book of abstracts - efmc-ismc 2022

497
Book of Abstracts International Symposium on Medicinal Chemistry Virtual Event Aug. 29-Sept. 2, 2021 EFMC-ISMC www.efmc-ismc.org EUROPEAN FEDERATION FOR MEDICINAL CHEMISTRY AND CHEMICAL BIOLOGY On behalf of Organised by

Upload: khangminh22

Post on 30-Jan-2023

0 views

Category:

Documents


0 download

TRANSCRIPT

Book ofAbstracts

International Symposium on Medicinal ChemistryVirtual EventAug. 29-Sept. 2, 2021

EFMC-ISMC

www.efmc-ismc.org

EUROPEAN FEDERATION FOR MEDICINAL CHEMISTRYAND CHEMICAL BIOLOGY

On behalf of

Organised by

2 | EFMC-ISMC

ContentWelcome 4

Committees 5

EFMC 6

Division of Medicinal Chemistry & Chemical Biology (DMCCB) of the Swiss Chemical Society (SCS) 7

Sponsors 8

Exhibitors 9

Daily Programme at a Glance 12

Company Workshops 16

Virtual Conference Center 20

Virtual Poster Hall 21

Virtual Exhibition 21

Plenary Lectures 27

Award & Prize Lectures 33

Invited Lectures & Oral Communications 43

EFMC-ISMC | 3

Posters - Chemical Biology 147

A. Carbohydrate Recognition and Drug Design 147

B. Chemical Approaches to Stem Cell Differentiation 165

C. Chemical Probes for Target Discovery and Validation 169

D. Molecular Imaging Tools for Chemical Biology 187

E. Photochemistry in Drug Discovery: Photopharmacology, Phototoxicity and Synthesis 195

Posters - Technologies 201

H. Application of Artificial Intelligence in Drug Discovery Projects 201

I. Biocatalysis & Late Stage Functionalisation 205

L. Innovative Synthesis for Medicinal Chemistry 207

M. Novel Strategies and Methods for Drug Discovery and Development 239

N. Prodrug Strategies in Drug Design and Discovery 299

Posters - Drug Discovery Projects 303

O. Addressing Neglected and Emerging Viral Diseases with Small Molecules 303

Q. Chemical Tools and Drug Discovery for Neuroinflammation 311

R. New Strategies and Agents for Targeting Gram-Negative Pathogens 321

T. Recent Advances in Anticancer Drug Discovery 339

U. Targeting Fibrotic Diseases with Small Molecules 381

V. Tissue and Cell Specific Drug Delivery 387

Posters - Diversified topics 393

W. Emerging Topics 393

X. First time disclosures 397

Z. Other 403

Index of Abstracts 447

Index of Authors 463

Index of Participants 485

4 | EFMC-ISMC

Welcome

Dear Colleagues,

A warm welcome to the 26th EFMC International Symposium on Medicinal Chemistry (EFMC-ISMC)!

Following a tradition established over half a century ago, the symposium is organised by the Division of Medicinal Chemistry and Chemical Biology of the Swiss Chemical Society, under the auspices of the European Federation for Medicinal Chemistry and Chemical Biology (EFMC). However, this edition of the EFMC-ISMC is special in more than one way: After the ravages caused by the SARS-CoV-2 pandemic forced us to postpone the symposium from 2020 to 2021, we finally have to run it (mostly) virtually, rather than on site in the beautiful city of Basel. A real shame, which will be rectified, however, in 2026 when the EFMC-ISMC will yet come to Basel, the city of Paracelcus, Reichstein and many other giants of science. Fortunately, the virtual nature of the symposium will have no effect on the quality of the science to be presented and we are grateful to all the lecturers and poster presenters who have committed to contribute to the event and help to make it a success.

Science and technology are at the heart of the Swiss economy, and they are strongly promoted by the govern-ment and the private sector. Indeed, the pharmaceutical industry in Switzerland is well-known for its investments in research, making Basel the main research centre for drug discovery in Europe. Today, the greater Basel area is home to a multitude of pharmaceutical companies and ten aca-demic centres, and it counts over 50’000 employees in the life sciences. Basel is a city of science, but it is also a city of culture, and we will tell you more about its histo-ry, museums and attractions when the EFMC-ISMC finally comes here in 2026, after the symposia in Nice in 2022 and in Rome in 2024. Hopefully, the pandemic will be under control long before this.

EFMC-ISMC 2021 offers you the opportunity to follow lec-tures by more than 100 speakers covering the chemical biology - medicinal chemistry continuum, and to access hundreds of posters describing the latest research in the field. The symposium is organised along three overarch-ing thematic streams, with parallel sessions focusing on chemical biology, drug discovery, and enabling technol-ogies. The questions that will be discussed in these ses-sions range from the development of novel tools to probe biological systems to the discovery of drug candidates

and their development. In addition, there are sessions on first time disclosures on Monday and Tuesday, as well as plenary lectures, company workshops, an exhibition and networking opportunities.

Naturally, not all relevant aspects of modern drug discov-ery can be covered by a single symposium. However, we believe that the breadth of the program of EFMC-ISMC 2021 and the timeliness of the topics offer a meaningful perspective on the state of the field and where it is mov-ing. We hope that the lectures, poster presentations and discussions (even if only virtual) will provide you with new insights and, above all, with new ideas and inspiration for your own work.

EFMC-ISMC 2021 also features lectures by the recipi-ents of the prestigious awards bestowed by the EFMC on a biennial basis for excellent scientific contribu-tions to drug discovery in the broadest sense: the Nauta Pharmacochemistry Award for Medicinal Chemistry and Chemical Biology, the UCB-Ehrlich Award for Excellence in Medicinal Chemistry, and the Prous Institute-Overton and Meyer Award for New Technologies in Drug Discovery. EFMC-ISMC 2021 for the first time will also host the cere-mony and lecture for the new Klaus Grohe Award.

This symposium would not have been possible without the help and support of many individuals and organisa-tions. In particular, we would like to thank the International Organising Committee, the Local Organising Committee and the Scientific Advisory Board for their contributions and hard work in helping put together this beautiful scien-tific programme.

The Symposium will start on Sunday with an opening lec-ture by Karin Briner (Novartis), and close on Thursday Sept 2 after Stefan Sieber’s Klaus Grohe Award lecture. We look forward to your active participation, and to welcom-ing you in our virtual convention centre for a week of excit-ing science.

With kind regards,

Karl-Heinz AltmannEFMC-ISMC 2021 Symposium Chairman

Yves P. AubersonEFMC President 2018-2020

EFMC-ISMC | 5

Committees

INTERNATIONAL ORGANISING COMMITTEEChairmanProf. Karl-Heinz ALTMANN (ETH Zürich, Zürich, Switzerland)

MembersProf. Marko ANDERLUH (University of Ljubljana, Ljubljana,

Slovenia)

Dr Yves P. AUBERSON (Novartis Institutes for Biomedical

Research, Basel, Switzerland)

Dr Karl-Heinz BARINGHAUS (Sanofi, Frankfurt, Germany)

Dr Werngard CZECHTIZKY (AstraZeneca, Mölndal, Sweden)

Prof. Beatriz DE PASCUAL-TERESA (CEU San Pablo

University, Madrid, Spain)

Dr Luc VAN HIJFTE (Symeres, Nijmegen, The Netherlands)

LOCAL ORGANISING COMMITTEEProf. Karl-Heinz ALTMANN (ETH Zürich, Zürich, Switzerland)

Dr Yves P. AUBERSON (Novartis Institutes for Biomedical

Research, Basel, Switzerland)

Dr Fides BENFATTI (Syngenta International, Basel,

Switzerland)

Mr Stefan BONSELS (Basel Tourismus, Basel, Switzerland)

Prof. Dennis GILLINGHAM (University of Basel, Basel,

Switzerland)

Prof. Christian HEINIS (Ecole Polytechnique Fédérale de

Lausanne (EPFL), Lausanne, Switzerland)

Dr Georg JAESCHKE (F. Hoffmann-La Roche, Basel,

Switzerland)

Dr Gerhard MÜLLER (Gotham Therapeutics GmbH,

Martinsried, Germany)

Prof. Jean-Louis REYMOND (University of Bern, Bern,

Switzerland)

Prof. Leonardo SCAPOZZA (University of Geneva, Genève,

Switzerland)

Mr David SPICHIGER (Swiss Chemical Society, Basel,

Switzerland)

Dr Cornelia ZUMBRUNN (Idorsia Pharmaceuticals Ltd.,

Allschwil, Switzerland)

INTERNATIONAL SCIENTIFIC COMMITTEEProf. Esin AKI-YALCIN (Ankara University, Ankara, Turkey)

Prof. Carolina H. ANDRADE (Federal University of Goias ,

Goiânia, Brazil)

Prof. Jonathan BAELL (Monash University, Parkville, Australia)

Prof. Ayla BALKAN (Hacettepe University, Ankara, Turkey)

Dr Jonathan BENNETT (Merck Sharp & Dohme, Boston,

United States)

Prof. Andrzej BOJARSKI (Institute of Pharmacology Polish

Academy of Sciences, Krakow, Poland)

Dr Martin BOLLI (Idorsia, Allschwil, Switzerland)

Dr Nicholas CARRUTHERS (Janssen Research &

Development (retired), San Diego, United States)

Prof. Violetta CECCHETTI (Università di Perugia, Perugia,

Italy)

Dr Jacobo CRUCES (GalChimia, S.A., O Pino, A Coruna, Spain)

Dr Hans-Michael EGGENWEILER (Merck KGaA, Darmstadt,

Germany)

Dr Ola ENGKVIST (AstraZeneca, Mölndal, Sweden)

Prof. Raphaël FREDERICK (UCLouvain, Brussels-Woluwe,

Belgium)

Prof. Alexander HILLISCH (Bayer, Wuppertal, Germany)

Prof. Jan KIHLBERG (Uppsala University, Uppsala, Sweden)

Dr Christian KUTTRUFF (Boehringer Ingelheim Pharma GmbH

& Co. KG, Biberach, Germany)

Prof. Thierry LANGER (University of Vienna, Vienna, Austria)

Prof. Emmanuel MIKROS (University of Athens, Athens,

Greece)

Dr Martin MISSBACH (Novartis, Basel, Switzerland)

Prof. Christa MÜLLER (University of Bonn, Bonn, Germany)

Prof. Adam NELSON (University of Leeds, Leeds, United

Kingdom)

Dr Jean-Claude ORTUNO (Institut de Recherches Servier,

Suresnes, France)

Prof. Roland PIETERS (Utrecht University, Utrecht, The

Netherlands)

Dr Alleyn T. PLOWRIGHT (Wren Therapeutics Ltd, Cambridge,

United Kingdom)

Prof. Vladimir POROIKOV (Institute of Biomedical Chemistry,

Moscow, Russia)

Prof. Thomas POULSEN (Aarhus University, Aarhus C,

Denmark)

Dr Raphaël RODRIGUEZ (Institut Curie, Paris, France)

Prof. Gianluca SBARDELLA (University of Salerno, Fisciano,

Italy)

Dr David SPRING (University of Cambridge, Cambridge, United

Kingdom)

Prof. Edgars SUNA (Latvian Institute of Organic Synthesis,

Riga, Latvia)

Dr Atli THORARENSEN (Pfizer, New-York, United States)

Dr Tihomir TOMASIC (University of Ljubljana, Ljubljana,

Slovenia)

Dr Adrijana VINTER (Fidelta Ltd, Zagreb, Croatia)

Prof. Nicolas WINSSINGER (University of Geneva, Geneva,

Switzerland)

Dr Clemens ZWERGEL (University of Rome La Sapienza,

Rome, Italy)

Dr Andrija ŠMELCEROVIĆ (University of Nis, Nis, Serbia)

6 | EFMC-ISMC

EFMC

The European Federation for Medicinal Chemistry and Chemical Biology (EFMC) is an independent association founded in 1970 that represents 27 scientific organisations from 25 European countries. Its objective is to advance the science of medicinal chemistry and chemical biology by promoting cooperation and encouraging strong links between the national adhering organisations in order to deepen contacts and exchanges between medicinal chemists and chemical biologists across Europe and around the world.

Its most important organisation is the biennial International Symposium on Medicinal Chemistry (EFMC-ISMC). These symposia, with an average attendance of 1,200 delegates, are highly international with a broad range of speakers and attendees from the pharmaceutical industry and aca-demia. Next to the EFMC-ISMC, EFMC is involved in the organisation of the EFMC|ACSMEDI Medicinal Chemistry Frontiers and the International Symposium on Advances in Synthetic and Medicinal Chemistry (EFMC-ASMC). EFMC also organises intensive short courses on specific topics in medicinal chemistry. The Young Medicinal Chemists’ Symposium (YMCS) on its side, has become an annual concentration of young talents.

Another important aspect of EFMC activities is the spon-sorship of national scientific meetings and Medicinal Chemistry Schools. EFMC also awards travel grants for younger scientists to attend EFMC symposia and schools.

EFMC also acknowledges the excellence of medicinal chemists’ work, by conferring three major awards: the Nauta Pharmacochemistry Award for Medicinal Chemistry and Chemical Biology, the UCB-Ehrlich Award for Excellence in Medicinal Chemistry and the Prous Institute-Overton and Meyer Award for New Technologies in Drug Discovery, which are given every two years for outstand-ing achievements in the field of Medicinal Chemistry. From 2010 on, EFMC established two prizes to acknowledge the scientific accomplishments of young medicinal chem-ists, both in industry and academia.

The EFMC has very strong links with the Medicinal Chemistry Division of the American Chemical Society (ACS), the European Federation for Pharmaceutical Sciences (EUFEPS), the European Association for Chemical and Molecular Sciences (EuCheMS), the Asian Federation of Medicinal Chemistry (AFMC), the Chinese Pharmaceutical Association (CPA) and the International Chemical Biology Society (ICBS) which gives the EFMC a broad international footprint.

More recently, the EFMC launched the Young Scientists Network which aims to inspire, connect and provide opportunities to medicinal chemists and chemical biolo-gists in their Early Career. They do so in organising vari-ous activities from networking events, webinars, soft-skill trainings, or mentoring programs. The EFMC-YSN Prize for the best PhD candidate has been launched in 2020 to rec-ognise excellence and merits is important since the early stage.

EFMC-ISMC | 7

Division of Medicinal Chemistry & Chemical Biology (DMCCB)

of the Swiss Chemical Society (SCS)

The DMCCB comprises about 750 scientists interested in medicinal chemistry, chemical biology and related fields of research. Beyond Switzerland, the DMCCB interacts with other country organisations to represent the Swiss medicinal chemistry and chemical biology community. The Division is also a member of the European Federation of Medicinal Chemistry (EFMC).

THE AIMS OF THE DMCCB ARE → to foster an international network of medicinal chemists, chemical biologists and scientists working in related fields → to facilitate contacts with leading experts in the field → to organise symposia, seminars and advanced training courses → to network and encourage the exchange of ideas

MAIN ACTIVITIES → Organise the bi-annual DMCCB Basel Symposium on a cutting-edge topic. → Organise the annual, international Peptide Therapeutics Forum in Basel. → Chair the MedChem & ChemBio sessions at the SCS Fall Meetings. → Collaborate with the medicinal chemistry and chemical biology divisions of chemical societies in neighbouring

countries to organise joint conferences. → Provide education to community members with the bi-annual Swiss Course on Medicinal Chemistry in Leysin or the

Swiss Summer School on Chemical Biology in Villars/Les Diablerets. → Publish the CHIMIA column ‘Highlights in Medicinal Chemistry’. → Inform the community via Social Media channels and with the DMCCB newsletter.

8 | EFMC-ISMC

SponsorsORGANISED BY ON BEHALF OF

GOLD SPONSORS

SILVER SPONSORS

BRONZE SPONSORS

AWARD & PRIZE SPONSORS

EFMC-ISMC | 9

Exhibitors

We also thank all our exhibitors for their support and invite you to visit them in the virtual exhibition hall

1. APEX Molecular 16 Iktos

2. Aragen Life Sciences 17. Instruct-ERIC

3. BioSolveIT 18. Jubilant Biosys

4. Biotage 19. Key Organics

5. Cambridge Crystallographic Data Centre 20. Kishida Chemical

6. Charles River 21. Liverpool ChiroChem

7. Charnwood Molecular 22. Merck

8. Chemical Computing Group 23. NovAliX

9. Collaborative Drug Discovery 24. Oncodesign

10. Concept Life Sciences 25. Schrodinger

11. EU-OPENSCREEN 26. Symeres

12. Eurofins Advinus 27. Synple Chem

13. Fidelta 28. Taros Chemicals

14. Fluorochem 29. UBE

15. Galchimia 30. WuXi AppTec

You need commitment, focus and passion to find new ways to fight the diseases of this world: innovation is at the heart of it.

Innovation for better health. Our commitment is to bring to patients around the world quality medicines for use in diagnosing, combating and preventing disease. Every day we work against time, researching new pathways, new molecules, new technologies – complementing our own capabilities with expertise of innovative partners from science and industry.

The success of this work is evidenced in new medicines for areas with significant unmet medical need such as oncology, cardiovascular and blood diseases, as well as gynecology and ophthalmology. Our aim is a better quality of life for all.

www.pharma.bayer.com

Curiosity is in our DNA. It has transformed us from a small pharmacy in 1668 to the global science and technology company we are today. As we look to the future, we can only imagine the breakthroughs it will make possible. Can you?

Discover more: curiosity.merckgroup.com

WITHOUT CURIOSITYthe earth would still be flat

12 | EFMC-ISMC

SUNDAY, AUGUST 29, 2021

10:00 EFMC-ISMC 2021 Opening Ceremony & Plenary Lecture

10:55 Nauta Award Ceremony & Lecture

11:45 Break

13:10 UCB-Ehrlich Award Ceremony & Lecture

14:00 Prous Institute-Overton and Meyer Award Ceremony & Lecture

14:40 EFMC Fellows Ceremony

15:15 End of the Day

MONDAY, AUGUST 30, 2021

09:30 Plenary Session

10:15 Session 1 Chemical BiologyMolecular Imaging Tools for

Chemical Biology

Session 2 TechnologiesNovel Strategies and Methods for Drug Discovery and Development

(CPA Session)

Session 3 Drug Discovery Projects

Chemical Tools and Drug Discovery for Neuroinflammation

(EuChemS Session)

12:00 End of Morning Session

12:30 Company WorkshopSchrödinger |LiveDesign: a Collaborative Environment for Real-world Drug Discovery Workflows

13:15 Poster Session 1, Exhibition & Networking

15:30 Session 4 Chemical BiologyPhotochemistry in Drug

Discovery: Photopharmacology, Phototoxicity and Synthesis

(ACSMEDI Session)

Session 5 TechnologiesCryo-EM as an Emerging Tool for Small-Molecule Drug Discovery

Session 6 Drug Discovery Projects

Next Generation Drugs for Heart Failure

17:15 Session 7.1.First Time Disclosures (Part I)

18:45 End of the Day

TUESDAY, AUGUST 31, 2021

09:30 EFMC Prize Ceremony & Lectures 2020

10:15 Session 8 Chemical BiologyChemical Probes for Target

Discovery and Validation

Session 9 TechnologiesInnovative Synthesis for

Medicinal Chemistry

Session 10 Drug Discovery Projects

Addressing Neglected and Emerging Viral Diseases with

Small Molecules (AFMC Session)

12:00 End of Morning Session

12:30 Company Workshops- Chemical Computing Group | Designing Molecules in the Binding Site

- WuXi AppTec | DEL Screening Technology as an Enabling Tool for Medicinal Chemists

13:15 Poster Session 2, Exhibition & Networking

15:30 Session 11 Chemical BiologyChemical Approaches to Stem

Cell Differentiation(ICBS Session)

Session 12 TechnologiesExpanding Chemical Space

through AI - Linking Synthesis Prediction and Automation

Session 13 Drug Discovery Projects

Breathing Life into Inhaled Drug Discovery: Challenges and Breakthroughs to the Clinic

(ACSMEDI Session)

17:15 Session 7.2.First Time Disclosures (Part II)

18:45 End of the Day

Daily Programme at a Glance

EFMC-ISMC | 13

WEDNESDAY, SEPTEMBER 1, 2021

09:30 EFMC Prize Ceremony & Lectures 2021

10:15 Session 14 Chemical BiologyCarbohydrate Recognition and

Drug Design

Session 15 TechnologiesApplication of Artificial

Intelligence in Drug Discovery Projects

Session 16 Drug Discovery Projects

New Strategies and Agents for Targeting Gram-Negative

Pathogens

12:00 End of Morning Session

12:30 Company Workshops- NovAliX | DNA-Encoded Library Technology: Strengths and Development Opportunities

- Charles River | Developing Inhaled Drugs for Respiratory Diseases: A Medicinal Chemistry Perspective

13:15 Poster Session 3, Exhibition & Networking

15:30 Session 17 Chemical BiologySmall Molecules Targeting RNA

Function and Processing

Session 18Highlights of Medicinal Chemistry

Session 19 Drug Discovery Projects

Tissue and Cell Specific Drug Delivery

(EUFEPS Session)

17:15 Plenary Session

18:00 End of the Day

THURSDAY, SEPTEMBER 2, 2021

09:30 Plenary Session

10:15 Session 20Emerging Topics

Session 21 TechnologiesProdrug Strategies in Drug

Design and Discovery

Session 22 Drug Discovery Projects

Targeting Fibrotic Diseases with Small Molecules

12:00 End of Morning Session

13:15 Poster Session 4, Exhibition & Networking

15:30 Session 23 Chemical BiologyTarget Deconvolution Strategies

in Drug Discovery

Session 24 TechnologiesBiocatalysis & Late Stage

Functionalisation

Session 25 Drug Discovery Projects

Recent Advances in Anticancer Drug Discovery

17:15 Plenary SessionThe Klaus Grohe Award Ceremony & Lecture

18:05 EFMC-ISMC Closing Remarks

18:20 End of the Symposium

By putting lives first, we’ve created a legacy that lastsFor 130 years, we have tackled some of the world’s biggest health challenges and provided hope in the fight against disease, for both people and animals. Today, we continue our commitment to be the premier research-intensive biopharmaceutical company in pursuit of medical breakthroughs that benefit patients and society for today, tomorrow and generations to come.

Copyright © 2021 Merck Sharp & Dohme Corp., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA. All rights reserved. HQ-NON-00309 02/20

Novartis Institutes for BioMedical Research Global Discovery Chemistry

Changing the practice of medicineAt Novartis, we harness the innovation power of chemical biology and medicinal chemistry to address some of society’s most challenging healthcare issues. Our researchers work to push the boundaries of science, broaden our understanding of diseases and develop novel products in areas of great unmet medical need. We are passionate about discovering new ways to extend and improve patients’ lives.

16 | EFMC-ISMC

Company Workshops

A number of company workshops will take place during EFMC-ISMC, in between the morning and afternoon sessions. Participation in the workshops is free of charge, but we kindly ask you to register for the workshops of your interest via your online registration.

MONDAY, AUGUST 30 | 12:30-13:15 CEST - SCHRÖDINGER → LiveDesign: a Collaborative Environment for Real-world Drug Discovery Workflows

Presenter: Stephan EhrlichLiveDesign is Schrodinger’s Enterprise Informatics platform, providing a powerful web-based data capture, analysis, and workflow interface to the underlying experimental data, chemoinformatics and modeling tools that drive modern drug discovery. The ability to assess validated advanced computational results side by side with assay and structure data in a real-time fashion has allowed Schrodinger and many global pharma companies to shorten molecular designs cycles and made exploration of large chemical spaces easy. At the same time, LiveDesign democratizes simpler computational workflows like docking, machine learning or virtual compound generation.This workshop will provide a hands-on impression of the platform, focusing on ideation of new com-pounds, visualization of those ideas in 3D, and enabling truly collaborative design decisions. Access to the platform will be provided to participants; please make sure you have a recent version of either the Chrome or Firefox browsers installed on your machine.

TUESDAY, AUGUST 31 | 12:30-13:15 CEST - CHEMICAL COMPUTING GROUP → Designing Molecules in the Binding Site

Presenter: Sarah WitzkeComputational techniques may be applied on one or more existing ligand-bound crystal structures to produce the best – and most easily synthesised – molecules to fit within a binding site. This is illus-trated using the drug Tarceva bound to its EGFR receptor, and applying capabilities within the MOE (Molecular Operating Environment) software system;

ț The structure is analysed by inspecting molecular surfaces and maps, and similar ligands are docked using a pharmacophore filter.

ț A database of replacement scaffolds is scanned to find new potential molecular frameworks that match or improve on the existing ligand.

ț A specific chemical reaction is applied to the molecule in the context of the binding site, with a database of reagents, to find easily-synthesised molecules that offer enhanced properties.

EFMC-ISMC | 17

TUESDAY, AUGUST 31 | 12:30-13:15 CEST - WUXI APPTEC → DNA-encoded libraries in drug discovery

Presenter: Dr Alex Satz DNA-Encoded library (DEL) screening is now commonly used in the pharmaceutical industry to find novel chemical matter that modulates protein targets of interest. A DEL is a mixture of millions of drug-like small-molecules, where each molecule is conjugated to a DNA-oligomer that encodes its chemical structure. The composition of a DEL mixture can be readily interrogated before and after interaction with a protein target by Next Generation Sequencing, and small-molecules that selectively bind the target identified.We’re excited to invite you to a webinar on this topic that Dr Alex Satz, Senior director of DEL strategy and operations at WuXi AppTec, will talk about the current learnings in DEL design, synthesis, screen-ing, and present some case studies.

WEDNESDAY, SEPTEMBER 1 | 12:30-13:15 CEST - NOVALIX → DNA-Encoded Library Technology: Strengths and Development Opportunities

Presenter: Olivier Querolle, Drug Discovery DirectorSince several years now, DNA-encoded library (DEL) technology has emerged as a valuable and robust platform for hit identification of multiple biological targets. In this workshop, NovAliX will review in detail the overall process and key principles of DEL meaning library design and production, affinity screening and hit identification. Then, through several examples, the power of this technology for the discovery of new chemical matter will be highlighted.

However, even if DEL displays several advantages compared to traditional screening methods, there is still room for scientific and technical developments which will reinforce the power of this technology. As an example, continuous emergence of novel DNA-compatible reactions and the increasing acces-sibility of new building blocks offer new avenues for library design and optimization of the chemical space coverage. Therefore, the last part of the workshop will be dedicated to share our view on the developments to come in the DEL field.

WEDNESDAY, SETPEMBER 1 | 12:30-13:15 CEST - CHARLES RIVER → Developing Inhaled Drugs for Respiratory Diseases: A Medicinal Chemistry Perspective

Presenter: Elisa Pasqua, Group LeaderDespite the devastating impact of many lung diseases on human health, there is still a significant unmet medical need in respiratory diseases, where inhaled delivery represents a critical strategy. There are many advantages to delivering a therapeutic directly to the lungs, but designing a drug for inhaled delivery is challenging. Watch this presentation to learn more about designing drugs for inhaled delivery.

ExtraordinaryAt Roche, we are committed to both. Discovering and developing innovative medicines and diagnostic tests to help people live better, longer lives.

medicine requires extraordinary science.

2101

_ELD

_F

Life is a health journey, with its ups and downs, and its challenges. These can be big or small, lifelong or momentary.

Everyone, from children to elder people, can be facing health challenges and needs, wherever they are.

We, at Sanofi, are there for those challenges, by the side of people, as a health journey partner.

Sanofi is about Empowering Life.

www.sanofi.de

20 | EFMC-ISMC

Virtual Conference Center

Our Virtual Conference Center will be the platform to use for your participation at the event. Similarly to a real-life conference center, you’ll have access to different rooms: auditorium, workshops, exhibition and posters, as well as the networking area. After the event, the replays of the talks will also be available from here.

Our platform is very user-friendly, but feel free to have a look at the Tutorial which you can find under Information & Tutorial once logged in.

In addition to the scientific sessions, the programme includes a series of networking moments. We invite you to take some time to interact with other delegates, to have a look at the virtual posters and to visit the exhibi-tion during these moments.

We advise you to turn your status to ‘available’ as much as possible during the networking moments, to maximise interaction with other delegates.

EFMC-ISMC | 21

Virtual Poster Hall

All accepted abstracts will be on display in the virtual post-er hall. The posters will be accessible by all registered del-egates starting from 1 week ahead of the official opening of the symposium and until 2 weeks after the end date.

For each poster, the visitor will be able to do the following: → • view the PDF poster and its corresponding abstract → • view a short video presentation of the poster → • chat with the author

The Poster Sessions will be organised according to the following schedule:

→ Poster Session 1 (posters with even numbers)Monday, August 30, from 13:15 to 15:30

→ Poster Session 2 (posters with odd numbers)Tuesday, August 31, from 13:15 to 15:30

→ Poster Session 3 (posters with even numbers)Wednesday, September 1, from 13:15 to 15:30

→ Poster Session 4 (posters with odd numbers)Thursday, September 2, from 13:15 to 15:30

Poster presenters are requested to be virtually pres-ent during the sessions assigned to their poster, in order to answer questions in the live chat. Chat messages will be visible in real-time for those online and remain visible throughout the symposium.

Posters are grouped into 4 main themes: → Chemical Biology → Technologies → Drug Discovery Projects → Diversified Topics

Within each theme, sub-categories can be selected for an ease of navigation.

Virtual Exhibition

30 companies are participating in the virtual exhibition of EFMC-ISMC. For a user-friendly experience they have been grouped into Categories:

→ Biotech Companies → Contract Research Organisations → Equipment Providers → Fine Chemicals → Software & Database Providers

In each exhibition booth participants can view compa-ny-related information and get in touch with the represen-tatives of each company, either via chat or via the Book a Meeting assistant when available.

Image: Boehringer Ingelheim

www.boehringer-ingelheim.com

As a leading research-driven biopharmaceutical company,

Boehringer Ingelheim is working on breakthrough therapies

in areas of high unmet medical need. Founded in 1885

and family-owned ever since, Boehringer Ingelheim takes

a long-term perspective. Around 52,000 employees serve

more than 130 markets in the three business areas,

Human Pharma, Animal Health, and Biopharmaceutical

Contract Manufacturing.

Value through Value through Value through innovationinnovationinnovation

Improving the health of humans and animals – Our goal.

ABCD

BI_Corporate-Ad-2021_210x297mm.indd 1BI_Corporate-Ad-2021_210x297mm.indd 1 04.06.21 10:2404.06.21 10:24

We research, we innovate, we care. Since 1901

www.gedeonrichter.com

LZD: 2021.07.12. PR/101/2021

Health is our mission

More science – Bursting with ideas

The purpose of Idorsia is to discover, develop and commercialize innovative medicines to help more patients.

www.idorsia.com

Idorsia supports the recognition of scientific excellence.

“Crossing boundaries to Build Strong Networks for Early Career Researchers”

The EFMC-Young Scientists Network aims to inspire, connect and facilitate medicinal chemists and chemical

biologists in their Early Career.

Registration is free of charge and open to all!

Visit www.efmc.info/ysn for more information, or mail us at [email protected]

Follow us and spread the word!

YoungSciNet

@YoungSciNet

Our activities

Communication› Interview of scientists› Quizzes› Photo Competition

› Networking activities› “Meet & Greet” events for young people› Networking evenings at the EFMC-YMCS

› Training activities› “Soft-skills” workshops› Career fairs› Mentoring programme

› Support to young scientists› Travel grants to attend EFMC Events› YSN Prize for best PhD/Post-doc› Job & academic positions portal

› Events & Meetings› EFMC-YMCS (www.efmc-ymcs.org)› EFMC-YSN MedChemBioOnline (www.efmc.info/efmc-ysn-medchembioonline)

With the support of

changeWe don't wait for change, we drive

EFMC-ISMC | 27

Plenary Lectures

28 | EFMC-ISMC

PL001

CREATING THE NEXT GENERATION OF THERAPEUTICS -

MEDICINAL CHEMISTRY UNLEASHED

Karin Briner

Novartis,Head of Global Discovery Chemistry

250 Massachusetts AvenueMA 02139 Cambridge

United States

No abstract

EFMC-ISMC | 29

PL002

MEDICINAL CHEMISTRY: MORE THAN EVER

Jean-Paul Clozel

Idorsia Pharmaceuticals Ltd - Hegenheimermattweg 91 - 4123 Allschwil - Switzerland

Idorsia, the company which I am leading, was created after the acquisition of Actelion by Johnson & Johnson. At

its creation Idorsia retained all the early research projects, could keep the labs, all Drug Discovery researchers

and about a third of Actelion’s Drug Development groups.

Since the creation of Idorsia we decided to focus on organic chemistry as a tool for discovering new drugs. At a

time when a large portion of new drugs are antibodies, at a time when gene therapy, gene editing, CART cells

are in the forefront of the drug industry news, this decision may seem counterintuitive.

During my talk I will explain why we believe organic chemistry has fantastic potential, will remain a key source

of new drugs and is the best approach for many diseases. I will also explain how organic chemistry has allowed

Idorsia to build a very diverse pipeline with more than 14 drugs in clinical development.

30 | EFMC-ISMC

PL003

TRANSLATIONAL CHEMISTRY

Phil Baran

The Scripps Research Institute, Department of Chemistry10550 N. Torrey Pines Rd, BCC-169

CA 92037 La JollaUnited States

There can be no more noble undertaking than the invention of medicines. Chemists that make up the engine of

drug discovery are facing incredible pressure to do more with less in a highly restrictive and regulated process

that is destined for failure more than 95% of the time. How can academic chemists working on natural products

help these heroes of drug discovery – those in the pharmaceutical industry? With selected examples from our lab

and others, this talk will focus on that question highlighting interesting findings in fundamental chemistry and

new approaches to scalable chemical synthesis.

EFMC-ISMC | 31

PL004

TARGETING PROTEIN SCAFFOLDING FUNCTION IN KINASES

Stefan Knapp (1,2)

1) Johann Wolfgang Goethe-University, Institute for Pharmaceutical Chemistry, Max-von-Laue-Str. 9, D-60438 Frankfurtam Main, Germany

2) Johann Wolfgang Goethe-University, Structural Genomics Consortium, Buchmann Institute for Life Sciences;Max-von-Laue-Str. 15, D-60438 Frankfurt am Main, Germany

In living cells, proteins are organized in large complexes comprising adapter proteins, enzymes and regulatory

proteins. The roles of the protein components of such large multifunctional complexes are usually assigned

based on simplistic models and the complexity of regulation of multiprotein complexes is neglected in the

development of inhibitors and drugs targeting classical enzymes. However, studies on the dynamics of protein

interactions and signalling networks demonstrated that scaffolding is not only an important function of structural

and non-enzymatically active proteins, but it is also a feature of most classical enzymes that should not be

neglected during drug development. In this talk, I will exemplify the implications of altering protein interactions

by allosteric small molecules as well as by canonical ATP competitive inhibitors using protein kinases as an

example. I will demonstrate how different binding modes that alter protein conformation and dynamics may

result in diverse effects on cellular signalling as well as on phenotypic responses. Targeting scaffolding roles

may also establish new target classes such as catalytically inactive pseudokinases, that represent a considerable

group of largely unexplored targets which have been linked to the development of many diseases.

EFMC is an independent association founded in 1969, representing 27 societies from 25 European countries, and more than 7500 scientists.It’s main objective is to advance the science of medicinal chemistry and chemical biology.

EFMC-YSN MedChemBioOnlineWebinars mixing science, so�-skills training & round table discussionswww.efmc.info/efmc-ysn-medchembioonline

EFMC 16th Short Course on Medicinal ChemistryNew Opportunities in GPCR Drug DiscoveryOegstgeest, The Netherlands | May 8-11, 2022

EFMC-ISMC 2022XXVII EFMC International Symposium on Medicinal ChemistryNice, France | September 4-8, 2022

EFMC-YMCS 20229th EFMC Young Medicinal Chemists’ SymposiumNice, France | September 8-9, 2022

— The Nauta Pharmacochemistry Award for Medicinal Chemistry and Chemical Biology— The “UCB-Ehrlich Award for Excellence in Medicinal Chemistry”— Prous Institute - Overton and Meyer Award for New Technologies in Drug DiscoveryVisit www.efmc.info/awards for more information

— EFMC Prizes for Young Medicinal Chemists in Industry & AcademiaVisit www.efmc.info/prizes for more information

Upc

omin

g Ev

ents

Aw

ards

Priz

esEF

MC

-YSN The Young Scientists Network

Building a strong network at an early stage in your career is crucial!The aim of the EFMC-YSN is to inspire, connect and provide opportunities to medicinal chemists and chemical biologists in their Early Career. Visit www.efmc.info/ysn for more information

WEBSITE EFMC.INFO AND SUBSCRIBE TO THE MONTHLY NEWSLETTER FOLLOW EFMC ON

@EuroMedChem

European Federation for Medicinal Chemistryand Chemical Biology

EFMC

MedChemBioOnline

EFMCYoung ScientistsNetwork

YSN

International Symposium on Medicinal ChemistryNice, FranceSeptember 4-8, 2022

EFMC-ISMC

Young Medicinal Chemists' SymposiumNice, FranceSeptember 8-9, 2022

EFMC-YMCS

16th Short Courseon Medicinal ChemistryOegstgeest, NetherlandsMay 8-11, 2022

EFMC

EFMC-ISMC | 33

Award & Prize Lectures

34 | EFMC-ISMC

AL001

PARTIAL AGONISM IN THE PICTURE

Ad P. IJzerman

Leiden Academic Centre for Drug Research - Division of Drug Discovery and SafetyPO Box 9502, 2300RA Leiden, The Netherlands

One of the first pharmacologists to address the concept of partial agonism on G protein-coupled receptors was

Evert Ariëns (1918-2002). In his typical way of phrasing he used to compare a partial agonist with a pianist

having one hand bound, somehow suggesting that partial agonism is troublesome. A first realization is that

partial agonism is not only compound- but also context-dependent. As an example, an adenosine A

1

receptor

agonist may be a partial agonist in heart tissue but emerge as a full agonist when studied on fat pad lipolysis, as a

consequence of differences in receptor densities. Secondly, with an evolving understanding of cellular

pharmacology it is now also appreciated that even within a cell a ligand may act as a full agonist for certain

cellular pathways, e.g. G protein activation, while in others it is only partially so, e.g. in beta-arrestin

modulation. This biased behavior may offer opportunities for drug discovery, but it seems that most

pharmaceutical companies, keeping Ariëns’s metaphor in mind, are hesitant to spend time and other resources on

the phenomena of both partial and biased agonism.

Over the years my team together with colleagues over the world has studied partial agonism on the four subtypes

of adenosine receptors, aspects of which I will integrate into this lecture. Our joint efforts have recently

culminated in elucidating the structure of the adenosine A

2A

receptor in the presence of a partial agonist. This

detailed 3D architecture provides atomic resolution of the principle of partial agonism with clear hints for future

drug discovery.

EFMC-ISMC | 35

AL002

EXPERIENCES & FUTURE DIRECTION FOR MEDICINAL

CHEMISTRY IN CARDIOVASCULAR, RENAL AND METABOLISM

DISEASES

Malin Lemurell

Department of Medicinal Chemistry, Research and Early Development Cardiovascular, Renal and Metabolism,BioPharmaceuticals R&D, AstraZeneca

The presentation will highlight project examples and experiences from medicinal chemistry and drug discovery

within the areas of Cardiovascular, Renal and Metabolic diseases. Patients frequently suffer from co-morbidities

across these diseases and the intertwined disease-driving pathways require a broad range of target classes to be

tackled. Furthermore, the nature of these diseases invariably demand chronic dosing and a wide therapeutic

window. Today, we build new systems biology knowledge graphs using ‘omics and AI and as medicinal

chemists we are challenged to find new ways to modulate more and more  demanding target classes. Examples

of how we are expanding our toolbox,  in the classical small molecule drug design and through embedding New

Modalities and capabilities towards difficult targets, incl. RNA, will be discussed. Reflecting on the pace that the

science is advancing and the opportunities ahead of us, the role of a medicinal chemist today is truly fascinating.

36 | EFMC-ISMC

AL003

DE NOVO DRUG DESIGN WITH MACHINE INTELLIGENCE

Gisbert Schneider (1,2)

1) ETH Zurich, Department of Chemistry and Applied Biosciences, Vladimir-Prelog-Weg 4, 8093 Zurich, Switzerland2) ETH Singapore Ltd, 1 CREATE Way, #06-01 CREATE Tower, Singapore

Drug design may be regarded as a pattern recognition process. Medicinal chemists are skilled in visual chemical

structure recognition and their association with (retro)synthesis routes and pharmacological properties. In this

context, various “artificial intelligence” (AI) methods have emerged as potentially enabling technology for drug

discovery and automation, because these systems aim to mimic the chemist’s pattern recognition process and

take it to the next level by considering the available domain–specific data and associations during model

development. Part of the appeal of applying AI methods in drug design lies in the potential to develop

data-driven, implicit model building processes to navigate vast datasets and to prioritize alternatives. This

represents at least a partial transfer of decision power to a machine intelligence, and could be viewed as

synergistic with human intelligence; that is, a domain-specific implicit AI that would augment the capabilities of

medicinal chemists in drug design and selection. More ambitiously, the ultimate challenge for drug design with

AI is to autonomously generate new chemical entities with the desired properties from scratch (de novo). We

will review the principles of AI methods for de novo drug design, emphasizing ligand-based approaches that

have proven useful and reliable in “little-data” situations. Selected prospective case studies will be presented,

ranging from targeted molecular design to fully automated design-make-test-analyze cycles. We will also

provide a critical assessment of the possibilities and limitations of this approach and dare forecasting the future

of drug design with machine intelligence.

References

1) Schneider, P., Walters, W. P., Plowright, A. T., Sieroka, N., Listgarten, J., Goodnow Jr., R. A., Fisher, J., Jansen, J. M.,

Duca, J. S., Rush, T. S., Zentgraf, M., Hill, J. E., Krutoholow, E., Kohler, M., Blaney, J., Funatsu, K., Luebkemann, C.,

Schneider, G. (2020) Rethinking drug design in the artificial intelligence era. Nature Reviews Drug Discovery 19, 353–364.

2) Schneider, G. (2019) Mind and machine in drug design. Nature Machine Intelligence 1, 128–130.

3) Schneider, G. (2018) Automating drug discovery. Nature Reviews Drug Discovery 17, 97–113.

4) Schneider, P., Schneider, G. (2016) De novo design at the edge of chaos. Journal of Medicinal Chemistry 59, 4077–4086.

EFMC-ISMC | 37

AL004

CHEMICAL-PROTEOMIC STRATEGIES TO FIGHT

MULTIRESISTANT BACTERIA

Stephan Sieber

Technical University Munich, Department of ChemistryLichtenbergstr. 4 - 85747 Garching, Germany

Multiresistant bacterial pathogens such as Methicillin-resistant Staphylococcus aureus (MRSA) are responsible

for a variety of severe infections that pose a significant threat to global health. To approach this challenge new

chemical entities with an unprecedented mode of action are desperately needed. This presentation will cover our

latest efforts to identify new anti-bacterial targets and corresponding chemical inhibitors. A proteome mining

approach will be presented to identify cofactor-dependent enzymes as novel antibiotic targets. Small molecule

cofactor mimics infiltrate the bacterial metabolic machinery leading to their incorporation in PLP-dependent

enzymes. Their analysis via mass-spectrometry revealed the function of uncharacterized proteins in important

bacterial pathways as well as the mechanism of action of known antibiotics.

In a separate approach we identified new synthetic or natural product derived compound classes that effectively

kill pathogenic bacteria. Chemical synthesis of improved derivatives led to the identification of active molecules

with nanomolar potency and suitable metabolic stability. The mode of action was investigated by diverse

methodologies including affinity based protein profiling (AfBPP). For example, one compound stimulates a

signal peptidase correlating with enhanced secretion of extracellular proteins. These included essential cell-wall

remodeling enzymes whose dysregulation likely explains the associated antibiotic effects.

38 | EFMC-ISMC

PR001

DISCOVERY OF POTENT SELECTIVE GABA

A

ALPHA5 POSITIVE

ALLOSTERIC MODULATORS (PAMs) FOR THE TREATMENT OF

NEUROLOGICAL DISORDERS

Giuseppe Cecere

Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Grenzacherstrasse 124, 4070Basel, Switzerland

[email protected]

GABA

A

receptors are ligand-gated chloride channels and the main mediators of inhibitory synaptic transmission

in the human brain. There are 19 genes encoding for GABA

A

receptor subunits that assemble as pentamers, with

the most common stoichiometry of two α, two β, and one γ subunit. The α5 subunit-containing GABA

A

receptors are of particular interest given their specific expression pattern and physiological properties (Sur et al.,

1999; Mohler, 2011). Multiple lines of evidence suggest that excessive neural activity in selected brain regions

with consequent imbalance between excitatory/inhibitory neurotransmission underlie a variety of neurological

disorders such as epilepsy, Autism Spectrum Disorder (ASD), Schizophrenia and Alzheimer’s disease. The

presentation will highlight our effort to identify highly potent, selective GABA

A

 α5 PAMs from a program that

already led to a clinical NAM asset (Basmisanil). Key medicinal chemistry concepts involved in the optimization

of the ligands and structural determinants underlining the NAM-to-PAM switch will be disclosed. Finally, proof

of concept studies with selected PAMs in disease-relevant animal models will be presented.

EFMC-ISMC | 39

PR002

WITHIN OUR CONTROL? ILLUMINATING HOW EPHEMERAL

ELECTROPHILES REWIRE CELL DECISION-MAKING PROCESSES

Yimon Aye

EPFL SB ISIC LEAGOCH A2 397 (Batiment CH)

Station 61015 Lausanne

Switzerland

Precisely timed and spatially regulated electrophilic chemical signals are slowly being implicated as bona fide

signaling events in numerous cells. However, modeling these low-stoichiometry signaling events and defining

the precise biological impacts of localized signals under physiologic conditions has proven to be highly

challenging

1

. The first half of the presentation will spotlight a unique set of proximity-directed chemical biology

tools that enables interrogation into functional consequences of specific redox-linked events: namely, T-REX™

precision electrophile delivery and G-REX™ electrophile-ligandability profiling in living systems

2

, and how

using these technologies have enabled us to identify bona fide “first responders” that interact with native

signaling electrophiles under close to endogenous redox signaling conditions (i.e., “k

cat

/K

m

”-like). Our data

show that these first responders lie at nexuses between electrophile- and canonical-signaling pathways. Thus,

these proteins translate information encoded by electrophiles to phosphate or ubiquitin to reroute signaling

pathway flux, even at the organismal level. The second half of the presentation will relate to our latest data of

unique relevance to medicinal chemistry research: i.e., our new ability to discover and functionally decipher

precision electrophile signaling mechanisms

3

toward targeted therapeutics and novel target discovery

4

. Here,

I will discuss successful small-molecule targeting of otherwise hard-to-treat pten-null triple-negative breast

cancers in cells and xenograft mouse tumor models

5

, guided by our recent identification of

protein-isoform-specific native electrophilic metabolite sensing in living cells and larval zebrafish

6

.

References

1) https://www.ncbi.nlm.nih.gov/pubmed/30913473 Genie in a Bottle: Controlled Release Helps Tame Natural

Polypharmacology? Current Opinion in Chemical Biology 2019 51 48 (with Long et al.)

2) https://www.ncbi.nlm.nih.gov/pubmed/30765181 Interrogating Precision Electrophile Signaling, Trends in Biochemical

Sciences 2019 44 380 (with Poganik et al.)

3) 2020 (in Revision in Nature Chemical Biology) (with Zhao et al.)

4) https://www.ncbi.nlm.nih.gov/pubmed/28648380 Privileged Electrophile Sensors: A Resource for Covalent Drug

Development, Cell Chemical Biology (Invited Perspective) 2017 24 787 (with Long)

5) Liu, Long, and Aye, 2018 PCT Patent filed; Liu et al., 2020 (in Revision in ACS Central Science)

6) https://www.ncbi.nlm.nih.gov/pubmed/28114274 Akt3 is A Privileged First Responder in Isozyme-Specific Electrophile

Response, Nature Chemical Biology 2017 13 333 (with Long and Parvez et al.)

40 | EFMC-ISMC

PR003

INTEGRIN ANTAGONISTS FOR TREATMENT OF IDEOPATHIC

PULMONARY FIBROSIS

Niall Anderson

GlaxoSmithKlineMedicines Research Centre

Gunnels Wood RoadStevenage

SG1 2NY HertfordshireUnited Kingdom

Fibrotic diseases lead to progressive loss of tissue function and organ failure and are estimated to contribute

˜45% of deaths in the developed world. Idiopathic pulmonary fibrosis (IPF) is one of these and is poorly treated

with a survival rate lower than that of most cancers. It is characterised by excessive scarring - unregulated wound

healing - in the lung with transforming growth factor (TGF) thought to play a pivotal role. Integrins are cell

surface receptors (involved in cell adhesion inter alia) involved in activating TGF and it is thought that their

inhibition - and in particular the integrin avb6  - may have a therapeutic benefit in treating IPF. This presentation

will focus on our work on the synthesis and properties of ανβ6 inhibitors which led to a clinical candidate.

 

EFMC-ISMC | 41

PR004

COVID MOONSHOT – CROWDSOURCING A COVID-19 CURE

Nir London

The Weizmann Institute of Science, Organic Chemistry76100 Rehovot, Israel

COVID-19, caused by SARS-CoV-2, lacks effective therapeutics. Additionally, no antiviral drugs or vaccines

were developed against the closely related coronavirus, SARS-CoV-1 or MERS� CoV, despite previous zoonotic

outbreaks. To identify starting points for such therapeutics, we performed a large-scale screen of electrophile and

non-covalent fragments through a combined mass spectrometry and X-ray approach against the SARS-CoV-2

main protease. The results with more than 71 co-crystal structures that span the entire active site, were released

to the public and we implemented a crowed-sourcing platform to solicit next generation design ideas. Thousands

of suggestions were submitted, of which we made and tested >1,500, through a global consortium including

academic labs, chemical vendors, pharma advisors and more. This effort resulted in 100nM inhibitors with

crystallographic confirmation and activity in live virus assays. Crowd computing (Folding@Home) was

harnessed for compound optimization; AI (via PostEra) for prioritization and retrosynthesis. This is still an

on-going live project, that can benefit from the input of medicinal chemists world-wide. We hope this initiative

can serve as a template for an alternative drug discovery pipeline for areas that are unappealing to traditional

pharma such as pandemic preparedness and antibiotic development.

42 | EFMC-ISMC

PR005

THE DISCOVERY OF RELPAX

®

AND NURTEC ODT

®

- TWO

DIFFERENT TREATMENTS FOR MIGRAINE HEADACHES

John Macor

Global Head Integrated Drug Discovery, SANOFI, Waltham, MA02541, USA

The discovery of Relpax® (eletriptan) and Nurtec ODT® (rimegepant) will be presented.  Eletriptan is a 5-HT

1D

receptor agonist (a triptan) and has been on the market for over 20 years, while Rimegepant is a CGRP receptor

antagonist and was launched in March 2020.  The medicinal chemistry behind both molecules will be presented. 

EFMC-ISMC | 43

Invited Lectures & Oral Communications

44 | EFMC-ISMC

LE001

TRANSLATIONAL CHEMICAL BIOLOGY

Gonçalo Bernardes

Yusuf Hamied Department of ChemistryUniversity of Cambridge, UK

Our research uses chemistry principles to address questions of importance in life sciences and molecular

medicine. This lecture will cover recent examples of two projects in my research group:

i) we computationally identify and experimentally validate TRPV2 as a target for piperlongumine and establish a

link between allosteric target engagement and impaired glioblastoma progression in vivo;

ii) we describe a small molecule-based platform to hijack RNA methylation by guided degradation for high

resolution profiling of modified substrates.

EFMC-ISMC | 45

LE002

CHEMICAL TOOLS TO STUDY REDOX BIOLOGY

Pablo Rivera-Fuentes (1), Jade Nguyen (1,2), Sarah Hübner (1), Henriette Lämmermann (1)

1) Institute of Chemical Sciences and Engineering, EPF Lausanne, Switzerland2) Department of Chemistry and Applied Biosciences, ETH Zurich, Switzerland

Redox homeostasis is essential for organelle function in eukaryotes and its disruption is associated with cancer,

metabolic and neurodegenerative diseases. In eukaryotes, however, subcellular compartments have vastly

different redox environments ranging from reducing potentials in mitochondria to oxidizing conditions in the

endoplasmic reticulum. The redox potential in cells and their organelles are largely defined by the ratio of

reduced to oxidzed glutathione (GSH:GSSG) because this is the redox pair that is present in the highest

concentrations. In this lecture, I will present fluorescent sensors that we developed recently to measure absolute

concentrations of glutathione in selected eukaryotic organelles, small-molecule chemical probes that can be used

to disrupt the GSH:GSSG ratio, and genetically encoded gene reporters that help elucidate the stress response

pathways that are activated upon disruption of redox homeostasis in specific organelles.

46 | EFMC-ISMC

LE003

SENSING ENZYMATIC ACTIVITY WITH QUANTUM DOTS IN

PATIENT-DERIVED NEURODEGENERATIVE DISEASE MODELS

Valle Palomo (1,2)

1) Centro de Investigaciones Biológicas Margarita Salas, CSIC , Ramiro de Maeztu 9 , 28040 Madrid , Spain.2) Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED) , Instituto Carlos III ,

28031 Madrid , Spain.

Fluorescence-based assays for monitoring enzyme activity with Quantum Dots represents a powerful technique

that allows for unique sensing opportunities.

1

We developed a reliable system that allows the quenching of

fluorescent particles by simple canonic amino acid side chains to find broad utility for peptide-based sensors and

that would be advantageous for exopeptidases. Using this system we described a quantum dot (QD)-based

electron transfer (ET) sensor able to allow direct, quantitative sensing of both endopeptidase and exopeptidase

activity. 3,4-dihydroxyphenylalanine (DOPA) is a nonproteinogenic amino acid that can be incorporated into a

peptide sequence allowing for the quenching of QD photoluminescence through an ET mechanism. DOPA can

replace a phenylalanine or tyrosine residue without severely altering structural properties, allowing for its

introduction at multiple positions within a biologically active peptide substrate.

2

Consequently, the quenching

system presented here is ideally suited for incorporation into diverse peptide substrates for enzyme recognition,

digestion, and activity sensing. Our findings suggest a broad utility of a small ET-capable amino acid side chain

in detecting enzyme activity through ET-mediated QD luminescence quenching.

 

Following this successful implementation, we have started to work in the development of a BACE1 sensor for in

vivo protease monitoring in living cells from patients. While BACE1 is a validated drug target for Alzheimer’s

disease it has also proven to have a role in other neurodegenerative diseases.

3

We are combining the possibilities

of enzyme activity monitoring with quantum dots in human models derived from patients to allow for a

personalized analysis of pathological mechanisms together with a tailored selection for treatment for specific

patients.

References

1) Energy Transfer with Semiconductor Quantum Dot Bioconjugates: A Versatile Platform for Biosensing, Energy

Harvesting, and Other Developing Applications. Hildebrandt N, Spillmann CM, Algar WR, Pons T, Stewart MH, Oh E,

Susumu K, Díaz SA, Delehanty JB, Medintz IL. Chem Rev. 2017, 117, 536-711.

2) 3,4-Dihydroxyphenylalanine Peptides as Nonperturbative Quantum Dot Sensors of Aminopeptidase. Palomo V, Díaz SA,

Stewart MH, Susumu K, Medintz IL, Dawson PE. ACS Nano. 2016, 10, 6090-9.

3) Pharmacological BACE Inhibition Improves Axonal Regeneration in Nerve Injury and Disease Models. Tallon C,

Marshall KL, Kennedy ME, Hyde LA, Farah MH. Neurotherapeutics. 2020, 17, 973-88.

EFMC-ISMC | 47

LE004

PATTERN-GENERATING FLUORESCENT MOLECULAR PROBES

FOR CHEMICAL BIOLOGY

Leila Motiei

Weizmann Institute of ScienceChemical and Structural Biology

234 Herzl Street - 7610001 RehovotIsrael

Fluorescent molecular probes have become a powerful tool in protein research. However, these probes are less

suitable for analyzing specific populations of proteins in their native environment. In this talk I will give an

overview of a new class of fluorescent molecular probes

1-5

recently developed in our group, and show how they

can be used to detect individual proteins, protein combinations, as well as binding interactions and dynamic

changes that occur on their surfaces. In the second part of this talk, I will present a new class of fluorescent

molecular sensors that combines the properties of small molecule-based probes and cross-reactive sensor arrays

(the so-called chemical nose/tongue’) and explain how these pattern-generating probes could expand the

fluorescent toolbox currently used to detect and image proteins.

6

Specifically, I will show how such systems can

be used to identify combinations of specific protein families within complex mixtures and to discriminate among

protein isoforms in living cells, where macroscopic arrays cannot access.

6

References

1) Rout, B.; Milko, P.; Iron, M. A.; Motiei, L.; Margulies, D. J. Am. Chem. Soc. 2013, 135, 15330.

2) Unger-Angel, L.; Rout, B.; Ilani, T.; Eisenstein, M.; Motiei, L.; Margulies, D. Chem. Sci. 2015, 6, 5419.

3) Nissinkorn, Y.; Lahav-Mankovski, N.; Rabinkov, A.; Albeck, S.; Motiei, L.; Margulies, D. Chem. Eur. J. 2015, 21, 15981.

4) Motiei, L.; Pode, Z.; Koganitsky, A.; Margulies, D. Angew. Chem. Int. Ed. 2014, 53, 9289.

5) Hatai, J.; Motiei, L.; Margulies D. J. Am. Chem. Soc. 2017, 139, 2136.

6) Pode, Z.; Peri-Naor, R.; Georgeson, J. M.; Ilani, T.; Kiss, V.; Unger, L.; Markus, B.; HM, B.; Motiei, L.; Margulies, D.

Nature Nanotechnol. 2017, 12, 1161.

48 | EFMC-ISMC

LE005

ALONG THE ALLOSTERY STREAM: RECENT ADVANCES IN

COMPUTATIONAL METHODS FOR ALLOSTERIC DRUG

DISCOVERY

Zhengtian Yu, Jian Zhang

Shanghai Jiaotong UniversityNutshell Therapeutics, Shanghai, China

Allostery is a universal, biological phenomenon in which orthosteric sites are fine-tuned by topologically distal

allosteric sites triggered by perturbations, such as ligand binding, residue mutations, or post-translational

modifications. Allosteric regulation is implicated in a variety of physiological and pathological conditions and is

thus emerging as a novel avenue for drug discovery. Allosteric drugs have traditionally been discovered by

serendipity through large-scale experimental screening. Recently, we have witnessed significant progress in

biophysics, particularly in structural bioinformatics, which has facilitated the in-depth characterization of

allosteric effects and the accurate detection of allosteric residues and exosites. These advances improve our

understanding of allosterism and promote allosteric drug discovery, thereby revolutionizing the shift from the

traditional serendipitous route used to discover allosteric drugs to the updated path centered on rational

structure-based design. In this talk, recent advances in computational methods applied to allosteric drug

discovery were summarized. We comprehensively reviewed these achievements along various levels of allosteric

events, from the construction of allosteric databases to the identification and analysis of allosteric residues,

signals, sites, and modulators. We expect to increase the awareness of the discovery of allosteric drugs using

structure-based computational methods.

 

EFMC-ISMC | 49

LE006

MOLECULAR GLUE DESIGN FOR TARGETED CANCER THERAPY

Jie Chen (1), Peihao Chen (1), Lu Lv (1), Nan Liu (2), Youwei Ai (1), Yuxing Sun (1), Longzhi Cao (1),

Yuanxun Wang (1), Dianrong Li (1), Hongwei Wang (2), Niu Huang (1), Ting Han (1), Xiaodong Wang

(1), Xiangbing Qi (1)

1) National Institute of Biological Sciences, Beijing, 1022062) Tsinghua University, Beijing, 100084

Molecular glue is a class of small molecule that acts on the interface of protein and induces the protein-protein

interaction (PPI). Usually there are two types of molecular glue, either inducing the formation of a complex with

higher binding affinity and exerting a stronger downstream function (stabilizer); or inducing protein interaction

for degradation and inhibiting the function of targeted protein (degrading agent). In terms of molecular

glue-induced protein degradation, we developed a new type of molecular glue HQ461 and revealed that HQ461

induces the interaction of CDK12 and DDB1, leading to the CDK12 binding protein cyclin K polyubiquitin and

degradation. In terms of molecular glue-enhanced protein-protein interaction, based on the discovery of

PDE3A/SLFN12, a new target for inducing apoptosis, we discovered that Anagrelide, as a new molecular glue,

can induce the binding of PDE3A and SLFN12, thereby activating the apoptosis pathway by the SLFN12 protein

stabilization. We also solved the complex structure of PDE3A/SLFN12/Anagrelide through high-resolution

cryo-electron microscopy. Based on the structure, we developed a more active molecular glue A6 (IC50, 0.3 nM)

and the in-vivo tumor growth inhibition activity was also verified. The novel mechanism of molecular glue

provides more opportunities for broader range of drug targets, laying the foundation for the development of

molecular glue drugs.

Keywords: molecular glue, PPI, protein degradation, apoptosis, Targeted cancer therapy

References

1) Lu Lv, Peihao Chen, Longzhi Cao, Yamei Li, Zhi Zeng, Yue Cui, Qingcui Wu, Jiaojiao Li, Jian-Hua Wang, Meng-Qiu

Dong, Xiangbing Qi* and Ting Han*, “Discovery of a molecular glue promoting CDK12-DDB1 interaction to trigger Cyclin

K degradation”, eLife, 2020;9:e59994 (Journal)

2) Youwei Ai*, Haibing He, Peihao Chen, Bo Yan, Wenbin Zhang, Zhangcheng Ding, Dianrong Li, Jie Chen, Yan Ma, Yang

Cao, Jie Zhu, Jiaojiao Li, Jinjie Ou, Shan Du, Xiaodong Wang, Jianzhang Ma, Shuanhu Gao* and Xiangbing Qi*, “An

alkaloid initiates phosphodiesterase 3A–schlafen 12 dependent apoptosis without affecting the phosphodiesterase activity”,

Nature Communication, 2020, (11), 3236 (Journal)

50 | EFMC-ISMC

LE007

DISCOVERY OF NEW SARS-COV-2 M

pro

INHIBITORS WITH

POTENT IN VITRO AND IN VIVO ANTIVIRAL ACTIVITY

Shengyong Yang

State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China.

The pandemic of COVID-19 due to the SARS-CoV-2 infection poses serious threats to global public health. The

main protease (M

pro

) of SARS-CoV-2 is an attractive drug target because of its central role in viral replication.

We designed and synthesized totally 32 new bicycloproline-containing M

pro

inhibitors. These compounds

exhibited vigorous potency against SARS-CoV-2 M

pro

, and the co-crystal structure of M

pro

in complex with one

of the most active compounds revealed an interaction mode. Of note, two compounds (MI-09 and MI-30)

showed excellent antiviral activity in cell-based assays. In a SARS-CoV-2 infected transgenic mouse model,

oral/ intraperitoneal treatment with MI-09 or MI-30 significantly reduced lung viral loads and lung lesions.

Meanwhile, they displayed good pharmacokinetic properties and safety in rats, suggesting their potentials to be

developed as efficient SARS-CoV-2 antiviral drugs.

EFMC-ISMC | 51

LE008

DEVELOPMENT OF A NEW TARGET IDENTIFICATION SYSTEM

FOR SMALL MOLECULES IN TRYPANOSOMA BRUCEI PARASITES

Suzanne Sherihan Sahraoui (1,2), Oscar Vadas (3), Sébastien Tardy (1,2), Oliver Petermann (1,2), Aurélie

Gouiller (1,2), Leonardo Scapozza (1,2)

1) Pharmaceutical Biochemistry/Chemistry Group, School of Pharmaceutical Sciences, University of Geneva, CMU - RueMichel-Servet 1, CH-1211 Geneva 4, Switzerland

2) Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CMU - Rue Michel-Servet 1, CH-1211Geneva 4, Switzerland

3) Department of Microbiology and Molecular Medicine, University of Geneva, CMU - Rue Michel-Servet 1, CH-1211Geneva 4, Switzerland

Neglected diseases as the Human African Trypanosomiasis relies on drugs associated with many side effects

which cause a real problem for the effective treatment of populations (1).

The discovery of current drugs used against Human African Trypanosomiasis has been made possible by using

phenotypic screening measuring the effects of molecules on parasite growth and survival. Although this

approach has been successful, the targeted proteins, the mechanism of action, and often the cause of the side

effects remain unsolved.

To address this question and to support drug design and lead optimization, scientists started to develop methods

to identify cellular targets and elucidate the mechanism of action of lead compounds (2).  Up to now, many

target identification techniques have been developed and successfully applied. However, some drawbacks such

as the loss of some parts of the proteome or strong background signals have been reported (3, 4).

To overcome these limitations, this project was designed to develop a novel system made of the merging of two

recently developed techniques based on BioID and SNAP-tag technology, that will allow the identification of

small molecules targeting proteins in the cytosol and organelles of the sub-proteome of Trypanosoma brucei as

well as breaking the boundaries of the two methods taken separately.

In this paper, we will report the results achieved while addressing the different challenges linked to the

development of the method. Indeed, designing a plasmid enabling homologous recombination and a

polycistronic gene expression in Trypanosoma brucei represents the first challenge that has been thrivingly

accomplished. The ability to obtain stable cell lines correctly integrating the gene of interest remains arduous and

uncertain. The work leading to successful transduction will be presented.

The synthesis of BG-PEG4-C1 was chosen as the first test compound and BG-PEG4-Mtx as the second for the

proof of concept, the results of their cell permeation studies will be reported. Finally, the assessment of the

correct biotinylation of targets by the tool and the ability to identify them will be described.

Keywords: Target deconvolution, Trypanosoma brucei, BioID, BirA*, SNAP-tag, organelles, sub-proteome,

BG-PEG4-C1, BG-PEG4-Mtx (Mtx: Methotrexate)

 

References

1) Babokhov P, Sanyaolu AO, Oyibo WA, Fagbenro-Beyioku AF, Iriemenam NC. A current analysis of chemotherapy

strategies for the treatment of human African trypanosomiasis. Pathog Glob Health. 2013;107(5):242-52.

2) Swinney DC. Phenotypic vs. target-based drug discovery for first-in-class medicines. Clinical pharmacology and

therapeutics. 2013;93(4):299-301.

3) Morriswood B, Havlicek K, Demmel L, Yavuz S, Sealey-Cardona M, Vidilaseris K, et al. Novel bilobe components in

Trypanosoma brucei identified using proximity-dependent biotinylation. Eukaryotic cell. 2013;12(2):356-67.

4) Roux KJ, Kim DI, Burke B. BioID: a screen for protein-protein interactions. Current protocols in protein science.

2013;74:Unit 19.23.

52 | EFMC-ISMC

LE009

MICROGLIA IN NEUROINFLAMMATION: AN INTRIGUING

TARGET TOWARDS NEUROPROTECTION AND

NEUROREGENERATION

Barbara Monti

Department of Pharmacy and Biotechnology,University of Bologna,

Bologna, Italy

Neuroinflammation is a common feature of most neuropathologies, including neurodegenerative,

neurodevelopmental, neuropsychiatric and demyelinating disorders. Therefore, understanding the cellular and

molecular basis of this process may lead to the identification of new potential therapeutic targets to counteract

these devastating neuropathological conditions. Many different nervous and glial cells are involved in

neuroinflammation, but, in the last years, attention has been mainly focused on microglia. These are the

predominant type of immune cells in the brain, which play an important role in many physiological processes,

during both development and adulthood, being involved in synapsis formation, function and plasticity, as well as

in neuronal trophic support and protection from infections. However, in pathological conditions, microglia

undergo activation and change their role from physiological to pathological one, thus playing a key role in

neuroinflammation. This functional change is related to a microglia phenotypic shift: in physiological

conditions, microglia is surveying the brain parenchyma, but in neuropathologies microglia become activated. In

early stages of brain diseases, microglia acquire a M2 activated phenotype, which is able to phagocyte protein

aggregates and cell debris, to release neuroprotective and neurotrophic factors. Later on microglia acquire a M1

phenotype, which loses the M2 neuroprotective features and become neurotoxic, further contributing to

neuroinflammation and neurodegeneration. The immunomodulatory approach, i.e. the ability to revert the

microglial phenotype from the neurotoxic M1 to the neuroprotective M2 one, seems to be a promising strategy to

counteract neuroinflammation and neurodegeneration, as well as to support regeneration of both neurons and

glial cells. In this framework, in collaboration with many research groups, we were able to identify and modulate

several biochemical and molecular targets in microglial cells (kinases as GSK3beta, epigenetic enzymes as

HDACs, receptors as CBRs, etc.), thus leading to immunomodulation during neuroinflammatory events and

therefore promoting neuroprotective and neuroregenerative processes.

EFMC-ISMC | 53

LE010

DEVELOPMENT OF FLUORINATED PET TRACERS FOR IMAGING

OF SIGMA-1 RECEPTORS IN THE BRAIN

Bernhard Wünsch

Institut für Pharmazeutische und Medizinische Chemie, WWU Münster, Corrensstraße 48, D-48149 Münster, Germnay

It has been shown that σ

1

receptors play an important role in several human diseases including depression,

schizophrenia, Alzheimer’s disease, as well as drug/alcohol addiction. In particular, antagonists at the σ

1

receptor

potentiate the pain-relieving effects of opioid analgesics and can be used for the treatment of neuropathic pain.

Due to their overexpression in several human tumor cell lines, σ

1

and σ

2

receptors are interesting targets for

tumor therapy and diagnosis.

In the talk, the development of a PET (positron emission tomography) tracer for labeling of s

1

receptors in the

brain is presented. Synthetic strategies including late stage diversification and enantioselective synthesis,

relationships between the structure and s

1

affinity and receptor selectivity are reported. Some general features for

analyzing the pharmacokinetic properties (logD

7.4

value, metabolic stability, formed metabolites) are discussed.

Finally, the results of a first in men clinical trial are shown.

54 | EFMC-ISMC

LE011

BOOSTING THE RESOLUTION OF INFLAMMATION THROUGH

FORMYL PEPTIDE RECEPTOR 2 (FPR2) AGONISTS AS A NOVEL

STRATEGY IN NEUROINFLAMMATION-ASSOCIATED CENTRAL

NERVOUS SYSTEM DISORDERS

Enza Lacivita (1), Margherita Mastromarino (1), Antonio Carrieri (1), Igor A. Schepetkin (2), Lilyia N.

Kirpotina (2), Ewa Trojan (3), Kinga Tylek (3), Mark T. Quinn (2), Agnieszka Basta-Kaim (3), Marcello

Leopoldo (1)

1) Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari, Via Orabona, 4, 70125 Bari, Italy2) Department of Microbiology and Immunology, Montana State University, Bozeman, MT 59717, USA

3) Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 SmetnaStreet, 31-343 Kraków, Poland

Chronic or unresolved inflammation is a key pathological process in various diseases, including

neurodegenerative disorders. Successful resolution of inflammation requires the activation of endogenous

pathways that can switch from the production of pro-inflammatory to specialized pro-resolving mediators

(SPMs). New insights into such pathways are offering novel opportunities to pharmacologically manipulate the

resolution of inflammation and, eventually, to open new therapeutic approaches for chronic inflammation [1].

The formyl peptide receptor 2 (FPR2), a receptor modulated by several SPMs, such as lipoxin A4 and resolvins,

is one of the key players in the resolution of inflammation [2]. Recently, we have identified a class of

non-peptidic FPR2 agonists with a ureidopropanamide scaffold having neuroprotective properties in several invitro and in vivomodels of Central Nervous System (CNS) disorders characterized by chronic neuroinflammation

[3,4]. Here we report on the structure-activity relationships of new potent ureidopropanamide-based FPR2

agonists specifically designed to cross the blood-brain barrier.The effect of the most potent compounds on

viability/metabolic activity, necrotic death, and production of pro-and anti-inflammatory mediators in primary

microglial cells and hippocampal organotypic cultures in normal conditions and after LPS or b-amyloid

stimulation will be illustrated by highlighting the potential of these agonists in the treatment of CNS diseases

characterized by neuroinflammation.

References

1) Fullerton, J. N.; Gilroy, D. W. Resolution of inflammation: a new therapeutic frontier. Nat. Rev. Drug Discov. 2016,

15(8):551-67

2) Park, J.; Langmead, C.J; Riddy, D.M. New advances in targeting the resolution of inflammation: implications fro

Specialized Pro-Resolving Mediator GPCR drug discovery. ACS Pharmacol. Transl. Sci. 2020, 3, 88-106

3) Stama, M. L.; Ślusarczyk, J.; Lacivita, E. et al. Novel ureidopropanamide based N-formyl peptide receptor 2 (FPR2)

agonists with potential application for central nervous system disorders characterized by neuroinflammation. Eur. J. Med.

Chem. 2017, 141:703-720.

4) Trojan, E.; Tylek, K.; Schroder, N.; et al. The N-Formyl Peptide Receptor 2 (FPR2) agonist MR39 suppresses amyloid

beta (1-42)-induced neuroinflammation in the ex-vivo and in vivo mouse model of Alzheimer’s disease. Mol. Neurobiol.

Submitted

EFMC-ISMC | 55

LE012

SMALL MOLECULE DEGRADERS TARGETING FOLDING

INTERMEDIATES: THE PRION PROTEIN CASE STUDY

Andrea Astolfi (1), Giovanni Spagnolli (2,3), Tania Massignan (2,3,4), Petro Faccioli (5,6), Emiliano

Biasini (2,3), Maria Letizia Barreca (1)

1) Department of Pharmaceutical Sciences, University of Perugia, 06123 Perugia, PG, Italy.2) Department of Cellular, Computational and Integrative Biology, University of Trento, 38123 Povo, TN, Italy.

3) Dulbecco Telethon Institute, University of Trento, 38123 Povo, TN, Italy.4) Sibylla Biotech SRL, 37121 Verona, VR, Italy.

5) Department of Physics, University of Trento, Povo, Trento, TN, Italy.6) INFN-TIFPA, University of Trento, Povo, Trento, TN, Italy.

Decades of research efforts have conclusively provided overwhelming evidence that the cellular prion protein

(PrP

C

) represents an optimal pharmaceutical target to tackle prion diseases, a set of fatal and incurable

neurodegenerative disorders characterized by the conformational conversion of the physiological PrP

C

into a

misfolded and infectious isoform referred to as PrP scrapie (PrP

Sc

). Indeed, PrP

C

plays a key role in the disease

etiology and knock-out experiments demonstrated that its therapeutic suppression can be considered safe.[1]

Over the years different strategies have been proposed to tackle this target based on traditional drug discovery

approaches, such as the identification of small molecules able to promote the PrP

C

relocalization from cellular

membrane to intracellular endosomes, as well as PrP

C

binders that prevent its conversion to PrP

Sc

. However no

therapy is yet available, and prion disease still represents a currently unmet medical need. [2]

Very recently, we have exploited the potential of a radically different therapeutic rationale by blocking the

folding pathway of PrP instead of targeting its native state. We refer to this strategy as Pharmacological Protein

Inactivation by Folding Intermediate Targeting (PPI-FIT).[3] The rationale underlying PPI-FIT is that targeting a

folding intermediate with small ligands could promote its degradation by the cellular quality control machinery,

which recognizes such artificially stabilized intermediates as improperly folded species.

In detail, the reconstruction of the PrP folding pathway through all-atoms MD simulation allowed the

identification of a metastable intermediate of the PrP folding pathway characterized by a druggable pocket.

Virtual screening of a commercial small molecule library resulted in the identification of thirty potential binders,

four of which capable of selectively lowering the load of PrP into the cellular membrane and promote its

degradation. Additionally, one of these compounds inhibits prion replication in a dose-dependent fashion. Our

data collectively suggest that these small molecule degraders are worthy of further development and

investigations, as they could represent a unique pharmacological resource for the treatment of prion diseases and

much more frequent disorders of the nervous system, such as Alzheimer's and Parkinson's diseases, in which a

harmful role of PrP has recently been identified.[4]

References

1) Colini Baldeschi, A.; Vanni, S.; Zattoni, M.; Legname, G. Novel regulators of PrPC expression as potential therapeutic

targets in prion diseases. Expert Opin. Ther. Targets 2020, 00, 1–18

2) Astolfi, A.; Spagnolli, G.; Biasini, E.; Barreca, M.L. The Compelling Demand for an Effective PrPC-Directed Therapy

against Prion Diseases. ACS Med. Chem. Lett. 2020, 11, 2063–2067.

3) Spagnolli, G.; Massignan, T.; Astolfi, A.; et al. Pharmacological inactivation of the prion protein by targeting a folding

intermediate. Commun. Biol. 2021, 4, 1–16

4) Laurén, J.; Gimbel, D.A.; Nygaard, H.B.; Gilbert, J.W.; Strittmatter, S.M. Cellular prion protein mediates impairment of

synaptic plasticity by amyloid-Β oligomers. Nature 2009, 457, 1128–1132

56 | EFMC-ISMC

LE013

PHOTOPHARMACOLOGY: TOWARDS LIGHT-CONTROLLED

THERAPY

Wiktor Szymanski

Medical Imaging Center, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9747AG Groningen,The Netherlands;

Current pharmacological treatments rely on using bioactive compounds that evoke a response by interacting with

molecular targets in the human body. The selectivity of this interaction is crucial and the lack of it leads to the

emergence of severe side-effects in the body and toxicity in the environment. This is especially the case in

chemotherapy, which employs inherently cytotoxic molecules, and in the antibiotic therapy, where the overuse

of bioactive substances has led to the emergence of antimicrobial resistance.

To solve this problem, drugs could be introduced whose activity could be reversibly or irreversibly turned on

with light, enabling local activation with high spatiotemporal resolution. The aim of this presentation is to

describe the emerging concept of photopharmacology (Figure A),[1] which is currently being developed and

applied to precisely control the activity of drugs using UV/visible/NIR light. Light offers unparalleled

advantages in regulation of bioactivity. Importantly, light of the same energy is also used as an input/output

signal in medical (mostly optical/optoacoustic) imaging. Combination of those two paradigms along the

principles of theranostics requires light-responsive tools that empower both therapy and imaging. 

The presentation will focus on our efforts towards bridging light and medicine, focusing first on new

light-operated tools [2] (molecular photoswitches [3,4,5] and photocages [6], Figure B). Next, I will highlight the

synergies between medical imaging and therapy, offered by light, through photoresponsive optical [7] and

magnetic resonance [8,9] imaging agents. The examples of light-controlled bioactive molecules presented will

include small molecules [10] and proteins.[11] Finally, using those examples, I will discuss the structural aspects

[12] and design principles [13] in photopharmacology.

Figure. The principle of photopharmacology (A) and its key molecular tools (B).

References

1) M. W. H. Hoorens et al. Trends. Biochem. Sci. 43, 567 (2018);

2) I. M. Welleman et al. Chem. Sci. 11, 11672-11691 (2020);

3) L. N. Lameijer et al. Angew. Chem. Int. Ed. 59, 21663-21670 (2020);

4) M. W. H. Hoorens et al. Nature Comm. 10, 2390 (2019);

5) M. W. H. Hoorens et al. Chem. Sci. 12, 4588-4898 (2021);

6) K. Sitkowska et al. ChemComm, 56, 5480-5483 (2020);

7) F. Reessing et al. ACS Omega 5, 22071-22080 (2020);

8) F. Reessing et al. Curr. Opin. Biotech. 58, 9 (2019);

9) F. Reessing et al. Chem. Comm. 55, 10784 (2019);

10) D. Kolarski et al. Nat. Commun. ASAP (2021);

11) N. Mutter et al. J. Am. Chem. Soc. 141, 14356 (2019);

12) V. Arkhipova et al. J. Am. Chem. Soc., 143, 1513–1520 (2021);

13) D. Kolarski et al. Org. Biomol. Chem. 19, 2312 – 2321 (2021).

EFMC-ISMC | 57

LE014

MAPPING CELL-CELL INTERACTIONS IN TUMOR

MICROENVIRONMENT VIA PHOTOCATALYTIC PROXIMITY

LABELING

Niyi Fadeyi

Merck & Co, Cambridge, United States

Membrane proteins play essential roles in an extensive range of cellular functions.  One notable example is the

surface interaction between immunomodulatory receptors (IMRs) to initiate and regulate immune responses. 

The success in modulating these interactions with checkpoint inhibitors such as anti-PDL1/CTLA4 demonstrates

the tremendous therapeutic value in understanding how IMRs interact and the impact of neighboring proteins on

modulating IMR function.  Protein proximity labeling represents a powerful approach for the unbiased

assessment of protein-protein interactions or bystander proteins with effector function on the cell surface.  A

number of enzyme-based proximity labeling strategies have been developed over the last decade that either

generate a reactive labeling species in proximity to the protein of interest, or physically "stamp" neighboring

proteins.  The success of these approaches has led to the consideration of non-enzyme based methods that are

smaller in size, can be temporally controlled, and/or can avoid harsh treatment conditions.  This talk will

describe the development of novel photocatalytic-based proximity labeling approaches whereby protein residues

are labeled in the presence of visible light and a photocatalyst.  Applications of this technology on the cell

surface and within cell interaction environments will also be showcased.

 

58 | EFMC-ISMC

LE015

AZAAURONES AS NOVEL CHEMOTYPES AGAINST

MYCOBACTERIUM TUBERCULOSIS: SAR, ADME PROFILING AND

PHOTO-SWITCHING PROPERTIES

André Campaniço (1), Shrika Harjivan (1), Elisabete Freitas (1), Audrey Jordaan (2), Digby Warner (2),

Rui Moreira (1), Francisca Lopes (1)

1) Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, University of Lisbon, Av. Prof. Gama Pinto,1649-003 Lisboa, Portugal

2) Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch 7701, South Africa;SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, Department of Pathology, University of Cape Town,Rondebosch 7701, South Africa; Welcome Centre for Infectious Diseases Research in Africa, University of Cape Town,

Rondebosch 7701, South Africa

Tuberculosis (TB) is a deadly disease caused by a single infectious agent, Mycobacterium tuberculosis (M.tb).

The complexity and duration of the treatment lead to misuse and low compliance by patients, increasing disease

burden and the appearance of multidrug-resistant strains of M.tb. Thus, new antibiotics active against

drug-resistant M.tb and useful for short period therapeutic regimens at lower required doses are urgently needed.

[1,2]

A family of azaaurone-based derivatives, from a chemical library developed in iMed.ULisboa, revealed to be

active against M.tb, including multidrug- and extensively drug-resistant tuberculosis from clinical isolates, at a

submicromolar level. [3] Despite the promising activities, this new scaffold displayed poor ADME properties.

We now report the complete SAR exploration and ADME profiling of newly synthesized derivatives. Along

with an enhanced metabolic stability and solubility, rings A and B as well as N-substitutions were extensively

explored. (Figure 1) The double bond within the scaffold was also reduced to a single bond, generating a new

family of saturated azaaurones. Furthermore, the E and Z isomers were isolated, allowing a differential study of

each and revealing biological- and photo-conversion.

Figure 1 Azaaurones as potent antitubercular derivatives.

Acknowledgements: This research was funded by projects UIDB/04138/2020 and UIDP/04138/2020 (Fundação

para a Ciência e Tecnologia (FCT), Portugal) and PTDC/MED-FAR/30266/2017 and

LISBOA-01-014-FEDER-030266 (FCT and FEDER). We also acknowledge FCT for fellowship

SFRH/BD/131896/2017 (A.C.). This work was also supported by the Strategic Health Innovation Partnerships

(SHIP) initiative of the South African Medical Research Council with funds from the National Treasury under its

Economic Competitiveness and Support Package (D.F.W.).

References

1) Campaniço A, Moreira R, Lopes F, Eur J Med Chem., 2018, 150, 525-545.

2) Ehrt S et al, Cell Microbiol., 2009, 11, 1170-1178.

3) Campaniço A, Carrasco MP, Njoroge M, Seldon R, Chibale K, Perdigão J, Portugal I, Warner DF, Moreira R, Lopes F.,

ChemMedChem, 2019, 14, 1537-1546

EFMC-ISMC | 59

LE016

IN VIVO ADRENERGIC MODULATION WITH

PHOTOPHARMACOLOGY

Davia Prischich (1,2), Alexandre M. J. Gomila (1,2), Santiago Milla-Navarro (3), Gemma Sangüesa (4,5),

Rebeca Diez-Alarcia (6,7), Beatrice Preda (1), Carlo Matera (1,2), Montserrat Batlle (4,5), Laura Ramírez

(3), Ernest Giralt (8,9), Jordi Hernando (10), Eduard Guasch (4,5), J. Javier Meana (6,7), Pedro de la

Villa (3,11), Pau Gorostiza (1,2,12)

1) Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology, Barcelona, Spain2) Centro de Investigación Biomédica en Red – Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain

3) Department of Systems Biology, University of Alcalá (UAH), Madrid, Spain4) Institut Clínic Cardiovascular, Hospital Clinic, University of Barcelona (UB), IDIBAPS, Barcelona, Spain

5) Centro de Investigación Biomédica en Red – Enfermedades Cardiovasculares (CIBER-CV), Spain6) Department of Pharmacology, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain

7) Centro de Investigación Biomédica en Red - Salud Mental (CIBER-SAM), Spain.8) Department of Inorganic and Organic Chemistry, University of Barcelona (UB), Barcelona, Spain

9) Institute for Research in Biomedicine (IRB), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain10) Departament de Química, Universitat Autònoma de Barcelona (UAB), Cerdanyola del Vallès, Spain

11) Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain12) Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain

Adrenoceptors are ubiquitous and regulate most vital functions in the human body, including heart and

respiratory rate, digestion, smooth-muscle contraction, gland secretion, and pupil diameter among others. In

addition, adrenergic neurons firing from the locus coeruleus towards different areas of the central nervous

system mediate alertness, responses to acute stress and danger, pain modulation, arousal, sleep-wake cycles, as

well as neuroplasticity and cognitive behaviour. Despite the physiological relevance of adrenergic

neurotransmission, molecular methods to precisely modulate the activity of endogenous adrenoceptor and to

functionally dissect their pathways in vivo are not available. 

Here we present a set of photochromic ligands, that we call adrenoswitches, to switch on and off adrenoceptor

activity with high spatio-temporal resolution. Using a non-canonical azologization approach, we have designed

novel arylazoheteroarene units that we have characterized in vitro and in two animal models (zebrafish

locomotion and pupillary reflex in mice). The drug-like properties of these molecules, their efficacy and absence

of acute toxicity in zebrafish larvae, and most remarkably the fact that specific adrenergic photomodulation was

readily and reversibly achieved in the mammalian eye by topical application without formulation, all indicate

that adrenoswitches could be a disruptive tool to dissect physiological adrenergic signaling and to develop safe

and effective therapies. For example, photocontrol of adrenoceptors at specific locations might allow to single

out individual adrenergic projections from the locus coeruleus, or to selectively decouple pupil tone from

environmental illumination.  

 

60 | EFMC-ISMC

LE017

CRYO-EM IN GPCR STRUCTURE BASED DRUG DISCOVERY

Stacey Southall

Sosei Heptares,Steinmetz Building, Granta Park

Great Abington, Cambridge CB21 6DGUnited Kingdom

Structure-Based Drug Discovery (SBDD) for GPCRs has been revolutionized by the wealth of new structural

information that has become available in recent years, enhancing our understanding of their function and the

mechanism of action of ligands. X-ray crystallography has been the method of choice for GPCR structure

determination and has been routinely used at Sosei Heptares, driven by access to stabilised receptors (StaR®),

[1-3]. Cryo-electron microscopy (cryo-EM) has now become established as a complementary approach to

determine novel structures and drive SBDD. Indeed, since 2017, more structures of GPCRs in the active state

coupled to heterotrimeric G proteins have been determined by cryo-EM than by X-ray crystallography [3].

We will present how Sosei Heptares has integrated cryo-EM to our SBDD workflow, including the role of

proprietary mini G technology [4] for understanding ligand-induced GPCR activation and as a tool for

facilitating structure determination. Exploitation of cryo-EM is now providing additional opportunities to solve

structures of GPCRs for which limited tool ligands are available, which we will exemplify with cryo-EM

enabled GPCR SBDD case studies.

Cryo-EM has clearly improved accessibility of challenging receptors to SBDD. In addition to assisting hit and

lead generation for these valuable targets, the understanding of novel modes of binding opens new opportunities

for modulating receptor function in disease. The structural revolution for GPCRs is now revealing the

mechanistic details of this diverse range of orthosteric, allosteric and biased modulators and should drive SBDD

of new therapeutic agents.

 

References

1) Congreve M, Andrews SP, Doré AS, Hollenstein K, Hurrell E, Langmead CJ, Mason JS, Ng IW, Tehan B, Zhukov A,

Weir M, Marshall FH. Discovery of 1,2,4-triazine derivatives as adenosine A(2A) antagonists using structure-based drug

design. J Med Chem. 2012, 55: 1898-1903. doi: 10.1021/jm201376w

2) Christopher JA, Aves SJ, Bennett KA, Doré AS, Errey JC, Jazayeri A, Marshall FH, Okrasa K, Serrano-Vega MJ, Tehan

BG, Wiggin GR, Congreve M. Fragment and Structure-Based Drug Discovery for a Class C GPCR: Discovery of the mGlu5

Negative Allosteric Modulator HTL14242 (3-Chloro-5-[6-(5-fluoropyridin-2-yl)pyrimidin-4-yl]benzonitrile). J Med Chem.

2015, 58: 6653-6664. doi: 10.1021/acs.jmedchem.5b00892

3) Congreve M, de Graaf C, Swain NA, Tate CG. Impact of GPCR Structures on Drug Discovery. Cell. 2020. 181:81-91. doi:

10.1016/j.cell.2020.03.003

4) Nehmé R, Carpenter B, Singhal A, Strege A, Edwards PC, White CF, Du H, Grisshammer R, Tate CG. Mini-G proteins:

Novel tools for studying GPCRs in their active conformation. PLoS One. 2017, 12: e0175642. doi:

10.1371/journal.pone.0175642

EFMC-ISMC | 61

LE018

ALLOSTERIC REGULATION OF GTP CYCLOHYDROLASE I

Herbert Nar

Boehringer Ingelheim GmbH & Co. KG, Biberach, Germany

GTP Cyclohydrolase I (GCH1) is a homodecameric protein complex of approximately 250kDa molecular

weight. GCH1 catalyses the conversion of guanosine triphosphate (GTP) to dihydroneopterin triphosphate

(H2NTP). The proposed complex reaction involves the hydrolytic opening of the imidazole ring of GTP and the

formation of a formamido-pyrimidine intermediate. Release of formate, Amadori rearrangement of the ribose,

and closure of the dihydropyrazine ring affords the product. This reaction is the initiating step in the biosynthesis

of tetrahydrobiopterin (BH4) (1).

BH4 functions as co-factor in neurotransmitter biosynthesis. The BH4 biosynthetic pathway and GCH1 have

been identified as promising targets to treat pain disorders in patients (2).

The function of mammalian GCH1s is regulated by a metabolic sensing mechanism involving a regulator

protein, GCH1 feedback regulatory protein (GFRP). GFRP is a pentamer of 9.5kDa subunits, which binds to

GCH1 to form inhibited or activated complexes dependent on availability of co-factor ligands, BH4 and

phenylalanine, respectively (3).

We determined high resolution structures of human GCH1-GFRP complexes by cryoEM and X-ray

crystallography. cryoEM revealed structural flexibility of specific and relevant surface lining loops, which were

unresolved by X-ray crystallography due to crystal packing. The overall resolution of the cryoEM structures of

2.9 and 3.0Å allows for visualization of ligand binding to functional sites of the regulated complexes. Further,

we studied allosteric regulation of isolated GCH1 by X-ray crystallography. Using the combined structural

information we are able to obtain a comprehensive picture of the mechanism of allosteric regulation.

Local rearrangements in the allosteric pocket upon BH4 binding result in drastic changes in the quaternary

structure of the enzyme leading to a more compact, tense form of the inhibited protein and translocate to the

active site, leading to an open, more flexible structure of its surroundings. Inhibition of the enzymatic activity is

not a result of hindrance of substrate binding, but rather a consequence of accelerated substrate binding kinetics

as shown by STD-NMR and site-directed mutagenesis (4).

References

1) Nar, H. (2004). GTP cyclohydrolase I, Handbbok of Metalloproteins, John Wiley & Sons Ltd.

2) Tegeder, I., et al. (2006). GTP cyclohydrolase and tetrahydrobiopterin regulate pain sensitivity and persistence. Nat Med

12(11): 1269-1277

3) Harada, T., et al. (1993). Feedback regulation mechanisms for the control of GTP cyclohydrolase I activity. Science

260:1507–1510

4) Ebenhoch R., et al. (2021). A hybrid approach reveals the allosteric regulation of GTP cyclohydrolase. PNAS 117, 50,

31838-31849

62 | EFMC-ISMC

LE019

THE ROLE OF CRYO-EM IN FRAGMENT-BASED DRUG

DISCOVERY

Judith Reeks

Astex Pharmaceuticals, 436 Cambridge Science Park, Cambridge, CB4 0QA, UK

Structural biology is routinely used throughout the drug discovery process, from target and hit identification

through to clinical candidate selection. Structure-based drug design (SBDD) uses protein-ligand structures to

guide iterative cycles of design to produce potent lead molecules. Historically, X-ray crystallography has been

the method of choice for generating such structures. However, recent advances in cryo-EM have shown that it

too can be used to generate structures of sufficient quality to guide drug discovery. Certain protein classes that

are often refractory to crystallisation, such as membrane proteins and multiprotein complexes, have seen success

in cryo-EM, thus widening the pool of targets to which SBDD can be applied. In this talk the role of cryo-EM

for SBDD will be discussed, including the current state of the art and future perspectives. This will be

exemplified by the adoption of cryo-EM at Astex and our evaluation of the utility of cryo-EM for

fragment-based drug discovery (FBDD). (1)

References

1) Fragment-based drug discovery using cryo-EM. M. Saur, et al. Drug Discovery Today, 2019.

EFMC-ISMC | 63

LE020

STRUCTURE BASED DRUG DISCOVERY TARGETING

HELICOBACTER PYLORI, USING CRYO-EM

Eva S. Cunha (1), Xiaorui Chen (2,6), Marta Sanz-Gaitero (1), Deryck J. Mills (3), Hartmut Luecke

(1,2,4,5)

1) Structural Biology and Drug Discovery Group, Centre for Molecular Medicine Norway, Nordic EMBL Partnership,University of Oslo and Oslo University Hospital, 0318 Oslo, Norway

2) Department of Molecular Biology and Biochemistry, University of California, Irvine, CA 92697, USA3) Department of Structural Biology, Max Planck Institute of Biophysics, Frankfurt am Main, Germany

4) Department of Medical Biochemistry, University of Oslo and Oslo University Hospital, 0372 Oslo, Norway5) Department of Physiology and Biophysics, University of California, Irvine, CA 92697, USA

6) Genomics Research Center, Academia Sinica, 128 Academia Road, Sect. 2, Nankang District, Taipei, Taiwan

Infection of the human stomach by Helicobacter pylori remains a worldwide problem and greatly contributes to

peptic ulcer disease and gastric cancer. Without active intervention approximately 50% of the world population

will continue to be infected with this gastric pathogen. Current eradication, called triple therapy, entails a

proton-pump inhibitor and two broadband antibiotics, however resistance to either clarithromycin or

metronidazole is greater than 25% and rising. Therefore, there is an urgent need for a targeted, high-specificity

eradication drug. Gastric infection by H. pylori depends on the expression of a nickel-dependent urease in the

cytoplasm of the bacteria. We have solved a 2.0 Å resolution structure of the 1.1 MDa urease in complex with an

inhibitor by cryo-electron microscopy and compare it to a β-mercaptoethanol-inhibited structure at 2.5 Å

resolution. We also present a new map at 1.68 Å resolution with a novel inhibitor bound. The structural

information is of sufficient detail to aid in the development of inhibitors with high specificity and affinity.

64 | EFMC-ISMC

LE021

IDENTIFICATION OF POTENT AND LONG ACTING SINGLE-CHAIN

PEPTIDE MIMETICS OF HUMAN RELAXIN-2 FOR

CARDIOVASCULAR DISEASES

Sergio MALLART

Emerging Chemical Modalities, Integrated Drug Discovery, Sanofi R&D, Chilly Mazarin, France

In recent years, peptides have gained interest as therapeutic solutions. Their potency and specificity as well as

their intermediate size between small molecules and biologics have contributed to this success. Using natural

occurring peptides as starting points, and application of traditional rational design to improve their weaknesses,

such as their chemical and physical properties have led to numerous approved or clinical candidates. We will

present a multiparametric optimization of the B-chain of relaxin that led to the discovery of potent single chain

relaxin mimetics. Modifications in the B chain of relaxin, such as the introduction of specific mutations and the

trimming of the sequence to an optimal size resulted in potent, structurally simplified peptide agonists of the

relaxin receptor RXFP1. Introduction of suitable spacers and fatty acids, led to the identification of single chain

lipidated peptide agonists of RXFP1, with sub-nanomolar activity, high subcutaneous bioavailability, extended

half-lives and in vivoefficacy. These long lasting peptides represent a new class of RXFP1 agonists, compatible

with once daily subcutaneous administration in patients that could open the door to new treatments for chronic

fibrotic and cardiovascular diseases.

EFMC-ISMC | 65

LE022

DISCOVERY OF AFICAMTEN (CK-274): A NOVEL, SMALL

MOLECULE, CARDIAC MYOSIN INHIBITOR FOR THE POTENTIAL

TREATMENT OF HYPERTROPHIC CARDIOMYOPATHIES (HCM)

Grace Chuang, Peadar Cremin, James Hartman, Yangsong Wu, Darren Hwee, Jeanelle Zamora, Jingying

Wang, Laura Robertson, Fady Malik, Bradley Morgan

The underlying pathological hypertrophy and fibrosis in genetic hypertrophic cardiomyopathies (HCM) is likely

caused by hypercontractility of the cardiac sarcomere. A novel cardiac myosin inhibitor that directly decreases

cardiac muscle contractility, Aficamten (CK-274) was discovered starting with a unique hit from a high

through-put screen, compound 1.  The major optimization advancement came from the modification of

compound 1 to compound 12. The less restrictive structure-activity relationship of 12 allowed for the rapid

improvement of drug-like properties.  Key to the design of Aficamten was to provide a predicted human half-life

(t

1/2

)appropriate for once a day (q.d.) dosing, to reach steady state within two weeks, to have no substantial

cytochrome P450 (CYP) induction or inhibition, and to have a wide therapeutic window in vivo with a clear

pharmacokinetic (PK) / pharmacodynamic (PD) relationship.  Aficamten demonstrated a human t

1/2

similar to

predictions and was able to reach steady state concentration within the desired two-week window in a phase 1

clinical trial.  A phase 2 clinical trial of aficamtenin patients with symptomatic hypertrophic cardiomyopathy and

left ventricular outflow tract obstruction recently announced positive results supporting advancement into phase

3.

66 | EFMC-ISMC

LE023

MAP4K4 INHIBITORS FOR THE SUPPRESSION OF CARDIAC

MUSCLE CELL DEATH

Gary Newton (1), Kathryn Chapman (2), Rehan Aqil (2), Robert Yan (2), Mohamed Bellahcene (3),

Melanie Bayford (2), Nicholas Chapman (2), Nick Martin (2), Ashley Jarvis (2), Pelin Golforoush (3),

Lorna Fielder (3), Maaike te Lintel Hekkert (4), Daphne Merkus (4), Dirk Duncker (4), Trevor Perrior

(2), Michael Schneider (3)

1) Institute of Cancer Research, Sutton, UK2) Domainex, Chesterford Research Park, UK

3) National Heart and Lung Institute, Imperial College London, UK4) Erasmus MC, University Medical Center Rotterdam, The Netherlands

Heart disease is a paramount cause of global death and disability, most typically in the form of coronary artery

narrowing and its urgent manifestation as myocardial infarction (“heart attacks”). It has also emerged as a major

and growing alternative cause of death for cancer survivors, because of the cardiotoxicity of several routinely

used anti-cancer drugs. Notably, cardiac side effects are commonly seen in patients receiving anthracyclines,

such as doxorubicin, the anti-HER2 receptor antibody trastuzumab, tyrosine kinase inhibitors, immune

checkpoint inhibitors, and other agents. In short, cardiac muscle cell death plays an instrumental role in

mediating both acute and chronic heart failure, driving dysfunction of the heart as a biomechanical pump.

Despite this, few therapeutic approaches have sought to directly protect the cardiomyocytes by opposing the

pathways that are engaged by these initiators of cell death.  We have recently reported that genetic and

pharmacological inhibition of MAP4K4, a serine threonine kinase, protects rodent and human cardiomyocytes

from death induced by a variety of stimuli, including oxidative stress and anthracyclines [1-3]. Herein we

describe the development of a series of MAP4K4 inhibitors, members of which are suitable for either oral or

intravenous administration and which have subsequently been used to investigate the potential of MAP4K4

inhibition as a novel strategy for cardioprotection.

References

1) Fielder et al. MAP4K4 inhibition promotes survival of human stem cell-derived cardiomyocytes and reduces infarct size in

vivo, Cell Stem Cell, 2019, 24, 579-591

2) Golforoush et al. Selective protection of human cardiomyocytes from anthracycline cardiotoxicity by small molecule

inhibitors of MAP4K4, Sci Rep, 2020, 10, 12060

3) Te Lintel Hekkert et al. Preclinical trial of a MAP4K4 inhibitor to reduce infarct size in the pig: does cardioprotection in

human stem cell derived myocytes predict success in large mammals?, Basic Research in Cardiology, 2021, 116, 34

EFMC-ISMC | 67

LE024

DEVELOPMENT OF NON-CODING RNA BASED

NEXT-GENERATION HEART FAILURE THERAPEUTICS

Thomas Thum

Director, Hannover Medical School and Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover,Germany

Despite some progress in cardiac pharmacotherapies, heart failure (HF) is a growing pandemic with increasing

burden. Treatments are mainly symptomatic and only few are based on the root of the disease. MicroRNAs

(miRNA) are transcriptional regulators and essential drivers of disease progression. We previously demonstrated

that miR-132 is both necessary and sufficient to drive the pathological cardiomyocytes growth, a hallmark of

adverse cardiac remodelling. We recently showed translational evidence for an optimized, synthetic locked

nucleic acid antisense oligonucleotide inhibitor (antimiR-132) for heart failure therapy in 3 large animal models

of subacute and chronic heart failure. We demonstrated favourable pharmacokinetics, safety, tolerability,

dose-dependent PK/PD relationships and high clinical potential for the antimiR-132 treatment scheme. Recently

we completed a world-wide first study testing an antisense molecule in heart failure patients. The miR-132

blocker CDR132L was safe and well tolerated, confirmed linear plasma pharmacokinetics with no signs of

accumulation, and suggested cardiac functional improvements. Although this study was limited by the small

patient numbers, the indicative efficacy of this drug is very encouraging justifying additional clinical studies to

confirm the beneficial CDR132L pharmacodynamic effects for the treatment of HF.

68 | EFMC-ISMC

LE025

DESIGN AND PRECLINICAL CHARACTERIZATION PROGRAM

TOWARDS BAY 2433334, AN ORAL FACTOR XIa INHIBITOR FOR

THE PREVENTION AND TREATMENT OF THROMBOEMBOLIC

DISORDERS

Susanne Roehrig

Bayer AG, R&D Pharmaceuticals, Wuppertal, Germany

Coagulation factor XIa (FXIa) contributes to the development of thrombosis but plays only a minor role in

hemostasis. Inhibition of FXIa may therefore have the potential to reduce the risk for thrombosis without

increasing the risk for bleeding which would offer antithrombotic protection to a broader patient population and

to new patient groups. For chronic treatment, reversible and orally available FXIa inhibitors are particularly

desirable but despite significant efforts and time, the research aiming for such FXIa inhibitors has been a

challenge.

In this paper, we describe our drug discovery efforts leading to the identification of BAY 2433334, a potent,

direct and reversible inhibitor of FXIa for oral treatment. Its potential as anticoagulant with a broad therapeutic

window is currently being investigated in a phase 2 development program.

EFMC-ISMC | 69

LE026

FIRST TIME DISCLOSURE OF BI 1265162, AN ENaC INHIBITOR FOR

THE TREATMENT OF CYSTIC FIBROSIS

Jörg Thomas Kley (1), Sara Frattini (2), Dieter Hamprecht (2), Armin Heckel (1), Jörg Hehn (1), Thomas

Veser (1), Dieter Wiedenmayer (1)

1) Boehringer Ingelheim Pharma, Medicinal Chemistry Germany, Biberach, Germany2) Boehringer Ingelheim Research Italy, Milan, Italy

The epithelial sodium channel (ENaC) is an important regulator of airway surface liquid volume and it is

hyperactivated in cystic fibrosis (CF)

1

. ENaC inhibition is a potential strategy for a mutation-agnostic therapy in

CF, alone and in synergy with CFTR modulators

2

. The ENaC inhibitor BI 1265162 has shown safety in phase I

studies

3

and entered clinical phase II.

This talk will discuss the requirements for an inhaled ENaC inhibitor for the treatment of CF. Our approaches to

translate these requirements into compound design eventually lead to the discovery of BI 1265162.

While our starting points already showed attractive in vitro potency, we had to overcome numerous hurdles

during optimization, among them achieving

-          high in vivo potency and efficacy

-          high target selectivity

-          high and lasting lung exposure, but no relevant effect on ENaC in kidney

-          low permeability and minimal oral bioavailability

-          formidable aqueous solubility and hydrolytic stability allowing administration via the Respimat® Soft

Mist™ inhaler

-          a crystalline and non-hygroscopic salt form

The successful move of BI 1265162 into clinical phase II highlights the value of considering non-standard

structural motifs to tackle specific problems in multi-parameter optimization.

 

References

1) Patrick J. Moore & Robert Tarran. The epithelial sodium channel (ENaC) as a therapeutic target for cystic fibrosis lung

disease. Expert Opinion on Therapeutic Targets (2018), 22(8), 687-701

2) Mall MA. ENaC inhibition in cystic fibrosis: potential role in the new era of CFTR modulator therapies. Eur Respir J

(2020), 56(6), 2000946

3) Mackie A, Rascher J, Schmid M, et al. First clinical trials of the inhaled ENaC inhibitor BI 1265162 in healthy volunteers.

ERJ Open Res (2021), 7(1)

70 | EFMC-ISMC

LE027

DISCOVERY OF THE CLINICAL CANDIDATE AZD4604, A POTENT

AND SELECTIVE JANUS KINASE 1 INHIBITOR, AS AN INHALED

TREATMENT FOR RESPIRATORY DISEASE

Magnus Nilsson (1), Kristina Berggren (1), Susanne Berglund (1), Göran Dahl (2), David Elmqvist (3),

Ramon Hendrickx (4), Johan Johansson (1), Matti Lepistö (1), Amir Smailagic (5), James Pinkerton (6),

Torben Österlund (7)

1) Medicinal Chemistry, Respiratory & Immunology (R&I), Research & Early Development, BioPharmaceuticals R&D,AstraZeneca, Gothenburg, Sweden.

2) Biophysics and FBLG, Discovery Sciences, Research & Early Development, BioPharmaceuticals R&D, AstraZeneca,Gothenburg, Sweden.

3) Formulation & Biopharmaceutics, RIA Inhaled PDU, Research & Early Development, BioPharmaceuticals R&D,AstraZeneca, Gothenburg, Sweden.

4) DMPK, Respiratory & Immunology (R&I), Research & Early Development, BioPharmaceuticals R&D, AstraZeneca,Gothenburg, Sweden.

5) Bioscience cough and in vivo, Respiratory & Immunology (R&I), Research & Early Development, BioPharmaceuticalsR&D, AstraZeneca, Gothenburg, Sweden.

6) Respiratory Pharmacology Group, Division of Airway Disease, NHLI, Imperial College London, United Kingdom7) Discovery Biology, Discovery Sciences, Research & Early Development, BioPharmaceuticals R&D, AstraZeneca,

Gothenburg, Sweden.

Aberrant Janus Kinase (JAK) signal transduction and activation of transcription (STAT) signaling is implicated

in several key inflammatory pathways associated with asthma. Potent inhibition of JAK1 blocks

pSTAT-mediated signaling which has broad anti-inflammatory effects through inhibition of these

JAK1-dependent cytokine signaling pathways. Here we describe the optimization leading to AstraZeneca

compound AZD4604, an inhaled JAK1-selective inhibitor as a potential treatment of asthma. AZD4604 has a

differentiated lung retention driver as compared to a second inhaled JAK1-selective inhibitor candidate

(AZD0449). The optimization was guided by physicochemical properties, such as solubility, lipophilicity,

crystallinity and melting point, while maintaining high potency and selectivity to the primary target. By tuning

these properties for increased tissue binding, we could transform a weakly basic lead compound, originating

from a separate lead series intended for oral dosing, into AZD4604 (1).

In vivo lung-PK profiles upon dry powder inhalation indicated good lung retention and favourable lung to

plasma exposures, corroborating suitable properties for inhaled dosing. Target engagement was also

demonstrated in a pre-clinical rodent model of allergic asthma (2). AZD4604 will be tested in a Phase 1 clinical

trial (NCT04769869).

References

1) Qibin Su et al., Discovery of (2R)‑N‑[3-[2-[(3-Methoxy-1-methyl-pyrazol-4-

yl)amino]pyrimidin-4-yl]‑1H‑indol-7-yl]-2-(4-methylpiperazin-1- yl)propenamide (AZD4205) as a Potent and Selective

Janus Kinase 1 Inhibitor. J. Med. Chem. 2020, 63, 4517-4527. https://dx.doi.org/10.1021/acs.jmedchem.9b01392

2) James Pinkerton et al., Profiling the impact of two JAK inhibitors in a pre-clinical model of allergic asthma. European

Respiratory Journal 2020; 56: Suppl. 64, 3302. https://dx.doi.org/10.1183/13993003.congress-2020.3302

EFMC-ISMC | 71

LE028

CHEMICAL PROBES TO EXPLORE CANCER BIOLOGY AND

VALIDATE DRUG TARGETS

Paul Workman

Institute of Cancer ResearchHaddow Laboratories

15 Cotswold RoadSM2 5NG LondonUnited Kingdom

In my keynote lecture, I will cover four topics:

� Importance and essential properties of high-quality chemical probes

� Need for outreach to biomedical researchers and resources available

� Examples of the use of chemical probes and controlled polypharmacology from my own research in

oncology drug discovery – focusing on overcoming cancer evolution and drug resistance

� Recent research to drug oncogenic transcription factors

Here, my definition of a chemical probe is a well-characterised small-molecule compound that can be used to

modulate the activity of a target protein and investigate with confidence the effects of that target protein on

biological and disease phenotypes. Also referred to as chemical tools and pharmacological probes – and most

commonly acting as inhibitors – chemical probes are valuable reagents for application in fundamental biology

and target validation. But to be effective they must be of high quality, particularly showing high potency and

selectivity, evidence of target modulation in cells, and for application in vivo – for example in mice and rats –

they must also have adequate PK/PD properties to ensure suitable exposure to free (unbound) compound. These

required features are very well known to the expert community of medicinal chemists, drug discovery biologists

and chemical biologists, but understanding across the broad biomedical research community is very low. Despite

the increasing usage of chemical probes, choice of poor quality compounds and misuse of chemical probes

remains very frequent and systemically problematic. Thus alongside the continued drive for greater numbers of

high quality probes for a higher proportion of the proteome, it is equally important to reach out to and educate the

broader research community, journal editors and funders. Resources and programmes I am involved with – such

as the Chemical Probes Portal (https://new.chemicalprobes.org), Probe Miner (https://probeminer.icr.ac.uk),

canSAR (https://cansarblack.icr.ac.uk) and the Target 2035 federation (https://www.target2035.net) – are playing

a valuable role in driving best practice while expanding proteome coverage.

In collaborative studies, we have progressed to a first-in-child clinical trial evaluation of the dual-selective

cyclin-dependent kinase (CDK) 9/2 inhibitor fadraciclib (CYC065). This drug was discovered by our ICR team

in collaboration with Cyclacel. We then discovered its therapeutic potential in models of aggressive paediatric

neuroblastoma that is driven by the pathogenic transcription factor, MYCN. MYCN is a highly validated target

but hard to drug directly. However, its expression is decreased by fadraciclib’s inhibition of the transcriptional

kinase CDK9. The additional inhibition of CDK2 by fadraciclib induces apoptotic cell death in MYCN

neuroblastoma cells and produces prolonged tumour response and survival in mouse models. Another

transcription factor that we are seeking to drug is brachyury which is pathogenic in the spinal tumour, chordoma.

We are evaluating fadraciclib and in collaboration with the Structural Genomics Consortium and Chordoma/Mark

Foundation we have identified fragments with potential for elaboration as inhibitors or degraders.

Following our discovery of the Heat Shock Protein 90 molecular chaperone inhibitors and progression of

luminespib to the clinic (with Vernalis and Novartis) we switched our attention to discover inhibitors of the

transcription factor Heat Shock Factor 1 or HSF1. HSF1 plays an important role in oncogenesis and the

maintenance of the cancer state. HSF1 is itself hard to drug directly and so we adopted a phenotypic screening

approach. I will describe our more than fourteen-year journey from the initial screen to the discovery of a

chemical probe and then a clinical candidate HSF1 pathway inhibitor that shows particular potential for the

treatment of ovarian clear cell carcinoma and multiple myeloma.

72 | EFMC-ISMC

LE029

CHEMICAL PROBES TO STUDY LIPID SIGNALING

Mario van der Stelt

Leiden University & Oncode Institute, Einsteinweg 55, Leiden, The Netherlands

Lipid transmitters, such as endocannabinoids and eicosanoids, play important roles in the central nervous system

and regulate physiological processes in health and disease that include pain, emotion, addition and

neuroinflammation. Chemical probes that perturb lipid transmitter biosynthesis and metabolism are needed to

understand the function of these pathways in the nervous system. Here, I will present our work on the

development of chemical probes to study endocannabinoid biology. I will discuss how they can be used in

various stages of drug discovery from target validation to profiling of clinical candidates. (1 - 4)

References

1) Ogasawara et al., Proc. Natl. Acad. Sci. USA, 2016, 113, 26

2) Van Esbroeck et al., Science, 2017, 356, 1084

3) Mock et al., Naturę Chem. Biol., 2020, 16, 667

4) Mock et al., J. Med. Chem., 2021, 64, 481

EFMC-ISMC | 73

LE030

DISCOVERY OF MRK-740, A FIRST-IN-CLASS, POTENT, SELECTIVE

AND CELL ACTIVE PRDM9 CHEMICAL PROBE

Christian Fischer

Discovery Chemistry, Merck & Co., Inc., Boston, Massachusetts, USA

PRDM9 is a PR domain containing protein which trimethylates histone 3 on lysine 4 and 36. Its normal

expression is restricted to germ cells and attenuation of its activity results in altered meiotic gene transcription,

impairment of double-stranded breaks and pairing between homologous chromosomes. There is growing

evidence for a role of aberrant expression of PRDM9 in oncogenesis and genome instability. In this presentation

we will detail our screening, hit finding and lead optimization efforts leading to the discovery of MRK-740, a

first-in-class, potent, selective and cell active PRDM9 inhibitor.  This project was pursued in close collaboration

between the SGC in Toronto and MSD where we utilized our complementary capabilities to accelerate the

development of chemical matter which had the ability to inhibit this previously untouched clade of the

methyltransferase family.  We initially identified several potential starting points through screening MSD

diversity and focused libraries, however, we quickly began to focus on a particularly promising series of

compounds, where we were able to rapidly gain biochemical potency, albeit at the expense of overall properties,

cellular activity and off-target profile.  A complete re-optimization of the scaffold focusing on predicted and

measured properties was ultimately required to install all needed properties into our molecules and to arrive at

our candidate chemical probe MRK-740, as well as a closely related inactive control compound.

Subsequently, researchers at the SGC were able to obtain an x-ray co-crystal structure of MRK-740 in PRDM9

which displayed several intriguing and unanticipated elements.  MRK-740 was found to bind in the

substrate-binding pocket, but with unusually extensive interactions with the cofactor S-adenosylmethionine

(SAM), conferring SAM-dependent substrate competitive inhibition. To the best of our knowledge, such a

binding mode has to date not been described for an inhibitor of any SAM-utilizing enzyme.  We will review the

biostructural information and discuss hypotheses how this may be responsible for the exquisite selectivity of

MRK-740.

In summary we will report the joint SGC-MSD discovery of the first potent and selective PRDM9 inhibitor and

its negative control.

74 | EFMC-ISMC

LE031

QUENCHED ACTIVITY-BASED PROBES AS NEW CHEMICAL

TOOLS TO ANALYSE RESISTANCE TO ANTIBIOTICS

Rita Félix (1), Ana Mallo-Abreu (2,3), Luís A. Carvalho (1), Carlos A. M. Afonso (1), Rui Moreira (1)

1) iMed.ULisboa, Faculdade de Farmácia da Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal2) Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS)

3) Departamento de Química Orgánica, Facultade de Farmacia. Universidade de Santiago de Compostela, 15782. Santiagode Compostela, Spain

Beta-lactamases comprise several serine and metallo-hydrolases that are responsible for the bacterial resistance

to beta-lactam antibiotics (BLAs), thus posing a serious threat to the treatment of bacterial infections [1]. Since

pathogens have evolved to express previously rare or unknown beta-lactamases, serine-based enzymes have

become the subject of several medicinal chemistry campaigns, highlighting the urgent need for developing new

broad spectrum enzyme inhibitors. There has been an increasing interest in developing methods to profile

enzyme activity, identifying new therapeutic targets and biomarkers, and further understanding their molecular

mechanisms.

Quenched activity-based probes (qABP) are molecules that contain in their structure a fluorophore (F) and a

quencher (Q), covalently tagging active enzymes but not their inactive form. In this particular case, qABP only

shows fluorescence when it is linked to the enzyme, working as a mechanism-based (suicide) inhibitor (Fig 1).

In this work we report the development and optimization of a synthetic methodology to obtain several qABPs,

with different reactivity with the target enzyme and using different linkers and quenchers. The synthesized

qABPs showed an increase of the quantum fluorescence yield after the reaction with sodium hydroxide reaching

a final QY up to 92%. The synthesized qABPs are currently being tested in gel-based studies to understand the

activity against beta-lactamase, and other serine hydrolases. In order to detect beta-lactamase in biological

matrices,gel-based studies using HEK cells  lysates and multidrug-resistant bacteria lysates will be further

performed.

Figure 1 - qABP targeting beta-lactamase mechanism and fluorescence spectrum of qABPs before and after

reaction with NaOH.

Thanks to FCT through by the funding grants SFRH/BD/137459/2018 andPTDC/BBB-BEP/2463/2014.

References

1) Bush, K., International Journal of Antimicrobial Agents, 2015, 46, 483-493.

2) Biondi, S; Long S; Panunzio M; Qin WL., Current Medicinal Chemistry, 2011, 18, 4223-4236.

3) Hu, M., Multicolor, Journal of American Chemical Society, 2011, 113, 12009-12020.

EFMC-ISMC | 75

LE032

RECENT DEVELOPMENTS IN STRATEGIES AND TACTICS

TOWARDS COMPLEX SECONDARY METABOLITES

Erick M. Carreira

Department of Chemistry and Applied Biosciences, ETH-Zürich, Switzerlandwww.carreira.ethz.ch

The talk will include discussion and analysis of recent natural product targets that have been synthesized in the

group.  It will focus on target oriented synthesis as an engine for the generation of novel methods and approaches

to bioactive agents.  The methods involve novel, unexpected reactivity and unusual building blocks that are fully

integrated to lead to efficient routes.

 

76 | EFMC-ISMC

LE033

INNOVATIVE DNA-BASED ASYMMETRIC CATALYSIS

Stellios Arseniyadis (1), Nicolas Duchemin (1), Sidonie Aubert (1), Jinlei Zhang (1), Michael Smietana (2)

1) Department of Chemistry, Queen Mary University of LondonMile End Road, E1 4NS London (UK)https://arseniyadislab.sbcs.qmul.ac.uk

2) Institut des Biomolécules Max Mousseron, Université de MontpellierPlace Eugène Bataillon, 34095 Montpellier (France)

DNA-based artificial metalloenzymes have recently drawn considerable attention because of their unique

features that comprise a chemically stable chiral double helix associated with many programmable secondary

structures. Since the pioneering work of Roelfes and Feringa [1], the field of DNA-based asymmetric catalysis

has been thriving resulting in the development of a handful of highly selective synthetic transformations by

several groups [2] including ours [3]. The concept of DNA-based asymmetric catalysis lies in embedding an

achiral transition metal catalyst within a DNA double helix that provides the necessary chiral microenvironment

to induce enantioselectivity. The most recent efforts to unveil new reactivities have been accompanied with the

willingness to understand the mechanisms by which the chirality is transferred. I’ll present some of the group’s

most recent results.

 

References

1) Roelfes, G.; Feringa, B. L. Angew. Chem. Int. Ed. 2005, 44, 3230.

2) (a) Drienovska, I.; Roelfes, G. Isr. J. Chem. 2015, 55, 21. (b) Park, S.; Sugiyama, H. Molecules 2012, 17, 12792. (c)

Boersma A. J.; Megens, R. P.; Feringa, B. L.; Roelfes, G. Chem. Soc. Rev. 2010, 39, 2083.

3) (a) Mansot, J.; Lauberteaux, J.; Lebrun, A.; Mauduit, M.; Vasseur, J.-J.; Marcia de Figueiredo, R.; Arseniyadis, S.;

Campagne, J.-M.; Smietana, M. Chem. Eur. J. 2020, 26, 3519. (b) Mansot, J.; Aubert, S.; Duchemin, N.; Vasseur, J.-J.;

Arseniyadis, S.; Smietana, M. Chem. Sci. 2019, 2875. (c) Duchemin, N.; Skiredj, A.; Mansot, J.; Leblanc, K.; Vasseur, J.-J.;

Beniddir, M. A.; Evanno, L.; Poupon, E.; Smietana, M.; Arseniyadis, S. Angew. Chem. Int. Ed. 2018, 57, 11786. (d)

Duchemin, N.; Benedetti, E.; Bethge, L.; Vonhoff, S.; Klussmann, S.; Vasseur, J.-J.; Cossy, J.; Smietana, M.; Arseniyadis, S.

Chem. Commun. 2016, 52, 8604. (e) Amirbekyan, K.; Duchemin, N.; Benedetti, E.; Joseph, R.; Colon, A.; Markarian, S. A.;

Bethge, L.; Vonhoff, S.; Klussmann, S.; Cossy, J.; Vasseur, J.-J.; Arseniyadis, S.; Smietana, M. ACS Catal. 2016, 6, 3096. (f)

Benedetti, E.; Duchemin, N.; Bethge, L.; Vonhoff, S.; Klussmann, S.; Vasseur, J.-J.; Cossy, J.; Smietana, M.; Arseniyadis, S.

Chem. Commun. 2015, 51, 6076. (g) Wang, J.; Benedetti, E.; Bethge, L.; Vonhoff, S.; Klussmann, S.; Vasseur, J.-J.; Cossy,

J.; Smietana, M.; Arseniyadis, S. Angew. Chem. Int. Ed. 2013, 52, 11546.

EFMC-ISMC | 77

LE034

SCAFFOLD-ASSISTED PEPTIDE CYCLIZATIONS: TOWARDS

PROTEIN MIMICS

Gaston Richelle, Dieuwertje Streefkerk, Timo Nuijens, Peter Timmerman, Jan van Maarseveen

University of AmsterdamScience Park 904, 1098XH Amsterdam

The Netherlands

In nature, multicyclic peptides constitute a versatile molecule class with various biological functions. For their

pharmaceutical exploitation, orthogonal chemical methodologies that enable selective consecutive

macrocyclizations are required. It will be presented that a combination of CLIPS alkylation, enzymatic

macrocyclization, and CuAAC macrocyclization or oxime ligation provides access up to hexacyclic

peptides. For this, we designed multivalent scaffolds containing two benzyl bromides and two alkyne or

alkoxyamine functionalities that react via CLIPS/CuAAC/Oxime ligation reactions with cysteines and

azides/aldehydes in the peptide. As will be discussed, it became clear that for the exclusive formation of

isomerically pure products flexible small molecular scaffolds are required. There seems no practical limitation to

the loop size and type of canonical amino acids present.

78 | EFMC-ISMC

LE035

SYNTHESIS OF NEW BUILDING BLOCKS FROM THE CHEMICAL

UNIVERSE DATABASE GDB

Aline L. Carrel, Prof. Dr. Jean-Louis Reymond

University of Bern, Freiestrasse 3, 3012 Bern/CH

Drug discovery is in constant need of new molecules to develop drugs addressing unmet medical needs. To

assess the chemical space available for drug design our group developed tools to enumerate, visualize and search

chemical space. The Generated DataBases (GDBs) list billions of possible organic small molecules following

simple rules of chemical stability and synthetic accessibility. [1a-1d] These databases are treasure troves of new

molecules because the vast majority of GDB molecules (>99%) is unknown. Much of the originality and novelty

of GDB molecules lies in new combinations of rings including 3D-shaped saturated ring systems, often

containing chiral and quarternary centers. Molecules showcasing these properties have recently been recognized

as desirable building blocks for drug candidates. [2] This has been demonstrated in our group by the synthesis of

Triquinazine, a selective inhibitor of Janus Kinase I related to the marketed drug Tofacitinib. [3]

To identify interesting ring systems for drug design we analyzed the GDB4c database containing 916’130

possible ring systems. We focused our attention on novel aliphatic bicyclic diamines which represent interesting

building blocks for medicinal chemistry. Here we present versatile synthetic strategies to access libraries of

fused and spirocyclic diamines starting from simple commercially available building blocks.

References

1) a) T. Fink, J.-L. Reymond, J. Chem. Inf. Model. 2007, 47, 342-353. b) L. C. Blum, J.-L. Reymond, J. Am. Chem. Soc.

2009, 131, 8732-8733. c) L. Ruddigkeit, R. van Deursen, L. C, Blum, J.-L. Reymond, J. Chem. Inf. Model. 2012, 52,

2864-2875. d) R. Visini, J. Arús-Pous, M. Awale, J.-L. Reymond, J. Chem. Inf. Model. 2017, 57, 2707-2718.

2) O. O. Grygorenko, D. S. Radchenko, D. M. Volochnyuk, A. A. Tolmachev, I. V. Komarov, Chem. Rev. 2011, 111,

5506-5568.

3) K. Meier, J. Arús-Pous, J.-L. Reymond, Angew. Chem. Int. Ed., 2021, 60, 2704-2077.

EFMC-ISMC | 79

LE036

DISCOVERY OF A NOVEL CLASS OF SMALL MOLECULE DENGUE

VIRUS INHIBITORS

Tim H. M. Jonckers

Janssen Pharmaceutica, Medicinal Chemistry, Discovery Product Development and Supply, Janssen Research &Development, Turnhoutseweg 30, 2340 Beerse, Belgium

Although a vaccine is now approved for the prevention of dengue virus in individuals aged 9 to 45 years, it’s use

is limited to those people that had a previous dengue exposure confirmed by a point-of-care diagnostic test. In

addition, small molecule antivirals useful for treatment or prophylaxis of dengue are non-existent at this stage.

We previously reported on the identification of a novel series of 3-acyl-indole derivatives as promising dengue

virus inhibitors. Here, we present a series of compounds that display improved pan-serotype antiviral efficacy,

chiral stability and optimized in vitro ADME properties, as well as a dose dependent PK/PD relationship against

DENV-2 in an AG129 mouse model.

80 | EFMC-ISMC

LE037

ANTIVIRALS AGAINST CHIKUNGUNYA VIRUS: ANALYZING THE

CURRENT SITUATION TO IDENTIFY NEW OPPORTUNITIES

María-Jesús Pérez-Pérez

Instituto de Química MédicaJuan de la Cierva, 3

28006 MadridSpain

Chikungunya virus (CHIKV) is a re-emerging alphavirus transmitted to humans by the bites of infected Aedesmosquitos. The morbidity associated to CHIKV infections, with million of cases in tropical and subtropical

areas, has an increasing impact on health with important social and economic consequences. Large phenotypic

screening campaigns and drug repurposing approaches have identified active compounds against CHIKV

replication, in a few cases reaching clinical trials, although no drug has been approved yet.

1

Fortunately, our

knowledge at the structural level and at the functional role of some of the clue non-structural proteins required

for viral replication is increasing in the latest two years. Also significant advances are being performed in

clarifying the interactions of the virus with the host.

Our research group, in close collaboration with different partners, has described

3-aryl-triazolo[3,4-d]pyrimidines as the first nsP1 inhibitors of CHIKV.

2–4

The viral nsP1 possess

methyltransferase (MTase) and guanylyltransferase (GTase) activities required for the 5´-capping of the nascent

mRNAs, a crucial step for viral replication.

5

Very recently, it has been shown that nsP1 is highly palmitoylated

and serves as the anchoring of the replication complex to cellular membranes.

6

Indeed CHIKV infection

remodelates the plasma membrane creating invaginations known as spherules, where the new RNA is

synthesized. Also very recently, the cryo-EM structure of CHIKV nsP1 has been described, showing its

assembly in a dodecameric ring, where the ring shape favors the exit of the capped mRNA.

7

These important

advances related to nsP1 open new opportunities for antiviral intervention.

 

Acknowledgments. Our research on arbovirus is supported by the Spanish AEI (PID2019-105117RR-C22).

 

 

References

1) Pérez-Pérez, M. J.; Delang, L.; Ng, L. F. P.; et al. Expert Opin. Drug Discov. 2019, 14, 855–866.

https://doi.org/10.1080/17460441.2019.1629413

2) Gómez-SanJuan, A.; Gamo, A.-M.; Delang, L.; et al. ACS Infect. Dis. 2018, 4, 605–619.

https://doi.org/10.1021/acsinfecdis.7b00219.

3) Gigante, A.; Gómez-SanJuan, A.; Delang, L.; et al. Antiviral Res. 2017, 144, 216–222.

https://doi.org/10.1016/j.antiviral.2017.06.003.

4) Gigante, A.; Canela, M.-D.; Delang, L.;et al. J. Med. Chem. 2014, 57, 4000–4008. https://doi.org/10.1021/jm401844c.

5) Delang, L.; Li, C.; Tas, A.; et al.. Sci. Rep. 2016, 6, 31819. https://doi.org/10.1038/srep31819

6) Bakhache, W.; Neyret, A.; Bernard, E et al. J. Virol. 2020, 94, 1–20. https://doi.org/10.1128/JVI.02183-19.

7) Jones, R.; Bragagnolo, G.; Arranz, R.; Reguera, J. Nature 2021, 589, 615–619.

https://doi.org/10.1038/s41586-020-3036-8.

EFMC-ISMC | 81

LE038

ARTIFICIAL INTELLIGENCE FOR EMERGING AND NEGLECTED

DISEASES DRUG DISCOVERY

Carolina Horta Andrade

LabMol - Laboratory for Molecular Modeling and Drug Design, Faculdade de Farmácia, Universidade Federal de Goiás,Goiânia, GO, Brazil.

Only ~1% of all drug candidates against Neglected Tropical Diseases (NTDs) have reached clinical trials in the

last decades, underscoring the need for new, safer and effective treatments for these debilitating group of

diseases. Artificial intelligence (AI) is a cutting-edge area of computational research that allows rapid

identification of potentially active compounds with appropriate pharmacokinetic and toxicological properties,

shortening the drug discovery process while leading to a higher success rate and reducing  costs. Thus,

combining drug discovery and AI approaches has the potential to transform drug discovery from a slow,

sequential and high-risk process to a fast, integrated model with diminished risk of failure. In this talk, we will

present the development and application of AI and computational approaches such as structure-based drug

design (SBDD) and ligand-based drug design (LBDD) to accelerate drug discovery for the treatment of NTDs

and Emerging Diseases, such as Zika and COVID-19, by the identification of hits suitable for optimization.

82 | EFMC-ISMC

LE039

INNOVATIVE DIHYDROOROTATE DEHYDROGENASE CLINICAL

READY INHIBITORS AS PAN-CORONAVIRUS ANTIVIRALS:

TARGETING THE UNEXPECTED WITH INNOVATION

M. L. Lolli (1), S. Sainas (1), A. Luganini (2), A. Calistri (3), G. Sibille (2), B. Mognetti (2), V. Conciatori

(3), C. Del Vecchio (3), M. Giorgis (1), A. C. Pippione (1), R. Bagnati (5), A. Passoni (5), P. Circosta (4), V.

Gaidano (4), A. Cignetti (4), G. Saglio (4), C. Parolin (3), D. Boschi (1), G. Gribaudo (2)

1) Department of Science and Drug Technology, University of Turin, Turin (IT).2) Department of Life Sciences and Systems Biology, University of Turin, Turin (IT).

3) University of Padua, Padua (IT).4) Department of Clinical and Biological Sciences, University of Turin, Turin (IT).

5) Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan (IT).

The devastating effects of the new COronaVIrus Disease (COVID-19) have taught the world the weakness and

general unpreparedness in tackling such a viral pandemic. SARS and MERS outbreaks indeed indicate that

SARS-CoV-2 is not the first zoonotic coronavirus (CoVs) that humanity has to face, and it would likely not be

the last. Already at the beginning of a novel CoVs emergence, effective antivirals should be available to slow

down the spread of infection, saving life, and gaining time while waiting for the specific CoV vaccine

development. Thus, pan-CoVs antivirals effective for the management of SARS-CoV-2 infection and that can

be rapidly deployed against future emerging CoVs are urgently needed. Host-Targeting Antivirals (HTAs), by

targeting host biochemical pathways and cellular factors hijacked by CoVs for their replication, are targets of

choice for the development of broad-spectrum agents effective against the pandemic virus, even its

vaccine-escaping mutants, or in and future epidemic or pandemic CoVs threat. To contribute to this

preparedness, here we present the pathway that allowed the discovery of a class of potent human dihydroorotatedehydrogenase (hDHODH) inhibitors based on the hydroxypyrazole[1,5-a]pyridine scaffold [1]. Among this

class of molecules, MEDS433 has been shown to be a potent HTA and it is being developed until preclinical

level. By affecting the host de novo pyrimidine biosynthesis, MEDS433 in fact inhibits the in vitro replication of

several hCoVs, such as SARS-CoV-2 [2], hCoV-OC43 and hCoV-229E, as well as a large virus panel [3], with

EC

50

s always in the low nM range and an incredibly effective Safety Index (SI) (see figure). Due to the favorable

PK and toxicity in mice, MEDS433 is proposed as dual acting pan-CoVs HTA, effective against SARS-CoV-2

and other CoVs infections by directly inhibiting virus replication through pyrimidine depletion and by

stimulating the antiviral innate immune responses. In order to pave its future entrance in clinical trials for

COVID-19, drug combination strategies to enhance MEDS433’s antiviral effectiveness are also being explored.

References

1) Stefano Sainas, et al Targeting Acute Myelogenous Leukemia using potent human dihydroorotate dehydrogenase

inhibitors based on the 2-hydroxypyrazolo[1,5-a]pyridine scaffold: SAR of the biphenyl moiety. J Med Chem 2021, in press.

2) Calistri, A.; et al The new generation hDHODH inhibitor MEDS433 hinders the in vitro replication of SARS-CoV-2.

bioRxiv 2020, 2020.12.06.412759.

3) Anna Luganini, et al Effective deploying of a novel DHODH inhibitor against herpes simplex type 1 and type 2

replication. Antiviral Research 2021, 189, 105057;

EFMC-ISMC | 83

LE040

A CHEMICAL APPROACH TO CONTROLLING CELL FATE

Sheng Ding

School of Pharmaceutical Sciences, Tsinghua UniversityBeijing, China

Recent advances in stem cell biology may make possible new approaches for the treatment of a number of

diseases. A better understanding of molecular mechanisms that control stem cell fate as well as an improved

ability to manipulate them are required. Toward these goals, we have developed and implemented high

throughput cell-based screenings of chemical libraries, and identified and further characterized small molecules

that can control stem cell fate in various systems. This talk will provide latest examples of discovery efforts in

my lab that have advanced our ability and understanding toward controlling stem cell fate, including

self-renewal, survival, differentiation and reprogramming of stem cells.

84 | EFMC-ISMC

LE041

DISCOVERY OF SMALL MOLECULES TO MANIPULATE CELL

FATE IN VIVO: TOWARDS NEW THERAPIES FOR DEGENERATIVE

DISEASES

Angela J. Russell

Departments of Chemistry and Pharmacology, Chemistry Research Laboratory, University of Oxford, Mansfield Road,Oxford OX1 3TA, U.K.

With an increasingly ageing population, chronic diseases including cancers, dementia and heart failure are

placing huge demand on society and healthcare services. Regenerative medicine approaches seek to transform

healthcare management strategies, improving outcomes for patients suffering from degenerative diseases.  Over

the past two decades the majority of studies have been focussed on the transplantation of therapeutic

cells.  Several thousands of clinical trials have been conducted involving cell transplantation and while there

have been signs of efficacy in some cases, major hurdles exist to the routine adoption of such therapies in the

clinic.  Moreover, we now understand that in most cases these cells act not as a cell replacement therapy but

rather through the stimulation of endogenous repair pathways already present within the body.  This has opened

up a whole new avenue of research in the development of agents to directly stimulate these tissue repair and

regeneration processes in the treatment of chronic degenerative diseases and injury, negating the need for cell

transplantation.  

The field of drug discovery for regenerative medicine will be introduced and the impact this is beginning to have

on the diseases of ageing described. Our own research in the discovery of small molecules to modulate utrophin

for the treatment of the muscle degenerative disease, Duchenne Muscular Dystrophy will be described;

translation of the first generation utrophin modulator to the clinic and deconvolution of its molecular target and

mechanism. Our extension of this approach into regenerative medicine will then be described, exemplified by the

discovery of small molecules to stimulate neurogenesis in vitro and in vivo.

Leading References

Davies, S.G.; Kennewell, P.D.; Russell, A.J.*; Seden, P.T.; Westwood, R.; Wynne, G.M.  Stemistry: the Control

of Stem Cells in situ using Chemistry. J. Med. Chem. 2015, 58, 2863-2894. DOI: 10.1021/jm500838d.

Wilkinson, I.V.L.; Perkins, K.J.; Dugdale, H.; Moir. L.; Vuorinen, A.; Chatzopoulou, M.; Squire, S. E.;

Monecke, S.; Lomow, A.; Geese, M.; Charles, P.D.; Burch, P.; Tinsley, J.M.; Wynne, G.M.; Davies, S.G.;

Wilson, F.X.; Rastinejad, F.; Mohammed, S.; Davies, K.E.; Russell, A.J.* Chemical Proteomics and Phenotypic

Profiling Identifies the Aryl Hydrocarbon Receptor as a Molecular Target of the Utrophin Modulator

Ezutromid. Angew. Chem. Int. Ed. 2020; DOI: 10.1002/anie.201912392.

Babbs, A.; Berg, A.; Chatzopoulou, M.; Davies, K.E.; Davies, S.G.; Edwards, B.; Elsey, D.J.; Emer, E.; Guiraud,

S.; Harriman, S.; Lecci, C.; Moir, L.; Peters, D.; Robinson, N.; Rowley, J.A.; Russell, A.J.*; Squire, S.E.;

Tinsley, J.M.; Wilson, F.X.; Wynne, G.M. 2-Arylbenzo[d]oxazole phosphinate esters as second-generation

modulators of utrophin for the treatment of Duchenne Muscular Dystrophy. J. Med. Chem. 2020 ; DOI:

10.1021/acs.jmedchem.0c00807.

EFMC-ISMC | 85

LE042

PROBING EMBRYONIC MESODERMAL DIFFERENTIATION

ENABLES IDENTIFICATION OF SMALL MOLECULE BONE

MORPHOGENETIC PROTEIN ACTIVATORS

Fabian Wesseler (1,2,3), Daniel Riege (3), Stefan Lohmann (3), Jonas Halver (1), Mahesh Puthanveedu (1),

Jessica Bertrand (4), Andrey Antonchick (1), Slava Ziegler (5), Sepand Rastegar (6), Sonja Sievers (2,5),

Herbert Waldmann (1,5), Dennis Schade (3,7)

1) Faculty of Chemistry and Chemical Biology, Technical University Dortmund, 44227 Dortmund, Germany2) Compound Management and Screening Center, 44227 Dortmund, Germany

3) Department of Pharmaceutical/Medicinal Chemistry, Christian-Albrechts-University of Kiel, 24118 Kiel, Germany4) Department of Orthopedic Surgery, Otto-von-Guericke University, 39120 Magdeburg, Germany

5) Max-Planck-Institute of Molecular Physiology, 44227 Dortmund, Germany6) Institute of Biological and Chemical Systems-Biological Information Processing, Karlsruhe Institute of Technology, 76021

Karlsruhe, Germany7) Partner Site Kiel, DZHK, German Center for Cardiovascular Research, 24105 Kiel, Germany

Bone Morphogenetic Proteins (BMPs) belong to the superfamily of Transforming Growth Factor-β (TGFβ)

ligands and play central roles during embryonic development, tissue homeostasis and regeneration. The quality

and quantity of BMP signaling outputs largely depend on the cellular context and is regulated at multiple levels.

Owing to their osteoinductive activity, recombinant BMPs mostly gained attention for therapeutic approaches to

address skeletal trauma and osteopenic diseases.

Chemical modalities that serve as genuine BMP mimetics or potentiators are highly desirable, not only for bone

regeneration but also for Xeno-free methods in stem cell technology. However, the discovery and development

of such small molecule growth factor/cytokine activators is intrinsically challenging.

We aimed at devising a physiological, morphogenic cellular screening system that is focused on the BMP

pathway, yet target-agnostic, to increase chances for expanding the druggable space of potential BMP effectors.

Building on the multiphasic roles of BMPs during embryonic development, our chemical biology approach

identified a time frame during mesodermal structuring of murine embryonic stem cells that enabled probing of

the BMP pathway in a high-throughput format. Specifically, the orchestration of signaling cues is exploited in

the context of mesendodermal structuring towards cardiovascular cell fates. Proof-of-concept is presented from a

screen of >7.000 compounds. A panel of secondary, orthogonal BMP-dependent assays established stringent

filtering and validation of hits.

Our approach harnesses embryonic development as an enabling technology for drug discovery in regenerative

medicine. Here, we disclose novel chemotypes as osteogenic BMP activators and provide insights into the

compounds‘ mechanism of action.

86 | EFMC-ISMC

LE043

POTENTIAL OF STEM CELL TECHNOLOGY IN DISCOVERY OF

DRUGS FOR NEURODEGENERATIVE DISEASES

Colin Pouton

Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia

There is an urgent need to develop therapies that reduce the rate of neurodegeneration in Alzheimer’s disease

(AD). Recent clinical trials have focused on attempts to reduce the accumulation of amyloid-beta in the brain. 

While formation of amyloid-beta plaques is a classical pathological hallmark of AD, it is not necessarily the

cause of AD, and attention is turning to a search for new opportunities.  A paradigm shift is required to identify

new targets for drug discovery. Human pluripotent stem cell (hPSC) technology offers a new approach to target

identification and lead development.

hPSC technology allows relevant human brain cell phenotypes (i.e. specific mature neurons, astrocytes, and

microglia) to be produced by in vitro differentiation (IVD).  This approach allows complex models to be

assembled, using 2D or 3D mixed cultures, representing the early stages of neurodegeneration, characterised by

damage to synapses, which generally precedes the death of neurons.  Such models represent a holistic approach

to AD, allowing identification of therapeutic targets using models of adequate complexity.  To explore genetic

factors directly, the genomes of hPSCs can be edited to introduce AD-associated polymorphisms, allowing

mechanisms of disease to be studied at the molecular level.  In addition, panels of genotyped patient-derived

induced PSCs (iPSCs) will allow identification of common pathways with the potential to reduce the rate of

degeneration in late-onset AD patients.

An important goal is the design of quantitative assays of neuronal health and microglial function that are quick

and easy to use, but can operate in a complex mixed cell environment.  Ideally an assay would determine the

health and function of synapses, to identify strategies for neuroprotection, using freely available plate readers. 

Such an assay would facilitate screening, using known drugs or gene editing strategies, with adequate throughput

and replication of experiments. 

Over the past ten years we have developed expertise in engineering hPSCs by homologous recombination (i.e.

gene editing) to introduce fluorescent or enzymatic reporters of gene expression.  Such reporters are valuable to

identify and isolate specific phenotypes during differentiation.  Enzymatic reporters allow high-throughput

assays of gene expression to be developed.  We are now using similar gene targeting techniques to engineer

more complex reporters of synaptic activity.

EFMC-ISMC | 87

LE044

AI ASSISTED AND AUTOMATED CHEMICAL SYNTHESIS

Klavs F. Jensen

Department of Chemical EngineeringMassachusetts Institute of Technology (MIT)

77 Massachusetts Avenue, Cambridge MA 02139, U.S.A.

This presentation explores opportunities in integration of computer aided synthesis planning with an automated,

robotic synthesis platform.  Computer aided synthesis planning (CASP) based on machine-learning (ML)

techniques, a subset of AI methods, have gained increasing interest in the past decade, in part because of their

scalability in incorporating new reactions from publications, patents, and electronic lab notes into trained

models.

1,2

  The ML-based CASP platform, ASKCOS, developed by MIT researchers is discussed with examples

of applications in medicinal chemistry.

3,4

 With the aim of accelerating ideation and planning of chemical

synthesis, the platform accomplishes the three primary tasks of computer-aided synthesis planning, (i)

retrosynthetic planning, (ii) reaction condition recommendation, and (iii) forward-reaction prediction (Figure 1).

The retrosynthesis Monte Carlo Tree-Search algorithm speeds synthesis assessment by generating proposed

synthetic routes that can be used to prioritize pathway by synthesis metrics (e.g., ease, cost, and compound

availability).  ML models are trained on reaction data to make condition recommendations, but they are limited

by lack of high-quality data, primarily inadequate records of concentrations, reaction times, and order of addition

of reagents and catalysts.  When trained on a broad set of reactions, machine-learning models can generalize

chemical information to enable evaluation of the feasibility of suggested reaction steps.  Examples illustrate the

ability of ASKCOS’s combined ML models for the CASP tasks to produce a rank ordered list of reaction paths

to realize a molecular target from purchasable starting materials as well as related predictions of reaction

selectivity, impurities, and green chemistry conditions.   

Integration of the CASP suggested routes with actual synthesis is exemplified with a modular continuous flow

platform automatically reconfigured and executed by a robot.

3

The CASP conditions are reviewed by expert

chemists, who add residence times, stoichiometries, and concentrations compatible with continuous flow –

elements difficult to predict because of the lack of data. To execute the planned syntheses, a robot assembles

modular reactors and separators into a continuous flow path and connects reagent lines. The integration of CASP

and flow chemistry is demonstrated for medicinally relevant small molecules, including ACE inhibitors and

nonsteroidal anti-inflammatory compound libraries. Integration of the system with HPLC and FTIR monitoring

modules (Figure 2) is shown to enable automated optimization of reaction conditions.

5

In addition to continuous

variables (e.g., temperature, concentrations, and residence time), the robot allows optimization of categorical

variables (e.g. reagents, solvents, and reactor modules), which otherwise would require manual interruptions of

the optimization process.  Finally, challenges and opportunities in further integration of CASP and synthesis

platforms are discussed, including generation of data for machine-learning models.

6

The presenter would like to thank colleagues and coworkers for contributions to the work presented. 

References

1) Thakkar, A., Johansson, S.; Jorner, K.; Buttar, D,; Reymond, J.-L.; Engkvist, O., React. Chem. Eng., 2021, 6, 27.

2) Jorner, K., Tomberg, A.; Christoph Bauer, C.; Sköld, C.; Norrby, P.O., Nat. Rev. Chem. 2021, 5, 240.

3) Coley, C.W., et al., Science, 2019, 365, 557.

4) Struble, T.J., et al., J. Med. Chem. 2020,63, 8667.

5) Breen, C.P.; Nambiar, A.M.K.; Jamison, T. F.; Jensen, K.F., Trends Chem., 2021, 3 373.

6) Eyke, N.S.; Koscher, B.A.; Jensen, K.F., Trends Chem., 2021, 3 120.

88 | EFMC-ISMC

LE045

UNIVERSAL SYNTHESIS MACHINES AND CHEMPUTATION

Leroy (Lee) Cronin

University of Glasgow, Cronin Laboratory, School of ChemistryJoseph Black Building - G12 8QQ Glasgow, United Kingdom

Today it is possible to design and synthesize many of the physically allowed molecules and materials

conceivable if practical, yet paradoxically it is not possible to reproduce or rerun these successful procedures

with high reliability. This is because many of the conditions devised for the manual or semi-manual synthesis are

not uniformly recorded. The situation is even worse when the literature is investigated. For example, our

preliminary studies reveal that >80% of all the published procedures fail to replicate without the help of an

expert.

In this talk I will outline how we have solved this problem by devising a universal approach to chemical

synthesis that allows us to translate all procedures, manual or automatic, to a new interchange format, XDL, that

allows chemistry to be universally communicated. Furthermore, this new approach maps into a universal

programming language for chemistry that is accessible to ALL synthetic chemists and will work on ALL robotic

systems (subject to suitable specification).  We demonstrate that the process is universal, and by analogy with

computation, we call systems capable of universally turning code into reliable chemistry and materials processes

Chemputation, see Figure.

 

Figure: Depiction of a chemical state machine (CSM) for synthesis that is capable of Chemputation. The input is

a combination of reagents, process information and hardware addresses. The CSM organizes the reagents and the

processes by using a scheduler that then gets executed in the hardware as a function of the available state until

the product is formed.

I will also explain why our Chemical Synthesis Machine is really a Chemical State* Machine (CSM) and why

you should care. This because our CSM can be used with AI and machine learning to search for new reactivity,

reactions, materials, and molecules – that once discovered, can be reproduced using the CSM. This allows us to

build the ultimate system that can both reproduce, search, discover, and update chemical knowledge

autonomously in real time. I will show this working with real world examples from energy materials to small

molecule drug candidates.

References

1) J. Granda, L. Donina, V. Dragone, D. –L. Long, L. Cronin 'Controlling an organic synthesis robot with machine learning

to search for new reactivity', Nature, 2018, 559, 377-381.

2) P. Kitson, G. Marie, J. –P. Francoia, S. Zalesskiy, R. Sigerson, J. S. Mathieson, L. Cronin 'Digitization of multistep

organic synthesis in reactionware for on-demand pharmaceuticals', Science, 2018, 359, 314-319.

3) S. Steiner, J. Wolf, S. Glatzel, A. Andreou, J. Granda, G. Keenan, T. Hinkley, G. Aragon-Camarasa, P. J. Kitson, D.

Angelone, L. Cronin 'Organic synthesis in a modular robotic system driven by a chemical programming language', Science,

2019, 363, 144-152

4) P. S. Gromski, A. Henson, J. Granda, L. Cronin 'How to explore chemical space using algorithms and automation', Nat

Rev Chem., 2019, 3, 119-128.

5) S. Hessam M. Mehr, M. Craven, A. Leonov, G. Keenan, L. Cronin 'A universal system for digitization and automatic

execution of the chemical synthesis literature', Science, 2020, 370, 101-108.

EFMC-ISMC | 89

LE046

AI IN THE LAB: AUTOMATING CHEMICAL SYNTHESIS. WHERE

NEXT FROM HERE?

Teodoro Laino, Matteo Manica, Alain Vaucher, Antonio Cardinale, Alessandro Castrogiovanni,

Aleksandros Sobczyk, Phlippe Schwaller, Alessandra Toniato, Heiko Wolf

IBM Research Europe, Saeumerstrasse 4, CH-8803 Rueschlikon (Switzerland)

Designing and making new molecules is one of the most impactful outcomes of chemistry. The use of domain

knowledge accumulated over multiple decades of laboratory experience, has been a crucial element for the

synthesis of many new molecular structures. Still, most synthetic success stories are accompanied by long hours

of unsuccessful exploration. No more than 20 years ago, automation systems were designed to support chemists

in repetitive laboratory tasks. While this proved very effective in a few areas, such as high-throughput chemistry,

the use of automation for general purpose tasks remains an incredible challenge even today. It requires chemistry

operators to write different software for different tasks, each codifying a specific and different type of chemistry.

Meanwhile, Artificial Intelligence (AI) has emerged as a valuable complement to human knowledge and

creativity in organic chemistry - for tasks like predicting chemical reactions [1-2], retrosynthetic routes [3],

digitizing chemical literature [4] or predicting entire sequences of experimental protocols [5]. Here, we present

the first implementation of a cloud-based AI-driven autonomous laboratory.

The remote laboratory is made accessible to chemists through the cloud and is equipped with automation

technologies. The AI assists remote chemists with several tasks: designing retrosynthetic trees and suggesting

the correct sequence of operational actions (reaction conditions and procedures), or ingesting literature on

synthetic procedures to convert them into an executable program. Following supervision by synthetic chemists,

the AI self-programs the automation layer and makes decisions on the synthesis execution using feedback loops

from analytical chemistry instruments. I will present the platform architecture and its performance across various

classes of synthetic tasks.

 

References

1) IBM Research Europe, Chem. Sci., 2018, 9, 6091-6098

2) IBM Research Europe, ACS Cent. Sci. 2019, 5, 9, 1572-1583

3) IBM Research Europe, Chem. Sci., 2020, 11, 3316-3325

4) IBM Research Europe, Nat. Comm., 2020, 11, 3601

5) IBM Research Europe, Nat. Comm., 2021, 12, 2573

6) https://rxn.res.ibm.com

90 | EFMC-ISMC

LE047

PROGRESS AND LIMITATIONS IN EXPLORING THE CHEMICAL

SPACE WITH AI

Ola Engkvist

AstraZeneca R&DPepparedsleden 1431 50 Mölndal

Sweden

Artificial Intelligence has become impactful during the last few years in chemistry and the life sciences, pushing

the scientific boundaries forward as exemplified by the recent success of AlphaFold2. In this presentation I will

provide an overview of how AI have impacted the chemical space relevant for drug design in the last few years,

where we are now and what progress we can reasonably expect in the coming years. The presentation will have a

focus on deep learning based molecular de novo design, however, also aspects of synthesis prediction, molecular

property predictions and chemistry automation will be covered.

EFMC-ISMC | 91

LE048

INHALED NEW MODALITIES IN RESPIRATORY DISEASE: PAST,

PRESENT, FUTURE

Werngard Czechtizky

Medicinal Chemistry, Research & Early Development, Respiratory & Immunology (R&I), BioPharmaceuticals R&D,AstraZeneca, Gothenburg, Sweden.

Inhalation of small molecule drugs has proven very efficacious for the treatment of respiratory diseases due to

enhanced efficacy and a favorable therapeutic index. It enables targeted delivery to the tissue or even cells of

interest with rapid onset of therapeutic action, low systemic drug exposure, and thereby reduced systemic side

effects.

An increasing number of pharmaceutical companies and biotechs are investing in new modalities – for this

presentation defined as synthetic molecules with a molecular weight >700 Da and beyond inhaled small

molecule space. As of yet, the general expertise with inhalation of synthetic modalities such as PROTACs,

peptides and oligonucleotides (antisense oligonucleotides, siRNAs, miRs and antagomirs) for treatment of

respiratory diseases is still limited. The retention and metabolization of these types of molecules in lung tissue /

fluid is under full investigation and will contribute to our understanding which of these modalities are suited for

inhaled RoA.

This presentation will provide an overview of the scientific expertise around the inhalation of PROTACs,

peptides and oligonucleotides to treat respiratory diseases. Available information on physicochemical and

metabolic stability of new modalities in healthy and diseased lung as well as formulation, aerosolization and

delivery aspects will be outlined. Finally, first clinical trial outcomes with inhaled new modalities will be

summarized.

92 | EFMC-ISMC

LE049

DISCOVERY OF NOVEL INHALED PI3Kδ INHIBITOR BY LUNG

RETENTION OPTIMIZATION

Montse Erra, Joan Taltavull, Francisco Javier Bernal, Juan Francisco Caturla, Marta Carrascal, Lluís

Pagès, Marta Mir, Sònia Espinosa, Jordi Gràcia, María Domínguez, Mar Sabaté, Stéphane Paris, Mónica

Maldonado, Begoña Hernández, Mónica Bravo, Elena Calama, Montserrat Miralpeix, Martin Lehner,

Marta Calbet, Marisa Viñals, Mireia Verdú

Almirall R&D, Laureà Miró, 408-410 Sant Feliu de Llobregat (Spain)

PI3Kδ  is a lipid kinase that belongs to the PI3K class I family along with PI3Ka, PI3Kb,  PI3Kg, the first two

isoforms being ubiquitously expressed and PI3Kd and PI3Kg showing a more restricted expression pattern that

seems to play a major role in leukocytes.

In this sense, PI3Kδ is expressed in several immune cells modulating relevant functions that include cell

migration, cytokine production, antibody production, and even steroid resistance. Such effects suggest that

selective pharmacological inhibition of PI3Kδ could be a promising approach for treating respiratory diseases

such as asthma and COPD.

Considering that oral PI3Kδ inhibitors approved as anticancer agent,  such as Idelalisib, have reported serious

adverse effects including hepatic toxicity, severe diarrhoea, colitis, pneumonitis, infections and intestinal

perforation, our internal approach was focused on reducing systemic side effects by administering the

compounds by the inhaled route. Compounds administered by inhalation are delivered directly to the site of

action and may improve the therapeutic index of a drug, minimizing undesired side effects.  

Thus, a Medicinal Chemistry exercise focused on optimizing both the in vitro potency and the ADME profile of

a series of novel pyrrolotriazinones will be presented, with a particular emphasis on the identification of the

suitable chemical space that can lead to optimal physicochemical properties translating into better lung retention

and avoiding low solubility. This exercise culminated with the identification of LAS195319 as a candidate for

clinical development, and its complete in vitro and in vivo profile will be also presented.

EFMC-ISMC | 93

LE050

INTRODUCTION TO PRECLINICAL INHALED DRUG DOSING AND

ITS APPLICATION TO MEASURE THE THERAPEUTIC INDEX OF

INHALED STEROIDS

Jonathan Phillips

Amgen, Newbury Park, United States

There are many advantages to inhaled administration of drugs for the treatment of respiratory diseases. Inhaled

delivery applies the therapeutic agent directly to the site of action, the lungs. A high local concentration of the

drug in the lungs offers a significant advantage that minimizes dose and systemic exposure, and maximizes

efficacy. This is an important advantage that can greatly increase the therapeutic index (TI, ratio of dose of drug

that causes a side-effect over the dose of drug that provides efficacy) of a drug.  Pre-clinically, inhaled

compounds are optimized to increase the TI, which requires simultaneous measurements of in vivo efficacy and

side effect.  An introduction to the methods available to deliver drugs topically to the lungs of rodents will be

presented, focusing on dry powder nose-only inhalation (NOI) techniques.  The TI of steroids delivered by NOI

to an asthmatic rat disease model are compared.  This model could be used to identify novel inhaled steroids

optimized to have a greater TI than those currently available.

