synthesis of novel dual acting cardio- protective...

311
Synthesis of Novel Dual Acting Cardio- protective Agents VIRAJ H MANKAR [Masters of Science (Drug Chemistry)] A Thesis Submitted in Fulfilment of the Requirements for the Degree of Doctor of Philosophy. School of Chemistry, Physics and Mechanical Engineering Science and Engineering Faculty Queensland University of Technology Brisbane, Australia [2016]

Upload: hoangque

Post on 13-Apr-2018

217 views

Category:

Documents


4 download

TRANSCRIPT

Synthesis of Novel Dual Acting Cardio-

protective Agents

VIRAJ H MANKAR

[Masters of Science (Drug Chemistry)]

A Thesis Submitted in Fulfilment of the Requirements for the Degree of

Doctor of Philosophy.

School of Chemistry, Physics and Mechanical Engineering

Science and Engineering Faculty

Queensland University of Technology

Brisbane, Australia

[2016]

II

Keywords

Adenosine, adenosine receptors, allosteric modulator, antioxidants, chemistry,

nitroxides, free radicals, synthesis, cardio-protection, ischaemia, oxidative

stress, dual-acting drug.

III

Abstract

Adenosine and adenosine derivatives are recognised as cardioprotective

agents due to their ability to activate myocardial adenosine receptors.

Antioxidants such as nitroxide radicals have also been demonstrated to provide

cardioprotective effects by scavenging oxygen based damaging free radicals.

In this thesis, three series of C2-linked adenosine 5-N-ethylcarboxamide

derivatives bearing antioxidant nitroxide and phenolic moieties were designed

and synthesised. It was envisaged that linking of the antioxidant moiety to the

adenosine skeleton would produce adenosine analogues that bind to and

activate adenosine receptors and, in addition, scavenge harmful oxygen-

centered free radicals generated in the body.

Evaluation of the prepared compounds as A2AAR agonists using the cAMP

accumulation assay identified a number of potent and selective agonists. Of the

compounds examined, the adenosine 5–N-ethylcarboxamide compounds,

possessing various nitroxides linked through para-(2-aminoethyl)aniline

linkages (namely compounds designated as 162, 164, 166, 168 and 170 in this

Thesis) displayed high selectivity, with more than 200-fold selectivity for the

adenosine A2A receptors over adenosine A1 receptors, whilst retaining

reasonable selectivity over adenosine A3 receptors.

Of the adenosine compounds examined, PROXYL nitroxide (166) and tetraethyl

isoindoline nitroxide (170) possessing a para-(2-aminoethyl)aniline linked

adenosine 5–N-ethylcarboxamide unit approached high selectivity for the A2A

receptors over the other adenosine receptor subtypes. Four of the

IV

highly-selective adenosine compounds (162, 164, 166, and 170) were further

investigated in the simulated ischaemia model using rat atrial cardiomyoblast

cells. All four of these compounds showed strong protective effects in this

assay. There were some additional cardioprotective effects observed at the

highest agonist concentration in the presence of the antagonist. This result

indicates that the antioxidant moiety may contribute to the compounds

cardioprotective effects at high levels of agonist concentration.

Ethynyl- and ethyl-linked structural classes of adenosine 5–N-ethylcarboxamide

compounds were also synthesised bearing tetraalkylisoindoline nitroxides. All

compounds from these series are less-potent A2A agonists than the previously-

mentioned candidates. However, the ethynyl linked tetramethylisoindoline (178)

and tetraethylisoindoline nitroxide (182) bearing adenosine 5-N-

ethylcarboxamide and ethyl linked tetraethylisoindoline nitroxide bearing

adenosine 5-N-ethylcarboxamide (184) have a higher selectivity for A2B

receptors than other adenosine receptor subtypes.

This project has resulted in the synthesis and characterization of twelve novel

C2-substituted adenosine analogues with three different linker groups. Most of

the target compounds were tested for their binding ability to adenosine receptor

subtypes, and some analogues were found to be potent and selective

adenosine receptor subtype agonists.

V

Table of Contents

Keywords II

Abstract III

Table of Contents V

List of Figures XIV

List of Schemes XVI

List of Tables XIX

List of Abbreviations XX

Statement of Original Authorship XXVI

Acknowledgements XXVII

Chapter 1: Introduction & Literature Review 1

1.1 Cardiovascular diseases 2

1.1.1 Ischaemic heart disease 3

1.1.2 Reperfusion injury 5

1.1.3 Free radicals 6

1.1.4 Oxidative stress 7

1.2 Antioxidants 9

1.2.1 Nitroxide radical 13

1.2.2 Application of nitroxides 18

1.2.2.1 Radical trapping and spin labelling 18

1.2.2.2 Nitroxide antioxidants 20

1.2.3 Phenolic antioxidants 21

1.3 Adenosine 25

1.3.1 Structural features of adenosine 25

VI

1.3.2 Adenosine receptors 26

1.3.2.1 Adenosine A1 receptors 31

1.3.2.2 Adenosine A2A receptors 32

1.3.2.2 Adenosine A2B receptors 34

1.3.2.2 Adenosine A3 receptors 35

1.3.3 The concept of agonist and antagonists 36

1.3.3.1 Adenosine agonists 37

1.3.3.2 Adenosine antagonists 38

1.3.4 Active adenosine A2AAR agonists and antagonists in clinical

trials

40

1.3.4.1 Adenosine A2AAR agonists 40

1.3.4.2 Adenosine A2AAR antagonists 42

1.3.4.3 Adenosine A2BAR agonists 45

1.3.4.4 Adenosine A2BAR antagonists 46

1.4 Dual action adenosine agonists: -Rationale for the research 48

Chapter 2: Result and Discussion: Part A: Adenosine analogue with

para-aminophenylethylamine linker possessing antioxidant moieties

57

2.1 Preliminary considerations 58

2.2 Synthesis of C2-substituted TMIO adenosine analogues

possessing substituent attached to ‘aminoethylaniline

linker’ moiety (162)

64

2.2.1 Synthesis of antioxidant carboxy TMIO nitroxide

intermediates possessing substituent attached to

‘aminoethylaniline linker’ moiety (127)

65

2.2.1.1 Synthesis of antioxidant carboxy TMIO intermediate (106) 65

2.2.1.2 Synthesis of antioxidant carboxy TMIO linked

aminoethylaniline chain linker intermediate (127)

67

2.2.2 Synthesis of C2-substituted adenosine intermediates (146) 72

VII

2.2.2.1 Convergent approach to synthesis of adenosine

intermediate

73

2.2.2.2 Adenosine intermediate for C2-functionalization with a

Linear approach

75

2.2.3 Synthesis of TMIO adenosine analogue with

aminoethylaniline linker (161) (Approach 1).

90

2.2.4 Synthesis of TMIO adenosine analogue with

aminoethylaniline linker (161) (Approach 2).

94

2.3 Synthesis of C2-substituted CTEMPO adenosine analogues

possessing substituent attached to ‘aminoethylaniline linker’

moiety (164)

101

2.4 Synthesis of C2-substituted CPROXYL adenosine

analogues possessing substituent attached to

‘aminoethylaniline linker’ moiety (166).

104

2.5 Synthesis of C2-substituted MCTMIO adenosine analogues

possessing substituent attached to ‘aminoethylaniline linker’

moiety (168).

106

2.6 Synthesis of C2-substituted CTEIO adenosine analogues

possessing substituent attached to ‘aminoethylaniline linker’

moiety (170)

110

2.6.1 Synthesis of antioxidant carboxy TEIO intermediate (114) 110

2.6.2 Coupling of carboxy TEIO intermediate with

aminoethylaniline linked adenosine moiety (169)

111

2.7 Synthesis of C2-substituted di-tert-butylhydroxycinnamic

acid adenosine analogues possessing substituent attached

to ‘aminoethylaniline linker’ moiety (172)

114

2.7.1 Synthesis of antioxidant di-tert-butylhydroxycinnamic acid

linked ‘aminoethylaniline linker’ intermediate (133)

115

2.7.2 Synthesis of C2-substituted di-tert-butylhydroxycinnamic

acid adenosine analogue possessing substituent attached

by ‘aminoethylaniline linker’ moiety (171) (Approach 1).

118

2.7.3 Synthesis of C2-substituted di-tert-butylhydroxycinnamic

acid adenosine analogue possessing substituent attached

by ‘aminoethylaniline linker’ moiety (172) (Approach 2).

120

VIII

2.8 Result of Biological testing of adenosine analogue 123

2.8.1 Adenosine receptor binding assays 123

2.8.2 Simulated ischemia assay. 129

Chapter 3: Result and Discussion: Part B: Adenosine analogue

with ethynyl linker possessing antioxidant moieties.

132

3.1 Synthesis of C2-substituted TMIO adenosine analogues

possessing substituent attached to ‘ethynyl linker’ moiety

(178)

134

3.1.1 Synthesis of the 2-iodo-2,3-O-isopropylideneadenosine-5-

N-ethylcarboxamide intermediate (149).

134

3.1.2 Synthesis of ethynyl TMIO (ETMIO) intermediate (120). 137

3.1.3 Coupling of ETMIO with iodo-adenosine intermediate(178). 139

3.2 Synthesis of C2-substituted TEIO adenosine analogues

possessing substituent attached to ‘ethynyl linker’ moiety

(182)

143

3.3 Synthesis of C2-substituted phenyl adenosine analogues

possessing substituent attached to ‘ethynyl linker’ moiety

(185)

146

3.4 Result of biological testing of adenosine analogue 147

Chapter 4: Result and Discussion: Part C: Adenosine analogue

with ethyl linker possessing antioxidant moieties

151

4.1 Synthesis of C2-substituted TMIO adenosine analogues

possessing substituent attached to ‘ethyl linker’ moiety (180)

153

4.2 Synthesis of C2-substituted TEIO adenosine analogues

possessing substituent attached to ‘ethyl linker’ moiety (184)

156

4.3 Synthesis of C2-substituted non-nitroxide phenyl adenosine

analogues possessing substituent attached to ‘ethyl linker’

moiety (188)

158

4.4 Result of Biological testing of adenosine analogue 161

Chapter 5: Conclusion 165

5.1 Synthetic chemistry 166

IX

5.1.1 Adenosine based antioxidant compounds with para-

aminoethylaniline linker

167

5.1.2 Adenosine based antioxidant compounds with ethynyl linker 168

5.1.3 Adenosine based antioxidant compounds with ethyl linker 169

5.2 Biological analysis 170

5.2.1 Adenosine based antioxidant compounds with para-

aminoethylaniline linker

170

5.2.2 Adenosine based antioxidant compounds with ethynyl linker 172

5.2.3 Adenosine based antioxidant compounds with ethyl linker 172

Chapter 6: Future work 174

Chapter 7: Experimental 177

7.1 General experimental 178

7.2 Synthetic procedures 180

7.2.1 Synthesis of 2-benzyl phthalimide (100) 180

7.2.2 Synthesis of 2-benzyl-1, 1, 3, 3- tetramethylisoindoline (101) 181

7.2.3 Synthesis of 5-bromo-1,1,3,3-tetramethylisoindoline (102) 182

7.2.4 Synthesis of 5-bromo-1, 1, 3, 3- tetramethylisoindoline-2-

yloxyl (103)

183

7.2.5 Synthesis of 5-iodo -1, 1, 3, 3- tetramethylisoindoline (104) 185

7.2.6 Synthesis of 5-iodo-1, 1, 3, 3- tetramethylisoindoline-2-yloxyl

(105)

186

7.2.7 Synthesis of 5-carboxy-1, 1, 3, 3- tetramethylisoindoline-2-

yloxyl (106)

187

7.2.8 Synthesis of 2-benzyl-5-methylphthalimide (108) 189

7.2.9 Synthesis of 2-benzyl-5-methyl-1,1,3,3-tetraethylisoindoline

(109)

190

7.2.10 Synthesis of 5-methyl-1,1,3,3-tetraethylisoindoline (110) 191

X

7.2.11 Synthesis of 5-methyl-1,1,3,3-tetraethylisoindoline-2-yloxyl

(111)

192

7.2.12 Synthesis of 2-acetoxy-5-methyl-1,1,3,3-tetraethylisoindoline

(112)

193

7.2.13 Synthesis of 2-acetoxy-5-carboxy-1,1,3,3-

tetraethylisoindoline (113)

194

7.2.14 Synthesis of 5-carboxy-1,1,3,3-tetraethylisoindoline-2-yloxyl

(114)

195

7.2.15 Synthesis of 5-(3-hydroxy-3-methyl)butynyl-1,1,3,3-

tetramethylisoindoline-2-yloxyl) (119)

196

7.2.16 Synthesis of 5-ethynyl-1,1,3,3-tetramethylisoindoline-2-yloxyl

(120)

197

7.2.17 Synthesis of N-tert-butoxycarbonyl-2-(4-

aminophenyl)ethylamine (125)

198

7.2.18 Synthesis of N-tert-butoxycarbonyl-2-(4-N-(5-carboxy-

1,1,3,3-tetramethylisoindoline-2-

yloxyl)aminophenyl)ethylamine (126)

199

7.2.19 Synthesis of 2-(4-N-(5-carboxy-1,1,3,3-

tetramethylisoindoline-2-yloxyl)aminophenyl)ethylamine

(127)

201

7.2.20 Synthesis of (E)-tert-4-(3-(3,5-di-tert-butyl-4-

hydroxyphenyl)acrylamide)phenethylcarbamate (132)

202

7.2.21 Synthesis of (E)-N-(4-(2-aminoethyl)phenyl)-3-(3,5-di-tert-

butyl-4-hydroxyphenyl)acrylamide (133)

204

7.2.22 Synthesis of 2,3-O-isopropylideneguanosine (139) 205

7.2.23 Synthesis of 2,3-O-isopropylideneguanosine-5-carboxylic

acid (140)

206

7.2.24 Synthesis of 2,3-O-isopropylideneguanosine-5-

ethylcarboxylate (142)

207

7.2.25 Synthesis of 2,3-O-isopropylideneguanosine-5-N-

ethylcarboxamide (143)

209

7.2.26 Synthesis of O6-(benzotriazol-1-yl)-2′,3′-O-

isopropylideneguanosine-5′-N-ethylcarboxamide (144)

210

XI

7.2.27 Synthesis of 2-fluoro-O6-benzotriazole-1-yl-2,3-O-

isopropylideneguanosine-5-N-ethylcarboxamide (145)

211

7.2.28 Synthesis of 2-fluoro-2,3-O-isopropylideneadenosine-5-N-

ethylcarboxamide (146)

213

7.2.29 Synthesis of 2-iodo-O6-benzotriazole-1-yl-2,3-O-

isopropylideneguanosine-5-N-ethylcarboxamide (148)

214

7.2.30 Synthesis of 2-iodo-2,3-O-isopropylideneadenosine-5-N-

ethylcarboxamide (149)

215

7.2.31 Synthesis of 2-(2-(4-aminophenyl))ethylamino-2,3-O-

isopropylideneadenosine-5-N-ethylcarboxamide (159)

217

7.2.32 Synthesis of 2-(2-(4-N-(5-carboxy-1,1,3,3-

tetramethylisoindoline-2-yloxyl)aminophenyl))ethylamino-

2,3-O-isopropylideneadenosine-5-N-ethylcarboxamide

(161).

219

7.2.33 Synthesis of 2-(2-(4-N-(5-carboxy-1,1,3,3-

tetramethylisoindoline-1-

yloxyl)aminophenyl))ethylaminoadenosine-5-N-

ethylcarboxamide (162)

221

7.2.34 Synthesis of 2-(2-(4-N-(4-carboxy-2,2,6,6-

tetramethylpiperidine-1-yloxyl)aminophenyl))ethylamino-

2,3,-O-isopropylidineadenosine-5-N-ethylcarboxamide

(163)

223

7.2.35 Synthesis of 2-(2-(4-N-(4-carboxy-2,2,6,6-

tetramethylpiperidine-1-

yloxyl)aminophenyl))ethylaminoadenosine-5-N-

ethylcarboxamide (164)

224

7.2.36 Synthesis of 2-(2-(4-N-(3-carboxy-2,2,5,5-

tetramethylpyrrolidine-1-yloxyl)aminophenyl))ethylamino-2,

3,-O-isopropylidineadenosine-5-N-ethylcarboxamide (165)

226

7.2.37 Synthesis of 2-(2-(4-N-(3-carboxy-2,2,5,5-

tetramethylpyrrolidine-1-

yloxyl)aminophenyl))ethylaminoadenosine-5-N-

ethylcarboxamide(166)

227

XII

7.2.38 Synthesis of 2-(2-(4-N-(5-methylcarboxy-1,1,3,3-

tetramethylisoindoline-2-yloxyl)aminophenyl))ethylamino-

2,3-O-isopropylideneadenosine-5-N-ethylcarboxamide

(167)

229

7.2.39 Synthesis of 2-(2-(4-N-(methylcarboxy-1,1,3,3-

tetramethylisoindoline-2-

yloxyl)aminophenyl))ethylaminoadenosine-5-N-

ethylcarboxamide (168)

230

7.2.40 Synthesis of 2-(2-(4-N-(5-carboxy-1,1,3,3-

tetraethylisoindoline-2-

yloxyl)aminophenyl))ethylaminoadenosine-5-N-

ethylcarboxamide (169)

232

7.2.41 Synthesis of 2-(2-(4-N-(5-carboxy-1,1,3,3-

tetraethylisoindoline-2-

yloxyl)aminophenyl))ethylaminoadenosine-5-N-

ethylcarboxamide (170)

234

7.2.42 Synthesis of 2-(2-(4-N-(3,5-di-tert-butyl-4-

hydroxyphenyl)acrylamido) phenyl))ethylamino-2,3-O-

isopropylideneadenosine-5-N-ethylcarboxamide (171)

236

7.2.43 Synthesis of 2-(2-(4-N-(3,5-di-tert-butyl-4-

hydroxyphenyl)acrylamido) phenyl))ethylaminoadenosine-5-

N-ethylcarboxamide (172)

238

7.2.44 Synthesis of 2-((1,1,3,3-tetramethylisoindoline-2-yloxyl)-5-

ethynyl)-2,3-O-isopropylideneadenosine-5-N-

ethylcarboxamide (177)

240

7.2.45 Synthesis of 2-((1,1,3,3-tetramethylisoindoline-2-yloxyl)-5-

ethynyl)adenosine-5-N-ethylcarboxamide (178)

241

7.2.46 Synthesis of 2-((1,1,3,3-tetramethylisoindoline-2-yloxyl)-5-

ethyl)-2,3-O-isopropylideneadenosine-5-N-

ethylcarboxamide (179)

243

7.2.47 Synthesis of 2-((1,1,3,3-tetramethylisoindoline-2-yloxyl)-5-

ethyl)adenosine-5-N-ethylcarboxamide (180)

244

7.2.48 Synthesis of 2-((1,1,3,3-tetraethylisoindoline-2-yloxyl)-5-

ethynyl)-2,3-O-isopropylideneadenosine-5-N-

ethylcarboxamide (181)

246

XIII

7.2.49 Synthesis of 2-((1,1,3,3-tetraethylisoindoline-2-yloxyl)-5-

ethynyl)adenosine-5-N-ethylcarboxamide (182)

247

7.2.50 Synthesis of 2-((1,1,3,3-tetraethylisoindoline-2-yloxyl)-5-

ethyl)-2,3-O-isopropylideneadenosine-5-N-

ethylcarboxamide (183)

249

7.2.51 Synthesis of 2-((1,1,3,3-tetraethylisoindoline-2-yloxyl)-5-

ethyl)adenosine-5-N-ethylcarboxamide (184)

250

7.2.52 Synthesis of 2-(2-phenylethynyl)-2,3-O-

isopropylideneadenosine-5-N-ethylcarboxamide (185)

251

7.2.53 Synthesis of 2-(2-phenylethyl)-2,3-O-

isopropylideneadenosine-5-N-ethylcarboxamide (187)

253

7.2.54 Synthesis of 2-(2-phenylethyl)-2,3-O-

isopropylideneadenosine-5-N-ethylcarboxamide (188)

254

Chapter 8: Bibliography 256

XIV

List of Figures

Figure 1.1 The chemical structure of triphenylmethyl radical. 6

Figure 1.2 Dietary and synthetic antioxidants. 14

Figure 1.3 General chemical structure of a nitroxide. 14

Figure 1.4 Structures of stable nitroxide radicals. 16

Figure 1.5 The chemical structure of nitroxide, TEMPOL. 19

Figure 1.6 Phenolic and radical antioxidants 23

Figure 1.7 Examples of derivatives of hydroxycinnamic acids. 24

Figure 1.8 Structural features of adenosine. 26

Figure 1.9 Three-dimensional crystal structure of the G-protein coupled

receptor (GPCR) embedded in the cell membrane attached to

heterotrimeric G protein.

28

Figure 1.10 Intracellular adenosine synthesis. 29

Figure 1.11 Adenosine receptor activation. 30

Figure 1.12 Mode of action of agonists. 38

Figure 1.13 Mode of action of competitive and non-competitive antagonists. 39

Figure 1.14 Adenosine A2AAR agonists. 40

Figure 1.15 Pharmacologically active A2AAR agonists. 42

Figure 1.16 Adenosine A2AAR antagonists. 43

Figure 1.17 Comparison of receptor-ligand interaction binding for the A2AR

with the agonists adenosine (39) and NECA (62) along with

antagonists ZM 241385 (51).

44

Figure 1.18 Adenosine A2BAR agonists. 46

Figure 1.19 Adenosine A2BAR antagonists. 47

Figure 1.20 Dual action compounds effective for ischaemia. 48

Figure 1.21 Bifunctional adenosine receptor-binding analogues and binary

conjugates.

49

Figure 1.22 Pharmacologically active A2AAR and A3AR selective analogues. 51

Figure 1.23 Pharmacologically dual-action antioxidant coupled A2AAR and

A3 AR-selective analogues.

52

XV

Figure 1.24 Adenosine receptor crystal structure in complex with A) UK432,

097 (42) B) compound VCP728 (70).

53

Figure 1.25 Antioxidants coupled p-aminoethylaniline linked adenosine

target compounds.

55

Figure 1.26 Antioxidants coupled with ethynyl and ethyl linked adenosine

target compounds.

56

Figure 2.1 Adenosine A2AAR selective compound CGS21680 (46). 60

Figure 2.2 Candidate moieties and their actions in the first structural class

of target adenosine analogues.

63

Figure 2.3 Dual acting A2AAR agonists - antioxidants decrease cell death

within an in vitro model of simulated ischemia. A)

Concentration-dependent, decrease in cell death and B). The

decrease in cell death was inhibited in the presence of the

A2AAR antagonist, SCH442415.

130

Figure 3.1 Candidate moieties and their actions in the second structural

class of target adenosine analogues.

133

Figure 3.2 Adenosine A3AR agonists PENECA (186). 134

Figure 4.1 Candidate moieties and their actions in the third structural class

of target adenosine analogues

152

XVI

List of Schemes

Scheme 1.1 DPPH radical scavenging assay principle. 13

Scheme 1.2 Resonance stabilisation of a nitroxide radical. 14

Scheme 1.3 Bimolecular degradation of a phenyl substituted nitroxide. 15

Scheme 1.4 β-Hydrogen induced disproportion of nitroxide. 16

Scheme 1.5 Redox reaction of nitroxide. 18

Scheme 2.1 Overview of first approach synthesis of 161. 64

Scheme 2.2 Overview of second approach synthesis of 161. 65

Scheme 2.3 Synthesis of 5-carboxy-1,1,3,3-tetramethylisoindoline-2-yloxyl

(C-TMIO) 106.

66

Scheme 2.4 Overview of N-Boc protection of linker amine. 67

Scheme 2.5 Mechanism of N-Boc protection of linker amine. 68

Scheme 2.6 Overview of EDCI amide coupling of nitroxide to para-

(aminoethyl)aniline linker moiety (127).

70

Scheme 2.7 Mechanism of EDCI promoted amide formation. 71

Scheme 2.8 Convergent approach to adenosine A2AAR agonists. 74

Scheme 2.9 Overview of synthesis of the key adenosine intermediate. 76

Scheme 2.10 Overview of 2, 3-alcohol protection (139). 77

Scheme 2.11 Overview of the 5-hydroxyl oxidation of (140). 79

Scheme 2.12 Mechanism of primary-hydroxyl oxidation . 80

Scheme 2.13 Overview of 5-ethyl ester formation (142). 82

Scheme 2.14 Overview of 5-amide formation (143). 83

Scheme 2.15 Overview of leaving group formation (144). 85

Scheme 2.16 Mechanism of leaving group formation. 86

Scheme 2.17 Overview of fluoro substitution at the C2 position (145). 88

Scheme 2.18 Overview of N6 amino substitution (146). 89

Scheme 2.19 Overview of coupling of nitroxide with aminoethylaniline linker

to adenosine intermediate (Approach 1) (161).

91

XVII

Scheme 2.20 SNAr Mechanism for the synthesis of C2-substituted target

compound (161)

92

Scheme 2.21 Overview of coupling of carboxy nitroxides with

aminoethylaniline linked adenosine intermediate (161)

(Approach 2)..

95

Scheme 2.22 Overview of coupling of aminoethylaniline linker to adenosine

intermediate (159)..

95

Scheme 2.23 Synthesis of TMIO-substituted adenosine analogue

possessing a substituted aniline linker moiety (161)

(Approach 2).

98

Scheme 2.24 Synthesis of 3,5-dinitrobenzoic acid-coupled adenosine

analogue possessing a substituted aniline linker.

100

Scheme 2.25 Reaction of coupling reagents with adenosine analogue

possessing a substituted aniline linker.

101

Scheme 2.26 Synthesis of TEMPO-substituted adenosine analogue

possessing a substituted aniline linker moiety (164).

102

Scheme 2.27 Synthesis of proxyl-substituted adenosine analogue

possessing a substituted aniline linker moiety (166).

104

Scheme 2.28 Synthesis of MCTEIO-substituted adenosine analogue

possessing a substituted aniline linker moiety (168).

107

Scheme 2.29 Synthesis of 5-Carboxy-1,1,3,3-tetraethylisoindoline-2-yloxyl

(CTEIO) 114.

110

Scheme 2.30 Synthesis of CTEIO-substituted adenosine analogue

possessing a substituted aniline linker moiety (170).

112

Scheme 2.31 Overview of synthesis of di-tert-butylhydroxycinnamic acid

coupled aminoethylaniline linker moiety (133).

115

Scheme 2.32 Overview of coupling of di-tert-butylhydroxycinnamic acid with

linker to adenosine (Approach 1) (171).

118

Scheme 2.33 Synthesis of di-tert-butylhydroxycinnamic acid adenosine

analogue possessing a substituted aniline linker moiety

(Approach 2).

120

Scheme 3.1 Overview of synthesis of C2 iodo adenosine intermediate

(149).

135

Scheme 3.2 Synthesis of 5-ethyne-1,1,3,3-tetraethylisoindoline-2-yloxyl

(E-TMIO) (120).

138

XVIII

Scheme 3.3 Sonogshira coupling reaction mechanism. 140

Scheme 3.4 Synthesis of ETMIO-substituted adenosine analogue (178). 141

Scheme 3.5. Synthesis of ETEIO-substituted adenosine analogue (182). 144

Scheme 3.6. Synthesis of the phenylacetylene-substituted adenosine

analogue (185).

146

Scheme 4.1. Overview the synthesis of the TMIO-nitroxide ethyl linked

adenosine compound (180).

154

Scheme 4.2. Overview synthesis of TEIO-nitroxide ethyl linked adenosine

compound (184).

156

Scheme 4.3. Overview synthesis of the non-nitroxide phenyl ethyl linked

adenosine compound (188).

159

XIX

List of Tables

Table 2.1 cAMP analysis of aminoethylaniline linked adenosine

compounds

126

Table 3.1 cAMP analysis of ethynyl linked adenosine compounds. 148

Table 4.1 cAMP analysis of ethyl linked adenosine compounds. 163

XX

LIST OF ABBREVIATIONS

3 h — 3 hours

1 M — One molar

2 M — Two molar

4 M — Four molar

7TM — 7-Trans membrane

AR — Adenosine receptors

A1R — Adenosine A1 receptors

A2AR — Adenosine A2A receptors

A2BR — Adenosine A2B receptors

A3R — Adenosine A3 receptors

AMP — Adenosine monophosphate

ADP — Adenosine diphosphate

ATP — Adenosine triphosphate

ADORA2A — Adenosine A2A receptors

BAIB — Bis(acetoxy)iodobenzene

BOP—(Bezotriazole-1-yloxy)tris(dimethylamino)phosphonium

hexafluorophospohate

XXI

BHT — Butylhydroxytoluene

BOC — Butyloxycarboxyl

CDCl3 — Deuterated chloroform

13CNMR — 13C nuclear magnetic resonance

CHO — Chinese hamster ovary

CO2 — Carbon dioxide

cAMP — Cyclic adenosine monophosphate

CAD — Coronary Artery Disease

CHD — Chronic heart Disease

CRP — C-reactive protein

CSC — (E)-8-(3-chlorostyryl)caffine

CTMIO — 5-Carboxy-1,1,3,3-tetramethylisoindoline-2-yloxyl

CTEIO — 5-Carboxy-1,1,3,3-tetraethylisoindoline-2-yloxyl

CTEMPO — 4-Carboxy-2,2,6,6-tetramethylpieridine-1-yloxyl

CPROXYL — 3-Carboxy- 2,2,5,5-tetramethylpyrrolidine-1-yloxyl

CuI — Copper iodide

DABCO — 1,4-Diazabicyclo(2.2.2)octane

DBU — 1,8-Diazabicycloundec-7-ene

DCM — Dichloromethane

XXII

DIPEA — N,N-diisopropylethylamine

DMF — N,N-dimethylformamide

DMPX — 3,7-Dimethyl-1-propargylxanthine

DMAP — 4-Dimethylaminopyridine

DMSO — Dimethyl sulfoxide

DNA — Deoxyribonucleic acid

DPPH — 2,2-Diphenyl-1-picrylhydrazyl

DTBN — Di-tert-butylnitroxide

ET — Electron transfer

EtOAc — Ethyl acetate

EI — Electron ionisation

E-TMIO — 5-Ethynyl-1,1,3,3-tetramethylisoindoline-2-yloxyl

E-TEIO — 5-Ethynyl-1,1,3,3-tetraethylisoindoline-2-yloxyl

EDCI — 1-Ethyl-3-(3-dimethylaminopropyl)carbodiimide

Et2O — Diethyl ether

EtOH — Ethanol

EIMS — Electron ionisation mass spectroscopy

GDP — Guanosine diphosphate

GTP — Guanosine triphosphate

XXIII

GSH-Px — Glutathione peroxidase

GPCR — G-coupled protein receptor

H2O2 — Hydrogen peroxide

HCl — Hydrochloric acid

HCA — Hydroxycinnamic acid

HF — Hydrofluoric acid

HOBt — Hydroxy benzotriazole

HPLC — High-pressure liquid chromatography

HRMS — High-resolution mass spectroscopy

H9c2 — Rat cardiomyoblast cell line

HENECA — 2-Hexynyl-5-N-ethylcarboxamide

IL-4 — Interleukin-4

I2 — Iodine

IOP — Internal ocular pressure

Ki — Inhibitory constant

KMnO4 — Potassium permanganate

KOH — Potassium hydroxide

LC/MS — Liquid chromatography-mass spectroscopy

LDL — Low-density lipoprotein

XXIV

MeCN — Acetonitrile

MeOH — Methanol

MCTMIO — 5-Methylcarboxy-1,1,3,3-tetramethylisoindoline-2-yloxyl

MS — Mass spectroscopy

MPLC — Medium pressure liquid chromatography

MPI — Myocardial perfusion imaging

mRNA — Messenger ribonucleic acid

NADPH — Nicotinamide adenine dinucleotide phosphate

NaOH — Sodium hydroxide

Na2WO4.2H2O — Sodium Tungstate dihydrate

nM — Nanomolar

NECA — N-ethylcarboxamide

NMR — Nuclear magnetic resonance

NO — Nitric oxide

n-BuLi — n-Butyl Lithium

O2 — Oxygen

PbO2 — Lead dioxide

Pd — Palladium

Pd/C — Palladium on carbon

XXV

Pd(PPh3)2Cl2 — Palladium bis(triphenylphosphene) dichloride

PS — Petroleum spirit

PTSA — Para-toluene sulphonic acid

PROXYL — 2,2,5,5-Tetramethylpyrrolidine-1-yloxyl

pEC50 — Logarithimic 50% effective concentration

ROS — Reactive oxygen species

RNS — Reactive nitrogen species

SAR — Structure-activity relationship

SNAr — Nucleophilic aromatic substitution

SI — Simulated Iscaemia

SOD — Superoxide dismutase

TEA — Triethylamine

THF — Tetrahydrofuran

TEMPO — 2,2,6,6-Tetramethylpiperidin-1-yloxyl

TEMPOL — 2,2,6,6-Tetramethylpiperidin-1-yloxyl

TEIO — 1,1,3,3-Tetraethylisoindoline-2-yloxyl

TMIO — 1,1,3,3-Tetramethylisoindoline-2-yloxyl

TLC — Thin layer chromatography

XXVI

Statement of Original Authorship

The work contained in this thesis has not been previously submitted for a

degree or diploma from this or any other higher education institution. To the

best of my knowledge and belief, the thesis contains no material previously

published or written by another person except where due reference is made.

Signature

Date: ------------------------------------------

QUT Verified Signature

XXVII

Acknowledgements

Thanks to Professor Steven Bottle for his guidance and continuous support for

allowing me to move thousands of kilometres away from my home country and

further develop my skills and expertise.

Thanks to Professor Peter Scammells and co-workers from Monash Institute of

Pharmaceutical Sciences Melbourne for the biological testing of my synthesised

chemical compounds and providing guidance and their assistance throughout

the project.

Thanks to Dr Kathryn Fairfull-Smith for her valuable guidance and support, and

help in understanding the policies and procedures in QUT.

Thanks to Dr James Blinco and Dr Kye Masters for their support inside and

outside the laboratory.

Thanks to my parents and siblings for their love and support and

encouragement throughout the Ph.D. tenure.

Thanks to all friends and colleagues Mr Komba Thomas, Mr Liam Walsh, Mr

Jason Morries, Ms Vanessa Luccini, Mr Paul Lederhose, Mr. Chong, Mr Viraj

Jayawardhana and Mr Kai Anders-Hanson for their support and friendly

environment in and outside the laboratory.

Thanks to all other inhabitants of M6 laboratory who directly or indirectly helped

me.

Thanks all the QUT chemistry technicians, Dr Chris Carvalho, Dr Mark Wellard

and all technical staff for their support in learning the instrument handling.

XXVIII

Thanks to QUT administrative staff and all research centre staff for their direct

or indirect support throughout my QUT life.

Thanks to QUT counselling and medical centre services for their support.

Thanks to professional editor Laurel Mackinnon, provided copyediting and

proofreading services, according to the guidelines laid out in the university-

endorsed guidelines and the Australian standards for editing research theses’.

Thanks to the Australian Research Council and Centre of Excellence for Free

Radical Chemistry and Biotechnology for a living allowance scholarship.

1

Introduction & Literature Review 1

2

1.1 Cardiovascular diseases

Cardiovascular diseases are a leading public health concern throughout the

world. Globally, more than 17 million people die from cardiovascular diseases

every year.1 Of these, 80% of deaths occur in low-income and middle-income

people or developing countries. Heart disease, a type of cardiovascular

disease, is the most common cause of death in developed countries.1

Cardiovascular disease is classified further as peripheral vascular diseases and

cerebrovascular diseases, and includes coronary heart disease (CHD),

rheumatic heart disease, stroke, chronic heart failure, heart attack (myocardial

infarction), and diabetes.2

The pathophysiology of cardiovascular diseases is very complex because of the

involvement of several biological pathways, which all originate in the vascular

endothelium.3 Interference in these pathways is considered to be the main

factor in cardiovascular risk. Activation of the vascular endothelium and

oxidative stress play important roles in atherosclerosis4 and hypertension,5, 6

which eventually result in the progression of cardiovascular damage. High

cholesterol levels,7 impaired glucose tolerance8 and inflammation also

contribute to cardiovascular damage.

During the past several decads, extensive research has focused on treating

cardiovascular diseases from a different perspective such as the use of

precautionary therapies and curative medications. Several factors have been

found to contribute to cardiovascular diseases. These factors are classified into

3

two types: unavoidable and avoidable factors. Unavoidable factors include age,

sex, menopausal status and genetic composition. Avoidable factors can include

environmental pollution, diet, smoking, some diseases (e.g., diabetes,

hypertension), lack of physical activity9 and ethanol consumption.10

Cardiovascular diseases result from an inadequate blood supply to the brain,

heart and peripheral vasculature.2 The term ischaemia describes the deficiency

or shortage of blood supply (to hold back blood).11 These ischaemia-related

heart diseases are the most common cause of death in the world,12 and

research has focused on finding new ways to design and synthesise drugs that

will achieve cardioprotection.

1.1.1 Ischaemic heart diseases

Coronary artery disease (CAD) is also called ischaemic heart disease.12

According to the World Health Organization, in 2008 ischaemic heart disease

accounted for 7.25 million deaths worldwide and, by 2015, 25% of the global

adult population will be affected by the major risk of hypertension.13 Ischaemic

heart disease involves a narrowing of the coronary arteries, the vessels that

supply blood to the mayocardium, mainly because of deposition of plaque in the

arterial walls, a process called atherosclerosis.14 Plaques are composed of

cholesterol-rich fatty deposits, collagen, other proteins, and excess smooth

muscle cells.14 Thickening and narrowing of the arterial walls, as well as

impeding the flow of blood and starving the heart of oxygen, are the symptoms

of this disease (also called “ischaemia”).15 This disease can cause a muscle

cramp-like chest pain called angina pectoris.16

4

Ischaemia occurs where there is an interruption or occlusion of the blood supply

to an organ or tissues, which deprives the cells of oxygen and nutrients, and the

ability to generate energy, a condition called hypoxia.11 This hypoxia can

initiate a series of chemical events that result in cell dysfunction, cellular chaos,

interstitial oedema, and necrosis, and eventually lead to anaerobic cell death.11

Hypoxia can affect various parts of the body, resulting in cardiac ischaemia,

brain ischaemia,17 cutaneous ischaemia, and ischaemia of the bowel and

limb.11 Hypoxia can cause serious disorders by damaging metabolically active

tissues and eventually various organs linked by the circulatory system.11 A

deficiency of oxygen supply (hypoxia) increases the intercellular concentration

of lactic acid through a process called acidosis, which alters normal enzyme

kinetics within cells.11

Ischaemia often occurs during surgical procedures, especially vascular

procedures and transplantations.18 Abnormalities can be detected in the

electrocardiogram (ECG), which may show the first indications of ischaemic

heart disease.18 These abnormalities may occur even with an absence of

symptoms of angina or myocardial infarction and may indicate the possible

presence of ischaemic heart disease.18 In hypoxic muscle cells, glucose

consumption can increase by 12-fold.15 The depletion of cellular energy stores,

particularly of adenosine 5-triphosphate (ATP) is considered to reflect the

failure of cellular homoeostasis and is diagnosed by the loss of the ion gradient

across the cell membrane.15

5

Hypoxia develops within 10 seconds of the coronary blockage.11 Different

tissues can remain hypoxic for different periods. Skeletal muscles can recover

after some period of ischaemia; however, irreversible neuronal injury typically

occurs within minutes.19 Cardiac cells can survive for about 20 minutes under

ischaemic conditions and, if blood flow is restored within this time, the normal

aerobic mechanism resumes.15 In aerobic condition, the toxic metabolites are

removed from the site of the ischaemic event. However, this restoration of the

blood supply may also lead to further local tissue injury.11 Ischaemia and

reperfusion injuries are the root causes of the pathophysiology of cerebral

ischaemia, stroke, myocardial infarction and some of the problems with organ

transplantation.20

1.1.2 Reperfusion injury

Reperfusion is the restoration of blood supply to ischaemic tissues, which

facilitates the removal of toxic metabolites from these tissues.11 These toxic

metabolites flow into and throughout the systemic circulation and can lead to

serious injury including ischaemic tissue injury, called ischaemia–reperfusion

injury.11 This restoration of blood initiates a series of events that can lead to

further tissue injury.21 Once the blood supply is restored, a cascade of

biochemical and molecular changes occur that lead to free radical-mediated

damage.21 Oxygen-centred free radicals, including superoxide radicals, are

mainly involved in ischaemia–reperfusion injury.21 These free radicals are

produced intracellularly within the mitochondria in the post-ischaemic

endothelium and may be responsible for major changes in the endothelium.15

Superoxide radicals may be responsible for the production of highly reactive

6

and damaging hydroxyl radicals.22 Superoxide radical production is controlled

naturally by the endogenous enzyme superoxide dismutase in normal and non-

stressed physiological conditions.23-26 This natural defence mechanism is

overwhelmed in ischaemia and reperfusion conditions, and the hydroxyl radicals

produced can cause damage to a variety of biological components including

amino acids, proteins, enzymes and nucleic acids.22, 27-30

1.1.3 Free radicals

Free radicals are ions, atoms or molecules with one or more unpaired electrons

in their outer shell. Radicals are formed by homolytic bond cleavage. Free

radicals can react readily with other radicals to form stable species and can be

oxidised to cations or reduced to anions. In 1900, the first organic free radical

triphenylmethyl radical (Figure 1.1) was identified by Moses Gomberg.31

Figure 1.1: Chemical structure of a triphenylmethyl radical.

Free radicals are generated in biological systems by a wide variety of processes

such as ionising radiation, inflammation or exposure to toxic xenobiotics, and as

metabolites of membrane lipid transformation.32 These radical species also can

be generated by external sources including cigarette smoke33 and other forms

of pollution, by exposure to radiation and by exposure to chemical processes

7

such as those involving photosensitized decomposition and the decomposition

of non-endogeneous diacetyl peroxide, peresters and azo compounds.34

However, not all radicals are highly reactive or damaging. Free radicals can

also play an important role as mediators of cell signalling processes that

regulate vascular function.35, 36

1.1.4 Oxidative stress

Oxidation and reduction reactions represent the basis for several biochemical

mechanisms that generate metabolic changes in biological systems.37

Oxidative stress is defined as the imbalance between the increased production

of reactive oxygen species (ROS) and reactive nitrogen species (RNS) and the

lowered activity of radical-scavenging mechanisms in biological systems.38

Oxidative stress is involved in the various mechanisms of injury in many types

of disease processes. The free radical species ROS and RNS are produced in

low amount in all layers of the vascular membrane, such as the endothelium

and smooth muscle and in several metabolic processes via the actions of

various enzymes.39 These enzymes include nicotinamide adenine dinucleotide

phosphate (NADPH) oxidase, xanthine oxidase, nitric oxide (NO) synthases,

myeloperoxidase (MPO) and some mitochondrial enzymes.40

The involvement of ROS and RNS is widely recognised as being important in

several aspects of pathophysiological processes.39, 41 ROS are known to play

an important role in oxidative stress. Increased oxidative stress can cause

damage to cellular structures. The overproduction of radical species (the active

8

form of oxygen or nitrogen) can damage biological molecules including proteins,

amino acids, nucleic acids and DNA molecules by various mechanisms.42

The toxic effects of oxygen were investigated in several animal species. In

1954, Gerschman et. al. reported that the observed oxygen toxicity could be

attributed to the partially reduced form of oxygen.43 These ROS and RNS

encompass hydrogen peroxide (H2O2), superoxide anion radicals (O2–),

hydroxyl radicals (OH), nitric oxide (NO), and peroxynitrite (ONOO–) radicals.44,

45 An excess of the active forms of these radicals (R) or other ROS, leads to

the oxidation of other cellular components and forms the peroxyl radical

(HOO), which can initiate the chain oxidation reaction. This radical chain

process leads to more damage and generates oxidative stress conditions.

ROS are responsible for the increased oxidative stress and decreased lifespan

in various living species.46 The free radical theory of ageing states that the

organism’s age depends on the accumulation of damage over time, attributed to

oxidative stress induced by ROS.47 This theory is supported by the imbalance

between the ROS produced by mitochondria, and the activity of internal radical-

scavenging defence mechanisms.48

Physical exercise, can also increase oxidative stress. Intense physical activity

increases mitochondrial activity in skeletal muscles and the subsequent

production and leakage of ROS from mitochondria.49 Ischaemia–reperfusion

injury and activation of neutrophils, may occur during intense physical exercise,

and also further increase in the production of ROS.50 Cigarette smoke can also

9

increase oxidative stress because it contains a large number of free radicals

that can cause endothelial cell dysfunction and mitochondrial damage.33, 51

In living organisms, free radicals may cause more damage to the immature

brain than the adult's brain because of underdeveloped free radical-scavenging

mechanisms and the presence of a higher concentration of iron that can

promote radical formation.17 As a result, free radical damage in infants can

cause neurological complications such as birth asphyxia, perinatal brain injury

and ischaemic injury etc.17 However, antioxidant analogues are recognised to

provide protective effects for this type of free radical damage.

1.2 Antioxidants

Antioxidants are compounds that prevent or defend against the cellular damage

caused by free radicals via inhibition of oxidation of atoms or molecules.32

Oxidation is a chemical reaction in which loss of a hydrogen atom or an electron

or addition of oxygen to a molecule.52 Oxidation can generate free radicals,

which then initiate a chain radical reaction that can damage the cellular

components of biological systems.53 Antioxidants terminate this chain reaction

by scavenging the free radical intermediates and preventing further oxidation of

other species.53 The antioxidant activity of species such as thiols, ascorbic acid

and polyphenols is due to their ability to act as reducing agents (i.e. they

undergo oxidation themselves).53 The antioxidant mechanism of

cardioprotection typically involves either the generation of lynchpin radicals in

ischaemia–reperfusion injury or the direct scavenging of the superoxide anion.54

Clinical studies have shown that anti-inflammatory agents act as antioxidants

10

significantly reduce the levels of C-reactive protein (CRP) and membrane attack

complex (MAC) protein in infarcted tissues.54 These CRP and MAC protein

levels increase during inflammatory and cardiovascular conditions.55

In biological systems, the potential damaging effects of free radicals are

regulated through intracellular body fluids by the actions of several enzymes

including superoxide dismutase (SOD), glutathione peroxidase (GSH-Px), and

glutathione-S-transferase.56 The family of glutathione-S-transferase enzymes

catalyses the transformation of thiols, glutathione and some reactive

electrophiles to thioethers called S-conjugates.56 These reactive intermediates

are formed by several mechanisms including NADPH-dependent enzymatic

dethiolation and thiol exchange or radical-mediated processes.56

Non-enzymatic defence mechanisms act by preventing the excessive

production of free radicals. The body employs several means to prevent the

overproduction of free radicals. The first method is metal chelation, which

involves binding metal ions, particularly iron and copper ions, at the site of

initiation of the chain reaction. This method is a potent technique for preventing

the radical production and it is used as a major strategy for preventing lipid

peroxidation and DNA fragmentation.57 Metal binding to the proteins transferrin,

ceruloplasmin and metallothionein is broadly effective to counteract radical

generation. A second process involves the modification of the target site to

make it resistant to the metal ion-dependent oxidation.53 This process can

protect cells from radiation-derived oxidation; for example, to protect melanin

11

from ultraviolet radiation and carotenoid from electronically excited species such

as singlet oxygen.37

A third biological process that prevents the overproduction of free radicals is the

introduction of non-enzymatic antioxidants to protect the tissues. The main

issue is that once the radical species are formed, they initiate a radical chain

reaction and produce more radical compounds. This radical generation can be

prevented by the production of the non-radical and unreactive end products.

This process of deactivation of the radical chain reaction by inserting

antioxidants or other radical species prevents further damage.

Another strategy is to move radical species away from potential target species

to avoid cellular damage. High-efficiency breakup mechanisms are used in

cellular systems, where only a few antioxidant molecules are required to

deactivate thousands of potential target molecules. Such chain-breaking

mechanisms often involve polyphenolic antioxidants including -tocopherol,

which reduces peroxyl radicals and is itself converted to the tocopheroxyl

radical. This radical is then scavenged by external dietary antioxidants

including ascorbate vitamin C (2) and thiols.58 Several other naturally occurring

dietary antioxidants include retinol vitamin A (8), kaempferol (4) and quercetin

(5) (Figure 1.2).

12

Figure 1.2 Dietary and synthetic antioxidants.

A number of techniques can be used to measure the antioxidant activity of

phenolic compounds. All antioxidant compounds exhibit a moderate

electroactivity. The following electron transfer (ET) based assay evaluates the

capacity of an antioxidant to reduce an oxidant that usually changes colour

when reduced59. The ET based radical scavenging assay encompasses one of

the most popular antioxidant assays that contains 2,2-diphenyl-1-picrylhydrazyl

(DPPH) radical 9 as well known radical scavenger (Scheme 1.1). The ET

technique is also a powerful tool to measure the antioxidant activity of

hydroxycinnamic acid derivatives.60

13

Scheme 1.1: DPPH radical scavenging assay principle.

Several synthetic compounds have also been reported as radical scavengers

that have different mechanisms of action. After the discovery of superoxide

dismutase (SOD) activity in 1969, there has been a significant development in

the understanding of radical chemistry61 and, since 1988, in the arginine-

dependant synthesis of NO radicals.62 It has also been demonstrated that

nitroxides are very efficient radical scavengers that exhibit antioxidant actions.63

For example, several synthetic compounds reported as radical scavengers

including AAD-2004 (6)64, Neu-2000 (3)65 and NXY-059 (7)66, 67 (Figure 1.2)

exhibit anti-inflammatory and antioxidant properties by spin trapping in cortical

neuron culture.

1.2.1 Nitroxide radicals

Nitroxides (also known as aminoxyls or nitroxyls) are synthetically stable free

radicals. In 1965, Griffith and McConnell demonstrated that nitroxides are free

radical species with paramagnetic properties.68 Nitroxides exhibit paramagnetic

behaviour because of an unpaired electron. The general molecular formula is

R2NO (Figure 1.3).

14

Figure 1.3: General chemical structure of a nitroxide.

The majority of stable nitroxides are secondary amine N-oxide radical species

with quaternary α–carbon atoms. The stability of a nitroxide radical depends on

several factors. The delocalization of an unpaired electron (resonance) over the

N–O bond and hetero atoms provides remarkable kinetic and thermodynamic

stability to the nitroxide moiety (Scheme 1.2). Therefore, it shows resistance to

dimerization.69

Scheme 1.2: Resonance stabilisation of a nitroxide radical.

The groups attached to the radical centre can also strongly influence the

stability of the nitroxide radical. Nitroxides become more stable with bulky

groups around the nitroxyl moiety. This steric effect resists the bimolecular

combination reaction that occurs between a nitroxide and a more reactive

radical.

The stability of nitroxides is also maintained by their resistance towards the

formation of stable adducts with other oxygen centred radical species. Although

nitroxides can undergo reaction with a variety of reactive oxygen species, this is

not through direct interaction with the oxygen centred radicals. Dimerization

does not occur due to the loss in delocalisation stabilisation of two nitroxide

15

species. The dimerization of two nitroxide molecules is also energetically

unfavourable (2 x ~130 kJ/mol) because of the weak nature of the resultant N–

O–O–N bond (~140 kJ/mol) that would form.70, 71 This nature maintains the

stability of nitroxides and is also useful for protecting the nitroxide radical from

dimerization.72, 73

The stability of nitroxides is dependent on the unpaired electron remaining

delocalised over the two hetero atoms. The nitroxide stability is, therefore,

dependent on the absence of degradation pathways, which mainly arise through

the substituents present on the carbon α to the nitroxide moiety.74 The

delocalisation of unpaired electron spin density in the aromatic ring of tert-butyl

phenyl nitroxide increases the thermodynamic stability of the nitroxide system.

However, the reactive carbon centred radical thereby formed promotes the

bimolecular degradation of the nitroxide moiety to give the corresponding amine

and nitrone species74 (Scheme 1.3).

Scheme 1.3: Bimolecular degradation of a phenyl substituted nitroxide.

The degradation pathways that affect the stability of nitroxides arise mainly due

to the presence of methyl (CH3-), methylene (RCH2-) and methine (R2CH-)

16

hydrogen atoms adjacent to the nitroxide moiety. Nitroxides possessing β–

hydrogen atoms become unstable because of facile disproportion to give the

hydroxylamine and nitrone (Scheme 1.4).74

Scheme 1.4: β–Hydrogen induced disproportion of a β–H substituted nitroxides.

Non-cyclic bis(tert-alkyl) nitroxides such as di-tert-butylnitroxide (DTBN), as well

as cyclic nitroxides, are stable species also known as aminoxyls and nitroxyls.

These stable radicals possess methyl groups (that have no β–Hydrogen) on the

carbons α to the nitroxide.75 Methyl groups impact stability to the nitroxide

radical by limiting access to reactive species and avoiding radical dimerization.

There are several common types of cyclic nitroxide radicals that have been

exploited in research studies including, 1,1,3,3-tetramethylisoindoline-2-oxyl

(TMIO); 2,2,5,5-tetramethylpyrrolidine-1-oxyl (PROXYL) and 2,2,6,6-

tetramethylpiperidine-1-oxyl (TEMPO) nitroxides (Figure 1.4).

Figure 1.4: Structures of stable nitroxide radicals.72, 73

17

Nitroxides have been reported as effective ROS scavengers with some

advantages over other reactive radical trapping agents. Specifically, the

isoindoline-based aminoxyls, namely 1,1,3,3-tetramethylisoindoline-2-oxyl

(TMIO)76 (22) and its analogues, are known to have several advantages77

compared to other nitroxides including the PROXYL (25) and TEMPO (24)

containing species. The fused aromatic ring of the isoindoline skeleton provides

resistance to ring-opening reactions over the decomposition pathways of other

aminoxyls and demonstrates excellent electron paramagnetic resonance (EPR)

properties with narrower line-widths. In addition, these nitroxides demonstrated

excellent thermal and chemical stability in several chemical reactions.

Substitution on the aromatic ring of the isoindoline allows the creation of more

complex structures suitable for several applications with little impact on the

stability and reactivity of nitroxide radical species.78, 79 These modifications of

isoindoline nitroxide can be beneficial in increasing the water solubility of

nitroxides, and this has been used to expand the potential scope of the

application of nitroxides in various therapeutic contexts.80 Isoindoline nitroxides

are implicated in radical trapping and as potent antioxidants in biological

systems.81, 82

As nitroxides are more stable than other reactive radical species, their use has

been exploited in various applications. Nitroxides demonstrate their effect

through electron transfer mechanisms, including redox metabolism via one or

two electron-reduction reactions involving either an interconvertible

hydroxylamine, an oxoammonium cation or a radical ion. These three-stage

18

oxidative electron-transfer processes also explain the ROS-metabolizing action

of nitroxides (Scheme 1.5).81, 83

Biological studies have found that some nitroxides can accumulate in cells at

high concentration with lifetimes of over 100 minutes.84 This property of

nitroxides has been exploited in EPR imaging and biomarkers.75 These findings

have initiated further research into nitroxides and their applications in the

therapeutic field.85

Scheme 1.5: Redox reactions of a nitroxide.81, 83

1.2.2 Applications of nitroxides:

Radical trapping and spin labelling: 1.2.2.1

The biological activity of nitroxides was first investigated in 1964 by Emmerson

and Howard-Flanders.86 Further studies on nitroxides described the protective

antitumor activity of nitroxides,87 and also their roles as powerful bioregulatory

molecules in the nervous system, immune system and cardiovascular system.88

Nitroxides are kinetically stable systems compared with other radical species,

19

and their intracellular lifespan has been found to be long enough to be detected

by electron paramagnetic resonance (EPR) spectroscopy. Therefore, these

stable nitroxide radicals have been used in several animal models of disease

and also in human diseases to try to find ways to avoid the production of ROS,

which can generate oxidative stress.

