Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic...

206
1 Evaluating the genotoxic potential of oligonucleotide pharmaceuticals Reshat Reshat 2012 Thesis submitted for the Degree of Doctor of Philosophy Department of Biomolecular Medicine Division of Surgery and Cancer, Faculty of Medicine Imperial College London SW7 2AZ UK

Transcript of Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic...

Page 1: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

1

Evaluating the genotoxic potential of

oligonucleotide pharmaceuticals

Reshat Reshat

2012

Thesis submitted for the Degree of Doctor of Philosophy

Department of Biomolecular Medicine

Division of Surgery and Cancer,

Faculty of Medicine

Imperial College London

SW7 2AZ

UK

Page 2: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

2

Abstract

According to regulatory guidelines, routine genotoxicity tests are not appropriate for

biotechnology derived pharmaceuticals, including oligonucleotide based therapeutics, as

they are not expected to interact with genomic DNA. However, reports of oligonucleotides

capable of binding duplex DNA in a sequence specific manner to form triple-helix (triplex)

or displacement-loop (D-loop) structures that in turn cause mutation have raised concern.

The European Medicines Agency (EMA) has questioned the capability of antisense

oligonucleotide (ASO) therapeutics to form such structures at genomic DNA.

Additionally, concern has been expressed regarding the fate of chemically modified

ASO degradation products (nucleotide analogues). It is well established that non-canonical

antiretroviral nucleoside analogues, employed in antiretroviral therapy, result in gross

chromosome aberrations following incorporation into genomic DNA.

This study has addressed these concerns by evaluating the genotoxic potential of a

triplex forming oligonucleotide (TFO) and a D-loop forming ASO targeting genomic DNA.

Furthermore, the incorporation efficiency and genotoxicity of nucleotide analogues derived

from ASO degradation was investigated.

Data presented here demonstrate a TFO targeting genomic DNA was not capable

of inducing mutation above the detection limit of this assay. However, a biologically active

ASO molecule induced sequence specific mutation ~4.4 fold above control in a system

where RAD51 protein expression was induced. Additionally, DNA polymerase was capable

of incorporating various ASO derived nucleotide analogues into a primed DNA template

with reduced efficiency. Treatment with phosphorothioate nucleotide analogue, one of the

most common chemical modifications used in ASO design, induced mutation ~100 fold

above control.

To conclude, ASO and their putative degradation products appeared to be capable

of off-target mutagenesis providing favourable conditions were met. However, as

genotoxicity data has been presented for a single ASO and nucleotide analogue, it seems

plausible to suggest this work can provide a foundation to test future ASO therapeutics

and putative degradation products.

Page 3: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

3

Statement of originality

I certify that the research presented within this thesis is a product of my own work and that

ideas, quotations or work from other people are fully acknowledged. This thesis has not

been submitted for any other award.

The copyright of this thesis rests with the author and no quotation from it or information

derived from it may be published without the prior written consent of the author.

Page 4: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

4

Acknowledgements

To begin, I want to thank both of my supervisors Prof. Nigel J. Gooderham and Dr.

Catherine Priestley who tirelessly read abstracts, manuscripts and even my thesis. It

seems only yesterday I sat across you both in Nigel’s office for my PhD interview. My

passion and diligence towards my project is a testament to the brilliance of Nigel and

Catherine as supervisors. Without you both, I would not be writing this thesis which has

opened avenues of opportunities for me. It seems almost insufficient for me to simply say,

thank you, from the bottom of my heart.

I also want to acknowledge the help and guidance from Dr. Mike O’Donovan and Mick

Fellows as well as the entire Gen.Tox team at AstraZeneca.

I especially thank my mother, father, brothers, sisters, grandparents and friends for their

limitless support, advice and comfort throughout my education. I also want to apologise for

the occasions I have been unavailable, but I can assure you, I was working.

A special acknowledgement goes to my girlfriend Tabitha Jones and her family. Without

Tabitha’s love, support and friendship I could not have completed this PhD.

Although I have gained so much throughout my PhD, one of the most valuable assets I

have taken from Imperial are the friendships I have made. I hope that with time, our

friendships grow even stronger and we stay in touch. Corrinne, Costas, Mike, Cancy,

Rhiannon, Nurul, Wayne and Abdullah, I thank you all for the “mini-symposia” we held by

our desks. Those lively debates are priceless memories, and Costas, I look forward to

opening a laboratory together in Cyprus. I await your call.

My advice to future PhD students that read this thesis is regarding my times of low. To get

me by, I remembered the following saying, “you either want a PhD or you don’t” (Nigel J.

Gooderham). The way I interpreted this was to simply crack on because you only have

one shot. Good luck.

Page 5: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

5

List of abbreviations

Abbreviation Definition

µg Microgram

µL Microliters

µM Micromolar

µm micrometer

2’-OMe 2’-O-methyl-ribose

2’OMe-ATP ATP modified to replace 2’OH group on ribose moiety with

O-methyl

4NQO 4-Nitroquinoline 1-oxide

5Me-dCTP dCTP modified to contain a methyl group in position 5 of cytidine

6FAM 6-carboxyfluorescein

6-TG 6-thioguanine

AD3-HPRTPM Antisense oligonucleotide with sequence homology to the

ADAMTS3 and HPRT locus

ADAMTS3 A disintegrin and metalloproteinase with thrombospondin type 1,

motif 3

AIDS Acquired immunodeficiency syndrome

ANOVA Analysis of variance

ASO Antisense oligonucleotide

ATP adenosine triphosphate

AZT 3’-azido-2’, 3’-dideoxythymidine

BCA Bicinchoninic acid

bp Base pairs

BSA Bovine serum albumin

cDNA Complementary DNA

CHO Chinese hamster ovary

CMV Cytomegalovirus

dAMP 2’-deoxyadenosine monophosphate

dAMPαS dAMP modified to replace a non-bridging oxygen with sulphur on

the α-phosphate group

dATP 2’-deoxyadenosine triphosphate

dATPαS dATP modified to replace a non-bridging oxygen with sulphur on

the α-phosphate group

dCTP 2’-deoxycytidine triphosphate

dGTP 2’-deoxyguanosine triphosphate

Didanosone 2’,3’-dideoxyinosine

D-loop Displacement loop

Dloop27 D-loop oligonucleotide targeting the TFO27 HPRT target

sequence

DMD Duchenne muscular dystrophy

DMSO Dimethylsulphoxide

DNA Deoxyribonucleic acid

Page 6: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

6

dNTP Deoxyribonucleoside triphosphate

DSBs Double strand breaks

dTTP 2’-deoxythymidine triphosphate

EBI European Bioinformatics Institute

EDTA Ethylenediaminetetraacetic acid

eGFP Enhanced green fluorescent protein

EMA European Medicines Agency

EMS Ethyl methanesulphonate

EMSA Electrophoretic mobility shift assay

FDA Food and Drug Administration

GA motif Purine motif

Ganciclovir 9-[(1,3-dihydroxy-2-propoxy)-methyl]-guanine

GAPDH Glyceraldehyde 3-phosphate dehydrogenase

gDNA Genomic DNA

GFP Green fluorescent protein

GT motif Mixed motif

h hour(s)

HAART Highly active antiretroviral therapy

HIFBS Heat inactivated foetal bovine serum

HIHS Heat inactivated horse serum

HIV-1 Human immunodeficiency virus type 1

HPRT Hypoxanthine-guanine phosphoribosyltransferase

HR Homologous recombination

HRP Horse radish peroxidise

ICH International Conference on Harmonisation of Technical

Requirements for Registration of Pharmaceuticals for Human Use

Kd Dissociation constant

Lamivudine 2’, 3’-dideoxy-3’-thiacytidine

LNA Locked nucleic acid

MDM2 Murine double minute 2

mins Minutes

mL Millilitres

MLA Mouse lymphoma assay

MLH-1 Mut L homologue 1

mM Millimolar

MMR Mismatch repair

MMS Methyl methanesulphonate

mRNA messenger RNA

MSH2 Mut S homologue 2

NCBI-BLAST National Center for Biotechnology Information - Basic Local

Alignment Search Tool

NER Nucleotide excision repair

ng Nanogram

NHEJ Non homologous end joining

Page 7: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

7

nM Nanomolar

nm Nanometer

OECD Organisation for Economic Co-operation and Development

PBS Phosphate buffered saline

PCR Polymerase chain reaction

PKC-α Protein kinase C-α

PNA Peptide nucleic acid

poly-rA Polyriboadenylic acid

poly-rU Polyribouradylic acid

Pso-SCR27-biot SCR27 conjugated 5’ to psoralen and 3’ biotin

Pso-TFO27-biot TFO27 conjugated 5’ to psoralen and 3’ biotin

PVDF Polyvinylidine fluoride

R0 RPMI 1640 without serum (incomplete media)

R10HIFBS RPMI 1640 media containing 10%w/v HIFBS (complete media)

R10HIHS RPMI 1640 media containing 10%w/v HIHS (complete media)

RFLP Restriction fragment length polymorphism

RISC complex RNA-induced silencing (RISC) complex

RNA Ribonucleic acid

RNAi RNA interference

RPD Relative population doubling

RSG Relative suspension growth

RTG Relative total growth

SCE Sister chromatid exchange

SCR27 Control oligonucleotide used in triplex studies

SD Standard deviation

SDS Sodium dodecyl sulphate

siRNA Small interfering RNA

SV40 Simian virus 40

TA (Primer) annealing temperature

TBE Tris, boric acid and EDTA

TC motif Pyrimidine motif

TFO Triple-helix forming oligonucleotide

TFO27 TFO targeting exon 3 of the HPRT locus

TFT Trifluorothymidine

TK Thymidine kinase

TM (Primer) melting temperature

TNE Targeted nucleotide exchange

Triplex Triple-helix

V Volts

Vmax Maximum speed of incorporation

Page 8: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

8

Table of contents

List of figures ................................................................................................................... 12

List of tables ..................................................................................................................... 15

Chapter 1. General introduction ..................................................................................... 16

1.1. Introduction ............................................................................................................. 16

1.2. Antisense oligonucleotides (ASO) ........................................................................... 17

1.2.1. Chemical modifications ..................................................................................... 17

1.2.2. Biological activity of ASO .................................................................................. 21

1.2.3. ASO mechanism of action ................................................................................ 24

1.2.4. ASO used in clinical trials ................................................................................. 26

1.3. Concerns with oligonucleotide based pharmaceuticals ........................................... 29

1.3.1. Triplex forming oligonucleotides (TFO) ............................................................. 30

1.3.1.1. Mutagenicity of TFO ................................................................................... 33

1.3.2. D-loop forming oligonucleotides........................................................................ 35

1.3.2.1. Mutagenicity of D-loop forming oligonucleotides ........................................ 36

1.3.3. Nucleotide analogues ....................................................................................... 44

1.3.3.1. Mutagenicity of non-canonical nucleotide analogues ................................. 46

1.3.3.2. Mutagenicity of canonical nucleotides ........................................................ 49

1.4. Aims and objectives ................................................................................................ 52

Chapter 2. Materials and Methods .................................................................................. 54

2.1. Oligonucleotides ...................................................................................................... 54

2.1.1. Triple-helix forming oligonucleotide design ....................................................... 54

2.1.2. Displacement loop/ antisense oligonucleotide design ....................................... 55

2.1.3. Serum stability of oligonucleotides .................................................................... 56

2.1.4. Oligonucleotide third strand binding assay ....................................................... 58

2.1.5. Detecting triplex formation at genomic DNA ..................................................... 58

2.1.6. DNase I protection assay .................................................................................. 61

2.1.7. Quadruplex forming ability of SupFG1 .............................................................. 62

2.1.8. Primer extension assay .................................................................................... 66

2.2. General cell culture ................................................................................................. 68

2.2.1. Human lymphoblastoid TK6 cells ...................................................................... 68

Page 9: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

9

2.2.2. Human colon epithelial HCT116 cells ............................................................... 70

2.2.3. Genomic DNA extraction from cultured cells .................................................... 70

2.2.4. RNA extraction.................................................................................................. 71

2.2.5. Protein extraction and quantification ................................................................. 72

2.3. Cell treatments ........................................................................................................ 73

2.3.1. MSH2 protein knockdown in TK6 cells using Silencer Select siRNA ................ 73

2.3.2. Induction of RAD51 protein expression in TK6 cells ......................................... 74

2.3.3 Oligonucleotide transfection efficiency using siPORT NeoFX and NTER in TK6

cells ............................................................................................................................ 74

2.3.4. Treatment of TK6 cells with oligonucleotide ..................................................... 75

2.3.5. Treatment of TK6 cells with nucleosides and nucleotides ................................ 77

2.4. HPRT and TK forward mutation assays .................................................................. 78

2.4.1. Determining the TK mutant frequency (Day 3) ................................................. 78

2.4.2. Determining the HPRT mutant frequency (Day 7) ............................................ 79

2.4.3. The in vitro micronucleus assay ........................................................................ 80

2.4.4. Treatment of HCT116 cells with oligonucleotide ............................................... 81

2.4.5. Cytotoxicity following treatment of HCT116 cells .............................................. 81

2.4.6. Determining the HPRT mutant frequency following treatment of HCT116 cells 81

2.5. Polymerase Chain Reaction (PCR) ......................................................................... 82

2.5.1. First strand cDNA synthesis from RNA extracts ............................................... 82

2.5.2. PCR primer design ........................................................................................... 82

2.5.3. PCR amplification of genomic DNA and cDNA ................................................. 83

2.5.4. Agarose gel electrophoresis ............................................................................. 83

2.5.5. Restriction enzyme digestion of PCR products ................................................. 84

2.5.6. PCR product purification and DNA sequencing ................................................ 84

2.6. Immunoblot ............................................................................................................. 86

2.6.1. Preparation and electrophoresis of samples ..................................................... 86

2.6.2. Transfer of protein ............................................................................................ 87

2.6.3. Immunoblotting ................................................................................................. 87

Chapter 3. The Genotoxicity of a triplex forming oligonucleotide targeting genomic

DNA ................................................................................................................................... 90

3.1. Introduction ............................................................................................................. 90

3.2. Experimental outline ................................................................................................ 92

3.3. Methods .................................................................................................................. 93

Page 10: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

10

3.4. Results .................................................................................................................... 94

3.4.1. High affinity triplex formation............................................................................. 94

3.4.2. Transfection efficiency ...................................................................................... 96

3.4.3. Evaluating the genotoxic potential of a TFO at genomic DNA .......................... 98

3.4.4. Serum stability of oligonucleotides .................................................................. 104

3.4.5. Human TK6 cells contain the wild-type HPRT sequence ................................ 105

3.4.6. Method development to detect triplex formation at genomic DNA .................. 107

3.4.7. The previously employed SupFG1 triplex target sequence is capable of DNA

quadruplex formation ................................................................................................ 112

3.5. Discussion ............................................................................................................. 115

Chapter 4. The Genotoxicity of an antisense oligonucleotide via targeted nucleotide

exchange ........................................................................................................................ 120

4.1. Introduction ........................................................................................................... 120

4.2. Experimental outline .............................................................................................. 121

4.3. Methods ................................................................................................................ 123

4.4. Results .................................................................................................................. 124

4.4.1. Genotoxicity of Dloop27 .................................................................................. 124

4.4.2. Biological activity of AD3-hprtPM .................................................................... 126

4.4.3. The of role of mismatch repair in antisense oligonucleotide genotoxicity ....... 128

4.4.4. The of role of homologous recombination repair in antisense oligonucleotide

genotoxicity ............................................................................................................... 137

4.5. Discussion ............................................................................................................. 145

Chapter 5. Incorporation and genotoxicity of oligonucleotide degradation products

into the genome ............................................................................................................. 149

5.1. Introduction ........................................................................................................... 149

5.2. Experimental outline .............................................................................................. 151

5.3. Methods ................................................................................................................ 152

5.4. Results .................................................................................................................. 153

5.4.1. In vitro incorporation efficiency of nucleotide analogues ................................. 153

5.4.2. Genotoxic fate of canonical nucleotides ......................................................... 158

5.4.3. Genotoxic fate of a phosphorothioate nucleotide analogue ............................ 162

5.5. Discussion ............................................................................................................. 168

Chapter 6. General discussion and conclusions ........................................................ 173

Page 11: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

11

6.1. Future work ........................................................................................................... 180

References ..................................................................................................................... 183

Appendix 1. Buffers and reagents ................................................................................ 205

Publications ................................................................................................................... 206

Page 12: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

12

List of figures

Figure 1.1. Chemical modifications used in ASO design................................................... 20

Figure 1.2. Exon skipping by ASO targeting the DMD pre-mRNA. ................................... 24

Figure 1.3. ASO mechanism of action............................................................................... 26

Figure 1.4. DNA triplex formation. ..................................................................................... 31

Figure 1.5. Hoogsteen hydrogen bonding between nucleotides of TFO and duplex DNA.32

Figure 1.6. Classification of TFO. ...................................................................................... 33

Figure 1.7. Putative mechanism of TNE by D-loop forming oligonucleotides. ................... 43

Figure 1.8. Chemical structure of antiretroviral nucleoside analogues used in HAART .... 45

Figure 2.1. Overview of the methodology used to detect triplex formation within genomic

DNA. .................................................................................................................................. 61

Figure 3.1. DNA quadruplex formation. ............................................................................. 92

Figure 3.2. Investigating the triplex forming capability of TFO27 and SCR27 ................... 95

Figure 3.3. Investigating the capability of oligonucleotides to protect duplex DNA from

DNase I cleavage. ............................................................................................................. 95

Figure 3.4. Evaluating the transfection efficiency of TK6 cells using siPORT NeoFX and

NTER. ................................................................................................................................ 97

Figure 3.5. Investigating the genotoxic effect of 1μM oligonucleotide treating 3x106 human

TK6 cells. ......................................................................................................................... 100

Figure 3.6. Investigating the genotoxic effect of 1μM, 2μM and 5μM oligonucleotide

treating 3x106 human TK6 cells. ...................................................................................... 100

Figure 3.7. Investigating the genotoxic effect of 1μM, 2μM and 5μM oligonucleotide

treating 10x106 human TK6 cells using a regulatory acceptable mutation assay ............ 102

Figure 3.8. Investigating the genotoxic effect of TFO27 in a regulatory acceptable

mutation assay treating 10x106 TK6 cells. ....................................................................... 102

Figure 3.9. Investigating the genotoxic effect of TFO27 up to 9.8μM in a regulatory

acceptable mutation assay treating 10x106 TK6 cells. ..................................................... 103

Figure 3.10. The serum stability of oligonucleotides. ...................................................... 105

Figure 3.11. Validating the putative TFO27 target sequence in exon 3 of the HPRT locus

in human lymphoblastoid TK6 cells. ................................................................................ 106

Figure 3.12. Optimising PCR conditions for triplex detection. ......................................... 108

Page 13: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

13

Figure 3.13. Detecting triplex formation and optimising streptavidin bead concentration.

......................................................................................................................................... 109

Figure 3.14. Optimising genomic DNA purification. ......................................................... 110

Figure 3.15. Optimising purification of genomic DNA using the Qiagen DNA extraction

column.. ........................................................................................................................... 111

Figure 3.16. Investigating the quadruplex forming ability of the SupFG1 system. ........... 113

Figure 3.17. Confirmation of DNA quadruplex formation in the SupFG1 duplex. ............ 114

Figure 4.1. The Dloop27 target sequence in exon 3 of the HPRT locus. ........................ 123

Figure 4.2. The AD3-HPRTPM target sequence in exon 3 of the HPRT locus. .............. 123

Figure 4.3. Investigating the mutagenic effect of Dloop27 at the HPRT locus after treating

10x106 human lymphoblastoid TK6 cells. ........................................................................ 125

Figure 4.4. Evaluating the clastogenic activity of Dloop27 in human lymphoblastoid TK6

cells using the in vitro micronucleus assay.. .................................................................... 125

Figure 4.5. The antisense activity of AD3-HPRTPM. ...................................................... 127

Figure 4.6. Investigating the mutagenic effect of an ASO containing a single mismatched

base. ................................................................................................................................ 129

Figure 4.7. Quantitation of MSH2 knockdown in human TK6 cells treated with (S) or

without (U) 10nM Silencer Select siRNA for 24h. ............................................................ 130

Figure 4.8. Optimising MSH2 knockdown using Silencer Select siRNA. ......................... 130

Figure 4.9. Investigating the genotoxic effect of AD3-HPRTPM using human TK6 cells

pre-treated with MSH2 Silencer Select siRNA. ................................................................ 131

Figure 4.10. Investigating the genotoxic effect of AD3-HPRTPM using human TK6

cells.................................................................................................................................. 132

Figure 4.11. Mbo I restriction enzyme digestion of PCR products. ................................. 134

Figure 4.12. Investigating the genotoxic effect of Val-HPRTPM using human TK6 cells.136

Figure 4.13. Investigating the genotoxic effect of AD3-HPRTPM and Val-HPRTPM in a

mismatch repair deficient cell line. ................................................................................... 136

Figure 4.14. Inducing the homologous recombination repair pathway. ........................... 138

Figure 4.15. Investigating the genotoxic effect of AD3-HPRTPM in a HR repair induced

system. ............................................................................................................................ 140

Figure 4.16. Mbo I restriction enzyme digestion of HPRT mutant PCR products. ........... 143

Figure 4.17. Distribution of AD3-HPRTPM mediated mutation in exon 3 of the HPRT

locus.. .............................................................................................................................. 144

Page 14: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

14

Figure 5.1. Template used in the in vitro primer extension assay. .................................. 152

Figure 5.2. The chemical structure of adenine based nucleotides. ................................. 153

Figure 5.3. Single nucleotide incorporation of dATP.. ..................................................... 154

Figure 5.4. Incorporation of dATP using an extended template. ..................................... 154

Figure 5.5. Incorporation kinetics of phosphorothioate nucleotide analogue dATPαS. ... 155

Figure 5.6. Incorporation kinetics of 2’O-methyl ribose modified nucleotide analogue

2’OMe-ATP. ..................................................................................................................... 156

Figure 5.7. The chemical structure of cytidine based nucleotides.. ................................. 157

Figure 5.8. Incorporation kinetics of dCTP and 5Me-dCTP. ............................................ 158

Figure 5.9. Genotoxicity of canonical nucleotides. .......................................................... 161

Figure 5.10. Genotoxicity of a non-canonical phosphorothioate nucleotide analogue. ... 164

Figure 5.11. Clastogenic/ aneugenic activity of dAMPαS in human TK6 cells. ............... 166

Figure 5.12. PCR amplification of cDNA from isolated dAMPαS induced TK mutants. ... 167

Page 15: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

15

List of tables

Table 2.1. Triplex Forming Oligonucleotide sequences ..................................................... 57

Table 2.2. D-loop forming oligonucleotides ....................................................................... 57

Table 2.3. Oligonucleotides used in gel retardation assays............................................... 64

Table 2.4. Primer sequences used for PCR amplification. ................................................ 65

Table 2.5. Oligonucleotides used in primer extension assays ........................................... 67

Table 2.6. Antibodies used in immunoblot analysis. ......................................................... 89

Page 16: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

16

Chapter 1

General introduction

1.1. Introduction

Approximately 7.1 million deaths worldwide are attributed to cancer, according to the 2002

World Health Report, with lung, stomach, colon, liver, breast and oesophageal cancers the

most prevalent (http://www.who.int/whr/2002/en/). More frightful is that in 20 years the

incidence of cancer related death is expected to increase by 50%

(http://www.iarc.fr/en/publications/pdfs-online/wcr/2003/index.php). Sequencing of the

human genome has helped inform the role of genes involved in cancer development/

susceptibility and provide novel drug targets for human therapy (Dean & Bennett, 2003,

Pirollo et al., 2003). One of the major limitations for drug development against molecular

targets is the time required to design a functional molecule (Dean & Bennett, 2003).

However, this limitation can be avoided by using antisense oligonucleotide (ASO)

technologies which can be used to modulate expression of theoretically any gene

(Davidson & McCray, 2011, Pirollo et al., 2003, Agrawal, 1999); although the testing and

validation procedure is the same for small molecule drugs and oligonucleotide based

pharmaceuticals, the time taken to design an ASO is faster (Davidson & McCray, 2011,

Pirollo et al., 2003). It is important to acknowledge the difference between ASO and small

interfering ribonucleic acids (siRNA). siRNA are short double stranded ribonucleic acid

(RNA) sequences which require processing and assembly into the RNA-induced silencing

(RISC) complex for target messenger RNA (mRNA) binding and degradation (Elbashir et

al., 2001, Zamore, 2001, Hammond et al., 2000, Zamore et al., 2000). However, ASO are

single stranded oligonucleotide sequences that do not require endogenous processing and

Page 17: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

17

bind to target mRNA independent of the RISC complex. This study has focused on ASO

therapeutics and not siRNA.

1.2. Antisense oligonucleotides (ASO)

ASO are short oligonucleotide sequences, typically 12-25 nucleotides, designed to bind

complimentary to the mRNA sequence via Watson-Crick base pairing (Geary, 2009, Dean

& Bennett, 2003, Pirollo et al., 2003, Dias & Stein, 2002, Stephenson & Zamecnik, 1978,

Zamecnik & Stephenson, 1978). In theory, providing the mRNA sequence is known,

expression of any gene can be modulated (Davidson & McCray, 2011, Pirollo et al., 2003,

Agrawal, 1999). Antisense activity of ASO therapeutics is governed by several factors,

including: the stability of oligonucleotide from nuclease digestion, cellular uptake, selective

and high affinity binding to target mRNA and the ability to evoke target degradation by

RNase H (described in section 1.2.3), repress translation or modulate alternative splicing

(Dean & Bennett, 2003, Pirollo et al., 2003, Sazani & Kole, 2003, Dias & Stein, 2002,

Agrawal, 1999).

1.2.1. Chemical modifications

The first ASO contained phosphodiester linkages and was reported to inhibit Rous

sarcoma virus translation and transformation of cells (Stephenson & Zamecnik, 1978,

Zamecnik & Stephenson, 1978). The limitation of using unmodified ASO was that they

were found to be rapidly degraded (Akhtar, Kole & Juliano, 1991). For ASO to be efficient

therapeutic tools improving resistance to serum and intracellular nucleases and effective

cellular uptake is critical for their future use (Pirollo et al., 2003). Amongst the first

chemical modifications employed in ASO design was the replacement of a non-bridging

oxygen in the phosphate backbone with either a CH3 group (methylphosphonates) (Miller,

Page 18: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

18

1989, Miller et al., 1979) or a sulphur atom (phosphorothioate) (Stein & Cohen, 1988)

(Fig.1.1).

From the various chemical modifications that can be adopted in ASO design, the

most prevalent amongst ASO used in clinical trials are phosphorothioate linkages (Pirollo

et al., 2003). Besides the increased nuclease resistance provided by this modification

(Eckstein, 1985), phosphorothioate ASO are one of the few capable of stimulating (RNase

H mediated) degradation of hybridised mRNA which offers potent antisense activity (Stein

et al., 1988). However, phosphorothioate ASO administration in humans can result in

thrombocytopenia, anaemia, hyperplasia of the liver, spleen and kidneys and activation of

the immune system in a sequence independent manner (Geary, 2009, Krieg, 1999,

Agrawal et al., 1997, Henry et al., 1997). In particular, ASO containing unmethylated CpG

dinucleotide motifs are capable of stimulating B cell proliferation through Toll-like receptor

recognition (TLR 7,8 and9) and activation of the mitogen-activated protein kinase pathway

(Bafica et al., 2004, Hemmi et al., 2000, Krieg, 1999, Yi & Krieg, 1998, Krieg et al., 1995,

Tanaka, Chu & Paul, 1992). The activation of B cells by CpG ASO is thought to be a result

of immune system recognition of an invading pathogen as bacterial deoxyribonucleic acid

(DNA) has higher frequency of CpG motifs that are unmethylated compared to vertebrate

DNA (Krieg, 1999, Krieg et al., 1995, Kuramoto et al., 1992, Adrian P., 1987). However,

replacing the cytosine with 5’-methyl-cytosine in the CpG motif mitigates immune system

activation by oligonucleotides (Krieg et al., 1995). These limitations have created a niche

for new chemical modifications which provide improved ASO pharmacokinetics,

pharmacodynamics and reduced side effects associated with administration.

Modification of the sugar-phosphate backbone has also been adopted in ASO

design in hope of replacing phosphorothioate linkages. Such modifications include peptide

nucleic acids (PNA) (Egholm et al., 1993), phosphoroamidates (Froehler, Ng & Matteucci,

1988) and morpholinos (Summerton & Weller, 1997) (Fig.1.1) (Egholm et al., 1993).

Page 19: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

19

Although PNA, methylphosphonates, phosphoroamidates and morpholino oligonucleotides

are almost completely nuclease resistant and can hybridise to mRNA targets with very

high affinity, they cannot penetrate the cell membrane, have reduced solubility and cannot

elicit degradation of target mRNA via RNase H and are often found to be weak antisense

molecules (Dias & Stein, 2002, Dias et al., 1999, Summerton & Weller, 1997, Egholm et

al., 1993, Furdon, Dominski & Kole, 1989, Miller, 1989, Miller et al., 1979).

More recent modifications of ASO include replacing the 2’-OH group in the ribose

moiety with O-methyl (2’-OMe) (Fig.1.1). Complete modification of ASO with 2’-OMe

permits higher binding affinity to target mRNA compared to phosphorothioate ASO; for

example, a phosphorothioate ASO hybridised to target mRNA had a melting temperature

of 45°C whereas the 2’-OMe ASO increased the melting temperature by ~50% (Metelev,

Lisziewicz & Agrawal, 1994). However, 2’-OMe ASO do not possess potent antisense

activity because they cannot elicit degradation of target mRNA via RNase H; ASO which

are RNase H independent can only exert antisense activity by physically blocking the

progression/initiation of the translation or splicing machinery (discussed in section 1.2.3)

(Sazani & Kole, 2003, Dias & Stein, 2002, Shibahara et al., 1989). To improve on this,

ASO have been further engineered to contain central phosphorothioate linkages which are

flanked by 2’-OMe ribose nucleotides. These phosphorothioate/ 2’-OMe ASO are reported

to have reduced side effects, increased nuclease resistance and target binding affinity

whilst retaining the ability to evoke an RNase H response (Yoo et al., 2004, Agrawal &

Zhao, 1998, Peng Ho et al., 1998, Yu et al., 1996, Metelev, Lisziewicz & Agrawal, 1994,

Shibahara et al., 1989); for example, the addition of a single 2’-OMe nucleotide at the 5’

and 3’ end increased the melting temperature of the ASO/mRNA duplex by 3.4°C.

Furthermore, snake venom phosphodiesterase fully degraded a phosphorothioate

oligonucleotide within 7 hours (h) but failed to degrade a 2’-OMe and phosphorothioate

modified oligonucleotide (Metelev, Lisziewicz & Agrawal, 1994).

Page 20: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

20

Figure 1.1. Chemical modifications used in ASO design. Figure taken and manipulated from Dias et

al. 2002

Page 21: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

21

A 2’-OMe modified ASO has been reported to retain antisense activity by eliciting

RNase H activation providing at least 4 or 5 deoxyribonucleosides, containing

phosphorothioate linkages, are included (Yoo et al., 2004, Monia et al., 1993). In one

particular study, including a stretch of 6 deoxyribonucleosides on the 5’ end of a

2’-OMe modified ASO, as opposed to the middle or 3’ end, radically improved the ability of

the hybridised ASO to elicit RNase H mediated cleavage of target mRNA by 2-4 fold

(Shen, Kandimalla & Agrawal, 1998). In the design of these 2’-OMe phosphorothioate

ASO, the 2’-OMe domain serves to increase ASO target binding affinity and the

deoxyribonucleosides activate RNase H. This observation has also been made for other

ASO chemical modifications which fail to elicit RNase H activity. Using human U1 small

nuclear RNA as a target, an unmodified (canonical phosphodiester linkages, sugar and

base moieties) and phosphorothioate ASO were biologically active in downregulating U1

small nuclear RNA whereas a methylphosphonate modified ASO failed to have such

effect. However, inclusion of 6 phosphorothioate linkages into the methylphosphonate

ASO was reported to possess antisense activity through RNase H activation (Agrawal et

al., 1990).

The objective of engineering ASO with various chemical modifications is to enhance

biologically stability and efficacy such that ASO can be administered less frequently to

exert pharmacological activity which would also reduce toxic side effects associated with

current ASO administration (Agrawal, 1999).

1.2.2. Biological activity of ASO

ASO are carefully designed to contain various chemical modifications to facilitate biological

activity. Often ASO are employed to modulate splicing of pre-mRNA or down regulate

protein expression either through translational repression or degradation of target mRNA

Page 22: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

22

in a sequence specific manner. Examples are discussed below and ASO used in clinical

trials are discussed in section 1.2.4.

The murine double minute 2 (MDM2) protein is over expressed in 40-60% of human

osteogenic sarcomas and 30% of soft tissue sarcomas implicating a role of the MDM2

gene in the development of these cancers (Cordon-Cardo et al., 1994, Oliner et al., 1992).

MDM2 protein function is to bind p53 protein in a negative feedback loop and inhibit its

function as a transcription factor (Momand et al., 1992). Using 100-400nM of a

phosphorothioate ASO, MDM2 protein expression was reduced 3-5 fold in JAR

(choriocarcinoma) and SJSA (osteosarcoma) cell lines by eliciting RNase H mediated

degradation of MDM2 mRNA (Chen et al., 1998). Down regulation of MDM2 protein

expression increased susceptibility of treated cells to apoptosis and the cytotoxic effects of

camptothecin by up to 30 fold.

Including novel chemical modifications in ASO design such as locked nucleic acids

(LNA), which contain a methylene bridge that connects the 2’-oxygen of ribose with the 4’

carbon (Petersen & Wengel, 2003, Orum & Wengel, 2001), facilitates potent antisense

activity compared to phosphorothioate molecules (Grunweller et al., 2003, Kurreck et al.,

2002). In a comparative study, an ASO containing a central deoxyribonucleoside moiety of

8-10 nucleotides, to activate RNase H, flanked by 4-5 LNA monomers was almost 1000

fold more potent than its comparative phosphorothioate ASO in mediating knockdown of a

green fluorescent protein (GFP) based reporter construct in monkey COS-7 cells

(Grunweller et al., 2003). Even though phosphorothioate ASO have been reported to be

more potent than 2’-OMe, PNA and methylphosphonate ASO in inhibiting reverse

transcription of the human immunodeficiency virus type 1 (HIV-1) (Boulme et al., 1998),

the combination of chemical modifications (once optimised) in ASO design may eventually

become prevalent in clinical trials to mitigate human diseases.

Page 23: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

23

ASO have also been engineered to modulate alternative splicing of pre-mRNA

which occurs in up to 60% of human genes and almost 50% of genetic disorders are a

result of defective pre-mRNA splicing, including various cancers, Duchenne Muscular

dystrophy (DMD), thalassemia and cystic fibrosis (Sazani & Kole, 2003). ASO can be

designed to target mutations in pre-mRNA that cause aberrant splicing, to physically block

the aberrant splice site or induce exon skipping, to restore function to a defective gene. In

the design of these ASO, chemical modifications adopted must not elicit RNase H to

degrade target mRNA; as discussed above, ASO completely modified with 2’-OMe,

methylphosphonate, LNA and phosphoroamidate can bind with high affinity to target

mRNA without activation of RNase H.

For example, DMD is caused by mutations which disrupt the reading frame or result

in a premature stop codon in the dystrophin gene creating a non-functional protein (Koenig

et al., 1989, Sicinski et al., 1989, Monaco et al., 1988). DMD, characterised by progressive

muscle wasting and weakness, affects 1 in 3500 male births (Koenig et al., 1989, Koenig,

Monaco & Kunkel, 1988). Almost 90% of those with DMD do not make it to their 20th

birthday due to cardiac and respiratory complications (Hoffman, Brown & Kunkel, 1987).

ASO have been employed for the treatment of DMD using the mdx mouse animal model.

The mdx mouse model has a premature stop codon in exon 23 of the dystrophin gene

resulting in a non-functional protein (Bulfield et al., 1984). ASO targeting the 5’ splice site

of intron 23 in pre-mRNA have been reported to cause skipping of exon 23 restoring the

reading frame of mRNA from immortalised mdx myoblast cells in vitro (Wilton et al., 1999)

and in vivo, following intramuscular injection into mdx mice, resulting in the production of a

truncated semi-functional protein which produces the milder phenotype of Becker

muscular dystrophy (Fig. 1.2) (Mann et al., 2001). A similar observation has been made

with LNA ASO directed to the splice site of exon 46 in the dystrophin pre-mRNA (Aartsma-

Rus et al., 2004). Treating primary human myoblasts from a DMD patient with 500nM LNA

Page 24: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

24

ASO caused exon skipping in pre-mRNA up to 75% compared to 20% with the 2’-OMe

ASO. Currently, an ASO (AVI-4658, AVI BioPharma) that has finished Phase I/II clinical

trials was reported to be well tolerated for treatment of DMD by modulating alternative

splicing of pre-mRNA (http://clinicaltrials.gov/ct2/show/NCT00159250?term=antisense+

oligonucleotide&rank=2).

1.2.3. ASO mechanism of action

ASO can be categorised into two groups depending on their mechanism of action: (1)

RNase H dependent ASO which result in degradation of target mRNA and (2) RNase H

independent ASO which physically inhibit the initiation or progression of splicing or

translational machinery (Fig.1.3) (Sazani & Kole, 2003, Dias & Stein, 2002, Dias et al.,

1999, Baker et al., 1997).

RNase H is an intracellular enzyme that cleaves the RNA strand of a DNA/RNA

duplex producing shorter oligonucleotide fragments (Hausen & Stein, 1970, Stein &

Figure 1.2. Exon skipping by ASO targeting the DMD pre-mRNA. ASO targeting to the 5’ splice site of intron 23 in pre-mRNA results in exclusion of exon 23 in the mature mRNA by physically blocking splicing machinery access. Elimination of exon 23 carrying the premature stop codon (*) restores the open reading frame resulting in a truncated yet semi-functional protein. Vertical arrows indicate the intron/exon splice sites.

Page 25: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

25

Hausen, 1969). RNase H from E. coli was reported to be vital for DNA replication in the

bacterium (Zamaratski, Pradeepkumar & Chattopadhyaya, 2001, Dasgupta, Masukata &

Tomizawa, 1987). Following this, it was proposed the enzyme may be important for

degradation/elimination of the RNA primer in the RNA/DNA duplex following DNA

replication.

The majority of ASO in current clinical trials depend on RNase H activation to exert

antisense activity provided by the phosphorothioate backbone (Dias & Stein, 2002).

RNase H mediated degradation of target mRNA is highly efficient reaching up to 95%

knockdown of protein expression (Dias & Stein, 2002). Further to this, RNase H dependent

ASO can be theoretically directed to any region of the target mRNA whilst ASO

independent of RNase H activity are only biologically active when directed to the AUG

translation start codon or to a splice site (Sazani & Kole, 2003, Dean et al., 1994, Larrouy

et al., 1992). For example, phosphorothioate ASO targeting the translation start codon in

the Protein kinase C (PKC)-α mRNA have been reported to be the most biologically active

(Dean et al., 1994). However, phosphorothioate ASO targeting most positions in the

mRNA down regulated PKC-α protein expression; ASO containing 2’-OMe ribose failed to

reduce PKC-α protein expression unless the AUG initiation codon was targeted and in this

case, PKC-α mRNA levels were not affected suggesting an RNase H independent

mechanism which confirms translational repression as the mechanism of action (Dean et

al., 1994).

Page 26: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

26

1.2.4. ASO used in clinical trials

ASO have been actively recruited as therapeutic tools to modulate protein expression to

treat genetic diseases. In 2003, there were at least 46 ASO in clinical trials, 10 of which

were in phase III and 20 in phase II, the majority of which contained phosphorothioate

modifications (Dean & Bennett, 2003, Pirollo et al., 2003, Hogrefe, 1999). According to the

United States National Institute of Health, there are currently 51 ASO in clinical trials that

have recently completed Phase I or are still on-going (www.clinicaltrials.gov). Almost half

of the 2003 ASO trials were designed as anticancer therapeutics including molecular

targets such as C-RAF-1, RAS and Protein Kinase A- type I (Cunningham et al., 2001,

Chen et al., 2000, Cunningham et al., 2000). Many tumours have altered or increased

Figure 1.3. ASO mechanism of action. ASO oligonucleotide enter the cytoplasm via endocytosis and then the nucleus (1 and 2) (Lorenz et al., 2000, Loke et al., 1989, Yakubov et al., 1989). ASO hybridises to complimentary target mRNA (3) and reduces target protein expression by physically blocking initiation/progression of mRNA translation (4-6) or stimulating RNase H mediated degradation of mRNA (7 and 8). Image extrapolated from Dean et al. 2003.

Page 27: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

27

gene expression, and since ASO act in a sequence specific manner, they can be designed

to modulate expression of mutant or tumour associated genes without affecting expression

in normal cells (Pirollo et al., 2003).

The lack of site specific delivery methods for ASO remains the biggest obstacle in

their use as therapeutics (Akhtar et al., 2000). ASO are typically administered systemically,

but the dose limiting toxicity associated with intravenous administration includes

hypotension, fever and fatigue, as described in section 1.2.1. (Pirollo et al., 2003). The

similarity in the toxicity profile between different ASO is a reflection on the

phosphorothioate backbone common to most of the compounds tested. Sequence

independent interactions of phosphorothioate ASO with hydrophilic groups of serum

albumin reduces elimination and increases systemic cellular uptake mainly into the liver,

kidneys, bone marrow and lymph nodes resulting in the toxic side effects that are reported

following administration (Geary, 2009, Levin, 1999).

The first ASO approved, by the United States Food and Drug Administration (FDA),

was a phosphorothioate ASO called Vitravene™ (ISIS 2922, Isis Pharmaceuticals) for the

treatment of cytomegalovirus (CMV) retinitis in 1998 (Crooke, 1998). Treating CMV

infected human fibroblast cells with 1µM ISIS 2922 inhibited antigen production by 99%

and virus production by 104 fold demonstrating the efficacy of antisense technology (Cinatl

et al., 2000).

G3139 (Genta Inc., CA, USA) is an 18 nucleotide phosphorothioate ASO targeting

the first 6 codons of the BCL-2 gene mRNA (Banerjee, 2001). The BCL-2 gene is an anti-

apoptotic mitochondrial protein which is over expressed in many cancers (Gross,

McDonnell & Korsmeyer, 1999, Reed, 1999). Daily subcutaneous treatment of 16 non-

Hodgkin’s lymphoma patients with G3139, for 14 days, in phase I trials resulted in

reduction of BCL-2 expression in 7 patients (Waters et al., 2000). In a larger cohort, 35

patients with advanced solid tumours (predominantly in the prostate) were treated with

Page 28: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

28

G3139 with continuous intravenous infusion over 14 or 21 days (Morris et al., 2002).

Although antitumour activity was not seen following treatment, immunoblot analysis using

peripheral blood mononuclear cells showed a reduction in BCL-2 protein expression.

G3139 has also been used in combination with the chemotherapeutic drug

mitoxantrine to treat patients with prostate cancer (Chi et al., 2001). The ASO was

intravenously administered continuously for 14 days with a single administration of

mitoxantrine on day 8. BCL-2 protein expression from peripheral blood mononuclear cells

was decreased in all 5 patients. The addition of G3139 with dacarbazine for treatment of

malignant melanoma (currently in Phase III clinical trial; WWW.clinicaltrials.gov) was shown

to increase the median survival rate by 2 months which was paralleled with an increase in

the median progression free survival from 49 days (dacarbazine alone) to 78 days (G3139

+ dacarbazine) (Dean & Bennett, 2003, Banerjee, 2001).

Similarly to G3139, ISIS 3521 (Isis Pharmaceuticals) is a 20 nucleotide

phosphorothioate ASO designed to target the 3’ untranslated region of the PKC-α gene

mRNA (McGraw et al., 1997, Dean et al., 1996). PKC-α is a serine-threonine protein

kinase involved in signal transduction pathways that controls cell proliferation;

overexpression of PKC-α has been implicated in many cancers making it an appropriate

molecular target for therapeutic agents (McGraw et al., 1997, Yazaki et al., 1996, Gescher,

1992). Mice with subcutaneous glioblastoma U-87 xenografts treated daily for 21 days with

2 or 20mg/kg ISIS 3521 resulted in suppression of tumour growth by 3 or 5 fold,

respectively, compared to control oligonucleotide (Yazaki et al., 1996). ISIS 3521 also

suppressed intracranial U-87 tumour growth in mice treated daily for up to 82 days with

ISIS 3521 whereas control oligonucleotide treated mice did not survive past day 42.

Page 29: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

29

1.3. Concerns with oligonucleotide based pharmaceuticals

Genotoxicity testing of pharmaceuticals that are not biotechnology derived is regulated by

the International Conference on Harmonisation of Technical Requirements for Registration

of Pharmaceuticals for Human Use (ICH) guidelines S2A and S2B

(http://www.ich.org/fileadmin/Public_Web_Site/ICH_Products/Guidelines/Safety/S2_R1/St

ep4/S2R1_Step4.pdf). The battery of genotoxicity tests are designed to inform the

potential for compounds to induce genetic damage. Providing the methodology of each

test does not deviate from the standardised protocol set by the Organisation for Economic

Co-operation and Development (OECD) without scientific justification, those compounds

which are found to be genotoxic are likely to be human mutagens/carcinogens that cause

heritable alterations to the DNA sequence. The standard genotoxicity battery of tests is

comprised of the following:

(1) A bacterial gene reverse mutation assay, typically using Salmonella typhimurium

(Ames test) or Escherichia coli to detect point mutations and frameshift mutations.

(2) An in vitro gene mutation assay such as the Thymidine kinase (TK) forward

mutation assay using mouse lymphoblastoid L5178Y cells or a cytogenetics assay

for evaluation of chromosomal damage using mammalian cells such as the in vitro

micronucleus assay.

(3) An in vivo test for chromosome damage such as the in vivo bone marrow

micronucleus assay using rodents where the frequency of micronuclei in

polychromatic erythrocytes is a marker of clastogenic/ aneugenic activity.

According to the ICH S6 guideline for pre-clinical safety evaluation of biotechnology

derived pharmaceuticals, including ASO therapeutics and monoclonal antibodies, routine

genotoxicity testing as described above is not appropriate and these molecules are not

Page 30: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

30

expected to interact with DNA (www.ich.org/fileadmin/Public_Web_Site/ICH_Products/

Guidelines/Safety/S6_R1/Step4/S6_R1_Guideline.pdf).