94 | EFMC-ISMC

LE051

DESIGN AND DEVELOPMENT OF INHALED MOLECULES TO

TARGET THE PULMONARY VASCULATURE

Duncan Shaw, Mark Healy

NIBR 22 Windsor Street, Cambridge MA 02139 USA

Inhaled delivery has been shown to give real therapeutic advantages for drugs that are targeted to the airway

epithelial surface and to bronchial smooth muscle layers. It is however, an open question whether drugs

delivered to the airway can be targeted to tissues deeper within the lung.

Two Novartis projects sought to answer this question experimentally. The overarching aim was to enable the

delivery of powerful therapeutics that can intervene in a wider range of lung diseases, such as pulmonary arterial

hypertension (PAH), while reducing the burden of systemic side effects.

In response to the IMPRESS clinical trial

1

, which showed efficacy for Imatinib in PAH, a series of potent dual

PDGFR/cKIT inhibitors have been identified. The series was optimized for duration of action in the lung. A

novel kinase occupancy assay was used to directly measure target engagement after intratracheal dosing and

allowed the development of apotent compound with a long inhaled duration of action.

Prostacyclin receptor (IPR) agonists have long been the standard of care in PAH and we will describe how a cell

affinity assay was developed and used to optimize inhaled IPR agonists. This novel screening method identified

compounds which achieved long duration of action, by binding selectively to lung tissue and achieving

significant improvements in preclinical therapeutic index compared to the inhaled standard of care treprostinil.

The learnings from these projects will be synthesized to provide a medicinal chemistry strategy that can be used

to develop improved inhaled therapeutics for PAH and address further unmet needs in other respiratory diseases

across a range of protein targets and drug modalities.

EFMC-ISMC | 95

LE052

DISCOVERY OF AN ORAL, RO5 COMPLIANT, INTERLEUKIN 17A

PROTEIN-PROTEIN INTERACTION MODULATOR FOR THE

TREATMENT OF PSORIASIS AND OTHER INFLAMMATORY

DISEASES

KEVIN DACK

LEO PHARMA. COPENHAGEN, DENMARK

Injectable biologics, such as the IL-17A pathway mAbs brodalumab, secukinumab, and ixekizumab have

become established as highly effective and safe treatments for moderate to severe psoriasis. These mAbs prevent

the cytokine from activating the IL-17 Receptor and hence reduce the inflammatory drive to allow resolution of

the inflammation.

An Oral therapy, with similar efficacy and safety to these biologics, is desirable and in this presentation we will

disclose our 1st small molecule (Rule-of-5 compliant) orally bioavailable candidate that binds to IL-17A to

modulate its structure and function. We will describe key SAR, the binding mode in IL-17A and some of the

preclinical data that supported its nomination as a Development Candidate for the Oral Treatment of Psoriasis.

96 | EFMC-ISMC

LE053

INSIGHTS TO THE DISCOVERY AND DESIGN OF ATAXIA

TELANGIECTASIA MUTATED (ATM) KINASE INHIBITORS M3541

AND M4076 WITH STRONG ANTI-TUMOR EFFICACY IN

COMBINATION THERAPY APPROACHES

Thomas Fuchss, Ulrich Graedler, Kai Schiemann, Daniel Kuhn, Holger Kubas, Ulrich Pehl, Astrid

Zimmermann, Heike Dahmen, Axel Becker, Mireille Krier, Christoph Saal

Merck KGaA, Frankfurter Strasse 250, 64293 Darmstadt, Germany

M3541 and M4076 are ATP-competitive and sub-nanomolar potent small molecule inhibitors of the Ataxiatelangiectasia mutated (ATM) kinase, which target tumor cell survival and growth by inhibiting double-strand

break (DSB) repair as well as checkpoint control. DSB repair is crucial for survival of malignant tumor cells,

especially under treatment with DNA damaging chemo- and radiotherapy. As such, the rationale of

pharmacological inhibition of ATM is to increase and maintain the extent of unrepaired DNA damage generated

by radio-, chemotherapy, and targeted therapies to drive tumor cells into cell death.

We present discovery, optimization, structure-activity relationships as well as physicochemical profiles and

preclinical characterization using biochemical, cellular and in vivo human tumor models.  

The axial chiral ATM inhibitor M4076 is currently being investigated in a Phase I clinical trial for the treatment

of cancer.

EFMC-ISMC | 97

LE054

DISCOVERY OF JDQ443 A STRUCTURALLY UNIQUE, HIGHLY

POTENT, SELECTIVE AND ORALLY BIOAVAILABLE KRAS

G12C

COVALENT INHIBITOR

S. Cotesta, E. Lorthiois, A. Vaupel, C. Leblanc, R. Wilcken, C. Bomio-Confaglia, S. Stutz, P. Manley, P.

Rigollier, R. Machauer, R. Mah, E. Altmann, N. Schneider, K. Beyer, D. Guthy, D. Sterker, D. Erdmann,

N. Ostermann, B. Shrestha, J. Ottl, P. Skaanderup, J. Giovannoni, R. Stringer, R. De Kanter, E. Jimenez

Nunez, T. Widmer, D. Roman, P. Wessels, V. Head, L. McGregor, M. Schirle, J. Kearns, H. Voshol, D.

Graus Porta, M. Maira, R. Sedrani, F. Hoffman, J, Engelmann, F. Zecri, A. Weiss , SM. Brachmann, M.

Gerspacher

Novartis, Chemistry, 4002 Basel, Switzerland

RAS is the most frequently mutated oncogene in cancer, with KRAS G12C mutations most commonly found in

lung adenocarcinoma, colorectal cancer, and other solid tumor malignancies. First generation KRAS

G12C

inhibitors show anti-tumor efficacy in early phase clinical trials. Here we report the identification and

optimization of a new KRAS

G12C

chemo-type discovered by structure based de-novo design. The chemical series

binds to the KRAS

G12C

switch II pocket with a novel binding mode, exploiting unique interactions with the

GDP-bound form of the KRAS

G12C

protein. Optimization of the chemo-type by structure-based design and

medicinal chemistry led to the discovery of NVP-JDQ443 (JDQ443), a novel, highly potent, selective and orally

bioavailable KRAS

G12C

covalent inhibitor. NVP-JDQ443 shows dose-dependent anti-tumor activity in mice

bearing KRAS G12C mutated tumor xenografts.  A Phase Ib/II study of NVP-JDQ443 in patients with advanced

solid tumors harboring the KRAS G12C mutation is currently ongoing (NCT04699188).

98 | EFMC-ISMC

LE055

C-TYPE LECTIN RECEPTORS AS DRUG TARGETS

Christoph Rademacher (1,2)

1) University of Vienna, Department of Pharmaceutical Sciences, Althanstrasse 14, 1090 Vienna, Austria2) University of Vienna, Department of Microbiology, Immunology and Genetics, Max F. Perutz Labs, Biocenter 5, 1030

Vienna, Austria

The C-type lectin protein family is the largest and most complex family of mammalian carbohydrate-binding

proteins. The distinct expression pattern on defined cellular subsets has spurred interest in their targeting for

many decades already. As a prime example, the asialoglycoprotein receptor (ASGPR) has been utilized as

receptor present on hepatocytes to enable specific delivery of therapeutic siRNA (Gvosiran, Alnylam

Pharmaceuticals). The ASGPR recognizes carbohydrates conjugated to its RNA cargo and allows receptor

mediated endocytosis. Thus, it can be hijacked for the delivery of therapeutics. Other members of the C-type

lectin family are expressed on defined sets of cells of the innate immune system. In this case, these cell surface

receptors promote pathogen recognition either triggering immune cell activation or endocytosis, finally leading

to antigen presentation. Overall, these characteristics of having a defined expression pattern and being able to

trigger an immune response, render C-type lectin receptors excellent drug targets for immunomodulation. 

Here, I will summarize our efforts to make use of the structural plasticity of C-type lectins for the development

of small molecule modulators of their receptor function. Previously, we have highlighted the discrepancy

between the static picture of these proteins as inferred from X-ray crystallography and the experimental

description of susceptibility of these proteins for drug-like molecule binding. We found that besides the shallow

and featureless carbohydrate recognition site, several secondary sites exist that are partially druggable and offer

possibilities for inhibitor design against C-type lectins [1]. These insights are complemented by our studies into

the receptor flexibility using protein NMR in combination with molecular dynamics simulations revealing an

allosteric network of communicating amino acid sidechains [2, 3]. To showcase the application of our C-type

lectin ligands, I will present some of your latest work on the targeted delivery of therapeutics to Langerhans cell

in the human skin via targeting of langerin (CD207) [4]. 

References

1) Aretz, J., Baukmann, H., Shanina, E., Hanske, J., Wawrzinek, R., Zapol'skii, V.A., Seeberger, P.H., Kaufmann, D.E., and

Rademacher, C. (2017). Identification of Multiple Druggable Secondary Sites by Fragment Screening against DC-SIGN.

Angew Chem Int Ed Engl 56, 7292-7296.

2) Hanske, J., Aleksic, S., Ballaschk, M., Jurk, M., Shanina, E., Beerbaum, M., Schmieder, P., Keller, B.G., and Rademacher,

C. (2016). Intradomain Allosteric Network Modulates Calcium Affinity of the C-Type Lectin Receptor Langerin. J Am

Chem Soc 138, 12176-12186.

3) Aretz, J., Anumala, U.R., Fuchsberger, F.F., Molavi, N., Ziebart, N., Zhang, H., Nazare, M., and Rademacher, C. (2018).

Allosteric Inhibition of a Mammalian Lectin. J Am Chem Soc 140, 14915-14925.

4) Wamhoff, E. C., Schulze, J., Bellmann, L., Rentzsch, M., Bachem, G., Fuchsberger, F. F., Rademacher, J., Hermann, M.,

Del Frari, B., van Dalen, R., Hartmann, D., van Sorge, N. M., Seitz, O., Stoitzner, P., and Rademacher, C.* (2019) A

Specific, Glycomimetic Langerin Ligand for Human Langerhans Cell Targeting, ACS Cent Sci 5, 808-820.

EFMC-ISMC | 99

LE056

DESIGN, SYNTHESIS, AND DEVELOPMENT OF LECTIN LIGAND

MIMETICS

Ulf Nilsson

Lund University, Department of Chemistry, Centre for Analysis and SynthesisP.O. Box 124 - 221 00 Lund, Sweden

Galectins are glycoconjugate-binding lectins that bind galactopyranose-containing glycans to influence e.g.

glycoprotein trafficking, localization, residence times, which in turn may impact glycoprotein functions in cell

proliferation, inflammatory processes, tumor growth, and tumor metastasis.

1

 Consequently, this has sparked a

growing interest in developing galectin inhibitors as research tools and drug leads. Herein we will present our

efforts towards discovery and development of high-affinity, selective, and orally available galectin-3-inhibiting

compounds,

2,3

with emphasis on biophysical aspects

4-6

 of ligand-protein interactions such as conformational

entropy, entropy-entropy compensation, and halogen bond-solvation thermodynamics. Furthermore, validation

of galectin-3 as a target in cancer and fibrosis and key aspects for selecting and advancing galectin inhibitors to 

the clinic will be presented.

References

1) Johannes, L., Jacob, R. & Leffler, H. Galectins at a glance. J. Cell. Sci. 131, doi:10.1242/jcs.208884 (2018).

2) Delaine, T. et al. Galectin-3-Binding Glycomimetics that Strongly Reduce Bleomycin-Induced Lung Fibrosis and

Modulate Intracellular Glycan Recognition. ChemBioChem 17, 1759-1770, doi:10.1002/cbic.201600285 (2016).

3) Zetterberg, F. R. et al. Monosaccharide Derivatives with Low-Nanomolar Lectin Affinity and High Selectivity Based on

Combined Fluorine-Amide, Phenyl-Arginine, Sulfur-π, and Halogen Bond Interactions. ChemMedChem 27, 133-137,

doi:papers3://publication/doi/10.1002/cmdc.201700744 (2018).

4) Verteramo, M. L. et al. Interplay between Conformational Entropy and Solvation Entropy in Protein–Ligand Binding. J.

Am. Chem. Soc. 141, 2012-2026, doi:papers3://publication/doi/10.1021/jacs.8b11099 (2019).

5) Stenström, O., Diehl, C., Modig, K., Nilsson, U. J. & Akke, M. Mapping the energy landscape of protein–ligand binding

via linear free energy relationships determined by protein NMR relaxation dispersion. RSC Chem. Biol. 2, 259-265,

doi:10.1039/d0cb00229a (2021).

6) Wallerstein, J. et al. Entropy–Entropy Compensation between the Protein, Ligand, and Solvent Degrees of Freedom

Fine-Tunes Affinity in Ligand Binding to Galectin-3C. JACS Au 1, 484-500, doi:10.1021/jacsau.0c00094 (2021).

100 | EFMC-ISMC

LE057

LECTINS FROM PATHOGENS: FROM STRUCTURAL

GLYCOBIOLOGY TO ANTIADHESIVE STRATEGIES

Anne Imberty

CERMAV, CNRS and Université Grenoble Alpes, Grenoble, France

A large number of pathogenic microorganisms display receptors for specific recognition and adhesion to the

glycoconjugates present on human tissues.  In addition to membrane-bound adhesins, soluble lectins are

involved in lung infections caused by the bacteria Pseudomonas aeruginosa  and  Burkholderia cepacia  and by

the fungus Aspergillus fumigatus that are responsible  for hospital-acquired diseases. The multivalency of lectin

is proposed to play a role in their strong avidity for glycosylated cell surfaces, their specific binding to somec

cell types, and also in their ability to affect membrane dynamics by clustering glycosphingolipids, resulting in

some cases in internalization of intracellular pathogens.

Accumulated knowledge about the structures of the lectins and the interactions with host glycoconjugates has led

to the design of powerful glyco-derived inhibitors that can serve as antimicrobial therapeutic agents, as a

complement to or an alternative to antibiotic therapy. Several strategies are developed : glycoderivatives with

increased affinity, glycomimetics with no carbohydrate moiety or multivalent compounds that make use of the

receptor architecture.

References

1) Sommer, R., K. Rox, S. Wagner, D. Hauck, S. Henrikus, S. Newsad, T. Arnold, et al. 2019. 'Anti-biofilm agents against

Pseudomonas aeruginosa: a structure-activity relationship study of C-glycosidic LecB inhibitors', J. Med. Chem., 62:

9201-16.

2) Kuhaudomlarp, S., L. Cerofolini, S. Santarsia, E. Gillon, M. Denis, S. Fallarini, S. Giuntini, et al. 2020. 'Fucosylated

ubiquitin and orthogonally glycosylated mutant A28C: Conceptually new ligands for Burkholderia ambifaria lectin (BambL)

', Chem. Sci., 11: 12662–70.

3) Lal, K., R. Bermeo, J. Cramer, F. Vasile, B. Ernst, A. Imberty, A. Bernardi, A. Varrot, L. Belvisi. in press. 'Prediction and

validation of a druggable site on virulence factor of drug resistant Burkholderia cenocepacia. ', Chem. Eur. J.

4) Kuhaudomlar, S., E. Siebs, E. Shanina, J. Topin, I. Joachim, P. da Silva Figueiredo Celestino Gomes, A. Varrot, et al.

2021. 'Non-carbohydrate glycomimetics as inhibitors of calcium(II)-binding lectins', Angew. Chem. Int. Ed., 60: 2-13.

EFMC-ISMC | 101

LE058

DEVELOPMENT OF GLYCOMIMETIC COLLECTIN-11

ANTAGONISTS TO REDUCE COMPLEMENT-MEDIATED

ISCHEMIA-REPERFUSION INJURIES

Rachel Hevey (1), Deborah Berta (1), Mergim Maraj Martinez (1), Tobias Wichers (1), Tobias Brunner

(1,2), Martin Smiesko (2), Said Rabbani (1), Daniel Ricklin (1)

1) Molecular Pharmacy, Dept. Pharmaceutical Sciences, University of Basel, Switzerland2) Computational Pharmacy, Dept. Pharmaceutical Sciences, University of Basel, Switzerland

Glycans are ubiquitous in nature, decorating the extracellular surface of cells and acting as a first point of contact

with neighbouring cells and biomolecules. They play a critical role in numerous biological processes, including

inflammation, cell adhesion, and host-pathogen recognition, and as a result carbohydrate-binding proteins have

been rapidly gaining interest as targets in drug development programs. Although carbohydrate structures have

extensive structural diversity, they suffer from weak binding affinities and inherently poor pharmacokinetic

properties. In order to improve their drug-like properties, ‘glycomimetic’ compounds can be designed which

mimic the structure and function of the native glycan, yet show improved affinity, specificity, and bioavailability

[1].

Collectin-11 (CL-11, CL-K1) is a protein of the complement innate immune pathway that is highly relevant in

the context of ischemia-reperfusion-associated injury in renal transplantation, having recently been demonstrated

to play an important role in both acute kidney injury and late-stage/chronic renal inflammation. In a murine renal

transplantation model, preventing CL-11 binding to carbohydrate structures at the renal cell surface reduced

complement-mediated tissue damage, reduced neutrophil infiltration, and improved renal function [2-4].

Given the inherently weak affinities of native carbohydrate ligands for their lectin targets, we have been focused

on developing glycomimetic inhibitors of CL-11 with enhanced affinities and improved pharmacokinetic

properties. Based on insights from in silico studies and an available X-ray structure [5], we have constructed

several glycomimetic libraries and evaluated them using a thermal denaturation-based nano-differential scanning

fluorimetry assay, and a polymer-based competitive binding assay. Initial glycomimetic leads based on

mannobiose have demonstrated nearly 100-fold improvements in affinity, advancing our efforts at developing a

therapeutic entity against ischemia-reperfusion injuries and further evaluating the biological role of CL-11.

References

1) Hevey, R. Pharmaceuticals 2019, 12, 55.

2) Farrar, C.A.; et al. J. Clin. Invest. 2016, 126, 1911-1925.

3) Wu, W.; et al. J. Am. Soc. Nephrol. 2018, 29, 168-181.

4) Howard, M.C.; et al. FASEB J. 2020, 34, 822-834.

5) Girija, U.V.; et al. BMC Biology 2015, 13, 27.

102 | EFMC-ISMC

LE059

ARTIFICIAL INTELLIGENCE IN CHEMICAL BIOLOGY AND DRUG

DISCOVERY – DATA, APPLICATIONS, AND ILLUSIONS

Andreas Bender

Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Cambridge, UKDigital Life Sciences, Nuvisan/ICB, Berlin, Germany

Artificial Intelligence (AI) has had a profound impact on areas such as image and speech recognition; however,

comparable advances in drug discovery are still rare. This contribution will explore some of the reasons why this

is the case, starting firstly by embedding the application of AI in drug discovery into its historical context. Next,

we will explore which types of improvements – namely to speed, cost, or quality of decisions – are able to have

the most profound impact on the success of drug discovery projects, and we will see that it is quality of decisionsin clinical phases that matters most for project success. However, while computational algorithms are

tremendously powerful today, much of the data we have available in drug discovery projects comes from the

preclinical, proxy domain – which possesses only limited utility to make predictions for clinical stages. Hence,

while numerically we are at least to an extent becoming better at modelling proxy endpoints, this leads to a

‘models of models’ situation, which only translates to a very limited extent to the safety and efficacy of

compounds which will reach patients in a clinical setting. Even more fundamentally, there are intrinsic aspects

of chemical and biological data which make labelling data much more difficult than in the image- or

speech-domain, such as its conditionality (e.g. the dependence of effects on dose), which pose conceptual

problems to being able to apply computational algorithms in the drug discovery domain. The contribution will

conclude with aspects of model validation and perceptions of AI in society which pose additional problems for

their unbiased evaluation and development, and point out developments which are needed for applications of AI

in drug discovery to contribute to the clinical safety and efficacy of future medicines.

 

 

References

1) Andreas Bender and Isidro Cortés-Ciriano. Artificial intelligence in drug discovery: what is realistic, what are illusions?

Part 1: Ways to make an impact, and why we are not there yet. Drug Discovery Today, 2021 (26) 2, 511 – 524.

https://doi.org/10.1016/j.drudis.2020.12.009

2) Andreas Bender and Isidro Cortes-Ciriano. Artificial intelligence in drug discovery: what is realistic, what are illusions?

Part 2: a discussion of chemical and biological data. Drug Discovery Today, 2021 (26) 4 1040 - 1052.

https://doi.org/10.1016/j.drudis.2020.11.037

EFMC-ISMC | 103

LE060

AUGMENTING DRUG HUNTERS WITH GENERATIVE CHEMISTRY

MODELS

Nadine Schneider (2), Finton Sirockin (2), Nikolaus Stiefl, Marc Brockschmidt (1), Pashmina Cameron

(1), Nikolas Fechner (2), William Jose Godinez-Navarro (2), Rishi Gupta (2), Henry Jackson-Flux (1),

Jessica Lanini (2), Richard Lewis (2), Krzysztof Maziarz (1), Hubert Misztela (2), Nadya Paleyes (1),

Michal Pikusa (2), Marwin Segler (1), Qurrat Ul-Ain (2)

1) Microsoft Research - MSR, Cambridge, UK2) Novartis Institutes for BioMedical Research & Novartis Pharma AG, Novartis Campus,

Basel, Switzerland

[email protected]

Small-Molecule Drug discovery is a multi-objective optimization problem in which finding the next drug

candidate depends on various characteristics of compounds including efficacy, pharmacokinetics and safety. In

the design process of small molecule drugs, medicinal chemistry project teams routinely face this complex

multidimensional optimization challenge. Given the massive size of the relevant “chemical space” (estimated to

be in the range of up to 1060 drug-like molecules), the key question for medicinal chemists is “What is the best

compound to make and test next”'. While humans are extremely good in understanding the bigger picture,

computers/algorithms are potentially much better in coming up with and evaluating a large body of

complementary solutions – such as the described multidimensional optimization problem.

Novartis has partnered with Microsoft Research to explore the potential of Generative Chemistry in “real life”

conditions. We have built an in silico decision-support system that assists medicinal chemists in multi-objective

compound design, selection and prioritization. Compared to humans, GenChem is not biased by past experience,

thus it complements the chemist’s experience with independent ideas driven by data.

This presentation will describe the computational workflow that is being developed. It combines a diverse set of

generative models to suggest novel, high-quality compounds. The compounds are optimized in a continuous

latent space to fit a pre-defined property profile. Predictive models and scoring functions guide the generation of

promising candidate molecules. The application to different stages of the drug discovery process and future areas

of development will be discussed.

104 | EFMC-ISMC

LE061

NAVIGATING CHEMICAL SPACE BY ARTIFICIAL INTELLIGENCE

Gerhard Hessler, Christoph Grebner, Hans Matter

Sanofi-Aventis Deutschland GmbH, R&D, Integrated Drug Discovery,Industriepark Hoechst, D-65926 Frankfurt am Main, Germany

 The identification and optimization of novel drug candidates for clinical development is a challenging,

time-consuming process. It often requires synthesis of several hundreds to thousands of molecules in a

multiparameter optimization process. This process typically involves parallel optimization of multiple properties

like the biological activity against the desired target, selectivity over undesired targets as well as ADMET

properties. Here, artificial intelligence (AI)-based data analysis and compound design offer exciting

opportunities to mine the chemical space to accelerate the identification of drug candidates with the suitable

profile.

AI-based de novo design is used in lead generation as well as in lead optimization. Both scenarios require

different strategies to mine chemical space. In lead generation, including scaffold hopping as well, a broad

search of the chemical space is desired to identify novel chemotypes, while in lead optimization analogues of the

current lead series are explored to find the compounds with the best property profile. In this presentation, we will

describe approaches to tune AI-based exploration of chemical space between different scenarios and highlight

the integration of these technologies into discovery projects.

EFMC-ISMC | 105

LE062

GENERATIVE RECURRENT NEURAL NETWORKS (RNN) FOR DE

NOVO DRUG DESIGN

Suneel Kumar BVS, Rochish Gundabathula, Pooja Deshmukh, Victoria Steadman, Veena MM

Sai Lifesciences pvt limited, Unit-II, Plot No. DS-7, ICICI knowledge Park, Turkaplly Village, Shameerpet, Hyderabad,Telangana 500078

De novo drug design is a well-established method to computationally generate novel molecules for purchase or

synthesis  which have good potency and affinity towards the biological target. Generative models are a more

recent concept in de novo drug design, which allow the user to rapidly explore chemical space and to generate

novel and potential molecules of interest, filtered for desirable properties. In this study, we have implemented

deep and recurrent neural networks (RNNs) to develop generative model of sequences for SMILES strings from

a dataset of known active compounds. The learned pattern probabilities are used for generation of de novo

SMILES. Transfer learning approaches were implemented further to fine-tune the RNN's predictions. We have

implemented generative RNN-LSTM modelling for multiple scenarios such as targets with huge data sets, also

targets with small-data sets,  followed by fragment based molecular design. During the presentation, we will be

sharing our learnings on generative chemistry on a diverse range of targets, targets with small data sets,

observations, and limitations.

106 | EFMC-ISMC

LE063

THERAPIES FOR GRAM-NEGATIVE BACTERIAL INFECTIONS:

NEW APPROACHES AND FURTHER GENERATIONS OF EXISTING

SERIES

Michael J. Dawson

Antimicrobial Research Consultancy, Hitchin, UK

The need for new antibiotics is driven by two imperatives. Antimicrobial resistance not surprisingly grabs most

of the headlines, but we also need to improve the efficacy of therapies against some infections which are not

well-treated even when the pathogens are susceptible.  

Despite a strong and diverse therapeutic armamentarium with activity against Gram-positive bacteria, therapy of

many infections remains challenging. Whilst susceptibility in the laboratory given the alternatives available is

rarely the issue, achieving adequate drug concentrations at the target site, preventing damage to the host by

multiple toxins, dealing with biofilms and intracellular pathogens, whilst minimising collateral damage to the

normal microbiome all remain key challenges.

Additionally with Gram-negative bacteria pan-resistance is never far away. Whilst new developments to existing

classes of antibiotics are playing an important role and saving lives the path to resistance is well-precedented.

The major challenge here is the two membranes sandwiching a periplasm rich in antibiotic-degrading enzymes

and efflux pumps. Due to these barriers we need relatively high external drug concentrations to drive entry and

we need to achieve these concentrations at diverse body sites where infection may occur, leading to high doses

and a difficult balance of efficacy versus toxicity.

Our new drugs must not only be active against emerging pathogens and new strains resistant to the existing

armamentarium, but must be non-inferior for the treatment of susceptible strains to the antibiotic classes that

have been honed through multiple generations of improvements over the last 50+ years. Against this background

it is no surprise that most projects select advanced starting points from previous generations and aim to add the

additional functionality needed. 

Nevertheless, there are also exciting new developments in terms of new targets, new drug classes for existing

targets and even new modalities which treat infection in fundamentally different ways. Amongst these are agents

which have potential to raise the bar for treatment efficacy as well as to extend the status quo to MDR bacteria.   

The presentation will aim to review some selected developments of interest. Diversity of approaches will be

illustrated by an example of improving an existing class, the polymyxins, as well as a new discovery strategy

involving phage display of bicyclic peptides as leads for new antibacterials (Bicycles®). 

EFMC-ISMC | 107

LE064

TETHERED MACROCYCLIC PEPTIDES, A NOVEL ANTIBIOTIC

CLASS TARGETING ACINETOBACTER BAUMANNII

Patrizio Mattei

Small Molecule ResearchpRED Therapeutic ModalitiesRoche Innovation Center Basel

CH-4070 Basel

In the light of the rapidly rising number of people dying from bacterial infections and the lack of effective

antibiotics, the Tethered Macrocyclic Peptide (MCP) Acinetobacter baumannii project addresses the urgent need

to fight multi-drug resistant bacteria.

The Tethered Macrocyclic Peptides represent a structurally distinct compound class selectively targeting

Acinetobacter baumannii.  The lecture will present the discovery and optimization of this compound class

culminating in potent molecules that in vitro overcome pre-existing antibiotic resistance mechanisms and show

in vivo efficacy in murine models of infection.

108 | EFMC-ISMC

LE065

DISCOVERY OF SUBMICROMOLAR INHIBITORS OF THE

VIRULENCE FACTOR LASB FROM PSEUDOMONAS AERUGINOSA

USING RATIONAL DESIGN

Anna K. H. Hirsch (1,2)

1) Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) – Helmholtz Center for Infection Research (HZI),Department of Drug Design and Optimization, Campus Building E8.1, 66123 Saarbrücken, Germany

2) Department of Pharmacy, Saarland University Campus Building E8.1, 66123 Saarbrücken, Germany

Pseudomonas aeruginosa is a Gram-negative bacterium, typically affecting the lungs, urinary tract and wounds,

leading to severe infections. Treatment is becoming increasingly challenging due to the rapid emergence of

drug-resistant strains. Recently, significant efforts have been put into the development of the ‘pathoblockers’ –

agents capable of blocking bacterial virulence by disarming the pathogen, rather than killing it. Among a vast

number of virulence factors secreted by P. aeruginosa, elastase (LasB) plays a crucial role in the infection

process and is considered as a promising target for the development of new inhibitors.

1,2

 

Here, we report on the structure-based optimization of our N-arylmercaptoacetamides, resulting in two highly

potent chemical classes. We pursued rigidification and a structure-based fragment growing. Freezing the active

conformation in the form of succinimides enhanced the activity toward LasB two-fold compared to our

previously published inhibitors and improved chemical stability with regard to disulfide formation.

3,4

On the

other hand, exploiting structure-based design, fragment growing of the original hit led to a substantial 16-fold

boost in activity.

5

In addition to the substantial increase in the potency, our new derivatives show no cytotoxicity

and are highly selective for the bacterial metalloproteases over human matrix metalloproteases. Having

demonstrated an excellent in vivo effect in a Galleria mellonella infection model, one of the selected inhibitors

was further evaluated for its pharmacokinetic profile in mice and was subjected to an advanced SafetyScreen44

panel. Taken together, our inhibitors hold a lot of potential as novel therapeutics in the form of an adjunctive

therapy for P. aeruginosa-derived infections. 

References

1) Bassetti et al. Drugs Context 2018, 7:212527.

2) Calvert et al. J. Org. Chem. 2018, 14, 2607–2617.

3) Kany et al. ACS Infect. Dis. 2018, 4, 988–997.

4) Konstantinović et al. J. Med. Chem. 2020, 63, 8359–8368.

5) Ducho C., Hartmann R.W., Haupenthal J., Hirsch A.K.H, Kany A.M., Kaya C., Konstantinović J., Voos K., Walter I.,

Yahiaoui S. 2020. Germany, 25th August 2020. EP 20 192 608.6. Patent Pending.

EFMC-ISMC | 109

LE066

DOTAM-BASED SIDEROMYCINS TO VISUALIZE AND TREAT MDR

BACTERIAL PATHOGENS IN VIVO AND IN VITRO

Carsten Peukert (1,2), Dr. Kevin Ferreira (1,2), Anna Tutov (1,2), Bianka Karge (1), Dr. Hans Prochnow

(1), Dr. Sven-Kevin Hotop (1), Dr. Hazel Fuchs (1), Laura Langer (4), Prof. Dr. Tobias Ross (4), Prof. Dr.

Mark Brönstrup (1,2,3)

1) Department of Chemical Biology, Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany2) Institute for Organic Chemistry, Leibniz University Hannover (LUH), 30167 Hannover, Germany

3) Deutsches Zentrum für Infektionsforschung (DZIF), 38124 Braunschweig, Germany4) Radiopharmaceutical Chemistry, Medical School Hannover (MHH), 30625 Hannover, Germany

Infections caused by MDR Gram-negative bacteria contribute to the global increase of mortality and morbidity.

Remarkably, all pathogens that received a ‘critical’ status by the recently established WHO priority list were

Gram-negative species.

[1]

The reasons for the limited success of pharmaceutical research programs to generate

Gram-negative effective antimicrobials have been evaluated: The main scientific hurdle is the limited

understanding how to assure a sufficient translocation of bioactive molecules across the complex Gram-negative

cell wall.

[2]

[3]

Therefore, the development of innovative as well as specific approaches to identify, target and

treat Gram-negative bacterial pathogens represents an imminent challenge for medicinal chemistry.

In this work, we aimed to develop targeted conjugates with an improved bacterial penetration by exploiting the

bacterial iron transport machinery.

[5]

Ferric iron, a poorly available and essential nutrient for metabolism and

growth, is complexed by so-called siderophores - small-molecule, bacterial chelators, that harbor a strong

affinity for Fe

3+

. In a host organism, these siderophores sequester ferric iron from transferrin or hemes and

subsequently deliver the metal into bacteria, thereby enabling bacterial growth and pathogenesis. The Fe

3+

-complexed siderophore is recognized by a bacterial ligand-specific outer membrane receptor and actively

transported into the periplasm, released and made bioavailable.

[5] [6] 

In a Trojan Horse approach, (synthetic) Fe

3+

-siderophores can be loaded with effectors and exploited for theranostic purposes. Furthermore, it was shown

that the rule-of-thumb that only molecules with a weight of approx. 600 Da or lower can passively diffuse into

Gram-negative bacteria is not valid for actively transported siderophore conjugates.

[7]

[8] 

The

1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic amide (DOTAM), an established, synthetic siderophore is

employed as a carrier/targeting moiety, showing up to 500-times higher bacterial penetration in comparison to

the free payload. The DOTAM siderophore enables efficient targeting of MDR Gram-negative pathogens and

was successfully applied in a variety of approaches:

(I) Semi-synthetically modified inhibitors of gyrase and RNA-polymerase as well as lipopeptide antibiotics are

attached to the DOTAM siderophore, either by covalent or self-immolative linkers. These conjugates are

evaluated for their bacterial penetration and antimicrobial efficacy under iron-reduced cation-adjusted

conditions.

(II) Variation of the DOTAM siderophore with subsequent screening in siderophore-deficient E. coli mutants

yielded a first generation of PET tracers, which significantly distinguished microbial infections from sterile

inflammation in a first in vivo experiment.

(III) The combination of antimicrobial conjugates with imaging entities yielded a first generation of theranostics.

Taken together, the afforded antimicrobials and in particular the theranostics could allow for strain-specific

treatment and monitoring of bacterial infections, addressing a major-medical need expressed by the WHO.

References

1) E. Tacconelli, E. Carrara, A. Savoldi, S. Harbarth, M. Mendelson, D. L. Monnet, C. Pulcini, G. Kahlmeter, J. Kluytmans,

Y. Carmeli, M. Ouellette, K. Outterson, J. Patel, M. Cavaleri, E. M. Cox, C. R. Houchens, M. L. Grayson, P. Hansen, N.

Singh, U. Theuretzbacher, N. Magrini, W. H. O. P. P. L. W. Group, Lancet Infect. Dis. 2018, 18, 318-327.

2) R. Tommasi, D. G. Brown, G. K. Walkup, J. I. Manchester, A. A. Miller, Nat. Rev. Drug Discov. 2015, 14, 529-542.

3) D. J. Payne, M. N. Gwynn, D. J. Holmes, D. L. Pompliano, Nat. Rev. Drug Discov. 2007, 6, 29-40.

4) S. S. Kelkar, T. M. Reineke, Bioconjug. Chem. 2011, 22, 1879-1903.

5) U. Mollmann, L. Heinisch, A. Bauernfeind, T. Kohler, D. Ankel-Fuchs, BioMetals 2009, 22, 615-624.

6) J. H. Crosa, A. R. Mey, S. M. Payne, Iron transport in bacteria, ASM Press, Washington, 2004.

7) P. Klahn, M. Bronstrup, Nat. Prod. Rep. 2017, 34, 832-885.

8) G. S. Tillotson, Infect. Dis. (Auckl.) 2016, 9, 45-52.

110 | EFMC-ISMC

LE067

TARGETING RNA WITH DRUG-LIKE SMALL MOLECULES

Jennifer Petter

Arrakis Therapeutics828 Winter St

Waltham, MA 02451USA

RNA offers a broad array of folded, three-dimensional structures that mediate or regulate the functional roles

played by those RNAs. Our drug discovery platform at Arrakis Therapeutics is directed at the intervention of

those functions to therapeutic benefit using drug-like small molecules that bind the folded, RNA structures.

Specifically, we have focused on binding to and modulating the function of structures in pre-mRNA and mRNA

that govern splicing and translation, respectively.

The construction of this broad discovery platform presents many unique challenges: characterization of

endogenous RNA structures, sub-target selection and validation, high-throughput screening, assessment of target

engagement, new cellular assays, and demonstration of on-target mechanism. This presentation will touch on

some of these challenges, describing novel methods and provide early data on specific RNA targets. Particular

attention will be given to methods for the demonstration of target engagement.

EFMC-ISMC | 111

LE068

DRUGGING RNA MODIFYING ENZYMES – METTL3 INHIBITORS

David Hardick

Storm Therapeutics LtdBabraham Research Campus

CambridgeU.K.

METTL3 is an RNA methyltransferase that is responsible for the deposition of N-6-methyladenosine (m

6

A) on

selected mRNA targets, thereby regulating their stability and translation. We, and others, have recently shown

that METTL3 is a promising therapeutic target for acute myeloid leukaemia (AML), see Yankova et al, 2021*.

We have developed and characterised novel, selective, small molecule inhibitors of METTL3, and used them to

demonstrate their potential therapeutic utility to treat AML and solid tumours.

Chemical starting points for METTL3 inhibitors were identified from both unbiased HTS and knowledge-based

approaches. Using structure-guided design, potent and selective small molecule inhibitors of METTL3 from

distinct chemical series were prepared.  Optimised compounds were assessed for their ability to inhibit m

6

A

formation on mRNA in vivo using mass spectrometry and the best compounds from these PK/PD studies were

progressed to in vivo disease models. Testing these inhibitors in a range of in vivo cancer studies demonstrated

profound efficacy in AML models, including AML PDX models, as well as inhibition of cancer xenograft

growth in solid cancer models.

The route towards identification of key METTL3 inhibitors will be presented, with some of the challenges

encountered and how these were overcome.  Some inhibitor SAR will be described in the presentation along

with key in vivo data from selected cancer models.

*Yankova et al, Nature DOI:10.1038/s41586-021-03536-w

112 | EFMC-ISMC

LE069

SPLICE-SWITCHING SMALL MOLECULES AS INDUCERS OF

APOPTOSIS

Emma Campbell (1), Olivia I. Rutherford (1), Andrea Taladriz-Sender (1), Carika Weldon (2), Laurence

Hurley (3), Cyril Dominguez (2), Ian C. Eperon (2), Glenn A. Burley (1)

1) Department of Pure and Applied Chemistry, University of Strathclyde, Glasgow, UK2) Department of Biochemistry, University of Leicester, Leicester, UK

3) College of Pharmacology and Toxicology, University of Arizona, Tucson, Arizona 85721, United States

The Bcl-2 protein family are essential regulators of apoptosis.[1] One such protein, Bcl-x, is of particular interest

as a therapeutic target. It has two splicing isoforms: pro-apoptotic Bcl-xS and the anti-apoptotic Bcl-xL, the

latter of which is upregulated in a variety of cancers.

Exogenous regulation of Bcl-x splicing to bias the pathway towards the pro-apoptotic Bcl-xS isoform could

provide a new, novel mechanism for cancer therapy (Figure 1).[2,3]

 

Figure 1: Alternative splicing pathways of Bcl-x towards the shorter isoform (Bcl-xS) and the longer isoform

(Bcl-xL). Structure of GQC-05 (a known splice-switcher of Bcl-x) shown in the box.[4]

A focused structure-activity-relationship profile revealed key functional requirements for splice-switching

activity in this panel of ellipticine compounds. A one-pot, modular route for the synthesis of ellipticine

analogues was developed to allow access to an extensive library of small molecules, which can be used to probe

the mechanism of binding and act as potential therapeutic leads. In this poster, we present a suite of small

molecules that induce switching of the splicing pathway of Bcl-x in favour of the production of the pro-apoptotic

Bcl-xS isoform.[4]

References

1) Yip, K. W.; Reed, J. C. Bcl-2 Family Proteins and Cancer. Oncogene, 2008, 27, 6398 – 6406.

2) Naryshkin, N. et al. SMN2 Splicing Modifiers Improve Motor Function and Longevity in Mice with Spinal Muscular

Atrophy. Science, 2014, 345, 6197.

3) Sivaramakrishnan, M. et al. Binding To SMN2 Pre-mRNA-Protein Complex Elicits Specificity For Small Molecule

Splicing Modifiers. Nat. Commun., 2017, 8.

4) Weldon, C. et al. Identification Of G-Quadruplexes In Long Functional RNAs Using 7 Deazaguanine RNA. Nucleic Acids

Res., 2018, 46, 886 – 896.

EFMC-ISMC | 113

LE070

MODULATING VIRAL RNAS WITH AMILORIDE SMALL

MOLECULES

Amanda Hargrove

Duke UniversityChemistry Department

124 Science DriveDurham, NC 27708 USA

Viral RNA genomes contain three-dimensional structures that are critical for replication and merit consideration

as drug targets. This is particularly true for positive-sense RNA viruses, which must directly hijack translation

machinery upon entry into the cell and whose transcripts are present at high levels during replication. As part of

our efforts to improve small molecule targeting strategies for RNA and gain fundamental insights into small

molecule:RNA recognition, we have analyzed patterns in both RNA-biased small molecule chemical space and

RNA topological space privileged for differentiation. We recently applied these principles to functionally

modulate conformations of an enterovirus (EV71) IRES structure, where we identified an amiloride small

molecule that bound the EV71 IRES SLII bulge structure and decreased viral translation and replication in

cell-based studies in a dose-dependent manner with no significant toxicity. Structural, biophysical, and

biochemical characterization supported an allosteric mechanism in which DMA-135 induces a dramatic

conformational change in the RNA structure that stabilizes a ternary complex with the AUF1 protein that

represses translation. This mechanism was further supported by pull-down experiments in cell culture.

We have now expanded this strategy to target functional RNAs in the SARS-CoV-2 genome and have identified

amiloride-based small molecules that inhibit OC43 and SARS-CoV-2 replication through targeting of conserved

structured elements within the viral 5’-end. Dual luciferase assays supported reduction of viral translation, and

NMR-based structural studies revealed specific amiloride interactions with stem loops 1, 4, 5a, and 6, all of

which contain bulge like structures and were predicted to be strongly bound by the lead amilorides in

retrospective docking studies. As such, amilorides represent the first antiviral small molecules that target RNA

structures within the 5’-UTR and proximal region of the CoV genomes.

This combined work supports modulation of RNA conformational and structural landscapes as a promising

method to impact viral RNA function.

114 | EFMC-ISMC

LE071

DISCOVERY AND DEVELOPMENT OF A MKK-4 INHIBITORS TO

INCREASE LIVER REGENERATION

Stefan Laufer (1), Bent Praefke (1), Philip Klövekorn (1), Michael Juchum (1), Sabrina Klotz (2), Julia

Maier (6), Roland Selig (3), Wolfgang Albrecht (3), Lars Zender (2,4,5)

1) Department of Pharmaceutical/Medicinal Chemistry, Eberhard Karls Universität, Auf der Morgenstelle 8, TübingenCenter for Academic Drug Discovery, 72076 Tübingen, DE, Germany

2) Department of Medical Oncology and Pneumology (Internal Medicine VIII), University Hospital Tuebingen, Tuebingen,DE, Germany

3) HepaRegenix GmbH, Eisenbahnstraße 63, D-72072 Tübingen4) Cluster of Excellence ‘Image Guided and Functionally Instructed Tumor Therapies’ (iFIT), Eberhard Karls University of

Tübingen, Tübingen, Germany5) German Consortium for Translational Cancer Research (DKTK), Partner Site Tübingen, German Cancer Research

Center (DKFZ), Heidelberg, Germany6) Department of Pharmaceutical Biotechnology, Eberhard Karls Universität, Auf der Morgenstelle 8, 72076 Tübingen, DE,

Germany

Currently, the therapeutic options for treatment of liver failure are very limited. In an in vivo RNAi screen,

mitogen-activated protein kinase kinase 4 (MKK4) has recently been identified as a major regulator in

hepatocyte regeneration. By functional genetic silencing of MKK4. the target was validated in various

experimental disease models. These data strongly supported the concept, that selective MKK4-inhibition with a

small molecule-represents a promising and attractive approach for treatment of a complex and multi-factorial

disease. Further to the observation that the approved BRAF

V600E

inhibitor vemurafenib shows a high affinity to

and moderate functional inhibition potency against MKK4, our hit optimization concept included classical

iterative SAR-optimization but also  a scaffold-hopping approach by changing the core heterocycle from 1H-pyrrolo[2,3-b]pyridine to 1H-pyrazolo[2,3-b]pyridine. Both approaches followed a mandatory multiparameter

optimization. In vivo RNAi experiments also revealed, that MKK7 and JNK1 are anti-targets and thereby

defined the specification regarding the kinase selectivity. In both series, highly selective MKK4-inhibitors down

to low nanomolar range with excellent selectivity profile against MKK7/JNK1 could be achieved.

LN3118 was identified as a tool compound with an IC

50

-value against MKK4 of 0,1 µM and an attractive

selectivity profile and was used to validate MKK4 as a druggable target for treatment of liver disease. In

experimental 2/3-hepatectomy in the mouse, orally administered LN3118 achieved a two-fold increase of

hepatocyte proliferation. In acute CCl

4

-induced liver injury, cell death was prevented by 70%. Furthermore,

LN3118 demonstrated therapeutic activity in two subchronic animal models. LN3118 reduced alcohol-induced

steatosis and significantly reversed CCl

4

-induced liver fibrosis.

The latest generation of compounds included highly potent MKK4-inhibitors with a pharmacological,

toxicological and pharmacokinetic profile, which meet the specification of a clinical development candidate.

Latest profiling and selection strategy for candidate selection will be presented.

References

1) Wuestefeld, T.; Pesic, M.; Rudalska, R.; Dauch, D.; Longerich, T.; Kang, T.-W.; Yevsa, T.; Heinzmann, F.; Hoenicke, L.;

Hohmeyer, A.; Potapova, A.; Rittelmeier, I.; Jarek, M.; Geffers, R.; Scharfe, M.; Klawonn, F.; Schirmacher, P.; Malek, Nisar

P.; Ott, M.; Nordheim, A.; Vogel, A.; Manns, Michael P.; Zender, L. A Direct In Vivo RNAi Screen Identifies MKK4 as a

Key Regulator of Liver Regeneration. Cell 2013, 153, 389-401.

2) Willenbring, H.; Grompe, M. A Therapy for Liver Failure Found in the JNK Yard. Cell 2013, 153, 283-284.

3) Kloevekorn, P.; Pfaffenrot, B; Juchum, M; Selig, R; Albrecht, W; Zender, L; Laufer, SA: From off-to on-target: New

BRAF-inhibitor-template-derived compounds selectively targeting mitogen activated protein kinase kinase 4 (MKK4),

EUROPEAN JOURNAL OF MEDICINAL CHEMISTRY (2021) DOI: 10.1016/j.ejmech.2020.112963

4) Bent Pfaffenrot; PhilipKlövekorn; Michael Juchum; Roland Selig; Wolfgang Albrecht; Lars Zender; Stefan A.Laufer:

Design and Synthesis of 1H-pyrazolo[3,4-b]pyridines targeting mitogen-activated protein kinase kinase 4 (MKK4) - a

promising target for liver regeneration; European Journal of Medicinal Chemistry,in press, available online 17 March 2021

EFMC-ISMC | 115

LE072

TARGETING ONCOGENIC BRAF USING A NOVEL ALLOSTERIC

SITE

Bogos Agianian, Xiomaris Cotto-Rios, Nadege Gitego, Emmanouil Zachariouadakis, Evripidis

Gavathiotis

Albert Einstein College of Medicine, New York, USA

 

BRAF mutants are present in ~50% of melanoma patients as well as in colorectal (~10%), thyroid carcinomas

(~40%) and other malignancies. FDA-approved RAF inhibitors Vemurafenib, Dabrafenib and Encorafenib show

remarkable responses in melanoma patients with BRAF

V600E

tumors. However, these drugs ultimately become

ineffective due to adaptive and acquired resistance mechanisms, many of which depend on signaling by poorly

inhibited BRAF homo- or hetero-dimers. In about 20-30% of melanoma tumors, intrinsic resistance is driven by

a constitutive BRAF dimer splice variant, p61-BRAF

V600E

, while in resistant colorectal and thyroid tumors RAF

dimerization is maintained by frequent RAS activation. An adaptive resistance mechanism depends on

inhibitor-induced relief of ERK-mediated feedback pathway inhibition, which stimulates RAS-GTP and

increases RAF dimers. Finally, oncogenic non-V600E BRAF species in some tumors signal as obligated dimers.

Challenged by the clinical relevance of BRAF dimerization, we initiated a systematic quest for novel inhibitors

that specifically recognize BRAF

V600E

dimers. We developed a phenotypic cellular assay to screen for inhibitors

of ERK-signaling, using Vemurafenib-resistant melanoma cells that express p61-BRAF

V600E

dimers. We

discovered new potent BRAF inhibitors, including a FDA approved kinase inhibitor. We obtained crystal

structures of this drug with BRAF

V600E

and BRAF

WT

and identified an unprecedented binding interaction of the

drug with a distinct αC-helix conformation of BRAF kinase. Using these structural insights and scaffold-based

drug-design methods we developed an orally bioavailable PHI1 inhibitor. Remarkably, the crystal structure of

BRAF

V600E

/PHI1 complex revealed that this compound engages a new BRAF allosteric site, which uniquely

modulates the αC-helix conformation. In vitro and cellular studies showed that PHI1 is potent and selective

against cells with BRAF mutant dimers and has no activity for BRAF

 

monomers. In addition, SAR analysis

indicated that selectivity for dimer inhibition is lost upon modification of PHI1’s moiety targeting the allosteric

pocket, suggesting that this site modulates specificity for BRAF dimers. Co-treatment experiments revealed that

PHI1 inhibits the second site within the dimers with positive cooperativity driven from first site occupancy, in

contrast to current RAF inhibitors that exhibit no, or negative, cooperativity. Based on these data, co-treatment

of allosteric BRAF inhibitors with various clinical RAF and MEK inhibitors was investigated in BRAF

dimer-dependent tumors resulting in promising synergistic activity. This work provides a new class of allosteric

BRAF inhibitors that may specifically diminish clinically untargeted oncogenic signaling dependent on BRAF

dimers.

116 | EFMC-ISMC

LE073

FURAN-OXIDATION MEDIATED TECHNOLOGY: FROM IN VITRO

ANALYSIS OF PROTEIN-PROTEIN INTERACTIONS TO

GPCR-LIGAND INTERACTIONS ON LIVE CELLS

Laia Miret-Casals (1), Willem Vannecke (1), Kurt Hoogewijs (1), Marta Vilaseca (3), Christophe Ampe

(2), Marleen Van Troys (2), Annemieke Madder (1)

1) Organic and Biomimetic Chemistry Research Group, Ghent University, Faculty of Science, B-9000 Ghent, Belgium2) Department of Biomolecular Medicine, Faculty of Medicine and Health Sciences, Ghent University, B-9000 Ghent,

Belgium3) Institute for Research in Biomedicine, The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain

Our research group has developed a novel furan cross-link technology for oligonucleotides, further applicable to

the investigation of peptide-protein interactions. Furan, a small aromatic compound, is incorporated in peptides

by means of the unnatural amino acid 2-furyl-L-alanine. The furan moiety can be selectively oxidized to a

reactive aldehyde upon generation of singlet oxygen. A site-selective cross-link reaction can then occur between

the oxidized furan moiety and sulfhydryl and/or amine groups present in the target protein if sufficiently

proximal. Here, we describe the application of this fast and highly efficient furan-oxidation mediated technology

to protein-protein

1

and peptide-protein

2

interactions in two different systems.

Initially, we studied the weak and dynamic protein-protein interaction between actin, the major cytoskeletal

protein of the cell that forms filaments, and Tß4 that regulates the polymerization of actin and keeps actin in the

monomeric form. Furan-armed-Tß4 analogues were still able to sequester monomeric actin with comparable

capacity as wildtype Tß4 and were shown to efficiently cross-link to monomeric actin upon singlet oxygen

generation

1

. The cross-linked Fua-Tß4/actin complex was separated on SDS-PAGE and submitted to in-gel

tryptic digestion. The isolated tryptic peptides were analyzed by MS-analysis and searches for cross-linked

peptides were performed using xiSEARCH v1.7.6.1. A cross-link is formed between Tß4-Fua24 and

Actin-Lys61 and a plausible chemical structure of the covalent linkage between Lys and the activated Fua was

confirmed by the MS-data

1

.

The furan cross-link technology was further optimized to enable cross-linking of furan-modified peptide ligands

to GPCR proteins on live cells relying on the spontaneous endogenous oxidation of the furan moiety. We studied

the neuropeptide kisspeptin-10 and its G-protein coupled receptor GPR54, which play a role in breast cancer and

in the regulation of mammalian reproduction. We described selective cross-linking of a furan-modified

kisspeptin-10 analogue to its membrane receptor GPR54 in live cells, with no toxicity and high efficiency

2,3

. In

addition, we have been able to pull-down the peptide-receptor complex using a Biotin-furan-modified

kisspeptin-10. Currently, we are working towards characterization of the cross-linked peptide by MS analysis.

References

1) Miret-Casals, L; Vannecke, W; Hoogewijs, K; et al. Chem. Commun., 2021, 57, 6054-6057.

2) Vannecke, W; Van Troys, M; Ampe, C; Madder, A. ACS Chem. Biol. 2017, 12, 2191-2200.

3) Van Troys, M; Vannecke, W; Ampe, C; Madder, A. G Protein-Coupled Receptor Signaling/Methods and Protocols. 2019,

1947, 81–102.

EFMC-ISMC | 117

LE074

LNP023: DISCOVERY AND SYNTHESIS OF A FIRST-IN-CLASS,

ORAL FACTOR B INHIBITOR FOR TREATMENT OF RARE RENAL

AND HEMATOLOGICAL DISEASES

Anna Schubart (1), Karen Anderson (2), Holger Sellner (1), Christopher Adams (2), Nello Mainolfi (2),

Rajeshri Karki (2), Finton Sirockin (1), Philipp Lustenberger (1), Stefanie Flohr (1)

1) Novartis Institutes for BioMedical Research, Novartis Pharma AG, CH-4056 Basel, Switzerland2) Novartis Institutes for BioMedical Research, Cambridge, Massachusetts 02139, United States

The alternative pathway (AP) of the complement system is a key contributor to the pathogenesis of several

human diseases including age-related macular degeneration, paroxysmal nocturnal hemoglobinuria (PNH),

atypical hemolytic uremic syndrome (aHUS), and various glomerular diseases. The serine protease factor B (FB)

is a key node in the AP and is integral to the formation of C3 and C5 convertase. Despite the prominent role of

FB in the AP, selective orally bioavailable inhibitors, beyond our own efforts, have not been reported previously.

We report the discovery of LNP023 from a fragment-like hit by structure based drug discovery. LNP023 is a

First-in-Class oral LMW Factor B inhibitor with excellent selectivity against proteases, enzymes, GPCRs and

transporters. We will share efficacy data from animal models, patient samples as well as from ongoing clinical

studies. LNP023 displays an excellent preclinical and clinical safety profile: the hERG liability related to the

central piperidine core was successfully mitigated by a zwitterionic approach. In the development of the large

scale synthesis biocatalysis was introduced. 

118 | EFMC-ISMC

LE075

ANTIBODY-DRUG CONJUGATES (ADC) AND SMALL

MOLECULE-DRUG CONJUGATES (SMDC): A COMPARATIVE

EVALUATION

Dario Neri

CEO of Philogen (www.philogen.com) and Professor of ETH ZürichVia Bellaria, 35

53018 Sovicille (SI) – ITALY

Cytotoxic agents used for conventional cancer therapy do not preferentially localize to the tumor site. This

pharmacokinetic limitation may limit dose escalation to therapeutically active regimens and may cause

unnecessary toxicity. For many application it may be desirable to couple cytotoxic drugs to antibodies or to

small organic ligands, specific to a suitable accessible tumor-associated antigen, so that the resulting conjugates

may preferentially accumulate at the site of disease, helping spare normal tissues.

In this talk, I will present results from a collaboration between my group at ETH Zürich and the group of Dr.

Samuele Cazzamalli at Philochem AG, in which we aimed at comparing the performance of antibody-drug

conjugates (ADCs) and small molecule-drug conjugates (SMDCs).  Both classes of therapeutics can be

extremely efficacious for the treatment of "difficult-to-cure" tumors in animal models of the disease. However,

in our experience, SMDCs extravasate from tumor blood vessels more rapidly than ADCs. This feature leads, in

many cases, to a higher drug uptake in the tumor, to better tumor:organ ratios and to a deeper penetration of the

conjugate into the neoplastic mass.

I will present results from preclinical comparisons of ADCs and SMDCs, as well as translational activities, in

which the tumor-targeting properties of selected candidates were evaluated both in mouse models of cancer and

in patients.

References

1) Cazzamalli et al. (2018) J. Am. Chem. Soc., 140: 1617

2) Millul et al. (2021) Proc. Natl. Acad. Sci. U.S.A., 118: e2101852118

EFMC-ISMC | 119

LE076

NUCLEIC ACIDS AS HOUSEHOLD MEDICINE: LIVING IN THE

WORLD OF RNA THERAPEUTICS

Muthiah Manoharan

Alnylam Pharmaceuticals, Cambridge, MA [email protected]

Synthetic small interfering RNAs (siRNAs) are potent inhibitors of gene expression; these agents act through the

natural RNA interference (RNAi) pathway.  Our laboratory demonstrated the very first in vivo RNAi mediated

silencing in mouse liver and jejunum in 2004. To deliver therapeutic siRNAs into liver hepatocytes, we have

further developed a three-pronged approach with the goals of enabling delivery to hepatocytes after both

intravenous and subcutaneous administration. These methods include chemical modification of siRNAs, lipid

nanoparticle (LNP) formulation of siRNAs, and multivalent N-acetylgalactosamine (GalNAc) conjugation of

siRNAs.

The LNP strategy with a partially chemically modified siRNA resulted in the first RNAi therapeutic,

ONPATTRO

®

, approved in 2018 is used to treat polyneuropathy in patients with hereditary ATTR amyloidosis.

 The approval of ONPATTRO

®

paved the way for a whole new class of RNA-based medicines and further

validated LNP platform-based delivery of Nucleic Acids for human therapeutics including mRNA based

vaccines, Gene-Editing and related applications.

The Ashwell-Morell, or the asialoglycoprotein (ASGPR) receptor, was discovered in 1965 and fully

characterized in 1974. However, the human therapeutic utility of this receptor-ligand pair for delivery of nucleic

acids was fully realized only in 2019. By combining the chemical modifications of oligonucleotides with the

triantennary N-acetylgalactosamine (GalNAc) ligand, our research group enabled human therapeutic applications

of hepatocyte-targeting GalNAc-conjugated oligonucleotides. This delivery platform has revolutionized the

nucleic acid-based therapeutics field. Three GalNAc-conjugated RNAi therapeutics have been approved so far:

GIVLAARI® (givosiran, 2019) for treating acute hepatic porphyria, OXLUMO® (lumasiran, 2020) for the

treatment of primary hyperoxaluria type 1, and Leqvio® (inclisiran, 2020) for treatment of

hypercholesterolemia. Several other GalNAc-conjugated siRNAs such as vutrisiran are currently in advanced

clinical testing.  This presentation will cover the molecular basis of this success including the chemical

modifications used in each RNA strand to ensure Argonaute2 recognition, silencing efficiency, metabolic

stability, and safety. We will also present our current explorations of this platform driven by rational design of

novel chemical modifications and motifs.

References

1) Akinc, A.; Maier, M.A.; Manoharan, M.et al.; “The ONPATTRO story and the clinical translation of nanomedicines

containing nucleic acid-based drugs.” Nat. Nanotechnol. 2019, 14, 1084-1087.

2) Egli, M.; Manoharan, M. “Re-Engineering RNA Molecules into Therapeutic Agents.” Acc. Chem. Res. 2019, 52,

1036-1047.

3) Nair, J.K et al; “Multivalent N-Acetylgalactosamine-Conjugated siRNA Localizes in Hepatocytes and Elicits Robust

RNAi-Mediated Gene Silencing.” J. Am. Chem. Soc. 2014, 136, 16958-16961

4) Jayaraman, M. et al.; “Maximizing the potency of siRNA lipid nanoparticles for hepatic gene silencing in vivo.” Angew.

Chem. Int. Ed. 2012, 51, 8529-8533.

120 | EFMC-ISMC

LE077

OMICS-BASED DEVELOPMENT OF NANOMEDICINES FOR SAFE

AND EFFECTIVE DRUG DELIVERY IN CANCER

Erem Bilensoy

Hacettepe University Faculty of Pharmacy Department of Pharmaceutical Technology 06100 Ankara Turkey

Nanomedicines are the focus of research in the biopharmaceutical field especially after the formulation of

several Covid-19 vaccines with lipid nanoparticles. In fact, as of 2021 there are more than 60 different

nanomedicines on the global pharmaceutical market. These nanomedicines are considered non-biological

complex drugs in the sense that they alter the pharmacokinetic properties and the bioavailability of the active

molecule encapsulated in the nanoparticle structure. Both the API and the nanocarrier act as the pharmaceutical

product and affect the body at the molecular level. 

Proteomics and metabolomics data obtained from cells treated with nanoparticles can provide in-depth and new

information on how these nanomedicines affect the body with our without the entrapped drug. This lecture will

discuss several case studies involving the effect of non-ionic and polycationic nanoparticles on different breast

cancer ceel lines and hepatocellular carcinoma cells to elucidate the intrinsic apoptotic effects of the

nanoparticles and determine which metabolic pathways are induced by the nanoparticles prepared from different

amphiphilic cyclodextrin derivatives. Finally the effect of methotrexate in free form and bound to nanoparticles

will be covered through a global omics approach. 

QTOF-based untargeted or targeted protemic and metabolomic analysis can provide an important tool for the

development of non-biological complex drugs and nanoparticulate therapeutics and vaccines. 

Acknowledgement:This study was financially supported by The Scientific and Technological Research Council

of Turkey (TUBITAK) (Project ID:115S456)

 

References

1) Ercan A., Çelebier M., Varan G., Öncül S., Nenni M, Kaplan, O., Bilensoy E., “Global Omics Strategies to Investigate the

Effect of Cyclodextrin Nanoparticles on MCF-7 Breast Cancer Cells”, European Journal of Pharmaceutical Sciences,

https://doi.org/10.1016/j.ejps.2018.07.060 (2018)

2) Varol, I, Kaplan, O; Erdoğar, N; Öncül, S., Nielsen TT; Ercan, A; Bilensoy, E., Çelebier, M., Folate-Conjugated

Cyclodextrins Directly Intervene the Energy Metabolism in Triple Negative Breast Cancer Cells: An Untargeted

Metabolomics Study, Current Pharm. Analysis, DOI: 10.2174/1573412917999201020205745 (2021)

3) Ercan, A., Celebier, M., Oncul, S., Varan, G., Kocak, E., Benito, JM, Bilensoy, E., Polycationic Cyclodextrin

Nanoparticles Induced Cell Death on Hepatocellular Carcinoma Cells: An Evaluation at The Molecular Level, Int. J. Pharm.

https://doi.org/10.1016/j.ijpharm.2021.120379 (2021)

EFMC-ISMC | 121

LE078

TARGETING THE SPECIFICITIES OF THE TUMOR

MICROENVIRONMENT FOR CANCER THERAPY AND DIAGNOSIS

Sébastien Papot

Seekyo, Technopole Grand Poitiers, Site CEI, bat Téléport 1, 2, avenue Galilée, BP 30153, 86961 Futuroscope, France.University of Poitiers, UMR 7285 (IC2MP), 4 rue Michel Brunet, 86022 Poitiers, France.

E-mail: [email protected]

The controlled delivery of anticancer agents in malignant tissues is an emerging therapeutic strategy that reduces

dose-limiting adverse effects associated with traditional chemotherapy. The vast majority of drug delivery

systems have been designed to recognise a specific cell surface marker (e.g. antigens and receptors), penetrate

inside cancer cells through endocytosis and trigger the release of highly toxic compounds in response to an

intracellular biochemical stimulus. Numerous internalising ligand- and antibody-drug conjugates have been

assessed in human, leading to the marketing of several antibody-drug conjugates (ADCs) for applications in

oncology. However, the scope of such targeting devices is only restricted to the treatment of tumors expressing a

high level of the targeted cell surface marker and is limited by the high tumor heterogeneity. Moreover, ADCs

poorly penetrate solid tumors which represents a major drawback for their efficacy, hence highlighting the

urgent need for the development of novel therapeutic strategies.

Thus, the use of nontoxic drug delivery systems (DDS) that can be activated by an enzyme present in the tumor

microenvironment has been proposed as an attractive alternative to internalizing ligand- and antibody-drug

conjugates. In this approach, the active drug is released in the extracellular space following a catalytic process

and can then enter surrounding cancer cells whatever their membrane specificities, thus offering substantial

advantages due to the high heterogeneity of cells in malignant tissues.

In this context, we developed enzyme-responsive DDS programmed for (1) the transport in the body of potent

anticancer agents in an innocuous manner toward safe tissues, (2) the efficient recognition of enzymatic

specificities of the tumor microenvironment and (3) the controlled release of the parent drug exclusively at the

tumor site. In parallel, we also designed volatile organic compounds (VOC)-based probes targeting the same

malignant specificities for diagnostic and prognostic purposes.

Main results obtained in the course of this research program will be presented.

122 | EFMC-ISMC

LE079

TOWARD A NEW THERAPY FOR IMMUNE-MEDIATED TISSUE

INJURY

Julie Spicer (1), Jiney Jose (1), Christian Miller (1), Patrick O'Connor (1), Anna Giddens (1), Jagdish

Jaiswal (1), Stephen Jamieson (1), Matthew Bull (1), Eleanor Leung (2), Raymond Norton (2), Ruby Law

(3), James Whisstock (3), Hedieh Akhlaghi (4), Joseph Trapani (4), Geoff Hill (5), Karshing Chang (5),

Kate Gartlan (5)

1) Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, NewZealand

2) Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Melbourne, Australia3) Department of Biochemistry and Molecular Biology, Monash University, Clayton, Melbourne, Australia

4) Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Australia5) QIMR Berghofer Medical Research Institute, Queensland, Australia

The pore-forming protein perforin is a key component of the human immune response, performing a crucial role

in the granule exocytosis pathway employed by natural killer (NK) cells and cytotoxic T lymphocytes (CTL) to

eliminate virus-infected and transformed cells. However inappropriate CTL and NK cell activity has also been

implicated in autoimmune diseases (e.g., insulin-dependent diabetes) and therapy-induced conditions (e.g.,

allograft rejection, graft-versus-host disease). Because perforin is expressed exclusively by cells of the immune

system, inhibition of its activity could provide a highly selective strategy for the treatment of these conditions

compared to conventional immunosuppressive treatments that indiscriminately depress immune function. This is

especially so in the context of bone marrow stem cell transplantation where early rejection of

immunologically-mismatched grafts is driven by the recipient’s natural killer cells, which overwhelmingly use

perforin to kill their targets. In this presentation I will discuss our identification of a novel series of

benzenesulfonamide-based perforin inhibitors, detailing the in vitro structure-activity relationships and

optimisation of in vivo physicochemical and pharmacokinetic properties. This work resulted in the identification

of 1 - the first reported small molecule able to prevent rejection of transplanted bone marrow stem cells in vivo by blocking perforin function.

1

 

Reference 1: ‘Inhibition of the cytolytic protein perforin prevents rejection of transplanted bone marrow stem

cells in vivo.’ Spicer, J. A. et al, Journal of Medicinal Chemistry; DOI: 10.1021/acs.jmedchem.9b00881.

EFMC-ISMC | 123

LE080

A PHENOTYPIC SCREEN IDENTIFIES A SMALL MOLECULE THAT

INDUCES DIFFERENTIATION OF AML CELLS IN VITRO AND

SHOWS ANTI-TUMOUR EFFECTS IN VIVO

Laia Josa-Culleré (1), Thomas R. Jackson (2), Thomas J. Cogswell (1), Aini Vuorinen (1), Douzi Zhang

(2), Stephen G. Davies (1), Paresh Vyas (2), Thomas Milne (2), Graham M. Wynne (1), Angela J. Russell

(1)

1) Chemistry Research Laboratory, University of Oxford, 12 Mansfield Road, Oxford OX1 3TA, UK2) MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, NIHR Oxford Biomedical Research

Centre Haematology Theme, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DS, UK

Acute myeloid leukemia (AML) is the most aggressive type of blood cancer, and the second most common

leukemia in adults. AML arises from genetic mutation(s) which cause an arrest of differentiation at the

progenitor or precursor stage, blocking production of downstream blood lineages.

1

 The standard of care for

many years has been intensive cytotoxic chemotherapy, which often results in numerous side effects and low

rates of complete remission. An alternative approach that has gained much interest in the recent years is to

relieve the differentiation block of AML cells, instead of directly inducing apoptosis.

2

 Such therapies are

expected to be more effective and less toxic. However, the reported examples are targeted to specific mutations

and are only effective to narrow patient populations.

Our goal was to identify novel small molecules that induce differentiation to AML cells regardless of their

genetic status. For this purpose, we developed a robust in vitro screening assay which we utilised to perform a

pilot screen of over 1,000 small molecules. The screen was followed by a validation strategy which identified a

range of structurally distinct hits that could differentiate AML cells of several subtypes. An extensive medicinal

chemistry program to improve the properties of these compounds delivered highly potent molecules with

favorable physicochemical properties, enabling the progression to in vivo studies.

One of the compound classes proved to decrease tumour volume and increase survival in a xenograft model of

AML. Subsequent intensive mechanism of actions studies, including RNA sequencing and chemoproteomics,

identified the biological target driving the observed differentiation effect.

 

This work identified novel small molecules able to induce differentiation in wider patient populations, which

have the potential to be more effective and better tolerated than current agents. It also proposed an unknown role

of the identified target towards the induction of differentiation in AML.

References

1) Khwaja, A.; Bjorkholm, M.; Gale, R. E.; Levine, R. L.; Jordan, C. T.; Ehninger, G.; Bloomfield, C. D.; Estey, E.; Burnett,

A.; Cornelissen, J. J.; Scheinberg, D. A.; Bouscary, D.; Linch, D. C. Acute Myeloid Leukaemia. Nat. Rev. Dis. Prim. 2016, 2,

16010.

2) De Thé, H. Differentiation Therapy Revisited. Nat. Rev. Cancer 2018, 18 (2), 117–127.

124 | EFMC-ISMC

LE081

ISOFORM-SELECTIVE TAU TUBULINE KINASE 1 INHIBITORS

REDUCE TDP-43 HYPERPHOSPHORYLATION: A NEW HOPE IN

THE TREATMENT OF TDP-43 PROTEINOPATHIES

V. NOZAL (1), L. MARTÍNEZ-GONZÁLEZ (1), R. BENÍTEZ-FERNÁNDEZ (1), M.A.

MARTÍN-REQUERO (1), V. PALOMO (1,2), A. MARTÍNEZ (1,2)

1) Centro de Investigaciones Biológicas-CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain2) Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos

III, 28031 Madrid, Spain

TAR DNA binding protein of 43 kDa (TDP-43) aggregates are pathological hallmarks present in the 97% of

amyotrophic lateral sclerosis patients, independently of their familiar or sporadic origin. Within these inclusions

post-translational modifications such as ubiquitination, truncation and hyperphosphorylation are common

features. Few kinases have been described to be involved in this pathological phosphorylation: GSK3β, CK1,

CDC7, MAPK/ERK and TTBK1/2 

1

. TTBK1/2 are the most recently discovered kinases, therefore, few

inhibitors have been described so far and they have not been tested in the modulation of TDP-43 phosphorylation

in any cellular model 

2,3,4

.

In this work, we present the design, synthesis and biological evaluation of new TTBK1/2 inhibitors. We have

payed special attention to the selectivity of the new compounds towards isoform 1 which role in the human body

is related to pathological events, whereas TTBK2 is involved in ciliogenesis and other important physiological

functions. After filtering the synthetized compounds based on their kinase inhibition potency, their permeability

over the blood-brain barrier and their preliminary cellular activity in SH-5YSY and ALS patients cell lines, we

have obtained a hit compound that has been further evaluated in a tg TDP-43

A315T

mouse model. The reduction

of TDP-43 phosphorylation as well as immunomodulatory action and motoneurons preservation activity has

been confirmed for this hit compound and the role of TTBK1 inhibitors in the treatment of TDP-43

proteinopathies is demonstrated for the first time.

References

1) ACS Chem. Neurosci. 2019, 10, 3, 1183−1196. Palomo V. et al.

2) ChemMedChem. 2013, 8, 11, 1846-1854. Xue Y. et al.

3) Acta Crystallogr F Struct Biol Commun. 2014, 70, 2, 173-181 Kiefer S.E. et al.

4) PloS ONE, 2020, 15, 4, e0228771. Dillon G.M. et al.

EFMC-ISMC | 125

LE082

TARGET-TEMPLATED DE NOVO DESIGN OF DRUG-LIKE CYCLIC

PEPTIDES: PD-1/PD-L1 INHIBITORS

Salvador Guardiola (1,2), Ernest Giralt (1)

1) Institute for Biomedical Research (IRB), Barcelona Science Park, Spain2) Ona Therapeutics, Barcelona Science Park, Spain

Peptides, together with antibodies, are potent biochemical tools capable of modulating difficult protein-protein

interactions. However, current structure-based methods are largely limited to natural peptides and are not

suitable for designing target-specific binders with improved pharmaceutical properties, such as macrocyclic

peptides. In this work, we report a general framework that leverages the computational power of Rosetta for

large-scale backbone sampling and energy scoring, followed by side-chain composition, to design heterochiral

cyclic peptides that bind to a protein surface of interest. To showcase the applicability of our approach, we

identified two hits (PD-i3 and PD- i6) that target PD-1, a key immune checkpoint,

1

and work as protein ligand

decoys.

2

A comprehensive biophysical evaluation confirmed their binding mechanism to PD-1 and their

inhibitory effect on the PD-1/PD-L1 interaction.

3

 Finally, elucidation of their solution structures by NMR served

as validation of our de novo design approach. We anticipate that our results will provide a general framework for

designing target-specific drug-like peptides.

 

References

1) Chinai, J. M. et al. New immunotherapies targeting the PD-1 pathway. Trends Pharmacol. Sci. 36, 587–595 (2015).

2) Zak, K. M. et al. Structural Biology of the Immune Checkpoint Receptor PD-1 and Its Ligands PD-L1/PD-L2. Structure

25, 1163–1174 (2017).

3) Guardiola, S. et al. Target-templated de novo design of macrocyclic D-/L-peptides: discovery of drug-like inhibitors of

PD-1. Chemical Science (2021) doi 10.1039/D1SC01031J

126 | EFMC-ISMC

LE083

DISCOVERY OF LAS200019, A NOVEL TOPICAL JAK INHIBITOR

FOR THE TREATMENT OF INFLAMMATORY SKIN DISEASES

Elena Gomez

ALMIRALL, Medicinal ChemistryRDA General Mitre 151

08022 BarcelonaSpain

JAK proteins are key signaling molecules of several cytokine receptors. Oral JAK inhibitors are approved for the

treatment of cancer and inflammatory diseases in which they have proven to be efficacious but also to exhibit

significant adverse events related with immune-suppression, thromboembolism, gastrointestinal perforations and

alterations in laboratory parameters.

Topical drug delivery presents a huge opportunity for the treatment of inflammatory dermatological diseases.

Topical treatment with JAK inhibitors may cause local immunomodulation and lesion improvement,  with a

significant reduction or avoidance of systemic side effects previously described. This is particularly relevant in

indications such as atopic dermatitis, which is a pediatric indication demanding large safety margins. Thus, a

comprehensive medicinal chemistry exercise for the optimization of a series of aminopyrimidines that led to the

discovery of LAS200019 as a topical clinical candidate for the treatment of inflammatory skin diseases will be

presented. Key aspects of the development of topical drugs such as phototoxicity risk and solubility in topically

relevant solvents will be addressed.

 

EFMC-ISMC | 127

LE084

PRODRUGS STRATEGIES IN MEDICINAL CHEMISTRY

Jarkko Rautio

School of Pharmacy - University of Eastern Finland - Yliopistonranta 1 - FI-70211 Kuopio - Finland

The current interest in prodrugs is evident. In the past 10 years, the FDA has approved over 30 prodrugs, and

approximately 10% of all marketed drugs worldwide can be considered prodrugs. Prodrug strategies are versatile

and powerful tools to improve the problematic characteristics of molecules. Those have traditionally been

embarked to address ADME properties and risks of marketed drugs or as a tool in late-stage problem solving for

drug candidates in development phases. However, prodrug design is now being integrated into early drug

discovery.

Admittedly, embarking a prodrug strategy can certainly present its own challenges, but depending on the

chemical nature of the parent drug and the therapeutic target, many times the prodrug design can represents a

comparable smaller challenge than the alternative of searching for a new therapeutically active molecule that

also inherently possesses the desired ADMET properties. This presentation will demonstrate the versatility of

prodrugs strategies in medicinal chemistry to overcome various ADMET barriers with examples of successful

prodrugs developed in industry and in academia.

128 | EFMC-ISMC

LE085

NEW EXAMPLES FOR N-HYDROXYAMIDINE PRODRUGS

Bernd Clement

Christian-Albrechts-Universität zu Kiel, Department of Pharmaceutical and Medicinal Chemistry, Gutenbergstraße 76,24118 Kiel, Germany

The mitochondrial amidoxime reducing component (mARC) is the most recently discovered molybdenum

enzyme in mammals. In concert with the electron transport proteins, NADH cytochrome b5 reductase and

cytochrome b5 it reduces N-oxygenated structures [1]. This plays a major role in N-reductive drug metabolism

[2].

The discovery of the enzyme in my laboratory was connected to the activation of N-hydroxyamidines (=

amidoximes) as prodrug for amidines [1-3]. The old prodrug principle amidoximes instead of amidines [3] is

based on the lower basicity of amidoximes, which are not protonated under physiological conditions and are thus

absorbed from the gastrointestinal tract [1-3].

After absorption, the amidoximes are reduced to the active amidines by mARC. N-hydroxyguanidines can also

be used as prodrugs for guanidines [1-3].

In the lecture, new results of the structure and function of mARC1 and mARC2 will be presented [4-7].

Furthermore, new N-hydroxyamidine prodrugs like succinyl-dabigatran will be introduced using amidoxime

esters as prodrugs which are hydrolysed by esterases to amidoximes and then reduced to amidines by mARC1

and mARC2.

Dabigatran is an orally administered direct thrombin inhibitor and belongs to the class of anticoagulants. It has a

zwitterionic structure and is not absorbed after oral administration. It is used as dabigatran etexilate, a carbamate

prodrug, which is converted to dabigatran by esterase-catalysed hydrolysis in the liver and plasma [8]. Problems

of dabigatran etexilate and the active dabigatran are mainly based on its poor solubility [9, 10] and have been

solved by the development of the new prodrug succinyl-dabigatran. This prodrug of dabigatran has a similar

bioavailability and fast activation as dabigatran-etexilate, but it also shows excellent solubility in a relevant

physiological pH range. It can be used for intravenous injection, but also for an oral solution. F.ex. a pediatric

formulation is possible. 

The suitability of succinyl-dabigatran as a prodrug has been shown in animal experiments and in a first study in

humans. Succinyl-dabigatran is the first highly soluble representative of the new class of orally available

anticoagulants.

References

1) A. Havemeyer, F. Bittner, S. Wollers, R. Mendel, T. Kunze, B. Clement, Identification of the missing component in the

mitochondrial benzamidoxime prodrug converting system as a novel molybdemum enzyme. Journal of Biological Chemistry,

2006, 281, 34796-34802

2) S. Grünewald, B. Wahl, F. Bittner, H. Hungeling, S. Kanzow, J. Kotthaus, U. Schwering, R.R. Mendel, B. Clement, The

fourth molybdenum containing enzyme mARC: cloning and involvement in the activation of N-hydroxylated prodrugs.

Journal of Medicinal Chemistry, 2008, 51, 8173-8177

3) G. Ott, A. Havemeyer, B. Clement, The mammalian molybdenum enzymes of mARC. Journal of Biological Inorganic

Chemistry, 2015, 20(2), 265-275

4) C. Kubitza, F. Bittner, C.n Ginsel, A. Havemeyer, B. Clement, and A. J. Scheidig, Crystal structure of human mARC1

reveals its exceptional position among eukaryotic molybdenum enzymes. Proceedings of the National Academy of Sciences

of the United States of America, 2018, 115 (47) 11958-11963

5) C. Ginsel, B. Plitzko, D. Froriep, D. A. Stolfa, M. Jung, C. Kubitza, A. J. Scheidig, A. Havemeyer and B. Clement, The

Involvement of the Mitochondrial Amidoxime Reducing Component (mARC) in the Reductive Metabolism of Hydroxamic

Acids. Drug Metabolism and Disposition, 2018, 46(10), 1396-1402

6) S. Rixen, A. Havemeyer, A. Tyl-Bielicka, K. Pysniak, M. Gajewska, M. Kulecka, J. Ostrowski, M. Mikula, B. Clement,

Mitochondrial Amidoxime reducing Component 2 (mARC2) has a significant role in N-reductive activitiy and energy

metabolism. Journal of Biological Chemistry, 2019, 46: 17593-17602

7) P. Indorf, C. Kubitza, A.J. Scheidig, T. Kunze, B. Clement, Drug Metabolism by the Mitochondrial Amidoxime Reducing

Component (mARC2): Rapid Assay and Identification of New Substrates. Journal of Medicinal Chemistry, 2020, 63,

6538-6546

8) M.R. Lassen, Recent developments in the use of oral anticoagulants. Expert Opinion in Pharmacotherapy, 2009, 10,

1769-1781

9) M. Sanford & G. L. Plosker, Dabigatran etexilate. Drugs, 68, 1699-1709

10) J. Stangier, H. Stähle, K. Rathgen, R. Fuhr, Pharmacokinetics and pharmacodynamics oft he direct oral thrombin

inhibitor dabigatran in healthy elderly subjects. Clinical Pharmacokinetics, 2008, 57, 47-59

EFMC-ISMC | 129

LE086

NELADENOSON BIALANATE HYDROCHLORIDE – PRODRUG

EXPLORATION OF A PARTIAL ADENOSINE A1 AGONIST

Daniel Meibom

Bayer AG, Aprather Weg 18A, 42113 Wuppertal, Germany

Adenosine, a purine nucleoside, exerts a variety of physiological actions by binding to adenosine cell surface

receptor subtypes (A1, A2a, A2b, and A3) which are widely expressed in the body. A1 receptor activation by

full A1 agonists results in desired (e.g. cardioprotection) and non-desired (e.g. sedation) effects. Partial A1

receptor agonists can work as agonists with lower efficacy which might lead to organ-specific beneficial

pharmacological responses.

The presentation will begin by briefly touching upon the hit to candidate optimization, starting from an HTS hit

and leading to Neladenoson, focusing on the degree of partial agonism needed to arrive at a suitable therapeutic

window. Neladenoson, while safe and efficacious in animal models, showed low solubility in aqueous as well as

organic media prohibiting tablet development. Furthermore, only 1% bioavailability was observed after oral

administration of a suspension of crystalline material to rats. A dipeptide prodrug approach was used to improve

solubility and bioavailability.

Details on the prodrug campaign including the screening cascade used will be shown. Dipeptide ester prodrugs

were tested for their hydrolytic profile in aqueous media, their bioavailability from solution and crystallinity.

Neladenoson bialanate hydrochloride emerged as the compound with the best characteristics. The presentation

will also highlight the prodrug’s solubility and PK properties from crystalline material (e.g. 52% bioavailability).

Sufficient shelf life of the bis-alanate prodrug of Neladenoson allowed for tablet development and two PhIIb

studies.

130 | EFMC-ISMC

LE087

TARGETED (PRO)DRUGS FOR IMPROVED TREATMENT OF BRAIN

TUMORS

Kristiina M. Huttunen

University of Eastern Finland, School of Pharmacy

During the drug development, it has been very challenging to create potent compounds with perfect

pharmacokinetic properties. Therefore, for example, the majority of central nervous system (CNS) drugs fail in

clinical trials as they are not delivered to their site of actions, and thus, they lack efficacy and cause toxic side

effects. The blood-brain barrier (BBB) prevents the uptake of numerous drug molecules into the brain, however,

the cell membranes and the delivery to specific cell types, such as cancer cells, may also create a secondary

barrier for drugs. In this presentation, I will show how L-type amino acid transporter 1 (LAT1) can be utilized

for targeting brain tumors by introducing two examples; 1) LAT1-utilizing prodrugs of a chemotherapeutic and

2) LAT1-utilizing derivative of an efflux transporter inhibitor that can increase the accumulation of another

chemotherapeutic. The presentation will also cover the main challenges that can be faced when working with this

approach.

EFMC-ISMC | 131

LE088

NOVEL MOLECULAR TARGETS FOR THE TREATMENT OF

FIBROSIS

Jörg H.W. Distler

Department of Internal Medicine 3, University Hospital Erlangen, Germany

Persistent activation of fibroblasts is a common denominator of fibrotic diseases but mechanistically

incompletely defined. In contrast to physiologic tissue repair responses, fibroblasts remain persistently active in

fibrotic diseases and continue to release excessive amounts of extracellular matrix. While fibroblasts are only

transiently activated in normal wound healing, they are persistently active in fibrotic diseases. We will discuss

novel insights into the molecular mechanisms underlying the uncontrolled activation of fibroblasts in fibrotic

diseases and potential therapeutic implications for targeted antifibrotic therapies.

132 | EFMC-ISMC

LE089

DISCOVERY OF GLPG1205, A FIRST IN CLASS GPR84 ANTAGONIST

IN PHASE II CLINICAL TRIAL

Romain Gosmini

Galapagos, Chemistry102 AVENUE GASTON ROUSSEL - 93230 Romainville

France

Medium chain Free Fatty acid activated GPR84 receptor has been identified as playing an important role in

inflammation. In this presentation the identification of potent dihydropyrimidino-isoquinolinones GPR84

antagonists will be described. Setting up of the biological cascade and optimization of in vitro potency and

ADME/PK properties of the Hit series leading to the identification of GLPG1205 will be addressed. in vivo

studies in IBD animal model as well as pharmacological data supporting current progression of GLPG1205 in

phase II clinical trial in IPF will be presented. In addition studies establishing the particular binding mode of

GLPG1205 and its close analogues will be documented.

EFMC-ISMC | 133

LE090

INVESTIGATING THE CHAMELEONIC PROPERTIES OF RGD

INTEGRIN ANTAGONISTS FOR THE TREATMENT OF IDIOPATHIC

PULMONARY FIBROSIS

Dr James D.F. Thompson (1,2), Mr John M. Pritchard (1), Professor William J. Kerr (2)

1) GlaxoSmithKline, Gunnels Wood Road, Stevenage, Herts. SG1 2NY, United Kingdom2) Dept. of Pure and Applied Chemistry, Thomas Graham Building, 295 Cathedral Street, Glasgow. G1 1XL, United

Kingdom

 

 

This work describes an investigation into the unusual permeability of a candidate-quality RGD integrin

antagonist (1) for the treatment of Idiopathic Pulmonary Fibrosis, in particular its ability to show chameleonic

behaviour depending on the polarity of its environment, as shown in the figure above. The importance of

molecular chameleons is gaining appreciation, as new, more difficult-to-drug targets require molecules that fall

beyond the ‘rule of 5’, but still possess oral bioavailability. In this case, this behaviour allows for the compound

to transiently appear more lipophilic during permeation, then to reveal its polar functionality upon binding.

The zwitterionic nature of α

V

integrin inhibitors means that obtaining passive permeability is a challenge. During

development of 1, it was found to have enhanced permeability over its diastereomer. This enhanced permeability

was not predicted, and it was hypothesised that this enhancement may be a conformational effect, since the

compounds have identical measured lipophilicities and pK

a

s.

Several analogues have been synthesised to investigate how changing the flexibility of the core of the compound

affects the permeability. Additionally, the largely untapped resource of

15

N NMR has been used in a unique

fashion to show the presence of a greater extent of intramolecular hydrogen bonding in 1 than its diastereomer,

which is supported by computational investigations and physicochemical experiments. These methods have

subsequently been used to rationalise the permeability of other more recent series of RGD integrin antagonists.

Based on these findings, work is ongoing to show the use of this method predictively, to allow ranking of

possible future targets based on predicted permeability. Initial results of these predictions are very positive. This

will aid future medicinal chemistry work, especially for pharmacophores where designing passively permeable

compounds is a challenge, such as those beyond the ‘rule of 5’.

134 | EFMC-ISMC

LE091

DISCOVERY OF THE HIGHLY POTENT AND SELECTIVE S1P2

ANTAGONIST GLPG2938, A PRECLINICAL CANDIDATE FOR THE

TREATMENT OF IDIOPATHIC PULMONARY FIBROSIS

Aline Palisse (1), Oscar Mammoliti (1), Caroline Joannesse (1), Beatrice Coornaert (1), Kathleen Sonck

(1), Inge Duys (1), Thierry Christophe (1), Nicolas Houvenaghel (1), Bertrand Heckmann (2)

1) Galapagos NV, Generaal De Wittelaan L11 A3, 2800 Mechelen, Belgium2) Galapagos SASU, 102 avenue Gaston Roussel, 93230 Romainville, France

Despite several studies showing that blocking S1P2 receptor (S1PR2) signaling could be effective for the

treatment of idiopathic pulmonary fibrosis (IPF), only a few antagonists have been so far disclosed and there is

no report of clinical studies.

The presentation will describe the different strategies and optimization to obtain GLPG2938, a highly potent and

selective S1P2 antagonist, tested also in a bleomycin induced model of pulmonary fibrosis.

Our program started with a chemical enablement strategy to led us to the discovery of a 5-pyrazol-yl-pyridine

series with good antagonist activity. A scaffold hopping exercise delivered a pyrazine series with improved

lipophilic efficiency (LipE), to finally reach, after further optimization, GLPG2938.

EFMC-ISMC | 135

LE092

MECHANISTIC INSIGHTS OF A CDK9 INHIBITOR VIA

ORTHOGONAL PROTEOMICS METHODS

Paola Castaldi (1), J. Adam Hendricks (2), Nigel Beaton (3), Alexey Chernobrovkin (4), Eric Miele (5),

Ghaith M. Hamza (5), Piero Ricchiuto (6), Ronald C. Tomlinson (2), Tomas Friman (4), Cassandra

Borenstain (8), Bernard Barlaam (9), Sudhir Hande (10), Chris DeSavi (12), Rick Davies (14), Martin

Main (11), Joakim Hellner (4), Kristina Beeler (3), Yuehan Feng (3), Roland Bruderer (3), Lukas Reiter

(3), Daniel Martinez Molina (4)

1) LifeMine Therapeutics, 30 Acorn Park Drive, Cambridge, MA 02140 (USA)2) Civetta Therapeutics, 10 Wilson Road, Cambridge, MA 021403) Biognosys AG, Wagistrasse 21, 8952 Schlieren, Switzerland

4) Pelago Bioscience AB, Banvaktsvägen 20, 17148 Solna, Sweden5) Discovery Sciences, AstraZeneca, Boston, Massachusetts 02451, United States

6) Alexion Pharmaceuticals, 121 Seaport Blvd, Boston, MA 02210 (USA)7) Cedilla Therapeutics, 38 Sidney Street, Cambridge, MA 02139 (USA)

8) Biogen, 133 Boston Post Rd, Weston, MA 02493 (USA)9) Oncology R&D, AstraZeneca, Cambridge, CB4 0WG, United Kingdom

10) Oncology R&D, Boston, Massachusetts 02451, United States11) Molecular, Cellular and Biomedical Sciences, University of New Hampshire, Durham, NH 03824, USA

12) Kymera Therapeutics, 200 Arsenal Yards Blvd., Suite 230, Watertown, MA 0247213) Medicine Discovery Catapult, Alderley Park, Alderley Edge, Cheshire SK104TG, UK

14) Discovery Sciences, AstraZeneca, Cambridge, CB4 0WG, United Kingdom

Cyclin-dependent-kinases (CDKs) are members of the serine/threonine kinase family and are highly regulated by

cyclins, a family of regulatory subunits that bind to CDKs. CDK9 represents one of the most studied examples of

these transcriptional CDKs. CDK9 forms a heterodimeric complex with its regulatory subunit cyclins T1, T2 and

K to form the positive transcription elongation factor b (P-TEFb). This complex regulates transcription via the

phosphorylation of RNA polymerase II (RNAPolII) on Ser-2, facilitating promoter clearance and transcription

elongation and thus remains an attractive therapeutic target.

Herein, we have utilized classical affinity purification chemical proteomics, kinobeads assay, compressed

CEllular Thermal Shift Assay (CETSA®)-MS and Limited Proteolysis (LiP) to study the selectivity, target

engagement and downstream mechanistic insights of a CDK9 tool compound.

The above experiments highlight the value of quantitative mass spectrometry approaches to drug discovery,

specifically proteome wide target identification and selectivity profiling. The approaches utilized in this study

unanimously indicated that the CDK family of kinases are the main target of the compound of interest, with

CDK9, showing the highest target affinity with remarkable consistency across approaches.

We aim to provide guidance to the scientific community on the available chemical biology/proteomic tools to

study advanced lead molecules and to highlight pros and cons of each technology while describing our findings

in the context of the CDKs biology.

136 | EFMC-ISMC

LE093

PROTEOMES IN 3D

Paola Picotti

ETH Zürich, Zürich, Switzerland

Protein structural changes induced by external perturbations or internal cues can profoundly influence protein

activity and thus modulate cellular physiology. Mass spectrometry (MS)-based proteomic techniques are

routinely used to measure changes in protein abundance, post-translational modification and protein interactors,

but much less is known about protein structural changes. In my talk, I will present a structural proteomics

method that enables analysis of protein structural changes on a proteome-wide scale and directly in complex

biological extracts. The approach relies on the coupling of limited proteolysis (LiP) tools and MS. LiP-MS can

detect subtle alterations in secondary structure content, larger scale movements such as domain motions, and

more pronounced transitions such as the switch between folded and unfolded states. I will describe how we are

applying this approach to study the molecular bases of protein aggregation diseases and to the identification of

protein-small molecule interactions (e.g drug targets). I will also propose that monitoring protein structural states

on a proteome-wide scale can serve as a new powerful readout to pinpoint altered protein functional states and

the (de)regulation of biochemical pathways. Last, I will discuss the power and limitations of the new approach.

EFMC-ISMC | 137

LE094

A NOVEL BACTERIAL THREE-HYBRID SYSTEM FOR TARGET

IDENTIFICATION IN BACTERIA

Radhia El Phil (1,2), Sébastien Tardy (1,2), Aurélie Gouiller (1,2), Olivier Petermann (1,2), Oscar Vadas

(1,3),

1) Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland2) Pharmaceutical Biochemistry/Chemistry Group, School of Pharmaceutical Sciences, University of Geneva, Geneva,

Switzerland3) Department of microbiology and molecular medicine, Faculty of medicine, University of Geneva, Geneva, Switzerland

As part of the success of target hunting in drug discovery, the implementation of new tools and technologies is

essential to explore uncharted pathways and validate targets.

In this paper, we report a novel in vivo tool for small molecule-target identification, a bacterial three-hybrid

system. Unlike its yeast counterpart the handling should be easier and screens should be less time consuming (1).

The snap-tag labelling technique is used to anchor the molecule of interest to the transcription machinery

allowing an increased sensitivity of the system. The target proteins used for the profiling of drugs will be

obtained from ORF libraries introduced via the Gateway® technique. This will allow the easy, and quick cloning

of genes into the single vector of the system already containing the elements for the transcription machinery. The

use of a unique vector will contribute to the decrease of the number of experiment to perform. The latter exploits

the complementarity of two proteins T25 and T18 taken from an adenylate cyclase domain (2). When separated,

the proteins are unable to generate cyclic adenosine monophosphate (cAMP). However, the interaction of the

molecule of interest synthesized as an O

6

-benzylguanine (BG) derivative  with the target protein produced as a

fusion with T18 will allow the indirect interaction with T25 produced as a fusion with the Snap protein resulting

in the reconstitution of the adenylate cyclase activity in E. coli deficient in this activity.  The read-out of the

system exploits this reconstitution as the synthesized cAMP forms a complex with the catabolite activator

protein and triggers the lacZ gene encoding for the β-galactosidase. The expression of this enzyme and therefore

the occurrence of an interaction can be easily detected on MacConkey medium.

In order to develop the system, several elements had to be prepared. A chemical synthesis of the BG derivative

have been achieved by combining 3 elements: a BG moiety, a PEG linker and the molecule of interest. The

single plasmid carrying the transcription machinery had to be validated by testing plasmids constructed with the

fused proteins either at the C-terminus or N-terminus and the Trypanosoma brucei adenosine kinase gene has

been inserted to serve as a positive control. The expression of the proteins within the cells has been confirmed.

Finally, we could assess that the synthetized BG enter the bacterial cell and that the snap-tag labelling have been

successfully tested in E. coli.

References

1) Chidley C, Haruki H, Pedersen MG, Muller E, Johnsson K. A yeast-based screen reveals that sulfasalazine inhibits

tetrahydrobiopterin biosynthesis. Nat Chem Biol. 2011;7(6):375-83.

2) Battesti A, Bouveret E. The bacterial two-hybrid system based on adenylate cyclase reconstitution in Escherichia coli.

Methods. 2012;58(4):325-34.

138 | EFMC-ISMC

LE095

DESIGN AND SYNTHESIS OF CHEMICAL TOOLS TO PROBE THE

FUNCTION OF TRIM33

Jessica Reynolds (1), Stuart Conway (1), James Dodd (2)

1) Department of Chemistry, University of Oxford, United Kingdom2) Vertex Pharmaceuticals, Abingdon, United Kingdom

Ripartite motif-containing (TRIM) protein 33 is a transcriptional regulator that contains a C-terminal dual

PHD-bromodomain motif, which recognises methylation states and acetylation of lysine residues respectively.

TRIM33 functions as part of the PARP-dependent DNA damage response pathway. Developing ligands for the

TRIM33 bromodomain and exploring in-cell degradation will enable validation of the protein as a therapeutic

target in situations where DNA damage is increased. To achieve this aim, we are taking two approaches;

targeting the bromodomain using small molecule inhibition, and proteolysis-targeting chimera

(PROTAC)-induced degradation of TRIM33 in cells.

The group has previously identified compound 1 containing the benzimidazolone core as a ligand for the

TRIM33 bromodomain, and TRIM24. SAR studies guided by docking and molecular dynamics allowed a

greater understanding of the TRIM33 bromodomain binding pocket and led to development of ligand 3 and 4

(Figure 1). Having achieved selectivity over the structurally similar bromodomain of TRIM24,

heterobifunctional PROTAC molecules are being synthesised, which contain TRIM33 ligands linked to an E3

ubiquitin ligase warhead. These compounds could recruit TRIM33 to the E3 ligase VHL and allow degradation

in cells (Figure 2).  To date, a series of PROTACs with different linkers were synthesised, but cell permeability

was marginal. Despite this, a promising degradation profile was seen for one PROTAC, so a further series of

PROTACs is being synthesised with more desirable cell permeability. These parallel strategies for probing

TRIM33 will allow examination of the difference between inhibition and degradation, and subsequently explores

the role of TRIM33 and its bromodomain in disease.

References

1) Kulkarni et al., J. Biol. Chem., 2013, 288, 32357-32369.

EFMC-ISMC | 139

LE096

REPROGRAMMING NONRIBOSOMAL PEPTIDE BIOSYNTHESIS

Donald Hilvert

Laboratory of Organic Chemistry, ETH Zürich, 8093 Zurich, Switzerland

Nonribosomal peptides are a structurally diverse class of secondary metabolites that exhibit a broad range of

biological activities. These molecules are naturally produced by huge, multifunctional enzymes called

nonribosomal peptide synthetases (NRPSs). Although synthetic or semisynthetic modification of these molecules

can enhance their efficacy and even provide new therapeutic leads, the structural complexity of most

nonribosomal peptides makes systematic chemical manipulation challenging. Consequently, there is

considerable interest in harnessing biosynthetic routes for the discovery and sustainable production of new

chemotypes with enhanced pharmacological properties or novel bioactivities. In this lecture, an evolutionary

platform for engineering these assembly-line biocatalysts in high throughput will be discussed. It is based on the

display of libraries of functional NRPS modules on the surface of yeast cells and subsequent screening for

variants that perform a novel catalytic function. For example, this approach has been successfully used to

reprogram the substrate specificity of the gatekeeper adenylation domains responsible for building block

selection. The resulting modules retain their function in the context of a full-length NRPS cluster, enabling

robust assembly of natural product analogs.  Building blocks possessing diverse chemical and physical

properties, including bioorthogonal handles, backbone modifications, crosslinking groups, fluorophores, caging

moieties, spectroscopic probes and the like, can be incorporated site specifically into nonribosomal peptides in

this way, expanding the nonribosomal code and opening the door to customized peptide therapeutics.

140 | EFMC-ISMC

LE097

FLAVOENZYME BIOCATALYSIS IN BIOORGANIC AND

MEDICINAL CHEMISTRY – CHALLENGES AND OPPORTUNITIES

Marko D. Mihovilovic

TU Wien, Institute of Applied Synthetic Chemistry,Getreidemarkt 9/163, A-1060 Vienna, Austria; [email protected]

Over the last two decades flavoenzymes have attracted considerable attention in academic research mainly due to

the diverse nature of biotransformations, which can be achieved with these redox biocatalysts. Baeyer-Villiger

monooygenases (BVMOs) represent a particularly interesting class of biocatalysts based on their high substrate

promiscuity and excellent selectivity in combination with remarkable stereoelectronic behavior in generating

chiral building blocks. The identification of enantiocomplementary wild-type enzymes together with an

increasing understanding to engineer stereospecificity of these biocatalysts resulted in a number of facile

applications for the preparation of biologically active compounds. Several case-studies will be presented

employing the diverse biocatalytic behavior of BVMOs (kinetic resolution, desymmetrization, regiodivertent

biooxidation) and outlining the potential of this enzyme class in bioorganic synthesis.

As an important step towards broader scale application of BVMOs, only recently it was demonstrated that their

generally alleged instability under industrial process conditions can be overcome and their large-scale

application can be both economically and ecologically viable. Key challenges in improving these enzymes for

their operational stability within biotransformations will be outlined and recent progress in understanding the

intricate interplay between protein scaffold and essential cofactors affecting biocatalytic activity will be

discussed.

EFMC-ISMC | 141

LE098

UTILIZATION OF IRON/ALPHA-KETOGLUTARATE-DEPENDENT

ENZYMES FOR STEREOCONTROLLED C-H FUNCTIONALIZATION

Rebecca Buller

Zurich University of Applied Sciences, Institute for Chemistry and Biotechnology, Einsiedlerstrasse 31, 8820 Wädenswil,Switzerland

Modification of aliphatic C–H bonds in a regio- and stereoselective manner can pose a formidable challenge in

organic chemistry. In this context, Fe(II)/α-ketoglutarate-dependent dioxygenases, enzymes which are capable of

halogenating and hydroxylating sp

3

 carbons with high stereo- and regiocontrol under benign conditions, have

attracted particular attention. Using directed evolution, we engineered iron/α-ketoglutarate dependent

dioxygenases and halogenases for the functionalization of molecules of pharmaceutical interest, ranging from

non-natural amino acids

1

 to alkaloids

2

. Notably, our enzyme engineering approach allowed us to rapidly identify

more active enzyme variants increasing the apparent kcat and the turnover number of the enzymes by orders of

magnitude. In addition, we succeeded to modulate the catalyzed chemistry in the active site of a Fe(II)/α

-ketoglutarate-dependent dioxygenase, generating desaturated, hydroxylated or halogenated products in function

of the active site architecture. The biochemical characterization of the (novel) enzymes further highlights their

application potential.

References

1) F. Meyer*, R. Frey*, M. Ligibel, E. Sager, K. Schroer, R. Snajdrova, R. Buller, Modulating chemoselectivity in a Fe(II)/

α-ketoglutarate dependent dioxygenase for the oxidative modification of a non-proteinogenic amino acid, ACS Catal 2021,

11 (10), 6261-6269

2) T. Hayashi, M. Ligibel, E. Sager, M. Voss, J. Hunziker, K. Schroer, R. Snajdrova, R. Buller, Evolved aliphatic

halogenases enable regiocomplementary C-H functionalization of an added-value chemical, Angew Chem Int Ed 2019, 58,

18535 - 18539.

142 | EFMC-ISMC

LE099

INTEGRATION OF BIOCATALYSIS INTO MEDICINAL CHEMISTRY

PROGRAMS FOR LATE-STAGE OXIDISED DERIVATIVES USING

POLYCYPS ENZYMES

Frank Scheffler, Julia Shanu-Wilson, Liam Evans, Renia Gemmell, Emily Hopkins, Aksana Khan, Ravi

Manohar, Kinga Nytko, Richard Phipps, Vincent Poon, Jonathan Steele, Stephen Wrigley

Hypha Discovery Limited154B Brook Drive, Milton Park, Abingdon,

Oxfordshire, OX14 4SD, UK

Late-stage oxidation of C-H bonds in drug compounds permits the diversification of complex structures to alter

drug properties, without the need for de novo synthesis. The selective introduction of oxidised functionality,

acting as hydrogen bond donors and acceptors in three dimensional space, enables drug molecules to interact

with hitherto unexploited sites within a target protein. Thus, pharmacological properties such as potency and

selectivity can be influenced, as well as DMPK properties and physical attributes such as solubility and polarity.

Being challenging to achieve synthetically, C-H bond oxidation is common in nature in the metabolism of

xenobiotics and biotransformation via microbes, liver S9 fractions or recombinant enzymes offers an alternative

route to rapidly produce and analyse oxidised derivatives of drug candidates.

Hypha have developed a diverse set of cytochrome P450s – PolyCYPs® – recombinant enzymes cloned from

actinomycete bacteria and expressed in E.coli. PolyCYPs enzymes, which permit production of a broad range of

oxidised derivatives from multiple enzyme starting points, including the biosynthesis of human CYP-derived

metabolites. Due to the wide range of organic molecules that CYPs are able to oxidise, they can also be used for

late-stage oxidation of drug candidates to generate multiple oxidised derivatives in parallel, whilst

simultaneously exploring the possibility of active metabolites.

To expedite the process, series of lead compounds can be rapidly screened against a set of PolyCYPs enzymes to

determine their susceptibility to late-stage oxidation. Compounds, for which usable conversions to oxidised

products are observed, can then be prioritised for scale-up reactions to generate sufficient material for

pharmacological testing and subsequent structural identification of active derivatives.

The development of PolyCYPs as tools for late-stage oxidation of drug leads will be discussed and exemplified

using case studies.

EFMC-ISMC | 143

LE100

TARGETING CANCER

Daniel Rauh (1,2)

1) Department of Chemistry and Chemical Biology, TU Dortmund University, Otto-Hahn-Strasse 4a, 44227 Dortmund(Germany)

2) Drug Discovery Hub Dortmund (DDHD) am Zentrum für integrierte Wirkstoffforschung (ZIW), 44227 Dortmund(Germany)

eMail: [email protected], Twitter: @DDHDortmund, Web: https://www.rauh-lab.de

The treatment of genetically defined cancer is experiencing a revolution. Over the last 15 years, the knowledge

gained about the biochemical features of biomarkers and their predictive power has led to the development of

targeted small-molecule molecules that have improved the quality of life and increased the survival of cancer

patients. However, the occurrence of inevitable drug resistance limits these approaches and requires the constant

development of next-generation precision medicines

1

.

 

Against this background, we employ protein X-ray

crystallography

2

, structure-based design approaches

3

, organic synthesis

4

, as well as biochemical

5

and cellular

compound screening

6

 to understand the mechanisms of acquired resistance better and to develop next-generation

inhibitors. We are also interested in devising means to bridge the innovation gap between academic research and

practical application. We recently established the “Zentrum für integrierte Wirkstoffforschung” (ZIW) as well as

the Drug Discovery Hub Dortmund (DDHD) to serve as incubators for the translation of basic academic research

into pharmaceutical applications. Some of our current endeavors will be outlined during the talk.

Keywords: protein kinase, academic drug research, inhibitor, covalent, Ras

References

1) a) J. Lategahn, et al., Angew Chem Int Ed Engl, 2018, 57, 2307-13; b) S. Grunewald, et al., Cancer Discov., 2021, 11,

108-125.

2) a) J. Niggenaber, et al., J Med Chem, 2020, 63, 40-51., b) J. Niggenaber, et al., ACS Med Chem Lett.,2020, 11,

2484-2490.

3) a) J. Engel, et al., Angew Chem Int Ed Engl, 2016, 55, 10909-12; b) J. Lategahn, et al., Chem Sci, 2019, 10, 10789-01; c)

L. Quambusch, et al., Angew Chem Int Ed Engl, 2019, 58, 18823-29.

4) a) J. Weisner, et al., Angew Chem Int Ed Engl, 2015, 54, 10313-16; b) J. Weisner, et al., Cancer Res, 2019, 79, 2367-78;

c) N. Uhlenbrock, et al., Chem Sci, 2019, 10, 3573-85.

5) a) S. C. Mayer-Wrangowski, et al., Angew Chem Int Ed Engl, 2015, 54, 4379-82; b) R. Schneider, et al., J Am Chem Soc,

2013, 135, 6838-41; c) J. R. Simard, et al., Nat Chem Biol, 2009, 5, 394-6.

6) J. Fassunke, et al., Nat Commun, 2018, 9, 4655.

144 | EFMC-ISMC

LE101

STRUCTURE-BASED OPTIMIZATION AND SYNTHESIS OF M3258, A

POTENT AND SELECTIVE INHIBITOR OF THE

IMMUNOPROTEASOME SUBUNIT LMP7 (beta5i) DEMONSTRATING

STRONG EFFICACY IN MULTIPLE MYELOMA MODELS.

Markus Klein (1), Michael Busch (1), Manja Friese-Hamin (1), Stefano Crossignani (1), Thomas Fuchss

(1), Djordje Musil (1), Felix Rohdich (1), Michael Sanderson (1), Jeyaprakashnarayanan Seenisamy (2),

Gina Walter-Bausch (1), Ugo Zanelli (1), Philip Hewitt (1), Christina Esdar (1), Oliver Schadt (1)

1) Merck KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany2) Syngene International Limited, Biocon Park, Plot2&3, Bommasandra-Jigani Link Road, 560 099 Bangalore, India

Proteasomes are large, multi-subunit proteolytic complexes that are key components of the ubiquitin-dependent

protein degradation pathway. While the constitutive proteasome (cP) is broadly expressed, the

immunoproteasome (iP) is the predominant isoform in normal and malignant hematopoietic cells.

Both proteasome forms contain 3 catalytically active subunits (beta1, beta2 and beta5) with unique structural and

proteolytic features. The iP subunit LMP7 (beta5i) plays an essential role in restoring protein homeostasis under

conditions of elevated proteotoxic or oxidative stress; a hallmark of selected cancer cell types such as multiple

myeloma. Clinically effective pan-proteasome inhibitors (PIs) target LMP7 and other iP and cP subunits in a

comparable potency range. This lack of selectivity is associated with diverse toxicities and thus can limit the

therapeutic utility of these drugs. We hypothesized that selective LMP7 inhibition could potentially overcome

the limitations of pan-PIs. This presentation will describe our LMP7 drug discovery program including

structure-based hit optimization, SAR development, improvement of ADME properties, and synthesis of

new amido boronic acids which selectively inhibit LMP7 whilst sparing all other cP and iP subunits. The

exploitation of structural differences of the proteasome subunits led to the identification of the highly potent and

exquisitely selective LMP7 inhibitor cpd 50 (M3258). Despite the presence of a benzofuran and a bicyclic ether,

M3258 displayed favorable pharmacokinetics and oral bioavailability. M3258 demonstrated strong anti-tumor

activity in several multiple myeloma xenograft models in mice. These data supported the initiation of a phase I

clinical trial of M3258 in relapsed/refractory multiple myeloma patients.

References

1) Klein et al, J. Med. Chem. (submitted)

2) Klein, M.; Busch, M.; Esdar, C.; Friese-Hamim, M.; Krier, M.; Musil, D.; Rohdich, F.; Sanderson, M.; Walter, G.; Schadt,

O.; Zanelli, U.; Ma, J. Abstract LB-054: Discovery and profiling of M3258, a potent and selective LMP7 inhibitor

demonstrating high efficacy in multiple myeloma models. Cancer Research 2019, 79, LB-054-LB-054

3) Sanderson et al, Molecular Cancer Therapeutics, 2021, DOI: 10.1158/1535-7163.MCT-21-0005

EFMC-ISMC | 145

LE102

MAGIC IN THE MOONLIGHT: OUR CONTRIBUTION TO THE

DEVELOPMENT OF IDO1 INHIBITORS FOR CANCER

IMMUNOTHERAPY

Tracey Pirali (1), Maria Teresa Pallotta (2), Alberto Massarotti (1), Silvia Fallarini (1), Stefano Ugel (3),

Silvio Aprile (1)

1) Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy2) Department of Experimental Medicine, University of Perugia, Perugia, Italy

3) University Hospital and Department of Medicine, Section of Immunology, University of Verona, Verona, Italy

Immunotherapy has revolutionized the clinical practice in oncology, as exemplified by the advent of immune

checkpoint inhibitors in the treatment of melanoma. Nevertheless, the clinical benefit of cancer immunotherapy

might be limited by the establishment of an immunosuppressive microenvironment as cancer progresses.

From this perspective, one of the key proteins in promoting cancer immune escape is indoleamine

2,3-dioxygenase 1 (IDO1). In the effort to discover a therapeutic agent capable of boosting the anti-tumor

immune responses, hundreds of inhibitors that interfere with IDO1 biochemical activity (i.e. tryptophane

catabolism) have been developed over the years.

In 2018 one of them, epacadostat, did not outperform treatment

with pembrolizumab alone in a Phase 3 clinical trial, raising the hypothesis that some features of IDO1 biology

might have been overlooked. 

It has now become clear that IDO1 is a far more complex protein than first thought, shaping up to be a

moonlighting protein (i.e. a multitasking protein, that, besides its canonical function, displays unpredictable

secondary activities according to its localization in the cell and the interaction with binding partners).

Besides

its well-known biochemical activity, IDO1 acts as a signal-transducing molecule: it interacts with molecular

partners, resulting in the upregulation of its gene expression or in its degradation by the proteasomal system.

It is also able to self-regulate its catalytically active form (holo-IDO1), shifting toward a heme-free conformation

(apo-IDO1).

After years of focusing on inhibitors of the catalytic activity of IDO1,

4,5,6

 we have serendipitously discovered a

class of small molecules that bind to the apo-form, compete with the heme cofactor and are able to affect IDO1

signalling functions. In this communication, the starting chemotypes, the performed SAR study to improve the

drug-likeness and the biological data obtained so far will be disclosed.

The available information point to an unprecedented interplay between inhibition of apo-IDO1 and modulation

of its signalling functions, a peculiarity that might confer a unique competitive advantage over the IDO1

inhibitors discovered so far. 

References

1) Feng, X. et al. Development of indoleamine 2,3-dioxygenase 1 inhibitors for cancer therapy and beyond: a recent

perspective. J. Med. Chem. 2020, 63, 15115-15139

2) Adamo, A. et al. Moonlighting proteins are important players in cancer immunology. Front. Immunol. 2021, 11, 613069

3) Pallotta, M.T. et al. Indoleamine 2,3-dioxygenase is a signaling protein in long-term tolerance by dendritic cells. Nat.

Immunol. 2011, 12, 870-878

4) Fallarini, S. et al. In silico-driven multicomponent synthesis of 4,5- and 1,5-disubstituted imidazoles as indoleamine

2,3-dioxygenase inhibitors. Med. Chem. Comm. 2016, 7, 409

5) Griglio, A. et al. A multicomponent approach in the discovery of indoleamine 2, 3-dioxygenase 1 inhibitors: Synthesis,

biological investigation and docking studies. Bioorg. Med. Chem. Lett. 2018, 28, 651-657

6) Serafini, M. et al. Discovery of highly potent benzimidazole derivatives as indoleamine 2,3-dioxygenase-1 (IDO1)

inhibitors: from structure-based virtual screening to in vivo pharmacodynamic activity. J. Med. Chem. 2020, 63, 3047-3065

146 | EFMC-ISMC

LE103

DESIGN AND OPTIMIZATION OF A FIRST-IN-CLASS NACK

INHIBITOR: A NOVEL PATH TO NOTCH INHIBITION

Tanya Kelley (1), Dr. Giulia Diluvio (2), Ellen Kolb (3), Dr. Rhett Kovall (3), Dr. Anthony Capobianco (2),

Dr. Stephan Schürer (1)

1) Department of Molecular and Cellular Pharmacology, Sylvester Comprehensive Cancer Center Epigenetics Program,University of Miami Miller School of Medicine, Miami, FL, USA

2) Department of Surgery, Sylvester Comprehensive Cancer Center Tumor Biology Program, University of Miami MillerSchool of Medicine, Miami, FL, USA

3) Department of Molecular Genetics, University of Cincinnati College of medicine, Cincinnati, OH, USA

Notch Activation Complex Kinase (NACK) is a key player in Notch-mediated tumorigenesis and an attractive

novel target for the treatment of esophageal adenocarcinoma, and other Notch-rich cancers. NACK, annotated in

the kinome as SgK223, is an atypical pseudokinase and an established Notch transcriptional co-activator.

However, there is no known endogenous or exogenous ligand, no existing co-crystal structure and no reported

biological data. To identify a scaffold for NACK inhibition, a machine learning approach was utilized. Over 6

million commercially available compounds were screened against established kinase machine learning

classifiers, and nearly 8000 were prioritized based on the predicted probability of being active. A homology

structure model of the NACK kinase domain was generated and further optimized by all-atom explicit water

molecular dynamics (MD) simulations, followed by virtual screening of prioritized compounds. Top-scoring

compounds were purchased and screened in in-vitro and in-vivo assays. Commercially available compound

Z271-0326 displayed low micromolar inhibitory activity across several notch-rich cancer cell lines, and was

further validated in rodent models. A robust novel chemical synthesis for Z271-0326 was accomplished in six

steps with an overall yield of 26%. Current efforts are aimed towards optimizing Z271-0326 into the first NACK

molecular probe. We optimized our virtual NACK kinase domain structure model via MD simulations, and

performed a structure-guided optimization approach to better understand putative binding interactions.

Analogues were designed and synthesized with the goal of improving binding affinity and inhibitory activity.

Preliminary assay results demonstrate that our hit compound is optimizable as analogue UM-323 demonstrates

an IC

50

 of 45 nM in OE33 cells, and displays NACK target engagement via Differential Scanning Fluorimetry.

Recent efforts are focused towards improving lipophilicity and metabolic stability while attenuating affinity so

that we may arrive at an advanced preclinical NACK inhibitor. 

EFMC-ISMC | 147

A. Carbohydrate Recognition and Drug DesignPosters - Chemical Biology

148 | EFMC-ISMC

A001

NANOMUG: A NOVEL GLYCOSYLATED PROTEIN BASED

DELIVERY SYSTEM

Sonja Visentin (1), Cosmin Stefan Butnarasu (1), Giuseppe Guagliano (2), Francesco Bracotti (1), Livia

Visai (3), Paola Petrini (2)

1) Department of Molecular Biotechnology and Health Sciences, University of Torino, via Quarello 15, Italy2) Politecnico di Milano, Dipartimento Chimica, Materiali e Ingegneria Chimica “G. Natta”, Piazza L. da Vinci, 32, 20133

Milano3) Molecular Medicine Department, UdR INSTM, University of Pavia, Viale Taramelli 3/B, 27100 Pavia, Italy

NanoMuG stems from the willingness to ensure readiness for future outbreaks and reduce social impact of

frightening pathologies while improving quality of life. Our objective is to the reinvent drug delivery with a

transversal platform to lead the research towards a change of paradigm, thinking smaller and smarter.

We are developing the NanoMug system, a patented technology that could be considered a new class of

nanoparticles, defined as mucosomes, made of glycoproteins with mucoadhesive properties, specifically

advantageous for the delivery/interaction with mucus, although not limited to this. In Figure 1 some advantages

of mucosomes are described.

The protein structure makes the nanoparticles  biocompatible; studies have shown how NanoMuGs are able to

adhere to mucus and the presence of glycans, sugars involved in cell recognition (1,2) allows for a specific

direction of nanoparticles. The possibility, then, of being loaded with different drugs means that the NanoMuGs

are suitable for new drugs or for  the repositioning of drugs currently on the market. Finally, the perfect recipe

comes from the stability of the powder, from the lean, agile synthesis that makes NanoMuGs unique and from

the team interdisciplinarity.

References

1) Beat Ernst et al., Nature Reviews, Drug Discovery, 2009, 8. 661-677.

2) Daniel Passos da Silva et al., Nature Communications, 2019, 10, 1-11.

EFMC-ISMC | 149

A002

DESIGN, SYNTHESIS AND EVALUATION OF SIALIC ACID

DERIVATIVES THAT INHIBIT SIAT SYMPORTERS AND

ATTENUATE BACTERIAL GROWTH

Tiago Bozzola (1,2), Ulf J. Nilsson (1), Ulf Ellervik (1)

1) Centre for Analysis and Synthesis, Department of Chemistry, Lund University, Sweden2) Department of Pharmaceutical Sciences, University of Basel, Switzerland

According to the World Health Organization (WHO), antibiotic resistance is a serious threat to our society.

[1]

Due to the significance of sialic acid for bacterial growth and proliferation, the sialic acid uptake inhibition could

represent a potent and novel pathway to develop new antibacterial drugs.

[2]

We have designed, synthesized and evaluated a library of sialic acid derivatives to test the previous hypothesis.

Here we present the first compounds able to bind and inhibit the sialic acid uptake of the SiaT transporters from

P. mirabilis[3]

and S. aureus.[4]

The compounds were evaluated with nanoDSF as a screening method and ITC to

determine the affinity constant. The best compound showed an affinity constant in the mid nanomolar range,

with nearly 300-fold affinity increase compared to sialic acid. Molecular dynamics supported the analysis of

possible binding modes and in explaining the affinity enhancements. A proteoliposome assay confirmed that the

compounds competitively inhibit sialic acid transport via the SiaT transporters from P. mirabilis and S. aureus.

Bacterial growth assays revealed that the compounds possess bacteriostatic effects likely due to sialic acid

uptake inhibition. These findings confirm the validity of our approach and open the way for a potential new class

of antibacterial drugs.

References

1) WHO. Antimicrobial resistance. Global Report on Surveillance. Bull. World Health Organ. 2014, 61, 383–94.

2) Vimr, E. Unified theory of bacterial sialometabolism: how and why bacteria metabolize host sialic acids. ISRN Microbiol.,

2013, 816713.

3) Wahlgren, W. Y. et al. Substrate-bound outward-open structure of a Na+-coupled sialic acid symporter reveals a new Na+

site. Nat. Commun. 2018, 1–14.

4) North, R. A. et al. The Sodium Sialic Acid Symporter from Staphylococcus aureus has altered substrate specificity. Front.

Chem. (2018) 6, 1–11 .

150 | EFMC-ISMC

A003

HIGH-AFFINITY GLYCOMIMETIC LIGANDS FOR HUMAN

SIGLEC-8

Gabriele Conti (1,2), Blijke Kroezen (1), Jonathan Cramer (1), Benedetta Girardi (1), Said Rabbani (1),

Oliver Schwardt (1), Daniel Ricklin (1), Roland Pieters (2), Beat Ernst (1)

1) Molecular Pharmacy, Department of Pharmaceutical Sciences, University of Basel, 4056 Basel, Switzerland2) Chemical Biology and Drug Discovery, Department of Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht,

The Netherlands

We present here the identification of glycomimetics with low µM affinity towards the human Siglec-8. Siglec-8

is an immunoglobulin-type lectin solely expressed on eosinophils and mast cells, and weakly on basophils. Many

pathological conditions are associated with altered functions and/or numbers of these cells, among which allergic

inflammation and asthma

1

. Despite the only partially known biological mechanism of action, the

pharmacological importance of Siglec-8 has been demonstrated as eosinophil apoptosis and inhibition of mast

cell degranulation could be achieved by means of anti-Siglec-8 monoclonal antibodies or synthetic

glycopolymers decorated with Siglec-8 ligands

2

. However, no small molecules targeting Siglec-8 have been

described so far. Such molecules could be useful to better elucidate the apoptotic cellular pathway and

potentially provide a new pharmacological approach for eosinophil and mast cell associated diseases.

The glycan epitope recognized by Siglec-8 is the tetrasaccharide 6’-sulfo sialyl Lewis

x

(6’S-sLe

x

)

3

. While the

sialic acid carboxylate and the sulfate group on the galactose are involved in two crucial salt bridges, fucose and

glucosamine show minor contributions to binding. Therefore, not surprisingly, we discovered that the related

disaccharide Neu5Ac-Gal6S represents the minimal binding epitope (Fig. 1). This disaccharide served as lead

compound for our search of new ligands with improved affinity and drug-like properties. In addition, it has been

recently reported that sulfonamide modifications at the 9-position of the sialic acid moiety lead to compounds

with increased activity

4

.

Applying different strategies, such as Gal6S replacement with non-carbohydrate moieties, bioisostere

modifications, and extension of the glycerol side chain (Fig. 1), we synthesized a new series of glycomimetic

structures. The best representative exhibits a low µM affinity, i.e. an almost 20-fold improved affinity compared

to tetrasaccharide 6’S-sLe

x

. ITC measurements revealed that binding of 6’S-sLe

x

is punished with a substantial

entropic penalty, whereas the disaccharide mimetics exhibit beneficial entropic and enthalpic contributions.

Our study made available potent small-molecule Siglec-8 antagonists, which can be used to further explored the

biological role of Siglec-8.

Figure 1. Chemical structure of Neu5Ac-Gal6S and the various modifications exploited for the discovery of

high-affinity ligands.

References

1) H. Floyd, J. Ni, A. L. Cornish, Z. Zeng, D. Liu, K. C. Carter, J. Steel, P. R. Crocker; J. Biol. Chem. 2000, 275, 861-866.

2) a) S. A. Hudson, N. V. Bovin, R. L. Schnaar, P. R. Crocker, B. S. Bochner; J. Pharmacol. Exp. Ther. 2009, 330, 608-612;

b) B. A. Youngblood, E. C. Brock, J. Leung, R. Falahati, B. S. Bochner, H. S. Rasmussen, K. Peterson, C. Bebbington, N.

Tomasevic; JCI Insight 2019, 4, e126219.

3) J. M. Pröpster, F. Yang, S. Rabbani, B. Ernst, F. H.-T. Allain, M. Schubert; PNAS 2016, 113, E4170-E4179.

4) C. M. Nycholat, S. Duan, E. Knuplez, C. Worth, M. Elich, A. Yao, J. O’Sullivan, R. McBride, Y. Wei, S. M. Fernandes, Z.

Zhu, R. L. Schnaar, B. S. Bochner, J. C. Paulson; J. Am. Chem. Soc. 2019, 141, 14032-14037.

EFMC-ISMC | 151

A004

A GLYCOMIMETIC STRATEGY AGAINST DC-SIGN-MEDIATED

CELL ENTRY AND DISSEMINATION OF SARS-COV-2

Jonathan Cramer (1,2), Adem Lakkaichi (1), Butrint Aliu (1), Roman P. Jakob (3), Sebastian Klein (1),

Ivan Cattaneo (1), Xiaohua Jiang (1), Said Rabbani (1), Oliver Schwardt (1), Timm Maier (3), Beat Ernst

(1)

1) University of Basel, Department of Pharmaceutical Sciences, Institute of Molecular Pharmacy, Klingelbergstrasse 50,4056 Basel, Switzerland

2) Heinrich-Heine-University of Düsseldorf, Department of Pharmaceutical Sciences, Institute of Pharmaceutical andMedicinal Chemistry, Universitätsstraße 1, 40225 Düsseldorf, Germany

3) University of Basel, Biozentrum, Institute of Structural Biology, Klingelbergstrasse 70, 4056 Basel, Switzerland

The C-type lectin receptor DC-SIGN is a pattern recognition receptor expressed on macrophages and dendritic

cells. It has been identified as a promiscuous entry receptor for many pathogenic agents, including pandemic

viruses such as SARS-CoV-2, ebola, and HIV.

1-5

In the context of the recent SARS-CoV-2 pandemic, DC-SIGN-mediated stimulation of innate immune

responses and virus dissemination through the lymphatic system have been implicated as potential factors in the

development of severe COVID-19.

2-5

Inhibition of virus binding to DC-SIGN, thus, represents an attractive

host-directed strategy to attenuate the progression of the disease and prevent overshooting innate immune

responses.

In this study, we report the discovery of a new class of potent glycomimetic DC-SIGN antagonists from a

focused library of triazole-based mannose analogs. Structure-based optimization of an initial screening hit

yielded a glycomimetic ligand with an over 100-fold improved binding affinity compared with methyl

mannoside. Analysis of binding thermodynamics revealed an enthalpy-driven improvement of binding affinity

that was enabled by hydrophobic interactions with an adjacent loop region and displacement of a conserved

water molecule. The identified ligand was employed for the synthesis of a multivalent glycopolymer that was

able to inhibit SARS-CoV-2 spike glycoprotein binding to cells expressing DC-SIGN at nanomolar

concentrations. Furthermore, the glycompolymer efficiently abrogated DC-SIGN-mediated trans-infection of

susceptible Vero E6 cells by SARS-CoV-2 pseudotyped VSV viruses.

The identified glycomimetic ligands reported here open promising perspectives for the development of highly

potent and fully selective DC-SIGN-targeted therapeutics, not only for severe COVID-19, but also for a broad

spectrum of other viral infections with pandemic potential.

 

References

1) Monteiro, J.; Lepenies, B. Myeloid C-Type Lectin Receptors in Viral Recognition and Antiviral Immunity. Viruses 2017,

9 (3), 59.

2) Gao, C.; Zeng, J.; Jia, N. et al. SARS-CoV-2 Spike Protein Interacts with Multiple Innate Immune Receptors. bioRxiv

2020, 2020.07.29.227462.

3) Chiodo, F.; Bruijns, S. C. M.; Rodriguez, E. et al. Novel ACE2-Independent Carbohydrate-Binding of SARS-CoV-2 Spike

Protein to Host Lectins and Lung Microbiota. Biorxiv 2020, 2020.05.13.092478.

4) Thepaut, M.; Luczkowiak, J.; Vives, C. et al. DC/L-SIGN Recognition of Spike Glycoprotein Promotes SARS-CoV-2

Trans-Infection and Can Be Inhibited by a Glycomimetic Antagonist. Biorxiv 2020, 2020.08.09.242917.

5) Katz, D. H.; Tahir, U. A.; Ngo, D.; et al. Proteomic Profiling in Biracial Cohorts Implicates DC-SIGN as a Mediator of

Genetic Risk in COVID-19. Medrxiv 2020, 2020.06.09.20125690.

152 | EFMC-ISMC

A005

MULTIVALENT FUCOSYLATED INHIBITORS TARGETING

BETA-PROPELLER LECTINS AS PROMISING ANTI-ADHESIVE

DRUGS

Margherita Duca (1,2), Annabelle Varrot (1), Roland J. Pieters (2)

1) Université Grenoble Alpes, CNRS, CERMAV, Grenoble, France2) Utrecht University, UIPS, Chemical Biology and Drug Discovery Department, Utrecht, The Netherlands

Bacterial and fungal pathogens often use lectins to mediate adhesion to glycoconjugates at the surface of host

tissues, especially those involved in lung infections of immunocompromised patients. Fucose-binding lectins

presenting a six bladed β-propeller fold, such as BambL[1] from Bulkholderia ambifaria, AFL1/FleA[2] from

Aspergillus fumigatus and SapL1[3] from Scedosporium apiospermum, are an example. Given the rapid

emergence of resistance to the treatments on current use, these lectins have become appealing targets for

alternative therapies that interfere with the host-pathogens recognition process, such as the development of

antiadhesive glycodrugs.

Multivalency[4] (i.e.: the presence of multiple binding sites) plays a major role in such events, allowing

simultaneous low affinity interactions between carbohydrate ligands and their receptors, leading to avidity and

stronger affinity. To block the adhesion of the pathogen to host tissues, we synthesized a set of inhibitors for the

targeted lectins that can display up to six fucosides units. They feature a mercaptomethyl-benzene core, a PEG

spacer and a sugar ligand: key structural elements that allow to optimize valency, size and flexibility of the final

molecule. The new antagonists showed nanomolar affinity for their protein targets, as measured by diverse

biophysical methods: fluorescence polarisation, isothermal titration calorimetry, and bio-layer interferometry.

References

1) A. Audfray, J. Claudinon, S. Abounit, N. Ruvoёn-Clouet, G. Larson, D. F. Smith, M. Wimmerová, J. Le Pendu, W.

Rёmer, A. Varrot, and A. Imberty, Fucose-binding Lectin from Opportunistic Pathogen Burkholderia ambifaria Binds to

Both Plant and Human Oligosaccharidic Epitopes, The Journal of Biological Chemistry, 2012, 287, 6, 4335–4347.

2) J. Houser, J. Komarek, N. Kostlanova, G. Cioci, A. Varrot, S. C. Kerr, M. Lahmann, V. Balloy, J. V. Fahy, M. Chignard,

A. Imberty, M. Wimmerová, A Soluble Fucose-Specific Lectin from Aspergillus fumigatus Conidia - Structure, Specificity

and Possible Role in Fungal Pathogenicity, PLOS ONE, 2013, 8, 12, e83077.

3) D. Martínez-Alarcón, J-P. Bouchara, R. J. Pieters, A. Varrot, SapL1: A New Target Lectin for the Development of

Antiadhesive Therapy Against Scedosporium Apiospermum, bioRxiv, 2020, https://doi.org/10.1101/2020.11.10.376251.

4) R. J. Pieters, Maximising multivalency effects in protein–carbohydrate interactions, Org. Biomol. Chem., 2009, 7,

2013–2025.

EFMC-ISMC | 153

A006

ORGANORUTHENIUM(II) INHIBITORS OF HUMAN HEXOKINASE 2

AS HEPATOCELLULAR ANTICANCER AGENTS

Joana Gonçalves (1), Joana Amaral (1), Cecília Rodrigues (1), Rui Moreira (1), Vanessa Almeida (2),

Margarida Frazão (2), Sara Garcia (3), Matilde Marques (3), Nissim Hay (4), Paulo Costa (5), Pedro

Florindo (1)

1) Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia da Universidade de Lisboa, Av. Prof.Gama Pinto, 1649-003 Lisboa (Portugal)

2) Instituto de Tecnologia Química e Biológica António Xavier (ITQB-NOVA), Av. da República, 2780-157 Oeiras(Portugal)

3) Centro de Química Estrutural, Instituto Superior Técnico (CQE-IST), Av. Rovisco Pais 1, 1049-001, Lisboa (Portugal)4) University of Illinois at Chicago, Department of Biochemistry and Molecular Genetics, 900 S. Ashland (M/C 669)

Chicago, IL 60607 (USA)5) Biosystems and Integrated Sciences Institute (BioSys), Faculdade de Ciências da Universidade de Lisboa, Campo Grande,

1749-016 Lisboa (Portugal)

Hexokinases (HK) catalyse the first committed step in glycolysis, converting glucose to glucose-6-phosphate

(G6P). There are four HK isoforms (HK1-4) that share high sequence homology but significantly differ in their

kinetics, subcellular distribution, and regulation suited to their specific metabolic functions. HK2 is

overexpressed in most cancer cells to comply with increased anabolic demands required for proliferation. 

Hepatocellular Cancer (HCC) displays the most comprehensive glycolytic reprogramming: while normal

hepatocytes rely on HK4 (glucokinase, GCK) for glucose phosphorylation, HCC cells express HK2 in high

levels, repressing GCK expression. Since systemically delivered drugs tend to accumulate first in the liver,

relatively low doses of HK2 inhibitors can selectively target HCC cells over normal hepatocytes, profiling it as

an excellent target for HCC chemotherapy [1].

As molecular shape is one of the most important factors in molecular recognition by biomolecules, metal

complexes are an ideal starting point to tackle a novel chemical space in drug design. With a well-established

synthetic chemistry, ruthenium(II) compounds provide access to a huge diversity of kinetically-inert

three-dimensional scaffolds [2].  Our team very recently identified potent HK2 hit inhibitors within a series of

ruthenium(II) glycoconjugates. Indeed, best compound demonstrated low µM potency in HK2 inhibition,

inhibition of glycolysis along with a strong antiproliferative effect (IC

50

) against HCC cells (Figure 1), and to

selectively inhibits glycolysis in HK2 expressing CHO cells, versus HK1- and GCK-expressing cells.

We will disclose on biochemical characterization of hit inhibitors, efforts for HK2-hit co-crystallization, and

chemical design towards hit-to-lead optimization of HK2 inhibitory potency.

 

Figure 1. Left: preliminary docking results of lead inhibitor in HK2 (PDB: 5HG1). Right: Glycolysis Stress

Test. Hepatocellular cancer cells (Huh7) incubated with vehicle (D) or hit inhibitor (6) for 2h (10 µM).

Compound strongly inhibits glycolysis in HCC cells.

 

References

1) D. DeWaal et al., Hexokinase-2 depletion inhibits glycolysis and induces oxidative phosphorylation in hepatocellular

carcinoma and sensitizes to metformin. Nat. Commun. 9 (2018) 446

2) G. Gasser, N. Metzler-Nolte, The potential of organometallic complexes in medicinal chemistry. Curr. Opin. Chem. Biol.

16 (2012) 84–91

154 | EFMC-ISMC

A007

1,3-SUBSTITUTED GALACTOSIDES AS SELECTIVE MONOVALENT

GALECTIN-8 LIGANDS

Benedetta Maria Girardi (1,2), Martina Manna (2), Ulf Nilsson (3), Tihomir Tomasic (2), Marko

Anderluh (2), Janez Mravljak (2), Daniel Ricklin (1), Oliver Schwardt (1)

1) Molecular Pharmacy Group, Pharmacenter, University of Basel, CH-4056 Basel, Switzerland2) Faculty of Pharmacy, University of Ljubljana, 1000 Ljubljana, Slovenia

3) Centre for Analysis and Synthesis, Department of Chemistry, Lund University, Box 124, Lund, Sweden

Galectin-8 (Gal-8) is a tandem-repeat type lectin widely expressed in tissues, both in normal and cancer cells. It

mainly regulates cell-cell and cell-matrix interaction, playing a role in many physiological and pathological

processes.

1,2

Regulating the differentiation of T cells, plasma cells and bone cells, Gal-8 has a crucial function in

inflammation and bone remodeling.

3,4

It also helps immune system in recognizing and eliminating bacteria by

activating selective autophagy process.

5

Overexpression of Gal-8 has been linked to different pathologies, like

inflammatory disorders, autoimmune diseases and cancer: in particular the protein stimulates lymphangiogenesis

and the adhesion and dissemination of tumor cells.

6,7

Thus, Gal-8 is a new interesting pharmacological target for

the treatment of many diseases.

Gal-8 has two carbohydrate binding domains (CRDs), one at the N-terminal and the other at the C-terminal, with

different binding specificities. Just recently, the crystal structure of Gal-8N with its preferred ligand, the

3-SiaLac, has been solved. By investigating the main interactions between this ligand and the protein, the

minimal binding epitope was identified and crystallized: it consists of a free galactose bearing a lactic acid in

position 3.

8

Starting from the methyl-3-O-(2-lactyl)galactopiranoside core structure, in this work we aimed to improve its

affinity and the selectivity for Gal-8 by modifying the lactic acid moiety in position 3 and by introducing a

3,4-dichlorophenyl ring in position 1 by S-glycosidic bond, a group known to enhance the binding to galectins

(Figure). Affinity data measured by Fluorescence Polarization show that we have obtained a focused library of

galactosides with the most potent compound reaching affinity for Gal-8 with a K

D

of 12 μM. Reasonable

selectivity versus other galectins was achieved making the highlighted compound a valuable molecular probe.

Our results represent an optimal starting point for the development of novel selective and potent ligands for

Gal-8 as molecular probes to study the protein’s role in cell lines and in vivo.

References

1) Tazhitdinova, R.; Timoshenko, A. V. Cells 2020, 9 (8), 1792.

2) Zick, Y.; Eisenstein, M.; Goren, R. A.; Hadari, Y. R.; Levy, Y.; Ronen, D. Glycoconj. J. 2002, 19 (7–9), 517–526.

3) Tribulatti, M. V.; Carabelli, J.; Prato, C. A.; Campetella, O. Glycobiology 2020, 30 (3), 134–142.

4) Vinik, Y.; Shatz-Azoulay, H.; Vivanti, A, Hever N.; Levy, Y.; Karmona, R.; Brumfeld, V.; Baraghithy, S.; Attar-Lamdar,

M.; Boura-Halfon, S.; Bab, I.; Zick, Y. Elife. 2015, 4: e05914.

5) Thurston, T.; Wandel, M., von Muhlinen, N.; Foeglein, A.; Randow, F. Nature 2012, 482, 414–418.

6) Chen, W. S.; Cao, Z.; Sugaya, S.; Lopez, M. J.; Sendra, V. G.; Laver, N.; Leffler, H.; Nilsson, U. J.; Fu, J.; Song, J.; Xia,

L.; Hamrah, P.; Panjwani, N. Nat. Commun. 2016, 7, 11302.

7) Shatz-Azoulay, H.; Vinik, Y.; Isaac, R.; Kohler, U.; Lev, S.; Zick, Y. Sci. Rep. 2020, 10 (1), 7375.

8) Bohari, M. H.; Yu, X.; Kishor, C.; Patel, B.; Go, R. M.; Eslampanah Seyedi, HA.; Vinik, Y.; Grice, I. D.; Zick, Y.;

Blanchard, H. ChemMedChem. 2018, 13(16), 1664-1672.

EFMC-ISMC | 155

A008

DESIGN, SYNTHESIS AND EVALUATION OF D-GALACTAL

DERIVATIVES AS SELECTIVE INHIBITORS OF GALECTIN-8

N-TERMINAL DOMAIN

Mujtaba Hassan (1,2), Floriane Baussière (1), Samo Guzelj (2), Anders Sundin (1), Maria Håkansson (3),

Björn Walse (3), Carl Diehl (3), Hakon Leffler (4), Fredrik Zetterberg (5), Tihomir Tomašič (2), Marko

Anderluh (2), Žiga Jakopin (2), Ulf J. Nilsson (1)

1) Centre for Analysis and Synthesis, Department of Chemistry, Lund University, Box 124, SE-221 00 Lund, Sweden2) University of Ljubljana, Department of Medicinal Chemistry, Faculty of Pharmacy, Aškerčeva cesta 7, 1000 Ljubljana,

Slovenia3) SARomics Biostructures AB, Medicon Village, SE-223 63 Lund, Sweden

4) Department of Laboratory Medicine, Section MIG, Lund University BMC-C1228b, Klinikgatan 28, 221 84 Lund, Sweden5) Galecto Biotech AB, Sahlgrenska Science Park, Medicinaregatan 8 A, SE-413 46 Gothenburg, Sweden

Galectins are a family of proteins that bind β-D-galactopyranoside-containing glycoproteins through their

carbohydrate recognition domains (CRD) to form lattices.

1

These lattices modulate the cellular mechanisms and

the immune system.

2,3

Galectin-8 is a member of the galectins family with two CRDs in tandem joined by a

peptide linker. It plays a key role in autophagy,

4

modulation of the innate and adaptive immunity,

5,6

and

pathological lymphangiogenesis, which is implicated in tumor growth and metastasis, and inflammatory

conditions.

7

Thus galectin-8 represents a promising target for the discovery of antiinflammatory and antitumor

drugs.

We have designed and synthesized a library of C-3 substituted D-galactal derivatives. Evaluation of the

compounds as inhibitors of the human galectin-1, 2, 3, 4N (N-terminal domain), 4C (C-terminal domain), 7, 8N,

8C, 9N, and 9C  by a competitive fluorescence polarization assay has led to the discovery of a

D-galactal-benzimidazole hybrid that represents the most selective galectin-8N inhibitor to date 15 folds

selectivity over galectin-3. X-ray crystallography followed by molecular dynamics simulation and quantum

mechanical calculations has revealed that the molecular orbitals interaction between the LUMO of Arg45 and the

HOMO of the olefine of as well as the HOMO of O4 of the D-galactal are responsible for the high affinity and

selectivity compared to the galactose derivatives. Treating the triple-negative breast cancer cells MDA-MB-231

cells with the D-galactal-benzimidazole compound reduced the secretion of the proinflammatory cytokines IL-6

and IL-8 in a dose-dependent manner. Evaluation of the cytotoxicity of the compound on a panel of cell lines in

MTS assay has shown that this compound does not affect the cell viability at the tested concentrations.

Therefore, the D-galactal-benzimidazole hybrid represents a safe and promising lead compound for the design

and synthesis of potent and selective galectin-8 inhibitors as potential antitumor and anti-inflammatory agents.

References

1) Lajoie, P.; Goetz, J. G.; Dennis, J. W.; Nabi, I. R. J. Cell Biol. 2009, 185 (3), 381 LP – 385.

2) Boscher, C.; Dennis, J. W.; Nabi, I. R. Curr. Opin. Cell Biol. 2011, 23 (4), 383–392.

3) Liu, F.-T.; Rabinovich, G. A. Nat. Rev. Cancer 2005, 5, 29–41.

4) Thurston, T. L. M.; Wandel, M. P.; Von Muhlinen, N.; Foeglein, Á.; Randow, F. Nature 2012, 482 (7385), 414–418.

5) Norambuena, A.; Metz, C.; Vicuña, L.; Silva, A.; Pardo, E.; Oyanadel, C.; Massardo, L.; González, A.; Soza, A. J. Biol.

Chem. 2009, 284 (19), 12670–12679.

6) Sampson, J. F.; Suryawanshi, A.; Chen, W. S.; Rabinovich, G. A.; Panjwani, N. Immunol. Cell Biol. 2016, 94 (2),

213–219.

7) Chen, W.-S.; Cao, Z.; Sugaya, S.; Lopez, M. J.; Sendra, V. G.; Laver, N.; Leffler, H.; Nilsson, U. J.; Fu, J.; Song, J.; Xia,

L.; Hamrah, P.; Panjwani, N. Nat. Commun. 2016, 7, 11302.

156 | EFMC-ISMC

A009

DIFFERENT DRUG DESIGN APPROACHES TO TACKLE O-GLCNAC

TRANSFERASE INHIBITION

Elena M. Loi (1,2), Matjaž Weiss (1), Cyril Balsollier (1,2), Tihomir Tomašič (1), Roland J. Pieters (2),

Marko Anderluh (1)

1) Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ljubljana, Ljubljana, 1000, Slovenia2) Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University,

3584 CG Utrecht, The Netherlands

O-GlcNAc transferase (OGT) is an essential enzyme that catalyzes the transfer of N-acetylglucosamine

(GlcNAc) onto Serine and Threonine protein residues. This dynamic post-translational modification occurs on

hundreds of cellular targets, and its impairment is linked with severe human pathologies such as Diabetes and

Cancer. Unfortunately, the role of OGT in these pathogenic processes is still not completely understood. Hence,

selective and cell-permeable OGT inhibitors are strongly needed to further study the O-GlcNAc cycle and

validate OGT as a therapeutic target.

This communication will cover our recent efforts in exploring the chemical space of OGT inhibitors by

combining different drug design approaches.

Structure-based virtual screening of fragment-like and drug-like libraries led to the identification of novel uridine

mimetic scaffolds that target the enzyme's active site. As an alternative strategy, the presence of new, potentially

druggable pockets in the protein structure was investigated.

The molecular modelling results served as the basis for the rational design and synthesis of new OGT inhibitors

that exhibited IC

50

values in the micromolar range.

EFMC-ISMC | 157

A010

RECOMBINANT LECTIN FROM TEPARY BEAN (Phaseolus acutifolius)WITH SPECIFIC CYTOTOXIC EFFECT OF COLON CANCER CELLS:

PRODUCTION, STRUCTURAL CHARACTERIZATION AND TARGET

IDENTIFICATION.

Dania Martínez-Alarcón (1,2,3,4), Annabelle Varrot (1), José L. Dena-Beltán (2), Elaine Fitches (4), John

A. Gatehouse (4), Min Cao (4), Prashant Pyati (4), Alejandro Blanco-Labra (3), Teresa García-Gasca (2)

1) University of Grenoble Alpes, CNRS, CERMAV, 38000 Grenoble, France2) Faculty of Natural Sciences, Autonomous University of Querétaro. Santiago de Querétaro, 76230. Querétaro, Mexico

3) Biotechnology and Biochemistry Department, CINVESTAV, Irapuato 36821, Guanajuato, México4) Department of Biosciences, Durham University, Durham, DH1 3LE, UK

A Tepary bean (Phaseolus acutifolius) lectin fraction (TBLF) recognizes aberrant glycoconjugates and induces

apoptosis on colorectal cancer cells

2

. In vivo studies have shown that TBLF toxicity is very low and that its

intragastric administration inhibits up to 70% of early tumorigenesis in rats with chemically-induced colorectal

cancer

3

. Therefore, it is considered as a potential therapeutic agent for malignant transformations of the

gastrointestinal tract. Nevertheless, its implementation as drug has not been carried out because the purification

process is expensive, slow and provide low yields.

Here, we report the large-scale production of a recombinant Tepary bean lectin (rTBL-1), its structural

characterization, identification of its ligand on cancer cell surfaces and a mapping the intracellular signaling

pathway induced by this lectin. rTBL-1 was expressed in Pichia pastoris yielding to 316 mg per liter of culture.

Its cytotoxic activity was assayed on colon cancer cells (HT-29), where a strong apoptotic effect was identified

after 24 h of treatment (LC50; 1.2 µg·mL

-1

). Its 3D-structure was solved by X-ray diffraction at 1.9 Å resolution

and a screening for potential targets, performed by glycan array, reveals that rTBL-1 binds to β1-6 branched

N-glycans, whose presence is increased in cancer cells due the upregulation of the enzyme MGAT5. The

presence of α1-6 core fucose, also tumor-associated, improved carbohydrate recognition. 

rTBL-1 mechanism of action was linked to the phosphorylation and eventual degradation of the Epidermal Grow

Factor Receptor (EGFR), which was induced through recognition of its glycosylated portion by rTBL-1. EGFR

degradation inhibits activation of Akt and triggers a caspase-dependent apoptosis by activation of p38, PARP,

HA2X and p-53. Furthermore, internalization of EGFR disrupts lattice formation through galectin binding and

decreases the prolonged transduction of signals that enhances cancer progression. rTBL-1 affinity for a broad

spectrum of mono and disaccharides was evaluated by isothermal titration calorimetry (ITC), however, no

interaction was detected, corroborating that carbohydrate recognition is highly specific and requires large ligands

for binding. This would explain for the differential recognition between healthy and cancer cells by Tepary bean

lectins. Our results indicate that the strategy followed provides an efficient platform for rTBL-1 production and

open the gates for its possible implementation as coadjuvant therapy of colorectal cancer treatments.

References

1) Martínez-Alarcón, D.; Varrot, A.; Fitches, E.; et al. Recombinant Lectin from Tepary Bean (Phaseolus acutifolius) with

Specific Recognition for Cancer-Associated Glycans: Production, Structural Characterization and Target Identification.

Biomolecules 2020, 10(4), 654;

2) Moreno, U.; López, F.J.; Cervantes, R.; et al. Tepary bean (Phaseolus acutifolius) lectins induce apoptosis and cell arrest

in G0/G1 by p53(ser46) phosphorylation in colon cancer cells. Molecules 2020, 25

3) Moreno, U.; López, F.J.; Cervantes, R.; et al. Phaseolus acutifolius lectin fractions exhibit apoptotic effects on colon

cancer: preclinical studies using dimethilhydrazine or azoxi-methane as cancer induction agents. Molecules 2017, 22, 1670.

158 | EFMC-ISMC

A011

SYNTHESIS OF LACTOSE-DRUG CONJUGATES FOR

LIVER-TARGETED DELIVERY

Morgan Morris, Paul Evans

Centre for Synthesis and Chemical Biology,School of Chemistry and Chemical Biology,

University College Dublin,Belfield, Dublin 4, Ireland.

Asialoglycoprotein receptor (ASGP-R) is a transmembrane C-type lectin expressed exclusively on hepatocytes

that recycles degraded glycans.

1

It recognizes, and internalizes, glycans which have cleaved sialic acid residues

and exposed sugar residues containing a cis-3,4-diol subunit. Furthermore, binding is enhanced through

multivalency since the lectin comprises three distinct subunits that may each bind independently to a saccharide

residue. ASGP-R has thus long been recognized as a promising vector for targeted drug delivery to the liver.

This may be achieved through conjugation of a suitable sugar to liver chemotherapeutics via an appropriate

linker system.

2

Such glycoconjugates have been used in the development of hepatocellular carcinoma and

malaria therapeutics.

3

Lactose- a waste product of the Irish dairy industry- represents a cheap alternative to

traditionally employed ligands in these delivery systems and may prove just as effective in ASGP-R mediated

uptake of drug treatments.

Herein, we describe the rational design and synthesis of a series of PEGylated lactose-drug conjugates with

suitable spatial geometries and solubility profiles to facilitate endocytosis by ASGP-R. We report the

development of glycoconjugates of both the anticancer drug camptothecin and of the fluorescent probe

fluorescein, with reference made to improved pharmacokinetics in the case of the former. A series of

monoantennary and triantennary systems have been employed as carriers and conjugated to these structures

through “click” chemistry protocols.

4

References

1) A. A. D'Souza, P. V. Devarajan, Journ. Contr. Rel. 2015, 203, 126.

2) G. Y. Wu, C. H. Wu, J. Biol. Chem. 1988, 263, 588.

3) L. Fiume, G. D. Stefano, C. Busi, A. Mattioli, F. Bonino, M. Torrani-Cerenzia, G. Verme, M. Rapicetta, M. Bertini, G.B.

Gervasi, Journ. Vir. Hep. 1997, 4, 363.

4) R. A. Petrov, S. Y. Maklakova, Y. A. Ivanenkov, Bioorg. Med. Chem. Lett. 2018, 28, 382.

EFMC-ISMC | 159

A012

TARGETING TLR4 WITH SYNTHETIC MOLECULES: INNOVATIVE

THERAPEUTICS FOR INFECTIOUS AND INFLAMMATORY

DISEASES

Francesco Peri, Alessio Romerio, Andrea Luraghi, Nicole Gotri, Monsoor Shaik, Ana Rita Franco, Fabio

A. Facchini

Department of Biotechnology and Biosciences, University of Milano-Bicocca.Piazza della Scienza, 2; 20126 Milano, Italy

Toll-Like Receptor 4 (TLR4) is one of the receptors of innate immunity, it is activated by Pathogen- and

Damage-Associated Molecular Patterns (PAMPs and DAMPs). Mild TLR4 stimulation by non-toxic molecules

resembling its natural agonist (lipid A), provided MPLA, a clinically approved vaccine adjuvant. TLR4

excessive activation by Gram-negative bacteria lipopolysaccharide (LPS) leads to sepsis, while TLR4

stimulation by DAMPs is a common mechanism in several inflammatory and autoimmune diseases. TLR4

inhibition by small molecules and antibodies could therefore provide access to innovative therapeutics targeting

sepsis, acute and chronic inflammations.[1] TLR4 antagonists can also block the violent inflammation and

cytokine storm caused by several viral diseases, including COVID-19.

The most recent achievement of our group in the development of synthetic, glycolipid-based TLR4 agonists and

antagonists will be presented. Monosaccharide-based TLR4 agonists potently stimulate innate immunity and are

in preclinical development as adjuvants in antiviral and antibacterial vaccines. TLR4 antagonist[2] efficiently

block inflammation in cell models and in animal models of sepsis, influenza virus lethality,[3] vascular

inflammations,[4] neuroinflammations,[5] and inflammatory bowel diseases (IBDs).[6]

Computer-assisted docking of synthetic monosaccharides active as TLR4 modulators to the MD-2/TLR4 dimer

 

References

1) A. Romerio, F. Peri (2020), Front. Immunol. (11), 1210.

2) F. Facchini et al. (2018), J. Med. Chem. (61), 2895–2909.

3) L. Perrin-Cocon et al., (2017), Sci. Rep. (7), 40791.

4) C. Huggins et al. (2015), Atherosclerosis (242), 563–570.

5) De Paola, M. et al. (2016), Pharmacol. Res. (103), 180–187.

6) Facchini, F. A. et al. (2020), Eur. J. Clin. Pharmacol. (76), 409–418.

160 | EFMC-ISMC

A013

DESIGN, SYNTHESIS AND EVALUATION OF

IMMUNOSTIMULATING ACTIVITIES OF DESMURAMYL PEPTIDES

CONTAINING 2-AMINOADAMANTANE-2-CARBOXYLIC ACID

Rosana Ribić (1), Marija Paurević (2), Ranko Stojković (3), Lidija Milković (3), Mariastefania Antica (3),

Srđanka Tomić (4)

1) University Center Varaždin, University North, Jurja Križanića 31b, HR-42000 Varaždin, Croatia2) Josip Juraj Strossmayer University Osijek, Ulica cara Hadrijana 8/A, HR-31000 Osijek, Croatia

3) Ruđer Bošković Institute, Bijenička cesta 54, HR-10000 Zagreb, Croatia4) Faculty of Science, University of Zagreb, Horvatovac 102a, HR-10000 Zagreb, Croatia

Muramyl dipeptide (MDP, N-acetylmuramyl-L-alanyl-D-isoglutamine) is the smallest peptidoglycan fragment

capable of replacing the whole Mycobacteria in Freund’sadjuvant. MDP acts as a pathogen-associated molecular

pattern and activates the NOD2 receptor. Numerous structural variations of MDP have been performed in order

to improve its pharmacological properties. The main parameter for the improvement is lipophilicity. We have

designed and prepared desmuramyl peptides containing lypophilic 2-aminoadamantane-2-carboxylic acid, and

furthermore modified it with mannose moieties. Mannose receptors presented on immunocompetent cells are

considered to be pattern-recognition receptors, as well as NOD2, and therefore they can affect the immune

reactions.

 Here we present the synthesis and biological evaluation of desmuramyl peptides containing

2-aminoadamantane-2-carboxylic acid, and its mono- and di-mannosylated derivatives. Their

immunostimulating activities were evaluated in vivo in the mouse model using ovalbumin as an antigen. The

effects on the secondary humoral response were determined by measuring overall anti-OVA IgG antibodies, and

the subclasses anti-OVA IgG1 and anti-OVA IgG2a. Mannosylated desmuramyl peptides exhibited an improved

immunological activity and the di-mannoslyated derivative showed to be the most active one.