The nitroxide TEMPOL (4-hydroxy-2, 2, 6, 6-tetramethylpiperidine-N-oxyl) 28

(Figure 1.5) is the most widely studied nitroxide, and it has been shown to be

permeable through cell membranes and to reduce the formation of toxic

hydroxyl radicals.89

Figure 1.5: The chemical structure of nitroxide, TEMPOL.

In 1990, Schnackenberg, Welch, and Wilcox90 reported that the intravenous

administration of TEMPOL decreased blood pressure and lipid peroxidation in a

hypertensive rat model. TEMPOL also reduced glutathione levels in human

blood cells,91 and was found to protect cells and tissues from the damaging

effects of ROS. In vivo, nitroxides rapidly undergo reduction to the

corresponding hydroxylamine, which demonstrates their involvement in redox

biochemistry.92

20

Nitroxide antioxidants 1.2.2.2

Nitroxides act as antioxidants against free radical-induced oxidative damage in

biological systems and provide chemo-protection and radioprotection.93

Extensive research has focused on the action of nitroxides as antioxidants in

several biological systems ranging from isolated molecules to membranes, cells

to organs and in whole animals.94, 95 These nitroxides can operate at different

levels in the cell, for example, preventing radical formation, intercepting formed

radicals, repairing oxidative damage, and increasing the elimination of damaged

molecules as well as promoting the death of cells with excessively damaged

DNA96. Antioxidant mechanisms are studied in cells using isolated

biomolecules created by microsomal preparation methods.97 Cell protection is

observed by preserving the colony-formation ability of the cells and by avoiding

DNA degradation. The mechanism proposed to explain this protection is that

nitroxides oxidise reduced metals and limit the production of H2O2 and,

consequently, the production of ROS.

Nitroxide radicals react with free radicals and intercept them before they can

damage lipids, proteins and DNA molecules. These radicals also act as

cytotoxic mediators in the immune system and as neurotransmitters in the

central nervous system.98 Nitroxide analogues and their derivatives have been

investigated as potential protective agents in rat models of myocardial

ischaemia–reperfusion injury.99 Experiments have confirmed that, under the

ischaemic condition, nitroxides are completely reduced in the heart, and this is

decreased by ~33% in non-ischaemic conditions.100

21

Several stable nitroxides have been reported to exhibit intracellular radical-

scavenging actions with recycling capacities.101, 102 These nitroxides were

developed and tested for their free radical-scavenging and antioxidant

abilities.63, 75, 76 TEMPO and its analogues have been shown to protect against

cerebral and cardiovascular ischaemia103 and reperfusion injury89 by decreasing

the infract size. This discovery of the actions of nitroxide radicals has led to an

interest in the role of dietary antioxidants and evidence suggesting that

antioxidants may protect against many neurological and cardiovascular

diseases.104-107 Several hindered phenolic and polyphenolic compounds also

exhibit antioxidant and anticancer activity.108-111

1.2.3 Phenolic antioxidants

Phenolic compounds also exhibit antioxidant properties similar to those of

nitroxides. In particular, hindered phenolic compounds have been reported to

demonstrate excellent antioxidant activity and are known as “natural”

antioxidants. These sterically hindered phenolic compounds have improved

reactivity towards small free radicals and are more biologically stable than non-

hindered phenolic species when oxidised. Phenolic antioxidants including

butylated hydroxytoluene (BHT) generate stable free radicals by donating a

hydrogen atom to diffuse the more reactive radical species. This phenoxy

radical is more stable, but it can still react with other radicals or can be

polymerised, although these reactions are slower than those that are initiated by

more reactive species.

22

Substituents on the ring play an important role in determining the antioxidant

activity of phenolic antioxidants. Electron-donating substituents on the phenyl

ring increase the radical-scavenging potency of phenolic compounds by

weakening the phenolic O–H bond.112 Substitutions at the ortho position

determine the stability of the phenoxy radical. A bulkier substituent produces a

more hindered phenoxy radical, which helps increase the radical-scavenging

activity. Substitution at the para position avoids the dimerization of the phenolic

radical compounds.

A variety of compounds incorporating the phenolic structures have been

reported. Some if these phenolic compounds have been claimed to be

responsible for the trapping and stabilising of reactive free radicals, including

single oxygen, hydroxyl, superoxide and peroxyl radicals and also lipid soluble

radicals. These properties of phenolic compounds (some examples of which

are shown in Figure 1.6) provide desirable effects in various biological

functions.

In particular, the hindered phenolic species have been reported to reduce low-

density lipoprotein (LDL) oxidation and morbidity in the lipoprotein and have

reactivity towards radicals.112 Many novel phenolic antioxidant compounds

capable of oxidative modification of LDL have been reported. LDL oxidation

has been found to be a major contributor to atherosclerosis, and logically its

inhibition by antioxidants should be beneficial in preventing and treating

atherosclerosis. The well-known phenolic compound -tocopherol (vitamin E)

23

exhibits antioxidant activity by trapping and stabilising reactive free radicals and

efficiently inhibiting lipid peroxidation.113

The antioxidant capacity of a compound is measured by its reactivity towards

the reactive radical species. Likewise vitamin E (29), Galvinoxyl (34) is also a

free radical inhibitor that is often used to investigate the antioxidant activity.

The rates of reaction of other phenolic compounds have been compared with

that of Galvinoxyl. BO-653 (31) and vitamin E (29) were found to have similar

reactivity, which is higher than that of another commonly used hindered

phenolic compound Probucol (32)114(Figure 1.6).

Figure 1.6: Phenolic and radical antioxidants.

The hydroxycinnamic acids (HCA) are also a class of phenolic antioxidants,

which are highly abundant in food and may account for one-third of the phenolic

compounds in our diet. Hydroxycinnamic acid compounds are of increasing

interest in health research because they are recognised as potent

antioxidants.115, 116 These compounds have been reported as radical chain

24

breaking antioxidants that achieve this action through radical scavenging

activity. This activity has been attributed to hydrogen or electron-donating

capacity and their ability to delocalise or stabilise the resulting radical within the

structure. The additional unsaturation incorporated in hindered

hydroxycinnamic acid compounds gives extra stabilisation to the phenoxy

radical species accordingly increasing their antioxidant activity. These active

phenolic compounds are employed in various biological applications.117, 118

HCA derivatives display their effects on cell membrane stabilisation through

their anti-hypoxic, hepatoprotective and anti-inflammatory mode of action. It

has been reported that there is a direct relationship between the antioxidant

activity and the membrane stabilisation activity of these compounds. HCA

derivatives (p-caumaric (35), ferulic (36), and caffeic (37) acids) also

demonstrate high antibacterial, antifungal activity and also antiviral activity119.

Some of the alkyl and cycloalkylamino derivatives of cinnamic acids also exhibit

hypolipidemic and antiatherosclerotic effects120 (Figure 1.7). Several adenosine

combining hindered phenolic antioxidant species are found to be biologically

active and useful in various therapeutic applications.121

Figure 1.7: Examples of derivatives of hydroxycinnamic acids.

25

1.3 Adenosine

Antioxidants are not only radical scavenging and H-transfer molecules, but they

can also generate an effect through switching on endogenous cellular

antioxidant mechanisms. Many clinical and experimental studies have

confirmed that stimulation of the adenosine receptor activates the antioxidant

mechanism of enzymes.122, 123 By stimulating neutrophils, adenosine inhibits

the superoxide anion generation. This adenosine-induced inhibition acts via

activation of cell surface receptors without cellular uptake of adenosine.124

Neuronal cells are highly sensitive to free radical induced oxidative stress.

Adenosine is used as a neuro-protective agent in the condition of free radical-

induced ischaemia-reperfusion injury and ischaemic stroke.125

1.3.1 Structural features of adenosine

Adenosine (39) is a naturally occurring nucleoside, formed during physiological

processes by degradation of the adenosine triphosphate (ATP) nucleoside (a

cell energy source). Its structure is composed of an adenine-based purine

nucleoside attached to a ribose sugar at the N9-position. The ribose sugar ring

is numbered independently in the chemical structure (Figure 1.8). Adenosine

derivatives are usually synthesised by modification at the exocyclic amine (N6),

the heteroaromatic ring carbon (C2) or the exocyclic (C5) ribose ring methylene

group. Adenosine is the only endogenous agonist for adenosine receptors and

regulates a number of physiological processes through the activation of

adenosine receptors.

26

Figure 1.8: Chemical structure of adenosine (39)

1.3.2 Adenosine receptors

A receptor is a protein macromolecular complex often found on the surfaces of

the cell membrane that receives or transmits chemical signals through the cell

wall. The chemical signals are generated through the binding of a particular

agent to these receptors which is extremely structurally selective. A ligand

binds to the receptor through various interactions including covalent, ionic,

dipole–dipole, and ion–dipole, hydrogen bonding and the charge-transfer

complex. The binding of a ligand generates a chemical signal, which is further

converted to a specific cellular and/or tissue response. Several ‘drug-receptor

interaction theories’ have been proposed to explain how a drug can generate or

stimulate a specific biological response.126

Several types of receptors are involved in various physiological processes. One

of these types of receptors is called a purinergic receptor. They are involved in

signalling actions mediated by purine nucleosides or nucleotides. The term

purinergic receptor is used to describe classes of plasma membrane receptors

that are also called purinoceptors. Purinergic receptors are involved in a

27

number of cellular functions including the migration of neuronal stem cells,

proliferation, vascular action, apoptosis and cytokine secretion.127, 128 The

purinergic receptors are categorised into three types: P1 (adenosine) receptors,

P2Y (all nucleotides) and P2X (ATP), where P1 and P2Y are G-protein-coupled

receptors (GPCR) and P2X are ligand-gated ion channels.129

GPCRs are receptors with a peptide sequence that pass through the cell

membrane seven times, and therefore, are known as seven-transmembrane

domain receptors or 7TM receptors. GPCRs are the most heavily targeted

receptors in the pharmaceutical industry, because of their significant role in

many diseases. Resulting in 40% of modern drugs being based on targeting

these receptors.130 GPCRs interact with different types of proteins and

participate in several important biological functions. They interact with hetero-

trimeric G-proteins (Figure 1.9) and with the accessory proteins called GPCR-

interacting proteins (GIP).131

The GPCR couples with the membrane-associated heterotrimeric G-protein

made up of G, G and G subunits.132 These subunits are in complex form

when in the resting state. The G subunit is the flexible signalling subunit. In

the inactive complex form, the G subunit binds to guanosine diphosphate

(GDP). After the activation of this receptors, a nucleotide-exchange reaction

occurs on the G subunit and the GDP is replaced by guanosine triphosphate

(GTP).133

28

These G-proteins exist in three different families of proteins including Gi, Gs and

Gq, which express selectivity for particular receptors. Gs and Gi proteins

stimulate and inhibit the adenylate cyclase enzyme, and Gq protein interacts

with the enzyme phospholipase-C respectively.134 The G subunits of these G-

proteins differ in structure (Figure 1.9).

Figure 1.9: Three-dimensional crystal structure of the G-protein coupled receptor (GPCR) embedded in the cell membrane attached to heterotrimeric G protein.

135

Adenosine receptors are the P1-type G-protein coupled receptor, which are

activated by the adenosine nucleotide released in cells. The adenosine

nucleotide is found in living cells and is involved in several metabolic functions

through both intracellular and extracellular mechanisms. In cells, adenosine

participates in all metabolic processes, and its concentration is modified by

pathophysiological conditions (i.e. during disease or disorder).136, 137

29

Adenosine is produced intracellularly by two metabolic pathways, which both

involve the hydrolysis of AMP to adenosine by 5-nucleotidase followed by the

catabolism of S-adenosylhomocysteine137 (Figure 1.10). The adenosine

concentration increases within the cell membrane during pathophysiological

conditions and is released from the cell. The released adenosine can then bind

to adenosine receptors and acts as a local metabolic regulator or, what has

been described as a “retaliatory metabolite”.138 Adenosine is also involved in

the regulation of several types of cells including endothelial cells, smooth

muscle cells and macrophages.139

Figure 1.10: Intracellular adenosine synthesis.140

Several studies have demonstrated the strong effect of adenosine purine

nucleoside on the regulation of various physiological and biochemical

30

functions.141 The local tissue concentration of adenosine increases when

exposed to oxidative stress. This ischaemic event protects the heart from the

further ischaemic injury, which has focused attention on understanding the

protective effects of adenosine.142-144 Adenosine exerts its effect through

several receptors called adenosine receptors. Adenosine receptors are

classified into four different receptor subtypes, namely A1, A2A, A2B, and A3

subtypes20 (Figure 1.11). These receptor subtypes are identified according to

their pharmacological properties and cloning studies. These adenosine

receptor subtypes are members of the GPCR family, which are classified

according to the receptor coupling to adenylate cyclase through Gi (A1, A3) and

Gs (A2A, A2B) protein. The receptor subtypes are involved in the inhibition (A1,

A3) and stimulation (A2A, A2B) of adenylate cyclase activity and subsequent

cyclic adenosine monophosphate (cAMP) production.145, 146

Figure 1.11: Adenosine receptor activation.147

31

Adenosine receptors were first investigated as potential drug targets in 1981.148,

149 Adenosine A1 receptor (A1AR) agonists are involved in cardiac arrhythmias

and neuropathic pain. Adenosine A2A receptor (A2AAR) agonists are useful for

myocardial perfusion imaging and as anti-inflammatory agents. Adenosine A2B

receptor (A2BAR) agonists are used in the treatment of cardiac ischaemia. The

Adenosine A3 receptor (A3AR) subtype is involved in inflammation, cancer and

cardioprotection. These cardioprotective effects are generated through binding

interactions of the four extracellular adenosine receptor subtypes. The

protective effect of adenosine in reperfusion injury has also been reported.150 A

study of acute myocardial infarction showed that administration of adenosine

reduced the infarct size.151, 152 In particular, adenosine plays a role in ischaemic

preconditioning to protect the heart from ischaemia153 and also protects against

acute myocardial infarction.

Adenosine A1 receptors (A1AR) 1.3.2.1

The A1 receptor is the first subtype of the G-protein coupled receptors that was

discovered. It is involved in a wide range of biological functions through

different signalling pathways, which are based on the coupling of the A1AR to

the different G-proteins.154, 155 In this mechanism, the A1AR agonist binds to the

Gi1/2/3 and G0 proteins and inhibits the enzyme adenylate cyclase by decreasing

the concentration of the secondary messenger cAMP.156, 157 The binding of the

A1AR activates K+ ion channels and inhibits Ca2+ ion channels that activate the

enzyme phospholipase C158-160 throughout the body.

32

The A1AR are distributed widely throughout the body and mediate different

biological functions. This receptor is present in the brain, spinal cord, heart,

liver, testis and white adipose tissues in higher concentrations.161 The A1AR is

found in lower concentrations in the lungs, kidney and small intestine162, 163 and

is present in the smooth muscles throughout the vascular system.164

Activation of the A1AR is involved in several myocardial pathological conditions

including ischaemia, reperfusion injury in arrhythmogenesis, coronary and

ventricular dysfunction, and chronic heart failure.165-168 In the heart, stimulation

by the A1AR acts as a myocardial depressant by decreasing the conduction of

electrical impulses and suppressing the pacemaker cell function, resulting in a

decrease of the heart rate. A1AR have also been detected in several types of

cancer, with suspicion that they may be involved in the stimulation of

chemotaxis of tumour cells.169

Adenosine A2A receptors (A2AAR) 1.3.2.2

The adenosine A2AR was first suggested in 1979 by Calker et al. 170 and in

1980 by Londos and co-workers.157 A2AR are classified further according to

their ligand binding affinity as A2AAR (high affinity for adenosine 0.1-1.0 µM) and

A2BAR (low affinity for adenosine ≥ 10 µM) by Bruns171 on the basis of initial

work by Daly and co-workers.172 It is a type of GPCR, whose activity is

mediated by G-proteins and stimulates adenylate cyclase and the synthesis of

cAMP.

33

A2AAR are present throughout the body and are found in low abundance in the

heart, lungs, blood vessels, and other areas of the brain.160. These receptors

are highly expressed in blood platelets, leukocytes, thymus, spleen and

olfactory bulb. These receptors have been involved in several

pathophysiological conditions. Current research has focused on developing

new A2AAR agonists in order to determine the structure of the A2AAR receptor.

This development has been achieved by cloning the A2AAR from several

species including rat,173 human brain,174 mouse175 and guinea pig brain.176 The

A2AAR agonists have also been found to increase blood flow during a cardiac

nuclear stress test.177

The A2AAR agonists play a key role in several diseases including

inflammation,178 ischaemia-reperfusion injury179 and cancer180 and have

cardioprotective effects.141 The A2AAR provides cardioprotection via

vasodilatation by increasing myocardial blood flow, and by supporting the

synthesis of new blood vessels as well as protecting tissues from collateral

inflammatory tissue damage.181 The A2AAR is beneficial in controlling

inflammatory leukocytes by reducing oxidative metabolites such as superoxide

and H2O2 in activated neutrophils.124, 182, 183

The activation of A2AAR may contribute in the pathophysiology of glaucoma due

to the increase in ocular perfusion pressure.184 Current evidence suggests that

the increased internal ocular pressure (IOP) leads to blindness via induction of

ischaemia and lowering IOP via blood reperfusion to cause ischemia-

34

reperfusion injury and retinal inflammation.185 Intravenous infusion of

adenosine in healthy volunteers led to the reduction of IOP and increasing

blood flow to the optic nerves. Experimental evidence has also shown that

application of adenosine and selective A2AAR agonists induced dilation of retinal

blood vessels via activation of A2AAR.186 Therefore, A2AAR agonists might be

an ideal agent in the treatment of glaucoma.

In addition, activation of the A2AAR on cells of the immune system plays a role

in controlling the activity of inflammatory cells including neutrophils,

macrophages and T lymphocytes. Therefore, A2AAR agonists may be

applicable as endogenous regulators of inflammation and cardioprotection.187,

188 However, A2AAR also negatively regulates over-reactive immune cells. The

A2AAR influence the activity of the indirect pathway of the basic ganglia in the

brain. In the brain, the A2AAR mediate the release of glutamate and dopamine,

making them a potential therapeutic target for neurological diseases.

Adenosine A2B receptors (A2BAR) 1.3.2.3

The A2BAR are low-affinity GPCRs that are involved in the activation of

adenylate cyclase.160 These receptors are small in number, and a high

concentration of adenosine is needed to generate a response. The adenosine

A2BAR has been implicated in the proliferation and differentiation of mast cell-

mediated angiogenesis.189-191 Additionally, adenosine stimulates the production

of interleukin 4 (IL-4) and IL-13 in mast cells via A2BAR activation.192

35

A2BAR have been suggested to be present in a relatively high density in, the

gastrointestinal tract, caecum, large intestine and urinary bladder. They are

thought to be present at a lower concentration in the lung, blood vessels, eye

and mast cells.193 Adipose tissue and the adrenal gland, brain, kidney, liver,

ovary and pituitary gland are thought to have a very low concentration of the

A2BAR.160

A2BAR are involved in several pathophysiological conditions including coronary

artery disorders,194 atherosclerosis,195 pulmonary hypertension associated with

lung disease,196 liver ischaemia–reperfusion injury,197 colitis198 and type II

diabetes.193

Adenosine A3 receptors (A3AR) 1.3.2.4

A3AR are GPCRs that bind specifically to Gi/ Gq proteins and are involved in

many pathophysiological functions. Activation of the A3AR inhibits adenylate

cyclase activity, which decreases the cAMP level and stimulates phospholipase

C and D, resulting in the elevation of intracellular inositol 1,4,5-triphosphate and

Ca2+ levels.199 The A3AR is involved in neutrophil degranulation in neutrophil-

induced tissue injury and the regulation of other immune cells of the innate and

adaptive immune systems. The A3AR also participates in intracellular signalling

and stimulates mitogen-activated protein kinases including extracellular signal-

regulated kinase1/2 and p38 by upstream activation of protein PI3K.200

A3AR are distributed widely in the body. The A3AR can be detected using radio-

ligand binding and functional assays in organs from various species. The

36

highest levels of these receptors are present in the testis, liver and brain. They

are present at low levels in the central nervous system, placenta, heart, bladder,

uterus, spleen, and eye of sheep, rats and humans.201-203 Some of the

functional A3AR are detected on the cell surface.204 Expression of the A3AR

can be detected at the mRNA level in both rodent and human organs. The

A3AR is overexpressed in cancer and inflammatory cells, rheumatoid arthritis,

psoriasis and Crohn's disease and exhibits low expression in normal cells.205-207

Adenosine expresses itself in all the receptor-binding and biological functions

via agonist and antagonist mechanisms.

1.3.3 The concept of agonism and antagonism

Endogeneous natural ligands, including hormones and neurotransmitters bind

to their receptors, which induces a further biological response. The ligand of a

adenosine receptors induces a conformational change in the receptor (a

transmembrane protein) which initiates the intracellular response. The agonism

and antagonism concept explains the different types of binding of a drug

molecule to its receptor.

Adenosine receptor selective ligands are synthesised by first studying their

binding interaction with GPCRs and are usually categorised as (partial)

agonists, neutral antagonists, (partial) inverse agonists, and positive and

negative allosteric modulators.

37

Over the past two decades, it has become clear that the available GPCR

ligands preferentially stimulate one signal-transduction pathway over another

signalling pathway. This phenomenon is referred to as functional selectivity or

biased signalling.208-210 Biased signalling implies that an agonist for one

pathway may act as an antagonist or inverse agonist for another signal-

transduction cascade. Exploiting the functional selectivity of compounds is an

important research field that will help develop drugs with fewer side

effects.209,211 Selective activation of the beneficial signalling pathways without

simultaneous activation of side effect-inducing pathways can be used to

improve the therapeutic window of a drug.

An agonist is a compound that binds to a receptor, reorients receptor and

protein conformation, and causes similar biological actions as the endogenous

species (such as hormones and neurotransmitters) which triggers the natural

response. An antagonist is a chemical or biological entity that inhibits the

effects of a natural agonist ligands (hormone, neurotransmitter) and is also

called a blocker of the receptor binding site or competitive inhibitors.

Adenosine agonists 1.3.3.1

An agonist can cause complete or partial effects when it binds to its receptor

under different physiological conditions. Agonists can be exogenous ligands

(such as drugs) or endogenous ligands for the receptor. Agonists are

categorised according to the binding ability of the ligand as full agonists, partial

agonists and inverse agonists (Figure 1.12). Full agonists activate all sites of

38

adenosine receptors, and side effects are associated with receptor activation.

Partial agonists bind to their receptor and show a partial response, and that can

provide the solution to avoiding side effects by activating only the specific site of

the receptor.212 The inverse agonists show a negative intrinsic effect or partially

or fully block the receptor, resulting in low or no biological response and are

also called antagonists.

Figure 1.12: Mode of action of a agonists.213

Adenosine antagonists 1.3.3.2

Antagonists that bind at the same site as the natural ligands are called

competitive antagonists, and agents that bind to mixed-receptor sites are known

as nonselective or non-competitive antagonists (Figure 1.13). Caffeine,

39

theophylline and other methylxanthines are non-selective antagonists for

adenosine receptors. Several antagonists are used in therapeutic

applications.214 It has been proposed recently that antagonists of distinct

adenosine receptor subtypes may be beneficial in the treatment of asthma215, 216

or certain neurological diseases, such as Parkinson’s disease.217

Figure 1.13: Mode of action of a competitive and noncompetitive antagonists.213

Adenosine mediates various physiological and pathophysiological functions via

agonist and antagonist mechanisms. All four adenosine receptors can be

influenced differently via these mechanisms. Structural modifications to the

parent adenosine molecule can significantly change activity towards these

receptors, which are essential in maintaining various pathophysiological

conditions. Specifically, cardiovascular diseases are a major health concern in

40

the world and therefore research has been focused towards the adenosine A2

receptor subtypes, which are believed to be mainly involved in the

pathophysiology of cardiovascular diseases.

1.3.4 Active adenosine A2AR agonists and antagonists in clinical trials

Adenosine A2AAR agonists 1.3.4.1

In the past two decades, a number of biologically important agonists and

antagonists that that can selectively exhibit A2AAR affinity, have been

synthesised. The study of these compounds has progressed further with the

discovery of the importance of the A2AAR agonist–ligand interactions. The C5-

modification of adenosine produces the potent and nonselective adenosine

receptor agonist 5-N-ethylcarboxamide (NECA) (Figure 1.14).

Figure 1.14: Adenosine A2AAR agonists.

Substitution at the C2-position of adenosine has been determined to be

essential for the A2AAR activity and selectivity. Most of the A2AAR agonists are

synthesised by C2-modification of NECA (40). In this regard, the C2-modified

ammonium salt of 4-[2-[(6-amino-9-b-D-ribofuranosyl-9H-purin-2-yl) thio] ethyl]

benzenesulfonic acid (PSB 0777, 41) is a potent full A2AAR agonist that has

41

been reported to stimulate acetylcholine-induced contractions in rat intestine

segments.218 This compound is expected to be potentially useful for the

treatment of inflammatory bowel disease (Figure 1.14).

C2-modified alkynyl linked adenosine compounds are also known to possess

A2AAR agonist activity and selectivity. In this regard, Apadenoson (48) is a

highly selective A2AAR agonist’s which has potential utility as a pharmacological

stress agent in myocardial perfusion imaging (MPI).219 The selective A2AAR

agonist YT 146 (46) acts as a vasodilator that also inhibits the synthesis of

inflammatory mediators to reduce ischemia/reperfusion injury.220 Furthermore,

this alkynyl analogue HENECA (YT146, 46), is being studied for

cardioprotective effects.221

A synthetic analogue A2AAR-selective agonist UK-432097 (42) was designed for

the treatment of chronic obstructive pulmonary disease, but it failed phase II

clinical trials and still being used as an internal probe to examine the active site

structure. The protein-binding capacity of C2-terminal amino acid conjugates,

the A2AAR agonist CGS 21680222 has been studied and it has shown 140 times

greater selectivity for A2AAR compared with the A1AR and it has effects on

neuronal transmission.223-225

The highly selective A2AAR agonists Apadenoson (48) (StedivazeTM) and

Binodenoson (CorVueTM) are in phase III trials for myocardial perfusion

42

imaging.226 Regadenoson (Lexican, 43) is used as a coronary vasodilator and

Sonedenoson is being evaluated as a new therapy for wound healing and

human diabetic foot ulcers.227 ATL313 (47), a A2AAR selective agonist

profoundly protect the mouse liver from reperfusion injury (Figure 1.15).

Figure 1.15: Pharmacologically active A2AAR agonists.

Adenosine A2AAR antagonists 1.3.4.2

A2AAR antagonists also play important roles in elucidating structure activity

relationships (SARs) and are synthesised primarily by modification of the

xanthine ring with caffeine and theophylline (from tea) as well-known examples.

43

The xanthine synthetic analogue DMPX (49)228 and the caffeine analogue (E)-8-

(3-chlorostyryl)caffine (CSC)229 were reported to be selective antagonists of the

A2AAR. In addition, the A2AAR antagonist Istradefylline (KW 6002, 51) has been

used in the treatment of Parkinson’s disease to reduce the movement disorder

(dyskinesia) caused by conventional anti-Parkinson’s drug therapy (Figure

1.16).230, 231

Further insights into the allowable structural diversity of receptor antagonists for

these receptor subtypes were discovered through the X-Ray crystal analysis of

a non-xanthine-based analogue ZM 241385 (51) bound to the A2AAR. This

compound has a 90-fold selectivity for the A2AAR over the A2BAR.232

Preladenant (SCH420814, 52) and several other similar analogues were

developed with longer hydrophobic chains at the C2-position. These

compounds were found to be highly potent and selective adenosine A2AAR

antagonists and were intended for use in the treatment of Parkinson’s

disease233 (Figure 1.16).

Figure 1.16: Pharmacologically active A2AAR antagonists.

44

The hypotensive and bradycardic activity of adenosine was first investigated in

1929 by Drury and Sezent Gyorgyi.234 Adenosine is currently used in the

treatment of several cardiovascular diseases under the brand name Adenocard

and Adenoscan. The recent determination of the crystal structure of some

adenosine receptors confirms that the 7TM topology is common between

various families of GPCR receptors. Adenosine receptor agonists have more

beneficial effects than do antagonists, and the crystal structure provides

information about the binding interaction of agonists to the protein receptors, as

shown in Figure 1.17.

Figure 1.17: Comparison of receptor-ligand interaction binding for the A2AR with the agonists adenosine

(39) and NECA (62) along with antagonists ZM 241385 (51).235

Structure of human A2AR in cartoon

representation are bound to the ligands (a) ZM 241385; (b) NECA; and (c) adenosine; (d,e) polar and non-polar interactions involved in agonist binding to human A2AR are shown for NECA and adenosine. Figure b and c the hydrogen bond interactions (H3 and H7) activates the A2A receptor. However, these bonding is abscent in antagonists Figure a. Figure d and e van der waals interactions with amino acid residues.

45

There is a great deal of interest in studying GPCR–drug interactions, particularly

those concerning the A2AARs. A2AAR agonists that act in the periphery to cause

vasodilation and to decrease blood pressure, can also induce cardiovascular

side effects such as hypotension.188 These effects can trigger a baroreflex

(body’s homeostatic mechanism to maintain blood pressure) in the heart,

leading to increases in heart rate and cardiac output, and therefore the demand

for oxygen. These conditions would be seriously detrimental under prolonged

ischaemic conditions and would work against any cardioprotective effects of

A2AAR activation in cardiomyocytes. However, it has been demonstrated that

adenosine itself has cardioprotective effects during myocardial ischemia236 and

reperfusion,237 and is expected that an A2AAR subtype-specific agent would

cause fewer side effects.

Adenosine A2BAR agonists 1.3.4.3

Several ligands for the A2BAR have been identified in recent years193, 238 and

have been studied as pharmacological targets and to evaluate their therapeutic

potential. However, the important advances in identifying A2BAR agonists

through improved in vitro pharmacological profiling have been recently

published.239 Previous SAR studies of NECA and adenosine derivatives have

indicated that N6 is a useful position for A2B receptor binding-site recognition.

Some examples of N6-substituted adenosine derivatives endowed with

satisfactory levels of A2BAR potency have been reported. In particular, the

introduction of (substituted) phenyl rings at the N6-position of adenosine led to

the A2BAR selective compounds.238

46

Based on some patent claims, a series of substituted 2-amino-4-phenyl-6-

phenylsulfanylpyridine-3,5-dicarbonitriles were synthesised as agonists for

adenosine receptors.240 A series of five 2-amino-6-(1H-imidazol-2-

ylmethylsulfanyl)-4-(substituted) phenyl pyridine-3,5-dicarbonitrile derivatives

has been also reported to display high-potency agonistic activity for the A2BAR

over the A3AR subtype.241 The A2BAR agonist 2-phenylaminoadenosine

(CV1808, 53) has been shown to be effective as a coronary vasodilator and as

an antihypertensive and antipsychotic agent.242, 243 The potent and highly

selective non-adenosine compound A2BAR agonist 2-((6-amino-3,5-dicyano-4-

(4-(cyclopropylmethoxy)phenyl)-2-pyridinyl)thio)-acetamide (BAY60-6583, 54)

has been shown to have similar potency to NECA 244, 245 (Figure 1.18).

Figure 1.18: Adenosine A2BAR agonists

Adenosine A2BAR antagonists 1.3.4.4

Several A2BAR antagonists with high affinity and good selectivity have been

identified among structures based upon a xanthine core suitably substituted at

the 1-, 3- and 8- positions.246 Kim et al.247 reported that a substituted

phenylcarbamoyl-methoxy-phenyl chain at the 8 position of a series of 1,3-

dipropylxanthines could specifically direct the antagonist activity to the A2BAR.

47

C8-modifications of adenosine analogues without a ribose ring are found to give

the selective A2BAR antagonists that are also involved in cardiac actions. In this

regard, 3-ethyl-3,9-dihydro-1-propyl-8-[1-[[3-(trifluoromethyl)phenyl]methyl]-1H-

pyrazol-4-yl]-1H-purine-2,6-dione (GS6201, 55) attenuates an inflammatory

response during acute myocardial infarction in mice and reduces caspase-1

activity in the heart.195 The highly potent human A2BAR inverse antagonist N-(4-

acetylphenyl)-2-[4-(2,3,6,7-tetrahydro-2,6-dioxo-1,3-dipropyl-1H-purin-8-

yl)phenoxy]acetamide (MRS 1706, 56) with extended chain was reported to be

a selective A2BAR antagonist with Ki value of 1.93 nM. (Figure 1.19).

Figure 1.19: Adenosine A2BAR antagonists

Despite the number of selective A2AAR agonists and antagonists that have been

reported, only a few adenosine derivatives have succeeded in clinical trials for

cardiovascular diseases. This failure is due to widespread signalling effects of

adenosine and unwanted side effects. It was believed that dual acting

adenosine molecule would be beneficial to overcome these complications.

48

1.4 Dual action adenosine agonists-Rationale for the research

The focus of current research is shifting towards the development of lead

compounds that can act on more than one target. This modern approach to

drug design combines two or more active parts of different molecules within one

molecule to generate a multitargeted drug.

The novel dual acting antioxidant 5-(3-methyl-pyrazole-5-ol-1-yl)-1,1,3,3-

tetramethylisoindoline-2-yloxyl (57) with an isoindoline nitroxide within the

Edaravone structure is act as a potential active drug for cardiovascular101 and

cerebrovascular ischemia in the brain-protection.248 The dual acting compound

UA-8 ( 13-(3-propylureido)tridec-8-enoic acid, 58) provides the cardioprotective

effect against the ischaemia-reperfusion injury249 (Figure 1.20).

Figure 1.20: Dual action compounds in Ischaemia

The majority of research on multi-targeted adenosine ligands has been carried

out by Ken Jacobson and co-workers. In 1985, Jacobson et. al. first published

their investigation into adenosine “binary conjugates” molecules. These

compounds are coupled to organic molecules such as peptides and amines and

act as ligands at adenosine receptors (Figure 1.21). This work initiated

research that resulted in the coupling of both adenosine agonists and

antagonists to biotin (a compound that forms a complex with glycoprotein

avidin), substance P-receptor binding entities as well as lipids. At the time,

49

some of the work was patented and many additional publications detailed the

synthesis of bifunctional adenosine analogues.250

Figure 1.21: Bifunctional adenosine receptor-binding analogues and binary conjugates.

The broader interest in the field of binary conjugates of adenosine A1AR and

A3AR agonists commenced with a publication in 2000, 251 and a continuance of

the original patent was granted in 2003.252 Further work in the field of

bifunctional adenosine drugs rose again in 2003 when Liang and Jacobson filed

a patent for the use of adenosine receptor agonists and antagonists together as

a novel approach for treating ischaemic injury to the heart.252 In the patent, the

activity of the binary conjugate MRS1528 (62) was explained, i.e. it acts as an

agonist for the A3AR and as an antagonist at the A2AAR.

50

In 1986, Burns et al characterised the A2 adenosine receptors and termed them

A2AR. During the 1989s, Hutchison and co-workers found that C2-substituted

adenosine compounds were selective A2AR agonist ligands and possessed

hypotensive activity.253 In 1992, Cristalli and colleagues published further

research that demonstrated that C2-substituted alkynyl linked with 5-

ethylcarboxamide adenosine compounds increased A2AAR selectivity.254 In

1997 255 and 2001,183 Martin and Sullivan et al further developed C2-substituted

adenosine compounds by preparing compounds containing a 2-cyclohexyl

linked with hydrazine chain (63) and an alkynyl chain (64). These compounds

enhanced A2AAR selectivity and were effective in coronary vasodilation and

inhibiting human neutrophil oxidation. Pfizer patented several C2-, N6- and C5-

modified adenosine compounds (65 and 66) as anti-inflammatory agents with

inhibition of neutrophil function at a concentration of < 40 nM 256 (Figure 1.22).

51

Figure 1.22: Pharmacologically active A2AAR and A3AR selective analogues.

Interest in A2AAR has been rekindled by a finding that suggests both agonisms

of this receptor and its inhibition by antagonists may have therapeutic

applications. In 2001, Ohta and Sitkovsky reported that A2AARs are

imperatively involved in the limitation and termination of the prolonged

inflammation.257

In 2007, Gregg and co-workers published new research on dual acting A1AR

agonists that contained antioxidant species and had the potential to produce

cardioprotective effects.258 Furthermore, in 2009 and 2012, Foitzik and Hausler

52

reported the cardioprotective effects of A2AAR selective adenosine

compounds259 attached by an aminoethylaniline linker possessing antioxidant

functionality, which were shown to reduce ROS activity. The C2- and C5-

modified adenosine compounds VCP874 (69) and VCP728 (70) attached by an

aminoethylaniline linker were shown to have an excellent A2AAR potency and

selectivity. They demonstrated good EC50 values of 45 nM and 6.6 nM,

respectively121 (Figure 1.23).

Figure 1.23: Pharmacologically dual-action antioxidant coupled A2AAR and A3AR-selective analogues.

Adenosine compounds that act via A2AAR on leucocytes significantly inhibit the

inflammatory process. Adenosine compounds with the para-(2-

aminoethyl)aniline linker at the C2-position are known to have A2AAR selectivity.

Attachment of the antioxidant structure by a flexible linker is predicted to

contribute to the A2AAR pharmacophore, improving A2AAR selectivity and thus

53

localising antioxidant function to the desired site of action. Once positioned, this

can move in the vicinity of its intended site of action, scavenging cytotoxic ROS

before they can cause further injury (the antioxidant species will be active from

the moment of administration and the ROS scavenging properties are therefore

not strictly limited to A2AAR orthosteric site, but apply more generally) (Figure

1.24).

The value of this approach has been given support by binding studies

undertaken using molecular modelling that have analysed the crystal structure

of bifunctional compound 70. Here the attached antioxidant moiety would

extend beyond the orthosteric binding site of the adenosine derivative. The 2.7

Ǻ-resolution crystal structure of the human A2AAR agonist in complex with a

selective A2AAR agonist UK-432097 has shed light on the ligand-induced GPCR

binding and activation260 (Figure 1.24).121

A B

Figure 1.24: Adenosine receptor crystal structure in complex with A)UK-432, 097 (42), B) compound VCP728 (70).

54

Currently, there is a need for potent A2AAR agonists with combined antioxidant

species to test this theory. In addition, cardiovascular and other inflammatory

diseases have complex pathogenesis, and currently available adenosine and

antioxidant compounds are still not able to do enough on their own, treating only

a limited number of these complications. Treatment involving adenosine

analogues have selectivity problems and hence forming unwanted side effects

and low efficacy. Antioxidant therapy is more effective in a combination of other

related treatments.

These days, it is common practice to administer a cocktail of drugs with

complementary effects to patients. However, this combined drug therapy

introduces complications associated with pharmacokinetics, distribution, toxicity,

and patient's compliance and elevate the risk of side effects. To circumvent

some of these issues, the focus of current research is shifting towards the

generation of multi-targeted lead compounds. This new generation of designed

drug compounds combines two or more active parts in a single molecule that

each initiates independent but complementary action when administered. A

single administration of multi-targeted compounds can have benefit in the

predictability of their pharmacokinetics (PK) and pharmacodynamics (PD)

properties and also in improved patient compliance. It is anticipated that, this

bifunctional approach could have beneficial effects from the combined entities

which would not only be additive but synergistic in nature, resulting in increased

efficacy.

55

It has been demonstrated that both the stimulation of adenosine receptors and

presence of antioxidants such as nitroxide radicals have independently shown

beneficial effects in reducing the duration of ischemia. These effects also limit

ischemia-reperfusion injury in models of acute myocardial infraction. It was

proposed that compounds that possess both adenosine-receptor binding and

antioxidant capabilities would demonstrate potent cardioprotective actions and

be more effective than the two independent moieties.

The goal of this project therefore was to explore multi-targeted compounds that

can act as selective A2AAR agonists. These compounds (some initial target

structures are shown in Figure 1.25 and 1.26) would also possess a moiety that

is able to react with ROS (and other free radical species). Thus, these

compounds could potentially act as effective preventive treatments for

inflammatory diseases and also provide cardiovascular protection.

Figure 1.25: Antioxidant coupled p-aminoethylaniline linked adenosine target compounds (Series 1).

56

Figure 1.26: Antioxidant coupled with ethynyl (Series 2) and ethyl (Series 3) linked adenosine target compounds.

The aim of this project therefore is to synthesise a range of bifunctional

compounds which would ideally provoke cardioprotective responses when

administered to people experiencing ischaemia, and at the same time limit

reactive radical-based reperfusion injury.

57

Results and Discussion Part A: Adenosine 2analogues with para-aminoethylaniline linker

possessing antioxidant moieties

58

2.1 Preliminary Considerations

There have been many efforts to attempt to elucidate the various SARs for the

adenosine receptor subtypes. These efforts are continuing, and the

understanding of key structural features has improved, which is essential to

enhance subtype affinity and selectivity.256 Most of the current adenosine

receptor ligands are based on the structure of endogenous adenosine, and

therefore tend to be polar in nature.261 The nitrogen atoms at position 3 and 7

on the adenosine structure are particularly essential for high affinity for the

adenosine receptors (Figure 2.1).256, 262

The ribose moiety attached to the N9-position of the adenosine molecule is the

β-D-ribofuranose stereoisomer. The experimental evidence clearly indicates

that the ribose group binds to the receptor pocket via its hydroxyl groups and,

therefore, is essential for binding affinity and adenosine receptor activation.263

Further evidence shows that the adenosine molecule is arranged in an anti-

conformation within the receptor binding pocket, and thus the essential

pharmacophore must possess this particular ribose stereoisomer.264 This

ribose stereoisomer with at least one free 2- or 3-hydroxyl group is necessary

for the agonist activity at adenosine receptors.

Although all available ribose hydroxyl groups are hydrogen-bonded to the

receptor pocket, modifications at the C5- and even C3-position are known to

be tolerated.256 Even the furanose oxygen can be replaced by a carbon with

retention of activity.261 In addition, C2-256 and N6-positions on the purine ring

59

are most tolerant towards modifications with other groups. Indeed, structural

modification at C2 and N6 is thought to be responsible for the increased

receptor subtype selectivity and affinity.265 Most of the C6-modifications on the

purine ring undertaken to date lead to more potent and selective A1AR agonists.

However, co-substitution at the C2- and N6-positions does not necessarily lead

to additional affinity for the A2AAR over the A1AR.265, 266

The C6–amino proton appears to be necessary for high receptor affinity.263

This property may indicate either some unfavourable steric interaction for

N,N-disubstituted adenosine analogues or a hydrogen bond forming the role of

the C6-amino proton with the N6 binding domain of the orthosteric binding site.

However, certain bulky N6-substituents are known to be A2AAR tolerated such

as the hydrophobic 6-diphenylethylamino group. Almost all other N6

modifications considered favour A1AR affinity. Non-bulky modification at the

endogenous C6–amino223, 262, 267-269 group is also known to be tolerated by

A2AAR.270 Consequently, the additions of non-bulky side chains at the

C6-amino group were employed for all of the synthetic targets used in this

project.

The 5-modification most essential for an increase in agonist receptor affinity

such as the N-ethylcarboxamide moiety (5-NECA). The NECA itself is a

prototypic A2A adenosine receptor agonist.223, 266, 268, 271 This modification is

also known to improve pharmacokinetic stability to adenosine derivatives

because of the lack of interaction with adenosine deaminase.270 An ethyl amide

60

at the C5- position is better tolerated by the A2AAR, however, larger alkyl

groups bound to the amide decrease the receptor affinity. These characteristics

suggest either a critical role for the amide proton in forming a hydrogen bond

with the C5- binding domain of the orthosteric site or a severe steric limitation

encountered within the C5-binding domain.270 Therefore, the 5-NECA moiety

was selected as the only modification at the ribose 5-position.121 The critical

points of chemical reactivity for the adenosine derivatives are labelled in Figure

2.1.

Figure 2.1: Adenosine A2AAR selective compound CGS21680 (74).

Compounds possessing C2-substituted groups are typically selective for the

A2AAR, and exhibit high affinity and specificity for the A2AAR. Much attention

has focused on the development of aryl-alkyl amino substituents with the aim of

increasing the potency and selectivity of A2AAR agonists272 (in fact, these are

reported mainly as having decreased A1AR affinity and activity).223 The C2-

binding domain of the orthosteric site, therefore, sits at least largely (if not

totally) distal to the N6 and purine riboside binding domains, and has a critical

polar interaction adjacent to C2.266 It is otherwise hydrophobic in nature,

preferring aromatic rings attached via two methylene groups to the polar C2-

61

linking atom (Figure 2.1). The size of the hydrophobic C2-domain appears to

be better suited to an attached aromatic ring because the affinity of the

saturated cyclohexyl-bearing compound is significantly less than that of C2-

arylalkyl amino compounds.265

It is proposed that a linker involving a ‘bulky’ substituent linked with a two

carbon chain at the C2-position of adenosine could endow the generated

analogue with some subtype selectivity at adenosine receptors. C2-

substituents are well tolerated, and a range of different substituents display

variations in subtype selectivity. In particular, A2AAR selectivity appears to be

maximised by an electronegative bonding atom,261-263, 265, 267, 270, 272 a two or

three-atom spacer and a bulky, nonpolar group such as a cyclohexyl or phenyl

ring,259, 263 which can be itself further substituted.

It has been further proposed that the introduction of radical species attached to

the bulky linker substitution at C2-position of adenosine may improve the

receptor subtype selectivity and also be able to scavenge free radicals present

at the site. To test this theory, it was speculated that the antioxidant moieties

be tethered to the receptor binding adenosine using a ‘linker side chain’ that

would extend outside the binding pocket of the receptor. In such compounds,

the antioxidant functionality would still be joined to the receptor binding entity,

but, the ability of such dual action compounds to interact with the receptor

would not be compromised.

62

Another advantage of the ‘linker’ approach with an aniline-based unit such as

the aryl alkylamino chain linker would be that the antioxidant portion of the

molecule would be located outside the hydrophobic part of receptor pocket.

The antioxidant fuctionality would consequently interact more easily with

reactive free radical species.121

In the literature of adenosine analogues, a number of different linker groups

have been used. This linker groups were used to bridge from the receptor-

binding adenosine functionality to another moiety, including a fluorescent group,

a radiolabelled group, or another receptor binding entity. The compound

CGS21680 (46) has been shown to possess high affinity and specificity at the

A2AAR. By incorporating the commercially available para-(2-aminoethyl)aniline

(124), which has an aniline ring attached through an ethylamino group at the

C2-position in the target compounds. This type of compound preserves the 6-

carbon distance between two amino groups and also maintains high A2AAR

selectivity (Figure 2.1 and 2.2).

63

Figure 2.2: Candidate moieties and their actions in the initial structural class of target adenosine analogues.

In this study, all three series of compounds were synthesised with C2-

substitution on the adenosine NECA molecule. Further design and different

extensions to this structural C2-modification on adenosine was achieved with

appropriate linker groups and the desired antioxidant function. This approach

confers a bifunctional mode of action upon the target molecules without

compromising binding efficiency.

Nucleophilic C2-substitutions on adenosine can be accessed via an SNAr

(nucleophilic aromatic substitution) process. The literature provides an example

of the most nucleophilic atoms (i.e. S265, 273, 274, O261, 266 and N,121, 223, 259, 274),

which are primary choices of nucleophilic atoms that can be used to attack at

the C2-position of adenosine. This position is activated for SNAr by the

installation of a lower (highly electronegative) halogen atom (F, Cl, I). These

synthetic variables are discussed below in further detail.

64

2.2 Synthesis of C2-substituted TMIO adenosine analogues possessing substituents attached by the ‘aminoethylaniline linker’ moiety

The initial adenosine-based synthetic targets of this work can be synthesised

using two different approaches. In the first approach, the appropriate carboxy

nitroxide bearing an amine functionalised linker is generated and then coupled

to the adenosine intermediate 146 (Approach 1)(Scheme 2.1).

Scheme 2.1: Overview of first approach retro-synthesis of 161. Reagents and conditions: EtOH, DIPEA

75-80 °C;

In the second approach, the amine functionalised linker is first attached to the

adenosine intermediate to give compound 159 which is then coupled with the

appropriate carboxy nitroxide (Approach 2) )(Scheme 2.2).

65

Scheme 2.2: Overview of second approach retro-synthesis of 161. Reagents and conditions: EDCI/HOBt,

DMF, DIPEA 25 °C.

The published methodology using approach 1 for achieving these targets first

involves the synthesise of an appropriately functionalized carboxy nitroxide and

its subsequent reaction with the 4-(aminoethyl)aniline linker chain (127). The

resulting compound can then coupled with C2- and C5- modified adenosine

intermediates (146). To generate the desired target 127, the first step was to

synthesise the appropriate carboxy isoindoline nitroxide 106.

2.2.1 Synthesis of antioxidant carboxy nitroxide intermediates

possessing substituent attached to ‘aminoethylaniline linker’

moiety (127).

Synthesis of antioxidant carboxy TMIO intermediate (106) 2.2.1.1

The stable nitroxide 5–carboxy-1,1,3,3-tetramethylisoindoline-2-yloxyl (CTMIO)

(106) is typically synthesised from commercially available phthalic anhydride

(98) using the five-step procedure published first by Bottle et al.275 However, a

66

useful intermediate, brominated isoindoline precursor 5-bromo-isoindoline

(102), is synthesised according to the method developed by Micallef et al. in

1999276 from an adaptation of published methods (Scheme 2.3).

Scheme 2.3: Synthesis of 5-carboxy-1,1,3,3-tetramethylisoindoline-2-yloxyl (C-TMIO) (106).Reagents and

conditions:(i) Glacial CH3COOH, reflux 75 min., 83%; (ii) Mg, CH3I, toluene, reflux 3.0 h; 31% (iii) Br2, DCM, AlCl3, 0

oC 1.0 h, 65% (iv) dry CO2, THF, n-BuLi, 30 min, 83% (v) Na2WO4.2H2O, H2O2, NaHCO3,

Stir 72 h. 77%.

Commercially available phthalic anhydride (98) and benzylamine (99) were

reacted in a nucleophilic acyl substitution reaction to generate N-benzyl

phthalimide (100) in an 83% yield. Compound 100 was then tetramethylated in

a Grignard reaction using methylmagnesium iodide to form 2-benzyl-1,1,3,3-

tetramethylisoindoline (101) in 31% yield. Compound 101 was then brominated

and subsequently debenzylated in a single-step procedure to obtain 5-bromo-

1,1,3,3-tetramethylisoindoline (102) in a 65% yield. Compound 102 was

lithiated and quenched with carbon dioxide to give intermediate compound 5-

carboxy-1,1,3,3-tetramethylisoindoline. In situ sodium tungstate and hydrogen

peroxide oxidation of the crude intermediate compound afforded the desired

67

nitroxide 5-carboxy-1,1,3,3-tetramethylisoindoline-2-yloxyl (106) in a yield of

77% over two steps.(Scheme 2.3).

The antioxidant carboxy TMIO 106 was then in hand to react with linker 4-(2-

aminoethyl)aniline moiety (124) to generate the appropriate antioxidant bearing

side chain to synthesise the C2-modified adenosine target compound (161).

Synthesis of antioxidant carboxy TMIO coupled with para-2.2.1.2

(aminoethyl)aniline chain linker (127).

The antioxidant-bearing side chain 127 was initially synthesised separately in a

three-step reaction sequence involving coupling of the core compound 124 with

the antioxidant carboxy isoindoline nitroxide 106 to form a stable amide (125).

In the first-step, the more reactive primary aliphatic amine of the commercially

available compound 124 was protected with di-tert-butyl dicarbonate to give the

mono-Boc-protected compound N–tert-butoxycarbonyl-2-(4-aminophenyl)

ethylamine (125) (Scheme 2.4).

Scheme 2.4: Overview of N-Boc protection of linker amine. Reagents and conditions: (i) (Boc)2O, DCM, 100 min., 89%.

This was accomplished using a standard amine N-boc-protection method using

di-tert-butyl dicarbonate.277, 278 Dichloromethane (DCM) was substituted for

chloroform as the solvent for convenience, and the reaction was maintained at 0

°C to minimise undesirable reactivity at the 4–aminophenyl group. After 100

68

min, the starting material was consumed and < 5% of the undesired di-boc-

protected species was recovered. The reaction was conducted on a 1–2 g

scale.

The primary aliphatic amine is a stronger nucleophile than the aniline and

attacks the anhydride carbonyl to form the sp3 hybridised intermediate.

Reformation of the carbonyl eliminates a tert-butyloxycarboxylate anion, which

quickly releases CO2 and the tert-butoxide anion. This anion abstracts the extra

proton from the Boc-protected amine to form the product 125 and tert-butyl

alcohol (Scheme 2.5). The addition of the boc group to the primary amine

lowers the overall polarity of the compound, giving an Rf value of 0.67 in (9:1)

DCM/MeOH.

Scheme 2.5: Mechanism of N-boc protection of linker amine

69

The structure of 125 was supported by 1H NMR and 13C NMR spectroscopy and

mass spectrometry (MS) with the obtained spectra in agreement with the

published values. The Boc-protected amine was characterised using electron

ionisation MS (EIMS), which showed the expected molecular ion of M+Na at

259.1433 m/z (EI+ (HRMS) showed a deviation of 0.0011 from a calc. mass of

C13H20NaN2O2) and by a melting point of 56–58°C which supported the

successful synthesis of 125.121 In the 1H NMR spectrum, a new peak appeared

as a singlet at 1.45 ppm for the nine protons of the tert-butyl group. In the 13C

NMR spectrum, three additional peaks appeared. A strong peak corresponding

to the equivalent Boc carbons appeared at 28.4 ppm, the quaternary Boc

carbon appeared at 79.1 ppm, and the new Boc-amide carbonyl appeared at

155.9 ppm.

The Boc-protected amine linker (125) was coupled with the carboxylic acid

functionality of isoindoline nitroxide antioxidant 106 to form an amide. The

standard peptide coupling agent EDCI and reaction conditions were employed

in the formation of the amide.

The novel compound N-tert-butoxycarbonyl-2(4-N-(5-carboxy-1,1,3,3-

tetramethylisoindoline)aminophenyl)ethylamine (126) was synthesized from

compound 125 following the published procedure for amide coupling by

employing the carboxy isoindoline nitroxide compound 106 and EDCI in the

presence of HOBt to give N-tert-butoxycarbonyl-2(4-N-(5-carboxy-1,1,3,3-

70

tetramethylisoindoline)aminophenyl)ethylamine (126) in 77% yield (Scheme

2.6).

Scheme 2.6: Overview of EDCI amide coupling of CTMIO nitroxide with para-aminoethylaniline linker moiety (127), Reagents and conditions: (i) EDCI/HOBt, DMF, DIPEA 25 °C, 77%; (ii) 4 M HCl solution, 1,4-

dioxane:water(1:1), 55–60 °C, 5 h.,89%.

