medicinal chemistry introduction, part 1

40
Medicinal Chemistry Introduction, Part 1 9 Nov 2017 University Tor Vergata Steven Harper Ph.D.

Upload: others

Post on 29-Jan-2022

18 views

Category:

Documents


2 download

TRANSCRIPT

Page 1: Medicinal Chemistry Introduction, Part 1

Medicinal Chemistry Introduction, Part 1

9 Nov 2017University Tor Vergata

Steven Harper Ph.D.

Page 2: Medicinal Chemistry Introduction, Part 1

A bit about this course...

• Medicinal chemistry part of the course split into 4 lessons

• 2 lessons of introduction

– Today (Steven Harper)

› Broad introduction to medicinal chemistry and drug discovery

› Where do drugs come from and what features do drug molecules need to have

› Introduction to the early phases of rational drug design & how they work

– Future (Vincenzo Summa)

› More on drug candidate optimisation

› Toxicity and safety studies / clinical phases of drug discovery

• 2 lessons of more applied medicinal chemistry

– Problem solving on medicinal chemistry projects

– Case studies from medicinal chemistry programs

› Still to decide exactly what to present based on what is included in some of the upcoming lessons on

peptide chemistry, pharmacokinetics.

Page 3: Medicinal Chemistry Introduction, Part 1

What is medicinal chemistry?

• Simple idealistic answer: using organic chemistry to synthesise new drugs

• More realistic answer:

Medicinal chemistry is an infinitely complicated multi-disciplinary

science that uses organic chemistry, biochemistry, physics, biology,

computational chemistry and many other scientific disciplines to

drive the discovery and development of new drug therapies

• Medicinal chemistry can be exciting and rewarding

– the decision making engine that brings forward new drugs

– opportunity to do both applied science and be involved in creative/innovative research

• Medicinal chemistry is always demanding

– developing medicines is a long, expensive and far from easy process

Good: Bad:

Isentress Thalidomide

Page 4: Medicinal Chemistry Introduction, Part 1

Drugs are everywhere....

• Drugs are compounds that have an effect on human (or animal) biological systems

Everyday Drugs Recreational Drugs Pharmaceuticals

Red Bull contains taurine

- originally isolated from ox bile

- cardiovascular effects

- blood pressure changes

- vision impairment

Diamorphine / Heroin

- analgesic

- euphoria

- highly addictive

Morphine

- analgesic

- still addictive

very little distinction between drugs and drugs

Page 5: Medicinal Chemistry Introduction, Part 1

Drug discovery through history – ‘pre-scientific’ drugs

• Nature was the original source of drugs and has been making them for a long time

– no need for medicinal chemistry if you can find the finished drug in nature

– lots of drugs have come in full or in part from nature either pre-scientific or post-scientific

- chew willow bark to ease fever and

reduce inflammation

- white willow = Salix alba

- contains salicylic acid (aspirin)

Salicylic acid Aspirin (Prodrug)

- sweet wormwood extracts from

Chinese folklore dramatically blocked

plasmodium parasite growth

- sweet wormwood = Artemisa annua

- active species identified in 1972

Artemisinin – 2015 Nobel Prize in Medicine

Hippocrates (c.400BC) Tu Youyou (1930 to date)

Page 6: Medicinal Chemistry Introduction, Part 1

Drug discovery through history – serendipitous discoveries

Alexander Fleming 1928

- Staphylococci bacterial cultures left

beside open window during Fleming’s

vacation grew moldy

- mold appeared to kill some of the

bacteria

- penicillin antibiotics isolated by Florey &

Chain in time to save thousands of lives

in 2’nd world war

Penicillin G

Pfizer (1998)

- compound discovered in 1991 and reached

clinical trials for chest pain / angina

- found to be fairly ineffective treatment for

heart disease

- some male patients in the trials reported

unexpected side effects (and some refused

to give back their left over pills!)