However, publication of oligonucleotide mediated mutagenesis using engineered

reporter constructs has raised concern from the EMA regarding the capability of

oligonucleotide based pharmaceuticals, such as ASO, to bind genomic DNA and cause

heritable sequence alterations (European Medicines Agency, 2004). Single strand

oligonucleotides have been reported to be capable of forming secondary structures with

duplex DNA, in a sequence specific manner, to form triplex or D-loop structures which

have been reported to be mutagenic in engineered reporter constructs (Bonner & Kmiec,

2009, Maguire & Kmiec, 2007, Radecke et al., 2006, Dekker, Brouwers & te Riele, 2003,

Faruqi et al., 2000, Vasquez et al., 1999, Wang, Seidman & Glazer, 1996, de Wind et al.,

1995).

As discussed in section 1.2.1, ASO therapeutics are chemically modified to offer

pharmacological activity. The EMA has also highlighted concern regarding the fate of

degradation products from chemically modified oligonucleotide therapeutics (European

Medicines Agency, 2004). Degradation of chemically modified ASO may release

nucleotide analogues that may enter intracellular deoxyribonucleoside triphosphate

(dNTP) pools and become substrates for DNA polymerase facilitating insertion into newly

synthesised genomic DNA. If base pairing with nucleotide analogue occurs with reduced

fidelity the probability of mutation is increased.

1.3.1. Triplex forming oligonucleotides (TFO)

The first triplex structure was described in 1957 by Felsenfeld et al. (Felsenfeld, Davies &

Rich, 1957) composed of two polyribouradylic acid chains (poly-rU) and one

polyriboadenylic acid chain (poly-rA). The structure was described as having an increased

sedimentation rate relative to the duplex formed from single chains of poly-rA and poly-rU.

Page 31: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

31

The triplex structure appeared to be stabilised in the presence of divalent cations

presumed to neutralise the negative charges of the phosphate backbones in the

oligoribonucleotide strands.

Since its discovery, DNA containing a purine rich stretch has been shown to be

capable of binding a third oligonucleotide strand in a sequence specific manner to form a

triplex structure (Fig.1.4) (Duca et al., 2008, Jain, Wang & Vasquez, 2008, Kalish et al.,

2005, Guntaka, Varma & Weber, 2003, Washbrook & Fox, 1994, Durland et al., 1991,

Hoogsteen, 1959). TFO bind in the major groove of duplex DNA where acceptor and donor

groups present in the purine nucleotides are capable of hydrogen bonding with the

nucleotides of the TFO (Fig.1.5) (Guntaka, Varma & Weber, 2003, Cooney et al., 1988).

The hydrogen bonds between duplex DNA and TFO are not identical to the interactions

between Watson-Crick base pairs and are referred to as Hoogsteen type hydrogen bonds

(Hoogsteen, 1959).

Figure 1.4. DNA triplex formation. TFO bind the major groove of duplex DNA containing purine tracts via Hoogsteen hydrogen bonds. Blue and green strands form duplex DNA. The red strands represents the TFO. Image taken from Guntaka et al. 2003.

Page 32: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

32

There are three classifications of TFO, in which the rules for triplex formation are

well described (Fig.1.6) (Buchini & Leumann, 2003, Chan & Glazer, 1997, Frank-

Kamenetskii & Mirkin, 1995); (i) The pyrimidine (TC) motif TFO composed of thymidine

and cytosine binds parallel to the triplex target sequence forming T:A•T and C:G•C triplets.

Triplex formation by TC motif TFO is limited by pH as the cytosine requires protonation in

order to bind duplex DNA. Hence, pyrimidine motif TFO are not readily applicable for

triplex formation at physiological pH without replacing the cytosine base with 5’-methyl-

cytosine to mitigate the pH dependence of binding (Xodo et al., 1991, Lee et al., 1984).

(ii) The purine (GA) motif TFO composed of guanine and adenine is capable of binding

antiparallel to the triplex target sequence independent of pH, forming C:G•G and G:A•A

triplets. (iii) The mixed (GT) motif TFO composed of guanine and thymidine is capable of

binding parallel or antiparallel to the triplex target sequence forming C:G•G and T:A•T

triplets depending on the sequence composition [“:” indicates Watson-Crick hydrogen

bonds; “•” indicates Hoogsteen hydrogen bonds].

Figure 1.5. Hoogsteen hydrogen bonding between nucleotides of TFO and duplex DNA. ‘IIIII’ indicates Watson-Crick hydrogen bonding between base pairs in duplex DNA and ‘••••’ indicates Hoogsteen hydrogen bonds between nucleotides in TFO and purine nucleotides in duplex DNA to form A•A:T and G•GC triplets. Image taken from Chan and Glazer, 1997.

Page 33: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

33

1.3.1.1. Mutagenicity of TFO

Using mammalian cells, triplex formation has been reported to induce sequence specific

repression of target gene transcription (Kochetkova & Shannon, 1996, Grigoriev et al.,

1992, Cooney et al., 1988) and sequence specific mutagenesis using reporter constructs

containing selectable loci or transgenic mice; in these studies, triplex mediated

mutagenesis was described for populations of cells containing an engineered target

sequence (Kalish et al., 2005, Datta et al., 2001, Vasquez et al., 2001, Faruqi et al., 2000,

Xu, Glazer & Wang, 2000, Vasquez et al., 1999, Wang, Seidman & Glazer, 1996); with

TFO covalently linked to a DNA intercalator (psoralen or acridine) (Macris & Glazer, 2003,

Vasquez et al., 2001, Xu, Glazer & Wang, 2000, Vasquez et al., 1999); by pre-incubating

the target vector with TFO under non-physiological conditions (Ye, Guntaka & Mahato,

2007, Datta et al., 2001, Xu, Glazer & Wang, 2000) or with electroporation to deliver

oligonucleotide (Puri et al., 2002, Puri et al., 2001, Faruqi et al., 2000, Majumdar et al.,

1998) and thus do not necessarily reflect biologically relevant events. To the best of my

knowledge, no study has examined triplex mediated mutagenesis at genomic coding

sequences using a non-conjugated TFO.

Figure 1.6. Classification of TFO. TFO (red) are classified according to nucleotide composition and binding orientation relative to the polypurine target sequence (green) in duplex DNA. Image taken and modified from Duca et al. 2008.

Page 34: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

34

Nevertheless, Cooney et al. (Cooney et al., 1988) demonstrated a 27mer

oligonucleotide was capable of binding to a plasmid construct encompassing the c-myc

gene. The TFO target was a purine rich stretch upstream of the transcription initiation site.

Through the use of electrophoretic mobility shift assays (EMSA) and DNase 1 protection

assays TFO binding was estimated to have a dissociation constant (Kd) around 4nM. The

TFO was shown to be effective in downregulation of c-myc gene transcription through

inhibition of transcription initiation. This was hypothesised to be a result of the occupancy

of a putative protein binding site vital to the target gene’s expression, which was later

confirmed in several systems (Kochetkova & Shannon, 1996, Grigoriev et al., 1992).

Wang et al. (Wang, Seidman & Glazer, 1996) demonstrated the capability of a TFO

to induce mainly point mutations and small deletions around the triplex target sequence.

Within monkey COS cells, three phosphodiester oligonucleotides (AG10; AG20; AG30

named according to TFO composition and length) targeted a Simian virus 40 (SV40)

based shuttle vector containing the supFG1 reporter gene. Modified to enclose a purine

stretch, the supFG1 reporter was used to assess the mutagenic potential of the TFO.

Wang et al. (1996) reported mutation frequencies using the supFG1 system in the order of

0.27% (2700x10-6), 13 fold above background, using 2micromolar (μM) AG30 TFO (Kd;

20nM) incorporated into the cell growth media for 48h. AG10 (Kd; 30μM) and AG20 (Kd;

300nM) TFO produced a mutant frequency 3 and 5 fold above background, respectively. It

is notable that the reported background mutant frequency in this model is extraordinarily

high.

More recently, Kalish et al. (Kalish et al., 2005) reasoned the mutagenic potential of

TFO to be dependent on the helical distortion produced upon triplex formation inferred

from a nuclear magnetic resonance based structural study; oligonucleotides containing

substitutions at the 2’ position of ribose were found to produce less helical distortion i.e.

the relative degrees of distortion were DNA>RNA>2’-OMe (Asensio et al., 1999). In

Page 35: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

35

agreement with the mutant frequency in their system, those chemical modifications

promoting the C3’-endo conformation (RNA and 2’-OMe) were not mutagenic; the

deduction was that those TFO which produced a significant helical distortion possess the

ability to provoke the DNA repair pathway, initiated by the recruitment of the XPA/RPA

DNA damage recognition complex to the triplex site, resulting in mutation (Vasquez et al.,

2002).

A possible reason for using engineered reporter constructs, such as SupF and

GFP, to demonstrate triplex mediated mutagenesis is due to the fact triplex formation is

governed by such stringent conditions i.e. a purine stretch and divalent cations. The

availability of a 20-30 nucleotide purine stretch is limited in selectable genomic loci that

can be used as a reporter of mutation but these reporters readily allow manipulation of

reporter constructs and high-throughput screening using previously established assays.

1.3.2. D-loop forming oligonucleotides

Single strand oligonucleotides have also been shown to be capable of binding to

complementary sequences in duplex DNA during transcription and DNA replication such

that one of the duplex strands becomes displaced forming a D-loop structure (Wu et al.,

2005). Oligonucleotides which have a mismatched base upon binding to their

complementary sequence in duplex DNA are capable of directing mutation to the site of

the mismatched base in a process known as targeted nucleotide exchange (TNE).

Oligonucleotide mediated mutagenesis via TNE is thought to be a 2-stage process: firstly,

the oligonucleotide binds to its complimentary strand in duplex DNA to form a D-loop

structure and secondly, mutation is directed to the mismatched base (Maguire & Kmiec,

2007, Wu et al., 2005). Typically, oligonucleotides used in TNE are over 45 nucleotides in

length and chemically modified to contain phosphorothioate linkages to provide nuclease

resistance (Kenner et al., 2002).

Page 36: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

36

Unlike TFO where binding to duplex DNA is dictated by the presence of a purine

stretch, in theory, D-loop forming oligonucleotides can bind to any complementary

sequence in duplex DNA. Also, D-loop formation is irrespective of divalent cations which

are reported to be necessary for stabilisation of triplex structures (Kalish et al., 2005,

Felsenfeld, Davies & Rich, 1957). For example, an ASO designed to bind within the coding

region of mRNA may also be capable of binding to the same complementary sequence in

genomic DNA and induce mutation via TNE but in order for ASO to form a triplex at

genomic DNA they must bind a purine stretch otherwise triplex formation will not occur.

1.3.2.1. Mutagenicity of D-loop forming oligonucleotides

D-loop forming oligonucleotides have been reported to be capable of inducing site specific

mutation in a process known as TNE providing the oligonucleotide contains a mismatched

base upon binding to its complimentary sequence in duplex DNA. Oligonucleotides

(typically >45 nucleotides) are usually engineered to target reporter constructs such as

enhanced GFP (eGFP) (Bonner & Kmiec, 2009, Olsen et al., 2009, Papaioannou, Disterer

& Owen, 2009, Morozov & Wawrousek, 2008, Maguire & Kmiec, 2007, Aarts et al., 2006,

Radecke et al., 2006, Olsen et al., 2005, Dekker, Brouwers & te Riele, 2003, de Wind et

al., 1995, te Riele, Maandag & Berns, 1992). This system allows high throughput

screening using flow cytometry by monitoring the correction of a point mutation that reverts

the mutant eGFP protein functional and fluorescent.

The precise mechanism of TNE by oligonucleotides is not known but the efficiency

of TNE has been reported to be modulated by transcription (Igoucheva et al., 2003), DNA

replication (Brachman & Kmiec, 2005, Olsen et al., 2005, Wu et al., 2005), the mismatch

repair (MMR) pathway (Dekker et al., 2011, Papaioannou, Disterer & Owen, 2009,

Maguire & Kmiec, 2007, Aarts et al., 2006, Dekker, Brouwers & te Riele, 2003, de Wind et

Page 37: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

37

al., 1995) and homologous recombination (HR) repair (Radecke et al., 2006, te Riele,

Maandag & Berns, 1992). See Fig.1.7 for an overview.

The rate of transcription has been reported to positively modulate TNE. Using a

LacZ reporter construct integrated into two different cell populations, from the same cell

line, with different levels of transcription (20 fold difference in LacZ protein expression), a

45mer oligonucleotide was able to direct site specific mutation in cells with elevated LacZ

gene transcription almost 100 fold greater than the same cell line with reduced LacZ

transcription (Igoucheva et al., 2003). This was proposed to be a result of an open

chromatin structure around highly transcribed genes making the target site more

accessible for oligonucleotide binding.

Oligonucleotides targeting the non-transcribed (sense) strand have also been

reported to be more efficient in gene correction via TNE, rather than oligonucleotides

targeting the transcribed (antisense) strand, by up to 200 fold (Bonner & Kmiec, 2009, Wu

et al., 2005, Igoucheva et al., 2003). It was postulated that the strand bias is due to limited

accessibility of the transcribed strand during transcription (Igoucheva et al., 2003).

Furthermore, oligonucleotides bound to the transcribed strand have been reported to be

unwound by a progressing transcription fork, thus, preventing TNE (Liu et al., 2002).

Controversially, Dekker and colleagues did not report a difference in the frequency of TNE

when oligonucleotides were directed to the sense or antisense strands (Dekker, Brouwers

& te Riele, 2003).

To investigate the effect of cell cycle on TNE, Wu and co-workers (Wu et al., 2005)

synchronised HeLa cells, containing a mutant eGFP plasmid, at G1, S and M phase by

treatment with specific inhibitors, L-mimosine, thymidine and nocodazole, respectively,

followed by treatment with oligonucleotide. Correction of mutant eGFP was up to 0.54%

(5,400x10-6) in S phase treated cells whilst much lower in the remaining phases. Taking

the study a step further, they used various S phase inhibitors (thymidine, hydroxyurea and

Page 38: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

38

aphidicolin), which reduce the rate of DNA replication to varying degrees (Saintigny et al.,

2001), to investigate the relationship between the rate of replication and frequency of TNE.

They observed that reducing the rate of replication, using 2mM thymidine, was optimum

for TNE whilst complete inhibition of replication, using hydroxyurea or aphidicolin,

mitigated mutant eGFP gene correction (Wu et al., 2005). Thus, reducing (not inhibiting)

the rate of DNA replication during S phase has a positive modulatory effect on TNE which

is in contrast to the modulatory effects of transcription (Igoucheva et al., 2003). The

authors proposed this effect was a result of a prolonged open chromatin structure that

facilitates efficient annealing of oligonucleotide to duplex DNA by increasing the “window

of opportunity” for binding (Wu et al., 2005). This conclusion was also made by Bachman

and Kmiec (Brachman & Kmiec, 2005); pre-treating cells with 2’,3’-dideoxycytidine

nucleoside resulted in an extended S-phase by stalling replication forks and increased

oligonucleotide mediated gene correction of mutant eGFP by 3 fold. In this study also,

gene correction was absent when DNA replication was completely inhibited. Similar to Wu

et al. (Wu et al., 2005), the authors proposed a prolonged open chromatin structure

increases target site availability for oligonucleotide binding.

Bonner and co-workers reported that prior to electroporation of HCT116 cells with

8µM oligonucleotide, synchronisation of cells in early S phase, using aphidicolin, resulted

in gene correction frequencies of 2% (Bonner & Kmiec, 2009). Importantly, the authors

reported that gene correction of mutant eGFP was coupled to DNA double strand breaks

(DSBs), measured using the alkaline comet assay. However, DNA DSBs following

oligonucleotide treatment were not observed above control (spontaneous or control

oligonucleotide treatments) until the average comets per field were normalised to the cell

number following treatment (i.e. cytotoxicity) which is an unorthodox approach for this

assay. It seems the authors correlated the extent of cytotoxicity following treatment with

the frequency of DNA DSBs. However, cytotoxicity following treatment is not necessarily

Page 39: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

39

caused by DNA damage and therefore, is not the most appropriate method of

normalisation for this assay.

The DNA repair pathways have also been implicated as modulators of TNE. For

instance, the MMR protein, mut S homologue 2 (MSH2), has been proposed to be a

negative modulator of TNE which contradicts initial speculation on the mechanism of gene

correction (Gamper et al., 2000, Cole-Strauss et al., 1999). MSH2 can complex with MSH6

or MSH3 to form MutS-α or MutS-β, respectively, both of which are involved in mismatch

recognition and initiation of DNA repair (Kunkel & Erie, 2005); MutS-α preferentially

recognises 1 or 2 base mismatches whereas MutS-β recognises larger mismatches. It is

also reported that the MSH2 protein possesses anti-recombinogenic activity, if there is

greater than 0.6% sequence divergence between homologous pairs, by directly interacting

with HR repair protein factors and inhibiting their function, (Brown et al., 2003, Wang et al.,

2000, de Wind et al., 1995, te Riele, Maandag & Berns, 1992). Further to this, it is believed

that oligonucleotides employed in TNE physically incorporate into duplex DNA as a result

of HR repair or by oligonucleotides acting as Okazaki fragments during DNA replication

(Radecke et al., 2006, Parekh-Olmedo et al., 2005). Thus, it has been proposed that

reducing MSH2 protein expression increases the efficiency of TNE by mitigating the

associated antirecombinogenic activity (Papaioannou, Disterer & Owen, 2009, Maguire &

Kmiec, 2007, Aarts et al., 2006, Dekker, Brouwers & te Riele, 2003, de Wind et al., 1995,

te Riele, Maandag & Berns, 1992). Alternatively, oligonucleotides may act as primers

during DNA synthesis where the mismatched base stimulates the MMR pathway in wild-

type cells to correct the mismatched base in the newly synthesised strand including the

oligonucleotide (Parekh-Olmedo et al., 2005, Dekker, Brouwers & te Riele, 2003); a

deficiency in MMR may allow the cell to tolerate the mismatched base until cell division

where the mutation becomes fixed.

Page 40: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

40

In mouse embryonic fibroblast cells, a 47mer oligonucleotide was able to correct a

single base deletion in an eGFP reporter plasmid to restore the wild-type sequence but

silencing of MSH2 protein, by RNA interference (RNAi), enhanced gene correction by 3

fold (Maguire & Kmiec, 2007). Furthermore, oligonucleotide mediated gene correction of a

neomycin gene carrying (i) a single point mutation (ATG → AAG) that inactivates the

transcription start codon or (ii) a two base pair insertion to disrupt the open reading frame,

was only observed in mouse embryonic stem cells when MSH2 protein expression was

reduced by RNAi, up to a frequency of 70x10-6 (Aarts et al., 2006). Using this MSH2

deficient system, an oligonucleotide with 4 mismatched bases was also capable of

restoring the neomycin gene open reading frame at a frequency of 20x10-6. In a similar

study, gene correction of 2 tandem frameshift mutations, in a neomycin gene, was

reported in ~32 mouse embryonic stem cells when 7x105 were treated with

oligonucleotide, in an MSH2 deficient system (Dekker, Brouwers & te Riele, 2003). In wild-

type cells, gene correction was as low as 0.5x10-7.

Data has been presented that implicates the involvement of the HR repair pathway

(or at least proteins involved in the initiation of HR repair) in the process of TNE. Co-

injection of a 74mer oligonucleotide targeting a mutant αβ-crystallin gene with recombinant

RAD51 protein (important for HR repair initiation) was capable of correcting a premature

stop codon at a frequency of 2.8% (Morozov & Wawrousek, 2008). Targeted gene

correction was absent with oligonucleotide treatment alone. However, microinjection of

oligonucleotide and an antibody against the Ku70/80 heterodimer increased the correction

efficiency up to 9.9%. The Ku70/80 heterodimer is a critical DNA DSB binding protein vital

for signalling the non-homologous end joining (NHEJ) repair pathway (Jeggo, 1998,

Takata et al., 1998). It is believed that resolution of DNA DSBs is governed by competitive

binding of critical initiation factors to direct repair via NHEJ or HR repair (Pierce et al.,

2001); silencing of Ku protein expression is reported to enhance the HR repair pathway.

Page 41: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

41

This study was the first to highlight the positive and negative modulatory roles of HR and

NHEJ repair on oligonucleotide mediated TNE, respectively (Morozov & Wawrousek,

2008).

In fact, Radecke and colleagues (Radecke et al., 2006) demonstrated that a

biotinylated oligonucleotide was capable of physically incorporating into the genome of

cells following TNE with correction frequencies up to 5%. Most interesting was that

modification of the 5’ or 3’ oligonucleotide termini with 5’-OH or 3’-H in place of a 5’-

phosphate moiety or 3’-OH group, respectively, significantly reduced the efficiency of gene

correction suggesting canonical termini were important for TNE, supporting oligonucleotide

integration into the host cell genome prior to gene correction. A low level of gene

correction was observed possibly as a result of exonuclease metabolism of

oligonucleotides releasing terminal nucleotides carrying non-canonical ends resulting in

oligonucleotides with termini capable of integration. The authors proposed that TNE was a

result of physical incorporation of targeting oligonucleotides into duplex DNA possibly

through (i) HR repair or (ii) oligonucleotides serving as Okazaki fragments during DNA

replication.

A similar observation was made by Olsen et al. (Olsen et al., 2005) when the 5’ and

3’ ends of an oligonucleotide were blocked by an octyl moiety. Targeted gene correction of

a mutant GFP gene was reduced to 16% of that observed with oligonucleotide treatment

without the octyl blocking groups. The authors suggested that functional 5’ and 3’ moieties

may be important for physical incorporation of oligonucleotides into duplex DNA.

To summarise, the rate of gene transcription has been reported to positively

modulate TNE possibly by facilitating oligonucleotide annealing through increased

accessibility of the target sequence due to an open chromatin structure (Fig.1.7, step 1).

The contrary mechanism may apply to oligonucleotide annealing during S-phase DNA

replication; reducing the rate of DNA replication may increase the probability of

Page 42: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

42

oligonucleotide annealing due to a prolonged open chromatin structure (Fig.1.7, step1).

RAD51 protein is likely to facilitate strand invasion of duplex DNA by oligonucleotides to

form a D-loop structure (Fig.1.7). This aberrant structure may result in the stimulation of

the HR repair pathway. Following this, oligonucleotides may physically incorporate into

duplex DNA via HR repair creating a mismatched base with the opposing strand (Fig.1.7,

step 2). Alternatively, oligonucleotides may physically integrate into the host cell genome

by serving as a primer for DNA replication (Fig.1.7, step 2). If the MMR repair pathway is

active, the mismatched base is removed and restored, or, HR of oligonucleotide is

inhibited due to the anti-recombinogenic activity of MSH2 protein. In either case, TNE will

not occur. Inactivation of the MMR pathway, by silencing MSH2 protein expression, may

facilitate HR of oligonucleotides and tolerance of the mismatched base until cell division

where it becomes fixed in the genome of a daughter cell (Fig.1.7, step 3).

Studies addressing the influence of other critical MMR proteins on TNE may clarify

the mechanism of oligonucleotide mediated gene correction. For example, other proteins

in MMR, such as mut L homolog 1 (MLH-1), are vital for downstream signalling (Li, 2008,

Kunkel & Erie, 2005). Silencing of MLH-1 mitigates activity of the MMR pathway but MSH2

protein remains active as an anti-recombinogenic protein (Li, 2008). Thus, if TNE in MLH-1

deficient cells is less than that in MSH2 deficient cells, this may imply that MMR may not

be involved in TNE and it is the HR repair pathway which incorporates oligonucleotide into

genomic DNA when the anti-recombinogenic activity of MSH2 is silenced. Alternatively, if

MLH-1 and MSH2 deficient cells have a similar magnitude of TNE, it may imply that the

MMR pathway serves to correct the mismatched base within newly synthesised DNA upon

incorporation via serving as a DNA primer and that the HR repair pathway may not be

involved. These studies may highlight RAD51 as an important factor in the annealing

process of oligonucleotides but not in HR repair. An important consideration is that

Page 43: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

43

Replication/ Transcription

HR repair

MMR Replication

oligonucleotide incorporation into genomic DNA may not be unique to a single mechanism

but possibly a combination of both.

Figure 1.7. Putative mechanism of TNE by D-loop forming oligonucleotides. Step1: Oligonucleotide binds to its complimentary target sequence in duplex DNA, during DNA replication or transcription, forming a D-loop structure. Duplex DNA contains a mismatched base relative to the oligonucleotide sequence (*). Step 2: oligonucleotide physically incorporates into genomic DNA via HR repair or by acting as an Okazaki fragment during DNA replication. Step 3: once oligonucleotide is incorporated, the mismatched base is either corrected by MMR or tolerated until another cycle of DNA replication fixing the mutation. Image adapted from Radecke et al.2006.

Step 1

Step 2

Step 3

Page 44: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

44

1.3.3. Nucleotide analogues

Unmodified oligonucleotides containing a canonical phosphodiester backbone, sugar and

base moieties tend to be degraded by nucleases to release monophosphate nucleotide

units (Spitzer & Eckstein, 1988). Degradation of oligonucleotides tends to initiate on the 3’

end as 3’ exonucleases are more prevalent in serum than 5’ exonucleases or

endonucleases (Shaw et al., 1991, Tidd & Warenius, 1989, Wickstrom, 1986). Due to such

limitations, it has become routine to chemically modify oligonucleotides to facilitate

biological activity, as discussed in section 1.2.1. Most of these modifications do not offer

complete resistance to nucleases and because of this are slowly degraded to release

nucleotide analogues (Shaw et al., 1991).

Although nucleotide analogues can be derived from oligonucleotide degradation,

nucleoside analogues have also been engineered as antiretroviral therapies to be used as

part of the highly active antiretroviral therapy (HAART) to treat HIV-1 (Fig.1.8) (Olivero,

2008, Olivero, 2007, Wutzler & Thust, 2001).

It has been estimated that around 30 million adults and 2.5 million children

worldwide were living with HIV-1 or acquired immunodeficiency syndrome (AIDS) by the

end of 2009 (http://www.avert.org/worldstats.htm). Typically, antiretroviral nucleoside

analogues are targeted to virus encoded reverse transcriptase. The antiretroviral activity of

nucleoside analogues is mediated by their ability to cause viral DNA chain termination

and/or block the HIV-1 reverse transcriptase nucleotide binding site (Wutzler & Thust,

2001, Huang, Farquhar & Plunkett, 1990, St Clair et al., 1987, Furman et al., 1986).

AZT (3’-azido-2’, 3’-dideoxythymidine; Fig.1.8A) has been recommended for use

since 1994 to prevent vertical transmission of HIV-1 from infected mothers to foetus

(Olivero, 2007). Since then, AZT (Zidovudine) has become one of the most important

components of HAART to treat HIV-1 (Olivero, 2008, Olivero, 2007). AZT administration to

the mother during pregnancy and labour and to the new born baby following birth reduces

Page 45: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

45

the frequency of HIV-1 transmission to children from 30% to 8% (Connor et al., 1994).

AZT used in combination therapy with the antiretroviral nucleoside analogue Lamivudine

(2’, 3’-dideoxy-3’-thiacytidine; Fig.1.8B) and coupled to caesarean section can reduce

vertical transmission to 2% (Panburana et al., 2004).

The biological efficacy of antiretroviral nucleoside analogues depends on cytosolic

phosphorylation to mono-, di- and tri-phosphate forms (Wutzler & Thust, 2001, St Clair et

al., 1987). For example, AZT is mono, di and triphosphorylated by TK, thymidylate kinase

and pyrimidine nucleoside diphosphate kinase (St Clair et al., 1987). This highlights

Figure 1.8. Chemical structure of antiretroviral nucleoside analogues used in HAART. AZT (A), Lamuvidine (B), Ganciclovir (C) and Didanosone (D). Image taken from Wutzler et al. 2001.

A B

C D

Page 46: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

46

potential for nucleoside analogue entry into endogenous dNTP pools following

phosphorylation by endogenous pathways.

Although AZT is a potent HIV-1 reverse transcriptase inhibitor the drug limiting

toxicity of AZT has been proposed to be a result of genomic insult to host cell DNA. This is

supported by previous studies demonstrating AZT incorporation into the genomic DNA of

mouse, hamster and human cell lines (Olivero, Beland & Poirier, 1994).

1.3.3.1. Mutagenicity of non-canonical nucleotide analogues

The majority of nucleoside analogues subject to the battery of genotoxicity testing

(described in section 1.3) are not positive in bacterial gene mutation assays, such as the

Ames test, and tend not to induce point mutations (Wutzler & Thust, 2001); this effect has

been hypothesised to be a result of the mechanism of nucleoside analogue mediated

genotoxicity which is thought to be gross chromosomal damage as a result of obligate

chain termination following nucleoside analogue incorporation. According to Wutzler et al.

(Wutzler & Thust, 2001), most nucleoside analogues tested cause chromosomal damage

albeit at concentrations above physiological plasma levels used in therapeutic treatments.

Ganciclovir (9-[(1,3-dihydroxy-2-propoxy)-methyl]-guanine; Fig.1.8C) causes

cytogenetic damage including chromosome aberrations and sister chromatid exchange

(SCE) (Thust, Schacke & Wutzler, 1996). In Chinese hamster ovary cells expressing the

herpes virus TK gene, Ganciclovir is a very potent genotoxin causing chromosome breaks,

translocations and SCE at concentrations as low as 1nM (Thust et al., 2000a, Thust et al.,

2000b). These data suggest that the drug can be monophosphorylated for its utilisation by

endogenous cellular machinery (Wutzler & Thust, 2001). This is supported by several

groups demonstrating incorporation of radiolabelled Ganciclovir into genomic DNA,

presumably during DNA replication, where chromosomal damage was hypothesised to be

at sites of Ganciclovir incorporation (Thust et al., 2000a, Thust et al., 2000b, Rubsam,

Page 47: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

47

Davidson & Shewach, 1998, St Clair, Lambe & Furman, 1987). Lifetime carcinogenicity

testing in mice was found to be positive with tumours induced in the forestomach (male

and female mice), mammary gland and preputial gland in females. Thus, Ganciclovir is

considered to be a human carcinogen (Physicians' Desk Reference, 2000).

AZT has been reported to be positive in genotoxicity tests including the mouse

lymphoma assay (MLA) using L5178Y cells, in vitro micronucleus assay and in vivo bone

marrow micronucleus assay, SCE, in vitro cell transformation and in vivo carcinogenicity in

mice (Sussman et al., 1999, Ayers et al., 1996, Gonzalez Cid & Larripa, 1994, Phillips et

al., 1991) (http://monographs.iarc.fr/ENG/ Monographs/vol76/mono76.pdf). AZT is also

reported to epigenetically alter the methylation profile of the TK gene causing its

inactivation (Wu et al., 1995, Nyce et al., 1993).

Treatment of human blood lymphocytes in vitro with AZT from 50 micrograms (µg)

per millilitre (ml) (187μM) induced micronuclei and SCE as well as inducing transformed

foci in BALB/c 3T3 cells from 2μM (Ayers et al., 1996, Gonzalez Cid & Larripa, 1994).

Transformation assays are ideally used for suspected non-genotoxic carcinogens which

act via altering the epigenome without genomic sequence alteration. The assay relies on

normal growth of cells and contact inhibition; following treatment with a drug,

transformation of cells results in the development of foci that are no longer regulated by

contact inhibition (Heidelberger et al., 1983, Thust, 1976) (http://ecvam.jrc.it/publication/

WorkshopReport39.pdf).

Oral administration of 30mg/kg/day AZT to female mice induced vaginal carcinomas

and vaginal epithelial neoplasias in rats after administration of 300mg/kg/day (Ayers et al.,

1996). Tumourigenic activity of AZT was not observed in male mice. This was proposed to

be a result of local vaginal epithelium exposure to urine containing high levels of the drug

(Ayers et al., 1996). In a separate study, oral administration of AZT to female mice resulted

in incorporation of AZT into the genomic DNA of vaginal epithelial cells which may explain

Page 48: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

48

the mechanism of AZT tumourigenesis (Olivero et al., 1994, Olivero, Beland & Poirier,

1994).

The exact mechanism of DNA damage caused by nucleoside analogues is not

known. It is likely that genomic insult can occur via two processes: (1) nucleoside

analogues perturb the intracellular balance of dNTP pools altering the fidelity of DNA

polymerase and/or (2) physical incorporation of nucleoside analogues into genomic DNA;

the majority of nucleoside analogues have unmodified bases so it is presumed the fidelity

of base pairing is not affected (Wutzler & Thust, 2001, Kunz et al., 1994, Kunz, 1988).

However, they are modified on the side chain to contain a single functional hydroxyl group

similar to the 5’ hydroxyl group of the sugar ring in standard nucleotides. They do not

contain a 3’ hydroxyl group capable of forming phosphodiester linkages (Olivero, 2007).

Thus, incorporation of nucleoside analogues into newly synthesised DNA causes chain

termination. It has been hypothesised that exonuclease mediated removal of the terminal

nucleoside may trigger error prone DNA repair and when the repair pathway becomes

saturated the apoptotic pathway is triggered (Meng et al., 2002, Wutzler & Thust, 2001,

Olivero et al., 1999a, Olivero et al., 1999b).

The above describe that antiretroviral nucleoside analogues can act as substrates

for nuclear DNA polymerase mediated incorporation into genomic DNA. However,

mitochondrial DNA polymerase γ mediated incorporation of antiretroviral nucleoside

analogues into mitochondrial DNA can also result in dose limiting toxicity (Olivero, 2007,

Johnson et al., 2001). Johnson et al. (Johnson et al., 2001) tested the kinetics of

incorporation and excision of all FDA approved antiretroviral nucleoside analogues into

mitochondrial DNA. The study reported a correlation of highly toxic nucleoside analogues

such as 2’,3’-dideoxyinosine (Didanosone; Fig.1.8D) with increased rate of incorporation

into mitochondrial DNA with inefficient removal. Low toxicity associated with nucleoside

Page 49: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

49

analogues such as AZT and Lamivudine correlated with reduced mitochondrial DNA

incorporation and high rates of removal.

Concerns over toxicity and potential carcinogenicity, associated with antiretroviral

nucleoside analogues (in particular AZT treatment used to prevent vertical transmission of

HIV-1 in pregnant women) have been expressed by regularity authorities in South Africa

where the HIV/AIDS pandemic is on the rise (Birmingham, 2000). This concern is

supported by data confirming AZT incorporation into maternal and infant leukocyte DNA up

to 350 AZT molecules per 106 nucleotides following treatment as part of the HAART

protocol (Olivero et al., 1999b). However, conclusive carcinogenicity in humans by

nucleoside analogues is lacking and suggestions of carcinogenicity are speculative and

based on the fact that these drugs interfere with nucleic acid metabolism and utilisation

therefore make hereditary sequence alterations possible (Wutzler & Thust, 2001). The

validity of these concerns will become clear over the next decade.

An important point to consider is that the genotoxicity of antiretroviral nucleoside

analogues is well established, however, these nucleoside analogues are not used in ASO

design and such genotoxicity data cannot be used to extrapolate mutagenicity of

nucleotide analogues derived from ASO degradation. The data can be used to highlight

the potential for such aberrant nucleoside structures to be substrates for intracellular

kinases and DNA polymerase.

1.3.3.2. Mutagenicity of canonical nucleotides

A continuous and balanced supply of intracellular dNTP pools is essential to maintain the

integrity of DNA during replication (Fersht, 1979, Kunkel & Loeb, 1979, Reichard, 1978).

Supplementing Chinese hamster ovary cells with 1millimolar (mM) thymidine has been

reported to reduce the rate of DNA synthesis by 90% as well as cause an expansion in the

2’-deoxyadenosine triphosphate (dATP), 2’-deoxyguanosine triphosphate (dGTP) and 2’-

Page 50: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

50

deoxythymidine triphosphate (dTTP) pools while decreasing the 2’-deoxycytidine

triphosphate (dCTP) pools 10 fold (Bjursell & Reichard, 1973). Thymidine deprivation and

excess can also induce DNA breaks, deletions, SCE and heritable sequence alterations

(Seno et al., 1985, Perry, 1983, Potter, 1971).

Treatment of human CEM cells with 50μM 2’-deoxyguanosine for 6h increased the

dGTP levels by 43 fold and induced the HPRT mutant frequency 40 fold above control

(Mattano, Palella & Mitchell, 1990). A similar induction of HPRT mutants was observed

with 20μM 2’-deoxyadenosine treatment.

The magnitude and duration of dNTP pool perturbation can affect the rate of

misincorporation of nucleotides in excess (Mattano, Palella & Mitchell, 1990). It is likely

that DNA polymerase slippage at sites of repeating bases is enhanced following

perturbation of dNTP pools facilitating nucleotide misincorporation (Mattano, Palella &

Mitchell, 1990).

DNA polymerase tends to misincorporate the nucleotide in excess to produce

mainly transition point mutations (Phear, Nalbantoglu & Meuth, 1987). In a molecular

study, Phear et al. sequenced several adenine phosphoribosyltransferase gene mutant

clones derived from excess thymidine treatment (Phear, Nalbantoglu & Meuth, 1987).

Sequencing data revealed mainly C→T transition mutations at sites that were followed by

thymidine. For example, mixed sequences containing cytosine and thymidine were

mutated following excess thymidine treatment to runs of thymidine.

A similar affect was observed in thymidine depleted conditions (high cytosine to

thymidine ratio); T→C transition mutations were reported when cytosine was the 3’

nucleotide in sequence. The authors proposed the local sequence context influences the

position of base substitutions. Typically, the nucleotide in excess is downstream of most

misincorporated nucleotides. This mechanism of mutation by excess nucleotides is known

as the next nucleotide effect model (Phear, Nalbantoglu & Meuth, 1987, Bernardi & Ninio,

Page 51: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

51

1978); an outcome based on a compromise between the proof-reading activity of the 3’ to

5’ exonuclease activity and the rate of polymerisation; when a misincorporated nucleotide

is followed 3’ by a nucleotide in excess, polymerisation of this nucleotide is favoured rather

than excision of the incorrect one thereby causing a point mutation (Fersht, 1979).

Nucleotide pool perturbations have also been reported to increase the susceptibility

of Chinese hamster ovary (CHO) cells to mutation by the DNA alkylating agents ethyl

methanesulphonate (EMS) and N-methyl-N’-nitro-N-nitrosoguanidine (Meuth, 1981a,

Peterson et al., 1978). For instance, Meuth et al. (Meuth, 1981a) reported an imbalance of

dCTP to dTTP increased the cytotoxic and mutagenic activity of EMS. When intracellular

dCTP concentrations were 10 fold greater than dTTP, the concentration of EMS that

induced 10% survival was 3mM; the converse resulted in 10% survival with 1mM EMS

treatment.

In CHO cells, mutation at the sodium-potassium exchanging adenosine

triphosphatase gene co-treated with 0.4mM EMS and 100µM thymidine produced a similar

mutagenic response as 1.6mM EMS treatment alone (Meuth, 1981a). Increasing the

thymidine concentration was coupled to an increase in the mutagenicity of EMS. The

authors proposed that an increased dCTP to dTTP ratio can protect cells from the lethality

of EMS treatment by reducing the misincorporation frequency of thymidine at O6-

alkylguanine residues (Meuth, 1981a). Competitive incorporation of the correct nucleotide

(dCTP) prevents a transition mutation that would occur if dTTP were incorporated. Thus,

increasing the dTTP to dCTP ratio facilitates misincorporation of thymidine to

O6-alkylguanine enhancing the cytotoxicity and mutagenicity of EMS.

Page 52: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

52

1.4. Aims and objectives

Subjecting ASO pharmaceuticals to the standard battery of genotoxicity testing is not

appropriate due to the putative mechanisms of oligonucleotide-genomic DNA interactions;

the sequence specific nature for triplex and D-loop formation makes it highly unlikely for

routine tests to detect genomic insult. As antiretroviral nucleoside analogues used as part

of HAART are not used in ASO design the established genotoxicity associated with these

nucleoside analogues cannot be used to extrapolate the genotoxic risk posed by

nucleotide analogues released as degradation products from chemically modified ASO.

Hypothesis under investigation: ASO as therapeutic modalities do not carry a

genetic toxicology risk.

Thus, this study has aimed to:

Characterise the binding affinity and genotoxicity of a TFO targeting genomic DNA

in viable human cells.

Design and evaluate the genotoxicity of an ASO capable of D-loop formation at

genomic DNA and inform the modulatory roles of distinct DNA repair pathways in

the mechanism of mutation.

Inform the potential for nucleotide analogues, derived from ASO degradation, to be

utilised by DNA polymerase and cause mutation.

The outcomes from this study can be used to highlight the risk posed by oligonucleotide

based pharmaceuticals that may be used in human therapies and the requirement for

Page 53: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

53

novel pre-clinical safety assessment methods for such drug candidates on a case-by-case

basis.

Page 54: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

54

Chapter 2

Materials and Methods

2.1. Oligonucleotides

All oligonucleotides contained a deoxyribose sugar moiety. Oligonucleotides were

synthesised by Sigma Genosys (Haverhill, UK) and purified (>95%) by reverse phase

cartridge. Psoralen and biotin conjugated oligonucleotides were HPLC purified. All

oligonucleotides were chemically modified to contain 4 terminal phosphorothioate linkages

at the 5’ and 4 at the 3’ ends to increase nuclease resistance (Kenner et al., 2002), unless

otherwise stated. Oligonucleotides were supplied lyophilised and stored at -20°C. On the

day of use, dried oligonucleotides were dissolved to a final concentration of 200μM using

sterile DNase/RNase free water (Invitrogen, Paisley).

2.1.1. Triple-helix forming oligonucleotide design

A purine motif TFO, twenty seven nucleotides in length (TFO27), was designed to target a

purine rich segment within exon 3 of the human Hypoxanthine-guanine

phophoribosyltransferase (HPRT) locus (gene ID 3251). The TFO27 triplex target

sequence (nucleotides 113-139 from exon 3) is highlighted in Table 2.1. Antiparallel

binding of TFO27 to the triplex target sequence forms A•A:T and G•G:C triplets stabilised

by reverse Hoogsteen hydrogen bonds as discussed in further detail in section 1.3.1. A

scrambled oligonucleotide (SCR27), with the same base composition as TFO27 but in a

random order was employed as an oligonucleotide control throughout experiments.

SCR27 has no sequence similarity to the HPRT locus, confirmed using National Center for

Biotechnology Information-Basic Local Alignment Search Tool (NCBI-BLAST). TFO

sequences are listed in Table 2.1.

Page 55: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

55

2.1.2. Displacement loop/ antisense oligonucleotide design

An oligonucleotide, twenty seven nucleotides in length, was engineered to bind to the

HPRT gene which was complementary to the TFO27 triplex target sequence via D-loop

formation (Dloop27). Dloop27 contains four terminal phosphorothioate linkages at either

end to facilitate nuclease resistance (Kenner et al., 2002) (Table 2.2).

In a similar approach using NCBI-BLAST, an ASO, twenty three nucleotides in

length, was engineered to bind complementary to the A disintegrin and metalloproteinase

with thrombospondin type 1, motif 3 (ADAMTS3) gene (gene ID 9508) mRNA as well as

contain sequence homology to the HPRT locus except a single mismatched base (AD3-

HPRTPM). AD3-HPRTPM was engineered to act as an ASO to modulate ADAMTS3

protein expression. Additional to this, it was proposed that off-target genotoxicity caused

by the presence of AD3-HPRTPM would result in point mutation at the site of the

mismatched base within the HPRT locus. The single point mutation would result in a G>T

transversion changing the codon from AGA (arginine) to ATA (isoleucine). AD3-HPRTPM

mediated mutation at the HPRT locus was informed using the HPRT mutation assay

described in section 2.4. AD3-HPRTPM was chemically modified to contain four terminal

phosphorothioate linkages at either end to facilitate nuclease resistance (Kenner et al.,

2002) (Table 2.2).

Further to AD3-HPRTPM, a positive control oligonucleotide (Val-HPRTPM) was

engineered to contain sequence homology to the HPRT locus, except a single mismatched

base (Table 2.2). Point mutation at the site of the mismatched base in Val-HPRTPM has

previously been reported to result in a mutant HPRT phenotype (Meng et al., 2002).

Hence, Val-HPRTPM is predicted to cause a C>T transition mutation in exon 3 (position 61

from exon 3 or position 202 in mRNA) changing the codon from leucine to phenylalanine.

Page 56: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

56

Throughout D-loop studies, a negative control oligonucleotide (control), which

contained the same base composition as AD3-HPRTPM but in a random order, was

employed (Table 2.2).

2.1.3. Serum stability of oligonucleotides

The serum stability of TFO27 and SCR27 (Table 2.1) was determined as previously

described by (Grunweller et al., 2003) with slight modification. Oligonucleotides were

diluted in RPMI 1640 media supplemented with 10% horse serum to a final concentration

of 10μM in DNase/RNase free eppendorf tubes in triplicate. Samples were incubated at

37°C and 10μL aliquots removed at 0 minutes (mins), 15 mins, 30 mins, 1 hour (h), 2h, 4h,

8h and 24h. Reactions were terminated by 10μL addition of urea-TBE loading buffer

(1xTBE, 7M urea and 0.1%w/v bromophenol blue) and subsequent freezing in -80°C.

Oligonucleotides were resolved on 1.5% agarose gels containing 0.5μg/mL ethidium

bromide at 80 volts (V) for 1h. Gels were visualised under UV using BioRad ChemiDoc

XRS+ and bands quantified by densitometry using Image Lab software. Data was

processed using GraphPad prism 5.0 (GraphPad software, USA). Average band

intensities were normalised to the average band intensity of the 0h oligonucleotide.

Page 57: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

57

Table 2.1. TFO sequences

Sequence (5’ to 3’) Length

TFO target sequence AAAGCACTGAATAGAAATAGTGATAGA 27

TFO27 A*G*A*T*AGTGATAAAGATAAGTCAC*G*A*A*A 27

SCR27 A*A*G*A*ATTAGAGCAGTAGTAACAG*A*A*T*A 27

PO-TFO27 AGATAGTGATAAAGATAAGTCACGAAA 27

6FAM-TFO27 6FAM - A*G*A*T*AGTGATAAAGATAAGTCAC*G*A*A*A 27

Pso-TFO27-biot Psoralen (C6) - A*G*A*T*AGTGATAAAGATAAGTCAC*G*A*A*A – biotin (C2) 27

Pso-SCR27-biot Psoralen (C6) - A*A*G*A*ATTAGAGCAGTAGTAACAG*A*A*T*A – biotin (C2) 27

* indicates a phosphorothioate linkage. C6 indicates a 6 carbon linker to the 5’ phosphate moiety. C2 indicates a 2 carbon linker to the 3’ hydroxyl moiety.