EFMC-ISMC | 161

A014

MOLECULAR RECOGNITION OF GLYCOSAMINOGLYCANS BY

VASCULAR ENDOTHELIAL GROWTH FACTOR AND THEIR DUAL

ROLE ON ANGIOGENIC PROCESSES

Gloria Ruiz-Gómez (1), Linda Koehler (2), Kanagasabai Balamurugan (1), Sandra Rother (2), Joanna

Freyse (3), Stephanie Möller (4), Matthias Schnabelrauch (4), Sebastian Köhling (3), Snezana Djordjevic

(5), Dieter Scharnweber (2), Jörg Rademann (3), Vera Hintze (2), M. Teresa Pisabarro (1)

1) Structural Bioinformatics, BIOTEC TU Dresden, Tatzberg 47-51, 01307 Dresden, Germany2) Institute of Materials Science, Max Bergmann Center of Biomaterials, TU Dresden, Budapester Straße 27, 01069

Dresden, Germany3) Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Straße 2+4, 14195 Berlin, Germany

4) Biomaterials Department, INNOVENT e.V., Prüssingstraße 27 B, 07745 Jena, Germany5) Institute of Structural and Molecular Biology, University College London, Gower Street, Darwin Building, WC1E 6BT

London, United Kingdom

The aging of population has led to an increase in the number of multimorbid patients in which several pathologic

conditions such as psoriasis, osteoporosis, age-related blindness, cardiovascular disease or stroke are related to

angiogenesis [1,2]. Therefore, modulation of biomacromolecules related to angiogenesis such as vascular

endothelial growth factor (VEGF) has relevant applications in regenerative medicine and tissue engineering. The

development of innovative biomaterials containing glycosaminoglycans (GAG) is emerging as strategy to

modulate the biological function of different targets such as growth factors. In these lines, GAG have been

previously found to interact with VEGF [3].

We apply molecular modeling and dynamics (MD) simulations in combination with experimental techniques (

i.e. surface plasmon resonance and isothermal titration calorimetry (ITC)) to investigate the role of GAG in

angiogenesis through the detailed analysis of the molecular recognition of chemically modified sulfated GAG by

the heparin-binding domain (HBD) of VEGF

165

and its signaling receptor VEGFR-2. In addition, the biological

consequences of GAG recognition by VEGF

165

have been evaluated in 2D and 3D in vitro cell experiments

using umbilical vein endothelial cells (HUVECs).

Our MD-based theoretical models have predicted that sulfated GAG act as a “molecular staple” bridging the two

HBD of the dimeric VEGF

165

. Such HBD-GAG-HBD stacking configuration has been validated by ITC

experiments. Furthermore, our experimental and computational studies have revealed that the sulfation degree

and pattern in GAG play a relevant role on their strength of binding towards VEGF

165

. Moreover, GAG

recognition by VEGF

165

has been found to impair the interaction of the growth factor with its receptor

VEGFR-2, preventing downstream signaling and reducing HUVEC spheroid sprouting. Interestingly, sulfated

GAG promoted sprouting of HUVEC spheroids in the absence of growth factor [4]. The dual pro- and

anti-angiogenic function exhibited by sulfated GAG could be translated into innovative biomaterials able to

modulate angiogenesis and, therefore, healing processes by tuning their composition and selected GAG type.

References

1) Carmeliet, P. Nat. Med. 2003, 9, 653-660.

2) Martino, M. M. et al. Front. Bioeng. Biotechnol. 2015, 3, 45.

3) Zhao, W. et al. Biosci. Rep. 2012, 32, 71-81.

4) Koehler, L.; Ruiz-Gómez, G. et al. Sci. Rep. 2019, 9:18143.

162 | EFMC-ISMC

A015

HOW PHTHALAZINONE-DERIVATIVES BIND THE GALECTIN-8N

CARBOHYDRATE RECOGNITION DOMAIN WITH EXCELLENT

SELECTIVITY

Sjors van Klaveren (1,2), Mujtaba Hassan (1,2), Anders Sundin (2), Marko Anderluh (1), Tihomir

Tomašič (1), Ulf J. Nilsson (2)

1) Chair of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ljubljana, Slovenia2) Centre for Analysis and Synthesis, Department of Chemsitry, Faculty of Science, Lund University, Sweden

Galectins are sugar-binding protein with specificity for β-galactoside sugars. Galectin-8 has immunomodulatory

properties and is involved in tumour development and metastasis. A combination of protein–protein interactions

and carbohydrate recognition is involved in the function of galectin-8. To delineate the role of galectin-8

sugar-binding, selective ligands of the galectin-8 N-terminal carbohydrate recognition domain (CRD) are

essential tools.

We present a series of highly selective galectin-8N ligands with a phthalazinone scaffold. We will explain the

selectivity amongst these highly conserved CRDs through our molecular dynamics studies. The tools for

studying the biological function of galectin-8 carbohydrate recognition are now at hand. This series may also

feed the drug discovery pipeline and inspire those looking for selectivity within the galectin family.

EFMC-ISMC | 163

A016

CARBOHYDRATE PROBES FOR BACTERIAL LABELING

Boris Vauzeilles

Université Paris-Saclay, CNRS, Institut de Chimie des Substances Naturelles, UPR 2301, 91198, Gif-sur-Yvette, France.E-mail: [email protected]

Infectious disease represent a serious and actual societal challenge. 

In the pre-antibiotic era, bacterial infections could have serious consequences, and some epidemic outbreaks

often proved dramatic. During the 20

th 

century, the discovery of these molecules considerably impacted our life

conditions. Some bacteria remain however difficult to treat or to detect, and the development of resistant strains,

combined with their rapid diffusion within our globalized societies, have considerably reduced our antibiotic

arsenal. Epidemic outbreaks can regularly have severe sanitary, but also economic impact. Rapid detection and

identification of bacteria remains therefore a major subject of interest.

We are developing an approach to address this question, relying on metabolic labeling of the bacterial cell

surface. As an example, the external membrane of Gram-negative bacteria is covered by a dense

lipopolysaccharide layer (LPS) which is involved in cell integrity, but also in the virulence of some strains. 

We have shown that, when metabolically active, Gram-negative bacteria, can specifically incorporate a

chemically modified, azide-containing monosaccharide within their LPS. This bioorthogonal reporter group can

then be used to "reveal" labeled bacteria, using a click-chemistry ligation method. This strategy allows for rapid

detection of live pathogenic bacteria such as Legionella pneumophila. 

We have also recently developed a bioorthogonal reporter for the labeling of the mycomembrane

of Corynebacteria, which are close relatives of pathogenic Mycobacteria. We will present our most recent results

in this field.

References

1) A. Dumont, A. Malleron, M. Awwad, S. Dukan, B. Vauzeilles Angew. Chem. Int. Ed. 2012, 51, 3143-3146.

2) J. Mas Pons, A. Dumont, G. Sautejeau, E. Fugier, A. Baron, S. Dukan, B. Vauzeilles Angew. Chem. Int. Ed. 2014, 53,

1275-1278.

3) E. Fugier, A. Dumont, A. Malleron, E. Poquet, J. Mas Pons, A. Baron, B. Vauzeilles, S. Dukan, PLoS ONE 2015, 10(6):

e0127700.

4) C. Cabriel, N. Bourg, P. Jouchet, G. Dupuis, C. Leterrier, A. Baron, M.-A. Badet-Denisot, B. Vauzeilles, E. Fort, S.

Lévêque-Fort, Nature Commun., 2019, 10:1980 | https://doi.org/10.1038/s41467-019-09901-8

5) E. Lesur, A. Baron, C. Dietrich, M. Buchotte, G. Doisneau, D. Urban, J.-M. Beau, N. Bayan, B. Vauzeilles, D.

Guianvarc’h, Y. Bourdreux, Chem Commun., 2019, DOI: 10.1039/C9CC05754D

164 | EFMC-ISMC

NOTES

EFMC-ISMC | 165

B. Chemical Approaches to Stem Cell Differentiation

Posters - Chemical Biology

166 | EFMC-ISMC

B001

PHORBOL DIESTERS INDUCE TGFα RELEASE AND ADULT MOUSE

NEUROGENESIS

Escobar-Montaño Felipe (1), Ezzanad Abdellah (1), Gómez-Oliva Ricardo (2), Díez-Salguero Mónica (2),

Geribaldi-Doldan Noelia (2), Dominguez-Garcia Samuel (2), Botubol-Ares José Manuel (1), Durán-Patrón

Rosa (1), Nunez-Abades Pedro (3), Macías-Sánchez Antonio José (1), Castro González Carmen (2),

Hernández-Galán Rosario (1)

1) Departamento de Química Orgánica, Facultad de Ciencias, Universidad de Cádiz, Puerto Real, Cádiz, Spain and Institutode Investigación en Biomoléculas (INBIO), e-mail: [email protected]

2) Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain and Instituto de Investigación e InnovaciónBiomédica de Cádiz (INiBICA).

3) Departamento de Fisiología, Facultad de Farmacia, Universidad de Sevilla, 41012 Sevilla, Spain

Brain damage caused by neurological and vascular disorders or by traumatic injuries is tightly associated with an

irreversible neuronal loss, which may lead to cognitive impairment, motor dysfunction, and even alterations of

the personality. Neuronal replacement in damaged brain regions rarely occurs, complicating functional recovery.

Thus, this field demands the discovery of new drugs that regenerate damaged brain regions, facilitating

functional recovery.

[1]

In this study, a small library of phorbol 12,13-diester bearing low lipophilicity ester chains was prepared from

commercial Croton oil as potential neurogenic agents in the adult brain. Phorbol-20-trityl ether (P20T) was

obtained from phorbols esters present in Croton oil through a modified procedure described by Bertolini et al.

[2]

Once the hydroxyl group at C-20 was protected, a set of phorbol esters was prepared by selective

transformations at C-12, C-13 (Scheme 1).

The ability of these compounds to stimulate transforming growth factor alpha (TFGα) release, to increase neural

progenitor cell (NPC) proliferation, and to stimulate neurogenesis was evaluated. All compounds that facilitated

TGFα release promoted NPC proliferation when evaluated in vitro. The presence of two acyloxy moieties on the

tigliane skeleton led to higher levels of activity, which decreased when a free hydroxyl group was at C-12.

Remarkably, the compound bearing isobutyryloxy groups was the most potent on the TGFα assay and in the invitro NPC proliferation assay, also leading to enhanced neurogenesis in vivo when administered intranasally to

mice. These results point out at phorbol diesters as relatively simple-to-manufacture pharmacological agents in

the development of treatments for disorders associated with a reduction in neurogenesis and memory impairment

such as neurodegenerative diseases.

[3]

References

1) Schachar, R. J.; Park, L. S.; Dennis, M. Can. Acad. Child Adolesc. Psychiatr. 2015, 24, 100−108

2) Bertolini, T. M.; Giorgione, J.; Harvey, D. F.; Newton, A. C. J. Org. Chem. 2003, 68, 5028–5036

3) Ezzanad, A.; Ricardo Gómez-Oliva, R.; Escobar-Montaño, F.; Díez-Salguero, M.; Geribaldi-Doldán, N.;

Domínguez-García, S.; Botubol-Ares, J. M.; de los Reyes Jiménez, C.; Durán Patrón, R. M.; Nuñez-Abades, P.;

Macías-Sánchez, A. J.; Castro-González, C.; Hernández-Galán, R. J. Med. Chem. 2021, 64, 6070-6084.

EFMC-ISMC | 167

B002

UHPLC-HRMS-GUIDED DISCOVERY AND CHARACTERIZATION

OF NEW 12-DEOXY-16-HYDROXYPHORBOL-13,16-DIESTERS FROM

EUPHORBIA RESINIFERA: SELECTIVE PURIFICATION OF

NEUROGENIC AGENTS

Abdellah Ezzanad (1), Ricardo Gómez-Oliva (2,3), Felipe Escobar-Montaño (1), Mónica Díez-Salguero

(2), Noelia Geribaldi-Doldan (4), Samuel Dominguez-Garcia (2,3), José Manuel Botubol-Ares (1,3),

Carolina de los Reyes (1), Rosa Durán-Patrón (1,3), Pedro Nunez-Abades (5), Antonio José

Macías-Sánchez (1,3,6), Carmen Castro (2,3), Rosario Hernández-Galán (1,3,6)

1) Departamento de Química Organica, Facultad de Ciencias, Universidad de Cadiz, 11510 Puerto Real, Cádiz (Spain)2) Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, 11002, Cádiz (Spain)

3) Instituto de Investigación e Innovación Biomédica de Cádiz (INIBICA), 11009, Cádiz (Spain)4) Departamento de Anatomía y Embriología Humanas, Universidad de Cádiz, 11002 Cádiz, (Spain)5) Departamento de Fisiología, Facultad de Farmacia, Universidad de Sevilla, 41012 Sevilla (Spain)

6) Instituto de Investigacion en Biomoleculas (INBIO), Universidad de Cadiz, 11510 Puerto Real, Cádiz (Spain)

Phorbol derivatives such as 12-deoxyphorbol-13-acetate (Prostratin), phorbol 12-myristate-13-acetate (PMA),

12-deoxyphorbol 13-phenylacetate (DPP), 12-deoxyphorbol 13-isobutyrate (DPB) and 12-deoxyphorbol

13-angelate (DPA), are capable of induction of adult neural progenitor cell (NPC) proliferation via PKC

activation and of stimulating neurogenesis in the adult brain neurogenic niche of the subventricular zone (SVZ)

and dentate gyrus (DG).

[1]

,

[2]

Unlike PMA, Prostratine, DPP, DPB and DPA do not show tumour promoting

activities, suggesting 12-deoxyphorbols 13-acyl esters as good candidates to produce potent pharmacological

drugs that promote neurogenesis and facilitate functional repair of damaged brain tissue. Interestingly, the nature

of C-13 acyl chains is a key element to define the potency of these compounds in the activation of PKC and

promotion of neurogenesis in vivo.

[1]

,

[3]

Consequently, the influence of both the nature and location of these acyl

moieties on the activity of the resulting derivatives deserves further exploration.

This communication describes the detection, isolation and characterization of 12-deoxy-16-hydroxyphorbol

13,16-diesters, and the evaluation of their activity as neurogenesis-stimulating small molecules, through a

ultra-high performance liquid chromatography / high-resolution mass spectroscopy (UHPLC-HRMS) assisted

screening of the latex of E. resinifera. Resulting structure−activity relationships may provide helpful insights for

the development of this class of compounds as effective neurogenic agents.

References

[1]) Geribaldi-Doldán, N.; Flores-Giubi, E.; Muriilo-Carretero, M.; García-Bernal, F.; Carrasco, M.; Macías-Sánchez, A.J.;

Domínguez-Riscart, J.; Verástegui, C.; Hernández-Galán, R.; Castro, C.. Int.J. Neuropsychopharmacol. 2016, 19, 1-14,

doi:10.1093/ijnp/pyv085

[2]) Domínguez-García, S.; Gómez-Oliva, R.;Geribaldi-Doldán, N.; Hierro-Bujalance, C.; Sendra, M.; Ruiz, F.A.; Carrascal,

L.; Macías-Sánchez, A.J.; Verástegu, C.; Hernández-Galán, R.; García-Alloza, M.; Nunez-Abades, P.; Castro, C.

Neuropsychopharmacology. 2021, 46, 1207–1219, doi:10.1038/s41386-020-00934-y

[3]) Ezzanad, A., Ricardo Gómez-Oliva, R., Escobar-Montaño, F., Díez-Salguero, M., Geribaldi-Doldán, N.,

Domínguez-García, S., Botubol-Ares, J. M., de los Reyes, C., Durán Patrón, R. M., Nuñez-Abades, P., Macías-Sánchez, A.

J., Castro-González, C., Hernández-Galán, R. J. Med. Chem. 2021, 64, 6070-6084. doi.org/10.1021/acs.jmedchem.1c00156

168 | EFMC-ISMC

NOTES

EFMC-ISMC | 169

Posters - Chemical Biology C. Chemical Probes for Target Discovery and Validation

170 | EFMC-ISMC

C001

NOVEL ACTIVITY‐BASED PROBES AND NANOMOLAR

PEPTIDOMIMETIC INHIBITORS AGAINST THE MAIN PROTEASE

OF SARS‐CoV‐2

Marta Barniol-Xicota (1), Roeland Vanhoutte (1), Merel van de Plassche (1), Steven Verhelst (1,2)

1) Laboratory of Chemical Biology, Department of Cellular and Molecular Medicine.KU Leuven – University of Leuven.Herestraat 49 box 802, 3000 Leuven (Belgium)

2) AG Chemical Proteomics, Leibniz Institute for Analytical Sciences ISAS, Otto-Hahn-Str. 6b, 44227 Dortmund (Germany)

The global pandemic caused by SARS‐CoV‐2 calls for the fast development of antiviral drugs against this

particular coronavirus. Chemical tools to facilitate inhibitor discovery as well as detection of target engagement

by hit or lead compounds from high‐throughput screens are therefore in urgent need. We here report novel,

selective activity‐based probes that enable detection of the SARS‐CoV‐2 main protease. The probes are based on

acyloxymethyl ketone reactive electrophiles combined with a peptide sequence including unnatural amino acids

that targets the nonprimed site of the main protease substrate-binding cleft. They are the first activity‐based

probes for the main protease of coronaviruses and display target labeling within a human proteome without

background

1

. We expect that these reagents will be useful in the drug‐development pipeline, not only for the

current SARS‐CoV‐2, but also for other coronaviruses. Proof of this is our probes have enabled the discovery of

a novel class of peptidomimetic inhibitors, which display nanomolar potencies against the SARS‐CoV‐2 main

protease. Here we report the structures and antiviral activity of our new class of inhibitors and dwell into their

binding mode thanks to the co-crystal structure of our most potent inhibitor with Mpro

2

.

References

1) van de Plassche MAT* Barniol-Xicota M* Verhelst SHL. Peptidyl Acyloxymethyl Ketones as Activity-Based Probes for

the Main Protease of SARS-CoV-2. Chembiochem. 2020;10.1002/cbic.202000371. doi:10.1002/cbic.202000371. *Shared

first-authorship. Journal Cover

2) Unpublished results

EFMC-ISMC | 171

C002

BENZOTHIAZOLYLUREA-BASED 17β-HSD10 INHIBITORS -

DESIGN, SYNTHESIS, IN VITRO AND IN VIVO EVALUATION

Ondrej Benek (1,2), Monika Schmidt (1,2), Laura Aitken (3), Michaela Vaskova (1), Adam Skarka (1),

Jana Hatlapatkova (2,4), Lucie Zemanova (1), Jana Zdarova Karasova (2,4), Frank Gunn-Moore (3),

Kamil Musilek (1,2)

1) University of Hradec Kralove, Faculty of Science, Department of Chemistry, Hradec Kralove, Czech Republic;*[email protected]

2) University Hospital, Biomedical Research Center, Sokolska 581, 500 05 Hradec Kralove, Czech Republic3) University of St. Andrews, School of Biology, Medical and Biological Sciences Building, North Haugh, St. Andrews,

United Kingdom4) University of Defence, Faculty of Military Health Sciences, Department of Toxicology and Military Pharmacy, Hradec

Kralove, Czech Republic

There is a well-documented connection between Alzheimer’s disease (AD) and mitochondrial dysfunction. One

of mitochondrial enzymes affected in AD is 17β-hydroxysteroid dehydrogenase type 10 (17β-HSD10), also

known as amyloid-β binding alcohol dehydrogenase (ABAD). Importantly, it has been shown that inhibition of

this enzyme is beneficial in AD and also protects against amyloid-β toxicity

1–3

. We have developed several

series of benzothiazolyl urea compounds in order to find novel 17β-HSD10 inhibitors applicable for pre-clinical

studies

4,5

.

Compounds were prepared and screened in an enzymatic assay to determine their inhibitory ability towards

purified 17β-HSD10. For active compounds were determined IC

50

values and inhibition kinetics. Selected

compounds were tested in the cellular assay using fluorescent substrate CHANA. Two most promising

compounds were selected for bioavailability study when they were administered to mice or rats by i.v. injection

or p.o. gavage and compared to experimental inhibitor AG18051.

More than 100 novel compounds were designed and synthesized. All compounds were evaluated for 17β-HSD10

inhibitory ability in vitro, where several compounds showed promising inhibitory activity in both enzymatic (IC

50

< 2 µM) and cellular (EC

50

< 5 µM) assays.  The novel compounds were found to be uncompetitive inhibitors

of 17β-HSD10 with selectivity towards the enzyme-substrate complex. The experimental inhibitor AG18051, as

well as the two novel leads, showed, however, only minimal CNS exposure after p.o. administration to rats.

References

1) Muirhead, K. E. A.; Borger, E.; Aitken, L.; Conway, S. J.; Gunn-Moore, F. J. The Consequences of Mitochondrial

Amyloid β-Peptide in Alzheimer’s Disease. Biochemical Journal 2010, 426 (3), 255–270.

https://doi.org/10.1042/BJ20091941.

2) Benek, O.; Aitken, L.; Hroch, L.; Kuca, K.; Gunn-Moore, F.; Musilek, K. A Direct Interaction Between Mitochondrial

Proteins and Amyloid-β Peptide and Its Significance for the Progression and Treatment of Alzheimer's Disease. Current

Medicinal Chemistry 2015, 22 (9), 1056–1085. https://doi.org/10.2174/0929867322666150114163051.

3) Vinklarova, L.; Schmidt, M.; Benek, O.; Kuca, K.; Gunn‐Moore, F.; Musilek, K. Friend or Enemy? Review of

17β-HSD10 and Its Role in Human Health or Disease. Journal of Neurochemistry 2020, 155 (3), 231–249.

https://doi.org/10.1111/jnc.15027.

4) Aitken, L.; Benek, O.; McKelvie, B. E.; Hughes, R. E.; Hroch, L.; Schmidt, M.; Major, L. L.; Vinklarova, L.; Kuca, K.;

Smith, T. K.; Musilek, K.; Gunn-Moore, F. J. Novel Benzothiazole-Based Ureas as 17β-HSD10 Inhibitors, A Potential

Alzheimer’s Disease Treatment. Molecules 2019, 24 (15), 2757. https://doi.org/10.3390/molecules24152757.

5) Schmidt, M.; Benek, O.; Vinklarova, L.; Hrabinova, M.; Zemanova, L.; Chribek, M.; Kralova, V.; Hroch, L.; Dolezal, R.;

Lycka, A.; Prchal, L.; Jun, D.; Aitken, L.; Gunn-Moore, F.; Kuca, K.; Musilek, K. Benzothiazolyl Ureas Are Low

Micromolar and Uncompetitive Inhibitors of 17β-HSD10 with Implications to Alzheimer’s Disease Treatment. International

Journal of Molecular Sciences 2020, 21 (6), 2059. https://doi.org/10.3390/ijms21062059.

6) This work was supported by the Ministry of Education, Youth and Sports of the Czech Republic (project ESF no.

CZ.02.1.01/0.0/0.0/18_069/0010054), University of Hradec Kralove (Faculty of Science, no. VT2019-2021, SV2113-2019,

and Postdoctoral job positions at UHK), Wellcome Trust (204821/Z/16/Z), The Rosetrees Trust, and the RS MacDonald

Charitable Trust.

172 | EFMC-ISMC

C003

4-IODOPYRIMIDINE IS A POTENT AND SELECTIVE COVALENT

INHIBITOR AND ACTIVITY-BASED PROBE FOR MIF FAMILY

PROTEINS

Zhangping Xiao, Fabian Mulder, Deng Chen, Frank Dekker

Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen,Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands

Electron-deficient halogenated (hetero)aryls are a group of reactive fragments that have been employed to

covalently and unspecifically label cysteine on proteins via a nucleophilic aromatic substitution.

4-Iodo-6-phenylpyrimidine (4-IPP) is a covalent inhibitor for both macrophage migration inhibitory factor (MIF)

and D-dopachrome tautomerase (DDT) in the MIF family. More importantly, 4-IPP also exhibits inhibition of

MIF-related proliferative and proinflammatory activity in cells and animal models. However, the proteome-wide

selectivity of 4-IPP is elusive. In our study, we synthesized a series of analogs of 4-IPP to explore the

structure-activity relationships. To construct a probe for protein detection and enrichment, one of the most potent

derivative was tagged with fluorephore or biotin, which did not weaken the potency of it. In the experiments

with purified proteins, the probe react with both MIF and DDT in a dose- and time-dependent manner, and the

reacted residue was 1-proline. By specifying the incubation time and using a MIF-selective blocker molecule, we

can selectively label MIF or DDT with our probe. In a more complex cell lysate labeling assay, the probe

exhibited a surprising selectivity on a band around 15 kDa, which contains both MIF and DDT. Of note, the

probe was capable to localizing intercellular MIF and DDT in A549 and HeLa cells. Therefore, our study not

only demonstrates the selectivity of 4-IPP derivatives, but also provides a powerful tool for MIF and DDT

research.

References

1) Shannon, D. Alexander, et al. J Am Chem Soc 2014, 136, 3330.

2) Schardon, Christopher L., et al. Chembiochem 2017, 18, 1551.

3) Zheng, Lin, et al. FASEB J 2019, 33, 7667.

EFMC-ISMC | 173

C004

MOLECULAR PROBE TO ISOLATE DLODP: AN ENZYME

INVOLVED IN TYPE I CONGENITAL DISORDERS OF

GLYCOSYLATION (CDG-I)

Michael BOSCO (1), Su-Jin PAIK (2), Isabelle CHANTRET (2), Stuart E.H. MOORE (2), Patricia

BUSCA (1), Christine GRAVIER-PELLETIER (1)

1) Université de Paris, UMR 8601 CNRS, LCBPT, F-75006 Paris, FRANCE2) Université de Paris, INSERM U1149, Faculté de Médecine Xavier Bichat, Paris, FRANCE

Protein N-glycosylation occurs by the transfer of an oligosaccharide from a lipid linked precursor (Glc

3

Man

9

GlcNAc

2

-PP-dolichol, DLO) onto newly synthesized proteins in the lumen of the endoplasmic reticulum. This

process is essential for life, and mutations in genes required for the biosynthesis of DLO underlie several of the

rare Type I Congenital Disorders of Glycosylation (CDG-I).

1

These autosomal recessive metabolic disorders are

characterised at the biochemical level by hypoglycosylated serum glycoproteins.

2,3

DLO diphosphatases

(DLODPs), orphan activities whose proteins/genes have not been identified yet, cleave DLO intermediates seen

in certain pathophysiological situations like CDG-I,

4,5

but their roles in normal and disease cells remain to be

determined. We hypothesize that DLODPs destroy toxic truncated DLO intermediates, which are generated

under pathological/stress conditions like CDG-I, that would otherwise cause cell pathology. In order to identify

DLODP genes and test this hypothesis our goal is to synthesize a probe with a photoactivable moiety able to

crosslink the diphosphatase.

 

In this communication,

6

we will describe the synthesis of this molecular probe which involves the preparation of

three building blocks: a biotinylated azide A, a phosphorylated alkyne B and a solanesyl phosphate C.

References

1) Haltiwanger RS, Lowe JB. Annu Rev Biochem. 2004, 73:491–537.

2) Haeuptle MA, Hennet T. Hum Mutat. 2009, 30(12):1628–41.

3) Jaeken J, Matthijs G. Annu Rev Genomics Hum Genet. 2001, 2:129–51.

4) Massarweh A, Bosco M, Iatmanen-Harbi S, et al. J Lipid Res. 2016, 57:1029–42.

5) Massarweh A, Bosco M, Iatmanen-Harbi S, et al. J Lipid Res. 2016, 57:1477–91.

6) The financial support of this work by ANR is gratefully acknowledged (ANR-18-CE44-0007-02).

174 | EFMC-ISMC

C005

DEVELOPMENT AND APPLICATION OF SUBTYPE-SELECTIVE

FLUORESCENT ANTAGONISTS FOR THE STUDY OF THE HUMAN

A1AR ADENOSINE RECEPTOR IN LIVING CELLS.

Eleonora Comeo (1,3,5), Stephen John Hill (4,5), Barrie Kellam (3,5), Lauren T. May (2), Michelle Halls

(2), Peter J. Scammells (1)

1) Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, VIC, Australia2) Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, VIC, Australia

3) Division of Biomolecular Sciences and Medicinal Chemistry, School of Pharmacy, University of Nottingham,Nottinghamshire, UK

4) Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham,Nottinghamshire, UK

5) Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, TheMidlands, UK

The adenosine A

receptor (A

1

AR) is a purinergic receptor that provides important therapeutic opportunities for

a number of conditions including congestive heart failure, tachycardia and neuropathic pain.

However, despite

the therapeutic potential offered by this important membrane protein, the translation of the A

1

AR ligands from

the bench to the clinic has been hampered by many challenges, mainly associated to the ubiquitous distribution

of this receptor in the body, leading to on-target side effects. Real-time monitoring of the differences in A

1

AR

pharmacology across tissues, cell types and subcellular compartments could provide additional insights on its

modulatory role in both health and disease and support the development of more efficacious drugs. This

additional knowledge could be achieved by employing fluorescent ligands which specifically target the A

1

AR in

its native cellular context.2,3 

In the present study, we set to develop a novel class of fluorescent ligands for the A

1

AR exploring two different

positions of the prototypical xanthine scaffold for linker/fluorophore elaboration.

After a preliminary screening

of a series of amino functionalized congeners equipped with linkers of different length and physiochemical

composition, it was identified the best template upon which append a series of commercially available

fluorophores. Our synthetic efforts, which involved over 15 steps synthesis, led to two families of fluorescent

analogues, including probes featuring the 4,4-difluoro-4-bora-3a,4a-diaza-s-indacene (BODIPY) dyes alongside

water-soluble dyes such as sulfonated cyanine-5 (Sulfo-Cy5). This focused library of eight fluorescent ligands

was thoroughly characterised pharmacologically, showing that these novel probes retained binding affinity to the

target receptor, functional activity (antagonism of A

1

AR) and most importantly, selectivity with respect to

binding the other three adenosine receptor subtypes (A

2A

AR, A

2B

AR and A

3

AR). Subsequently, we evaluated

the utility of the novel probes as tracers in competition ligand binding assays, ultimately determining binding

affinities of known unlabelled A

1

AR ligands that are in very good agreement with literature values. Lastly, we

used three fluorescent A

1

AR antagonists for visualizing specific fluorescent labelling of engineered A

1

AR

(SNAP-tagged) localized at the membrane of living cells by confocal imaging and Total Internal Reflection

Fluorescence Microscopy (TIRF-M). We envisage that these novel A

1

AR fluorescent probes will be valuable

pharmacological tools to the wider scientific community to aid in understanding of the molecular mechanisms

underlying A

1

AR pharmacology in health and disease.

5

References

1) Gao, Z.-G. et al., The Adenosine Receptors, 2018, 34, 59-89

2) L.A. Stoddart et al., Nature Methods, 2015, 12 (7), 661-663

3) Comeo, E. et al., J Med Chem, 2020, 63 (5), 2656-2672

4) W.F. Kiesman et al., J. Med. Chem, 2006, 49, 7119-7131

EFMC-ISMC | 175

C006

INVERSE AGONISTS OF 5-HT6 RECEPTOR-OPERATED SIGNALING

AS A NEW PARADIGM TO TREAT NEUROPATHIC PAIN AND

COGNITIVE CO-MORBIDITIES

Marcin Drop (1,2), Pierre-Yves Martin (5), Stéphane Doly (5), Vittorio Canale (1), Séverine

Chaumont-Dubel (3), Maria Walczak (1), Al Mahdy Hamieh (5), Eric Chapuy (5), Grzegorz Satała (4),

Paulina Koczurkiewicz (1), Andrzej J. Bojarski (4), Elżbieta Pękala (1), Xavier Bantreil (2), Gilles Subra

(2), Jean Martinez (2), Maciej Pawłowski (1), Frederic Lamaty (2), Philippe Marin (3), Alain Eschalier (5),

Christine Courteix (5), Paweł Zajdel (1)

1) Faculty of Pharmacy, Jagiellonian University Medical College, 30-688 Kraków, Poland2) IBMM, Université de Montpellier, CNRS, ENSCM, Montpellier, France

3) Institut de Génomique Fonctionelle, Université de Montpellier, CNRS INSERM,34094 Montpellier, France

4) Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Kraków, Poland5) Université Clermont Auvergne, INSERM U1107, NEURO-DOL, F-63000, Clermont-Ferrand, France

An increasing body of evidence supports the pronociceptive effects of serotonin and puts forward serotonin type

6 receptor (5-HT6R) as a potential target for the treatment of neuropathic pain. The high level of constitutive

activity together with the complex signaling network operated by 5-HT6R enable to design ligands with a

particular functional profile (neutral antagonism or inverse agonism) to follow distinct pharmacological

responses [1, 2]. In line with our findings indicating a non-physiological activation of mTOR pathway by

constitutively active spinal 5-HT6R, application of 5-HT6R inverse agonists might be a valuable strategy [3, 4].

Recently, our investigations identified a compound PZ-1388 designed on novel 2-phenyl-1H-pyrrole-3-carboxamide framework [3]. The selected compound displayed an inverse agonism of the 5-HT6R at

Gs and Cdk5 signaling as well as inhibited 5-HT6R-operated mTOR signaling pathway. PZ1388 showed high

selectivity over off-targets, high metabolic stability and was brain-penetrant. Finally, PZ1388 dose-dependently

alleviated painful symptoms in two preclinical models of traumatic (spinal nerve ligation) and toxic (oxaliplatin

administration) neuropathy as well as reverced cognitive deficits in neuropathic rats. Of note, neutral antagonists

(CPPQ [5]) failed to produce effect in the above tests. These data suggest that targeting constitutively active

5-HT6Rs migth be efficient strategy to alleviate painfull symptoms and associated cognitive comorbidities in

neuropathic pain.

 

The study was supported from Agence Nationale de la Recherche (ANR) (N° ANR-17-CE16-0010-01,

ANR-17-CE16-0013-01, ANR-19-CE18-0018-01 and program “Investissements d’Avenir’’16-IDEX-0001 CAP

20-25), the Institut National de la Sante et de la Recherche Medicale (INSERM), The Centre National de la

Recherche Scientifique (CNRS), the Auvergne-Rhone-Alpes Region (France), the Universite Clermont

Auvergne, the Universite de Montpellier and the National Science Centre, Poland (grant N°

2016/21/B/NZ7/01742).

References

1) Barnes N.M., Ahern G.P., Becamel C. et al. Pharmacol. Rev. 2021, 73, 310–520.

2) Chaumont-Dubel S., Dupuy V., Bockaert J. et al. Neuropharmacology 2020, 172, 107839.

3) Martin P.Y., Doly S. Hamieh A.M., et al. Prog. Neurobiol. 2020, 193, 101846.

4) Zajdel P., Drop M., Canale V. et al. WO2020117075A1, 2020.

5) Grychowska K., Satała G., Kos T. et al. ACS Chem. Neurosci. 2016, 7, 7, 972-983.

176 | EFMC-ISMC

C007

SPECTROPHOTOMETRIC STUDIES OF NOVEL FLUORESCENT

SENSORS AND THEIR BIOLOGICAL APPLICATION FOR ALBUMIN

DETECTION

Patryk Szymaszek, Joanna Ortyl

Cracow University of Technology, Department of Chemical Engineering and Technology, Poland

Albumin is the main protein of blood serum. It is also the main transport protein in the body and acts as a

transporter of many important compounds such as hormones, fatty acids, metal ions, amino acids and drugs. The

ability of albumin to bind with toxic substances in order to excrete them from the body demonstrates its

importance in the process of keeping the body's organs and tissues in good condition. Therefore, the search for

simple and convenient methods to determine albumin content is important in medical diagnosis. Considering the

importance of developing new sensors, it was decided to investigate new 3-(2-pyridyl)chromen-2-one derivatives

for the role of fluorescent sensors. For this purpose, absorbance and fluorescence studies as well as the

interaction of the obtained derivatives with BSA were carried out in order to determine the binding parameters.

The derivatives under study have a high molar extinction coefficient value of over 10,000 dm

3

·mol

-1

·cm

-1

at

absorption maxima suggesting their high potential as fluorescent sensors. Investigated derivatives bind to bovine

serum albumin and exhibit high sensitivity to changes in protein concentration. Calculated values of LOD and

LOQ are of the order of magnitude 10

-8

, while the concentration of albumin in blood is of the order of magnitude

10

-5

. Moreover, the determined binding parameters of the interaction of the sensors with albumin indicate static

BSA fluorescence quenching mechanism by formation of a complex. Obtained thermodynamic parameters

suggests that the binding of the probes to BSA is due to a spontaneous complexation. Moreover, FRET

experiment demonstrated that energy transfer between BSA and probe can occur with high probability

confirming static quenching mechanism. Analyzing the obtained results we can state that the investigated probes

can be potentially used for the role of non-covalent fluorescent sensors.

 

The present work was funded by the Ministry of Science and Higher Education (MNiSW) under the Diamond

Grant project, contract number 0072/DIA/2020/49.

EFMC-ISMC | 177

C008

HDAC10 SELECTIVE CHEMICAL PROBES FROM PAN-INHIBITOR

BY SUBSTITUTION OF A SINGLE METHYLENE IN VORINOSTAT

(SAHA)

Raphael R. Steimbach, Johanna Hummel-Eisenbeiß, Nikolas Gunkel, Aubry K. Miller

Cancer Drug Development Group, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg,Germany

Histone deacetylases (HDACs) have roles beyond histone deacetylation and epigenetic regulation with

non-histone and even non-peptide substrates. Class IIB member HDAC10 is known to be an effective N8

-acetylspermidine deacetylase

1

and may be involved in chemotherapy resistance of neuroblastoma via

autophagy.

2

HDAC10 deletion in mice was shown to increase the suppressive function of T-regulatory cells,

therefore playing an important role in transplant rejection and inflammatory disorders.

3

Selective inhibition of

HDAC10 has potential for therapeutic applications, but has been challenging so far, especially within the same

HDAC class.

4

 

We report the development of selective HDAC10 inhibitors (HDAC10i) by rational design based on the SAHA

scaffold. We mimicked the polyamine structure of the HDAC10 substrate by replacing a single methylene group

in the SAHA linker region with an amino functionality. This transformed the pan-inhibitor into a selective

HDAC10i with increased potency, while mostly eliminating affinity for Class I enzymes and the other Class IIB

member HDAC6. An SAR study yielded HDAC10 binders with IC

50

values in the low nanomolar range in a

cellular target engagement assay, while maintaining several hundred-fold selectivity over HDAC6 and all Class I

enzymes. We confirmed the remarkable HDAC10 selectivity of our chemical probes by Western blotting against

off-target HDAC activity markers for class I (acetyl-H3) and HDAC6 (acetyl-tubulin). Enzymatic inhibition of

HDAC10 in cells is demonstrated dose-dependently by targeted metabolomics of acetyl spermidines.

 

The unprecedented HDAC10 selectivity makes our chemical probes valuable tools to investigate HDAC10

biology and offers potential therapeutic applications in co-treatment cancer therapy strategies or as

immunomodulators.   

 

References

1) Y. Hai et al., Nat. Commun. 2017, 8, 15368.

2) I. Oehme et al., PNAS 2013, 110, E2592–2601.

3) S. Dahiya et al., Sci. Rep. 2020, 10, 424.

4) M. Géraldy et al., J. Med. Chem. 2019, 62, 4426–4443.

178 | EFMC-ISMC

C009

TARGETING THE PROTEIN KINASE S6K2 (p70S6Kβ) WITH

NON-CANONICAL ELECTROPHILIC WARHEADS

Stefan Gerstenecker, Lisa Haarer, Stefan Laufer, Matthias Gehringer

Institute of Pharmaceutical Sciences, University of Tuebingen, Auf der Morgenstelle 8, 72076 Tuebingen, Germany

The 70 kDa ribosomal protein S6 kinases S6K1 (p70S6Kα) and S6K2 (p70S6Kβ) constitute a sub-family of the

RSK serine/threonine kinases. As a part of the mTOR (mechanistic target of rapamycin) signaling pathway,

which is a key regulator of cell proliferation and growth that has been targeted in the treatment of cancer and

immune disorders,

1

both kinases are activated by the mTOR complex 1 (mTORC1). Due to their high degree of

similarity, a redundant functional profile was initially assumed and most research conducted so far has focused

exclusively on the role of S6K1. However, it is becoming increasingly apparent that each of these enzymes has

distinct functions in mTOR signaling.

2

Our understanding of S6K2-specific functions is poor and partly limited by the lack of suitable chemical probes.

While reasonably selective S6K1 inhibitors have already been developed, no such compounds are available for

S6K2.

3

Achieving selectivity between these two kinases is a challenge because of their virtually identical ATP

binding site differing only by a single amino acid. The latter residue is located at the gatekeeper+2 (GK+2)

position in the so-called "hinge region", where S6K1 harbors a tyrosine (Tyr-151) which is a cysteine (Cys-150)

in S6K2. Owing to the nucleophilic properties of the side chain thiol, cysteines can be covalently targeted via

weakly electrophilic functional groups often termed "warheads".

4

Since cysteines with an equivalent placement

are present in only four other, structurally more diverse kinases (namely FGFR4, TTK, MAPKAPK2 and

MAPKAPK3),

5

covalent bond formation can serve as a filter to promote selectivity against S6K1 and the

remaining kinome.

Addressing non-catalytic cysteines with "targeted covalent inhibitors" (TCIs) has recently gained popularity in

drug discovery and chemical biology, especially in protein kinase research. So far, cysteines at the kinases'

GK+2 position have been addressed only in the receptor tyrosine kinase FGFR4, e.g. by the covalent inhibitor

BLU9931 and similar compounds.

6

In order to target the equivalent cysteine in S6K2, we first generated hybrid

structures combining the warhead portion of BLU9931 with the scaffold of the S6K1 inhibitor PF-4708671.

3

These acrylamide-based compounds, however, showed relatively weak inhibitory potencies. By employing

non-canonical electrophilic warheads based on nucleophilic aromatic substitution reactions (S

N

Ar), we were

recently able to generate a second generation of S6K2 inhibitors. While featuring low intrinsic thiol reactivity,

these compounds showed strongly increased (low nanomolar) potency and high selectivity against other kinases

with an analogous cysteine but also in the kinome. Besides the generation of isoform-specific S6K2 inhibitors,

this approach may serve as a blueprint for the use of non-canonical warhead chemistry in the development of

cysteine-targeted chemical probes or drugs.

References

1) Saxton, R. A., Sabatini, D. M. Cell 2017, 168(6), 960–976.

2) Pardo, O. E., Seckl, M. J. Front. Oncol. 2013, 3, 191.

3) Pearce, L. R., Alton, G. R., Richter, D. T., Kath, J. C., Lingardo, L., Chapman, J., Hwang, C., Alessi D. R. Biochem J.

2010, 431(2), 245−255.

4) Gehringer, M., Laufer, S. A. J. Med. Chem. 2019, 62(12), 5673–5724

5) Chaikuad, A., Koch, P., Laufer, S. A., Knapp, S. Angew. Chem. Int. Ed. 2018, 57(16), 4372−4385.

6) Hagel, M., Miduturu, C., Sheets, M., Rubin, N., Weng, W., Stransky, N. et al. Cancer Discov. 2015, 5(4), 424−437

EFMC-ISMC | 179

C010

A COST-EFFECTIVE AND EASY-TO-USE METHOD TO ASSESS

CELL PERMEABILITY OF NEW PROTAC E3 LIGASE BINDERS

Sara Pannilunghi (1,2), Sébastien Tardy (1,2), Aurélie Gouiller (1,2), Leonardo Scapozza (1,2)

1) School of Pharmaceutical Sciences, University of Geneva, Switzerland2) Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Switzerland

Proteolysis-Targeting Chimeras (PROTACs) represent a novel and emerging tool in targeted protein

degradation that is currently receiving much attention for its therapeutic potential. PROTACs are bispecific

molecules containing an E3 ligase binder, a linker of various lengths, and a binder of the target of interest (TOI).

PROTACs are designed to create a selective bridge between the E3 ligase and the TOI, promoting

polyubiquitination and subsequent proteasomal degradation of disease-causing proteins. While more than 600

different E3 ligases are predicted to be encoded by the human genome, so far only a few of them have been

exploited by PROTACs, including cereblon (CRBN), Von Hippel–Lindau (VHL), Mouse Double Minute 2

homolog (MDM2), and cellular Inhibitor of Apoptosis Protein 1 (cIAP1).

(1)

In this perspective, it is clear that

identifying new small molecules to broaden the E3 ligase binder toolbox would be strongly desirable for

improved compartment-specific protein degradation. Despite the tremendous advancements and breakthroughs

that pushed the first chimeric degraders into clinical trials,

(2)

application of PROTAC technology is often

affected by poor cellular permeability and the mechanism of cell-penetration itself has been largely

underexplored.

(3)

Emerging biological tools yielding information on target engagement,

(4)

and quantitatively

measuring cell penetration of biomolecules

(5)

are now available for the in vitro evaluation of PROTACs in-cell

accumulation but they usually require specific and sophisticated technological settings.

This project aims to provide medicinal chemists, who are currently working on the conception and development

of PROTACs, with a cost-effective and easy-to-use method for the rapid evaluation of cell permeability for new

linker-conjugated E3 ligase binders, by exploiting the fluorochromaisa phenomenon and the combinatorial

potential of click chemistry. Knowing that several thalidomide-based CRBN PROTACs have been showing

activity within cells,

(6) 

we first chose to synthesize a 4-hydroxylthalidomide PROTAC as a positive control.

Click chemistry reaction was selected for in parallel coupling of the tetraethylene glycol-conjugated E3 ligase

binder and a fluorescent tracer, leading to a fluorescein-based PROTAC assembly. In a second time, we

investigated a novel E3 ligase binder, the Nedd-4 ligand, in the future perspective of exploiting the HECT

ubiquitin E3 ligase as a putative degrader tool for membrane proteins. In vitro studies carried out to evaluate and

quantify cell penetration of the two fluorescein-based PROTACs in Hela and RCC 769P cells have revealed very

promising results, paving the way for our model to be further employed in monitoring cell permeability

parameters of other PROTAC components.

References

1) Schapira M, Calabrese MF, Bullock AN, Crews CM. Targeted protein degradation: expanding the toolbox. Nat Rev Drug

Discov. 2019;18(12):949-63.

2) Proof-of-Concept with PROTACs in Prostate Cancer. Cancer discovery. 2020;10(8):1084.

3) oley CA, Potjewyd F, Lamb KN, James LI, Frye SV. Assessing the Cell Permeability of Bivalent Chemical Degraders

Using the Chloroalkane Penetration Assay. ACS chemical biology. 2020;15(1):290-5.

4) Guo WH, Qi X, Yu X, Liu Y, Chung CI, Bai F, et al. Enhancing intracellular accumulation and target engagement of

PROTACs with reversible covalent chemistry. Nat Commun. 2020;11(1):4268.

5) Peraro L, Deprey KL, Moser MK, Zou Z, Ball HL, Levine B, et al. Cell Penetration Profiling Using the Chloroalkane

Penetration Assay. J Am Chem Soc. 2018;140(36):11360-9.

6) Collins I, Wang H, Caldwell JJ, Chopra R. Chemical approaches to targeted protein degradation through modulation of the

ubiquitin-proteasome pathway. Biochem J. 2017;474(7):1127-47.

180 | EFMC-ISMC

C011

SYNTHESIS OF NEW FLUORESCENT CHEMICAL PROBES

TARGETING BACTERIAL EFFLUX TO EARLY DETECT AND FIGHT

THE FIRST BARRIER IN BACTERIA ANTIBIOTIC RESISTANCE

Johan Revol-Tissot, Julia Vergalli, Gérard Boyer, Jean-Marie Pagès, Jean-Michel Bolla, Sandrine Alibert

Aix Marseille Univ, INSERM, SSA, MCT, FAC PHARM, Marseille, France

Antimicrobial resistance (AMR) is one of the more serious problem of Public Health. The 2014 WHO’s global

report [1] and the more recent of J. O’Neill [2] outlined worrying levels of AMR worldwide involving

therapeutic failure especially in Gram-negative bacterial diseases. Resistance is a natural response of

microorganisms to counteract pharmacological effects of active agents. A lot of mechanisms as membrane

impermeability, enzymatic and intracellular target alterations contribute to the Multi-Drug Resistance (MDR)

phenotypes. Furthermore, efflux overexpression is a major early-stage trigger in the MDR setting up.

Resistance-Nodulation-Cell-Division (RND) efflux pump superfamily constitutes a tripartite protein system

which is the resistance first line in Gram-negative bacteria [3]. They can extrude structurally different substances

outside the cell space; hence antibiotics struggle to reach effective intracellular concentrations. Thanks to the

spectrofluorometric method developed in our laboratory [4], we are working on the use of new natural product

derivatives with high and linear fluorescent signal with their intracellular concentration. Their efflux substrate

properties are useful reference to detect the capacity of antibiotics to cross membranes and to accumulate in

bacteria especially the ones which express MDR resistance. These compounds must not have cytotoxic effects to

minimize their resistance impact. Starting from a screening of chemical libraries of natural products and by a

drug design approach, pharmacophores and their positions were identified on the fluorescent scaffold. Then,

Structure-Activity Relationship studies allowed us to highlight physicochemical properties responsible not only

for a high fluorescence intensity but also for substrate efflux-pump features. These results will guide the design

of a real-time diagnosis trial allowing antibiotic drug accumulation monitoring, identification, minimization, and

prevention in the early appearance of efflux bacterial resistance in clinic [5] while decreasing patient exposure to

antibiotics.

References

1) Antimicrobial Resistance: Global Report on Surveillance 2014. WHO. Available from:

http://www.who.int/drugresistance/documents/surveillancereport/en/

2) The Review on Antimicrobial Resistance Chaired by Jim O’Neill. Tackling Drug-Resistant Infections Globally: final

report and recommendations. 19 May 2016.

3) S. Alibert, J. N’gompaza Diarra, J. Hernandez, et al. Multidrug efflux pumps and their role in antibiotic and antiseptic

resistance: a pharmacodynamic perspective, Expert Opinion on Drug Metabolism & Toxicology. Nov 2016; 13(3):301-309

4) Vergalli, J., Atzori, A., Pajovic, J. et al. The challenge of intracellular antibiotic accumulation, a function of

fluoroquinolone influx versus bacterial efflux. Commun Biol 3, 198 (2020).

5) Puttaswamy S. et al. A Comprehensive Review of the Present and Future Antibiotic Susceptibility Testing (AST) Systems.

Arch Clin Microbiol. 2018, Vol No: 9 Iss No: 3:83.

EFMC-ISMC | 181

C012

THE NOVEL 1,3,5-TRIAZINE DERIVATIVES AS INNOVATIVE

POTENTIAL ACHE AND BACE1 INHIBITORS

Dawid Maliszewski (1), Agnieszka Wróbel (1), Beata Kolesińska (2), Justyna Frączyk (2), Danuta

Drozdowska (1)

1) Department of Organic Chemistry, Faculty of Pharmacy, Medical University of Bialystok,Mickiewicza 2 street, 15-089 Bialystok, Poland

2) Institute of Organic Chemistry, Technical University of Lodz, Poland

Objectives: The main goal of research was design, synthesis and biological evaluation of the novel hybrid

2,4,6-trisubstituted-1,3,5-triazine derivatives. The designed compounds contained three different functional

groups (methoxy group, alkylating group and dipeptide group). Obtained compounds presented inhibitory

properties of both acetylcholinesterase (AChE) and β-secretase (BACE1) enzymes.

 

Scheme 1.Synthesis of the new analogs of triazine.

Methods: As a starting material was used 2-methoxy-1,3,5-triazine. Dipeptide was obtained by solid phase

synthesis. Derivatives 4a-4h (Scheme 1) were obtained in two-steps reaction by adding dipeptide (first step) and

DABCO (second step). Product was analyzed by thin layer chromatography experiments (TLC), RP-HPLC,

mass spectroscopy (MS) and

1

H-NMR and

13

C-NMR spectra. The inhibitory activity of triazine derivatives

4a-4h was evaluated in vitro against AChE (from electrophorus electricus) using Ellman's method and BACE1

(from equine serum) using a forester resonance energy transfer (FRET).

Result and Conclusions: All compounds displayed considerable AChE and BACE1 inhibition. The most active

against both AChE and BACE1 enzymes was 4a with an inhibitory concentration of (AChE IC50 = 0.051µM;

0,055 µM ) and (BACE1 IC50 = 9.00 µM; 11.09 µM) respectively.

Funding: Publication financed under the project№ POWR.03.02.00-00-I051/16 from European Union funds ,

PO WER 2014-2020, grant№ 07/IMSD/G/2019

References

1) Kolesińska, B.; Drozdowska, D.; Kamiński, Z.J. The new analogues of nitrogen mustard with one, two or three

2-chloroethyloamino fragments. Reactions with nucleophiles. Acta Pol. Pharm. 2008, 65, 709–714.

2) Wróbel, A.; Kolesińska, B.; Frączyk, J.; Kamiński, Z.J.; Tankiewicz-Kwedło, A.; Hermanowicz, J.; Czarnomysy, R.;

Maliszewski, D.; Drozdowska, D. Synthesis and cellular effects of novel 1,3,5-triazine derivatives in DLD and Ht-29 human

colon cancer cell lines. Invest. New Drugs 2020, 4, 990–1002.

3) Han, J.; Ji, Y.; Youn, K.; Lim, G.; Lee, J.; Kim, D. H.; Jun, M. Baicalein as a Potential Inhibitor against BACE1 and

AChE: Mechanistic Comprehension through In Vitro and Computational Approaches. Nutrients 2019, 11, 2694.

4) Yiannopoulou, K. G.; Papageorgiou, S. G. Current and Future Treatments in Alzheimer Disease: An Update. J. Cent.

Nerv. Syst. Dis. 2020, 12, 1179573520907397.

182 | EFMC-ISMC

C013

VALIDATING TARGETS FOR TARGETED PROTEIN DEGRADATION

USING DTAG – A COMPREHENSIVE WORKFLOW SOLUTION

Pouya Amrollahi (2), Marnie Wallin (3), Nithya Jesuraj (3), Jeff Cooper (3), Chris Heger (3), Charles

Haitjema (2), Hannah Maple (1)

1) Bio-Techne, The Watkins Building, Atlantic Road, Avonmouth, Bristol, BS11 9QD2) Bio-Techne, 3001 Orchard Parkway, San Jose, CA

3) Bio-Techne, 614 McKinley Place NE, Minneapolis, MN 55413

Degraders and the field of Targeted Protein Degradation offer a mechanistically differentiated way to modulate

target proteins using small molecules. While a small molecule inhibitor will block or modulate a specific protein

domain or function (for example an enzymatic role), a Degrader will knockdown the entire protein, removing all

possible functions. Inhibition therefore does not always phenocopy degradation and careful assessment of

potential targets for new Degrader development programs is vital. Target validation is therefore an important

stage of the Degrader development project workflow.

TAG degradation technology offers a generalizable strategy to degrade, in principle, any intracellular protein of

interest (POI). Its key benefit is that it does not rely on the pre-existence of a ligand or PROTAC® for the POI.

The broad applicability that it offers makes this a useful strategy for exploration and validation of targets,

particularly in the context of new Degrader development programs.

Here we present two case studies illustrating the dTAG workflow. We present data to demonstrate the value of

testing both CRBN- and VHL-recruiting dTAG Degraders for each POI target as well as the use of negative

control Degraders. We also show the utility of using a FKBP12 antibody for detecting POI∙dTAG fusion

molecules, allowing for a more simplified procedure when probing multiple targets.

EFMC-ISMC | 183

C014

DESIGN AND SYNTHESIS OF A LIBRARY OF

PYRAZOLO[1,5-A]PYRIDINE FLUOROPHORES FOR DEVELOPING

NEW FLUORESCENT PROBES

Elena Martino (1), Stefano Sainas (1), Francesco Bavo (2), Claudio Garino (1), Donatella Boschi (1), Bente

Frølund (2), Marco Lolli (1)

1) Department of Science and Drug Technology, Universita' degli Studi di Torino, via Giuria 9, 10125-Torino, Italy2) Department of Medicinal Chemistry, University of Copenaghen, Jagtvej 162, 2100 København Ø, Denmark

Fluorescence imaging is a non-invasive technique useful to understand and visualize biological processes in

living organisms.

1

 Most of the available dyes normally used for this aim, suffer for various limitations such as

large size, poor water solubility or low cell permeability.

2

 In recent years, several research has been focused on

developing novel small fluorescent dyes with low molecular weight, good signal to noise ratio and optimized

proprieties that can be applied to most biological samples without disturbing the physiological envirorment.

Pyrazolo[1,5-a]pyridin-2-ol is a small scaffold with a strong emission in the blue region, recently studied

as bioisoster of the carboxylic acid moiety.

4,5

 With the purpose to increase the fluorescent properties of this

scaffold, different strategies were investigating. Generally, the donor–π-acceptor moiety is the key structural

framework modulated to give fluorescence to small organic compounds. To improve the intrinsic properties of

our probe (absorption/emission/Φ/Stokes shift), the latter was modulated with different stronger electronic

drawing and accepting groups. Furthermore, the scaffold was studied by expanding the π-conjugated network

with the introduction of a wider structure to improve the emission profile of the system (see Figure). Preliminary

data of the fluorescent proprieties of the designed target compounds, showed a bathochromic (red) shift in the

UV–vis absorption and emission spectra. Further studies need to be conducted to establish the best attachment

position to use our molecule as an imaging probe. Theoretical design, synthesis and fluorescent proprieties will

be here presented and discussed.

Figure: Design of a new library based of pyrazolo[1,5-a]pyridin-2-ol scaffold.

References

1) Xia, T., Li, N., & Fang, X. Annual Review of Physical Chemistry, (2013), 64(1), 459–480

2) Joomyung V. Jun, David M. Chenoweth and E. James Petersson, Org. Biomol. Chem., (2020), 18, 5747

3) Neto, B. A. D., Carvalho, P. H. P. R., & Correa, J. R. Accounts of Chemical Research, (2015) 48(6), 1560–1569

4) S. Sainas, A. C. Pippione, E. Lupino, M. Giorgis, P. Circosta, V. Gaidano, P. Goyal, D. Bonanni, B. Rolando, A. Cignetti,

A. Ducime, M. Andersson, M. Jarva, R. Friemann, M. Piccinini, C. Ramondetti, B. Buccinna, S. Al-Karadaghi, D. Boschi, G.

Saglio, and M. L. Lolli. J. Med. Chem. (2018), 61, 6034-6055

5) Sainas, S. et al. J. Med. Chem., (2021), in press.

184 | EFMC-ISMC

C015

DISCOVERY OF SELECTIVE FRAGMENT-SIZED

IMMUNOPROTEASOME INHIBITORS

Matic Proj (1), Levente Kollár (2), Martina Gobec (1), Bence Szilágyi (2), Damijan Knez (1), Péter

Ábrányi-Balogh (2), Dávid Bajusz (2), György G. Ferenczy (2), Stanislav Gobec (1), György M. Keserű

(2), Izidor Sosič (1)

1) University of Ljubljana, Faculty of Pharmacy, Chair of Pharmaceutical Chemistry, Ljubljana, Slovenia2) Hungarian Academy of Sciences, Research Centre for Natural Sciences, Medicinal Chemistry Research Group, Budapest,

Hungary

Proteasomes contribute to the maintenance of protein homeostasis, and their inhibition is a viable approach for

the treatment of certain types of cancer and autoimmune diseases. To limit the undesirable side-effects,

considerable efforts have been made to selectively inhibit the immunoproteasome (iCP). The aim of our study

was to discover new chemotypes capable of selectively inhibiting the iCP using a fragment-based approach. This

strategy has not previously been performed with the iCP, but is based on its success against a wide range of

pharmacological targets. [1]

 

Screening of an in-house fragment library led to the identification of benzo[d]oxazole-2(3H)-thiones, benzo[d]thiazole-2(3H)-thiones, benzo[d]imidazole-2(3H)-thiones, and 1-methylbenzo[d]imidazole-2(3H)-thiones as

selective inhibitors of the chymotrypsin-like (β5i) subunit of the iCP. Exploration of the structure-activity

relationships provided us with insights into possible vectors for fragment growth. Thorough characterization by

thiol reactivity assays suggested that these initial hits inhibit the iCP by forming a disulfide bond with the Cys48

specifically present in the β5i active site. To obtain a more biologically tractable covalent interaction, a warhead

scan was performed. We discovered benzoXazole-2-carbonitriles as promising starting points for the

development of selective immunoproteasome inhibitors with non-peptidic scaffolds (Figure 1). [2] We are now

performing a fragment growing campaign to further optimize these compounds.

Figure 1: Schematic representation of the workflow that led us to the discovery of immunoproteasome inhibitors

References

1) W. Jahnke, J. Med. Chem. 2020, 63, 24, 15494–15507.

2) L. Kollár, 2021, manuscript under review.

EFMC-ISMC | 185

C016

TOWARDS PROTAC-MEDIATED DEGRADATION OF CBP/EP300

Leonardo Palaferri (1), Iván Cheng-Sánchez (1), Katherine Rollins (1,2), Mariia Kirillova (1), Eleen Laul

(1), Anna Müller (2), Lars Wiedmer (2), Amedeo Caflisch (2), Cristina Nevado (1)

1) Department of Chemistry, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland2) Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland

CREB Binding Protein (CBP) and EP300 are two proteins with a high sequence homology that participate in

chromatin remodelling and act as co-activators of several transcription factors.

1

Their lysine acetyl transferase

domain (KAT) and bromodomain (BRD), which respectively acetylate and bind acetylated lysine residues on

histone tails and other proteins, are crucial for these functions.

2

CBP/EP300 are often overexpressed, mutated or

dysregulated in cancer, making them extremely interesting targets for medicinal chemistry.

1,2,3

After developing CBP/EP300 bromodomain-binding molecules with sub-micromolar potency and promising

bioavailability,

4

we now aim to elucidate the contribution of CBP/EP300 to gene regulation by inducing their

degradation through PROteolysis TArgeting Chimeras (PROTACs). PROTACs consist of a binder for a protein

of interest (POI) connected to an E3 ligase ligand through a linker. With a catalytic mechanism, PROTACs

promote the formation of a ternary complex with the POI and the E3 ligase leading to polyubiquitination of the

POI and consequent proteasome-mediated degradation. In such way, PROTACs enable loss of function of the

whole protein independently from the bound domain.

5

Starting from the analysis of X-ray crystal structures of our in-house

4

and other published probes

6

in complex

with CBP/EP300, we have designed and synthesised a series of bifunctional compounds with the ability to

simultaneously bind either the BRD or KAT domain and a E3 ligase. Empirical exploration of several linkers

and E3 ligands allowed optimisation of the probes’ potency, cell permeability and target engagement. Binding

affinity and ternary complex formation have been measured with biochemical assays while cellular target

engagement, degradation and phenotypical effects were evaluated in multiple cell systems. In conclusion, we

developed a library of compounds with some valid candidates to enable CBP/EP300 degradation as a powerful

therapeutic alternative to classical protein inhibition.

References

1) R. Kemler et al. EMBO J. 2000, 19, 1839-1850

2) P. A. Cole et al. Chem. Rev. 2015, 115, 2419-2452

3) J. S.Carroll et al. Nat. Rev. Cancer 2007, 7, 713-722

4) C. Nevado et al. ACS Med. Chem. Lett. 2018, 9, 929-934

5) C. M. Crews et al. Nat. Rev. Drug Discov. 2017, 16, 101-114

6) J. R. Levell et al. ACS Med. Chem. Lett. 2020, 11, 1324-1320

186 | EFMC-ISMC

C017

SYNTHESIS AND IN VITRO STUDIES OF BIS-BENZIMIDAZOLE

DERIVATIVES TARGETING KINETOPLASTID PARASITES

J. Jonathan Nué Martinez, Christophe Dardonville

Instituto de Química Médica CSIC, Juan de la Cierva 3, Madrid 28006, Spain

Human infection by kinetoplastid parasites of the family Trypanosomatidae (Leishmania and Trypanosoma) is a

significant public health problem with widespread disease and limited therapeutic options. These diseases are

particularly devastating and are a cause of great morbidity and mortality in the developing world.

Chagas disease, caused by Trypanosoma cruzi, is a major illness in Latin America with 6–7 million infected

individuals, 25 million at risk of infection, and an annual death toll of over 10 000. It is also an emerging public

health problem in the United States, Canada and Europe due to immigration; an estimated 300,000 infected

individuals reside in the United States and 67,000 live in Spain.

1

The drugs currently used to treat Chagas

disease, nifurtimox and benznidazole, are decades old and have many limitations, including severe side-effects,

and low to medium efficacy and selectivity. Hence, new safe and effective drugs for the treatment of the

prevalent chronic stage of the disease, where the traditional drugs show low efficacy, are urgently needed.

Leishmaniosis is endemic in 97 countries, putting 350 million people at risk, and there are an estimated 12

million individuals currently infected, and 26 000–65 000 deaths worldwide each year. Leishmaniosis still

represent a global scourge and new therapeutic tools are necessary to replace the current treatments that are

expensive, difficult to administer, and nonspecific. The toxicity of current anti-parasite drugs coupled with the

rapid emergence of drug resistant Leishmania strains remain significant challenges for disease control.

Recent examples in the literature have shown that the mitochondrial DNA (kDNA) of kinetoplastid parasites is a

valuable target for chemotherapy. In fact, AT-specific minor groove binders that disrupt DNA-protein

interactions are an effective strategy to kill kinetoplastid parasites.

2-5

In this project, a series of bis-benzimidazole compounds based on the N-phenylbenzamide scaffold were

synthesized as potential kDNA binders. Their antiparasite activity was tested in vitro against three kinetoplastid

parasites: L. donovani, T. cruzi and T. brucei. The cytotoxicity against mammalian cells was also determined to

calculate the selectivity index (SI) of the compounds.

EFMC-ISMC | 187

Posters - Chemical Biology D. Molecular Imaging Tools for Chemical Biology

188 | EFMC-ISMC

D001

DISCOVERY OF THE NEXT GENERATION TAU PET TRACER

PI-2620 FOR THE ASSESSMENT OF TAU PATHOLOGY IN

ALZHEIMER'S DISEASE AND OTHER TAUOPATHIES

Emanuele Gabellieri (1), Heiko Kroth (1), Jerome Molette (1), Francesca Capotosti (1), Cèdric Boudou

(1), Vincent Darmency (1), Tanja Juergens (1), Yvan Varisco (1), Efthymi Vokali (1), David Hickman (1),

Sonia Poli (1), Pfeifer Pfeifer (1), Andre Mueller Mueller (2), Mathias Berndt (2), Hanno Schieferstein (2),

Felix Odenb Odenb (2), Heribert Schmitt Willichb (2), Ludger Dinkelborg Dinkelborg (2), Andrew

Stephens (2)

1) AC Immune SA, EPFL Innovation Park, Building B , 1015 Lausanne, Switzerland2) Life Molecular Imaging GmbH, Tegeler Strasse 6-7, 13353 Berlin, Germany

Tau deposition is a key pathologic feature of Alzheimer’s disease (AD) and other neurodegenerative disorders.

The spreading of Tau neurofibrillary tangles across defined brain regions corresponds to the observed level of

cognitive decline in AD.

Positron-emission tomography (PET) has proven to be an important tool for the detection of amyloid-beta

aggregates in the brain and is currently explored for detection of pathological misfolded Tau in AD and other

non-AD tauopathies.

Several PET tracers targeting Tau have been discovered and tested in humans. However, limitations have been

reported especially regarding their off-target binding and ability to detect Tau aggregates in non-AD tauopathies.

To address these issues, we have designed and synthesized a series of ten fluoro-pyridine regioisomers attached

to the pyrrolo[2,3-b:4,5-c']dipyridine tricyclic core as part of our Morphomer

TM

library.

To enable in vitro and in vivo evaluation, the ten non-radioactive compounds and their corresponding precursors

were prepared. The nitro-group was selected as the leaving group for nucleophilic substitution with fluorine-18

(18F) to prepare the corresponding 18F-labeled compounds.

The affinity of the ten regioisomers to aggregated Tau was assessed using AD brain homogenates. Off-target

binding was evaluated in competition assays using radiolabeled MAO A / B and Aβ binders. The

pharmacokinetic profiles of the 18F-labeled compounds were evaluated in mice. In addition, the binding of

selected compounds to recombinant 4R Tau aggregates, Progressive supranuclear palsy (PSP) brain homogenate,

and Pick’s disease (PiD) brain homogenate was evaluated in competition experiments and by autoradiography.

PI-2620 was identified as the most promising candidate for clinical validation, showing high affinity to AD and

non-AD Tau aggregates, as well as excellent selectivity and pharmacokinetic properties in mice. The PK of

PI-2620 was further investigated in non-human primates confirming the mice data.

Thus, PI-2620 is currently being evaluated in several clinical trials to define its pharmacokinetic properties and

metabolic profile in order to confirm its suitability to image pathological Tau aggregates/aggregate folds in AD

and non-AD tauopathies.

EFMC-ISMC | 189

D002

MOLECULAR TOOLS TO STUDY AND CHARACTERIZE THE

LYSOSOMAL OLIGOSACCHARIDE TRANSPORTER (LOST)

Haifei GAO (1), Younes BOUZIDI (2), Morgane MAYE (3), Michaël BOSCO (1), Isabelle CHANTRET

(2), Sébastien FORT (3), Stuart MOORE (2), Patricia BUSCA (1), Christine GRAVIER-PELLETIER (1)

1) Université de Paris, LCBPT, UMR 8601CNRS, F-75006 Paris, France2) Inserm U1149, Faculté de Médecine Xavier Bichat, Paris, France

3) CERMAV-CNRS UPR 5301, Domaine Universitaire de Saint-Martin d'Hères, Grenoble, France

Glycoconjugates and oligosaccharides derived from pathogens

1

and the extracellular matrix

2

are naturally

detected by the innate immune system. Free oligosaccharides (fOS) generated during protein N-glycosylation

become proinflammatory (fOSp)

3

after partial demannosylation by an unknown process and contribute to certain

rare inherited inflammatory diseases. A lysosomal oligosaccharide transporter (LOST) assures intralysosomal

fOS demannosylation, but its role in the metabolism of fOSp is unclear. LOST is difficult to study because its

gene has not been identified.

In order to assay, characterize and identify LOST, we need different fluorescent, biotinylated and photoactivable

oligosaccharidyl probes. In this communication, we will describe the design, synthesis and biological activities

of these probes.

4

 

References

1) Franchi, L.; Muñoz-Planillo, R.; Núñez, G. Nat. Immunol. 2012, 13, 325. doi: 10.1038/ni.2231

2) Misra, S.; Hascall, V. C.; Markwald, R. R.; Ghatak, S. Front. Immunol. 2015, 6, 201. doi: 10.3389/fimmu.2015.00201

3) Fermaintt, C. S.; Sano, K.; Liu, Z.; Ishii, N.; Seino, J.;Dobbs, N.; Suzuki, T.; Fu, Y. -X.; Lehrman, M. A.; Matsuo, I.; Yan,

N. Nature Commun. 2019, 10, 2377. doi.org/10.1038/s41467-019-10319-5

4) We gratefully acknowledged the financial support of this work by FRM (DCM20181039551).

190 | EFMC-ISMC

D003

NEW BORINIC PROBES FOR FAST DETECTION AND IMAGING OF

HYDROGEN PEROXIDE

Blaise Gatin-Fraudet (1,2), Thomas Le Saux (3), Stéphanie Norsikian (1), Mathilde Pucher (2), Marie

Erard (4), Ludovic Jullien (3), Dominique Guianvarc'h (2), Dominique Urban (2), Boris Vauzeilles (1)

1) Université Paris-Saclay, CNRS, Institut de Chimie des Substances Naturelles, UPR 2301, 91198, Gif-sur-Yvette, France.E-mail: [email protected]

2) Université Paris-Saclay, CNRS Institut de Chimie Moléculaire et des Matériaux d'Orsay, UMR CNRS 8182, 91405, Orsay,France. E-mail: [email protected]

3) PASTEUR, Département de chimie, École Normale Supérieure PSL University, Sorbonne Université, CNRS, 75005 Paris,France

4) Université Paris-Saclay, CNRS, Insitut de Chimie Physique, UMR CNRS 8000, 91405, Orsay, France

Reactive oxygen species (ROS: hydrogen peroxide, hydroxyl and superoxide radicals) are by-products of aerobic

metabolism. Among them, hydrogen peroxide (H

2

O

2

) plays a crucial role in a wide range of physiological

processes in human.[1] However, when our cells are subjected to oxidative stress conditions, its overproduction

is directly or indirectly responsible for numerous damages at the molecular level, which can affect cellular

mechanisms. This process is associated with aging, as well as cancer and several neuro-degenerative diseases

such as Alzheimer’s or Parkinson’s.[2] The development of selective and sensitive tools allowing H

2

O

detection

in a biological context represents a great challenge for a better understanding of H

2

O

2

-mediated signaling in

physiological and pathological processes 

To date, several “off-on” small fluorescent probes triggered by H

2

O

have been developed for its detection.

Among them, probes based on the boronate oxidation are amongst the most effective for the detection of H

2

O

incellula.[3] But these probes also suffer from lack of reactivity, which is not fully satisfactory for biological

applications. To address this issue, we envisioned the use of borinic acids which due to electronic effects could

be more prone to oxidation compared to their boronic acid counterpart. 

Herein, we report the design, synthesis, and kinetic properties of the first borinic sensor for the fast detection of

hydrogen peroxide.[4] Furthermore, a comparative study with its boronic acid counterpart is presented in vitro and in a cellular context. 

 

References

1) a) H. Sies, Redox. Biol. 2017, 11, 613-619; b) A. van der Vliet, Y. M. Janssen-Heininger, J. Cell. Biochem. 2014, 115,

427-435.

2) a) K. Ishikawa et al., Science 2008, 320, 661-664; b) M. P. Mattson, Nature 2004, 430, 631-639.

3) a) B. C. Dickinson, C. Huynh, C. J. Chang, J. Am. Chem. Soc. 2010, 132, 5906-5915; b) L. Du, M. Li, S. Zheng, B. Wang,

Tetrahedron Lett. 2008, 49, 3045-3048; c) C. Yik-Sham Chung, G. A. Timblin, K. Saijo, C. J. Chang, J. Am. Chem. Soc.

2018, 140, 6109-6121.

4) B. Gatin-Fraudet, R. Ottenwelter, T. Le Saux, T. Lombès, A. Baron, Ph. Durand, S. Norsikian, G. Doisneau, Y.

Bourdreux, D. Guianvarc’h, M. Erard, L. Julien, D. Urban, B. Vauzeilles, ChemRxiv, 2020,

https://doi.org/10.26434/chemrxiv.12032670.v1

EFMC-ISMC | 191

D004

FLUORESCENT PROBES FOR IMAGING OF PROTEIN

NANOENVIRONMENT

Anna Wychowaniec, Karolina Konsewicz, Michal Jakubczyk, Michal Gladysz, Jacek L. Kolanowski

Institute of Bioorganic Chemistry Polish Academy of Sciences, 61-704, Poland

The necessity of a balanced pH level in different segments of biological object was proved to be crucial for the

functioning of a whole organism, individual cells or even single proteins. In case of proteins, changes in overall

pH of molecular microenvironment affect the attractions and reactivities between side chain groups. As a result,

changes in protein conformation and/or activity can be observed

[1]

affecting the affinity of ligands,

protein-protein interactions and enzymatic functions

[2]

,which influence physiological and pathological

processes.

[3]

In our work we aim at development of chemical tools to look into particular pH changes in protein

nanoenvironment (near or at the surface of it). These tools are fluorescent probes, covalently labeled to a chosen

endogenous protein of interest (POI)

[4]

. With use of probes responsive to pH changes

[5]

and super-resolution

microscopy techniques, information about local pH differences at a single protein may be obtained. This opens

up a possibility of analyzing physiological and pathological processes at currently inaccessible level and can be

extended to other analytes beyond pH. This work is funded by National Science Centre in Poland through OPUS

grant scheme (grant no. 2018/29/B/ST4/01498).

References

1) Zhou, H., Pang, X., Chem Rev. 2018, 118(4), 1691–1741.

2) Li, Z.Q. et al., J. Insect Physiol 2013, 59(3), 263-272.

3) Santos, J. et al. Cells 2020, 9(1), 145.

4) Tamura, T., Hamachi, I., J. Am. Chem. Soc. 2019, 141(7), 2782–2799.

5) Hou, J., Chem. Soc. Rev., 2017, 46, 2076-2090.

192 | EFMC-ISMC

D005

ONE CURIE LEVEL PRODUCTION OF [18F]SynVesT-1 TRACER AT

HIGH MOLAR ACTIVITY USING THE COPPER MEDIATED

FLUORODESTANNYLATION METHOD

Adrien Stouse, Sylvestre Dammicco, Christian Lemaire, André Luxen

Université de Liège,Bât. B30 CRC In vivo Imaging-Radiochemistry

Quartier Agora, allée du 6 Août 8, 4000 Liège, Belgique

The use of aromatic compounds labelled with fluorine-18 as radiopharmaceuticals is of major importance in PET

imaging. Recently a lot of work has been done to develop new approaches to access complex compounds

containing [

18

F]fluoroarenes which are  hard to obtain using classical Nucleophilic Aromatic Substitution

mechanism (S

N

Ar). One of these recent methods, introduced by Makavarage in 2016

1

, involves the use of

trialkyltin aromatic precursors and a copper (II) catalyst. In contrast to S

N

Ar, the fluorodestannylation doesn’t

require any activating group at the para or ortho position, which allows for late stage radiolabelling even from

structurally complex molecules. In 2019, this approach was simultaneously used by two research groups

2,3

to

synthesize SynVesT-1, a tracer for imaging SV2-A protein. This compound was successfully synthesized with a

radiochemical yield of 15-35% decay corrected and a molar activity of 6.5-8Ci/µmol. In this work, we present

how we were able to synthesize 900mCi of this radiopharmaceutical as a racemic mixture, and improve its molar

activity up to 36Ci/µmol after process modifications based on the 2017 study by Zarrad

4

.

 

 

References

1) Makaravage, K. J., Brooks, A. F., Mossine, A. V., Sanford, M. S. & Scott, P. J. H. Copper-Mediated Radiofluorination of

Arylstannanes with [18F]KF. Org. Lett. 18, 5440–5443 (2016).

2) Constantinescu, C. C. et al. Development and In Vivo Preclinical Imaging of Fluorine-18-Labeled Synaptic Vesicle

Protein 2A ( SV2A ) PET Tracers. Mol. Imaging Biol. 21, 509–518 (2019).

3) Li, S. et al. Synthesis and in Vivo Evaluation of a Novel PET Radiotracer for Imaging of Synaptic Vesicle Glycoprotein

2A (SV2A) in Nonhuman Primates. ACS Chem. Neurosci. 10, 1544–1554 (2019).

4) Zarrad, F., Zlatopolskiy, B., Krapf, P., Zischker, J. & Neumaier, B. A Practical Method for the Preparation of

[18F]-Labeled Aromatic Amino Acids from Nucleophilic [18F]Fluoride and Stannyl Precursors for Electrophilic

Radiohalogenation. Molecules 22, 2231 (2017).

EFMC-ISMC | 193

D006

STUDY OF RADIOSENSITIZING PROPERTIES INDUCED BY

PEGYLATED MAGNETIC NANOPARTICLES

Indiana Ternad (1), Dimitri Stanicki (1), Thomas Vangijzegem (1), Sébastien Penninckx (3), Robert

Muller (1,2), Stéphane Lucas (3), Sophie Laurent (1,2)

1) General, Organic and Biomedical Chemistry Unit, NMR and Molecular Imaging Laboratory, University of Mons(UMONS), 7000 Mons, Belgium

2) Center for Microscopy and Molecular Imaging (CMMI), 6041 Gosselies, Belgium3) Research Center for the Physics of Matter and Radiation (PMR-LARN), Namur Research Institute for Life sciences

(NARILIS), University of Namur, 5000 Namur, Belgium

In the field of radiosensitizing compounds, high-Z nanoparticles (NPs) have received growing interest. Up to

now, reports have highlighted the ability of such nanoparticles to increase tumor cell death upon irradiation,

improving thus, the radiation treatment efficiency. Despite some extensive studies led in the field (especially

focused on golds NPs (GNPs)), the mechanism(s) responsible for the radiosensitization effect of GNPs remains

poorly understood, and mainly focused on physical effects (i.e. dose increase due to secondary particle emission

after collision between the NPs and the ionizing radiations). Interestingly, recent studies suggest the central role

of some biochemical mechanisms on the observed radiosensitizing effect [1]. A significant correlation has been

made between the inhibition of the detoxification enzyme in GNP-treated cells and the magnitude of

radiosensitizing effect [2]. These enzymes play a key role in the detoxification system of cells by catalyzing the

transformation of reactive oxygen species into stable oxygen compounds such as O

2

 and H

2

O. 

In view of these elements, we were interested to study if such inhibition behavior may be evidenced for other

kind of NPs. Owing to their biocompatibility and superparamagnetic properties, iron oxide nanoparticles

(SPION) were selected. Stable SPION were obtained by coprecipitation, followed by a surface treatment using a

carboxylated-silane (i.e. TEPSA). PEG chains were further introduced by a subsequent coupling reaction [3].

The size, magnetic properties, surface modification were characterized using respectively transmission electron

microscopy (TEM), NMRD profile, dynamic light scattering. In addition, a feasibility study was first approached

by biocompatibility study and the cellular uptake evaluation. Irradiation of a lung carcinoma cell line incubated

with iron oxide nanoparticles evidenced a potent radiosensitizing effect, which suggests the use of our

nano-objects as a theranostic platform.

References

1) S.Penninckx, et.al. « Gold Nanoparticles as a Potent Radiosensitizer: A Transdisciplinary Approach from Physics to

Patient ». Cancers 12, 2020: 2021.

2) Penninckx, S. et al.« Thioredoxin Reductase Activity Predicts Gold Nanoparticle Radiosensitization Effect ».

Nanomaterials 9, 2 (2019): 295.

3) Stanicki, D. et al. « Carboxy-Silane Coated Iron Oxide Nanoparticles: A Convenient Platform for Cellular and Small

Animal Imaging ». J. Mater. Chem. B 2, 4 (2014): 387 97.

194 | EFMC-ISMC

D007

ILLUMINATING CANNABINOID RECEPTORS: FLUORESCENT

PROBES FOR SPECIFIC VISUALIZATION OF THE CANNABINOID

TYPE 2 RECEPTOR

Marc Nazare

Leibniz Research Institute for Molecular Pharmacology (FMP), Berlin, Germany

The cannabinoid type 2 receptor (CB2R) is a fundamental part of the endocannabinoid signaling system and

plays an important role in a number of pathophysiological processes like inflammatory pain, diabetic neuro- and

nephropathy, liver cirrhosis, and ischemic-reperfusion injury. However, due to the lack of suitable antibodies,

there are quite a few persisting questions on target occupancy, expression and activation levels in tissues that

remain to be clarified. Selective fluorescently labeled chemical probes are powerful tools to study CB2R in a

spatiotemporal controlled manner in living native cells. Here the tremendous drug discovery efforts that have

been devoted to the development of selective CB2R agonists have delivered a rich foundation of synthetic

drug-like ligands suitable for conversion into fluorescent probes. This presentation will illustrate successful

reverse design strategies for development of a toolbox of potent and efficacious drug derived CB2R-selective

fluorescent probes as well as their universal applicability in imaging and mechanism of action studies as well as

for the radiolabel free generation of binding affinity and kinetic data.

References

1) T. Gazzi, B. Brennecke, U. Grether, M. Nazare et al. ChemRxiv (2019) 1-14; DOI: 10.26434/chemrxiv.10283027.v1

EFMC-ISMC | 195

Posters - Chemical Biology E. Photochemistry in Drug Discovery: Photopharmacology,

Phototoxicity and Synthesis

196 | EFMC-ISMC

E001

MODIFICATIONS OF QUINOLINE PHOTOCAGES

Dóra Csorba (1), Tekla Abonyi (1), Dávid Szabó (1), Attila Csomos (2), Zoltán Mucsi (2), Petra Dunkel (1)

1) Semmelweis University, Department of Organic Chemistry1092 Budapest, Hőgyes Endre utca 7, Hungary2) Femtonics Limited, Chemistry Department

1094 Budapest, Tűzoltó utca 59, Hungary

Since the first examples in the 1970s, photocages became a routine experimental tool for studying dynamic

biological processes (1,2) and recently there is a growing number of studies on diverse prodrug applications

(3,4). Photocages act as protecting groups for masking temporarily the bioactivity of substrates linked via a

covalent bond, the cleavage of which with light irradiaton unleashes the activity in a spatiotemporally controlled

manner.

Quinoline caging groups were first described in 2002 by Dore and coworkers (5). Since then several structural

modifications to improve the properties and responsiveness of quinoline photocages were studied, as well as

various experimental applications thereof. On the one hand, type and position of ring substituents were found to

affect significantly the efficiency of the photolysis (6,7), on the other hand more complex dimer/trimer

constructs were designed and tested (8). Of note, quinoline photocages are efficient for two-photon activation,

exploiting (NIR) wavelengths more appropriate for addressing living systems (vs the UV operation range of

several conventional probes).

 

In the present work first we prepared reference quinoline photocages - BHQ, 7-DMAQ, 8-DMAQ, 3’-PyHQ –

according to the described protocols for comparison with our novel probes under the same experimental

conditions. Next, starting from the known 8-DMAQ, we studied modifications of the 8-amino substituent

(introducing cyclic aliphatic groups, substituted piperazines, benzofused heterocycles) to address on the one

hand the effect on the photophysical-photochemical properties and to provide on the other hand photoactivatable

scaffolds for the future design of more complex delivery systems. Of the novel set of photocages thus prepared

and characterised (18 new photocages, acetic acid used as model substrate), several derivatives showed similar

or better uncaging than the reference compounds.

Acknowledgements: The research was funded by the National Research, Development and Innovation Office(NKFIH), grant no. SNN 135825 and by the ÚNKP-20-5 New National Excellence Program of the Ministry ofInnovation and Technology. P.D. is recipient of the János Bolyai Research Scholarship of the HungarianAcademy of Sciences.

References

1) Chem. Rev. 2013, 113, 119-191

2) Nat. Methods 2007, 4, 619-628

3) J. Control. Release 2019, 298, 154-176

4) Curr. Med. Chem. 2017, 24, 4905-4950

5) Org. Lett. 2002, 4, 3419-3422

6) J. Org. Chem. 2020, 85, 726-744

7) Org. Lett. 2012, 14, 6366-6369

8) Chem. Eur. J. 2017, 23, 1860-1869

EFMC-ISMC | 197

E002

PHOTOSWITCHING MOLECULES FOR THE SPATIOTEMPORAL

TARGETING OF CANCER STEM CELLS WITH LIGHT

Laia Josa-Culleré, Amadeu Llebaria

Institute of Advanced Chemistry of Catalonia (IQAC-CSIC), Jordi Girona 18-26, 08034, Barcelona, Spain

Cancer treatment still represents a major challenge, with continuous increase in incidence, morbidity and relapse.

It is now accepted that resistance to conventional chemotherapy is often caused by a small population of cells

that can self-renew and differentiate into the cells that constitute the bulk of the tumour, designated cancer stem

cells (CSCs).

1

CSCs can adopt a quiescent state, which is not affected by standard anti-proliferative

chemotherapy. Since the CSC concept was first demonstrated,

2

the field of CSCs has seen an enormous advance,

and to date CSCs have been identified, isolated and characterised from various human cancers. However,

paradoxically approaches to avoid tumour relapse caused by CSCs are scarce.

The lack of selectivity of small molecule drugs, which often leads to side effects, is caused by the inability to

control their biological activity in time and space. This remains a major issue in the care of cancer patients

amongst others. Photopharmacology aims to use light as an external non-invasive element to control drug

activity, allowing for the activation of drugs with high spatial and temporal precision.

3

The design of

light-responsive molecules is based on photochromism, where a light-regulated moiety is incorporated into a

drug, leading to changes in polarity, geometry, and end-to- end distance upon isomerisation. These changes can

be designed to alter its biological activity.

Given the similarity of CSCs to normal stem cells, targeting only the former is a big challenge. However, doing

so is essential for a treatment that is well-tolerated. We believe that this challenge can be overcome by designing 

drugs that can be activated only in the tumour area by external light.

 

We will present our first proof of concept results. We have synthesised histone deacetylase inhibitors (HDACis)

that change their conformation under illumination with different wavelengths. We have photocharacterised them

to determine the optimal switching conditions and half-time, and tested them in enzymatic and cellular assays.

We have optimised them to be activable under visible light and to increase the activity difference between dark

vs light conformations. These constitute the first reported examples of photoactivable molecules targeted to

eliminate CSCs with external spatiotemporal control.

References

1) Batlle, E.; Clevers, H. Cancer Stem Cells Revisited. Nat. Med. 2017, 23 (10), 1124–1134

2) Lapidot, T.; Sirard, C.; Vormoor, J.; Murdoch, B.; Hoang, T.; Caceres-Cortes, J.; Minden, M.; Paterson, B.; Caligiuri, M.

A.; Dick, J. E. A Cell Initiating Human Acute Myeloid Leukaemia after Transplantation into SCID Mice. Nature 1994, 367

(6464), 645–648.

3) Hüll, K.; Morstein, J.; Trauner, D. In Vivo Photopharmacology. Chem. Rev. 2018, 118 (21), 10710–10747.

198 | EFMC-ISMC

E003

RATIONAL DESIGN OF LIGHT-CONTROLLED BIOACTIVE

COMPOUNDS FOR PHOTOPHARMACOLOGY

Piermichele Kobauri (1), Fangyuan Cao (2), Sebastian Thallmair (3,4), Siewert J. Marrink (3), Martin D.

Witte (5), Frank J. Dekker (2), Wiktor Szymanski (6), Ben L. Feringa (1)

1) Stratingh Institute for Chemistry, University of Groningen, Nijenborgh 4, 9747 AG, Groningen, The Netherlands2) Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, A.

Deusinglaan 1, 9713 AV Groningen, The Netherlands3) Groningen Biomolecular Sciences and Biotechnology Institute & Zernike Institute for Advanced Materials, University of

Groningen, Nijenborgh 7, 9747 AG Groningen, The Netherlands4) Frankfurt Institute for Advanced Studies, Ruth-Moufang-Straße 1, 60438 Frankfurt am Main, Germany

5) Chemical Biology II, Stratingh Institute for Chemistry, University of Groningen, 9747AG Groningen, The Netherlands6) Medical Imaging Center, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ,

Groningen, The Netherlands

Photopharmacology employs light to control the bioactivity of drugs with exceptional bio-orthogonality and

spatiotemporal precision, thus potentially increasing site- and organ-selectivity in pharmaceutical applications.

1–3

The incorporation of molecular photoswitches (such as azobenzenes) into drugs enables this non-invasive

control, since their irradiation induces reversible changes in the structure and properties of the drug.

2

In most

applications, photopharmacology aims to design photoswitchable drugs that are more active in the metastable

state, i.e. “cis-on”

4

in the case of classical azobenzenes. In particular, if this property is combined with a large

difference in potency between the photoisomers, it ensures effective control of biological processes.

5

Here we present the description of general criteria for the rational design of cis-on photoswitchable drugs. To

explore the requirements for molecular similarity with cis-azobenzene, we have analyzed the geometrical and

electrostatic properties of two-atom-linked biaryl systems. The biaryl sulfonamide motif was selected as an

example of this approach because of its bent geometry and favorable dipole moment, as well as its high

incidence in medicinal chemistry. Azologization of cisoid substructures may provide a rich source of inspiration

for photopharmacology and has the potential to guide the rational design of light-controlled bioactive

compounds.

References

1) W. A. Velema, W. Szymanski and B. L. Feringa, J. Am. Chem. Soc., 2014, 136, 2178–2191

2) M. M. Lerch, M. J. Hansen, G. M. van Dam, W. Szymanski and B. L. Feringa, Angew. Chem., Int. Ed., 2016, 55,

10978–10999

3) J. Broichhagen, J. A. Frank and D. Trauner, Acc. Chem. Res., 2015, 48, 1947–1960

4) C. Matera, A. M. J. Gomila, N. Camarero, M. Libergoli, C. Soler and P. Gorostiza, J. Am. Chem. Soc., 2018, 140,

15764–15773

5) I. M. Welleman, M. W. H. Hoorens, B. L. Feringa, H. H. Boersma and W. Szymanski, Chem. Sci., 2020, 11, 11672–11691

EFMC-ISMC | 199

E004

PHYTOCHEMISTRY AND CELLULAR TOLERANCE OF THREE

ANTIDIARRHEA PLANTS OF GABON

Joanna Grace Ombouma (1), Alban G. Houngbeme (2), Pauline Deguenon (2,4,5), Raymonde Mboma (3),

Marcel RB. Houinato (4), Fernand A. Gbaguidi (5)

1) Département de Pharmacologie. Faculté de Pharmacie. Université des Sciences de la Santé, B.P 4009 Libreville-Gabon.2) Laboratoire de Pharmacognosie/Institut de Recherche et d’Expérimentation en Médecine et Pharmacopée Traditionnelles

(IREMPT)/Centre Béninois de la Recherche Scientifique et de l’Innovation (CBRSI) /UAC, 01 BP 06 Oganla Porto-Novo,Bénin.

3) Département de Biologie et d’Ecologie végétale. Institut de Recherche en Ecologie Tropicale (IRET), Gabon.4) Faculté des Sciences Agronomiques de l’Université d’Abomey-Calavi, 01 BP 526 Cotonou, Benin.

5) Laboratoire de Chimie Pharmaceutique Organique, Ecole de Pharmacie, Faculté des Sciences de la Sante, Universitéd'Abomey-Calavi, Campus du Champ de Foire, 01 BP 188, Cotonou, Benin.

In Gabon, the Mitsogho populations of Estuaire use the leaves of Psidium guineense Sw. (Myrtaceae) and the

bark of Ceiba pentandra (L.) Gaertn. (Malvaceae) and Coula edulis Baill. (Olacaceae) to treat childhood

diarrhea. This work aims to provide scientific evidence for their use by determining the secondary metabolites

contained in said plants and the tolerance of their extract to 9PS and 9KB cells (human nasopharyngeal

carcinoma)

[1]

,  A-549 cells of lung carcinoma and HT-29 cells of colon carcinoma

[2]

. The phytochemical

analysis carried out on the various powders revealed the presence of steroids, tannins (catechetical and gallic),

reducing compounds and flavonoids. It should also be noted that there are no toxic compounds such as

cyanogenic derivatives, quinonics and cardiotonic glycosides. These results were corroborated by thin layer

chromatography with a Toluene-ethyl acetate-formic acid (36-12-5; v/v/v) solvent system as the mobile phase.

The toxicity on the brine shrimp larvae of the aqueous decocts has shown that these plants are not toxic about the

acquired LC50 which are between 3.61 ± 0.06 and 4.71 ± 0.15 mg/mL. The non-toxic nature of these extracts,

revealed by this general toxicity test, justifies the metabolites identified during phytochemical analysis.

 

Keywords: Medicinal plants, Phytochemical screening, chromatography, toxicity, Gabon.

References

1) Pelka M, Distler W, Petschelt A and Dent J. A new screening test toxicity testing of dental materials. Journal of Dentistry,

2000; 28: 341-345.

2) Carballo JL, Hernandez-Inda ZL, Pérez P and Garcia-Gràvalos MD. A comparison between two brine shrimp assays to

detect in vitro cytotoxicity in marine natural products. BMC Biotechnology, 2002; 2:17.

200 | EFMC-ISMC

E005

ONE FOR ALL? TOWARDS THE DEVELOPMENT OF POLYAMINE

BASED- PHOTOTHERANOSTIC CONJUGATES FOR CANCER

APPLICATIONS

Uliassi Elisa (1), Rossi Michele (1), Di Giosia Matteo (2), Calvaresi Matteo (2), Conti Amalia (3), Rhoden

Kerry J. (3), Bolognesi Maria Laura (1)

1) Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Via Belmeloro 6, 40126,Bologna, Italy

2) Department of Chemistry "Giacomo Ciamician," Alma mater Studiorum-Università di Bologna, Via Selmi 2, 40126,Bologna, Italy

3) Department of Medical and Surgical Sciences, Medical Genetics Unit, University of Bologna School of Medicine, ViaMassarenti 9, 40138 Bologna, Italy

Despite decades of intensive drug discovery research, cancer still ranks as the second-leading cause of mortality

globally and stands as an important barrier to increasing life expectancy [1]. In the search of innovative and

effective approaches, photodynamic therapy (PDT) has emerged as a promising option [2]. Being a

light-activated therapy, PDT allows precise controllability, minimal invasiveness, and high spatiotemporal

accuracy, when compared to conventional chemotherapy [2]. In fact, the core structural element of PDT, i.e., the

photosensitizer (PS), only works after it has been activated by light irradiation. The subsequent photon energy

transfer to surrounding oxygen molecules produces reactive oxygen species (ROS), which kill tumor cells. In

addition, as PS are generally fluorescent, PDT offers a unique advantage of developing molecules acting

simultaneously as both therapeutic and diagnostic agents (i.e., theranostics) [3]. As a further step toward a

fine-tuned delivery, the conjugation of a PS with cancer-targeting moieties, might allow a targeted delivery

selectively in cancer cells. Our idea is based on the knowledge that the polyamine transport system is

hyperactivated in certain cancers and that polyamines may act as carrier molecules to selectively guide the PS to

malignant cells [4]. Herein, we report on a small library of polyamine-based photodynamic conjugates for

selectively targeting cancer cells. The library has been designed by combining natural and synthetic polyamines,

which differ in chain length and number of nitrogen atoms within the carbon chains, to a reported naphthalimide

PS dye [3]. The presence of the naphthalimide moiety will allow selective fluorescence imaging and

photodynamic therapy (upon irradiation), while the presence of the polyamine tail will enable selective delivery.

The synthesized conjugates have been first characterized for their photophysical and photochemical properties.

Their uptake has been investigated in normal (MCF-10A) and cancer (MCF-7 and MDA-MB-231) cells by

fluorescence imaging studies, giving promising results for longer chain derivatives. Transport and competition

studies with natural polyamines have been performed. Furthermore, the conjugates display negligible to low

cytotoxicity in the dark in MCF-7 cells (up to 10 μM). In parallel, in vitro photodynamic cytotoxic activities are

being assessed.

Collectively, photodynamic conjugates consisting of a naphthalimide photosensitizer and a polyamine carrier

might represent innovative single molecules combining (i) targeting, (ii) diagnosis, and (iii) therapy.

References

1) Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, Bray F. CA Cancer J Clin 2021, 1-41.

2) Simões JCS, Sarpaki S, Papadimitroulas P, Therrien B, Loudos G. J Med Chem 2020, 63, 14119-150.

3) Zhang L, Lei K, Zhang J, Song W, Zheng Y, Tan S, Gao Y, Xu Y, Liu J, Qian X. MedChemComm 2016, 7, 1171-5.

4) Kaur N, Delcros JG, Imran J, Khaled A, Chehtane M, Tschammer N, Martin B, Phanstiel O4. J Med Chem 2008, 51,

1393-401.

EFMC-ISMC | 201

H. Application of Artificial Intelligence in Drug Discovery Projects

Posters - Technologies

202 | EFMC-ISMC

H001

BEYOND BINARY: SCALED MOLECULAR FINGERPRINTS FOR

MAXIMUM DIVERSITY PICKING

Vladimir Berishvili, Alexei Pushechnikov

ChemDiv, 12760 High Bluff Drive, Ste. 370 San Diego, CA 92130, USA.

The task of choosing the most diverse subset of chemical compounds is often encountered in the practice of a

medical chemist. This task occurs most often while conducting virtual high-throughput screening for selecting

compounds to probe a novel biological target. Given a limited budget, selecting a diverse subset of compounds

from a larger library can maximize the chemical space coverage, thereby improving chances of finding good

starting points for a discovery program.

The diversity picking involves three key steps: (1) the choice of molecular representation, (2) the choice of either

similarity or distance metric, and (3) the choice of an algorithm that leads to a global (or local) optimum for a

given metric of diversity. We focus in this report on the first step.

A widely used combination of binary fingerprints (e.g. ECFP, MACCS [1,2]) with either Tversky [3] or cosine

similarities advocates for an absolute equality of all bits (molecular features) in a representation. Despite the

simplicity of this approach, it is probably not the most efficient one for a task of a chemical diversity selection,

given the huge differences in the frequencies of different features (compare, for example, the chances of finding

a carbon atom versus phosphorus in a randomly picked organic compound).

Using term frequency normalization (akin to the popular natural language processing algorithm TF-IDF), one

could obtain not only the representation of a molecular structure as such, but also the representation of a specific

molecular structure in the context of a chemical compound library. Thus, the resulting floating-point vector

encodes the information about relative frequencies of various molecular features across an entire library. We

have shown that using this approach one can get the significantly more meaningful results compared to results

obtained via standard fingerprints. Additionally, using popular fingerprints benchmarks [4], we have

demonstrated that the proposed normalization can be a quite useful addition to a virtual screening workflow.

References

1) Rogers D, Hahn M. Extended-connectivity fingerprints. J Chem Inf Model., 50 (5): 742-754, 2010.

2) Joseph L Durant, Burton A Leland, Douglas R Henry, and James G Nourse. Reoptimization of MDL keys for use in drug

discovery. Journal of chemical information and computer sciences, 42:1273–1280, 2002.

3) Tversky A. Features of Similarity. Psychological Review, 84 (4): 327–352, 1977.

4) Riniker S, Landrum GA. Open-source platform to benchmark fingerprints for ligand-based virtual screening. J

Cheminform 5, 26, 2013.

EFMC-ISMC | 203

H002

FRAGMENT GROWING WITH REACTION PREDICTION FOR HIT

DISCOVERY

Hamza Tajmouati (1), Anna Kriukova (2), Christopher Housseman (3), Quentin Perron (4)

1) [email protected]) [email protected]

3) [email protected]) [email protected]

Molecular generation using AI approaches have gained wide recognition, thanks to the availability of numerous

approaches (LSTM, VAE, GAN…).

1

Unfortunately those algorithms suffer from producing complex to

unfeasible molecules in terms of synthetic feasibility.

2

Recently new AI approaches involving chemical reactions

have been described.

3

Mixing an initial molecular structure with commercial starting materials in the context of a

reaction is a natural way to define a policy for generating new compounds. This method ensures synthetic

accessibility of the generated molecules as the synthetic scheme is inherently obtained during the design process.

In this work we built a library of molecules starting from a defined fragment possessing two exit vectors where

commercial starting materials can react with.  The reaction prediction was performed with a template-based

neural network coupled with an applicability domain estimator. The goal was to find hits for the PIM-1 protein,

4

an isozyme of PIM kinase found in many cancers which inhibition is a promising approach to stop cell-growing

and reproduction.

The library was generated under the constraints of drug likeness metrics and structure-based scoring. The best

scoring compounds were profiled by medicinal chemist and some of them were found similar to known PIM-1

inhibitors. According to our knowledge it is the first report that uses a generative AI incorporating synthetic

feasibility by design, under structure-based constraints with a single fragment as starting point. This clearly

demonstrates the tremendous potential of such approach to easily generate valuable new starting points in the

context of a hit discovery program.

References

1) Nathan Brown, Artificial Intelligence in Drug Discovery - Drug Discovery - The Royal Society of Chemistry 2021

2) The Synthesizability of Molecules Proposed by Generative Models; Wenhao Gao and Connor W. Coley; J. Chem. Inf.

Model. 2020, 60, 12, 5714–5723

3) (a) Sai Krishna Gottipati, Boris Sattarov, Sufeng Niu, Yashaswi Pathak, Haoran Wei, Shengchao Liu, Simon Blackburn,

Karam Thomas, Connor Coley, Jian Tang, et al. Learning to navigate the synthetically accessible chemical space using

reinforcement learning. In International Conference on Machine Learning, pages 3668–3679. PMLR, 2020. (b) Bradshaw J,

Paige B, Kusner MJ, Segler, MH, Hernández-Lobato JM (2020) Barking up the right tree: an approach to search over

molecule synthesis dags. NeurIPS 2020 workshop on machine learning for molecules

4) Tursynbay, Y.; Zhang, J.; Li, Z.; Tokay, T.; Zhumadilov, Z.; Wu, D.; Xie, Y. PIM-1 kinase as cancer drug target: An

update. Biomed. Rep. 2016, 4, 140–146.

204 | EFMC-ISMC

H003

APPLICATION OF ARTIFICIAL INTELLIGENCE ON DRUG

DISCOVERY PROJECT: AN EVOTEC INSIGHT

Ferruccio Palazzesi (1), Mike Brunavs (2), Nathaniel Monck (2), Richard Rathmell (2), Adam Davenport

(2), Dimitar Hristozov (2), Alfonso Pozzan (1)

1) Computational Drug Discovery, Aptuit, an Evotec Company, Via A. Fleming 4, 37135 Verona, Italy2) Evotec (UK) Ltd. 114 Innovation Drive, Milton Park, Abingdon, Oxfordshire OX14 4RZ, UK

In recent years, the use of Artificial Intelligence (AI) approaches into drug discovery projects became a very

powerful tool. 

AI methods can play an important role in many different aspects of the design-make-test-analyse cycle.

1) 

Ligand-

and structure-based deep generative methods can be used to design novel compounds. Quantitative

structure-activity/property relationship AI models can be applied to help compound prioritization or to guide denovo generation of compounds towards a desirable region of chemical space. AI approaches can also aid the

bench chemists, predicting and optimizing chemical reactions, and identifying novel retrosynthetic routes.  

However, from a recent analysis, we found that few practical examples reported in the literature about the

application of these approaches, and in particular de novo drug design, to real life drug discovery projects.

2) 

For

this reason, in this poster presentation we decided to describe the application of AI approaches to one such

project.

In particular, we demonstrate the use of AI for designing potent and relevant compounds considering only

information available in the literature.

Constrained by specific project needs, many of the AI-suggested compounds remain structurally similar to

compounds present in the dataset used for training. However, these compounds were not obtained by either

human knowledge-based design or by extensive brute-force virtual enumeration. Instead, they emerged from an

efficient de novo structure generation procedure optimised for the exploration of desirable physical property

space, followed by an accurate analysis of the results by drug designers.

By sharing our experience, we hope to shed new light on the application of AI approaches on drug discovery

problems. In particular, we believe this work will help scientists to have a better understanding on how AI

methods work, and the outcomes we can expect from the application of these advanced technologies.

 

References

1) Schneider, Petra, et al. “Rethinking drug design in the artificial intelligence era.” Nature Reviews Drug Discovery 19.5

(2020): 353-364

2) Palazzesi, Ferruccio and Pozzan, Alfonso “Deep learning applied to ligand-based de novo drug design” Artificial

Intelligence in Drug Design, Humana, New York, NY, 2021 in press

EFMC-ISMC | 205

Posters - TechnologiesI. Biocatalysis & Late Stage Functionalisation

206 | EFMC-ISMC

I001

3-BROMOTETRAZINE: LABELLING OF MACROMOLECULES VIA

MONOSUBSTITUTED BIFUNCTIONAL S-TETRAZINES

Simon D. Schnell, Lukas V. Hoff, Advaita Panchagnula, Simon Sieber, Anthony Linden, Karl Gademann

Department of Chemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland

In recent years, the inverse electron demand Diels-Alder (IEDDA) reaction gained interest in the field of

chemical biology, due to its bioorthogonality, fast kinetics and biocompatibility.

[1]

Especially s-tetrazines

emerged as powerful precursors for IEDDA reactions with strained alkenes and alkynes.

[1]

However, it still

remains a challenge to incorporate s-tetrazines into macromolecules and current techniques mainly rely on amide

bond formation of a carboxylic acid derived tetrazine based precursor and an amine.

[2]

Not only the site of

labelling is thus strictly limited to amines, but the presence of a the bulky linker usually has a negative effect on

the physiochemical properties and the three-dimensional structure of the moiety attached to the s-tetrazine.

[3]

 

 

 

Herein, we report the synthesis of 3-halogenated, monosubstituted s-tetrazines and demonstrate their potential as

precursors for the installation of a minimal tetrazine unit selectively into natural products and proteins.

[4]

Additionally, chemoselective probes are synthesized for the labelling of thiols in cell extracts in the context of

targeted natural product isolation and in living cells.

[4]

References

1) B. L. Oliveira, Z. Guo, G. J. L. Bernardes, Chem. Soc. Rev. 2017, 46, 4895–4950.

2) W. D. Lambert, Y. Fang, S. Mahapatra, Z. Huang, C. W. am Ende, J. M. Fox, J. Am. Chem. Soc. 2019, 141,

17068–17074.

3) T. Cañeque, S. Müller, R. Rodriguez, Nat. Rev. Chem. 2018, 2, 202–215; M. Baalmann, M. J. Ziegler, P. Werther, J.

Wilhelm, R. Wombacher, Bioconj. Chem. 2019, 30, 1405–1414.

4) S. D. Schnell, L. V. Hoff, M. H. H. Wurzenberger, T. M. Klapötke, S. Sieber, A. Linden, K. Gademann, Chem. Sci. 2020,

11, 3042-3047.

ABSTRACT CANCELLED

EFMC-ISMC | 207

Posters - TechnologiesL. Innovative Synthesis for Medicinal Chemistry

208 | EFMC-ISMC

L001

DISCOVERY OF POTENT AND SELECTIVE A2A ANTAGONISTS

WITH EFFICACY IN ANIMAL MODELS OF PARKINSON'S DISEASE

AND DEPRESSION

Sujay Basu

Eurofins Advinus#21 & 22 Phase II, Peenya Industrial Area

Bangalore 560058, India

Adenosine A2A receptor (A2AAdoR) antagonism is a nondopaminergic approach to Parkinson’s Disease

treatment that is under development. We herein described a novel series of [1,2,4]triazolo[5,1-f]purin-2-one

derivatives that displays functional antagonism of the A2A receptor with a high degree of selectivity over A1,

A2B, and A3 receptors. Compounds from this new scaffold resulted in the discovery of highly potent, selective,

stable, and moderate brain penetrating compound 33. Compound 33 endowed satisfactorily in vitro and in vivo

pharmacokinetic properties. Compound 33 demonstrated robust oral efficacies in two commonly used models of

Parkinson’s Disease (haloperidol-induced catalepsy  and 6-OHDA lesioned rat models) and depression (TST and

FST mice models).

EFMC-ISMC | 209

L002

PYROGLUTAMIC ACID AS A NATURAL PLATFORM TOWARDS

NEW ANTIMICROBIAL AGENTS

Muriel BILLAMBOZ (1,2), Anca-Elena DASCALU (1,2), Antoine FAYEULLE (3), Benoît RIGO (1,2),

Alina GHINET (1,2)

1) Univ. Lille, Inserm, CHU Lille, Institut Pasteur Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculairesdes maladies liées au vieillissement, F-59000 Lille, France

Junia, Health and Environment, Laboratory of Sustainable Chemistry and Health, F-59000 Lille, France2) Junia, Health and Environment, Laboratory of Sustainable Chemistry and Health, F-59000 Lille, France

3) Université de technologie de Compiègne, ESCOM, TIMR, Centre de recherche Royallieu - CS 60 319 - 60 203 CompiègneCedex

The γ-lactam known as pyrrolidin-2-one is a heterocyclic moiety present in many natural and unnatural

compounds covering lots of biological activities. In the frame of green chemistry, the valorization of

pyroglutamic acid 1, a natural amino acid referred as the “forgotten amino acid”, sounds of greater interest for

proposing new γ-lactam derivatives. Pyroglutamic acid, byproduct from the sugar beet industry, issued from the

cyclization of glutamine and glutamic acid. This “forgotten” amino-acid has been studied since decades in our

team.

Based on an optimized solvent-free catalytic process,

1

series of N,N’-aminals, N,O, N,S-acetals and C

5

-substituted γ-lactams have been obtained and evaluated as potential antimicrobial agents against a panel of

fungal, bacterial and yeast strains. From this screening, biologically active compounds have been discovered and

early structure-activity relationships have been stated by variation of the linker.

2,3,4

These chemical

modifications had dramatic impacts on the potency and spectrum of antimicrobial activity. All these results are

paving the way to further development of tailored-made antimicrobials.

 

References

1) A.-E. Dascalu, A. Ghinet, E. Lipka, M. Collinet, B. Rigo, M. Billamboz. Cesium salts as superior catalysts for solvent-less

modifications of biosourced pterolactam. Mol. Cat. 2019, 470, 32-39.

2) A.-E. Dascalu, A. Ghinet, E. Lipka, C. Furman, B. Rigo, A. Fayeulle, M. Billamboz. Design, synthesis and evaluation of

hydrazine and acyl hydrazone derivatives of 5-pyrrolidin-2-one as antifungal agents. Bioorg. Med. Chem. Lett. 2020, 30(13),

127220.

3) A.-E. Dascalu, A. Ghinet, E. Lipka, C. Furman, B. Rigo, A. Fayeulle, M. Billamboz. Design, synthesis and antifungal

activity of pterolactam-inspired amide Mannich bases. Fitoterapia, 2020, 143, 104581.

4) M. Billamboz, A.E. Dascalu, A. Ghinet, R. Hartkoorn, E. Lipka-Belloli, C. Ple, B. Rigo. Hydrazide derivatives and their

specific use as antibacterial agents. PCT/EP2020/054392 (2020)

210 | EFMC-ISMC

L003

ONE-POT PROCEDURE FOR THE PREPARATION OF

1,5-DISUBSTITUTED 1,2,3-TRIAZOLES BY IN-SITU GENERATION

OF AZIDES.

Virgyl Camberlein (1), Nicolas Kraupner (1), Emmanuelle Lipka (2), Rebecca Deprez-Poulain (1), Benoit

Deprez (1), Damien Bosc (1)

1) Univ. Lille, Inserm, Institut Pasteur de Lille, U1177—Drugs and Molecules for Living Systems, Lille, France2) Univ. Lille, Inserm, RID-AGE U1167, F-59000, Lille, France

1,2,3-triazoles are broadly found in several domains such as materials science and medicinal chemistry. Whereas

the 1,4-disubstituted 1,2,3-triazoles are often encountered thanks to its easiness of access by CuAAC, their

1,5-regioisomers suffer from the lack of efficient methods for their synthesis. There is a crucial need to access to

this 1,5-disubstituted scaffold regarding its potential wide applications such as in drug discovery as disulfide

bond surrogate or cis-peptide bond mimetic.

Most of the methodologies developed to provide 1,5-disubstituted 1,2,3-triazoles require the handling of isolated

organic azides, which are not often commercially available and known to be unstable. Some azide-free

procedures were described but they are tied to limited substrate scope, and the request of specific conditions,

which have limited their use.

We developed and described in this poster a one-pot method, that converts functionalized bromides into azides,

which could then react in the same vessel with alkynes by NiAAC. This method has a broad scope and affords

1,5-disubstituted 1,2,3-triazoles with high regioselectivity.

EFMC-ISMC | 211

L004

NEW TRYPTANTHRIN-BASED PETASIS ADDUCTS - DESIGN,

SYNTHESIS AND BIOLOGICAL ACTIVITY EVALUATION

Pedro Brandão (1,2), Carolina Marques (2), Eugénia Pinto (3,4), Anthony J. Burke (2,5), Marta Pineiro

(1)

1) University of Coimbra, CQC, Department of Chemistry, 3004-535, Coimbra, Portugal2) LAQV-REQUIMTE, University of Évora, Rua Romão Ramalho, 59, 7000, Évora, Portugal

3) Laboratório de Microbiologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto,Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal

4) CIIMAR—Interdisciplinary Centre of Marine and Environmental Research, Terminal de Cruzeiros do Porto de Leixões,4450-208 Matosinhos, Portugal

5) University of Évora, Department of Chemistry, Rua Romão Ramalho, 59, 7000, Évora, Portugal

Tryptanthrin is a golden yellow alkaloid with great relevance in medicinal chemistry. This natural tetracyclic

compound, which can be synthesized through different reaction pathways, has shown the ability to interact with

multiple biological targets, being a suitable candidate for polypharmacological approaches. Among its multiple

biological activities, which includes anti-inflammatory, antioxidant and antiallergic, tryptanthrin and its

derivatives are also known by their activity against several microorganisms.[1] The increasing number of

microorganisms resistant to conventional drugs highlights the need to develop new drug candidates targeting

infectious diseases.[2]

In our group, the application of multicomponent reactions (MCRs) in drug discovery is gaining attention, as this

methodology is suitable to achieve structural diversity in a fast and efficient manner.[3] Therefore, engaging

tryptanthrin in a MCR approach emerged as a suitable plan to unlock the structural diversity of new tryptanthrin

derivatives. Literature research evidenced that, despite the great interest of this molecules in medicinal

chemistry, this approach has been neglected, with only one example of its application in MCRs reported,

consisting of a Knoevenagel-initiated MCR.[4] We decided to apply the Petasis MCR to obtain novel

tryptanthrin-derivatives. Reaction conditions (including solvent, temperature, catalyst) were optimized and the

best reaction conditions suitable to prepare a library (20 compounds) of druglike molecules bearing the

tryptanthrin tetracyclic moiety were obtained. An asymmetric version of the reaction was also found giving good

yields and high enantioselectivities. The prepared library was evaluated in silico in what concerns their

physical-chemical properties and druglikeness, using the SwissADME free web-based tool,[5] and it is currently

undergoing antifungal and antibacterial bioactivity evaluation.

Acknowledgements

Coimbra Chemistry Centre (CQC) is supported by the Portuguese Agency for Scientific Research, “Fundação

para a Ciência e a Tecnologia” (FCT) through project UIDB/00313/2020 and UIDP/00313/2020, co-funded by

COMPETE2020-UE. NMR data was collected at the UC-NMR facility which is supported in part by FEDER –

European Regional Development Fund through the COMPETE Programme (Operational Programme for

Competitiveness) and by National Funds through FCT through grants REEQ/481/QUI/2006,

RECI/QEQ-QFI/0168/2012, CENTRO-07-CT62-FEDER-002012, and Rede Nacional de Ressonância

Magnética Nuclear (RNRMN). This work also received financial support from FCT/MCTES through the project

UIDB/50006/2020. Pedro Brandão acknowledges financial support from PhD fellowship (PD/BD/128490/2017)

of the Programa Doutoral FCT “Catalysis and Sustainability” CATSUS (PD/00248/2012).

References

1) a) A. M. Tucker et al., Arkivoc, 2012, 546-569. b) R. Kaur et al., Bioorg. Med. Chem., 2017, 25, 4533-4552.

2) a) J. R. Perfect, Exp. Op. Emerg. Drugs, 2016, 21 (2), 129-131. b) E. D. Brown et al., Nature, 2016, 529, 336-343.

3) a) P. Brandão et al., Eur. J. Med. Chem., 2021, 211, 113102. b) P. Brandão et al., Chem. Rec., 2021, in press.

4) M. Beyrati et al., Tetrahedron Lett., 2017, 58, 1947-1951.

5) A. Daina et al., Sci. Rep., 2017, 7.

212 | EFMC-ISMC

L005

DESIGN AND SYNTHESIS OF LINKERS FOR GLYCAN-COATED

GOLD NANOPARTICLES USED IN DRUG DISCOVERY

Begona Pampín Casal, Julie Masse, Jacobo Cruces Colado, Monica Carreira

GalChimia S.A., Parque Empresarial, 15822, Fonte Díaz, Touro, A Coruna, Spain

The great appeal of nanopharmaceuticals lies in their ability to exploit the unique properties of nanoscale

materials to address some of the most challenging problems of medical therapy and diagnosis.

Glycan-coated gold nanoparticles (GNPs) represent the latest generation of nanomedicines. This technology is

designed to control the biodistribution and pharmacokinetic properties, thus allowing for improved therapeutic

indexes of medicines.

The union of a metallic core with active molecules, which can be either biological or chemical, is required to

obtain a functional GNP. This union is made through a chemical linker. The choice of linker(s) and their

distribution will have several impacts on the final medicine:

- Stabilizing the metallic core and making the particle water-soluble and biocompatible.

- Establishing the union between the nanoparticle and the active product.

- Controlling the release of the active product.

In this work, different linkers based on carbohydrates or glycans were designed, synthesized, and optimized for

attachment to the gold core of GNPs via gold–sulfur bonds.

References

1) Golding, Jon; Williams, Phillip; Roskamp, Meike; Nanoparticles and Their Use in Medicine, PCT Int. Appl. 2016, WO

2016/102613 A1.

2) McAteer, Martina; Dayan, Professor Colin Mark; Birchall, Professor James Caradoc; Nanoparticles-based antigen specific

immunotherapy. PCT Int. Appl. 2016, WO 2016/162495 A1.

EFMC-ISMC | 213

L006

SYNTHESIS AND BIOLOGICAL ACTIVITY OF

TRITHIOLATO-BRIDGED DINUCLEAR RUTHENIUM(II)-ARENE

COMPLEXES WITH NUCLEIC BASES

Oksana Desiatkina (1), Martin Mösching (1), Emilia Păunescu (1), Julien Furrer (1), Nicoleta Anghel (2),

Ghalia Boubaker (2), Yosra Amdouni (2), Andrew Hemphill (2)

1) Department of Chemistry, Biochemistry and Pharmaceutical sciences, University of Bern, Freiestrasse 3, 3012 Bern,Switzerland

2) Institute of Parasitology, Vetsuisse Faculty, University of Bern, Länggass-Strasse 122, 3012 Bern, Switzerland

Symmetric and mixed cationic trithiolato dinuclear ruthenium(II)-arene complexes (general formula [(η6

-arene)

2

Ru

2

(μ2

-SR)

3

]

+

and, respectively, [(η6

-arene)

2

Ru

2

(μ2

-SR

1

)

2

(μ2

-SR

2

)]

+

) have shown high in vitro activity against

the apicomplexan parasites Toxoplasma gondii1 and Neospora caninum2

which encouraged the development of

new compounds based on this scaffold.

Attaching a bioactive compound (metabolite, drug) to a ruthenium(II)-arene moiety is an approach used to

modulate the medicinal properties of organometallic complexes

3,4

. The resulting hybrid molecules can be

directed towards specific sub-cellular targets, and can present new modes of action. T. gondii cannot synthesize

de novo purines and must uptake these metabolites from the host cells

5

. Conjugating nucleic bases to trithiolato

diruthenium units could be a promising strategy which could favor the uptake of the metalorganic complexes

into the parasite.

A new functionalization strategy based of copper(I)-catalyzed azide-alkyne 1,3-cycloaddition (CuAAC) was

successfully implemented for obtaining the conjugates. 14 novel 'hybrids' nucleic base-diruthenium complex

were synthesized and fully characterized. A first antiparasitic activity screening on T. gondii grown in human

foreskin fibroblasts (HFFs) was performed by applying 1 and 0.1 µM of each compound of interest

(intermediates and complexes). 9 compounds were further submitted to IC

50

determination and shown inhibition

of T. gondii proliferation in the 0.03-0.66 µM range.

References

1) J. Furrer, G. Süss-Fink, Coord. Chem. Rev. 2016, 309, 36-50.

2) T. Mede, M. Jäger, U. S. Schubert, Chem. Soc. Rev. 2018, 47, 7577-7627.

3) E. Ratzon, Y. Najajreh, R. Salem, H. Khamaisie, M. Ruthardt, J. Mahajna, BMC Cancer 2016, 16, 140.

4) S. Savino, C. Marzano, V. Gandin, J. D. Hoeschele, G. Natile, N. Margiotta, Int. J. Mol. Sci. 2018, 19, 2050.

5) E. C. Krug, J. J. Marr, R. L. Berens, J. Biol. Chem. 1989, 264, 18, 10601-10607.

214 | EFMC-ISMC

L007

DESIGN AND SYNTHESIS OF RED-SMU1 INTERACTION

DISRUPTORS AS NEW ANTIVIRAL AGENTS AGAINST INFLUENZA

A VIRUS

Alessandra Corio (1), Laurent Le Corre (1), Usama Ashraf (2), Pierre-Olivier Vidalain (3), Nicolas

Pietrancosta (4), Thibaut Crepin (5), Nadia Naffakh (2), Christine Gravier-Pelletier (1), Patricia Busca (1)

1) Université de Paris, LCBPT, UMR 8601CNRS, 75006 Paris, France2) Institut Paster, Biologie des ARN et Virus Influenza, 75015 Paris, France

3) Ecole normale supériore, Centre International de Recherche en Infectiologie- inserm, Lyon, France4) Institut de Biologie Paris Seine, UMR8246 Neurosciences, 75252 Paris, France

5) Institut de Biologie Structurale, Groupe Machines de Réplication Virale, 38000 Grenoble, France

Influenza virus A (IVA) represent a major global health threat. Despite the existence of a seasonal vaccine and

the availability of antiviral drugs, each year IVA causes between 3 and 5 million of severe cases and the death of

more than 300 thousand people in the world. Moreover, due to the rapid antigenic variation of the virus, the

available drugs become inefficient and the vaccine needs to be reformulated each year. Consequently, there is an

urging need for the development of innovative antiviral agents.

1

During the last years, Naffakh’s team and collaborators demonstrated that the complex of Red and SMU1, which

are human spliceosomal factors, is necessary for the replication cycle of the IVA. In this way Red-SMU1 can be

considered as a promising target to fight against Influenza A.

2

Small molecules acting as Red-SMU1 disruptors were shown to inhibit viral replication without targeting viral

components: this new approach belongs to the so-called “host directed therapies” and virtually eliminates the

risk of drug-resistance development.

3

As preliminary results, a hit compound (ALG61) was identified and validated through i) the in silico screening

of more than 3000 compounds from our in-house chemolibrary, ii) the biological evaluations in vitro as well as

in cellulo of the best candidates and iii) the co-crystallization with SMU1. ALG61, a small molecule that belongs

to the pyrido[2,3-d]pyrimidines family, was found to inhibit both the RED-SMU1 interaction (EC

50

= 15 μM) 

and the viral replication.

4

With the objective of improving the biological activity of the hit and its selectivity toward the Red-SMU1

interaction, we designed series of ALG61 analogues, with different central scaffolds or substitution patterns.

 

The recent results regarding their synthesis and biological activity will be presented in this communication.

References

1) F. Krammer, G.J.D. Smith, R.A.M. Fouchier, M. Peiris, K. Kedzierska, P.C. Doherty, P. Palese, M.L. Shaw, J. Treanor,

R.G. Webster, A. García-Sastre, Influenza, Nature Reviews Disease Primers. 4 (2018) 3.

2) G. Fournier, C. Chiang, S. Munier, A. Tomoiu, C. Demeret, P.-O. Vidalain, Y. Jacob, N. Naffakh, Recruitment of

RED-SMU1 Complex by Influenza A Virus RNA Polymerase to Control Viral mRNA Splicing, PLoS Pathogens. 10 (2014)

e1004164.

3) S.H.E. Kaufmann, A. Dorhoi, R.S. Hotchkiss, R. Bartenschlager, Host-directed therapies for bacterial and viral infections,

Nature Reviews Drug Discovery. 17 (2018) 35–56.

4) U. Ashraf, L. Tengo, L. Le Corre, G. Fournier, P. Busca, A.A. McCarthy, M.-A. Rameix-Welti, C. Gravier-Pelletier,

R.W.H. Ruigrok, Y. Jacob, P.-O. Vidalain, N. Pietrancosta, T. Crépin, N. Naffakh, Destabilization of the human RED–SMU1

splicing complex as a basis for host-directed antiinfluenza strategy, Proceedings of the National Academy of Sciences. 116

(2019) 10968–10977.

EFMC-ISMC | 215

L008

AZIDE/ALKENE 1,3-DIPOLAR CYCLOADDITION FOR THE

SYNTESIS OF 1,4,5-TRISUBSTITUTED 1H-1,2,3-TRIAZOLES AS

POTENTIAL ANTIPROLIFERATIVE AGENTS

Mario De-Juan-Alberdi (1,2), Irene De Celis (1), Noelia Monja (1), Concepción Pérez-Melero (1,2,3)

1) Pharmaceutical Sciences Department, University of Salamanca. Campus Miguel de Unamuno. 37007 Salamanca, Spain.2) Institute of Biochemical Research of Salamanca (IBSAL). Paseo de San Vicente, 58-182. 37007 Salamanca, Spain.

3) Tropical Diseases Research Centre (CIETUS). School of Pharmacy, University of Salamanca. 37007 Salamanca, Spain.

Eg5 is a kinesin protein that participates only in mitosis (prophase and prometaphase) by producing spindle-pole

separation. It is essential in cell division, and its inhibition leads to cell cycle arrest and ultimately to cell death.

Therefore, Eg5 is a promising target for developing new antimitotic agents, which would prevent the

neurotoxicity caused by the anti-tubulin drugs currently in use

1

.

In this context, a series of 1H-1,2,3-triazoles trisubstituted at positions 1, 4 and 5 have been designed as Eg5

inhibitors by binding at the L5 allosteric site and thus as potential antiproliferative agents. In this work, the

synthesis of 1H-1,2,3-triazoles has been carried out by means of a 1,3-dipolar cycloaddition reaction between

benzylic azides and the double bond of a chalcone (Figure 1). The azides were prepared from commercial

hydroxybenzaldehydes in a four-step route. The chalcone was prepared by a Claisen-Schmidt condensation

between substituted benzaldehydes and a benzyl protected hydroxyacetophenone. The cycloaddition reactions

between the heterochalcone and benzyl-azides were carried out by using TBAHS as catalyst

2

under a sequential

inert and oxidizing atmosphere. In some cases, TEMPO was added as co-oxidant

3

.

Fig 1. Synthesis of 1,4,5-trisubstituted 1H-1,2,3-triazoles by 1,3-dipolar cycloaddition (60 % TEMPO is added in

step 1 when the benzyloxy group of the azide is at position 2).

Docking studies show that the designed 1H-1,2,3-triazoles fit into the L5 allosteric binding site of human Eg5.

 

 

 

Acknowlegdements: Financial support from Junta de Castilla y León (SA072G19) and “Memoria de D. Samuel

Solórzano Barruso” Foundation (FS/18-2020). MDJA thanks Universidad de Salamanca and FundaciónSalamanca de Cultura y Saberes for financial support.

References

1) Rath, O.; Kozielski, F. Kinesins and Cancer. Nature Rev. Cancer 2012, 12, 527-539.

2) Arya A.; Gupt, M. P.; Devender, N.;·Tripathi, R. P. Accessing a small library of pluripotent 1,4,5-trisubstituted

1H-1,2,3-triazoles via diversity-oriented synthesis Mol. Divers. 2012, 16, 335-350.

3) Gangaprasad, D.; Raj, J. P.; Kiranmye, T.; Karthikeyan, K.; Elangovan, J. Another example of organo-click reactions:

TEMPO-promoted oxidative azide-olefin cycloaddition for the synthesis of 1,2,3-triazoles in water. Eur. J. Org. Chem. 2016,

5642–5646.

216 | EFMC-ISMC

L009

SYNTHESIS OF DIARYL HYDROXYL DICARBOXYLIC ACIDS FROM

AMINO ACIDS

Aleksi Eronen, Jere Mannisto, Karina Moslova, Martin Nieger, Eeva Heliövaara, Timo Repo

Department of Chemistry, University of Helsinki, A. I. Virtasenaukio 1, 00014, Helsinki, Finland

In this presentation, we report a unique method for preparing diaryl hydroxyl dicarboxylic acids. The reaction

occurs in three-component system between amino acid, aromatic aldehyde and primary alcohol in alkaline

solutions under microwave-assisted conditions. The diacid products are composed in situ generated pyruvic acid

and cinnamaldehyde which react together via a [3,3]-sigmatropic rearrangement, followed by Na

+

-ion assisted

hydride transfer. The diacids form with up to four stereocenters in the diastereospecific manner. As further

shown, the diacids are easily turned into corresponding δ-lactones.

References

1) Eronen, A. E. K.; Mannisto, J. K.; Moslova, K; Nieger, M; Heliövaara, E; Repo, T. Synthesis of Diaryl Hydroxyl

Dicarboxylic Acids from Amino Acids. J. Org. Chem. 2020, 85, 5799-5806.

EFMC-ISMC | 217

L010

A NEW APPROACH TO THE SYNTHESIS OF ZOLPIDEM AND

FLUORINATED DERIVATIVES

Nikola Fajkis-Zajączkowska (1), Monika Marcinkowska (1), Beata Gryzło (1), Anna Krupa (2), Marcin

Kołaczkowski (1)

1) Department of Medicinal Chemistry, Jagiellonian University Medical College, Medyczna 9 Street, 30-688 Kraków,Poland.

2) Department of Pharmaceutical Technology and Biopharmaceutics, Jagiellonian University Medical College, Medyczna 9Street, 30-688 Kraków, Poland.

More than three decades ago, zolpidem was successfully introduced to the pharmaceutical market that is set to

induce sleep initiation. The great advantage of this is the fact that the inversion to benzodiazepines does not

result in a hangover the next day [1].

 

Literature reports indicate that zolpidem has a promising pharmacological response in clinical trials. It has been

proved that in post-stroke patients an improvement in motor and neurological functions was observed [2].

 

Therefore, there is a great need for large-scale synthesis of zolpidem with high purity for advanced biological

research. The synthetic pathways leading to zolpidem have already been described in literature. However,

the paths required the use of toxic reagents and large amounts of solvents [3].

 

We developed a zolpidem synthesis path which consisted of three steps and was carried out in microwave

reactors. This approach made it possible to obtain final compounds with high yields and purity in a very short

time and with a small amount of solvent.

 

Research financed by the National Science Center, project number 2018/30/E/NZ7/00247.

References

1) E. Sanna et al., European Journal of Pharmacology, 2002, 13, 451.

2) J. Sutton et al., Brain Injury, 2017, 8, 31.

3) W. Shucheng et al., Advances in Pharmacology, 2016, 68.

218 | EFMC-ISMC

L011

SYNTHESIS AND ANTIVIRAL ACTIVITY OF C-NUCLEOSIDE

ANALOGUES OF RIBAVIRIN

Simon Gonzalez (1), Abdelhakim Ouarti (1), Carmen Solarte (1), Gabriela Brzuska (2), Angélique Ferry

(1), Florian Gallier (1), Jacques Uziel (1), Ewelina Krol (2), Nadège Lubin-Germain (1)

1) CY Cergy-Paris Université, CNRS, BioCIS, Equipe de Chimie Biologique, Cergy, France2) Intercollegiate Faculty of Biotechnology

University of Gdańsk and Medical University of Gdańsk, Gdańsk, Poland

Ribavirin (RBV) is a well-known broad-spectrum antiviral and anticancer nucleosidic therapeutic agent. RBV

has been extensively studied for its benefic activity in various pathologies such as leukemias, lymphomas,

carcinomas, respiratory syncytial virus-related diseases, hemorrhagic fevers and hepatitis viruses.

1,2

Nonetheless, its use often comes with adverse effects, particularly anemia. In an attempt to improve both

stability and pharmacological profile, we developed a C-nucleoside analogue of RBV, named SRO-91, bearing a

1,2,3-triazole moiety in lieu of the native 1,2,4-triazole. SRO-91 was accessed by an indium-mediated

alkynylation of ribose creating the C-C nucleosidic linkage, and a Huisgen cycloaddition to form the triazole

ring, serving as a base analogue.

3

Interestingly, SRO-91 showed a potent anticancer activity against ovarian

cancer cells, melanomas, epideroid carcinomas and gliomas while being non-toxic against healthy cells.

4,5

In

parallel, we developed a pharmacomodulation approach on our C-nucleoside scaffold by introducing different

carboxamide groups (R

1

), hydroxyl protecting groups (R

2

and R

3

) and quaternarization at position 2 (R

4

). In

addition, we prepared conjugates in position 5 (R

5

) with galactosides as liver targeting agents or with previously

studied membranotropic peptides as cell-penetrating agents.

6

Finally, in order to study the potent broad-spectrum

activity of the newly synthesized RBV analogues, the antiviral activity in cell culture system was evaluated

against two important viral human pathogens - hepatitis C virus and Zika virus. The results clearly indicated that

SRO-91 analogues feature antiviral activity against hepatitis C virus as well as Zika virus. Additionally, some C-nucleoside analogues demonstrated strong to moderate inhibitory activity against both viruses.

References

1) Casaos, J.; Gorelick, N. L.; Huq, S.; Choi, J.; Xia, Y.; Serra, R.; Felder, R.; Lott, T.; Kast, R. E.; Suk, I.; Brem, H.; Tyler,

B.; Skuli, N. The Use of Ribavirin as an Anticancer Therapeutic: Will It Go Viral? Mol. Cancer Ther. 2019, 18 (7),

1185–1194.

2) Nyström, K.; Waldenström, J.; Tang, K.-W.; Lagging, M. Ribavirin: Pharmacology, Multiple Modes of Action and

Possible Future Perspectives. Future Virol. 2019, 14 (3), 153–160.

3) Solarte, C.; Dos Santos, M.; Gonzalez, S.; Miranda, L.; Guillot, R.; Ferry, A.; Gallier, F.; Uziel, J.; Lubin-Germain, N.

Synthesis of C-Ribosyl-1,2,3-Triazolyl Carboxamides. Synthesis 2017, 49 (09), 1993–2002.

4) Wambecke, A.; Laurent-Issartel, C.; Leroy-Dudal, J.; Giffard, F.; Cosson, F.; Lubin-Germain, N.; Uziel, J.; Kellouche, S.;

Carreiras, F. Evaluation of the Potential of a New Ribavirin Analog Impairing the Dissemination of Ovarian Cancer Cells.

PLOS ONE 2019, 14 (12), e0225860.

5) Sabat, N.; Migianu-Griffoni, E.; Tudela, T.; Lecouvey, M.; Kellouche, S.; Carreiras, F.; Gallier, F.; Uziel, J.;

Lubin-Germain, N. Synthesis and Antitumor Activities Investigation of a C-Nucleoside Analogue of Ribavirin. Eur. J. Med.

Chem. 2020, 188, 112009.

6) Gonzalez, S.; Gallier, F.; Kellouche, S.; Carreiras, F.; Novellino, E.; Carotenuto, A.; Chassaing, G.; Rovero, P.; Uziel, J.;

Lubin-Germain, N. Studies of Membranotropic and Fusogenic Activity of Two Putative HCV Fusion Peptides. Biochim.

Biophys. Acta BBA - Biomembr. 2019, 1861 (1), 50–61.

EFMC-ISMC | 219

L012

AN ORIGINAL PETASIS APPROACH TOWARDS THE SYNTHESIS

OF NOVEL MCL-1 INHIBITORS

Soufyan Jerhaoui, Frederik J. R. Rombouts, Gaston S. M. Diels, Michel Surkyn, Yves E. M. Van

Roosbroeck

Janssen Research and Development, Turnhoutseweg 30, 2340 Beerse (Belgium)

Myeloid Cell Leukemia 1 (Mcl-1) is a member of the Bcl-2 protein family, which are critical regulatory proteins

in the mitochondrial apoptotic pathway. It is overexpressed and amplified in various tumours such as myeloid

leukemia

1,2

 and promotes the survival of cancer cells by inhibiting the apoptosis process, thus making Mcl-1 an

attractive target in oncology.

3,4,5

We herein disclose the discovery of novel fluoroallylamine containing macrocycles, ideated via a structure-based

drug design approach, showing favourable physicochemicalproperties as well as a good level of inhibition of the

Mcl-1 enzyme.

6

The synthesis of these complex macrocycles was elucidated applying an original and diastereoselective Petasis

approach between fragments A, B and C,

7

followed by a one-pot palladium-catalysed CO insertion and

amidation reactions.

8

References

1) Kelly, G. L., Strasser, A. Annual Review of Cancer Biology, 2020, 4, pp. 299-313

2) Hird, A. W., Tron, A. E. Pharmacology & Therapeutics, 2019, 198, pp. 59-67

3) Brown, S. P., Hughes, P. E., et al. Cancer Discovery, 2018, 8, pp. 1582-1597

4) Hird, A. W., et al. Nat Commun., 2018, 9, pp. 5341-5355

5) Geneste, O., et al. Nature, 2016, 538, pp. 477-482

6) Rombouts, F. J. R., Diels, G. S. M., Jerhaoui, S., Surkyn, M., Van Roosbroeck, Y. E. M. WO 2021/005043 A1

7) Petasis, N. A., Akritopoulou, I. A. Tetrahedron Lett., 1993, 34, pp. 583–586

8) Van der Wildt, B., Shen, B., Chin, F. T. Chem. Commun., 2019, 55, pp. 3124-3127

220 | EFMC-ISMC

L013

NEW FIDAXOMICIN ANTIBIOTICS: COMBINING METABOLIC

ENGINEERING AND SEMISYNTHESIS

Erik Jung (1), Maja Remškar (2), Daniel Schäfle (3), Andrea Dorst (1), David Dailler (1), Peter Sander

(3,4), Rolf Müller (2), Karl Gademann (1)

1) Department of Chemistry, University of Zurich, Winterthurerstrasse 190, CH-8057 Zürich, Switzerland2) Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Department of Microbial Natural Products, HelmholtzCentre for Infection Research and Department of Pharmaceutical Biotechnology, Saarland University, Campus E8.1, 66123

Saarbrücken, Germany3) Institute of Medical Microbiology, University of Zurich, Gloriastrasse 28/30, 8006 Zurich, Switzerland4) National Center for Mycobacteria, University of Zurich, Gloriastrasse 28/30, 8006 Zurich, Switzerland

Fidaxomicin (tiacumicin B, lipiarmycin A3)

[1]

is a glycosylated macrocyclic antibiotic with potent activity

against various Gram-positive bacteria through inhibition of the RNA-polymerase (RNAP).

[2]

In 2015 our

research group accomplished the first total synthesis of this complex natural product.

[3,4]

Fidaxomicin is

marketed to treat Clostridium difficile infections in the gut. Moreover, its antibacterial activity against resistant

strains of Mycobacterium tuberculosis and Staphylococcus aureus is of great interest, as these strains still pose a

global problem.

[5,6]

However, treatment of these systemic infections with fidaxomicin is prevented by its low water solubility. The

central goal of this study was the synthesis of new fidaxomicin derivatives with reduced structural complexity

that retain or improve their antibiotic activities, while improving other parameters like solubility. Our

investigation pursued two strategies: the isolation of shunt metabolites from a mutant fidaxomicin producing

strain followed by their chemical modification and semisynthesis of new antibiotics by chemical modification of

the natural product itself. The antibiotic activity of derivatives was rationalised by modelling their binding to

bacterial RNAP.

References

1) W. Erb, J. Zhu, Nat Prod Rep 2013, 30, 161–174.

2) W. Lin, K. Das, D. Degen, A. Mazumder, D. Duchi, D. Wang, Y. W. Ebright, R. Y. Ebright, E. Sineva, M. Gigliotti, A.

Srivastava, S. Mandal, Y. Jiang, Y. Liu, R. Yin, Z. Zhang, E. T. Eng, D. Thomas, S. Donadio, H. Zhang, C. Zhang, A. N.

Kapanidis, R. H. Ebright, Mol. Cell 2018, 70, 60 71.e15 and references therein.

3) E. Kaufmann, H. Hattori, H. Miyatake-Ondozabal, K. Gademann, Org. Lett. 2015, 17, 3514–3517.

4) S. Norsikian, C. Tresse, M. François‐Eude, L. Jeanne‐Julien, G. Masson, V. Servajean, G. Genta‐Jouve, J.-M. Beau, E.

Roulland, Angew. Chem. Int. Ed. 2020, 59, 6612–6616.

5) World Health Organization. Global Tuberculosis Report 2019, Geneva: WHO; 2019.

6) European Centre for Disease Prevention and Control. Surveillance of antimicrobial resistance in Europe 2018, Stockholm:

ECDC; 2019.

EFMC-ISMC | 221

L014

SYNTHESIS OF THE OXA-BRIDGED TRICYCLE CORE STRUCTURE

OF DAPHNANE DITERPENOID ORTHOESTERS VIA PRINS-TYPE

CYCLIZATION REACTION

Ahmed Zakaria (1), Young Seub Kim (2), Sun-Joon Min (2)

1) Beni-Suef University, Egypt2) Hanyang University, Korea

The daphnane diterpenoid orthoesters (DDOs), generally isolated from the plants families of Thymelaeaceae and

Euphorbiaceae, have been demonstrated to possess a wide range of biological activities such as cytotoxic,

anti-HIV, antifertility, neurotrophic, analgesic, antihyperglycemic, and skin irritant activities

1

.

Because of their structural complexity and promising biological activities, the daphnane-family natural products

have become attractive synthetic targets. A number of elegant synthetic approaches toward the core ring

structures of this family have been reported

2

.

Despite these synthetic efforts, concise and highly efficient synthetic routes toward these complex DDOs are still

worthy. Herein, we describe a concise and amenable synthetic route to the 7,6-membered ring core structure.

The key step in this approach is the development of a stereocontrolled Prins-type cyclization reaction.

References

1) (a) Liao, S.-G.; Chen, H.-D.; Yue, J.-M. Chem. Rev. 2009, 109, 1092 (b) Adolf, W.; Sorg, B.; Hergenhahn, M.; Hecker, E.

J. Nat. Pro. 1982, 45, 347 (c) He, W.; Cik, M.; Appendino, G.; van Puyvelde, L.; Leyson, J. E.; De Kimpe, N. Mini-Rev.

Med. Chem. 2002, 2, 185.

2) Examples of synthetic approaches to DDOs (a) Li, Y.; Wei, M.; Dai, M. Tetrahedron 2017, 73, 4172. (b) Hassan, A. H. E.;

Lee, J. K.; Pae, A. N.; Min, S.-J.; Cho, Y. S. Org. Lett. 2015, 17, 2672. (c) Tong, G.; Liu, Z.; Li, P. Org. Lett. 2014, 16, 2288.

222 | EFMC-ISMC

L015

SUSTAINABLE SYNTHESIS OF NEW CHOLINE KINASE

INHIBITORS

Pilar María Luque Navarro (1,2), M. Paz Carrasco-Jiménez (3), Inmaculada Pérez-Prieto (3), Carmen

Marco (3), Daniela Lanari (1), Luisa Carlota López-Cara (2)

1) Department of Pharmaceutical Sciences, University of Perugia, Via del Liceo 1, 06123, Perugia, Italy2) Department of Pharmaceutical and Organic Chemistry, Faculty of Pharmacy, Campus de Cartuja s/n, 18071, Granada,

Spain3) Department of Biochemistry and Molecular Biology I, Faculty of Sciences, Campus Fuentenueva, Granada, Spain

Keywords: Cancer, choline kinase, green chemistry.

The need for new safer and greener chemical pathways is a strong tendency in nowadays drug discovery

industry.

[1]

Not only environmental aspects but also economic ones, are taken in account while designing a

synthetic pathway.

Lately, we have been interested in the synthesis of bioisosteric inhibitors of the enzyme Choline Kinase. This

cytosolic enzyme is established as a new therapeutic target because of its overexpression in ras-transformed

cells. Its role is the catalytic transformation of choline into phosphocholine by using Mg

2+

and ATP in the first

step of the Kennedy pathway. As a result, the increase of phosphatidylcholine, which is one of the principal

components of the plasma membrane of eukaryotic cells, allows cell proliferation and is associated with

tumorigenesis. Studies using RMN analysis, have recognized this effect in a lot of types of cancer as bladder,

breast, colon, and prostate.

[2]

So the synthesis of ChoK inhibitors has opened a promising window to common

therapies for cancer. Our group has focused on the synthesis of new inhibitors that are characterized for the

bioisosteric exchange

[3, 4]

of some carbons to sulphur atoms, giving room to better interaction with the amino

acidic residues of the enzyme pocket and conserving the antiproliferative activity of the previous ones. The

synthesis of such molecules has been performed in a sustainable and efficient way, paying special attention to

the use of reaction conditions able to generate the minimal amount of waste. Good yields in each step and no

purification of intermediates make of this synthesis a good approach towards sustainability.

 

Biological assays are currently being performed to confirm our hypothesis.

References

1) Louise Summerton, Helen F. Sneddon, Leonie C. Jones, James H. Clark. Green and Sustainable Medicinal Chemistry.

Methods, tools and strategies for the 21st Century Pharmaceutical Industry. RSC Green Chemistry No. 46, Cambridge, 2016.

2) R. Katz-Brull, R. Margalit, P. Bendel, H. Degani. Magnetic Resonance Materials in Biology, Physics and Medicine 6

(1998) 44-52.

3) Francisco Fermín Castro-Navas, Santiago Schiaffino-Ortega, María Paz Carrasco-Jiménez, et al. Future Med. Chem. 2015;

7 (4), 417-436.

4) Santiago Schiaffino-Ortega, et al. Scientific Reports, 2016, 31, 6:23793.

EFMC-ISMC | 223

L016

NOVEL CLASS OF NECROPTOTIC CELL DEATH INHIBITORS:

SYNTHETIC APPROACH AND IN VITRO ASSAYS

André F. S. Luz (1), Andreia C. Vieira (1), Vanda Marques (1), Catarina Pinto (1), Cecília M. P.

Rodrigues (1), Rita C. Guedes (1), Lígia C. Gomes-da-Silva (2), Rui Moreira (1)

1) iMed.UL, Faculty of Pharmacy - University of Lisbon, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal2) Chemistry Department, University of Coimbra, Rua Larga, 3004-535 Coimbra, Portugal

email address: [email protected]

 

Necroptosis is a regulated form of cell death that is associated with several inflammatory and degenerative

diseases. The identification of necroptosis’ pathway mediators, such as receptor interacting protein kinase-1

(RIPK1), and the prevalence of this type of cell death in critical human illnesses, led to an intensive, but yet

unaccomplished, search for high quality necroptosis inhibitors.

1

To find novel necroptosis inhibitors, a high-throughput cell-based phenotypic screening was performed in the

iMed.ULisboa. A group of novel RIPK1 inhibitors, with a IC

50

in the low micromolar range, were identified

(Figure 1).

2

Aiming to modulate the interaction with the RIKP1’s binding pocket, a library of necroptosis inhibitors based on

the hits, was prepared and studied in enzymatic and cell-based assays (Figure 1). The diversified groups

introduced allowed the understanding of the structural requirements for activity and the development of potent

RIPK1 and necroptosis inhibitors.

Figure 1: A library of necroptosis inhibitors, based on novel RIPK1 inhibitors, was synthesised and studied in

enzymatic and in vitro assays leading to structure-activity relationships.

 

This research was funded by projects UIDB/04138/2020 and UIDP/04138/2020 (Fundação para a Ciência e

Tecnologia (FCT), Portugal). Joint funding from FCT and the COMPETE Program through grant

SAICTPAC/0019/2015 is gratefully acknowledged. AFSL also thanks FCT for a PhD fellowship under the

MedChemTrain program (PD/BD/143122/2019).

References

1) Zhuang, C.; Chen, F. J. Med. Chem. 2020, 63, 4, 1490–1510

2) Brito, H.; Marques, V.; Afonso, M. B.; Brown, D. G.; Börjesson, U.; Selmi, N.; Smith, D. M.; Roberts, I. O.; Fitzek, M.;

Aniceto, N.; Guedes, R. C.; Moreira, R.; Rodrigues, C. M. P. Cell Death Discov. 2020, 6 (1)

224 | EFMC-ISMC

L017

THE ROLE OF GUANIDINE-CO

2

ADDUCTS IN CARBOXYLATIONS:

MECHANISTIC INSIGHTS

Jere K. Mannisto (1), Ljiljana Pavlovic (2), Tony Tiainen (1), Martin Nieger (1), Aleksi Sahari (1), Kathrin

H. Hopmann (2), Timo Repo (1)

1) Department of Chemistry, University of HelsinkiP.O. Box 55, A.I. Virtasen aukio 1, 00014 Helsinki, Finland

2) Department of Chemistry, UiT The Arctic University of NorwayN-9037 Tromsø, Norway

Complex pharmaceutically relevant amines are carboxylated selectively using CO

2

and superbases, for example

1,1,3,3-tetramethylguanidine (TMG). The resulting carbamate ion is transformed into various sp

3

-rich

2-oxazolidinones, including fluorinated derivatives, using our method (Scheme 1).

[1]

Scheme 1. Selected examples of CO

2

-derived 2-oxazolidinones.

We have investigated the superbase mode of action experimentally and computationally.

[2]

Interestingly, in the

absence of a CO

2

atmosphere, both zwitterionic TMG–CO

2

and the corresponding bicarbonate (TMGH

+

HCO

3

-

)

are able to act as a stoichiometric source of CO

2

for carboxylation of various nucleophiles. The presented

chemistry of a pre-prepared superbase–CO

2

adduct enables carboxylations without pressurized reactors.

Mechanistically, zwitterionic TMG–CO

2

is not an active carboxylation agent. Instead, once a nucleophile is

added, TMG–CO

2

dissociates, leading to concerted carboxylation, where TMG deprotonates the nucleophile as

it is attacking a free CO

2

(Scheme 2).

Scheme 2. Stoichiometric carboxylation using TMG–CO

2

.

References

1) J. K. Mannisto, A. Sahari, K. Lagerblom, T. Niemi, M. Nieger, G. Sztanó, Timo Repo, Chem. Eur. J. 2019, 25, 10284 –

10289, DOI: 10.1002/chem.201902451

2) J. K. Mannisto, L. Pavlovic, T. Tiainen, M. Nieger, A. Sahari, K. H. Hopmann, T. Repo, Manuscript in preparation

EFMC-ISMC | 225

L018

COPPER (II) BROMIDE AS AN EFFICIENT CATALYST FOR THE

SELECTIVE PROTECTION AND DEPROTECTION OF ALCOHOLS

Roufia Mezaache (1), Yénimégué Albert Dembelé (2), Yann Bikard (3), Jean-Marc Weibel (3), Aurélien

Blanc (3), Patrick Pale (3)

1) Département de Pharmacie, Faculté de Médecine, Université de Batna2, 05000, Algeria.2) Laboratoire de chimie, FMPOS, Université de Bamako, Mali.

3) Laboratoire de synthèse et réactivité organiques, associé au CNRS, Institut de Chimie, Université de Strasbourg, France.

The syntheses of highly functionalized molecules usually require several steps dealing with the protection and

deprotection of those functional groups.

1, 2

The choice of protecting groups is often critical for synthesis success,

specially for the total synthesis of complexe natural products and analogs.

2,3

Benzyl type protecting groups are

among the most commonly used, due to their deprotection conditions orthogonal to other protecting and

functional groups

1-3

, and they have been applied to the protection of alcohols, thiols, amines, and acids.

1, 2

Nevertheless, their introduction is not always simple due to the basic or acid condition required,

in order to

solve this problem, we recently described a convenient and efficient method based on Copper bromide catalyst

(CuBr

2

) has been developed for the protection of Primary and secondary alcohols with

bis(4-methoxyphenyl)méthanol (BMPMOH) in good yield using CuBr

2

as catalyst in acetonitrile at room

temperature.

4

Deprotection could easily be achieved using the same catalyst but in ethanol. Both Cu-catalyzed

protection and deprotection were orthogonal to other methods and fully compatible with other functional groups.

The mildness of these protection and deprotection methods as well as their selectivity render them very useful

tools for total synthesis.

References

1-) Greene, T. W.; Wuts, P. G. M. Protective Groups in Organic Synthesis, 3rd ed. ; J. Wiley & Sons: New York, NY, 1999.

2-) . Kocienski, P. J. Protecting groups, 3rd ed .; G. Thieme: Stuttgart, New York, NY, 2004.

3-) . Nicolaou, K. C.; Snyder, S. A Classics in Total Synthesis II; Wiley-VCH: Weinheim, 2003.

4-) Mezaache, R. ; Dembelé, Y. A. ; Bikard, Y. ; Weibel, J.M. ; Blanc, A. ; Pale, P. Tetrahedron Letters, 2009, 50,

7322–7326.

226 | EFMC-ISMC

L019

FLUOLEAD, A VERSATILE AND SAFE NUCLEOPHILIC

FLUORINATION AGENT

Norimichi Saito (1), Tomochika Iwata (1), Southida Nachampassak (2)

1) UBE Industries, Ltd., Tokyo, Japan2) UBE Europe GmbH, Düsseldorf, Germany

Versatile, safe, shelf-stable and easy-to-handle fluorination agents are strongly desired in both academic and

industrial arenas, since fluorinated compounds have attracted considerable interest in many areas, such as drug

discovery, due to the unique effects of fluorine atoms when incorporated into molecules.

We have discovered and characterized 4-tert-butyl-2,6-dimethylphenylsulfur trifluoride (FLUOLEAD

TM

), as a

crystalline solid which has versatile fluorination capability as deoxo-fluorinating agent in addition to possessing

high thermal stability and unusual resistance to aqueous hydrolysis, compared to traditional agents, such as

DAST and its analogues (see table 1 below).

Table 1. Comparison of FLUOLEAD

TM

 vs competitive products

FLUOLEAD

TM

fluorinates alcohols, aldehydes, ketones and carboxylic acids to give the corresponding

monofluoro, difluoro, and trifluoro products (Scheme 1). The reaction conditions shall be acid, non-polar solvent

and with catalysts e.g Pyridine-HF.

Scheme 1. Application of FLUOLEAD

TM

 for deoxo-fluorination

To summarize, FLUOLEAD

TM

has a high thermal stability, is easy to handle and needs only one step for

deoxo-fluorination. It is now available in large scale and UBE Industries can provide fluorinating technology

under GMP.

References

1) Umemoto, T.; Singh, R. P.; Xu Y.; Saito N. J. Am. Chem. Soc. 2010, 132, 18199. The article can be downloaded freely

from: http://pubs.acs.org/journal/jacsat.

EFMC-ISMC | 227

L020

NOVEL ARYL SULFONAMIDES TARGETING THE

PROTEIN-PROTEIN INTERACTION OF AIMP2-DX2 WITH HSP70 AS

POTENTIAL CANCER THERAPEUTICS

Aneesh Sivaraman (1), Kyoung ho Min (1), Minkyoung Kim (1), Hwa Young Lee (1), YongSeok Choi (2),

Sung hoon Kim (3), Kyeong Lee (1)

1) College of Pharmacy, Dongguk University-Seoul, Goyang, 10326, Republic of Korea.2) Department of Biotechnology, Korea University,Seoul 02841, Republic of Korea

3) College of Pharmacy & College of Medicine, Yonsei University, Incheon 21983, Republic of Korea

The tumorigenic factor AIMP2-DX2, AIMP2 lacking exon 2, is often up regulated in many cancers. Heat-shock

protein HSP70 was identified as a critical determinant for the cellular level of AIMP2-DX2. Binding of the

substrate-binding domain of HSP70 to the N-terminal flexible region and GST domain of DX2 stabilizes DX2

and protects it from Siah1-mediated ubiquitination. Preventing HSP70 binding to DX2 with small molecules

provides a promising therapeutic strategy for AIMP2-DX2-dependent cancers. A structure-activity relationship

study using a sulfonamide hit compound (BCA-1093, IC

50

= 40.14 μM) identified the novel compound

BCL-848 which showed improved inhibition against AIMP2-DX2 (IC

50

= 0.92 μM) with more than 100-fold

selectivity over AIMP2 in a luciferase assay. A molecular docking study using the in house homology model of

DX2 showed that BCL-848 binds to the hydrophobic pocket of AIMP2-DX2, which is proximal to its HSP70

binding domain. Analysis of binding configuration revealed that, BCL-848 was positioned very close to L80,

T82, F116, T117, and K129 residues in the hydrophobic pocket. Interestingly, BCL-848 forms two hydrogen

bonds with the hydrophobic pocket via T82 and D52. Thus BCL-848 effectively induces cancer cell apoptosis

by specifically interrupting the interaction between DX2 and HSP70, which leads to the degradation of DX2 via

Siah1-mediated ubiquitination.