1-Ethyl-3-(3-dimethylaminopropyl)carbodiimide hydrochloride (EDCI) acts as an

acid activating agent, forming O-acylisourea a reactive ester intermediate in situ

with the carboxylic acid of the antioxidant moiety. This O-acylisourea formed

has an activated leaving group and is attacked by the 1-Hydroxybenzotriazole

hydrate (HOBt) to form HOBt ester. A thermodynamically stable urea by-

product is released. The nucleophilic aniline is reacted with HOBt ester to give

the novel amide target (Scheme 2.7).

71

Scheme 2.7: Mechanism of EDC promoted amide formation.

Compound 126 appeared on a TLC plate as a single spot, at a Rf value of 0.64

in ethyl acetate/pet ether (8:2). Compound 126 contained a free radical species

and was difficult to characterise by 1H NMR and 13C NMR spectroscopy as

paramagnetic broadening was observed, however signals of the aniline linker

moiety could be detected. The melting point of product 126 was 158–160 °C.

The structure of 126 was supported by HRMS, which showed the expected

molecular ion of M+Na at 475.2450 m/z (EI+ (HRMS) showed a deviation of

0.0010 from a calc. mass of C26H34NaN3O4).

The acid-labile Boc-protecting groups of the amide linkers were then removed

using 4 M HCl to provide the novel primary amine 2-(4-N-(5-carboxy-1,1,3,3-

tetramethylisoindoline)4-aminophenyl)ethylamine (127) (Scheme 2.6). The

nitroxide containing linker compound 126 was stirred at 55–60 °C for 2–3 h in a

72

mixture of 1,4-dioxane and water with a dropwise addition of 4 M HCl over 10

minute. Analysis by TLC indicated a loss of starting material, and ninhydrin

staining highlighted the generation of a product expected to be the reformed

primary amine 127 on the TLC plate with an intense purple colour with an Rf of

0.46 in chloroform/methanol (9:1). The compound was purified by column

chromatography to give an 89% yield of the deprotected primary amine product

127 with the sharp melting point of 176–178 °C.

The structure of 127 was supported HRMS, which showed the expected

molecular ion of M+H at 353 m/z (EI+ (HRMS) showed a deviation of 0.0011

ppm from a calc. mass of C21H27N3O2). In the 1H NMR spectrum, paramagnetic

broadening was observed due to the nitroxide radical that resulted in some

signals not being detected or giving poor integration. However, signals

corresponding to the aniline and the ethylene linker moiety could be detected,

the aromatic signals at 7.21 and 7.61ppm.

2.2.2 Synthesis of C2-substituted adenosine intermediates (146)

Adenosine receptors were first investigated as a potential drug target in 1981148,

149 and the compound NECA was quickly identified as the prototypic A2

adenosine receptor agonist.279 Adenosine analogues with amide functionality at

the C5-position have been the focus of research since that time. Further

investigation of the A2A adenosine receptor in 1990280 and the discovery that the

C5 amide adenosine analogues are active at this receptor has subsequently

prompted more work in this area.

73

Convergent approach to synthesis of adenosine intermediate 2.2.2.1

A general synthesis of C2-modified N-alkyl-5-N-carboxamide was published by

Jacobson and Kim et al. in 1994 and has been subsequently adopted as the

standard procedure for generating such analogues. The synthetic route most

commonly reported in the literature 223, 259, 263, 272, 274, 281-283 has several

drawbacks for the targets of this research project. Convergent synthesis has

been performed on numerous occasions with varying degrees of success, and

this approach was initially explained here. Such synthetic methods involve

separate modification of the appropriate –D-ribose sugar, followed by a

Vorbrüggen coupling of the anomeric N9 of the purine ring compound. This

compound has been modified at C6 and which can then be substituted at C2 as

required259, 262, 270, 272, 274, 281, 282 (Scheme 2.8).

74

Scheme 2.8: Convergent approach to adenosine A2AAR agonists: Reagents and conditions: (i) Candida rugosa lipase, 1,4-dioxane, 0.1 M sodium phosphate buffer (pH = 7); (ii) TEMPO/BAIB, MeCN/H2O (1:1);

(iii) EDCI, DMAP, MeOH; (iv) R-NH2, t-BuOH, 80 °C; (v) HMDS, MeCN, (NH4)2SO4, TMSOTf; (vi) EtNH2, THF; (vii) R-NH2, EtOH, 120 °C.

The literature shows that the convergent approach gives poorer yields than a

linear approach. Modification of the ribose ring has proved challenging, time-

consuming, and only produced moderate yields. The Vorbrüggen reaction was

difficult to scale up, and substitution at the 2-chloro requires sufficiently harsh

conditions, which threaten the stability of the ribose and antioxidant moieties.

Instead, a linear approach proved more reliable than the convergent method

and afforded acceptable yields.259

75

A linear synthetic approach to this class of compounds tends to have many

steps and, therefore, provides reduced overall yields. The literature proves that

the potential starting materials, including inosine284, 285 and xanthosine286, 287

derivatives, are often difficult to work with. This difficulty is because of their

poor solubility, which requires the use of harsh reaction conditions including the

use of DMSO, DMF, and strongly acidic media. In addition, the similarity in the

reactive groups, which make the regioselectivity modification difficult. This

linear synthetic approach towards C2-substituted adenosine compounds was

based on the work of Hutchison and was improved upon here by modifying the

methods of Foitzik121, 259 and Scammells.121 This work began from guanosine

as the starting material and progressed to selective N6-, C2- and C5-modified

adenosine derivatives. The single change of 5-ethylcarboxylate was

introduced instead of 5-methylcarboxylate in this scheme because of

convenience and because it produced a better yield and improved the quality of

the product synthesised. The synthetic methodology was published121, 259 and

is presented here in detail as it pertains to this project.

Adenosine intermediate for C2-functionalization with a linear 2.2.2.2

approach121

The planned series of adenosine target compounds was obtained successfully

via the 2-fluoro-2,3-O-isopropylideneadenosine-5-N-ethylcarboxamide (146)

intermediate, which was synthesised from guanosine in seven steps of efficient

synthesis (Scheme 2.9).

76

Scheme 2.9: Overview of synthesis of the key adenosine intermediate (146).

Because all of the target compounds bear the 5–NECA modification (Figure

1.15 and 2.1), this was installed first. Then C6-modifications were achieved

with regioselective substitution by the installation of an active, leaving group at

the C6-position (144). The nucleophilic aromatic substitution (SNAr)-reactive

fluorine was achieved at C2 (145). The C6-leaving group was quickly replaced

by the amino group (146). The detailed reaction steps are discussed below.

The secondary 2,3-alcoholic groups of guanosine (138) are more nucleophilic

than the primary 5-alcoholic group, which is the required target for our initial

part of the synthesis. To avoid any side reactions from these secondary

alcoholic groups throughout the process, they were protected by forming a

cyclic acetal with an isopropylidene-protecting group, according to the

procedure originally outlined by Hampton288 (Scheme 2.10).

77

Scheme 2.10: Overview of 2, 3-alcohol protection (139)288

Reagents and conditions: (i) Acetone; 2, 2-

dimethoxypropane, 19 h. 75%.

Another benefit of the formation of protected guanosine is the increasing

solubility of the substrate in a variety of solvents other than DMSO. Solubility of

guanosine is one of the obstacles in the development of the efficient synthesis,

and guanosine is a highly polar species and is less soluble in solvents other

than DMSO.

The reaction was carried out using the acid catalyst para-toluene sulfonic acid

under anhydrous conditions with acetone as the solvent, however, acetal

formation was hindered by the presence of water with 2, 2-dimethoxypropane.

The workup involved using water and sodium bicarbonate to neutralise the

acidic environment and to precipitate the desired product 2,3-O-

isopropylideneguanosine (139) in a yield of 75%. Complete drying of product

139 was very difficult because of the aqueous work-up, so a quick methanol

wash was introduced after filtration of the product from the aqueous medium.

The use of para-toluenesulfonic (tosic) acid in 10-fold excess as the acid

catalyst aided the solubility of the substrate in this reaction because it formed

acetone-soluble salts with the purine nucleoside.288 The isopropylidene

78

protecting group is very labile under acidic conditions, and all further reactions

were carried out using neutral or basic conditions.

Because of the overall length of the synthetic sequence, the first reaction was

undertaken on a scale of 10 g of starting material. Yields were typically

excellent (75-90%). It remains a highly effective reaction in terms of its ease

and general applicability to protect nucleoside 2,3–hydroxyl groups, and it

continues to feature prominently in the recent literature.289,290,291,271, 292,293

The reaction gives the single reaction product (as identified on TLC) with a Rf of

the product of 0.72 in EtOAc/MeOH (7:3). In the 1H NMR (d6-DMSO) spectrum

of compound 139, two singlets appeared at 1.5 ppm and 1.3 ppm with an

integral corresponding to three protons each. In the 13C NMR spectrum, two

additional signals corresponding to the two methyl groups of the acetal moiety

appeared at 25.6 ppm and 27.5 ppm. The melting point of product 139 is 260–

262 °C and is consistant with literature value. The HRMS of the isolated

compound supported the synthesis of 139, which showed the expected

molecular ion of M+H at 324.1286, (EI+ HRMS showed a deviation of 0.0014

ppm from the calc. mass of C13H18N5O5).

The next step in the synthesis of 146 was the oxidation of the 5-hydroxy group

of 139. The traditional oxidising agents used to oxidise primary alcohols involve

harsh conditions and toxic reagents (e.g., Cr(VI) oxides; periodate/ruthenium;294

79

KMnO4; Pd/O2).295 The standard methods of oxidation of the 5-hydroxy group

gave poor yields and were carried out in strong acidic or alkaline conditions,

which resulted in degradation of the substrate.223 These methods can also

create a problem with regioselectivity for compounds with more than one

oxidisable group. The 5-hydroxy group of compound 139 was then oxidised to

the corresponding 2,3-O-isopropylideneguanosine-5-carboxylic acid (140).

The Epp and Widlanski296 oxidation implemented using the mild oxidising agent

system including, the free radical species TEMPO and a stoichiometric amount

of the organic oxidant bis(acetoxy)iodobenzene (BAIB) (Scheme 2.11).295

Scheme 2.11: Overview of the 5-hydroxyl oxidation (140). Reagents and conditions: (i) [Bis (acetoxy)

iodo] benzene (BAIB), TEMPO, MeCN, H2O, 18 h,79%.

This oxidation progressed under aqueous conditions (1:1 solution of

acetonitrile/water) with the two-step mechanism that led to the formation of the

aldehyde which was oxidised further to give the 5-carboxylic acid (Scheme

2.11). The aqueous conditions were used to allow the reaction to stir overnight

(Epp & Widlanski296 (3 h) and Middleton292 (4 h). In this process, the

hydroxylamine formed was reoxidised to TEMPO in the presence of BAIB,

which contains hypervalent (+3) iodine 279. The nitroxide radical is very stable

because of the absence of -protons on the ring, which can cause

disproportionate reactions and form a cyclic hydroxylamine and nitrone.297

80

Although it is not completely proven, the following mechanism was proposed by

de Mico (Scheme 2.12).293

Scheme 2.12: Mechanism of the primary hydroxyl oxidation.

The reaction has several additional benefits including the use of relatively non-

toxic reagents that can be used safely in the presence of a wide range of

functional groups and protecting groups such as acetal and ethers296 and

secondary alcohols.279 These reagents are highly selective for the oxidation of

primary alcohols.279 The by-products of oxidation were iodobenzene and acetic

acid, which could be easily removed via precipitation of the product from

acetone and diethyl ether.298,289 Often, in oxidation reactions, the solution is

homogeneous and opaque; however; in these reaction conditions, the solution

was heterogeneous and biphasic before the workup.

81

The workup, which involved adding some acetone and then adding the solution

to a large volume of Et2O, produced a precipitate. This precipitate was filtered

and gave reliable crude yields in the range of 70–80%. The Rf of compound

140 in 4:1 EtOAc/MeOH returned to 0 (as identified on TLC) because of the

increase in polarity around the 5-carbon from the introduction of the

electronegative oxygen to form the carbonyl group. The purification of the acid

was not successful using recrystallisation and column chromatography.

There are two main characteristics of the 1H NMR (d6-DMSO) spectrum of

crude compound 140. The signal at 3.54 ppm for the 5-methylene protons was

absent because of their removal by the successful oxidation of the 5-carbon.

Conversion to the carboxylic acid also removed the signal observed at 5.06

ppm for the remaining hydroxyl proton. In the 13C NMR spectrum, the signal for

the oxidised 5-carbon moved 112 ppm downfield from the corresponding signal

for 139 because of the powerful deshielding effect of the electron-withdrawing

carboxyl group and reappeared at 171.37 ppm. The melting point of the desired

acid 140 was 210–212 °C259 and is consistent with literature value. The

synthesis of compound 140 supported by HRMS, which showed the expected

molecular ion of M+H at 338.1134 m/z, (EI+ HRMS showed a deviation of

0.0033 ppm from the calc. mass of C13H16N5O6).

Despite evidence that the compound produced was desired target 140, purity of

the carboxylic acid was not satisfactory. Forming a novel intermediate ethyl

ester 142 at the 5–carboxyl group was a more reliable and higher-yielding route

82

to the desired 2,3-O-isopropylideneguanosine-5-N-ethylcarboxamide (143).299

Novel 5-ethylester 142 was synthesised under standard esterification

conditions (Scheme 2.13).

Scheme 2.13: Overview of 5–ethyl ester formation (142). Reagents and conditions: (iii) SOCl2, ethanol, 15

h, 64%.

Compound 140 was reacted at 0 °C in absolute ethanol with a fourfold excess

of thionyl chloride to give the reactive acid chloride in situ. The mixture was

stirred at room temperature overnight, which allowed the ethyl ester to form

through nucleophilic acyl substitution.300 The alcohol was used in large excess

(as solvent) optimising the yield of 2,3-isopropylideneguanosine-5-

ethylcarboxylate (142) (Scheme 2.13). The reaction solution was then basified

with an excess saturated NaHCO3 solution to quench any remaining thionyl

chloride and HCl. The acidity of the conditions did not appear to affect the

purine riboside linkage adversely.

The workup involved removal of ethanol by distillation after basification, and the

residue was stirred with the water to remove excess NaHCO3. The desired

product 142 was obtained in 65% yield. The Rf of the product on TLC in 7:3

83

EtOAc/MeOH increased to 0.71 because of the decrease in polarity around the

5-carbon from the introduction of the ethyl group to form the ester group.

In the 1H NMR (d6-DMSO) spectrum of 142, a multiplet appeared at 3.7–3.9

ppm with an integral corresponding to two protons and a triplet appeared at 0.9

ppm for three protons of the ethyl group of the new ester substituent. An extra

two peaks corresponding to the carbon of the ethyl group also appeared in the

13C NMR spectrum at 61.1 ppm and 13.8 ppm, respectively. The melting point

(272–274 °C) of the isolated compound 142 was consistant and sharp. The

high-resolution mass spectrum of the isolated compound supported the

synthesis of 142, which showed the expected molecular ion of M+H at 366.1449

m/z, (EI+ HRMS showed a deviation of 0.0035 ppm from the calc. mass of

C15H20N5O6).

Compound 142 was converted to the desired 2,3-O-isopropylideneguanosine-

5-N-ethylcarboxamide (NECA) (143) by using liquid ethylamine (Scheme 2.14).

Scheme 2.14: Overview of 5-amide formation (143). Reagents and conditions: (iv) Ethylamine, EtOH, -

20°C, 3 h. 88%.

84

Formation of the NECA 143 was achieved following a novel procedure by

combining the 5-ethyl ester 142 directly in an excess of dry ethylamine that was

used both as a reagent and as a solvent at a temperature of –20 °C for 3.0 h.

The reaction mixture was allowed to room temperature and stirred overnight at

room temperature. This overnight stirring was used for to react the unreacted

starting material and to remove the excess of ethylamine. The reaction mixture

was then diluted with absolute ethanol. The product mixture was then adsorbed

on silica and separated by column chromatography to obtain pure compound

143 in an 82 % yield.

The conditions were sufficient to enable nucleophilic acyl substitution of the

ethoxy group by the nucleophilic amine to form the amide in excellent yield.

The reaction was conducted no more than a 3 g scale. Formation of the amide

bond increased the polarity of the molecule, and the Rf value of compound 143

declined to 0.43 (9:1 DCM/MeOH).

The successful reaction to produce 143 was analysed by the 1H NMR (d6-

DMSO) spectroscopy. A multiplet appeared at 2.7–2.9 ppm with an integral

corresponding to the two protons and a triplet appeared at 0.6 ppm for three

protons of the ethyl group of the new amide substituent. An extra two peaks

corresponding to the carbons of the ethyl group also appeared in the 13C NMR

spectrum at 61.1 ppm and 13.8 ppm, respectively. The results of analysis using

HRMS and the melting point (278–280 °C) of the isolated compound were

consistent with the expected results for desired compound 143, which showed

85

the expected molecular ion of M+H at 365.1574 m/z, (EI+ HRMS showed a

deviation of 0.0001 ppm from the calc. mass of C15H21N6O5).

Installation of a stable and effective leaving group was achieved by the

(bezotriazole-1-yloxy)tris(dimethylamino)phosphoniumhexafluorophosphate

(BOP)/ 1,8-diazabicycloundec-7-ene (DBU) method (Scheme 2.15) of Bae and

Lakshman’s group.301 As the substrate was only sparingly soluble, the addition

of DBU assisted the dissolution of the substrate, and stirring was continued

overnight.

Scheme 2.15: Overview of leaving group formation (144). Reagents and conditions: (v) MeCN, BOP, DBU,

16 h, 25°C 98%.

The role of the base is critical because it abstracts the N1-proton from the

purine ring, which forms a tautomer with the nucleophilic C6-oxo anion. The

attack of the BOP reagent at the electrophilic phosphonium, forming an O6-

phosphonium intermediate and the nucleophilic OBt anion. The OBt anion then

attacks the now activated (partially positively charged) C6 in a SNAr addition–

substitution reaction, forming a Meisenheimer intermediate.299 Rearomatisation

expels the phosphonium substituent to give the desired product and produces a

highly thermodynamically stable phosphine oxide in the process (Scheme 2.16).

86

Scheme 2.16: Mechanism of leaving group formation.

Different initiating bases have been explored in the literature302 and because

DBU is widely considered the superior reagent, it was used here. Initial studies

used BOP in its normal capacity as a coupling reagent to create a reactive in

situ intermediate that would then undergo further immediate nucleophilic

aromatic substitution.302,277 However, the intermediate itself, containing the O6-

benzotriazol-1-yl (6–OBt) moiety, is very stable and can be easily isolated in

high yield and purity.301,289 This useful intermediate can be activated for

nucleophilic aromatic substitution at the C6-position of the purine ring to then be

one relatively straightforward diazotisation step away from the 2-halo series.

The halogenated compounds provide starting material from which further

substitutions at the critical C2-position of the purine ring in the A2AAR can be

made.

87

The introduction of the benzotriazole group decreased the polarity around the

C6-position of guanosine molecule to form the better leaving group. The Rf

value of the product O6-(benzotriazol-1-yl)-2,3-O-isopropylideneadenosine-5-

N-ethylcarboxamide (144) decreased to 0.38 in 8:2 DCM/MeOH. In the 1H

NMR spectrum (d6-DMSO) of 144, four additional peaks from the phenyl

protons of the heteroaromatic OBt substituent were located between 7.5 ppm

and 8.3 ppm, with a multiplet at 7.55 ppm with an integral corresponding to one

proton, a multiplet at 7.66 ppm with an integral corresponding to two protons,

and a doublet at 8.19 ppm with an integral corresponding to one proton. The

multiplet at 7.66 ppm corresponds to the two aromatic protons furthest from the

triazole heterocyclic and the doublet to the aromatic proton most proximal to the

nitrogen–oxygen bond. Six new peaks corresponding to the two quaternary and

four proton-bearing OBt phenyl carbons appeared in the 13C NMR spectrum

between 110 ppm and 130 ppm. The melting point of the isolated compound

144 was 140–142 °C. The HRMS supported the synthesis of 144, which

showed the expected molecular ion of M+H at 482.1898 m/z, (EI+ HRMS

showed a deviation of 0.0002 ppm from the calc. mass of C21H24N9O5).

Functional group interconversion of the C2-amine of compound 144 to the

nucleophilic aromatic substitution-promoting fluorine was accomplished using

the method of Kim et al.273 (Scheme 2.17). This method proceeded smoothly

with good yields (60–75%) of 2-fluoro-O6-(benzotriazol-1-yl)-2,3-O-

isopropylideneadenosine-5-N-ethylcarboxamide (145) obtained on scales

ranging from 50 mg to 2.0 g.

88

Scheme 2.17: Overview of fluoro substitution at the C2-position (145). Reagents and conditions: (vi) t-

BuONO, 70% HF in pyridine, 68%.

The reaction conditions employed were reasonably harsh with HF (70% in

pyridine) used as the fluorinating agent and pyridine as the solvent. This

reaction required the temperature to be kept between –50 °C and –30 °C.

Although distinguishable by TLC, the Rf value for the product 145 was 0.45 in

1:4 P.S./EtOAc (Rf of 0.37 for the C2-amino compound 144).This change in Rf

value indicates that the polarity of the molecule was decreased only slightly by

the introduction of the highly electronegative fluorine atom.

In the 1H NMR spectrum (d6-DMSO), the broad singlet at 6.68 ppm

corresponding to the heteroaromatic aniline at the C2-position in 144 was not

present in the product 145. In the 13C NMR spectrum of the product 145, the

peak for the C2-carbon of the purine ring was split by coupling with the fluorine

to produce a doublet at 157.1 ppm with a coupling constant of 221.8 Hz.

Further, less pronounced splitting occurred at C4 and C6 (J = 16.4 Hz). No

splitting was observed at sites more remote from C2. The melting point of the

isolated compound 145 was 145–147 °C. The synthesis of isolated compound

89

145 was also supported using HRMS, which showed the expected molecular

ion of M+H at 485.1699 m/z, (EI+ HRMS showed a deviation of 0.0007 ppm

from the calc. mass of C21H22FN8O5).

The labile benzotriazolyl leaving group present in compound 145 is readily

displaced by nucleophilic amines.299, 302 Ammonia solution was used to form a

6-amino product 2-fluoro-2,3-O-isopropylideneadenosine-5-N-

ethylcarboxamide (146) (Scheme 2.18). This SNAr reaction occurred smoothly

overnight in acetonitrile at room temperature.

Scheme 2.18: Overview of N6-amino substitution (146) Reagents and conditions: (vii) 28% ammonia sol,

MeCN, 2 h, 85%.

The Rf of the product 146 decreased as the benzotriazolyl group was lost, and

the polarity increased slightly. The Rf of compound 146 was determined to be

0.62 in ethyl acetate/MeOH (7:3). Purification via column chromatography gave

good yields of a white solid for 250 mg to 1 g scale reactions. A second

component was separated during the column chromatography was identified as

a small amount of amino-substituted product at the C2-position. The diamino

substitution was observed to be present in < 5% most typically when longer

90

reaction times led to some of the fluorine group being replaced by the amino

group.

In d6-DMSO, the major features of the 1H NMR spectrum of the fluoro

adenosine intermediate 146 are the absence of the signals from the

benzotriazole aromatic protons, and the appearance at 7.85 ppm of a broad

singlet with an integral corresponding to the two N6 protons. The signals from

the 2’- and 3’-protons completely coalesce from two resolved doublets in the

starting material to a singlet peak at 5.35 ppm with an integral corresponding to

two protons in the product. Likewise, in the 13C NMR spectrum, six peaks for

the benzotriazolyl aromatic carbons are no longer present. Two identifiable

signals, separated by 0.03 ppm were observed for the 2’- and 3’-carbons. The

melting point of the product 146 was 194–196 °C. The synthesis of compound

146 was supported by HRMS spectrum of 146, which showed the expected

molecular ion of M+ at 366.1451 m/z, (EI+ HRMS showed a deviation of 0.0049

ppm from the calc. mass of C15H19FN6O4).

2.2.3 Synthesis of TMIO adenosine analogues with aminoethylaniline

linker (161)(Approach 1)

Coupling of the antioxidant linker with functionalised adenosine was the next

step in the process towards target 161. In this work, the novel C2-substituted

target compound 2-(2-(4-N-(5-carboxy-1,1,3,3-tetramethylisoindoline-1-yloxyl)

aminophenyl)) ethylamino-2,3,-O-isopropylideneadenosine-5-N-

ethylcarboxamide (161) was synthesised through the reaction of the 2-fluoro

adenosine analogue (146) with an appropriate antioxidant-bearing

91

aminoethylaniline linker moiety 127 (Scheme 2.19) by following the

Hutchison’s223 synthetic procedure.

Scheme 2.19: Overview of coupling of nitroxide with aminoethylaniline linker to adenosine intermediate (approach 1) (161) Reagents and conditions: (i) DIPEA, Ethanol, 75–80 °C, 7 days, 58%.

The reaction proceeded via a nucleophilic aromatic substitution (SNAr) reaction

according to Hutchison’s method, and the reaction mechanism is explained in

Scheme 2.20.

92

Scheme 2.20: SNAr mechanism for the synthesis of C2-substituted target compounds.

Trivedi265, Caddell272 and Kim274 documented that nucleophilic substitution at a

C2-bromo and chloro moiety, however, these substitutions was considered

insufficiently reactive and time inefficient and were therefore not attempted.

Instead, efforts were directed towards the more reactive C2-fluoro species.

However, Takvorian has reported reaction temperatures well above the

200°C,303 which are perhaps unnecessarily. The rate-limiting step in the

reaction is the initial nucleophilic attack on C2-carbon and the formation of a

Meisenheimer intermediate, and this explains why the 2-fluoro compound is

more reactive with respect to SNAr substitutions. In this regard, the substituent

with the highest electronegativity produces a more partial positive charge at the

C2-position of adenosine and, therefore, electronic conditions are more prone to

nucleophilic attack.

93

The synthesis of product 161 was achieved in ethanol using Hunig’s base (N,

N-diisopropylethylamine)(DIPEA) by stirring at 75–80 °C for 7 days. The Rf

value of the product on TLC in EtOAc/MeOH (9:1) was 0.51 and the compound

was purified by column chromatography using this solvent mixture to generate

product 161 in 58% yield.

The structure of the nitroxide-coupled adenosine product 161 was supported by

the HRMS, which showed the expected molecular ion of M+H at 699.3506 m/z,

(EI+ HRMS showed a deviation of 0.0017 ppm from the calc. mass of

C36H45N9O6). In the 1H NMR spectrum, paramagnetic broadening was

observed as expected due to the nitroxide radical that resulted in some peaks

not being detected and giving poor integration. However, the signal of the

ethylene group from aniline linker could be detected at 2.91 and 2.98 ppm and

the adenosine ribose ethylamide proton signals could be detected at 0.71 ppm

and acetal two methyl protons appeared at 1.24 and 1.42 ppm. The melting

point of the isolated compound 161 was 66–68°C.

In this approach, the reaction conditions used were very harsh, and the product

was only generated in low yields. This low yield was attributed to

decomposition of the starting reactants or product formed. In addition, this

approach required extended reaction times (7 days). Accordingly, an

alternative approach was explored. This approach first involved the attachment

of the aminoethylaniline linker to the adenosine intermediate, followed by

subsequent coupling with the carboxy nitroxide. As the amine linkers could be

94

sourced in large scale, greater efficiency for the synthesis was thought to be

possible through coupling the adenosine intermediate 146 with excess amine

124 and then coupling this system to the nitroxide.

2.2.4 Synthesis of TMIO adenosine analogues with aminoethylaniline

linker (161) (Approach 2)

As outlined in previous section, the reaction of 2-fluoro adenosine precursor 146

with an excess of amine 124 was undertaken in order to give the maximum

possible levels of the preliminary target 2-(2-(4-aminophenyl)) ethylamino-2,3-

O-isopropylideneadenosine-5-N-ethylcarboxamide (159).

In this approach, the para-(aminophenyl)ethylamine-linked adenosine

intermediate 159 was synthesised first and then the appropriate carboxy-

nitroxide was coupled directly to the adenosine intermediate 159. The

advantage of this approach is that various carboxy-nitroxides such as 5-

carboxy-1,1,3,3-tetramethylisoindoline-2-yloxyl (CTMIO, 106), 4-carboxy-

2,2,6,6-tetramethylpieridine-1-yloxyl (CTEMPO, 116), 3-carboxy- 2,2,5,5-

tetramethylpyrrolidine-1-yloxyl (CPROXYL, 117), 5-methylcarboxy-1,1,3,3-

tetramethylisoindoline-2-yloxyl (MCTMIO, 115) and 5-carboxy-1,1,3,3-

tetraethylisoindoline-2-yloxyl (CTEIO, 114) could also be coupled to the para-

aminoethylaniline linked adenosine intermediate 159 using very mild amide

coupling conditions (Scheme 2.21).

95

Scheme 2.21: Overview of coupling of carboxy nitroxides with aminoethylaniline linked adenosine intermediate (Approach 2)

Firstly, the adenosine intermediate 159 was synthesised from the reaction of

compound 146 with amine 124 by using Hunig’s base DIPEA in 97% yields

(Scheme 2.22). The reaction was proceeded by an SNAr reaction mechanism,

which is explained in Scheme 2.20.

Scheme 2.22: Overview of coupling of aminoethylaniline linker to adenosine intermediate (159), Reagents

and conditions: (i) DIPEA, MeCN, MW 180 °C;7 h.

96

The adenosine intermediate 159 was synthesised by using the conventional

method of heating in oil bath. This synthetic method takes 7 days to complete

the reaction and only gives moderate yields (50%). This methodology is time-

consuming and failed to achieve desired yield of product 159. This low yield

(50%) was attributed to the longer heating time which may decompose the

starting reagents, or the product formed. Accordingly, an alternative approach

was implemented which successfully prepared the desired C2-

aminoethylaniline adenosine analogue 159 through a substitution reaction using

microwave assisted conditions in the reduced time span of 7 h with an excellent

yield of 97%. This approach provided a convenient synthetic route to the

synthesis of the desired adenosine analogues.

The reaction was carried out successfully either in ethanol or acetonitrile with oil

bath heating (at 75–80 °C). For complete conversion of the starting material,

the reaction still took 7 days to complete. This longer reaction time, is avoided

by undertaken using microwave irradiation in which compound 146, compound

124 and DIPEA were combined in acetonitrile, sealed in a microwave tube and

heated at 180 °C. Multiple experiments at various reaction times were

conducted under microwave conditions and after 7 h, the complete conversion

of the starting material was observed. After which time, the reaction mixture

was purified using column chromatography.

The structure of 159 was supported by 1H NMR and 13C NMR spectroscopy and

MS spectrometry. In the 1H NMR spectrum, a new peak corresponding to the

97

ethylene protons of the linker appeared in the aliphatic region as two multiplets

at 2.82 and 2.86 ppm integrated for four protons. Two additional doublets

corresponding to the aromatic ring of the linker appeared at 6.67 and 7.06 ppm

which integrated for four protons. In the 13C NMR spectrum, three additional

signals appeared as strong peaks corresponding to the two methyl carbons at

33.7 and 34.3 ppm. The additional aromatic ring signals appeared at 79.1 ppm.

The C2-substituted linked adenosine compound 159 was supported by HRMS,

which showed the expected molecular ion of M+H at 483.2482 m/z, (EI+ HRMS

showed a deviation of 0.022 ppm from the calc. mass of C23H31N8O4). The

melting point of 150-152 °C of isolated product 159 is consistent. The obtained

data were in agreement with the published values.121

Compound 159 was then coupled with nitroxide compound (CTMIO) 106 using

EDCI/HOBt coupling reagent and Hunig’s base in dry DMF to give 2-(2-(4-N-(5-

carboxy-1,1,3,3-tetramethylisoindoline-2-yloxyl)aminophenyl))ethylamino-2,3-

O-isopropylideneadenosine-5-N-ethylcarboxamide (161) (Scheme 2.23). HOBt

helps in the formation of active esters in the reaction mixture by reacting with

the carboxylic acid at ambient temperatures.

98

Scheme 2.23: Synthesis of TMIO-substituted adenosine analogue possessing a substituted aniline linker moiety (161) (approach 2). Reagents and conditions: (i) EDCI/HOBt, DMF, DIPEA 25 °C; (ii) 1 M HCl

solution, MeCN, 55–60 °C, 5 h.

The reaction of 159 with 106 gave two reaction products (as identified by TLC),

which were isolated using medium-pressure liquid chromatography (MPLC).

The main product isolated was attributed to the expected adduct 161 in 81%

yield. In the 1H and 13C NMR spectra of the reaction product 161, paramagnetic

broadening by the nitroxide radical results in some peaks not being detected or

giving poor integration. However, the signal of the ethylene group from aniline

linker could be detected at 2.91 and 2.98 ppm and the adenosine ribose

ethylamide proton signals could be detected at 0.71 ppm and acetal two methyl

protons appeared at 1.24 and 1.42 ppm. The melting point of the isolated

product 159 was 66–68 °C. The synthesis of desired amide 161 was supported

by HRMS, which showed the expected molecular ion of M+H at 699.3506 m/z,

(EI+ HRMS showed a deviation of 0.0013 ppm from the calc. mass of

99

C36H45N9O6). The purity was found to be 97.6% by analytical HPLC in 80%

methanol/20% water system.

Deprotection of the isopropylidene group from compound 161 was achieved

using 1 M HCl at at 55–60 °C in 4.0 h to give 2-(2-(4-N-(5-carboxy-1,1,3,3-

tetramethylisoindoline-2-yloxyl) aminophenyl)) ethylaminoadenosine-5-N-

ethylcarboxamide (162) (Scheme 2.23).

The deprotection reaction gave a single reaction product 162 (as identified on

TLC), which was isolated using MPLC and partial crystallisation in a mixture of

methanol/ water solvent system with a yield of 70%. The structure of

deprotected product 162 was supported by HRMS, which showed the expected

molecular ion of M+H at 659.3197 m/z, (EI+ HRMS showed a deviation of

0.0010 ppm from the calc. mass of C33H41N9O6). A melting point of 241–243 °C

(decomp) was obtained for the product 162. The purity was found to be 99.9%

using analytical HPLC in an 80% methanol/20% water system. In the proton

NMR spectrum, paramagnetic broadening was observed which resulted in

some peaks not being detected and giving poor integration of compound 162.

However, signals of the aniline linker and adenosine moieties could be

detected.

The coupling reaction also gave a small amount of unknown impurity at the

nonpolar region (as identified on TLC) in each reaction. To confirm the

structure of the by-product, which was suspected to be the dimer, several

100

experiments were undertaken. Firstly, the compound 159 was coupled with 3,5-

dinitrobenzoic acid (175) using EDCI/HOBT-coupling reagents (Scheme 2.24).

The same reaction conditions gave the product 176 with a similar impurity to

those of the coupling reactions with radical species (a identified by TLC). The

coupled compound was not isolated further.

Scheme 2.24: Synthesis of 3,5-dinitrobenzoic acid-coupled adenosine analogue possessing a substituted aniline linker moiety (176) Reagents and conditions: (i) EDCI/HOBt, DMF, DIPEA 25 °C.

We next performed another experiment by assuming that the impurity may have

formed because of a side reaction or decomposition of the starting material 159

(Scheme 2.25). This assumption is confirmed by stirring the aniline linked

adenosine compound 159 under the same reaction conditions and without any

carboxy acid starting material. However, no change was observed on TLC,

after three days of continued stirring at room temperature and starting

compound 159 was recovered.

101

Scheme 2.25: Reaction of coupling reagents with adenosine analogue possessing a substituted aniline linker, Reagents and conditions: (i) EDCI/HOBt, DMF, DIPEA 25 °C.

From this experiment, it was confirmed that the unknown compound impurity

was arising from the other starting material the carboxy nitroxide. After analysis,

it was found that the impurity interferring in this reaction was the dimer

compound (anhydried) formed from two CTMIO molecules.

2.3 Synthesis of C2-substituted CTEMPO adenosine analogues possessing substituent attached to ‘aminoethylaniline linker’ moiety (164)

Compound 159 was coupled with commercially available 4-carboxy- 2,2,6,6 -

tetramethylpiperidine-1-yloxyl (CTEMPO) (116) using the EDCI/HOBT amide

coupling reagent to give the novel compound 2-(-2-(4-N-(4-carboxy-2,2,6,6

tetramethylpieridine-2-yloxyl)aminophenyl))ethylamino-2,3-O-

isopropylideneadenosine-5-N-ethylcarboxamide (163) in 60% yield (Scheme

2.26).

102

Scheme 2.26: Synthesis of TEMPO-substituted adenosine analogue possessing a substituted aniline linker moiety (164), Reagents and conditions: (i) EDCI/HOBt, DMF, DIPEA 25 °C; (ii) 1 M HCl solution,

MeCN, 55–60 °C, 5 h.

The reaction was carried out in freshly distilled DMF under an argon

atmosphere at room temperature. The reaction went to completion in 3 days

(as identified by TLC).

The reaction of 116 with 159 produced two products (as identified by TLC)

which were isolated and purified twice by MPLC, as the two components ran

very close to each other. In the 1H spectrum of the reaction product 163,

paramagnetic broadening by the nitroxide radical resulted in some peaks not

being detected or giving poor integration. However, the signal of the aniline

linker and adenosine moieties could be detected. The results of HRMS of the

isolated compound were consistent and supported the structure of 163, which

showed the expected molecular ion of M+H at 665.3658 m/z, (EI+ HRMS

showed a deviation of 0.0018 ppm from the calc. mass of C33H47N9O6). A purity

103

of 99.4% was found using analytical HPLC in an 80% methanol/20% water

system. The melting point of compound 163 was measured as 148–150 °C.

The second spot isolated was found to be the mass corresponding to the

anhydride of the CTEMPO (116), which was found according to the mass

analysis.

Deprotection of the isopropylidene group from compound 163 was achieved

using similar procedure as used for synthesis of compound 162 by employing 1

M HCl at elevated temperatures to give the novel target compound 2-(2-(4-N-(4-

carboxy-2,2,6,6-tetramethylpieridine-1-

yloxyl)aminophenyl))ethylaminoadenosine-5-N-ethylcarboxamide (164)

(Scheme 2.26). The reaction went to completion in 5.0 h at 55–60 °C.

The reaction gave a single reaction product (as identified on TLC) with an Rf

value of 0.43 in (8:2) IPA: MeOH, and which was purified using MPLC and

partial crystallisation using a 75% methanol/25% water system to give 164 in

70% yield. The melting point of 211-213 °C (decomp) was obtained for the

isolated product 164. The structure of 164 was supported by HRMS, which

showed the expected molecular ion of [M+H]: 625.3341 m/z (EI+ HRMS showed

a deviation of 0.0011 from the calc. mass of C30H43N9O6). In the 1H spectrum of

the reaction product 164, paramagnetic broadening by the nitroxide radical

resulted in some peaks not being detected or giving poor integration, however

the signal of the aniline linker and adenosine moieties could be detected.

104

2.4 Synthesis of C2-substituted CPROXYL adenosine analogues possessing substituent attached by ‘aminoethylaniline linker’ moiety (166).

The compound 159 was coupled with commercially available 3-carboxy-2,2,5,5-

tetramethylpyrrolidine-1-oxyl (CPROXYL) (117) using similar reaction conditions

as used to synthesize compound 163 with EDCI/HOBT as the amide coupling

reagent to give the novel compound 2-(2-(4-N-(3-carboxy-2,2,5,5-

tetramethylpyrrolidine-1-yloxyl)aminophenyl))ethylamino-2,3-O-

isopropylideneadenosine-5-N-ethylcarboxamide (165) in 74% yield (Scheme

2.27).

Scheme 2.27: Synthesis of the PROXYL-substituted adenosine analogue possessing a substituted aniline linker moiety (166) Reagents and conditions: (i) EDCI/HOBt, DMF, DIPEA 25 °C; (ii) 1 M HCl solution,

MeCN, 55–60 °C, 5 h.

The reaction was carried out in freshly distilled DMF under an argon

atmosphere at room temperature. The reaction went to completion in 24 h (as

105

identified by TLC). The reaction of 117 with 159 gives two products (as

identified by TLC), which were subsequently isolated by MPLC in a 75%

methanol/25% water system. The results of HRMS of the main component

isolated were consistent and supported structure of 165, which showed the

expected molecular ion of M+H at 651.3493 m/z, (EI+ HRMS showed a

deviation of 0.0003 ppm from the calc. mass of C32H45N9O6). The melting point

of the isolated product 165 was found to be 140–142 °C. The purity was found

to be 98% by analytical HPLC in a 75% methanol/25% water system. The

second component isolated from the reaction mixture was found to be the

unreacted substrate compound 159 by MS analysis.

The novel compound 2-(2-(4-N-(3-carboxy-2,2,5,5-tetramethylpyrrolidine-1-

yloxyl) aminophenyl)) ethylaminoadenosine-5-N-ethylcarboxamide (166) was

synthesised by deprotection of the isopropylidene group from compound 165

using 1 M HCl at 50–60 °C for 5.0 h in 62.5% yield. (Scheme 2.27).

The reaction gave a single reaction product 166 (as identified on TLC), which

was isolated using MPLC and partial crystallisation using 75% methanol/25%

water system. The results of HRMS of the isolated compound were consistent

with the expected results for 166, which showed the expected molecular ion of

M+H at 611.3183 m/z, (EI+ HRMS showed a deviation of 0.0001 ppm from the

calc. mass of C29H41N9O6). In the 1H spectrum of the compound 166,

paramagnetic broadening by the nitroxide radical resulted in some peaks not

being detected or giving poor integration of compound 166, however signals of

106

the aniline linker and adenosine moieties could be detected. A melting point of

176–178 °C (decomp) was measured for the isolated product 166. The purity

was found to be >99.9% using analytical HPLC in a 75% methanol/25% water

system.

2.5 Synthesis of C2-substituted MCTMIO adenosine analogues possessing substituent attached to “aminoethylaniline linker” moiety (168)

A novel adenosine compound was synthesised using a mixture of the one

carbon extended methylene carboxy isoindoline nitroxide 115, and compound

106 which was synthesised from compound 102 and obtained from advanced

stage synthesis in the laboratory. The compound 159 was coupled with a

mixture of nitroxide 5-methylenecarboxy-1,1,3,3-tetramethylisoindoline-2-yloxyl

(MCTMIO, 115) and 106 using EDCI/HOBT as the amide coupling reagent to

give a mixture of 2-(2-(4-N-(5-methylenecarboxy-1,1,3,3-tetramethylisoindoline-

2-yloxyl)aminophenyl))ethylamino-2,3-O-isopropylideneadenosine-5-N-

ethylcarboxamide (167) and compound 161 (Scheme 2.28).

107

Scheme 2.28: Synthesis of MCTEIO-substituted adenosine analogue possessing a substituted aniline linker moiety (168). Reagents and conditions: (i) EDCI/HOBt, DMF, DIPEA 25 °C; (ii) 1 M HCl solution,

MeCN, 55–60 °C, 5 h.

The reaction was carried out in freshly distilled DMF under an argon

atmosphere at room temperature. Addition of the mixture of nitroxides after the

coupling reagents, improves the reaction completion time from 48 h to 24 h and

product 167 was obtained in a high yield. Moreover, complete consumption of

the starting material was observed. The addition of EDCI and HOBt first to

DMF under an argon atmosphere followed by the addition of carboxy nitroxide

115 and DIPEA, and later the portion-wise addition of the linker amine 159 led

to completion of the reaction in 24 h instead of 3 days at room temperature (as

identified by TLC). The work-up involved filtration of the coupled solid product

formed at 0 °C after the 30 min reaction, quenching and washing many times

108

with chilled water. As per TLC analysis, the similar Rf values were observed for

the mixture of compounds 167 and 161.

The synthesis of the one carbon extended isoindoline nitroxide derivative of

adenosine compound 167 was attempted by following a similar amide coupling

procedure used for the synthesis of compound 165. The reaction of mixture of

115 and 106 with 159 produced three components (as identified by MPLC),

however, a TLC analysis shows only two components. These components was

firstly purified by using MPLC. The first fraction was found to be unreacted

starting amine 159, which was isolated from the product. The second fraction

isolated from the 300mg reaction by MPLC was 150 mg of a pale yellow

component. The second component on TLC, what appeared to be a single new

product has two close components (as identified by HPLC). However, it was

found difficult to isolate these two compounds present in the sample by MPLC

and therefore, this mixture was used as it is for the next step of deprotection

reaction.

The mixture was analysed using analytical HPLC in a 75% methanol/25% water

system and was found to be a 60:40 mixture of compound 167 with compound

161 respectively. The results of HRMS of the isolated product were consistent

with the expected results for 167 and 161, which showed the expected

molecular ions of M+H at 713.5496 and 699.9465 m/z respectively. The NMR,

MS and HPLC analysis revealed that two structures-related isoindoline

compounds were present and determined that a component had a molecular

109

mass corresponding to the desired MCTMIO adenosine derivative 167 with the

CTMIO adenosine derivative 161 in a 60:40 mixture.

A mixture of compound 167 and 161 was used as starting material for the

deprotection reaction. Deprotection of the isopropylidene group from a mixture

of compound 167 and compound 161 was achieved using 1 M HCl at elevated

temperatures (Scheme 2.28). The reaction went to completion in 5.0 h at 50–

55 °C and produced a mixture of the final target compound 2-(2-(4-N-(5-

methylcarboxy-1,1,3,3-tetramethylisoindoline-2-

yloxyl)aminophenyl))ethylaminoadenosine-5-N-ethylcarboxamide (168) and

162 as an off-white solid compound. (Note: compound 162 is present because

the starting material contained two components). The product was filtered after

quenching of the reaction using saturated sodium bicarbonate solution at 0 °C

and was washed with plenty of chilled water.

The reaction gave a mixture of compounds 168 with 162 (single spot as

identified on TLC) that was not isolated using MPLC but partially crystallised in

75% methanol/25% water system. The results of HRMS of the isolated

compound were consistent with the expected results for 168 and 162, which

showed the expected molecular ion of M+2H at 674.6586, and 661.0205 m/z.

The isolated off-white solid was found to be a 60:40 mixture of compound 168

with 162 using analytical HPLC in a 75% methanol/25% water mobile-phase

system. This compound was analysed by 1H NMR spectroscopy, which

indicated clearly the absence of the two methyl signals of isopropylidene

protection.

110

2.6 Synthesis of C2-substituted adenosine analogue possessing TEIO nitroxide substituent attached to the ‘aminoethylaniline linker’ moiety (170).

2.6.1 Synthesis of carboxy TEIO nitroxide antioxidant

The stable nitroxide 5-carboxy-1,1,3,3-tetraethylisoindoline-2-yloxyl (CTEIO)

114 was synthesised in six steps as per the procedure first published by our

group 102, 275 (Scheme 2.29).

Scheme 2.29: Synthesis of 5-Carboxy-1,1,3,3-tetraethylisoindoline-2-yloxyl (CTEIO) 114.102

Reagents and

conditions: (i) Glacial CH3COOH, reflux 75 min, 80%; (ii) Mg, C2H5I, toluene, reflux 3.0 h, 32%; (iii) 60 psi H2, 10% Pd/C catalyst, glacial CH3COOH, 3 h, 92%; (iv) Na2WO4. 2H2O, H2O2, NaHCO3, stirred 72 h, 47%; (v) AcCl, Pd/C,Et3N, 99%; (vi) KMnO4 , MgSO4, 78%; (vii) LiOH, 89%.

Commercially available 5-methyl phthalic anhydride (107) and benzylamine

were reacted in a nucleophilic acyl substitution to generate 5-methyl-N-benzyl

phthalimide (108) in 80% yield. Compound 108 was tetraethylated in a

Grignard reaction using ethylmagnesium iodide to form 5-methyl-N-benzyl-

1,1,3,3-tetraethylisoindoline (109) in 32% yield. The 2-benzyl group of 109 was

111

cleaved through hydrogenation of the protected isoindoline in a Parr-apparatus

using 50 psi H2 gas pressure to generate 5-methyl-1,1,3,3-tetraethylisoindoline

amine (110) in 92% yield. The debenzylated amine compound 110 was then

oxidized304 with 30% H2O2 in the presence of a Na2WO4.2H2O catalyst to form

the desired nitroxide 5-methyl-1, 1, 3, 3-tetraethylisoindoline-2-yloxyl (111) in

47% yield. Compound 111 was subsequently reduced with hydrogen using a

Pd/C catalyst and then acylated305 with acetyl chloride to form 2-acetoxy-5-

methyl-1,1,3,3-tetraethylisoindoline (112) in 99% yield.102 The methyl group of

compound 112 was then oxidized using 0.4 M solution of potassium

permanganate in the presence of magnesium sulfate306 at 70 °C to generate the

desired carboxylic acid 2-acetoxy-5-carboxy-1,1,3,3-tetraethylisoindoline (113)

in 78% yield.102 Compound 113 was finally hydrolysed by lithium hydroxide and

reoxidised by lead oxide to generate the desired nitroxide 5-carboxy-1,1,3,3-

tetraethylisoindoline-2-yloxyl (114) in 89% yield (Scheme 2.29). The

characterization of all compounds in this scheme is in agreement with the

published data.102

2.6.2 Coupling carboxy TEIO adenosine compound with

aminoethylaniline linker (170)

The compound 159 was coupled with a 5-carboxy-1,1,3,3-tetraethylisoindoline-

2-yloxyl (CTEIO) (114) using the EDCI/HOBT as the amide coupling reagent to

give the novel compound 2-(2-(4-N-(5-carboxy-1,1,3,3-tetraethylisoindoline-2-

yloxyl)aminophenyl))ethylamino-2,3-O-isopropylideneadenosine-5-N-

ethylcarboxamide (169) in 70% yield. (Scheme 2.30)

112

Scheme 2.30: Synthesis of CTEIO-substituted adenosine analogue possessing a substituted aniline linker moiety (170). Reagents and conditions: (i) EDCI/HOBt, DMF, DIPEA 25 °C; (ii) 1 M HCl solution, MeCN,

55–60 °C, 5 h.

The reaction was carried out in freshly distilled DMF under an argon

atmosphere at room temperature. The nitroxide was added after the coupling

reagents that led to completion of the reaction in 24 h instead of 3 days and

also produced a better yield of 79%. Addition of EDCI and HOBt first in DMF in

an argon atmosphere followed by carboxy nitroxide 114 and DIPEA, and later

portion-wise addition of the linker amine led to completion of the reaction at

room temperature (as identified on TLC). The work-up involved filtration of the

coupled solid product formed at chilling temperature after 30 min of reaction

quenching and washing many times with chilled water.

113

The reaction of 114 with 159 produced two products (as identified by TLC),

which were isolated by MPLC. HRMS of one isolated product was consistent

with the expected results for 169, which showed the expected molecular ion of

M+H at 755.4118 m/z, (EI+ HRMS showed a deviation of 0.0001 ppm from the

calc. mass of C40H53N9O6). The purity was found to be 95% by HPLC in a 75%

methanol/25% water system. The second component isolated was found to be

starting material. In the1H NMR spectroscopic analysis of compound 169,

signals of the aniline linker and the adenosine moieties could be detected.

Deprotection of the isopropylidene group from compound 169 was achieved

using 1 M HCl at elevated temperatures. The reaction went to completion in 5.0

h at 50–55 °C, to give the novel target compound 2-(2-(4-N-(5-carboxy-1,1,3,3-

tetraethylisoindoline-2-yloxyl) aminophenyl)) ethylaminoadenosine-5-N-

ethylcarboxamide (170) as a pale yellow solid compound (Scheme 2.30). The

product was filtered after quenching of the reaction using saturated sodium

bicarbonate solution at 0 °C and washed with plenty of chilled water.

The reaction gave a single reaction product 170 (as identified on TLC), which

was isolated using MPLC and partial crystallisation using 75% methanol/25%

water system, to give product in 71% yields. The results of the HRMS of the

isolated compound were consistent with the expected results for 170, which

showed the expected molecular ion of M+Na at 737.3506 m/z, (EI+ HRMS

showed a deviation of 0.0021 ppm from the calc. mass of C37H48NaN9O6). The

purity of product 170 was found to be 98.5% using analytical HPLC in a 75%

114

methanol/25% water mobile-phase system. This compound was analysed by

1H NMR spectroscopy, which indicated paramagnetic broadening by the

nitroxide radical resulting in some peaks not being detected or giving poor

integration, however the signals of the aniline linker and adenosine moieties

and the disappearance of the two acetal methyl signals at 1.17 and 1.19 ppm

could be detected.

2.7 Synthesis of C2-substituted di-tert-butylhydroxycinnamic acid adenosine analogues possessing substituent attached to ‘aminoethylaniline linker’ moiety (172).

Phenolic compounds also exhibit antioxidant properties similar to those of

nitroxides. In particular, hindered phenolic compounds, including BHT with

adenosine molecule demonstrated excellent results in the biological analysis.

Similarly, the phenolic compound hydroxycinnamic acid is also a potent

antioxidant. Additionally, this may deliver an excellent antioxidant activity and

A2AAR selectivity results when attached to the adenosine molecule.

It was speculated that the nitroxide compounds were degrading through out the

reaction conditions and having low yield using first approach synthesis,

therefore to confirm this behavoiur the novel hydroxy cinnamic acid based

adenosine compound 171 was synthesised using two different synthetic

approaches in a manner similar to synthesise compound 161. Firstly coupling

of the antioxidant moiety 118 with the linker amine is performed to form

compound 133 and then 133 is coupled to the adenosine intermediate 146.

115

2.7.1 Synthesis of di-tert-butylhydroxycinnamic acid possessing

substituent attached by ‘aminoethylaniline linker’ intermediate (133)

The novel compound N-tert-butoxycarbonyl-2(4-N-(3,5-ditert-butyl-4-

hydroxycinnamic acid)aminophenyl)ethylamine (132) was synthesized from

compound 125 by amide coupling with 3,5-ditert-butyl-4-hydroxycinnamic acid

(118) using EDCI in the presence of a catalytic amount of HOBt and DIPEA

along with using DMF as the basic solvent in 99% yield (Scheme 2.31).

Scheme 2.31: Overview of synthesis of di-tert-butyl-4-Hydroxycinnamic acid coupled aminophenylethylamine linker moiety (133). Reagents and conditions: (i) EDCI/HOBt, DMF, DIPEA 25 °C,

99%; (ii) 1 M HCl solution, MeCN, 55–60 °C, 5 h., 65% .

Compound 132 was synthesized by following the same amide coupling

procedure as that used to generate the nitroxide linker compounds. EDCI was

used as an acid activating agent, forming a reactive ester intermediate in situ

with the carboxylic acid of the hydroxy cinnamic acid antioxidant moiety. This

carboxylic acid ester has an activated leaving group that is attacked by the

116

nucleophilic amine group from the linker aniline. A thermodynamically stable

urea is released to give the novel desired amide product 132 with the amide

coupling mechanism explained in Scheme 2.7. HOBt was used to drive the

reaction in the desired direction, which limits side reactions and improves yields.