Table 2.2. D-loop forming oligonucleotides

Sequence (5’ to 3’) Length

Dloop27 T*C*T*A*TCACTATTTCTATTCAGTG*C*T*T*T 27

AD3-HPRTPM A*C*A*G*TCATAGGAATGGATa*T*A*T*C 23

Val-HPRTPM T*A*G*C*CCCCCTTGAaCAC*A*C*A*G 21

Control A*C*C*T*TGATGGCAAATAGGT*A*A*T*A 23

* indicates a phosphorothioate linkage. Lowercase letters represent the site of the mismatched base

Page 58: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

58

2.1.4. Oligonucleotide third strand binding assay

Third strand binding efficiency of TFO27 and SCR27 to the HPRT target motif enclosed

within a synthetic duplex was measured using EMSA. Duplex DNA 35 base pairs (bp) in

length was made by annealing HPRT-S-35 and HPRT-AS-35 (Table 2.3) in annealing

buffer 1 (50mM sodium chloride) at 60°C for 30 mins followed by cooling at room

temperature for 60 mins.

Serial dilutions of TFO27 or SCR27 were incubated with a fixed concentration of

duplex DNA (0.2µM) in either triplex binding buffer (10mM Tris, pH 7.2 and 10mM MgCl2)

or triplex binding buffer supplemented with 2.5mM spermidine in a 20 microlitre (µL)

reaction volume at room temperature for 3-4 h. The negative control in each experiment

was duplex DNA without oligonucleotide. Samples (20μL) were mixed with 4μL of 6x gel

loading buffer (0.25%w/v bromophenol blue and 40% w/v sucrose in water) then subject to

electrophoresis through 15% polyacrylamide gel at 70-80V in triplex running buffer

(17.8mM Tris pH 7.2, 17.8mM boric acid and 10mM MgCl2) on ice. Gels were stained with

0.5µg/mL ethidium bromide for 25-30 mins followed by a 5 minute wash in demineralised

water. Gels were visualised using the Kodak IS4000MM with an excitation/emission of 535

nanometers (nm)/600nm.

2.1.5. Detecting triplex formation at genomic DNA

The ability of TFO27 to bind the triplex target sequence in genomic DNA (gDNA) within

exon 3 of the HPRT locus was investigated in an approach adapted from Besch et al.

(Besch et al., 2004) (Fig. 2.1).

For this, TFO27 was conjugated to a psoralen moiety on the 5’ end via a 6 carbon

linker and to a biotin moiety on the 3’ end via a 2 carbon linker (Pso-TFO27-biot; Table

2.1). The control oligonucleotide, SCR27, was modified the same way (Pso-SCR27-biot;

Table 2.1).

Page 59: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

59

Two or sixteen micrograms of gDNA extracted from human lymphoblastoid TK6

cells (described in section 2.2.3) was incubated with 20μM Pso-TFO27-biot or Pso-

SCR27-biot in triplex binding buffer (10mM Tris, pH 7.2 and 10mM MgCl2) in a 25μL

volume (Fig.2.1, step A). Samples were allowed to incubate at room temperature for 3h.

Following this, samples were irradiated with UVA (5J/cm2) to covalently crosslink the

psoralen moiety to the 5’-AT-3’ sequence upstream of the HPRT triplex target sequence

within exon 3 (Fig. 2.1, step A).

Following covalent linkage of Pso-TFO27-biot to the gDNA, samples were digested

overnight at 37°C with 10-15 units of Bgl II restriction enzyme in a 30μL volume. Bgl II

digestion of gDNA yields a 3.4kb fragment of the HPRT locus containing the covalently

bound Pso-TFO27-biot (Fig.2.1, step B).

To remove unbound biotinylated oligonucleotide from the reaction mixture (Fig. 2.1,

step C), samples were either purified through the Qiagen DNA extraction column (as

described in section 2.2.3) or subject to electrophoresis through a 0.6%w/v agarose gel

containing 0.5μg/mL ethidium bromide for 10-15 mins (agarose gel electrophoresis is

described in section 2.5.4). Gels were visualised under UV and bands excised and purified

using the Qiaquick gel extraction kit as described in section 2.5.6.

Meanwhile, the desired concentration of BioMag® streptavidin coated paramagnetic

beads (Qiagen, Crawley) were aliquoted into a 0.2mL nuclease free polymerase chain

reaction (PCR) tube. Beads were pelleted using a magnet for 1 minute and the

supernatant carefully discarded. Pelleted beads were resuspended in biotin binding buffer

(20mM Tris, pH 7.8, 1M NaCl, 1mM ethylenediaminetetraacetic acid (EDTA), 0.02%v/v

Triton X-100). To isolate the biotinylated 3.4kb HPRT fragment, the required concentration

of streptavidin beads were mixed with 70μL of purified gDNA (containing the covalently

linked Pso-TFO27-biot) in biotin binding buffer in a 100μL volume for 30 mins at room

Page 60: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

60

temperature (Fig. 2.1, step C). Samples were agitated every 8-10 mins to avoid beads

sedimenting to the bottom of the reaction tube.

Following incubation, beads were pelleted using a magnet for 1 minute and the

supernatant discarded. Beads were gently washed three times in 50μL of DNase/RNase

free water and suspended in a final volume of 20μL. Resuspended beads bound to the

biotinylated oligonucleotide linked to a 3.4kb HPRT fragment were subject to PCR

amplification of a 364 bp stretch downstream of the TFO27 triplex target sequence using

primer pair 1 (Table 2.4) (Fig. 2.1, step D). PCR amplification is described in section 2.5.

Page 61: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

61

Figure 2.1. Overview of the methodology used to detect triplex formation within genomic DNA. In brief, TFO27 conjugated on 5’ to psoralen and 3’ biotin was incubated with genomic DNA to allow triplex formation within the HPRT locus, then UVA irradiated to form psoralen crosslinks with DNA (A). Genomic DNA containing covalently linked TFO27 was digested overnight with Bgl II restriction enzyme (B). Samples were then purified using a column or by electrophoresis through a gel to remove free biotinylated TFO27 (C). Purified samples were then incubated with streptavidin coated paramagnetic beads to isolate genomic DNA covalently linked to Pso-TFO27-biot (C). Following several washes, samples were PCR amplified using primers specific to HPRT (D).

Incubate and UVA

Bgl II digestion

Purification and Isolation

Magnet

PCR

A

B

C

D

Page 62: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

62

2.1.6. DNase I protection assay

The ability of TFO27 to protect the triplex target sequence within a synthetic duplex was

measured by means of a DNase I protection assay, as previously reported by (Washbrook

& Fox, 1994) with slight modification.

Duplex DNA, 50 bp in length, was made by annealing oligonucleotides HPRT-S-50

and HPRT-AS-50 as described in section 2.1.4 (Table 2.3). Duplex DNA (5μM) was

incubated overnight at 37°C with 2μM or 5μM TFO27 in triplex binding buffer (10mM Tris,

pH 7.2 and 10mM MgCl2) supplemented with 2.5mM spermidine, in a final reaction volume

of 20μL. Duplex DNA incubated without oligonucleotide and with 5μM SCR27 was used as

a control.

Complexes were digested using 5μL of 0.01units/mL DNase 1 for 15 mins at room

temperature. Digestion was stopped by adding 10μL formamide containing 10mM EDTA.

Products of digestion were mixed with 6x gel loading buffer and resolved on 10%w/v

polyacrylamide (19:1) gels at 60V for 7 h in triplex running buffer (17.8mM Tris pH 7.2,

17.8mM boric acid and 10mM MgCl2). Gels were stained with 0.5μg/mL ethidium bromide

and visualised under UV using BioRad ChemiDoc XRS+ with Image Lab software.

2.1.7. Quadruplex forming ability of SupFG1

The quadruplex forming ability of the SupFG1 purine stretch previously described (Kalish

et al., 2005, Datta et al., 2001, Faruqi et al., 2000, Vasquez et al., 1999, Wang, Seidman &

Glazer, 1996) was assessed using a 40bp duplex. Duplex DNA was made by annealing

SupF-S and SupF-AS or SupF-S-4MM and SupF-AS-4MM (Table 2.3) as described in

section 2.1.4. A fixed concentration of duplex DNA (0.2μM) was incubated with serial

dilutions of targeting oligonucleotide (AG30 or AG30-4MM; Table 2.3) in quadruplex buffer

(10mM Tris pH 7.2, 0.1mM MgCl2 and 140mM KCl) in a 20µL reaction volume for 3h at

room temperature. Samples were then electrophoresed through 15% polyacrylamide gel at

Page 63: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

63

70-80V in triplex running buffer (17.8mM Tris, pH 7.2, 17.8mM boric acid and 10mM

MgCl2). Gels were stained with 0.5μg/mL ethidium bromide and visualised using a Kodak

Imaging Station 4000MM with an excitation/emission of 535nm/600nm. Bands were

quantitated by densitometry using Kodak Imaging software.

Page 64: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

64

Table 2.3. Oligonucleotides used in gel retardation assays

Sequence (5’ to 3’) Length

AG30 AGGAAGGGGGGGGTGGTGGGGGAGGGGGAG 30

AG30-4MM AGGAAGGAGGAGGTGGTGGAGGAGGAGGAG 30

HPRT-S-35 CATCAAAGCACTGAATAGAAATAGTGATAGATCCA 35

HPRT-AS-35 TGGATCTATCACTATTTCTATTCAGTGCTTTGATG 35

HPRT-S-50 CTGGATTACATCAAAGCACTGAATAGAAATAGTGATAGATCCATTCCTAT 50

HPRT-AS-50 ATAGGAATGGATCTATCACTATTTCTATTCAGTGCTTTGATGTAATCCAG 50

SupF-S GAGGGGGAGGGGGTGGTGGGGGGGGAAGGATTCGAACCTT 40

SupF-AS AAGGTTCGAATCCTTCCCCCCCCACCACCCCCTCCCCCTC 40

SupF-S-4MM GAGGAGGAGGAGGTGGTGGAGGAGGAAGGATTCGAACCTT 40

SupF-AS-4MM AAGGTTCGAATCCTTCCTCCTCCACCACCTCCTCCTCCTC 40

Underlined sequence highlights the position of the replaced guanine nucleotide

‘S’ means sense strand of duplex; ‘AS’ means antisense strand of duplex

Bold sequence highlights the TFO27 triplex target sequence within exon 3 of the HPRT locus

Page 65: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

65

Primer Pair

Genomic or cDNA

Target sequence

Sequence (5’―> 3’)

Primer annealing

Temperature (°C)

PCR Polymerisati

on time (secs)

Length of

product (bp)

1 Forward

Reverse

Genomic HPRT locus

Intron 3

CCTGTAGGTGATTGGGCAGCCA

TGATGGGGATGGGGAAGGAGGC 66 60 364

2 Forward

Reverse Genomic

HPRT locus

exon 3

CTGCTGGATTACATCAAAGCACTG

TGAAAGCAAGTATGGTTTGCAGAGA 64 30 177

3 Forward

Reverse

Genomic

HPRT locus

exon 3

AGGGCAAAGGATGTGTTACG

AGTGGTTTCTGGTGCGACTT 58 60 1018

4 Forward

Reverse Genomic GAPDH*

GTATTGGGCGCCTGGTCAC

CTCCTGGAAGATGGTGATGG 60 30 290

5 Forward

Reverse CDNA TK

TACTGCGGGACGGCCTTGGA

GAGTGGCTGGGCAGCATGCA 64 60 837

* Primer sequences were taken from Donovan et al. (Donovan et al., 2000)

Table 2.4. Primer sequences used for PCR amplification.

Page 66: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

66

2.1.8. Primer extension assay

The ability of DNA polymerase to incorporate nucleotide analogues was investigated using

a primer extension assay extrapolated from Lacenere et al. (Lacenere et al., 2006).

Nucleotide analogues were obtained from Jena Bioscience, Germany. Unmodified

nucleotides were obtained from Sigma Aldrich, Poole.

Primed template was made by annealing template strand with the complementary 5’

6-carboxyfluorescein labelled primer strand (25nt) in annealing buffer 2 (75mM Tris, pH

7.2 and 75mM NaCl). Oligonucleotide sequences are listed in Table 2.5. The mixture was

heated to 95°C for 5 mins and allowed to cool for 1h at room temperature. A typical

reaction mixture contained 25nM of the primed template, 4U Tfi DNA polymerase, 3.5mM

MgCl2, 1x Tfi PCR buffer and nucleotide in a 20μL volume. As a negative control in

experiments, nucleotide was omitted. Reactions were allowed to incubate for the required

length of time at 72°C to permit extension of the primed template. Following this, 5μL of

1M EDTA was added to stop the reaction. Primed template in the reaction mixture was

then denatured by adding 25μL of urea gel loading buffer (1.5M sucrose, 7M urea, 10mM

EDTA, 0.1% bromophenol blue) and heated at 95°C for 5 mins with immediate transfer to

ice slurry. Samples were then subject to electrophoresis through a 14% 8M urea-

polyacrylamide gel at 120V for 3h, unless stated otherwise. 6-carboxyfluorescein (6FAM)

labelled primer strand was visualised at 490nm/530nm using the Kodak imaging station

4000MM.

Page 67: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

67

Table 2.5. Oligonucleotides used in primer extension assays

Sequence (5’ to 3’) Length

Primer strand 6FAM-TGTGTCTGGGCTTTTGGTTTGTGGG 25

Template strands

A1-template TCCCACAAACCAAAAGCCCAGACACA 26

A5-template TTTTTCCCACAAACCAAAAGCCCAGACACA 30

C5-template GGGGGCCCACAAACCAAAAGCCCAGACACA 30

Underlined and bold sequence highlights the position of the overhang

Page 68: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

68

2.2. General cell culture

RPMI 1640, heat inactivated horse serum (HIHS), heat inactivated foetal bovine serum

(HIFBS), L-glutamine and penicillin/streptomycin and RNase/DNase free water (sterile)

were obtained from Invitrogen (Paisley, UK) while the remaining reagents were obtained

from Sigma Aldrich (Poole, UK), unless otherwise stated. Tissue culture consumables

were obtained from VWR (Leicestershire, UK). All cell culture procedures were carried out

in a Class II biological safety cabinet. Cell lines were grown in a humidified incubator at

37°C, 5% CO2.

2.2.1. Human lymphoblastoid TK6 cells

Human lymphoblastoid TK6 cells were obtained from ATCC (Teddington, UK). TK6 cells

were cultured in RPMI 1640 media containing 10%v/v HIHS, 2mM L-Glutamine,

100units/mL penicillin and 100µg/mL streptomycin, referred to as complete media

(R10HIHS). Culture media without serum is referred to as incomplete media (R0).

TK6 cells were resuscitated from a frozen vial containing 2-3x106 cells stored in the

vapour phase of liquid nitrogen. In brief, vials of frozen cells were rapidly thawed in a pre-

warmed water bath (37°C). The cells were transferred to a 15mL centrifuge tube and

centrifuged at 200xg for 5mins. The cell pellet was slowly resuspended drop-wise in pre-

warmed R10HIHS media and transferred to a 75cm2 flask followed by further drop-wise

addition of pre-warmed R10HIHS media up to a final volume of 10mL and incubated as

described above.

TK6 cells were maintained in exponential growth by passaging populations between

5x104/mL and 1x106/mL. In brief, cultures were gently shaken to ensure single cell

suspensions. Accurate quantification of viable cells was carried out using the trypan blue

exclusion assay. An 80μL aliquot of the single cell suspension was mixed with 20μL of

trypan blue solution (0.4%w/v) with incubation at room temperature for 1-2 mins to allow

Page 69: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

69

uptake/exclusion of the dye. An aliquot (10μL) was transferred to a Neubauer

haemocytometer by capillary action. Viable cells were scored as those which excluded the

blue dye and appear clear, as opposed to non-viable cells appearing blue. The cell

concentration per millilitre was determined as follows:

The average count from 4 grids x (1/10-4; volume of grid cm3) x dilution factor.

The Beckman Coulter counter Z2 series (Beckman Coulter UK ltd) was used for

rapid quantification of cell populations during high throughput experiments. The lower and

upper size for TK6 cells were found to be optimum between 7.45 micrometers (μm) and

18.62μm (cell counts were in consensus with the trypan blue exclusion assay using the

haemocytometer). In the first instance, Isoton II diluent (excluding cells) was counted to

ensure the background count was below 150. As described above, cultures were briefly

shaken to ensure single cell suspensions as multiple cell clumps are not differentiated by

the coulter counter. A 0.5mL aliquot of single cell suspension was added to 9.5mL Isoton II

diluent, shaken and counted. The concentration of cell suspensions were adjusted to

maintain exponential growth, as described above.

Human TK6 cells were grown for up to 7 days to accumulate sufficient cells to store

aliquots with the same passage number as working stocks for subsequent mutation

assays. This was to reduce variability between experiments. For cryopreservation, 1mL of

2-3x106 TK6 cells in R10HIHS media containing 10%v/v dimethylsulphoxide (DMSO) were

transferred to 1.5mL cryovials and stored in a container overnight at -80°C before being

transferred to liquid nitrogen. Cells were thawed as described above for subsequent

experiments.

Page 70: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

70

2.2.2. Human colon epithelial HCT116 cells

Human colon epithelial cells were obtained from ATCC. HCT116 cells were cultured in

RPMI 1640 media containing 10% HIFBS, 2mM L-Glutamine, 100units/mL penicillin and

100µg/mL streptomycin, referred to as complete media (R10HIFBS). Culture media without

serum is referred to as incomplete media (R0).

HCT116 cells were resuscitated as described in section 2.2.1. Confluent flasks

were passaged by aspirating the media and washing cells with 5mL phosphate buffered

saline (PBS). In order to detach cells, 3-4mL of Trypsin-EDTA (1x; 0.05%) was added per

75cm2 flask for 3-5 mins at 37°C. Flasks were examined by light microscope to ensure all

cells had become detached. Trypsin was inactivated by adding an equal volume of

R10HIFBS. Detached cells were transferred to 15mL centrifuge tubes and centrifuged at

200xg for 5 mins. Cell pellets were then suspended in R10HIFBS and counted using the

haemocytometer as described in section 2.2.1. and maintained in exponential growth by

seeding at least 0.5x106-1x106 cells per 75cm2 flask with regular passaging every 2-3

days.

2.2.3. Genomic DNA extraction from cultured cells

gDNA was extracted from cultured cells using the QIAamp DNA Blood mini kit (Qiagen,

Crawley) according to manufacturer’s instructions. Lysis buffer AL, wash buffers AW1 and

AW2, proteinase K solution and elution buffer AE were provided in the manufacturer’s kit.

In brief, up to 5x106 TK6 cells were centrifuged at 200xg for 5 mins. The pellet was

suspended in 200μL PBS and transferred to a 1.5mL eppendorf tube. Qiagen proteinase K

(20μL) was added along with 200μL lysis buffer AL. The samples were then mixed by

pulse vortexing and incubated at 56°C for 10 mins. Samples were briefly centrifuged and

200μL ethanol (>99.6%) added with gentle inverting to precipitate gDNA. The mixture was

applied to a spin column in a collection tube (provided with the manufacturer’s kit) and

Page 71: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

71

centrifuged at 6,000xg for 1 min. The spin column was placed in a fresh collection tube

and the filtrate discarded. gDNA in the spin column was washed with 500μL ethanol wash

buffer AW1 and centrifuged at 6,000xg for 1 min. The spin column was placed in a fresh

collection tube and the filtrate discarded. Five hundred microliters (500µL) ethanol wash

buffer AW2 was added to the spin column and centrifuged at 20,000xg for 3 mins. The

spin column was placed into a fresh RNase/DNase free eppendorf tube and the filtrate

discarded. The spin column was centrifuged once more at 20,000xg for 1 minute to

remove traces of ethanol. DNA was eluted out of the spin column by addition of 50-200μL

buffer AE and incubation for 5 mins at room temperature followed by centrifugation at

6,000xg for 1 minute. DNA was quantified using the NanoDrop ND1000

spectrophotometer (Labtech, Sussex). Samples with 260nm/280nm ratios above 1.8 were

used in subsequent experiments and stored at -20°C.

2.2.4. RNA extraction

Cellular RNA was extracted using the SV total RNA Isolation System (Promega,

Southampton) according to manufacturer’s protocol. RNA lysis buffer, RNA dilution buffer,

RNA wash solution, DNase I and DNase stop solution were provided with the

manufacturer’s kit.

In brief, cells were pelleted at 200xg for 5 mins and washed in cold PBS. Cell

pellets were suspended in 175μL RNA lysis buffer and transferred to a 1.5mL eppendorf

tube with the subsequent addition of 350μL RNA dilution buffer. Extracts were inverted

several times and heated at 70°C for 3 mins. Samples were centrifuged at 12,000xg for 10

mins and the supernatant collected. Ethanol (200µL, >99.6%) was added to the

supernatant and inverted several times. Samples were then transferred to a spin column

with a collection tube (provided with the manufacturer’s kit) and centrifuged at 12,000xg for

1 min. Columns were washed with 600μL ethanol based RNA wash solution and again

Page 72: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

72

centrifuged. DNase (50μL) was added to each column for 15 mins at room temperature

followed by the addition of 200μL DNase stop solution. Columns were then centrifuged at

12,000xg for 1 minute. Columns were washed twice with RNA wash solution. The spin

column was placed in a fresh eppendorf tube and RNA eluted by the addition of RNase

free water followed by centrifugation at 14,000xg for 1 minute. Samples were then stored

at -70°C until the time of analysis. RNA samples were quantified as described in Section

2.2.3.

2.2.5. Protein extraction and quantification

Total cell protein content was extracted as described previously (Sambrook, 2001). In

brief, cell pellets were suspended in 100μL freshly made master lysis buffer (1μL Halt

protease inhibitor + 99μL lysis buffer [150mM NaCl, 0.1%v/v igepal, 1mM Tris, pH 7.4 and

1mM EDTA]) on ice for 20 mins. Samples were then centrifuged at 10,000xg for 10 mins at

4°C. Supernatants were collected in fresh eppendorf tubes and stored at -20°C.

Total protein was determined using the bicinchoninic acid (BCA) assay (Thermo

Scientific, Surrey UK), according to manufacturer’s protocol. In brief, protein extracts were

diluted 10 or 20 fold and 10μL transferred to a single well of a 96 well plate. Additionally,

known concentrations of bovine serum albumin were used to produce a standard curve.

Samples were overlaid with 200μL of BCA working reagent (made by mixing 50 parts of

reagent A with 1 part reagent B provided with the manufacturer’s kit) and shaken gently for

30 seconds. Plates were then incubated for 30 mins at 37°C and allowed to cool to room

temperature. The absorbance at 562nm was measured and protein concentration in

extracts was determined from the standard curve.

Page 73: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

73

2.3. Cell treatments

2.3.1. MSH2 protein knockdown in TK6 cells using Silencer Select siRNA

Where MSH2 protein knockdown was required, exponentially growing TK6 cells were

treated for 24h with Silencer Select siRNA (Applied Biosystems, UK) using siPORT NeoFX

transfection reagent (Applied Biosystems, UK). The following day, TK6 cells were treated

with oligonucleotide (section 2.3.4).

As the manufacturer’s protocol is designed for low volume transfection ie 2.5mL,

transfection was scaled to treat 8x106 TK6 cells in a 50mL volume to provide sufficient

cells for subsequent mutagenicity studies. To treat 8x106 TK6 cells in a 50mL volume with

10nM MSH2 siRNA, 500μL 1μM Silencer Select siRNA was diluted up to 4mL with R0

media. The siRNA dilution was mixed with 4mL of diluted siPORT (210μL of siPORT

NeoFX diluted up to 4.2mL with R0) in a flask and allowed to stand for 10 mins. The

siRNA/siPORT transfection complexes were overlaid with 8x106 TK6 cells in a 42mL

volume and allowed to incubate for 24h at 37°C, 5%CO2. As a negative control, the

siRNA/siPORT mixture was replaced with R0 culture media

Following siRNA transfection, TK6 cells were harvested by centrifugation and

suspension in R10HIHS media. siRNA treated TK6 cells were counted using the coulter

counter (section 2.2.1) and diluted for oligonucleotide treatment (section 2.3.4).

To confirm Silencer Select siRNA mediated knockdown of MSH2 protein

expression, 5x106 TK6 cells were removed and washed in ice cold PBS for immunoblot

analysis at 24h, 48h and 72h after treatment, unless stated otherwise. Protein extraction

was performed as described in section 2.2.5. Protein expression was determined by

immunoblot analysis as described in section 2.6.

Page 74: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

74

2.3.2. Induction of RAD51 protein expression in TK6 cells

Exponentially growing TK6 cells were treated with methyl methanesulphonate (MMS) for

24h to induce RAD51 protein expression. In a 50mL volume, 8x106 TK6 cells were treated

for 24h with 0μg/mL, 0.1μg/mL, 0.2μg/mL or 0.5μg/mL MMS (0.1%v/v DMSO solvent).

Following treatment, TK6 cells were washed in R10HIHS media and maintained in

exponential growth for up to 72h. Aliquots of treated TK6 cells (5mL) were removed at 2h,

4h, 8h, 24h, 48h and 72h and then washed in 1mL PBS and pellets stored at -20°C for

protein extraction (section 2.2.5). RAD51 protein expression was determined by

immunoblot analysis as described in section 2.6.

The concentration of MMS found to significantly induce RAD51 protein expression

was used to pre-treat TK6 cells for 24h the day before oligonucleotide treatment (section

2.3.4).

2.3.3 Oligonucleotide transfection efficiency using siPORT NeoFX and NTER in TK6

cells

siPORT NeoFX (Applied Biosystems, UK) and NTER (Sigma Aldrich, UK) transfection

facilitators were evaluated for transfection efficiency in human TK6 cells using 6-

carboxyfluorescein labelled TFO27 (6FAM-TFO27; see Table 2.1). The ratio of

transfection facilitator (μL) to oligonucleotide (μg) was explored for optimal delivery of 1μM

and 2μM 6FAM-TFO27. The ratios of oligonucleotide (μg): transfection facilitator (μL)

examined were 2:1, 1:1, 1:2 and 1:4.

For siPORT NeoFX, the respective volume of transfection reagent was diluted in R0

media up to a volume of 10μL, in triplicate wells of a 96-well plate and allowed to stand for

10 mins. Following this, 6FAM-TFO27 was diluted in serum free media and 10μL added to

the siPORT mixture in each respective well and allowed to incubate for 10 mins to allow

complex formation.

Page 75: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

75

For NTER, the respective volume of transfection reagent followed by 6FAM-TFO27

was diluted in serum free media up to a volume of 20μL, in triplicate wells of a 96-well

plate. Complex formation was allowed to occur by incubation at room temperature for 20

mins.

Meanwhile, TK6 cells were counted using the coulter counter (section 2.2.1) and

adjusted to 1.9x106 cells/mL in R10HIHS media. An 80μL aliquot of cells was added to each

well in the 96-well plate and transfection was carried out at 37°C for 4 h.

As a blank for each concentration of oligonucleotide, a mock transfection was

carried out in parallel as described above, except the oligonucleotide was replaced with

water.

Following transfection, TK6 cells were pelleted at 200xg for 5 mins, washed in PBS

twice and re-suspended in 100μL PBS. Fluorescence was quantified using the Polarstar

Galaxy (BMG labtech, UK) with excitation/emission around 490nm/520nm. Following

measurement, TK6 cells in each well were counted using the trypan blue exclusion assay

on the haemocytometer (section 2.2.1) to normalise arbitrary fluorescence units per viable

104 cells.

2.3.4. Treatment of TK6 cells with oligonucleotide

Oligonucleotides do not require metabolic activation like meat derived carcinogens and so

cell treatments were conducted in the absence of a metabolic activation system (Yu et al.,

2009, Boobis et al., 1994).

In the first instance, oligonucleotides were thawed at room temperature while the

siPORT NeoFX was allowed to equilibrate to room temperature. The optimal volume of

siPORT required for transfection of oligonucleotide was determined previously (section

2.3.3). A 2:1, DNA (μg): siPORT (μL) ratio was employed for large scale oligonucleotide

transfection.

Page 76: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

76

Exponentially growing TK6 cells (or pre-treated TK6 cells with MSH2 siRNA section

2.3.1 or MMS section 2.3.2) were counted using the coulter counter (section 2.2.1). TK6

cells were aliquoted to 50mL centrifuge tubes and centrifuged at 200xg for 5 mins. The

supernatant from cell pellets was collected. Cells were then suspended at a density of

1.9x106/mL in conditioned R3.125 media (made by diluting the cultured R10HIHS

supernatant from the cell pellets with R0). Three million cells (1.6mL) from the cell

suspension were aliquoted to 15mL centrifuge tubes per culture. To treat 10x106 cells,

5.6mL of cells were aliquoted per tube.

Meanwhile, the siPORT NeoFX was diluted in R0 media and mixed with

oligonucleotide to allow complex formation. For example, to transfect 3x106 cells with 5μM

oligonucleotide per culture, 108μL of siPORT was mixed with 542μL of R0 media and

allowed to stand for 10 mins. Oligonucleotide (158μL) was diluted with 472μL of R0 media

and overlaid with 630μL of the siPORT mixture. The oligonucleotide/siPORT mixture was

allowed to incubate at room temperature for 10 mins to allow complex formation. Following

this, 0.4mL of the oligonucleotide/siPORT mixture was added to 1.6mL of cells (3x106) or

scaled further such that 1.4mL of oligonucleotide/siPORT mixture was added to 5.6mL of

cells to treat 10x106.

To measure the spontaneous mutant frequency at the HPRT and TK (gene ID

7083) loci, TK6 cells were treated as above, except the oligonucleotide/siPORT NeoFX

mixture was replaced with R0 media. As a positive control, 5µg/mL EMS or 1µM 4-

nitroquinoline-1-oxide (4NQO) were used (0.1%v/v DMSO solvent).

Transfection was allowed to occur for 4h at 37°C, 5%CO2. Following this, cells were

pelleted at 200xg for 5 mins and washed with 5mL of R10HIHS media. Cell pellets were

then suspended up to 10mL and transferred to a 75cm2 flask. Flasks were allowed to

incubate overnight at 37°C, 5%CO2.

Page 77: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

77

TK6 cells were counted using the coulter counter (section 2.2.1) and adjusted to

3x105/mL in a minimum volume of 10mL R10HIHS. Treated cultures with cell counts below

3x105/mL were not adjusted. Cells were counted daily up to Day 3 to determine the

relative suspension growth (RSG) and plated to determine the TK mutant frequency as

described in section 2.4.1. RSG is a measure of cell death and proliferative ability of cells

following treatment and calculated as previously described (Clements, 2000, Clive et al.,

1995). Cells were also maintained in exponential growth up to Day 7 to determine the

HPRT mutant frequency as described in section 2.4.2.

2.3.5. Treatment of TK6 cells with nucleosides and nucleotides

Exponentially growing TK6 cells were counted using the coulter counter (section 2.2.1).

TK6 cells were aliquoted to 50mL centrifuge tubes and centrifuged at 200xg for 5 mins.

The supernatant from cell pellets was collected. Cells were then suspended at a density of

8x105/mL in conditioned R10 media (made by using the cultured R10HIHS supernatant from

the cell pellets). Four million cells (5mL) from the cell suspension were aliquoted into

75cm2 flasks per culture.

Nucleotides and nucleosides were supplied in a lyophilised state and stored as

recommended by the manufacturer (Jena Bioscience, Germany). On the day of use, a

weighed amount of nucleotide was dissolved in R0 media up to a 2mM solution. From this,

serial dilutions were made using R0 media so nucleotides were as 2x solutions. A 5mL

aliquot of test compound was used to supplement 5mL of cells (4x106) in R10 media into

flasks. This gave a final treatment volume of 10mL in conditioned R5 media (equal parts of

R0 and R10 media). As the negative control, 5mL of R0 media replaced nucleotides. As a

positive control, 5μg/mL EMS was used. Cells were exposed to test compounds for 24 h.

Following treatment, cells were pelleted at 200xg for 5 mins and washed with 5mL

of R10HIHS media. Cell pellets were then suspended in 10mL of R10HIHS media and

Page 78: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

78

counted using the coulter counter (section 2.2.1). Cells were adjusted to 3x105/mL in a

minimum volume of 12mL R10HIHS. Treated cultures with cell counts below 3x105/mL were

not adjusted.

Cells were counted daily up to Day 3 to determine the RSG (described in section

2.3.4) and plated to determine the TK mutant frequency as described in section 2.4.1.

Cells were also maintained in exponential growth up to Day 7 to determine the HPRT

mutant frequency as described in section 2.4.2.

2.4. HPRT and TK forward mutation assays

The HPRT and TK forward mutation assays were conducted as recommended by the

OECD guideline 476 (http://www.oecd.org/dataoecd/18/32/1948426.pdf).

2.4.1. Determining the TK mutant frequency (Day 3)

TK6 cells were counted using the coulter counter (section 2.2.1) and adjusted to 1x105/mL

in a minimum volume of 30mL of R10HIHS for a further 4 days phenotypic expression of

HPRT mutants.

To determine the TK mutant frequency, remaining TK6 cells were adjusted to

1x105/mL in 65mL of R20HIHS media. From this, a 100μL aliquot of cells (1x105/mL) was

removed and diluted up to 10mL in R10HIHS media (1000cells/mL). From this, a 360μL

aliquot was removed and added to 45mL R20HIHS media (8cells/mL). To determine cloning

efficiency following treatment and plating efficiency during mutant selection, 200μL of this

cell suspension was plated in each well of two 96 well plates (1.6cells/well). This cloning

efficiency was used to correct the RSG to give the relative total growth (RTG) as a

measure of cytotoxicity following treatment (Clements, 2000, Clive et al., 1995).

For TK mutant selection, cells adjusted to 1x105/mL in 65mL were supplemented

with 0.65mL trifluorothymidine (TFT, 400µg/mL) in subdued light. From this, a 200μL

Page 79: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

79

aliquot of cells was plated in each well of three 96 well plates (20,000 cells/ well). Plates

were incubated at 37°C, 5%CO2 for a minimum of 14 days to allow colony formation and

empty wells scored using a light box. The mutant frequency was calculated as previously

described (Clements, 2000, Clive et al., 1995). The proportion of small and large TK

mutant colonies were also noted for each treatment. A small TK mutant clone is defined as

one which has a diameter less than ¼ of the well width in the microtitre plate (Clements,

2000). An increase in small TK mutant colonies may suggest large inter-gene deletions or

chromosome aberrations involving the TK gene in these clones which may affect the rate

of growth due to the extent of genomic insult or perhaps a result of mutation within an

adjacent growth regulatory gene (Liechty et al., 1998, Applegate et al., 1990, Moore et al.,

1985, Hozier et al., 1981).

Clones selected for molecular analysis were picked for expansion in a well of a 6-

well plate. Individual clones were grown for 7-10 days in R10HIHS culture media. Following

expansion, clones were isolated, centrifuged and pellets stored at -20°C for gDNA

extraction (section 2.2.3).

2.4.2. Determining the HPRT mutant frequency (Day 7)

TK6 cells were maintained in exponential growth from Day 3 until Day 7 by routine

culturing. To determine the HPRT mutant frequency, TK6 cells were counted using the

coulter counter (section 2.2.1) and adjusted to 1x105/mL in 65mL of R20HIHS media. From

this, a 100μL aliquot of cells (1x105/mL) was removed and diluted up to 10mL in R10HIHS

media (1000 cells/mL). From this, a 360μL aliquot was then removed and added to 45mL

R20HIHS media (8 cells/mL). To determine the plating efficiency during mutant selection,

200μL of this cell suspension was plated in each well of two 96 well plates (1.6 cells/well).

For HPRT mutant selection, cells adjusted to 1x105/mL in 65mL were supplemented with

0.65mL of 500µg/mL 6-thioguanine (6-TG). From this, 200μL of cells were plated in each

Page 80: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

80

well of three 96 well plates (20,000 cells/well). Plates were incubated at 37°C, 5%CO2 for

a minimum of 14 days to allow colony formation and empty wells scored using a light box

in aseptic conditions. The mutant frequency was calculated as previously described

(Clements, 2000, Clive et al., 1995). Clones selected for molecular analysis were picked

for expansion in a well of a 6-well plate. Individual clones were grown for 7-10 days in

R10HIHS culture media. Following expansion, clones were isolated, centrifuged and pellets

stored at -20°C for gDNA extraction (section 2.2.3).

2.4.3. The in vitro micronucleus assay

The in vitro micronucleus assay was conducted as recommended by the OECD guideline

487 (http://iccvam.niehs.nih.gov/SuppDocs /FedDocs/OECD/OECD-TG487.pdf).

Following two population doublings of treated TK6 cells, 2x105 cells were removed,

centrifuged at 200xg for 5 mins and suspended in 2mL of R02P (R0 media containing

0.2%v/v pluronic F-68). Cells were mixed thoroughly then 200μL were cytospun on to clean

microscope slides at 200xg for 5 mins. Two microscope slides were used per culture.

Slides were allowed to air dry for 20 mins, then fixed in 100% methanol for 1 minute.

Slides were washed in PBS for 1 minute and then stained in acridine orange (0.12mg/mL)

for 1 minute. Stained slides were washed in PBS for 10 mins then again in fresh PBS for

15 mins. Slides were visualised using the Leitz Diaplan fluorescence microscope. One

thousand viable mononucleate cells were scored per slide whilst scoring the incidence of

micronucleated mononucleate cells, binucleate cells and micronucleated binucleate cells.

Cytotoxicity and proliferative ability of cells following treatment was informed by the relative

population doubling as described in OECD guideline 487 (http://iccvam.niehs.nih.gov/

SuppDocs /FedDocs/OECD/OECD-TG487.pdf).

Page 81: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

81

2.4.4. Treatment of HCT116 cells with oligonucleotide

HCT116 cells were detached from flasks and counted using a haemocytometer (sections

2.2.1 and 2.2.2). Cells were adjusted to 1.9x106/mL and transfected as described in

section 2.3.4 but in 25cm2 flasks. Transfection was allowed to occur for 6 h. Following this,

the transfection media was collected and replaced with 2mL of fresh R10HIFBS. The

transfection media was centrifuged at 200xg for 5 mins to pellet any unattached cells. Cell

pellets were suspended in 2mL of R10HIFBS media and replaced back into the respective

flask for incubation overnight.

2.4.5. Cytotoxicity following treatment of HCT116 cells

Following overnight incubation of treated HCT116 cells, cells were washed with PBS, and

detached using trypsin for counting using a haemocytometer (sections 2.2.1and 2.2.2).

The volume for 10,000 viable cells was added to 10mL of R10HIFBS media (1000 cells/mL).

From this, 450μL was added to 22mL of R10HIFBS media (20 cells/mL). Three millilitres of

this cell suspension was plated per well in an entire 6 well plate (60 cells/well) to determine

cloning efficiency. Plates were incubated for 10 days to allow colony formation and scored

using a light box. The remaining treated cells were maintained in exponential growth for a

further 6 days to allow phenotypic expression of HPRT mutants.

2.4.6. Determining the HPRT mutant frequency following treatment of HCT116 cells

HCT116 cells were washed with PBS, detached using trypsin and counted using a

haemocytometer (section 2.2.1). The volume for 1.32x106 viable cells was added up to

99mL of R10HIFBS media. From this, 18μL was added to 12mL of R10HIFBS media

(20 cells/mL). Three millilitres of this cell suspension was plated per well, in 3 wells of a 6

well plate (60cells per well) to determine plating efficiency. HPRT mutants were screened

by adding 0.99mL of 500µg/mL 6-TG to cells diluted in 99mL media. From this, 3mL of

Page 82: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

82

cells were plated per well in five 6 well plates (40,000 cells/well). Plates were incubated for

10 days to allow colony formation and scored using a light box under aseptic conditions.

(Mutant frequency (x10-6) was calculated as the total number of 6-TG resistant clones

divided by the total number of cells plated multiplied by 106 and corrected for plating

efficiency).

2.5. Polymerase Chain Reaction (PCR)

2.5.1. First strand cDNA synthesis from RNA extracts

First strand complementary DNA (cDNA) was synthesised from RNA extracts using the

superscript VILO cDNA synthesis kit (Invitrogen, Paisley) according to manufacturer’s

protocol. In brief, each cDNA synthesis reaction contained 2μL of superscript enzyme mix,

RNA (<2.5μg) and 4μL of 5x VILO mix composed of random hexamer primers, MgCl2 and

dNTP in a total reaction volume of 20μL. Reactions were performed using a Peltier thermal

cycler (PTC-200, MJ Research) with a heated lid to prevent sample evaporation;

conditions for cDNA synthesis were as follows: 10mins at room temperature, 60 mins at

42°C and termination at 85°C for 5 mins. cDNA samples were then stored at -20°C until

analysis. As a negative control, RNA was omitted in a reverse transcription reaction and

replaced with water to evaluate the possibility of RNA introduction during experimentation.

Additionally, to eliminate the possibility of genomic DNA amplification in ensuing

experiments, superscript enzyme was omitted from a cDNA synthesis reaction containing

RNA.

2.5.2. PCR primer design

Unless otherwise stated, primer sequences were designed using NCBI-primer BLAST

design tool (http://www.ncbi.nlm.nih.gov/tools/primer-blast/index.cgi?LINK_LOC=

BlastHome). Primers were selected to be approximately 20-25 nucleotides in length. The

Page 83: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

83

melting temperature (TM) of primers was calculated using the following formula: 4(G+C) +

2(A+T). The difference in annealing temperature (TA) between primer pairs was no greater

than 4°C. The primers designed using NCBI-primer BLAST were subject to further analysis

using Integrated DNA Technologies oligonucleotide analyser

(http://eu.idtdna.com/analyzer/applications/oligoanalyzer/). Primer sequences were

assessed for the capability to form intramolecular hairpins, self-dimers and hetero-dimers.

Those which failed to do so were used in subsequent PCR reactions. PCR primers were

synthesised by Invitrogen (Paisley, UK). The primer sequences used for PCR amplification

are listed in Table 2.4.

2.5.3. PCR amplification of genomic DNA and cDNA

All PCR reagents were obtained from Invitrogen (Paisley, UK). PCR was performed using

a Peltier thermal cycler (PTC-200, MJ Research) using a heated lid. In a final reaction

volume of 50μL, a typical reaction mixture contained at least 300ng of gDNA or 1-2µL of

cDNA, 200nM of each forward and reverse primer (Table 2.4), 200μM dNTPs, 5 units of Tfi

polymerase and 1x Tfi PCR buffer supplemented with 2mM MgCl2.

Unless otherwise stated, standard PCR amplification conditions were as follows:

initial denaturation step at 94°C for 4 mins, followed by 30 cycles of denaturing at 94°C for

30 secs, primer annealing at 64°C for 25secs and polymerisation at 72°C for 30secs. The

final polymerisation step was extended to 7 mins and samples stored at 4°C until analysis.

PCR conditions for each primer pair can be found in Table 2.4.

2.5.4. Agarose gel electrophoresis

All PCR products were resolved using agarose gel electrophoresis. Aliquots of PCR

products (20μL) were mixed with 4μL of 6x gel loading buffer (0.25%w/v bromophenol blue

and 40% w/v sucrose) and typically electrophoresed through a 1.2%w/v agarose gel

Page 84: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

84

containing 0.5μg/mL ethidium bromide. Electrophoresis was carried out for 60-90 mins at

100V in 1x TBE buffer (89 mM Tris, pH 8.3, 89mM boric acid and 2 mM EDTA). Gels were

visualised under UV using BioRad ChemiDoc XRS+ with Image Lab software.

2.5.5. Restriction enzyme digestion of PCR products

PCR products from HPRT amplification (primer pairs 2 and 3; Table 2.4) were subject to

Mbo I restriction enzyme digestion to inform the integrity of the 5’-GATC-3’ restriction

enzyme recognition sequence. A wild-type 5’-GATC-3’ sequence results in cleavage of

PCR product into two fragments; 137 and 40bp for 177bp PCR product (primer pair 2) or

474bp and 544bp for 1018bp PCR product (primer pair 3). Mutation within the 5’-GATC-3’

sequence results in resistance to Mbo I digestion. Typically, 5-15μL of PCR product were

restriction enzyme digested with 1 unit of Mbo I for 1h-2h at 37°C. A typical reaction

mixture contained PCR product, 1x enzyme reaction buffer K and 1 unit of Mbo I enzyme

in a 20μL volume. Digested PCR products were resolved on 1.2%w/v agarose gels and

visualised as described in section 2.5.4.

2.5.6. PCR product purification and DNA sequencing

PCR products were extracted from agarose gels and purified using the QIAquick gel

extraction kit (Qiagen, Crawley) according to manufacturer’s protocol. Gel solubilisation

buffer QG, wash buffer PE and elution buffer EB were included in the manufacturer’s kit. In

brief, the DNA fragment(s) were excised from the agarose gel and weighed. Up to 400mg

of the gel fragment containing the PCR product was placed in a DNase/RNase free

eppendorf tube overlaid with three volumes of gel solubilisation buffer QG i.e. 1.2mL of

buffer QG per 0.4g gel piece. The samples were incubated at 50°C for at least 10 mins

with occasional mixing until the agarose gel slice had dissolved. For optimal recovery, one

gel volume of isopropanol was added to the dissolved gel slice and mixed. The solution

Page 85: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

85

was then added to the extraction column within a wash tube and centrifuged at 18,000xg

for 1 minute. The filtrate was discarded and 500μL of gel solubilisation buffer QG added to

the column and centrifuged at 18,000xg for 1 minute. The flow through was discarded and

0.75mL of wash buffer PE added to the column and incubated for 5 mins at room

temperature. The column was centrifuged at 18,000xg for 1 minute and the filtrate

discarded. The spin column was placed in a fresh wash tube and centrifuged again at

18,000xg for 1 minute to remove residual PE wash buffer. The spin column was then

transferred to a clean eppendorf tube (DNase/RNase free) with the addition of 20-50μL of

elution buffer EB. Columns were incubated for 1 minute at room temperature and

centrifuged at 18,000xg for 1 minute. Eluant containing the purified PCR product was

stored at -20°C until the time of analysis.

