final master thesis revised 11.08.06

79
ABSTRACT Synthesis of Protected Amino Thymidines and New Thiol Derivatives of the Vascular Targeting Agent Combretastatin A-4 Daniel Abel Ramirez Mentor: Robert R. Kane, Ph.D. Protected amino thymidines were synthesized as part of a project aimed at developing reagents for the site-specific, sequence selective DNA cleavage. Modified DNA oligonucleotides have the capacity to behave like artificial restriction enzymes in addition to being agents for the targeted scission of DNA and RNA in antiviral and gene therapy. Sulfur-containing analogues of the vascular targeting agent (VTA) Combretastatin A4 (CA4) were also synthesized. Vascular targeting agents are a class of anticancer compounds made to target and restrict tumor neovasculature. Disruption of these new tumor-associated capillaries prevents the blood flow necessary to feed the tumor, resulting in tumor cell starvation, the build-up of toxins, and massive necrotic cell death. One of the best characterized small molecule vascular targeting agents is Combretastatin A-4 (CA4), which is currently in Phase II clinical trials.

Upload: others

Post on 09-Dec-2021

10 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Final Master Thesis revised 11.08.06

ABSTRACT

Synthesis of Protected Amino Thymidines and New Thiol Derivatives of the Vascular Targeting Agent Combretastatin A-4

Daniel Abel Ramirez

Mentor: Robert R. Kane, Ph.D.

Protected amino thymidines were synthesized as part of a project aimed at

developing reagents for the site-specific, sequence selective DNA cleavage. Modified

DNA oligonucleotides have the capacity to behave like artificial restriction enzymes in

addition to being agents for the targeted scission of DNA and RNA in antiviral and gene

therapy. Sulfur-containing analogues of the vascular targeting agent (VTA)

Combretastatin A4 (CA4) were also synthesized. Vascular targeting agents are a class of

anticancer compounds made to target and restrict tumor neovasculature. Disruption of

these new tumor-associated capillaries prevents the blood flow necessary to feed the

tumor, resulting in tumor cell starvation, the build-up of toxins, and massive necrotic cell

death. One of the best characterized small molecule vascular targeting agents is

Combretastatin A-4 (CA4), which is currently in Phase II clinical trials.

Page 2: Final Master Thesis revised 11.08.06
Page 3: Final Master Thesis revised 11.08.06

Copyright © 2006 by Daniel A. Ramirez

All rights reserved

Page 4: Final Master Thesis revised 11.08.06

iii

TABLE OF CONTENTS

List of Figures v

List of Schemes vi

List of Tables vi

List of Abbreviations vii-viii

Acknowledgments ix-x

Dedication xi

Chapter One: Introduction 1

Project 1: Oligonucleotides 1

Project 2: Thio-combretastatin Analog 4

Chapter Two: Background 7

DNA Cleavage and Oligonucleotides 7

Chapter Three: Materials and Methods 11

Synthesis of 5’-(Monomethoxytrityl)-3’-(2-cyanoethyldiisopropylamino) thymidine

Synthesis of 3’-(Monomethoxytrityl) thymidine

Chapter Four: Results and Discussion 19

Chapter Five: Background 21

Tubulin: A Target for Anti-Cancer Drug Development 21

Colchicine 24

Combretastatins 26

Prodrugs 29

Page 5: Final Master Thesis revised 11.08.06

iv

Chapter Six: Materials and Methods 30

Synthesis of Thiol Combretastatin A-4 Analog

Synthesis of Disulfide Combretastatin A-4 Analogs

Chapter Seven: Results and Discussion 37

Chapter Eight: Conclusions 44

Appendices 46

References 63

Page 6: Final Master Thesis revised 11.08.06

v

LIST OF FIGURES Figure 1: Structures of Modified Thymidine Phosphoramidites previously

developed by the Kane group. 2

Figure 2: DNA Cleavage via an EDTA-linked ODN 2

Figure 3: Diagram of the Proposed DNA Cleavage using two IDA

oligonucleotides 3 Figure 4: Structures of Modified Thymidine Phosphoramidates 3 Figure 5: Various Combrestatin A-4 Analogs that have been developed. 5 Figure 6: Cis and trans isomers of the Thiol Prodrug 6 Figure 7: The Site Specific Cleavage of a 167-mer ODN by a Complementary

Base Pairing, Fe2+ - EDTA tethered 19-Mer. 8 Figure 8: Schematic diagram of the site specific cleavage for the triple helix

sequence of Chromosome III by an oligonucleotide-(EDTA·Fe)2. 9 Figure 9: Compounds 1 and 2. 19 Figure 10: Tubulin dimmer 21 Figure 11: Microtubule Structure and Subunits 22 Figure 12: Role of Tubulin in Cell Division and Interaction with Tubulin Ligands

Colchicine Binding Site 23 Figure 13: Natural and Synthetic ligands that bind to tubulin 24 Figure 14: Structure of Colchicine 25 Figure 15: A Diagrammatic Representation of the Proposed Interaction

of Colchicine with Tubulin. 26 Figure 16: Structures of Combretastatin Series Compounds 28 Figure 17: The Newman-Kwart Rearrangement 37

Page 7: Final Master Thesis revised 11.08.06

vi

Figure 18: Wittig reaction using Sodium Hydride 38 Figure 19: Wittig reaction using Butyllithium 38 Figure 20: Thiol cis-isomer crystal structure 39 Figure 21: Thiol trans-isomer crystal structure 40 Figure 22: Purity of Cis-Isomer determined by HPLC 41 Figure 23: Thiocarbamate deprotection 41 Figure 24: Acetal Protection of Aldehyde 42 Figure 25: Formation of Disulfide 43 Figure 26: Wittig Reaction for Disulfide 43

Page 8: Final Master Thesis revised 11.08.06

vii

LIST OF SCHEMES Scheme 1: Synthesis of 5’-(monomethoxytritylamino)-3’-

(2-cyanoethyldiisopropyl) thymidine phosphoramidite 17 Scheme 2: Synthesis of 3'-(Monomethoxytritylamino)-5'-

(2-cyanoethyldiisopropyl) thymidine phosphoramidite 18

Page 9: Final Master Thesis revised 11.08.06

viii

LIST OF TABLES Table 1: In Vitro Human Tumor Cell Line Assay for Combretastatin A-4. 29 Table 2: Crystal Data for trans and cis-isomers. 40 Table 3: Blood flow in % of the control. 42

Page 10: Final Master Thesis revised 11.08.06

ix

LIST OF ABBREVIATIONS

CA-4 Combretastatin A-4

CA-4P Combretastatin A-4 Prodrug

MMTCl Monomethoxytrityl chloride

MMT Monomethoxytrityl

13C NMR Carbon 13 Nuclear Magnetic Resonance

DMAP N, N-dimethylaminopyridine

DMF Dimethyl Formamide

EDTA Ethylenediaminetetraacetic acid

EtOAc Ethyl Acetate

EtOH Ethanol

GC-MS Gas Chromatography Mass Spectrometry

1H NMR Proton Nuclear Magnetic Resonance

Hz Hertz

J Coupling Constant

MeOH Methanol

MHz Megahertz

Min Minutes

mL Milliliter

mmol Millimoles

n-BuLi n-Butyllithium

Page 11: Final Master Thesis revised 11.08.06

x

ppm Parts per Million

r.t. Room Temperature

hrs. Hours

TsCl Tosyl Chloride

THL Tetrahydrofuran

TLC Thin-Layer Chromatography

μM Micromolar

TEA Triethylamine

p-TsOH para-Toluenesulphonic acid

Page 12: Final Master Thesis revised 11.08.06

xi

ACKNOWLEDGMENTS

I would first like to acknowledge Dr. Bob Kane for his dedication, kindness, and

patience during my studies at Baylor University. I would not have had the opportunities I

have today if it wasn’t for his support and guidance. I deeply appreciate the opportunity

he has given me to pursue my dreams.

I would like to thank Dr. Carlos Manzanares and Dr. Marianna Busch for giving

me a chance to attend Baylor University. I would like to extend my special thanks to Dr.

Garner for his help and support; Dr. Kevin Klausmeyer for his assistance with the X-ray

crystallography.

I would like to thank all my friends in the Kane group, both past and present

members including Dr. John Zhang, Dr. Hui-Min Chang, De Wayne Davidson, and

Aruna Perera for the support they have given me. I would like to thank those in the

chemistry department for their encouragement and support.

I would like to express my appreciation to my committee for spending their

valuable time for this completion.

I would like to extend a special thank you to Tori Strong, David Kuo, Ed Ambros,

and De Wayne Davidson for all the moral support they have given me in and outside the

lab. I would not have made it without their companionship.

Last but not least my family, Jessie, Josephine, Adam, Veronica Ramirez and

Fred Koenigs, for their support, love and encouragement to always do my best. They

have always been there through good times and bad.

Page 13: Final Master Thesis revised 11.08.06

xii

I would like to acknowledge the Department of Chemistry and Biochemistry for

giving me the opportunity to study and provide assistantship during my stay here. I am

also thankful to the Welch Foundation for the funding during the course of study.

Page 14: Final Master Thesis revised 11.08.06

xiii

DEDICATION

To my mother, father and brother

Page 15: Final Master Thesis revised 11.08.06

1

CHAPTER ONE

Introduction

Oligonucleotides An ongoing goal of the Kane research group is to develop reagents for site-

specific, sequence selective DNA cleavage. Appropriately modified

oligodeoxynucleotides (ODNs) have the potential to act as “active antisense” agents that

bind and catalyze the destruction of a target complementary DNA or RNA sequence. An

example of their function is as artificial restriction endonucleases (REs), which cut

nucleotide polymers at sequence-selective points that can’t be cleaved by natural

restriction enzymes. RE’s are enzymes that cut DNA into fragments by recognizing and

cutting pallindromic DNA sequences that are 4 to 6 base pairs in length. These enzymes

used extensively in chromosomal mapping, gene isolation, and DNA sequencing. One

problem with these enzymes is that they are limited in number and scope of recognition

sequences and sites. As such, the need for artificial RE’s that are fine tuned in selectivity

and recognition sequence size has been widely recognized.1

In preliminary work, the Kane group modified a previous approach developed by

Dervan et. al. that utilized a Fe2+ -EDTA tethered complementary DNA oligomer.2 Our

research involved a system utilizing two iminodiacetic acid (IDA) tethered DNA

oligonucleotides (Figure 2).

Page 16: Final Master Thesis revised 11.08.06

2

ON

O

N

O

ON

P

O

O

EtO

EtO

NO

NC

ON

N

O

OOPN

O

CN

N

OO

OEt OEt

A B

Figure 1: Structures of Modified Thymidine Phosphoramidites previously developed by the Kane group.