- eventually marketed as Viagra

Sildenafil

Page 7: Medicinal Chemistry Introduction, Part 1

Drug discovery through history – modern approach

• Most new drugs in current times come from rational drug design

– many different approaches can be used but they more or less follow a standard workflow

– simplified timeline for rational drug discovery and some of the key scientific disciplines involved at

each stage shown in the scheme below

Target Identification

Hit DiscoveryHit to Lead Research

Lead Optimisation

Preclinical Development

Clinical Development

Biology

Biology Biochemistry

Comp. Sci.

Chemistry Biochemistry Biophysics

Chemistry Comp. Sci.

Phramacology

Phys. Chem

Pharmacology Toxicology

Medicine Statistics

1-3years 3-6months 3months 2years 6months-1year > 5years

• Cost of making a new drug is around $3 Billion

– these costs need to be recovered in a short space of time by drug companies due to patent rights

– drugs are expensive to make, and therefore expensive for patients

Page 8: Medicinal Chemistry Introduction, Part 1

Today’s Talk

Target Identification

Hit Discovery

Hit to Lead Research

What are the main families of

targets for drug molecules

How do compounds interact

with these targetsrs onths2years 6months-1year > 5years

Once we know what

our target is, how do

we start to find a drug?

- What are the

approaches for finding

compounds that bind

to the target

If we’re going to invest

$3Bn we need to be

sure we have a good

chance of success in

finding a drug

How do we choose

which compound

series to follow up on

What’s the difference

between a hit and a

lead?

Page 9: Medicinal Chemistry Introduction, Part 1

Target ID : knowing the target is key for drug discovery

• Biological targets where drugs interact are usually (but not always) proteins

Target Identification

• 3 main protein target types:

– Enzymes : catalyse specific chemical processes that influence biological events and pathways

› Proteases – hydrolysis reactions, usually protein hydrolysis

› Kinases – phosphorylation reactions

› Polymerases – facilitate DNA / RNA synthesis

– Receptors : bind naturally occuring molecules and the binding triggers a physiological event

› Neurotransmitter receptor – neuronal signalling

› Growth hormone receptors – switch on and off the pathways that lead tissues to undergo growth

– Protein-protein interactions : two (or more) proteins that bind together to influence a downstream

biochemical event in a physiological pathway

› Protein-protein interactions are extremely challenging targets for medicinal chemistry

Target identification/validation timeline : many months - many years

Page 10: Medicinal Chemistry Introduction, Part 1

Target ID – Enzymes

• Enzymes convert substrates (S) to products (P) at an ACTIVE SITE that usually mimics the

reaction transition state (TS)

– enzymes essentially lower the barrier to reactions by making the transition state more viable

TS

S

P

Progress of Reaction

Fre

e E

ne

rgy

Active Site Inhibitors - Inhibitors that bind in the active site

instead of the enzymes natural

substrate

- competitive inhibitors

- inhibitor binding blocks access to the substrate

Suicide Inhibitors - Inhibitors that covalently bind to the

enzyme to inactive its mechanism of

action

- also known as irreversible inhibitors

- inhibitor binding chemically inactivates the enzyme

preventing it from turning over any further substrate

Page 11: Medicinal Chemistry Introduction, Part 1

Target ID – Enzymes

• Enzymes convert substrates (S) to products (P) usually at an ACTIVE SITE that mimics the

reaction transition state (TS)

– enzyme essentially lower the barrier to reactions by making the transition state more viable

TS

S

P

Progress of Reaction

Fre

e E

ne

rgy

Allosteric Inhibitors - Inhibitors that bind at sites distal to

the active site.