References

1) J. Med. Chem. 2020, 63, 5139−5158

228 | EFMC-ISMC

L021

DEOXYFLUORINATION OF ACYL FLUORIDES TO

TRIFLUOROMETHYL COMPOUNDS BY FLUOLEAD/OLAH'S

REAGENT UNDER SOLVENT-FREE CONDITIONS

Norimichi SAITO (1), Southida NACHAMPASSAK (2)

1) UBE Industries, Ltd., Tokyo, Japan2) UBE Europe GmbH, Düsseldorf, Germany

A new protocol enabling the formation of trifluoromethyl compounds from acyl fluorides has been developed.

The combination of FLUOLEAD

TM

 (4-tert-butyl-2,6-dimethylphenylsulfur trifluoride) and Olah’s reagent in

solvent-free conditions at 70°C initiated the significant deoxyfluorination of the acyl fluorides and resulted in the

corresponding trifluoromethyl products with high yields (up to 99%). This strategy showed a great tolerance for

various acyl fluorides containing aryloyl, (heteroaryl)oyl, or aliphatic acyl moieties, providing good to excellent

yields of the trifluoromethyl (CF

3

) products. Synthetic drug-like molecules were also transformed into the

corresponding trifluoromethyl compounds under the same reaction conditions. A representative reaction example

is proposed below and highlighting the optimized reaction conditions to get CF

3

via FLUOLEAD

TM

 : 

The reason why we chose the acyl-fluoride as the starting material is: (i) acyl-fluoride is thermally and

hydrolytically more stable than the acyl chloride, and (ii) acyl-fluoride is readily available from the

corresponding carboxylic acid as well as aldehyde and alcohol , thus the acyl-fluoride could be a good precursor

for this deoxyfluorination reaction with FLUOLEAD

TM 1)

.

The conversion of acylfluoride to the corresponding CF

3

was conducted by using FLUOLEAD and

nHF-Pyridine under neat, 24h and 70°C with an extreme high yield (99%), while the reaction did not proceed

without nHF-pyridine. In the second example, if we use DAST instead of FLUOLEAD

TM

as deoxofluorinating

agent, the reaction occurred but at a very low yield only 25%. In case that we used the other deoxyfluorination

reagents such as Deoxofluor, Xtalfluor or TBAT, the reaction did not proceed at all

2)

.

Deoxyfluorination can be applied to several types of groups: Aromatics, Heterocyclic and Aliphatic. 

Finally, we discovered recently a new successful work-up method based on amino-alcohol which can completely

remove the side product of FLUOLEAD

TM 

(Ar-SOF), and will be introduced at the conference.

References

1) Y. Liang, Z. Zhao, A. Taya, and N. Shibata., Organic Letters, 2021, 23 (3), 847-852

2) Y. Liang, A. Taya, Z. Zhao, N. Saito, and N. Shibata, Beilstein J. Org. Chem., 2020, 16, 3052-3058

EFMC-ISMC | 229

L022

SYNTHESIS OF SPIROCYCLIC-DHEA DERIVATIVES AS

NEUROTROPHIN MIMETICS

Daniele Narducci (1), Thanasis Rogdakis (2,3), Despoina Charou (2,3), Canelif Yilmaz (4), Kyriakos C.

Prousis (1), Ioannis Charalampopoulos (2,3), Vasileia Ismini Alexaki (4), Achille Gravanis (2,3), Theodora

Calogeropoulou (1)

1) National Hellenic Research Foundation, Institute of Biology, Medicinal Chemistry and Biotechnology, 48 VassileosConstantinou Avenue, 11635 Athens, Greece.

2) Institute of Molecular Biology & Biotechnology Foundation of Research & Technology-Hellas, 100 Plastira Street, 70013Heraklion, Greece.

3) Dept of Pharmacology, Medical School, University of Crete, Stavrakia-Voutes, 71003, Heraklion, Greece.4) Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, TU Dresden, 01307

Dresden, Germany.

Neurotrophins (NTs) compose a small family of Neurotrophic Growth Factors constituted of four orthologues,

associated by structure and function: Nerve Growth Factor (NGF), Brain-Derived Growth Factor (BDNF),

Neurotrophin-3 (NT-3) and Neurotrophin-4 (NT-4, also known as NT-5 and NT-4/5). These large proteins after

being secreted, both centrally and peripherally, bind with high affinity and selectivity to the Tyrosine Receptor

Kinases (TrkA, TrkB and TrkC), to produce powerful neuroprotective and neurogenic effects. On the other hand,

the Pan Neurotrophin Receptor 75 (p75

NTR

) is also bound by NTs with low affinity and by their immature forms

(pro-neurotrophins, pro-NTs) with high affinity, generally resulting in cell death. This small and general

introduction is just the tip of the iceberg of the complex equilibrium of Neurotrophin signaling. Based on these

premises, many pharmacological strategies were attempted, but an optimal solution is yet to be identified. Due to

their polypeptidic nature, in fact, NTs are not suitable for therapeutic use and small druggable molecules,

mimicking neurotrophins’s beneficial actions are highly desirable.

1

Following the discovery that the endogenous steroid precursor Dehydroepiandrosterone (DHEA) activates the

neurotrophin receptor TrkA exerting an activity that is both neuroprotective

2

and anti-neuroinflammatory

3

, our

group synthesized a small library of variously substituted dehydroepiandrosterone derivatives. The

17-spiro-epoxy derivative BNN27 showed anti-apoptotic and neuroprotective activity, resulting from the

selective activation of the TrkA receptor.

4-6

  BNN27, in contrast to DHEA, not only possesses improved affinity

for TrkA, but does not exhibit hormonal side effects.

As a continuation of our studies on steroidal neurotrophin mimetics, we embarked on the synthesis of

C17-spiro-DHEA derivatives bearing five or six-membered rings substituted with a variety of pharmacophore

groups in order to probe the stereoelectronic requirements for optimum neurotrophic/neuroprotective/neurogenic

activity. The new derivatives were evaluated for their agonistic activity for TrkA and TrkB receptors as well as

for their neuroprotective properties on appropriate cellular models.  Moreover, their effect on inflammatory

responses was tested using microglia cells.

Acknowledgement. This project has received funding from the European Union’s Horizon 2020 research and

innovation programme under the Marie Skłodowska-Curie grant agreement No. 765704

(www.euroneurotrophin.eu).

References

1) S. Josephy-Hernandez, S. Jmaeff, I. Pirvulescu, T. Aboulkassim, H. U. Saragovi. Neurotrophin receptor agonists and

antagonists as therapeutic agents: An evolving paradigm. Neurobiol. Dis. 2017, 97, 139-155.

2) I. Lazaridis, I. Charalampopoulos, V.I. Alexaki, N. Avlonitis, I. Pediaditaki, P. Efstathopoulos, T. Calogeropoulou, E.

Castanas, A. Gravanis. Neurosteroid dehydroepiandrosterone interacts with nerve growth factor (NGF) receptors, preventing

neuronal apoptosis.; PLoS Biol. 2011, 9 (4): e1001051.

3) V.I. Alexaki, G. Fodelianaki, A. Neuwirth, C. Mund, A. Kourgiantaki, E. Ieronimaki, K. Lyroni, M. Troullinaki, C. Fujii,

W. Kanczkowski, A. Ziogas, M. Peitzsch, S. Grossklaus, B. Sönnichsen, A. Gravanis, S.R. Bornstein, I. Charalampopoulos,

C. Tsatsanis, T. Chavakis. DHEA inhibits acute microglia-mediated inflammation through activation of the

TrkA-Akt1/2-CREB-Jmjd3 pathway. Mol Psychiatry. 2018, 23(6):1410-1420.

4) T. Calogeropoulou, N. Avlonitis, V. Minas, X. Alexi, A. Pantzou, I. Charalampopoulos,M. Zervou, V. Vergou, E.S.

Katsanou, I. Lazaridis, M. N. Alexis, A. Gravanis. Novel dehydroepiandrosterone derivatives with antiapoptotic,

neuroprotective activity; J. Med. Chem. 2009, 52, 6569-87.

5) I. Pediaditakis, A. Kourgiantaki, K. Prousis, C. Potamitis, K. P. Xanthopoulos, M. Zervou, et al. BNN27, a 17-Spiroepoxy

Steroid Derivative, Interacts With and Activates p75 Neurotrophin Receptor, Rescuing Cerebellar Granule Neurons from

Apoptosis. Front. Pharmacol. 2016, 7, 1-14.

6) I. Pediaditakis, P. Efstathopoulos, K. C. Prousis, M. Zervou, J. C. Arévalo, V. I., E. Nikoletopoulou, C. Potamitis, N.

Tavernarakis, T. Chavakis, A. N. Margioris, M. Venihaki, T. Calogeropoulou, I. Charalampopoulos, A. Gravanis. Selective

and differential interactions of BNN27, a novel C17-spiroepoxy steroid derivative, with TrkA receptors, regulating neuronal

survival and differentiation. Neuropharmacol. 2016, 111:266-282.

230 | EFMC-ISMC

L023

BIS(OXABOROLES) AND BIS(SILOXABOROLES) AS POTENT

ANTIMICROBIAL AGENTS

Krzysztof Nowicki (1), Joanna Krajewska (2), Piotr Pacholak (1,3), Patrycja Wińska (1), Agnieszka E.

Laudy (2), Sergiusz Luliński (1)

1) Warsaw University of Technology, Faculty of Chemistry, Noakowskiego 3, 00-664 Warsaw, Poland2) Department of Pharmaceutical Microbiology, Medical University of Warsaw, Oczki 3, 02-007 Warsaw, Poland

3) University of Warsaw, Faculty of Chemistry, Pasteura 1, 00-093 Warsaw, Poland

Over the past two decades, great attention has been paid to boron heterocycles, especially benzoxaboroles. They

have emerged as a novel class of small-molecule therapeutic agents possessing excellent antifungal,

antibacterial, and anti-inflammatory activity. Thanks to the extensive research carried out in recent years, two

benzoxaborole derivatives have already been successfully marketed, while several others are in clinical trials.

Recently, we have presented silicon-based analogues of benzoxaboroles - benzosiloxaboroles [1,2]. The

introduction of a SiMe

2

group to the oxaborole ring in the place of a methylene group results in the increased

Lewis acidity and lipophilicity, which may be favourable for biological activity. Our preliminary

microbiological studies on simple fluorinated benzosiloxaboroles showed their promising activity against

selected yeast strains [1], whereas more extended systems were found to be potent KPC β‑lactamase inhibitors

[3].

Herein, we present the research devoted to synthesis, structural and physicochemical characterization of the

series of bis(siloxaboroles) and related bis(oxaboroles) based on halogenated biphenyl scaffolds. Following the

basic chemical research, a comprehensive antimicrobial activity evaluation was performed. The title compounds

were prepared starting with symmetrically halogen‑substituted biphenyls such as

4,4’-dibromo-2,2’-difluorobiphenyl and 3,3’-dibromo-5,5’-dihalobiphenyls (Hal = F, Cl, Br). For this reason,

they can be regarded as pseudo-dimers of simple halogenated benzosiloxaboroles and benzoxaboroles.

Investigation of antimicrobial activity showed that obtained bis(siloxaboroles) based on the biphenyl scaffold 1 

demonstrate high activity (MIC < 3.13 mg/L) against Gram-positive cocci, including multi-drug resistant (MDR)

clinical strains (S. aureus MRSA, E. faecalis VanA and VanB and E. faecium VanA and VanB). Moreover,

checkerboard assay revealed synergistic activity against E. faecalis VanA for gentamycin combinations with two

bis(siloxaborole) derivatives. Additionally, cytotoxicity tests were performed.

In conclusion, a series of new bis(oxaborole) and bis(siloxaborole) systems were obtained and comprehensively

characterized. Conducted research showed the high therapeutic potential of tested compounds as three of them

exhibit potent antibacterial activity. Furthermore, physicochemical properties of studied bis(siloxaboroles) are

beneficial from the viewpoint of applications in medicinal chemistry.

The work was supported by the National Science Centre (Poland), Grant No UMO‑2018/31/B/ST5/00210.

References

1) Brzozowska, A.; Ćwik, P.; Durka, K.; Kliś, T.; Laudy, A. E.; Luliński, S.; Serwatowski, J.; Tyski, S.; Urban, M.;

Wróblewski, W. Organometallics 2015, 34, 2924-2932

2) Czub, M.; Durka, K.; Luliński, S.; Łosiewicz, J.; Serwatowski, J.; Urban, M.; Woźniak, K. Eur. J. Org. Chem. 2017, 4,

818-826.

3) Durka, K.; Laudy, A. E.; Charzewski, Ł.; Urban, M.; Stępień, K.; Tyski, S.; Krzyśko, K. A.; Luliński, S. Eur. J. Med.

Chem. 2019, 171, 11-24

EFMC-ISMC | 231

L024

SYNTHESIS OF NEW MraY INHIBITORS AND THEIR BIOLOGICAL

EVALUATION

Martin OLIVER, Raja BEN OTHMAN, HongWei WAN, Mélanie POINSOT, Laurent LE CORRE,

Sandrine CALVET-VITALE, Christine GRAVIER-PELLETIER

Université de Paris, LCBPT, UMR 8601CNRS, F-75006 Paris, France

In the context of Multidrug Resistance (MDR) bacteria, the discovery and development of novel compounds,

efficient against MDR strains, appears to be crucial for the future.

The bacterial transferase MraY is an interesting target for the discovery of new antibiotics. Indeed, it is still an

unexploited target in clinic so there is no resistance that has been developed so far. Moreover, MraY is ubiquitous

in procaryotes and is essential for the bacteria survival. Many natural MraY inhibitors such as liposidomycins or

muraymycins contain an aminoribosyl-O-uridine scaffold shown to be essential for their biological activity

1

. Recently, a crystal structure of the integral membrane enzyme MraY

AA

in complex with muraymycin D2 has

been solved

2

 and allowed us to build a map of the interactions between aminoacids of the active site and this

natural inhibitor (Figure1).

 

 

Our goal is to synthesize new MraY inhibitors with structurally simplified structures as compared to that of

naturally occurring inhibitors and endowed with antibacterial activity. We previously developed the synthesis of

original and simplified inhibitors based on this scaffold with C- and N- triazole linkers

3

.

In the continuity of these results, new families of inhibitors containing various linkers were synthesized (Figure

2) and the biological evaluation of all the synthetized compounds on the MraY

AA

activity was performed. We

will present all these results that have led to promising molecules with activities against purified MraY

AA

in the

micromolar range.

References

1) a. C. Dini, Curr. Top. Med. Chem. 2005, 5, 1221−1236. b) S. Ichikawa, M. Yamaguchi, A. Matsuda. Curr. Med. Chem.

2015, 22, 3951-3979

2) C. Chung, E.H. Mashalidis, T. Tanino, M. Kim, A. Matsuda, J. Hong, S. Ichikawa, S.Y. Lee, Nature 2016, 557-560

3) a. M. J. Fer, S. Olatunji, A. Bouhss, S. Calvet-Vitale, C. Gravier-Pelletier, J. Org. Chem. 2013, 78, 10088–10105. b. M.J.

Fer, A. Bouhss, M. Patrão, L. Le Corre, N. Pietrancosta, A. Amoroso, B. Joris, D. Mengin-Lecreulx, S. Calvet-Vitale, C.

Gravier-Pelletier, Org. Biomol. Chem, 2015, 13, 7193-7222. c. M.J. Fer, L. Le Corre, N. Pietrancosta, N. Evrard-Todeschi, S.

Olatunji, A. Bouhss, S. Calvet-Vitale, C. Gravier-Pelletier, Curr. Med. Chem. 2018, 25, 42, 6013-6029.

232 | EFMC-ISMC

L025

RETROSYNTHETIC SOFTWARE FOR PRACTICING CHEMISTS:

NOVEL AND EFFICIENT IN SILICO PATHWAY DESIGN

VALIDATED AT THE BENCH

Lindsey Rickershauser

Manager, SYNTHIA™ Sales and MarketingCheminformatics Technologies

MilliporeSigmaA business of Merck KGaA, Darmstadt, Germany

In a continuously evolving landscape of in silico chemical intelligence and machine learning, computer assisted

synthetic planning has come to the forefront of discussion in the cheminformatics space. Herein, we describe the

use of SYNTHIA™,a retrosynthetic design software in drug discovery, industrial, and academic laboratories all

over the world. As a product of over 20 years of research, this unique tool is poised to not only get better with

time, but also revolutionize the way chemists approach designing pathways to their complex targets.

SYNTHIA™’s unique approach to building our expert database of known reactions by hand coding each

transformation has allowed this tool to become a bench chemist’s ally by ‘learning’ chemistry much like a

chemist would themselves, and suggesting diverse pathways towards their targets, thus generating ideas and

providing cost effective routes based on each user’s unique needs.

References

1) • Bartosz A. Grzybowski et al., Efficient Syntheses of Diverse, Medicinally Relevant Targets Planned by Computer and

Executed in the Laboratory. Chem, 2018, 3, 522-532.

2) • https://www.nature.com/articles/s41586-020-2855-y

EFMC-ISMC | 233

L026

FLUORINATED OXINDOLE SYNTHESIS VIA ORGANOCATALYTIC

REACTIONS WITH THIOESTER ENOLATES

Dominik Zetschok, Helma Wennemers

Laboratory of Organic Chemistry, D-CHAB, ETH Zurich, Vladimir-Prelog Weg 3, CH-8093 Zurich, Switzerland

Thioester enolate surrogates are utilized by nature for the synthesis of polyketides and fatty acids

1

and are

valuable in organic chemistry for C–C bond formations.

2

Our group developed alkylated and fluorinated malonic

acid half thioesters (MAHTs) and monothiomalonates (MTMs) as masked thioester enolate equivalents and used

them in organocatalytic addition reactions to several electrophiles, including nitroolefins, imines,

isatin-ketimines and aldehydes. The products were obtained in excellent yields and stereoselectivities.

3

We expanded this methodology to reactions of fluorinated thioester enolate equivalents with isatins and

isatin-ketimines to access fluorinated oxindoles. Oxindoles are prominent motifs in many therapeutically active

compounds

4

and fluorine substituents can be expected to further enhance their potency.

5

Fluorinated malonic acid half thioesters (F-MAHTs) were used as thioester enolate equivalents in

organocatalyzed addition reactions to isatins. The products from a range of different N-protected and

nonprotected isatins were obtained under mild reaction conditions in high yields and enantioselectivities. The

unique reactivity of the thioester moiety enabled diverse derivatization and allowed for the straightforward

access to a fluorinated analogue of the anticancer agent (S)-YK-4-279, a therapeutically active compound against

Ewing’s sarcoma (Scheme 1a).

6

The addition of fluorinated monothiomalonates (F-MTMs) to isatin-ketimines using cinchona alkaloids as

catalysts, provided 3-amino oxindoles in excellent yields and stereoselectivities (Scheme 1b). Remarkably low

catalyst loadings and short reaction times sufficed to obtain the product with two adjacent tetrasubstituted

stereocenters without the need for a protecting group at the isatin lactam.

The poster will present the scope of the organocatalytic reactions and the preparation of fluorinated downstream

analogs of the chiral 3-hydroxy and 3-amino oxindoles.

References

1) J. Staunton, K. J. Weissman, Nat. Prod. Rep. 2001, 18, 380–416; A. M. Hill, Nat. Prod. Rep. 2006, 23, 256–320; C.

Khosla, D. Herschlag, D. E. Cane, C. T. Walsh, Biochemistry 2014, 53, 2875–2883; M. C. Walker, B. W. Thuronyi, L. K.

Charkoudian, B. Lowry, C. Khosla, M. C. Y. Chang, Science 2013, 341, 1089–1094.

2) G. Lalic, A. D. Aloise, M. D. Shair, J. Am. Chem. Soc. 2003, 125, 2852–2853.

3) E. Cosimi, J. Saadi, H. Wennemers, Org. Lett. 2016, 18, 6014–6017; E. Cosimi, O. D. Engl, J. Saadi, M. Ebert, H.

Wennemers, Angew. Chem. Int. Ed. 2016, 55, 13127–13131; O. D. Engl, S. P. Fritz, H. Wennemers, Angew. Chem. Int. Ed.

2015, 54, 8193–8197; J. Saadi, H. Wennemers, Nat. Chem. 2016, 8, 276–280.

4) C. Marti, E. M. Carreira, Eur. J. Org. Chem. 2003, 2209; B. M. Trost, M. K. Brennan, Synthesis 2009, 3003.

5) Y. Li, X. Wang, D. Xiao, M. Liu, Y. Du, J. Deng, Adv. Synth. Catal. 2018, 36; J. Qian, W. Yi, X. Huang, J. P. Jasinski,

W. Zhang, Adv. Synth. Catal. 2016, 358, 2811.

6) D. Zetschok, L. Heieck, H. Wennemers Org. Lett. 2021, 23, 1753–1757.

234 | EFMC-ISMC

L027

GOLD CATALYSIS FOR THE SYNTHESIS OF

HETEROSPIROCYCLES

Kossi Efouako SOKLOU, Hamid MARZAG, Sylvain ROUTIER, Karen PLÉ

Institut de Chimie Organique et Analytique (ICOA), UMR 7311, Université d’Orléans, Pôle de chimie, Rue de Chartres,45100 Orléans, France

The conception and synthesis of new biologically active molecules, coupled with an increased exploration of

new chemical space is an important goal in medicinal chemistry. The advantage of 3D fragments over 2D

fragments, giving better physicochemical properties, has been widely documented.

1, 2

Due to their molecular

rigidity, and an increased number of Csp

3

carbons, interest in the synthesis of new and original spirocyclic

compounds is constant, in academia as well as in the pharmaceutical industry. The increasing occurrence of

heterospirocycles that possess a heteroatom directly attached to the quaternary spirocyclic carbon is a driving

force for the development of general synthetic strategies for these heterospirocycles. Starting from commercial

piperidone derivatives and quinic acid, suitable substrates for the target heterospirocycles were prepared. A

screening of different transition metal catalysts and the tuning of different reaction parameters led to the best

cyclization conditions. The optimized strategy was applied to several substrates, exploring molecular diversity

and studying the scope of the reaction. Selected heterospirocycles were directed towards a medicinal chemistry

program exploring new potent kinases inhibitors.

 

References

1) M. Aldeghi, S. Malhotra, D. L. Selwood, A. W. E. Chan, Chem. Biol. Drug Des., 2014, 83, 450-461

2) F. Lovering, J. Bikker, C. Humblet, J. Med. Chem., 2009, 52, 6752-6756. B) Lovering, F. MedChemComm 2013, 4,

515-519.

EFMC-ISMC | 235

L028

STEREOSELECTIVE REDUCTION OF STEROIDAL ENONES

LEADING TO BIOLOGICALLY IMPORTANT SKELETONS

Eszter Szanti-Pinter, Eva Kudova

Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo namesti 2, 16610 Prague 6, CzechRepublic

Our research is focused on the reduction of enone functionalities on steroid skeletons, leading to biologically

important 5β-steroid structures. For example, bile acids, neurosteroids and their synthetic analogues, neuroactive

steroids possess a 5β-steroid backbone. Bile acids play a role in the digestion and absorption of lipids, also they

are important signaling molecules with endocrine functions.

1

In addition, neurosteroids play a role in neuronal

functions, including cognition, memory processes, and neuroprotection. Consequently, neurosteroids and

neuroactive steroids have been implicated in the treatment of variety of central nervous system diseases, i.e.

anxiety, schizophrenia and neurodegenerative diseases such as Alzheimer’s disease.

2

The asymmetric hydrogenation of unsaturated organic compounds is an important method to obtain optically

active molecules. Besides the traditional hydrogenation using hydrogen gas, new alternative methods were

developed in the recent years. The organocatalytic reduction of enones with biomimetic reductants leads to high

yields and stereoselectivities.

3

Diastereoselective synthesis of some 5β-steroids has been reported via

organocatalytic transfer hydrogenation.

4

Besides organocatalytic transfer hydrogenations, copper-catalyzed

stereoselective conjugate reductions were developed in the recent years, using an alternative reductant

polymethylhydrosiloxane.

5

As the starting materials, commercially available steroids – progesterone and testosterone were selected. The

previously mentioned strategies for reduction of enone moiety on both steroids were tested and the selectivity

affording desired 5β-isomers were evaluated. The results of this study will be described and discussed.

 

This work was supported by the Czech Science Foundation (GACR), grant 19-05318S and Research Project ofthe AS CR RVO, RVO Grant 61388963.

References

1) Houten, S.M.; Watanabe, M.; Auwerx, J. EMBO J. 2006, 25, 1419.

2) Ratner, M.H.; Kumaresan V.; Farb, D.H. Front. Endocrinol, 2019, 10, 169.

3) Phillips, A.M.F.; Pombeiro, A.J.L. Org. Biomol. Chem., 2017, 15, 2307.

4) Ramachary, D.B.; Sakthidevi, R.; Reddy, P.S. RSC Adv., 2013, 3, 13497.

5) Moritani, Y.; Appella, D.H.; Jurkauskas, V.; Buchwald, S.L. J. Am. Chem. Soc. 2000, 122, 6797.

236 | EFMC-ISMC

L029

PEPTIDE CATALYSTS FOR CONJUGATE ADDITION REACTIONS

WITH C-SUBSTITUTED MALEIMIDES

Greta Vastakaite, Claudio Grünenfelder, Helma Wennemers

Laboratory of Organic Chemistry, ETH Zürich, Vladimir-Prelog-Weg 3, Zürich, Switzerland

Peptides of the type H-Pro-Pro-Xaa (Xaa = any amino acid) are highly reactive and stereoselective catalysts for

organocatalytic C–C bond formations, such as aldol reactions

[1]

 as well as conjugate addition reactions of

aldehydes to, for example, nitroolefins

[2] 

and unprotected maleimide.

[3] 

The peptide catalysts are so reactive that

loadings of less than 1 mol% suffice to obtain the products in high yields,  enantio- and diastereoselectivities and

the peptides can be immobilized and used in flow chemistry.

[4] 

ESI‑MS, React-IR and NMR spectroscopic

studies allowed for detailed insight into the mechanism.

[5]

The poster will focus on the expansion of the scope of peptide catalyzed conjugate addition reactions to reactions

with C-substituted maleimides that provide chiral succinimides with 3 contiguous stereogenic centres. We show

that the reaction proceeds with high regio- and chemoselectivity and without major side reactions of the

unprotected maleimides. We also present insights into the reaction mechanism.

References

1) P. Krattiger, R. Kovasy, J. D. Revell, S. Ivan, H. Wennemers, Org. Lett. 2005, 7, 6, 1101.

2) a) M. Wiesner, M. Neuburger, H. Wennemers, Chem. Eur. J. 2009, 15, 10103. b) J. Duschmalé, H. Wennemers, Chem.

Eur. J. 2012, 18, 1111. c) R. Kastl, H. Wennemers, Angew. Chem. Int. Ed. 2013, 52, 7228, d) T. Schnitzer, A. Budinska, H.

Wennemers, Nature Catalysis 2020, 3, 143, e) J. S. Möhler, T. Schnitzer, H. Wennemers, Chem. Eur. J. 2020, 15623.

3) C. Grünenfelder, J. Kisunzu, H. Wennemers, Angew. Chem. Int. Ed. 2016, 55, 8571.

4) Y. Arakawa, H. Wennemers, ChemSusChem 2013, 6, 242.

5) a) F. Bächle, J. Duschmalé, C. Ebner, A. Pfalz, H. Wennemers, Angew. Chem. Int. Ed. 2013, 25, 4511. b) J. Duschmalé, J.

Wiest, M. Wiesner, H. Wennemers, Chem. Sci. 2013, 4, 1312. c) C. Rigling, J. K. Kisunzu, J. Duschmalé, D. Häussinger, M.

Wiesner, M. O. Ebert, H. Wennemers, J. Am. Chem. Soc. 2018, 140, 10829. d) J. S. Möhler, T. Schnitzer, H. Wennemers,

Chem. Sci. 2020, 11, 1942. e) T. Schnitzer, H. Wennemers, J. Org. Chem. 2020, 85, 7633.

EFMC-ISMC | 237

L030

FROM INNOVATION TO THE MARKET: ADDING VALUE TO THE

COMPOUNDS FROM ACADEMIC RESEARCH AND TEACHING.

Divneet Kaur (1), Steve Brough (2)

1) School of Chemistry, Business Partnership Unit, University of Nottingham, University Park, Nottingham NG7 2RD2) Key Organics, Camelford, Cornwall PL32 9RA, UK

Research and teaching activities at universities produces a large number of novel chemical compounds that are

unavailable on the global market. However, on completion of  research projects, many of these compounds have

no further use and end up being stored before being sent for disposal. To maximise the value of these chemicals,

a unique initiative, called Nottingham Research Chemicals (NRC), was established in the School of Chemistry

of the University of Nottingham. This pioneering project allows the introduction of chemicals from research and

teaching to the market via collaboration with our industrial partner – Key Organics Ltd. Since mid-2015, the

NRC project has introduced 169 (as of May 2021) various compounds that resulted from teaching (summer

studentships) and research (PhD and post-doctoral) and continues to grow its portfolio. Further details of the

latest compounds identified and commercialised by NRC and Key Organics are described below.

Research and teaching activities at universities produces a large number of novel chemical compounds that are

unavailable on the global market. However, on completion of  research projects, many of these compounds have

no further use and end up being stored before being sent for disposal. To maximise the value of these chemicals,

a unique initiative, called Nottingham Research Chemicals (NRC), was established in the School of Chemistry

of the University of Nottingham. This pioneering project allows the introduction of chemicals from research and

teaching to the market via collaboration with our industrial partner – Key Organics Ltd. Since mid-2015, the

NRC project has introduced 169 (as of May 2021) various compounds that resulted from teaching (summer

studentships) and research (PhD and post-doctoral) and continues to grow its portfolio. Further details of the

latest compounds identified and commercialised by NRC and Key Organics are described below.

References

1) R. H. Beddoe, K. G. Andrews, V. Magné, J. D. Cuthbertson, J. Saska, A. L. Shannon-Little, S. E. Shanahan, H. F.

Sneddon, R. M. Denton, Science, 2019, 365, 910–914. V. Magn and L. T. Ball, Chem. Eur. J., 2019, 25, 8903 – 8910. K. G.

Andrews, R. Falzova and R. D. Denton, Nature Comm., 2017, 8, 15913. J. Li, S. Jiang, G. Procopiou, R. A. Stockman, G.

Yang, Eur. J. Org. Chem., 2016, 21, 3500

2) V. Magn and L. T. Ball, Chem. Eur. J., 2019, 25, 8903 – 8910.

3) . G. Andrews, R. Falzova and R. D. Denton, Nature Comm., 2017, 8, 15913.

4) J. Li, S. Jiang, G. Procopiou, R. A. Stockman, G. Yang, Eur. J. Org. Chem., 2016, 21, 3500

238 | EFMC-ISMC

L031

SYNTHESIS AND ANTICHOLINESTERASE ACTIVITY OF NOVEL

COUMARIN DERIVATIVES CONTAINING AMINOALCOHOL

GROUP

Ebru Didem COSAR, Belma ZENGİN KURT

Bezmialem Vakıf University, Faculty of Pharmacy, Department of Pharmaceutical Chemistry

Alzheimer’s disease (AD) is the most common neurodegenerative disease with symptoms of memory loss,

cognition defect and behavioural impairment.  AD is associated with a selective loss of cholinergic neurons in

the brain and decreasing levels of acetylcholine (ACh). The classical hypothesis of AD is the cholinergic

hypothesis, which suggests that acetylcholinesterase inhibitors (AChEI) could increase the levels of ACh in AD

patients through the inhibition of AChE and, therefore, relieve some symptoms experienced by AD patients.

Therefore, the search for potent AChE inhibitors plays an important role in the treatment of AD (1).

Coumarin (1,2H-chromen-2-one or 2H-1-benzopyran-2-one) is a bicyclic heterocycle compound, consisting of

benzene and 2-pyrone rings. Coumarins are also a wide class of natural and synthetic compounds that showed

versatile pharmacological activities (2). As seen in previous studies by our team, compounds containing

coumarin ring have potential anticholinesterase inhibition effects (3, 4). Considering these studies, we were

aimed that the synthesis of new coumarin derivatives with cholinesterase inhibition.

In this study, 9 new coumarin derivatives containing the aminoalcohol group were synthesized (Figure 1). All

the new compounds were characterized by

1

H NMR,

13

C NMR, IR, and MS. Molecular modelling studies were

performed for the compounds and according to these results, an anticholinesterase inhibition experiment was

performed for compound 3i by Elmann Method (5). According to the result of this experiment, inhibition of

acetylcholinesterase of 3i was found to be IC

50

=59.7 µM and the inhibition of butyrylcholinesterase was found to

be IC

50

=52.0 µM concentration.

As a result of molecular docking studies of the designed compounds to the active site of acetylcholinesterase

(PDB ID: 1DX6, resolution: 2.3 A) and butyrylcholinesterase (PDB ID: 4BDS, resolution: 2.1 A) enzymes, it is

estimated that there are compounds with possible anticholinesterase activity (Schrodinger Release 2021-1; Glide,

Schrodinger, LLC, New York, NY, 2021).

Acknowledgement: This work was supported by the Bezmialem Research Fund of the Bezmialem Vakif

University, Project no: 20210220.

References

1) Kurt BZ, Gazioglu I, Sonmez F, Kucukislamoglu M. Synthesis, antioxidant and anticholinesterase activities of novel

coumarylthiazole derivatives. Bioorg Chem. 2015;59:80-90.

2) Kurt BZ, Kandas NO, Dag A, Sonmez F, Kucukislamoglu M. Synthesis and biological evaluation of novel

coumarin-chalcone derivatives containing urea moiety as potential anticancer agents. Arabian Journal of Chemistry.

2020;13(1):1120-9.

3) Sonmez F, Kurt BZ, Gazioglu I, Basile L, Dag A, Cappello V, et al. Design, synthesis and docking study of novel

coumarin ligands as potential selective acetylcholinesterase inhibitors. J Enzym Inhib Med Ch. 2017;32(1):285-97.

4) Kurt BZ, Gazioglu I, Sevgi E, Sonmez F. Anticholinesterase, Antioxidant, Antiaflatoxigenic Activities of Ten Edible Wild

Plants from Ordu Area, Turkey. Iran J Pharm Res. 2018;17(3):1047-56.

5) Ellman GL, Courtney KD, Andres V, Jr., Feather-Stone RM. A new and rapid colorimetric determination of

acetylcholinesterase activity. Biochem Pharmacol. 1961;7:88-95.

EFMC-ISMC | 239

Posters - TechnologiesM. Novel Strategies and Methods for Drug Discovery and

Development

240 | EFMC-ISMC

M001

SYNTHESIS OF NOVEL 3-AMINOINDOLE DERIVATIVES AND

THEIR BIOLOGICAL EVALUATION AGAINST MYCOBACTERIUM

TUBERCULOSIS

David Rikhotso, Winston Nxumalo, Tlabo Leboho

Department of Chemistry, University of Limpopo, Sovenga, South Africa, 0727

Tuberculosis (TB), a bacterial infection caused by Mycobacterium tuberculosis (M. tb) is one of the deadly

diseases in the world, mostly in developing continents such as Africa and Asia. World Health Organization

(WHO) reported that in 2018, 1.5 million people died from TB while an estimated 10 million worldwide (and

counting) were infected with this disease.

1

Several drugs have been developed to address this including the first

line (Isoniazid, Pyrazinamide, Rifampicin, Ethambutol), second line (Levofloxacin, Streptomycin, Capreomycin,

Ofloxacin) and those developed later especially for MDR-TB such as Linezolid, Clofazimine, and Amoxicillin)

2

.

However, the use of these drugs is under threat due to the emergence of multi-drug and extensively-drug

resistant TB strains, thus hampering efforts to combat this disease. To add to this, the time required for the

treatment of TB using the recent regimen of drugs ranges anywhere between 3 – 9 months, thus increasing the

chances of patients relapsing from the treatment

3

.  The indole motif is present in a large number of therapeutic

substances such as oxypertine, acemetacin, and panobinostat.

4

Owing to their diverse biological properties, of

particular interest are 3-aminoindole derivatives as potential anti-TB agents. Aminoindoles were found to

possess various biological activities, for example, 7-aminoindoles were found to be active against diabetes,

cancer, and necrosis inhibitor.

5

5-Aminoindole derivatives were found to be HIV protease inhibitors, while

melatonin was used for the treatment of strokes.

6

Additionally,  Schiff bases of indoline-2,3-dione (isatin) were

found to exhibit anti-TB activity.

7

Therefore, in this poster presentation, I will be presenting the synthesis of

3-aminoindole derivatives (Figure 1) starting from 2-iodoanilines (one-pot synthesis) and their biological activity

against Mycobacterium tuberculosis (H37Rv).

 

References

1.) https://www.who.int/news-room/fact-sheets/detail/tuberculosis accessed on 20/02/2020.

2.) Rupesh V. Chikhale, Mahesh A. Barmade, Prashant R. Murumkar, and Mange Ram Yadav; J. Med. Chem. 2018, 61, 19,

8563-8593.

3.) Katherine A. Abrahams, Jonathan A. G. Cox, Vickey L. Spivey, Nicholas J. Loman, Mark J. Pallen, Chrystala

Constantinidou, Raquel Fernández, Carlos Alemparte, Modesto J. Remuiñán, David Barros, Lluis Ballell, Gurdyal S. Besra;

PLoS ONE 7(12): e52951.

4.) Hao Zhang, Hao-Yang Wang, Yixin Luo, Chaohuang Chen, Yimiao Cao, Pinhong Chen, Yin-Long Guo, Yu Lan,

Guosheng Liu; ACS Catal. 2018, 8, 3, 2173-2180.

5.) In-Chang Hwang, Ju-Young Kim, Ji-Hyun Kim, Joo-Eun Lee, Ji-Yun Seo, Jae-Won Lee, Jonghanne Park, Han-Mo Yang,

Soon-Ha Kim, Hyun-Jai Cho, and Hyo-Soo Kim; Hypertension. 2018; 71: 1143–1155.

6.) Patent Corporation Treaty, “Indolylmaleimide derivatives”, World intellectual property organization, 2004.08.26.

7.) Tarek Aboul-Fadl, Fayzah A.S.Bin-Jubair, Omima Aboul-Wafa; Eur. J. Med. Chem., 2010, 45, 10, 4578-4586.

EFMC-ISMC | 241

M002

Δ-QUANTUM MACHINE LEARNING FOR DRUG-LIKE MOLECULES

WITH E(3)-EQUIVARIANT GRAPH NEURAL NETWORKS

Kenneth Atz (1), Clemens Isert (1), Markus Böcker (1), José Jiménez-Luna (1,2), Gisbert Schneider (1,3)

1) ETH Zurich, Department of Chemistry and Applied Biosciences, RETHINK, Vladimir-Prelog-Weg 4, 8093 Zurich,Switzerland.

2) Department of Medicinal Chemistry, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Strasse 65, 88397Biberach an der Riss, Germany.

3) ETH Singapore SEC Ltd, 1 CREATE Way, #06-01 CREATE Tower, Singapore 138602, Singapore.

Drug discovery and the material sciences benefit from fast and accurate calculations of quantum-chemical

molecular properties [1]. Geometric deep learning models [2] have shown promising results for small organic

molecules [3]. However, the application of such neural networks for larger drug-like molecules such as peptides,

macrocycles or steroids is still underexplored. ∆-Quantum machine learning, which is a technique that aims to

learn corrections to computationally inexpensive methods to approximate more accurate but expensive quantum

methods, has shown superior performance to directly predicting the property of interest [4]. Here we present an

open-source Python suite for the ∆-learning of quantum-chemical properties at the density functional theory (ω

B97X-D/def2-SVP) level-of-theory using semiempirically determined equivalents on the GFN2-xTB

level-of-theory [5]. We present E(3)-equivariant graph neural networks trained on the newly-released QMugs

data collection [6]. QMugs contains the quantum-chemical properties of approximately 2M conformers of 665k

bioactive molecules. We show significant predictive improvements for ∆-machine learning over direct density

functional theory (DFT) learning for a variety of quantum observables. The software suite provides both a Bash

interface and a fully-documented Python API to enable fast and accurate property prediction for drug-like

molecules.

References

1) Jiménez-Luna, J., Grisoni, F., Weskamp, N. and Schneider, G. Artificial intelligence in drug discovery: Recent advances

and future perspectives. Expert Opinion on Drug Discovery, 1–11 (2021).

2) Bronstein, M. M., Bruna, J., Cohen, T. & Veličković, P. Geometric deep learning: Grids, groups, graphs, geodesics, and

gauges. arXiv preprint arXiv:2104.13478 (2021).

3) Schütt, K. T., Unke, O. T. & Gastegger, M. Equivariant message passing for the prediction of tensorial properties and

molecular spectra. arXiv preprint arXiv:2102.03150 (2021).

4) Bogojeski, M., Vogt-Maranto, L., Tuckerman, M. E., Müller, K.-R. & Burke, K. Quantum chemical accuracy from density

functional approximations via machine learning. Nature communications, 1–11 (2020).

5) Bannwarth, C. et al. Extended tight-binding quantum chemistry methods. Wiley Interdisciplinary Reviews: Computational

Molecular Science, e01493 (2020).

6) Isert, C., Atz, K., Jiménez-Luna, J. & Schneider, G. QMugs: Quantum mechanical properties of drug-like molecules.

submitted (2021).

242 | EFMC-ISMC

M003

DEVELOPMENT OF p53 ACTIVATORS TO TARGET COLORECTAL

CANCER: HIT OPTIMIZATION OF TRYPTOPHANOL-DERIVED

ISOINDOLINONES

Valentina Barcherini (1), Joana Almeida (2), Elizabeth A. Lopes (1), Mi Wang (3), Alexandra M. M.

Antunes (4), Mattia Mori (5), Shaomeng Wang (3), Lucília Saraiva (2), Maria M.M. Santos (1)

1) Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal;2) Department of Biological Sciences, Universidade do Porto, Porto, Portugal;

3) Roger Cancer Center, Medical School, University of Michigan, Ann Arbor, USA;4) Centro de Química Estrutural, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal;

5) Department of Biotecnology, Chemistry and Pharmacy, University of Siena, Italy

Colorectal cancer (CRC) is the third most recurrent cancer worldwide and often diagnosed in advanced clinical

stage. Poor therapeutic options are available for its treatment, with surgery, neoadjuvant radiotherapy and

adjuvant chemotherapy the ones with highest rate of success. The tumor suppressor p53 is a protein expressed in

all types of human cancers. In 53% of CRC cases, the TP53 gene undergoes hemizygous loss and patients with

mutated p53 gene gain multidrug resistance leading to therapy failure

1

. p53, in its wild-type status, is also found

inactivated by its principal transcriptional targets from the murine double minute protein family (MDMs),

MDM2 and MDMX. Hence, tackle full reactivation of protein p53 represents an appealing anticancer strategy

2

.

In this area of research, we have identified a small molecule (SLMP53-1), reactivator of wild-type and mutant

p53, with promising in vitro and in vivo p53-dependent antitumor activity in CRC

3

. In this communication, we

will present our most recent results in the hit optimization of SLMP53-1, including the identification and

synthesis of the Phase I metabolites identified for the most promising derivatives. The target compounds were

evaluated as potential anticancer agents, showing promising anti-proliferative activities in human colorectal

carcinoma HCT116 cells. From this process, we obtained a compound six-fold more active, and also more

selective for HCT116 cells expressing p53 over cells without p53, and with low toxicity in normal cells (Figure

1)

4

Figure 1 Tryptophanol-derived isoindolinone 1 as potential new lead compound in human colorectal cancer.

 

Acknowledgments

We thank FCT (Fundação para a Ciência e a Tecnologia) for financial support through UIDB/04138/2020,

UIDB/50006/2020, PTDC/QUI-QOR/1304/2020, PTDC/QUI-QOR/29664/2017, PD/BD/143126/2019 and

SFRH/BD/137544/2018

 

References

[1]) Van der Jeught, K.; et al. World J. Gastroenterol. 2018, 24(34): 3834

[2]) a) Espadinha, M.; et al. Curr. Top. Med.Chem. 2018, 18(8), 647; b) Lopes E.A.; et al. Curr. Med. Chem. 2019, 26(41),

7323

[3]) a) Soares, J.; et al. Oncotarget 2016, 7(4), 4326; b) Gomes, A.S.; et al. Biochim. Biophys. Acta (B.B.A.), 2020, 1864(1),

129440

[4]) Barcherini, V.; et al. ChemMedChem 2021, 16(1), 250-258

EFMC-ISMC | 243

M004

A MULTIDISCIPLINARY APPROACH TO DRUG DISCOVERY

REVEALS NEW PHARMACEUTICAL TOOLS AGAINST

NEURODEGENERATIVE DISEASES

Runfola Massimiliano (1), Gul Sheraz (2), Perni Michele (3), Marchese Maria (4), Mero Serena (4),

Chiellini Grazia (5), Santorelli Filippo (4), Vendruscolo Michele (3), Rapposelli Simona (1)

1) Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126, Pisa, Italy2) Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Schnackenburgallee 114, 22525 Hamburg,

Germany3) Centre for Misfolding Diseases, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK

4) Molecular Medicine, IRCCS Stella Maris, Via dei Giacinti 2, 56128 Calambrone (Pisa), Italy5) School of Medicine, University of Pisa, Via Savi 10, Pisa, 56126, Italy

As life expectancy rises worldwide, we are confronted with a worrisome increase in the number of people

affected by neurodegenerative disorders (NDDs), such as Alzheimer’s (AD) and Parkinson’s diseases. The

dramatic status of drug discovery in this field, with no disease-modifying drug approved by the FDA to date, is

also underlying the weaknesses of currently available strategies. In this context, a paradigm-shift towards

advanced pharmaceutical approaches to develop safe drugs able to counteract the onset and progression of NDDs

is urgently needed. Several decades of research in this field are leading to the idea that a tangled network of

pathogenic mechanisms affecting the fragile neuronal homeostasis lies behind NDDs. Protein homeostasis

collapse, malfunctioning of lipid metabolism, neuroinflammation, and oxidative stress are just a few examples of

the large number of mechanisms associated with neurodegenerative processes. This complexity largely

contributes to the high attrition rate characterizing the drug discovery process in this area of research. Here, we

propose an innovative multidisciplinary approach to drug discovery encompassing the combination of the use of

prodrug, phenotypic drug discovery, and polypharmacology.

 

We previously reported a class of small molecules sharing a diphenylmethane core obtained through chemical

manipulations of triiodothyronine (T3) and its metabolite 3-iodothyronamine (1, 2, 3). Analogously to parent

compounds, these small molecules show an interesting and unique polypharmacology including modulation of

lipids metabolism, autophagy, neuronal development, and oligodendrogenesis (4). Within this class, ten

compounds showed a safe profile against cardiotoxicity, cytotoxicity, metabolic stability, and off-target liability.

Further evaluations using a high accurate tracking platform for C. elegans (Wide-Field Nematode Tracking

Platform) showed also a promising recovery of pathological conditions in AD-affected models consequent to

protein aggregates clearance mediated by autophagy promotion (5). Finally, safety and non-toxicity of selected

compounds were confirmed in Zebrafish. These results illustrate the potential of the drug discovery process that

we have developed.  

References

1) Chiellini, G. et al. Design, Synthesis, and Evaluation of Thyronamine Analogues as Novel Potent Mouse Trace Amine

Associated Receptor 1 (m TAAR1) Agonists. Journal of medicinal chemistry 58, 5096-5107 (2015).

2) Chiellini, G. et al. Hit-to-Lead Optimization of Mouse Trace Amine Associated Receptor 1 (mTAAR1) Agonists with a

Diphenylmethane-Scaffold: Design, Synthesis, and Biological Study. J Med Chem 59, 9825-9836,

doi:10.1021/acs.jmedchem.6b01092 (2016).

3) Runfola, M. et al. Design, synthesis and biological evaluation of novel TRβ selective agonists sustained by

ADME-toxicity analysis. European Journal of Medicinal Chemistry 188, 112006 (2020).

4) Bellusci, L. et al. Endogenous 3-Iodothyronamine (T1AM) and Synthetic Thyronamine-like Analog SG-2 Act as Novel

Pleiotropic Neuroprotective Agents Through the Modulation of SIRT6. Molecules. 2020 Jan;25(5):1054.

10.3390/molecules25051054

5) Koopman, M. et al. Assessing motor-related phenotypes of Caenorhabditis elegans with the wide field-of-view nematode

tracking platform. Nat Protoc 15, 2071-2106, doi:10.1038/s41596-020-0321-9 (2020).

244 | EFMC-ISMC

M005

NEUTRAL 5-HT6 RECEPTOR ANTAGONISTS WITH

GLIOPROTECTIVE AND COGNITIVE-ENHANCING PROPERTIES:

AN IMPACT OF STRUCTURAL MODIFICATIONS ON VARIOUS

CONFORMATIONAL STATES OF THE RECEPTOR

David Vanda (1), Vittorio Canale (2), Severine Chaumont-Dubel (3), Rafał Kurczab (4), Grzegorz Satała

(4), Paulina Koczurkiewicz-Adamczyk (2), Martyna Krawczyk (4), Wojciech Pietruś (4), Klaudia Blicharz

(2), Elżbieta Pękala (2), Andrzej J. Bojarski (4), Piotr Popik (4), Philippe Marin (3), Miroslav Soural (1),

Paweł Zajdel (2)

1) Faculty of Science, Palacký University, 12 17.listopadu Street, 771-46 Olomouc, Czech Republic2) Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Kraków, Poland

3) Institut de Génomique Fonctionnelle, CNRS UMR 5203, INSERM U1191, Université de Montpellier, Montpellier Cedex 5,France

4) Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, 31-324 Kraków, Poland

             In contrast to the initial view, G-protein coupled receptors (GPCRs) exist in an equilibrium of multiple

conformational states, including different active states, which depend on the nature of the bound ligand.             

In consequence, different conformational states can initiate specific signal transduction pathways [1,2].           

The challenge is to characterize the ligands acting on or stabilizing specific receptor conformations.

             To investigate these effects at the 5-HT

6

receptor, a focused library of 3H-imidazo[4,5-b]pyridine and   

1H-imidazo[4,5-c]pyridine derivatives was designed [3,4]. The study identified compound PZ-1727

(1-[(3-chlorophenyl)sulfonyl]-2-ethyl-4-(piperazin-1-yl)-1H-imidazo[4,5-c]pyridine), which acts as a potent

5-HT

6

receptor neutral antagonist at Gs signaling, and does not impact neurite growth, in a process controlled   

by Cdk5 signaling pathways. Molecular dynamic simulations highlighted intramolecular constraints and receptor

conformational changes at transmembrane helices 6, 5, and 4, resulting in different ligand-receptor interactions

of the identified neutral antagonist compound PZ-1727 and the previously reported inverse agonist PZ-1444.   

 In cell-based assays, neutral antagonists of the 5-HT

6

receptor (compound PZ-1727 and CPPQ), but not inverse

agonists (SB-258585, intepirdine and PZ-1444), displayed glioprotective properties against 6-OHDA- and

doxorubicin- induced cytotoxicity. Notably, both observed effects suggest that targeting the activated

conformational state of 5-HT

6

R with neutral antagonists implicates the protecting properties of astrocytes.

Additionally, compound PZ-1727 prevented scopolamine-induced learning deficits in the novel object

recognition (NOR) test in rats. We propose compound PZ-1727 as a molecular probe that can be used for further

understanding of the functional outcomes of the different conformational states of the 5-HT

6

receptor.

References

1) Chaumont-Dubel, S. et al., Neuropharm. 2020, 172, 107839-107853.

2) Pujol, C. et al., Sci. Signal. 2020, 13, 1-14.

3) Vanda, D. et al., Eur. J. Med. Chem. 2018, 144, 716-729.

4) Vanda, D. et al., J. Med. Chem. 2021, 64, 1180-1196.

EFMC-ISMC | 245

M006

STRUCTURAL CHARACTERIZATION AND STABILITY OF

PROTEINS IN SOLID FORMS

Aljoša Bolje, Stanislav Gobec

Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, SI-1000 Ljubljana, Slovenia

Protein therapeutics are becoming increasingly important as an alternative treatment for a variety of diseases. For

better stability, proteins are often formulated as solid dosage forms, the most common of which are lyophilizates.

Their stability depends on the preservation of the native structure of the proteins during lyophilization, as well as

in the lyophilizates. During lyophilization cycle, proteins are exposed to various stress factors that, in

combination with the excipients, can affect the protein structure in the final solid form. If the native structure of

the protein is not maintained during lyophilization, this may be reflected in an unstable final pharmaceutical

product and consequently in its quality, safety and efficiency. Characterization of proteins in solid form is less

established, as most analytical methods evaluate critical properties in solution, which is not necessarily

indicative of adequate stabilization of the protein in the solid phase and thus long-term stability of the

pharmaceutical form. Characterization of proteins in solid form can be used to evaluate both secondary and

tertiary structure during formulation development.

 

In addition to structural characterization, monitoring protein aggregation is also very important. Together with

denaturation and surface adsorption, aggregation can affect the activity and stability of lyophilized proteins,

leading to poorer quality, safety and efficiency of the final drug product.

1

Only formulations that retain their

structure in the solid state are then included in stability studies, which are of particular importance for the

development of protein drugs. In this work, the study of protein structure and stability in solid dosage forms

using analytical methods such as FTIR, NIR, Raman, solid-state fluorescence, solid-state UV-Vis and solid-state

NMR spectroscopy, as well as circular dichrosim, DSC and X-ray powder diffraction is presented. Aggregation

phenomena were also studied by size exclusion chromatography and dynamic light scattering.

2

References

1) Bolje, A.; Gobec, S. (2021) (under peer-review)

2) Bolje, A. et al. (2021) (unpublished results)

246 | EFMC-ISMC

M007

SCAFFOLD HOPPING FOR THE DIVERSIFICATION OF SMALL

MOLECULES TARGETING PROTEIN AGGREGATION

Timothy B. Callis (1), Hei Wun Alison Cheng (2), Andrew P. Montgomery (1), William Jorgensen (1),

Jonathon J. Danon (1), Eryn L. Werry (2), Michael Kassiou (1)

1) School of Chemistry, University of Sydney, Sydney, NSW2) School of Medical Sciences (Pharmacology), University of Sydney, Sydney, NSW

Dementia encompasses a wide range of neurodegenerative diseases that cause the physical and mental capacity

of patients to deteriorate and ultimately leads to death. Dementia disease states are estimated to currently affect

50 million people globally with costs exceeding US$1 trillion in 2018.

[1]

Without significant medical

breakthroughs, the number of people suffering from dementia and the associated cost is predicted to almost triple

by 2050.

[1]

Treatments currently available for dementia patients only provide short-term relief from common

symptoms without targeting the pathological causes of these diseases.

[2]

We have explored pharmacophores

reported to reduce neurotoxic aggregates of transactive-response DNA binding protein 43 kDa (TDP-43).

Aggregates of TDP-43 are implicated in frontotemporal dementia (FTD) and recently identified

limbic-predominant age-related TDP-43 encelopathy (LATE) amongst other dementia disease states.

[3-4]

Using a

scaffold hopping approach we have synthesised small molecule candidates derived from reportedly active

compounds that are structurally distinct but contain similar pharmacophoric features. This library of compounds

has been evaluated in a range of assays for their activity against TDP-43 pathology. These results have produced

some interesting biological results and have highlighted some of the difficulties faced in CNS drug discovery.

References

1) Prince, M., et al., World Alzheimer Report 2015. The Global Impact of Dementia. An Analysis of Prevalence, Incidence,

Cost and Trends. 2015.

2) Yiannopoulou, K. G., et al., Ther. Adv. Neurol. Disord. 2013, 6 (1), 19-33.

3) Nelson, P. T., et al., Brain 2019, 142 (6), 1503-1527.

4) Mackenzie, I. R. A., et al., Curr. Opin. Neurol. 2008, 21 (6), 693-700.

EFMC-ISMC | 247

M008

TARGETING HIF-2α IN CLEAR CELL RENAL CELL CARCINOMA

WITH PROTAC TECHNOLOGY

Carlotta Cecchini (1,2), Federico Costanzo (3), Margaux Héritier (1,2), Sébastien Tardy (1,2),

Jean-Philippe Theurillat (3), Leonardo Scapozza (1,2)

1) School of Pharmaceutical Sciences, University of Geneva, Rue Michel-Servet 1, 1206 Genève2) Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva

3) Functional Cancer Genomics Group, Institute of Oncology Research, Via Vela 6, 6500 Bellinzona

Dysregulation of the Ubiquitin-Proteasome System (UPS), such as the presence of non-functional E3 ubiquitin

ligases, may result in oncogenic substrates overexpression.

(1)

In particular, the lack of VHL E3 ubiquitin ligase in

von Hippel-Lindau (VHL) disease has shown to increase cell accumulation of HIF-1α and HIF-2α transcription

factors, leading to angiogenesis and tumors’ high vascularization.

(2, 3) 

Close to 60% of patients with VHL disease

develop clear cell renal cell carcinoma (ccRCC) or renal cysts.

(4)

While the role of HIF-1α in tumorigenesis is

still debated

(5)

, the growing evidence that the accumulation of HIF-2α is promoting human ccRCC fostered the

development of HIF-2α antagonists.

(6, 7)

In this work, we aim at inducing HIF-2α knockdown by taking advantage of the promising targeted protein

degradation (TPD) approach.

(8),(9)

More specifically, we generated Proteolysis Targeting Chimeras (PROTACs)

targeting HIF-2α to induce its ubiquitylation and proteasomal degradation. Inspired by a potent HIF-2α inhibitor

(PT2385)

(10)

, we designed and synthesized the warhead of our degraders, responsible for engaging HIF-2α. In

parallel, as the HIF-2 α binding site is completely buried, we conducted molecular modeling studies to identify

linker attachment points allowing maintaining the major antagonist-target interactions and thus the binding

affinity. Polyethylene glycol (PEG) linkers have been initially selected as a linkage between the warhead and the

ligase binders, which are specific for recruiting the corresponding E3 ubiquitin ligase (CRBN and MDM2).

Multiple functional assays have been performed to assess the in vitro activity of PROTACs. In particular, both

degraders showed activity at the nanomolar range after 4h treatment in RCC cells that lack VHL. In perspective,

we plan proteomic analysis to quantitatively study protein expression in cells and we consider performing in vivostudies. So far, our PROTACs are the first and unique class of degraders targeting HIF-2α present in the

literature. Besides their applicability in hampering HIF-2α accumulation in ccRCC, they also represent a

powerful tool to investigate dysregulation of the Ubiquitin-Proteasome System in cancer.

References

1) Jara JH, Frank DD, Özdinler PH. Could dysregulation of UPS be a common underlying mechanism for cancer and

neurodegeneration? Lessons from UCHL1. Cell biochemistry and biophysics. 2013;67(1):45-53.

2) Kim E, Zschiedrich S. Renal Cell Carcinoma in von Hippel-Lindau Disease-From Tumor Genetics to Novel Therapeutic

Strategies. Front Pediatr. 2018;6:16-.

3) Clark PE. The role of VHL in clear-cell renal cell carcinoma and its relation to targeted therapy. Kidney International.

2009;76(9):939-45.

4) Rednam SP, Erez A, Druker H, Janeway KA, Kamihara J, Kohlmann WK, et al. Von Hippel–Lindau and Hereditary

Pheochromocytoma/Paraganglioma Syndromes: Clinical Features, Genetics, and Surveillance Recommendations in

Childhood. Clinical Cancer Research. 2017;23(12):e68.

5) Hoefflin R, Harlander S, Schäfer S, Metzger P, Kuo F, Schönenberger D, et al. HIF-1α and HIF-2α differently regulate

tumour development and inflammation of clear cell renal cell carcinoma in mice. Nature Communications. 2020;11(1):4111.

6) Chen W, Hill H, Christie A, Kim MS, Holloman E, Pavia-Jimenez A, et al. Targeting renal cell carcinoma with a HIF-2

antagonist. Nature. 2016;539(7627):112-7.

7) Martínez-Sáez O, Gajate Borau P, Alonso-Gordoa T, Molina-Cerrillo J, Grande E. Targeting HIF-2 α in clear cell renal

cell carcinoma: A promising therapeutic strategy. Critical Reviews in Oncology/Hematology. 2017;111:117-23.

8) Nalawansha DA, Crews CM. PROTACs: An Emerging Therapeutic Modality in Precision Medicine. Cell Chemical

Biology. 2020;27(8):998-1014.

9) Cecchini C, Tardy S, Ceserani V, Theurillat JP, Scapozza L. Exploring the Ubiquitin-Proteasome System (UPS) through

PROTAC Technology. Chimia. 2020;74(4):274-7.

10) Wehn PM, Rizzi JP, Dixon DD, Grina JA, Schlachter ST, Wang B, et al. Design and Activity of Specific

Hypoxia-Inducible Factor-2α (HIF-2α) Inhibitors for the Treatment of Clear Cell Renal Cell Carcinoma: Discovery of

Clinical Candidate ( S)-3-((2,2-Difluoro-1-hydroxy-7-(methylsulfonyl)-2,3-dihydro-1 H-inden-4-yl)oxy)-5-fluorobenzonitrile

(PT2385). Journal of medicinal chemistry. 2018;61(21):9691-721.

248 | EFMC-ISMC

M009

PROCESS OPTIMIZATION OF THE SYNTHESIS OF UAMC-00050, A

NOVEL uPA INHIBITOR

Davide Ceradini, Kirill Shubin

Aizkraukles Iela 21, Riga, Latvia

The α-aminophosphonate UAMC-00050, a newly developed trypsin-like serine protease inhibitor, has shown

promising results for the treatment of dry eye syndrome and ocular inflammation.

1

A laboratory scale synthetic

route was initially developed at University of Antwerp. Preparation of larger amounts of UAMC-00050, required

in the advanced steps of the project, proved to be difficult, due to the usage of environmentally unfriendly

solvents and hazardous reagents. A new process was developed with greener alternatives and less toxic reagents.

Every reaction was investigated in order to obtain the maximum yield, all the flash chromatography were

replaced with plug filtration and slurry purifications. The overall yield was increased from a 3% of the

discovered route to a 33% of the process development route.

References

1) Joossen, C., Baán, A., Moreno-Cinos, C. et al. A novel serine protease inhibitor as potential treatment for dry eye

syndrome and ocular inflammation. Sci Rep 10, 17268 (2020).

EFMC-ISMC | 249

M010

COMBINATION OF MEMANTINE AND 6-CHLOROTACRINE AS

NOVEL MULTI-TARGET COMPOUND AGAINST ALZHEIMER’S

DISEASE

Zofia Chrienova (1), Martina Kaniakova (2,4), Eugenie Nepovimova (1), Lenka Kleteckova (5), Kristyna

Skrenkova (4), Kristina Holubova (5), Vendula Hepnarova (6), Tomas Kucera (6), Tereza Kobrlova (3),

Karel Vales (5), Jan Korabecny (3), Ondrej Soukup (3), Martin Horak (2,4)

1) University of Hradec Kralove, Faculty of Science, Department of Chemistry, Rokitanskeho 62, 50003 Hradec Kralove,Czech Republic

2) Institute of Physiology of the Czech Academy of Sciences of the Czech Republic, Videnska 1083, 14220 Prague 4, CzechRepublic

3) Biomedical Research Centre, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic4) Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 14220 Prague 4, Czech Republic

5) National Institute of Mental Health, Topolova 748, 250 67 Klecany, Czech Republic6) Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, University of Defence, Trebesska

1575, Hradec Kralove 500 05, Czech Republic

Alzheimer´s disease as the most common form of dementia in the aging population is still opening a number of

questions in terms of its triggering mechanism. Although there are several hypotheses trying to explain the

etiology of the disease (amyloid, tau, oxidative stress, cholinergic and glutamatergic)

1–4

, none of them gives

a satisfying answer. Current pharmacotherapy is built around cholinergic and glutamatergic hypotheses using

acetylcholinesterase (AChE) inhibitors and the N-methyl-D-aspartate receptors (NMDARs) antagonist. A novel

compound combining 6-chlorotacrine (6-Cl-THA) and memantine was inspired by memagal structure (molecule

combining galantamine and memantine

5

). The design of the molecule uses a strategy known as „multi-target-

directed ligands” when two pharmacophores are combined in order to aim different pharmacological targets

6

.

Obtained molecule connects the 6-Cl-THA scaffold with memantine to support the cholinergic system and

simultaneously alleviates the burden caused by glutamate excitotoxicity. Indeed, the novel hybrid maintains high

AChE inhibitory activity and shows potent NMDARs inhibition, surpassing those of the parent 6-Cl-THA and

memantine. The hybrid molecule varies mechanism of NMDARs inhibition leading to a substance with a potent

neuroprotective effect in vivo, meanwhile having the ability to cross the blood-brain barrier via passive diffusion.

In conclusion, a single molecule with a synergistic mechanism of action offers promising results in NMDARs

mediated CNS disorders associated with cognitive decline and excitotoxicity.

This work was supported by University of Hradec Kralove (Faculty of Science, no. SV2105-2020), Ministry of

Health of the Czech Republic (no. NV19-09-00578), the project from the Czech Science Foundation (no.

16-08554S) and from European Regional Development Fund: Project "PharmaBrain" (no.

CZ.02.1.01/0.0/0.0/16_025/0007444).

References

1) Bartus RT, Dean RL, Beer B, Lippa AS. The cholinergic hypothesis of geriatric memory dysfunction. Science. 217(4558):

408–414 (1982).

2) Hardy J, Selkoe DJ. The amyloid hypothesis of Alzheimer’s disease: progress and problems on the road to therapeutics.

Science. 297(5580): 353–356 (2002).

3) Maccioni RB, Muñoz JP, Barbeito L. The molecular bases of Alzheimer’s disease and other neurodegenerative disorders.

Arch Med Res. 32(5): 367–381 (2001).

4) Swomley AM, Butterfield DA. Oxidative stress in Alzheimer disease and mild cognitive impairment: evidence from

human data provided by redox proteomics. Arch Toxicol. 89(10): 1669–1680 (2015).

5) Simoni E, Daniele S, Bottegoni G, Pizzirani D, Trincavelli ML, Goldoni L, et al. Combining galantamine and memantine

in multitargeted, new chemical entities potentially useful in Alzheimer’s disease. J Med Chem. 55(22): 9708–9721 (2012).

6) Prati F, Cavalli A, Bolognesi ML. Navigating the Chemical Space of Multitarget-Directed Ligands: From Hybrids to

Fragments in Alzheimer’s Disease. Mol Basel Switz. 21(4): 466 (2016).

250 | EFMC-ISMC

M011

A VIRTUAL SCREENING PROTOCOL TO IDENTIFY NOVEL

INHIBITORS OF GLUTAMINYL CYCLASE

Judite R.M. Coimbra (1,2), Paula I. Moreira (2,3), Armanda E. Santos (2,4), Jorge A.R. Salvador (1,2)

1) Laboratory of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal.2) Center for Innovative Biomedicine and Biotechnology (CIBB), Center for Neuroscience and Cell Biology (CNC),

University of Coimbra, 3004-504 Coimbra, Portugal.3) Laboratory of Physiology, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal.

4) Laboratory of Biochemistry and Biology, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal.

Glutaminyl Cyclase (QC) enzymes catalyze the post-translational processing of several substrates with

N-terminal glutamine or glutamate to form the pyroglutamate (pE) residue by releasing either ammonia or a

water molecule [1,2]. Emerging evidence demonstrates that human QCs play a part in pathological processes in

diverse diseases such as Alzheimer’s disease (AD) [3], inflammatory [4], and cancer diseases [5]. Thus, reducing

QC activity may achieve therapeutic effects [6-7]. This research project, which aims to discover new small

molecules that effectively inhibit QC, is focused on a reliable protocol of virtual screening (Figure 1) including i)

investigation of the QC binding site to explore the potential ligand-binding hot spots and druggability of QC

enzyme; ii) pharmacophore generation based on structural data of QC crystal complexes and the hot spots

previously identified; iii) pharmacophore-based virtual screening to quickly screen drug-like compound libraries

and molecular docking simulations to predict the binding mode and affinity of the retrieved hits. Based on this

approach, 93 high-scoring compounds were selected for a coupled-enzyme screening assay (adapted from [8]),

which resulted in novel hit-compounds with QC inhibitory activity at the micromolar level. These may represent

a starting point for further structural refinement in a hit-to-lead optimization process.

References

1) Fischer WH, Spiess J. Identification of a mammalian glutaminyl cyclase converting glutaminyl into pyroglutamyl

peptides. Proc. Natl Acad. Sci. USA. 1987;84:3628–3632.

2) Schilling S, Niestroj AJ, Rahfeld JU, et al. Identification of human glutaminyl cyclase as a metalloenzyme Potent

inhibition by imidazole derivatives and heterocyclic chelators. J Biol Chem. 2003;278:49773-49779.

3) Morawski M, Schilling S, Kreuzberger M, et al. Glutaminyl cyclase in human cortex: correlation with

(pGlu)-amyloid-beta load and cognitive decline in Alzheimer's disease. J Alzheimers Dis. 2014;39:385-400.

4) Cynis H, Hoffmann T, Friedrich D, et al. The isoenzyme of glutaminyl cyclase is an important regulator of monocyte

infiltration under inflammatory conditions. EMBO Mol med. 2011;3:545-558.

5) Logtenberg MEW, Jansen JHM, Raaben M, et al. Glutaminyl cyclase is an enzymatic modifier of the CD47- SIRPα axis

and a target for cancer immunotherapy. Nat Med. 2019;25:612-619.

6) Schilling S, Zeitschel U, Hoffmann T, et al. Glutaminyl cyclase inhibition attenuates pyroglutamate Abeta and Alzheimer's

disease-like pathology. Nat Med. 2008;14:1106-1111.

7) Coimbra JR, Sobral PJ, Santos AE, et al. An overview of glutaminyl cyclase inhibitors for Alzheimer’s disease. Future

Med Chem. 2019;11:3179-3194.

8) Schilling S, Hoffmann T, Wermann M, et al. Continuous spectrometric assays for glutaminyl cyclase activity. Anal

Biochem. 2002;303:49-56.

EFMC-ISMC | 251

M012

ANTI-LEISHMANIAL ACTIVITY OF 2-AMINO ALKANOLS AND

ALKYL-1,2-DIAMINES

de la Vega Fernández Jennifer (1), Rivas Luis (2), Gamarro Francisco (3), del Olmo Esther (1)

1) Departamento de Ciencias Farmacéuticas, Facultad de Farmacia (CIETUS, IBSAL). Universidad de Salamanca.Salamanca, Spain

2) Centro de Investigaciones Biológicas (CIB-CSIC), Madrid, Spain3) Instituto de Parasitología y Biomedicina López-Neyra (IPBLN-CSIC), Granada, Spain

Leishmaniasis is distributed worldwide infecting 12 million people, with about 1.5 million new cases each year.

The disease shows different clinical manifestations including cutaneous leishmaniasis (CL, self-healing skin

ulcers), mucosal leishmaniasis (ML), and the potentially fatal visceral leishmaniasis (VL), also known as

kala-azar. VL is caused by L. donovani or L. infantum spp, and infects 200,000–400,000 people annually, mostly

in Brazil, Sudan, Bangladesh, and in India, causing more than 20,000 deaths.

1

Chemotherapy represents the main weapon against Leishmaniasis. Recommended first line therapies include the

use of pentavalent antimonials (Sb

V

), obsolete in some endemic areas of India, amphotericine B (AmB) and

mainly its lipid formulation (Ambisome), the oral drug miltefosine and the aminoglycoside paromomycin.

However, the present chemotherapy have several drawbacks including the reduce arsenal of drugs, the cost of

treatment and the significant increase in therapeutic failure/drug resistance. In consequence, it is necessary to

search for new therapeutic strategies like drug combinations and new drugs more effectives.  Therefore, new

therapeutic hits are needed. The aim of this study was synthetized different 2-amino alkanol (type 1) and

alkyl-1,2-diamine (type 2) derivatives and to test their leishmanicidal activity against L. infantum promastigotes

and amastigotes.

Compounds type 1 and type 2 were synthesised as indicated in figure 1.

2

Twelve compounds of type 1 and ten of type 2 were obtained and tested in vitro against L. infantum, using

amphotericin B as reference drug.  Six out of twenty compounds showed EC50 below 3 microM in L.

infantum promastigotes. Further studies are currently in progress.

Acknowledgements. Financial support by Grants from MINECO-Retos, Spain: AGL2016-79813-C2-2-R), and

PID2019‐106222RB‐C32/AEI/10.13039/501100011033 are acknowledged

References

1) Leishmaniasis [World Health Organization]. Visited on June, 2021. Website:

https://www.who.int/news-room/fact-sheets/detail/leishmaniasis

2) Molecules. 2015 Jun; 20(6): 11554–11568.

252 | EFMC-ISMC

M013

DEVELOPMENT OF MULTITARGET-DIRECTED LIGANDS (MTDLS)

PROTEIN KINASES INHIBITORS TO TACKLE CENTRAL NERVOUS

SYSTEM (CNS)-RELATED DISORDERS.

Demuro Stefania (1,2), Di Martino Rita Maria Concetta (1), Ortega José Antonio (1), Sauvey Conall (3),

Shi Da (3), Tripathi Shailesh (1), Balboni Beatrice (1,2), Girotto Stefania (1), Russo Debora (4), Penna

Ilaria (4), Bandiera Tiziano (4), Ballatore Carlo (3), Abagyan Ruben (3), Cavalli Andrea (1,2)

1) Computational and Chemical Biology, Italian Institute of Technology, 16163 Genoa, Italy2) Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy

3) Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California92093, United States

4) D3 Pharma Chemistry, Italian Institute of Technology, 16163 Genova, Italy

The human kinome plays a crucial role in several pathways, and its dysregulation is associated with several

multifactorial disorders, such as cancer and neurodegenerative diseases. Among these latter, tauopaties,

pathologically defined by the presence of intracellular tau-positive inclusions and clinically characterized by

dementia and/or parkinsonism, have a drastic impact in the aging population. Nowadays, protein kinases (PKs)

have not been largely investigated in the neuroscience space, and a kinase-targeting drug has not yet been

approved to treat neurological disorders

1

. Moreover, the multifactorial nature of the CNS-related diseases has

clearly demonstrated the inadequacy of single-target drugs to achieve a therapeutic effect and has suggested the

great potential of purposely designed multi-target compounds to provide better efficacy and safer profile

compared to single-targeted ones

2

.

GSK-3β, Fyn and DYRK1A are three close-related PKs, which play a pivotal role in microtubule‐associated tau

protein hyperphosphorylation, and in turn self-aggregation and fibrillization leading to neurofibrillary

degeneration and neuronal death

3,4,5

. These may be at the basis of unmet medical needs in neurology including

Alzheimer’s disease and frontotemporal dementia. The simultaneous modulation of these drug targets by

applying the multitarget-directed ligands (MTDLs) approach could be a successful strategy to identify

disease-modifying therapies.

In this scenario, we aimed to develop the first-in-class triple inhibitors of GSK-3β, FYN and DYRK1A as

promising modulators of the abnormal tau hyperphosphorylation. We selected ARN25068 among a large set of

small molecules tested against 172 different PKs

6

.  ARN25068 has shown a promising inhibitory profile against

the three kinases. Computational studies predicted the compound binding pose at the ATP pockets of the targets.

ARN25068 binds the kinases via the typical H-bond pattern of ATP-competitive inhibitors. Microscale

thermophoresis investigations and X-Ray crystallographic studies corroborated the computational studies

highlighting the key binding features. Moreover, in vitro enzymatic assays confirmed the inhibitory potency of

ARN25068 towards GSK‐3β and Fyn in the nanomolar range and DYRK1A in the submicromolar range. Tau

phosphorylation assay on Tau0N4R-TM-tGFP U2OS stable cell line confirmed its capability to reduce the extent

of tau phosphorylation and, therefore, to promote tau and microtubule bundles.

In the light of these outcomes, ARN25068 has emerged as a promising prototype for further SAR investigations.

 Computer-aided drug design (CADD) protocols and synthetic effort are currently combined to develop novel

series of analogs with a balanced inhibiting profile against GSK-3β, FYN and DYRK1A towards innovative

chemical tools to study tauopathies.

References

1) Krahn A. I. et al. ACS Chem Neurosci 2020, 11 (13), 1871-1886.

2) Ramsay R. R. et al. Clinical and Translational Medicine 2018, 7 (1), 3.

3) Wagner U. et al. J Cell Sci 1996, 109 (Pt 6), 1537-43.

4) Tang S. J. et al. Acta Neuropathologica Communications 2020, 8 (1).

5) Guedj F. et al. Neurobiol Dis 2012, 46 (1), 190-203.

6) Metz J. T. et al. Nat Chem Biol 2011, 7 (4), 200-2.

EFMC-ISMC | 253

M014

PROTEIN-TEMPLATED SYNTHESIS DELIVERS THE FIRST

NANOMOLAR SELECTIVE ERAP2 INHIBITORS FOR

IMMUNO-ONCOLOGY

Virgyl Camberlein, Pierre Sierocki, Ronan Gealageas, Charlotte Tabey, Valentin Guillaume, Damien

Bosc, Laetitia Lesire, Sandrine Waremghem, Florence Leroux, Benoit Deprez, Rebecca Deprez-Poulain

INSERM U1177 Drugs and Molecules for Living Systems; Drug Discovery unit; Institut Pasteur de Lille, University of Lille,3 rue du Professeur Laguesse, 59000 Lille, France

Protein-templated ligand synthesis has emerged as an elegant and powerful lead- and drug-discovery strategy. It

is based on the use of the protein of interest as a vessel to catalyze the formation of its own ligands is a rather

novel concept. In the protein-templated strategy called Kinetic Target-Guided Synthesis (KTGS) (1), the

biological target accelerates an irreversible reaction between a pair of reagents by stabilizing a productive

configuration of the ternary complex. If the product is structurally similar to the transition state, its affinity for

the protein is significantly improved compared to the affinity of reagents. We previously were successful in

using KTGS for the discovery of drug-like ligands of insulin-degrading enzyme that were used as probes for invivo studies (2).

To tackle new targets for immune-oncology with small molecules (3), we got interested into the intracellular

endoplasmic reticulum aminopeptidases (ERAP1 and 2) that are zinc aminopeptidases playing a key role in the

antigen presentation pathway. These enzymes trim peptide precursors resulting from proteins degradation by the

proteasome and generate mature antigens for presentation by major histocompatibility complex class I (MHCI)

molecules. The cytotoxic T-cells recognition of the cell surface peptidome triggers immune response. Thereby

ERAPs are major regulators of adaptive immune responses acting upstream current therapeutic strategies

targeting cytokines, immune checkpoints or downstream signalling pathways.

The impact of ERAP2 polymorphic variations on NK cell activity against cancer cell lines has been shown and

low levels of ERAP2 were associated with improved response to anti-PDL1 treatments. As a result ERAP2 is a

tractable pharmacological target for modulating the cellular immunopeptidome with applications in the treatment

of cancer in a personalized approach. So far only a handful inhibitors with poor potencies, properties and

selectivities are available (4).

We report here the successful use of Kinetic Target Guided Synthesis to discover the first nanomolar,

selective and drug-like inhibitors of ERAP-2 with high potential for immuno-oncology.

 

References

1) Bosc, D., Camberlein, V., Gealageas, R., Castillo-Aguilera, O., Deprez, B., & Deprez-Poulain, R. Kinetic Target-Guided

Synthesis: reaching the age of maturity. J. Med. Chem.,2020: 3817−3833.10.1021/acs.jmedchem.9b01183

2) Deprez-Poulain R, Nathalie H, Bosc D et al.: Catalytic site inhibition of insulin-degrading enzyme by a small molecule

induces glucose intolerance in mice. Nature Comm. 2015 6: 8250. OPEN ACCESS doi: 10.1038/ncomms9250 .

3) Huck, B. R., Kötzner, L., & Urbahns, K. Small Molecules Drive Big Improvements in Immuno-Oncology Therapies.

Angewandte Chemie int. Ed.,2018, 57(16): 4412-4428.

4) Medve, L., Gealageas, R., Lam, B. V., Guillaume, V., Castillo-Aguilera, O., Camberlein, V., Rosell, M., Fleau, C.,

Warenghem, S., Charton, J., Dumont-Ryckembusch, J., Bosc, D., Leroux, F., van Endert, P., Deprez, B., & Deprez-Poulain,

R. Modulators of hERAP2 discovered by High-Throughput Screening. Eur. J. Med. Chem.,2021,

113053.10.1016/j.ejmech.2020.113053

254 | EFMC-ISMC

M015

DISCOVERY OF NOVEL SMALL MOLECULE DNA GYRASE

INHIBITORS BY STRUCTURE-BASED VIRTUAL SCREENING

Satish N. Dighe (1,2), Marina Matthew (1), Trudi A. Collet (1)

1) Innovative Medicines Group, Institute of Health & Biomedical Innovation, School of Clinical Sciences, QueenslandUniversity of Technology, Brisbane, QLD 4059, Australia.

2) Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia.

Antibiotics were considered “Super Drugs” when first discovered in the 20

th

century. Since then, they have been

used as first-line treatments for many bacterial infections. However, over the years, bacteria have developed

resistance to these antimicrobial agents, thereby making treatment of antibiotic-resistant bacterial infections

difficult. DNA gyrase is a type IIA topoisomerase, which catalyses changes in DNA during replication by

introducing negative supercoils and, hence, is a well validated drug target for antibacterial drug discovery.

Currently, fluoroquinolone and aminocoumarin classes of antibiotics act by inhibiting bacterial DNA gyrase.

However, fluoroquinolones are primarily used for gram-negative bacterial infections as the activity against

gram-positive bacteria is limited. Similarly, novobiocin, an aminocoumarin class of antibiotic initially approved

for clinical use, was later withdrawn from the market due to safety issues and a poor pharmacological profile.

Computer-aided drug design has emerged as an important tool for drug discovery within the last decade.

Therefore, in the proposed work, we utilised a structure-based virtual screening approach (SBVS) to identify

novel DNA gyrase inhibitors. In the SBVS study, the CoCoCo database containing seven million molecules was

subjected to an in-silico docking-based virtual screening workflow against DNA gyrase (Protein Data Bank

entry: 2XCR). Thirty-eight molecules were selected and evaluated for their antimicrobial activity. Of these, four

non-fluoroquinolone class of compounds were shown to elicit a minimum inhibitory concentration value

between 0.5-8 µg/mL against eight strains of bacteria including two clinical isolates of MRSA. In future, all four

lead compounds will be tested for their DNA gyrase inhibitory potential and used as a basis to develop DNA

gyrase targeted antibiotics. 

EFMC-ISMC | 255

M016

COMPUTATIONAL EVALUATION OF PROTAC-MEDIATED

PROTEIN DEGRADATION: CASE STUDIES AND RECENT

DEVELOPMENTS

Michael Drummond

Chemical Computing Group, Suite 910, 1010 Sherbrooke Street West, Montreal H3A 2R7, Canada

Targeted protein degradation utilizing bivalent small molecules (PROTACs) represents a new modality in drug

development, with numerous advantages vis-à-vis traditional inhibitor design. However, numerous challenges

exist in PROTAC development, particularly concerning the rational design of efficacious molecules. In this

presentation, multiple computational methods that enable the a priori evaluation of putative PROTAC molecules

will be discussed. Numerous case studies will be offered, where the application of these computational tools can

successfully recommend both potent PROTAC candidates and molecules to avoid. Results from scenarios across

different target proteins, E3 ligases, and PROTAC architectures will be presented.

256 | EFMC-ISMC

M017

HYDROPHOBIC LIGAND AUGMENTATION AS A STRATEGY OF

CARBONIC ANHYDRASE ISOZYME ACTIVE SITE EXPLORATION

Virginija Dudutienė (1), Asta Zubrienė (1), Visvaldas Kairys (2), Alexey Smirnov (1), Daumantas Matulis

(1)

1) Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University,Saulėtekio 7, Vilnius LT-10257, Lithuania

2) Department of Bioinformatics, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio 7, VilniusLT-10257, Lithuania

Carbonic anhydrases are involved in numerous physiological and pathological processes and are considered as

important therapeutic targets with the potential to treat a wide range of disorders. Carbonic anhydrase inhibitors

have been in clinical use for decades. However, all of them possess some drawbacks due to lack of isozyme

selectivity. A challenge in the design of selective inhibitors is related to the high number of isoforms and high

similarity of their active site pockets.

The major class of carbonic anhydrase inhibitors is aromatic compounds possessing a primary sulfonamide

group. Among them, benzenesulfonamides are the most abundant inhibitors. We have synthesized

benzensulfonamides bearing hydrophobic substituents. Variation of the substituent length and bulkiness was

performed on the benzenesulfonamide scaffold by three approaches: substituting 3,5-positions; substituting

2,4,6-positions, and extending the condensed ring system. The gradual augmentation of the inhibitor size

allowed determination of the maximal contact area and maximal size that could fit in the protein pocket. This led

to exploration of active site cavities giving new insights into the rational design of isozyme-selective inhibitors.

Furthermore, such approach could find use in drug design for other ligand-enzyme systems.

 

EFMC-ISMC | 257

M018

SYNTHESIS, ANTIMICROBIAL ACTIVITY, AND DOCKING STUDIES

OF NOVEL QUATERNARY AMMONIUM

FLUOROQUINOLONE-BASED ANTIBACTERIAL AGENTS

Joanna Fedorowicz (1), Małgorzata Morawska (1,2,3), Shella Gilbert-Girard (3), Liliana Mazur (4),

Cristina Durante-Cruz (3), Heidi Mäkkylä (3), Kirsi Savijoki (3), Adyary Fallarero (3), Päivi Tammela

(3), Jarosław Sączewski (2)

1) Department of Chemical Technology of Drugs, Medical University of Gdańsk, Al. Gen. J. Hallera 107, 80-416 Gdańsk,Poland

2) Department of Organic Chemistry, Medical University of Gdańsk, Al. Gen. J. Hallera 107, 80-416 Gdańsk, Poland3) Faculty of Pharmacy, University of Helsinki, Yliopistonkatu 4, 00100 Helsinki, Finland

4) Faculty of Chemistry, Maria Curie-Sklodowska University, Plac Marii Curie-Skłodowskiej 520-031 Lublin, Poland

Antibiotic resistance was identified by the WHO as one of the three biggest threats to human health and an

especially dangerous problem for people with a compromised immune system. Infections caused by pathogens

resistant to antimicrobial drugs lead to higher health care costs and extended hospital stays. A single drug is not

always able to adequately control the illness, therefore, a combination of medicines with different

pharmacotherapeutic profiles may be favorable. Hybrid drugs are molecules designed to act at multiple

molecular targets. Remarkably, the fluoroquinolones fused with other antibacterials constitute the most

comprehensively described hybrid compounds [1].

In our latest research, we developed the novel fluorescent fluoroquinolone hybrids featuring combined

quaternary quinolone-triazolinium moiety that exhibit antibacterial activity against various pathogens, including

biofilm-forming Pseudomonas aeruginosa. The reported derivatives displayed delayed antimicrobial resistance

development, caused a defect in DNA decatenation, and were potent DNA gyrase inhibitors comparable to the

reference drug, ciprofloxacin [2-4].

The present study aimed to assess the biological activity of new antibacterials incorporating a quaternary

ammonium group and a fluoroquinolone core to evaluate the hypothesis that a new class of hybrid drugs exhibits

an unique dual mechanism of action: perturbation of bacterial membranes due to the presence of quaternary

nitrogen atom and inhibition of bacterial type II topoisomerases caused by fluoroquinolone portion.

Novel fluoroquinolone derivatives were design and synthesized by exhaustive alkylation to give compounds

incorporating permanent positive charge on the nitrogen atom. The products were characterized by NMR, IR,

MS, X-ray crystallography, and elemental analysis. Subsequently, the obtained derivatives were screened invitro for antimicrobial activity against a panel of Gram-positive and Gram-negative bacterial strains. The most

active molecules exhibited MICs in the low micro- and nanomolar range, especially towards resistant species

and pathogens from the ESKAPE group. Molecular docking experiments revealed that all the synthesized

compounds were able to interact at the active sites of bacterial type II topoisomerases in the

fluoroquinolone-binding mode.

The Project was financed by the Polish National Agency for Academic Exchange as part of the Bekker

Scholarship Programme and Medical University of Gdańsk subsidies.

References

1) J. Fedorowicz, J. Sączewski, Modifications of quinolones and fluoroquinolones: hybrid compounds and dual-action

molecules, Monatschefte fur Chemie, 2018, 149(7), 1199-1245

2) J. Fedorowicz, J. Sączewski, A. Konopacka, K. Waleron, D. Lejnowski, K. Ciura, T. Tomašič, Ž. Skok, K. Savijoki, M.

Morawska, S. Gilbert-Girard, A. Fallarero, Synthesis and biological evaluation of hybrid quinolone-based quaternary

ammonium antibacterial agents, European Journal of Medicinal Chemistry, 2019, 179, 576-590

3) K. Ciura, J. Fedorowicz, F. Andrić, K.E. Greber, Al. Gurgielewicz, W. Sawicki, J. Sączewski, Lipophilicity determination

of quaternary (fluoro)quinolones by chromatographic and theoretical approaches, International Journal of Molecular

Sciences, 20(21), 5288

4) K. Ciura, J. Fedorowicz, H. Kapica, A. Adamkowska, W. Sawicki, J. Saczewski, Affinity of fluoroquinolone-safirinium

dye hybrids to phospholipids estimated by IAM-HPLC, Processes, 8(9), 1148

258 | EFMC-ISMC

M019

ORGANOSELENIUM BASED COMPOUNDS AS POTENT INHIBITORS

OF BACTERIAL UREASE

Saurabh Loharch, Wojciech Tabor, Łukasz Berlicki

Department of Bioorganic Chemistry, Faculty of Chemistry, Wrocław University of Science and Technology, WybrzeżeWyspiańskiego 27, 50-370 Wrocław, Poland

Organoselenium compounds have received significant attention in medicinal chemistry owing to their wide

spectrum of biological activity [1]. Several Se-containing compounds have been reported for their potential

applications as enzyme inhibitors, antioxidants, immunomodulators, and antitumor, anti-inflammatory and

anti-infective agents [1, 2]. Herein, we report the discovery of novel organoselenium compounds as inhibitors of

Sporosarcina pasteurii and Helicobacter pylori ureases, which is in line with our previous study [3]. The urease

enzymes are the principal virulence factor of many microorganisms that facilitates the colonization and infection

by increasing the pH at the target site [4]. This increase in pH leads to numerous health complications in

humans, such as occurrence of urinary stones, pyelonephritis, gastritis, ammonia encephalopathy, hepatic coma,

peptic ulceration, and catheters blocking [5, 6]. Consequently, drug discovery efforts have led to discovery of

various classes of urease inhibitors including phosphoramides, hydroxamic acids, heterocyclic compounds,

quinones, heavy metal ions, and thiols [7]. Unfortunately, the most of existing urease inhibitors exhibit

unfavourable features, such as relatively low hydrolytic stability or toxicity. Therefore, it is highly desirable to

design and develop novel urease inhibitors with drug-like characteristics: oral administration, wide therapeutic

window and chemical stability. However, the design of urease inhibitors is tricky and challenging as the active

site is very small and thereby restricts the available space for structural modifications in inhibitor design. An

effective strategy is to target the conserved Cysteine residue (Cys-322 in S. pasteurii urease and Cys-321 in H.pylori urease) present at the entrance of active site, making it inaccessible, thereby, enabling effective inhibition

of the enzyme. As the organoselenium compounds are known for their cysteine-binding properties [8], we

profiled a series of diselenides (Se-Se) with varying scaffolds. We envisage that the ‘Se-Se’ will interact with the

conserved Cys residue and the varying scaffolds might add to the specificity of the inhibitors. We were able to

identify 15 diselenide based inhibitors with high activity. Further, the study was complemented by whole

cell-based assays to demonstrate the bactericidal activity of identified inhibitors.

References

1) Mangiavacchi, F., et al., Sweet Selenium: Synthesis and Properties of Selenium-Containing Sugars and Derivatives.

Pharmaceuticals (Basel), 2020. 13(9).

2) Singh, F.V. and T. Wirth, Chapter 3 Synthesis of Organoselenium Compounds with Potential Biological Activities, in

Organoselenium Compounds in Biology and Medicine: Synthesis, Biological and Therapeutic Treatments. 2018, The Royal

Society of Chemistry. p. 77-121.

3) Macegoniuk, K., et al., 1,2-Benzisoselenazol-3(2H)-one Derivatives As a New Class of Bacterial Urease Inhibitors. J Med

Chem, 2016. 59(17): p. 8125-33.

4) Rutherford, J.C., The emerging role of urease as a general microbial virulence factor. PLoS Pathog, 2014. 10(5): p.

e1004062.

5) Mora, D. and S. Arioli, Microbial urease in health and disease. PLoS Pathog, 2014. 10(12): p. e1004472.

6) Konieczna, I., et al., Bacterial urease and its role in long-lasting human diseases. Curr Protein Pept Sci, 2012. 13(8): p.

789-806.

7) Kafarski, P. and M. Talma, Recent advances in design of new urease inhibitors: A review. J Adv Res, 2018. 13: p.

101-112.

8) Ganther, H.E., Selenium metabolism, selenoproteins and mechanisms of cancer prevention: complexities with thioredoxin

reductase. Carcinogenesis, 1999. 20(9): p. 1657-66.

EFMC-ISMC | 259

M020

FRAGMENT BASED DESIGN OF MYCOBACTERIAL THIOREDOXIN

REDUCTASE INHIBITORS: FROM A FRAGMENT SCREENING TO

NOVEL INHIBITORS.

Friederike Füsser (1,2,4), Jan Wollenhaupt (3), Manfred Weiss (3), Daniel Kümmel (4), Oliver Koch (1,2)

1) Institute of Pharmaceutical and Medicinal Chemistry, University of Münster2) German Center of Infection Research, University of Münster3) Macromolecular Crystallography, Helmholtz-Zentrum Berlin

4) Institute of Biochemistry, University of Münster

The resurgence of tuberculosis, caused primarily by Mycobacterium tuberculosis (Mtb), and the appearance of

multi-drug and extensively drug resistant strains leads to an urgent need for new antituberculotic drugs with

alternative modes of action. As part of the thioredoxin system the thioredoxin reductase (TrxR) is essential for

thiol redox homeostasis [1]. The mycobacterial TrxR shows a substantial difference in sequence, mechanism and

structure to eukaryotic TrxRs leading to the expectation that the mycobacterium tuberculosis TrxR is a selective

and promising target for a tuberculosis treatment. The druggability was already shown with a compound class

derived from a docking-based virtual screening approach [2].

For the identification of new fragment-based starting points and for the investigation of new potential interaction

sites for new drugs, a crystallographic fragment screening was performed. TrxR crystals that reproducibly

showed high-resolution diffraction (~1.7 Å) were soaked with the 96 structurally diverse fragments of the

F2X-Entry Screen[3]. The diffraction data were collected at the MX beamlines at BESSY II [4] and were

processed and refined by a largely automated software pipeline including the FragMAXapp and a multi dataset

analysis approach called PanDDa [5,6]. 40 fragments were found bound to nine binding sites, of which four sites

are positioned at binding pockets or important interaction sites and therefore show high potential for possible

inhibition. The identified binding sites and fragments represent interesting starting points for the development of

new and potent TrxR inhibitors.

Further studies will include a detailed analysis of all fragments and their binding sites. Promising fragments will

be used for virtual screening including a substructure search and docking to gain structurally optimised

compounds. A selection of these will be tested and characterised in biochemical and biophysical assays and will

be used for further crystallisation to determine the binding conformations.

References

1) K. Lin, K. M. O'Brien, C. Trujillo, R. Wang, J. B. Wallach, D. Schnappinger, S. Ehrt, PLoS pathogens 2016, 12,

e1005675.

2) O. Koch, T. Jäger, K. Heller, P. C. Khandavalli, J. Pretzel, K. Becker, L. Flohé, P. M. Selzer, Journal of medicinal

chemistry 2013, 56, 4849.

3) J. Wollenhaupt, A. Metz, T. Barthel, G. M. A. Lima, A. Heine, U. Mueller, G. Klebe, M. S. Weiss, Structure (London,

England : 1993) 2020, 28, 694-706.e5.

4) U. Mueller, R. Förster, M. Hellmig, F. Huschmann, A. Kastner, P. Malecki, S. Pühringer, M. Röwer, K. Sparta, M.

Steffien, M. Ühlein, P. Wilk & *M. S. Weiss*, Eur. Phys. J. Plus 2015 ,130, 141-150.

5) G. M. A. Lima, E. Jagudin, V. O. Talibov, L. S. Benz, M. Costantino, T. Barthel, J. Wollenhaupt, M. S. Weiss and U.

Mueller, Acta crystallographica. Section D, Biological crystallography 2021, D77, 799-808

6) Pearce, N., Krojer, T., Bradley, A. et al. A multi-crystal method for extracting obscured crystallographic states from

conventionally uninterpretable electron density. Nat Commun 2017, 8, 15123.

260 | EFMC-ISMC

M021

DETERMINATION OF TRPA1 CHANNEL BINDING SITE FOR

METHYLXANTHINE ANTAGONISTS USING MOLECULAR

MODELING APPROACH

Alicja Gawalska, Adam Bucki, Marcin Kołaczkowski

Department of Medicinal Chemistry, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland

TRPA1 is a transmembrane nonselective cation channel, one of the most promising and studied targets in

preclinical pharmaceutical research, especially in the context of chronic and acute pain and respiratory diseases

[1,2]. Its whole structure has already been experimentally resolved, but the binding site of TRPA1 antagonists,

e.g. HC-030031, a model methylxanthine derivative, remains unknown.

The aim of the study was to determine the potential binding site of xanthine antagonists and to describe the

binding mode of their representative, compound HC-030031, using molecular modeling approach.

To this end, an approach using the available molecular modeling tools predicting binding sites of biological

targets, e.g. Sitemap, MetaPocket 2.0, DeepSite, CASTp 3.0 was applied. The TRPA1 channel model was

prepared using a properly optimized 6PQQ three dimensional atomic structure provided by cryo-EM [3]. The

obtained results were analyzed in terms of scoring functions, pocket size and frequency of occurrence.

Compound HC-030031 was docked to the selected binding pockets using Induced Fit Docking procedure, and

the obtained HC-030031-TRPA1 complexes served as a source of conformational models of the protein, and

were further tested using retrospective virtual screening and molecular dynamics simulations.

The research showed that HC-030031 binds to a pocket formed by the TRP-like domain and the pre-S1, S4, S5

helices of one subunit and establishes crucial interactions: hydrogen bond with Asn-855 and π-π stacking with

Trp-711, which might play an equally important role in recognition of other xanthine derivatives and their

bioisosteres.

These results allow for a more specific search for new potential TRPA1 antagonists in the group of xanthine

derivatives as well as their bioisosteres and hence new potential drugs for the treatment of neuropathic pain,

asthma, and COPD. This study represents also the first attempt to determine the binding site and describe

interactions of HC030031 - the approach that brings together reports on site-directed mutagenesis and the

latest cryo-EM structure of the other antagonist bound to TRPA1.

The study was financially supported by the National Science Centre, Poland (grant no. 2020/37/N/NZ7/02365)

and Jagiellonian University Medical College (grant no. N42/DBS/000187)

References

1) Logashina, Y. et al., TRPA1 Channel as a Regulator of Neurogenic Inflammation and Pain: Structure, Function, Role in

Pathophysiology, and Therapeutic Potential of Ligands, Biochemistry Mosc. 2019, 84 (2), 101–118

2) Moran, M. et al., Transient Receptor Potential Channels as Therapeutic Targets. Nat Rev Drug Discov 2011, 10 (8),

601–620

3) Suo Yang et al., Structural Insights into Electrophile Irritant Sensing by the Human TRPA1 Channel, Neuron, 105, 5,

2020, 882-894

EFMC-ISMC | 261

M022

KINETIC EVALUATION OF SULFUR(VI) FLUORIDE COVALENT

WARHEADS TO ENABLE THE RAPID DEVELOPMENT OF

THERAPEUTICS

Katharine Gilbert (1,2), Aini Vuorinen (1,3), Katrin Rittinger (3), Glenn Burley (2), Jacob Bush (1,3)

1) Chemical Biology, GlaxoSmithKline, Medicines Research Centre, Stevenage, SG1 2NY2) Pure and Applied Chemistry, University of Strathclyde, Thomas Graham Building, Glasgow, G1 1XL

3) The Francis Crick Institute, 1 Midland Road, London, NW1 1AT

Expanding the range of biological systems that can be investigated and targeted by chemical biology approaches

is becoming increasingly crucial to the development of novel therapeutics. Covalent warheads capture specific

compound-target interactions, so can be leveraged as tools to facilitate the discovery and development of new

medicines.

1,2

Established warhead functionalities, including photoreactive and cysteine-reactive moieties, hold

limitations that result in limited applicability across the proteome. Sulfur(VI) fluorides have emerged as attractive

covalent warheads, due to the ability to target numerous nucleophilic amino acids and achieve quantitative

modification, thus expanding the druggable proteome.

3

Sulfur(VI) fluoride (S

VI

-F) warheads are inherently

electrophilic and so are susceptible to hydrolysis, forming an inactive sulfonic acid species. Thus, it is crucial to

strike a balance between stability and reactivity of S

VI

-F warheads prior to application.

Here, we present a biologically relevant evaluation of S

VI

-F warhead reactivity, and build understanding of the

parameters that influence the modification of targets by covalent tools (Figure 1).

 

Solution studies on fragments containing various S

VI

-F warheads revealed a vast range of intrinsic reactivities

and aqueous stabilities, reinforcing the need for informed warhead selection. Further, computational LUMO

energy calculations displayed excellent correlation, enabling the future prediction of warhead reactivity.

To achieve biologically-relevant insight, the S

VI

-F warheads were appended to small molecule binders for

anchorage within protein targets. Intact protein LCMS was utilised to measure time-dependent protein

modification yields and conduct kinetic profiling, to investigate the factors that drive covalent modification.

We further demonstrated the cell-compatibility of S

VI

-F warheads and conducted chemoproteomic studies with

warhead-substituted kinase probes in live cells. The proteome-wide engagement of the probes correlated with the

warhead intrinsic reactivity, while multiple warheads exhibited unique selectivity for specific protein targets.

 

Based on these initial studies it is envisaged that S

VI

-F warheads will be a valuable addition to the reactive tools

strategy. The use of S

VI

-F warheads expands the ligandable proteome and thus assists the development of novel

medicines.

References

1) K. M. Backus, B. E. Correia, K. M. Lum, S. Forli, B. D. Horning, G. E. González-Páez, S. Chatterjee, B. R. Lanning, J. R.

Teijaro, A. J. Olson, D. W. Wolan and B. F. Cravatt, Nature, 2016, 534, 570–574.

2) E. Grant, D. Fallon, M. Hann, K. Fantom, C. Quinn, F. Zappacosta, R. Annan, C. Chung, P. Bamborough, D. Dixon, P.

Stacey, D. House, V. Patel, N. C. O. Tomkinson and J. Bush, Angew. Chem. Int. Ed., 2020, 58, 17322–17327.

3) E. C. Hett, H. Xu, K. F. Geoghegan, A. Gopalsamy, R. E. Kyne, C. A. Menard, A. Narayanan, M. D. Parikh, S. Liu, L.

Roberts, R. P. Robinson, M. A. Tones and L. H. Jones, ACS Chem. Biol., 2015, 10, 1094–1098.

ABSTRACT CANCELLED

262 | EFMC-ISMC

M023

KINETIC EVALUATION OF SULFUR(VI) FLUORIDE COVALENT

WARHEADS TO ENABLE THE RAPID DEVELOPMENT OF

THERAPEUTICS

Katharine Gilbert (1,2), Aini Vuorinen (1,3), Katrin Rittinger (3), Glenn Burley (2), Jacob Bush (1,3)

1) Chemical Biology, GlaxoSmithKline, Medicines Research Centre, Stevenage, SG1 2NY2) Pure and Applied Chemistry, University of Strathclyde, Thomas Graham Building, Glasgow, G1 1XL

3) The Francis Crick Institute, 1 Midland Road, London, NW1 1AT

Expanding the range of biological systems that can be investigated and targeted by chemical biology approaches

is becoming increasingly crucial to the development of novel therapeutics. Covalent warheads capture specific

compound-target interactions, so can be leveraged as tools to facilitate the discovery and development of new

medicines.

1,2

Established warhead functionalities, including photoreactive and cysteine-reactive moieties, hold

limitations that result in limited applicability across the proteome. Sulfur(VI) fluorides have emerged as

attractive covalent warheads, due to the ability to target numerous nucleophilic amino acids and achieve

quantitative modification, thus expanding the druggable proteome.

3

Sulfur(VI) fluoride (S

VI

-F) warheads are

inherently electrophilic and so are susceptible to hydrolysis, forming an inactive sulfonic acid species. Thus, it is

crucial to strike a balance between stability and reactivity of S

VI

-F warheads prior to application.

 

Here, we present a biologically relevant evaluation of S

VI

-F warhead reactivity, and build understanding of the

parameters that influence the modification of targets by covalent tools (Figure 1).

Solution studies on fragments containing various S

VI

-F warheads revealed a vast range of intrinsic reactivities

and aqueous stabilities, reinforcing the need for informed warhead selection. Further, computational LUMO

energy calculations displayed excellent correlation, enabling the future prediction of warhead reactivity.

To achieve biologically-relevant insight, the S

VI

-F warheads were appended to small molecule binders for

anchorage within protein targets. Intact protein LCMS was utilised to measure time-dependent protein

modification yields and conduct kinetic profiling, to investigate the factors that drive covalent modification.

We further demonstrated the cell-compatibility of S

VI

-F warheads and conducted chemoproteomic studies with

warhead-substituted kinase probes in live cells. The proteome-wide engagement of the probes correlated with the

warhead intrinsic reactivity, while multiple warheads exhibited unique selectivity for specific protein targets.

 

Based on these initial studies it is envisaged that S

VI

-F warheads will be a valuable addition to the reactive tools

strategy. The use of S

VI

-F warheads expands the ligandable proteome and thus assists the development of novel

medicines.

 

References

1) K. M. Backus, B. E. Correia, K. M. Lum, S. Forli, B. D. Horning, G. E. González-Páez, S. Chatterjee, B. R. Lanning, J. R.

Teijaro, A. J. Olson, D. W. Wolan and B. F. Cravatt, Nature, 2016, 534, 570–574.

2) E. Grant, D. Fallon, M. Hann, K. Fantom, C. Quinn, F. Zappacosta, R. Annan, C. Chung, P. Bamborough, D. Dixon, P.

Stacey, D. House, V. Patel, N. C. O. Tomkinson and J. Bush, Angew. Chem. Int. Ed., 2020, 58, 17322–17327.

3) E. C. Hett, H. Xu, K. F. Geoghegan, A. Gopalsamy, R. E. Kyne, C. A. Menard, A. Narayanan, M. D. Parikh, S. Liu, L.

Roberts, R. P. Robinson, M. A. Tones and L. H. Jones, ACS Chem. Biol., 2015, 10, 1094–1098.

EFMC-ISMC | 263

M024

THE DESIGN, PREPARATION AND SCREENING OF BRPF1

DEGRADERS

Daniel Glynn (1), Rosie Crampton (1), Thomas Pesnot (1), Anne-Chloe Nassoy (1), Ralph Kirk (1), Daniele

Narducci (1), Andrew Scott (2), Gary Nelson (1), Lynette Ongeri (1), Matilda Bingham (1), Rhoanne

McPherson (2), Darryl Turner (2), Justyna Rzepecka (2)

1) Concept Life Sciences, Frith Knoll Rd, Chapel-en-le-Frith, High Peak SK23 0PG, United Kingdom2) Concept Life Sciences, Edinburgh BioQuarter, 9 Little France Road, Edinburgh, EH16 4UX, United Kingdom

Given the recent promising clinical results, PROteolysis TArgeting Chimeras (PROTACs) are attracting great

attention both from academia and industry. PROTACs regulate protein function by degradation instead of

inhibition and their novel chemical knockdown approach is perfect for the validation of new and emerging

biological targets.

Bromodomain and PHD finger-containing protein 1 (BRPF1) a multivalent chromatin regulator possessing three

histone-binding domains, is involved in the epigenetic regulation of gene expression and is implicated in

neurodevelopmental disorders. Brpf1 has been shown to form a stable complex with Moz-Tif2, which can lead

to the development of human acute myeloid leukemia (AML). The role of BRPF1 in carcinogenesis however

still remains elusive and warrants additional investigation.

To understand the role of BRPF1 in cancer disease biology we designed and synthesised a range of BRPF1

degraders (based on previously reported inhibitors (1)) in a highly desirable physicochemical space utilising

in-silico modelling. The prepared PROTACs which were designed to elicit the recruitment of a range of E3

ligases were evaluated as BRPF1 degraders in THP-1 cells. Finally, an in-vitro ADMET profiling was carried

out to evaluate the suitability of these degraders for in-vivo target validation.

References

1) R. J. Watson, ACS Med. Chem. Lett. 2016, 7, 552–557

264 | EFMC-ISMC

M025

SYNTHESIS OF SMALL MOLECULE INHIBITORS OF

CYCLOPHILIN D AND THEIR IN VITRO EVALUATION FOR

NEUROPROTECTIVE TREATMENT OF ALZHEIMERS DISEASE

Annamaria Haleckova, Ondrej Benek, Michaela Vaskova, Lucie Zemanova, Kamil Musilek

University of Hradec Kralove, Faculty of Science, Department of Chemistry, Hradec Kralove, Czech Republic;*[email protected]

Mitochondrial enzyme cyclophilin D (CypD) is associated with mitochondrial dysfunction and apoptosis of

neuronal cells and plays a pivotal role in the formation and opening of the mitochondrial permeability transition

pore (mPTP). Several recent studies have found that CypD promotes the formation of the mPTP upon binding to

amyloid-β (Aβ) peptide inside brain mitochondria, suggesting that neuronal CypD has a potential to be a

promising therapeutic target in Alzheimer's disease.

1

Compounds able to influence the enzyme by inhibiting its

activity or its interaction with Aβ may prevent mitochondrial destruction, however, only limited number of

CypD inhibitors are currently available - mostly cyclosporin A derivatives, which serve only limited purposes

due to their undesirable properties.

1

Dynamic development of much more preferable selective small molecule

inhibitors is expected in this regard since they can be readily modified and optimized to have desired properties

as possible drug candidates. We have synthesized series of compounds based on the previously published

structural scaffold

2,3,4

(Fig. 1) being particularly focused on resolving the optimal configuration on the two chiral

centers. 

References

1) Park, I. et al. Discovery of non-peptidic small molecule inhibitors of cyclophilin D as neuroprotective agents in

Aβ-induced mitochondrial dysfunction. J Comput Aided Mol Des, 31 (10), 929–941 doi: 10.1007/s10822-017-0067-9

(2017).

2) Ahmed-Belkacem, A. et al. Fragment-based discovery of a new family of non-peptidic small-molecule cyclophilin

inhibitors with potent antiviral activities. Nat. Commun. 7:12777 doi: 10.1038/ncomms12777 (2016).

3) Shore, E. R. et al. Small molecule inhibitors of cyclophilin D to protect mitochondrial function as a potential treatment for

acute pancreatitis. Journal of Medicinal Chemistry 59 (6), 2596-2611 doi: 10.1021/acs.jmedchem.5b01801 (2016).

4) De Simone, A. et al. A computationally designed binding mode flip leads to a novel class of potent tri-vector cyclophilin

inhibitors. Chem. Sci. 10, 542–547 doi: 10.1039/c8sc03831g (2019).

EFMC-ISMC | 265

M026

PROTEIN DEGRADER BUILDING BLOCKS FOR STREAMLINED

SYNTHESIS OF HETEROBIFUNCTIONAL DEGRADER LIBRARIES

Kaelyn Kilke

Senior Product ManagerEmerging Chemical Synthesis

MilliporeSigmaA business of Merck KGaA, Darmstadt, Germany

Targeted protein degradation is a novel strategy that uses small molecules to hijack endogenous proteolysis

systems to degrade disease-relevant proteins, thus reducing their abundance in cells. Not only is this of interest

as a research tool to study the impact of selective protein knockdowns, but it has quickly been adopted by the

global drug discovery community for its advantages over occupancy-based inhibition and drugging proteins

traditionally intractable to small molecules. Heterobifunctional protein degraders contain a target-binding

warhead on one end and an E3 ubiquitin ligase-targeting ligand on the other, connected by a linker in the middle.

While their potential for modular assembly is appealing, the design and synthesis of degraders remains an

empirical and chemistry-intensive process that explores varied permutations of E3 ligand derivatives, exit

vectors, linker diversity, and target warheads for optimal ternary complex formation and target degradation. To

accelerate access to degrader libraries for initial screening and degrader hit identification, we have developed a

toolbox of building blocks that simplify library generation from one starting target warhead. This suite of

degrader templates contains strategic combinations of E3 ligands for CRBN, VHL, IAP, and RNF4; exit vectors;

and diverse linkers ranging in length, flexibility, and conjugation chemistry. Molecules showing targeted

degradation in cells are starting points for further lead optimization studies.

References

1) Schlesiger, S.; Toure, M.; Wilke, K. E.; Huck, B. R. Accelerating the Discovery of Next-Generation Small-Molecule

Protein Degraders. Aldrichimica Acta, 2019, 52, 35–49.

266 | EFMC-ISMC

M027

RAPID HIT DISCOVERY WITH DNA-ENCODED LIBRARY

TECHNOLOGY USING DyNAbind® KITS

John Fetter

Merck KGaA, 3050 Spruce St., Saint Louis, MO 63103, USA

[email protected]

DNA-encoded libraries have revolutionized hit and lead discovery, enabling researchers to simultaneously

screen large libraries of compounds against their protein target of interest. Though powerful, the barrier to entry

for this technology has been high, requiring either a robust, in-house DEL technology platform, or an expensive,

milestone-driven service agreement with an outside party. In collaboration with DyNAbind, Merck KGaA has

introduced two off-the-shelf DELs, granting access to this powerful technology for a fraction of the usual cost.

We are offering two kits for screening: one for fragment screening and the second a 10 million compound

standard library. Fragment-based screening offers a way to find hits for challenging targets. The fragment kit is

based on dynamic fragment screening technology developed at DyNAbind, in which a fragment binding pair

becomes enriched during selection from >370,000 possible pairs. Also, the small library enables the QC of each

fragment in the kit, improving the overall quality of the results. The 10 million compound library offers larger

more drug-like molecules to provide hits farther along in the development process than when starting with

fragments. After PCR and nextgen sequencing by the user, data can be loaded on our free and secure portal that

reveals the structures for the top hits. Learn more about these kits and the science behind it.

EFMC-ISMC | 267

M028

DEVELOPMENT OF A FLUORESCENCE REPORTER ASSAY TO

STUDY BACTERIAL TYPE III SECRETION SYSTEM

(T3SS)-MODULATING COMPOUNDS

Julia Hotinger (1,2), Heather Pendergrass (1), Adam Johnson (1), Aaron May (1)

1) Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA, (USA)2) [email protected]

Enteropathogenic Escherichia coli (EPEC) is a major cause of infantile diarrhea worldwide. EPEC and the

closely related murine model of EPEC infection, Citrobacter rodentium, utilize a type III secretion system

(T3SS) to propagate infection.

[1]

Inhibiting or activating the T3SS with a therapeutic could treat the infection

without causing harm to commensal bacteria and have lower selective pressure than traditional antibiotics,

reducing the rate of resistance formation. Studying modulation of the T3SS usually requires a BSL-2 laboratory

designation and eukaryotic host cells while giving no indication of the mechanism of action.

[1,2]

We have

designed and developed a BSL-1 assay that does not require mammalian cell culture using an enzyme

carboxypeptidase G2 (CPG2) expression system in C. rodentium for screening potential T3SS modulators.

[3]

[image 1]

CPG2 was conjugated to the secretion tag of an EPEC T3SS effector protein, EspF, and expressed on a pBAD

plasmid under arabinose (ARA) induction to allow for CPG2’s secretion through the T3SS. After secretion,

CPG2 cleaves the yellow substrate, Glu-CyFur (Figure 2, 1), to release a purple and fluorescent CyFur (λ

ex

=

563 nm, λ

em

= 610 nm, Figure 1, 2). We screened known T3SS inhibitors to validate this assay, including

(-)-epigallocatechin-3-gallate (EGCG), regacin, and aurodox.

[4]

We then screened multiple natural product

libraries, upwards of 1,000 compounds total. One important discovery was that the known quorum sensing

inhibitor tannic acid (TA), a natural product found in red wines, is a potent T3SS inhibitor with an IC

50

of 0.65 ±

0.09 μM.

[4]

[image 2]

The incomplete T3SS induction in our system allows us a unique opportunity to observe T3SS activation.

Multiple compounds with similar structural characteristics (structures not shown for IP purposes) were found to

dose-dependently increase the activity of the T3SS (representative compounds E3, F5, & F6. Figure 2). These

compounds also showed little to no cytotoxicity, increasing the likelihood of T3SS activation. Further

experimentation to validate this result and determine their mechanism of action is ongoing.

References

1) Fasciano, A.C.; Shaban, L.; Mecsas, J. Promises and challenges of the type three secretion system injectisome as an

antivirulence target. EcoSal Plus 2019, 8, 10.

2) Yount, J.S.; Tsou, L.K.; Dossa, P.D.; Kullas, A.L.; van der Velden, A.W.M.; Hang, H.C. Visible fluorescence detection of

type III protein secretion from bacterial pathogens. J. Am. Chem. Soc. 2010, 132, 8244–8245.

3) Pendergrass, H., Johnson, A., Hotinger, J., May A. Fluorescence detection of type III secretion using a Glu-CyFur reporter

system in Citrobacter rodentium. Microorganisms 2020, 8, 1953.

4) Pendergrass, H.A.; May, A.E. Natural product type III secretion system inhibitors. Antibiotics 2019, 8, 162.

268 | EFMC-ISMC

M029

A STUDY OF THE EFFECT OF COMMON

ELECTRON-WITHDRAWING GROUPS ON THE

PHYSICOCHEMICAL PROPERTIES OF

2-AMINOTETRAHYDROPYRIDINE BACE1 INHIBITORS

Meng-Yang Hsiao, Frederik Rombouts, Ann Vos, Harrie Gijsen

Janssen Research & Development, Turnhoutseweg 30, B-2340 Beerse.

A common challenge for medicinal chemists is to reduce the pK

a

of strongly basic groups’ conjugate acids into a

range that preserves the desired effects, usually potency and/or solubility, but avoids undesired effects like high

volume of distribution (V

d

), limited membrane permeation, and off-target binding to, notably, the hERG channel

and monoamine receptors. We faced this challenge with a 2-aminotetrahydropyridine scaffold harboring an

amidine, a key structural component of potential inhibitors of BACE1, the rate-limiting enzyme in the

production of Aß species that make up amyloid plaques in Alzheimer’s disease. In our endeavor to balance

potency with desirable properties to achieve brain penetration, we introduced a diverse set of groups in beta

position of the amidine that modulate logD, PSA and pK

a

. Given the synthetic challenge to prepare these highly

functionalized warheads, we first developed a design flow including docking and predicted physicochemical

parameters which allowed us to select only the most promising candidates for synthesis. For this we evaluated a

set of commercial packages to predict physicochemical properties, which can guide medicinal chemists in their

endeavors to modulate pK

a

values of amidine and amine bases.

References

1) Hsiao, C. C.; Rombouts, F.; Gijsen H. J. M. New evolutions in the BACE1 inhibitor field from 2014 to 2018. Bioorg.

Med. Chem. Lett. 2019, 29, 761-777.

ABSTRACT CANCELLED

EFMC-ISMC | 269

M030

ONE RING TO RULE THEM ALL: THE INDOLE IS INDISPENSABLE

FOR ANTITRYPANOSOMAL PAULLONES

Irina Ihnatenko (1,2), Marco J. Müller (1,2), Diego Benítez (3), Cecilia Ortíz (3), Estefanía Dibello (3),

Marcelo A. Comini (3), Conrad Kunick (1,2)

1) Institut für Medizinische und Pharmazeutische Chemie, Technische Universität Braunschweig, Beethovenstraße 55, 38106Braunschweig, Germany

2) Center of Pharmaceutical Engineering (PVZ), Technische Universität Braunschweig, Franz-Liszt-Straße 35A, 38106Braunschweig, Germany

3) Laboratory Redox Biology of Trypanosomes, Institut Pasteur de Montevideo, Mataojo 2020, 11400 Montevideo, Uruguay

Parasites from the genera Trypanosoma and Leishmania1

cause certain neglected tropical diseases. As treatment

is scarce, new strategies to combat these diseases are required, including chemotherapeutic approaches. In our

approach, the molecular target is a vital enzyme of the parasites’ unique trypanothione-based thiol redox

metabolism: the trypanothione synthetase (TryS)

2

. Previous work has shown that N5

-substituted paullones like

MOL2008 display antitrypanosomal activity as well as TryS inhibition

3

.

We aimed to investigate how the paullone scaffold can be modified to improve activity, selectivity and aqueous

solubility. The novel compounds’ design is partly based on molecular docking studies on TryS homology

models. Different series of compounds were obtained with significant modifications, including replacement of

the indole and variation of the N5

side chain. Minor changes included substitutions in the 3- and 9-position of the

paullone scaffold and the amine functional group in the sidechain. We tested the activity of the synthesised

compounds against the infective stage of Trypanosoma brucei brucei and recombinant TryS from T. brucei. On

the one hand, alternative 9-substituents and bulkier residues in the sidechain led to sustained antitrypanosomal

activity. On the other hand, major modifications such as molecule downsizing negatively impacted the biological

activity, rendering the indole moiety indispensable for a compound to be active.

References

1) World Health Organization. Integrating neglected tropical diseases into global health and development: Fourth WHO

report on neglected tropical diseases; World Health Organization, Geneva, 2017.

2) Torrie, L. S.; Wyllie, S.; Spinks, D.; Oza, S. L.; Thompson, S.; Harrison, J. R.; Gilbert, I. H.; Wyatt, P. G.; Fairlamb, A.

H.; Frearson, J. A. J. Biol. Chem. 2009, 284, 36137–36145.

3) a) Benítez, D.; Medeiros, A.; Fiestas, L.; Panozzo-Zenere, E. A.; Maiwald, F.; Prousis, K. C.; Roussaki, M.;

Calogeropoulou, T.; Detsi, A.; Jaeger, T.; Šarlauskas, J.; Peterlin Mašič, L.; Kunick, C.; Labadie, G. R.; Flohé, L.; Comini,

M. A. PLoS Neglected Trop. Dis. 2016, 10, e0004617; b) Orban, O. C. F.; Korn, R. S.; Benítez, D.; Medeiros, A.; Preu, L.;

Loaëc, N.; Meijer, L.; Koch, O.; Comini, M. A.; Kunick, C. Bioorg. Med. Chem. 2016, 24, 3790–3800;

270 | EFMC-ISMC

M031

THE APPLICATION OF COMPUTER-AIDED DRUG DESIGN IN THE

SEARCH FOR CARBONYL REDUCTASE 1 (CBR1) INHIBITORS AS

COMPOUNDS SUPPORTING CANCER CHEMOTHERAPY.

Marek Jamrozik (1), Adam Bucki (1), Michał Sapa (1), Kamil Piska (2), Paulina

Koczurkiewicz-Adamczyk (2), Elżbieta Pękala (2), Marcin Kołaczkowski (1)

1) Department of Medicinal Chemistry, Jagiellonian University Medical College, Medyczna 9 st, Kraków, Poland,2) Department of Pharmaceutical Biochemistry, Jagiellonian University Medical College, Medyczna 9 st, Kraków, Poland

The search for effective anticancer drugs is one of the biggest challenges of modern pharmacotherapy.

Anthracycline antibiotics (ANT) are still among the most widely used group of anticancer drugs. Unfortunately,

ANT metabolism, which consists in the two-electron reduction of a carbonyl moiety to a hydroxy group,

performed mainly by carbonyl reductase 1 (CBR1), leads to the formation of metabolites with decreased activity.

Additionally, it is postulated that those metabolites are responsible for the cardiotoxic effect that significantly

limits the use of ANT. Inhibition of CBR1-related ANT metabolism can lead to improvement of the

pharmacological action of this group of drugs [1,2].

This study presents the application of computational methods in the optimization of CBR1 crystal structures

(PDB codes 1WMA and 3BHJ) to improve their ability to effectively distinguish CBR1 ligands from

non-ligands. Optimized models were then used in prospective virtual screenings leading to a selection of new

chemotypes of potential CBR1 inhibitors.

Optimization of CBR1 crystals was performed using Induced-fit docking, with 14 CBR1 ligands. The obtained

models were validated in retrospective virtual screening. Their quality was assessed using two parameters:

BEDROC

α=20

and EF

1%

. Good-quality models were additionally checked in 20 ns molecular dynamics

simulations. The best models were used in prospective virtual screening with over 2 million compounds.

Models with higher BEDROC

α=20

and EF

1%.

values, comparing to non-optimized crystals, were obtained as a

result of optimization of CBR1 crystals. Models 1WMA_27 (BEDROC

α=20

: 0.690; EF

1%

: 18) and 3BHJ_11

(BEDROC

α=20

: 0.826; EF

1%

: 63) have been selected to prospective virtual screening. Both models were stable

during 20 ns molecular dynamics simulations (based on the measurement of the distances between the ligand and

CBR1 Tyr193 residue, as well as between ligand and cofactor (NADPH) molecule). After the virtual screening,

ADMET properties of 52 compounds were predicted using ADMET Predictor software. 4 most promising

compounds (considering both docking simulations and ADMET properties prediction) were purchased and

submitted for in vitro research.

 

Acknowledgements:

This study was funded by Jagiellonian University Medical College (N42/DBS/000188).

References

1) Minotti G. et al.; Chem Res Toxicol. 2000 Dec;13(12):1336-41,

2) Olson RD. et al.; Proc Natl Acad Sci U S A. 1988 May;85(10):3585-9.

EFMC-ISMC | 271

M032

SYNTHESIS AND BIOLOGICAL EVALUATION OF NOVEL

INDOLE-BASED DERIVATIVES AS COMPETITIVE AND SELECTIVE

HUMAN MONOAMINE OXIDASE B INHIBITORS

Ahmed Elkamhawy (1,2), Jiyu Woo (1), Noha Goud (1), Joo Roh (3,4), Jungsook Cho (1), Ki Duk Park

(4,5), Kyeong Lee (1)

1) College of Pharmacy, Dongguk University-Seoul, Goyang 10326, Republic of Korea2) Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt3) Chemical Kinomics Research Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea4) Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul 02792,

Republic of Korea5) Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and

Technology (KIST), Seoul 02792, Republic of Korea6) KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul 02447, Republic of Korea

Since there is no disease-modifying treatment has been discovered yet for Parkinson’s disease (PD). On the basis

of the recent patent literatures, novel selective MAO inhibitors have been developed for new therapeutic

opportunities such as cancer, hair loss, muscle dystrophies, cocaine addiction and inflammation along with the

classical therapeutic window as promising therapeutically active candidates for PD patients. In recent years, we

report rational design and synthesis of a new series of Indole-based final compounds as potential human MAO-B

(hMAO-B) selective inhibitors. Out of twenty-four synthesized derivatives, five compounds exhibited inhibition

more than 70% over MAO-B at a single dose of 10uM. A molecular modeling study was performed to explain

the binding mode of the new series to MAO-B binding site. More detailed biological results will be presented.

272 | EFMC-ISMC

M033

TARGETED THERANOSTIC AGENTS FOR THE TREATMENT OF

GLIOBLASTOMA MULTIFORME

Jiney Jose (1), Peter J. Choi (1), Elizabeth Cooper (2), Thomas I.-H. Park (2,3), William A Denny (1),

Mike Dragunow (2,3), Richard Faull (3), Edward Mee (3,4), Patrick Schweder (3,4), Katsuya Noguchi (5)

1) Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Private Bag 92019,Auckland 1142, New Zealand

2) Department of Pharmacology & The Centre for Brain Research, University of Auckland, Private Bag 92019, Auckland1142, New Zealand

3) Neurosurgical Research Unit, The Centre for Brain Research, University of Auckland, Private Bag 92019, Auckland 1142,New Zealand

4) Department of Neurosurgery, Auckland City Hospital, Private Bag 92024, Auckland 1142, New Zealand5) Dojindo Laboratories Co., Ltd, Techno-Research Park Tabaru 2025-5 Mashiki-machi, Kamimashiki-gun, 861-2202,

Japan

We describe the synthesis of drug-dye conjugate 1 between anaplastic lymphoma kinase inhibitor Crizotinib and

heptamethine cyanine dye IR-786. The drug-dye conjugate 1 was evaluated in three different patient-derived

adult and paediatric glioblastoma cell lines and showed potent cytotoxic activity with nanomolar potency (EC50:

50.9 nM). We also demonstrate evidence for antiproliferative activity of 1 with single digit nanomolar potency

(IC50: 4.7 nM). Furthermore, the cytotoxic effects conveyed a dramatic, 110-fold improvement over Crizotinib.

This improvement was even more pronounced (492-fold) when 1 was combined with Temozolomide, the

standard drug for treatment for glioblastoma. This work lays the foundation for future exploration of similar

tyrosine kinase inhibitor drug-dye conjugates for the treatment of glioblastoma.

References

1) Choi, P. J.; Cooper, E.; Schweder, P.; Mee, E.; Faull, R.; Denny, W. A.; Dragunow, M.; Park, T. I.; Jose, J., The synthesis

of a novel Crizotinib heptamethine cyanine dye conjugate that potentiates the cytostatic and cytotoxic effects of Crizotinib in

patient-derived glioblastoma cell lines. Bioorg Med Chem Lett 2019, 29, 2617-2621.

EFMC-ISMC | 273

M034

HIT-OPTIMIZATION USING TARGET-DIRECTED DYNAMIC

COMBINATORIAL CHEMISTRY

Ravindra P. Jumde (1), Melissa Guardigni (2), Robin M. Gierse (1,3,4), Alaa Alhayek (1,3), Di Zhu (1,3,4),

Zhoor Hamid (1,3), Sandra Johannsen (1,3), Walid A. M. Elgaher (1), Philipp J. Neusens (1,3), Christian

Nehls (5), Jörg Haupenthal (1), Norbert Reiling (5,6), Anna K. H. Hirsch (1,3)

1) Department of Drug Design and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) –Helmholtz Centre for Infection Research (HZI), Campus Building E8.1, 66123 Saarbrücken, Germany

2) D3-PharmaChemistry, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy3) Department of Pharmacy, Saarland University, Campus Building E8.1, 66123 Saarbrücken, Germany

4) Stratingh Institute for Chemistry, University of Groningen, Nijenborgh 7, 9747 AG Groningen, The Netherlands5) RG Biophysics, Research Center Borstel, Leibniz Lung Center, Borstel, Germany

6) RG Microbial Interface Biology, Research Center Borstel, Leibniz Lung Center, Borstel, Germany7) German Center for Infection Research (DZIF), Partner site Hamburg-Lübeck-Borstel-Riems, Borstel, Germany

Antimicrobial resistance is emerging as a serious threat to global public health.

1,2

 Current therapeutics are

becoming increasingly ineffective as drug resistance spreads globally. The current slow speed of drug

development and disinvestment of the pharmaceutical industry in the field of anti-infectives is aggravating this

problem. In this alarming situation, innovative strategies for drug development and novel targets for which

inhibitors with an unprecedented mode of action can be developed are urgently required. Our study aims to use

the target‑directed dynamic combinatorial chemistry (tdDCC) for identification and multiparameter optimization

of ligands for underexplored anti-infective target 1-deoxy-D-xylulose-5-phosphate synthase (DXPS).

We report the use of tdDCC to first identify and subsequently optimize binders/inhibitors of the target DXPS.

The initial hits were also optimized for their antibacterial activity against E. coli and M. tuberculosis during

subsequent tdDCC runs. Using tdDCC, we were able to generate acylhydrazone-based inhibitors of DXPS. The

tailored tdDCC runs also provided insights into this novel class of DXPS inhibitors' structure–activity

relationship. Furthermore, the competition tdDCC runs provided important information about the mode of

inhibition of acylhydrazone-based inhibitors. This approach holds the potential to expedite the drug discovery

process and apply to various biological targets.

3

 

References

1) A. H. Fairlamb, et al., Nat. Microbiol. 2016, 1, 16092

2) L. Munkholm, O. Rubin, Globalization and Health, 2020, 16, 109

3) R. P. Jumde, M. Guadigni, R. M. Gierse, A. Alhayek, D. Zhu, Z. Hamid, S. Johannsen, W. A. M. Elgaher, P. J. Neusens,

C. Nehls, J. Haupenthal, N. Reiling and A. K. H. Hirsch, Chemical Science, 2021, 12, 7775

274 | EFMC-ISMC

M035

DEVELOPING DIARYLIMIDAZOLES AS ANTIBIOTICS USING AN

OPEN SOURCE APPROACH

Dana Klug (1), Edwin Tse (1), Alexandra Vaideanu (1), Ireno Demmangngewa (1), Paul Stapleton (1),

Álvaro Lorente Macías (2), Chris Swain (3), Anthony Sama (4), Andreas Schätzlein (1), William Zuercher

(2), Lee Graves (2), Susan Charman (5), Lori Ferrins (6), Matthew Todd (1)

1) UCL School of Pharmacy, 29-39 Brunswick Square, London WC1N 1AX, UK2) UNC Eshelman School of Pharmacy, 301 Pharmacy Lane, CB#7355, Chapel Hill, NC 27599-7355, USA

3) Cambridge Med Chem Consulting, 8 Mangers Lane, Duxford, Cambridge CB22 4RN, UK4) Email: [email protected]

5) Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University (ParkvilleCampus), Parkville, VIC 3052, Australia

6) Northeastern University Department of Chemistry and Chemical Biology, 360 Huntington Ave, Boston, MA 02115, USA

Antimicrobial resistance (AMR) poses an increasing threat to global public health.

1

However, development of

new antibiotics has slowed, and despite a clear and increasingly urgent medical need, large pharmaceutical

companies have been pulling out of antibiotic drug development, and small-to-medium-sized entities that have

successfully brought new antibiotics to market have subsequently gone bankrupt.

2

It is clear, therefore, that the

current incentives are not enough to solve this problem. The failure of traditional market forces to foster

innovation in antibiotic drug development presents an opportunity for the use of an open source approach

encompassing maximal data and resource sharing to efficiently deliver a public good.

2

To that end, the Open

Source Antibiotics (OSA) consortium

3

has been created. We here describe the open source drug discovery

approach, using OSA’s hit-to-lead development of a series of diarylimidazoles to treat methicillin-resistant S.aureus (MRSA) infection as an example. Starting with a small array of compounds which were found to have

activity against MRSA when screened by the Community for Open Antimicrobial Drug Discovery (CO-ADD),

the OSA consortium have elucidated detailed structure-activity relationships of this chemotype, as well as

determined sites of metabolism and performed preliminary investigations into the mechanism of action.

 

References

1) Hernando-Amado, S. et al. Defining and combating antibiotic resistance from One Health and Global Health perspectives.

Nature Microbiol. 2019, 4: 1432-1442. https://doi.org/10.1038/s41565-019-0503-9

2) Klug, D. et al. There is no market for new antibiotics: this allows an open approach to research and development [version

1; peer review: 1 approved]. Wellcome Open Res. 2021, 6: 146. https://doi.org/10.12688/wellcomeopenres.16847.1

3) Open Source Antibiotics, available at https://github.com/opensourceantibiotics

EFMC-ISMC | 275

M036

MACROLIDE INSPIRED MACROCYCLES AS MODULATORS OF

THE IL-17A/IL-17RA INTERACTION

Sanja Koštrun (1), Andreja Fajdetić (1), Dijana Pešić (1), Karmen Brajša (1), Vlatka Bencetić Mihaljević

(1), Dubravko Jelić (1), Adriana Petrinić Grba (1), Ivaylo Elenkov (1), Renata Rupčić (1), Samra Kapić

(1), Ivana Ozimec Landek (1), Kristina Butković (1), Ana Grgičević (1), Dinko Žiher (1), Ana Čikoš (1),

Jasna Padovan (1), Gordon Saxty (1), Kevin Dack (2), Hakan Bladh (2), Tine Skak-Nielsen (2), Simon

Feldbaek Nielsen (2), Maja Lambert (2), Martin Stahlhut (2)

1) Fidelta Ltd, Prilaz baruna Filipovića 29, 10000 Zagreb, Croatia2) LEO Pharma A/S, Industriparken 55, 2750 Ballerup, Denmark

Interleukin 17 (IL-17) cytokines promote inflammatory pathophysiology in many autoimmune diseases,

including psoriasis, multiple sclerosis, rheumatoid arthritis and inflammatory bowel disease.

1

Such broad

involvement of IL-17 in various autoimmune diseases makes it an ideal target for drug discovery. Psoriasis is a

chronic inflammatory disease characterized by numerous defective components of the immune system.

2,3

Significantly higher levels of IL-17A have been noticed in lesions of psoriatic patients, if compared to non-lesion

parts.

4

IL-17A/IL-17 RA interaction surface is, as other protein-protein interactions, characterized with hot-spots

spread over the wide and flat protein surface. Such interaction sites are mainly intractable with traditional small

molecule drugs. This has resulted in efforts to exploit the chemical space of natural products and macrocyclic

molecules.

5

Presented work is focused on the macrolide inspired macrocycles as potential IL-17A/IL-17RA modulators and

covers the molecular design, synthesis, and in vitro profiling. Macrocycles are prepared using FideltaMacro

technology which is consisting of coupling the macrolide fragments with variety of natural or synthetic

fragments ranging from amino-acids known to interact with PPI hot-spots, different heterocycles, alkyl

substituents or sugars.

6

Macrocycles, as potential IL-17A modulators, are designed to diversify and enrich

chemical space through different ring sizes, stereochemistry combinations and side-chains modifications

resulting in the variety of three-dimensional shapes. Structural insights into possible binding interactions have

been used to design and prioritize target molecules. Inhibitors in the nM range were identified in both target

based and phenotypic assays. In vitro ADME as well as in vivo PK properties will be discussed.

This work contributes to the body of literature demonstrating that preparation of diverse synthetic and

semi-synthetic macrocyclic compounds as modulators of challenging protein-protein interactions is an effective

strategy to bridge the space between biologicals and small molecules.

References

1) Brembilla, N. C.; Senra, L.; Boehncke, W.-H. The IL-17 Family of Cytokines in Psoriasis: IL-17A and Beyond. Front.

Immunol. 2018, 9, 1682.

2) Parisi, R.; Symmons, D. P. M.; Griffiths, C. E. M.; Ashcroft, D. M.; Identification and Management of Psoriasis and

Associated ComorbidiTy (IMPACT) project team. Global Epidemiology of Psoriasis: A Systematic Review of Incidence and

Prevalence. J. Inv. Derm. 2013, 133(2), 377-385.

3) World Health Organization. WHO Global report on Psoriasis; Geneva, Switzerland, 2016.

4) Blauvelt, A.; Chiricozzi, A. The Immunologic Role of IL-17 in Psoriasis and Psoriatic Arthritis Patho-genesis. Clin. Rev.

Allerg. Immunol. 2018, 55(3), 379-390.

5) Dougherty PG et al. Macrocycles as protein-protein interaction inhibitors. Biochem J 2017; 474(7):1109-1125.

6) Fajdetić, A.; Matanović Škugor, M.; Elenkov, I.; Kragol, G.; Bukvić, M.; Marušić Ištuk, Z.; Koštrun, S.; Žiher, D.; Rupčić,

R.; Butković, K., Dukši M.; Ozimec Landek, I.; Pešić, D.; Hutinec, A.; Mesić, M.; Saxty, G.; Poljak, V. Preparation of seco

macrolide ami-noglycosides for use in the treatment of prophylaxis. Int. Pat. Appl. WO2017194452A1, 2017.

276 | EFMC-ISMC

M037

DESIGN, SYNTHESIS, AND DEVELOPMENT OF NEUROSTEROIDS

AS THERAPEUTICS FOR CENTRAL NERVOUS SYSTEM DISEASES

Eva Kudova

Institute of Organic Chemistry and Biochemistry of the Czech Academy of SciencesFlemingovo namesti 2, 16610 Prague 6, Czech Republic

Neurosteroids (NS) are endogenous steroids that are synthesized from cholesterol in the brain. NS produce rapid

effects on neuronal excitability and synaptic function. The central effects of these compounds can be mediated

by interactions with various ligand-gated ion channels or voltage-gated ion channels.

1

We design and synthesize

NS as modulators of NMDA,

2,3,4

GABA

A

/glycine,

5,6

and P2X receptors.

7,8

Then, their ADME/Tox profile is

evaluated.

9

Selected compounds are further tested in vitro on a particular biological target in collaboration with

the Institute of Physiology, Academy of Sciences of the Czech Republic. The “hit molecules” are tested in vivoin various models of neuropathic pain and seizure diseases. The results of our studies on NS are covered by

patent applications and patents.

10

This work was supported by the Technology Agency of the Czech Republic: Czech National Centres of

Competence, project “PerMed” Personalized Medicine – Diagnostics and Therapy TN01000013 and by the

Academy of Sciences of the Czech Republic (AS CR) – grant RVO 61388963.

References

1) Kudova E.: Neurosci. Lett. 2021, 750, 135771.

2) Kudova E. et al.: J. Med. Chem. 2015, 58(15), 5950-66.

3) Slavikova B. et al.: J. Med. Chem. 2016, 59(10), 4727-4739.

4) Krausova B. et al.: J. Med. Chem. 2018, 61, 4505-4516.

5) Bukanova J.V. et al.: Neurochem. Int. 2018, 118, 145-151.

6) Bukanova J.V. et al.: Front. Mol. Neurosci. 2020, 13, 14.

7) Sivcev S. et al.: J. Neurochem. 2019, 150, 28-43.

8) Sivcev S. et al.: J. Steroid Biochem. Mol. Biol. 2020, 202, 105725.

9) Matousova M. et al.: Steroids 2019, 147, 4-9.

10) Kudova et al.: PCT/CZ2020/050017 (04/02/2020); Kudova et al.: US 10,017,535 (07/10/2018); Vyklicky L. and Kudova

E.: CZ 307648 (01/30/2019).

EFMC-ISMC | 277

M038

EXPLOITING DNA-ENCODED LIBRARY TECHNOLOGY FOR THE

DISCOVERY OF NOVEL ANTIBODY RECRUITING MOLECULES

AGAINST LOX-1

Katherine Macfarlane (1,4), Svetlana Belyanskaya (2), Carol Mulrooney (2), Eric Shi (2), Sopheary Op

(2), Jeff Messer (2), Yao Chen (3), Matthew Sender (3), Thomas Clohessy (4), Craig Jamieson (1)

1) Department of Pure and Applied Chemistry, University of Strathclyde, Thomas Graham Building, 295 Cathedral Street,Glasgow, UK

2) Encoded Library Technology Department, GlaxoSmithKline, 200 Cambridge Park Drive, 6th Floor, Cambridge, MA02140, USA

3) Chemical Biology Department, GlaxoSmithKline, 1250 S Collegeville Rd, Collegeville, PA 19426, USA4) Medicinal Chemistry Department, GlaxoSmithKline, Gunnels Wood Road, Stevenage, SG1 2NY, UK

Bifunctional small molecules, such as Proteasome Targeting Chimeras (PROTACs),

1

Lysosome Targeting

Chimeras (LYTACs),

2

and Antibody Recruiting Molecules (ARMs),

3

are used increasingly in medicinal

chemistry, both as tool compounds and potential therapeutics, due to their ability to induce protein-protein

interactions and influence biology.  Known small molecule ligands can be excellent starting points for the design

of bifunctional molecules, but many proteins of interest do not yet have known binders.  Moreover, most

traditional drug discovery techniques were developed for the identification of monovalent small molecule drugs,

and are not necessarily well suited to bifunctional drug discovery.  The choice and creative use of drug discovery

platforms can therefore be critical to success.

This work involves the identification and synthesis of novel ARMs against Lectin-type Oxidized LDL Receptor

1 (LOX-1) based on a DNA-encoded library selection.  This technology enables simultaneous screening of

billions of compounds against the target protein in a single experiment.

4

  Unlike a phenotypic assay, it identifies

all binders regardless of activity, which is highly complementary to the ARM modality: non-functional binders

may still make functional ARMs.  Even more conveniently, each molecule in the DNA-encoded library is

attached to a DNA “barcode” for identification, meaning a suitable linker trajectory is already established for

every hit.

SAR-driven analysis of the LOX-1 selection data narrowed 13,000 hits to 17 compounds for off-DNA synthesis,

and a machine learning model was applied to identify potential binders within the GSK collection.  Off-DNA

compounds were synthesised with a functional handle at the position used for DNA conjugation, facilitating

ARM synthesis in a single step.  Eight compounds were confirmed as binders by Affinity Selection Mass

Spectrometry (ASMS).  The fully elaborated novel ARMs will now be progressed to quantitative binding and

cell killing assays, the ultimate goal being selective killing of Polymorphonuclear Myeloid-Derived Suppressor

Cells (PMN-MDSCs).  This could be highly therapeutically beneficial, given the immune suppressive role of

PMN-MDSCs in many cancers, and their strong association with poor checkpoint inhibitor response.

5

References

1) Gu, S.; Cui, D.; Chen, X.; Xiong, X.; Zhao, Y. Bioessays 2018, 40, e1700247.

2) Spiegel, D.; Muthusamy, V.; McDonald, D.; Gong, A.; Sabbasani, V.; Chirkin, E.; Branham, E.; Swartzel, J.; Ray, J.;

Zhang, M.; Caianiello, D. Bifunctional Small Molecules That Mediate the Degradation of Extracellular Proteins, Preprint

2020.

3) Achilli, S.; Berthet, N.; Renaudet, O. RSC Chem. Biol. 2021, Advance Article

4) Madsen, D.; Azevedo, C.; Micco, I.; Petersen, L. K.; Hansen, N. J. V. Prog. Med. Chem. 2020, 59, 181.

5) Yang, Z.; Guo, J.; Weng, L.; Tang, W.; Jin, S.; Ma, W. J Hematol. Oncol. 2020, 13, 10.

278 | EFMC-ISMC

M039

NOVEL BENZENESULFONATE SCAFFOLDS WITH A HIGH

ANTICANCER ACTIVITY AND G2/M CELL CYCLE ARREST IN

GLIOBLASTOMA MULTIFORME

Katarzyna Malarz (1), Jacek Mularski (2), Michal Kuczak (1,2), Anna Mrozek-Wilczkiewicz (1), Robert

Musiol (2)

1) A. Chelkowski Institute of Physics and Silesian Centre for Education and Interdisciplinary Research, University of Silesiain Katowice, 75 Pulku Piechoty 1a, 41-500 Chorzow, Poland

2) Institute of Chemistry, University of Silesia in Katowice, 75 Pulku Piechoty 1a, 41-500 Chorzów, Poland

Sulfonates, unlike their derivatives, sulphonamides, have rarely been investigated for their anticancer activity.

Unlike the well-known sulphonamides, esters are mainly used as convenient intermediates in synthesis [1]. Here,

we present the first in-depth investigation of quinazoline sulfonates. A small series of derivatives were

synthesized and tested for their anticancer activity. Based on their structural similarity, these compounds

resemble tyrosine kinase inhibitors and the p53 reactivator CP-31398 [2]. The compounds had a good level of

antiproliferative activity and selectivity in a panel of cancer cell lines. Especially, we noticed the high anticancer

activity against leukemia and glioblastoma. Among synthesized compounds, the tosylate of

7-chloro-2-styrylquinazoline appeared to be the most active at a sub-micromolar level and was considerably

higher than both of the positive control drugs. Regardless of their structural similarity to the p53 reactivator and

kinase inhibitors, the activity of the tested sulfonates seems to be unrelated to these targets. However, some of

the dependence on the p53 status that was observed can be explained as a signal of a multitargeted action. The

anticancer activity of tosylates corresponded with a strong cell cycle arrest in GBM and mitotic inhibition similar

or higher than that of paclitaxel. Moreover, the cell cycle arrest may be possibly, if not exclusively, associated

with the interaction with microtubules. Our studies exhibited that tested tosylates caused a significant

enhancement of tubulin polymerization. Interestingly, the observed effect was much stronger than that of the

well-known enhancer of this process, paclitaxel. Apoptosis and autophagy were confirmed as the cell death

modes that corresponded with the inhibition of metabolic activity and the activation of the p53-dependent and

p53-independent pathways. Namely, there was a strong activation of the p62 protein and GADD44. Other

proteins such as cdc2 were also expressed at a higher level [3]. It can therefore be concluded that the sulfonates

of quinazolines can be regarded as promising scaffolds for developing anticancer agents. Additionally, this can

be hypothesized that strong G2/M inhibitors may be effectively used for overcoming drug resistance or adverse

effects as is known for paclitaxel.

 

This work was financed by the National Science Centre grant 2019/35/B/NZ5/04208 (K.M.).

References

1) Elder, D.P.; Delaney, E.; Teasdale, A.; Eyley, S.; Reif, V.D.; Jacq, K.; Facchine, K.L.; Oestrich, R.S.; Sandra, P.; David,

F. The utility of sulfonate salts in drug development. J. Pharm. Sci. 2010, 99, 2948–2961.

2) Mularski, J.; Malarz, K.; Pacholczyk, M.; Musiol, R. The p53 stabilizing agent CP-31398 and multi-kinase inhibitors.

Designing, synthesizing and screening of styrylquinazoline series. Eur. J. Med. Chem. 2019, 163, 610–625.

3) Malarz, K.; Mularski, J.; Kuczak, M.; Mrozek-Wilczkiewicz, A.; Musiol, R. Novel Benzenesulfonate Scaffolds with a

High Anticancer Activity and G2/M Cell Cycle Arrest. Cancers 2021, 13, 1790.

EFMC-ISMC | 279

M040

LIGAND IDENTIFICATION FROM NON-ENCODED CHEMICAL

MATTER USING AFFINITY SELECTION MASS SPECTROMETRY

Renaud Prudent, Hugues Lemoine, Glorianne Jouravel, Christophe Nicoud, Jean-Yves Ortholand, Didier

Roche

Edelris, 60 Avenue Rockefeller, 69008 Lyon, France

High throughput screening (HTS) has been the main driver for hit generation since its introduction in the

mid-1980s. Pharma companies have routinely relied upon screening decks of 0.5-3 million compounds, at a

throughput of 1 x 10

6

compounds/day.

1

Although HTS has contributed to many approved drugs, alternative

paradigms aiming at improving R&D productivity are slowly emerging. Notably Fragment Based Drug

Discovery

2

(FBDD) and DNA-Encoded Library Technology (DELT)

3

 have opened up new possibilities to

address targets that failed to deliver using traditional HTS. DELT is becoming particularly attractive due to the

recent emergence of PROTACs

4

resulting in a quest for the identification of binders versus functional hits. Yet,

the DELT approach requires a strong effort in post screening deconvolution and the chemical optimization of

identified ligands is hampered by the reaction conditions initially used for DNA library generation.

Developed in the mid-1990s but not broadly used by global drug-discovery organizations until the last decade,

affinity selection mass spectrometry (AS-MS) has become an important HTS technique for drug discovery.

As a

binding assay, AS-MS enables the identification of novel chemical entities with diverse mechanisms of action.

The hits may serve as allosteric inhibitors or activators, may stabilize inactive enzyme conformations or be

warheads for targeted protein degradation. We will illustrate why AS-MS is powerful technique to explore a

broad chemical space and identify small-molecule tools and lead compounds that bind new and challenging

biological targets. AS-MS is shifting from being a simple screening alternative to traditional HTS or DELT to

being a firmly established approach in chemical biology.

References

1) R. Macarron, M.N. Banks, D. Bojanic, D.J. Burns, D.A. Cirovic, T. Garyantes, D.V. Green, R.P. Hertzberg, W.P. Janzen,

J.W. Paslay, U. Schopfer, G.S. Sittampalam, "Impact of high-throughput screening in biomedical research". Nat. Rev. Drug.

Discov. 2011, 10, 188–195.

2) D. A. Erlanson, R. S. McDowell, T. O'Brien , "Fragment-Based Drug Discovery" J. Med. Chem. 2004, 47, 3463-3482.

3) R. M. Franzini, L.H. Yuen, "Achievements, Challenges, and Opportunities in DNA-Encoded Library Research: An

Academic Point-of-View", ChemBioChem, 2017, 18, 829-836.

4) M. Toure, CM.Crews, "Small-Molecule PROTACS: New Approaches to Protein Degradation". Angew. Chem. Int. Ed.

Engl. 2016, 55, 1966-1973.

5) R. Prudent, D. A. Annis, P. J. Dandliker, J.Y. Ortholand and D. Roche, "Exploring new targets and chemical space with

affinity selection-mass spectrometry”.Nature Reviews Chemistry, 2020

280 | EFMC-ISMC

M041

METATACS: A STRATEGY FOR METASTASIS PREVENTION

THROUGH TARGETED FASCIN DEGRADATION

Sarah Memarzadeh (1), Nikki Paul (2), Laura Machesky (2), Justin Bower (2), David J. France (1)

1) School of Chemistry, University of Glasgow, Glasgow2) Beatson Institute, Cancer Research UK, Glasgow

The lifetime risk to develop cancer is estimated to be around 50%, and – although survival rates have vastly

improved over the last few decades – cancer remains the second leading cause of death worldwide.

1

Metastasisis the major contributor to cancer mortality, and describes the ability of cancer cells to detach from the primary

tumour, invade the body and colonise at a distal site. Fascin, a structural protein which is overexpressed inaggressive metastatic cancers with poor survival rates, is integral to the formation of actin-rich membrane

protrusions (invadopodia) necessary for cancer cells to invade and migrate through tissue (Figure 1). Inhibition

of fascin's actin-bundling activity has been shown to dramatically reduce tumour cell invasion and formation of

metastases.

2

Targeted protein degradation induced by PROTACs is a promising strategy that could overcome the challenges

of drugging a structural protein and disrupting its protein–protein interaction encountered with traditional

small-molecule therapeutics. These bifunctional molecules are designed to bring fascin in close proximity with

an E3 ligase, inducing ubiquitination of the pro-metastatic target, and tagging it for selective degradation by the

proteasome. Depletion of fascin levels by degradation and the catalytic nature of the PROTAC are likely to

results in a prolonged therapeutic response whilst minimising risk of side effects.

3

 

Given the difficulty of developing small molecule ligands for structural proteins, HaloPROTACs were chosen as

proof-of-concept for selective fascin degradation via the PROTAC mechanism. Instead of binding through a

ligand, HaloPROTACs attach to HaloTag fusion proteins covalently.4 Accordingly, a series of compounds was

designed and synthesised (Figure 2a), and a HaloTag-fascin fusion protein was cloned and expressed in different

cancer cell lines. Dose-response and time-course assays showed degradation of HaloTag-fascin by up to 80%with as little as 0.1 µM HaloPROTAC concentration. This decrease was observed as early as 4 hours after

treatment, with sustained degradation beyond 72 hours (Figure 2b).

 

The impact of selectively degrading endogenous fascin on metastasis is evaluated through functional and

phenotypic assays. Failure to form the invasive structures in the absence of fascin could inhibit cancer cell

invasion altogether, paving the way for the development of anti-metastatic PROTACs (MetaTACs) able to

sustainably reduce and prevent metastasis in cancer patients.

 

References

1) CRUK, https://www.cancerresearchuk.org/health-professional/cancer-statistics/survival/all-cancers-combined (accessed

June 2021).

2) A. Li et al, Curr Biol, 2010, 20, 339-345.

3) A. C. Lai and C. M. Crews, Nat Rev Drug Discov, 2017, 16, 101-114.

4) G. V. Los et al, ACS Chem Biol, 2008, 3, 373-382.

EFMC-ISMC | 281

M042

QUINOLONYL DKA DERIVATIVES DISRUPT THE INTERACTION

BETWEEN V-RNA AND INTEGRASE OF HIV-1

Antonella Messore (1), Alessandro De Leo (1), Davide Ialongo (1), Luigi Scipione (1), Mamuka

Kvaratskhelia (2), Roberto Di Santo (1), Roberta Costi (1)

1) Istituto Pasteur-Fondazione Cenci Bolognetti, Dipartimento di Chimica e Tecnologie del Farmaco, Sapienza University ofRome, P. le Aldo Moro 5, 00185 Rome, Italy

2) Division of Infectious Diseases, University of Colorado School of Medicine, Aurora, United States

AIDS remains one of the most important global health challenges, especially in sub-Saharan Africa.

Antiretroviral therapy (ART) comprises a multitarget regimen combining antiviral drugs with orthogonal

mechanisms of action. Although HAART is purposely devised to overcome the high viral genetic variability and

the subsequent emergence of resistance, multi-drug resistant strains can still be detected in some patients and

long-term drug toxicities still represent an unresolved concern in HIV management [1]. HIV integrase (IN) is a

nucleic-acid-processing enzyme present in retroviruses that is critical for viral propagation, and it is an important

therapeutic target. Integrase strand transfer inhibitors (INSTIs) which bind to the active site of the viral enzyme,

have proven to be highly effective, becoming a potent first-line therapy to treat infected patients. On the other

hand, allosteric inhibitors of IN (termed ALLINIs) are a promising new class of antiretroviral agents, which are

not yet a clinically approved therapeutic option, but represent a relevant scientific area of interest for anti-HIV

research. These inhibitors act differently in respect to INSTIs, in fact, they interfere with viral maturation in the

late steps of the infection, exhibiting only modest activity in the early steps. They induce a higher order

multimerization of IN and they impair the correct IN-vRNA interaction which is fundamental for the generation

of fully competent virions. As a result, defective viral particles with greatly reduced infective potential are

produced and mis-localization of the vRNA outside the viral capsid is generally observed [2]. We therefore

decided to deepen the mechanism of action of a small subset of quinolyl based compounds belonging to our

in-house library, previously screened against IN. We assessed the capability of our derivatives to inhibit at low

micromolar concentrations both the IN 3’-processing (3’-P) and strand transfer (ST) reactions in a LEDGF/p75

independent assay. In addition, we performed in vitro binding assays, and we found that our quinolonyl

derivatives are able to disrupt the IN-vRNA interaction, that is vital for a correct generation of a functional

infective virion. In conclusion, we identified quinolonyl derivatives endowed with a unique mechanism of action

based on the interference on both 3’-P and ST steps of the IN activity, and the ability to hamper the IN-vRNA

interaction during the late steps of HIV life cycle. The discovery of this unique mechanism of action can pave

the way for the development of a new inhibition strategy against IN.

References

1) R. Costi, M. Métifiot, S. Chung, G. Cuzzucoli Crucitti, K. Maddali, L. Pescatori, A. Messore, V.N. Madia, G. Pupo, L.

Scipione, S. Tortorella, F.S. Di Leva, S. Cosconati, L. Marinelli, E. Novellino, S.F.J. Le Grice, A. Corona, Y. Pommier, C.

Marchand, R. Di Santo, J. Med. Chem. 2014, 57(8), 3223–3234.