In this system the two IDA oligonucleotides were, attached by an adjacent

complementary base pairing to a template oligonucleotide to form a site for metal

chelation (Figure 3). The resulting ternary complex could potentially chelate metal ions

in an EDTA-like fashion, hopefully resulting in the metal ion catalyzed, hydrolytic or

oxidative, sequence selective cleavage of the complementary strand.3

5'-Biotin-...G T A C T T A C A------------------------------------G C G T A T A...-3'

3'-C A T G A AT G T NH

N

O

OO-

NFe2+

Point of Cleavage

5' - EDTA modified oligo

DNA Strand

O

O-O

-O

Figure 2: DNA Cleavage via an EDTA-linked ODN2

The Kane group analyzed the ability of the complex in Figure 3 to afford

oxidative DNA cleavage. 5’-IDA modified 13-mer and 3’-IDA modified 12-mer were

Page 17: Final Master Thesis revised 11.08.06

3

bound to 44 and 45-mer DNA substrate templates in the presence of Fe(II) and

dithiothreitol (DTT) as described by Dervan et al.

5'-Biotin-...G T A C T T A C A-------------------------------------------------------A C G T A T A...-3'

3'-C A T G A AT G T

5' - IDA modified oligo made from A

DNA Strand

NT G C A T A T-5'

O

OO-

O-

N

O

O-O

-O

Fe2+

3' - IDA modified oligo made from B

Point of Cleavage

Figure 3: Diagram of the Proposed DNA Cleavage using two IDA oligonucleotides

However, these experiments were inconclusive in demonstrating observable

cleavage with the 2xIDA system.3 In order to study additional structural motifs,

compounds 1 and 2 were chosen as synthetic targets, as they would provide useful 3’ and

5’ amino oligonucleotides.

O

NH

NHN

O

OO

OCH3

PO

NC

N

CNP

N

O

O

O

NHN

O

OHNH3CO

3'-(Monomethoxytrityl)-5'-(2-cyanoethyldiisopropylamino) thymidinephosphoramidite, 2

5'-(Monomethoxytrityl)-3'-(2-cyanoethyldiisopropylamino) thymidine phosphoramidite, 1

Figure 4: Structures of Modified Thymidine Phosphoramidites

Page 18: Final Master Thesis revised 11.08.06

4

Unfortunately, the synthesis of these phosphoramidite derivatives was more

challenging than expected and we were unable to produce useful quantities of the pure

compounds. However, the experience gained during these syntheses facilitated progress

on a second project.

Thio-combretastatin Analog

Cancer is a disease in which abnormal cells divide without control, in many cases

producing a mass of tissue called a malignant tumor. The cancer associated with these

solid masses spreads when cells break away from the malignant tumor and enter the

blood stream. Tumors can be surgically removed or treated with chemotherapeutic

agents or radiation. However, complete recovery is often limited to early stage cancers

and cures are rare. A major group of antitumor agents that have proven to be effective in

the treatment of cancer are the antimitotic drugs.

One target of antimitotic drugs is tubulin, a heterodimeric protein made up of α

and β subunits approximately 50 kDa in size.4 Tubulin polymerizes to form

microtubules, cytosketal polymers essential for intracellular transport and mitosis in all

eukaryotes. Vinca alkaloids, taxoids, colchicine, cryptophycin and combretastatin A-4

are antimitotic agents that are known to have a binding affinity to tubulin. Three well-

characterized sites that the antimitotic agents can bind to on tubulin are commonly called

the colchicine site, the vinca alkaloid site and the taxol site. The combretastatin prodrugs

developed by Professor George R. Pettit (Arizona State University) appear to selectively

target the vascular system of tumors by affecting the microtubules that maintain the shape

of the endothelial cells lining the tumor vasculature.

Page 19: Final Master Thesis revised 11.08.06

5

The use of such vascular targeting agents is a relatively recent approach to the

treatment of solid tumors.5,6 This approach to cancer therapy takes advantage of the

observation that tumors that enlarge beyond a certain mass can no longer be supported by

peripheral blood or by one main blood vessel, and therefore become vascularized.

Disruption of new tumor-associated capillaries (neovascalature) prevents the blood flow

necessary to feed the tumor, resulting in tumor cell starvation, the build-up of toxins, and

massive necrotic cell death. One of the best characterized small molecule vascular

targeting agents is Combretastatin A-4 (CA4) (3).7, 8, 9 While this compound is known to

be an extremely effective inhibitor of the assembly of tubulin into microtubules,10 its

potential as a vascular targeting agent was only revealed after its formulation into the

water-soluble prodrug, disodium phosphate Combretastatin A-4 (CA4DP) (4).11

Although the prodrug 4 is inactive towards tubulin, it is dephosphorylated in the

neovasculature of the tumor to reveal the parent tubulin polymerization inhibiting drug 3

(Figure 5). The resulting disruption of the microtubule skeleton of the endothelial cells in

the neovasculature causes them to lose their characteristic flat shape and become bloated,

thereby occluding the narrow capillaries. The loss of blood flow to the tumor mass results

in extensive necrotic cell death.12

The amine analogue (5) of CA4 is also a very active tubulin

polymerization inhibitor.13 The formulated serine amide 6 shows significant promise as a

vascular targeting agent for the treatment of cancer.14 As is true for the CA4DP prodrug

of CA4, compound 6 is inactive until converted to the parent drug 5. However, the

activation chemistry for the two prodrugs is very different (dephosphorylation for 4, 3

compared to deacylation for 6, 5).

Page 20: Final Master Thesis revised 11.08.06

6

H3CO OCH3

H3CO

OCH3

X

3 X= OH4 X= OPO3Na25 X= NH26 X= NHCH(CO2H)CH2OH7 X= SH

Figure 5: Various Combrestatin A-4 Analogs that have been developed.

This prompted us to investigate the thiol analogue (7) of compounds 3 and 5, and to study

the formulation of this novel compound into reductively activated prodrugs.

H3CO OCH3

H3CO

OCH3

SN

O H3CO

OCH3

H3CO

OCH3

S

NO

8 9

Figure 6: Cis and trans isomers of the Thiol Prodrug In the course of this work a crystalline derivative of compound 8 and its trans

stereoisomer 9 were isolated and structurally characterized in order to unequivocally

confirm their molecular structure.

Page 21: Final Master Thesis revised 11.08.06

7

CHAPTER TWO

Background

Our understanding of DNA and RNA based biomolecules like ribozymes and

DNAzymes has increased due to the considerable amount of research performed. One

major area of study is the development of polynucleotides which can function as

sequence selective chemical nucleases, and particularly, those that derive their activity

from an incorporated metal ion/ligand.2

One common system studied for oligonucleotide cleavage is the iron-mediated

hydroxyl radical generating system often called “Fenton” chemistry.15 Fenton chemistry

comprises the catalyzed reduction of hydrogen peroxide to generate hydroxide anion and

a reactive hydroxyl radical by a redox active metal (M),

Mn+ + H2O2 M(n+1)+ + .OH + OH- (1)

A cyclical reaction will occur in the presence of a metal that can be reduced back to its

active form. This reaction will repeat itself in the presence of hydrogen peroxide and

work as a generator of a hydroxide anion and radical. The superoxide anion is very

effective as a reductant as can be shown in Equation 2.

M(n+1)+ + O2. - Mn+ + O2 (2)

The overall reaction of this process can be seen in Equation 3.

O2. - + H2O2 O2 + .OH + OH- (3)

A very good catalytic system for Fenton chemistry was found to be Fe (II)-EDTA

(ethylenediaminetetraacetic acid) because it is easily reduced by superoxides or ascorbic

Page 22: Final Master Thesis revised 11.08.06

8

acid and is oxidized effectively by hydrogen peroxide, while its compact structure

restricts reactivity with hydroxyl radicals that it generates. These hydroxyl radicals

abstract a deoxyribose hydrogen atom which damages the DNA strand, inducing strand

breakage.16

Using this strategy, Dervan and coworkers harnessed the cleavage ability of Fe2+-

EDTA. Dervan et al illustrated that Fe2+-EDTA tethered via modified thymidine (T*) to

the center of a 19-mer sequence selectively cleaved a single stranded 167-mer with

Fenton like chemistry in the presence of O2 and dithiothreitol (Figure 7).2

3'- -5'

-3'5'-

5'-3'-

-3'-5'

-3'5'-

Fe

-OHO2

T* =

5'- TAACGCAGT*CAGGCACCGT-3'

1.) Heat (denature), anneal2.) Fe (II), DTT

19-mer

167 - mer

73 bases 94 bases

ON

O

N

O

OO

O

N

N O

N O

OEt

N

OO

OEtEtO

Figure 7: The Site Specific Cleavage of a 167-mer ODN by a Complementary Base Pairing, Fe2+-EDTA tethered 19-Mer.2

In an elegant example of utilizing chemistry for the development of artificial

restriction endonucleases (REs), Dervan’s group constructed oligonucleotides, tethered at

Page 23: Final Master Thesis revised 11.08.06

9

both ends with Fe2+ -EDTA that could bind sequence specifically to double stranded

DNA by triple helix formation to produce double stranded cleavage.2

Figure 8: Schematic diagram of the site-specific cleavage for the triple-helix sequence of Chromosome III by an oligonucleotide-(EDTA·Fe)2.17

Dervan’s group synthesized a 20-mer oligonucleotide with Fe2+ -EDTA attached

to both the 5’ and 3’ ends via the derivatized thymidine phosphoramidite in figure 7.

Using this metal-containing oligonucleotide, targeted to a sequence in the 340-kilobase

pair chromosome III of the yeast Saccharomyces cerevisiae, double-strand cleavage was

demonstrated to be limited to a single target site, distinguishing between almost 14

megabase pairs of DNA (Figure 8).17 However, the actual cleavage site was distributed

over several bases around the target site. This suggests that while the gross cleavage site

on the chromosome was well defined, because of the dispersive nature of the hydroxyl

Page 24: Final Master Thesis revised 11.08.06

10

radicals produced and the flexibility of the tether they were unable to achieve extremely

precise DNA cleavage. Other problems in achieving sequence specific DNA cleavage is

the fact that selecting a target sequence is limited by pH, temperature and nature of the

co-solvents. The difference between Dervan’s Fe2+ -EDTA linker our previous IDA

system was that our 2x IDA system is more rigid and as a result could potentially place a

metal ion in such a way that it is able to achieve improved sequence selectivity.

Page 25: Final Master Thesis revised 11.08.06

11

CHAPTER THREE

Materials and Methods

General Section

Chemicals used for synthesis were obtained from Sigma, Aldrich, Fisher (Acros),

Crystal Chem. Inc. and used directly as purchased. All solvents such as ethyl acetate,

hexanes, methylene chloride, methanol, ethanol, tetrahydrofuran were received from

commercial sources and distilled before using. Pyridine and TEA were obtained from

commercial sources in DrySolv bottles and use directly as purchased. When dry

methylene chloride (CH2Cl2) was used, it was distilled over calcium hydride and used

immediately. Tetrahydrofuran (THF) was dried and distilled over potassium metal and

benzophenone according to standard procedure prior to use.