For DNA sequencing, purified PCR products were quantitated using a quantitative

agarose gel (section 2.5.4). An aliquot of purified PCR product (5μL) was mixed with 1μL

of 6x gel loading buffer and electrophoresed simultaneously with a quantitative DNA ladder

(Fisher Scientific, Leicester) through a 1.2%w/v agarose gel in 1x TBE. Samples were

diluted to 10ng/μL in a 10μL volume to be sent for sequencing. In separate PCR tubes,

each primer used for PCR amplification was diluted to 3.2μM in a 10μL volume per sample

to be sequenced. Diluted DNA and primers were sent for DNA sequencing by Source

Bioscience ltd (University College London, UK).

DNA sequence data was visualised using the Applied Biosystems Sequence

Scanner version 1.0 (Applied Biosystems, UK). Sequence data was compared to the wild-

type sequence using the European Bioinformatics Institute (EBI) ClustalW2 multiple

sequence alignment tool. Individual sequences were manually screened to confirm

mutation using NCBI-BLAST.

Page 86: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

86

2.6. Immunoblot

All antibodies were obtained from Abcam (Cambridge, UK), unless specified otherwise.

Chemiluminescent ladder was obtained from Cell Signalling Technology (Hertfordshire,

UK). Tracking ladder was obtained from Invitrogen (Paisley, UK).

Protein from cell pellets was extracted and quantitated as described in section

2.2.5. Protein expression was assessed using a denaturing immunoblot.

2.6.1. Preparation and electrophoresis of samples

Typically, between 20-40μg of protein were loaded per well depending on the protein

under investigation. For example, protein extracts (10μL) were diluted with water (up to

16μL) and mixed with 4μL (5x) sodium dodecyl sulphate (SDS) loading buffer (66mM Tris

pH6.8, 26%w/v glycerol, 2.1%w/v SDS, 0.01%w/v bromophenol blue and 5%v/v β-

mercaptoethanol (only added on the day of use)). Samples were then heated at 95°C for 5

mins to denature native protein. Meanwhile, 3μL of chemiluminescent ladder was heated

to 95°C for 2 mins. Following heating, samples were briefly centrifuged and loaded on to a

reducing SDS polyacrylamide gel. To ensure resolution of the intended protein, a pre-

stained protein tracking ladder was used.

Gels were cast such that 1/3 of the top of the gel was a 5%w/v stacking gel (5%w/v

acrylamide, clean water, 0.07%w/v ammonium persulphate (made fresh) and 1x SDS

stacking gel buffer composed of 0.125M Tris pH6.8 and 0.1%w/v SDS) whilst the lower 2/3

of the gel was a 10%w/v resolving gel (10%w/v acrylamide, water, 0.07%w/v ammonium

persulphate (made fresh) and 1x SDS resolving gel buffer composed of 0.6M Tris pH8.8

and 0.16%w/v SDS).

Samples were electrophoresed at 60V for ~30 mins in 1x SDS running buffer

(2.5mM Tris pH8.3, 25mM glycine, 0.01%w/v SDS) through the stacking gel and then 100V

through the resolving gel. Typical length of electrophoresis ranged from 90-120mins.

Page 87: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

87

2.6.2. Transfer of protein

Protein within SDS polyacrylamide gel was transferred to a polyvinylidine fluoride (PVDF)

membrane for immunoblotting as follows. To make the transfer sandwich, two sponges

were soaked in freshly prepared 1x transfer buffer (25mM Tris, 186mM glycine and 20%

methanol). Two filter cards (cut to the size of the PVDF membrane) were overlaid on to a

sponge followed by the SDS polyacrylamide gel. The PVDF membrane was briefly soaked

in water then placed over the gel in the 1x transfer buffer taking care not to introduce

bubbles. A further two filter cards were placed on top of the PVDF membrane followed by

a pre-soaked sponge. The transfer sandwich was in the following order:

Sponge - 2x filter card - SDS gel – PVDF membrane – 2x filter card – sponge

The transfer of the protein occurs from the gel on to the PVDF membrane to facilitate

immunoblotting. Thus, the SDS gel should be closest to the negative electrode whilst the

PVDF membrane to the positive. Transfer was allowed to occur in 1x transfer buffer at

150V, 400mA at 4°C for 75 mins.

To confirm transfer of protein on to the PVDF membrane, a Ponceau S stain was

employed. In brief, membranes are removed from the transfer apparatus and stained with

20mL of Ponceau S stain for 5 mins. Images were visualised on the Kodak IS4000MM.

The blots were destained by three 5 minute washes in PBS-T wash buffer (1x phosphate

buffered saline and 0.1%v/v Tween-20).

2.6.3. Immunoblotting

Following successful transfer of protein on to PVDF membrane, immunoblotting was used

to determine protein expression.

Page 88: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

88

Membranes were blocked for 1h in 20mL blocking buffer (PBS-T plus 5%w/v semi-

skimmed milk powder). Blots were then stained overnight at 4°C with primary antibody

diluted in blocking buffer to the desired concentration (see Table 2.6 for individual antibody

dilutions). Following this, membranes were washed for 5 mins three times in 25mL PBS-T

wash buffer. Membranes were then stained with horse radish peroxidise (HRP) conjugated

secondary antibody for 1h at room temperature. The secondary antibody used was

(1:2000) goat anti-mouse HRP conjugated, unless otherwise stated (see Table 2.6 for

individual antibody targets). Membranes were then washed for 5 mins three times in 25mL

PBS-T wash buffer.

Blots were visualised using 4mL of the west pico chemiluminescent substrate

(Thermo Scientific, Surrey UK) with a 5 minute incubation at room temperature on the

Kodak IS4000MM.

As a loading control and for normalisation of target protein expression, either

β-ACTIN or glyceraldehyde 3-phosphate dehydrogenase (GAPDH) protein expression was

assessed. Membranes were washed to remove chemiluminescent substrate three times

for 5 mins in PBS-T wash buffer.

Membranes were blocked in blocking buffer for 3 h then overlaid with a primary

antibody specific to either β-ACTIN or GAPDH. Primary antibody staining for β-ACTIN was

for 1h at room temperature whilst GAPDH staining was overnight at 4°C (see Table 2.6).

Following staining, the primary antibody was removed and membranes washed

three times in PBS-T for 5 mins. Membranes were further stained with secondary antibody

for 1 h at room temperature (See Table 2.6 for details). Blots were washed three times for

5 mins in PBS-T wash buffer and visualised using the west pico chemiluminescent

substrate as described above.

All buffers described above are listed in appendix 1.

Page 89: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

89

1 Primary and secondary antibodies were obtained from Abcam (Cambridge, UK).

2 Primary GAPDH antibody was obtained from Cell Signalling Technology (Hertfordshire, UK).

3 RT: room temperature.

4 All 2° Antibodies are conjugated to HRP.

Table 2.6. Antibodies used in immunoblot analysis.

Protein

Primary (1°) Ab1 1° Dilution Incubation length(h)/

temperature(°C) Secondary (2°) Ab 4 2° dilution

MSH2 Mouse anti-human 1:1,000 Overnight/4°C Goat anti-mouse 1:2,000

ADAMTS3

Rabbit anti-human 1:2,000 Overnight/4°C Donkey anti-rabbit 1:10,000

RAD51 Mouse anti-human 1:2,000 Overnight/4°C Goat anti-mouse 1:2,000

β-ACTIN Mouse anti-human 1:40,000 1h/RT 3 Goat anti-mouse 1:2,000

GAPDH Rabbit anti-human 2 1:1,000 Overnight/4°C Donkey anti-rabbit 1:10,000

Page 90: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

90

Chapter 3

The Genotoxicity of a triplex forming oligonucleotide targeting

genomic DNA

3.1. Introduction

As described in section 1.3.1, purine tracts in duplex DNA can bind third oligonucleotide

strands in a sequence specific manner to form triplex structures (Duca et al., 2008,

Hoogsteen, 1959). TFO designed to bind within engineered reporter constructs have been

reported to be mutagenic (Kalish et al., 2005, Puri et al., 2002, Puri et al., 2001, Vasquez

et al., 2001, Faruqi et al., 2000, Vasquez et al., 1999, Majumdar et al., 1998, Wang,

Seidman & Glazer, 1996).

In particular, a 30 nucleotide TFO was reported to be capable of inducing mutation

in the order of 0.27% (13 fold above the spontaneous mutant frequency) in a SupFG1

based reporter construct (Wang, Seidman & Glazer, 1996). Sequencing mutant SupFG1

clones revealed mainly point mutations and some small deletions and insertions around

the triplex target sequence. The mechanism of mutation was proposed to be a result of a

stalled transcription fork at the site of the triplex generating a re-iterative repair patch

(Hanawalt, 1994). Repeated attempts in “gratuitous” and “error prone” repair, (introduced

by the natural error frequency of DNA-repair polymerases) may have resulted in mutation

around the triplex target sequence (Wang, Seidman & Glazer, 1996). It was further

postulated that multiple cycles of triplex formation, transcription inhibition and stimulation

of repair can occur at a single site increasing the probability of a mutagenic event.

Triplex formation has also been reported to induce HR (Faruqi et al., 2000). A TFO

conjugated to the DNA intercalator psoralen was reported to induce recombination

between two tandem supF genes, each containing an inactivating point mutation, at a

Page 91: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

91

frequency of 0.4%. Triplex induced recombination produced a single functional supF gene

detected by a phenotypic change (White vs. blue colonies). These non-conservative

events are controversial in comparison to the study presented by Luo et al. (Luo et al.,

2000); there, they used a reporter construct composed of two tandem TK genes, flanking

the triplex target sequence, each containing an inactivating insertion mutation. These cells

formed a wild-type TK gene through conservative events at a frequency of 1%, while

maintaining one of the mutant TK loci.

Although ASO appear to be powerful tools for sequence specific manipulation of

gene expression (Grunweller et al., 2003, Pirollo et al., 2003, Kurreck et al., 2002,

Zamaratski, Pradeepkumar & Chattopadhyaya, 2001), the studies described above

present a concern for oligonucleotide based therapies; specifically, they are suggestive

that triplex formation at genomic DNA may be ensued with heritable sequence alterations.

Indeed, the EMA has raised concern as to whether such oligonucleotide based

pharmaceuticals could present with off-target genotoxicity through triplex formation at

genomic DNA (European Medicines Agency, 2004).

Of particular relevance to these highlighted concerns is that previous reports of

triplex mediated mutagenesis have been described for populations of cells containing an

engineered target sequence with (1) TFO covalently linked to a DNA intercalator (psoralen

or acridine) (Macris & Glazer, 2003, Vasquez et al., 2001, Xu, Glazer & Wang, 2000,

Vasquez et al., 1999); (2) by pre-incubating the target vector with TFO under non-

physiological conditions to assess a mutation endpoint (Ye, Guntaka & Mahato, 2007,

Datta et al., 2001, Xu, Glazer & Wang, 2000); (3) using electroporation to deliver

oligonucleotide (Puri et al., 2002, Puri et al., 2001, Faruqi et al., 2000, Majumdar et al.,

1998) and thus do not necessarily reflect biologically relevant events.

Additionally, critical review of the 30 nucleotide SupFG1 target sequence previously

used (Kalish et al., 2005, Datta et al., 2001, Vasquez et al., 2001, Faruqi et al., 2000,

Page 92: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

92

Figure 3.1. DNA quadruplex formation. Guanine rich sequences in duplex DNA can adopt a DNA

quadruplex structure provding there are 4 units of GGG each separated by at least one nucleotide.

Vasquez et al., 1999, Wang, Seidman & Glazer, 1996) has highlighted the particular

abundance of guanine nucleotides (23nt). In a study by Todd (Todd, 2007), a sequence

with a guanine composition of 23/30 nucleotides is almost 90% likely to form a DNA

quadruplex structure (Fig. 3.1). However, a targeting oligonucleotide has never been

reported to interact with a duplex to facilitate or stabilise quadruplex formation.

Considering the limitations of previous studies, the purpose of this study was to

examine the hypothesis that triplex formation within the coding region of genomic DNA is

mutagenic. Furthermore, the study has assessed the potential for DNA quadruplex

formation by the previously employed SupFG1 target sequence.

3.2. Experimental outline

To investigate triplex mediated mutagenesis at genomic DNA, a TFO (TFO27),

twenty seven nucleotides in length, was engineered to target the genomic coding region of

the hemizygous HPRT locus in human lymphoblastoid TK6 cells. Following treatment of

TK6 cells, TFO27 mediated mutagenesis at the targeted HPRT locus was assessed using

the regulatory acceptable HPRT forward mutation assay. Mutation at the HPRT locus can

be detected and quantified through cellular resistance to the toxic effects of 6-TG. The

Page 93: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

93

HPRT forward mutation assay has been previously employed using TK6 cells to assess

the genotoxic potential of small molecule compounds (Carter et al., 2007, Meng et al.,

2002, Zhuang et al., 2000). Additionally, non-target specific genotoxicity caused by the

presence of oligonucleotide could be screened through cellular resistance to TFT

informing the integrity of the TK locus (gene ID 7083).

As a negative control, an oligonucleotide with the same base composition as

TFO27 but in a random order was adopted (SCR27). SCR27 served as the reference for

which the biological activity of TFO27 was compared. The spontaneous mutant frequency

at the genomic HPRT or TK loci was assessed using an untreated population of TK6 cells;

the known mutagen, EMS, was used as a positive genotoxic control.

In the first instance, oligonucleotide target binding affinity was determined using an

EMSA and a DNase I protection assay. Following this, the mutagenicity of oligonucleotides

treating 3x106 and then 10x106 TK6 cells was determined using the HPRT forward

mutation assay. DNA quadruplex formation by the SupFG1 target sequence was also

assessed using an EMSA technique.

3.3. Methods

1. TFO design is described in section 2.1.1.

2. EMSA are described in section 2.1.4 and 2.1.7.

3. DNase I protection assay is described in section 2.1.6.

4. Serum stability of oligonucleotides is described in section 2.1.3.

5. Oligonucleotide transfection efficiencies and mutation assays are described in

sections 2.3 and 2.4.

6. Detecting triplex formation at genomic DNA is described in section 2.1.5.

7. Genomic DNA extraction and PCR amplification/sequencing is described in section

2.2.3 and 2.5.

Page 94: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

94

3.4. Results

3.4.1. High affinity triplex formation

In the first instance, it was important to assess the capability of TFO27 to form a triplex at

its target motif. For this, an EMSA was employed. Duplex DNA encompassing the purine

rich tract in exon 3 of the HPRT locus was incubated with serial dilutions of TFO27 or

SCR27. Following this, samples were electrophoresed through polyacrylamide gel. Bands

of reduced migration relative to duplex are indicative of triplex formation. In triplex binding

buffer, TFO27 bound duplex DNA with high affinity at concentrations as low as 10-10M (Fig.

3.2A). As expected, SCR27 failed to form triplex structures at concentrations as high as

10-5M (Fig. 3.2B). These data indicate sequence specific triplex formation by TFO27 at its

target motif with a Kd in the sub-nanomolar range.

Previous studies examining triplex mediated mutagenesis have incubated target

plasmid DNA with TFO in the presence of spermidine (Ye, Guntaka & Mahato, 2007, Datta

et al., 2001, Xu, Glazer & Wang, 2000). Using similar conditions, triplex binding buffer

containing 2.5mM spermidine, we found both TFO27 and SCR27 to be capable of

producing bands of reduced migration (Fig. 3.2, C and D).

Additionally, the ability of TFO27 to protect the triplex target sequence in a 50mer

duplex from DNase I mediated degradation was assessed (Fig. 3.3). TFO27 appeared to

protect duplex DNA from digestion in a dose dependent manner. SCR27 also appeared to

partially protect duplex DNA from digestion. This effect is likely to be due to the presence

of spermidine in the reaction buffer, in support of our initial observation (Fig. 3.2D).

Page 95: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

95

Figure 3.3. Investigating the capability of oligonucleotides to protect duplex DNA from DNase I cleavage. A synthetic 50mer duplex containing the triplex target sequence in exon 3 of the HPRT locus was annealed using oligonucleotides HPRT-S-50 and HPRT-AS-50 (Table 2.3). Duplex DNA was incubated without oligonucleotide (lane 1), with 5μM SCR27 (lane 2), 2μM TFO27 (lane 3) or 5μM TFO27 (lane 4) in triplex binding buffer supplemented with 2.5mM spermidine. Samples were then digested with DNase I and electrophoresed. TFO27 appeared to protect duplex DNA from DNase I digestion in a dose dependent manner. SCR27 also appeared to partially protect duplex DNA from DNase I digestion. This is likely to be caused by the presence of spermidine in the reaction buffer.

Figure 3.2. Investigating the triplex forming capability of TFO27 and SCR27. A synthetic 35mer duplex containing the triplex target sequence in exon 3 of the HPRT locus was annealed using oligonucleotides HPRT-S-35 and HPRT-AS-35 (Table 2.3) and incubated with serial dilutions of oligonucleotide in triplex binding buffer (A and B) or triplex binding buffer containing 2.5mM spermidine (C and D). Samples were then electrophoresed through polyacrylamide gel. In triplex binding buffer, bands of reduced migration are observed with TFO27 at nanomolar concentrations representing triplex formation (A) whereas SCR27 fails to form such structures (B). High affinity triplex formation is also evident with TFO27 (C) in the presence of spermidine. However, SCR27 also appears capable of associating to duplex DNA in a sequence independent manner in the presence of spermidine (D). Bands ‘T’ and ‘D’ represent the triplex and duplex structures, respectively.

A B

D

C

Page 96: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

96

3.4.2. Transfection efficiency

Due to the scale of transfection and concentration of oligonucleotide required for ensuing

mutagenicity studies, the manufacturer’s protocol for small interfereing RNA (siRNA)

transfection was not appropriate. Thus, the transfection efficiency of human TK6 cells was

investigated, in a 96 well format, using 6-carboxyfluorescein (6FAM) labelled TFO27

(6FAM-TFO27) with siPORT NeoFX (Applied Biosystems) or NTER (Sigma Aldrich) as the

transfection facilitators. Transfection of TK6 cells was allowed to occur at 37°C for 4 h.

Following fluorescence measurements, the viable cell population in each well was

determined using the trypan blue exclusion assay. Arbitrary fluorescence units were

normalised to cell number following transfection to correct for cell death during treatment.

The following ratios of oligonucleotide (μg) to transfection facilitator (μL) were

evaluated for both siPORT and NTER using 1μM and 2μM 6FAM-TFO27; 2:1, 1:1, 1:2 and

1:4, respectively (Fig. 3.4).

Using 1μM 6FAM-TFO27, 2:1 DNA (μg): transfection facilitator (μL) appeared to be

the most efficient transfection ratio for both siPORT NeoFX and NTER. However, no

significant difference was observed between either transfection facilitator using 1μM

oligonucleotide (Fig. 3.4A). Transfection efficiency between siPORT NeoFX and NTER

appeared to be markedly increased using 2μM 6FAM-TFO27 with an almost 3 fold

difference in transfection efficiency between siPORT and NTER using 2:1 or 1:1 DNA (μg)

to transfection facilitator (μL) (Fig. 3.4B). Furthermore, no difference in transfection

efficiency between 2:1 or 1:1 DNA (μg) to siPORT NeoFX (μL) was observed using either

1μM or 2μM 6FAM-TFO27.

Based on these results, siPORT NeoFX was selected as the transfection facilitator

using a 2:1 DNA (μg) to siPORT (μL) ratio for large scale transfection of TK6 cells in

oligonucleotide mutagenicity studies.

Page 97: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

97

Figure 3.4. Evaluating the transfection efficiency of TK6 cells using siPORT NeoFX and NTER. Human TK6 cells were transfected for 4h with 6FAM-TFO27 using either siPORT NeoFX or NTER using 2:1, 1:1, 1:2 or 1:4 DNA (μg) to transfection facilitator (μl) ratios. siPORT NeoFX appeared to be more efficient than NTER in delivery of 1μM (A) and 2μM (B) 6FAM-TFO27 using a 2:1 or 1:1 DNA (μg) to transfection facilitator (μl) ratio. Data represent mean ± standard deviation (SD) of three independent wells of a 96 well plate.

A

B

1µM 6FAM-TFO27

2µM 6FAM-TFO27

Page 98: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

98

3.4.3. Evaluating the genotoxic potential of a TFO at genomic DNA

To evaluate the genotoxicity of TFO27 at the genomic HPRT locus, the HPRT forward

mutation assay was employed using the human lymphoblastoid TK6 cell line. Off-target

genotoxicity caused by the presence of oligonucleotide was determined by screening the

mutant frequency at the non-targeted TK locus.

TK6 cells were transfected with oligonucleotide using siPORT NeoFX over 4h since

electroporation stress induces an altered biochemical and genetic milieu and such cells

tend to be more susceptible to mutation (Oberly & Garriot, 1996, Seetharam & Seidman,

1992, Trump & Berezesky, 1987). In this study, cytotoxicity following treatment was

assessed by the RTG, which accounts for cell death during treatment and subsequent

effects on cell growth and cloning ability (Clements, 2000, Clive et al., 1995). Cytotoxicity

was maintained within OECD guideline 476 (http://www.oecd.org/dataoecd/

18/32/1948426.pdf) and therefore cell survival was no less than the minimum accepted

20% RTG to avoid biasing the mutation endpoint (Oberly & Garriot, 1996).

In the first instance, 3x106 TK6 cells were transfected with 1μM oligonucleotide in a

single culture experiment (Fig. 3.5). The spontaneous mutant frequency at each locus was

measured using an untreated population of cells (Background). The positive genotoxic

control was 5μg/mL EMS. Cytotoxicity was not observed with oligonucleotide treatment

(Fig. 3.5A). TFO27 appeared to result in a marginal increase in mutant frequency at the

targeted HPRT locus, whereas SCR27 had no such effect (Fig. 3.5B). The TK mutant

frequency was not determined in this initial experiment; thus, the mode of action of TFO27

may not necessarily be locus specific. To reproduce this observation, 3x106 TK6 cells

were treated in single culture with 1μM, 2μM and 5μM oligonucleotide (Fig. 3.6). Cell

survival following treatment decreased with increasing dose (Fig. 3.6A), but the RTG was

above the minimum accepted 20%. In this case, TFO27 appeared to induce sequence

specific and dose dependent HPRT mutants (Fig. 3.6B), whilst no induction of TK mutants

Page 99: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

99

were observed suggesting a locus specific mode of action (Fig. 3.6C). As expected,

SCR27 failed to elicit a genotoxic response.

Page 100: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

100

Background

SCR27

TFO27

SCR27

TFO27

SCR27

TFO27EM

S

0

10

20

30

40

50

60

70

80

Mu

tan

t fr

eq

ue

ncy

x10

-6 v

iab

le c

ells

Background

SCR27

TFO27

SCR27

TFO27

SCR27

TFO27EM

S

0

10

20

30

40

50

60

70

80

90

100

110

Re

lati

ve

To

tal G

row

th (

%)

Background

SCR27

TFO27

SCR27

TFO27

SCR27

TFO27EM

S

0

20

40

60

80

100

120

140

160

Mu

tan

t fr

eq

ue

ncy

x10

-6 v

iab

le c

ells

Background

SCR27

TFO27EM

S

0

10

20

30

40

50

60

70

80

90

100

110R

ela

tiv

e T

ota

l G

row

th (

%)

Background

SCR27

TFO27EM

S

0

10

20

30

40

50

60

70

80

Mu

tan

t fr

eq

ue

ncy

x10

-6 v

iab

le c

ells

1M Oligo

2M Oligo

5M Oligo

Figure 3.6. Investigating the genotoxic effect of 1μM, 2μM and 5μM oligonucleotide treating 3x106 human TK6 cells. Viability following treatment was above the minimum 20% accepted RTG, although a dose dependent increase in cell death was evident (A). TFO27 appeared to cause a dose dependent increase in HPRT mutants relative to the control oligonucleotide (B) but failed to induce mutation at the non-targeted TK locus (C). As expected, the positive control EMS (5μg/ml) induced mutation at the TK and HPRT loci (B and C). Data represent the value of a single treatment.

Figure 3.5. Investigating the genotoxic effect of 1μM oligonucleotide treating 3x106 human TK6 cells. Cytotoxicity following treatment was not observed (A). TFO27 appeared to produce a marginal increase in HPRT mutants relative to SCR27 (B). As expected, the positive control EMS (5μg/ml) induced mutation at the HPRT locus (B). Data represent the value of a single treatment.

A

B C

A B

Page 101: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

101

Conforming to a regulatory protocol outlined in the OECD guideline for the MLA

(http://www.oecd.org/dataoecd/18/32/1948426.pdf), 10x106 TK6 cells were treated with

oligonucleotide to reproduce the initial observation treating 3x106 cells. In this case, four

independent negative control cultures were adopted to measure the spontaneous mutant

frequency, whereas the remaining treatments were in duplicate cultures. One micromolar

of 4NQO was used as a positive genotoxic control. Treating 10x106 TK6 cells with 1μM,

2μM and 5μM oligonucleotide using a regulatory acceptable mutation assay was not

cytotoxic (Fig 3.7A), but contrary to treating 3x106 cells, TFO27 failed to induce a dose

dependent increase in HPRT mutants (Fig. 3.7B). As expected, 4NQO induced a

significant increase in HPRT mutants.

A more comprehensive dose range from 2μM to 12.2μM was used to explore the

mutagenicity of TFO27 up to maximum acceptable cytotoxicity (Fig. 3.8). Treatment of TK6

cells up to 12.2μM of TFO27 appeared to result in a dose dependent decrease in the RSG;

a measure of cell death and proliferating ability following treatment (Fig. 3.8A). Three

doses of TFO27 (2μM, 5μM and 6.25μM) were selected that resulted in maximum

acceptable cytotoxicity following treatment, estimated using the RSG, and carried forward

to determine the RTG, TK and HPRT mutant frequency. The selected doses of TFO27

were not found to be cytotoxic (~60% RTG, Fig. 3.8B). Additionally, no induction of TK or

HPRT mutants were observed (Fig. 3.8, C and D). As expected, the positive control

significantly increased the mutant frequency at both loci.

Page 102: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

102

Figure 3.8. Investigating the genotoxic effect of TFO27 in a regulatory acceptable mutation assay treating 10x106 TK6 cells. Cytotoxicity following treatment, estimated from the RSG, appeared to be evident from 7.8μM (A). Three doses of TFO27 (2μM, 5μM and 6.25μM) that gave maximum acceptable cytotoxicity were selected to determine the RTG (B), TK mutant frequency (C) and HPRT mutant frequency (D). Cytotoxicity up to 6.25μM was no greater than approximately 40% (A) but no induction of TK (C) or HPRT mutants (D) were observed. As expected the positive control 4NQO (1μM) was genotoxic at the TK and HPRT loci (B and C). Error bar represents mean ± SD of four independent treatments, whereas the remaining data are the mean of two independent treatments.

Background M2

M5M

6.25 4NQO

0

20

40

60

80

100

120

140

160

Mu

tan

t fr

eq

ue

ncy

x10

-6 v

iab

le c

ells

Background M2

M5M

6.25 4NQO

0

10

20

30

40

50

60

70

80

90

100

110

Re

lati

ve

To

tal G

row

th (

%)

C

A B

D

Background

SCR27

TFO27

TFO27

SCR27

SCR27

TFO27

4NQO

0

10

20

30

40

50

60

70

80

90

100

110

Re

lati

ve

To

tal G

row

th (

%)

Background

SCR27

TFO27

TFO27

SCR27

SCR27

TFO27

4NQO

0

10

20

30

40

50

60

70

80

90

100

Mu

tan

t fr

eq

ue

ncy

x10

-6 v

iab

le c

ells

Background M2

M5M

6.25 4NQO

0

5

10

15

20

25

30

35

40

Mu

tan

t fr

eq

ue

ncy

x10

-6 v

iab

le c

ells

1M Oligo

2M Oligo

5M Oligo

Background M2

M5M

6.25

M7.8

M9.8

M12.2 4NQO

0.0

0.1

0.2

0.3

0.4

0.5

0.6

0.7

0.8

0.9

1.0

1.1

Re

lati

ve

su

sp

en

sio

n g

row

th

A B

Figure 3.7. Investigating the genotoxic effect of 1μM. 2μM and 5μM oligonucleotide treating 10x106 human TK6 cells using a regulatory acceptable mutation assay. Cytotoxicity following treatment was not observed (A) and TFO27 failed to induce HPRT mutants (B). As expected the positive control 4NQO (1μM) was genotoxic at the HPRT locus (B). Error bar represents mean ± SD of four independent treatments, whereas the remaining data are the mean of two independent treatments.

Page 103: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

103

A

B C

Figure 3.9. Investigating the genotoxic effect of TFO27 up to 9.8μM in a regulatory acceptable mutation assay treating 10x106 TK6 cells. cytotoxicity following treatment was no greater than 30% (A). Treatment of TK6 cells with oligonucleotide failed to result in an increase in TK (B) or HPRT mutants (C). Error bars represent mean ± SD of three independent cultures.

Background

SCRTFO

SCRTFO

SCRTFO

EMS

0

10

20

30

40

50

60

70

80

90

100

110

120

130

Re

lati

ve

To

tal G

row

th (

%)

Background

SCRTFO

SCRTFO

SCRTFO

EMS

0

5

10

15

20

Mu

tan

t fr

eq

ue

ncy

x10

-6 v

iab

le c

ells

Background

SCRTFO

SCRTFO

SCRTFO

EMS

0

10

20

30

40

50

60

70

80

Mu

tan

t fr

eq

ue

ncy

x10

-6 v

iab

le c

ells

6.25M Oligo

7.8M Oligo

9.8M Oligo

6.25M Oligo

7.8M Oligo

9.8M Oligo

In a definitive study to explore the mutagenicity of TFO27 at genomic DNA, 10x106

TK6 cells were treated in triplicate cultures up to 9.8μM (Fig. 3.9). The RTG following

treatment was above the minimum accepted 20% (Fig 3.9A). TFO27 failed to induce

mutation in human TK6 cells at both the TK (Fig. 3.9B) and HPRT loci (Fig. 3.9C) up to

9.8μM. The positive control (EMS) induced mutation at both loci suggesting a viable

experiment.

Page 104: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

104

3.4.4. Serum stability of oligonucleotides

As described above, TFO27 targeting the genomic HPRT locus in human TK6 cells was

found to lack reproducible genotoxic activity up to 9.8μM when treating up to10x106 cells.

A potential cause for this may be rapid nuclease mediated degradation of oligonucleotides

during treatment. Oligonucleotides used in mutagenicity studies were modified with four

terminal phosphorothioate linkages at either end to offer nuclease resistance (Kenner et

al., 2002). To validate this, a serum stability assay was employed. Additionally, we

assessed the serum stability of TFO27 with canonical phosphodiester linkages (PO-

TFO27).

Oligonucleotides were incubated at 37°C in the presence of cell culture media,

which contained 10%v/v horse serum. Aliquots were removed at select time points up to

24h and electrophoresed through a 1.5% agarose gel. Bands were quantified by

densitometry and made relative to the zero hour band. TFO27 and SCR27 were resistant

to degradation in cell culture media for at least 24h (Fig. 3.10). In contrast, PO-TFO27 was

found to be significantly degraded in cell culture media within 3h (Fig. 3.10). Thus, four

terminal phosphorothioate linkages provided nuclease resistance for TFO27 and SCR27.

As degradation of oligonucleotides is likely to occur 3’ to 5’ by serum 3’

exonucleases (Shaw et al., 1991, Tidd & Warenius, 1989, Wickstrom, 1986) the agarose

gel is unlikely to resolve between n-1 or n-2 oligonucleotides whereas polyacrylamide gel

electrophoresis may have been more appropriate in discerning full length and truncated

products. However, TFO that have lost 1 or 2 terminal nucleotides still have the potential

for triplex formation so the method used here accounts for these truncated

oligonucleotides also, as well as the full length product.

Page 105: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

105

Figure 3.10. The serum stability of oligonucleotides. Oligonucleotides, were incubated at 37°C with cell culture media containing 10%v/v horse serum. Aliquots were removed at select time points up to 24h. Samples were electrophoresed through a 1.5% agarose gel. Bands were analysed by densitometry. Intensities are relative to the zero hour. TFO27 and SCR27 with 4 terminal phosphorothioate linkages at either end appeared to be resistant to nuclease degradation up to 24h (PS-TFO and PS-SCR). However, TFO27 containing canonical phosphodiester linkages was rapidly degraded within 3h (PO-TFO). Data represent mean ± SD of three independent samples.

3.4.5. Human TK6 cells contain the wild-type HPRT sequence

An alternative explanation for the lack of genotoxicity by TFO27 may be mutation of the

triplex target sequence in exon 3 of the HPRT locus in human TK6 cells. If the wild-type

sequence was lost, this would present with reduced target binding affinity of TFO27 with

subsequent effects on the potential to block a transcription fork and cause mutation.

To address this, a 177bp stretch encompassing the TFO27 target sequence in exon

3 of the HPRT locus was PCR amplified using genomic DNA from human TK6 cells.

Products were resolved on agarose gels and extracted for DNA sequencing. Sequencing

confirmed that the TK6 cell line used in mutagenicity studies contained the proposed

TFO27 target sequence and therefore the potential for triplex formation (Fig. 3.11, A and

B).

Page 106: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

106

Figure 3.11. Validating the putative TFO27 target sequence in exon 3 of the HPRT locus in human lymphoblastoid TK6 cells. Genomic DNA from TK6 cells was extracted and used for PCR amplification around the TFO27 target sequence. PCR products were resolved on agarose gels. The corresponding band was excised, purified and sequenced. The raw sequence data (A) was extrapolated to NCBI-BLAST (B) which revealed the wild-type sequence of the complementary strand to the TFO27 target sequence in exon 3, highlighted with the red box.

B

A

Page 107: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

107

3.4.6. Method development to detect triplex formation at genomic DNA

To investigate the potential for TFO27 to bind to its target sequence in genomic DNA, a

method was developed to detect triplex formation, a procedure adapted from Besch et al.

(Besch et al., 2004).

An overview of the procedure is outlined in Fig. 2.1 in section 2.1.5. In brief,

genomic DNA extracted from TK6 cells was mixed with TFO27 or SCR27. However,

oligonucleotides were conjugated 5’ to psoralen and 3’ to biotin (sequences listed in Table

2.1). Incubation of Pso-TFO27-biot or Pso-SCR27-biot with genomic DNA in triplex binding

buffer allows triplex formation and intercalation of psoralen into genomic DNA (Fig. 2.1

step A). Once TFO27 forms a triplex at the HPRT locus, UVA (365nm) irradiation of

samples (5J/cm2) covalently reacts the psoralen linked oligonucleotide to genomic DNA at

5’-AT-3’ upstream of the TFO27 target sequence (Ramaswamy & Yeung, 1994, Song &

Tapley, 1979).

Once Pso-TFO27-biot was covalently bound to DNA, samples were restriction

enzyme digested overnight using Bgl II (Fig. 2.1, step B). This produces a HPRT fragment

of ~3.4kb encompassing the covalently bound Pso-TFO27-biot.

To prevent competitive binding of unbound Pso-TFO27-biot with the biotinylated

3.4kb HPRT fragment containing triplex DNA, unbound oligonucleotide was removed

either using electrophoresis through an agarose gel followed by extraction or passing

samples through a Qiagen DNA column (Fig. 2.1 step C).

Following purification, 3.4kb HPRT fragments containing covalently bound

oligonucleotide were separated by their biotin moiety using streptavidin coated

paramagnetic beads (Fig. 2.1, step C). After several washes to remove unbound DNA,

beads bound to triplexed HPRT fragments via the biotin moiety were subject to PCR

amplification across intron 3 of the HPRT locus (downstream of TFO27 target sequence)

to yield a 364bp product using primer pair 1, Table 2.4 (Fig. 2.1, step D).

Page 108: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

108

Figure 3.12. Optimising PCR conditions for triplex detection. In brief, 2µg of Bgl II digested genomic DNA from TK6 cells was used for PCR amplification of a 364bp stretch in intron 3 of the HPRT locus. In the first instance, the primer annealing temperature and MgCl2 concentration was optimised (A). The values above each row in part A represent the concentration (mM) of MgCl2 used in each PCR reaction. Lane 0 represents the water control. Optimum PCR was found to be with 1mM MgCl2 and a 66°C primer annealing temperature. Following this, the lowest PCR detection limit was determined from 60,000 template copies to 10 (B). Lane C in part B represents a water control.

B

A

In the first instance, Bgl II digested genomic DNA extracted from TK6 cells was

used to optimise conditions for PCR amplification of intron 3 of the HPRT locus to detect

triplex formation (Fig. 3.12A). The concentration of MgCl2 (1mM, 2mM and 3mM) was

optimised at primer TA of 66°C, 67°C and 68°C. Under these conditions, 1mM MgCl2 and

Ta 66°C were found to be best resulting in a distinct band around 364bp.

Secondly, it was important to assess the minimum copy number of template the

PCR could detect using these optimised parameters. For this, Bgl II digested TK6 genomic

DNA was diluted from 60,000 copies to 10 and PCR amplified (Fig. 3.12B). The PCR

method was able to detect a minimum of 500 copies of template.

Following optimisation, 16μg of genomic DNA from TK6 cells was incubated with

20μM Pso-TFO27-biot or Pso-SCR27-biot for 3h at room temperature, UVA irradiated and

Bgl II restriction enzyme digested overnight. As a control, genomic DNA was incubated

with water to replace oligonucleotide. Samples were purified through an agarose gel and

Page 109: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

109

A

Figure 3.13. Detecting triplex formation and optimising streptavidin bead concentration. Briefly, 16µg genomic DNA extracted from TK6 cells was incubated without or with 20µM Pso-SCR27-biot (S) or Pso-TFO27-biot (T) at room temperature for 3h in triplex binding buffer (A). Samples were gel purified and mixed with 50µg of streptavidin beads, washed and PCR amplified. No PCR signal is observed with any mixture including the positive genomic DNA control (A). Following this, the influence of streptavidin coated paramagnetic beads on PCR efficiency was evaluated (B). Each PCR reaction contained 16µg genomic DNA mixed with streptavidin beads from 0.5µg up to 5µg. Clearly, above 0.5µg of beads, the PCR reaction is inhibited significantly.

B

isolated using 50μg of streptavidin coated beads. Here, no PCR signal was observed in

any sample including the genomic DNA control (Fig. 3.13A).

To investigate the influence of streptavidin beads, 16μg of Bgl II digested genomic

DNA was mixed with serial dilutions of beads from 50μg down to 10μg and PCR amplified.

No signal was obtained using these bead concentrations (data not shown). Repeating this

experiment with fewer streptavidin beads (5μg down to 0.5μg) for PCR amplification of

genomic DNA showed the presence of beads drastically inhibited the efficiency of PCR

amplification (Fig. 3.13B). Streptavidin beads at 0.5μg was the only concentration not to

effect PCR amplification of 16μg genomic DNA. Thus, 0.5μg of streptavidin beads was

used in subsequent experiments.

TK6 genomic DNA (16μg) incubated with 20μM oligonucleotide, UVA irradiated, Bgl

II digested was purified through an agarose gel as described above. Here, genomic DNA

Page 110: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

110

Figure 3.14. Optimising genomic DNA purification. Briefly, 16µg genomic DNA extracted from TK6 cells was incubated without or with 20µM Pso-SCR27-biot (S) or Pso-TFO27-biot (T) at room temperature for 3h in triplex binding buffer, purified through agarose gel, then mixed with 0.5µg streptavidin beads for PCR amplification (A). All samples were gel purified except sample PCR gDNA which served as a PCR control. No signal detected in any purified sample. Comparing the control gDNA and PCR gDNA suggests a problem with the purification technique as no signal was obtained following purification of DNA (A). Additionally, the above procedure was repeated using a Qiagen DNA extraction column to purify samples (B). No signal was obtained from any sample including the positive control.

A

B

without oligonucleotide was also gel extracted (control gDNA), but additionally, genomic

DNA without oligonucleotide and purification (PCR gDNA) was used as a PCR control

(Fig. 3.14A). Most peculiar was that genomic DNA purified using agarose gel produced no

distinct PCR signal except a faint smear >2kb whereas the PCR control worked. This

suggested a problem with the purification technique and so the above experiment was

repeated except a Qiagen DNA extraction column was employed to purify DNA (Fig.

3.14B). In this case also, purified genomic DNA control failed to produce a distinct 364bp

band except a smear >2kb.

Page 111: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

111

Figure 3.15. Optimising purification of genomic DNA using the Qiagen DNA extraction column. Briefly, 16µg genomic DNA extracted from TK6 cells was digested by Bgl II then used to optimise agarose gel purification. Extraction 1 included the addition of 20µL of ethanol (>99.6%) to a 20µL sample before addition to the spin column. Extraction 2 included addition of 20µL sample directly on to the spin column. Extraction 3 included addition of 100µL ethanol (>99.6%) and 100µL lysis buffer AL to 20µL sample before addition to the spin column. The remaining procedure is the same for all samples as described in section 2.2.3. Following elution of samples in a 50µL, 20µL was used for PCR amplification with or without 0.5µg streptavidin beads. No distinct band is apparent in any sample apart from a smear >2kb in length most intense with extraction 3. Also, no difference is observed between signals with/without beads.

In a further attempt, DNA purification using the Qiagen spin column was investigated.

Three different protocols were used for purification of genomic DNA. Extraction 1 involved

the addition of 20μL of ethanol (>99.6%) before addition to the spin column, extraction 2

was the direct addition of 20μL sample to spin column and extraction 3 involved the

addition of 100μL ethanol (>99.6%) and 100μL of lysis buffer AL before addition to the spin

column. The remaining procedure is as outlined in section 2.2.3. Following elution of DNA,

samples were divided for PCR amplification with or without the addition of 0.5μg

streptavidin beads (Fig. 3.15). In this case also, no distinct bands around 364bp were

observed except a smear >2kb. The most intense smear was using extraction 3, and no

difference was noted with the addition of the streptavidin beads.

Further work is required to determine why a smear is present instead of a distinct

band and to optimise the purification technique either using the gel or the Qiagen spin

column.

Page 112: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

112

3.4.7. The previously employed SupFG1 triplex target sequence is capable of DNA

quadruplex formation

Previous reports of triplex mediated mutagenesis have focused on targeting extra-

chromosomal DNA mainly using the SupFG1 reporter (Kalish et al., 2005, Datta et al.,

2001, Vasquez et al., 2001, Faruqi et al., 2000, Vasquez et al., 1999, Wang, Seidman &

Glazer, 1996). In this system, a 30 bp purine stretch composed primarily of guanine (23nt)

is targeted using a purine motif TFO. On the basis that guanine rich sequences have been

implicated in DNA quadruplex formation (Todd, 2007), the ability of the SupFG1 triplex

target sequence to adopt a DNA quadruplex structure was investigated using an EMSA. In

this case, a band of reduced migration relative to the triplex structure is indicative of DNA

quadruplex formation.

In triplex binding buffer, the SupFG1 target sequence was capable of DNA triplex

and quadruplex formation in the presence of the targeting oligonucleotide AG30 (Fig

3.16A, Lane 5). Densitometric quantitation of lane 5 (Fig. 3.16A) revealed primarily triplex

formation (79%) as opposed to quadruplex formation (18%). As a control, the same

sequence as AG30 and the SupFG1 duplex was employed, except that 4 guanine

nucleotides were replaced with adenine to reduce the potential for quadruplex formation

(AG30-4MM and SupF-4MM, Table 2.3). In triplex binding buffer (Fig. 3.16A), replacing

four guanine nucleotides was found to partially attenuate quadruplex formation (6.5%,

Lane 2). Repeating the experiments using a quadruplex buffer containing potassium (Fig

3.16B), where potassium has been reported to stabilise quadruplex structures (Sun et al.,

2005), quadruplex formation by AG30 and the wild-type SupFG1 target sequence was

almost as prevalent as triplex formation (17% versus 27%, respectively; Lane 5). This

observation was confirmed in an independent experiment (Fig. 3.17). In the quadruplex

buffer (Fig. 3.16B), the level of quadruplex formation by AG30-4MM and SupF-4MM was

about 50% (Lane 2) of the wild-type SupFG1 sequence (Lane 2; 9.5% versus Lane 5;

Page 113: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

113

Figure 3.16. Investigating the quadruplex forming ability of the SupFG1 system. Oligonucleotide sequences are listed in Table 2.3. Duplex DNA containing the SupFG1 sequence was incubated with targeting oligonucleotide in triplex binding buffer (A) or quadruplex buffer (B). As a control to the wild-type SupFG1 sequence, 4 guanine residues were replaced in the target duplex and targeting oligonucleotide (SupF-4MM and AG30-4MM, respectively). In figures A and B, lanes 1-3 are 0.2μM of the SupF-4MM duplex incubated with 10-6M, 10-5M or zero AG30-4MM, respectively. Lanes 4-6 are 0.2μM of the wild-type SupFG1 duplex incubated with zero, 10-5M or 10-6M AG30, respectively. Quadruplex formation is observed with the wild-type SupFG1 duplex incubated with AG30, to a lesser extent in triplex binding buffer (Lane 5, A) than quadruplex buffer (Lane 5, B). SupF-4MM and AG30-4MM is less efficient in quadruplex formation in either buffer relative to the wild-type sequence (Lane 2 vs. Lane 5, A and B). Bands ‘Q’, ‘T’ and ‘D’ represent the quadruplex, triplex and duplex structures, respectively.

B

A

17%). It may be possible that AG30 interacts with the guanine based target sequence

within the SupFG1 duplex such that it complexes with or stabilises/ facilitates quadruplex

formation. Replacing four guanine residues to interrupt the contiguous run of guanines

within AG30 and the SupFG1 duplex was found to partially attenuate quadruplex

formation.

Page 114: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

114

Figure 3.17. Confirmation of DNA quadruplex formation in the SupFG1 duplex. Duplex DNA containing the SupFG1 sequence was incubated with targeting oligonucleotide (AG30) in quadruplex buffer. Lanes 1-4 are 0.2μM of the wild-type SupF duplex incubated with zero, 10-5M, 10-6M, 10-7M AG30, respectively. Quadruplex formation is observed with the wild-type SupFG1 duplex incubated with 10-5M AG30 (Lane 2), although less prevalent to triplex formation (16% vs. 31%, respectively). Bands ‘Q’, ‘T’ and ‘D’ represent the quadruplex, triplex and duplex structures, respectively.

Page 115: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

115

3.5. Discussion

The potential for oligonucleotides to interact with duplex DNA, in a sequence specific

manner, with ensuing mutation has raised concern in regards to oligonucleotide therapy

(European Medicines Agency, 2004). Oligonucleotides have previously been reported to

induce mutation through recognition and binding of purine rich sequences within

engineered reporter constructs to form triplex structures which block progression of

elongating transcription forks (Datta et al., 2001, Vasquez et al., 2001, Faruqi et al., 2000,

Vasquez et al., 1999, Wang, Seidman & Glazer, 1996). Here, the potential for

oligonucleotide mediated mutagenesis via triplex formation at genomic DNA was

addressed.

