SYNTHETIC STUDIES OF (+)-CHETOMIN AND (–) - Mountain ...

416
DISSERTATION EPIDITHIODIOXOPIPERAZINES: SYNTHETIC STUDIES OF (+)-CHETOMIN AND (–)-SPORIDESMIN A Submitted by Timothy R. Welch Department of Chemistry In partial fulfillment of the requirements For the Degree of Doctor of Philosophy Colorado State University Fort Collins, Colorado Fall 2012 Doctoral Committee: Advisor: Robert M. Williams Tomislav Rovis John L. Wood Chris Ackerson Douglas H. Thamm

Transcript of SYNTHETIC STUDIES OF (+)-CHETOMIN AND (–) - Mountain ...

DISSERTATION

EPIDITHIODIOXOPIPERAZINES: SYNTHETIC STUDIES OF

(+)-CHETOMIN AND (–)-SPORIDESMIN A

Submitted by

Timothy R. Welch

Department of Chemistry

In partial fulfillment of the requirements

For the Degree of Doctor of Philosophy

Colorado State University

Fort Collins, Colorado

Fall 2012

Doctoral Committee: Advisor: Robert M. Williams Tomislav Rovis John L. Wood Chris Ackerson Douglas H. Thamm

Copyright by Timothy Ryan Welch 2012

All Rights Reserved  

ii  

ABSTRACT

EPIDITHIODIOXOPIPERAZINES: SYNTHETIC STUDIES OF

(+)-CHETOMIN AND (–)-SPORIDESMIN A

This dissertation documents efforts toward the asymmetric total syntheses of the

natural products (+)-chetomin and (–)-sporidesmin A. Synthetic methods have been

developed to efficiently construct the dioxopiperazine core of both molecules.

Additionally, a simple epidithiodioxopiperazine has been synthesized to demonstrate a

general method for the addition of a sulfur bridge to a dioxopiperazine ring. The work

described herein, while not totally successful, provides a basis for future completion of

the asymmetric total syntheses of these two epidithiodioxopiperazines and other related

fungal metabolites.

iii  

ACKNOWLEDGEMENTS

I am truly thankful for all of the supportive individuals who have motivated,

inspired, and encouraged me throughout my education. This work and much of my

personal and professional development would not have been possible without you. I

would first like to thank my advisor, Professor Robert M. Williams, for accepting me into

the Williams group and providing a dynamic research environment. It has been an honor

to work for you, and I am grateful for all of the advice and insight you have provided me

over the years. Thanks to all members of the Williams group, past and present, for all of

the ideas, encouragement, and shenanigans. Karen Kahler and Elizabeth McCoy, thank

you for making my life at CSU a little easier. Special mention must go to all of my

climbing partners; thanks for catching my numerous falls, throwing entertaining

wobblers, helping me clear my head, and inspiring me to try harder.

Without question, I would not be here were it not for two professors at KU and

several teachers from high school. Professor Brian Blagg, thank you for introducing me

to the field of organic synthesis and for your continued support. Professor Givens, it was

your skillful course instruction that first inspired me to pursue a career in academia. In

high school, I was blessed to have Kristin Seaton (Gunn), John Wachholz, and Linda

Nelson as teachers and advisors; thank you all for your encouragement and for always

challenging me to do better.

Lastly, I extend my utmost gratitude to my family. My parents taught me the

value of hard work and persistence and have always encouraged me to strive for

excellence. It was their encouragement and support that nurtured my intellectual

curiosity. Mom, Dad, Eric, and Emily, thank you for all of your love and support.

iv  

TABLE OF CONTENTS

ABSTRACT ........................................................................................................................ ii

ACKNOWLEDGEMENTS ............................................................................................... iii

TABLE OF CONTENTS ................................................................................................... iv

LIST OF FIGURES .......................................................................................................... vii

LIST OF SCHEMES ....................................................................................................... xiv

Chapter 1: Epidithiodioxopiperazines

1.1: Introduction ................................................................................................1

1.2: Epidithiodioxopiperazines Derived from Phenylalanine or Tyrosine ...3

1.3: Tryptophan-Derived Epidithiodioxopiperazines ..................................13

1.4: Biosynthetic Investigations of Epidithiodioxopiperazines ...................23

1.5: Concluding Remarks ...............................................................................28

Chapter 2: Total Syntheses of Epidithiodioxopiperazines

2.1: Introduction ..............................................................................................29

2.2: Early Epidithiodioxopiperazine Syntheses (1973-1981) .......................30

2.3: Recent Epidithiodioxopiperazine Syntheses (2009-2012) .....................35

2.4: Other Relevant Syntheses: Formation of the C3-N1’ Bond .................45

2.5: Concluding Remarks ...............................................................................49

v  

Chapter 3: Studies Toward the Total Synthesis of Chetomin

3.1: Introduction ..............................................................................................51

3.2: Goals and Early Studies

3.2.1: Goals of the Project ..........................................................................51

3.2.2: Iminium Ion Approach .....................................................................52

3.2.3: Pinacol-type Rearrangement ............................................................53

3.3: Evolution of Coupling Strategy

3.3.1: Witkop’s Pyrroloindole ....................................................................55

3.3.2: Indole-Aniline Coupling ..................................................................57

3.3.3: Coupling to exo-3-bromopyrroloindoline ........................................59

3.3.4: Attempted Coupling to Tetracyclic Bromide ...................................60

3.4: Epidithiodioxopiperazine Formation .....................................................62

3.5: Attempted Core Construction

3.5.1: Problematic Peptide Couplings ........................................................63

3.6: Synthesis of Heptacyclic Core

3.6.1: Synthesis of Dioxopiperazines .........................................................65

3.6.2: Initial N-Methyl Amino Acid Incorporation ....................................66

3.6.3: Bypassing the Serine Side Chain with Sarcosine ............................68

3.6.4: Completion of the Carbon Skeleton of Chetomin ...........................69

3.6.5: Attempted Epidithiodioxopiperazine Formation .............................69

3.7: Proposed Future Studies

3.7.1: Proposed Synthesis of Tetraol .........................................................70

3.7.2: Bypassing the Tetraol ......................................................................72

vi  

3.7.3: Late-Stage Alkylation of Sarcosine Analogue .................................72

3.8: Concluding Remarks ...............................................................................73

Chapter 4: Synthetic Approach to Sporidesmin A

4.1: Introduction ..............................................................................................75

4.2: Retrosynthetic Analysis ...........................................................................76

4.3: Synthetic Approach to Sporidesmin A

4.3.1: Preparation of Dinitrostyrene Derivative .........................................78

4.3.2: Indole Synthesis and Sharpless Asymmetric Aminohydroxylation 80

4.3.3: Dioxopiperazine Formation .............................................................81

4.3.4: Oxidative Cyclization Attempts .......................................................83

4.3.5: Toward a Formal Synthesis of Sporidesmin A ................................86

4.4: Future Direction .......................................................................................88

4.5: Concluding Remarks ...............................................................................89

References .........................................................................................................................91

Chapter 5: Experimental Procedures ......................................................................104

Appendix I: Publications ............................................................................................346

Appendix II: Research Proposal ..................................................................................382

List of Abbreviations .....................................................................................................395

vii  

LIST OF FIGURES

CHAPTER 1

Figure 1.1 Epidithiodioxopiperazines derived from tyrosine and/or phenylalanine .....2

Figure 1.2 Tryptophan-derived epidithiodioxopiperazines ...........................................2

Figure 1.3 Gliotoxins .....................................................................................................3

Figure 1.4 Redox cycling of epidithiodioxopiperazines ...............................................4

Figure 1.5 Mixed disulfide formation ...........................................................................5

Figure 1.6 Simple biologically active epidithiodioxopiperazines .................................6

Figure 1.7 Hyalodendrins and related compounds ........................................................7

Figure 1.8 Silvatins .......................................................................................................7

Figure 1.9 Aranotins ......................................................................................................8

Figure 1.10 Emethallicins ................................................................................................9

Figure 1.11 Emestrins and related metabolites .............................................................11

Figure 1.12 Epicorazines ...............................................................................................11

Figure 1.13 Scabrosin esters ..........................................................................................12

Figure 1.14 Sirodesmins ................................................................................................13

Figure 1.15 Sporidesmins ..............................................................................................15

Figure 1.16 Metabolites of the fungus Chaetomium cochliodes ...................................16

Figure 1.17 Chetoseminudins ........................................................................................16

Figure 1.18 HIF-1 hypoxia response pathway ..............................................................17

Figure 1.19 Chaetocins and related metabolites ............................................................19

Figure 1.20 Fungal metabolites related to the chaetocins .............................................20

Figure 1.21 Verticillin A and related metabolites .........................................................20

Figure 1.22 Verticillin-type epipolythiodioxopiperazines ............................................21

Figure 1.23 Leptosins ....................................................................................................23

viii  

Figure 1.24 Putative epidithiodioxopiperazine gene clusters for sirodesmin PL (A) and

gliotoxin (B) ...............................................................................................26

CHAPTER 3

Figure 3.1 Chetomin and related fungal metabolites ..................................................52

CHAPTER 4

Figure 4.1 Sporidesmin A and related fungal metabolites ..........................................76

CHAPTER 5

Figure 5.1a 1H NMR spectrum of compound 3.5 .......................................................106

Figure 5.2a 1H NMR spectrum of compound 3.6 .......................................................108

Figure 5.2b 13C NMR spectrum of compound 3.6 ......................................................108

Figure 5.3a 1H NMR spectrum of compound 3.15 .....................................................110

Figure 5.4a 1H NMR spectrum of compound 3.12 .....................................................112

Figure 5.5a 1H NMR spectrum of compound 3.16 .....................................................114

Figure 5.6a 1H NMR spectrum of compound 3.17 .....................................................116

Figure 5.6b 13C NMR spectrum of compound 3.17 ....................................................116

Figure 5.7a 1H NMR spectrum of compound 3.21 .....................................................118

Figure 5.8a 1H NMR spectrum of compound 3.31 .....................................................120

Figure 5.9a 1H NMR spectrum of compound 3.32 .....................................................122

Figure 5.10a 1H NMR spectrum of compound 3.35 .....................................................124

Figure 5.11a 1H NMR spectrum of compound 3.40 .....................................................126

Figure 5.12a 1H NMR spectrum of compound 3.41 .....................................................128

Figure 5.13a 1H NMR spectrum of compound 3.42 .....................................................130

Figure 5.14a 1H NMR spectrum of (bromoethynyl)trimethysilane ..............................131

Figure 5.15a 1H NMR spectrum of compound 3.43 .....................................................133

Figure 5.16a 1H NMR spectrum of compound 3.44 .....................................................135

Figure 5.17a 1H NMR spectrum of compound 3.47 .....................................................137

ix  

Figure 5.18a 1H NMR spectrum of compound 3.48 .....................................................139

Figure 5.19a 1H NMR spectrum of compound 3.56 .....................................................141

Figure 5.20a 1H NMR spectrum of compound 3.57a ...................................................143

Figure 5.21a 1H NMR spectrum of compound 3.58a ...................................................145

Figure 5.22a 1H NMR spectrum of compound 3.57c ....................................................147

Figure 5.22b 13C NMR spectrum of compound 3.57c ..................................................147

Figure 5.23a 1H NMR spectrum of compound 3.57d ...................................................149

Figure 5.23b 13C NMR spectrum of compound 3.57d ..................................................149

Figure 5.24a 1H NMR spectrum of compound 3.58d ...................................................151

Figure 5.24b 13C NMR spectrum of compound 3.58d ..................................................151

Figure 5.25a 1H NMR spectrum of compound 3.57e ....................................................153

Figure 5.25b 13C NMR spectrum of compound 3.57e ..................................................153

Figure 5.26a 1H NMR spectrum of compound 3.58e ....................................................155

Figure 5.27a 1H NMR spectrum of compound 5.1 .......................................................157

Figure 5.28a 1H NMR spectrum of compound 5.2 .......................................................159

Figure 5.28b 13C NMR spectrum of compound 5.2 ......................................................159

Figure 5.29a 1H NMR spectrum of compound 5.3 .......................................................161

Figure 5.29b 13C NMR spectrum of compound 5.3 ......................................................161

Figure 5.30a 1H NMR spectrum of compound 3.56 .....................................................163

Figure 5.31a 1H NMR spectrum of compound 3.55e ....................................................165

Figure 5.32a 1H NMR spectrum of compound 3.60 .....................................................167

Figure 5.33a 1H NMR spectrum of compound 3.61 .....................................................169

Figure 5.34a 1H NMR spectrum of compound 3.63 .....................................................172

Figure 5.34b 13C NMR spectrum of compound 3.63 ....................................................172

Figure 5.35a 1H NMR spectrum of compound 3.59 .....................................................174

Figure 5.36a 1H NMR spectrum of compound 3.34 .....................................................176

Figure 5.37a 1H NMR spectrum of compound 3.64 .....................................................178

Figure 5.38a 1H NMR spectrum of compound 3.65 .....................................................180

Figure 5.38b 13C NMR spectrum of compound 3.65 ....................................................180

Figure 5.39a 1H NMR spectrum of compound 3.66 .....................................................182

Figure 5.39b 13C NMR spectrum of compound 3.66 ....................................................182

x  

Figure 5.40a 1H NMR spectrum of compound 3.67 .....................................................184

Figure 5.40b 13C NMR spectrum of compound 3.67 ....................................................184

Figure 5.41a 1H NMR spectrum of compound 3.68 .....................................................186

Figure 5.42a 1H NMR spectrum of compound 3.69 .....................................................188

Figure 5.43a 1H NMR spectrum of compound 5.4 .......................................................190

Figure 5.44a 1H NMR spectrum of compound 3.71 .....................................................192

Figure 5.45a 1H NMR spectrum of compound 5.5 .......................................................194

Figure 5.46a 1H NMR spectrum of compound 3.74 .....................................................196

Figure 5.46b 13C NMR spectrum of compound 3.74 ....................................................196

Figure 5.47a 1H NMR spectrum of compound 3.75 .....................................................198

Figure 5.47b 13C NMR spectrum of compound 3.75 ....................................................198

Figure 5.48a 1H NMR spectrum of compound 5.6 .......................................................200

Figure 5.48b 13C NMR spectrum of compound 5.6 ......................................................200

Figure 5.49a 1H NMR spectrum of compound 3.76 .....................................................203

Figure 5.49b 13C NMR spectrum of compound 3.76 ....................................................203

Figure 5.50a 1H NMR spectrum of compound 3.77 .....................................................205

Figure 5.51a 1H NMR spectrum of compound 5.7 .......................................................207

Figure 5.51b 13C NMR spectrum of compound 5.7 ......................................................207

Figure 5.52a 1H NMR spectrum of compound 3.80 .....................................................209

Figure 5.52b 13C NMR spectrum of compound 3.80 ....................................................209

Figure 5.53a 1H NMR spectrum of compound 3.81 .....................................................211

Figure 5.54a 1H NMR spectrum of compound 3.82 .....................................................214

Figure 5.55a 1H NMR spectrum of compound 5.9 .......................................................216

Figure 5.56a 1H NMR spectrum of compound 3.88 .....................................................218

Figure 5.56b 13C NMR spectrum of compound 3.88 ....................................................218

Figure 5.57a 1H NMR spectrum of compound 5.10 .....................................................220

Figure 5.58a 1H NMR spectrum of compound 3.90A ..................................................223

Figure 5.58b 13C NMR spectrum of compound 3.90A .................................................223

Figure 5.59a 1H NMR spectrum of compound 3.90B ...................................................224

Figure 5.59b 13C NMR spectrum of compound 3.90B ..................................................224

Figure 5.60a 1H NMR spectrum of compound 5.11 .....................................................226

xi  

Figure 5.60b 13C NMR spectrum of compound 5.11 ....................................................226

Figure 5.61a 1H NMR spectrum of compound 4.15 .....................................................228

Figure 5.61b 13C NMR spectrum of compound 4.15 ....................................................228

Figure 5.62a 1H NMR spectrum of compound 4.17 .....................................................230

Figure 5.62b 13C NMR spectrum of compound 4.17 ....................................................230

Figure 5.63a 1H NMR spectrum of compound 4.18 .....................................................232

Figure 5.63b 13C NMR spectrum of compound 4.18 ....................................................232

Figure 5.64a 1H NMR spectrum of compound 4.19 .....................................................233

Figure 5.65a 1H NMR spectrum of compound 4.7 .......................................................235

Figure 5.65b 13C NMR spectrum of compound 4.7 ......................................................235

Figure 5.66a 1H NMR spectrum of compound 4.6 .......................................................237

Figure 5.66b 13C NMR spectrum of compound 4.6 ......................................................237

Figure 5.67a 1H NMR spectrum of compound 5.12 .....................................................239

Figure 5.67b 13C NMR spectrum of compound 5.12 ....................................................239

Figure 5.68a 1H NMR spectrum of compound 4.20 .....................................................241

Figure 5.68b 13C NMR spectrum of compound 4.20 ....................................................241

Figure 5.69a 1H NMR spectrum of compound 4.5 .......................................................243

Figure 5.69b 13C NMR spectrum of compound 4.5 ......................................................243

Figure 5.70a 1H NMR spectrum of compound 4.4 .......................................................245

Figure 5.70b 13C NMR spectrum of compound 4.4 ......................................................245

Figure 5.71a 1H NMR spectrum of compound 4.23 .....................................................247

Figure 5.71b 13C NMR spectrum of compound 4.23 ....................................................247

Figure 5.72a 1H NMR spectrum of compound 4.24 .....................................................249

Figure 5.73a 1H NMR spectrum of compound 4.25 .....................................................251

Figure 5.74a 1H NMR spectrum of compound 4.26 .....................................................253

Figure 5.74b 13C NMR spectrum of compound 4.26 ....................................................253

Figure 5.75a 1H NMR spectrum of compound 4.27 .....................................................255

Figure 5.76a 1H NMR spectrum of compound 4.28 .....................................................257

Figure 5.77a 1H NMR spectrum of compound 4.29 .....................................................259

Figure 5.77b 13C NMR spectrum of compound 4.29 ....................................................259

Figure 5.78a 1H NMR spectrum of compound 4.30 .....................................................261

xii  

Figure 5.79a 1H NMR spectrum of compound 4.31 .....................................................263

Figure 5.80a 1H NMR spectrum of compound 4.32 .....................................................265

Figure 5.80b 13C NMR spectrum of compound 4.32 ....................................................265

Figure 5.81a 1H NMR spectrum of compound 4.3 .......................................................267

Figure 5.81b 13C NMR spectrum of compound 4.3 ......................................................267

Figure 5.82a 1H NMR spectrum of compound 4.40 .....................................................269

Figure 5.83a 1H NMR spectrum of compound 4.47 .....................................................271

Figure 5.84a 1H NMR spectrum of compound 4.50 .....................................................274

Figure 5.85a 1H NMR spectrum of compound 4.45 .....................................................276

Figure 5.86a 1H NMR spectrum of compound 4.46 .....................................................278

Figure 5.87a 1H NMR spectrum of compound 5.14 .....................................................281

Figure 5.88a 1H NMR spectrum of compound 5.16 .....................................................283

Figure 5.89a 1H NMR spectrum of compound 5.17 .....................................................285

Figure 5.90a 1H NMR spectrum of compound 5.18 .....................................................287

Figure 5.91a 1H NMR spectrum of compound 5.19 .....................................................289

Figure 5.92a 1H NMR spectrum of compound 5.20 .....................................................291

Figure 5.93a 1H NMR spectrum of compound 5.22 .....................................................294

Figure 5.94a 1H NMR spectrum of compound 5.23 .....................................................296

Figure 5.95a 1H NMR spectrum of compound 5.24 .....................................................298

Figure 5.96a 1H NMR spectrum of compound 5.25 .....................................................300

Figure 5.97a 1H NMR spectrum of compound 5.26 .....................................................302

Figure 5.98a 1H NMR spectrum of compound 5.27 .....................................................304

Figure 5.99a 1H NMR spectrum of compound 5.28 .....................................................306

Figure 5.100a 1H NMR spectrum of compound 5.29 .....................................................308

Figure 5.101a 1H NMR spectrum of compound 5.30 .....................................................310

Figure 5.102a 1H NMR spectrum of compound 5.31 .....................................................312

Figure 5.103a 1H NMR spectrum of compound 5.32 .....................................................314

Figure 5.104a 1H NMR spectrum of compound 5.33 .....................................................316

Figure 5.105a 1H NMR spectrum of compound 5.34 .....................................................318

Figure 5.106a 1H NMR spectrum of compound 5.35 .....................................................320

Figure 5.106b 13C NMR spectrum of compound 5.35 ....................................................320

xiii  

Figure 5.107a 1H NMR spectrum of compound 5.36 .....................................................322

Figure 5.108a 1H NMR spectrum of compound 5.37 .....................................................324

Figure 5.108b 13C NMR spectrum of compound 5.37 ....................................................324

Figure 5.109a 1H NMR spectrum of compound 5.38 .....................................................326

Figure 5.109b 13C NMR spectrum of compound 5.38 ....................................................326

Figure 5.110a 1H NMR spectrum of compound 5.39 .....................................................328

Figure 5.110b 13C NMR spectrum of compound 5.39 ....................................................328

Figure 5.111a 1H NMR spectrum of compound 5.40 .....................................................330

Figure 5.111b 13C NMR spectrum of compound 5.40 ....................................................330

Figure 5.112a 1H NMR spectrum of compound 5.41 .....................................................332

Figure 5.112b 13C NMR spectrum of compound 5.41 ....................................................332

Figure 5.113a 1H NMR spectrum of compound 5.42 .....................................................334

Figure 5.113b 13C NMR spectrum of compound 5.42 ....................................................334

Figure 5.114a 1H NMR spectrum of compound 5.43 .....................................................336

Figure 5.114b 13C NMR spectrum of compound 5.43 ....................................................336

Figure 5.115a 1H NMR spectrum of compound 5.44 .....................................................338

Figure 5.115b 13C NMR spectrum of compound 5.44 ....................................................338

Figure 5.116a 1H NMR spectrum of compound 5.45a ...................................................340

Figure 5.116b 13C NMR spectrum of compound 5.45a ..................................................340

Figure 5.117a 1H NMR spectrum of compound 5.45b ...................................................341

Figure 5.117b 13C NMR spectrum of compound 5.45b ..................................................341

Figure 5.118a 1H NMR spectrum of compound 5.46 .....................................................343

Figure 5.118b 13C NMR spectrum of compound 5.46 ....................................................343

Figure 5.119a 1H NMR spectrum of compound 1.12 .....................................................345

Figure 5.119b 13C NMR spectrum of compound 1.12 ....................................................345

xiv  

LIST OF SCHEMES

CHAPTER 1

Scheme 1.1 Proposed biosynthesis of gliotoxin ............................................................24

Scheme 1.2 Proposed oxepine ring formation ...............................................................24

Scheme 1.3 Proposed biosynthetic pathway of sirodesmin PL .....................................27

CHAPTER 2

Scheme 2.1 Total synthesis of (±)-sporidesmin A ........................................................31

Scheme 2.2 Synthesis of (±)-sporidesmin B .................................................................31

Scheme 2.3 Synthesis of (±)-dehydrogliotoxin .............................................................32

Scheme 2.4 Synthesis of (+)-gliotoxin ..........................................................................33

Scheme 2.5 Synthesis of (±)-hyalodendrin ....................................................................34

Scheme 2.6 Total synthesis of (±)-gliovictin and epi-gliovictin ...................................34

Scheme 2.7 Rastetter’s total synthesis of (±)-hyalodendrin ..........................................35

Scheme 2.8 Biomimetic total synthesis of (+)-11,11’-dideoxyverticillin A .................36

Scheme 2.9 Sodeoka’s total synthesis of (+)-chaetocin ................................................37

Scheme 2.10 Movassaghi’s total synthesis of (+)-chaetocin ...........................................38

Scheme 2.11 Total syntheses of chaetocin C and 12,12’-dideoxychetracin A ................39

Scheme 2.12 Synthesis of (+)-gliocladine C ...................................................................41

Scheme 2.13 Total synthesis of (+)-gliocladin B ............................................................42

Scheme 2.14 Synthesis of (+)-12-deoxybionectin A and (+)-gliocladin B .....................42

Scheme 2.15 Total synthesis of epicoccin G ...................................................................43

Scheme 2.16 Key pyrrolidine synthesis ..........................................................................44

Scheme 2.17 Completion of the total synthesis of (–)-acetylaranotin (1.35) ..................45

Scheme 2.18 Synthesis of (±)-psychotrimine via novel C3-N1’ bond formation ...........46

xv  

Scheme 2.19 Total synthesis of kapakahine B ................................................................47

Scheme 2.20 Rainier’s synthetic C3-N1’ bond forming method ....................................47

Scheme 2.21 Total synthesis of kapakahine F .................................................................48

Scheme 2.22 Concise total synthesis of (+)-pestalazine B ..............................................49

CHAPTER 3

Scheme 3.1 Proposed model study for C3-N1’ bond formation ...................................52

Scheme 3.2 Attempted iminium ion formation .............................................................53

Scheme 3.3 Retrosynthesis of key intermediate 3.8 through a pinacol-type

rearrangement ............................................................................................54

Scheme 3.4 Synthesis of 2-alkynyl aniline derivative 3.17 ...........................................54

Scheme 3.5 Synthesis of 2,3-dihydrotryptophan alkyne derivative ..............................55

Scheme 3.6 Witkop’s pyrroloindole inspiration ............................................................56

Scheme 3.7 Retrosynthetic plan using pyrroloindoline .................................................56

Scheme 3.8 Attempted synthesis of 3-bromopyrroloindole ..........................................57

Scheme 3.9 Formation of key C-3 quaternary center ....................................................57

Scheme 3.10 Attempted formation of functionalized C3-N bond ...................................58

Scheme 3.11 Key alkyne synthesis ..................................................................................58

Scheme 3.12 Larock indole synthesis of tryptophan dimer .............................................59

Scheme 3.13 Coupling of 3-bromopyrroloindoline .........................................................59

Scheme 3.14 Retrosynthetic analysis of (+)-chetomin ....................................................60

Scheme 3.15 Coupling with more advanced bromopyrroloindoline ...............................61

Scheme 3.16 Optimized peptide coupling and dioxopiperazine formation .....................61

Scheme 3.17 Microwave assisted dioxopiperazine synthesis .........................................62

Scheme 3.18 Attempted coupling with indole .................................................................62

Scheme 3.19 Formation of the disulfide bridge ..............................................................63

Scheme 3.20 Coupling to 3-bromopyrroloindoline and attempted dioxopiperazine

formation ....................................................................................................64

Scheme 3.21 Attempted synthesis of alternate dioxopiperazine precursor .....................65

Scheme 3.22 Synthesis of dioxopiperazine 3.77 .............................................................66

xvi  

Scheme 3.23 Synthesis of (N-Me)3 dioxopiperazine .......................................................67

Scheme 3.24 Attempted protection and deprotection of 3.82 .........................................68

Scheme 3.25 Attempted peptide coupling with N-Me,Boc-Ser(OH) ..............................68

Scheme 3.26 Synthesis of sarcosine-derived dioxopiperazine ........................................69

Scheme 3.27 Synthesis of (N-Me)3 dioxopiperazine .......................................................69

Scheme 3.28 Attempted epidithiodioxopiperazine formations .......................................70

Scheme 3.29 Key tetraols and iminium intermediates ....................................................71

Scheme 3.30 Possible oxidations ....................................................................................72

Scheme 3.31 Alternative sulfenylation ............................................................................72

Scheme 3.32 Proposed conversion of 3.91 to chetomin ..................................................73

CHAPTER 4

Scheme 4.1 Retrosynthetic analysis of (–)-sporidesmin A ............................................77

Scheme 4.2 Nitration of vanillin ....................................................................................78

Scheme 4.3 Dimethoxyindole synthesis ........................................................................79

Scheme 4.4 Synthesis of 5-chloro-dinitrostyrene derivative .........................................79

Scheme 4.5 Undesired acetal formation ........................................................................80

Scheme 4.6 Asymmetric aminohydroxylation ..............................................................80

Scheme 4.7 Preparation of (DHQD)2AQN ....................................................................81

Scheme 4.8 Formation of the dioxopiperazine and an undesired elimination ...............81

Scheme 4.9 Preparation of dipeptide .............................................................................82

Scheme 4.10 Cyclization to the dioxopiperazine ............................................................82

Scheme 4.11 Improved synthesis of dioxopiperazine 4.3 ...............................................83

Scheme 4.12 First attempts at oxidative cyclization .......................................................84

Scheme 4.13 Model cyclization study .............................................................................84

Scheme 4.14 More oxidative cyclization attempts ..........................................................85

Scheme 4.15 Progression of cyclization ..........................................................................86

Scheme 4.16 Attempted cyclization ................................................................................86

Scheme 4.17 Kishi’s final steps in the sporidesmin A synthesis .....................................87

Scheme 4.18 Preparation of N-Me,Fmoc-Ala-OH ..........................................................87

xvii  

Scheme 4.19 Incorporation of N-Me-Ala ........................................................................88

Scheme 4.20 Full panel of oxidation conditions to screen ..............................................89

Scheme 4.21 Proposed epidithiodioxopiperazine formation ...........................................89

1

CHAPTER 1

Epidithiodioxopiperazines

1.1: Introduction

Nearly twenty distinct families of epidithiodioxopiperazine fungal metabolites

have been characterized since the seminal discovery of gliotoxin in 1936. This unique

class of natural products is characterized by a sulfur-bridged dioxopiperazine (1.1), a

feature generally requisite for the potent biological activity prevalent among the class.1-5

All natural epidithiodioxopiperazines discovered to date contain at least one

aromatic amino acid. Representative molecules from each family to be discussed are

shown in Figure 1.1 (tyrosine- and/or phenylalanine-derived) and Figure 1.2

(tryptophan-derived). In this chapter, we present an overview of the structures of

naturally occurring epidithiodioxopiperazines, relevant physiological properties, and

some of the more interesting of the proposed fungal biogeneses.

NN

O

O

R4

R2

R1

R3

SS

1.1

2

Figure 1.1. Epidithiodioxopiperazines derived from tyrosine and/or

phenylalanine.

Figure 1.2. Tryptophan-derived epidithiodioxopiperazines.

gliotoxin (1.2) hyalodendrin (1.3) dithiosilvatin (1.4)

NNMe

O

O

OH

OHH

SS MeN

NMe

O

O

OHSS

MeNNMe

O

OH

SS

O

NN

O

O

SS

O

O

OHH

HAcO

aranotin (1.5)

NN

O

O

SS

O

O

OH

HO

emethallicin A (1.6)

O Bn

O PhHO

NNMe

O

O

SSO

OH

emestrin (1.7)

HO

OH

OOH

O

OMe

NN

O

O

SS

epicorazine A (1.8)

HO

O

O

OH

H

H

H

H NN

O

O

SS

scabrosin ester I (1.9)

H

HO

OAc

O

AcO

sirodesmin A (1.10)

NNMe

O

O

OHSS

HO

OAcO

Me

Me MeO

H

sporidesmin A (1.11)

NNMe

O

OMe

SS

NMe

HOCl

MeOOMe

H

HO

chetomin (1.12)

NNMe

O

O

SS

NH

HOH

MeNNMe

O

O

SS

NOH

NNMe

O

O

SS

NH

HOH

NMeN

O

O

SS

HNH

HO

chaetocin (1.13)

NNMe

O

OMe

SS

NH

H

NMeN

O

OMe

SS

HNH

verticillin A (1.14)

OHHO

NNMe

O

O

SS

NH

H

NMeN

O

O

SS

HNH

leptosin (1.15)

OHHO

Me

Me

HO

3

1.2: Epidithiodioxopiperazines Derived from Phenylalanine or Tyrosine

In 1936, a novel substance with substantial antifungal and antiviral activity was

isolated from the wood fungus Gliocladium fimbriatum by Weindling and Emerson.6 A

putative structure (1.16, Figure 1.3) was proposed for the metabolite based on

degradation studies. This structure, however, could not account for some experimental

observations, leading Johnson and Woodward to propose a revised structure for gliotoxin

(1.2) in 1958.7 Absolute stereochemistry was later determined by x-ray analysis.8

Gliotoxin and related metabolites (Figure 1.3) have since been isolated from a variety of

fungi—including several Penicillium and Aspergillus species, Gliocladium,

Thermoascus, and Candida—and have been the focus of numerous synthetic and

biosynthetic studies that have formed the basis for much of the research discussed

herein.2,3

Figure 1.3. Gliotoxins.

The initial interest in the chemotherapeutic potential of gliotoxin as an antifungal

or antiviral agent waned as in vivo studies revealed gliotoxin to be generally cytotoxic.9

Moreover, gliotoxin has been implicated as a virulence factor of Aspergillus fumigatus,

NNMeO

OH

O

SS

OH1.16 dehydrogliotoxin (1.18)

N NMe

O

O OH

SS

RO H

gliotoxin (1.2, R=H)gliotoxin acetate (1.17, R=Ac)

N NMe

O

O OH

SS

HO

bisdethiodi(methylthio)gliotoxin (1.19)

N NMe

O

O OHOH H

MeS

SMe

bisdethiodi(methylthio)dehydrogliotoxin (1.20)

N NMe

O

O OHOH

MeS

SMe

N NMe

O

O OH

S3

HO H

gliotoxin E (1.21)

N NMe

O

O OH

S4

HO H

gliotoxin G (1.22)

4

the main source of invasive aspergillosis and leading cause of death in

immunocompromised patients.10 However, interest in the molecule was renewed when it

was discovered that gliotoxin displayed selective toxicity to cells of the hematopoietic

system.11,12 Specifically, gliotoxin exhibits antiproliferative activity against T and B cells

and inhibits phagocytic activity with considerable selectivity toward immune system

cells, leading to promising studies that have demonstrated that gliotoxin prevents graft-

versus-host disease after bone marrow transplantation.13

Generally, the toxicity of gliotoxin and other epidithiodioxopiperazines can be

attributed to two mechanisms: generation of reactive oxygen species (Figure 1.4) and

mixed disulfide formation (Figure 1.5). In the presence of a suitable reducing agent such

as glutathione or dithiothreitol, epidithiodioxopiperazines are reduced to the

corresponding dithiols (1.23). Autooxidation back to the disulfide (1.1) occurs with the

production of superoxide ions and hydrogen peroxide, known to cause oxidative damage

to cells.14,15 Gliotoxin has specifically been shown to induce single- and double-stranded

DNA damage in the presence of glutathione, presumably by hydroxyl radicals generated

in this redox process.16

Figure 1.4. Redox cycling of epidithiodioxopiperazines.

Evidence suggests that gliotoxin and other epidithiodioxopiperazines are also

capable of forming mixed disulfides with free thiol groups in cells.17,18 Following

NN

O

O

R4

R2

R1

R3

SS

1.1

NN

O

O

R4

R2

1.23

R1HS

R3

SH

2O22O2

cellular reductant

electron transfer

H2O2

5

incubation with radiolabelled gliotoxin, cells were shown to contain protein-bound [35S]-

gliotoxin (1.24).3 This result could be reversed if cells were treated with both gliotoxin

and excess reducing agent (dithiothreitol), suggesting a covalent interaction. Moreover,

the antiviral activity of gliotoxin is lost in the presence of excess dithiothreitol. This

evidence supports a mechanism of toxicity resulting from mixed disulfide formation

between gliotoxin and protein.19

Figure 1.5. Mixed disulfide formation.

Toxicity of epidithiodioxopiperazines would seem to rely on an intact disulfide

ring or the reduced dithiol. Indeed, dethiogliotoxin lacks the antibacterial activity of

gliotoxin. Reduction and methylation of the disulfide bridge

(bisdethiodi(methylthio)gliotoxin, 1.19) also results in a loss of antiviral activity.4,20 Not

surprisingly, the simple disulfide 1.25 and dithiol 1.26 exhibit potent biological activity,

highlighting the importance of the fragment to the observed toxicity of

epidithiodioxopiperazines.21 The great structural diversity in epidithiodioxopiperazines

may have evolved only to mask the core disulfide moiety to prevent degradation by target

organisms, rather than to impart any sort of selectivity or increased toxicity.2

N NMe

O

O OHOH H

S

SH

S

HS

N NMe

O

O OH

SS

OH H+

[35S]-gliotoxin

1.24

6

Figure 1.6. Simple biologically active epidithiodioxopiperazines.

Like gliotoxin, the hyalodendrins (Figure 1.7) are derived naturally from

phenylalanine and serine. Hyalodendrin (1.3) was originally isolated by Strunz in 1974

from Hyalodendron sp.22 The same fungus was later shown to produce the

bis(methylthio) derivative (1.29)23 and epitetrasulfide 1.28.24 Epitrithiohyalodendrin

(1.27) has only been observed as a product of the unidentified fungus NRRL 3888, along

with 1.3 and 1.29.25 Not surprisingly, hyalodendrin and the epitri- and epitetrasulfide

derivatives exhibit antibacterial activity, while the bisdethiodi(methylthio) analogue is

inactive against fungi and bacteria and relatively non-toxic to mice.22-25 Interestingly, it

was observed that hyalodendrin could be converted to epitetrasulfide 1.28 in the presence

of HCl with heating in methanol and the culture medium. Racemic tetrasulfide was

isolated when HCl was omitted from the same conditions.

Enantiomers of the hyalodendrins (except for epitrisulfide 1.27) have been

isolated from both terrestrial and marine sources. Gliovictin (1.32) was first isolated from

Helminthosporium victoriae in 1974,26 the same year that researchers at Eli Lilly reported

the isolation of the same structure (named A26771E) along with the disulfide (A26771A,

1.30) and epitetrasulfide (A26771C, 1.31) from Penicillium turbatum.27 Fenical has also

isolated gliovictin from the marine deuteromycete Asteromyces cruciatus.28 Predictably,

1.30 and 1.31 both showed antiviral and antibacterial activity, while gliovictin–lacking

sulfur atoms capable of redox cycling or mixed disulfide formation–was inactive.27

MeNNMe

O

O

H

H

SS

1.25

MeNNMe

O

O1.26

HHS

HSH

7

Figure 1.7. Hyalodendrins and related compounds.

Kawahara and coworkers reported the isolation of dithiosilvatin (1.4) and

silvathione (1.33) in 1987 from Apergillus silvaticus (Figure 1.8).29

Dioxopiperazinethiones such as 1.33 are rare and are possible intermediates in the

formation of trioxopiperazines from epidithiodioxopiperazines. The authors reported the

conversion of 1.4 to the bisdethiodi(methylthio) derivative (1.34) by reductive

methylation (NaBH4, MeI), a compound previously isolated by Hanson and O’Leary

from Gliocladium deliquescens.30

Figure 1.8. Silvatins.

The next subgroup of epidithiodioxopiperazines to be discussed is characterized

by the presence of at least one seven-membered dihydrooxepine ring, and includes the

MeNNMe

O

OssMeN

NMeO

O

sssss

MeNNMe

MeN

O

OS

NMeO

O

MeS

SMe

S

MeNNMe

O

OS MeN

NMeO

O

MeS

SMeMeN

NMeO

O

S

gliovictin (A26771E) (1.32)

A26771A (1.30)

hyalodendrin (1.3) bisdethiodi(methylthio)-hyalodendrin (1.29)

A26771C (1.31)

hyalodendrin-S4 (1.28)hyalodendrin-S3 (1.27)

OH

OH OH OH

OHOH

ssss

OH

MeNNMe

O

O

O

H

SSMeN

NMe

O

O

O

S

MeNNMe

O

O

OMeS

bisdethiodi(methylthio)silvatin (1.34)

silvathione (1.33)dithiosilvatin (1.4)

SMe

8

aranotins (Figure 1.9), emethallicins (Figure 1.10), and emestrins (Figure 1.11).

Aranotin (1.5) and acetylaranotin (1.35) have been isolated from Arachniotus aureus and

Aspergillus terreus and exhibit antiviral activity that is apparently selective for RNA

viruses such as the polio, Coxsackie (A21), rhino-, and parainfluenza viruses through

inhibition of viral RNA synthesis.31-37 Structurally, the aranotins are related to gliotoxin,

with the similarities most evident in apoaranotin (1.37). Apoaranotin can be considered a

chimera of gliotoxin and aranotin, containing the cyclohexadienol of gliotoxin and the

dihydrooxepine ring of aranotin. Asteroxepin (1.39) was the first monooxepine derivative

to be isolated and is further unique in that it contains one unsubstituted amide. This

dioxopiperazine may provide evidence for the sequence of steps in the biosynthesis of the

more complex aranotins.

Figure 1.9. Aranotins.

Closely related to the aranotins are the emethallicins (Figure 1.10). Both families

share the same absolute stereochemistry, and emethallicin A (1.6) differs apoaranotin

(1.37) only in the ester moieties. This observation was experimentally confirmed by

hydrolysis of 1.6 followed by acetylation to afford compound 1.37, identical to naturally

NN

O

O

O

OOH

SS N

NO

O

O

OOAc

SS

NN O

O

O

SS

NN

O

O

O

O

MeS

SMe

NN O

O

O

MeS

SMeN

NHO

O

OOAcH

MeS

SMe

asteroxepin (1.39)bisdethiodi(methythio)acetylapoaranotin (1.38)

bisdethiodi(methylthio)acetylaranotin (1.36)

apoaranotin (1.37)

acetylaranotin(1.35)aranotin (1.5)

AcOH

AcOH

HH

HAcO

HOAc

HOH

AcOH

AcOH

HOAc

Ph

9

occurring apoaranotin.38 Emethallicins B, D, and F (1.40, 1.42, and 1.44) share this same

monooxepine core and differ apoaranotin only in ester substitution and sulfur content of

the epipolythiodioxopiperazine ring. Emethallicin C (1.41) is symmetrical and the only

emethallicin to contain two dihydrooxepine rings, more closely resembling aranotin and

acetylaranotin.

Figure 1.10. Emethallicins.

Emethallicin A (1.6) was first isolated from Emericella heterothallica in 1989 by

Kawai and coworkers, who later reported the isolation of emethallicins B-F (1.40-1.44)

from the same fungus.38-41 Interestingly, Kawai was unable to convert synthetic

emethallicin D monoacetate (1.42, R=Ac) to the naturally occurring metabolite (1.42,

R=H). Basic hydrolytic conditions only succeeded at forming the disulfide and

N

N

O

O

OOO

O OH

H

HO

S

N

NS

O

OO

OO O

HH

SSN

N

O

OOO

O

OOH

HS

sSN

N

O

O

OOO

O O

HO

s

N

N

O

O

OOO

OO

OHH

HO

s

s

N

N

O

OOO

OO O

HH

RO

s

s

OH

s ssss

emethallicin F (1.44)emethallicin E (1.43)

emethallicin D (1.42)R= H, Ac

emethallicin C (1.41)

emethallicin B (1.40)emethallicin A (1.6)

H

H

10

tetrasulfide monoacetates of 1.6 and 1.40, respectively.39 This disproportionation of the

trisulfide is similar to a result obtained by Waring and coworkers, who similarly

converted trisulfide gliotoxin E (1.21) to the disulfide, gliotoxin (1.2), and the

tetrasulfide, gliotoxin G (1.22).42

All of the emethallicins exhibit fairly strong inhibitory activities upon histamine

release from mast cells, with IC50 values ranging from 1.0 x 10-6 to 2.0 x 10-8 M.

Generally, activity is stronger for the original emethallicins than for the acetate

derivatives. Micromolar inhibition of 5-lipoxygenase has also been reported.43

The final class of known epipolythiodioxopiperazine metabolites known to

contain at least one dihydrooxepine ring is the macrocyclic emestrins (Figure 1.11).

Emestrin (1.7) was isolated in 1985 from the fungus Emericella striata, and later from E.

quadrilineata, E. foveolata, E. acristata, and E. parvathecia.44-46 Trisulfide emestrin B

(1.45), piperazinethione aurantioemestrin (1.47), and trioxopiperazine dethiosecoemestrin

(1.48) were later isolated from E. striata.46-49 It has been postulated that the latter two

compounds are derived biosynthetically from emestrin. Emestrin displays potent

antifungal and antibacterial activity, but is also very toxic to mammals.

Recently, Kanda and coworkers reported the isolation of MPC1001 (1.46) and its

analogues (not shown) from Cladorrhinum sp. KY4922, contributing eight new members

to the emestrin family of natural products.50 MPC1001 contains a methoxy group rather

than the free phenol found in emestrin, but is otherwise structurally and stereochemically

identical. MPC1001 and its epipolysulfide analogues all showed antiproliferative activity

in the DU145 human prostate cancer cell line.50

11

Figure 1.11. Emestrins and related metabolites.

Epicorazines (Figure 1.12) have been isolated from several organisms, including

Epicoccum nigrum (epicorazine A and B, 1.8 and 1.49),51-53 E. purpurascens (epicorazine

B),54 and Stereum hirsutum (epicorazine C, 1.50).55 The only difference between 1.8 and

1.49 is the absolute stereochemistry at C6. This cis configuration is shared between 1.49

and 1.50.

Figure 1.12. Epicorazines.