Instrumentation

Proton (1H), carbon (13C) and Phosphorous (31P) NMR were obtained on a Bruker

AVANCE 300 NMR running XWIN-NMR 3.1 software at 300 MHz. Chemical shifts

are expressed in ppm (δ), peaks are listed as singlet (s), doublet (d), triplet (t), or

multiplet (m), with the coupling constant (J) expressed in Hz.

High resolution mass spectra were obtained from the University of California,

Riverside Mass Spectrometry Facility

Page 26: Final Master Thesis revised 11.08.06

12

Synthesis of 5’-(Monomethoxytrityl)-3’-(2-cyanoethyldiisopropylamino) thymidine phosphoramidite

5´-Tosylthymidine (10)3 A solution of thymidine (7.24 g, 30.0 mmol) in 50 ml of dry pyridine was cooled

in an ice bath to 0 °C. A solution of p-tolylsulfonyl choride (9.80 g, 51.5 mmoles) in 50

ml of dry pyridine (also cooled to 0 °C) was added under argon with stirring. The

mixture was allowed to react at 0 °C for over 24 hrs, after which it is decomposed with a

small piece of ice. The mixture was poured into 500 ml of deionized ice water and

extracted three times with 250 mL portions of chloroform. The chloroform was then

washed twice with 200 ml portions of saturated aqueous sodium bicarbonate followed by

two washings of 300 ml of deionized water. The washed chloroform layer was dried over

sodium sulfate and evaporated under reduced pressure to yield a crude brownish solid.

The solid was recrystalized from an ethanol and gave 5.52 g of white crystals (total yield

46 %). The reaction was followed by TLC (1% triethylamine, 10% methanol, methylene

chloride) and visualized by UV and stained with anisaldehyde. Rf thymidine = 0.22, Rf

5´-tosylthymidine = 0.44.

5´-Azidothymidine (11)3 A solution of 0.48 g (1.2 mmoles) 5´-tosylthymidine in 35 mL of deionized water

was reacted with 0.24 g (3.7 mmoles) of sodium azide with stirring under argon for over

12 hours until reaction was complete. The product was then extracted with 3 washings of

100mL of ethyl acetate. The ethyl acetate solution was evaporated off and dried

overnight. The resulting solid was then dissolved in ethanol to which 5.25 g silica gel

was added. The ethanol was then evaporated off to yield crude product adsorbed to the

Page 27: Final Master Thesis revised 11.08.06

13

silica gel. The crude product was then packed into a Biotage FLASH sample injection

module (SIM) cartridge which was then eluted through a KP-SilTMSilica FLASH 40S

cartridge using 3.6% ethanol in methylene chloride using the Biotage FLASH 40

chromatography system. The fractions with product were pooled and evaporated down to

yield 0.32 g product shown pure by TLC and NMR (100 % yield). TLC’s were developed

in 3.6% ethanol in methylene chloride. Plates were visualized by UV light and with

anisaldehyde (5´-tosylthymidine stains black, 5´-azidothymidine stains yellow). Rf 5´-

azidothymidine = 0.17

5´-Aminothymidine (12) A solution of 0.32 g (1.20 mmoles) of 5´-azidothymidine in 70 ml of a

pyridine/water mixture (70/30) was reacted with 0.26 ml (2.64 mmoles) 1, 3-

propanedithiol and 0.37 ml (2.66 mmoles) triethylamine under a static atmosphere of

argon with stirring. It should be noted that when we did the analogous reaction with 3´-

azidothymidine that a solvent of 70:30 v/v pyridine-water (ref. 9 in Bayley paper)

considerably increased the rate of reaction. The reaction generated tiny bubbles of

nitrogen gas and was left stirring for 4 days (we believe that this ensures the oxidation

and precipitation of all the excess dithiol, which appears as a white disulfide powder that

coats the walls of the reaction vessel). The solution was filtered to remove the disulfide

and rotovapped down to give a white precipitate. This was dissolved in deionized water

upon which a further amount of disulfide precipitates and was filtered out. The water

solution is then washed three times with 150 ml of methylene chloride and then

rotovapped to dryness to yield 1.02 g (94 % yield) of an off-white product. The 5´-

aminothymidine rection was followed by TLC with 10% methanol in methylene chloride.

Page 28: Final Master Thesis revised 11.08.06

14

5’-Monomethyoxytritylamino thymidine (13) 5’-aminothymidine (1.0 g, 4.1 mmol) was washed three times with pyridine and

filtered. Under reduced pressure the material was dried overnight with a drying tube

while flushing with nitrogen. The dried 5’-aminothymidine was then dissolved in

anhydrous pyridine under nitrogen. At room temperature monomethoxytrityl chloride

(1.9 g, 6.2 mmol), triethylamine (4.3 ml, 30.8 mmol) and 4-dimethylaminopyridine

(0.025g, 0.21mmol) were added to the pyridine mixture, turning the solution a dark

green. After stirring for 30 minutes the solution turned black in color. The reaction

mixed for additional 3.5 hours and followed by TLC with a methylene chloride/ TEA

mixture (9.5/0.5, Rf = 0.25). Once the reaction was completed, it was then concentrated

under reduced pressure overnight and flashed chromatographed using a Biotage Flash

column system. The final product was a light-brown powder (1.8 g, 86% yield):

1H NMR (300 MHz, CDCl3, δ of residual CHCl3 set to 7.26): (δ, ppm): 7.46 (s, 1 H, J =

7.1 Hz), 7.36 (d, 4 H, J = 9.0 Hz), 7.29 (t, 2 H, J = 7.4 Hz), 7.20 (d, 4 H, J = 7.2 Hz), 7.05

(d, 2 H, J = 1.2 Hz), 6.82 (d, 2 H, J = 9.0 Hz), 6.27 (t, H, J = 6.5 Hz), 4.32 (m, H), 3.97

(m, H), 3.78 (s, OCH3), 3.75 (m, H), 2.36 (m, 2H), 2.12 (m, H), 1.85 (d, CH3, J = 1.2 Hz)

13C NMR (300MHz, CDCL3, δ of residual CHCl3 set to 7.26): (δ, ppm): 163.7, 157.9,

150.3, 145.9, 137.7, 135.1, 129.7, 128.4, 127.9, 126.4, 113.2, 111.1, 86.0, 84.3, 72.1,

70.2, 63.2, 55.2, 52.9, 46.1, 45.9, 40.3, 12.5, 9.6 and 7.9.

Synthesis of 5’-(monomethoxytritylamino)-3’-(2-cyanoethyldiisopropyl) thymidine phosphoramidite (1) 5’-(monomethoxytrityl) thymidine was dried in a dessicator with phosphorus

pentoxide for two days. The trityl compound (0.185 mmol, 95 mg) was dissolved in

Page 29: Final Master Thesis revised 11.08.06

15

anhydrous CH2Cl2 (4 ml) and cooled to 0°C and left to stir for 10 minutes under nitrogen.

Hunig’s base (120 μl) was added followed by 2-cyanoethyl-N, N-

diisopropylchlorophosphoramidite (0.268 mmol, 63 mg, 60 μl) was then added. The

reaction stirred at 0°C for 15 minutes and then allowed to stir for 1 hour at room

temperature. The reaction was followed by TLC and upon completion the reaction was

concentrated under reduced pressure and immediately chromatographed (anhydrous ethyl

ether containing 2%TEA). The final product was a brown gel (20 mg, 15 % yield):

1HNMR (300 MHz, DMSO-d6, δ of residual DMSO set to 2.48 ppm): (δ, ppm): 7.49 (s,

1H), 7.41 (d, aromatic 4H, J = 7.6 Hz), 7.30 (d, aromatic 2H, J = 8.8 Hz), 7.27 (d,

aromatic 2H, J = 7.4 Hz), 7.17 (t, aromatic 2H, J = 7.22 Hz), 6.83 (d, aromatic 2H, J = 8.8

Hz), 6.12 (t, H, J = 6.8 Hz), 4.51 (m, H), 3.98 (m, H), 3.86 (m, 2H), 3.75 (m, 2H), 3.71 (s,

CH3), 3.64 (m, H), 3.55 (m, H), 2.82 (t, 2H, J = 5.8 Hz), 2.30 (m, 2H), 1.69 (s, CH3), 0.94

(m, 12H)

13C NMR (300 MHz, DMSO-d6, δ of residual DMSO set to 39.43): (ppm): 163.5, 157.3,

150.2, 146.1, 137.6, 136.1, 129.5, 128.2, 127.6, 126.0, 118.7, 112.9, 109.5, 83.6, 73.5,

69.7, 58.3, 55.5, 54.8, 45.6, 42.5, 42.4, 24.2, 19.7 and 11.9.

31P NMR (300MHz, DMSO-d6): (ppm): 147.21

Synthesis of 3’-aminothymidine (14) A solution of 0.50 g (1.9 mmoles) of 3´-azidothymidine in 150 ml of methanol

mixture (70/30) was reacted with 0.38 ml (3.8 mmoles) of 1, 3-propanedithiol and 0.53

ml (3.8 mmoles) triethylamine under a static pressure of argon with stirring. The reaction

generated tiny bubbles of nitrogen gas and was left stirring for 4 days. The solution was

filtered to remove the disulfide and rotovapped down to give a white solid. This solid

Page 30: Final Master Thesis revised 11.08.06

16

was then dissolved in deionized water upon which a further amount of disulfide

precipitated and was filtered out. The aqueous solution was then washed three times with

150 ml of methylene chloride and then the water was removed by evaporation to yield

0.45 g (98 % yield) of off-white product. The 3´-aminothymidine reaction was followed

by TLC with 10% methanol in methylene chloride.

Synthesis of 3’-Monomethoxytritylamino thymidine (15) In a similar reaction to produce compound 13, 3’-aminothymidine (1.86 mmol,

0.450 g) was washed three times with pyridine and dried overnight with a drying tube

while flushing with nitrogen and co-evaporating with pyridine. The material was then

dissolved in anhydrous pyridine under nitrogen. At room temperature

monomethoxytrityl chloride (0.861 g, 2.79 mmol), triethylamine (14.0 mmol, 1.96 mL)

and 4-dimethylaminopyridine (0.011 g, 0.093 mmol) were added to the mixture turning

the solution a dark green. After stirring for 30 minutes the solution turned black in color.

