synthesis and characterization of novel platinum … · 2018. 1. 8. · platinum complexes –...

164
SYNTHESIS AND CHARACTERIZATION OF NOVEL PLATINUM COMPLEXES – THEIR ANTICANCER BEHAVIOUR By JOLANDA MYBURGH Submitted in fulfilment of the requirements for the degree of Magister Scientiae at the Nelson Mandela Metropolitan University January 2009 Supervisor: Prof JGH du Preez

Upload: others

Post on 30-Jan-2021

3 views

Category:

Documents


0 download

TRANSCRIPT

  • SYNTHESIS AND CHARACTERIZATION OF NOVEL PLATINUM COMPLEXES – THEIR ANTICANCER

    BEHAVIOUR

    By

    JOLANDA MYBURGH

    Submitted in fulfilment of the requirements for the degree of

    Magister Scientiae at the Nelson Mandela Metropolitan

    University

    January 2009

    Supervisor: Prof JGH du Preez

  • To my mother..

  • i

    ACKNOWLEDGEMENTS

    I wish to express my sincere thanks to:

    Prof. J.G.H. du Preez for his motivation, support and guidance.

    Dr. Mauritz Wentzel and Malcolm Taylor from Shimoda Biotech and Dr. L. Fourie

    from the University of Potchefstroom for the HPLC and mass spectroscopy work.

    Debbie du Plessis-Stoman and Anzel Bredenkamp under the supervision of Dr.

    M. van de Venter of the Biochemistry department of the NMMU, for the anticancer

    testing.

    Dr. Eric Hosten of the NMMU for the ICP analysis

    Henk Schalekamp for his assistance in the laboratory.

    Dr. M. Fernandes of the University of the Witwatersrand for the acquisition of the

    crystallographic data.

    Dr. B.J.A.M. van Brecht of the NMMU for the analysis of and the contributions

    towards the solution and refinement of the crystal structures.

    Prof. C.W. Mc Clelland of the NMMU for assisting in the molecular modelling.

    Mrs Erica Wagenaar for her expertise in the formatting of this dissertation.

    My colleagues, especially Yatish Jaganath, Lukas Oosthuizen, Marissa Louw and

    Yolanda Bouwer for their support, guidance, chemical advice and/or assistance

    with the experimental work.

    My family and friends for all the moral support

    God because by the Grace of God I Am What I Am

  • ii

    CONTENTS

    Page

    ACKNOWLEDGEMENTS ........................................................................................ i LIST OF FIGURES ................................................................................................... vii LIST OF TABLES ..................................................................................................... xiiiii ABBREVIATIONS .................................................................................................... xvi SUMMARY……………………………………………………………………………..xvi

    CHAPTER 1: INTRODUCTION

    1 HISTORICAL BACKGROUND ................................................................. 1 1.2 PLATINUM COMPOUNDS COMMONLY USED ...................................... 3 1.3 CHEMICAL CHARACTERISTIC OF PLATINUM(II) AND PLATINUM(IV) 5 1.4 IMPORTANT FACTORS REGARDING THE ANTICANCER ACTION OF

    PLATINUM COMPLEXES ........................................................................ 8 1.4.1 Kinetics ....................................................................................................... 8 1.4.2 Stereochemistry ......................................................................................... 10 1.4.3 S-N interaction (N7) .................................................................................... 10 1.4.4 Biocompatibility ......................................................................................... 13 1.5 RECENT DEVELOPMENTS IN PLATINUM ANTICANCER RESEARCH .... 14 1.6 OBJECTIVES OF THIS DISSERTATION ................................................. 15

    CHAPTER 2: EXPERIMENTAL PROCEDURES

    2 INTRODUCTION ....................................................................................... 17 2.1 ANALYSIS TECHNIQUES ........................................................................ 17 2.1.1 Elemental Analysis ..................................................................................... 17 2.1.2 Spectrophotometric Analysis ................................................................... 17 2.1.3 Solubility Studies ....................................................................................... 20 2.1.4 Crystallographic Analysis ......................................................................... 20 2.1.5 Cytotoxicity Studies ................................................................................... 21 2.1.6 Methods applied for the Ionization Studies ............................................. 22 2.2 PREPARATION OF STARTING MATERIALS ......................................... 24

  • iii

    2.2.1 K2PtCl6 ......................................................................................................... 24 2.2.2 K2PtCl4 ......................................................................................................... 24 2.2.3 K2Pt(C2O4)2.2H2O ........................................................................................ 25 2.3 SYNTHESIS OF THE (NS)-DONOR LIGANDS ........................................ 25 2.3.1 1-Methyl-2-methylthioalkylimidazole ........................................................ 25 2.3.1.1 1-Methyl-2-methylthiomethylimidazole(mmtmi) ...................................... 25 2.3.1.2 1-Methyl-2-methylthioethylimidazole (mmtei) .......................................... 27 2.3.1.3 1-Methyl-2-methylthiopropylimidazole (mmtpi) ....................................... 27 2.3.2 1-Butyl-2-methylthiomethylimidazole(bmtmi): ........................................ 28 2.4 SYNTHESIS OF PLATINUM(II) COMPLEXES WITH BIDENTATE (NS)-

    LIGANDS .................................................................................................. 28 2.4.1 Synthesis of the (NS)- haloplatinum(II) complexes ................................. 29 2.4.1.1 Preparation of the (NS)-iodoplatinum(II) complexes ............................... 29 2.4.1.2 Preparation of the (NS)-chloroplatinum(II) complexes ........................... 29 2.4.1.3 Preparation of the (NS)-bromoplatinum(II) complexes ........................... 30 2.4.2 Synthesis of the (NS)-oxalatoplatinum(II) complexes ............................. 31 2.5 SYNTHESIS OF PLATINUM(II) COMPLEXES WITH N-DONOR

    LIGANDS – MONODENTATE AND BIDENTATE .................................... 32 2.5.1 Synthesis of the aminehalo- and oxalatoplatinum(II) complexes .......... 32 2.5.2 Synthesis of platinum(II) complexes of the type PtL1L2X2 (L1 and L2

    are two different monodentate amines) ................................................... 33 2.6 ELEMENTAL ANALYSIS OF PLATINUM(II) COMPLEXES .................... 34 2.7 ANTICANCER ACTION OF THE (NS)-HALOPLATINUM(II) COMPLEXES . 36 2.8 SYNTHESIS OF PLATINUM(IV) COMPLEXES ....................................... 38 2.8.1 Synthesis of Platinum(IV) complexes: …………………………………38

    CHAPTER 3: THE (NS)-HALOPLATINUM(II) COMPLEXES

    3 INTRODUCTION ....................................................................................... 40 3.1 SYNTHETIC METHODS AND THE IMPLICATIONS THEREOF ............. 42 3.1.1 Introduction ................................................................................................ 42 3.1.2 Method A - Direct ligand exchange from K2PtCl4 .................................... 42 3.1.3 Method B - Silver Method .......................................................................... 44 3.2 (NS)-PLATINUM(II) COMPLEXES - THEIR PHYSICAL PROPERTIES .. 46

  • iv

    3.2.1 Infrared Spectroscopy ............................................................................... 46 3.3 MASS SPECTROMETRY .......................................................................... 50 3.3.1 (NS)-iodoplatinum(II) complexes .............................................................. 50 3.3.2 (NS)-bromo- and chloroplatinum(II) complexes ...................................... 56 3.4 IONIZATION OF THE (NS)-HALOPLATINUM(II) COMPLEXES .............. 61 3.5 ANTICANCER BEHAVIOUR .................................................................... 66 3.6 SOLUBILITY STUDIES ............................................................................ 67 3.7 CONCLUSION .......................................................................................... 67 3.8 APPENDIX ................................................................................................. 70

    CHAPTER 4: THE (NS)-DICARBOXYLATOPLATINUM(II) COMPLEXES

    4 INTRODUCTION ....................................................................................... 70 4.1 SYNTHETIC METHODS AND THE IMPLICATIONS THEREOF ............. 71 4.2 (NS)-OXALATOPLATINUM(II) COMPLEXES THEIR PHYSICAL

    PROPERTIES ........................................................................................... 74 4.2.1 Infrared Spectroscopy ............................................................................... 74 4.2.2 Mass Spectrometry .................................................................................... 75 4.3 IONIZATION STUDIES .............................................................................. 75 4.4 ANTICANCER ACTION ............................................................................ 77 4.4.1 Final comments .......................................................................................... 80 4.5 CONCLUSION ........................................................................................... 82 4.6 APPENDIX ................................................................................................ 82

    CHAPTER 5: PLATINUM(II) COMPLEXES WITH N-LIGAND

    5 INTRODUCTION ....................................................................................... 86 5.1 NON-LEAVING GROUPS ......................................................................... 88 5.2 AMINEIODOPLATINUM(II) COMPLEXES ............................................... 89 5.3 AMINEBROMOPLATINUM(II) COMPLEXES ........................................... 94 5.4 AMINECHLOROPLATINUM(II) COMPLEXES ......................................... 97 5.5 AMINEOXALATOPLATINUM(II) COMPLEXES ........................................ 102 5.6 INFRARED SPECTROSCOPY ................................................................. 109

  • v

    5.7 COMPARISON OF IONIZATION BEHAVIOUR ....................................... 114 5.8 ANTICANCER BEHAVIOUR .................................................................... 118 5.9 SOLUBILITY STUDIES ............................................................................ 121 5.10 CONCLUSION .......................................................................................... 121