Epicorazine A, B, and C display only marginal activity against Gram-positive

bacteria, including methicillin-resistant Staphylococcus aureus (MRSA) and

vancomycin-resistant Enterococcus faecalis (VRE), and are inactive against Gram-

N

NMe

O

O

O

O

OO

MeO

HO

S S

OH

H

OH

NNMe

O

O

O

OO

O

MeO

HO

N

CHO

N

NMe

O

O

O

O

OO

MeO

HO

s

OH

H

OH

s s

S

H NMe

O

O

O

OO

O

MeO

HO

CHO

O

H

emestrin (1.7)

aurantioemestrin (1.47) desthiosecoemestrin (1.48)

emestrin B (1.45)

N

NMe

O

O

O

O

OO

MeO

MeO

S SH

OH

MPC1001 (1.46)

OH

NNO

OH H

H

S S

H

NNO

OH H

H

S S

H

epicorazine A (1.8) epicorazine B (1.49)

OO

HO HO

O

OH OH

O

NNO

OH H

H

S S

H

epicorazine C (1.50)

O

HO OH

O

OH

NNO

OS S

3822-A (1.51)

O

HO OH

O

12

negative organisms. All three exhibit antiproliferative effects against L929 mouse

fibroblast cells and K562 human leukemia cells, as well as cytotoxicity toward the HeLa

human cervical carcinoma cell line.55

The scabrosin esters were originally isolated from the lichen Xanthoparmelia

scabrosa in 1978, but it was not until 1999 that the correct structures were determined

(Figure 1.13).56,57 The isolation of these compounds was of particular interest, as this

report marked the first epidithiodioxopiperazine to be isolated from lichenized fungi.

Submicromolar activity, similar to that of gliotoxin (1.2), was observed for the scabrosins

against the murine P815 mastocytomia cell line, as well as low nanomolar activity in

MCF7 human breast carcinoma cell line. Scabrosin esters have also been shown to induce

apoptosis concomitantly with a large increase in mitochondrial membrane potential and

significant decrease in total cellular ATP. Mitochondrial ATP synthase is the proposed

cellular target of the scabrosins.58

Figure 1.13. Scabrosin esters.

The sirodesmins (Figure 1.14) were first discovered in 1977 as metabolites of

Sirodesmium diversum59 and later from the unrelated fungus Phoma lingam.60

Sirodesmins A-H (1.10, 1.56-1.60) are characterized by a spirofused tetrahydrofuran

cyclopentylpyrrolidine skeleton. Sirodesmins A-C are epimers of G and H at the

spirocenter. Notably, sirodesmin H was the first example of a naturally occurring

monosulfide.

NN

O

O

SS

scabrosin ester 1 (R1=CH3, R2=CH3, 1.9)scabrosin ester 2 (R1=CH3, R2=C3H7, 1.52)scabrosin ester 3 (R1=C3H7, R2=C3H7, 1.53)scabrosin ester 4 (R1=CH3, R2=C5H11, 1.54)scabrosin ester 5 (R1=C3H7, R2=C5H11, 1.55)H

HO

O

O

O

OR1

OR2

13

Figure 1.14. Sirodesmins.

Sirodesmins are potent antiviral agents, particularly against the rhinovirus.59

Sirodesmin G (originally named sirodesmin PL, 1.58) has specifically been shown to

exhibit activity against Gram-positive bacteria,61 although it has also been implicated as

the causative agent of blackleg disease in canola crops (along with phomalirazine, 1.61).

Metabolites 1.58 and 1.61 have both been isolated from the ascomycetous fungus

Leptosphaeria maculans, the organism known to be responsible for blackleg disease.62

1.3: Tryptophan–Derived Epidithiodioxopiperazines

The remaining epidithiodioxopiperazine alkaloids to be discussed are all derived

from tryptophan (Figure 1.2). Of this subset, the sporidesmins (Figure 1.15) are the most

densely functionalized and the only members to contain a substituted aromatic ring.

NNMe

O

H

OHAcO

Me

Me

MeO

O

OS

OH

NNMe

O

H

OHAcO

Me

Me

MeO

O

OS

OH

NNH

O

OOH

H

MeMeMe

S

O

OS

OH

s

SN

NMe

O

H

OHHO

Me

Me

MeO

O

OS

OH

S

NNMe

O

H

OHAcO

Me

Me

MeO

O

O

S

OH

S

s sNNMe

O

H

OHAcO

Me

Me

MeO

O

O

OH

sirodesmin H (1.60)

desacetyl-sirodesmin PL (1.59)sirodesmin G (sirodesmin PL, 1.58)

sirodesmin C (1.57)sirodesmin A (1.10)

phomalirazine (1.61)

sss

sN

NMe

O

H

OHAcO

Me

Me

MeO

O

O

OHsirodesmin B (1.56)

14

Sporidesmin was discovered by researchers investigating the source of the disease facial

eczema that plagued sheep in New Zealand and Australia. The disease caused extensive

liver damage in infected sheep and ultimately resulted in death. Thornton and Percival

eventually established that ingestion of pasture grasses on which the fungus Pithomyces

chartarum (previously known as Sporidesmium bakeri) was growing was the cause of the

serious disease.63,64 Sporidesmin (1.11) was isolated and implicated as the main toxic

agent produced by P. chartarum.65 The structure and absolute configuration were

subsequently determined by crystallographic means.8,66,67

As an interesting aside, veterinarians discovered that zinc sulfate doses gave

sheep protection from the effects of sporidesmin.68 Transition metals such as zinc are

now known to inhibit generation of the superoxide anion radical, with

epidithiodioxopiperazines shown to form a 2:1 complex with zinc ion.69,70

Extensive amounts of research have focused on the sporidesmins, producing the

complete characterization of all nine derivatives (1.11, 1.62-1.69). All contain a densely

functionalized, tryptophan–derived pyrroloindoline core coupled to an alanine residue.

Sporidesmin C (1.63)71 is the most unusual, containing a novel trisulfide [4.3.3] ring

system.

A great deal of chemistry applicable to most of the epipolythiodioxopiperazines

was discovered through investigations of the sporidesmins. For example, the trisulfide

sporidesmin E (1.65) is readily converted to the disulfide (1.11) upon treatment with

triphenyl phosphine. Alternatively, di- and trisulfides can be converted to tetrasulfides,

achieved using hydrogen polysulfide or dihydrogen disulfide. This was demonstrated by

the conversion of sporidesmins A (1.11) and E (1.65) into sporidesmin G (1.67).

15

Figure 1.15. Sporidesmins.

In 1944, Waksman and Bugie reported the isolation of a new antibiotic metabolite

of the fungus Chaetomium cochliodes that they named chetomin.72 It was not until 30

years later that Walter and coworkers determined the structure of chetomin (1.12),

revealing a nearly dimeric core likely formed from two molecules each of tryptophan and

serine.73 The two fragments are joined by a bond between the β-pyrrolidinoindoline

carbon and the indole nitrogen, a common feature of all five chaetocins (Figure 1.16).74

Chetomin is the only molecule in the family to contain a disulfide bridge within both

dioxopiperazine rings. Dethio-tetra(methylthio)chetomin (1.70) and chaetocochin C

(1.73) differ only in the oxidation state of the sulfurs, while chaetocochins A (1.71) and B

(1.72) are macrocyclic analogues of 1.70 and 1.73, each containing a novel 14-membered

ring.

NNMe

N

O

OMeMeO

MeO

ClHO

H

SS

OH

Me NNMe

N

O

OMeMeO

MeO

ClHO

H

SS

Me NNMe

N

O

OMeMeO

MeO

ClHO

H

SS

OH

S

NNMe

N

O

OMeMeO

MeO

ClHO

H

MeS

SMe

HO

Me NNMe

N

O

OMeMeO

MeO

ClHO

H

s

OH

Me NNMe

N

O

OMeMeO

MeO

ClHO

H

MeSHO

NNMe

N

O

OMeMeO

MeO

ClHO

H

sOH

Me NNMe

N

O

OMeMeO

MeO

Cl

H

SS NMe

NMe

N

O

OHMeO

MeO

HO

H

SS

MeCl

HO

ss

s ss

sporidesmin G (1.67)

sporidesmin E (1.65)

sporidesmin J (1.69)sporidesmin H (1.68)

sporidesmin F (1.66)sporidesmin D (1.64)

sporidesmin C (1.63)sporidesmin B (1.62)sporidesmin A (1.11)

16

Figure 1.16. Metabolites of the fungus Chaetomium cochliodes.

Chetomin was recently isolated from Chaetomium seminudum by Fujimoto and

coworkers in 2004, along with three new metabolites named the chetoseminudins (Figure

1.17).75 Chetoseminudin A (1.74) is merely the pentasulfide homolog of chetomin. From

a biosynthetic viewpoint, the more interesting discoveries are chetoseminudins B-D

(1.75-1.77), monomeric bisdethiodi(methylthio) structures that potentially provide insight

as to the biosynthetic sequence that produces chetomin, the chaetocochins, and other

related epipolythiodioxopiperazines derived from tryptophan and serine.

Figure 1.17. Chetoseminudins.

Chetomin (1.12) has an unprecedented mechanism of action as a cancer

chemotherapeutic agent. Solid tumors must adapt to oxygen deprivation through

NH

N

NNMeS

S

O

O

OH

NMeMeN

O

HO

O

SS

chetomin (1.12)

NH

NNMe

O

O

dethio-tetra(methylthio)chetomin (1.70)

SMe

MeS

NH

NNMe

O

O

chaetocochin C (1.73)

SS

NN

NMe

O

O

SS

chaetocochin B (1.72)

N

N

NNMe

O

OSMe

MeS

O

chaetocochin A (1.71)

H

NNMeMeN

O

MeSO

SMe

H

NMeMeN

O

MeSO

SMe

N

O

NMeMeN

O

MeSO

SMe

NNMeMeN

O

MeSO

SMe

OH

HO

OHOH

HO

OH

NH

N

NNMeS

S

O

O

OH

NMeMeN

O

HO

O

SS

chetoseminudin A (1.74)

H

S NH

HNNMe

O

O

MeS

SMeOH NH

HNNH

O

O

MeS

SMeOR

chetoseminudin B (1.75) chetoseminudin C (R=H, 1.76)chetoseminudin D (R=Ac, 1.77)

17

induction of the heterodimeric transcription factor hypoxia-inducible factor 1 (HIF-1) in

order to survive. Overexpression of HIF-1 is associated with radioresistance in tumors,

increased risk of metastasis, and a poor prognosis for patients.76-78 In normal cells, the !-

subunit of HIF-1 (HIF-1!) is hydroxylated and degraded by vHL proteosome (Figure

1.18). As oxygen levels decrease and become the rate-limiting reagent in the

hydroxylation reaction, HIF-1! accumulates and binds to transcriptional coactivators

p300 and CREB binding protein (CBP). Consequent to this binding is the transcription of

proteins requisite to the survival of hypoxic cancer cells, facilitating tumor growth and

progression.79

Figure 1.18. HIF-1 hypoxia response pathway.

Chetomin has been shown to inhibit the interaction between HIF-1 and p300 both

in vitro and in cells, despite extensive surface interactions between the two proteins.

Specifically, Kung and coworkers have shown that chetomin disrupts the tertiary

structure of p300, inhibiting the transcriptional activity of HIF-1.76 No other small

molecule has been identified to mediate an antitumor response through this mechanism of

action. More recently, Hilton and coworkers proposed that chetomin and other

HIF-1!FIH-1

O2

low [O2]

HIF-1! HIF-1!

HIF-1! HIF-1!

HIF-1! HIF-1!

HIF-1!

HO

OH

OH

vHLProteosome

HIF-1!p300/CBP

transcriptiontumorgrowth

chetomin

p300/CBP

chetomin

tumordeath

18

epidithiodioxopiperazines bind zinc at the CH1 domain of p300, ultimately resulting in

ejection of a stable zinc–epidithiodioxopiperazine complex. Loss of zinc from the CH1

domain causes the previously observed disruption of the p300 tertiary structure.1,80

Chaetocin A (1.13, Figure 1.19) is a dimeric epidithiodioxopiperazine also

derived from two molecules each of tryptophan and serine. It was isolated from

Chaetomium minutum in 1970.81,82 Fifteen years passed before the penta- and hexasulfide

homologs chaetocin B and C (1.78 and 1.79) were isolated from Chaetomium spp., along

with the novel tetrasulfide chetracin A (1.80).83 In 2012, several related metabolites were

isolated from Oidiodendron truncatum, including the tetra-, penta- and hexasulfide

homologs melinacidin IV, chetracins B and C (1.82, 1.83, and 1.84), and the

dethiotetra(methylthio) derivative chetracin D (1.81).84

The three chaetocin metabolites (and likely the chetracins) can be interconverted

through either desulfurization of 1.78 and 1.79 with triphenyl phosphine to generate

chaetocin (1.13), or by sulfurization of chaetocin with phosphorus pentasulfide in carbon

disulfide to afford a mixture of chaetocins B and C.83

Recently, chaetocin A was identified as the first known inhibitor of lysine-specific

histone methyltransferases.85 Histone methylation is an important process in controlling

gene expression patterns, especially during cellular differentiation and embryonic

development. The activity of histone methyltransferases is disregulated in some tumors,

making chaetocin an attractive tool for the study of the molecular mechanism of histone

methylation.85 Additionally, melinacidin IV and chetracin B display nanomolar (3 – 54

nM) activity against five human cancer cell lines (HCT-8, Bel-7402, BGC-823, A549,

and A2780).84

19

Figure 1.19. Chaetocins and related metabolites.

Several metabolites related to the chaetocins have recently been reported,

possessing a C3-C3’ linkage to indole rather than the additional monomer found in the

chaetocins (Figure 1.20). T988 A, B, and C (1.86-1.88) were originally isolated from

Tilachlidium sp., although recently it was shown that Oidiodendron truncatum also

produces the same metabolites, in addition to oidioperazine A (1.89) and the chetracins

(1.83-1.85).75,86 Chetoseminudin C (1.76) was also isolated from O. truncatum,

suggesting that it may be a common intermediate to all of the tryptophan- and serine-

derived epipolythiodioxopiperazines discussed thus far. T988 A and B are cytotoxic to

P388 leukemia cells.86

NNMe

O

O

SS

NH

HOH

NMeN

O

O

SS

HNH

HO

chaetocin A (1.13)

NNMe

O

O

SS

NH

HOH

NMeN

O

O

SS

HNH

HO

chaetocin B (1.78)

S

NNMe

O

ONH

HOH

NMeN

O

O

SS

HNH

HO

chaetocin C (1.79)

S

SSS

NNMe

O

ONH

HOH

NMeN

O

O HNH

HO

chetracin A (1.80)

ss s

s

ss s

s

NNMe

O

ONH

HOH

NMeN

O

O HNH

HO

12,12'-dideoxychetracin A (1.81)

ss s

s

ss s

sOH

HO

melinacidin IV (x=y=2, 1.82)chetracin B (x=2, y=3, 1.83)chetracin C (x=3, y=3, 1.84)

NNMe

O

ONH

HOH

NMeN

O

O HNH

HO Sx

OHHO

Sy

chetracin D (1.85)

NNMe

O

ONH

HOH

NMeN

O

O HNH

HO

OHHO

MeS

SMe

MeS

SMe

20

Figure 1.20. Fungal metabolites related to the chaetocins.

Verticillin A (1.14, Figure 1.21) is quite similar to chaetocin A (1.13), derived

from two molecules of alanine rather than serine. Only verticillins A, D and E (1.14, 1.92

and 1.93) are symmetrical, while the two tryptophan residues of the remaining verticillins

are coupled to different amino acids on the two halves (either alanine, serine, or tyrosine).

Verticillin B (1.90), for instance, contains alanine and serine residues on the northern and

southern halves of the molecule, respectively. Verticillins A-C were isolated from

Verticillium sp., while the remaining compounds in Figure 1.21 are produced by

Gliocladium sp.87-89

Figure 1.21. Verticillin A and related metabolites.

Gliocladines A-E (Figure 1.22, 1.97-1.101) were isolated in 2005 from

Gliocladium roseum, along with verticillin A, Sch52900, and Sch52901 (1.14, 1.95, and

1.96).90 Gliocaldines A and B are simply the penta- and hexasulfide homologs of

verticillin A, thus it is not surprising that the same organism produces all three

compounds. The structures of gliocladines C and D (1.99 and 1.100) should also look

NNMe

O

ONH

HOH

HN

T988 A (1.86)

SSS

OH

NNMe

O

ONH

HOH

HN

T988 B (1.87)

OH

NNMe

O

ONH

HOH

HN

T988 C (1.88)

SS

OHMeS

SMeN

NMe

O

ONCHO

HOH

HN

oidioperazine A (1.89)

OHMeS

SMe

NNMe

O

OMe

SS

NH

H

NMeN

O

OMe

SS

HNH

verticillin A (1.14)

OHHO

NNMe

O

OR2

SS

NH

H

NMeN

O

OR1

SS

HNH verticillin B (R1=CH3, R2=CH2OH, 1.90)

verticillin C (R1=CH3, R2=CH2OH, S5 homolog 1.91)verticillin D (R1=CH(OH)CH3, R2=CH(OH)CH3, 1.92)verticillin E (R1=COCH3, R2=COCH3, 1.93)verticillin F (R1=COCH3, R2=CH(OH)CH3, 1.94)Sch52900 (R1=CH(OH)CH3, R2=CH3, 1.95)Sch52901 (R1=CH2CH3, R2=CH3, 1.96)

OHHO

21

familiar, as they are the alanine derivatives of T988 A and C (Figure 1.20). Bionectra

byssicola F120 has also been shown to produce bionectins A and B (1.102, 1.103), the

glycine and tyrosine derivatives of T988C.91

Figure 1.22. Verticillin-type epipolythiodioxopiperazines.

The dimeric subset of epipolythiodioxopiperazines increased greatly in number

with the discovery of the leptosins (Figure 1.23) from a strain of Leptosphaeria sp.

attached to the marine alga Sargassum tortile.92-94 Leptosins A-K (1.104-1.118) all

contain at least one valine residue, a feature unique among all

epipolythiodioxopiperazines to this family. Leptosins A-C, G, H, and K (1.104-1.113)

share the 12,12’-dihydroxylated, octacyclic core of the verticillins, gliocladines, and

chetracins. The C3-C3’ linkage to indole is once again produced in leptosins D-F (1.114-

1.116), the valine derivatives of the T988s, gliocladines C-E, and the bionectins. In 1994,

two remarkable additions to this family were discovered, the epimers leptosin I and J

(1.117 and 1.118).94 These two compounds are characterized by a C12-C11’ ether

linkage, introducing an additional ring to the structure that prohibits the possibility of

topoisomers. Leptosins are generally toxic to P388 leukemia cells.

NNMe

O

OMe

SS

NH

H

NMeN

O

OMe

Sx

HNH

gliocladine A (x=3, 1.97)gliocladine B (x=4, 1.98)verticillin A (x=2, 1.14)

OHHO

NNMe

O

OMe

NH

H

HN

gliocladine C (x=2, 1.99)gliocladine D (x=3, 1.100)gliocladine E (x=4, 1.101)

Sx

OH

NNMe

O

OR

NH

H

HN

bionectin A (R=H, 1.102)bionectin B (R=CH(OH)CH3, 1.103)

SS

OH

22

Figure 1.23. Leptosins.

In summary, over 100 epipolythiodioxopiperazine alkaloids have been isolated

and characterized to date. While this was intended to be a comprehensive review of all

known members of this class of natural products, some were undoubtedly and

inadvertently omitted. One cannot help but marvel at the remarkable biological activity of

the class as a whole, although it is unlikely that any epipolythiodioxopiperazine will ever

achieve therapeutic utility in the clinic due to the general cytotoxicity inherent in the

disulfide through mechanisms discussed above. Certainly others have and will continue

to argue otherwise, but it is our opinion that the true contribution to medicine will be

realized by employing epipolythiodioxopiperazines as tools for the study of novel

biological pathways. To date, the natural products chetomin (1.12), gliotoxin (1.2), and

chaetocin A (1.13) have already contributed to our understanding of the respective

biological targets of the molecules.

NNMe

O

O

Sy

NH

H

NMeN

O

O

Sx

HNH

leptosin A (x=4, y=2, 1.104)leptosin B (x=3, y=2, 1.105)leptosin C (x=2, y=2, 1.106)leptosin G (x=4, y=3, 1.107)leptosin G1 (x=3, y=3, 1.108)leptosin G2 (x=2, y=3, 1.109)leptosin H (x=2, y=4, 1.110)

OHHO

HO

Me

Me

NNMe

O

O

Sx

NH

H

NMeN

O

O

S

HNH

leptosin K (x=2, 1.111)leptosin K1 (x=3, 1.112)leptosin K2 (x=4, 1.113)

OHHO

Me

Me

Me

Me

S

N

NMeO

O

Sx

NH H

N NMe

O

OHN

H

leptosin I (R1=CH2OH, R2=H, 1.117)leptosin J (R1=H, R2=CH2OH, 1.118)

OOH

Me

Me

R2R1

NNMe

O

ONH

H

HN

leptosin D (x=2, 1.114)leptosin E (x=3, 1.115)leptosin F (x=4, 1.116)

Sx

OH

Me

Me

23

Many of the families (particularly those containing a C3-C3’ indoline linkage)

share a great deal of structural similarity beyond the epipolythiodioxopiperazine moiety

that defines the class. It is likely that hundreds of additional variants exist in nature and

have yet to be discovered. While numerous biosynthetic studies have been conducted, we

still have only a limited understanding of the ease with which nature is able to assemble

epipolythiodioxopiperazines, molecules that have for over fifty years taunted synthetic

chemists. In the last section of this chapter, we present the notable biosynthetic

discoveries reported to date.

1.4: Biosynthetic Investigations of Epidithiodioxopiperazines

Researchers have investigated the biosynthesis of gliotoxin (1.2) for nearly forty-

five years, but still very little experimental evidence exists to support the numerous

proposals. In 1958 and 1960, Suhadolnik reported the only undisputed incorporation

study, demonstrating that Trichoderma viride incorporates isotopically labeled

phenylalanine (1.119) and serine (1.120) into gliotoxin (Scheme 1.1).95,96 Walsh recently

implicated the nonribosomal peptide synthetase GliP in the catalysis of the peptide

coupling and dioxopiperazine cyclization reactions.97 Some debate has occurred as to

whether cyclo-Phe-Ser (1.121) is a biosynthetic intermediate to gliotoxin. Although

doubly labeled 1.121 is incorporated into gliotoxin by T. viride, the fungus Penicillium

terlikowskii poorly incorporates the same compound.98-100 Walsh observed that release of

the dioxopiperazine from the enzyme is slow, allowing for some speculation that further

transformations may occur to the enzyme bound compound prior to release.

24

Scheme 1.1. Proposed biosynthesis of gliotoxin.

Arene oxide 1.122 was proposed as a biosynthetic intermediate to both the

gliotoxins and aranotins by Neuss in 1968. Cyclization at the ortho position provides the

tricyclic core of gliotoxin (1.123), whereas a ring enlarging tautomerization of the arene

oxide could give the oxepine ring (1.129) characteristic of the aranotins (Scheme

1.2).33,34

Scheme 1.2. Proposed oxepine ring formation.

gliotoxin (1.2)

NNMe

O

O

OH

OHH

SS

1.123

NNH

O

O

OH

OHH

1.124

NNH

O

O

OH

OHH OH

HOGliC GliG

GSH

1.125

NNH

O

O

OH

OHH

G S

S G

NNH

O

O

OH

OHH S

NH

HN

O

CO2HO

HO2C

NH2

S

HNN

H

O

HO2CO

CO2H

NH2

NNH

O

O

OH

OHH SH

HS

1.128

NNH

O

O

OH

OHH

SS

1.126

1.127

GliT

O2 H2O2

CO2H

NH2OHHO2C

NH2

phenylalanine (1.119) serine (1.120)

+HN

NH

O

O

OHGliP GliC/GliF

O2, NADPH

HNNH

O

O

OH

1.122

O

1.121

GliJ/GliI

GliN

HNNH

O

O

OH

1.122

OHN

NH

O

O

OH

1.129

OHN

NH

O

O

OH

1.130

O

O

[O]

25

The incorporation of sulfur into epidithiodioxopiperazines was until recently

poorly understood. Sulfur transfer readily occurs among various potential donors,

including methionine, cysteine, and sodium sulfate, complicating [35S] feeding

studies.2,101 In 2011, two groups independently provided evidence supporting a GliC–

mediated bishydroxylation of a dioxopiperazine (i.e. 1.123 to 1.124).102,103 Elimination of

water to generate diiminium 1.125 could be followed by nucleophilic attack of the

cysteine thiolate residues of two glutathione (GSH) molecules, catalyzed by the

specialized glutathione S-transferase GliG.102-104 Sequential GliJ peptidase and GliI

thioesterase activity is proposed to reveal the free dithiol.102 Scharf and coworkers have

unequivocally shown that GliT catalyzes the oxidation to the disulfide (i.e. 1.128),

leaving N-methylation to complete the biosynthesis of gliotoxin.105 The order of tailoring

events is up for debate, but recent advances in genomics and proteomics tools to study

and manipulate secondary metabolite production will certainly produce much more

accurate insight as to the formation of gliotoxin in nature.

Recall that the toxicity of epidithiodioxopiperazines generally arises from redox

cycling or mixed disulfide formation (Figures 1.4 and 1.5). How is it then that producing

fungi, such as Aspergillus fumigatus, are immune to toxicity inherent in the structure of

gliotoxin? Several studies published in the last two years have provided convincing

evidence that GliT is responsible for the self-resistance of A. fumigatus to gliotoxin.105,106

Deletion of the gliT gene renders A. fumigatus mutants highly sensitive to gliotoxin,

toxicity that can be reversed by addition of glutathione. Moreover, concentrations of

reduced gliotoxin increase with the concomitant depletion of intracellular glutathione

26

levels. It is likely that alterations of the cellular redox status and mixed disulfide

formation contributes to the growth inhibition observed in the absence of gliT.

Perhaps the cytotoxicity of gliotoxin is an indirect consequence of the true role of

gliotoxin and other epidithiodioxopiperazines produced by fungi. The same redox cycling

that produces deleterious consequences in naïve organisms may, in Aspergillus

fumigatus, serve as a buffer against cellular oxidative stress.106 Gliotoxin and other

epidithiodioxopiperazines may have evolved as simple antioxidants, with the host

protected from unwanted redox cycling and mixed disulfide formation by GliT.

Sirodesmin PL (1.58) is the only other epidithiodioxopiperazine to have been

extensively studied from a biosynthetic perspective. Putative biosynthetic gene clusters

for sirodesmin and gliotoxin identified from Leptosphaeria maculans and Aspergillus

fumigatus, respectively, are given in Figure 1.24.107-109 Based on recent advances to our

understanding of gliotoxin biosynthesis and the identification of homologs in the

sirodesmin gene cluster, we are able to predict many of the experimentally unverified

steps in the biosynthesis of sirodesmin PL (Scheme 1.3).

Figure 1.24. Putative epidithiodioxopiperazine gene clusters for sirodemsin PL

(A) and gliotoxin (B).109

Feeding studies have shown that Leptosphaeria maculans incorporate labeled

tyrosine and serine into both sirodesmin PL (1.58) and presumed intermediate

phomamide (1.132).59,110-112 We propose the incorporation of sulfur to proceed

27

analogously to the gliotoxin biosynthesis, through dihydroxylation (1.133), imine

formation, and glutathione addition (1.134). Unmasking of the dithiol (1.135) and

oxidation to the disulfide (1.136) could be followed by oxidation and Claisen

rearrangement to give 1.137, which after cyclization and oxidation provides known L.

maculans metabolite phomalirazine (1.61).59,111 Oxidative spirorearrangement, N-

methylation, and ketone reduction would lead to another known metabolite, desacetyl-

sirodemin PL (1.59). Acetylation by SirH would complete the biosynthesis of sirodesmin

PL (1.58).2,65,107,109

Scheme 1.3. Proposed biosynthetic pathway of sirodesmin PL.

CO2H

NH2OHHO2C

NH2

tyrosine (1.131) serine (1.120)

+HN

NH

O

O

OH

SirD/SirP SirC

phomamide (1.132)

HO O

MeMe

HNNH

O

O

OH

1.133

O

MeMe HO

OHHN

NH

O

O

OH

1.134 (G=glutathione)

O

MeMe GS

SG

SirJ/SirISirGGSH HN

NH

O

O

OH

1.135

O

MeMe HS

SH

SirTHN

NH

O

O

OH

1.136

O

MeMe

SS HN

NH

O

O

OH

1.137

SSO

Me

MeMe

O[O]

ClaisenRearrangement

[O]

NNH

O

O

OH

phomalirazine (1.61)

SS

[O], N-Me (SirN), reduction

OMe

MeMe

OHO

H

NNMe

OH

HOHO

Me

Me

MeO

O

O

SOHS

desacetyl-sirodesmin PL (1.59)

SirH NNMe

OH

OHAcO

Me

Me

MeO

O

O

SOHS

sirodesmin G (sirodesmin PL, 1.58)

28

1.5: Concluding Remarks

Epidithiodioxopiperazine alkaloids possess an astonishing array of molecular

architecture that has for decades challenged and inspired synthetic chemists. Biosynthetic

studies presented in this chapter provide a glimpse at the efficiency and elegance with

which Nature is able to assemble these compounds. Synthetic chemists strive to mimic

and in turn better understand the mechanisms by which microorganisms are able to

produce such complexity, hoping to channel some of Nature’s efficiency into novel

synthetic pathways. In the next chapter, we present many of the synthetic advances made

toward several of the epidithiodioxopiperazines presented above.

29

CHAPTER 2

Total Syntheses of Epidithiodioxopiperazines

2.1: Introduction

Over 100 naturally occurring epidithiodioxopiperazines have been isolated.

However, relatively few have succumbed to total synthesis despite decades of effort,

highlighting the challenging synthetic nature of the class of molecules. Initial interest in

the biological activity of epidithiodioxopiperazines sparked the synthetic interest of

several groups, with six different naturally occurring members of the class yielding to

synthetic chemists between 1973 and 1981. Three decades passed before renewed interest

in epidithiodioxopiperazines arose, sparked by isolation reports of new metabolites,

exciting results from the biological community detailing novel mechanisms of action in

cells, and advances in genomics that invigorated interest in elucidating the biosynthesis

of these secondary metabolites. Between 2009 and 2012 alone, syntheses of eight

additional epidithiodioxopiperazine alkaloids have been reported. A casual glance

through Chapter 1 is enough to impress upon the reader the great degree of structural

similarity shared among and between families of epidithiodioxopiperazines. We

anticipate that in the coming decade total syntheses of entire families of this class of

fungal metabolites will be completed concomitantly with great advances in synthetic

methods and biosynthetic understanding. In this chapter, we present a comprehensive

review of total syntheses of epidithiodioxopiperazines.

30

2.2: Early Epidithiodioxopiperazine Syntheses (1973-1981)

The total synthesis of sporidesmin A was completed by Kishi and coworkers in

1973. In a series of communications Kishi described a novel strategy for the synthesis of

epidithiodioxopiperazines using a dithioacetal moiety as a protecting group for the

disulfide bridge.113-115 Thus protected, the dithioacetal is stable to acidic, basic, and

reducing conditions, allowing for the introduction of thiol groups at an early stage in a

total synthesis. Synthesis of the sporidesmins began with the treatment of

dioxopiperazine 2.1 with the dithiane derivative of p-anisaldehyde in the presence of acid

to afford dithioacetal-protected dioxopiperazine 2.2 (Scheme 2.1).113 Condensation with

acid chloride 2.3 and subsequent methoxymethyl deprotection gave compound 2.4.

Treatment of ketone 2.4 with DIBAL-H at -78 °C resulted in stereoselective reduction to

the alcohol, which was then converted into acetate 2.5 in 80% yield. Cyclization to the

diacetate (2.6) proceeded upon addition of iodosobenzene diacetate, and hydrolysis of the

acetates gave the corresponding diol. Treatment of the diol with m-chloroperbenzoic acid

(mCPBA) afforded an intermediate sulfoxide, which decomposed to the disulfide upon

exposure to strong Lewis acid, revealing (±)-sporidesmin A (1.11).

Intermediate 2.5 was also used by Kishi in a total synthesis of (±)-sporidesmin B

(Scheme 2.2).116 Reduction of the acetate gave the methylene (2.7), which underwent an

oxidative cyclization similar to that reported in the sporidesmin A synthesis to benzoate

2.8. The disulfide was revealed as described above, completing the synthesis of (±)-

sporidesmin B (1.62).

31

Scheme 2.1. Total synthesis of (±)-sporidesmin A.

Scheme 2.2. Synthesis of (±)-sporidesmin B.

Fukuyama and Kishi reported the first synthesis of dehydrogliotoxin (1.18,

Scheme 2.3) in 1973.114 Benzoic acid derivative 2.10 was coupled to dioxopiperazine 2.9

and treated with diazomethane to give methyl ester 2.11. Radical bromination with NBS

and benzoyl peroxide was followed by treatment with potassium thioacetate to give

dithioacetate 2.12. Cleavage of the thioacetates and addition of p-methoxybenzaldehyde

NNMe

O

O

MeO

AcS

p-MeOC6H4CH2S2H2,H+

NMe

COClCl

MeOOMe

+

50%

1. n-BuLi2. HCl, THF3. NaOH

NMe

Cl

MeOOMe

O

HNNMe

O

OMe

SS Ar

NNMe

O

OMe

SS ArMeO

80%

1. DIBAL-H2. Ac2O

NMe

Cl

MeOOMe

HNNMe

O

OMe

SS Ar

AcO

30%

iodosobenzenediacetate

NMe

Cl

MeOOMe

N

25%

1. NaOH, MeOH2. mCPBA

H

AcOAcO

3. BF3!Et2O

(±)-sporidesmin A (1.11)

2.1 2.2 2.3

2.4 2.5

2.6

NMe

O

OMe

SS Ar

NMe

Cl

MeOOMe

NH

HOHO

NMe

O

OMe

SS

NMe

Cl

MeOOMe

HNNMe

O

OMe

SS Ar

AcO

90%

NaCNBH3

2.5

AcOHNMe

Cl

MeOOMe

HNNMe

O

OMe

SS Ar

2.7

NMe

Cl

MeOOMe

NH

NMe

O

OMe

PhOCO

20%

(PhCO2)2, DME

4,4'-thiobis-(6-t-butyl-3-methylphenol)

SS Ar

26%

1. KOH, MeOH, THF2. mCPBA

3. BF3!Et2O NNMe

N

O

OMeMeO

MeO

ClHO

H

SS

Me

(±)-sporidesmin B (1.62)2.8

32

and boron trifluoride etherate gave a 1:1 diasteromeric mixture of thioacetals 2.13 in

good yield. This masked disulfide is stable to a variety of conditions that the disulfide

would not otherwise survive, allowing Kishi to introduce sulfur at an early stage in the

synthesis.

Scheme 2.3. Synthesis of (±)-dehydrogliotoxin.

A three-step conversion to the primary chloride (2.14) was followed by addition

of phenyllithium to affect cyclization on the desired bridgehead carbon in 79% yield

(2.15). Addition of phenyllithium with benzyl chloromethyl ether efficiently installed the

benzyl protected serine side chain (2.16). Cleavage of the benzyl and methyl ethers and

conversion to the epidithiodioxopiperazine gave (±)-dehydrogliotoxin (1.18).

The biosynthesis of gliotoxin (1.2) is believed to proceed through the

intramolecular nucleophilic ring-opening of a phenylalanine-derived arene oxide (1.122)

and has been the subject of considerable speculation and interest. Kishi and coworkers

drew inspiration from this biogenetic hypothesis in devising a brilliant total synthesis of

gliotoxin. The total synthesis of (±)-gliotoxin was completed in 1976 utilizing the same

HNNMe

O

O

I

OMe

O OH 1. CuI, K2CO3 PhNO2, 170 °C2. CH2N2

35%

NNMe

O

O

MeO2C

OMe

+

NBS, (PhCO2)2, CCl4;

AcSK, CH2Cl2 NNMe

O

O

MeO2C

OMe

AcS

SAc

2. p-MeO-PhCHO, BF3!OEt2, CH2Cl2

72%

NNMe

O

O

MeO2C

OMe

S SAr

1. HCl, MeOH, 50 °C

61%N

NMe

O

OOMe

S SAr1. MeOH, NaBH4

2. MsCl, Et3NCl

65%

3. LiCl, DMF THF, -78 °C79%

PhLi

2.9 2.10 2.11 2.12

2.13 2.14

NNMe

O

O

S SAr

THF, -78 °C;BOMCl

71%

PhLi

2.15OMe

NNMe

O

O

S SAr

48%

1. BCl3, CH2Cl2, 0 °C2. mCPBA, CH2Cl2

2.16OBn

OBn

(±)-dehydrogliotoxin (1.18)

NNMe

O

O

OHSS

OH

3. BF3!Et2O

33

disulfide protecting strategy as employed above for the sporidesmins, and was re-

engineered in 1981 by the same route starting from optically pure dithioacetal 2.17

obtained from resolution (Scheme 2.4).117,118 Coupling of 2.17 with t-butoxy arene oxide

2.18 in the presence of triton B afforded 2.19 and 2.20 in a 2 :1 ratio. Acetylation,

deprotection, mixed anhydride formation, and reduction gave alcohol 2.21 in 77% yield

from 2.19. Primary alcohol 2.21 was converted to the chloride following mesylation, and

the secondary ester deprotected to reveal alcohol 2.22. The key stereoselective

cyclization-alkylation reaction was achieved upon addition of phenyllithium to 2.22 and

benzoxymethyl chloride, affording cycloadduct 2.23 in modest yield (53%). The primary

alcohol was revealed upon removal of the benzyl group, and the thioacetal oxidatively

removed to afford either (±)- or (+)-gliotoxin (1.2).

Scheme 2.4. Synthesis of (+)-gliotoxin.

Strunz and Kakushima followed the precedent set by Kishi and completed a total

synthesis of hyalodendrin (1.3, Scheme 2.5).119 Known thioacetal 2.24 was alkylated

HNN

O Triton B

O MeSS Ar

O

O

OtBu+N

N

O

O MeSS Ar

O

OtBu

HON

N

O

O MeSS Ar

O

OtBu

HO+

1. Ac2O2. TFA

3. ClCO2Et4. NaBH4

77%

98%

NN

O

O MeSS Ar

OH

AcO1. MsCl2. LiCl3. NaOMe

90%N

N

O

O MeSS Ar

Cl

HOPhLi, BOMCl

53%N

N

O

O MeSS ArHO H

OBn

1. BCl32. mCPBA

35%

NN

O

O MeSSHO H

OH

3. HClO4

2.17 2.182.19 2.20

2.19:2.20 = 2:1

2.21 2.22 2.23

(+)-gliotoxin (1.2)

34

sequentially with chloromethyl methyl ether and benzyl bromide, then deprotected to

reveal hyalodendrin (1.3).

Scheme 2.5. Synthesis of (±)-hyalodendrin.

In 1979 and 1980, Williams and Rastetter reported the total synthesis of gliovictin

(1.32, Scheme 2.6).120,121 Sarcosine anhydride (2.25) was converted in three steps to silyl

enol ether 2.27. Alkylation with benzyl bromide and concomitant sulfenylation and

deprotection gave methyl sulfide 2.29. Additional sulfenylation gave a 3:1 mixture of

diastereomers (2.30), favoring the undesired anti isomer. Reduction of the mixture of

aldehydes completed the total synthesis of gliovictin (1.32) and epi-gliovictin (2.31).

Scheme 2.6. Total synthesis of (±)-gliovictin and epi-gliovictin.

A synthesis of hyalodendrin (1.3, Scheme 2.7) was achieved from silyl enol ether

2.28, used previously in the gliovictin synthesis.120 Addition of monoclinic sulfur to the

MeNNMe

O

O

S SAr

2.24

1. n-BuLi; MOMCl2. n-BuLi; BnBr3. mCPBA4. HClO45. BCl3, -70 °C

5% (±)-hyalodendrin (1.3)

MeNNMe

O

O

OHSS

MeNNMe

O

O

EtOCHO, NaOMeMeN

NMe

O

O

1. KOtBu

O

H

H

MeNNMe

O

O

LDA, -78 °C;2. TBDPSCl

H

OTBDPS BnBr

MeNNMe

O

O

LDA, -78 °C;

H

OTBDPS MeSSMe, -78 °C MeNNMe

O

O

1. Et3N

O

H 2. MeSCl, -100 °C

MeS

H

MeNNMe

O

O

LiAl(t-BuO)3HMeS

SMeCHO

95%80% (3 steps)

87%

MeNNMe

O

OMeS

SMeOH

MeNNMe

O

OMeS

SMeOH

(±)-epi-gliovictin (2.31)40%

(±)-gliovictin (1.32)16%

2.25 2.26 2.27

2.28 2.29

2.30, anti:syn = 3:1

+

35

enolate of 2.28 followed by reductive workup provided thiol 2.32. Conversion to the

enolic methyl disulfide, deprotection of the silyl group, and sulfenylation with

triphenylmethyl chlorodisulfide gave a mixture of diastereomers, unfortunately favoring

the undesired anti isomer (2.34). Reduction and oxidation of 2.34 gave hyalodendrin (1.3)

in 29% yield.

Scheme 2.7. Rastetter’s total synthesis of (±)-hyalodendrin.

2.3: Recent Epidithiodioxopiperazine Syntheses (2009-2012)

Several total syntheses of epidithiodioxopiperazines have been reported in the last

three years. Movassaghi and coworkers were the first to publish in this modern synthetic

revival, reporting the synthesis of (+)-11,11’-dideoxyverticillin A (2.43, Scheme 2.8).122

The authors showed that the amine resulting from cleavage of the N-Boc carbamate of

2.35 was readily cyclized to dioxopiperazine 2.36 in morpholine. Exposure of 2.36 to

bromine produced 3-bromopyrroloindoline 2.37, and the amides were subsequently

methylated upon treatment with iodomethane. Addition of tris(triphenylphosphine)cobalt

chloride gave the desired dimeric intermediate 2.38. After extensive investigation

searching for appropriate stereoselective hydroxylation conditions, the dimer was

eventually oxidized with bis(pyridine)silver(I) permanganate to a fragile octacyclic

MeNNMe

O

O 1. LDA, -78 °C; !-sulfur2. HCl

H

OTBDPS 3. NaBH4

1. MeSCl, Et3N, -78 °C2. HCl, H2O

Ph3CSSCl, Et3N

>99%

2.28 2.32

2.33

MeNNMe

O

O

H

OTBDPS

HS

MeNNMe

O

O

O

H

S

H

MeS

MeNNMe

O

OS

SCHO

2.34, anti:syn = 2:1

MeS

SCPh3

-78 °C1. NaBH4

2. KI3

(±)-hyalodendrin (1.3)

MeNNMe

O

O

OHSS

51%

29%

36

tetraol (2.39). Exposure of 2.39 to Fu’s (R)-(+)-4-pyrrolidinopyridinyl(pentamethyl-

cyclopentadienyl)iron (PPY) catalyst with t-butyldimethylsilyl chloride (TBSCl) gave

selectively the alanine-derived protected hemiaminals (2.40). Removal of the

benzenesulfonyl groups with sodium amalgam revealed diaminodiol 2.41. Treatment of

2.41 with K2CS3 followed by ethanolamine gave diaminotetrathiol 2.42, which readily

oxidized to (+)-11,11’-dideoxyverticillin A (2.43) when partitioned between aqueous

hydrochloric acid and dichloromethane and treated with potassium triiodide.

Scheme 2.8. Biomimetic total synthesis of (+)-11,11’-dideoxyverticillin A.

NHBocO

HN

Me

CO2Me

PhSO2N

1. TFA

84%PhSO2N

59%

HNNH

O

OMe

1. Br2

NSO2Ph

NNMeBr

H

O

OMe2. MeI

46%

CoCl(PPh3)3

NSO2Ph

NNMe

H

O

OMe

SO2PhN

NMeN

H

O

OMe

63%

Py2AgMnO4

NSO2Ph

NNMe

H

O

O Me

SO2PhN

NMeN

H

O

OMeHO

OHHO

OH55%

TBSCl

NSO2Ph

NNMe

H

O

O Me

SO2PhN

NMeN

H

O

OMeTBSO

OHHO

OTBS

PPY

87%

Na(Hg)

NH

NNMe

H

O

O Me

HN

NMeN

H

O

OMeTBSO

OHHO

OTBS 56%

1. K2CS3, TFA

2. ethanolamine

NH

NNMe

H

O

O Me

HN

NMeN

H

O

OMeHS

SHHS

SH 87%

KI3pyridine

NH

NNMe

H

O

O

HN

NMeN

H

O

OMe

Me

SS

SS

(+)-11,11'-dideoxyverticillin A (2.43)

2.35 2.36 2.37

2.38 2.39

2.40 2.41

2.42

2. morpholine

37

Chaetocin (1.13) is quite similar in structure to 11,11’-dideoxyverticillin A,

derived from two molecules of serine rather than alanine. Substituted dioxopiperazine

2.46 was prepared in five steps from N-Me,Cbz-D-Ser (2.44) and D-Trp-OMe

(2.45).123,124 Bromocyclization gave tetracycle 2.47, converted to the tribromide (2.48)

under radical conditions and hemiaminal 2.49 following addition of water. Movassaghi’s

reductive dimerization conditions using a Co(I) complex afforded the desired dimer

(2.50) in modest yield. Addition of 2.50 to condensed hydrogen sulfide and BF3⋅OEt2

formed the tetrathiol, oxidized to the bis(disulfide) upon addition of iodine to complete

the synthesis of (+)-chaetocin (1.13).

Scheme 2.9. Sodeoka’s total synthesis of (+)-chaetocin.

Sodeoka originally planned to form the tetraol from the product resulting from

dimerization of 2.47. It was noted that this dimer was not stable to the radical conditions

employed above, so the synthetic plan was altered to that shown in Scheme 2.9 to

NNMe

O

O

SS

NH

HOH

NMeN

O

O

SS

HNH

HO

(+)-chaetocin (1.13)

NNMe

O

ONBoc

HOTBS

NMeN

O

O BocNH

TBSO

2.50

OH

HOOH

HO

NNMe

O

ONBoc

HOTBS

OH

HOBr

NNMe

O

ONBoc

HOTBS

Br

BrBr

NNMe

O

ONBoc

HOTBS

H

HBr

HNNMe

O

ONBoc

OTBS

NMeCbzHO2C

HONH

NH2

CO2Me

+ NBS

88%

5 steps

69%

NBS, V-70 H2O

47%

CoCl(PPh3)3

55%

H2S, BF3!OEt2;I2

44%

2.442.45 2.46

2.47 2.48 2.49

38

circumvent this problem. Recall that Movassaghi had similar difficulties in forming

related tetraol 2.39 and eventually settled on the mild oxidation discussed above.