The reaction was run overnight at room temperature. A workup procedure

involving pouring the reaction solution into excess water and extracting the

resulting precipitate with DCM and washing several times with water afforded

high yields. The compound 132 was purified by column chromatography using

a mixture of CHCl3: MeOH: 28% aqueous ammonia solution (7.9:2:0.1), to give

product 132 in 99% yields on a 50–100 mg scale. Compound 132 appeared on

a TLC plate as a single spot. In the 1H NMR (CDCl3) spectrum of 132, a

multiplet appeared at 1.4 ppm with an integral corresponding to the eighteen

protons from the two tert-butyl groups and two triplet appeared at 2.73 and 3.36

ppm for the four protons of the ethyl linker group from the new aminoethylaniline

substituent. An extra two peaks corresponding to the carbons of the ethene

group also appeared in the 13C NMR spectrum at 117.3 ppm and 136.3 ppm,

respectively. The melting point of 126–128 °C of the isolated compound 132

was consistent and sharp. The structure of 132 was supported by MS analysis,

which showed the expected molecular ion of M+Na at 517.3006 m/z (EI+

(HRMS) showed a deviation of 0.0034 from the calc. mass of C30H42NaN2O4).

117

The novel target compound primary amine (E)-N-(4-(2-aminoethyl)phenyl)-3-

(3,5-di-tert-butyl-4-hydroxyphenyl)acrylamide (133) was obtained in 65% yield

from the Boc-protected amine 132 (Scheme 2.31). Removal of the acid-labile

Boc-protecting groups of amine 132 can be effected by reaction with 4 M HCl.

By following the same procedure as that used for the deprotection of 169, the

desired target 133 was obtained from the protected precursor 132. The

hydroxycinnamic acid containing linker compound 132 was stirred at 55–60 °C

for 2–3 h in a mixture of 1,4-dioxane and water with dropwise addition of 4 M

HCl over ten minutes. Analysis by TLC indicated consumption of starting

material, and ninhydrin staining highlighted the reformed primary amine 133 on

the TLC plate with a strong purple colour with an Rf of 0.13 in 9:1

chloroform/methanol. The compound was purified by column chromatography

to give 65% yield of the deprotected primary amine product 133 with a sharp

melting point of 130–132 °C.

In the 1H NMR (CDCl3) spectrum of 133, a multiplet appeared at 1.43 ppm with

an integral corresponding to eighteen protons from two tert-butyl groups and a

broad singlet appeared at 2.95 ppm for the two primary amine protons. An

extra two peaks corresponding to the carbons of the ethene group also

appeared in the 13C NMR spectrum at 125.0 ppm and 136.6 ppm, respectively.

The structure of 133 was supported by MS, which showed the expected

molecular ion of M+H at 395.2651 m/z (EI+ (HRMS) showed a deviation of

0.0048 from the calc. mass of C25H35N2O2).

118

2.7.2 Synthesis of C2-substituted di-tert-butylhydroxycinnamic acid

adenosine analogue possessing substituent attached by

‘aminoethylaniline linker’ moiety (171) (Approach 1)

Coupling of the antioxidant linker with the functionalized adenosine was the next

step in the process towards target compound 2-(2-(4-N-(3,5-di-tert-butyl-4-

hydroxyphenyl)acrylamido)aminophenyl))ethylamino-2,3,-O-

isopropylideneadenosine-5-N-ethylcarboxamide (171). In this work, a novel

C2-substituted adenosine analogue 171 was synthesised through the reaction

of 2-fluoro adenosine intermediate (146) with the isoindoline nitroxide

substituted aminoethylaniline linker moiety (133) (Scheme 2.32) by following the

Olsson synthetic procedure and as per mechanism explained in Scheme 2.20.

Scheme 2.32: Overview of coupling of di-tert-butylhydroxycinnamic acid with linker to adenosine (171)

(approach 1) Reagents and conditions: (i) DIPEA 75-80 °C, 7 days, 51%.

Synthesis of compound 171 was achieved by a coupling reaction in ethanol

using Hunig’s base DIPEA at 75–80 °C for 7 days. The reaction of 146 and 133

gives a single product (as identified on TLC). The compound was purified by

119

column chromatography using ethyl acetate and ammonia solvent system in

50% yield.

The structure of the hydroxycinnamic acid-coupled adenosine compound 171

was supported by MS, which showed the expected molecular ion of M+H at

741.4490 m/z by the HRMS (EI+ HRMS showed a deviation of 0.040 ppm from

the calc. mass of C40H53N8O6). The consistent sharp melting point of 138–140

°C supported the synthesis of the isolated compound 171.

Similarly, to the nitroxide possessing adenosine analogues the hydroxycinnamic

acid coupling reaction with approach 1 only gives moderate yields (50%). The

reaction conditions were very harsh. As this approach required harsh reaction

conditions and extended reaction times (7 days) an alternative approach

(approach 2) was explored. As the amine linkers could be sourced in large

scale, greater efficiency for the synthesis was thought to be possible through

first coupling the adenosine intermediate 146 with excess amine 124 and then

coupling this system to the hydroxycinnamic acid 118. Moreover, the number of

steps are also reduced by using adenosine intermediate 159 as the starting

material.

120

2.7.3 Synthesis of C2-substituted di-tert-butylhydroxycinnamic acid

adenosine analogues possessing substituent attached by

‘aminoethylaniline linker’ moiety (172) (Approach 2):

Compound 159 was coupled with 3, 5-di tert-butyl-4-hydroxycinnamic acid (118)

using EDCI/HOBt amide coupling reagent and DIPEA in dry DMF to give a

novel compound 2-(2-(4-N-(3,5-di-tert-butyl-4-

hydroxyphenyl)acrylamido)aminophenyl))ethylamino-2,3,-O-

isopropylideneadenosine-5-N-ethylcarboxamide (171) in 85% yield (Scheme

2.33).

Scheme 2.33: Synthesis of di-tert-butylhydroxycinnamic acid adenosine analogue possessing a substituted aniline linker moiety (172) (Approach 2). Reagents and conditions: (i) EDCI/HOBt, DMF, DIPEA

25 °C; (ii) 1 M HCl solution, MeCN, 55–60 °C, 5 h.

The reaction of 159 with 118 gave a single reaction product (as identified by

TLC), which was purified by using column chromatography to give 171 in 85%

121

yield. In the 1H NMR (CD3OD) spectrum, of the product 171, two alkene

protons appeared at 7.27 and 6.65 ppm as doublets and the phenolic hydroxy

proton appeared as a broad singlet at 5.57 ppm. Two multiplets at 2.77 ppm

with an integral corresponding to two protons and a triplet appeared at 0.59

ppm with an integral corresponding to three ethyl group protons from the ribose

amide moiety. An extra two peaks corresponding to the carbon of the ethene

group also appeared in the 13C NMR spectrum at 117 ppm and 138 ppm,

respectively.

The structure of 171 was supported by high-resolution mass spectrometry,

which showed the expected molecular ion of M+H at 741.4490 m/z (EI+ HRMS

showed a deviation of 0.040 ppm from the calc. mass of C40H53N8O6). The

melting point of 138–140 °C of the isolated compound 171 was sharp. The

purity of compound 171 was found to be >99.9% by analytical high-pressure

liquid chromatography (HPLC).

Deprotection of the isopropylidene group from compound 171 was achieved

using 1 M HCl at at 55–60 °C. in 4.0 h to give 2-(2-(4-N-(3,5-di-tert-butyl-4-

hydroxyphenyl)acrylamido)phenyl))ethylamino)adenosine-5-N-

ethylcarboxamide (172) in 70% yield (Scheme 2.33).

The deprotection reaction gave a single reaction product 172 (as identified on

TLC), which was isolated and purified using column chromatography and partial

122

crystallisation with 75% methanol/ 25% water system. In the 1H NMR

(CD3COCD3) spectrum of 172, a doublet of two alkene protons appeared at

7.24 and 5.92 ppm and the phenolic hydroxy proton appeared as a broad

singlet at 5.59 ppm. Two multiplets at 3.07 and 3.36 ppm with an integral

corresponding to two protons and a triplet appeared at 1.03 ppm with an

integral corresponding to three protons for the ethyl group from the ribose

amide substituent. An extra two peaks corresponding to the carbon of the ethyl

group linker also appeared in the 13C NMR spectrum (CD3OD) at 34.1 ppm and

42.5 ppm, respectively and two ethene group carbons appears at 117.2 and

142.6 ppm. The structure of the deprotected product 172 was supported by

MS, which showed the expected molecular ion of M+H at 701.4096 m/z, (EI+

HRMS showed a deviation of 0.032 ppm from the calc. mass of C37H49N8O6).

The melting point of 241–243 °C (decomposed) of isolated compound 172 was

sharp. The purity of compound 172 was found to be >99.9% using analytical

HPLC.

All the novel C2-substituted para-aminoethylaniline linked adenosine analogues

joined with antioxidant moieties were found to be difficult to purify and isolate by

column chromatography. The main products generated have close Rf values

similar to starting materials and a byproduct. The use of medium pressure

chromatography (MPLC) was found to be a better technique to purify those

components.

123

2.8 Result of biological testing of adenosine analogues

2.8.1 Adenosine receptor-binding assay

A number of C2-modified nitroxide adenosine compounds with various linker

groups were successfully synthesised. The potency and agonist activity (rather

than antagonists or inverse agonists) of each adenosine analogues was

assessed with a functional cAMP assay using transfected Chinese Hamster

Ovary (CHO) cell lines, in which cells overexpress one of the relevant human

A1, A2A, A2B, and A3 adenosine receptor subtypes. This analysis was undertaken

at the Monash Pharmaceutical Institute, Melbourne.

These functional assays provide an indication of the physiologically-relevant

mode of interaction between test compounds and the target receptor

subtypes307 and determination of the agonist activity of the compound in this

case at human adenosine receptor subtypes (hAXAR, X=1, 2A, 3). Functional

assays that measure a cAMP production were used to assess the (A1, A2A, A2B,

and A3) receptor binding affinity for each of the novel compounds.

This assay is based on a competition between cAMP attached to special

fluorescence material present in the test well with the test cells, and the

stimulation or inhibition of intracellular cAMP production by receptor agonism.

The generation of fluorescent material is relative to the total amount of cAMP at

the end of the experiment, and is measured by photo-stimulating the fluorescent

material and measuring the degree of fluorescence.

124

The potency for the A2AAR, and A2BAR is measured by pEC50, which is the

logarithmic value of concentration at which exactly half of the receptor

population is in a fully-activated state (i.e. the concentration of an agonists that

produces a 50% of physiological response). The A1AR and A3AR data is

reported as a pIC50 value, which is also a measure of potency, but more

commonly used for antagonists (this is because A1AR and A3AR act

biochemically in opposition to the A2AAR) and therefore the assay reports the

inverse of the pEC50 for the A1AR and A3AR.121 In this case, both assays are

equivalent measures of relative agonist activity at the adenosine receptor

subtype in question, and are expressed here in nanomoles (nM).

Next, the activity of a compound can be compared to that of other compounds

for each of the adenosine receptor subtypes assayed. The values are an

indication of relative functional selectivity and are expressed as a unitless ratio.

Functional selectivity ratios are calculated simply by dividing the pEC50 of the

compound at A2AAR by the pIC50 of the compound at the A1AR (or A3AR)

subtype. The value is an indication of how much more functionally active the

compound is at A2AAR than the other receptor subtypes (i.e. a value of A2A/ A1

of 100 means the compound is 100 times more potent in terms of functional

selectivity at A2AAR than A1AR). Desirable figures of this measure tend towards

more than a thousand fold in favour of the target receptor subtype relative to all

other receptor subtypes. The assays were conducted upon all three structural

classes of compounds at A1AR, A2AAR, A2BAR and A3AR receptor subtypes,

and the results are presented in Table 1.

125

cAMP Accumulation Assay. Adenosine is found naturally in nanomolar

concentrations in human interstitial fluid, and this level is essential for any

potential drug acting at the adenosine receptor. However, in this work,

compounds that exhibited nanomolar pEC50 and pIC50 values were described

here, which could act as desirable targets for further development as

cardioprotective agents.

The receptor binding results generated by the adenosine compounds from this

structural class are listed in Table 1.

126

Table 2.1: cAMP analysis of aminoethylaniline linked adenosine compounds (pEC50 and pIC50) in nM.

No. Compound Structure pIC50 - A1R pEC50 -

A2AR

pEC50 -

A2BR pIC50 - A3R A2A/A1 A2A/A3 A2A/A2B

162

5.2 ± 0.3 8.1 ± 0.2 7.4 ± 0.3 7.3 ± 0.2 794 6.3 5

164

5.7 ± 0.2 8.1 ± 0.1 7.8 ± 0.3 7.0 ± 0.2 251 13 2

166

5.5 ± 0.2 8.7 ± 0.3 7.7± 0.2 6.9 ± 0.3 1585 63 10

168

5.5 ±0.3 8.0 ± 0.2 6.4 ± 0.5 7.1 ± 0.1 316 8 40

170

N.D.(< 5) 8.2 ± 0.1 7.7 ± 0.1 7.7 ± 0.3 <1585 3 3

127

The dual action of adenosine agonist compounds possessing effective radical

scavenger nitroxide moieties and the C2-binding domain of A2AAR selective

compounds has not been explored to date. To investigate such selectivity, a

number of C2-modified nitroxide adenosine compounds were synthesised with

various linker groups. The potency and agonist activity (rather than antagonists

or inverse agonists) of the adenosine analogues was assessed with a this type

of downstream cAMP assay using transfected CHO lines. These cells

overexpress one of the human A1, A2A, A2B, and A3 adenosine receptor

subtypes. In general the C2-substituted nitroxides with 4-(2-aminoethyl)aniline

linked N6-aminoadenosine-5-N-ethylcarboxamide compounds (162-172) have

shown a good A2AAR affinity.

It is well known that the C2-binding domain with particular linkers can

accommodate a wide range of aryl and cycloalkyl groups. The incorporation of

very bulky tetramethylisoindoline-2-yloxyl, tetraethylisoindoline-2-yloxyl,

tetramethylpiperidine-1-yloxyl, and tetramethylpyrrolidine-1-yloxyl nitroxide

groups in this position afforded agonists with excellent affinity for the A2AAR. In

order to improve receptor affinity, the antioxidant nitroxide moiety was attached

via an aminoethylaniline linker. Linkers of this type have previously proven to

be effective in connecting adenosine receptor agonists with antioxidant moieties

(compounds 48 and 49).121

128

In some cases, large selectivity increases were seen with C2-substituted

nitroxides possessing (4-aminophenyl) ethylaminoadenosine-5-N-

ethylcarboxamide (namely compounds 162, 164, 166, 168 and 170) showing

more than 200-fold selectivity for the A2AAR over A1AR and reasonable

selectivity over A3AR.

All adenosine compounds in the first structural class with aminoethylaniline

linked series are potent A2AAR agonists with pEC50 values ranging from 8.0-8.7.

Compound 166 is marginally the most potent adenosine analogue with pEC50

value of 8.7. These compounds also showed good selectivity versus A1AR (251

to 1585) and modest selectivity versus A3AR (up to 63 fold). Similarly, these

compounds also have modest selectivity versus A2BAR (2 to 40 fold).

In a comparison of structurally related antioxidant groups in the first structural

class with aminoethylaniline linked series series, structural modification has

limited effect on the A2AAR selectivity. For example the tetramethylisoindoline

nitroxide 162 has a pEC50 of 8.1 at the A2AAR. The incorporation of a

methylene spacer in compound 168 has no (statistically significant) effect on

A2AAR activity (168; pEC50 of 8.0 ± 0.2). Likewise, tetraethyl substitution had no

statistical effect on A2A potency (cf. 162 v 170 with pEC50 values of 8.1 ± 0.2

and 8.2 ± 0.1, respectively). The above facts support the concept that the

antioxidant group is protruding into the extracellular space when the agonist is

bound to the A2AAR.

129

2.8.2 Simulated Ischemia Assay121.

A number of potent and selective C2-modified aminoethylaniline linked nitroxide

adenosine agonists were successfully assessed with this type of simulated

ischaemia assay. This assay is one measure of investigating cardioprotective

action of agonists. This ischaemia assay was conducted using a previously

reported cell culture hypoxia model to determine cell protection in the ischemic

induced rat arterial cardiomyoblast cell line H9c2.258, 308-310 This cell line cells

are incubating in hypoxic simulated ischaemia. This analysis was undertaken at

the Monash Pharmaceutical Institute, Melbourne.

The assay was conducted using the most potent and selective dual functionality

adenosine compounds 162, 164, 166, and 170 along with known compound

VCP874 (49) for further analysis. All compounds possess antioxidant moieties

combined with aminoethylaniline linked adenosine analogues. These

antioxidants possessing aminoethylaniline linked adenosine compounds have

good A2AAR selectivity (with pEC50 values of 8.0-8.7 nM) and have

demonstrated positive control in the study of ischemic cell protection as shown

in Figure 2.3 (panel A).

Specifically, the adenosine compounds 162 and 170 were selected for further

examination in an ischemic cell culture hypoxia assay. These compounds were

analysed by combining with a known A2AAR antagonist SCH442415, to observe

the effect of the antioxidants. This study shows that these dual acting ligands

gave significant cardioprotective effects, however, these effects were largely

130

inhibited by A2AAR antagonist SCH442415. This means that these effects were

primarily mediated by A2AAR activation and a limited effect by the nitroxide

antioxidant species was evident. There were some additional cardioprotective

effects observed at the highest concentration in the presence of antagonist.

These results may indicate that the antioxidant activity makes some contribution

in cardioprotection at that point of high concentration as shown in Figure 2.3

(panel B).

A B

Figure 2.3: Dual acting A2AAR agonists - antioxidants decrease cell death within an in vitro model of simulated ischemia. A) 162 (), 164 (), 166 () and 170 () stimulate an equivalent, concentration-

dependent, decrease in cell death to the reference ligand VCP874 (). B) The decrease in cell death mediated by 162 () and 170 () was inhibited in the presence of the A2AAR antagonist, SCH442415 ( 162 + 100 nM SCH442415; 170 + 100 nM SCH442415). Cell death was assessed by determining the

proportion of non-viable, propidium iodide positive, cells; N=2-4.

The smaller ring nitroxide adenosine compounds linked with the

aminoethylaniline linker group were found to be more potent and selective to

the adenosine receptors than other larger nitroxides containing adenosine

analogues. Furthermore, tetraethyl modified nitroxide adenosine compounds

also have better adenosine agonist activity. However, these compounds have

no major differences in the selectivities to the particular adenosine receptors. In

addition, side chains do not limit binding and are not detrimental to bioassays.

-8 -7 -6 -5SI

60

80

100

120

[Agonist] Log M

Ce

ll D

ea

th (%

of S

I)

VCP874

1VHM157C

1VHM168C2

1VHM170B

1VHM187

-8 -7 -6 -5SI

60

80

100

120

[Agonist] Log M

Ce

ll D

ea

th (%

of S

I)

1VHM157C

1VHM157C + 100 nM SCH442416

1VHM187

1VHM187 + 100 nM SCH442416

131

This means that even small or large group modifications are also tolerated in

the adenosine receptor selectivity; therefore, a small spacer linker group such

as alkynyl-linked nitroxide possessing adenosine compounds were synthesised.

In addition, it has been previously reported that the alkyne containing

compounds at adenosine C2-position are selective to the A2AAR. In order to

expand the possible side chain variations all of which still possess a nitroxide or

phenolic antioxidant, a further target of ethynyl-linked extended groups were

developed.

132

Result and Discussion Part B: Adenosine 3analogues with ethynyl linker possessing

antioxidant moieties.

133

Compounds having groups linked by an alkyne joined at the C2-position of the

adenosine are reported to be selective adenosine receptor agonists. A series of

2-alkynyl-5-N-ethylcarboxamide compounds were previously synthesised.311

These compounds were studied for the binding and functional assays to assess

their potencies for the A2AR compared to the A1AR. These compounds are

known to have desirable effects on vasorelaxation without impacting on heart

rate. C2-alkynyl-5-N-ethylcarboxamide compounds were found to be selective

A2AR agonists243 (Figure 3.1). Moreover, some of these alkynyl-linked

compounds were also found to possess good A1AR/A2AR affinity. Some of the

hydroxy alkynyl adenosine derivatives are potent inhibitors of platelet

aggregation and could be beneficial in the treatment of cardiovascular

disorders.311

Figure 3.1: Candidate moieties and their actions in the second structural class of target adenosine analogues.

The alkynyl linker was intended to promote specific receptor recognition.

Binding studies312 have shown that the presence of a phenylethynyl group in the

134

C2-position of adenosine 65 (Figure 3.2) favoured the interaction with A3AR,

with the resulting compounds displaying high affinity and selectivity for the

adenosine A3AR. Additional substitution at the N6- and C4-positions interact

with the hydrophilic pocket on the adenosine receptor to increase both the A3AR

agonists affinity and also the H-bonding at the 3- and 5-positions which are

required for A3AR agonism.313-315

Figure 3.2: Adenosine A3AR agonists (PENECA) 65

The initial adenosine synthetic targets of this work can be synthesised from the

adenosine intermediate 2-iodo-2,3-O-isopropylideneadenosine-5-N-

ethylcarboxamide (149) by coupling with the appropriate alkyne isoindoline

nitroxide (120, and 122) or non-nitroxide moiety. The published methodology

for achieving these targets involves the Sonogashira coupling reaction.

3.1 Synthesis of C2-substituted TMIO adenosine analogues possessing substituents attached by an ‘ethynyl linker’moiety (178)

3.1.1 Synthesis of the 2-iodo-2,3-O-isopropylideneadenosine-5-N-

ethylcarboxamide intermediate (149)

Functional group interconversion of the C2-amine of O6-(benzotriazol-1-yl)-2,3-

O-isopropylideneadenosine-5-N-ethylcarboxamide (144) to the nucleophilic

135

aromatic substitution-promoting iodine was accomplished using the method of

Kim273 (Scheme 3.1) The reaction proceeded smoothly achieving 2-iodo-O6-

(benzotriazol-1-yl)-2,3-O-isopropylideneadenosine-5-N-ethylcarboxamide

(148) in 86% yield on a scale ranging from 50-200 mg.

Scheme 3.1: Overview of synthesis of C2-iodo adenosine intermediate (149) Reagents and conditions: (i)

CH2I2, t-BuONO, in MeCN, 68 %. (ii) 28% aq. ammonia MeCN.

The reaction conditions were reasonably harsh because diiodomethane was

used as an iodination agent and MeCN as the solvent, and the reaction was

carried out at a temperature of 65–70 °C. The solvent and excess of

diiodomethane was removed by evaporating the reaction mixture under vacuum

for a long time. The impurity was successfully removed by excessive washing

with ethyl acetate. The Rf value for the product 148 was 0.42 in 1:4

DCM/MeOH and was distinguishable from 144 by TLC, (Rf. 0.37 for the C2-

amino compound 144). These values indicates that the polarity of the molecule

slightly decreased with the introduction of the electronegative iodine atom and

the removal of the amine.

136

In the 1H NMR spectrum (d6-DMSO), the broad singlet at 6.67 ppm

corresponding to the heteroaromatic amino at C2-position of compound 144

was not present. The C8-proton appeared to be a sharp singlet. This singlet

was shifted to the more deshielded region from 8.04 to the 8.19 ppm for the

iodo substituted adenosine compound 148. In the 13C NMR spectrum, a peak

for the C2-carbon of the purine ring appeared at 144.0 ppm. Additionally, there

was a signal at 168 ppm which was assigned to the amide carbonyl carbon.

The melting point of 194–196 °C for the product 148 was sharp. The structure

of deprotected product 148 was supported by HRMS, which showed the

expected molecular ion of M+Na at 615.0808 m/z, (EI+ HRMS showed a

deviation of 0.0371 ppm from the calc. mass of C21H21INaN8O5)

Ammonia solution was used to displace the more labile benzotriazolyl leaving

group from compound 148 with nucleophilic amines262, 282 and form a 6-amino

product 2-iodo-2,3-O-isopropylideneadenosine-5-N-ethylcarboxamide (149)

(Scheme 3.1). This SNAr reaction proceeded smoothly in acetonitrile at room

temperature overnight. Purification via column chromatography gave good

yields between 70–80% of an off-white solid on a 250 mg to 1 g scale. A

second component separated by column chromatography was identified as a

small amount of amino-substituted product at the C2-position.

As expected, the Rf value of 149 on TLC decreased as the benzotriazolyl group

was lost and the polarity increased slightly. The Rf value was measured at 0.62

using 7:3 ethyl acetate/MeOH. In d6-DMSO, the major features of the 1H NMR

137

spectrum of 149 were the absence of signals from the benzotriazolyl aromatic

protons and the broad singlet signal which appeared at 7.85 ppm with an

integral corresponding to two N6-protons. Signals from the 2- and 3-protons

coalesced completely from two resolved doublets to a singlet peak at 5.35 ppm

with an integral corresponding to two protons. Similarly, in the 13C NMR

spectrum, signals from the benzotriazolyl aromatic carbons disappeared.

Furthermore, the signals for the 2- and 3-carbons coalesced to give two

identifiable signals separated by 0.03 ppm. The melting point of 196–198 °C of

the product 149 was sharp. The structure of the product 149 was supported by

MS, which showed the expected molecular ion of M+H at 497.0390 m/z, (EI+

HRMS showed a deviation of 0.0030 ppm from the calc. mass of

C15H19INaN6O4). The purity of product 149 was found to be 99.99% by

analytical HPLC in a 80% methanol/20% water system.

A number of adenosine molecules were generated with either tetramethyl and

tetraethyl nitroxide or a phenyl side chain joined by with an ethynyl linker by

modification of adenosine at the C2-position and with 5-N-ethylcarboxamide

modification of the ribose ring.

3.1.2 Synthesis of ethynyl TMIO (ETMIO) intermediate (120)

In this synthesis, firstly the nitroxide moiety 5-ethynyl-1,1,3,3-

tetramethylisoindoline-2-yloxyl (ETMIO) nitroxide 120 was synthesised from 5-

bromo-tetramethylisoindoline (102) according to the procedure reported in the

literature316 (Scheme 3.2).

138

Scheme 3.2: Synthesis of 5-ethynyl-1,1,3,3-tetraethylisoindoline-2-yloxyl (ETMIO) (120) 316

. Reagents and conditions: (i) I2, n-BuLi, H2O2, Stir 30 min, 69% (ii) Na2WO4.2H2O,H2O2,NaHCO3, Stir 72 h. 79%. (iii)

DABCO, Pd(OAc)2, 2-methyl-3-butyn-2-ol, MeCN, 80 %; (iv) KOH, Toluene, 90 %.

5-Bromo-1,1,3,3-tetramethylisoindoline (102) was reacted with n-BuLi in dry

THF and then quenched with iodine to obtain a di-iodo intermediate, in which

iodination occurred on the ring as well as on the nitrogen atom. This product

was subsequently treated with H2O2 to generate a mono-iodo compound 5-iodo-

1,1,3,3-tetramethylisoindoline (104) in 69% yield. Compound 104 was then

oxidised by 30% H2O2 in the presence of the Na2WO4.2H2O catalyst in MeCN to

give 5-iodo-1,1,3,3-tetramethylisoindoline-2-yloxyl (105) in 79% yield. The

compound 105 was then coupled with 2-methyl-3-butyne-2-ol (160) using

palladium diacetate (Pd(OAc)2) in the presence of 1,4-diazabicyclo(2.2.2)

octane (DABCO) as a base using triethylamine as a solvent to obtain 5-(2-

methyl-3-butyne-2-ol)-1,1,3,3-tetramethylisoindoline-2-yloxyl (119)316 in 80%

yield.

The reaction mechanism of alkyne coupling with the palladium reagent is

detailed in Scheme 3.3. Deprotection of the dimethylpropanol group from

compound 119 with solid KOH in refluxing MeCN resulted in the formation of E-

139

TMIO 120 in 90% yield (Scheme 3.2). The melting point of 96–98 °C of the

recrystallised product was consistent and in agreement with the published

values316 for the desired product 120.

3.1.3 Coupling of E-TMIO with iodo-adenosine intermediate (178)

The ethynyl isoindoline nitroxides and phenyl acetylene were then coupled in

separate reactions with the adenosine iodo intermediate under Sonogashira

coupling conditions to achieve coupling products 177, 181 and 185. The details

of the reaction mechanisms are explained as follows.

The palladium-catalysed alkynylation called the Sonogashira reaction is a

convenient method to synthesise aryl and other acetylene-substituted

compounds. This methodology was first discovered in 1975 by Cassar,317

Dieck, Heck318 and Sonogashira et al.319 The method used was an

amalgamation of the Castro–Stephens reaction320, 321, involving Cu-promoted

alkynylation and alkyne implementation of the Heck reaction. Sonogashira et

al. used Cu (I) in the presence of the Pd (0) catalyst, which favours the alkynyl

transformation at room temperature. The reaction was achieved under much

milder conditions and is now commonly referred to as the Sonogshira coupling

reaction.

140

Scheme 3.3: Sonogshira coupling reaction mechanism.

This coupling of terminal alkynes with aryl halides typically proceeds with two

catalysts, palladium (0) (Pd(PPh3)2Cl2) and copper (I) co-catalyst (CuI) using

triethylamine as base. This reaction requires anhydrous conditions; however

newer techniques have recently been developed, where these restrictions are

not required.322 Reactions can be carried out under mild conditions using

mixtures of solvents at room temperature. The aryl halide first forms a complex

with the palladium catalyst by oxidative addition. The presence of base results

in the formation of an alkyne π-complex, which makes the terminal carbon of

the alkyne more acidic and leads to the formation of a copper-acetylide

complex. Trans-metalation with palladium and the copper acetylide along with

leaving of the halide ion leads to the formation of an alkyne palladium complex,

141

which can undergo reductive elimination to give rise to the desired C–C

coupling product (Scheme 3.3).

Using this approach 5-iodo-2,3-O-isopropylideneadenosine-5-N-

ethylcarboxamide 149 was coupled with 5-ethynyl-1,1,3,3-

tetramethylisoindoline-2-yloxyl (E-TMIO) 120. This reaction was carried out

using Pd(PPh3)2Cl2 and copper iodide as the Sonogashira coupling reagent

along with a catalytic amount triethylamine in dry DMF. The reaction gave the

novel compound 2-(1,1,3,3-tetramethylisoindoline-2-yloxyl-5-ethynyl)-2,3-O-

isopropylideneadenosine-5-N-ethylcarboxamide (177) in 76% yield (Scheme

3.4) .

Scheme 3.4: Synthesis of ETMIO-substituted adenosine analogue (178) Reagents and conditions: (i) Pd

(PPh3)2Cl2, CuI, TEA in DCM/MeCN, 76%. (ii) 1 M HCl in MeCN 55-60 °C, 98%.

142

The reaction was carried out in a mixture of freshly distilled DCM and dry MeCN

under an argon atmosphere at room temperature. The reaction was followed by

TLC and went to completion in 20 h with a product Rf value of 0.64 in

chloroform: ammonia solution (9.8:0.2) as the mobile phase solvent system.

The reaction of 120 with 149 produced a single adduct 177 along with some

alkyne nitroxide starting material (<5%) (as identified by TLC). The product was

isolated and purified by column chromatography using ethyl acetate and

aqueous ammonia solution (9.8:0.2) as the mobile phase. In the 1H NMR

spectrum, paramagnetic broadening was observed as expected due to the

nitroxide radical. This broadening resulted in some peaks not being detected

and giving poor integration. However, the signal of the adenosine moieties

could be detected. The synthesis of product 177 was supported by HRMS,

which showed the expected molecular ion of M+H at 561.2682 m/z, (EI+ HRMS

showed a deviation of 0.0018 ppm from the calc. mass of C29H35N7O5). The

melting point of 146–148 °C of the isolated compound 177 was sharp. The

purity of product 177 was found to be 97.4% by analytical HPLC in an 80%

methanol/20% water system.

Deprotection of the isopropylidene group from compound 177 was achieved

using 1 M HCl at 55–60 °C in 8.0 h. The reaction gave the novel target

compound 2-(1,1,3,3-tetramethylisoindoline-2-yloxyl-5-ethynyl)-adenosine-5-N-

ethylcarboxamide 178 in 98% yield (Scheme 3.4).

143

The deprotection reaction gave a single product 178 (as identified on TLC),

which was isolated by column chromatography using (9.8:0.2) chloroform and

aqueous ammonia solution as the mobile phase. In the 1H NMR spectrum,

paramagnetic broadening was observed as expected due to the nitroxide

radical. This broadening resulted in some peaks not being detected and giving

poor integration. However, the signal of the adenosine moieties could be

detected. The synthesis of product 178 was supported by HRMS, which

showed the expected molecular ion of M+Na at 543.2281 m/z, (EI+ HRMS

showed a deviation of 0.0075 ppm from the calc. mass of C26H30NaN7O5). The

melting point of 261 °C (decomp) for the isolated compound 178 was sharp.

The purity of product 178 was found to be 99.9% using analytical HPLC in an

80% methanol/20% water system.

3.2 Synthesis of C2-substituted TEIO adenosine analogues possessing substituents attached by an ‘ethynyl linker’ moiety (182)

A novel compound 5-ethynyl-1,1,3,3-tetraethylisoindoline-2-yloxyl (E-TEIO) 122,

which was synthesized from compound 105 and obtained from advanced stage

synthesis in the laboratory. Compound 149 was coupled with compound 122

using Pd(PPh3)2Cl2 and copper iodide as the coupling reagents. The reaction is

employed a catalytic amount of triethylamine in dry DMF to produce a novel

compound 2-(1,1,3,3-tetraethylisoindoline-2-yloxyl-5-ethynyl)-2,3-O-

isopropylideneadenosine-5-N-ethylcarboxamide (181) in 83% yield (Scheme

3.5).

144

Scheme 3.5: Synthesis of ETEIO-substituted adenosine analogue (182) ,Reagents and conditions: (i) Pd

(PPh3)2Cl2, CuI, TEA in DCM/MeCN, 83%. (ii) 1 M HCl in MeCN 55-60 °C, 88%.

The reaction was carried out with a mixture of freshly distilled DCM and dry

MeCN under an argon atmosphere at room temperature. The reaction went to

completion in 20 h (as identified by TLC) with Rf value of product 0.47 in (DCM /

MeOH, 9:1).

The reaction of 122 with 149 produced a single product 181 along with some

alkynyl nitroxide starting material (<5%) (as identified by TLC). The product

was isolated and purified by column chromatography in 83% yield. In the 1H

NMR spectrum, paramagnetic broadening was observed as expected due to the

nitroxide radical. This broadening resulted in some peaks not being detected

and giving poor integration. However, the signal of the adenosine moieties

145

could be detected. The synthesis of product 181 was supported by HRMS,

which showed the expected molecular ion of M+H at 617.3296 m/z, (EI+ HRMS

showed a deviation of 0.0030 ppm from the calc. mass of C33H43N7O5). The

melting point of 188–190 °C of the isolated compound 181 was sharp. The

purity of the product 181 was found to be 97.4% using analytical HPLC in an

80% methanol/20% water system.

The deprotection of isopropylidene group from compound 181 was achieved

using 1 M HCl at 55–60 °C in 8.0 h. The reaction generated the novel target

compound 2-(1,1,3,3-tetraethylisoindoline-2-yloxyl-5-ethynyl)-adenosine-5-N-

ethylcarboxamide (182) (Scheme 3.5).

The deprotection reaction of 181 gave a single product 182 (as identified on

TLC), which was purified by column chromatography to give product 182 in 88%

yield. In the 1H NMR spectra, paramagnetic broadening was observed as

expected due to the nitroxide radical that resulted in some peaks not being

detected and giving poor integration. However, signals of the adenosine moiety

could be detected. The synthesis of product 182 was supported by HRMS,

which showed the expected molecular ion of M+Na at 577.3012 m/z, (EI+

HRMS showed a deviation of 0.0001 ppm from the calc. mass of C30H39N7O5).

The melting point of 196–198 °C (decomp) of the isolated compound 182 was

sharp. The purity of the product 182 was found to be 99.9% using analytical

HPLC in an 80% methanol/20% water system.

146

3.3 Synthesis of C2-substituted non-nitroxide phenyl adenosine analogues possessing substituents attached by an ‘ethynyl linker’ moiety (185)

The compound 149 was coupled with phenylacetylene (123) using Pd(PPh3)2Cl2

and copper iodide as the coupling reagent as well as a catalytic amount of

triethylamine in dry DMF. The coupling reaction produced the expected product

2-(2-phenylethynyl)-2,3-O-isopropylideneadenosine-5-N-ethylcarboxamide

(185) in 97% yield (Scheme 3.6).

Scheme 3.6: Synthesis of the phenylacetylene-substituted adenosine analogue (185). Reagents and

conditions: (i) Pd (PPh3)2Cl2, CuI, TEA in DCM/MeCN, 97%.

The reaction was carried out with a mixture of freshly distilled DCM and dry

MeCN under an argon atmosphere. The reaction went to completion in 15 h (as

identified by TLC) at room temperature.

The reaction of 149 with 123 produced a single adduct 185 along with some

detectable alkyne nitroxide starting material (< 5%) (as identified by TLC). The

product was isolated and purified by column chromatography using100%

chloroform. In the 1H NMR spectrum, a new peak appeared in the aromatic

region as a multiplet for three protons from the phenyl group at 7.35–7.40 ppm

along with one multiplet for two aromatic protons which appeared at 7.58 ppm.

147

In the 13C NMR spectrum, three additional signals appeared as strong peaks

corresponding to the aromatic phenyl carbons at 128.4, 129.5 and 132.3 ppm.

The other signals appeared at 84.9 and 114.9 ppm for the alkyne carbons. The

synthesis of product 185 was supported by HRMS, which showed the expected

molecular ion of M+H at 449.1881 m/z, (EI+ HRMS showed a deviation of

0.0057 ppm from the calc. mass of C23H25N6O4). The melting point of 174–176

°C of the isolated compound 185 was sharp. The purity of product 185 was

found to be 99.9% using analytical HPLC in a 80% methanol/20% water

system.

3.4 Results of biological testing of adenosine analogues

3.4.1 Adenosine receptor binding assays

A number of C2-modified nitroxide adenosine compounds were synthesised

with ethynyl linker groups. The potency and agonist activity (rather than

antagonists or inverse agonists) of the adenosine analogues were assessed

with this type of downstream cAMP assay. This test was implemented using

transfected CHO lines in which cells overexpress one of the human A1, A2A, A2B,

and A3 adenosine receptor subtypes. This assay was carried out by our

collaborators at the Monash University Melbourne.

148

Table 3.1: cAMP analysis of ethynyl linked adenosine compounds(pEC50 and pIC50) in nM

No. Compound Structure pIC50 -

A1R

pEC50 -

A2AR

pEC50 -

A2BR

pIC50 -

A3R A2A/A1 A2A/A3 A2A/A2B A2B/A1 A2B/A3 A2B/A2A

178

6.6 ± 0.2 6.2 ± 0.2 7.0 ± 0.4 6.5 ± 0.1 0.4 0.5 0.5 3 3 6

182

6.3± 0.2 5.6 ± 0.2 7.0 ± 0.7 N.D.(< 5) 0.2 4 0.5 5 100 25

149

The molecules from this series of adenosine linked nitroxide were found to have

low A2AAR selectivity with pEC50 values ranging from 5.6-6.2 nM. The alkyne

(ethynyl) linked tetramethylisoindoline-2-yloxyl nitroxide adenosine compounds

(178; pEC50 of 6.2 ± 0.2) and tetraethylisoindoline-2-yloxyl nitroxide adenosine

(182; pEC50 of 5.6 ± 0.2) were significantly less selective for A2AAR over the

other receptor subtypes.

However, this structural class of nitroxide linked with ethynyl moieties to

adenosine displayed a slightly higher selectivity for A2BAR with pEC50 values

(178; pEC50 of 7.0 ± 0.4) and (182; pEC50 of 7.0 ± 0.7) respectively. The above

results indicate that the nitroxide adenosine compounds with two carbon

(ethynyl) linker are better in A2BAR potency and selectivity than the non-

nitroxide adenosine compounds.

These analogues demonstrate slightly higher selectivity for the adenosine

A2BAR over the other adenosine receptor subtypes. Specifically, the adenosine

analogue 178 shows 6 fold better selectivity for A2BAR over A2AAR and 3 fold

better selectivity over other receptor subtypes. Adenosine analogue 182 shows

25 fold selectivity for adenosine A2BAR over A2AAR and 5 and 100 fold

selectivity over adenosine A1AR and A3AR subtypes respectively ( Table 3.1).

These ethynyl linked nitroxide adenosine compounds synthesised have no

significant impact on the affinity of the A2AAR. However, these modified

150

adenosine analogues were found to be selective for the A2BAR. This

modification shows the promising characteristics to develop the new A2BAR

selective compounds. It was postulated that the flexible chain linker might

increase the selectivity for the A2AAR agonists. To find out the effect of

flexibility on the adenosine receptor selectivity, it was decided to reduce the

ethynyl triple bond between the adenosine and antioxidant nitroxide moiety in

order to generate a new class of compounds.

151

Results and Discussion Part C: Adenosine 4analogues with ethyl linker possessing

antioxidant moieties

152

The adenosine synthetic targets of this work have been synthesised starting

from adenosine-5-N-ethylcarboxamide coupled with nitroxides and non-

nitroxide compounds joined by an ethynyl linker chain to generate nitroxides

and non-nitroxides possessing ethyl linked adenosine compounds. These

ethylene-linked adenosine targets possessing nitroxide antioxidant moieties

were expected to be highly selective towards the A2AR. This selectivity is

expected to arise based on previous reports showing that A2AR selectivity is

provided through the incorporation of 2 (or 3) atom chain spacers connected to

the adenosine C2-position. In this project, a number of nitroxide and non-

nitroxide compounds possessing a 5-amide group (the 5-NECA modification)

were synthesised by modification of the C2-position using a two methylene

carbon linkage, with no polar atoms attached. It was expected that this would

provide good A2AR specificity whilst allowing a range of structurally-diverse

antioxidant groups to be incorporated (Figure 4.1).

Figure 4.1: Candidate moieties and their actions in the third structural class of target adenosine analogues.

153

A number of adenosine molecules were generated with tetramethyl and

tetraethylisoindoline nitroxides as well as a phenyl group as the non-radical,

non-antioxidant analogue and each was joined to adenosine at the adenosine

C2-position by an ethyl linker were achieved by the reaction of the appropriate

alkyne at the adenosine C2-position. These compounds also possesse the 5-

N-ethylcarboxamide modification to enhance receptor subtype specificity.

In this approach, the alkynyl linked nitroxides and non-nitroxide adenosine

compounds were reacted further with palladium on carbon under hydrogen gas

at room temperature in methanol to reduce the triple bond and afford the

‘saturated-linker’ adenosine compounds.

4.1 Synthesis of C2-substituted TMIO adenosine analogues possessing substituents attached by an ‘ethyl linker’ moiety (180)

2-(1,1,3,3-Tetramethylisoindoline-2-yloxyl-5-ethynyl)-2,3-

isopropylideneadenosine-5-N-ethylcarboxamide (177) was reacted with

palladium on carbon under a hydrogen atmosphere at room temperature to

afford the novel reduced compound 2-(1,1,3,3-tetramethylisoindoline-2-yloxyl-5-

ethyl)-2,3-isopropylideneadenosine-5-N-ethylcarboxamide (179) in 84% yield

(Scheme 4.1).

154

Scheme 4.1: Overview the synthesis of the TMIO-nitroxide with ethyl linked adenosine compound (180),Reagents and conditions: (i) 10% Pd/C, H2, MeOH, 84%. (ii) PbO2 , MeOH.(iii)1 M HCl, MeCN, 55–60

°C, 79%.

The reaction went to completion in methanol in 12 h (as identified on TLC). The

Rf value of the product 179 was 0.023 in 1:4 DCM/MeOH (Rf. = 0.37 for the C2-

amino compound 177), indicating that the polarity of the molecule had

decreased markedly by reduction of the triple bond.

The reduction reaction of 177 produced a single product (as identified by TLC).

In this reaction, the nitroxide radical was also hydrogenated to form the N-

hydroxy compound. This hydroxy compound was not isolated or characterised

(Scheme 4.1). The crude mixture was subjected to reoxidation using lead oxide

(PbO2) in methanol to regenerate the nitroxide radical compound 179. The

product 179 was isolated and purified by column chromatography using a

mixture of CHCl3: MeOH (9:1) mobile phase. In the 1H NMR spectrum,

paramagnetic broadening was observed as expected due to the nitroxide

155

radical which resulted in some peaks not being detected and giving poor

integration, however the signal of the adenosine moieties could be detected.

The synthesis of product 179 was supported by HRMS, which showed the

expected molecular ion of M+H at 564.3276 m/z, (EI+ HRMS showed a

deviation of 0.033 ppm from the calc. mass of C29H38N7O5). The melting point

of 108–110 °C of the isolated compound 179 was sharp. The purity of the

product 179 was found to be 96.7% using analytical HPLC in an 80%

methanol/20% water system.

Deprotection of the isopropylidene group from compound 179 was achieved

using 1 M HCl at 55–60 °C in 8.0 h and generated the novel target compound 2-

(1,1,3,3-tetramethylisoindoline-2-yloxyl)-5-ethyl)-adenosine-5-N-

ethylcarboxamide (180) in 79% yield (Scheme 4.1).

The deprotection of compound 179 produced a single product 180 (as identified

by TLC). The Rf value of the product 180 was 0.16 in CHCl3/MeOH (8:2). The

product was isolated and purified by column chromatography using a mixture of

CHCl3: MeOH (9:1) mobile phase. In the 1H NMR spectra, paramagnetic

broadening was observed as expected due to the nitroxide radical which

resulted in some peaks not being detected and giving poor integration, however

the signal of the adenosine moieties could be detected. The synthesis of

product 180 was supported by HRMS, which showed the expected molecular

ion of M+H at 525.2833 m/z, (EI+ HRMS showed a deviation of 0.013 ppm from

the calc. mass of C26H35N7O5). The melting point of 122–124 °C of the isolated

156

compound 180 was sharp. The purity of product 180 was found to be 99.9%

using analytical HPLC in a 80% methanol/20% water system.

4.2 Synthesis of C2-substituted TEIO adenosine analogues possessing substituents attached by ‘ethyl linker’ moiety (184).

2-(2-(1,1,3,3-Tetraethylisoindoline-2-yloxyl)-5-ethynyl)-2,3-

isopropylideneadenosine-5-N-ethylcarboxamide (181) was reacted with

palladium on carbon under hydrogen atmosphere using a hydrogen gas balloon

at room temperature afford the novel reduced compound 2-(1,1,3,3-

tetraethylisoindoline-2-yloxyl-5-ethyl)-2,3-isopropylideneadenosine-5-N-

ethylcarboxamide (183) in 97% yield (Scheme 4.2).

Scheme 4.2: Overview synthesis of TEIO-nitroxide with ethyl linked adenosine compound (184) Reagents

and conditions: (i) 10% Pd/C, H2, MeOH, 97%. (ii) PbO2 , MeOH. (iii) 1 M HCl, MeCN, 55–60 °C, 70%.

157

The reaction was carried out in a mixture of methanol with 10% palladium on

carbon as the solvent under a hydrogen atmosphere at room temperature. The

reaction went to completion in 12 h (as identified on TLC). The Rf value of

product 183 was 0.023 in a mixture of DCM/MeOH (1:4), indicating that the

polarity of the molecule was decreased markedly by reduction of the triple bond.

The reduction of compound 181 produced a single component (as identified by

TLC). In this reaction, the nitroxide radical also hydrogenated to form N-

hydroxy compound. This hydroxy compound was not isolated or characterised

(Scheme 4.2). The crude mixture was subjected to reoxidation using lead oxide

(PbO2) in methanol to regenerate the nitroxide radical product 183. The product

183 was isolated and purified by column chromatography using DCM/MeOH

(9:1) mobile phase. In the 1H NMR spectrum, paramagnetic broadening was

observed as expected due to the nitroxide radical which resulted in some peaks

not being detected and giving poor integration, however the signal of the

adenosine moieties could be detected. The synthesis of product 183 was

supported by HRMS, which showed the expected molecular ion of M+H at

621.3630 m/z, (EI+ HRMS showed a deviation of 0.0009 ppm from the calc.

mass of C33H47N7O5). The melting point of 88–90 °C of the isolated compound

183 was sharp. The purity of product 183 was found to be 89% using analytical

HPLC in an 80% methanol/20% water system.

Deprotection of the isopropylidene group from compound 183 was achieved

using 1 M HCl at 55–60 °C in 8.0 h and generated a novel target compound 2-

158

(1,1,3,3-tetraethylisoindoline-2-yloxyl-5-ethyl)adenosine-5-N-ethylcarboxamide

(184) in 70 % yield (Scheme 4.2).

The deprotection of compound 183 produced a single product 184 (as identified

by TLC). The Rf value of the product 184 was 0.69 in DCM/MeOH (9:1). The

product was isolated and purified by column chromatography using (100%)

chloroform as mobile phase. In the 1H NMR spectra, paramagnetic broadening

was observed as expected due to the nitroxide radical which resulted in some

peaks not being detected and giving poor integration, however the signal of the

adenosine moieties could be detected. The synthesis of product 184 was

supported by HRMS, which showed the expected molecular ion of M+H at

581.3277 m/z, (EI+ HRMS showed a deviation of 0.0049 ppm from the calc.

mass of C30H43N7O5). The melting point of 138–140 °C of the isolated

compound 184 was consistent. The purity of product 184 was found to be

>99.9% using analytical HPLC in 80% methanol/20% water system.

4.3 Synthesis of C2-substituted non-nitroxide phenyl adenosine analogues possessing substituents attached by an ‘ethyl linker’ moiety (188).

2-(2-Phenylethynyl)-2,3-isopropylideneadenosine-5-N-ethylcarboxamide (185)

was reacted with palladium on carbon under hydrogen atmosphere at room

temperature to afford the novel reduced compound 2-(2-phenylethyl)-2,3-

isopropylideneadenosine-5-N-ethylcarboxamide (187) in 93% yield (Scheme

4.3).

159

Scheme 4.3: Overview of synthesis of the non-nitroxide phenyl ethyl linker to the adenosine compound (188) Reagents and conditions: (i) 10% Pd/C, H2, 93% MeOH. (ii) 1 M HCl, MeCN, 55–60 °C, 94%.

The reaction was carried out at room temperature in a mixture of methanol with

10% palladium on carbon under hydrogen atmosphere at room temperature.

The reaction went to completion in 12 h (as identified on TLC). The Rf value of

the product 187 was 0.023 in DCM/MeOH (9:1), indicating that the polarity of

the molecule was decreased markedly compared to starting material by

reduction of the triple bond.

The reduction reaction of 185 produced a single product 187 (as identified by

TLC), which was purified and isolated twice by column chromatography using

DCM/MeOH (9:1) mobile phase. In CDCl3, the major features of the 1H NMR

spectrum of the hydrogenated adenosine analogue 187 was the appearance at

3.05-3.11 ppm a multiplet integrating for four protons corresponding to the two

methylene groups, as well as the signal for the methyl group from the acetal

protection appeared at the 1.37 and 1.58 ppm. Likewise, in the 13C NMR

160

spectrum, two methylene carbon signals appeared at 33.7 and 40.5 ppm. The

synthesis of the product 187 was supported by HRMS, which showed the

expected molecular ion of M+H at 453.2243 m/z, (EI+ HRMS showed a

deviation of 0.0007 ppm from the calc. mass of C23H29N6O4). The melting point

of 70–72 °C of the isolated compound 187 was sharp. The purity of the product

187 was found to be 98% using analytical HPLC in an 80% methanol/20%

water system.

Deprotection of the isopropylidene group from compound 187 was achieved

using 1 M HCl at 55–60 °C in 8 h and generated the novel target compound 2-

(2-phenylethyl)adenosine-5-N-ethylcarboxamide (188) in 94% yield (Scheme

4.3). The deprotection of compound 187 produced a single product 188 (as

identified by TLC). The Rf value of the product 188 was 0.69 in DCM/MeOH

(1:4), The product was purified and isolated by column chromatography using

100% chloroform as the mobile phase. In CDCl3, the major features of the 1H

NMR spectrum of deprotected adenosine analogue 188 were the

disappearance of the methyl signals from the acetal protection at the 1.37 and

1.58 ppm. Likewise, in the 13C NMR spectrum, two methylene carbon signals

disappeared at 25.2 and 26.9 ppm. The synthesis of the product 188 was

supported by HRMS, which showed the expected molecular ion of M+H at

413.2019 m/z, (EI+ HRMS showed a deviation of 0.0082 ppm from the calc.

mass of C20H25N6O4). The melting point of 216–217 °C (decomp). of the

isolated compound 188 was sharp. The purity of the product 188 was found to

be >99.9% using analytical HPLC in an 80% methanol/ 20% water system.

161

4.4 Results of Biological testing of adenosine analogues

A number of C2-modified nitroxide adenosine analogues were synthesised with

ethyl linker groups. The potency and agonist activity (rather than antagonists or

inverse agonists) of the adenosine analogues was assessed with a downstream

cAMP assay using transfected CHO lines. These cells overexpress one of the

human A1, A2A, A2B, and A3 adenosine receptor subtypes. The details about the

cAMP Accumulation Assay have recently been described.121 This assay was

carried out at Monash University in Melbourne.

The tetraethylisoindoline-2-yloxyl nitroxide adenosine analogue 184 possessing

a two carbon chain (ethyl) linker moiety showed less A2AAR affinity (184; pEC50

of 5.2 ± 0.2) than non-nitroxide species (188; pEC50 of 6.0 ± 0.1) when

compared to all other receptors subtypes. The compound 188 with ethylene

linked phenyl was prepared in this structural class of comppounds is to compare

the effect of radical and non-radical species on adenosine receptors. The

compound 180 with tetramethyl isoindoline nitroxide linker was not tested for

cAMP assay because it was synthesised after the last batch of compounds was

tested.

All compounds in the final series are less-potent A2AAR agonists with pEC50

values ranging from 5.2–6.0. However, compound 184 is a slightly higher

potent adenosine analogue for A2BAR with pEC50 of 7.0 ± 0.5) compared to

other receptor subtypes. The non-nitroxide adenosine analogue also showed

162

somewhat good selectivity for A1AR and A3AR (pEC50 of 7.1± 0.2 and 7.0± 0.2)

respectively, compared to other adenosine receptor subtypes.

163

Table 4.1: cAMP analysis of ethyl linked adenosine compounds (pEC50 and pIC50 values in nM.

No. Compound Structure pIC50 -

A1R

pEC50 -

A2AR

pEC50 -

A2BR

pIC50 -

A3R A2A/A1 A2A/A3

A2A/A2

B A2B/A1 A2B/A3

A2B/

A2A

184

6.0± 0.2 5.2± 0.2 7.0± 0.5 N.D.(< 5) 0.2 <2 <2 10 100 63

188

7.1± 0.2 6.0± 0.1 6.3± 0.6 7.0 ± 0.2 0.08 0.1 0.1

A1/A2B

13

A3/A2B

10

-

164

Specifically, radical species (TEIO nitroxide) possessing adenosine analogue

184 with an ethyl linker moiety has been demonstrated to have 63 fold better

selectivity at the adenosine A2BAR over the adenosine A2AAR subtype.

However, it was 10 and 100 fold selective for the A1AR and A3AR subtype

respectively. Non-radical, non-nitroxide species (phenyl group) possessing

ethyl linked adenosine derivative 188, exhibited slightly increased selectivity

towards the adenosine A1AR and A3AR subtypes (10–13 fold), over the

adenosine A2AAR and A2BAR subtypes.