The reaction was left to mix for additional 3.5 hours and followed by TLC with a

methylene chloride/ TEA mixture (9.5/0.5, Rf = 0.25). Upon completion the reaction was

then concentrated under reduced pressure overnight and flashed chromatographed using a

Biotage Flash column system. The final product was a light brown powder (0.600 g,

86% yield):

1H NMR (300 MHz, CDCl3, δ of residual CHCl3 set to 7.26): (δ, ppm): 7.51(d, 1 H, J =

8.2 Hz), 7.41(d, 4 H, J = 6.8 Hz), 7.29 (t, 2 H, J = 7.4 Hz), 7.21 (d, 4H, J = 6.11 Hz), 7.17

(s, H), 6.82 (d, 2H, J = 8.94 Hz), 6.03 (t, H, J = 6.14 Hz, 6.68 Hz), 3.86 (dd, H, J = 12.0,

2.2), 3.78 (s, OCH3), 3.66 (dd, H, J = 12.0, 3.4), 3.33 (m, 3H), 2.08 (s, OH), 1.96 (s, NH),

1.83 (s, CH3)

Page 31: Final Master Thesis revised 11.08.06

17

13C NMR (300 MHz, CDCl3, δ of residual CHCl3 set to 7.26): (δ, ppm): 163.5, 158.2,

150.2, 146.3, 138.1, 136.0, 129.8, 128.5, 128.1, 126.7, 113.4, 110.8, 86.6, 84.9, 70.7,

62.1, 55.2, 53.2, 40.0 and 12.5.

(R)(R)

O(R)

OH

NH

N

O

OTsO

(Z)

(R)(R)

O(R)

OH

NH

N

O

OHO

(Z)

(R)(R)

O(R)

OH

NH

N

O

ON3

(Z)

(S)(R)

O(R)

OH

NH

N

O

OH2N

(Z)

(R)(R)

O(R)

OH

NHN

O

OHNH3CO

(Z)

PN

OCN

(R)(R)

O(R)

O

NHN

O

OHNH3CO

(Z)

ClH3CO

N

NCH3H3C

P

N

OCN

ClN

CH3CH3

CH3H3C

H3C

H3C SO

OCl

11

5'-(Monomethoxytritylamino)-3'-(2-cyanoethyldiisopropyl) thymidine phosphoramidite: 1 (15 % yield)

13

10Thymidine

5 eq. NaN3

H2O

100 oC, reflux

2 eq. HSCH2CH2CH2SH2 eq. (CH3CH2)3N

pyridine/water(70/30)

12

1.5 eq.

1.5 eq. (CH3CH2)3N

pyridine

0oC - RT

0.05 eq.

dry CH2Cl2

1.4 eq

46% yield 33 % yield

94 % yield86 % yield

Scheme 1: Synthesis of 5’-(monomethoxytritylamino)-3’-(2-cyanoethyldiisopropyl) thymidine phosphoramidite

Page 32: Final Master Thesis revised 11.08.06

18

(R)(R)

O(S)

NH

N

O

OHO

NH

OCH3

(Z)

(R)(R)

O(S)

NH2

NH

N

O

OHO

(Z)

(R)(R)

O(S)

N3

NH

N

O

OHO

(Z)

P O

CN

(R)(R)

O(S)

NH

N

O

OO

N

NH

OCH3

(Z)

15

3'-(Monomethoxytritylamino)-5'-(2-cyanoethyldiisopropyl) thymidine phosphoramidite, 2

3'- azidothymidine 14

ClH3CO

N

NCH3H3C

1.5 eq.

1.5 eq. (CH3CH2)3N

0.05 eq.

P

N

OCN

Cl

dry CH2Cl2

1.4 eq

NCH3

CH3

CH3H3C

H3C

2 eq. HSCH2CH2CH2SH2 eq. (CH3CH2)3N

methanol

86% yield

98 % yield

Scheme 2: Synthesis of 3'-(Monomethoxytritylamino)-5'-(2-cyanoethyldiisopropyl) thymidine phosphoramidite

Page 33: Final Master Thesis revised 11.08.06

19

CHAPTER FOUR

Results and Conclusion

The goal of my oligonucleotide research was to synthesize compounds 1 and 2,

which could be useful in the development of novel functional oligonucleotides single

base specific cleavage.

O

NH

NHN

O

OO

OCH3

PO

NC

N

CNP

N

O

O

O

NHN

O

OHNH3CO

3'-(Monomethoxytritylamino)-5'-(2-cyanoethyldiisopropyl) thymidinephosphoramidite, 2

5'-(Monomethoxytritylamino)-3'-(2-cyanoethyldiisopropyl) thymidine phosphoramidite, 1

Figure 9: Compounds 1 and 2.

Different changes to the original synthesis were attempted to increase the overall

yield. One problem that was prevalent in the original synthesis of 5’-azidothymidine was

the use of DMF as the solvent. Because of the nature of DMF it was difficult to remove

all the solvent from the 5’-azidothymidine product. As a result DMF could be seen by

NMR throughout the rest of the synthesis. In order to avoid this problem an azeotrope

Page 34: Final Master Thesis revised 11.08.06

20

method was incorporated which produced a more pure product but with a lower yield.

Despite the low yield, in that step, the biggest setback in the overall synthesis was the

decomposition of the final product and the instability of the reagent-2-

cyanoethyldiisopropyl phosphorus chloride. One of the problems was that the reagent

would decompose rapidly even when kept in the freezer. Additionally, the final product

would further decompose while in the column which made it difficult to get any

measurable quantity to characterize or to proceed further. Even when the column was ran

at a faster rate the decomposition was still a problem. We were able to get enough of the

5’-(Monomethoxytritylamino)-3’-(cyanoethyldiisopropyl) thymidine phosphoramidite to

characterize by NMR but unfortunately not enough of the product was isolated to proceed

with the synthesis of modified oligonucleotides. In the end we learned a variety of

chromatography techniques, as well as synthetic techniques that could be applied to other

synthesis.

Page 35: Final Master Thesis revised 11.08.06

21

CHAPTER FIVE

Background

In a recent paper in Nature Review, Mary Ann Jordon wrote “Highly dynamic

mitotic-spindle microtubules are among the most successful targets for anticancer

therapy”.18 The major component of microtubules is tubulin, which is a heterodimeric

protein that consists of two polypeptides subunits α-tubulin and β-tubulin with

dimensions of 4nm x 5nm x 8nm and approximately 100,000 daltons.4 The structures of

these subunits are similar, as can be seen in Figure 1.

Figure 10: Tubulin dimer19

Polymerization of tubulin results in microtubules that are the essential

cytoskeleton polymers that effect cell shape, cell transport and cell division. The initial

Page 36: Final Master Thesis revised 11.08.06

22

stage of microtubule formation is called nucleation, where the alpha and beta tubulin

molecules join to form a heterodimers. In the second stage (elongation) the heterodimers

then attach to other dimers in a head to tail fashion to form protofilaments which then

join together to form a microtubule. Each microtubule contains 13 protofilaments and is

20-25 nm in diameter (Figure 11).4

Figure 11: Microtubule Structure and Subunits20

The rate of polymerization differs at the two ends of the microtubule, as assembly

and disassembly occurs at both the plus and minus ends of the microtubule. This

behavior is known as dynamic instability, which is a process that allows the ends of the

microtubule to switch phases between growth and shortening. The plus end is where the

microtubule rate of growth is the fastest. The assembly and disassembly of the

microtubule is a result of the binding and hydrolysis of guanosine triphosphate (GTP) by

the tubulin subunits. Each α/β-tubulin subunit is bound together by one molecule of GTP

which is non-exchangeable. However, the GTP that is attached to the beta monomer is

exchangeable. Due to this exchangeability when the tubulin subunits bind to the

microtubule the GTP is hydrolyzed to guanosine diphosphate (GDP). Upon release of the

Page 37: Final Master Thesis revised 11.08.06

23

GTP-tubulin subunits, depolymerization results and the tubulin subunits exchange GDP,

on beta tubulin, for GTP and can undergo another round of polymerization.

Prevention of the formation of microtubules is the focus of a significant body of

cancer research. There are three well characterized ligand binding sites found in

microtubules; the taxane site, the vinca alkaloid site, and the colchicine binding site. It

has been shown that each binding site has an influence on a different part of mitosis. The

taxane site effects the depolymerization of microtubules, where as the vinca alkaloid and

colchicine sites effects the polymerization of microtubules. By preventing

polymerization or depolymerization the ability for cancer cells to reproduce is affected,

thereby causing a reduction in cellular division.

Figure 12: Role of Tubulin in Cell Division and Interaction with Tubulin Ligands Colchicine Binding Site21

There have been many drugs developed or discovered in nature that disrupt

microtubule polymerization by binding to tubulin (Figure 13).22 These antimitotic drugs

Page 38: Final Master Thesis revised 11.08.06

24

bind to either of the three binding sites of tubulin; the colchicine site, the vinca alkaloid

site, or the taxoid site.

OCH3

H3CO

H3CO

OCH3

OH

O

H3COOCH3

H3CO

OCH3

OH

Combretastatin A-422 Phenstatin23

H3COH3CO

H3CO

OCH3

O

NHCOCH3

O

OO

OHH

H O

H

H3COOCH3

OCH3

Colchicine24 Podophyllotoxin25

N

H O

R

H

R OOAc

HOBz

OHO

OHAcO

O

Ph

NH

OH

Ph

O O

3-Formyl-2-phenylindoles26 Taxol27

N

S

O OH O

O

OH

Me

NR'H

O

R

Epothilone B28 2-phenyl-4-quinolones29

Figure 13: Natural and Synthetic ligands that bind to tubulin

Of special interest is the colchicine binding site. Colchicine (Figure 14) is a

natural product that can be extracted from the meadow saffron, and which has historically

Page 39: Final Master Thesis revised 11.08.06

25

been used as a treatment for gout. Although each molecule of colchicine binds tightly to

tubulin and prevents polymerization, once tubulin has polymerized into a microtubule

colchicine will not have an effect upon it. The rapid disappearance of the mitotic spindle

caused by colchicine indicates a chemical equilibrium between the spindle microtubules

and the free tubulin.4 The site on β-tubulin which colchicine binds is termed the

“colchicine binding site” and is located at the interface of the α / β subunits of tubulin.

Antimitotic drugs that bind to the colchicine site block tubulin polymerization.18 The fact

that colchicine is a powerful antitumor agent its use in the clinic is limited because of its

toxicity.

H3COH3CO

H3CO(S)

(E)

(E)

(E)

O

OCH3

NHCOCH3A B

C

Figure 14: Structure of Colchicine

A novel “footprinting” method developed by Chaudhuri30 and coworkers allows

the drug-binding sites as well as the domain of tubulin affected by drug-induced

conformational changes to be determined. In this method, tubulin is treated with

concentrations of urea low enough to allow the ligand to bind to tubulin, yet high enough

to loosen the conformation of the protein so that any cysteine residue not affected by the

ligand can react with unlabeled N-ethylmaleimide. Afterwards, the ligand is removed

and tubulin is reacted with labeled N-ethylmaleimide to identify the residues which were

blocked when the ligand was originally bound. It was determined in this study that

Page 40: Final Master Thesis revised 11.08.06

26

colchicine binds to tubulin at the α/β interface with the B-ring on the α-subunit and the A

and C-rings on the β-subunit. In addition, the B-ring of the colchicine plays a major role

in the stability of colchicine with little effect on the A and C-rings.