Mere months after Sodeoka reported the first total synthesis of chaetocin,

Movassaghi published the synthesis of (+)-chaetocin (Scheme 2.10), the hexasulfide (+)-

chaetocin C (1.79), and the octasulfide (+)-12,12’-dideoxychetracin A (1.81, Scheme

2.11), all from the natural amino acids L-serine and L-tryptophan.125

Scheme 2.10. Movassaghi’s total synthesis of (+)-chaetocin.

Bromocyclization of dioxopiperazine 2.51 gave endo-tetracyclic bromide 2.52. N-

methylation, protecting group exchange, and key reductive radical dimerization afforded

the dimeric octacycle (2.53). Selective tetrahydroxylation was achieved using

Py2AgMnO4. Tetraol 2.54 is analogous to 2.50 used in Sodeoka’s synthesis, but

NNMe

O

O

SS

NH

H

NMeN

O

O

SS

HNH

(+)-chaetocin (1.13)

NNMe

O

ONH

HOAc

NMeN

O

O HNH

AcOO

O O

Me Me

SSCPh3

SS

Ph3C

O

MeMe

NNMe

O

ON HOAc

NMeN

O

O SO2PhNH

AcOO

O O

Me Me

S

S

O

MeMe

O

Me

Me

O

Me

MeN

NMe

O

ONOAc

NMeN

O

O SO2PhNH

AcOHO

OH

HOOH

NNMe

O

ON H

NMeN

O

O SO2PhNHH

H

HH

PhO2SH

PhO2S

NNH

O

ONSO2Ph

Br

HHN

NH

O

ONSO2Ph

OAc

AcO

OTBDPS OTBDPS

Br2

59%

1. MeI, LHMDS2. HF!py; AcCl3. CoCl(PPh3)3

36%

Py2AgMnO4

55%

1. H2S, TFA

53%2. i-PrCOCl

1. h" (350 nm), L-ascorbic acid, 1,4-dimethoxy- naphthalene

46%

2. N2H4, 0 °C; TrSSCl, Et3N

1. BF3!OEt2, 2,6- di-t-Bu-4-Me-py., Et3SiH

75%2. Otera's cat.

OH

OH

2.51 2.522.53

2.54 2.55

2.56

PhO2S

39

interestingly the acetate allows for differentiation of the hemiaminals, resulting in

regioselective substitution of hydrogen sulfide. Protection of resulting thiohemiaminal as

the dithioisobutyrate (2.55) served both to prevent opening of the hemiaminal under polar

protic conditions and to activate the hemiaminal to mild ionization in future steps. Mild

deprotection of the sulfonyl group was followed by chemoselective hydrazinolysis and

addition of triphenylmethanesulfenyl chloride to give disulfane 2.56. Ionization of the

isobutyrates and cyclization to the epidithiodioxopiperazines with concomitant loss of a

triphenylmethyl cation was followed by removal of the acetates using Otera’s catalyst to

afford (+)-chaetocin (1.13).

Chaetocin C (1.79) and 12,12’-dideoxychetracin A (1.81), the epitri- and

epitetrathiodioxopiperazine analogues of chaetocin, were similarly synthesized from a

common intermediate (Scheme 2.11).125

Scheme 2.11. Total syntheses of chaetocin C and 12,12’-dideoxychetracin A.

NNMe

O

ONH

HOAc

NMeN

O

O HNH

AcOO

O O

Me Me

SSn

CPh3

SSn

Ph3C

O

MeMe

NNMe

O

ONH

HOAc

NMeN

O

O HNH

AcOO

O O

Me Me

S

S

O

MeMe

O

Me

Me

O

Me

Me

(n=2): N2H4, 0 °C;TrSSCl, Et3N (86%)

or

2.57

(n=3): N2H4, 0 °C;TrSSSCl, Et3N (74%)

2.58 (n=2)2.59 (n=3)

NNMe

O

O

Sm

N H

NMeN

O

O

Sm

NHO

R

OR

2.60 (m=3, R=CF3)2.61 (m=4, R=H)

(m=3): TFAA, di-t-Bu-4-Me-py.; BF3!OEt2 (91%)

or(m=4): HCO2Ac; BF3!OEt2 (60%)

(m=3): Otera's cat., 90 °C (95%)

or(m=4): HCl, MeOH

(52%)

OAc

AcO

NNMe

O

O

Sm

NH

H

NMeN

O

O

Sm

HNH

(+)-chaetocin C (1.79), m=3(+)-12,12'-dideoxychetracin A (1.81), m=4

OH

HO

40

Hydrazinolysis of diaminodithioisobutyrate 2.57 was followed by treatment with

the corresponding sulfur source to give 2.58 or 2.59. It was necessary to protect the

indoline nitrogens as the trifluoroacetates before addition of Lewis acid to form the

polysulfide bridges (2.60 and 2.61), presumably to prevent decomposition pathways

encountered over longer reactions times necessary to the formation of the larger

polysulfide bridges. Global deprotection gave either (+)-chaetocin C (1.79) or (+)-12,12’-

dideoxychetracin A (1.81).

In 2011, Overman and coworkers reported the total synthesis of (+)-gliocladine C

(1.99, Scheme 2.12).126 Known 2-indolinone 2.62 (prepared from isatin and indole) was

reduced and Boc-protected to afford compound 2.63. Conversion to the oxindole ester

(2.64) proceeded efficiently upon treatment with 2,2,2-trichloro-1,1-dimethylethyl

chloroformate, triethyl amine, and 10 mol % of Fu’s (S)-(–)-4-pyrrolidinopyrindinyl-

(pentamethylcyclopentadienyl)iron catalyst. The oxindole was elaborated to indoline 2.65

over four steps in 54% yield. Aldol condensation with the lithium enolate of the

piperazinedione provided exclusively the Z isomer of 2.66, which readily cyclized to

hexacycle 2.67 upon treatment with BF3·OEt2. Grignard addition, silyl protection of the

resultant alcohol, asymmetric dihydroxylation, and acetylation gave advanced

intermediate 2.68. Addition of the epimeric mixture of silyl ethers to condensed hydrogen

sulfide and BF3·OEt2 gave the dithiol, oxidized to the disulfide upon exposure to oxygen.

Removal of the acetate revealed (+)-gliocladine C (1.99).

41

Scheme 2.12. Synthesis of (+)-gliocladine C.

Boyer and Movassaghi recently completed the total synthesis of two compounds

related to the gliocladines. The most direct route to the first, (+)-gliocladin B (2.73), is

detailed in Scheme 2.13.127 Bromocyclization proceeded as above, providing tetracycle

2.70 in good yield with excellent stereoselectivity (97:3 endo:exo). A Friedel–Crafts type

coupling of N-TIPS-5-bromoindole with 2.70 formed the desired 3-3’ bond. Several

indole derivatives were screened, and the 5-bromo derivative was found to give the best

yield and selectivity. The bromine and silyl protecting group were removed and the

resulting compound oxidized to diol 2.72. Exposure of this diol to sodium thiomethoxide

and trifluoracetic acid gave the bismethylthio derivative. (+)-Gliocladin B (2.73) was

revealed upon removal of the benzenesulfonyl group.

NNMe

O

OMe

NH

H

HN

(+)-gliocladine C (1.99)

S

OH

NH

O

OHHN

NBoc

O

HBocN

NBoc

O

BocN O

NBoc

OMe

BocN O

H

1. TFA, Et3SiH

2. Boc2O, DMAP; MeOH

68%

88%, 98:2 er

O

OCl ClCl

Cl

Et3N, Fu's cat., 40 °C

1. NaBH42. HC(OMe)3, PPTS, 65 °C3. LiBH4-MeOH, r.t. to 40 °C4. Dess-Martin

54%

O CCl3

MeMe

2.62 2.63 2.64

2.65

LDA, -78 °C;

75%

NNMe

O

OMeO

2.65, -78 °C; AcOH, -78 °C to r.t.

NBoc

OMe

BocN

N

NMe

OMe

O

O

80%-78 °C to -40 °C

BF3·OEt2

2.66

NNMe

O

OO

NBoc

H

BocN

62%

3. AD-mix-!, H2NSO2Me, K2OsO4·2H2O, (DHQ)2PHAL, >14:1 dr)4. Ac2O, DMAP

1. MeMgCl, -78 °C (9:1 dr)2. TBSOTf, DMAP, Et3N (3:2 dr)

NNMe

O

ONBoc

H

BocN AcOAcO

OTBSMe 47%

2. La(OTf)3, 40 °C

1. H2S, BF3·OEt2; O2

S

2.67

2.68

42

Scheme 2.13. Total synthesis of (+)-gliocladin B.

Although it has not to date been isolated from a natural source, it is worth noting

that the authors were able to convert diol 2.72 into the disulfide (+)-12-deoxybionectin A

(2.76) using conditions analogous to those discussed above in the chaetocin syntheses

(Scheme 2.14).127 Disulfide 2.76 is also easily converted to (+)-gliocladin B (2.73) by

reduction of the disulfide with sodium borohydride in the presence of iodomethane.

Scheme 2.14. Synthesis of (+)-12-deoxybionectin A and (+)-gliocladin B.

Epiccocin G (2.84) lacks a disulfide bridge and was thus not discussed in Chapter

1. Nonetheless, the structure is remarkably similar to that of epicorazine A (1.8), and a

discussion of Nicolaou’s recent total synthesis is certainly relevant in this context.128

NNMe

O

ONSO2Ph

Br

HHN

NMe

O

ONSO2Ph

Br2

75%

2.69 2.70

NNMe

O

ONSO2Ph

H

AgBF4, DTBMP, 0 °C

83%

2.71

TIPSN

BrTIPSN

Br

NNMe

O

ONSO2Ph

H

2.72

HN

OH

HO1. H2, Pd/C; Et3N·3HF2. n-Bu4NMnO4

41%

1. MeSNa, TFA-MeNO2

2. h! (350 nm), 1,4-di- methoxynaphthalene, ascorbic acid, sodium ascorbate

68%

NNMe

O

ONH

H

(+)-gliocladin B (2.73)

HN

SMe

MeS

NNMe

O

ONSO2Ph

H

2.72

HN

OH

HO

NNMe

O

ONH

H

2.74

HN

O

S

O Me

MeO

Me

Me

1. H2S, TFA-CH2Cl2, 0 °C; i-PrCOCl, pyr.2. h! (350 nm), 1,4-di- methoxynaphthalene, ascorbic acid, sodium ascorbate

47%

N2H4, 0 °C;Ph3CSCl, Et3N, 0 °C

81%

NNMe

O

ONH

H

2.75

HN

OH

SSCPh3

Hf(OTf)4

NNMe

O

ONH

H

HN

SS

NaBH4, MeI,pyr.

80%80% NNMe

O

ONH

H

(+)-gliocladin B (2.73)

HN

SMe

MeS

(+)-12-deoxybionectin A (2.76)

43

Hydroxy enone 2.77 (prepared in two steps from N-Boc-tyrosine) was converted in four

steps to hydroxy methyl ester 2.78 (Scheme 2.15). Separate deprotections to either the

amine or carboxylic acid gave two derivatives that were coupled to form amide 2.79.

Deprotection and cyclization to the dioxopiperazine was followed by conversion to the

bistrifluoroacetate (2.80), which eliminated to bisdiene 2.81 upon exposure to Pd(PPh3)4

catalyst. Treatment with S8 and NaHMDS gave a mixture of oligosulfenylated

compounds (2.82) that were readily converted to the bisdimethylthio compound (2.83)

upon reduction with sodium borohydride and addition of iodomethane. Bisendoperoxide

2.85 was generated on reaction with singlet oxygen, and addition of DBU induced a

Kornblum–DeLaMare rearrangement to give a bishydroxy enone. Reduction to the

ketone completed the total synthesis of epicoccin G (2.84).

Scheme 2.15. Total synthesis of epicoccin G.

NBoc

CO2Me

OH

HO

1. Ac2O2. Zn, AcOH3. DBU4. NaBH4, CeCl3

NBoc

CO2Me

H

HHO

1. TFA (a) or LiOH (b)2. BOP-Cl, a + b

NBoc

H

HHO NMeO2C

H

H OH

O1. TFA; Et3N2. (CF3CO)2O

N

H

HF3COCO N

O

OH

H

OCOCF3

N

H

H N

O

OH

H

Pd(PPh3)4

N

H

H N

O

OH

H

S8, NaHMDS;2.81, NaHMDS

SSnS

SSnS

2.82, n=0-6

N

H

H N

O

OH

H

NaBH4; MeI

SMe

MeS

1. O2, h!, TPP; DBU2. H2, Pd(OH)2

N

H

H N

O

OH

HSMe

MeSOH

O

OH

O

N

H

H N

O

OH

HSMe

MeS

OO

OO

H

HB:

:B

via:

2.77 2.78

2.79 2.80

2.81 2.83

epicoccin G (2.84) 2.85

47%84%

53%90%

58% (3 steps)

45%

44

The last epidithiodioxopiperazine total synthesis to be discussed was reported

recently by the Reisman group, who completed an elegant synthesis of (–)-acetylaranotin

(1.35).129 Pyrrolidine 2.88 was synthesized with >98% ee by (1,3)-dipolar cycloaddition

of cinnamaldimine 2.87 to t-butyl acrylate (2.86) and subsequent cleavage of the t-butyl

ester (Scheme 2.16). Protection of the amine and ozonolytic cleavage of the alkene

provided lactone 2.89 in good yield. Ethynylmagnesium bromide was added to the

hydroxylactone to form the hydroxy acid, which upon addition of triphenylphosphine and

DIAD underwent Mitsunobu lactonization to 2.90. Reduction to the diol (2.91) was

followed by bis-TBS protection of the alcohols and selective deprotection of the primary

alcohol. The aldehyde obtained following oxidation with Dess–Martin periodinane was

efficiently converted to chlorohydrin 2.92.

Scheme 2.16. Key pyrrolidine synthesis.

Formation of the dihydrooxepine (2.93) was realized upon treatment of alkyne

2.92 with catalytic [Rh(cod)-Cl]2 and tris(4-fluorophenyl)phosphine (Scheme 2.17).129

Elimination of HCl and hydrolysis of the ester gave acid 2.94, while deprotection of the

Teoc group and HCl elimination of the same compound gave amine 2.95. BOP-Cl–

mediated coupling of 2.94 and 2.95 delivered amide 2.96, which readily cyclized to

dioxopiperazine 2.97 after TBAF·(t-BuOH)4–induced desilylation. Synthesis of (–)-

N CO2Et

t-BuO2C

+1. CuI, Brucin-OL, DBU2. TFA, Et3SiH

39%NH

HO2C

CO2EtPh·TFA

2.88 (>98% ee)

NTeoc

CO2EtO

H

H

HO

O1. Teoc-OSu, Et3N2. O3, -78 °C; DMS

77%

NTeoc

CO2EtO

H

HO

-78 °C to 0 °C;PPh3, DIAD, 0 °C

76%

MgBr

NaBH4

86% NTeoc

CO2EtHHO

HO

NTeoc

CO2EtHTBSO

HO Cl1. TBSOTf, 2,6-lutidine, 0 °C2. AcOH3. DMP, pyr.4. pyrrolidine·TFA, NCS; NaBH4

58%

2.86

2.87 2.89

2.90 2.91 2.92

45

acetylaranotin (1.35) was completed following formation of tetrasulfide 2.98,

bisacetylation, mild reduction to the dithiol, and oxidation to the natural disulfide. This

publication marked the first total synthesis of a dihydrooxepine-containing

epidithiodioxopiperazine natural product.

Scheme 2.17. Completion of the total synthesis of (–)-acetylaranotin (1.35).

2.4: Other Relevant Syntheses: Formation of the C3-N1’ Bond

Recall that chetomin (1.12) contains a C3-N1’ linkage between two indole–

derived fragments. This unique structural feature was synthetically unprecedented when

we began our synthetic efforts toward a total synthesis of chetomin. In the last few years,

two different methods for the formation of this bond were reported in the literature. We

NN

O

O

O

OOH

(!)-acetylaranotin(1.35)

HO H H

NTeoc

CO2EtHTBSO

HO Cl

2.92

[Rh(cod)Cl]2,(4-FC6H4)3P,

85 °C

88% NTeoc

CO2EtH

ClO

TBSO

1. LiCl, Li2CO3, 100 °C2. Me3SnOH, 80 °C

1. TBAF, 0 °C2. LiCl, Li2CO3, 100 °C

48%

55%

2.94

2.95

BOP-Cl

NTeoc

CO2HH

O

TBSO

NH

CO2EtH

O

TBSO

87%

2.93

N H

O

OTBSN CO2EtH

O

TBSOO

Teoc

TBAF·(t-BuOH)4, 70 °C, N2 atm

76%

NN

O

O

O

OOH

HO H H

S8, NaHMDS;2.97, NaHMDS;

40%

S4

NaHMDS

1. AcCl, DMAP2. Et3N; O2

HS SH NN

O

O

O

OOAc

AcO H HS S

2.96 2.97

2.9832%

46

feel it prudent to discuss in the remainder of this chapter methods for the formation of the

challenging C3-N1’ bond as it relates to the synthesis of chetomin.

In 2008, Newhouse and Baran reported the total synthesis of (±)-psychotrimine

(Scheme 2.18).130,131 The key C3-N1’ bond forming reaction was completed by treating

tryptamine derivative 2.99 with N-iodosuccinimide and 2-iodoaniline to afford compound

2.100. Larock indole synthesis with alkyne 2.101 completed the carbon framework for

the top tryptamine fragment (2.102). A total synthesis of psychotrimine (2.103) was

completed in two steps from 2.102.

Scheme 2.18. Synthesis of (±)-psychotrimine via novel C3-N1’ bond formation.

Baran used the same method in the synthesis of kapakahine B (2.108, Scheme

2.19).130,132 In this instance, dipeptide 2.104 was cyclized and coupled to 2-iodoaniline to

give 2.105. Indole formation by a Larock annulation with TES-alkyne 2.106 gave indole

2.107, converted in several steps to the natural product kapakahine B (2.108).

NH

NHCO2Me

I

NH2

NIS

67%NH

NCO2Me

NH

H

I

2.992.100

Br

NH

NCO2Me

N

H

2.102

Br

Br

NHCO2Me

TMS

NHCO2Me

Pd(II)

85%

NH

NMe

N

HN

NHMe

MeHN

2.101

psychotrimine (2.103)

47

Scheme 2.19. Total synthesis of kapakahine B.

In 2008, Espejo and Rainier reported a new method for the synthesis of C3-N1’

heterodimeric indolines from bromopyrroloindoline 2.110 (Scheme 2.20, prepared in one

step from N,N’-Boc2-L-Trp-OMe).133

Scheme 2.20. Rainier’s synthetic C3-N1’ bond forming method.

Addition of potassium tert-butoxide to a stirring solution of 2.110 and an indole

(i.e. 2.113-2.117) in acetonitrile at 0 °C effects the desired bond formation in 30 minutes.

NH

NHCbz

HNO

CO2MeBn

I

NH2

NIS

65%NH

NCbz

NH

H

I

NH

O

CO2Me

Bn

BocHN NH

HN CO2Bn

TES

O Me

O

Me

Me

Pd(dppf)Cl255%

NH

NCbz

N

H

NH

O

CO2Me

Bn

BocHN HN

O Me

O

NH

CO2Bn

Me

Me

N

N

NH HN

O Me

O

NH

N

OBn

O

NOH

Me Me

OH2N

Ph

2.104 2.105

2.106

2.107 kapakahine B (2.108)

NBoc

NBoc

Br

H

CO2Me indole(2.113 - 2.117)

KOtBu, CH3CN, 0 °C

NBoc

NBoc

N

H

CO2Me

R

NBoc

NBoc

N

H

CO2Me

R

NBoc

CO2Me

NHBocNBS, PPTS

CH2Cl292%

38-82%3:1 to >95:5 endo:exo

endo (2.111) exo (2.112)2.109 2.110

NH

NH

Me

NH

NHBoc

NH

CO2Me

NHBoc

NH

NBn

PMBN

O

O

indole =

29% (5:1)2.113

81%, (3:1)2.114

48% (>95:5)2.115

38% (>95:5)2.116

76% (5:1)2.117

+

48

This method is far more general and cost effective than that reported by Baran, as a

variety of indole derivatives can be coupled directly without the added Larock annulation

step.

The utility of this method was demonstrated through the total synthesis of

kapakahine F (2.121, Scheme 2.21).134 Pyrroloindoline dimer 2.118 was synthesized as

described above and transformed to phenylalanine derivative 2.119 over several steps.

This substrate was originally intended to serve as a model for the subsequent trimethyl

aluminum induced rearrangement, but unfortunately the product resulting from the

coupling of 2.110 to a tryptophan derivative decomposed under the rearrangement

conditions. Thus, indole 2.120 was ultimately used to complete the synthesis of

kapakahine F (2.121).

Scheme 2.21. Total synthesis of kapakahine F.

The final total synthesis to be discussed in this chapter featuring a C3-N1’ bond

forming step is that of (+)-pestalazine B (2.126, Scheme 2.22), a natural product with

similar carbon framework to chetomin (1.12).135 Pestalazine B is unique in this discussion

in that, like chetomin, it is enantiomeric at the pyrroloindoline core compared to the

NBoc

NBoc

Br

H

CO2Me

KOtBu, 0 °CNBoc

NBoc

N

H

CO2Me

82%5:1 endo:exo

indole

NH

N

N

H

O

NH

CO2Et

Bn

AlMe3

0 °C to 30 °C

80% (1.5:1)N

N

N

OBn

NH2

N

N

NH2 HN

O Me

O

NH

N

OBn

O

NOH

MeMe

kapakahine F (2.121)

2.110 2.118 2.119

2.120

SO2Ph

49

above examples. Accordingly, D-tryptophan was selected as the starting material,

converted to bromopyrroloindoline 2.122 using known methods. Rainier’s coupling

methodology was employed to join 2.122 with dioxopiperazine 2.123 to form key

intermediate 2.124. Removal of the Boc groups and peptide coupling produced

tetrapeptide 2.125. Treatment of 2.125 with diethyl amine resulted in concomitant Fmoc

deprotection and cyclization to the naturally occurring dioxopiperazine, completing the

total synthesis of pestalazine B (2.126).

Scheme 2.22. Concise total synthesis of (+)-pestalazine B.

2.5: Concluding Remarks

Epidithiodioxopiperazine alkaloids possess an astonishing array of molecular

architecture and, with that, corresponding synthetic challenges to construct such

substances. The biosynthesis of many of the natural metabolites touched on in Chapter 1

has certainly inspired many of the chemists cited in this chapter, as several have sought to

exploit insights from Nature’s strategic bond constructions in a synthetic laboratory

NBoc

NBoc

N

H

CO2Me

NH

HNO

O

Ph

NBoc

NBoc

Br

H

CO2MeKOtBu, 12 °C

30%

2.122

NH

NH

HN

O

O

Ph

2.123

2.124

+

1. TMSI, 0 °C2. N-Fmoc-D-Leu, HATU, Et3N

NH

N

N

H

CO2Me

NH

HNO

O

Ph

2.125

O

NHFmoc

MeMeNH

N

N

H

NH

HNO

O

Ph

NH

O

OMe

Me

(+)-pestalazine B (2.126)

Et2NH

48% (3 steps)

50

context. Our own synthetic efforts toward chetomin and sporidesmin A as described in

the following chapters were undertaken before all of the recent reports were published,

although insight and inspiration were drawn from each as new developments in the

modern synthetic renaissance of epidithiodioxopiperazines emerged.

51

CHAPTER 3

Studies Toward the Total Synthesis of Chetomin

3:1: Introduction

Our interest in chetomin stems both from its novel mechanism of action as a

potential cancer chemotherapeutic agent and the synthetic challenge posed by the unique

architecture of the molecule. Chetomin (1.12) and related metabolites isolated from

Chaetomium sp. (Figure 3.1) are the only known epidithiodioxopiperazine alkaloids to

contain a nearly dimeric structure joined by a C3-N1’ linkage.73,74 The densely

functionalized structure contains five tetrasubstituted carbons and six stereocenters. The

synthetic challenge imposed by the epidithiodioxopiperazine rings is also not to be

overlooked, as the disulfide bridge is sensitive to oxidative, reductive, basic, and strongly

acidic conditions.

3.2: Goals and Early Studies

3.2.1: Goals for the Project

The primary goal of this project was to develop a scalable, divergent synthesis of

(+)-chetomin (1.12). We hoped to bring in as much functionality as possible when

forming the C3-N1’ bond to exploit the pseudo-dimeric architecture. Introduction of the

disulfide bridges would be reserved for a late stage in the synthesis to circumvent the

troubling sensitivity of the functional group. However, no precedent existed in the

52

infancy of this project for the formation of the key C3-N1’ bond. Our initial synthetic

efforts were focused on this bond.

Figure 3.1. Chetomin and related fungal metabolites.

3.2.2: Iminium Ion Approach

A simple model study was designed to gain access to oxindole 3.1 (Scheme 3.1).

A side chain mimicking the amino acid fragment could be added by nucleophilic addition

to iminium 3.2, formed from condensation of isatin (3.3) with indoline (3.4).

Scheme 3.1. Proposed model study for C3-N1’ bond formation.

Condensation of indoline (3.4) on N-Boc isatin showed promise at forming the

desired iminium ion (3.7), especially after we observed addition of vinyl Grignard

NH

N

NNMeS

S

O

O

OH

NMeMeN

O

HO

O

SS

chetomin (1.12)

NH

NNMe

O

O

dethio-tetra(methylthio)chetomin (1.70)

SMe

MeS

NH

NNMe

O

O

chaetocochin C (1.73)

SS

NN

NMe

O

O

SS

chaetocochin B (1.72)

N

N

NNMe

O

OSMe

MeS

O

chaetocochin A (1.71)

H

NNMeMeN

O

MeSO

SMe

H

NMeMeN

O

MeSO

SMe

N

O

NMeMeN

O

MeSO

SMe

NNMeMeN

O

MeSO

SMe

OH

HO

OHOH

HO

OH

NH

N+

NH

O

ONH

ONH

N

O

addition condensation

X

3.1 3.2 3.3

3.4

53

reagent to the condensation product (Scheme 3.2). However, further structural analysis

by NMR revealed that water was not eliminated in the condensation, and hemiaminal 3.6

was instead formed through direct addition of indoline to the C3 carbonyl of isatin. Vinyl

Grignard reagent was adding to the lactam carbonyl carbon. Numerous conditions were

attempted to effect elimination of water, but all failed to produce the desired iminium ion

(3.7).

Scheme 3.2. Attempted iminium ion formation.

Looking back, we did achieve our goal of forming a tetrasubstituted carbon on an

indole surrogate. However, no reasonable plan existed for the incorporation of the amino

acid residue, let alone with any stereocontrol.

3.2.3: Pinacol-type Rearrangement

Inspired by past Williams group chemistry and aryl pinacol-type rearrangements

recently reported by Movassaghi, we next envisioned forming the C3 tetrasubstituted

center through a pinacol-type rearrangement of tryptophan dimer 3.9 to oxindole 3.8

(Scheme 3.3).136-138 The amino acid fragment of indole 3.9 could be installed through

alkylation of 3.10, which was expected to arise from copper-mediated indole cyclization

NH

Boc2O, DMAP

NBoc

THF, 0 °-r.t.

O

O

O

O

NH

4Å m.s.toluene, reflux

K2CO3

NBoc

O

NHO

NBoc

O

N+

3.3 3.5

3.4

3.6 3.7

70%>99%

54

of aniline 3.11. The nitro alkene dimer could be accessed via coupling of bromoalkyne

3.12 with 2,3-dihydrotryptophan (3.13).

Scheme 3.3. Retrosynthesis of key intermediate 3.8 through a pinacol-type

rearrangement.

Bromoalkyne 3.12 was prepared in two steps by treating 2-nitrobenzaldehyde

with carbon tetrabromide and triphenylphosphine to afford dibromoalkene 3.15 in 91%

yield, which under basic conditions eliminated to form 3.12 in quantitative yield (Scheme

3.4).139

Scheme 3.4. Synthesis of 2-alkynyl aniline derivative 3.17.

Standard copper-mediated coupling of the bromoalkyne with indoline (3.4) failed

to proceed in appreciable yields.140 Microwave-assisted coupling successfully formed

alkyne 3.16, albeit in 37% yield after optimization on small scale (<200 mg). Reduction

Cu-mediatedcouplingcyclization

alkylationPinacol-type

rearrangement

NH

O

NNH2

CO2H

CO2HH2N

NH

N NH2

CO2HNH2

CO2H

NH

N NH2

CO2H

NH2

N

NH2

CO2H

NO2

Br

NH

NH2

CO2H

reduction

3.8 3.9

3.10

3.11 3.12 3.13

NO2

BrCuSO4, K3PO4

NH

+

NO2

N

toluene, µw

CHO

NO2CH2Cl2

CBr4, PPh3Br

BrNO2

CH2Cl2

KOH/H2OBnEt3NCl

Zn, NH4Cl

NH2

N

EtOH DMF / Eth.Gly. NH

NCuI

3.15 3.12

3.16 3.17 3.18

3.1491% 100% 37%

61%

3.4

55

of the nitro group using zinc and ammonium chloride gave aniline 3.17 in 61% yield,

although copper iodide mediated indole formation failed to produce the desired indole

(3.18).

Protected 2,3-dihydrotryptophan was synthesized in parallel to this model study to

be used in place of indoline (3.4) above. N-Cbz tryptophan was treated with iodomethane

to form the methyl ester (3.20) in quantitative yield (Scheme 3.5). Reduction of 3.20

using BH3·Me2S in THF and TFA gave 2,3-dihydrotryptophan derivative 3.21 in 79%

yield. Coupling using the previously optimized conditions afforded alkyne 3.22 in 45%

yield, although reduction to aniline 3.23 gave only trace amounts of the desired product.

Scheme 3.5. Synthesis of 2,3-dihydrotryptophan alkyne derivative.

3.3: Evolution of Coupling Strategy

3.3.1: Witkop’s Pyrroloindole

At this point, we took a step back and looked to nature for inspiration. It is

unlikely that fungi synthesize chetomin by such contrived routes. If chetomin is derived

naturally from two molecules each of tryptophan and serine, why not start with

tryptophan and serine–optically pure, readily available compounds? Moreover, it is likely

NH

NHCbz

CO2H

MeI, KHCO3

DMFNH

NHCbz

CO2Me

BH3·Me2STHF/TFA

NH

NHCbz

CO2Me

CuSO4, K3PO4

toluene,µw

NO2

NNHCbz

CO2Me

Zn, NH4ClEtOH

NH2

NNHCbz

CO2Me

3.12,

3.20 3.21

3.22 3.23

100% 79%

45% trace

3.19

56

that the pyrroloindoline core is formed in nature prior to coupling with the second half of

the molecule, if not concomitantly. Witkop’s pyrroloindole (3.24) was identified as a

synthetically useful intermediate in the synthesis of chetomin that could be formed

readily from tryptophan.141 Halogenation of the indole double bond would provide a

handle that could be substituted with another indole, ideally tryptophan, all with the

potential for some stereocontrol (Scheme 3.6).

Scheme 3.6. Witkop’s pyrroloindole inspiration.

Retrosynthetically, we could form advanced intermediate 3.26 from the coupling

of bromopyrroloindoline 3.28 and a suitably protected tryptophan derivative (3.27,

Scheme 3.7). Both of these coupling partners could be synthesized in a few steps from

tryptophan.

Scheme 3.7. Retrosynthetic plan using pyrroloindoline.

Tryptophan methyl ester (3.30) was protected as the trifluoroacetate amide (3.31),

then cyclized to pyrroloindole 3.32 using t-BuOCl in 63 % overall yield (Scheme 3.8).

Treatment of 3.32 with sodium hydride and NBS did not provide the desired

bromopyrroloindole.142

NH

NAc

CO2Me

NNAc

CO2MeX

Witkop's pyrroloindole (3.24) 3.25

NR'

NR'

CO2RBr

H

NH

R'HN

RO2C

NR'

N

NR'

CO2R

NR'

NHR'

CO2R

3.29

halocyclizationcoupling

RO2C

R'HN

3.26 3.28

3.27

57

Scheme 3.8. Attempted synthesis of 3-bromopyrroloindole.

3.3.2: Indole–Aniline Coupling

This stepwise procedure to 3.33 was put on hold after a report from the Baran

group was published, showing an analogous sequence that showed promise at reducing

our synthesis of 3.26 to one step. Baran showed that treatment of N-Boc-Trp-OMe (3.34)

and 2-iodoaniline with N-iodosuccinimide in acetonitrile afforded the desired C-N bond

and C-3 quaternary center in one pot (Scheme 3.9).130-132

Scheme 3.9. Formation of key C-3 quaternary center.

We repeated this chemistry using indoline in place of 2-iodoaniline, but only

starting material was recovered (Scheme 3.10). Ideally, this reaction would be done with

tryptophan or 2,3-dihydrotryptophan, eliminating the need for a Larock indole synthesis

with the iodoaniline. To our disappointment, the reaction only worked as originally

reported with 2-iodoaniline, despite numerous attempts at the reaction with indoline (3.4),

aniline (3.37), or 7-iodoindoline143 (3.36).

NH

NH2

CO2Me

EtCO2CF3

MeOH, Et3NNH

NH

CO2Me

O CF3

t-BuOClCH2Cl2, Et3N

NH

NCOCF3

CO2MeNaH, NBS

NNCOCF3

CO2MeBr

100%

3.31

63%

3.32 3.33

3.30

NH

NHBoc

CO2Meo-Iodoaniline,

NISCH3CN

-42°C to -30°CNH

NBoc

CO2MeNH

I

H78%

3.353.34

58

Scheme 3.10. Attempted formation of functionalized C3-N bond.

A suitable TMS-acetylene derivative was synthesized from L-serine methyl ester

(3.39) and TMS-acetylene (Scheme 3.11). Serine methyl ester (3.39) was N-Boc

protected (3.40) and the alcohol converted to the iodide (3.41) upon treatment with

triphenylphosphine, imidazole, and iodine. TMS-acetylene bromide (prepared in one

step, 72% yield) was coupled to serine-derived halide 3.41 to afford the desired alkyne

(3.42) in 40% yield.132

Scheme 3.11. Key alkyne synthesis.

Alkyne 3.42 was subjected to Larock annulation with iodoaniline 3.12 to form

indole 3.43. Saponification to the diacid (3.44) proceeded in good yield. While this route

afforded access to advanced intermediate 3.44 in decent yields, it suffered from high step

count due to the necessary Larock annulation and provided access to a diastereomer of

chetomin. We again looked back to the convergent, stepwise approach that we had

NH

NH

NHBoc

CO2Me

CH3CN-42°C to -30°C

NH

NBoc

CO2MeN

NH

I

NH2

NISR1

R2

R3

+

or

or H3.34

3.4

3.36

3.373.38

MeO2C NH2

OH Boc2OCH3CN, Et3N MeO2C NHBoc

OH PPh3, imidazoleI2, CH2Cl2

MeO2C NHBoc

I

CuCN, Zn MeO2C NHBoc

Br TMS

TMS

78% 3.40 55%

3.41 40% 3.42

3.39

59

previously envisioned (Scheme 3.7) and attempted the desired sequence in a stepwise

manner.

Scheme 3.12. Larock indole synthesis of tryptophan dimer.

3.3.3: Coupling to exo-3-bromopyrroloindoline

N-Cbz-Trp-OMe (3.20) was synthesized in >99% yield from tryptophan and the

indole nitrogen protected as the Boc carbamate (3.46). Per a report published by Espejo

and Rainier in 2008, addition of NBS and PPTS to 3.46 afforded exo-3-

bromopyrroloindoline 3.47 in 89% yield (Scheme 3.13).133,144 Deprotonation by KOtBu

in the presence of indole formed the C3-N bond in 28% yield and resulted in

epimerization of the methyl ester to the thermodynamically more favorable endo position

(3.48).133

Scheme 3.13. Coupling of 3-bromopyrroloindoline.

NH

NBoc

CO2MeN

NHBoc

CO2Me

LiOHTHF, H2O

NH

NBoc

CO2HN

NHBoc

CO2H

H H70%

3.43

92%

3.44

NH

NBoc

CO2MeNH

I

H

3.35

DMF

Pd(OAc)2, NaOAc,LiCl

3.42

NH

NH2

CO2H

CbzClMeOH, NaHCO3

NH

NHCbz

CO2H

MeI, KHCO3

DMFNH

NHCbz

CO2Me

Boc2O, NaOHBu4NHSO4

CH2Cl2

NBoc

NHCbz

CO2Me

NBoc

NCbz

CO2MeBrNBS, PPTS

CH2Cl2

indole,KOtBuCH3CN

NBoc

NCbz

CO2MeN

HH

100%

3.19

100%

3.2098%

89%

3.46 3.47

28%

3.48

3.45

60

This result evolved into our current strategy for the formation of the C3-N1’ bond.

The stereochemistry of 3.48 matches the enantiomer of what would be needed for a

synthesis of chetomin (i.e. 3.51, Scheme 3.14). We could either form the

bromopyrroloindoline from L-tryptophan and to it couple D-tryptophan and D-serine to

synthesize the enantiomer of chetomin, or we could synthesize the D-tryptophan-derived

pyrroloindoline (3.52) and use the natural amino acid isomers for the remaining three

couplings in a synthesis of (+)-chetomin (1.12). Initially, however, only the cheaper L-

isomers were used for all four fragments as conditions were worked out for the remaining

steps.

Scheme 3.14. Retrosynthetic analysis of (+)-chetomin.

3.3.4: Attempted Coupling to Tetracyclic Bromide

The next goal was to test a more divergent approach. Ideally, the dioxopiperazine

ring could be formed prior to coupling with indole (Scheme 3.15).

NH

NNH

O

O

OTBSH

N

NMe

MeN

O

O

OTBS

tetra-thiolation

C3-N1' coupling(L-Trp)

HOOH

HO

OH

oxidation

NNMe

O

O

SS

NH

HOH

MeNNMe

O

O

SS

NOH

(+)-chetomin (1.12)

NH

NNMe

O

O

OTBSH

N

NMe

MeN

O

O

OTBS

L-Ser (2 eq), cyclization,

N-methylation

NH

NHCO2Me

H

N

CO2Me

NHBoc

NBoc

NBocCO2Me

Br

H NH

NH2

CO2H

D-tryptophan (3.53)

bromo-cyclization

3.52

3.49 3.50

3.51

61

Scheme 3.15. Coupling with more advanced bromopyrroloindoline.

Unfortunately, synthesis of 3.55 was more challenging than expected. Peptide

coupling of tryptophan derivative (3.56) with N-Me-L-Ser-OMe initially suffered from

poor yield. Eventually, the coupling was optimized using EDCI as the coupling reagent to

afford dipeptide 3.57a in good, reproducible yield (Scheme 3.16). Cyclization to the

dioxopiperazine (3.58a) failed under methods traditionally used in the Williams group

(i.e. refluxing toluene with catalytic 2-hydroxy pyridine following Boc deprotection).

However, microwave heating of neat N-Boc dipeptide 3.57a at 180 °C effected

deprotection and cyclization to the dioxopiperazine (3.58a) in two minutes. The

recovered product did not require any purification before proceeding to the next step.

Scheme 3.16. Optimized peptide coupling and dioxopiperazine formation.

Gratifyingly, this procedure was generally applicable to a variety of related

dipeptides. Dioxopiperazines 3.58a-e were all synthesized with consistently good yield

on scales up to 500 mg (Scheme 3.17).

Several of the dioxopiperazines were cyclized to the corresponding

bromopyrroloindolines (3.55, Scheme 3.18). Despite numerous attempts, coupling of

indole failed using conditions analogous to those first used in Scheme 3.13.

NN

NR2

OR3

O

O

Br

HR1N

NNR2

OR3

O

O

N

HR1

3.54 3.55

NHN

NMeOH

O

OSO2PhNNHBoc

NMe

O

SO2Ph

CO2Me

OH

NNHBoc

SO2Ph

CO2H N-Me-L-Ser-OMe,EDCI, iPr2NEt

CH2Cl2

3.56 3.57a 3.58a

µw (180 °C)neat

86% 94%

62

Scheme 3.17. Microwave assisted dioxopiperazine synthesis.

Scheme 3.18. Attempted coupling with indole.

3.4: Epidithiodioxopiperazine Formation

While unsuccessfully attempting to couple indole or tryptophan to 3.55, we also

began to explore methods for the introduction of sulfur to the same dioxopiperazine.

Epidithiodioxopiperazine 3.63, essentially a monomer of chetomin, was synthesized by a

general method recently described by the Sodeoka group in Japan (Scheme 3.19).123,124

Benzenesulfonamide protection of L-tryptophan (3.59) gave compound 3.56, to which L-

Ser-OMe was coupled to afford dipeptide 3.57d. The primary alcohol was protected as

the silyl ether (3.57e) and suffered microwave-induced deprotection and cyclization to

dioxopiperazine 3.58e. Treatment with bromine affected the bromocyclization to endo-

pyrroloindoline 3.55e. Addition of iodomethane successfully methylated the secondary

amide, but also resulted in the loss of the silyl ether to primary alcohol 3.60. After

reprotecting the alcohol, diol 3.62 was formed following radical bromination and

NNHBoc

N

O

R1

CO2Me

OR3

µw (180 °C)neat

NHN

NR2

OR3

O

OR1

R2

75-98%

3.57a (R1=SO2Ph, R2=Me, R3=H)3.57b (R1=H, R2=Me, R3=H)3.57c (R1=H, R2=H, R3=H)3.57d (R1=SO2Ph, R2=H, R3=H)3.57e (R1=SO2Ph, R2=H, R3=TBS)

3.58a-e

NHN

NR2

OR3

O

OR1 NN

NR2

OR3

O

O

Br

HR1 N

NNR2

OR3

O

O

N

HR1

Br2CH3CN

indole,KOtBuCH3CN

3.58 3.55 3.5438-63%

63

substitution of 3.61. Addition of the diol to condensed hydrogen sulfide and BF3·OEt2

and oxidation of the resultant disulfide with iodine gave epidithiodioxopiperazine 3.63.

Scheme 3.19. Formation of the disulfide bridge.

3.5: Attempted Core Construction

3.5.1: Problematic Peptide Couplings

With a method for the formation of an epidithiodioxopiperazine in hand, we went

back to the method described in Scheme 3.13 for construction of the key C3-N1’ bond.

N-Boc-tryptophan methyl ester (3.34) was prepared and further protected as the

benzenesulfonamide (3.64, Scheme 3.20). Cyclization to the bromopyrroloindoline

(3.65) proceeded readily upon treatment with NBS and PPTS, and coupling of 3.65 with

N-Boc-Trp-OMe (3.34) gave 3.66 in poor yield (34%). Saponification to diacid 3.67 was

followed by peptide coupling with serine methyl ester to give peptide 3.68. TBS

NH

CO2H

NHBoc 1) LHMDS2) PhSO2Cl

NSO2Ph

CO2H

NHBocL-Ser-OMe,

EDCIiPr2NEt, CH2Cl2 N

HPhO2SN NHBoc

O

CO2Me

OH

TBSCl, imidDMF N

HPhO2SN NHBoc

O

CO2Me

OTBSµw, 180°C

NSO2Ph

HNNH

O

O

OTBS

NSO2Ph

NNH

O

O

OTBS

Br

Br2

MeCN

NSO2Ph

NNMe

O

O

OH

BrK2CO3, MeIacetone

NSO2Ph

NNMe

O

O

OTBS

BrTBSCl

1. NBS, V-70

NSO2Ph

NNMe

O

O

OTBS

Br

OH

OH

2. H2O, MeCN1. BF3!OEt2, H2S2. I2

NSO2Ph

NNMe

O

O

OH

BrSS

3.59 3.56 3.57d

3.57e 3.58e

3.55e 3.60 3.61

3.62 3.63

79% 66%

90%98%

38%

98%47%

30% 42%

64

protection of the primary alcohols proceeded in good yield, although cyclization to the

dioxopiperazines (3.70) using the previously described microwave conditions failed, as

did the reaction with traditional thermal conditions.

Scheme 3.20. Coupling to 3-bromopyrroloindoline and attempted

dioxopiperazine formation.

Deprotection of the Boc groups of intermediate 3.66 and peptide couplings on the

resultant amines gave tripeptide 3.71 (Scheme 3.21). Intriguingly, formation of the

tetrapeptide of 3.66 was never achieved, despite numerous attempts with varying

coupling conditions and protecting groups on the amino acid functionalities.

NH

CO2H

NHBocDMF, KHCO3

NH

CO2Me

NHBocBu4NHSO4,

NaOH

MeI PhSO2Cl

NSO2Ph

CO2Me

NHBoc

CH2Cl2

NBS, PPTS

NSO2Ph

NBocCO2Me

BrNH

CO2Me

NHBoc

KOtBu

EDCI, iPr2NEt,L-Ser-OMe

NSO2Ph

NBoc

NCO2Me

MeO2C

BocHN

NSO2Ph

NBoc

NBocHN O

NH CO2Me

OH

O

NH

MeO2C

HO

THF/H2O

NSO2Ph

NBoc

NCO2H

HO2C

BocHN

LiOH

TBSCl, imidDMF

NSO2Ph

NBoc

NBocHN O

NH CO2Me

OTBS

O

NH

MeO2C

TBSO

µw, 180°C

NSO2Ph

N

NNH

NH

HN

O

OTBSO

O

O

TBSO

3.59 3.34 3.64

3.65

3.66

3.67 3.68

3.69 3.70

3.34

95%>99%

>99% 34% 97%

33% 87%CH2Cl2

65

Scheme 3.21. Attempted synthesis of alternate dioxopiperazine precursor.