However, it was confirmed that the nitroxide possessing adenosine compounds

with ethyl linkers also have the similar receptor subtype affinity and selectivity at

A2BAR as the ethyne linked nitroxide adenosine compounds from the previous

chapter. In addition, it was also confirmed that flexibility of the linker chain has

no significant impact on receptor-binding .

165

Conclusion 5

166

5.1 Synthetic chemistry

This project focused on the design, synthesis and biological evaluation of novel

adenosine compounds that incorporate radical antioxidant species. These

hybrid analogues are designed to act potentially as selective adenosine

receptor agonists. They are specifically designed to target the A2A adenosine

receptors. The A2A adenosine receptors are one of the primary drug targets in

the management of ischaemia reperfusion injury. It has previously been

reported that having a potent antioxidant molecule within a molecular framework

of A2AAR agonists significantly increases their therapeutic efficacy. Therefore,

this work was based on linking/combining antioxidant molecules with known

adenosine receptor agonists. The main classes of antioxidant molecules

explored are nitroxides and phenolic compounds. These compounds were

linked via enzymatically cleavable (esters, amides) and non-clevable

(aminoethylaniline, ethynyl, ethyl) bonds.

Facilitating this approach, three structural classes of adenosine analogues were

designed and synthesised using a linear methodology. In total, twelve novel

hybrid adenosine compounds were synthesised in nine to twelve synthetic

steps. In eleven of those new adenosine analogues, the antioxidant moieties

were incorporated at the C2-position of the adenosine skeleton. These

analogues were designed to act as bifunctional ligands at the adenosine A2AAR

subtype. In the twelveth adenosine compound, the C2-position was modified

with a phenyl group instead of an antioxidant. This was intended to act as a

control to compare the effect of the presence of an antioxidant moiety.

167

5.1.1 Adenosine based antioxidant compounds with para-

aminoethylaniline linkers

In the first set of antioxidant-linked adenosine analogues, the C2-position of the

adenosine intermediate 146 was functionalised with a series of sterically

hindered antioxidant nitroxides via a para-aminoethylaniline linker. Different

ring classes of nitroxides, having tetramethyl substitution predominantly at the

α-carbon (some with tetraethyl substituent), were used. Adenosine intermediate

146 was synthesised in high yield from commercially available guanosine 138 in

seven steps. The key step in the synthesis of adenosine N-ethylcarboxamide

(NECA) 146 involves the generation of 143 which was achieved through the

synthesis of a novel guanosine-5-ethyl ester analogue 142. The synthesis of

143 was accomplished successfully in high yield by the esterification of

appropriately substituted guanosine-5-carboxylic acid 140.

Addition rate and addition sequence of the reagents and reactants make a

significant difference in the time span in completion of the amide coupling

reaction. The amide coupling reaction was complete in 21 h compared to 48 h,

when the carboxy nitroxide reactant was added after the addition of the other

coupling reagents to give the C2-substituted adenosine analogue in reduced

time.

The deprotection of the isopropylidene group was carried out using dilute

hydrochloric acid (HCl). Using this easier and cheaper deprotection reagent

gave better yields and compared to the reported resin mediated isopropylidene

168

deprotection.323 However, the isopropylidene deprotection of adenosine

derivatives with dilute HCl gave the deprotected adenosine analogues in (50–

71%) moderate yields. These moderate yields were attributed to the low

solubility of the deprotected adenosine analogues. The synthesis of the target

adenosine analogues was achieved in a with ten step reaction scheme starting

from guanosine and achieved in an overall yield of 9–12%.

Purification and isolation of the novel antioxidant possessing aminoethylaniline

linked adenosine analogues was found to be difficult using silica gel column

chromatography. The product generated has an Rf value close to the starting

materials and a byproduct. The use of medium pressure chromatography

(MPLC) with a reverse phase column was found to be a better technique to

purify those components. All the adenosine were analogues generated in >

98% pure and all the novel target adenosine analogues were isolated with >

99.9% purity (as analysed by HPLC).

5.1.2 Adenosine based antioxidant compounds with ethynyl linkers

In a second structural class of adenosine compounds, two novel alkynyl

(ethynyl) linked adenosine analogues were successfully synthesised

possessing sterically bulky isoindoline nitroxide substituents attached at the C2-

position of the adenosine skeleton. In this synthesis, the key step was the

formation of the ethynyl coupled product between the 2-iodo adenosine 149 and

the appropriate terminal alkyne functionality of 120 and 122 and 123. This

synthesis was achieved using the Sonogshira coupling procedure under mild

reaction conditions. The ethynyl linked adenosine product was successfully

169

synthesised in good overall yields of 53% and by 58% respectively. All the

novel target adenosine analogues were isolated in this class were > 99.9%

purity (as analysed by HPLC).

5.1.3 Adenosine based antioxidant compounds with ethyl linkers

In a third structural class of adenosine compound, three novel adenosine

analogues were successfully synthesised possessing ethyl linked sterically

bulky isoindoline nitroxides and a phenyl group substituents at the C2-position

of the adenosine skeleton. In this project, a four-stage reaction scheme was

employed to synthesise the target adenosine analogues. The isoindoline

nitroxide possessing ethynyl linked adenosine analogues 177 and 181 and 185

(from the chapter 3) were the key materials used as starting compounds. The

target adenosine analogues were achieved in excellent overall yields of 62%

and 64% respectively. In addition, a single non-nitroxide ethyl linked adenosine

derivative was synthesised using a three-step reaction scheme from the iodo

adenosine intermediate 149 in a high overall yield of 74%. All the novel target

adenosine analogues isolated in this class were > 99.9% purity (as analysed by

HPLC).

170

5.2 Biological Testing

In this project, three structural classes comprising of twelve novel hybrid

adenosine compounds were synthesised. In eleven of those novel compounds,

an antioxidant functionality was incorporated into the adenosine framework to

generate the bifunctional compounds. Each structural class of adenosine

analogue was tested for the adenosine agonist activity and selectivity at A1AR,

A2AAR, A2BAR, and A3AR. All three structural classes of adenosine analogues

were found to act as adenosine agonists to all adenosine receptor subtypes.

5.2.1 Adenosine based antioxidant compounds with para-

aminoethylaniline linkers

In this work, seven novel antioxidants possessing aminoethylaniline linked

adenosine analogues were found to be selective as the human adenosine

receptor agonists. Specifically, these adenosine analogues are acted as

selective bifunctional ligands at the adenosine A2AAR subtype. The adenosine

A2AARs are one of the primary drug targets in the pathophysiology of

cardiovascular diseases. The activation of adenosine A2AAR is required to

achieve higher cardioprotection via ischemic preconditioning. In addition, these

receptor subtype are strongly involved in anti-ischemic action to avoid damage

induced by reperfusion injury. In this regard, all these synthesised adenosine

analogues were shown to have very promising characteristics and may be

implicated in potential cardiac treatments.

The nitroxide radical functionality and the ring size of the cyclic nitroxide moiety

have no significant difference in the receptor-binding ability at any of the four

171

adenosine receptor subtypes. From this, it can be postulated that the nitroxide

functionality of the five compounds did not interact in any great extent within the

binding pocket of the adenosine receptors.

However, all these adenosine derivatives retained more than 200-fold selectivity

to adenosine A2AAR over the A1AR subtype. Specifically, the adenosine

analogue possessing PROXYL nitroxide (166) and TEIO nitroxide (170)

substituents being displayed more than 800 fold selectivity for the A2AAR over

the A1AR. These compounds have low selectivity versus A2BAR (3 and 10 fold).

In addition, these analogues demonstrate modest selectivity for A2AAR over the

A3AR (63 and three fold). Therefore, it is beneficial to implicate some of these

adenosine A2AAR selective analogues in further ischemic evaluations.

All the adenosine analogues from this first structural class were analysed in the

further cardioprotection testing by using cell-based simulated ischemia assay.

All compounds have demonstrated positive control in the study of an ischaemia

cell protection assay. The test indicates that, the positive cardioprotective effect

is primarily mediated by A2AAR activation. Furthermore, additional

cardioprotective effects were observed at the highest concentration is may be

due to the contribution of antioxidant activity. However, it can be said with

confidence that the incorporation of antioxidant functionality does not have a

measurable adverse effect on selectivity as adenosine A2AAR agonists. It is

reemphasized here that selectivity at the receptor subtype is an equally

important characteristic than compounds potency. This feature justifies the

172

original hypothesis that the C2-binding domain can accommodate sufficient

structural variations. This property makes feasible the design and application of

bifunctional compounds of this nature in the cardioprotection.

5.2.2 Adenosine based antioxidant compounds with ethynyl linkers

Compounds from a second structural class include antioxidant linked adenosine

analogues with ethynyl linkers. These adenosine analogues demonstrated no

significant impact on selectivity at adenosine A2AAR. However, these

analogues show slightly higher selectivity for the adenosine A2BAR over the

other adenosine receptor subtypes. This characteristic activity of these

analogues as a selective adenosine A2BAR agonists may be implicated in their

therapeutic applications.

5.2.3 Adenosine based antioxidant compounds with ethyl linkers

Compounds from a third structural class include two adenosine analogues

possessing various antioxidant isoindoline nitroxides linked with two carbon

chain (ethyl) linker moieties. Similarly, to the second class nitroxide adenosine

compounds, these nitroxide compounds exhibit high selectivity at A2BAR over

the other receptor subtypes. However, no significant selectivity at A2AAR was

observed.

However, non-nitroxide phenyl group possessing ethyl linked adenosine

derivative 188, exhibited slightly increased selectivity towards the adenosine

A1AR and A3AR subtypes over the adenosine A2AAR and A2BAR subtypes.

There is no significant difference in selectivities of non-nitroxide adenosine

173

analogues with an ethynyl or ethyl linkers, all are selective at adenosine A1AR

and A3AR subtypes.

In this structural class of compounds, the comparison between adenosine

compounds possessing antioxidant groups with non-antioxidant groups was

found to have significant effect on the selectivity of particular adenosine

receptor subtypes. The adenosine compound possessing antioxidant groups

has higher selectivity at the adenosine A2BAR subtype. However, the

adenosine compound possessing non-antioxidant groups has greater selectivity

at the A1AR and A3AR subtypes compared to other receptor subtypes. In this

regard, the characteristics of this adenosine analogues possessing nitroxide

antioxidant moiety as A2BAR selective analogues may be implicated in the

further testing in the potential treatment as coronary vasodilator or

antihypertensive agents.

174

Future work 6

175

No further pharmacological testing of the adenosine compounds was able to be

conducted within the time frame of the project and this remains an missing

piece of this work. While the functional data has confirmed that the compounds

possess agonists function, testing for compound antioxidant efficacy remains to

be performed.

The functional efficacy of the antioxidant combined adenosine analogues would

be more challenging to effectively measure. The work described in this thesis

has been an initial exploration into the concept of ligands for selective

adenosine receptors that also possess an antioxidant species. No further

pharmacological testing was conducted within the time frame of this project.

Although this research has resulted in the synthesis of compounds that are

potent adenosine agonists ligands, there are three main areas that can be

identified where there is opportunity for further development of the concept of

dual action adenosine receptor ligands. These areas are i) Synthesis of novel

adenosine compounds that are designed to be selective adenosine A2AAR

agonists with propynyl linkers linked to nitroxide antioxidant capabilities. This

joining of three carbon distance with a triple bond (alkyne) compounds

increases the selectivity for A2AAR and antioxidant function will also have

additional effects. ii) Synthesis of tetraethyl modified smaller ring nitroxides with

aminoethylaniline linked adenosine compounds will increase the selectivity for

A2AAR. The nitroxide functionality will be blocked at some extent by more

hindered tetraethyl group. This phenomenon will achieve better

cardioprotection. iii) More extensive biological evaluation of the novel

compounds synthesised in this work remained to be performed, as the

176

functional data have confirmed that the compounds possess agonist function,

testing for compound antioxidant efficacy remains to be performed.

More extensive biological testing of the novel adenosine derivatives described

in this work is required in order to measure their potential as cardioprotective

treatments. Although the antioxidant activity of both BHT and TMIO and other

nitroxide functional groups are mentioned in the literature, the novel compounds

synthesised in this work have not yet been tested for the comprehensive

assessment of antioxidant activity. In vitro, evaluation of the antioxidant

capabilities of these antioxidant coupled adenosine analogues should be

performed. In addition, the further biological assay should be performed in a

more complex biological system such as isolated perfused rat hearts

(Langendorff heart assay) and live animal models which stimulate heart attack

and ischaemia-reperfusion injury. However, only after the biological evaluation

of these compounds in a more complex system, their viability can be assessed

as a potential cardioprotective treatment.

177

Experimental work 7

178

7.1 General Experimental

All starting materials and required chemicals were purchased from Sigma-

Aldrich with purities of more than 95% and all solvents used were of analytical

reagent (AR) grade except for HPLC. Solvents were dried according the

appropriate drying procedure under an argon atmosphere. Solvents used such

as acetone, tetrahydrofuran (THF), Dichloromethane (DCM), Chloroform

(CHCl3), and methanol (MeOH), N, N-dimethylformamide (DMF) for air and

moisture sensitive reactions were distilled under an argon atmosphere. The

solvents such as diethyl ether (Et2O), and toluene were dried under sodium wire

and acetonitrile (MeCN) was dried under predried molecular sieves and used

absolute ethanol (EtOH) for moisture sensitive reactions. Demineralised (DM)

water was used for all reaction work-ups. Thin layer chromatography was

performed on aluminium backed silica F254 plates with UV light (254 nm) for

chemical compound detection. Column chromatography was performed using

kieselgel 60 silica gel (200- 300 mesh) with eluents.

Nuclear magnetic resonance spectra were recorded on a Bruker Advance 300

WB (1H at 400 MHz and 13C at 75 MHz) or a Varian (1H NMR at 400 MHz and

13C NMR at 101 MHz) FT- NMR spectrometer instrument. NMR spectra are

reported based on integration, chemical shift (δ ppm) and multiplicity (s =

singlet, d = doublet, dd = doublets of doublet, t = triplet, q = quartet, m =

multiplet, br = broad, coupling constant and assignment. 13C NMR spectra are

reported according to the chemical shift, multiplicity, coupling constant (if

appropriate) and assignment. Deuterated solvents acetone (1H NMR 2.05/13C

179

NMR 29.84 ppm), chloroform (1H NMR 7.26/13C NMR 77.16 ppm); DMSO (1H

NMR 2.50/13C NMR 39.53 ppm) and methanol (1H NMR 3.31/13C NMR 49.00

ppm) was used for recording nuclear magnetic resonance spectra with their

respective solvent residual peaks.

High resolution mass analysis were performed on the Micromass platform II

single quadrupole mass spectrometer equipped with the dual electron spray ion

source. Melting points were measured using a Gallenkamp melting point

apparatus with a calibrated thermometer. All the target final compounds (161-

174 and 177-188) were purified using a medium pressure liquid

chromatography (MPLC) instrument using filtered HPLC grade methanol and

HPLC grade water using C18 reverse phase column with 1 mL/ minute flow rate

of mobile phase and 264 nM UV detector. The purities of novel compounds

(161-174 and 177-188) were analysed using analytical high performance liquid

chromatography (HPLC) using a C18 reverse phase column with a 1 mL/minute

flow rate with various solvent (methanol/water) ratios of mobile phase and 264

nM UV detector.

180

7.2 Synthetic procedure

7.2.1 Synthesis of 2-benzyl phthalimide (100).

A suspension of phthalic anhydride (98) (20 g, 135 mmol, 1 equiv) in acetic acid

(93.5 mL) was treated with benzylamine (99) (22.42 mL, 205 mmol, 1.52 equiv).

The reaction mixture was heated at 120 °C and stirred for 1 h. The resulting

solution was poured onto an ice/ water mixture (150 mL) and collect the white

precipitate by filteration. The solid was recrystallised from ethanol to give 2-

benzyl phthalimide (100) as a white solid (26.5 g, 83%). Rf = 0.83 (EtOAc /

Hexane, 1:1).

1H NMR (400 MHz, CDCl3) δ ppm: 4.85 (s, 2 H) 7.29 - 7.35 (m, 3 H) 7.43 (d,

J=6.99 Hz, 2 H) 7.67 - 7.72 (m, 2 H) 7.84 (dd, J=5.40, 3.06 Hz, 2 H).

13C NMR (101MHz, CDCl3) δ ppm: 41.6, 123.3, 127.8, 128.6, 128.7, 132.1,

134.0, 136.3, 168.1. The obtained spectroscopic data was consistent with that

previously reported.324

Melting point: 116–118 °C (lit.325 116 °C).

HRMS (ESI): m/z calcd for C15H11NNaO2+ [M+Na]: 260.0687, found: 260.0813

181

7.2.2 Synthesis of 2-benzyl-1,1,3,3-tetramethylisoindoline (101).

A solution of compound 100 (40 g,168 mmol,1.0 equiv) in anhydrous toluene

(320 mL) was treated with methyl magnesium iodide [freshly prepared from

methyl iodide (62.75 mL,1008 mmol, 6.0 equiv) and pre-dried magnesium

turnings (33 g, 1348 mmol, 8.0 equiv) in anhydrous Et2O (400 mL)]. The Et2O

was distilled off via a Dean-stark. The reaction mixture was heated to reflux,

stirred for 3 h and then concentrated to half its volume. Hexane (600 mL) was

added, and the resulting mixture was filtered through celite and washed

thoroughly with extra hexane (600 mL). The filtrate was passed through a

column of basic alumina and evaporated under reduced pressure to give a

colourless oil that solidified at ambient temperature. This solid was

recrystallised in MeOH to give 2-benzyl-1,1,3,3-tetramethylisoindoline (101) as

a white crystalline solid (14 g, 31%), Rf = 0.9 (EtOAc / Hexane, 1:1).

1H NMR (400 MHz, CDCl3) δ ppm: 1.33 (s, 12 H) 4.02 (s, 2 H) 7.16 (dd, J=5.52,

3.17 Hz, 2 H) 7.26 (br. s., 2 H) 7.27 - 7.34 (m, 3 H) 7.49 (d, J=7.23 Hz, 2 H).

13C NMR (101 MHz, CDCl3) δ ppm: 28.4, 46.3, 65.2, 120.6, 122.1, 126.0, 127.2,

127.5, 128.6, 129.0, 143.5 and 147.9. The obtained spectroscopic data was

consistent with that previously reported.76

Melting point: 63–65 °C (lit.76 63–64 °C).

182

HRMS (ESI): m/z calcd for C19H24N+ [M+H]: 266.1909, found: 266.1936.

7.2.3 Synthesis of 5-bromo-1,1,3,3-tetramethylisoindoline (102)275

A solution of compound 101 (5 g, 18.8 mmol, 1 equiv) in DCM (30 mL) under an

atmosphere of argon was cooled to 0 °C in an ice bath. A solution of liquid Br2

(2.15 mL, 41.47 mmol, 2.2 equiv) in DCM (42 mL) was cautiously added

followed by the immediate addition of anhydrous AlCl3 (9.5 g, 65.8 mmol, 3.5

equiv). The reaction was stirred at 0 °C for 1 h and then poured onto ice (200

mL) and stirred vigorously for 30 minutes after which time the solution was

basified with 10 M NaOH solution up to pH 14 and extracted with DCM (3 × 50

mL). The combined DCM layers were washed with brine (3 × 50 mL) and

evaporated under reduced pressure to give 2, 5-dibromoisoindoline as a yellow

oily residue.

The residue was dissolved in methanol (30 mL), and NaHCO3 (280 mg) was

added to the solution. Aqueous H2O2 (30% aqueous solution, 15 mL) was then

added slowly until no further effervescence could be detected (ensuring that

some NaHCO3 remained). The mixture was acidified with 2 M H2SO4 aqueous

solution (up to pH 1) and extracted with DCM (3 × 50 mL) to remove the

benzaldehyde by-product. The combined organic layers were back extracted

with 2 M H2SO4 to recover any removed product. The combined aqueous

phases were washed with DCM (3 × 50 mL), after which time they were cooled

183

in ice bath, basified with 10 M NaOH aqueous solution (up to pH 14) and

extracted with DCM (3 × 50 mL). The combined organic phases were washed

with brine (3 × 50 mL) and dried (over anhydrous Na2SO4). The solvent was

removed under reduced pressure to give a golden oil which immediately

crystallised to give yellowish-white solid, which was purified by silica gel column

chromatography using a mixture of DCM: MeOH (8:2) to give 5-bromo-1,1,3,3-

tetramethylisoindoline (102) as a pale yellow solid (3.15 g, 64.5%). Rf = 0.65

(EtOAc: Hexane, 1:1).

1H NMR (400 MHz, CDCl3): δ ppm 1.45 (s, 6 H) 1.47 (s, 6 H) 1.80 (br s, 1 H)

7.00 (d, J=8.03 Hz, 2 H) 7.25 (d, J=1.58 Hz, 2 H) 7.35 - 7.39 (m, 3 H).

13C NMR (101 MHz, CDCl3): δ ppm 31.8, 62.7, 120.8, 123.1, 123.2, 124.7,

147.9, 151.3. (The obtained spectroscopic data was consistent with that

previously reported)276

Melting point: 58–60 °C (lit.276 58–60 °C).

HRMS (ESI): m/z calcd for C12H17NBr+ [M+H]: 254.0544 and 256.0524, found:

254.0568 and 256.0549.

7.2.4 Synthesis of 5-Bromo-1,1,3,3-tetramethylisoindoline-2-yloxyl

(103)276.

184

To a solution of compound 102 (1.0 g, 3.92 mmol, 1 equiv), NaHCO3 (400 mg,

4.312 mmol, 1.1 equiv) and Na2WO4.2 H2O (169 mg, 0.78 mmol, 0.2 equiv) in

methanol (15 mL) was added 30% H2O2 solution (2.7 mL, 28.6 mmol, 7.3

equiv). The reaction mixture was stirred for 24 h, after which time added a

second portion of NaHCO3 (400 mg, 4.312 mmol, 1.1 equiv), Na2WO4.2 H2O

(169 mg, 0.78 mmol, 0.2 equiv) and 30% H2O2 (2.7 mL, 28.6 mmol, 7.3 equiv)

were added and stirring was continued for 48 hours. Water (40 mL) was then

added into the reaction solution and the resulting mixture was extracted with

DCM (3× 40 mL). The combined organic phases were washed with 2 M HCl

solution (40 mL), brine solution (3 × 40 mL) and dried over anhydrous Na2SO4.

The solvent was concentrated under reduced pressure. The residue was

recrystallised (MeCN) to give 5-bromo-1, 1, 3, 3- tetramethylisoindoline-2-yloxyl

(103) as a yellowish crystalline solid (1.0 g, 95%), Rf = 0.01 (EtOAc / MeOH,

8:2).

Melting point: 108–110 °C (lit.276 109 °C).

HRMS (ESI): m/z calcd for C12H16NBrO+ [M+2H]: 270.0494, found: 270.0487.

185

7.2.5 Synthesis of 5-iodo-1,1,3,3- tetramethylisoindoline (104)276.

Compound 102 (300 mg, 1.176 mmol, 1 equiv) was dissolved in dry THF (3.4

mL) under an atmosphere of argon. The solution was cooled in a dry ice and

acetone bath at –78 °C. A solution of n-BuLi (1.6 M in hexanes, 1.95 mL, 3.22

mmol, 2.74 equiv) was added dropwise to the stirring mixture, followed by the

addition of a solution of iodine (0.9 g, 65.8 mmol, 6 equiv) in dry THF (7.5 mL).

The reaction mixture was cooled to room temperature and then quenched by

pouring onto an ice water mixture (150 mL). The mixture was basified with 5 M

NaOH solution (up to pH 14). The resulting mixture was extracted with DCM (3

× 25 mL), the combined organic phases were washed with brine (3 × 25 mL)

and dried over anhydrous Na2SO4 and the solvent was concentrated under

reduced pressure to give a yellow oily residue.

The residue was taken up in MeOH (7.5 mL) and NaHCO3 (37.5 mg) was

added, followed by the addition of 30% H2O2 solution, until no further

effervescences could be detected. The mixture was treated with 2 M H2SO4 (50

mL) and washed with DCM (3 × 25 mL). The combined organic layers were

back extracted with 2 M H2SO4 to recover any removed product. The combined

aqueous phases were washed with DCM (25 mL) after which time they were

cooled in ice bath and basified with 5 M NaOH solution (up to pH 14). The

resulting solution was extracted with DCM (3 × 25 mL). The combined organic

phases were washed with brine (3 × 25 mL), dried over anhydrous Na2SO4 and

186

the solvent was evaporated under reduced pressure to give a golden oil which

immediately crystallised to give 5-iodo -1, 1, 3, 3- tetramethylisoindoline (104)

as a pale yellow solid (243 mg, 69%). Rf = 0.67 (EtOAc / Hexane, 3:7).

1H NMR (400 MHz, CDCl3) δ ppm: 1.43 (s, 6 H) 1.44 (s, 6 H) 1.93 (br. s., 1 H)

6.88 (d, J=7.92 Hz, 1 H) 7.44 (d, J=1.03 Hz, 1 H) 7.56 (dd, J=7.92, 1.22 Hz, 1

H).

13C NMR (101 MHz, CDCl3) δ ppm: 31.7, 62.7, 92.2, 123.5, 130.8, 136.1, 148.6,

and 151.4. (The obtained spectroscopic data was consistent with that previously

reported)79

ESMS calcd for C12H17IN+ [M+H]: 302.0406, found: 302.0400.

7.2.6 Synthesis of 5-iodo-1,1,3,3-tetramethylisoindoline-2-yloxyl (105)79

To a stirring solution of compound 104 (180 mg, 0.59 mmol, 1 equiv) in MeOH

(2.7 mL), NaHCO3 (60 mg, 0.705 mmol, 1.195 equiv), Na2WO4.2H2O (25.4 mg,

0.705 mmol, 0.135 equiv) and 30% H2O2 (0.5 mL, 4.307 mmol, 7.3 equiv) was

added and the reaction mixture was stirred at room temperature for 24 h. A

second portion of NaHCO3 (60 mg, 0.705 mmol, 1.195 equiv), Na2WO4.2H2O

(25.4 mg, 0.705 mmol, 0.135 equiv) and 30% H2O2 solution (0.5 mL, 4.307

mmol, 7.3 equiv) was then added. After 48 h, water (30 mL) was added and the

resulting mixture was extracted with DCM (3 × 20 mL). The combined organic

phases were washed with 2 M HCl solution (25 mL) and brine solution (3 × 25

187

mL) and dried over anhydrous Na2SO4. The solvent was concentrated under

reduced pressure to give a yellow crystalline solid and which was recrystallised

(MeCN) to give 5-iodo-1,1,3,3-tetramethylisoindoline-2-yloxyl (105) as an

orange crystalline solid (150 mg, 79%). Rf = 0.71 (EtOAc / Hexane, 3:7).

Melting point: 133–135 °C (lit.79 133–135 °C).

ESMS calcd for C12H16INNaO+ [M+Na]: 339.0096, found: 339.0090.

7.2.7 Synthesis of 5-carboxy-1,1,3,3-tetramethylisoindoline-2-yloxyl

(106)275.

A solution of n-BuLi (1.6 M in hexanes, 1.95 mL, 3.22 mmol, 2.74 equiv ) was

added dropwise to a stirring solution of compound 102 (300 mg, 1.176 mmol, 1

equiv) in a freshly distilled dry THF (3.4 mL) at –78 °C under an atmosphere of

argon. After stirring for 10 min, the solution was added slowly to a mixture of

crushed dry ice (0.9 g, 65.8 mmol, 6 equiv) in freshly distilled dry THF (7.5 mL).

The reaction mixture was warmed to room temperature under an atmosphere of

argon and then concentrated under reduced pressure. 2 M HCl (15 mL) was

added to the resulting residue and the solution was washed with Et2O (3 × 10

mL). The combined organic phases were washed with 2 M HCl (15 mL). The

combined aqueous phases were neutralised with saturated Na2CO3 solution (15

mL) and extracted with Et2O (3 × 20 mL). The ether layers were washed with

188

water (3 × 20 mL) and brine (3 × 20 mL) and dried over anhydrous Na2SO4.

The solvent was evaporated under reduced pressure to give yellow oily residue.

The yellow oily residue was dissolved in MeOH (2 mL) at room temperature.

NaHCO3 (40 mg, 3.92 mmol, 1.1 equiv), Na2WO4.2H2O (24 mg, 0.78 mmol, 0.2

equiv) and 30% H2O2 solution (0.56 mL, 28.6 mmol, 7.3 equiv) were added and

the reaction mixture was stirred for 24 h. A second portion of NaHCO3 (40 mg,

3.92 mmol, 1.1 equiv), Na2WO4.2 H2O (24 mg, 0.78 mmol, 0.2 equiv) and 30%

H2O2 (0.56 mL, 28.6 mmol, 7.3 equiv) was added. The reaction mixture was

stirred again for 48 h, after which time water (30 mL) was added to the reaction

mixture and the resulting mixture was extracted with DCM (3× 40 mL). The

combined organic extracts were washed with 2 M HCl solution (40 mL) and

brine solution (40 mL) and dried over Na2SO4. The solvent was concentrated

under reduced pressure to give a yellow crystalline solid, which was

recrystallised (MeCN) to give 5-carboxy-1, 1, 3, 3- tetramethylisoindoline-2-

yloxyl (106) as a yellowish crystalline solid (140 mg, 76.5%). Rf = 0.01 (EtOAc /

MeOH, 8:2).

IR: 2977 (alkylCH3)), 1680 (C=O), 1423, 1357 (N-O), 1256, 1160, 908, 840,

774, 744.

Melting point: 216–216 °C (lit.275 214–218 °C).

ESMS calcd for C13H17 NO3+ [M+H+]: 235.1208, found: 235.1198.

EPR (methanol): typical 3-line nitroxide signal, g 1.9823, aN 1.5007 mT.

189

7.2.8 Synthesis of 2-benzyl-5-methylphthalimide(108)102.

Benzylamine (20.5 mL, 92.6 mmol, 1.52 equiv) was added to a stirring

suspension of 5-methylphthalic anhydride (107) (20.0 g, 123 mmol, 1 equiv) in

acetic acid (94 mL). The reaction mixture was refluxed at 120 °C for 1 h and

then poured onto ice/ water mixture (500 mL) and stirred vigorously for 30

minutes and the obtained white precipitate was filtered. The residue was

recrystallised from EtOH to give 2-benzyl-5-methylphthalimide (108) as a white

solid (24.8g, 80%) Rf = 0.83 (100% EtOAc),

1H NMR (400 MHz, CDCl3) δ ppm: 2.45 - 2.54 (m, 3 H) 4.83 (s, 2 H) 7.22 - 7.35

(m, 3 H) 7.39 - 7.51 (m, 3 H) 7.60 - 7.67 (m, 1 H) 7.67 - 7.78 (m, 1 H).

13C NMR (101 MHz, DMSO- d6) δ ppm: 22.0, 41.5, 123.3, 123.9, 127.7, 128.5,

128.6, 129.5, 132.9, 134.5, 136.8, 145.3, 168.1 and 168.2. (The obtained

spectroscopic data were consistent with that previously reported).102

Melting point:128–130 °C .(lit 102 128–130 °C)

HRMS (ESI): m/z calcd for C16H14NO2+ [M+H]: 252.1025, found: 252.1067.

190

7.2.9 Synthesis of 2-benzyl-5-methyl-1,1,3,3-tetraethylisoindoline(109)102.

A solution of compound 108 (10.0 g, 39.8 mmol, 1 equiv) in anhydrous toluene

(80 mL) was treated with ethyl magnesium iodide [freshly prepared from ethyl

iodide (19.2 mL, 239 mmol, 6.0 equiv) and pre-dried magnesium turnings (7.78

g, 318.4 mmol, 8.0 equiv.) in anhydrous Et2O (100 mL)]. The Et2O was distilled

off via dean-stark. The reaction mixture was heated to reflux, stirred for 3 h and

then concentrated to half its volume. Hexane (4 x 200 mL) was added to the

reaction mixture. The resulting mixture was filtered through celite and washed

thoroughly with extra hexane (600 mL). The extracted hexane was bubbled

overnight to dryness. The residue was dissolved in hexane (600 mL) and

passed through a column of basic alumina. The resulting filtrate was

evaporated under reduced pressure to give 2-benzyl-5-methyl-1,1,3,3-

tetraethylisoindoline (109) as a colourless oil (4.25 g, 32%). Rf = 0.92 (100%

EtOAc).

1H NMR (400 MHz, CDCl3) δ ppm: 0.78 (td, J=7.34, 2.93 Hz, 12 H) 1.53 (dd,

J=13.90, 7.30, 3.30 Hz, 4 H) 1.84 - 1.97 (m, 4 H) 2.38 (s, 3 H) 4.01 (s, 2 H) 6.87

(s, 1 H) 6.95 (d, J=7.78 Hz, 1 H) 7.00 - 7.06 (m, 1 H) 7.24 (br. s., 1 H) 7.27 -

7.34 (m, 2 H) 7.46 (d, J=7.04 Hz, 2 H).

13C NMR (75 MHz, CDCl3) δ: 9.6, 21.5, 30.3, 46.8, 71.0, 71.2, 123.2, 124.0,

125.3, 126.5, 127.7, 128.2, 129.1, 129.3, 135.0, 141.7, 142.5, and 144.8; (The

191

obtained spectroscopic data was consistent with that previously reported for this

compound).102

HRMS (ESI): m/z calcd for C24H34N+ [M+H]: 336.2691, found: 336.2677.

7.2.10 Synthesis of 5-methyl-1,1,3,3-tetraethylisoindoline (110).

To a solution of compound 109 (4.2 g, 12.5 mmol, 1.0 equiv) in acetic acid (100

mL) was added palladium (10% charcoal = 1.0 g). The reaction mixture was

shaken under hydrogen pressure (50 psi in Parr apparatus) at room

temperature for 3 h. The mixture was filtered through celite and concentrated

under reduced pressure. The resulting residue was dissolved in DCM (150 mL)

and the resulting solution was washed with saturated sodium bicarbonate

(Na2HCO3) solution (3 x 50 mL). The organic phase was dried over anhydrous

Na2SO4 and concentrated under reduced pressure to give 5-methyl-1,1,3,3-

tetraethylisoindoline (110) as a yellow orange oil (2.82 g, 92%). Rf = 0.92

(EtOAc: MeOH, 8:2).

1H NMR (400 MHz, CDCl3) δ ppm: 0.80 - 0.91 (m, 12 H) 1.59 - 1.78 (m, 8 H)

2.35 (s, 3 H) 3.49 (s, 1 H) 6.86 (s, 1 H) 6.91 - 6.97 (m, 1 H) 7.01 (s, 1 H).

13C NMR (101 MHz, CDCl3) δ ppm: 9.1, 21.5, 33.9, 47.0, 68.1, 68.2, 122.3,

123.2, 127.5, 136.2, and 147.9. (The obtained spectroscopic data was

consistent with that previously reported for this compound)102.

192

HRMS (ESI): m/z calcd for C17H28N+ [M+H]: 246.2222, found: 246.2226.

7.2.11 Synthesis of 5-methyl-1,1,3,3-tetraethylisoindoline-2-yloxyl (111).

A solution of compound 110 (2.8 g,11.4 mmol, 1 equiv), NaHCO3 (1.04 g,125

mmol, 1.1 equiv) and Na2WO4.2 H2O (0.42 g,15.5 mmol, 1.15 equiv) in MeOH

(80 mL), was added dropwise 30% H2O2 solution (9.2 mL,105 mmol, 7.3 equiv

). The reaction mixture was stirred at room temperature for 24 h after which

time a second portion of NaHCO3 (1.04 g,125 mmol, 1.1 equiv) and Na2WO4.2

H2O (0.42 g,15.5 mmol, 1.15 equiv) and then 30% H2O2 solution (9.2 mL,105

mmol, 7.3 equiv) were added and the stirring was continued for an additional 48

h. Water (160 mL) was then added to a reaction mixture and the resulting

mixture was extracted in DCM (3 x 50 mL). The combined organic phases were

washed with 2 M H2SO4 solution (3 x 50 mL), brine (3 x 50 mL) solution and

dried over anhydrous Na2SO4, filtered. The solvent was evaporated under

reduced pressure. The resulting residue was purified by silica gel column

chromatography (eluent 100 % DCM) to give 5-methyl-1,1,3,3-

tetraethylisoindoline-2-yloxyl (111) as a yellowish orange oil (1.35 g, 47%). Rf =

0.53 (1:1 DCM/ Hexane).

HRMS (ESI): m/z calcd for C17H27NaN+ [M+Na]: 283.191, found: 283.1934. (The

obtained spectroscopic data was consistent with that previously reported for this

compound).102

193

7.2.12 Synthesis of 2-acetoxy-5-methyl-1,1,3,3-tetraethylisoindoline (112).

To a stirring solution of compound 111 (1.35 g, 5.17 mmol, 1 equiv) in a dry

THF (70 mL) was added palladium (10% charcoal, 137 mg). The reaction

mixture was stirred under balloon of hydrogen for 1 h. The reaction mixture was

cooled at 0 °C in ice bath and, triethylamine (1.44 mL, 10.34 mmol, 2 equiv) and

acetyl chloride (0.92 mL) were added. A mixture was allowed to warm up to 25

°C and stirred for 1 h. Argon gas bubbled over the mixture for 10 minutes and

the reaction mixture was filtered through celite and evaporated under reduced

pressure. Water was poured onto the residue and the resulting mixture was

extracted with EtOAc (3 x 50 mL). The organic extracts were dried over

anhydrous Na2SO4 and evaporated by distillation under reduced pressure. The

resulting residue was purified by silica gel column chromatography (eluent 1:1

DCM/ Hexane) to give 2-acetoxy-5-methyl-1,1,3,3-tetraethylisoindoline (112) as

a colourless oil which solidifies upon standing (1.55 g, 99%). Rf = 0.78 (DCM/

Hexane, 1:1).

1H NMR (400 MHz, CDCl3) δ ppm: 0.79 (br. s., 6 H) 0.95 (d, J=4.70 Hz, 6 H)

1.60 - 1.80 (m, 4 H) 1.83 - 2.02 (m, 4 H) 2.10 (s, 3 H) 2.36 (s, 3 H) 6.86 (s, 1 H)

6.94 (d, J=7.63 Hz, 1 H) 7.06 (d, J=7.48 Hz, 1 H).

13C NMR (101 MHz, CDCl3) δ ppm: 8.7, 9.6, 19.6, 21.7, 29.0, 30.4, 73.7, 73.8,

123.5, 124.2, 127.6, 136.3, 138.8, 141.9, and 170.7. (The obtained

194

spectroscopic data was consistent with that previously reported for this

compound).102

Melting point: 78–80 °C (Lit102 76–78 °C).

HRMS (ESI): m/z calcd for C19H30NO2+ [M+H]: 304.2277, found: 304.2293.

7.2.13 Synthesis of 2-acetoxy-5-carboxy-1,1,3,3-tetraethylisoindoline(113).

A solution of compound 112 (1.55 g, 5.12 mmol, 1 equiv) in tert-butyl alcohol

(35 mL) was warmed to 40 °C. Anhydrous MgSO4 (0.62 g, 5.12 mmol, 1 equiv)

and potassium permanganate (0.4 M solution in water, 60 mL, 20.68 mmol, and

4.04 equiv) were added to the solution and heated to 70 °C for 24 h and the

reaction progress was followed by TLC analysis DCM: EtOAc (6:4). The

solution was cooled to 40 °C, was treated with 2-propanol (50 mL) and the

reaction mixture was stirred at room temperature for 16 h, after which time it

was filtered through celite. The filtrate was then evaporated to half its volume

and acidified with 2 M HCl solution (50 mL) up to pH 2. Water was added to the

mixture (50 mL) and extracted with Et2O (4 x 50 mL). The organic layers were

dried over anhydrous Na2SO4. The combined organic phases were evaporated

under reduced pressure. The resulting residue was purified by using silica gel

column chromatography (eluent DCM/ EtOAc, 6:4) to give 2-acetoxy-5-carboxy-

1,1,3,3-tetraethylisoindoline (113) as a white solid (1.32 g, 78%).

195

1H NMR (400 MHz, CDCl3) δ ppm: 0.80 (br. s., 6 H) 0.97 (br. s., 6 H) 1.57 - 1.85

(m, 4 H) 1.85 - 2.06 (m, 4 H) 2.12 (s, 3 H) 7.17 (d, J=8.07 Hz, 1 H) 7.80 (s, 1 H)

8.03 (d, J=7.92 Hz, 1 H).

13C NMR (101 MHz, CDCl3) δ ppm: 8.5, 9.4, 19.3, 28.9, 30.2, 73.7, 74.0, 123.7,

125.4, 128.0, 128.9, 142.3, 148.1 , 170.3., 170.8 (The obtained spectroscopic

data was consistent with that previously reported for this compound).102

Melting point: 168–170 °C. (Lit102 168–170 °C).

HRMS (ESI): m/z calcd for C19H28NO4+ [M+H]: 334.2018, found: 334.2042.

7.2.14 Synthesis of 5-carboxy-1,1,3,3-tetraethylisoindoline-2-yloxyl (114).

Compound 113 (1.3 g, 3.91 mmol, 1 equiv) was suspended in water (26 mL)

and the mixture was cooled using ice/salt mixture up to 0–5 °C. Lithium

hydroxide (LiOH) (0.462 g, 19.47 mmol, 4.98 equiv) was added to the reaction

mixture and ice bath was removed. The mixture was stirred at room

temperature for 20 h and the reaction progress was followed by TLC analysis

EtOAc: MeOH (8:2) The resulting yellow solution was again cooled in ice bath

and acidified with 2 M HCl (50 mL) solution and extracted with Et2O (5 x 50 mL).

The combined ether layers were treated with lead oxide (PbO2) (0.462 g, 1.95

mmol, 0.5 equiv) and stirred for 30 minutes. The ether extract was dried using

anhydrous Na2SO4, filtered. The filtrate was evaporated under reduced

196

pressure to give yellow oil that solidifies upon standing, which was recrystallised

(MeCN) to give 5-carboxy-1,1,3,3-tetraethylisoindoline-2-yloxyl (114) as yellow

crystals. (1.02 g, 89%).

Melting point: 102–104 °C. (lit.102 97–99 °C)

HRMS (ESI): m/z calcd for C17H24NNaO3+ [M+Na]: 313.1654, found: 313.1680.

EPR (methanol): typical 3-line nitroxide signal, g 1.9928, aN 1.4418 mT.

7.2.15 Synthesis of 5-(3-hydroxy-3-methyl)butynyl-1,1,3,3-

tetramethylisoindoline-2-yloxyl) (119).

To a solution of compound 105 (770 mg, 2.435 mmol, 1 equiv) in an anhydrous

triethylamine (15 mL) was added in a stirring solution of Pd(OAc)2 (13 mg,

0.021 mmol, 0.1 equiv), and DABCO (820 mg, 7.31 mmol, 3 equiv) and 2-

methyl-3-butyne-2-ol (204). The argon was bubbled for 10 minutes in a mixture.

The reaction mixture was stirred at 80–85 °C for 3 days and after which time,

CHCl3 (50 mL) was added to the reaction mixture. The organic phase was

washed with water (3 x 50 mL) and brine solution (3 x 50 mL) and dried over

anhydrous Na2SO4. The solvent was evaporated under reduced pressure and

the resulting residue was purified by silica gel column chromatography using a

mixture of (1:9) EtOAc: Hexanes to give 5-(3-hydroxy-3-methyl)butynyl-1,1,3,3-

197

tetramethylisoindoline-2-yloxyl) (119) as a brown-oil (530 mg, 80 %), Rf = 0.71

(DCM/ Hexanes, 2:8).

HRMS (ESI): m/z calcd for C17H22NO2+ [M+]: 272.1651, found: 272.1654. (The

obtained spectroscopic data was consistent with that previously reported for this

compound).316

7.2.16 Synthesis of 5-ethynyl-1,1,3,3-tetramethylisoindoline-2-yloxyl (120).

To a solution of compound 119 (200 mg, 0.734 mmol, 1 equiv), in an anhydrous

MeCN (15 mL) added solid KOH (330 mg, 5.87 mmol, 8.0 equiv). The reaction

mixture was stirred at 80–85 °C for 4 h and after which time water (30 mL) was

added to the reaction mixture. The resulting mixture was extracted with EtOAc

(3 x 50 mL). The combined organic phases were washed with water (15 mL)

and brine solution (15 mL) and dried over an anhydrous Na2SO4, filtered. The

solvent was evaporated under reduced pressure and the resulting residue was

purified on silica gel column chromatography using a mixture of DCM: Hexanes

(9:1) to give 5-ethynyl-1,1,3,3-tetramethylisoindoline-2-yloxyl (120) as a

yellowish-purple oil (141 mg, 90 %). Rf = 0.71 (DCM/ Hexanes, 2:8).

Ʋmax (ATR-FTIR) cm-1: 3726 (aryl C-H), 3192 (ΞC-H), 3044, 2978 (alkyl CH3),

2166 (CΞC), 1487 (aryl C-C), and 1428 (N-O).

Melting point: 96–98 °C (lit).

198

HRMS (ESI): m/z calcd for C14H17NO+ [M+Na]: 237.1130, found: 237.1126.

(The obtained spectroscopic data was consistent with that previously reported

for this compound).316

EPR (methanol): typical 3-line nitroxide signal, g 1.9908, aN 1.1266 mT.

7.2.17 Synthesis of N-tert-butoxycarbonyl-2-(4-aminophenyl)ethylamine

(125)277, 326

A solution of 2-(4-aminophenyl)ethylamine (124) (550 mg, 4.04 mmol, 1 equiv)

in DCM (50 mL) was cooled to –5 °C and tert-butoxycarbonyl anhydride (882

mg, 4.04 mmol, 1 equiv) was slowly added. The reaction mixture was stirred for

100 minutes at –5 to –10 °C, and the reaction progress was followed by TLC

analysis using DCM / MeOH (9:1) and poured onto an ice/ water mixture. After

stirring vigorously for 30 minutes, the organic phase was separated and washed

with water (3 × 25 mL). The combined aqueous layers were then washed with

DCM (3 × 25 mL). The combined organic layers were washed with brine

solution (3 × 25 mL) and dried, over anhydrous MgSO4, filtered. The solvent

was evaporated under reduced pressure and the resulting residue purified by

silica gel column chromatography to give N-tert-butoxycarbonyl-2-(4-

aminophenyl)ethylamine (125) as an orange solid (847 mg, 88.7%). Rf = 0.71

(DCM / MeOH, 9:1).

Melting point: 56–58 °C (lit.121 55–60 °C).

199

1H NMR (400 MHz, DMSO-d6) δ ppm 1.37 (s, 9 H) 2.95 - 3.07 (m, 2 H) 3.17 (d,

J=4.36 Hz, 2 H) 4.12 (d, J=5.06 Hz, 1 H) 4.85 (s, 2 H) 6.47 (d, J=8.33 Hz, 2 H)

6.82 (d, J=8.25 Hz, 2 H).

13C NMR (101 MHz, CDCl3) δ ppm: 28.4, 35.2, 41.9, 79.1, 115.3, 128.8, 129.6,

144.7, and 155.9.

Ʋmax (ATR-FTIR) cm-1: 3369 (NH2), 3188 (aryl C-H), 2976 (alkyl CH3), 1678

(NC=OO), 1511 (aryl C-C), 1362 (O-C(CH3)3), 1244 (H-NCOO),

ESMS calcd for C13H20NaN2O2+ [M + Na]: 259.1422, found: 259.1433.

7.2.18 Synthesis of N-tert-butoxycarbonyl-2-(4-N-(5-carboxy-1,1,3,3-

tetramethylisoindoline-2-yloxyl)aminophenyl)ethylamine (126).

Compound 125 (244 mg, 1.03 mmol, 1.21 equiv) was added to a solution

containing 5-carboxy-1,1,3,3-tetramethylisoindoline-2-yloxyl (106) (200 mg,

0.853 mmol, 1 equiv), EDCI (174 mg, 0.904 mmol, 1.06 equiv), HOBt (126 mg,

0.929 mmol, 1.09 equiv), and DIPEA (160 µL, 0.895 mmol, 1.05 equiv) in dry

DMF (10 mL). The reaction mixture was stirred at room temperature for 16 h

and the reaction progress was followed by TLC analysis using (DCM / EtOAc,

7:3). The solution was quenched using water (30 mL) at 0–5 °C and the oily

reaction mixture was extracted in EtOAc (3 × 25 mL). The combined extracts

were washed with water (30 mL) and brine solution (30 mL) and dried over

200

anhydrous MgSO4. The solvent was evaporated under reduced pressure and

the resulting residue purified by silica gel column chromatography using a (8:2)

mixture of EtOAc and petroleum ether to give N-tert-butoxycarbonyl-2-(4-N-(5-

carboxy-1,1,3,3-tetramethylisoindoline-2-yloxyl)aminophenyl)ethylamine (126)

as a brownish-white solid (220 mg, 57%). Rf = 0.64 (DCM / EtOAc, 7:3).

1H NMR (400 MHz, CDCl3) δ ppm 1.47 (br. s., 9 H) 1.57 (br. s., 12 H) 2.82 (br.

s., 3 H) 3.42 (br. s., 4 H) 4.55 (br. s., 1 H) 7.62 (br. s., 3 H) 7.79 (br. s., 1 H)

Note that paramagnetic broadening by the nitroxide radical results in some

peaks not being detected or giving poor integration in the proton NMR

spectrum.

Melting point: 158–160 °C.

ESMS calcd for C26H34NaN3O4+ [M+Na]: 475.2440, found: 475.2450.

Elemental analysis: found C 68.58, H 7.51, N 8.88, expected C 69.00, H 7.57,

and N 9.28.

HPLC: 91%

Ʋmax (ATR-FTIR) cm-1: 3360 (NH), 3289 (NH), 2969 (alkylCH3); 2929 (aryl CH),

1689 (NC=O), 1662, 1599 (amide C=O), 1514 (H-NCOO), 1359 (O-C(CH3)3),

1319 and 1356 (NO), 1276-1248 (N-CO-O), 1168 (H-NCOO).

EPR (methanol): typical 3-line nitroxide signal g 1.9922, aN 1.4875 mT.

201

7.2.19 Synthesis of 2-(4-N-(5-carboxy-1,1,3,3-tetramethylisoindoline-2-

yloxyl) aminophenyl) ethylamine (127).

4 M HCl (0.75 mL) was added to a solution of compound 126 (100 mg, 0.94

mmol, 1.0 equiv) in a (1:1) mixture of 1, 4–dioxane (4.3 mL) and water (4.3 mL).

The reaction mixture was stirred for 2 h at 55–60 °C and the reaction progress

was followed by TLC analysis using DCM / EtOAc (7:3). Upon completion, the

solution was evaporated to half its volume under reduced pressure. Water (30

mL) was added and the resulting solution was basified with 28% aqueous

ammonia solution and extracted with EtOAc (3 x 25 mL). The combined

organic layers were washed with water (30 mL) and brine solution (30 mL), and

dried over anhydrous MgSO4, filtered. The filtrate was concentrated under

reduced pressure and the resulting residue purified by silica gel column

chromatography using a (8.9:1:0.1) mixture of CHCl3: methanol: 28% aqueous

ammonia solution to give (127) as a yellowish solid (95 mg, 91%), Rf = 0.26

(DCM / EtOAc, 7:3).

1H NMR (400 MHz, CDCl3) δ ppm 0.79 - 0.97 (m, 2 H) 1.28 (br. s., 13 H) 1.38 -

1.63 (m, 12 H) 2.06 (s, 2 H) 2.62 (s, 1 H) 2.68 - 2.86 (m, 3 H) 3.17 (br. s., 1 H)

3.81 (br. s., 2 H) 4.14 (d, J=6.90 Hz, 1 H) 7.21 (br. s., 3 H) 7.61 (br. s., 4 H).

Note that paramagnetic broadening by the nitroxide radical results in some

202

peaks not being detected or giving poor integration in the proton NMR

spectrum.

Ʋmax (ATR-FTIR) cm-1: 3285 (NH2), 2972 (alkylCH3); 2921(aryl CH), 1654

(C=O), 1598 and 1574 (H-NCO), 1514 (aryl C-C), 1356, and 1316 and (NO).

Melting point: 176–178 °C.

ESMS calcd for C21H27N3O2+ [M + H]: 353.2100, found: 353.2110.

EPR (methanol): typical 3-line nitroxide signal, g 1.9924, aN 1.4738 mT..

HPLC: 97%

7.2.20 Synthesis of (E)-tert-4-(3-(3,5-di-tert-butyl-4-

hydroxyphenyl)acrylamide)phenethylcarbamate (132).

3, 5-di tert-butyl-4-hydroxycinnamic acid (118) (135 mg, 0.486 mmol, 1.15

equiv) was added to a mixture of EDCI (93.5 mg, 0.486 mmol, 1.15 equiv),

HOBt (65.6 mg, 0.486 mmol, 1.15 equiv), DIPEA (85 µL, 0.486 mmol, 1.15

equiv), and compound 125 (100 mg, 0.423 mmol, 1 equiv) in anhydrous DMF (2

mL). The reaction mixture was stirred to room temperature for 21 h and the

reaction progress was followed by TLC analysis in IPA/ MeOH (8:2). DCM was

added in to the reaction mixture and the solution was quenched by using water

(30 mL) at 0–5 °C. The resulting mixture was extracted in DCM (3 × 25 mL).

203

The combined extracts were washed with water (3 × 25 mL) and brine solution

(2 × 25 mL) and dried over anhydrous MgSO4, filtered. The solvent was

evaporated under reduced pressure and the oily residue was purified by silica

gel column chromatography using a (8:2) mixture of isopropanol: methanol to

give (E)-tert-4-(3-(3,5-di-tert-butyl-4-

hydroxyphenyl)acrylamide)phenethylcarbamate (132) as a brownish-oil (208

mg, 99.5 %), Rf = 0.78 (CHCl3/ MeOH, 8:2),

1H NMR (400 MHz, CDCl3) δ ppm: 1.40 - 1.50 (m, 18 H) 1.60 (br. s., 9H) 2.73 -

2.81 (m, 2 H) 3.36 (d, J=5.53 Hz, 2 H) 5.30 (s, 1 H) 5.49 (s, 1 H) 6.39 (d,

J=15.41 Hz, 1 H) 7.17 (d, J=8.27 Hz, 2 H) 7.29 (s, 1 H) 7.39 (s, 2 H) 7.55 (d,

J=8.17 Hz, 1 H) 7.70 (d, J=15.41 Hz, 1 H).

13C NMR (101 MHz, CDCl3) δ ppm: 28.4, 30.2, 34.3, 35.6, 41.8, 117.3, 120.1,

125.3, 125.9, 129.4, 136.3, 143.4, and 155.9.

Ʋmax (ATR-FTIR) cm-1: 3624 (NH2), 3282 (OH), 2956 (alkyl CH3), 1690 (NC=O),

1619 (C=C), 1511, 1425 (aryl C-C), 1390, 1365 (tBu-O), 1238 (H-NCO), 1168,

978, 849.

Melting Point: 126–128°C.

HRMS (ESI): m/z calcd for C30H42NaN2O4+ [M+Na]: 517.3042, found: 517.3006.

204

7.2.21 Synthesis of (E)-N-(4-(2-aminoethyl)phenyl)-3-(3,5-di-tert-butyl-4-

hydroxyphenyl)acrylamide (133).