In contrast to previous reports using reporter systems, this study suggests that a

non-conjugated TFO fails to induce mutation at genomic DNA in human TK6 cells in a

regulatory acceptable mutation assay conducted to OECD guidelines (OECD guideline

476; http://www.oecd.org/dataoecd/18/32/1948426.pdf). TK6 cells were treated with a

dose range of oligonucleotide that spans the concentrations employed in previous studies

examining triplex mediated mutagenesis using reporter constructs (Puri et al., 2002, Puri

et al., 2001, Vasquez et al., 2001, Faruqi et al., 2000, Vasquez et al., 1999, Wang,

Seidman & Glazer, 1996). Despite the lack of genotoxicity by TFO27, it was capable of

binding to its target motif with high affinity and was not a substrate for rapid nuclease

degradation. Additionally, the TK6 cells contained the putative HPRT target sequence

highlighting the potential for triplex formation although it was not that TFO27 could directly

interact with its target sequence.

To the best of our knowledge, this is the first study to report cytotoxicity coupled to a

mutation endpoint for an oligonucleotide. It is crucial to assess the state of cells following

treatment, as stressed cells are more susceptible to mutation thereby biasing an

experimental endpoint (Oberly & Garriot, 1996, Seetharam & Seidman, 1992, Trump &

Page 116: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

116

Berezesky, 1987). For this reason, oligonucleotides were delivered using a transfection

facilitator rather than electroporation which has been regularly employed when assessing

triplex mediated mutagenesis (Puri et al., 2002, Puri et al., 2001, Faruqi et al., 2000,

Majumdar et al., 1998). However, as the arbitrary fluorescence units are reported here per

104 viable cells, and not the percentage of cells that are fluorescent, it is difficult to

determine the variation of transfection efficiency amongst the population of treated cells.

For this reason, confocal microscopy may have been more appropriate to quantitatively

assess the proportion of cells that have been successfully transfected and to what extent.

One may speculate that the transfection procedure in this study may not have been

adequate for TFO27 to overcome the hurdles of the intranuclear milieu for triplex formation

and subsequent mutation. However, in chapter 4, it is shown that an oligonucleotide that

binds to genomic DNA in an alternative mechanism can induce mutation using the same

transfection procedure so this speculation is not warranted.

Nevertheless, using siPORT NeoFX as the transfection agent, which was found to

be more efficient than NTER, excessive cytotoxicity was not found in our studies, thus,

was not expected to influence the endpoint of the assay. Lepik et al. (Lepik et al., 2003)

suggested that electroporation can change the balance of intracellular ions and even

damage cellular DNA directly. This may then lead to activation of p53-dependent and

independent cell cycle arrest or apoptosis. Indeed, this was reported to be the case with

mock electroporation which increased the level of p53 and nuclear localisation of the

protein (Lepik et al. 2003); coupling electroporation with carrier DNA augmented this

observation further. As p53 can regulate cell cycle checkpoints, apoptosis and DNA repair

(Vousden & Lu, 2002), this may confer an increase in cell susceptibility to triplex mediated

mutagenesis following electroporation and therefore does not necessarily reflect

biologically relevant events.

Page 117: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

117

Oligonucleotides used throughout the mutation study were DNA based and

chemically modified to contain 4 terminal phosphorothioate linkages which we found to

permit nuclease resistance in contrast to the unmodified counterpart. Additionally, DNA

based oligonucleotides are reported to be more likely to induce mutation by causing a

greater helical distortion than those which are RNA based (Kalish et al., 2005, Asensio et

al., 1999). Despite high affinity binding of TFO27 to its target motif, mutation at the

targeted genomic HPRT locus was not reproducibly observed. In the preliminary

experiments treating 3x106 cells in single cultures a dose dependent increase in HPRT

mutants was observed. It is noteworthy, that the oligonucleotides used in the preliminary

studies were from the same synthesised batch, which would be consistent with the results

obtained. However, this observation was never reproduced treating a viable population of

10x106 cells in three independent cultures with oligonucleotide obtained from several

different batches of synthesis.

It is noteworthy, that the presence of spermidine facilitated sequence independent

DNA aggregation of SCR27 in binding studies. Thus, the conditions employed previously

(Ye, Guntaka & Mahato, 2007, Datta et al., 2001, Xu, Glazer & Wang, 2000) incubating

plasmid DNA with a TFO in the presence of spermidine to demonstrate triplex formation

with a mutation endpoint should be interpreted with caution if they are to be extrapolated to

the genomic context as they do not necessarily reflect biologically relevant events.

It is pertinent to add that the triplex target sequence in the supFG1 based reporter

construct previously used (Kalish et al., 2005, Datta et al., 2001, Vasquez et al., 2001,

Faruqi et al., 2000, Vasquez et al., 1999, Wang, Seidman & Glazer, 1996) has been

reported to be a mutagenic hotspot (Hegan et al., 2006); in a contiguous run of guanines

located in the TFO target sequence, spontaneous small deletions and insertions have

been reported to localise upstream, likely due to a polymerase slippage mechanism at this

site (Narayanan et al., 1997). In the present study, the SupFG1 triplex target sequence

Page 118: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

118

was also found to be capable of DNA quadruplex formation in the presence of the

targeting oligonucleotide. Replacing 4 guanine residues in the SupFG1 duplex and

targeting oligonucleotide partially attenuated quadruplex formation at this sequence. A

plausible explanation for the partial and not complete reduction of quadruplex formation in

the mutant SupFG1 construct is that the quadruplex structure may still be capable of

folding with four repeating units of 2 guanine residues, but with reduced efficiency. In

further support was that quadruplex formation in the SupFG1 sequence was favoured in a

potassium buffer. Moreover, a DNA stretch of 30 bases containing 23 guanines (as is the

case in the SupFG1 target sequence) is almost 90% likely to form a DNA quadruplex

structure (Todd, 2007). It may be plausible to suggest that previous oligonucleotide

targeting to this mutation hotspot may be facilitating or stabilising the DNA quadruplex

structure such that it increases the susceptibility of this sequence to mutation. In further

support of this and in accordance with previous results (Wang, Seidman & Glazer, 1996),

DNA quadruplex formation can also result in truncated transcripts and has been implicated

in transcriptional regulation and genomic instability (Broxson, Beckett & Tornaletti, 2011,

Basundra et al., 2010).

Similar to this study, Majumdar et al. (Majumdar et al., 1998) employed a psoralen

conjugated TFO to target an intronic purine rich tract adjacent to exon 5 of the genomic

hprt locus in Chinese hamster ovary cells. The psoralen TFO was electroporated into the

cells and UVA treated after 90 mins. This UVA treatment caused psoralen to covalently

react with both strands of the duplex to generate interstrand cross-links. Following this,

hprt mutant frequencies were reported to be induced to 122 per million cells. In agreement

with the results presented here, omitting the UVA treatment or the psoralen conjugation

from the TFO resulted in the cessation of mutation. However, mutation caused by the un-

conjugated TFO in the intronic sequence may not have resulted in a 6-TG resistant

phenotype. As reported here, a TFO targeting a genomic coding region was also not

Page 119: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

119

capable of inducing mutation above the spontaneous mutation rate of this assay. This

outcome suggests that unless triplex formation is accompanied with a covalent link to the

genomic DNA i.e. psoralen conjugation, the structure is unlikely to result in mutation.

Of relevance to the results presented here, nucleosome core particles have been

shown to inhibit TFO binding to duplex DNA by hindering access to the target sequence

(Brown & Fox, 1996). Moreover, a DNA helicase has been shown to be capable of

unwinding triplex and duplex structures with similar efficiency (Maine & Kodadek, 1994).

Thus, it is probable that triplex formation at genomic DNA is unlikely to block the

progression of a transcription fork and generate a reiterative repair patch which results in

mutation.

Page 120: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

120

Chapter 4

The Genotoxicity of an antisense oligonucleotide via targeted

nucleotide exchange

4.1. Introduction

The potential for ASO to modulate protein expression by translational repression or RNase

H mediated degradation has application as a powerful therapeutic tool (Denli & Hannon,

2003, Zamore, 2001). However, as previously described in chapter 3, recent studies

demonstrating the potential for oligonucleotides to induce site directed mutation have

given rise to concerns from the EMA (European Medicines Agency, 2004). In addition to

oligonucleotide mediated mutagenesis by triplex formation (chapter 3), oligonucleotides

have also been reported to induce mutation in a process known as TNE.

In the process of TNE (section 1.3.2), oligonucleotides, typically over 45 nucleotides

in length, are designed to bind complementary to the non-transcribed (sense) strand within

duplex DNA to form a D-loop structure where the transcribed (antisense) strand of duplex

DNA becomes displaced. Oligonucleotides are engineered to carry a single mismatched

base relative to its homologous sequence such that mutation is directed to the site of the

mismatch in duplex DNA. In a previous study, a mutant GFP construct carrying a single

inactivating point mutation was reported to be corrected to the wild-type sequence,

restoring fluorescence, using a 74-mer oligonucleotide with correction frequencies in the

order of 2% (Bonner & Kmiec, 2009).

The mechanism of TNE has been proposed to be a result of physical incorporation

of oligonucleotides into duplex DNA as a result of (i) HR repair activity or (ii)

oligonucleotides acting as Okazaki fragments during DNA replication (Morozov &

Wawrousek, 2008, Radecke et al., 2006). In either case, the MMR protein MSH2 has been

Page 121: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

121

shown to negatively modulate TNE (Papaioannou, Disterer & Owen, 2009, Maguire &

Kmiec, 2007, Aarts et al., 2006, Dekker, Brouwers & te Riele, 2003, de Wind et al., 1995,

te Riele, Maandag & Berns, 1992).

It is pertinent to highlight that the genotoxicity observed with oligonucleotides

previously employed in TNE cannot be extrapolated to ASO because (i) the length of

oligonucleotides are greater than 21-23 nucleotides used in ASO design (Geary, 2009)

and (ii) oligonucleotides are engineered to target synthetic reporter constructs and so do

not reflect the complexity of the intranuclear milieu. The study presented here has

addressed the genotoxic potential of an ASO targeting genomic DNA in human TK6 cells,

whilst investigating the modulatory roles of MSH2 protein expression and HR repair activity

in TNE.

4.2. Experimental outline

In a study by Bonner et al. (Bonner & Kmiec, 2009), a targeting oligonucleotide (74mer)

was reported to induce DNA DSBs following correction of a mutant GFP construct,

assessed using the alkaline Comet assay. However, DNA DSBs, measured as the comet

tail intensity, following oligonucleotide treatment was not significantly different from control

unless the comet tail intensity was normalised to cell count following treatment which is an

unconventional approach to the analysis. Furthermore, targeting oligonucleotides were

electroporated into aphidicolin synchronised cells where such treatments have been

previously reported to result in DNA DSBs (Lepik et al., 2003).

In this study, we investigated the mutagenic/ clastogenic activity of a 27mer D-loop

forming oligonucleotide (Dloop27) which was engineered to bind the TFO27 triplex target

sequence in exon 3 of the HPRT locus (Fig. 4.1). Dloop27 is complementary to the target

sequence so genotoxicity resulting in cellular resistance to 6-TG is not a result of a

mismatched base. In this instance, the oligonucleotide SCR27 (which lacks sequence

Page 122: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

122

homology to the HPRT locus) was employed as a negative oligonucleotide control. The

genotoxin 4-nitroquinoline (1µM) was used as a positive control.

In a novel approach, an ASO (AD3-HPRTPM), 23 nucleotides in length, was

engineered to bind complementary to the ADAMTS3 gene mRNA as well as contain

sequence homology to the HPRT locus except a single mismatched base (described in

section 2.1.2; Fig. 4.2). It was proposed that AD3-HPRTPM would act as an ASO in

modulating ADAMTS3 protein expression whilst the HPRT locus served as a genomic

reporter of off-target genotoxicity caused by AD3-HPRTPM which could be quantified

using the established HPRT forward mutation assay. The integrity of the genomic TK locus

served as a reporter of sequence independent genotoxicity following oligonucleotide

treatment. As a negative control, an oligonucleotide with the same base composition as

AD3-HPRTPM but in a random order was employed (control) (Fig. 4.2). Similarly, two

other ASO molecules containing homology to a protein target and a selectable locus (such

as HPRT or TK) were designed; however, following ASO sequence examination both were

found to be capable of forming stable hair-pin structures that would render the

oligonucleotides incapable of target binding and so were abandoned for further testing

(data not shown).

In the first instance, the genotoxicity of AD3-HPRTPM was investigated using a

system where MSH2 protein expression was modulated using RNAi. Following this, the

genotoxicity of AD3-HPRTPM was investigated using a system where HR repair activity

was stimulated.

All oligonucleotides were chemically modified at the 5’ and 3’ ends to contain four

terminal phosphorothioate linkages to offer nuclease resistance, as described in section

3.4.4. Oligonucleotide sequences are listed in Table 2.2.

Page 123: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

123

Figure 4.1. The Dloop27 target sequence in exon 3 of the HPRT locus. Dloop27 is engineered to bind complementary to the TFO27 triplex target sequence through a displacement loop structure. Dloop27 contains four terminal phosphorothioate linkages at either end to facilitate nuclease resistance (Table 2.2). The red underlined sequence in duplex DNA highlights the target sequence.

Figure 4.2. The AD3-HPRTPM target sequence in exon 3 of the HPRT locus. AD3-HPRTPM is engineered to bind complementary, except a single mismatched base (underlined in red), to the target sequence (underlined in red) through a D-loop structure. The control oligonucleotide contains the same base composition as AD3-HPRTPM but in a random order and has no sequence homology to the HPRT locus. AD3-HPRTPM and control oligonucleotide contain four terminal phosphorothioate linkages at either end to facilitate nuclease resistance (Table 2.2).

4.3. Methods

1. Antisense and Dloop oligonucleotide design is described in section 2.1.2.

2. Cell treatments and mutation assays are described in section 2.3 and 2.4.

3. Protein extraction is described in section 2.2.5.

4. Immunoblot is described in section 2.6.

5. Genomic DNA extraction is described in section 2.2.3.

6. PCR and DNA sequencing is described in section 2.5.

7. Restriction fragment length polymorphism (RFLP) assay is described in section

2.5.5.

Page 124: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

124

4.4. Results

4.4.1. Genotoxicity of Dloop27

Treating 10x106 human TK6 cells with 1μM, 2μM or 5μM Dloop27 or SCR27 was not

found to be cytotoxic (Fig. 4.3A). Contrary to the study by Bonner et al. (Bonner & Kmiec,

2009) which targeted a synthetic reporter construct, Dloop27 targeting a genomic

sequence lacked mutagenic activity at the targeted HPRT locus (Fig. 4.3B). The in vitro

micronucleus assay was employed to assess the clastogenic activity of Dloop27 in TK6

cells conducted in accordance with OECD guideline 487

(http://iccvam.niehs.nih.gov/SuppDocs /FedDocs/OECD/OECD-TG487.pdf). Here,

Dloop27 targeting genomic DNA failed to induce micronuclei above background in 2000

viable mononucleate cells scored (Fig. 4.4A). Proliferative ability and cytotoxicity following

treatment of TK6 cells was informed by the relative population doubling (RPD) according

to OECD guideline 487 (http://iccvam.niehs.nih.gov/SuppDocs /FedDocs/OECD/OECD-

TG487.pdf). Proliferation of TK6 cells following oligonucleotide treatment was greater than

the minimum 45% recommended for this assay to allow expression of micronuclei and

avoid artifactual positive results caused by excess cytotoxicity (Fig. 4.4B). Thus, the

inability of Dloop27 to exert a clastogenic effect is not an artefact resulting from cells that

have not undergone mitosis.

It is noteworthy that the genotoxicity observed in previous studies employed

oligonucleotides which contained a single mismatched base whereas Dloop27 did not.

Thus, the presence of the mismatched base may be the cause for genotoxicity and was

explored further.

Page 125: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

125

Background

SCR27

Dloop27

SCR27

Dloop27

SCR27

Dloop27

4NQO

0

10

20

30

40

50

60

70

80

90

100

110

Re

lati

ve

To

tal G

row

th (

%)

Background

SCR27

Dloop27

SCR27

Dloop27

SCR27

Dloop27

4NQO

0

10

20

30

40

50

60

70

80

90

100

Mu

tan

t fr

eq

ue

ncy

x10

-6 v

iab

le c

ells

Background

SCR27

Dloop27

SCR27

Dloop27

SCR27

Dloop27

4NQO

0

10

20

30

40

50

60

70

80

90

100

110

120

130

140

Mic

ron

ucle

ate

d m

on

on

ucle

ate

ce

lls

B A

Background

SCR27

Dloop27

SCR27

Dloop27

SCR27

Dloop27

4NQO

0

10

20

30

40

50

60

70

80

90

100

110

120

Rela

tive p

op

ula

tio

n d

ou

blin

g

2M Oligo

5M Oligo

10M Oligo

B A

2M Oligo

5M Oligo

10M Oligo

Figure 4.4. Evaluating the clastogenic activity of Dloop27 in human lymphoblastoid TK6 cells using the in vitro micronucleus assay. An untreated population of cells was used as a negative control (Background) and 1μM 4NQO was used as a positive genotoxin. Dloop27 failed to induce the incidence of micronuclei in 2000 mononucleate cells (A). The relative population doubling of oligonucleotide treated TK6 cells was greater than the minimum 45% accepted for this assay (B). Error bar is mean ± SD of four independent treatments whilst the remaining are the mean of two independent treatments.

Figure 4.3. Investigating the mutagenic effect of Dloop27 at the HPRT locus after treating 10x106 human lymphoblastoid TK6 cells. An untreated population of cells was used to measure the spontaneous mutant frequency (Background) and 1μM 4NQO was used as a positive genotoxin. Cytotoxicity is not evident treating TK6 cells up to 5μM oligonucleotide (A). Dloop27 failed to induce mutation at the targeted genomic HPRT locus (B). Error bar is mean ± SD of four independent treatments whilst the remaining are the mean of two independent treatments.

Page 126: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

126

4.4.2. Biological activity of AD3-hprtPM

Having reported the lack of genotoxicity of Dloop27 at genomic DNA, we sought to

examine the consequence of introducing a single mismatched base.

In the first instance, the antisense activity of AD3-HPRTPM was examined by

quantifying ADAMTS3 protein expression by immunoblot (Fig. 4.5). In brief, TK6 cells were

treated with 2µM, 5µM or 10µM AD3-hprtPM or control oligonucleotide and aliquots

removed at 24h, 48h and 72h (Fig. 4.5, A-C). Protein expression was corrected for

GAPDH protein expression as a loading control. ADAMTS3 protein expression following

AD3-HPRTPM treatment was normalised to ADAMTS3 expression following control

oligonucleotide treatment, at the corresponding dose and time (Fig. 4.5D).

AD3-HPRTPM induced a clear dose and time dependent knockdown of ADAMTS3

protein expression (Fig. 4.5D). ADAMTS3 protein expression was reduced by 20-30% with

5μM and 10μM AD3-HPRTPM treatment by 48h. This reduction in ADAMTS3 protein

expression was 45-70% by 72h with 2µM, 5μM and 10μM AD3-HPRTPM. Thus, AD3-

HPRTPM was found to be a biologically active ASO.

Page 127: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

127

24h 48h 72h

0

10

20

30

40

50

60

70

80

90

100

110

120

130

140

150

*

*

Time (hours)

rela

tiv

e A

DA

MT

S3 e

xp

ressio

n (

%)

Figure 4.5. The antisense activity of AD3-HPRTPM. TK6 cells were treated with 2μM, 5μM or 10μM control or AD3-HPRTPM oligonucleotide. Aliquots of cells were removed for protein extraction. ADAMTS3 protein expression at 24h (A), 48h (B) and 72h (C) was determined by western blot. ADAMTS3 expression was quantified by densitometry and normalised to GAPDH protein expression as a loading control. ADAMTS3 expression from AD3-HPRTPM treatment was made relative to control oligonucleotide treatment (D). Data represent the mean ± SD of three independent treatments. Normalised ADAMTS3 protein expression from AD3-HPRTPM treatment is compared to the control oligonucleotide at the same dose and time using a two-way students t-test. *p<0.05.

B

A

C

D 2M Oligo

5M Oligo

10M Oligo

Page 128: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

128

4.4.3. The of role of mismatch repair in antisense oligonucleotide genotoxicity

Previous reports have highlighted the involvement of the MMR protein MSH2 as a

negative modulator of TNE (Papaioannou, Disterer & Owen, 2009, Maguire & Kmiec,

2007, Aarts et al., 2006, Dekker, Brouwers & te Riele, 2003, de Wind et al., 1995, te Riele,

Maandag & Berns, 1992). In the first instance, TK6 cells were pre-treated with 10nM

MSH2 siRNA to knockdown MSH2 protein expression followed by 5µM AD3-HPRTPM or

control oligonucleotide treatment, to assess the influence of MMR on ASO mediated

cytotoxicity and genotoxicity at the HPRT locus (Fig. 4.6). Cytotoxicity was not observed

with oligonucleotide treatment (Fig. 4.6A). Interestingly, AD3-HPRTPM induced HPRT

mutants ~2 fold above control oligonucleotide, although to a similar magnitude as the

spontaneous HPRT mutant frequency (Fig. 4.6B). AD3-HPRTPM failed to induce mutation

at the non-targeted TK locus suggesting a sequence specific and locus specific mode of

action (Fig. 4.6C).

During MSH2 siRNA pre-treatment of TK6 cells, aliquots of cells were removed for

analysis of MSH2 protein expression by immunoblot to confirm protein knockdown (Fig.

4.7A). MSH2 protein expression was quantified by densitometry and corrected for the β-

ACTIN loading control and normalised to the untreated population of cells (Fig. 4.7B). The

Silencer Select siRNA failed to knockdown MSH2 protein expression up to 72h after

treatment. Treating TK6 cells with a dose range of MSH2 siRNA up to 20nM failed to

knockdown protein expression (Fig. 4.8). In fact, treating TK6 cells with siRNA for 24h

appeared to result in a dose dependent increase in MSH2 protein expression which

peaked at 24h up to ~1.6 fold above control (Fig. 4.8B). Although this Silencer Select

siRNA is guaranteed by the manufacturer to give a 100% knockdown, this claim was not

experimentally verified here. However, MSH2 protein knockdown in a sub population of

cells is not likely to be detected using an immunoblot as this technique informs average

protein expression within a population.

Page 129: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

129

Background

Control

AD3-hprtP

MEM

S

0

10

20

30

40

50

60

70

80

90

100

110

120

Re

lati

ve

To

tal G

row

th (

%)

Background

Control

AD3-hprtP

MEM

S

0

5

10

15

20

25

30

35

40

**

Mu

tan

t fr

eq

ue

ncy

x10

-6 v

iab

le c

ells

Figure 4.6. Investigating the mutagenic effect of an ASO containing a single mismatched base. Human TK6 cells were pre-treated for 24h with 10nM MSH2 Silencer Select siRNA and then treated with 5μM oligonucleotide. An untreated population of cells was used to measure the spontaneous mutant frequency at each locus (Background) and 5μg/ml EMS was used as a positive genotoxin. Cytotoxicity is not evident following oligonucleotide treatment (A). AD3-HPRTPM induced mutation at the targeted HPRT locus ~2 fold above the control oligonucleotide (B) but failed to induce mutation at the non-targeted TK locus (C). Error bars represent mean ± SD of three independent treatments. **p<0.01 using two way students t-test.

Background

Control

AD3-hprtP

MEM

S

0

10

20

30

40

50

60

Mu

tan

t fr

eq

ue

ncy

x10

-6 v

iab

le c

ells

A

C

B

Page 130: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

130

0 5 10 15 20 25 30 35 40 45 50 55 60 65 70 750.0

0.1

0.2

0.3

0.4

0.5

0.6

0.7

0.8

0.9

1.0

1.1

1.2

1.3

Time (hours)

No

rmalised

MS

H2 e

xp

ressio

n

Figure 4.7. Quantitation of MSH2 knockdown in human TK6 cells treated with (S) or without (U) 10nM Silencer Select siRNA for 24h. Aliquots of TK6 cells at the desired time point were harvested for protein extraction. MSH2 protein expression was determined by immunoblot. In brief, 20μg of protein was electrophoresed through an SDS-polyacrylamide gel and transferred to a PVDF membrane. Membrane was stained with primary mouse anti-human MSH2 antibody (1:1000) overnight and visualised (A). β-ACTIN protein expression was used as a loading control using primary mouse anti-human β-ACTIN antibody (1:40,000) for 1h (A). Protein expression was quantitated using densitometry. MSH2 protein expression was corrected for the β-ACTIN loading control and normalised to the untreated population of cells at the respective time point (B).

0 5 10 15 20 25 30 35 40 45 50 55 60 65 70 750.0

0.1

0.2

0.3

0.4

0.5

0.6

0.7

0.8

0.9

1.0

1.1

1.2

1.3

1.4

1.5

1.6 5nM

10nM

20nM

Time (h)

No

rmalised

MS

H2 e

xp

ressio

n

Figure 4.8. Optimising MSH2 knockdown using Silencer Select siRNA. Human TK6 cells were treated without siRNA (0nM) or with 5nM, 10nM or 20nM MSH2 siRNA for 24h, washed and cultured for the remaining time. Aliquots of cells were harvested for protein extraction at the selected time points. MSH2 protein expression was determined by immunoblot. In brief, 20μg of protein was electrophoresed through an SDS-polyacrylamide gel and transferred to a PVDF membrane. Membrane was stained with primary mouse anti-human MSH2 antibody (1:1000) overnight and visualised (A). β-ACTIN protein expression was employed as a loading control using primary mouse anti-human β-ACTIN antibody (1:40,000) for 1h (A). Protein expression was quantitated using densitometry. MSH2 protein expression was corrected for the β-ACTIN loading control and

normalised to the untreated population of cells (0nM) at the respective time point (B).

A

B

A

B

Page 131: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

131

Background

Control

AD3-hprtP

M

Control

AD3-hprtP

M

Control

AD3-hprtP

M

EMS

0

20

40

60

80

100

120

140

160

Re

lati

ve

To

tal G

row

th (

%)

Background

Control

AD3-hprtP

M

Control

AD3-hprtP

M

Control

AD3-hprtP

M

EMS

0

10

20

30

40

50

60

Mu

tan

t fr

eq

ue

ncy

x10

-6 v

iab

le c

ells

Background

Control

AD3-hprtP

M

Control

AD3-hprtP

M

Control

AD3-hprtP

M

EMS

0

10

20

30

40

50

60

70

80

Mu

tan

t fr

eq

ue

ncy

x10

-6 v

iab

le c

ells

Figure 4.9. Investigating the genotoxic effect of AD3-HPRTPM using human TK6 cells pre-treated with MSH2 Silencer Select siRNA. Following 10nM siRNA pre-treatment for 24h, 3x106 TK6 cells were treated with 2μM, 5μM or 10μM oligonucleotide. An untreated population of cells was used to measure the spontaneous mutant frequency (Background) and 5μg/ml EMS was used as a positive genotoxin. Cytotoxicity was not evident following oligonucleotide treatment up to 10μM (A). AD3-HPRTPM failed to induce mutation at the targeted HPRT locus (B) and the non-targeted TK locus (C). Data represent mean ± SD of three independent treatments.

B

A

C

2M Oligo

5M Oligo

10M Oligo

Nevertheless, AD3-HPRTPM appeared to induce mutation ~2 fold above control

oligonucleotide treatment at a single dose and so was important to reproduce this

observation using a dose range of AD3-HPRTPM (Fig. 4.6). Thus, human TK6 cells were

pre-treated with 10nM MSH2 siRNA as before, followed by oligonucleotide from 2µM up to

10µM (Fig. 4.9). In this experiment, up to 45% cytotoxicity was observed with 10µM

oligonucleotide treatment (Fig. 4.9A). However, AD3-HPRTPM failed to induce mutation at

the HPRT and TK loci (Fig. 4.9B and C, respectively). It is important to note that although

AD3-HPRTPM induced mutation ~2 fold above the control oligonucleotide (Fig. 4.6B), this

mutant frequency was similar to the untreated population of cells measuring the

spontaneous mutant frequency.

Page 132: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

132

Background

Control

AD3-hprtP

M

Control

AD3-hprtP

M

Control

AD3-hprtP

M

EMS

0

10

20

30

40

50

60

70

80

90

100

110

120

130

Re

lati

ve

To

tal G

row

th (

%)

Background

Control

AD3-hprtP

M

Control

AD3-hprtP

M

Control

AD3-hprtP

M

EMS

0

5

10

15

20

25

30

35

40

Mu

tan

t fr

eq

ue

ncy

x10

-6 v

iab

le c

ells

Background

Control

AD3-hprtP

M

Control

AD3-hprtP

M

Control

AD3-hprtP

M

EMS

0

10

20

30

40

50

60

70

80

Mu

tan

t fr

eq

ue

ncy

x10

-6 v

iab

le c

ells

Figure 4.10. Investigating the genotoxic effect of AD3-HPRTPM using human TK6 cells. An untreated population of cells was used to measure the spontaneous mutant frequency (Background) and 5μg/ml EMS was used as a positive genotoxin. Cytotoxicity is not evident following treatment of 3x106 TK6 cells with 2μM, 5μM or 10μM oligonucleotide (A). AD3-HPRTPM failed to induce mutation at the targeted HPRT locus (B) and the non-targeted TK locus (C). Data represent mean ± SD of three independent treatments.

B

A

C

2M Oligo

5M Oligo

10M Oligo

The above experiments describe TK6 cells pre-treated with siRNA to knockdown

MSH2 protein expression. However, average MSH2 protein expression following 10nM

siRNA treatment increased 1.2-1.4 fold above control by 24h (Fig. 4.7 and 4.8). Thus, at

the time of treatment of TK6 cells with AD3-HPRTPM, average MSH2 protein expression

was above basal level. The next plausible approach was to examine the genotoxicity of

AD3-HPRTPM without siRNA treatment (Fig. 4.10). Cytotoxicity following oligonucleotide

treatment up to 10µM was no greater than 30% (Fig. 4.10A). However, AD3-HPRTPM

failed to induce mutation at the targeted HPRT and non-targeted TK loci above the

detection limits of this assay (Fig. 4.10B and C, respectively).

Page 133: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

133

Ergo, the initial genotoxicity observed with AD3-HPRTPM was not reproduced with/

without MSH2 siRNA pre-treatment using a dose range of oligonucleotide up to 10µM.

Thus, it was important to validate the initial observation as a true mutagenic event. For this

reason, a restriction fragment length polymorphism (RFLP) assay was designed.

Mutant HPRT clones were picked from 96 well plates and expanded in 6-well plates

for a week. Genomic DNA from clones was extracted and used for PCR amplification of

exon 3 in the HPRT locus. In particular, a 177bp stretch encompassing the AD3-HPRTPM

target sequence was amplified. According to TNE, AD3-HPRTPM mediated mutation

should occur at the site of the mismatched base. AD3-HPRTPM was designed to induce a

point mutation (G→T transversion) in exon 3 of the HPRT locus such that it causes the

loss of a 5’-↓GATC-3’ Mbo I restriction enzyme recognition sequence. Thus, subjecting

177bp PCR fragments to Mbo I restriction digestion should yield resistant fragments if

AD3-HPRTPM induced site specific mutation. If mutation does not alter the 5’-GATC-3’

recognition sequence then PCR fragments will cleave into two fragments (~140bp and

37bp).

A typical PCR reaction is shown in Fig. 4.11A yielding a PCR fragment close to

200bp (177bp). Out of 20 HPRT mutant clones from the control oligonucleotide treatment

only 14 successfully PCR; Mbo I restriction enzyme digestion of 14 PCR products from

control clones showed cleavage into two fragments (Fig. 4.11B). Thus, mutation in the

control samples did not alter the 5’-GATC-3’ sequence within exon 3.

Similarly, 35 HPRT mutant clones from the AD3-HPRTPM treatment were subject

to PCR and only 28 were successful. These 28 PCR products were subject to Mbo I

restriction digestion. Unexpectedly, all 28 PCR clones were cleaved by Mbo I into two

fragments (Fig. 4.11C). As mentioned above, AD3-HPRTPM induced HPRT mutants ~2

fold above the control oligonucleotide, but the magnitude of this mutation was similar to the

spontaneous mutant frequency. Furthermore, this genotoxicity was not reproduced under

Page 134: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

134

Figure 4.11. Mbo I restriction enzyme digestion of PCR products. Isolated HPRT mutant clones from control (1-14) or AD3-HPRTPM (21-48) oligonucleotide treatments from Fig. 4.6B were subject to genomic DNA extraction followed by 30 cycles of PCR to yield a 177bp product. PCR reactions (50μl) contained 300ng of genomic DNA, 5U Tfi DNA polymerase, 200μM dNTP, 200nM primer pair 2 (Table 2.4) and 2mM MgCl2. A typical PCR reaction using genomic DNA from control oligonucleotide clones is shown in panel A. From 20 control clones 14 successfully PCR and all were cleaved by Mbo I restriction enzyme (B). From 35 AD3-HPRTPM induced clones 28 successfully PCR and all cleaved by Mbo I restriction enzyme (C).

B

C

A

the same conditions in subsequent experiments. These data suggest that AD3-HPRTPM

failed to induce site specific mutation above control oligonucleotide treatment and the

spontaneous mutant frequency at the HPRT locus, under the conditions investigated so

far.

A particular concern was that AD3-HPRTPM mediated site specific mutation within

exon 3 of the HPRT locus would not result in a 6-TG resistant phenotype. To explore this

further, an oligonucleotide was engineered to contain sequence homology to exon 3 of the

HPRT locus except a single mismatched base (Val-HPRTPM). Point mutation induced by

Page 135: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

135

Val-HPRTPM (C>T transition) has been previously reported to result in a 6-TG resistant

phenotype (Meng et al., 2002). Mutation at this site is expected to change the codon from

CTC (Leucine) to TTC (phenylalanine). Treating human TK6 cells with 2μM, 5μM and

10μM Val-HPRTPM was not cytotoxic but nor was it genotoxic at the HPRT or TK loci (Fig.

4.12). This supports the observations discussed above. However, it does not directly

address whether AD3-HPRTPM mediated mutation results in a 6-TG resistant phenotype.

As knockdown of MSH2 protein expression using RNAi technology failed, a MMR

deficient cell line was employed to explore the role of MMR in oligonucleotide mediated

genotoxicity. The HCT116 cells line has a mutation in the MLH-1 gene which renders the

MMR signalling pathway redundant (Papadopoulos et al., 1994).

HCT116 cells were treated with 5μM control, AD3-HPRTPM or Val-HPRTPM and

plated to determine cytotoxicity and genotoxicity. Oligonucleotide treatment was not found

to be cytotoxic nor was it genotoxic at the HPRT locus (Fig. 4.13). It is noteworthy that

400μg/mL EMS was used as a positive genotoxin using HCT116 cells as opposed 5μg/mL

EMS for TK6 cells to elicit a similar mutagenic response. Also, the spontaneous HPRT

mutant frequency in HCT116 cells was ~200x10-6 as opposed to 5-10x10-6 for TK6 cells, in

line with previous reports (Meng et al., 2002, Glaab, Kort & Skopek, 2000). This suggests

a higher mutation rate in HCT116 cells as well as a requirement for greater genotoxin

concentrations (80 fold) to elicit a mutagenic response. Thus, due to these factors, subtle

induction of HPRT mutants by AD3-HPRTPM would be less likely to be detected using

HCT116 cells as it would be masked by the spontaneous mutant frequency.

Page 136: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

136

Background

Control

Val-hprtP

M

Control

Val-hprtP

M

Control

Val-hprtP

MEM

S

0

5

10

15

20

25

30

35

40

Mu

tan

t fr

eq

ue

ncy

x10

-6 v

iab

le c

ells

Figure 4.12. Investigating the genotoxic effect of Val-HPRTPM using human TK6 cells. Val-HPRTPM was engineered to induce a point mutation within the HPRT locus which has previously been reported to result in a 6-TG resistant phenotype. An untreated population of cells was used to measure the spontaneous mutant frequency (Background) and 5μg/ml EMS was used as a positive genotoxin. Cytotoxicity is not evident following treatment of 3x106 TK6 cells with 2μM, 5μM or 10μM oligonucleotide (A). Val-HPRTPM failed to induce mutation at the targeted HPRT locus (B) and the non-targeted TK locus (C). Data represent mean ± SD of three independent treatments.

Background

Control

AD3-hprtP

M

Val-hprtP

MEM

S

0

10

20

30

40

50

60

70

80

90

100

110

120

Clo

nin

g E

ffic

ien

cy

(%)

Background

Control

AD3-hprtP

M

Val-hprtP

MEM

S

0

200

400

600

800

1000

1200

1400

1600

1800

2000

Mu

tan

t fr

eq

ue

ncy

x10

-6 v

iab

le c

ells

Figure 4.13. Investigating the genotoxic effect of AD3-HPRTPM and Val-HPRTPM in a mismatch repair deficient cell line. Human colon epithelial HCT116 cells were treated with 5μM oligonucleotide. An untreated population of cells was used to measure the spontaneous mutant frequency (Background) and 400μg/ml EMS was used as a positive genotoxin. Cytotoxicity following treatment was not evident (A). All oligonucleotides failed to induce mutation at the targeted HPRT locus (B). Data represent mean ± SD of three independent treatments.

B C

A

2M Oligo

5M Oligo

10M Oligo

Background

Control

Val-hprtP

M

Control

Val-hprtP

M

Control

Val-hprtP

MEM

S

0

10

20

30

40

50

60

70

80

Mu

tan

t fr

eq

ue

ncy

x10

-6 v

iab

le c

ells

Background

Control

Val-hprtP

M

Control

Val-hprtP

M

Control

Val-hprtP

MEM

S

0

10

20

30

40

50

60

70

80

90

100

110

120

130

140

Re

lati

ve

To

tal G

row

th (

%)

B

A

Page 137: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

137

4.4.4. The of role of homologous recombination repair in antisense oligonucleotide

genotoxicity

The mechanism of mutation for oligonucleotide mediated TNE has been proposed to

involve the HR repair pathway (Morozov & Wawrousek, 2008, Radecke et al., 2006). To

investigate this further, human TK6 cells were pre-treated with the genotoxin MMS to

induce the HR repair pathway. The RAD51 protein, which is involved in strand alignment

during HR (Gupta et al., 1997, Sung & Robberson, 1995), was used as a marker for

induction of HR repair activity.

The concentration range of MMS used to induce RAD51 protein expression in TK6

cells was carefully considered, as a genotoxic response in subsequent mutagenicity

studies may have masked subtle mutation induced by AD3-HPRTPM treatment. On this

basis, TK6 cells were treated with 0μg/mL, 0.1μg/mL, 0.2μg/mL or 0.5μg/mL MMS for 24h.

Following treatment, TK6 cells were washed to remove the treatment media and passaged

for a further 48h. Treated TK6 cells were harvested at 2h, 4h, 8h, 24h, 48h and 72h for

analysis of RAD51 protein expression by immunoblot (Fig. 4.14A). MMS induced RAD51

protein expression was corrected for the GAPDH loading control and normalised to the

control (0μg/mL) at the respective time. MMS treatment of TK6 cells showed a clear

induction of RAD51 protein expression (Fig. 4.14B). At the lowest dose of MMS

(0.1μg/mL), RAD51 protein expression was induced ~2.5 fold above control at 48h

returning to basal level by 72h. The highest RAD51 protein induction occurred with

0.2μg/mL MMS; RAD51 protein expression was almost 3 fold by 24h peaking up to ~5.5

fold at 48h. The 0.5μg/mL MMS treatment weakly induced RAD51 protein expression (~2

fold by 24h returning to basal by 48h). The lack of induction at this dose may be a result of

the activation of an alternative repair mechanism or perhaps apoptosis due to the extent of

cellular insult.

Page 138: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

138

Figure 4.14. Inducing the homologous recombination repair pathway. TK6 cells were treated with the genotoxin MMS for 24h, washed and maintained in exponential growth for the desired time. Aliquots of cells were harvested at 2h, 4h, 8h, 24h, 48h and 72h. RAD51 protein expression was determined by immunoblot (A). GAPDH was used as a loading control (A). Lanes 1-4 are 0μg/ml, 0.1μg/ml, 0.2μg/ml and 0.5μg/ml MMS, respectively. Densitometry was used to quantitate protein expression. RAD51 protein expression was corrected for the GAPDH loading control and normalised to 0µg/ml solvent control at the corresponding time (B).

0 5 10 15 20 25 30 35 40 45 50 55 60 65 70 750

1

2

3

4

5

60.1g/ml

0.2g/ml

0.5g/ml

Time (h)

No

rmalised

Rad

51 f

old

ch

an

ge

B

A

RAD51 protein expression was found to be maximally induced from 24h to 48h with

0.2μg/mL MMS. Thus, TK6 cells were pre-treated with 0.2μg/mL MMS for 24h, washed

and treated with oligonucleotide (Fig. 4.15). In this study, Val-HPRTPM was included as an

oligonucleotide control targeting the HPRT locus where mutation at the site of the

mismatched bases would result in 6-TG resistance, as previously discussed (section

4.4.3). Additionally, untreated TK6 cells (Untreated) i.e. without 24h MMS treatment were

included to investigate the cytotoxic/genotoxic effect of the MMS pre-treatment.

Page 139: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

139

Following oligonucleotide treatment, dose dependent cytotoxicity was observed up

to 10μM oligonucleotide (Fig. 4.15A). Cytotoxicity was no greater than 65% which is less

than the maximum accepted 80% for this assay (Clements, 2000, Clive et al., 1995).

The HPRT and TK mutant frequency between the untreated and MMS pre-treated

cultures were not significantly different (Fig. 4.15B and C, comparing untreated and

background). Thus, the MMS pre-treatment was not found to be significantly genotoxic.

Most interestingly, AD3-HPRTPM induced a dose dependent increase in HPRT

mutants. Treatment of TK6 cells with 5µM AD3-HPRTPM induced the HPRT mutant

frequency ~1.5 fold above 5µM control oligonucleotide treatment whereas 10µM treatment

induced the HPRT mutant frequency ~4.4 fold (Fig. 4.15B). Similarly, 10μM Val-HPRTPM

induced HPRT mutants ~4 fold above 10µM control oligonucleotide treatment (Fig. 4.15B).

As expected, both AD3-HPRTPM and Val-HPRTPM failed to induce mutation at the non-

targeted TK locus (Fig. 4.15C). Thus, both AD3-HPRTPM and Val-HPRTPM were capable

of sequence specific and locus specific mutation when RAD51 protein expression was

induced in human TK6 cells.

Page 140: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

140

Figure 4.15. Investigating the genotoxic effect of AD3-HPRTPM in a HR repair induced system. TK6 cells were pre-treated with 0.2μg/ml MMS for 24h to induce HR repair activity, except the untreated treatment which measures the cytotoxic/genotoxic effect of the MMS pre-treatment. The background treatment measures the spontaneous mutant frequency following MMS pre-treatment and 5μg/ml EMS was used as a positive genotoxin. Viability below the minimum accepted 20% RTG is not evident following treatment of 3x106 TK6 cells with 2μM, 5μM or 10μM oligonucleotide (A). AD3-HPRTPM induced a dose dependent increase in HPRT mutants up to 4.4 fold (B). Val-HPRTPM (10μM) induced HPRT mutants to a similar magnitude as 10μM AD3-HPRTPM (B). Both oligonucleotides failed to induce mutation at the non-targeted TK locus (C). Data represent mean ± SD of three independent treatments. * p<0.05 using two-way students t-test.

Untreate

d

Background

Control

AD3-hprtP

M

Control

AD3-hprtP

M

Control

AD3-hprtP

M

Val-hprtP

MEM

S

0

10

20

30

40

50

60

70

80

90

100

110

120

130

Re

lati

ve

To

tal G

row

th (

%)

Untreate

d

Background

Control

AD3-hprtP

M

Control

AD3-hprtP

M

Control

AD3-hprtP

M

Val-hprtP

MEM

S

0

20

40

60

80

100

120

140

160

*

ns

ns

Mu

tan

t fr

eq

ue

ncy

x10

-6 v

iab

le c

ells

Untreate

d

Background

Control

AD3-hprtP

M

Control

AD3-hprtP

M

Control

AD3-hprtP

M

Val-hprtP

MEM

S

0

10

20

30

40

50

60

70

80

90

100

Mu

tan

t fr

eq

ue

ncy

x10

-6 v

iab

le c

ells

B

A

C

2M Oligo

5M Oligo

10M Oligo

Untreated; No MMS

Background; Spontaneous with MMS

According to TNE, mutation within duplex DNA is directed to the site of the mismatched

base (Bonner & Kmiec, 2009, Olsen et al., 2009, Morozov & Wawrousek, 2008, Aarts et

al., 2006, Radecke et al., 2006, Olsen et al., 2005, Dekker, Brouwers & te Riele, 2003).

Thus, to confirm sequence specific mutation by AD3-HPRTPM at the HPRT locus, a RFLP

assay was designed as discussed in section 4.4.3. In this instance, the target region of

AD3-HPRTPM, within exon 3 of the HPRT locus, was PCR amplified to yield a 1kb

fragment. AD3-HPRTPM mediated mutation at the site of the mismatched base would be

expected to result in a G>T transversion causing the loss of the 5’-GATC-3’ Mbo I

Page 141: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

141

restriction enzyme recognition sequence to 5’-TATC-3’. Thus, PCR fragments containing

the mutant sequence would be rendered resistant to Mbo I digestion. Those which

retained the wild-type sequence would yield 2 cleavage products of ~550bp and ~450bp.

HPRT mutant clones from 10µM AD3-HPRTPM or control oligonucleotide treatment

were isolated, expanded for up to 10 days and harvested for genomic DNA extraction and

PCR amplification. Interestingly, Mbo I digestion of 14 control and 29 AD3-HPRTPM

induced HPRT mutant clones all resulted in cleavage of all PCR products into 2 fragments,

contradicting the expected mechanism of mutagenesis (Fig. 4.16A and B). Those which

failed to PCR across the 1kb sequence in exon 3, were subject to PCR amplification

across a 177bp stretch and Mbo I digested (Fig. 4.16C). Those which failed to PCR across

exon 3 of the HPRT locus using primer pairs 2 and 3 (Table 2.4) were subject to PCR

amplification across the GAPDH locus to inform the integrity of the genomic DNA (primer

pair 4, Table 2.4). From the 4 control and 5 AD3-HPRTPM induced clones, all successfully

PCR across the GAPDH locus (Fig. 4.16D). This suggests there may be a problem across

or within exon 3 in these samples such that the primer (s) could not bind to PCR amplify

the region.