3.6: Synthesis of Heptacyclic Core

3.6.1: Synthesis of Dioxopiperazines

We reasoned that the failed cyclization attempts in Scheme 3.20 and peptide

coupling problems in Scheme 3.21 could be due to steric congestion caused by the

indoline protecting group. Thus, N,N’-Boc2-D-Trp-OMe was prepared from D-tryptophan

(3.73, Scheme 3.22). Bromocyclization proceeded stereoselectively to exo-

bromopyrroloindoline 3.74. Coupling with N-Boc-L-Trp-OMe gave endo-intermediate

3.75 in decent yield, while deprotection of the carbamate gave the key intermediate for

peptide coupling. In the absence of an aniline protecting group, peptide coupling

proceeded in 44% yield to tetrapeptide 3.76. With the Fmoc amines, deprotection and

cyclization to dioxopiperazine 3.77 proceeded in one pot upon treatment with morpholine

in THF with gentle heating. At this point, all of the amides needed to be methylated prior

to the introduction of sulfur, which first required protection of the aniline nitrogen.

Selective Boc protection at this position was never achieved, nor was protection of

tetrapeptide 3.76 to 3.78.

NSO2Ph

NBoc

NCO2Me

MeO2C

BocHN

NSO2Ph

NH

NCO2Me

MeO2C

NH

2. N-Cbz-L-Ser-OTBS EDCI, iPr2NEt

O

TBSOCbzHN

3.66 3.71

1. TFA

73%

66

Scheme 3.22. Synthesis of dioxopiperazine 3.77.

3.6.2: Initial N-Methyl Amino Acid Incorporation

It was clear that N-methyl tryptophan and serine derivatives needed to be prepared

to avoid late stage N-methylation. Months of laborious chemistry went in to preparing

these seemingly simple derivatives, and ultimately bromopyrroloindoline 3.74 was

coupled to N-Me,Boc-L-Trp-OMe (Scheme 3.23). Deprotection of the two Boc groups

gave 3.80, a key intermediate used extensively in the remainder of this chapter. Peptide

coupling with N-Me,Boc-Ser(OBn) gave tetrapeptide 3.81, which was cyclized to

NBoc

NBocCO2Me

Br

KOtBu, MeCNN-Boc-L-Trp-OMe

NH

NH2

CO2H

3. NBS

1. SOCl2, MeOH2. Boc2O

H

NBoc

NBocCO2Me

H

N

CO2Me

NHBoc

58%

2. N-Fmoc-Ser(OTBS), T3P, iPr2NEt

NH

NCO2Me

H

N

CO2Me

HNO

NHFmoc

OTBS

O

NHFmoc

OTBS

1. TFA

NH

NNH

O

O

OTBSH

N

NH

HN

O

O

OTBS

morpholineTHF

44% 85%

conditionsBoc2O, Et3N, MeOHBoc2O, EtOHBoc2O, Et3N, DMFBoc2O, Et3N, THFBoc2O, DMAP, CH3CN

NBoc

NNH

O

O

OTBSH

N

NH

HN

O

O

OTBS

3.79

NRNRNRNRInseparable

Boc2O, DMAP, CH3CN(inseparable mixture)

NBoc

NCO2Me

H

N

CO2Me

HNO

NHFmoc

OTBS

O

NHFmoc

OTBS

3.73 3.74

3.75

3.76 3.77

3.78

45%

67

dioxopiperazine 3.82 in excellent yield after Boc deprotection and addition of

morpholine.

Scheme 3.23. Synthesis of (N-Me)3 dioxopiperazine.

Initially we attempted to again protect the aniline nitrogen before subjecting the

material to radical bromination conditions. All attempts to Boc or TBS protect 3.82 failed

(Scheme 3.24). We decided to move forward to try the bromination anyway on diol 3.84,

but the benzyl ether resisted all deprotection attempts.

A serine derivative lacking the benzyl group was added to diamine 3.80, but this

coupling failed to give the desired tetrapeptide (Scheme 3.25). The benzyl group seemed

to be a poor choice of protecting group for this intermediate, necessitating the synthesis

of N-Me,Fmoc-Ser(OTBS).

NBoc

NBocCO2Me

Br

2. TFA

1. N-Me,Boc-L-Trp-OMe KOtBu, MeCN

H

NH

NHCO2Me

H

N

CO2Me

NHMe

40%

N-Me,Boc-Ser(OBn),T3P, iPr2NEt

42%

3.80

NH

NCO2Me

H

N

CO2Me

MeNO

NMeBoc

OBn

O

NMeBoc

OBn

3.81

NH

NNMe

O

O

OBnH

N

NMe

MeN

O

O

OBn

2. morpholine, THF1. TFA

98%

3.82

3.74

CH2Cl2

68

Scheme 3.24. Attempted protection and deprotection of 3.82.

Scheme 3.25. Attempted peptide coupling with N-Me,Boc-Ser(OH).

3.6.3: Bypassing the Serine Side Chain with Sarcosine

While working out the synthesis of an appropriate N-Me serine derivative, the

entire serine side chain was eliminated from the synthesis. N-Fmoc sarcosine (3.87) was

prepared and coupled to diamine 3.80 (Scheme 3.26). Cyclization to dioxopiperazine

3.88 proceeded smoothly. However, the radical bromination and hydroxylation did not

produce the desired tetraol (3.89).

NH

NNMe

O

O

OBnH

N

NMe

MeN

O

O

OBn

conditions

NR

NNMe

O

O

OBnH

N

NMe

MeN

O

O

OBn

3.83a (R=Boc)3.83b (R=TBS)

NH

NNMe

O

O

OBnH

N

NMe

MeN

O

O

OBn

Pd/C or Pd(OH)2

NH

NNMe

O

O

OHH

N

NMe

MeN

O

O

OH

MeOH

conditionsBoc2O, DMAPBoc2O, NaOH, Bu4NHSO4Boc2O H3PW12O40TBSCl, Et3N, DMAP

NRNRNRNR

3.82

3.82 3.84

NH

NHCO2Me

H

N

CO2Me

NHMe

NH

NCO2Me

H

N

CO2Me

MeNO

NMeBoc

OH

O

NMeBoc

OH

N-Me,Boc-Ser,T3P, iPr2NEt

3.80 3.85

CH2Cl2

69

Scheme 3.26. Synthesis of sarcosine-derived dioxopiperazine.

3.6.4: Completion of the Carbon Skeleton of Chetomin

Meanwhile, N-Me,Fmoc-Ser(OTBS) was successfully prepared and coupled to

3.80 (Scheme 3.27). Dioxopiperazine 3.90 was easily formed as described above.

Scheme 3.27. Synthesis of (N-Me)3 dioxopiperazine.

3.6.5: Attempted Epidithiodioxopiperazine Formation

Numerous attempts have been made to convert both the serine- and sarcosine-

derived dioxopiperazines (3.90 and 3.88) to the corresponding epidithiodioxopiperazines

CO2H

NHMe

FmocOSu, Na2CO3 CO2H

NMeFmocdioxane, H2O>99%

+1. T3P, iPr2NEt2. morpholine, THF

NH

NHCO2Me

H

N

CO2Me

NHMe

52%

NH

NNMe

O

OH

N

NMe

MeN

O

O 1. V-70, NBS2. H2O, MeCN pH = 7

NH

NNMe

O

OH

N

NMe

MeN

O

O

HO OH

OH

HO

3.86 3.87

3.80

3.88 3.89

1. T3P, iPr2NEt, N-Me,Fmoc-Ser(OTBS)2. morpholine, THF

NH

NHCO2Me

H

N

CO2Me

NHMe

30%

NH

NNMe

O

OH

N

NMe

MeN

O

O

OTBS

OTBS

3.903.80

70

(1.12 and 3.91, Scheme 3.28). Much to our disappointment, both have refused to yield to

our continued efforts.

Scheme 3.28. Attempted epidithiodioxopiperazine formations.

3.7: Proposed Future Studies

3.7.1: Proposed Synthesis of Tetraol

We have successfully synthesized the carbon framework of chetomin. The final

key to a total synthesis is the oxidation of this core (3.90 or 3.88) to tetraols 3.92 or 3.89

(Scheme 3.29). Numerous examples (see Chapter 2) suggest that this compound should

readily form intermediate iminium ions such as 3.93 and 3.94. Addition of sulfur should

occur at the less hindered faces of the dioxopiperazines.122-125,127

NH

NNMe

O

OH

N

NMe

MeN

O

O

OTBS

OTBS

NNMe

O

O

SS

NH

HOH

MeNNMe

O

O

SS

NOH

1. NBS, V-70; H2O2. H2S; I2

NH

NNMe

O

OH

N

NMe

MeN

O

O

NNMe

O

O

SS

NH

H

MeNNMe

O

O

SS

N

3.90 1.12

3.88 3.91

1. NBS, V-70; H2O2. H2S; I2

71

Scheme 3.29. Key tetraols and iminium intermediates.

Synthesis of the tetraols should be possible either by the method previously

attempted (radical bromination and substitution) or by mild oxidation with

bis(pyridine)silver(I) permanganate (Scheme 3.30). In the synthesis of gliocladin B, n-

Bu4NMnO4 was also used as an oxidant for this reaction.127 Repeated trials will be

necessary to produce optimal conditions, requiring sufficient time, material, and

persistence. Three potential problems may complicate this route: (1) The free indoline

nitrogen may require protection if it proves to be reactive under the proposed radical or

oxidation conditions. This has previously been a difficult protection, and while a Boc or

silyl group would be ideal, a trifluoroacetate group would also be appropriate (although it

would introduce an additional deprotection step). (2) The desired hemiaminal may very

well be unstable and require additional functionalization after it is formed before sulfur

can be introduced. (3) The serine-derived bis(dioxopiperazine) (3.90) is susceptible to

elimination of the primary alcohol.

NH

NNMe

O

OH

N

NMe

MeN

O

O

OTBS

NH

NNMe

O

OH

N

NMe

MeN

O

O

3.92

3.89

HOOH

OTBS

OH

HO

HO OH

OH

HO

NH

NNMe

O

OH

N

NMe

MeN

O

O

OTBS

OTBS

NH

NNMe

O

OH

N

NMe

MeN

O

O

3.94

3.93

72

Scheme 3.30. Possible oxidations.

3.7.2: Bypassing the Tetraol

It would also be worth attempting the addition of sulfur directly to

dioxopiperazines 3.90 and 3.88. Both Nicolaou and Reisman took this approach in their

respective epidithiodioxopiperazine syntheses, using S8 and NaHMDS (Scheme

3.31).128,129

Scheme 3.31. Alternative sulfenylation.

3.7.3: Late-Stage Alkylation of Sarcosine Analogue

Lastly, if access to sarcosine-derived epidithiodioxopiperazine 3.91 proves to be

the most efficient route, conversion to chetomin can occur using inspiration from Kishi’s

early syntheses of sporidesmin and gliotoxin (Scheme 3.32).113-118 Reduction of the

NH

NNMe

O

OH

N

NMe

MeN

O

O

R

R

3.90 (R=CH2OTBS)3.88 (R=H)

NH

NNMe

O

OH

N

NMe

MeN

O

O

R

R

3.92 (R=CH2OTBS)3.89 (R=H)

HOOH

OH

HO

NBS, V-70; H2Oor

Py2AgMnO4

NNMe

O

OR

SS

NH

H

MeNNMe

O

OR

SS

N

1.12 (R=CH2OTBS)3.91 (R=H)

NH

NNMe

O

OH

N

NMe

MeN

O

O

R

R

3.90 (R=CH2OTBS)3.88 (R=H)

1. S8, NaHMDS2. deprotection3. I2

73

disulfide bridges and protection as the thioacetals would give 3.95. Alklation with

BOMCl would install the protected serine side chain (3.96), which could be deprotected

and the disulfide revealed to give chetomin (1.12).

Scheme 3.32. Proposed conversion of 3.91 to chetomin.

3.8: Concluding Remarks

It will be difficult to leave this project behind with the total synthesis of chetomin

so near to completion. However, short of a completed synthesis, we did achieve the

remaining goals for this project in the synthesis of dioxopiperazines 3.88 and 3.90. The

key C3-N1’ bond formation works for a variety of substrates and is a convergent

approach to the core structure. A variety of peptide couplings were optimized for this

system, producing di- and tetrapeptides in respectable yields. Cyclization of the

dioxopiperazine rings is now performed consistently and in high yields. Moreover, the

entire sequence is scalable and allows quick access to gram quantities of material. All

NNMe

O

O

SS

NH

H

MeNNMe

O

O

SS

N

3.91

NNMe

O

ONH

H

MeNNMe

O

O

SS

N

3.95

Ar

SS

Ar1. NaBH4

2. p-MeO-PhCHO

NNMe

O

ONH

H

MeNNMe

O

O

SS

N

3.96

Ar

SS

Ar

OBn

OBn

PhLi, BOMCl

NNMe

O

O

SS

NH

HOH

MeNNMe

O

O

SS

NOH

1.12

1. BCl32. mCPBA3. BF3·OEt2

74

that remains is synthesis of the disulfide bridges to complete a total synthesis of (+)-

chetomin.

75

CHAPTER 4

Synthetic Approach to Sporidesmin A

4:1: Introduction

In 2010, we had the opportunity to write a book chapter on biomimetic syntheses

of tryptophan-derived dioxopiperazines, in which Kishi’s elegant synthesis of (±)-

sporidesmin A (1.11) was featured (Figure 4.1).145 We were drawn to the densely

functionalized structure of sporidesmin A, including the polysubstituted aromatic ring,

pyrroloindoline core, hydroxyl groups at C3 and C12, and the epidiothiodioxopiperazine

ring. We hoped that chemistry developed for the synthesis of chetomin (see Chapter 3)

could be applied to an improved, stereoselective synthesis of sporidesmin A.

As detailed in Chapter 1, sporidesmin A was isolated from Pithomyces chartarum

in 1959 as part of a forty-year nationwide search for the cause of facial eczema, a disease

that was plaguing sheep herds of New Zealand.63-65 Facial eczema causes extensive liver

damage in sheep and has been responsible for massive economic losses in New Zealand

since the late 1890s. A dental nurse allegedly discovered that the addition of zinc sulfate

to drinking water could prevent the disease, a measure still endorsed by veterinarians.146

Epidithiodioxopiperazines are known to form a 2:1 complex with zinc ions, inhibiting the

generation of the superoxide anion radical and likely providing the observed protective

effects of zinc against sporidesmin.69,70 The toxicity of sporidesmin toward sheep was

responsible for many of the early advances made in the study of

76

epidithiodioxopiperazines, including the isolation and characterization of sporidesmin

and related metabolites (Figure 4.1).

Figure 4.1. Sporidesmin A and related fungal metabolites.

No reasonable argument can be made advocating that a new synthesis of

sporidesmin A will have any direct impact on mankind, medicine, or even sheep herds in

New Zealand. Our interest is purely academic. Sporidesmin A is a synthetically

challenging molecule, and pursuit of a total synthesis will allow us both to explore the

scope of similar reactions employed toward the total synthesis of chetomin and to expand

our general understanding of the reactivity of epidithiodioxopiperazine alkaloids.

4.2: Retrosynthetic Analysis

The primary goal of this project was to develop a scalable total synthesis of (–)-

sporidesmin A (1.11). Compared to chetomin (1.12), sporidesmin A presents several

unique synthetic challenges, the most significant of which is a highly substituted aromatic

NNMe

N

O

OMeMeO

MeO

ClHO

H

SS

OH

Me NNMe

N

O

OMeMeO

MeO

ClHO

H

SS

Me NNMe

N

O

OMeMeO

MeO

ClHO

H

SS

OH

S

NNMe

N

O

OMeMeO

MeO

ClHO

H

MeS

SMe

HO

Me NNMe

N

O

OMeMeO

MeO

ClHO

H

s

OH

Me NNMe

N

O

OMeMeO

MeO

ClHO

H

MeSHO

NNMe

N

O

OMeMeO

MeO

ClHO

H

sOH

Me NNMe

N

O

OMeMeO

MeO

Cl

H

SS NMe

NMe

N

O

OHMeO

MeO

HO

H

SS

MeCl

HO

ss

s ss

sporidesmin G (1.67)

sporidesmin E (1.65)

sporidesmin J (1.69)sporidesmin H (1.68)

sporidesmin F (1.66)sporidesmin D (1.64)

sporidesmin C (1.63)sporidesmin B (1.62)sporidesmin A (1.11)

77

ring. We hoped to develop an efficient synthesis of the requisite substituted L-tryptophan

derivative and to apply the previously used bromocyclization reaction (Scheme 3.13) to

form the pyrroloindoline core.

As with chetomin, we planned to introduce the disulfide bridge of (–)-sporidesmin

A (1.11) at a late stage in the synthesis from hemiaminal 4.1 (Scheme 4.1). The

hemiaminal could be formed by oxidation of tetracycle 4.2 after forming the

pyrroloindoline core through oxidative cyclization and N-methylation of dioxopiperazine

4.3. This intermediate is similar to many of the dioxopiperazines prepared in the previous

chapter and was envisioned to arise via coupling of 4.4 with alanine, followed by

cyclization.

Scheme 4.1. Retrosynthetic analysis of (–)-sporidesmin A.

One key to an asymmetric synthesis is the preparation of β-hydroxytryptophan

derivative 4.4. We hoped to introduce both stereocenters and the amino acid portion of

this intermediate through Sharpless asymmetric aminohydroxylation of unsaturated ester

4.5. This ester could be formed by Wittig olefination of the aldehyde product of a

CHO

OMeHO

OMeMeO

Cl

NO2

NO2

OMeMeO

Cl

NH

OMeMeO

Cl

NMe

CO2Me

OMeMeO

Cl

NMe

CO2MeHONHCbz

NMe

Cl

MeOOMe

HNNH

O

OMe

TBSO

NMe

Cl

MeOOMe

NH

TBSOTBSO

NMe

O

OMe

(!)-sporidesmin A (1.11)

NMe

Cl

MeOOMe

NH

HOHO

NMe

O

OMe

SS

NMe

Cl

MeOOMe

NH

TBSOTBSO

NMe

O

O

Me

HO

OH

thiolation oxidation

oxidativecyclization

peptidecoupling

amino-hydroxylation

alkylation,Wittig

indoleformation substitution

4.1 4.2

4.3 4.4 4.5

4.6 4.7 vanillin (4.8)

78

Vilsmeier–Haack reaction of indole 4.6. Modified dinitrostyrene Batcho–Leimgruber

conditions were planned for the formation of the indole from styrene 4.7, prepared in

several precedented steps from vanillin (4.8).

4.3: Synthetic Approach to Sporidesmin A

4.3.1: Preparation of Dinitrostyrene Derivative

Nitration of O-acetyl vanillin (4.9) afforded 4.10 when fuming nitric acid (>90%)

was used as the nitronium source (Scheme 4.2). Fuming nitric acid was originally

prepared fresh by distillation from a slurry of potassium nitrate and concentrated sulfuric

acid, but this procedure seemed quite dangerous on scales producing more than 50 mL of

nitric acid, prompting us to obtain the reagent from a commercial source.

Scheme 4.2. Nitration of vanillin.

Following nitration, 4.10 was heated to reflux in aqueous KOH to afford phenol

4.11, then O-methylation with dimethyl sulfate gave 4.12 in 99% yield (Scheme 4.3).

The Henry reaction was used to prepare dinitrostyrene derivative 4.13 through addition of

nitromethane to the aldehyde and subsequent elimination of water. Indole 4.14 was then

prepared in low yield using modified Batcho–Leimgruber conditions.147

Originally, we had envisioned preparing chloroindole 4.6 from indole 4.14.

However, it seemed more prudent to chlorinate vanillin, as we would be less concerned

with yield and could minimize the amount of chlorine gas used. Accordingly, 5-Cl-

CHO

OMeHO

Ac2O, Et3NDMAPTHF

CHO

OMeAcO

conditionsCHO

OMeAcO NO2

conditionsHNO3 (70%)H2SO4, HNO3HNO3 (>90%)

NRNR47%

4.8 4.9 4.1090%

yield

79

vanillin (4.15) was prepared by bubbling chlorine gas through a solution of vanillin in

acetic acid (Scheme 4.4).148 Nitration, deacetylation, and methylation proceeded in good

yield to provide aldehyde 4.18. The Henry reaction gave dinitrostyrene 4.7 in low

yield.149

Scheme 4.3. Dimethoxyindole synthesis.

Scheme 4.4. Synthesis of 5-chloro-dinitrostyrene derivative.

We determined that the small amount of ethyl acetal 4.19 formed during the

methylation reaction was detrimental to the yield of the Henry reaction (Scheme 4.5). To

circumvent this problem, the crude mixture of 4.18 and 4.19 was treated with TFA in

chloroform to cleave the acetal, affording pure 4.18. Use of clean material greatly

increased the ease of recrystallization and yield of the product of the Henry reaction (4.7).

CHO

OMeAcO NO2

KOHCHO

OMeHO NO2

Me2SO4, NaOHCHO

OMeMeO NO2

MeNO2, NaOH

OMeMeO NO2

NO2Fe, SiO2, AcOHPhCH3, reflux

OMeMeO N

H

H2O, reflux H2O, EtOH

H2O, MeOH

4.10 4.11 4.12

4.13 4.14

47% (2 steps) 99%

21% 7%

CHO

OMeHO

1. Cl2, AcOH2. Ac2O, Et3N DMAP, THF

CHO

OMeAcO

HNO3 (>90%)Cl

CHO

OMeHO

Cl

NO2OMe

MeO

Cl

NO2

NO2CHO

OMeMeO

Cl

NO2

CHO

OMeAcO

Cl

NO2

KOHH2O, reflux

Me2SO4, NaOHH2O, EtOH

MeNO2, NaOHH2O, MeOH

<10%

4.8 4.15 4.16

4.17 4.18 4.7

64%76% (2 steps)

>99%

80

Scheme 4.5. Undesired acetal formation.

4.3.2: Indole Synthesis and Sharpless Asymmetric Aminohydroxylation

Synthesis of indole 4.6 proceeded in good yield using modified Batcho–

Leimgruber conditions (Scheme 4.6). Following N-methylation, an aryl aldehyde was

installed at C3 via the Vilsmeier–Haack reaction.150 This aldehyde (4.20) was

immediately converted to the unsaturated ester (4.5) upon Wittig reaction with the

requisite phosphonium ylide. Sharpless asymmetric aminohydroxylation gave β-

hydroxytryptophan derivative 4.4, albeit in modest yield.151

Scheme 4.6. Asymmetric aminohydroxylation.

While it is commercially available, we opted to prepare the ligand for the

Sharpless reaction as described in the literature (Scheme 4.7).152 Acylation of

CHO

OMeHO

Cl

NO2

OMeMeO

Cl

NO2

NO2

CHO

OMeMeO

Cl

NO2

Me2SO4, NaOHH2O, EtOH

MeNO2, NaOHH2O, MeOH

+

OMeMeO

Cl

NO2

OEt

OEt

TFACHCl3

CHO

OMeMeO

Cl

NO2

64%

4.17 4.18 4.19

4.18 4.7

>99%

Fe, SiO2, AcOHPhCH3, reflux

OMeMeO

1. MeI

Ph3P=CHCO2Me

Cl

NH

OMeMeO

Cl

NMe

CO2Me K2OsO2(OH)4,(DHQD)2AQN (4.24),

CbzNClNa

OMeMeO

Cl

NMe

CO2MeHONHCbz

n-PrOH, H2O

91%

35%

OMeMeO

Cl

NMe

CHO

OMeMeO

Cl

NO2

NO2

2. POCl3, DMF; NaOH, H2O

PhCH3, reflux

4.7 4.6 4.20

4.5 4.4

93%

80%

81

difluorobenzene gave dione 4.23, which was alkylated to form the dihydroquinidinyl

anthraquinone ligand (4.24) in good yield.

Scheme 4.7. Preparation of (DHQD)2AQN.

4.3.3: Dioxopiperazine Formation

β-hydroxytryptophan derivative 4.4 was protected as the silyl ether (4.25) and the

Cbz group cleaved to reveal the free amine (4.26, Scheme 4.8). EDCI-mediated coupling

of N-Boc-Ala-OH provided dipeptide 4.27 in 59% yield.

Scheme 4.8. Formation of the dioxopiperazine and an undesired elimination.

Attempts to form the desired dioxopiperazine directly from 4.27 using the

microwave conditions described in the previous chapter resulted in decomposition.

O

O

O 1. AlCl3, reflux, 48 h

F

F

+2. polyphosphoric acid, 140°C

O

O F

F

DHQD, n-BuLiTHF

O

O DHQD

DHQD(DHQD)2AQN (4.24)4.21 4.22 4.23

14%78%

NMe

Cl

MeOOMe

HNNH

O

OMe

H2, Pd/C

EDCI, iPr2NEt,N-Boc-Ala-OH

NMe

Cl

MeOOMe

HNNHBoc

CO2Me

OMe

TBSO

2-OH-pyridine

OMeMeO

Cl

NMe

CO2MeHONHCbz

OMeMeO

Cl

NMe

CO2MeTBSONHCbzTBSCl, imid

OMeMeO

Cl

NMe

CO2MeTBSONH2

NMe

Cl

MeOOMe

HNNH2

CO2Me

OMe

TBSOTMSOTf,

2,6-lutadineCH2Cl2

PhCH3, reflux

4.4 4.25 4.26

4.27 4.28

4.29

DMF>99%

MeOH94%

CH2Cl259% 59%

61%

82

Following more traditional methods, we first removed the Boc group, then heated the

crude product (4.28) in toluene in the presence of 2-hydroxypyridine. The

dioxopiperazine was formed, but unfortunately the conditions also caused elimination of

the silyl ether to afford 4.29.

An alternative cyclization precursor was synthesized from 4.25 by saponification

of the methyl ester and coupling of L-Ala-OMe (Scheme 4.9). The low yielding coupling

step forced a quick screen of coupling reagents, through which T3P was identified as the

most efficient of those tested, providing dipeptide 4.31 in a mere 34% yield.

Scheme 4.9. Preparation of dipeptide.

The unoptimized reaction allowed us access to enough material to attempt the

cyclization reaction. Dipeptide 4.31 was converted to the free amine and heated gently in

toluene with catalytic 2-hydroxypyridine (Scheme 4.10). A mixture of the elimination

product (4.29) and the desired dioxopiperazine (4.3) was obtained in poor yield.

Scheme 4.10. Cyclization to the dioxopiperazine.

OMeMeO

Cl

NMe

CO2MeTBSONHCbz

NMe

Cl

MeOOMe

NHCbz

TBSO O

NH

CO2Me

Me

OMeMeO

Cl

NMe

CO2HTBSONHCbzLiOH

H2O, THF

L-Ala-OMe,iPr2NEt,

conditions

conditionsEDCIHATUT3P

6%29%34%

4.25 4.30 4.31

CH2Cl287%

OP O P

OPO

OO

!T3P

NMe

Cl

MeOOMe

NHCbz

TBSO O

NH

CO2Me

Me

NMe

Cl

MeOOMe

HNNH

O

OMe2. 2-OH-pyridine

PhCH3, 50 °C NMe

Cl

MeOOMe

HNNH

O

OMe

TBSO

1. H2, Pd/C +

22%4.31 4.29 4.3

83

We returned to amine 4.26 and to it coupled N-Fmoc-L-Ala (Scheme 4.11).

Again, our standard EDCI-mediated coupling produced dipeptide 4.32 in very poor yield.

Use of T3P nearly doubled the yield and greatly simplified purification of 4.32. Gentle

heating of the dipeptide in THF and morpholine resulted in concomitant Fmoc

deprotection and cyclization to the desired dioxopiperazine (4.3).

Scheme 4.11. Improved synthesis of dioxopiperazine 4.3.

4.3.4: Oxidative Cyclization Attempts

Elated to finally have access to dioxopiperazine 4.3, we pressed forward and

attempted the oxidative cyclization to the pyrroloindoline. Treatment of 4.3 with NBS or

bromine resulted in products containing two bromine atoms. We turned to the conditions

reported in Kishi’s synthesis, but only isolated starting material from the reaction mixture

(Scheme 4.12).113 Alcohol 4.34 was expected to result from aqueous conditions, but only

a small amount of 4.35, formed by oxidative fission of the indole double bond and

elimination of TBSOH, was recovered with starting material.153

N-Fmoc-L-Ala,iPr2NEt, x

NMe

Cl

MeOOMe

HNNHFmoc

CO2Me

OMe

TBSO

OMeMeO

Cl

NMe

CO2MeTBSONH2

CH2Cl2x=EDCI, 30%x=T3P, 59%

NMe

Cl

MeOOMe

HNNH

O

OMe

TBSO

morpholineTHF, 35 °C

4.26 4.32 4.3

78%

84

Scheme 4.12. First attempts at oxidative cyclization.

Dioxopiperazine 4.3 was precious material at this point in the synthesis, and we

certainly did not have large enough quantities to attempt a variety of conditions.

Therefore, simple dioxopiperazine 4.36 was prepared and subjected numerous times to

variations of the above conditions (Scheme 4.13). Unfortunately, model compound 4.36

also failed to react. Dioxopiperazine 4.36 is nearly insoluble in most solvents, so we were

never able to determine if this was the true cause of the failure. The unique electronics of

dioxopiperazine 4.3 and the poor solubility of any readily accessible surrogate forced us

to study the cyclization using advanced material.

Scheme 4.13. Model cyclization study.

We next attempted the oxidative cyclization using DMDO in acetone and

dichloromethane (Scheme 4.14).154,155 No product (4.34 or 4.39) was detected from the

NMe

Cl

MeOOMe

HN

TBSONH

O

OMe

NMe

Cl

MeOOMe

NH

AcOTBSO

NH

O

OMe

PhI(OAc)2

Me2S, MeCN

NMe

Cl

MeOOMe

HN

TBSONH

O

OMe

NMe

Cl

MeOOMe

NH

HOTBSO

NH

O

OMe

PhI(OAc)2

H2O, MeCNNHAc

Cl

MeOOMe

O

NH

HNO Me

O

4.3

4.3

4.33

4.344.35

NH

HNNH

O

O NH

NH

AcO NH

O

O

PhI(OAc)2

Me2S, MeCN

NH

HNNH

O

O NH

NH

HO NH

O

O

PhI(OAc)2

H2O, MeCN

OTBS OTBS

OTBS OTBS

4.36 4.37

4.36 4.38

85

reaction of either dioxopiperazine 4.3 or amine 4.26. However, trace amounts of 3-

hydroxypyrroloindoline 4.40 were detected from the reaction with Cbz protected

derivative 4.25.

Scheme 4.14. More oxidative cyclization attempts.

Interestingly, only the TBS-protected derivative (4.25) would cyclize. The

unprotected analogue (4.4) also failed to react when treated with DMDO to form the

desired tricycle (4.41, Scheme 4.15). Unfortunately, even the successful cyclization (4.25

to 4.40) had limited potential, as the best yield obtained for this sequence was 9%,

obtained from the crude reaction mixture by a difficult separation. The recovered alcohol

needed to be protected before moving forward, but only trace amounts of the bis-TBS

product were obtained. Steric congestion of the two neighboring silyl groups likely

contributed to the poor yield of 4.42.

NMe

Cl

MeOOMe

HN

TBSONH

O

OMe

NMe

Cl

MeOOMe

NH

HOTBSO

NH

O

OMe

DMDOCH2Cl2/acetone

NMe

Cl

MeOOMe

TBSO

NMe

Cl

MeOOMe

NHH

HOTBSO

CO2MeDMDO

CH2Cl2/acetone

CO2Me

NH2

NMe

Cl

MeOOMe

TBSO

NMe

Cl

MeOOMe

NCbzH

HOTBSO

CO2MeDMDO

CH2Cl2/acetone

CO2Me

NHCbz

4.3 4.34

4.26 4.39

4.25 4.40

86

Scheme 4.15. Progression of cyclization.

We also attempted the cyclization using other oxidants, including Davis’s

oxaziridine and benzoyl peroxide (Scheme 4.16).116,156 Only starting material was

recovered, with no trace of the desired products (4.34 and 4.43).

Scheme 4.16. Attempted cyclizations.

4.3.5: Toward a Formal Synthesis of Sporidesmin A

These repeated attempts at forming the requisite 3-hydroxypyrroloindoline were

frustrating, especially considering that Kishi had accomplished the same reaction using a

NMe

Cl

MeOOMe

NCbzH

HOHO

CO2Me

DMDOCH2Cl2/acetone

OMeMeO

Cl

NMe

CO2MeHONHCbz

TBSCl, imid

OMeMeO

Cl

NMe

CO2MeTBSONHCbz

>99%

NMe

Cl

MeOOMe

NCbzH

HOTBSO

CO2Me

DMDOCH2Cl2/acetone 9%

TBSCl, imidNMe

Cl

MeOOMe

NCbzH

TBSOTBSO

CO2Me

4.4 4.25

4.41 4.40 4.42

trace

NMe

Cl

MeOOMe

HN

TBSONH

O

OMe

NMe

Cl

MeOOMe

NH

HOTBSO

NH

O

OMe

Davis's oxaziridineCH2Cl2

NMe

Cl

MeOOMe

HN

TBSONH

O

OMe

NMe

Cl

MeOOMe

NH

PhOCOTBSO

NH

O

OMe

(PhCO2)2

DME

4.3 4.34

4.3 4.43

87

similar substrate. That said, it should be noted that in the original sporidesmin synthesis,

the authors only recovered diacetate 2.6 in 30% yield and completed the synthesis using

authentic diacetate prepared from the degradation of sporidesmin A (Scheme 4.17).113

One can only conclude that they also were unable to access appreciable quantities of the

pyrroloindoline. Moreover, in Kishi’s later synthesis of sporidesmin B, benzoyl peroxide

was used as the oxidant instead of iodosobenzene diacetate, yielding only 20% of the

desired oxidative cyclization product.116 We recognized that intermediate 2.5 reported in

the Kishi sporidesmin A synthesis could be intercepted from alcohol 4.4.

Scheme 4.17. Kishi’s final steps in the sporidesmin A synthesis.

A suitable N-Me alanine derivative needed to be prepared for the formal synthesis

of sporidesmin A. N-Fmoc-Ala-OH (4.44) was converted to the oxazolidinone (4.45),

then reduced to afford N-Me,Fmoc-Ala-OH (4.46, Scheme 4.18).157

Scheme 4.18. Preparation of N-Me,Fmoc-Ala-OH.

Alcohol 4.4 was acetylated by standard procedures to give 4.47. Amine 4.48 was

revealed following deprotection of the Cbz group, then coupled to N-Me alanine

NMe

Cl

MeOOMe

HNNMe

O

OMe

SS Ar

AcO

30% NMe

Cl

MeOOMe

N

25%

1. NaOH, MeOH2. mCPBA

H

AcOAcO

3. BF3!Et2O

(±)-sporidesmin A (1.11)

2.5 2.6

NMe

O

OMe

SS Ar

NMe

Cl

MeOOMe

NH

HOHO

NMe

O

OMe

SS

PhI(OAc)2

NHFmoc

Me CO2H

(HCHO)n, p-TsOH

PhCH3, reflux

CF3CO2H, Et3SiHOFmocN

MeO CHCl3

NMeFmoc

Me CO2H4.44 4.45 4.4655% (2 steps)

88

derivative 4.46 to afford dipeptide 4.49. This substrate, however, did not cyclize to the

dioxopiperazine when heated in morpholine and THF. Only 4.50, the product resulting

from Fmoc-deprotection and elimination of acetic acid, was recovered.

Scheme 4.19. Incorporation of N-Me-Ala.

4.4: Future Direction

Given more time, I would like to prepare bis(N,N’-Cbz) tryptophan derivatives

4.51 and 4.53 from indole 4.6 (Scheme 4.20). The bromocyclizations that were carried

out for the synthesis of chetomin were all performed on tryptophan derivatives bearing an

electron-withdrawing group on the indole nitrogen. Cyclization to the pyrroloindoline

should be more favorable with these substrates than for the N-Me derivatives. Experience

thus far with the oxidative cyclization reaction shows that a complex mixture of products

will likely be recovered from each reaction, so sufficient starting material will be required

to obtain adequate characterization of the reaction products.

OMeMeO

Cl

NMe

CO2MeHONHCbz

N-Fmoc,Me-Ala-OH,iPr2NEt, T3P morpholine

NMe

Cl

MeOOMe

HN

AcO

NMeFmocCO2Me

OMe THF N

Me

Cl

MeOOMe

HNNHMe

CO2Me

OMe

OMeMeO

Cl

NMe

CO2MeAcONHCbzAc2O H2, Pd/C

OMeMeO

Cl

NMe

CO2MeAcONH2

4.4 4.47 4.48

4.49 4.50

56%70%

MeOH

CH2Cl218%

89

Scheme 4.20. Full panel of oxidation conditions to screen.

The proposed installation of the epidithiodioxopiperazine ring may also be

problematic, especially considering the attempts made in the chetomin synthesis. Still, we

propose forming (–)-sporidesmin A (1.11) through oxidation of the disulfide prepared by

reaction of hemiaminal 4.1 with hydrogen sulfide (Scheme 4.21). The hemiaminal could

be prepared from dioxopiperazine 4.2 or a related derivative.

Scheme 4.21. Proposed epidithiodioxopiperazine formation.

4.5: Concluding Remarks

In summary, we have developed an asymmetric approach toward the synthesis of

(–)-sporidesmin A. Advanced dioxopiperazine 4.3 can be prepared in decent overall yield

and is theoretically three to four steps away from the target natural product. Moreover,

OMeMeO

Cl

NCbz

CO2MeTBSONHCbz

NCbz

Cl

MeOOMe

NCbzH

ROTBSO

CO2Me

NCbz

Cl

MeOOMe

HN

TBSONH

O

OMe

NCbz

Cl

MeOOMe

NH

ROTBSO

NH

O

OMe

conditions

conditions

conditions

a. PhI(OAc)2, Me2Sb. PhI(OAc)2, H2Oc. DMDOd. Davis's oxaziridinee. (PhCO2)2f. N-PSP; mCPBAg. Br2; AgCN, H2Oh. O2, methylene blue, sunlamp; Me2S

AcHHHCOPhHHH

R

4.51 4.52

4.53 4.54

NMe

Cl

MeOOMe

NH

TBSOTBSO

NMe

O

OMe

(!)-sporidesmin A (1.11)

NMe

Cl

MeOOMe

NH

HOHO

NMe

O

OMe

SS

NMe

Cl

MeOOMe

NH

TBSOTBSO

NMe

O

O

Me

HO

OH

thiolation oxidation

4.1 4.2

90

many of the synthetic advances made over the course of this synthesis were applied in

our approach to chetomin. Particularly, use of N-Fmoc amino acids as precursors to

dioxopiperazine rings eliminated a deptrotection step from each synthesis while

improving the cyclization yields. While it is difficult to leave this project behind

unfinished, I have no doubt that capable hands will pick it up in the future to achieve the

total synthesis of (–)-sporidesmin A.

91

REFERENCES

1. Cook, K. M.; Hilton, S. T.; Mecinovic, J.; Motherwell, W. B.; Figg, W. D.; Schofield, C. J. Epidithiodiketopiperazines Block the Interaction between Hypoxia-Inducible Factor-1alpha (Hif-1alpha) and P300 by a Zinc Ejection Mechanism. J. Biol. Chem. 2009, 284, 26831-26838.

2. Gardiner, D. M.; Waring, P.; Howlett, B. J. The Epipolythiodioxopiperazine (Etp) Class of Fungal Toxins: Distribution, Mode of Action, Functions and Biosynthesis. Microbiology 2005, 151, 1021-1032.

3. Waring, P.; Beaver, J. Gliotoxin and Related Epipolythiodioxopiperazines. Gen. Pharmacol. 1996, 27, 1311-1316.

4. Waring, P.; Eichner, R. D.; Mullbacher, A. The Chemistry and Biology of the Immunomodulating Agent Gliotoxin and Related Epipolythiodioxopiperazines. Med. Res. Rev. 1988, 8, 499-524.

5. Iwasa, E.; Hamashima, Y.; Sodeoka, M. Epipolythiodiketopiperazine Alkaloids: Total Syntheses and Biological Activities. Isr. J. Chem. 2011, 51, 420-433.

6. Weindling, R.; Emerson, O. H. The Isolation of a Toxic Substance from the Culture Filtrate of Trichoderma. Phytopathology 1936, 26, 1068-1070.

7. Bell, M. R.; Johnson, J. R.; Wildi, B. S.; Woodward, R. B. The Structure of Gliotoxin. J. Am. Chem. Soc. 1958, 80, 1001-1001.

8. Beecham, A. F.; Fridrichsons, J.; Mathieson, A. M. Structure and Absolute Configuration of Gliotoxin and Absolute Configuration of Sporidesmin. Tetrahedron Lett. 1966, 3131.

9. Jordan, T. W.; Cordiner, S. J. Fungal Epipolythiodioxopiperazine Toxins Have Therapeutic Potential and Roles in Disease. Trends Pharmacol. Sci. 1987, 8, 144-149.

10. Kwon-Chung, K. J.; Sugui, J. A. What Do We Know About the Role of Gliotoxin in the Pathobiology of Aspergillus Fumigatus? Med. Mycol. 2009, 47 Suppl 1, S97-103.

11. Mullbacher, A.; Eichner, R. D. Immunosuppression Invitro by a Metabolite of a Human Pathogenic Fungus. Proc. Natl. Acad. Sci., Biol. 1984, 81, 3835-3837.

12. Mullbacher, A.; Waring, P.; Eichner, R. D. Identification of an Agent in Cultures of Aspergillus-Fumigatus Displaying Anti-Phagocytic and Immunomodulating Activity Invitro. J. Gen. Microbiol. 1985, 131, 1251-1258.

92

13. Mullbacher, A.; Moreland, A. F.; Waring, P.; Sjaarda, A.; Eichner, R. D. Prevention of Graft-Versus-Host Disease by Treatment of Bone-Marrow with Gliotoxin in Fully Allogeneic Chimeras and Their Cyto-Toxic T-Cell Repertoire. Transplantation 1988, 46, 120-125.

14. Munday, R. Studies on the Mechanism of Toxicity of the Mycotoxin, Sporidesmin I. Generation of Superoxide Radical by Sporidesmin. Chem.-Biol. Interact. 1982, 41, 361-374.

15. Chai, C. L.; Waring, P. Redox Sensitive Epidithiodioxopiperazines in Biological Mechanisms of Toxicity. Redox Rep. 2000, 5, 257-264.

16. Eichner, R. D.; Waring, P.; Geue, A. M.; Braithwaite, A. W.; Mullbacher, A. Gliotoxin Causes Oxidative Damage to Plasmid and Cellular DNA. J. Biol. Chem. 1988, 263, 3772-3777.

17. Cordiner, S. J.; Jordan, T. W. Inhibition by Sporidesmin of Hepatocyte Bile Acid Transport. Biochem. J. 1983, 212, 197-204.

18. Mason, J. W.; Kidd, J. G. Effects of Gliotoxin and Other Sulfur-Containing Compounds on Tumor Cells in Vitro; with Observations on the Mechanism of Action of Gliotoxin. J. Immunol. 1951, 66, 99-106.

19. Hurne, A. M.; Chai, C. L.; Waring, P. Inactivation of Rabbit Muscle Creatine Kinase by Reversible Formation of an Internal Disulfide Bond Induced by the Fungal Toxin Gliotoxin. J. Biol. Chem. 2000, 275, 25202-25206.

20. Trown, P. W.; Bilello, J. A. Mechanism of Action of Gliotoxin: Elimination of Activity by Sulfhydryl Compounds. Antimicrob. Agents Chemother. 1972, 2, 261-266.

21. Mullbacher, A.; Waring, P.; Tiwari-Palni, U.; Eichner, R. D. Structural Relationship of Epipolythiodioxopiperazines and Their Immunomodulating Activity. Mol. Immunol. 1986, 23, 231-235.

22. Stillwell, M. A.; Magasi, L. P.; Strunz, G. M. Production, Isolation, and Antimicrobial Activity of Hyalodendrin, a New Antibiotic Produced by a Species of Hyalodendron. Can. J. Microbiol. 1974, 20, 759-764.

23. Strunz, G. M.; Heissner, C. J.; Kakushima, M.; Stillwell, M. A. Metabolites of Hyalodendron Sp - Bisdethiodi(Methylthio)Hyalodendrin. Can. J. Chem. 1974, 52, 325-326.

24. Strunz, G. M.; Kakushima, M.; Stillwell, M. A. Epitetrathiodioxopiperazine with 3s,6s Configuration from Hyalodendron Sp. Can. J. Chem. 1975, 53, 295-297.

25. DeVault, R. L.; Rosenbrook, W., Jr. A Novel Class of Diketopiperazines. J. Antibiot. 1973, 26, 532-534.

93

26. Dorn, F.; Arigoni, D. Gliovictin, New Metabolite of Helminthosporium-Victoriae. Experientia 1974, 30, 134-135.

27. Michel, K. H.; Chaney, M. O.; Jones, N. D.; Hoehn, M. M.; Nagarajan, R. Epipolythiopiperazinedione Antibiotics from Penicillium Turbatum. J. Antibiot. 1974, 27, 57-64.

28. Shin, J. H.; Fenical, W. Isolation of Gliovictin from the Marine Deuteromycete Asteromyces-Cruciatus. Phytochemistry 1987, 26, 3347.