4 M HCl (2 mL) was slowly added to a stirring solution of compound 33 (205

mg, 0.414 mmol, 1 equiv) in a (1:1) mixture of 1, 4-dioxane (9 mL) and water (9

mL). The solution was stirred for 2 h at 55–60 °C and the reaction progress

was followed by TLC analysis in CHCl3/ MeOH (8:2). Upon completion, the

solution was treated with water (25 mL) and basified up to pH 14 by using 28%

aqueous ammonia solution. The resulting mixture was extracted with EtOAc (3

x 25 mL). The combined organic phases were washed with water (3 x 25 mL)

and brine solution (2 x 25 mL) and dried over anhydrous Na2SO4, filtered. The

filtrate was evaporated under reduced pressure and the resulting residue was

purified by silica gel column chromatography using a mixture of CHCl3: MeOH:

28% aqueous ammonia (7.9:2:0.1) solution to give (E)-N-(4-(2-

aminoethyl)phenyl)-3-(3,5-di-tert-butyl-4-hydroxyphenyl)acrylamide (133) as a

yellowish-green solid (105 mg, 65%), Rf = 0.13 (CHCl3/ MeOH, 8:2).

1H NMR (400 MHz, CDCl3) δ ppm: 1.43 (s, 18 H) 2.70 - 2.74 (m, 2 H) 2.95 (br.

s., 2 H) 5.30 (s, 1 H) 6.48 (d, J=15.41 Hz, 1 H) 7.11 (d, J=7.24 Hz, 2 H) 7.37 (s,

2 H) 7.49 (d, J=6.94 Hz, 2 H) 7.70 (d, J=15.36 Hz, 1 H).

205

13C NMR (101 MHz, CDCl3): δ ppm: 30.1, 34.3, 55.9, 63.9, 125.0, 125.3, 126.0,

129.2, 136.2, 136.6 137.9, 143.2 146.2, 153.5 and 155.8.

Ʋmax (ATR-FTIR) cm-1: 3624 (NH2), 2955 (alkyl CH3), 1660 (NC=O), 1617

(C=C), 1513, 1421 (aryl C-C), 1237 (H-NCO), 1174, 979, 848, 771, 565, 508.

Melting point: 130–132 °C.

HRMS (ESI): m/z calcd for C25H35N2O2+ [M+H]: 395.2699, found: 395.2651.

7.2.22 Synthesis of 2,3-O-isopropylideneguanosine (139).288

Guanosine 138 (10.00 g, 35.3 mmol, 1.0 equiv) and para-toluenesulfonic acid

monohydrate (6.72 g, 35.3 mmol, 1.0 equiv) were added to a stirring solution of

acetone (350 mL) and 2, 2-dimethoxypropane (88.7 mL, 723 mmol, 20.5 equiv).

The reaction was stirred overnight at 25 °C. The reaction mixture was

evaporated to dryness and water (250 mL) and saturated NaHCO3 solution (70

mL) was added. The mixture was stirred for a further 1 h. The resulting

suspension was filtered and washed with cold water (250 mL) to give 2,3-O-

isopropylideneguanosine (139) as a white solid (8.6 g, 75%), Rf = 0.72 (EtOAc /

MeOH, 7:3).

1H NMR (400 MHz, DMSO-d6) δ ppm:1.34 (s, 3 H) 1.49 (s, 3 H) 3.49- 3.55 (m,

2H) 4.10- 4.15 (m, 1 H) 4.97 (dd, J=6.31, 3.08, 1 H) 5.05 (br. s., 1 H) 5.19(dd,

206

J=6.24, 2.86 Hz, 1 H) 5.93 (d, J=2.79, 1 H) 6.52 (s, 2 H) 7.92 (s, 1 H) 10.70 (s,

1 H).

13C NMR (101 MHz, DMSO-d6) δ ppm: 25.2, 27.0, 62.1, 81.2, 83.6, 86.6, 88.4,

113.0, 116.7, 135.8, 150.7, 153.7, 156.7.

Melting point: 260–262 °C (lit.259 260–262 °C)

HRMS (ESI): m/z calcd for C13H18N5O5+ [M + H]: 324.1300, found: 324.1286.

(The obtained spectroscopic data was consistent with that previously reported

for this compound).259

7.2.23 Synthesis of 2,3-O-isopropylideneguanosine-5-carboxylic acid

(140).259

Compound 139 (6.7 g, 21 mmol, 1 equiv), TEMPO (809 mg, 5.25 mmol, 0.25

equiv) and bis (acetoxy) iodobenzene (BAIB) (13.5 g, 42 mmol, 2.25 equiv)

were combined in MeCN/ water (1:1, 88 mL). The reaction mixture was stirred

overnight at 25 °C and the reaction progress was followed by TLC analysis

using a EtOAc / MeOH (4:1). After which time, acetone (160 mL) was added

and the solution was stirred for 1 h. The solution was poured into a well-stirred

volume of Et2O (930 mL) and the mixture was stirred for a further 1 h. The

resulting precipitate was filtered to give 2,3-O-isopropylideneguanosine-5-

carboxylic acid (140) as a pale orange solid (5.5 g, 78.8%). Rf = 0.16 (4:1

EtOAc / MeOH).

207

1H NMR (400 MHz, DMSO- d6) δ ppm: 1.34 (s, 3 H) 1.49 (s, 3 H) 4.61 (s, 1 H)

5.31 (d, J=6.02 Hz, 1 H) 5.56 (d, J=6.02 Hz, 1 H) 6.10 (s, 1 H) 6.46 (br. s., 2 H)

7.80 (s, 1 H) 10.68 (s, 1 H).

13C NMR (101 MHz, DMSO- d6) δ ppm: 25.4, 26.9, 84.0, 84.2, 85.9, 89.5,

112.9, 116.9, 137.1, 151.3, 153.8, 157.2, and 171.4.

Melting point: 210–212 °C (lit.259 210–212 °C).

HRMS (ESI): m/z calcd for C13H16N5O6+ [M + H]: 338.1101, found: 338.1134.

(The obtained spectroscopic data was consistent with that previously reported

for this compound).259

7.2.24 Synthesis of 2,3-O-isopropylideneguanosine-5-ethylcarboxylate

(142).

A suspension of 140 (5 g, 15.7 mmol, 1 equiv) in EtOH (500 mL) was stirred at

0 °C for 30 minutes. SOCl2 (4.5 mL, 67.6 mmol, 4.3 equiv) was slowly added.

The reaction mixture was warmed to room temperature and stirred overnight

and the reaction progress was followed by TLC analysis using (7:3) EtOAc /

MeOH. The reaction mixture was basified up to pH 8 by adding saturated

NaHCO3 solution and the resulting mixture was filtered. The filtrate was

evaporated under reduced pressure. The resulting residue was purified by

silica gel column chromatography using a mixture of EtOAc / MeOH (7:3) to

208

give 2,3-O-isopropylideneguanosine-5-ethylcarboxylate (142) as an off-white

solid (4.5 g, 84%). Rf = 0.71 (7:3 EtOAc / MeOH).

1H NMR (400 MHz, DMSO-d6) δ ppm: 0.98 (t, J=7.19 Hz, 3 H) 1.35 (s, 3 H) 1.49

(s, 3 H) 3.71 - 3.81 (m, 1 H) 3.82 - 3.91 (m, 1 H) 4.74 (s, 1 H) 5.34 (d, J=6.02

Hz, 1 H) 5.72 (d, J=5.87 Hz, 1 H) 6.18 (s, 1 H) 6.43 (br. s., 2 H) 7.78 (s, 1 H)

10.69 (br. s., 1 H).

13C NMR (101 MHz, DMSO-d6) δ ppm: 13.8, 25.3, 26.7, 61.1, 83.9, 84.8, 86.3,

89.9, 112.7, 117.1, 137.7, 151.1, 153.7, 157.2 and 169.8.

Ʋmax (ATR-FTIR) cm-1: 3413 (NH2), 3162 (NH), 2731 (alkyl CH3), 1696 (OC=O),

1628, 1596 (aryl C-N), 1540, 1488 (aryl C-C), 1382 (Et-O), 1271, 1205, 1157,

1057 (C-O-C), 866, 782.

Melting point: 272–274 °C.

HRMS (ESI): m/z calcd for C15H20N5O6+ [M + H]: 366.1414, found: 366.1449.

HPLC purity: - 99.99 %.

Elemental analysis: found C 49.30, H 5.17, and N 19.06. Expected C 49.31, H

5.24, and N 19.17

209

7.2.25 Synthesis of 2,3-O-isopropylideneguanosine-5-N-

ethylcarboxamide (143)259.

Compound 142 (774 mg, 2.12 mmol, 1 equiv) and ethylamine (22 mL) were

stirred at 0 to –20 °C for 3 h. The reaction progress was followed by TLC

analysis using (8:2 DCM: MeOH) and after which time the solution was allowed

to warm to room temperature and stirred overnight and the reaction progress

was followed by TLC analysis using a DCM / MeOH (7:3). The reaction mixture

was added to a stirring Et2O (50 mL) and the resulting mixture was stirred for 1

h, after which time the obtained precipitate was filtered and washed with Et2O

(25 mL). The resulting residue was purified by silica gel column

chromatography using a mixture of Pet Spirits/ EtOAc (2:8) to give 2,3-O-

isopropylideneguanosine-5-N-ethylcarboxamide (143) as a bluish-white solid

(725 mg, 94%), Rf = 0.85 (DCM / MeOH, 8:2).

1H NMR (400 MHz, DMSO-d6) δ ppm: 0.75 (t, J=7.19 Hz, 3 H) 1.33 (s, 3 H) 1.51

(s, 3 H) 2.78 - 2.90 (m, 1 H) 2.90 - 3.01 (m, 1 H) 4.47 (d, J=2.20 Hz, 1 H) 5.27

(dd, J=6.09, 1.25 Hz, 3 H) 5.44 (dd, J=6.09, 2.27 Hz, 3 H) 6.13 (s, 1 H) 6.42 (br.

s., 2 H) 7.37 (t, J=5.80 Hz, 1 H) 7.81 (s, 3 H) 10.65 (br s, 2 H).

13C NMR (101 MHz, DMSO-d6) δ ppm: 14.7, 15.8, 17.5, 25.4, 75.9, 76.0, 80.1,

82.9, 105.1, 108.6, 130.3, 142.8, 145.6, 149.9, and 162.5.

Melting point: 278–280 °C (lit.259 199–200 °C).

210

HRMS (ESI): m/z calcd for C15H21N6O5+ [M + H]: 365.1573, found: 365.1574.

(The obtained spectroscopic data was consistent with that previously reported

for this compound).259

7.2.26 Synthesis of O6-(benzotriazol-1-yl)-2′,3′-O-isopropylideneguanosine-

5′-N-ethylcarboxamide (144).259

Compound 143 (285 mg, 0.78 mmol, 1 equiv) was suspended in MeCN (27

mL).and BOP (554 mg, 1.25 mmol, 1.6 equiv) and DBU (187 µL, 1.25 mmol, 1.6

equiv) were added. The reaction mixture was stirred at room temperature for

20 h and the reaction progress was followed by TLC analysis using (Pet Spirits:

EtOAc, 1:4). The reaction mixture was diluted with EtOAc (90 mL) and the

resulting solution was washed with water (3 x 90 mL) and brine solution (90

mL). The EtOAc layer was filtered and dried over anhydrous MgSO4. The

solvent was evaporated under reduced pressure and the resulting residue was

purified via silica gel column chromatography using a gradient from (1:1)

petroleum spirits/ EtOAc to (1:4) petroleum spirits/ EtOAc to give O6-

(benzotriazol-1-yl)-2′,3′-O-isopropylideneguanosine-5′-N-ethylcarboxamide

(144) as a pale yellow solid (500 mg, 98%), Rf = 0.95 (Pet Spirits / EtOAc, 1:4).

211

1H NMR (400 MHz, DMSO-d6): δ ppm 0.67 (t, J=7.19 Hz, 3 H) 1.35 (s, 3 H) 1.52

(s, 3 H) 2.71 - 2.98 (m, 2 H) 4.53 (s, 1 H) 5.40 (d, J=6.16 Hz, 1 H) 5.52 (d,

J=6.02 Hz, 1 H) 6.29 (s, 1 H) 6.68 (s, 2 H) 7.48 (t, J=5.72 Hz, 1 H) 7.51 - 7.58

(m, 1 H) 7.66 (d, J=3.81 Hz, 2 H) 8.18 (d, J=8.51 Hz, 1 H) 8.21 (s, 1 H).

13C NMR (101 MHz, DMSO-d6) δ ppm: 14.4, 25.4, 27.0, 33.5, 83.6, 87.0, 90.5,

109.5, 113.4, 120.6, 126.1, 128.6, 130.2, 143.1, 147.4, 155.0, 156.1, 156.2,

159.8, 160.0 and 168.4.

Melting point: 140–142 °C (lit.259 140–141 °C).

HRMS (ESI): m/z calcd for C21H24N9O5+ [M+H]: 482.1900, found: 482.1898.

(The obtained spectroscopic data was consistent with that previously reported

for this compound)259.

7.2.27 Synthesis of 2-fluoro-O6-benzotriazole-1-yl-2,3-O-

isopropylideneguanosine-5-N-ethylcarboxamide (145).259

Pyridine (2.0 mL) was cooled to –50 °C and 70% HF in pyridine (6.5 mL) was

added slowly followed by compound 144 (1 g, 2.08 mmol, 1 equiv) with vigorous

stirring. The temperature was allowed to rise up to –30 °C and the reaction

mixture was stirred to dissolve all solid. Tert-butylnitrite (2.1 mL, 17.68 mmol,

8.5 equiv) was added dropwise into the reaction mixture and the mixture was

212

stirred at –30 °C for a further 15 minutes. The reaction progress was followed

by TLC analysis using (2:8 Pet Spirits: EtOAc). The reaction mixture was

poured onto iced water (200 mL) and stirred for 30 minutes. The resulting

suspension was extracted in DCM (3 x 50 mL) and the combined DCM extracts

were washed with water (3 x 50 mL) and brine solution (3 x 50 mL). The DCM

extract was dried over anhydrous MgSO4, filtered. The filtrate was evaporated

under reduced pressure and the resulting residue was purified by silica gel

column chromatography using a mixture of Pet Spirits / EtOAc (2:8) to give 2-

fluoro-O6-benzotriazole-1-yl-2,3-O-isopropylideneguanosine-5-N-

ethylcarboxamide (145) as an off-white solid (0.68 g, 68%). Rf = 0.52 (2:8 Pet

Spirits / EtOAc).

1H NMR (400 MHz, DMSO-d6): δ ppm 0.65 (t, J=7.12 Hz, 3 H) 1.36 (s, 3 H) 1.54

(s, 3 H) 2.66 - 2.98 (m, 2 H) 4.65 (s, 1 H) 5.38 (d, J=4.40 Hz, 1 H) 5.45 (d,

J=5.87 Hz, 1 H) 6.46 (s, 1 H) 7.59 (t, J=6.0 Hz, 1 H) 7.66 - 7.82 (m, 3 H) 8.24

(d, J=8.36 Hz, 1 H) 8.78 (s, 1 H).

13C NMR (101 MHz, DMSO-d6) δ ppm: 14.4, 25.5, 27.0, 33.5, 83.7, 87.1, 90.6,

109.6, 113.5, 118.2, 120.6, 126.1, 128.7, 130.2, 143.2, 147.5, 155.0, 157.1,

159.9, 168.4.

Melting point: 145–147 °C (decomp) (lit.259 140–141 °C).

HRMS (ESI): m/z calcd for C21H22FN8O5+ [M+H]: 485.1692, found: 485.1699.

(The obtained spectroscopic data was consistent with that previously reported

for this compound).259

213

7.2.28 Synthesis of 2-fluoro-2,3-O-isopropylideneadenosine-5-N-

ethylcarboxamide (146).259

A solution of compound 145 (700 mg, 1.24 mmol. 1 equiv)) in MeCN (9 mL) was

cooled at 0 °C in ice bath and 28% ammonia solution (500 µL) was cautiously

added to the stirring solution. The reaction mixture was allowed to rise to room

temperature and the stirring was continued for 2 h and the reaction progress

was followed by TLC analysis using EtOAc: MeOH (7:3). The reaction mass

was partitioned between water (50 mL) and EtOAc (50 mL). The aqueous

phase was extracted with EtOAc (3 x 30 mL) and the combined organic phases

were washed with water (3 x 50 mL) and brine solution (3 x 50 mL) and dried

over anhydrous MgSO4, filtered. The filtrate was evaporated under reduced

pressure. The resulting residue was purified by silica gel column

chromatography using EtOAc as the eluent to give 2-fluoro-2,3-O-

isopropylideneadenosine-5-N-ethylcarboxamide (146) as a pale yellow solid

(450 mg, 85%). Rf = 0.63 (EtOAc / MeOH, 7:3).

1H NMR (400 MHz, DMSO-d6): δ ppm 0.66 (t, J=7.19 Hz, 3 H) 1.34 (s, 3 H) 1.53

(s, 3 H) 2.72 - 2.97 (m, 2 H) 4.54 (s, 1 H) 5.36 (br s, 2 H) 6.27 (s, 1 H) 7.55 (t,

J=5.58 Hz, 1 H) 7.75 (br s, 2 H) 8.23 (s, 1 H).

214

13C NMR (101 MHz, DMSO-d6) δ ppm: 14.4, 25.5, 27.1, 33.5, 83.5, 83.7, 86.5,

89.8, 113.3, 117.9, 141.0, and 150.6 (d, J = 20.1 Hz), 158.0 (d, J = 21.1 Hz),

158.2, 168.5.

Melting point: 194–196 °C (lit 259 194–196 °C).

HRMS (ESI): m/z calcd for C15H19FN6O4+ [M+]: 366.1500, found: 366.1451. (The

obtained spectroscopic data was consistent with that previously reported for this

compound).259

7.2.29 Synthesis of 2-iodo-O6-benzotriazole-1-yl-2,3-O-

isopropylideneguanosine-5-N-ethylcarboxamide (148)259.

Tert-butylnitrite (0.2 mL, 10.39 mmol, 4 equiv) was added to a mixture of

compound 144 (200 mg, 0.415 mmol, 1 equiv) and diiodomethane (1 mL, 49.92

mmol, 30 equiv) in MeCN (3 mL). The reaction mixture was vigorously stirred at

65–70 °C for 4 h under argon atmosphere and the reaction progress was

followed by TLC analysis using DCM: MeOH (9:1). The reaction solution was

diluted with EtOAc (50 mL), and washed with water (3 x 50 mL), and brine

solution (3 x 50 mL) and dried over anhydrous MgSO4. The filtrate solvent was

evaporated under reduced pressure and the resulting residue was purified by

silica gel column chromatography using a mixture of DCM and MeOH (9:1) to

215

give 2-iodo-O6-benzotriazole-1-yl-2,3-O-isopropylideneguanosine-5-N-

ethylcarboxamide (148) as a brown solid (212 mg, 86%). Rf = 0.42 (DCM /

MeOH, 9:1).

1H NMR (400 MHz, DMSO-d6) δ ppm: 0.64 (t, J=7.19 Hz, 3 H) 1.33 (s, 3 H) 1.50

(s, 3 H) 2.54 - 2.59 (m, 2 H) 4.58 (d, J=2.20 Hz, 1 H) 5.38 (m, 2 H) 6.44 (br. s., 1

H) 7.53 - 7.57 (m, 2 H) 7.64 - 7.67 (m, 2 H) 8.17- 8.20 (m, 1 H) 8.63 (s, 1 H).

13C NMR (101 MHz, DMSO-d6) δ ppm: 14.7, 25.1, 27.0, 30.9, 34.2, 82.9, 83.2,

86.3, 91.6, 108.3, 114.9, 116.3, 120.0, 120.7, 125.0, 128.6, 129.0, 143.4, 143.9,

154.2, 157.9 and 167.8.

Melting point: 194–196 °C (lit 259 195–196 °C).

HRMS (ESI): m/z calcd for C21H21INaN8O5+ [M+Na]: 615.0577, found:

615.0808. (The obtained spectroscopic data was consistent with that previously

reported for this compound).259

7.2.30 Synthesis of 2-iodo-2,3-O-isopropylideneadenosine-5-N-

ethylcarboxamide (149).259

An aqueous ammonia (28%) solution (1.1 mL) was added to the solution of

compound 148 (652 mg, 1.1 mmol, 1 equiv) in MeCN (52 mL) at 25 °C. The

solution was stirred for three days at room temperature, and the reaction

216

progress was followed by TLC analysis using EtOAc: MeOH (7:3). After which

time, the reaction mass was partitioned between water (50 mL) and EtOAc (50

mL). The aqueous phase was extracted with EtOAc (4 x 70 mL). The

combined organic phases were washed with water (3 x 50 mL), and brine

solution (4 x 50 mL) and dried over anhydrous MgSO4. The organic phase was

filtered, and the solvent was evaporated under reduced pressure. The resulting

residue was purified by silica gel column chromatography using 100% EtOAc to

give 2-iodo-2,3-O-isopropylideneadenosine-5-N-ethylcarboxamide (149) as a

pale yellow solid (400 mg, 77%), Rf = 0.63 (EtOAc / MeOH, 7:3).

1H NMR (400 MHz, CDCl3) δ ppm 0.92 (t, J=7.24 Hz, 3 H) 1.37 (s, 3 H) 1.59 (s,

3 H) 3.11 - 3.35 (m, 2 H) 4.66 (d, J=1.81 Hz, 2 H) 5.27 (d, J=2.74 Hz, 1 H) 5.28

(d, J=2.64 Hz, 1 H) 5.32 (d, J=1.96 Hz, 1 H) 5.33 (d, J=2.01 Hz, 1 H) 6.00 (d,

J=2.69 Hz, 2 H) 6.02 (br. s., 2 H) 6.48 (br. s., 1 H) 7.74 (s, 1 H).

13C NMR (101 MHz, CDCl3) δ ppm: 14.7, 25.2, 27.1, 30.9, 34.3, 82.6, 83.4,

85.9, 91.5, 114.7, 120.1, 139.8, 149.7, 155.2, and 168.2.

Ʋmax (ATR-FTIR) cm-1: 3390 (NH2), 3308 (NH), 3168, 3079 (aryl C-H), 2982

(alkyl C-H), 1647 (NC=O), 1586 (aryl C-N), 1520 (aryl C-C), 1456 (N-CO), 1256

(H-NCO) 1183, 1091 (C-O-C), 1052.

Melting point: 196–198 °C(lit 259 196–198 °C).

HRMS (ESI): m/z calcd for C15H19INaN6O4+ [M+Na]: 497.0410, found:

497.0390.

217

7.2.31 Synthesis of 2-(2-(4-aminophenyl))ethylamino-2,3-O-

isopropylideneadenosine -5-N-ethylcarboxamide (159).

Approach 1

Compound 146 (150 mg, 0.41 mmol, 1 equiv) was combined with compound

124 (93 mg, 0.389 mmol, 1.25 equiv), and DIPEA (1.1 mL) in absolute EtOH

(3.0 mL). The reaction mixture was stirred at 70–75 °C for 7 days, after which

time the reaction mixture was treated with DM water at 0–5 °C. The resulting

oily reaction mixture was extracted in DCM (4 x 30 mL), and the combined DCM

extract was washed with water (50 mL), saturated NaHCO3 solution (50 mL)

and brine solution (50 mL). The solvent was dried over anhydrous MgSO4 and

evaporated under reduced pressure. The resulting residue was purified by

silica gel column chromatography using a mixture of 75% MeOH and 25%

water, to give 2-(2-(4-aminophenyl))ethylamino-2,3-O-

isopropylideneadenosine-5-N-ethylcarboxamide (159) as a yellowish solid.

Approach 2

Compound 146 (400 mg, 0.33 mmol, 1 equiv), 4-(2-aminoethyl)anilne 124 (83

mg, 0.69 mmol, 2 equiv), and DIPEA (1.5 mL) in MeCN (36 mL) was stirred in a

218

sealed reaction vessel at 180 °C in microwave conditions for 7 h. The reaction

progress was followed by TLC analysis in a mixture of (9.9:0.1) EtOAc and 28%

aqueous ammonia solution. After which time, the reaction mixture was

evaporated under reduced pressure. The resulting residue was purified by

silica gel column chromatography using a mixture of (9.9:0.1) EtOAc and 28%

aqueous ammonia solution to give 2-(2-(4-aminophenyl))ethylamino-2,3-O-

isopropylideneadenosine-5-N-ethylcarboxamide (159) as a yellowish solid (510

mg, 97%), Rf = 0.32 (EtOAc/ 28% NH3 solution, 9.5:0.5).

1H NMR (400 MHz, CDCl3) :δ ppm 0.71 (t, J=7.26 Hz, 3 H) 1.40 (s, 3 H) 1.62 (

s., 3 H) 2.82 (t, J=6.82 Hz, 2 H) 2.86 - 3.08 (m, 2 H) 3.45 - 3.61 (m, 1 H) 3.62 -

3.72 (m, 1 H) 4.68 (s, 1 H) 4.85 (br. s., 1 H) 5.26 (br. s., 2 H) 5.54 (d, J=6 Hz, 1

H) 5.63 (d, J=4.84 Hz, 1 H) 6.04 (br. s., 2 H) 6.67 (d, J=8.22 Hz, 2 H) 7.06 (d,

J=8.07 Hz, 2 H) 7.53 (s, 1 H).

13C NMR (101 MHz, CDCl3) δ ppm: 13.9, 20.1, 25.0, 26.7, 33.8, 34.8, 83.3,

84.0, 87.7, 91.4, 105.7, 113.7, 115.4, 129.3, 129.6, 137.0, 144.7, 151.3, 155.6,

159.5 and 168.9.

Ʋmax (ATR-FTIR) cm-1: 3350 (NH2), 3292 (NH), 2968 (alkyl CH3), 1689 (NC=O),

1662, 1602 (aryl C-N), 1513 (aryl C-C), 1437 (N-CO), 1364, 1253 (H-NCO),

1170 (C-O-C).

Melting point: 150–152 °C (lit 121 95–104 °C).

HRMS (ESI): m/z calcd for C23H31N8O4+ [M+H]: 483.2460, found: 483.2482.

(The obtained spectroscopic data was consistent with that previously reported

for this compound).121

219

7.2.32 Synthesis of 2-(2-(4-N-(5-carboxy-1,1,3,3-tetramethylisoindoline-2-

yloxyl)aminophenyl))ethylamino-2,3-O-isopropylideneadenosine -

5-N-ethylcarboxamide (161).

Approach 1

Compound 146 (25 mg, 0.068 mmol, 1 equiv) was combined with compound

127 (51 mg, 0.143 mmol, 2.1 equiv), and DIPEA (75 µL) in absolute EtOH (1.5

mL). The reaction mixture was stirred at 70–75 °C for 7 days. The reaction

mixture was cooled to 0–5 °C and treated with DM water, and the resulting oily

mixture was extracted in DCM (4 x 30 mL). The combined organic layer was

washed with water (30 mL) and saturated NaHCO3 solution (30 mL) and brine

solution (30 mL). The solvent was dried over anhydrous MgSO4 and

evaporated under reduced pressure. The resulting residue was purified by

MPLC instrument using a mixture of 75% MeOH and 25% water, to give 2-(2-(4-

N-(5-carboxy-1,1,3,3-tetramethylisoindoline-2-yloxyl)aminophenyl))ethylamino-

2,3-O-isopropylideneadenosine-5-N-ethylcarboxamide (161) as a pale yellow

solid (40 mg, 58%).

220

Approach 2

Compound 159 (143 mg, 0.311mmol, 1 equiv) was added to a mixture

containing 5-carboxy-1,1,3,3-tetramethylisoindoline-1-yloxyl 106 (88 mg, 0.37

mmol, 1.25 equiv), EDCI (96 mg, 0.356 mmol, 1.2 equiv), HOBt (100 mg, 0.356

mmol, 1.2 equiv), and DIPEA (1 mL) were combined in dry DMF (3 mL) under

argon atmosphere. The reaction mixture was stirred at room temperature for 2

days. The solution was quenched by using water (30 mL) at 0–5 °C and the

resulting oily reaction mixture was extracted in DCM (4 x 30 mL). The

combined DCM extract was washed with water (30 mL), saturated NaHCO3

solution (30 mL) and brine solution (30 mL). The solvent was evaporated under

reduced pressure and the resulting residue was purified by MPLC instrument

using a mixture of 75% MeOH and 25% water to give 2-(2-(4-N-(5-carboxy-

1,1,3,3-tetramethylisoindoline-2-yloxyl)aminophenyl))ethylamino-2,3-O-

isopropylideneadenosine-5-N-ethylcarboxamide (161) as a pale yellow solid

(168 mg, 81%), Rf = 0.48 (DCM / IPA/ MeOH, 4.5:4.5:1).

1H NMR (400 MHz, CDCl3) δ ppm: 0.71 (t, 3 H) 1.23 (d, J=6.13 Hz, 12 H) 1.42

(s, 3 H) 1.62 (s, 3 H) 2.91 (s, 2 H) 2.98 (s, 2 H) 4.69 - 4.70 (m, 1 H) 5.32 (s, 1 H)

6.05 - 6.07 (m, 1 H) 6.24 - 6.26 (m, 1 H) 7.00 - 7.03 (m, 1 H) 7.55 (s, 1 H) 8.03 -

8.05 (m, 1 H) Note that paramagnetic broadening by the nitroxide radical results

in some peaks not being detected or giving poor integration in the proton NMR

spectrum.

Melting point: 66–68 °C (decomp.)

HRMS (ESI): m/z calcd for C36H45N9O6+ [M + H]: 699.3493, found: 699.3506.

EPR (methanol): typical 3-line nitroxide signal, g 1.9823, aN 1.4871 mT.

221

HPLC purity- 97.64%.

7.2.33 Synthesis of 2-(2-(4-N-(5-carboxy-1,1,3,3-tetramethylisoindoline-1-

yloxyl)aminophenyl))ethylaminoadenosine-5-N-ethylcarboxamide

(162).

1 M HCl (3.3 mL) was added to a solution of compound 161 (101 mg, 0.94

mmol) in MeCN (8.0 mL). The solution was stirred for 4 h at 55–60 °C, and the

reaction progress was followed by TLC. Upon completion, the reaction mixture

was basified with saturated NaHCO3 solution up to pH 8 at 0–5 °C, the resulting

precipitate was extracted with EtOAc (3 x 25 mL). The combined organic phase

was washed with water (3 x 25 mL), brine solution (3 x 25 mL) and dried over

anhydrous MgSO4. The solvent was filtered and evaporated under reduced

pressure. The resulting residue was purified by MPLC instrument using a

mixture of 80% MeOH and 20% water with partial crystallisation to give 2-(2-(4-

N-(5-carboxy-1,1,3,3-tetramethylisoindoline-1-

yloxyl)aminophenyl))ethylaminoadenosine-5-N-ethylcarboxamide (162) as a

pale yellow solid (66 mg, 69.5%), Rf = 0.65 (EtOAc: MeOH, 8:2).

1H NMR (400 MHz, MeOH-d4) δ ppm: 1.00 - 1.12 (m, 3 H) 1.27 (s, 2 H) 2.12 -

2.25 (m, 1 H) 2.61 - 2.69 (m, 1 H) 2.86 - 3.00 (m, 1 H) 3.09 - 3.22 (m, 1 H) 3.33

(br. s., 20 H) 3.46 - 3.61 (m, 1 H) 3.63 - 3.77 (m, 1 H) 4.36 - 4.47 (m, 1 H) 4.49 -

4.55 (m, 1 H) 4.58 - 4.72 (m, 1 H) 4.87 (s, 74 H) 5.01 - 5.09 (m, 1 H) 5.90 - 6.02

222

(m, 1 H) 7.54 - 7.75 (m, 1 H) 7.93 - 8.04 (m, 1 H) 8.05 - 8.12 (m, 1 H) Note that

paramagnetic broadening by the nitroxide radical results in some peaks not

being detected or giving poor integration in the proton NMR spectra.

Ʋmax (ATR-FTIR) cm-1: 3400-3000 (NH2, NH, OH), 2926 (alkyl CH3), 1691

(NC=O), 1597 (aryl C-N), 1529 (aryl C-C), 1408 (OC-N), 1258 (H-NCO), 1044

(C-O-C).

Melting point: 241-243 °C (decomp).

HRMS (ESI): m/z calcd for C33H41N9O6+ [M+H]: 659.318, found: 659.3197.

EPR (methanol): typical 3-line nitroxide signal, g 1.9950, aN 1.4928 mT.

HPLC purity: >99.9 %.

Elemental analysis: found C 57.64, H 6.10, N 17.91. Expected C 60.17, H 6.12,

N 19.14

223

7.2.34 Synthesis of 2-(2-(4-N-(4-carboxy-2,2,6,6-tetramethylpiperidine-1-

yloxyl)aminophenyl))ethylamino-2,3,-O-isopropylidineadenosine-

5-N-ethylcarboxamide (163).

Compound 159 (300 mg, 0.622 mmol, 1 equiv), was added to a mixture

containing 4-carboxy-TEMPO-1-yloxyl 116(156 mg, 0.7789 mmol, 1.25 equiv),

EDCI (179 mg, 0.933 mmol, 1.5 equiv), and HOBt (101 mg, 0.746 mmol, 1.2

equiv), and DIPEA (2 mL) in dry DMF (10 mL) under argon atmosphere. The

resulting reaction mixture was stirred at room temperature for two days. The

solution was quenched by adding water (30 mL) at 0–5 °C and the resulting oily

mixture was extracted in DCM (3 x 30 mL). The combined extracts were

washed with water (2 x 30 mL), and saturated NaHCO3 solution (2 x 30 mL) and

brine solution (2 x 30 mL) and dried over anhydrous MgSO4. The solvent was

evaporated under reduced pressure. The resulting brownish residue was

purified by MPLC instrument using a mixture of 75% MeOH and 25% water to

give 2-(2-(4-N-(4-carboxy-2,2,6,6-tetramethylpiperidine-1-

yloxyl)aminophenyl))ethylamino-2,3,-O-isopropylidineadenosine-5-N-

ethylcarboxamide (163) as a yellowish solid (346 mg, 82%). Rf = 0.43 (IPA:

MeOH, 8:2).

224

Ʋmax (ATR-FTIR) cm-1: 3400-3000 (NH, NH2), 2976 (alkyl CH3), 1655 (NC=O),

1596 (aryl C-N), 1530 (aryl C-C), 1411 (N-CO), 1326 (N-O), 1239 (H-NCO)

1087 (C-O-C), 789.

Melting point: 148–150 °C decomp.

HRMS (ESI): m/z calcd for C33H47N9O6+ [M + H]: 665.364, found: 665.3658.

EPR (methanol): typical 3-line nitroxide signal, g 1.9823, aN 1.4871 mT.

HPLC purity: 99.38%.

Elemental analysis: found: C 59.58, H 7.35, and N 18.43. Expected: C 59.62, H

6.97, and N 18.96.

7.2.35 Synthesis of 2-(2-(4-N -(4- carboxy-2,2,6,6-tetramethylpiperidine-1-

yloxyl)aminophenyl))ethylaminoadenosine-5-N-ethylcarboxamide

(164).

1 M HCl (11 mL) was slowly added to a solution of compound 163 (177 mg,

0.267 mmol, 1 equiv) in MeCN (6.0 mL). The reaction mixture was stirred for 5

h at 55–60 °C, and the reaction progress was followed by TLC in IPA: MeOH

(8:2). Upon completion, the solution was basified up to pH 8 by adding

saturated NaHCO3 solution at 0–5 °C, and the resulting precipitate was

extracted with EtOAc (3 x 20 mL). The combined organic phase was washed

225

with water (3 x 20 mL), brine solution (3 x 20 mL) and dried over anhydrous

MgSO4, filtered. The solvent was evaporated under reduced pressure and the

resulting residue was purified by MPLC instrument using a mixture of 70%

MeOH and 30% water with a partial crystallisation to give 2-(2-(4-N-(4-carboxy-

2,2,6,6-tetramethylpiperidine-1-yloxyl)aminophenyl))ethylaminoadenosine-5-N-

ethylcarboxamide (164) as a pale yellow solid (100 mg, 60 %). Rf = 0.43 (IPA:

MeOH, 8:2).

Ʋmax (ATR-FTIR) cm-1: 3400-3100 (NH2, NH, OH), 2926 (alkyl CH3), 1691

(NC=O), 1650, 1600 (aryl C-N), 1532 (aryl C-C), 1411 (OC-N), 1240 (H-NCO),

1324 (N-O), 1044 (C-O-C).

Melting point: 211-213°C (decomp).

HRMS (ESI): m/z calcd for C30H43N9O6+ [M+H]: 625.333, found: 625.3341.

EPR (methanol): typical 3-line nitroxide signal, g 1.9893, aN 1.6011 mT.

HPLC Purity – <99.9 %.

Elemental analysis: found C 59.79, H 7.25, and N 19.32. Expected C 57.68, H

6.78, and N 20.18.

226

7.2.36 Synthesis of 2-(2-(4-N-(3-carboxy-2,2,5,5-tetramethylpyrrolidine-1-

yloxyl) aminophenyl)) ethylamino-2,3,-O-isopropylidineadenosine-

5-N-ethylcarboxamide (165).

3-Carboxy-PROXYL 117(145 mg, 0.778 mmol, 1.25 equiv) was added to a

stirring solution containing compound 159 (300 mg, 0.622 mmol, 1 equiv), EDCI

(179 mg, 0.933 mmol, 1.5 equiv), HOBt (101 mg, 0.746 mmol, 1.2 equiv), and

DIPEA (2 mL) were combined in dry DMF (10.5 mL) under argon atmosphere.

The reaction mixture was stirred at room temperature for two days. The

resulting solution was quenched by adding water (20 mL) at 0–5 °C and the

resulting oily reaction mixture was extracted in DCM (3 x 20 mL). The

combined extracts were washed with water (3 x 25 mL) and saturated NaHCO3

solution (3 x 25 mL) and brine solution (3 x 25 mL) and dried over anhydrous

MgSO4. The solvent was evaporated under reduced pressure and the resulting

residue was purified by MPLC instrument using a mixture of 75% MeOH and

25% water to give 2-(2-(4-N-(3-carboxy-2,2,5,5-tetramethylpyrrolidine-1-

yloxyl)aminophenyl))ethylamino-2,3,-O-isopropylidineadenosine-5-N-

ethylcarboxamide (165) as a yellowish-white solid (300 mg, 74 %), Rf = 0.29

(IPA / MeOH, 9:1).

Ʋmax (ATR-FTIR) cm-1: 3500-3000 (OH, NH, NH2), 2923 (alkyl CH3), 1670

(NC=O), 1600 (aryl C-N), 1532 (aryl C-C), 1411 (N-CO), 1356-1320 (N-O),

1195, 1087 (C-O-C), 789.

227

Melting point: 140–142 °C.

HRMS (ESI): m/z calcd for C32H45N9O6+ [M + H]: 651.349, found: 651.3493,

EPR (methanol): typical 3-line nitroxide signal, g 1.9808, aN 1.5239 mT.

HPLC purity- 97.96 %.

Elemental analysis: found C 59.57, H 7.30, and N 19.08. Expected C 59.06, H

6.82, and N 19.37.

7.2.37 Synthesis of 2-(2-(4-N -(3-carboxy-2,2,5,5-tetramethylpyrrolidine-1-

yloxyl) aminophenyl))ethylaminoadenosine-5-N-ethylcarboxamide

(166).

1 M HCl (3.3 mL) was added to a stirring solution of compound 165 (101 mg,

0.94 mmol, 1 equiv) in MeCN (8.0 mL). The reaction mixture was stirred for 4 h

at 55–60 °C, and the reaction progress was followed by TLC analysis using

(DCM: IPA: MeOH, 4:5:1). Upon completion, the solution was basified by using

saturated NaHCO3 solution up to pH 8 at 0–5 °C and the resulting precipitate

was extracted with EtOAc (4 x 20 mL). The organic phases were washed with

water (3 x 20 mL) and brine solution (3 x 20 mL) and dried over anhydrous

MgSO4, filtered. The solvent was evaporated under reduced pressure and the

resulting residue was purified by MPLC instrument using a mixture of 70%

methanol and 30% water with a partial crystallisation of solid to give 2-(2-(4-N-

(3-carboxy-2,2,5,5-tetramethylpyrrolidine-1-

228

yloxyl)aminophenyl))ethylaminoadenosine-5-N-ethylcarboxamide(166) as a

brownish white solid (65 mg, 62.5 %), Rf = 0.10 (DCM: IPA: MeOH, 4:5:1).

1H NMR (400 MHz, CD3OD) δ ppm: 0.97 - 1.11 (m, 3 H) 2.17 (s, 1 H) 2.82 -

3.01 (m, 1 H) 3.05 - 3.19 (m, 1 H) 3.33 (d, J=1.47 Hz, 18 H) 3.46 - 3.59 (m, 1 H)

3.61 - 3.74 (m, 1 H) 4.37 - 4.54 (m, 1 H) 4.97 - 5.12 (m, 1 H) 5.87 - 5.99 (m, 1

H) 7.14 - 7.33 (m, 1 H) 7.38 - 7.61 (m, 1 H) 7.91 - 8.04 (s, 1 H). Note that

paramagnetic broadening by the nitroxide radical results in some peaks not

being detected or giving poor integration in the proton NMR spectrum.

Ʋmax (ATR-FTIR) cm-1: 3322-3000 (OH, NH, NH2), 2926 (alkyl CH3), 1634

(NC=O), 1597 (aryl C-N), 1538 (aryl C-C), 1411 (N-CO), 1354 (N-O), 1255 (H-

NCO), 1048 (C-O-C).

Melting point: 176–178 °C (decomp).

HRMS (ESI): m/z calcd for C29H41N9O6+ [M+H]: 611.3184, found: 611.3183.

EPR (methanol): typical 3-line nitroxide signal, g 1.9939, aN 1.5006 mT.

HPLC purity- >99.9%

Elemental analysis: found C 56.40, H 7.19, and N 19.22. Expected C 57.04, H

6.60, and N 20.64.

229

.

7.2.38 .Synthesis of 2-(2-(4-N-(5-methylcarboxy-1,1,3,3-

tetramethylisoindoline-2-yloxyl)aminophenyl))ethylamino-2,3-O-

isopropylideneadenosine -5-N-ethylcarboxamide (167).

Compound 159 (100 mg, 0.207 mmol, 1 equiv), was added in a stirring mixture

of (60:40) 5-methylcarboxy-1,1,3,3-tetramethylisoindoline-2-yloxyl (115) and

compound 8 (65 mg, 0.259 mmol, 1.25 equiv), EDCI (64 mg, 0.33 mmol,

1.6equiv), HOBt (35 mg, 0.259 mmol, 1.25 equiv), and DIPEA (1.4 mL) in

anhydrous DMF (8 mL). The reaction mixture was stirred at room temperature

for 4 days and the reaction progress was followed by TLC analysis (EtOAc:

MeOH, 8:2). The solution was quenched by adding of water (50 mL) at 0–5 °C

and the oily reaction mixture was extracted in DCM (3 x 50 mL). The combined

extracts were washed with water (3 x 50 mL) and brine solution (3 x 50 mL).

The solvent was evaporated under reduced pressure. The resulting residue

was purified by MPLC instrument using a mixture of 75% MeOH and 25% water

to give a (60:40) mixture of 2-(2-(4-N-(5-methylcarboxy-1,1,3,3-

tetramethylisoindoline-2-yloxyl)aminophenyl))ethylamino-2,3-O-

isopropylideneadenosine-5-N-ethylcarboxamide (167) and compound (161) as

a pale yellow solid (150 mg, 71% w/w)

230

Melting point: 175–180 °C (decomp).

HRMS (ESI): m/z calcd for C37H47N9O6+ [M + H] 713.3649 and C36H45N9O6+:

699.3493, found: 713.5496 and 699.9465.

HPLC purity: 60:40 mixtures of compound 167 and 161

EPR (methanol): typical 3-line nitroxide signal, g 1.9912, aN 1.5084 mT.

(This mixture was hard to separate so taken as it is for further for next reaction)

7.2.39 Synthesis of 2-(2-(4-N-(methylcarboxy-1,1,3,3-

tetramethylisoindoline-2-

yloxyl)aminophenyl))ethylaminoadenosine-5-N-ethylcarboxamide

(168).

1 M HCl (3.3 mL) was added slowly in to a mixture of compound 167 and 161

(86 mg, 0.94 mmol, 1 equiv)) in MeCN (8.0 ml). The solution was stirred for 9 h

at 55–60 °C, and the reaction progress was followed by TLC analysis in (DCM:

IPA: MeOH, 4:5:1). Upon completion, the solution was basified up to pH 8 by

slow adding saturated NaHCO3 solution and, the resulting mixture was

extracted with EtOAc (5 x 50 mL). The EtOAc layers were washed with water

(3 x 25 mL) and brine solution (3 x 25 mL), and dried over anhydrous MgSO4,

filtered. The filtrate was evaporated under reduced pressure and the resulting

231

residue was purified by MPLC instrument using 70% MeOH and 30% water to

give a mixture of 2-(2-(4-N-(methylcarboxy-1,1,3,3-tetramethylisoindoline-2-

yloxyl)aminophenyl))ethylaminoadenosine-5-N-ethylcarboxamide (168) and

compound (162) as an off-white solid (74 mg,). Rf = 0.37 (DCM: MeOH, 9:1).

Ʋmax (ATR-FTIR) cm-1: 3400-3117 (OH, NH, NH2), 2973 (alkyl CH3), 2927, 1656

(NC=O), 1599 (aryl C-N), 1409 (N-CO), 1355 (N-O), 1256 (H-NCO), 1045 (C-O-

C).

Melting point: 175–180 °C (decomp).

HRMS (ESI): m/z calcd for C34H43N9O6+ and C33H41N9O6

+ [M+2H]: 673.3336

and 659 56 found: 674.6586.and 661.0205.

HPLC purity: 60:40 mixture.

EPR (methanol): typical 3-line nitroxide signal, g 1.9928, aN 1.4952 mT.

232

7.2.40 Synthesis of 2-(2-(4-N-(5-carboxy-1,1,3,3-tetraethylisoindoline-2-

yloxyl) aminophenyl)) ethylaminoadenosine-5-N-ethylcarboxamide

(169).

Compound 159 (150 mg, 0.311 mmol, 1 equiv) was added in a mixture

containing 5-carboxy-1, 1, 3, 3-tetraethylisoindoline-2-yloxyl (114) (113 mg,

0.389 mmol, 1.25 equiv), EDCI (95.4 mg, 0.498 mmol, 1.6 equiv), HOBt (53 mg,

0.389 mmol, 1.25 equiv), and DIPEA (1.2 mL) were combined in anhydrous

DMF (10 mL). The reaction mixture was stirred at room temperature for 19 h

and the reaction progress was followed by TLC analysis (DCM: MeOH, 9:1).

The solution was quenched by adding water at 0–5 °C and the resulting oily

reaction mixture was extracted in DCM (5 x 50 mL). The combined DCM

extracts were washed with water (3 x 25 mL) and saturated NaHCO3 solution (3

x 25 mL) and brine solution (2 x 25 mL) and dried over anhydrous MgSO4,

filtered. The solvent was evaporated under reduced pressure. The resulting

residue was purified by MPLC instrument using a mixture of 75% MeOH and

25% water to give 2-(2-(4-N-(5-carboxy-1,1,3,3-tetraethylisoindoline-2-

yloxyl)aminophenyl))ethylaminoadenosine-5-N-ethylcarboxamide (169) as a

pale yellow solid (101 mg, 65%).

Rf = 0.80 (DCM / MeOH, 9:1).

233

1H NMR (400 MHz, METHANOL-d4) δ ppm 0.54 - 0.68 (t, 3 H) 1.17 - 1.19 (d, 6

H) 1.53 - 1.63 (m, 1 H) 2.71 - 2.86 (m, 1 H) 2.86 - 3.03 (m, 1 H) 3.44 - 3.58 (m,

1 H) 3.67 - 3.82 (m, 1 H) 5.56 - 5.64 (m, 1 H) 5.73 - 5.81 (m, 1 H) 6.20 - 6.27

(m, 1 H) 7.29 - 7.39 (m, 1 H) 7.59 - 7.73 (m, 1 H) 7.83 - 7.90 (m, 1 H) Note that

paramagnetic broadening by the nitroxide radical results in some peaks not

being detected or giving poor integration in the proton NMR spectrum.

Ʋmax (ATR-FTIR) cm-1:3400-3000 (NH2 and NH), 2924 (alkyl CH3), 1645

(NC=O), 1593 (C-N), 1528 (aryl C-C), 1409 (OC-N), 1256 (H-NCO), 1088 (C-O-

C).

Melting point: 138–140 °C dec;

HRMS (ESI): m/z calcd for C40H53N9O6+ [M + H]: 755.4119, found: 755.4118.

HPLC purity – <95%.

EPR (methanol): typical 3-line nitroxide signal, g 2.0288, aN 1.4456 mT.

Elemental analysis: found C 66.59, H 8.03, and N 15.97. Expected C 63.64, H

6.94, and N 16.70.

234

7.2.41 Synthesis of 2-(2-(4-N-(5-carboxy-1,1,3,3-tetraethylisoindoline-2-

yloxyl)aminophenyl))ethylaminoadenosine-5-N-ethylcarboxamide

(170).

1M HCl (6.5 mL) added slowly in to a stirring solution of compound 169 (100

mg, 0.132 mmol, 1 equiv) in MeCN (3.0 mL). The solution was stirred for 5 h at

55–60 °C and the reaction progress was followed by TLC analysis (EtOAc:

MeOH, 8:2) indicated that all starting material had been consumed. Upon

completion, the solution was basified up to pH 8 with saturated NaHCO3

solution, and the resulting mixture was extracted with EtOAc (3 x 50 mL). The

combined organic phases were washed with water (3 x 25 mL) and brine

solution (3 x 25 mL) and dried over anhydrous MgSO4, filtered. The filtrate was

evaporated under reduced pressure. The resulting residue was purified by

MPLC instrument using a mixture of 75% MeOH and 25% water as the eluent to

give 2-(2-(4-N-(5-carboxy-1,1,3,3-tetraethylisoindoline-2-yloxyl)

aminophenyl))ethylaminoadenosine-5-N-ethylcarboxamide (170) as a pale

yellow solid (67 mg, 71%), Rf = 0.48 (EtOAc: MeOH, 8:2).

1H NMR (400 MHz, METHANOL-d4) δ ppm 1.00 - 1.20 (m, 12 H) 2.95 (br. s., 8

H) 3.22 (br. s., 4 H) 3.56 - 3.84 (m, 7 H) 4.47 (br. s., 9 H) 5.99 (br. s., 4 H) 7.31

(br. s., 8 H) 7.67 (br. s., 7 H) 8.11 (br. s., 3 H) Note that paramagnetic

235

broadening by the nitroxide radical results in some peaks not being detected or

giving poor integration in the proton NMR spectrum.

Melting point: 175–180 °C (decomp).

Ʋmax (ATR-FTIR) cm-1: 3286-3000 (OH, NH, NH2), 2962 and 2926 (alkyl CH3),

1622 (NC=O), 1591 (aryl C-N), 1515 (aryl C-C), 1407 (OC-N), 1320 (N-O), 1050

(C-O-C).

HRMS (ESI): m/z calcd for C37H48NaN9O6+ [M+ Na]: 737.3625, found:

737.3883.

HPLC purity- 98.5%.

EPR (methanol): typical 3-line nitroxide signal, g 1.9923, aN 1.4380 mT.

Elemental analysis: found C 59.58, H 7.35, and N 18.43. Expected C 59.62, H

6.97, and N 18.96.

236

7.2.42 Synthesis of 2-(2-(4-N-(3,5-di-tert-butyl-4-

hydroxyphenyl)acrylamido) phenyl))ethylamino-2,3-O-

isopropylideneadenosine-5-N-ethylcarboxamide(171).

3, 5-di tert-butyl-4-hydroxycinnamic acid (118) (65 mg, 0.486 mmol, 1.2 equiv)

was added to a mixture of EDCI (85 mg, 0.486 mmol, 1.2 equiv), HOBt (35 mg,

0.486 mmol, 1.2 equiv), DIPEA (85 µL, 0.486 mmol, 1.2 equiv), and compound

159 (100 mg, 0.207 mmol, 1 equiv) in anhydrous DMF (2 mL). The reaction

mixture was stirred to room temperature for 21 h and the reaction progress was

followed by TLC analysis in (EtOAc/ Hexanes, 9:1). DCM was added in to the

reaction mixture and the solution was quenched by using water (30 mL) at 0–5

°C. The resulting mixture was extracted in DCM (3 × 25 mL). The combined

extracts were washed with water (3 × 25 mL) and brine solution (2 × 25 mL) and

dried over anhydrous MgSO4, filtered. The solvent was evaporated under

reduced pressure and the oily residue was purified by silica gel column

chromatography using a (9:1) mixture of EtOAc/ Hexanes to give 2-(2-(4-N-(3,5-

di-tert-butyl-4-hydroxyphenyl)acrylamido) phenyl))ethylamino-2,3-O-

isopropylideneadenosine-5-N-ethylcarboxamide (171) as a brownish-solid (146

mg, 95%), Rf = 0.54 (IPA/ MeOH, 8:2).

237

1H NMR (400 MHz, CD3OD) δ ppm: 0.59 (t, J=7.21 Hz, 3 H) 0.84 - 0.92 (t,

J=7.21 Hz, 3 H) 1.28 (s, 6 H) 1.45 (s, 9 H) 1.51 - 1.58 (m, 2 H) 1.97 (s, 1 H)

2.01 (s, 1 H) 2.13 - 2.20 (m, 1 H) 2.30 (d, J=6.80 Hz, 1 H) 2.62 (s, 1 H) 2.76 (dd,

J=13.74, 7.14 Hz, 1 H) 2.85 (s, 1 H) 2.87 - 2.95 (m, 1 H) 2.99 (s, 1 H) 3.48 (d,

J=6.50 Hz, 1 H) 3.66 - 3.76 (m, 1 H) 4.55 - 4.62 (m, 1 H) 5.58 (d, J=5.09 Hz, 1

H) 5.73 (d, J=5.87 Hz, 1 H) 6.21 (s, 1 H) 6.50 - 6.55 (m, 1 H) 6.63 (d, J=15.80

Hz, 1 H) 6.95 (s, 1 H) 7.13 (s, 1 H) 7.26 (d, J=7.87 Hz, 1 H) 7.42 (s, 1 H) 7.53 -

7.63 (m, 1 H) 7.84 (br. s., 1 H) 7.97 (s, 1 H)

13C NMR (101 MHz, CD3OD) δ ppm 12.5, 23.9, 25.5, 29.2, 29.3, 29.5, 33.4,

34.1, 83.6, 84.1, 87.9, 91.1, 113.0, 117.2, 119.8, 122.0, 124.6, 126.0, 128.9,

135.6, 136.9, 138.0, 142.5, 156.2, 159.4, 163.4, 165.9, 170.2 .

Ʋmax (ATR-FTIR) cm-1: 3621(NH2), 3500-3200 (NH, OH), 2922 and 2852 (alkyl

CH3), 1655 (NC=O), 1601 (C=C), 1515 (aryl C-N), 1467 (aryl C-C), 1425 (N-

CO), 1381, 1357, 1207 (H-NCO), 1184, 1118, 1087 (C-O-C).