To further investigate the molecular nature of mutation within exon 3 of HPRT

mutant clones, 1kb PCR products were sent for DNA sequencing. Raw sequence data

was analysed using EBI multiple sequence alignment tool CLUSTALW. Mutation in each

spectrum was then manually confirmed using NCBI-BLAST. Sequencing of PCR amplified

HPRT mutant clones (41 AD3-HPRTPM induced HPRT mutant clones and 29 control)

supported the retention of the wild-type 5’-GATC-3’ sequence at position 138 (Fig. 4.17).

However, DNA sequencing revealed a single base deletion (position 171; 34%) adjacent to

a single G>A transition mutation (position 172; 29%) downstream of the AD3-HPRTPM

target sequence (underlined; position 134-156). Importantly, these mutations were

primarily found in AD3-HPRTPM induced HPRT mutant clones and not control. All of the

Page 142: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

142

G>A transition mutations (position 172) were found to be adjacent to the single thymidine

deletion at position 171. The frequency of particular point mutations upstream (G>A

position 126; 58% control vs. 81% AD3-HPRTPM) and downstream (G>A position 158;

19% control vs. 51% AD3-HPRTPM) of the AD3-HPRTPM target sequence were also

found to be effected following AD3-HPRTPM treatment (Fig. 4.17). Also, ~86% of clones

harbouring the thymidine deletion at position 171 either had a G>A transition at position

175 or a G>A transition at position 158 (although the frequency of the G>A transition at

position 175 was greater than mutation at position 158; 57% vs. 43%, respectively).

Page 143: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

143

Mbo I

PCR

Mbo I

PCR

Mbo I Mbo I

A

B

C D

Figure 4.16. Mbo I restriction enzyme digestion of HPRT mutant PCR products. In brief, isolated 6-TG resistant clones from 10µM control (1-18) or AD3-HPRTPM (19-53) oligonucleotide treatments were subject to DNA extraction followed by 30 cycles of PCR to yield a 1018bp product encompassing the AD3-HPRTPM target sequence in exon 3. PCR reactions (50μl) contained 300ng of genomic DNA, 5U Tfi DNA polymerase, 200μM dNTP, 200nM primer pair 3 (Table 2.4) and 2mM MgCl2. A typical PCR reaction is shown for control (A) and AD3-HPRTPM (B) oligonucleotide clones. From 18 control HPRT clones, 14 successfully PCR and all cleaved by Mbo I restriction enzyme (A). From 35 AD3-HPRTPM induced HPRT clones 29 successfully PCR and all cleaved by Mbo I restriction enzyme (B). Those which failed to PCR across the 1kb HPRT sequence, were subjected to PCR across a 177bp region (C). Only clone 49 successfully PCR but was also cleaved by Mbo I. Those which failed to PCR across the HPRT locus were subject to PCR across the GAPDH locus using primer pair 4 (Table 2.4) (D). All clones successfully PCR across GAPDH.

PCR PCR

GAPDH

Mbo I Mbo I

Page 144: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

144

Figure 4.17. Distribution of AD3-HPRTPM mediated mutation in exon 3 of the HPRT locus. PCR products (1kb) from Fig. 4.16 were sent for DNA sequencing to inform the mechanism of AD3-HPRTPM mutation. Sequences are numbered from the first base of exon 3 (1 to 164). The AD3-HPRTPM target sequence is underlined from position 134-156. * above guanine (position 138) indicates the site of the mismatched base in AD3-HPRTPM where G>T transversion mutation was predicted. Numbers and letters within a circle represent the frequency (%) and type of mutation from 41 AD3-HPRTPM induced HPRT mutant clones whereas those in a square are from 29 control induced HPRT mutant clones. + indicates an insertion and ∆ indicates a deletion. Mutation common to both control and AD3-HPRTPM with the same frequency have been removed from the figure for clarity.

Page 145: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

145

4.5. Discussion

Data presented in this study suggest that a biologically active ASO, AD3-HPRTPM,

was capable of inducing mutation at genomic DNA in human lymphoblastoid cells when

RAD51 protein expression was induced. Oligonucleotides have been previously reported

to be capable of inducing mutation within a homologous sequence in duplex DNA, in a

process known as TNE, where mutation was a result of a mismatched base within the

oligonucleotide (Bonner & Kmiec, 2009, Olsen et al., 2009, Papaioannou, Disterer &

Owen, 2009, Morozov & Wawrousek, 2008, Maguire & Kmiec, 2007, Aarts et al., 2006,

Radecke et al., 2006, Olsen et al., 2005, Dekker, Brouwers & te Riele, 2003, de Wind et

al., 1995, te Riele, Maandag & Berns, 1992)(Dekker, Brouwers & te Riele, 2003)(Dekker,

Brouwers & te Riele, 2003). Furthermore, Bonner et al. (Bonner & Kmiec, 2009) reported

DNA DSBs were a consequence of TNE; however, DNA DSBs were only reported once

comet tail intensities were normalised to cytotoxicity following electroporation of

oligonucleotides; a treatment which itself can result in DNA DSBs (Lepik et al., 2003). In

this study, Dloop27 targeting genomic DNA was not mutagenic or clastogenic using the in

vitro micronucleus assay conducted to OECD guidelines

(http://iccvam.niehs.nih.gov/SuppDocs /FedDocs/OECD/ OECD-TG487.pdf).

An important point to consider is that oligonucleotides employed in previous studies

reporting TNE tend to be greater than 47 nucleotides in length and contain a single

mismatched base relative to the target sequence. Thus, in this study, AD3-HPRTPM (23

nucleotides in length) was engineered to reflect the length of an ASO therapeutic and

chemically modified to contain phosphorothioate linkages, which is the most common

modification used in ASO therapeutics (Geary, 2009, Buchini & Leumann, 2003).

Although AD3-HPRTPM was biologically active as an antisense molecule, reducing

ADAMTS3 expression by 70%, mutation at the targeted HPRT locus in untreated human

lymphoblastoid TK6 cells was not observed above the detection limit of the assay.

Page 146: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

146

However, in a system where RAD51 protein expression was stimulated prior to treatment

with oligonucleotide, AD3-HPRTPM induced locus and sequence specific mutation ~4.4

fold (40x10-6) above control (9x10-6). TNE by a micro-injected oligonucleotide was also

absent in one cell mouse embryos until coinjection of purified RAD51 protein with

oligonucleotide (Morozov & Wawrousek, 2008). In that instance, mutant frequencies were

in the order of 9.9% (99,000x10-6) when 313 cells were screened which is significantly

higher than reported here. A similar observation as this study was made using an E. Coli

strain which lacked functional RecA protein (a RAD51 homolog) where TNE to correct a

kanamycin resistance gene was absent until purified RecA was supplied with

oligonucleotide (Drury & Kmiec, 2003). However, in the study presented here, RAD51

protein was not exogenously supplied but rather endogenously induced.

Using an engineered mutant GFP construct, mutant frequencies by a 74mer

oligonucleotide were found to be in the order of 2% using the mismatch deficient HCT116

cell line (Bonner & Kmiec, 2009). In the study presented here, using a genomic DNA

target, AD3-HPRTPM failed to induce mutation using the HCT116 cell line, possibly a

result of higher spontaneous mutation in these cells masking subtle genotoxicity. In

support of this, the spontaneous HPRT mutant frequency was observed to be remarkably

higher in HCT116 cells (200x10-6) than in TK6 cells (~5x10-6). Additionally, HCT116 cells

have functional MSH2 protein so it can still exert its anti-recombinogenic activity (de Wind

et al., 1995, te Riele, Maandag & Berns, 1992); HR repair has been reported to be

inhibited by MSH2 protein when DNA in homologous pairing has greater than 0.6%

sequence divergence (de Wind et al., 1995). For example, a 47mer oligonucleotide with a

single mismatched base would correlate to ~2% sequence divergence, whereas AD3-

HPRTPM would have a sequence divergence of 4.3%. This may explain why AD3-

HPRTPM mediated genotoxicity was not observed until RAD51 protein expression was

sufficiently induced to overcome the inhibitory effects of MSH2 protein.

Page 147: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

147

Sequencing AD3-HPRTPM induced HPRT mutant clones revealed the predicted

mutation at the site of the mismatched base was absent in all clones. However, amongst

other mutations, a single base deletion and point mutation adjacent to but downstream of

the AD3-HPRTPM target sequence was observed in mutant clones primarily treated with

AD3-HPRTPM. AD3-HPRTPM binding to its target sequence on the sense strand (non-

transcribed) may be facilitated by the strand pairing properties of RAD51 protein perhaps

during DNA replication. Indeed, the ability of RAD51 to pair single stranded DNA with

homologous double stranded DNA in an adenosine triphosphate (ATP) dependent process

has been previously reported (Gupta et al., 1997, Baumann, Benson & West, 1996, Sung

& Robberson, 1995). Upon strand invasion of AD3-HPRTPM, a D-loop structure is formed

which results in the displacement of the antisense strand. Following this, the model can be

extrapolated from Hanawalt (Hanawalt, 1994) and Wang (Wang, Seidman & Glazer,

1996). The D-loop structure may result in a physical blockade to a progressing replication

fork causing it to revert back to a natural pause site generating a reiterative repair patch.

Repeated attempts in progression of a replication fork may result in mutation introduced by

the natural error frequency of the polymerase. It may be the bound AD3-HPRTPM is

removed by the helicase activity associated with a replication fork but repeated cycles of

binding inhibition of replication and re-iterative repair may increase the probability of a

mutagenic event.

Alternatively, perhaps through HR repair during S-phase (Johnson & Jasin, 2001,

Takata et al., 1998), AD3-HPRTPM physically incorporates into the genome (Radecke et

al., 2006). The influence of cell cycle progression has been reported to affect TNE with

optimum mutation occurring during early S phase (Olsen et al., 2005). Once incorporated,

AD3-HPRTPM with the mismatched base and the non-canonical phosphorothioate

linkages at either end may stimulate the nucleotide excision repair (NER) pathway in

recognising the sulphur within the phosphodiester backbone as a lesion, ultimately

Page 148: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

148

resulting in the excision of an encompassing fragment of 27-29nt in length (Svoboda et al.,

1993, Huang et al., 1992). In support of this, the introduction of phosphorothioate bonds

into a DNA double helix has been reported to cause structural alterations (Kanaori et al.,

1999), which may stimulate the repair pathway. This latter mechanism may explain why

the mismatched base from AD3-HPRTPM treatment was not observed in HPRT mutant

clones but observed in previous studies using oligonucleotides that are far greater in

length than the NER repair pathway is able to cleave.

Recent efforts have focused on efficient and site specific delivery methods to

improve bioavailability/ biological efficacy of ASO therapeutics and reduce susceptibility to

degradation by nucleases in blood (Morvan et al., 1993, Akhtar, Kole & Juliano, 1991,

Hoke et al., 1991). However, increasing intracellular concentration of ASO as a result of

efficient delivery systems may increase the probability of binding to genomic DNA, which

may in turn result in an increased risk of introducing heritable sequence alterations.

These data suggest that an ASO could induce mutation if it is involved in strand

invasion mediated by high RAD51 protein activity.

Page 149: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

149

Chapter 5

Incorporation and genotoxicity of oligonucleotide degradation

products into the genome

5.1. Introduction

ASO therapeutics can be used to modulate protein expression in a mechanism known as

RNAi (Denli & Hannon, 2003, Zamore, 2001). However, ASO therapeutics are chemically

modified to offer protection from nuclease mediated degradation in order to remain

biologically active. One of the most common chemical modifications used in ASO design is

the phosphorothioate linkage (Geary, 2009, Buchini & Leumann, 2003, Pirollo et al., 2003);

here, a non-bridging oxygen in the 5’ phosphate moiety, that forms the sugar phosphate

backbone, is replaced with sulphur. This confers resistance of the ASO to nuclease

degradation (Kenner et al., 2002, Eckstein, 1985). As described in section 3.4.4, a

phosphorothioate modified oligonucleotide had a half-life in cell culture media over 24h

whereas the unmodified counterpart (containing a phosphodiester backbone) was

degraded with a half-life of 3h (Fig. 3.10). Similarly, an unmodified oligonucleotide has

been reported to degrade in human serum within 30 mins (Akhtar, Kole & Juliano, 1991).

Although phosphorothioate linkages offer nuclease resistance, they can be

degraded over time at a much slower rate relative to phosphodiester linkages to release

monophosphate nucleotide analogues (Spitzer & Eckstein, 1988). The likelihood of

chemically modified oligonucleotide based therapeutics (e.g. ASO) to degrade and release

potentially genotoxic nucleotide analogues was a concern highlighted by the EMA

(European Medicines Agency, 2004); it was proposed that if nucleotide analogues could

enter endogenous nucleotide pools and become a substrate for DNA polymerase, base-

Page 150: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

150

pairing in newly synthesised DNA may occur with reduced fidelity increasing the probability

of mutation.

It may be that perturbation of intracellular dNTP pools by the presence of nucleotide

analogues may exert a mutagenic effect. Indeed, the balance of cellular dNTP pools is

known to be critical to maintain the fidelity of DNA replication and ensure the integrity of

the genome (Kunz, 1988, Meuth, 1984, Meuth, 1981b, Reichard, 1978). This pre-requisite

has been demonstrated in several studies, for example, supplementing mammalian cells

with 1mM or 3mM thymidine inhibited DNA synthesis by 90% (Bjursell & Reichard, 1973)

and induced GC→AT transition mutations in runs of guanine (Kresnak & Davidson, 1992),

respectively. Further to this, exogenous supply of 5µM deoxyadenosine was reported to

increase mutation at the genomic HPRT locus up to 59 fold in human CCRF-CEM

lymphoblast cells (Mattano, Palella & Mitchell, 1990).

Nucleoside analogues have also been adapted for use as viral therapeutics (Lee,

Hanes & Johnson, 2003, Ward & Duchin, 1997). In particular, AZT has been used in the

United States of America to decrease the rate of perinatal transmission of HIV-1 (Ward &

Duchin, 1997). However, AZT has been shown to be mutagenic, cause chromosome

aberrations and induce epithelial neoplasms in rats and mice (Sussman et al., 1999, Ayers

et al., 1996).

Nucleoside analogues used in viral therapy are not used in ASO therapeutic design.

Thus, it was important to inform the potential for nucleotide analogues used in

oligonucleotide based therapeutics to incorporate into newly synthesised DNA and cause

mutation.

Page 151: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

151

5.2. Experimental outline

The study presented here reports (i) the potential for nucleotide analogues, used in ASO

design, to incorporate into newly synthesised DNA and (ii) the genotoxicity of nucleotide

analogue incorporation in human lymphoblastoid TK6 cells.

In brief, the ability of Tfi DNA polymerase to incorporate various nucleotide

analogues into a primed DNA template, in vitro, was investigated using a method

extrapolated from Lacenere et al. (Lacenere et al., 2006). In this method, duplex DNA is

engineered to contain a 5’ fluorophore on the primer strand (25mer) whilst the template

strand has either one or five 5’ nucleotide overhang (Fig. 5.1). If DNA polymerase was

capable of extending the primer strand (5’ to 3’) it would be visible as a band of reduced

migration following electrophoresis through polyacrylamide gel. Under certain conditions,

Tfi DNA polymerase was capable of utilising nucleotide analogues, but could not fully

extend the primed template. In these instances, the reaction time was increased until the

template was fully extended. Comparing the time and dose required to fully extend the

primed template using nucleotide analogue with the time and dose required to extend the

primed template using the unmodified nucleotide counterpart informed the incorporation

efficiency of that particular nucleotide analogue.

Once a nucleotide analogue was shown to incorporate into a primed template, the

genotoxicity of this incorporation relative to the unmodified counterpart was assessed in

human TK6 cells using the HPRT and TK forward mutation assays. Clastogenic and

aneugenic activity following nucleotide analogue incorporation was assessed using the in

vitro micronucleus assay according to OECD guideline 487 (http://iccvam.niehs.nih.gov/

SuppDocs /FedDocs/OECD/OECD-TG487.pdf).

Page 152: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

152

Figure 5.1. Template used in the in vitro primer extension assay. Primer strand (25nt) was conjugated 5’ with 6FAM fluorophore for detection. Either, A1, A5 of C5 template strands were annealed to primer strand to produce a single or five 5’ nucleotide overhang. Primed template was incubated with nucleotide and Tfi DNA polymerase, denatured and electrophoresed. Extended primer strand (green arrow) appeared as a band of reduced migration relative to unextended primer strand if DNA polymerase could incorporate nucleotide. Oligonucleotide sequences used in the primer extension assay are listed in Table 2.5.

5.3. Methods

1. Primer extension assays are described in section 2.1.8.

2. The HPRT and TK forward mutation assay for nucleotide treatments are described

in sections 2.3.5 and 2.4.

3. The in vitro micronucleus assay is described in section 2.4.3.

4. RNA extraction is described in section 2.2.4.

5. PCR is described in section 2.5.

Primer strand

Template strand

Page 153: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

153

Figure 5.2. The chemical structure of adenine based nucleotides. Canonical dATP is shown in part A. A phosphorothioate nucleotide analogue (dATPαS) where a non-bridging oxygen in the α-phosphate group is replaced with sulphur is shown in B. A 2’O-methyl-ribose-ATP nucleotide analogue has an O-methyl group on the 2’ position of the ribose moiety of ATP is shown in C. The red circle depicts the position of the modification in each nucleotide analogue. Images were adapted from the manufacturer (Jena Bioscience, Germany).

5.4. Results

5.4.1. In vitro incorporation efficiency of nucleotide analogues

In the first instance, a primed template (Fig. 5.1) with a single thymidine nucleotide

overhang was used to measure incorporation of dATP up to 5µM (Fig. 5.2A). Single

nucleotide incorporation would allow determination of the Kd and maximum speed of

incorporation (Vmax) for each nucleotide analogue (Boosalis, Petruska & Goodman,

1987). However, gels could not clearly distinguish between unextended and extended

primed template using dATP, although it appeared to fully extend the primer strand around

0.5µM (Fig. 5.3). To clearly define incorporation of dATP, the overhang on the template

strand was extended to five thymidine nucleotides. Using this extended template, Tfi DNA

polymerase was clearly capable of fully extending the primer strand using 0.5µM dATP in

a 10 minute reaction (Fig. 5.4). Below 0.5µM, intermediate lengths of primer strand could

be seen. This concluded that an overhang of five nucleotides was appropriate for detecting

nucleotide incorporation and was used in subsequent experiments.

B

A

C

Page 154: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

154

Figure 5.3. Single nucleotide incorporation of dATP. Primer strand (25nt) and A1-template strand (26nt) were annealed together to produce a primed template with one 5’ thymidine nucleotide overhang (Table 2.5). The ability of Tfi DNA polymerase to extend the primer strand by one nucleotide can be observed as a band of reduced migration relative to unextended primer strand (0µM). The numbers above each lane are dose of nucleotide (μM). Samples were resolved through 15% urea-polyacrylamide gel. Tfi DNA polymerase appeared to fully extend primed template by one nucleotide using 0.5µM dATP in a 10 minute reaction. However, bands are not clearly resolved.

Figure 5.4. Incorporation of dATP using an extended template. Primer strand (25nt) and A5-template strand (30nt) were annealed together to produce a primed template with five 5’ thymidine nucleotide overhang (Table 2.5). The number above each lane is the concentration of nucleotide (μM). The ability of Tfi DNA polymerase to extend the primer strand by five nucleotides can be observed as a pronounced band of reduced migration relative to unextended primer strand (0µM). Tfi DNA polymerase clearly extended the primed template to completion using 0.5µM dATP in a 10 minute reaction.

To investigate the capability of Tfi DNA polymerase to incorporate nucleotide

analogues derived from ASO therapeutics, the above in vitro primer extension assay was

employed. Using a phosphorothioate nucleotide analogue (dATPαS) where a non-bridging

oxygen in the alpha phosphate moiety is replaced with sulphur (Fig. 5.2B); the most

common modification used in ASO therapeutics to increase nuclease resistance and

bioavailability (Geary, 2009, Buchini & Leumann, 2003), Tfi DNA polymerase was capable

of incorporating dATPαS to extend the primed template (Fig. 5.5A). Progressive bands of

reduced migration were evident with increasing dose of dATPαS and fully extended

Page 155: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

155

Figure 5.5. Incorporation kinetics of phosphorothioate nucleotide analogue dATPαS. Primer strand (25nt) and A5-template strand (30nt) were annealed together to produce a primed template with five 5’ thymidine nucleotide overhang (Table 2.5). The number above each lane is the concentration of nucleotide (μM). Primed template was incubated with dATPαS up to 5µM in a 10 minute (A) and 20 minute reaction (B). As a positive control in each reaction (lane C), 5µM dATP was used to depict a fully extended template. In a 10 minute reaction, Tfi DNA polymerase was capable of using dATPαS but could not extend the primed template to completion (A). Using a 20 minute reaction time, Tfi DNA polymerase fully extended primed template using 2µM dATPαS (B).

template can be seen from 1µM. However, Tfi DNA polymerase failed to completely

extend all the primed templates so the reaction time was increased to 20 mins using the

same concentrations of dATPαS. Using this extended reaction time, Tfi DNA polymerase

fully extended primed template using 2μM dATPαS (Fig. 5.5B). This utilisation of dATPαS

correlates to ~12% incorporation efficiency (8 fold less) compared to the unmodified

nucleotide counterpart dATP.

A common modification used to increase ASO target binding affinity is replacing the

2’hydroxyl group of the sugar moiety (within a ribonucleotide) with 2’O-methyl (Yoo et al.,

2004). A 2’O-methyl-adenosine triphosphate (2’OMe-ATP) nucleotide analogue was also

investigated to inform its potential to incorporate into a primed template (Fig. 5.2C).

B

A

Page 156: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

156

Figure 5.6. Incorporation kinetics of 2’O-methyl ribose modified nucleotide analogue 2’OMe-ATP. Primer strand (25nt) and A5-template strand (30nt) were annealed together to produce a primed template with five 5’ thymidine nucleotide overhang (Table 2.5). The number above each lane is the concentration of nucleotide (μM). Primed template was incubated with 2’OMe-ATP up to 5µM in a 10 minute (A) and 60 minute reaction (B). As a positive control in each reaction (lane C), 5µM dATP was used to depict a fully extended template. Tfi DNA polymerase failed to incorporate 2’OMe-ATP up to 5μM in a 10 minute (A) and 60 minute reaction (B) whereas the positive control worked suggesting a viable experiment.

Incubating Tfi DNA polymerase with 2’OMe-ATP up to 5µM in a 10 minute reaction failed

to extend the primed template so the reaction time was increased (Fig. 5.6A). Tfi DNA

polymerase failed to incorporate 2’OMe-ATP up to 5µM in an extended 60 minute reaction

(Fig. 5.6B). The positive control (dATP) in each experiment was fully incorporated

suggesting a viable experiment (Lane C, Fig. 5.6A and B). These data suggest that Tfi

DNA polymerase cannot use 2’OMe-ATP to extend a primed template in vitro and the

incorporation efficiency of 2’OMe-ATP relative to dATP is <<1.7%.

B

A

Page 157: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

157

Figure 5.7. The chemical structure of cytidine based nucleotides. Canonical dCTP is shown in part A. 5Me-dCTP is shown in part B. The red circle depicts the position of the modification in the nucleotide analogue. Images were taken from the nucleotide data sheets supplied by the manufacturer (Jena Bioscience, Germany).

An alternative modification used in pyrimidine motif based TFO and ASO

therapeutics is 5-methyl cytosine (Fig. 5.7; discussed in sections 1.2.1 and 1.3.1) which is

used to alleviate the pH dependence of triplex formation and avoid an immune response

with ASO therapeutics (Krieg, 1999, Xodo et al., 1991, Lee et al., 1984). The incorporation

efficiency of this nucleotide analogue and its unmodified counterpart was investigated

using a primed template as described above. In the first instance, the incorporation

efficiency of canonical dCTP was informed (Fig. 5.7A). Incubation of Tfi DNA polymerase

with dCTP in a 20 minute reaction fully extended the primed template using 5μM

nucleotide (Fig. 5.8A). Using 5-methyl dCTP (5Me-dCTP), Tfi DNA polymerase fully

extended primed template in a 40 minute reaction (Fig. 5.8B). The incorporation efficiency

of 5Me-dCTP is approximately 50% relative to dCTP.

B

A

Page 158: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

158

Figure 5.8. Incorporation kinetics of dCTP and 5Me-dCTP. Primer strand (25nt) and C5-template strand (30nt) were annealed together to produce a primed template with five 5’ guanine nucleotide overhang (Table 2.5). Primed template was incubated with dCTP up to 5µM in a 20 minute reaction (A) or with 5Me-dCTP in a 40 minute reaction (B). As a positive control in part B (lane C), 5µM dCTP was used to depict a fully extended template. Tfi DNA polymerase fully extended primed template using 5μM dCTP in a 20 minute reaction (A) whereas it took a 40 minute reaction using 5μM 5Me-dCTP (B).

5.4.2. Genotoxic fate of canonical nucleotides

Previous studies have reported that exogenous supply of canonical nucleotides can have

a genotoxic effect if perturbation of intracellular nucleotide pools occurs (Mattano, Palella

& Mitchell, 1990, Phear, Nalbantoglu & Meuth, 1987). If an ASO was not chemically

modified i.e. one that contained canonical bases, sugar moieties and a phosphodiester

backbone, it would be rapidly degraded by serum nucleases (Akhtar, Kole & Juliano,

1991); in section 3.4.4, it was shown that an unmodified 27mer oligonucleotide (PO-

TFO27) had a half-life of ~3h in cell culture media, whilst a 15mer was previously reported

to be completely degraded within 30 mins in human serum (Akhtar, Kole & Juliano, 1991).

Nuclease mediated degradation of an unmodified ASO therapeutic would release excess

conical nucleotides (Tanaka & Nyce, 2001) which may perturb endogenous nucleotide

B

A

Page 159: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

159

pools and cause mutation. In the study presented here, the genotoxic effect of

deoxyadenosine, deoxyadenosine monophosphate (dAMP) and dATP was investigated

using the established HPRT and TK forward mutation assay.

Human lymphoblastoid TK6 cells (4x106) were treated with nucleotide from 20µM

up to 1mM for 24h. Viability following treatment with all canonical nucleotides was not

below the accepted 20% RTG for this assay (Fig. 5.9A). Treatment with deoxyadenosine

and dAMP resulted in dose dependent cytotoxicity up to 40% cell death (Fig. 5.9A).

However, dATP failed to induce any cell death following treatment (Fig. 5.9A).

Treatment of TK6 cells with deoxyadenosine induced dose dependent mutation at

the HPRT and TK loci up to 3 and 1.8 fold above control (0mM), respectively, with an

increase in the proportion of small TK mutant clones from 14% in control up to 26% with

1mM treatment (Fig. 5.9B-D). An increase in small TK mutant colonies may suggest large

inter-gene deletions or chromosome aberrations involving the TK gene in these clones

which may affect the rate of growth due to the extent of genomic insult or perhaps a result

of mutation within an adjacent growth regulatory gene (Liechty et al., 1998, Applegate et

al., 1990, Moore et al., 1985, Hozier et al., 1981); although such a gene has never been

identified (Liechty et al., 1998).

Interestingly, treating TK6 cells with dAMP resulted in a dose dependent increase in

TK mutants up to 1.7 fold above control (Fig. 5.9B). In this case also, an increase in the

proportion of small clones was evident (Fig. 5.9C). However, dAMP failed to induce HPRT

mutants above control (Fig. 5.9D). This may suggest the TK locus was more sensitive in

detecting mutation induced by dAMP or that dAMP mediated mutation within the HPRT

locus was lethal to the cell or did not result in a 6-TG resistant phenotype. Certain types of

mutation within the HPRT locus are known to be lethal to a cell; for example inter-gene

deletions, whilst the TK locus is a more robust reporter capable of detecting point

mutations, deletions, chromosome aberrations and even recombination events (Doak et

Page 160: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

160

al., 2007, Liechty et al., 1998, McGregor et al., 1996, Applegate et al., 1990, Yandell, Dryja

& Little, 1986).

Most peculiar was the treatment of TK6 cells with dATP up to 1mM did not induce

TK mutants (Fig. 5.9B) but induced dose dependent mutation at the HPRT locus up to 2.7

fold (Fig. 5.9D). However, the increase in mutant frequency was not statistically significant

(p>0.05). Perhaps dATP mediated mutation at the TK locus did not result in a TFT

resistant phenotype.

Page 161: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

161

Figure 5.9. Genotoxicity of canonical nucleotides. TK6 cells (4x106) were treated with nucleotide up to 1mM for 24h. Cytotoxicity following treatment with nucleotides was not below the accepted 20% RTG for this assay (A). Deoxyadenosine and dAMP induced dose dependent mutation at the TK locus up to 1.8 fold whilst treatment with dATP did not (B). Deoxyadenosine and dAMP treatment resulted in an increase in the proportion of small TK mutant clones (C). Deoxyadenosine and dATP treatment resulted in dose dependent mutation at the HPRT locus up to 3 fold and 2.7 fold, respectively, whilst treatment with dAMP failed to induce mutation at the HPRT locus (D). Data represent mean ± SD of three independent treatments. Data is compared to negative control (0mM) for each nucleotide using one-way analysis of variance (ANOVA) with Dunnett’s post-hoc test. * p<0.05.

B A

C D

Page 162: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

162

5.4.3. Genotoxic fate of a phosphorothioate nucleotide analogue

Having shown a phosphorothioate nucleotide analogue could be used by Tfi DNA

polymerase to extend a primed template in vitro (section 5.4.1); the potential for utilisation

in vivo was investigated using human TK6 cells. Mutation caused by nucleotide analogue

utilisation/ incorporation into genomic DNA was informed using the HPRT and TK forward

mutation assay whilst aneugenic/ clastogenic activity was informed using the micronucleus

assay conducted to OECD guidelines (OECD guideline 487; http://iccvam.niehs.nih.gov/

SuppDocs /FedDocs/OECD/OECD-TG487.pdf).

Of important consideration was that degradation of an oligonucleotide would not

yield nucleotides as a triphosphate but rather as monophosphates. Thus, to reflect a true

biologically relevant event, taking into account the prerequisite for phosphorylation of

monophosphate nucleotides into triphosphates before utilisation by DNA polymerase,

human TK6 cells were treated with monophosphate phosphorothioate nucleotide dAMPαS.

Human lymphoblastoid TK6 cells (4x106) were treated with dAMPαS up to 1mM for

24h. As a nucleotide control, TK6 cells were also treated with 1mM of the unmodified

nucleotide counterpart, dAMP. This would inform the genotoxicity caused by the single

substitution of oxygen for sulphur in the 5’ phosphate group in dAMPαS (Fig. 5.2B).

Treating TK6 cells with dAMPαS up to 0.5mM was not cytotoxic above the

maximum recommended 80% for this assay (30% RTG; Fig. 5.10A). However, 1mM

dAMPαS was significantly cytotoxic (10% RTG) which prompted caution when interpreting

genotoxicity data. Interestingly, the magnitude of cytotoxicity following 1mM dAMP

treatment (54% RTG) was much less than treatment with 1mM dAMPαS suggesting the

nucleotide analogue was more cytotoxic (Fig. 5.10A).

Phenomenally, treating TK6 cells with 0.5mM and 1mM dAMPαS resulted in TK

mutant frequencies of 317x10-6 and 1540x10-6, which is equivalent to an increase of ~20

fold and ~96 fold above control, respectively (Fig. 5.10A). Noteworthy was that treatment

Page 163: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

163

with 1mM dAMPαS induced TK mutant clones in every well of three 96 well microtitre

plates for each independent treatment at that dose. To allow calculation of mutant

frequency, one empty well was assumed from each treatment. This infers that the

frequency of mutation at the TK locus was a minimum of 96 fold greater than control with

1mM dAMPαS treatment.

To support mutation at the TK locus following 0.5mM dAMPαS treatment, randomly

selected clones were expanded in TFT media for 7-10 days. All clones tested grew

successfully informing mutation at the TK locus.

dAMPαS treatment did not increase the proportion of small TK mutant clones (Fig.

5.10C). In fact, treatment with 1mM dAMPαS produced entirely large TK colonies.

Interestingly, 1mM dAMP treatment only induced the TK mutant frequency ~1.7 fold above

control (Fig. 5.10B), similar to the initial observation (Fig. 5.9B). This correlates the

genotoxicity of 1mM dAMPαS to be at least 56 fold more potent than the unmodified

counterpart. Most interesting was that dAMP and dAMPαS treatment, even up to 1mM,

failed to induce mutation at the HPRT locus (Fig. 5.10D). This infers a cytotoxicity based

mechanism of mutation at the TK locus could be excluded with 1mM dAMPαS treatment.

Page 164: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

164

Figure 5.10. Genotoxicity of a non-canonical phosphorothioate nucleotide analogue. TK6 cells (4x106) were treated with dAMPαS up to 1mM for 24h. As a negative nucleotide control, TK6 cells were also treated with 1mM of the unmodified counterpart dAMP. EMS (5µg/ml) was used as a positive genotoxic control. Treatment with dAMPαS resulted in dose dependent cytotoxicity up to 10% RTG (A). Treatment with dAMP was not cytotoxic (A). Phenomenally, treatment with 0.5mM and 1mM dAMPαS resulted in significant mutation above control at the TK locus ~20 fold and ~96 fold, respectively (B). dAMP treatment induced the TK mutant frequency ~1.7 fold (B). There was no shift in the proportion of small TK mutant clones following dAMPαS treatment (C). All nucleotide treatments failed to induce mutation at the HPRT locus (D). Data represent mean ± SD of three independent treatments. Data is compared to negative control (0mM) using one-way ANOVA with Dunnett’s post-hoc test. * p<0.05; **p<0.01;***p<0.001. EMS and 1mM dAMPαS are excluded from statistical analysis.

0 0.02 0.05 0.1 0.2 0.5 1.0 dAMP EMS

0

10

20

30

40

50

60

70

80

90

100

110

120

**

***

***

***

Dose (mM)

Rela

tive T

ota

l G

row

th (

%)

0 0.02 0.05 0.1 0.2 0.5 1.0 dAMP EMS

0

50

100

150

200

250

300

350

1400

1500

1600

1700

1800

***

Dose (mM)

Mu

tan

t fr

eq

ue

ncy

x10

-6 v

iab

le c

ells

B A

C D

Page 165: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

165

Following 48h of treatment with nucleotide, TK6 cells were harvested to inform the

frequency of micronuclei as a marker of clastogenic/ aneugenic activity. Surprisingly,

treatment with either dAMPαS up to 1mM or 1mM dAMP failed to induce the frequency of

micronuclei in mononucleate cells despite significant mutation at the TK locus (Fig. 5.11A).

The positive control, 5µg/mL EMS, induced the frequency of micronuclei in mononucleate

cells over 2 fold suggesting a viable experiment (Fig. 5.11A). Cytotoxicity in the

micronucleus assay was not evident following nucleotide treatment of TK6, assessed

using the RPD according to OECD guideline 487 (Fig. 5.11B)

(http://iccvam.niehs.nih.gov/SuppDocs /FedDocs/OECD/OECD-TG487.pdf). The RPD is

important to ascertain whether cells have undergone mitosis to facilitate expression of

micronuclei. Cells which fail to undergo mitosis do not express micronuclei and can be a

factor for false negative results. According to OECD guideline 487, cytotoxicity should not

be greater than 55% to avoid false positive results which is in concert with the results

obtained here (Fig. 5.11B) (http://iccvam.niehs.nih.gov/SuppDocs /FedDocs/OECD/OECD-

TG487.pdf).

Interestingly, treatment with 1mM dAMP and 0.2mM, 0.5mM and 1mM dAMPαS

produced a dose dependent increase in the frequency of binucleate cells and

micronucleated binucleate cells (Fig. 5.11C and D, respectively). The frequency of

binucleate cells with 1mM dAMP treatment was almost twice that observed with 1mM

dAMPαS treatment (Fig. 5.11C).

Page 166: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

166

Figure 5.11. Clastogenic/ aneugenic activity of dAMPαS in human TK6 cells. Human TK6 cells (4x106) were treated with 1mM dAMP and dAMPαS up to 1mM for 24h. After 48h from treatment, TK6 cells were sampled to score the incidence of micronuclei in mononucleate cells (A), cytotoxicity following treatment measured as the relative population doubling (B), binucleate cells (C) and binucleate cells containing micronuclei (D). Treatment with nucleotides failed to induce micronuclei in 2000 mononucleate cells scored per treatment (A) and cytotoxicity was no greater than the recommended 55% for this assay (B). Treatment with nucleotide caused a dose dependent increase in binucleate cells and micronucleated binucleate cells (C and D, respectively). Data represent mean ± SD of three independent treatments. Data is compared to negative control (0mM) using one-way ANOVA with Dunnett’s post-hoc test. * p<0.05; **p<0.01;***p<0.001. EMS is excluded from statistical analysis.

0 0.2 0.5 1.0 dAMP EMS

0

10

20

30

40

50

60

70

80

90

100

110

120

**

***

Dose (mM)

Bin

ucle

ate

cells/2

000 m

on

on

ucle

ate

cells

0 0.2 0.5 1.0 dAMP EMS

0

5

10

15

20

25

30

35

40

Dose (mM)

MN

/2000 m

on

on

ucle

ate

cells

0 0.2 0.5 1.0 dAMP EMS

0

2

4

6

8

10

12

14

16

18

20

22

24

Dose (mM)

MN

bin

ucle

ate

/2000 m

on

on

ucle

ate

cells

0 0.02 0.05 0.1 0.2 0.5 1 dAMP EMS

0

10

20

30

40

50

60

70

80

90

100

110

Dose (mM)

Rela

tive p

op

ula

tio

n d

ou

blin

g

B A

C D

Page 167: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

167

Figure 5.12. PCR amplification of cDNA from isolated dAMPαS induced TK mutants. Control (0mM) and 0.5mM dAMPαS TK mutant clones were isolated and expanded for total RNA extraction. RNA extracts were reverse transcribed and used to PCR amplify 837bp of the TK mRNA using primer pair 5 (Table 2.4). To inform possible RNA contamination during reverse transcription, RNA was omitted from a mixture including the reverse transcriptase enzyme (RT+). To inform the potential for genomic DNA contamination and PCR amplification using primer pair 5, reverse transcriptase was omitted from a reaction mixture containing RNA (RT-). To inform the potential of cDNA contamination and amplification during PCR reactions, cDNA was replaced with water (W). Only 4 out of 6 control TK mutants successfully PCR (A). From 15 dAMPαS induced TK mutants only 2 clones efficiently PCR to yield products capable of sequencing (B).

To inform the mechanism of dAMPαS mediated mutation, TK mutant clones were

expanded for RNA extraction. Following reverse transcription of RNA into cDNA, extracts

were subject to PCR across the TK mRNA (837bp). PCR primers (Table 2.4) were

designed to include the entire coding region of the TK gene. Following successful PCR,

products would be sent for DNA sequencing. However, following PCR of 6 control (0mM)

TK mutant clones, only 4 successfully PCR amplified (Fig. 5.12A). Nevertheless, 15 TK

clones from 0.5mM dAMPαS treatment were also amplified. Although almost all clones

produced a visible band ~850bp only 2 PCR reactions efficiently permitted DNA

sequencing (Fig. 5.12B). This posed a problem for molecular analysis as an accurate

mechanism of action could not be elucidated from the sequence of 2 mutant clones. Due

to time restraints, this work could not be pursued any further and so DNA sequence

analysis of clones could not be completed.

B

A

Page 168: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

168

5.5. Discussion

In a reflection paper by the EMA, concern was raised regarding the fate of degradation

products from chemically modified oligonucleotide based pharmaceuticals (European

Medicines Agency, 2004). It was proposed that nucleotide analogues released from

degradation of ASO therapeutics may enter endogenous nucleotide pools and become

substrates for DNA polymerase. If base-pairing with nucleotide analogues in newly

synthesised DNA occurred with reduced fidelity, mutation would be likely. Indeed, the

genotoxicity associated with non-canonical nucleotides used in treatment of HIV has been

well documented and supports the concern presented by the EMA (Lee, Hanes &

Johnson, 2003, Sussman et al., 1999, Ward & Duchin, 1997, Ayers et al., 1996). The

objective of this study was to inform (i) the capability of putative nucleotide analogues,

released from ASO degradation, to be utilised by DNA polymerase to extend a DNA

template and (ii) the genotoxicity associated with this incorporation in vivo.

Data presented here suggest that Tfi DNA polymerase can utilise some of the

nucleotide analogues commonly found in ASO therapeutics. Tfi DNA polymerase was

capable of incorporating dATPαS and 5Me-dCTP into a primed DNA template, albeit with

reduced affinity (8 and 2 fold compared to the unmodified nucleotide control, respectively).

However, Tfi DNA polymerase failed to use 2’OMe-ATP even in extended reactions. It may

be plausible to suggest that nucleotide analogue incorporation entirely depends on the

nature of the chemical modification.

Although 5-methyl cytosine is not a non-canonical base per se (cytosine can be

methylated in vivo on the 5’ position to epigenetically alter gene expression) unexpected

incorporation of 5Me-dCTP released from oligonucleotide degradation, may change the

methylation profile of highly regulated genes and may be deleterious to the cell (Esteller,

2008, Weber et al., 2007, Herman & Baylin, 2003, Greger et al., 1989).

Page 169: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

169

To inform genotoxicity following nucleotide incorporation into genomic DNA in vivo,

TK6 cells were treated for 24h. In the first instance, cells were treated with canonical

adenine based nucleotides. Here, genotoxicity was no greater than 3 fold at either the TK

or HPRT loci. Treating TK6 cells with 0.5mM and 1mM phosphorothioate nucleotide

analogue dAMPαS, derived from the most common chemical modification used in ASO

therapeutics (Geary, 2009, Buchini & Leumann, 2003, Pirollo et al., 2003), was

phenomenally genotoxic (>96 fold) at the genomic TK locus. Surprisingly, mutation at the

HPRT locus was not observed. This phenomenon can be explained from previous studies

suggesting a large proportion of mutations at the HPRT locus can be deleterious to the cell

whilst the TK locus is a more robust reporter capable of detecting point mutations, large

inter-gene deletions, chromosome aberrations and even recombination events (Doak et

al., 2007, Liechty et al., 1998, McGregor et al., 1996, Applegate et al., 1990, Yandell, Dryja

& Little, 1986). Perhaps the extent of genomic insult at the HPRT locus following

nucleotide analogue treatment was lethal to the cell, or, if phosphorothioate analogue

mutation was the result of a delayed mechanism, then perhaps there was not sufficient

phenotypic expression of HPRT mutants. Premature selection in 6-TG media would result

in lethality to cells that contained residual wild type HPRT activity. For example,

phenotypic expression of TK mutants is recommended to be 1-2 days whereas HPRT is 6-

7 days. In this study, TK and HPRT mutants are selected on Day 3 and 7, respectively. If

mutation by dAMPαS was 48h from treatment, this would still provide TK6 cells sufficient

phenotypic expression of TK mutants but not HPRT. To confirm this speculation, the

genotoxicity experiments could be repeated with a longer phenotypic expression period for

HPRT. However, one would still expect some recovery of HPRT mutants during selection,

even those prematurely selected by 1 day, especially when mutation at the TK locus is

~100 fold above control.

Page 170: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

170

Although TK mutant clones were not sequenced, having considered the threshold

effect observed at the TK locus, we suggest the mechanism of mutation by dAMPαS

treatment may be in accordance with the next nucleotide effect model (Phear, Nalbantoglu

& Meuth, 1987). The speed at which DNA polymerase extends an elongating strand (5’ to

3’) is governed by the availability of the next (3’) nucleotide in sequence. Thus, if a

misinserted nucleotide is followed 3’ by a highly abundant nucleotide, polymerisation of

this next nucleotide is favoured rather than the excision of the incorrect one by the 3’ to 5’

exonuclease activity associated to DNA polymerase (Fersht, 1979). However, in this case,

the insertion of the phosphorothioate analogue would render the bond nuclease resistant

sealing the error. This mechanism of mutation would also explain why micronuclei (as a

marker of DNA double strand breaks) were not observed following nucleotide treatment

despite significant mutation at the TK locus.

Further to this is that dATPαS incorporation into newly synthesised genomic DNA

may still occur below 0.5mM without mutation, as the critical balance of nucleotide pools

dictates the fidelity of DNA polymerase and concentrations of dAMPαS ≥ 0.5mM may

perturb these pools such that mutation results from misincorporation of canonical

nucleotides by DNA polymerase which becomes sealed by dATPαS. It is also noteworthy

that the human TK6 cells were treated with the monophosphate analogue (dAMPαS) and

so for incorporation into genomic DNA, not only must the concentration of dATPαS exceed

beyond a threshold for insertion of analogue rather than canonical dATP, but importantly,

dAMPαS must first become a substrate for various kinases (possibly Adenylate Kinase

(EC 2.7.4.3) and nucleoside-diphosphate kinase (EC 2.7.4.6) (Stadtman & et al., 1973)) to

convert the monophosphate into a triphosphate for utilisation by DNA polymerase. In

essence, these limitations would contribute to a genotoxic threshold as observed in the

data presented here.

Page 171: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

171

One cannot dismiss the possibility of an epigenetic mechanism of mutation by

dAMPαS. It is reported that AZT can epigenetically silence TK gene expression by

inducing promoter hypermethylation (Hoever et al., 2003, Wu et al., 1995, Nyce et al.,

1993). This is thought to be the cause of AZT drug resistance. DNA hypermethylation is

thought to be one of the cellular responses to drugs that inhibit DNA synthesis (Nyce et al.,

1989, Nyce, 1989). This hypermethylation silences expression of genes that may be

involved in activation/phosphorylation of the drug. Thus, TK gene silencing increases

resistance to AZT treatment because AZT is not monophosohorylated by TK. Similarly,

dAMPαS may be inducing global hypermethylation similar to that reported following AZT

treatment (Nyce et al. 1993). Furthermore, once dAMPαS is phosphorylated to dATPαS, it

may become oxidised to ATPαS which can then be utilised for S-adenosylmethionine

biosynthesis, which is the methyl donor for methylation of 5’-CpG dinucleotide motifs by

DNA methyltransferase (Mato et al., 1997). If this confers increased 5’-CpG methylation

potential by DNA methyltransferase this would also explain an epigenetic mechanism of

gene silencing.

