polymeric micelles for sirna and aon delivery · polymeric micelles for sirna and aon delivery...

84
Polymeric Micelles for SiRNA and AON Delivery by Dianna Chan A thesis submitted in conformity with the requirements for the degree of Master’s of Applied Science Department of Chemical Engineering and Applied Chemistry University of Toronto © Copyright by Dianna Chan 2012

Upload: others

Post on 11-Mar-2020

4 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

Polymeric Micelles for SiRNA and AON Delivery

by

Dianna Chan

A thesis submitted in conformity with the requirements

for the degree of Master’s of Applied Science

Department of Chemical Engineering and Applied Chemistry

University of Toronto

© Copyright by Dianna Chan 2012

Page 2: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

ii

Polymeric Micelles for SiRNA and AON Delivery

Dianna Chan

Master’s of Applied Science

Department of Chemical Engineering and Applied Chemistry

University of Toronto

2012

Abstract

Immuno-nanoparticles of poly(ᴅ,ʟ-lactide-co-2-methyl-2-carboxytrimethylene carbonate)-g-poly(ethylene

glycol) (poly(LA-co-TMCC)-g-PEG) have been used to target breast cancer cells through the specific

binding of trastuzumab antibodies to over-expressed human epidermal growth factor receptor 2 (HER2).

Small interfering RNA (siRNA) and antisense oligonucleotides (AONs) disrupt the synthesis of select

proteins. It is hypothesized that oligonucleotides coupled to polymeric immuno-nanoparticles can be used

for gene silencing and specifically to target luciferase. The first objective is to demonstrate the capacity to

create dual functional micelles with antibodies and oligonucleotides. The second objective is in vitro

testing of the nanoparticle for gene silencing activity.

Oligonucleotides are conjugated to the nanoparticle by sequential click reactions of Diels Alder chemistry

and copper catalyzed azide-alkyne cycloadditions, respectively. A luciferase assay is used to quantify

knockdown of luciferase levels in SKOV-3luc cells (HER2+, luc+). When used in conjunction with a

targeted drug delivery vehicle, the nanoparticles provide selective interactions with SKOV-3luc cells.

Page 3: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

iii

Acknowledgements

I would like to thank my supervisor, Dr. Molly Shoichet, for her motivation and advice during my

Master’s degree. Her guidance has taught me to think critically within and outside of research problems

and I am grateful for the opportunity to work among such bright minds. It has been a great pleasure to

work with the lab members of the Shoichet group. In particular, I would like to thank ‘Team

Nanoparticle’ and our collaborators, Dr. Masad Damha and Glen Deleavey, for their support and

feedback on my thesis project. I would like to thank Dr. Shawn Owen for his continuous counsel and

contributions to my thesis project. Lastly, I would like to thank my family and friends for their continuous

support. I would also like to thank OGSST and NSERC for my funding.

Page 4: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

iv

Table of Contents

Abstract ........................................................................................................................................... ii

Acknowledgements ........................................................................................................................ iii

Table of Contents ........................................................................................................................... iv

List of Figures ............................................................................................................................... vii

List of Tables ............................................................................................................................... viii

List of Abbreviations ..................................................................................................................... ix

1. Introduction ........................................................................................................................... 1

1.1 Rationale ......................................................................................................................... 1

1.2 Hypothesis and objectives............................................................................................... 1

1.3 Polymers for targeted drug delivery ............................................................................... 2

1.3.1 Passive tissue targeting ............................................................................................... 2

1.3.2 Active cellular targeting .............................................................................................. 3

1.4 Small Interference RNA (siRNA) and Antisense Oligonucleotides (AON) .................. 4

1.4.1 Chemical modifications to siRNA and AONs ............................................................ 5

1.5 Nanoparticle modification by click chemistry ................................................................ 8

1.5.1 Click chemistry ........................................................................................................... 8

1.5.2 Multi-functional nanoparticles .................................................................................. 10

1.6 Cellular Uptake and Release ......................................................................................... 10

1.7 Summary of Research ................................................................................................... 11

1.8 References ..................................................................................................................... 13

2. Double click: Dual functionalized polymeric micelles ..................................................... 17

2.1 Abstract ......................................................................................................................... 17

2.2 Introduction ................................................................................................................... 17

2.3 Materials and Methods .................................................................................................. 19

2.3.1 Polymer Synthesis ..................................................................................................... 21

2.3.2 Micellization ............................................................................................................. 23

2.3.3 Conjugation of Trastuzumab and FLAG Peptides to the Nanoparticles................... 24

2.3.4 Cell Culture and In Vitro Assessment of Dual Functionalized Micelle Uptake ....... 27

2.4 Results ........................................................................................................................... 27

2.4.1 Polymer Synthesis ..................................................................................................... 27

Page 5: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

v

2.4.2 Micellization ............................................................................................................. 28

2.4.3 Conjugation of Trastuzumab and FLAG Peptides to the Nanoparticles................... 28

2.5 Discussion ..................................................................................................................... 32

2.6 Conclusion .................................................................................................................... 34

2.7 References ..................................................................................................................... 34

3. Delivery of SiRNA and Antisense Oligonucleotides using Dual Functional Polymeric

Immuno-nanoparticles ............................................................................................................... 36

3.1 Abstract ......................................................................................................................... 36

3.2 Introduction ................................................................................................................... 36

3.3 Materials and Methods .................................................................................................. 38

3.3.1 Polymer Synthesis and Characterization .................................................................. 39

3.3.2 Oligonucleotide Synthesis and Characterization ...................................................... 40

3.3.3 Nanoparticle Formation and Characterization .......................................................... 40

3.3.4 Luciferase Characterization ...................................................................................... 41

3.4 Results ........................................................................................................................... 42

3.5 Discussion ..................................................................................................................... 50

3.6 Conclusion .................................................................................................................... 52

3.7 References ..................................................................................................................... 53

4. Conclusions .......................................................................................................................... 56

5. Recommendations and Future Directions ........................................................................ 57

Appendix A: Supplemental Information .................................................................................. 61

A1. Characterization of NH2-PEG-azide ............................................................................. 61

A2. Characterization of NH2-PEG-furan ............................................................................. 62

A3. Characterization of P(LA-co-TMCC) ........................................................................... 64

A4. Characterization of P(LA-co-TMCC)-g-PEG ............................................................... 66

A5. DA Reaction with NH2-PEG-Fu ................................................................................... 68

A6. SPAAC Reaction with NH2-PEG-N3 ............................................................................ 68

A7. Size Exclusion Column Purification of Micelles .......................................................... 69

A8. Confocal Images of SKOV-3luc controls ..................................................................... 70

A9. Click Chemistry Reactions ........................................................................................... 71

Page 6: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

vi

Appendix B: siRNA and AON Synthesis and Characterization ............................................. 72

B1. Synthesis of siRNA and AON sequences ..................................................................... 72

B2. Cleavage and Purification ............................................................................................. 72

B3. Quantitation and characterization ................................................................................. 73

B4. Annealing of oligonucleotides. ..................................................................................... 73

Appendix C: Statistical Analysis

C1. SiRNA Delivery ............................................................................................................ 75

C2. AON Delivery ............................................................................................................... 75

Page 7: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

vii

List of Figures

1. Introduction

1.1 Structure of poly(LA-co-TMCC)-g-PEG ........................................................................... 3

1.2 Mechanisms of action for siRNA and AONs. .................................................................... 4

1.1 Overview of oligonucleotide delivery through non-viral vehicles ..................................... 7

1.3 Click reaction schemes ....................................................................................................... 9

1.4 Flowchart of plan .............................................................................................................. 12

1.5 Poly(LA-co-TMCC)-g-PEG nanoparticle coupled with siRNA or AONs ....................... 12

2. Double click: Dual functionalized polymeric micelles

2.1 Synthesis of NH2-PEG-azide and NH2-PEG-furan .......................................................... 21

2.2 Synthesis of P(LA-co-TMCC)-g-PEG-X (X=azide or furan) .......................................... 23

2.3 FPLC purification of nanoparticles ................................................................................... 31

2.4 Confocal images of SKOV-3luc cells ............................................................................... 32

3. Delivery of SiRNA and AONs using Dual Functional Polymeric Immuno-nanoparticles

3.1 siRNA and AON Delivery Strategies. .............................................................................. 43

3.2 Luciferase levels of cells treated with siRNA immuno-nanoparticles .............................. 45

3.3 Luciferase levels of cells treated with AON immuno-nanoparticles ................................ 47

3.4 Luciferase levels of cells treated with controls. ................................................................ 48

3.5. Persistence effect of siRNA immuno-nanoparticles. ....................................................... 49

3.6 Persistence effect of AON immuno-nanoparticles. .......................................................... 50

Appendix A: Supplemental Information

A1. Characterization of NH2-PEG-azide by FTIR. ................................................................ 61

A2. Characterization of NH2-PEG-furan by 1H NMR ............................................................ 63

A3. Characterization of P(LA-co-TMCC) by 1H NMR.......................................................... 65

A4. Characterization of P(LA-co-TMCC)-g-PEG-X by 1H NMR ......................................... 67

A5. DA Reaction with NH2-PEG-Fu.. .................................................................................... 68

A6. SPAAC Reaction with NH2-PEG-N3 and Alexa 488-dibenzylcyclooctyne. ................... 68

A7. Size exclusion column purification of mixed micelles. ................................................... 69

A8. Confocal images of SKOV-3luc cells controls. ............................................................... 70

Page 8: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

viii

List of Tables

1. Introduction

1.1 Overview of oligonucleotide delivery through non-viral vehicles ................................. 7

2. Double click: Dual functionalized polymeric micelles

2.1 Dual-functional micelle reactions with maleimide- and dibenzylcyclooctyne-functional

moieties ............................................................................................................................... 29

3. Delivery of SiRNA and AONs using Dual Functional Polymeric Immuno-nanoparticles

3.1. Characterization of poly (LA-co-TMCC) .................................................................... 39

3.2. Characterization of poly(LA-co-TMCC)-g-PEG-X graft copolymer .......................... 40

3.3. Coupling efficiency of oligonucleotides to micelles ................................................... 41

5. Recommendations and Future Directions

5.1. Potential siRNA target genes ....................................................................................... 58

C. Statistical Analysis

C1. SiRNA Delivery ........................................................................................................... 75

C2. AON Delivery .............................................................................................................. 75

Page 9: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

ix

List of Abbreviations

2′F-ANA, 2′F-RNA 2′-Deoxy-2′-fluoroarabino nucleic acid

AON Antisense oligonucleotide

CuAAC Copper catalyzed azide-alkyne cycloaddition

DA Diels Alder

DBCO Dibenzylcyclooctyne

Di Dicer

Fu Furan

LipA Lipofectamine

MI Maleimide

ON Oligonucleotide

Poly(LA-co-TMCC) Poly(ᴅ,ʟ-lactide-co-2-methyl-2-carboxytrimethylene carbonate)

SiRNA Small interfering ribonucleic acid

SPAAC Strain promoted azide-alkyne cycloaddition

Page 10: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

1

1. Introduction

1.1 Rationale

Among women, breast cancer is the most commonly diagnosed cancer. It accounts for 14% of cancer-

related mortality; the second deadliest cancer for women. In 2008, 1.38 million women worldwide were

newly diagnosed with breast cancer [1]. Breast cancer is a slowly progressive disease and its costs to

individuals are measured in years or decades. The cost of follow-up procedures and management of

recurrent disease was estimated to cost over $454 million Canadian in one year [2].

Chemotherapeutic drugs, such as taxanes, are commonly used for the treatment of cancer. However,

systemic toxicity can have negative side effects on healthy tissue. Based on literature review, gene

therapy with siRNA and AONs are less toxic to the body than chemotherapeutics. These gene silencing

strategies will only interrupt the translation of proteins in mRNA with matching base pairs [3]. Thus, the

selectivity can minimize the negative effects of systemic drug distribution.

Typically gene therapy is limited by the instability of siRNA or AON in serum. However, the modified

siRNA and AONs provided from the Damha Group at the University of McGill will address this problem

[4, 5]. Combined with a biocompatible copolymer such as poly(LA-co-TMCC)-g-PEG-azide, the system

can be used for targeted delivery. Our goal is to prevent cancer cell growth by using copolymer

nanoparticles to deliver siRNA.

1.2 Hypothesis and objectives

It is hypothesized that siRNA or AONs coupled to polymeric immuno-nanoparticles will have targeted

gene silencing activity. SiRNA or AONs can be used for selective gene knockdown of cells. Polymeric

immuno-nanoparticles formed by poly(LA-co-TMCC)-g-PEG have been prepared to target HER2. HER2

is overexpressed in breast cancer cells and specifically binds to the antibody trastuzumab (Herceptin®).

Poly(LA-co-TMCC)-g-PEG has also been used as a biodegradable delivery vehicle for the

chemotherapeutic drug doxorubicin. Use of oligonucleotides for selective gene knockdown may provide

an alternative to chemotherapeutics in the copolymer drug delivery vehicle. Gene knockdown can be

modelled by selecting an oligonucleotide sequence specific to firefly luciferase. Measurements of

bioluminescence of cells expressing firefly luciferase, SKOV-3luc cells, will demonstrate the

effectiveness of gene knockdown.

Page 11: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

2

The first objective is to demonstrate the capacity to create dual functional micelles. The second objected

is to demonstrate that the immuno-nanoparticles have a gene knockdown effect.

1.3 Polymers for targeted drug delivery

Nanocarriers have shown promise for therapeutic applications that can exploit its controllable size and

material properties to protect drugs or therapeutic moieties from degradation, increase drug solubility,

enhance delivery through physiological barriers and modify its pharmacokinetics and distribution profile.

The choice of material for the carrier can range from lipids, to metal nanoparticles and polymers [1].

Polymer therapeutics is a general term that includes the use of polymers in the form of micelles

covalently bound to drugs, polymer-drug conjugates, polymer-protein conjugates, or multi-component

polyplexes [2]. Drugs delivered by polymeric vehicles display features shown by macromolecular drugs,

but include the versatility of synthetic chemistry. Adjusting the molecular weight or adding bioresponsive

elements allow the therapeutic vehicle to be tailored [2]. Polymers are a logical option for targeted drug

delivery because they can be modified to optimize size, hydrophobicity or hydrophilicity, and targeting

properties. Most polymeric nanoparticles are amphiphilic; there is a hydrophobic core and hydrophilic

shell. The core allows hydrophobic drugs to be encapsulated [3]. With cancer therapy, polymeric micelles

face the challenge of increasing drug efficacy and reducing toxicity by controlling its biodistribution and

improving intracellular penetration [1].

1.3.1 Passive tissue targeting

Use of polymeric anticancer therapeutics is rationalized by the ‘enhanced permeability and retention’

(EPR) effect. Hyperpermeable angiogenic tumour vasculature allows long circulation times for polymer

therapeutics [2]. There are two main features that result in the EPR effect: the disorganized pathology of

tumour vasculature and the lack of effective tumour lymphatic drainage. As a result, solid tumours of

breast cancer are hyperpermeable to circulating polymeric nanoparticles. Thus, the nanocarriers are able

to accumulate [4, 5]. Polymeric micelles are promising drug carriers that can benefit from the passive

targeting due to its size. However, the following design criteria are required for effective drug delivery:

the micelle must be between 10-200 nm to penetrate into tissue, be biocompatible, be tuneable for

stability and pharmacokinetic profiles, and be able to interact and locate targeted cells[6]. The micelles

must circulate long enough to accumulate in the targeted tissue. Opsonin proteins in the blood serum can

bind to nanoparticles to signal removal by the macrophages in the mononuclear phagocytic (MPS) system

Page 12: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

3

[7]. One common strategy to address this issue is the grafting of the biocompatible poly(ethylene glycol)

(PEG) to decrease opsonization of the nanoparticles.

1.3.2 Active cellular targeting

Active targeting is another strategy used to deliver the nanoparticles and its cargo to tumour tissue.

Specific ligand targeting proteins or antibodies are expressed on cancer cell membranes or cells lining the

blood vessels of the tumour. Conjugation of proteins, antibodies, or ligands can improve the specificity of

the drug delivery vehicle [1].

In order to improve drug targeting to breast cancer cells, a drug delivery system using poly(LA-co-

TMCC)-g-PEG (Figure 1.1) to form nanoparticle micelles has been previously characterized. Poly(LA-

co-TMCC)-g-PEG forms stable nanoparticles, has a low polydispersity index, and the carboxylate group

can be used to graft amine-terminated PEG. The poly(LA-co-TMCC)-g-PEG molecules can self assemble

into nanoparticles [8].

Figure 1.1: Structure of poly(LA-co-TMCC)-g-PEG. Each polymer chain contains one pyrene molecule

from the pyrene butanol initiator. Dispersed segments of ᴅ,ʟ-Lactide (LA) and 2-methyl 2-

carboxytrimethylene carbonate (TMCC) monomer units compose the polymer chain. Poly(ethylene

glycol) is grafted onto approximately one TMCC unit per polymer backbone. X represents a functional

group of furan or azide. The degree of polymerization is controlled by the quantity of monomers used for

synthesis [8].

LA TMCC

Page 13: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

4

The critical aggregation concentration (CAC) is the minimum polymer concentration required for self-

assembled structures to form. The CAC reflects thermodynamic stability. The low values (50 nM for

poly(LA-co-TMCC) and 200 nM for poly(LA-co-TMCC)-g-PEG) indicate that limited dissociation will

occur. The polymers also successfully encapsulated docetaxel. This shows the ability of the nanoparticle

to deliver a hydrophobic cancer drug [9]. Further modification through the attachment of antibodies for

specific binding to cells overexpressing HER2 antigens to these micelles has demonstrated increased drug

targeting [10].

1.4 Small Interference RNA (siRNA) and Antisense Oligonucleotides (AON)

Oligonucleotides have been shown to inhibit target RNA functions by interfering with translation of

mRNA. Two strategies include the use of synthetic antisense oligonucleotides and RNA interference [11].

Antisense therapy uses antisense oligonucleotides (AONs) that are typically modified to improve serum

stability. RNA interference (RNAi) uses small interfering RNA (siRNA). In both cases, the mechanism of

action begins when AONs or siRNA bind to target RNA and activate nucleases that causes RNA

degradation (Figure 1.2) [12].

Figure 1.2: Mechanisms of action for siRNA and AONs.

DNA

mRNA

Small interference RNA (siRNA)

Antisense oligonucleotide (AON)

dsDNA

Dicer RISC

RNase H

Page 14: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

5

Antisense RNAs were discovered in 1981 when it was shown that RNA regulators could control the copy

numbers of Escherichia coli plasmids [13, 14]. Antisense RNAs are sequences that target complementary

sense RNA for translational blocking or activation of ribonuclease H (RNase H). RNase is found naturally

in most cells and can act on immature and mature mRNA [12, 15].

RNAi was later discovered in 1998 by Fire et al. [16]. Post-transcriptional gene silencing was later shown

in mammalian cells by Elbashir et al. for the specific knockdown of proteins in HeLa cells [17]. It has

been demonstrated that synthetic short RNA duplexes can display high potency in gene knockdown.

SiRNA is an oligonucleotide composed of approximately 20 to 25 base pairs [18]. It enters the cell and

associates with an RNA induced silencing complex (RISC). There, it cleaves one of the RNA strands and

leaves the single stranded RNA incorporated with the RISC. The complex downregulates specific genes

[19]. The Dicer is the active nuclease that catalyzes the cleavage of dsRNA [20].

Oligonucleotide therapy is advantageous over small molecule drugs because the RNA and DNA

sequences can be designed for specific inhibition of the target of interest [21]. AON and siRNA therapy

display strong potency and specificity in cell culture once cellular uptake can be achieved. However, gene

therapy faces the obstacles of poor nuclease stability leading to rapid degradation, poor unassisted cellular

uptake, and unintended side-effects from “off-target” silencing and immunostimulation [21, 22].

Currently, these challenges are addressed through the use of delivery vehicles and chemical modifications

to the oligonucleotide backbones to prevent nuclease degradation and undesired side effects [18, 19, 22].

1.4.1 Chemical modifications to siRNA and AONs

Phosphorothioate modifications to DNA can increase serum stability of AONs while maintaining their

ability to activate RNase H [23]. One drawback of these modifications is their toxicity and non-specific

binding to proteins [24]. The Damha group at McGill University has chemically modified

oligonucleotides for improved stability. Modifications include changes to the sugar, phosphate linkage,

base, overhangs, and termini [18]. Protection at the 2′-hydroxyl group has shown increased stability of

siRNA. Chemical modifications include 2′-O′methyl, 2′-fluoro, and 2’-O-(2-methoxyethyl) groups [19].

Fluorination of the 2′-carbon of sugar rings (2′F-ANA) has become a useful chemical modification in

both siRNA and AON gene silencing applications [22, 25-30]. With AON treatment, 2′F-ANA combined

with the phosphorothioate backbone modification enhances nuclease stability [32-35]. For siRNA

applications, 2′F-ANA improves siRNA nuclease stability and can be incorporated in the siRNA

Page 15: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

6

passenger strand [26, 27]. When used in combination with 2′F-RNA, siRNAs exhibit improved potency

and reduced immunostimulatory properties[28].

Another limitation of oligonucleotides is the inability of the negatively charged molecules to cross the cell

membrane and begin the antisense or RNAi response [19]. Oligonucleotides alone have a low transfection

efficiency, poor tissue penetration and non-specific immune stimulation. Nucleic acids are highly charged

and will not cross the cell membrane by free diffusion [21].

Additionally, research by Santel et al. evaluated the biodistribution of naked siRNA and concluded that

siRNA does not target any specific tissue. After injection, most of the siRNA was eliminated by renal

clearance [21]. Thus, attachment to a delivery vehicle is necessary. Both viral and nonviral delivery

methods have been demonstrated [19]. Viral vectors are very effective, but clinically limited. Viral

vectors can have potential mutagenicity or host immune responses [19]. Non-viral delivery vehicles are a

good alternative. Different systems include: chemical modification of siRNA or AONs, cationic

polymers, lipids, and targeted delivery [19].