Melting point: 138–140 °C.

HRMS (ESI): m/z calcd for C40H53N8O6+ [M+H]: 741.4088, found: 741.4490.

HPLC purity: >99.9%.

238

7.2.43 Synthesis of 2-(2-(4-N-(3,5-di-tert-butyl-4-

hydroxyphenyl)acrylamido) phenyl))ethylaminoadenosine-5-N-

ethylcarboxamide (172) (2VHM162)

1 M HCl (3.3 mL) was added to a solution of compound 171 (121 mg, 0.94

mmol) in MeCN (8.0 mL). The solution was stirred for 4 h at 55–60 °C, and the

reaction progress was followed by TLC. Upon completion, the reaction mixture

was basified with saturated NaHCO3 solution up to pH 8 at 0–5 °C, the resulting

precipitate was extracted with EtOAc (3 x 25 mL). The combined organic phase

was washed with water (3 x 25 mL), brine solution (3 x 25 mL) and dried over

anhydrous MgSO4. The solvent was filtered and evaporated under reduced

pressure. The resulting residue was purified by MPLC instrument using a

mixture of 80% MeOH and 20% water with partial crystallisation to give 2-(2-(4-

N-(3,5-di-tert-butyl-4-hydroxyphenyl)acrylamido) phenyl))ethylaminoadenosine-

5-N-ethylcarboxamide (172) as a pale yellow solid (46 mg, 50%), Rf = 0.65

(EtOAc: MeOH, 8:2).

1H NMR (400 MHz, (CD3)2CO) δ ppm 1.02 (t, J=7.09 Hz, 3 H) 1.23 - 1.34 (m, 9

H) 1.44 - 1.52 (m, 9 H) 2.05 (d, J=1.03 Hz, 2 H) 3.07 - 3.17 (m, 1 H) 3.26 - 3.36

(m, 1 H) 3.51 - 3.58 (m, 1 H) 3.59 - 3.69 (m, 1 H) 4.36 (s, 1 H) 4.46 (br, s, 1 H)

5.02 - 5.09 (m, 2 H) 5.59 - 5.63 (s, 1 H) 5.91 (d, J=6.55 Hz, 1 H) 6.33 (br, s, 1 H)

239

6.69 (d, J=15 Hz, 2 H)) 7.23 (d, J=8.12 Hz, 2 H) 7.38 (s, 1H) 7.45 (s, 2 H) 7.64

(s, 1 H) 7.69 (s, 1 H) 7.70 (d, J=7.87 Hz, 2 H) 7.87 (s, 1 H) 9.26 (s, 1 H).

13C NMR (101 MHz, CD3OD) δ ppm 21.4, 29.1, 33.4 , 34.9, 35.5, 42.5, 83.6,

84.1, 87.9, 91.1, 113.9, 117.2, 119.8, 122.0, 126.0, 128.9, 135.7, 136.8, 137.0,

138.1, 142.6, 152.4 156.1, 156.2, 159.3, 159.5, 163.5, 165.9, 170.2.

Ʋmax (ATR-FTIR) cm-1: 3700-3000 (NH2, NH, OH), 2955 and 2852 (alkyl CH3),

1620 (C=C), 1596 (NC=O), 1514 (aryl C-N), 1467 (aryl C-C), 1424 (N-CO),

1356, 1236 (H-NCO), 1206, 1184, 1116 (C-O-C).

Melting point: 241-243 °C (decomp).

HRMS (ESI): m/z calcd for C37H49N8O6+ [M+H]: 701.3775, found: 701.4096.

HPLC purity: >99.9%.

240

7.2.44 Synthesis of 2-((1,1,3,3-tetramethylisoindoline-2-yloxyl)-5-ethynyl)-

2,3-O-isopropylideneadenosine-5-N-ethylcarboxamide (177).

Compound 149 (190 mg, 0.4 mmol,1 equiv), triethylamine (550 µL),

Pd(PPh3)2Cl2 (28.5 mg, 0.04 mmol, 0.1 equiv), 5-ethyne-1,1,3,3-

tetramethylisoindoline-2-yloxyl (120) (94.5 mg, 0.44 mmol,1.1 equiv), and

copper(I)iodide (15.0 mg, 0.08 mmol, 0.2 equiv), were combined in a mixture of

freshly distilled DCM (8.0 mL) and anhydrous MeCN (4.0 mL) The reaction

mixture was stirred at room temperature for 20 h. The reaction mixture was

quenched by adding chloroform (50 mL) and the organic layer was washed with

water (3 x 25 mL), brine solution (3 x 25 mL) and dried over anhydrous NaSO4.

The solvent was evaporated under reduced pressure, and the resulting residue

was purified by silica gel column chromatography using chloroform: 28%

aqueous ammonia solution (9.9:0.1) to give 2-((1,1,3,3-tetramethylisoindoline-2-

yloxyl)-5-ethynyl)-2,3-O-isopropylideneadenosine-5-N-ethylcarboxamide (177)

as a yellowish solid (170 mg, 76%), Rf = 0.57 (chloroform: MeOH, 9:1).

1H NMR (400 MHz, CDCl3) δ ppm: 1.00 (br. s., 3 H) 1.40 (s, 3 H) 1.65 (s, 3 H)

2.18 (s, 1 H) 2.63 - 2.65 (m, 1 H) 4.72 (s, 1 H) 5.31 - 5.34 (m, 1 H) 6.05 (br. s., 1

H) 7.12 - 7.22 (m, 1 H) 7.32 - 7.39 (m, 1 H) 7.40 - 7.44 (m, 1 H) 7.45 - 7.53 (m,

1 H) 7.84 - 7.89 (m, 1 H) Note that paramagnetic broadening by the

241

nitroxide radical results in some peaks not being detected or giving poor

integration in the proton NMR spectrum.

Ʋmax (ATR-FTIR) cm-1: 3634 (NH2), 3050 (alkyl CH3), 2220 (CΞC), 1668

(NC=O), 1572 (aryl C-N), 1490 (aryl C-C), 1454 (N-CO), 1377 (N-O), 1206 (H-

NCO), 1084 (C-O-C).

Melting point: 146–148 °C.

HRMS (ESI): m/z calcd for C29H35N7O5+ [M+H]: 561.2700, found: 561.2682.

HPLC purity: 97.4%.

EPR (methanol): typical 3-line nitroxide signal, g 1.9919, aN 1.4972 mT.

7.2.45 Synthesis of 2-((1,1,3,3-tetramethylisoindoline-2-yloxyl)-5-

ethynyl)adenosine-5-N-ethylcarboxamide (178).

1 M HCl solution (3.5 mL) was added slowly in a stirring solution of compound

177 (55 mg, 0.98 mmol, 1 equiv) in MeCN (3 mL). The reaction mixture was

stirred for 8 h at 55–60 °C and the reaction was followed by TLC analysis.in

(EtOAc: MeOH, 8:2). Upon completion, the pH of the solution was adjusted up

to 8 via the slow addition of saturated NaHCO3 solution (25 mL). The resulting

mixture was extracted with EtOAc (5 x 50 mL). The organic phases were

washed with water (3 x 25 mL), brine solution (3 x 25 mL), and dried over

242

anhydrous Na2SO4. The solvent was filtered and the filtrate was evaporated

under reduced pressure to give 2-((1,1,3,3-tetramethylisoindoline-2-yloxyl)-5-

ethynyl)adenosine-5-N-ethylcarboxamide (178) as a pale yellow solid (50 mg,

98%), Rf = 0.57 (EtOAc: MeOH, 8:2).

1H NMR (400 MHz, acetone-d6) δ ppm: 1.10 (br. s., 12 H) 4.37 (br. s., 2 H) 4.43

(s, 3 H) 4.68 - 4.81 (m, 2 H) 4.91 (br. s., 6 H) 6.04 (d, J=6.55 Hz, 3 H) 7.96 -

7.98 (m, 1 H) 8.31 (s, 3 H) 8.58 - 8.61 (m, 1 H). Note that paramagnetic

broadening by the nitroxide radical results in some peaks not being detected or

giving poor integration in the proton NMR spectrum.

Ʋmax (ATR-FTIR) cm-1: 3634 (NH2), 3050 (alkyl CH3), 2220 (CΞC), 1668

(NC=O), 1572 (aryl C-N), 1490 (aryl C-C), 1454 (N-CO), 1377 (N-O), 1206 (H-

NCO), 1084 (C-O-C).

Melting point: 261 °C (decomp).

HRMS (ESI): m/z calcd for C26H30NaN7O5+ [M+Na]: 543.2206, found: 543.2281.

EPR (methanol): typical 3-line nitroxide signal, g 1.9928, aN 1.4876 mT.

HPLC purity: 99.99%.

243

7.2.46 Synthesis of 2-((1,1,3,3-tetramethylisoindoline-2-yloxyl)-5-ethyl)-

2,3-O-isopropylideneadenosine-5-N-ethylcarboxamide (179).

Palladium (10% charcoal = 190 mg) was added to a stirring solution of

compound 177 (190 mg, 0.178 mmol, 1.equiv) in MeOH (50 mL). The reaction

mixture was stirred at room temperature using a balloon of H2 gas for 12 h and

the reaction progress was followed by TLC analysis in (CHCl3: MeOH, 9:1).

The reaction mixture was filtered through celite and the solvent was evaporated

under reduced pressure. The resulting residue was dissolved in MeOH and

added lead oxide (100 mg). The mixture was stirred for 1 h at room

temperature after which time filtered and dried through anhydrous Na2SO4 and

the solvent was evaporated under reduced pressure. The resulting residue was

purified by silica gel column chromatography using CHCl3: MeOH (9:1) to give

2-((1,1,3,3-tetramethylisoindoline-2-yloxyl)-5-ethyl)-2,3-O-

isopropylideneadenosine-5-N-ethylcarboxamide (179) as an off-white solid (161

mg, 84%). Rf = 0.023 (CHCl3: MeOH, 9:1),

1H NMR (400 MHz, CDCl3) δ ppm 0.67 (t, J=7.29 Hz, 3 H) 1.40 (s, 3 H) 1.60 (s,

3 H) 1.75 - 1.87 (m, 12 H) 2.75 - 2.87 (m, 1 H) 2.95 - 3.04 (m, 1 H) 3.04 - 3.11

(m, 2 H) 3.11 - 3.18 (m, 2 H) 3.48 (s, 1 H) 4.69 (d, J=1.37 Hz, 1 H) 5.42 (d,

244

J=6.80 Hz, 1 H) 5.59 - 5.67 (m, 3 H) 6.04 (t, J=5.40 Hz, 1 H) 6.13 (s, 1 H) 7.00

(s, 1 H) 7.05 (d, J=7.82 Hz, 1 H) 7.29 (s, 1 H) 7.81 (s, 1 H).

13C NMR (101 MHz, CDCl3) δ ppm 14.1, 25.2, 26.8, 33.7, 34.3, 40.6, 83.4, 83.8,

87.4, 91.4, 114.0, 121.3 (d, J=5.34 Hz, 1 C) 139.8, 155.2, 168.5. Note that

paramagnetic broadening by the nitroxide radical results in some peaks not

being detected or giving poor integration in the NMR spectrum.

Melting point: 108–110 °C. (Decomp.)

HRMS (ESI): m/z calcd for C29H38N7O5+ [M+]: 564.2934, found: 564.3276.

EPR (methanol): typical 3-line nitroxide signal, g 1.9928, aN 1.5066 mT.

HPLC purity- 96.72%.

7.2.47 .Synthesis of 2-((1,1,3,3-tetramethylisoindoline-2-yloxyl)-5-

ethyl)adenosine-5-N-ethylcarboxamide (180).

1 M HCl (10 mL) was slowly added in a stirring solution of compound 179 (150

mg, 0.155 mmol, 1 equiv) in MeCN (12.0 mL). The reaction mixture was stirred

for 5 h at 55–60 °C, and the reaction progress was followed by TLC analysis in

(CHCl3: MeOH, 8:2). Upon completion, the solution was basified up to pH 8 by

slow addition of saturated NaHCO3 solution (50 mL). The resulting suspension

245

was extracted with EtOAc (5 x 50 mL). The combined organic layers were

washed with water (3 x 50 mL) and brine solution (3 x 50 mL) and dried over

anhydrous Na2SO4, filtered. The solvent was evaporated under reduced

pressure to give a tan solid which was recrystallised (MeCN) to give 2-((1,1,3,3-

tetramethylisoindoline-2-yloxyl)-5-ethyl)adenosine-5-N-ethylcarboxamide (180)

as a pale yellow solid (110 mg, 79%). Rf = 0.16 (CHCl3: MeOH, 8:2).

1H NMR (400 MHz, Acetone-d6) δ ppm: 1.08 (t, J=7.16 Hz, 3 H) 1.31 - 1.31 (m,

1 H) 1.31 - 1.42 (m, 16 H) 2.98 - 3.08 (m, 2 H) 3.08 - 3.16 (m, 2 H) 3.26 (d,

J=7.09 Hz, 1 H) 3.31 - 3.43 (m, 1 H) 3.55 - 3.61 (m, 1 H) 4.38 - 4.44 (m, 2 H)

4.88 (dd, J=7.24, 4.60 Hz, 1 H) 6.02 (d, J=7.43 Hz, 1 H) 6.68 (br. s., 1 H) 7.00

(s, 1 H) 7.04 - 7.09 (m, 1 H) 7.09 - 7.15 (m, 1 H) 8.20 (s, 1 H) 8.23 (br. s., 1 H).

13C NMR (101 MHz, Acetone-d6) δ ppm: 14.7, 31.5, 31.6, 33.5, 34.7, 41.3, 62.0

(d, J=13.73 Hz, 1 C) 72.0, 73.7, 84.9, 88.7, 121.2 (d, J=3.05 Hz, 1 C) 127.1,

140.5, 140.7, 146.6, 149.1, 150.3, 156.2, 164.5, 169.2. Note that paramagnetic

broadening by the nitroxide radical results in some peaks not being detected or

giving poor integration in the NMR spectra

Ʋmax (ATR-FTIR) cm-1: 3198(OH), 2924 (alkyl CH3), 1636 (NC=O), 1581, 1388

(N-O), 1120, 1046, 797.

Melting point: 122-124 °C

HRMS (ESI): m/z calcd for C26H35N7O5+ [M+H]: 525.2700, found: 525.2833.

EPR (methanol): typical 3-line nitroxide signal, g 1.9939, aN 1.5006 mT.

HPLC purity- >99.9%.

246

7.2.48 Synthesis of 2-((1,1,3,3-tetraethylisoindoline-2-yloxyl)-5-ethynyl)-

2,3-O-isopropylideneadenosine-5-N-ethylcarboxamide (181).

Compound 149 (100 mg, 0.211 mmol, 1 equiv), 5-ethyne-(1,1,3,3-

tetraethylisoindoline-2-yloxyl (122) (57 mg, 0.211 mmol, 1 equiv), Pd(PPh3)2Cl2

(15 mg, 0.021 mmol, 0.1 equiv), copper iodide (8 mg, 0.042 mmol, 0.2 equiv),

and triethylamine (0.25 mL) were combined in a mixture of freshly distilled DCM

(6 mL) and anhydrous MeCN (3 mL). Argon was bubbled for 10 min, and the

reaction mixture was stirred at room temperature for 3 days and the reaction

progress was followed by TLC analysis using (DCM / MeOH, 9:1). The

chloroform (25 mL) was added to the reaction mixture and the organic layer was

washed with water (3 x 25 mL), and brine solution (3 x 25 mL) and dried over

anhydrous Na2SO4. The solvent was evaporated under reduced pressure and

the resulting residue was purified by silica gel column chromatography using a

mixture of (9:1) EtOAc: Hexane to give 2-((1,1,3,3-tetraethylisoindoline-2-

yloxyl)-5-ethynyl)-2,3-O-isopropylideneadenosine-5-N-ethylcarboxamide (181)

as a pale yellow solid (107 mg, 83%), Rf = 0.47 (DCM / MeOH, 9:1).

1H NMR (400 MHz, CDCl3)(Me trap) δ ppm: 0.58 - 1.05 (m, 5 H) 1.23 (br. s., 8

H) 1.57 (br. s., 13 H) 1.85 - 2.18 (m, 3 H) 2.97 (s, 5 H) 3.47 (br. s., 1 H) 3.66 (br.

s., 1 H) 4.63 (br. s., 1 H) 4.89 (br. s., 1 H) 5.60 - 5.92 (m, 1 H) 7.02 (d, J=6.85

Hz, 1 H) 7.36 - 7.57 (m, 2 H) 7.58 - 7.69 (m, 1 H) 7.89 (br. s., 1 H). Note that

247

paramagnetic broadening by the nitroxide radical results in some peaks not

being detected or giving poor integration in the proton NMR spectrum.

Ʋmax (ATR-FTIR) cm-1: 3400-3100 (OH, NH2), 2971 and 2960 (alkyl CH3 and

CH2), 2214 (CΞC), 1668 (NC=O), 1593 (aryl C-N), 1521 (aryl C-C), 1487 (N-

CO), 1374 (N-O), 1259, 1206 (H-NCO), 1156, 1083 (C-O-C).

Melting point: 188–190 °C.

HRMS (ESI): m/z calcd for C33H43N7O5+ [M + H]: 617.3326, found: 617.3296.

EPR (methanol): typical 3-line nitroxide signal, g 1.9913, aN 1.4200 mT.

HPLC purity- >99.9%.

7.2.49 Synthesis of 2-((1,1,3,3-tetraethylisoindoline-2-yloxyl)-5-

ethynyl)adenosine-5-N-ethylcarboxamide (182).

1 M HCl solution (5 mL) was added slowly.in a stirring solution of compound

181 (70 mg, 0.94 mmol, 1 equiv) was dissolved in MeCN (5.0 mL). The

reaction mixture was stirred for 3 h at 55–60 °C, and the reaction progress was

followed by TLC analysis in 100% EtOAc. Upon completion, the solution was

basified up to pH 8 by adding saturated NaHCO3 solution (25 mL). The

resulting suspension was extracted with EtOAc (5 x 50 mL). The organic

extracts were washed with water (3 x 25 mL) and brine solution (3 x 25 mL) and

dried over anhydrous Na2SO4, filtrated. The solvent was evaporated under

248

reduced pressure and the resulting residue was purified by silica gel column

chromatography using EtOAc to give 2-((1,1,3,3-tetraethylisoindoline-2-yloxyl)-

5-ethynyl)adenosine-5-N-ethylcarboxamide (182) as a tan solid (57 mg, 88%),

Rf = 0.53 (100% EtOAc).

1H NMR (400 MHz, CDCl3) δ ppm: 0.73 (br. s., 3 H) 0.81 - 1.00 (m, 8 H) 1.11

(s., 2H)1.23 (br. s., 15 H) 2.02 (br. s., 5 H) 2.97 (s, 3 H) 3.47 (br. s., 2 H) 3.54 -

3.74 (m, 4 H) 4.49 - 4.69 (m, 1 H) 4.89 (br. s., 1 H) 5.68 - 5.88 (m, 1 H) 7.00 (d,

J=5.18 Hz, 1 H) 7.44 (d, J=1.52 Hz, 1 H) 7.52 (d, J=6.46 Hz, 1 H) 7.58 - 7.69

(m, 1 H) 7.90 (br. s., 1 H) Note that paramagnetic broadening by the nitroxide

radical results in some peaks not being detected or giving poor integration in the

proton NMR spectrum.

Ʋmax (ATR-FTIR) cm-1: 3400-3100 (OH, NH2), 2971 and 2960 (alkyl CH3 and

CH2), 2214 (CΞC), 1668 (NC=O), 1593, 1521, 1487, 1374 (N-O), 1259, 1206,

1156, 1083, 861,796.

Melting Point: 196–198 °C (decomp).

HRMS (ESI): m/z calcd for C30H39N7O5+ [M+H]: 577.3013, found: 577.3012.

EPR (methanol): typical 3-line nitroxide signal, g 1.9928, aN 1.4113 mT.

HPLC purity- >99.9%.

249

7.2.50 Synthesis of 2-((1,1,3,3-tetraethylisoindoline-2-yloxyl)-5-ethyl)-2,3-

O-isopropylideneadenosine-5-N-ethylcarboxamide (183).

Palladium (10 % charcoal = 50 mg) was added in a stirring solution of

compound 181 (68 mg, 0.11 mmol, 1.0 equiv) in MeOH (4 mL). The reaction

mixture was stirred at room temperature under a balloon full of H2 gas for 12 h

and the reaction progress was followed by TLC analysis in (DCM: MeOH, 9:1).

The mixture was filtered through celite and the solvent was evaporated under

reduced pressure. The resulting residue was dissolved in MeOH and added

lead oxide (100 mg). The mixture was stirred for 1 h at room temperature after

which time filtered and dried through anhydrous Na2SO4 and the solvent was

evaporated under reduced pressure. The resulting residue was purified by

silica gel column chromatography using CHCl3 to give 2-((1,1,3,3-

tetraethylisoindoline-2-yloxyl)-5-ethyl)-2,3-O-isopropylideneadenosine-5-N-

ethylcarboxamide (183) as a yellow solid (66 mg, 96.5%), Rf = 0.76 (DCM:

MeOH, 9:1).

1H NMR (400 MHz, CDCl3) δ ppm 0.68 (t, J=6.90 Hz, 3 H) 1.38 (s, 12 H) 1.52 -

1.65 (m, 11 H) 2.84 - 2.86 (m, 2 H) 3.07 - 3.12 (m, 4 H) 4.68 (s., 1 H) 5.52 (br s,

2 H) 6.09-6.12 (s, 1 H) 6.18 (br s, 2 H) 6.89 (s, 1 H) 6.94 - 6.99 (m, 1 H) 7.12 -

7.14 (d, 1 H) 7.79 (br. s., 1 H).

250

13C NMR (101 MHz, CDCl3) δ ppm: 9.0, 25.3, 26. 9, 33.8, 34.4, 67.9, 82.1,

83.1, 83.3, 83.8, 87.3, 90.0, 91.4, 103.7, 110.0, 114.0, 118.3, 139.7, 168.5,

185.2, 217.3.

Melting point: 88–90 °C.

HRMS (ESI): m/z calcd for C33H47N7O5+ [M+H]: 621.3639, found: 621.3630.

HPLC purity: 88.72%.

EPR (methanol): typical 3-line nitroxide signal, g 1.9892, aN 1.4462 mT.

7.2.51 Synthesis of 2-((1,1,3,3-tetraethylisoindoline-2-yloxyl)-5-

ethyl)adenosine-5-N-ethylcarboxamide (184).

1 M HCl solution (5 mL) was added slowly in a stirred solution of compound 183

(66 mg, 0.106 mmol, 1 equiv) in MeCN (5.0 mL). The solution was stirred for 5

h at 55–60 °C and the reaction progress was followed by TLC analysis in DCM:

MeOH (9:1). Upon completion, the solution was basified up to pH 8 by adding

saturated NaHCO3 solution (25 mL). The resulting suspension was extracted

with EtOAc (5 x 50 mL). The combined organic phases were washed with

water (3 x 25 mL) and brine solution (3 x 25 mL) and filtered and dried over

anhydrous Na2SO4. The solvent was evaporated under reduced pressure

251

to give a tan solid, which was recrystallised (MeCN) to give 2-((1,1,3,3-

tetraethylisoindoline-2-yloxyl)-5-ethyl)adenosine-5-N-ethylcarboxamide (184)

as a pale yellow solid (43 mg, 70%). Rf = 0.69 (DCM: MeOH, 9:1).

Ʋmax (ATR-FTIR) cm-1: 3400-3100 (NH2, OH,), 2925 (alkyl CH3), 2214 (CΞC),

1636 (NC=O), 1581(aryl C-N), 1459 (N-CO), 1389 (N-O), 1119, 1046 (C-O-C).

Melting point: 138–140 °C

HRMS (ESI): m/z calcd for C30H43N7O5+ [M+H]: 581.3326, found: 581.3277.

HPLC purity:- 99.99%.

EPR (methanol): typical 3-line nitroxide signal, g 1.9921, aN 1.4609 mT.

7.2.52 Synthesis of 2-(2-Phenylethynyl)-2,3-O-isopropylideneadenosine -

5-N-ethylcarboxamide (185).

Compound 149 (100 mg, 0.211 mmol, 1 equiv), phenylacetylene 123 (57 mg,

0.232 mmol, 1.1 equiv), Pd(PPh3)2Cl2 (15 mg, 0.021 mmol, 0.1 equiv), copper

iodide (8 mg, 0.042 mmol, 0.2 equiv), and triethylamine (0.3 mL) were

combined in a mixture of freshly distilled DCM (6 mL) and anhydrous MeCN (3

mL). Argon was bubbled through a mixture for 10 minutes. The reaction

mixture was stirred at room temperature for 15 h and the reaction progress was

252

followed by TLC analysis in (DCM: MeOH, 9:1). The CHCl3 was added to a

mixture and resulting solution was washed with water (3 x 25 mL) and brine

solution (3 x 25 mL). The solvent was evaporated under reduced pressure and

the resulting residue was purified by silica gel column chromatography using

CHCl3 to give 2-(2-Phenylethynyl)-2,3-O-isopropylideneadenosine-5-N-

ethylcarboxamide (185) as a brownish solid (91 mg, 97%).

1H NMR (400 MHz, CDCl3) δ ppm: 0.94 - 1.00 (m, 3 H) 1.36 (s, 3 H) 1.62 (s, 3

H) 3.16 - 3.30 (m, 1 H) 3.37 (dt, J=13.60, 6.94 Hz, 1 H) 4.69 (s, 1 H) 5.24 (d,

J=5.18 Hz, 1 H) 5.30 (br. s., 1 H) 6.00 (d, J=1.37 Hz, 1 H) 7.35 (br. s., 1 H) 7.35

- 7.40 (m, 3 H) 7.58 – 7.61(m, 2H).

13C NMR (101 MHz, CDCl3) δ ppm: 14.7 , 25.3 , 27.3 , 34.1 , 76.7 , 77.0 , 77.2 ,

77.3 , 82.0 , 83.4 , 84.9 , 114.9 , 128.4 , 129.5 , 132.3 , 168.5 .

Ʋmax (ATR-FTIR) cm-1: 3356 (NH2), 3156 (OH), 2214 (CΞC), 1654 (NC=O),

1588 (aryl C-N), 1522 (aryl C-C), 1492, 1457 (N-CO), 1380, 1203 (H-NCO),

1155, 1082 (C-O-C).

Melting point: 174–176 °C ;(lit )

HRMS (ESI): m/z calcd for C23H25N6O4+ [M + H]: 449.1937, found: 449.1881.

HPLC purity- 99.99%.

253

7.2.53 Synthesis of 2-(2-phenylethyl)-2,3-O-isopropylideneadenosine-5-

N-ethylcarboxamide (187).

Palladium (10% charcoal = 80 mg) was added to a stirred solution of compound

185 (80 mg, 0.178 mmol, 1.0 eq) in MeOH (10 mL). The reaction mixture was

stirred at room temperature using a balloon of H2 gas atmosphere for 12 h and

the reaction progress was followed by TLC analysis in (DCM: MeOH, 9:1). The

mixture was filtered through celite and the filtrate was evaporated under

reduced pressure. The resulting residue was purified by silica gel column

chromatography using a DCM: MeOH (9:1) to give 2-(2-phenylethyl)-2,3-O-

isopropylideneadenosine-5-N-ethylcarboxamide (187) as a yellow solid (75 mg,

93 %). Rf = 0.56 (DCM: Methanol, 9:1).

1H NMR (400 MHz, CDCl3): δ ppm 0.67 (t, J=7.24 Hz, 4 H) 1.37 (s, 3 H) 1.58 (s,

3H) 2.77 - 2.86 (m, 1 H) 2.99 - 3.05 (m, 1 H) 3.05 - 3.14 (m, 4 H) 4.65 (d, J=1.66

Hz, 1 H) 5.25 (d, J=4.65 Hz, 1 H) 5.47 - 5.55 (m, 3 H) 6.05 (d, J=1.37 Hz, 1 H)

7.15 (d, J=6.60 Hz, 1 H) 7.19 - 7.23 (m, 3 H) 7.76 (s, 1 H).

13C NMR (75 MHz, DMSO- d6): δ ppm: 14.1, 25.2, 26.9, 33.7, 34.5, 40.5, 83.2,

83.8, 86.2, 91.4, 114.0, 118.3, 125.8, 128.3, 128.4, 139.7, 141.5, 155.5, 165.4

and 168.5.

254

Melting point: 70–72 °C.

HRMS (ESI): m/z calcd for C23H29N6O4+ [M+H]: 453.2250, found: 453.2243.

HPLC purity- 98.32%.

7.2.54 Synthesis of 2-(2-phenylethyl)adenosine-5-N-ethylcarboxamide

(188).

1 M HCl solution (6 mL) was added slowly in a stirring solution of compound

187 (70 mg, 0.155 mmol, 1 equiv) in MeCN (6.0 mL). The reaction mixture was

stirred for 5 h at 55–60 °C and the reaction progress was followed by TLC

analysis in (DCM: MeOH, 9:1). Upon completion, the solution was basified up

to the pH 8 by adding saturated NaHCO3 solution (25 mL). The resulting

suspension was extracted with EtOAc (5 x 25 mL) and the combined organic

layers were washed with water (3 x 25 mL) and brine solution (3 x 25 mL) and

dried over anhydrous Na2SO4. The solvent was evaporated under reduced

pressure to give a brownish solid which was recrystallised (MeCN) to give 2-(2-

phenylethyl)adenosine-5-N-ethylcarboxamide (188) as a pale yellow solid (60

mg, 94%). Rf = 0.38 (DCM: MeOH, 9:1).

1H NMR (400 MHz, CDCl3) δ ppm: 1.06 (t, J=7.26 Hz, 3 H) 2.03 (s, 2 H) 3.07 (d,

J=4.55 Hz, 4 H) 3.16 - 3.21 (m, 1 H) 3.37 - 3.43 (m, 1 H) 4.57 (s, 1 H) 4.63 -

255

4.68 (m, 2 H) 5.63 (br. s, 2 H) 5.83 (d, J=5.48 Hz, 1 H) 7.15 - 7.19 (m, 3 H) 7.23

(br. s., 1 H) 7.24(br. s, 1 H) 7.84 (s, 1 H).

13C NMR (101 MHz, CDCl3) δ ppm: 14.7 , 25.3 , 27.3 , 34.1 , 76.7 , 77.0 , 77.2 ,

77.3 , 82.0 , 83.4 , 84.9 , 114.9 , 128.4 , 129.5 , 132.3 and 168.5.

Ʋmax (ATR-FTIR) cm-1: 3330 (NH2), 3225 (OH), 2921 (aryl C-H), 1643 (NC=O),

1588 (aryl C-N), 1522 (aryl C-C), 1467 (N-CO), 1396, 1315, 1203 (H-NCO),

1196, 1143, 1115, 1080 (C-O-C), 1043, 968, 870.

Melting point: 216-217 °C.(decomp.)

HRMS (ESI): m/z calcd for C20H25N6O4+ [M+H]: 413.1937, found: 413.2019.

HPLC purity- 100%.

256

BIBLIOGRAPHY 8

257

1. Ogunseitan, Oladele ed., I. Cardiovascular Diseases. Green Health: An A-to-Z Guide. SAGE Publications, Inc. SAGE Publications, Inc.: Thousand Oaks, CA, 2011, 9.

2. Guttmacher, A. E. M. D.; Collins, F. S. M. D. P.; Nabel, E. G. M. D. Cardiovascular disease. The New England Journal of Medicine 2003, 349, 60-72.

3. Cai, H.; Harrison, D. G. Endothelial dysfunction in cardiovascular diseases: the role of oxidant stress. Circulation Research 2000, 87, 840-844.

4. Peluso, I.; Morabito, G.; Urban, L.; Ioannone, F.; Serafi, M. Oxidative stress in atherosclerosis development: the central role of LDL and oxidative burst. Endocrine, Metabolic & Immune Disorders-Drug Targets (Formerly Current Drug Targets-Immune, Endocrine & Metabolic Disorders) 2012, 12, 351-360.

5. Touyz, R. M. Reactive oxygen species, vascular oxidative stress, and redox signaling in hypertension what is the clinical significance? Hypertension 2004, 44, 248-252.

6. Rodrigo, R.; González, J.; Paoletto, F. The role of oxidative stress in the pathophysiology of hypertension. Hypertension Research 2011, 34, 431-440.

7. Anderson, K. M.; Odell, P. M.; Wilson, P. W.; Kannel, W. B. Cardiovascular disease risk profiles. American Heart Journal 1991, 121, 293-298.

8. Ceriello, A.; Motz, E. Is oxidative stress the pathogenic mechanism underlying insulin resistance, diabetes, and cardiovascular disease? The common soil hypothesis revisited. Arteriosclerosis, Thrombosis, and Vascular Biology 2004, 24, 816-823.

9. Ganzit, G. P.; Stefanini, L. Cardiovascular diseases and physical activity. In 1 ed.; SEEd: Torino, 2012.

10. Abegunde, D. O.; Mathers, C. D.; Adam, T.; Ortegon, M.; Strong, K. The burden and costs of chronic diseases in low-income and middle-income countries. The Lancet 2007, 370, 1929-1938.

11. Grace, P. A. Ischaemia-reperfusion injury. British Journal of Surgery 1994, 81, 637-647.

12. Bhatia, S. K. Biomaterials for clinical applications. Springer Science & Business Media: 2010.

13. Mittal, B. V.; Singh, A. K. Hypertension in the developing world: challenges and opportunities. American Journal of Kidney Diseases 2010, 55, 590-598.

14. Selzer, A., M.D. Understanding heart disease. . In University of California Press: Berkeley, 1992.

258

15. McCord, J. M. Oxygen-derived free radicals in postischemic tissue injury. New England Journal of Medicine 1985, 312, 159-163.

16. Gutterman, D. D. Silent myocardial ischemia. Circulation Journal 2009, 73, 785-797.

17. Blomgren, K.; Hagberg, H. Free radicals, mitochondria, and hypoxia–ischemia in the developing brain. Free Radical Biology and Medicine 2006, 40, 388-397.

18. Randall, O. S. R., Deborah S. Segerson, Nathan M. . The encyclopedia of the heart and heart disease. In 2nd ed.; Facts On File New York, NY, USA 2010.

19. Chervu, A.; Moore, W. S.; Homsher, E.; Quinones-Baldrich, W. J. Differential recovery of skeletal muscle and peripheral nerve function after ischemia and reperfusion. Journal of Surgical Research 1989, 47, 12-9.

20. Kerrigan, C. L., Stotland, Mitchell A. Ischemia reperfusion injury: A review. Microsurgery 1993, 14, 165-175.

21. Anaya-Prado, R.; Toledo-Pereyra, L. H.; Lentsch, A. B.; Ward, P. A. Ischemia/reperfusion injury. Journal of Surgical Research 2002, 105, 248-258.

22. Halliwell, B.; Gutteridge, J.; Cross, C. Free radicals, antioxidants, and human disease: where are we now? The Journal of Laboratory and Clinical Medicine 1992, 119, 598.

23. Wink, D. A.; Mitchell, J. B. Chemical biology of nitric oxide: insights into regulatory, cytotoxic, and cytoprotective mechanisms of nitric oxide. Free Radical Biology and Medicine 1998, 25, 434-456.

24. Granger, D. N. Role of xanthine oxidase and neutrophils in ischemia-reperfusion injury in rabbit lung. 1990; Vol. 69, p 2012-2018.

25. Tyler, D. D. Polarographic assay and intracellular distribution of superoxide dismutase in rat liver. Biochem. J 1975, 147, 493-504.

26. Granger, D. N. Role of xanthine oxidase and granulocytes in ischemia-reperfusion injury. American Journal of Physiology-Heart and Circulatory Physiology 1988, 255, H1269-H1275.

27. Halliwell, B.; Gutteridge, J. Biologically relevant metal ion-dependent hydroxyl radical generation An update. Febs Letters 1992, 307, 108-112.

28. Wink, D. A.; Wink, C. B.; Nims, R. W.; Ford, P. C. Oxidizing intermediates generated in the Fenton reagent: kinetic arguments against the intermediacy of the hydroxyl radical. Environmental Health Perspectives 1994, 102, 11.

29. Halliwell, B.; Clement, M. V.; Ramalingam, J.; Long, L. H. Hydrogen peroxide. Ubiquitous in cell culture and in vivo? IUBMB life 2000, 50, 251-257.

259

30. Halliwell, B.; Clement, M. V.; Long, L. H. Hydrogen peroxide in the human body. Febs Letters 2000, 486, 10-13.

31. Gomberg, M. An instance of trivalent carbon: Triphenylmethyl. Journal of the American Chemical Society 1900, 22, 757-771.

32. Fang, Y.-Z.; Yang, S.; Wu, G. Free radicals, antioxidants, and nutrition. Nutrition 2002, 18, 872-879.

33. Kalra, J.; Chaudhary, A.; Prasad, K. In Cigarette-smoking and oxygen free-radicals producing activity of polymorphonuclear leukocytes, Clinical Pharmacology & Therapeutics, 1989; Mosby-year book Inc 11830 Westline Industrial Dr, St Louis, Mo 63146-3318: 1989; pp 178-178.

34. Nonhebel, D. a. J. W. Free radical chemistry: structure and mechanism

1974.

35. Mittler, R.; Vanderauwera, S.; Suzuki, N.; Miller, G.; Tognetti, V. B.; Vandepoele, K.; Gollery, M.; Shulaev, V.; Van Breusegem, F. ROS signaling: the new wave? Trends in Plant Science 2011, 16, 300-309.

36. Zhang, M.; Shah, A. M. ROS signalling between endothelial cells and cardiac cells. Cardiovascular Research 2014, 102, 249-257.

37. Valko, M.; Rhodes, C.; Moncol, J.; Izakovic, M.; Mazur, M. Free radicals, metals and antioxidants in oxidative stress-induced cancer. Chemico-Biological Interactions 2006, 160, 1-40.

38. Maddika, S.; Elimban, V.; Chapman, D.; Dhalla, N. S. Role of oxidative stress in ischemia-reperfusion-induced alterations in myofibrillar ATPase activities and gene expression in the heart. Canadian Journal of Physiology & Pharmacology 2009, 87, 120-129.

39. Westerblad, H.; Allen, D. G. Emerging roles of ROS/RNS in muscle function and fatigue. Antioxidants & Redox Signaling 2011, 15, 2487-2499.

40. Rodrigo, R.; Libuy, M.; Feliú, F.; Hasson, D. Oxidative stress-related biomarkers in essential hypertension and ischemia-reperfusion myocardial damage. Disease Markers 2013, 35, 773-790.

41. Fubini, B.; Hubbard, A. Reactive oxygen species (ROS) and reactive nitrogen species (RNS) generation by silica in inflammation and fibrosis. Free Radical Biology and Medicine 2003, 34, 1507-1516.

42. Binger, C. A.; Faulkner, J. M.; Moore, R. L. Oxygen poisoning in mammals. The Journal of Experimental Medicine 1927, 45, 849.

43. Gerschman, R.; Gilbert, D. L.; Nye, S. W.; Dwyer, P.; Fenn, W. O. Oxygen poisoning and x-irradiation: a mechanism in common. Science of Aging Knowledge Environment 2005, 2005, cp1.

260

44. Michaelson, L. P.; Iler, C.; Ward, C. W. ROS and RNS signaling in skeletal muscle: Critical signals and therapeutic targets. Annual Review of Nursing Research 2013, 31, 367-X.

45. Szabó, C.; Ischiropoulos, H.; Radi, R. Peroxynitrite: biochemistry, pathophysiology and development of therapeutics. Nature Reviews Drug Discovery 2007, 6, 662-680.

46. Finkel, T.; Holbrook, N. J. Oxidants, oxidative stress and the biology of ageing. Nature 2000, 408, 239-247.

47. Harman, D. Aging: a theory based on free radical and radiation chemistry. University of California Radiation Laboratory: 1955.

48. Harman, D. The biologic clock: the mitochondria? Journal of the American Geriatrics Society 1972, 20, 145-147.

49. Leeuwenburgh, C.; Heinecke, J. W. Oxidative stress and antioxidants in exercise. Current Medicinal Chemistry 2001, 8, 829-38.

50. Preiser, J.-C. Oxidative stress. Journal of Parenteral and Enteral Nutrition 2012, 36, 147-154.

51. Morrow, J. D.; Frei, B.; Longmire, A. W.; Gaziano, J. M.; Lynch, S. M.; Shyr, Y.; Strauss, W. E.; Oates, J. A.; Roberts, L. J. Increase in circulating products of lipid peroxidation (F2-isoprostanes) in smokers—smoking as a cause of oxidative damage. New England Journal of Medicine 1995, 332, 1198-1203.

52. Wikipedia. Oxidation http://simple.wikipedia.org/wiki/Oxidation.

53. Sies, H. Oxidative stress: oxidants and antioxidants. Experimental Physiology 1997, 82, 291-295.

54. Lockwood, S. F.; Gross, G. J. Disodium disuccinate astaxanthin (CardaxTM): antioxidant and antiinflammatory cardioprotection. Cardiovascular Drug Reviews 2005, 23, 199-216.

55. Pepys, M. B.; Hirschfield, G. M.; Tennent, G. A.; Gallimore, J. R.; Kahan, M. C.; Bellotti, V.; Hawkins, P. N.; Myers, R. M.; Smith, M. D.; Polara, A. Targeting C-reactive protein for the treatment of cardiovascular disease. Nature 2006, 440, 1217-1221.

56. Dafré, A. L.; Sies, H.; Akerboom, T. Protein S-thiolation and regulation of microsomal glutathione transferase activity by the glutathione redox couple. Archives of Biochemistry and Biophysics 1996, 332, 288-294.

57. Aronovitch, Y.; Godinger, D.; Israeli, A.; Krishna, M. C.; Samuni, A.; Goldstein, S. Dual activity of nitroxides as pro- and antioxidants: Catalysis of copper-mediated DNA breakage and H2O2 dismutation. Free Radical Biology & Medicine 2007, 42, 1317-1325.

261

58. Brivba K., S. H. Non enzymatic antioxidant defense systems, in Natural Antioxidants in Human Health and Disease. San Diego: Academic Press: Orlando, FL, USA., 1994. ; p pp. 107–128.

59. Huang, D.; Ou, B.; Prior, R. L. The chemistry behind antioxidant capacity assays. Journal of Agricultural and Food Chemistry 2005, 53, 1841-1856.

60. Teixeira, J.; Gaspar, A.; Garrido, E. M.; Garrido, J.; Borges, F. Hydroxycinnamic acid antioxidants: an electrochemical overview. BioMed Research International 2013, 2013.

61. McCord, J. M.; Fridovich, I. SOD enzyme function for erythrocuprein. J. Biol. Chem. 1969, 224, 6049-6055.

62. Palmer, R. M. J.; Ashton, D. S.; Moncada, S. Vascular endothelial cells synthesize nitric oxide from L-arginine. Nature 1988, 333, 664-666.

63. Lam, M. A.; Pattison, D. I.; Bottle, S. E.; Keddie, D. J.; Davies, M. J. Nitric oxide and nitroxides can act as efficient scavengers of protein-derived free radicals. Chemical Research in Toxicology 2008, 21, 2111-2119.

64. Cho, J.; Son, S.; Ko, S.; Lee, M.; Byun, S.; Lee, Y.; Park, S.; Kim, J.; Gwag, B. In Nonclinical safety and pharmacokinetic profile of AAD-2004 as a putative disease-modifying drug for neurodegenerative diseases, The 9th international conference AD/PD, 2009, pp 11-15.

65. Shin, J. H.; Lee, Y. A.; Lee, J. K.; Lee, Y. B.; Cho, W.; Im, D. S.; Lee, J. H.; Yun, B. S.; Springer, J. E.; Gwag, B. J. Concurrent blockade of free radical and microsomal prostaglandin E synthase-1-mediated PGE2 production improves safety and efficacy in a mouse model of amyotrophic lateral sclerosis. Journal of Neurochemistry 2012, 122, 952-961.

66. Jackson, D.; Sammut, I. Oxygen free radical traps for the treatment of ischemia-associated organ injury. Current Opinion in Investigational Drugs (London, England: 2000) 2004, 5, 50-54.

67. Lees, K. R.; Zivin, J. A.; Ashwood, T.; Davalos, A.; Davis, S. M.; Diener, H.-C.; Grotta, J.; Lyden, P.; Shuaib, A.; Hårdemark, H.-G. NXY-059 for acute ischemic stroke. New England Journal of Medicine 2006, 354, 588-600.

68. Griffith, O. H.; McConnell, H. M. A nitroxide-maleimide spin label. Proceedings of the National Academy of Sciences of the United States of America 1966, 55, 8.

69. Griffith, O. H.; Cornell, D. W.; McConnell, H. M. Nitrogen hyperfine tensor and g tensor of nitroxide radicals. The Journal of Chemical Physics 2004, 43, 2909-2910.

70. Forrester, A. R.; Hepburn, S. P. Nitroxide radicals. Part XV. p-Methoxy-and p-phenoxy-phenyl t-butyl nitroxides. Journal of the Chemical Society, Perkin Transactions 1 1974, 2208-2213.

262

71. Mukherjee, S. Syntheses of nitroxide diradicals and tetraradicals. Ph.D., The University of Nebraska - Lincoln, Ann Arbor, 2006.

72. Nonhebel, D. C.; Walton, J. C. Free-radical chemistry; structure and mechanism. CUP Archive: 1974.

73. Hicks, R. Stable radicals: fundamentals and applied aspects of odd-electron compounds. John Wiley & Sons: 2011.

74. Volodarsky, L. B.; Reznikov, V. A.; Ovcharenko, V. I. Synthetic chemistry of stable nitroxides. CRC Press Boca Raton, FL: 1994.

75. Soule, B. P.; Hyodo, F.; Matsumoto, K.-i.; Simone, N. L.; Cook, J. A.; Krishna, M. C.; Mitchell, J. B. The chemistry and biology of nitroxide compounds. Free Radical Biology and Medicine 2007, 42, 1632-1650.

76. Griffiths, P.; Moad, G.; Rizzardo, E. Synthesis of the radical scavenger 1,1,3,3-Tetramethylisoindolin-2-yloxyl. Australian Journal of Chemistry 1983, 36, 397-401.

77. Bottle, S.; Busfield, W.; Grice, I.; Heiland, K.; Jenkins, I.; Meutermans, W.; Monteiro, M. Some recent developments in the aminoxyl radical trapping technique. In Progress in Pacific Polymer Science 3, Springer: 1994; pp 85-97.

78. Keddie, D. J.; Johnson, T. E.; Arnold, D. P.; Bottle, S. E. Synthesis of profluorescent isoindoline nitroxides via palladium-catalysed Heck alkenylation. Organic and Biomolecular Chemistry 2005, 3, 2593-2598.

79. Fairfull-Smith, K. E.; Blinco, J. P.; Keddie, D. J.; George, G. A.; Bottle, S. E. A novel profluorescent dinitroxide for imaging polypropylene degradation. Macromolecules 2008, 41, 1577-1580.

80. Hosokawa, K.; Chen, P.; Lavin, F. M.; Bottle, E. S. The impact of carboxy nitroxide antioxidants on irradiated ataxia telangiectasia cells. Free Radical Biology and Medicine 2004, 37, 946-952.

81. Krishna, M. C.; Halevy, R. F.; Zhang, R. L.; Gutierrez, P. L.; Samuni, A. Modulation of streptonigrin cytotoxicity by nitroxide sod mimics. Free Radical Biology and Medicine 1994, 17, 379-388.

82. Mitchell, J. B.; Xavier, S.; DeLuca, A. M.; Sowers, A. L.; Cook, J. A.; Krishna, M. C.; Hahn, S. M.; Russo, A. A low molecular weight antioxidant decreases weight and lowers tumor incidence. Free Radical Biology and Medicine 2003, 34, 93-102.

83. Kuppusamy, P.; Chzhan, M.; Vij, K.; Shteynbuk, M.; Lefer, D. J.; Giannella, E.; Zweier, J. L. Three-dimensional spectral-spatial EPR imaging of free radicals in the heart: a technique for imaging tissue metabolism and oxygenation. Proceedings of the National Academy of Sciences 1994, 91, 3388-3392.

263

84. Kao, J. P.; Barth, E. D.; Burks, S. R.; Smithback, P.; Mailer, C.; Ahn, K.

H.; Halpern, H. J.; Rosen, G. M. Very‐low‐frequency electron paramagnetic resonance (EPR) imaging of nitroxide‐loaded cells. Magnetic Resonance in Medicine 2007, 58, 850-854.

85. Brustad, T.; Bugge, H.; Jones, W.; Wold, E. Reactions between organic nitroxyl free radicals and radiation-induced transients in the DNA bases. International Journal of Radiation Biology 1972, 22, 115-129.

86. Emmerson, P. T.; Howard-Flanders, P. Sensitization of anoxic bacteria to x-rays by di-t-butyl nitroxide and analogues. Nature 1964, 204, 1005-1006.

87. Gubskaya, V. P.; Berezhnaya, L. S.; Gubaidullin, A. T.; Faingold, I. I.; Kotelnikova, R. A.; Konovalova, N. P.; Morozov, V. I.; Litvinov, I. A.; Nuretdinov, I. A. Synthesis, structure and biological activity of nitroxide malonate methanofullerenes. Organic & Biomolecular Chemistry 2007, 5, 976-981.

88. Likhtenshtein, G. I. Novel fluorescent methods for biotechnological and biomedical sensoring: assessing antioxidants, reactive radicals, NO dynamics, immunoassay, and biomembranes fluidity. Applied Biochemistry and Biotechnology 2009, 152, 135-155.

89. Kato, N.; Yanaka, K.; Hyodo, K.; Homma, K.; Nagase, S.; Nose, T. Stable nitroxide Tempol ameliorates brain injury by inhibiting lipid peroxidation in a rat model of transient focal cerebral ischemia. Brain Research 2003, 979, 188-193.

90. Wilcox, C. S.; Pearlman, A. Chemistry and antihypertensive effects of tempol and other nitroxides. Pharmacological Reviews 2008, 60, 418-469.

91. Quinu, J.; Delbridge, L.; Proietto, J.; Ritchie, R. In Cardioprotective actions of the antioxidant tempol in GLUT4-deficient mice, Clinical and Experimental Pharmacology and Physiology, 2004; Blackwell Publishing Asia 54 University St, Po Box 378, Carlton, Victoria 3053, Australia: 2004; pp A25-A25.

92. Goldstein, S.; Samuni, A.; Russo, A. Reaction of cyclic nitroxides with nitrogen dioxide: the intermediacy of the oxoammonium cations. Journal of the American Chemical Society 2003, 125, 8364-8370.

93. Tabaczar, S.; Talar, M.; Gwoździński, K. [Nitroxides as antioxidants-possibilities of their application in chemoprevention and radioprotection]. Postepy Higieny i Medycyny Doswiadczalnej (Online) 2010, 65, 46-54.

94. Weil, J. T.; Van der Veen, J.; Olcott, H. Stable nitroxides as lipid antioxidants. 1968.

95. Halliwell, B.; Whiteman, M. Measuring reactive species and oxidative damage in vivo and in cell culture: how should you do it and what do the results mean? British Journal of Pharmacology 2004, 142, 231-255.

264

96. Gutteridge, J. Lipid peroxidation and antioxidants as biomarkers of tissue damage. Clinical Chemistry 1995, 41, 1819-1828.

97. Chen, X.; Sun, F.; Ma, L.; Wang, J.; Qin, H.; Du, G. In vitro evaluation on the antioxidant capacity of triethylchebulate, an aglycone from Terminalia chebula Retz fruit. Indian Journal of Pharmacology 2011, 43, 320.

98. Vincent, S. R. Nitric oxide: A radical neurotransmitter in the central nervous system. Progress in Neurobiology 1994, 42, 129-160.

99. Chatterjee, P. K.; Cuzzocrea, S.; Brown, P. A.; Zacharowski, K.; Stewart, K. N.; Mota-Filipe, H.; Thiemermann, C. Tempol, a membrane-permeable radical scavenger, reduces oxidant stress-mediated renal dysfunction and injury in the rat. Kidney International 2000, 58, 658-673.

100. Zhu, X.; Zuo, L.; Cardounel, A. J.; Zweier, J. L.; He, G. Characterization of in vivo tissue redox status, oxygenation, and formation of reactive oxygen species in postischemic myocardium. Antioxidants and Redox Signaling 2007, 9, 447-455.

101. Walker, J. R.; Fairfull-Smith, K. E.; Anzai, K.; Lau, S.; White, P. J.; Scammells, P. J.; Bottle, S. E. Edaravone containing isoindoline nitroxides for the potential treatment of cardiovascular ischaemia. MedChemComm 2011, 2, 436-441.

102. Fairfull-Smith, K. E.; Brackmann, F.; Bottle, S. E. The synthesis of novel isoindoline nitroxides bearing water-solubilising functionality. European Journal of Organic Chemistry 2009, 2009, 1902-1915.

103. Samuni, A. M.; Degraff, W.; Krishna, M. C.; Mitchell, J. B. Nitroxides as antioxidants: Tempol protects against EO9 cytotoxicity. Molecular and Cellular Biochemistry 2002, 234-235, 327-33.

104. Wu, G.; Meininger, C. J. Regulation of nitric oxide synthesis by dietary factors. Annual Review of Nutrition 2002, 22, 61-86.

105. Cong, J.-H.; Sun, C.-P.; Tian, X.-H.; Fang, Y.-Z. Effect of Lu-Duo-Wei on Scavenging Superoxide and Hydroxyl Radicals in vitro. The American Journal of Chinese Medicine 1998, 26, 153-158.

106. Jackson, M. J. An overview of methods for assessment of free radical activity in biology. Proceedings of the Nutrition Society 1999, 58, 1001-1006.

107. Gilbert, D. L. Fifty years of radical ideas. Annals of the New York Academy of Sciences 2000, 899, 1-14.

108. Rice-Evans, C.; Miller, N.; Paganga, G. Antioxidant properties of phenolic compounds. Trends in Plant Science 1997, 2, 152-159.

109. Cai, Y.; Luo, Q.; Sun, M.; Corke, H. Antioxidant activity and phenolic compounds of 112 traditional Chinese medicinal plants associated with anticancer. Life Sciences 2004, 74, 2157-2184.

265

110. Rice-Evans, C. A.; Miller, N. J.; Paganga, G. Structure-antioxidant activity relationships of flavonoids and phenolic acids. Free Radical Biology and Medicine 1996, 20, 933-956.

111. Wright, J. S.; Johnson, E. R.; DiLabio, G. A. Predicting the activity of phenolic antioxidants: theoretical method, analysis of substituent effects, and application to major families of antioxidants. Journal of the American Chemical Society 2001, 123, 1173-1183.

112. Noguchi, N.; Niki, E. Phenolic antioxidants:: A rationale for design and evaluation of novel antioxidant drug for atherosclerosis. Free Radical Biology and Medicine 2000, 28, 1538-1546.

113. Pryor, W. A. Vitamin E and heart disease:: basic science to clinical intervention trials. Free Radical Biology and Medicine 2000, 28, 141-164.