  • vi

    CHAPTER 6: MONONITRO PLATINUM(IV) COMPLEXES

    6 INTRODUCTION ....................................................................................... 123 6.1 THE REACTION OF THE (NS)-PLATINUM(II) COMPLEXES WITH NO2 123 6.2 EFFORTS TO SYNTHESIZE PLATINUM(IV) ANALOGUES OF THE

    (NS)-OXALATOPLATINUM(II) COMPLEXES .......................................... 124 6.3 EFFORTS TO SYNTHESIZE PLATINUM(IV) ANALOGUES OF THE

    AMINEPLATINUM(II) COMPLEXES ........................................................ 126 6.4 MECHANISM OF OXIDATION ................................ ……………………130 6.5 CONCLUSION ..................................................................................... 133

    REFERENCES

  • vii

    LIST OF FIGURES

    Page

    Figure 1:1 Structure of cisplatin ............................................................................ 1

    Figure 1:2 Platinum complex of the type PtA2X2Y2 ............................................... 2

    Figure 1:3 Structure of carboplatin ........................................................................ 3

    Figure 1:4 Structure of oxaliplatin ......................................................................... 4

    Figure 1:5 Structures of Lobaplatin (a) and Nedaplatin (b) ................................... 5

    Figure 1:6 The dissociative (SN1) and associative (SN2) mechanisms ................. 6

    Figure 1:7 Structure of the amino acids methionine (a) and cysteine (b) ............. 8

    Figure 1:8 Space filling model (a) and charge distribution values (b) of

    adenine. ............................................................................................... 11

    Figure 1:9 Space filling model (a) and charge distribution values (b) of guanine .. 11

    Figure 1:10 Space filling model(a) and charge distribution values (b) of cysteine .. 12

    Figure 2.1 Experimental set up for the synthesis of the platinum(IV) complexes

    Figure 3:1 (NS)-Donor Ligands ............................................................................. 41

    Figure 3:2 Full Scan Mass Spectra of J13.2 (FAB method) .................................. 43

    Figure 3:3a Full Scan Mass Spectra (electronspray method) of J13.2D in

    positive ion mode (top) and negative ion mode (bottom) –

    (M=monomolecular species and A=auto-ionized species). .................. 43

    Figure 3:3b Possible structures of the detected ions - auto-ionized and

    monomolecular species ....................................................................... 42

    Figure 3:4 Full scan mass spectrum of J13.2 (FAB method) ................................ 45

    Figure 3:5 Full scan mass spectrum of J13.2 (electron spray method) in

    positive (top) and negative (bottom) ion modes ................................... 46

    Figure 3:6 Infrared spectrum of ligand J7 showing the characteristic infrared

    stretches .............................................................................................. 47

    Figure 3:7 Infrared spectrum of J7.2 showing characteristic stretches of the

    (NS)-haloplatinum(II) complexes ......................................................... 47

    Figure 3:8 Full Scan mass spectrum of J13.5 (FAB method) ................................ 51

    Figure 3:9 Full Scan mass spectra of J13.5 in positive (top) and negative

    (bottom) ion modes. Possible structures of detected ions are given

    below spectra ....................................................................................... 52

  • viii

    Figure 3:10 Full Scan mass spectra of J5.5 in positive (top) and negative

    (bottom) ion modes. (M=monomolecular species and A=auto-

    ionized) ................................................................................................ 53

    Figure 3:11 Full Scan mass spectra of J7.5 in positive (top) and negative

    (bottom) ion modes. (M=monomolecular species and A=auto-

    ionized species.) The possible structures of some of the detected

    ions are given below the spectra. ........................................................ 54

    Figure 3:12 Full Scan mass spectra of J12.5 in positive (top) and negative

    (bottom) ion mode. (M=monomolecular species and A=auto-

    ionized). Possible structures of some of the detected ions are given

    below spectra ....................................................................................... 55

    Figure 3:13a Full Scan mass spectra of J7.2 in positive (top) and

    negative(bottom) ion modes ............................................................... 56

    Figure 3:13b Possible structures of some of the detected ions ................................. 53

    Figure 3:14 Full Scan mass spectra of J7.3 in positive (top) and negative

    (bottom) ion modes. Possible structures of some of the detected

    ions are given below spectra ............................................................... 57

    Figure 3:15 Full Scan mass spectra of J13.3 in positive (top) and

    negative(bottom) ion modes .............................................................. 58

    Figure 3:16 Full Scan mass spectra of J5.2 in positive (top) and

    negative(bottom) ion modes. Possible structures of detected ions

    are given below spectra ....................................................................... 59

    Figure 3:17 Full Scan mass spectra of J5.3 in positive (top) and negative

    (bottom) ion modes. Possible structures of detected ions are given

    below spectra ....................................................................................... 60

    Figure 3:18 Full Scan mass spectra of J7.2 showing ionization behaviour in

    CH3CN and H2O at room temperature at T= 0 and 80ºC at T = 18 ...... 61

    Figure 3:19 Full Scan mass spectra of J5.2 showing ionization behaviour in

    CH3CN and H2O at room temperature at T= O and 80ºC at T = 18 ..... 62

    Figure 3:20 Effect of the ligand on the ionization rate for the molecular species

    [M+NH4] + for (a) the chloro, (b) bromo and (c) iodo species ............... 63

    Figure 3:21 Effect of the leaving groups on the ionization rate for the molecular

    species [M+NH4] + for (a) J7, (b) J5 and (c) J12. ................................ 64

    Figure 3:22: Effect of the leaving groups on the hydrolysis rate for the

  • ix

    monocationic species [ M-X+S]+ for (a) J7, (b) J5 and (c) J12. ........... 65

    Figure 4:1 Graph detailing the drop in cisplatin concentration in water (37°C)

    over time .............................................................................................. 70

    Figure 4:2 Graph indicating the stability in carboplatin concentration in water

    (37°C) over time ................................................................................. 70

    Figure 4:3 Graph demonstrating the stability in oxaliplatin concentration in

    water (37°C) over time ......................................................................... 71

    Figure 4:4 Full Scan Mass Spectrum of J7.1 synthesized by the direct method

    (method A) in positive and negative ion modes ................................... 72

    Figure 4:5 Full Scan Mass Spectrum of J7.1 synthesized by the silver method

    (method B) in positive and negative ion modes. Possible structures

    of detected ions are given below spectra ......................................... 73

    Figure 4:6 LC-UV separation in H2O of J7.1 ......................................................... 74

    Figure 4:7 Full Scan Mass Spectrum of J7.1 synthesized by the silver method

    (method B) in positive mode in CH3CN and H2O at room

    temperature at T=O and 80ºC at T= 60Hr. .......................................... 76

    Figure 4:8 Full Scan Mass Spectrum of J5.1 synthesized by the silver method

    (method B) in positive mode in CH3CN and H2O at room

    temperature at T=0 and 80ºC at T=60Hr ............................................ 76

    Figure 4:9 Anticancer action of J7.1; percentage inhibition as a function of

    concentration of J7.1 in μM for HT29 ................................................... 77

    Figure 4:10 Anticancer action of J7.1; percentage inhibition as a function of

    concentration of J7.1 in μM for HeLa ................................................... 78

    Figure 4:11 Anticancer action of J7.1; percentage inhibition as a function of

    concentration of J7.1 in μM for MCF7 .................................................. 78

    Figure 4:12 2-methylthiomethylpyridine .................................................................. 80

    Figure 4:13 Full scan mass spectra of J5.1 in positive ion mode (top) and

    negative ion modes (bottom) ............................................................... 82

    Figure 4:14 Full scan mass spectra of J12.1 synthesised via Method A in

    positive (top) and negative (bottom) ion modes ................................... 82

    Figure 4:15 Full Scan mass spectra of J12.1 synthesised via Method B in

    positive (top) and negative(bottom) ion modes .................................... 83

    Figure 4:16 Full scan mass spectra of J13.1 synthesised via Method A in

    positive (top) and negative (bottom) ion modes ................................... 83

  • x

    Figure 4:17 Full scan mass spectra of J13.1 synthesised via Method B in

    positive (top) and negative (bottom) ion modes ................................... 84

    Figure 4:18 Full scan mass spectra of J12.1 synthesized by method b in

    positive ion mode in CH3CN and H2O at T=0 and at 80°c at T= 60Hr .. 84

    Figure 4:19 Full scan mass spectra of J13.1 synthesized by method B in positive

    ion mode in CH3CN and H2O at T=0 and at 80°C at T= 60Hr .............. 85

    Figure 5:1 Structure of cisplatin and its hydrolysis products ................................. 86

    Figure 5:2 Structure of picoplatin ......................................................................... 87

    Figure 5:3 Full scan mass spectrum of JMY13.5 in positive (top spectrum) and

    negative (bottom spectrum) ion modes. Possible structures of some

    of the detected ions are given below spectra ....................................... 90

    Figure 5:4 Full scan mass spectrum of JMY14.5 in positive (top spectrum) and

    negative (bottom spectrum) ion modes. Possible structures of some

    of the ions are given below the spectra. ............................................... 91

    Figure 5:5 Full scan mass spectra of JMY15.5 in positive (top spectrum) and

    negative (bottom spectrum) ion mode. Possible structures of some

    of the detected ions are given below spectra. ...................................... 92

    Figure 5:6 Full scan mass spectrum of JMY16.5 in positive ion mode. Possible

    structures of some of the detected ions are given below spectra ........ 93

    Figure 5:7 Full scan mass spectrum of JMY17.5 in positive ion mode and

    negative ion mode (M = molecular species and S = symmetrical

    species). Possible structures of some of the detected ions are given

    below spectra ....................................................................................... 94