A hurdle with gene therapy is the ability to internalize siRNA or AONs specifically into the cell. Cell-

specific delivery has been achieved using targeting ligands, such as antibodies and cell membrane

receptors [21]. Methods used to deliver oligonucleotides in vitro are summarized in Table 1.1.

Page 16: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

7

Table 1.1: Overview of oligonucleotide delivery through non-viral vehicles [31]

Category Carrier Description Limitations Lipid

complex

Cationic liposomes

(DOTAP, DOTMA,

Lipofectamine) [32]

Cationic lipids form lipoplexes

with negatively charged siRNA

for high transfection.

Liposomes can also interact with

serum proteins and aggregate.

Lipofectamine also has dose-

dependent toxicity.

Neutral liposome

(DOPC, DOPE) [33, 34]

SiRNA is encapsulated using

simple mixing and incubation.

Ligands are conjugated for

specific delivery.

Stability; fusion of liposomes to

form large particles that clear

quickly.

Lipoplexes (DOTAP and

poly(arginine)) [32, 35,

36]

Lipoplexes are cationic

liposomes complexed with

nucleic acids. The lipoplex is

delivered through the cellular

membrane. Less toxicity is

exhibited than cationic liposome

alone.

Little control of interaction

between siRNA and plasmid DNA

or RNA used to deliver the

lipoplex through the membrane.

This leads to excessive sizes and

low stability.

Conjugated

polymers

Targeting molecule

(peptides, antibodies,

aptamers) [35]

Conjugation can be attached to

either the sense or antisense

strand without compromising the

activity of siRNA silencing.

Antibodies and proteins are large

molecules and alter the drug

delivery vehicle size. For

aptamers, the major challenge is

internalization of cargo after the

ligand interacts with the receptor.

Polymer-PEG [35, 37,

38]

PEG prevents non-specific

binding of proteins. PEG can be

linked to siRNA through a pH

sensitive bond that breaks when

internalized and exposed to the

acidic endosome.

Pegylation can decrease the in

vitro activity of proteins, but this

is usually offset by its increased

half-life.

Cationic

polymers

Synthetic polymers (PEI)

[21, 39]

The high cationic charge density

allows PEI to exhibit a proton

sponge effect to swell

endosomes and cause release of

siRNA in the cell.

PEI is cytotoxic in vivo. It may

induce nonspecific interferon

response.

Natural polymers

(chitosan, atellocollagen)

[36, 45]

The polymers are biodegradable.

They form complexes with

negatively charged siRNA.

Maximizing the activity of

chitosan carriers is being

investigated by Howard, et al.

Cyclodextrin [36, 46] Cyclodextrin molecules have a

hydrophilic outer surface and

hydrophobic cavity. Molecules

form vesicles that carry siRNA.

For improved uptake and release,

modifications with polycations,

targeting ligands and PEG are

being investigated.

DOPC – 1,2-Oleoyl-sn-Glycero-3-phosphocholine

DOPE – 1,2-Dioleoyl-sn-Glycero-3-phosphoethanolamine

DOTAP – 1-oleoyl-2-[6-[(7-nitro-2-1,3-benzoxadiazol-4-yl)amino]hexanoyl]-3-trimethylammonium propane

DOTMA – N-[1-(2,3-dioleoyloxy)propyl]-N,N,N-trimethyl-ammonium methyl sulphate

PEI – polyethyleneimine

Although siRNA and AON both target mRNA for gene silencing, their differences in mechanism of

action result in different levels of knockdown in similar conditions. SiRNA shows better inhibition of

mRNA at lower doses; approximately 10-fold higher concentrations of AONs were required to achieve

inhibition comparable to siRNA [47]. Nevertheless, both AON and siRNA provide potential strategies for

Page 17: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

8

the therapeutic treatment of cancer. Antisense therapy has the benefit of being a more mature technology,

while siRNA has the advantage of increased sensitivity at low concentrations.

1.5 Nanoparticle modification by click chemistry

Nanocarriers have often been surface modified to achieve targeted cellular and intracellular delivery or to

monitor cell uptake and release [40]. The covalent coupling of antibodies, proteins, peptides, fluorescent

molecules, or contrast agents provides additional functionality and versatility [41]. In contrast to the

encapsulation of hydrophobic drugs in the core, targeting moieties require exposure to receptors on or

within the cell. For diagnostic purposes, MRI contrast agents that require the chelation of metal atoms

require exposure to the aqueous environment for enhanced contrast properties [42].

However, surface modification of micelles is not limited to targeting or diagnostic applications.

Therapeutic molecules can also be conjugated to the micelle shell. Doxorubicin has been coupled to the

PEG terminus of poly(LA-co-TMCC)-g-PEG-furan micelles using Diels Alder chemistry with

maleimide-modified doxorubicin [43].

To maintain the bioactivity of these molecules, the chemistry required for conjugations have restrictions

with pH, temperature and exposure to organic solvents [41]. Early chemistries include activated esters for

condensation with amines[20], maleimide-thiol reactions [24], aldehydes and amines [11, 23], and biotin

binding to avidin [44]. An increasingly popular approach uses click chemistry reactions [52-54].

1.5.1 Click chemistry

Click chemistry reactions are classified as reactions that are wide in scope, produce high yields, and

generate mild byproducts. Two types of click chemistry reactions are Diels Alder chemistry and the

Huisgen dipolar cycloaddition between azides and alkynes [45]. The reaction schemes are shown in

Figure 1.3.

Page 18: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

9

Figure 1.3: Click reaction schemes. All reactions can be completed in water at room temperature.

The Diels Alder reaction is a cycloaddition reaction between an electron-rich diene and electron-poor

dienophile. Rideout and Breslow showed that the rate of Diels Alder reactions could be increased in

aqueous environments by bringing together hydrophobic interactions of the reactive groups [46]. The

mild reaction conditions and quantitative yields allow nanocarriers to be modified with antibodies,

peptides, fluorescent probes, affinity tags and oligonucleotides [47, 48].

The most common click reaction is the copper catalyzed alkyne-azide (CuAAC) reaction. The addition of

a copper catalyst increases the rate of reaction by forming a coordination complex with the acetylide. The

rate of reaction has been shown to increase by seven orders of magnitude and the yield of CuAAC

reactions in water range from 40-90% [49].

Alternatively, a copper-free reaction can be achieved using a strain-promoted alkyne-azide cycloaddition

(SPAAC) reaction [53, 59]. SPAAC reactions overcome the potential cytotoxicity of metal ion

contamination and degradation of oligonucleotides [50, 51]. Commonly, strained cyclooctynes are used to

Diels Alder diene dienophile cyclohexene Huisgen dipolar cycloaddition Copper Catalyzed Alkyne-Azide

Cu(I) azide acetylene 1,4-triazole Strain Promoted Alkyne-Azide

azide cyclooctyne 1,4-triazole

Page 19: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

10

enable the click reaction [50]. Fluorinated cyclooctyne rings or dibenzocyclooctynes are commonly used

to react with azides [50, 52]. Azide modified peptides have been coupled to PEO-PCL micelles with

dibenzocyclooctyne moieties with 58-76% yield [52].

1.5.2 Multi-functional nanoparticles

An ideal therapeutic carrier needs to incorporate multiple features: therapeutic cargo, ligands to facilitate

cellular or subcellular targeting, the ability to maintain stable, and a safe profile [53]. To meet these

criteria, multi-functional nanoparticles have been synthesized to combine various moieties onto a single

vehicle. Weissleder first synthesized triple labelled nanoparticles with a magnetic core and fluorescent

and isotopic tags [54]. More recently, “octa-functional” PLGA nanoparticles were used to deliver siRNA.

The eight functions included: 1) the polymeric micelle vehicle, 2) the therapeutic moiety (siRNA), 3)

PEG to improve nanoparticle circulation, 4) a small molecule enhancer for siRNA, 5) an endosomal

escape agent, 6) cell penetrating peptides, 7) peptides for endosomal escape, and 8) tumor targeting

peptides [53]. Most nanocarriers include variations of the first three functions listed.

With the addition of multiple ligands and cargo to micelles, there is added complexity to the synthesis

process. One-step reactions and minimal purification steps are advantageous in terms of maximizing

yields and minimizing reaction time. Double click reaction strategies combine orthogonal reactions for

post-modification of polymers [55]. CuAAC and Diels Alder reactions have been used by Hizal, et al. in a

one pot reaction to synthesize linear triblock copolymers. The orthogonal and independent reactions with

azide-alkyne and anthracene-maleimide cycloadditions produced PMMA-b-PS-b-PEG [56]. Variations

with CuAAC and Diels Alder double click reactions have been used to produce graft copolymers [57, 58],

star polymers [59], dendrimers [60, 61] and cyclic polymers [62]. More recently, Gunay et al. synthesized

a 3-arm star terpolymer with sequential Diels-Alder, then CuAAC and nitroxide radical coupling

reactions. PEG with maleimide functionality, poly(ε-caprolactone) with tetramethylpiperidine-1-oxyl

functionality and polystyrene with azide functionality were conjugated to a multifunctional core [63].

1.6 Cellular Uptake and Release

The charge of the delivery system will affect siRNA uptake. Highly charged particles can bind to proteins

and lead to immune stimulation and rapid clearance. Another factor for cell uptake and clearance is size

[21]. If a carrier system is used, it is typically taken up by endocytosis. After entering the cell, the siRNA

must escape from the endosomes [64].

Page 20: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

11

Escape from the endosome is not well understood, but it is hypothesized that the endosome undergoes a

‘proton sponge effect’: the acidic pH of the endosome interacts with the negatively charged nanoparticle

to escape into the cytoplasm [21]. A common vehicle used for siRNA is polyethyleneimine (PEI). The

PEI acts as a buffer in the endosome and supplies protons into the endosome. To maintain a charge

balance, there is an influx of chloride ions. Osmotic swelling occurs and the endosome bursts. By

escaping the endosome, the siRNA escapes the lysosomal trafficking pathway [21]. Negatively charged

PLGA nanoparticles were also able to deliver siRNA after surface modification with positively charged

PEI [65]. Thus, the charge of the polymer is a major factor which affects the endosomal escape [21].

Other types of co-polymers used to deliver siRNA have been shown by Kataoka et al. PEG-polycation di-

block copolymers have been engineered to have high endosomal escape. A diamine side chain has a

distinct pKa that results in siRNA encapsulation in polyplex micelles [21]. Sun et al. developed a cationic

triblock copolymer mPEG45-bPCL100-b-PPEEA12 for the delivery of siRNA. The vehicle was

biocompatible, biodegradable and demonstrated effective gene silencing in 40-70% of the targeted cells

[76]. The choice of polymer will affect the biodistribution and release kinetics of the drug.

Liposomes have proven to be good carriers for oligonucleotides [28]. Cationic liposomes complex with

negatively charged oligonucleotides to overcome the electrostatic repulsion of the cell surface and induce

local destabilization of the plasma membrane [66, 67]. Anionic lipids in the endosome membrane

complex with the cationic lipids. As a consequence, AONs are translocated into the cytoplasm [80, 81].

1.7 Summary of Research

Polymers can be effective vehicles for drug delivery due to their biocompatibility and potential for

attaching targeting moieties. Attachment of antibodies, siRNA and AONs increase drug delivery to cancer

cells and limit the exposure of toxic drugs to noncancerous cells. However, micellar drug delivery

vehicles have limitations in stability. As well, endosomal escape must be considered when developing a

drug carrier. If modifications are made to increase the stability of siRNA and the polymer delivery vehicle

permits endosomal escape, then siRNA has the potential to increase the effectiveness of the drug delivery

system.

An overview of the plan is shown in Figure 1.4. The first objective is to synthesize and characterize the

nanoparticle. The second objective is in vitro testing of gene silencing activity.

Page 21: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

12

Figure 1.4: Flowchart of plan. Two objectives have been defined. The first part requires the synthesis and

characterization of a siRNA- or AON- conjugated nanoparticle. The second part tests the nanoparticle for

its activity.

Using established protocols, the poly(LA-co-TMCC) backbone is synthesized. Next, PEG is coupled to

the copolymer to produce poly(LA-co-TMCC)-g-PEG-X (X = azide or furan functional groups). Dual-

functional nanoparticles are formed using the dialysis method. The nanoparticles can be characterized for

particle size and polydispersity. Sequential click reactions to the furan and azide functionalities are

completed with trastuzumab-SMCC and oligonucleotide-alkyne moieties. Figure 1.5 displays the

schematic of the proposed nanoparticle.

Figure 1.5: Poly(LA-co-TMCC)-g-PEG nanoparticles coupled with siRNA or AONs. The hydrophilic

PEG regions will interact with the aqueous environment. SiRNA or AONs and trastuzumab antibodies

will attach to the PEG chains on the exterior of the nanoparticle.

The Damha group at McGill University has modified siRNA and AONs for increased stability.

Additionally, the group has added an acetylene group and fluorescent dye to their oligonucleotides. There

SiRNA or AON

PEG

Poly(LA-co-TMCC)

Syn

thes

is &

Ch

arac

teri

zati

on

In

Vit

ro T

est

s

Poly(LA-co-TMCC)-g-PEG Can be modified with

Herceptin to target breast cancer cells

SiRNA/AON Target model enzyme

luciferase

Nanoparticle Cell targeting ability (HER2) Gene silencing ability (luc)

Cell Assay Gene knockdown

Trastuzumab

Page 22: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

13

is evidence that the use of CuAAC chemistry will allow the azide group on the nanoparticles to react with

the acetylene group on the oligonucleotides. Thus, click chemistry will use the acetylene group to attach

the siRNA to the nanoparticles. The coupling efficiency of siRNA or AONs to the nanoparticle can be

evaluated using fluorescence. The oligonucleotides have a Cy5 dye that will allow quantification of

siRNA or AONs.

The sequential double click reaction was achieved with model compounds of Alexa 488-

dibenzylcyclooctyne and Alexa 647-SMCC. The coupling efficiency was measured by fluorescence.

Later, dual functional nanoparticles were prepared with trastuzumab and FLAG peptides to monitor cell

uptake.

The second part of the project involves quantifying the selective gene knockdown of the siRNA or AON

immuno-nanoparticles to cells. An oligonucleotide sequence that targets firefly luciferase is coupled to

the nanoparticle. Selectivity of the nanoparticles can be evaluated by introducing the siRNA-modified

nanoparticles to cells expressing firefly luciferase. Gene knockdown decreases the bioluminescence from

cells. Nanoparticles prepared with scrambled sequences are used as negative controls. Meanwhile,

delivery using a common transfection reagent, Lipofectamine, is used as a positive control.

1.8 References

[1] P. Couvreur and C. Vauthier, "Nanotechnology: Intelligent design to treat complex disease,"

Pharmaceutical Research, vol. 23, pp. 1417-1450, Jul 2006.

[2] R. Duncan, "The dawning era of polymer therapeutics," Nature Reviews Drug Discovery, vol. 2,

pp. 347-360, May 2003.

[3] K. Kataoka, et al., "Block-Copolymer Micelles as Vehicles for Drug Delivery," Journal of

Controlled Release, vol. 24, pp. 119-132, May 1 1993.

[4] Y. Matsumura and H. Maeda, "A New Concept for Macromolecular Therapeutics in Cancer-

Chemotherapy - Mechanism of Tumoritropic Accumulation of Proteins and the Antitumor Agent

Smancs," Cancer Research, vol. 46, pp. 6387-6392, Dec 1986.

[5] M. Egeblad, et al., "Tumors as Organs: Complex Tissues that Interface with the Entire

Organism," Developmental Cell, vol. 18, pp. 884-901, Jun 15 2010.

[6] S. C. Owen, et al., "Polymeric micelle stability," Nano Today, vol. 7, pp. 53-65, Feb 2012.

[7] D. E. Owens and N. A. Peppas, "Opsonization, biodistribution, and pharmacokinetics of

polymeric nanoparticles," International Journal of Pharmaceutics, vol. 307, pp. 93-102, Jan 3

2006.

[8] J. Lu and M. S. Shoichet, "Self-Assembled Polymeric Nanoparticles of Organocatalytic

Copolymerizated D,L-Lactide and 2-Methyl 2-Carboxytrimethylene Carbonate,"

Macromolecules, vol. 43, pp. 4943-4953, Jun 8 2010.

[9] M. Shi and M. S. Shoichet, "Furan-functionalized co-polymers for targeted drug delivery:

characterization, self-assembly and drug encapsulation," Journal of Biomaterials Science-

Polymer Edition, vol. 19, pp. 1143-1157, 2008.

Page 23: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

14

[10] M. Shi, et al., "Immuno-polymeric nanoparticles by Diels-Alder chemistry," Angewandte

Chemie-International Edition, vol. 46, pp. 6126-6131, 2007.

[11] Y. Yamamoto, et al., "Surface charge modulation of poly(ethylene glycol)-poly(D, L-lactide)

block copolymer micelles: conjugation of charged peptides," Colloids and Surfaces B-

Biointerfaces, vol. 16, pp. 135-146, Nov 1999.

[12] T. V. Achenbach, et al., "Oligonucleotide-based knockdown technologies: Antisense versus RNA

interference," Chembiochem, vol. 4, pp. 928-935, Oct 6 2003.

[13] P. Stougaard, et al., "Rnas Involved in Copy-Number Control and Incompatibility of Plasmid

R1," Proceedings of the National Academy of Sciences of the United States of America-Biological

Sciences, vol. 78, pp. 6008-6012, 1981.

[14] J. Tomizawa, et al., "Inhibition of Cole1 Rna Primer Formation by a Plasmid-Specified Small

Rna," Proceedings of the National Academy of Sciences of the United States of America-

Biological Sciences, vol. 78, pp. 1421-1425, 1981.

[15] S. Brantl, "Antisense-RNA regulation and RNA interference," Biochimica Et Biophysica Acta-

Gene Structure and Expression, vol. 1575, pp. 15-25, May 3 2002.

[16] A. Fire, et al., "Potent and specific genetic interference by double-stranded RNA in

Caenorhabditis elegans," Nature, vol. 391, pp. 806-811, Feb 19 1998.

[17] S. M. Elbashir, et al., "Duplexes of 21-nucleotide RNAs mediate RNA interference in cultured

mammalian cells," Nature, vol. 411, pp. 494-498, May 24 2001.

[18] J. K. Watts, et al., "Chemically modified siRNA: tools and applications," Drug Discovery Today,

vol. 13, pp. 842-855, Oct 2008.

[19] W. J. Kim and S. W. Kim, "Efficient siRNA Delivery with Non-viral Polymeric Vehicles,"

Pharmaceutical Research, vol. 26, pp. 657-666, Mar 2009.

[20] S. W. Choi, et al., "Surface modification of functional nanoparticles for controlled drug delivery,"

Journal of Dispersion Science and Technology, vol. 24, pp. 475-487, 2003 2003.

[21] D. Reischl and A. Zimmer, "Drug delivery of siRNA therapeutics: potentials and limits of

nanosystems," Nanomedicine-Nanotechnology Biology and Medicine, vol. 5, pp. 8-20, Mar 2009.

[22] A. Kalota, et al., "2 '-Deoxy-2 '-fluoro-beta-D-arabinonucleic acid (2 ' F-ANA) modified

oligonucleotides (ON) effect highly efficient, and persistent, gene silencing," Nucleic Acids

Research, vol. 34, pp. 451-461, 2006.

[23] M. Oba, et al., "Polyplex Micelles with Cyclic RGD Peptide Ligands and Disulfide Cross-Links

Directing to the Enhanced Transfection via Controlled Intracellular Trafficking," Molecular

Pharmaceutics, vol. 5, pp. 1080-1092, Nov-Dec 2008.

[24] C. B. Hansen, et al., "Attachment of antibodies to sterically stabilized liposomes: Evaluation,

comparison and optimization of coupling procedures," Biochimica Et Biophysica Acta-

Biomembranes, vol. 1239, pp. 133-144, Nov 1995.

[25] G. F. Deleavey and M. J. Damha, "Designing Chemically Modified Oligonucleotides for

Targeted Gene Silencing," Chemistry & Biology, 2012.

[26] T. Dowler, et al., "Improvements in siRNA properties mediated by 2 '-deoxy-2 '-fluoro-beta-D-

arabinonucleic acid (FANA)," Nucleic Acids Research, vol. 34, pp. 1669-1675, 2006.

[27] G. F. Deleavey, et al., "Synergistic effects between analogs of DNA and RNA improve the

potency of siRNA-mediated gene silencing," Nucleic Acids Research, vol. 38, pp. 4547-4557, Jul

2010.

[28] A. E. Felber, et al., "siRNA nanocarriers based on methacrylic acid copolymers," Journal of

Controlled Release, vol. 152, pp. 159-167, May 2011.

[29] K. L. Min, et al., "Oligonucleotides comprised of alternating 2 '-deoxy-2 '-fluoro-beta-D-

arabinonucleosides and D-2 '-deoxyribonucleosides (2 ' F-ANA/DNA 'altimers') induce efficient

RNA cleavage mediated by RNase H," Bioorganic & Medicinal Chemistry Letters, vol. 12, pp.

2651-2654, Sep 2002.

Page 24: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

15

[30] J. K. Watts and M. J. Damha, "2 ' F-arabinonucleic acids (2 ' F-ANA) - History, properties, and

new frontiers," Canadian Journal of Chemistry-Revue Canadienne De Chimie, vol. 86, pp. 641-

656, Jul 2008.

[31] B. Ozpolat, et al., "Nanomedicine based approaches for the delivery of siRNA in cancer,"

Journal of Internal Medicine, vol. 267, pp. 44-53, Jan 2010.