- non-competitive inhibitors

- can be many allosteric sites on a single enzyme

- inhibitor binding often blocks a conformation of the

protein that prevents productive processing of the

substrate

Page 12: Medicinal Chemistry Introduction, Part 1

Target ID – Enzymes: an example – the protease family

• Proteases hydrolyse the peptide bonds in polypeptides and proteins

– essential enzymes in a multitude of biological processes from dietary to viral infections

– peptide bonds are highly stable and difficult to break, making the proteases a remarkable family

• Proteases hydrolyse the peptide bonds in polypeptides and proteins

– essential enzymes in a multitude of biological processes from dietary to viral infections

– proteases have pockets (S,S’) that recognize substrate side chains (P,P’) on either side of the

bond that they cleave

Peptide Substrate Four Families of Proteases

Serine Proteases

Cysteine Proteases

Metallo Proteases

Aspartyl Proteases

Scissile

BondFamily Nucleophile

Nucleophilic attack catalysed

by protease enzyme

N-Terminus C-Terminus

Serine CH2OH

Cysteine CH2SH

Water

Water

Page 13: Medicinal Chemistry Introduction, Part 1

Target ID – Enzymes: an example – the protease family

• Proteases also known as proteinases and peptidases

– highly specific specific enzymes – they do not hydrolyse peptides indiscriminately

– each different enzyme has a different preference for the specific groups at P1, P2, P3 etc

– P-side often crucial for selectivity with the S1 pocket being known as the ‘specificity pocket’

Enzyme

Substrate

Page 14: Medicinal Chemistry Introduction, Part 1

Target ID : Receptors

• Receptors are usually extracellular / cell-surface protein bundles

– Binding of an endogenous small molecule to the receptor can set off

a chain of complicated events INSIDE the cell

• Medicinal chemists can design small molecules that interact at the receptor

– binding of an exogenous small molecule to the receptor can achieve different outcomes

Antagonist

Exogenous

Small Molecule

Intracellular

SignalDescription

Agonist

Inverse Agonist

Antagonists are the receptor equivalent of enzyme inhibitors

- bind to receptor but do not produce intramolecular events

- block the binding of endogenous ligand

Off

Agonists are molecules that bind and switch on the receptor

- produce same effect as natural ligand, but usually stronger

(Partial Agonist)

On

Partly On

Inverse agonists bind to the same receptor but produce

the OPPOSITE effect inside the cell

(Partial Inverse Agonist)

Reversed

Partly reversed

Page 15: Medicinal Chemistry Introduction, Part 1

Hit discovery: what is a hit?

• A hit is a molecule that binds to the target protein at the site of interest

– classic model of interaction is Emil Fischer’s ‘lock and key’ theory

– protein binding sites are the locks, small molecule ligands are the keys

– early compounds in drug development and only need to bind & have a nice ‘tractable’ structure

– cell-based activity can be optimised later

Hit Discovery

Keap-1 protein with published ligand

Page 16: Medicinal Chemistry Introduction, Part 1

Hit discovery: what is a hit?

• A hit is a molecule that binds to the target protein at the site of interest

– small molecules (ligands) bind to proteins through a range of potential interactions

– overall binding follows the laws of thermodynamics

Direct binding involves enthalpy-driven

interactions between protein/ligand

Flexible structures can be less

complementary to the protein ‘lock’ but are

beneficial to entropy

- can ‘pre-pay’ entropy penalty needed to

get the binding conformation by making a

constrained inhibitor

Page 17: Medicinal Chemistry Introduction, Part 1

Hit binding is nearly always an equilibrium process

[P:D]

[P] x [D]

Both Enthalpy (DH) and Entropy (DS) changes affect binding strength

DG=-RTlnK and DG=DH-TDS

Gibbs Free Energy Changes

[Protein] + [Drug] [P:D]

K

For a binding Equilibrium between a Protein & a Drug

DG

Drug ProteinDrugProtein

K =

Page 18: Medicinal Chemistry Introduction, Part 1

Molecular interactions that make hits bind to proteins

• Charge-charge interactions (ionic interactions)

– Ionic groups on a protein surface can interact strongly with oppositely charged ions on a ligand

– Classic example is early thrombin inhibibitors

Thrombin

(Serine Protease)

Strong charge-charge

interaction between a basic

group on the inhibitor and a

conserved aspartic acid

residue in the P1 specificity

pocket

Page 19: Medicinal Chemistry Introduction, Part 1

• Van der Waals forces between hydrocarbon (aliphatic/hydrophobic) groups

– weakest intermolecular force (0.5-1.0 kcal/mole) but often there are lots of themHit Discovry