    Figure 5:8 Full scan mass spectrum of JMY13.3 in positive (top spectrum) and

    negative (bottom spectrum) ion modes. Possible structures of some

    of the detected ions are given below spectra ....................................... 95

    Figure 5:9 Full scan mass spectra of JMY14.3 in positive and negative ion

    mode. Possible structures of detected ions are given below spectra ... 96

    Figure 5:10 Full scan mass spectra of JMY15.3 in CH3CN/H2O solution, positive

    (top spectrum) and negative (bottom spectrum) ion mode ................... 97

    Figure 5:11 Full scan mass spectra of JMY13.2 in positive (top spectrum) and

    negative (bottom spectrum) ion modes. Possible structures of some

    of the detected ions are given below spectra ....................................... 98

    Figure 5:12 Full scan mass spectra of JMY14.2 in positive (top spectrum) and

  • xi

    negative (bottom spectrum) ion modes. Possible structures of some

    of the ions are given below spectra ..................................................... 99

    Figure 5:13 Full scan mass spectra of JMY15.2 in positive (top spectrum) and

    negative (bottom spectrum) ion modes. Possible structures of some

    of the detected ions are given below spectra ....................................... 100

    Figure 5:14 Full scan mass spectrum of JMY16.2 in positive ion mode. Possible

    structures of detected ions are given below spectrum ......................... 101

    Figure 5:15 Full scan mass spectra of JMY13.1 in positive (top spectrum) and

    negative (bottom spectrum) ion modes. Possible structures of some

    of the detected ions are given below spectra ....................................... 102

    Figure 5:16 Full scan mass spectra of JMY14.1 in H2O, positive (top spectrum)

    and negative (bottom spectrum) ion modes. Possible structures of

    detected ions are given below spectra ................................................. 103

    Figure 5:17 Full scan mass spectra of JMY15.1 in positive (top spectrum) and

    negative (bottom spectrum) ion modes. Possible structures of some

    of the detected ions are given below spectra ....................................... 104

    Figure 5:18 Full scan mass spectrum of JMY16.1 in positive ion mode. Possible

    structures of some of the detected ions are given below spectra ........ 105

    Figure 5:19 Full scan mass spectra of JMY17.1 in positive (top spectrum) and

    negative (bottom spectrum) ion modes. Possible structures of some

    of the detected ions are given below spectra ................................. 106

    Figure 5:20 LC-UV separation of JMY17.1 in 25% CH3CN (JMY17.1 at peak

    4.30) ..................................................................................................... 107

    Figure 5:21 LC-MS separation of JMY17.1 in 25% CH3CN solution(positive ion

    mode) ................................................................................................... 107

    Figure 5:22 LC-MS separation of JMY17.1 in 25% CH3CN solution (negative ion

    mode) ................................................................................................... 108

    Figure 5:23 (a) ball and stick model of JMY15.2, lowest energy conformer. (b) an

    alternative ball and stick model for JMY15.2 ........................................ 109

    Figure 5:24 Infrared spectrum of JMY13.2 showing the characteristic stretches

    of amineplatinum(II) complexes ........................................................... 110

    Figure 5:25 Full scan mass spectra of JMY13.5 in H2O/CH3CN solution, showing

    ionization behaviour at different time intervals at 37°C ........................ 114

    Figure 5:26 Full scan mass spectra of JMY13.3 in H2O/CH3CN solution, showing

  • xii

    ionization behaviour at different time intervals at 37°C ........................ 115

    Figure 5:27 Full scan mass spectra of JMY13.2 in H2O/CH3CN solution, showing

    ionization behaviour at different time intervals at 37°C ........................ 115

    Figure 5:28 (a) Effect of the leaving group on the ionization rate for the

    molecular species [M+NH4] + and (b) the monocationic species [M-

    X+S] for the chloro, bromo and iodo species; X = Cl, Br or I and S =

    CH3CN. ................................................................................................ 116

    Figure 5:29 A comparison between the extent of ionization of the (NN)- and

    (NS)-complexes for the monocationic species [M-X+S]+ ( X=Cl, Br or

    I and S=CH3CN). (a) Comparison between the dichlorocomplexes,

    (b) the dibromo complexes. and (c) the diodo complexes .................... 117

    Figure 6:1 Structure of CPA-7 ............................................................................... 123

    Figure 6:2 Full Scan mass spectra of J12.1.4 in positive (top) and negative

    (bottom) ion modes. Possible structures of detected ions are given

    below spectra ....................................................................................... 125

    Figure 6:3 Mass spectra of JMY15.1.4 in positive ion mode (top) and negative

    ion mode (bottom). Possible structures of some of the detected ions

    are given below spectra ....................................................................... 127

    Figure 6:4 LC-UV separation of sample JMY15.1.4 in 25% CH3CN ..................... 127

    Figure 6:5 Full scan mass spectra of JMY16.1.4 in positive ion mode (top) and

    negative ion mode (bottom). Possible structures of some of the

    detected ions are given below spectra ................................................. 128

    Figure 6:6 Full scan mass spectra of JMY13.1.4 in positive ion mode (top) and

    negative ion mode (bottom). Possible structures of some of the

    detected ions are given below spectra ................................................. 129

    Figure 6:7 Trans-aqua(nitrosyl)tetranitroplatinate(IV) anion .................................. 130

    Figure 6:8 Trans-dinitro-trans-dichloro-trans-nitrosylchloroplatinum(IV) ion ......... 131

    Figure 6:9

    X-ray crystallographic structure of platinum(IV) analogue of

    oxaliplatin ............................................................................................. 132

    Figure 6:10 Possible stucture of JMY15.1.4 ........................................................... 132

  • xiii

    LIST OF TABLES

    Page

    Table 2:1 General ICP settings used ................................................................... 18

    Table 2:2 Colour and yields of the (NS)- iodoplatinum(II) complexes . ................ 29

    Table 2:3 Colour and yields of the (NS)-chloroplatinum(II) complexes prepared

    via method A ........................................................................................ 30

    Table 2:4 Colour and yields of the (NS)-chloroplatinum(II) complexes prepared

    via method B ........................................................................................ 30

    Table 2:5 Colour and yields of the (NS)-bromoplatinum(II) complexes

    prepared via method B ......................................................................... 31

    Table 2:6 Colour and yields of the (NS)-oxalatoplatinum(II) complexes

    prepared via method A ......................................................................... 31

    Table 2:7 Colour and yields of the (NS)-oxalatoplatinum(II) complexes

    prepared via method B ......................................................................... 32

    Table 2:8 Colour and yields of the aminehaloplatinum(II) complexes prepared

    via method B. ....................................................................................... 32

    Table 2:9 Colour and yields of the amine-oxalatoplatinum(II) complexes

    prepared via method B. ........................................................................ 33

    Table 2:10 Elemental analysis of the (NS)-chloroplatinum(II) complexes via

    method A ............................................................................................. 34

    Table 2:11 Elemental analysis of the iodoplatinum(II) complexes ............................ 34

    Table 2:12 Elemental analysis of the chloroplatinum(II) complexes via method B 35

    Table 2:13 Elemental analysis of the bromoplatinum(II) complexes via method

    B .......................................................................................................... 35

    Table 2:14 Elemental analysis of the oxalatoplatinum(II) complexes via method A ..... 36

    Table 2:15 Elemental analysis of the oxalatoplatinum(II) complexes via method B ..... 36

    Table 2:16 Cell growth inhibition assay results for (NS)-haloplatinum(II)

    complexes on HT29 (colon cancer) cell lines at two different

    concentrations in μM. ........................................................................... 36

    Table 2:17 Cell growth inhibition assay results for (NS)-haloplatinum(II)

    complexes on Hela (cervical cancer) cell lines at two different

    concentrations in μM ............................................................................ 37

  • xiv

    Table 2:18 Cell growth inhibition assay results for (NS)-haloplatinum(II)

    complexes on MCF7 (breast cancer) cell lines at two different

    concentrations in μM ............................................................................ 37

    Table 2:19 Chemicals used ................................................................................... 39

    Table 3:1 Infrared data for the (NS)-donor ligands .............................................. 48

    Table 3:2 Infrared data for complexes of J5 ........................................................ 48

    Table 3:3 Infrared data for complexes of J7 ....................................................... 48

    Table 3:4 Infrared data for complexes of J12 ..................................................... 49

    Table 3:5 Infrared data for complexes of J13 ..................................................... 49

    Table 3:6 The ν(C-N), ν(CH2S) def and ν(CH2S) wag shifts of the (NS)-

    chloroplatinum(II) complexes ............................................................... 50

    Table 3:7 Summary of the major ion clusters (pos ion mode) observed in the

    mass spectra of the samples in CH3CN and water and in the

    aqueous samples after ≥ 18Hr at 80ºC ................................................ 66

    Table 3:8 Solubility of the (NS)-haloplatinum(II) complexes ................................ 67

    Table 4:1 Infrared data for the (NS)-oxalatoplatinum(II) complexes .................... 74

    Table 4:2 The ν(CH2S) def and ν(CH2S) wag shifts of the (NS)-

    oxalatoplatinum(II) complexes ............................................................. 75

    Table 4:3 IC50 values obtained for J7.1 and cisplatin ......................................... 79

    Table 4:4 Cell growth inhibition assay results for (NS)-oxalatoplatinum(II)

    complexes on HT29 (colon cancer) cell lines using two different

    complex concentrations. ...................................................................... 79

    Table 4:5 Cell growth inhibition assay results for (NS)-oxalatoplatinum(II)

    complexes on HeLa (cervical cancer) cell lines using two different

    complex concentrations ....................................................................... 79