[32] C. L. Zhang, et al., "siRNA-containing liposomes modified with polyarginine effectively silence

the targeted gene," Journal of Controlled Release, vol. 112, pp. 229-239, May 15 2006.

[33] C. N. Landen, et al., "Therapeutic EphA2 gene targeting in vivo using neutral liposomal small

interfering RNA delivery," Cancer Research, vol. 65, pp. 6910-6918, Aug 1 2005.

[34] A. Sharma and U. S. Sharma, "Liposomes in drug delivery: progress and limitations,"

International Journal of Pharmaceutics, vol. 154, pp. 123-140, Aug 26 1997.

[35] P. X. Guo, et al., "Engineering RNA for Targeted siRNA Delivery and Medical Application,"

Advanced Drug Delivery Reviews, vol. 62, pp. 650-666, Apr 30 2010.

[36] S. Spagnou, et al., "Lipidic carriers of siRNA: Differences in the formulation, cellular uptake, and

delivery with plasmid DNA," Biochemistry, vol. 43, pp. 13348-13356, Oct 26 2004.

[37] M. Oishi, et al., "Lactosylated poly(ethylene glycol)-siRNA conjugate through acid-labile ss-

thiopropionate linkage to construct pH-sensitive polyion complex micelles achieving enhanced

gene silencing in hepatoma cells," Journal of the American Chemical Society, vol. 127, pp. 1624-

1625, Feb 16 2005.

[38] J. M. Harris and R. B. Chess, "Effect of pegylation on pharmaceuticals," Nature Reviews Drug

Discovery, vol. 2, pp. 214-221, Mar 2003.

[39] F. Y. Xie, et al., "Harnessing in vivo siRNA delivery for drug discovery and therapeutic

development," Drug Discovery Today, vol. 11, pp. 67-73, Jan 2006.

[40] V. P. Torchilin, "Multifunctional nanocarriers," Advanced Drug Delivery Reviews, vol. 58, pp.

1532-1555, Dec 1 2006.

[41] L. Nobs, et al., "Current methods for attaching targeting ligands to liposomes and nanoparticles,"

Journal of Pharmaceutical Sciences, vol. 93, pp. 1980-1992, Aug 2004.

[42] V. P. Torchilin, "Micellar Nanocarriers: Pharmaceutical Perspectives," Pharmaceutical Research,

vol. 24, p. 16, 2006.

[43] M. Shi, et al., "Doxorubicin-Conjugated lmmuno-Nanoparticles for Intracellular Anticancer Drug

Delivery," Advanced Functional Materials, vol. 19, pp. 1689-1696, Jun 9 2009.

[44] A. K. Salem, et al., "Synthesis and characterisation of a degradable poly(lactic acid)-

poly(ethylene glycol) copolymer with biotinylated end groups," Biomacromolecules, vol. 2, pp.

575-580, Sum 2001.

[45] H. C. Kolb, et al., "Click chemistry: Diverse chemical function from a few good reactions,"

Angewandte Chemie-International Edition, vol. 40, pp. 2004-+, 2001.

[46] D. C. Rideout and R. Breslow, "HYDROPHOBIC ACCELERATION OF DIELS-ALDER

REACTIONS," Journal of the American Chemical Society, vol. 102, pp. 7816-7817, 1980.

[47] T. M. Allen, "Ligand-targeted therapeutics in anticancer therapy," Nature Reviews Cancer, vol. 2,

pp. 750-763, Oct 2002.

[48] R. Tona and R. Haner, "Synthesis and bioconjugation of diene-modified oligonucleotides,"

Bioconjugate Chemistry, vol. 16, pp. 837-842, Jul-Aug 2005.

[49] M. Meldal and C. W. Tornoe, "Cu-catalyzed azide-alkyne cycloaddition," Chemical Reviews, vol.

108, pp. 2952-3015, Aug 2008.

[50] M. Manoharan, et al., "Non-Nucleoside Building Blocks for Copper-Assisted and Copper-Free

Click Chemistry for the Efficient Synthesis of RNA Conjugates," Organic Letters, vol. 12, pp.

5410-5413, Dec 3 2010.

[51] C. J. Burrows and J. G. Muller, "Oxidative nucleobase modifications leading to strand scission,"

Chemical Reviews, vol. 98, pp. 1109-1151, May 1998.

[52] G. J. Boons, et al., "Surface Modification of Polymeric Micelles by Strain-Promoted Alkyne-

Azide Cycloadditions," Chemistry-a European Journal, vol. 16, pp. 13360-13366, 2010.

Page 25: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

16

[53] J. Zhou, et al., "Octa-functional PLGA nanoparticles for targeted and efficient siRNA delivery to

tumors," Biomaterials, vol. 33, pp. 583-91, Jan 2012.

[54] M. Lewin, et al., "Tat peptide-derivatized magnetic nanoparticles allow in vivo tracking and

recovery of progenitor cells," Nature Biotechnology, vol. 18, pp. 410-414, Apr 2000.

[55] H. Durmaz, et al., "Double click reaction strategies for polymer conjugation and post-

functionalization of polymers," Polymer Chemistry, vol. 3, pp. 825-835, 2012.

[56] H. Durmaz, et al., "One-pot synthesis of ABC type triblock copolymers via in situ click [3+2] and

Diels-Alder [4+2] reactions," Macromolecules, vol. 40, pp. 191-198, Jan 23 2007.

[57] A. Dag, et al., "Heterograft Copolymers via Double Click Reactions Using One-Pot Technique,"

Journal of Polymer Science Part a-Polymer Chemistry, vol. 46, pp. 6969-6977, Oct 15 2008.

[58] A. Dag, et al., "Block-Brush Copolymers via ROMP and Sequential Double Click Reaction

Strategy," Journal of Polymer Science Part a-Polymer Chemistry, vol. 49, pp. 886-892, Feb 15

2011.

[59] H. Durmaz, et al., "One-Pot Synthesis of Star-Block Copolymers Using Double Click Reactions,"

Journal of Polymer Science Part a-Polymer Chemistry, vol. 46, pp. 7091-7100, Nov 1 2008.

[60] A. Sharma, et al., "Facile Construction of Multifunctional Nanocarriers Using Sequential Click

Chemistry for Applications in Biology," Macromolecules, vol. 44, pp. 521-529, Feb 8 2011.

[61] A. Vieyres, et al., "Combined Cu-I-catalysed alkyne-azide cycloaddition and furan-maleimide

Diels-Alder "click" chemistry approach to thermoresponsive dendrimers," Chemical

Communications, vol. 46, pp. 1875-1877, 2010 2010.

[62] H. Durmaz, et al., "Cyclic Homo and Block Copolymers Through Sequential Double Click

Reactions," Journal of Polymer Science Part a-Polymer Chemistry, vol. 48, pp. 5083-5091, Nov

15 2010.

[63] U. S. Gunay, et al., "3-miktoarm star terpolymers using triple click reactions: Diels-Alder,

copper-catalyzed azide-alkyne cycloaddition, and nitroxide radical coupling reactions," Journal

of Polymer Science Part a-Polymer Chemistry, vol. 50, pp. 729-735, Feb 2012.

[64] J. Panyam, et al., "Rapid endo-lysosomal escape of poly(DL-lactide-co-glycolide) nanoparticles:

implications for drug and gene delivery," Faseb Journal, vol. 16, pp. -, Aug 2002.

[65] X. Y. Yuan, X. D., et al., "Recent advances of siRNA delivery by nanoparticles," Expert Opinion

on Drug Delivery, vol. 8, pp. 521-536, Apr 2011.

[66] X. H. Zhou and L. Huang, "DNA Transfection Mediated by Cationic Liposomes Containing

Lipopolylysine - Characterization and Mechanism of Action," Biochimica Et Biophysica Acta-

Biomembranes, vol. 1189, pp. 195-203, Jan 19 1994.

[67] B. Dalby, et al., "Advanced transfection with Lipofectamine 2000 reagent: primary neurons,

siRNA, and high-throughput applications," Methods, vol. 33, pp. 95-103, Jun 2004.

Page 26: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

17

2. Double click: Dual functionalized polymeric micelles*

*This chapter is comprised of a paper submitted to Bioconjugate Chemistry.

Dianna P.Y. Chan, Shawn C. Owen, and Molly S. Shoichet, “Double click: Dual functionalized polymeric

micelles,” submitted for publication.

Chan and Owen are co-first authors. Owen synthesized the FLAG peptide and stained the cells for confocal

microscopy. Chan completed all other experiments outlined in this chapter. The discussion was written by Chan

with input and editing by all other authors.

2.1 Abstract

Polymeric nanoparticle micelles provide a possible platform for theranostic delivery, combining

role of therapeutics and diagnostics in one vehicle. To explore dual-functional micelles, the

amphiphilic copolymer of poly(ᴅ,ʟ-lactide-co-2-methyl-2-carboxytrimethylene carbonate)-graft-

poly(ethylene glycol)-X (P(LA-co-TMCC)-g-PEG-X) was self-assembled to form micelles, with

X representing either azide or furan. Micelles of P(LA-co-TMCC)-g-PEG-azide and P(LA-co-

TMCC)-g-PEG-furan terminal functional groups were used to conjugate dibenzylcyclooctyne

and maleimide-modified probes, respectively, taking advantage of orthogonal coupling

chemistry. To verify the utility of the dual-functional micelles, trastuzumab-maleimide

antibodies and FLAG- dibenzylcyclooctyne peptides were covalently bound by sequential click

chemistry reactions. SKOV-3luc cells that were treated with the dual-functionalized micelles

showed co-localization of the antibodies and peptides by confocal imaging, demonstrating the

promise of this approach.

2.2 Introduction

Biodegradable copolymers that self assemble into nanoparticle micelles are promising drug

delivery vehicles due to their tuneable physical properties [1]. Polymeric micelles can be tailored

to improve the solubility of hydrophobic drugs, control the release rate of drugs, reduce the

elimination rate associated with the host immune system, increase the circulation time and

enhance the preferential delivery of drugs to solid tumors [2]. Polymeric micelles are unique

drug delivery vehicles because they remain intact at concentrations above the critical micelle

concentration (CMC) and dissociate slowly when below the CMC to release encapsulated drugs

[2].

Page 27: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

18

Polymeric micelles can capitalize on their unique amphiphilic properties by loading hydrophobic

drugs into the core to improve drug solubility while incorporating targeting and/or imaging

ligands on the surface as well. For example, micelles can be conjugated to antibodies, peptides,

lectins, saccharides or hormones[3] for targeted drug delivery. Targeting cell surface receptors

using antibodies is a promising method to improve the specific binding of micelles to desired

cells [4]; however, intracellular trafficking may be required if the target is within the nucleus or

organelles. Cell penetrating peptides and fusogenic proteins facilitate the targeting to

intracellular locations and can be added to the drug carrier system [5, 6]. The co-delivery of

additional therapeutics can also improve the overall efficacy [7]. Likewise, micelles that are

labelled with a molecular probe or contrast agent can aid to diagnose disease state or track the

kinetics of the drug delivery system [8]. A recent shift towards multi-functional “theranostic”

treatments aim to use the polymeric micelle as a drug carrier with diagnostic capacity, thereby

serving a dual purpose.

To covalently attach multiple, unique diagnostic agents, targeting ligands or therapeutic agents,

the micelle must have multiple, accessible functional groups [2]. A mixed micelle system

comprised of a blend of block copolymers could produce multi-functional micelles. Mixed

micellar systems are formed by weak hydrophobic interactions, hydrogen bonding,

stereocomplexation, ionic interaction or chemical crosslinking [1]. In one system of self-

assembled micelles, a graft copolymer was combined with a diblock copolymer to conjugate two

moieties, Cy5.5 and folate, with the size and polydispersity dependent on the proportion of each

polymer[9].

Micelles have been surface-functionalized with a single type of functional group, using either

Diels Alder or Huisgen 1,3-cycloaddition click chemistries [4, 10], but never both. While the

most common click reaction is the copper-catalyzed alkyne-azide (CuAAC) reaction [11], the

copper-free reaction using a strain-promoted alkyne-azide cycloaddition (SPAAC) reaction is

gaining popularity as it overcomes the potential cytotoxicity associated with copper ion

contamination[12]. The Diels Alder reaction similarly avoids potential metal ion contamination

and is a clean and simple click reaction that is effective in aqueous conditions.

Page 28: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

19

Poly(ᴅ,ʟ-lactide-co-2-methyl-2-carboxytrimethylene carbonate)-graft-poly(ethylene glycol)

(P(LA-co-TMCC)-g-PEG) is an amphiphilic polymer that self-assembles to form stable micelles

with the capacity to encapsulate hydrophobic drugs and to be covalently modified with targeting

ligands[13-15]. Here we show that a P(LA-co-TMCC)-g-PEG-X copolymer, where X represents

one of two click-able functional groups (furan or azide), can self assemble into micelles and be

functionalized with two distinct biomolecules (maleimide-trastuzumab and dibenzylcyclooctyne-

FLAG peptide), thereby achieving dual functionalized micelles through click chemistry. In

contrast to mixed micelles formed from two dissimilar block copolymers, the use of a single

copolymer with two functional groups produces a stable micelle that retains the characteristics of

a single population. We take advantage of the Diels Alder reaction between furan and maleimide

and the copper-free reaction between azide and dibenzylcyclooctyne on pre-formed micelles in

aqueous conditions. The strained alkyne bond of dibenzylcyclooctyne has increased

electrophilicity over simple alkynes, allowing the reaction to proceed without a catalyst[16]. The

versatility of binding different moieties with click chemistry reactions provides a platform for the

incorporation of unique combinations of ligands with potential use in therapeutic and diagnostic

applications or in primary/secondary targeting conditions. The ability to conjugate biomolecules

on the surface of pre-formed micelles in aqueous conditions is particularly important for ligands

that are sensitive to the organic solvents used during micellization, for targeting ligands that

require direct binding to cells, or for MRI contrast agents that have enhanced contrast properties

when exposed to aqueous environments[17].

2.3 Materials and Methods

Synthesis of 5-methyl-5-benzyloxycarbonyl-1,3-trimethylene carbonate (benzyl-protected

TMCC, TMCC-Bn) was carried out as previously reported[4]. 3,6-dimethyl-1,4-dioxane-2,5-

dione and 1-[3,5-bis(trifluoromethyl)phenyl]-3-[(1R,2R)-(–)-2(dimethylamino) cyclohexyl]

thiourea (Strem Chemicals, Newburyport, MA) were used as received to synthesize P(LA-co-

TMCC)[18]. Boc-NH-PEG(10K)-NHS (Rapp Polymere, Tübingen, Germany) was modified

using previously reported protocols for furan and azide-functional groups[4, 10]. N,N’-

diisopropyl carbodiimide, N,N’-diisopropylethylamine and hydroxybenzotriazole (TRC, Toronto,

ON) were used as received. Amino acid derivatives were purchased from Novabiochem

Page 29: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

20

(Billerica, MA). Anti-FLAG (Rabbit) antibodies were purchased from Cell Sorting (Boston,

MA), anti-human IgG (Rat) FITC from Sigma-Aldrich (St. Louis, MO), and anti-Rabbit (Goat)

Alexa-647 from Invitrogen (Grand Island, NY). All solvents and reagents were purchased from

Sigma-Aldrich and were used as received, unless otherwise noted.

The Alexa 488 C5 maleimide was obtained from Invitrogen (Burlington, ON), the Alexa 488-

dibenzylecylooctyne (DBCO) was purchased from Click Chemistry Tools (Scottsdale, AZ) and

sulfosuccinimidyl 4-(N-maleimidomethyl)cyclohexane-1-carboxylate (sulfo-SMCC) was

purchased from Pierce (Rockford, IL). Trastuzumab was purchased through Hoffmann-La Roche

Limited (Mississauga, ON). 5-((2-(and-3)-S-(acetylmercapto)succinoyl)amino) (SAMSA)

fluorescein (Invitrogen, Eugene, OR) was used to quantify the amount of SMCC that coupled to

the trastuzumab. The human ovary cancer cell line SKOV-3luc was purchased from Cell

Biolabs, Inc (San Diego, CA).

The 1H NMR spectra were recorded at 400 MHz at room temperature using a Varian Mercury

400 spectrometer. The chemical shifts are in ppm. The molecular weights and polydispersity of

P(LA-co-TMCC) were measured by gel permeation chromatography (GPC) in THF relative to

polystyrene standards at room temperatures on a system with two-column sets (Viscotek

GMHHR-H) and a triple detector array (TDA302) at a flow rate of 0.6 mL/min. Polymeric

micelle size and distribution were measured by dynamic light scattering (DLS) using the

Zetasizer Nano ZS system (Malvern, UK). The NH2-PEG-azide was characterized using the

Perkin-Elmer FT-IR Spectrum 1000. The Sepharose CL-4B column was prepared by soaking the

beads in distilled water overnight and packing the beads in a column (5 x 15 cm). The column

was washed with distilled water for 1 h before use and the flow rate was determined by gravity.

The Sephadex G-25 column was prepared using the same method, except PBS buffer (1x, pH

7.4) was used as the eluent and the beads were packed in a column (1.5 x 15 cm). FPLC of the

micelles used a Superdex 200 column. The column was washed with distilled water for 20 min

and PBS buffer (1x, pH 7.4) for 20 min at a flow rate of 1.5 mL/min before use. Fluorescence

measurements were completed with the Tecan Infinite M200 Pro fluorescent plate reader and

absorbance was quantified using the NanoDrop Spectrophotometer.

Page 30: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

21

2.3.1 Polymer Synthesis

Synthesis of NH2-PEG-azide. As previously shown, bifunctional tert-butyloxycarbonyl-

protected amine-PEG activated acid (BocNH-PEG-(N-hydroxysuccinimide)), 0.523 g, 52 μmol)

was modified to incorporate a terminal azide group[10] by reacting with 11-azido-3,6,9-

trioxaundecan-1-amine (Sigma Aldrich, 1.0 mL, 5 mmol) in DMF (Figure 2.1). The final product

yielded NH2-PEG-N3 (0.3629 g, 80.6%). The presence of the terminal azide group (2101 cm-1

),

terminal amine (3389 cm-1

), C–H alkane (2888 cm-1

), C=O amide (1692 cm-1

), and C–O ether

(1101 cm-1

) was characterized by FTIR (Appendix Figure A 1).

Figure 2.1. Synthesis of (a) NH2-PEG-azide and (b) NH2-PEG-furan by reacting NHS-activated PEG

chains with furfurylamine or amine-terminated triethylene glycol-azide, respectively, followed by BOC

deprotection.

Synthesis of NH2-PEG-furan. Similar to the procedure used to synthesize PEG-azide, BocNH-

PEG-NHS (1.00 g, 0.1mmol) and furfurylamine (1.00 mL, 11.2 mmol) were reacted to yield

bocNH-PEG-furan (0.8021 g, 80.2% yield). 1H NMR (CDCl3, 400 MHz): δ 1.44 (s, Boc CH3),

2.32 (t, J=14 Hz , –CH2–), 3.61 (m, –OCH2CH2–) , 4.42 (s, –CH2-furan), 6.22 (d, J= 3Hz, furan

CH-3), 6.30 (dd, J=5 Hz, furan CH-4) and 7.34 (d, J=3 Hz, furan CH-5) . For NMR spectra, see

Page 31: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

22

Appendix Figure A 2. The product was deprotected with treatment of 15% trifluoroacetic acid in

CH2Cl2 for a final yield of NH2-PEG-N3 (0.6614 g, 81.5%).

Grafting of PEG to P(LA-co-TMCC). Synthesis of P(LA-co-TMCC) by ring opening

polymerization has previously been shown [18]. 1H NMR (CDCl3, 400MHz): δ 1.22 (br s, CH3

from TMCC), 1.59 (m, CH3 from LA), 4.31 (br s, CH2 from TMCC), 5.15 (m, CH from LA),

7.33 (m, benzyl Ar) and 8.02 (m, pyrene). Absence of the benzyl group peaks in the 1H NMR

after the hydrogenolysis of P(LA-co-TMCC-Bn) showed full deprotection of the polymer

backbone. See Appendix Figure A 3 for 1H NMR spectra.

The modified H2N-PEG (PEG-X) was grafted onto the P(LA-co-TMCC) as previously shown

(Figure 2.2) [18]. P(LA-co-TMCC) (100 mg, 8.3 μmol) was dissolved in 3mL of DMF with N,N’-

diisopropyl carbodiimide (DIC, 100 μL, 0.65 mmol), hydroxybenzotriazole (HOBt, 63.1mg, 0.48

mmol) and N,N’-diisopropylethylamine (DIPEA, 0.52 mmol) and stirred for 30 min. The PEG-

X (100 mg, 10 μmol) was dissolved in 3 mL of DMF and added to the P(LA-co-TMCC) solution

dropwise. An additional 90 μL of DIPEA was added, the mixture was sealed under argon for 2

min and left to react overnight.

Borate buffer (400 μL, 500 mM, pH 9.0) was added to the P(LA-co-TMCC)-g-PEG-X solution

before dialyzing against distilled water with a 2 kDa RC membrane. The P(LA-co-TMCC)-g-

PEG-X was purified using the Sepharose CL-4B column, then freeze dried (37.5 mg, 38%).

From the 1H NMR spectra, it was calculated that there was an average of 0.9 PEG-azide chains

per P(LA-co-TMCC) backbone. 1H NMR (CDCl3, 400 MHz): δ 1.25 (br s, CH3 from TMCC),

1.57 (m, CH3 from LA), 3.61-3.64 (m, –OCH2CH2–), 4.35 (m, CH2 from TMCC) and 5.17 (m,

CH from LA). See Appendix Figure A 4 for 1H NMR spectra.

The same procedure was carried out to graft PEG-furan to P(LA-co-TMCC). An average of 0.7

PEG-furan chains per backbone was calculated using the 1H NMR spectra.