Most surprising was that dAMPαS treatment was not clastogenic in the in vitro

micronucleus assay despite the mutagenic response observed at the TK locus. However,

this would support the above hypothesis of a mechanism of mutation through perturbation

of intracellular nucleotides pools resulting in excess nucleotide misincorporation and

subsequent point mutation or epigenetic silencing of TK gene expression. Despite this, a

dose dependent increase in binucleate cells with and without micronuclei was observed

following nucleotide treatments. It seems likely that the presence of excess nucleotide

analogue results in perturbation of intracellular nucleotide pools. In an E. Coli system,

treatment of cells with adenine caused a two fold increase in ATP levels with a three to

four fold decrease in GTP levels (Levine & Taylor, 1982). In a lymphoblastoid cell line,

treating cells with 20µM deoxyadenosine increased the dATP/dGTP ratio almost 28 fold

Page 172: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

172

(Mattano, Palella & Mitchell, 1990). It is possible that perturbation of nucleotide pools and

subsequent reduction in GTP levels following dAMPαS treatment affects the

efficiency/kinetics of cytokinesis which is a GTP dependent process (reviewed by Lacroix

and Maddox (Lacroix & Maddox, 2011)).

With increasing focus to produce more efficient delivery systems and chemical

modifications for oligonucleotide based pharmaceuticals to enhance in vivo bioavailability/

function (Morvan et al., 1993, Akhtar, Kole & Juliano, 1991, Hoke et al., 1991), the risk of

intracellular accumulation of ASO degradation products may also become a factor that

needs considering. Data presented here suggest that some ASO degradation products

can be used by DNA polymerase and, if the concentration of nucleotide analogue is

sufficient, mutation at genomic DNA can occur.

Page 173: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

173

Chapter 6

General discussion and conclusions

ASO therapeutics are powerful therapeutic tools theoretically capable of modulating

protein expression of any gene (Davidson & McCray, 2011, Pirollo et al., 2003, Agrawal,

1999). Currently, there are several ASO in clinical trials for treatment of various diseases,

predominantly cancer (Cunningham et al., 2001, Chen et al., 2000, Cunningham et al.,

2000). The current genotoxicity test battery is deemed inappropriate for safety assessment

of biotechnology derived pharmaceuticals, which includes oligonucleotide based

therapeutics, according to the ICH S6 guidelines

(www.ich.org/fileadmin/Public_Web_Site/ICH_Products/Guidelines/Safety/S6_R1/Step4/S

6_R1_Guideline.pdf). Previous studies have presented data demonstrating oligonucleotide

mediated mutagenesis through triplex and D-loop formation (sections 1.3.1.1 and 1.3.2.1).

These studies have raised concern from the EMA regarding the capability of

oligonucleotide based therapeutics, such as ASO, to form such structures at genomic DNA

and cause mutation (European Medicines Agency, 2004). However, an important point to

consider is that previous studies have employed plasmid based reporter systems to

evaluate genotoxicity of oligonucleotides and do not take into account ASO design (length

and chemical modification). Of further consideration when using engineered reporter

systems is the number of copies of plasmid/ vector in each cell; those with more copies

are more likely to undergo oligonucleotide mediated mutation due to the increased number

of available target sites and, thus, does not necessarily reflect mutagenesis at a genomic

target where a single target sequence is likely to exist within one or two alleles complexed

with histone proteins.

Additionally, the EMA questioned the fate of degradative products (nucleotide

analogues) from chemically modified ASO therapeutics as chromosomal damage following

Page 174: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

174

antiretroviral nucleoside analogue incorporation into genomic DNA has been previously

established (Carter et al., 2007, Meng et al., 2002, Wutzler & Thust, 2001, Thust et al.,

2000a, Thust et al., 2000b, Sussman et al., 1999, Thust, Schacke & Wutzler, 1996).

Importantly, antiretroviral nucleoside analogues are not used in ASO design and so the

genotoxicity associated with antiviral nucleoside analogue therapy cannot be extrapolated

to putative ASO degradation products.

This study has addressed these concerns by testing the genotoxic potential of

oligonucleotides designed to form triplex and D-loop structures as well as assessing the

mutagenic potential of a phosphorothioate nucleotide analogue at genomic DNA.

Data presented in this study shows a TFO targeting genomic DNA could not induce

mutation above the detection limit of the assay despite high affinity binding to the target

motif (chapter 3). It is likely that the complexity of the chromatin structure hinders

accessibility of TFO27 to its binding site within the HPRT locus or that the triplex structure

is unwound by a progressing replication/transcription fork. This study has also shown that

previous reports of triplex mediated mutagenesis using the supF based reporter system

should be interpreted with caution as the target sequence is also capable of DNA

quadruplex formation in the presence of targeting oligonucleotide (Kalish et al., 2005,

Datta et al., 2001, Vasquez et al., 2001, Faruqi et al., 2000, Vasquez et al., 1999, Wang,

Seidman & Glazer, 1996). Indeed, DNA quadruplex structures are known to be sources of

genomic instability and it may have been DNA quadruplex and not triplex formation that

contributed to mutagenesis in supF based systems previously employed (Broxson, Beckett

& Tornaletti, 2011, Basundra et al., 2010).

This study has also shown a biologically active ASO molecule to be capable of

inducing sequence specific mutation at genomic DNA by forming a D-loop structure

(chapter 4). D-loop formation is more likely to be a concern/ consideration for future

oligonucleotide pharmaceutical design as triplex formation is dictated by divalent cations

Page 175: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

175

and more importantly a purine stretch (Duca et al., 2008, Kalish et al., 2005, Hoogsteen,

1959, Felsenfeld, Davies & Rich, 1957) whereas D-loop formation is not governed by such

stringent conditions. Treating TK6 cells with AD3-hprtPM, in a system where RAD51

protein expression was induced, led to mutation downstream of the target sequence,

contrary to the expected mechanism of TNE where mutation was predicted to be at the

site of the mismatched base. This observation may be particularly important as a

mismatched base may not be a pre-requisite for mutation following D-loop formation by

oligonucleotides. Thus, an ASO molecule perfectly complimentary to a genomic sequence

may also result in mutation; however, this will need to be experimentally confirmed.

An important limitation to the data set is that genotoxicity of a single TFO and ASO

molecule has been investigated. This presents with several questions: (1) Can a TFO

targeting a different genomic sequence induce mutation? (2) Can ASO molecules

homologous to difference coding regions of genomic DNA induce mutation similar to that

observed at the HPRT locus and does mutagenicity depend on RAD51 protein

expression? These limitations were acknowledged during the early stages of the study and

an attempt was made to resolve them. An effort was made to design a TFO to target the

TK and other regions of the HPRT locus but as triplex formation is limited to purine

stretches, a TFO could not successfully be engineered. Similarly, two other ASO

molecules containing homology to a protein target and a selectable locus such as HPRT

and TK were designed; both ASO were tested in a preliminary mutation study with

negative results but upon ASO sequence examination, both were found to be capable of

forming stable hair-pin structures rendering the oligonucleotides biologically inactive for

target binding and so abandoned for further testing.

Despite these limitations, these data still suggest an ASO molecule may present

mutation potential, providing RAD51 protein expression is sufficient to mediate strand

invasion. Although ASO mediated mutagenesis appeared to depend on RAD51 protein

Page 176: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

176

induction, the possibility of mutagenesis occurring below the detection limit of the assay

cannot be excluded in the absence of RAD51 induction.

The dependence of AD3-hprtPM mediated mutagenesis on RAD51 protein

induction is clinically relevant to patients with p53 mutant tumours that are often found to

have elevated RAD51 protein expression; it has been proposed that RAD51

overexpression may contribute to drug resistance and genomic instability (Klein, 2008).

Furthermore, the majority of ASO currently in clinical trials are for cancer therapy and

reportedly p53 is inactivated in half of human cancers (Soussi & Lozano, 2005). Since p53

negatively regulates RAD51 gene expression these patients may also present with

elevated RAD51 protein expression (Hannay et al., 2007, Arias-Lopez et al., 2006). For

example, the extent of RAD51 protein expression in invasive ductal breast cancer

correlated with the histological grading of tumours and RAD51 is reportedly induced 2-7

fold in several cancer cell lines derived from prostate cancer, colon cancer and breast

cancer, similar to that reported here (Raderschall et al., 2002, Maacke et al., 2000).

However, how these in vitro genotoxicity data translate to an in vivo system is unknown

and warrants further investigation.

Noteworthy is that in this study, human TK6 cells were transfected with

oligonucleotide for 4h and then removed from treatment. It seems plausible to suggest that

exposure of cells to oligonucleotide for longer periods of time may increase the probability

for ASO hybridisation to genomic DNA and mutation. For example, in a Phase I/II clinical

trial using a 2’OMe/phosphorothioate ASO (PRO051, Prosensa Therapetucis), plasma

half-life of oligonucleotide was between 19 and 56 days (Goemans et al., 2011).

Additionally, subcutaneous injection of a phosphorothioate ASO has been shown to rapidly

distribute to the liver in mice and remain there with a half-life up to 19 days (Yu et al.,

2001). In fact, ASO elimination from the liver was reduced with increasing ASO dose. ASO

distribution is not found to be isolated to the liver; administration of a single dose of a

Page 177: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

177

phosphorothioate ASO (30mg/kg) has also been shown to accumulate ASO in the spleen

(8µg/g of tissue), heart (4µg/g of tissue), small and large intestines (~10µg/g of tissue),

lung (1µg/g of tissue) and testes (7µg/g of tissue) (Agrawal, Temsamani & Tang, 1991).

Analysis of extracted tissues informed the concentration of oligonucleotide remained

consistent in the tissues listed above from 1h to 48h. The concentrations of oligonucleotide

found in these tissues are a fraction of those used in the study presented here, but daily

treatment of mice (intraperitoneal injection) for 21 days with 20mg/kg ISIS 3521 reached

intra-tumoural concentrations of 2µM which are very similar to the concentrations used

here (Yazaki et al., 1996). Thus, accumulation and constant exposure of cells to ASO may

increase the probability of ASO hybridisation to genomic DNA and subsequent mutation;

repeated cycles of ASO binding may further increase the likelihood of mutation.

This hypothesis is supported from a study by Leonetti et al. (Leonetti et al., 1991)

and Chin et al. (Chin et al., 1990) where micro-injected oligonucleotides were shown to

rapidly accumulate in the cell nucleus and not the cytoplasm. Thus, ASO binding to

genomic DNA is even more likely if ASO reside in the cell nucleus.

The bioavailability of ASO for gene silencing is a major limitation to their use as

therapeutic tools, to address this, focus has been drawn to optimise biological efficacy

through effective design and efficient delivery of ASO to target tissue (Dias & Stein, 2002).

However, as discussed above, increasing intracellular concentrations of ASO through

efficient delivery methods may further facilitate intracellular accumulation of ASO

increasing the likelihood of an ASO molecule binding to a complimentary sequence in

genomic DNA.

It seems premature to suggest novel safety assessment techniques are required for

ASO therapeutics, especially when the genotoxicity data presented here is based on

mutagenesis by a single ASO. However, the data cannot be entirely dismissed as the

possibility for heritable sequence alterations is now evident. If novel pre-clinical safety

Page 178: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

178

assessment techniques were to be developed to screen sequence specific off-target

mutagenesis by oligonucleotide pharmaceuticals, it is most likely that a fluorescent

activated cell sorting (FACS) based method will be employed. Sequence specific mutation

by oligonucleotides at genomic DNA would render gene products non-functional and

mutant cells could be detected/ isolated as those without a fluorescent signal. DNA

sequencing of mutant clones could be used to confirm sequence specific mutation.

Using new chemical modifications for ASO design may increase biological efficacy,

but may also present with new concerns; ASO degradation, mainly from the 3’ end (Shaw

et al., 1991, Tidd & Warenius, 1989, Wickstrom, 1986), may release monophosphate

nucleotide analogues which may be substrates for DNA polymerase and incorporate into

newly synthesised genomic DNA. Degradation of unmodified phosphodiester

oligonucleotides has previously been shown to release monophosphate nucleotides

(Vaerman et al., 1997). In this study, data is presented illustrating the use of

phosphorothioate and 5-methyl-cytosine modified nucleotide analogues by DNA

polymerase, albeit with reduced efficiency (chapter 5). However, DNA polymerase could

not use a 2’O-methyl-ribose modified nucleotide most likely because it resembles an RNA

molecule and not DNA. Treatment of TK6 cells with 1mM phosphorothioate nucleotide

analogue resulted in mutation ~100 fold above control. A limitation that should be

acknowledged is that the genotoxicity of an adenine based phosphorothioate nucleotide

was investigated without evaluating potential differences in genotoxicity for those

containing cytosine, guanine or thymidine as the nucleobase. Although the concentration

of dAMPαS found to be genotoxic (≥0.5mM) is greater than the physiological dNTP

concentration in mammalian cells (24µM dATP; 5.2µM dGTP; 29µM dCTP; 37µM dTTP

according to Traut (Traut, 1994)), intracellular accumulation of ASO is also likely to be

paralleled to the level of nucleotide analogues available for entry into endogenous dNTP

Page 179: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

179

pools. Similar to ASO described above, prolonged exposure to nucleotide analogues may

increase the risk of utilisation/ incorporation and mutation.

Although the genotoxicity of 5-Me-dCTP was not investigated in this study DNA

polymerase was shown to be capable of incorporating 5-Me-dCTP into a primed DNA

template. Methylation of cytosine at CpG dinucleotide motifs in genomic DNA is a method

of epigenetically altering gene expression and so 5-Me-dCTP may not necessarily be

considered a non-canonical nucleotide (Esteller, 2008, Weber et al., 2007, Herman &

Baylin, 2003, Greger et al., 1989). ASO such as Vitravene, G3139 and ISIS 3521 are

commonly modified to contain 5-methyl-cytosine residues at CpG dinucleotide motifs to

prevent immune stimulation, as described in section 1.2.1 (Agrawal & Kandimalla, 2004,

Krieg et al., 1995). If these chemically modified ASO are degraded by endogenous

nucleases they are likely to release 5-methyl-cytosine monophosphate nucleotides.

Incorporation of 5-methyl-cytosine into genomic DNA, thus, has the potential to alter the

epigenome and this may lead to altered gene expression and changes to certain

biochemical processes such as cell proliferation.

The data generated in this study shows that an ASO was capable of off-target

genotoxicity by directly interacting with genomic DNA or through utilisation of it’s

degradative product during DNA synthesis. The concentration of oligonucleotide coupled

to the conditions used during treatments (i.e. MMS pre-treatment to induce RAD51 protein

expression) that elicited a genotoxic response suggest ASO entities are not likely to result

in genomic insult at physiological concentrations that are employed for therapeutic

application. However, the data informs the potential for mutation providing favourable

conditions are met. It is likely that further testing of ASO and putative degradation products

is required to draw final conclusions.

Page 180: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

180

6.1. Future work

To bolster the data presented in this study and the mechanism of mutation by ASO and

putative degradation products, several experiments can be suggested:

The mutagenicity data for triplex formation that has been presented in this study is

based on a single TFO targeting a selectable genomic locus due to the limitations for

triplex formation, as discussed in section 1.3. However, to confirm the results for TFO27

targeting the HPRT locus, the genotoxicity of TFO targeting distinct gene loci can also be

investigated using a FACS based method that can be optimised for each target by

monitoring a decrease in cellular fluorescence.

Furthermore, developing the method outlined in section 3.4.6. to detect and quantify

triplex formation at genomic DNA will allow a correlation between the binding capacity and

mutagenicity of TFO.

Data presented in this study also implicates the SupF gene target sequence as a

site for DNA quadruplex formation and triplex formation in the presence of targeting

oligonucleotide. Mutation of key guanine residues in the engineered triplex target

sequence of the SupF gene was shown to reduce DNA quadruplex formation (chapter 3).

By employing this engineered system as a reporter for mutation, DNA quadruplex and not

triplex formation may be implicated in the mutagenicity of oligonucleotides, that contain a

high guanine content, previously used by Wang and co-workers (Kalish et al., 2005, Datta

et al., 2001, Vasquez et al., 2001, Faruqi et al., 2000, Vasquez et al., 1999, Wang,

Seidman & Glazer, 1996).

The study presented here also assessed the genotoxicity of a single ASO targeting

a genomic locus. To design an ASO that contained homology to a selectable and a

functional protein target was limited to sites of homology. However, by employing a FACS

based method the genotoxicity of ASO can also be investigated at several distinct genomic

Page 181: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

181

loci. Additionally, AD3-HPRTPM was only mutagenic in a RAD51 induced system.

Reproducing this observation for ASO targeting different genomic loci will be important.

Additionally, although RAD51 induction appeared to be vital for AD3-HPRTPM

mediated mutation, the effect of silencing KU70 protein expression using RNAi should also

be investigated to further confirm the putative mechanism of mutation. KU70 protein

expression has been reported to negatively modulate HR repair (Morozov & Wawrousek,

2008, Pierce et al., 2001), thus, silencing KU70 protein expression may increase the

mutagenicity of AD3-HPRTPM. This would imply physical incorporation of oligonucleotide

into genomic DNA through HR repair. If silencing did not alter the potency this would imply

ASO incorporation by serving as an Okazaki fragment following strand annealing by

RAD51.

Mutation by AD3-HPRTPM at the HPRT locus did not appear to be dependent on

the mismatched base as described for TNE. Reproducing this result with AD3-HPRTPM

without a mismatched will be important to confirm the mechanism of action suggested in

chapter 4, as well as investigating the extent of mismatched bases, positional effects and

different chemical modifications.

Data presented in this study also highlighted the genotoxicity of an adenine based

phosphorothioate monophosphate nucleotide analogue. It would be interesting to

investigate the genotoxicity of phosphorothioate nucleotide analogues containing different

nucleobases i.e. thymidine, guanine and cytosine. Due to time restrictions, the genotoxicity

of 5-methyl-dCTP could not be investigated although its incorporation was shown using a

primer extension assay. Ideally, epigenetic effects could be assessed using a

transformation assay with BALB/c 3T3 cells.

A primer extension assay was employed to assess the incorporation efficiency of

nucleotide analogues using Tfi DNA polymerase. Ideally, the method should have used a

recombinant human DNA polymerase. The primer extension assay can also be used to

Page 182: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

182

investigate the fidelity of base pairing with nucleotide analogues. For example, can a

thymidine based analogue base pair with guanine. A more relevant method to assess

incorporation of nucleotide analogues would be to use a radiolabeled counterpart and

quantitate the radioactivity in genomic DNA following DNA extraction. These methods

could then be used to correlate the incorporation efficiency and mutagenicity of nucleotide

analogues using the HPRT and TK forward mutation assay and the in vitro micronucleus

assay.

Recently, improved efficacy and toxicity with ASO containing phosphorothioate and

2’O-methyl ribose has been described (section 1.2.1). The incorporation efficiency and

mutagenicity of such second generation nucleotide analogues can also be investigated

using the methods described above.

Page 183: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

183

References

Aarts, M., Dekker, M., de Vries, S., van der Wal, A. & te Riele, H. (2006) Generation of a mouse mutant by oligonucleotide-mediated gene modification in ES cells. Nucleic Acids Research. 34 (21), e147.

Aartsma-Rus, A., Kaman, W. E., Bremmer-Bout, M., Janson, A. A., den Dunnen, J. T., van Ommen, G. J. & van Deutekom, J. C. (2004) Comparative analysis of antisense oligonucleotide analogs for targeted DMD exon 46 skipping in muscle cells. Gene Therapy. 11 (18), 1391-1398.

Adrian P., B. (1987) CpG islands as gene markers in the vertebrate nucleus. Trends in Genetics. 3 (0), 342-347.

Agrawal, S. (1999) Importance of nucleotide sequence and chemical modifications of antisense oligonucleotides. Biochimica Et Biophysica Acta. 1489 (1), 53-68.

Agrawal, S. & Kandimalla, E. R. (2004) Role of Toll-like receptors in antisense and siRNA [corrected. Nature Biotechnology. 22 (12), 1533-1537.

Agrawal, S., Mayrand, S. H., Zamecnik, P. C. & Pederson, T. (1990) Site-specific excision from RNA by RNase H and mixed-phosphate-backbone oligodeoxynucleotides. Proceedings of the National Academy of Sciences of the United States of America. 87 (4), 1401-1405.

Agrawal, S., Temsamani, J. & Tang, J. Y. (1991) Pharmacokinetics, biodistribution, and stability of oligodeoxynucleotide phosphorothioates in mice. Proceedings of the National Academy of Sciences of the United States of America. 88 (17), 7595-7599.

Agrawal, S. & Zhao, Q. (1998) Mixed backbone oligonucleotides: improvement in oligonucleotide-induced toxicity in vivo. Antisense & Nucleic Acid Drug Development. 8 (2), 135-139.

Agrawal, S., Zhao, Q., Jiang, Z., Oliver, C., Giles, H., Heath, J. & Serota, D. (1997) Toxicologic effects of an oligodeoxynucleotide phosphorothioate and its analogs following intravenous administration in rats. Antisense & Nucleic Acid Drug Development. 7 (6), 575-584.

Akhtar, S., Hughes, M. D., Khan, A., Bibby, M., Hussain, M., Nawaz, Q., Double, J. & Sayyed, P. (2000) The delivery of antisense therapeutics. Advanced Drug Delivery Reviews. 44 (1), 3-21.

Akhtar, S., Kole, R. & Juliano, R. L. (1991) Stability of antisense DNA oligodeoxynucleotide analogs in cellular extracts and sera. Life Sciences. 49 (24), 1793-1801.

Applegate, M. L., Moore, M. M., Broder, C. B., Burrell, A., Juhn, G., Kasweck, K. L., Lin, P. F., Wadhams, A. & Hozier, J. C. (1990) Molecular dissection of mutations at the heterozygous thymidine kinase locus in mouse lymphoma cells. Proceedings of the National Academy of Sciences of the United States of America. 87 (1), 51-55.

Page 184: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

184

Arias-Lopez, C., Lazaro-Trueba, I., Kerr, P., Lord, C. J., Dexter, T., Iravani, M., Ashworth, A. & Silva, A. (2006) p53 modulates homologous recombination by transcriptional regulation of the RAD51 gene. EMBO Reports. 7 (2), 219-224.

Asensio, J. L., Carr, R., Brown, T. & Lane, A. N. (1999) Conformational and thermodynamic properties of parallel intramolecular triple helices containing a DNA, RNA, or 2'- OMeDNA third strand. Journal of the American Chemical Society. 121 (48), 11063-11070.

Ayers, K. M., Clive, D., Tucker, W. E.,Jr, Hajian, G. & de Miranda, P. (1996) Nonclinical toxicology studies with zidovudine: genetic toxicity tests and carcinogenicity bioassays in mice and rats. Fundamental and Applied Toxicology : Official Journal of the Society of Toxicology. 32 (2), 148-158.

Bafica, A., Scanga, C. A., Schito, M., Chaussabel, D. & Sher, A. (2004) Influence of coinfecting pathogens on HIV expression: evidence for a role of Toll-like receptors. Journal of Immunology (Baltimore, Md.: 1950). 172 (12), 7229-7234.

Baker, B. F., Lot, S. S., Condon, T. P., Cheng-Flournoy, S., Lesnik, E. A., Sasmor, H. M. & Bennett, C. F. (1997) 2'-O-(2-Methoxy)ethyl-modified anti-intercellular adhesion molecule 1 (ICAM-1) oligonucleotides selectively increase the ICAM-1 mRNA level and inhibit formation of the ICAM-1 translation initiation complex in human umbilical vein endothelial cells. The Journal of Biological Chemistry. 272 (18), 11994-12000.

Banerjee, D. (2001) Genasense (Genta Inc). Current Opinion in Investigational Drugs (London, England : 2000). 2 (4), 574-580.

Basundra, R., Kumar, A., Amrane, S., Verma, A., Phan, A. T. & Chowdhury, S. (2010) A novel G-quadruplex motif modulates promoter activity of human thymidine kinase 1. The FEBS Journal. 277 (20), 4254-4264.

Baumann, P., Benson, F. E. & West, S. C. (1996) Human Rad51 protein promotes ATP-dependent homologous pairing and strand transfer reactions in vitro. Cell. 87 (4), 757-766.

Bernardi, F. & Ninio, J. (1978) Biochimie. 60, 762-778.

Besch, R., Giovannangeli, C., Schuh, T., Kammerbauer, C. & Degitz, K. (2004) Characterization and quantification of triple helix formation in chromosomal DNA. Journal of Molecular Biology. 341 (4), 979-989.

Birmingham, K. (2000) UN acknowledges HIV/AIDS as a threat to world peace. Nature Medicine. 6 (2), 117.

Bjursell, G. & Reichard, P. (1973) Effects of thymidine on deoxyribonucleoside triphosphate pools and deoxyribonucleic acid synthesis in Chinese hamster ovary cells. The Journal of Biological Chemistry. 248 (11), 3904-3909.

Bonner, M. & Kmiec, E. B. (2009) DNA breakage associated with targeted gene alteration directed by DNA oligonucleotides. Mutation Research. 669 (1-2), 85-94.

Boobis, A. R., Lynch, A. M., Murray, S., de la Torre, R., Solans, A., Farre, M., Segura, J., Gooderham, N. J. & Davies, D. S. (1994) CYP1A2-catalyzed conversion of dietary

Page 185: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

185

heterocyclic amines to their proximate carcinogens is their major route of metabolism in humans. Cancer Research. 54 (1), 89-94.

Boosalis, M. S., Petruska, J. & Goodman, M. F. (1987) DNA polymerase insertion fidelity. Gel assay for site-specific kinetics. The Journal of Biological Chemistry. 262 (30), 14689-14696.

Boulme, F., Freund, F., Moreau, S., Nielsen, P. E., Gryaznov, S., Toulme, J. J. & Litvak, S. (1998) Modified (PNA, 2'-O-methyl and phosphoramidate) anti-TAR antisense oligonucleotides as strong and specific inhibitors of in vitro HIV-1 reverse transcription. Nucleic Acids Research. 26 (23), 5492-5500.

Brachman, E. E. & Kmiec, E. B. (2005) Gene repair in mammalian cells is stimulated by the elongation of S phase and transient stalling of replication forks. DNA Repair. 4 (4), 445-457.

Brown, K. D., Rathi, A., Kamath, R., Beardsley, D. I., Zhan, Q., Mannino, J. L. & Baskaran, R. (2003) The mismatch repair system is required for S-phase checkpoint activation. Nature Genetics. 33 (1), 80-84.

Brown, P. M. & Fox, K. R. (1996) Nucleosome core particles inhibit DNA triple helix formation. The Biochemical Journal. 319 ( Pt 2) (Pt 2), 607-611.

Broxson, C., Beckett, J. & Tornaletti, S. (2011) Transcription Arrest by a G Quadruplex Forming-Trinucleotide Repeat Sequence from the Human c-myb Gene. Biochemistry. 50 (19), 4162-4172.

Buchini, S. & Leumann, C. J. (2003) Recent improvements in antigene technology. Current Opinion in Chemical Biology. 7 (6), 717-726.

Bulfield, G., Siller, W. G., Wight, P. A. & Moore, K. J. (1984) X chromosome-linked muscular dystrophy (mdx) in the mouse. Proceedings of the National Academy of Sciences of the United States of America. 81 (4), 1189-1192.

Carter, M. M., Torres, S. M., Cook, D. L.,Jr, McCash, C. L., Yu, M., Walker, V. E. & Walker, D. M. (2007) Relative mutagenic potencies of several nucleoside analogs, alone or in drug pairs, at the HPRT and TK loci of human TK6 lymphoblastoid cells. Environmental and Molecular Mutagenesis. 48 (3-4), 239-247.

Chan, P. P. & Glazer, P. M. (1997) Triplex DNA: fundamentals, advances, and potential applications for gene therapy. Journal of Molecular Medicine (Berlin, Germany). 75 (4), 267-282.

Chen, H. X., Marshall, J. L., Ness, E., Martin, R. R., Dvorchik, B., Rizvi, N., Marquis, J., McKinlay, M., Dahut, W. & Hawkins, M. J. (2000) A safety and pharmacokinetic study of a mixed-backbone oligonucleotide (GEM231) targeting the type I protein kinase A by two-hour infusions in patients with refractory solid tumors. Clinical Cancer Research : An Official Journal of the American Association for Cancer Research. 6 (4), 1259-1266.

Chen, L., Agrawal, S., Zhou, W., Zhang, R. & Chen, J. (1998) Synergistic activation of p53 by inhibition of MDM2 expression and DNA damage. Proceedings of the National Academy of Sciences of the United States of America. 95 (1), 195-200.

Page 186: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

186

Chi, K. N., Gleave, M. E., Klasa, R., Murray, N., Bryce, C., Lopes de Menezes, D. E., D'Aloisio, S. & Tolcher, A. W. (2001) A phase I dose-finding study of combined treatment with an antisense Bcl-2 oligonucleotide (Genasense) and mitoxantrone in patients with metastatic hormone-refractory prostate cancer. Clinical Cancer Research : An Official Journal of the American Association for Cancer Research. 7 (12), 3920-3927.

Chin, D. J., Green, G. A., Zon, G., Szoka, F. C.,Jr & Straubinger, R. M. (1990) Rapid nuclear accumulation of injected oligodeoxyribonucleotides. The New Biologist. 2 (12), 1091-1100.

Cinatl, J.,Jr, Kotchetkov, R., Weimer, E., Blaheta, R. A., Scholz, M., Vogel, J. U., Gumbel, H. O. & Doerr, H. W. (2000) The antisense oligonucleotide ISIS 2922 prevents cytomegalovirus-induced upregulation of IL-8 and ICAM-1 in cultured human fibroblasts. Journal of Medical Virology. 60 (3), 313-323.

Clements, J. (2000) The mouse lymphoma assay. Mutation Research. 455 (1-2), 97-110.

Clive, D., Bolcsfoldi, G., Clements, J., Cole, J., Homna, M., Majeska, J., Moore, M., Muller, L., Myhr, B. & Oberly, T. (1995) Consensus agreement regarding protocol issues discussed during the mouse lymphoma workshop: Portland, Oregon, May 7, 1994. Environmental and Molecular Mutagenesis. 25 (2), 165-168.

Cole-Strauss, A., Gamper, H., Holloman, W. K., Munoz, M., Cheng, N. & Kmiec, E. B. (1999) Targeted gene repair directed by the chimeric RNA/DNA oligonucleotide in a mammalian cell-free extract. Nucleic Acids Research. 27 (5), 1323-1330.

Connor, E. M., Sperling, R. S., Gelber, R., Kiselev, P., Scott, G., O'Sullivan, M. J., VanDyke, R., Bey, M., Shearer, W. & Jacobson, R. L. (1994) Reduction of maternal-infant transmission of human immunodeficiency virus type 1 with zidovudine treatment. Pediatric AIDS Clinical Trials Group Protocol 076 Study Group. The New England Journal of Medicine. 331 (18), 1173-1180.

Cooney, M., Czernuszewicz, G., Postel, E. H., Flint, S. J. & Hogan, M. E. (1988) Site-specific oligonucleotide binding represses transcription of the human c-myc gene in vitro. Science (New York, N.Y.). 241 (4864), 456-459.

Cordon-Cardo, C., Latres, E., Drobnjak, M., Oliva, M. R., Pollack, D., Woodruff, J. M., Marechal, V., Chen, J., Brennan, M. F. & Levine, A. J. (1994) Molecular abnormalities of mdm2 and p53 genes in adult soft tissue sarcomas. Cancer Research. 54 (3), 794-799.

Crooke, S. T. (1998) Vitravene--another piece in the mosaic. Antisense & Nucleic Acid Drug Development. 8 (4), vii-viii.

Cunningham, C. C., Holmlund, J. T., Geary, R. S., Kwoh, T. J., Dorr, A., Johnston, J. F., Monia, B. & Nemunaitis, J. (2001) A Phase I trial of H-ras antisense oligonucleotide ISIS 2503 administered as a continuous intravenous infusion in patients with advanced carcinoma. Cancer. 92 (5), 1265-1271.

Cunningham, C. C., Holmlund, J. T., Schiller, J. H., Geary, R. S., Kwoh, T. J., Dorr, A. & Nemunaitis, J. (2000) A phase I trial of c-Raf kinase antisense oligonucleotide ISIS 5132 administered as a continuous intravenous infusion in patients with advanced cancer.

Page 187: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

187

Clinical Cancer Research : An Official Journal of the American Association for Cancer Research. 6 (5), 1626-1631.

Dasgupta, S., Masukata, H. & Tomizawa, J. (1987) Multiple mechanisms for initiation of ColE1 DNA replication: DNA synthesis in the presence and absence of ribonuclease H. Cell. 51 (6), 1113-1122.

Datta, H. J., Chan, P. P., Vasquez, K. M., Gupta, R. C. & Glazer, P. M. (2001) Triplex-induced recombination in human cell-free extracts. Dependence on XPA and HsRad51. The Journal of Biological Chemistry. 276 (21), 18018-18023.

Davidson, B. L. & McCray, P. B.,Jr. (2011) Current prospects for RNA interference-based therapies. Nature Reviews.Genetics. 12 (5), 329-340.

de Wind, N., Dekker, M., Berns, A., Radman, M. & te Riele, H. (1995) Inactivation of the mouse Msh2 gene results in mismatch repair deficiency, methylation tolerance, hyperrecombination, and predisposition to cancer. Cell. 82 (2), 321-330.

Dean, N., McKay, R., Miraglia, L., Howard, R., Cooper, S., Giddings, J., Nicklin, P., Meister, L., Ziel, R., Geiger, T., Muller, M. & Fabbro, D. (1996) Inhibition of growth of human tumor cell lines in nude mice by an antisense of oligonucleotide inhibitor of protein kinase C-alpha expression. Cancer Research. 56 (15), 3499-3507.

Dean, N. M. & Bennett, C. F. (2003) Antisense oligonucleotide-based therapeutics for cancer. Oncogene. 22 (56), 9087-9096.

Dean, N. M., McKay, R., Condon, T. P. & Bennett, C. F. (1994) Inhibition of protein kinase C-alpha expression in human A549 cells by antisense oligonucleotides inhibits induction of intercellular adhesion molecule 1 (ICAM-1) mRNA by phorbol esters. The Journal of Biological Chemistry. 269 (23), 16416-16424.

Dekker, M., Brouwers, C. & te Riele, H. (2003) Targeted gene modification in mismatch-repair-deficient embryonic stem cells by single-stranded DNA oligonucleotides. Nucleic Acids Research. 31 (6), e27.

Dekker, M., de Vries, S., Aarts, M., Dekker, R., Brouwers, C., Wiebenga, O., de Wind, N., Cantelli, E., Tonelli, R. & Te Riele, H. (2011) Transient suppression of MLH1 allows effective single-nucleotide substitution by single-stranded DNA oligonucleotides. Mutation Research. 715 (1-2), 52-60.

Denli, A. M. & Hannon, G. J. (2003) RNAi: an ever-growing puzzle. Trends in Biochemical Sciences. 28 (4), 196-201.

Dias, N., Dheur, S., Nielsen, P. E., Gryaznov, S., Van Aerschot, A., Herdewijn, P., Helene, C. & Saison-Behmoaras, T. E. (1999) Antisense PNA tridecamers targeted to the coding region of Ha-ras mRNA arrest polypeptide chain elongation. Journal of Molecular Biology. 294 (2), 403-416.

Dias, N. & Stein, C. A. (2002) Antisense oligonucleotides: basic concepts and mechanisms. Molecular Cancer Therapeutics. 1 (5), 347-355.

Page 188: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

188

Doak, S. H., Jenkins, G. J., Johnson, G. E., Quick, E., Parry, E. M. & Parry, J. M. (2007) Mechanistic influences for mutation induction curves after exposure to DNA-reactive carcinogens. Cancer Research. 67 (8), 3904-3911.

Donovan, J. W., Ladetto, M., Zou, G., Neuberg, D., Poor, C., Bowers, D. & Gribben, J. G. (2000) Immunoglobulin heavy-chain consensus probes for real-time PCR quantification of residual disease in acute lymphoblastic leukemia. Blood. 95 (8), 2651-2658.

Drury, M. D. & Kmiec, E. B. (2003) DNA pairing is an important step in the process of targeted nucleotide exchange. Nucleic Acids Research. 31 (3), 899-910.

Duca, M., Vekhoff, P., Oussedik, K., Halby, L. & Arimondo, P. B. (2008) The triple helix: 50 years later, the outcome. Nucleic Acids Research. 36 (16), 5123-5138.

Durland, R. H., Kessler, D. J., Gunnell, S., Duvic, M., Pettitt, B. M. & Hogan, M. E. (1991) Binding of triple helix forming oligonucleotides to sites in gene promoters. Biochemistry. 30 (38), 9246-9255.

Eckstein, F. (1985) Nucleoside phosphorothioates. Annual Review of Biochemistry. 54, 367-402.

Egholm, M., Buchardt, O., Christensen, L., Behrens, C., Freier, S. M., Driver, D. A., Berg, R. H., Kim, S. K., Norden, B. & Nielsen, P. E. (1993) PNA hybridizes to complementary oligonucleotides obeying the Watson-Crick hydrogen-bonding rules. Nature. 365 (6446), 566-568.

Elbashir, S. M., Martinez, J., Patkaniowska, A., Lendeckel, W. & Tuschl, T. (2001) Functional anatomy of siRNAs for mediating efficient RNAi in Drosophila melanogaster embryo lysate. The EMBO Journal. 20 (23), 6877-6888.

Esteller, M. (2008) Epigenetics in cancer. The New England Journal of Medicine. 358 (11), 1148-1159.

European Medicines Agency. Doc. Ref.EMEA/CHMP/SWP/199726/2004. , .

Faruqi, A. F., Datta, H. J., Carroll, D., Seidman, M. M. & Glazer, P. M. (2000) Triple-helix formation induces recombination in mammalian cells via a nucleotide excision repair-dependent pathway. Molecular and Cellular Biology. 20 (3), 990-1000.

Felsenfeld, G., Davies, D.,R. & Rich, ,Alexander. (1957) Formation of a three-stranded polynucleotide molecule. Journal of the American Chemical Society. 79 (8), 2023-2024.

Fersht, A. R. (1979) Fidelity of replication of phage phi X174 DNA by DNA polymerase III holoenzyme: spontaneous mutation by misincorporation. Proceedings of the National Academy of Sciences of the United States of America. 76 (10), 4946-4950.

Frank-Kamenetskii, M. D. & Mirkin, S. M. (1995) Triplex DNA structures. Annual Review of Biochemistry. 64, 65-95.

Froehler, B., Ng, P. & Matteucci, M. (1988) Phosphoramidate analogues of DNA: synthesis and thermal stability of heteroduplexes. Nucleic Acids Research. 16 (11), 4831-4839.

Page 189: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

189

Furdon, P. J., Dominski, Z. & Kole, R. (1989) RNase H cleavage of RNA hybridized to oligonucleotides containing methylphosphonate, phosphorothioate and phosphodiester bonds. Nucleic Acids Research. 17 (22), 9193-9204.

Furman, P. A., Fyfe, J. A., St Clair, M. H., Weinhold, K., Rideout, J. L., Freeman, G. A., Lehrman, S. N., Bolognesi, D. P., Broder, S. & Mitsuya, H. (1986) Phosphorylation of 3'-azido-3'-deoxythymidine and selective interaction of the 5'-triphosphate with human immunodeficiency virus reverse transcriptase. Proceedings of the National Academy of Sciences of the United States of America. 83 (21), 8333-8337.

Gamper, H. B.,Jr, Cole-Strauss, A., Metz, R., Parekh, H., Kumar, R. & Kmiec, E. B. (2000) A plausible mechanism for gene correction by chimeric oligonucleotides. Biochemistry. 39 (19), 5808-5816.

Geary, R. S. (2009) Antisense oligonucleotide pharmacokinetics and metabolism. Expert Opinion on Drug Metabolism & Toxicology. 5 (4), 381-391.

Gescher, A. (1992) Towards selective pharmacological modulation of protein kinase C--opportunities for the development of novel antineoplastic agents. British Journal of Cancer. 66 (1), 10-19.

Glaab, W. E., Kort, K. L. & Skopek, T. R. (2000) Specificity of mutations induced by the food-associated heterocyclic amine 2-amino-1-methyl-6-phenylimidazo-[4,5-b]-pyridine in colon cancer cell lines defective in mismatch repair. Cancer Research. 60 (17), 4921-4925.

Goemans, N. M., Tulinius, M., van den Akker, J. T., Burm, B. E., Ekhart, P. F., Heuvelmans, N., Holling, T., Janson, A. A., Platenburg, G. J., Sipkens, J. A., Sitsen, J. M., Aartsma-Rus, A., van Ommen, G. J., Buyse, G., Darin, N., Verschuuren, J. J., Campion, G. V., de Kimpe, S. J. & van Deutekom, J. C. (2011) Systemic administration of PRO051 in Duchenne's muscular dystrophy. The New England Journal of Medicine. 364 (16), 1513-1522.

Gonzalez Cid, M. & Larripa, I. (1994) Genotoxic activity of azidothymidine (AZT) in in vitro systems. Mutation Research. 321 (1-2), 113-118.

Greger, V., Passarge, E., Hopping, W., Messmer, E. & Horsthemke, B. (1989) Epigenetic changes may contribute to the formation and spontaneous regression of retinoblastoma. Human Genetics. 83 (2), 155-158.

Grigoriev, M., Praseuth, D., Robin, P., Hemar, A., Saison-Behmoaras, T., Dautry-Varsat, A., Thuong, N. T., Helene, C. & Harel-Bellan, A. (1992) A triple helix-forming oligonucleotide-intercalator conjugate acts as a transcriptional repressor via inhibition of NF kappa B binding to interleukin-2 receptor alpha-regulatory sequence. The Journal of Biological Chemistry. 267 (5), 3389-3395.

Gross, A., McDonnell, J. M. & Korsmeyer, S. J. (1999) BCL-2 family members and the mitochondria in apoptosis. Genes & Development. 13 (15), 1899-1911.

Grunweller, A., Wyszko, E., Bieber, B., Jahnel, R., Erdmann, V. A. & Kurreck, J. (2003) Comparison of different antisense strategies in mammalian cells using locked nucleic

Page 190: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

190

acids, 2'-O-methyl RNA, phosphorothioates and small interfering RNA. Nucleic Acids Research. 31 (12), 3185-3193.

Guntaka, R. V., Varma, B. R. & Weber, K. T. (2003) Triplex-forming oligonucleotides as modulators of gene expression. The International Journal of Biochemistry & Cell Biology. 35 (1), 22-31.

Gupta, R. C., Bazemore, L. R., Golub, E. I. & Radding, C. M. (1997) Activities of human recombination protein Rad51. Proceedings of the National Academy of Sciences of the United States of America. 94 (2), 463-468.

Hammond, S. M., Bernstein, E., Beach, D. & Hannon, G. J. (2000) An RNA-directed nuclease mediates post-transcriptional gene silencing in Drosophila cells. Nature. 404 (6775), 293-296.

Hanawalt, P. C. (1994) Transcription-coupled repair and human disease. Science (New York, N.Y.). 266 (5193), 1957-1958.

Hannay, J. A., Liu, J., Zhu, Q. S., Bolshakov, S. V., Li, L., Pisters, P. W., Lazar, A. J., Yu, D., Pollock, R. E. & Lev, D. (2007) Rad51 overexpression contributes to chemoresistance in human soft tissue sarcoma cells: a role for p53/activator protein 2 transcriptional regulation. Molecular Cancer Therapeutics. 6 (5), 1650-1660.

Hausen, P. & Stein, H. (1970) Ribonuclease H. An enzyme degrading the RNA moiety of DNA-RNA hybrids. European Journal of Biochemistry / FEBS. 14 (2), 278-283.

Hegan, D. C., Narayanan, L., Jirik, F. R., Edelmann, W., Liskay, R. M. & Glazer, P. M. (2006) Differing patterns of genetic instability in mice deficient in the mismatch repair genes Pms2, Mlh1, Msh2, Msh3 and Msh6. Carcinogenesis. 27 (12), 2402-2408.

Heidelberger, C., Freeman, A. E., Pienta, R. J., Sivak, A., Bertram, J. S., Casto, B. C., Dunkel, V. C., Francis, M. W., Kakunaga, T., Little, J. B. & Schechtman, L. M. (1983) Cell transformation by chemical agents--a review and analysis of the literature. A report of the U.S. Environmental Protection Agency Gene-Tox Program. Mutation Research. 114 (3), 283-385.

Hemmi, H., Takeuchi, O., Kawai, T., Kaisho, T., Sato, S., Sanjo, H., Matsumoto, M., Hoshino, K., Wagner, H., Takeda, K. & Akira, S. (2000) A Toll-like receptor recognizes bacterial DNA. Nature. 408 (6813), 740-745.

Henry, S. P., Zuckerman, J. E., Rojko, J., Hall, W. C., Harman, R. J., Kitchen, D. & Crooke, S. T. (1997) Toxicological properties of several novel oligonucleotide analogs in mice. Anti-Cancer Drug Design. 12 (1), 1-14.

Herman, J. G. & Baylin, S. B. (2003) Gene silencing in cancer in association with promoter hypermethylation. The New England Journal of Medicine. 349 (21), 2042-2054.

Hoever, G., Groeschel, B., Chandra, P., Doerr, H. W. & Cinatl, J. (2003) The mechanism of 3'-azido-2',3'-dideoxythymidine resistance to human lymphoid cells. International Journal of Molecular Medicine. 11 (6), 743-747.

Page 191: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

191

Hoffman, E. P., Brown, R. H.,Jr & Kunkel, L. M. (1987) Dystrophin: the protein product of the Duchenne muscular dystrophy locus. Cell. 51 (6), 919-928.

Hogrefe, R. I. (1999) An antisense oligonucleotide primer. Antisense & Nucleic Acid Drug Development. 9 (4), 351-357.

Hoke, G. D., Draper, K., Freier, S. M., Gonzalez, C., Driver, V. B., Zounes, M. C. & Ecker, D. J. (1991) Effects of phosphorothioate capping on antisense oligonucleotide stability, hybridization and antiviral efficacy versus herpes simplex virus infection. Nucleic Acids Research. 19 (20), 5743-5748.

Hoogsteen, K. (1959) The structure of crystals containing a hydrogen-bonded complex of 1-methylthymine and 9-methyladenine. Acta Crystallographica. 12, 822-823.