    Table 4:6 Cell growth inhibition assay results for (NS)-oxalatoplatinum(II)

    complexes on MCF7 (breast cancer) cell lines using two different

    complex concentrations ....................................................................... 79

    Table 5:1 Infrared data for amine ligands ............................................................ 110

    Table 5:2 Infrared data for aminehaloplatinum(II) complexes ............................. 111

    Table 5:3 Infrared data of the amine-oxalatoplatinum(II) complexes ................... 112

    Table 5:4 The ν(NH2-stretch) ν(NH2-def) and ν(C-N) shifts of the

    amineplatinum(II) complexes ............................................................... 113

  • xv

    Table 5:5 Cell growth inhibition assay results for aminehaloplatinum(II)

    complexes on the HT29 (colon cancer) cell line at two different

    concentrations in μM. ........................................................................... 118

    Table 5:6 Cell growth inhibition assay results for aminehaloplatinum(II)

    complexes on the HeLa (cervical cancer) cell line at two different

    concentrations in μM ............................................................................ 119

    Table 5:7 Cell growth inhibition assay results for aminehaloplatinum(II)

    complexes on the MCF7 (breast cancer) cell line at two different

    concentrations in µM ............................................................................ 119

    Table 5:8 Cell growth inhibition assay results for amine-oxalatoplatinum(II) -

    complexes on the MCF7 (breast cancer) cell line at two different

    concentrations in μM ............................................................................. 120

    Table 5:9 Cell growth inhibition assay results for amine-oxalatoplatinum(II)

    complexes on the HT29 (colon cancer) cell line at two different

    concentrations in μM ............................................................................. 120

    Table 5:10 Cell growth inhibition assay results for amine-oxalatoplatinum(II)

    complexes on the HeLa (cervical cancer) cell line at two different

    concentrations in μM ............................................................................. 120

    Table 5:11 Solubility of the amineplatinum(II) complexes ...................................... 121

  • xvi

    ABBREVIATIONS

    mmtmi: 1-methyl-2-methylthiomethylimidazole

    mmtei: 1-methyl-2-methylthioethylimidazole

    mmtpi: 1-methyl-2-methylthiopropylimidazole

    bmtmi: 1-butyl-2-methylthiomethylimidazole

  • xvii

    SUMMARY

    In this dissertation novel non-leaving groups were employed to synthesize

    platinum complexes which can contribute to the understanding or improvement of

    anticancer action. These complexes basically consist of (NS)-chelate and

    amineplatinum complexes.

    Bidentate (NS)-donor ligands were used as non-leaving ligands in the syntheses

    of platinum(II) complexes with iodide, chloride, bromide and oxalate anions as

    leaving groups. These complexes were synthesized and studied since many

    questions regarding the interaction of sulfur donors and platinum still exists.

    These relate to thermodynamic and kinetic factors and their influence on

    anticancer action. In this dissertation the properties of novel platinum(II)

    complexes of a bidentate ligand having an aromatic nitrogen-donor atom in

    combination with a thioethereal sulfur atom capable of forming a five membered

    ring with platinum(II) were studied. The general structure of the (NS) -ligands

    used were N-alkyl-2-methylthioalkyl imidazole. Alkyl groups used were methyl,

    ethyl and propyl.

    Although amine complexes of platinum have been extensively studied there are

    some new aspects of these that are worthwhile investigating. In this dissertation

    amines having planar attachments which will be at an angle with the coordination

    plane viz. benzylamine and amines having cyclic aliphatic groups namely

    cyclopropyl and cyclohexyl were investigated.

    Some of the (NS) - and amineplatinum(II) complexes were oxidised to their

    mononitroplatinum(IV) analogues . The motivation for the synthesis of these

    complexes was the greater kinetic stability of platinum(IV) and recent research

    has shown that a specific type of platinum(IV) compound shows suitable

    properties as an anticancer agent.

    These complexes were characterised by a variety of spectral means (IR, NMR,

    mass spectroscopy) as well as elemental analysis, solubility determinations,

    thermal analysis (TGA), ionization studies and finally their anticancer behaviour

    towards three different cell lines(Hela, MCF 7, Ht29) and in this process they

  • xviii

    were compared to the behaviour of cisplatin as a reference. A few have shown

    promising anticancer behaviour.

  • 1

    | 1

    CHAPTER 1: INTRODUCTION

    1 HISTORICAL BACKGROUND

    The interest in the pharmacological properties of platinum compounds was

    initiated by Rosenberg’s accidental discovery of the inhibition of cell-division by

    platinum complexes in 1965.1 It was found that the complexes responsible for the

    effects were cis-diamminedichloroplatinum(II) (cis-PtCl2(NH3)2, cisplatin) and cis-

    diamminetetrachloroplatinum(IV) (cis-PtCl4(NH3)2). 2,3

    H3N

    H3N Cl

    Cl

    Pt

    Figure 1:1 Structure of cisplatin

    Cisplatin is effective in the treatment of a broad range of solid tumours and has

    become one of the most widely used anticancer drugs since 1971 when it entered

    clinical trials for the first time.4,5

    Although it is commonly used cisplatin has several side effects. Side effects of

    cisplatin include cumulative peripheral neuropathy, ototoxicity, hair loss, nausea

    and vomiting.

    The presence of the chloride anions as leaving

    ligands makes cisplatin biologically compatible with the body. In blood and other

    body fluids chloride is found in different concentrations.

    6 Cisplatin has become the prototype for research and development

    of platinum-based antitumour drugs with improved pharmacological properties

    and a reduction of negative effects commonly found with cisplatin. The bulk of

    these compounds is uncharged cis-configured square planar platinum(II)

    complexes with the general formula cis-PtA2X2 where A2 is either two amines or a

    diamine as neutral ligands and X2 either a monodentate or bidentate anionic

    leaving group such as a halo or a dicarboxylato group. Platinum(IV) complexes of

    the form PtA2X2Y2 are also undergoing clinical trials,.7 Y2 being two other

  • 2

    | 2

    monodentate anionic leaving groups.

    A

    A X

    X

    Pt

    Y

    Y

    Figure 1:2 Platinum complex of the type PtA2X2Y2

    The only differences between the two types of anticancer platinum complexes are

    the higher oxidation state and the two axial leaving groups. Platinum(IV) is less

    reactive than platinum(II) and platinum(IV) drugs can possibly be administrated

    orally. However these compounds are reduced to platinum(II) in the body

    accompanied by the loss of the two axial ligands and thus rather than being seen

    as a new type of drug it can be seen as prodrugs.6 Research has shown that

    trans-diamminedichloroplatinum(II) (transplatin) is less effective than cisplatin.

    Transplatin is kinetically more reactive than cisplatin and more susceptible to

    deactivation. 8

    A major limitation in the therapeutic success of cisplatin and its derivatives is the

    fact that some cancer cells become resistant to the drugs.9,10 Efforts are being

    made to overcome resistance. One of the main reasons cells become resistant to

    cisplatin is that insufficient amounts of cisplatin reach and bind to the DNA and

    this leads to failure of cell death which takes place after binding of platinum to

    DNA.6 Reduced platinum accumulation and cytoplasmic detoxification by

    glutathione and / or metallothioneins are the most important reasons why

    inadequate amounts of platinum reach DNA. Mechanisms within the cell can also

    lead to platinum resistance after platinum binding to the DNA has occurred.

    These mechanisms include increased DNA repairs of adducts and the ability to

    withstand greater levels of DNA damage failure to engage programmed cell death

    pathways.8

  • 3

    | 3

    1.2 PLATINUM COMPOUNDS COMMONLY USED

    Since the introduction of cisplatin numerous platinum complexes have been

    synthesized and evaluated but only a limited number is used worldwide.9 Most of these

    complexes are cisplatin analogues. The most important of these drugs is diammine-

    1,1-cyclobutanedicarboxylato-(2)-O,O’-platinum(II) (carboplatin, ([(NH3)2Pt(C4H6O4)]).5

    O

    OPt

    H3N

    H3N

    O

    O

    Figure 1:3 Structure of carboplatin

    On inspection it can be seen that carboplatin differs from cisplatin by the

    replacement of the chloride anions with a dicarboxylic chelate ligand with a

    hydrophobic substituent. This derivative of malonic acid is a relatively weak acid.

    The weak acidity and the resulting six-membered chelate ring enhance its stability

    and thus limit the rate of detoxification and the number of negative side effects.5

    Due to the enhanced stability of carboplatin, its ligand exchange reactions occur

    at a slower rate than those of cispaltin. Carboplatin is also more readily

    transferred across the cell wall due to the presence of the hydrophobic

    component.11 The half life of carboplatin is approximately 30h in body fluid. It

    resists attack by thiols more readily than cisplatin and the four oxygen atoms

    increase its aqueous solubility. Carboplatin is less toxic but also less reactive than

    cisplatin and it must be administered in a higher dose.12 A further advantage of

    carboplatin is its ability to circumvent the dose limiting nephrotoxicity of

    cisplatin.13 It is clinically proven that carboplatin is active against the same range

    of tumours than cisplatin but because of its lower toxicity patients tolerate it better

    and thus patients who could not continue with cisplatin treatment can continue

    with carboplatin treatment.14 The main toxicity of carboplatin is myelosuppression.

    A disadvantage of carboplatin is that it also reacts with glutathione to form

    inactive platinum-thiol complexes and thus is not active against cisplatin-resistant

    tumours.15

  • 4

    | 4

    More recently trans-R,R-1,2-diaminocyclohexaneoxalatoplatinum(II) (oxaliplatin)

    has gained interest. This can be regarded as the first non-cisplatin analogue.