1H NMR (CDCl3, 400

MHz): δ 1.22 (br s, CH3 from TMCC), 1.56 (m, CH3 from LA), 3.59-3.69 (m, –OCH2CH2–),

4.31 (m, CH2 from TMCC) and 5.15 (m, CH from LA). See Appendix Figure A 4 for 1H NMR

spectra.

Page 32: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

23

Figure 2.2. Synthesis of P(LA-co-TMCC)-g-PEG-X (X=azide or furan) by grafting NH2-PEG-X with

DIC chemistry.

2.3.2 Micellization

The micelles were prepared by co-self-assembly of P(LA co-TMCC)-g-PEG-furan and P(LA-co-

TMCC)-g-PEG-azide by membrane dialysis, as previously shown for single functionalized

chains [19]. To form the dual-functional micelles, P(LA-co-TMCC)-g-PEG-furan (2.2 mg, 18.2

kDa) and P(LA-co-TMCC)-g-PEG-azide (1.8 mg, 20.2 kDa) were combined for an average of 80

nmol each of furan and azide functional groups. The average number of furan and azide

functional groups per micelle accounted for the difference in grafting efficiency (0.7 PEG and

0.9 PEG per backbone, respectively) by adjusting the ratio of each polymer dissolved in 1mL of

DMF. Borate buffer (50 μL, 0.5 M, pH 9.0) was added dropwise to the solution and an additional

500 μL of distilled water was added to form the micelles. The solution was dialysed against

Page 33: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

24

distilled water using a dialysis membrane with MWCO of 2 kDa at room temperature overnight

to form micelles. The water was changed every 2 h for the first 6 h. The micelles were filtered

using a 0.45 μm nylon filter and brought to a concentration of 50 μM of polymer before

characterization by DLS.

Azide-only and furan-only micelles were prepared using the same dialysis method as controls for

the coupling chemistry. P(LA-co-TMCC)-g-PEG-azide (4.0 mg, 20.2 kDa) was used for the

azide micelles and P(LA-co-TMCC)-g-PEG-furan (4.0 mg, 18.2 kDa) was used for the furan

micelles. The average diameter was measured by DLS of the furan micelles at 66.43 nm with

PDI=0.218; the average diameter of the azide micelles was measured at 102.9 nm with PDI=

0.165.

2.3.3 Conjugation of Trastuzumab and FLAG Peptides to the Nanoparticles

Synthesis of Trastuzumab-SMCC. Trastuzumab (6.6 mg, 45.5 nmol) was dialyzed overnight (2

kDa MWCO) against Tris buffer (100 mM, pH 8.5) to remove the storage solution of sodium

azide. Sulfo-SMCC (0.5 mg, 1.15 µmol) was added to the trastuzumab solution and left to react

for 4 h. The SAMSA fluorescein (10 mg/mL in DMSO) was dissolved in 500 µL of NaOH (0.1

M) and incubated for 15 min at room temperature. The solution was neutralized using 7 µL of

HCl (6 M) and buffered with 100 µL Na2HPO4 (0.5 M). From this solution, the SAMSA

fluorescein (0.55 µmol, 63 µL) was mixed with trastuzumab-SMCC (55 nmol, 100 µL) and

incubated at room temperature for 30 min. The mixture was dialyzed against Tris buffer

overnight. A standard curve was prepared using the activated SAMSA fluorescein solution and

was used to quantify the trastuzumab-SMCC at an excitation wavelength of 480 nm and

emission at 530 nm. An average of 1 fluorescein molecule per antibody translated to 1 SMCC

group available per antibody.

Synthesis of dibenzylcyclooctyne-modified FLAG peptide. The FLAG peptide

(DYKDDDDK) was synthesized by standard Fmoc-based solid phase peptide chemistry on a

CEM Liberty 1 microwave peptide synthesizer. The resin-bound FLAG peptide (100 mg, 1.1

meq/g, 0.11 mmol) was swollen in CH2Cl2 and then rinsed several times with N,N-

Page 34: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

25

diisopropylethylamine (DIPEA). Dibenzylcyclooctyne-NHS (80 mg, 0.165 mmol) was dissolved

in DMF (~2 mL), added to the resin, and allowed to react overnight. The resin was washed

several times with DMF and CH2Cl2. The product was deprotected and cleaved from the resin

with treatment of 95% trifluoroacetic acid in CH2Cl2. Solvent was removed from the resulting

product and peptide precipitated in cold Et2O. Product was purified by HPLC and analyzed by

electrospray ionization mass spectrometry (ESI+ MS). Expected 1327.5, found 1329.6 (M+2H).

Coupling of Alexa 488-maleimide (MI) on Furan Micelles. P(LA-co-TMCC)-g-PEG-furan

micelles (2 mg/mL, 50 nmol) were reacted for 24 h with Alexa 488-C5-MI (2 mg/mL, 100 nmol)

in MES buffer (100 mM, pH 5.5, 100 μL). Using the G-25 Sephadex column, the unreacted

Alexa 488-MI was separated. Fractions containing the micelles were detected using Bradford

reagent and quantified by absorbance at 260 nm. Fluorescence was used to quantify the Alexa

488 that conjugated to the micelle.

Controls were prepared by quenching the Alexa 488-MI (100 nmol) with furfurylamine (10

μmol) and cysteine (10 μmol) in MES buffer (100 mM, pH 5.5, 100 μL) before the coupling

reaction. To fully quench the maleimide with the cysteine, tris(2-carboxyethyl)phosphine) (80

μmol) was added for the 48 h reaction. The same purification procedure was followed to remove

the unreacted dye.

The Diels Alder (DA) reaction was characterized by reacting NH2-PEG-furan (100 nmol) with

Alexa 647-MI (150 nmol) overnight. The solution was dialyzed to remove the unreacted Alexa

647-MI with 2 kDa MWCO dialysis tubing in distilled water. The sample was lyophilized before

forming a KBr pellet. The reacted NH2-PEG-furan was compared with an unreacted sample to

confirm the DA reaction. A decrease of the characteristic peaks of furan C=C (1466 cm-1

) and

the appearance of the C=C Diels Alder adduct peak (1459 cm-1

) indicate that the Diels Alder

reaction occurred. See Appendix Figure A 5 for FTIR spectra.

Coupling of Alexa 488-dibenzylcyclooctyne (DBCO) on Azide Micelles. P(LA-co-TMCC)-g-

PEG-azide micelles (2 mg/mL, 100 nmol) were reacted for 24 h with Alexa 488-DBCO (0.5

Page 35: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

26

mg/mL, 200 nmol). Using the G-25 Sephadex column, the unreacted Alexa 488-DBCO was

removed.

As a control, the reaction was repeated with Alexa 488-DBCO where the Alexa 488-DBCO (200

nmol) was quenched by incubation with 11-azido-3,6,9-trioxaundecan-1-amine (400 µL, 2

mmol) prior to the click chemistry reaction. The micelles were purified using the same procedure

with a size exclusion column.

To characterize the SPAAC reaction, NH2-PEG-N3 (100 nmol) was reacted with Alexa 488-

DBCO (500 nmol) overnight. Similar to the procedure outlined previously, the sample was

prepared for FTIR analysis to compare the unreacted and reacted NH2-PEG-N3. The presence of

the terminal azide group was characterized by the FTIR analysis. See Appendix Figure A 6 for

FTIR spectra.

Coupling of Alexa 488-DBCO and Alexa 647-MI on Dual-Functional Micelles by Click

Chemistry. P(LA-co-TMCC)-g-PEG micelles with azide and furan functional groups (50 μM)

were sequentially reacted with maleimide and dibenzylcyclooctyne-functional moieties. First,

Alexa 647-MI (100 nmol) was added at room temperature with MES buffer (100 mM, pH 5.5)

for 24 h. The solution was dialyzed overnight against PBS buffer (1x, pH 7.4) using 2 kDa

MWCO dialysis tubing. The buffer was changed every 2 h for the first 6 h. Then, the solution

was transferred to a glass vial to react with the Alexa 488-DBCO (100 nmol) at room

temperature for 24 h. The reacted micelles were purified using size exclusion chromatography on

a Sephadex G-25 column with PBS buffer (1x, pH 7.4) as the eluent. Fractions were collected in

1 mL volumes for a total of 20 fractions. Fluorescence of the Alexa 647 (ex. 640 nm, em. 675

nm) and Alexa 488 (ex. 490 nm, em. 525 nm) were used to quantify the conjugation efficiency.

The concentrations of the micelles were measured by absorbance at 260 nm. Controls were

repeated by quenching either the Alexa 647-MI or Alexa 488-DBCO with three orders of

magnitude excess of furfurylamine or 11-azido-3,6,9-trioxaundecan-1-amine, respectively,

before reacting with the micelles. See Appendix Figure A 7 for fluorescence and absorbance

measurements from the size exclusion column.

Page 36: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

27

Coupling of FLAG-DBCO and Trastuzumab-SMCC on Dual-Functional Micelles by Click

Chemistry. The dual-functional micelles were prepared using the same procedure as other

micelles described. As with the Alexa dyes, the trastuzumab-SMCC and FLAG-DBCO were

conjugated sequentially. Micelles were purified by FPLC (GE Healthcare AKTA Purifier)

monitoring absorbance at 215 and 280 nm.

2.3.4 Cell Culture and In Vitro Assessment of Dual Functionalized Micelle Uptake

SKOV-3luc cells were cultured in McCoy’s 5A media containing 10% FBS, 10 UI/mL

penicillin, and 10 μg/mL streptomycin under standard culture conditions (37°C, 5% CO2, 100%

humidity). Cells were seeded in 16-well chamber slides at 10,000 cells/well and allowed to

adhere for 22 h. Dual functionalized micelles having both trastuzumab and FLAG peptides (100

µL, 100 nM trastuzumab) were added. Cells were supplemented with an additional 200 μL of

media and incubated for 3 h. Cells were fixed in 4% paraformaldehyde before staining and

analysis. Cells were incubated with 1% BSA in PBS with 0.05% Tween 20 (PBST) for 30 min to

block unspecific binding of the antibodies. Cells were then incubated with the primary antibody

(anti-FLAG or anti-human IgG) diluted in 1% BSA in PBST for 1 hr at room temperature.

Secondary antibodies were incubated in the same manner. Cells were then washed several time

with PBS and mounted with medium containing DAPI. Images were collected by confocal

microscopy (Olympus FV1000) - for DAPI, excitation at 405 nm, emission at 460; for FITC,

excitation at 485 nm, emission at 520 nm; for Alexa-647, excitation at 650nm, emission at

675nm.

2.4 Results

2.4.1 Polymer Synthesis

Synthesis of P(LA-co-TMCC)-g-PEG-X. The number average molecular weight and

polydispersity of P(LA-co-TMCC) was determined by GPC to be 11.2 kDa and 1.12,

respectively. Based on 1H NMR integrated peak areas for TMCC methylene (4.35 ppm) and LA

methyl (5.15 ppm), the molar percentage of TMCC in the copolymer was determined to be 7.1%.

The copolymer composition was calculated using the 1H NMR spectra based on the integrated

Page 37: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

28

peak areas of LA methyl groups and TMCC methylene groups relative to the single pyrene

initiator group (8.02 ppm) present on each polymer chain: P(LA)130-co-(TMCC)10. These data

are consistent with the GPC measurement.

Using DIC chemistry, we grafted NH2-PEG-furan or NH2-PEG-azide onto the carboxylic acid

groups present on the TMCC segments of the copolymer. We estimated the ratio of grafted PEG

to P(LA-co-TMCC) by comparing the integrated peak areas of the PEG methylene (3.61-3.64

ppm) to the TMCC methylene (4.35 ppm). On average, there was approximately 0.7 PEG-furan

or 0.9 PEG-azide chains grafted to every P(LA-co-TMCC) backbone. The number average molar

mass of P(LA-co-TMCC)-g-PEG was calculated according to equation (1):

Mn(P(LA-co-TMCC)-g-PEG) = Mn(backbone) + (Mn(PEG) x # of PEG per copolymer backbone) (1)

Thus, the P(LA130-co-TMCC10)-g-PEG10kDa-furan had a number average molar mass of 18.2 kDa

and the P(LA130-co-TMCC10)-g-PEG10kDa-azide had a slightly larger number average molar mass

of 20.2 kDa.

2.4.2 Micellization

Estimation of the Aggregation Number of Micelles. The micelles formed from P(LA-co-

TMCC)-g-PEG-X have an average diameter of 96.2 nm and distribution is 0.166 as determined

by dynamic light scattering. The aggregation number (Nagg) was estimated for these micelles, as

previously described[10], according to equation (2), where k is a scaling factor between the

radius of the polymeric micelle, Rmicelle, and the aggregation number.

Nagg = kRmicelle2 (2)

For these micelles, with average diameter of 96.2 nm, the aggregation number is approximately

3500.

2.4.3 Conjugation of Trastuzumab and FLAG Peptides to the Nanoparticles

Coupling of Alexa 488-DBCO and Alexa 647-MI on Dual-Functional Micelles by Click

Chemistry. We first verified the coupling reactions in our system using two functionalized dye

Page 38: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

29

molecules, Alexa 647-MI and Alexa 488-DBCO, as surrogate ligands for the trastuzumab and

FLAG peptide, respectively. The NH2-PEG-furan was reacted with Alexa 647-MI to demonstrate

the DA reaction. In order to verify the specificity of the two click reactions with our polymeric

micelles, we tested multiple combinations of reactants and controls (Table 2.1). The first reaction,

(a), showed efficient binding from sequential click reactions between nanoparticle micelles and

Alexa 647-MI followed by Alexa 488-DBCO. Three controls demonstrated the selectivity of the

DA and SPAAC reactions: the functional groups of either the Alexa 647-MI, Alexa 488-DBCO,

or both, were quenched prior to the click chemical reaction with dual functionalized micelles

that had an equal number of azide and furan groups.

For one control (b), both dye molecules were quenched prior to the double click reaction. The

results confirmed that there was little non-specific binding between the dyes and micelles. When

only the DBCO was quenched (c), we observed the selective reactivity between the maleimide

moiety and available furan groups on the micelle: the average number of Alexa 647-MI groups

that bound to each micelle was similar to reaction (a). By contrast, when only the maleimide was

quenched (d), the number of Alexa 488-DBCO which bound to the micelle was greater than

reaction (a), where the available furan sites were occupied by the Alexa 647-MI, indicating that

the DBCO reacted with both azide and furan groups.

Table 2.1 Dual-functional micelle reactions with maleimide- and dibenzylcyclooctyne-functional

moieties (R1= Alexa 647, R2= Alexa 488). The average number of each moiety bound to a dual-functional

micelle with equal furan and azide groups are tabulated (n=3, mean ± standard deviation).

Average R1-MI/micelle Average R2-DBCO/micelle

(a) Micelles + R1-MI + R2-

DBCO

88 ± 11 270 ± 28

(b) Micelles + R1-MI

(quenched) + R2-DBCO

(quenched)

11 ± 7 18 ± 18

(c) Micelles + R1-MI

+ R2-DBCO (quenched)

110 ± 42 14 ± 11

(d) Micelles + R1-MI

(quenched) + R2-DBCO

18 ± 11 510 ± 7

Page 39: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

30

We confirmed that the conjugations were due to DA and SPAAC reactions with two additional

control reactions: (1) between a furan-only micelle and Alexa 647-MI; and (2) between an azide-

only micelle and Alexa 488-DBCO. The DA reaction with the furan micelle and Alexa 488-MI

showed an average of 1 dye for every 83 polymer chains (i.e. 1:83 dye/polymer). When the

Alexa 488-MI was first quenched with either furfurylamine or cysteine prior to reaction with

furan-micelles, the average number of dyes per polymer decreased significantly to 1:1200

dye/polymer chains for furfurylamine pre-quenched controls and 1:8200 dye/polymer chains for

cysteine pre-quenched controls. These demonstrate that there is little non-specific binding

between Alexa 488 and our polymeric micelles, which is consistent with previous reports of the

DA reaction between furan and maleimide groups, as confirmed by FTIR [20].

For the SPAAC reaction with the azide micelle and Alexa 488-DBCO, we detected an average of

1 dye for every 13 polymer chains (i.e 1:13 dye/polymer). When the DBCO was first quenched

with excess 11-azido-3,6,9-trioxaundecan-1-amine, the number of dyes per polymer dropped to

an average of 1:200 dye/polymer chains, demonstrating that there was little non-specific binding

between Alexa 488 and the polymeric micelles.

Coupling of FLAG-DBCO and Trastuzumab-SMCC on Dual-Functional Micelles by Click

Chemistry. To demonstrate the functional utility of a dual-labelled polymeric micelle, we

performed sequential click reactions with the SMCC-functionalized antibody (trastuzumab-

SMCC) and DBCO-functionalized peptide (FLAG-DBCO). Trastuzumab is a humanized

antibody that binds human epidermal growth factor receptor 2 (HER2). A FLAG peptide was

selected as a simple, model peptide sequence that can be recognized by an antibody. We chose

SKOV-3luc cells, a human ovarian cancer cell line that overexpresses HER2, as a target to verify

cell binding of the dual-functionalized micelles, through anti-HER2 and HER2 interactions.

After the double-click reaction described above, micelle constructs were separated from free,

unreacted ligands by size-exclusion FPLC. Micelle elution peaks were completely resolved from

unreacted material (Figure 2.3)

Page 40: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

31

Figure 2.3 FPLC purification of nanoparticles with trastuzumab and FLAG peptide. Nanoparticles

conjugated with trastuzumab and FLAG were separated from the unreacted moieties. Fractions between 8

and 12 mL were incubated with SKOV-3luc cells to observe cell uptake.

Cells were treated with fluorescently-labeled antibodies against trastuzumab/FLAG to reveal the

cellular localization of the antibody and peptide. Fluorescence images show trastuzumab and

FLAG co-localized on the cell surface and inside the cells (Figure 2.4). Cells treated with non-

functionalized micelles showed no uptake (Appendix Figure A 8), thereby demonstrating receptor

mediated cell targeting. If trastuzumab was denatured or was not accessible on the micelle

surface, no binding would occur. Likewise, positive staining for the FLAG peptide indicated that

the peptide remained intact and accessible on the micelle surface.

Nanoparticle with

Herceptin and FLAG

peptide (8-12mL)

Unreacted FLAG

peptide, Herceptin

Page 41: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

32

Figure 2.4 Confocal images of SKOV-3luc cells. (a) Cell nuclei are stained with DAPI (blue); (b)

trastuzumab is fluorescently-tagged with Alexa Fluor 488 (green); (c) FLAG peptide is fluorescently-

tagged with Alexa Fluor 647 (purple); and (d) Overlay of all channels showed cell nuclei surrounded by

trastuzumab/FLAG-tagged micelles.

2.5 Discussion

Polymeric nanoparticle micelle chemical modifications are limited to aqueous reaction

conditions, yet many of these involve cytotoxic catalysts or result in undesirable side

reactions[12]. The dual functional micelles synthesized herein provide a platform for facile

orthogonal conjugation chemistry, resulting in clean and versatile ligand modification. By

forming the micelles prior to the conjugation reaction and using aqueous-based chemistry, we are

able to preserve the native structure and activity of the ligands and micelles. Furthermore, a

single purification step after ligand bonding is beneficial for maximizing yields and maintaining

micelle stability.

Interestingly, we observed a difference in the reactivity of Diels Alder and azide-alkyne click

chemistries. When the dual functionalized micelles were reacted sequentially with MI and then

Page 42: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

33

DBCO, more DBCO conjugated to the micelle than MI. This was also observed for micelles with

a single functional group: the SPAAC reaction between the DBCO and azide functionalized

micelles was more efficient than the DA reaction between the MI and furan micelle. While

SPAAC reactions can be completed at pH 7.4, DA reactions between the furan and SMCC

moiety are most efficient at acidic pH. Hydrolysis of the maleimide group will form maleamic

acid that will not react with the furan [21].

Using either click reaction, not all functional groups on the micelle were reacted - only

approximately 7.7% of azide groups and 1.2% of furan groups reacted on average. Although the

functional groups are on the PEG termini, they are not all accessible at the micelle surface for

reaction, possibly because the terminal groups are buried within the flexible PEG chain.

Importantly, the click conjugated ligands remain active to facilitate interactions with cell surface

receptors and antibody probes.

For the double labelling to be most effective, the DA reaction had to be completed prior to the

SPAAC reaction because the DBCO was able to bind to both azide and furan whereas maleimide

only reacted with furan (see Appendix Table A1 for a summary of possible reactions).

Notwithstanding this requirement for the sequential DA and then SPAAC reactions, importantly,

both reactions were carried out in the same vessel and only one purification step was required

after modification.

The use of two reactive groups improves control of the chemical modifications and composition.

In a micelle system with multiple ligands binding to a single reactive group, the reactions are

competitive. With dual functional micelles, each ligand reacts independently with a specific

receptor, eliminating competition. In addition, multiple functionalities allow for the most

effective reaction partners to be selected, based on the properties of each ligand. Importantly, the

aqueous-based coupling chemistries used in our system result in bioactive functionalized

micelles: trastuzumab interacts with HER2 receptors expressed on SKOV-3 cells and FLAG

provides a marker of the micelle for cell trafficking. Since there is, on average, less than one

functional group available on each polymer chain, if the micelles were to dissociate, the FLAG

peptides would not co-localize with trastuzumab labeled micelles. Thus, the co-localization of

Page 43: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

34

trastuzumab and FLAG in the cells suggest that the micelles remain intact after receptor-

mediated endocytosis, which is consistent with previous reports.

Nanoparticle micelles have shown potential as drug delivery vehicles due to their tuneable

physical properties. One of the challenges with modifying micelles is controlling surface

chemistry while maintaining micelle stability [4, 22]. In our system, the aqueous-based click

reactions are completed after the micelles have formed, preserving the size of the micelles and

the bioactivity of the conjugated ligands[4]. The approach of coupling multiple ligands onto a

single micelle in a simple manner provides the versatility needed to achieve: selective cellular

and subcellular targeting, cell targeting with therapeutic delivery, or facilitated co-delivery of

therapeutic agents with diagnostic probes.