29. Kawahara, N.; Nozawa, K.; Nakajima, S.; Kawai, K. Studies on Fungal Products 13. Isolation and Structures of Dithiosilvatin and Silvathione, Novel Dioxopiperazine Derivatives from Aspergillus-Silvaticus. J. Chem. Soc., Perkin Trans. 1 1987, 2099-2101.

30. Hanson, J. R.; Oleary, M. A. New Piperazinedione Metabolites of Gliocladium-Deliquescens. J. Chem. Soc., Perkin Trans. 1 1981, 218-220.

31. Nagarajan, R.; Huckstep, L. L.; Lively, D. H.; Delong, D. C.; Marsh, M. M.; Neuss, N. Aranotin and Related Metabolites from Arachniotus Aureus I. Determination of Structure. J. Am. Chem. Soc. 1968, 90, 2980.

32. Nagarajan, R.; Neuss, N.; Marsh, M. M. Aranotin and Related Metabolites 3. Configuration and Conformation of Acetylaranotin. J. Am. Chem. Soc. 1968, 90, 6518.

33. Neuss, N.; Boeck, L. D.; Brannon, D. R.; Cline, J. C.; DeLong, D. C.; Gorman, M.; Huckstep, L. L.; Lively, D. H.; Mabe, J.; Marsh, M. M.; Molloy, B. B.; Nagarajan, R.; Nelson, J. D.; Stark, W. M. Aranotin and Related Metabolites from Arachniotus Aureus (Eidam) Schroeter Iv. Fermentation, Isolation, Structure Elucidation, Biosynthesis, and Antiviral Properties. Antimicrob. Agents Chemother. 1968, 8, 213-219.

34. Neuss, N.; Nagarajan, R.; Molloy, B. B.; Huckstep, L. L. Aranotin and Related Metabolites 2. Isolation Characterization and Structures of 2 New Metabolites. Tetrahedron Lett. 1968, 4467.

35. Trown, P. W.; Lindh, H. F.; Milstrey, K. P.; Gallo, V. M.; Mayberry, B. R.; Lindsay, H. L.; Miller, P. A. Ll-S88-Alpha, an Antiviral Substance Produced by Aspergillus Terreus. Antimicrob. Agents Chemother. 1968, 8, 225-228.

36. Cosulich, D. B.; Nelson, N. R.; Van den Hende, J. H. Crystal and Molecular Structure of L-S88alpha, an Antiviral Epidithiapiperazinedione Derivative from Aspergillus Terreus. J. Am. Chem. Soc. 1968, 90, 6519.

37. Murdock, K. C. Antiviral Agents. Chemical Modifications of a Disulfide Antibiotic, Acetylaranotin. J. Med. Chem. 1974, 17, 827-835.

94

38. Kawahara, N.; Nakajima, S.; Yamazaki, M.; Kawai, K. Structure of a Novel Epidithiodioxopiperazine, Emethallicin a, a Potent Inhibitor of Histamine Release from Emericella Heterothallica. Chem. Pharm. Bull. 1989, 37, 2592-2595.

39. Kawahara, N.; Nozawa, K.; Yamazaki, M.; Nakajima, S.; Kawai, K. Structures of Novel Epipolythiodioxopiperazines, Emethallicins B, C, and D, Potent Inhibitors of Histamine Release, from Emericella Heterothallica. Chem. Pharm. Bull. 1990, 38, 73-78.

40. Kawahara, N.; Nozawa, K.; Nakajima, S.; Kawai, K.; Yamazaki, M. Novel Epitetrathiodioxopiperazines, Emethallicin-B and Emethallicin-C, as Potent Inhibitors of Compound 48/80-Induced Histamine-Release, from Emericella-Heterothallica. J. Chem. Soc., Chem. Commun. 1989, 951-952.

41. Kawahara, N.; Nozawa, K.; Yamazaki, M.; Nakajima, S.; Kawai, K. Studies on Fungal Products 32. Novel Epidithiodioxopiperazines, Emethallicin-E and Emethallicin-F, from Emericella-Heterothallica. Heterocycles 1990, 30, 507-515.

42. Waring, P.; Eichner, R. D.; Tiwaripalni, U.; Mullbacher, A. Gliotoxin-E - a New Biologically-Active Epipolythiodioxopiperazine Isolated from Penicillium-Terlikowskii. Aust. J. Chem. 1987, 40, 991-997.

43. Ueno, Y.; Umemori, K.; Niimi, E.; Tanuma, S.; Nagata, S.; Sugamata, M.; Ihara, T.; Sekijima, M.; Kawai, K.; Ueno, I.; et al. Induction of Apoptosis by T-2 Toxin and Other Natural Toxins in Hl-60 Human Promyelotic Leukemia Cells. Nat. Toxins 1995, 3, 129-137.

44. Seya, H.; Nozawa, K.; Nakajima, S.; Kawai, K. I.; Udagawa, S. I. Studies on Fungal Products 8. Isolation and Structure of Emestrin, a Novel Antifungal Macrocyclic Epidithiodioxopiperazine from Emericella-Striata - X-Ray Molecular-Structure of Emestrin. J. Chem. Soc., Perkin Trans. 1 1986, 109-116.

45. Seya, H.; Nakajima, S.; Kawai, K.; Udagawa, S. Structure and Absolute-Configuration of Emestrin, a New Macrocyclic Epidithiodioxopiperazine from Emericella-Striata. J. Chem. Soc., Chem. Commun. 1985, 657-658.

46. Kawai, K.; Nozawa, K.; Seya, H.; Kawahara, N.; Udagawa, S.; Nakajima, S. Studies on Fungal Products 12. Structure of Aurantioemestrin from Emericella-Striata. Heterocycles 1987, 26, 475-479.

47. Kawahara, N.; Nozawa, K.; Nakajima, S.; Kawai, K. Aurantioemestrin from Emericella-Striata and Silvathione from Aspergillus-Silvaticus, Possible Key Intermediates from Epidithiodioxopiperazines to Trioxopiperazines. J. Chem. Soc., Chem. Commun. 1986, 1495-1496.

48. Seya, H.; Nozawa, K.; Udagawa, S.; Nakajima, S.; Kawai, K. Studies on Fungal Products 9. Dethiosecoemestrin, a New Metabolite Related to Emestrin, from Emericella-Striata. Chem. Pharm. Bull. 1986, 34, 2411-2416.

95

49. Nozawa, K.; Udagawa, S. I.; Nakajima, S.; Kawai, K. I. Studies on Fungal Products 14. Emestrin-B, a New Epitrithiodioxopiperazine, from Emericella-Striata. Chem. Pharm. Bull. 1987, 35, 3460-3463.

50. Onodera, H.; Hasegawa, A.; Tsumagari, N.; Nakai, R.; Ogawa, T.; Kanda, Y. Mpc1001 and Its Analogues: New Antitumor Agents from the Fungus Cladorrhinum Species. Org. Lett. 2004, 6, 4101-4104.

51. Deffieux, G.; Gadret, M.; Leger, J. M.; Carpy, A. Crystal Structure of Original Fungal Metabolite of 3,6-Epidithio-2,5-Dioxopiperazine Group - Epicorazine-A. Acta Crystallogr. B. 1977, 33, 1474-1478.

52. Deffieux, G.; Baute, M. A.; Baute, R.; Filleau, M. J. New Antibiotics from Fungus Epicoccum-Nigrum 2. Epicorazine-a - Structure Elucidation and Absolute Configuration. J. Antibiot. 1978, 31, 1102-1105.

53. Deffieux, G.; Filleau, M. J.; Baute, R. New Antibiotics from Fungus Epicoccum-Nigrum 3. Epicorazine-B - Structure Elucidation and Absolute Configuration. J. Antibiot. 1978, 31, 1106-1109.

54. Brown, A. E.; Finlay, R.; Ward, J. S. Antifungal Compounds Produced by Epicoccum-Purpurascens against Soil-Borne Plant Pathogenic Fungi. Soil Biol. Biochem. 1987, 19, 657-664.

55. Kleinwachter, P.; Dahse, H. M.; Luhmann, U.; Schlegel, B.; Dornberger, K. Epicorazine C, an Antimicrobial Metabolite from Stereum Hirsutum Hki 0195. J. Antibiot. 2001, 54, 521-525.

56. Begg, W. R.; Elix, J. A.; Jones, A. J. Nonacyclic Amides from Lichens of Genus Xanthoparmelia. Tetrahedron Lett. 1978, 1047-1050.

57. Ernst-Russell, M. A.; Chai, C. L. L.; Hurne, A. M.; Waring, P.; Hockless, D. C. R.; Elix, J. A. Structure Revision and Cytotoxic Activity of the Scabrosin Esters, Epidithiopiperazinediones from the Lichen Xanthoparmelia Scabrosa. Aust. J. Chem. 1999, 52, 279-283.

58. Moerman, K. L.; Chai, C. L. L.; Waring, P. Evidence That the Lichen-Derived Scabrosin Esters Target Mitochondrial Atp Synthase in P388d1 Cells. Toxicol. Appl. Pharmacol. 2003, 190, 232-240.

59. Curtis, P. J.; Greatbanks, D.; Hesp, B.; Cameron, A. F.; Freer, A. A. Sirodesmins a, B, C, and G, Antiviral Epipolythiopiperazine-2,5-Diones of Fungal Origin - X-Ray Analysis of Sirodesmin-a Diacetate. J. Chem. Soc., Perkin Trans. 1 1977, 180-189.

60. Ferezou, J. P.; Riche, C.; Quesneauthierry, A.; Pascardbilly, C.; Barbier, M.; Bousquet, J. F.; Boudart, G. Structures of 2 Toxins Isolated from Fungus Cultures

96

Phoma-Lingam Tode - Sirodesmin Pl and Deacetylsirodesmin Pl. Nouv. J. Chim. 1977, 1, 327-334.

61. Boudart, G. Antibacterial Activity of Sirodesmin Pl Phytotoxin: Application to the Selection of Phytotoxin-Deficient Mutants. Appl. Environ. Microbiol. 1989, 55, 1555-1559.

62. Elliott, C. E.; Gardiner, D. M.; Thomas, G.; Cozijnsen, A.; Van De Wouw, A.; Howlett, B. J. Production of the Toxin Sirodesmin Pl by Leptosphaeria Maculans During Infection of Brassica Napus. Mol. Plant Pathol. 2007, 8, 791-802.

63. Thornton, R. H.; Sinclair, D. P. Sporidesmium Bakeri and Facial Eczema of Sheep in the Field. Nature 1959, 184, 1327-1328.

64. Thornton, R. H.; Percival, J. C. A Hepatotoxin from Sporidesmium Bakeri Capable of Producing Facial Eczema Diseases in Sheep. Nature 1959, 183, 63.

65. Synge, R. L. M.; White, E. P. Sporidesmin - a Substance from Sporidesmium-Bakeri Causing Lesions Characteristic of Facial Eczema. Chem. Ind. 1959, 1546-1547.

66. Fridrichsons, J.; Mathieson, A. M. Structure of Methylene Dibromide Adduct of Sporidesmin at - 150 Degrees C. Acta Crystallogr. 1965, 18, 1043.

67. Fridrichsons, J.; Mathieson, A. M. The Structure of Sporidesmin - Causative Agent of Facial Eczema in Sheep. Tetrahedron Lett. 1962, 1265-1268.

68. Smith, B. L.; Embling, P. P.; Towers, N. R.; Wright, D. E.; Payne, E. The Protective Effect of Zinc Sulphate in Experimental Sporidesmin Poisoning of Sheep. New Zealand veterinary journal 1977, 25, 124-127.

69. Waring, P.; Egan, M.; Braithwaite, A.; Mullbacher, A.; Sjaarda, A. Apoptosis Induced in Macrophages and T Blasts by the Mycotoxin Sporidesmin and Protection by Zn2+ Salts. Int. J. Immunopharmacol. 1990, 12, 445-457.

70. Munday, R. Studies on the Mechanism of Toxicity of the Mycotoxin Sporidesmin 3. Inhibition by Metals of the Generation of Superoxide Radical by Sporidesmin. J. Appl. Toxicol. 1984, 4, 182-186.

71. Hodges, R.; Shannon, J. S. Isolation and Structure of Sporidesmin C. Aust. J. Chem. 1966, 19, 1059.

72. Waksman, S. A.; Bugie, E. Chaetomin, a New Antibiotic Substance Produced by Chaetomium Cochliodes I. Formation and Properties. J. Bacteriol. 1944, 48, 527-530.

73. Mcinnes, A. G.; Taylor, A.; Walter, J. A. Structure of Chetomin. J. Am. Chem. Soc. 1976, 98, 6741-6741.

97

74. Li, G. Y.; Li, B. G.; Yang, T.; Yan, J. F.; Liu, G. Y.; Zhang, G. L. Chaetocochins a-C, Epipolythiodioxopiperazines from Chaetomium Cochliodes. J. Nat. Prod. 2006, 69, 1374-1376.

75. Fujimoto, H.; Sumino, M.; Okuyama, E.; Ishibashi, M. Immunomodulatory Constituents from an Ascomycete, Chaetomium Seminudum. J. Nat. Prod. 2004, 67, 98-102.

76. Kung, A. L.; Zabludoff, S. D.; France, D. S.; Freedman, S. J.; Tanner, E. A.; Vieira, A.; Cornell-Kennon, S.; Lee, J.; Wang, B.; Wang, J.; Memmert, K.; Naegeli, H. U.; Petersen, F.; Eck, M. J.; Bair, K. W.; Wood, A. W.; Livingston, D. M. Small Molecule Blockade of Transcriptional Coactivation of the Hypoxia-Inducible Factor Pathway. Cancer Cell 2004, 6, 33-43.

77. Staab, A.; Loeffler, J.; Said, H. M.; Diehlmann, D.; Katzer, A.; Beyer, M.; Fleischer, M.; Schwab, F.; Baier, K.; Einsele, H.; Flentje, M.; Vordermark, D. Effects of Hif-1 Inhibition by Chetomin on Hypoxia-Related Transcription and Radiosensitivity in Ht 1080 Human Fibrosarcoma Cells. BMC Cancer 2007, 7, 213.

78. Mohammed, K. A.; Jadulco, R. C.; Bugni, T. S.; Harper, M. K.; Sturdy, M.; Ireland, C. M. Strongylophorines: Natural Product Inhibitors of Hypoxia-Inducible Factor-1 Transcriptional Pathway. J. Med. Chem. 2008, 51, 1402-1405.

79. Melillo, G. Hypoxia-Inducible Factor 1 Inhibitors. Methods Enzymol. 2007, 435, 385-402.

80. Block, K. M.; Wang, H.; Szabo, L. Z.; Polaske, N. W.; Henchey, L. K.; Dubey, R.; Kushal, S.; Laszlo, C. F.; Makhoul, J.; Song, Z.; Meuillet, E. J.; Olenyuk, B. Z. Direct Inhibition of Hypoxia-Inducible Transcription Factor Complex with Designed Dimeric Epidithiodiketopiperazine. J. Am. Chem. Soc. 2009, 131, 18078-18088.

81. Hauser, D.; Weber, H. P.; Sigg, H. P. Isolation and Configuration of Chaetocin. Helv. Chim. Acta 1970, 53, 1061-1073.

82. Weber, H. P. Molecular Structure and Absolute Configuration of Chaetocin. Acta Crystallogr. B. 1972, B 28, 2945.

83. Saito, T.; Suzuki, Y.; Koyama, K.; Natori, S.; Iitaka, Y.; Kinoshita, T. Chetracin-a and Chaetocin-B and Chaetocin-C, 3 New Epipolythiodioxopiperazines from Chaetomium Spp. Chem. Pharm. Bull. 1988, 36, 1942-1956.

84. Li, L.; Li, D.; Luan, Y.; Gu, Q.; Zhu, T. Cytotoxic Metabolites from the Antarctic Psychrophilic Fungus Oidiodendron Truncatum. J. Nat. Prod. 2012, 75, 920-927.

98

85. Greiner, D.; Bonaldi, T.; Eskeland, R.; Roemer, E.; Imhof, A. Identification of a Specific Inhibitor of the Histone Methyltransferase Su(Var)3-9. Nat. Chem. Biol. 2005, 1, 143-145.

86. Feng, Y.; Blunt, J. W.; Cole, A. L.; Munro, M. H. Novel Cytotoxic Thiodiketopiperazine Derivatives from a Tilachlidium Sp. J. Nat. Prod. 2004, 67, 2090-2092.

87. Minato, H.; Matsumoto, M.; Katayama, T. Studies on the Metabolites of Verticillium Sp. Structures of Verticillins a, B, and C. J. Chem. Soc., Perkin Trans. 1 1973, 17, 1819-1825.

88. Joshi, B. K.; Gloer, J. B.; Wicklow, D. T. New Verticillin and Glisoprenin Analogues from Gliocladium Catenulatum, a Mycoparasite of Aspergillus Flavus Sclerotia. J. Nat. Prod. 1999, 62, 730-733.

89. Chu, M.; Truumees, I.; Rothofsky, M. L.; Patel, M. G.; Gentile, F.; Das, P. R.; Puar, M. S.; Lin, S. L. Inhibition of C-Fos Proto-Oncogene Induction by Sch 52900 and Sch 52901, Novel Diketopiperazine Produced by Gliocladium Sp. J. Antibiot. 1995, 48, 1440-1445.

90. Dong, J. Y.; He, H. P.; Shen, Y. M.; Zhang, K. Q. Nematicidal Epipolysulfanyldioxopiperazines from Gliocladium Roseum. J. Nat. Prod. 2005, 68, 1510-1513.

91. Zheng, C. J.; Kim, C. J.; Bae, K. S.; Kim, Y. H.; Kim, W. G. Bionectins a-C, Epidithiodioxopiperazines with Anti-Mrsa Activity, from Bionectra Byssicola F120. J. Nat. Prod. 2006, 69, 1816-1819.

92. Takahashi, C.; Minoura, K.; Yamada, T.; Numata, A.; Kushida, K.; Shingu, T.; Hagishita, S.; Nakai, H.; Sato, T.; Harada, H. Potent Cytotoxic Metabolites from a Leptosphaeria Species - Structure Determination and Conformational-Analysis. Tetrahedron 1995, 51, 3483-3498.

93. Takahashi, C.; Numata, A.; Ito, Y.; Matsumura, E.; Araki, H.; Iwaki, H.; Kushida, K. Leptosins, Antitumor Metabolites of a Fungus Isolated from a Marine Alga. J. Chem. Soc., Perkin Trans. 1 1994, 1859-1864.

94. Takahashi, C.; Numata, A.; Matsumura, E.; Minoura, K.; Eto, H.; Shingu, T.; Ito, T.; Hasegawa, T. Leptosins I and J, Cytotoxic Substances Produced by a Leptosphaeria Sp. Physico-Chemical Properties and Structures. J. Antibiot. 1994, 47, 1242-1249.

95. Winstead, J. A.; Suhadolnik, R. J. Biosynthesis of Gliotoxin 2. Further Studies on the Incorporation of Carbon-14 and Tritium-Labeled Precursors. J. Am. Chem. Soc. 1960, 82, 1644-1647.

99

96. Suhadolnik, R. J.; Chenoweth, R. G. Biosynthesis of Gliotoxin 1. Incorporation of Phenylalanine-1-C-14 and Phenylalanine-2-C-14. J. Am. Chem. Soc. 1958, 80, 4391-4392.

97. Balibar, C. J.; Walsh, C. T. Glip, a Multimodular Nonribosomal Peptide Synthetase in Aspergillus Fumigatus, Makes the Diketopiperazine Scaffold of Gliotoxin. Biochemistry 2006, 45, 15029-15038.

98. Bulock, J. D.; Leigh, C. Biosynthesis of Gliotoxin. J. Chem. Soc., Chem. Commun. 1975, 628-629.

99. Kirby, G. W.; Patrick, G. L.; Robins, D. J. Cyclo-(L-Phenylalanyl-L-Seryl) as an Intermediate in Biosynthesis of Gliotoxin. J. Chem. Soc., Perkin Trans. 1 1978, 1336-1338.

100. Macdonald, J. C.; Slater, G. P. Biosynthesis of Gliotoxin and Mycelianamide. Can. J. Biochem. 1975, 53, 475-478.

101. Kirby, G. W.; Rao, G. V.; Robins, D. J. New Co-Metabolites of Gliotoxin in Gliocladium-Virens. J. Chem. Soc., Perkin Trans. 1 1988, 301-304.

102. Davis, C.; Carberry, S.; Schrettl, M.; Singh, I.; Stephens, J. C.; Barry, S. M.; Kavanagh, K.; Challis, G. L.; Brougham, D.; Doyle, S. The Role of Glutathione S-Transferase Glig in Gliotoxin Biosynthesis in Aspergillus Fumigatus. Chem. Biol. 2011, 18, 542-552.

103. Scharf, D. H.; Remme, N.; Habel, A.; Chankhamjon, P.; Scherlach, K.; Heinekamp, T.; Hortschansky, P.; Brakhage, A. A.; Hertweck, C. A Dedicated Glutathione S-Transferase Mediates Carbon-Sulfur Bond Formation in Gliotoxin Biosynthesis. J. Am. Chem. Soc. 2011, 133, 12322-12325.

104. Scharf, D. H.; Heinekamp, T.; Remme, N.; Hortschansky, P.; Brakhage, A. A.; Hertweck, C. Biosynthesis and Function of Gliotoxin in Aspergillus Fumigatus. Appl. Microbiol. Biotechnol. 2012, 93, 467-472.

105. Scharf, D. H.; Remme, N.; Heinekamp, T.; Hortschansky, P.; Brakhage, A. A.; Hertweck, C. Transannular Disulfide Formation in Gliotoxin Biosynthesis and Its Role in Self-Resistance of the Human Pathogen Aspergillus Fumigatus. J. Am. Chem. Soc. 2010, 132, 10136-10141.

106. Schrettl, M.; Carberry, S.; Kavanagh, K.; Haas, H.; Jones, G. W.; O'Brien, J.; Nolan, A.; Stephens, J.; Fenelon, O.; Doyle, S. Self-Protection against Gliotoxin--a Component of the Gliotoxin Biosynthetic Cluster, Glit, Completely Protects Aspergillus Fumigatus against Exogenous Gliotoxin. PLoS Pathog. 2010, 6, e1000952.

100

107. Gardiner, D. M.; Cozijnsen, A. J.; Wilson, L. M.; Pedras, M. S.; Howlett, B. J. The Sirodesmin Biosynthetic Gene Cluster of the Plant Pathogenic Fungus Leptosphaeria Maculans. Mol. Microbiol. 2004, 53, 1307-1318.

108. Gardiner, D. M.; Howlett, B. J. Bioinformatic and Expression Analysis of the Putative Gliotoxin Biosynthetic Gene Cluster of Aspergillus Fumigatus. FEMS Microbiol. Lett. 2005, 248, 241-248.

109. Fox, E. M.; Howlett, B. J. Biosynthetic Gene Clusters for Epipolythiodioxopiperazines in Filamentous Fungi. Mycol. Res. 2008, 112, 162-169.

110. Ferezou, J. P.; Quesneauthierry, A.; Barbier, M.; Kollmann, A.; Bousquet, J. F. Structure and Synthesis of Phomamide, a New Piperazine-2,5-Dione Related to the Sirodesmins, Isolated from the Culture-Medium of Phoma-Lingam Tode. J. Chem. Soc., Perkin Trans. 1 1980, 113-115.

111. Bulock, J. D.; Clough, L. E. Sirodesmin Biosynthesis. Aust. J. Chem. 1992, 45, 39-45.

112. Kremer, A.; Li, S. M. A Tyrosine O-Prenyltransferase Catalyses the First Pathway-Specific Step in the Biosynthesis of Sirodesmin Pl. Microbiology 2010, 156, 278-286.

113. Kishi, Y.; Nakatsuka, S.; Fukuyama, T.; Havel, M. Total Synthesis of Sporidesmin-A. J. Am. Chem. Soc. 1973, 95, 6493-6495.

114. Kishi, Y.; Fukuyama, T.; Nakatsuka, S. Total Synthesis of Dehydrogliotoxin. J. Am. Chem. Soc. 1973, 95, 6492-6493.

115. Kishi, Y.; Fukuyama, T.; Nakatsuka, S. New Method for Synthesis of Epidithiodiketopiperazines. J. Am. Chem. Soc. 1973, 95, 6490-6492.

116. Nakatsuka, S.; Fukuyama, T.; Kishi, Y. Total Synthesis of D,L-Sporidesmin B. Tetrahedron Lett. 1974, 1549-1552.

117. Fukuyama, T.; Kishi, Y. Total Synthesis of Gliotoxin. J. Am. Chem. Soc. 1976, 98, 6723-6724.

118. Fukuyama, T.; Nakatsuka, S.; Kishi, Y. Total Synthesis of Gliotoxin, Dehydrogliotoxin and Hyalodendrin. Tetrahedron 1981, 37, 2045-2078.

119. Strunz, G. M.; Kakushima, M. Total Synthesis of (+/-) Hyalodendrin. Experientia 1974, 30, 719-720.

120. Williams, R. M.; Rastetter, W. H. Syntheses of the Fungal Metabolites (+/-)-Gliovictin and (+/-)-Hyalodendrin. J. Org. Chem. 1980, 45, 2625-2631.

101

121. Williams, R. M.; Rastetter, W. H. Efficient Synthesis of D,L-Gliovictin - Construction of the Hydroxymethyl Moiety Via a 3-Formyl-2,5-Piperazinedione. Tetrahedron Lett. 1979, 1187-1190.

122. Kim, J.; Ashenhurst, J. A.; Movassaghi, M. Total Synthesis of (+)-11,11'-Dideoxyverticillin A. Science 2009, 324, 238-241.

123. Iwasa, E.; Hamashima, Y.; Fujishiro, S.; Higuchi, E.; Ito, A.; Yoshida, M.; Sodeoka, M. Total Synthesis of (+)-Chaetocin and Its Analogues: Their Histone Methyltransferase G9a Inhibitory Activity. J. Am. Chem. Soc. 2010, 132, 4078-4079.

124. Iwasa, E.; Hamashima, Y.; Fujishiro, S.; Hashizume, D.; Sodeoka, M. Total Syntheses of Chaetocin and Ent-Chaetocin. Tetrahedron 2011, 67, 6587-6599.

125. Kim, J.; Movassaghi, M. General Approach to Epipolythiodiketopiperazine Alkaloids: Total Synthesis of (+)-Chaetocins a and C and (+)-12,12'-Dideoxychetracin A. J. Am. Chem. Soc. 2010, 132, 14376-14378.

126. DeLorbe, J. E.; Jabri, S. Y.; Mennen, S. M.; Overman, L. E.; Zhang, F. L. Enantioselective Total Synthesis of (+)-Gliocladine C: Convergent Construction of Cyclotryptamine-Fused Polyoxopiperazines and a General Approach for Preparing Epidithiodioxopiperazines from Trioxopiperazine Precursors. J. Am. Chem. Soc. 2011, 133, 6549-6552.

127. Movassaghi, M.; Boyer, N. Concise Total Synthesis of (+)-Gliocladins B and C. Chem. Sci. 2012, 3, 1798-1803.

128. Nicolaou, K. C.; Totokotsopoulos, S.; Giguere, D.; Sun, Y. P.; Sarlah, D. Total Synthesis of Epicoccin G. J. Am. Chem. Soc. 2011, 133, 8150-8153.

129. Codelli, J. A.; Puchlopek, A. L. A.; Reisman, S. E. Enantioselective Total Synthesis of (-)-Acetylaranotin, a Dihydrooxepine Epidithiodiketopiperazine. J. Am. Chem. Soc. 2012, 134, 1930-1933.

130. Newhouse, T.; Lewis, C. A.; Eastman, K. J.; Baran, P. S. Scalable Total Syntheses of N-Linked Tryptamine Dimers by Direct Indole-Aniline Coupling: Psychotrimine and Kapakahines B and F. J. Am. Chem. Soc. 2010, 132, 7119-7137.

131. Newhouse, T.; Baran, P. S. Total Synthesis of (+/-)-Psychotrimine. J. Am. Chem. Soc. 2008, 130, 10886-10887.

132. Newhouse, T.; Lewis, C. A.; Baran, P. S. Enantiospecific Total Syntheses of Kapakahines B and F. J. Am. Chem. Soc. 2009, 131, 6360-6361.

133. Espejo, V. R.; Rainier, J. D. An Expeditious Synthesis of C(3)-N(1') Heterodimeric Indolines. J. Am. Chem. Soc. 2008, 130, 12894-12895.

102

134. Espejo, V. R.; Rainier, J. D. Total Synthesis of Kapakahine E and F. Org. Lett. 2010, 12, 2154-2157.

135. Perez-Balado, C.; de Lera, A. R. Concise Total Synthesis and Structural Revision of (+)-Pestalazine B. Org. Biomol. Chem. 2010, 8, 5179-5186.

136. Greshock, T. J.; Williams, R. M. Improved Biomimetic Total Synthesis of D,L-Stephacidin A. Org. Lett. 2007, 9, 4255-4258.

137. Movassaghi, M.; Schmidt, M. A.; Ashenhurst, J. A. Stereoselective Oxidative Rearrangement of 2-Aryl Tryptamine Derivatives. Org. Lett. 2008, 10, 4009-4012.

138. Poriel, C.; Lachia, M.; Wilson, C.; Davies, J. R.; Moody, C. J. Oxidative Rearrangement of Indoles: A New Approach to the Efhg-Tetracyclic Core of Diazonamide A. J. Org. Chem. 2007, 72, 2978-2987.

139. Yao, P. Y.; Zhang, Y.; Hsung, R. P.; Zhao, K. A Sequential Metal-Catalyzed C-N Bond Formation in the Synthesis of 2-Amido-Indoles. Org. Lett. 2008, 10, 4275-4278.

140. Istrate, F. M.; Buzas, A. K.; Jurberg, I. D.; Odabachian, Y.; Gagosz, F. Synthesis of Functionalized Oxazolones by a Sequence of Cu(Ii)- and Au(I)-Catalyzed Transformations. Org. Lett. 2008, 10, 925-928.

141. Ohno, M.; Spande, T. F.; Witkop, B. Cyclization of Tryptophan and Tryptamine Derivatives to 2,3-Dihydropyrrolo[2,3-B]Indoles. J. Am. Chem. Soc. 1970, 92, 343-348.

142. Fuchs, J. R.; Funk, R. L. Total Synthesis of (+/-)-Perophoramidine. J. Am. Chem. Soc. 2004, 126, 5068-5069.

143. Yamada, Y.; Arima, S.; Okada, C.; Akiba, A.; Kai, T.; Harigaya, Y. Preparation of 7-Halo-Indoles by Thallation of N-Formylindoline and Their Attempted Use for Synthesis of the Right-Hand Segment of Chloropeptin. Chem. Pharm. Bull. 2006, 54, 788-794.

144. Lopez, C. S.; Perez-Balado, C.; Rodriguez-Grana, P.; de Lera, A. R. Mechanistic Insights into the Stereocontrolled Synthesis of Hexahydropyrrolo[2,3-B]Indoles by Electrophilic Activation of Tryptophan Derivatives. Org. Lett. 2008, 10, 77-80.

145. Welch, T. R.; Williams, R. M. Biomimetic Synthesis of Alkaloids Derived from Tryptophan: Dioxopiperazine Alkaloids. In Biomimetic Organic Synthesis, Poupon, E.; Nay, B., Eds. Wiley-VCH: 2011, 117-148.

146. Di Menna, M. E.; Smith, B. L.; Miles, C. O. A History of Facial Eczema (Pithomycotoxicosis) Research. New Zeal. J. Agr. Res. 2009, 52, 345-376.

103

147. Rege, P. D.; Tian, Y.; Corey, E. J. Studies of New Indole Alkaloid Coupling Methods for the Synthesis of Haplophytine. Org. Lett. 2006, 8, 3117-3120.

148. Ojo, B.; Findsen, L. A.; Igarashi, N.; Kong, B.; Chowdhury, B. K. Synthesis and Bronchodilatory Activity of Four New Derivatives of Deoxyvasicine. Drug Des. Discovery 1996, 14, 1-14.

149. Blunt, J. W.; Erasmuson, A. F.; Ferrier, R. J.; Munro, M. H. G. Syntheses of Haptens Related to the Benzenoid and Indole Portions of Sporidesmin-a - C-13 Nmr-Spectra of Indole-Derivatives. Aust. J. Chem. 1979, 32, 1045-1054.

150. Ishikawa, H.; Elliott, G. I.; Velcicky, J.; Choi, Y.; Boger, D. L. Synthesis of (-)- and Ent-(+)-Vindoline and Related Alkaloids. J. Am. Chem. Soc. 2006, 128, 10596-10612.

151. Koketsu, K.; Oguri, H.; Watanabe, K.; Oikawa, H. Identification and Stereochemical Assignment of the Beta-Hydroxytryptophan Intermediate in the Echinomycin Biosynthetic Pathway. Org. Lett. 2006, 8, 4719-4722.

152. Becker, H.; Sharpless, K. B. A New Ligand Class for the Asymmetric Dihydroxylation of Olefins. Angew. Chem., Int. Ed. 1996, 35, 448-451.

153. Ritchie, R.; Saxton, J. E. Studies on Indolic Mold Metabolites - Total Synthesis of L-Prolyl-2-Methyltryptophan Anhydride and Deoxybrevianamide-E. Tetrahedron 1981, 37, 4295-4303.

154. Zhao, L.; May, J. P.; Huang, J.; Perrin, D. M. Stereoselective Synthesis of Brevianamide E. Org. Lett. 2012, 14, 90-93.

155. Schkeryantz, J. M.; Woo, J. C. G.; Siliphaivanh, P.; Depew, K. M.; Danishefsky, S. J. Total Synthesis of Gypsetin, Deoxybrevianamide E, Brevianamide E, and Tryprostatin B: Novel Constructions of 2,3-Disubstituted Indoles. J. Am. Chem. Soc. 1999, 121, 11964-11975.

156. Greshock, T. J.; Grubbs, A. W.; Williams, R. M. Concise, Biomimetic Total Synthesis of D,L-Marcfortine C. Tetrahedron 2007, 63, 6124-6130.

157. Luo, Y.; Evindar, G.; Fishlock, D.; Lajoie, G. A. Synthesis of N-Protected N-Methyl Serine and Threonine. Tetrahedron Lett. 2001, 42, 3807-3809.

104

CHAPTER 5

Experimental Procedures

Unless otherwise noted, all reagents were obtained from commercial suppliers

and were used without further purification. All air or moisture sensitive reactions were

performed under a positive pressured of argon in flame-dried glassware. Tetrahydrofuran

(THF), toluene, diethyl ether (Et2O), dichloromethane, benzene (PhH), acetonitrile

(MeCN), triethylamine (Et3N), pyridine, diisopropyl amine, methanol (MeOH),

dimethylsulfoxide (DMSO), and N,N-dimethylformamide (DMF) were obtained from a

dry solvent system (Ar degassed solvents delivered through activated alumina columns,

positive pressure of argon). Column chromatography was performed on Merck silica gel

Kieselgel 60 (230-400 mesh). Melting points were determined in open-end capillary

tubes and are uncorrected. 1HNMR and 13CNMR spectra were recorded on Varian 300, or

400 MHz spectrometers. Chemical shifts are reported in ppm relative to CHCl3 at δ 7.27

(1HNMR) and δ 77.23 (13CNMR). Mass spectra were obtained on Fisons VG Autospec.

IR spectra were obtained from thin films on a NaCl plate using a Bruker Tensor 27 FT-IR

spectrometer. Optical rotations were collected at 589 nm on a Rudolph Research

automatic polarimeter Autopol III.

105

Tert-butyl 2,3-dioxoindoline-1-carboxylate (3.5). To a solution of isatin (4.0 g, 27.2

mmol) in THF (100 mL) at 0°C was added Boc2O (7.1 g, 32.6 mmol) and DMAP (195

mg, 1.6 mmol). The resulting mixture was stirred overnight at r.t. under Ar, then

concentrated under reduced pressure. The product was recrystallized in EtOAc/hexanes

to afford 3.5 (4.7 g, 70% yield).

1H-NMR (300 MHz; DMSO-d6): δ 7.92 (d, J = 8.2 Hz, 1H), 7.76-7.65 (m, 2H), 7.29 (t, J

= 7.5 Hz, 1H), 1.56 (s, 9H).

REF: TRW-I-175, TRW-I-179.

NBoc

O

O

3.5

106

Figure 5.1a. 1H NMR spectrum of compound 3.5.

8.38.3

1.01.0

2.02.0

1.01.0

ppm0 01122334455667788-19.97

1.56

7.26

7.29

7.31

7.65

7.66

7.68

7.68

7.71

7.71

7.73

7.74

7.76

7.76

7.91

7.93

107

Boc-Hemiaminal (3.6). To a flame-dried RBF was added 3.5 (500 mg, 2.02 mmol) and

toluene (20 mL). Indoline (134 µL, 1.2 mmol), K2CO3 (372 mg, 2.7 mmol), and 4 Å M.S.

(5 g) were subsequently added, and the resulting suspension heated to reflux for 12 h

under Ar. The reaction mixture was filtered through celite and the fitrate concentrated

under reduced pressure to afford 419 mg of 3.6 as a tan solid (100% yield) that was taken

on without further purification.

1H-NMR (400 MHz; CDCl3): δ 10.59 (s, 1H), 8.55 (d, J = 8.7 Hz, 1H), 8.26 (d, J = 8.1

Hz, 1H), 7.74 (dd, J = 8.1, 1.4 Hz, 1H), 7.57 (dd, J = 8.6, 7.2 Hz, 1H), 7.24-7.20 (m, 1H),

7.13-7.11 (m, 1H), 7.03-6.99 (m, 1H), 3.97 (t, J = 8.4 Hz, 2H), 3.15 (t, J = 8.3 Hz, 2H),

1.54 (s, 9H); 13C-NMR (101 MHz; CDCl3): δ 193.7, 163.4, 152.9, 144.1, 141.7, 137.0,

133.9, 132.0, 129.2, 128.0, 125.5, 121.7, 119.3, 117.7, 81.4, 48.1, 28.49, 28.42.

REF: TRW-I-180, TRW-I-185, TRW-I-201.

NBoc

O

NHO

3.6

108

Figure 5.2a. 1H NMR spectrum of compound 3.6.

Figure 5.2b. 13C NMR spectrum of compound 3.6.

10.210.2

2.42.4

2.12.1

1.21.2

1.11.1

1.41.4

1.31.3

1.01.0

1.01.0

1.01.0

1.21.2

ppm0 0224466881010-17.45

1.54

3.13

3.15

3.17

3.95

3.97

3.99

6.99

6.99

7.01

7.01

7.01

7.03

7.11

7.11

7.11

7.12

7.13

7.13

7.20

7.21

7.23

7.23

7.24

7.24

7.26

7.55

7.57

7.57

7.59

7.73

7.73

7.75

7.75

8.25

8.27

8.54

8.56

10.5

9

ppm0 05050100100150150200200-298.35

28.4

228

.49

48.1

0

81.3

7

117.

6611

9.32

121.

7012

5.51

127.

9712

9.23

132.

0113

3.94

137.

0214

1.72

144.

09

152.

90

163.

42

193.

74

109

1-(2,2-dibromovinyl)-2-nitrobenzene (3.15). 2-nitrobenzaldehyde (3.14, 2.0 g, 13.2

mmol) and CBr4 (6.57 g, 19.8 mmol) were dissolved in CH2Cl2 (100 mL). The resulting

solution was cooled to 0 °C and a solution of PPh3 (10.38 g, 39.6 mmol) in CH2Cl2 (80

mL) slowly added. The reaction was stirred an additional 30 minutes at 0 °C, then

concentrated under reduced pressure. The residue was taken up in chloroform (30 mL),

filtered, and the solid washed with chloroform (2 x 30 mL). The filtrate was concentrated,

then purified by SiO2 chromatography (8:1 hexanes:EtOAc) to afford 3.15 (3.69 g, 91%

yield).

1H-NMR (300 MHz; CDCl3): δ 8.12 (dd, J = 8.2, 1.3 Hz, 1H), 7.78 (s, 1H), 7.71-7.65

(m, 1H), 7.60-7.51 (m, 2H).

REF: TRW-I-286, TRW-I-337, TRW-I-363.

Br

BrNO2

3.15

110

Figure 5.3a. 1H NMR spectrum of compound 3.15.

2.02.0

1.11.1

1.01.0

1.01.0

ppm0 011223344556677889-19.97

7.51

7.52

7.54

7.54

7.55

7.57

7.57

7.58

7.58

7.58

7.60

7.65

7.66

7.68

7.68

7.68

7.70

7.71

7.78

8.10

8.11

8.13

8.14

111

1-(bromoethynyl)-2-nitrobenzene (3.12). To a solution of dibromoalkene 3.15 (3.60 g,

11.7 mmol) and BnEt3NCl (1.33 g, 5.85 mmol) in CH2Cl2 (46 mL) at 0 °C was added a

solution of KOH (30 g) in 23 mL of H2O. The resulting solution was stirred for 1 h at 0

°C, then extracted with CH2Cl2. The organic extracts were dried over Na2SO4, filtered,

concentrated under reduced pressure, and purified by SiO2 chromatography (10-20%

EtOAc in hexanes) to afford 3.12 in quantitative yield.

1H-NMR (300 MHz; CDCl3): δ 8.06 (dd, J = 8.2, 1.4 Hz, 1H), 7.66 (dd, J = 7.7, 1.7 Hz,

1H), 7.58 (td, J = 7.5, 1.4 Hz, 1H), 7.49 (td, J = 7.8, 1.6 Hz, 1H).

REF: TRW-I-291, TRW-I-338, TRW-I-364.

NO2

Br

3.12

112

Figure 5.4a. 1H NMR spectrum of compound 3.12.

1.01.0

1.11.1

1.11.1

1.01.0

ppm1 122334455667788-19.97

7.46

7.47

7.49

7.49

7.51

7.52

7.56

7.56

7.58

7.59

7.61

7.61

7.65

7.65

7.67

7.68

8.05

8.05

8.07

8.08

113

1-((2-nitrophenyl)ethynyl)indoline (3.16). Bromoalkyne 3.12 (185 mg, 0.82 mmol) was

dissolved in toluene (2.75 mL). Indoline (110 µL, 0.98 mmol), K3PO4 (418 mg, 1.97

mmol), CuSO4⋅5H2O (40 mg, 0.16 mmol), and 1,10-phenanthroline (60 mg, 0.33 mmol)

were added successively. The resulting suspension was heated to 120 °C for 4 h in a

microwave. Upon completion of the reaction, the solution was cooled and diluted with

ether, then filtered through celite. The filtrate was concentrated under reduced pressure

and purified by SiO2 chromatography (10-20% EtOAc in hexanes) to afford 3.16 (80 mg,

37% yield).

1H-NMR (300 MHz; CDCl3): δ 8.38 (d, J = 7.9 Hz, 1H), 8.15 (d, J = 8.7 Hz, 1H), 7.68

(d, J = 9.1 Hz, 1H), 7.40 (t, J = 7.7 Hz, 1H), 7.30-7.26 (m, 2H), 7.19 (td, J = 10.2, 6.6 Hz,

2H), 4.72 (t, J = 8.3 Hz, 2H), 3.32 (t, J = 8.3 Hz, 2H).

REF: TRW-I-328, TRW-I-334, TRW-I-342, TRW-I-343, TRW-I-353, TRW-I-360,

TRW-I-362, TRW-I-385.

NO2

N

3.16

114

Figure 5.5a. 1H NMR spectrum of compound 3.16.

2.22.2

2.12.1

2.12.1

2.42.4

1.41.4

1.11.1

1.01.0

1.01.0

ppm1 122334455667788-19.973.

293.

323.

35

4.69

4.72

4.74

7.14

7.16

7.18

7.20

7.21

7.23

7.26

7.26

7.27

7.28

7.30

7.37

7.40

7.42

7.66

7.69

8.13

8.16

8.37

8.39

115

2-(indolin-1-ylethynyl)aniline (3.17). Nitroalkyne 3.16 was dissolved in a 4 to 1

acetone/water mixture (4.3 mL). Zinc dust (141 mg, 2.15 mmol) and NH4Cl (228 mg,

4.30 mmol) were added successively, and the resulting suspension stirred briskly for 10

minutes at r.t. The mixture was concentrated under reduced pressure, diluted with EtOAc,

washed with water, washed with brine, dried over Na2SO4 and concentrated. Purification

by SiO2 chromatography (3:1 hexanes:EtOAc, 1:1 hexanes:EtOAc, then 100% EtOAc)

afforded pure aniline derivative 3.17 (61 mg, 61% yield).

1H-NMR (300 MHz; CDCl3): δ 8.33 (d, J = 8.1 Hz, 1H), 7.26 (d, J = 15.6 Hz, 2H), 7.20-

7.04 (m, 4H), 6.72 (dd, J = 7.4, 2.3 Hz, 2H), 4.39-4.14 (m, 2H), 3.96 (td, J = 10.4, 6.1

Hz, 1H), 3.54 (td, J = 10.4, 7.0 Hz, 1H), 3.21-2.99 (m, 2H); 13C-NMR (75 MHz; CDCl3):

δ 131.5, 130.1, 129.1, 127.9, 124.98, 124.95, 122.2, 118.8, 117.6, 117.3, 112.5, 71.6,

47.5, 28.4.

REF: TRW-I-331, TRW-I-336, TRW-I-352, TRW-I-357, TRW-I-361, TRW-I-366.

NH2

N

3.17

116

Figure 5.6a. 1H NMR spectrum of compound 3.17.

Figure 5.6b. 13C NMR spectrum of compound 3.17.