Hozier, J., Sawyer, J., Moore, M., Howard, B. & Clive, D. (1981) Cytogenetic analysis of the L5178Y/TK+/- leads to TK-/- mouse lymphoma mutagenesis assay system. Mutation Research. 84 (1), 169-181.

Huang, J. C., Svoboda, D. L., Reardon, J. T. & Sancar, A. (1992) Human nucleotide excision nuclease removes thymine dimers from DNA by incising the 22nd phosphodiester bond 5' and the 6th phosphodiester bond 3' to the photodimer. Proceedings of the National Academy of Sciences of the United States of America. 89 (8), 3664-3668.

Huang, P., Farquhar, D. & Plunkett, W. (1990) Selective action of 3'-azido-3'-deoxythymidine 5'-triphosphate on viral reverse transcriptases and human DNA polymerases. The Journal of Biological Chemistry. 265 (20), 11914-11918.

Igoucheva, O., Alexeev, V., Pryce, M. & Yoon, K. (2003) Transcription affects formation and processing of intermediates in oligonucleotide-mediated gene alteration. Nucleic Acids Research. 31 (10), 2659-2670.

Jain, A., Wang, G. & Vasquez, K. M. (2008) DNA triple helices: biological consequences and therapeutic potential. Biochimie. 90 (8), 1117-1130.

Jeggo, P. A. (1998) DNA breakage and repair. Advances in Genetics. 38, 185-218.

Johnson, A. A., Ray, A. S., Hanes, J., Suo, Z., Colacino, J. M., Anderson, K. S. & Johnson, K. A. (2001) Toxicity of antiviral nucleoside analogs and the human mitochondrial DNA polymerase. The Journal of Biological Chemistry. 276 (44), 40847-40857.

Johnson, R. D. & Jasin, M. (2001) Double-strand-break-induced homologous recombination in mammalian cells. Biochemical Society Transactions. 29 (Pt 2), 196-201.

Kalish, J. M., Seidman, M. M., Weeks, D. L. & Glazer, P. M. (2005) Triplex-induced recombination and repair in the pyrimidine motif. Nucleic Acids Research. 33 (11), 3492-3502.

Kanaori, K., Tamura, Y., Wada, T., Nishi, M., Kanehara, H., Morii, T., Tajima, K. & Makino, K. (1999) Structure and stability of the consecutive stereoregulated chiral phosphorothioate DNA duplex. Biochemistry. 38 (49), 16058-16066.

Page 192: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

192

Kenner, O., Kneisel, A., Klingler, J., Bartelt, B., Speit, G., Vogel, W. & Kaufmann, D. (2002) Targeted gene correction of hprt mutations by 45 base single-stranded oligonucleotides. Biochemical and Biophysical Research Communications. 299 (5), 787-792.

Klein, H. L. (2008) The consequences of Rad51 overexpression for normal and tumor cells. DNA Repair. 7 (5), 686-693.

Kochetkova, M. & Shannon, M. F. (1996) DNA triplex formation selectively inhibits granulocyte-macrophage colony-stimulating factor gene expression in human T cells. The Journal of Biological Chemistry. 271 (24), 14438-14444.

Koenig, M., Beggs, A. H., Moyer, M., Scherpf, S., Heindrich, K., Bettecken, T., Meng, G., Muller, C. R., Lindlof, M. & Kaariainen, H. (1989) The molecular basis for Duchenne versus Becker muscular dystrophy: correlation of severity with type of deletion. American Journal of Human Genetics. 45 (4), 498-506.

Koenig, M., Monaco, A. P. & Kunkel, L. M. (1988) The complete sequence of dystrophin predicts a rod-shaped cytoskeletal protein. Cell. 53 (2), 219-228.

Kresnak, M. T. & Davidson, R. L. (1992) Thymidine-induced mutations in mammalian cells: sequence specificity and implications for mutagenesis in vivo. Proceedings of the National Academy of Sciences of the United States of America. 89 (7), 2829-2833.

Krieg, A. M. (1999) Mechanisms and applications of immune stimulatory CpG oligodeoxynucleotides. Biochimica Et Biophysica Acta. 1489 (1), 107-116.

Krieg, A. M., Yi, A. K., Matson, S., Waldschmidt, T. J., Bishop, G. A., Teasdale, R., Koretzky, G. A. & Klinman, D. M. (1995) CpG motifs in bacterial DNA trigger direct B-cell activation. Nature. 374 (6522), 546-549.

Kunkel, T. A. & Erie, D. A. (2005) DNA mismatch repair. Annual Review of Biochemistry. 74, 681-710.

Kunkel, T. A. & Loeb, L. A. (1979) On the fidelity of DNA replication. Effect of divalent metal ion activators and deoxyrionucleoside triphosphate pools on in vitro mutagenesis. The Journal of Biological Chemistry. 254 (13), 5718-5725.

Kunz, B. A. (1988) Mutagenesis and deoxyribonucleotide pool imbalance. Mutation Research. 200 (1-2), 133-147.

Kunz, B. A., Kohalmi, S. E., Kunkel, T. A., Mathews, C. K., McIntosh, E. M. & Reidy, J. A. (1994) International Commission for Protection Against Environmental Mutagens and Carcinogens. Deoxyribonucleoside triphosphate levels: a critical factor in the maintenance of genetic stability. Mutation Research. 318 (1), 1-64.

Kuramoto, E., Yano, O., Kimura, Y., Baba, M., Makino, T., Yamamoto, S., Yamamoto, T., Kataoka, T. & Tokunaga, T. (1992) Oligonucleotide sequences required for natural killer cell activation. Japanese Journal of Cancer Research : Gann. 83 (11), 1128-1131.

Page 193: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

193

Kurreck, J., Wyszko, E., Gillen, C. & Erdmann, V. A. (2002) Design of antisense oligonucleotides stabilized by locked nucleic acids. Nucleic Acids Research. 30 (9), 1911-1918.

Lacenere, C., Garg, M. K., Stoltz, B. M. & Quake, S. R. (2006) Effects of a modified dye-labeled nucleotide spacer arm on incorporation by thermophilic DNA polymerases. Nucleosides, Nucleotides & Nucleic Acids. 25 (1), 9-15.

Lacroix, B. & Maddox, A. S. (2011) Cytokinesis, ploidy and aneuploidy. The Journal of Pathology. , .

Larrouy, B., Blonski, C., Boiziau, C., Stuer, M., Moreau, S., Shire, D. & Toulme, J. J. (1992) RNase H-mediated inhibition of translation by antisense oligodeoxyribonucleotides: use of backbone modification to improve specificity. Gene. 121 (2), 189-194.

Lee, H., Hanes, J. & Johnson, K. A. (2003) Toxicity of nucleoside analogues used to treat AIDS and the selectivity of the mitochondrial DNA polymerase. Biochemistry. 42 (50), 14711-14719.

Lee, J. S., Woodsworth, M. L., Latimer, L. J. & Morgan, A. R. (1984) Poly(pyrimidine) . poly(purine) synthetic DNAs containing 5-methylcytosine form stable triplexes at neutral pH. Nucleic Acids Research. 12 (16), 6603-6614.

Leonetti, J. P., Mechti, N., Degols, G., Gagnor, C. & Lebleu, B. (1991) Intracellular distribution of microinjected antisense oligonucleotides. Proceedings of the National Academy of Sciences of the United States of America. 88 (7), 2702-2706.

Lepik, D., Jaks, V., Kadaja, L., Varv, S. & Maimets, T. (2003) Electroporation and carrier DNA cause p53 activation, cell cycle arrest, and apoptosis. Analytical Biochemistry. 318 (1), 52-59.

Levin, A. A. (1999) A review of the issues in the pharmacokinetics and toxicology of phosphorothioate antisense oligonucleotides. Biochimica Et Biophysica Acta. 1489 (1), 69-84.

Levine, R. A. & Taylor, M. W. (1982) Mechanism of adenine toxicity in Escherichia coli. Journal of Bacteriology. 149 (3), 923-930.

Li, G. M. (2008) Mechanisms and functions of DNA mismatch repair. Cell Research. 18 (1), 85-98.

Liechty, M. C., Scalzi, J. M., Sims, K. R., Crosby, H.,Jr, Spencer, D. L., Davis, L. M., Caspary, W. J. & Hozier, J. C. (1998) Analysis of large and small colony L5178Y tk-/- mouse lymphoma mutants by loss of heterozygosity (LOH) and by whole chromosome 11 painting: detection of recombination. Mutagenesis. 13 (5), 461-474.

Liu, L., Rice, M. C., Drury, M., Cheng, S., Gamper, H. & Kmiec, E. B. (2002) Strand bias in targeted gene repair is influenced by transcriptional activity. Molecular and Cellular Biology. 22 (11), 3852-3863.

Luo, Z., Macris, M. A., Faruqi, A. F. & Glazer, P. M. (2000) High-frequency intrachromosomal gene conversion induced by triplex-forming oligonucleotides

Page 194: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

194

microinjected into mouse cells. Proceedings of the National Academy of Sciences of the United States of America. 97 (16), 9003-9008.

Maacke, H., Opitz, S., Jost, K., Hamdorf, W., Henning, W., Kruger, S., Feller, A. C., Lopens, A., Diedrich, K., Schwinger, E. & Sturzbecher, H. W. (2000) Over-expression of wild-type Rad51 correlates with histological grading of invasive ductal breast cancer. International Journal of Cancer.Journal International Du Cancer. 88 (6), 907-913.

Macris, M. A. & Glazer, P. M. (2003) Transcription dependence of chromosomal gene targeting by triplex-forming oligonucleotides. The Journal of Biological Chemistry. 278 (5), 3357-3362.

Maguire, K. K. & Kmiec, E. B. (2007) Multiple roles for MSH2 in the repair of a deletion mutation directed by modified single-stranded oligonucleotides. Gene. 386 (1-2), 107-114.

Maine, I. P. & Kodadek, T. (1994) Efficient unwinding of triplex DNA by a DNA helicase. Biochemical and Biophysical Research Communications. 204 (3), 1119-1124.

Majumdar, A., Khorlin, A., Dyatkina, N., Lin, F. L., Powell, J., Liu, J., Fei, Z., Khripine, Y., Watanabe, K. A., George, J., Glazer, P. M. & Seidman, M. M. (1998) Targeted gene knockout mediated by triple helix forming oligonucleotides. Nature Genetics. 20 (2), 212-214.

Mann, C. J., Honeyman, K., Cheng, A. J., Ly, T., Lloyd, F., Fletcher, S., Morgan, J. E., Partridge, T. A. & Wilton, S. D. (2001) Antisense-induced exon skipping and synthesis of dystrophin in the mdx mouse. Proceedings of the National Academy of Sciences of the United States of America. 98 (1), 42-47.

Mato, J. M., Alvarez, L., Ortiz, P. & Pajares, M. A. (1997) S-adenosylmethionine synthesis: molecular mechanisms and clinical implications. Pharmacology & Therapeutics. 73 (3), 265-280.

Mattano, S. S., Palella, T. D. & Mitchell, B. S. (1990) Mutations induced at the hypoxanthine-guanine phosphoribosyltransferase locus of human T-lymphoblasts by perturbations of purine deoxyribonucleoside triphosphate pools. Cancer Research. 50 (15), 4566-4571.

McGraw, K., McKay, R., Miraglia, L., Boggs, R. T., Pribble, J. P., Muller, M., Geiger, T., Fabbro, D. & Dean, N. M. (1997) Antisense oligonucleotide inhibitors of isozymes of protein kinase C: in vitro and in vivo activity, and clinical development as anti-cancer therapeutics. Anti-Cancer Drug Design. 12 (5), 315-326.

McGregor, D. B., Riach, C., Cattanach, P., Edwards, I., Shepherd, W. & Caspary, W. J. (1996) Mutagenic responses of L5178Y mouse cells at the tk and hprt loci. Toxicology in Vitro : An International Journal Published in Association with BIBRA. 10 (5), 643-647.

Meng, Q., Su, T., O'Neill, J. P. & Walker, V. E. (2002) Molecular analysis of mutations at the HPRT and TK loci of human lymphoblastoid cells after combined treatments with 3'-azido-3'-deoxythymidine and 2',3'-dideoxyinosinedagger. Environmental and Molecular Mutagenesis. 39 (4), 282-295.

Page 195: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

195

Metelev, V., Lisziewicz, J. & Agrawal, S. (1994) Study of antisense oligonucleotide phosphorothioates containing segments of oligodeoxynucleotides and 2'-O-methyloligoribonucleotides Bioorganic & Medicinal Chemistry Letters. 4, 2929-2934.

Meuth, M. (1984) The genetic consequences of nucleotide precursor pool imbalance in mammalian cells. Mutation Research. 126 (2), 107-112.

Meuth, M. (1981a) Role of deoxynucleoside triphosphate pools in the cytotoxic and mutagenic effects of DNA alkylating agents. Somatic Cell Genetics. 7 (1), 89-102.

Meuth, M. (1981b) Sensitivity of a mutator gene in Chinese hamster ovary cell to deoxynucleoside triphosphate pool alterations. Molecular and Cellular Biology. 1 (7), 652-660.

Miller, P. (1989) Non-ionic antisense oligonucleotides: oligonucleotide methylphosphonates. In: Cohen, J. S. (ed.).Oligodeoxynucleotides: Anti-sense Inhibitors of Gene Expression. Boca Raton, Fl, USA, CRC Press. pp. 85-92.

Miller, P. S., Yano, J., Yano, E., Carroll, C., Jayaraman, K. & Ts'o, P. O. (1979) Nonionic nucleic acid analogues. Synthesis and characterization of dideoxyribonucleoside methylphosphonates. Biochemistry. 18 (23), 5134-5143.

Momand, J., Zambetti, G. P., Olson, D. C., George, D. & Levine, A. J. (1992) The mdm-2 oncogene product forms a complex with the p53 protein and inhibits p53-mediated transactivation. Cell. 69 (7), 1237-1245.

Monaco, A. P., Bertelson, C. J., Liechti-Gallati, S., Moser, H. & Kunkel, L. M. (1988) An explanation for the phenotypic differences between patients bearing partial deletions of the DMD locus. Genomics. 2 (1), 90-95.

Monia, B. P., Lesnik, E. A., Gonzalez, C., Lima, W. F., McGee, D., Guinosso, C. J., Kawasaki, A. M., Cook, P. D. & Freier, S. M. (1993) Evaluation of 2'-modified oligonucleotides containing 2'-deoxy gaps as antisense inhibitors of gene expression. The Journal of Biological Chemistry. 268 (19), 14514-14522.

Moore, M. M., Clive, D., Hozier, J. C., Howard, B. E., Batson, A. G., Turner, N. T. & Sawyer, J. (1985) Analysis of trifluorothymidine-resistant (TFTr) mutants of L5178Y/TK+/- mouse lymphoma cells. Mutation Research. 151 (1), 161-174.

Morozov, V. & Wawrousek, E. F. (2008) Single-strand DNA-mediated targeted mutagenesis of genomic DNA in early mouse embryos is stimulated by Rad51/54 and by Ku70/86 inhibition. Gene Therapy. 15 (6), 468-472.

Morris, M. J., Tong, W. P., Cordon-Cardo, C., Drobnjak, M., Kelly, W. K., Slovin, S. F., Terry, K. L., Siedlecki, K., Swanson, P., Rafi, M., DiPaola, R. S., Rosen, N. & Scher, H. I. (2002) Phase I trial of BCL-2 antisense oligonucleotide (G3139) administered by continuous intravenous infusion in patients with advanced cancer. Clinical Cancer Research : An Official Journal of the American Association for Cancer Research. 8 (3), 679-683.

Morvan, F., Porumb, H., Degols, G., Lefebvre, I., Pompon, A., Sproat, B. S., Rayner, B., Malvy, C., Lebleu, B. & Imbach, J. L. (1993) Comparative evaluation of seven

Page 196: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

196

oligonucleotide analogues as potential antisense agents. Journal of Medicinal Chemistry. 36 (2), 280-287.

Narayanan, L., Fritzell, J. A., Baker, S. M., Liskay, R. M. & Glazer, P. M. (1997) Elevated levels of mutation in multiple tissues of mice deficient in the DNA mismatch repair gene Pms2. Proceedings of the National Academy of Sciences of the United States of America. 94 (7), 3122-3127.

Nyce, J. (1989) Drug-induced DNA hypermethylation and drug resistance in human tumors. Cancer Research. 49 (21), 5829-5836.

Nyce, J., Leonard, S., Canupp, D., Schulz, S. & Wong, S. (1993) Epigenetic mechanisms of drug resistance: drug-induced DNA hypermethylation and drug resistance. Proceedings of the National Academy of Sciences of the United States of America. 90 (7), 2960-2964.

Nyce, J., Mylott, D., Leonard, S., Willis, L. & Kataria, A. (1989) Detection of Drug-Induced DNA Hypermethylation in Human Tumor Cells Exposed to Cancer Chemotherapy Agents. Journal of Liquid Chromatography. 12 (8), 1313-1321.

Oberly, T. J. & Garriot, M. L. (1996) Influence of cytotoxicity on test results in the L5178Y TK+/- mouse lymphoma assay. Environmental and Molecular Mutagenesis. 27 (1), 75-78.

Oliner, J. D., Kinzler, K. W., Meltzer, P. S., George, D. L. & Vogelstein, B. (1992) Amplification of a gene encoding a p53-associated protein in human sarcomas. Nature. 358 (6381), 80-83.

Olivero, O., Beland, F. & Poirier, M. (1994) Immunofluorescent localization and quantitation of 3'-azido-2',3'-dideoxythymidine (azt) incorporated into chromosomal DNA of human, hamster and mouse-cell lines. International Journal of Oncology. 4 (1), 49-54.

Olivero, O. A. (2008) Relevance of experimental models for investigation of genotoxicity induced by antiretroviral therapy during human pregnancy. Mutation Research. 658 (3), 184-190.

Olivero, O. A. (2007) Mechanisms of genotoxicity of nucleoside reverse transcriptase inhibitors. Environmental and Molecular Mutagenesis. 48 (3-4), 215-223.

Olivero, O. A., Beland, F. A., Fullerton, N. F. & Poirier, M. C. (1994) Vaginal epithelial DNA damage and expression of preneoplastic markers in mice during chronic dosing with tumorigenic levels of 3'-azido-2',3'-dideoxythymidine. Cancer Research. 54 (23), 6235-6242.

Olivero, O. A., Parikka, R., Poirier, M. C. & Vahakangas, K. (1999a) 3'-azido-3'-deoxythymidine (AZT) transplacental perfusion kinetics and DNA incorporation in normal human placentas perfused with AZT. Mutation Research. 428 (1-2), 41-47.

Olivero, O. A., Shearer, G. M., Chougnet, C. A., Kovacs, A. A., Landay, A. L., Baker, R., Stek, A. M., Khoury, M. M., Proia, L. A., Kessler, H. A., Sha, B. E., Tarone, R. E. & Poirier, M. C. (1999b) Incorporation of zidovudine into leukocyte DNA from HIV-1-positive adults and pregnant women, and cord blood from infants exposed in utero. AIDS (London, England). 13 (8), 919-925.

Page 197: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

197

Olsen, P. A., Randol, M., Luna, L., Brown, T. & Krauss, S. (2005) Genomic sequence correction by single-stranded DNA oligonucleotides: role of DNA synthesis and chemical modifications of the oligonucleotide ends. The Journal of Gene Medicine. 7 (12), 1534-1544.

Olsen, P. A., Solhaug, A., Booth, J. A., Gelazauskaite, M. & Krauss, S. (2009) Cellular responses to targeted genomic sequence modification using single-stranded oligonucleotides and zinc-finger nucleases. DNA Repair. 8 (3), 298-308.

Orum, H. & Wengel, J. (2001) Locked nucleic acids: a promising molecular family for gene-function analysis and antisense drug development. Current Opinion in Molecular Therapeutics. 3 (3), 239-243.

Panburana, P., Sirinavin, S., Phuapradit, W., Vibhagool, A. & Chantratita, W. (2004) Elective cesarean delivery plus short-course lamivudine and zidovudine for the prevention of mother-to-child transmission of human immunodeficiency virus type 1. American Journal of Obstetrics and Gynecology. 190 (3), 803-808.

Papadopoulos, N., Nicolaides, N. C., Wei, Y. F., Ruben, S. M., Carter, K. C., Rosen, C. A., Haseltine, W. A., Fleischmann, R. D., Fraser, C. M. & Adams, M. D. (1994) Mutation of a mutL homolog in hereditary colon cancer. Science (New York, N.Y.). 263 (5153), 1625-1629.

Papaioannou, I., Disterer, P. & Owen, J. S. (2009) Use of internally nuclease-protected single-strand DNA oligonucleotides and silencing of the mismatch repair protein, MSH2, enhances the replication of corrected cells following gene editing. The Journal of Gene Medicine. 11 (3), 267-274.

Parekh-Olmedo, H., Ferrara, L., Brachman, E. & Kmiec, E. B. (2005) Gene therapy progress and prospects: targeted gene repair. Gene Therapy. 12 (8), 639-646.

Peng Ho, S., Livanov, V., Zhang, W., Li, J. & Lesher, T. (1998) Modification of phosphorothioate oligonucleotides yields potent analogs with minimal toxicity for antisense experiments in the CNS. Brain Research.Molecular Brain Research. 62 (1), 1-11.

Perry, P. E. (1983) Induction of sister-chromatid exchanges (SCEs) by thymidine and the potentiation of mutagen-induced SCEs in Chinese hamster ovary cells. Mutation Research. 109 (2), 219-229.

Petersen, M. & Wengel, J. (2003) LNA: a versatile tool for therapeutics and genomics. Trends in Biotechnology. 21 (2), 74-81.

Peterson, A. R., Landolph, J. R., Peterson, H. & Heidelberger, C. (1978) Mutagenesis of Chinese hamster cells is facilitated by thymidine and deoxycytidine. Nature. 276 (5687), 508-510.

Phear, G., Nalbantoglu, J. & Meuth, M. (1987) Next-nucleotide effects in mutations driven by DNA precursor pool imbalances at the aprt locus of Chinese hamster ovary cells. Proceedings of the National Academy of Sciences of the United States of America. 84 (13), 4450-4454.

Page 198: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

198

Phillips, M. D., Nascimbeni, B., Tice, R. R. & Shelby, M. D. (1991) Induction of micronuclei in mouse bone marrow cells: an evaluation of nucleoside analogues used in the treatment of AIDS. Environmental and Molecular Mutagenesis. 18 (3), 168-183.

Physicians' Desk Reference. (2000) Healthcare Ser. 103, .

Pierce, A. J., Hu, P., Han, M., Ellis, N. & Jasin, M. (2001) Ku DNA end-binding protein modulates homologous repair of double-strand breaks in mammalian cells. Genes & Development. 15 (24), 3237-3242.

Pirollo, K. F., Rait, A., Sleer, L. S. & Chang, E. H. (2003) Antisense therapeutics: from theory to clinical practice. Pharmacology & Therapeutics. 99 (1), 55-77.

Potter, C. G. (1971) Induction of polyploidy by concentrated thymidine. Experimental Cell Research. 68 (2), 442-448.

Puri, N., Majumdar, A., Cuenoud, B., Natt, F., Martin, P., Boyd, A., Miller, P. S. & Seidman, M. M. (2002) Minimum number of 2'-O-(2-aminoethyl) residues required for gene knockout activity by triple helix forming oligonucleotides. Biochemistry. 41 (24), 7716-7724.

Puri, N., Majumdar, A., Cuenoud, B., Natt, F., Martin, P., Boyd, A., Miller, P. S. & Seidman, M. M. (2001) Targeted gene knockout by 2'-O-aminoethyl modified triplex forming oligonucleotides. The Journal of Biological Chemistry. 276 (31), 28991-28998.

Radecke, S., Radecke, F., Peter, I. & Schwarz, K. (2006) Physical incorporation of a single-stranded oligodeoxynucleotide during targeted repair of a human chromosomal locus. The Journal of Gene Medicine. 8 (2), 217-228.

Raderschall, E., Stout, K., Freier, S., Suckow, V., Schweiger, S. & Haaf, T. (2002) Elevated levels of Rad51 recombination protein in tumor cells. Cancer Research. 62 (1), 219-225.

Ramaswamy, M. & Yeung, A. T. (1994) Sequence-specific interactions of UvrABC endonuclease with psoralen interstrand cross-links. The Journal of Biological Chemistry. 269 (1), 485-492.

Reed, J. C. (1999) Dysregulation of apoptosis in cancer. Journal of Clinical Oncology : Official Journal of the American Society of Clinical Oncology. 17 (9), 2941-2953.

Reichard, P. (1978) From deoxynucleotides to DNA synthesis. Federation Proceedings. 37 (1), 9-14.

Rubsam, L. Z., Davidson, B. L. & Shewach, D. S. (1998) Superior cytotoxicity with ganciclovir compared with acyclovir and 1-beta-D-arabinofuranosylthymine in herpes simplex virus-thymidine kinase-expressing cells: a novel paradigm for cell killing. Cancer Research. 58 (17), 3873-3882.

Saintigny, Y., Delacote, F., Vares, G., Petitot, F., Lambert, S., Averbeck, D. & Lopez, B. S. (2001) Characterization of homologous recombination induced by replication inhibition in mammalian cells. The EMBO Journal. 20 (14), 3861-3870.

Sambrook, J. (2001) Molecular Cloning: A Laboratory Manual (Third Edition).

Page 199: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

199

Sazani, P. & Kole, R. (2003) Therapeutic potential of antisense oligonucleotides as modulators of alternative splicing. The Journal of Clinical Investigation. 112 (4), 481-486.

Seetharam, S. & Seidman, M. M. (1992) Modulation of ultraviolet light mutational hotspots by cellular stress. Journal of Molecular Biology. 228 (4), 1031-1036.

Seno, T., Ayusawa, D., Shimizu, K., Koyama, H., Takeishi, K. & Hori, T. (1985) Thymineless death and genetic events in mammalian cells. Basic Life Sciences. 31, 241-263.

Shaw, J. P., Kent, K., Bird, J., Fishback, J. & Froehler, B. (1991) Modified deoxyoligonucleotides stable to exonuclease degradation in serum. Nucleic Acids Research. 19 (4), 747-750.

Shen, L. X., Kandimalla, E. R. & Agrawal, S. (1998) Impact of mixed-backbone oligonucleotides on target binding affinity and target cleaving specificity and selectivity by Escherichia coli RNase H. Bioorganic & Medicinal Chemistry. 6 (10), 1695-1705.

Shibahara, S., Mukai, S., Morisawa, H., Nakashima, H., Kobayashi, S. & Yamamoto, N. (1989) Inhibition of human immunodeficiency virus (HIV-1) replication by synthetic oligo-RNA derivatives. Nucleic Acids Research. 17 (1), 239-252.

Sicinski, P., Geng, Y., Ryder-Cook, A. S., Barnard, E. A., Darlison, M. G. & Barnard, P. J. (1989) The molecular basis of muscular dystrophy in the mdx mouse: a point mutation. Science (New York, N.Y.). 244 (4912), 1578-1580.

Song, P. S. & Tapley, K. J.,Jr. (1979) Photochemistry and photobiology of psoralens. Photochemistry and Photobiology. 29 (6), 1177-1197.

Soussi, T. & Lozano, G. (2005) P53 Mutation Heterogeneity in Cancer. Biochemical and Biophysical Research Communications. 331 (3), 834-842.

Spitzer, S. & Eckstein, F. (1988) Inhibition of deoxyribonucleases by phosphorothioate groups in oligodeoxyribonucleotides. Nucleic Acids Research. 16 (24), 11691-11704.

St Clair, M. H., Lambe, C. U. & Furman, P. A. (1987) Inhibition by ganciclovir of cell growth and DNA synthesis of cells biochemically transformed with herpesvirus genetic information. Antimicrobial Agents and Chemotherapy. 31 (6), 844-849.

St Clair, M. H., Richards, C. A., Spector, T., Weinhold, K. J., Miller, W. H., Langlois, A. J. & Furman, P. A. (1987) 3'-Azido-3'-deoxythymidine triphosphate as an inhibitor and substrate of purified human immunodeficiency virus reverse transcriptase. Antimicrobial Agents and Chemotherapy. 31 (12), 1972-1977.

Stadtman, E. R. & et al. (1973) The Enzymes: Group Transfer. USA, ACADEMIC PRESS, INC.

Stein, C. A. & Cohen, J. S. (1988) Oligodeoxynucleotides as inhibitors of gene expression: a review. Cancer Research. 48 (10), 2659-2668.

Stein, C. A., Mori, K., Loke, S. L., Subasinghe, C., Shinozuka, K., Cohen, J. S. & Neckers, L. M. (1988) Phosphorothioate and normal oligodeoxyribonucleotides with 5'-linked

Page 200: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

200

acridine: characterization and preliminary kinetics of cellular uptake. Gene. 72 (1-2), 333-341.

Stein, H. & Hausen, P. (1969) Enzyme from calf thymus degrading the RNA moiety of DNA-RNA Hybrids: effect on DNA-dependent RNA polymerase. Science (New York, N.Y.). 166 (903), 393-395.

Stephenson, M. L. & Zamecnik, P. C. (1978) Inhibition of Rous sarcoma viral RNA translation by a specific oligodeoxyribonucleotide. Proceedings of the National Academy of Sciences of the United States of America. 75 (1), 285-288.

Summerton, J. & Weller, D. (1997) Morpholino antisense oligomers: design, preparation, and properties. Antisense & Nucleic Acid Drug Development. 7 (3), 187-195.

Sun, D., Guo, K., Rusche, J. J. & Hurley, L. H. (2005) Facilitation of a structural transition in the polypurine/polypyrimidine tract within the proximal promoter region of the human VEGF gene by the presence of potassium and G-quadruplex-interactive agents. Nucleic Acids Research. 33 (18), 6070-6080.

Sung, P. & Robberson, D. L. (1995) DNA strand exchange mediated by a RAD51-ssDNA nucleoprotein filament with polarity opposite to that of RecA. Cell. 82 (3), 453-461.

Sussman, H. E., Olivero, O. A., Meng, Q., Pietras, S. M., Poirier, M. C., O'Neill, J. P., Finette, B. A., Bauer, M. J. & Walker, V. E. (1999) Genotoxicity of 3'-azido-3'-deoxythymidine in the human lymphoblastoid cell line, TK6: relationships between DNA incorporation, mutant frequency, and spectrum of deletion mutations in HPRT. Mutation Research. 429 (2), 249-259.

Svoboda, D. L., Taylor, J. S., Hearst, J. E. & Sancar, A. (1993) DNA repair by eukaryotic nucleotide excision nuclease. Removal of thymine dimer and psoralen monoadduct by HeLa cell-free extract and of thymine dimer by Xenopus laevis oocytes. The Journal of Biological Chemistry. 268 (3), 1931-1936.

Takata, M., Sasaki, M. S., Sonoda, E., Morrison, C., Hashimoto, M., Utsumi, H., Yamaguchi-Iwai, Y., Shinohara, A. & Takeda, S. (1998) Homologous recombination and non-homologous end-joining pathways of DNA double-strand break repair have overlapping roles in the maintenance of chromosomal integrity in vertebrate cells. The EMBO Journal. 17 (18), 5497-5508.

Tanaka, M. & Nyce, J. W. (2001) Respirable antisense oligonucleotides: a new drug class for respiratory disease. Respiratory Research. 2 (1), 5-9.

Tanaka, T., Chu, C. C. & Paul, W. E. (1992) An antisense oligonucleotide complementary to a sequence in I gamma 2b increases gamma 2b germline transcripts, stimulates B cell DNA synthesis, and inhibits immunoglobulin secretion. The Journal of Experimental Medicine. 175 (2), 597-607.

te Riele, H., Maandag, E. R. & Berns, A. (1992) Highly efficient gene targeting in embryonic stem cells through homologous recombination with isogenic DNA constructs. Proceedings of the National Academy of Sciences of the United States of America. 89 (11), 5128-5132.

Page 201: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

201

Thust, R. (1976) Screening of potential chemical carcinogens by means of mammalian cells in vitro. Archiv Fur Geschwulstforschung. 46 (7), 538-548.

Thust, R., Schacke, M. & Wutzler, P. (1996) Cytogenetic genotoxicity of antiherpes virostatics in Chinese hamster V79-E cells. I. Purine nucleoside analogues. Antiviral Research. 31 (1-2), 105-113.

Thust, R., Tomicic, M., Klocking, R., Voutilainen, N., Wutzler, P. & Kaina, B. (2000a) Comparison of the genotoxic and apoptosis-inducing properties of ganciclovir and penciclovir in Chinese hamster ovary cells transfected with the thymidine kinase gene of herpes simplex virus-1: implications for gene therapeutic approaches. Cancer Gene Therapy. 7 (1), 107-117.

Thust, R., Tomicic, M., Klocking, R., Wutzler, P. & Kaina, B. (2000b) Cytogenetic genotoxicity of anti-herpes purine nucleoside analogues in CHO cells expressing the thymidine kinase gene of herpes simplex virus type 1: comparison of ganciclovir, penciclovir and aciclovir. Mutagenesis. 15 (2), 177-184.

Tidd, D. M. & Warenius, H. M. (1989) Partial protection of oncogene, anti-sense oligodeoxynucleotides against serum nuclease degradation using terminal methylphosphonate groups. British Journal of Cancer. 60 (3), 343-350.

Todd, A. K. (2007) Bioinformatics approaches to quadruplex sequence location. Methods (San Diego, Calif.). 43 (4), 246-251.

Traut, T. W. (1994) Physiological concentrations of purines and pyrimidines. Molecular and Cellular Biochemistry. 140 (1), 1-22.

Trump, B. F. & Berezesky, I. K. (1987) Ion regulation, cell injury and carcinogenesis. Carcinogenesis. 8 (8), 1027-1031.

Vaerman, J. L., Moureau, P., Deldime, F., Lewalle, P., Lammineur, C., Morschhauser, F. & Martiat, P. (1997) Antisense oligodeoxyribonucleotides suppress hematologic cell growth through stepwise release of deoxyribonucleotides. Blood. 90 (1), 331-339.

Vasquez, K. M., Christensen, J., Li, L., Finch, R. A. & Glazer, P. M. (2002) Human XPA and RPA DNA repair proteins participate in specific recognition of triplex-induced helical distortions. Proceedings of the National Academy of Sciences of the United States of America. 99 (9), 5848-5853.

Vasquez, K. M., Dagle, J. M., Weeks, D. L. & Glazer, P. M. (2001) Chromosome targeting at short polypurine sites by cationic triplex-forming oligonucleotides. The Journal of Biological Chemistry. 276 (42), 38536-38541.

Vasquez, K. M., Wang, G., Havre, P. A. & Glazer, P. M. (1999) Chromosomal mutations induced by triplex-forming oligonucleotides in mammalian cells. Nucleic Acids Research. 27 (4), 1176-1181.

Vousden, K. H. & Lu, X. (2002) Live or let die: the cell's response to p53. Nature Reviews.Cancer. 2 (8), 594-604.

Page 202: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

202

Wang, G., Seidman, M. M. & Glazer, P. M. (1996) Mutagenesis in mammalian cells induced by triple helix formation and transcription-coupled repair. Science (New York, N.Y.). 271 (5250), 802-805.

Wang, Y., Cortez, D., Yazdi, P., Neff, N., Elledge, S. J. & Qin, J. (2000) BASC, a super complex of BRCA1-associated proteins involved in the recognition and repair of aberrant DNA structures. Genes & Development. 14 (8), 927-939.

Ward, J. W. & Duchin, J. S. (1997) The epidemiology of HIV and AIDS in the United States. AIDS Clinical Review. , 1-45.

Washbrook, E. & Fox, K. R. (1994) Comparison of antiparallel A.AT and T.AT triplets within an alternate strand DNA triple helix. Nucleic Acids Research. 22 (19), 3977-3982.

Waters, J. S., Webb, A., Cunningham, D., Clarke, P. A., Raynaud, F., di Stefano, F. & Cotter, F. E. (2000) Phase I clinical and pharmacokinetic study of bcl-2 antisense oligonucleotide therapy in patients with non-Hodgkin's lymphoma. Journal of Clinical Oncology : Official Journal of the American Society of Clinical Oncology. 18 (9), 1812-1823.

Weber, M., Hellmann, I., Stadler, M. B., Ramos, L., Paabo, S., Rebhan, M. & Schubeler, D. (2007) Distribution, silencing potential and evolutionary impact of promoter DNA methylation in the human genome. Nature Genetics. 39 (4), 457-466.

Wickstrom, E. (1986) Oligodeoxynucleotide stability in subcellular extracts and culture media. Journal of Biochemical and Biophysical Methods. 13 (2), 97-102.

Wilton, S. D., Lloyd, F., Carville, K., Fletcher, S., Honeyman, K., Agrawal, S. & Kole, R. (1999) Specific removal of the nonsense mutation from the mdx dystrophin mRNA using antisense oligonucleotides. Neuromuscular Disorders : NMD. 9 (5), 330-338.

Wu, S., Liu, X., Solorzano, M. M., Kwock, R. & Avramis, V. I. (1995) Development of zidovudine (AZT) resistance in Jurkat T cells is associated with decreased expression of the thymidine kinase (TK) gene and hypermethylation of the 5' end of human TK gene. Journal of Acquired Immune Deficiency Syndromes and Human Retrovirology : Official Publication of the International Retrovirology Association. 8 (1), 1-9.

Wu, X. S., Xin, L., Yin, W. X., Shang, X. Y., Lu, L., Watt, R. M., Cheah, K. S., Huang, J. D., Liu, D. P. & Liang, C. C. (2005) Increased efficiency of oligonucleotide-mediated gene repair through slowing replication fork progression. Proceedings of the National Academy of Sciences of the United States of America. 102 (7), 2508-2513.

Wutzler, P. & Thust, R. (2001) Genetic risks of antiviral nucleoside analogues--a survey. Antiviral Research. 49 (2), 55-74.

Xodo, L. E., Manzini, G., Quadrifoglio, F., van der Marel, G. A. & van Boom, J. H. (1991) Effect of 5-methylcytosine on the stability of triple-stranded DNA--a thermodynamic study. Nucleic Acids Research. 19 (20), 5625-5631.

Xu, X. S., Glazer, P. M. & Wang, G. (2000) Activation of human gamma-globin gene expression via triplex-forming oligonucleotide (TFO)-directed mutations in the gamma-globin gene 5' flanking region. Gene. 242 (1-2), 219-228.

Page 203: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

203

Yandell, D. W., Dryja, T. P. & Little, J. B. (1986) Somatic mutations at a heterozygous autosomal locus in human cells occur more frequently by allele loss than by intragenic structural alterations. Somatic Cell and Molecular Genetics. 12 (3), 255-263.

Yazaki, T., Ahmad, S., Chahlavi, A., Zylber-Katz, E., Dean, N. M., Rabkin, S. D., Martuza, R. L. & Glazer, R. I. (1996) Treatment of glioblastoma U-87 by systemic administration of an antisense protein kinase C-alpha phosphorothioate oligodeoxynucleotide. Molecular Pharmacology. 50 (2), 236-242.

Ye, Z., Guntaka, R. V. & Mahato, R. I. (2007) Sequence-specific triple helix formation with genomic DNA. Biochemistry. 46 (40), 11240-11252.

Yi, A. K. & Krieg, A. M. (1998) Rapid induction of mitogen-activated protein kinases by immune stimulatory CpG DNA. Journal of Immunology (Baltimore, Md.: 1950). 161 (9), 4493-4497.

Yoo, B. H., Bochkareva, E., Bochkarev, A., Mou, T. C. & Gray, D. M. (2004) 2'-O-methyl-modified phosphorothioate antisense oligonucleotides have reduced non-specific effects in vitro. Nucleic Acids Research. 32 (6), 2008-2016.

Yu, D., Iyer, R. P., Shaw, D. R., Lisziewicz, J., Li, Y., Jiang, Z., Roskey, A. & Agrawal, S. (1996) Hybrid oligonucleotides: synthesis, biophysical properties, stability studies, and biological activity. Bioorganic & Medicinal Chemistry. 4 (10), 1685-1692.

Yu, R. Z., Geary, R. S., Flaim, J. D., Riley, G. C., Tribble, D. L., vanVliet, A. A. & Wedel, M. K. (2009) Lack of pharmacokinetic interaction of mipomersen sodium (ISIS 301012), a 2'-O-methoxyethyl modified antisense oligonucleotide targeting apolipoprotein B-100 messenger RNA, with simvastatin and ezetimibe. Clinical Pharmacokinetics. 48 (1), 39-50.

Yu, R. Z., Zhang, H., Geary, R. S., Graham, M., Masarjian, L., Lemonidis, K., Crooke, R., Dean, N. M. & Levin, A. A. (2001) Pharmacokinetics and pharmacodynamics of an antisense phosphorothioate oligonucleotide targeting Fas mRNA in mice. The Journal of Pharmacology and Experimental Therapeutics. 296 (2), 388-395.

Zamaratski, E., Pradeepkumar, P. I. & Chattopadhyaya, J. (2001) A critical survey of the structure-function of the antisense oligo/RNA heteroduplex as substrate for RNase H. Journal of Biochemical and Biophysical Methods. 48 (3), 189-208.

Zamecnik, P. C. & Stephenson, M. L. (1978) Inhibition of Rous sarcoma virus replication and cell transformation by a specific oligodeoxynucleotide. Proceedings of the National Academy of Sciences of the United States of America. 75 (1), 280-284.

Zamore, P. D. (2001) RNA interference: listening to the sound of silence. Nature Structural Biology. 8 (9), 746-750.

Zamore, P. D., Tuschl, T., Sharp, P. A. & Bartel, D. P. (2000) RNAi: double-stranded RNA directs the ATP-dependent cleavage of mRNA at 21 to 23 nucleotide intervals. Cell. 101 (1), 25-33.

Zhuang, J. C., Wright, T. L., deRojas-Walker, T., Tannenbaum, S. R. & Wogan, G. N. (2000) Nitric oxide-induced mutations in the HPRT gene of human lymphoblastoid TK6

Page 204: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

204

cells and in Salmonella typhimurium. Environmental and Molecular Mutagenesis. 35 (1), 39-47.

Page 205: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

205

Appendix 1. Buffers and reagents

Annealing buffer 1 50mM sodium chloride

Annealing buffer 2 75mM Tris pH 7.2 and 75mM NaCl

Biotin binding buffer 20mM Tris (pH 7.8), 1M NaCl, 1mM EDTA, 0.02%v/v Triton X-100

Blocking buffer PBS-T wash buffer plus 5%w/v semi-skimmed milk powder.

Prepared fresh on the day

Gel loading buffer (6x) 0.25%w/v bromophenol blue and 40% w/v sucrose.

Stored at 4°C

Master lysis buffer 10μL Halt protease inhibitor + 990μL lysis buffer [150mM NaCl, 0.1%v/v igepal, 1mM Tris (pH 7.4) and 1mM EDTA]

PBS 10 mM phosphate buffer (pH 7.4), 2.7mM potassium chloride and 137mM sodium chloride.

PBS-T wash buffer 1x PBS plus 0.1%v/v Tween-20.

Prepared fresh on the day.

Quadruplex buffer 10mM Tris pH 7.2, 0.1mM MgCl2 and 140mM KCl

SDS loading buffer (5x) 66mM Tris (pH) 6.8, 26%w/v glycerol, 2.1% SDS, 0.01%w/v bromophenol blue.

SDS resolving gel buffer (2.5x)

1.5M Tris (pH 8.8, 0.4%w/v SDS.

Autoclaved and stored at 4°C

SDS running buffer (10x) 25mM Tris (pH 8.3), 250mM glycine and 0.1%w/v SDS.

Stored at 4°C

SDS stacking gel buffer (4x) 0.5M Tris (pH 6.8) and 0.4%w/v SDS.

Autoclaved and stored at 4°C

TBE buffer (1x) 89 mM Tris (pH 8.3), 89mM boric acid and 2 mM EDTA.

Tfi PCR buffer (5x) 200 mM Hepes-KOH (pH 8.1), 75 mM KCl, 75 mM (NH4)2S04, 10% Glycerol and proprietary stabilizers

Transfer buffer (10x) 250mM Tris and 1.86M glycine.

Stored at 4°C

Transfer buffer (1x)

100mL transfer buffer (10x), 200mL 100% methanol and 700mL water.

Prepared on the day of use.

Triplex binding buffer 10mM Tris (pH 7.2) and 10mM MgCl2

Triplex Running buffer 17.8mM Tris (pH 7.2), 17.8mM boric acid and 10mM MgCl2

Urea gel loading buffer 1.5M sucrose, 7M urea, 10mM EDTA, 0.1% bromophenol blue

Urea-TBE loading buffer 1xTBE, 7M urea and 0.1%w/v bromophenol blue

Page 206: Evaluating the genotoxic potential of oligonucleotide … · 2020. 2. 14. · LNA Locked nucleic acid MDM2 Murine double minute 2 mins Minutes mL Millilitres MLA Mouse lymphoma assay

206

Publications

Reshat, R., Priestley, C., and Gooderham N.J. (2012). Paper in review: Mutagenesis by an antisense oligonucleotide and it’s degradation product. Reshat, R., Priestley, C., and Gooderham N.J. (2012). Paper in review: A triple-helix forming oligonucleotide targeting genomic DNA fails to induce mutation. Li, J., Reshat, R., Wu, Q., Ashrafian, H., Bueter, M., le Roux, C.W., Darzi, A., Athanasiou, T., Marchesi, J.R., Nicholson, J.K., Holmes, E., and Gooderham, N.J.(Research Article). Experimental Bariatric Surgery in Rats Generates a Cytotoxic Chemical Environment in the Gut Contents. Front. Microbiol. (2011); 2: 183 Reshat, R., Priestley, C., Fellows,, M., O’Donovan, M. and Gooderham NJ. (Abstract). Is a non-conjugated triple-helix forming oligonucleotide targeting the genomic HPRT locus capable of sequence specific mutagenesis? Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011, Abstract 5413. Reshat, R., Priestley, C., Fellows,, M., O’Donovan, M. and Gooderham NJ (2010). Is a triple-helix forming oligonucleotide targeting a genomic locus capable of sequence specific mutagenesis? Abstract presented at the UK Environmental Mutagen Society (UKEMS) annual meeting. Mutagenesis 25, pp632-633.

Reshat, R., Priestley, C., Fellows,, M., O’Donovan, M. and Gooderham NJ (2009). A Triplex-forming oligonucleotide targeting a genomic locus is capable of sequence specific mutagenesis in human lymphoblastoid TK6 cells. Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010, Abstract 6.