2.6 Conclusion

Using two independent, sequential click reactions, we were able to prepare dual-functional

micelles. Sequential reactions with maleimide- and dibenzylcyclooctyne- Alexa dyes allowed us

to quantify the conjugation reactions and demonstrate the specificity between the DA and

SPAAC reactions. To further demonstrate applicability of the dual-functional micelle, we

conjugated trastuzumab antibodies and FLAG peptides onto the self-assembled, polymeric

micelles by DA and SPAAC reactions, respectively. The ability to selectively bind two different

moieties onto the same micelle results in a platform for future combination strategies, such as

multi-functional micelles of therapeutic moieties, diagnostic agents, targeting ligands and cell

penetrating/trafficking peptides.

2.7 References

[1] A. B. E. Attia, et al., "Mixed micelles self-assembled from block copolymers for drug delivery,"

Current Opinion in Colloid & Interface Science, vol. 16, pp. 182-194, Jun 2011.

[2] G. S. Kwon and T. Okano, "Polymeric micelles as new drug carriers," Advanced Drug Delivery

Reviews, vol. 21, pp. 107-116, Sep 16 1996.

[3] V. P. Torchilin and A. N. Lukyanov, "Peptide and protein drug delivery to and into tumors:

challenges and solutions," Drug Discovery Today, vol. 8, pp. 259-266, Mar 15 2003.

[4] M. Shi, et al., "Immuno-polymeric nanoparticles by Diels-Alder chemistry," Angewandte

Chemie-International Edition, vol. 46, pp. 6126-6131, 2007.

Page 44: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

35

[5] M. Ogris, et al., "Melittin enables efficient vesicular escape and enhanced nuclear access of

nonviral gene delivery vectors," Journal of Biological Chemistry, vol. 276, pp. 47550-47555, Dec

14 2001.

[6] M. Lindgren, et al., "Translocation properties of novel cell penetrating transportan and penetratin

analogues," Bioconjugate Chemistry, vol. 11, pp. 619-626, Sep-Oct 2000.

[7] Y. Wang, et al., "Co-delivery of drugs and DNA from cationic core-shell nanoparticles self-

assembled from a biodegradable copolymer," Nature Materials, vol. 5, pp. 791-796, Oct 2006.

[8] S. M. Janib, et al., "Imaging and drug delivery using theranostic nanoparticles," Advanced Drug

Delivery Reviews, vol. 62, pp. 1052-1063, Aug 30 2010.

[9] H. C. Tsai, et al., "Graft and diblock copolymer multifunctional micelles for cancer

chemotherapy and imaging," Biomaterials, vol. 31, pp. 2293-2301, Mar 2010.

[10] J. Lu, et al., "Click Chemistry Functionalized Polymeric Nanoparticles Target Corneal Epithelial

Cells through RGD-Cell Surface Receptors," Bioconjugate Chemistry, vol. 20, pp. 87-94, Jan

2009.

[11] M. Meldal and C. W. Tornoe, "Cu-catalyzed azide-alkyne cycloaddition," Chemical Reviews, vol.

108, pp. 2952-3015, Aug 2008.

[12] M. Manoharan, et al., "Non-Nucleoside Building Blocks for Copper-Assisted and Copper-Free

Click Chemistry for the Efficient Synthesis of RNA Conjugates," Organic Letters, vol. 12, pp.

5410-5413, Dec 3 2010.

[13] J. Lu, et al., "Stability of Self-Assembled Polymeric Micelles in Serum," Macromolecules, vol.

44, pp. 6002-6008, Aug 9 2011.

[14] K. Ho, et al., "Tunable immunonanoparticle binding to cancer cells: thermodynamic analysis of

targeted drug delivery vehicles," Soft Matter, vol. 5, pp. 1074-1080, 2009.

[15] K. S. Ho, et al., "Amphiphilic micelles of poly(2-methyl-2-carboxytrimethylene carbonate-co-

D,L-lactide)-graft-poly(ethylene glycol) for anti-cancer drug delivery to solid tumours,"

Biomaterials, vol. 33, pp. 2223-2229, Mar 2012.

[16] G. J. Boons, et al., "Surface Modification of Polymeric Micelles by Strain-Promoted Alkyne-

Azide Cycloadditions," Chemistry-a European Journal, vol. 16, pp. 13360-13366, 2010.

[17] V. P. Torchilin, "Micellar Nanocarriers: Pharmaceutical Perspectives," Pharmaceutical Research,

vol. 24, p. 16, 2006.

[18] J. Lu and M. S. Shoichet, "Self-Assembled Polymeric Nanoparticles of Organocatalytic

Copolymerizated D,L-Lactide and 2-Methyl 2-Carboxytrimethylene Carbonate,"

Macromolecules, vol. 43, pp. 4943-4953, Jun 2010.

[19] M. Shi and M. S. Shoichet, "Furan-functionalized co-polymers for targeted drug delivery:

characterization, self-assembly and drug encapsulation," Journal of Biomaterials Science-

Polymer Edition, vol. 19, pp. 1143-1157, 2008.

[20] C. M. Nimmo, et al., "Diels-Alder Click Cross-Linked Hyaluronic Acid Hydrogels for Tissue

Engineering," Biomacromolecules, vol. 12, pp. 824-830, Mar 2011.

[21] M. Pereira and E. P. Lai, "Capillary electrophoresis for the characterization of quantum dots after

non-selective or selective bioconjugation with antibodies for immunoassay.," Journal of

Nanobiotechnology, vol. 6, p. 15, 2008.

[22] S. C. Owen, et al., "Polymeric micelle stability," Nano Today, vol. 7, pp. 53-65, Feb 2012.

Page 45: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

36

3. Delivery of SiRNA and Antisense Oligonucleotides using Dual

Functional Polymeric Immuno-nanoparticles*

*This chapter comprises a paper submitted to Angewandte Chemie International.

Dianna P.Y. Chan, Glen F. Deleavey, Shawn C. Owen, Masad J. Damha, and Molly S. Shoichet, “Delivery of

siRNA and Antisense Oligonucleotides using Dual Functional Polymeric Immuno-nanoparticles for Targeted Gene

Silencing,” submitted for publication.

Chan and Deleavey are co-first authors. Deleavey synthesized and characterized the oligonucleotides. Chan

completed all other experiments outlined in this chapter and contributed to the discussion relating to siRNA and

AON delivery using nanoparticles and Lipofectamine. The manuscript was written by Chan and Deleavey with input

from all authors.

3.1 Abstract

Efficient and targeted cellular delivery of gene silencing oligonucleotides (small interfering RNAs

(siRNAs), antisense oligonucleotides (AONs)) is a major challenge facing oligonucleotide-based

therapeutics. The majority of current delivery strategies employ either conjugated ligands or

oligonucleotide encapsulation within delivery vehicles to facilitate cellular uptake. Chemical modification

of the oligonucleotides (ONs) themselves can improve potency and duration of activity, usually as a result

of improved nuclease resistance. Here, dual functionalized polymeric immuno-nanoparticles have allowed

formation of nucleic acid-functionalized polymeric micelles enabling antibody-targeted delivery of

siRNAs or AONs for gene silencing. ONs are carried on the exterior PEG corona of the functionalized

nanoparticles, and results show that phosphorothioate (PS), 2′F-ANA, and 2′F-RNA backbone chemical

modifications improve siRNA and AON potency and duration of activity. Gene silencing potencies of

nanoparticle-delivered nucleic acids were comparable to those observed with Lipofectamine transfections.

3.2 Introduction

RNA interference (RNAi) and antisense gene silencing strategies are ON-based therapeutic approaches

that utilize small interfering RNAs (siRNAs) or antisense oligonucleotides (AONs), respectively, to

silence the expression of a target gene through sequence-specific mRNA knockdown[1-3]. Both strategies

feature excellent potency and specificity once cellular uptake can be achieved. Unfortunately, potential

nucleic acid therapeutics are disadvantaged by poor nuclease stability leading to rapid degradation, poor

unassisted cellular uptake, and unintended side-effects from “off-target” silencing[4] and

immunostimulation [5]. Many of these obstacles are addressed through the use of effective new delivery

technologies and chemical modifications to the ON backbones to prevent nuclease degradation and

unwanted side effects [1, 3, 6, 7].

Page 46: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

37

2′-Deoxy-2′-fluoroarabino nucleic acid (2′F-ANA) has proven a particularly useful chemical modification

in both siRNA and AON gene silencing applications[7-13]. In AON applications, when combined with

the phosphorothioate (PS) backbone modification [14-16], 2′F-ANA enhances nuclease stability[7, 8], is

fully compatible with RNase H-mediated cleavage[7, 17], and can improve binding stability, duration of

activity, and potency [7, 12, 13]. 2′F-ANA modified PS-AONs can also function in gymnotic cellular

delivery, in which unassisted AON cellular uptake can be achieved in certain cell lines at high

concentrations and extended incubation time[18]. In siRNA, 2′F-ANA enhances siRNA nuclease stability

[9, 10], can be readily incorporated in the siRNA passenger strand[9, 10], and can be used in combination

with the rigid North-type nucleoside analogues 2′F-RNA and locked nucleic acid (LNA) to create fully

modified siRNAs with improved potency, reduced immunostimulatory properties, and a thermodynamic

bias for antisense strand RISC loading [10].

ON delivery methods[19-21] typically employ nucleic acid conjugates or delivery vehicles to complex

with ONs to facilitate uptake. Examples include liposomes [22, 23], cationic polymers[24-27], and

polyvalent siRNA structures[28]. ON encapsulation typically requires polycation block copolymers to

complex with the negatively charged siRNA[11, 26, 29]. Lipofectamine is a widely used transfection

reagent in cell culture; however, dose dependent toxicity[22] has been observed, and delivery by cationic

polymers is non-specific and requires a mechanism for endosomal escape.

Polymeric micelles with controllable size and material properties can be modified to achieve active tumor

targeting for therapeutic applications[30]. As well, PEGylated surfaces have the ability to decrease

opsinization of plasma proteins [31]. Although small molecule drugs are typically encapsulated within

micelles for drug delivery, there are examples in which conjugation to the surface of NPs can be

beneficial for achieving cellular uptake[32]. Therapeutics with low affinity for the hydrophobic NP core

are good candidates for surface conjugation, since encapsulation strategies result in rapid diffusion out of

the core[32]. The caveat for this strategy is the release of the therapeutic entity at the site of action. This

can be achieved by designing linkers to be susceptible to the microenvironment[33-35] or endogenous

proteins in the cell[29].

Here, the efficient cellular delivery of ONs conjugated to the polymeric NP shell of poly(ᴅ,ʟ-lactide-co-2-

methyl-2-carboxytrimethylene carbonate)-graft-poly(ethylene glycol) (P(LA-co-TMCC)-g-PEG) NPs is

demonstrated. Because the ONs are on the NP shell, delivery of natural and chemically modified siRNAs

and AONs was investigated to observe possible improvements in nuclease resistance. Using orthogonal

Page 47: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

38

click reactions (Diels-Alder and copper catalyzed azide-alkyne cycloadditions (CuAAC)), NPs were

conjugated first with maleimide-modified antibodies, followed by 3′alkyne-modified gene silencing ONs.

ON sequences were designed to facilitate enzyme-mediated release upon cellular uptake. Results

demonstrate that the combination of chemically modified ONs, P(LA-co-TMCC) micelles, and

trastuzumab (Herceptin®) antibodies provides effective targeted gene silencing. All three components are

required for cell-specific targeted ON delivery and gene silencing comparable to Lipofectamine. These

strategies are amenable to the delivery of both siRNAs and AONs, as evidenced by knockdown of firefly

luciferase expression from a pGL3 vector in SKOV-3luc cells (HER2+, luc+).

3.3 Materials and Methods

5-methyl-5-benzyloxycarbonyl-1,3-trimethylene carbonate (benzyl-protected TMCC, TMCC-Bn) was

synthesized as previously reported [36]. 3,6-dimethyl-1,4-dioxane-2,5-dione and 1-[3,5-

bis(trifluoromethyl)phenyl]-3-[(1R,2R)-(–)-2(dimethylamino) cyclohexyl] thiourea (Strem Chemicals,

Newburyport, MA) were used following previous protocols to synthesize P(LA-co-TMCC) [37]. Boc-

NH-PEG(10K)-NHS (Rapp Polymere, Tübingen, Germany) was modified as previously reported

protocols for shown furan and azide-functional groups [36, 38]. N,N’-diisopropyl carbodiimide, N,N’-

diisopropylethylamine and hydroxybenzotriazole (TRC, Toronto, ON) were used as received. The human

ovary cancer cell line SKOV-3luc was purchased from Cell Biolabs, Inc (San Diego, CA). The

Lipofectamine LTX® and Plus Reagent were purchased from Invitrogen (Burlington, ON) and the

Luciferase Assay System was from Promega (Madison, WI). All solvents and reagents were purchased

from Sigma-Aldrich and were used as received, unless otherwise noted.

The 1H NMR spectra were recorded at 400 MHz at room temperature using a Varian Mercury 400

spectrometer. The chemical shifts are in ppm. The molecular weights and polydispersity of P(LA-co-

TMCC) were measured by gel permeation chromatography (GPC) in THF relative to polystyrene

standards at room temperatures on a system with two-column sets (Viscotek GMHHR-H) and a triple

detector array (TDA302) at a flow rate of 0.6 mL/min. Polymeric micelle size and distribution were

measured by dynamic light scattering (DLS) using the Zetasizer Nano ZS system (Malvern, UK). The

NH2-PEG-azide was characterized using the Perkin-Elmer FT-IR Spectrum 1000. The Sepharose CL-4B

column was prepared by soaking the beads in distilled water overnight and packing the beads in a column

(5 x 15 cm). The column was washed with distilled water for 1 h before use and the flow rate was

determined by gravity. The Sephadex G-25 column was prepared using the same method, except PBS

buffer (1x, pH 7.4) was used as the eluent and the beads were packed in a column (1.5 x 15 cm). FPLC of

Page 48: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

39

the micelles used a Superdex 200 column. The column was washed with distilled water for 20 min and

PBS buffer (1x, pH 7.4) for 20 min at a flow rate of 1.5 mL/min before use. Fluorescence measurements

were completed with the Tecan Infinite M200 Pro fluorescent plate reader and absorbance was quantified

using the NanoDrop Spectrophotometer. Luminescence was measured with the MicroLumat Plus LB96V

(EG&G Berthold, Bad Wildbad, Germany).

3.3.1 Polymer Synthesis and Characterization

Synthesis of P(LA-co-TMCC), modification of NH2-PEG-NHS for furan and azide functionalities and

grafting of the PEG onto the polymer backbone, have all been previously shown [37]. Detailed

characterization for the batches of polymer used in these experiments are shown in Chapter 2.

Synthesis of (P(LA-co-TMCC). 1H NMR (CDCl2, 400MHz): δ 1.22 (br s, CH3 from TMCC), 1.59 (m,

CH3 from LA), 4.31 (br s, CH2 from TMCC), 5.15 (m, CH from LA), 7.33 (m, benzyl) and 8.02 (m,

pyrene).

Synthesis of NH2-PEG-azide. Perkin-Elmer FT-IR Spectrum 1000: terminal azide group (2100cm-1

),

terminal amine (3389 cm-1

), C–H alkane (2888 cm-1

), C=O amide (1692 cm-1

), and C–O ether (1101

cm-1

).

Synthesis of NH2-PEG-furan. 1H NMR (CDCl3, 400 MHz): δ 1.44 (s, Boc CH3), 2.32 (t, J=14 Hz , –

CH2–), 3.61 (m, –OCH2CH2–) , 4.42 (s, –CH2-furan), 6.22 (d, J= 3Hz, furan C-3), 6.30 (dd, J=5 Hz,

furan C-4) and 7.34 (d, J=3 Hz, furan C-5).

Synthesis of P(LA-co-TMCC)-g-PEG-X. For X=azide, 1H NMR (CDCl2, 400 MHz): δ 1.25 (br s, CH3

from TMCC), 1.58 (m, CH3 from LA), 3.59-3.69 (m, –OCH2CH2–), 4.35 (br s, CH2 from TMCC) and

5.17 (m, CH from LA). For X= furan, 1H NMR (CDCl2, 400 MHz): δ 1.22 (br s, CH3 from TMCC),

1.56 (m, CH3 from LA), 3.59-3.69 (m, –OCH2CH2–), 4.31 (br s, CH2 from TMCC) and 5.15 (m, CH

from LA).

Table 3.1. Characterization of poly (LA-co-TMCC) copolymer backbone

Copolymer

backbone

Mn (GPC)a

(kDa)

Mw/Mn (GPC)a TMCC content

(theoretical)b

(mol%)

TMCC content

(cal)c

(mol%)

P(LA-co-TMCC) 11.2 1.12 10 7.1 a Determined by GPC measurements in THF with poly(styrene) standards;

Page 49: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

40

b At quantitative monomer conversion

c Determined using

1H NMR measurements in CDCl3. Calculated by comparing the integral of the

methylene groups in TMCC (4.31 ppm) with the methyl groups of LA (5.15ppm)

Table 3.2. Characterization of poly(LA-co-TMCC)-g-PEG-X graft copolymer

Graft copolymer Average PEGa grafts/copolymer

backbone

Mn (1H NMR)

(kDa)

P(LA-co-TMCC)-g-PEG-azide 0.9 18.2

P(LA-co-TMCC)-g-PEG-furan 0.7 20.2 a PEG: Mn= 10kDa from manufacturer

b Estimated from

1H NMR and calculated by comparing the integral of the methylene groups of the PEG

(3.61-3.64 ppm) and the methylene groups of TMCC (4.31 ppm)

3.3.2 Oligonucleotide Synthesis and Characterization

All oligonucleotides were synthesized by Glen Deleavey; a PhD student working in Dr. Masad

Damha’s Lab at McGill University. Details relating to the procedure and characterization of the

oligonucleotides are in Appendix B.

3.3.3 Nanoparticle Formation and Characterization

Micelle formation. The micelles were prepared by co-self-assembly of P(LA co-TMCC)-g-PEG-furan

and P(LA-co-TMCC)-g-PEG-azide by membrane dialysis, as previously shown for single functionalized

chains [39]. P(LA-co-TMCC)-g-PEG-furan (2.5 mg, 18.2 kDa) and P(LA-co-TMCC)-g-PEG-azide (1.3

mg, 20.2 kDa) were combined to form the dual-functional micelles with an average ratio of 1:2 furan to

azide functional groups available. Dynamic light scattering (DLS) measurements of the micellar

nanoparticles determined a hydrodynamic diameter of 63.35 nm and polydispersity of 0.186.

Double-click reaction. P(LA-co-TMCC)-g-PEG micelles with azide and furan functional groups (680

nmol, 200 μM) were sequentially reacted with maleimide and alkyne-functional moieties. First,

trastuzumab-SMCC (250 nmol, 80 mg/mL) was added at room temperature with MES buffer (1mL, 100

mM, pH 5.5) for 24 h. The solution was dialyzed overnight against PBS buffer (1x, pH 7.4) using 2 kDa

MWCO dialysis tubing. The buffer was changed every 2 h for the first 6 h. The trastuzumab-nanoparticles

were concentrated to 100 µM using centrifuge concentrators at 1600 rpm.

Page 50: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

41

The trastuzumab-nanoparticles (50 nmol, 40 µM) were transferred to a glass vial to react with the alkyne-

modified oligonucleotide (35 nmol, 30 µM), copper sulphate (CuSO4, 40 µM), sodium ascorbate (NaAsc,

113 µM) and tris[(1-benzyl-1H-1,2,3-triazol-4-yl)methyl]amine (TBTA, 60 µM) in 3% methanol at room

temperature for 24 h. Micelles were purified by FPLC (GE Healthcare AKTA Purifier) monitoring

absorbance at 215 and 280 nm. Free oligonucleotides were eluted at 7-10 mL and purified nanoparticles

were eluted at 15-20 mL. Fluorescence of the Cy5 (ex. 640 nm, em. 675 nm) was used to quantify the

conjugation of the oligonucleotides. Absorbance at 260 nm was use to determine the micelle

concentration after subtracting absorbance contributions from the oligonucleotides. The aggregation

number of the micelles have previously been calculated to be 3500. There was approximately 30 ± 10

trastuzumab and 120 ± 40 siRNA molecules per micelle (Table 3.3).

Table 3.3. Coupling efficiency of oligonucleotides to micelles. A 1:2 ratio of furan to azide groups were

available for reaction on an average nanoparticle. There was an average of 1800 azide groups per micelle.

Number of

oligonucleotides per

micelle

% of azide groups

reacted

Di-siRNA 165 9.2

Mod-Di-siRNA 95 5.3

D-AON 145 8.1

Mod-D-AON 90 5.0

3.3.4 Luciferase Characterization

Cell Culture with oligonucleotide-nanoparticles. SKOV-3luc cells were cultured in McCoy’s 5A media

containing 10% FBS and 1% penicillin/streptomycin under standard culture conditions (37°C, 5% CO2,

100% humidity). Cells were seeded in 96 well plates at 7000 cells/well and allowed to adhere overnight.

The cell media was replaced with serum free media and dual functionalized micelles having both

trastuzumab and oligonucleotides were added. The cells were incubated for 24h. For each type of

nanoparticle, 8 replicates were prepared. Untreated cells were prepared simultaneously as a baseline for

comparison. Controls of unmodified nanoparticles and trastuzumab-nanoparticles were used to observe

the effect of the nanoparticles on luciferase activity. As well, oligonucleotides without nanoparticles were

delivered using a cationic lipid delivery system, Lipofectamine, to confirm the effectiveness of the AON

and siRNA sequences. Scrambled sequences of oligonucleotides were also included as a negative control.