In a similar study done by Hamel31 and coworkers, cysteine 354 of β-tubulin was

determined to be the binding site for the A ring of colchicine (Figure 14). In this study 3-

chloroacetyl-3-dimethylthiocolchicine (3CTC), an analog of colchicine and a competitive

inhibitor of the binding of colchicine to tubulin, was reacted with β-tubulin containing

radiolabeled peptides. Once these peptides were cleaved from the intact protein and

isolated it was determined that a strong covalent reaction between [14C]3CTC and β-

tubulin occurred at cysteine 354. Conveniently, the calculated length of the chloroacetyl

moiety in 3CTC is 3 Å, and it was determined that there is a 3 Å distance between the C-

3 oxygen of colchicine and the Cys-354 sulfur atom. This suggests that the colchicine A

ring lies somewhere between Cys-354 and Cys-239 which is known to have a distance of

9 Å.

Combretastatins Combretastatins are antimitotic agents isolated and characterized by Pettit and

coworkers in 1982 from the bark of the South Afican bush willow Combretum caffrum.32

A series of naturally occurring combretastatins (Figure 15)28-32 were isolated and shown

to function as antimitotic agents. The combretastatin A series has been the focus of much

research, especially the combretastatins A-1, A-2 and A-4. These simple cis-stilbenes

differ according to various substituents on the A and B-rings. The combretastatin B

series consist of two aryl rings connected by a saturated hydrocarbon bridge.

Page 41: Final Master Thesis revised 11.08.06

27

Figure 15: A Diagrammatic Representation of the Proposed Interaction of Colchicine with Tubulin31

Combretastatin C is a single compound which is similar to combretastatin A-1

with a unique tricyclic core. The D series has an ether linkage that connects the two aryl

rings which are joined together by a macrocyclic lactone. Even though these compounds

are similar in structure to colchicine they are more attractive as anti-cancer targets

because of their decreased toxicity to normal healthy cells.

Among the most potent compounds in the combretastatin family is the

combretastatin A-4 (CA-4). This compound inhibits the polymerization of tubulin into

microtubules by binding to the colchicine site on β-tubulin. CA-4 is similar to colchicine

in that it contains two aromatic rings connected by a hydrocarbon bridge in which one

aryl ring has a 3, 4, 5-trimethoxyphenyl moiety. CA-4 has shown to have excellent

activity as an anti-tubulin agent with an inhibition constant (IC50) of 2-3uM for

inhibition of tubulin assembly.33 The cytotoxic properties of CA4 can be seen in the

following table.

Page 42: Final Master Thesis revised 11.08.06

28

H3COH3CO

H3CO

OH

OH

OCH3

H3COOH

OCH3

O

O

Combretastatin A-131 Combretastatin A-232

H3COH3CO

H3CO

OH

OH

OCH3

Combretastatin B-131 Combretastatin C-134

OH3CO

H3CO

OHOH

O

O

O

O

O

OH

Combretastatin D-135

O

O

O

OH

Combretastatin D-236

Figure 16: Structures of Combretastatin Series Compounds

Extensive SAR studies have demonstrated that one of the requirements for these

compounds is to have the Z-geometry for the stilbenoid system. The E-configuration

shows a considerable drop in antitubulin activity and cancer cell inhibition.33

In addition, the 3, 4, 5-trimethoxyphenyl substitution and the 3-hydroxy-4-

methoxy rings are both vital parts of CA-4 binding to the colchicine site on tubulin.37

From molecular recognition studies performed by Dr. Pinney’s group at Baylor

Page 43: Final Master Thesis revised 11.08.06

29

University, it was determined that the distance between the aryl rings of CA-4 is

important for tubulin polymerization inhibition.

Table 1: In Vitro Human Tumor Cell Line Assay for Combretastatin A-4.23

Cell Type Cell Line GI50

(�g/mL)

Ovarian OVCAR-3 <1.0 x 10-3 CNS SF-295 <1.0 x 10-3

Lung-NSC NCI-H460 5.6 x 10-4 Colon KM20L2 6.1 x 10-2

Melanoma SK-MEL-5 2.0 x 10-4 Pancreas BXPC-3 3.9 x 10-1 Pharynx FADU 6.5 x 10-4 Thyroid SW1736 7.1 x 10-4 Prostate DU-145 7.6 x 10-3

Prodrugs Despite the exciting antitubulin activity of CA-4, its availability in aqueous

environments is unfortunately low. Therefore, modifications to CA-4 were necessary to

achieve an appropriate prodrug. The prodrug form of CA-4, combretastatin A-4

phosphate disodium salt (CA-4P)11 was developed by the functionalization of the free

hydroxyl moiety. Prodrugs are agents that are transformed after administration, either by

metabolism or spontaneous chemical breakdown, to form a pharmacologically active

species.36 Once in the bloodstream enzymes convert the water-soluble prodrug into the

fat-soluble active form of the drug. This activation chemistry for the CA4 prodrug

prompted us to investigate the thiol analogue (8) CA4, and to study the formulation of

this novel compound into reductively activated prodrugs. In the course of this work a

crystalline derivatives of compound 8 and its trans stereoisomer 9 were isolated and

structurally characterized in order to unequivocally confirm their molecular structure.

Page 44: Final Master Thesis revised 11.08.06

30

CHAPTER SIX

Materials and Methods

General Section Chemicals used for synthesis were obtained from Sigma, Aldrich, Fisher (Acros),

BACHEM and used directly as purchased. All solvents such as ethyl acetate, hexanes,

methylene chloride, methanol, ethanol, tetrahydrofuran were received from commercial

sources and distilled before using. When dry methylene chloride (CH2Cl2) was used it

was distilled over calcium hydride and used immediately. Tetrahydrofuran (THF) was

dried and distilled over potassium metal and benzophenone according to standard

procedure prior to use.

Instrumentation

Proton (1H), carbon (13C) and Phosphorous (31P) NMR were obtained on a Bruker

AVANCE 300 NMR running XWIN-NMR 3.1 software at 300 MHz. Chemical shifts

are expressed in ppm (δ), peaks are listed as singlet (s), doublet (d), triplet (t), or

multiplet (m), with the coupling constant (J) expressed in Hz.

3-(N, N-dimethylthiocarbamoyloxy)-4-methoxybenzaldehyde38 (16) A solution of N, N-dimethylcarbamoyl chloride (4.07 g, 32.9 mmol) in THF was

added dropwise at 0° C to a solution of 3-Hydroxy-4-methoxybenzaldehyde (5.03 g, 32.9

mmol) and potassium hydroxide (1.84 g, 32.9 mmol) in water (22 mL). A white

precipitate was filtered using suction filtration, washed with water and dried by vacuum.

Page 45: Final Master Thesis revised 11.08.06

31

Yield 6.72 g (85 %) of 3-(N, N-dimethylthiocarbamoyloxy)-4-methoxybenzaldehyde.

1H NMR (300 MHz, CDCl3, δ of residual CHCl3 set to 7.26 ppm): (δ, ppm): 9.86 (s, 1H,

CHO), 7.77 (dd, H, J = 8.5, 2.0 Hz), 7.57 (d, H J = 2.0 Hz), 7.06 (d, H, J = 8.5 Hz), 3.90

(s, OCH3), 3.45 and 3.35 (2s, N(CH3)2).

13C NMR (300 MHz, CDCl3, δ of residual CHCl3 set to 77.0 ppm): (ppm): 38.8, 43.5,

56.3, 112.2, 124.9, 129.8, 143.2, 156.8, 187.2 and 190.1.

3-(N, N-dimethylcarbamoylthio)-4-methoxybenzaldehyde39 (17) A solution of 3-(N, N-dimethylcarbamoylthio)-4-methoxybenzaldehyde (4.93 g,

20.6 mmol) in 200 mL of diphenyl ether was heated to 245 -250 °C under argon for 2 hr

10 min. The mixture was cooled to room temperature then added to 1L of hexanes and

the precipitate formed was collected by suction filtration, washed with hexanes and dried

by vacuum. Yield 3.77 g (76.5 %) of 3-(N, N-dimethylcarbamoylthio)-4-

methoxybenzaldehyde as a light brown precipitate.

1H NMR (300 MHz, CDCl3, δ of residual CHCl3 set to 7.26 ppm): (δ, ppm): 9.87 (s, 1H

CHO), 7.99 (d, 1H J = 2.1 Hz), 7.94 (dd, 1H J = 8.53, 2.16 Hz), 7.06 (d, 1H J = 8.54 Hz),

3.95 (s, OCH3), 3.08 (2 broad s, N(CH3)2).

13C NMR (300 MHz, CDCl3, δ of residual CHCl3 set to 77.0 ppm): (ppm): 190.1, 165.4,

164.8, 140.3, 133.2, 130.0, 118.4, 111.5, 56.6 and 37.0.

3, 4, 5-Trimethoxybenzyl-triphenylphosphonium bromide40 (18) To a solution of CBr4 (22.9 g, 69.0 mmol) in anhydrous acetone (170 mL) at 0 °C

under argon, 3, 4, 5-trimethoxybenzyl alcohol (10.0 g, 50.4 mol) and triphenylphosphine

(17.99 g, 68.0 mmol) were added. The reaction was left overnight and then the solvent

Page 46: Final Master Thesis revised 11.08.06

32

was removed under reduced pressure to obtain 3, 4, 5-trimethoxybenzyl bromide crude as

a brown gel. The crude product was dissolved in toluene (200 mL) and

triphenylphosphine (14.54 g, 55 mmol) was added. The reaction was refluxed for 2

hours. After refluxing the reaction was allowed to stir overnight, and a sticky orange

precipitate was formed. The solvent was removed under reduced pressure and the

precipitate was dried under vacuum. Once the material was dry, it was recrystalized

using ethanol in the cold overnight. The solvent was then removed under reduced

pressure and the white crystals were dried under vacuum. Yield 17.64 g (66.8 %) of 3, 4,

5-trimethoxybenzyltriphenylphosphonium bromide.