114. Noguchi, N.; Iwaki, Y.; Takahashi, M.; Komuro, E.; Kato, Y.; Tamura, K.; Cynshi, O.; Kodama, T.; Niki, E. 2,3-Dihydro-5-hydroxy-2,2-dipentyl-4,6-di-tert-butylbenzofuran: Design and evaluation as a novel radical-scavenging antioxidant against lipid peroxidation. Archives of Biochemistry and Biophysics 1997, 342, 236-243.

115. Agadzhanyan, V.; Oganesyan, E.; Abaev, V. Targeted search for a lead compound in a series of cinnamic acid derivatives possessing antiradical activity. Pharmaceutical Chemistry Journal 2010, 44, 360-365.

116. Durand, E.; Bayrasy, C.; Laguerre, M.; Barouh, N.; Lecomte, J.; Durand,

T.; Balas, L.; Wrutniak‐Cabello, C.; Cabello, G.; Villeneuve, P. Regioselective synthesis of diacylglycerol rosmarinates and evaluation of their antioxidant activity in fibroblasts. European Journal of Lipid Science and Technology 2015.

117. Kwak, S.-Y.; Yang, J.-K.; Choi, H.-R.; Park, K.-C.; Kim, Y.-B.; Lee, Y.-S. Synthesis and dual biological effects of hydroxycinnamoyl phenylalanyl/prolyl hydroxamic acid derivatives as tyrosinase inhibitor and antioxidant. Bioorganic & Medicinal Chemistry Letters 2013, 23, 1136-1142.

118. Fresco, P.; Borges, F.; Diniz, C.; Marques, M. New insights on the anticancer properties of dietary polyphenols. Medicinal Research Reviews 2006, 26, 747-766.

119. Simonyan, A. Activity of cinnamic acid derivatives and new methods for their synthesis (review). Pharmaceutical Chemistry Journal 1993, 27, 92-100.

120. Shepherd, R. G. Certain cinnamic acid or propiolic acid derivatives. 1983.

121. Hausler, N. E.; Devine, S. M.; McRobb, F. M.; Warfe, L.; Pouton, C. W.; Haynes, J. M.; Bottle, S. E.; White, P. J.; Scammells, P. J. Synthesis and pharmacological evaluation of dual acting antioxidant A2A adenosine receptor agonists. Journal of Medicinal Chemistry 2012, 55, 3521-3534.

266

122. Arch, J. R.; Newsholme, E. A. The control of the metabolism and the hormonal role of adenosine. Essays in Biochemistry 1978, 14, 82-123.

123. Maggirwar, S. B.; Dhanraj, D. N.; Somani, S. M.; Ramkumar, V. Adenosine acts as an endogenous activator of the cellular antioxidant defense system. Biochemical and Biophysical Research Communications 1994, 201, 508-515.

124. Cronstein, B. N.; Kramer, S. B.; Weissmann, G.; Hirschhorn, R. Adenosine: a physiological modulator of superoxide anion generation by human neutrophils. The Journal of Experimental Medicine 1983, 158, 1160-1177.

125. Williams-Karnesky, R. L.; Stenzel-Poore, M. P. Adenosine and stroke: Maximizing the therapeutic potential of adenosine as a prophylactic and acute neuroprotectant. Current Neuropharmacology 2009, 7, 217.

126. Silverman, R. B. The organic chemistry of drug design and drug action. Academic press: 2004.

127. Ulrich, H.; Abbracchio, M.; Burnstock, G. Extrinsic purinergic regulation of neural stem/progenitor cells: Implications for CNS development and repair. Stem Cell Reviews and Reports 2012, 8, 755-767.

128. Burnstock, G. Introduction to purinergic signalling in the brain. In Glioma Signaling, Barańska, J., Ed. Springer Netherlands: 2013; Vol. 986, pp 1-12.

129. King, B. F.; Burnstock, G. Purinergic receptors. Understanding G Protein-coupled Receptors and their Role in the CNS 2002, 422-438.

130. Overington, J. P.; Al-Lazikani, B.; Hopkins, A. L. How many drug targets are there? Nature Reviews Drug Discovery 2006, 5, 993-996.

131. Bockaert, J.; Fagni, L.; Dumuis, A.; Marin, P. GPCR interacting proteins (GIP). Pharmacology & Therapeutics 2004, 103, 203-221.

132. Hurowitz, E. H.; Melnyk, J. M.; Chen, Y.-J.; Kouros-Mehr, H.; Simon, M. I.; Shizuya, H. Genomic characterization of the human heterotrimeric G protein α, β, and γ subunit genes. DNA Research 2000, 7, 111-120.

133. Hamm, H. E. The many faces of G protein signaling. Journal of Biological Chemistry 1998, 273, 669-672.

134. Dickenson, J. M.; Hill, S. J. Involvement of G-protein βγ subunits in coupling the adenosine A1 receptor to phospholipase C in transfected CHO cells. European Journal of Pharmacology 1998, 355, 85-93.

135. Snyder, B. Where are the new drugs. The push to improve the pipeline.(Article) Picture by William Oldham 2005.

136. Berne, R. Cardiac nucleotides in hypoxia: possible role in regulation of coronary blood flow. 1963; Vol. 204, p 317-322.

267

137. Olsson, R.; Pearson, J. Cardiovascular purinoceptors. Physiological Reviews 1990, 70, 761-845.

138. Newby, A. C. Adenosine and the concept of ‘retaliatory metabolites’. Trends in Biochemical Sciences 1984, 9, 42-44.

139. Müller, C. E. Adenosine receptors: Therapeutic aspects for inflammatory and immune diseases. Edited by Gyorgy Hasko, Bruce N. Cronstein and Csaba Szabo. ChemMedChem 2008, 3, 1789-1789.

140. Best, B. Is Caffeine a health hazard. The American Journal of Psychiatry 1999, 156, 223-228.

141. Headrick, J. P.; Peart, J. A3 adenosine receptor-mediated protection of the ischemic heart. Vascular Pharmacology 2005, 42, 271-279.

142. Miura, T.; Ogawa, T.; Iwamoto, T.; Shimamoto, K.; Iimura, O. Dipyridamole potentiates the myocardial infarct size-limiting effect of ischemic preconditioning. Circulation 1992, 86, 979-985.

143. Tsuchida, A.; Miura, T.; Miki, T.; Shimamoto, K.; Iimura, O. Role of adenosine receptor activation in myocardial infarct size limitation by ischaemic preconditioning. Cardiovascular Research 1992, 26, 456-461.

144. Miura, T.; Iimura, O. Infarct size limitation by preconditioning: its phenomenological features and the key role of adenosine. Cardiovascular Research 1993, 27, 36-42.

145. Poulsen, S. A.; Quinn, R. J. Adenosine receptors: New opportunities for future drugs. Bioorganic & Medicinal Chemistry 1998, 6, 619-641.

146. Ralevic, V.; Burnstock, G. Receptors for purines and pyrimidines. Pharmacological Reviews 1998, 50, 413-492.

147. Ham, J.; Evans, B. A. An emerging role for adenosine and its receptors in bone homeostasis. Frontiers in endocrinology 2012, 3.

148. Daly, J. W.; Padgett, W.; Thompson, R. D.; Kusachi, S.; Bugni, W. J.; Olsson, R. A. Structure-activity-relationships for N6-substituted adenosines at a brain adenosine-A1-receptor with a comparison to an adenosine-A2 receptor regulating coronary blood-flow. Biochemical Pharmacology 1986, 35, 2467-2481.

149. Daly, J. W. Adenosine receptors: targets for future drugs. Journal of Medicinal Chemistry 1982, 25, 197-207.

150. Yang, Z.; Day, Y. J.; Toufektsian, M. C.; Xu, Y.; Ramos, S. I.; Marshall, M. A.; French, B. A.; Linden, J. Myocardial infarct-sparing effect of adenosine A2A receptor activation is due to its action on CD4+ T lymphocytes. Circulation 2006, 114, 2056-2064.

268

151. Mahaffey, K. W.; Puma, J. A.; Barbagelata, N. A.; Dicarli, M. F.; Leesar, M. A.; Browne, K. F.; Eisenberg, P. R.; Bolli, R.; Casas, A. C.; Molina-Viamonte, V.; Orlandi, C.; Blevins, R.; Gibbons, R. J.; Califf, R. M.; Granger, C. B. Adenosine as an adjunct to thrombolytic therapy for acute myocardial infarction - Results of a multicenter, randomized, placebo-controlled trial: the acute myocardial infarction study of adenosine (AMISTAD) trial. Journal of the American College of Cardiology 1999, 34, 1711-1720.

152. Ross, A. M.; Gibbons, R. J.; Stone, G. W.; Kloner, R. A.; Alexander, R. W. A randomized, double-blinded, placebo-controlled multicenter trial of adenosine as an adjunct to reperfusion in the treatment of acute myocardial infarction (AMISTAD-II). Journal of the American College of Cardiology 2005, 45, 1775-1780.

153. Ninomiya, H.; Otani, H.; Lu, K.; Uchiyama, T.; Kido, M.; Imamura, H. Complementary role of extracellular ATP and adenosine in ischemic preconditioning in the rat heart. American Journal of Physiology-Heart and Circulatory Physiology 2002, 282, H1810-H1820.

154. Freissmuth, M.; Schütz, W.; Linder, M. E. Interactions of the bovine brain A1-adenosine receptor with recombinant G protein alpha-subunits. Selectivity for rGi alpha-3. Journal of Biological Chemistry 1991, 266, 17778-17783.

155. Munshi, R.; Pang, I. H.; Sternweis, P. C.; Linden, J. A1 adenosine receptors of bovine brain couple to guanine nucleotide-binding proteins Gi1, Gi2, and Go. Journal of Biological Chemistry 1991, 266, 22285-9.

156. Calker, D. V.; Muller, M.; Hamprecht, B. Adenosine inhibits the accumulation of cyclic AMP in cultured brain cells. Nature 1978, 276, 839-841.

157. Londos, C.; Cooper, D. M.; Wolff, J. Subclasses of external adenosine receptors. Proceedings of the National Academy of Sciences 1980, 77, 2551-2554.

158. Iredale, P. A.; Alexander, S. P. H.; Hill, S. J. Coupling of a transfected human brain A1 adenosine receptor in CHO-K1 cells to calcium mobilisation via a pertussis toxin-sensitive mechanism. British Journal of Pharmacology 1994, 111, 1252-1256.

159. Megson, A. C.; Dickenson, J. M.; Townsend-Nicholson, A.; Hill, S. J. Synergy between the inositol phosphate responses to transfected human adenosine A1-receptors and constitutive P2-purinoceptors in CHO-K1 cells. British Journal of Pharmacology 1995, 115, 1415-1424.

160. Fredholm, B. B.; IJzerman, A. P.; Jacobson, K. A.; Klotz, K.-N.; Linden, J. International union of pharmacology. XXV. Nomenclature and classification of adenosine receptors. Pharmacological Reviews 2001, 53, 527-552.

161. Reppert, S. M.; Weaver, D. R.; Stehle, J. H.; Rivkees, S. A. Molecular cloning and characterization of a rat A1-adenosine receptor that is widely expressed in brain and spinal cord. Molecular Endocrinology 1991, 5, 1037-1048.

269

162. Stehle, J. H.; Rivkees, S. A.; Lee, J. J.; Weaver, D. R.; Deeds, J. D.; Reppert, S. M. Molecular cloning and expression of the cDNA for a novel A2-adenosine receptor subtype. Molecular Endocrinology 1992, 6, 384-393.

163. Dixon, A. K.; Gubitz, A. K.; Sirinathsinghji, D. J. S.; Richardson, P. J.; Freeman, T. C. Tissue distribution of adenosine receptor mRNAs in the rat. British Journal of Pharmacology 1996, 118, 1461-1468.

164. Tawfik, H. E.; Schnermann, J.; Oldenburg, P. J.; Mustafa, S. J. Role of A1 adenosine receptors in regulation of vascular tone. American Journal of Physiology-Heart and Circulatory Physiology 2005, 288, H1411-H1416.

165. Reichelt, M. E.; Willems, L.; Molina, J. G.; Sun, C.-X.; Noble, J. C.; Ashton, K. J.; Schnermann, J.; Blackburn, M. R.; Headrick, J. P. Genetic deletion of the A1 adenosine receptor limits myocardial ischemic tolerance. Circulation Research 2005, 96, 363-367.

166. Liao, Y.; Takashima, S.; Asano, Y.; Asakura, M.; Ogai, A.; Shintani, Y.; Minamino, T.; Asanuma, H.; Sanada, S.; Kim, J.; Ogita, H.; Tomoike, H.; Hori, M.; Kitakaze, M. Activation of adenosine A1 receptor attenuates cardiac hypertrophy and prevents heart failure in murine left ventricular pressure-overload model. Circulation Research 2003, 93, 759-766.

167. Peart, J.; Headrick, J. P. Intrinsic A1 adenosine receptor activation during ischemia or reperfusion improves recovery in mouse hearts. American Journal of Physiology-Heart and Circulatory Physiology 2000, 279, H2166-H2175.

168. Finegan, B. A.; Lopaschuk, G. D.; Gandhi, M.; Clanachan, A. S. Inhibition of glycolysis and enhanced mechanical function of working rat hearts as a result of adenosine A1 receptor stimulation during reperfusion following ischaemia. British Journal of Pharmacology 1996, 118, 355-363.

169. Merighi, S.; Mirandola, P.; Varani, K.; Gessi, S.; Leung, E.; Baraldi, P. G.; Tabrizi, M. A.; Borea, P. A. A glance at adenosine receptors: novel target for antitumor therapy. Pharmacology & Therapeutics 2003, 100, 31-48.

170. Calker, D. v.; Müller, M.; Hamprecht, B. Adenosine regulates via two different types of receptors, the accumulation of cyclic AMP in cultured brain cells. Journal of Neurochemistry 1979, 33, 999-1005.

171. Bruns, R. F.; Lu, G. H.; Pugsley, T. A. Characterization of the A2 adenosine receptor labeled by [3H] NECA in rat striatal membranes. Molecular Pharmacology 1986, 29, 331-346.

172. Daly, J. W.; Butts-Lamb, P.; Padgett, W. Subclasses of adenosine receptors in the central nervous system: interaction with caffeine and related methylxanthines. Cellular and Molecular Neurobiology 1983, 3, 69-80.

173. Chern, Y.; King, K.; Lai, H.-L.; Lai, H.-T. Molecular cloning of a novel adenosine receptor gene from rat brain. Biochemical and Biophysical Research Communications 1992, 185, 304-309.

270

174. Furlong, T. J.; Pierce, K. D.; Selbie, L. A.; Shine, J. Molecular characterization of a human brain adenosine A2 receptor. Molecular Brain Research 1992, 15, 62-66.

175. Ledent, C.; Vaugeois, J.-M.; Schiffmann, S. N.; Pedrazzini, T.; El Yacoubi, M.; Vanderhaeghen, J.-J.; Costentin, J.; Heath, J. K.; Vassart, G.; Parmentier, M. Aggressiveness, hypoalgesia and high blood pressure in mice lacking the adenosine A2a receptor. Nature 1997, 388, 674-678.

176. Meng, F.; Xie, G.-x.; Chalmers, D.; Morgan, C.; Watson Jr, S. J.; Akil, H. Cloning and expression of the A2A adenosine receptor from guinea pig brain. Neurochemical Research 1994, 19, 613-621.

177. Botvinick, E. H. Current methods of pharmacologic stress testing and the potential advantages of new agents. Journal of Nuclear Medicine Technology 2009, 37, 14-25.

178. Lappas, C. M.; Sullivan, G. W.; Linden, J. Adenosine A2A agonists in development for the treatment of inflammation. Expert Opinion on Investigational Drugs 2005, 14, 797-806.

179. Laubach, V. E.; French, B. A.; Okusa, M. D. Targeting of adenosine receptors in ischemia–reperfusion injury. Expert Opinion on Therapeutic Targets 2011, 15, 103-118.

180. Fishman, P. J., K. A.; Ochaion, A.; Cohen, S.; Bar-Yehuda, S. The anti-cancer effect of A3 adenosine receptor agonists: a novel, targeted therapy. Immun., Endocr. Metab. Agents in Med. Chem 2007, 7.

181. de Lera Ruiz, M.; Lim, Y.-H.; Zheng, J. Adenosine A2A receptor as a drug discovery target. Journal of Medicinal Chemistry 2013, 57(9), 3623-3650.

182. Roberts, P.; Newby, A.; Hallett, M.; Campbell, A. Inhibition by adenosine of reactive oxygen metabolite production by human polymorphonuclear leucocytes. Biochem. J 1985, 227, 669-674.

183. Sullivan, G. W.; Rieger, J. M.; Michael Scheld, W.; Macdonald, T. L.;

Linden, J. Cyclic AMP‐dependent inhibition of human neutrophil oxidative activity by substituted 2‐propynylcyclohexyl adenosine A2A receptor agonists. British Journal of Pharmacology 2001, 132, 1017-1026.

184. Grieshaber, M. C.; Flammer, J. Blood flow in glaucoma. Current Opinion in Ophthalmology 2005, 16, 79-83.

185. Leske, M. C. Ocular perfusion pressure and glaucoma: clinical trial and epidemiologic findings. Current Opinion in Ophthalmology 2009, 20, 73.

186. Konno, T.; Uchibori, T.; Nagai, A.; Kogi, K.; Nakahata, N. Effect of 2-(6-cyano-1-hexyn-1-yl) adenosine on ocular blood flow in rabbits. Life sciences 2007, 80, 1115-1122.

271

187. Sitkovsky, M. V.; Lukashev, D.; Apasov, S.; Kojima, H.; Koshiba, M.; Caldwell, C.; Ohta, A.; Thiel, M. Physiological control of immune response and inflammatory tissue damage by hypoxia-inducible factors and adenosine a2a receptors*. Annual Review Immunology 2004, 22, 657-682.

188. Cronstein, B. N. Adenosine, an endogenous anti-inflammatory agent. Journal of Applied Physiology 1994, 76, 5-13.

189. Feoktistov, I.; Goldstein, A. E.; Biaggioni, I. Role of p38 mitogen-activated protein kinase and extracellular signal-regulated protein kinase kinase in adenosine a2breceptor-mediated interleukin-8 production in human mast cells. Molecular Pharmacology 1999, 55, 726-734.

190. Feoktistov, I.; Ryzhov, S.; Goldstein, A. E.; Biaggioni, I. Mast cell–mediated stimulation of angiogenesis cooperative interaction between A2B and A3 adenosine receptors. Circulation Research 2003, 92, 485-492.

191. Auchampach, J. A.; Jin, X.; Wan, T. C.; Caughey, G. H.; Linden, J. Canine mast cell adenosine receptors: cloning and expression of the A3 receptor and evidence that degranulation is mediated by the A2B receptor. Molecular Pharmacology 1997, 52, 846-860.

192. Ryzhov, S.; Goldstein, A. E.; Matafonov, A.; Zeng, D.; Biaggioni, I.; Feoktistov, I. Adenosine-activated mast cells induce IgE synthesis by B lymphocytes: an A2B-mediated process involving Th2 cytokines IL-4 and IL-13 with implications for asthma. The Journal of Immunology 2004, 172, 7726-7733.

193. Volpini, R.; Costanzi, S.; Vittori, S.; Cristalli, G.; Klotz, K.-N. Medicinal chemistry and pharmacology of A2B adenosine receptors. Current Topics in Medicinal Chemistry 2003, 3, 427-443.

194. Eckle, T.; Krahn, T.; Grenz, A.; Köhler, D.; Mittelbronn, M.; Ledent, C.; Jacobson, M. A.; Osswald, H.; Thompson, L. F.; Unertl, K. Cardioprotection by ecto-5′-nucleotidase (CD73) and A2B adenosine receptors. Circulation 2007, 115, 1581-1590.

195. Toldo, S.; Zhong, H.; Mezzaroma, E.; Van Tassell, B. W.; Kannan, H.; Zeng, D.; Belardinelli, L.; Voelkel, N. F.; Abbate, A. GS-6201, a selective blocker of the A2B adenosine receptor, attenuates cardiac remodeling after acute myocardial infarction in the mouse. Journal of Pharmacology and Experimental Therapeutics 2012, 343, 587-595.

196. Karmouty-Quintana, H.; Zhong, H.; Acero, L.; Weng, T.; Melicoff, E.; West, J. D.; Hemnes, A.; Grenz, A.; Eltzschig, H. K.; Blackwell, T. S. The A2B

adenosine receptor modulates pulmonary hypertension associated with interstitial lung disease. The FASEB Journal 2012, 26, 2546-2557.

197. Zimmerman, M. A.; Grenz, A.; Tak, E.; Kaplan, M.; Ridyard, D.; Brodsky, K. S.; Mandell, M. S.; Kam, I.; Eltzschig, H. K. Signaling through hepatocellular A2B adenosine receptors dampens ischemia and reperfusion injury of the liver. Proceedings of the National Academy of Sciences 2013, 110, 12012-12017.

272

198. Frick, J.-S.; MacManus, C. F.; Scully, M.; Glover, L. E.; Eltzschig, H. K.; Colgan, S. P. Contribution of adenosine A2B receptors to inflammatory parameters of experimental colitis. The Journal of Immunology 2009, 182, 4957-4964.

199. Gessi, S.; Merighi, S.; Varani, K.; Leung, E.; Mac Lennan, S.; Borea, P. A. The A3 adenosine receptor: An enigmatic player in cell biology. Pharmacology & Therapeutics 2008, 117, 123-140.

200. Schulte, G.; Fredholm, B. B. Signalling from adenosine receptors to mitogen-activated protein kinases. Cellular Signalling 2003, 15, 813-827.

201. Zhou, Q.-Y.; Li, C.; Olah, M. E.; Johnson, R. A.; Stiles, G. L.; Civelli, O. Molecular cloning and characterization of an adenosine receptor: the A3 adenosine receptor. Proceedings of the National Academy of Sciences 1992, 89, 7432-7436.

202. Linden, J. Cloned adenosine A3 receptors: pharmacological properties, species differences and receptor functions. Trends in Pharmacological Sciences 1994, 15, 298-306.

203. Rivkees, S. A.; Thevananther, S.; Hao, H. Are A3 adenosine receptors expressed in the brain? Neuroreport 2000, 11, 1025-1030.

204. Haskó, G.; Linden, J.; Cronstein, B.; Pacher, P. Adenosine receptors: therapeutic aspects for inflammatory and immune diseases. Nature Reviews Drug Discovery 2008, 7, 759-770.

205. Gessi, S.; Cattabriga, E.; Avitabile, A.; Gafa, R.; Lanza, G.; Cavazzini, L.; Bianchi, N.; Gambari, R.; Feo, C.; Liboni, A.; Gullini, S.; Leung, E.; Mac-Lennan, S.; Borea, P. A. Elevated expression of A3 adenosine receptors in human colorectal cancer is reflected in peripheral blood cells. Clinical Cancer Research 2004, 10, 5895-5901.

206. Madi, L.; Ochaion, A.; Rath-Wolfson, L.; Bar-Yehuda, S.; Erlanger, A.; Ohana, G.; Harish, A.; Merimski, O.; Barer, F.; Fishman, P. The A3 adenosine receptor is highly expressed in tumor versus normal cells: Potential target for tumor growth inhibition. Clinical Cancer Research 2004, 10, 4472-4479.

207. Ochaion, A.; Bar-Yehuda, S.; Cohen, S.; Barer, F.; Patoka, R.; Amital, H.; Reitblat, T.; Reitblat, A.; Ophir, J.; Konfino, I.; Chowers, Y.; Ben-Horin, S.; Fishman, P. The anti-inflammatory target A3 adenosine receptor is over-expressed in rheumatoid arthritis, psoriasis and Crohn's disease. Cellular Immunology 2009, 258, 115-122.

208. Rajagopal, S.; Rajagopal, K.; Lefkowitz, R. J. Teaching old receptors new tricks: biasing seven-transmembrane receptors. Nature Reviews Drug Discovery 2010, 9, 373-386.

209. Patel, C. B.; Noor, N.; Rockman, H. A. Functional Selectivity in Adrenergic and Angiotensin Signaling Systems. Molecular Pharmacology 2010, 78, 983-992.

273

210. Kenakin, T. Functional Selectivity and Biased Receptor Signaling. Journal of Pharmacology and Experimental Therapeutics 2011, 336, 296-302.

211. Whalen, E. J.; Rajagopal, S.; Lefkowitz, R. J. Therapeutic potential of β-arrestin- and G protein-biased agonists. Trends in Molecular Medicine 2011, 17, 126-139.

212. Albrecht-Küpper, B. E.; Leineweber, K.; Nell, P. G. Partial adenosine A1 receptor agonists for cardiovascular therapies. Purinergic Signalling 2012, 8, 91-99.

213. Jakubowski, H. Transport and kinetics, biochemistry online: An approach based on chemical logic. In Agonist and antagonist of ligand binding, biowiki ucdavis.edu

214. Moro, S.; Gao, Z. G.; Jacobson, K. A.; Spalluto, G. Progress in the pursuit of therapeutic adenosine receptor antagonists. Medicinal Research Reviews 2006, 26, 131-159.

215. Feoktistov, I.; Biaggioni, I.; Polosa, R.; Holgate, S. Adenosine A2B receptors: a novel therapeutic target in asthma? Trends in pharmacological sciences 1998, 19, 148-153.

216. Fozard, J.; McCarthy, C. Adenosine receptor ligands as potential therapeutics in asthma. Current opinion in investigational drugs (London, England: 2000) 2002, 3, 69-77.

217. Richardson, P. J.; Kase, H.; Jenner, P. G. Adenosine A2A receptor antagonists as new agents for the treatment of Parkinson's disease. Trends in Pharmacological Sciences 1997, 18, 338-344.

218. El-Tayeb, A.; Michael, S.; Abdelrahman, A.; Behrenswerth, A.; Gollos, S.; Nieber, K.; Muller, C. E. Development of polar adenosine A2A receptor agonists for inflammatory bowel disease: synergism with A2B antagonists. ACS Medicinal Chemistry Letters 2011, 2, 890-895.

219. Hendel, R.; Stilley, W. B.; Williams, S. P. Unit Dosage of Apadenoson. U. S. Patent application No 14/103,130, 2013.

220. Harada, N.; Okajima, K.; Murakami, K.; Usune, S.; Sato, C.; Ohshima, K.; Katsuragi, T. Adenosine and selective A2A receptor agonists reduce ischemia/reperfusion injury of rat liver mainly by inhibiting leukocyte activation. Journal of Pharmacology and Experimental Therapeutics 2000, 294, 1034-1042.

221. Yoneyama, F.; Yamada, H.; Satoh, K.; Taira, N. Vasodepressor mechanisms of 2-(1-octynyl)-adenosine (YT-146), a selective adenosine A2 receptor agonist, involve the opening of glibenclamide-sensitive K+ channels. European Journal of Pharmacology 1992, 213, 199-204.

222. Deflorian, F.; Kumar, T. S.; Phan, K.; Gao, Z.-G.; Xu, F.; Wu, H.; Katritch, V.; Stevens, R. C.; Jacobson, K. A. Evaluation of molecular modeling of agonist

274

binding in light of the crystallographic structure of an agonist-bound A2A adenosine receptor. Journal of Medicinal Chemistry 2011, 55, 538-552.

223. Hutchison, A. J.; Williams, M.; De Jesus, R.; Yokoyama, R.; Oei, H. H.; Ghai, G. R.; Webb, R. L.; Zoganas, H. C.; Stone, G. A.; Jarvis, M. F. 2-(Arylalkylamino)adenosin-5'-uronamides: a new class of highly selective adenosine A2 receptor ligands. Journal of Medicinal Chemistry 1990, 33, 1919-1924.

224. Mayer, C. A. Adenosine A2A receptors mediate GABAergic inhibition of respiration in immature rats. 2006; Vol. 100, p 91-97.

225. Diógenes, M. J.; Fernandes, C. C.; Sebastião, A. M.; Ribeiro, J. A. Activation of adenosine A2A receptor facilitates brain-derived neurotrophic factor modulation of synaptic transmission in hippocampal slices. The Journal of Neuroscience 2004, 24, 2905-2913.

226. Zoghbi, G. J. M. D.; Iskandrian, A. E. M. D. Selective adenosine agonists and myocardial perfusion imaging. Journal of Nuclear Cardiology 2012, 19, 126-41.

227. Müller, C. E.; Jacobson, K. A. Recent developments in adenosine receptor ligands and their potential as novel drugs. Biochimica et Biophysica Acta (BBA) - Biomembranes 2011, 1808, 1290-1308.

228. Brackett, L. E.; Daly, J. W. Relaxant effects of adenosine analogs on guinea pig trachea in vitro: xanthine-sensitive and xanthine-insensitive mechanisms. Journal of Pharmacology and Experimental Therapeutics 1991, 257, 205-213.

229. Mathôt, R. A.; Gubbens-Stibbe, J. M.; Soudijn, W.; Jacobson, K. A.; Ijzerman, A. P.; Danhof, M. Quantification of the in vivo potency of the adenosine A2 receptor antagonist 8-(3-chlorostyryl)caffeine. Journal of Pharmacology and Experimental Therapeutics 1995, 275, 245-53.

230. LeWitt, P. A.; Guttman, M.; Tetrud, J. W.; Tuite, P. J.; Mori, A.; Chaikin,

P.; Sussman, N. M. Adenosine A2A receptor antagonist istradefylline (KW‐6002) reduces “off” time in Parkinson's disease: A double‐blind, randomized, multicenter clinical trial (6002‐US‐005). Annals of Neurology 2008, 63, 295-302.

231. Mizuno, Y.; Hasegawa, K.; Kondo, T.; Kuno, S.; Yamamoto, M. Clinical efficacy of istradefylline (KW-6002) in Parkinson's disease: A randomized, controlled study. Movement Disorders 2010, 25, 1437-1443.

232. Jaakola, V.-P.; Griffith, M. T.; Hanson, M. A.; Cherezov, V.; Chien, E. Y. T.; Lane, J. R.; Ijzerman, A. P.; Stevens, R. C. The 2.6 angstrom crystal structure of a human [image] adenosine receptor bound to an antagonist. Science 2008, 322, 1211-1217.

233. Salamone, J. D. Preladenant, a novel adenosine A (2A) receptor antagonist for the potential treatment of parkinsonism and other disorders. IDrugs: The Investigational Drugs Journal 2010, 13, 723-731.

275

234. Drury, A.; Szent-Györgyi, A. v. The physiological activity of adenine compounds with especial reference to their action upon the mammalian heart. The Journal of Physiology 1929, 68, 213-237.

235. Lebon, G.; Warne, T.; Edwards, P. C.; Bennett, K.; Langmead, C. J.; Leslie, A. G. W.; Tate, C. G. Agonist-bound adenosine A2A receptor structures reveal common features of GPCR activation. Nature 2011, 474, 521-5.

236. Ely, S.; Berne, R. Protective effects of adenosine in myocardial ischemia. Circulation 1992, 85, 893-904.

237. Peart, J.; Paul Matherne, G.; Cerniway, R. J.; Headrick, J. P. Cardioprotection with adenosine metabolism inhibitors in ischemic–reperfused mouse heart. Cardiovascular Research 2001, 52, 120-129.

238. Baraldi, P. G.; Tabrizi, M. A.; Fruttarolo, F.; Romagnoli, R.; Preti, D. Recent improvements in the development of A2B adenosine receptor agonists. Purinergic Signalling 2008, 4, 287-303.

239. Auchampach, J. A.; Kreckler, L. M.; Wan, T. C.; Maas, J. E.; van der Hoeven, D.; Gizewski, E.; Narayanan, J.; Maas, G. E. Characterization of the A2B adenosine receptor from mouse, rabbit, and dog. Journal of Pharmacology and Experimental Therapeutics 2009, 329, 2-13.

240. Rosentreter, U.; Henning, R.; Bauser, M.; Krämer, T.; Vaupel, A.; Hübsch, W.; Dembowsky, K.; Salcher-Schraufstätter, O.; Stasch, J.-P.; Krahn, T. Substituted 2-thio-3, 5-dicyano-4-aryl-6-aminopyridines and the use thereof. U.S. Patent No. 7,135,486, 2006.

241. Beukers, M. W.; Chang, L. C.; von Frijtag Drabbe Künzel, J. K.; Mulder-Krieger, T.; Spanjersberg, R. F.; Brussee, J.; IJzerman, A. P. New, non-adenosine, high-potency agonists for the human adenosine A2B receptor with an improved selectivity profile compared to the reference agonist N-ethylcarboxamidoadenosine. Journal of Medicinal Chemistry 2004, 47, 3707-3709.

242. Kawazoe, K.; Matsumoto, N.; Tanabe, M.; Fujiwara, S.; Yanagimoto, M.; Hirata, M.; Kikuchi, K. Coronary and cardiohemodynamic effects of 2-phenylamino-adenosine (CV-1808) in anesthetized dogs and cats. Arzneimittelforschung Drug Research 1980.

243. Abiru, T.; Miyashita, T.; Watanabe, Y.; Yamaguchi, T.; Machida, H.; Matsuda, A. Nucleosides and nucleotides. 107. 2-(cycloalkylalkynyl) adenosines: adenosine A2 receptor agonists with potent antihypertensive effects. Journal of Medicinal Chemistry 1992, 35, 2253-2260.

244. Thimm, D.; Schiedel, A. C.; Sherbiny, F. F.; Hinz, S.; Hochheiser, K.; Bertarelli, D. C.; Maaß, A.; Muller, C. E. Ligand-specific binding and activation of the human adenosine A2B receptor. Biochemistry 2013, 52, 726-740.

276

245. Hinz, S.; Lacher, S. K.; Seibt, B. F.; Müller, C. E. BAY60-6583 acts as a partial agonist at adenosine A2B receptors. Journal of Pharmacology and Experimental Therapeutics 2014, 349, 427-436.

246. Kim, S.-A.; Marshall, M. A.; Melman, N.; Kim, H. S.; Müller, C. E.; Linden, J.; Jacobson, K. A. Structure-activity relationships at human and rat A2B adenosine receptors of xanthine derivatives substituted at the 1-, 3-, 7-, and 8-positions. Journal of Medicinal Chemistry 2002, 45, 2131-2138.

247. Kim, Y.-C.; Ji, X.-d.; Melman, N.; Linden, J.; Jacobson, K. A. Anilide derivatives of an 8-phenylxanthine carboxylic congener are highly potent and selective antagonists at human A2B adenosine receptors. Journal of Medicinal Chemistry 2000, 43, 1165-1172.

248. Watanabe, T.; Tanaka, M.; Watanabe, K.; Takamatsu, Y.; Tobe, A. [Research and development of the free radical scavenger edaravone as a neuroprotectant]. Yakugaku Zasshi Journal of the Pharmaceutical Society of Japan 2004, 99-111.

249. Batchu, S.; Lee, S.; Qadhi, R.; Chaudhary, K.; El‐Sikhry, H.; Kodela, R.; Falck, J.; Seubert, J. Cardioprotective effect of a dual acting epoxyeicosatrienoic acid analogue towards ischaemia reperfusion injury. British Journal of Pharmacology 2011, 162, 897-907.

250. Jacobson, K. A.; Gao, Z.-G. Adenosine receptors as therapeutic targets. Nature Reviews Drug discovery 2006, 5, 247-264.

251. Jacobson, K. A.; Xie, R.; Young, L.; Chang, L.; Liang, B. T. A novel pharmacological approach to treating cardiac ischemia binary conjugates of A1 and A3 adenosine receptor agonists. Journal of Biological Chemistry 2000, 275, 30272-30279.

252. Liang, B. T.; Jacobson, K. A. Methods and compositions for reducing ischemic injury of the heart by administering adenosine receptor agonists and antagonists. U. S. Patents 6586413, 2003.

253. Hutchison, A.; Oei, H.; Ghai, G.; Williams, M. CGS21680, an A2 selective adenosine (ADO) receptor agonist with preferential hypotensive activity. FASEB J 1989, 3, A281.

254. Cristalli, G.; Eleuteri, A.; Vittori, S.; Volpini, R.; Lohse, M. J.; Klotz, K. N. 2-Alkynyl derivatives of adenosine and adenosine-5'-N-ethyluronamide as selective agonists at A2 adenosine receptors. Journal of Medicinal Chemistry 1992, 35, 2363-2368.

255. Martin, P. L.; Barrett, R. J.; Linden, J.; Abraham, W. M. Pharmacology of

2‐cyclohexylmethylidenehydrazinoadenosine (WRC‐0470), a novel, short‐acting adenosine A2A receptor agonist that produces selective coronary vasodilation. Drug Development Research 1997, 40, 313-324.

277

256. Yan, L.; Burbiel, J. C.; Maaß, A.; Müller, C. E. Adenosine receptor agonists: from basic medicinal chemistry to clinical development. Expert Opinion on Emerging Drugs 2003, 8, 537-576.

257. Ohta, A.; Sitkovsky, M. Role of G-protein-coupled adenosine receptors in downregulation of inflammation and protection from tissue damage. Nature 2001, 414, 916-920.

258. Gregg, A.; Bottle, S. E.; Devine, S. M.; Figler, H.; Linden, J.; White, P.; Pouton, C. W.; Urmaliya, V.; Scammells, P. J. Dual acting antioxidant A1 adenosine receptor agonists. Bioorganic & Medicinal Chemistry Letters 2007, 17, 5437-5441.

259. Foitzik, R. C.; Devine, S. M.; Hausler, N. E.; Scammells, P. J. Linear and convergent approaches to 2-substituted adenosine-5′-N-alkylcarboxamides. Tetrahedron 2009, 65, 8851-8857.

260. Xu, F.; Wu, H.; Katritch, V.; Han, G. W.; Jacobson, K. A.; Gao, Z.-G.; Cherezov, V.; Stevens, R. C. Structure of an agonist-bound human A2A adenosine receptor. Science 2011, 332, 322-327.

261. Mantell, S.; Jones, R.; Trevethick, M. Design and application of locally delivered agonists of the adenosine A2A receptor. Expert Review of Clinical Pharmacology 2010, 3, 55-72.

262. Bosch, M. P.; Campos, F.; Niubó, I.; Rosell, G.; Díaz, J. L.; Brea, J.; Loza, M. I.; Guerrero, A. Synthesis and biological activity of new potential agonists for the human adenosine A2A receptor. Journal of Medicinal Chemistry 2004, 47, 4041-4053.

263. Francis, J. E.; Webb, R. L.; Ghai, G. R.; Hutchison, A. J.; Moskal, M. A.; DeJesus, R.; Yokoyama, R.; Rovinski, S. L.; Contardo, N. Highly selective adenosine A2 receptor agonists in a series of N-alkylated 2-aminoadenosines. Journal of Medicinal Chemistry 1991, 34, 2570-2579.

264. Jacobson, M. A. Adenosine receptor agonists. Expert Opinion on Therapeutic Patents 2002, 12, 489-501.

265. Trivedi, B. K.; Bruns, R. F. C2, N6-Disubstituted adenosines: synthesis and structure-activity relationships. Journal of Medicinal Chemistry 1989, 32, 1667-1673.

266. Cristalli, G.; Lambertucci, C.; Taffi, S.; Vittori, S.; Volpini, R. Medicinal chemistry of adenosine A2A receptor agonists. Current Topics in Medicinal Chemistry 2003, 3, 387-401.

267. Niiya, K.; Thompson, R. D.; Silvia, S. K.; Olsson, R. A. 2-(N'-aralkylidenehydrazino) adenosines: potent and selective coronary vasodilators. Journal of Medicinal Chemistry 1992, 35, 4562-4566.

278

268. Rieger, J. M.; Brown, M. L.; Sullivan, G. W.; Linden, J.; Macdonald, T. L. Design, synthesis, and evaluation of novel A2A adenosine receptor agonists. Journal of Medicinal Chemistry 2001, 44, 531-539.

269. Manera, C.; Saccomanni, G. A2A receptor ligands: past, present and future trends. Current Topics in Medicinal Chemistry 2010, 10, 902-922.

270. Hutchinson, S.; Scammells, P. A1 adenosine receptor agonists: medicinal chemistry and therapeutic potential. Current Pharmaceutical Design 2004, 10, 2021-2039.

271. Adachi, H.; Palaniappan, K. K.; Ivanov, A. A.; Bergman, N.; Gao, Z.-G.; Jacobson, K. A. Structure−activity relationships of 2,N6,5‘-substituted adenosine derivatives with potent activity at the A2B adenosine receptor. Journal of Medicinal Chemistry 2007, 50, 1810-1827.

272. Caddell, J. M.; Chapman, A. M.; Cooley, B. E.; Downey, B. P.; LeBlanc, M. P.; Jackson, M. M.; O'Connell, T. M.; Phung, H.-M.; Roper, T. D.; Xie, S. Efficient synthesis of an adenosine A2A agonist: Glycosylation of 2-haloadenines and an N 2-alkyl-6-chloroguanine. The Journal of Organic Chemistry 2004, 69, 3212-3215.

273. Kim, H. S.; Ohno, M.; Xu, B.; Kim, H. O.; Choi, Y.; Ji, X. D.; Maddileti, S.; Marquez, V. E.; Harden, T. K.; Jacobson, K. A. 2-Substitution of adenine nucleotide analogues containing a bicyclo[3.1.0]hexane ring system locked in a northern conformation:  Enhanced potency as P2Y1 receptor antagonists. Journal of Medicinal Chemistry 2003, 46, 4974-4987.

274. Kim, H. O.; Ji, X.-d.; Siddiqi, S. M.; Olah, M. E.; Stiles, G. L.; Jacobson, K. A. 2-Substitution of N6-benzyladenosine-5'-uronamides enhances selectivity for A3 adenosine receptors. Journal of Medicinal Chemistry 1994, 37, 3614-3621.

275. Bottle, S. E.; Gillies, D. G.; Hughes, D. L.; Micallef, A. S.; Smirnov, A. I.; Sutcliffe, L. H. Synthesis, single crystal X-ray structure and W-band (95 GHz) EPR spectroscopy of a new anionic isoindoline aminoxyl: synthesis and characterisation of some derivatives. Journal of the Chemical Society, Perkin Transactions 2 2000, 1285-1291.

276. S. Micallef, A.; C. Bott, R.; E. Bottle, S.; Smith, G.; M. White, J.; Matsuda, K.; Iwamura, H. Brominated isoindolines: precursors to functionalised nitroxides. Journal of the Chemical Society, Perkin Transactions 2 1999, 65-72.

277. Howell, S. J.; Spencer, N.; Philp, D. Recognition-mediated regiocontrol of a dipolar cycloaddition reaction. Tetrahedron 2001, 57, 4945-4954.

278. Wuts, P. G.; Greene, T. W. Greene's protective groups in organic synthesis. John Wiley & Sons: 2006.

279. Olsson, R. A.; Kusachi, S.; Thompson, R. D.; Ukena, D.; Padgett, W.; Daly, J. W. N6-Substituted N-alkyladenosine-5'-uronamides: bifunctional ligands

279

having recognition groups for A1 and A2 adenosine receptors. Journal of Medicinal Chemistry 1986, 29, 1683-1689.

280. Maenhaut, C.; Van Sande, J.; Libert, F.; Abramowicz, M.; Parmentier, M.; Vanderhaegen, J.-J.; Dumont, J. E.; Vassart, G.; Schiffmann, S. RDC8 codes for an adenosine A2 receptor with physiological constitutive activity. Biochemical and Biophysical Research Communications 1990, 173, 1169-1178.

281. Devine, S. M.; Scammells, P. J. An efficient convergent synthesis of adenosine-5′-N-alkyluronamides. Tetrahedron 2008, 64, 1772-1777.

282. Bookser, B. C.; Raffaele, N. B. High-throughput five minute microwave accelerated glycosylation approach to the synthesis of nucleoside libraries. The Journal of Organic Chemistry 2007, 72, 173-179.

283. Hutchinson, S. A.; Baker, S. P.; Linden, J.; Scammells, P. J. New potent and selective A1 adenosine receptor agonists. Bioorganic & Medicinal Chemistry 2004, 12, 4877-4884.

284. Ahmadibeni, Y.; Dash, C.; Le Grice, S. F.; Parang, K. Solid-phase synthesis of 5′-O-β, γ-methylenetriphosphate derivatives of nucleosides and evaluation of their inhibitory activity against HIV-1 reverse transcriptase. Tetrahedron Letters 2010, 51, 3010-3013.

285. El-Farargy, A.; Ghoneim, A. A. Synthesis of Some Purine Nucleoside Derivatives with Expected Biological Activity. Current Organic Chemistry 2009, 13, 1842-1847.

286. Chen, G. S.; Chen, C. S.; Chien, T. C.; Yeh, J. Y.; Kuo, C. C.; Talekar, R.

S.; Chern, J. W. Nucleosides. IX. Synthesis of purine N 3, 5′‐cyclonucleosides and N 3, 5′‐Cyclo‐2′, 3′‐seconucleosides via Mitsunobu reaction as TIBO‐like derivatives. Nucleosides, Nucleotides and Nucleic Acids 2004, 23, 347-359.

287. Aerschot, A. V.; Mag, M.; Herdewijn, P.; Vanderhaeghe, H. Double protection of the heterocyclic base of xanthosine and 2′-deoxyxanthosine. Nucleosides & Nucleotides 1989, 8, 159-178.

288. Hampton, A. Nucleotides. II.1 A new procedure for the conversion of ribonucleosides to 2',3'-O-isopropylidene derivatives. Journal of the American Chemical Society 1961, 83, 3640-3645.

289. Ashton, T. D.; Scammells, P. J. An improved synthesis of 5′-fluoro-5′-deoxyadenosines. Bioorganic & Medicinal Chemistry Letters 2005, 15, 3361-3363.

290. Blum, T.; Ermert, J.; Wutz, W.; Bier, D.; Coenen, H. H. First no-carrier-added radioselenation of an adenosine-A1 receptor ligand. Journal of Labelled Compounds and Radiopharmaceuticals 2004, 47, 415-427.

291. Ciuffreda, P.; Loseto, A.; Santaniello, E. Deamination of 5′-substituted-2′,3′-isopropylidene adenosine derivatives catalyzed by adenosine deaminase (ADA, EC 3.5.4.4) and complementary enzymatic biotransformations catalyzed

280

by adenylate deaminase (AMPDA, EC 3.5.4.6): a viable route for the preparation of 5′-substituted inosine derivatives. Tetrahedron 2002, 58, 5767-5771.

292. Middleton, R. J.; Briddon, S. J.; Cordeaux, Y.; Yates, A. S.; Dale, C. L.; George, M. W.; Baker, J. G.; Hill, S. J.; Kellam, B. New fluorescent adenosine A1-receptor agonists that allow quantification of ligand−receptor interactions in microdomains of single living cells. Journal of Medicinal Chemistry 2007, 50, 782-793.

293. De Mico, A.; Margarita, R.; Parlanti, L.; Vescovi, A.; Piancatelli, G. A versatile and highly selective hypervalent iodine (iii)/2,2,6,6-tetramethyl-1-piperidinyloxyl-mediated oxidation of alcohols to carbonyl compounds. The Journal of Organic Chemistry 1997, 62, 6974-6977.

294. Singh, A. K.; Varma, R. S. Ruthenium tetraoxide: a mild reagent for the oxidation of 2′,3′-O-isopropylidene purine nucleosides. Tetrahedron Letters 1992, 33, 2307-2310.

295. Marcus M. Sá, G. P. S., Marcelo S. Castilho, Fernando Pavão and Glaucius Oliva. Synthesis of acylated nucleosides and ribonic-1,4-lactones as inhibitors of trypanosomal glyceraldehyde-3-phosphate dehydrogenase (gGAPDH) (AP-507HP) ARKIVOC: (Gainesville, FL United States) 2002, 8, 112-124.

296. Epp, J. B.; Widlanski, T. S. Facile preparation of nucleoside-5'-carboxylic acids. The Journal of Organic Chemistry 1999, 64, 293-295.

297. Nooy, A. E. J. d.; Besemer, A. C.; Bekkum, H. v. On the use of stable organic nitroxyl radicals for the oxidation of primary and secondary alcohols. Synthesis 1996, 1996, 1153-1176.

298. Smith, M. B., March, J. . March’s Advanced Organic Chemistry: Reactions, Mechanisms, and Structure.; John Wiley & Sons, Inc. Hoboken, N.J.: 6 ed. 2007, 1254-1255.

299. Wan, Z.-K.; Wacharasindhu, S.; Binnun, E.; Mansour, T. An efficient direct amination of cyclic amides and cyclic ureas. Organic Letters 2006, 8, 2425-2428.

300. Bae, S.; Lakshman, M. K. Synthetic utility of an isolable nucleoside phosphonium salt. Organic Letters 2008, 10, 2203-2206.

301. Smith, M. B., March, J. March’s Advanced Organic Chemistry: Reactions, Mechanisms, and Structure. John Wiley & Sons, Inc.: Hoboken, N.J 6 th ed, 2007, 854-857.

302. Kang, F.-A.; Sui, Z.; Murray, W. V. Phosphonium Coupling in the Direct Bond Formations of Tautomerizable Heterocycles via C–OH Bond Activation. European Journal of Organic Chemistry 2009, 2009, 461-479.

281

303. Takvorian, A. G.; Combs, A. P. Microwave-assisted organic synthesis using minivials to optimize and expedite the synthesis of diverse purine libraries. Journal of Combinatorial Chemistry 2004, 6, 171-174.

304. Sholle, V. D. Akad. Nauk. SSSR, Ser, Khim. 1969, 1, 149-151.

305. Keana, J. F. W.; Heo, G. S.; Gaughan, G. T. Stereospecific synthesis of difunctionalized 2,5-disubstituted cis-2,5-dimethylpyrrolidine (azethoxyl) nitroxides by oxidative cleavage of protected 8-azabicyclo[3.2.1]octane precursors. The Journal of Organic Chemistry 1985, 50, 2346-2351.

306. Rogister, F.; Laeckmann, D.; Plasman, P.-O.; Van Eylen, F.; Ghyoot, M.; Maggetto, C.; Liégeois, J.-F.; Géczy, J.; Herchuelz, A.; Delarge, J.; Masereel, B. Novel inhibitors of the sodium–calcium exchanger: benzene ring analogues of N-guanidino substituted amiloride derivatives. European Journal of Medicinal Chemistry 2001, 36, 597-614.

307. Ferguson, G. N. Synthesis and pharmacological evaluation of novel heteroaromatic ligands of the A1 adenosine receptor. . PhD, Monash University, Melbourne 2010.

308. Urmaliya, V. B.; Church, J. E.; Coupar, I. M.; Rose'Meyer, R. B.; Pouton, C. W.; White, P. J. Cardioprotection induced by adenosine A1 receptor agonists in a cardiac cell ischemia model involves cooperative activation of adenosine A2A and A2B receptors by endogenous adenosine. Journal of Cardiovascular Pharmacology 2009, 53, 424-433.

309. Urmaliya, V. B.; Pouton, C. W.; Devine, S. M.; Haynes, J. M.; Warfe, L.; Scammells, P. J.; White, P. J. A novel highly selective adenosine A1 receptor agonist VCP28 reduces ischemia injury in a cardiac cell line and ischemia–reperfusion injury in isolated rat hearts at concentrations that do not affect heart rate. Journal of Cardiovascular Pharmacology 2010, 56, 282-292.

310. Kolamunne, R. T.; Clare, M.; Griffiths, H. R. Mitochondrial superoxide anion radicals mediate induction of apoptosis in cardiac myoblasts exposed to chronic hypoxia. Archives of biochemistry and biophysics 2011, 505, 256-265.

311. Cristalli, G.; Volpini, R.; Vittori, S.; Camaioni, E.; Monopoli, A.; Conti, A.; Dionisotti, S.; Zocchi, C.; Ongini, E. 2-Alkynyl derivatives of adenosine-5'-N-ethyluronamide: selective A2 adenosine receptor agonists with potent inhibitory activity on platelet aggregation. Journal of Medicinal Chemistry 1994, 37, 1720-1726.

312. Dal Ben, D.; Lambertucci, C.; Taffi, S.; Vittori, S.; Volpini, R.; Cristalli, G.; Klotz, K.-N. Molecular modelling study of 2-phenylethynyladenosine (PEAdo) derivatives as highly selective A3 adenosine receptor ligands. Purinergic Signalling 2006, 2, 589-594.

313. Kim, Y.-C.; Ji, X.-d.; Jacobson, K. A. Derivatives of the triazoloquinazoline adenosine antagonist (CGS15943) are selective for the human A3 receptor subtype. Journal of Medicinal Chemistry 1996, 39, 4142-4148.

282

314. Kim, S.-K.; Gao, Z.-G.; Jeong, L. S.; Jacobson, K. A. Docking studies of agonists and antagonists suggest an activation pathway of the A3 adenosine receptor. Journal of Molecular Graphics and Modelling 2006, 25, 562-577.

315. Jacobson, K. A.; Klutz, A. M.; Tosh, D. K.; Ivanov, A. A.; Preti, D.; Baraldi, P. G. Medicinal chemistry of the A3 adenosine receptor: agonists, antagonists, and receptor engineering. In Adenosine Receptors in Health and Disease, Springer: 2009; pp 123-159.

316. Keddie, D. J.; Fairfull-Smith, K. E.; Bottle, S. E. The palladium-catalysed copper-free Sonogashira coupling of isoindoline nitroxides: a convenient route to robust profluorescent carbon-carbon frameworks. Organic & Biomolecular Chemistry 2008, 6, 3135-3143.

317. Cassar, L. Synthesis of aryl-and vinyl-substituted acetylene derivatives by the use of nickel and palladium complexes. Journal of Organometallic Chemistry 1975, 93, 253-257.

318. Dieck, H.; Heck, F. Palladium catalyzed synthesis of aryl, heterocyclic and vinylic acetylene derivatives. Journal of Organometallic Chemistry 1975, 93, 259-263.

319. Sonogashira, K.; Tohda, Y.; Hagihara, N. A convenient synthesis of acetylenes: catalytic substitutions of acetylenic hydrogen with bromoalkenes, iodoarenes and bromopyridines. Tetrahedron letters 1975, 16, 4467-4470.

320. Stephens, R.; Castro, C. The Substitution of Aryl Iodides with Cuprous Acetylides. A Synthesis of Tolanes and Heterocyclics1. The Journal of organic chemistry 1963, 28, 3313-3315.

321. Castro, C.; Stephens, R. Substitutions by ligands of low valent transition metals. A preparation of tolanes and heterocyclics from aryl iodides and cuprous acetylides. Journal of Organic Chemistry 1963, 28, 3313-3315.

322. Liang, B.; Dai, M.; Chen, J.; Yang, Z. Copper-free Sonogashira coupling reaction with PdCl2 in water under aerobic conditions. The Journal of Organic Chemistry 2005, 70, 391-393.

323. Park, K., Yun, YJ., Lee, SG. Efficient clevage of terminal acetonide group:Chirospecific synthesis of 2, 5-dideoxy-2, 5-imino-D-mannitol. Tetrahedran Letters 1994, 9737.

324. Hsieh, J.-C.; Cheng, C.-H. Nickel-catalyzed coupling of isocyanates with 1,3-iodoesters and halobenzenes: a novel method for the synthesis of imide and amide derivatives. Chemical Communications 2005, 4554-4556.

325. Mankske, R. H. F. Organic Synthesis 1932, XII, 10-11.

326. Greene, T. W., Wuts, P.G.M. Protective Groups in Organic Synthesis. John Wiley & Sons, Inc.: Brisbane, 1999, p 518–525.

283