2) P.C. Koneru, A.C. Francis, N. Deng, S.V. Rebensburg, A.C. Hoyte, J. Lindenberg, D. Adu-Ampratwum, R.C. Larue, M.F.

Wempe, A. N. Engelman, D. Lyumkis, J.R. Fuchs, R.M. Levy, G.B. Melikyan, M. Kvaratskhelia, eLife. 2019, e46344.

282 | EFMC-ISMC

M043

IMMOBILIZATION OF ZINC(II) PHTHALOCYANINES ON

GRAPHENE OXIDE FOR DNA RECOGNITION

Ana R. Monteiro (1,2), Catarina I. V. Ramos (2), Leandro M. O. Lourenço (2), Sara Fateixa (1), Joana

Rodrigues (3), Maria G. P. M. S. Neves (2), Tito Trindade (1)

1) CICECO, Department of Chemistry of University of Aveiro, 3810-193, Aveiro, Portugal.2) LAQV-REQUIMTE, Department of Chemistry of University of Aveiro, 3810-193 Aveiro, Portugal.

3) I3N, Department of Physics of University of Aveiro, 3810-193 Aveiro, Portugal.

The preparation of graphene-based nanomaterials for the detection of deoxyribonucleic acid (DNA) structures

has great interest for developing cancer diagnostic, and theranostic agents.

1

Zinc(II) phthalocyanines (ZnPcs)

have been described as effective DNA G-quadruplex stabilizers,

2

as well as non-covalent and covalent modifiers

of pristine graphene and graphene oxide (GO).

3,4

Here, we report a non-covalent functionalization route of GO

with octa-substituted thiopyridinium zinc(II) phthalocyanine (ZnPcB) and using mechanical exfoliation (Figure

1). The ensuing product (ZnPcB@GO) is a few-layered hybrid nanomaterial (interlayer distance 0.16 nm)

resulting from π-π stacking and electrostatic interactions between ZnPcB and GO. Concerning the UV-Vis

spectrum of ZnPcB, the hybrids showed pronounced bathochromic shifts on the Q-bands (Δ = 56 nm), which are

ascribed to changes in the molecular stacking of ZnPcB assembled onto the GO sheets. The interactions of

ZnPcB and ZnPcB@GO with different DNA oligonucleotides were studied by spectrophotometric and

spectrofluorimetric titrations. The role of the carbon nanomaterial in the recognition of duplex- or

G-Quadruplex-forming sequences will be discussed alongside the potential application of these nanomaterials in

DNA-based drugs for cancer therapies.

 

Figure 1 – Recognition of DNA structures using ZnPcB@GO.

 

 

References

1) Monteiro, A. R.; Ramos, C. I. V.; Fateixa, S.; Moura, N. M. M.; Neves, M. G. P. M. S.; Trindade, T., ACS Omega, 2018,

3, 1184.

2) Ramos, C. I. V.; Almeida, S.; Lourenço, L. M. O.; Pereira, P. M. R.; Fernandes, R.; Faustino, M. A. F.; Tomé, J. P. C.;

Carvalho, J.; Cruz, C.; Neves, M. G. P. M. S., Molecules, 2019, 24, 733.

3) Wibmer, L.; Lourenço, L. M. O.; Roth, A.; Katsukis, G.; Neves, M. G. P. M. S.; Cavaleiro, J. A. S.; Tomé, J. P. C.; Torres,

T.; Guldi, D. M., Nanoscale, 2015, 7, 5674.

4) Zhu, J.; Li, Y.; Chen, Y.; Wang, J.; Zhang, B.; Zhang, J.; Blau, W. J., Carbon, 2011, 49, 1900.

EFMC-ISMC | 283

M044

USE OF IMIDE CONDENSATION FOR PREPARING

CORE-DIVERSIFIED G-QUADRUPLEX LIGANDS: ANTICANCER

AND ANTIPARASITIC ACTIVITY

Steven S.T. Street (1), Pablo Peñalver (2), Michael O'Hagan (3), Gregory J. Hollingworth (4), Juan Carlos

Morales (2), Carmen Galan (3)

1) Department of Chemistry, University of Victoria, Victoria BC V8P 5C2 (Canada)2) Instituto de Parasitología y Biomedicina López Neyra, CSIC, PTS Granada, Avenida del Conocimiento, 17, 18016,

Armilla, Granada (Spain)3) School of Chemistry, University of Bristol Cantocks Close, Bristol, BS8 1TS (UK)

4) Novartis Institutes for Biomedical Research, Novartis Campus, 4002, Basel (Switzerland)

G-quadruplexes (G4) are DNA secondary structures, which play important roles in the regulation of gene

expression in human cells. They have been proposed as therapeutic targets in cancer. At the same time, putative

G-quadruplex forming sequences have also been found on the genome of parasites T. brucei, L. major and P.

falciparum suggesting they could also be explored as therapeutic targets.

G-quadruplex ligands are frequently formed by a heterocyclic aromatic structure modified with positively

charged groups. Here, we describe a facile imide coupling strategy for the one-step preparation of G-quadruplex

ligands with varied core chemistries. We prepared a small library of compounds and examined their ability to

bind DNA G-quadruplex using FRET melting experiments, CD, UV-Vis, fluorescence and NMR titrations. The

best G4 ligand was based on a perylene scaffold and exhibited 40-fold selectivity for a telomeric G-quadruplex

over duplex DNA. The perylene diimide ligand showed nanomolar inhibition of Trypanosoma brucei with

78-fold selectivity over MRC5 cells. The perylene derivative localized outside the nucleus and kinetoplast what

could be due to strong fluorescence quenching in the presence of small amounts of DNA.

1

References

1) Imide Condensation as a Strategy for the Synthesis of Core-Diversified G-Quadruplex Ligands with Anticancer and

Antiparasitic Activity. Steven T. G. Street, Pablo Penalver, Michael P. O’Hagan, Gregory J. Hollingworth, Juan C. Morales,

and M. Carmen Galan. Chem. Eur. J. 2021, 27, 7712–7721.

284 | EFMC-ISMC

M045

NEW PEX14 LIGANDS - A NEW STRATEGY TO INTERRUPT

GLYCOSOME BIOGENESIS

Piotr Mróz, Monika Marciniak, Maciej Dawidowski

Department of Drug Technology and Pharmaceutical Biotechnology, Faculty of Pharmacy, Medical University of Warsaw,Poland

      Glycosomes are organelles unique to kinetoplasts, including Trypanosoma spp. They contain enzymes

necessary for glycolysis - the source of ATP in the parasite cell and for other important metabolic pathways.

Accordingly, glycosomes are essential for the parasite cell and have long been considered attractive targets for

the development of new drugs against the diseases related to Trypanosoma infections [1,2].

      Glycosomes do not produce their own enzymes, therefore glycosomal proteins have to be transported from

the cytosol in a post-translational process. This is mediated by a cascade of interactions of small peroxisomal

proteins, called peroxins (PEX#). The PEX14-PEX5 protein-protein interaction (PPI) plays an important role in

this process. The PEX5 import receptor can bind short peroxisome targeting sequences (PTS) of the cargo

enzymes. The resulting PEX5-cargo complex binds PEX14 that sits at the glycosomal membrane, which allows

for cargo translocation into the matrix of the organelle [1,2].

      The aim of our research is to synthesize small-molecule inhibitors of PEX14-PEX5 protein-protein

interaction (PPI). It has previously been shown that disruption of complex formation between these two proteins

results in mislocalisation of glycosomal enzymes, ATP depletion and parasite death [3,4]. The compounds are

designed using high-resolution X-ray structures of PEX14-PEX5 PPI interface and synthesized by conventional

methods as well as by use of multicomponent reactions. The ultimate goal is to find new preclinical candidates

for Trypanosoma infections as well as to provide new molecular probes for glycosomal biogenesis testing.

 

Acknowledgements:

 

The research  tasks performer at Medical University of Warsaw within this topic is supported by National Centre

for Science (grant UMO-2016/23/B/NZ7/03339).

References

1) M. GualdroN-Lopez; A. N. A. Brennand; L. Avilan; P. A. Michels, Parasitology 140 (2013) 1.

2) J. Moyersoen; J. Choe; A. Kumar; F. G. Voncken; W. G. Hol; P. A. Michels, P. A. Eur. J. Biochem. 270 (2003) 2059.

3) M. Dawidowski; L. Emmanouilidis; V. C. Kalel; K. Tripsianes; K. Schorpp; K. Hadian; M. Kaiser; P. Mäser; M. Kolonko;

S. Tanghe; A. Rodriguez; W. Schliebs; R. Erdmann; M. Sattler; G. M. Popowicz, Science 355 (2017) 1416.

4) M. Dawidowski, V.C. Kalel, V. Napolitano, R. Fino, K. Schorpp, L. Emmanouilidis, D. Lenhart, M. Ostertag, M. Kaiser,

M. Kolonko, B. Tippler, W. Schliebs, G. Dubin, P. Maser, I. V. Tetko, K. Hadian, O. Plettenburg, R. Erdmann, M.Sattler, G.

M. Popowicz, J. Med. Chem. 2020, 63, 847−879

EFMC-ISMC | 285

M046

CHEMICAL SPACE DOCKING: MINING BILLIONS OF ON-DEMAND

MOLECULES IN 3D

Alexander Neumann, Marcus Gastreich, Christian Lemmen

BioSolveIT, An der Ziegelei 79, 53757 Sankt Augustin, Germany

The search for novel intellectual property (IP) in vast chemical spaces that comprise make-on-demand molecules

is experiencing an unseen wave of success [1]. Many research organizations and compound makers encode their

in-house chemical reaction knowledge as reaction-driven chemical spaces with sizes currently exceeding 10

20

accessible compounds [2,3, and the 2020 NIH Meeting on Ultralarge Databases in Chemistry].

 

It is highly desirable to exploit checmical space beyond mere "collections" using molecular docking.

Unfortunately, docking efforts are so far limited by brute-force algorithms applied to enumerated libraries.

Utilizing cloud-computing docking is possible, in a massively parallel fashion, entailing enormous amounts of

CPU/GPU time, and questionable power consumption. But brute force approaches fail, by and large, when it

comes to screening vast chemical spaces of several billions of compounds [4,5].

This is why we have devised clever algorithms that scale far better in this scenario. On the one hand side, we

developed a similarity search strategy that operates largely on the building blocks which are the foundation of

these chemical spaces and in combination lead to valid product molecules [6–9]. While you only need 1,000 plus

1,000 building blocks, these multiply and generate 1,000,000 virtual products which are available, on demand,

from compound suppliers. Based on that same foundation, we introduce here Chemical Space Docking, a new

method for flexible docking of billions-sized compound spaces. Algorithmically, we exploit an anchor-and-grow

strategy with reaction-based connections between formalized building blocks, reflecting real chemistry. Opposed

to any pre-filtered method, or methods that are limited by size, ths makies the entire, full chemical space

amenable as a result, in principle. The method has been applied successfully on a number of internal projects.

Here we will present the method and disclose recent results.

References

1) Hoffmann, T.; Gastreich, M. The next level in chemical space navigation: going far beyond enumerable compound

libraries. Drug Discov. Today 2019, 24, 1148–1156.

2) Grebner, C. Webinar: “Exploration and Mining of Large Virtual Chemical Spaces” Available online:

https://youtu.be/fMrI11SXwpU.

3) Knehans, T.; Klingler, F.-M.; Kraut, H.; Saller, H.; Herrmann, A.; Rippmann, F.; Eiblmaier, J.; Lemmen, C.; Krier, M.

Merck AcceSSible InVentory (MASSIV): In silico synthesis guided by chemical transforms obtained through bootstrapping

reaction databases. In Proceedings of the Abstracts of Papers, 254th ACS National Meeting & Exposition; Washington, DC,

USA, 2017.

4) Lyu, J.; Irwin, J.J.; Roth, B.L.; Shoichet, B.K.; Levit, A.; Wang, S.; Tolmachova, K.; Singh, I.; Tolmachev, A.A.; Che, T.;

et al. Ultra-large library docking for discovering new chemotypes. Nature 2019, 566, 224–229.

5) Gorgulla, C.; Boeszoermenyi, A.; Wang, Z.-F.; Fischer, P.D.; Coote, P.; Padmanabha Das, K.M.; Malets, Y.S.;

Radchenko, D.S.; Moroz, Y.S.; Scott, D.A.; et al. An open-source drug discovery platform enables ultra-large virtual screens.

Nature 2020, 1–8.

6) Hartenfeller, M.; Zettl, H.; Walter, M.; Rupp, M.; Reisen, F.; Proschak, E.; Weggen, S.; Stark, H.; Schneider, G. DOGS:

Reaction-driven de novo design of bioactive compounds. PLoS Comput. Biol. 2012, 8, 1–12.

7) Rarey, M.; Stahl, M. Similarity searching in large combinatorial chemistry spaces. J. Comput. Aided. Mol. Des. 2001, 15,

497–520.

8) Nicolaou, C.A.; Watson, I.A.; Hu, H.; Wang, J. The Proximal Lilly Collection: Mapping, Exploring and Exploiting

Feasible Chemical Space. J. Chem. Inf. Model. 2016, 56, 1253–1266.

9) Klingler, F.M.; Gastreich, M.; Grygorenko, O.O.; Savych, O.; Borysko, P.; Griniukova, A.; Gubina, K.E.; Lemmen, C.;

Moroz, Y.S. SAR by space: Enriching hit sets from the chemical space. Molecules 2019, 24, 3096–3106.

286 | EFMC-ISMC

M047

FIRST TOTAL SYNTHESIS OF CUDRAISOFLAVONE J AS

POTENTIAL NEUROPROTECTIVE AGENT

Lu Qili (1), Harmalkar Dipesh S. (1,2), Quan Guofeng (1), Kwon Haeun (3), Lee Donghoo (3), Lee Kyeong

(1)

1) College of Pharmacy, Dongguk University-Seoul, Goyang, 10326, Republic of Korea2) Department of Biotechnology, Korea University, Seoul, 02841, Republic of Korea

3) Department of Plant Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republicof Korea

C. tricuspidata, is widely used in east Asia as a traditional medicine or health supplement. Cudraisoflavone J,

which is isolated from C. tricuspidata, showed potent activity against 6-hydroxydopamine (6-OHDA) - induced

cell death in human neuroblastoma SH-SY

5

Y cells, with EC

50

value of 0.5 µM. Thus, cudraisoflavone J could be

considered as a candidate for further research for therapeutic purposes into neurodegenerative diseases such as

Parkinson's disease. To the best of our knowledge, neither the total synthesis nor the synthetic approach for the

absolute configuration of cudraisoflavone J has ever been reported. In this study, we report the first total

synthesis of racemic cudraisoflavone J by using Claisen rearrangement, and Suzuki coupling reaction as key

reactions. The racemic secondary alcohol was kinetically resolved to give (+) - and (−) - cudraisoflavone J with

up to 96.8% ee and 88.0% ee, respectively. The absolute configurations were determined by the modified

Mosher’s method. Furthermore, the synthetic methodology is well suited to the development of new structural

analogues.

EFMC-ISMC | 287

M048

SYNTHESIS OF NOVEL AMIDE TYPE HARMICINES

Marina Marinović, Zrinka Rajić

University of Zagreb Faculty of Pharmacy and Biochemistry, A. Kovačića 1, Zagreb, Croatia

Malaria, one of the most dangerous parasitic diseases, causes almost half a million deaths each year (1). In an

effort to reduce the growing resistance of the parasite to the available drugs, the approach of molecular

hybridization has been used to produce amide type (AT) harmicines, i.e. hybrids composed of β-carboline

alkaloid harmine and cinnamic acid derivatives (CADs) linked via amide bond. In our previous work we showed

that AT harmicines prepared at O-7 and N-9 of the β-carboline core exert increased antimalarial activity

compared to harmine (2). Encouraged by those results we decided to prepare novel AT harmicines at the

positions C-1, C-3 and C-6 of the β-carboline core (4a-h, 5a-h, 6a-h).

                                                                                                                                           

Scheme. Synthesis of novel harmicines.

Synthesis of harmicines (4a-h, 5a-h, 6a-h) involves preparation of harmine-based primary amines 1-3 and

coupling reactions between amines and different CADs (Scheme). Synthesis of amines 1-3 included several

reaction steps. C-1, C-3 and O-6 substituted β-carbolines were generated by Pictet-Spengler reaction between

tryptamine (C-1), L-tryptophan methyl ester (C-3) or 5-methoxytryptamine (O-6) and the corresponding

aldehyde (2,2-dimethoxyacetaldehyde/acetaldehyde dimethyl acetal) in TFA, followed by their aromatisation at

rt using KMnO

4

 in DMF (C-1) or in the microwave reactor using 10% Pd/C in EtOH (C-3 and O-6). Reduction

of aldehyde (C-1) or ester (C-3) with LiAlH

yielded alcohols, which were transformed to azides using

ADMP/DBU. Subsequent catalytic hydrogenation of azides generated amines 1 and 2. On the other hand, ether

at O-6 was hydrolysed in AcOH/HBr and the obtained phenol alkylated with 2-(Boc-amino)ethyl bromide. The

removal of the Boc-protecting group under acidic conditions resulted in the preparation of amine 3. Finally, the

targeted harmicines (4a-h, 5a-h, 6a-h) were prepared by coupling reactions between CADs and harmine-based

amines (1-3) in the presence of HATU/DIEA in dichloromethane. Structures of newly prepared harmicines were

confirmed by spectroscopic and spectrometric methods (IR, MS, 

1

H and 

13

C NMR). Evaluation of their

antimalarial activity and cytotoxicity is in progress.

This work was fully supported by the Croatian Science Foundation under the project number UIP-2017-05-5160and by the Young researcher’s career development project – training of doctoral students of the CroatianScience Foundation founded by the European Union from the European Social Fund.

References

1) https://www.who.int/publications/i/item/9789240015791 (date of access 18 March 2021)

2) M. Marinović et al., Molecules 2020, 25, 4376.

288 | EFMC-ISMC

M049

SYNTHESIS OF C-1- AND C-3-TETHERED TRIAZOLE TYPE

HARMICINES

Goran Poje, Ivana Perković, Zrinka Rajić

University of Zagreb Faculty of Pharmacy and Biochemistry, A. Kovačića 1, 10000 Zagreb

As malaria remains a global health threat, there is a great need for new and effective antimalarials. Novel and

emerging approach in the development of antimalarial agents is molecular hybridization, i.e. preparation of

hybrid compounds (1). Harmicines are hybrids consisting of two moieties with antimalarial properties, namely

harmine, a β-carboline alkaloid and cinnamic acid derivatives (CADs), linked via triazole ring or amide bond. In

our previous work, we have shown that triazole type (TT) harmicines prepared at O-7 and N-9 positions of the

β-carboline core exert significant antiplasmodial activities (2). To broaden our knowledge on their

structure-activity relationship, we decided to expand the series of TT harmicines by preparing new generation of

hybrids tethered at C-1 and C-3 positions of the β-carboline ring, 4a-e and 5a-e.

Scheme. Synthesis of novel harmicines.

Novel harmicines were prepared via “click” reaction of CAD-based alkynes (1a-e) and β-carboline azides (2

 and 3), resulting in a triazole type linker (Scheme). “Click” reaction proceeded smoothly with Na ascorbate and

CuSO

4

 × 5H

2

O as a source of Cu(I), in a t-BuOH/water mixture (1:1). Synthesis of CAD-based alkynes (1a-e)

was straightforward and it was accomplished by the reaction of a corresponding CAD with propargyl bromide in

presence of K

2

CO

3

. On the other hand, synthesis of azides 2 and 3 involved several steps. Pictet-Spengler

condensation of tryptamine and 2,2-dimethoxyacetaldehyde (C-1) or tryptophan methyl ester and acetaldehyde

dimethyl acetal (C-3), followed by the oxidation with KMnO

4

 at rt or Pd/C under microwave irradiation yielded

substituted β-carbolines. Their reduction with LiAlH

4

, i.e. reduction of aldehyde (C-1) or ester (C-3), resulted in

the synthesis of the corresponding alcohols, which were efficiently transformed to azides using

ADMP/DBU. Structures of novel harmicines were confirmed by MS, IR, 

1

H and 

13

C NMR. Evaluation of their

antiplasmodial activity and cytotoxicity is in progress.

This work was fully supported by the Croatian Science Foundation under the project numberUIP-2017-05-5160.  

References

1) D. Agarwal at al., Antimicrob. Agents Chemother. 2017, 61, e00249-17.

2) I. Perković et al., Eur. J. Med. Chem. 2020, 187, 111927.

EFMC-ISMC | 289

M050

SYNTHESIS OF O- AND N-HARMIRINES, NOVEL

HARMINE-COUMARIN HYBRIDS

Kristina Pavić, Goran Poje, Zrinka Rajić

University of Zagreb Faculty of Pharmacy and Biochemistry, A. Kovačića 1, 10000 Zagreb, Croatia

Coumarin and harmine, naturally occurring compounds, and their derivatives exhibit a wide range of biological

activities, such as anticancer, antibacterial, antimalarial, or antifungal (1, 2). Numerous papers show that

coumarins exert their anticancer activity through different pathways (cell cycle arrest, inhibition of kinases,

aromatase, Hsp90, telomerase, and angiogenesis) (1). Moreover, harmine, as well as other β-carboline alkaloids,

also interact with different anticancer drug targets, such as DNA (intercalation, groove binding), topoisomerase,

kinases, tubulin, etc. (2). Thus, hybridization of coumarin and harmine could be a promising strategy to improve

their activity, reduce side effects, and overcome drug resistance. To this end, we have prepared harmirines, new

hybrid molecules comprising both motifs, linked by a 1,2,3-triazole spacer.

The title compounds were obtained by applying the standard Cu(I)-catalyzed azide-alkyne cycloaddition

(CuAAC) (Scheme). To prepare harmirines, we synthesized coumarin-based azides 2a-d and harmine-based

terminal alkynes 3-5. Azides 2a-d were prepared in a two-step procedure from 4-hydroxycoumarins. The first

step included chlorination of coumarins with phosphorus oxychloride. The obtained coumarin chlorides 1a-d

 were subsequently transformed to azides 2a-d using sodium azide. The preparation of harmine-based terminal

alkynes 3-5 at positions 6, 7, and 9 of the β-carboline core was previously described (3, 4). Synthesized azides

and alkynes were the starting compounds for the CuAAC reactions that resulted in three types of hybrids: 6- and

7-substituted O-harmirines 6a-d and 7a-d, respectively, and 9-substituted N-harmirines 8a-d. ‘Click’ reactions

proceeded using Cu(II)-acetate precatalyst in methanol. The scheme outlines the general route leading to the title

compounds. The structures of newly prepared compounds 6-8 were confirmed by standard methods (IR, 

1

H, 

13

C

NMR, MS). The evaluation of their antiproliferative activity is in progress.

Scheme. Synthesis of harmirines.

This work was fully supported by the Croatian Science Foundation under the project numberUIP-2017-05-5160.

References

1) A. Thakur et al., Eur. J. Med. Chem. 2015, 101, 476.

2) S. Aaghaz et al., Eur. J. Med. Chem. 2021, 216, 113321.

3) I. Perković et al., Eur. J. Med. Chem. 2020, 187, 111927.

4) M. Marinović et al., in preparation.

290 | EFMC-ISMC

M051

TARGETING TRPV1 SOFTLY: SYNTHESIS AND DEVELOPMENT OF

CAPSAICINOID MODULATORS WITH IN VIVO EFFICACY IN

MOUSE MODELS OF SKIN DISEASES

Marta Serafini (1), Asia Fernandez-Carvajal (2), Antonio Ferrer-Montiel (2), Tracey Pirali (1)

1) Department of Pharmaceutical Sciences; Università del Piemonte Orientale; Novara, 28100, Italy2) Instituto de Biología Molecular y Celular, Universitas Miguel Hernandez, Av de la Universidad s/n, 03202 Elche, Spain

Transient receptor potential vanilloid 1 (TRPV1) channel is an ion channel that plays a crucial role in the

pathogenesis of skin disorders, especially related to inflammation and pruritus.

1

Over the last decades, capsaicin

and other agonists have been developed for topical application, but their important side effects, i.e. the initial

burning sensation, frequent erythema reactions and skin carcinogenesis related to the long residence time in the

skin, have always hampered the use of these compounds. Besides agonists, several topical antagonists have been

discovered

2

but their clinical development has been undermined by hyperthermia induction and promotion of

skin cancer.

In our work, 42 capsaicinoid soft analogues have been synthesized and developed.

3

These soft drugs are able to

undergo topical deactivation by the hydrolyzing activity of skin esterases after having played their topical effect.

4

The implanting of an ester group in the lipophilic side chain of capsaicinoids using the Passerini

multicomponent reaction affords both agonists and antagonists with good TRPV1 modulating activity. At the

same time, these soft analogues are susceptible to in situ topical deactivation, avoiding the side effects related to

the long-term residence in the skin and to the systemic distribution of the compounds. Among the antagonists,

compound AG1529 potently and competitively blocked capsaicin-evoked activation of hTRPV1, mildly affected

pH and voltage-induced activation, and did not alter heat responses. Moreover, the compound abolished

histaminergic and inflammatory sensitization of TRPV1 in nociceptors. Finally, topical application of AG1529

dose-dependently attenuated histaminergic pruritus in mice, without increasing body temperature. Taken

together, these pre-clinical results substantiate this capsaicin-based soft antagonist as a promising candidate for

the treatment of psoriatic pruritus.

References

1) Caterina, M. J.; Pang, Z. Pharmaceuticals 2016, 9, 77-104.

2) Trevisani, M.; Gatti, R. Open Pain J. 2013, 6, 108-118

3) a) Serafini, M.; Griglio, A.; Aprile, S.; Seiti, F.; Travelli, C. et al. J. Med. Chem. 2018, 61, 4436-4455; b) Devesa, G. I.;

Genazzani, A. A.; Pirali, T.; Fernandez-Carvajal, A.; Ferrer-Montiel, A. V. May 11, 2017. EP17382266.9; c) Lockwood, D.

Chili pepper compound made to self-destruct. C&EN 2018, 96(25).

4) Aprile, S.; Serafini, M.; Pirali, T. Drug Discov. Today 2019, 24, 2234-2246.

EFMC-ISMC | 291

M052

PROTAC-MEDIATED INACTIVATION OF E3 LIGASES: FROM

TOOLS TO CANCER TREATMENT

Christian Steinebach (1), Yuen Lam Dora Ng (3), Aleša Bricelj (2), Izidor Sosič (2), Jan Krönke (3),

Michael Gütschow (1)

1) University of Bonn, Pharmaceutical Institute, Bonn, Germany2) University of Ljubljana, Faculty of Pharmacy, Ljubljana, Slovenia

3) Charité, Berlin, Germany

Targeted protein degradation is a rapidly growing field in drug discovery. In addition to their emerging role in

the PROTAC technology, E3 ligases represent attractive therapeutic targets.[1] However, the development of E3

inhibitors is complicated by the absence of a catalytic domain.

To investigate whether a ligase itself can be targeted for degradation by PROTACs, we developed homodimeric

PROTACs that caused a self-directed inactivation of cereblon (CRBN) via ternary complex formation,

subsequent ubiquitination, and degradation.[2] Such homo-PROTACs might be a considerable leap forward to

unravel the biological complexities of the E3 ligase CRBN and to provide additional insights into the mechanism

of CRBN-targeting drugs. However, homodimers were hampered by the self-limiting reduction of the E3 ligase

component and neomorphic activities on the transcription factors IKZF1 and IKZF3. A superior set of

compounds was assembled from a CRBN and a Von Hippel-Lindau (VHL) ligand. For the first time,

PROTAC-induced heterodimerization of two E3 ligases with unidirectional ubiquitination and efficient

degradation of CRBN was described.[3]

Inspired by these results, our PROTAC-mediated depletion strategy was applied to therapeutically relevant E3

ligases. A combinatorial library of IAP/CRBN/VHL-based PROTACs was designed, synthesized, and

investigated for their capability to target different BIRC domains.[4] The entire set of PROTACs spans pan-IAP

degraders, XIAP-selective compounds, as well as dual-active entities. The homolog-selective modulation of

degradation targets was not yet possible with small-molecule IAP inhibitors. Furthermore, our bivalent platform

could potentially be used for targeting the E3 ligase Keap1, which was recently found to be upregulated in

relapsed multiple myeloma patients. [5]

References

1) Huang et al., Cell Res., 2016

2) Steinebach et al., ACS Chem. Biol., 2018

3) Steinebach et al., Chem. Commun., 2019

4) unpublished data

5) Ng et al., in revision

292 | EFMC-ISMC

M053

STRUCTURAL INSIGHTS INTO PROTAC-MEDIATED

DEGRADATION OF BCL-XL

Christopher P. Tinworth (1), Chun-wa Chung (2), Han Dai (1), Esther Fernandez (1), Ian Churcher (1),

Jenni Cryan (1), John D. Harling (1), Agnieszka Konopacka (1), Markus A. Queisser (1), Christopher J.

Tame (1), Gillian Watt (1), Fan Jiang (3), Dongming Qian (3), Andrew B. Benowitz (1)

1) Protein Degradation Group, GlaxoSmithKline, Stevenage SG1 2NY, United Kingdom2) Protein, Cellular & Structural Sciences, GlaxoSmithKline, Stevenage SG1 2NY, United Kingdom

3) Viva Biotech, Ltd., Shanghai 201202, China

The Bcl-2 family of proteins, such as Bcl-xL and Bcl-2, play key roles in cancer cell survival.

1

Structural studies

of Bcl-xL formed the foundation for the development of the first Bcl-2 family inhibitors and FDA approved

drugs. Recently, Proteolysis Targeting Chimeras (PROTACs) that degrade Bcl-xL have been proposed as a

therapeutic modality with the potential to enhance potency and reduce toxicity versus antagonists.

2

However, no

ternary complex structures of Bcl-xL with a PROTAC and an E3 ligase have been successfully determined to

guide this approach.

We report the design, characterization, and X-ray structure of a VHL E3 ligase-recruiting Bcl-xL PROTAC

degrader.

3

The 1.9 Å heterotetrameric structure, composed of (ElonginB:ElonginC:VHL):PROTAC:Bcl-xL,

reveals an extensive network of neo-interactions, between the E3 ligase and the target protein, and between

noncognate parts of the PROTAC and partner proteins. This work illustrates the challenges associated with the

rational design of bifunctional molecules where interactions involve composite interfaces.

 

References

1) Semin. Cancer Biol. 2003, 13, 115-123

2) Nat. Med. 2019, 25, 1938–1947

3) ACS Chem. Biol. 2020, 15, 2316–2323

EFMC-ISMC | 293

M054

DISCOVERY OF NOVEL ANTITUBERCULOSIS AGENTS

TARGETING AMINOACYL-TRNA SYNTHETASES

Galyna Volynets (1), Mariia Rybak (2), Sergiy Starosyla (1), Volodymyr Bdzhola (1), Mykola Protopopov

(1,3), Sergiy Yarmoluk (1), Michail Tukalo (2)

1) Department of Medicinal Chemistry, Institute of Molecular Biology and Genetics, NAS of Ukraine, 150 Zabolotnogo St.,03143, Kyiv, Ukraine

2) Department of Protein Synthesis Enzymology, Institute of Molecular Biology and Genetics, NAS of Ukraine, 150Zabolotnogo St., 03143, Kyiv, Ukraine

3) Scientific Services Company Otava Ltd., 150 Zabolotnogo St., 03143, Ukraine

Antibiotic resistance is a major problem of tuberculosis treatment. This provides the stimulus for the search of

novel approaches and molecular targets to overcome resistance in Mycobacterium tuberculosis.

Nowadays, multitarget drug discovery is considered as a promising approach to reduce or forestall resistance,

since mutations are necessary in all targets for resistance development.

In order to identify hit compounds targeting simultaneously two enzymes – M. tuberculosis leucyl-tRNA

synthetase (LeuRS) and methionyl-tRNA synthetase (MetRS), we generated ligand-based pharmacophore

models and performed pharmacophore screening of OTAVA database containing about 100,000 compounds.

According to virtual screening results we have selected 156 compounds for investigation of inhibitory activity

toward mycobacterial LeuRS and MetRS. In vitro aminoacylation assay revealed five compounds belonging to

different chemical classes inhibiting both enzymes. Among them the most active compound –

3-(3-chloro-4-methoxy-phenyl)-5-[3-(4-fluoro-phenyl)-[1,2,4]oxadiazol-5-yl]-3H-[1,2,3]triazol-4-ylamine

decreases the activity of mycobacterial LeuRS and MetRS with IC

50

values of 13 µM and 13.8 µM,

correspondingly. Unfortunately, the identified inhibitor does not demonstrate antibacterial activity toward M.tuberculosis.

To find aminoacyl-tRNA synthetase inhibitors among this chemical class possessing antibacterial activity toward

mycobacterial resistant strains we carried out semi-flexible docking of

3-phenyl-5-(1-phenyl-1H-[1,2,3]triazol-4-yl)-[1,2,4]oxadiazole derivatives library into active sites of LeuRS and

MetRS and selected 41 compounds for study of their inhibitory activity against recombinant MetRS and LeuRS

and antibacterial activity against M. tuberculosis mono-resistant strains. In vitro aminoacylation assay revealed

10 compounds with inhibitory activity against MetRS and 3 compounds active against LeuRS. The antibacterial

microdilution screening resulted in 4 compounds possessing activity against M. tuberculosis strains in the range

of concentrations 2-20 mg/L. It was found, that among compounds with antibacterial activity only one –

3-(5-Chloro-2-methoxy-phenyl)-5-[3-(4-fluoro-phenyl)-[1,2,4]oxadiazol-5-yl]-3H-[1,2,3]triazol-4-ylamine,

demonstrates significant enzyme inhibitory activity against M. tuberculosis MetRS with IC

50

value of 148.5 µM.

This compound is not cytotoxic toward human HEK293 and HepG2 cell lines.

Therefore, the derivatives of 3-phenyl-5-(1-phenyl-1H-[1,2,3]triazol-4-yl)-[1,2,4]oxadiazole can be promising

candidates for further chemical optimization to develop novel types of antituberculosis agents.

294 | EFMC-ISMC

M055

NOVEL UNNATURAL NUCLEOSIDE ANALOGUES AS PRMT5

INHIBITORS

Murray Wan, Shuhei Kawamura, Brooke Swalm, Benjamin Nicholson, Catherine Gerdt, Charles Yeung,

Christopher Sondey, Craig Gibeau, Danica Rankic, Dapeng Chen, David Candito, David Sloman, David

Witter, Doug Linn, Erin DiMauro, Haiyan Xu, Jonathan Hughes, Jongwon Lim, Michelle Machacek,

Mayankbhai Patel, Michael Altman, Michael Reutershan, My Mansueto, Nicole Follmer, Patrick Fier,

Paul Ciaccio, Pierre Daublain, Phieng Siliphaivanh, Rachel Kubiak, Robert Hayes, Ryan Quiroz1, Sandra

Lee, Sebastian Schneider, Steven Silverman, Sulagna Sanyal, Tiffany Piou, Timothy Cernak, Timothy

Henderson, Wonsuk Chang, Yingchun Ye, Yves Ducharme, Zangwei Xu

Merck Research Laboratories33 Avenue Louis Pasteur

Boston, MA 02115

Protein arginine methyl transferase 5 (PRMT5) mediates post translational modification of proteins via

symmetric dimethylation of arginine residues. The dysregulation of this process has been implicated in the

development and progression of various types of cancers. Furthermore, preclinical mouse models have provided

strong evidence that PRMT5 inhibition can induce tumor growth regression in vivo. As a result, identification of

novel inhibitors has been a topic of significant interest within the pharmaceutical industry. Herein we describe

structure-based drug design efforts that drove the discovery of 3’-C-methyl-7-deazaadenosine nucleoside-derived

PRMT5 inhibitors. Specifically, we will highlight how leveraging ligand conformational strain energy

calculations, binding site water mapping, and novel binding site interactions enabled identification of highly

selective and potent inhibitors. Furthermore, we will describe a property-based optimization approach that

resulted in discovery of molecules with excellent pre-clinical pharmacokinetics and overall favorable properties.

Finally, in vivo efficacy data with these novel inhibitors will be described, providing insights into the evaluation

of a PRMT5 inhibitor for use as a therapy.

EFMC-ISMC | 295

M056

BENZATHIAZOLE ANALOGS AS BIOLOGICALLY ACTIVE AGENTS

Ajayrajsinh Zala , Premlata Kumari

Department of Applied Chemistry, S.V. National Institute of Technology, Surat, Gujarat, India

Benzothiazole has a heterocyclic structure, fundamental in nature with a wide assortment of dynamic

destinations. As a little scaffold, it contains electronegative atoms due to which it is achievable for different

moieties to frame conjugates. Alluding to the bioorganic and medicinal chemistry, the moiety can possibly give

dynamic pharmacophores to grow new therapeutically important compound. Benzothiazole subordinates were

found to show adequacy against some intense illnesses like cancer, neurodegeneration, neuropathic torment,

infectious diseases, epilepsy, etc. In this book chapter, benzothiazole derivatives are given anticonvulsant and

anticancer activity. A series of semicarbazones, pyrimidinetriones some of the derivatives are shown major

activity in MES test as 22.8, 13.9, 25.4 and isothiourea two derivatives shows major activity in MES test as 30

and shown promising anticonvulsant activities. A series of acetohydrazide and amino benzothiazole derivatives

are given highest GI50 value of 7.18 _ 10_8 M against HOP-92 Lung Cancer cell line and derivatives of

benzothiazoles are evaluated for their antitumor activity.

References

1) Siddiqui, N., Rana, A., Khan, S. A., Bhat, M. A., and Haque, S. E. (2007). Synthesis of benzothiazole semicarbazones as

novel anticonvulsants- The role of hydrophobic domain. Bioorganic & medicinal chemistry letters, 17(15), 4178-4182.

2) Siddiqui, N., and Ahsan, W. (2009). Benzothiazole incorporated barbituric acid derivatives: synthesis and anticonvulsant

screening. Archiv der Pharmazie: An International Journal Pharmaceutical and Medicinal Chemistry, 342(8), 462-468.

3) Siddiqui, N., Alam, M. S., Sahu, M., Naim, M. J., Yar, M. S., and Alam, O. (2017). Design, synthesis, anticonvulsant

evaluation and docking study of 2-[(6-substituted benzo [d] thiazol-2-ylcarbamoyl) methyl]-1-(4-substituted phenyl)

isothioureas. Bioorganic chemistry, 71, 230-243.

4) Noolvi, M. N., Patel, H. M., and Kaur, M. (2012). Benzothiazoles: search for anticancer agents. European journal of

medicinal chemistry, 54, 447-462.

5) Osmaniye, D., Levent, S., Karaduman, A., Ilgın, S., Özkay, Y., and Kaplancıklı, Z. (2018). Synthesis of New

Benzothiazole Acylhydrazones as Anticancer Agents. Molecules, 23(5), 1054.

296 | EFMC-ISMC

M057

UNDERSTANDING THE MECHANISM OF SECONDARY

SULFONAMIDE BINDING TO CARBONIC ANHYDRASE

Asta Zubriene (1), Denis Baronas (1), Virginija Dudutiene (1), Vaida Paketuryte (1), Visvaldas Kairys (2),

Daumantas Matulis (1)

1) Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University,Sauletekio 7, Vilnius LT-10257, Lithuania

2) Department of Bioinformatics, Institute of Biotechnology, Life Sciences Center, Vilnius University, Sauletekio 7, VilniusLT-10257, Lithuania

Metalloenzyme carbonic anhydrase (CA), containing Zn(II) in the active site, exhibits up to picomolar affinity

for primary sulfonamides that form a coordination bond between the negatively charged amino group of the

compound and the zinc ion. Secondary, the N-alkyl-substituted, sulfonamides bind CA with much lower affinity.

A series of secondary sulfonamides were synthesized and, together with analogous primary sulfonamides, the

affinities for twelve catalytically active human CA isoforms were determined by the fluorescent thermal shift

assay, stopped-flow assay of inhibition of enzymatic activity and isothermal titration calorimetry. The binding

profile of a secondary sulfonamide as a function of pH revealed the same U-shape dependence known for

primary sulfonamides. This dependence demonstrated that there were binding-linked protonation reactions that

should be dissected for the estimation of the intrinsic binding constants. X-ray crystallographic structures of

secondary sulfonamides bound to CA II were determined and compared to analogous primary sulfonamide

complexes showing the altered binding orientation. Computational modeling dissected the atomic contributions

to the binding energetic and molecular docking yielded correct orientation of bound compounds providing

insight into the sulfonamide binding mechanism to CA.

EFMC-ISMC | 297

M058

A STUDY OF THE EFFECT OF COMMON

ELECTRON-WITHDRAWING GROUPS ON THE PHYSIOCHEMICAL

PROPERTIES OF 2-AMINOTETRAHYDORPYRIDINE BACE1

INHIBITORS

Meng-Yang Hsiao, Frederik Rombouts, Ann Vos, Harrie Gijsen

Janssen Research & Development, Turnhoutseweg 30, B-2340 Beerse

Medicinal chemists are often facing the challenge of reducing the pK

a

of strongly basic groups present in drug

molecules to a range that is optimal for their potency and drug-like properties such as solubility, permeability,

volume of distribution (V

d

), and off-target toxicity. The 2-aminotetrahydorpyridine core is a key component of

potent inhibitors of BACE1 – the rate-limiting enzyme in the production of Aβ amyloid proteins which lead to

plaques formation in Alzheimer’s disease. During our work, we strived to balance potency and physiochemical

properties to achieve brain penetration by introducing diverse groups on the 2-aminotetrahydropyridine core to

modulate its logD, PSA, and pK

a

parameters. To prioritize the synthetic efforts of these highly functionalized

warheads, we implemented a design-synthesis-design workflow which includes: (1) docking simulations, and (2)

MPO scoring based on predicted physiochemical properties. To achieve this, we evaluated a set of commercially

available software packages which are specialized in predicting the physicochemical properties of drug leads.

We believe that this work provides a conceptual framework to guide medicinal chemists in their endeavors to

optimizing the pK

a

values of amidine and amine bases present in their drug leads.

References

1) Hsiao CC, Rombouts F, Gijsen HJM. New evolutions in the BACE1 inhibitor field from 2014 to 2018. Bioorg Med Chem

Lett. 2019; 29(6): 761-777.

298 | EFMC-ISMC

NOTES

EFMC-ISMC | 299

Posters - TechnologiesN. Prodrug Strategies in Drug Design and Discovery

300 | EFMC-ISMC

N001

TARGETING HYPOTHALAMIC FREE FATTY ACID RECEPTORS

WITH LYSOPHOSPHATIDYLCHOLINE DRUG CARRIERS

Athanasios Papangelis, Trond Ulven

University of Copenhagen, Universitetsparken 22100 København Ø, Denmark

Hypothalamic inflammation has been identified as an important factor in the development of obesity.

1

Recent

studies have shown that free fatty acid receptors FFA1 and FFA4, expressed in the hypothalamus, control energy

homeostasis and inflammation.

2, 3

An attractive strategy against obesity is therefore to target the hypothalamic

FFA1 and FFA4 via a lysophosphatidylcholine (LPC)-conjugate that could be actively transported into the brain

via the Mfsd2a. Recently, the transporter Mfsd2a was identified as essential for the uptake of DHA and other

fatty acids through the blood-brain barrier as LPC conjugates.

4

Herein, we report the design, synthesis, and

initial evaluation of synthetic LPC analogues bearing free fatty acid receptor agonists as pharmacological tools to

target these receptors in the CNS.

References

1) Wisse, B. E.; Schwartz, M. W., Does Hypothalamic Inflammation Cause Obesity? Cell Metabolism 2009, 10 (4), 241-242.

2) Dragano, N. R. V.; Solon, C.; Ramalho, A. F.; de Moura, R. F.; Razolli, D. S.; Christiansen, E.; Azevedo, C.; Ulven, T.;

Velloso, L. A., Polyunsaturated fatty acid receptors, GPR40 and GPR120, are expressed in the hypothalamus and control

energy homeostasis and inflammation. J Neuroinflammation 2017, 14 (1), 91.

3) Engel, D. F.; Bobbo, V. C. D.; Solon, C. S.; Nogueira, G. A.; Moura-Assis, A.; Mendes, N. F.; Zanesco, A. M.;

Papangelis, A.; Ulven, T.; Velloso, L. A., Activation of GPR40 induces hypothalamic neurogenesis through p38- and

BDNF-dependent mechanisms. Scientific Reports 2020, 10 (1), 11047.

4) Nguyen, L. N.; Ma, D.; Shui, G.; Wong, P.; Cazenave-Gassiot, A.; Zhang, X.; Wenk, M. R.; Goh, E. L. K.; Silver, D. L.,

Mfsd2a is a transporter for the essential omega-3 fatty acid docosahexaenoic acid. Nature 2014, 509 (7501), 503-506.

ABSTRACT CANCELLED

EFMC-ISMC | 301

N002

SYNTHESIS OF FLUORESCENCE-LABELED

TRIPPPRO-COMPOUNDS FOR CELLULAR UPTAKE STUDIES IN

VARIOUS CANCER CELL LINES

Julian Witt (1), Simon Remus (2), Udo Schumacher (2), Chris Meier (1)

1) Organic Chemistry, Chemistry Department, University of Hamburg, Hamburg, Germany2) Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany

Nucleoside and nucleobase analogues (NNAs) are well established and widely used as chemotherapeutic agents.

However, efficient distribution of chemotherapeutic agents and more specifically their penetration into tumor

tissue and cells remains challenging. The lipophilic microenvironment of solid tumors in combination with

lacking vascular systems in these tissues leads to poor uptake of anticancer drugs and consequently their

therapeutic effect is highly limited. In case of many NNAs, another limiting factor lies within intracellular

metabolism to form the nucleoside analogue (NA) triphosphate as active metabolite, which is then incorporated

into DNA and RNA during synthesis, resulting in chain termination and apoptosis.

We have previously established a prodrug technology for intracellular delivery of NA triphosphates that uses

lipophilic masking units at the γ-phosphate. This TriPPPro-approach bypasses inefficient phosphorylation steps

of NAs inside the cells and at the same time it enables compounds to penetrate into lipophilic tissue and cells.

The TriPPPro-technology has been applied to NAs used in antiviral therapy which strongly increased their

biological activity compared to parent NAs.

To visualize the uptake of TriPPPro-compounds into tumor cells, we synthesized various TriPPPro-compounds

of fluorescent bicyclic nucleoside analogues (BCNAs). Compared to UV/Vis-absorption-based detection,

fluorescence achieves about 1000-times higher sensitivity. Additionally, interference is low as most of the cells’

components do not fluoresce. These compounds were taken-up by various cancer cell lines and importantly, even

those cells that are so far not treatable with chemotherapeutic agents due to insufficient penetration into the cells

and tissue. Studies to apply the TriPPPro-concept to nucleoside analogues already used in chemotherapy are

ongoing with the aim to significantly improve their efficacy.

 

 

302 | EFMC-ISMC

N003

PLEIOTROPIC PRODRUGS: A NOVEL POLYPHARMACOLOGY

APPROACH TO TREAT NEURODEGENERATIVE DISEASES

Christophe Rochais, Patrick Dallemagne

Centre d Etudes et de Recherche sur le Medicament de Normandie (CERMN), Normandie Univ., UNICAEN, UFR Sante - BdBecquerel, F- 14032 Caen, France

Today, treatment of Alzheimer's Disease (AD) mainly involves acetylcholinesterase inhibitors (AChEIs).

AChEIs display solely a symptomatic benefit, alleviating the cognitive disorders associated to AD through a

temporary restoration of the cholinergic neurotransmission impaired by the neurodegeneration. The gradual loss

of efficiency for AChEIs led to associate them to drugs exhibiting potential disease-modifying properties. The

Multi-Target-Directed Ligands (MTDLs) were used in the recent years with a great potential benefit towards

multiple targets implicated in the complex AD,

1

as well as other neurodegenerative syndromes, which involve

multiple pathogenic factors. Our contribution to the field led recently to the discovery of Donecopride, the first

5-HT

4

R partial agonist, which further possesses important acetylcholinesterase (AChE) inhibition properties.

Donecopride is currently under preclinical development.

2,3,4

Based on this experience, we have recently

developed a novel pleiotropic prodrug approach. to generate promising in vivo active compounds. Based on the

model of rivastigmine, novel MTDLs were designed, acting as prodrugs, able to temporarily covalently bind and

inhibit AChE (for a symptomatic effect) and to secondarily release a drug able to selectively reach another AD

target (for a potential disease-modifying effect). This concept was applied to a several secondary targets,

including several 5-HT receptors of interest (typically 5-HT

4

and 5-HT

6

subtypes)

5

for the treatment of AD. The

concept, the synthetic development, as well as the in vitro and in vivo evaluation of two novel candidates, will be

presented in this communication.

6,7

Undisclosed results concerning a repositioning strategy and an innovativeself-immolative linkerwill be also presented for the first time in this communication.

References

1) Cavalli, A.; et al. J. Med. Chem.; 2008, 51 (3), 347–372.

2) Lecoutey, C. et al.; Proc. Natl. Acad. Sci. USA, 2014, 111(36), E3825–E3830

3) Rochais, C. et al. J. Med. Chem., 2015, 58 (7), 3172–3187

4) Rochais, C. et al. Br. J. Pharmacol. 2020, https://doi.org/10.1111/bph.14964.

5) Lalut, J. et al. Future Med. Chem. 2017 9(8), 781–795.

6) Toublet, F.X. et al Molecules 2019, 24(15), 2786.

7) Toublet, F.X. Eur. J. Med. Chem. 2021, 210, 113059

EFMC-ISMC | 303

Posters - Drug Discovery ProjectsO. Addressing Neglected and Emerging Viral Diseases with Small

Molecules

304 | EFMC-ISMC

O001

DISCOVERY OF NITD-688: A POTENT PAN-SEROTYPE INHIBITOR

OF DENGUE VIRUS NS4B PROTEIN

Stephanie A. Moquin (1,3), Oliver Simon (2,4), Fumiaki Yokokawa (1), Suresh B. Lakshminarayana (1),

Feng Wang (3), Bryan K. S. Yeung (2), Feng Gu (1)

1) Novartis Institute for Tropical Diseases, Emeryville, CA 94608, USA2) Novartis Institute for Tropical Diseases, Singapore 138670, Singapore

3) Novartis Institutes for BioMedical Research, Emeryville, CA 94608, USA4) Novartis (Singapore) Pte Ltd, Singapore 117432, Singapore

Dengue fever is a mosquito-borne viral disease caused by the four serotypes of dengue virus (DENV), which is

widespread in tropical and subtropical countries. To date, antiviral agents to specifically treat dengue virus are

not available, consequently, there is an urgent need to develop an effective and safe pan-serotype dengue virus

inhibitor. We performed a high-throughput phenotypic screen of 1.5 million compounds in the Novartis

proprietary library using a single-point cytopathic effect (CPE) assay against DENV-2 in Huh-7 cells. Active hits

(EC

50

< 5 uM, 160 compounds) were tested for activity against all four serotypes in a DENV-infected A549

cell–based flavivirus immunodetection (CFI) assay. To help rule out host-targeting compounds, we also

performed a counter screen against Chikungunya virus, a closely related virus from the Alphaviridae family, as

well as cytotoxicity assays using HepG2 and Huh-7 cells. This screening campaign led to the identification of

tetrahydrobenzothiophene 1 as a validated hit with submicromolar potency against all four dengue serotypes.

Following extensive structure-activity relationship (SAR) studies, an optimization of physicochemical properties

resulted in the identification of the N-substituted tetrahydrothienopyridine derivative NITD-688 (2), which

showed a strong potency against all four serotypes (EC

50

= 0.008–0.038 uM) and showed excellent oral efficacy

in the DENV-2 infected AG129 mouse model, with a 1.44 log viremia reduction at 30 mg/kg twice daily for 3

days. Importantly, NITD-688 (2) treatment also resulted in a 1.16-log reduction in viremia when mice were

treated 48 hours after infection. Selection of resistance mutations and binding studies with recombinant protein

using NMR analysis indicated that the molecular target of NITD-688 (2) is NS4B protein, which is a

nonenzymatic transmembrane protein and a component of the viral replication complex. NITD-688 (2) was

shown to have a long half-life and good oral bioavailability in pharmacokinetic studies in rats and dogs.

Moreover, extensive in vitro safety profiling, combined with rat and dog toxicology studies, demonstrated that

NITD-688 (2) was well tolerated after 7-day repeat dosing. With its pan-serotypic activity, efficacy in both acute

and delayed in vivo models, and favorable pharmacokinetic properties and safety profile, NITD-688 (2) was

demonstrated to be a promising preclinical candidate for the treatment of dengue fever.

References

1) Moquin, S.A. et al. Sci. Transl. Med. 2021, 13(579): eabb2181.

EFMC-ISMC | 305

O002

DESIGN AND SYNTHESIS OF NOVEL SELENOESTERS AND

SELENOCYANATES FOR THE TREATMENT OF CHAGAS DISEASE

Verónica Alcolea (1), Alejandro Jiménez-Sánchez (2), Clotilde Marín (2), Silvia Pérez-Silanes (1)

1) Universidad de Navarra, ISTUN Instituto de Salud Tropical, Irunlarrea 1, 31008, Pamplona, Spain; School of Pharmacyand Nutrition, Department of Pharmaceutical Technology and Chemistry, Universidad de Navarra, Campus Universitario,

31008, Pamplona, Spain2) Departamento de Parasitología, Instituto de Investigacion Biosanitaria Ibs, University of Granada, Severo Ochoa s/n,

E-18071, Granada, Spain.

Chagas disease, caused by the parasite Trypanosoma cruzi (T. cruzi) is one of the most prevalent tropical

neglected diseases and causes high mortality and morbidity in endemic countries. Current treatments for this

disease, nifurtimox and benznidazole, are ineffective in the chronic phase and produce severe adverse effects.

Therefore, novel therapies are urgently required. The trace element selenium (Se) has an important role in human

health, due to its antioxidant, antiinflammatory, and pro-immune properties. Recently, novel Se-containing

compounds with in vitro activity against T. cruzi have been described (1). Among them, selenocyanate

derivatives showed the best results, with IC

50 

values in the nanomolar range and high selectivity indexes.

Therefore, in this work, we have designed and synthesized a series of novel selenocyanates with potential

activity against T. cruzi. The molecules also include a group selenoester as a strategy to potentiate the effect and

introduce structural variability. The novel compounds were tested for their anti-T. cruzi activity and their

cytotoxicity was evaluated in Vero cells to establish their selectivity indexes. Besides, their potential against 

Leihsmania major was also explored. 

References

1) Alcolea V. and Pérez-Silanes S.Selenium as an interesting option for the treatment of Chagas disease: A review. European

Journal of Medicinal Chemistry. 2020;206:112673.

306 | EFMC-ISMC

O003

DESIGN AND SYNTHESIS OF MANNICH BASE-TYPE DERIVATIVES

CONTAINING IMIDAZOLE AND BENZIMIDAZOLE AS LEAD

COMPOUNDS FOR DRUG DISCOVERY IN CHAGAS DISEASE

Mercedes Rubio-Hernández (1), Iván Beltran-Hortelano (1), Richard L. Atherton (2), Julen Sanz-Serrano

(3), Verónica Alcolea (1), John M. Kelly (2), Francisco Olmo (2), Silvia Pérez-Silanes (1)

1) Universidad de Navarra, ISTUN Instituto de Salud Tropical; School of Pharmacy and Nutrition, Department ofPharmaceutical Technology and Chemistry, Campus Universitario, 31008, Pamplona, Spain.

2) Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK.3) Universidad de Navarra, Pharmacy and Nutrition Faculty, Department of Pharmacology and Toxicology, Irunlarrea 1,

31008, Pamplona, Spain.

The protozoan parasite Trypanosoma cruzi is the causative agent of Chagas Disease, the most important parasitic

infection in Latin America. The only treatments currently available are nitro-derivative drugs that are

characterised by high toxicity and limited efficacy. Therefore, there is an urgent need for more effective, less

toxic therapeutic agents. We have previously identified the potential for Mannich [1,2] base derivatives as novel

inhibitors of this parasite. To further explore this family of compounds, we synthesized a panel of 69 new

analogues, based on multi-parametric structure-activity relationships, which allowed optimization of both

anti-parasitic activity, physicochemical parameters and ADME properties. Additionally, we optimized our invitro screening approaches against all three developmental forms of the parasite, allowing us to discard the least

effective and trypanostatic derivatives at an early stage. We ultimately identified derivative 3c, which

demonstrated excellent trypanocidal properties; both its druggability and low-cost production make this

compound a promising candidate for the preclinical, in vivo assays of the Chagas disease drug-discovery

pipeline.

References

1) Moreno-Viguri E, Jiménez-Montes C, Martín-Escolano R et al. J. Med. Chem. 2016, 59, 10929-10945.

2) Paucar R, Martín-Escolano R, Moreno-Viguri E et al. Bioorg & Med Chem. 2019, 27, 3902-3917.

EFMC-ISMC | 307

O004

IN SITU CLICK CHEMISTRY APPLIED TO BUNYAVIRALES: FROM

CONVENTIONAL DRUG DESIGN TO ENZYMES ASSEMBLING

THEIR OWN INHIBITORS

Laura Garlatti, Mikael Feracci, Sergio Hernandez, Bruno Canard, Juan Reguera, François Ferron,

Karine Alvarez

AFMB, AMU, CNRS UMR 7257, 163 Avenue de Luminy, Campus de Luminy, 13288 Marseille

With a worldwide repartition and limited therapeutic options reported, neglected Bunyavirales viruses represent

a major public health issue. The replication machinery of these viruses is governed by the intricate L-protein that

displays RNA-dependent RNA-polymerase activity (RdRp) and endonuclease (EndoN) activity in its N-terminal

end.

1,2

This key protein, responsible for the crucial cap-snatching mechanism that allows the viral transcription,

was identified as a promising target to develop pan-genus antivirals. Its catalytic mechanism of RNA hydrolysis

mediated by Mg

2+

ions enables the development of diketo-acids (DKA) metal-chelating inhibitors.

3,4

Recently, rational drug-design concepts have made their way with the emergence of Target-Guided-Synthesis

(TGS), a powerful method that directly involves the target that assembles its own inhibitors In Situ like LEGOs®

. This strategy was successfully employed for the development of few therapeutic molecules.

5

Herein, we describe the use of Bunyavirales EndoN active sites as reaction vessels for the In Situ generation of

their own highly specific metal-chelating inhibitors. DKA anchor molecules bearing an azide moiety were

synthesized using an optimized pathway and EndoNs affinity towards them was assessed. The In Situ Click

Chemistry experiment was designed in 96-well plates using biochemical conditions and various combinations of

DKA-azide and alkyne fragments to produce 1,4-triazolyl-diketo-acids (NT-DKAs) in the presence of the

enzyme. Treatment of the reaction was performed using Ultrafiltration (UF). HIT molecules resulting from this

fragment-based screening are identified by HPLC-MS using an optimized method and will be synthesized on

large scale to enabled their full biological characterization by TSA, FRET or mini-genome assay.

References

1) Reguera, J. et al., PLoS Pathog., 2010, 6(9), e1001101

2) Morin, B. et al., PLoS Pathog., 2010, 6(9), e1001038

3) Saez-Ayala, M. et al., IUCrJ, 2018, 5(2), 223-235

4) Saez-Ayala, M. et al., Antivir. Res., 2019, 162, 79-89

5) Prakasam, T. et al., Chem. Rev. 2013, 113(7), 4905–4979

308 | EFMC-ISMC

O005

DEVELOPMENT OF INNOVATIVE MICROARRAYS TO DETECT

INTERACTION OF SMALL MOLECULES WITH ZIKA VIRUS

STRUCTURAL PROTEINS

Francisca Carvalhal (1,2), Akul Mehta (3), Ana Rita Neves (1,2), Andreia Palmeira (1,2), Emília Sousa

(1,2), Marta Correia-da-Silva (1,2), Richard Cummings (3)

1) Laboratory of Organic and Pharmaceutical Chemistry/ Department of Chemical Sciences, FFUP - Faculty of Pharmacy,University of Porto, Rua Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal

2) CIIMAR – Interdisciplinary Centre of Marine and Environmental Research, Terminal de Cruzeiros do Porto de Leixões,Matosinhos, Portugal

3) Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, CLS 11087 - 3 Blackfan Circle,Boston, MA 02115, USA

Zika virus (ZIKV) infection is a “neglected tropical disease” associated with brain development abnormalities

and fetal death

1

. Since its first isolation in 1947 in Africa

2

, ZIKV was ignored, until its major outbreak in the

Americas in 2015, when WHO declared Public Health Emergency of International Concern

1

. There is still no

specific medicine or vaccine to treat ZIKV infection, leading to the urgent need to improve our understanding of

ZIKV pathogenesis and therapeutic strategies

1

.

ZIKV is a member of the virus family Flaviviridae and is an RNA virus. ZIKV RNA is translated into a single

polyprotein encoding three structural proteins: (i) capsid (C), (ii) membrane (M) generated from its precursor

pre-membrane (prM), (iii) and envelope (E). These form the virus particle, along with seven nonstructural

proteins (NS1, NS2A, NS2B, NS3, NS4A, NS4B, and NS5), that assist in replication and packaging of the

genome, as well as in subverting the host pathways in favor of the virus

3

. To trigger viral infection,

glycosaminoglycans displayed at the surface of host cells must form an initial complex with proteins embedded

on the envelope of the viral particle, similar to other enveloped RNA viruses

4

.

In our group, we have obtained several small molecules in the last decade, and promising hits were discovered

particularly with anticoagulant

5

, antiviral

6

, and antifouling activity

7

. In this work, new compounds

functionalized with primary amines were synthesized (Figure 1A) and immobilized on NHS-ester microarray

slides (Figure 1B). An innovative microarray technique was developed to explore the putative binding of these

small molecules to ZIKV structural proteins present in ZIKV virus-like particles (VLP).

Figure 1. A) Small molecules functionalized with primary amines. B) Immobilization of compounds with

primary amine in NHS-ester slides.

A bile acid derivative hit compound was able to bind to the structural proteins of ZIKV in a dose-dependence

manner. Following, virtual studies predicted the ability of this small molecule to interact with the envelope

protein of ZIKV. Taken together, development of these small molecules and subsequent microarray technology

could yield potential therapeutic or diagnostic tools.

Acknowledgements: This research was supported by national funds through FCT- Foundation for Science and

Technology within the scope of UIDB/04423/ 2020 and UIDP/04423/2020 and under the Project

POCI-01-0145-FEDER-028736, co-financed by COMPETE 2020, Portugal 2020 and ERDF. FC also

acknowledges Luso-American Foundation for the scholarship Proj.2/2019 and FCT support through the PhD

fellowship 2020.07873.BD.

References

1) Baud, D., et al., Lancet 2017, 390(10107), 2099-2109.

2) Dick, G.W., et al., Trans. R. Soc. Trop. Med. Hyg. 1952, 46(5), 509-520.

3) . Sirohi, D. and R.J. Kuhn, J. Infect. Dis. 2017, 216(suppl10): p. S935-S944.

4) Pomin, V.H., Interdiscip. Perspect. Infect. Dis. 2017, 8.

5) a) Correia-da-Silva, M., et al. J. Med. Chem. 2011, 54(1), 95-106. b) Correia-da-Silva, M., et al., J. Med. Chem. 2011,

54(15), 5373-84. c) Correia-da-Silva, M., et al. Eur. J. Med. Chem. 2011, 46(6), 2347-58.

6) Lima, R.T., et al. Chem. Biol. Drug Des. 2013, 81(5), 631-44.

7) a) Almeida, J.R., et al. Sci. Rep. 2017, 7, 42424. b) Neves, A.R., et al. Ecotoxicol. Environ. Saf. 2020, 187, 109812.

EFMC-ISMC | 309

O006

ILLUMINATING THE RARE DISEASE PROTEOME

Tudor Oprea

Translational Informatics Division, University of New Mexico School of Medicine, Albuquerque NM 87131, USA

More than 10,000 rare diseases (RDs) have been identified (1). Of these, only a small number have appropriate

therapies (2). The Illuminating the Druggable Genome (IDG) program (3) has developed a number of integrative

approaches, with focus on the rare disease proteome (4). Of the 20412 human proteins curated in UniProt (5),

6143 are associated with RDs. Of these, 462 are Mode of Action drug targets ("Tclin"), and 868 are associated

with active small molecules ("Tchem"). Another 331 proteins are significantly understudied ("Tdark").

Pathways to illuminate the RD proteome include associations (or lack thereof) with significant mouse

phenotypes, based on International Mouse Phenotype Consortium data (6), as well as associations with known

drug data extracted from DrugCentral (7). The systematic illumination of the RD proteome will be discussed

with examples from the Tclin (therapeutically actionable) and Tdark (high risk) categories.

These examples include the orphan G-protein coupled receptor GPR153, associated with childhood-onset

schizophrenia (Tdark); and the Tumour-associated calcium signal transducer 2 (TACSTD2; Tclin), a drug target

used in triple-negative breast cancer, which is associated with gelatinous drop-like corneal dystrophy. As the

IDG subset of Reactome (8) shows TACSTD2 involvement in the "formation of the cornified envelope" (9), this

suggests that the humanized anti-TACSTD2 antibody may be suitable for eye drop formulation. This antibody

and its conjugate govitecan are FDA-approved under the brand name Trodelvy. At the other end of the drug

repurposing spectrum, childhood-onset schizophrenia - a very rare and severe chronic psychiatric condition (10)

- could perhaps be managed by finding suitable drugs to block GPR153.

References

1) Haendel M. et al., Nature Rev Drug Discov 2020, 19:77-78

2) Fetro C, Scherman D. Therapie 2020, 75:157-160

3) Oprea T.I., et al., Nature Rev Drug Discov 2018, 17:317-332

4) Tambuyzer E., et al, Nature Rev Drug Discov 2020, 19:93-111

5) UniProt Consortium. Nucleic Acids Res. 2019, 47:D506-D515

6) Koscielny G, et al., Nucleic Acids Res. 2014, 42:D802-D809

7) Avram S, et al., Nucleic Acids Res. 2021, 49:D1160-D1169

8) Jassal B, et al., Nucleic Acids Res. 2020, 48:D498-D503

9) https://idg.reactome.org/PathwayBrowser/#/R-HSA-6805567&PATH=R-HSA-1266738

10) Fernandez A., et al., Front Genet. 2019, 10:1137

310 | EFMC-ISMC

O007

RATIONALIZING THE MOLECULAR INTERACTIONS BETWEEN

HUMAN AND VIRAL KEY ELEMENTS INVOLVED IN SARS-COV-2

INFECTION: LIMITS AND SUCCESSES TO DESIGN A BETTER

FUTURE

Marco Marazzi (1), Cristina García-Iriepa (1), Antonio Francés-Monerris (2,3), Cécilia Hognon (3), Tom

Miclot (3,4), Isabel Iriepa (5), Giampaolo Barone (4), Antonio Monari (3)

1) Dept. of Analytical Chemistry, Physical Chemistry and Chemical Engineering & IQAR, Universidad de Alcalá, Madrid,ES

2) Dept. de Química Física, Universitat de València, 46100 Burjassot, ES3) Université de Lorraine and CNRS, LPCT UMR 7019, F-54000 Nancy, FR

4) Dept. of Biological, Chemical and Pharma. Sciences and Technologies, Università degli Studi di Palermo, IT5) Dept. of Organic & Inorganic Chemistry & IQAR, Universidad de Alcalá, Madrid, ES

The coronavirus disease (COVID-19) global pandemic caused by the severe-and-acute respiratory syndrome

coronavirus 2 (SARS-CoV-2) has largely spread worldwide in 2020 and 2021 constituting a major public health

threath. The occurrence of severe respiratory syndromes and major inflammatory responses, especially in elderly

patients or in subjects presenting comorbidities, is at the base of the pressure imposed by SARS-CoV-2

infections on the public health systems. This is also aggravated by the high infectivity of SARS-CoV-2 and the

appearance of apparently more transmissible variants,(1) that may result in uncontrolled bursts of infections

hampering the pandemic control.

Here, we show the results obtained by our international task force by applying a combination of computational

and simulation methods (blind docking, extensive all-atom molecular dynamics, free energy calculations), aimed

at first at rationalizing the molecular recognition of the Angiotensin-Converting Enzyme 2 (ACE2) by the virus

Receptor Binding Domain (RBD), i.e. explaining the basis of the chemical interactions happening at the virus

gate entry into the human body.(2) Moreover, as a general recognition mechanism common to all coronaviruses,

we described the energetics of the human G-quadruplex RNA trapping into the viral SARS-Unique Domain,

SUD (see Figure), i.e. a part of the non-structural protein 3 (Nsp3), thus hindering at least partially the human

immune system response.(3)

Further, such deep and detailed rationalization gave us the opportunity to focus on the search of drugs possibly

limiting or hampering the SARS-CoV-2 effects, mainly dedicating our efforts to small drugs repurposing.

Especially, the effects of different aureolic acids, flavonoids,(2) and ivermectin(4) were and are under

investigation, with the aim of interfering with the ACE2/RBD and RNA/SUD binding interactions. Moreover,

the eventual drug activity with respect to two of the most know virus proteases, 3-Chymotrypsin-Like (3CLpro)

and Papain-Like (PLpro) proteases, was also taken into account.(5)

Finally, the relevance of our in silico approach will be discussed, pointing out limits and potentialities to predict

the effect of present and eventually future virus variants.(6)

References

1) Y. J. Hou, S. Chiba, P. Halfmann, C. Ehre, M. Kuroda, K. H. Dinnon, S. R. Leist, A. Schäfer, N. Nakajima, K. Takahashi,

et al. Science 2020, 370, 1464–1468.

2) C. García-Iriepa, C. Hognon, A. Francés-Monerris, I. Iriepa, T. Miclot, G. Barone, A. Monari, M. Marazzi, J. Phys. Chem.

Lett. 2020, 11, 21, 9272–9281.

3) C. Hognon, T. Miclot, C. García-Iriepa, A. Francés-Monerris, S. Grandemange, A. Terenzi, M. Marazzi, G. Barone, A.

Monari, J. Phys. Chem. Lett. 2020, 11, 5661–5667.

4) A. Francés-Monerris, C. García-Iriepa, I. Iriepa, C. Hognon, T. Miclot, G. Barone, A. Monari, M. Marazzi, Comput.

Struct. Biotechnol. J. 2020, under revision.

5) A. Francés-Monerris, C. Hognon, T. Miclot, C. García-Iriepa, I. Iriepa, A. Terenzi, S. Grandemange, G. Barone, M.

Marazzi, A. Monari, J. Proteome Res. 2020, 19, 11, 4291–4315.

6) A. Francés-Monerris, C. García-Iriepa, I. Iriepa, C. Hognon, T. Miclot, G. Barone, A. Monari, M. Marazzi, Sci. Rep. 2020,

in preparation.

EFMC-ISMC | 311

Posters - Drug Discovery ProjectsQ. Chemical Tools and Drug Discovery for Neuroinflammation

312 | EFMC-ISMC

Q001

LOCKED ANALOGUES OF L-ISOSERINE AS POTENTIAL

SUBTYPE-SELECTIVE GAT3 INHIBITORS

Francesco Bavo (1), Maria E.K. Lie (1), Stefanie Kickinger (2), Gerard F. Ecker (2), Petrine Wellendorph

(1), Bente Frølund (1)

1) University of Copenhagen, Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, 2100Copenhagen, Denmark

2) University of Vienna, Department of Pharmaceutical Chemistry, Althanstrasse 14, 1090 Vienna, Austria

In the brain, extrasynaptic γ-aminobutyric acid (GABA) maintains a persistent “tonic” inhibition through the

activation of GABA-A receptors. Extrasynaptic GABA levels are mainly regulated by the astrocytic GABA

reuptake transporters (GATs) GAT3 and BGT1.[1] During an ischemic stroke, the surface expression of GAT3

is reduced, causing increased tonic inhibition.[2] Augmented extrasynaptic levels of GABA leads to

neuroprotection in the acute phase, but impairs the formation of new structural and functional circuits required

for recovery in the chronic phase. Oppositely, reduced tonic inhibition after a stroke facilitates functional

recovery.[3]

In 2017, Lie et al. demonstrated that post-stroke delayed administration of L-isoserine, a moderately selective

GAT3 substrate inhibitor, increases GAT3 expression, reduces tonic inhibition and facilitates post-stroke

functional recovery in mice models.[2]

Therefore, L-isoserine is a useful scaffold for the investigation of the SARs that underlie GAT3 inhibition and

subtype-selectivity.

Particularly, this part of the project is dedicated to the identification of locked analogue of L-isoserine with

increased GAT3 potency and selectivity, that will represent a unique platform for further investigation of the

SARs.

Upon assessment of GAT3 inhibition and subtype-selectivity in the [3H]GABA competition uptake assay, the

most promising compounds will be evaluated in the FLIPR membrane potential assay to define their activity as

substrate or non-substrate inhibitors and further evaluated for in-vivo activity in post-stroke animal models.

References

1) Schousboe, A; Larsen, PK et al. Astrocytic GABA Transporters: Pharmacological Properties and Target for Antiepileptic

Drugs. Adv Neurobiol. 2017, 16, 283–296.

2) Lie, MEK; Clarkson, AN et al. GAT3 Selective Substrate L-Isoserine Upregulates GAT3 Expression and Increases

Functional Recovery after a Focal Ischemic Stroke in Mice. J. Cereb. Blood Flow Metab. 2017, 39, 74-88.

3) Clarkson, AN; Carmichael, ST et al. Reducing Excessive GABA-Mediated Tonic Inhibition Promotes Functional

Recovery after Stroke. Nature, 2010, 468, 305–309.

EFMC-ISMC | 313

Q002

NEW BRAIN PENATRANT COMPOUNDS IN ADVANCED STUDIES:

MULTIPOTENCY SCREENING AND EVALUATION AGAINST

PARKINSON'S DISEASE

Nikolay Tzvetkov (1), Andrew Lowerson (2), Steve Brough (2)

1) NTZ Lab Ltd., Krasno selo 198, 1618 Sofia, BG, [email protected]) Key Organics Ltd, Highfield Road Industrial Estate, Camelford, Cornwall, PL32 9RA, UK

A new collection of structurally optimized compounds addressing the central nervous system

(CNS) diseases, which have been recently entered into a development and marketing

agreement between Key Organics Ltd. and NTZ Lab Ltd., is now available in the BIONET

collection [1]. This screening collection with experimentally-determined bioactivity, ADMET,

bioavailability, and toxicity will address the need of new, robust, and multi-potent small

molecules for the treatment and diagnosis of CNS diseases, such as Parkinson´s disease,

Alzheimer´s disease, dementia and/or other neurodegenerative diseases

References

1) Tzvetkov, N. T. PCT Patent WO 2014/107771 A1, NTZ Lab Ltd., 2014.

2) Tzvetkov,et al. Eur. J. Med. Chem., 2017, 127, 470–492.

3) Tzvetkov,et al. J. Med. Chem., 2014, 57, 6679–6703

3) Tzvetkov,et al. J. Enz. Inh. Med. Chem., 2017, 32, 960–967.

5) www.biosolveit.de/SeeSAR (SeeSAR v.8.0, 2018).

314 | EFMC-ISMC

Q003

DEVELOPMENT OF NOVEL NECROPTOSIS INHIBITORS:

TARGETING RIPK1

Lara Fidalgo, Rui Moreira, Cecília Rodrigues, Ana S. Ressurreição

iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa. Av. Professor Gama Pinto, 1649-003 Lisbon, Portugal

Necroptosis, a regulated form of necrosis, is the major mechanism of cellular death upon extracellular

inflammatory signalling and is crucially dependent on the kinase activity of RIPK1 and its downstream

mediators: RIPK3 and pseudokinase MLKL. Consequently, RIPK1 has emerged as a promising therapeutic

target for the treatment of a wide range of human neurodegenerative, autoimmune, and inflammatory diseases.

[1-6]

To address the lack of chemotypes targeting necroptosis and RIPK1, our group recently developed a phenotypic

high-throughput screening strategy to identify novel necroptosis inhibitors. This collaborative effort led to the

discovery of promising compounds possessing different scaffolds from those reported in the literature.

Taking into account the gathered knowledge, and following Computer-Aided Drug Design (CADD)

investigations, we have synthetized a highly diversified library of thiazole-based analogues in order to identify

structural features responsible for RIPK1 potency and selectivity.

Acknowledgements: We thank the Fundação para a Ciência e Tecnologia for financial support through the PhD

fellowship grant PD/BD/143157/2019 awarded to Lara Fidalgo.

References

1) Yuan, J.; Amin, P.; Ofengeim, D. Necroptosis and RIPK1-Mediated Neuroinflammation in CNS Diseases. Nat. Rev.

Neurosci. 2018, 20 (1), 19–33

2) Degterev, A.; Ofengeim, D.; Yuan, J. Targeting RIPK1 for the Treatment of Human Diseases. Proc. Natl. Acad. Sci. 2019,

116 (20), 9714-9722

3) Harris, P. A.; Marinis, J. M.; Lich, J. D.; Berger, S. B.; Chirala, A.; Cox, J. A.; Eidam, P. M.; Finger, J. N.; Gough, P. J.;

Jeong, J. U.; et al. Identification of a RIP1 Kinase Inhibitor Clinical Candidate (GSK3145095) for the Treatment of

Pancreatic Cancer. ACS Med. Chem. Lett. (2019), 10 (6), 857–862.

4) Brito, H.; Marques, V.; Afonso, M. B.; Brown, D. G.; Börjesson, U.; Selmi, N.; Smith, D. M.; Roberts, I. O.; Fitzek, M.;

Aniceto, N.; et al. Phenotypic High-Throughput Screening Platform Identifies Novel Chemotypes for Necroptosis Inhibition.

Cell Death Discov. (2020), 6 (1)

EFMC-ISMC | 315

Q004

NOVEL THIOMORPHOLINE DERIVATIVES WITH INCREASED

ANTIOXIDANT ACTIVITY AGAINST NEURODEGENERATION

Ariadni Tzara, Angeliki Kourounakis

Department of Medicinal Chemistry, School of Pharmacy, National and Kapodistrian University of Athens*[email protected]

As scientific research evolves, more and more complex diseases, such as metabolic syndrome, cancer and

neurodegeneration, reveal their multifactorial nature, shifting research interest towards a multitargeted approach.

Compounds bearing a combination of two pharmacophores, each endowed with different pharmacological

properties, seem to be a more suitable approach to tackle such pathological conditions. This project involves a

series of amides, combining acids with known antioxidant and anti-inflammatory properties and a potent

thiomorpholinic squalene synthase (SQS) inhibitor, previously designed and studied by our research group

1

.

Apart from acting as antihyperlipidemic agents, SQS inhibitors have been further correlated with various other

potential therapeutic targets, among them Alzheimer’s disease (AD); imbalance in cell cholesterol levels may

lead to alterations in neuronal cell membranes, influencing neuronal survival and therefore the course of AD

2

.

Additional acetylcholinesterase inhibitory properties, as part of a multitargeting approach, may assist the

neuroprotective action of the cholinergic system against AD neurological events such as neuroinflammation, Aβ

accumulation, and abnormal tau phosphorylation

3

.

The newly designed compounds were synthesized in good yields, characterized via

1

H and

13

C NMR

spectroscopy and pharmacologically evaluated- both in vitro and in vivo - for their antioxidant,

anti-inflammatory and anti-neurodegenerative activity. All new derivatives successfully maintained or even

exceeded the antioxidant activity of their parent molecules, in corresponding assays of free radical scavenging

and lipid peroxidation. They also demonstrated satisfactory results in an in vitro lipoxygenase (LOX) inhibition

assay, bearing IC

50

values 2 to 4-fold lower than the parent compounds, whereas one derivative showed a

significant improvement of activity, with an IC

50

value lowered by two orders of magnitude. The new

compounds also reduced in vivo carrageenan-induced paw edema by 35-65%. Finally, the preliminary results of

the in vitro anti-neurodegenerative related assays of iron chelation (ferrozine assay) and acetylcholinesterase

inhibition are considered to extend the design purpose of the new compounds, rendering them a promising

approach for further investigation.

References

1) Matralis, AN; Kourounakis, AP. ACS Med. Chem. Lett. 2019, 10(1), 98-104

2) Kourounakis AP; Bavavea E. Arch. Pharm. (Weinheim) 2020, 353(9):e2000085

3) Douchamps, V; Mathis, C. Behav. Pharmacol. 2017, 28, 112-123

316 | EFMC-ISMC

Q005

NEW BIOLOGICAL ACTIVE HYBRIDS COMBINING ENONE AND

PYRIMIDINE SCAFFOLDS

Thalia Liargkova, Dimitra Hadjipavlou-Lititna,

Department of Pharmaceutical Chemistry, School of Pharmacy, Faculty of Health Sciences, Aristotle University ofThessaloniki, 54124, Thessaloniki, [email protected], [email protected]

Chalcones are biogenetic precursors of flavonoids in higher plants displaying a wide variety of pharmacological

properties. They are well known intermediates for the synthesis of various heterocyclic compounds. Cyclization

of chalcones, leading to thiazines, pyrimidines, pyridazines, attracted a developing research increase within the

heterocyclic chemistry for several years due to their quick accessibility and the broad spectrum of biological

activities. These observations led us to synthesize [1, 2, 3, 4] new bis-chalcones and their corresponding

bis-pyrimidines in the direction of new bioactive hybrids. The new compounds were spectrometrically identified.

Chemistry: [1, 2 ,3, 4]

 

The compounds were tested in vitro for their ability to: a) inhibit in vitro AchE, b) inhibit lipid peroxidation of

linoleic acid, c) inhibit soybean lipoxygenase and d) interact with DPPH. The results were discussed in terms of

structural characteristics and physicochemical properties of the molecules.

References

1) Liargkova T, Eleftheriadis N, Dekker F, Voulgari E, Avgoustakis C, Sagnou M, Mavroidi B, Pelecanou M,

Hadjipavlou-Litina D.” Small Multitarget Molecules Incorporating the Enone Moiety”. Molecules. 2019 Jan 7; 24(1):199

2) Liargkova T., Hadjipavlou-Litina D.J., koukoulitsa C., Voulgari E., Avgoustakis C., J Enzyme Inhib Med Chem, 2015,23,

1-12

3) Asiri A.M., Khan S.A., “Synthesis and Anti-Bacterial Activities of a Bis-Chalcone Derived from Thiophene and Its

Bis-Cyclized Products”, Molecules 2011, 16, 523-531

4) Sodani R.S., Choudhary P.C., Sharma H.O., Verma B.L., E-journal of Chemistry, 2010, 7(3), 763-769

EFMC-ISMC | 317

Q006

MOLECULAR MODELLING AND DESIGN OF NOVEL

SUBNANOMOLAR AMPA RECEPTOR MODULATORS

Vladimir A. Palyulin (1), Mstislav I. Lavrov (1), Dmitry S. Karlov (1), Eugene V. Radchenko (1), Elena B.

Averina (1), Kseniya N. Sedenkova (1), Dmitry A. Vasilenko (1), Anna A. Nazarova (1), Nadezhda S.

Temnyakova (1), Polina N. Veremeeva (1), Vladimir L. Zamoyski (2), Vladimir V. Grigoriev (1,2)

1) Department of Chemistry, Lomonosov Moscow State University, Moscow, 119991, Russia2) Institute of Physiologically Active Compounds RAS, Severny Proezd 1, Chernogolovka, Moscow Region, 142432, Russia

A growing attention is attracted recently to AMPA receptors as a promising target for the development of drugs

for the treatment of serious neurological and psychiatric disorders, such as schizophrenia, depression, multiple

sclerosis, age-related cognitive and memory disorders, Parkinson's disease, Alzheimer's disease. The positive

AMPA receptor allosteric modulators reveal such neurophysiological effects as induction of long-term

potentiation of synaptic excitation, considered as a substrate for learning and memory, and significant increase of

nerve growth factors expression. This makes them promising compounds for the development of nootropic

agents and neuroprotectors. The negative modulators of AMPA receptors are also applicable as drugs and can be

used as antiepileptics.

In this communication, a combined approach to the de novo design of AMPA receptor modulators is considered

including molecular modelling and molecular dynamics simulation of modulator-agonist-receptor complexes.

That allowed us to find both positive and negative highly potent allosteric modulators of AMPA receptors with

new scaffolds. They include derivatives of 3,7-diazabicyclo[3.3.1]nonane, substituted bis(pyrimidines) and

bis-amides with various linkers/spaces. Convenient synthetic approaches were elaborated for the designed

compounds. Electrophysiological patch clamp experiments have demonstrated the pronounced influence of the

studied compounds in sub-nanomolar concentrations on the kainate-induced currents recorded for Purkinje

neurons from rat cerebellum. A series of other in vitro and in vivo studies has shown neuroprotective and

cognition-enhancing properties for the positive modulators selected in the patch clamp studies. Several designed

compounds have successfully passed preclinical studies.

This work was supported by the Russian Science Foundation (grant No. 17-15-01455).

 

References

1) Radchenko E.V., Karlov D.S., Lavrov M.I., Palyulin V.A., Mendeleev Commun., 2017, 27, 623.

2) Nazarova A.A., Sedenkova K.N., Karlov D.S., Lavrov M.I., Grishin Y.K., Kuznetsova T.S., Zamoyski V.L., Grigoriev

V.V., Averina E.B., Palyulin V.A., MedChemComm, 2019, 10, 1615.

3) Lavrov M.I., Karlov D.S., Voronina T.A., Grigoriev V.V., Ustyugov A.A., Bachurin S.O., Palyulin V.A., Mol. Neurobiol.,

2020, 57, 191.

318 | EFMC-ISMC

Q007

NOVEL COMPOUNDS WITH ANTIOXIDANT PROPERTIES

CONTAINING CNS-ACTING MOIETIES AGAINST

NEURODEGENERATION

Georgios Papagiouvannis (1), Panagiotis Theodosis-Nobelos (1), Eleni A. Rekka (2)

1) Department of Pharmacy, School of Health Sciences, Frederick University, Nicosia, 1036, Cyprus2) Department of Medicinal Chemistry, School of Pharmacy, Faculty of Health Sciences, Aristotle University of Thessaloniki,

Thessaloniki 54124, Greece

Alzheimer’s Disease (AD) is one of the most common neurodegenerative disorders and a major health problem

to society, affecting millions of people worldwide. It has a multifactorial character and is developed as a

complex network of interconnected events leading to the evolution of the disease. Thus, the concept of the

multitarget approach is particularly applicable to AD and the neurodegenerative disorders in general.

Oxidative stress is one of the main causes of neuronal death in AD and oxidative damage is a key process in AD

pathogenesis. Increased hydrogen peroxide formation and elevated free iron concentrations, due to decreased

amount of ferritin, observed in AD patients, generate more reactive oxygen species (ROS). ROS oxidise lipids

and damage membranes in the AD brain. Protein, DNA and RNA oxidation products are increased in several

brain regions in AD patients. Inflammation and glia activation are observed in AD patients; thus, inflammation is

a key target in AD drug development. 5-Lipoxygenase is overexpressed in AD and contributes to neuronal

vulnerability. Moreover, acetylcholinesterase is currently the main pharmacological target against AD, since

cholinergic neuron loss is one of the most important factors, contributing to AD development.

In this research, we have designed and synthesised novel compounds that contain phenolic acids with antioxidant

activity, such as trolox or ferulic acid and moieties, such as gabapentin and nipecotic acid, aiming to multitarget

ligand design against neuron loss. Gabapentin is a neuroprotective agent and nipecotic acid acts as a GABA

reuptake inhibitor. The compounds were synthesized by amidation reactions using

N,N-dicyclohexylcarbodiimide (DCC) or carbonyldiimidazole (CDI) as coupling agents. They were purified by

flash column chromatography and identified spectrometrically (

1

H-NMR,

13

C-NMR, MS)

The synthesised compounds were found to have in vitro antioxidant activity as lipid peroxidation inhibitors,

DPPH radical scavengers and oxidative protein glycation inhibitors, to inhibit lipoxygenase and cycloxygenase

activity and to exert in vivo anti-inflammatory activity, assessed as paw oedema reduction. Furthermore, some of

them could inhibit acetylcholinesterase.

With the design of the described derivatives, we aimed to compounds that would acquire a series of biological

properties able to prevent or restore several pathological changes implicated in AD and appearing in the

demented brain. This study has demonstrated that, in general, the synthesised compounds possess a combination

of the desired properties integrated in their molecules.

 

EFMC-ISMC | 319

Q008

BIVALENT LIGANDS AS PHARMACOLOGICAL TOOLS FOR

STUDYING THE ROLE OF SIGMA-1 RECEPTOR AGONISM IN

NEUROPROTECTION AND NEURODEGENERATIVE DISEASES

Giacomo Rossino (1), Pasquale Linciano (1), Marta Rui (1), Daniela Rossi (1), Marco Peviani (2), Dirk

Schepmann (3), Bernhard Wuensh (3), Julio Caballero (4), Daniela Curti (2), Simona Collina (1)

1) Department of Drug Science, University of Pavia, viale Taramelli 12, Pavia, 27100, Italy2) Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, via Ferrata 9, Pavia, 27100, Italy

3) Institute of Pharmaceutical and Medicinal Chemistry, University of Muenster, Correnstrasse 48, 48149 Munster, Germany4) Center for Bioinformatics and Molecular Simulation, Universidad de Talca, 1 Poniente, 1141 Talca, Chile

Sigma-1 receptors (S1R) are chaperone proteins highly express in central nervous system cells. Activation of

S1Rs is associated with neuroprotection and neuroplasticity, and promotes neuronal survival via multiple

mechanisms, including stimulation of mitochondrial functions, decrease of the oxidative stress, and suppression

of the neuroinflammatory cascade. For this reason, S1R are recognized as important drug targets and they were

deeply investigated over the past 20 years for the identification of novel drugs for the treatment of

neurodegenerative diseases.

1

 Although, thousands of S1R ligands were disclosed, the biochemical basis for

agonism or antagonism at the S1R still remain a primary challenge. Stuides suggested that the activity of S1R

might be related to its oligomerization state. Agonists may favour S1R dimer and monomer forms that represent

the functional chaperone states, whereas antagonists may stabilize S1R in inactive higher oligomer complexes,

that may act as a repository for the active forms. Moreover, the recent disclosure of the crystal structures of S1R

in complex with agonists and antagonists has lifted the veil on additional riddles. Indeed, the crystal structures

showed that the binding pocket of S1R is highly occluded from solvent; therefore, it is not clear how ligands

access this site. In our previous work, we applied molecular dynamics simulations to open the occluded

ligand-binding pocket in the S1R crystal structure and to determine the preferred ligand pathway to enter and

exit the binding site.

2

However, the mechanism for the recognition of the ligand and its access into the binding

site is still unclear.

Based on the previous outcomes and considerations, we suggest the presence of a ligand recognition site on the

surface of S1R, necessary for selecting ligands that will enter into the binding site. By following the same

approach previously reported,2 a series of homo- and hetero-bivalent compounds were designed and synthesized

as pharmaceutical tools to verify the proposed mechanism and to shed light on the molecular basis behind S1R

activity. The bivalent ligands were designed so that the first monomer is identified by the recognition site, it

enters the binding site, and places the second monomer near the residues that recognize the ligands. Such

bivalent symmetric and asymmetric ligands were designed using as template (R)-RC-33, our best-in-class S1R

agonist.

3

Diverse linkers were exploited to study the distance and the position of the recognition site with the

respect to the binding site. Since (R)-RC-33 presents one chiral center, an enantioselective synthetic procedure

was set up for the synthesis of the corresponding bivalent ligands. The chiral purity of the intermediates and final

compounds was assessed via chiral HPLC. X-ray crystallography was performed on one representative

compound to verify the retainment of configuration, thus confirming that the products are homochiral, and no

racemization occurred during the synthetic steps. The binding affinity (Ki) was determined for all the synthesized

bivalent compounds and computational studies were performed to rationalize the results. In particular, one

homo-bivalent compound showed a binding affinity comparable with the Ki of the single monomer RC-33. As

stated by molecular dynamic, this derivative is the one with the proper linker length and flexibilit, thus being

capable to fits both the occluded binding site and the recognition site. The other symmetric homo-bivalent

ligands range from moderate to low affinity and this agrees with the computational results, since the second

moiety might not reach the recognition site because of the improper linkers. Similarly, the asymmetric

hetero-bivalent ligands, lacking the second pharmacophoric moiety that might interact with the recognition site,

displayed a significant lower affinity as expected. Lastly, the pharmacological profile of the two bivalent ligands

with low nanomolar affinity was assessed by evaluating the ability to enhance NGF-induced neurite outgrowth

in PC12 cells, confirming a S1R agonist behaviour. Taken together, these results support our initial hypothesis.

The agonist activity is strictly related to the stabilization of the open conformation of the receptor that induces a

conformational change that disrupts the oligomeric architecture. Further studies will be directed to the validation

of the role of S1R agonism as molecular mechanism critically implicated in neuroprotection and related

pathologies.

References

1) Front. Cell. Neurosci. 2018; 12:314

2) J. Chem. Inf. Model. 2020, 60, 2, 756–765

3) Bioorg. Med. Chem. 2013; 21(9):2577-86

320 | EFMC-ISMC

Q009

DISCOVERY OF POTENT AND SELECTIVE A2A ANTAGONISTS

WITH EFFICACY IN ANIMAL MODELS OF PARKINSON'S DISEASE

AND DEPRESSION

Sujay Basu

Eurofins Advinus#21 & 22 Phase II, Peenya Industrial Area

Bangalore 560058, India

Adenosine A2A receptor (A2AAdoR) antagonism is a nondopaminergic approach to Parkinson’s Disease

treatment that is under development. We herein described a novel series of [1,2,4]triazolo[5,1-f]purin-2-one

derivatives that displays functional antagonism of the A2A receptor with a high degree of selectivity over A1,

A2B, and A3 receptors. Compounds from this new scaffold resulted in the discovery of highly potent, selective,

stable, and moderate brain penetrating compound 33. Compound 33 endowed satisfactorily in vitro and in vivo

pharmacokinetic properties. Compound 33 demonstrated robust oral efficacies in two commonly used models of

Parkinson’s Disease (haloperidol-induced catalepsy  and 6-OHDA lesioned rat models) and depression (TST and

FST mice models).

EFMC-ISMC | 321

Posters - Drug Discovery ProjectsR. New Strategies and Agents for Targeting Gram-Negative Pathogens

322 | EFMC-ISMC

R001

TARGETING A PATHOGENIC LECTIN: DESIGN, SYNTHESIS AND

EVALUATION OF BC2L-C INHIBITORS

Rafael Bermeo (1,2), Kanhaya Lal (1,2), Laura Belvisi (2), Anne Imberty (1), Anna Bernardi (2),

Annabelle Varrot (1)

1) Université Grenoble Alpes, CNRS, CERMAV. Grenoble. France2) Department of Chemistry, Università degli Studi di Milano. Milano. Italy

Multi-drug resistant (MDR) pathogens have become a high-profile threat to public health. In particular,

opportunistic MDR pathogens such as Burkholderia cenocepacia are responsible for healthcare-associated

infections and increase mortality, especially for patients admitted with cystic fibrosis or immuno-compromising

conditions. As other opportunistic Gram-negative bacteria, this pathogen establishes virulence and biofilms

through lectin-mediated adhesion. The “anti-adhesion” therapy is a tactic devised to fight against this mechanic:

by inhibiting the lectin-mediated adhesion of bacteria to host tissues, the infectious process is blocked at its

initial stage without leading to selective pressure and resistance.

B. cenocepacia’s BC2L-C is a superlectin featuring two distinct lectin domains, and thus displays dual

carbohydrate specificity.

[1,2]

Consequently, BC2L-C is believed to enable cross-linking of B. cenocepacia to

human epithelial cells during pulmonary infection and “Cepacia Syndrome”: rapid decline of respiratory

function leading to sepsis and high mortality. The interactions between BC2L-C’s N-terminal and human

histo-blood group epitopes is particularly interesting to target for inhibition.

[1]

As an anti-adhesion tactic, we aim

to design glycomimetic inhibitors of BC2L-C-Nter.

Here, we report the extensive structural study of the targeted interaction, which led to 3 crystal structures in

complex with human oligosaccharides (Figure 1A).

[3]

In order to design BC2L-C-Nter antagonists,

computational study of the structures identified ligandable pockets adjacent to the main binding site (Figure 1A

). In turn, fragment screening provided a small library of ligand candidates (Figure 1B), later validated through

crystallography.

[4]

We further report a successful campaign of modular synthesis towards a glycomimetic panel

of C- and N- fucosides (Figure 1C). Evaluation of the generated structures includes STD-NMR, SPR and ITC

experiments against target BC2L-C-Nt. The leading inhibitor provided satisfactory affinity and resulted in a

crystal structure of the antagonist/lectin complex (Figure 1D). With these results, a second generation of

BC2L-C-Nt inhibitors can be designed. Our latest results will be described in the communication.

This project has received funding from the European Union’s Horizon 2020 research and innovation programme under the Marie Skłodowska-Curiegrant agreement No 765581.

References

1) Sulak, O.; Cioci, G. et al. A TNF-like trimeric lectin domain from Burkholderia cenocepacia with specificity for

fucosylated human histo-blood group antigens. Structure 2010, 18, 59–72.

2) Sulak, O.; Cioci, G. et al. Burkholderia cenocepacia BC2L-C is a super lectin with dual specificity and proinflammatory

activity. PLoS Pathog. 2011, 7 (9), e1002238.

3) Bermeo, R.; Bernardi, A.; Varrot A. BC2L-C N-Terminal Lectin Domain Complexed with Histo Blood Group

Oligosaccharides Provides New Structural Information. Molecules 2020, 25(2), 248.

4) Lal, K.; Bermeo, R. et al. Prediction and Validation of a Druggable Site on Virulence Factor of Drug Resistant

Burkholderia cenocepacia. Chem. Eur. J. 2021, https://doi.org/10.1002/chem.202100252

EFMC-ISMC | 323

R002

ASSAULT, SIEGE OR TROJAN HORSE STRATEGY: USE OF

NATURAL PRODUCTS TO FIGHT BACTERIAL INFECTIONS

Mark Brönstrup

Helmholtz Centre for Infection Research, Braunschweig, Germany

 Multidrug resistant bacterial pathogens have become a major health concern. Especially infections by

gram-negative bacteria are challenging, since their complex cell membrane architecture strongly impedes the

uptake of drugs. Because microbial natural products continue to be the prime source to tackle these issues, we

have investigated natural products as the basis for novel antibiotic.

A broad spectrum of gram-positive and gram-negative pathogens is addressed by cystobactamids,

oligo-arylamids originally isolated from Cystobacter sp. Our efforts to optimize the antibiotic properties of the

cystobactamids by medicinal chemistry to in vivo active compounds will be presented.[1]  

Beyond a classic ‘assault’ of bacteria with such antibiotics, the conjugation of natural products to targeting

functions has been beneficial to improve their drug properties.[2] In the so-called Trojan Horse Strategy,

antibiotics are conjugated to siderophores to hijack the bacterial siderophore transport system, and thereby

enhance the intracellular accumulation of drugs.[3] We synthesized novel artificial siderophores, characterized

their transport and resistance mechanisms, and their efficacy when coupled to antibiotic natural products.[4]

Finally, we present a novel approach for the selective bacterial targeting and infection-triggered release of

antibiotic conjugates in the alternative siege concept, using the lipopeptide colistin as the antibiotic effector.[4]

References

1) G. Testolin et al., Chem. Sci. 2020, 11, 1316 – 1334.

2) P. Klahn, M. Brönstrup, Nat. Prod. Rep., 2017, 34, 832 – 885.

3) K. Ferreira et al., Angew. Chem. Int. Ed. 2017, 56, 8272-8276.

4) C. Peukert, L. Pinkert, Y. Lai, W. Tegge, unpublished.

324 | EFMC-ISMC

R003

A STUDY WITH PEPTIDE DENDRIMERS REVEALS AN EXTREME

pH DEPENDENCE OF ANTIBIOTIC ACTIVITY ABOVE pH 7.4

Xingguang Cai (1), Sacha Javor (1), Bee Ha Gan (1), Thilo Kohler (2), Jean-Louis Reymond (1)

1) Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, 3012 Bern,Switzerland

2) Department of Microbiology and Molecular Medicine, University of Geneva, Geneva

Recently we reported antimicrobial peptide dendrimer (AMPD) G3KL and T7 with potent activities against P.aeruginosa and A. baumannii, two of the most problematic antibiotic-resistant nosocomial pathogens

[1][2]

. In our

efforts to develop new AMPDs against Gram-negative bacteria, we investigated their activity at acidic and basic

pH, which correspond to the conditions of the site of bacterial infections on skin or biofilms and chronic wounds

respectively. Removing the eight low pKa amino termini by substituting the N-terminal lysine residues with

aminohexanoic acid in our reference dendrimer G3KL provided the modified peptide dendrimer XC1 with a

broader pH-activity range. Furthermore, we discovered that raising the pH to 8.0 reveals strong activities against 

Klebsiella pneumoniae and methicillin at pH 7.4, an effect also observed with polymyxin B and tentatively

assigned to stronger binding to the bacteria at higher pH as observed with a fluorescence labeled dendrimer

analog. This work has been published in Chemical Communication.

[3]

References

1) M. Stach, T. N. Siriwardena, T. Köhler, C. van Delden, T. Darbre, J.-L. Reymond, Angew. Chem. Int. Ed. 2014, 53,

12827–12831.

2) T. N. Siriwardena, A. Capecchi, B.-H. Gan, X. Jin, R. He, D. Wei, L. Ma, T. Köhler, C. van Delden, S. Javor, J.-L.

Reymond, Angew. Chem. Int. Ed. 2018, 57, 8483–8487.

3) X. Cai, S. Javor, B.-H. Gan, T. Kohler, J.-L. Reymond, Chem. Commun. 2021, 10.1039.D1CC01838H.

EFMC-ISMC | 325

R004

NON-HEMOLYTIC ANTIMICROBIAL PEPTIDES DESIGNED BY

MACHINE LEARNING

Xingguang Cai (1), Alice Capecchi (1), Hippolyte Personne (1), Thilo Kohler (2,3), Christian van Delden

(2,3), Jean-Louis Reymond (1)

1) Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, 3012 Bern,Switzerland

2) Department of Microbiology and Molecular Medicine, University of Geneva3) Service of Infectious Diseases, University Hospital of Geneva, Geneva, Switzerland

Antimicrobial peptides (AMPs) offer a unique opportunity to address antibiotic resistance, which is one of the

major global public health threats [1]. Most AMPs are membrane disruptive amphiphiles, unfortunately, this

activity is often associated with toxicity to human red blood cells [2]. Here, we have trained a combination of

supervised and unsupervised recurrent neural networks (RNN) with hemolysis and activity data from DBAASP

(Database of Antimicrobial Activity and Structure of Peptides) [3] to design short non-hemolytic AMPs. The

synthesis and test of 28 generated peptides led to the identification of eight new non-hemolytic AMPs against 

Pseudomonas aeruginosa, Acinetobacter baumannii, and methicillin-resistant Staphylococcus aureus (MRSA).

This work has been published in Chemical Science. [4]

References

1) Magana, M.; Pushpanathan, M.; Santos, A. L.; Leanse, L.; Fernandez, M.; Ioannidis, A.; Giulianotti, M. A.; Apidianakis,

Y.; Bradfute, S.; Ferguson, A. L.; Cherkasov, A.; Seleem, M. N.; Pinilla, C.; de la Fuente-Nunez, C.; Lazaridis, T.; Dai, T.;

Houghten, R. A.; Hancock, R. E. W.; Tegos, G. P. The Value of Antimicrobial Peptides in the Age of Resistance. Lancet

Infect. Dis. 2020, 20 (9), e216–e230. https://doi.org/10.1016/S1473-3099(20)30327-3.

2) Greco, I.; Molchanova, N.; Holmedal, E.; Jenssen, H.; Hummel, B. D.; Watts, J. L.; Håkansson, J.; Hansen, P. R.;

Svenson, J. Correlation between Hemolytic Activity, Cytotoxicity and Systemic in Vivo Toxicity of Synthetic Antimicrobial

Peptides. Sci. Rep. 2020, 10 (1), 13206.

3) Gogoladze, G.; Grigolava, M.; Vishnepolsky, B.; Chubinidze, M.; Duroux, P.; Lefranc, M.-P.; Pirtskhalava, M. DBAASP:

Database of Antimicrobial Activity and Structure of Peptides. FEMS Microbiol. Lett. 2014, 357 (1), 63–68.

https://doi.org/10.1111/1574-6968.12489.

4) Capecchi, A., Cai, X., Personne, H., Kohler, T., van Delden, C. and Reymond, J.L., 2021. Machine Learning Designs

Non-Hemolytic Antimicrobial Peptides. Chemical Science.

326 | EFMC-ISMC

R005

RIGIDIFIED SHIKIMIC ACID DERIVATIVES TO IMPRINT

INHIBITOR EFFICACY AGAINST SHIKIMATE KINASE ENZYME - A

TARGET FOR ANTIBIOTIC DRUG DISCOVERY

Marina Pernas, Rafael Canabal-Falcón, Beatriz Blanco, Emilio Lence, Adrián Monteiro, Concepción

González-Bello*

Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS), Departamento de QuímicaOrgánica, Universidade de Santiago de Compostela, Jenaro de la Fuente s/n, 15782 Santiago de Compostela, Spain.

*e-mail: [email protected]

The ability of antibiotics to cure bacterial infections is at serious risk due to the emergence and worldwide spread

of superbugs. Convinced that the development of novel chemical entities targeting unexploited targets can

provide solutions to address this long-persisting problem, our research group is exploring the potential of

unexploited enzymes such as shikimate kinase (SK). SK is a recognized target for: (i) Mycobacteriumtuberculosis, which is responsible for tuberculosis; (ii) Helicobacter pylori, which is the causative agent of

gastric and duodenal ulcers and has also been classified as a type I carcinogen; and (iii) Pseudomonasaeruginosa, which is one of the most common pathogens in healthcare-associated infections. SK catalyzes the

stereospecific phosphorylation of the C3 hydroxyl group of shikimic acid by transferring the γ-phosphate group

of ATP to the hydroxyl group to provide shikimate 3-phosphate and ADP. This is achieved by an exquisitely

designed stabilization of the high-energy conformation of shikimic acid. Based on this recognition, we have

applied a very attractive strategy that is used in drug design, i.e. the pre-organization of the ligand conformation

or stabilization of the required high-energy active arrangements through the introduction of conformational

constraints.

1

To this end, diverse rigidified shikimic acids derivatives have been synthesized by ring-closing

metathesis of allyloxy vinyl derivatives as the key step. The rigidification of the ligand conformation was used to

maximize the effectiveness of the substituents introduced in the ether carbon bridge of the scaffold by

pre-orienting their interaction with key residues and enzyme domains that are essential for catalysis and enzyme

motion. Molecular Dynamics simulation studies on the enzyme/ligand complexes revealed marked differences in

the positioning of the ligand substituent in the active site of the two enzymes studied (SK from M. tuberculosisand H. pylori) and this explains their greater efficacy against one of the enzymes. This enhancement is due to the

distinct induced-fit motion of the two homologous enzymes.

Acknowledgements

Financial support from the Spanish Ministry of Science and Innovation (PID2019-105512RB-I00), the Xunta de

Galicia [Centro singular de investigación de Galicia accreditation 2019–2022 (ED431G 2019/03)] and the

European Regional Development Fund is gratefully acknowledged.

References

1) (a) Blanco, B.; Prado, V.; Lence, E.; Otero, J. M.; García-Doval, C.; van Raaij, M. J.; Llamas-Saiz, A. L.; Lamb, H.;

Hawkins, A. R.; González-Bello, C. J. Am. Chem. Soc. 2013, 135, 12366–12376. (b) Pernas, M.; Blanco, B.; Lence, E.;

Thompson, P.; Hawkins, A. R.; González-Bello, C. Org. Chem. Front. 2019, 6, 2514–2528.

EFMC-ISMC | 327

R006

SYNTHESIS OF HEPTOSE MIMETICS AS INHIBITORS OF

BACTERIAL ENZYMES OR MODULATORS OF INFLAMMATION

Jun CAO, Stéphane Vincent

Laboratory of Bioorganic Chemistry, Unamur, 61 rue de Bruxelles, 5000, Belgium

Anti-virulence has recently emerged as an alternative strategy to discover novel antibacterial drug candidates

[1,2].In contrast to traditional antibiotics that kill or impair bacterial viability, this new strategy aims to disarm

the pathogen. Small molecules that can annihilate bacterial virulence are a promising approach to treat infections.

Lipopolysaccharides (LPS) is a pivotal virulence factor of Gram-negative bacteria. LPS can be divided into lipid

A, the oligosaccharide core (core OS), and the oligosaccharide O-antigen (O-PS). Inhibition at the

oligosaccharide core level generates bacteria with a truncated LPS that are much less virulent and more sensitive

towards antibiotics. Thus, the inhibition of bacterial heptose, the first sugar unit that links Kdo

2

-lipid A moiety

and core oligosaccharide, represents an attractive target to discover anti-virulence agents [3].

 Our current project aims to design and synthesize novel antibacterial agents based on targeting bacterial heptose

[fig. 1].

 

                                               Fig.1 Biosynthesis of nucleotide-activated heptoses.

References

1) Seth W. Dickey, Gordon Y. C. Cheung, Michael Otto, Nat. Rev. Drug Discov. 2017, 16, 457-471.

2) A. E. Clatworthy, E. Pierson, D. T. Hung, Nat. Chem. Biol. 2007, 3, 541-548.

3) C. R. Raetz, C. M. Reynolds, M. S. Trent, R. E. Bishop, Annu. Rev. Biochem. 2007,7, 6, 295-329.

328 | EFMC-ISMC

R007

PENICILLIN-BASED SULFONE Β-LACTAMASE INHIBITORS TO

RESTORE CARBAPENEM EFFICACY AGAINST SUPERBUGS

Diana Rodríguez (1), Esther Colchón (1), María Maneiro (1), Juan C. Vázquez-Ucha (2), Alejandro

Beceiro (2), Concepción González-Bello* (1)

1) Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS), Departamento de QuímicaOrgánica, Universidade de Santiago de Compostela, Jenaro de la Fuente s/n, 15782 Santiago de Compostela, Spain.

*E-mail: [email protected]) Servicio de Microbiología do Complexo Hospitalario Universitario da Coruña (CHUAC), Instituto de Investigación

Biomédica da Coruña (INIBIC), Xubias de Arriba, 84, 15006 A Coruña, Spain

Current research efforts to fight against multidrug-resistant bacteria are focused on restoring the efficacy of

antibiotics in clinical use, which have proven to be safe and effective over the years. This successful and

growing area of investigation involves the administration of the antibiotic in combination with an “antibioticadjuvant”, which is a compound that either blocks a certain bacterial resistance mechanism or potentiates the

action of the drug by facilitating its permeabilization into the bacterium. Among them, β-lactamase inhibitors are

the most prominent since are able to disable the most prevalent cause of antibiotic resistance in Gram-negative

bacteria, i.e., the enzymatic deactivation of the most widely used antibiotics, penicillins, cephalosporines,

carbapenems, and monobactams, by β-lactamase enzymes.

1

In recent years, our research group has been

exploring the therapeutic potential of 6-arylmethylidene penicillin-based sulfones as new β-lactamase inhibitors

that could recover antibiotic efficacy. We report here the ability of 6-(aryl)methylidene penicillin-based sulfones

1–7 to restore β-lactam antibiotics activity with bacterial species that carry carbapenem-hydrolyzing class D

β-lactamases (OXA-23, OXA-24/40 and OXA-48), as well as with class A (TEM-1, CTX-M-2) and class C

(CMY-2, DHA-1) enzymes.

2

The most relevant inhibitors of the series were bromine 3 and fluoride 4. Thus, the

combinations imipenem/3 and imipenem/4 restored almost completely the antibiotic efficacy in OXA-23 and

OXA-24/40 carbapenemase-producing A. baumannii strains (1 μg mL

–1

), and also provided good results for

OXA-48 carbapenemase-producing K. pneumoniae strains (4 μg mL

–1

). Inhibition studies, protein mass

spectrometry analysis and docking studies allowed us to gain an insight into the inhibition mechanism and the

experimentally observed differences between the ligands.

Acknowledgements

Financial support from the Spanish Ministry of Science and Innovation (SAF2016-75638-R,

PID2019-105512RB-I00), the Xunta de Galicia [ED431B 2018/04 and Centro singular de investigación de

Galicia accreditation 2019–2022 (ED431G 2019/03)] and the European Regional Development Fund is

gratefully acknowledged.

 

References

1) González-Bello, C.; Rodríguez, D.; Pernas, M.; Rodríguez, A.; Colchón, E. J. Med. Chem. 2020, 63, 1859-1881.

2) Rodríguez, D.; Maneiro, M.; Vázquez-Ucha, J.; Beceiro, A.; González-Bello, C. J. Med. Chem. 2020, 63, 3737-3755.

EFMC-ISMC | 329

R008

AN IN SILICO PIPELINE IDENTIFIES INHIBITORS WITH

CROSS-CLASS ACTIVITY ON CLINICALLY RELEVANT SERINE-

AND METALLO-Β-LACTAMASES

Eleonora Gianquinto (1), Matteo Santucci (2), Lorenzo Maso (3), Simon Cross (4), Filomena Sannio (5),

Federica Verdirosa (5), Filomena De Luca (5), Jean-Denis Docquier (5), Laura Cendron (2), Donatella

Tondi (2), Alberto Venturelli (6), Gabriele Cruciani (7), Maria Paola Costi (8), Francesca Spyrakis (1)

1) Department of Drug Science and Technology, University of Turin, Via Pietro Giuria 9, 10125, Turin, Italy.2) Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 103, 41125, Modena, Italy

3) Department of Biology, University of Padua, Viale G. Colombo 3, 35121, Padua, Italy4) Molecular Discovery Limited, Centennial Avenue, Unit 501 Centennial Park, Borehamwood, Hertfordshire, WD6 3FG,

UK5) Department of Medical Biotechnology, University of Siena, Viale Bracci 16, 53100, Siena, Italy

6) TYDOCK PHARMA S.R.L., Strada Gherbella 294/b, 41126, Modena, Italy7) Department of Chemistry, Biology and Biotechnology, University of Perugia, Via Elce di Sotto 8, 06123, Perugia, Italy

8) Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 103, 41125, Modena, Italy

Antibiotic resistance is worryingly spreading worldwide, especially among the most dreaded ESKAPE bacteria

[1]. A main resistance mechanism involves the production of beta lactamases (BLs, i.e. enzymes able to degrade

beta lactam antibiotics), which are conventionally divided in four classes (A, B, C, or D) [2]. In particular, class

A, C and D enzymes use a catalytic serine to degrade beta lactam antibiotics, while class B enzymes exploit a

zinc-based catalytic hydrolysis. As the road to new classes of antibiotics is long and increasingly problematic,

the inhibition of BLs constitutes an effective strategy to potentiate or restore the activity of current antibiotics. In

this work, we focused on finding inhibitors against five relevant BLs (class A CTX-M-15 and KPC-2, class B

NDM-1 and VIM-2 MBLs, and the class C AmpC). A first in silico screening of a commercially available

library selected a pool of promising candidates, which were purchased and consequently tested against clinically

relevant strains of bacteria producing BLs. The results highlighted that most effective compounds share electron

donor moieties. In particular, KPC-2 is preferentially inhibited by sulfonamide and tetrazole-based derivatives,

NDM-1 by compounds bearing a thiol, a thiosemicarbazide or thiosemicarbazone moiety, while VIM-2 by

triazole-containing molecules. Moreover, we found few inhibitors able to target more than one class of BLs, thus

exerting a “broad-spectrum” inhibition against both serine-based and zinc-based hydrolysis. In vitro tests

highlighted that compound 40 enhances imipenem activity against an NDM-1 producing E. coli clinical isolate.

Finally, X-ray structures of the two most promising compounds in VIM-2 and NDM-1 gave important insights

about the binding mode of triazole-thiol scaffolds. Being NDM-1 and VIM-2 two worldwide emergent

carbapenemases, the following optimization of these promising compounds could lead to the generation of new

tools to fight antiobiotc resistance. 

References

1) WHO. Antimicrobial Resistance. Global Report on Surveillance (World Health Organization, Geneva, 2014).

2) Ambler, R. P. The structure of beta-lactamases. Philos. Trans. R. Soc. Lond. B. Biol. Sci. 289, 321–331 (1980).

330 | EFMC-ISMC

R009

DISCOVERY AND OPTIMIZATION OF VIRTUAL SCREENING AND

FRAGMENT HITS AS NOVEL BACTERIAL SLIDING CLAMP DNAN

INHIBITORS

Uladzislau Hapko (1), Walid A.M. Elgaher (1), Sari Rasheed (2), Peer Lukat (3), Jennifer Herrmann (2),

Wulf Blankenfeldt (3), Rolf Müller (2), Anna K. H. Hirsch (1)

1) Department of Drug Design and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS)*,Campus Building E8.1, 66123 Saarbrücken, Germany

2) Department of Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS)*, CampusBuilding E8.1, 66123 Saarbrücken, Germany

3) Structure and Function of Proteins, Helmholtz Center for Infection Research (HZI), Inhoffenstrasse 7, 38124 Brunswick,Germany

The emergence of the global antimicrobial resistance problem necessitates the discovery and development of

antibiotics with an unprecedented mode of action. The β subunit of the prokaryotic DNA polymerase III (sliding

clamp, DnaN) can be considered in this respect an emerging antibacterial target [1,2]. This protein is essential

for DNA replication and highly conserved in bacteria (yet different from its eukaryotic counterpart) making it a

promising target for the discovery of efficient, broad-spectrum and selective antibacterials. However, known

DnaN inhibitors so far exhibit either modest or strain-limited antibacterial effects [1,2]. Herein, we report new

small-molecule binders of DnaN, discovered by implementing virtual and biophysical screening strategies. We

established a structure-based virtual screening workflow with a pharmacophore filter using the X-Ray structure

of Mtb DnaN in complex with cyclohexylgriselimycin [1]. Evaluation of hits for DnaN binding and antibacterial

activity resulted in a novel chemotype with a promising antibacterial profile. In the fragment-based approach, we

pursued a modified three-step biophysical screening cascade [3]. It included microscale thermophoresis (MST)

for primary screening, surface plasmon resonance (SPR) and STD NMR for hit validation. The structure–activity

relationship studies and optimization of hits from both approaches led to improved activity (MIC 0.5 μM/l)

against mycobacteria and extended the spectrum to both Gram-positive and Gram-negative bacteria.

* - (HIPS) – Helmholtz Center for Infection Research (HZI)

References

1) Kling, A.; Lukat, P.; Almeida, D. V.; et al., Science 2015, 348, 1106–1112.

2) Yin, Z.; Whittell, L. R.; Wang, Y.; et al., J. Med. Chem. 2015, 58, 4693–4702.

3) Mashalidis, E. H.; Śledź, P.; Lang, S.; Abell, C., Nat. Protoc. 2013, 8, 2309–2324.

EFMC-ISMC | 331

R010

MOLECULAR BASIS OF BICYCLIC BORONATE β-LACTAMASE

INHIBITORS OF ULTRABROAD EFFICACY ─ INSIGHTS FROM

MOLECULAR DYNAMICS SIMULATION STUDIES

Emilio Lence, Concepción González-Bello

Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de QuímicaOrgánica, Universidade de Santiago de Compostela. Jenaro de la Fuente s/n, Santiago de Compostela 15782, Spain. *email:

[email protected]

The use of β‐lactamase inhibitors in combination with β‐lactam antibiotics is an emerging area in drug

discovery.

1

This strategy allows the restoration of the therapeutic efficacy of these antibiotics in clinical use

against MDR bacteria. These pathogens elude the therapeutic action of these antibiotics by expressing

β-lactamase enzymes that catalyse the hydrolysis of their β-lactam ring to give inactive products, which is one of

the most relevant resistance mechanisms in deadly pathogens such as Pseudomonas aeruginosa, Acinetobacterbaumannii and Enterobacteriaceae. From the drug development point of view, the design of an efficient

β-lactamase inhibitor able to block this antibiotic resistance mechanism and restore β-lactam antibiotics efficacy

is challenging. This is due to: (i) the huge structural diversity of these enzymes; (ii) the distinct hydrolytic

capability against different types of substrates; (iii) the variety of enzyme mechanisms of action employed, either

involving covalent catalyzed processes (serine hydrolases) or non-covalent catalysis (zinc-dependent

hydrolases); and (iv) the increasing emergence and spread of bacterial pathogens capable of simultaneously

producing distinct β-lactamases. The recent development of taniborbactam (formerly VNRX-5133) and

QPX7728, which are both bicyclic boronate inhibitors currently under clinical development, represents a huge

step towards the development of ultrabroad-spectrum inhibitors able to inhibit all types of β-lactamases. Using

Molecular Dynamic simulations and other computational tools, we shed light to the molecular basis of the

ultrabroad-spectrum of activity of these boron-based inhibitors.

2

The efficacy of bicyclic boronates

taniborbactam and QPX7728 is compared with the cyclic boronate inhibitor vaborbactam, which is the first

boron-based β-lactamase inhibitor approved by the FDA and presents little utility in the clinical treatment of

infections caused by bacteria that produces classes B and D β-lactamases.

Acknowledgements: Financial support from the Spanish Ministry of Science and Innovation

(PID2019-105512RB-I00), the Xunta de Galicia [ED431B 2018/04 and Centro singular de investigación de

Galicia accreditation 2019–2022 (ED431G 2019/03)], and the European Regional Development Fund is

gratefully acknowledged. The authors also thank the Centro de Supercomputación de Galicia (CESGA) for use

of the Finis Terrae computer.

 

References

1) 1. González-Bello, C.; Rodríguez, D.; Pernas, M.; Rodríguez, A.; Colchón, E. J. Med. Chem. 2020, 63, 1859.

2) 2. Lence, E.; González‐Bello, C. Adv. Therap. 2021, 2000246.

332 | EFMC-ISMC

R011

METHYLHYDROXYLAMMONIUM DERIVATIVES -

LYSINE-TARGETED IRREVERSIBLE INHIBITORS FOR THE

ANTI-VIRULENCE BACTERIAL TARGET TYPE I

DEHYDROQUINASE

María Maneiro (1), Emilio Lence (1), Marta Sanz-Gaitero (2), José M. Otero (1), Mark J. van Raaij (2),

Concepción González-Bello (1)

1) Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS), Departamento de QuímicaOrgánica, Universidade de Santiago deCompostela, Jenaro de la Fuente s/n, 15782 Santiago de Compostela, Spain. E-mail:

[email protected]) Departamento de Estructura de Macromoléculas, Centro Nacional de Biotecnología (CSIC), Campus Cantoblanco, 28049

Madrid, Spain

In recent years, irreversible inhibitors have gained momentum in the drug development pipeline with numerous

clinical candidates under study and more than 50 FDA-approved drugs.[1] Their enormous advantages, as well

as the more detailed knowledge available today on the real risks associated with these types of compounds,

explain its increasing presence in the drug discovery programs.[2] Nonetheless, the design of this type of

compounds is still a challenge, because they must combine selectivity (target affinity) and reactivity (covalent

modification) in a single chemical entity, thus enabling inhibitory potency without triggering off-target effects.

In recent years, a great deal of effort has been devoted to the development of ligands bearing functional groups,

often referred to as latent electrophiles, that become activated towards covalent modification upon binding. Most

of them are meant to modify cysteine residues, whereas the available latent electrophiles for selective lysine

residues modification remains sparse. Herein we report the first example of a methylhydroxylammonium

derivative that causes a specific covalent modification of an active-site and a sterically inaccessible lysine

residue of the type I dehydroquinase enzyme (DHQ1), which is a recognized target for the development of new

anti-virulence agents.[3] DHQ1 is present in pathogenic bacteria such as Salmonella typhiand Staphylococcusaureus. These first-in-class compounds 1–3 proved to cause the irreversible inhibition of the DHQ1 enzyme

from S. typhi and S. aureus by covalently modification of the catalytic Lys170/Lys160 through a stable amine

bond, which was demonstrated by resolution by X-ray crystallography at 1.08–1.25 Å of the corresponding

crystal structures of the enzyme/ligand adducts.[4] A two-dimensional QM/MM umbrella sampling simulation

study of the covalent modification mechanism suggested that the direct displacement by nucleophilic attack of

the catalytic lysine residue with the release of hydroxylammonium would be plausible. These studies might open

up new opportunities for the development of novel lysine-targeted irreversible inhibitors bearing a

methylhydroxylammonium moiety as a latent electrophile.

Acknowledgments: Financial support from the Spanish Ministry of Science and Innovation (SAF2016-75638-R,

PID2019-105512RB-I00), the Xunta de Galicia [ED431B 2018/04 and Centro singular de investigación de

Galicia accreditation 2019–2022 (ED431G 2019/03)] and the European Regional Development Fund is

gratefully acknowledged. MM acknowledges the Xunta de Galicia for her postdoctoral fellowship.

References

1) (a) Singh, J.; Petter, R. C.; Baillie, T. A.; Whitty, A. Nat. Rev. Drug Discov. 2011, 10, 307–317. (b) De Vita, E. Future

Med. Chem. 2021, 13, 193–210.

2) González-Bello, C. ChemMedChem 2016, 11, 22-30.

3) González-Bello, C. Future Med. Chem. 2015, 7, 2371-2383.

4) Maneiro, M.; Lence, E.; Sanz-Gaitero, M.; Otero, J. M.; Van Raaij, M. J.; Thompson, P.; Hawkins, A. R.; González-Bello,

C. Org. Chem. Front. 2019, 6, 3127–3135.

EFMC-ISMC | 333

R012

X-RAY CRYSTAL STRUCTURES OF SHORT MIXED CHIRALITY

A-HELICAL ANTIMICROBIAL PEPTIDES

Hippolyte Personne (1), Stéphane Baeriswyl (1), Ivan Di Bonaventura (1), Thilo Khöler (2), Christian van

Delden (2), Achim Stocker (1), Sacha Javor (1), Jean-Louis Reymond (1)

1) Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, CH-3012 Bern,Switzerland

2) Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland

Peptide α-helicity mostly depends on its amino acid sequence and is right or left-handed depending on amino

acids chirality (respectively L- or D-). However, mixed-chirality sequences are usually unfolded. In case of

antimicrobial peptides (AMPs), an amphiphilic α-helix is generally required to be active and research on mixed

chirality AMPs is poorly documented. We recently reported the first X-ray crystal structures of mixed chirality

short bicyclic and linear AMPs as complexes of fucosylated analogs with the bacterial lectin LecB.

1

Our data

suggest that this secondary structure is conserved both in a membrane environment and as helix bundle

determined by X-ray crystallography in water. Furthermore, mixed chirality linear peptide displays lower

hemolytic activity and better stability in human serum while keeping its antimicrobial behavior compare to its

homochiral parent.

References

1) Stéphane Baeriswyl, Hippolyte Personne, Ivan Di Bonaventura, Thilo Köhler, Christian van Delden, Achim Stocker,

Sacha Javor, Jean-Louis Reymond, ChemRxiv, Preprint, 2021, DOI:10.26434/chemrxiv.14052293.v3 

334 | EFMC-ISMC

R013

DISABLING PSEUDOMONAS AERUGINOSA VIRULENCE BY

MODULATION OF THE MASTER REGULATOR OF QUORUM

SENSING LasR

Ángela Rodríguez, Concepción González-Bello*

Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de QuímicaOrgánica, Universidade de Santiago de Compostela, Jenaro de la Fuente s/n, 15782 Santiago de Compostela, Spain.

e-mail: *[email protected]

In recent years, a great deal of effort has been devoted to the development of alternative therapies that target the

bacterial pathogenicity.

The attenuation of bacterial virulence makes the bacterium less able to establish

infection and, in consequence, it is cleared by the host immune system. Anti-virulence drugs will therefore

disarm bacteria and will create an in vivo scenario similar to that achieved by vaccination with a live attenuated

strain. The bacterial infection mechanism is based on a sophisticated cell-cell communication system known as

Quorum Sensing (QS) that relies on the activation of receptor proteins by small diffusible molecules, and which

results in the regulation of important cellular processes including virulence, antibiotic resistance and biofilm

formation. This phenomenon is particular relevant in Pseudomonas aeruginosa, which is one of the three most

critical, common and highly antibiotic-resistant nosocomial pathogens in the world. The LasR receptor is known

to initiate the QS regulatory system in this bacterial species,and therefore, it has recently garnered significant

interest as a target for QS modulationbyantagonists of QS (namely Quorum Quenching, QQ) as treatments of a

variety of human infections caused by this pathogen.

2

It is believed that the binding of the cognate

ligand, C12-AHL,to the ligand binding domain triggers a conformational change in the polypeptide, presumably

enabling the binding of the protein to specific promoters leading to transcriptional activation of QS genes. Based

on the latter, inthis project the development of analogues of the natural autoinducer C12-AHL that will be able to

deactivate the QS gene expression in P. aeruginosaby the formation of complexes LasR/ligand of inappropriate

conformation for binding with specific promoters is proposed. Our recent results in this project will be presented.

Fig.1. Schematic representation of the QS and QQ processes. LB = ligand binding domain. DB = DNA binding

domain.

Acknowledgements

Financial support from the Spanish Ministry of Science and Innovation (SAF2016-75638-R,

PID2019-105512RB-I00), the Xunta de Galicia [ED431B 2018/04 and Centro singular de investigación de

Galicia accreditation 2019–2022 (ED431G 2019/03)] and the European Regional Development Fund is

gratefully acknowledged. AR acknowledges the Spanish Ministry of Science, Innovation and Universities for her

FPI fellowship (BES-2017-080946).

References

1) A. E. Clatworthy, E. Pierson, D. T. Hung, Nat. Chem. Biol. 2007, 3, 541−548.

2) R. C. Allen, R. Popat, S. P. Diggle, S. P. Brown, Nat. Rev. Microbiol. 2014, 12, 300−308.

EFMC-ISMC | 335

R014

IN SEARCH OF NOVEL BACTERIAL RNA POLYMERASE

INHIBITORS

Klara Kuret (1,2,3), Georgi Belogurov (4), Marko Jukić (5,6), Outi Salo-Ahen (1)

1) Åbo Akademi University, Pharmaceutical Sciences Laboratory & Structural Bioinformatics Laboratory, Faculty ofScience and Engineering, Turku, Finland

2) University of Ljubljana, Faculty of Pharmacy, Ljubljana, Slovenia3) Present address: Dept. of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia

4) University of Turku, Department of Life Technologies, Biochemistry, Turku, Finland5) University of Maribor, Faculty of Chemistry and Chemical Engineering, Maribor, Slovenia

6) University of Primorska, Faculty of Mathematics, Natural Sciences and Information Technologies, Koper, Slovenia

The continuously increasing antimicrobial resistance poses a serious a threat to human health and creates thus an

urgent need for the discovery and development of novel antibiotics. Bacterial RNA polymerase (RNAP) is an

ideal broad-spectrum antimicrobial target that is present in both Gram-negative and Gram-positive bacteria and

is sufficiently different from its human counterpart [1]. Currently, there are just two classes of RNAP inhibitors

that are used as antibacterial drugs in the clinic: (i) rifamycins (e.g. rifampicin) that bind adjacent to the

enzyme’s active center, and (ii) lipiarmycins (fidaxomicin) that bind to an allosteric site distant from the active

center [2]. So far, only one nucleoside-analog inhibitor (NAI), pseudouridimycin has been reported to function

by binding to the enzyme’s active site [2,3].

In this work, we present the development of a virtual screening protocol for the discovery of novel bacterial

RNAP inhibitors (non-NAIs) targeting the enzyme’s active site. Molecular docking with GOLD and GLIDE

programs was utilized in the structure-based screening process. Rescoring of the top-ranked binding poses was

performed by estimating the free energy of binding of the docked compounds. Seven virtual hit compounds were

tested for their inhibitory activity in vitro, using the recombinant transcription-elongation complex of the E. coliwild-type RNAP. One of the tested compounds showed relatively high inhibitory activity against the enzyme

while two other hits exhibited only modest RNAP inhibition.

References

[1]) Mosaei, H., & Harbottle, J. (2019). Mechanisms of antibiotics inhibiting bacterial RNA polymerase. Biochemical

Society Transactions, BST20180499. doi:10.1042/bst20180499

[2]) Maffioli, S.I., Zhang, Y., Degen, D. et al. Antibacterial Nucleoside-Analog Inhibitor of Bacterial RNA Polymerase. Cell.

2017 Jun 15;169(7):1240-1248.e23. doi: 10.1016/j.cell.2017.05.042

[3]) Maffioli, S.I., Sosio, M., Ebright, R.H. et al. Discovery, properties, and biosynthesis of pseudouridimycin, an

antibacterial nucleoside-analog inhibitor of bacterial RNA polymerase. J Ind Microbiol Biotechnol 46, 335–343 (2019).

https://doi.org/10.1007/s10295-018-2109-2

336 | EFMC-ISMC

R015

THIOXANTHONES AS ADJUVANTS TO REVERT ANTIMICROBIAL

RESISTANCE

Fernando Durães (1,2), Andreia Palmeira (1,2), Joana Freitas-Silva (2), Nikoletta Szemerédi (3), Gabriella

Spengler (3), Paulo Costa (2,4), Eugénia Pinto (2,5), Madalena Pinto (1,2), Emília Sousa (1,2)

1) LQOF - Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade deFarmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal

2) CIIMAR - Centro Interdisciplinar de Investigação Marinha e Ambiental, Terminal de Cruzeiros do Porto de Leixões, Av.General Norton de Matos S/N, 4450-208 Matosinhos, Portugal

3) Department of Medical Microbiology and Immunobiology, Faculty of Medicine, University of Szeged4) ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228,

4050-313 Porto, Portugal5) Laboratório de Microbiologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto,

Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal

Thioxanthones are S-heterocycles closely related to xanthones, being likewise a privileged structure [1]. Old

drugs include antischistosomal and antitumor lucanthone and new agents with dual antitumor and P-glycoprotein

inhibitory activities with potential to overcome drug resistance have been reported by some of us [1-3]. This

initial study led us to investigate thioxanthones as antibacterial adjuvants more deeply and synergistic activity

with antimicrobials, in resistant strains was discovered [3], pointing out their potential as “antibiotic helpers”, a

very interesting therapeutic strategy [4]. We hypothesized thioxanthones to be bacterial efflux pump inhibitors

with potential to fight against antimicrobial resistance, since the overexpression efflux pumps is one of the major

pathways of antimicrobial resistance [5].

Herein, results showing the potential of aminated thioxanthones as bacterial efflux pump inhibitors (EPI) are

described. In this scope, a virtual library of 1000 aminated thioxanthones was designed and docked into the three

components of the AcrAB-TolC efflux pump, one of the major efflux systems of Gram-negative efflux pumps,

and on a homology model of NorA, an important efflux pump pertaining to Gram-positive bacteria [5]. The best

hits with feasible synthesis were obtained and tested for their antibacterial and antifungal activity and synergy

with antimicrobials. Two aminated thioxanthones emerged as potential adjuvants, as they could decrease the

minimum inhibitory concentration of cefotaxime and oxacillin in resistant strains of Escherichia coli and

Staphylococcus aureus, respectively. This library of compounds was also tested for specific mechanisms of

resistance and virulence, such as EPI activity, inhibition of biofilm formation, and quorum sensing. The assay

used to study the inhibition of the efflux pumps was real-time accumulation of ethidium bromide. The results

obtained suggest that the thioxanthones tested can have potential in the circumvention of antibacterial resistance,

with seven compounds displaying EPI activity, six inhibiting the formation of biofilm and eight being effective

in inhibiting quorum sensing.

 

Acknowledgements: This research was partially supported by the Strategic Funding UIDB/04423/2020,

UIDP/04423/2020 (Group of Natural Products and Medicinal Chemistry) and under the project

PTDC/SAU-PUB/28736/2017 (reference POCI-01-0145-FEDER-028736), co-financed by COMPETE 2020,

Portugal 2020 and the European Union through the ERDF and by FCT through national funds. Also funded by

R&D&I ATLANTIDA - Platform for the monitoring of the North Atlantic Ocean and tools for the sustainable

exploitation of the marine resources (reference NORTE-01-0145-FEDER-000040), NORTE2020, ERDF.

Fernando Durães acknowledges his FCT grant (SFRH/BD/144681/2019).

References

1) Paiva, A.M.; Pinto, M.M.; Sousa, E. Curr Med Chem 2013, 20, 2438-2457.

2) Palmeira, A.; Vasconcelos, M.H.; Paiva, A.; Fernandes, M.X.; Pinto, M.; Sousa, E. Biochem Pharmacol 2012, 83, 57-68.

3) Bessa, L.J.; Palmeira, A.; Gomes, A.S.; Vasconcelos, V.; Sousa, E.; Pinto, M.; Martins da Costa, P. Microb Drug Resist

2015, 21, 404-415.

4) Bueno, J. J Microb Biochem Technol 2016, 8, 169-176, doi:10.4172/1948-5948.1000281.

5) Durães, F.; Pinto, M.; Sousa, E. Curr Med Chem 2018, 25, 6030-6069.

EFMC-ISMC | 337

R016

EQATA: EQUITABLE ACCESS TO QUALITY ANTIBIOTIC

THERAPIES IN AFRICA

Marija K. Zacharova, Yunpeng Wang, Viktorija Stikonaite, Rebecca J. M. Goss, Gordon J. Florence

School of Chemistry, University of St Andrews, St Andrews, United Kingdom

Antibiotic treatable infections are still a global health and mortality burden, causing 5.7 million deaths annually,

the majority of which occur in low- and middle-income countries (LMICS). In Africa, mortality rate from lower

respiratory infections and diarrhoeal diseases outnumbers that from HIV/AIDS, TB and malaria combined,

1,2

and

rising antimicrobial resistance will further increase the mortality rates associated with bacterial diseases. CCDEP

identified a number of barriers to treating infectious diseases LMICS face which include low antibiotic

affordability and unreliable supply chains, which in combination with poor quality control, counterfeit

pharmaceuticals entering the market, and poor stewardship lead to widespread misuse and overuse of antibiotics,

further compounding the emergence of antimicrobial resistance.

1

To tackle these challenges, two strategies have been proposed:

Sustainable and supply chain-safeguarded fermentative production of Access antibiotics

A robust yet simple and cost-efficient fermentation rig constructed from an adapted commercially available

home brewing system will be developed for the fermentative production of precursors to two Access antibiotics

3

 - amikacin and doxycycline, followed by optimization of their semi-synthesis to be cost-efficient and suitable

for semi-industrial scale.

Drug discovery capacity building via community-driven discovery of novel bioactive molecules

In collaboration with our partners with Kenya, Tanzania and Nigeria, a citizen science-driven microbial

discovery project will be launched. Run in collaboration with Tiny Earth,

4

the project will involve

community-driven sampling soil and water and screening for presence of microorganisms producing novel,

promising bioactive molecules.

References

1) Access Barriers to Antibiotics, CDDEP, April 2019

2) WHO report on surveillance of antibiotic consumption: 2016-2018 early implementation, WHO, 2018

3) The State of the World's Antibiotics 2021: A Global Analysis of Antimicrobial Resistance and Its Drivers, CDDEP, 2021

4) https://tinyearth.wisc.edu/

338 | EFMC-ISMC

NOTES

EFMC-ISMC | 339

Posters - Drug Discovery ProjectsT. Recent Advances in Anticancer Drug Discovery

340 | EFMC-ISMC

T001

STRUCTURE-BASED DESIGN AND SYNTHESIS OF SELECTIVE

CLASS I HDAC INHIBITORS AS POTENTIAL ANTICANCER AGENTS

Mohamed Abdelsalam (1), Hany Ibrahim (1), Matthes Zessin (2), Yanira Zeyn (3), Emre Bülbül (1), Anita

Vecchio (1), Frank Erdmann (1), Matthias Schmidt (1), Mike Schutkowski (2), Oliver H. Krämer (3),

Wolfgang Sippl (1)

1) Institute of Pharmacy, Martin-Luther-University of Halle-Wittenberg, 06120 Halle, Germany2) Institute of Biochemistry, Martin-Luther-University of Halle-Wittenberg, 06120 Halle, Germany

3) Institute of Toxicology, Johannes-Gutenberg University of Mainz, 55131 Mainz, Germany

Class I Histone deacetylases (HDACs) play a significant role in regulating gene expression and cellular

proliferation; therefore they are considered as a clinically relevant epigenetic drug targets for treatment of

different types of cancers [1,2].  Most of the currently available HDAC inhibitors are pan-inhibitors with limited

specificity for different HDAC isoforms which might result in undesirable side effects [3,4]. In the present study

we carried out a structure-based optimization of HDAC1,2,3 inhibitors based on the scaffold of the potent

HDACi Quisinostat [5] by replacing the hydroxamic acid functionality with 2-anilinobenzamide as zinc binding

group (ZBG). The newly synthesized compounds were designed to have various substitution patterns in ZBG to

enhance the isoform selectivity. In addition, some modifications in the linker and capping groups were

performed to provide a complete structural activity relationship study. General structure of the synthesized

compounds is shown in the figure below.

                                                        Figure 1. Design of selective HDAC1,2,3 inhibitors

The new compounds were tested in vitro against HDAC1,2,3 and 8. Most of the compounds exhibit good HDAC

1,2,3 inhibitory activity in the low micro molar range. Interestingly, two compounds showed potent inhibitory

activity against HDAC1 in the low nano molar range and showed promising activity in several leukemia cell

lines.

References

1) West AC & Johnstone RW. The Journal of clinical investigation. 2014, 124(1):30-39.

2) Millard CJ. et al. Trends in Pharmacological Sciences. 2017, 38(4):363-377.

3) Yang F. et al. Rsc Advances. 2019, 9(34):19571-19583.

4) D. Liao. Drug Discovery Today: Technol., 2015, 18, 24–28.

5) Arts J, et al. Clinical Cancer Research. 2009, 15(22):6841-6851.

EFMC-ISMC | 341

T002

EVALUATION OF [1,2]OXAZOLE DERIVATIVES IN LYMPHOMA

MODELS

Marilia Barreca (1,2), Virginia Spanò (1), Maria Valeria Raimondi (1), Alessandra Montalbano (1), Rouli

Bai (3), Eugenio Gaudio (2), Stefano Alcaro (4), Ernest Hamel (3), Francesco Bertoni (2), Paola Barraja

(1)

1) Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo,Italy

2) Institute of Oncology Research (IOR), USI University, Bellinzona, Switzerland3) Screening Technologies Branch, Developmental Therapeutics Program, Frederick National Laboratory for Cancer

Research, National Cancer Institute, Maryland, United States4) Department of Health Sciences, University Magna Græcia of Catanzaro, Italy

Non-Hodgkin lymphoma (NHL) is one of the most common haematological malignancy in the world, with more

than 30 distinct subtypes divided into aggressive and indolent. Anti-tubulin agents are widely used in the

treatment of lymphoma both alone and in combination chemotherapy regimens such as ABVD and R-CHOP.

1

The isoxazole moiety is a valuable chemical feature for the design of new tubulin-binding agents, hence we

synthesized a class of new [1,2]oxazole derivatives for the treatment of different NHL subtypes.

All compounds were screened in the NCI’s panel of 60 human cancer cell lines and some of them showed potent

activity with GI

50

values reaching the nanomolar level.

2,3

They were further tested in four lymphoma histotypes:

germinal center B-cell and activated diffuse large B cell lymphoma (GCB-DLBCL and ABC-DLBCL), marginal

zone lymphoma (MZL) and mantle cell lymphoma (MCL). Cell proliferation was measured with the MTT test

after 72 h treatment. Compounds were pre-screened at the dose of 1 μM in SU-DHL-10, HBL1, VL51 and

MINO cell lines. Those with percentage of proliferating cells down to 60% proceeded to screenings with a wider

range of concentrations (0.15–10 µM). Several derivatives showed anti-proliferative activity with IC

50

values

between the low micromolar and the nanomolar range. The most potent derivatives, SIX2-G and SIX13-U,

reached nanomolar activity in the majority of cell lines. Studies on the mechanism of action revealed strong

inhibition of tubulin assembly and colchicine binding.

Structure-activity relationships (SAR) suggested that N-methoxybenzyl substitution at the pyrrole nitrogen plays

an important role in the modulation of activity. In particular, methoxy groups in position 3,4 and/or 5 are

relevant. Ring expansion in the tricyclic system did not affect the activity. However, the presence of a

cyclohexyl ring and condensation of the pyrrole moiety in position [5,4-e] lead to very potent compounds which

deserve further evaluation.

In conclusion, [1,2]oxazole derivatives are very promising anti-proliferative agents with activity in different

lymphoma histotypes, among which MINO cell line (MCL) is the most sensitive. Results will be discussed.

References

1) Barreca, M., et al.; Pharmacology & Therapeutics, 2020, 211, 107552;

2) Spanò, V., et al.; Eur. J. Med. Chem. 2016, 124, 840–851;

3) Spanò, V., et al.; J. Med. Chem. 2020, 63, 20, 12023-12042.

342 | EFMC-ISMC

T003

DESIGN AND SYNTHESIS OF 3,5-SUBSTITUTED

1,2,4-OXADIAZOLES AS CATALYTIC INHIBITORS OF HUMAN DNA

TOPOISOMERASE IIΑ

Kaja Bergant Loboda (1,2), Katja Valjavec (1), Martina Štampar (3), Gerhard Wolber (4), Bojana Žegura

(3), Metka Filipič (3), Marija Sollner Dolenc (2), Andrej Perdih (1,2)

1) National Institute of Chemistry, Hajdrihova 19, SI 1000 Ljubljana, Slovenia2) University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, SI 1000 Ljubljana, Slovenia

3) National Institute of Biology, Department of Genetic Toxicology and Cancer Biology, Večna pot 111, 1000 Ljubljana,Slovenia

4) Institute of Pharmacy, Freie Universität Berlin, Königin Luise-Strasse 2+4, 14195 Berlin, Germany

DNA topoisomerases are enzymes that catalyse topological changes of the DNA molecule and act as important

validated anticancer targets in chemotherapy [1]. Clinically used inhibitors of type II topoisomerases, topo II

poisons, such as etoposide and doxorubicin, although efficient, suffer from severe side effects such as

cardiotoxicity and induction of secondary malignancies. This occurs mainly because they induce the formation

of DNA double strands breaks (DSB) [2]. In our research we are investigating a new catalytic inhibition

mechanism of human topo IIα that could prevent the occurrence of severe side effects associated with topo II

poisons [3].

We designed, synthesized, and evaluated compounds from the class of 3,5-substituted 1,2,4-oxadiazoles that act

as catalytic inhibitors of topo IIα. By rigidization of initial oxadiazole derivatives we aimed to enhance their

interactions with the targeted topo IIα ATP binding site. Obtained compounds inhibited topo IIα as catalytic

inhibitors and displayed binding to the isolated ATPase domain. Proposed binding mode of a new inhibitor was

evaluated by classical molecular dynamics simulations and derived dynophore models served to evaluate the

components of molecular recognition. Selected compounds were cytotoxic on the MCF-7 breast cancer cell line

and did not induce DSB, indicating a different mechanism of cytotoxicity compared to topo II poisons also at the

cellular level. 3,5-substituted 1,2,4-oxadiazoles appear to be promising compounds for further anticancer

development [4].

References

1) Y. Pommier, Y. Sun, S.N. Huang, J.L. Nitiss, Roles of eukaryotic topoisomerases in transcription, replication and genomic

stability, Nat. Rev. Mol. Cell Biol., 17 (2016) 703-721.

2) B. Pogorelčnik, A. Perdih, T. Solmajer, Recent developments of DNA poisons - human DNA topoisomerase IIα inhibitors

- as anticancer agents, Curr. Pharm. Des., 19 (2013) 2474-2488.

3) K. Bergant, M. Janežič, A. Perdih, Bioassays and in silico methods in the identification of human DNA topoisomerase IIα

inhibitors, Curr. Med. Chem., 25 (2018) 3286-3318.

4) K.B. Loboda, K. Valjavec, M. Stampar, G. Wolber, B. Zegura, M. Filipic, M.S. Dolenc, A. Perdih, Design and synthesis

of 3,5-substituted 1,2,4-oxadiazoles as catalytic inhibitors of human DNA topoisomerase IIalpha, Bioorg. Chem., 99 (2020)

103828.

EFMC-ISMC | 343

T004

PRECISION DRUGS: A COVALENT STRATEGY TO MINIMIZE SIDE

EFFECTS OF PI3K INHIBITOR CANCER THERAPY

Chiara Borsari (1), Erhan Keles (1), Jacob McPhail (2), Alexander Schäfer (3), Rohitha Sriramaratnam

(1), Matthias Gstaiger (3), John Burke (2), Matthias Wymann (1)

1) Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland2) Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia V8W 2Y2, Canada

3) Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Otto-Stern-Weg 3, 8093 Zürich, Switzerland

Inhibitors of the phosphatidylinositol 3-kinase (PI3K) – protein kinase B (PKB/Akt) - mechanistic target of

rapamycin (mTOR) axis are considered as valuable assets in cancer therapy. A considerable effort has been

dedicated to the development of drugs targeting the PI3K-mTOR axis

[1-4]

, and some of them are currently

evaluated in preclinical and clinical studies.

Herein we present a strategy to convert a phase II clinical candidate, a pan-PI3K inhibitor (PQR309, bimiralisib)

[1,5]

, into highly selective PI3Kα-covalent inhibitors aiming to minimize the on-target metabolic side effects of

PI3K inhibitor cancer therapy. We exploited a rational approach to increase target selectivity by covalently

targeting a PI3Kα non-conserved nucleophilic amino acid side chain, namely Cys862. A reactive moiety, so

called warhead, was introduced into a chemically modified bimiralisib.

A combination of warhead activity design, proximity and orientation allows a tight control of reversible inhibitor

binding and isoform selective covalent binding. To avoid off-target reactions, we have set up a method to

quantitatively evaluate warheads’ reactivity and optimize for selective Cys862 modification. An extensive

Structure Activity Relationship (SAR) study was performed and a wide range of linear and restricted rotation

linkers introduced. A comprehensive understanding of the kinetics of irreversible inhibition allowed to interpret

SAR and identify compounds with optimal k

inact

(maximum potential rate of inactivation). X-ray crystallography

and mass spectrometry experiments validated the covalent modification of Cys862. Our pilot compounds exceed

specificity and potency over an experimental dimethyl-substituted enone, CNX-1351

[6]

. Moreover, they are

metabolically stable in rat liver microsomes and outperform the rapidly metabolized CNX-1351.

Our strategy to investigate and tune warheads’ reactivity represents a major step forward in the rational design of

covalent chemical tools, overcoming the serendipity in the discovery of irreversible compounds. Moreover, we

provide highly selective chemical tools to dissect PI3K isoform signaling in physiology and disease. A

clarification of the role of the different PI3K isoforms in insulin signaling allows to address the challenges in

isoform selectivity and to develop PI3K inhibitors showing ideal isoform specificity.

References

1) Beaufils F et. al. J Med Chem. 2017, 60 (17), 7524-7538.

2) Rageot R et. al. J Med Chem. 2019, 62 (13), 6241-6261.

3) Borsari C et. al. ACS Med Chem Lett. 2019, 10 (10), 1473-1479.

4) Borsari C et. al. J Med Chem. 2020, 63 (22), 13595-13617.

5) Wicki A et. al. Eur J Cancer. 2018, 96, 6-16.

6) Nacht M et. al. J Med Chem. 2013, 56 (3), 712-721.

344 | EFMC-ISMC

T005

INHIBITION OF THE ACTIVITY OF THE TRANSMEMBRANE FORM

OF CHLORIDE INTRACELLULAR CHANNEL 1 (tmCLIC1): A NOVEL

PHARMACOLOGICAL TOOL IN MULTIPLE TARGETED THERAPY

FOR GLIOBLASTOMA TREATMENT

Donatella Boschi (1), Agnese Chiara Pippione (1), Noemi Villella (1), Marco Lucio Lolli (1), Rosa Maria

Vitale (2), Michele Mazzanti (3), Alessandra Pattarozzi (4), Federica Barbieri (4), Tullio Florio (4)

1) Department of Science and Drug Technology, University of Turin, Via P. Giuria, 9, 10126 Torino, Italy2) Institute of Biomolecular Chemistry (ICB)-CNR, Via Campi Flegrei 34, 80078 Pozzuoli, Italy

3) Department of Bioscience, University of Milano, Via Celoria, 26, 20133, Milan, Italy4) Department of Internal Medicine, University of Genova, viale Benedetto XV, n°2, 16132, Genova, Italy

Glioblastoma (GB) prognosis has not improved in the last 20 years and current standard of care allows a life

expectancy of about 2 years. Therapeutic failure is mainly due to the presence of drug resistant GB stem cells

(GSCs). Here we developed novel pharmacological tool to target GSCs to be used in combined drug therapy.

Our study starts from the observation that the antidiabetic drug metformin exerts antitumor activity on GSCs

inhibiting the activity of the transmembrane form of Chloride Intracellular Channel 1 (tmCLIC1, Fig. (2)),

without significant systemic toxicity.

1

However, its clinical use in oncology is limited by low potency (IC50 in

mM range) and blood-brain barrier (BBB) permeability. In particular, due to high basicity that limits the

unprotonated fraction at physiological pH (pKa 2.8 and 11.5) as well as the poor lipid solubility of the unionized

metformin species (logP -1.43), metformin (chemiotypes A in Fig. (1) is characterized by poor membrane

diffusion, oral absorption and tissue distribution. From the structural point of view several studies have

confirmed that biguanides should be represented as the tautomer B reported in Fig. (1) where an intramolecular

H-bond forms a bridge connecting two nitrogen atoms that can be mimicked by the cyclic biguanidine C and by

the bioisosteric aminopyrazoles D and E that have a pKa significantly lower (9-10 value) as well as the

percentage of the cationic form at physiologic pH. All the chemotypes (linear A and B or cyclic C, D and E) are

able to form several polar and ionic contacts, thus maximizing the interaction with the receptor (Fig. (1)). We

choose them as hits for structural modifications aimed at improving their activity towards the target and

generated the corresponding four derivative series that retained CLIC1 blocker activity but showed significant

higher potency (EC50 in the microM range, Fig. (3)). Specificity has been verified using Clic1-/- GSCs (Fig.

(4)), or cells proliferating independently from tmCLIC1 activity. Although still not good enough for the

translation in a clinical setting, these preliminary results paved the way toward the development of novel

potential drugs with high potency and specificity for GSCs.

References

1) Barbieri F, Wurth R, Pattarozzi A, Verduci I, Mazzola C, Cattaneo MG, et al. Inhibition of Chloride Intracellular Channel

1 (CLIC1) as Biguanide Class-Effect to Impair Human Glioblastoma Stem Cell Viability. Frontiers in pharmacology.

2018;9:899.

EFMC-ISMC | 345

T006

DE NOVO DESIGN OF FOLDAMERIC MINI-PROTEINS AND THEIR

USE AS PD-1/PD-L1 INTERACTION INHIBITORS

Agnieszka Ciesiołkiewicz, Katarzyna Ożga, Łukasz Berlicki

Department of Bioorganic Chemistry, Wrocław University of Science and Technology, Wybrzeże Wyspiańskiego 27, 50-370Wrocław

Immunotherapies are currently a very promising approach in cancer treatment. Therefore, the blocking of

immune checkpoints has been extensively studied in recent years.

1

One of them, PD-1/PD-L1 immune

checkpoint is a favorable but difficult target for inhibition, because of the large, flat, hydrophobic interaction

surfaces of both proteins. However, 6 antibodies, inhibiting PD-1/PD-L1, have been already approved by FDA.

2

Unfortunately, antibody-based therapies show numerous disadvantages such as immunogenicity, poor solid

tumor penetration, and poor control of pharmacokinetics. Our goal is the utilization of peptide-based foldamers

containing β-amino acids for inhibition of PD-1/PD-L1 interaction as an improved way for cancer treatment. The

incorporation of β-amino acids into peptide scaffolds can lead to exhibit numerous beneficial features for the

construction of protein-protein interaction inhibitors such as high resistance to enzymatic proteolysis and low

toxicity.

3

In this work, mini-proteins were designed de novo assuming structure containing two β-strands, α-helix, and two

β-strands. The surface of β-strands was utilized for interaction with target PD-L1 and the foldameric helix was

obtained by introduction of a cyclic β-amino acid - trans-β-aminocyclopentanecarboxylic acid (trans-ACPC).

The proposed methodology is based on molecular modeling strategies and includes three major steps: design of

the scaffold with β-amino acids, iterative remodeling of the scaffold for obtaining improved properties, and

incorporation of activity into the scaffold. Mini-proteins were synthesized using microwave-assisted automated

solid phase synthesis. Their secondary structure and thermal stability were determined by circular dichroism.

Binding kinetic analyses and inhibition measurements were carried out using BLI (biolayer interferometry), and

HTRF (homogeneous time-resolved fluorescence).

References

1) Riley RS, June CH, Langer R, Mitchell MJ. Delivery technologies for cancer immunotherapy. Nat Rev Drug Discov.

2019;18(3):175-196. doi:10.1038/s41573-018-0006-z

2) Vaddepally RK, Kharel P, Pandey R, Garje R, Chandra AB. Review of indications of FDA-approved immune checkpoint

inhibitors per NCCN guidelines with the level of evidence. Cancers (Basel). 2020;12(3):1-19. doi:10.3390/cancers12030738

3) Cabrele C, Martinek TA, Reiser O, Berlicki Ł. Peptides containing β-amino acid patterns: Challenges and successes in

medicinal chemistry. J Med Chem. 2014;57(23):9718-9739. doi:10.1021/jm5010896

346 | EFMC-ISMC

T007

STRUCTURAL INSIGHTS ON THE BINDING MODE OF A SERIES OF

NOVEL DUAL INHIBITORS AGAINST CK2, HDAC1 AND HDAC6

Claire Coderch, Laura Marquez-Cantudo, Irene Ortín , José María Zapico, Ana Ramos, Beatriz de

Pascual-Teresa

Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities,Urbanización Montepríncipe, 28925, Alcorcón, Madrid, Spain.

The design of dual-targeted molecules is a hot topic in modern drug design. Dual inhibitors are more effective

than drugs addressed to only one biological target and reduce the risk of drug-drug interactions compared to the

treatments with drug coctails.

1,2

 In this line, our research group has extended experience on the design and

development of compounds aimed to selectively inhibit proteins that are implicated in cell division: Casein

Kinase 2 (CK2) and Histone DeACetylases (HDACs). CK2 is a constitutively active kinase present in many cell

compartments and tightly implicated in cell growth, differentiation and apoptosis, gene expression and signal

transduction.

3,4

The large HDAC family consists of epigenetic enzymes that regulate DNA packing by

catalyzing the hydrolysis of acetylated lysines in histones, thus they play an important role in transcriptional

regulation, cell cycle progression, and apoptosis.

5, 6

Here we present the design strategy of a series of novel

dual-inhibitors against CK2 and two HDAC family members: HDAC1 and HDAC6.

Acknowledgements: Financial support from RTI2018-093539-B-I00 (MICIU/FEDER, UE) is kindlyacknowledged.

References

1) S. Frantz, Nat. Rev. Drug Discovery, 2006, 5, 881–882.