Gene silencing response to nanoparticles. The cells were treated with three concentrations of AON or

siRNA. After the coupling reaction with the oligonucleotides, the number of AON or siRNA per

Page 51: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

42

nanoparticle was calculated. The nanoparticles were diluted with unmodified immuno-nanoparticles for a

final nanoparticle concentration of 1.7 µM and AON or siRNA concentrations of 56 nM, 14 nM or 2.8

nM (100 µL final volume in each well). Concentrations of siRNA were based on protocols targeting

firefly luciferase in HeLa X1/5 cells [10]. Each well was treated with 50 µL of nanoparticles and an

additional 50 µL of serum free McCoy’s 5A media was added. For the positive controls, the AON or

siRNA (5 µL) were complexed with the Lipofectamine (10 µL) and Plus reagent (10 µL) for 30 min, then

added to the cells. β-galactosidase was co-transfected with the free siRNA so the luminescence readings

could be normalized to account for transfection efficiency. The final volume of cell media was adjusted to

100 µL using serum free media.

Persistence of gene silencing duration. Following the procedure outlined above, the cells were treated

with AON- or siRNA-nanoparticles for 1, 3, 4 and 7 days. 7 x 105 cells were seeded into T-25 flasks and

allowed to adhere overnight. The cells were treated with 5 mL of serum free media and 5 mL of

nanoparticles (final concentrations of 1.7 µM nanoparticles, 56 nM AON or siRNA) for 5 h of

transfection. Then the cell media was replaced with complete media. The cells were split when confluent

and cell media was replaced every 2 days. 5 repeats of each sample were prepared. At each of the time

points, the luciferase activity was measured by the luciferase cell assay.

Luciferase cell assay. The cell media was removed and each well was washed three times using PBS

buffer (1x, pH 7.4) before adding 20 µL of lysis buffer. After 20 min, the 10 µL of lysate was transferred

to a white bottom 96 well plate. Following the Promega protocol, the luciferase assay reagent was

prepared. The auto-injector was used to add 25 µL of luciferase reagent to each well and measure

luminescence.

For each sample where β-galactosidase was co-transfected, another 10 µL of lysate was transferred to a

clear 96 well plate. A fresh mixture of Na2HPO4•7H2O (81 mM), NaH2PO4•H2O (18 mM), MgCl2 (2.3

mM), β-mercaptoethanol (49 mM) and ortho-nitrophenyl-β-galactoside (ONPG, 3.3 mM) was prepared.

90 µL of the ONPG mixture was added to the lysate and incubated for 30 min. The absorbance of the o-

nitrophenol formed was measured by the Tecan plate reader at 420 nm.

3.4 Results

P(LA-co-TMCC)-g-PEG-X micelles (D = 63.35 ± 6.95 nm, PDI= 0.186 ± 0.03) with X as both furan and

azide reactive groups on the PEG-X corona were sequentially reacted with trastuzumab-SMCC and

Page 52: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

43

alkyne-functionalized ONs, respectively, as shown in Figure 3.1. For siRNA delivery, nanoparticles were

prepared using Dicer substrate [40] 27nt versions of both native unmodified siRNAs (Di-siRNA) and 2′F-

ANA/2′F-RNA-modified dicer substrate siRNAs (Mod-Di-siRNA) corresponding to potent designs

observed in Deleavey et al. (2010) (sequences and modifications are outlined in Figure 3.2). Negative

control sequences consisted of non-targeting dicer substrate (i.e. scrambled) siRNAs: unmodified (Sc-Di-

siRNA) and modified (Mod-Sc-Di-siRNA) (Figure 3.2). For comparisons to standard ON transfection

methods, the activity of 21-mer siRNAs (siRNA) and modified siRNAs (Mod-siRNA) delivered by

Lipofectamine were measured. siRNA sense strands were modified with an alkyne functionality at the 3′

terminus for reaction with the azide groups on the micelle via CuAAC reactions [41]. A Cy5 label was

added to the 5′ end of the sense strands for quantification, revealing an average of 120 ± 40 siRNA

molecules and 30 ± 10 antibodies per micelle .

Figure 3.1 siRNA and AON Delivery Strategies. (a) siRNA delivery constructs. Immuno-NPs carry

dicer-substrate type siRNA constructs. Unmodified dicer-substrate siRNAs (Di-siRNA) are shown on top;

2′F-ANA/2′F-RNA modified Di-siRNAs (Mod-Di-siRNA) corresponding to active designs from Deleavey

et al. (2010) are shown below. (b) AON delivery constructs. Immuno-NPs carry AONs annealed to RNA

complement strands. Both a PS-DNA AON (D-AON, top) and a PS-2′F-ANA gapmer AON (Mod-D-

AON, bottom) were utilized. (c) NPs were dual functional, allowing antibody attachment through Diels

Page 53: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

44

Alder cycloaddition, and ON attachment via a CuAAC reaction. (d) Chemically modified ON backbones

used in these siRNA and AON constructs.

SKOV3-luc cells expressing HER2 and firefly luciferase were treated with the siRNA- and trastuzumab-

functionalized immuno-nanoparticles (Her-NPs for Herceptin®-Nanoparticles). Knockdown was

quantified by measuring luminescence, where a lower luminescent signal corresponds to greater gene

knockdown (Figure 2). The 2′F-ANA/2′F-RNA-modified Mod-Di-siRNA was equally effective when

delivered via NPs or Lipofectamine; however, the unmodified Di-siRNA showed somewhat greater

potency using Lipofectamine delivery (p<0.05), perhaps due to nuclease degradation and underscoring

the importance of Di-siRNA modification. When comparing NP delivery of modified and unmodified

siRNA, Mod-Di-siRNA demonstrated greater knockdown (p<0.05) whereas with Lipofectamine

transfection, there was no statistical difference (p>0.05). All statistics were completed with a one-way

ANOVA, unless otherwise noted. When 56 nM and 14 nM of a targeting siRNA sequence were delivered,

a knockdown effect was observed.

Among non-targeting negative controls, there was no statistical difference in luciferase levels regardless

of treatment type (unmodified/modified ON, Her-NP or LipA) or concentration (p>0.05, two-way

ANOVA). Targeting sequences resulted in lower luciferase levels when compared to relevant non-

targeting sequences at the same concentration (p≤0.012) with the exception of 2.8 nM of siRNA delivered

by Lipofectamine (p=0.213) and 2.8 nM of Di-siRNA Her-NP. While at 2.8 nM, the siRNA is near the

minimum concentration required to see a significant knockdown effect, the knockdown observed with

modified Mod-Di-siRNA (at 2.8 nM), by both immuno-nanoparticle and Lipofectamine, demonstrates its

enhanced stability and potency.

Page 54: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

45

0

0.2

0.4

0.6

0.8

1

1.2

Mod-Di-

siRNA

Her-NP

Mod-

siRNA

+ LipA

Di-siRNA

Her-NP

siRNA

+ LipA

Sc-Mod-

Di-siRNA

Her-NP

Sc-Mod-

siRNA

+ LipA

Sc-Di-

siRNA

Her-NP

Sc-siRNA

+ LipA

Rel

ativ

e Lu

cife

rase

Lev

el

56 nM

14 nM

2.8 nM

Duplex Sequence Di-siRNA 5' Cy5 – CAG AAG CUA UGA AAC GAU AUG GGC U – alkyne 3'

3' AC GUC UUC GAU ACU UUG CUA UAC CCG A 5'

Mod-Di-siRNA 5' Cy5 – CAG AAG CTA UGA AAC GAU ATG GGC U – alkyne 3'

3' AC GUC UUC GAU ACU UUG CUA UAC CCG A 5'

Sc-Di-siRNA 5' Cy5 – GCU UGA UUU CUG AAA UUA AUU CUG C – alkyne 3'

3' AC CGA ACU AAA GAC UUU AAU UAA GAC G 5'

Sc-Mod-Di-siRNA

5' Cy5 – GCT UGA TTT CUG AAA UTA ATT CUG C – alkyne 3'

3' AC CGA ACU AAA GAC UUU AAU UAA GAC G 5'

siRNA 5' CAG AAG CUA UGA AAC GAU AUG 3'

3' AC GUC UUC GAU ACU UUG CUA U 5'

Mod-siRNA 5' CAG AAG CTA UGA AAC GAU ATG 3'

3' AC GUC UUC GAU ACU UUG CUA U p 5'

Sc-siRNA 5' GCU UGA UUU CUG AAA UUA AUU 3'

3' AC CGA ACU AAA GAC UUU AAU U 5'

Sc-Mod-siRNA 5' GCT UGA TTT CUG AAA UTA ATT 3'

3' AC CGA ACU AAA GAC UUU AAU U p 5'

Figure 3.2 Modified and unmodified siRNA (sequences in Table) were delivered to SKOV3-luc cells by

conjugation to immuno-nanoparticles (Her-NP). Positive controls were prepared by delivering 21mer

siRNA with Lipofectamine (LipA), and negative controls used non-targeting, scrambled (Sc) sequences.

Luciferase levels were normalized to untreated cells. Each treatment was repeated 8 times (mean ±

standard deviation plotted). Bars with the same lower case letter denote no statistical significance between

pairs letters (one-way ANOVA, p>0.05). Bars with different letters are statistically different (one-way

ANOVA, p<0.01). Statistical significance (one-way ANOVA) is denoted by * (p≤0.05), ** (p≤0.01),

***(p≤0.001). Scrambled sequences were compared to active sequences by a two-way ANOVA and

*** *

*** ***

*

a a b c b c d a b d

d

Page 55: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

46

showed a statistical difference (p<0.05). Legend for sequences: RNA, 2′F-ANA, 2′F-RNA, Cy5 =

Cyanine 5, alkyne = 3′ alkyne modification, p = 5′ phosphate.

SKOV3-luc cells were also treated with AON-functionalized immuno-nanoparticles and tested to

luciferase knockdown relative to controls. In this construct, PS-DNA (D-AON) and PS-2′F-ANA gapmer

(Mod-D-AON) were annealed to complementary RNA strands, which were modified with 3′ alkyne and

5′ Cy5 functionalities, to allow for click-coupling to azide-functionalized NPs and subsequent

quantification. Upon cellular uptake, ON duplexes may be substrates for RNase H, resulting in RNA

strand cleavage and release of active AONs. Firefly luciferase gene silencing experiments using PS-DNA

and PS-2′F-ANA AONs with Lipofectamine and Her-NP mediated delivery are shown in Figure 3.3.

Lipofectamine transfections were completed with the RNA-AON duplex, which showed similar activity

to the single strand AONs. The D-AON and Mod-D-AON sequences showed greater knockdown potency

using Lipofectamine delivery (p<0.001) when compared to the Her-NPs; however, at the lowest AON

concentration (2.8 nM), there was no statistical difference between the two methods of delivery for Mod-

D-AON and D-AON (p>0.05).

Immuno-nanoparticle delivery of Mod-D-AON was significantly more potent than that with D-AON

(p<0.001), underscoring the importance of stability achieved with modification; however, this difference

was not observed with Lipofectamine delivery (p>0.05). Both D-AON and Mod-D-AON were more

effective at luciferase knockdown with Lipofectamine vs. NP delivery. The dimunition of D-AON (PS-

DNA) potency, and retention of potency for the Mod-D-AON (PS-2′F-ANA), suggests that nuclease

degradation prior to mRNA cleavage may play a role in determining gene silencing success when AONs

are delivered on the surface of these Her-NPs, although uptake and AON release efficiency may also

contribute to the observed differences.

Luciferase levels reflected a gene silencing effect to AON treatments. In all cases, active AONs showed

greater gene knockdown/lower luciferase levels when compared to equivalent treatments using non-

targeting PS-DNA (Sc-D-AON) or PS-2′F-ANA gapmer (Sc-Mod-D-AON) AONs, with the exception of

the 2.8 nM concentrations of AON delivered by either Lipofectamine or Her-NPs (p>0.05). At 2 nM, the

AON is likely outside the effective concentration range. There is no statistical difference between

delivery methods and scrambled sequences at this concentration.

Page 56: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

47

0

0.2

0.4

0.6

0.8

1

1.2

Mod-D-

AON

Her-NP

Mod-D-

AON

+ LipA

D-AON

Her-NP

D-AON

+ LipA

Sc Mod-D-

AON

Her-NP

Sc-Mod-D-

AON

+ LipA

Sc-D-AON

Her-NP

Sc D-AON

+ LipA

Rel

ativ

e Lu

cife

rase

Lev

el

56nM

14nM

2.8nM

D-AON 5' Cy5 – AGA CAA AGG CUA UCA GGU GGC CU – alkyne 3'

3' t* g*t*t* t*c*c* g*a*t* a*g*t* c*c*a* c*c*g* 5'

Mod-D-AON 5' Cy5 – AGA CAA AGG CUA UCA GGU GGC CU – alkyne 3'

3' t* g*t*t* t*c*c* g*a*t* a*g*t* c*c*a* c*c*g* 5'

Sc-D-AON 5' Cy5 – GCU UGA UUU CUG AAA UUA AUU CUG C – alkyne 3'

3' a* a*c*t* a*a*a* g*a*c* t*t*t* a*a*t* t*a*a 5'

Sc-Mod-D-AON 5' Cy5 – GCU UGA UUU CUG AAA UUA AUU CUG C – alkyne 3'

3' a* a*c*t* a*a*a* g*a*c* t*t*t* a*a*t* t*a*a 5'

Figure 3.3 Mod-D-AON and unmodified D-AON were delivered to SKOV3-luc cells by immuno-

nanoparticles (Her-NP). Positive controls were prepared by delivery with Lipofectamine (LipA), and

negative controls used scrambled sequences. Luciferase levels were measured by luminescence and

normalized to untreated cells. Each treatment was repeated 8 times (mean ± standard deviation reported).

The same lower case letter denotes no statistical significance between pairs (one-way ANOVA, p>0.05).

Bars with different letters are statistically different (one-way ANOVA, p≤0.05). Statistical significance

(one-way ANOVA) is denoted by * (p≤0.05), ** (p≤0.01), ***(p≤0.001). Scrambled sequences were

compared to active sequences by a two-way ANOVA and showed a statistical difference (p<0.05).

To understand the role of the nanoparticle, siRNA and trastuzumab to the gene knockdown observed with

immuno-nanoparticle delivery of siRNA, SKOV-3luc cells were treated with siRNA delivered by the

Her-NP system vs. NPs alone, NPs derivatized with only trastuzumab or siRNA (not both), and a mixture

of Her-NPs and siRNA (without covalent attachment of the siRNAs) (Figure 3.4). and measured average

luciferase levels of were observed. In all cases, luciferase levels of these controls were 0.891 or greater,

which is significantly greater than the 0.416 ± 0.087 measured for Mod-Di-siRNA Her-NPs (p<0.001),

*** ***

** **

*** ***

*** ***

**

a b c a d b c d

d

Page 57: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

48

demonstrating the importance of these three components in achieving a transfection-competent gene

silencing NP complex. There was no statistical difference among the controls (p>0.05).

0

0.2

0.4

0.6

0.8

1

1.2

Di-siRNA

Her-NP

NP Her-NP Di-siRNA NP Di-siRNA

mixed with

Her-NP

Rel

ativ

e Lu

cife

rase

Lev

el

Figure 3.4 Luciferase levels for NP controls demonstrate targeting effects of the immuno-nanoparticle.

From left to right, treatments consisted of unmodified Di-siRNA functionalized Her-NPs, NP alone, Her-

NP alone, Di-siRNA functionalized NPs, or Di-siRNA mixed (but not chemically conjugated) with Her-

NPs. In all treatments, the polymer concentration was maintained at 1.7 µM and the siRNA concentration

was 14 nM. 5 repeats were collected for each control group, and 8 repeats for the siRNA Her-NP (mean ±

standard deviation shown).

Chemical modification with phosphorothioates, 2′F-ANAs, and 2′F-RNAs is expected to not only protect

AONs and siRNAs from degradation during cellular delivery, but also extend the duration of gene

silencing activity. To observe differences in duration of effect, NP-delivered siRNA activity was

monitored over 7 days (Figure 3.5). Both modified and unmodified siRNAs showed a persistence of

effect for up to 4 days, after which the cells were split due to cell confluency, thereby possibly diluting

out the knockdown. The modified siRNA and siRNA delivered with immuno-nanoparticles demonstrated

greater knockdown effect relative to scrambled controls and the modified siRNA-Her-NP showed the

greatest knockdown at all time points until full rebound at day 7 (p≤0.02).

***

Page 58: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

49

0

0.2

0.4

0.6

0.8

1

1.2

0 2 4 6 8

Days after treatment

Rel

ativ

e Lu

cife

rase

Lev

el

Mod-Di-siRNA NP

Di-siRNA NP

Sc-Mod-Di-siRNA NP

Sc-Di-siRNA NP

Figure 3.5. Persistence study for siRNA NPs at 56nM concentration. SKOV-3luc cells were treated with

siRNA-carrying Her-NPs for 24h. Luciferase levels were measured at 1, 3, 4 and 7 day intervals

following treatment. The knockdown effect for siRNA-NPs was persistent for up to 4 days. Confluent

cells were split at day 4. By day 7, all treatment groups ceased to report a gene silencing effect. Each

sample was repeated 5 times.

Differences in the duration of activity for D-AON and Mod-D-AON AONs delivered via NPs were also

observed. Compared to the D-AON NPs and NPs with non-targeting sequences, the Mod-D-AON NPs

displayed a persistent effect (Figure 3.6). Gene knockdown was observed for up to 4 days after which the

cells were split. At day 7, luciferase levels returned to untreated levels.

The cells were split at day 4 due to cell confluency. The modified siRNA demonstrated a greater

knockdown effect at all time points until full rebound at day 7 (p≤0.02). Non-targeting controls did not

affect expression levels.

Page 59: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

50

0

0.2

0.4

0.6

0.8

1

1.2

0 2 4 6 8

Days after AON Her-NP Treatment

Rel

ativ

e Lu

cife

rase

Lev

el

Mod-D-AON NP

D-AON NP

Sc-Mod-D-AON NP

Sc-D-AON NP

Figure 3.6 Persistence study for AON NPs at 56nM concentration. SKOV-3luc cells were treated with

AON-modified Her-NPs for 24h. Luciferase levels were measured after 1, 3, 4 and 7 days. The

knockdown effect for Mod-D-AON NPs was persistent for up to 4 days. Confluent cells were split at day

4. By day 7, all treatment groups all treatment groups ceased to report a gene silencing effect. Each

sample was repeated 5 times (mean ± standard deviation shown).

3.5 Discussion

Both unmodified and 2′F-ANA/2′F-RNA-modified Di-siRNAs were effectively delivered by Her-NPs in

an antibody-targeted fashion. Modified Di-siRNA showed similar levels of activity when delivered by

either Lipofectamine or Her-NPs, demonstrating that Her-NPs covalently modified with Mod-Di-siRNAs

are equipotent to Lipofectamine delivery and cell-specific. By achieving equipotency without the toxicity

often associated with Lipofectamine, the Her-NP-Mod-Di-siRNA provides a more desirable siRNA

delivery vehicle . This delivery strategy requires a combination of 3 components to achieve effective ON

delivery: a biodegradable[36] polymer-based nanoparticle core, antibody functionalization for active

targeting, and stabilizing ON chemical modifications (PS, 2′F-ANA, 2′F-RNA). Furthermore, chemical

modification of siRNAs with 2′F-ANA and 2′F-RNA has been shown to reduce siRNA-triggered

immunostimulation[10], which can be particularly important when siRNAs are introduced to cells via a

delivery vehicle[5].

AONs can also be delivered using the Her-NP strategy, although Her-NP-mediated delivery was only

successful with the PS-2′F-ANA gapmer AON, and not with a PS-DNA AON. Even with the PS-2′F-

ANA AON, gene silencing levels were slightly reduced compared with Lipofectamine-mediated delivery

(as expected, PS-DNA and PS-2′F-ANA AONs both effected potent silencing when delivered by

Page 60: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

51

Lipofectamine). Results indicate that the reduction in potency of the PS-2′F-ANA AONs when delivered

by Her-NPs was only approximately 2-fold compared with Lipofectamine, indicating that Her-NP-

mediated delivery can maintain AON potency within a very reasonable concentration range when

chemically modified AONs are used. On the Her-NPs, AONs are partially protected by the PEG corona,

but exposure to nucleases could cause degradation and/or premature AON release prior to uptake. In

contrast, Lipofectamine complexes with the negatively charged ONs, protecting them from nucleases in

addition to facilitating uptake. Kim, et al. (2010) have shown that liposomes and PEGylated liposomes

can efficiently protect siRNA from RNase degradation[42]. These differences could help explain the

observed differences in gene silencing potency between PS-DNA and PS-2′F-ANA AONs.

The antibody-targeted delivery strategy implemented here is particularly interesting for the delivery of

ON therapeutics, where tissue-specific uptake is highly desirable. Studies on the pharmacokinetics and

tumor localization of siRNA delivered by polycationic carriers showed little difference versus

unformulated siRNA [43]. When siRNA was delivered with targeted NPs to mice, Bartlett et. al saw

evidence that more functional siRNA could be delivered to the tumor, even though non-targeted NPs also

accumulated in the tumor microenvironment to a similar extent [44]. This underscores the importance of

targeted delivery to the cell and not just the tumour. Based on previous studies where Her-NPs enter the

cell via receptor-mediated endocytosis [32, 36, 45], ON immuno-nanoparticles also likely accumulate in

the cell cytoplasm after endosomal escape, thereby achieving target specificity through a specific cell

surface receptor[32, 46, 47]. This cell-specific delivery differs from that of Lipofectamine which

complexes with the ONs to overcome the electrostatic repulsion of the cell surface, and induces local

destabilization of the plasma membrane allowing translocation into the cytoplasm in a non-specific

fashion [23, 48]. The Her-NP delivery strategy we have identified features active antibody-targeting, and

gene silencing potencies comparable with Lipofectamine delivery but with greater cell specificity and

without the associated cytotoxicity.