• Dipole-Dipole interactions such as hydrogen bonding

– electrostatic interaction between electron rich and electron poor atoms

– can be a strong interaction in a water free region of a protein (10kcal/mole)

– hydrogen bonds are directional and strength depends strongly on distance

• Also numerous other types of interactions that can contribute to binding

– cation-pi interactions

– pi-pi interactions

– ion-dipole interactions

N: H O

H

d-C

O

O H

O

H

H

d- d+

d+

d+

d-

Molecular interactions that make hits bind to proteins

Page 20: Medicinal Chemistry Introduction, Part 1

Entropy – hard to understand but important

• Based on the properties of the drug molecule, need to think about entropic effects involved in

dissolution – drugs need to be free from solvent to undergo enthalpic interactions with protein

When a hydrophobic drug is placed into water, the structure of

the water around the drug is more ordered.

This creates more ordered H2O-H2O H-bonding

This is lower entropy and is not favoured

Water molecules are preferably in a highly

disordered state.

Each molecule maximises H-bonds to other

water molecules

Page 21: Medicinal Chemistry Introduction, Part 1

Break

Page 22: Medicinal Chemistry Introduction, Part 1

How are hits found? – High Throughput Screening

• High Throughput Screening (HTS)

– most companies have collections of potential hits from 50,000 – 3 million compounds!

– test ALL of these compounds at one or two different concentrations to see if they bind

– select only the compounds that bind and then measure them at 10-20 concentrations to determine

IC50

IC50 – concentration of drug that

causes 50% inhibition of enzyme or

receptor activity

EC50 – effective concentration of

drug that causes 50% inhibition of

enzyme or receptor activity

- e.g. cell based assays

CC50 – concentration of drug that

causes 50% cytotoxicity (cell death)

in a cell based assay

Page 23: Medicinal Chemistry Introduction, Part 1

• Therapeutic window / therapeutic index can be important even for hits (selectivity index)

– therapeutic index is the ratio of toxic dose (TD50) / effective dose (ED50)

- usually needs to be >100

- higher TI means safer medicines

- two ways to improve the TI

make cpds more effective

make cpds less toxic

• For hits a selectivity index can be used to prioritize compounds

– selectivity index is the ratio of CC50 / EC50 or even IC50

How are hits found? – High Throughput Screening

Page 24: Medicinal Chemistry Introduction, Part 1

What properties do compound collections target

• IRBM’s compound collection is 120 000 compouds with carefully monitored properties

Structural diversity is a key to covering

‘Chemical Space’

- Redundancy needed to avoid false

negatives but should not be too high

- Murcko scaffolds often used to

compare molecules computationally

Mwt influences many things %Csp3 impacts solubility PSA influences absorption

Page 25: Medicinal Chemistry Introduction, Part 1

• Fragment based approaches based on a biophysical method

– modern approach to hid identification and lead identification

Fragments - typically libraries of very small molcules with molecular weight below 300

Biophysical Methods - X-ray crystallography (massive improvements in recent years)

Nmr spectroscopy

Surface plasmon resonance

How are hits found? – Fragment Based Approaches

Page 26: Medicinal Chemistry Introduction, Part 1

• Design from a natural product or endogenous ligand

– many drugs have come from natural receptor ligands as the original hits

How are hits found? – Other Approaches

Adrenalin Salbutamol (GlaxoSmithKline for asthma)

• ‘Me too!’ approaches

– copying other companies drugs or patent literature is an art

– not always easy to copy, but can be a very productive way to get to new starting points

– competition in pharmaceutical industry is huge, and highlights the importance of patents

› need to cover broad range of patent space to block me-too approaches from competitors

Viagra (Pfizer) Levitra (Bayer)

Page 27: Medicinal Chemistry Introduction, Part 1

Hit to lead efforts – where medicinal chemistry starts to get

interesting

• Hits are compounds that inhibit the target protein with a measurable IC50

Hit to Lead Research

• Leads are compounds that show improved potency with respect to the hit AND have some

other key properties

– the specific properties that medicinal chemists look for in a lead compound vary

– some key features are:

› chemical stability

› lead-likeness (tractable structure, suitable physicochemical properties)

› suitable early DMPK (Drug Metabolism & Pharmacokinetic) profile

Hit to Lead Medicinal Chemistry

Structure Activity Relationships

Page 28: Medicinal Chemistry Introduction, Part 1

What properties do drugs need to have?