(E/Z)-1-(3'-N, N-dimethylthiocarbamoyl-4'-methoxy)-2-(3", 4", 5"-trimethoxyphenyl) ethane (3 and 4) A well stirred solution of 3, 4, 5-trimethoxybenzyltriphenylphosphonium bromide

(4.37 g, 8.35 mmol) and n-BuLi (0.535 g, 8.36 mmol) in THF, 3-(N,N-

dimethylcarbamoylthio)-4-methoxybenzaldehyde (1.00 g, 4.18 mmol) was added drop

wise at -15 °C. Once at room temperature the reaction was followed by thin-layer

chromatography. When reaction was complete the mixture was quenched with ice-cold

water. The products were then extracted with ether. The etherate solution was then

washed with ice-cold water and dried over Na2SO4. Ether was removed in vacuo and the

resulting brownish gel was subjected to flash chromatography (silica gel, 20%

EtOAc/60% hexanes), in order to obtained the E-isomer as a pale-yellow crystalline

solid. (Z: E, 2:1 as determined by integration of 1H NMR signals). Yield 0.71 g (42 %)

of Z-1-(3'-N, N-dimethylthiocarbamoyl-4'-methoxy)-2-(3", 4", 5"-trimethoxyphenyl)

Page 47: Final Master Thesis revised 11.08.06

33

ethane and 0.34 g (21 %) of E-1-(3'-N, N-dimethylthiocarbamoyl-4'-methoxy)-2-(3", 4",

5"-trimethoxyphenyl) ethane.

1H NMR Z-isomer (300 MHz, CDCl3, δ of residual CHCl3 set to 7.26 ppm): (δ, ppm):

7.44 (d, 1H, J = 2.3 Hz), 7.31 (dd, 1H, J = 8.6, 2.3 Hz), 6.82 (d, 1H, J = 8.6 Hz), 6.51 (s,

2H), 6.45 (d, 1H, J = 12.4 Hz), 6.40 (d, 1H, J = 12.2 Hz), 3.84 (s, 3H), 3.82 (s, 3H), 3.70

(s, 6H), 3.03 (broad, N(CH3)2).

13C NMR Z-isomer (300 MHz, CDCl3, δ of residual CHCl3 set to 77.0 ppm): (ppm): 14.2,

21.1, 37.0, 56.0, 56.2, 60.4, 60.9, 105.8, 111.1, 116.6, 128.6, 129.2, 130.0, 132.3, 132.6,

137.1, 138.5, 153.0, 159.3, and 165.9.

1H NMR E-isomer (300 MHz, CDCl3, δ of residual CHCl3 set to 7.26 ppm): (δ, ppm):

7.64 (d, 1H, J = 2.3Hz), 7.51 (dd, 1H, J = 8.6 Hz, J = 2.3 Hz), 6.94 (d, 1H, J = 8.6 Hz),

6.93 (d, 1H, J = 16.1 Hz), 6.89 (d, 1H, J = 16.1 Hz), 6.69 (s, 2H), 3.90 (s, 6H), 3.89 (s,

3H), 3.85 (s, 3H), 3.11 (broad, N(CH3)2).

13C NMR Z-isomer (300 MHz, CDCl3, δ of residual CHCl3 set to 77.0 ppm): (ppm): 37.1,

56.1, 56.3, 61.0, 103.4, 111.7, 117.3, 127.0, 127.4, 129.5, 130.5, 133.3, 136.0, 137.8,

153.4 and 159.6.

(Z/E)-1-(3'-thiol-4'-methoxy)-2-(3", 4", 5"-trimethoxyphenyl) ethane (9) A solution of 3 (0.640 g, 1.58 mmol) in THF was collected to -42 °C and allowed

to mix for 15 minutes. To the cooled mixture LiAlH4 (0.36 g, 9.48 mmol) was added in

portions over a 10 min period. As the LiAlH4 was added the mixture began to bubbled

and turn to a dull grayish color. The reaction was followed by TLC and was determined

after 2 ½ hours that no change was observed. The reaction was then diluted with ether

and allowed to mix for 15 minutes. The mixture was then added to a flask containing

Page 48: Final Master Thesis revised 11.08.06

34

HPLC water to neutralize any remaining LiAlH4. While adding the mixture to water a

white precipitate (lithium hydroxide) was formed. The precipitate was filtered off and

the remaining solution was washed with 3 portions of methylene chloride. The

methylene chloride mixture was then washed with water and dried overnight by vacuum

to give an off-white crude product. Yield 0.22 g (40%) of (Z/E)-1-(3'-thiol-4'-methoxy)-

2-(3", 4", 5"-trimethoxyphenyl) ethane.

1H NMR E-isomer (300 MHz, CDCl3, δ of residual CHCl3 set to 7.26 ppm): 7.24 (d, H, J

= 2.4 Hz), 7.04 (dd, H, J = 6.4, 2.1 Hz), 6.72 (d, H, J = 8.5 Hz), 6.51 (s, 2H), 6.42 (s, 2H),

3.86 (s, 3H), 3.84 (s, 3H), 3.70 (s, 6H).

13C NMR Z-isomer (300 MHz, CDCl3, δ of residual CHCl3 set to 77.0 ppm): (ppm):

206.9, 154.0, 153.0, 129.8, 129.3, 128.7, 127.2, 110.3, 106.1, 61.0, 56.0, 56.0 and 31.0.

Acetal Protected 3-(N, N-dimethylcarbamoylthio)-4-methoxybenzaldehyde (19) To a well stirred solution of 17 (4.00 g, 16.7 mmol) and ethylene glycol (8.89 g, 8

mL, 143 mmol) in benzene, para- toluenesulphonic acid (0.066 g, 0.347 mmol) was

added. The temperature was then increased to 120 °C and allowed to react for 2hrs. The

reaction was monitored by thin-layer chromatography and determined that it was

complete. The benzene was removed in vacuo to give a brownish crude. The crude was

subjected to flash chromatography (silica gel, 50% EtOAc/50% hexanes, Rf = 0.23), in

order to obtain the acetal protected as a pale-yellow solid (yield 4.40 g).

1H NMR (300 MHz, CDCl3, δ of residual CHCl3 set to 7.26 ppm): (δ, ppm): 7.58 (d, H, J

= 2.2 Hz), 7.51 (dd, H, J = 2.2, 8.5 Hz), 6.96 (d, H, J = 8.5 Hz), 5.79 (s, H), 4.05 (m, 4H),

3.88 (s, 3H), 3.11 (b, 3H), 3.01 (b, 3H)

Page 49: Final Master Thesis revised 11.08.06

35

3-Thio-4-methoxybenzaldehyde (20)

To a well stirred solution of 19 (2.00 g, 7.06 mmol) in methanol, sodium

hydroxide (2.24 g, 3.50 mol) was added and allowed to reflux overnight. The reaction

was monitored by thin-layer chromatography (50% EtOAc/50% hexanes, Rf = 0.65).

The mixture was acidified to a pH 6.0 and allowed to mix an additional 30 minutes. The

methanol was removed by vacuum filtration and the product was dried overnight to give

a white solid (yield 1.45 g).

1H NMR (300 MHz, CDCl3, δ of residual CHCl3 set to 7.26 ppm): (δ, ppm): 9.84 (s,

COH), 7.81 (d, 1H, J = 2.02 Hz), 7.66 (dd, 1H, J = 2.05, 8.44 Hz), 6.97 (d, 1H, J = 12.05

Hz), 3.99 (s, 3H), 3.89 (s, SH)

3-(3’-carboxypropyl)-4-methoxybenzaldehyde disulfide (21) To a well stirred solution of 17 (2.00 g, 7.06 mmol) in methanol, sodium

hydroxide (2.24 g, 3.50 mol) was added and allowed to reflux overnight. The 3-

carboxypropyl disulfide was added in excess and allowed to mix for 1 hour. At this time

the reaction was followed by TLC until it was determined that the reaction had gone to

completion. The mixture was then acidified to a pH 6.0 and allowed to stir for 30

minutes. The methanol was removed by vacuum filtration to leave a crude white-

brownish solid. The crude product was subjected to flash chromatography (silica gel,

50% EtOAc/50% hexanes, Rf = 0.25) to give a white solid (yield 1.25 g).

1H NMR (300 MHz, CDCl3, δ of residual CHCl3 set to 7.26 ppm): (δ, ppm): 9.91 (s,

COH), 8.25 (d, 1H, J = 2.0 Hz), 7.75 (dd, 1H, J = 2.02, 8.38 Hz), 6.97 (d, 1H, J = 8.4 Hz),

3.99 (s, 3H), 2.79 (t, 2H, J = 7.10 Hz), 2.50 (t, 2H, J = 7.2 Hz), 2.04 (m, 2H, J = 7.0, 7.1,

7.3 Hz)

Page 50: Final Master Thesis revised 11.08.06

36

13C NMR (300 MHz, CDCl3, δ of residual CHCl3 set to 77.0 ppm): (ppm): 190.6, 177.9,

161.2, 130.8, 130.3, 128.5, 127.1, 110.4, 56.4, 37.3, 32.1, 29.7 and 23.7

Page 51: Final Master Thesis revised 11.08.06

37

CHAPTER SEVEN

Results and Discussion The goal of this research was to develop sulfur derivatives of Combretastatin A-4.

The thiol derivatives would then be converted to several novel disulfide prodrug

formulations, as well as reductively- and hydrolytically-activated compounds and the

antitumor activity of these new sulfur derivatives would be examined.

The initial problem was how we would put a thiol in the 3 position of the A-ring.

This problem was resolved by using a transformation based on the “Newman-Kwart

Rearrangement”38.

Figure 17: The Newman-Kwart Rearrangement38

This rearrangement was the key step in placing the thiol on C-3 of the A-ring.

There are two parts to this step that had a dramatic effect on the yield. If the temperature

OMe

O

HO

S

NMe

Me OMe

S

HO

O

NMe

Me

OMe

O

HO

S

NMe

Me OMe

O

HO

S

NMe

Me OMe

S

HO

O

NMe

Me

diphenyl ether245 C - 250 C

Mechanism:

Page 52: Final Master Thesis revised 11.08.06

38

was not kept within 245 °C – 250 °C the rearrangement would not be as effective which

would affect the yield and if the solution was added too rapidly to hexanes precipitation

would not occur efficiently. To allow as much of the product to precipitate out the

solution was added in portions over a 30 minute period. The next step that posed a

challenge was whether or not the Wittig reaction would give us our cis isomer in a high

enough yield. Two of the reactions can be seen below.