Transfected ONs are internalized by endocytosis into early and late endosomes, and subsequently

lysosomes. While the mechanism of endosomal escape by Her-NP delivered siRNAs and AONs is still

under investigation, potential explanations include lysosomal degradation or endo-lysosomal escape[18,

49]. Poly(ᴅ,ʟ-lactide-co-glycolide) (PLGA) NPs have a negative charge in physiological pH, but acquire a

positive charge in the acidic endo-lysosomal compartments and escape with a similar mechanism to

cationic lipids [49]. Our nanoparticles have a similar backbone comprised primarily of lactide monomers

and net negative charge [37] and may follow a similar mechanism. Endosomal escape of the lipoplex is

better understood: the complex destabilizes the endosomal membrane by switching the anionic lipids in

Page 61: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

52

the membrane with the cationic lipid. As a consequence, AONs are translocated into the cytoplasm [50,

51].

It is hypothesized that active siRNA is released from the Her-NPs when Dicer processes the dicer

substrate siRNA sequences [51, 52]. In our experiments, we attributed gene silencing to the effective

release of siRNA by Dicer cleavage. However, Dicer efficiency could also be measured by labelling the 5′

and 3′ ends of siRNA with a fluorescent donor and quencher. Upon cleavage by the Dicer, the

fluorescence intensity of the donor would increase [53]. Monitoring Dicer cleavage could provide insight

into whether the knockdown efficiency is hindered by incomplete release of siRNA. AONs might be

released following RNase H-mediated cleavage of the RNA complement strands covalently attached to

the NPs. Sequence designs for the siRNA and AON delivery strategies are illustrated in Figure 1. The

dicer substrate was designed to allow siRNA release in the cellular environment: Di-siRNAs on the

surface of the NPs are a potential substrate for cellular nucleases, which would furnish active siRNAs

following enzymatic processing. Dicer substrate siRNA designs were selected featuring a 2nt 3′-CA

overhang on the antisense strand of the construct. It has been shown that this overhang facilitates better

Dicer processing versus blunt ended duplexes, although this overhang is also associated with increased

degradation in serum [50, 51]. The 3′ sense strand terminus was selected as the point of attachment to the

NPs, again to facilitate Dicer processing to produce the desired active siRNA[52].

Unmodified and modified siRNAs (and modified AONs) delivered by Her-NPs have a duration of

activity up to at least 4 days. While the effect was attenuated by day 7, a dilution effect likely contributes

to observed diminution of activity because the cells reached confluency at day 4 and were split. Indeed,

SKOV3-luc cells double every 20 h, so the number of cells increases exponentially and the concentration

of ONs are likely below an effective concentration at day 7. The full recovery of luciferase levels

confirms that the NPs re non-cytotoxic.

3.6 Conclusion

P(LA-co-TMCC)-g-PEG NPs, targeted with trastuzumab and carrying unmodified or modified siRNAs

or AONs, are potent vehicles for tissue specific targeted gene silencing through either an RNAi or an

antisense mechanism. siRNA NPs achieved potencies comparable in effectiveness to the standard in

siRNA cell transfection protocol: Lipofectamine 2000. Although AON NPs showed reduced potency

compared with Lipofectamine delivery, gene silencing can still be readily achieved when PS-2′F-ANA

AONs are used. We have shown two new ON delivery systems based on antibody-targeted polymeric

Page 62: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

53

micelles carrying chemically modified ONs conjugated on their exterior shell. The described chemical

modifications improve the stability of ON sequences, and the attachment of antibodies to the micelle

allow for targeted delivery, both of which are essential for the success of this delivery strategy. The use of

NPs as an alternative to cytotoxic cationic transfection reagents for the delivery of siRNA or AONs

demonstrates the benefits achieved through incorporation of targeting ligands and chemically modified

ONs with a competent drug delivery vehicle.

3.7 References

[1] W. J. Kim and S. W. Kim, "Efficient siRNA Delivery with Non-viral Polymeric Vehicles,"

Pharmaceutical Research, vol. 26, pp. 657-666, Mar 2009.

[2] E. R. Rayburn and R. W. Zhang, "Antisense, RNAi, and gene silencing strategies for therapy:

Mission possible or impossible?," Drug Discovery Today, vol. 13, pp. 513-521, Jun 2008.

[3] G. F. Deleavey and M. J. Damha, "Designing Chemically Modified Oligonucleotides for

Targeted Gene Silencing," Chemistry & Biology, 2012.

[4] A. L. Jackson, et al., "Position-specific chemical modification of siRNAs reduces "off-target''

transcript silencing," Rna-a Publication of the Rna Society, vol. 12, pp. 1197-1205, Jul 2006.

[5] K. A. Whitehead, et al., "Silencing or Stimulation? siRNA Delivery and the Immune System,"

Annu. Rev. Chem. Biomol. Eng., vol. 2, pp. 77–96, 2011.

[6] J. K. Watts, et al., "Chemically modified siRNA: tools and applications," Drug Discovery Today,

vol. 13, pp. 842-855, Oct 2008.

[7] A. Kalota, et al., "2 '-Deoxy-2 '-fluoro-beta-D-arabinonucleic acid (2 ' F-ANA) modified

oligonucleotides (ON) effect highly efficient, and persistent, gene silencing," Nucleic Acids

Research, vol. 34, pp. 451-461, 2006.

[8] J. K. Watts, et al., "Studies on the hydrolytic stability of 2 '-fluoroarabinonucleic acid (2 ' F-

ANA)," Organic & Biomolecular Chemistry, vol. 7, pp. 1904-1910, 2009.

[9] T. Dowler, et al., "Improvements in siRNA properties mediated by 2 '-deoxy-2 '-fluoro-beta-D-

arabinonucleic acid (FANA)," Nucleic Acids Research, vol. 34, pp. 1669-1675, 2006.

[10] G. F. Deleavey, et al., "Synergistic effects between analogs of DNA and RNA improve the

potency of siRNA-mediated gene silencing," Nucleic Acids Research, vol. 38, pp. 4547-4557, Jul

2010.

[11] A. E. Felber, et al., "siRNA nanocarriers based on methacrylic acid copolymers," Journal of

Controlled Release, vol. 152, pp. 159-167, May 2011.

[12] K. L. Min, et al., "Oligonucleotides comprised of alternating 2 '-deoxy-2 '-fluoro-beta-D-

arabinonucleosides and D-2 '-deoxyribonucleosides (2 ' F-ANA/DNA 'altimers') induce efficient

RNA cleavage mediated by RNase H," Bioorganic & Medicinal Chemistry Letters, vol. 12, pp.

2651-2654, Sep 2002.

[13] J. K. Watts and M. J. Damha, "2 ' F-arabinonucleic acids (2 ' F-ANA) - History, properties, and

new frontiers," Canadian Journal of Chemistry-Revue Canadienne De Chimie, vol. 86, pp. 641-

656, Jul 2008.

[14] F. Eckstein, "Nucleoside Phophorothioates," Journal of the American Chemical Society, vol. 88,

pp. 4292-&, 1966.

[15] F. Eckstein, "A Dinucleoside Phosphorothioate," Tetrahedron Letters, pp. 1157-&, 1967.

[16] F. Eckstein, "Phosphorothioate oligodeoxynucleotides: What is their origin and what is unique

about them?," Antisense & Nucleic Acid Drug Development, vol. 10, pp. 117-121, Apr 2000.

Page 63: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

54

[17] M. J. Damha, et al., "Hybrids of RNA and arabinonucleic acids (ANA and 2 ' F-ANA) are

substrates of ribonuclease h," Journal of the American Chemical Society, vol. 120, pp. 12976-

12977, Dec 1998.

[18] N. Souleimanian, et al., "Antisense 2'-Deoxy-2'-Fluoroarabino Nucleic Acids (2'F-ANAs): In

Vitro Gymnotic Silencers of Gene Expression Whose Potency Is Enhanced by Fatty Acids,"

Molecular Therapy-Nucleic Acids, 2012.

[19] R. L. Juliano, et al., "The Chemistry and Biology of Oligonucleotide Conjugates," Accounts of

Chemical Research, vol. 45, pp. 1067-1076, Jul 2012.

[20] G. R. Rettig and M. A. Behlke, "Progress Toward In Vivo Use of siRNAs-II," Molecular

Therapy, vol. 20, pp. 483-512, Mar 2012.

[21] X. D. Yuan, et al., "Recent advances of siRNA delivery by nanoparticles," Expert Opinion on

Drug Delivery, vol. 8, pp. 521-536, Apr 2011.

[22] C. L. Zhang, et al., "siRNA-containing liposomes modified with polyarginine effectively silence

the targeted gene," Journal of Controlled Release, vol. 112, pp. 229-239, May 15 2006.

[23] B. Dalby, et al., "Advanced transfection with Lipofectamine 2000 reagent: primary neurons,

siRNA, and high-throughput applications," Methods, vol. 33, pp. 95-103, Jun 2004.

[24] J. F. Guo, et al., "Therapeutic targeting in the silent era: advances in non-viral siRNA delivery,"

Molecular Biosystems, vol. 6, pp. 1143-1161, 2010.

[25] B. Ozpolat, et al., "Nanomedicine based approaches for the delivery of siRNA in cancer,"

Journal of Internal Medicine, vol. 267, pp. 44-53, Jan 2010.

[26] K. Kataoka, "Smart polymeric micelles as nanocarriers for gene and drug delivery," 2004

International Conference on Mems, Nano and Smart Systems, Proceedings, pp. 4-5, 2004.

[27] K. A. Howard, et al., "RNA interference in vitro and in vivo using a chitosan/siRNA nanoparticle

system," Molecular Therapy, vol. 14, pp. 476-484, Oct 2006.

[28] J. I. Cutler, et al., "Polyvalent Nucleic Acid Nanostructures," Journal of the American Chemical

Society, vol. 133, pp. 9254-9257, Jun 22 2011.

[29] M. Elsabahy, et al., "Delivery of Nucleic Acids through the Controlled Disassembly of

Multifunctional Nanocomplexes," Advanced Functional Materials, vol. 19, pp. 3862-3867, Dec

2009.

[30] P. Couvreur and C. Vauthier, "Nanotechnology: Intelligent design to treat complex disease,"

Pharmaceutical Research, vol. 23, pp. 1417-1450, Jul 2006.

[31] V. P. Torchilin, et al., "Poly(Ethylene Glycol) on the Liposome Surface - on the Mechanism of

Polymer-Coated Liposome Longevity," Biochimica Et Biophysica Acta-Biomembranes, vol.

1195, pp. 11-20, Oct 12 1994.

[32] M. Shi, et al., "Doxorubicin-Conjugated lmmuno-Nanoparticles for Intracellular Anticancer Drug

Delivery," Advanced Functional Materials, vol. 19, pp. 1689-1696, Jun 9 2009.

[33] H. J. Kim, et al., "PEG-detachable cationic polyaspartamide derivatives bearing stearoyl moieties

for systemic siRNA delivery toward subcutaneous BxPC3 pancreatic tumor," Journal of Drug

Targeting, vol. 20, pp. 33-42, Jan 2012.

[34] S. Takae, et al., "PEG-detachable polyplex micelles based on disulfide-linked block catiomers as

bioresponsive nonviral gene vectors," Journal of the American Chemical Society, vol. 130, pp.

6001-6009, May 7 2008.

[35] G. Saito, et al., "Drug delivery strategy utilizing conjugation via reversible disulfide linkages:

role and site of cellular reducing activities," Advanced Drug Delivery Reviews, vol. 55, pp. 199-

215, Feb 10 2003.

[36] M. Shi, et al., "Immuno-polymeric nanoparticles by Diels-Alder chemistry," Angewandte

Chemie-International Edition, vol. 46, pp. 6126-6131, 2007.

[37] J. Lu and M. S. Shoichet, "Self-Assembled Polymeric Nanoparticles of Organocatalytic

Copolymerizated D,L-Lactide and 2-Methyl 2-Carboxytrimethylene Carbonate,"

Macromolecules, vol. 43, pp. 4943-4953, Jun 2010.

Page 64: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

55

[38] J. Lu, et al., "Click Chemistry Functionalized Polymeric Nanoparticles Target Corneal Epithelial

Cells through RGD-Cell Surface Receptors," Bioconjugate Chemistry, vol. 20, pp. 87-94, Jan

2009.

[39] M. Shi and M. S. Shoichet, "Furan-functionalized co-polymers for targeted drug delivery:

characterization, self-assembly and drug encapsulation," Journal of Biomaterials Science-

Polymer Edition, vol. 19, pp. 1143-1157, 2008.

[40] D. H. Kim, et al., "Synthetic dsRNA Dicer substrates enhance RNAi potency and efficacy,"

Nature Biotechnology, vol. 23, pp. 222-226, Feb 2005.

[41] M. Meldal and C. W. Tornoe, "Cu-catalyzed azide-alkyne cycloaddition," Chemical Reviews, vol.

108, pp. 2952-3015, Aug 2008.

[42] H. K. Kim, et al., "Enhanced siRNA delivery using cationic liposomes with new polyarginine-

conjugated PEG-lipid," International Journal of Pharmaceutics, vol. 392, pp. 141-147, Jun 15

2010.

[43] H. K. de Wolf, et al., "Effect of cationic carriers on the pharmacokinetics and tumor localization

of nucleic acids after intravenous administration," International Journal of Pharmaceutics, vol.

331, pp. 167-175, Mar 1 2007.

[44] D. W. Bartlett, et al., "Impact of tumor-specific targeting on the biodistribution and efficacy of

siRNA nanoparticles measured by multimodality in vivo imaging," Proceedings of the National

Academy of Sciences of the United States of America, vol. 104, pp. 15549-15554, Sep 25 2007.

[45] K. Ho, et al., "Tunable immunonanoparticle binding to cancer cells: thermodynamic analysis of

targeted drug delivery vehicles," Soft Matter, vol. 5, pp. 1074-1080, 2009.

[46] H. S. Yoo and T. G. Park, "Folate-receptor-targeted delivery of doxorubicin nano-aggregates

stabilized by doxorubicin-PEG-folate conjugate," Journal of Controlled Release, vol. 100, pp.

247-256, Nov 24 2004.

[47] J. W. Park, et al., "Development of Anti-P185(Her2) Immunoliposomes for Cancer-Therapy,"

Proceedings of the National Academy of Sciences of the United States of America, vol. 92, pp.

1327-1331, Feb 28 1995.

[48] X. H. Zhou and L. Huang, "DNA Transfection Mediated by Cationic Liposomes Containing

Lipopolylysine - Characterization and Mechanism of Action," Biochimica Et Biophysica Acta-

Biomembranes, vol. 1189, pp. 195-203, Jan 19 1994.

[49] J. Panyam, et al., "Rapid endo-lysosomal escape of poly(DL-lactide-co-glycolide) nanoparticles:

implications for drug and gene delivery," Faseb Journal, vol. 16, pp. -, Aug 2002.

[50] A. Vermeulen, et al., "The contributions of dsRNA structure to Dicer specificity and efficiency,"

Rna-a Publication of the Rna Society, vol. 11, pp. 674-682, May 2005.

[51] P. C. Patel, et al., "Duplex End Breathing Determines Serum Stability and Intracellular Potency

of siRNA-Au NPs," Molecular Pharmaceutics, vol. 8, pp. 1285-1291, Jul-Aug 2011.

[52] S. C. Kow, et al., "Dicer-Labile PEG Conjugates for siRNA Delivery," Biomacromolecules, vol.

12, pp. 4301-4310, Dec 2011.

[53] J. P. DiNitto, et al., "Continuous fluorescence-based method for assessing Dicer cleavage

efficiency reveals 3 ' overhang nucleotide preference," Biotechniques, vol. 48, pp. 303-309, Apr

2010.

Page 65: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

56

4. Conclusions

The overall purpose of this work was to determine whether oligonucleotides (siRNA or AONs) could be

coupled to the exterior PEG shell of polymer nanoparticles and maintain gene silencing activity. First, we

developed a strategy to sequentially label dual functional poly(LA-co-TMCC)-g-PEG micelles by double

click reactions. Secondly, we employed these methods to covalently attach antibodies and siRNA or

AONs to the nanoparticles. Cell assays used to measure luciferase levels of SKOV-3luc cells

overexpressing HER2 and luciferase validated the activity of the nanoparticles. The following

conclusions can be derived from this work:

1. Dual functional micelles can be formed by the co-dialysis of poly(LA-co-TMCC)-g-PEG-furan

and poly(LA-co-TMCC)-g-PEG-azide. The functional groups can be modified using sequential

Diels Alder and strain promoted azide-alkyne cycloadditions with maleimide- and

dibenzylcyclooctyne- modified moieties, respectively.

2. Immuno-nanoparticles formed using dual functional micelles can be further modified with

peptide sequences. A FLAG peptide was covalently coupled to trastuzumab-nanoparticles.

Confocal microscope imaging showed that the dual functional micelles remain intact to deliver

the peptide to the cells that overexpress HER2.

3. The siRNA and AON sequences target firefly luciferase in SKOV-3luc cells. A gene silencing

effect was observed when the oligonucleotides were delivered by immuno-nanoparticles or

Lipofectamine. In contrast, scrambled sequences did not demonstrate gene silencing activity.

4. Knockdown activity was only observed when active oligonucloetide sequences and trastuzumab

were covalently bound to the nanoparticle.

5. Of all the sequences used for gene silencing, modified 2’F-ANA siRNA was the most effective.

Modified siRNA showed equally effective levels of knockdown when delivered by immuno-

nanoparticles or by Lipofectamine.

6. Unmodified sequences delivered by the nanoparticle showed loss of activity and require 2’F-

ANA modification for effective gene silencing.

Page 66: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

57

5. Recommendations and Future Directions

In this work, we reported the gene silencing activity of siRNA and AON sequences delivered by immuno-

nanoparticles. Some recommended studies for future research:

1. Monitor cell uptake and release of nanoparticles using confocal microscopy.

Both the siRNA and AON mechanism require entry into the cell cytoplasm for Dicer and RNase enzymes

to be activated. Based on our knowledge of receptor-mediated endocytosis of immuno-nanoparticles, it is

hypothesized that the nanoparticles are taken up by the cells into endosomes or lysosomes [1]. However,

it is unclear how our negatively charged nanoparticle escapes the endosome. Literature has shown that

cationic liposomes or polymers can escape through the “proton sponge effect” [2]. One potential

explanation for endosomal escape stems from research by Panyam, et al. with PLGA nanoparticles [3].

Our poly(LA-co-TMCC)-g-PEG nanoparticle backbone is primarily composed of lactide units, thus its

net charge is similar to PLGA. PLGA nanoparticles were able to escape the endosome by selective

reversal of the surface charge from anionic to cationic in acidic endo-lysosomal compartments [3].

One proposed study uses confocal microscopy to gain a mechanistic understanding oligonucleotide

delivery using micelles. The siRNA and AONs have been modified with a Cy5 fluorophore that can be

used to track the nanoparticle. Unreacted TMCC units on the backbone can also be modified with

fluorophores to label the micelle. Confocal microscopy can monitor both the nanoparticle and the siRNA

to determine how both components are taken into the cell. No co-localization of oligonucleotides and

micelles would indicate separation of the siRNA/AON from the micelle.

A potential challenge is the low concentration of Cy5 since siRNA and AONs are delivered in picomolar

concentrations. If the concentration of oligonucleotides is too high, there could be cell cytotoxicity. In the

case where the Cy5 signal is too weak, the nanoparticles could be further modified with fluorescent dyes.

Unreacted azide and furan groups could be coupled with maleimide- and dibenzylcyclooctyne- modified

labels. The cells could be co-stained with endosomal markers to observe endocytosis of the nanoparticles.

This could address the question of whether the nanoparticles are taken into early/late endosomes or

lysosomes.

Since trastuzumab nanoparticles are shown to bind to the cell membrane after one hour and have receptor-

mediated uptake within 3 hours [4], it would be interesting to monitor the cells at these time points.

Page 67: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

58

2. Deliver clinically relevant modified Di-siRNA and D-AONs with immuno-nanoparticles.

Having demonstrated the gene silencing capability of modified siRNA and AON-nanoparticles, one

logical direction involves the use of clinically relevant oligonucleotide sequences for breast cancer. From

our results, we concluded that the fluorinated modification (2′F-ANA) improves siRNA and AON

stability and potency and is necessary for nanoparticle delivery. RNAi technology provides a potential

therapeutic strategy because of its high efficacy and specificity for down-regulating gene expression [5].

Current research with siRNA for cancer therapy involves knockdown of genes involved in oncogenesis

pathways, apoptosis, tumor-host interaction and resistance to conventional therapies [6]. Some potential

protein targets are shown in Table 5.1. These sequences could be modified by the Damha group and

protein levels can be quantified by qPCR [7].

Table 5.1: Potential siRNA target genes

Approach Target Gene Description Oncogenesis C-raf [5, 8] - C-raf plays a key role in a mitogen-activated protein kinase

cascade to promote tumor progression

- SiRNA against c-raf was tested by transfection to A549,

PC-3, MDA-MB-231 and SKOV-3 cancer cells with

liposomes

Heat shock protein 90

(HSP90) [9] - HSP90 plays a role in the conformational stabilization and

maturation of mutant oncogenic signalling proteins.