    Oxaliplatin belongs to a generation of drugs that contain different types of chiral

    amine molecules6,16

    Figure 1:4 Structure of oxaliplatin

    Oxaliplatin has shown antitumour activity in colorectal cancer and is used for the

    treatment of metastatic colorectal cancer. Oxaliplatin has shown to be effective in

    the treatment of cisplatin resistant tumours.6 Oxaliplatin is able to avoid mismatch

    repair and replicate bypass. It has reasoned that the 1,2-

    diaminocyclohexane(DACH) ligand projects into the major grooves of DNA, which

    play an important role in these mechanisms.

    17

    In addition to oxaliplatin being effective against cisplatin resistant cell lines a

    further advantage is that it is less toxic. The decreased toxicity is due to the

    oxalato leaving group being ionised more slowly and to a smaller extent than the

    chloro leaving groups of cisplatin. Oxaliplatin does not show nephrotoxicity,

    myelosuppression and ototoxicity.17,18. However, side effects are present.

    Neuropathy and emesis are the toxicities related to oxaliplatin treatment.19 This

    chelated diamine ligand has a stereochemical effect. Due to crowding, thiol

    interference is less and a larger amount of platinum reaches the DNA thus

    circumventing platinum resistance further.20

    A few other platinum compounds have achieved regional approval. Lobaplatin

    (lactatodiaminomethylcyclobutaneplatinum(II)) has been approved in China for the

    treatment of breast cancer21 and Nedaplatin (cis-glycolatodiamineplatinum(II)) in

    Japan for the treatment of head and neck, testicular, bladder and ovarian cancer.22

    H2NPt

    H2N

    O

    O

    O

    O

    ,5

  • 5

    | 5

    a)

    H2N

    NH2

    Pt

    O

    O

    b)

    Pt

    H3N

    H3N

    O

    O

    O

    Figure 1:5 Structures of Lobaplatin(a) and Nedaplatin(b)

    1.3 CHEMICAL CHARACTERISTICS OF PLATINUM(II) AND PLATINUM(IV)

    Why is platinum effective in the treatment of cancer? Platinum, being a later 5d

    transition, metal has the required properties which renders it suitable for

    anticancer action. It is highly covalent in its bonding (Platinum(II) is a soft metal

    ion indicated by its Marcus softness σ value of +0.33 as compared to K+ with a σ

    value of -0.5),23 It forms kinetically stable complexes (ligand exchange rate in

    solution is slow) and it prefers bonding to the so-called soft ligands like aliphatic

    and aromatic nitrogen-donor ligands and sulfur-donor ligands (e.g. SH- has a σ

    value of +0.65; Cl- = -0.09, Br-=+0.17 and I-=+0.50 on a scale where OH- =0)23

    Platinum(II) is diamagnetic and its complexes have a square planar symmetry.

    Ligand exchange of its complexes can occur via an associative (SN2) or a

    dissociative (SN1) mechanism. Ligand exchange of the square planar platinum(II)

    complexes occurs via a distorted pentacoordinated species either a trigonal

    bipyramidal or square pyramidal intermediate complexes thus by an associative

    process. (In very recent literature it was indicated that even the

    tetradiaquaplatinum(II) consists of a distorted species with four equal H2O

    molecules in the plane and one extended molecule on the apex of a distorted

    square pyramid)24

  • 6

    | 6

    M

    R

    R

    XR

    M

    R

    R

    R

    M

    R

    R

    R

    Y

    X

    Y

    M

    R

    R

    YR

    X

    Yfast

    SN2 Path

    SN1 Path

    X

    5-coordinate intermediate

    3-coordinate intermediate

    Y = nucleophileX = leaving group

    Figure 1:6 The dissociative (SN1) and associative (SN2) mechanisms

    Platinum(II) and platinum(IV) mainly differ by the higher covalency and greater

    kinetic stability of the latter. Square planar platinum(II) is a 16 electron system

    with a vacant low lying orbital which allows for coordination expansion as

    indicated by the SN2 mechanism pathway which is normally lower in activation

    energy than a SN1 mechanism thus more reactive. In contrast octahedral

    platinum(IV) is an 18 electron system in which case only a SN1 mechanism

    occurs thus kinetically more stable.

    The above behaviour can be used to explain the anticancer action of

    dichloroplatinum(II) complexes such as cisplatin. In aqueous solution and

    biological fluids (blood and cell fluid) the chloro ligands of platinum(II) complexes

    including cisplatin are replaced by aqua ligands e.g. ionization of cisplatin in

    blood. The chloride concentration in extracellular fluids is high (100µM in blood)

    and lowers the ionization rate whereas in the cell the chloride concentration is low

    (4µM) promoting ionization. However chemical equilibrium is not obtained within

    the body. 6, 24 See equation 1 and 2 for the stepwise ionization of cisplatin.

  • 7

    | 7

    Equation one and two:

    Pt

    Cl

    Cl

    H3N

    H3NPt

    Cl

    OH2

    H3N

    H3N

    Pt

    OH2

    OH2

    H3N

    H3N

    2

    -Cl +H2O -Cl +H2O

    equation 1 equation 2

    It is believed that the small amount of aqua species is the actual anticancer agent since it bonds to the aromatic nitrogen of guanine in the DNA strand with two of such atoms thus forming an intrastrand crosslink. This ultimately leads to apoptosis. Deprotonation can occur lowering the reactivity since the hydrolysed species is less reactive.7 See equation 3 -5 for possible deprotonation reactions.

    Equation three:

    -H

    equation 3

    Pt

    Cl

    OH2

    H3N

    H3N

    Pt

    Cl

    OH

    H3N

    H3N

    Equation four and five:

    Pt

    OH

    OH2

    H3N

    H3N

    H-Pt

    OH

    OH

    H3N

    H3N

    -H

    equation 4 equation 5

    Pt

    OH2

    OH2

    H3N

    H3N

    2

    A number of nitrogen (amines) and sulfur-donor ligands are present in the blood

    serum and organs. A too high rate of ionization leads to the interaction of such

    species with the platinum compound before the platinum reaches the cell. It has

    been found that chloride can be directly exchanged for certain sulfur donors such

    as the amino acids methionine and cysteine without the need of prior aquation.7

  • 8

    | 8

    a) NH2

    S

    O

    HO

    b)

    H2N

    SH

    O

    OH

    Figure 1:7 Structure of the amino acids methionine (a) and cysteine (b)

    These effects lead to relatively low efficiency of haloplatinum(II) complexes

    including cisplatin. Too fast rates of ionization lead to detoxification before these

    complexes reach the cell and this results in great toxicity to the rest of the body.

    Too slow rates cause inefficiency.24,25

    1.4 IMPORTANT FACTORS REGARDING THE ANTICANCER ACTION OF PLATINUM COMPLEXES

    In the previous section the relevant basic chemistry of platinum which has a

    bearing on anticancer activity was briefly discussed. It is now necessary to

    consider all the parameters of the platinum species i.e. combining the platinum

    metal properties with the various ligands and their properties thus the total

    platinum anticancer agent. These factors are taken into consideration with

    specific reference to the design of new anticancer agents having superior action.

    The following factors will now be considered namely kinetics, stereochemistry, S-

    N interaction (N7) and biocompatibility.

    1.4.1 Kinetics

    In the previous section the effect of the kinetics of diamminedichloroplatinum(II)

    complexes like cisplatin was considered from a basic viewpoint. The importance

    of ligand exchange rate was emphasised. Too fast kinetics means high toxicity,

    too slow kinetics means inefficient anticancer action. There are a number of

    secondary factors of which the trans effect and its variations are very significant.

    The trans effect i.e. the capability of a coordinated ligand to labilise a ligand trans to it

    in a square-planar platinum complex can in general be applied to ligand exchange in

    platinum complexes. The normal order of trans directing is:

  • 9

    | 9

    CN- ~ CO~ olefins > thioethers ~ H+ ~ arsines ~phosphines > SC(NH2)2 ~ CH3 >

    C6H5- ~ NO2- ~ I- ~SCN- > Br- > Cl- > amines ~NH3 ~ OH- ~ H2O.26

    i. Coligands can have a great effect on the coordinated ligand in covalent

    transition metal chemistry like that of platinum(II) and platinum(IV). E.g.

    coligands with high Eneg like N and O cause a high effective nuclear charge

    on the platinum leading to a sulfur-donor ligand with a smaller trans effect

    than predicted. Such properties are referred to as biphilic. 27

    From the above

    sequence it appears that sulfur-donor ligands will act differently from Cl- , water and

    amines which are the ligands commonly found in the body. Sulfur-donor ligands are

    used to demetallate heavy metals and it has been found that cisplatin is removed via

    urine bonded to sulfur-donor ligands.27 Considering the above factors it is clear that a

    study of the all the factors influencing ligand exchange is necessary when designing

    new anticancer agents in an effort to optimise their anticancer properties.

    Although the trans effect is important as a general rule there are a number of

    other factors also influencing ligand exchange rate.

    ii. Sterically hindered systems also influence the nucleophilicity sequence. In

    these systems the size and shape of the neucleophile becomes more

    important than the order of the sequence. In some cases reversal of

    nucleophilicity order occurs. The position of the steric effect is also important,

    e.g. a 2-methyl substituted pyridine cis to leaving group has a much bigger

    effect than when trans to the leaving group. Stereochemical bulk in a ligand in

    the close vicinity of platinum provides an energy barrier for the formation of

    the activated intermediate thus negatively influencing the SN2 mechanism and

    therefore lowering the ligand exchange rate.7

    iii. Lability is also influenced by bond lengths, force constants and stretching

    frequencies e.g. the Pt-Cl bond length is 2.21Å trans to NH3 and 2.37Å

    trans to a P in Pme3.27

    iv. The effect of the cis ligand (cis effect) is very important in Pt(NH3)2Cl2.