OCH3

OCH3H3CO

PPh3 Br

OCH3

S

HO

O

NCH3

CH3

H3CO

H3CO

OCH3

OCH3

S

O

NCH3

CH3

H3CO

H3CO

OCH3

OCH3

S

O N

CH3

CH3

NaH (6eq.) / CH2Cl2 25 C , ~ 7hrs

ratio: cis / trans 1 / 1

Cis - Isomer

Trans - Isomer

Figure 18: Wittig reaction using Sodium Hydride

n-BuLi , THF- 50 C

ratio: cis / trans 2 / 1

OCH3

OCH3H3CO

PPh3 Br

OCH3

S

HO

O

NCH3

CH3

H3CO

H3CO

OCH3

OCH3

S

O

NCH3

CH3

H3CO

H3CO

OCH3

OCH3

S

O N

CH3

CH3

Cis - Isomer

Trans - Isomer

Figure 19: Wittig reaction using Butyllithium

Page 53: Final Master Thesis revised 11.08.06

39

A comparison of the crude NMR’s of the reactions revealed that the cis to trans

ratio for the n-BuLi reaction was the most favorable (which is normally the best reagent

for this purpose) as it provided the products in a ratio of 2 to 1. The two isomers were

then carefully separated by column chromatography and each isomer was crystallized.

To verify that the NMR-based isomer assignments were correct, crystal structures of each

isomer were determined. It was found that crystals of these compounds could be grown

by dissolving the crude product in methylene chloride and initiating crystallization with

hexanes. These crystals had a yellow film around them which was washed off with cold

MeCl2, which left a clear crystalline solid. The yellow film was dried and crystallized

using the same procedure and produced a clear yellowish solid. Crystals of both isomers

were taken and crystal structures were determined by X-ray crystallography. The

structures are shown below (Figure 20 & 21) along with a summary of the

crystallographic data (Table 2) determined in our department by Dr. Kevin Klausmeyer.41

Figure 20: Thiol cis-isomer crystal structure41

Page 54: Final Master Thesis revised 11.08.06

40

Figure 21: Thiol trans-isomer crystal structure41

Table 2: Crystal Data for trans and cis- isomers41

Formula C21H25NO5S C21H25NO5S

Formula weight 403.48 403.48

Crystal System monoclinic monoclinic

Space Group (No.) C2/c P21/n

a (Å) = 21.468(5) 11.7825(7)

b (Å) = 1.932(1) 11.561(1)

c (Å) = 23.949(3) 14.911(1)

V (Å)3 = 4006(1) 2027.8(3)

Dcalc. (Mg m-3) = 1.338 1.322

Once the crystal structures of both the cis and trans isomers were determined

and the purity confirmed by HPLC we proceeded to the next step. After characterizing

the cis-isomer the next step was to convert the thiol-carbamate to a thiol. This step

became the most difficult to accomplish.

Page 55: Final Master Thesis revised 11.08.06

41

Figure 22: Purity of Cis-Isomer determined by HPLC

Figure 23: Thiocarbamate deprotection

Many variables that needed to be controlled during the deprotection step included

the dryness of the THF solution, temperature, isomerization and the deprotonation of the

thiol isomer during column chromatography. The one complication that we were not able

to avoid was the decomposition of the product and as a result the yield was very low. A

potential reason for this is that thiols can possibly react with the ethylene linker,

potentially forming polymeric products. After trying numerous different solvents for

column chromatography we determined that we would not be able to obtain the desired

H3CO

H3CO

OCH3

OCH3

SH

H3CO

H3CO

OCH3

OCH3

S

O

NMe

Me

H3CO

H3CO

OCH3

OCH3

SH

Cis - isomer

Trans - isomer

6eq LiAlH4, dry-THF, -50 C

Cis - isomer

+

Page 56: Final Master Thesis revised 11.08.06

42

product in a suitable enough yield. However, we were able to obtain enough of the pure

product in order to obtain an NMR spectrum and to test the blood flow shutdown.

Table 3: Blood flow in % of the control

CA4 (control) 100 ± 0

Oxi-com-224 100mg/kg 54.1 ± 11

Oxi-com-224 10mg/kg 83.3 ± 8.3

The above results showed us that the thiol analog of CA-4 was not as effective as CA4.

However, the limited activity could possibly be due to the decomposition of the thiol

analog.

At this point it was decided to try a different synthetic target. A variety of other

synthetic approaches were explored but the one compound that showed the most promise

was a disulfide combretastatin analog. The new synthetic scheme can be seen below,

OCH3

S

HO

O

NCH3

CH3

OH

OH

OCH3

S

O

NCH3

CH3

OO

OCH3

OO

S Na

p-TsOH+

1.) MeOH/NaOH/reflux

Benzene

Figure 24: Acetal Protection of Aldehyde

Once the aldehyde was protected the carbamate was removed using sodium

hydroxide in methanol under refluxing conditions, producing a protected thiol salt. The

thiol salt was then reacted with 3-carboxypropyl disulfide and acidified to a pH of 6.0.

Page 57: Final Master Thesis revised 11.08.06

43

OCH3

OO

S Na

HO

OCH3

S S

O

OH

1.) 3-Carboxypropyl disulf ide / pH 112.) Acidify to pH 6.0

Figure 25: Formation of Disulfide

The Wittig reaction was then performed on the deprotected aldehyde.

OCH3

OCH3H3CO

PPh3 Br HO

OCH3

S S

O

OH

n-BuLi , THF- 10 C / -75 C

H3CO

H3CO

H3COS S

H3CO

OH

O

Figure 26: Wittig Reaction for Disulfide

Despite many attempts we were unable to get complete separation of the cis and trans

products of this reaction.

Page 58: Final Master Thesis revised 11.08.06

44

CHAPTER EIGHT

Conclusion Various design and synthetic procedures were performed throughout both

research projects. During the oligonucleotide project much was learned through the

synthetic process. We were able to develop a clean reaction for 5’-

(Monomethoxytritylamino)-3’-(cyanoethyldiisopropyl) thymidine phosphoramidite using

an azeotrope method which produced a more pure product. We were also able to place a

MMT group on both the 5’- amine and 3’- amine ends of the thymidine compound.

Running the final product through the column at a faster rate allowed us to get enough of

the 5’-(Monomethoxytritylamino)-3’-(cyanoethyldiisopropyl) thymidine

phosphoramidite to characterize it by NMR and determine that we were able to obtain the

product but not in enough yield to proceed. In the end we learned a variety of separation

techniques such as reverse-, normal phase chromatography and preparatory TLC. As

well as synthetic techniques that could be applied to other synthesis such as an azetrope

method, using phosphoramidates.

Obtaining the thiol derivative of CA4 through a new route for a potential new

prodrug was developed. A variety of synthetic, separation and crystallization techniques

were learned while developing the CA4 thiol analog. The Newman-Kwart rearrangement

was successfully performed and was the key step in placing the thiol at the C-3 position.

Using X-ray crystallography it was determined that the CA4 thiol carbamate analog was

made. We were able to deprotect the thiol carbamate to give the desired thiol product

Page 59: Final Master Thesis revised 11.08.06

45

and obtain the blood flow shutdown data. Due to possible decomposition of the thiol

product a disulfide analog was explored. The disulfide Wittig reaction was attempted but

problems occurred that prevented us from obtaining the final disulfide product, despite

several approaches. Another approach that was not explored was to determine if n-BuLi

has more of an affinity to react with a disulfide or an aldehyde. If n-BuLi has a higher

affinity to the disulfide then a different reactant will be needed. Once this disulfide is

made this will allow a library of other CA4 disulfides to be developed. With this library

of disulfides there can be other compounds that will open the door to explore of research

areas.

Page 60: Final Master Thesis revised 11.08.06

46

APPENDICES

Page 61: Final Master Thesis revised 11.08.06

47

APPENDIX A

1H NMR for 3’-(Monomethoxytrityl) thymidine

ppm

(f1)

0.0

1.0

2.0

3.0

4.0

5.0

6.0

7.0

8.0

0 500

1000

1500

2000

7.525

7.498

7.433

7.4037.308

7.284

7.2647.258

7.222

7.198

7.173

6.833

6.8036.046

6.025

6.003

4.156

4.132

4.109

4.0853.882

3.835

3.777

3.749

3.681

3.640

3.381

3.357

3.326

3.311

3.287

3.264

2.079

2.046

1.964

1.830

1.283

1.259

1.235

3.851.963.922.87

1.88

1.00

0.77

0.934.480.96

2.42

0.75 1.20 0.514.40

2.06

3.80

Page 62: Final Master Thesis revised 11.08.06

48

APPENDIX B

13C NMR for 3’-(Monomethoxytrityl) thymidine

O

NH

N

O

OHO

HN

O

ppm (t1)50100150

0

10000

20000

30000

40000

50000

60000

163.

474

158.

202

150.

143

129.

752

128.

469

128.

094

126.

714

113.

387

110.

808

86.6

1284

.867

70.6

72

62.0

68

55.2

2053

.200

40.0

35

12.5

02

146.

331

138.

080

135.

973

Page 63: Final Master Thesis revised 11.08.06

49

APPENDIX C

1H NMR for 5’-(Monomethoxytrityl) thymidine

ONH

O

OH

N

NH(Z)

O

O

ppm (f1)0.01.02.03.04.05.06.07.08.0

0

50

100

150

200

250

300

3507.46

97.

445

7.37

67.

346

7.29

17.

268

7.24

27.

203

7.17

97.

095

6.82

16.

791

6.31

56.

293

6.27

1

5.45

7

4.31

14.

288

4.07

74.

054

3.76

93.

734

3.58

93.

564

3.54

03.

516

2.92

62.

902

2.87

82.

853

1.81

41.

451

1.42

71.

403

1.27

81.

254

1.23

0

0 0 0 0 0 0 0 0 0 0 0 0 0 0 3

ppm (f1)6.806.907.007.107.207.307.40

010203040506070

Page 64: Final Master Thesis revised 11.08.06

50

APPENDIX D

13C NMR for 5’-(Monomethoxytrityl) thymidine

ONH

O

OH

N

NH

O

O

ppm (f1)50100150

0

500

163.

632

157.

873

150.

303

145.

845

137.

672

135.

109

129.

689

128.

396

127.

857

126.

330

113.

159

111.

102

85.9

6284

.261

72.1

0470

.149

55.1

3952

.868

46.1

2245

.848

40.2

98

12.5

259.

571

7.92

1

Page 65: Final Master Thesis revised 11.08.06

51

APPENDIX E

1H NMR for 5’-(Monomethoxytrityl)-3’-(2-cyanoethyldiisopropylamino) thymidine

P

N

O

O

O

NHN

O

OHNO

N

H

ppm (f1)1.02.03.04.05.06.07.08.0

0

500

1000

1500

2000

7.5

7.4

7.4

7.3

7.3

7.3

7.3

7.2

7.2

7.2

7.2

6.9

6.8

6.2

6.1

6.1

4.5

4.5

4.0

4.0

4.0

4.0

3.9

3.9

3.7

3.7

3.7

3.6

3.6

3.6

2.9

2.8

2.8

2.3

1.7

1.0

0.9

0.9

0.894.106.412.06

2.31

0.84

0.71

0.930.284.333.19

0.57

3.04

2.88

8.80

Page 66: Final Master Thesis revised 11.08.06

52

APPENDIX F

13C NMR for 5’-(Monomethoxytrityl)-3’-(2-cyanoethyldiisopropylamino) thymidine

P

N

O

O

O

NHN

O

OHNO

N

H

ppm (t1)050100150

0

5000

10000

163.