• Whole range of properties and behaviours that are needed for them to be successful

– needs to be soluble in WATER

› all chemical reactions / interactions in humans occur in an aqueous solvent

› apart from bones, humans are well over 80% water

– needs to be stable in acidic environment of the stomach

– needs to get into the human bloodstream (absorption)

– needs to remain in circulation (metabolism / elimination as the body tries to get rid of the drug)

– needs to get to the site of action (distribution)

– needs to be non-toxic

– needs to be novel / patentable

• The fewer weak points (liabilities) a lead has in terms of these long term targets the higher

the probability of success of evolving the lead compound to a drug

– hit to lead work aims to get rid of any obvious issues in the hit compound

Page 29: Medicinal Chemistry Introduction, Part 1

Hit to lead: introduction to ADME properties

• Drug compounds need to have potency (usually inhibition of a target protein)

– activity is usually the easiest property to optimise

• ADME properties can be the most difficult to achieve

– to choose good hits we need to understand all the things that happen after swallowing a pill

The % of the oral dose

reaching the systemic

circulation is an

important PK parameter

F% = oral bioavailability

Page 30: Medicinal Chemistry Introduction, Part 1

Hit to Lead – example of a hit-to-lead process

HTS Screening Hit

IC50 19uM

Solubility >3mg/mL

Not patentable

Medicinal Chemistry Analysis

R = Me: IC50 200 uM

R = H: IC50 2 uM

Acid group

is needed

but better at

6-position

SAR at the Acid Group

R = CH3 IC50 90 uM

R = Ph IC50 120 uM

R = Chex IC50 0.7 uM

R = Cbut IC50 5 uM

SAR at the N-Alkyl Group

A cycloalkyl

group is

needed

SAR at the C2 Aromatic Group

R = CH3 IC50 Inactive

R = 4-Cl-Ph IC50 0.4 uM

R = Chex IC50 Inactive

An aryl

group is

needed at

C2

Page 31: Medicinal Chemistry Introduction, Part 1

Hit to Lead – example of a hit-to-lead process

Pharmacophore

- the parts of a ligand that are key to

the activity at a biological target

IC50 0.4 uM IC50 0.08 uM

Solubility < 0.01 mg/mLIC50 1.4 uM

Solubility >10 mg/mL

F% (rat) 37%

SAR Addressing SelectivitySAR at Core Group

Lead Compound

• The lead compound is not necessarily the most active compound that’s been made

– the lead is the compound judged to be the most developable starting point

– a lead typically has a well understood pharmacophore (or a pharmacophore model)

Early Optimised Hit

• Structure activity relationships (SAR) defines a pharmacophore

Core

modifications

are tolerated

Page 32: Medicinal Chemistry Introduction, Part 1

Overview – take home messages

• Seen the 3 main types of protein targets that drugs interact with

– Enzymes, receptors, protein protein interactions

Target Identification

Hit Discovery

Hit to Lead Research

• Should know some of the key interactions that are involved in protein-ligand intearactions

– Different energy contributions from VdW, electrostatic interactions

• Drugs and where they often originate

– Nature, good fortune or hard work

• Broad overview of the three early stages of drug discovery from a medicinal chemistry point

of view

Page 33: Medicinal Chemistry Introduction, Part 1

not used

Page 34: Medicinal Chemistry Introduction, Part 1

A bit about me.....