    When adjacent ligands have weak trans effects the cis effect has a greater

    influence in the behaviour of the coordinated ligands. Cl- > NH3 is normal,

    but in reality the opposite occurs.27

    v. The chelate effect in the case of anions slows down ligand exchange due

  • 10

    | 10

    to its stabilising effect on the platinum anion bonding.

    It should be borne in mind that more than one of the above factors could be

    acting simultaneously.

    1.4.2 Stereochemistry

    i. Crowding nitrogen-donor ligands with alkyl groups does not only slow

    down the ligand exchange rate, but it also prevents donation of the large

    sulfur atoms present in biological systems which can detoxify the agent by

    creating a barrier.16 Crowding also limits organ toxicity.

    ii. Stereochemistry also plays a role in the level of replicative bypass of Pt-DNA

    adducts and in the repair mechanism. In the case of DACH non-leaving group

    it is anticipated that the DACH co-ligand interacts with the major groove of the

    DNA assisting in the platination of the aromatic nitrogen of DNA.27

    iii. Increased stereochemistry in a non-leaving group results in increased

    hydrophobicity which facilitates passive diffusion through the cell wall and

    promotes the uptake of anticancer agents.

    28

    1.4.3 S-N interaction (N7)

    The large nucleophilicity of the sulfur-donor ligands give them a trans labilizing

    effect as previously discussed. However, this varies from sulfur donor to sulfur

    donor. Two types of sulfur-donor ligands exist namely thioethers and sulfoxides

    which are known for their σ donor π acceptor action, and secondly thiols and

    thiourea and similar compounds that interact via σ donor π donor bonding. It is

    known that the stronger σ donor ligands like thiols form more stable complexes

    with platinum than the weaker ones depending on π back donation as seen in the

    case of thioethers. The σ donor π donor group interacts irreversibiliy with

    platinum(II) e.g. the aromatic nitrogen (N7) of guanine cannot replace these. The

    former group is thermodynamically less stable and can be replaced by the N7 of

    guanine.29

    Platinum drugs exert their antitumour effects via damage to DNA. Platinum reacts

    with DNA preferentially to the N7 atoms of purine nucleobases. The rate of the

  • 11

    | 11

    initial binding step and the closure to form the bifunctional adduct are dependent

    on the kinetics of the hydrolysis rate. The DNA adducts consist of intrastrand

    cross-links involving adjacent bases, most frequently involving adjacent guanine

    bases and less frequently adenine adjacent to guanine.

    It is of interest that previous ligand exchange studies performed on platinum(II)

    anticancer compounds under in vivo conditions i.e. pH7 in the presence of Cl- ions, it

    was found that the order of replacement is thiols (glutathione) > GN7 > thioether

    (methionine) > AN7..31

    A comparative study was done on the two relevant N7 atoms namely adenine and

    guanine. Space filling models and charge distribution values of these ligands

    were obtained to investigate the possible reasons for the above sequence. (See

    figure 1.8 and 1.9).

    a) b)

    Figure 1:8 Space filling model (a) and charge distribution values (b) of adenine.

    a) b)

    Figure 1:9 Space filling model (a) and charge distribution values (b) of guanine

    From studying the above, the following can be concluded:

    i. The N7 atom of guanine is stereochemically less crowded than the N7 atom

    of adenine

  • 12

    | 12

    ii. The negative charges on the two atoms adjacent to adenine N7 atoms are

    lower than those of the guanine, thus guanine has a greater potential of

    induction by the platinum(II).30

    iii. Due to the greater σ basicity of the adenine N7 atom it will get protonated

    to a greater extent at pH7 than the guanine N7 atom. The adenine N7 atom

    is thus less available for platinum(II).

    31

    iv. Stronger H-bonding capability of guanine also plays a role in stability of its

    platinum-DNA adduct.

    There is also a distinct difference between the two types of sulfur atoms. The

    thioethers can be regarded as virtually neutral sulfur atoms, but the thiol sulfur

    can readily be deprotonated by the very soft 5d transition metal platinum in effect

    changing it from a neutral to a monoanionic ligand with enough covalent

    character to satisfy the nephelauxetic character of platinum(II).

    A space filling model and the charge distribution values of the biothio compound

    cysteine was obtained to study the effect of sulfur donation. The large size of the

    sulfur atoms immediately becomes clear as well as the slight negative charge

    already present before deprotonation

    a) . b)

    Figure 1:10 Space filling model (a) and charge distribution values (b) of cysteine

    If the size of the sulfur atom is compared to that of the aromatic nitrogen-donor

    atom then the difference is obvious. Ligand exchange of platinum(II) complexes

    occurs via an intermediate distorted pentacoordinated complex and therefore

    stereochemical crowding of the platinum(II) square planar complex will have

    bigger negative effects on ligand exchange.32 Due to the size difference between

  • 13

    | 13

    the bulky sulfur atoms and the aromatic nitrogen-donor atom it appears that there

    ligand exchange will be influenced more negatively for the sulfur donor than the

    nitrogen donor.

    Chelated nitrogen-donor ligands are used to control the effect of the sulfur donors

    such as thioether (methionine). The usage of chelated nitrogen donors increases

    the thermodynamic stability and provides a bigger barrier towards the

    replacement by the thioether sulfur atoms.34

    However, it should be borne in mind that chemical potential is proportional to

    concentration and there is more aromatic nitrogen donors in body fluid and cells

    than sulfur-donor atoms and this can influence the situation described above.

    1.4.4 Biocompatibility

    There are several factors that enhance the biocompatibility of a platinum

    compound.

    Solubility – In the design of anticancer agents a compromise needs to be reached

    between hydrophobicity (which normally lowers the aqueous solubility) and the

    aqueous solubility. Too low solubility is to be avoided since enough platinum

    compound must be introduced into the body to be effective.5

    Membrane transport – Studies have shown that these drugs initially enter the

    body through passive diffusion and facilitated diffusion through a gated channel.33

    Since the interior of the cell membrane consists of hydrophobic hydrocarbon tails,

    increasing the hydrophobicity or lipophilicity of an anticancer agent thus plays a

    role in the uptake of these compounds.34, 35

    It has been shown that the greater the lipophilic nature of complexes, the more

    readily it enters the cells and thus resistance can be circumvented due to

    decreased platinum accumulation. Carboxylate, carbonate or carbamate ligands

    used in such complexes are suitable ligands since they contain polar and

    lipophilic regions. Examples of such complexes are aminedibutylratodichloro

    An example of this is JM216

    (bis(acetato)ammine-dichloro(cyclohexylamine)platinum(IV), its increased

    lipophilicity enhanced its uptake via passive diffusion.35

  • 14

    | 14

    (cyclohexylamine)platinum(IV) (JM221). NNDP is a lipophilic platinum compound

    containing the DACH carrier ligand and lipophilic neodecanoic acid leaving

    groups encapsulated in a liposome. The cell uptake for L-NNDP is 8-10 fold

    higher compared to cisplatin because of its increased lipophilicity.

    I,2-intrastrand crosslinks formed by platinum binding to DNA can create a

    hydrophobic notch at the damage site, which can be bound by specific damage

    recognition proteins, thus shielding it from NER activity. Thus by using more

    hydrophobic platinum complexes, resistance can be prevented and this can lead

    to higher antitumour activity.

    Biologically active carrier groups – In some cases third generation drugs have a

    carrier molecule attached to the normal chloride binding areas, which are

    molecules the tumour cells may desire in particular and are more readily taken

    into the tumour cells. An example includes the platinum moiety to deoxorubicin

    and oestrogen analogues that bind to estrogen receptors. Carrier groups with the

    ability to direct platinum to specific organs as well as DNA intercalators that will

    localize the platinum in the vicinity of the DNA have been investigated. However,

    there have been no clinically significant advances that have been developed from

    attaching biologically active groups to platinum compounds.

    1.5 RECENT DEVELOPMENTS IN PLATINUM ANTICANCER RESEARCH

    Initial studies on the improvement of the anticancer action of platinum complexes

    centred around cisplatin and its structure e.g. the synthesis of various

    diamineplatinum(II) complexes with different amine non-leaving groups. Extensive

    anticancer studies showed that these ‘cisplatin analogues’ have very similar

    anticancer behaviour to cisplatin with reference to cell resistance and toxicity.

    Some of these properties were modified by using leaving groups that slows down

    ligand exchange and thus also slow down detoxification e.g. carboplatin. This was

    followed by the so-called second generation anticancer drugs e.g. oxaliplatin. In

    the case of oxaliplatin the non-leaving group had a positive effect on the

    platination of the N7 of guanine.

  • 15

    | 15

    The next development in the design of new platinum anticancer drugs was to

    produce platinum(IV) compounds which are inert and can thus be taken orally.

    The latter two modifications are still being investigated. The effect of variation of

    structure and nature of platinum complexes on the anticancer action is still under

    investigation. Platinum(IV) proved to be effective against cancer and thus studies

    on platinum(IV) compounds are still continued. More recently specific platinum(IV)

    compounds appeared in the literature which proved to have promising anticancer

    action and Stat-3 behaviour. 36,37

    1.6 OBJECTIVES OF THIS DISSERTATION

    STAT (signal transduction and translation) proteins migrate during the translation

    phase to the cell nucleus where it binds to promoter elements of DNA molecules,

    leading to expression of target genes. STAT proteins are normal constituents of

    cells, but in STAT-3 activity is frequently up in regulated in many human tumours.