2) R. Morphy and Z. Rankovic, J. Med. Chem., 2005, 48, 6523– 6543.

3) N. A. St-Denis and D. W. Litchfeld, Cell. Mol. Life Sci., 2009, 66, 1817–1829.

4) K. A. Ahmad, G. Wang, G. Unger, J. Slaton and K. Ahmed, in Advances in Enzyme Regulation, ed. G. Weber, C. E.

ForrestWeber and L. Cocco, 2008, vol. 48, pp. 179–187.

5) M. Haberland, R. L. Montgomery and E. N. Olson, Nat. Rev. Genet., 2009, 10, 32–42.

6) H. Lehrmann, L. L. Pritchard and A. Harel-Bellan, Adv. Cancer Res., 2002, 86, 41–65.

EFMC-ISMC | 347

T008

A NEW LEAD COMPOUND FOR TRIPLE NEGATIVE BREAST

CANCER THERAPY

Marta Costa (1), Olívia Pontes (1), Sofia Oliveira-Pinto (1), Céline S. Gonçalves (1), Sara Duarte-Silva (1),

Belém Sampaio-Marques (1), Carlos Bessa (1), Paula Ludovico (1), Bruno M. Costa (1), Patrícia Maciel

(1), Fernanda Proença (2), Fátima Baltazar (1)

1) Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus of Gualtar, Braga,Portugal; ICVS/3B’s - PT Government Associate Laboratory, Braga/Guimarães, Portugal

2) Department of Chemistry, School of Sciences, University of Minho, Campus of Gualtar, Braga, Portugal

Breast cancer (BC) is the most diagnosed cancer worldwide and the main cause of cancer-related death in

women (1). BC is considered a heterogeneous disease composed by different molecular subtypes, (Luminal A,

Luminal B, HER2-enriched and Triple Negative) that presents diverse prognostic, risk factors and responses to

treatment (2). Triple-negative breast cancer (TNBC) represents 15-20% of all breast cancers, being considered

the most aggressive subtype (3).  TNBC is unresponsive to existing endocrine or HER2-targeted therapy.

Chemotherapy, surgery and radiation are the current standard therapeutic options, but present very limited results

and induce severe side-effects (4,5). Metastasis formation, the low response to existing therapy and the high rate

of relapse (50% of early-stage patients) are major problems in TNBC.  Also, 37% of early-stage and 90% of

late-stage patients experience a 5-year mortality rate. Thus, there is an urgent medical need for new effective and

safe approaches for TNBC therapy.

Chromene-based scaffolds have been explored as medicinal agents and their biological activity has been

extensively studied (6). Also called benzopyrans, chromenes are heterocyclic compounds comprising a benzene

ring fused to a pyran nucleus. The continuous effort in drug discovery revealed numerous chromenes with

significant anticancer properties in many different cancer cell models, with several molecular targets involved in

cancer progression. New chromene-based compounds were synthesized using innovative experimental methods.

The antiproliferative potential of these chromenes and several cancer aggressiveness features were investigated

using different breast cancer cell lines (MCF-7, Hs578t and MDA-MB-468). The synthesized novel compounds

revealed potent anticancer activity and selectivity towards cancer cells, especially for TNBC cells (IC

50

in the

nM range). These molecules interfere with several cancer aggressiveness features in vitro, inducing regulated

cell death, G

2

/M cell cycle arrest, microtubule destabilization and decreasing cell migration and proliferation.

These results permitted the identification of a lead compound, MC408, comprising the best bioactive profile.

Efficacy studies with MC408, using the TNBC cells, in the ex vivo CAM model revealed significant reduction in

tumor perimeter and blood vessel formation. These results were supported by treatment of mice orthotopic

TNBC xenografts, revealing an excellent capacity to inhibit tumour growth, in a dose dependent manner.

Toxicity of MC408 was studied in vivo using the Caenorhabditis elegans and mouse models and no adverse

effects were detected, being indicative of a safe preclinical profile. These new compounds and their use as

anticancer agents are protected by an International Patent (PCT/IB2020/056131), due to their demonstrated

promising anticancer profile.

The present study provides evidence for the effectiveness and safety of a new drug candidate for TNBC therapy,

offering a new hope for BC patients suffering from this aggressive disease. Ultimately, we hope to translate the

results into the clinics, helping to improve the efficacy and safety of existing therapies, leading to the better

survival rates and overall life quality of survivors.

References

1) Sung H, Ferlay J, Siegel R.L, Laversanne M, Soerjomataram I, Jemal A, Bray F, Global Cancer Statistics 2020:

GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin.

2021;0:1–41.

2) Prat A, Pineda E, Adamo B, Galvan P, Fernandez A, Gaba L, et al. Clinical implications of the intrinsic molecular

subtypes of breast cancer. Breast. 2015;24 Suppl 2:S26-35.

3) Lee KL, Kuo YC, Ho YS, Huang YH. Triple-Negative Breast Cancer: Current Understanding and Future Therapeutic

Breakthrough Targeting Cancer Stemness. Cancers (Basel). 2019;11(9).

4) Rakha EA, Chan S. Metastatic triple-negative breast cancer. Clin Oncol (R Coll Radiol). 2011;23(9):587-600.

5) Ajithkumar TV, Hatcher HM. Specialist Training in ONCOLOGY. Elsevier M, 2011. 115-33.

6) Costa M, Dias TA, Brito A, Proenca F. Biological importance of structurally diversified chromenes. Eur J Med Chem.

2016;123:487-507.

348 | EFMC-ISMC

T009

STRUCTURE-ACTIVITY RELATIONSHIPS OF

BENZOTHIAZOLE-BASED HSP90 C-TERMINAL DOMAIN

INHIBITORS

Jaka Dernovšek, Živa Zajec, Tihomir Tomašič

Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000 Ljubljana, Slovenia

Heat shock protein 90 kDa (Hsp90) is a chaperone that consists of C-terminal domain, middle domain, and

N-terminal ATP-binding domain. This chaperone is responsible for the maturation of more than 300 client

proteins. Many of those such as Akt, CDK-4, Raf, p53 and others play crucial roles in the pathophysiology of

various cancers

[1]

. Moreover, Hsp90 is usually overexpressed in cancer cells

[2]

. Therefore, it is not surprising

that Hsp90 has been extensively studied as a potential target for the development of new anticancer agents.

Furthermore, at least 18 Hsp90 ATP-competitive N-terminal domain inhibitors have reached clinical trials.

Unfortunately, all of them have failed due to undesirable induction of heat shock response (HSR), resulting in

treatment inefficiency and some other side effects

[3]

. More recently new ways to circumvent these problems

have been described. One of those is the design of C-terminal allosteric modulators of this chaperone. C-terminal

Hsp90 inhibition leads to the desired downregulation of Hsp90 clients but does not simultaneously activate the

HSR

[2]

.

Therefore, our goal was to design, synthesise, and evaluate Hsp90 C-terminal allosteric modulators. The

compounds were designed by a combination of molecular docking and molecular dynamics simulations.

Although the exact C-terminal binding site is not yet known, a molecular modelling study has indicated that the

distance between cationic centre and aromatic ring is important. This is due to the interactions between Glu489A

and the cationic centre on one side and between the aromatic ring and the hydrophobic pocket on the other side

of the inhibitor

[4]

. It is also known that the benzothiazole ring is a useful structural feature in Hsp90 inhibitors,

but no connection between these two features has yet been established

[5,6]

. Therefore, we have decided to take a

deeper look in the structure-activity relationship of such benzothiazole Hsp90 C-terminal inhibitors. We have

prepared a library of about 30 compounds in which we have introduced different substituents at positions 2 and 6

of the benzothiazole ring as shown in Figure 1. At position 6 we have placed a differently substituted aromatic

ring, while we have introduced changes on position 2 to better elaborate the relevance of the basic centre in the

molecule.

We have evaluated these compounds in cell-based assays, where they have shown low micromolar

antiproliferative IC

50

values. We have shown that the cationic centre is critical for the efficiency of our inhibitors,

while some changes in the distance relative to the aromatic ring are tolerated but affect potency. Varying

substituents on the aromatic ring has also had some effect on potency. Most importantly, we have elucidated the

influence of halogens on binding affinity. These results represent important starting points for further

optimization.

Figure 1. Benzothiazole ring can serve as an appropriate linker between the cationic centre and aromatic ring

(A) as the compounds can reach low micromolar range antiproliferative IC

50

values in MCF-7 breast cancer cell

line (B). 

References

1) S. E. Jackson, in Molecular Chaperones (Ed.: S. Jackson), Springer, Berlin, Heidelberg, 2013, pp. 155–240.

2) K. M. Goode, D. P. Petrov, R. E. Vickman, S. A. Crist, P. E. Pascuzzi, T. L. Ratliff, V. J. Davisson, T. R. Hazbun,

Biochim Biophys Acta Gen Subj 2017, 1861, 1992–2006.

3) H.-K. Park, N. G. Yoon, J.-E. Lee, S. Hu, S. Yoon, S. Y. Kim, J.-H. Hong, D. Nam, Y. C. Chae, J. B. Park, B. H. Kang,

Experimental & Molecular Medicine 2020, 52, 79–91.

4) T. Tomašič, M. Durcik, B. M. Keegan, D. G. Skledar, Ž. Zajec, B. S. J. Blagg, S. D. Bryant, International Journal of

Molecular Sciences 2020, 21, 6898.

5) A. Lamut, M. Gjorgjieva, L. Naesens, S. Liekens, K.-E. Lillsunde, P. Tammela, D. Kikelj, T. Tomašič, Bioorganic

Chemistry 2020, 98, 103733.

6) K. W. Pugh, Z. Zhang, J. Wang, X. Xu, V. Munthali, A. Zuo, B. S. J. Blagg, ACS Med. Chem. Lett. 2020, 11, 1535–1538.

EFMC-ISMC | 349

T010

DEVELOPMENT OF THE NOVEL HIF INHIBITOR LW1564 THAT

TARGETS CANCER METABOLISM

Minkyoung Kim, Dongik Kwak, Sreenivasulu Godesi, Hammouda Hossam, Kyeong Lee

College of Pharmacy, Dongguk University, Goyang 10326, Korea

Hypoxia inducible factor (HIF) 1 is a transcription factor that functions as a mast regulator in the response of

growing tumor to hypoxia. Hypoxia-inducible factor-1 (HIF-1), a heterodimeric transcription factor consisting of

HIF-1α and HIF-1β, functions as a master regulator in response to tumor hypoxia and plays a key role in the

metabolic reprogramming of cancer cells.

Targeting cancer metabolism has emerged as an important cancer therapeutic strategy. The purpose of this study

was to identify a potential cancer drug that inhibits HIF-1 and targets cancer metabolism. We found that

LW1564 significantly inhibited the growth of HepG2 cells by suppressing mitochondrial respiration.

Mechanistically, LW1564 targeted mitochondrial ETC complex I, alleviating tumor hypoxia to promote HIF-1

degradation. In addition, the reduction in ATP level activated AMPK and inhibited mTOR pathway signaling,

which suppressed fatty acid synthesis. Therefore, LW1564 significantly inhibited tumor growth in a HepG2

xenograft mouse model, suggesting that LW1564 be developing novel therapeutics that target cancer

metabolism.

References

1) Experimental & Molecular Medicine (2020) 52:1845–1856

350 | EFMC-ISMC

T011

ANTITUMOR ACTIVITY AND FLUORESCENCE OF TETRACYCLIC

THIOXANTHENES: NEW THERANOSTIC AGENTS?

Fernando Durães (1,2), Patrícia M. A. Silva (3), Pedro Novais (3,4), Cátia I. C. Esteves (5), Samuel Guieu

(5,6), Hassan Bousbaa (3), Madalena Pinto (1,2), Emília Sousa (1,2)

1) Laboratory of Organic and Pharmaceutical Chemistry, Department of Chemical Sciences, Faculty of Pharmacy,University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal

2) CIIMAR-Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Novo Edifício do Terminalde Cruzeiros do Porto de Leixões, Avenida General Norton de Matos, S/N, 4450-208 Matosinhos, Portugal

3) CESPU, Institute of Research and Advanced Training in Health Sciences and Technologies (IINFACTS), Rua Central deGandra, 1317, 4585-116 Gandra, Portugal

4) Department of Biology, Faculty of Sciences, University of Porto, Rua do Campo Alegre, s/n, 4169-007 Porto, Portugal5) LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal

6) CICECO-Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal

Thioxanthones are bioisosteres of xanthones and are likewise privileged structures. Their tricyclic scaffold

constitutes an useful chemical substrate for molecular modification, which can lead to a wide range of new

compounds, with diverse bioactivities [1]. In fact, thioxanthones have been described as antitumor agents,

modulators of P-glycoprotein [2], antibacterial agents and have displayed synergy with antimicrobials in

resistant strains [3]. They have additionally been studied for photoinitiator properties [4].

In this work, we obtained an unexpected tetracyclic scaffold by coupling of thioxanthones, generating a

thioxanthene scaffold, with amines and tested their cell growth inhibitory potential against three cell lines: breast

adenocarcinoma (MCF-7), melanoma (A375-C5), and non-small cell lung cancer (NCI-H460). The results

obtained suggest two tetracyclic thioxanthenes as potent inhibitors of tumor cell growth.

Furthermore, these compounds are based on the D-π-A structure (“push-pull” system), architected by an electron

donor group (C-4 propoxy chain), an electron acceptor group (C-1 guanidine/urea) and bridged by a conjugated

π system, which is common to most near-infrared fluorescence probes. As such, photophysics studies were

performed, in order to understand the emission and absorption spectra of the compounds.

Fluorescence microscopy and flow cytometry analysis on cells treated with these compounds revealed that one

compound emitted a bright cytoplasmic fluorescence in the green channel, when cells were excited with 470 nm

light. Live-cell imaging showed that the fluorescent compound was internalized soon after its addition to cell

medium, and accumulated within intracellular endosome-like structures. Besides its high cytotoxicity, this

fluorescence propriety of the compound highlights their potential as theranostic cancer drug candidates. This can

lead to the application of these tetracyclic thioxanthones as theranostic agents, a concept that has been gathering

particular importance in the cancer field [5].

 

Acknowledgements: This research was partially supported by the Strategic Funding UIDB/04423/2020,

UIDP/04423/2020 (Group of Natural Products and Medicinal Chemistry) and under the project

PTDC/SAU-PUB/28736/2017 (reference POCI-01-0145-FEDER-028736), co-financed by COMPETE 2020,

Portugal 2020 and the European Union through the ERDF and by FCT through national funds. Fernando Durães

acknowledges his FCT grant (SFRH/BD/144681/2019). Hassan Bousbaa acknowledges funding from CESPU

(IMPLDEBRIS-PL-3RL-IINFACTS-2019). This research was funded by Fundação para a Ciência e a

Tecnologia (FCT) through research units UIDB/50006/2020, UID/CTM/50011/2019, UIDB/50011/2020 &

UIDP/50011/2020, and by the European Union (FEDER program) through project

CENTRO-01-0145-FEDER-000003.

References

1) Durães, F.; Pinto, M.; Sousa, E. Curr Med Chem 2018, 25, 6030-6069.

2) Palmeira, A.; Vasconcelos, M.H.; Paiva, A.; Fernandes, M.X.; Pinto, M.; Sousa, E. Biochem Pharmacol 2012, 83, 57-68.

3) Bessa, L.J.; Palmeira, A.; Gomes, A.S.; Vasconcelos, V.; Sousa, E.; Pinto, M.; Martins da Costa, P. Microb Drug Resist

2015, 21, 404-415.

4) Hassan, S.I.; Haque, A.; Jeilani, Y.A.; Ilmi, R.; Faizi, M.S.H.; Khan, I.; Mushtaque, M. J Mol Struct 2021, 1224, 129004.

5) Penet, M.-F.; Chen, Z.; Kakkad, S.; Pomper, M.G.; Bhujwalla, Z.M. Eur J Radiol 2012, 81 Suppl 1, S124-S126.

EFMC-ISMC | 351

T012

DYNOPHORE-BASED APPROACH TO INHIBITOR DESIGN:

DEVELOPMENT OF HUMAN DNA TOPOISOMERASE IIA

CATALYTIC INHIBITORS

Matej Janežič (1,2), Katja Valjavec (1), Kaja Bergant Loboda (1,3), Barbara Herlah (1,3), Gerhard

Wolber (4), Andrej Perdih (1,3)

1) National Institute of Chemistry, Hajdrihova 19, SI 1000 Ljubljana, Slovenia2) Laboratory for Structural Bioinformatics, RIKEN Center for Biosystems Dynamics Research, 1-7-22 Suehiro-cho,

Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan3) University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, SI 1000 Ljubljana, Slovenia

4) Institute of Pharmacy, Freie Universität Berlin, Königin Luise-Strasse 2+4, 14195 Berlin, Germany

DNA topoisomerases represent a diverse family of complex molecular motors that catalyze the topological

changes of the DNA molecule and are established targets of chemotherapy [1]. Clinically used inhibitors of

human type II DNA topoisomerases, topo II poisons, such as etoposide and doxorubicin, possess severe side

effects such as cardiotoxicity and induction of secondary malignancies. These occur mainly because such

compounds induce formation of DNA double strands breaks (DSB). To steer away for these unfavorable

attributes, we are investigating a new alternative catalytic topo IIα inhibition paradigm developing molecules

that would bining to its ATP binding site. [2,3]. 

             

In this study, we utilized a human DNA topo IIα as a model target to outline a dynophore-based approach to

catalytic inhibitor design. Based on MD simulations of a known catalytic inhibitor [4] and a native ATP ligand

we derived a joint dynophore model that supplemented the structure based-pharmacophore information with a

dynamic component. Subsequently derived pharmacophore models were employed in a virtual screening

campaign using a library of natural compounds. Experimental evaluation returned 5 flavonoid compounds,

possessing promising topo IIα catalytic inhibition. Binding studies also suggested the interaction with the

ATPase domain. The proposed binding modes of hit compounds were investigated in extensive MD simulations,

essential dynamics and MM-PBSA free energy calculations to reconnect the obtained results to the initial

dynophore-based screening model. Besides, a new design strategy that incorporates a dynamic component of

molecular recognition, the discovered flavonoids also comprise new hit compounds acting via an alternative

inhibition mechanism on this establish anticancer target.

References

1) J.L. Nitiss, DNA topoisomerase II and its growing repertoire of biological functions. Nat. Rev. Canc., 2009, 9, 327-337.

2) B. Pogorelčnik, A. Perdih, T. Solmajer, Recent developments of DNA poisons - human DNA topoisomerase IIα inhibitors

- as anticancer agents, Curr. Pharm. Des., 2013, 19, 2474-2488.

3) K. Bergant, M. Janežič, A. Perdih, Bioassays and in silico methods in the identification of human DNA topoisomerase IIα

inhibitors, Curr. Med. Chem., 2018, 25, 3286-3318.

4) B. Pogorelčnik,, M. Janežič,, I. Sosič, , S. Gobec,, T. Šolmajer, A. Perdih. 4,6- substituted-1,3,5-triazin-2(1H)-ones as

monocyclic catalytic inhibitors of human DNA topoisomerase IIα targeting the ATP binding site. Bioorg. & Med. Chem.

2015, 23, 4218-4229.

352 | EFMC-ISMC

T013

KEY ASPECTS OF STRUCTURE-BASED DRUG DESIGN

STRATEGIES FROM THE DEVELOPMENT OF A POTENT CLASS OF

SMALL MOLECULE INHIBITORS OF THE MDM2-P53

PROTEIN-PROTEIN INTERACTION

Rhian Holvey

On behalf of Astex Pharmaceuticals, 436 Cambridge Science Park, Milton Road, Cambridge, CB4 0QA, UKand the Cancer Research UK Newcastle Drug Discovery Unit, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK

The p53 tumour suppressor is activated in response to cellular stress to modulate cell cycle progression, DNA

repair, and cell death. The activity of p53 is tightly regulated by MDM2, an E3 ubiquitin ligase that targets p53

for proteasomal degradation. Inhibition of the MDM2-p53 interaction in tumours carrying wild-type p53 can

therefore reactivate p53 and elicit an anti-cancer effect. Small molecule inhibitors of the MDM2-p53 interaction

remains a promising strategy for cancer therapy and a number of these compounds are in clinical development.

Structure-based drug design was successfully applied to a previously reported isoindolinone series to identify

single digit nM MDM2-p53 inhibitors with in vivo activity. A combination of X-ray co-crystal structures, NMR

conformational analysis and computational methods were used to improve the binding affinity against the target

and improve physical chemical properties of our initial hit compound.

We will also present our efforts in identifying the metabolites of this series of compounds and the strategy used

to improve their in vivo PK profile, including deuteration of the metabolically labile soft spot. This resulted in a

lead compound with sub µM cellular activity in the SJSA-1 wild-type p53 cell line and improved stability in

CYP3A4 which showed marked tumour growth delay when dosed to mice bearing human SJSA-1 tumour

xenografts.

EFMC-ISMC | 353

T014

DESIGN OF FIRST-IN-CLASS DUAL EZH2/HDAC INHIBITOR:

BIOCHEMICAL ACTIVITY AND BIOLOGICAL EVALUATION IN

CANCER CELLS

Clemens Zwergel (1,2), Annalisa Romanelli (1), Giulia Stazi (1), Rossella Fioravanti (1), Elisabetta Di

Bello (1), Silvia Pomella (3), Clara Perrone (3), Cecilia Battistelli (4), Raffaele Strippoli (4,5), Marco

Tripodi (4,5), Donatella Del Bufalo (6), Rossella Rota (3), Daniela Trisciuoglio (6,7), Antonello Mai (1),

Sergio Valente (1)

1) Department of Drug Chemistry and Technologies, Sapienza University of Rome, P. le A. Moro 5, 00185 Rome, Italy2) Department of Medicine of Precision, University of Campania Luigi Vanvitelli, Via L. De Crecchio 7, 80138 Naples, Italy

3) Department of Oncohematology, Bambino Gesù Children’s Hospital, IRCCS, Viale San Paolo 15, 00146 Rome, Italy4) Department of Molecular Medicine, Sapienza University of Rome, P. le A. Moro 5, 00185 Rome, Italy

5) National Institute for Infectious Diseases L. Spallanzani, IRCCS, Via Portuense 292, 00149 Rome, Italy6) Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, Viale Regina Elena

291,295, 00100 Rome, Italy7) Institute of Molecular Biology and Pathology, National Research Council (CNR), Via Degli Apuli 4, Rome 00185, Italy

Since the histone modifying enzymes EZH2 and HDACs control a lot of epigenetic-dependent carcinogenic

pathways [1-2], we designed the first-in-class dual EZH2/HDAC inhibitor 5 (Figure 1) displaying

(sub)micromolar inhibition against the both targets. When tested in several cancer cell lines, the hybrid 5

impaired cell viability at low micromolar level, and in leukaemia U937 and rhabdomyosarcoma RH4 cells

provided subG1 arrest, apoptotic induction and increased differentiation, together with modulation of acetyl-H3,

acetyl-α-tubulin, and H3K27me3 levels. In glioblastoma U87 cells, 5 hampered epithelial to mesenchymal

transition by increasing the E-cadherin expression, thus proposing itself as a useful candidate for anticancer

therapy [4].

 

Figure 1: Design strategy and summary of the main biological effects of 5

References

1) Luan, Y. et al. J Med Chem 2019, 62 (7), 3171-3183; 2) Fioravanti, R. et al. Chem Rec 2018, 18 (12), 1818-1832; 3)

Mellini, P. et al. Philos Trans R Soc Lond B Biol Sci 2018, 373 (1748); 4) Romanelli, A. et al. ACS Med Chem Lett 2020

accepted manuscript

354 | EFMC-ISMC

T015

AROMATIC AND HETEROAROMATIC SULFONAMIDES AS

INHIBITORS OF CARBONIC ANHYDRASES - SYNTHESIS AND

BIOLOGICAL EVALUATION

Jekaterina Ivanova (1), Raivis Žalubovskis (1,2)

1) Latvian Institute of Organic Synthesis, 21 Aizkraukles St., LV-1006, Riga, Latvia2) Institute of Technology of Organic Chemistry, Faculty of Materials Science and Applied Chemistry, Riga Technical

University, 3/7 Paula Valdena St., LV-1048, Riga, Latvia

Carbonic anhydrases (CA) are zinc containing enzymes that catalyze CO

2

conversion to bicarbonate anion and

proton (Picture 1). hCAs (human CAs) are involved in a plenty of important physiological processes, such as gas

exchange, transport of CO

2

 and HCO

3

 across membranes, biosynthetic reactions, etc.

1-2

Fifteen different CA isoforms have been identified and characterized in human so far.

3

   CA I and CA II have

been found in a number of organs and tissues. CA IX is a hypoxia-inducible enzyme that regulates cellular pH to

promote cancer cell survival. Both CA IX and CA XII are tumor-associated isoforms. The aim of our research is

to obtain a series of selective CA IX/CA XII inhibitors.

Sulfonamides A, B (Picture 2) and their analogues have shown CA inhibitory activity.

4

The X-Ray structures of

CA IX-A and CA IX-B allowed us to design new potential selective CA IX inhibitors.

A series of A and B derivatives with various aryl and hetaryl fragments instead of triazole moiety (Picture 3,

derivative C) was synthesized via Suzuki-Miyaura coupling. Thiophene ring was replaced with benzene ring as

well (Picture 3, derivatives D, E).

Biological screening of all the synthesized compounds was performed (in vitro enzymatic on target CA IX and

CA XII and off-target CA I and CA II).

 

Acknowledgement: This project „Novel Inhibitors of Metalloenzymes as Anti-Cancer Agents” is supported byEuropean Regional Development Fund (PostDoc Latvia), project number 1.1.1.2/VIAA/3/19/576.

References

1) Alterio, V., et al. Chem. Rev., 2012, 112, 4421.

2) Supuran, C.T. J. Enz. Inhib. Med. Chem. 2013, 28, 229.

3) Nocentini, A., et al. Carbonic Anhydrases, 2019, Chapter 8, 151.

4) Leitans, J., et al. Bioorg. Med. Chem. Lett. 2013, 21, 5130.

EFMC-ISMC | 355

T016

SYNTHESIS AND ANTICANCER EVALUATION OF SPIROCYCLIC

BROMOTYROSINE ANALOGS INSPIRED FROM MARINE

COMPOUND CLAVATADINE C

Piyush Patel (1), Tanja Bruun (1), Polina Ilina (2), Heidi Mäkkylä (2), Antti Lempinen (1), Jari

Yli-Kauhaluoma (1), Päivi Tammela (2), Paula Kiuru (1)

1) Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, P.O. Box 56,00014 University of Helsinki, Finland

2) Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, P.O. Box 56, 00014 University ofHelsinki, Finland

Marine sponges and other marine organisms are a rich source of novel compounds with drug-like

properties.1 They are considered as an extremely valuable source of new anticancer drugs.

2

Quinn and

co-workers identified two new bromotyrosine compounds clavatadine C (I) and D (II) (Fig. 1) from the marine

sponge Suberea clavata, and these showed anti-coagulation properties.

3

The spirocyclic bromotyrosines are

structurally more rigid and occupy better the chemical space than open-chain bromotyrosines making them

interesting scaffolds for medicinal chemistry.

4

In our earlier research we reported a set of some simplified

open-chain bromotyrosines of purpurealidin I with potential antiproliferative activity.5 In order to understand

better the structure-activity relationships (SAR) of bromotyrosines as selective cytotoxic agents towards cancer

cells, we synthesized a small library of spirocyclic clavatadine analogs (III) that were tested against a melanoma

cell line (A-375) and normal human skin fibroblast cell line (Hs27) for cytotoxicity.

6

Several of these

compounds showed anticproliferative activity but moderate selectivity against A-375 cell line over the Hs27 cell

line. This shows that the marine compounds are promising scaffolds for developing cytotoxic agents and further

optimization of simplified clavatadine derivatives is in progress to understand the elements of their SAR and

improve the required selectivity.

References

1) Kiuru, P. D'Auria, V. M., Muller, C. D., Tammela, P., Yli-Kauhaluoma, J., Vuorela, H. Exploring marine resources for

bioactive compounds. Planta Med. 2014, 80, 1234-1246.

2) https://www.midwestern.edu/departments/marinepharmacology/clinical-pipeline.xml 1.4.2020

3) Buchanan, M., Carroll, A., Wessling, D., Hooper, J., Quinn, R. J. Clavatadines C-E, Guanidine Alkaloids from the

Australian Sponge Suberea clavata. J. Nat. Prod. 2009, 72, 973–975.

4) Zheng, Y. J., Tice, C. M., The utilization of spirocyclic scaffolds in novel drug discovery. Expert Opin. Drug Dis. 2016,

11, 831-834.

5) Bhat, C., Tilli, I., Ilina, P., Voráčová, M., Barba, V., Hribernik, N., Lillsunde, K-E., Bruun, T., Mäki-Lohiluoma, E.,

Yli-Kauhaluoma, J., Kiuru, P., Tammela, P. Synthesis and antiproliferative activity of marine bromotyrosine purpurealidin I

and its derivatives. Mar. Drugs 2018, 16, 481.

6) Patel, P. A., Bruun, T., Ilina, P., Mäkkylä, H., Lempinen, A., Yli-Kauhaluoma, J., Tammela, P., Kiuru, P. S. Synthesis and

anticancer evaluation of spirocyclic bromotyrosine clavatadine C analogs. Marine Drugs 2021, (submitted)

356 | EFMC-ISMC

T017

ATROPOISOMERISM – A FRIEND OR A FOE? - THE EXAMPLE OF

MCL1 INHIBITORS

Zoltan Szlavik (1), Marton Csekei (1), Attila Paczal (1), Zoltan B Szabo (1), Szabolcs Sipos (1), Gabor

Radics (1), Agnes Proszenyak (1), Balazs Balint (1), James Murray (2), Ijen Chen (2), Roderick E

Hubbard (2), Ana-Leticia Maragno (3), Alain Bruno (3), Maia Chanrion (3), Olivier Geneste (3), Andras

Kotschy (1)

1) Servier Research Institute of Medicinal Chemistry, Zahony u 7., H-1031 Budapest, Hungary2) Vernalis (R&D) Ltd., Granta Park, Cambridge CB21 6GB, UK

3) Institute de Recherche Servier, 125 Chemin de Ronde, 78290 Croissy-sur-Seine, France

The interaction of drug molecules with their biological targets is driven by two major factors: strong, usually

directional interactions such as electrostatic forces and hydrogen bonds, and the shape complementarity of

hydrophobic surfaces also known as hydrophobic interactions. While for the majority of historic, “druggable”

targets the strong interactions play a dominant role in the drug-target interaction, for more recent target classes

(e.g. protein-protein interaction inhibitors) the complementarity of the shape of the drug and the target surface

are of great significance.

The inhibition of the interaction between the proteins Mcl-1, an antiapoptotic protein having a key role in tumor

survival, and its ligands has long been recognized as a promising way to target cancer cells but little progress

was made until recently [1-3] due to the difficulty of targeting the shallow and hydrophobic interaction surface.

 

The presentation will describe how we and other groups managed to overcome these difficulties promoting drug

candidates into clinical research for a target that was once considered undruggable. The emphasis will be on the

tools medicinal chemists can use to address the shape complementarity challenge, in particular sharing the

lessons we learnt about atropoisomerism while developing a fragment starting point into our clinical candidate

S64315/MIK665.

References

1) Kotschy, A. et al. Nature 2016, 538, 477−482.

2) Caenepeel, S. et al. Cancer Discovery 2018, 8, 1582.

3) Tron, A.E. et al. Nature Comm. 2018, 9, 5341.

ABSTRACT CANCELLED

EFMC-ISMC | 357

T018

WW-DOMAIN BASED INHIBITORS OF PD-1/PDL-1 INTERACTION

Juan Lizandra Pérez, Łukasz Berlicki

Department of Bioorganic Chemistry, Wrocław University of Science and Technology, Wybrzeże Wyspiańskiego 27, 50-370Wrocław

Programmed death 1 protein (PD-1) interaction with programmed death 1 ligand (PDL-1), modulates the evasion

of the immune system by cancer cells. This immunosuppressive mechanism induces apoptosis of tumor specific

T-cells. Blockade of this immune checkpoint can boost the host’s immune system, and hence, stimulate the

inherent ability of our immune system to attack tumor cells. So far, therapeutics, that counteract the unbalanced

immunoregulation of PD-1/PDL-1 interaction have been the monoclonal antibodies (mAbs), with some of them

already on the market

1

. Yet mAbs have strong specificity for the target, they lack penetration cell membrane and

present low oral administration in comparison with small molecules. To beat these drawbacks, we propose the

use of foldameric mini-proteins

2

, which can also reduce the intrinsic disadvantages of conventional

peptidomimetics like instability of physicochemical properties.

To achieve our goal, we propose to develop an iterative approach. Initially, with molecular modelling techniques

as Rosetta Fast Design, we generate inhibitors with affinity towards PDL-1, based on WW-domains

3

. With a

βββ topology they are closely related to the buried surface of PD-1. After characterization and activity studies,

the selected sequences are then remodeled towards higher affinity for PDL-1 based on experimental and

computational results. These motifs are simultaneously extended with addition β-amino acids containing

secondary structures, to attain higher stability and induce folding. These modifications will allow us mutations,

without losing the structure of the inhibitors, crucial for this category of drugs.

References

1) Waldman, A. D., Fritz, J. M., & Lenardo, M. J. A guide to cancer immunotherapy: from T cell basic science to clinical

practice. Nature Reviews Immunology, 2020, 20(11), 651–668.

2) Ran, X., & Gestwicki, J. E. Inhibitors of protein–protein interactions (PPIs): an analysis of scaffold choices and buried

surface area. Current Opinion in Chemical Biology, 2018, 44, 75–86.

3) Macias, M. J., & Gervais, V. Structural analysis of WW domains and design of a WW prototype. America, 2000, 375–379.

358 | EFMC-ISMC

T019

ENHANCING BINDING OF SPIROPYRAZOLINE OXINDOLES TO

MDMS BY IN SILICO HIT-TO-LEAD OPTIMIZATION

Elizabeth A. Lopes (1), Margarida Espadinha (1), Mi Wang (2), Shaomeng Wang (2), Cecília M. P.

Rodrigues (1), Daniel J. D. A. Santos (1,3), Maria M. M. Santos (1)

1) Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof Gama Pinto1649-003, Lisbon, Portugal.

2) Rogel Cancer Center, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109 USA.3) LAQV@REQUIMTE/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Rua do Campo

Alegre, 4169-007, Porto, Portugal

The p53 tumor suppressor protein is involved in several biological mechanisms that assure the integrity of the

human genome. However, in all types of human cancers, the p53 tumor suppressor function is inactivated by

mutation or gene deletion or by overexpression of negative regulators such as MDM2 and/or MDMX. In the

latter case, great efforts have been done to develop p53-MDM2 protein-protein interaction (PPI) inhibitors which

some have achieved clinical trials. Still, these efforts were not enough for a full reactivation of p53. Later, the

negative regulator MDMX has also emerged as an important target. Both MDM2 and MDMX interact with p53

by four amino acids, Phe19, Leu22, Trp23, and Leu26. Though, the structural and conformational differences

between the regulators hinder the use of p53-MDM2 PPI inhibitors as efficient p53-MDMX PPI inhibitors. For

this reason, the discovery of potent small molecules acting as dual p53-MDM2/X PPI inhibitors is still a

challenge. For a full reactivation of p53, development of drugs that disrupt the interaction of p53 with both

negative regulators is still an essential matter[1].

Our research team has been working on the development and optimization of indole-based compounds to obtain

dual p53-MDM2/X PPI inhibitors[2,3]. Previously, we have identified a spiropyrazoline oxindole derivative that

induces apoptosis and cell cycle arrest at G0/G1 phase, upregulates p53 steady-state levels, and leads to a

decrease of MDM2 levels. However, the compound did not bind to MDM2 in competitive binding assays[2].

Moreover, molecular docking studies have revealed that this derivative did not have important interactions

necessary to bind to MDM2. For this reason, we have been focusing our efforts on the development of new

spiropyrazoline oxindole derivatives that target both MDM2 and MDMX. In this communication, we report the

construction of virtual libraries of spiropyrazoline oxindoles derivatives by adding fragments to the scaffold to

obtain dual p53-MDM2/X PPIs inhibitors by structure-based virtual screening of these libraries over the

MDM2/X structures. Moreover, the hit compounds identified by in silico screening were further optimized and

30 new molecules were synthesized and evaluated in cancer cell lines and competitive binding assays for MDM2

and MDMX.

 

This work was supported by national funds through FCT - Fundação para a Ciência e a Tecnologia, I.P., under

the project PTDC/QUI-QOR/29664/2017, iMed.ULisboa (UIDB/04138/2020) and fellowships

SFRH/BD/137544/2018 (E. A. Lopes) and SFRH/BD/117931/2016 (M. Espadinha).

References

1) Espadinha M, Barcherini V, Lopes EA, Santos MMM. Curr. Top. Med. Chem. 2018, 18, 647-660.

2) Nunes R, Ribeiro CJA, Monteiro A, Rodrigues CMP, Amaral JD, Santos MMM. Eur. J. Med. Chem. 2017, 139, 168-179.

3) Amaral JD, Silva D, Rodrigues CMP, Solá S, Santos MMM Frontiers in Chemistry. 2019, 7, article 15.

EFMC-ISMC | 359

T020

DESIGN, SEMISYNTHESIS AND ANTIESTROGENIC ACTIVITY OF

LIGNAN DERIVATIVES FROM NATURAL

DIBENZYLBUTYROLACTONES

Priscila López-Rojas (1), Ángel Amesty (1), Miguel Guerra-Rodríguez (2), Leandro Fernández-Pérez (2),

Ana Estévez-Braun (1)

1) Instituto Universitario de Bio-Orgánica Antonio González, CIBICAN, Departamento de Química Orgánica, Universidadde La Laguna, Avda. Astrofísico Francisco Sánchez 2, 38296, La Laguna, Tenerife, España.

2) Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS). Departamento de Ciencias Clínicas,BIOPHARM. Universidad de Las Palmas de Gran Canaria. Las Palmas de Gran Canaria, 35001, España.

Natural products are an important source of new drugs and are also a suitable platform for the design and

preparation of new therapeutic agents. Lignans are a family of natural products with a wide range of biological

activities, including estrogenic activity. In fact, some of them are considered as phytoestrogens since they can act

as endocrine disruptors, selective estrogen receptor modulators (SERMs), and may have an estrogenic or

anti-estrogenic action depending on the tissue in which they are found. Marketed SERMs as raloxifen and

tamoxifen have achieved remarkable success in clinical therapies against breast cancer and menopause

symptoms, but its insufficient subtype selectivity on the ERα and ERβ causes adverse effects and drug resistance

has appeared in the last decades. Therefore, discovery of new SERMs is of great importance for the treatment of

breast cancer.

In this communication we will report the design, semisynthesis and antiestrogenic activity of a library of lignans

obtained from two natural dibenzylbutyrolactone lignans isolated from Bupleurum salicifolium.

Acknowledgment: We thank to MICINN (RTI2018-094356-B-C21), Gobierno de Canarias (ProID

2017010071) and FEDER for the financial support. P.L.R and M.G.R. thank MECD for a predoctoral grant A.A

thanks to Agustín de Betancort program.

References

1) M. Lahlou, Pharmacol. Pharm. 2013, 4, 17-31.

2) A. Solyomváry et al. Mini Rev. Med. Chem. 2017, 17, 1053-1074

3) F. Mauvais-Jarvis et al. Endocr Rev. 2013, 34, 309-38

4) Y. Zhu et al. PloS one, 2017, 12, e0171390

5) G. Guedes et al. Chem. Med. Chem., 2015, 10, 1403-1412

360 | EFMC-ISMC

T021

RATIONAL DESIGN, SYNTHESIS AND IN VITRO TESTING OF

SELECTIVE HDAC6 AND SIRT2 INHIBITORS

Dusan Ruzic (1), Nemanja Djokovic (1), Milos Petkovic (2), Danica Agbaba (1), Sheraz Gul (3), Maija

Lahtela-Kakkonen (4), A. Ganesan (5), Juan F. Santibanez (6), Tatjana Srdic-Rajic (7), Katarina Nikolic

(1)

1) Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11000Belgrade, Serbia; E-mail of the presenting author: [email protected]

2) Department of Organic Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11000 Belgrade,Serbia

3) Fraunhofer-IME SP, Hamburg, Germany4) School of Pharmacy (Pharmaceutical Chemistry) University of Eastern Finland, Kuopio, Finland

5) School of Pharmacy, University of East Anglia, Norwich Research Park, NR4 7TJ Norwich, United Kingdom6) Group for Molecular Oncology, Institute for Medical Research, University of Belgrade, Dr. Subotića 4, 11129 Belgrade,

Serbia7) Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade,

Serbia

Histone deacetylases (HDACs) are epigenetic enzymes involved in regulation of histone posttranslational

modifications and gene expression. Since changed function of HDACs is involved in pathogenesis of cancer

1-3

,

the HDAC inhibitors are extensive examined as promising anticancer agents. In our in silico study we have

combined structure-based, ligand-based, and fragment-based methodologies to design selective inhibitors against

cytoplasmic isoforms of HDAC, such as histone deacetylase 6 inhibitors (HDAC6) and SIRT2. The drug design

study has defined several promising selective HDAC6 and SIRT2 inhibitors for further synthesis and in vitrotesting. Based on the in vitro activities of the novel compounds in a panel of biochemical HDAC assays as well

as various cell-based assays were selected the most promising candidates for further investigation.

References

1) Lee, Ju-Hee, et al. "Development of a histone deacetylase 6 inhibitor and its biological effects." Proceedings of the

National Academy of Sciences 110.39 (2013): 15704-15709.

2) Hai, Yang, and David W. Christianson. "Histone deacetylase 6 structure and molecular basis of catalysis and inhibition."

Nature Chemical Biology (2016).

3) Rumpf, T. et al. Selective Sirt2 inhibition by ligand-induced rearrangement of the active site. Nat Commun. 2015, 6, 6263.

EFMC-ISMC | 361

T022

NOVEL PD-1-TARGETED INHIBITORS OF PD-1/PD-L1

INTERACTION DESIGNED BASING ON THE MVAT MINI-PROTEIN

STRUCTURE

Paweł Noceń, Łukasz Berlicki

Wybrzeże Wyspiańskiego street 27, 50-370 Wrocław, Poland

Pivotal role of the PD-1/PD-L1 interaction in the development of numerous cancer types is widely known and

precisely described in the literature. PD-1 protein is a receptor present on the surface of T-cells and constitutes a

checkpoint that activates the apoptotic pathway under the influence of certain stimuli, while PD-L1 is a ligand

protein which is overexpressed on the surface of various tumour cells. A strong interaction that occurs between

both the ligand and the receptor lead to lack of apoptotic response of the immune system for the appearance of

neoplasm[1,2].

Following the discovery of this crucial point in carcinogenesis, multiple therapeutic approaches had been

proposed and are currently in use, including usage of specific antibodies[3], cyclic peptides and other small

biologically active molecules that are shown to bring some, predominantly moderate, curative outcome[4].

 However, due to the fact that it is very difficult to efficiently inhibit the abovementioned interaction, there is

still a need to search for new strategies and propose novel, distinct structures of higher effectiveness to improve

the therapeutic benefits. These might arise owing to use of purposefully designed and synthesized peptides as

anti-cancer agents[5].

Employment of the rational drug design, especially computer-aided design approach supported with the

knowledge regarding the influence of single substitutions in range of a certain sequence, is one of the scientific

methodologies that gets more attention nowadays[6].

Our aim is to obtain an inhibitor of PD-1/PD-L1 interaction, targeted towards PD-1 receptor, basing on a known

scaffold derived from MvaT mini-protein (pdb: 2mxe)[7]. In order to implement a designed peptide as an actual

partner that would interact with the target protein, remodelling of the structure is applied. The idea is to make use

of structural properties of a synthesized peptide and the chemical characteristics of its amino acid residues

exposed towards cavities in the surface of the target protein. Besides affinity towards the receptor, another

crucial parameter to be taken into consideration during designing and synthesis of the peptide, is the

conformational stability of the product.

Firstly, designed peptides had been synthesized with use of microwave-assisted solid phase synthesis. Then, by

means of circular dichroism spectroscopy, the structural analyses were done. Binding of the products to the

target protein was investigated with use of biolayer interferometry method.

References

1) Okazaki, T., Honjo, T., Int Immunol., 2007, 19(7):813-24.

2) Chamoto, K. et al., Curr Top Microbiol Immunol., 2017, 410:75-97.

3) Hellmann, M.D., et al., N Engl J Med., 2019, 381(21):2020-2031.

4) Shaabani S, et al., Expert Opin Ther Pat., 2018, 28(9):665-678.

5) Fortuna, P. et al., Eur J Med Chem., 2020, 15; 208:112814.

6) Kuhlman, B., J Biol Chem., 2019, 294(50):19436-19443.

7) Ding, P. et al., PLoS Pathog, 2015, 11(6), e1004967.

362 | EFMC-ISMC

T023

SYNTHESIS OF NOVEL CK2/HDAC DUAL INHIBITORS BASED ON

PHARMACOPHORE HYBRIDIZATION STRATEGY

Irene Ortin, Claire Coderch, Jose Maria Zapico, Ana Ramos, Beatriz de Pascual-Teresa

Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities,Urbanización Montepríncipe, 28925, Alcorcón, Madrid, Spain.

One advancement in small molecule drug design is the multi-target approach. Designing a single entity that

efficiently inhibits more than one target instead of administering separate entities for different targets not only

improves patients’ adherence to therapy, but also reduces pharmacokinetic interactions, sides effects, and

manufacturing costs.[1] This multi-target activity can be achieved by emerging two pharmacophores

(dual-inhibitors) giving raise to what is known as “hybrid compounds”.

Protein Kinase 2 (CK2) and Histone DeAcetylases (HDACs) are enzymes with a significant role in oncogenesis,

where a variety of cancers have reported overexpression of both enzymes, thus, they are regarded as promising

drug targets for cancer therapy. Although there are many HDAC inhibitors (HDACi) in clinical trials, due to

drug resistance[2] the results of solid cancer treatments have not been satisfactory.[3] Interestingly, a SAR study

confirms that the cap group in HDACi is flexible and tolerates modification. Thus, the zinc-binding group

present in most of them can easily bind to different fragments with antitumor activity, thus enhancing the effect

of HDACi.[4] Since under hypoxic conditions,[5] CK2 phosphorylates HDAC leading to HDAC activation

which contributes to tumor growth and vasculogenesis, we predict that simultaneously inhibiting these two

targets by a bifunctional molecule should improve efficacy compared to single-target agents. Hence, here we

report novel potential multi-target single hybrid molecules that concurrently inhibit CK2/HDAC.

Acknowledgements: Financial support from RTI2018-093539-B-I00 (MICIU/FEDER, UE) is kindlyacknowledged.

 

 

References

1) Bérubé, G. An overview of molecular hybrids in drug discovery. Expert Opin Drug Discov, 2016, 11:3, 281–305.

2) Ni, X.; Li, L.; Pan, G., HDAC inhibitor-induced drug resistance involving ATP-binding cassette transporters (Review).

Oncol Lett 2015, 9 (2), 515-521.

3) Luan, Y.; Li, J.; Bernatchez, J. A.; Li, R., Kinase and histone deacetylase hybrid inhibitors for cancer therapy. J Med

Chem 2019, 62 (7), 3171-3183.

4) Stazi G, Fioravanti R, Mai A, Mattevi A, Valente S. Histone deacetylases as an epigenetic pillar for the development of

hybrid inhibitors in cancer. Curr Opin Chem Biol. 2019;50:89–100

5) Pluemsampant, S.; Safronova, O. S.; Nakahama, K.; Morita, I., Protein kinase CK2 is a key activator of histone

deacetylase in hypoxia-associated tumors. Int J Cancer 2008, 122(2), 333-41

EFMC-ISMC | 363

T024

DEVELOPMENT OF NOVEL TRYPTOPHANOL-DERIVED

OXAZOLOPYRROLIDONE LACTAMS WITH ANTIPROLIFERATIVE

ACTIVITY AGAINST HUMAN GASTRIC ADENOCARCINOMA (AGS)

CELL LINE.

Paulo A. F. Pacheco (1), Margarida Espadinha (1), Valentina Barcherini (1), Lídia M. Gonçalves (1), Elies

Molins (2), Alexandra M. M. Antunes (3), Maria M. M. Santos (1)

1) Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto,1649-003 Lisboa, Portugal

2) Institut de Ciència de Materials de Barcelona (ICMAB-CSIC), Campus UAB, 08193 Bellaterra, Spain3) Centro de Química Estrutural, Instituto Superior Técnico, ULisboa, 1049-001 Lisboa, Portugal

Gastric cancer (GC) is the fifth most common type of cancer and second leading cause of cancer-related death

worldwide (1). To date, radical surgery is the only treatment with curative potential, although its efficacy is

limited by the difficulty to eliminate all neoplastic cells (1). Previously, our research group has shown a

promising antitumor activity for tryptophanol-derived isoindolinones (2). In this work, we synthetized a series of

novel enantiopure tryptophanol-derived oxazolopyrrolidone lactams (Figure 1), following a chiral-induced

cyclocondensation reaction between the enantiopure tryptophanol and commercially available keto acids (3). The

target compounds were evaluated for their antiproliferative activity against human gastric adenocarcinoma

(AGS) cell line. Our results indicated that (R)-tryptophanol-derived oxazolopyrrolidone lactams were more

active than the corresponding enantiomers. In addition, we were able to identify a hit compound 5a (IC

50

= 3.4 ±

0.2 mM) that was subjected to chemical derivatization via Suzuki-Miyaura cross-coupling reaction. The

cytotoxicity assays with AGS and other tumoral cell lines revealed two promising derivatives (6a/b) with high

selectivity towards all cancer cell lines (3). Moreover, mechanistic studies indicated that these compounds

induced a significant increase of caspase 3/7 activity, suggesting that the antiproliferative activity is due to

apoptosis induction. Finally, we also performed in vitro stability studies in human plasma and human liver

microsomes and the identification of metabolites. The results indicated that the compounds are stable, and that

the main metabolites of these molecules are mono- and di-hydroxylated derivatives.

Figure 1 - Tryptophanol-derived oxazolopyrrolidones (4-6) with antiproliferative activity studied in this work.

Keywords: anticancer, apoptosis, oxazolopyrrolidone lactam, gastric cancer.

Acknowledgments: We thank FCT (Fundação para a Ciência e a Tecnologia) for financial support through

UIDB/04138/2020, UIDB/50006/2020, project PTDC/QUI-QOR/29664/2017, PD/BD/143126/2019 and

SFRH/BD/137544/2018

References

1) Sexton RE, Al Hallak MN, Diab M, Azmi AS. Gastric cancer: a comprehensive review of current and future treatment

strategies. Cancer Metastasis Rev. 2020;39(4):1179–203.

2) Soares J, Espadinha M, Raimundo L, Ramos H, Gomes AS, Gomes S, et al. DIMP53-1: A novel small-molecule dual

inhibitor of p53-MDM2/X interactions with multifunctional p53-dependent anticancer properties. Mol Oncol.

2017;11(6):612–27.

3) Espadinha M, Barcherini V, Gonçalves LM, Molins E, Antunes AMM, Santos MMM. Tryptophanol‐derived

oxazolopyrrolidone lactams as potential anticancer agents against gastric adenocarcinoma. Pharmaceuticals. 2021;14(3).

364 | EFMC-ISMC

T025

NEW POTASSIUM ION CHANNELS KV1.3 AND KV10.1 INHIBITORS

AS NEW ANTICANCER COMPOUNDS

Špela Gubič (1), Tihomir Tomašič (1), Žan Toplak (1), Louise Antonia Hendrickx (2), Jan Tytgat (2), Luis

A. Pardo (3), Lucija Peterlin Mašič (1)

1) University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, 1000 Ljubljana, Slovenia2) University of Leuven, Toxicology and Pharmacology, Campus Gasthuisberg, Onderwijs en Navorsing 2, He-restraat 49,

PO Box 922, 3000 Leuven, Belgium3) AG Oncophysiology, Max-Planck Institute for Experimental Medicine, Hermann-Rein-Str. 3, 37075 Göttingen, Germany

Exploitation of genuinely novel classes of cancer targets provides great opportunities for the discovery of new

anticancer compounds. The voltage-gated potassium channel K

V

10.1 is considered a near- universal tumour

marker and represents a promising new target for the discovery of novel anticancer drugs.

1

K

V

1.3 channels

relevance in cancer therapy stems from the combination of its direct implication in apoptosis and its properties as

an immunomodulator in the tumour microenvironment. Therefore, K

v

1.3 inhibitors have the potential to

eliminate resistance to apoptosis in tumour cells and at the same time to minimise the pro-tumoral actions of

immune cells in the microenvironment.

2

We utilized the ligand-based drug discovery methodology using 3D pharmacophore modelling and medicinal

chemistry approaches to prepare a novel structural class of K

V

10.1 inhibitors, which inhibited the channel in a

voltage-dependent manner consistent with the action of a gating modifier.

3

Structural optimization resulted in the

most potent K

V

10.1 inhibitor of the series with an IC

50

value of 740 nM, which was potent on the MCF-7 cell

line expressing high K

V

10.1 levels and low hERG levels, induced significant apoptosis in tumour spheroids of

Colo-357 cells, and was not mutagenic.

3

We also identified a new structural class of K

v

1.3 inhibitor based on the marine alkaloid clathrodinand based on

3D similarity searches.

4

With the aid of new homology models of human K

v

1.3 channels, recent developments

have been based on available X-ray structures of related K

v

1.2 channels, and these have opened up possibilities

for discovery of new K

v

1.3 inhibitors as potential anticancer compounds that act via novel and unique

mechanisms. We also synthesized Kv1.3 inhibitors for selective delivery to mitochondria, and these offer

therapeutic potential for the treatment of cancers.

Computational ligand-based drug design methods can be successful in the discovery of new potent K

V

10.1 and

K

V

3.1 inhibitors.

References

1.) Toplak Ž, Hendrickx LA, Abdelaziz R, Shi X, Peigneur S, Tomašič T, Tytgat J, Peterlin Mašič L, Pardo LA. Overcoming

challenges of HERG potassium channel liability through rational design: Eag1 inhibitors for cancer treatment. Accepted in

Medicinal Research Reviews, 2021.

2.) Gubič Š, Hendrickx LA, Toplak Ž, Sterle M, Peigneur S, Tomašič T, Pardo LA, Tytgat J. Zega A, Peterlin Mašič

L.Discovery of KV1.3 ion channel inhibitors: medicinal chemistry approaches and challenges. Accepted in Medicinal

Research Reviews, 2021.

3. ) TOPLAK Ž, HENDRICKX LA, GUBIČ Š, MOŽINA Š, ŽEGURA B, ŠTERN A, NOVAK M, SHI X, TOMAŠIČ T,

PARDO LA, PETERLIN MAŠIČ L. 3D pharmacophore-based discovery of novel Kv10.1 inhibitors with antiproliferative

activity. Cancers, 2021, 13, 1-24.

4.) HENDRICKX LA, DOBRIČIĆ V, TOPLAK Ž, PEIGNEUR S, PETERLIN-MAŠIČ L, TOMAŠIČ T, TYTGAT J.

Design and characterization of a novel structural class of Kv1.3 inhibitors. Bioorganic chemistry, 2020, 98, 1-9.

EFMC-ISMC | 365

T026

MULTIPLE-TARGETING LIGANDS FOR AKR1C3 ENZYME AND

ANDROGEN RECEPTOR TO TARGET PROSTATE CANCER

Agnese Chiara Pippione (1), Zuhal Kilic-Kurt (2), Stefano Sainas (1), Iole Mannella (1), Barbara Rolando

(1), Sandra Kovachka (1), Francesca Spyrakis (1), Annamaria Buschini (3), Serena Montalbano (3),

Simonetta Oliaro Bosso (1), Marco Lolli (1), Donatella Boschi (1)

1) University of Torino, Dept of Science and Drug Technology, via Pietro Giuria 9, Torino, Italy2) Ankara University, Faculty of Pharmacy, Dept of Pharmacology, Yenimahalle, 06100, Ankara, Turkey

3) University of Parma, Dept of Chemistry, Life and Env. Sust. Sciences, Parco Area delle Scienze 11/a, Parma, Italy

The enzyme aldo-keto reductase 1C3 (AKR1C3) is considered an attractive target in Castration Resistant

Prostate Cancer (CRPC) for its role in the biosynthesis of androgens (testosterone and DHT).

1

At the same time,

antagonists to Androgen Receptor (AR) as enzalutamide and darolutamide are used as anticancer drugs in CRPC

patients. The two targets are compatible in terms of ligand accommodation as they are both able to interact with

DHT as product and ligand, respectively.

Our group already discovered new potent AKR1C3 inhibitors, where hydroxylated heterocycles were used to

mimic acidic functions.

1,2

We here propose new derivatives for both the hydroxybenzoisoxazole and the

hydroxytriazole series, and move our attention on obtaining multiple ligands for both AKR1C3 and AR.

To do this, we identified structural elements required for activity on AR and found some AKR1C3 inhibitors

able to play AR antagonism activity; this represent a starting point for multiple-targeting ligand development

applied to CRPC. The potency and dual action of new compounds are translated into citotoxicity against CRPC

cellular models.

In silico design, synthesis and biological activity on AKR1C3 enzyme of new compounds, as well as their

capability to address AR, are here described.

References

1) Pippione, A.C.; Carnovale, I.M.; Bonanni, D.; Boschi, D.; Oliaro-Bosso, S., Lolli M.L et al. Eur. J. Med. Chem. 2018;

150, 930-945.

366 | EFMC-ISMC

T027

NEW CHROMENE DERIVATIVES: OVERCOMING RENAL CELL

CARCINOMA DRUG RESISTANCE

Olívia Pontes (1,2), Sofia Oliveira-Pinto (1,2), Belém Sampaio-Marques (1,2), Ana Carolina Laus (3),

Luciane S. da Silva (3), Paula Ludovico (1,2), Rui M. Reis (1,2,3), Fátima Baltazar (1,2), Fernanda

Proença (4), Marta Costa (1,2)

1) Life and Health Sciences Research Institute (ICVS), University of Minho, Campus of Gualtar, Braga, Portugal2) ICVS/3Bs - PT Government Associate Laboratory, Braga/Guimarães, Portugal

3) Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo, Brazil4) Department of Chemistry, University of Minho, Campus of Gualtar, Braga, Portugal

Renal cell carcinoma (RCC) is a common type of cancer in the urological system and accounts for 90 to 95% of

malignant neoplasms that arise from the kidney. The number of RCC patients is expected to increase drastically

until 2040. The estimated incidence of RCC and mortality will rise in 62% and 78%, respectively, comparing

with 2020 statistics. Due to late diagnosis, metastatic RCC is very recurrent, requiring systemic treatment

combined with surgery. However, drug resistance eventually becomes problematic due to genetic and epigenetic

modifications or downstream pathway activation.

Inspired by in silico screening results, new chromene scaffolds fused to an imidazole moiety were synthetized

using optimized and innovative experimental methods. The anticancer potential of this set of new derivatives

was explored in several RCC cell lines (786-O, Caki-2 and A498) and non-neoplastic HK2 cells. Moreover,

RCC cells were subjected to drug pressure with stepwise increasing dosage over time to develop resistance to

commercially available drugs and then attempting to overcome it with chromenes treatment. After in vitrostudies, C. elegans model was used to study toxicity in vivo and the Chorioallantoic Membrane (CAM) assay to

assess in vivo tumor cell proliferation and angiogenesis.

After several months, cells developed drug resistance to Rapamycin 7-fold and Cediranib 4-fold higher than the

IC

50

value of the respective parental cell line. This resistance was confirmed by increased expression levels of

their target proteins (mTOR and VEGFR). Chromenes tested on parental, drug-resistant and non-neoplastic cells

presented IC

50

values in the nanomolar range and high selectivity indexes. Moreover, the new chromenes

inhibited cell proliferation, induced apoptosis, arrested cell cycle in S and G

2

/M phases and inhibited tumour

metabolism. In vivo toxicity studies showed that the compounds were well-tolerated and safe, even at

significantly higher concentrations than those used in the in vitro tests. Finally, in vivo tumour regression and

inhibition of angiogenesis were also potentiated by these novel chromenes.

This study demonstrates the effectiveness and safety of these new chromene derivatives to treat RCC. More

importantly, resistance features were overcame successfully using the synthetized chromenes, offering a new

chance for RCC patients with this aggressive disease. Overall, our study opens new perspectives for the

treatment of resistant renal cell cancer to targeted therapy.

References

Sung H, Ferlay J, Siegel R.L, Laversanne M, Soerjomataram I, Jemal A, Bray F, Global Cancer Statistics 2020:

GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin.

2021;0:1–41.

Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA Cancer J Clin 2019; 69(1): 7-34.

Nizam A, Schindelheim JA, Ornstein MC. The role of active surveillance and cytoreductive nephrectomy in metastatic renal

cell carcinoma. Cancer Treat Res Commun 2020; 23: 100169.

Ravaud A, Gross-Goupil M. Overcoming resistance to tyrosine kinase inhibitors in renal cell carcinoma. Cancer Treat Rev

2012; 38(8): 996-1003.

EFMC-ISMC | 367

T028

EPIGENETIC DRUG DISCOVERY: NOVEL PROMISING EZH2

INHIBITORS AGAINST CANCER

Filipa Ramilo-Gomes (1,2), Sharon D. Bryant (3), Riccardo Martini (4), Thierry Langer (4), Sheraz Gul

(5), Oliver Keminer (5), Luís Sobral (6), Rita C. Guedes (2), M. Matilde Marques (1)

1) Centro de Química Estrutural, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa,Portugal

2) iMed.ULisboa, Faculdade de Farmácia da Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal3) Inte:Ligand Software Entwicklungs und Consulting, Mariahilferstrasse 74B, 1070 Vienna, Austria

4) Department of Pharmaceutical Chemistry, Faculty of Life Sciences, University of Vienna, Althanstraße 14, A-1090Vienna, Austria

5) Department of Pharmaceutical Chemistry, Faculty of Life Sciences, University of Vienna, Althanstraße 14, A-1090Vienna, Austria; e) Fraunhofer IME-SP, Schnackenburgallee 114, 22525 Hamburg, Germany

6) Hovione Farmaciência SA, Sete Casas, 2674-506 Loures, Portugal

Cancer figures among the leading causes of morbidity and mortality worldwide. Recently, epigenetic pathways

have been recognized as determinants to cancer development and progression. Polycomb repressive complex 2

(PRC2) is an epigenetic regulator that catalyzes the trimethylation of lysine 27 in Histone 3 (H3K27me3), a

process that facilitates chromatin compaction and gene silencing [1]. The overexpression of EZH2, the catalytic

subunit of PRC2, is implicated in the development and progression of a variety of cancers with the worst

prognosis. Thus, the therapeutic targeting of EZH2 emerged as a hot topic and the development of selective

small-molecule EZH2 inhibitors is currently a promising research challenge for drug discovery [2].

A combination of state-of-the-art techniques, from computational drug design to synthetic methodologies and

biological testing, are being used to develop the new molecules. We performed a computer-aided drug design

campaign to design new EZH2 inhibitors using LigandScout [3]. A panel of unique pharmacophore models were

generated, validated and optimized. The prioritized models were used for two hit finding campaigns: Virtual

Screening and De Novo Design. For the Virtual Screening approach, several databases (e.g., DrugBank, NCI,

MuTaLig Chemotheca, and our in-house libraries) were computed and screened. Interesting virtual hit molecules

with high inhibition potential were found and tested in order to determine their EZH2 profiles. Notably, we

found several hits with inhibition profiles comparable to the reference compounds. In parallel, we started a De

Novo Design campaign based on selected pharmacophore models and we found a new scaffold for EZH2

inhibitors. Those from de novo design were synthesized and tested. The potential toxicity issues were also

assessed through metabolism studies. Finally, selectivity and binding mode of the most promising compounds

are being elucidated. The best drug candidates are expected to proceed to in vivo testing.

 

Thanks are due to grant PD/BD/128320/2017 from Fundação para a Ciência e a Tecnologia (FCT) and projects

UID/QUI/00100/2019, UID/DTP/04138/2019, PTDC/QUI-QAN/32242/2017, and SAICTPAC/0019/2015,

funded by national funds through FCT and when appropriate co-financed by FEDER under the PT2020

Partnership Agreement. This communication is based upon work from COST Action CA15135, supported by

COST.

References

1) A. Brooun, K. S. Gajiwala, Deng, Y.-L, et al. Nat. Comm. 2016, 7 11384

2) K. H. Kim and C. W. M. Roberts, Nat Med. 2016, 22 128–134

3) LigandScout Molecular Design Software from InteLigand GmbH (http://www.inteligand.com)

368 | EFMC-ISMC

T029

DRUG DESIGN AGAINST CARCINOGENESIS: DEVELOPMENT OF

NEW NADPH OXIDASE INHIBITORS AS A PATHWAY TO CANCER

THERAPY

Joana Reis (1), Christoph Gorgulla (2,3), Marta Massari (1), Sara Marchese (1), Haribabu Arthanari

(2,3), Andrea Mattevi (1)

1) Department of Biology and Biotechnology “L. Spallanzani,” University of Pavia, 27100 Pavia, Italy2) Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, USA

3) Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, USA

Reactive Oxygen Species (ROS) is associated to cancer biology, from cell transformation, proliferation,

invasion, or tumor survival. ROS production is also involved in oncogene expression through mitogenic action,

and in the mechanism of many if not most anticancer drugs [1].

NADPH-oxidases (NOXs) are the only known enzymes whose sole function is ROS generation and are known

to modulate pathways that induce and sustain cell proliferation (Figure 1). This notion is exemplified by the vital

role of NOX4 in the development of the pancreatic ductal carcinoma, possibly the deadliest cancer. NOX4 is

also investigated as target for fibrosis and its overexpression has been associated to tumour progression in lung

cancer cells, renal cell carcinoma, colorectal cancer, melanoma, and ovarian cancer [2].

Our research group has been successful in accomplishing the first crystal structure of the cytosolic

dehydrogenase (PDB: 5o0x) and trans-membrane (PDB: 5o0t) domains of NOX using a bacterial ortholog which

is highly similar (40% sequence identity) to human NOX5 [3]. Taking advantage of the fact that we have a

highly conserved active site between all NOX isoforms we have employed the same NOX5 dehydrogenase

domain as our initial platform to carry on a drug design campaign. To this end, together with our collaborators at

Harvard Medical School and Dana Farber Cancer Institute, we have conducted an ultra large-library

computational screening using our NOX5 dehydrogenase domain PDB structure. We have evaluated the chosen

library using a robust and high throughput workflow comprising primary and orthogonal biochemical assays as

well as control assays to probe assay interfering compounds and ROS scavengers [4]. Furthermore, binding

assays such as microscale thermophoresis, surface plasmon resonance and thermal shift experiments were also

performed for inhibitor validation purposes. Protein crystallography studies of the protein with the best hits have

yielded for the first-time a crystal structure of this class of enzymes in complex with our studied inhibitors.

Overall, our results show compounds acting in the submicromolar range displaying a mechanism of inhibition

dependant on the retention of the FAD cofactor within the active site. Accordingly, our current efforts rely on the

study and development of new and effective isoform-specific NOX inhibitors and the understanding of their

effect on cancer model cells in which NOXs have a key role. As a result, this work will give us insight into the

biology of ROS signalling and establish this family of enzymes as potential targets in chemotherapeutic

treatment with a focus on NOX4 and its roles in carcinogenesis.

 

Figure 1 - The biological role of NOXs in ROS signalling and cancer as well as the importance of developing

bonafide NOX inhibitors as opposite to ROS scavengers.

References

1) Irani, K. et al. (1997) Science 275, 1649-1652

2) Lambeth, J.D. et al. (2014) Annu. Rev. Pathol. 9, 119-145.

3) Magnani, F. et al. (2017) Proc. Natl. Acad. Sci USA 114, 6764-6769

4) Reis, J. et al. (2020) Redox Biology 32, 101466

EFMC-ISMC | 369

T030

DISCOVERY AND OPTIMIZATION OF

OXIDATIVE-PHOSPHORYLATION INHIBITORS FROM A

PHENOTYPIC SCREEN

Florian Richalet, Mahmoud El Shemerly, Laurenz Kellenberger, Lane Heidi, Sven Weiler, Dimitri Robay

Basilea Pharmaceutica AG Int, 487, Grenzacherstrasse, CH-4005 Basel

Mitochondria are considered the powerhouse cells, generating most of the ATP which is used as a source of

chemical energy (1). In addition, mitochondria are involved in functions, such as cellular signaling,

differentiation and death, as well as maintaining control of the cell cycle and cell growth (2).

Cancer cells reprogram their metabolism in favour of glycolysis, regardless of oxygen presence, according to a

phenomenon known as aerobic glycolysis. This so-called “Warburg phenotype” involves high glucose uptake

and a high glycolytic activity (3). Nevertheless, cancer cells are also dependent on mitochondria for ATP

production through oxidative phosphorylation (OXPHOS) (4, 5).

Mitochondrial metabolism is now recognized as a potential target for anticancer agents due to the metabolic

characteristic of cancer cells. Indeed, human cancer is associated with mitochondrial dysregulation, which

promotes cancer cell survival, tumor progression and metastases as well as resistance to current anticancer

therapies (4, 6, 7). 

Change of carbon sources in growth medium for HeLa cells from glucose (Glc) to galactose (Gal) allows the

switch of metabolism from glycolysis towards OXPHOS . Therefore, cancer cells are expected to show

differential sensitivity for an OXPHOS inhibitor depending on whether the cells are grown in Gal or in Glc

media.

Using the above concept, we carried out an HTS with ~100 000 in-house compounds. Triaging yielded B-001 as

a hit with promising differential sensitivity. Additional mode of action studies revealed that B-001 inhibits ATP

production and decreases oxygen consumption through inhibition of complex I of the electron transport chain

(ETC).

SAR investigations rapidly yielded potent leads of OXPHOSi with low nM activity but rather high xlogP. We

therefore embarked on a lead optimization campaign based on lipophilicity-driven hypothesis. Further improved

analogues displayed improved activity, significantly increased Lipophilic Ligand Efficiency - LLE (8) and –in

line with the working hypothesis- reduced promiscuity in off-target assays (hERG, CEREP).

Advanced analogues showed promising efficacy in xenograft models upon oral dosing. In order to mitigate

on-target related side effects different dosing regimen were tested in vivo and the search for suitable combination

partners is currently ongoing.

References

1) Campbell N.A., Williamson B., Heyden R.J. Biology: Exploring Life 2006th Edition, Publisher: Pearson Prentice Hall,

2006

2) McBride H.M. et.al., Curr. Biol., vol. 16, no.14, R551-60, 2006

3) Warburg O., Science, vol. 123, no. 3191, pages 309-314, 1956

4) Marchetti P. et al., International Journal of Cell Biology, vol. 2015, pages 1-17, 2015

5) Solaini G. et al., Biochim. Biophys. Acta,, vol. 2, page: 314-323, 2010

6) Boland M.L. et al., Frontieres in Oncology, vol. 3, Article 292, pages 1-28, 2013

7) Solaini G. et al., Biochim. Biophys. Acta, vol. 1797, pages 1171-1177, 2010

8) Leeson P.D., Springthorpe B., Nat. Rev. Drug Discov. 2007;6(11):881-90

370 | EFMC-ISMC

T031

DESIGN AND SYNTHESIS OF SPIROCYCLIC AZOLE Mcl-1

INHIBITORS

Fedor Romanov-Michailidis, Adriana Ingrid Velter, Meng-Yang Hsiao, Ann Vos, Frederik Rombouts

Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340 Beerse, Belgium

Avoidance of apoptosis is critical for the development and sustained growth of tumors. The pro-survival protein

myeloid cell leukemia 1 (Mcl-1) is an antiapoptotic member of the Bcl-2 family of proteins which is

overexpressed in many cancers. Upregulation of Mcl-1 in human cancers is associated with high tumor grade,

poor survival, and resistance to chemotherapy. Therefore, pharmacological modulation of Mcl-1 represents an

attractive approach to cancer therapy. Here we disclose the design and synthesis of potent and selective

small-molecule inhibitors of Mcl-1. We show that the introduction of two salient structural elements—an azole

heteroaromatic ring and a macrocycle—into a spirocyclic scaffold leads to compounds with single-digit

nanomolar inhibitory activity and provides a foundation for further exploration of potent and physchem-balanced

Mcl-1 inhibitors.

ABSTRACT CANCELLED

EFMC-ISMC | 371

T032

SYNTHESIS OF SECOND GENERATION FLUORESCENT TRIPPPRO

COMPOUNDS FOR CELLULAR UPTAKE STUDIES

Maria Roßmeier (1), Simon Remus (2), Udo Schumacher (2), Chris Meier (1)

1) Organic Chemistry, Depatment of Chemistry University of HamburgMartin-Luther-King-Platz 6, 20146 Hamburg (Germany)

2) Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Martinistraße 52,20251 Hamburg (Germany)

Nucleoside analogues (NAs) are widely used drugs in chemotherapy. The active metabolite of these compounds

is the NA 5’-triphosphate (NA-TP) and therefore, NAs have to undergo intracellular metabolism, specifically

kinase-catalyzed phosphorylation. Unfortunately, these phosphorylation steps are often inefficient and

importantly, reversible thus highly limiting the drugs’ biological effect. Consequently, the desired therapeutic

effect is only reached by high doses of the drug, which also increases undesired/off-target effects.

We have previously developed the TriPPPro technology to deliver antiviral NA-TPs into cells and thereby

bypass limiting metabolism and thus increase the biological effect. In TriPPPro-compounds, two cleavable

masks are attached to the g-phosphate of the TP. These groups compensate the negative charges of the TP so that

it can penetrate the cell membrane. Intracellularly, the masks are cleaved by unspecific esterases/lipases to

release the biologically active NA-TP. Previous studies demonstrated that replacing one cleavable mask with a

non-cleavable alkyl group greatly increases stability of the thus modified TP in biological media and, at the same

time, enhances selectivity of the biological effect and reduces off-targets effects.

We here applied our second generation TriPPPro-technology (TriPPPro

2

) to probe compound uptake and

metabolism in cancer cells with an intrinsically fluorescent bicyclic nucleoside analogue (BCNA).

Various TriPPPro

2

-BCNAs were synthesized that combine different cleavable masks and stable modifications.

The combination of different chain lengths for the TriPPPro

2

-BCNAs led to different lipophilicity of the

compounds. Under chemical (PBS) and enzymatical (PLE) conditions/media, the compounds demonstrated high

stability and, as expected, the main product of hydrolysis was the g-alkylated BCNA-TP. In cellular uptake

studies, all TriPPPro

2

compounds were able to penetrate the cell membrane and again, as desired, released the

g-alkylated BCNA-TP as main product.

These (proof-of-principle) results underline that the TriPPPro

2

-technology may increase the efficiency of NA

anticancer drugs and relating studies are currently ongoing in our labs.

372 | EFMC-ISMC

T033

SYNTHESIS, MOLECULAR MODELLING AND BIOLOGICAL

CHARACTERIZATION OF NOVEL ANTIMIGRATORY AND

ANTIINVASIVE 1-BENZHYDRYL PIPERAZINE DERIVATIVES

Ruzic Dusan (1,3), Petkovic Milos (2), Ellinger Bernhard (3), Gul Sheraz (3), Santibanez Juan F. (4),

Srdic-Rajic Tatjana (5), Nikolic Katarina (1)

1) Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221Belgrade, Serbia

2) Department of Organic Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade,Serbia

3) Fraunhofer Institute for Molecular Biology and Applied Ecology Screening Port, D-22525 Hamburg, Germany4) Group for Molecular Oncology, Institute for Medical Research, University of Belgrade, Dr. Subotića 4, 11129 Belgrade,

Serbia5) Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade,

Serbia

Human epigenetic metalloenzymes that modulate the acetylation status of histones, alter cancer cell morphology

and cell survival are histone deacetylases (HDACs). Of particular importance, histone deacetylase 6 is studied as

a cytoplasmic isoform implicated in the microtubule dynamics in cancer

1

. Still, more efforts need to be

undertaken to make these inhibitors reach to global oncology market

2

. In this study, we probed the 1-benzhydryl

piperazine as the capping (CAP) group to selectively target the HDAC6 isoform and alter the migration and

invasiveness of the breast cancer cell lines. Nine different 1-benzhydryl piperazine derivatives were synthesized

and the structure-activity relationship study was postulated with combined ligand-based (3D-QSAR) and

structure-based (molecular docking) in silico approaches

3,4

. We performed wound healing, matrigel invasion

and transwell migration assays to search for the inhibitor that shows antimigratory and antiinvasive properties of

the breast cancer cell lines (MDA-MB-231 and MCF-7). Most of the synthesized compounds induce apoptosis in

high concentrations (> 60 μM) in cell viability assay, whereas the antimigratory and antiinvasive effects were

significantly pronounced at subapoptotic concentrations (5 μM). One of the nine synthesized inhibitors showed

excellent non-cytotoxic, antimigratory and antiinvasive profile in breast cancer cell lines, which is in agreement

with the proposed cellular roles of HDAC6 in cancer.

The work presented in this study integrates in silico modelling, synthesis and in vitro biological profiling to

discover selective HDAC6 inhibitor. Identification of potent HDAC6 inhibitor that alters migration and

invasiveness of breast cancer cell lines opens up new horizons to treat metastatic diseases.

References

1) Banik, Debarati, et al. "HDAC6 plays a noncanonical role in the regulation of antitumor immune responses, dissemination,

and invasiveness of breast cancer." Cancer Research 80.17 (2020): 3649-3662.

2) He, Xingrui, et al. "Novel Selective Histone Deacetylase 6 (HDAC6) Inhibitors: A Patent Review (2016-2019)." Recent

patents on anti-cancer drug discovery 15.1 (2020): 32-48.

3) Ruzic, Dusan, et al. "Combined Ligand and Fragment‐based Drug Design of Selective Histone Deacetylase–6 Inhibitors."

Molecular informatics 38.5 (2019): 1800083.

EFMC-ISMC | 373

T034

A DIFFERENTIATING AND APOPTOTIC THERAPY FOR ACUTE

MYELOID LEUKAEMIA USING MEDS433, A POTENT HUMAN

DIHYDROOROTATE DEHYDROGENASE INHIBITOR

S. Sainas (1), P. Circosta (2), M. Giorgis (1), A.C. Pippione (1), M. Marraudino (3), M. Houshmand (3), B.

Bonaldo (3), V. Gaidano (4), A. Cignetti (4), S. Gotti (3), G. Saglio (4), S. Al-Karadaghi (5), D. Boschi (1),

M.L. Lolli (1)

1) Department of Science and Drug Technology, Università degli Studi di Torino, Torino, Italy2) Molecular Biotechonolgy Center, Università degli Studi di Torino, Torino, Italy

3) Neuroscience Institute Cavalieri Ottolenghi NICO, Università degli Studi di Torino, Orbassano, taly4) Department of Clinical and Biological Sciences,Università degli Studi di Torino, Orbassano, Italy

5) Department of Biochemistry and Structural Biology, Lund University (Sweden);

In acute myeloid leukaemia (AML), blasts lose their ability to differentiate into mature cells and undergo

apoptosis. Accordingly, a proapoptotic and differentiating therapy (arsenic and all trans retinoic acid, ATRA) has

dramatically improved survival in acute promyelocytic leukaemia; however, such combination therapy is not

available for other AML subtypes. While, in 2016, inhibition of dihydroorotate dehydrogenase (DHODH), a key

enzyme of the pyrimidine biosynthesis, was found to induce differentiation in several AML models. In fact,

brequinar (BRQ) was utilized in vivo studies.

1

We are optimising hDHODH inhibitors to improve potency and

drug-like proprieties. Moreover, we would like to evaluate how different parameters such as, pKa

, LogD

7.4

of

different carboxylic acid bioisosteres can influence in vitro and in vivo studies. The main objective is to identify

the best inhibitor suitable for use in in vivo studies on AML animal model. In this work we will present a new

generation of hDHODH inhibitors able to reach the enzymatic BRQ inhibition potency levels. Our data showed

that MEDS433, the best of two series, was found able to restore the myeloid differentiation in leukaemia cell

lines (U937 and THP1) at concentrations one digit lower than those achieved in experiments with BRQ.

Furthermore, we characterized MEDS433 with in vitro and in vivo experiments, showing that it had a significant

pro-apoptotic effect in several AML and CML cell lines, which was at least partially independent from the

differentiating effect.

3,4

Furthermore, MEDS433 had a significant pro-apoptotic effect on several AML cell

lines, but not on non-AML cell lines. Finally, our preliminary results from in vivo experiments showed that i)MEDS433 wasn’t toxic on Balb/c mice after 5 weeks of intraperitoneal administration at two different doses 10

and 25 mg/Kg and during acute toxicity experiment was not toxic ad dose of 1 g/Kg; ii) the half-life was limited

to 3-4 hours and iii) MEDS433 had a good antileukemic activity (approximately 50% reduction of the tumour

volume compared with control, after 18 days of treatment in THP1-xenograft models obtained from NSG mice).

Theoretical design, modeling, synthesis, SAR, X-ray crystallographic data, biological assays, Drug-Like

proprieties, pharmacokinetic studies and in vivo evaluations on AML models will be here presented and

discussed.

References

1) Sykes DB, et al. Inhibition of Dihydroorotate Dehydrogenase Overcomes Differentiation Blockade in Acute Myeloid

Leukemia. Cell 2016, 167(1):171-186.e15.

2) S. Sainas, et al. Targeting myeloid differentiation using potent 2-hydroxypyrazolo[1,5-a]pyridine scaffold-based human

dihydroorotate dehydrogenase inhibitors. J Med Chem 2018, 61 (14), 6034-6055.

3) Gaidano V., et al. The Synergism Between DHODH Inhibitors and Dipyridamole Leads to Metabolic Lethality in Acute

Myeloid Leukemia.Cancer 2021 13(5), 1003,

4) Stefano Sainas, et al Targeting Acute Myelogenous Leukemia using potent human dihydroorotate dehydrogenase

inhibitors based on the 2-hydroxypyrazolo[1,5-a]pyridine scaffold: SAR of the biphenyl moiety. J Med Chem 2021, in press.

374 | EFMC-ISMC

T035

CYCLIC DINUCLEOTIDE STING AGONISTS WITH A NOVEL

SULFAMATE LINKER SHOW ANTI-TUMOR ACTIVITY IN MOUSE

MODELS FOLLOWING SYSTEMIC ADMINISTRATION

Gilles C. Bignan (2), Stuart L. Emanuel (2), Marcel Viellevoye (1), Jan W. Thuring (1), Peter J. Connolly

(2), James P. Edwards (2), Szeman R. Chan (2), Leopoldo L. Luistro (2), Melissa Smith (2), Heng K. Lim

(2), Shefali Patel (2), Daniel J. Krosky (2), Salam Ibrahim (2), Victoria Wong (2), Jason Rowand (2),

Michael Allegrezza (2), Glenn Van Aller (2), Cynthia Milligan (2), Paul Shaffer (2), Wim Schepens (1)

1) Janssen Research & Development, Beerse, Antwerp, Belgium2) Janssen Research & Development, Spring House, PA, United States

Natural cyclic dinucleotides (CDNs), such as the human variant 2’,3’-cyclic GMP-AMP (2’,3’-cGAMP), have

been identified as ligands of the Stimulator of Interferon Genes (STING) protein. Upon CDN binding, STING

triggers a signalling cascade resulting in interferon-α and -β (IFN) production, which in turn induce innate and

adaptive anti-tumor immune responses. This finding has sparked huge interest from the scientific community to

develop CDN-based STING agonists as potential immunotherapeutic agents for cancer therapy. Natural CDNs

suffer however from poor resistance towards nucleases and low cell-membrane permeability which limit their

use in vivo and/or requires the need for intra-tumoral delivery, which would be of significant disadvantage in a

clinical setting. Substantial research to address this liability - mainly attributed to the presence of a phosphate

backbone - has been conducted over the past years, leading to the disclosure of CDN’s with non-natural furanose

and nucleobase replacements, as well as bio-isosteric phosphate modifications. The reported anti-tumor activity

in syngeneic mouse models of such modified CDN’s was impressive, however these were most often only

achieved by intra-tumoral administration. To overcome this shortcoming, we focused our design on modified

CDN analogues harboring a charge neutral sulfamate linker as a bioisosteric phosphate replacement. Within this

series, molecules have been identified that bind human STING and elicit a stronger activation of the protein

compared to 2’,3’-cGAMP, resulting in significantly increased secretion of signature cytokine IFN-β in a THP1

monocyte cell-line. In addition, we discovered that properly positioning the sulfamate linkage confers improved

chemical and in vitro enzymatic stability by eliminating the ENPP1 (ectonucleotide pyrophosphatase /

phosphodiesterase-1) cleavage site. Further in vivo evaluation of a selected compound in CT26 and MC38 colon

carcinoma syngeneic mouse models showed strong antitumor efficacy after intratumoral, subcutaneous and

intravenous administration.  In conclusion, this study demonstrates antitumor activity of a CDN derived STING

agonist in clinically relevant carcinoma mouse models following systemic administration. 

EFMC-ISMC | 375

T036

TARGETING TRANSCRIPTIONAL REPROGRAMMING IN CANCER:

EFFICACY OF CDK8/19 INHIBITORS ALONE AND IN

COMBINATION WITH CONVENTIONAL TREATMENTS

Alexander Shtil (1), Mengqian Chen (2), Victor Tatarskiy (3), Eugenia Broude (2), Igor Roninson (2)

1) Blokhin Cancer Center, 24 Kashirskoye shosse, Moscow 115478, Russia2) University of South Carolina, 715 Sumter Street, Columbia 29208 SC USA

3) Institute of Gene Biology, 34/5 Vavilov Street, Moscow 119334, Russia

Plasticity of the gene transcription machinery, that is, its ability to adapt mRNA synthesis to various conditions,

is a remarkable property of living systems. Development of chemical tools for regulation of specific

transcriptional events such as transcription factor targeting, prevention of binding to the DNA, inhibition of

RNA polymerase II, etc. makes it possible to select an individual compound for inactivation of a particular

mechanism. Given that these instruments frequently affect transcription in a genome wide manner, minimization

of general toxicity is a prerequisite for their clinical applicability. Transcriptional reprogramming, a fine tuning

of expression of actively transcribed genes, is vital in the embryo but largely dispensable in the adult organism in

which it is confined to gene regulation in a stimulus- and cell type specific context. This mechanism is mediated

via a CDK8/19 kinase module of the Mediator complex. Selective inhibitors of these kinases (CDK8/19i), being

non-toxic for adult cells in the course of a long-term exposure, efficiently prevented activation of a subset of

genes upon engagement of nuclear factor kappa B or estrogen receptor pathways [1,2]. Taking advantage of

water solubility, stability and bioavailability of CDK8/19i, we challenged these compounds as single agents

and/or components of antitumor combinations in several experimental settings. We found that, at submicromolar

concentrations, Senexin B and two structurally unrelated CDK8/19i induced an apoptotic death in acute

myelogenous leukemia cells whereas normal blood cells remained unaffected. These pivotal results suggest that

CDK8/19i can be an alternative to the cytotoxic treatment especially for elderly patients. Furthermore,

CDK8/19i synergized with the prototypic imatinib and the 3d generation Bcr-Abl inhibitors in killing chronic

myelogenous leukemia cells. Also, CDK8i potentiated the efficacy of γ-irradiation in HCT116 colon carcinoma

cells. Together with attenuation of emergence of resistance to drugs acting via the epidermal growth factor

receptor [3], we claim that CDK8/19i represent a novel class of drug candidates for the malignancies driven by

transcriptional reprogramming.

References

1) Chen et al. PNAS 2017, 114, 10208.

2) McDermott et al. Oncotarget 2017, 8, 12558.

3) Sharko et al. Cells 2021, 10, 144.

376 | EFMC-ISMC

T037

MEDICINAL CHEMISTRY DISCOVERY STORY OF AZD3229: A

PAN-MUTANT KIT INHIBITOR FOR THE TREATMENT OF GIST

James Smith

Oncology, R&D, AstraZeneca,Cambridge UK

While the treatment of gastrointestinal stromal tumors (GISTs) has been significantly advanced by the

application of targeted tyrosine kinase inhibitors able to inhibit KIT-driven proliferation, diverse mutations to

this kinase can drive resistance to established therapies such as Imatinib. We embarked upon a drug discovery

program focused on the development of KIT inhibitors which could address such resistance issues through the

pursuit of a profile with strong in vitro inhibitory activities against multiple clinically-relevant KIT mutants.  

We begun our medicinal chemistry program starting from a series of previously reported phenoxyquinazoline

and quinoline based ‘type 2’ kinase inhibitors of PDGFRα (e.g. 1).  We optimised potency against a diverse

panel of mutant KIT driven Ba/F3 cell lines, with a particular focus on reducing activity against a Ba/F3 derived

KDR driven cell model in order to minimise the potential for hypertension commonly seen in current second and

third line GIST therapies.  During the course of the medicinal chemistry development program, we investigated

compounds with a structural shift to an alternative amide linker of the original series, and further establishment

of SAR for this new scaffold allowed us to optimise towards more potent analogues with suitable kinase

selectivity and DMPK properties.  

As a result of this progress, and aided through X-ray protein crystallography showing key ligand binding modes,

we identified clinical candidate N-(4-{[5-Fluoro-7-(2-methoxyethoxy)quinazolin-4-yl]-

amino}phenyl)-2-[4-(propan-2-yl)-1H-1,2,3-triazol-1-yl]- acetamide (AZD3229) which demonstrates potent

single digit nM growth inhibition across a range of cell lines, and a good margin to KDR-driven effects. 

AZD3229 demonstrates excellent cross-species pharmacokinetics, shows robust in vivo pharmacodynamic

inhibition of KIT mutants, and is active in several disease-relevant in vivo models of GIST.

 

References

1) Kettle, J. et. al. J Med. Chem. 2018, 61, 8797-8810.

2) Ple, P. A. et. al. Bioorg. Med. Chem. Lett. 2012, 22, 262−266.

3) Ple, P. A et. al. Bioorg. Med. Chem. Lett. 2012, 22, 3050−3055.

EFMC-ISMC | 377

T038

SYNTHESIS AND CYTOTOXIC ACTIVITY EVALUATION OF NOVEL

HYDRAZONE DERIVATIVES AS POTENTIAL ANTICANCER

AGENTS

Hayrunnisa Tasci (1), Melda Sarıman (2), Engin Ulukaya (3), Birsen Tozkoparan (1), Nesrin Gokhan

Kelekci (1)

1) Hacettepe University, Faculty of Pharmacy Dept of Pharmaceutical Chemistry, 06100 Ankara-Turkey2) Istinye University, Molecular Cancer Research Center, 34010 Istanbul-Turkey

3) Istinye University, Faculty of Medicine, Dept of Clinical Biochemistry, 34010 Istanbul-Turkey

Hydrazone structure that constituted from an “azomethine” R-C(H)=N-N(H)-Ar group is a privileged

pharmachophoric group and has significant position in the field of medicinal chemistry. They have been

demonstrated to possess antibacterial, anticonvulsant, antitubercular, antidepressant, analgesic,

anti-inflammatory, antiviral, antiplatelet, antimalarial activities (1). In addition to, investigations revealed that

this group has also significant role as antitumor agent (2). As known, cancer is defined as a disease in which a

group of abnormal cells grow uncontrollably by out of the normal process of cellular division. This pathological

circumstance proceeds from several reason which some are clarified. Disruption of the structure of proteins,

formation of oncogenes from protooncogenes, loss of function of tumor suppressor genes (p53) and

malfunctioning of apoptosis mechanism assumed as that have a part in constitution of cancer. Continuing

searches have demonstrated that various hydrazone derivatives exhibited anticancer activity by inhibiting RNA

and DNA synthesis (3), inhibiting mitosis (3), induced caspase-dependent apoptosis (4), inhibiting tubulin

polymerization and cause cell cycle arrests in G2/M phase (4), cancer cycle arrest at sub G1/G0 phase(5), tumor

cell apoptosis (6) in different cancer cells (Figure 1).

In this study, we aimed to synthesize and evaluate of some new hydrazone derivatives bearing benzoxazolinone

through antitumor activity. These derivatives were synthesized by reacting

3-(5-methyl-2-benzoxazolinone)propanoicacid hydrazide with various aldehydes, their structures were

confirmed using IR,

1

H-NMR, mass and elemental analysis. The cytotoxicity and anticancer activity of these

compounds were evaluated against different cancer cell lines using a Sulphorhodamine-B (SRB) test.

Acknowledgments

Funding for this project was provided by the Hacettepe University, Scientific Research Projects Coordination

Unit (Project number: THD-2020-18624).

References

1) Kumar V, J Pharmacogn Phytochem, 2018, 7(2): 40-43.

2) Kumar D, Kumar NM, Ghosh S, Shah K. Bioorg & Med Chem Lett, 2012, 22, 212-215.

3) Kaplanek R, Havlik M, Dolensky B, Rak J, Dzubak P, Konecny P, Hajduch M, Kralova J, Kral V. Bioorg & Med Chem,

2015, 23:1651-1659.

4) Tantak MP, Klingler L, Arun L, Kumar A, Sadana R, Kumar D. Eur. J Med. Chem., 2017, 136:184-194.

5) Senkardes S, Basu NK, Durmaz I, Manvar D, Basu A, Atalay R and Kucukguzel SG. Eur J Med Chem., 2016,

108:301-308.

6) Shen S, Chen H, Zhu T, Ma X, Xu J, Zhu W, Chen R, Xie J, Ma T, Jia L, Wang Y, Peng C. Oncology. Letters, 2017,

13:3169- 3176.

378 | EFMC-ISMC

T039

NEW HSP90 C-TERMINAL INHIBITORS: HIT DISCOVERY, HIT

OPTIMIZATION AND EVALUATION

Tihomir Tomašič, Živa Zajec, Jaka Dernovšek

University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, 1000 Ljubljana, Slovenia

To date, mainly Hsp90 N-terminal domain (NTD) inhibitors have been developed for cancer treatments;

however, several of these have not been successful in clinical trials. One of the major drawbacks associated with

Hsp90 NTD inhibition is induction of the pro-survival heat-shock response. This response results in increased

levels of Hsp90 and antiapoptotic proteins, which initiate antiapoptotic cascades and promote drug resistance. In

contrast, Hsp90 C-terminal domain (CTD) inhibitors do not induce the heat shock response and are therefore

promising novel agents for cancer treatment.

One challenge associated with CTD inhibitors is the lack of a co-crystallized complex, requiring the use of a

predicted allosteric apo pocket, limiting structure-based (SB) design approaches. To address this, a unique

approach that enables the derivation and analysis of interactions between ligands and proteins from molecular

dynamics (MD) trajectories was used to derive pharmacophore models for virtual screening (VS) and identify

suitable binding sites for SB design. Furthermore, ligand-based (LB) pharmacophores were developed using a

set of CTD inhibitors to compare VS performance with the MD derived models. Virtual hits identified by VS

with both SB and LB models were tested for antiproliferative activity. The most potent VS hit displayed

antiproliferative activities in MCF-7 and Hep G2 cancer cell lines, inhibited Hsp90-dependent refolding of

denatured luciferase and induced the degradation of Hsp90 clients without concomitant induction of Hsp70

levels. In addition, novel Hsp90 CTD inhibitors with unique scaffolds were identified by structure-based VS

using docking. Structure-activity relationship data from hit expansion studies on different scaffolds of Hsp90

CTD inhibitors now provide a solid background for the ongoing hit-to-lead optimization program.

Acknowledgements:

This study was funded by the Slovenian Research Agency (Grant No. P1-0208, J1-1717, BI-US/18-19078).

References

1) Tomašič T et al. (2020). Int J Mol Sci. 21 (18): 6898.

EFMC-ISMC | 379

T040

IMPROVEMENT OF METABOLIC WEAKNESS OF NEW HUMAN

DIHYDROOROTATE DEHYDROGENASE INHIBITORS BASED ON

2-HYDROXYPYRAZOLO[1,5-A]PYRIDINE SCAFFOLD

C. Vigato (1), S. Sainas (1), M. Giorgis (1), P. Circosta (2), A. Passoni (3), A. C. Pippione (1), R. Bagnati

(3), G. Saglio (4), D. Boschi (1), M. L. Lolli (1)

1) Department of Science and Drug Technology, University of Torino (Italy)2) Molecular Biotechonolgy Center, University of Torino (Italy)

3) Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri2 - 20156 Milano (Italy)

4) Department of Clinical and Biological Sciences, University of Torino (Italy)

Human Dihydroorotate Dehydrogenase (hDHODH), a mitochondrial enzyme that plays a pivotal role in the de

novo pyrimidine biosynthesis, has been associated to Acute Myelogenous Leukemia (AML), as hDHODH

inhibitors are able to restore myeloid differentiation (1). In recent years, we designed potent hDHODH inhibitors

applying a scaffold-hopping approach to brequinar’s structure (2). By investigating the lead compound’s SAR,

we recently discovered compound 1 (Figure), a candidate superior to brequinar in terms of in vitro potency.

Unfortunately, compound 1 showed in vitro metabolic instability, as it soon undergoes to hydroxylation on

alkoxy side chain by microsomal enzymes (3). In this occasion, we investigated on the metabolic hydroxylation

site through the synthesis of the possible products of microsomal metabolism according to literature (4) and their

comparison to compound 1’ s own metabolite in high resolution mass spectrometry (HR-MS); then we designed

a new generation of hDHODH inhibitors protected from metabolic oxidation on the alkoxy side chain. Design,

synthesis, in vitro metabolism and biological characterization of the new developed compounds are here

described and discussed.

References

1) Sykes, D. B. et al. Inhibition of Dihydroorotate Dehydrogenase Overcomes Differentiation Blockade in Acute Myeloid

Leukemia. Cell. 2016, 167(1), 171-186

2) Sainas, S. et al. Targeting Myeloid Differentiation Using Potent 2-Hydroxypyrazolo[1,5- a]pyridine Scaffold-Based

Human Dihydroorotate Dehydrogenase Inhibitors. J. Med. Chem. 2018, 61, 6034-6055.

3) Sainas, S. et al. Targeting Acute Myelogenous Leukemia using potent human dihydroorotate dehydrogenase inhibitors

based on the 2-hydroxypyrazolo[1,5- a]pyridine scaffold: SAR of the biphenyl moiety. J. Med. Chem. 2021, in press.

4) Testa, B. and Clement, B. Biotransformation Reactions and Their Enzymes. The Practice of Medicinal Chemistry: Fourth

Edition. 2015, 561-584

380 | EFMC-ISMC

T041

HOW pH COULD AFFECT THE INTERACTION STRENGTH DURING

PD-L1 INHIBITORS RECOGNIZING

Alessandra Riccio (1), Andrea Mammoli (1), Alice Coletti (2), Elisa Bianconi (1), Andrea Carotti (1),

Antonio Macchiarulo (1)

1) Department of Pharmaceutical SciencesUniversity of Perugia

via del liceo n.1, 06123 Perugia (Italy)2) Department of Medicine and Surgery University of Perugia

P.le Gambuli, 06132 Perugia (Italy)

PD-1/PD-L1 belong to immune checkpoint molecules which provide the correct functioning of the immune

cells.[1] Recently, it has been proposed that inhibition of PD-1/PD-L1 axis could lead to a promising approach in

order to defect cancer, avoiding immune-escape mechanism from immune cells exploited by tumor.[2] Taking

into account the pH shift which occurs in tumor-microenvironment, we developed an integrated approach to

identify new selective PD-L1 inhibitors which are resilient to variable pH condition and therefore, effective

against cancer.[3] This work aims to identify both the critical residues on target surface and ionizable groups on

the ligand involved in electrostatic interactions understanding how these are strengthened or weakened by shift

of pH in tumor microenvironment. Once observed in silico the critical polar interactions between PD-L1 and

inhibitors, we performed experimental pKa measurements and MST assays in order to reveal potential ligand

physical-chemical features required to interact with PD-L1.Collectively, the results obtained will be able to

provide further insights into structure activity relationship of PD-L1 inhibitors that are useful for the design of

next generation small molecules inhibitors with physical chemical properties that are resilient to variable pH

conditions of tumor microenvironment.

References

1) Zak, K. M. et al. Structure 23, 2341–2348 (2015)

2) Constantinidou, A., et al. Pharmacol. Ther. 194, 84–106 (2019)

3) Korenchan, D. Cancers, 11, 1026 (2019)

EFMC-ISMC | 381

Posters - Drug Discovery ProjectsU. Targeting Fibrotic Diseases with Small Molecules

382 | EFMC-ISMC

U001

GPR84 A DRUGGABLE PROINFLAMMATORY RECEPTOR

Loukas Ieremias (1), Mads Kaspersen (1,2), Asmita Manandhar (1), Christoffer Vogsen Heidtmann (2),

Elisabeth Rexen Ulven (1), Laura Jenkins (3), Graeme Milligan (3), Trond Ulven (1)

1) Department of Drug Design and Pharmacology, Faculty of Health, University of Copenhagen, Copenhagen, Denmark2) Department of Physics, Chemistry and Pharmacy, Faculty of Science, University of Southern Denmark, Odense, Denmark

3) Institute for Molecular, Cell and System Biology, College of Medical, Veterinary and Life Sciences, University ofGlasgow, Glasgow, UK

GPR84 is a proinflammatory receptor expressed mainly in the bone marrow and leukocytes and in lesser extent

on the lung, kidney and adipose tissue

1

. Though still considered an orphan receptor, it is activated by medium

chain fatty acids

1

and mediates release of inflammatory cytokines by leukocytes

2-4

. Activation of GPR84

reduces atherogenesis

5

and may be beneficial for obesity and type-II diabetes

6, 7

. Protective effects towards

osteoporosis

8

, rheumatoid arthritis

8

, multiple sclerosis

9

and Alzheimer’s disease

9

have also been reported. On the

contrary, GPR84 is upregulated in diseases such as neuropathic pain

3

, acute myeloid leukemia

10

, lupus

erythematosus

11

, NASH

12

and fibrosis

13

in several organs. Despite GPR84 being involved in several poorly

treated diseases, drugs targeting the receptor have not been developed yet. We here present our work on design,

synthesis and pharmacological characterization of new ligands modulating GPR84.

References

1) Wang, J.; Wu, X.; Simonavicius, N.; Tian, H.; Ling, L., Medium-chain Fatty Acids as Ligands for Orphan G

Protein-coupled Receptor GPR84. J. Biol. Chem. 2006, 281 (45), 34457-34464.

2) Suzuki, M.; Takaishi, S.; Nagasaki, M.; Onozawa, Y.; Iino, I.; Maeda, H.; Komai, T.; Oda, T., Medium-chain Fatty

Acid-sensing Receptor, GPR84, Is a Proinflammatory Receptor. J. Biol. Chem. 2013, 288 (15), 10684-10691.

3) Nicol, L. S. C.; Dawes, J. M.; La Russa, F.; Didangelos, A.; Clark, A. K.; Gentry, C.; Grist, J.; Davies, J. B.; Malcangio,

M.; McMahon, S. B., The Role of G-Protein Receptor 84 in Experimental Neuropathic Pain. J. Neurosci. 2015, 35 (23),

8959-8969.

4) Recio, C.; Lucy, D.; Purvis, G. S. D.; Iveson, P.; Zeboudj, L.; Iqbal, A. J.; Lin, D.; O’Callaghan, C.; Davison, L.;

Griesbach, E.; Russell, A. J.; Wynne, G. M.; Dib, L.; Monaco, C.; Greaves, D. R., Activation of the Immune-Metabolic

Receptor GPR84 Enhances Inflammation and Phagocytosis in Macrophages. Front. Immunol. 2018, 9:1419.

5) Gaidarov, I.; Anthony, T.; Gatlin, J.; Chen, X.; Mills, D.; Solomon, M.; Han, S.; Semple, G.; Unett, D. J., Embelin and its

derivatives unravel the signaling, proinflammatory and antiatherogenic properties of GPR84 receptor. Pharmacol. Res. 2018,

131, 185-198.

6) Feltrin, K. L.; Little, T. J.; Meyer, J. H.; Horowitz, M.; Rades, T.; Wishart, J.; Feinle-Bisset, C., Comparative effects of

intraduodenal infusions of lauric and oleic acids on antropyloroduodenal motility, plasma cholecystokinin and peptide YY,

appetite, and energy intake in healthy men. Am. J. Clin. Nutr. 2008, 87 (5), 1181-1187.

7) Symonds, E. L.; Peiris, M.; Page, A. J.; Chia, B.; Dogra, H.; Masding, A.; Galanakis, V.; Atiba, M.; Bulmer, D.; Young,

R. L.; Blackshaw, L. A., Mechanisms of activation of mouse and human enteroendocrine cells by nutrients. Gut 2015, 64 (4),

618-626.

8) Park, J.-W.; Yoon, H.-J.; Kang, W. Y.; Cho, S.; Seong, S. J.; Lee, H. W.; Yoon, Y.-R.; Kim, H.-J., G protein-coupled

receptor 84 controls osteoclastogenesis through inhibition of NF-κB and MAPK signaling pathways. J. Cell. Physiol. 2018,

233 (2), 1481-1489.

9) Bouchard, C.; Pagé, J.; Bédard, A.; Tremblay, P.; Vallières, L., G protein-coupled receptor 84, a microglia-associated

protein expressed in neuroinflammatory conditions. Glia 2007, 55 (8), 790-800.

10) Dietrich, P. A.; Yang, C.; Leung, H. H. L.; Lynch, J. R.; Gonzales, E.; Liu, B.; Haber, M.; Norris, M. D.; Wang, J.;

Wang, J. Y., GPR84 sustains aberrant β-catenin signaling in leukemic stem cells for maintenance of MLL leukemogenesis.

Blood 2014, 124 (22), 3284-3294.

11) Zhu, H.; Mi, W.; Luo, H.; Chen, T.; Liu, S.; Raman, I.; Zuo, X.; Li, Q.-Z., Whole-genome transcription and DNA

methylation analysis of peripheral blood mononuclear cells identified aberrant gene regulation pathways in systemic lupus

erythematosus. Arthrit. Res. Ther. 2016, 18 (1), 162.

12) Puengel, T.; De Vos, S.; Hundertmark, J.; Kohlhepp, M.; Guldiken, N.; Pujuguet, P.; Auberval, M.; Marsais, F.; Shoji, K.

F.; Saniere, L.; Trautwein, C.; Luedde, T.; Strnad, P.; Brys, R.; Clément-Lacroix, P.; Tacke, F., The Medium-Chain Fatty

Acid Receptor GPR84 Mediates Myeloid Cell Infiltration Promoting Steatohepatitis and Fibrosis. J. Clin. Med. 2020, 9 (4),

1140.

13) Gagnon, L.; Leduc, M.; Thibodeau, J.-F.; Zhang, M.-Z.; Grouix, B.; Sarra-Bournet, F.; Gagnon, W.; Hince, K.;

Tremblay, M.; Geerts, L.; Kennedy, C. R. J.; Hebert, R. L.; Gutsol, A.; Holterman, C. E.; Kamto, E.; Gervais, L.;

Ouboudinar, J.; Richard, J.; Felton, A.; Laverdure, A.; Simard, J.-C.; Letourneau, S.; Cloutier, M.-P.; Leblond, F. A.; Abbott,

S. D.; Penney, C.; Duceppe, J.-S.; Zacharie, B.; Dupuis, J.; Calderone, A.; Nguyen, Q. T.; Harris, R. C.; Laurin, P., A Newly

Discovered Antifibrotic Pathway Regulated by Two Fatty Acid Receptors: GPR40 and GPR84. The American journal of

pathology 2018, 188 (5), 1132-1148.

EFMC-ISMC | 383

U002

SYNTHESIS OF

2-((1H-INDOL-3-YL)METHYL)-5-(ALKYLTHIO)-1,3,4-OXADIAZOLES

AND THEIR PROTECTIVE ACTIVITY AGAINST OXIDATIVE

STRESS

Vida Malinauskienė (1), Alena Kadlecová (2), Jiří Voller (2,3), Lucie Janovská (4,5), Monika Iškauskienė

(1), Asta Žukauskaitė (6), Algirdas Šačkus (1)

1) Department of Organic Chemistry, Kaunas University of Technology, Radvilėnų pl. 19, LT-50254 Kaunas, Lithuania2) Department of Experimental Biology, Faculty of Science, Palacký University, Šlechtitelů 27, CZ-78371 Olomouc, Czech

Republic3) Department of Clinical and Molecular Pathology, Institute of Molecular and Translational Medicine, Faculty of Medicine

and Dentistry, Palacký University, Hněvotínská 3, CZ-77515Olomouc, Czech Republic

4) Department of Microbiology, Faculty of Medicine and Dentistry, Palacký University, Hněvotínská 3, CZ-77515 Olomouc,Czech Republic

5) Laboratory of Growth Regulators, Institute of Experimental Botany of the Czech Academy of Sciences & Faculty ofScience, Palacký University, Šlechtitelů 27, CZ-78371 Olomouc, Czech Republic

6) Department of Chemical Biology, Faculty of Science, Palacký University, Šlechtitelů 27, CZ-78371 Olomouc, CzechRepublic

It is already known that 5-alkyl/aryl substituted indole-containing 1,3,4-oxadiazoles demonstrate antifungal

activity [1], while amino bridge-bearing analogues show anticonvulsant activity [2]. Biological activities of

analogous sulphur bridged compounds are understudied, with an exception of anticancer activity possessing

2-((1H-indol-3-yl)methyl)-5-((ethoxymethyl)thio)-1,3,4-oxadiazole [3]. To fill in this gap, in the present work

we present fast and effective route towards a collection of new sulphur bridge-containing 1,3,4-oxadiazoles

starting from indole-3-acetic acid (IAA) esters and assessment of their biological activities [4].

A small library of 2-((1H-indol-3-yl)methyl)-5-(alkylthio)-1,3,4-oxadiazoles was prepared starting from

indole-3-acetic acid methyl ester and its 5-methyl substituted derivatives via formation of corresponding

intermediate hydrazides. Their immediate base-mediated treatment with carbon disulfide, followed by in situacidification gave rise to corresponding 5-[(1H-indol-3-yl)methyl]-1,3,4-oxadiazole-2(3H)-thione derivatives.

Finally, S-alkylation reactions were performed using various alkylating agents.

All 5-[(1H-indol-3-yl)methyl]-1,3,4-oxadiazole-2(3H)-thiones and their S-alkylated derivatives were screened

for their toxic and protective effects in vitro and in vivo. Three of the prepared compounds protected

FA-fibroblasts against glutathione depletion induced by glutathione synthesis inhibitor BSO. The activity of the

most active compound was comparable to that of R-lipoic acid. Subsequent toxicity evaluation on a panel of

human cells showed that in contrast to R-lipoic acid, these compounds marginally decrease viability of cultures

at 60 µM concentration. Therefore, their possible use in FA, where cell homeostasis is severely perturbed at

multiple levels, requires caution.

References

1) Zhang M-Z, Mulholland N, Beattie D, et al (2013) Synthesis and antifungal activity of 3-(1,3,4-oxadiazol-5-yl)-indoles

and 3-(1,3,4-oxadiazol-5-yl)methyl-indoles. Eur J Med Chem 63:22–32. DOI: 10.1016/J.EJMECH.2013.01.038.

2) Siddiqui N, Alam MS, Ahsan W (2008) Synthesis, anticonvulsant and toxicity evaluation of

2-(1H-indol-3-yl)acetyl-N-(substituted phenyl)hydrazine carbothioamides and their related heterocyclic derivatives. Acta

Pharm 58:445–454. DOI: 10.2478/v10007-008-0025-0.

3) Abu-Zaied M A .Z., Nawwar G. A. M., Swellem R H., et al (2012) Synthesis and Screening of New

5-Substituted-1,3,4-oxadiazole-2-thioglycosides as Potent Anticancer Agents. Pharmacol & Pharm 03:254–261.

DOI:10.4236/pp.2012.32034.

4) Iškauskienė M., Kadlecová A., Voller J., Janovská L., Malinauskienė V., Žukauskaitė A., Šačkus A. Synthesis of

5-[(1H-indol-3-yl)methyl]-1,3,4- oxadiazole-2(3H)-thiones and their protective activity against oxidative stress, Archiv der

Pharmazie, Accepted 2021. DOI: 10.1002/ardp.202100001

384 | EFMC-ISMC

U003

DESIGN, SYNTHESIS, AND STRUCTURE-ACTIVITY RELATIONSHIP

STUDIES OF DUAL INHIBITORS OF SOLUBLE EPOXIDE

HYDROLASE AND 5-LIPOXYGENASE

Kerstin Hiesinger (1), Jan Sebastian Kramer (1), Sandra Beyer (2), Timon Eckes (2), Steffen Brunst (1),

Cathrin Flauaus (3), Sandra Kerstin Wittmann (1), Lilia Weizel (1), Astrid Kaiser (1), Sven George (1),

Simon Kretschmer (1), Carlo Angioni (4), Jan Heering (5), Gerd Geisslinger (4,5), Manfred

Schubert-Zsilavecz (1), Achim Schmidtko (3), Denys Pogoryelov (6), Josef Pfeilschifter (2), Bettina

Hofmann (1), Dieter Steinhilber (1,5), Stephanie Schwalm (2), Ewgenij Proschak (1,5)

1) Institute of Pharmaceutical Chemistry, Goethe-University of Frankfurt, D-60438 Frankfurt a.M.,Germany

2) Institute of General Pharmacology and Toxicology, Pharmazentrum Frankfurt, ZAFES, D-60590Frankfurt a.M., Germany

3) Institute of Pharmacology and Clinical Pharmacy, Goethe-University of Frankfurt, D-60438 Frankfurt a.M., Germany4) Institute of Clinical Pharmacology, Pharmazentrum Frankfurt, ZAFES, D-60590 Frankfurt a.M., Germany

5) Branch for Translational Medicine and Pharmacology, Fraunhofer Institute for Molecular Biology and Applied EcologyIME, D-60590 Frankfurt a.M., Germany

6) Institute of Biochemistry, Goethe-University of Frankfurt, D-60438 Frankfurt a.M., Germany

Over the past years designed multitarget drugs gained importance in medicinal chemistry. Modulating more than

one target can lead to synergistic effects which result in higher efficacy. Moreover, the safety profile and patient

compliance can be improved. One area where synergistic anti-inflammatory effects play an important role is the

inhibition of the arachidonic acid (ara) cascade.[1] Metabolites of the ara cascade induce inflammation and

allergic reactions. Inhibition of 5-lipoxygenase (5-LOX) reduces levels of leukotrienes. In addition, inhibition of

soluble epoxide hydrolase (sEH) leads to increased levels of the anti-inflammatory epoxyeicosatrienoic acid.

Merging pharmacophores of sEH and 5-LOX derived a dual sEH/5-LOX inhibitor, which was successfully

optimized in context of target affinity and metabolic stability. The lead structure showed its potency on cellular

level in human polymorphonuclear leukocytes, acceptable metabolic stability, oral bioavailability, and target

engagement in vivo. We demonstrated the anti-inflammatory and anti-fibrotic efficiency of the dual sEH/5-LOX

inhibitor using a kidney injury model (unilateral ureteral obstruction) in mice. Further evaluation in other

fibrosis- and inflammation-related disease models could demonstrate the superior effect of dual target

engagement.

References

1) J.-Y. Liu, J. Yang, B. Inceoglu, H. Qiu, A. Ulu, S.-H. Hwang, N. Chiamvimonvat, B. D. Hammock, Biochem. Pharmacol.

2010, 79, 880−887.

EFMC-ISMC | 385

U004

EFFECTS OF DATE FRUIT EXTRACTS (PHOENIX DACTYLIFERA

L.) ON PANCREATIC CANCER CELLS IN-VITRO

Younis Baqi

Chemistry Department, College of Science, Sultan Qaboos University, Muscat, [email protected]

Background: Date palm fruit (Phoenix dactylifera L.) is an endemic functional food, with great nutritional and

economic importance due to its phytochemical compositions. The microenvironment of pancreatic cancer

consists of cellular and acellular components, including fibroblasts, myofibroblasts, pancreatic stellate cells

(PSCs), immune cells, blood vessels, extracellular matrix (ECM) and soluble proteins, such as cytokines and

growth factors.

Objectives: The aim of this study is to prepare and fractionate natural product extracts of the edible part of date

palm fruit. The extracts and its fractions were investigated for antifibrotic and antiproliferative activities in tumor

necrosis factor (TNF) stimulated PSCs.

Methods: Maceration method was employed to prepare four different extract of the date fruit utilizing a single

solvent system including water, ethanol, acetone, and ethyl acetate. Column chromatography technique was used

to fractionate the ethyl acetate crude extract using methanol/dichloromethane solvent system. PSCs were imaged

using a Leica DM IRBE microscope. FN1 and α-SMA were determined using immunoblotting analysis. The

antiproliferative activity of the date fruit extracts was determined based on the Resazurin assay, and DCFH-DA

was used to measure the reactive oxygen species (ROS).

Results: Date fruit fractions reduced fibrosis, decreased PSC activity and reversed the PSCs’ fibrotic phenotype.

The results indicate the presence of a compound or mixture of compounds, which promote the reversal of PSCs

to their quiescent state, detectable by suppression of the secretion of α-SMA.

Conclusion: The findings suggest a new approach for targeting pancreatic cancer through the modulation of

PSC activity, thereby possibly enhancing the effect of known anticancer drugs. Moreover, date palm fruit

appears to have chemopreventive activity protecting from pancreatic and probably other types of cancer.

386 | EFMC-ISMC

NOTES

EFMC-ISMC | 387

Posters - Drug Discovery ProjectsV. Tissue and Cell Specific Drug Delivery

388 | EFMC-ISMC

V001

SELEC-PROTACS: NOVEL CANCER TARGETING PROTEIN

DEGRADERS

Eleen Laul, Katherine Rollins, Cristina Nevado

Department of Chemistry, University of Zurich, Zurich, Switzerland

PROteolysis TArgeting Chimeras (PROTACs) are bifunctional probes capable of hijacking the cells’ natural

proteasomal degradation machinery to completely degrade the protein of interest (POI). PROTACs have enabled

the targeting of many proteins so far considered undruggable by a classical occupancy-based inhibition

mechanism, thereby expanding therapeutic prospects for various pathologies including cancer.

1

Despite PROTACs being an efficient catalytic method to degrade malfunctioning proteins in cancer cells, many

of their targets are also expressed in healthy cells, where they play an essential role in the cells’ functioning.

Hence, on-site side effects are one of the major drawbacks of PROTACs, due to the lack of selectivity between

healthy and cancerous cells.

2

To tackle these issues, this work has focused on developing cancer cell selective and bioresponsive degraders

called SelecPROTACs. These small-molecule prodrugs have been designed to capitalize on some of the key

characteristics of cancer cells to ensure targeted delivery and PROTAC release only upon reaching the cancerous

environment.

3

We have synthesized proof-of-concept probes based on the BRD4 PROTAC MZ1

4

, bearing a cancer-cell

favoring delivery ligand appended through a bioresponsive cleavable linker, to enable cancer cell specific

activation. These inactive prodrugs have sown to release the active PROTAC via the expected mechanism and

have comparable POI degradation ability to the parent PROTAC in cancerous cells. As desired, these prodrugs

show reduced degradation activity in non-cancerous cells, owing to decreased uptake or drug release.

However, a negative control analogue with a non-cleavable linker also maintains some limited biological

activity, leading us to question the stability of the key connection sites and the linker on the molecule. Hence,

future efforts will be focused on modifying linkage mechanism for enhanced stability in biological conditions

and to further improve the spatio-temporal control and selectivity of the release at the site of interest.

References

1) G. M. Burslem and C. M. Crews, Cell, 2020, 181, 102–114.

2) K. Moreau, M. Coen, A. X. Zhang, F. Pachl, M. P. Castaldi, G. Dahl, H. Boyd, C. Scott and P. Newham, Br. J.

Pharmacol., 2020, 177, 1709–1718

3) X. Zhang, X. Li, Q. You and X. Zhang, Eur. J. Med. Chem., 2017, 139, 542–563

4) M. Zengerle, K. H. Chan and A. Ciulli, ACS Chem. Biol., 2015, 10, 1770–1777.

EFMC-ISMC | 389

V002

SYNTHESIS AND BIOLOGICAL EVALUATION OF PSMA

TARGETED FLUORESCENT CONJUGATES

Aleksei Machulkin (1), Radik Shafikov (1), Anastasia Uspenskaya (1), Stanislav Petrov (1), Dmitry

Skvortsov (1), Maxim Abakumov (3), Vladimir Polshakov (5), Yan Ivanenkov (1,2,3,4), Elena

Beloglazkina (1), Alexander Majouga (1,3), Elena Khazanova (6)

1) Lomonosov Moscow State University, Chemistry Dept., Leninskie gory, Building 1/3, GSP-1, Moscow, 119991, RussianFederation.

2) Moscow Institute of Physics and Technology (State University), 9 Institutskiy lane, Dolgoprudny City, Moscow Region,141700, Russian Federation.

3) National University of Science and Technology MISiS, 9 Leninskiy pr, Moscow, 119049, Russian Federation.4) The Federal State Unitary Enterprise Dukhov Automatics Research Institute, Moscow, 127055, Russia

5) Center for Magnetic Tomography and Spectroscopy, Faculty of Fundamental Medicine, M.V. Lomonosov Moscow StateUniversity, 119991, Moscow, Russian Federation.

6) LLC Izvarino-Pharma, v. Vnukovskoe, Vnukovskoe sh., 5th km., building 1, Moscow, 108817, Russian Federation.

Prostate cancer (PC) is the most prevalent malignancy among men and the second leading cause of

cancer-related death. There were 1.3 million new cases of PC worldwide in 2018 and about 192 thousand cases

in the US in 2020.

1

 Radical prostatectomy is the most effective and typical cancer treatment in the primary

therapy of prostate cancer. Deletion of all malignant cells leads to the absence of relapse in 50% of cases and

could extend the overall survival of patients. In the case of prostate cancer, PSMA (prostate-specific membrane

antigen) is one of the prominent targets for the visualization of malignant cells, due to its overexpression in

prostate cancer, in comparison to healthy prostate cells. This makes it a promising target for drug delivery,

prostate cancer diagnostics and intraoperative navigation. Fluorescence-guided surgery makes it possible to

highlight cancerous cells and to guide a surgeon to resect tumor tissue in real-time.

In this work we synthesized a series of 21 new DCL-based PSMA ligands and have evaluated their affinity to

PSMA. The most promising ligand  demonstrated a potent affinity with IC

50

=22 nM far exceeding the standard

Glu-urea-Lys PSMA ligand DCL (IC

50

=547 nM) and 2-PMPA (IC

50

=80 nM) at the same test. The synthesized

ligands demonstrated the absence of any strong toxicity on Hek293T, VA13, MCF7 and A549 cell lines. The

series of three fluorescent dye conjugates PSMA-FAM-5, PSMA-SulfoCy5 and PSMA-SulfoCy7 based on the

most potent ligand were synthesized. In vivo experiment demonstrated the significant accumulation of both

conjugates PSMA-SulfoCy5 and PSMA-SulfoCy7 in PSMA-overexpressing 22Rv1 xenografts 24 hours after a

single intravenous injection. Also, these conjugates demonstrated a significant difference between 22Rv1 and

PC-3 accumulation in in vivo experiment. One of the most significant facts of conjugate PSMA-SulfoCy7 is its

low toxicity. This conjugate demonstrated a 200-times higher maximum tolerated dose than the dose which

allows us to effectively visualize PSMA-bearing tumors. At the same time, the subchronic evaluation did not

show pronounced toxic manifestations of the conjugate. In connection with the data presented, it can be assumed

that the PSMA-SulfoCy7 conjugate can be a good candidate for further investigations as a means of

intraoperative diagnostics and imaging of prostate cancer.

2,3

The reported study was funded by RFBR, project number 20-33-70089.

References

1) Siegel, R. L.; Miller, K. D.; Jemal, A. Cancer statistics, 2020. CA. Cancer J. Clin. 2020, 70, 7–30.

2) Machulkin; Shafikov; Uspenskaya et al. Synthesis and Biological Evaluation of PSMA Ligands with Aromatic Residues

and Fluorescent Conjugates Based on Them. J. Med. Chem. 2021, 64 (8), 4532–4552.

3) Machulkin A.E., Uspenskaya A.A., Ber A.P. et.al. Conjugate of Fluorescent Dye for the Visualization of PSMA

Expressing Cells. Patent WO2021002771A1, Feb 7, 2021.

390 | EFMC-ISMC

V003

SYNTHESIS AND BIOLOGICAL EVALUATION OF CONJUGATE

DRUGS BEARING BILE ACID CARRIERS TO TARGET LIVER

TUMOURS

Alvaro G. Temprano (1,2), Beatriz Sánchez de Blas (1,2), Mattia Mori (3), Francisco Bermejo (4), Marta

R. Romero (1,2,5), Jose J.G. Marin (1,2,5), Concepcion Perez-Melero (2,6,7)

1) Experimental Hepatology and Drug Targeting (HEVEPHARM), University of Salamanca, Spain.2) Institute of Biochemical Research of Salamanca (IBSAL), Salamanca, Spain.

3) Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Italy.4) Organic Chemistry Department, University of Salamanca, Spain.

5) National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Madrid, Spain.6) Pharmaceutical Chemistry Laboratory, Pharmaceutical Sciences Department, University of Salamanca, Spain.

7) Tropical Diseases Research Centre (CIETUS), University of Salamanca, Spain.

Hepatocellular carcinoma (HCC) is the most frequent primary liver cancer, being the second deadliest cancer [1].

Pharmacological treatment of HCC is often impaired by the development of chemoresistance. Several

chemoresistance mechanisms have been reported, including reduced drug uptake by cells and increased drug

efflux, among others [2].

Bile acids (BAs) are endogenous steroids that perform essential functions for the digestion and absorption of

lipids and lipid-soluble vitamins. BAs are located to the liver/gall bladder/intestine system, where they are

continuously released and taken up by specific transporters in a process known as enterohepatic circulation.

Therefore, due to the marked enterohepatic organotropism of these compounds, BAs can be used as carriers to

target drugs towards tissues located in the enterohepatic system [3].

In this work we have designed and synthesized two conjugate compounds, named BATKIs, formed by a BA

carrier (cholic or glycocholic acid) and the first-line drug in the treatment of HCC, i.e. sorafenib (a tyrosine

kinase inhibitor, TKI). The linker contains a 1H-1,2,3-triazole ring, obtained by a 1,3-dipolar cycloaddition

reaction between an azide derived from the BA and a sorafenib analogue bearing an alkyne moiety [4] (Fig. 1).

Fig 1. Structure of the conjugated compounds designed by combination of a BA and sorafenib, a tyrosine kinase

inhibitor used as first-line treatment for HCC.

The antiproliferative activity of the obtained compounds has been evaluated in vitro on several liver tumour cell

lines expressing or not specific liver transporters for organic cations and anions, and BAs. Uptake experiments

revealed that OATP1B3, a BA transporter, efficiently transports BATKIs. Moreover, BATKIs displayed similar

antiproliferative activity to sorafenib. These results were further supported by molecular modelling studies,

showing that the target compounds are substrates of OATP1B3 and interact with the active site of tyrosine kinase

receptors (TKs).

Acknowlegdements: AGT thanks University of Salamanca/Santander for a predoctoral grant. The authors thank

Cesar Raposo (Mass spectrometry service, University of Salamanca) and Anna Lithgow (NMR service,

University of Salamanca).

References

1) Ferlay, J; Colombet, M; Soerjomataram, I; Parkin, DM et.al. F. Int. J. Cancer 2021, 149, 778.

2) Marin, JJG; Macias, RIR; Monte, MJ; Romero, MR et al. Cancers 2020, 12, 1663.

3) Kramer, W; Wess, G. Eur. J. Clin. Invest. 1996, 26, 715.

4) Rostovtsev, VV; Green, LG; Fokin, VV; Sharpless, KB. Angew. Chem. Int. Ed. 2002, 41, 2596.

EFMC-ISMC | 391

V004

ENCAPSULATION OF CHARGED CHOLINESTERASE

REACTIVATORS USING APOFERRITIN MAY PROLONG THEIR

HALFLIFE AND ENHANCE BIOAVAILABILITY IN CNS

David Malinak (1), Eliska Prchalova (1), Simona Rex (2), Martina Sukupova (2), Rudolf Andrys (1),

Adam Skarka (1), Zbynek Heger (2), Kamil Musilek (1)

1) University of Hradec Kralove, Faculty of Science, Department of Chemistry, Rokitanskeho 62, 500 03 Hradec Kralove,Czech Republic

2) Mendel University in Brno, Department of Chemistry and Biochemistry, Zemedelska 1, 613 00 Brno, Czech Republic

The cholinesterase reactivators (so called oximes) are used as causal antidotes for organophosphorus reactivators

(i.e. nerve agents, insecticides) [1]. They are able to split organophosphorus moiety from cholinesterase active

site using nucleophilic oximate anion. They are usually permanently charged molecules that are applied in high

doses (i.e. 300-1000 mg) via i.m. administration in an autoinjector. Thus, such oximes have to possess high

water solubility. However, such charged molecules have rapid body elimination and minimal CNS

bioavailability, and thus minimal reactivation of brain acetylcholinesterase. For this reason, there is high demand

to develop novel molecular entities for cholinesterase reactivation that would be able to enter CNS. Although

there were increasing attempts to develop uncharged cholinesterase reactivators that could be CNS penetrable,

the effectiveness and solubility of uncharged oximes or other nucleophiles are so far unsatisfactory [1].

Our group has developed very efficient charged cholinesterase reactivators (e.g. K027, K203, K868) that were

proved to reactivate cholinesterases both in vitro and in vivo [2]. But their CNS penetrability was found very

low. For this reason, the various apoferritins were introduced for preparation of nanocarriers that could be able to

deliver charged oximes/molecules to CNS [3]. For this purpose, particular oxime reactivators were tagged with

fluorescent BODIPY-like dye for their easier scavenging inside the nanocarriers and tissues. The nanocarriers

containing oxime or oxime-BODIPY conjugates were successfully prepared and characterized. The long-term

stability of loaded nanocarriers was confirmed and pH dependent oxime release was determined with very

promising results. The animal experiments are in progress.

This work was supported by the Czech Science Foundation (project no. 19-13628S), University of Hradec

Kralove (project no. VT2019-2021) and CEITEC2020 LQ1601.

References

1) Musilek, K. et. al In: Gupta, R.C. (ed). Handbook of Toxicology of Chemical Warfare Agents. 3rd Ed, 2020, p.

1161-1177.

2) Zorbaz, T. et al. J. Med. Chem. 2018, vol. 61, no. 23, p. 10753−10766.

3) Tesarova, B. et al. J. Control. Release. 2020, vol. 325, no. 1, p. 176-190.

392 | EFMC-ISMC

NOTES

EFMC-ISMC | 393

Posters - Diversified topicsW. Emerging Topics

394 | EFMC-ISMC

W001

INHIBITION OF MATRIX METALLOPROTEINASES TO FIGHT

ALZHEIMER’S DISEASE

Pauline Zipfel (1), Julien Lalut (1), Camille Denis (1), Ronan Bureau (1), Peggy Suzanne (1), Audrey Davis

(1), Aurelie Malzert-Freon (1), Kevin Baranger (2), Michel Khrestchatisky (2), Santiago Rivera (2),

Christophe Rochais (1), Patrick Dallemagne (1)

1) Centre d’Etudes et de Recherche sur le Medicament de Normandie (CERMN), Normandie Univ, UNICAEN, 14000 Caen,France

2) Institute of Neuropathophysiology (INP), UMR7051, CNRS, Aix Marseille Universite, France

Alzheimer's disease (AD), the most common form of dementia (

~

70%), is a neurodegenerative and incurable

brain disorder, for which only 4 symptomatic treatments are available at this time. In 2018, the number of people

living with dementia in the world was estimated at 50 million and this number will likely more than triple by

2050.

(a)

Because of the heavy economic and societal impacts, there is an urgent need to find new treatments that

target the molecular causes of the neurodegenerative process, but these remain elusive in 2020.

Indeed, abnormal aggregates of beta-amyloid peptide (Aβ), that result from the proteolytic processing of amyloid

precursor protein (APP), have long been considered one of the main pathological features of AD. They are better

known as extracellular amyloid plaques (also known as senile plaques). However, the generally accepted vision

of APP metabolism has been recognized as simplistic in recent years. In this context, matrix metalloproteinases

(MMPs) have been highlighted in the literature as potential new relevant biological targets in AD. Indeed,

independent works from two teams have shown that MT5-MMP, a membrane-type MMP, is able to process APP

through its η-secretase activity and leads to the generation of new fragments, including Aη-α which has

neurotoxic effects in vitro and in vivo.

(b),(c),(d)

  

Our innovative project aims therefore to design first inhibitors of MT5-MMP. However, the design of selective

inhibitors of MMPs remains quite challenging, considering the high structural homology MMPs share.

Therefore, we are exploring different drug design strategies in the search for first-in-class small molecule

inhibitors of MT5-MMP. For a start, we focused on an ambitious in silico approach, starting with the virtual

screening of the chemical library of our laboratory (>19,000 molecules) on a homology model of MT5-MMP. At

the moment, we are investigating a second “ligand-based drug design” approach, based on the knowledge gained

for other members of the MMP family, notably regarding the structural requirements of this enzyme inhibition.

For the first time, our experimental results concerning in silico approach and chemical synthesis will be

disclosed in this presentation.

 

References

(a) Patterson, C. World Alzheimer Report 2018. London Alzheimer’s Dis. Int. 2018.

(b) Baranger, K. et al., Cell. Mol. Life Sci. 2016, 73(1), 217–236.

(c) Willem, M. et al., Nature 2015, 526(7573), 443–447.

(d) Baranger, K. et al., Front. Mol. Neurosci. 2017, 9 (1), 1–17.

EFMC-ISMC | 395

W002

DEVELOPMENT OF SMALL MOLECULE ALLOSTERIC BETA 2

ADRENOCEPTOR MODULATORS

Stephanie Tiedt, Harald Hübner, Peter Gmeiner

Department of Chemistry and Pharmacy, Medicinal Chemistry,Friedrich Alexander University, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany

Bronchoconstriction is a major symptom in diseases like COPD and bronchial asthma, which can be effectively

treated with selective β

2

AR agonists, like salbutamol or the long-acting agent salmeterol.[1,2] The β

2

AR belongs

to the class of G protein-coupled receptors. A high-resolution crystal structure of the β

2

AR bound to the partial

agonist salmeterol reveals that the saligenin ethanolamine moiety binds to the orthosteric binding pocket,

whereas the hydrophobic aryloxyalkyl appendage interacts with an allosteric site of the receptor, which is

topographically distinct from the orthosteric site. Interestingly, a similar but not identical allosteric pocket was

identified in the β

1

AR.[3] Thus, addressing this site can lead to receptor subtype selectivity, which displays a

major advantage of allosteric modulators, as allosteric binding pockets are typically less conserved in their

sequence than orthosteric sites.[4]

We developed small molecule allosteric modulators targeting the exosite of salmeterol. Taking advantage of the

β

2

AR-salmeterol crystal structure [3], novel ligands were designed in an iterative docking approach by formally

removing the orthosteric moiety of the salmeterol template and modifying the aryloxyalkyl appendage. Hence,

we designed compounds bearing a variety of heterocycles connected to differently substituted benzyl ethers by

an alkyl linker. Various derivatives were synthesized and their allosteric effects were investigated in functional

assays at the β

2

AR and its congener β

1

AR.

References

1) Procopiou, P. A. et al., J. Med. Chem., 2014, 57, 159.

2) Moore, R.H. et al., Cell Mol. Biol., 2007, 36, 254.

3) Masureel, M. et al., Nature Chemical Biology, 2018, 14, 1059.

4) Conn, P.J. et al., Nature Reviews. Drug Discovery, 2009, 8, 41.

396 | EFMC-ISMC

W003

NOVEL METHOD TO IMPROVE API'S STABILITY IN

MICROGRAVITY ENVIRONMENT: CASE STUDY FOR COVID-19

DRUG FORMULATION IN SPACE

Gergo Mezohegyi (1), Balazs Buchholcz (1), Istvan Puskas (2), Lajos Szente (2), Tamas Sohajda (2),

Ferenc Darvas (1)

1) InnoStudio Inc., Záhony u. 7, Budapest 1031, Hungary2) CycloLab Cyclodextrin R&D Laboratory Ltd., Illatos út 7, Budapest 1097, Hungary

Research for drug stability has expanded over recent years to include the effects on stability of storage and

transport, repackaging and importantly the potential clinical outcomes for patients who may be exposed to toxic

effects of drug degradants. Drug stability is also critical in drug regulation and quality as regulators focus their

attention on counterfeit medicines. Here we first time account on our experiments how to use microgravity to

improve the stability of an important pharmaceutical ingredient (pharmaceutical active principle, API).

Veklury®, a remdesivir-based (REM) medicine produced by Gilead Sciences is one of the most widely used

drug for treatment of COVID-19 infections worldwide. Its active component, to be applicable for intravenous

infusion, requires solubilization by a cyclodextrin derivative called SBECD, which contains also sodium ions.

Unfortunately, the associated high sodium content of the REM-SBECD complex hinders the treatment of

COVID-19 patients with kidney and other problems.

We have been investigating the effect of microgravity on the complex formation of REM and SBECD, that

would potentially allow to tailor the formulation process in order to reach elevated drug efficiency for

COVID-19 treatment and/or a product with improved characteristics such as potentially elevated stability of the

active compound.

The experiments in space were carried out in Dec 2020 – Jan 2021, in ESA’s Columbus laboratory on the

International Space Station (ISS). Characterization of returned and reference samples on ground included a

comprehensive analysis of the samples separated to solid and liquid phases, which involved investigations by

optical microscopy, scanning electron microscopy, X-ray powder diffraction, Raman spectroscopy and HPLC.

Morphological investigations showed that remdesivir crystals in samples staying on ISS consisted of particles

with significantly smaller size, compared to REM crystals formed in reference samples (on ground). The

highlighted result of the study is that in the space sample formulated at neutral pH, significantly less degradation

rate of REM in the REM-SBECD complex was observed, compared to that of the ground sample. The results

suggest that the complex formation, perhaps also REM degradation took place via different chemical mechanism

in space and on Earth. These findings may induce a very new approach to both design of drug molecules with

improved stability and medicinal discovery studies in space.

As far as we know, the present experiment was the very first ocassion when anti-COVID-19 research efforts

were brought to space. Moreover, this is the first time when microgravity (which is in general known to diminish

drug stability) was used to increase stability of a small molecule API.

EFMC-ISMC | 397

Posters - Diversified topicsX. First time disclosures

398 | EFMC-ISMC

X001

TOWARDS NEXT GENERATION COMPLEMENT C3 INHIBITORS

WITH PROLONGED TARGET RESIDENCE TIME ? LATEST

INSIGHTS IN STRUCTURE-ACTIVITY RELATIONSHIP AND

PHARMAKOKINETIC STUDIES

Christina Lamers (1), Beatrice Wagner (1), Sarah Hughes (2), Merita Rumano (2), Martin Smiesko (3),

Piet Gros (4), John D. Lambris (2), Daniel Ricklin (1)

1) Molecular Pharmacy, Department of Pharmaceutical Sciences, University Basel, Basel, Switzerland2) Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania,

Philadelphia, USA3) Computational Pharmacy, Department of Pharmaceutical Sciences, University Basel, Basel, Switzerland

4) Crystal and Structural Chemistry, Department of Chemistry, Utrecht University, Utrecht, The Netherlands

The complement system as “first line of defence” gets activated upon injurious stimuli and invaders and leads to

pathogen clearance and opsonic cell killing. Yet complement may be inadvertently triggered on human cells or

biomaterial surfaces, thereby contributing to clinical complications in the pathogenesis of various autoimmune,

inflammatory and age-related diseases as well as transplant rejection. The involvement of dysregulated

complement activation in inflammatory and autoimmune diseases is now widely recognized[1], therefore, it has

gained increasing interest as potential target for intervention. So far, only C5-targeting drugs have reached the

market, while inhibitors of FB, FD and C3, amongst other targets, are in clinical evaluation[2]. C3 inhibitors are

of high interest as they have the potential to broadly prevent complement activation in acute-phase and other

clinical conditions.

In this presentation, we reflect on the development of Cp40, a picomolar derivative of the compstatin peptide.

Originally identified by phage display[3] the C3 inhibitor compstatin could be improved from micro- to

picomolar affinity. Based on a novel co-crystal structure of Cp40 in complex with its complement target C3b, we

used site-specific modifications/deletions, nonproteinogenic amino acids and tailor-made building blocks to

elucidate the SAR in detail and identify key interaction determinants.

In particular, the presented study discloses for the first time recent structure-activity relationship investigations,

which led to the identification of a next generation derivative with prolonged target residence time and increased

binding affinities. In addition to the in-depth study of binding kinetics and thermodynamics, we here present

pharmacokinetic data of this next generation compstatin derivative.

References

1) Ricklin D, et al.: J Immunol. 2013; 190: 3831-8.

2) Ricklin D, et al.: Nature Rev Neph 2017; 14: 26-47.

3) Sahu et al.: J Immunol. 1996; 157: 884-91.

EFMC-ISMC | 399

X002

STRUCTURAL OPTIMIZATION OF LONG-CHAIN VITAMIN E

METABOLITES YIELDS DERIVATIVES THAT INDUCE A LIPID

MEDIATOR CLASS SWITCH FROM INFLAMMATION TO

RESOLUTION

Stephan Permann (1), Konstantin Neukirch (1), Jean-Jacques Helesbeux (2), Guillaume Viault (2),

Chau-Phi Dinh (2), Elena Brunner (3), Oliver Werz (3), Denis Seraphin (2), Pascal Richomme (2),

Andreas Koeberle (1)

1) Michael Popp Institut, University of Innsbruck, 6020 Innsbruck, Austria2) Pharmacy & Health Sciences SONAS, University Angers, 49045 Angers, France

3) Pharmaceutical/ Medical Chemistry, Friedrich-Schiller-University Jena, 07747 Jena, Germany

Inflammation is a complex process driven by innate immune cells that produce pro-inflammatory lipid mediators

during acute phase and specialized pro-resolving lipid mediators (SPM) in the resolution phase. Macrophages of

the M2 subtype play a major role in inflammation resolution as they strongly express ALOX15 and ALOX15B

and effectively release SPM, including resolvins, protectins and maresins. This lipid mediator class switch

terminates inflammatory processes and restores tissue homeostasis. Current anti-inflammatory therapy largely

targets pro-inflammatory cascades, without considering resolution. We have recently shown that endogenous

metabolites of vitamin E cumulate in innate immune cells at inflammatory sites, limit inflammation by targeting

5-lipoxygenase (5-LOX) and raise systemic resolvin levels. Starting from an in-house library of 150 isolated and

semi-synthesized vitamin derivatives, we searched for structurally optimized compounds that combine potent

inhibition of 5-LOX with enhanced SPM biosynthesis. Lipid mediator profiles of activated and non-activated

macrophages (M1 and M2), peripheral blood mononuclear cells and neutrophils were assessed. We identified

specific ω-oxidized tocotrienol derivatives that selectively trigger the biosynthesis of protectins among SPM

while maintaining the 5-LOX inhibitory activity. These lead compounds are of high interest as tools for studying

protectin function and open the door to a novel class of anti-inflammatory/pro-resolving drug candidates that

induce a switch from leukotrienes to protectins.

400 | EFMC-ISMC

X003

POSITIVE ALLOSTERIC MODULATORS OF GLUN2B-CONTAINING

NMDARS AS ENHANCERS OF SYNAPTIC PLASTICITY

Carlos M. Martínez-Viturro (1), José M. Cid (1), Josep Llaveria (1), Jesús Alcazar (1), Ann Vos (2),

Murali Subramanian (2), Hilde Lavreysen (2), Ineke Fonteyn (2), Ilse Van Der Linden (2), Juan D.

Pita-Almenar (2)

1) Janssen Research & Development, Division of Janssen-Cilag S.A., Jarama 75A, Toledo 45007, Spain2) Janssen Research & Development, Janssen Pharmaceutica N.V., Turnhoutseweg 30, Beerse B-2340, Belgium

Synaptic plasticity is the ability of synapses to strengthen or weaken over time, in response to increases or

decreases in their activity. This phenomenon is proposed to be the basis for learning and memory of neuronal

networks and has the potential to recover from synaptic loss. Substantial evidence exists correlating synaptic

dysfunction/loss with degree of cognitive impairment and late stages of Alzheimer's Disease (AD) for which

synaptic plasticity represents a potential therapeutic approach.

NMDA receptors are glutamatergic receptors that can be activated precisely during the coincidence of excitatory

synaptic transmission with neuronal activity. This precise activation of the NMDA receptor is key for synaptic

plasticity and remodeling neuronal circuits. Each NMDA receptor contains different subunits: two NR1 and two

NR2 divided into four subtypes A-D. The NR2B subunit caught our attention.  Its expression is developmentally

regulated: a switch in NR2B-containing receptors by NR2A-containing receptors is seen after development

which is associated with the closure of the “critical period of synaptic plasticity during brain development”.

NR2B re-expression can restore critical period-like plasticity in adult sensory cortex, thus promoting learning

and memory. Our aim is to identify PAMs of NR2B function to enhance synaptic plasticity and improve learning

and memory in neuronal circuits with synaptic loss such as aged/AD brains.

 

Here we report the structure activity relationship of a series of NR2B PAMs pyridopyrazoles originated from a

HTS campaign. Further characterization of advanced compounds revealed their potential as in vivo tools to

understand the role of NR2B in synaptic plasticity and their potential application in neurodegenerative diseases.

EFMC-ISMC | 401

X004

DEVELOPMENT OF THE FIRST DUAL ACTIVATORS OF

NEUROTROPHIN AND SEROTONIN 5-HT4 RECEPTORS, A

PROMISING STRATEGY IN THE BATTLE AGAINST

NEURODEGENERATIVE DISORDERS

Mirjana Antonijevic (1), Despoina Charou (2,3), Thanasis Rogdakis (2,3), Marianna Papadopoulou (2,3),

Achille Gravanis (2,3), Ioannis Charalampopoulos (2,3), Patrick Dallemagne (1), Christophe Rochais (1)

1) Normandie Univ., UNICAEN, CERMN, 14000 Caen, France2) Department of Pharmacology, Medical School, University of Crete, Heraklion, Greece

3) Institute of Molecular Biology & Biotechnology, Foundation of Research & Technology-Hellas (IMBB-FORTH),Heraklion, Greece

Numerous studies have been published about the implication of the neurotrophin tyrosine kinase receptor - TrkB

in the pathogenesis of several neurodegenerative conditions

[1]

. Brain-derived neurotrophic factor (BDNF) and

neurotrophin-4/5 (NT-4/5) activate the TrkB receptor with high potency and specificity, promoting neuronal

survival, differentiation and synaptic function. On the other side, activation of the p75 neurotrophin receptor (a

member of the tumour necrosis factor receptor family) can activate several signalling cascades. The TRAF6

(TNF Receptor Associated Factor 6) cascade which is inducing cell death and the RIP2 (receptor-interacting

protein 2) cascade that propagates cell survival

[2]

. Based on all these findings we developed two strategies in

order to design and synthesize small molecules, able to prevent neuronal death and to increase

neuroregeneration. The first strategy was to use the main structural characteristics of LM22A-4

[3]

, a known

activator of the TrkB receptor, and modify it in order to obtain the compounds which will be not only ligands for

TrkB receptor but also act as partial 5-HT

4

receptor agonists. There are evidences that the partial 5-HT

4

receptor

agonist (RS67333) can increase the concentration of BDNF

[4]

. The second strategy was to modify LM11A-31

[5]

,

a molecule that is able to activate a specific cascade of the p75 neurotrophin receptor, inducing cell survival, and

to merge it with TrkB activator, LM22A-4. As a result of our study, we have developed two new datasets of

small molecules, potential TrkB/5-HT

4

receptors ligands and TrkB/p75 receptors ligands, which will be used for

further biological research and hit to lead optimisation studies.

This project has received funding from the European Union's Horizon 2020 framework programme for research

and innovation under grant agreement No 765704.

References

1) a) Simmons, D. A., et al., The Journal of neuroscience, 2013, 33(48), 18712-27; b) Nomura, T., et al., The Journal of

Neuroscience, 2017, 37(47), 11298–11310.; c) Zheng, H., Niu, S., Zhao, H. et al., Metab. Brain Dis., 2018, 33, 1961.;

2) a) Meldolesi, J., et al., Pharmacological Research, 2017, 121, 129–137.; b) Kisiswa, L., Ferna, D., Sergaki, M.C., Ibanez,

C.F., Cell Survival, 2018, 1013–24.;

3) Massa, S. M., et al., J Clin Invest, 2010, 120(5), 1774–1785.;

4) Pascual-Brazo, J., et al., International Journal of Neuropsychopharmacology, 2012, 15(5), 631–643.;

5) Shi, J., Longo, F.M., Massa, S.M., Stem Cells, 2013, 31(11):2561–74.

402 | EFMC-ISMC

NOTES

EFMC-ISMC | 403

Posters - Diversified topicsZ. Other

404 | EFMC-ISMC

Z001

COVALENT INHIBITION OF BACTERIAL UREASE BY

BIFUNCTIONAL CATECHOL-BASED PHOSPHONATES AND

PHOSPHINATES

Aikaterini Pagoni (1), Lukasz Berlicki (2), Stamatia Vassiliou (1)

1) National and Kapodistrian University of Athens, Department of Chemistry, Laboratory of Organic Chemistry, Athens,Greece

2) Department of Bioorganic Chemistry, Faculty of Chemistry, Wrocław University of Science and Technology, WybrzeżeWyspiańskiego 27, 50-370 Wrocław, Poland

Urease activity has been identified as a primary factor in the colonization and development of persistent

infections of several microbial species. The priority pathogen list indicated by the World Health Organization for

the research and development of new antibiotics includes urease-dependent antibiotic-resistant bacteria, several

of which are involved in bacterial infections of the respiratory apparatus, and it is remarkable that half of patients

who died of the recent COVID-19 epidemics in Wuhan (China) became co-infected with bacteria in the lungs

and also required antibiotics.

1

In this study, a new class of bifunctional inhibitors of bacterial ureases, important molecular targets for

antimicrobial therapies, was developed. The structures of the inhibitors consist of a combination of a

phosphonate or (2-carboxyethyl)phosphinate functionality with a catechol-based fragment, both of which are

located on the short propionate scaffold. The β-substituting phosphorus-based moieties were dedicated to

complexation of the catalytic nickel ions, while catechol was used to provide simultaneous covalent bonding

with the thiol group of Cys322(Figure 1).

The author thanks Special Account for Research Grants and National and Kapodistrian University of Athens forfunding to attend the meeting.

 

References

1) L. Mazzei, F. Musiani, S. Ciurli, Journal of Biological Inorganic Chemistry, 2020,

https://doi.org/10.1007/s00775-020-01808-w

EFMC-ISMC | 405

Z002

SYNTHESIS OF NOVEL CINNAMIC ACID DERIVATIVES AS

POSSIBLE ANTI-ALZHEIMER AGENTS

Niki Gouleni (1), Antonio diStefano (2), Ivana Cacciatore (2), Stamatia Vassiliou (1)

1) National and Kapodistrian University of Athens, Department of Chemistry, Laboratory of Organic Chemistry, 15771,Athens, Greece

2) Department of Pharmacy, “G. D'Annunzio” University of Chieti-Pescara, Chieti Scalo, CH 66100, Italy

Cinnamic acid and its derivatives display a broad spectrum of biological activitiesand they have been studied as

anti-AD agents due to their anti-neuroinflammatory properties and their ability to inhibit Aβ aggregation by

scavenging oxidants.

1

Considering the poor drugability of carboxylic acids and the limited ability of cinnamic acids to cross the

blood-brain barrier, we intended to replace the carboxyl group with other structures. Therefore, the synthesis of

cinnamic acid derivatives containing sulfone and thiazole portion was devised. Additionally, Ugi 4-component

adducts using cinnamic acids were elaborated (Figure 1).

The author thanks Special Account for Research Grants and National and Kapodistrian University of Athens forfunding to attend the meeting.

References

1) N. Ruwizhi and B. A. Aderibigbe , Cinnamic Acid Derivatives and Their Biological Efficacy, Int. J. Mol. Sci. 2020, 21,

5712-5746

406 | EFMC-ISMC

Z003

SYNTHESIS AND ANTIOXIDATIVE ACTIVITY OF NOVEL

ACRYLONITRILE IMIDAZO[4,5-b]PYRIDINES

Ida Boček (1), Kristina Starčević (2), Lucija Hok (3), Robert Vianello (3), Ivana Novak Jovanović (4),

Marijana Hranjec (1)

1) Department of Organic Chemistry, Faculty of Chemical Engineering and Technology, University of Zagreb, Marulićev trg19, HR-10000 Zagreb, Croatia

2) Department of Chemistry and Biochemistry, Faculty of Veterinary Medicine, University of Zagreb, Heinzelova 55,HR-10000 Zagreb, Croatia

3) Division of Organic Chemistry and Biochemistry, Ruđer Bošković Institute, Bijenička cesta 54, HR-10000 Zagreb, Croatia4) Institute for Medical Research and Occupational Health, Ksaverska c. 2,

HR-10000 Zagreb, Croatia

Imidazo[4,5-b]pyridines, as a structural analogues of naturally occurring purines, form similar interactions with

important biomacromolecules. Their derivatives posses a wide range of biological activities and some have

already found application in medicine, while many others are currently in clinical testing. Imidazo-pyridines

showed inhibitory activity against tumor cells and bacteria resistant to common chemotherapeutics, antioxidative

activity etc.

This work presents the synthesis and structural characterization of novel N-substituted acrylonitrile derived

imidazo[4,5-b]pyridines. For the synthesis of novel targeted compounds, classical organic synthesis reactions as

well as microwave assisted synthesis were used. Main precursors for the synthesis of targeted derivatives were

prepared by the uncatalyzed microwave assisted amination starting from corresponding halogeno-substituted

precursors followed by the reduction of nitro moiety to amino. 2-cyanomethlyimidazo[4,5-b]pyridines were

obtained in the cyclocondensation reaction with ethyl cyanoacetate, while targeted acrylonitriles were prepared

in condensation reaction with chosen aromatic aldehydes.

1

The structures of newly prepared compounds were

confirmed by means of 1H and 13C NMR spectroscopy as well as MS spectrometry.

All newly prepared compounds will be tested for their antioxidative activity in vitro by using several

spectroscopic (DPPH, ABTS and FRAP) as well as electrochemical methods. Additionally, the measured

antioxidative capacities will be rationalized by computational analysis.

2

Specifically, we will analyze the

influence of the type and the number of the substituents on the imidazo[4,5-b]pyridine scaffold in order to

choose lead compound(s), which will be further optimized as promising antioxidants.

 

Fig. 1: The structures of newly prepared derivatives

References

1) N. Perin, L. Hok, A. Beč, L. Persoons, E. Vanstreels, D. Daelemans, R. Vianello, M. Hranjec, Eur. J. Med. Chem. 211

(2021) 113003, 14

2) M. Cindrić, I. Sović, M. Mioč, L. Hok, I. Boček, P. Roškarić, K. Butković, I. Martin-Kleiner, K. Starčević, R. Vianello,

Antioxidants 8 (2019) 10, 477, 22

EFMC-ISMC | 407

Z004

SYNTHESIS AND BIOLOGICAL ACTIVITY OF NOVEL N-

SUBSTITUTED BENZIMIDAZOLE DERIVED ACRYLONITRILES

A. Beč (1), L. Hok (2), R. Vianello (2), L. Persoons (3), D. Daelemans (3), M. Hranjec (1)

1) Department of Organic Chemistry, Faculty of Chemical Engineering and Technology, University of Zagreb,Marulićev trg20, HR-10000 Zagreb, Croatia

2) Division of Organic Chemistry and Biochemistry, Ruđer Bošković Institute, Bijenička cesta 54, HR-10000 Zagreb, Croatia3) KU Leuven Department of Microbiology and Immunology, Laboratory of Virology and Chemotherapy, Rega Institute,

Leuven, Belgium

The study of heterocyclic chemistry is a vast and expanding area of chemistry because of obvious application of

compounds derived from heterocyclic rings in pharmacy, medicine and other fields. Among the various classes

of nitrogen‐containing heterocyclic compounds, benzimidazole is an important pharmacophore and a privileged

structure in medicinal chemistry. Nowadays, it is a moiety of choice which possesses many pharmacological

properties. Due to their special structural features and electron-rich environment, benzimidazole containing

drugs bind to a variety of therapeutic targets, thereby exhibiting a broad spectrum of bioactivities.

1

In this paper, we report the synthesis of novel biologically active N-substituted benzimidazole derivatives,

related to 2,3-disubstituted acrylonitriles. Novel compounds were prepared by classical reactions of organic

chemistry and by microwave assisted reactions. Acrylonitrile derivatives bearing benzimidazole nuclei were

prepared by aldol condensation of N-substituted 2-cyanomethylbenzimidazoles with corresponding hydroxy,

methoxy, 4-N,N-dimethyl-amino and 4-N,N-diethyl-amino subsituted benzaldehydes in moderate to good yields.

2

All newly prepared compounds will be screened for their antioxidant capacity using several spectroscopic

methods as well as antiproliferative activity on several human cancer cells in vitro.

 

Fig. 1: The structures of newly prepared acrylonitrile derivatives

 

References

1) M. Hranjec, G. Pavlović, M. Marjanović, G. Karminski-Zamola, Eur. J. Med. Chem. 45 (2010) 2405-2417.

2) N. Perin, L. Uzelac, I. Piantanida, G. Karminski-Zamola, M. Kralj, M. Hranjec, Bioorg. Med. Chem. 19 (2011)

6329-6339.

408 | EFMC-ISMC

Z005

COMPUTATIONAL CLUSTERING METHODS IN CHEMICAL

LIBRARIES: DEVELOPMENT OF A PROTEIN-PROTEIN

INTERACTION INHIBITOR FOCUSED LIBRARY

Bruno Di Geronimo, Marina Moreno, Javier Klett

Experimental Therapeutics Programme, Spanish National Cancer Research Centre (CNIO), Calle de Melchor FernándezAlmagro, 3, 28029 Madrid, Spain.

[email protected]

Given a growing interest in expanding chemical space, there are several chemical libraries containing hundreds

to billions of different molecules that remain inaccessible from the experimental point of view, as

High-Throughput Screening (HTS) approaches, and from the in-silico calculations, as Virtual Screening (VS)[1].

For these reason, classifying chemical libraries and obtaining a reduced representative sets of ligands by the use

of clustering methods can be an attractive tool to reduce the chemical space without losing chemical diversity

[2]

.