2.42.4

1.11.1

1.21.2

3.13.1

1.91.9

4.14.1

1.81.8

1.01.0

ppm0 01122334455667788-19.97

2.99

3.02

3.02

3.03

3.03

3.07

3.07

3.08

3.09

3.09

3.15

3.20

3.21

3.49

3.52

3.53

3.55

3.56

3.59

3.91

3.93

3.95

3.97

3.98

4.00

4.14

4.15

4.15

4.15

4.16

4.16

4.37

4.38

4.38

4.38

4.38

4.39

6.70

6.71

6.73

6.73

7.04

7.06

7.06

7.06

7.07

7.08

7.11

7.11

7.12

7.14

7.14

7.14

7.14

7.16

7.16

7.17

7.17

7.18

7.20

7.20

7.20

7.24

7.29

8.32

8.34

ppm0 05050100100150150-303.86

28.4

0

47.4

7

71.6

5

112.

4511

7.31

117.

5811

8.83

122.

2212

4.95

124.

9812

7.90

129.

1113

0.07

131.

55

117

Methyl 3-(benzyloxycarbonylamino)-4-(indolin-3-yl)butanoate (3.21). N-Cbz-L-Trp-

OMe (3.52 g, 10.0 mmol) was dissolved in THF/TFA (16.5 mL/16.5 mL) and cooled to 0

°C. BH3⋅Me2S was added dropwise and the resulting solution stirred 15 min. at 0 °C.

Water (33 mL) was added and the mixture stirred 15 min. at r.t. The solvent was removed

in vacuo and TFA azeotroped off with toluene (3x). The residue was taken up in EtOAc,

washed with 1M NaOH (3x), water (1x), and brine (1x) successively. The organic layer

was dried over Na2SO4, filtered, concentrated under reduced pressure, and purified by

SiO2 chromatography (2:1 – 1:1 hexanes:EtOAc) to afford indoline 3.21 (2.81 g, 79%

yield).

1H-NMR (300 MHz; CDCl3): δ 7.36 (s, 5H), 7.03 (t, J = 7.1 Hz, 2H), 6.74-6.71 (m, 1H),

6.64 (dd, J = 9.5, 1.8 Hz, 1H), 5.37 (dd, J = 26.1, 8.8 Hz, 1H), 5.12 (s, 2H), 4.53-4.44 (m,

1H), 3.74-3.68 (m, 4H), 3.36-3.22 (m, 2H), 2.11-2.06 (m, 2H).

REF: TRW-I-369.

NH

CO2Me

NHCbz

3.21

118

Figure 5.7a. 1H NMR spectrum of compound 3.21.

2.42.4

2.42.4

4.34.3

1.11.1

2.12.1

1.01.0

1.11.1

1.01.0

1.81.8

5.05.0

ppm1 1223344556677-19.97

2.06

2.08

2.08

2.08

2.10

2.11

3.22

3.25

3.25

3.31

3.33

3.34

3.36

3.36

3.36

3.68

3.71

3.74

4.44

4.45

4.46

4.46

4.46

4.48

4.48

4.49

4.51

4.52

4.53

5.12

5.31

5.34

5.40

5.43

6.63

6.63

6.66

6.66

6.71

6.73

6.74

7.01

7.03

7.06

7.36

119

(S)-methyl 3-(1H-indol-3-yl)-2-(2,2,2-trifluoroacetamido)propanoate (3.31). To a

solution of L-Trp-OMe·HCl (1.0 g, 3.92 mmol) in MeOH was added trifluoromethyl

propionate (0.94 mL, 7.84 mmol) and Et3N (1.1 mL, 7.84 mmol). After stirring 1 h at r.t.,

the reaction mixture was concentrated and the resulting residue dissolved in CH2Cl2. The

organic phase was washed with ammonium hydroxide (5%, 1x) and brine (1x), then dried

over Na2SO4 and concentrated to afford 1.23 g of the title compound, carried forward as

crude (>99% yield).

1H-NMR (300 MHz; CDCl3): δ 8.25-8.23 (bs, 1H), 7.49 (d, J = 7.8 Hz, 1H), 7.37 (d, J =

8.1 Hz, 1H), 7.21 (td, J = 7.5, 1.1 Hz, 1H), 7.13 (td, J = 7.4, 1.0 Hz, 1H), 6.98 (d, J = 2.4

Hz, 1H), 6.87-6.85 (bs, 1H), 4.94 (dt, J = 7.8, 5.0 Hz, 1H), 3.73 (s, 3H), 3.42 (d, J = 5.1

Hz, 2H).

REF: TRW-I-423, TRW-I-425.

NH

NH

CO2Me

O CF3

3.31

120

Figure 5.8a. 1H NMR spectrum of compound 3.31.

2.12.1

3.03.0

1.01.0

0.80.8

1.01.0

1.01.0

1.11.1

1.11.1

1.11.1

1.01.0

ppm1 122334455667788-19.973.

413.

43

3.73

4.91

4.92

4.93

4.94

4.95

4.96

6.85

6.85

6.87

6.87

6.87

6.97

6.98

7.10

7.11

7.13

7.13

7.15

7.16

7.18

7.19

7.21

7.21

7.23

7.24

7.36

7.39

7.48

7.50

8.23

8.23

8.24

8.24

8.24

8.24

8.24

8.25

121

(S)-methyl 1-(2,2,2-trifluoroacetyl)-1,2,3,8-tetrahydropyrrolo[2,3-b]indole-2-

carboxylate (3.32). To a solution of 3.31 (1.07 g, 3.4 mmol) in CH2Cl2 (34 mL) and Et3N

(1.9 mL, 13.6 mmol) at 0 °C was added t-BuOCl (0.41 mL, 3.4 mmol) dropwise. The

resulting solution was allowed to warm to r.t. O/N with stirring. Water was added and the

product extracted in to Et2O. The combined organic layers were washed with brine, dried

over Na2SO4, and concentrated. Recrystallization from EtOAc/hexanes provided the pure

title compound (670 mg, 63% yield).

1H-NMR (300 MHz; CDCl3): δ 9.17-9.16 (bs, 1H), 7.39 (td, J = 6.7, 2.6 Hz, 2H), 7.17

(ddd, J = 7.0, 4.3, 2.2 Hz, 2H), 5.55 (dt, J = 9.7, 1.8 Hz, 1H), 3.82 (s, 3H), 3.74 (dd, J =

14.8, 9.7 Hz, 1H), 3.36 (dd, J = 14.8, 2.3 Hz, 1H).

REF: TRW-I-424, TRW-I-427.

NH

NCOCF3

CO2Me

3.32

122

Figure 5.9a. 1H NMR spectrum of compound 3.32.

1.11.1

1.31.3

3.33.3

1.11.1

1.91.9

2.22.2

1.01.0

ppm2 233445566778899-19.97

3.33

3.34

3.38

3.39

3.70

3.73

3.75

3.78

3.82

5.53

5.53

5.54

5.56

5.57

5.57

7.15

7.15

7.16

7.17

7.18

7.18

7.19

7.36

7.37

7.38

7.39

7.40

7.41

9.16

9.16

9.17

123

(2S,3aR,8aS)-1-tert-butyl 2-methyl 3a-((2-iodophenyl)amino)-3,3a,8,8a-

tetrahydropyrrolo[2,3-b]indole-1,2(2H)-dicarboxylate (3.35). N-Boc-Trp-OMe (100

mg, 0.31 mmol) was dissolved in acetonitrile (6 mL), then 2-iodoaniline (81 mg, 0.37

mmol) added and the resulting solution cooled to -45 °C. Freshly recrystallized NIS (113

mg, 0.50 mmol) was added dropwise over 1 h as a solution in acetonitrile (1.5 mL). The

reaction was allowed to warm to -35 °C as it stirred an additional hour. The entire

reaction mixture was poured into a separatory funnel containing sat’d Na2S2O4 and

EtOAc. The product was extracted in EtOAc (3x) and the combined organic layers

washed with brine, dried over Na2SO4, and concentrated. Crude 3.35 was purified by

SiO2 chromatography (5-10% EtOAc in hexanes) to afford pure 3.35 in 78% yield (130

mg).

1H-NMR (300 MHz; CDCl3): δ 7.64 (t, J = 7.8 Hz, 1H), 7.12 (dt, J = 13.7, 7.3 Hz, 2H),

6.99 (t, J = 7.7 Hz, 1H), 6.75-6.65 (m, 2H), 6.40 (t, J = 7.6 Hz, 1H), 6.30 (dd, J = 16.2,

8.2 Hz, 1H), 5.89 (d, J = 34.7 Hz, 1H), 4.63 (d, J = 25.6 Hz, 1H), 4.39 (ddd, J = 25.8, 8.3,

4.3 Hz, 1H), 3.82 (s, 3H), 2.72 (qd, J = 15.3, 6.6 Hz, 2H), 1.46 (d, J = 30.4 Hz, 9H).

REF: TRW-I-435, TRW-I-441.

NH

NBoc

CO2MeNH

I

H

3.35

124

Figure 5.10a. 1H NMR spectrum of compound 3.35.

9.09.0

2.12.1

2.82.8

1.21.2

1.21.2

1.01.0

0.90.9

1.21.2

1.91.9

1.01.0

2.02.0

1.01.0

ppm0 011223344556677-19.97

1.42

1.52

2.63

2.66

2.68

2.71

2.74

2.76

2.79

2.80

3.82

4.33

4.34

4.35

4.37

4.41

4.43

4.44

4.45

4.59

4.68

5.82

5.94

6.26

6.28

6.31

6.34

6.38

6.40

6.43

6.65

6.67

6.70

6.73

6.75

6.96

6.99

7.01

7.07

7.10

7.12

7.14

7.17

7.61

7.64

7.67

125

L-serine-OMe (3.39). To a solution of L-serine (50.0 g, 0.48 mol) in MeOH (450 mL) at

0 °C was added SOCl2 (35 mL, 0.48 mol). The reaction was allowed to warm to r.t. as it

stirred O/N. The resulting solution was concentrated, taken up in ether, filtered, and the

solid washed with ether. The crude solid material was recrystallized in MeOH/ether to

afford pure crystalline methyl ester 3.39 as the HCl salt.

REF: TRW-I-462, TRW-II-118.

N-Boc-Ser-OMe (3.40). Boc anhydride (12.2 g, 56 mmol) was dissolved in acetonitrile

(190 mL), then Et3N (24 mL, 168 mmol) and L-Ser-OMe·HCl (8.74 g, 56 mmol) added.

The reaction was allowed to stir for 6 h at r.t., then CH2Cl2 (950 mL) and 1M HCl (665

mL) added. The organic layer was separated and washed with sat’d NaHCO3, then dried

over Na2SO4 and concentrated to afford the title material, used as crude (10.01 g, 82%

yield).

1H-NMR (300 MHz; CDCl3): δ 5.43 (m, J = 1.6, 0.8 Hz, 1H), 4.40-4.38 (m, 1H), 3.97-

3.91 (m, 2H), 3.79 (s, 3H), 2.20-2.19 (m, 1H), 1.46 (s, 9H).

REF: TRW-I-299, TRW-I-439.

MeO2C NH2

OH

3.39

MeO2C NHBoc

OH

3.40

126

Figure 5.11a. 1H NMR spectrum of compound 3.40.

9.49.4

0.90.9

3.03.0

2.12.1

0.90.9

0.90.9

ppm0 011223344556677-19.97

1.46

2.19

2.19

2.19

2.20

2.20

3.79

3.91

3.91

3.92

3.93

3.94

3.94

3.95

3.96

3.97

4.38

4.38

4.39

4.39

4.39

4.40

4.40

4.40

4.40

5.42

5.42

5.42

5.42

5.43

5.43

5.43

5.43

5.44

5.44

5.44

5.44

5.45

5.45

127

Methyl 2-(tert-butoxycarbonylamino)-3-iodopropanoate (3.41). To a solution of Ph3P

(2.13 g, 8.13 mmol) and imidazole (553 mg, 8.13 mmol) in CH2Cl2 (30 mL) at 0 °C was

added I2 (2.07 g, 8.13 mmol) in 3 portions. The reaction mixture was warmed to r.t. for

10 minutes, then cooled to 0 °C before adding dropwise a solution of Boc-Ser-OMe (1.42

g, 6.5 mmol) in 10 mL CH2Cl2. The resulting solution was stirred 1 h at 0 °C, then

allowed to warm to r.t. as it stirred an additional 1.5 h. The crude reaction mixture was

filtered through a plug of silica with 1:1 EtOAC/hexanes. The filtrate was concentrated

under reduced pressure and purified by SiO2 chromatography (5-20% EtOAc in hexanes)

to afford iodide 3.41 (1.17 g, 55% yield).

1H-NMR (300 MHz; CDCl3): δ 5.36-5.33 (m, 1H), 4.55-4.50 (m, 1H), 3.80 (s, 3H), 3.56

(dq, J = 9.8, 5.0 Hz, 2H), 1.46 (s, 9H).

REF: TRW-I-196, TRW-I-205, TRW-I-243, TRW-I-440

MeO2C NHBoc

I

3.41

128

Figure 5.12a. 1H NMR spectrum of compound 3.41.

10.210.2

2.02.0

3.03.0

0.90.9

0.90.9

ppm1 1223344556677-19.97

1.46

3.52

3.54

3.56

3.57

3.58

3.61

3.62

3.80

4.50

4.50

4.50

4.51

4.52

4.53

4.54

4.55

5.33

5.33

5.34

5.34

5.34

5.34

5.34

5.34

5.35

5.35

5.36

5.36

5.36

5.36

129

(S)-methyl 2-((tert-butoxycarbonyl)amino)-5-(trimethylsilyl)pent-4-ynoate (3.42). A

flask (Flask A) containing CuCN (123 mg, 1.37 mmol) and LiCl (115 mg, 2.74 mmol)

was heated to 150 °C under vacuum for 2 h, then cooled to r.t. DMF (4 mL) was added

and the suspension sonicated until full dissolution was observed. This solution was then

cooled to -20 °C. A second flask (Flask B) containing Zn dust (358 mg, 5.47 mmol) was

heated under vaccum, cooled to r.t., filled with Ar, then DMF (1.75 mL) and

dibromoethane (26 µL, 0.30 mmol) added. The suspension was heated to 80 °C for 30

min, then cooled to r.t. before addition of TMSCl (19 µL, 0.15 mmol). Following an

additional 30 min of stirring, N-Boc-Ser(I)-OMe (3.41, 500 mg, 1.52 mmol) was added

slowly as a solution in a minimal amount of DMF. After complete addition, this

suspension was transferred to Flask A. The combined reaction mixture was allowed to

stir for 15 min at -20 °C, then TMS-acetylene bromide (269 mg, 1.52 mmol) added

dropwise. The reaction mixture was allowed to warm to r.t. O/N. Water was added (15

mL) and the product extracted in EtOAc. The combined organic layers were washed with

brine, dried over Na2SO4, and concentrated. SiO2 chromatography (3:1 hexanes/EtOAc)

afforded the pure title compound (180 mg, 40% yield, stains with vanillin).

1H-NMR (300 MHz; CDCl3): δ 5.28 (d, J = 8.4 Hz, 1H), 4.43-4.37 (m, 1H), 3.70 (s, 3H),

2.75-2.59 (m, 2H), 1.39 (s, 9H), 0.07 (s, 8H).

REF: TRW-I-444, TRW-I-452, TRW-I-469.

MeO2C NHBoc

TMS

3.42

130

Figure 5.13a. 1H NMR spectrum of compound 3.42.

8.58.5

9.09.0

2.02.0

2.82.8

0.80.8

0.80.8

ppm-1 -10011223344556-19.97

0.07

1.39

2.59

2.61

2.65

2.67

2.68

2.69

2.73

2.75

3.70

4.37

4.39

4.40

4.40

4.42

4.43

5.27

5.30

131

(Bromoethynyl)trimethylsilane. To a solution of TMS-acetylene (5 mL, 35.5 mmol) in

acetone (120 mL) was added AgNO3 (604 mg, 3.55 mmol) and NBS (6.94 g, 39 mmol).

After stirring 3 h, the reaction was quenched by addition of ice water. The product was

extracted in pentane and the combined organic layers washed with brine, dried over

Na2SO4, and concentrated (bath at 0 °C) to afford pure (bromoethynyl)trimethylsilane

(4.51 g, 72% yield).

1H-NMR (300 MHz; CDCl3): δ 0.18 (s, 9H).

REF: TRW-I-442, TRW-I-457.

Figure 5.14a. 1H NMR spectrum of (bromoethynyl)trimethysilane.

Br TMS

9.09.0

ppm0 011223344-19.97

0.18

132

(2S,3aR,8aS)-1-tert-butyl 2-methyl 3a-(3-((S)-2-((tert-butoxycarbonyl)amino)-3-

methoxy-3-oxopropyl)-1H-indol-1-yl)-3,3a,8,8a-tetrahydropyrrolo[2,3-b]indole-

1,2(2H)-dicarboxylate (3.43). A flask containing 3.35 (300 mg, 0.56 mmol), 3.42 (370

mg, 1.24 mmol), NaOAc (321 mg, 3.92 mmol), LiCl (24 mg, 0.56 mmol), and Pd(OAc)2

(25 mg, 0.11 mmol) was vacated and backfilled with Ar. DMF (3 mL) was added and the

suspension sonicated for 15 minutes while bubbling Ar through reaction. Three freeze-

pump-thaw cycles were performed to complete the degassing procedure. The reaction

mixture was heated to 100 °C for 24 h, then diluted with toluene and concentrated. The

crude residue was taken up in EtOAc, filtered through celite, and washed with 3M HCl.

The organic fraction was further washed with brine, dried over Na2SO4, and concentrated

to afford the title compound (3.43, 250 mg, 70% yield).

1H-NMR (300 MHz; CDCl3): δ 7.62 (d, J = 8.1 Hz, 1H), 7.52 (d, J = 7.6 Hz, 1H), 7.35-

7.25 (m, 3H), 7.14 (d, J = 7.4 Hz, 1H), 6.92 (d, J = 7.5 Hz, 1H), 6.77 (d, J = 7.7 Hz, 1H),

6.64 (s, 1H), 6.05-5.91 (m, 1H), 5.02 (d, J = 7.9 Hz, 1H), 4.55-4.53 (m, 1H), 4.28-4.20

(m, 1H), 3.80 (d, J = 3.1 Hz, 3H), 3.58-3.47 (m, 3H), 3.36 (dd, J = 13.1, 9.3 Hz, 1H),

3.15-3.14 (m, 1H), 2.98 (dd, J = 13.2, 7.8 Hz, 1H), 1.42 (complex, J = 28.4 Hz, 18H).

REF: TRW-I-453, TRW-I-458.

NH

NBoc

CO2MeN

NHBoc

CO2Me

H

3.43

133

Figure 5.15a. 1H NMR spectrum of compound 3.43.

17.717.7

0.90.9

1.31.3

0.90.9

2.52.5

2.92.9

0.80.8

0.70.7

0.70.7

0.90.9

0.70.7

1.01.0

1.01.0

1.01.0

2.82.8

1.01.0

1.01.0

ppm0 0112233445566778-19.97

1.33

1.42

1.52

2.95

2.97

2.99

3.02

3.14

3.14

3.15

3.32

3.35

3.37

3.40

3.47

3.49

3.58

3.80

3.81

4.20

4.22

4.23

4.24

4.25

4.28

4.28

4.53

4.54

4.55

4.55

5.01

5.03

5.91

6.04

6.05

6.64

6.75

6.78

6.91

6.94

7.13

7.15

7.25

7.26

7.26

7.28

7.30

7.33

7.35

7.51

7.53

7.61

7.63

134

(2S,3aR,8aS)-1-(tert-butoxycarbonyl)-3a-(3-((S)-2-((tert-butoxycarbonyl)amino)-2-

carboxyethyl)-1H-indol-1-yl)-1,2,3,3a,8,8a-hexahydropyrrolo[2,3-b]indole-2-

carboxylic acid (3.44). To a solution of 3.43 (100 mg, 0.136 mmol) in THF and MeOH

(2:1, 0.40 mL:0.20 mL) was added LiOH (20 mg, 0.80 mmol). After stirring under Ar

O/N, 10% KHSO4 was added until the solution was acidic, then the product extracted in

EtOAc. The combined organic extracts were washed with brine, dried over Na2SO4, and

concentrated. Crude 3.44 recovered as such was carried forward to the next reaction (90.4

mg, 93% yield).

1H-NMR (300 MHz; CDCl3): δ 7.55-7.53 (m, 1H), 7.17 (d, J = 8.4 Hz, 2H), 7.06-7.00

(m, 1H), 6.90-6.86 (m, 1H), 6.79-6.62 (m, 3H), 6.48-6.32 (m, 1H), 6.24-6.13 (m, 1H),

4.63-4.34 (m, 2H), 3.56-3.47 (m, 1H), 3.20-3.18 (m, 1H), 3.04-2.82 (m, 2H), 1.50-1.38

(m, 18H).

REF: TRW-I-456, TRW-I-460.

NH

NBoc

CO2HN

NHBoc

CO2H

H

3.44

135

Figure 5.16a. 1H NMR spectrum of compound 3.44.

19.019.0

1.81.8

0.60.6

1.31.3

2.02.0

0.70.7

0.60.6

3.23.2

1.21.2

1.51.5

2.12.1

1.01.0

ppm0 011223344556677-19.97

1.38

1.40

1.50

2.82

2.82

2.84

2.87

2.87

3.02

3.04

3.04

3.18

3.18

3.18

3.19

3.19

3.19

3.20

3.20

3.20

3.20

3.47

3.47

3.47

3.48

3.48

3.48

3.49

3.50

3.51

3.51

3.53

3.53

3.53

3.53

3.55

3.56

4.34

4.36

4.36

4.36

4.57

4.60

4.63

6.13

6.14

6.24

6.32

6.32

6.45

6.45

6.45

6.46

6.46

6.47

6.47

6.47

6.47

6.48

6.48

6.62

6.65

6.69

6.69

6.70

6.72

6.74

6.76

6.79

6.86

6.87

6.87

6.88

6.88

6.90

7.00

7.04

7.04

7.05

7.06

7.15

7.18

7.53

7.55

7.55

7.55

136

(2S,3aR,8aR)-1-benzyl 8-tert-butyl 2-methyl 3a-bromo-3,3a-dihydropyrrolo[2,3-

b]indole-1,2,8(2H,8aH)-tricarboxylate (3.47). To a solution of N-Cbz,N’-Boc-Trp-OMe

(1.0 g, 2.2 mmol) in CH2Cl2 (100 mL) was added PPTS (552 mg, 2.2 mmol) and NBS

(392 mg, 2.2 mmol). The resulting solution was stirred O/N at r.t, then diluted with

CH2Cl2, washed with brine, dried over Na2SO4, and concentrated. Crude 3.47 was

purified by SiO2 chromatography (25% EtOAc in hexanes) to afford pure material in

88% yield (1.03 g).

1H-NMR (300 MHz; CDCl3): δ 7.37-7.28 (m, 8H), 7.12 (td, J = 7.5, 0.7 Hz, 1H), 6.43 (s,

1H), 5.19-5.16 (m, 2H), 3.97 (dd, J = 10.3, 6.5 Hz, 1H), 3.69-3.41 (m, 3H), 3.26 (dd, J =

12.7, 6.5 Hz, 1H), 2.84 (dd, J = 12.7, 10.3 Hz, 1H), 1.52 (s, 9H).

REF: TRW-I-477.

NBoc

NCbz

CO2MeBr

H

3.47

137

Figure 5.17a. 1H NMR spectrum of compound 3.47.

14.314.3

1.21.2

1.41.4

3.23.2

1.21.2

1.81.8

1.01.0

1.31.3

7.77.7

ppm1 1223344556677-19.97

1.52

2.81

2.84

2.85

2.88

3.22

3.25

3.27

3.29

3.41

3.66

3.66

3.67

3.67

3.68

3.69

3.69

3.94

3.97

3.98

4.00

5.16

5.17

5.17

5.18

5.18

5.18

5.19

5.19

6.43

7.10

7.10

7.12

7.12

7.15

7.15

7.26

7.27

7.28

7.28

7.28

7.28

7.29

7.30

7.31

7.32

7.32

7.32

7.33

7.34

7.35

7.37

138

(2R,3aR,8aR)-1-benzyl 8-tert-butyl 2-methyl 3a-(1H-indol-1-yl)-3,3a-

dihydropyrrolo[2,3-b]indole-1,2,8(2H,8aH)-tricarboxylate (3.48).

Bromopyrroloindoline 3.47 (500 mg, 0.94 mmol) was dissolved in acetonitrile (20 mL),

and to it added indole (74 mg, 0.63 mmol). The suspension was cooled to 0 °C, then

KOtBu (141 mg, 1.26 mmol) added slowly. Once the reaction was deemed complete by

TLC, the mixture was quenched by addition of sat’d NaHCO3 and the product extracted

in CH2Cl2. The combined organic layers were washed with brine, dried over Na2SO4,

concentrated, and purified by SiO2 chromatography (3:1 hexanes:EtOAc) to afford pure

3.48 (100 mg, 28% yield).

1H-NMR (300 MHz; CDCl3): δ 7.79-7.75 (bs, 1H), 7.65-7.62 (m, 1H), 7.44-7.27 (m,

8H), 7.19-7.15 (m, 3H), 6.96 (d, J = 3.0 Hz, 1H), 6.44 (d, J = 3.4 Hz, 1H), 5.30-5.20 (m,

2H), 4.97 (d, J = 8.9 Hz, 1H), 3.57 (dd, J = 13.0, 9.3 Hz, 1H), 3.22 (s, 3H), 3.07 (d, J =

13.1 Hz, 1H), 1.47 (bs, 9H).

REF: TRW-I-482, TRW-I-484.

NBoc

NCbz

CO2MeN

H3.48

139

Figure 5.18a. 1H NMR spectrum of compound 3.48.

8.48.4

0.90.9

2.62.6

0.90.9

0.80.8

1.71.7

0.70.7

1.01.0

2.72.7

8.18.1

1.01.0

1.01.0

ppm1 1223344556677-19.97

1.47

1.47

3.05

3.09

3.22

3.54

3.57

3.58

3.61

4.96

4.99

5.20

5.24

5.27

5.27

5.27

5.28

5.28

5.29

5.29

5.29

5.29

5.29

5.30

5.30

5.30

6.43

6.45

6.95

6.96

7.15

7.17

7.17

7.19

7.27

7.29

7.36

7.41

7.44

7.62

7.63

7.65

7.75

7.76

7.76

7.76

7.76

7.76

7.77

7.77

7.77

7.78

7.78

7.78

7.78

7.79

140

N-Boc-L-tryptophan-N-SO2Ph (3.56). N-Boc-Trp (14.8 g, 48.7 mmol) was dissolved in

THF (100 mL) and the resulting solution cooled to -78 °C. LHMDS (146 mL, 146.1

mmol) was added slowly over 5 minutes. The reaction was stirred at -78 °C for 1 h, then

PhSO2Cl (7.5 mL, 58.4 mmol) added in one portion. After stirring an additional 2 h at -78

°C, the reaction was quenched by addition of 1:1 AcOH:EtOAc then allowed to warm to

r.t. The suspension was diluted with 1M HCl and the product extracted in EtOAc. The

combined organic layers were dried and concentrated to afford crude 3.56, purified by

SiO2 chromatography (5% AcOH, 45% hexanes, 50% CH2Cl2) to afford pure 3.56 (17.12

g, 79% yield).

1H-NMR (300 MHz; DMSO-d6): δ 12.64-12.63 (m, 1H), 7.90-7.87 (m, 2H), 7.61 (t, J =

7.8 Hz, 2H), 7.56-7.50 (m, 2H), 7.25-7.20 (m, 2H), 7.16-7.13 (m, 2H), 4.20-4.17 (m, 1H),

3.11 (dd, J = 14.8, 4.2 Hz, 1H), 2.95 (dd, J = 14.8, 10.0 Hz, 1H), 1.31 (s, 9H).

REF: TRW-II-176, TRW-II-251.

NNHBoc

SO2Ph

CO2H

3.56

141

Figure 5.19a. 1H NMR spectrum of compound 3.56.

9.49.4

1.01.0

1.01.0

0.90.9

2.42.4

2.42.4

2.32.3

2.22.2

2.32.3

0.90.9

ppm0 02244668810101212-19.97

1.31

2.91

2.94

2.96

2.99

3.08

3.10

3.13

3.15

4.17

4.19

4.19

4.19

4.20

7.13

7.13

7.16

7.20

7.22

7.23

7.25

7.25

7.27

7.27

7.32

7.50

7.53

7.56

7.56

7.59

7.61

7.64

7.87

7.88

7.89

7.90

12.6

312

.63

12.6

312

.64

12.6

4

142

Dipeptide (3.57a). To a solution of N-Me-L-serine-OMe (2.0 g, 15 mmol) in CH2Cl2 (75

mL) was added iPr2NEt (2.61 mL, 15 mmol). After stirring 15 minutes, the suspension

was added to a stirring solution of 3.56 (6.66 g, 15 mmol) and EDCI (2.88 g, 15 mmol) in

CH2Cl2. The reaction was stirred O/N at r.t., then concentrated and purified through a

plug of SiO2 (100% EtOAc) to afford 3.57a (86% yield).

1H-NMR (300 MHz; DMSO-d6): δ 8.40 (d, J = 7.4 Hz, 1H), 7.87 (t, J = 8.8 Hz, 2H),

7.67 (dt, J = 21.1, 6.5 Hz, 2H), 7.54 (t, J = 7.7 Hz, 2H), 7.29 (complex, 2H), 6.95 (d, J =

8.9 Hz, 1H), 5.14 (dt, J = 11.2, 5.6 Hz, 1H), 4.47 (d, J = 5.7 Hz, 1H), 4.38-4.34 (m, 1H),

3.77-3.70 (m, 1H), 3.60 (s, 3H), 3.07-3.01 (m, 1H), 2.88-2.80 (m, 1H), 2.48 (s, 3H), 1.25

(s, 9H), 1.06 (s, 1H).

REF: TRW-II-129.

NNHBoc

NMe

O

SO2Ph

CO2Me

OH

3.57a

143

Figure 5.20a. 1H NMR spectrum of compound 3.57a.

1.11.1

6.76.7

1.81.8

0.80.8

0.90.9

3.03.0

0.90.9

1.41.4

0.60.6

1.01.0

0.70.7

2.32.3

1.71.7

2.42.4

2.42.4

0.70.7

ppm0 01122334455667788-19.97

1.06

1.25

2.48

2.80

2.83

2.83

2.84

2.84

2.88

2.88

3.01

3.01

3.02

3.02

3.02

3.03

3.06

3.06

3.07

3.07

3.60

3.70

3.71

3.72

3.73

3.74

3.75

3.77

3.77

4.34

4.35

4.36

4.37

4.37

4.38

4.38

4.46

4.48

5.10

5.12

5.14

5.16

5.18

6.93

6.96

7.24

7.26

7.28

7.30

7.33

7.51

7.54

7.56

7.62

7.63

7.65

7.68

7.70

7.73

7.84

7.87

7.89

8.39

8.41

144

Dioxopiperazine (3.58a). Dipeptide 3.57a (500 mg) was transferred to a microwave

tube, then heated neat in the microwave at maximum power to a safe temperature of 180

°C (microwave set to ‘open-vessel’). After cooling, the residue was suspended in CH2Cl2,

transferred to a vial, and concentrated to afford dioxopiperazine 3.58a (94% yield).

1H-NMR (300 MHz; DMSO-d6): δ 8.15 (d, J = 2.4 Hz, 1H), 8.03 (d, J = 2.0 Hz, 1H),

7.89 (dd, J = 24.0, 7.8 Hz, 3H), 7.64 (t, J = 3.6 Hz, 2H), 7.56 (d, J = 4.0 Hz, 1H), 7.33-

7.27 (m, 2H), 5.14 (t, J = 5.7 Hz, 1H), 4.47 (d, J = 5.6 Hz, 1H), 4.05-4.00 (m, 1H), 3.74

(d, J = 2.1 Hz, 1H), 3.65-3.60 (m, 1H), 3.42 (t, J = 4.2 Hz, 1H), 3.24 (dd, J = 59.1, 5.0

Hz, 3H).

REF: TRW-II-132.

NHN

NMeOH

O

OSO2Ph3.58a

145

Figure 5.21a. 1H NMR spectrum of compound 3.58a.

2.32.3

1.31.3

0.90.9

0.80.8

0.70.7

0.60.6

1.01.0

2.12.1

1.31.3

2.12.1

3.03.0

0.80.8

0.80.8

ppm1 122334455667788-19.97

3.14

3.15

3.33

3.35

3.41

3.42

3.44

3.60

3.60

3.64

3.65

3.65

3.73

3.74

4.00

4.00

4.00

4.01

4.01

4.01

4.01

4.02

4.03

4.04

4.04

4.04

4.05

4.05

4.46

4.48

5.13

5.14

5.16

7.27

7.28

7.29

7.30

7.33

7.33

7.53

7.55

7.56

7.57

7.58

7.59

7.63

7.64

7.66

7.84

7.86

7.92

7.94

8.02

8.03

8.14

8.15

146

(S)-methyl 2-((S)-2-((tert-butoxycarbonyl)amino)-3-(1H-indol-3-yl)propan-amido)-3-

hydroxypropanoate (3.57c). To a solution of L-Ser-OMe·HCl (2.56 g, 16.4 mmol) in

CH2Cl2 (80 mL) was added iPr2NEt (2.85 mL, 16.4 mmol). After stirring 15 minutes, N-

Boc-L-Trp (5.0 g, 16.4 mmol) and EDCI (3.15 g, 16.4 mmol) were added. The reaction

was stirred O/N at r.t., then concentrated and purified through a plug of SiO2 (100%

EtOAc) to afford 3.57c (5.71 g, 86% yield).

1H-NMR (400 MHz; CDCl3): δ 8.69 (bs, 1H), 7.52 (d, J = 7.8 Hz, 1H), 7.23 (d, J = 8.0

Hz, 1H), 7.06 (t, J = 7.4 Hz, 1H), 7.01-6.97 (m, 2H), 5.34 (d, J = 7.4 Hz, 1H), 4.42 (bs,

2H), 3.70 (bs, 2H), 3.54 (s, 3H), 3.16 (bs, 2H), 1.30 (s, 9H); 13C-NMR (101 MHz;

CDCl3): δ 172.4, 170.6, 155.8, 136.2, 127.5, 123.5, 122.0, 119.4, 118.5, 111.4, 109.8,

80.4, 62.5, 54.8, 52.6, 28.23, 28.14, 28.08.

REF: TRW-II-043.

NH

NHBocNH

O

CO2Me

OH

3.57c

147

Figure 5.22a. 1H NMR spectrum of compound 3.57c.

Figure 5.22b. 13C NMR spectrum of compound 3.57c.

8.98.9

2.42.4

3.03.0

2.02.0

2.02.0

1.01.0

2.52.5

1.11.1

1.11.1

1.01.0

1.01.0

ppm1 122334455667788-16.04

1.30

3.16

3.54

3.70

3.70

3.70

4.42

5.33

5.35

6.97

6.97

6.99

7.01

7.05

7.06

7.08

7.22

7.24

7.51

7.53

8.69

ppm0 05050100100150150-253.79

28.0

828

.14

28.2

3

52.5

654

.85

62.4

6

80.3

7

109.

8411

1.37

118.

5311

9.38

121.

9612

3.55

127.

51

136.

25

155.

77

170.

5717

2.39

148

Dipeptide (3.57d). To a solution of L-Ser-OMe·HCl (6.0 g, 38.5 mmol) in CH2Cl2 (200

mL) was added iPr2NEt (6.7 mL, 38.5 mmol). After stirring 15 minutes, the suspension

was added to a stirring solution of 3.56 (17.12 g, 38.5 mmol) and EDCI (7.39 g, 38.5

mmol) in CH2Cl2. The reaction was stirred O/N at r.t., then concentrated and purified

through a plug of SiO2 (100% EtOAc) to afford 3.57d (13.78 g, 66% yield).

1H-NMR (300 MHz; CDCl3): δ 7.96 (d, J = 8.1 Hz, 1H), 7.86 (d, J = 7.1 Hz, 2H), 7.52

(t, J = 7.0 Hz, 2H), 7.44 (dd, J = 13.3, 5.5 Hz, 3H), 7.31 (t, J = 7.1 Hz, 1H), 6.96 (d, J =

6.7 Hz, 1H), 5.24-5.21 (m, 1H), 4.59-4.56 (m, 1H), 4.44 (q, J = 7.2 Hz, 1H), 3.89 (dt, J =

1.3, 0.7 Hz, 2H), 3.72 (s, 3H), 3.18 (d, J = 6.0 Hz, 2H), 1.40 (s, 9H); 13C-NMR (101

MHz; CDCl3): δ 171.9, 170.7, 155.9, 138.0, 135.0, 133.7, 130.8, 129.2, 126.6, 124.72,

124.70, 123.2, 119.6, 117.9, 113.5, 80.3, 62.4, 54.7, 52.6, 28.16, 28.00.

REF: TRW-II-107, TRW-II-178, TRW-II-253.

NSO2Ph

NHBocNH

O

CO2Me

OH

3.57d

149

Figure 5.23a. 1H NMR spectrum of compound 3.57d.

Figure 5.23b. 13C NMR spectrum of compound 3.57d.

8.98.9

2.02.0

3.33.3

2.22.2

1.31.3

1.21.2

0.90.9

0.90.9

1.31.3

3.03.0

2.42.4

2.02.0

1.01.0

ppm1 122334455667788-19.97

1.40

3.17

3.19

3.72

3.89

3.89

3.89

3.89

3.89

4.40

4.43

4.45

4.47

4.56

4.57

4.57

4.58

4.59

5.21

5.24

5.24

5.24

5.24

6.95

6.97

7.29

7.31

7.34

7.40

7.43

7.45

7.46

7.50

7.52

7.54

7.84

7.87

7.94

7.97

ppm0 05050100100150150200-253.79

28.0

028

.16

52.6

054

.73

62.4

2

80.2

9

113.

4511

7.94

119.

5612

3.22

124.

7012

4.72

126.

6412

9.18

130.

8513

3.71

134.

9713

7.97

155.

87

170.

7317

1.94

150

(3S,6S)-3-(hydroxymethyl)-6-((1-(phenylsulfonyl)-1H-indol-3-yl)methyl)piperazine-

2,5-dione (3.58d). The general cyclization procedure given for compound 3.58a was

repeated.

1H-NMR (300 MHz; DMSO-d6): δ 8.14-7.86 (m, 4H), 7.64-7.56 (m, 4H), 7.30-7.28 (m,

J = 5.5, 0.8 Hz, 2H), 5.13-5.13 (m, 1H), 4.38-4.03 (m, 1H), 3.74-3.60 (m, 2H), 3.43-3.33

(m, 2H), 3.15-2.85 (m, 2H); 13C-NMR (101 MHz; DMSO): δ 167.5, 167.3, 137.6, 137.2,

134.8, 131.4, 130.2, 126.9, 125.9, 123.6, 120.4, 118.5, 113.40, 113.31, 63.0, 57.3, 54.7,

50.9.

REF: TRW-II-063, TRW-II-186.

NSO2Ph

HNNH

OH

O

O

3.58d

151

Figure 5.24a. 1H NMR spectrum of compound 3.58d.

Figure 5.24b. 13C NMR spectrum of compound 3.58d.

2.02.0

2.32.3

2.02.0

1.11.1

0.70.7

1.91.9

4.14.1

3.93.9

ppm0 0112233445566778899-19.97

2.85

2.85

2.85

3.14

3.15

3.15

3.15

3.15

3.33

3.33

3.34

3.34

3.42

3.43

3.60

3.60

3.74

3.74

3.74

4.03

4.36

4.37

4.38

5.13

5.13

5.13

5.13

5.13

7.28

7.28

7.28

7.30

7.30

7.30

7.56

7.57

7.63

7.64

7.64

7.86

7.87

7.91

7.91

7.92

8.14

8.14

8.14

ppm0 05050100100150150-253.79

50.8

854

.72

57.3

2

63.0

0

113.

3111

3.40

118.

4712

0.45

123.

6312

5.94

126.

9313

0.17

131.

3613

4.81

137.

2213

7.60

167.

2716

7.50

152

Dipeptide (3.57e). Alcohol 3.57d (2.9 g, 5.3 mmol) was dissolved in DMF (13 mL), and

to it added imidazole (864 mg, 12.7 mmol) and TBSCl (2.08 g, 13.8 mmol). The reaction

stirred O/N at r.t., then was concentrated, dissolved in EtOAc, washed with H2O (3x),

washed with brine, dried, and concentrated to afford silylether 3.57e (3.14 g, 90% yield).

1H-NMR (300 MHz; DMSO-d6): δ 8.14 (d, J = 7.8 Hz, 1H), 7.85 (t, J = 7.9 Hz, 3H),

7.65 (t, J = 7.2 Hz, 2H), 7.58-7.50 (m, 3H), 7.33-7.20 (m, 2H), 7.00 (d, J = 8.8 Hz, 1H),

4.46-4.40 (m, 1H), 4.34-4.30 (m, 1H), 3.86 (dd, J = 10.3, 4.7 Hz, 1H), 3.74 (dd, J = 10.3,

4.9 Hz, 1H), 3.60 (s, 3H), 3.05-2.99 (m, 1H), 2.89-2.80 (m, 1H), 1.25 (s, 9H), 0.82 (s,

9H), 0.00 (d, J = 3.2 Hz, 6H); 13C-NMR (101 MHz; CDCl3): δ 170.6, 170.2, 138.2,

135.1, 133.7, 130.8, 129.2, 126.7, 124.9, 124.6, 123.3, 119.6, 117.6, 113.6, 63.3, 54.2,

52.4, 28.20, 28.10, 25.70, 25.61, 18.1, -5.63, -5.77.

REF: TRW-II-196, TRW-II-222, TRW-II-254.

NSO2Ph

NHBocNH

O

CO2Me

OTBS

3.57e

153

Figure 5.25a. 1H NMR spectrum of compound 3.57e.

Figure 5.25b. 13C NMR spectrum of compound 3.57e.

6.36.3

8.98.9

8.58.5

1.21.2

1.21.2

3.43.4

1.21.2

1.31.3

1.01.0

1.21.2

1.01.0

2.42.4

3.23.2

2.32.3

3.33.3

1.01.0

ppm0 011223344556677889-19.97

-0.0

00.

01

0.82

1.25

2.80

2.84

2.84

2.86

2.88

2.89

2.99

2.99

3.00

3.01

3.04

3.04

3.05

3.05

3.05

3.60

3.71

3.73

3.75

3.76

3.84

3.86

3.87

3.89

4.30

4.30

4.31

4.31

4.32

4.33

4.33

4.33

4.34

4.34

4.34

4.34

4.40

4.42

4.42

4.44

4.46

4.46

6.99

7.02

7.20

7.21

7.23

7.25

7.26

7.27

7.28

7.30

7.30

7.32

7.33

7.50

7.53

7.55

7.58

7.58

7.62

7.65

7.67

7.83

7.85

7.88

8.13

8.16

ppm0 05050100100150150-253.79

-5.7

7-5

.63

18.0

525

.61

25.7

028

.10

28.2

0

52.4

254

.19

63.3

0

113.

6011

7.62

119.

5712

3.28

124.

5912

4.87

126.

7012

9.19

130.

7513

3.69

135.

1213

8.16

170.

2017

0.63

154

Dioxopiperazine (3.58e). Dipeptide 3.57e (500 mg) was transferred to a microwave tube,

then heated neat in the microwave at maximum power to a safe temperature of 180 °C

(microwave set to ‘open-vessel’). After cooling, the residue was suspended in CH2Cl2,

transferred to a vial, and concentrated to afford dioxopiperazine 3.57e (2.47 g over 6

batches, 98%).

1H-NMR (300 MHz; DMSO-d6): δ 7.92-7.83 (m, 4H), 7.64 (t, J = 3.7 Hz, 2H), 7.58-7.52

(m, 4H), 7.33-7.28 (m, 1H), 7.25-7.20 (m, 1H), 4.15-4.05 (m, 1H), 3.84-3.77 (m, 1H),

3.61-3.43 (m, 2H), 3.16-2.94 (m, 2H), 0.79 (s, 9H), -0.02 (s, 3H), -0.06 (s, 3H).

REF: TRW-II-226, TRW-II-255.

NSO2Ph

HNNH

OTBS

O

O

3.58e

155

Figure 5.26a. 1H NMR spectrum of compound 3.58e.

3.53.5

3.03.0

9.69.6

2.32.3

2.22.2

0.90.9

1.01.0

1.11.1

1.21.2

3.93.9

1.91.9

4.24.2

ppm0 01122334455667788-19.97

-0.0

6-0

.02

0.79

2.94

3.00

3.00

3.01

3.01

3.02

3.03

3.03

3.03

3.08

3.10

3.11

3.12

3.13

3.13

3.14

3.16

3.43

3.45

3.47

3.53

3.54

3.54

3.56

3.57

3.57

3.61

4.05

4.05

4.05

4.05

4.06

4.06

4.06

4.07

4.07

4.08

4.08

4.15

4.15

7.20

7.22

7.23

7.23

7.24

7.25

7.25

7.25

7.28

7.28

7.30

7.33

7.52

7.53

7.53

7.55

7.56

7.57

7.58

7.63

7.64

7.66

7.83

7.84

7.85

7.85

7.87

7.90

7.90

7.92

156

Dipeptide (5.1). Alcohol 3.57c (2.0 g, 4.9 mmol) was dissolved in DMF (12 mL), and to

it added imidazole (802 mg, 11.8 mmol) and TBSCl (1.92 g, 12.7 mmol). The reaction

stirred O/N at r.t., then was concentrated, dissolved in EtOAc, washed with H2O (3x),

washed with brine, dried, and concentrated to afford silylether 5.1 (2.38 g, 93% yield).