    It has been shown that some platinum anticancer agents interact with STAT-3

    inhibiting binding to its DNA target. 38

    There are still unanswered questions on specific aspects of sulfur-donor ligands.

    Targeting of the anticancer cells also became more important in view of the

    damage done to the normal cells (thus minimizing the negative side effects).

    Some properties of sulfur-donor ligands have been briefly discussed under

    kinetics. The dominant role of σ donor π donor atoms which irreversibly bond to

    platinum was mentioned as a negative factor. However σ donor π acceptor sulfur

    atoms like thioethers have some positive characteristics in so far that the bulky

    sulfur atom can protect the platinum through stereochemical crowding although it

    can be replaced by the N7 of guanine. The dominant trans directing power of

    sulfur can be limited. An example of this is a chelated compound where electron

    withdrawal from the sulfur atom can be achieved. In this research this specific

    property was studied by using a bidentate ligand having an aromatic nitrogen

    linked to a thioethereal sulfur via a methylene group in the form of a chelate

    ligand capable of forming a five membered ring. In this case it could be expected

    that some sulfur electron density will be drawn through the CH2 connection into

  • 16

    | 16

    the aromatic system thus weakening the effect.

    Platinum(II) complexes of such non-leaving groups having different N1 substituted

    imidazole ligands and varying leaving groups (Cl- , Br-, I- and oxalate) were

    synthesized, characterized and tested for anticancer action.

    Although amine complexes of platinum(II) have been extensively studied some

    amine non-leaving groups were selected for specific reasons e.g. having cyclic

    substituted groups with the potential to assist in platination and OH substituted

    groups for the improvement of aqueous solubility.

    The mononitrochloroplatinum(IV) complexes of some of the platinum(II)

    analogues especially those that showed significant anticancer action, were

    studied.

  • 17

    | 17

    -CHAPTER 2: EXPERIMENTAL PROCEDURES

    2 INTRODUCTION

    This chapter will be devoted to discussion of the experimental techniques applied

    for both analysis and characterization with special reference to the

    instrumentation involved. The instrumentation employed in this research project

    will be described here mentioning some of the analytical work done at other

    laboratories where instruments were not available in this department.

    This information will be followed by the general preparative methods and where

    required, alterations of it. This will be sequential to correspond to the later

    chapters.

    2.1 ANALYSIS TECHNIQUES

    2.1.1 Elemental Analysis

    Microanalysis for C, H, N, and S was performed at the University of Cape Town.

    The samples were analysed by a Thermo Flash 1112 Elemental Analyzer

    2.1.2 Spectrophotometric Analysis

    Infrared spectroscopy

    Infrared spectra were recorded using an Excalibur HE FTS3100

    spectrophotometer in the range of 4000-200 cm-1 using the single beam mode.

    Solid samples were compressed with dry potassium bromide into discs and

    scanned with nitrogen gas as reference. Liquid samples were spread as a thin

    film between two sodium chloride plates and scanned using the empty plates as

    reference.

  • 18

    | 18

    NMR spectrometry

    The 300.13 MHz 1H-NMR spectra were recorded on a Bruker 300 MHz

    spectrometer using SiMe4 as the internal standard. The samples were prepared

    using CDCl3 as solvent.

    Inductively-coupled plasma emission mass spectroscopy

    The platinum content of the samples prepared for the solubility studies were

    analysed using a Perkin Elmer SCIEX Elan-6100 ICP-MS Spectrometer. The ICP

    settings used are given below in Table 2.1

    Table 2:1 General ICP settings used

    The isotopes 195Pt and 196Pt were used for optimal analysis. The standards were

    prepared in an aqueous medium containing 0.1 M HCl in the range of 5-1000 µg/l.

    Samples for analysis were diluted appropriately and prepared in a 0.1 M HCl

    matrix.

    FAB Mass spectrometry

    FAB mass spectra were obtained from the University of Potchefstroom. The

    spectra were recorded on a VG70-70E mass spectrometer using a VG2035 data

    system. All samples were analysed under positive-ion operating conditions. An

    Ion Tech saddle-field using xenon as bombardment gas (8 kV, 1 mA) was

    employed to record the spectra. The mass spectrometer was operated at 6 kV

    and the magnetic analyser was scanned at 5 s per decade between 500-100 Da.

    The matrix used was glycerol or 3-nitrobenzylalcohol.

    Setting RF Power 1.4 kW

    Coolant gas 15 L/min

    Nebulizer gas flow 0.7L/min

    Dwell time per AMU 50ms

    Sweeps/reading 10

    Readings/Replicate 2

    Replicate 10

    Integration 5.0 s

  • 19

    | 19

    Electronspray Mass Spectrometry

    Electron Spray mass spectra were obtained from Platco laboratories at Shimoda-

    Biotech. Measurements were carried out on an LCQ Deca instrument (Finnigan,

    San Jose,CA, USA) in either positive or negative ionisation mode. For infusion

    studies the samples, in appropriate solvent, were directly infused at a flow rate of

    10 μM/min using a 500 μL unimetrics HPLC syringe and the instrument’s own

    syringe pump. A 50% CH3CN/10 mM ammonium formate/0.06% formic acid

    solution was used as sheath liquid at a flow rate of 150 μL/min. Direct contact

    with the analysis solution was allowed by positioning the tip of the fused silica

    capillary (i.d.100 μM, o.d. 190 μM) 0.5 to 1.0mm inside the tip of the stainless

    steel sprayer capillary. The parameters of the instrument were tuned utilising an

    infusion solution of Pt(dach)Cl2 in CH3CN (10 μM at 10 μL/min).

    Positive ion mode instrument settings - The instrument parameters were tuned

    using the ammonium adduct [M+NH4]+ with the following values: nitrogen sheath

    gas flow rate 80 arbitrary units, nitrogen auxiliary gas flow rate 20 arbitrary units,

    sprayer voltage 3.5 kV, capillary voltage 46.0 V, tube lens offset 55.0 V, multipole

    1 offset -5.75 V, lens voltage -28.0 V, multipole 2 offset -10.00 V, multipole RF

    amplitude (V p-p) 400 V, entrance lens -62.0 V and 100 ms maximum injection

    time using automatic gain control.

    Negative ion mode instrument settings - The parameters of the instrument were

    tuned using the formate adduct [M+HCOO]- to the following values: nitrogen

    sheath gas flow rate 80 arbitrary units, nitrogen auxiliary gas flow rate 20 arbitrary

    units, sprayer voltage 5.0 kV, capillary voltage -12.0 V, tube lens offset 5.0 V,

    multipole 1 offset 5.25 V, lens 49 voltage 46.0 V, multipole 2 offset 10.00 V,

    multipole RF amplitude (V p-p) 400 V, entrance lens 38.0 V and 100 ms

    maximum injection time using automatic gain control.

    LC-MS

    The LCQ Deca mass spectrometer was connected to a Thermo Surveyor HPLC

    system consisting of a quaternary gradient pump and autosampler. For LC-MS

    analyses, varying solvents were utilised but in each case, the solvents and

  • 20

    | 20

    modifiers complied with the overriding requirement of ESI-MS, in that all

    components had to be volatile. Thus, no separations could be effected with the

    help of phosphates or sulphonic acids and no sodium, potassium or chloride salts

    could be used. Instead, volatile buffer systems such as ammonium formate,

    ammonium acetate or diethylamine were used.

    HPLC-UV

    LC separations were performed on a Waters HPLC system consisting of a

    WISP717 autosampler, 600E quaternary gradient pump and 484 variable

    wavelength UV-Vis detector.

    HPLC separation columns used during this study:

    • YMC Hydrosphere C18 120 Å 5 μM 250 x 4.6 mm

    • Phenomenex Curosil PFP 5 μM 250 x 4.6 mm

    2.1.3 Solubility Studies

    The solubilities of the complexes were measured by suspending the compound in

    2 ml H2O and stirring at 25°C for 30 minutes after which the mixture was

    centrifuged for 5 minutes at 5000 rpm's. The supernatant liquor was diluted so

    that the platinum concentrations were suitable for ICP-MS analysis. 500 and 1000

    times dilutions were sufficient.

    2.1.4 Crystallographic Analysis

    Full X-ray structure determinations were attempted on some platinum complexes.

    The diffraction data was collected at the University of the Witwatersrand using a

    Bruker SMART 1K CCD area detector diffractometer with graphite

    monochromated MoKα radiation (50 kV, 30 mA). The collection method involved

    ω-scans of width 0.3°. Data reduction was carried out using SAINT+38 and

    absorption corrections were made using the program SADABS39

    The WinGX 1.64.02

    .

    40 suite of programs was used to solve the crystal structures

    using either direct methods or the Pattersen approach. Refinement was done

  • 21

    | 21

    using SHELXS-9741. Non-hydrogen atoms were first refined isotropically followed

    by anisotropic refinement by full matrix least-squares calculation based on F2.

    Hydrogen atoms were positioned geometrically and allowed to ride on their

    respective parent atoms with one exception. Torsional angles and least squares

    planes of selected atoms were calculated using PARST42. Diagrams were

    generated using ORTEP43

    2.1.5 Cytotoxicity Studies

    .

    All the complexes prepared in this dissertation were tested for anticancer action

    according to known literature methods44,45

    Cytotoxicity in cell lines was determined by a modified MTT assay (Sigma). Cells

    in exponential growth were trypsinized, counted using a haemocytometer and

    diluted to a density of 30 000 cells per ml. Aliquots (200µL) of cell suspensions

    were added to each well of a 96-well microtiterplate and incubated for 24 hours.