The aim of this project was to develop a fast, automatic and reliable computational tool for ligand clustering,

providing a representative set of molecules from a chemical library. We focused our approach to obtain a diverse

set of protein-protein interaction inhibitors (2P2I) by filtering a commercially available library using the

‘Rule-of-four’ (MW > 400 Da, aLogP > 4, HBA > 4, rings > 4) that characterize the already known 2P2I

inhibitors

[3]

. After that, we translated the chemical information into molecular descriptors considering 2D

fingerprints and 3D Electroshape descriptor. Then we computed the similarity between the filtered molecules

obtaining a similarity matrix. The similarity measure for the 2D fingerprints was the Tanimoto Coefficient, and

similarity between 3D Electroshape descriptors was computed using the Manhattan distance combined with a in

house approach considering the possible conformations of the molecule

 [4]

. Then we used the 2D and 3D

similarity matrices to perform different classifications using hierarchical clustering. Different methods were used

to perform the linkage analysis of the dataset (single, centroid, median and Ward), and clusters were retrieved

based on several criteria (inconsistent coefficient, cophenetic distance and a maximum number of clusters

allowed). Finally, we selected a set of representative molecules from the centroids of each cluster. Overall, 3D

classification led to better results than 2D, since it accounted better for the differences among molecules.

Moreover, the most efficient method for linkage analysis was centroid. This method provided different

classifications of representative compounds based on our data analysis protocol, by using different clustering

techniques comparing both physicochemical and conformational properties of organic compounds and allow us

to obtain a representative 2P2I library ranging from hundred to thousand compounds.

References

1) Fink, T.; Bruggesser, H.; Reymond, J.-L. Virtual Exploration of the Small-Molecule Chemical Universe below 160

Daltons. Angew. Chemie Int. Ed. 2005, 44 (10), 1504–1508.

2) Sander, T.; Freyss, J.; von Korff, M.; Rufener, C. DataWarrior: An Open-Source Program For Chemistry Aware Data

Visualization And Analysis. J. Chem. Inf. Model. 2015, 55 (2), 460–473.

3) Basse, M. J.; Betzi, S.; Bourgeas, R.; Bouzidi, S.; Chetrit, B.; Hamon, V.; Morelli, X.; Roche, P. 2P2Idb: a structural

database dedicated to orthosteric modulation of protein-protein interactions. Nucleic Acids Res. 2013, 41 (Database issue),

824–827.

4) Rácz, A.; Bajusz, D.; Héberger, K. Life beyond the Tanimoto coefficient: similarity measures for interaction fingerprints.

J. Cheminform. 2018, 10 (48), 1–12.

EFMC-ISMC | 409

Z006

SEROTONIN AND IMIDAZOLINE RECEPTOR LIGANDS, AND

RELATED COMPOUNDS - MODELING OF MIXED-MODE

INTERMOLECULAR INTERACTIONS IN CHROMATOGRAPHIC

SYSTEM

Darija Obradovic (1), Lukasz Komsta (2), Teresa Kowalska (3), Danica Agbaba (4)

1) Department of Pharmaceutical Chemistry, University of Belgrade-Faculty of Pharmacy, Serbia2) Chair and Department of Medicinal Chemistry, Faculty of Pharmacy, Medical University of Lublin, Poland

3) Institute of Chemistry, University of Silesia, Katowice, Poland4) Department of Pharmaceutical Chemistry, University of Belgrade-Faculty of Pharmacy, Serbia

In complex biological systems, heterogenic chemical interactions exist which facilitate passage of

pharmaceutical substances through biological membranes and their further interactions with the receptor sites.

Apart from interactions governed by lipophilicity of the molecules involved, therapeutic effects depend on the

ionic, dipol-dipol, van der Waals and other intermolecular interactions. Consequently, physico-chemical nature

of biological systems makes them multimodal and a real challenge for a more detailed examination. The

mixed-mode chromatographic systems can be used as the in vitro models for preliminary screening in the drug

development process. In such systems, numerous physicochemical interactions of hydrophobic and hydrophilic

type occur, which simulate pharmacokinetics of drugs in a live environment (1,2). Moreover, it is necessary to

develop proper chromatographic methods of testing complex pharmaceutical mixtures, to remain in conformity

with regulatory requirements in analytical control of drugs.

Chromatographic behaviour of the serotonin and imidazoline receptor ligands (and of the structurally related

compounds) was tested under the bimodal HILIC/RP conditions. The mixed-mode diol stationary phase and

acetonitrile + 20mM ammonium acetate mobile phase were used as the test system. Retention of the analytes

was examined in a wide range of the mobile phase compositions (where φ: volume fraction of the aqueous

component). The Box-Cox transformation was used to obtain best description of a bimodal retention, log k = f (φ). Optimal dependences were found for the following optimization parameter (λ) values: λ=-1, λ=-0.5, and λ= 1.

To find considerable proposals for the new equations, we examined the whole space for the following three

cases: log k  = β0

+ β’B(φ, λ1

) + β’’B(φ, λ2

), log k  = β0

+ β’B(φ, λ1

) + β’’B(φ+1, λ2

) and log k  = β0

+ β’B(φ, λ1

)

+ β’’B(φ-1, λ2

). With use of the Box-Cox transformations, seven formulas were selected and the unscaled PCA

approach confirmed that the newly developed formulas show satisfactory description of multimodal retention.

Summing up, it was demonstrated that the important physicochemical and pharmacokinetic properties of the

analytes (ionization, lipophilicity, aromacity, spherocirty) strongly influence the retention process in a

mixed-mode interactions system.

References

1) J. Vucicevic, M. Popovic, K. Nikolic, S. Filipic, D. Obradovic, D. Agbaba. Use of biopartitioning micellar

chromatography and RP-HPLC for the determination of blood–brain barrier penetration of α-adrenergic/imidazoline receptor

ligands, and QSPR analysis. SAR QSAR Environ Res. 2017 (28) 235-252.

2) M.S. Mohamed, S. Filipic, K. Nikolic, D. Agbaba. Estimation of lipophilicity and retention behaviour of some alpha

adrenergic and imidazoline receptor ligands using RP-TLC. J. Liq. Chromatogr. Rel. Technol. 2014 (37) 2829-2845.

410 | EFMC-ISMC

Z007

DESIGN, SYNTHESIS AND EVALUATION OF A PEPTIDE

CONJUGATE TO PROTECT BIOMATERIALS FROM UNDESIRED

IMMUNE ATTACK

Clément Bechtler (1), Ekaterina Umnyakova (1), Amal Hamid (1), Lukas Kaufmann (1), Richard Pouw

(1), Christoph Q. Schmidt (2), John D. Lambris (3), Daniel Ricklin (1)

1) Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland2) Institute of Pharmacology of Natural Products & Clinical Pharmacology, Ulm University, Helmholtzstraße 20, 89081

Ulm, Germany3) University of Pennsylvania School of Medicine, Department of Pathology & Laboratory Medicine, 401 Stellar-Chance

labs, 422 Curie Boulevard Philadelphia, PA 19104-6100, USA

Although important progress has been made to protect biomaterials such as transplants, implants or liposomes

from undesired host immune recognition and attack, several problems remain unsolved. One of these is the

involvement of the complement system, the humoral part of innate immunity. It broadly and swiftly recognises

non-self surfaces, leading to direct cell damage and induction of the adaptive and cellular innate immune system.

In order to restrict complement activation to non-self or degenerated surfaces, the organism tightly controls

complement through regulators in solution and on surfaces. One promising approach to protect biomaterials from

immune attack, inspired by microbial immune evasion, is to specifically recruit these large molecular

complement regulators with small to medium-sized molecules to surfaces and to prevent complement attack insitu.

Pursuing this idea, a disulphide-bridged cyclic peptide (5C6) was previously discovered by our group through

phage display screening. 5C6 showed nanomolar binding affinity to the plasma-borne, major complement

regulator Factor H (FH). Attractively, FH inhibits complement’s central self-amplificatory C3 convertases where

all three activation pathways converge. 5C6 could reduce complement activation when combined with

appropriate tethering motifs by acting as a bridge between FH and model surfaces [1,2].

Based on this initial hit, comprehensive structure-activity relationship studies were conducted in which all

residues having been identified for being important for the FH-5C6 interaction were investigated through

replacement with commercially available or tailor-made building blocks. Furthermore, changes in the

macrocycle size and efforts to replace the disulphide by a more bioinert functional group allowed us to further

improve the properties of 5C6. Moreover, the ideal position for the tether conjugation was determined by ELISA

and biophysical methods. Finally, functional tests in clinically relevant models are currently being undertaken to

assess the translational significance of these findings.

References

1) Wu, Y. Q.; Qu, H.; Sfyroera, G.; Tzekou, A.; Kay, B. K.; Nilsson, B.; Nilsson Ekdahl, K.; Ricklin, D.; Lambris, J. D.,

Protection of nonself surfaces from complement attack by factor H-binding peptides: implications for therapeutic medicine.

Journal of Immunology 2011, 186 (7), 4269-77.

2) Nilsson, P. H.; Ekdahl, K. N.; Magnusson, P. U.; Qu, H.; Iwata, H.; Ricklin, D.; Hong, J.; Lambris, J. D.; Nilsson, B.;

Teramura, Y., Autoregulation of thromboinflammation on biomaterial surfaces by a multicomponent therapeutic coating.

Biomaterials 2013, 34 (4), 985-94.

EFMC-ISMC | 411

Z008

FROM THE DESIGN TO THE IN VIVO EVALUATION OF

BENZOHOMOADAMANTANE-DERIVED SOLUBLE EPOXIDE

HYDROLASE INHIBITORS FOR THE TREATMENT OF ACUTE

PANCREATITIS

Sandra Codony (1), Christophe Morisseau (2), Elena Valverde (1), Isabel Loza (3), Jose M. Brea (3),

Concepción Pérez (4), María Isabel Rodríguez Franco (4), Bruce D. Hammock (2), Santiago Vázquez (1)

1) Laboratori de Química Farmacèutica (Unitat Associada al CSIC), Departament de Farmacologia, Toxicologia i QuímicaTerapèutica, Facultat de Farmàcia i Ciències de l’Alimentació, and Institute of Biomedicine (IBUB), Universitat de

Barcelona, Av. Joan XXIII, 27-31, 08028 Barcelona, Spain2) Department of Entomology and Nematology and Comprehensive Cancer Center, University of California, Davis, CA

95616, USA3) Drug Screening Platform/Biofarma Research Group, CIMUS Research Center. University of Santiago de Compostela

(USC), Santiago de Compostela, Spain4) Institute of Medicinal Chemistry, Spanish National Research Council (CSIC), C/Juan de la Cierva 3, 28006 Madrid, Spain

Soluble epoxide hydrolase (sEH) converts epoxyeicosatrienoic acids (EETs), endogenous chemical mediators

derived from arachidonic acid (AA), to their corresponding dihydroxyeicosatrienoic acids (DHETs). Taking into

account that EETs show anti-inflammatory, antihypertensive, analgesic, angiogenic, and antiatherosclerotic

effects, it has been proposed that inhibition of sEH may have therapeutic effects in various inflammatory

diseases.1 A number of very potent sEH inhibitors (sEHI) have been developed, several of them featuring an

adamantane moiety or an aromatic ring. Taking into account that both groups fit very well in the hydrophobic

pocket of the sEH and that the replacement of adamantane by larger polycyclic rings seems to be a promising

strategy to obtain more potent sEHI,2 herein we have designed and synthesized a novel series of sEHI bearing

the very versatile benzohomoadamantane scaffold as the hydrophobic moiety. This polycyclic, readily accessible

system, features a homoadamantane unit fused with an aromatic ring and permits several chemical

derivatizations in its structure. Most of the new compounds were endowed with low nanomolar or even

subnanomolar IC50 values against the human and murine sEH enzyme. Further in vitro profiling (human, mice

and rat microsomal stability, solubility, cytochromes inhibition, selectivity) and pharmacokinetic studies allowed

us to select a candidate for an in vivo efficacy study in a mice model of cerulein-induced acute pancreatitis (AP).

We found that our candidate, administered after the induction of AP by cerulein, significantly reduced pancreatic

damage and improved the health status of the animals. In summary, these results suggest that sEHI may be of

clinical interest for treating AP.

References

1) C. Morisseau; B. D. Hammock, Annu. Rev. Pharmacol. Toxicol. 2013, 53, 37-58.

2) Codony, S.; Valverde, E.; Leiva, R.; Brea, J.; Loza, M. I.; Morisseau, C.; Hammock, B. D.; Vázquez, S. Bioorg. Med.

Chem. 2019, 27, 115078.

412 | EFMC-ISMC

Z009

OPTIMIZATION OF A NOVEL FAST ACTING TRANSMISSION

BLOCKING ANTIMALARIAL AGENT

Laura Mourot (1), Marjorie Schmitt (1), Elisabeth Mouray (2), Isabelle Florent (2), Sébastien Albrecht (1)

1) Laboratoire d’Innovation Moléculaire et Applications, Université de Haute-Alsace, Université de Strasbourg, CNRS, 3bisrue Alfred Werner, 68093-Mulhouse, France

2) Laboratoire Molécules de Communication et Adaptation des Micro-organismes, CNRS, Muséum National d’HistoireNaturelle, 63 rue Buffon 75005-Paris, France.

Despite significant progress in the control of malaria with a net reduction of morbidity and mortality over the

past years, it remains as one of the deadliest infectious diseases in the world. New drugs with broad therapeutic

potential and novel modes of action to overcome emerging drug resistances are urgently needed. Key features of

the next-generation antimalarial, termed single-exposure radical cure and prophylaxis (SERCaP), have been

rationalized and resulted in the recommendation of a series of Target Candidate Profiles (TCPs). Notably, TCP1

requires rapid elimination of the initial parasite burden, at least as fast as chloroquine.

In this context, the quinazolinedione MMV665878 with its antimalarial activities against multiple life stages of

Plasmodium, fast acting and transmission blocking activities, has great potential to deliver useful drugs for

malaria parasite eradication. Indeed, it displays potent parasite growth inhibitory activities on erythrocytic

stages, but modest activity on liver stages,

1

activities on early ring blood stages inducing greater metabolic

perturbation than artemisinin

2

and an initial cytocidal activity greater than chloroquine but slower than

dihydroartemisinin.

3

It also inhibits mature stage V gametocytes, potentially preventing P. falciparum

transmission to mosquitoes.

4

More recently, it has been described as a potent red blood cells invasion inhibitor.

5

Moreover, this quinazolinedione-based scaffold shows remarkable selectivity window with a low toxicity for

human cells and no cardiotoxicity risk. However pharmacokinetic issues are encountered and include moderate

overall exposure and/or modest bioavailability,

1

issue probably caused by rapid metabolism and elimination.

Herein, we report our progress towards the optimization of this quinazolinedione-based antimalarial series.

References

1) Van Voorhis, W. C. et al. PLOS Pathogens 2016, 12, e1005763(1).

2) Creek, D. J. et al. Antimicrob. Agents Chemother. 2016, 60, 6650–6663.

3) Ullah, I. et al. J. Antimicrob. Chemother. 2017, 72, 717–726.

4) Almela, M. J. et al. PLOS ONE 2015, 10, e0135139.

5) Burns, A. L. et al. FEMS Microbiol. Rev. 2019, 43, 223–238.

EFMC-ISMC | 413

Z010

SYNTHESIS AND PHARMACOLOGY EVALUATION OF NOVEL 1-

BENZYL-N-(4-(4-ARYLPIPERAZIN-1-YL)PHENYL)PIPERIDIN-4-

CARBOXAMIDES, AS POTENTIAL ACETYLCHOLINESTERASE

INHIBITORS

Mihajlo Krunić (1), Jelena Penjišević (1), Ivana Jevtić (1), Milovan Ivanović (2), Slađana Kostić-Rajačić

(1)

1) Institute for Chemistry, Technology and Metallurgy, University of Belgrade, Njegoševa 12, 11000, Belgrade, Serbia2) Faculty of Chemistry, University of Belgrade, Studentski trg 12-16, 11000, Belgrade, Serbia

Alzheimer’s disease (AD) is a neurodegenerative disease which affects over 50 milion people worldwide.

1)

AD

mainly affects people ages 65 and above. Symptoms are memory loss, decline in learning capacity, and language

problems. One of currently available treatments for Alzheimer's disease consists of Cholinesterase (ChEIs)

inhibitors, which raise acetylcholine levels in the brain. The drugs currently approved by the FDA  for this

purpose are donepezil, rivastigmine and galantamine.

As a part of our research we designed and synthesized seven novel compounds (6a-g) as potential

acetylcholinesterase inhibitors (AChEIs). The compounds contain N-benzyl piperidine moiety, which is common

in many inhibitors including donepezil,

2) 

and N-aryl piperazine moieties.

3)

The synthetic route consists of three steps. The first step is SnAr substitution of Aryl-piperazines 1 with

4-fluoronitrobenzene 2.

4)

The second step is the reduction of the nitro group using tin in hydrocholoric acid. The

third and final step is N-acylation with 1-benzylpiperidine-4-carboxylic acid 5 using propanephosphonic acid

anhydrid (T3P) affording the final products 6a-g (Scheme 1).

Scheme 1. Synthesis of novel 1-benzyl-N-(4-(4-arylpiperazin-1-yl)phenyl)piperidin-4-carboxamides 6a-g

 

All synthesized compounds were tested for their AChE inhibitory activity, and showed moderate to high

inhibitory effect.

References

1) Alzheimer’s Disease International. 2019. World Alzheimer Report 2019: Attitudes to dementia. London: Alzheimer’s

Disease International.

2) Jin, W.; Zhi-Min, W.; Xue-Mei, L.; Fan, L.; Jia-Jia, W.; Ling-Yi, K.; Xiao-Bing, W. Bioorg. Med. Chem. 2016, 24,

4324-4338

3) Anna, W.; Tomasz, W.; Justyna, G.; Adam, B.; Monika, M.; Agata, S.; Krzysztof, W.; Paula, Z.; Damijan, K.; Monika, G.

L.; Grzegorz, K.; Gniewomir, L.; Kamil, M.; Marcin, K.; Jan, K.; Ondrej, S.; Marketa, B.; Katarzyna K. K.; Stanislav, G.;

Barbara, M. ACS Chem. Neurosci. 2018, 9, 1195-1214.

4) Manisha, K.; Santosh, K. R.; Sameena, A.; Anjana, V.; Manisha , T. Bioorg. Med. Chem. 2009, 17, 1890-1897.

414 | EFMC-ISMC

Z011

SYNTHESIS, CHARACTERIZATION, AND IN VITRO CYTOTOXIC

EVALUATION OF NOVEL

THIAZOLO[3,2-b]-1,2,4-TRIAZOLE-6(5H)-ONE DERIVATES

Ahmet Avci (1), Nazlihan Aztopal (2), Engin Ulukaya (3), Nesrin Gokhan Kelekci (1), Birsen Tozkoparan

(1)

1) Hacettepe University, Faculty of Pharmacy, Department of Pharmaceutical Chemistry, Ankara-Turkey2) Department of Molecular Biology and Genetics, Faculty of Science and Literature, İstinye University, İstanbul, Turkey

3) Department of Medical Biochemistry, Faculty of Medicine, İstinye University, İstanbul, Turkey

Cancer is a deadly disease group that starts with the uncontrolled division of cells as a result of DNA damage

and can spread to other tissues and organs through metastasis. It is the second most common cause of death in

the world (1). Treatment protocols vary according to the type and stage of cancer. However, chemotherapy is the

most important way amongst others. Although there have been great developments in the chemotherapeutics in

recent years, success in cancer’s definitive treatment isn't still satisfactory (2). On the other hand existing drugs

have some serious side effect, selectivity problems and resistance development potential (3). Therefore,

discovery of new drugs in this field is crucial and still maintains its importance.

1,2,4-Triazole ring system has been demonstrated to possess antibacterial, antifungal, anticonvulsant,

antitubercular, antidepressant, analgesic, anti-inflammatory, antiviral activities (4). One of the most interesting

and important biological activities of compounds containing 1,2,4-triazole ring is undoubtedly its anti-cancer

effect. FDA-approved aromatase inhibitors anastrozole, letrozole, vorozole and poly (ADP-ribose) polymerase

(PARP) enzyme inhibitor talazoparib are the compounds having 1,2,4-triazol ring that are still used in breast

cancer treatment effectively (5,6).  Thiazolidine-2,4-dione structure draw attention for their anti-hyperglycaemic,

antimicrobial, anti-inflammatory and anticancer activities and it has been reported by various researchers that

various 5-ylidene-thiazolidin-2,4-dione derivative compounds have a remarkable cytotoxic effect in different

cancer cell lines under in vitro conditions (7,8). The aim of this study is to obtain alternative chemotherapeutic

agents for the treatment of cancer. For this purpose, a new group of hybrid compounds with 5-arylidene-thiazole

[3,2-b][1,2,4]triazole-6(5H)-one structure combining 1,2,4-triazole and 5-benzylidenthiazolidinone ring systems.

Their structures have been elucidated and their cytotoxic activities have been investigated in various cancer cell

lines in this study. The results of the preliminary pharmacological data will be discussed in poster.

References

1) World Health Organisation. Cancer. https://www.who.int/news-room/fact-sheets/detail/cancer

2) Yadaw, P. et al. 2017. “Green synthesis and anticancer potential of chalcone linked-1,2,3-triazoles“, European Journal of

Medicinal Chemistry, 126, 944-953.

3) Hoelder, S., Clarke, P., Workman, P. 2012. “Discovery of small molecule cancer drugs: successes, challenges and

opportunities”, Molecular Oncology, 6, 155-176.

4) Aggarwal, R., Sumran, G. 2020. “An insight on medicinal attributes of 1,2,4-triazoles”, European Journal of Medicinal

Chemistry, 205, 112652.

5) Fabian, C.J. 2007. “The what, why and how of aromatase inhibitors: hormonal agents for treatment and prevention of

breast cancer”, Int. J. Clin. Pract., 61(12), 2051-2063.

6) Abou-Seri, S.M. 2010. “Synthesis and biological evaluation of novel 2, 4′-bis substituted diphenylamines as anticancer

agents and potential epidermal growth factor receptor tyrosine kinase inhibitors“, Eur. J.Med. Chem., 45, 4113-4121.

7) Wu, S., et al. 2006. “Anticancer activity of 5-benzylidene-2-phenylimino-1,3-thiazolidin-4-one (BPT) analogs”, Medicinal

Chemistry, 2(6), 597-605.

8) Liu, X., et al. 2010. “Discovery and SAR of thiazolidine-2,4-dione analogues as insulin-like growth factor-1 receptor

(IGF-1R) inhibitors via hierarchical virtual screening”, Journal of Medicinal Chemistry, 53(6), 2661-2665.

EFMC-ISMC | 415

Z012

ANTIOXIDANT CINNAMIC ACID DERIVATIVES WITH

ANTI-INFLAMMATORY AND HYPOLIPIDEMIC POTENCY

Georgios Papagiouvannis (1), Vaia-Marina Kotoula (2), Panagiotis Theodosis-Nobelos (1), Eleni A. Rekka

(2)

1) Department of Pharmacy, School of Health Sciences, Frederick University, Nicosia, 1036, Cyprus2) Department of Medicinal Chemistry, School of Pharmacy, Faculty of Health Sciences, Aristotle

University of Thessaloniki, Thessaloniki 54124, Greece

Reactive oxygen species (ROS) are involved in all aspects of aerobic life. If the balance between ROS

production and the biologic antioxidant defence is disturbed, they are able to cause oxidative damage. This

imbalance is involved in aging and age-related disorders, atherosclerosis, degenerative disorders and

carcinogenesis. Antioxidants are able to protect the organism against radical damage or to enhance the normal

cellular defence against oxidative injury. ROS and inflammation have been identified in the pathogenesis and

development of numerous diseases, including cancer, nervous system disorders, such as Alzheimer’s and

Parkinson’s diseases, cardiovascular like atherosclerosis, metabolic such as diabetes mellitus, gastrointestinal

and other autoimmune diseases. Vascular oxidative stress and increased production of reactive oxygen species

can promote atherogenesis. In hypercholesterolemic patients with coronary artery disease, NADPH oxidase and

xanthine oxidase appear to be the major sources of superoxide anion radical. Both native and oxidized LDL have

been shown to stimulate ROS production. Furthermore, considerable evidence indicates that many events

contribute to AD progression, including oxidative stress, inflammation and altered cholesterol homeostasis.

Lipoxygenase (LOX) inhibitors can exert anti-inflammatory activity and also limit the shifting of arachidonic

acid metabolism towards LOX pathway. Additionally, the activity of lipoxygenases 5, 12 and 15 in the

atherosclerotic plaques and their implication in the inflammation of lipid tissueis well established. Cinnamic acid

and cinnamalaldehyde exert antioxidant, anti-inflammatory and cytoprotective activity, increasing NO synthase

and decreasing COX-2 and NF-kB expression. Furthermore, we have recently reported that esters with cinnamyl

alcohol possess higher anti-inflammatory activity, in comparison with the parent drugs, as well as lipoxygenase

inhibitory action. Thiomorpholine derivatives have been found to acquire hypolipidemic and antioxidant actions

and 3-pyridinepropanol may offer neuroprotective properties.

In view of the above, cinnamic acid bearing moieties (ferrulic acid, methoxy ferrulic acid and sinnapic acid)

were combined with esterification of amidation with cinnamyl alcohol, thiomorpholine and 3-pyridinepropanol,

in order to exhibit two or more pharmacological actions, since multifunctional compounds may assist towards

the avoidance of adverse effects and better patient tolerance, in comparison to the multiple drug treatment.

The compounds were synthesized by amidation and esterification reactions using N,N-dicyclohexylcarbodiimide

(DCC) or carbonyldiimidazole (CDI) as coupling agents. They were purified by flash column chromatography

and identified spectrometrically (

1

H-NMR,

13

C-NMR, MS) and with elemental analysis.

Most of the synthesised compounds were found to have in vitro antioxidant activity as lipid peroxidation

inhibitors, act as DPPH radical scavengers. They were also moderate inhibitors of lipoxygenase and could

decrease paw oedema in vivo considerably. Furthermore, thiomorpholine derivatives could reduce all lipidemic

indices, partly similar to that of simvastatin.

In conclusion, with the design of the described compounds of cinnamic acid derivatives with potentially active

alcohols and thiomorpholine, we aimed to acquire a series of biological properties able to prevent or restore a

number of pathological changes in conditions like neurodegenerative, inflammatory and cardiovascular

disorders. The described compounds may assist towards this direction.

 

416 | EFMC-ISMC

Z013

DESIGN AND SYNTHESIS OF POTENT INHIBITORS OF

TREHALOSE MONOMYCOLATE TRANSPORTER MMPL3 OF

MYCOBACTERIUM TUBERCULOSIS

Marianna Stampolaki (1), Satish R. Malwal (2), Dimitrios Evangelopoulos (3), Nikolaos Naziris (4), Maria

Chountoulesi (4), Costas Demetzos (4), John M. Kelly (5), Sanjib Bhakta (6), Eric Oldfield (2), Antonios

Kolocouris* (1)

1) Department of Pharmaceutical Chemistry, Faculty of Pharmacy, National and Kapodistrian University of Athens, 15771Athens, Greece

2) Department of Chemistry, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, USA3) UCL Centre for Clinical Microbiology, University College London, London NW3 2PF, UK

4) Department of Pharmaceutical Technology, Faculty of Pharmacy, National and Kapodistrian University of Athens, 15771Athens, Greece

5) Department of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London WC1E 7HT,UK

6) Mycobacteria Research Laboratory, Institute of Structural and Molecular Biology, Department of Biological Sciences,Birkbeck, University of London, London WC1E 7HX, UK

Tuberculosis (TB) remains a most lethal disease worldwide, caused by the bacillus Mycobacterium tuberculosis

(Mtb). An estimated 10.0 million people fall ill with TB every year and it is the leading cause of death among

carriers of HIV. The main difficulty in curing TB is the emergence of multi drug-resistant (MDR) and

extensively drug-resistant (XDR) TB. Thus, there is an urgent need for new, highly potent anti-tubercular drugs

that also target drug-resistant TB cases. Several compounds have been developed as anti-tubercular agents, many

of which have reached phase III clinical trials. (1) The ethylenediamine derivative, SQ109

(N-Geranyl-N΄-(2-adamantyl)ethane-1,2-diamine) is currently in phase IIb/III of clinical trials and shows high

potency against resistant Mtb strains and other pathogens. (2) SQ109 inhibits the MmpL3 transporter,

responsible for the translocation of mycolic acids in the form of trehalose monomycolate (TMM) which are

essential for the assembly of the mycobacterial outer membrane. (3)

The main aim of this work is to develop highly potent anti-tubercular therapeutics via structure-based drug

design of SQ109 analogues. In an effort to understand features of SQ109’s binding to MmpL3, docking

calculations and MD simulations were applied to the ligand using the X-ray structure of SQ109 in complex with

MmpL3 (PDB ID: 6AJG) from M. smegmatis that became available in 2019. (4, 5) The MD simulations and

further binding free energy calculations suggest that the addition of alkyl adducts on adamantane moiety may

improve binding affinity. Following the computational predictions, we synthesized compounds AK116-AK128

with an additional alkyl adduct installed in C-2 or C-1 of the adamantane ring. Geranylamine was synthesized

from the low-cost geraniol and the new ethylenodiamines were obtained through a mild reduction of the

corresponding aminoamide precursors, applied for the first time in these derivatives. The final ethylenodiamines

and the aminoamide precursors were tested for their biological activity against Mycobacterium tuberculosis,

smegmatis and abscessus, Trypanosoma cruzi and brucei, Escherichia coli, Bacillus subtilis, and Leishmania

mexicana. The biological testing results showed that several compounds have promising activity against most

pathogens and that in some cases the activity is increased as the alkyl chain is growing, with AK124 having the

best activity profile. On-going structural studies using cryo-em focus on unraveling the molecular basis of

inhibition. 

Additionally, previous research (6) showed that SQ109 can inhibit MmpL3 indirectly by perturbing plasma

membrane and subsequently affecting ΔpH and thus the electrochemical potential across the membrane. We

applied Differential Scanning Calorimetry (DSC) to explore the interaction of the synthetic diamines

AK116-AK128 with lipid bilayers DMPC, DSPG as model membranes. The experimental calorimetric results

showed that compounds SQ109, AK116-AK128 provoke alteration of the calorimetric data of DMPC and DSPG

membrane, an indication that they interact strongly with the lipid bilayers. Membrane perturbation and proton

gradient alternation needed across the membrane can hamper MmpL3 function.

References

1) Mahesh S. Vasava et al., Indian J. Tuberc. 66, 12–19 (2019).

2) Z. Gil et al., Parasitol. Res., 649–657 (2020).

3) J. M. Belardinelli et al., ACS Infect. Dis. 2, 702–713 (2016).

4) B. Zhang et al., Cell. 176, 636--648.e13 (2019).

5) C.-C. Su et al., Proc Natl Acad Sci USA. 116, 11241–11246 (2019).

6) W. Li et al., Antimicrob. Agents Chemother. 58, 6413–6423 (2014).

EFMC-ISMC | 417

Z014

NEW MULTITARGET THERANOSTIC COMPOUNDS AGAINST

ALZHEIMER´S DISEASE

Álvaro Sarabia, Marta Piquero, Pilar López-Alvarado, José Carlos Menéndez

Department of Chemistry in Pharmaceutical Sciences, Unit of Organic and Pharmaceutical Chemistry, Faculty of Pharmacy,Universidad Complutense de Madrid

Alzheimer´s Disease (AD) is the most prevalent type of dementia, affecting more than 40 million people

worldwide

1

. AD is a neurodegenerative disease causing cognitive impairment and difficulties in carrying out

daily life tasks. Even though its aetiology remains incompletely understood, several processes and alterations are

involved, such as amyloid beta-peptide hyperphosphorylated tau protein deposition, an increase in oxidative

stress and neuroinflammation, mitochondrial and calcium homeostasis dysfunction or an imbalance in the

glutamatergic and cholinergic tone.

Among commercially available drugs for the treatment of AD, none of them addresses the cause of the problem

nor cures the disease. They only achieve a temporary amelioration of symptoms. Hence, a new approach is

needed in the treatment of AD. Multitarget drugs are promising candidates for AD since they can solve various

alterations at the same time, which is interesting considering the multifactorial aetiology of the disease. On the

other hand, theranostic compounds can deliver therapy and provide diagnostic information at the same time,

allowing assessment of molecule activity, organism response and pharmacokinetics. This is of special interest for

research purposes and future clinical applications related to personalised medicine, and plays a special role in

current AD research

2

.

In relation to this, our research group focuses on styrylquinolines as potential drug candidates against AD due to

their promising properties

3

. In a recent PhD thesis work

4

, a novel family of these molecules were presented with

remarkable features, such as: inhibition of tau aggregation, neuroprotective and antioxidant activity, while

exhibiting beta amyloid detection in the near-infrared range. In this work, new derivatives are being synthesised

to continue the evaluation of this class of compounds.

References

1) Lane CA, Hardy J, Schott JM. Alzheimer’s disease. Eur J Neurol. 2018;25(1):59-70. doi:10.1111/ene.13439

2) Yang J, Zeng F, Ge Y, Peng K, Li X, Li Y, Xu Y. Development of Near-Infrared Fluorescent Probes for Use in

Alzheimer’s Disease Diagnosis. Bioconjug Chem. 2020;31(1):2-15. doi:10.1021/acs.bioconjchem.9b00695

3) Staderini M, Aulić S, Bartolini M, et al. A fluorescent styrylquinoline with combined therapeutic and diagnostic activities

against Alzheimer’s and prion diseases. ACS Med Chem Lett. 2013;4(2):225-229. doi:10.1021/ml3003605

4) Piquero Martí M. New multitarget theranostic compounds against proteinopathies and infectious diseases. PhD Thesis,

Universidad Complutense de Madrid, 2020. Supervisors: P. López-Alvarado, J. C. Menéndez

418 | EFMC-ISMC

Z015

SYNTHESIS, X-RAY CRYSTAL STRUCTURE AND ANTITUMOUR

ACTIVITY OF NOVEL DEPHENYLATED (–)-GONIOFUFURONE

ANALOGUE

Sladjana Kekezović (1), Bojana Srećo Zelenović (1), Mirjana Popsavin (1), Vesna Kojić (2), Marko Rodić

(1), Velimir Popsavin (1,3)

1) University of Novi Sad, Faculty of Sciences, Department of Chemistry, Biochemistry and Environmental Protection, TrgDositeja Obradovića 3, 21000 Novi Sad, Serbia

2) University of Novi Sad, Faculty of Medicine, Oncology Institute of Vojvodina, Put Dr Goldmana 4, 21204 SremskaKamenica, Serbia

3) Serbian Academy of Sciences and Arts, Knez Mihajlova 35, Belgrade, Serbia

(–)-Goniofufurone (1) is the opposite enantiomer of naturally occurring lactone (+)-goniofufurone, with

significant antitumour activity.

Novel dephenylated (–)-goniofufurone analogue, bearing a methoxy group at the C-5 position, was prepared

from D-glucose

[1]

 as shown in Scheme 1. The single crystal structure was determined through X-ray diffraction.

In vitro cytotoxicity of newly synthesized analogue was evaluated against eleven human tumour cell lines and a

single normal cell line (MRC-5). Structure-activity relationship (SAR) of 1, new analogue 8 and few analogues

previously synthesized in our laboratory will be presented in detail.

Scheme 1. (a) H

5

IO

6

, EtOAc, rt, 3 h; (b) Ph

3

P:CHCO

2

Me, MeOH, 0°C, 0.5 h then rt, 1.5 h, 68.5% for 5, 7.3%

for 4 from 2;

(c) 2.5% H

2

SO

4

/MeOH, reflux, 2 h, NaHCO

3

, rt, 1 h, 70.8%; (d) 9:1 TFA/H

2

O, rt, 1 h; (e) NaBH

4

, MeOH, rt,

1.5 h, 57.4% from 6.

 

 

Acknowledgment: The authors acknowledge financial support of the Ministry of Education, Science andTechnological Development of the Republic of Serbia (Grant No. 451-03-9/2021-14/ 200125) and SerbianAcademy of Sciences and Arts (Grants No. F-65 and F-130)

References

1) Francuz, J., Popsavin, M., Djokić, S., Kojić, V., Srdić-Rajić, T., Rodić, M., Jakimov, D., Popsavin, V., Med. Chem.

Commun., 2018, 9, 2017.

EFMC-ISMC | 419

Z016

DESIGN, SYNTHESIS AND BIOLOGICAL EVALUATION OF NEW

INHIBITORS OF MYCOBACTERIAL METHIONINE

AMINOPEPTIDASE 1A

Martin Juhás, Jan Zitko

Charles University, Faculty of Pharmacy in Hradec Králové,Akademika Heyrovského 1203, Hradec Králové, Czech Republic

Tuberculosis (TB) is an infectious disease caused by Mycobacterium tuberculosis (Mtb), responsible for over a

million of deaths each year. Despite the advances, the treatment is still lengthy and complicated due to frequent

resistance to the first-line or second-line drugs. Thus, the development of new potential antimycobacterial agents

remains a worldwide concern.

In this work, we present rational design, synthesis, and biological evaluation of inhibitors of an indispensable

mycobacterial enzyme methionine aminopeptidase 1a (MtMetAP1a). Compounds of the similar structure

(Figure 1) have already showed high activity against some microbial MetAP1

1,2

and some were highly active invitro growth inhibitors of Mtb

3,4

.

Figure 1 Design and structure of new inhibitors of MtMetAP1a.

All presented derivatives have been tested in vitro on a broad spectrum of mycobacterial strains. The highest

activity was seen with Ni(II) cofactor in 2-substituted compounds, best IC

50

= 0.6 µM (R

1

= 2-Br), the lowest

with Co(II). Several compounds also showed mediocre in vitro potency against Mtb (best MIC = 15.625 µg/mL).

Acknowledgements

This research was supported by the Grant Schemes at CU (reg. no. CZ.02.2.69/0.0/0.0/19_073/00169359).

References

1) Cui, Y.M.; Huang, Q.Q.; Xu, J., et al. Bioorg Med Chem Lett, 2005, 15, 4130-4135.

2) Luo, Q. L.; Li, J. Y.; Liu, Z. Y., et al. J Med Chem, 2003, 46(13), 2631-2640.

3) Dolezal, M.; Palek, L.; Vinsova, J., et al. Molecules, 2006, 11, 242-256

4) Kratky, M.; Vinsova, J.; Novotna, E., et al. Tuberculosis (Edinb), 2012, 92(5), 434-439.

420 | EFMC-ISMC

Z017

AMIDO-QUINUCLIDINE BASED QACs AS A NEW SOFT

ANTIMICROBIAL AGENTS SUSCEPTIBLE TO PROTEASE

DEGRADATION

Crnčević Doris, Odžak Renata, Šprung Matilda

Faculty of Science, University of Split, Department of ChemistryRuđera Boškovića 33, Split

Quaternary ammonium compounds (QACs) are biologically active amphiphiles displaying potent antimicrobial

properties (M.D. Joyce et al., 2015). Given the alarming number of QACs resistant environmental isolates, the

aim of further QACs research is to synthetise soft QACs variants more prone to either spontanious or enzymatic

degradation. Our previous studies on QACs have shown that quinuclidine is a good starting structure for

quaternization and that its derivatives have potent antimicrobial activity (Bazina et al., 2019). It is also known

that amide bond is susceptible to peptidase degradation. This fact implies that compounds containing such bond

could be degraded more easily in the environment by enzymatic decomposition. For these reasons, we were set

to synthesize new amido-quinuclidine derivatives with long alkyl chains to yield biologically active and more

environmental friendly QACs variants. A series of six new compounds QAd12, MeQAd12, AlyQAd12,

QAd14, MeQAd14 and AlyQAd14 were synthesized and analyzed for their antimicrobial activity using a panel

of pahtogenic bacteria and fungus. All compounds display good antimicrobial potential at concentrations in μM

range. To investigate if those compounds are succesfully degraded by cellular peptidases, enzymatic digestion of

compounds using cell lysates will be preformed. This finding might shed light on new potential mechanism for

production of environmental friendly QACs.

References

1.) M.D. Joyce et al., 2015. Natural product-derived quaternary ammonium compounds with potent antimicrobial activity. J.

Antibiot. 69(4), 344–347.

2.) L. Bazina et al., 2019. Discovery of novel quaternary ammonium compounds based on quinuclidine-3-ol as new potential

antimicrobial candidates. Eur. J. Med. Chem. 163, 626–635.

EFMC-ISMC | 421

Z018

ALPHA-TRIAZOLYL-BORONIC ACIDS: A PROMISING SCAFFOLD

AS PROTEIN KINASE (PK) INHIBITORS IN ACUTE MYELOID

LEUKAEMIA (AML)

Lorenza Destro (1), Maria Luisa Introvigne (2,3), Luca Mologni (4), Emilia Caselli (2), Alfonso Zambon

(1)

1) Department of Chemical and Geological Sciences, University of Modena and Reggio Emilia, Via Campi 103, 41125Modena, Italy

2) Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy3) Clinical and Experimental Medicine PhD Programme, University of Modena and Reggio Emilia, Modena, Italy

4) Department of Medicine and Surgery, University of Milan-Bicocca, 20900 Monza, Italy

There is an active interest in the field of protein kinase (PK) inhibitors to develop dual-targeting agents to

improve treatment outcomes. Combined inhibition should lead to superior anticancer activity through a)

simultaneous suppression of cooperating oncogenic signals and b) abrogation of bypass signalling that can

promote resistance to therapy.

1

 FLT3 and RET protein kinases (PKs) are often concomitantly activated in acute

myeloid leukaemia (AML),

2

 and the development of dual inhibitors of RET and FLT3 could meet the medical

need of AML patients showing abnormal activities of both these proteins. Therefore, there is an ongoing interest

in identifying novel chemotypes able to inhibit simultaneously both these targets.

1

 Consistently with its presence

in nature, as a structural and metabolic component of plants and as a regulator of mammalian vitamin D levels

and bone health, Boron is now widely considered as safe for incorporation into pharmaceuticals. Since 1990’s

different boronic acids and esters have been studied and now there are five boron containing drugs on the market

and several compounds are under investigation in clinical trials.

Alpha-triazolyl-boronic acids demonstrated

good activity in the field of antibiotic resistance as Boronic Acid Transition State Inhibitors (BATSIs).

4

 These

are known reversible covalent inhibitors of beta-lactamases, due to the tetrahedral adduct formed with the

enzyme, mimicking the one formed with the beta-lactam/antibiotic complex. Conversely, boronic acids are only

scantly reported as PK inhibitors.

3

 To identify new boronic compounds able to target FLT3 and RET, we

performed a docking study on a targeted library of in-house alpha-triazolyl-boronic acids. Recurrent binding

interactions were observed in FLT3 with the salt bridge (Glu661-Lys644), the DFG motif

(Asp829-Phe830-Gly831) and the hinge site (Cys694-Cys695) of the ATP binding site, consistently with the

common binding mode of PK inhibitors.

Compounds with the highest docking scores were assessed

enzymatically, leading to the identification of several alpha-triazolyl-boronic acids as micromolar hits. The

binding mode of these compounds is characterised by the formation of strong hydrogen bonds between the

boronic acid and the salt bridge, the pi-stacking of the triazole with two Phe and the interaction of the R group

with the hinge site (Fig.1). These results open the way to the development of a new class of boronic compounds

as PKs inhibitors.

References

1) Mologni, L.; Marzaro, G.; Redaelli, S.; Zambon, A. Dual Kinase Targeting in Leukemia. Cancers 2021, 13 (1)

2) Rudat, S.; Pfaus, A.; Cheng, Y. Y.; Holtmann, J.; Ellegast, J. M.; Bühler, C.; Marcantonio, D. D.; Martinez, E.; Göllner,

S.; Wickenhauser, C.; Müller-Tidow, C.; Lutz, C.; Bullinger, L.; Milsom, M. D.; Sykes, S. M.; Fröhling, S.; Scholl, C.

RET-Mediated Autophagy Suppression as Targetable Co-Dependence in Acute Myeloid Leukemia. Leukemia 2018, 32 (10),

2189–2202

3) Plescia, J.; Moitessier, N. Design and Discovery of Boronic Acid Drugs. Eur. J. Med. Chem. 2020, 195, 112270

4) Caselli, E.; Fini, F.; Introvigne, M. L.; Stucchi, M.; Taracila, M. A.; Fish, E. R.; Smolen, K. A.; Rather, P. N.; Powers, R.

A.; Wallar, B. J.; Bonomo, R. A.; Prati, F. 1,2,3 -Triazolylmethaneboronate : A Structure Activity Relationship Study of a

New Class of β.Lactamase Inhibitors against Acinetobacter Baumannii Cephalosporinase ( ADC-7 ). ACS Infect. Dis. 2020,

6 (7), 1965–1975

5) Saturno, G.; Lopes, F.; Niculescu-Duvaz, I.; Niculescu-Duvaz, D.; Zambon, A.; Davies, L.; Johnson, L.; Preece, N.; Lee,

R.; Viros, A.; Holovanchuk, D.; Pedersen, M.; McLeary, R.; Lorigan, P.; Dhomen, N.; Fisher, C.; Banerji, U.; Dean, E.;

Krebs, M. G.; Gore, M.; Larkin, J.; Marais, R.; Springer, C. The Paradox-Breaking PanRAF plus SRC Family Kinase

Inhibitor, CCT3833, Is Effective in Mutant KRAS-Driven Cancers. Ann. Oncol. 2021, 32 (2), 269–278

6) Huse, M.; Kuriyan, J. The Conformational Plasticity of Protein Kinases. Cell 2002, 109 (3), 275–282

422 | EFMC-ISMC

Z019

PHARMACOPHORE-BASED VIRTUAL SCREENING FOR THE

IDENTIFICATION OF NEW MYCOBACTERIUM TUBERCULOSIS

PKNG INHIBITORS

Miguel Quiliano, Alicia Arica, Roberto Alcántara

Centre for Research and Innovation, Faculty of Health Sciences, Universidad Peruana de Ciencias Aplicadas (UPC), Lima15023, Peru

Tuberculosis is an infectious disease caused by Mycobacterium tuberculosis. According to the WHO, 10 million

people fall ill with tuberculosis (TB) every year. TB is a preventable and curable disease; however, 1.5 million

people die from TB each year (1). To face the panorama, PknG emerges as a new target; PknG is a

Serine/threonine protein kinase that plays a crucial role in regulatory processes inside the mycobacterial cell.

Inhibition of PknG resulted in fusion blocking of phagosome-lysosome as well as its role in intrinsic antibiotic

resistance (2). Therefore, the discovery of new PknG inhibitors is necessary. Our group report here a

pharmacophore-based virtual screening approach using Liganscout software and Amazon Web Service.

Pharmacophore models by structure-based and ligand-based approaches were developed and theoretically

validated. Compound databases Zinc and ChEMBL were used to perform the virtual screening. After that,

docking experiments were used for better insight into the predicted binding mode of identified hits. Twelve

compounds were finally selected for in vitro analysis. Our results serve to initiate future projects where the

activity of the best candidates can be optimized

References

1) World Health Organization (2021) link: https://www.who.int/health-topics/tuberculosis#tab=tab_1

2) Sipos A, Pató J, Székely R, Hartkoorn RC, Kékesi L, Őrfi L, Szántai-Kis C, Mikušová K, Svetlíková Z, Korduláková J,

Nagaraja V, Godbole AA, Bush N, Collin F, Maxwell A, Cole ST, Kéri G. Lead selection and characterization of

antitubercular compounds using the Nested Chemical Library. Tuberculosis (Edinb). 2015 Jun;95 Suppl 1:S200-6. doi:

10.1016/j.tube.2015.02.028

EFMC-ISMC | 423

Z020

N1-SUBSTITUTED QUINOXALINE-2,3-DIONES AS LIGANDS FOR

GLUTAMATE KAINATE RECEPTORS

Paulina Chałupnik (1), Alina Vialko (1), Ewa Szymańska (1)

1) Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College,30-688 Cracow, Medyczna 9, [email protected]

2) [email protected]

Ionotropic kainate receptors are involved in excitatory postsynaptic currents in numerous regions of the central

nervous system, such as the hippocampus, cerebral cortex, spinal cord, or retina. For this reason, their

dysfunction can lead to neuronal damage and neurodegenerative diseases such as Alzheimer's disease,

Parkinson's disease, Huntington's chorea and multiple sclerosis. Focusing on the development and functional

activity of potentially selective antagonists may contribute to understanding the function and location of

individual kainate receptors. These tetrameric receptors can be composed of: GluK1, GluK2 or GluK3 subunits

known as low affinity subunits and GluK4 and GluK5 which are high affinity subunits. In recent years,

antagonists for GluK1 receptors have been well studied and described, including several crystal structures, and

only a few compounds that bind preferentially to GluK2 or GluK3 subunits.

The present project is the continuation of recently published results of studies on the new series of

quinoxaline-2,3-dione derivatives acting as KAR antagonists with structural modifications within the N1-amide

moiety as well as 6- and 7-positions of the bicyclic core

1,2

. Based on the structures of the most active compounds

described, a series of novel quinoxalino-2,3-dione derivatives were designed with the aim of increasing affinity

for kainate receptors and selectivity to GluK3 receptors. 

Fig. 1. General structure for investigated compounds.

The target quinoxaline-2,3-diones were successfully synthesized and characterized in receptor binding studies at

recombinant homomeric ionotropic glutamate receptors (GluK1, GluK2, GluK3 and GluA2). In the present work

the obtained results are reported.

This work was supported by Jagiellonian University Medical College (grant to. N42/DBS/000113).

References

1) Pallesen, J. et al. N1 ‑substituted quinoxaline-2,3-diones as kainate receptor antagonists: x‑ray crystallography,

structure−affinity relationships, and in vitro pharmacology. ACS Chem. Neurosci. 10, 1841–1853 (2019).

2) Møllerud, S. et al. N-(7-(1H-imidazol-1-yl)-2,3-dioxo-6-(trifluoromethyl)-3,4-dihydroquinoxalin-1(2H)-yl)benzamide, a

new kainate receptor selective antagonist and analgesic: synthesis, x-ray crystallography, structure-affinity relationships, and

in vitro and in vivo pharmac. ACS Chem. Neurosci. 10, 4685–4695 (2019).

424 | EFMC-ISMC

Z021

WUXI HitS: UNLEASHING THE POTENTIAL OF HIT DISCOVERY

Laiyin Nie

Crelux, a WuXi Apptec companyAm Klopferspitz 19a

82152 Martinsried-PlaneggGermany

HitS is a unit under WuXi AppTec Research Service Division. With a focus on DNA Encoded Library (DEL)

technology, HitS offer a service covering the whole target to hit platform: protein production, DEL and

Fragment Based screening (FBDD), and hit validation (biophysical & biochemical panel, structure-based drug

discovery). HitS has four operational sites located in China, Germany and the USA and more than 170 proficient

scientists, assuring the high quality data delivery in a timely manner.

The protein production service covers from construct design/protein engineering to crystal grade protein

delivery. We have worked on a variety of targets, including nucleic acid binding proteins, DNA repairing factors

and transmembrane proteins. HitS provide three types of DEL services designed for different needs: DELpro

contains a library of 46+ billion molecules and offers a one-stop solution, DELight has 15+ billion molecules in

its library with unprecedented data release and DELopen provides free access to DEL for academic users. In

addition, our FBDD library has 3100 fragments. The advanced Dianthus device enables us to run and analyze the

carefully tendered HitS Fragment Library or your own library at an unparalleled speed. HitS has a

comprehensive biophysical and biochemical technology platform for hit confirmation, including innovative

biophysical technologies like TRIC (Dianthus) and SwitchSense, and state-of-the art biochemical approaches

like HTRF and AlphaScreen. The structure biology group utilize X-Ray crystallography, cryo-EM and NMR to

determine de novo structures. Taken together, the cutting edge technologies at HitS allows a more efficient and

productive drug discovery process.

 

EFMC-ISMC | 425

Z022

SYNTHESIS OF FERROCENE ESTER DERIVATIVES OF

DESMURAMYL PEPTIDE

Vesna Petrović Peroković (1), Željka Car (1), Rosana Ribić (2), Monika Kovačević (3), Lidija Barišić (3)

1) Department of Chemistry, Faculty of Science, University of Zagreb, Horvatovac 102a, 10000 Zagreb, Croatia2) University Center Varaždin, University North, Jurja Križanića 31b, 42000 Varaždin, Croatia

3) Faculty of Food Technology and Biotechnology, University of Zagreb, Pierottijeva 6, 10000 Zagreb, Croatia

Derivatives of muramyl dipeptide (MDP), a well-known peptidoglycan fragment, have been extensively studied

as possible adjuvants for human and animal vaccines. [1] MDP analogues without the N-acetylmuramyl moiety

are called desmuramyl peptides (DMP). Structure-activity studies of the MDP derivatives and their analogues

suggest that L-Ala-D-isoGln pharmacophore is essential for the immunostimulatory properties and that the

introduction of lipophilic substituent can increase its adjuvant activity. In our previous research we reported the

synthesis and immunostimulatory activity of DMP with an adamantane group on L-Ala-D-iGln fragment. [2] Furthermore, we prepared and biologically evaluated mannosylated adamantane derivatives of DMP which

showed enhanced immunostimulatory activity compared to compounds without the mannose subunit. The

acitivity of such compounds was additionally positively influenced by the glycolyl spacer between mannose unit

and desmuramyl peptide. [3] The two bioorganometallic analogues of MDP bearing lipophilic ferrocene subunit

were also previously synthesized by our group. [4] In this work we described the synthesis of novel class of

DMP derivatives in which lipophilic ferrocene unit was introduced at C-terminus of L-Ala-D-iGln via ester

bond, followed by mannosylation of N-terminus through glycolyl spacer. Furthermore, alkyl spacers of different

length will also be introduced between desmuramyl peptide and ferrocene unit to further increase lipophilicity of

the synthesized compounds.

This work has been fully supported by Croatian Science Foundation under the project IP-2020-02-9162.

References

1) R. Ribić, M. Paurević, S. Tomić, Croat. Chem. Acta 92 (2019) 153-161.

2) (a) R. Ribić, L. Habjanec, B. Vranešić, R. Frkanec, S. Tomić, Chem. Biodivers. 9 (2012) 777-788., (b) R. Ribić, L.

Habjanec, R. Frkanec, B. Vranešić, S. Tomić, Chem. Biodivers. 9 (2012) 1373-1381.

3) R. Ribić, R. Stojković, L. Milković, M. Antica, M. Cigler, S. Tomić, Beilstein J. Org. Chem. 15 (2019) 1805- 1814.

4) L. Barišić, M. Roščić, M. Kovačević, M. Čakić Semenčić, Š. Horvat, V. Rapić, Carbohydr. Res. 346 (2011) 678-684.

426 | EFMC-ISMC

Z023

NOVEL ANTHRANILIC ACID DERIVATIVES AS PROMISING

ANTI-TB AGENTS

Maja Beus (1,2), Lauri Paulamäki (3), Krisi Savijoki (4,), Jari Yli-Kauhaluoma (4), Mataleena Parikka

(3), Branka Zorc (2)

1) Institute for Medical Research and Occupational Health, Zagreb, Croatia2) Faculty of Pharmacy and Biochemistry, University of Zagreb, Zagreb, Croatia

3) Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland4) Faculty of Pharmacy, University of Helsinki, Helsinki, Finland

Tuberculosis (TB) is still one of the top 10 causes of death, based on the WHO 2020 report (1), although much

effort has been dedicated to the development of new anti-TB agents. After the discovery of bedaquilline, the

quinoline heterocycle was recognized as a promising scaffold in search of new anti-TBC drugs. Inspired by this

fact, our research team developed a new series of primaquine (PQ) derivatives showing high antitubercular

activity (2). Additionally, a recent study involving 1280 fragment-library revealed a fragment from the

anthranilic acid series as a lead to develop small molecules that inhibit Mycobacterium tuberculosis MabA

activity (3). Bearing this in mind, we have synthesized 19 novel anthranilamides, hybrid compounds bearing

chloroquine (CQ), PQ or harmine (HA) moiety in the amine part, and anthranilic derivatives in the carboxylic

part of the molecule.

The synthesis is outlined in the Scheme. The first step involved the coupling of PQ/CQ/HA with anthranilic acid

moiety in the presence of HATU/DIEA or T3P/TEA and the second step was the removal of the protecting

group. The Boc-protecting group was removed in the presence of TFA, whereas the benzyl group was removed

by catalytic hydrogenation or with ammonium formate. The structures of novel compounds were confirmed by

1

H and

13

C NMR, IR, and MS spectroscopy. Antimycobacterial evaluation is in progress.

The study was supported by the Croatian Science Foundation through the research project IP-09-2014-1501 and

the Jane and Aatos Erkko Foundation (Helsinki, Finland). The work of doctoral student M. Beus has been fully

supported by the Young researcher’s career development project – training of doctoral students of the Croatian

Science Foundation founded by the European Union from the European Social Fund.

References

1) WHO - Tuberculosis; https://www.who.int/news-room/fact-sheets/detail/tuberculosis

2) Pavić K, et al. Eur J Med Chem. 2018; 143:769779

3) Faïon L et al. Eur J Med Chem. 2020; 200: 112440

EFMC-ISMC | 427

Z024

RATIONALLY DERIVED INHIBITORS OF HEPATITIS C VIRUS P7

CHANNEL

J Shaw (1,2), R Gosain (2,3), M Mon Kalita (4), T L Foster (1,2), J Kankanala (2,3), D Ram Mahato (4), S

Abas (2,3), B J King (5), C Scott (1,2), E Brown (1,2), M J Bentham (1,2), L Wetherill (1,2), A Bloy (1,2), A

Samson (1), M Harris (2,6), J Mankouri (2,6), D Rowlands (2,6), A Macdonald (2,6), A W Tarr (5), W B

Fischer (4), R Foster (2,3), S Griffin (1,2)

1) Leeds Institute of Medical Research, School of Medicine, University of Leeds, St James’ University Hospital, UnitedKingdom

2) Astbury Centre for Structural Molecular Biology, University of Leeds, United Kingdom3) School of Chemistry, University of Leeds, United Kingdom

4) Institute of Biophotonics, National Yang-Ming University, Taiwan5) School of Life Sciences, University of Nottingham, Queen’s Medical Centre, United Kingdom

6) School of Molecular & Cellular Biology, University of Leeds, United Kingdom

Hepatitis C virus (HCV) represents a global clinical challenge as a major cause of chronic liver disease, with

severe complications including cirrhosis, liver failure and primary liver cancers. The HCV viroporin p7 plays

pivotal roles during virion assembly involving protein–protein interactions. Furthermore, p7 protein is capable of

oligomerising within membranes to form an ion channel complex with an essential role during virion secretion.

[1]  

Both hairpin- and triple-helix-based p7 channel structures retain an adamantine-binding site upon the channel

periphery. In previous work, our group described an in silico high throughput screening using a hairpin-based p7

channel complex as a template. It resulted in 12 chemical hits which displayed considerably improved potency

compared with rimantadine, the prototypic p7 inhibitor. However, initial hits lacked convergence around a

common pharmacophore and this prevented understanding of a structure-activity relationship (SAR). [2] 

Herein, we present a second-generation of lead-like oxindole based inhibitors of p7 channel activity with a

comprehensive SAR. 41 compounds were tested, 20 of them contributed directly to the rational SAR. The SAR

was largely consistent with the energetically preferred in silico docking predictions in the peripheral binding site

of hairpin-based p7 channels models, defining key determinants of its activity. In addition, molecular dynamics

supported the stable binding at the peripheral site of our lead compound. The lead compound of this new family

showed an excellent potency, comparable to sofosbuvir and improved to rimantidine. The resultant step forward

in potency and specificity has led to the identification of a second biological role for p7 channel activity during

virus entry. We have also generated a fluorescently labelled tool compound as a chemical probe that retained

biological activity and allowed to calculate the compound concentration by fluorimetry. [3] 

In conclusion, our work shows that it is possible to take a step-change in targeting viroporins, providing a new

approach to antiviral therapy and generating novel research tools with which to dissect viroporin function.

References

1) Gentzsch, J. et al. PLOS Pathog. 2013, 9, e1003355; Griffin, S. D. et al. FEBS Lett. 2003, 535, 34–38.

2) Foster, T. L. et al. Hepatology 2014, 59, 408–422.

3) Shaw, J. et al. eLife 2020, 9, e52555.

428 | EFMC-ISMC

Z025

DEVELOPMENT OF INNOVATIVE MULTITARGET DRUGS FOR

AMYOTROPHIC LATERAL SCLEROSIS BY TARGETING

NEUROINFLAMMATION AND MODULATING IRON

DYSREGULATION

Pedro Soares (1), Carlos Fernandes (1), Sergio F. Sousa (2), Ana Alfenim (1), Daniel Chavarria (1),

Fernando Cagide (1), Fernanda Borges (1)

1) CIQUP/Department of Chemistry and Biochemistry, Faculty of Sciences University of Porto, Rua do Campo Alegre1021/1055, 4169-007 Porto, Portugal.

2) UCIBIO/REQUIMTE, BioSIM, Department of Biomedicine, Faculty of Medicine University of Porto, Alameda Prof.Hernâni Monteiro, 4200-319 Porto, Portugal.

Amyotrophic lateral sclerosis (ALS), also known as Charcot disease, is a devastating disease characterized by

the progressive loss of motor neurons (MNs) in the brain stem, spinal cord and motor cortex.

1

The disease

typically affects adults between 40-60 years of age, the familial form of the disease has an early onset, which

rapidly loses their voluntary movements and ultimately die by respiratory failure 2-5 years after diagnosis.

1

The

drugs available for ALS, Riluzole and Edaravone, are only capable of modestly extend the survival of the

patients (average 3-4 months) or delay the symptoms of the disease in a subset of patients.

2

Hence, there is great

pressure for the development of more efficient therapeutic solutions for ALS.    

Neuroinflammation is one of the most striking marks of ALS. In the last decade, some studies suggested NF-κB

transcription factor as a relevant regulator of inflammation in ALS and that its inhibition mitigates the

progression of the disease in animal models.

3,4

An abnormal accumulation of iron was also observed in the cerebrospinal fluid of ALS patients. Iron is capable

of catalyzing the formation of the highly toxic hydroxyl radical and contribute to the oxidative damage observed

in the tissues of ALS patients.

5

Accordingly, the use of iron chelators could provide benefits for ALS patients.

Additionally, iron chelators are capable to induce a mild stabilization of HIFs and trigger the expression of

neuroprotection genes beneficial for ALS.

6

Therefore, we propose the development of novel multitarget small-molecules capable to modulate NF-κB

activity and, at the same time, present the neuroprotective properties of iron chelators. Herein, we present the

results of the docking studies performed against IKKβ, a protein crucial for NF-κB activation, that lead us to

prioritize a set of 20 compounds containing iron-chelating moieties. After the synthesis of the small library,

preliminary in-vitro screening studies were performed.  The data obtained so far will be presented in this

communication. 

This project was supported by the European Union’s Horizon 2020 research and innovation program under the

Marie Sklodowska-Curie grant agreement No 895144. This project was also supported by FCT/FEDER funds

through COMPETE and national funds (grants UID/QUI/00081/2020, PTDC/MED-QUI/29164/2017,

PTDC/MED-FAR/29391/2017)

References

1) Longinetti, E. et al. Epidemiology of amyotrophic lateral sclerosis: An update of recent literature. Curr. Opin. Neurol.,

2019, 32, 771–776.

2) Jaiswal, M.K. Riluzole and edaravone: A tale of two amyotrophic lateral sclerosis drugs. Med Res Rev., 2019, 39 (2),

733–748.

3) Frakes, A.E. et al. Microglia Induce Motor Neuron Death via the Classical NF-kB Pathway in Amyotrophic Lateral

Sclerosis, 2014, Neuron, 81, 1009-1023

4) Dutta, K. et al. Mitigation of ALS Pathology by Neuron-Specific Inhibition of Nuclear Factor Kappa B Signaling, 2020, J.

Neurosci., 40 (26), 5137-5154.

5) Devos, D. et al. Conservative iron chelation for neurodegenerative diseases such as Parkinson’s disease and amyotrophic

lateral sclerosis, 2020, 127, 189-203.

6) Mereli, A. et. al. Understanding the Role of Hypoxia Inducible Factor During Neurodegeneration for New Therapeutics

Opportunities, Curr Neuropharmacol., 2018, 16 (10), 1484-1498.

EFMC-ISMC | 429

Z026

BUILDING A DIVERSE AND EXPERIMENTALLY-CURATED

FRAGMENT LIBRARY

Andrew Lowerson (1), Steven LaPlante (2), Patrick McCarren (3), Michael Serrano-Wu (3)

1) Key Organics Ltd, Cornwall, UK2) NMX Research and Solutions, Montreal, CA

3) Broad Institute, Cambridge, MA, USA

Fragment libraries are commonly assembled by Rule of 3 filtering followed by manual curation.  However, the

robust experimental data that ensures the proper physicochemical attributes needed for high-concentration

screening is often lacking and replaced instead by in silico calculations of uncertain predictive value. A fragment

collection with experimentally-determined aqueous solubility will address a major source of false positives and

attrition in fragment screening libraries: Aggregation, Stability, and Solubility. 

1

H NMR spectral data in

aqueous buffer will further enable practitioners to rapidly build fragment pools and initiate screening.

Diversity selection methods in shape, scaffold, fingerprint, and predicted property space combined with

industry-standard substructure filtering were used to select over 2,500 Key Organics compounds for

experimental profiling.  NMR and LCMS analysis allowed the careful selection of highly-soluble fragments with

desirable physicochemical and stability characteristics.   Importantly, the curated molecules are enriched in

cyclic scaffolds commonly found in drug candidates, and spans chemical space that minimally overlaps with

existing commercial collections.  This poster will summarize the experimental and cheminformatic features of

this next generation Key Organics ‘BIONET Premium Fragment Library’.

430 | EFMC-ISMC

Z027

SYNTHESIS OF NEW HETEROCYCLIC DERIVATIVES BEARING

BENZIMIDAZOLE MOIETIES AND EVALUATION OF THEIR

BACTERIAL FLOCCULATION PROPERTIES

Isalyne Drewek (1,2), Maxence De Cock (1), Aurélie Pietka (1,2), Dimitri Stanicki (1), Ruddy Wattiez (2),

Sophie Laurent (1,3)

1) NMR and Molecular Imaging Laboratory, General, Organic and Biomedical Chemistry Unit, University of Mons, B-7000Mons, Belgium

2) Department of Proteomics and Microbiology, University of Mons, B-7000 Mons, Belgium3) Center for Microscopy and Molecular Imaging, B-6041 Charleroi, Belgium

Bacterial flocculation is a biological phenomenon that has shown an increasing interest in recent years due to its

various applications, ranging from wastewater treatment to biodegradation or biocatalytic processes. During this

phenomenon, bacterial cells form eye-visible flocs. This process can be observed naturally but can also be

provoked by the use of flocculating agents

1

. The mechanism of action of the existing agents is mainly based on

electrostatic interactions between the positive charge carried by these systems and the negatively charged

bacterial membranes

2

. However, this specific mechanism of action, combined with high cost and toxicity are

constraints preventing the use of the current flocculating agents on an industrial scale

1

.

As the benzimidazole moiety is found in many structures exhibiting interesting pharmacological activity (such as

anti-viral, anti-fungal or even anti-bacterial agents

3

), our work focused on the development of new heterocyclic

derivatives bearing benzimidazole units. In recent studies, we have highlighted that some of these heterocyclic

structures exhibit an important activity against both Gram (+) and Gram (-) bacterial strains characterized by the

rapid appearance (within a few minutes) of flocs when the bacterial suspension is incubated with a low

concentration of derivative.   

Interestingly, the synthesis and evaluation of this chemical library allowed to highlight given structural criteria

essential for the expression of the flocculation activity. Moreover, the structure of the evaluated heterocycles

suggests a non-conventional mechanism of action when compared to classical “positively-charged” flocculating

agents. To go further into the comprehension of the observed phenomenon, we decided to carry out some

additional studies including scanning electron microscopy (SEM) and metabolomic experiments alongside with

other biological characterizations of the bacterial flocs.

References

1) Ojima, Y., Azuma, M. & Taya, M. Inducing flocculation of non-floc-forming Escherichia coli cells. World J. Microbiol.

Biotechnol. 34, 185 (2018).

2) Yang, Z. et al. Flocculation of Escherichia coli Using a Quaternary Ammonium Salt Grafted Carboxymethyl Chitosan

Flocculant. Environ. Sci. Technol. 48, 6867–6873 (2014).

3) Pullagura, M. K. P., et al. Potent Biological Agent Benzimidazole - A review. Int. J. Pharm. Pharm. Sci. 8, 22 (2016).

EFMC-ISMC | 431

Z028

STRUCTURAL FINE-TUNING OF DESMURAMYLPEPTIDE NOD2

AGONISTS DEFINES THEIR IN VIVO ADJUVANT ACTIVITY

Samo Guzelj (1), Sanja Nabergoj (1), Martina Gobec (1), Stane Pajk (1), Veronika Klančič (1), Bram

Slütter (2), Ruža Frkanec (3), Adela Štimac (3), Primož Šket (4), Janez Plavec (4), Irena Mlinarič-Raščan

(1), Žiga Jakopin (1)

1) Faculty of Pharmacy, University of Ljubljana, SI-1000 Ljubljana, Slovenia2) Div. BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands

3) Centre for Research and Knowledge Transfer in Biotechnology, University of Zagreb, 10000 Zagreb, Croatia4) Slovenian NMR Centre, National Institute of Chemistry, SI-1000 Ljubljana, Slovenia

Nucleotide-binding oligomerization-domain-containing protein 2 (NOD2) is a well characterized innate immune

receptor in the pattern recognition receptor superfamily. It is responsible for detecting and responding to

fragments of bacterial peptidoglycan, with muramyl dipeptide (MDP) known as the smallest peptidoglycan

molecular signature still capable of activating NOD2.

1

Activation of NOD2 results in a wide range of innate and

adaptive immune responses, which make NOD2 an attractive target for the development of novel vaccine

adjuvants and cancer immunotherapeutics.

2,3

The discovery that the full glycopeptide scaffold is not mandatory for NOD2 agonism led to the concept of

desmuramylpeptides, a class of compounds that lack the carbohydrate moiety of MDP. In our study, which was

recently published in the Journal of Medicinal Chemistry,

4

we report on the design, synthesis and biological

evaluation of a series of novel desmuramylpeptides, which are based on the structures of promising NOD2

agonists previously reported by our group.

5

Several compounds emerged as potent nanomolar in vitro NOD2

agonists, with immunostimulatory effects on peripheral blood mononuclear cells at the protein and

transcriptional levels. These compounds also augmented the dendritic-cell-mediated activation of T cells in an

antigen presentation assay and enhanced the cytotoxic activity of peripheral blood mononuclear cells against

malignant cells. Importantly, a C

18

lipophilic tail was identified as a pivotal structural element that confers invivo adjuvant activity in conjunction with a liposomal delivery system. Accordingly, liposome‑encapsulated

desmuramylpeptides showed promising adjuvant activity in a mouse model of adjuvanticity, surpassing that of

MDP, while achieving a more balanced Th1/Th2 immune response, thus highlighting their potential as vaccine

adjuvants.

References

1) Philpott, D. J.; Sorbara, M. T.; Robertson, S. J.; Croitoru, K.; Girardin, S. E. Nat. Rev. Immunol. 2014, 14, 9–23.

https://doi.org/10.1038/nri3565.

2) Chen, G.; Shaw, M. H.; Kim, Y.-G.; Nuñez, G. Annu. Rev. Pathol. Mech. Dis. 2009, 4, 365–398.

https://doi.org/10.1146/annurev.pathol.4.110807.092239.

3) Negroni, A.; Pierdomenico, M.; Cucchiara, S.; Stronati, L. J. Inflamm. Res. 2018, Volume 11, 49–60.

https://doi.org/10.2147/JIR.S137606.

4) Guzelj, S.; Nabergoj, S.; Gobec, M.; Pajk, S.; Klančič, V.; Slütter, B.; Frkanec, R.; Štimac, A.; Šket, P.; Plavec, J.;

Mlinarič-Raščan, I.; Jakopin, Ž. J. Med. Chem. 2021. https://doi.org/10.1021/acs.jmedchem.1c00644.

5) Gobec, M.; Tomašič, T.; Štimac, A.; Frkanec, R.; Trontelj, J.; Anderluh, M.; Mlinarič-Raščan, I.; Jakopin, Ž. J. Med.

Chem. 2018, 61, 2707–2724. https://doi.org/10.1021/acs.jmedchem.7b01052.

432 | EFMC-ISMC

Z029

PYRIMIDINE MIMICS AS NEXT GENERATION CATCH AND

RELEASE DNA DECOYS FOR TRANSCRIPTION FACTOR

MODULATION

Samantha A. Kennelly, Ruben Silva Otero, Ramkumar Moorthy, Daniel A. Harki

Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota, USA

The Catch and Release DNA Decoy (CRDD) technology previously developed in the Harki laboratory was

expanded with the development of nitro-functionalized pyrimidine mimics that photochemically depurinate

similarly to their 7-nitroindole (7-NI) predecessor.

1

 Known nucleosides comprised of 2-nitropyrrole (2-NP) and

3-nitropyrrole (3-NP) nucleobases, as well as novel pyrimidine mimics containing 2-nitroimidazole (2-NI) and

2-nitrobenzene (2-NB) nucleobases were studied for their use as CRDDs. Current CRDD designs utilize

single-photon UV-irradiation to cleave the anomeric bond, leading to separation of the sugar and nucleobase.

Transcription factor (TF)-targeted CRDDs can be used to study TF-DNA interactions whereas the native CRDD

binds to the TF (catch) and photolysis of the CRDD results in the release of the TF-CRDD complex. In the

current work, we reveal through characterization of photoproducts that 2-NP and 2-NI can function in CRDDs,

whereas 3-NP and 2-NB result in complex products upon photolysis. DNA photolysis studies confirmed fast

photolysis kinetics of 2-NP (t

1/2

 = 1.8 mins), which is similar to 7-NI. We conclude that CRDD technology can

now be applied to both purine and pyrimidine scaffolds and that our new monomers, 2-NP and 2-NI, possess

even greater DNA stability when base-paired against 7-NI. 

References

1) Struntz, N. B., Harki, D. A. ACS Chem. Biol. 2016, 11, 1631-1638.

EFMC-ISMC | 433

Z030

NOVEL SYNTHETIC NUCLEOSIDES AND EVALUATION OF THEIR

BIOLOGICAL ACTIVITY

Mikhail S. Drenichev (1), Anastasia A. Zenchenko (1), Vladimir E. Oslovsky (1), Georgy A. Ivanov (1),

Evgeniya O. Dorinova (1), Anna A. Kozlova (1), Dirk Jochmans (2), Johan Neyts (2), Dmitry I. Osolodkin

(3), Alexandra L. Zakharenko (4), Nadezhda S. Dyrkheeva (4), Olga D. Zakharova (4), Irina A.

Chernyshova (4), Arina A. Chepanova (4), Olga I. Lavrik (4)

1) Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov Str. 32, 119991 Moscow, RussianFederation

2) KU Leuven - University of Leuven, Laboratory for Virology and Chemotherapy, Rega Institute for Medical Research,Minderbroedersstraat 10, 3000 Leuven, Belgium

3) FSBSI Chumakov FSC R&D IBP RAS, 108819 Moscow, Russian Federation4) Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of Russian Academy of Sciences, Lavrentiev

Ave, 8, 630090, Novosibirsk, Russian Federation

Natural nucleosides have a diverse structure; they are part of nucleotides, DNA, RNA, and coenzymes. More

than 140 minor nucleosides have been isolated from tRNA alone, and about 100 disaccharide nucleosides and

200 nucleoside antibiotics have been isolated from various natural sources, which contain additional functional

groups and hydrophobic residues. The library of natural nucleosides contains about 600 compounds which serve

as a basis for the creation of new biologically active compounds [1]. In this work we have elaborated several

series of nucleoside derivatives by synthetic modification of initial natural compounds.

Antiviral activity. Naturally occurring 6-benzylaminopurine riboside (BAR) exhibits potent antiviral effect on

human enterovirus EV71, but appears to be rather cytotoxic [2-3]. It was previously shown, that introduction of

bulky iodine atom into phenyl ring substantially decreased its toxicity on cells [4]. In order to overcome BAR

toxic effects various chemical modifications in phenyl ring were tested. The replacement of iodine atom in the

phenyl residue of N6

-(4-iodobenzyl)adenosine with bulky hydrocarbon ethynylcyclopropyl or

ethynylcyclopentyl fragment resulted in an 11-and 30-fold decrease in anti-enterovirus activity respectively. At

the same time, the compounds containing these substituents are characterized by low cytotoxicity. The

replacement of iodine atom in the phenyl residue of N6

-(3-iodobenzyl)adenosine with a bulky hydrocarbon

substituent led to 20-fold decrease of anti-enterovirus activity and increase of cytotoxicity. The optimal chemical

modification of BAR leading to low-toxic compounds with high antiviral effects was the introduction of small

groups (OH, Me, CF

3

) or iodine atoms (I) into phenyl ring.

Enzyme inhibition and combined chemotherapy. DNA-repair systems counteract the action of antitumor drugs.

Therefore, the therapeutic effect depends on the effectiveness of DNA repair systems in cancer cells.

Combination of anticancer drugs with inhibitors of DNA-repairing enzymes can increase the effectiveness of

chemotherapy. The second part of our work was an elaboration of nucleoside inhibitors for

tyrosyl-DNA-phosphodiesterase-1 (Tdp-1), a key DNA-repairing enzyme [5]. One possible approach to

inhibiting Tdp1 is a combination of deoxyribonucleoside and “tyrosine-mimicking” aromatic residue in a

structure. Therefore, a number of Tdp1 inhibitors based on lipophilic derivatives of thymidine, uridine and BAR

have been obtained. New effective Tdp1 inhibitors with values of IC

50

= 0.6-53 µM were found in a series of the

synthesized compounds.

 This work was supported by Russian Foundation for Basic Research (grant No 20-34-70116).

References

1) A.A. Zenchenko, M.S. Drenichev, I.A. Ilicheva, S.N. Mikhailov, Russ.J.Mol.Biol., 2021, in press.

2) M. Arita, T. Wakita, H. Shimizu. J. Gen. Virol., 2008, 89, 2518-2530.

3) M.S. Drenichev, V.E. Oslovsky, L. Sun, A. Tijsma, N.N. Kurochkin, V.I. Tararov, A.O. Chizhov, J. Neyts, C.

Pannecouque, P. Leyssen, P., S.N. Mikhailov. Eur J. Med. Chem., 2016, 111, 84 – 94.

4) P. Dolezel, P. Koudelkova, P. Mlejnek. Toxicol. In Vitro, 2010, 24, 2079-2083.

5) E.J. Brettrager, R.C.A.M. van Waardenburg. Cancer Drug Resist., 2019, 2, DOI: 10.20517/cdr.2019.91.

434 | EFMC-ISMC

Z031

THE CONSTRUCTION OF A VERSATILE CHEMINFORMATICS

WORKFLOW FOR THE DESIGN OF 3D FRAGMENT LIBRARIES

Tom Dekker (1), Mathilde A.C.H. Janssen (2), Rene W.M. Aben (2), Hans W. Scheeren (2), Daniel

Blanco-Ania (2), Floris P.J.T. Rutjes (2), Maikel Wijtmans (1), Iwan J.P. de Esch (1)

1) Division of Medicinal Chemistry, Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije UniversiteitAmsterdam, The Netherlands

2) Division of Synthetic Organic Chemistry, Institute for Molecules and Materials (IMM), Radboud University Nijmegen, TheNetherlands

Fragment-based drug discovery (FBDD) has grown into a well-established drug discovery approach, as is

evident from the five FDA-approved drugs and various clinical candidates it has produced up to June 2021.

Compared to high-throughput screening, smaller molecules are screened which allows for the use of smaller

libraries to sample the same degree of chemical space. Like most screening libraries, FBDD libraries are

dominated by flat molecules and this has resulted in a growing interest in 3D fragments. We are developing new

chemistry to generate novel 3D fragment libraries and have constructed an automated cheminformatics workflow

for efficient 3D library design. This workflow uses the open source platform KNIME and selects novel and

diverse fragments with suitable physicochemical properties and 3D character (using metrics such as PMI) from

virtually generated libraries. The workflow can be used to design large screening libraries but is also applicable

for diverse reagent selection in smaller and more focused chemistry programs.

References

1) Lovering F, Bikkering J, et al., J. Med. Chem, 2009, 52, 21

2) Lovering F, Med. Chem. Commun., 2013, 4, 515

3) Walters WP, Green J, et al., J. Med. Chem. 2011, 54, 19

4) Sauer WHB, Schwarz MK, J. et al., Chem. Inf. Comput. Sci, 2003, 43, 3

5) Congreve M, Carr M, et al., Drug Discov. Today, 2003, 8, 19

6) Hamilton DJ, Dekker T, et al. Drug Discov. Today Technol. 2021, https://doi.org/10.1016/j.ddtec.2021.05.001

EFMC-ISMC | 435

Z032

POTENT AND SELECTIVE INDOL-4-YL

PYRAZOLO[1,5-a]PYRIMIDINE-DERIVED PI3Kδ INHIBITORS AS

POTENTIAL CANDIDATES FOR TREATMENT OF COPD AND

ASTHMA

Stanisław Michałek (1,2), Mariola Stypik (1,2), Marcin Zagozda (1), Nina Orłowska (1,2), Martyna

Banach (1), Beata Zygmunt (1), Kamila Gala (1), Maciej Dziachan (1), Barbara Dymek (1), Paweł

Gunerka (1), Paweł Turowski (1), Krzysztof Dubiel (1), Jerzy Pieczykolan (1), Maciej Wieczorek (1)

1) Celon Pharma Innovative Drugs Research & Development Department, Celon Pharma S.A., Marymoncka 15,Kazun Nowy, Poland

2) Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland

Phosphoinositide 3-kinase δ (PI3Kδ), the member of the class I PI3K family, is very important signalling

molecule which regulates differentiation, proliferation, migration and survival of immune cells

1,2

. Different

PI3K isoforms regulate distinct cellular events, enabling the therapeutic opportunities treatment inflammatory

and autoimmune  diseases including asthma or COPD

3,4

. There are PI3K inhibitors in the clinic and many more

in the preclinical development. Herein, we developed a new small molecule inhibitors based on indol-4-yl

pyrazolo[1,5-a]pyrimidine derivatives with IC

50

values in the low nanomolar range (18-897 nM) and with high

selectivity against PI3Kδ. One of synthesized compounds was chosen as candidate for pre-clinical studies

5

.

References

1) Norman P., Selective PI3Kδ inhibitors, a review of the patent literature, Expert Opinion on Therapeutic Patents,

1773-1790, (2011), DOI:.1517/13543776.2011.629606

2) Koyasu, S. The role of PI3K in immune cells, Nat Immunol, 313–319, (2003), DOI:10.1038/ni0403-313

3) Walker C., Thomas M., Edwards M. J., Phosphoinositide 3-kinase (PI3K) family of signalling enzymes and their role in

asthma, Drug Discovery Today: Disease Mechanisms, 63-69, (2006), DOI:10.1016/j.ddmec.2006.02.004

4) Pirozzi F, Ren K, Murabito A, Ghigo A. PI3K Signaling in Chronic Obstructive Pulmonary Disease: Mechanisms,

Targets, and Therapy, Curr Med Chem., 2791-2800, (2016), DOI:10.2174/0929867325666180320120054

5) Gunerka P., Gala K., Banach M., Dominowski J., Hucz-Kalitowska J., Mulewski K., et al. Preclinical characterization of

CPL302-253, a selective inhibitor of PI3Kδ, as the candidate for the inhalatory treatment and prevention of Asthma, PLoS

ONE, (2020), DOI:10.1371/journal.pone.0236159

436 | EFMC-ISMC

Z033

NOVEL BENZIMIDAZOLE-DERIVED PI3Kδ INHIBITORS AS

HIGHLY POTENT DRUG CANDIDATES FOR SLE AND OTHER

INFLAMMATORY AND AUTOIMMUNE DISEASES

Mariola Stypik (1,2), Stanisław Michałek (1,2), Marcin Zagozda (1), Nina Orłowska (1,2), Martyna

Banach (1), Beata Zygmunt (1), Kamila Gala (1), Maciej Dziachan (1), Barbara Dymek (1), Paweł

Gunerka (1), Paweł Turowski (1), Krzysztof Dubiel (1), Jerzy Pieczykolan (1), Maciej Wieczorek (1)

1) Celon Pharma Innovative Drugs Research & Development Department, Celon Pharma S.A., Marymoncka 15,Kazun Nowy, Poland

2) Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland

PI3K (Phosphoinositide 3-kinase) is the family of lipid kinases which participates in many key cellular processes

like proliferation, growing up, migration, cytokines production, and apoptosis. Inhibition of PI3K (especially the

first class which contains four subunits: α, β, γ, δ) is regarded to be very attractive and potential mechanism for

the treatment of many diseases like SLE (Systemic Lupus Erythematosus), MS (Multiple Sclerosis), IBD

(Inflammatory Bowel Disease), and other inflammatory and autoimmune diseases [1-4]. In this work we present

a series of a new, very potent, active and selective small molecule inhibitors based on the structure of

appropriately substituted 5-(2-difluoromethylobenzimidazo-1-yl)pyrazolo[1,5-a]pyrimidine [5]. We synthesized

a library of very active benzimidazole derivatives (PI3Kδ  IC

50

= 0.02 – 1.07 μM). Then we developed the

process of lead compound(s) synthesis to select the preclinical candidate for SLE.

References

1) John G. Foster, Matthew D. Blunt, Edward Carter, Stephen G. Ward; “Inhibition of PI3K Signaling Spurs New

Therapeutic Opportunities in Inflammatory/Autoimmune Diseases and Hematological Malignancies”; Pharmacol Rev.; 2012

Oct.; 64(4):1027-54; doi: 10.1124/pr.110.004051.

2) Edward Banham-Hall, Menna R. Clatworthy nad Klaus Okkenhaug; “The Therapeutic Potential for PI3K Inhibitors in

Autoimmune Rheumatic Diseases”; Open Rheumatol J.; 2012; 6:245-58.; doi: 10.2174/1874312901206010245.

3) Anne-Katrien Stark, Srvidya Sriskantharajah, Edith M. Hessel, Klaus Okkenhaug; “PI3K inhibitors in inflammation,

autoimmunity and cancer”; Curr Opin Pharmacol.; 2015 Aug; 23:82-91.; doi: 10.1016/j.coph.2015.05.017.

4) Peter K. Vogt, Jonathan R. Hart, Marco Gymnopoulos, Hao Jiang, Sohye Kang, Andreas G. Bader, Li Zhao, Adam

Denley; “Phosphatidylinositol 3-kinase (PI3K): The Oncoprotein”; Curr. Top Microbiol. Immunol.; 2010; 347:79-104.; doi:

10.1007/82_2010_80.

5) EP3277687B1; “7-(Morpholin-4-yl)pyrazole[1,5-a]pyrimidine derivatives which are useful for the treatment of immune or

inflammatory diseases or cancer”.

EFMC-ISMC | 437

Z034

SYNTHESIS AND SAR OF PEPTIDE JUVENOIDS AS POTENT

AGONISTS SPECIFIC TO AN INSECT JH RECEPTOR

Adela Novotna (1), Ivan Snajdr (1), Sarka Tumova (2), Magesh Muthu (3), Matej Milacek (4), Lenka

Bittova (2), Pavel Jedlicka (1), Roman Tuma (4), David Sedlak (3), Pavel Majer (1), Marek Jindra (2)

1) Institute of Organic Chemistry and Biochemistry CAS, Prague, Czech Republic2) Biology Center CAS, Ceske Budejovice, Czech Republic

3) CZ-OPENSCREEN, Institute of Molecular Genetics CAS, Prague, Czech Republic4) Faculty of Science, University of South Bohemia Ceske Budejovice, Czech Republic

Juvenile hormones are signaling molecules that are important regulators of development and reproduction of

insects. The main objective of this project was to determine the structure-activity relationship (SAR) of potential

juvenile hormone receptor (JHR) agonists and to explore agonists with a higher activity than the previously

known analogues.

1

The activity of JHR mimics was tested using a 2-hybrid assay on orders of Diptera,Coleoptera, Blattodea and Heteroptera.  

The variation of the central amino acid (X) and two side chains (R

1

, R

2

) of the JH analogue affected the activity,

compared to the previously synthetized ethyl (S)-4-(2-pivalamidopropanamido)benzoate

2

. The biological

activity of the JH agonist was shown to depend on the length of the alkyl ester (R

2

) on the aminobenzoate the

bulkiness and hydrophobicity of the R

1

and X substituents.

References

1) Jindra M, et al. (2013) Annu Rev Entomol 58:181-204

2) Zaoral M, Slama K (1970) Science 170:92‐3

438 | EFMC-ISMC

Z035

SYNTHESIS AND BIOLOGICAL EVALUATION OF FINGOLIMOD

DERIVATIVES AS ANTIBACTERIAL AGENTS

Matej Zore (1), Shella Gilbert-Girard (2), Ines Reigada (2), Jayendra Patel (1), Kirsi Savijoki (2), Adyary

Fallarero (2), Jari Yli-Kauhaluoma (1)

1) Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University ofHelsinki, Viikinkaari 5 E, FI-00014 Helsinki, Finland

2) Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki,Viikinkaari 5 E, FI-00014 Helsinki, Finland

Bacterial infections are one of the most important challenges of modern medicine. New mechanisms of

resistance are evolving and spreading around the world, threatening our ability to treat even common infectious

diseases.

1

However, despite the recognised need for new antibiotics, only a few new classes have been brought

to market in the last decades.

2

Repurposing non-antibiotic drugs as antimicrobials is an approach with many

advantages, namely that a large amount of information about the drug’s activity, pharmacokinetics and safety is

already available, facilitating its development for a new purpose.3 In this sense, we have recently discovered that

fingolimod, an FDA-approved immunomodulating drug, has a potent antibacterial activity against

Staphylococcus aureus (MIC 15 µM).

4

Fingolimod also displays a high activity against Acinetobacter baumannii(MIC 25 µM) as well as a modest activity against Pseudomonas aeruginosa.

4

The aim of this study was to improve the antibacterial and antibiofilm activity of fingolimod, and to study

structure-activity relationships. We designed and synthesised a library of 28 fingolimod derivatives, and we

evaluated their efficacy against S. aureus grown in planktonic/single cell and biofilms. The most active

derivatives were further tested on pre-formed S. aureus biofilms and against gram-negative bacteria A.baumannii and P. aeruginosa. Compared to fingolimod, seven derivatives were more effective against S. aureus,

while five other derivatives showed improved activity against P. aeruginosa and/or A. baumannii, with no

apparent change in cytotoxicity on human cells. The information gathered in this work offers a promising

starting point and can guide further optimization towards less cytotoxic and more potent antibacterial molecules. 

References

1 ) Ferri, M. et al. Antimicrobial resistance: A global emerging threat to public health systems. Crit. Rev. Food Sci. Nutr.

2017, 57 (13), 2857-2876.

2) Hutchings M.I. et al. Antibiotics: past, present and future. Curr. Opin. Microbiol. 2019, 51, 72–80.

3) Miró-Canturri, A. et al. Drug Repurposing for the Treatment of Bacterial and Fungal Infections. Front. Microbiol. 2019,

10, 41.

4) Gilbert-Girard, S. et al. Screening of FDA-Approved Drugs Using a 384-Well Plate-Based Biofilm Platform: The Case of

Fingolimod. Microorganisms 2020, 8 (11)

EFMC-ISMC | 439

Z036

LEAD OPTIMIZATION OF A SERIES OF

[1,2,4]TRIAZOLO[1,5-A]PYRIMIDINE DERIVATIVES AS POTENT,

SELECTIVE AND BRAIN PENETRANT PDE2A INHIBITORS

Adriana Ingrid Velter, Peter Buijnsters, Yves Van Roosbroeck, Gary Tresadern, Edgar Jacoby, Luc Ver

Donck, Marijke Somers, Ilse Lenaerts, Harrie Gijsen

Janssen Pharmaceutica, Turnhoutseweg 30, Beerse 2340, Belgium

Phosphodiesterase 2A (PDE2A) is a member of the human cyclic nucleotide phosphodiesterase families

PDE1-PDE11, responsible for the hydrolysis of intracellular second messengers cAMP and cGMP. PDE2A is a

dual cGMP/cAMP phosphodiesterase, expressed predominantly in the brain, particularly in the hippocampus,

cortex and striatum, which govern cognitive functions such as learning and memory. Moreover, cAMP and

cGMP are implicated in the early and late stages of memory consolidation. PDE2A inactivates the biological

effects of cAMP and cGMP, and inhibition of PDE2A would increase these cyclic nucleotides' levels. It has been

hypothesized that this increase would lead to improvement of memory functions by increasing neural plasticity.

Thus, selective PDE2Ais could be useful as therapeutic agents for CNS disorders where cognitive function is

impaired, such as Alzheimer's disease (AD).

We have recently described the hit-to-lead exploration of a series of [1,2,4]triazolo[1,5-a]pyrimidine derivatives

as selective PDE2A inhibitors,[i] using structure and FEP -based guided design. This poster will present the lead

optimization efforts in this series, such as our strategy to improve metabolic stability and brain penetration while

maintaining the high PDE2 activity and selectivity.

References

i) Tresadern, G.; Velter, I.; Trabanco, A.A.; Van De Keybus F.; Macdonald, G. J. ; Somers, M. V.F.; Vanhoof, G.; Leonard,

P.M.; Lamers, M. B.A.C.; Van Roosbroeck, Y. E. M. ; Buijnsters. P. [1,2,4]triazolo[1,5-a]pyrimidine PDE2A Inhibitors:

Structure and Free Energy Perturbation- Guided exploration J. Med. Chem. 2020, 63, 12887.

440 | EFMC-ISMC

Z037

BENZIMIDAZOLE DERIVATIVES: SYNTHESIS AND

LEISHMANICIDAL ACTIVITY EVALUATION

Nerea Escala (1), Laura Pineda (2), Carmenza Spadafora (2), Esther del Olmo (1)

1) Department of Pharmaceutical Sciences; Pharmaceutical Chemistry. Faculty of Pharmacy, CIETUS, IBSAL, University ofSalamanca. 37007-Salamanca, Spain

2) Institute of Scientific Research and High Technology Services -INDICASAT AIP-, Ciudad del Saber, Panama, Rep. ofPanama

Leishmaniasis is a NTD caused by kinetoplastid protozoans of the genus Leishmania spp., which comprises over

20 species

1

, the disease is widespread and endemic in 98 countries. The disease shows different clinical

manifestations including cutaneous leishmaniasis (CL, self-healing skin ulcers), mucosal leishmaniasis (ML),

and the potentially fatal visceral leishmaniasis (VL), also known as kala-azar. VL is caused by L. donovani or L.

infantum spp, and infect 200,000–400,000 people annually, mostly in Brazil, East Africa and in India, causing

more than 20,000 deaths.

1

The available chemotherapeutic treatments are reduced to a few drugs (antimonial compounds, amphotericin B,

pentamidine, paromomycin and miltefosine) whose effectiveness is limited by rising drug resistance/therapeutic

failure, and noxious side-effects. Therefore, new therapeutic hits are needed. The aim of this study was to

synthetized different 2-aminobenzimidazole derivatives and test their leishmanicidal activity against L.donovani.

The benzimidazole derivatives were synthesized in two steps, Fig. 1. The 2-aminobenzimidazole intermediates

were obtained by the reaction of 4-substituted phenylendiamines with cyanogen bromide in methanol (v/v 50%)

at room temperature, providing the intermediates Ia and Ib in 90% yield.

2

Intermediates Ia and Ib were treated

with different carboxylic acids using 1,1-carbonildiimidazole (CDI) as coupling reagent to obtain compounds 1-

15 with yields 40-75%.

3

Compounds 1-15 were tested in vitro against L. donovani (LD-1S/MHOM/SD/00-1S strain) axenic amastigotes,

using as reference drug B amphotericin. The growth and survival of parasites was measured by using a

PicoGreen fluorochrome. Fifteen compounds were obtained and their in vitro leishmanicidal activity measured

on L. donovani. Six out of the fifteen compounds showed a growth inhibition higher than 60 % at 10 µM, for

those compounds the IC

50

was calculated. Compounds 1, 4 and 5 showed good IC

50

values of 1.9, 0.9 and 1.3

µM, respectively. Cytotoxicity assay in Vero cells of compounds 1, 4 and 5 are being carried out, to obtain the

selectivity indexes. Six of the fifteen obtained compounds showed good leishmanicidal activity, and three of

them showed IC

50

values of 1.9, 0.9 and 1.3 µM.

References

1) Leishmaniasis [World Health Organization]. Visited on June, 2021. Website:

https://www.who.int/news-room/fact-sheets/detail/leishmaniasis

2) Bansal, Y., Kaur, M., Silakari, O. (2015). Benzimidazoleeibuprofen/mesalamine conjugates: Potentialcandidates for

multifactorial diseases. European Journal of Medicinal Chemistry, 89, 671-682. doi: 10.1016/j.ejmech.2014.10.081

3) Escala, N., Valderas-García, E., Álvarez-Bardón, M., Castilla-Gómez de Agüero, V., Escarcena, R., López-Pérez, J.L.,

Rojo-Vázquez, F.A., San Feliciano, A., Balaña-Fouce, R., Mártinez-Valladares, M., del Olmo, E. (2020). Synthesis,

bioevaluation and docking studies of some 2-phenyl-1H-benzimidazole derivatives as anthelmintic agents against the

nematode Teladorsagia circumcincta. European Journal of Medicinal Chemistry, 208, 112554. doi:

10.1016/j.ejmech.2020.112554

EFMC-ISMC | 441

Z038

DESIGN, SYNTHESIS, AND ALDOSE REDUCTASE ACTIVITY OF

SOME NOVEL ARYL CARBOXYLIC ACID DERIVATIVES BEARING

2-SUBSTITUTED-6-OXO-PYRIDAZINONE MOIETY

Mevlüt Akdağ (1), Azime Berna Özçelik (1), Yeliz Demir (2), Şükrü Beydemir (3,4)

1) Gazi University, Faculty of Pharmacy, Department of Pharmaceutical Chemistry, Ankara, Turkey.2) Ardahan University, Nihat Delibalta Göle Vocational High School, Department of Pharmacy Services, Ardahan , Turkey.

3) Anadolu University, Faculty of Pharmacy, Department of Biochemistry, Eskişehir, Turkey4) Bilecik Şeyh Edebali University, The Rectorate of Bilecik Şeyh Edebali University, Bilecik,Turkey.

The polyol pathway is a two-step metabolic pathway in which glucose is reduced to sorbitol and then converted

to fructose. This pathway is associated with diabetic complications and other inflamatory patologies. Aldose

reductase (AR), first and rate limiting enzyme of polyol pathway, is an enzyme which belongs to aldo-keto

reductase super family. The enzyme is a drug target for treating diabetic complications (1, 2). The enzyme has an

active site covered with aromatic amino acids and thus, aryl moieties of inhibitor compounds are important to get

favourable interactions (3). Also, carboxylic acids are potent inhibitors of AR for they are able to interact

strongly with recognition site of AR (4, 5). Moreover, epalrestat which is the only inhibitor in the market is a

carboxylic acid derivative (6). Pyridazinone is a privileged structure found in some potent inhibitors of AR (7,

8). In this study, we designed some aryl carboxylic acid derivatives bearing 2-substituted-6-oxo-pyridazinone

moiety and evaluated the inhibitor activity of these derivatives against AR. Synthesized compounds were found

more active than epalrestat.  As a result of this study, these compounds were showed as novel AR enzyme

inhibitors.

Acknowlegment

Mevlüt Akdağ is a fellow of YÖK 100/2000 Scholarship Program.

 

This project was funded by Scientific Research Department of Gazi University (Gazi BAP) under grant No.

02/2020-15.

References

1) Petrash, J. M. (2004). All in the family: aldose reductase and closely related aldo-keto reductases. Cellular and Molecular

Life Sciences CMLS, 61(7-8), 737-749.

2) Chatzopoulou, M., Pegklidou, K., Papastavrou, N., & Demopoulos, V. J. (2013). Development of aldose reductase

inhibitors for the treatment of inflammatory disorders. Expert opinion on drug discovery, 8(11), 1365-1380.

3) El-Kabbani, O., Ruiz, F., Darmanin, C., & Chung, R. T. (2004). Aldose reductase structures: implications for mechanism

and inhibition. Cellular and Molecular Life Sciences CMLS, 61(7-8), 750-762.

4) Maccari, R.,& Ottana, R. (2015). Targeting aldose reductase for the treatment of diabetes complications and inflammatory

diseases: new insights and future directions. Journal of medicinal chemistry, 58(5), 2047-2067.

5) Koutsopoulos, K., Lavrentaki, V., Antoniou, I., Kousaxidis, A., Lefkopoulou, M., Tsantili-Kakoulidou, A., Kavacikova,

L., Stefek, M., & Nicolaou, I. (2020). Design synthesis and evaluation of novel aldose reductase inhibitors: The case of

indolyl–sulfonyl–phenols. Bioorganic & Medicinal Chemistry, 115575.

6) Iyer, S., Sam, F. S., DiPrimio, N., Preston, G., Verheijen, J., Murthy, K., Parton, Z., Tsang, H., Lao, J., Morova, E., &

Perlstein, E. O. (2019). Repurposing the aldose reductase inhibitor and diabetic neuropathy drug epalrestat for the congenital

disorder of glycosylation PMM2-CDG. Disease models & mechanisms, 12(11).

7) Oates, Peter J. "Aldose reductase inhibitors and diabetic kidney disease." Curr Opin Investig Drugs 11.4 (2010): 402-17.

8) Yaseen, R., Pushpalatha, H., Reddy, G. B., Ismael, A., Ahmed, A., Dheyaa, A., Ovais, S., Rathore, P., Samim, M.,

AKthar, M., Shafi, S., Singh., S., Javed, K., & Sharma, K. (2016). Design and synthesis of pyridazinone-substituted

benzenesulphonylurea derivatives as anti-hyperglycaemic agents and inhibitors of aldose reductase–an enzyme embroiled in

diabetic complications. Journal of enzyme inhibition and medicinal chemistry, 31(6), 1415-1427.

442 | EFMC-ISMC

Z039

COMBINED PEPTIDE AND SMALL-MOLECULE APPROACH

TOWARDS NON-ACIDIC TETRAHYDROISOQUINOLINE (THIQ)

INHIBITORS OF THE KEAP1/NRF2 INTERACTION

Fezzardi Paola (1), Biancofiore Ilaria (1), Ferrigno Federica (1), Torrente Esther (1), Colarusso Stefania

(1), Patsilinakos Alexandros (1), Kempf Georg (3), Augustin Martin (3), Steinbacher Stefan (3), Pacifici

Robert (2), Munoz-Sanjuan Ignacio (2), Park Larry (2), Bresciani Alberto (1), Dominguez Celia (2),

Bianchi Elisabetta (1), Toledo Sherman Letitia (2), Ontoria Jesus M. (1)

1) IRBM S.p.A., Departments of Chemistry and Biology, Via Pontina km 30,600, 00071 Pomezia, Rome, Italy2) CHDI Management/CHDI Foundation, 6080 Center Drive, Los Angeles, CA, USA

3) Proteros Biostructures GmbH, Bunsensraβe 7a, 82152 Planegg, Germany

Oxidative stresses exacerbate many diseases, including cancer and neurodegenerative disorders such as

Parkinson’s and Huntington’s disease. A crucial regulator of cell defense mechanisms against oxidative stress is

the nuclear factor erythroid-derived 2-like 2 (NRF2), a transcription factor which activates the transcription of

several antioxidants and detoxifying proteins by binding to the antioxidant response element (ARE) present in

their promoters. Under normal condition, NRF2 intracellular levels are kept low by the interaction with its main

negative regulator, Kelch-like ECH-associated protein 1 (KEAP1). Following oxidative stress, KEAP1 is

oxidized at cysteine residues, resulting in KEAP1 inactivation, NRF2 stabilization and translocation into the

nucleus where it stimulates the transcription of the NRF2-responsive genes.Thus, compounds that could increase

the amount of free NRF2 by disrupting the interaction KEAP1 may have a therapeutic potential for oxidative

stress-related diseases. Many small molecules known to be potent activators of ARE-related genes, such as

sulforaphane, are electrophiles and act as covalent indirect inhibitors of the KEAP1-NRF2 complex. The

potential toxic side effects of this approach are well documented. Several non-covalent small molecules have

been reported to be KEAP1 inhibitors, but most of them present high MW and an acidic moiety, due to the large

size of the pocket and the presence of many Arg residues in the Kelch domain, leading to a low cellular activity.

We describe the generation of new non-acidic tetrahydroisoquinolines (THIQs) that show improved binding to

KEAP1 by establishing new interactions at the P3 and P2 pockets. The initial identification of 5-substituted

THIQs with improved potency, thanks to additional H-bond interactions in the P3 pocket of KEAP1 lead to the

most potent THIQ KEAP1 binder known so far. A peptide library was prepared based on the known KEAP1

inhibitor, the LDEETGEFL nona-peptide, designed to specifically target the P2 region of the protein, with

non-acidic residues. Its screening allowed the identification of novel moieties that provided additional binding at

the P2 pocket. The introduction of these favourable moieties in our most active THIQ compounds led to

non-acidic analogs that show improved binding with KEAP1. The newly established interactions in the P3 and

P2 pockets, counterbalance the loss of the salt bridge between the former carboxylic acids with Arg415.

Crystallographic evidence of the novel interaction mode for the substituents at position 5 of the THIQ scaffold in

the P3 pocket and the non-acidic moiety in the P2 pocket will be provided.

EFMC-ISMC | 443

Z040

STRUCTURAL INSIGHTS INTO NATURAL STILBENOIDS AS

POTENTIAL TRPA1 MODULATORS

Atefeh Saadabadi (1,2), Outi Salo-Ahen (1), Patrik C. Eklund (2)

1) Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland2) Laboratory of Molecular Science and Engineering, Åbo Akademi University, Turku, Finland

Background: Stilbenoids comprise a class of plant polyphenols that have gained intense interest for their

diverse biological activities, such as anti-diabetic, anti-inflammatory and anti-cancer properties [1]. Recently,

studies showed pinosylvin, a natural stilbenoid, can inhibit TRPA1 (Transient Receptor Potential Ankyrin 1)

activity in vitro and in vivo [2]. This ion-channel is found in the pain-detecting sensory nerves [3] and activates

pain and inflammation pathways [4]. However, stilbenoids’ molecular mechanism (particularly resveratrol) in

many pathological conditions was almost revealed in recent years, their structural insight into transient TRP

family specifically TRPA1 has remained a mystery. The 3D structures of human TRPA1 provided valuable

information for structure-based drug design efforts. In essence, natural stilbenoids could serve as hits/lead

compounds for developing novel TRPA1 modulators. This study aimed to find a putative binding site for natural

stilbenoids (scheme 1) in TRPA1 to help us design more potent and selective TRPA1 modulators.

Scheme 1: Chemical structures of natural stilbenoids

Methods: The crystal structure of TRPA1 [PDB ID: 3J9P] was taken from Protein Data Bank. Molecular

dynamics simulation with the AMBER 16 simulation package was performed to create a representative ensemble

of binding site conformations for TRPA1. Known TRPA1 antagonists (A-967079 and HC-030031) were used as

references to select the best receptor conformations and best-binding ligands. GLIDE program (Schrödinger)

was conducted for the molecular docking of these compounds. Free energy of binding was calculated for the

ligand-receptor complexes with the Prime/MM-GBSA module of Maestro (Schrödinger). Pinosylvin, pinosylvin

monomethyl ether (PME), were isolated from pine knotwood and stilbenoid glycosides from Norway spruce

inner bark. The raw extracts were further purified by chromatography to yield pure compounds (purity > 95%).

The biological activities of these compounds were evaluated by FLIPR assay in hTRPA1-transfected HEK293

cells.

Figure 1. Pinosylvin (purple stick) in the A-967079 (left) and HC-030031 (right) binding sites of

TRPA1(deepteal ribbon). Hydrogen bond and pi-pi interactions are visualized in dashed yellow and red lines,

respectively.

Results: Molecular docking studies were executed at different conformers of TRPA1 using reference

compounds. Then natural stilbenoids were docked in the best receptor conformations in known active sites of

A-967079 and HC-030031. Their affinity to these binding sites was evaluated based on their interactions with

the essential residues and free energy of binding. Natural compounds except isorhapontin were in the same pose

of A-967079 and could make a hydrogen bond with Ser873 or Thr874 that get stable by making π -π interaction

between their phenyl rings and Phe877 or Phe909 or both. Although resveratrol, pinosylvin, and PME made a

hydrogen bond with Asn855, they were not in the same pose as HC-030031 (Figure 1). It seems bulky molecules

like astringin and isorhapontin imitated HC-030031’s pose and interactions more competently. The in vitro

results disclosed that our compounds could effectively interact with the TRPA1 channel and activate this channel

with EC50 values ranging from 6.1 to 13.3 µM except for stilbenoid glucosides that showed neither activity nor

inhibitory effect on this channel (Table 1).

Table 1: Molecular docking and biological results of natural stilbenoids in TRPA1 channel 

Conclusion: The docking studies revealed that natural stilbenoids have a higher affinity to the active site of

A-967079 compare to the HC-030031 binding site. In vitro studies also disclosed the natural compounds except

stilbenoid glucosides could inhibit TRPA1 channel with a desensitization mechanism. The stilbenoid glucosides

did not show any activity on human TRPA1 due to their bulky structures that prevented them from accessing the

active site. As the stilbenoid glucosides can be deglycosylated to the aglycones in vivo, they can be considered

sources of stilbenoids with higher solubility and the same biological activities.

References

1) Akinwumi, B. C.; Bordun, K. M.; Anderson, H. D. Biological Activities of Stilbenoids. Int. J. Mol. Sci. 2018, 19, 792.

2) Moilanen L. J.; Hämäläinen, M.; Lehtimäki, L.; Nieminen, R. M.; Muraki, K.; Moilanen, E. Pinosylvin Inhibits

TRPA1-Induced Calcium Influx In Vitro and TRPA1-Mediated Acute Paw Inflammation In Vivo. Basic & Clinical

Pharmacology & Toxicology. 2016, 118, 238-242.

3) Ramsey, I. S.; Delling, M.; Clapham, D. E. An introduction to TRP channels. Annu. Rev. Physiol. 2006, 68, 619–647.

4) Julius, D. TRP channels and pain. Annu. Rev. Cell Dev. Biol. 2013, 29, 355–384.

444 | EFMC-ISMC

Z041

ANTIPLASMODIUM ACTIVITY OF

PYRIMIDINO[1,2-A]BENZIMIDAZOLE AND

IMIDAZO[1,2-A]PYRIMIDINE DERIVATIVES

Radwan Alnajjar (1), Gurminder Kaur (1), Kelly Chibale (2,3)

1) Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa2) Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch 7701, South Africa.

3) South African Medical Research Council Drug Discovery and Development Research Unit, University of Cape Town,Rondebosch 7701, South Africa

In an attempt to improve physicochemical properties of previously identified antimalarial

pyrido[1,2-a]benzimidazole (PBI) derivatives

1-2

, Two novel series of pyrimidino[1,2-a]benzimidazole (I

1-33

) and

imidazo[1,2-a]pyrimidine (II

1-10

) were synthesized and evaluated for in vitro antiplasmodium activity against

chloroquine-sensitive (NF54) strain of the human malaria parasite Plasmodium falciparum(Pf). The

pyrimidino[1,2-a] benzimidazole series show good potency (IC

50

PfNF54 = 0.17 – 5.95 µM) with the frontrunner

derivative (I

1

) possessing better activity (IC

50

0.14 µM) and selectivity index (28.47) compared to the parent PBI

derivative (IC

50

0.32 µM, SI=4.19). This derivative also showed improved solubility over the parent PBI

molecule. On the other hand, the imidazo[1,2-a]pyrimidine series was found to be less active (IC

50

PfNF54 =

1.09 – 5.74 µM) except derivative (II

10

), which showed antiplasmodium activity with an  IC

50

value of 0.13 µM.

Beta hematin inhibition (βHI) assay data suggests that, like PBIs, inhibition of hemozoin formation might

contribute to the mechanism of action.

References

1) Ndakala, A. J.; Gessner, R. K.; Gitari, P. W.; October, N.; White, K. L.; Hudson, A.; Fakorede, F.; Shackleford, D. M.;

Kaiser, M.; Yeates, C.; Charman, S. A.; Chibale, K., Antimalarial Pyrido[1,2-a]benzimidazoles. Journal of Medicinal

Chemistry 2011, 54 (13), 4581-4589.

2) Singh, K.; Okombo, J.; Brunschwig, C.; Ndubi, F.; Barnard, L.; Wilkinson, C.; Njogu, P. M.; Njoroge, M.; Laing, L.;

Machado, M.; Prudêncio, M.; Reader, J.; Botha, M.; Nondaba, S.; Birkholtz, L.-M.; Lauterbach, S.; Churchyard, A.; Coetzer,

T. L.; Burrows, J. N.; Yeates, C.; Denti, P.; Wiesner, L.; Egan, T. J.; Wittlin, S.; Chibale, K., Antimalarial

Pyrido[1,2-a]benzimidazoles: Lead Optimization, Parasite Life Cycle Stage Profile, Mechanistic Evaluation, Killing Kinetics,

and in Vivo Oral Efficacy in a Mouse Model. Journal of Medicinal Chemistry 2017, 60 (4), 1432-1448.

EFMC-ISMC | 445

Z042

SYNTHESIS OF NOVEL BENZOXAZOLINONE-HYDRAZONE

DERIVATIVES AS POTENT AND SELECTIVE MAO A/B INHIBITORS

FOR TREATMENT OF NEURODEGENERATIVE DISEASES

Hayrunnisa Tasci (1), Begum Nurpelin Saglik (2), Yusuf Ozkay (2), Birsen Tozkoparan (1), Nesrin

Gokhan Kelekci (1)

1) Hacettepe University Faculty of Pharmacy Dept of Pharmaceutical Chemistry, 06100 Ankara-Turkey2) Anadolu University Faculty of Pharmacy Dept of Pharmaceutical Chemistry, 44280 Eskisehir-Turkey

Increased oxidation activity of MAO enzymes cause neurodegeneration and diseases that association with this

degeneration (1,2). As a results of these information, an approach for treating neurodegenerative and

stress-induced disorders is developed including selective MAO inhibition. According to this, MAO B inhibition

is targeted in Parkinson’s disease and Alzheimer’disease that decrase dopamin concentration and

plaque-associated astrocytes in the brains (3) Otherwise, depressive disorders are associated with elevated

MAO-A level and it is aimed to inhibition of MAO-A and the consequent ability to counter the decrement in

brain noradrenaline (NE), dopamine (DA), and especially serotonin (5-HT) levels during the depression (4,5).

Considering the therapeutical potential of MAO inhibitors, different chemical structures have been designed and

evaluated in terms of MAO inhibition. Hydrazine derivatives constitute the most important group of irreversible

MAO inhibitors. Iproniazid, originally designed as an antitubercular drug, is considered representative of this

group and has been released as an antidepressant (6). Presence of amino or imino group that playing a role in the

interaction of the MAO enzyme with its active site is common structure in hydrazone derivative inhibitors such

as iproniazid. On going research, It is aimed to obtain compounds that reversible, selective, more potent and

have less side effects.

In this study, some new N'-arylidene-3-(5-chloro-2-oxobenzo[d]oxazol-3(2H)-yl)propanehydrazide derivatives

(Figure 1) synthesised using 3-(5-chloro-2-benzoxazolinone)propanonic acid hydrazide and various aldehydes

and  their structures were confirmed using IR,

1

H-NMR, mass and elemental analysis. These compounds hMAO

inhibition potential has been evaluated in vitro tests. The compounds were investigated for their ability to inhibit

hMAO isoforms by in-vitro tests. Molecular docking studies were done to provide insights into enzyme-inhibitor

interactions and a rationale for the observed inhibition towards monoamine oxidases. In vitro activity results

revealed that synthesised compounds inhibited MAO A by 39-69 % and MAO B by 45-94 % in 10

-3

M

concentration and exhibite higher affinity for MAO B than MAO A. 

Acknowledgments Funding for this project was provided by the Hacettepe University, Scientific Research

Projects Coordination Unit (Project number: THD-2020-18624).

References

1) Binda, C.; Hubalek, F.; Li, M.; Edmondson, D. E.; Mattevi, A. FEBS Lett. 2004, 564, 225−228.

2) Binda, C., Newton-Vinson, P., Hubalek, F., Edmondson, D. E., & Mattevi, A. Nat. Struct. Biol., 2002, 9(1), 22-26,

3) Parambi, Della G. T; Combinatorial Chemistry & High Throughput Screening, 2020, 23(9), 847-861.

4) Salgin-Goksen U, Telli G, Erikci A, Dedecengiz E, Tel BC, Kaynak FB, Yelekci K, Ucar G, Gokhan-Kelekci N; J. Med.

Chem., 2021, 64, 4, 1989-2009.

5) Manzoor, S; Hoda, N; European J. Med. Chem., 2020, 206, 112787.

6) Cesura, M. A., Pletscher, A. Prog. Drug. Res. 1992, 38, 171-297.

446 | EFMC-ISMC

NOTES

EFMC-ISMC | 447

Index of Abstracts

448 | EFMC-ISMC

ABAS PRADES Sonia RATIONALLY DERIVED INHIBITORS OF HEPATITIS C VIRUS P7 CHANNEL

Z024

ABDELSALAM Mohamed STRUCTURE-BASED DESIGN AND SYNTHESIS OF SELECTIVE CLASS I HDAC INHIBITORS AS POTENTIAL ANTICANCER AGENTS

T001

AGBABA Danica SEROTONIN AND IMIDAZOLINE RECEPTOR LIGANDS, AND RELATED COMPOUNDS - MODELING OF MIXED-MODE INTERMOLECULAR INTERACTIONS IN CHROMATOGRAPHIC SYSTEM

Z006

AKDAG Mevlüt DESIGN, SYNTHESIS, AND ALDOSE REDUCTASE ACTIVITY OF SOME NOVEL ARYL CARBOXYLIC ACID DERIVATIVES BEARING 2-SUBSTITUTED-6-OXO-PYRIDAZINONE MOIETY

Z038

ALCOLEA Veronica DESIGN AND SYNTHESIS OF NOVEL SELENOESTERS AND SELENOCYANATES FOR THE TREATMENT OF CHAGAS DISEASE

O002

ALNAJJAR Radwn ANTIPLASMODIUM ACTIVITY OF PYRIMIDINO[1,2-A]BENZIMIDAZOLE AND IMIDAZO[1,2-A]PYRIMIDINE DERIVATIVES

Z041

ANDERSON Niall INTEGRIN ANTAGONISTS FOR TREATMENT OF IDIOPATHIC PULMONARY FIBROSIS

PR003

ANDRADE Carolina H. TITLE OF TALK TO BE CONFIRMED

LE038

ANTONIJEVIC Mirjana DEVELOPMENT OF THE FIRST DUAL ACTIVATORS OF NEUROTROPHIN AND SEROTONIN 5-HT4 RECEPTORS, A PROMISING STRATEGY IN THE BATTLE AGAINST NEURODEGENERATIVE DISORDERS

X004

ARSENIYADIS Stellios INNOVATIVE DNA-BASED ASYMMETRIC CATALYSIS

LE033

ASTOLFI Andrea SMALL MOLECULE DEGRADERS TARGETING FOLDING INTERMEDIATES: THE PRION PROTEIN CASE STUDY

LE012

ATZ Kenneth Δ-QUANTUM MACHINE LEARNING FOR DRUG-LIKE MOLECULES WITH E(3)-EQUIVARIANT GRAPH NEURAL NETWORKS

M002

AVCI Ahmet SYNTHESIS, CHARACTERIZATION, AND IN VITRO CYTOTOXIC EVALUATION OF NOVEL THIAZOLO[3,2-b]-1,2,4-TRIAZOLE-6(5H)-ONE DERIVATES

Z011

AYE Yimon WITHIN OUR CONTROL? ILLUMINATING HOW EPHEMERAL ELECTROPHILES REWIRE CELL DECISION-MAKING PROCESSES

PR002

BAQI Younis EFFECTS OF DATE FRUIT EXTRACTS (PHOENIX DACTYLIFERA L.) ON PANCREATIC CANCER CELLS IN-VITRO

U004

BARAN Phil TRANSLATIONAL CHEMISTRY

PL003

BARCHERINI Valentina DEVELOPMENT OF p53 ACTIVATORS TO TARGET COLORECTAL CANCER: HIT OPTIMIZATION OF TRYPTOPHANOL-DERIVED ISOINDOLINONES

M003

BARNIOL-XICOTA Marta NOVEL ACTIVITY-BASED PROBES AND NANOMOLAR PEPTIDOMIMETIC INHIBITORS AGAINST THE MAIN PROTEASE OF SARS‐CoV‐2

C001

BARRECA Marilia EVALUATION OF [1,2]OXAZOLE DERIVATIVES IN LYMPHOMA MODELS

T002

BASU Sujay DISCOVERY OF POTENT AND SELECTIVE A2A ANTAGONISTS WITH EFFICACY IN ANIMAL MODELS OF PARKINSON'S DISEASE AND DEPRESSION

Q009

BAVO Francesco LOCKED ANALOGUES OF L-ISOSERINE AS POTENTIAL SUBTYPE-SELECTIVE GAT3 INHIBITORS

Q001

BEC Anja SYNTHESIS AND BIOLOGICAL ACTIVITY OF NOVEL N- SUBSTITUTED BENZIMIDAZOLE DERIVED ACRYLONITRILES

Z004

BECHTLER Clément DESIGN, SYNTHESIS AND EVALUATION OF A PEPTIDE CONJUGATE TO PROTECT BIOMATERIALS FROM UNDESIRED IMMUNE ATTACK

Z007

BENDER Andreas ARTIFICIAL INTELLIGENCE IN CHEMICAL BIOLOGY AND DRUG DISCOVERY – DATA, APPLICATIONS, AND ILLUSIONS

LE059

BENEK Ondrej BENZOTHIAZOLYLUREA-BASED 17-HSD10 INHIBITORS - DESIGN, SYNTHESIS, IN VITRO AND IN VIVO EVALUATION

C002

EFMC-ISMC | 449

BERGANT LOBODA Kaja DESIGN AND SYNTHESIS OF 3,5-SUBSTITUTED 1,2,4-OXADIAZOLES AS CATALYTIC INHIBITORS OF HUMAN DNA TOPOISOMERASE IIA

T003

BERISHVILI Vladimir BEYOND BINARY: SCALED MOLECULAR FINGERPRINTS FOR MAXIMUM DIVERSITY PICKING

H001

BERMEO Rafael TARGETING A PATHOGENIC LECTIN: DESIGN, SYNTHESIS AND EVALUATION OF BC2L-C INHIBITORS

R001

BERNARDES Gonçalo TRANSLATIONAL CHEMICAL BIOLOGY

LE001

BEUS Maja NOVEL ANTHRANILIC ACID DERIVATIVES AS PROMISING ANTI-TB AGENTS

Z023

BILENSOY Erem OMICS-BASED DEVELOPMENT OF NANOMEDICINES FOR SAFE AND EFFECTIVE DRUG DELIVERY IN CANCER

LE077

BILLAMBOZ Muriel PYROGLUTAMIC ACID AS A NATURAL PLATFORM TOWARDS NEW ANTIMICROBIAL AGENTS

L002

BLICHARZ Klaudia NEUTRAL 5-HT6 RECEPTOR ANTAGONISTS WITH GLIOPROTECTIVE AND COGNITIVE-ENHANCING PROPERTIES: AN IMPACT OF STRUCTURAL MODIFICATIONS ON VARIOUS CONFORMATIONAL STATES OF THE RECEPTOR

M005

BOCEK Ida SYNTHESIS AND ANTIOXIDATIVE ACTIVITY OF NOVEL ACRYLONITRILE IMIDAZO[4,5-b] PYRIDINES

Z003

BOLJE Aljosa STRUCTURAL CHARACTERIZATION AND STABILITY OF PROTEINS IN SOLID FORMS

M006

BORSARI Chiara PRECISION DRUGS: A COVALENT STRATEGY TO MINIMIZE SIDE EFFECTS OF PI3K INHIBITOR CANCER THERAPY

T004

BOSC Damien ONE-POT PROCEDURE FOR THE PREPARATION OF 1,5-DISUBSTITUTED 1,2,3-TRIAZOLES BY IN-SITU GENERATION OF AZIDES

L003

BOSCHI Donatella INHIBITION OF THE ACTIVITY OF THE TRANSMEMBRANE FORM OF CHLORIDE INTRACELLULAR CHANNEL 1 (tmCLIC1): A NOVEL PHARMACOLOGICAL TOOL IN MULTIPLE TARGETED THERAPY FOR GLIOBLASTOMA TREATMENT

T005

BOSCO Michael MOLECULAR PROBE TO ISOLATE DLODP: AN ENZYME INVOLVED IN TYPE I CONGENITAL DISORDERS OF GLYCOSYLATION (CDG-I)

C004

BOZZOLA Tiago DESIGN, SYNTHESIS AND EVALUATION OF SIALIC ACID DERIVATIVES THAT INHIBIT SIAT SYMPORTERS AND ATTENUATE BACTERIAL GROWTH

A002

BRANDAO Pedro NEW TRYPTANTHRIN-BASED PETASIS ADDUCTS - DESIGN, SYNTHESIS AND BIOLOGICAL ACTIVITY EVALUATION

L004

BRINER Karin CREATING THE NEXT GENERATION OF THERAPEUTICS - MEDICINAL CHEMISTRY UNLEASHED

PL001

BROUGH Steve NEW BRAIN PENATRANT COMPOUNDS IN ADVANCED STUDIES: MULTIPOTENCY SCREENING AND EVALUATION AGAINST PARKINSON'S DISEASE

Q002

BROUGH Steve FROM INNOVATION TO THE MARKET: ADDING VALUE TO THE COMPOUNDS FROM ACADEMIC RESEARCH AND TEACHING

L030

BRÖNSTRUP Mark ASSAULT, SIEGE OR TROJAN HORSE STRATEGY: USE OF NATURAL PRODUCTS TO FIGHT BACTERIAL INFECTIONS

R002

BULLER Rebecca UTILIZATION OF IRON/ALPHA-KETOGLUTARATE-DEPENDENT ENZYMES FOR STEREOCONTROLLED C-H FUNCTIONALIZATION

LE098

CAI Xingguang A STUDY WITH PEPTIDE DENDRIMERS REVEALS AN EXTREME pH DEPENDENCE OF ANTIBIOTIC ACTIVITY ABOVE pH 7.4

R003

CAI Xingguang NON-HEMOLYTIC ANTIMICROBIAL PEPTIDES DESIGNED BY MACHINE LEARNING

R004

CALLIS Timothy SCAFFOLD HOPPING FOR THE DIVERSIFICATION OF SMALL MOLECULES TARGETING PROTEIN AGGREGATION

M007

CAMPANICO André AZAAURONES AS NOVEL CHEMOTYPES AGAINST MYCOBACTERIUM TUBERCULOSIS: SAR, ADME PROFILING AND PHOTO-SWITCHING PROPERTIES

LE015

450 | EFMC-ISMC

CAMPBELL Emma SPLICE-SWITCHING SMALL MOLECULES AS INDUCERS OF APOPTOSIS

LE069

CANABAL FALCON Rafael RIGIDIFIED SHIKIMIC ACID DERIVATIVES TO IMPRINT INHIBITOR EFFICACY AGAINST SHIKIMATE KINASE ENZYME - A TARGET FOR ANTIBIOTIC DRUG DISCOVERY

R005

CAO Jun SYNTHESIS OF HEPTOSE MIMETICS AS INHIBITORS OF BACTERIAL ENZYMES OR MODULATORS OF INFLAMMATION

R006

CARREIRA Erick M. RECENT DEVELOPMENTS IN STRATEGIES AND TACTICS TOWARDS COMPLEX SECONDARY METABOLITES

LE032

CARREIRA Monica DESIGN AND SYNTHESIS OF LINKERS FOR GLYCAN-COATED GOLD NANOPARTICLES USED IN DRUG DISCOVERY

L005

CARREL Aline SYNTHESIS OF NEW BUILDING BLOCKS FROM THE CHEMICAL UNIVERSE DATABASE GDB

LE035

CARVALHAL Francisca DEVELOPMENT OF INNOVATIVE MICROARRAYS TO DETECT INTERACTION OF SMALL MOLECULES WITH ZIKA VIRUS STRUCTURAL PROTEINS

O005

CASTALDI Paola MECHANISTIC INSIGHTS OF A CDK9 INHIBITOR VIA ORTHOGONAL PROTEOMICS METHODS

LE092

CECCHINI Carlotta TARGETING HIF-2 IN CLEAR CELL RENAL CELL CARCINOMA WITH PROTAC TECHNOLOGY

M008

CECERE Giuseppe DISCOVERY OF POTENT SELECTIVE GABAA ALPHA5 POSITIVE ALLOSTERIC MODULATORS (PAMS) FOR THE TREATMENT OF NEUROLOGICAL DISORDERS

PR001

CERADINI Davide PROCESS OPTIMIZATION OF THE SYNTHESIS OF UAMC-00050, A NOVEL uPA INHIBITOR

M009

CHALUPNIK Paulina N1-SUBSTITUTED QUINOXALINE-2,3-DIONES AS LIGANDS FOR GLUTAMATE KAINATE RECEPTORS

Z020

CHRIENOVA Zofia COMBINATION OF MEMANTINE AND 6-CHLOROTACRINE AS NOVEL MULTI-TARGET COMPOUND AGAINST ALZHEIMER’S DISEASE

M010

CHUANG Grace DISCOVERY OF AFICAMTEN (CK-274): A NOVEL, SMALL MOLECULE, CARDIAC MYOSIN INHIBITOR FOR THE POTENTIAL TREATMENT OF HYPERTROPHIC CARDIOMYOPATHIES (HCM)

LE022

CIESIOLKIEWICZ Agnieszka

DE NOVO DESIGN OF FOLDAMERIC MINI-PROTEINS AND THEIR USE AS PD-1/PD-L1 INTERACTION INHIBITORS

T006

CLEMENT Bernd NEW EXAMPLES FOR N-HYDROXYAMIDINE PRODRUGS

LE085

CLOZEL Jean-Paul MEDICINAL CHEMISTRY: MORE THAN EVER

PL002

CODERCH BOUE Claire STRUCTURAL INSIGHTS ON THE BINDING MODE OF A SERIES OF NOVEL DUAL INHIBITORS AGAINST CK2, HDAC1 AND HDAC6

T007

CODONY Sandra FROM THE DESIGN TO THE IN VIVO EVALUATION OF BENZOHOMOADAMANTANE-DERIVED SOLUBLE EPOXIDE HYDROLASE INHIBITORS FOR THE TREATMENT OF ACUTE PANCREATITIS

Z008

COIMBRA Judite A VIRTUAL SCREENING PROTOCOL TO IDENTIFY NOVEL INHIBITORS OF GLUTAMINYL CYCLASE

M011

COLCHON-PIERNA Esther PENICILLIN-BASED SULFONE B-LACTAMASE INHIBITORS TO RESTORE CARBAPENEM EFFICACY AGAINST SUPERBUGS

R007

COMEO Eleonora DEVELOPMENT AND APPLICATION OF SUBTYPE-SELECTIVE FLUORESCENT ANTAGONISTS FOR THE STUDY OF THE HUMAN A1AR ADENOSINE RECEPTOR IN LIVING CELLS

C005

CONTI Gabriele HIGH-AFFINITY GLYCOMIMETIC LIGANDS FOR HUMAN SIGLEC-8

A003

CORIO Alessandra DESIGN AND SYNTHESIS OF RED-SMU1 INTERACTION DISRUPTORS AS NEW ANTIVIRAL AGENTS AGAINST INFLUENZA A VIRUS

L007

COSTA Marta A NEW LEAD COMPOUND FOR TRIPLE NEGATIVE BREAST CANCER THERAPY

T008

COTESTA Simona DISCOVERY OF JDQ443 A STRUCTURALLY UNIQUE, HIGHLY POTENT, SELECTIVE AND ORALLY BIOAVAILABLE KRASG12C COVALENT INHIBITOR

LE054

EFMC-ISMC | 451

CRAMER Jonathan A GLYCOMIMETIC STRATEGY AGAINST DC-SIGN-MEDIATED CELL ENTRY AND DISSEMINATION OF SARS-COV-2

A004

CRNCEVIC Doris AMIDO-QUINUCLIDINE BASED QACs AS A NEW SOFT ANTIMICROBIAL AGENTS SUSCEPTIBLE TO PROTEASE DEGRADATION

Z017

CRONIN Leroy UNIVERSAL SYNTHESIS MACHINES AND CHEMPUTATION

LE045

CUNHA Eva STRUCTURE BASED DRUG DISCOVERY TARGETING HELICOBACTER PYLORI, USING CRYO-EM

LE020

CZECHTIZKY Werngard INHALED NEW MODALITIES IN RESPIRATORY DISEASE: PAST, PRESENT, FUTURE

LE048

DACK Kevin DISCOVERY OF AN ORAL, RO5 COMPLIANT, INTERLEUKIN 17A PROTEIN-PROTEIN INTERACTION MODULATOR FOR THE TREATMENT OF PSORIASIS AND OTHER INFLAMMATORY DISEASES

LE052

DAWSON Mike THERAPIES FOR GRAM-NEGATIVE BACTERIAL INFECTIONS: NEW APPROACHES AND FURTHER GENERATIONS OF EXISTING SERIES

LE063

DE JUAN ALBERDI Mario AZIDE/ALKENE 1,3-DIPOLAR CYCLOADDITION FOR THE SYNTESIS OF 1,4,5-TRISUBSTITUTED 1H-1,2,3-TRIAZOLES AS POTENTIAL ANTIPROLIFERATIVE AGENTS

L008

DE LA VEGA Jennifer ANTI-LEISHMANIAL ACTIVITY OF 2-AMINO ALKANOLS AND ALKYL-1,2-DIAMINES

M012

DEKKER Tom THE CONSTRUCTION OF A VERSATILE CHEMINFORMATICS WORKFLOW FOR THE DESIGN OF 3D FRAGMENT LIBRARIES

Z031

DEMURO Stefania DEVELOPMENT OF MULTITARGET-DIRECTED LIGANDS (MTDLS) PROTEIN KINASES INHIBITORS TO TACKLE CENTRAL NERVOUS SYSTEM (CNS)-RELATED DISORDERS.

M013

DEPREZ-POULAIN Rebecca

PROTEIN-TEMPLATED SYNTHESIS DELIVERS THE FIRST NANOMOLAR SELECTIVE ERAP2 INHIBITORS FOR IMMUNO-ONCOLOGY

M014

DERNOVSEK Jaka STRUCTURE-ACTIVITY RELATIONSHIPS OF BENZOTHIAZOLE-BASED HSP90 C-TERMINAL DOMAIN INHIBITORS

T009

DESIATKINA Oksana SYNTHESIS AND BIOLOGICAL ACTIVITY OF TRITHIOLATO-BRIDGED DINUCLEAR RUTHENIUM(II)-ARENE COMPLEXES WITH NUCLEIC BASES

L006

DESTRO Lorenza ALPHA-TRIAZOLYL-BORONIC ACIDS: A PROMISING SCAFFOLD AS PROTEIN KINASE (PK) INHIBITORS IN ACUTE MYELOID LEUKAEMIA (AML)

Z018

DI GERONIMO Bruno COMPUTATIONAL CLUSTERING METHODS IN CHEMICAL LIBRARIES: DEVELOPMENT OF A PROTEIN-PROTEIN INTERACTION INHIBITOR FOCUSED LIBRARY

Z005

DIGHE Satish Natha DISCOVERY OF NOVEL SMALL MOLECULE DNA GYRASE INHIBITORS BY STRUCTURE-BASED VIRTUAL SCREENING

M015

DING Sheng A CHEMICAL APPROACH TO CONTROLLING CELL FATE

LE040

DISTLER Joerg NOVEL MOLECULAR TARGETS FOR THE TREATMENT OF FIBROSIS

LE088

DONGIK Kwak DEVELOPMENT OF THE NOVEL HIF INHIBITOR LW1564 THAT TARGETS CANCER METABOLISM

T010

DRENICHEV Mikhail NOVEL SYNTHETIC NUCLEOSIDES AND EVALUATION OF THEIR BIOLOGICAL ACTIVITY

Z030

DREWEK Isalyne SYNTHESIS OF NEW HETEROCYCLIC DERIVATIVES BEARING BENZIMIDAZOLE MOIETIES AND EVALUATION OF THEIR BACTERIAL FLOCCULATION PROPERTIES

Z027

DRUMMOND Michael COMPUTATIONAL EVALUATION OF PROTAC-MEDIATED PROTEIN DEGRADATION: CASE STUDIES AND RECENT DEVELOPMENTS

M016

DUCA Margherita MULTIVALENT FUCOSYLATED INHIBITORS TARGETING BETA-PROPELLER LECTINS AS PROMISING ANTI-ADHESIVE DRUGS

A005

DUDUTIENE Virginija HYDROPHOBIC LIGAND AUGMENTATION AS A STRATEGY OF CARBONIC ANHYDRASE ISOZYME ACTIVE SITE EXPLORATION

M017

DUNKEL Petra MODIFICATIONS OF QUINOLINE PHOTOCAGES E001

452 | EFMC-ISMC

DURAES Fernando ANTITUMOR ACTIVITY AND FLUORESCENCE OF TETRACYCLIC THIOXANTHENES: NEW THERANOSTIC AGENTS?

T011

EBRU DIDEM COSAR SYNTHESIS AND ANTICHOLINESTERASE ACTIVITY OF NOVEL COUMARIN DERIVATIVES CONTAINING AMINOALCOHOL GROUP

L031

EL PHIL Radhia A NOVEL BACTERIAL THREE-HYBRID SYSTEM FOR TARGET IDENTIFICATION IN BACTERIA

LE094

ENGKVIST Ola TITLE OF TALK TO BE CONFIRMED

LE047

ERONEN Aleksi SYNTHESIS OF DIARYL HYDROXYL DICARBOXYLIC ACIDS FROM AMINO ACIDS

L009

ERRA SOLA Montse DISCOVERY OF NOVEL INHALED PI3Kδ INHIBITOR BY LUNG RETENTION OPTIMIZATION

LE049

ESCALA Nerea BENZIMIDAZOLE DERIVATIVES: SYNTHESIS AND LEISHMANICIDAL ACTIVITY EVALUATION

Z037

ESCOBAR MONTANO Felipe

PHORBOL DIESTERS INDUCE TGF RELEASE AND ADULT MOUSE NEUROGENESIS

B001

EZZANAD Abdellah UHPLC-HRMS-GUIDED DISCOVERY AND CHARACTERIZATION OF NEW 12-DEOXY-16-HYDROXYPHORBOL-13,16-DIESTERS FROM EUPHORBIA RESINIFERA: SELECTIVE PURIFICATION OF NEUROGENIC AGENTS

B002

FADEYI Niyi MAPPING CELL-CELL INTERACTIONS IN TUMOR MICROENVIRONMENT VIA PHOTOCATALYTIC PROXIMITY LABELING

LE014

FAJKIS-ZAJACZKOWSKA Nikola

A NEW APPROACH TO THE SYNTHESIS OF ZOLPIDEM AND FLUORINATED DERIVATIVES

L010

FEDOROWICZ Joanna SYNTHESIS, ANTIMICROBIAL ACTIVITY, AND DOCKING STUDIES OF NOVEL QUATERNARY AMMONIUM FLUOROQUINOLONE-BASED ANTIBACTERIAL AGENTS

M018

FELIX Rita QUENCHED ACTIVITY-BASED PROBES AS NEW CHEMICAL TOOLS TO ANALYSE RESISTANCE TO ANTIBIOTICS

LE031

FEZZARDI Paola COMBINED PEPTIDE AND SMALL-MOLECULE APPROACH TOWARDS NON-ACIDIC TETRAHYDROISOQUINOLINE (THIQ) INHIBITORS OF THE KEAP1/NRF2 INTERACTION

Z039

FIDALGO Lara DEVELOPMENT OF NOVEL NECROPTOSIS INHIBITORS: TARGETING RIPK1

Q003

FISCHER Christian DISCOVERY OF MRK-740, A FIRST-IN-CLASS, POTENT, SELECTIVE AND CELL ACTIVE PRDM9 CHEMICAL PROBE

LE030

FLOHR Stefanie LNP023: DISCOVERY AND SYNTHESIS OF A FIRST-IN-CLASS, ORAL FACTOR B INHIBITOR FOR TREATMENT OF RARE RENAL AND HEMATOLOGICAL DISEASES

LE074

FLORINDO Pedro ORGANORUTHENIUM(II) INHIBITORS OF HUMAN HEXOKINASE 2 AS HEPATOCELLULAR ANTICANCER AGENTS

A006

FUCHSS Thomas INSIGHTS TO THE DISCOVERY AND DESIGN OF ATAXIA TELANGIECTASIA MUTATED (ATM) KINASE INHIBITORS M3541 AND M4076 WITH STRONG ANTI-TUMOR EFFICACY IN COMBINATION THERAPY APPROACHES

LE053

FÜSSER Friederike FRAGMENT BASED DESIGN OF MYCOBACTERIAL THIOREDOXIN REDUCTASE INHIBITORS: FROM A FRAGMENT SCREENING TO NOVEL INHIBITORS

M020

GABELLIERI Emanuele DISCOVERY OF THE NEXT GENERATION TAU PET TRACER PI-2620 FOR THE ASSESSMENT OF TAU PATHOLOGY IN ALZHEIMER'S DISEASE AND OTHER TAUOPATHIES

D001

GARLATTI Laura IN SITU CLICK CHEMISTRY APPLIED TO BUNYAVIRALES: FROM CONVENTIONAL DRUG DESIGN TO ENZYMES ASSEMBLING THEIR OWN INHIBITORS

O004

GAVATHIOTIS Evripidis TARGETING ONCOGENIC BRAF USING A NOVEL ALLOSTERIC SITE

LE072

GAWALSKA Alicja DETERMINATION OF TRPA1 CHANNEL BINDING SITE FOR METHYLXANTHINE ANTAGONISTS USING MOLECULAR MODELING APPROACH

M021

GEHRINGER Matthias TARGETING THE PROTEIN KINASE S6K2 (p70S6K) WITH NON-CANONICAL ELECTROPHILIC WARHEADS

C009

GIANQUINTO Eleonora AN IN SILICO PIPELINE IDENTIFIES INHIBITORS WITH CROSS-CLASS ACTIVITY ON CLINICALLY RELEVANT SERINE- AND METALLO-B-LACTAMASES

R008

EFMC-ISMC | 453

GILBERT Kate KINETIC EVALUATION OF SULFUR(VI) FLUORIDE COVALENT WARHEADS TO ENABLE THE RAPID DEVELOPMENT OF THERAPEUTICS

M023

GIRARDI Benedetta 1,3-SUBSTITUTED GALACTOSIDES AS SELECTIVE MONOVALENT GALECTIN-8 LIGANDS

A007

GLYNN Daniel THE DESIGN, PREPARATION AND SCREENING OF BRPF1 DEGRADERS

M024

GOMEZ Elena DISCOVERY OF LAS200019, A NOVEL TOPICAL JAK INHIBITOR FOR THE TREATMENT OF INFLAMMATORY SKIN DISEASES

LE083

GONZALEZ Simon SYNTHESIS AND ANTIVIRAL ACTIVITY OF C-NUCLEOSIDE ANALOGUES OF RIBAVIRIN

L011

GOSMINI Romain DISCOVERY OF GLPG1205, A FIRST IN CLASS GPR84 ANTAGONIST IN PHASE II CLINICAL TRIAL

LE089

GRAVIER-PELLETIER Christine

MOLECULAR TOOLS TO STUDY AND CHARACTERIZE THE LYSOSOMAL OLIGOSACCHARIDE TRANSPORTER (LOST)

D002

GUARDIOLA Salvador TARGET-TEMPLATED DE NOVO DESIGN OF DRUG-LIKE CYCLIC PEPTIDES: PD-1/PD-L1 INHIBITORS

LE082

GUZELJ Samo STRUCTURAL FINE-TUNING OF DESMURAMYLPEPTIDE NOD2 AGONISTS DEFINES THEIR IN VIVO ADJUVANT ACTIVITY

Z028

HALECKOVA Annamaria SYNTHESIS OF SMALL MOLECULE INHIBITORS OF CYCLOPHILIN D AND THEIR IN VITRO EVALUATION FOR NEUROPROTECTIVE TREATMENT OF ALZHEIMERS DISEASE

M025

HAPKO Uladzislau DISCOVERY AND OPTIMIZATION OF VIRTUAL SCREENING AND FRAGMENT HITS AS NOVEL BACTERIAL SLIDING CLAMP DNAN INHIBITORS

R009

HARDICK David DRUGGING RNA MODIFYING ENZYMES - METTL3 INHIBITORS

LE068

HARGROVE Amanda MODULATING VIRAL RNAS WITH AMILORIDE SMALL MOLECULES

LE070

HASSAN Mujtaba DESIGN, SYNTHESIS AND EVALUATION OF D-GALACTAL DERIVATIVES AS SELECTIVE INHIBITORS OF GALECTIN-8 N-TERMINAL DOMAIN

A008

HERLAH Barbara DYNOPHORE-BASED APPROACH TO INHIBITOR DESIGN: DEVELOPMENT OF HUMAN DNA TOPOISOMERASE IIA CATALYTIC INHIBITORS

T012

HERNANDEZ Mercedes Rubio

DESIGN AND SYNTHESIS OF MANNICH BASE-TYPE DERIVATIVES CONTAINING IMIDAZOLE AND BENZIMIDAZOLE AS LEAD COMPOUNDS FOR DRUG DISCOVERY IN CHAGAS DISEASE

O003

HESSLER Gerhard NAVIGATING CHEMICAL SPACE BY ARTIFICIAL INTELLIGENCE

LE061

HEVEY Rachel DEVELOPMENT OF GLYCOMIMETIC COLLECTIN-11 ANTAGONISTS TO REDUCE COMPLEMENT-MEDIATED ISCHEMIA-REPERFUSION INJURIES

LE058

HIESINGER Kerstin DESIGN, SYNTHESIS, AND STRUCTURE-ACTIVITY RELATIONSHIP STUDIES OF DUAL INHIBITORS OF SOLUBLE EPOXIDE HYDROLASE AND 5-LIPOXYGENASE

U003

HILVERT Donald REPROGRAMMING NONRIBOSOMAL PEPTIDE BIOSYNTHESIS

LE096

HIRSCH Anna K. H. DISCOVERY OF SUBMICROMOLAR INHIBITORS OF THE VIRULENCE FACTOR LASB FROM PSEUDOMONAS AERUGINOSA USING RATIONAL DESIGN

LE065

HOLVEY Rhian KEY ASPECTS OF STRUCTURE-BASED DRUG DESIGN STRATEGIES FROM THE DEVELOPMENT OF A POTENT CLASS OF SMALL MOLECULE INHIBITORS OF THE MDM2-P53 PROTEIN-PROTEIN INTERACTION

T013

HOTINGER Julia DEVELOPMENT OF A FLUORESCENCE REPORTER ASSAY TO STUDY BACTERIAL TYPE III SECRETION SYSTEM (T3SS)-MODULATING COMPOUNDS

M028

HSIAO Meng-Yang A STUDY OF THE EFFECT OF COMMON ELECTRON-WITHDRAWING GROUPS ON THE PHYSIOCHEMICAL PROPERTIES OF 2-AMINOTETRAHYDORPYRIDINE BACE1 INHIBITORS

M058

HUTTUNEN Kristiina TARGETED (PRO)DRUGS FOR IMPROVED TREATMENT OF BRAIN TUMORS

LE087

IHNATENKO Irina ONE RING TO RULE THEM ALL: THE INDOLE IS INDISPENSABLE FOR ANTITRYPANOSOMAL PAULLONES

M030

454 | EFMC-ISMC

IJZERMAN Ad P. PARTIAL AGONISM IN THE PICTURE

AL001

IMBERTY Anne LECTINS FROM PATHOGENS: FROM STRUCTURAL GLYCOBIOLOGY TO ANTIADHESIVE STRATEGIES

LE057

IVANOVA Jekaterina AROMATIC AND HETEROAROMATIC SULFONAMIDES AS INHIBITORS OF CARBONIC ANHYDRASES - SYNTHESIS AND BIOLOGICAL EVALUATION

T015

JAMROZIK Marek THE APPLICATION OF COMPUTER-AIDED DRUG DESIGN IN THE SEARCH FOR CARBONYL REDUCTASE 1 (CBR1) INHIBITORS AS COMPOUNDS SUPPORTING CANCER CHEMOTHERAPY

M031

JENSEN Klavs F. AI ASSISTED AND AUTOMATED CHEMICAL SYNTHESIS

LE044

JERHAOUI Soufyan AN ORIGINAL PETASIS APPROACH TOWARDS THE SYNTHESIS OF NOVEL MCL-1 INHIBITORS

L012

JIYU Woo SYNTHESIS AND BIOLOGICAL EVALUATION OF NOVEL INDOLE-BASED DERIVATIVES AS COMPETITIVE AND SELECTIVE HUMAN MONOAMINE OXIDASE B INHIBITORS

M032

JONCKERS Tim DISCOVERY OF A NOVEL CLASS OF SMALL MOLECULE DENGUE VIRUS INHIBITORS

LE036

JOSA CULLERE Laia PHOTOSWITCHING MOLECULES FOR THE SPATIOTEMPORAL TARGETING OF CANCER STEM CELLS WITH LIGHT

E002

JOSA CULLERE Laia A PHENOTYPIC SCREEN IDENTIFIES A SMALL MOLECULE THAT INDUCES DIFFERENTIATION OF AML CELLS IN VITRO AND SHOWS ANTI-TUMOUR EFFECTS IN VIVO

LE080

JOSE Jiney TARGETED THERANOSTIC AGENTS FOR THE TREATMENT OF GLIOBLASTOMA MULTIFORME

M033

JUHAS Martin DESIGN, SYNTHESIS AND BIOLOGICAL EVALUATION OF NEW INHIBITORS OF MYCOBACTERIAL METHIONINE AMINOPEPTIDASE 1A

Z016

JUMDE Ravindra P. HIT-OPTIMIZATION USING TARGET-DIRECTED DYNAMIC COMBINATORIAL CHEMISTRY

M034

JUNG Erik NEW FIDAXOMICIN ANTIBIOTICS: COMBINING METABOLIC ENGINEERING AND SEMISYNTHESIS

L013

KEKEZOVIC Sladjana SYNTHESIS, X-RAY CRYSTAL STRUCTURE AND ANTITUMOUR ACTIVITY OF NOVEL DEPHENYLATED (-)-GONIOFUFURONE ANALOGUE

Z015

KELLEY Tanya DESIGN AND OPTIMIZATION OF A FIRST-IN-CLASS NACK INHIBITOR: A NOVEL PATH TO NOTCH INHIBITION

LE103

KENNELLY Samantha PYRIMIDINE MIMICS AS NEXT GENERATION CATCH AND RELEASE DNA DECOYS FOR TRANSCRIPTION FACTOR MODULATION

Z029

KIURU Paula SYNTHESIS AND ANTICANCER EVALUATION OF SPIROCYCLIC BROMOTYROSINE ANALOGS INSPIRED FROM MARINE COMPOUND CLAVATADINE C

T016

KLEIN Markus STRUCTURE-BASED OPTIMIZATION AND SYNTHESIS OF M3258, A POTENT AND SELECTIVE INHIBITOR OF THE IMMUNOPROTEASOME SUBUNIT LMP7 (BETA5I) DEMONSTRATING STRONG EFFICACY IN MULTIPLE MYELOMA MODELS.

LE101

KLEY Jörg FIRST TIME DISCLOSURE OF BI 1265162, AN ENaC INHIBITOR FOR THE TREATMENT OF CYSTIC FIBROSIS

LE026

KLUG Dana DEVELOPING DIARYLIMIDAZOLES AS ANTIBIOTICS USING AN OPEN SOURCE APPROACH

M035

KNAPP Stefan TARGETING PROTEIN SCAFFOLDING FUNCTION IN KINASES

PL004

KOBAURI Piermichele RATIONAL DESIGN OF LIGHT-CONTROLLED BIOACTIVE COMPOUNDS FOR PHOTOPHARMACOLOGY

E003

KOSTRUN Sanja MACROLIDE INSPIRED MACROCYCLES AS MODULATORS OF THE IL-17A/IL-17RA INTERACTION

M036

KOUROUNAKIS Angeliki NOVEL THIOMORPHOLINE DERIVATIVES WITH INCREASED ANTIOXIDANT ACTIVITY AGAINST NEURODEGENERATION

Q004

KUDOVA Eva DESIGN, SYNTHESIS, AND DEVELOPMENT OF NEUROSTEROIDS AS THERAPEUTICS FOR CENTRAL NERVOUS SYSTEM DISEASES

M037

EFMC-ISMC | 455

LACIVITA Enza BOOSTING THE RESOLUTION OF INFLAMMATION THROUGH FORMYL PEPTIDE RECEPTOR 2 (FPR2) AGONISTS AS A NOVEL STRATEGY IN NEUROINFLAMMATION-ASSOCIATED CENTRAL NERVOUS SYSTEM DISORDERS

LE011

LAINO Teodoro AI IN THE LAB: AUTOMATING CHEMICAL SYNTHESIS. WHERE NEXT FROM HERE?

LE046

LAMERS Christina TOWARDS NEXT GENERATION COMPLEMENT C3 INHIBITORS WITH PROLONGED TARGET RESIDENCE TIME - LATEST INSIGHTS IN STRUCTURE-ACTIVITY RELATIONSHIP AND PHARMAKOKINETIC STUDIES

X001

LAUFER Stefan DISCOVERY AND DEVELOPMENT OF A MKK-4 INHIBITORS TO INCREASE LIVER REGENERATION

LE071

LAUL Eleen SELEC-PROTACS: NOVEL CANCER TARGETING PROTEIN DEGRADERS

V001

LEBOHO Tlabo SYNTHESIS OF NOVEL 3-AMINOINDOLE DERIVATIVES AND THEIR BIOLOGICAL EVALUATION AGAINST MYCOBACTERIUM TUBERCULOSIS

M001

LEMURELL Malin EXPERIENCES & FUTURE DIRECTION FOR MEDICINAL CHEMISTRY IN CARDIOVASCULAR, RENAL AND METABOLISM DISEASES

AL002

LENCE Emilio MOLECULAR BASIS OF BICYCLIC BORONATE -LACTAMASE INHIBITORS OF ULTRABROAD EFFICACY - INSIGHTS FROM MOLECULAR DYNAMICS SIMULATION STUDIES

R010

LIARGKOVA Thalia NEW BIOLOGICAL ACTIVE HYBRIDS COMBINING ENONE AND PYRIMIDINE SCAFFOLDS

Q005

LIZANDRA PEREZ JUAN WW-DOMAIN BASED INHIBITORS OF PD-1/PDL-1 INTERACTION

T018

LOI Elena DIFFERENT DRUG DESIGN APPROACHES TO TACKLE O-GLCNAC TRANSFERASE INHIBITION

A009

LOLLI Marco INNOVATIVE DIHYDROOROTATE DEHYDROGENASE CLINICAL READY INHIBITORS AS PAN-CORONAVIRUS ANTIVIRALS: TARGETING THE UNEXPECTED WITH INNOVATION

LE039

LONDON Nir COVID MOONSHOT - CROWDSOURCING A COVID-19 CURE

PR004

LOPES Elizabeth A. ENHANCING BINDING OF SPIROPYRAZOLINE OXINDOLES TO MDMS BY IN SILICO HIT-TO-LEAD OPTIMIZATION

T019

LOPEZ ROJAS Priscila DESIGN, SEMISYNTHESIS AND ANTIESTROGENIC ACTIVITY OF LIGNAN DERIVATIVES FROM NATURAL DIBENZYLBUTYROLACTONES

T020

LOUKAS Ieremias GPR84 A DRUGGABLE PROINFLAMMATORY RECEPTOR

U001

LOWERSON Andrew BUILDING A DIVERSE AND EXPERIMENTALLY-CURATED FRAGMENT LIBRARY

Z026

LUQUE NAVARRO Pilar Maria

SUSTAINABLE SYNTHESIS OF NEW CHOLINE KINASE INHIBITORS

L015

LUZ André NOVEL CLASS OF NECROPTOTIC CELL DEATH INHIBITORS: SYNTHETIC APPROACH AND IN VITRO ASSAYS

L016

MACFARLANE Katherine EXPLOITING DNA-ENCODED LIBRARY TECHNOLOGY FOR THE DISCOVERY OF NOVEL ANTIBODY RECRUITING MOLECULES AGAINST LOX-1

M038

MACHULKIN Aleksei SYNTHESIS AND BIOLOGICAL EVALUATION OF PSMA TARGETED FLUORESCENT CONJUGATES

V002

MACOR John E. THE DISCOVERY OF RELPAX® AND NURTEC ODT® - TWO DIFFERENT TREATMENTS FOR MIGRAINE HEADACHES

PR005

MALARZ Katarzyna NOVEL BENZENESULFONATE SCAFFOLDS WITH A HIGH ANTICANCER ACTIVITY AND G2/M CELL CYCLE ARREST IN GLIOBLASTOMA MULTIFORME

M039

MALINAUSKIENE Vida SYNTHESIS OF 2-((1H-INDOL-3-YL)METHYL)-5-(ALKYLTHIO)-1,3,4-OXADIAZOLES AND THEIR PROTECTIVE ACTIVITY AGAINST OXIDATIVE STRESS

U002

MALISZEWSKI Dawid THE NOVEL 1,3,5-TRIAZINE DERIVATIVES AS INNOVATIVE POTENTIAL ACHE AND BACE1 INHIBITORS

C012

MALLART Sergio IDENTIFICATION OF POTENT AND LONG ACTING SINGLE-CHAIN PEPTIDE MIMETICS OF HUMAN RELAXIN-2 FOR CARDIOVASCULAR DISEASES

LE021

456 | EFMC-ISMC

MANEIRO Maria METHYLHYDROXYLAMMONIUM DERIVATIVES - LYSINE-TARGETED IRREVERSIBLE INHIBITORS FOR THE ANTI-VIRULENCE BACTERIAL TARGET TYPE I DEHYDROQUINASE

R011

MANNISTO Jere Kristian THE ROLE OF GUANIDINE-CO2 ADDUCTS IN CARBOXYLATIONS: MECHANISTIC INSIGHTS

L017

MANOHARAN Muthiah NUCLEIC ACIDS AS HOUSEHOLD MEDICINE: LIVING IN THE WORLD OF RNA THERAPEUTICS

LE076

MAPLE Hannah VALIDATING TARGETS FOR TARGETED PROTEIN DEGRADATION USING DTAG – A COMPREHENSIVE WORKFLOW SOLUTION

C013

MARAZZI Marco RATIONALIZING THE MOLECULAR INTERACTIONS BETWEEN HUMAN AND VIRAL KEY ELEMENTS INVOLVED IN SARS-COV-2 INFECTION: LIMITS AND SUCCESSES TO DESIGN A BETTER FUTURE

O007

MARTINEZ ALARCON Dania

RECOMBINANT LECTIN FROM TEPARY BEAN (phaseolus acutifolius) WITH SPECIFIC CYTOTOXIC EFFECT OF COLON CANCER CELLS: PRODUCTION, STRUCTURAL CHARACTERIZATION AND TARGET IDENTIFICATION.