1H-NMR (300 MHz; CDCl3): δ 8.18 (bs, J = 0.4 Hz, 1H), 7.66 (d, J = 7.6 Hz, 1H), 7.34

(d, J = 8.0 Hz, 1H), 7.20-7.18 (m, 1H), 7.16-7.11 (m, 2H), 6.55-6.52 (m, 1H), 5.19-5.16

(m, 1H), 4.54 (broad, J = 7.0, 4.1 Hz, 2H), 3.95 (dd, J = 10.1, 2.7 Hz, 1H), 3.66 (s, 3H),

3.62-3.58 (m, 1H), 3.31-3.30 (m, 1H), 3.21 (dd, J = 14.6, 6.9 Hz, 1H), 1.42 (s, 9H), 0.77

(s, 9H), -0.07 (s, 3H), -0.10 (s, 3H).

REF: TRW-III-196.

NH

NHBocNH

O

CO2Me

OTBS

5.1

157

Figure 5.27a. 1H NMR spectrum of compound 5.1.

3.13.1

3.03.0

9.49.4

9.49.4

1.21.2

1.01.0

0.80.8

3.33.3

1.11.1

1.91.9

0.80.8

1.01.0

2.22.2

0.90.9

1.21.2

1.11.1

1.01.0

ppm-1 -1001122334455667788-19.97

-0.1

0-0

.07

0.77

1.42

3.17

3.19

3.22

3.24

3.30

3.30

3.31

3.31

3.31

3.31

3.58

3.58

3.59

3.59

3.59

3.59

3.60

3.60

3.61

3.61

3.61

3.61

3.62

3.62

3.66

3.93

3.93

3.96

3.97

4.52

4.53

4.54

4.56

5.16

5.16

5.16

5.16

5.17

5.18

5.18

5.18

5.18

5.18

5.19

6.52

6.52

6.54

6.54

6.54

6.55

7.11

7.13

7.13

7.15

7.16

7.18

7.18

7.20

7.33

7.36

7.64

7.67

8.18

8.18

158

(3S,6S)-3-((1H-indol-3-yl)methyl)-6-(((tert-

butyldimethylsilyl)oxy)methyl)piperazine-2,5-dione (5.2). The general cyclization

procedure given for compound 3.58a was repeated.

1H-NMR (300 MHz; DMSO-d6): δ 10.88 (m, 1H), 7.95 (s, 1H), 7.60-7.48 (m, 1H), 7.29

(t, J = 7.8 Hz, 1H), 7.05-6.93 (m, 2H), 4.09-3.97 (m, 1H), 3.82-3.70 (m, 1H), 3.52-3.43

(m, 1H), 3.27-3.14 (m, 2H), 2.79 (d, J = 47.6 Hz, 1H), 0.81 (s, 9H), -0.02--0.05 (m, 6H);

13C-NMR (101 MHz; DMSO): δ 167.6, 165.5, 136.5, 127.9, 124.4, 121.3, 118.96,

118.77, 111.7, 109.5, 65.4, 57.5, 55.9, 31.5, 26.33, 26.24, 26.11, 26.05, 25.99, 18.7, -5.0.

REF: TRW-II-187.

NH

HNNH

OTBS

O

O

5.2

159

Figure 5.28a. 1H NMR spectrum of compound 5.2.

Figure 5.28b. 13C NMR spectrum of compound 5.2.

5.45.4

6.86.8

0.80.8

1.91.9

0.70.7

0.80.8

0.90.9

2.32.3

1.01.0

1.01.0

1.01.0

0.90.9

ppm0 0224466881010-19.97

-0.0

5-0

.03

-0.0

2

0.81

2.71

2.87

3.14

3.15

3.19

3.27

3.27

3.43

3.44

3.47

3.48

3.48

3.49

3.49

3.51

3.52

3.52

3.70

3.70

3.70

3.71

3.78

3.78

3.78

3.78

3.78

3.81

3.82

3.97

4.08

4.08

4.08

4.08

4.09

6.93

6.95

6.96

6.96

6.98

7.01

7.01

7.05

7.05

7.27

7.29

7.32

7.48

7.51

7.57

7.59

7.60

7.95

10.8

510

.90

ppm0 05050100100150150-253.79

-5.0

1

18.6

825

.99

26.0

526

.11

26.2

426

.33

31.5

3

55.9

457

.48

65.4

0

109.

5111

1.74

118.

7711

8.96

121.

2712

4.44

127.

87

136.

51

165.

5116

7.57

160

tert-butyl 3-(((2S,5S)-5-(((tert-butyldimethylsilyl)oxy)methyl)-3,6-dioxopiperazin-2-

yl)methyl)-1H-indole-1-carboxylate. The general cyclization procedure given for

compound 3.58a was repeated.

1H-NMR (300 MHz; CDCl3): δ 8.01 (d, J = 8.2 Hz, 1H), 7.47 (dd, J = 7.7, 0.4 Hz, 1H),

7.35 (s, 1H), 7.23-7.10 (m, 2H), 4.16-4.12 (m, 1H), 3.87-3.84 (m, 3H), 3.63 (dd, J = 10.1,

3.1 Hz, 1H), 3.31 (dd, J = 10.1, 5.8 Hz, 2H), 3.05 (dd, J = 14.4, 8.8 Hz, 1H), 1.54 (s, 9H),

0.75 (s, 9H), -0.09 (s, 6H); 13C-NMR (101 MHz; CDCl3): δ 173.3, 171.6, 135.4, 130.61,

130.58, 130.50, 128.6, 124.8, 121.0, 120.2, 89.7, 70.5, 62.7, 60.5, 54.7, 54.5, 36.5, 33.7,

31.4, 24.1, 0.06, -0.00.

REF: TRW-III-187.

NBoc

HNNH

OTBS

O

O

5.3

161

Figure 5.29a. 1H NMR spectrum of compound 5.3.

Figure 5.29b. 13C NMR spectrum of compound 5.3.

5.45.4

9.19.1

9.39.3

1.11.1

2.22.2

1.11.1

2.92.9

1.11.1

2.02.0

1.01.0

1.11.1

1.01.0

ppm0 01122334455667788-14.99

-0.0

9

0.75

1.54

3.02

3.05

3.06

3.09

3.29

3.31

3.32

3.34

3.61

3.62

3.64

3.65

3.84

3.84

3.85

3.85

3.86

3.87

4.12

4.13

4.15

4.15

4.16

7.10

7.11

7.13

7.15

7.16

7.18

7.18

7.21

7.23

7.23

7.35

7.46

7.46

7.48

7.49

7.99

8.02

ppm0 05050100100150150-253.79

-0.0

00.

06

24.0

7

31.4

533

.68

36.5

5

54.5

154

.72

60.5

362

.74

70.5

0

89.7

2

120.

1912

0.98

124.

7912

8.58

130.

5013

0.58

130.

6113

5.43

171.

5617

3.30

162

N-Boc-L-tryptophan-N’-SO2Ph (3.56). Compound 3.59 (14.8 g, 48.7 mmol) was

dissolved in THF (100 mL) and the resulting solution cooled to -78 °C. LHMDS (146

mL, 146.1 mmol) was added slowly over 5 minutes. The reaction was stirred at -78 °C

for 1 h, then PhSO2Cl (7.5 mL, 58.4 mmol) added in one portion. After stirring an

additional 2 h at -78 °C, the reaction was quenched by addition of 1:1 AcOH:EtOAc then

allowed to warm to r.t. The suspension was diluted with 1M HCl and the product

extracted in EtOAc. The combined organic layers were dried and concentrated to afford

crude 3.56, purified by SiO2 chromatography (5% AcOH, 45% hexanes, 50% CH2Cl2) to

afford pure 3.56 (17.12 g, 79% yield).

1H-NMR (300 MHz; DMSO-d6): δ 12.64-12.63 (m, 1H), 7.90-7.87 (m, 2H), 7.61 (t, J =

7.8 Hz, 2H), 7.56-7.50 (m, 2H), 7.25-7.20 (m, 2H), 7.16-7.13 (m, 2H), 4.20-4.17 (m, 1H),

3.11 (dd, J = 14.8, 4.2 Hz, 1H), 2.95 (dd, J = 14.8, 10.0 Hz, 1H), 1.31 (s, 9H).

REF: TRW-II-176, TRW-II-251.

NSO2Ph

CO2H

NHBoc

3.56

163

Figure 5.30a. 1H NMR spectrum of compound 3.56.

9.49.4

1.01.0

1.01.0

0.90.9

2.42.4

2.42.4

2.32.3

2.22.2

2.32.3

0.90.9

ppm0 02244668810101212-19.97

1.31

2.91

2.94

2.96

2.99

3.08

3.10

3.13

3.15

4.17

4.19

4.19

4.19

4.20

7.13

7.13

7.16

7.20

7.22

7.23

7.25

7.25

7.27

7.27

7.32

7.50

7.53

7.56

7.56

7.59

7.61

7.64

7.87

7.88

7.89

7.90

12.6

312

.63

12.6

312

.64

12.6

4

164

Bromopyrroloindoline (3.55e). A solution of dioxopiperazine 3.58e (2.47 g, 4.68 mmol)

in MeCN (50 mL) was cooled to 0 °C and to it added bromine (0.962 mL, 18.7 mmol)

dropwise. After stirring 5 minutes, 1:1 Na2S2O3:NaHCO3 was added and the product

extracted in EtOAc. The combined organic layers were washed with brine, dried over

Na2SO4 and concentrated. Purification by SiO2 chromatgraphy (10/40/50 iPrOH/hexanes/

CH2Cl2) afforded pure pyrroloindoline 3.55e in 38% yield (1.09 g).

1H-NMR (300 MHz; DMSO-d6): δ 8.06 (s, 1H), 7.87-7.84 (m, 2H), 7.66-7.53 (m, 4H),

7.42 (d, J = 2.3 Hz, 2H), 7.27-7.22 (m, 1H), 6.46 (s, 1H), 4.05 (d, J = 1.0 Hz, 1H), 3.77-

3.75 (m, 2H), 3.73-3.68 (m, 2H), 3.60-3.56 (m, 2H), 1.01 (d, J = 6.1 Hz, 9H), -0.02 (s,

6H).

REF: TRW-II-228, TRW-II-256.

NSO2Ph

NNH

O

O

OTBS

Br

3.55e

165

Figure 5.31a. 1H NMR spectrum of compound 3.55e.

1.81.8

1.81.8

1.81.8

1.21.2

0.80.8

0.90.9

1.81.8

4.34.3

2.22.2

1.01.0

ppm0 01122334455667788-19.97

-0.0

2

1.00

1.02

3.56

3.59

3.59

3.60

3.60

3.68

3.68

3.70

3.70

3.71

3.71

3.73

3.75

3.76

3.77

4.05

4.05

6.46

7.22

7.23

7.23

7.24

7.24

7.26

7.27

7.42

7.43

7.53

7.55

7.58

7.60

7.62

7.64

7.66

7.84

7.85

7.87

7.87

8.06

166

N-Me-bromopyrroloindoline (3.60). Dioxopiperazine 3.55e (2.09 g, 3.4 mmol) was

dissolved in acetone (50 mL). Potassium carbonate (11.73 g, 85 mmol) and MeI (27.5

mL, 442 mmol) were added, the flask covered with foil, and the reaction allowed to stir at

r.t. under Ar for 5 d. The crude reaction mixture was concentrated, partitioned between

EtOAc and H2O (200 mL each), and the product extracted in EtOAc. The combined

organic layers were dried and concentrated. SiO2 chromatography (10/40/50

iPrOH/hexanes/CH2Cl2) gave the pure title compound (1.69 g, 98% yield).

1H-NMR (300 MHz; CDCl3): δ 7.89-7.71 (m, J = 1.6 Hz, 2H), 7.69 (d, J = 8.2 Hz, 1H),

7.58-7.52 (m, 1H), 7.44 (ddd, J = 8.4, 6.8, 1.4 Hz, 2H), 7.38-7.32 (m, 2H), 7.21 (d, J =

7.0 Hz, 1H), 6.54 (d, J = 28.9 Hz, 1H), 4.08-3.78 (m, 4H), 3.17 (ddd, J = 28.8, 12.6, 5.2

Hz, 1H), 2.98 (s, 3H), 2.89-2.76 (m, 1H).

REF: TRW-II-235, TRW-II-258.

NSO2Ph

NNMe

O

O

OH

Br

3.60

167

Figure 5.32a. 1H NMR spectrum of compound 3.60.

1.31.3

3.23.2

1.31.3

4.64.6

1.01.0

0.90.9

1.91.9

2.32.3

1.01.0

0.90.9

2.02.0

ppm0 01122334455667788-19.97

2.76

2.77

2.78

2.81

2.82

2.83

2.87

2.89

2.98

3.09

3.11

3.14

3.15

3.19

3.21

3.23

3.25

3.78

3.79

3.82

3.82

3.83

3.90

3.91

3.92

3.92

3.93

3.94

3.95

3.96

3.96

3.97

3.97

3.99

3.99

4.00

4.00

4.01

4.02

4.03

4.04

4.05

4.07

4.08

6.48

6.58

7.22

7.26

7.32

7.33

7.33

7.34

7.34

7.35

7.35

7.36

7.36

7.36

7.38

7.38

7.41

7.42

7.43

7.44

7.44

7.46

7.46

7.52

7.53

7.53

7.55

7.55

7.56

7.58

7.68

7.71

7.85

7.85

7.86

7.86

7.87

7.87

7.89

7.89

168

OTBS-N-Me-bromopyrroloindoline (3.61). The same general procedure for TBS

protection was followed as for 3.57e. Instead of carrying the crude product forward as

before, it was necessary for the subsequent radical bromination to purify the product by

SiO2 chromatography (20-60% EtOAc in hexanes) to afford 3.61 (47% yield).

1H-NMR (300 MHz; CDCl3): δ 7.89 (dd, J = 8.1, 1.0 Hz, 2H), 7.70 (dd, J = 7.9, 0.6 Hz,

1H), 7.56-7.53 (m, 1H), 7.48-7.43 (m, 2H), 7.35-7.32 (m, 2H), 7.21-7.16 (m, 1H), 6.63

(s, 1H), 4.28 (dd, J = 10.2, 1.3 Hz, 1H), 4.06 (s, 1H), 3.94 (dd, J = 10.1, 2.7 Hz, 1H), 3.78

(dd, J = 12.4, 4.5 Hz, 1H), 3.09 (dd, J = 12.0, 4.5 Hz, 1H), 2.91 (s, 3H), 2.73 (t, J = 12.2

Hz, 1H), 0.89 (s, 9H), 0.10 (s, 6H).

REF: TRW-II-236, TRW-II-242.

NSO2Ph

NNMe

O

O

OTBS

Br

3.61

169

Figure 5.33a. 1H NMR spectrum of compound 3.61.

6.86.8

10.410.4

1.11.1

3.33.3

1.21.2

1.11.1

1.31.3

1.21.2

1.11.1

1.01.0

1.21.2

2.32.3

2.42.4

1.21.2

0.90.9

2.02.0

ppm0 01122334455667788-19.97

0.10

0.89

2.69

2.73

2.78

2.91

3.06

3.08

3.10

3.12

3.75

3.77

3.79

3.81

3.92

3.93

3.95

3.96

4.06

4.26

4.27

4.30

4.30

6.63

7.16

7.18

7.18

7.18

7.19

7.21

7.21

7.32

7.32

7.32

7.34

7.34

7.35

7.35

7.35

7.43

7.45

7.48

7.48

7.53

7.54

7.54

7.55

7.56

7.56

7.69

7.69

7.72

7.72

7.87

7.88

7.90

7.90

170

Diol (3.62). To a solution of Br-dioxopiperazine 3.61 (710 mg, 1.14 mmol) in CCl4 (40

mL) was added NBS (406 mg, 2.28 mmol) and V-70 (89 mg, 0.29 mmol). The resulting

suspension was stirred 8 h. at r.t., then filtered through a cotton plug, the solid washed

with CCl4, and the combined filtrate concentrated. The resulting residue was dissolved in

1:1 MeCN:pH 7 phosphate buffer (58 mL) and stirred 3 h. at r.t. The product was

extracted in EtOAc (3x), washed with brine, dried, and concentrated to afford the diol in

97% yield (720 mg), carried forward to the next step as the crude material.

REF: TRW-II-244, 245, TRW-II-368, 370.

NSO2Ph

NNMe

O

O

OTBS

Br

OH

OH

3.62

171

Epidithiodioxopiperazine (3.63). Hydrogen sulfide (ca. 8 mL) gas was condensed in a

sealed tube capped with a rubber septum at -78 °C. Diol 3.62 (125 mg, 0.19 mmol) and

BF3·OEt2 (0.119 mL, 0.95 mmol) were added as a solution in CH2Cl2 (7.6 mL). The

rubber septum was removed and the sealed tube capped with a teflon cap and placed

behind a blast shield. The reaction mixture stirred at r.t. for 90 min., recooled to -78 °C,

uncapped and sealed with a rubber septum, then allowed to warm to r.t. with 3 NaOH and

1 bleach trap in place as the gas vented. The resulting solution was diluted with EtOAc,

washed with sat’d NH4Cl, and the product extracted in EtOAc. Iodine (100 mg, 2 eq) was

added to the EtOAc extracts and the mixture allowed to stir 1 min. 10% Na2S2O3 was

added and the product extracted in EtOAc, dried, and concentrated. Purification by SiO2

chromatography (1:1 hexanes:EtOAc) afforded pure epidithiodioxopiperazine 3.63 in

42% yield (44.9 mg).

1H-NMR (300 MHz; CDCl3): δ 7.78 (dd, J = 8.4, 1.1 Hz, 2H), 7.64 (d, J = 8.2 Hz, 1H),

7.52-7.49 (m, 1H), 7.41-7.36 (m, 4H), 7.28-7.26 (m, 1H), 6.49 (s, 1H), 4.33 (d, J = 12.8

Hz, 1H), 4.18 (d, J = 12.7 Hz, 1H), 3.85 (d, J = 15.3 Hz, 1H), 3.21 (d, J = 15.3 Hz, 1H),

3.13 (s, 3H); 13C-NMR (101 MHz; CDCl3): δ 176.9, 163.7, 134.83, 134.82, 134.3, 133.8,

132.8, 131.23, 131.22, 131.0, 123.9, 114.6, 72.4, 66.2, 37.6, 31.3, 26.9, 23.7, 20.0;

HRMS (ESI-APCI): [M+H]+ 567.9670 calcd for C21H19BrN3O5S3, found: 567.9641. REF:

TRW-II-246, 247, TRW-II-372, 373.

NSO2Ph

NNMe

O

O

OH

BrSS

3.63

172

Figure 5.34a. 1H NMR spectrum of compound 3.63.

Figure 5.34b. 13C NMR spectrum of compound 3.63.

3.63.6

1.31.3

1.21.2

1.31.3

1.01.0

1.01.0

1.31.3

3.83.8

1.21.2

1.01.0

1.81.8

ppm0 0112233445566778-19.973.

133.

193.

24

3.83

3.88

4.16

4.20

4.32

4.36

6.49

7.25

7.25

7.26

7.27

7.28

7.33

7.34

7.36

7.38

7.38

7.40

7.40

7.41

7.43

7.49

7.49

7.49

7.50

7.51

7.52

7.52

7.63

7.65

7.77

7.77

7.80

7.80

ppm0 05050100100150150200200-253.79

20.0

223

.67

26.8

631

.26

37.6

2

66.2

0

72.3

9

114.

6212

3.92

130.

9913

1.22

131.

2313

2.81

133.

8413

4.33

134.

8213

4.83

163.

66

176.

95

173

N-Boc-L-tryptophan (3.59). To a solution of L-tryptophan (50 g, 245 mmol) in 1:1

THF:H2O (700 mL) was added NaOH (10.8 g, 270 mmol) and Boc2O (58.9 g, 270

mmol). The resulting solution was stirred O/N at r.t. The volume was reduced and the

product extracted in CH2Cl2. The aqueous layer was acidified with 1M HCl to pH 4, and

the product extracted in CH2Cl2. The combined organic layers were dried over Na2SO4

and concentrated to afford 3.59 (70.23 g, 94% yield).

1H-NMR (300 MHz; CDCl3): δ 7.50 (d, J = 7.6 Hz, 1H), 7.26 (d, J = 7.9 Hz, 1H), 7.09-

6.98 (m, 2H), 6.96 (s, 1H), 4.45 (t, J = 5.4 Hz, 1H), 3.28-3.17 (m, 2H), 1.31 (s, 9H).

REF: TRW-II-252.

NH

CO2H

NHBoc

3.59

174

Figure 5.35a. 1H NMR spectrum of compound 3.59.

9.19.1

2.02.0

0.80.8

1.11.1

1.91.9

1.31.3

1.01.0

ppm0 011223344556677-19.97

1.31

3.17

3.19

3.22

3.23

3.26

3.26

3.27

3.27

3.28

3.28

4.43

4.45

4.47

6.96

6.98

7.01

7.01

7.03

7.04

7.06

7.06

7.08

7.09

7.25

7.28

7.49

7.51

175

N-Boc-L-tryptophan-OMe (3.34). To a solution of N-Boc-L-tryptophan (30 g, 98.7

mmol) in DMF (200 mL) at 0 °C was added MeI (9.2 mL, 148 mmol) and KHCO3 (19.7

g, 197 mmol). After stirring O/N at r.t., the mixture was diluted with ether, washed with

H2O (2x), 1M KHSO4 (1x), and brine (1x). The combined organic layers were dried and

concentrated to afford the title compound (29.86 g, 95% yield).

1H-NMR (300 MHz; CDCl3): δ 8.10-8.08 (m, 1H), 7.55 (d, J = 7.9 Hz, 1H), 7.36 (d, J =

8.0 Hz, 1H), 7.22-7.09 (m, 2H), 7.01 (d, J = 2.4 Hz, 1H), 5.09-5.06 (m, 1H), 4.68-4.62

(m, 1H), 3.68 (s, 3H), 3.29 (d, J = 5.2 Hz, 2H), 1.43 (s, 9H).

REF: TRW-II-273, TRW-II-285, TRW-II-349.

NH

CO2Me

NHBoc

3.34

176

Figure 5.36a. 1H NMR spectrum of compound 3.34.

9.49.4

1.81.8

2.92.9

0.90.9

0.80.8

0.90.9

1.91.9

1.11.1

1.11.1

1.01.0

ppm0 01122334455667788-19.97

1.43

3.28

3.30

3.68

4.62

4.62

4.62

4.62

4.62

4.63

4.64

4.64

4.64

4.65

4.65

4.65

4.66

4.66

4.66

4.66

4.67

4.68

4.68

4.68

5.06

5.06

5.06

5.08

5.09

7.01

7.01

7.09

7.10

7.12

7.14

7.15

7.17

7.17

7.19

7.19

7.20

7.22

7.22

7.34

7.37

7.54

7.57

8.08

8.08

8.08

8.08

8.08

8.09

8.09

8.09

8.10

177

N-Boc-L-tryptophan-OMe-N’-SO2Ph (3.64). N-Boc-L-tryptophan-OMe (10.0 g, 31.4

mmol), NaOH (3.77 g, 94.2 mmol), and Bu4NHSO4 (544 mg, 1.6 mmol) were suspended

in CH2Cl2 (150 mL), then PhSO2Cl (4.8 mL, 37.7 mmol) added dropwise. After stirring 6

h. at r.t., aqueous NH4Cl (300 mL) and EtOAc (300 mL) were added and the product

extracted in EtOAc (3x). The combined organic extracts were dried and concentrated to

afford the title compound (14.4 g, >99% yield).

1H-NMR (300 MHz; CDCl3): δ 7.97 (d, J = 8.2 Hz, 1H), 7.84 (d, J = 7.5 Hz, 2H), 7.56-

7.50 (m, 1H), 7.47-7.44 (m, 2H), 7.42 (d, J = 7.2 Hz, 1H), 7.36 (s, 1H), 7.31-7.20 (m,

2H), 5.05 (dt, J = 7.9, 0.6 Hz, 1H), 4.66-4.59 (m, 1H), 3.61 (s, 3H), 3.27-3.11 (m, 2H),

1.44 (s, 9H).

REF: TRW-II-263, TRW-II-275, TRW-II-350.

NSO2Ph

CO2Me

NHBoc

3.64

178

Figure 5.37a. 1H NMR spectrum of compound 3.64.

8.88.8

1.91.9

2.72.7

0.90.9

0.90.9

2.12.1

1.01.0

0.70.7

2.12.1

1.11.1

1.81.8

1.01.0

ppm1 122334455667788-19.97

1.44

3.11

3.11

3.11

3.12

3.12

3.12

3.13

3.13

3.13

3.14

3.15

3.15

3.15

3.15

3.15

3.16

3.17

3.17

3.18

3.18

3.19

3.19

3.20

3.20

3.20

3.22

3.25

3.25

3.25

3.25

3.26

3.27

3.27

3.27

3.27

3.27

3.61

4.59

4.59

4.59

4.59

4.60

4.60

4.60

4.61

4.62

4.63

4.63

4.63

4.63

4.64

4.65

4.65

4.65

4.66

5.03

5.04

5.04

5.06

5.06

5.06

7.20

7.20

7.23

7.25

7.25

7.26

7.26

7.28

7.29

7.31

7.31

7.36

7.41

7.43

7.44

7.44

7.44

7.44

7.46

7.47

7.47

7.50

7.50

7.51

7.52

7.53

7.54

7.55

7.56

7.83

7.85

7.95

7.98

179

Bromopyrroloindoline (3.65). To a solution of N-Boc-L-tryptophan-OMe-N’-SO2Ph (42

g, 91.5 mmol) in CH2Cl2 (1000 mL) was added NBS (16.29 g, 91.5 mmol) and PPTS

(22.97 g, 91.5 mmol). The reaction was stirred O/N at r.t., then washed with 10%

NaHCO3 and 10% Na2S2O4 (1:1), and the organic extract concentrated to afford 3.65 in

quantitative yield.

1H-NMR (300 MHz; CDCl3): δ 7.83-7.79 (bs, 2H), 7.57 (d, J = 8.0 Hz, 1H), 7.46 (ddd, J

= 8.4, 6.4, 1.6 Hz, 1H), 7.34 (td, J = 7.7, 1.4 Hz, 3H), 7.27-7.25 (m, 1H), 7.17 (td, J = 7.5,

1.0 Hz, 1H), 6.33-6.31 (bs, 1H), 3.85-3.80 (m, 1H), 3.73 (s, 3H), 3.06 (dd, J = 12.6, 5.9

Hz, 1H), 2.81-2.73 (m, 1H), 1.54-1.53 (bs, 9H); 13C-NMR (101 MHz; CDCl3): δ 171.9,

138.0, 133.8, 133.4, 130.9, 129.2, 128.6, 128.3, 127.3, 126.6, 124.9, 124.3, 123.9, 123.3,

113.6, 86.7, 80.0, 59.4, 52.4, 28.26, 28.17, 27.8, 21.0, 14.2.

REF: TRW-II-264, TRW-II-277, TRW-II-351.

NSO2Ph

NBocCO2Me

Br

3.65

180

Figure 5.38a. 1H NMR spectrum of compound 3.65.

Figure 5.38b. 13C NMR spectrum of compound 3.65.

9.39.3

1.11.1

1.11.1

3.03.0

1.01.0

0.90.9

1.11.1

1.91.9

3.13.1

1.31.3

1.01.0

1.91.9

ppm0 0112233445566778-19.97

1.53

1.53

1.53

1.53

1.54

1.54

2.73

2.73

2.77

2.77

2.80

2.81

3.03

3.05

3.08

3.10

3.73

3.80

3.80

3.82

3.82

3.83

3.85

6.31

6.31

6.31

6.31

6.31

6.31

6.32

6.32

6.32

6.32

6.32

6.33

6.33

7.14

7.15

7.17

7.17

7.19

7.20

7.25

7.25

7.25

7.26

7.26

7.27

7.32

7.32

7.34

7.35

7.37

7.37

7.43

7.44

7.46

7.46

7.47

7.48

7.49

7.56

7.59

7.79

7.79

7.79

7.80

7.80

7.80

7.80

7.80

7.81

7.81

7.81

7.82

7.82

7.82

7.82

7.83

ppm0 05050100100150150200200-253.79

28.1

728

.26

52.2

852

.43

59.4

4

76.7

677

.08

77.4

0

113.

6412

3.28

123.

8912

4.90

126.

6112

8.28

128.

5912

9.21

130.

8713

3.39

133.

76

171.

91

181

Indole (3.66). Bromopyrroloindoline 3.65 (16.41 g, 30.6 mmol) was dissolved in

CH3CN, and to the resulting solution added N-Boc-L-tryptophan-OMe (6.49 g, 20.4

mmol). The solution was cooled to 0 °C, then KOtBu (4.57 g, 40.8 mmol) added. After

stirring 1 h at 0 °C, the reaction was quenched with NaHCO3, extracted in CH2Cl2,

washed with brine, dried, and concentrated. Purification by SiO2 chromatography (40%

EtOAc in hexanes) afforded 3.66 in 34% yield (5.36 g).

1H-NMR (300 MHz; CDCl3): δ 7.85-7.79 (m, 1H), 7.85-7.79 (m, 1H), 7.60-7.43 (m,

2H), 7.60-7.43 (m, 2H), 7.34 (t, J = 7.7 Hz, 3H), 7.34 (t, J = 7.7 Hz, 3H), 7.22-7.01 (m,

7H), 7.22-7.01 (m, 7H), 6.85 (s, 1H), 6.85 (s, 1H), 6.31 (bs, 1H), 6.31 (s, 1H), 5.06 (bs,

1H), 5.06 (s, 1H), 4.64-4.61 (m, 1H), 4.64-4.61 (m, 1H), 3.73 (s, 3H), 3.73 (s, 3H), 3.68

(s, 3H), 3.68 (s, 3H), 3.28-3.21 (m, 2H), 3.28-3.21 (m, 2H), 3.07 (dd, J = 12.6, 5.9 Hz,

1H), 3.07 (dd, J = 12.6, 5.9 Hz, 1H), 2.77 (t, J = 11.6 Hz, 1H), 2.77 (t, J = 11.6 Hz, 1H),

1.58-1.55 (bs, 9H), 1.58-1.55 (m, 9H), 1.43 (s, 9H), 1.43 (s, 9H); 13C-NMR (101 MHz;

CDCl3): δ 172.4, 161.2, 154.9, 143.2, 133.7, 132.9, 132.2, 131.61, 131.58, 131.52, 130.5,

128.9, 128.5, 128.1, 127.0, 126.46, 126.35, 125.2, 124.1, 122.4, 121.82, 121.76, 120.24,

120.17, 119.57, 119.43, 114.8, 82.2, 59.44, 59.36, 54.4, 53.3, 52.1, 51.6, 37.9, 37.7,

28.18, 28.14, 28.08, 28.01, 27.99, 27.97, 27.94, 27.93, 27.91, 27.79; HRMS (ESI-

APCI+): [M+Na]+ 797.2832 calcd for C40H46N4NaO10S, found: 797.2829.

REF: TRW-II-266, TRW-II-278, TRW-II-352.

NSO2Ph

NBoc

NCO2Me

MeO2C

BocHN

3.66

182

Figure 5.39a. 1H NMR spectrum of compound 3.66.

Figure 5.39b. 13C NMR spectrum of compound 3.66.

9.59.5

8.88.8

1.01.0

1.21.2

2.42.4

2.72.7

3.43.4

1.01.0

0.90.9

0.90.9

0.70.7

6.96.9

2.62.6

1.81.8

0.90.9

ppm0 01122334455667788-19.97

1.43

1.55

1.55

1.55

1.55

1.56

1.56

1.58

1.58

2.73

2.77

2.81

3.04

3.06

3.08

3.10

3.21

3.21

3.21

3.23

3.23

3.26

3.28

3.28

3.68

3.73

4.61

4.63

4.63

4.64

5.06

6.31

6.85

7.01

7.01

7.06

7.06

7.10

7.10

7.10

7.10

7.14

7.15

7.17

7.19

7.21

7.21

7.22

7.32

7.34

7.37

7.43

7.44

7.46

7.46

7.46

7.49

7.49

7.51

7.54

7.54

7.54

7.56

7.56

7.59

7.59

7.60

7.79

7.80

7.80

7.82

7.83

7.85

7.85

ppm50 50100100150150-253.79

27.7

927

.91

27.9

327

.94

27.9

727

.99

28.0

128

.08

28.1

428

.18

37.7

137

.92

51.5

952

.15

53.3

454

.35

59.3

659

.44

82.2

2

114.

7611

9.43

119.

5712

0.17

120.

2412

1.76

121.

8212

2.43

124.

1012

5.16

126.

3512

6.46

126.

9912

8.09

128.

4912

8.93

130.

5013

1.52

131.

5813

1.61

132.

2013

2.91

133.

6514

3.17

154.

8816

1.16

172.

42

183

Diacid (3.67). To a solution of 3.66 (5.35 g, 6.9 mmol) in 2:1 THF:H2O (24 mL) was

added LiOH (828 mg, 34.5 mmol). The reaction stirred O/N before acidifying with 10%

KHSO4. The product was extracted in EtOAc, and the combined organic extracts washed

with brine, dried, and concentrated. SiO2 chromatography (1:1 hexanes:EtOAc) gave the

pure diacid (5.01 g, 97% yield).

1H-NMR (300 MHz; CDCl3): δ 9.51 (bs, 2H), 7.70-7.67 (m, 1H), 7.55 (dt, J = 7.0, 0.8

Hz, 1H), 7.42-7.38 (m, 1H), 7.19-7.13 (m, 8H), 6.95-6.88 (m, 2H), 6.70 (d, J = 5.8 Hz,

1H), 6.06-5.93 (m, 1H), 4.99-4.92 (m, 1H), 4.54-4.40 (m, 1H), 3.43-3.34 (m, 1H), 3.19-

3.05 (m, 1H), 2.81-2.72 (m, 2H), 1.56 (s, 8H), 1.42 (d, J = 11.7 Hz, 9H); 13C-NMR (101

MHz; CDCl3): δ 178.03, 177.95, 159.41, 159.30, 146.7, 141.6, 137.56, 137.44, 136.8,

136.3, 135.3, 134.47, 134.33, 132.8, 132.4, 131.7, 130.8, 130.4, 129.5, 128.35, 128.18,

126.2, 123.97, 123.91, 123.64, 123.63, 123.58, 123.54, 123.52, 123.50, 123.47, 113.45,

113.42, 98.4, 86.33, 86.25, 83.81, 83.68, 64.5, 63.3, 32.0, 31.8.

REF: TRW-II-268, TRW-II-446.

NSO2Ph

NBoc

NCO2H

HO2C

BocHN

3.67

184

Figure 5.40a. 1H NMR spectrum of compound 3.67.

Figure 5.40b. 13C NMR spectrum of compound 3.67.

9.09.0

8.38.3

1.71.7

1.01.0

0.90.9

0.90.9

1.41.4

0.90.9

1.11.1

2.02.0

8.28.2

1.31.3

1.21.2

1.31.3

2.12.1

ppm0 011223344556677889910-19.97

1.40

1.44

1.56

2.72

2.73

2.76

2.76

2.77

2.77

2.77

2.78

2.78

2.80

2.80

2.81

2.81

2.81

2.81

3.05

3.05

3.05

3.06

3.06

3.06

3.08

3.14

3.14

3.14

3.15

3.15

3.16

3.18

3.19

3.34

3.35

3.35

3.35

3.37

3.37

3.38

3.38

3.38

3.38

3.38

3.39

3.39

3.39

3.41

3.42

3.42

3.42

3.43

4.40

4.41

4.41

4.42

4.42

4.51

4.51

4.51

4.52

4.52

4.52

4.53

4.53

4.53

4.53

4.54

4.92

4.92

4.92

4.93

4.93

4.93

4.93

4.94

4.94

4.96

4.96

4.96

4.96

4.96

4.97

4.99

4.99

4.99

4.99

5.93

5.93

5.93

6.02

6.02

6.03

6.03

6.03

6.04

6.04

6.05

6.05

6.05

6.06

6.69

6.71

6.88

6.88

6.90

6.92

6.92

6.93

6.93

6.93

6.93

6.93

6.94

6.94

6.95

6.95

7.13

7.14

7.14

7.16

7.16

7.16

7.17

7.17

7.17

7.17

7.18

7.18

7.18

7.19

7.19

7.19

7.37

7.38

7.40

7.42

7.42

7.42

7.54

7.54

7.54

7.56

7.56

7.57

7.67

7.70

7.70

7.70

9.51

ppm0 05050100100150150-253.79

31.8

432

.05

63.2

564

.49

83.6

883

.81

86.2

586

.33

98.4

211

3.42

113.

4512

3.47

123.

5012

3.52

123.

5412

3.58

123.

6312

3.64

123.

9112

3.97

126.

1612

8.18

128.

3512

9.46

130.

3613

0.81

131.

6713

2.40

132.

8113

4.33

134.

4713

5.28

136.

2613

6.81

137.

4413

7.56

141.

6214

6.74

159.

3015

9.41

177.

9517

8.03

185

Tetrapeptide (3.68). To a solution of L-serine-OMe·HCl (2.09 g, 13.4 mmol) in CH2Cl2

(70 mL) was added iPr2NEt (2.3 mL, 13.4 mmol). After stirring 15 minutes, the

suspension was added to a stirring solution of 3.67 (5.01 g, 6.69 mmol) and EDCI (2.57

g, 13.4 mmol) in CH2Cl2. The reaction was stirred O/N at r.t., then concentrated and

purified through a plug of SiO2 (100% EtOAc) to afford 3.68 (2.11 g, 33% yield).

1H-NMR (300 MHz; DMSO-d6): δ 8.10-8.08 (broad, 1H), 7.73-7.58 (m, 5H), 7.44-7.31

(m, 3H), 7.10-7.07 (m, 3H), 6.83-6.74 (m, 1H), 6.44-6.42 (m, 1H), 5.04-4.86 (m, 3H),

4.34-4.22 (m, 3H), 3.61-3.43 (m, 9H), 3.02-2.83 (m, 1H), 2.76-2.51 (m, 2H), 1.34-1.27

(m, 18H).

REF: TRW-II-270, TRW-II-447.

NSO2Ph

NBoc

NBocHN O

NH CO2Me

OH

O

NH

MeO2C

HO

3.68

186

Figure 5.41a. 1H NMR spectrum of compound 3.68.

13.713.7

2.02.0

1.11.1

8.88.8

2.92.9

2.62.6

0.70.7

1.11.1

2.62.6

2.82.8

4.84.8

1.01.0

ppm1 122334455667788-19.97

1.27

1.29

1.33

1.34

2.51

2.51

2.51

2.52

2.52

2.52

2.65

2.65

2.65

2.66

2.72

2.72

2.73

2.75

2.75

2.76

2.76

2.76

2.76

2.76

2.83

2.83

2.83

2.84

2.84

2.84

2.85

2.96

2.96

2.97

2.97

2.97

2.98

2.98

3.01

3.01

3.02

3.43

3.44

3.44

3.44

3.45

3.45

3.45

3.45

3.45

3.46

3.46

3.57

3.61

4.22

4.22

4.22

4.23

4.23

4.23

4.23

4.32

4.32

4.33

4.34

4.34

4.86

4.86

4.86

4.87

4.87

4.87

4.87

4.88

4.88

4.89

5.02

5.02

5.02

5.03

5.04

5.04

5.04

5.04

5.04

6.42

6.43

6.43

6.43

6.43

6.44

6.44

6.44

6.44

6.74

6.77

6.77

6.77

6.80

6.80

6.80

6.83

6.83

7.07

7.07

7.07

7.08

7.08

7.08

7.08

7.09

7.09

7.09

7.10

7.10

7.31

7.31

7.31

7.32

7.32

7.32

7.33

7.33

7.39

7.41

7.43

7.43

7.43

7.44

7.44

7.44

7.58

7.59

7.59

7.61

7.61

7.61

7.61

7.70

7.71

7.72

7.72

7.72

7.73

7.73

8.08

8.08

8.10

8.10

187

TBS2-Tetrapeptide (3.69). To a solution of diol 3.68 (2.11 g, 2.22 mmol) in DMF (12

mL) was added imidazole (728 mg, 10.7 mmol) and TBSCl (1.74 g, 11.5 mmol). The

reaction mixture stirred O/N at r.t., then was concentrated. The crude residue was taken

up in EtOAc and washed with water (3x), brine (1x), dried over Na2SO4, and

concentrated to afford the title compound (2.27 g, 87% yield).

1H-NMR (300 MHz; DMSO-d6): δ 8.02-7.99 (m, 1H), 7.72-7.42 (m, 6H), 7.37-7.21 (m,

2H), 7.13-6.86 (m, 4H), 6.48-6.39 (m, 1H), 4.92-4.89 (m, 1H), 4.43-4.39 (m, 1H), 4.21-

4.06 (m, 2H), 3.82-3.69 (m, 3H), 3.63-3.58 (m, 6H), 3.00-2.89 (m, 1H), 2.86-2.80 (m,

1H), 2.66-2.52 (m, 3H), 1.31 (dd, J = 22.1, 8.9 Hz, 18H), 0.81 (s, 18H), -0.01--0.03 (m,

12H).

REF: TRW-II-272.

NSO2Ph

NBoc

NBocHN O

NH CO2Me

OTBS

O

NH

MeO2C

TBSO

3.69

188

Figure 5.42a. 1H NMR spectrum of compound 3.69.

12.612.6

18.018.0

14.214.2

3.23.2

0.70.7

0.60.6

6.16.1

3.13.1

1.61.6

1.01.0

0.90.9

1.11.1

3.93.9

2.12.1

6.26.2

1.11.1

ppm0 01122334455667788-19.97

-0.0

3-0

.02

-0.0

10.

811.

261.

291.

341.

362.

522.

522.

522.

542.

552.

552.

552.

652.

652.

652.

662.

802.

802.

812.

812.

822.

832.

832.

832.

832.

842.

842.

842.

862.

892.

892.

902.

942.

942.

952.

952.

952.

962.

962.

992.

992.

993.

003.

583.

623.

633.

693.

693.

723.

743.

743.

803.

824.

064.

064.

074.

074.

204.

204.

204.

214.

214.

394.

394.

404.

404.

414.

424.

424.

424.

434.

434.

894.

894.

914.

914.

914.

914.

914.

926.

396.

396.

396.

406.

406.

406.

406.

416.

416.

466.

476.

476.

476.

476.

486.

866.

896.

896.

896.

896.

896.

966.

966.

967.

027.

037.

037.

037.

037.

047.

047.

047.

127.

127.

127.

137.

217.

227.

227.

227.

227.

227.

367.

367.

377.

377.

427.

437.

457.

557.

557.

567.

567.

567.

567.

607.

607.

677.

677.

697.

727.

727.

727.

997.

998.

018.

028.

028.

028.

02

189

Diamine (5.4). To a solution of 3.66 (2 g, 2.58 mmol) in CH2Cl2 (50 mL) at 0 °C was

added TFA (4.8 mL). The resulting solution was warmed to r.t. and stirred for 2 h under

Ar. The reaction was quenched with sat’d NaHCO3, and the product extracted in CH2Cl2.

The combined organic layers were dried and concentrated to afford 5.4 in quantitative

yield (1.48 g).

1H-NMR (300 MHz; DMSO-d6): δ 7.69 (d, J = 7.4 Hz, 2H), 7.63-7.58 (m, 2H), 7.50-

7.36 (m, 6H), 7.20-7.15 (m, 1H), 7.09-7.04 (m, 1H), 6.99-6.98 (m, 1H), 6.82-6.75 (m,

2H), 5.89 (d, J = 4.1 Hz, 1H), 4.51 (t, J = 4.2 Hz, 1H), 4.33-4.29 (m, 1H), 3.78 (s, 1H),

3.52 (d, J = 6.2 Hz, 3H), 3.39-3.32 (m, 3H), 2.95-2.78 (m, 2H), 2.71-2.65 (m, 2H),

HRMS (ESI-APCI-): [M-H]- 574.1886 calcd for C30H30N4O6S, found: 574.1867.

REF: TRW-II-282, TRW-II-289, TRW-II-292, TRW-II-300, TRW-II-335, TRW-II-353.

NSO2Ph

NH

NCO2Me

MeO2C

H2N

5.4

190

Figure 5.43a. 1H NMR spectrum of compound 5.4.

1.71.7

2.02.0

2.42.4

3.23.2

1.01.0

1.11.1

1.01.0

0.90.9

2.32.3

1.31.3

1.31.3

1.21.2

6.36.3

2.22.2

2.02.0

ppm0 01122334455667788-19.97

2.65

2.65

2.69

2.69

2.69

2.70

2.71

2.78

2.81

2.81

2.81

2.83

2.88

2.90

2.95

2.95

3.32

3.33

3.33

3.37

3.37

3.37

3.39

3.39

3.51

3.53

3.78

4.29

4.30

4.30

4.31

4.32

4.32

4.33

4.33

4.50

4.51

4.52

5.88

5.90

6.75

6.77

6.80

6.80

6.82

6.82

6.98

6.98

6.99

7.04

7.05

7.06

7.06

7.09

7.15

7.17

7.17

7.19

7.20

7.36

7.41

7.41

7.43

7.44

7.44

7.46

7.47

7.50

7.58

7.60

7.60

7.61

7.61

7.63

7.68

7.71

191

Tripeptide (3.71). N-Cbz-L-serine(OTBS) (2.52 g, 7.14 mmol), iPr2NEt (1.3 mL, 7.14

mmol), and EDCI (1.37 g, 7.14 mmol) were added to a solution of 5.4 (2.05 g, 3.57

mmol) in CH2Cl2 (20 mL). The reaction stirred O/N, then was concentrated and purified

through an SiO2 plug to afford pure 3.71 (2.37 g, 73% yield).