    The novel platinum containing compounds were screened for cytotoxicity using

    10 and 100µM concentrations. Cisplatin (10 and 100µM concentrations) served

    as a positive control. The spent medium was removed from the 96-well plate and

    aliquots (200 µl) of the treatments were added to the wells in quadruplicate. The

    plate was incubated in a 37°C-5% CO2 humidified incubator for 48 hours. A

    by the Biochemistry department of the

    Nelson Mandela Metropolitan University under the supervision of Dr. M. van de

    Venter. The compounds were tested on colon (HT29), breast (MCF 7) and

    cervical cancer (HeLa) cell lines. Cisplatin was used as reference. A summary of

    the general method is given below.

    The reduction of tetrazolium salts is now widely accepted as a reliable way to

    examine cell proliferation. The yellow tetrazolium salt MTT (3-(4,5-

    dimethylthiazolyl-2)-2, 5-diphenyltetrazolium bromide) is reduced by metabolically

    active cells, in part by the action of dehydrogenase enzymes. The resulting

    intracellular purple formazan can be solubilized and quantified by

    spectrophotometric means. The MTT Cell Proliferation Assay measures the cell

    proliferation rate and conversely, when metabolic events lead to apoptosis or

    necrosis, the reduction in cell viability. The MTT Reagent yields low background

    absorbance values in the absence of cells.

  • 22

    | 22

    5mg/ml stock solution of MTT was prepared in PBSA and used to prepare a

    1mg/ml solution of MTT in growth medium, to be applied to wells in 200µL

    aliquots. The plate was incubated for 3 hours before the MTT was removed and

    replaced by 200µL of DMSO to dissolve MTT formazan. The plates were agitated

    on a shaker for 5 minutes, before the absorbance was read at 540nm on a

    multiwell scanning spectrophotometer. The values obtained were used to

    determine the percentage inhibition of cell growth that these drugs cause.

    The treatment solutions (100µM) were made up in pre-warmed (37°C) RPMI

    1640 medium containing 10% foetal calf serum. The samples were vortexed for

    approximately 1 minute, sonicated for 10 minutes and then briefly vortexed again

    before it was ready for use. The 100µM solutions was filtered and sterilized using

    0.22µm syringe filter units. A 10x dilution was prepared in order to obtain a 10µM

    solution of the treatment. These solutions was applied to cells in 200 µL aliquots

    for 48 hours.

    All of the complexes synthesized went through a preliminary screening on the cell

    lines using the MTT Assay. The results obtained determined which complexes

    were used for further experiments.

    Dose response curves

    Dose response curves were prepared for each of the chosen complexes, as well

    as positive controls, in order to obtain IC50 values. The concentrations used was

    100, 50, 25, 10, 5 and 1 µM. IC50 values were calculated from the log-dose

    response curves using GraphPad Prism 4.

    2.1.6 Methods applied for the Ionization Studies

    Methods applied for ionization studies on (NS)-platinum(II) complexes- Following

    verification of structure by infusion mass spectrometry, the samples were ionized

    to determine the individual ionization rates. Two different methods were used as

    there was to be found a large difference in the ionization rates of the dichlorides

    and the oxalates. The method utilised to monitor the ionization of the dichlorides

    consisted of first dissolving the sample in a suitable solvent (600 µM/ CH3OH)

  • 23

    | 23

    where after an aliquot of the methanolic solution (50 µL) was diluted with water

    (450 µL) immediately prior to analysis. The dilution step took an average of 25-

    30s. The solution (5 µL) was then injected directly into the mass spectrometer at

    10s intervals. A total of 8 to 10 injections were performed.

    The oxalates were initially treated as the dichlorides above, but it was found that

    no ionization took place during the short time allowed for the fast loop test (ca.

    6min). Subsequently fresh samples were suspended in water (ca. 1mM) and the

    solutions were infused in a baseline spectrum. The sample was then allowed to

    stand at room temperature for 6 hours before a second mass spectrum was

    obtained. Finally the sample was placed in an oven at 80ºC for a 60 hour period

    prior to final analysis.

    Methods applied for ionization studies on (NN)-platinum(II) complexes –

    Experiments were performed to evaluate the ionization rates of a set of bidentate

    (NN)-dihalideplatinum(II) complexes and compare this with the data obtained for

    bidentate (NS)- dihalideplatinum(II) complexes.

    Methods - Structural verification was performed by infusing a solution of each

    compound in CH3CN (600μM) directly into the mass spectrometer. Unique

    molecular ions were obtained demonstrating that the correct structure had been

    prepared during synthesis and that the energy imparted by the instrument during

    the electronspray process was not excessive. The CH3CN solutions were diluted

    in water to 60uM (90% water). This was done to check whether these compounds

    underwent rapid ionization as in the case of the analogous (NS)-compounds.

    To try and characterize the ionization rate of each individual compound, aqueous

    solutions were prepared by diluting the original CH3CN stock with water (1%

    CH3CN / 99% H2O) and analysed over time (3Hr). The formation of the ionization

    product(s) [M-X+CH3CN]+ and the degradation of the molecular species [M+NH4]+

    were quantified, by integration of the peaks produced for each.

    Incubation method - The samples were dissolved in CH3CN (600μM). Prior to

    analysis (0.5min) an aliquot of the CH3CN solution was diluted with water (37ºC)

    to produce a solution of 6uM in 1% CH3CN (99% water). The samples were then

  • 24

    | 24

    placed in a heated tray (37ºC) and sampled periodically, typically monitoring

    transitions between 0.5 and 140min. The difficulty with this technique is that it can be

    influenced by instrument drift as the detector is on for the entire period. This implies

    that identical samples analysed 2 hours apart may show differences in the ion

    intensities and hence the perceived concentration of species. Such differences in ion

    intensities will detrimentally affect ionization curves leading to false ionization T1/2 times. The relative abundance of the species will however be reflected correctly.

    Therefore plotting the individual ion intensities as a function of the total ion current

    (TIC) was considered as a possible solution, the assumption being that all ionization

    and degradation products are detected and contribute to the TIC (unsafe

    assumption). The best way to compensate for the problem would be through the use

    of a suitable internal standard. In the absence of such a compound all steps were

    taken to ensure that the analysis time was kept to a minimum

    2.2 PREPARATION OF STARTING MATERIALS 2.2.1 K2PtCl6

    Pure platinum metal (99.99%) (10 g, 51 mmol) was dissolved in a 1:1 mixture of

    aqua regia and distilled water at an elevated temperature. The solution was

    concentrated by heating to a silvery paste. The latter was dissolved in

    concentrated HCl (100 ml) and again concentrated to a paste. This last step was

    repeated. The final paste was dissolved in 1M HCl and made up to 250 ml in a

    volumetric flask to act as a stock platinum solution.

    Chemically pure potassium chloride (KCl, 0.85 g, 11.4 mmol) was dissolved in 10

    ml distilled water and added drop wise to 25 ml stock platinum solution (+/- 1 g,

    5.1 mmol) while stirring. An equal volume of 95% ethanol was added to the

    platinum mixture and allowed to stand on ice for 45 minutes. The resultant yellow

    crystalline K2PtCl6 was filtered and washed twice with 20 ml portions of 50%

    ethanol, once with 20 ml 95% ethanol and 3 times with 20 ml portions of ether

    where after it was dried in an oven at 50°C. Yield: 95%

    2.2.2 K2PtCl4

    K2PtCl6 (4.7 g, 9.67 mmol) was suspended in 35 ml distilled water over a 90°C

  • 25

    | 25

    water bath. A saturated aqueous solution of sulfur dioxide (SO2) was added in

    0.6 ml portions over 2-3 minute intervals. The time intervals gradually lengthen

    as the reduction nears completion. The next addition can be performed when no

    more SO2 is emitted from the platinum(II) solution. Addition of SO2 was stopped

    when only a very small portion of solid K2PtCl6 remained in a very red solution.

    The platinum solution was filtered while hot to remove the unreacted K2PtCl6 after

    which the supernatant was concentrated and allowed to cool. The resultant red

    crystalline K2PtCl4 was filtered, washed once with 3 ml acetone and allowed to air

    dry in a 50°C oven.

    Yield: 65%

    2.2.3 K2Pt(C2O4)2.2H2O

    K2PtCl4 (2.08 g, 5.0 mmol) and 16 equivalents of K2(C2O4)2 (14.72 g, 79.9 mmol)

    were dissolved in 30 ml distilled water at 90°C and stirred for 1 hour maintaining

    the temperature at 90°C after which the reaction was allowed to cool for 2 hours

    at 5°C. The resultant K2Pt(C2O4)2.2H2O precipitate which formed was filtered and

    washed 5 times with 15 ml distilled water, once with 10 ml acetone and allowed to

    air dry at 50°C in an oven.

    Yield: 95%

    2.3 SYNTHESIS OF THE (NS)-DONOR LIGANDS 2.3.1 1-Methyl-2-methylthioalkylimidazole

    These ligands were prepared in three consecutive steps. All these compounds

    were prepared in the same manner as described below. The difference was that

    in the final step the appropriate alkylthiol was used.

    2.3.1.1 1-Methyl-2-methylthiomethylimidazole(mmtmi)

    Step 1:

    A solution of 1-methylimidazole (50 g, 0.61 mol) dissolved in1000 ml of 40 %

    formaldehyde was refluxed for 7 days. Sodium hydroxide (60 g, 1.5mol) and 2

  • 26

    | 26

    litres of water were subsequently added