- Knockdown of HSP90 leads to MIF (migration inhibitory

factor) degradation and triggers apoptosis of cancer cells

- In ErbB3 transgenic model of human HER2+ breast

cancer, genetic knockdown delays tumor progression and

prolongs overall survival of mice

- MDA-MB231 and MDA-MB468 cells were cultured to

observe gene knockdown

Her2/neu (HER2) [7] - Overexpression of HER2 gene provides mitogenic signals

to tumour cells to increase growth potential and resistance

to apoptotic factors

- siRNA sequences used against SKBR3, SKOV3, MCF-7

cell lines

Apoptosis Bcl-2 and xIAP [10] - Bcl-2 or xIAP may be responsible for apoptotic resistance

to cytotoxic drugs

- Use of siRNA to downregulate bcl-2 or xIAP enhances the

effects of etoposide and doxorubicin treatments

Tumor-host

interactions

Ras homologous A

(RhoA) and Ras

homologous C (RhoC)

[11]

- RhoA and RhoC promote cell proliferation and cell

invasion

- Aggressiveness of cancer is associated with increased

angiogenesis, which displays RhoA signalling

- siRNA used to inhibit cell proliferation and invasion in

MDA-MB-231 breast cancer cells in vitro

- Inhibited growth and angiogenesis of xenografted MDA-

MB-231 tumors in a nude mouse model

Page 68: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

59

3. Conjugate siRNA or AONs using SPAAC chemistry.

In these studies, the oligonucleotides were conjugated using copper catalyzed azide-alkyne

cycloadditions. The copper was removed with the unreacted antibodies and oligonucleotides during the

purification step. However, the concentration of copper in the purified nanoparticle solution was not

measured. Trace amounts of copper could be quantified by Inductively Coupled Plasma-Atomic Emission

Spectrometry (ICP-AES) or Inductively Coupled Plasma-Mass Spectrometry (ICP-MS).

The strain promoted azide-alkyne reaction provides an alternative strategy to avoid problems of cell

cytotoxicity associated with copper. The Damha group has been working on the synthesis of

dibenzylcyclooctyne terminated oligonucleotides. These sequences could be used for a copper-free

reaction. SPAAC reactions also have the added benefit of reducing the complexity of the click reaction;

the reducing agent and ligand used to protect the oligonucleotide from reactive copper species is no

longer required.

4. Use nanoparticles to encapsulate agents for the improvement of siRNA knockdown.

Previous studies have shown that our polymeric nanoparticle can encapsulate small molecule drugs. The

siRNA immuno-nanoparticle system could make use of the hydrophobic core to load and deliver small

molecules to improve cellular translocation or endosomal escape of siRNA. SiRNA can passively

transmit through nuclear pore complexes from the cytoplasm to the nucleus. However, permeability of the

nuclear membrane can be improved with the delivery of trans-cyclohexane-1,2-diol (TCHD). TCHD is an

amphipathic alcohol that can improve delivery efficiency by 6 times [12, 13]. Alternatively, endosomal

escape can be improved by the encapsulation and delivery of agents like chloroquine. Chloroquine buffers

the endosomal pH and has improved delivery efficiency by 7 times [14]. The encapsulation of small

molecules could strengthen the siRNA effects for complete gene knockdown.

References

[1] J. S. Park, et al., "N-acetyl histidine-conjugated glycol chitosan self-assembled nanoparticles for

intracytoplasmic delivery of drugs: Endocytosis, exocytosis and drug release," Journal of

Controlled Release, vol. 115, pp. 37-45, Sep 2006.

[2] Y. W. Cho, et al., "Polycation gene delivery systems: escape from endosomes to cytosol,"

Journal of Pharmacy and Pharmacology, vol. 55, pp. 721-734, Jun 2003.

[3] J. Panyam, et al., "Rapid endo-lysosomal escape of poly(DL-lactide-co-glycolide) nanoparticles:

implications for drug and gene delivery," Faseb Journal, vol. 16, pp. -, Aug 2002.

[4] H. Wartlick, et al., "Highly specific HER2-mediated cellular uptake of antibody-modified

nanoparticles in tumour cells," Journal of Drug Targeting, vol. 12, pp. 461-471, 2004.

Page 69: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

60

[5] M. Masiero, et al., "RNA interference: Implications for cancer treatment," Molecular Aspects of

Medicine, vol. 28, pp. 143-166, Feb 2007.

[6] J. F. Guo, et al., "Can non-viral technologies knockdown the barriers to siRNA delivery and

achieve the next generation of cancer therapeutics?," Biotechnology Advances, vol. 29, pp. 402-

417, Jul-Aug 2011.

[7] A. Choudhury, et al., "Small interfering RNA (siRNA) inhibits the expression of the Her2/neu

gene, upregulates HLA class I and induces apoptosis of Her2/neu positive tumor cell lines,"

International Journal of Cancer, vol. 108, pp. 71-7, Jan 1 2004.

[8] P. Y. Chien, et al., "Novel cationic cardiolipin analogue-based liposome for efficient DNA and

small interfering RNA delivery in vitro and in vivo," Cancer Gene Therapy, vol. 12, pp. 321-328,

Mar 2005.

[9] R. Schulz, et al., "Inhibiting the HSP90 chaperone destabilizes macrophage migration inhibitory

factor and thereby inhibits breast tumor progression," J Exp Med, vol. 209, pp. 275-89, Feb 13

2012.

[10] R. T. Lima, et al., "Specific downregulation of bcl-2 and xIAP by RNAi enhances the effects of

chemotherapeutic agents in MCF-7 human breast cancer cells," Cancer Gene Therapy, vol. 11,

pp. 309-316, May 2004.

[11] J. Y. Pille, et al., "Anti-RhoA and anti-RhoC siRNAs inhibit the proliferation and invasiveness of

MDA-MB-231 breast cancer cells in vitro and in vivo," Molecular Therapy, vol. 11, pp. 267-74,

Feb 2005.

[12] K. L. Douglas, "Toward development of artificial viruses for gene therapy: A comparative

evaluation of viral and non-viral transfection," Biotechnology Progress, vol. 24, pp. 871-883, Jul-

Aug 2008.

[13] R. E. Vandenbroucke, et al., "Nuclear accumulation of plasmid DNA can be enhanced by non-

selective gating of the nuclear pore," Nucleic Acids Research, vol. 35, Jun 2007.

[14] J. Cheng, et al., "Structure - Function correlation of chloroquine and analogues as transgene

expression enhancers in nonviral gene delivery," Journal of Medicinal Chemistry, vol. 49, pp.

6522-6531, Nov 2 2006.

Page 70: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

61

Appendix A: Supplemental Information

A1. Characterization of NH2-PEG-azide

Figure A 1. Characterization of NH2-PEG-Azide by FTIR. Terminal azide group (2100cm-1

), terminal

amine (3389 cm-1

), C–H alkane (2888 cm-1

), C=O amide (1692 cm-1

), and C–O ether (1101 cm-1

)

N–H

terminal amine

(3300-3500 cm-1

)

N≡N, azide

(2100 cm-1

)

C–O, ether

(1101 cm-1

)

C=O, amide

(1692 cm-1

)

Page 71: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

62

A2. Characterization of NH2-PEG-furan

(a)

Page 72: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

63

(b)

Figure A 2. Characterization of NH2-PEG-furan by 1H NMR (a) before boc deprotection.

1H NMR

(CDCl3, 400 MHz): δ 1.44 (s, boc protecting group), 2.32 (t, J=14 Hz , –CH2–), 3.64 (m, –OCH2CH2–) ,

4.42 (s, –CH2-furan), 6.21 (d, J= 3Hz, furan CH-3), 6.30 (dd, J=5 Hz, furan CH-4) and 7.33 (d, J=3 Hz,

furan CH-5). (b) NH2-PEG-furan after deprotection. 1H NMR (CDCl3, 400 MHz): δ 2.33 (t, J=14 Hz , –

CH2–), 3.64 (m, –OCH2CH2–) , 4.43 (s, –CH2-furan), 6.22 (d, J= 3Hz, furan CH-3), 6.30 (t, J=5 Hz,

furan CH-4) and 7.34 (d, J=3 Hz, furan CH-5)

Page 73: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

64

A3. Characterization of P(LA-co-TMCC)

(a)

Page 74: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

65

(b)

Figure A 3. Characterization of P(LA-co-TMCC) by 1H NMR: (a) P(LA-co-TMCC-Bn) shows presence

of the benzyl group. 1H NMR (CDCl3, 400MHz): δ 1.23 (br s, CH3 from TMCC), 1.58 (m, CH3 from LA),

4.31 (br s, CH2 from TMCC), 4.98 (s, CH-Bn), 7.33 (m, benzyl Ar) and 8.02 (m, pyrene). (b) Absence of

the benzyl group indicates complete deprotection. Due to the large polymer size, the pyrene group is

indistinguishable at the polymer concentration sampled. 1H NMR (CDCl3, 400MHz): δ 1.19 (br s, CH3

from TMCC), 1.51 (m, CH3 from LA), and 4.26 (br s, CH2 from TMCC).

Page 75: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

66

A4. Characterization of P(LA-co-TMCC)-g-PEG

(a)

Page 76: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

67

(b)

Figure A 4. Characterization of P(LA-co-TMCC)-g-PEG-X by 1H NMR: (a) Azide-functionalized PEG

grafted to the polymer backbone. 1H NMR (CDCl3, 400 MHz): δ 1.25 (br s, CH3 from TMCC), 1.58 (m,

CH3 from LA), 3.59-3.69 (m, –OCH2CH2–), 4.35 (br s, CH2 from TMCC) and 5.17 (m, CH from LA).

(b) Furan-functionalized PEG grafted to the polymer backbone. 1H NMR (CDCl3, 400 MHz): δ 1.22 (br s,

CH3 from TMCC), 1.57 (m, CH3 from LA), 3.59-3.69 (m, –OCH2CH2–), 4.37 (br s, CH2 from TMCC)

and 5.15 (m, CH from LA). Integrated peak areas of the PEG methylene to TMCC methylene was used

to determine the grafting efficiency: 0.7 PEG-furan or 0.9 PEG-azide per backbone.

Page 77: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

68

A5. DA Reaction with NH2-PEG-Fu

Figure A 5. DA Reaction with NH2-PEG-Fu. The decrease of the characteristic peaks of maleimide C=C

(1466 cm-1

) and the appearance of the C=C Diels-Alder adduct peak (1459 cm-1

) indicate that the Diels

Alder reaction has occurred.

A6. SPAAC Reaction with NH2-PEG-N3

Figure A 6. SPAAC Reaction with NH2-PEG-N3 and Alexa 488-DBCO. Decrease in the azide peak

(2109 cm-1

) indicates that the SPAAC reaction has occurred between the dibenzylcyclooctyne and azide.

C–H, alkane

(2888 cm-1

)

N≡N, azide

(2109 cm-1

)

C–O, ether

(1101 cm-1

)

C–H, alkane

(2887.8 cm-1

) C–O, ether

(1101.3 cm-1

)

C=C, furan

(1466 cm-1

)

C=C, D-A

adduct

(1459 cm-1

)

Page 78: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

69

A7. Size Exclusion Column Purification of Micelles

Figure A 7. Size exclusion column purification of mixed micelles with Alexa 647-MI and Alexa 488-

DBCO. Separation was completed using a Sephadex G-25 column. Fluorescence of the Alexa 647 (ex.

640 nm, em. 675 nm) and Alexa 488 (ex. 490 nm, em. 525 nm) was used to quantify the conjugation

efficiency. The concentration of the nanoparticles was measured by absorbance at 260 nm. Fractions 6

and 7 contain the dual labelled micelle.

Page 79: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

70

A8. Confocal Images of SKOV-3luc controls

(a)

(b)

Figure A 8. Confocal images of SKOV-3luc cells controls. (a) Cells without nanoparticles show neither

background signal of the Alexa Fluor 647 nor Alexa Fluor 488; (b) SKOV-3luc cells and Trastuzumab-

nanoparticles show no background signal of the Alexa Fluor 647 used to stain the FLAG (i) Cell nuclei

are stained with DAPI (blue); (ii) FLAG peptide (Alexa Fluor 647, purple); (ii) trastuzumab (Alexa Fluor

488, green); (iv) Overlay of all channels.

(i) (ii)

(iii) (iv)

(i) (ii)

(iii) (iv)

Page 80: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

71

A9. Click Chemistry Reactions

Table A1. Click Chemistry Reactions. In this system, the micelle was functionalized with azide and furan

groups. Potential binding partners include: maleimides, acetylenes and strained alkynes. Our studies used

the maleimide and strained alkyne.

Azide

Furan

Maleimide

No reaction DA [1]

Acetylene

CuAAC [2]

+ Cu

No reactiona

Strained alkyne

SPAAC [3]

DA

aAlthough there is no reaction between an acetylene group and furan, Martin-Matute, et al. have shown that the

addition of a PtCl2 catalyst will result in a Diels Alder reaction. The electrophilic catalyst forms a Pt carbine

intermediate which initiates a nucleophilic attack of the furan.

[1] M. Shi, et al., "Immuno-polymeric nanoparticles by Diels-Alder chemistry," Angewandte Chemie-International

Edition, vol. 46, pp. 6126-6131, 2007.

[2] M. Meldal and C. W. Tornoe, "Cu-catalyzed azide-alkyne cycloaddition," Chemical Reviews, vol. 108, pp. 2952-

3015, Aug 2008.

[3] G. J. Boons, et al., "Surface Modification of Polymeric Micelles by Strain-Promoted Alkyne-Azide

Cycloadditions," Chemistry-a European Journal, vol. 16, pp. 13360-13366, 2010.

Page 81: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

72

Appendix B: siRNA and AON Synthesis and Characterization

B1. Synthesis of siRNA and AON sequences

All oligonucleotides (ONs) were synthesized on an Applied Biosystems (ABI) 3400 DNA synthesizer at

1µmol scale. Unylink CPG (ChemGenes) was used for the syntheses of all ONs, except those modified

with 3′ alkyne functionality, which was introduced using a 3′ alkyne-modifier serinol CPG, which is

commercially available from Glen Research (Figure S2). 2′F-ANA, 2′F-RNA, Cyanine 5 (Glen Research)

and RNA phosphoramidites were prepared as 0.15M solutions in dry acetonitrile (ACN), DNA as 0.1M.

RNA amidites were 5′-DMT, 2′-TBDMS protected, and base protection was benzoyl (A), i-Bu (G) or

acetyl (C). 5-ethylthiotetrazole (0.25M in ACN, ChemGenes) was used to activate the phosphoramidites

from coupling. Detritylations were accomplished with 3% trichloroacetic acid in dichloromethane for

110s. Capping of failure sequences was achieved with acetic anhydride in tetrahydrofuran, and 16% N-

methylimidazole in tetrahydrofuran. Oxidations were done using 0.1M I2 in 1:2:10

pyridine:water:tetrahydrofuran, except for those following Cyanine 5 addition to ON 5′ termini, which

was accomplished with 0.02M I2 instead. AONs containing phosphorothioate linkages were sulfurized

using a 0.1M solution of Xanthane Hydride (TCI) in 1:1 vol/vol pyridine/ACN (anhydrous). The

sulfurization step was allowed to proceed for 2.5min, with new sulfurization reagent added after 1.25min.

Phosphoramidite coupling times were 600s for 2′F-ANA, 2′F-RNA, and RNA, with the exception of the

guanosine phosphoramidiates, which were allowed to couple for 900s. DNA coupling times were 110s,

and 270s for guanosine. Cy5 coupling times were extended to 20min.

B2. Cleavage and Purification

Base deprotection and cleavage from the solid support was accomplished with 1mL of 3:1 aqueous

NH4OH:EtOH for 48 hours at room temperature (for modified sequences), after which samples were

chilled on dry ice for 15min and subsequently lyophilised to dryness in a speedvac concentrator (Savant).

Standard RNA sequences were base deprotected with 1mL of 40% (w/v) aqueous methylamine at 65oC

for 10min, chilled on dry ice, and lyophilized to dryness. 2′-TBDMS protecting groups were removed

with 250µL neat triethylamine trihydrofluoride (TreatHF) for 48hr at room temperature (modified

sequences), or with 300µL of TreatHF/N-methyl pyrollidinone (NMP)/triethyl amine solution (prepared

by adding 0.75mL NMP, 1mL TEA, and 1.5mL TreatHF together at 65oC) at 65

oC for 3 hours. Following

desilylation, ONs were precipitated by the addition of 25µL 3M NaOAc and 1mL of n-butanol followed

by cooling on dry ice. The ON pellets were lyophilized to dryness.

Page 82: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

73

Oligonucleotides were desalted on NAP-25 sephadex size exclusion columns (GE Healthcare) according

to manufacturer protocol to prepare for HPLC purification. ONs were purified by either anion exchange

or reverse phase HPLC, on either a Waters 1525 or Agilent 1200 HPLC, using a Varian Pursuit 5

semipreparative reverse phase C18 column, or a Waters Protein Pak DEAE 5PW semipreparative anion

exchange column. For reverse phase purifications, a stationary phase of 100mM triethylammonium

acetate in water with 5% ACN (pH7) and a mobile phase of HPLC-grade ACN (Sigma) were used (with a

gradient of 0%-35% over 30min). Purified ONs were lyophilized to dryness, which also served to remove

excess triethylammonium acetate salts. For anion exchange purifications, a stationary phase of milliQ

H2O and a mobile phase of 1M LiClO4 in milliQ water was used (with a gradient of 0%-38% over

38min). Following anion exchange purification, excess LiClO4 salt was removed using a second desalting

with NAP-25 sephadex size exclusion columns (GE Healthcare) according to manufacturer protocol.

B3. Quantitation and characterization

All oligonucleotides were quantitated by UV (extinction coefficients were calculated using the online IDT

OligoAnalyzer tool (www.idtdna.com/analyzer/Applications/OligoAnalyzer); 2′F-ANA extinction

coefficients were calculated using DNA values). Oligonucleotides were characterized by LC-MS on a

Waters Q-TOF2 using an ESI NanoSpray source. A CapLC (Waters) with a C18 trap column was used

for LC prior to injections. Thermal denaturation measurements were performed for select siRNA

sequences on a Cary 300 UV/Vis spectrophotometer, by ramping from 15oC- 95

oC at a rate of 1

oC/min

using common ON buffer (140mM KCl, 1mM MgCl2, 5mM NaHPO4, pH 7.2). The Tm for the

unmodified 21mer siRNA targeting sequence is 63.8oC, well above the temperatures encountered during

the click reaction to attach siRNAs to the nanoparticles. To determine if ON backbone damage could be

expected following the copper-mediated click reaction to attach ONs to the nanoparticles, a Cy5-labelled

21mer poly-dT ON was treated with Sodium L-ascorbate, CuSO4.5H2O, and tris[(1-benzyl-1H-1,2,3-

triazol-4-yl)methyl]amine (TBTA), and followed at regular time points over a 30hr period. Strand

integrity was observed using 24% PAGE, and bands were visualized by both UV shadowing and using

Stains-all reagent (Sigma) in isopropanol (50mL formamide, 125mL isopropanol, 325mL water, soak gel

overnight). No strand cleavage was detected.

B4. Annealing of oligonucleotides.

Equimolar amounts of the sense and antisense strands (or AON and RNA complement strands for

antisense gene silencing) of each oligonucleotide duplex were combined in annealing buffer (140 mM

Page 83: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

74

KCl, 1mM MgCl2, 5 mM NaHPO4, pH 7.2) for a final concentration of 28 µM for each strand. The vial

was heated at 90°C for 1 min, then cooled to room temperature over an hour. The UV visible light

spectrophotometer was used to confirm annealing of the two strands by measuring absorbance at 260 nm.

References

[1] M. Shi, et al., "Immuno-polymeric nanoparticles by Diels-Alder chemistry," Angewandte Chemie-

International Edition, vol. 46, pp. 6126-6131, 2007.

[2] M. Meldal and C. W. Tornoe, "Cu-catalyzed azide-alkyne cycloaddition," Chemical Reviews, vol.

108, pp. 2952-3015, Aug 2008.

[3] G. J. Boons, et al., "Surface Modification of Polymeric Micelles by Strain-Promoted Alkyne-Azide

Cycloadditions," Chemistry-a European Journal, vol. 16, pp. 13360-13366, 2010.

[4] B. Martin-Matute, et al., "Pt-II-catalyzed intramolecular reaction of furans with alkynes," Angewandte

Chemie-International Edition, vol. 40, pp. 4754, 2001.

Page 84: Polymeric Micelles for SiRNA and AON Delivery · Polymeric Micelles for SiRNA and AON Delivery Dianna Chan Master’s of Applied Science Department of Chemical Engineering and Applied

75

Appendix C: Statistical Analysis

C1. SiRNA Delivery

In Figure 3.2, lower case letters were grouped bars with no statistical difference (one-way ANOVA,

p>0.05). Statistics were also completed between groups (labelled ‘a’, ‘b’, ‘c’ and ‘d’) and are summarized

in Table C1.

Table C1. P-values calculated by one-way ANOVA analysis of luciferase levels of SKOV-3luc cells

treated with chemically modified and unmodified siRNA with immuno-nanoparticles or Lipofectamine.

Label a b c d

Average Luciferase Level 0.1834 0.2690 0.5389 0.8693

Standard Deviation 0.0794 0.1240 0.1294 0.1535

p-value vs. a 0.007 < 0.001 < 0.001

b < 0.001 < 0.001

c < 0.001

d

C2. AON Delivery

In Figure 3.3, lower case letters were used to group bars with no statistical difference (one-way ANOVA,

p>0.05). Statistics were also completed between groups (labelled ‘a’, ‘b’, ‘c’ and ‘d’) and are summarized

in Table C2.

Table C2. P-values calculated by one-way ANOVA analysis of luciferase levels of SKOV-3luc cells

treated with chemically modified and unmodified siRNA with immuno-nanoparticles or Lipofectamine.

Label a b c d

Average Luciferase Level 0.6225 0.0589 0.0927 0.7984

Standard Deviation 0.1281 0.0332 0.0295 0.1265

p-value vs. a < 0.001 < 0.001 < 0.001

b 0.005 < 0.001

c < 0.001

d