A010

MARTINEZ VITURRO Carlos Manuel

POSITIVE ALLOSTERIC MODULATORS OF GLUN2B-CONTAINING NMDARS AS ENHANCERS OF SYNAPTIC PLASTICITY

X003

MARTINO Elena DESIGN AND SYNTHESIS OF A LIBRARY OF PYRAZOLO[1,5-A]PYRIDINE FLUOROPHORES FOR DEVELOPING NEW FLUORESCENT PROBES

C014

MATTEI Patrizio TETHERED MACROCYCLIC PEPTIDES, A NOVEL ANTIBIOTIC CLASS TARGETING ACINETOBACTER BAUMANNII

LE064

MEIBOM Daniel NELADENOSON BIALANATE HYDROCHLORIDE - PRODRUG EXPLORATION OF A PARTIAL ADENOSINE A1 AGONIST

LE086

MEMARZADEH Sarah METATACS: A STRATEGY FOR METASTASIS PREVENTION THROUGH TARGETED FASCIN DEGRADATION

M041

MESSORE Antonella QUINOLONYL DKA DERIVATIVES DISRUPT THE INTERACTION BETWEEN V-RNA AND INTEGRASE OF HIV-1

M042

MEZAACHE Roufia COPPER (II) BROMIDE AS AN EFFICIENT CATALYST FOR THE SELECTIVE PROTECTION AND DEPROTECTION OF ALCOHOLS

L018

MEZOHEGYI Gergo NOVEL METHOD TO IMPROVE API'S STABILITY IN MICROGRAVITY ENVIRONMENT: CASE STUDY FOR COVID-19 DRUG FORMULATION IN SPACE

W003

MICHALEK Stanislaw POTENT AND SELECTIVE INDOL-4-YL PYRAZOLO[1,5-a]PYRIMIDINE-DERIVED PI3Kδ INHIBITORS AS POTENTIAL CANDIDATES FOR TREATMENT OF COPD AND ASTHMA

Z032

MIHAJLO Krunic SYNTHESIS AND PHARMACOLOGY EVALUATION OF NOVEL 1- BENZYL-N-(4-(4-ARYLPIPERAZIN-1-YL)PHENYL)PIPERIDIN-4- CARBOXAMIDES, AS POTENTIAL ACETYLCHOLINESTERASE INHIBITORS

Z010

MIHOVILOVIC Marko D. FLAVOENZYME BIOCATALYSIS IN BIOORGANIC AND MEDICINAL CHEMISTRY - CHALLENGES AND OPPORTUNITIES

LE097

MIN Kyoungho NOVEL ARYL SULFONAMIDES TARGETING THE PROTEIN-PROTEIN INTERACTION OF AIMP2-DX2 WITH HSP70 AS POTENTIAL CANCER THERAPEUTICS

L020

MIRET CASALS Laia FURAN-OXIDATION MEDIATED TECHNOLOGY: FROM IN VITRO ANALYSIS OF PROTEIN-PROTEIN INTERACTIONS TO GPCR-LIGAND INTERACTIONS ON LIVE CELLS

LE073

MONTEIRO Ana IMMOBILIZATION OF ZINC(II) PHTHALOCYANINES ON GRAPHENE OXIDE FOR DNA RECOGNITION

M043

MONTI Barbara MICROGLIA IN NEUROINFLAMMATION: AN INTRIGUING TARGET TOWARDS NEUROPROTECTION AND NEUROREGENERATION

LE009

MORALES Juan Carlos USE OF IMIDE CONDENSATION FOR PREPARING CORE-DIVERSIFIED G-QUADRUPLEX LIGANDS: ANTICANCER AND ANTIPARASITIC ACTIVITY

M044

MORRIS Morgan SYNTHESIS OF LACTOSE-DRUG CONJUGATES FOR LIVER-TARGETED DELIVERY

A011

MOTIEI Leila PATTERN-GENERATING FLUORESCENT MOLECULAR PROBES FOR CHEMICAL BIOLOGY LE004

EFMC-ISMC | 457

MOUROT Laura OPTIMIZATION OF A NOVEL FAST ACTING TRANSMISSION BLOCKING ANTIMALARIAL AGENT

Z009

MROZ Piotr NEW PEX14 LIGANDS - A NEW STRATEGY TO INTERRUPT GLYCOSOME BIOGENESIS

M045

MUSILEK Kamil ENCAPSULATION OF CHARGED CHOLINESTERASE REACTIVATORS USING APOFERRITIN MAY PROLONG THEIR HALFLIFE AND ENHANCE BIOAVAILABILITY IN CNS

V004

NACHAMPASSAK Southida

DEOXYFLUORINATION OF ACYL FLUORIDES TO TRIFLUOROMETHYL COMPOUNDS BY FLUOLEAD/OLAH'S REAGENT UNDER SOLVENT-FREE CONDITIONS

L021

NACHAMPASSAK Southida

FLUOLEAD, A VERSATILE AND SAFE NUCLEOPHILIC FLUORINATION AGENT

L019

NAR Herbert ALLOSTERIC REGULATION OF GTP CYCLOHYDROLASE I

LE018

NARDUCCI Daniele SYNTHESIS OF SPIROCYCLIC-DHEA DERIVATIVES AS NEUROTROPHIN MIMETICS

L022

NAZARE Marc ILLUMINATING CANNABINOID RECEPTORS: FLUORESCENT PROBES FOR SPECIFIC VISUALIZATION OF THE CANNABINOID TYPE 2 RECEPTOR

D007

NERI Dario ANTIBODY-DRUG CONJUGATES (ADC) AND SMALL MOLECULE-DRUG CONJUGATES (SMDC): A COMPARATIVE EVALUATION

LE075

NEUMANN Alexander CHEMICAL SPACE DOCKING: MINING BILLIONS OF ON-DEMAND MOLECULES IN 3D

M046

NEWTON Gary MAP4K4 INHIBITORS FOR THE SUPPRESSION OF CARDIAC MUSCLE CELL DEATH

LE023

NIE Laiyin WUXI HitS: UNLEASHING THE POTENTIAL OF HIT DISCOVERY

Z021

NIKOLIC Katarina RATIONAL DESIGN, SYNTHESIS AND IN VITRO TESTING OF SELECTIVE HDAC6 AND SIRT2 INHIBITORS

T021

NILSSON Magnus DISCOVERY OF THE CLINICAL CANDIDATE AZD4604, A POTENT AND SELECTIVE JANUS KINASE 1 INHIBITOR, AS AN INHALED TREATMENT FOR RESPIRATORY DISEASE

LE027

NILSSON Ulf DESIGN, SYNTHESIS, AND DEVELOPMENT OF LECTIN LIGAND MIMETICS

LE056

NOCEN Pawel NOVEL PD-1-TARGETED INHIBITORS OF PD-1/PD-L1 INTERACTION DESIGNED BASING ON THE MVAT MINI-PROTEIN STRUCTURE

T022

NOVOTNA Adéla SYNTHESIS AND SAR OF PEPTIDE JUVENOIDS AS POTENT AGONISTS SPECIFIC TO AN INSECT JH RECEPTOR

Z034

NOWICKI Krzysztof BIS(OXABOROLES) AND BIS(SILOXABOROLES) AS POTENT ANTIMICROBIAL AGENTS

L023

NOZAL GARCIA Vanesa ISOFORM-SELECTIVE TAU TUBULINE KINASE 1 INHIBITORS REDUCE TDP-43 HYPERPHOSPHORYLATION: A NEW HOPE IN THE TREATMENT OF TDP-43 PROTEINOPATHIES

LE081

NUE Jonathan SYNTHESIS AND IN VITRO STUDIES OF BIS-BENZIMIDAZOLE DERIVATIVES TARGETING KINETOPLASTID PARASITES

C017

OLIVER Martin SYNTHESIS OF NEW MraY INHIBITORS AND THEIR BIOLOGICAL EVALUATION

L024

OMBOUMA JOANNA PHYTOCHEMISTRY AND CELLULAR TOLERANCE OF THREE ANTIDIARRHEA PLANTS OF GABON

E004

OPREA Tudor I. ILLUMINATING THE RARE DISEASE PROTEOME

O006

ORTIN REMON Irene SYNTHESIS OF NOVEL CK2/HDAC DUAL INHIBITORS BASED ON PHARMACOPHORE HYBRIDIZATION STRATEGY

T023

PACHECO Paulo DEVELOPMENT OF NOVEL TRYPTOPHANOL-DERIVED OXAZOLOPYRROLIDONE LACTAMS WITH ANTIPROLIFERATIVE ACTIVITY AGAINST HUMAN GASTRIC ADENOCARCINOMA (AGS) CELL LINE

T024

PALAFERRI Leonardo TOWARDS PROTAC-MEDIATED DEGRADATION OF CBP/EP300

C016

PALAZZESI Ferruccio APPLICATION OF ARTIFICIAL INTELLIGENCE ON DRUG DISCOVERY PROJECT: AN EVOTEC INSIGHT

H003

458 | EFMC-ISMC

PALISSE Adeline DISCOVERY OF THE HIGHLY POTENT AND SELECTIVE S1P2 ANTAGONIST GLPG2938, A PRECLINICAL CANDIDATE FOR THE TREATMENT OF IDIOPATHIC PULMONARY FIBROSIS

LE091

PALOMO Valle SENSING ENZYMATIC ACTIVITY WITH QUANTUM DOTS IN PATIENT-DERIVED NEURODEGENERATIVE DISEASE MODELS

LE003

PALYULIN Vladimir A. MOLECULAR MODELLING AND DESIGN OF NOVEL SUBNANOMOLAR AMPA RECEPTOR MODULATORS

Q006

PANNIUNGHI Sara A COST-EFFECTIVE AND EASY-TO-USE METHOD TO ASSESS CELL PERMEABILITY OF NEW PROTAC E3 LIGASE BINDERS

C010

PAPAGIOUVANNIS Georgios

NOVEL COMPOUNDS WITH ANTIOXIDANT PROPERTIES CONTAINING CNS-ACTING MOIETIES AGAINST NEURODEGENERATION

Q007

PAPAGIOUVANNIS Georgios

ANTIOXIDANT CINNAMIC ACID DERIVATIVES WITH ANTI-INFLAMMATORY AND HYPOLIPIDEMIC POTENCY

Z012

PAPOT Sebastien TARGETING THE SPECIFICITIES OF THE TUMOR MICROENVIRONMENT FOR CANCER THERAPY AND DIAGNOSIS

LE078

PASQUALE Linciano BIVALENT LIGANDS AS PHARMACOLOGICAL TOOLS FOR STUDYING THE ROLE OF SIGMA-1 RECEPTOR AGONISM IN NEUROPROTECTION AND NEURODEGENERATIVE DISEASES

Q008

PEREZ MELERO Maria Concepcion

SYNTHESIS AND BIOLOGICAL EVALUATION OF CONJUGATE DRUGS BEARING BILE ACID CARRIERS TO TARGET LIVER TUMOURS

V003

PEREZ-PEREZ Maria-Jesus ANTIVIRALS AGAINST CHIKUNGUNYA VIRUS: ANALYZING THE CURRENT SITUATION TO IDENTIFY NEW OPPORTUNITIES

LE037

PERI Francesco TARGETING TLR4 WITH SYNTHETIC MOLECULES: INNOVATIVE THERAPEUTICS FOR INFECTIOUS AND INFLAMMATORY DISEASES

A012

PERMANN Stephan STRUCTURAL OPTIMIZATION OF LONG-CHAIN VITAMIN E METABOLITES YIELDS DERIVATIVES THAT INDUCE A LIPID MEDIATOR CLASS SWITCH FROM INFLAMMATION TO RESOLUTION

X002

PEROKOVIC Vesna SYNTHESIS OF FERROCENE ESTER DERIVATIVES OF DESMURAMYL PEPTIDE

Z022

PERRON Quentin FRAGMENT GROWING WITH REACTION PREDICTION FOR HIT DISCOVERY

H002

PERSONNE Hippolyte X-RAY CRYSTAL STRUCTURES OF SHORT MIXED CHIRALITY A-HELICAL ANTIMICROBIAL PEPTIDES

R012

PETERLIN MASIC Lucija NEW POTASSIUM ION CHANNELS KV1.3 AND KV10.1 INHIBITORS AS NEW ANTICANCER COMPOUNDS

T025

PETTER Jennifer TARGETING RNA WITH DRUG-LIKE SMALL MOLECULES

LE067

PEUKERT Carsten DOTAM-BASED SIDEROMYCINS TO VISUALIZE AND TREAT MDR BACTERIAL PATHOGENS IN VIVO AND IN VITRO

LE066

PHILLIPS Jonathan INTRODUCTION TO PRECLINICAL INHALED DRUG DOSING AND ITS APPLICATION TO MEASURE THE THERAPEUTIC INDEX OF INHALED STEROIDS

LE050

PICOTTI Paola PROTEOMES IN 3D

LE093

PIPPIONE Agnese Chiara MULTIPLE-TARGETING LIGANDS FOR AKR1C3 ENZYME AND ANDROGEN RECEPTOR TO TARGET PROSTATE CANCER

T026

PIRALI Tracey MAGIC IN THE MOONLIGHT: OUR CONTRIBUTION TO THE DEVELOPMENT OF IDO1 INHIBITORS FOR CANCER IMMUNOTHERAPY

LE102

PONTES Olivia NEW CHROMENE DERIVATIVES: OVERCOMING RENAL CELL CARCINOMA DRUG RESISTANCE

T027

POOJA Deshmukh GENERATIVE RECURRENT NEURAL NETWORKS (RNN) FOR DE NOVO DRUG DESIGN

LE062

POUTON Colin POTENTIAL OF STEM CELL TECHNOLOGY IN DISCOVERY OF DRUGS FOR NEURODEGENERATIVE DISEASES

LE043

EFMC-ISMC | 459

PRISCHICH Davia IN VIVO ADRENERGIC MODULATION WITH PHOTOPHARMACOLOGY

LE016

PROJ Matic DISCOVERY OF SELECTIVE FRAGMENT-SIZED IMMUNOPROTEASOME INHIBITORS

C015

QI Xiangbing MOLECULAR GLUE DESIGN FOR TARGETED CANCER THERAPY LE006

QILI LU FIRST TOTAL SYNTHESIS OF CUDRAISOFLAVONE J AS POTENTIAL NEUROPROTECTIVE AGENT

M047

QUILIANO Miguel PHARMACOPHORE-BASED VIRTUAL SCREENING FOR THE IDENTIFICATION OF NEW MYCOBACTERIUM TUBERCULOSIS PKNG INHIBITORS

Z019

RADEMACHER Christoph C-TYPE LECTIN RECEPTORS AS DRUG TARGETS

LE055

RAJIC Zrinka SYNTHESIS OF NOVEL AMIDE TYPE HARMICINES

M048

RAJIC Zrinka SYNTHESIS OF C-1- AND C-3-TETHERED TRIAZOLE TYPE HARMICINES

M049

RAJIC Zrinka SYNTHESIS OF O- AND N-HARMIRINES, NOVEL HARMINE-COUMARIN HYBRIDS

M050

RAMILO-GOMES Filipa EPIGENETIC DRUG DISCOVERY: NOVEL PROMISING EZH2 INHIBITORS AGAINST CANCER

T028

RAUH Daniel TARGETING CANCER

LE100

RAUTIO Jarkko PRODRUGS STRATEGIES IN MEDICINAL CHEMISTRY

LE084

REEKS Judith THE ROLE OF CRYO-EM IN FRAGMENT-BASED DRUG DISCOVERY

LE019

REIS Joana DRUG DESIGN AGAINST CARCINOGENESIS: DEVELOPMENT OF NEW NADPH OXIDASE INHIBITORS AS A PATHWAY TO CANCER THERAPY

T029

REVOL-TISSOT Johan SYNTHESIS OF NEW FLUORESCENT CHEMICAL PROBES TARGETING BACTERIAL EFFLUX TO EARLY DETECT AND FIGHT THE FIRST BARRIER IN BACTERIA ANTIBIOTIC RESISTANCE

C011

REYNOLDS Jessica DESIGN AND SYNTHESIS OF CHEMICAL TOOLS TO PROBE THE FUNCTION OF TRIM33

LE095

RIBIC Rosana DESIGN, SYNTHESIS AND EVALUATION OF IMMUNOSTIMULATING ACTIVITIES OF DESMURAMYL PEPTIDES CONTAINING 2-AMINOADAMANTANE-2-CARBOXYLIC ACID

A013

RICCIO Alessandra HOW PH COULD AFFECT THE INTERACTION STRENGTH DURING PD-L1 INHIBITORS RECOGNIZING

RICHALET FLORIAN DISCOVERY AND OPTIMIZATION OF OXIDATIVE-PHOSPHORYLATION INHIBITORS FROM A PHENOTYPIC SCREEN

T030

RIVERA FUENTES Pablo CHEMICAL TOOLS TO STUDY REDOX BIOLOGY LE002 ROCHAIS Christophe PLEIOTROPIC PRODRUGS: A NOVEL POLYPHARMACOLOGY APPROACH TO TREAT

NEURODEGENERATIVE DISEASES

N003

ROCHE Didier LIGAND IDENTIFICATION FROM NON-ENCODED CHEMICAL MATTER USING AFFINITY SELECTION MASS SPECTROMETRY

M040

RODRIGUEZ-COSTA Angela

DISABLING PSEUDOMONAS AERUGINOSA VIRULENCE BY MODULATION OF THE MASTER REGULATOR OF QUORUM SENSING LasR

R013

ROEHRIG Susanne DESIGN AND PRECLINICAL CHARACTERIZATION PROGRAM TOWARDS BAY 2433334, AN ORAL FACTOR XIA INHIBITOR FOR THE PREVENTION AND TREATMENT OF THROMBOEMBOLIC DISORDERS

LE025

ROMANO Assunta RETROSYNTHETIC SOFTWARE FOR PRACTICING CHEMISTS: NOVEL AND EFFICIENT IN SILICO PATHWAY DESIGN VALIDATED AT THE BENCH

L025

ROMANO Assunta PROTEIN DEGRADER BUILDING BLOCKS FOR STREAMLINED SYNTHESIS OF HETEROBIFUNCTIONAL DEGRADER LIBRARIES

M026

ROMANO Assunta RAPID HIT DISCOVERY WITH DNA-ENCODED LIBRARY TECHNOLOGY USING DyNAbind® KITS

M027

ROSSMEIER Maria SYNTHESIS OF SECOND GENERATION FLUORESCENT TRIPPPRO COMPOUNDS FOR CELLULAR UPTAKE STUDIES

T032

460 | EFMC-ISMC

RUIZ-GOMEZ Gloria MOLECULAR RECOGNITION OF GLYCOSAMINOGLYCANS BY VASCULAR ENDOTHELIAL GROWTH FACTOR AND THEIR DUAL ROLE ON ANGIOGENIC PROCESSES

A014

RUNFOLA Massimiliano A MULTIDISCIPLINARY APPROACH TO DRUG DISCOVERY REVEALS NEW PHARMACEUTICAL TOOLS AGAINST NEURODEGENERATIVE DISEASES

M004

RUSSELL Angela DISCOVERY OF SMALL MOLECULES TO MANIPULATE CELL FATE IN VIVO: TOWARDS NEW THERAPIES FOR DEGENERATIVE DISEASES

LE041

RUZIC Dusan SYNTHESIS, MOLECULAR MODELLING AND BIOLOGICAL CHARACTERIZATION OF NOVEL ANTIMIGRATORY AND ANTIINVASIVE 1-BENZHYDRYL PIPERAZINE DERIVATIVES

T033

SAADABADI Atefeh STRUCTURAL INSIGHTS INTO NATURAL STILBENOIDS AS POTENTIAL TRPA1 MODULATORS

Z040

SAHRAOUI Suzanne Sherihan

DEVELOPMENT OF A NEW TARGET IDENTIFICATION SYSTEM FOR SMALL MOLECULES IN TRYPANOSOMA BRUCEI PARASITES

LE008

SAINAS Stefano A DIFFERENTIATING AND APOPTOTIC THERAPY FOR ACUTE MYELOID LEUKAEMIA USING MEDS433, A POTENT HUMAN DIHYDROOROTATE DEHYDROGENASE INHIBITOR

T034

SALO-AHEN Outi IN SEARCH OF NOVEL BACTERIAL RNA POLYMERASE INHIBITORS

R014

SARABIA VALLEJO Alvaro NEW MULTITARGET THERANOSTIC COMPOUNDS AGAINST ALZHEIMER’S DISEASE

Z014

SAURABH Loharch ORGANOSELENIUM BASED COMPOUNDS AS POTENT INHIBITORS OF BACTERIAL UREASE

M019

SCHADE Dennis PROBING EMBRYONIC MESODERMAL DIFFERENTIATION ENABLES IDENTIFICATION OF SMALL MOLECULE BONE MORPHOGENETIC PROTEIN ACTIVATORS

LE042

SCHEFFLER Frank INTEGRATION OF BIOCATALYSIS INTO MEDICINAL CHEMISTRY PROGRAMS FOR LATE-STAGE OXIDISED DERIVATIVES USING POLYCYPS ENZYMES

LE099

SCHEPENS Wim CYCLIC DINUCLEOTIDE STING AGONISTS WITH A NOVEL SULFAMATE LINKER SHOW ANTI-TUMOR ACTIVITY IN MOUSE MODELS FOLLOWING SYSTEMIC ADMINISTRATION

T035

SCHNEIDER Gisbert DE NOVO DRUG DESIGN WITH MACHINE INTELLIGENCE

AL003

SCHNEIDER Nadine AUGMENTING DRUG HUNTERS WITH GENERATIVE CHEMISTRY MODELS

LE060

SERAFINI Marta TARGETING TRPV1 SOFTLY: SYNTHESIS AND DEVELOPMENT OF CAPSAICINOID MODULATORS WITH IN VIVO EFFICACY IN MOUSE MODELS OF SKIN DISEASES

M051

SHAW Duncan DESIGN AND DEVELOPMENT OF INHALED MOLECULES TO TARGET THE PULMONARY VASCULATURE

LE051

SHTIL Alexander TARGETING TRANSCRIPTIONAL REPROGRAMMING IN CANCER: EFFICACY OF CDK8/19 INHIBITORS ALONE AND IN COMBINATION WITH CONVENTIONAL TREATMENTS

T036

SIEBER Stephan A. CHEMICAL-PROTEOMIC STRATEGIES TO FIGHT MULTIRESISTANT BACTERIA AL004 SMITH James MEDICINAL CHEMISTRY DISCOVERY STORY OF AZD3229: A PAN-MUTANT KIT INHIBITOR

FOR THE TREATMENT OF GIST

T037

SOARES Pedro DEVELOPMENT OF INNOVATIVE MULTITARGET DRUGS FOR AMYOTROPHIC LATERAL SCLEROSIS BY TARGETING NEUROINFLAMMATION AND MODULATING IRON DYSREGULATION

Z025

SOKLOU Kossi Efouako GOLD CATALYSIS FOR THE SYNTHESIS OF HETEROSPIROCYCLES

L027

SOUSA Maria Emilia THIOXANTHONES AS ADJUVANTS TO REVERT ANTIMICROBIAL RESISTANCE

R015

SOUTHALL Stacey CRYO-EM IN GPCR STRUCTURE BASED DRUG DISCOVERY

LE017

SPICER Julie TOWARD A NEW THERAPY FOR IMMUNE-MEDIATED TISSUE INJURY

LE079

STAMPOLAKI MARIANNA DESIGN AND SYNTHESIS OF POTENT INHIBITORS OF TREHALOSE MONOMYCOLATE TRANSPORTER MMPL3 OF MYCOBACTERIUM TUBERCULOSIS

Z013

STEIMBACH Raphael R. HDAC10 SELECTIVE CHEMICAL PROBES FROM PAN-INHIBITOR BY SUBSTITUTION OF A SINGLE METHYLENE IN VORINOSTAT (SAHA)

C008

EFMC-ISMC | 461

STEINEBACH Christian PROTAC-MEDIATED INACTIVATION OF E3 LIGASES: FROM TOOLS TO CANCER TREATMENT

M052

STOUSE Adrien ONE CURIE LEVEL PRODUCTION OF [18F]SynVesT-1 TRACER AT HIGH MOLAR ACTIVITY USING THE COPPER MEDIATED FLUORODESTANNYLATION METHOD

D005

STYPIK Mariola NOVEL BENZIMIDAZOLE-DERIVED PI3Kδ INHIBITORS AS HIGHLY POTENT DRUG CANDIDATES FOR SLE AND OTHER INFLAMMATORY AND AUTOIMMUNE DISEASES

Z033

SZANTI-PINTER Eszter STEREOSELECTIVE REDUCTION OF STEROIDAL ENONES LEADING TO BIOLOGICALLY IMPORTANT SKELETONS

L028

SZYMANSKI Wiktor PHOTOPHARMACOLOGY: TOWARDS LIGHT-CONTROLLED THERAPY

LE013

SZYMASZEK Patryk SPECTROPHOTOMETRIC STUDIES OF NOVEL FLUORESCENT SENSORS AND THEIR BIOLOGICAL APPLICATION FOR ALBUMIN DETECTION

C007

TASCI Hayrünnisa SYNTHESIS OF NOVEL BENZOXAZOLINONE-HYDRAZONE DERIVATIVES AS POTENT AND SELECTIVE MAO A/B INHIBITORS FOR TREATMENT OF NEURODEGENERATIVE DISEASES

Z042

TASCI Hayrünnisa SYNTHESIS AND CYTOTOXIC ACTIVITY EVALUATION OF NOVEL HYDRAZONE DERIVATIVES AS POTENTIAL ANTICANCER AGENTS

T038

TERNAD Indiana STUDY OF RADIOSENSITIZING PROPERTIES INDUCED BY PEGYLATED MAGNETIC NANOPARTICLES

D006

THOMPSON James INVESTIGATING THE CHAMELEONIC PROPERTIES OF RGD INTEGRIN ANTAGONISTS FOR THE TREATMENT OF IDIOPATHIC PULMONARY FIBROSIS

LE090

THUM Thomas DEVELOPMENT OF NON-CODING RNA BASED NEXT-GENERATION HEART FAILURE THERAPEUTICS

LE024

TIEDT Stephanie DEVELOPMENT OF SMALL MOLECULE ALLOSTERIC BETA 2 ADRENOCEPTOR MODULATORS

W002

TINWORTH Christopher STRUCTURAL INSIGHTS INTO PROTAC-MEDIATED DEGRADATION OF BCL-XL

M053

TOMASIC Tihomir NEW HSP90 C-TERMINAL INHIBITORS: HIT DISCOVERY, HIT OPTIMIZATION AND EVALUATION

T039

ULIASSI Elisa ONE FOR ALL? TOWARDS THE DEVELOPMENT OF POLYAMINE BASED- PHOTOTHERANOSTIC CONJUGATES FOR CANCER APPLICATIONS

E005

VAN DER STELT Mario CHEMICAL PROBES TO STUDY LIPID SIGNALING

LE029

VAN KLAVEREN Sjors HOW PHTHALAZINONE-DERIVATIVES BIND THE GALECTIN-8N CARBOHYDRATE RECOGNITION DOMAIN WITH EXCELLENT SELECTIVITY

A015

VAN MAARSEVEEN Jan SCAFFOLD-ASSISTED PEPTIDE CYCLIZATIONS: TOWARDS PROTEIN MIMICS

LE034

VASSILIOU Stamatia COVALENT INHIBITION OF BACTERIAL UREASE BY BIFUNCTIONAL CATECHOL-BASED PHOSPHONATES AND PHOSPHINATES

Z001

VASSILIOU Stamatia SYNTHESIS OF NOVEL CINNAMIC ACID DERIVATIVES AS POSSIBLE ANTI-ALZHEIMER AGENTS

Z002

VASTAKAITE Greta PEPTIDE CATALYSTS FOR CONJUGATE ADDITION REACTIONS WITH C-SUBSTITUTED MALEIMIDES

L029

VAUZEILLES Boris NEW BORINIC PROBES FOR FAST DETECTION AND IMAGING OF HYDROGEN PEROXIDE

D003

VAUZEILLES Boris CARBOHYDRATE PROBES FOR BACTERIAL LABELING

A016

VELTER Ingrid LEAD OPTIMIZATION OF A SERIES OF [1,2,4]TRIAZOLO[1,5-A]PYRIMIDINE DERIVATIVES AS POTENT, SELECTIVE AND BRAIN PENETRANT PDE2A INHIBITORS

Z036

VIGATO Chiara IMPROVEMENT OF METABOLIC WEAKNESS OF NEW HUMAN DIHYDROOROTATE DEHYDROGENASE INHIBITORS BASED ON 2-HYDROXYPYRAZOLO[1,5-A]PYRIDINE SCAFFOLD

T040

VISENTIN Sonja NANOMUG: A NOVEL GLYCOSYLATED PROTEIN BASED DELIVERY SYSTEM

A001

VOLYNETS Galyna DISCOVERY OF NOVEL ANTITUBERCULOSIS AGENTS TARGETING AMINOACYL-TRNA SYNTHETASES

M054

462 | EFMC-ISMC

WAN Murray NOVEL UNNATURAL NUCLEOSIDE ANALOGUES AS PRMT5 INHIBITORS

M055

WITT Julian SYNTHESIS OF FLUORESCENCE-LABELED TRIPPPRO-COMPOUNDS FOR CELLULAR UPTAKE STUDIES IN VARIOUS CANCER CELL LINES

N002

WORKMAN Paul CHEMICAL PROBES TO EXPLORE CANCER BIOLOGY AND VALIDATE DRUG TARGETS

LE028

WYCHOWANIEC Anna FLUORESCENT PROBES FOR IMAGING OF PROTEIN NANOENVIRONMENT

D004

WÜNSCH Bernhard DEVELOPMENT OF FLUORINATED PET TRACERS FOR IMAGING OF SIGMA-1 RECEPTORS IN THE BRAIN

LE010

YANG Shengyong DISCOVERY OF NEW SARS-COV-2 MPRO INHIBITORS WITH POTENT IN VITRO AND IN VIVO ANTIVIRAL ACTIVITY

LE007

YOKOKAWA Fumiaki DISCOVERY OF NITD-688: A POTENT PAN-SEROTYPE INHIBITOR OF DENGUE VIRUS NS4B PROTEIN

O001

ZACHAROVA Marija EQATA: EQUITABLE ACCESS TO QUALITY ANTIBIOTIC THERAPIES IN AFRICA

R016

ZAJDEL Pawel INVERSE AGONISTS OF 5-HT6 RECEPTOR-OPERATED SIGNALING AS A NEW PARADIGM TO TREAT NEUROPATHIC PAIN AND COGNITIVE CO-MORBIDITIES

C006

ZAKARIA Ahmed SYNTHESIS OF THE OXA-BRIDGED TRICYCLE CORE STRUCTURE OF DAPHNANE DITERPENOID ORTHOESTERS VIA PRINS-TYPE CYCLIZATION REACTION

L014

ZALA Ajayrajsinh BENZATHIAZOLE ANALOGS AS BIOLOGICALLY ACTIVE AGENTS

M056

ZETSCHOK Dominik FLUORINATED OXINDOLE SYNTHESIS VIA ORGANOCATALYTIC REACTIONS WITH THIOESTER ENOLATES

L026

ZHANG Jian ALONG THE ALLOSTERY STREAM: RECENT ADVANCES IN COMPUTATIONAL METHODS FOR ALLOSTERIC DRUG DISCOVERY

LE005

ZHANGPING Xiao 4-IODOPYRIMIDINE IS A POTENT AND SELECTIVE COVALENT INHIBITOR AND ACTIVITY-BASED PROBE FOR MIF FAMILY PROTEINS

C003

ZIPFEL Pauline INHIBITION OF MATRIX METALLOPROTEINASES TO FIGHT ALZHEIMER’S DISEASE

W001

ZORE Matej SYNTHESIS AND BIOLOGICAL EVALUATION OF FINGOLIMOD DERIVATIVES AS ANTIBACTERIAL AGENTS

Z035

ZUBRIENE Asta UNDERSTANDING THE MECHANISM OF SECONDARY SULFONAMIDE BINDING TO CARBONIC ANHYDRASE

M057

ZWERGEL Clemens DESIGN OF FIRST-IN-CLASS DUAL EZH2/HDAC INHIBITOR: BIOCHEMICAL ACTIVITY AND BIOLOGICAL EVALUATION IN CANCER CELLS

T014

EFMC-ISMC | 463

Index of Authors

464 | EFMC-ISMC

Lastname and firstname initial(s) Abstract number ABAKUMOV M. V002 ABAS S. Z024 ABDELSALAM M. T001 ABEN R. W. Z031 ABONYI T. E001 ABRANYI-BALOGH P. C015 ADAMS C. LE074 AFONSO C. A. M. LE031 AGBABA D. T021, Z006 AGIANIAN B. LE072 AI Y. LE006 AITKEN L. C002 AKDAG M. Z038 AKHLAGHI H. LE079 ALBRECHT W. LE071 ALBRECHT S. Z009 ALCANTARA R. Z019 ALCARO S. T002 ALCAZAR J. X003 ALCOLEA V. O002, O003 ALEXAKI V. I. L022 ALFENIM A. Z025 ALHAYEK A. M034 ALIBERT S. C011 ALIU B. A004 AL-KARADAGHI S. T034 ALLEGREZZA M. T035 ALMEIDA V. A006 ALMEIDA J. M003 ALNAJJAR R. Z041 ALTMAN M. M055 ALTMANN E. LE054 ALVAREZ K. O004 AMARAL J. A006 AMDOUNI Y. L006 AMESTY Ã. T020 AMPE C. LE073 AMROLLAHI P. C013 ANDERLUH M. A007, A008, A009,

A015 ANDERSON K. LE074 ANDERSON N. PR003 ANDRADE C. H. LE038 ANDREA C. M013 ANDRYS R. V004 ANGHEL N. L006 ANGIONI C. U003

ANTICA M. A013 ANTONCHICK A. LE042 ANTONIJEVIC M. X004 ANTUNES A.M.M. M003, T024 APRILE S. LE102 AQIL R. LE023 ARICA A. Z019 ARSENIYADIS S. LE033 ARTHANARI H. T029 ASHRAF U. L007 ASTOLFI A. LE012 ATHERTON R.L. O003 ATZ K. M002 AUBERT S. LE033 AUGUSTIN M. Z039 AVCI A. Z011 AVERINA E. B. Q006 AYE Y. PR002 AZTOPAL N. Z011 BAERISWYL S. R012 BAGNATI R. T040, LE039 BAI R. T002 BAJUSZ D. C015 BALAMURUGAN K. A014 BALSOLLIER C. A009 BALTAZAR F. T008, T027 BANACH M. Z032, Z033 BANTREIL X. C006 BAQI Y. U004 BARAN P. PL003 BARANGER K. W001 BARBIERI F. T005 BARCHERINI V. M003, T024 BARISIC L. Z022 BARLAAM B. LE092 BARNIOL-XICOTA M. C001 BARONAS D. M057 BARONE G. O007 BARRAJA P. T002 BARRECA M. L. LE012 BARRECA M. T002 BASTA-KAIM A. LE011 BASU S. Q009 BATLLE M. LE016 BATTISTELLI C. T014 BAUSSIERE F. A008 BAVO F. Q001, C014 BAYFORD M. LE023

EFMC-ISMC | 465

BDZHOLA V. M054 BEATON N. LE092 BEATRICE B. M013 BEC A. Z004 BECEIRO A. R007 BECHTLER C. Z007 BECKER A. LE053 BEELER K. LE092 BELLAHCENE M. LE023 BELOGLAZKINA E. V002 BELOGUROV G. R014 BELTRAN-HORTELANO I. O003 BELVISI L. R001 BELYANSKAYA S. M038 BEN OTHMAN R. L024 BENCETIC MIHALJEVIC V. M036 BENDER A. LE059 BENEK O. M025, C002 BENITEZ D. M030 BENITEZ-FERNANDEZ R. LE081 BENOWITZ A. B. M053 BENTHAM M. J. Z024 BERGANT LOBODA K. T012, T003 BERGGREN K. LE027 BERGLUND S. LE027 BERISHVILI V. H001 BERLICKI Å. T018, T022, T006,

M019, Z001 BERMEJO F. V003 BERMEO R. R001 BERNAL F. J. LE049 BERNARDES G. LE001 BERNARDI A. R001 BERNDT M. D001 BERNHARD E. T033 BERTA D. LE058 BERTONI F. T002 BERTRAND J. LE042 BESSA C. T008 BEUS M. Z023 BEYDEMIR Å. Z038 BEYER K. LE054 BEYER S. U003 BHAKTA S. Z013 BIANCHI E. Z039 BIANCOFIORE I. Z039 BIANCONI E. T041 BIASINI E. LE012

BIGNAN G.C. T035 BIKARD Y. L018 BILENSOY E. LE077 BILLAMBOZ M. L002 BINGHAM M. M024 BITTOVA L. Z034 BLADH H. M036 BLANC A. L018 BLANCO B. R005 BLANCO-ANIA D. Z031 BLANCO-LABRA A. A010 BLANKENFELDT W. R009 BLICHARZ K. M005 BLOY A. Z024 BOCEK I. Z003 BÖCKER M. M002 BOJARSKI A. J. C006, M005 BOLJE A. M006 BOLLA J.-M. C011 BOLOGNESI M.-L. E005 BOMIO-CONFAGLIA C. LE054 BONALDO B. T034 BORENSTAIN C. LE092 BORGES F. Z025 BORSARI C. T004 BOSC D. M014, L003 BOSCHI D. C014, T005, T026,

T034, T040, LE039 BOSCO M. C004, D002 BOTUBOL-ARES J.M. B001, B002 BOUBAKER G. L006 BOUDOU C. D001 BOUSBAA H. T011 BOUZIDI Y. D002 BOWER J. M041 BOYER G. C011 BOZZOLA T. A002 BRACHMANN S. LE054 BRACOTTI F. A001 BRAJSA K. M036 BRANDAO P. L004 BRAVO M. LE049 BREA J. M. Z008 BRESCIANI A. Z039 BRICELJ A. M052 BRINER K. PL001 BROCKSCHMIDT M. LE060 BRÖNSTRUP P. D. M. LE066, R002

466 | EFMC-ISMC

BROUDE E. T036 BROUGH S. Q002, L030 BROWN E. Z024 BRUDERER R. LE092 BRUNAVS M. H003 BRUNNER T. LE058 BRUNNER E. X002 BRUNST S. U003 BRUUN T. T016 BRYANT S. D. T028 BRZUSKA G. L011 BUCHHOLCZ B. W003 BUCKI A. M021, M031 BUIJNSTERS P. Z036 BÜLBÜL E. T001 BULL M. LE079 BULLER R. LE098 BUREAU R. W001 BURKE A. J. L004 BURKE J. T004 BURLEY G. A. LE069, M023, M022 BUSCA P. D002, C004, L007 BUSCH M. LE101 BUSCHINI A. T026 BUSH J. M023, M022 BUTKOVIC K. M036 BUTNARASU C. S. A001 BVS S. K. LE062 CABALLERO J. Q008 CACCIATORE I. Z002 CAFLISCH A. C016 CAGIDE F. Z025 CAI X. R003, R004 CALAMA E. LE049 CALBET M. LE049 CALISTRI A. LE039 CALLIS T. B. M007 CALOGEROPOULOU T. L022 CALVARESI M. E005 CALVET-VITALE S. L024 CAMBERLEIN V. M014, L003 CAMERON P. . LE060 CAMPANICO A. LE015 CAMPBELL E. LE069 CANABAL-FALCON R. R005 CANALE V. C006, M005 CANARD B. O004 CANDITO D. M055

CAO M. A010 CAO F. E003 CAO L. LE006 CAO J. R006 CAPECCHI A. R004 CAPOBIANCO D. A. LE103 CAPOTOSTI F. D001 CAR Å. Z022 CARDINALE A. LE046 CARLO B. M013 CAROTTI A. T041 CARRASCAL M. LE049 CARRASCO-JIMENEZ M. P. L015 CARREIRA M. L005 CARREIRA E. M. LE032 CARREL A. L. LE035 CARRIERI A. LE011 CARVALHAL F. O005 CARVALHO L. A. LE031 CASELLI E. Z018 CASTALDI P. LE092 CASTRO GONZALEZ C. B001, B002 CASTROGIOVANNI A. LE046 CATTANEO I. A004 CATURLA J. F. LE049 CECCHINI C. M008 CECERE G. PR001 CENDRON L. R008 CERADINI D. M009 CERNAK T. M055 CHALUPNIK P. Z020 CHAN S.R. T035 CHANG K. LE079 CHANG W. M055 CHANTRET I. D002, C004 CHAPMAN K. LE023 CHAPMAN N. LE023 CHAPUY E. C006 CHARALAMPOPOULOS I. X004, L022 CHARMAN S. M035 CHAROU D. X004, L022 CHAUMONT-DUBEL S. C006, M005 CHAVARRIA D. Z025 CHEN D. C003 CHEN J. LE006 CHEN P. LE006 CHEN X. LE020 CHEN Y. M038

EFMC-ISMC | 467

CHEN D. M055 CHEN M. T036 CHENG H. W. A. M007 CHENG-SANCHEZ I. C016 CHEPANOVA A. A. Z030 CHERNOBROVKIN A. LE092 CHERNYSHOVA I. A. Z030 CHIBALE K. Z041 CHO J. M032 CHOI Y. L020 CHOI P.J. M033 CHOUNTOULESI M. Z013 CHRIENOVA Z. M010 CHRISTOPHE T. LE091 CHUANG G. LE022 CHUNG C.-W. M053 CHURCHER I. M053 CIACCIO P. M055 CID J. M. X003 CIESIOLKIEWICZ A. T006 CIGNETTI A. T034, LE039 CIKOS A. M036 CIRCOSTA P. T034, T040, LE039 CLEMENT B. LE085 CLOHESSY T. M038 CLOZEL J.-P. PL002 CODERCH C. T007, T023 CODONY S. Z008 COGSWELL T. J. LE080 COIMBRA J. R. M011 COLARUSSO S. Z039 COLCHON E. R007 COLETTI A. T041 COLLET T. A. M015 COLLINA S. Q008 COMEO E. C005 COMINI M. A. M030 CONALL S. M013 CONCIATORI V. LE039 CONNOLLY P.J. T035 CONTI A. E005 CONTI G. A003 CONWAY S. LE095 COOPER J. C013 COOPER E. M033 COORNAERT B. LE091 CORIO A. L007 CORREIA-DA-SILVA M. O005

COSAR E. D. L031 COSTA B.M. T008 COSTA P. R015, A006 COSTA M. T008, T027 COSTANZO F. M008 COSTI M. P. R008 COSTI R. M042 COTESTA S. LE054 COTTO-RIOS X. LE072 COURTEIX C. C006 CRAMER J. A004, A003 CRAMPTON R. M024 CREMIN P. LE022 CREPIN T. L007 CRONIN L. (. LE045 CROSS S. R008 CROSSIGNANI S. LE101 CRUCES COLADO J. L005 CRUCIANI G. R008 CRYAN J. M053 CSOMOS A. E001 CSORBA D. E001 CUMMINGS R. O005 CUNHA E. S. LE020 CURTI D. Q008 CZECHTIZKY W. LE048 DA S. M013 DA SILVA L. S. T027 DACK K. LE052, M036 DAELEMANS D. Z004 DAHL G. LE027 DAHMEN H. LE053 DAI H. M053 DAILLER D. L013 DALLEMAGNE P. W001, X004, N003 DAMMICCO S. D005 DANON J. J. M007 DARDONVILLE C. C017 DARMENCY V. D001 DARVAS F. W003 DASCALU A.-E. L002 DAUBLAIN P. M055 DAVENPORT A. H003 DAVIES S. G. LE080 DAVIES R. LE092 DAVIS A. W001 DAWIDOWSKI M. M045 DAWSON M. J. LE063

468 | EFMC-ISMC

DE CELIS I. L008 DE COCK M. Z027 DE ESCH I. J. Z031 DE KANTER R. LE054 DE LA VILLA P. LE016 DE LEO A. M042 DE LOS REYES C. B002 DE LUCA F. R008 DE PASCUAL-TERESA B. T007, T023 DEBORA R. M013 DEGUENON P. E004 DE-JUAN-ALBERDI M. L008 DEKKER F. C003, E003 DEKKER T. Z031 DEL BUFALO D. T014 DEL OLMO E. Z037 DEL VECCHIO C. LE039 DEMBELE Y. A. L018 DEMETZOS C. Z013 DEMIR Y. Z038 DEMMANGNGEWA I. M035 DENA-BELTAN J. L. A010 DENIS C. W001 DENNY W. A. M033 DEPREZ B. M014, L003 DEPREZ-POULAIN R. M014, L003 DERNOVSEK J. T009, T039 DESAVI C. LE092 DESHMUKH P. LE062 DESIATKINA O. L006 DESTRO L. Z018 DI BELLO E. T014 DI BONAVENTURA I. R012 DI GERONIMO B. Z005 DI GIOSA M. E005 DI SANTO R. M042 DIBELLO E. M030 DIEHL C. A008 DIELS G. S. M. L012 DIEZ-ALARCIA R. LE016 DIEZ-SALGUERO M. B001, B002 DIGHE S. N. M015 DILUVIO D. G. LE103 DIMAURO E. M055 DING S. LE040 DINH C.-P. X002 DINKELBORG L. D. D001 DIPESH S. H. M047

DISTEFANO A. Z002 DISTLER J. H. LE088 DJOKOVIC N. T021 DJORDJEVIC S. A014 DOCQUIER J.-D. R008 DODD J. LE095 DOLY S. C006 DOMINGUEZ M. LE049 DOMINGUEZ Cy. LE069 DOMINGUEZ Ce. Z039 DOMINGUEZ-GARCIA S. B001, B002 DONGHOO L. M047 DORINOVA E. O. Z030 DORIS C. Z017 DORST A. L013 DRAGUNOW M. M033 DRENICHEV M. S. Z030 DREWEK I. Z027 DROP M. C006 DROZDOWSKA D. C012 DRUMMOND M. M016 DUARTE-SILVA S. T008 DUBIEL K. Z032, Z033 DUCA M. A005 DUCHARME Y. M055 DUCHEMIN N. LE033 DUDUTIENE V. M017, M057 DUNCKER D. LE023 DUNKEL P. E001 DURAES F. R015, T011 DURAN-PATRON R. B001, B002 DURANTE-CRUZ C. M018 DUSAN R. T033 DUYS I. LE091 DYMEK B. Z032, Z033 DYRKHEEVA N. S. Z030 DZIACHAN M. Z032, Z033 ECKER G. F. Q001 ECKES T. U003 EDWARDS J.P. T035 EKLUND P. C. Z040 EL PHIL R. LE094 EL SHEMERLY M. T030 ELENKOV I. M036 ELGAHER W. A. M. M034, R009 ELKAMHAWY A. M032 ELLERVIK U. A002 ELMQVIST D. LE027

EFMC-ISMC | 469

EMANUEL S.L. T035 ENGELMANN J. LE054 ENGKVIST O. LE047 EPERON I. C. LE069 ERARD M. D003 ERDMANN D. LE054 ERDMANN F. T001 ERNST B. A004, A003 ERONEN A. L009 ERRA M. LE049 ESCALA N. Z037 ESCHALIER A. C006 ESCOBAR-MONTANO F. B001, B002 ESDAR C. LE101 ESPADINHA M. T019, T024 ESPINOSA S. LE049 ESTEVES C. I. C. T011 ESTEVEZ-BRAUN A. T020 ESTHER D. O. M012 EVANGELOPOULOS D. Z013 EVANS P. A011 EVANS L. LE099 EZZANAD E. B001, B002 FACCHINI F. A. A012 FACCIOLI P. LE012 FADEYI N. LE014 FAJDETIC A. M036 FAJKIS-ZAJACZKOWSKA N. L010 FALLARERO A. M018, Z035 FALLARINI S. LE102 FATEIXA S. M043 FAULL R. M033 FAYEULLE A. L002 FECHNER N. LE060 FEDOROWICZ J. M018 FELDBAEK NIELSEN S. M036 FELIX R. LE031 FENG Y. LE092 FERACCI M. O004 FERENCZY G. G. C015 FERINGA B. L. E003 FERNANDES C. Z025 FERNANDEZ E. M053 FERNANDEZ-CARVAJAL A. M051 FERNANDEZ-PEREZ L. T020 FERREIRA K. LE066 FERRER-MONTIEL A. M051 FERRIGNO F. Z039

FERRINS L. M035 FERRON F. O004 FERRY A. L011 FETTER J. M027 FEZZARDI P. Z039 FIDALGO L. Q003 FIELDER L. LE023 FIER P. M055 FILIPIC M. T003 FILIPPO S. M004 FIORAVANTI R. T014 FISCHER W. B. Z024 FISCHER C. LE030 FITCHES E. A010 FLAUAUS C. U003 FLOHR S. LE074 FLORENCE G. J. R016 FLORENT I. Z009 FLORINDO P. A006 FLORIO T. T005 FOLLMER N. M055 FONTEYN I. X003 FORT S. D002 FOSTER R. Z024 FOSTER T. L. Z024 FRACZYK J. C012 FRANCE D. J. M041 FRANCES-MONERRIS A. O007 FRANCISCO G. M012 FRANCO A. R. A012 FRATTINI S. LE026 FRAZAO M. A006 FREITAS E. LE015 FREITAS-SILVA J. R015 FREYSE J. A014 FRIESE-HAMIN M. LE101 FRIMAN T. LE092 FRKANEC R. Z028 FROLUND B. Q001, C014 FUCHS H. LE066 FUCHSS T. LE101, LE053 FURRER J. L006 FÜSSER F. M020 GABELLIERI E. D001 GADEMANN K. L013 GAIDANO V. T034, LE039 GALA K. Z032, Z033 GALAN C. M044

470 | EFMC-ISMC

GALLIER F. L011 GAN B. H. R003 GANESAN A. T021 GAO H. D002 GARCIA S. A006 GARCIA-GASCA T. A010 GARCIA-IRIEPA C. O007 GARINO C. C014 GARLATTI L. O004 GARTLAN K. LE079 GASTREICH M. M046 GATEHOUSE J. A. A010 GATIN-FRAUDET B. D003 GAUDIO E. T002 GAVATHIOTIS E. LE072 GAWALSKA A. M021 GBAGUIDI F. A. E004 GEALAGEAS R. M014 GEHRINGER M. C009 GEISSLINGER G. U003 GEMMELL R. LE099 GEORGE S. U003 GERDT C. M055 GERIBALDI-DOLDAN N. B001, B002 GERSPACHER M. LE054 GERSTENECKER S. C009 GHINET A. L002 GIANQUINTO E. R008 GIBEAU C. M055 GIDDENS A. LE079 GIERSE R. M. M034 GIJSEN H. Z036, M029, M058 GILBERT K. M023, M022 GILBERT-GIRARD S. M018, Z035 GIORGIS M. T034, T040, LE039 GIOVANNONI J. LE054 GIRALT E. LE082, LE016 GIRARDI B. A003, A007 GITEGO N. LE072 GLADYSZ M. D004 GLYNN D. M024 GMEINER P. W002 GOBEC S. C015, M006 GOBEC M. C015, Z028 GODESI S. T010 GODINEZ-NAVARRO W. J. LE060 GOKHAN KELEKCI N. Z042, Z011, T038 GOLFOROUSH P. LE023

GOMES-DA-SILVA L. C. L016 GOMEZ E. LE083 GOMEZ-OLIVA R. B001, B002 GOMILA A. M. J. LE016 GONCALVES J. A006 GONCALVES C.S. T008 GONCALVES L. M. T024 GONZALEZ S. L011 GONZALEZ-BELLO C. R007, R013, R005,

R011, R010 GORGULLA C. T029 GOROSTIZA P. LE016 GOSAIN R. Z024 GOSMINI R. LE089 GOSS R. J. M. R016 GOTRI N. A012 GOTTI S. T034 GOUD N. M032 GOUILLER A. LE008, LE094, C010 GOULENI N. Z002 GRACIA J. LE049 GRAEDLER U. LE053 GRAUS PORTA D. LE054 GRAVANIS A. X004, L022 GRAVES L. M035 GRAVIER-PELLETIER C. D002, L024, C004,

L007 GRAZIA C. M004 GREBNER C. LE061 GRGICEVIC A. M036 GRIBAUDO G. LE039 GRIFFIN S. Z024 GRIGORIEV V. V. Q006 GROS P. X001 GRÜNENFELDER C. L029 GRYZLO B. L010 GSTAIGER M. T004 GU F. O001 GUAGLIANO G. A001 GUARDIGNI M. M034 GUARDIOLA S. LE082 GUASCH E. LE016 GUBIC Å. T025 GUEDES R. C. T028, L016 GUERRA-RODRIGUEZ M. T020 GUIANVARC'H D. D003 GUIEU S. T011 GUILLAUME V. M014 GUL S. T021, T028

EFMC-ISMC | 471

GUNDABATHULA R. LE062 GUNERKA P. Z032, Z033 GUNKEL N. C008 GUNN-MOORE F. C002 GUOFENG Q. M047 GUPTA R. LE060 GUTHY D. LE054 GÜTSCHOW M. M052 GUZELJ S. A008, Z028 HAARER L. C009 HADJIPAVLOU-LITITNA D. Q005 HAEUN K. M047 HAITJEMA C. C013 HAKANSSON M. A008 HALECKOVA A. M025 HALLS M. C005 HALVER J. LE042 HAMEL E. T002 HAMID Z. M034 HAMID A. Z007 HAMIEH A. M. C006 HAMMOCK B. D. Z008 HAMPRECHT D. LE026 HAMZA G. M. LE092 HAN T. LE006 HANDE S. LE092 HAPKO U. R009 HARDICK D. LE068 HARGROVE A. LE070 HARJIVAN S. LE015 HARKI D. A. Z029 HARLING J. D. M053 HARRIS M. Z024 HARTMAN J. LE022 HASSAN M. A008, A015 HATLAPATKOVA J. C002 HAUPENTHAL J. M034 HAY N. A006 HAYES R. M055 HEAD V. LE054 HEALY M. LE051 HECKEL A. LE026 HECKMANN B. LE091 HEERING J. U003 HEGER C. C013 HEGER Z. V004 HEHN J. LE026 HEIDI L. T030

HELESBEUX J.-J. X002 HELIÖVAARA E. L009 HELLNER J. LE092 HEMPHILL A. L006 HENDERSON T. M055 HENDRICKS J. A. LE092 HENDRICKX R. LE027 HENDRICKX L. A. T025 HEPNAROVA V. M010 HERITIER M. M008 HERLAH B. T012 HERNANDEZ B. LE049 HERNANDEZ S. O004 HERNANDEZ-GALAN R. B001, B002 HERNANDO J. LE016 HERRMANN J. R009 HESSLER G. LE061 HEVEY R. LE058 HEWITT P. LE101 HICKMAN D. D001 HIESINGER K. U003 HILL S. J. C005 HILL G. LE079 HILVERT D. LE096 HINTZE V. A014 HIRSCH A. K. H. R009, LE065, M034 HOFFMAN F. LE054 HOFMANN B. U003 HOGNON C. O007 HOK L. Z003, Z004 HOLLINGWORTH G. J. M044 HOLUBOVA K. M010 HOLVEY R. T013 HOOGEWIJS K. LE073 HOPKINS E. LE099 HOPMANN K. H. L017 HORAK M. M010 HOSSAM H. T010 HOTINGER J. M028 HOTOP S-K. LE066 HOUINATO M. R. E004 HOUNGBEME A. G. E004 HOUSHMAND M. T034 HOUSSEMAN C. H002 HOUVENAGHEL N. LE091 HRANJEC M. Z003, Z004 HRISTOZOV D. H003 HSIAO M.-Y. M029, M058

472 | EFMC-ISMC

HUANG N. LE006 HÜBNER S. LE002 HÜBNER H. W002 HUGHES J. M055 HUGHES S. X001 HUMMEL-EISENBEISS J. C008 HURLEY L. LE069 HUTTUNEN K. M. LE087 HWEE D. LE022 IALONGO D. M042 IBRAHIM H. T001 IBRAHIM S. T035 IEREMIAS L. U001 IHNATENKO I. M030 IJZERMAN A. P. AL001 ILARIA P. M013 ILINA P. T016 IMBERTY A. R001, LE057 INTROVIGNE M. L. Z018 IRIEPA I. O007 ISERT C. M002 ISKAUSKIENE M. U002 IVANENKOV Y. V002 IVANOV G. A. Z030 IVANOVA J. T015 IVANOVIC M. Z010 IWATA T. L019 JACKSON T. R. LE080 JACKSON-FLUX H. LE060 JACOBY E. Z036 JAISWAL J. LE079 JAKOB R. P. A004 JAKOPIN Å. A008, Z028 JAKUBCZYK M. D004 JAMIESON S. LE079 JAMIESON C. M038 JAMROZIK M. M031 JANEZIC M. T012 JANOVSKA L. U002 JANSSEN M. A. Z031 JARVIS A. LE023 JAVOR S. R003, R012 JEDLICKA P. Z034 JELIC D. M036 JENKINS L. U001 JENNIFER D. L. V. F. M012 JENSEN K. F. LE044 JERHAOUI S. L012

JESURAJ N. C013 JEVTIC I. Z010 JIANG X. A004 JIANG F. M053 JIMENEZ NUNEZ E. LE054 JIMENEZ-LUNA J. M002 JIMENEZ-SANCHEZ A. O002 JINDRA M. Z034 JOANNESSE C. LE091 JOCHMANS D. Z030 JOHANNSEN S. M034 JOHANSSON J. LE027 JOHNSON A. M028 JONCKERS T. H. M. LE036 JORDAAN A. LE015 JORGENSEN W. M007 JOSA-CULLERE L. E002, LE080 JOSE J. M033, LE079 JOSE ANTONIO O. M013 JOURAVEL G. M040 JUAN F. S. T033 JUCHUM M. LE071 JUERGENS T. D001 JUHAS M. Z016 JUKIC M. R014 JULLIEN L. D003 JUMDE R. P. M034 JUNG E. L013 KADLECOVA A. U002 KAIRYS V. M017, M057 KAISER A. U003 KANIAKOVA M. M010 KANKANALA J. Z024 KAPIC S. M036 KARGE B. LE066 KARKI R. LE074 KARLOV D. S. Q006 KASPERSEN M. U001 KASSIOU M. M007 KATARINA N. T033 KAUFMANN L. Z007 KAUR D. L030 KAUR G. Z041 KAWAMURA S. M055 KEARNS J. LE054 KEKEZOVIC S. Z015 KELES E. T004 KELLAM B. C005

EFMC-ISMC | 473

KELLENBERGER L. T030 KELLEY T. LE103 KELLY J.M. O003, Z013 KEMINER O. T028 KEMPF G. Z039 KENNELLY S. A. Z029 KERR W. J. LE090 KESERU G. M. C015 KHAN A. LE099 KHAZANOVA E. V002 KHÖLER T. R012 KHRESTCHATISKY M. W001 KICKINGER S. Q001 KILIC-KURT Z. T026 KILKE K. M026 KIM Y. S. L014 KIM S. H. L020 KIM M. T010, L020 KING B. J. Z024 KIRILLOVA M. C016 KIRK R. M024 KIRPOTINA L. N. LE011 KIURU P. T016 KLANCIC V. Z028 KLEIN S. A004 KLEIN M. LE101 KLETECKOVA L. M010 KLETT J. Z005 KLEY J. T. LE026 KLOTZ S. LE071 KLÖVEKORN P. LE071 KLUG D. M035 KNAPP S. PL004 KNEZ D. C015 KOBAURI P. E003 KOBRLOVA T. M010 KOCH O. M020 KOCZURKIEWICZ P. C006, M005, M031 KOEBERLE A. X002 KOEHLER L. A014 KOHLER T. R003, R004 KÖHLING S. A014 KOJIC V. Z015 KOLACZKOWSKI M. M021, L010, M031 KOLANOWSKI J. L. D004 KOLB E. LE103 KOLESINSKA B. C012 KOLLAR L. C015

KOLOCOURIS A. Z013 KOMSTA L. Z006 KONOPACKA A. M053 KONSEWICZ K. D004 KORABECNY J. M010 KOSTIC-RAJACIC S. Z010 KOSTRUN S. M036 KOTOULA V.-M. Z012 KOUROUNAKIS A. Q004 KOVACEVIC M. Z022 KOVACHKA S. T026 KOVALL R. LE103 KOWALSKA T. Z006 KOZLOVA A. A. Z030 KRAJEWSKA J. L023 KRAMER J. S. U003 KRÄMER O. H. T001 KRAUPNER N. L003 KRAWCZYK M. M005 KRETSCHMER S. U003 KRIER M. LE053 KRIUKOVA A. H002 KROEZEN B. A003 KROL E. L011 KRÖNKE J. M052 KROSKY D.J. T035 KROTH H. D001 KRUNIC M. Z010 KRUPA A. L010 KUBAS H. LE053 KUBIAK R. M055 KUCERA T. M010 KUCZAK M. M039 KUDOVA E. L028, M037 KUHN D. LE053 KUMARI P. M056 KÜMMEL D. M020 KUNICK C. M030 KURCZAB R. M005 KURET K. R014 KVARATSKHELIA M. M042 KWAK D. T010 KYEONG L. M047 LACIVITA E. LE011 LAHTELA-KAKKONEN M. T021 LAINO T. LE046 LAKKAICHI A. A004 LAKSHMINARAYANA S. B. O001

474 | EFMC-ISMC

LAL K. R001 LALUT J. W001 LAMATY F. C006 LAMBERT M. M036 LAMBRIS J. D. Z007, X001 LAMERS C. X001 LÄMMERMANN H. LE002 LANARI D. L015 LANGER L. LE066 LANGER T. T028 LANINI J. LE060 LAPLANTE S. Z026 LAUDY A. E. L023 LAUFER S. C009, LE071 LAUL E. C016, V001 LAURENT S. D006, Z027 LAUS A. C. T027 LAVREYSEN H. X003 LAVRIK O. I. Z030 LAVROV M. I. Q006 LAW R. LE079 LE CORRE L. L007, L024 LE SAUX T. D003 LEBLANC C. LE054 LEBOHO T. M001 LEE H. Y. L020 LEE S. M055 LEE K. T010, L020, M032 LEFFLER H. A008 LEHNER M. LE049 LEMAIRE C. D005 LEMMEN C. M046 LEMOINE H. M040 LEMPINEN A. T016 LEMURELL M. AL002 LENAERTS I. Z036 LENCE E. R011, R010, R005 LEOPOLDO M. LE011 LEPISTÖ M. LE027 LEROUX F. M014 LESIRE L. M014 LEUNG E. LE079 LEWIS R. LE060 LI D. LE006 LIARGKOVA T. Q005 LIE M. E. Q001 LIM J. M055 LIM H.K. T035

LINCIANO P. Q008 LINN D. M055 LIPKA E. L003 LIU N. LE006 LIZANDRA PEREZ J. T018 LLAVERIA J. X003 LLEBARIA A. E002 LOHARCH S. M019 LOHMANN S. LE042 LOI E. M. A009 LOLLI M. C014, T005, T026,

T034, T040, LE039 LONDON N. PR004 LOPES F. LE015 LOPES E. A. M003, T019 LOPEZ-ALVARADO P. Z014 LOPEZ-CARA L. C. L015 LOPEZ-ROJAS P. T020 LORENTE MACIAS Ã. M035 LORTHIOIS E. LE054 LOURENCO L. M. O. M043 LOWERSON A. Q002, Z026 LOZA I. Z008 LUBIN-GERMAIN N. L011 LUCAS S. D006 LUDOVICO P. T008, T027 LUECKE H. LE020 LUGANINI A. LE039 LUIS R. M012 LUISTRO L.L. T035 LUKAT P. R009 LULINSKI S. L023 LUQUE NAVARRO P. M. L015 LURAGHI A. A012 LUSTENBERGER P. LE074 LUXEN A. D005 LUZ A. F. S. L016 LV L. LE006 MACCHIARULO A. T041 MACDONALD A. Z024 MACFARLANE K. M038 MACHACEK M. M055 MACHAUER R. LE054 MACHESKY L. M041 MACHULKIN A. V002 MACIAS SANCHEZ A.J. B001, B022 MACIEL P. T008 MACOR J. PR005

EFMC-ISMC | 475

MADDER A. LE073 MAH R. LE054 MAI A. T014 MAIER T. A004 MAIER J. LE071 MAIN M. LE092 MAINOLFI N. LE074 MAIRA M. LE054 MAJER P. Z034 MAJOUGA A. V002 MÄKKYLÄ H. M018, T016 MALARZ K. M039 MALDONADO M. LE049 MALIK F. LE022 MALINAK D. V004 MALINAUSKIENE V. U002 MALISZEWSKI D. C012 MALLART S. LE021 MALLO-ABREU A. LE031 MALWAL S. R. Z013 MALZERT-FREON A. W001 MAMMOLI A. T041 MAMMOLITI O. LE091 MANANDHAR A. U001 MANEIRO M. R011, R007 MANICA M. LE046 MANKOURI J. Z024 MANLEY P. LE054 MANNA M. A007 MANNELLA I. T026 MANNISTO J. L009, L017 MANOHAR R. LE099 MANOHARAN M. LE076 MANSUETO M. M055 MAPLE H. C013 MARAJ MARTINEZ M. LE058 MARAZZI M. O007 MARCHESE S. T029 MARCINIAK M. M045 MARCINKOWSKA M. L010 MARCO C. L015 MARIA M. M004 MARIN C. O002 MARIN J. J. V003 MARIN P. C006, M005 MARINOVIC M. M048 MARQUES M. A006, T028 MARQUES C. L004

MARQUES V. L016 MARQUEZ-CANTUDO L. T007 MARRAUDINO M. T034 MARRINK S. J. E003 MARTIN P.-Y. C006 MARTIN N. LE023 MARTINEZ J. C006 MARTINEZ A. LE081 MARTINEZ MOLINA D. LE092 MARTINEZ-ALARCON D. A010 MARTINEZ-GONZALEZ L. LE081 MARTINEZ-VITURRO C. M. X003 MARTINI R. T028 MARTINO E. C014 MARTIN-REQUERO M. LE081 MARZAG H. L027 MASO L. R008 MASSARI M. T029 MASSAROTTI A. LE102 MASSE J. L005 MASSIGNAN T. LE012 MASSIMILIANO R. M004 MASTROMARINO M. LE011 MATERA C. LE016 MATILDA Å. Z017 MATTEI P. LE064 MATTER H. LE061 MATTEVI A. T029 MATTHEW M. M015 MATULIS D. M017, M057 MAY L. T. C005 MAY A. M028 MAYE M. D002 MAZIARZ K. LE060 MAZUR L. M018 MAZZANTI M. T005 MBOMA R. E004 MCCARREN P. Z026 MCGREGOR L. LE054 MCPHAIL J. T004 MCPHERSON R. M024 MEANA J. J. LE016 MEE E. M033 MEHTA A. O005 MEIBOM D. LE086 MEIER C. N002, T032 MEMARZADEH S. M041 MENENDEZ J. C. Z014

476 | EFMC-ISMC

MERKUS D. LE023 MESSER J. M038 MESSORE A. M042 MEZAACHE R. L018 MEZOHEGYI G. W003 MICHALEK S. Z032, Z033 MICHELE P. M004 MICHELE V. M004 MICLOT T. O007 MIELE E. LE092 MIHOVILOVIC M. D. LE097 MILACEK M. Z034 MILKOVIC L. A013 MILLA-NAVARRO S. LE016 MILLER A. K. C008 MILLER C. LE079 MILLIGAN C. T035 MILLIGAN G. U001 MILLS D. J. LE020 MILNE T. LE080 MILOS P. T033 MIN S.-J. L014 MIN K. H. L020 MIR M. LE049 MIRALPEIX M. LE049 MIRET-CASALS L. LE073 MISZTELA H. LE060 MLINARIC-RASCAN I. Z028 MM V. LE062 MOGNETTI B. LE039 MOLETTE J. D001 MOLINS E. T024 MÖLLER S. A014 MOLOGNI L. Z018 MON KALITA M. Z024 MONARI A. O007 MONCK N. H003 MONJA N. L008 MONTALBANO A. T002 MONTALBANO S. T026 MONTEIRO Ad. R005 MONTEIRO Ana. R. M043 MONTGOMERY A. P. M007 MONTI B. LE009 MOORE S. E. C004, D002 MOORTHY R. Z029 MOQUIN S. A. O001 MORALES J. C. M044

MORAWSKA M. M018 MOREIRA P. I. M011 MOREIRA R. LE015, L016, LE031,

A006, Q003 MORENO M. Z005 MORGAN B. LE022 MORI M. M003, V003 MORISSEAU C. Z008 MORRIS M. A011 MÖSCHING M. L006 MOSLOVA K. L009 MOTIEI L. LE004 MOURAY E. Z009 MOUROT L. Z009 MRAVLJAK J. A007 MROZ P. M045 MROZEK-WILCZKIEWICZ A. M039 MUCSI Z. E001 MUELLER A. M. D001 MULARSKI J. M039 MULDER F. C003 MULLER Robert D006 MÜLLER A. C016 MÜLLER M. J. M030 MÜLLER Rolf R009, L013 MULROONEY C. M038 MUNOZ-SANJUAN I. Z039 MUSIL D. LE101 MUSILEK K. M025, V004, C002 MUSIOL R. M039 MUTHU M. Z034 NABERGOJ S. Z028 NACHAMPASSAK S. L019, L021 NAFFAKH N. L007 NAR H. LE018 NARDUCCI D. L022, M024 NASSOY A.-C. M024 NAZARE M. D007 NAZAROVA A. A. Q006 NAZIRIS N. Z013 NEHLS C. M034 NELSON G. M024 NEPOVIMOVA E. M010 NERI D. LE075 NEUKIRCH K. X002 NEUMANN A. M046 NEUSENS P. J. M034 NEVADO C. C016, V001

EFMC-ISMC | 477

NEVES M. G. P. M. S. M043 NEVES A. R. O005 NEWTON G. LE023 NEYTS J. Z030 NG Y. L. D. M052 NGUYEN J. LE002 NICHOLSON B. M055 NICOUD C. M040 NIE L. Z021 NIEGER M. L009, L017 NIKOLIC K. T021 NILSSON M. LE027 NILSSON U. A007, LE056, A008,

A002, A015 NOCEN P. T022 NOGUCHI K. M033 NORSIKIAN S. D003 NORTON R. LE079 NOVAIS P. T011 NOVAK JOVANOVIC I. Z003 NOVOTNA A. Z034 NOWICKI K. L023 NOZAL V. LE081 NUE MARTINEZ J. J. C017 NUIJENS T. LE034 NUNEZ-ABADES P. B001, B002 NXUMALO W. M001 NYTKO K. LE099 OBRADOVIC D. Z006 O'CONNOR P. LE079 ODENB F. O. D001 O'HAGAN M. M044 OLDFIELD E. Z013 OLIARO BOSSO S. T026 OLIVEIRA-PINTO S. T008, T027 OLIVER M. L024 OLMO F. O003 OMBOUMA J. G. E004 ONGERI L. M024 ONTORIA J. Z039 OP S. M038 OPREA T. O006 ORLOWSKA N. Z032, Z033 ORTHOLAND J.-Y. M040 ORTIN I. T007, T023 ORTIZ C. M030 ORTYL J. C007 OSLOVSKY V. E. Z030

OSOLODKIN D. I. Z030 ÖSTERLUND T. LE027 OSTERMANN N. LE054 OTERO J. M. R011 OTTL J. LE054 OUARTI A. L011 ÖZCELIK A. B. Z038 OZGA K. T006 OZIMEC LANDEK I. M036 OZKAY Y. Z042 PACHECO P. A.F. T024 PACHOLAK P. L023 PACIFICI R. Z039 PADOVAN J. M036 PAGES J.-M. C011 PAGES L. LE049 PAGONI A. Z001 PAIK S.-J. C004 PAJK S. Z028 PAKETURYTE V. M057 PALAFERRI L. C016 PALAZZESI F. H003 PALE P. L018 PALEYES N. LE060 PALISSE A. LE091 PALLOTTA M. T. LE102 PALMEIRA A. R015, O005 PALOMO V. LE003, LE081 PALYULIN V. A. Q006 PAMPIN CASAL B. L005 PANNILUNGHI S. C010 PAPADOPOULOU M. X004 PAPAGIOUVANNIS G. Q007, Z012 PAPOT S. LE078 PARDO L. A. T025 PARIKKA M. Z023 PARIS S. LE049 PARK K. D. M032 PARK T.I.-H. M033 PARK L. Z039 PAROLIN C. LE039 PASSONI A. T040, LE039 PATEL M. M055 PATEL P. T016 PATEL S. T035 PATEL J. Z035 PATSILINAKOS A. Z039 PATTAROZZI A. T005

478 | EFMC-ISMC

PAUL N. M041 PAULAMÄKI L. Z023 PAUNESCU E. L006 PAUREVIC M. A013 PAVIC K. M050 PAVLOVIC L. L017 PAWLOWSKI M. C006 PEHL U. LE053 PEKALA E. C006, M005, M031 PENALVER P. M044 PENDERGRASS H. M028 PENJISEVIC J. Z010 PENNINCKX S. D006 PERDIH A. T003, T012 PEREZ C. Z008, L008, V003 PEREZ-PEREZ M.-J. LE037 PEREZ-PRIETO I. L015 PEREZ-SILANES S. O002, O003 PERI F. A012 PERKOVIC I. M049 PERMANN S. X002 PERNAS M. R005 PERRIOR T. LE023 PERRON Q. H002 PERRONE C. T014 PERSONNE H. R012, R004 PERSOONS L. Z004 PESIC D. M036 PESNOT T. M024 PETERLIN MASIC L. T025 PETERMANN O. LE008, LE094 PETKOVIC M. T021 PETRINI P. A001 PETRINIC GRBA A. M036 PETROV S. V002 PETROVIC PEROKOVIC V. Z022 PETTER J. LE067 PEUKERT C. LE066 PEVIANI M. Q008 PFEIFER P. D001 PFEILSCHIFTER J. U003 PHILLIPS J. LE050 PHIPPS R. LE099 PICOTTI P. LE093 PIECZYKOLAN J. Z032, Z033 PIETERS R. A003, A005, A009 PIETKA A. Z027 PIETRANCOSTA N. L007

PIETRUÅš W. M005 PIKUSA M. LE060 PINEDA L. Z037 PINEIRO M. L004 PINKERTON J. LE027 PINTO C. L016 PINTO E. R015, L004 PINTO M. R015, T011 PIOU T. M055 PIPPIONE A. C. T005, T026, T034,

T040, LE039 PIQUERO M. Z014 PIRALI T. M051, LE102 PISABARRO M. T. A014 PISKA K. M031 PITA-ALMENAR J. D. X003 PLAVEC J. Z028 PLE K. L027 POGORYELOV D. U003 POINSOT M. L024 POJE G. M049, M050 POLI S. D001 POLSHAKOV V. V002 POMELLA S. T014 PONTES O. T008, T027 POON V. LE099 POPIK P. M005 POPSAVIN M. Z015 POPSAVIN V. Z015 POUTON C. LE043 POUW R. Z007 POZZAN A. H003 PRAEFKE B. LE071 PRCHALOVA E. V004 PREDA B. LE016 PRISCHICH D. LE016 PRITCHARD J. M. LE090 PROCHNOW H. LE066 PROENCA F. T008, T027 PROJ M. C015 PROSCHAK E. U003 PROTOPOPOV M. M054 PROUSIS K. C. L022 PRUDENT R. M040 PUCHER M. D003 PUSHECHNIKOV A. H001 PUSKAS I. W003 PUTHANVEEDU M. LE042

EFMC-ISMC | 479

PYATI P. A010 QI X. LE006 QIAN D. M053 QILI L. M047 QUEISSER M. A. M053 QUILIANO M. Z019 QUINN M. T. LE011 QUIROZ1 R. M055 RABBANI S. A004, LE058, A003 RADCHENKO E. V. Q006 RADEMACHER C. LE055 RADEMANN J. A014 RAIMONDI M. V. T002 RAJIC Z. M048, M049, M050 RAM MAHATO D. Z024 RAMILO-GOMES F. T028 RAMIREZ L. LE016 RAMOS C. I. V. M043 RAMOS A. T007, T023 RANKIC D. M055 RASHEED S. R009 RASTEGAR S. LE042 RATHMELL R. H003 RAUH D. LE100 RAUTIO J. LE084 REEKS J. LE019 REGUERA J. O004 REIGADA I. Z035 REILING N. M034 REIS R. M. T027 REIS J. T029 REITER L. LE092 REKKA E. A. Q007, Z012 REMSKAR M. L013 REMUS S. N002, T032 RENATA O. Z017 REPO T. L009, L017 RESSURREICAO A. S. Q003 REUTERSHAN M. M055 REVOL-TISSOT J. C011 REX S. V004 REXEN ULVEN E. U001 REYMOND J-L LE035, R003, R012,

R004 REYNOLDS J. LE095 RHODEN K. E005 RIBIC R. A013, Z022 RICCHIUTO P. LE092

RICCIO A. T041 RICHALET F. T030 RICHELLE G. LE034 RICHOMME P. X002 RICKERSHAUSER L. L025 RICKLIN D. LE058, A007, A003,

Z007, X001 RIEGE D. LE042 RIGO B. L002 RIGOLLIER P. LE054 RIKHOTSO D. M001 RITA MARIA CONCETTA D. M. M013 RITTINGER K. M023, M022 RIVERA S. W001 RIVERA-FUENTES P. LE002 ROBAY D. T030 ROBERTSON L. LE022 ROCHAIS C. W001, X004, N003 ROCHE D. M040 RODIC M. Z015 RODRIGUES J. M043 RODRIGUES C. A006, Q003, L016,

T019 RODRIGUEZ D. R007 RODRIGUEZ Ã. R013 RODRIGUEZ FRANCO M. I. Z008 ROEHRIG S. LE025 ROGDAKIS T. X004, L022 ROH J. M032 ROHDICH F. LE101 ROLANDO B. T026 ROLLINS K. C016, V001 ROMAN D. LE054 ROMANELLI A. T014 ROMBOUTS F. J. R. L012, M029, M058 ROMERIO A. A012 ROMERO M. R. V003 RONINSON I. T036 ROSS T. LE066 ROSSI M. E005 ROSSI D. Q008 ROSSINO G. Q008 ROSSMEIER M. T032 ROTA R. T014 ROTHER S. A014 ROUTIER S. L027 ROWAND J. T035 ROWLANDS D. Z024 RUBEN A. M013

480 | EFMC-ISMC

RUBIO-HERNANDEZ M. O003 RUI M. Q008 RUIZ-GOMEZ G. A014 RUMANO M. X001 RUPCIC R. M036 RUSSELL A. J. LE080, LE041 RUTHERFORD O. I. LE069 RUTJES F. P. Z031 RUZIC D. T021 RYBAK M. M054 RZEPECKA J. M024 SAADABADI A. Z040 SAAL C. LE053 SABATE M. LE049 SACKUS A. U002 SACZEWSKI J. M018 SAGLIK B. N. Z042 SAGLIO G. T034, T040, LE039 SAHARI A. L017 SAHRAOUI S. S. LE008 SAINAS S. C014, T026, T034,

T040, LE039 SAITO N. L019, L021 SALO-AHEN O. R014, Z040 SALVADOR J. A. M011 SAMA A. M035 SAMPAIO-MARQUES B. T008, T027 SAMSON A. Z024 SANCHEZ DE BLAS B. V003 SANDER P. L013 SANDERSON M. LE101 SANGÜESA G. LE016 SANNIO F. R008 SANTIBANEZ J. F. T021 SANTOS M. M. M003 SANTOS A. E. M011 SANTOS D. J. D. A. T019 SANTOS M. M. M. T019, T024 SANTUCCI M. R008 SANYAL S. M055 SANZ-GAITERO M. LE020, R011 SANZ-SERRANO J. O003 SAPA M. M031 SARABIA Ã. Z014 SARAIVA L. M003 SARIMAN M. T038 SATALA G. C006, M005 SAVIJOKI K. M018, Z035, Z023

SAXTY G. M036 SCAMMELLS P. J. C005 SCAPOZZA L. LE008, C010, M008 SCHADE D. LE042 SCHADT O. LE101 SCHÄFER A. T004 SCHÄFLE D. L013 SCHARNWEBER D. A014 SCHÄTZLEIN A. M035 SCHEEREN H. W. Z031 SCHEFFLER F. LE099 SCHEPENS W. T035 SCHEPETKIN I. A. LE011 SCHEPMANN D. Q008 SCHIEFERSTEIN H. D001 SCHIEMANN K. LE053 SCHIRLE M. LE054 SCHMIDT M. C002 SCHMIDT M. T001 SCHMIDT C. Q. Z007 SCHMIDTKO A. U003 SCHMITT M. Z009 SCHNABELRAUCH M. A014 SCHNEIDER M. LE023 SCHNEIDER N. LE054, LE060 SCHNEIDER S. M055 SCHNEIDER G. M002, AL003 SCHUBART A. LE074 SCHUBERT-ZSILAVECZ M. U003 SCHUMACHER U. N002, T032 SCHÜRER S. LE103 SCHUTKOWSKI M. T001 SCHWALLER P. LE046 SCHWALM S. U003 SCHWARDT O. A004, A007, A003 SCHWEDER P. M033 SCIPIONE L. M042 SCOTT A. M024 SCOTT C. Z024 SEDENKOVA K. N. Q006 SEDLAK D. Z034 SEDRANI R. LE054 SEENISAMY J. LE101 SEGLER . M. LE060 SELIG R. LE071 SELLNER H. LE074 SENDER M. M038 SERAFINI M. M051

EFMC-ISMC | 481

SERAPHIN D. X002 SERENA M. M004 SERRANO-WU M. Z026 SHAFFER P. T035 SHAFIKOV R. V002 SHAIK M. A012 SHAILESH T. M013 SHANU-WILSON J. LE099 SHAW J. Z024 SHAW D. LE051 SHERAZ G. M004, T033 SHI E. M038 SHRESTHA B. LE054 SHTIL A. T036 SHUBIN K. M009 SIBILLE G. LE039 SIEBER St. AL004 SIEROCKI P. M014 SIEVERS S. LE042 SILIPHAIVANH P. M055 SILVA P. M. A. T011 SILVA OTERO R. Z029 SILVERMAN S. M055 SIMON O. O001 SIMONA R. M004 SIPPL W. T001 SIROCKIN F. LE074, LE060 SIVARAMAN A. L020 SKAANDERUP P. LE054 SKAK-NIELSEN T. M036 SKARKA A. V004, C002 SKET P. Z028 SKRENKOVA K. M010 SKVORTSOV D. V002 SLOMAN D. M055 SLÜTTER B. Z028 SMAILAGIC A. LE027 SMIESKO M. LE058, X001 SMIETANA M. LE033 SMIRNOV A. M017 SMITH M. T035 SMITH J. T037 SNAJDR I. Z034 SOARES P. Z025 SOBCZYK A. LE046 SOBRAL L. T028 SOHAJDA T. W003 SOKLOU K. E. L027

SOLARTE C. L011 SOLLNER DOLENC M. T003 SOMERS M. Z036 SONCK K. LE091 SONDEY C. M055 SOSIC I. C015, M052 SOUKUP O. M010 SOURAL M. M005 SOUSA S. F. Z025 SOUSA E. R015, T011, O005 SOUTHALL S. LE017 SPADAFORA C. Z037 SPAGNOLLI G. LE012 SPANO V. T002 SPENGLER G. R015 SPICER J. LE079 SPYRAKIS F. R008, T026 SRDIC-RAJIC T. T021 SRECO ZELENOVIC B. Z015 SRIRAMARATNAM R. T004 STAHLHUT M. M036 STAMPAR M. T003 STAMPOLAKI M. Z013 STANICKI D. D006, Z027 STAPLETON P. M035 STARCEVIC K. Z003 STAROSYLA S. M054 STAZI G. T014 STEADMAN V. LE062 STEELE J. LE099 STEFANIA G. M013 STEFANIA D. M013 STEIMBACH R. R. C008 STEINBACHER S. Z039 STEINEBACH C. M052 STEINHILBER D. U003 STEPHENS A. D001 STERKER D. LE054 STIEFL N. LE060 STIKONAITE V. R016 STIMAC A. Z028 STOCKER A. R012 STOJKOVIC R. A013 STOUSE A. D005 STREEFKERK D. LE034 STREET S. S. M044 STRINGER R. LE054 STRIPPOLI R. T014

482 | EFMC-ISMC

STUTZ S. LE054 STYPIK M. Z032, Z033 SUBRA G. C006 SUBRAMANIAN M. X003 SUKUPOVA M. V004 SUN Y. LE006 SUNDIN A. A008, A015 SURKYN M. L012 SUZANNE P. W001 SWAIN C. M035 SWALM B. M055 SZABO D. E001 SZANTI-PINTER E. L028 SZEMEREDI N. R015 SZENTE L. W003 SZILAGYI B. C015 SZYMANSKA E. Z020 SZYMANSKI W. E003, LE013 SZYMASZEK P. C007 TABEY C. M014 TABOR W. M019 TAJMOUATI H. H002 TALADRIZ-SENDER A. LE069 TALTAVULL J. LE049 TAME C. J. M053 TAMMELA P. T016, M018 TARDY S. LE008, LE094, C010,

M008 TARR A. W. Z024 TASCI H. Z042, T038 TATARSKIY V. T036 TATJANA S.-R. T033 TE LINTEL HEKKERT M. LE023 TEMNYAKOVA N. S. Q006 TEMPRANO A. G. V003 TERNAD I. D006 THALLMAIR S. E003 THEODOSIS-NOBELOS P. Q007, Z012 THEURILLAT J.-P. M008 THOMPSON J. D. LE090 THUM T. LE024 THURING J.W. T035 TIAINEN T. L017 TIEDT S. W002 TIMMERMAN P. LE034 TINWORTH C. P. M053 TIZIANO B. M013 TODD M. M035

TOLEDO SHERMAN L. Z039 TOMASIC T. A007, A008, A009,

A015, T009, T025, T039

TOMIC S. A013 TOMLINSON R. C. LE092 TONDI D. R008 TONIATO A. LE046 TOPLAK Å. T025 TORRENTE E. Z039 TOZKOPARAN B. Z042, Z011, T038 TRAPANI J. LE079 TRESADERN G. Z036 TRINDADE T. M043 TRIPODI M. T014 TRISCIUOGLIO D. T014 TROJAN E. LE011 TSE E. M035 TUKALO M. M054 TUMA R. Z034 TUMOVA S. Z034 TURNER D. M024 TUROWSKI P. Z032, Z033 TUTOV A. LE066 TYLEK K. LE011 TYTGAT J. T025 TZARA A. Q004 TZVETKOV N. Q002 UGEL S. LE102 UL-AIN Q. LE060 ULIASSI E. E005 ULUKAYA E. Z011, T038 ULVEN T. U001 UMNYAKOVA E. Z007 URBAN D. D003 USPENSKAYA A. V002 UZIEL J. L011 VADAS O. LE008, LE094 VAIDEANU A. M035 VALENTE S. T014 VALES K. M010 VALJAVEC K. T012, T003 VALVERDE E. Z008 VAN ALLER G. T035 VAN DE PLASSCHE M. C001 VAN DELDEN C. R012, R004 VAN DER LINDEN I. X003 VAN DER STELT M. LE029 VAN KLAVEREN S. A015

EFMC-ISMC | 483

VAN MAARSEVEEN J. LE034 VAN RAAIJ M. J. R011 VAN ROOSBROECK Y. E. M. L012, Z036 VAN TROYS M. LE073 VANDA D. M005 VANGIJZEGEM T. D006 VANHOUTTE R. C001 VANNECKE W. LE073 VARISCO Y. D001 VARROT A. R001, A010, A005 VASILENKO D. A. Q006 VASKOVA M. M025, C002 VASSILIOU S. Z001, Z002 VASTAKAITE G. L029 VAUCHER A. LE046 VAUPEL A. LE054 VAUZEILLES B. D003, A016 VAZQUEZ S. Z008 VAZQUEZ-UCHA J. C. R007 VECCHIO A. T001 VELTER A. I. Z036 VENTURELLI A. R008 VER DONCK L. Z036 VERDIROSA F. R008 VERDU M. LE049 VEREMEEVA P. N. Q006 VERGALLI J. C011 VERHELST S. C001 VESER T. LE026 VIALKO A. Z020 VIANELLO R. Z003, Z004 VIAULT G. X002 VIDALAIN P.-O. L007 VIEIRA A. C. L016 VIELLEVOYE M. T035 VIGATO C. T040 VILASECA M. LE073 VILLELLA N. T005 VINALS M. LE049 VINCENT S. R006 VISAI L. A001 VISENTIN S. A001 VITALE R. M. T005 VOGSEN HEIDTMANN C. U001 VOKALI E. D001 VOLLER J. U002 VOLYNETS G. M054 VOS A. X003, M029, M058

VOSHOL H. LE054 VUORINEN A. LE080, M023, M022 VYAS P. LE080 WAGNER B. X001 WALCZAK M. C006 WALDMANN H. LE042 WALLIN M. C013 WALSE B. A008 WALTER-BAUSCH G. LE101 WAN H. L024 WAN M. M055 WANG H. LE006 WANG X. LE006 WANG Y. LE006 WANG J. LE022 WANG F. O001 WANG Y. R016 WANG M. T019, M003 WANG S. T019, M003 WAREMGHEM S. M014 WARNER D. LE015 WATT G. M053 WATTIEZ R. Z027 WEIBEL J.-M. L018 WEILER S. T030 WEISS Matjaz A009 WEISS Manfred M020 WEISS A. LE054 WEIZEL L. U003 WELDON C. LE069 WELLENDORPH P. Q001 WENNEMERS H. L029, L026 WERRY E. L. M007 WERZ O. X002 WESSELER F. LE042 WESSELS P. LE054 WETHERILL L. Z024 WHISSTOCK J. LE079 WICHERS T. LE058 WIDMER T. LE054 WIECZOREK M. Z032, Z033 WIEDENMAYER D. LE026 WIEDMER L. C016 WIJTMANS M. Z031 WILCKEN R. LE054 WILLICHB H. S. D001 WINSKA P. L023 WITT J. N002

484 | EFMC-ISMC

WITTE M. D. E003 WITTER D. M055 WITTMANN S. K. U003 WOLBER G. T012, T003 WOLF H. LE046 WOLLENHAUPT J. M020 WONG V. T035 WOO J. M032 WORKMAN P. LE028 WRIGLEY S. LE099 WROBEL A. C012 WU Y. LE022 WUENSH B. Q008, LE010 WYCHOWANIEC A. D004 WYMANN M. T004 WYNNE G. M. LE080 XIAO Z. C003 XU H. M055 XU Z. M055 YAN R. LE023 YANG S. LE007 YARMOLUK S. M054 YE Y. M055 YEUNG C. M055 YEUNG B. K. S. O001 YILMAZ C. L022 YLI-KAUHALUOMA J. T016, Z023, Z035 YOKOKAWA F. O001 YU Z. LE005 ZACHARIOUADAKIS E. LE072 ZACHAROVA M. K. R016 ZAGOZDA M. Z032, Z033 ZAJDEL P. C006, M005 ZAJEC Å. T009, T039 ZAKARIA A. L014 ZAKHARENKO A. L. Z030 ZAKHAROVA O. D. Z030 ZALA A. M056 ZALUBOVSKIS R. T015 ZAMBON A. Z018 ZAMORA J. LE022 ZAMOYSKI V. L. Q006 ZANELLI U. LE101 ZAPICO J. M. T007, T023 ZDAROVA KARASOVA J. C002 ZECRI F. LE054 ZEGURA B. T003 ZEMANOVA L. M025, C002

ZENCHENKO A. A. Z030 ZENDER L. LE071 ZENGIN KURT B. L031 ZESSIN M. T001 ZETSCHOK D. L026 ZETTERBERG F. A008 ZEYN Y. T001 ZHANG Jinlei LE033 ZHANG D. LE080 ZHANG Jian LE005 ZHU D. M034 ZIEGLER S. LE042 ZIHER D. M036 ZIMMERMANN A. LE053 ZIPFEL P. W001 ZITKO J. Z016 ZORC B. Z023 ZORE M. Z035 ZUBRIENE A. M017, M057 ZUERCHER W. M035 ZUKAUSKAITE A. U002

ZWERGEL C. T014 ZYGMUNT B. Z032, Z033

EFMC-ISMC | 485

Index of Participants

486 | EFMC-ISMC

ABAS PRADES Sonia UNIVERSITY OF LEEDS, LEEDS, GB ABDELSALAM Mohamed UNIVERSITY OF HALLE, HALLE, DE ABLASSER Andrea EPFL, LAUSANNE, CH ÁBRÁNYI-BALOGH Peter RCNS, BUDAPEST, HU AGBABA Danica UNIVERSITY OF BELGRADE, BELGRADE, RS AHMAD ASLAM Nayyar KU LEUVEN, LEUVEN, BE AKAHOSHI Issei KAKEN PHARMACEUTICAL, KYOTO, JP AKDAG Mevlüt UNIVERSITY OF GAZI, ANKARA, TR AKI-YALCIN Esin ANKARA UNIVERSITY, ANKARA, TR ALAMOUDI Mohsen NEWCASTLE UNIVERSITY, NEWCASTLE, GB ALCOLEA Veronica UNIVERSITY OF NAVARRA, PAMPLONA, ES ALEN AMARO Claudia INSTRUCT-ERIC, OXFORD, GB ALI Amjad MERCK & CO, KENILWORTH, US ALNAJJAR Radwn UCT, CAPE TOWN, ZA ALSHABANI Lama CARDIFF UNIVERSITY, CARDIFF, GB ALSULAIMANY Marwa UNIVERSITY OF CARDIFF, CARDIFF, GB ALTMANN Karl-Heinz ETH ZÜRICH, ZÜRICH, CH ALZA Esther ICIQ, TARRAGONA, ES AMADO Patrícia UNIVERSITY OF ALGARVE, OLHÃO, PT ANDERLUH Marko UNIVERSITY OF LJUBLJANA, LJUBLJANA, SI ANDERSON Niall GSK, HERTFORDSHIRE, GB ANDRADE Carolina H. UFG, GOIÂNIA, BR ANDRES Jose Ignacio JOSE I. ANDRES, MADRID, ES ANTONIJEVIC Mirjana UNIVERSITY OF CAEN, CAEN, FR ARNAUD Bertrand ASTRAZENECA, MÖLNDAL, SE ARSENIYADIS Stellios QUEEN MARY UNIVERSITY OF LONDON, LONDON, GB ASTARLOA Iratxe FAES FARMA, LEIOA, ES ASTOLFI Andrea UNIVERSITY OF PERUGIA, PERUGIA, IT ATZ Kenneth ETH ZÜRICH, ZÜRICH, CH AUBERSON Yves P. NOVARTIS, BASEL, CH AUGUSTYNS Koen UANTWERP, ANTWERP, BE AVCI Ahmet HACETTEPE UNIVERSITY, ANKARA, TR AYE Yimon EPFL, LAUSANNE, CH BACCI Andrea UNIVERSITY OF PISA, PISA, IT BAKER Renate COLLABORATIVE DRUG DISCOVERY, CAMBRIDGE, GB BAQI Younis SULTAN QABOOS UNIVERSITY, MUSCAT, OM BARAN Phil SCRIPPS RESEARCH INSTITUTE, LA JOLLA, US BARCHERINI Valentina IMED ULISBONA, LISBON, PT BARINGHAUS Karl-Heinz SANOFI, FRANKFURT, DE BARNIOL-XICOTA Marta KU LEUVEN, LEUVEN, BE BARRECA Marilia UNIVERSITY OF PALERMO, PALERMO, IT BARTELS Bjoern F. HOFFMANN-LA ROCHE, BASEL, CH BARTOS Piia UNIVERSITY OF EASTERN FINLAND, KUOPIO, FI BASU Sujay EUROFINS ADVINUS, BANGALORE, IN BAVO Francesco UNIVERSITY OF COPENHAGEN, COPENHAGEN, DK BEC Anja UNIVERSITY OF ZAGREB, ZAGREB, HR BECHTLER Clément UNIVERSITY OF BASEL, BASEL, CH BEERLI René NOVARTIS, BASEL, CH BELUBBI Anirudh SUN PHARMA, VADODARA, IN BENDER Andreas UNIVERSITY OF CAMBRIDGE, CAMBRIDGE, GB BENEK Ondrej UNIVERSITY OF HRADEC KRALOVE, HRADEC KRALOVE, CZ BENNETT Jonathan MERCK, SHARP & DOHME, BOSTON, US BENNETT Jonathan MSD, BOSTON, US BERGANT LOBODA Kaja NATIONAL INSTITUTE OF CHEMISTRY SLOVENIA, LJUBLJANA, SI BERISHVILI Vladimir CHEMDIV, SAN DIEGO, CA, US BERMEO Rafael UNIVERSITY OF GRENOBLE ALPES, GRENOBLE, FR BERNARDES Gonçalo UNIVERSITY OF CAMBRIDGE, CAMBRIDGE, GB BERNIER David BAYER, LYON, FR BETSCHART Claudia NOVARTIS, BASEL, CH BETTS Jonathan CCDC, GB BEUS Maja INSTITUTE FOR MEDICAL RESEARCH AND OCCUPATIONAL HEALTH, ZAGREB, HR BEZENCON Olivier IDORSIA PHARMACEUTICALS, ALLSCHWIL, CH BILENSOY Erem HACETTEPE UNIVERSITY, ANKARA, TR BILLAMBOZ Muriel JUNIA, LILLE, FR BINCH Hayley F. HOFFMANN-LA ROCHE, BASEL, CH BIT Rino GSK, STEVENAGE, GB BLANKENSTEIN Jorg SANOFI, VITRY SUR SEINE, FR

BLICHARZ Klaudia JAGIELLONIAN UNIVERSITY, KRAKÓW, PL

EFMC-ISMC | 487

BLOM Petra ONCODESIGN, DIJON CEDEX, FR BOCEK Ida UNIVERSITY OF ZAGREB, ZAGREB, HR BOLJE Aljosa UNIVERSITY OF LJUBJANA, LJUBLJANA, SI BOMIO Claudio NOVARTIS, BASEL, CH BONASERA Thomas GSK, LONDON, GB BONNATERRE Florence GALAPAGOS, ROMAINVILLE, FR BONNET Maurinne ICN, NICE, FR BONNET Dominique UNIVERSITY OF STRASBOURG, ILLKIRCH, FR BONVIN Etienne UNIVERSITY OF BERN, FREIBURG, CH BORSARI Chiara UNIVERSITY OF BASEL, BASEL, CH BOSC Damien UNIVERSITY OF LILLE, LILLE, FR BOSCHI Donatella UNIVERSITY OF TURIN, TORINO, IT BOSCO Michael UNIVERSITY OF PARIS, PARIS, FR BOSS Christoph IDORSIA PHARMACEUTICALS, ALLSCHWIL, CH BOUCHÉ Léa F. HOFFMANN-LA ROCHE, BASEL, CH BOZZOLA Tiago LUND UNIVERSITY, LUND, SE BRAMBILLA Marta JANSSEN R&d, BEERSE, BE BRANDAO Pedro UNIVERSITY OF COIMBRA, COIMBRA, PT BRANDSTÄTTER Marco HOFFMANN LA ROCHE, BASEL, CH BRAUN CORNEJO Maria SPECS, ZOETERMEER, NL BRENNAN Paul UNIVERSITY OF OXFORD, OXFORD, GB BRINER Karin NOVARTIS, CAMBRIDGE, US BRÖNSTRUP Mark HELMHOLTZ CENTRE FOR INFECTION RESEARCH, BRAUNSCHWEIG, DE BROTSCHI Christine IDORSIA, ALLSCHWIL, CH BROUGH Steve KEY ORGANICS, CORNWALL, GB BRUSS Hanna ASTRAZENECA, MÖLNDAL, SE BULLER Rebecca ZHAW, WÄDENSWIL, CH BUSCHMANN Nicole NOVARTIS, BASEL, CH CAI Xingguang UNIVERSITY OF BERN, BERN, CH CALLIS Timothy UNIVERSITY OF SYDNEY, SYDNEY, AU CAMPANICO André UNIVERSITY OF LISBON, LISBOA, PT CAMPBELL Emma UNIVERSITY OF STRATHCLYDE, GLASGOW, GB CANABAL FALCON Rafael UNIVERSITY OF SANTIAGO DE COMPOSTELA, SANTIAGO DE COMPOSTELA, ES CAO Jun UNAMUR, NAMUR, BE CARAMENTI Paola JANSSEN, BEERSE, BE CARDONA Francisco BIAL, CORONADO (S. ROMÃO E S. MAMEDE), PT CARREIRA Monica GALCHIMIA, TOURO, ES CARREIRA Erick M. ETH ZÜRICH, ZÜRICH, CH CARREL Aline UNIVERSITY OF BERN, BERN, CH CARVALHAL Francisca UNIVERSITY OF PORTO, VILA NOVA DE FAMALICÃO, PT CASTALDI Paola LIFEMINE THERAPEUTICS, CAMBRIDGE, US CECCHINI Carlotta UNIVERSITY OF GENEVA, GENEVA, CH CECERE Giuseppe F. HOFFMANN-LA ROCHE, BASEL, CH CERADINI Davide LATVIAN INSTITUTE OF ORGANIC SYNTHESIS, RIGA, LV CEULEMANS Hugo JANSSEN, BRUSSELS, BE CHALUPNIK Paulina JAGIELLONIAN UNIVERSITY, CRACOW, PL CHARPENTIER Julie ROCHE, BASEL, CH CHARTIER Léa UNAMUR, NAMUR, BE CHINO Ayaka ASTELLAS PHARMA INC, IBARAKI, JP CHRIENOVA Zofia UNIVERSITY OF HRADEC KRALOVE, HRADEC KRALOVE, CZ CHRZANOWSKI Jacek ONCOARENDI THERAPEUTICS, WARSAW, PL CHUANG Grace CYTOKINETICS, SOUTH SAN FRANCISCO, US CIBA Marija UNIVERSITY OF COPENHAGEN, COPENHAGEN, DK CIESIOLKIEWICZ Agnieszka WROCLAW UNIVERSITY, WROCLAW, PL CLEMENT Bernd UNIVERSITY OF KIEL, KIEL, DE CLOZEL Jean-Paul IDORSIA PHARMACEUTICALS, ALLSCHWIL, CH CODERCH BOUÉ Claire USP-CEU, BOADILLA DEL MONTE, ES CODONY Sandra UNIVERSITY OF BARCELONA, BARCELONA, ES COIMBRA Judite UNIVERSITY OF COIMBRA, COIMBRA, PT COLBON Paul LIVERPOOL CHIROCHEM, LIVERPOOL, GB COLCHÓN-PIERNA Esther USC, SANTIAGO DE COMPOSTELA, ES COMEO Eleonora UNIVERSITY OF NOTTINGHAM/MONASH UNIVERSITY, NOTTINGHAM, GB CONTI Gabriele UNIVERSITY OF BASEL, BASEL, CH CORIO Alessandra UNIVERSITY OF PARIS, PARIS, FR COSTA Marta UNIVERSITY OF MINHO, BRAGA, PT COTESTA Simona NOVARTIS, BASEL, CH COX Rhona ASTRAZENECA, MÖLNDAL, SE CRAMER Jonathan UNIVERSITY OF BASEL, BASEL, CH CRNCEVIC Doris UNIVERSITY OF SPLIT, SPLIT, HR CRONIN Leroy UNIVERSITY OF GLASGOW, GLASGOW, GB

488 | EFMC-ISMC

CUMMING John F. HOFFMANN-LA ROCHE, BASEL, CH CUNHA Eva UNIVERSITY OF OSLO, OSLO, NO CUNNINGHAM Fraser UNIVERSITY OF DUNDEE, DUNDEE, GB CZECHTIZKY Werngard ASTRAZENECA, MÖLNDAL, SE DACK Kevin LEO PHARMA, BALLERUP, DK DANTI Giorgia UNIVERSITY OF WURZBURG, WÜRZBURG, DE DARMENCY Vincent AC IMMUNE, LAUSANNE, CH DAVIS Holly NOVATRIS, BASEL, CH DAWSON Mike MIKE DAWSON ANTIMICROBIAL RESEARCH CONSULTANCY, STEVENAGE, GB DE ESCH Iwan VU AMSTERDAM, AMSTERDAM, NL DE GRAAF Chris SOSEI HEPTARES, CAMBRIDGE, GB DE JUAN ALBERDI Mario UNIVERSITY OF SALAMANCA, SALAMANCA, ES DE LA VEGA Jennifer UNIVERSITY OF SALAMANCA, SALAMANCA, ES DE PASCUAL-TERESA Beatriz CEU SAN PABLO UNIVERSITY, MADRID, ES DE VILLEROCHÉ Anne NOVALIX, ILLKIRCH, FR DEKKER Tom UNIVERSITY OF AMSTERDAM, AMSTERDAM, NL DEMURO Stefania UNIVERSITY OF BOLOGNA, BOLOGNA, IT DEPREZ-POULAIN Rebecca UNIVERSITY OF LILLE / INSTITUT PASTEUR DE LILLE, LILLE, FR DERDAU Volker SANOFI, FRANKFURT, DE DERNOVSEK Jaka UNIVERSITY OF LJUBLJANA, LJUBLJANA, SI DESIATKINA Oksana UNIVERSITY OF BERN, BERN, CH DESTRO Lorenza UNIVERSITY OF MODENA, MODENA, IT DETERING Carsten SCHRÖDINGER, DE DEY Fabian F. HOFFMANN-LA ROCHE, BASEL, CH DEY Surjendu BIOTECH, MAINZ, DE DI GERONIMO Bruno CNIO, MADRID, ES DI SANTO Roberto UNIVERSITY OF ROME LA SAPIENZA, ROME, IT DIALER Clemens GRUNENTHAL, AACHEN, DE DIFFERDING Edmond DIFFERDING CONSULTING, LOUVAIN-LA-NEUVE, BE DIGHE Satish Natha QUESNSLAND UNIVERSITY OF TECHNOLOGY, BRISBANE, AU DILLON Michael IDEAYA BIOSCIENCES, SOUTH SAN FRANCISCO, US DING Sheng UCSF, SAN FRANCISCO, US DING Ke JINAN UNIVERSITY, GUANGZHOU, CN DINH Chau Phi UNIVERSITY OF LILLE, LILLE, FR DISTLER Joerg UNIVERSITY OF ERLANGEN-NUREMBERG, ERLANGEN, DE DODD Peter UNIVERSITY OF DUNDEE, DUNDEE, GB DOLLINGER Horst PRIVATE, SCHEMMERHOFEN, DE DONG HO Lee SK BIOPHARMACEUTICALS, SEONGNAM-SI, KR DONGIK Kwak DONGGUK UNIVERSITY, GOYANGSI, KR DORMAN György UNIVERSITY OF SZEGED, SZEGED, HU DRENICHEV Mikhail EIMB RAS, MOSCOW, RU DREWEK Isalyne UMONS, MONS, BE DRUMMOND Michael CHEMICAL COMPUTING GROUP, MONTREAL, CA DU Zhimin GILEAD SCIENCES, BELMONT, US DUBURS Gunars LATVIAN INSTITUTE OF ORGANIC SYNTHESIS, RIGA, LV DUCA Margherita UNIVERSTY OF UTRECHT, UTRECHT, NL DUCA Maria UNIVERSITY OF COTE D'AZUR, NICE, FR DUDUTIENE Virginija VILNIUS UNIVERSITY, VILNIUS, LT DUNKEL Petra UNIVERSITY OF SEMMELWEIS, BUDAPEST, HU DURAES Fernando UNIVERSITY OF PORTO, BARCELOS, PT EBRU DIDEM Cosar BEZMIALEM VAKIF UNIVERSITY, ISTANBUL, TR ECKER Gerhard UNIVERSITY OF VIENNA, VIENNA, AT EHMKE Veronika NOVARTIS, BASEL, CH EHRLICH Stephan SCHRÖDINGER, MUNICH, DE EL BKASSINY Sandy GALAPAGOS, MECHELEN, BE EL PHIL Radhia UNIGE, GENEVA, CH ENGKVIST Ola ASTRAZENECA, MÖLNDAL, SE ERDMANN Alexandre EVOTEC, TOULOUSE, FR ERONEN Aleksi UNIVERSITY OF HELSINKI, HELSINKI, FI ERRA SOLA Montse ALMIRALL, BARCELONA, ES ESCALA Nerea UNIVERSITY OF SALAMANCA, SALAMANCA, ES ESCOBAR MONTAÑO Felipe UNIVERSITY OF CADIZ, PUERTO REAL (CÁDIZ), ES ESPOSITO SALSANO Jasmine UNIVERSITY OF PISA, PISA, IT EZZANAD Abdellah UNIVERSITY OF CADIZ, CADIZ, ES FADEYI Niyi MERCK & Co, CAMBRIDGE, US FAHS Sara BIONTECH, MAINZ, DE FAJKIS-ZAJACZKOWSKA Nikola UNIVERSITY OF JAGIELLONIAN, CRACOW, PL FANG Emily ONCODESIGN, DIJON, FR FEDOROWICZ Joanna MEDICAL UNIVERSITY OF GDANSK, GDANSK, PL FÉLIX Rita ULISBOA, LISBOA, PT

EFMC-ISMC | 489

FERRARA Isabella UNIVERSITY OF ZURICH, ZURICH, CH FERNANDEZ-LLAMAZARES Ana Iris CLARIVATE, BARCELONA, ES FEZZARDI Paola IRBM, ROMA, IT FIDALGO Lara UNIVERSITY OF LISBON, LISBOA, PT FISCHER Janos EMERITUS MEMBER OF IUPAC, BUDAPEST, HU FISCHER Christian MERCK, SHARP & DOHME, BOSTON, US FISHER Martin NANNA THERAPEUTICS, CAMBRIDGE, GB FLASSHOFF Maren TU BRAUNSCHWEIG, BRAUNSCHWEIG, DE FLOHR Stefanie NOVARTIS, BASEL, CH FLORINDO Pedro UNIVERSITY OF LISBON, LISBON, PT FOITZIK Richard MYCRX, MELBOURNE, AU FORICHER Yann SANOFI, VITRY-SUR-SEINE, FR FOURNIER Jean-Francois GALAPAGOS, ROMAINVILLE, FR FRYCIA Anne UCB, BRAINE L'ALLEUD, BE FUCHSS Thomas MERCK KGaA, DARMSTADT, DE FUJIWARA Hideto OTSUKA PHARMACEUTICAL, TOKUSHIMA, JP FÜSSER Friederike WILHELMS-UNIVERSITY MÜNSTER, MÜNSTER, DE GABELLIERI Emanuele AC IMMUNE, LAUSANNE, CH GANDINI Annachiara NANNA THERAPEUTICS, CAMBRIDGE, GB GAO Xuri ENANTA PHARM, WATERTOWN, US GARCÍA Ana BelÉn SPANISH NATIONAL CANCER RESEARCH CENTER, MADRID, ES GARCIA JIMENEZ Diego UNIVERSITY OF TURN, TORINO, IT GARCÍA MARTÍN Aimar FAES FARMA, BIZKAIA, ES GARDELLI Cristina ASTRAZENECA, MÖLNDAL, SE GARDNER Chris WEHI, BUNDOORA, AU GARLATTI Laura AFMB, MARSEILLE, FR GASTREICH Marcus BIOSOLVEIT, ST. AUGUSTIN, DE GAUL Christoph NOVARTIS, BASEL, CH GAVATHIOTIS Evripidis ALBERT EINSTEIN COLLEGE OF MEDICINE, NEW YORK, US GAWALSKA Alicja UNIVERSITY OF JAGIELLONIAN, KRAKÓW, PL GEHRINGER Matthias UNIVERSITY OF TÜBINGEN, TUEBINGEN, DE GEHRTZ Paul MERCK KGAA, DARMSTADT, DE GENSKI Thorsten TAROS CHEMICALS, DORTMUND, DE GEORGIOU Irene UNIVERSIRY OF DUNDEE, DUNDEE, GB GHERBOVET Olga EVOTEC, TOULOUSE, FR GIACCHE Nicola TES PHARMA, PERUGIA, IT GIANQUINTO Eleonora UNIVERSITY OF TORINO, TORINO, IT GILBERT Kate GSK, STEVENAGE, GB GIRARDI Benedetta UNIVERSITY OF BASEL, BASEL, CH GIROUD Maude F. HOFFMANN-LA ROCHE, BASEL, CH GLYNN Daniel MALVERN PANALYTICAL, WORCESTERSHIRE MALVERN, GB GOBBI Luca F. HOFFMANN-LA ROCHE, BASEL, CH GOFFIN Eline KU LEUVEN, LEUVEN, BE GOMEZ Sylvie EVOTEC, TOULOUSE, FR GOMEZ Elena ALMIRALL, BARCELONA, ES GONCHARENKO Kristina SPIROCHEM, BASEL, CH GONDELA Andrzej RYVY THERAPEUTICS, KRAKOW, PL GONZALEZ Simon CERGY PARIS UNIVERSITY, CERGY, FR GONZALEZ CANTALAPIEDRA Esther SPANISH NATIONAL CANCER RESEARCH CENTER, MADRID, ES GOODWIN Nicole GSK, KING OF PRUSSIA, US GOSMINI Romain GALAPAGOS, ROMAINVILLE, FR GOUDE Marie-Line NOVARTIS, BASEL, CH GRAVEN SAMS Anette H. LUNDBECK, VALBY, DK GRAVIER-PELLETIER Christine UNIVERSITY PARIS DESCARTES, PARIS CEDEX 06, FR GROTZFELD Robert NOVARTIS, BASEL, CH GRUNDY Ian ARAGEN LIFE SCIENCES, GB GUARDIOLA Salvador ONA THERAPEUTICS, BARCELONA, ES GUBIC Spela UNIVERSITY OF LJUBLJANA, LJUBLJANA, SI GUERET Stephanie ASTRAZENECA, MÖLNDAL, SE GUIBOURDENCHE Christel NOVARTIS, BASEL, CH GUZELJ Samo UNIVERSITY OF LJUBJANA, LJUBLJANA, SI HAIMOV Elvira TEL AVIV UNIVERSITY, TEL AVIV, IL HALECKOVA Annamaria UNIVERSITY OF HRADEC KRALOVE, HRADEC KRALOVE, CZ HALLAND Nis SANOFI, FRANKFURT AM MAIN, DE HAN Wooseok CYRUS THERAPEUTICS, SONGPA-GU, KR HANCOCK Ashley GSK, STEVENAGE, GB HANSEN Finn UNIVERSITY OF BONN, BONN, DE HAPKO Uladzislau HIPS, SAARBÜCKEN, DE HARDICK David STORM THERAPEUTICS, CAMBRIDGE, GB HARGROVE Amanda DUKE UNIVERSITY, DURHAM, US

490 | EFMC-ISMC

HARKI Daniel UNIVERSITY OF MINNESOTA, MINNEAPOLIS, US HARTUNG Ingo MERCK HEALTHCARE KGAA, DARMSTADT, DE HASLETT Greg UCB, SLOUGH, GB HASSAN Mujtaba LUND UNIVERSITY, LUND, SE HAY Duncan VERTEX, ABINGDON, GB HEINELT Uwe SANOFI, FRANKFURT, DE HEMMERLE Horst NOVARTIS INSTITUTES FOR BIOMEDICAL RESEARCH, CAMBRIDGE, US HENDERSON Timothy J. Merck & Co, BOSTON, US HENRY Andrew CHEMICAL COMPUTING GROUP, CAMBRIDGE, GB HERBERT Simon BAYER, DE HERLAH Barbara NATIONAL INSTITUTE OF CHEMISTRY, LJUBLJANA, SI HERMANN Gary BAYER, WUPPERTAL, DE HERR Jasmin GDCh, FRANKFURT A.M., DE HESSLER Gerhard SANOFI, FRANKFURT AM MAIN, DE HEVEY Rachel UNIVERSITY BASEL, BASEL, CH HEWINGS David VERTEX PHARMACEUTICALS, ABINGDON, GB HIDAKA Kousuke TAKEDA, FUJISAWA, KANAGAWA, JP HIESINGER Kerstin GOETHE UNIVERSITY, FRANKFURT/MAIN, DE HILVERT Donald ETH ZÜRICH, ZURICH, CH HINRICHS Jürgen NOVARTIS, BASEL, CH HINTERMANN Samuel NOVARTIS, BASEL, CH HIRATA Yoshiyuki OSAKA UNIVERSITY, OSAKA, JP HIRSCH Anna K. H. HELMHOLTZ INSTITUTE HIPS, SAARBRÜCKEN, DE HOEGENAUER Klemens NOVARTIS, BASEL, CH HOFFMANN Remy JUBILANT BIOSYS, NOIDA, IN HOLLINGWORTH Gregory NOVARTIS, BASEL, CH HOLVEY Rhian ASTEX PHARMACEUTICALS, CAMBRIDGE, GB HONDA Takeshi DAIICHISANKYO, TOKYO, JP HOTINGER Julia VIRGINIA COMMONWEALTH, HENRICO, US HSIAO Meng-Yang JANSSEN, BEERSE, BE HUCK Jacques GALAPAGOS, ROMAINVILLE, FR HUTCHINGS Rosie WUXI APPTEC, LONDON, GB HUTTER Roman HOFFMANN LA ROCHE, BASEL, CH HUTTUNEN Kristiina UNIVERSITY OF EASTERN FINLAND, KUOPIO, FI IALONGO Davide UNIVERSITY OF ROME, ROME, IT IHNATENKO Irina TU BRAUNSCHWEIG, BRAUNSCHWEIG, DE IJZERMAN Ad P. LEIDEN UNIVERSITY, LEIDEN, NL IKEDA Zenichi TAKEDA, KANAGAWA, JP IMBERTY Anne CNRS/CERMAV, GRENOBLE, FR INTROVIGNE Maria Luisa UNIVERSITY OF MODENA, MODENA, IT IRALDE-LORENTE Leire UNIVERSITY OF SIENA, SIENA, IT IVANOVA Jekaterina LATVIAN INSTITUTE OF ORGANIC SYNTHESIS, RIGA, LV JACQUIER Sebastien NOVARTIS, BASEL, CH JAESCHKE Georg F. HOFFMANN-LA ROCHE, BASEL, CH JAMROZIK Marek UNIVERSITY OF JAGIELLONIAN, CRACOW, PL JANKOWSKA Elzbieta UNIVERSITY OF GDANSK, GDANSK, PL JANSSEN Freek PIVOT PARK SCREENING CENTRE, NIJMEGEN, NL JENSEN Klavs F. MIT, CAMBRIDGE, US JERHAOUI Soufyan JANSSEN, BEERSE, BE JIYU Woo DONGGUK UNIVERSITY, GYEONGGI-DO, KR JOLY Emilie NOVARTIS, RIXHEIM, FR JONCKERS Tim JANSSEN, BEERSE, BE JONCOUR Agnes GALAPAGOS, ROMAINVILLE, FR JONES Lee FLUOROCHEM, HADFIELD, GB JOSA CULLERE Laia IQAC-CSIC, BARCELONA, ES JOSE Jiney UNIVERSITY OF AUCKLAND, AUCKLAND, NZ JUDD Andrew ABBVIE, GRAYSLAKE, US JUHÁS Martin CHARLES UNIVERSITY, HRADEC KRÁLOVÉ, CZ JUHL Karsten LUNDBECK, VALBY, DK JUILLET Charlotte CNRS, GIF SUR YVETTE, FR JUMDE Ravindra P. HELMHOLTZ INSTITUTE FOR PHARMACEUTICAL RESEARCH SAARLAND, SAARBRÜCKEN, DE JUMPPANEN Mikael UNIVERSITY OF HELSINKI, HELSINKI, FI JUN MA Jun ENANTA, WATERTOWN, US JUNG Erik UNIVERSITY OF ZURICH, DIETLIKON, CH JUVENTHALA Rama Krishna ARAGEN LIFE SCIENCES, IN KAPADNIS Prashant CHARLES RIVER, HARLOW, GB KARLÉN Anders UPPSALA UNIVERSITY, UPPSALA, SE KEKEZOVIC Sladjana UNIVERSITY OF NOVI SAD, NOVI SAD, RS KELLEY Tanya UNIVERSITY OF MIAMI, MIAMI, US KENNELLY Samantha UNIVERSITY OF MINNESOTA, MINNEAPOLIS, US

EFMC-ISMC | 491

KESERU Gyorgy HUNGARIAN ACADEMY OF SCIENCES, BUDAPEST, HU KEYAERTS Jean UCB, BRAINE L'ALLEUD, BE KEZAMPOUR-DIZAJI Mohamad TEHRAN UNIVERSITY OF MEDICAL SCIENCES, TEHRAN, IR KHAN Ashiq SCHRÖDINGER, MUNICH, DE KHAN Tuhina INSTITUT PASTEUR DE LILLE, LILLE, FR KHANNA Avinash HANSOH BIO, ROCKVILLE, US KIEC-KONONOWICZ Katarzyna JAGIELLONIAN UNIVERSITY, KRAKOW, PL KILAJ Ardita NOVARTIS, BASEL, CH KILBURN Paul LUNDBECK, VALBY, DK KILONDA Amuri CISTIM, LEUVEN, BE KIURU Paula UNIVERSITY OF HELSINKI, HELSINKI, FI KLEIN Markus MERCK, DARMSTADT, DE KLEY Jörg BOEHRINGER INGELHEIM PHARMA, BIBERACH AN DER RISS, DE KLINGLER Franca MSD, LONDON, GB KLUG Dana UCL, LONDON, GB KNAPP Stefan GOETHE UNIVERSITY FRANKFURT, FRANKFURT AM MAIN, DE KNOEPFEL Thomas NOVARTIS, BASEL, CH KOBAURI Piermichele UNIVERSITY OF GRONINGEN, GRONINGEN, NL KONETZKI Ingo SKYHAWK THERAPEUTICS, BASEL, CH KOSSNER Markus CHEMICAL COMPUTING GROUP, KÖLN, DE KOSTRUN Sanja FIDELTA, ZAGREB, HR KOTSCHY Andras SERVIER, BUDAPEST, HU KOUROUNAKIS Angeliki NATIONAL AND KAPODISTRIAN UNIVERSITY OF ATHENS, ATHENS, GR KOVACHKA Sandra UNIVERSITY OF COTE D'AZUR, NICE, FR KRAJNC Alen UNIVERSITY OF OXFORD, OXFORD, GB KRAMER Christian F. HOFFMANN-LA ROCHE, BASEL, CH KUDOVA Eva CZECH ACADEMY OF SCIENCES, PRAGUE 6, CZ KUHN Bernd F. HOFFMANN-LA-ROCHE, BASEL, CH KUTTRUFF Christian BOEHRINGER INGELHEIM, BIBERACH, DE LACIVITA Enza UNIVERSITY OF BARI, BARI, IT LADOSZ Agnieszka IDORSIA, ALLSCHWIL, CH LAINO Teodoro IBM RESEARCH EUROPE, RUESCHLIKON, CH LAM Philip NIBR, CAMBRIDGE, US LAMBERT Heather FLUOROCHEM, HADFIELD, GB LAMERS Christina UNIVERSITY OF BASEL, BASEL, CH LAMY Cedric CHARLES RIVER, SAFFRON WALDEN, GB LANGLOIS Jean-Baptiste NOVARTIS, BASEL, CH LANMAN Brian AMGEN, THOUSAND OAKS, CA, US LANTHIER Caroline UNIVERSITY OF ANTWERPEN, ANTWERP, BE LAPOINTE Guillaume NOVARTIS, BASEL, CH LAUFER Stefan UNIVERSITY OF TÜBINGEN, TÜBINGEN, DE LAUL Eleen UNIVERSITY OF ZURICH, ZURICH, CH LAZARIDES Linos CHARNWOOD MOLECULAR, LOUGHBOROUGH, GB LE CORRE Laurent UNIVERSITY OF PARIS, PARIS, FR LEBOHO Tlabo UNIVERSITY OF LIMPOPO, SOVENGA, POLOKWANE, ZA LEE Maximillian MSD, LONDON, GB LEE Kyeong DONGGUK UNIVERSITY, GOYANG, KR LEFRANC Julien MERCK, DARMSTADT, DE EHR Philipp NOVARTIS, BASEL, CH LEMURELL Malin ASTRAZENECA, GOTHENBURG, SE LENCE Emilio UNIVERSITY OF SANTIAGO DE COMPOSTELA, SANTIAGO DE COMPOSTELA, ES LENDERS Stijn KU LEUVEN, LEUVEN, BE LESIRE Laetitia INSTITUT PASTEUR DE LILLE, LILLE, FR LEVANTO Stefano CHARLES RIVER, SAFFRON WALDEN, GB LEVET Gaspard UNIVERSITY OF ROUEN, MONT SAINT AIGNAN, FR LEYPOLD Mario ASTRAZENECA, MÖLNDAL, SE LIARGKOVA Thalia ARISTOTLE UNIVERSITY OF THESSALONIKI, THESSALONIKI, GR LINDHOF Jens TU BRAUNSCHWEIG, BRAUNSCHWEIG, DE LINDSTROM Johan SPRINT BIOSCIENCE, HUDDINGE, SE LISCIO Paride TES PHARMA, PERUGIA, IT LIU Zhiguo JOHNSON & JOHNSON CHINA, SHANGHAI, CN LIU Longbin CHDI FOUNDATION, LOS ANGELES, US LIU Haixia F. HOFFMANN-LA ROCHE, SHANGHAI, CN LIZANDRA PEREZ Juan POLITECHNIKA WROCLAWSKA, WROCLAW, PL LOI Elena UNIVERSITY OF LJUBLJANA, LJUBLJANA, SI LOLLI Marco UNIVERSITY OF TURIN, TORINO, IT LONDON Nir THE WEIZMANN INSTITUTE OF SCIENCE, REHOVOT, IL LOPES Elizabeth A. UNIVERSITY OF LISBON, LISBOA, PT LOPEZ RAMOS Miriam GALAPAGOS, ROMAINVILLE, FR LOPEZ ROJAS Priscila UNIVERSITY OF LA LAGUNA, SAN CRISTÓBAL DE LA LAGUNA, ES

492 | EFMC-ISMC

LOUKAS Ieremias UNIVERSITY OF COPENHAGEN, COPENHAGEN, DK LOWERSON Andrew KEY ORGANICS, CORNWALL, GB LUCAS Xavier HOFFMANN-LA ROCHE AG, BASEL, CH LUQUE NAVARRO Pilar María UNIVERSITY OF PERUGIA, PERUGIA, IT LUZ André UNIVERSITY OF LISBON, LAGOS, PT LYOTHIER Isabelle IDORSIA PHARMACEUTICALS, ALLSCHWIL, CH MA Zhihua KRONOS BIO, CAMBRIDGE, US MACFARLANE Katherine UNIVERSITY OF STRATHCLYDE, LONDON, GB MACHACEK Michelle MERCK, SHARP & DOHME, BOSTON, US MACHULKIN Aleksei LOMONOSOV MOSCOW STATE UNIVERSITY, MOSCOW, RU MACK Jürgen BOEHRINGER INGELHEIM, BIBERACH AN DER RISS, DE MACKAY Angela NOVARTIS, BASEL, CH MACKENZIE Claire UNIVERSITY OF DUNDEE, DUNDEE, GB MACOR John E. SANOFI, WALTHAM, US MAGALHAES E SILVA Diogo UNIVERSITY OF LISBON, LISBON, PT MALARZ Katarzyna UNIVERSITY OF SILESIA, KATOWICE, PL MALEBARI Azizah KING ABDULAZIZ UNIVERSITY, JEDDAH, SA MALINAUSKIENE Vida KAUNAS UNIVERSITY, KAUNAS, LT MALISZEWSKI Dawid MEDICAL UNIVERSITY OF BIALYSTOK, BIA&#322;YSTOK, PL MALLART Sergio SANOFI, CHILLY MAZARIN, FR MAMIDI Srinivas Reddy ARAGEN LIFE SCIENCES, AMSTERDAM SCHIPHOL, NL MAMMOLITI Oscar GALAPAGOS, ROMAINVILLE, FR MANEIRO María UNIVERSITY OF SANTIAGO DE COMPOSTELA, SANTIAGO DE COMPOSTELA, ES MANNISTO Jere Kristian UNIVERSITY OF HELSINKI, HELSINKI, FI MANOHARAN Muthiah ALNYLAM PHARMACEUTICALS, CAMBRIDGE, US MAPLE Hannah BIO-TECHNE, BRISTOL, GB MARAZZI Marco UNIVERSITY OF ALCALA, ALCALÁ DE HENARES, ES MARCHINI Mattia ITALFARMACO, CINISELLO BALSAMO, IT MARCINIAK Monika UNIVERSITY OF WARSAW, WARSAW, PL MARI Michele UNIVERSITY OF URBINO, URBINO, IT MARIAM Ezekiel CCDC, CAMBRIDGE, GB MARIC Hans UNIVERSITY WUERZBURG, WÜRZBURG, DE MARTIN Barrie CXA DISCOVERY, MANCHESTER, GB MARTIN Fernando PHARMACELERA, BARCELONA, ES MARTIN Rainer F. HOFFMANN-LA ROCHE, BASEL, CH MARTIN Laetitia F. HOFFMANN-LA ROCHE, BASEL, CH MARTINEZ ALARCON Dania UNIVERSITY OF GRENOBLE, GIERES, FR MARTINEZ VITURRO Carlos Manuel JANSSEN, TOLEDO, ES MARTINO Elena UNIVERSITY OF TORINO, TORINO, IT MARTIN-SANTAMARIA Sonsoles CIB-CSIC, MADRID, ES MARUYAMA Akinobu TEJIN PHARMA, TOKYO, JP MASSE Julie GALCHIMIA, O PINO A CORUNA, ES MATCHA Kiran JANSSEN, BEERSE, BE MATTEI Patrizio F. HOFFMANN-LA ROCHE, BASEL, CH MAZUNIN Dmitry F. HOFFMANN-LA ROCHE, BASEL, CH MCCLARRON Alexander UNIVERSITY OF YORK, READING, GB MEIBOM Daniel BAYER, WUPPERTAL, DE MEMARZADEH Sarah UNIVERSITY OF GLASGOW, GLASGOW, GB MÉNDEZ PÉREZ Maria SANOFI, FRANKFURT AM MAIN, DE MERCEDES Rubio Hernández UNIVERSITY OF NAVARRA, PAMPLONA, ES MESCH Stefanie HOFFMANN-LA ROCHE, BASEL, CH MESMOUDI Youri TAROS CHEMICALS, DORTMUND, DE MESSORE Antonella UNIVERSITY OF ROME, ROMA, IT METSÄNEN Toni ORION CORPORATION, ESPOO, FI MEVELLEC Laurence NOVALIX, VAL DE REUIL, FR MEZAACHE Roufia UNIVERSITY MOSTAFA BENBOULAID, BATNA, DZ MEZOHEGYI Gergo INNOSTUDIO INC, BUDAPEST, HU MIAH Afjal GLAXOSMITHKLINE, STEVENAGE, GB MICHAEL Wright NOVARTIS, BASEL, CH MICHALEK Stanislaw CELON PHARMA, KIELPIN, PL MICHALSKA Patrycja KAROLINASKA INSTITUTET, STOCKHOLM, SE MIGUELEZ RAMOS Javier ASTELLAS PHARMA, TSUKUBA, JP MIHAJLO Krunic UNIVERSITY OF BERLGRADE, BELGRADE, RS MIHOVILOVIC Marko D. TU WIEN, VIENNA, AT MILLET Antoine GW PHARMACEUTICALS, SITTINGBOURNE, GB MIN Kyoungho DONGGUK UNIVERSITY, GOYANG, KR MIRET CASALS Laia UGENT, GENT, BE MIRONOV Maksim NOVOSIBIRSK INSTITUTE OF ORGANIC CHEMISTRY, NOVOSIBIRSK, RU MISSBACH Martin NOVARTIS, BASEL, CH MITTENDORF Joachim BAYER, WUPPERTAL, DE

EFMC-ISMC | 493

MOFFATT Frank NOVALIX, ILLKIRCH, FR MONTEIRO Ana UNIVERSITY OF AVEIRO, AVEIRO, PT MONTI Barbara UNIVERSITY OF BOLOGNA, BOLOGNA, IT MORALES Juan Carlos CSIC, ARMILLA, ES MOREIRA Rui UNIVERSITY OF LISBON, LISBON, PT MORGAN Barry UNIVERSITY OF TEXAS, HOUSTON, US MORRIS Morgan UNIVERSITY COLLEGE DUBLIN, LOUGHLINSTOWN, IE MOTIEI Leila WEIZMANN INSTITUTE, REHOVOT, IL MOUROT Laura UHA, MULHOUSE, FR MRÓZ Piotr UNIVERSITY OF WARSAW, WARSAW, PL MÜLLER Marco TU BRAUNSCHWEIG, BRAUNSCHWEIG, DE MULLIRI Kleni UNIVERSITY OF BERN, BERN, CH MUSILEK Kamil UNIVERSITY OF HRADEC KRALOVE, HRADEC KRALOVE, CZ MUTTENTHALER Markus UNIVERSITY OF VIENNA, VIENNA, AT NACHAMPASSAK Southida UBE, DUSSELDORF, DE NAGABHUSHAN Smrithi ARAGEN LIFE SCIENCES, HYDERABAD, IN NAMOTO Kenji NOVARTIS, BASEL, CH NAR Herbert BOEHRINGER INGELHEIM, BIBERACH, DE NARDUCCI Daniele NATIONAL HELLENIC RESEARCH FOUNDATION, ATHENS, GR NAZARE Marc FMP, BERLIN, DE NERI Dario ETH ZÜRICH, ZÜRICH, CH NEUBIG Megan GILEAD SCIENCES, FOSTER CITY, US NEUMANN Alexander BIOSOLVEIT, ST. AUGUSTIN, DE NEVADO Cristina UNIVERSITY OF ZÜRICH, ZURICH, CH NEWTON Gary THE INSTITUTE OF CANCER RESEARCH, LONDON, GB NGUYEN Ngoc STRATHCLYDE UNIVERSITY, GLASGOW, GB NICHOLS Paula SYNPLE CHEM, ZURICH, CH NIE Laiyin WUXI APPTEC, DE NIKOLIC Katarina UNIVERSITY OF BELGRADE, BELGRADE, RS NILSSON Magnus ASTRAZENECA, MÖLNDAL, SE NILSSON Ulf LUND UNIVERSITY, LUND, SE NOCEN Pawel WROCLAW UNIVERSITY, WROCLAW, PL NORDQVIST Anneli ASTRAZENECA, GOTHENBURG, SE NOVAK Tibor SERVIER, BUDAPEST, HU NOVOTNÁ Adéla INSTITUTE OF ORGANIC CHEMISTRY AND BIOCHEMISTRY, PRAGUE, CZ NOWICKI Krzysztof WARSAW UNIVERSITY OF TECHNOLOGY, WARSAW, PL NOZAL GARCIA Vanesa CIB - CSIC, MADRID, ES NUÉ Jonathan CSIC, MADRID, ES OBIKA Shingo JAPAN TOBACCO, OSAKA, JP OBRINGER Michel NOVALIX, ILLKIRCH, FR OBST-SANDER Ulrike F.HOFFMANN - LA ROCHE, BASEL, CH OKA Yusuke TAISHO PHARMACEUTICALS, SAITAMA, JP OKAWA Tomohiro CARDURION PHARMACEUTICALS, FUJISAWA, KANAGAWA, JP OLIVER Martin UNIVERSITY OF PARIS, PARIS, FR OMBOUMA Joanna UNIVERSITY OF HEALTH SCIENCE, LIBREVILLE, GA ONODERA Yuta KAKEN PHARMACEUTICAL, KYOTO-CITY, JP OPREA Tudor I. UNIVERSITY OF NEW MEXICO, ALBUQUERQUE, US ORAIN David NOVARTIS, BASEL, CH ORTHOLAND Jean-Yves EDELRIS, LYON, FR ORTIN REMON Irene USPCEU, MADRID, ES PACHECO Paulo FARM-ID, LISBOA, PT PAESSENS Lutz NOVALIX, ILLKIRCH CEDEX, DE PALAFERRI Leonardo UZH, ZURICH, CH PALAZZESI Ferruccio APTUIT AN EVOTEC COMPANY, VERONA, IT PALISSE Adeline GALAPAGOS, MECHELEN, BE PALOMO Valle CSIC, MADRID, ES PALYULIN Vladimir A. LOMONOSOV MOSCOW STATE UNIVERSITY, MOSCOW, RU PANNIUNGHI Sara UNIVERSITY OF GENEVA, GENÈVE, CH PANOSETTI Marc UNIVERSITY OF COTE D'AZUR, MENTON, FR PAPAGIOUVANNIS Georgios FREDERICK UNIVERSITY CYPRUS, NICOSIA, CY PAPANGELIS Athanasios UNIVERSITY OF COPENHAGEN, COPENHAGEN, DK PAPOT Sebastien SCT & UNIVERSITY OF POITIERS, POITIERS, FR PARSY Christophe ONCODESIGN, DIJON, FR PASQUALE Linciano UNIVERSITY OF PAVIA, PAVIA, IT PASTOR Miryam LILLY, MADRID, ES PATINY Angélique F. HOFFMANN-LA ROCHE, BASEL, CH PÄTSI Henri UNIVERSITY OF HELSINKI, HELSINKI, FI PAWLICZEK Martin TAKEDA, FUJISAWA-CITY, JP PEIL Sebastian GMBH, AACHEN, DE PELLICCIARI Roberto TES PHARMA, CORCIANO, IT

494 | EFMC-ISMC

PEÑALVER PUENTE Pablo CSIC Granada, GRANADA, ES PEREZ MELERO Maria Concepcion UNIVERSITY OF SALAMANCA, SALAMANCA, ES PÉREZ-PÉREZ Maria-Jesus IQM-CSIC, MADRID, ES PERI Francesco UNIVERSITY OF MILAN BICOCCA, MILANO, IT PERKOVIC Ivana UNIVERSITY OF ZAGREB, ZAGREB, HR PERLÍKOVA Pavla VSCHT, PRAGUE 6, CZ PERMANN Stephan MICHAEL POPP INSTITUT - UNIVERSITY OF INNSBRUCK, AXAMS, AT PEROKOVIC Vesna UNIVERSITY OF ZAGREB, ZAGREB, HR PERRON Quentin IKTOS, PARIS, FR PERSONNE Hippolyte UNIVERSITY OF BERN, BERN, CH PERVAIZ Mehrosh EXSCIENTIA, OXFORD, GB PETERLIN MASIC Lucija UNIVERSITY OF LJUBJANA, LJUBLJANA, SI PETERSEN Nadia UPPSALA UNIVERSITY, UPPSALA, SE PETTER Jennifer ARRAKIS THERAPEUTICS, WALTHAM, US PETTUS Liping AMGEN, THOUSAND OAKS, US PEUKERT Carsten HELMHOLTZ CENTRE FOR INFECTION RESEARCH, BRAUNSCHWEIG, DE PHILLIPS Jonathan AMGEN, NEWBURY PARK, US PICOTTI Paola ETH ZÜRICH, ZÜRICH, CH PIGUEL Sandrine UNIVERSITY PARIS-SACLAY, ORSAY, FR PINARD Emmanuel F. HOFFMANN-LA ROCHE, BASEL, CH PIPPIONE Agnese Chiara UNIVERSITY OF TURIN, TORINO, IT PIRALI Tracey UNIVERSITY OF PIEMONTE, NOVARA, IT PIRARD Bernard NOVARTIS, BASEL, CH PLOWRIGHT Alleyn T. WREN THERAPEUTICS, CAMBRIDGE, GB PODLEWSKA Sabina MAJ INSTITUTE OF PHARMACOLOGY PAS, KRAKOW, PL POHLKI Frauke ABBVIE, LUDWIGSHAFEN, DE PONTES Olivia UNIVERSITY OF MINHO, BRAGA, PT POOJA Deshmukh SAI LIFESCIENCES, HYDERABAD, IN PORTRON Benoit BIOTAGE, HENGOED, GB POUTON Colin MONASH UNIVERSITY, CLAYTON, AU PRATI Federica ANGELINI PHARMA, ROME, IT PRIERI Marion Janssen Research & Development, BEERSE, BE PRISCHICH Davia IBEC, BARCELONA, ES PROJ Matic UNIVERSITY OF LJUBLJANA, LJUBLJANA, SI PROSZENYAK Agnes SERVIER, BUDAPEST, HU PUSKAS Laszlo AVICOR, SZEGED, HU QI Xiangbing NATIONAL INSTITUTE OF BIOLOGICAL SCIENCE, CN QILI Lu DONGGUK UNIVERSITY, GOYANG-SI, KR QUANCARD Jean NOVARTIS, BASEL, CH QUEROLLE Olivier NOVALIX, VAL DE REUIL, FR QUILIANO Miguel UNIVERSITY OF APPLIED SCIENCE OF PERU, LIMA, PE RADEMACHER Christoph UNIVERSITY OF VIENNA, VIENNA, AT RAGEOT Denise ELANCO ANIMAL HEALTH, BASEL, CH RAJIC Zrinka UNIVERSITY OF ZAGREB, ZAGREB, HR RAMILO-GOMES Filipa UNIVERSITY OF LISBON, LISBON, PT RANCATI Fabio CHIESI FARMACEUTICI, PARMA, IT RANIERI Beatrice ASTRAZENECA, MÖLNDAL, SE RAUH Daniel TU DORTMUND, DORTMUND, DE RAUTIO Jarkko UNIVERSITY OF EASTERN FINLAND, KUOPIO, FI REEKS Judith ASTEX PHARMACEUTICALS, CAMBRIDGE, GB REID Lauren MEDCHEMICA, MACCLESFIELD, GB REIS Joana UNIVERSITY OF PAVIA, PAVIA, IT REMEN Lubos IDORSIA PHARMACEUTICALS, ALLSCHWIL, CH RENAUD Jean-Paul URANIA THERAPEUTICS, OSTWALD, FR REVOL-TISSOT Johan UNIVERSITY OF MARSEILLE, MARSEILLE, FR REXEN ULVEN Elisabeth UNIVERSITY OF COPENHAGEN, COPENHAGEN, DK REY Jullien NOVATRIS AG, BASEL, CH REYNOLDS Jessica UNIVERSITY OF OXFORD, OXFORD, GB RIBIC Rosana UNIVERSITY NORTH, VARAZDIN, HR RICCI Antonio F. HOFFMANN-LA-ROCHE, BASEL, CH RICCIO Alessandra UNIVERSITY OF PERUGIA, PERUGIA, IT RICHALET Florian BASILEA PHARMACEUTICA, BASEL, CH RIEL Asia Marie UNIVERSITY OF MONTANA, MISSOULA, US RIEMENS Rick VU UNIVERSITY, UTRECHT, NL RIVERA FUENTES Pablo EPFL, LAUSANNE, CH ROCHAIS UNIVERSITY OF CAEN, CAEN, FR ROCHE Didier EDELRIS, LYON, FR RODRIGUES Daniel Alencar ROYAL COLLEGE OF SURGEON, DUBLIN, IE RODRIGUEZ SARMIENTO Rosa María F. HOFFMANN-LA ROCHE, BASEL, CH RODRÍGUEZ-COSTA Ángela UNIVERSITY OF SANTIAGO, SANTIAGO DE COMPOSTELA, ES

EFMC-ISMC | 495

ROEHRIG Susanne BAYER, WUPPERTAL, DE ROLANDO Catherine NOVARTIS, BASEL, CH ROMANO Assunta MERCK, MILANO, IT ROMANOV MICHAILIDIS Fedor JANSSAN PHARMACEUTICA, BEERSE, BE ROSA Carla TECNIMEDE GROUP, TORRES VEDRAS, PT ROSÉ Geoffroy NOVATRIS AG, ESCHENTZWILLER, FR ROSS Tatjana MERCK, DARMSTADT, DE ROßMEIER Maria UNIVERSITY OF HAMBURG, HAMBURG, DE ROZAS Isabel TRINITY COLLEGE, DUBLIN, IE RUECKRICH Thomas MERCK, TAUFKIRCHEN, DE RÜEDI Georg IDORSIA, ALLSCHWIL, CH RUIZ-GÓMEZ Gloria TUD, DRESDEN, DE RUNFOLA Massimiliano UNIVERSITY OF PISA, PISA, IT RUNTZ-SCHMITT Valerie F. HOFFMANN-LA ROCHE, BASEL, CH RUSSELL Angela UNIVERSITY OF OXFORD, OXFORD, GB RUZIC Dusan FACULTY OF PHARMACY BELGRADE, BELGRADE, RS RYDZIK Anna ASTRAZENECA, MÖLNDAL, SE SAADABADI Atefeh ABO AKADEMI UNIVERSITY, TURKU, FI SAHRAOUI Suzanne Sherihan UNIVERSITY OF GENEVA, GENEVA, CH SAINAS Stefano UNIVERSITY OF TORINO, TORINO, IT SAITO Norimichi UBE INDUSTRIES, JP SAKURAI Fumie TAKEDA, FUJISAWA, JP SALO-AHEN Outi UNIVERSITY OF ABO AKADEMI, TURKU, FI SALVADOR Jorge UNIVERSITY OF COIMBRA, COIMBRA, PT SANZ Ferran UNIVERSITY POMPEU FABRA, BARCELONA, ES SARABIA VALLEJO Álvaro UNIVERSITY COMPLUTENSE OF MADRID, MADRID, ES SATZ Alex WUXI APPTEC, CH SAURABH Loharch POLITECHNICA WROCLAWSKA, WROCLAW, PL SAUTIER Brice EVOTEC, TOULOUSE, FR SBARDELLA Gianluca UNIVERSITY OF SALERNO, FISCIANO (SA), IT SCARPELLINI Camilla UANTWERP, WILRIJK, BE SCHADE Dennis UNIVERSITY OF KIEL, KIEL, DE SCHEFFLER Frank HYPHA DISCOVERY, ABINGDON, GB SCHEPENS Wim JANSSEN, BEERSE, BE SCHIESSER Stefan ASTRAZENECA, MÖLNDAL, SE SCHIPP Kathrin NOVARTIS, BASEL, CH SCHLAPBACH Achim NOVARTIS, BASEL, CH SCHLESIGER Sarah MERCK, DARMSTADT, DE SCHNEIDER Gisbert ETH ZÜRICH, ZÜRICH, CH SCHNEIDER Nadine NOVARTIS, BASEL, CH SCHNIDER Patrick REJUVERON LIFE SCIENCES, SCHLIEREN, CH SCHREINER Peter R. JUSTUS-LIEBIG UNIVERSITY, GIESSEN, DE SCHUHMANN Tim NOVARTIS, BASEL, CH SCHULTE Clemens UNIVERSITY OF WUERZBURG, WUERZBURG, DE SCHUPPISSER Céline UNIVERSITY OF BERN, BERN, CH SCHWARDT Oliver UNIVERSITY OF BASEL, BASEL, CH SEAL Jon GSK, STEVENAGE, GB SERAFINI Marta UNIUPO, NOVARA, IT SHAW Duncan NOVARTIS, CAMBRIDGE, US SHIMIZU Takafumi MODULUS, TOKYO, JP SHOUKSMITH Andrew SELVITA SERVICES, KRAKÓW, PL SHTIL Alexander BLOKHIN CANCER CENTER, MOSCOW, RU SIEBER Stephan A. TU MUNICH, GARCHING, DE SIMIC Oliver NOVARTIS, BASEL, CH SMALLEY Adam UCB, SLOUGH, GB SMETS Wim CISTIM, HEVERLEE, BE SMITH James ASTRAZENECA, CAMBRIDGE, GB SNAJDROVA Radka NOVARTIS, BASEL, CH SOARES Pedro UNIVERSITY OF PORTO, PORTO, PT SOKLOU Kossi Efouako UNIVERSITY OF ORLEANS, ORLÉANS, FR SOUSA Maria Emilia UNIVERSITY OF PORTO, PORTO, PT SOUTHALL Stacey SOSEI HEPTARES, CAMBRIDGE, GB SPICER Julie UNIVERISTY OF AUCKLAND, AUCKLAND, NZ SPRUNG Matilda UNIVERSITY OF SPLIT, SPLIT, HR STAMPOLAKI Marianna NATIONAL AND KAPODISTRIAN UNIVERSITY OF ATHENS, ATHENS, GR STEADMAN Vicky SAI LIFE SCIENCES, ESSEX, GB STECHMANN Bahne EU-OPENSCREEN, BERLIN, DE STÉEN Johanna VU UNIVERSITY AMSTERDAM, AMSTERDAM, NL STEIMBACH Raphael R. GERMAN CANCER RESEARCH CENTER, HEIDELBERG, DE STEINEBACH Christian UNIVERSITY OF BONN, BONN, DE

496 | EFMC-ISMC

STEINER Sandra F. HOFFMANN-LA ROCHE, BASEL, CH STEPAN Antonia F. F. HOFFMANN-LA ROCHE, BASEL, CH STEWART Alison BENEVOLENTAI, LONDON, GB STILZ Hans Ulrich NOVO NORDISK, CAMBRIDGE, US STOLL Theodor F. HOFFMANN-LA-ROCHE, BASEL, CH STOUSE Adrien ULIEGE, LIÈGE, BE STRUWE Michel UNIVERSITY OF KIEL, KIEL, DE STYPIK Mariola CELON PHARMA, KIELPIN, PL SUZUKI Yamato DAIICHI SANKYO, TOKYO, JP SWAIN Christopher CAMBRIDGE MEDCHEM CONSULTING, DUXFORD, GB SZÁNTI-PINTÉR Eszter CZECH ACADEMY OF SCIENCES, PRAHA, CZ SZEREMETA-SPISAK Joanna RYVU THERAPEUTICS, KRAKOW, PL SZYMANSKI Wiktor UNIVERSITY OF GRONINGEN, GRONINGEN, NL SZYMASZEK Patryk POLITECHNIKA KRAKOWSKA, KRAKÓW, PL TASCI Hayrünnisa HACETTEPE UNIVERSITY, ANKARA, TR TAHTAOUI Chouaib ELANCO ANIMAL HEALTH, BASEL, CH TAKAHIRO Miyake KISHIDA CHEMICAL, OSAKA, JP TALUCCI Ivan JULIUS MAXIMILIANS UNIVERSITY WUERZBURG, WUERZBURG, DE TAMAYO Nuria AMGEN, THOUSAND OAKS, US TEIXEIRA-FOUCHARD Sylvie NOVARTIS, BASEL, CH TEOBALD Barry SITRYX THERAPEUTICS, OXFORD, GB TERNAD Indiana UMONS, CHAPELLE-LEZ-HERLAIMONT, BE TERSTIEGE Ina ASTRAZENECA, MÖLNDAL, SE THIEMANN Marie TU BRAUNSCHWEIG, BRAUNSCHWEIG, DE THOMA Gebhard NOVARTIS, BASEL, CH THOMPSON James GSK, STEVENAGE, GB THOMSON Phil APEX MOLECULAR, CHESHIRE, GB THUM Thomas HANNOVER MEDICAL SCHOOL, HANNOVER, DE TICHY Michal IOCB, PRAGUE, CZ TIEDT Stephanie FAU ERLANGEN, ERLANGEN, DE TIMMERMAN Henk VU AMSTERDAM, OEGSTGEEST, NL TINWORTH Christopher GSK, STEVENAGE, GB TITZ Alexander HELMHOLTZ INSTITUTE, SAARBRÜCKEN, DE TODA Narihiro DAIICHI SANKYO, TOKYO, JP TODOROV Milen NOVARTIS, BASEL, CH TOMASIC Tihomir UNIVERSITY OF LJUBLJANA, LJUBLJANA, SI TORMAKANGAS Olli ORION CORPORATION, TURKU, FI TORRENS Antoni ESTEVE PHARMACEUTICALS, BARCELONA, ES TORRES-GOMEZ Hector LEIBNIZ-HKI, JENA, DE TSCHAMMER Nuska WUXI APPTEC, DE TSIONOS Dimitris COLLABORATIVE DRUG DISCOVERY, CAMBRIDGE, GB TSIPA Maria IMPRS, ESSEN, DE TZARA Ariadni NATIONAL AND KAPODISTRIAN UNIVERSITY OF ATHENS, ATHENS, GR UBUKATA Minoru JT PHARMA, OSAKA, JP UHEREK Anna TAROS CHEMICALS, DORTMUND, DE ULIASSI Elisa UNIVERSITY OF BOLOGNA, BOLOGNA, IT ULLRICH Thomas NOVARTIS, BASEL, CH ULVEN Trond UNIVERSITY OF COPENHAGEN, KØBENHAVN Ø, DK VADLAMUDI Sree IKTOS, PARIS, FR VAISANEN Emilia ORION PHARMA, ESPOO, FI VALENTINI Alice UNIVERSITY OF COPENHAGEN, COPENHAGEN, DK VAN CALENBERGH Serge UGENT, GHENT, BE VAN DER STELT Mario LEIDEN UNIVERSITY, LEIDEN, NL VAN GOOL Michiel JANSSEN, TOLEDO, ES VAN HIJFTE Luc SYMERES, NIJMEGEN, NL VAN KLAVEREN Sjors UNIVERSITY OF LJUBLJANA, LJUBLJANA, SI VAN MAARSEVEEN Jan UNIVERSITY OF AMSTERDAM, AMSTERDAM, NL VANBELLINGHEN Alain UCB, BRAINE L'ALLEUD, BE VANDEVEER Harold BIOGEN, STONEHAM, US VANHERCK Jean-Christophe CISTIM, LEUVEN, BE VASCHETTO Mariana COLLABORATIVE DRUG DISCOVERY, CAMBRIDGE, GB VASSILIOU Stamatia NATIONAL AND KAPODISTRIAN UNIVERSITY OF ATHENS, ATHENS, GR VASTAKAITE Greta ETH ZURICH, ZURICH, CH VAUZEILLES Boris CNRS, GIF-SUR-YVETTE, FR VELCICKY Juraj NOVARTIS, BASEL, CH VELTER Ingrid JOHNSON & JOHNSON, BEERSE, BE VERWILST Peter KU LEUVEN, LEUVEN, BE VESELOVSKA Lucia IOCB, PRAGUE, CZ VIGATO Chiara UNIVERSITY OF TURIN, CASALE MONFERRATO, IT VISENTIN Sonja UNIVERSITY OF TORINO, TORINO, IT

EFMC-ISMC | 497

VITAL Paulo BIAL, SÃO MAMEDE DO CORONADO, PT VIVIANO Monica UNIVERSITY OF SALERNO, FISCIANO, IT VOLYNETS Galyna NAS OF UKRAINE, KYIV, UA VON NUSSBAUM Franz NUVISAN, BERLIN, DE VUJASINOVIC Ines FIDELTA, ZAGREB, HR WACH Jean-Yves F. HOFFMANN-LA ROCHE, BASEL, CH WAGNER Sven EUROFINS ADVINUS, DE WAN Murray MSD, BOSTON, US WANNER Benedikt SYNPLE CHEM, ZÜRICH, CH WARNOCK Tammy LIVERPOOL CHIROCHEM, LIVERPOOL, GB WATANABE Takumi BIKAKEN, TOKYO, JP WATANABE Keiichi KISHIDA CHEMICAL, OSAKA, JP WATT Alan NODTHERA, LITTLE CHESTERFORD, GB WEBSTER Robert BAYER, WUPPERTAL, DE WEIDNER Steffen NOVALIX, ILLKIRCH, FR WEISING Simon BIONTECH, MAINZ, DE WHITEHEAD David NOVARTIS, BASEL, CH WICHMANN Juergen F.HOFFMANN- LA ROCHE AG, BASEL, CH WIJTMANS Maikel UNIVERSITY OF AMSTERDAM, AMSTERDAM, NL WILDING Birgit BOEHRINGER INGELHEIM, VIENNA, AT WILKES Robin CHARNWOOD MOLECULAR, LOUGHBOROUGH, LEICS, GB WILLIAMS Pam CHARLES RIVER, US WILLIAMS Jodi IDORSIA PHARMACEUTICALS, ALLSCHWIL, CH WITT Julian UNIVERSITY OF HAMBOURG, MOORREGE, DE WITZKE Sarah CHEMICAL COMPUTING GROUP, CAMBRIDGE, GB WOLEK Barbara SELVITA, KRAKOW, PL WORKMAN Paul INSTITUTE OF CANCER RESEARCH, LONDON, GB WRIGHT Nathan GILEAD SCIENCES, SAN MATEO, US WU Yao ROCHE, SHANGHAI, CN WÜNSCH Bernhard UNIVERSITY OF MÜNSTER, MÜNSTER, DE WURNIG Silas UNIVERSITY OF BONN, BONN, DE WYCHOWANIEC Anna ICHB PAN, POZNAN, PL WYLER René NOVARTIS, BASEL, CH XIONG Huan UNIVERISTY OF PARIS, PARIS, FR XU Yongjin UNIVERSITY OF GOTHENBURG, GÖTEBORG, SE YANG Wu BRISTOL MYERS SQUIBB, PRINCETON, US YANG Shengyong SICHUAN UNIVERSITY, SICHUAN, CN YOKOKAWA Fumiaki NOVARTIS, EMERYVILLE, US YU Jianming ENANTA PHARMACEUTICALS, WATERTOWN, US ZACHAROVA Marija UNIVERSITY OF ST ANDREWS, ST ANDREWS, GB ZAJDEL Pawel JAGIELLONIAN UNIVERSITY, KRAKOW, PL ZAKARIA Ahmed BENI-SUEF UNIVERSITY, BENI-SUEF, EG ZALA Ajayrajsinh SARDAR VALLABHBHAI NATIONAL INSTITUTE OF TECHNOLOGY, SURAT, IN ZARZYCKA Barbara VRIJE UNIVERSITEIT AMSTERDAM, AMSTERDAM, NL ZAWADZKA Magdalena RYVU THERAPEUTICS, KRAKOW, PL ZETSCHOK Dominik ETH ZÜRICH, ZURICH, CH ZETTERBERG Fredrik GALECTO BIOTECH, GOTHENBURG, SE ZHANG Jian SHANGHAI JIAO TONG UNIVERSITY, SHANGHAI, CN ZHANGPING Xiao UNIVERSITY OF GRONINGEN, GRONINGEN, NL ZHU Wei F. HOFFMANN-LA ROCHE, SHANGHAI, CN ZIMMER Collin JOHANNES GUTENBERG UNIVERSITY, MAINZ, DE ZIMMERMANN Moritz TU BRAUNSCHWEIG, BRAUNSCHWEIG, DE ZIPFEL Pauline UNIVERSITY OF CAEN, CAEN, FR ZORC Branka UNIVERSITY OF ZAGREB, ZAGREB, HR ZORE Matej UNIVERSITY OF HELSINKI, HELSINKI, FI ZOU Ge ROCHE, SHANGHAI, CN ZUBRIENE Asta VILNIUS UNIVERSITY, VILNIUS, LT ZUMBRUNN Cornelia IDORSIA PHARMACEUTICALS, ALLSCHWIL, CH ZWERGEL Clemens UNIVERSITY OF ROME LA SAPIENZA, ROME, IT