• Born : Scotland a long time ago

• Studied: University of Dundee (Degree) / University of Sheffield (PhD)

• Worked:

University of Siena (1994)

– 1 year post doc.

IRBM / Merck Sharpe & Dohme (13 years)

– Senior research fellow / project leader

– Worked exclusively in antiviral reasearch, especially on hepatitis C virus

IRBM Science Park (6 years)

– Head of medicinal chemistry unit

• Experience:

– worked on several projects that led to compounds that reached clinic studies

– development of Grazopevir (HCV) a marketed drug for hepatitis C virus

– still learning new things every day/week about medicinal chemistry and drug discovery

Page 35: Medicinal Chemistry Introduction, Part 1

What features are needed for absorption

• Drug compounds in this presentation all have one feature in common

MW 446 MW 282 MW 474 MW 239

• Compounds that are orally absorbed (by the most usual mechanism of passive diffusion)

almost always have molecular weight below 500

• 1997 : Lipinski’s Rule of Five (Christopher Lipinski, Pfizer)

– Set of rules that are help medicinal chemists optimize hits towards orally absorbed leads / drugs

• Oral absorption favoured if a molecule

– has molecular weight below 500

– has less than 5 hydrogen bond donors (typically OH or NH groups)

– has less than 10 hydrogen bond acceptors (disubstituted O atoms or N atoms in heterocycles)

– has a partition co-efficient (log P) less than 5 (logP = octanol vs. water distribution of neutral drug)

Page 36: Medicinal Chemistry Introduction, Part 1

Mechanisms of drug absorption

Transcellular absorption

– Main route for most oral drugs

– Drug must be in solution at cell surface

– pKa important - drug must be unionised

– Lipophilicity important

– Lipinski’s ‘Rule of 5’

Paracellular absorption

– drug passes through gaps between cells

– inefficient – pores have << surface area than cellular surface so hard for drugs to find the pores

– restricted to small polar molecules

Active Transport

– drugs carried through membrane by a transporter – requires energy

– many transporters exist for nutrient molecules, eg glucose, amino acids

– SAR specific – few drugs absorbed by this route

Page 37: Medicinal Chemistry Introduction, Part 1

Hydrophobic Bonding : D Entropy

Water molecules are

in a highly disordered

state. Each molecule

maximises H-Bonds

to other molecules

of water.

When a hydrophobic drug

is placed into water, the

structure of the water around

the drug is more ordered.

This allows the H2O-H2O

H-bonds to be maintained.

This leads to lower entropy

and is not favoured.

Page 38: Medicinal Chemistry Introduction, Part 1

Hydrophobic Bonding : D Entropy

D E

• Hydrophobic interaction between protein and drug is favoured by entropy gains:

• Bulk water returns to less ordered state

• Water molecules may be expelled from being bound in active site.

• In addition enthalpy gains due to new bonds may also be favourable

(e.g. van der Waals interactions)

DE

Page 39: Medicinal Chemistry Introduction, Part 1

NB. When a drug moves from the aqueous medium into the ‘Binding Site’ it has to

break H-Bonds with water, de-solvate etc. These processes require energy, so the

net energy available for binding is only a fraction of the above bond energies.

Drug-Protein Interactions

Bond Example kJ/mol

Van der Waal Xe…Xe, alkyl groups 2

Hydrophobic Ph…Ph (p-stacking) 5

Dipole - Dipole C=O…HN-R (d+/d-)...(d+/d-) 5

Hydrogen H2O…H2O (X-H) …(Y-R) 35

Ion - Dipole F-…H2O (+/-ve)…(d+/d-) 170

Ion - Ion H+…Cl- (+ve)…(-ve) 450

Covalent C-O 350

Page 40: Medicinal Chemistry Introduction, Part 1

MOUTH

INTESTINE

Blood

Gut wallMetabolism

Elimination

Systemic

Circulation

STOMACHpH ~1

pH ~ 7

Liver

Portal vein

Solubility

Acid stability

Permeability

Metabolism – gut wall / 1’st pass metabolism