499

157.

280

150.

186

146.

045

137.

542

136.

045

129.

484

128.

150

127.

552

125.

938

118.

684

115.

026

112.

891

109.

502

84.7

1883

.626

73.4

8469

.684

58.3

3355

.552

54.8

2445

.559

42.5

5142

.385

24.1

8224

.124

24.0

3124

.283

23.9

7120

.495

19.5

9019

.685

11.9

31

Page 67: Final Master Thesis revised 11.08.06

53

APPENDIX G

31P NMR for 5’-(Monomethoxytrityl)-3’-(2-cyanoethyldiisopropylamino) thymidine

P

N

O

O

O

NHN

O

OHNO

N

H

ppm (t1)-100-50050100150200

0

50

100

147.

216

Page 68: Final Master Thesis revised 11.08.06

54

APPENDIX H

1H NMR for CA4 Carbamate Cis-Isomer

H3CO

H3CO

OCH3

OCH3

S

O

NMe

Me

Page 69: Final Master Thesis revised 11.08.06

55

APPENDIX I

1H NMR (expanded) for CA4 Carbamate Cis-Isomer

H3CO

H3CO

OCH3

OCH3

S

O

NMe

Me

Page 70: Final Master Thesis revised 11.08.06

56

APPENDIX J

1H NMR for CA4 Carbamate Trans-Isomer

H3CO

H3CO

OCH3

OCH3

S

O N

CH3

CH3

Page 71: Final Master Thesis revised 11.08.06

57

APPENDIX K

1H NMR for CA4 thiol Cis-Isomer

H3CO OCH3

H3CO

OCH3

SH

Page 72: Final Master Thesis revised 11.08.06

58

APPENDIX L

1H NMR for CA4 thiol Cis-Isomer (expanded)

H3CO OCH3

H3CO

OCH3

SH

Page 73: Final Master Thesis revised 11.08.06

59

APPENDIX M

13C NMR for CA4 thiol Cis-Isomer

H3CO OCH3

H3CO

OCH3

SH

ppm (f1)050100150200

0

10

20

30

40

50

206.

9

154.

015

3.0

129.

812

9.3

128.

7

127.

2

110.

3

106.

1

61.0

56.0

56.0

31.0

TMS

silicone grease

Page 74: Final Master Thesis revised 11.08.06

60

APPENDIX N 1H NMR for Acetal Protected 3-(N, N-dimethylcarbamoylthio)-4-methoxybenzaldehyde

OCH3

S

OO

O

NCH3

CH3

ppm (f1)0.01.02.03.04.05.06.07.08.0

0

1000

2000

3000

4000

7.6

7.6

7.5

7.5

7.5

7.5

7.0

6.9

5.8

4.1

4.1

4.1

4.1

4.1

4.1

4.1

4.1

4.0

4.0

4.0

4.0

4.0

4.0

4.0

4.0

3.9

3.1

3.0

1.001.07

1.07

1.05

4.443.23

6.47

CHCl3

TMS

silicone grease

Impurity

Page 75: Final Master Thesis revised 11.08.06

61

APPENDIX O

3-Thiol-4-methoxybenzaldehyde

OCH3

SH

HO

ppm (t1)0.02.55.07.510.0

9.8

7.8

7.8

7.7

7.7

7.6

7.6

7.0

6.9

4.0

3.9

1.08

1.051.06

1.12

3.251.04

TMSCDCl3

Impurity from CDCl3

Page 76: Final Master Thesis revised 11.08.06

62

APPENDIX P

3-(3’-carboxypropyl)-4-methoxybenzaldehyde disulfide

HO

OCH3

S S

O

OH

ppm (t1)0.05.010.0

9.9

8.3

8.2

7.8

7.8

7.7

7.7

7.0

7.0

4.0

2.8

2.8

2.8

2.5

2.5

2.5

2.1

2.1

2.0

2.0

2.0

1.00

1.01

1.07

1.06

3.09

2.14

2.32

2.43

TMS

CDCl3

Page 77: Final Master Thesis revised 11.08.06

63

REFERENCES

1. Schleif, R.; “Genetics and Molecular Biology” 2nd edition, 1993, 268-272. 2. Dreyer, G.B.; Dervan, P.B. Proc. Natl. Acad. Sci. USA, 1985, 82, 968-972. 3. Jonklass, M. D.; The synthesis of novel phosphate diester prodrugs of

combretastatin A4: DNA cleavage with a metal ion-iminodiacetic acid linked deoxyoligonucleotide system or by photoreaction with 4-amino-1, 8-naphthalimides, Baylor University. 2003, 1-62.

4. Alberts, B.; Bray, D.; Lewis, J.; Raff, M.; Roberts, K.; Watson, D.J.; “Molecular

Biology of the Cell” 3rd edition, 1994, 803. 5. Burrows, F.J.; Thorpe, P.E. Pharmacol. Ther. 1994, 64(1), 155-174. 6. Siemann, D.W.; Chaplin, D.J.; Horsman, M.R. Cancer 2004, 100(12), 2491-

2499. 7 Pettit, G.R.; Cragg, G.M.; Herald, D.L.; Schmidt, J.M.; Lohavanijaya, P. Can. J.

Chem. 1982, 60, 1374-1376. 8 Pettit, G.R.; Cragg, G. M.; Singh, S. B. J. Nat. Prod. 1987, 50, 386-391. 9 Pettit, G.R.; Lippert, J.W. III; Herald, D.C. J. Org. Chem. 2000, 65, 7438-7444. 10 Hamel, E.; Lin, C.M. Biochem. Pharmacol. 1983, 32, 3864-3867. 11 Pettit, G.R.; Rhodes, M.R. Anticancer Drug Des. 1998, 13, 183-191. 12 Molema, G. Pharm. Res. 2002, 19(9), 1251-1258. 13. Ohsumi, K.; Nakagawa, R.; Fukuda, Y.; Hatanaka, T.; Morinaga, Y.; Nihei, Y.;

Ohishi, K.; Suga, Y.; Akiyama, Y.; Tsuji, T. J. Med. Chem. 1998, 41(16), 3022-3032.

14. Hori, K.; Saito, S.; Nihei, Y.; Suzuki, M.; Sato Y. Jpn J Cancer Res. 1999, 90,

1026-1038. 15 Sigman, D.S.; Mazumder, A.; Perrin, D.M. Chem. Rev., 93, 2295-2316. 16 Muiras, M. L.; Giacomoni, P. U.; Tachon, P. Mutat Res. 1993, 295(1), 47-54.

Page 78: Final Master Thesis revised 11.08.06

64

17 Strobel, S. A.; Dervan, P. B. Science, 1990, 249, 73-75. 18 Jordon, M. A.; Wilson, L. Nature Reviews. 2004, 4, 253-265. 19 Nogales, E.; Wolf, S. G.; Downing, K. H. Nature, 1998, 391, 199-203. 20 Furniss, B.S.; Hannaford, A.J.;Smith, P.W.G.; Tatchell, A.R., Vogel’s Textbook of

Practical Organic Chemistry, 5th Ed., 1989, Addison Wesley Longman Limited, Essex, England.

21 Pinney, KG.; Mejia, M.P.; Villalobos, V.M.; Rosenquist, B.E.; Pettit, G.R.;

Verdier-Pinard, P.; Hamel, E. Bioorg. Med. Chem. 2001, 8,2417. 22 Pettit, G.R.; Singh, S.B.; Boyd, M.R.; Hamel, E.; Pettit, R.K.; Schmidt, J.M.;

Hogan, F.J. J.Med.Chem. 1995, 38, 1666. 23. Pettit, G.R.; Toki, B.; Herald, D.L.; Veridier-Pinard, P.; Boyd, M.R.; Hamel, E.;

Pettit, R.K. J. Med. Chem. 1998, 41, 1688. 24. Hamel, E. Med. Res. Rev. 1996, 16 207. 25. Jordan, A.; Hadfield, J.A.; Lawrence, N.J.; McGown, A.T. Med. Res. Rev. 1998,

8, 259. 26. Gastpar, R.; Goldbrunner, M.; Marko, D.; von Angerer, E. J. Med. Chem. 1998,

41, 4965. 27. Wani, M.C.; Taylor, H.L.; Wall, M.E.; Coggon, P.; McPhail, A.T. J. Am. Chem.

Soc. 1971, 93, 2325. 28. Mulzer, J.; Mantoulidis, A.; Ohler, E. J. Org. Chem. 2000, 65, 7456. 29. Li, L.; Wang, H.K.; Kuo, S.C.; Wu, T.S.; Mauger, A.; Lin, C.M.; Hamel, E.; Lee,

K.H. J. Med. Chem. 1994, 37, 3400. 30. Chaudhuri, A.R.; Seetharamalu, P.; Schwarz, P.M.; Hausheer, F.H.; Luduena,

R.F. J. Mol. Biol. 2000, 303, 679. 31. Bai, R; Pei, X.; Boy, O.; Getahun, Z.; Grover, S.; Bekisz, J.; Nguyen, N.Y.;

Brossi, A.; Hamel, E. J. Biol. Chem. 1996, 271, 12639. 32. Pettit, G. R.; Cragg, G.M.; Herald, D.L.; Schmidt, J.M.; Lohavanijaya P. Can. J.

Chem. 1982, 60, 1375. 33 Pettit, G.R.; Lippert J.W. Herald, D.L. J. Org. Chem. 2000, 65, 7438.

Page 79: Final Master Thesis revised 11.08.06

65

34. Pettit, G.R.; Singh, S.B.; Niven, M.L.; Hamel, E.; Schmidt, J.M., J. Nat. Prod. 1987, 50, 119.

35. Pettit, G.R.; Singh, S.B. Can. J. Chem. 1987, 65, 2390. 36. Pettit, G. R.; Singh, S.B. J. Org. Chem. 1989, 54, 4105. 37. Maya, A.B.S.; Del Rey, B.; Clairac, R.; Caballero, E.; Barasoain, I.; Andreu,

J.M.; Medarde, M. Bioorg. Med. Chem. Lett. 2000, 10, 2549. 38 Newman, M. S.; Karnes, H. A.; J. Org. Chem. 1966, 3980. 39 Relles, H. M.; Pizzolato, G.; J. Org. Chem. 1968, 2249. 40. Pettit, G.R.; Singh, S. B.; Niven, M. L.; Hamel, E.; Schmidt, J. M.; J. Nat. Prod.

1987, 50, 119. 41 Ramirez, D. A.; Zhang, J.; Klausmeyer, K.; Kane, R. R. Journal of Chemical

Crystallography, 2005, 35, 167-170.