1H-NMR (300 MHz; DMSO-d6): δ 8.48-8.37 (m, 1H), 7.70-7.58 (m, 5H), 7.48-7.42 (m,

3H), 7.37-7.29 (m, 8H), 7.09-6.99 (m, 3H), 6.79-6.71 (m, 2H), 5.90-5.87 (m, 1H), 5.03-

4.96 (m, 2H), 4.49-4.40 (m, 2H), 4.31-4.29 (m, 1H), 4.23-4.19 (m, 1H), 3.78 (s, 1H),

3.64-3.61 (m, 1H), 3.53 (d, J = 4.4 Hz, 3H), 3.41-3.34 (m, 1H), 3.19 (d, J = 9.1 Hz, 3H),

3.01-2.93 (m, 1H), 2.68-2.64 (m, 1H), 0.78 (d, J = 10.3 Hz, 9H), -0.02 (s, 6H); HRMS

(ESI-APCI): [M+H]+ 910.3517 calcd for C47H56N5O10SSi, found: 910.3543.

REF: TRW-II-354, TRW-II-341, TRW-II-359.

NSO2Ph

NH

NCO2Me

MeO2C

NH

O

TBSOCbzHN

3.71

192

Figure 5.44a. 1H NMR spectrum of compound 3.71.

6.86.8

9.09.0

1.21.2

1.41.4

2.82.8

0.70.7

3.13.1

1.41.4

1.41.4

1.11.1

1.11.1

2.32.3

2.22.2

0.90.9

2.22.2

3.13.1

8.48.4

3.23.2

5.05.0

1.31.3

ppm0 01122334455667788-19.97

-0.0

20.

760.

792.

642.

642.

662.

672.

672.

672.

682.

682.

682.

932.

932.

942.

942.

952.

952.

962.

962.

982.

982.

993.

003.

003.

003.

013.

173.

203.

523.

533.

613.

613.

623.

633.

633.

643.

643.

643.

643.

784.

194.

214.

214.

214.

214.

224.

234.

234.

294.

304.

314.

314.

314.

404.

404.

484.

494.

494.

494.

964.

994.

995.

035.

875.

885.

885.

895.

895.

906.

716.

726.

726.

726.

726.

736.

736.

786.

786.

796.

796.

996.

997.

067.

087.

097.

297.

317.

377.

377.

427.

447.

447.

447.

487.

507.

507.

587.

617.

617.

637.

637.

677.

677.

677.

697.

707.

70

193

(R)-tert-butyl 3-(2-((tert-butoxycarbonyl)amino)-3-methoxy-3-oxopropyl)-1H-indole-

1-carboxylate (5.5). Powdered NaOH (19.0 g, 475 mmol) was added to a stirring

suspension of D-Trp-OMe·HCl (24.14 g, 95 mmol) and Bu4NHSO4 (3.23 g, 9.5 mmol) in

CH2Cl2 (950 mL). After stirring 2.5 h, Boc2O (62.1 g, 285 mmol) was added, and the

resulting suspension stirred O/N at r.t. The mixture was filtered through celite,

concentrated, and purified by SiO2 chromatography (9:1 hexanes:EtOAc) to afford N,N’-

Boc2-D-Trp-OMe (18.53 g, 47% yield).

1H-NMR (300 MHz; CDCl3): δ 8.11 (d, J = 7.4 Hz, 1H), 7.49 (d, J = 7.6 Hz, 1H), 7.39

(s, 1H), 7.27 (td, J = 16.1, 8.4 Hz, 2H), 5.11 (d, J = 7.2 Hz, 1H), 4.65 (q, J = 6.4 Hz, 1H),

3.69 (s, 3H), 3.22 (qd, J = 13.5, 5.5 Hz, 2H), 1.66 (s, 9H), 1.43 (s, 9H).

REF: TRW-III-171, TRW-III-239.

NBoc

NHBoc

CO2H

5.5

194

Figure 5.45a. 1H NMR spectrum of compound 5.5.

9.99.9

10.510.5

2.22.2

3.33.3

0.90.9

0.90.9

2.62.6

1.11.1

1.21.2

1.01.0

ppm1 1223344556677889-14.99

1.43

1.66

3.14

3.15

3.19

3.21

3.23

3.25

3.28

3.29

3.69

4.62

4.63

4.66

4.68

5.10

5.12

7.20

7.23

7.25

7.28

7.31

7.33

7.39

7.47

7.50

8.09

8.12

195

Bromopyrroloindoline (3.74). To a solution of N,N’-Boc2-D-Trp-OMe (18.53 g, 44.3

mmol) in CH2Cl2 (1000 mL) was added NBS (7.89 g, 44.3 mmol) and PPTS (11.12 g,

44.3 mmol). The resulting solution was allowed to stir at r.t. O/N. The reaction mixture

was washed with brine, the organic layer dried (Na2SO4) and concentrated. Purification

by SiO2 chromatography (30% EtOAc in hexanes) gave the desired product in 96% yield

(21.20 g).

[α]D = +150.0 (CH2Cl2); 1H-NMR (400 MHz; CDCl3): δ 7.53-7.52 (bs, 1H), 7.34-7.24

(m, 2H), 7.09 (t, J = 7.6 Hz, 1H), 6.37 (s, 1H), 3.86 (dd, J = 10.3, 6.3 Hz, 1H), 3.71 (s,

3H), 3.18 (dd, J = 12.6, 6.3 Hz, 1H), 2.79 (dd, J = 12.6, 10.3 Hz, 1H), 1.56 (s, 9H), 1.37

(bs, 9H); 13C-NMR (101 MHz; CDCl3): δ 171.46, 171.41, 171.0, 152.1, 141.5, 130.6,

124.37, 124.35, 123.2, 83.8, 82.2, 59.4, 52.3, 36.4, 28.6, 28.2, 24.1, 21.0, 14.2; IR (NaCl,

film) 2980, 1719, 1395, 1334, 1163, 753 cm-1; HRMS (ESI-APCI): [M+Na]+ 519.1107

calcd for C22H29BrN2NaO6, found: 519.1102.

REF: TRW-III-174, TRW-III-246.

NBoc

NBocCO2Me

Br

H

3.74

196

Figure 5.46a. 1H NMR spectrum of compound 3.74.

Figure 5.46b. 13C NMR spectrum of compound 3.74.

8.88.8

9.19.1

1.21.2

1.21.2

3.03.0

1.11.1

0.90.9

1.21.2

2.42.4

1.01.0

ppm0 0112233445566778-16.04

1.37

1.56

2.76

2.79

2.79

2.82

3.16

3.17

3.19

3.20

3.71

3.84

3.85

3.86

3.88

6.37

7.07

7.09

7.11

7.24

7.27

7.27

7.29

7.30

7.31

7.32

7.34

7.52

7.52

7.52

7.53

7.53

ppm0 05050100100150150-253.79

14.1

6

20.9

924

.07

28.1

728

.57

36.3

9

52.3

4

59.4

3

82.2

383

.77

123.

1912

4.35

124.

3713

0.57

141.

49

152.

14

171.

0317

1.41

171.

46

197

(2S,3aS,8aS)-1,8-di-tert-butyl 2-methyl 3a-(3-((S)-2-((tert-butoxycarbonyl)amino)-3-

methoxy-3-oxopropyl)-1H-indol-1-yl)-3,3a-dihydropyrrolo[2,3-b]indole-

1,2,8(2H,8aH)-tricarboxylate (3.75). KOtBu (3.02 g, 27 mmol) was added to a solution

of bromopyrroloindoline 3.74 (10.0 g, 20.2 mmol) and N-Boc-Trp-OMe (4.29 g, 13.5

mmol) in CH3CN (1000 mL) at 0 °C at stirred 1h under Ar. NaHCO3 was added and the

product extracted in CH2Cl2, washed with brine, dried (Na2SO4), and concentrated.

Purification by SiO2 chromatography (40% EtOAc in hexanes) afforded pure dipeptide

(5.47 g, 55% yield).

[α]D = -7.2 (CH2Cl2); 1H-NMR (400 MHz; CDCl3): δ 7.70-7.64 (bs, 1H), 7.51 (t, J = 7.5

Hz, 1H), 7.37-7.32 (m, 1H), 7.26 (s, 1H), 7.20-7.05 (m, J = 7.8 Hz, 4H), 6.74 (s, 1H),

6.67 (s, 1H), 4.97 (t, J = 6.4 Hz, 1H), 4.88 (d, J = 7.2 Hz, 1H), 4.53 (s, 1H), 3.58 (s, 2H),

3.53-3.46 (m, 2H), 3.20 (s, 3H), 3.13 (s, 3H), 1.51-1.48 (m, 18H), 1.39 (d, J = 7.7 Hz,

9H); 13C-NMR (101 MHz; CDCl3): δ 172.7, 171.0, 155.0, 151.9, 143.6, 134.7, 131.00,

130.89, 124.80, 124.78, 122.9, 122.31, 122.30, 122.0, 120.0, 119.55, 119.37, 111.5,

109.7, 82.1, 79.7, 72.57, 72.54, 60.3, 54.2, 52.1, 28.29, 28.17; IR (NaCl, film) 2979,

1716, 1457, 1207, 739 cm-1; HRMS (ESI-APCI): [M+Na]+ 757.3425 calcd for

C39H50N4NaO10, found: 757.3427.

REF: TRW-III-183, TRW-III-194.

NBoc

NBocCO2Me

H

N

CO2Me

NHBoc

3.75

198

Figure 5.47a. 1H NMR spectrum of compound 3.75.

Figure 5.47b. 13C NMR spectrum of compound 3.75.

9.569.56

20.720.7

2.842.84

3.193.19

2.572.57

2.022.02

1.271.27

11

11

0.8610.861

1.021.02

4.124.12

0.8590.859

1.361.36

1.321.32

1.131.13

ppm0 01122334455667788-16.04

1.38

1.40

1.48

1.50

1.51

3.13

3.20

3.46

3.49

3.50

3.51

3.53

3.58

4.53

4.87

4.89

4.96

4.97

4.99

6.67

6.74

7.05

7.07

7.09

7.10

7.12

7.15

7.17

7.19

7.20

7.24

7.26

7.26

7.32

7.33

7.33

7.34

7.34

7.35

7.36

7.36

7.37

7.49

7.51

7.53

7.64

7.64

7.64

7.64

7.67

7.67

7.69

7.70

7.70

7.70

ppm0 05050100100150150200200-253.79

28.1

728

.29

52.1

354

.24

60.3

5

72.5

472

.57

79.7

282

.11

109.

7111

1.46

119.

3711

9.55

119.

9712

1.95

122.

3012

2.31

122.

8712

4.78

124.

8013

0.89

131.

0013

4.72

143.

6015

1.92

155.

05

171.

0017

2.73

199

(2S,3aS,8aS)-methyl 3a-(3-((S)-2-amino-3-methoxy-3-oxopropyl)-1H-indol-1-yl)-

1,2,3,3a,8,8a-hexahydropyrrolo[2,3-b]indole-2-carboxylate (5.6). TFA (15 mL) was

slowly added to a solution of the Boc-protected compound 3.75 (5.47 g, 7.5 mmol) in

CH2Cl2 (150 mL) at 0 °C. The reaction mixture was allowed to warm to r.t. as it stirred

O/N. NaHCO3 was added and the product extracted into CH2Cl2. The combined organic

extracts were dried (Na2SO4) and concentrated to afford the deprotected product (3.02 g,

93% yield), used without further purification.

[α]D = +80.0 (CH2Cl2); 1H-NMR (400 MHz; CDCl3): δ 7.52 (dt, J = 6.6, 3.0 Hz, 1H),

7.40-7.37 (m, 1H), 7.11-6.99 (m, 5H), 6.70 (t, J = 7.4 Hz, 1H), 6.62 (dd, J = 7.9, 3.4 Hz,

1H), 5.44 (s, 1H), 4.14 (dd, J = 7.8, 2.6 Hz, 1H), 3.72 (t, J = 6.0 Hz, 1H), 3.60 (d, J =

17.0 Hz, 3H), 3.40 (ddd, J = 13.0, 7.9, 2.3 Hz, 1H), 3.29 (s, 3H), 3.15 (dt, J = 14.4, 4.3

Hz, 1H), 2.95-2.86 (m, 2H), 1.99-1.90 (bs, 3H); 13C-NMR (101 MHz; CDCl3): δ 175.6,

173.8, 149.7, 135.2, 130.5, 129.80, 129.76, 127.5, 125.4, 125.0, 121.8, 119.5, 119.2,

112.1, 110.9, 109.7, 81.8, 75.7, 60.3, 55.0, 52.1, 40.6, 30.5, 28.4; IR (NaCl, film) 3373,

2950, 1735, 1459, 1207, 744 cm-1; HRMS (ESI-APCI): [M+H]+ 435.2032 calcd for

C24H27N4O4, found: 435.2036.

REF: TRW-III-184, TRW-III-195.

NH

NHCO2Me

H

N

CO2Me

NH2

5.6

200

Figure 5.48a. 1H NMR spectrum of compound 5.6.

Figure 5.48b. 13C NMR spectrum of compound 5.6.

3.23.2

2.02.0

1.41.4

2.72.7

1.11.1

3.03.0

1.61.6

1.21.2

1.01.0

1.11.1

1.21.2

5.45.4

1.11.1

1.31.3

ppm0 0112233445566778-16.04

1.90

1.91

1.91

1.92

1.92

1.93

1.93

1.93

1.99

1.99

2.86

2.87

2.88

2.89

2.90

2.90

2.91

2.93

2.95

3.12

3.13

3.14

3.15

3.16

3.17

3.29

3.37

3.37

3.39

3.39

3.40

3.40

3.42

3.42

3.58

3.62

3.71

3.72

3.74

4.13

4.14

4.15

4.16

5.44

6.60

6.61

6.62

6.63

6.68

6.70

6.72

6.99

7.00

7.03

7.04

7.06

7.06

7.07

7.08

7.09

7.11

7.37

7.38

7.39

7.39

7.40

7.51

7.51

7.52

7.53

7.54

ppm0 05050100100150150-253.79

28.4

430

.54

40.6

4

52.0

655

.00

60.2

9

75.7

2

81.8

3

109.

7311

0.85

112.

0811

9.19

119.

5412

1.83

124.

9612

5.43

127.

5412

9.76

129.

8013

0.48

135.

25

149.

69

173.

7917

5.58

201

(2S,3aS,8aR)-methyl 3a-(3-((S)-2-((S)-2-((((9H-fluoren-9-yl)methoxy)-

carbonyl)amino)-3-((tert-butyldimethylsilyl)oxy)propanamido)-3-methoxy-3-

oxopropyl)-1H-indol-1-yl)-1-((S)-2-((((9H-fluoren-9-yl)methoxy)carbonyl)-amino)-3-

((tert-butyldimethylsilyl)oxy)propanoyl)-1,2,3,3a,8,8a-hexahydro-pyrrolo[2,3-

b]indole-2-carboxylate (3.76). Diamine 5.6 (1.03 g, 2.4 mmol), N-Fmoc-L-Ser(OTBS)-

OH (2.12 g, 4.8 mmol), T3P (50% by weight in DMF, 3.51 g, 5.5 mmol), iPr2NEt (2.1

mL, 12 mmol), and CH2Cl2 (24 mL) were combined and stirred under Ar O/N at 35 °C.

Water was added, the product extracted in EtOAc, the combined extracts washed with

brine, dried (Na2SO4), concentrated, and purified by SiO2 chromatography (10-25%

EtOAc in hexanes) to afford the tetrapeptide (2.15 g, 70% yield).

[α]D = +61.9 (CH2Cl2); 1H-NMR (300 MHz; CDCl3): δ 7.77 (d, J = 7.4 Hz, 4H), 7.60 (d,

J = 8.0 Hz, 5H), 7.41 (t, J = 7.3 Hz, 5H), 7.35-7.30 (m, 4H), 7.26-7.15 (m, 4H), 6.82-6.67

(m, 2H), 5.68 (d, J = 7.7 Hz, 1H), 5.42 (s, 1H), 4.90 (s, 1H), 4.40-4.33 (m, 4H), 4.24-4.10

(m, 3H), 4.05-3.88 (m, 2H), 3.74 (s, 1H), 3.63 (d, J = 13.3 Hz, 3H), 3.20 (s, 4H), 2.95 (s,

1H), 0.94-0.82 (m, 18H), 0.08 (dd, J = 35.4, 2.8 Hz, 12H); 13C-NMR (101 MHz; CDCl3):

δ 177.5, 177.3, 175.7, 175.35, 175.20, 160.8, 156.0, 150.0, 149.48, 149.34, 149.30, 147.1,

146.9, 140.3, 137.11, 137.00, 135.96, 135.82, 133.25, 133.17, 132.6, 131.5, 130.75,

130.70, 130.64, 130.60, 130.57, 130.4, 130.1, 127.8, 125.65, 125.58, 125.48, 125.1,

124.7, 117.28, 117.22, 116.21, 116.13, 114.93, 114.83, 84.8, 78.3, 73.0, 72.78, 72.75,

NH

NCO2Me

H

N

CO2Me

HNO

NHFmoc

OTBS

O

NHFmoc

OTBS

3.76

202

72.62, 70.8, 68.7, 65.3, 59.06, 58.90, 58.4, 57.7, 56.0, 52.75, 52.72, 52.69, 35.3, 33.9,

33.4, 31.8, 31.52, 31.43, 24.1, 23.75, 23.71, 0.23, 0.11, 0.08, 0.03, -0.01; IR (NaCl, film)

2953, 1724, 1673, 1450, 1253, 1107, 740 cm-1; HRMS (ESI-APCI): [M+H]+ 1281.5764

calcd for C72H85N6O12Si2, found: 1281.5759.

REF: TRW-III-185, TRW-III-196, TRW-III-202.

203

Figure 5.49a. 1H NMR spectrum of compound 3.76.

Figure 5.49b. 13C NMR spectrum of compound 3.76.

9.29.2

15.515.5

1.01.0

3.93.9

3.33.3

0.80.8

2.32.3

3.23.2

4.04.0

0.80.8

0.80.8

1.21.2

2.22.2

4.24.2

4.34.3

4.94.9

4.94.9

4.04.0

ppm0 0112233445566778-14.99

0.01

0.02

0.13

0.14

0.82

0.84

0.94

0.94

2.95

3.20

3.61

3.65

3.74

3.88

4.03

4.05

4.10

4.11

4.14

4.16

4.17

4.19

4.22

4.24

4.33

4.36

4.36

4.38

4.38

4.40

4.40

4.90

5.42

5.67

5.70

6.67

6.69

6.70

6.77

6.80

6.82

7.15

7.15

7.16

7.17

7.26

7.30

7.32

7.34

7.35

7.38

7.41

7.43

7.58

7.61

7.76

7.79

ppm0 05050100100150150200-253.79

-0.0

10.

030.

080.

110.

23

23.7

123

.75

24.0

531

.43

31.5

231

.76

33.4

333

.89

35.3

252

.69

52.7

252

.75

55.9

857

.74

58.4

158

.90

59.0

665

.29

68.6

770

.79

72.6

272

.75

72.7

873

.00

78.3

084

.77

114.

8311

4.93

116.

1311

6.21

117.

2211

7.28

124.

7412

5.11

125.

4812

5.58

125.

6512

7.85

130.

0913

0.35

130.

5713

0.60

130.

6413

0.70

130.

7513

1.50

132.

6213

3.17

133.

2513

5.82

135.

9613

7.00

137.

1114

0.26

146.

9014

7.11

149.

3014

9.34

149.

4815

0.01

155.

9516

0.78

175.

2017

5.35

175.

7417

7.25

177.

45

204

Dioxopiperazine2 (3.77). Tetrapeptide 3.76 (2.18 g, 1.7 mmol) and morpholine (17 mL)

were dissolved in THF (70 mL) and heated to 35 °C. After stirring O/N, the reaction

mixture was concentrated and purified by SiO2 chromatography (5% MeOH in CH2Cl2)

to afford pure dioxopiperazine 3.77 (1.11 g, 85% yield).

1H-NMR (400 MHz; CDCl3): δ 7.68 (d, J = 8.0 Hz, 1H), 7.56 (s, 1H), 7.29 (d, J = 7.4

Hz, 1H), 7.16 (t, J = 7.4 Hz, 1H), 7.09 (d, J = 7.8 Hz, 1H), 7.00 (d, J = 7.7 Hz, 1H), 6.81-

6.79 (m, 2H), 6.63 (d, J = 17.9 Hz, 1H), 6.42 (s, 1H), 6.13 (d, J = 3.5 Hz, 1H), 5.53 (d, J

= 3.5 Hz, 1H), 4.83 (dd, J = 11.1, 6.1 Hz, 1H), 4.37-4.35 (m, 1H), 4.25 (q, J = 5.4 Hz,

2H), 4.11 (t, J = 3.8 Hz, 1H), 4.03 (dd, J = 10.0, 3.5 Hz, 1H), 3.85 (dd, J = 19.1, 8.2 Hz,

2H), 3.72 (d, J = 14.7 Hz, 2H), 3.59 (dd, J = 9.9, 8.0 Hz, 1H), 3.54 (s, 1H), 3.34-3.29 (m,

1H), 3.19 (dd, J = 14.6, 9.8 Hz, 1H), 2.83 (dd, J = 14.7, 11.4 Hz, 1H), 0.96 (s, 18H), 0.16

(s, 12H); HRMS (ESI-APCI): [M+Na]+ 795.3698 calcd for C40H56N6NaO6Si2, found:

795.3696.

REF: TRW-III-191, TRW-III-198.

NH

NNH

O

O

OTBSH

N

NH

HN

O

O

OTBS

3.77

205

Figure 5.50a. 1H NMR spectrum of compound 3.77.

10.710.7

15.715.7

1.31.3

1.11.1

0.90.9

0.80.8

1.31.3

2.42.4

1.81.8

0.90.9

1.01.0

1.81.8

1.21.2

1.01.0

0.90.9

1.21.2

0.90.9

1.41.4

2.72.7

1.01.0

1.01.0

1.11.1

1.31.3

1.01.0

1.41.4

ppm0 0112233445566778-16.04

0.16

0.96

2.80

2.83

2.84

2.87

3.16

3.18

3.19

3.22

3.29

3.30

3.34

3.54

3.57

3.59

3.59

3.61

3.71

3.74

3.82

3.83

3.86

3.88

4.02

4.03

4.04

4.05

4.11

4.11

4.12

4.23

4.24

4.26

4.27

4.35

4.35

4.35

4.37

4.81

4.83

4.84

4.86

5.52

5.53

6.13

6.14

6.42

6.60

6.65

6.79

6.80

6.81

6.99

7.01

7.08

7.10

7.15

7.16

7.18

7.28

7.30

7.33

7.56

7.67

7.69

206

(2S,3aS,8aS)-1,8-di-tert-butyl 2-methyl 3a-(3-((S)-2-((tert-butoxycarbonyl)

(methyl)amino)-3-methoxy-3-oxopropyl)-1H-indol-1-yl)-3,3a-dihydro-pyrrolo[2,3-

b]indole-1,2,8(2H,8aH)-tricarboxylate (5.7). KOtBu (3.04 g, 27.2 mmol) was added to

a solution of bromopyrroloindoline 3.74 (10.08 g, 20.3 mmol) and N-Me,Boc-Trp-OMe

(4.50 g, 13.6 mmol) in CH3CN (1000 mL) at 0 °C and stirred 1h under Ar. NaHCO3 was

added and the product extracted in CH2Cl2, washed with brine, dried (Na2SO4), and

concentrated. Purification by SiO2 chromatography (10-20% EtOAc in hexanes) afforded

pure dipeptide (5.41 g, 53% yield).

[α]D = -3.1 (CH2Cl2); 1H-NMR (400 MHz; CDCl3): δ 7.68-7.67 (bs, 1H), 7.56 (d, J = 7.5

Hz, 1H), 7.35-7.32 (m, 2H), 7.23 (s, 1H), 7.16-7.08 (m, 3H), 6.84 (d, J = 15.2 Hz, 1H),

6.74 (d, J = 3.3 Hz, 1H), 4.87-4.86 (bs, 1H), 4.73-4.56 (broad, 2H), 3.69 (broad, J = 7.1

Hz, 3H), 3.49-3.43 (m, 1H), 3.29 (dd, J = 15.0, 4.9 Hz, 1H), 3.20 (s, 3H), 3.02-2.91 (m,

2H), 2.67-2.59 (m, 3H), 1.57-1.11 (m, 27H); 13C-NMR (101 MHz; CDCl3): δ 171.7,

170.9, 151.9, 134.7, 130.97, 130.82, 129.9, 125.0, 123.3, 122.2, 119.88, 119.86, 119.2,

111.6, 110.6, 82.0, 79.9, 72.59, 72.57, 59.9, 59.32, 59.30, 52.12, 52.09, 52.05, 52.00,

51.97, 34.6, 31.5, 28.19, 28.17, 25.2, 22.6, 20.7, 14.1; IR (NaCl, film) 2978, 1703, 1482,

1394, 1157, 738 cm-1; HRMS (ESI-APCI): [M+Na]+ 771.3581 calcd for C40H52N4NaO10,

found: 771.3600.

REF: TRW-III-211, TRW-III-258.

NBoc

NBocCO2Me

H

N

CO2Me

NMeBoc

5.7

207

Figure 5.51a. 1H NMR spectrum of compound 5.7.

Figure 5.51b. 13C NMR spectrum of compound 5.7.

26.626.6

3.03.0

2.02.0

2.62.6

1.21.2

1.41.4

2.92.9

1.81.8

1.31.3

1.01.0

0.80.8

3.43.4

1.41.4

1.71.7

1.31.3

1.11.1

ppm0 01122334455667788-16.04

1.11

1.19

1.36

1.47

1.48

1.49

1.51

1.52

1.57

2.59

2.62

2.67

2.91

2.93

2.96

2.96

2.99

3.02

3.20

3.27

3.28

3.31

3.32

3.43

3.44

3.46

3.47

3.49

3.67

3.68

3.71

4.56

4.62

4.63

4.63

4.64

4.64

4.64

4.65

4.65

4.72

4.72

4.73

4.73

4.73

4.86

4.86

4.86

4.87

4.87

4.87

4.87

6.74

6.75

6.82

6.86

7.08

7.10

7.10

7.12

7.13

7.16

7.16

7.23

7.32

7.33

7.35

7.56

7.57

7.67

7.67

7.67

7.67

7.68

7.68

ppm0 05050100100150150-253.79

14.0

8

20.6

622

.61

25.2

428

.17

28.1

931

.54

34.6

2

51.9

752

.00

52.0

552

.09

52.1

259

.30

59.3

259

.94

72.5

772

.59

79.8

782

.01

110.

5511

1.56

119.

1911

9.86

119.

8812

2.19

123.

3512

5.05

129.

9313

0.82

130.

9713

4.72

151.

90

170.

9517

1.66

208

(2S,3aS,8aS)-methyl 3a-(3-((S)-3-methoxy-2-(methylamino)-3-oxopropyl)-1H-indol-

1-yl)-1,2,3,3a,8,8a-hexahydropyrrolo[2,3-b]indole-2-carboxylate (3.80). TFA (11 mL)

was slowly added to a solution of Boc-protected material 5.7 (4.11 g, 5.49 mmol) in

CH2Cl2 (110 mL) at 0 °C. The reaction mixture was allowed to warm to r.t. as it stirred

O/N. NaHCO3 was added and the product extracted into CH2Cl2. The combined organic

extracts were dried (Na2SO4) and concentrated to afford the deprotected product (2.35 g,

96% yield), used without further purification.

[α]D = +73.3 (CH2Cl2); 1H-NMR (300 MHz; CDCl3): δ 7.61-7.58 (m, 1H), 7.46-7.43 (m,

1H), 7.18-7.05 (m, 6H), 6.77 (t, J = 7.4 Hz, 1H), 6.68 (dd, J = 7.9, 0.9 Hz, 1H), 5.51 (s,

1H), 4.55 (bs, 1H), 4.21 (d, J = 7.6 Hz, 1H), 3.61 (d, J = 9.5 Hz, 3H), 3.53-3.46 (m, 2H),

3.36 (d, J = 1.1 Hz, 3H), 3.10-2.93 (m, 4H), 2.35 (s, 3H); 13C-NMR (101 MHz; CDCl3): δ

174.7, 173.8, 149.69, 149.66, 135.26, 135.23, 130.4, 129.7, 127.58, 127.54, 125.3,

125.00, 124.89, 121.8, 119.5, 119.30, 119.13, 112.1, 110.74, 110.71, 109.55, 109.49,

82.0, 75.72, 75.69, 63.71, 63.69, 60.2, 52.1, 51.7, 40.76, 40.70, 34.68, 34.62, 28.91,

28.81; IR (NaCl, film) 2950, 1734, 1459, 1206, 742 cm-1; HRMS (ESI-APCI): [M+H]+

449.2189 calcd for C25H29N4O4, found: 449.2179.

REF: TRW-III-216, TRW-III-267.

NH

NHCO2Me

H

N

CO2Me

NHMe

3.80

209

Figure 5.52a. 1H NMR spectrum of compound 3.80.

Figure 5.52b. 13C NMR spectrum of compound 3.80.

3.63.6

4.04.0

3.03.0

2.42.4

3.13.1

1.01.0

0.90.9

1.01.0

1.01.0

1.11.1

6.06.0

1.11.1

1.31.3

ppm0 0112233445566778-14.99

2.35

2.93

2.96

2.97

2.98

3.06

3.08

3.09

3.10

3.35

3.36

3.46

3.46

3.48

3.50

3.52

3.53

3.60

3.63

4.20

4.22

4.54

4.55

4.55

4.55

5.51

6.67

6.67

6.70

6.70

6.74

6.77

6.79

7.05

7.05

7.11

7.12

7.13

7.15

7.16

7.16

7.18

7.43

7.44

7.46

7.58

7.59

7.59

7.59

7.60

7.61

ppm0 05050100100150150200-253.79

28.8

128

.91

34.6

234

.68

40.7

040

.76

51.7

052

.05

60.2

363

.69

63.7

1

75.6

975

.72

81.9

9

109.

4910

9.55

110.

7111

0.74

112.

1011

9.13

119.

3011

9.51

121.

8012

4.89

125.

0012

5.28

127.

5412

7.58

129.

6613

0.42

135.

26

149.

6614

9.69

173.

7917

4.72

210

(2S,3aS,8aR)-methyl 3a-(3-((S)-2-((S)-3-(benzyloxy)-2-((tert-

butoxycarbonyl)(methyl)amino)-N-methylpropanamido)-3-methoxy-3-oxopropyl)-

1H-indol-1-yl)-1-((S)-3-(benzyloxy)-2-((tert-

butoxycarbonyl)(methyl)amino)propanoyl)-1,2,3,3a,8,8a-hexahydropyrrolo[2,3-

b]indole-2-carboxylate (3.81). The diamine (3.80, 430 mg, 0.96 mmol), N-Me,Boc-L-

Ser(OBn)-OH (591 mg, 1.91 mmol), T3P (50% by weight in DMF, 1.41 g, 2.21 mmol),

iPr2NEt (0.835 mL, 4.8 mmol), and CH2Cl2 (10 mL) were combined and stirred under Ar

O/N at 35 °C. Water was added, the product extracted in EtOAc, the combined extracts

washed with brine, dried (Na2SO4), concentrated, and purified by SiO2 chromatography

(60% EtOAc in hexanes) to afford the tetrapeptide (420 mg, 42% yield).

1H-NMR (300 MHz; CDCl3): δ 7.58-7.55 (m, 1H), 7.41-7.15 (m, 16H), 6.99 (s, 1H),

6.76-6.69 (m, 1H), 6.26-6.02 (m, 1H), 5.42-5.12 (m, 3H), 4.96-4.86 (m, 1H), 4.61-4.41

(m, 5H), 3.71-3.54 (m, 7H), 3.40-3.35 (m, 2H), 3.18 (d, J = 16.2 Hz, 5H), 2.93-2.67 (m,

8H), 1.42 (t, J = 8.2 Hz, 18H); HRMS (ESI-APCI): [M+H]+ 1031.5130 calcd for

C57H71N6O12, found: 1031.5126.

REF: TRW-III-231, TRW-III-237, TRW-III-253.

NH

NCO2Me

H

N

CO2Me

MeNO

NMeBoc

OBn

O

NMeBoc

OBn

3.81

211

Figure 5.53a. 1H NMR spectrum of compound 3.81.

18.018.0

7.67.6

4.74.7

2.12.1

6.96.9

4.74.7

1.01.0

3.33.3

1.01.0

1.21.2

1.01.0

16.316.3

1.41.4

ppm0 01122334455667788-14.99

1.40

1.41

1.45

2.67

2.82

2.85

2.93

3.15

3.21

3.35

3.36

3.36

3.40

3.54

3.56

3.57

3.57

3.57

3.64

3.66

3.71

4.41

4.42

4.56

4.56

4.61

4.86

4.86

4.86

4.87

4.87

4.95

4.96

5.12

5.12

5.12

5.13

5.18

5.20

5.25

5.35

5.40

5.42

5.42

6.02

6.03

6.04

6.24

6.25

6.25

6.26

6.69

6.70

6.71

6.72

6.75

6.75

6.76

7.15

7.26

7.29

7.30

7.31

7.31

7.41

7.55

7.55

7.58

212

(2S,3aS,8aR)-methyl 1-((S)-3-(benzyloxy)-2-(methylamino)propanoyl)-3a-(3-((S)-2-

((S)-3-(benzyloxy)-N-methyl-2-(methylamino)propanamido)-3-methoxy-3-

oxopropyl)-1H-indol-1-yl)-1,2,3,3a,8,8a-hexahydropyrrolo[2,3-b]indole-2-

carboxylate (5.8). TFA (0.2 mL) was slowly added to a solution of Boc-protected

material 3.81 (103 mg, 0.1 mmol) in CH2Cl2 (2 mL) at 0 °C. The reaction mixture was

allowed to warm to r.t. as it stirred O/N. NaHCO3 was added and the product extracted

into CH2Cl2. The combined organic extracts were dried (Na2SO4) and concentrated to

afford the deprotected product (81 mg, 98% yield), used without further purification.

REF: TRW-III-233, TRW-III-243, TRW-III-254.

NH

NCO2Me

H

N

CO2Me

MeNO

NHMe

OBn

O

NHMe

OBn

5.8

213

Dioxopiperazine2 (3.82). Tetrapeptide 5.8 (81 mg, 0.098 mmol) and morpholine (1 mL)

were dissolved in THF (4 mL) and heated to 35 °C. After stirring O/N, the reaction

mixture was concentrated and purified by SiO2 chromatography (5% MeOH in CH2Cl2)

to afford pure dioxopiperazine (75 mg, >99% yield).

1H-NMR (300 MHz; CDCl3): δ 7.53-7.05 (m, 23H), 6.91 (t, J = 7.1 Hz, 2H), 6.76-6.71

(m, 2H), 6.46-6.34 (m, 2H), 5.72 (d, J = 4.1 Hz, 1H), 4.99-4.92 (m, 1H), 4.63-4.52 (m,

3H), 4.29-4.23 (m, 2H), 3.96 (dd, J = 9.7, 4.1 Hz, 2H), 3.69-3.54 (m, 5H), 3.26-3.21 (m,

2H), 3.08 (s, 3H), 2.87 (d, J = 3.9 Hz, 2H), 2.71 (dd, J = 12.2, 2.8 Hz, 1H), 2.21 (s, 3H);

HRMS (ESI-APCI): [M+Na]+ 789.3377 calcd for C45H46N6NaO6, found: 789.3379.

REF: TRW-III-235, TRW-III-245, TRW-III-257.

NH

NNMe

O

O

OBnH

N

NMe

MeN

O

O

OBn

3.82

214

Figure 5.54a. 1H NMR spectrum of compound 3.82.

3.173.17

1.371.37

2.192.19

2.922.92

2.472.47

5.275.27

2.262.26

2.342.34

3.233.23

1.141.14

0.8710.871

1.711.71

1.981.98

2.172.17

2323

ppm0 01122334455667788-14.99

2.21

2.69

2.70

2.73

2.74

2.86

2.87

3.08

3.21

3.22

3.25

3.26

3.26

3.54

3.54

3.57

3.58

3.59

3.62

3.64

3.65

3.66

3.68

3.69

3.94

3.95

3.97

3.99

4.23

4.24

4.25

4.29

4.52

4.55

4.56

4.58

4.59

4.63

4.92

4.92

4.94

4.96

4.96

4.96

4.97

4.98

4.99

5.72

5.73

6.34

6.43

6.46

6.71

6.74

6.76

6.76

6.89

6.91

6.94

7.05

7.15

7.16

7.25

7.26

7.27

7.31

7.34

7.34

7.35

7.50

7.53

215

(2S,3aS,8aR)-methyl 3a-(3-((S)-2-(2-((((9H-fluoren-9-

yl)methoxy)carbonyl)(methyl)amino)-N-methylacetamido)-3-methoxy-3-oxopropyl)-

1H-indol-1-yl)-1-(2-((((9H-fluoren-9-yl)methoxy)carbonyl)(methyl)amino)acetyl)-

1,2,3,3a,8,8a-hexahydropyrrolo[2,3-b]indole-2-carboxylate (5.9). The diamine (3.80,

896 mg, 2.0 mmol), N-Fmoc-sarcosine-OH (1.24 g, 4.0 mmol), T3P (50% by weight in

DMF, 2.93 g, 4.6 mmol), iPr2NEt (1.7 mL, 10.0 mmol), and CH2Cl2 (20 mL) were

combined and stirred under Ar O/N at 35 °C. Water was added, the product extracted in

EtOAc, the combined extracts washed with brine, dried (Na2SO4), concentrated, and

purified by SiO2 chromatography (50-100% EtOAc in hexanes) to afford tetrapeptide 5.9

(1.17 g, 57% yield).

[α]D = +92.1 (CH2Cl2); 1H-NMR (300 MHz; CDCl3): δ 7.78-7.74 (m, 4H), 7.63-7.59 (m,

6H), 7.35 (dd, J = 19.2, 7.0 Hz, 8H), 7.16-7.04 (m, 5H), 6.78-6.65 (m, 2H), 6.10-6.00 (m,

1H), 4.54-4.20 (m, 7H), 3.99-3.86 (m, 2H), 3.72-3.61 (m, 5H), 3.37-3.33 (m, 3H), 3.23-

3.08 (m, 6H), 2.99-2.90 (m, 4H), 2.80-2.56 (m, 4H); IR (NaCl, film) 2951, 1703, 1451,

1229, 740 cm-1; HRMS (ESI-APCI): [M+H]+ 1035.4293 calcd for C61H59N6O10, found:

1035.4285.

REF: TRW-III-292.

NH

NCO2Me

H

N

CO2Me

MeNO

NMeFmoc

O

NMeFmoc

5.9

216

Figure 5.55a. 1H NMR spectrum of compound 5.9.

4.34.3

4.24.2

6.36.3

2.62.6

4.94.9

1.81.8

7.17.1

0.70.7

1.71.7

5.35.3

8.48.4

6.06.0

4.04.0

ppm1 12233445566778-14.99

2.56

2.59

2.66

2.66

2.70

2.80

2.90

2.95

2.97

2.99

3.08

3.09

3.10

3.23

3.33

3.34

3.34

3.34

3.34

3.35

3.36

3.36

3.37

3.37

3.37

3.61

3.62

3.72

3.86

3.86

3.87

3.87

3.87

3.88

3.92

3.99

4.20

4.20

4.20

4.21

4.22

4.22

4.29

4.30

4.39

4.51

4.52

4.52

4.52

4.54

6.00

6.00

6.07

6.10

6.65

6.65

6.65

6.66

6.68

6.68

6.68

6.69

6.70

6.77

6.78

7.00

7.04

7.12

7.14

7.16

7.31

7.33

7.38

7.40

7.59

7.59

7.61

7.63

7.63

7.74

7.76

7.78

217

(5aR,10bS,11aS)-10b-(3-(((S)-1,4-dimethyl-3,6-dioxopiperazin-2-yl)methyl)-1H-

indol-1-yl)-2-methyl-2,3,5a,6,11,11a-hexahydro-1H-pyrazino[1',2':1,5]pyrrolo[2,3-

b]indole-1,4(10bH)-dione (3.88). Tetrapeptide 5.9 (1.0 g, 0.97 mmol) and morpholine

(10 mL) were dissolved in THF (40 mL) and heated to 35 °C. After stirring O/N, the

reaction mixture was concentrated and purified by SiO2 chromatography (2-6% MeOH in

CH2Cl2) to afford pure dioxopiperazine 3.88 (470 mg, 92% yield).

[α]D = +145.5 (CH2Cl2); 1H-NMR (300 MHz; CDCl3): δ 7.49 (t, J = 7.3 Hz, 1H), 7.26-

7.15 (m, 2H), 7.09-6.91 (m, 3H), 6.79-6.73 (m, 2H), 6.01 (dd, J = 51.3, 3.1 Hz, 1H), 5.45

(dd, J = 53.9, 3.8 Hz, 1H), 4.57-4.49 (m, 1H), 4.18 (t, J = 3.2 Hz, 1H), 3.84-3.46 (m, 4H),

3.23-3.12 (m, 2H), 3.06-2.98 (m, 6H), 2.68 (ddd, J = 15.3, 11.5, 4.2 Hz, 1H), 2.33 (d, J =

32.5 Hz, 3H), 2.06-1.94 (m, 1H); 13C-NMR (101 MHz; CDCl3): δ 166.3, 166.0, 164.6,

164.2, 146.7, 135.4, 130.1, 129.1, 128.1, 127.8, 124.7, 124.4, 122.8, 122.5, 120.2, 119.7,

112.4, 110.2, 108.3, 82.6, 82.0, 74.1, 62.7, 57.2, 53.5, 40.1, 39.7, 33.5, 32.9, 32.0; IR

(NaCl, film) 2931, 1664, 1459, 1324, 1259 cm-1; HRMS (ESI-APCI): [M+H]+ 527.2407

calcd for C29H31N6O4, found: 527.2402.

REF: TRW-III-293.

NH

NNMe

O

OH

N

NMe

MeN

O

O

3.88

218

Figure 5.56a. 1H NMR spectrum of compound 3.88.

Figure 5.56b. 13C NMR spectrum of compound 3.88.

1.11.1

3.43.4

1.41.4

6.16.1

1.91.9

4.34.3

1.21.2

0.90.9

1.01.0

1.01.0

2.42.4

3.33.3

2.02.0

1.01.0

ppm1 1223344556677-14.99

1.94

2.00

2.06

2.27

2.38

2.63

2.64

2.67

2.68

2.69

2.72

2.73

2.98

3.02

3.05

3.06

3.12

3.13

3.15

3.16

3.18

3.20

3.21

3.23

3.46

3.48

3.51

3.52

3.53

3.57

3.57

3.61

3.62

3.63

3.66

3.68

3.71

3.76

3.78

3.84

4.17

4.18

4.19

4.49

4.51

4.53

4.54

4.57

5.36

5.37

5.54

5.55

5.92

5.93

6.09

6.11

6.73

6.75

6.76

6.76

6.77

6.79

6.89

6.91

6.93

6.96

6.99

7.01

7.05

7.07

7.09

7.15

7.17

7.20

7.22

7.22

7.25

7.26

7.26

7.47

7.49

7.52

ppm0 05050100100150150-250.03

32.0

032

.86

33.5

439

.74

40.0

7

53.5

457

.21

62.6

5

74.1

0

82.0

082

.64

108.

3411

0.19

112.

4011

9.68

120.

2212

2.55

122.

8312

4.39

124.

7112

7.84

128.

1112

9.12

130.

1413

5.36

146.

73

164.

2016

4.64

166.

0516

6.33

219

Tetrapeptide (5.10). The diamine (3.80, 327 mg, 0.73 mmol), N-Me,Fmoc-L-

Ser(OTBS)-OH (663 g, 1.46 mmol), T3P (50% by weight in DMF, 1.07 g, 1.68 mmol),

iPr2NEt (0.635 mL, 5.0 mmol), and CH2Cl2 (7.3 mL) were combined and stirred under Ar

O/N at 35 °C. Water was added, the product extracted in EtOAc, the combined extracts

washed with brine, dried (Na2SO4), concentrated, and purified by SiO2 chromatography

(35% EtOAc in hexanes) to afford tetrapeptide 5.10 (351 mg, 36% yield).

[α]D = -1.5 (CH2Cl2); 1H-NMR (300 MHz; CDCl3): δ 7.78-7.71 (m, 4H), 7.61-7.53 (m,

5H), 7.41-7.15 (m, 14H), 6.93-6.66 (m, 3H), 5.13-5.07 (m, 2H), 4.41 (d, J = 6.8 Hz, 2H),

4.16-4.01 (m, 5H), 3.93-3.89 (m, 1H), 3.75-3.66 (m, 5H), 3.40-3.35 (m, 2H), 3.25 (d, J =

10.7 Hz, 2H), 3.09-2.94 (m, 5H), 2.82-2.81 (m, 4H), 2.72-2.66 (m, 2H), 2.34 (dd, J = 6.3,

3.2 Hz, 1H), 2.10-1.95 (m, 2H), 0.87-0.81 (m, 18H), 0.09--0.02 (m, 12H); IR (NaCl,

film) 2953, 1696, 1651, 1451, 1317, 1154, 740 cm-1; HRMS (ESI-APCI): [M+H]+

1323.6234 calcd for C75H91N6O12Si2, found:1323.6253.

REF: TRW-III-299, TRW-III-314.

NH

NCO2Me

H

N

CO2Me

MeNO

NHMeFmoc

O

NMeFmoc

5.10

OTBS

OTBS

220

Figure 5.57a. 1H NMR spectrum of compound 5.10.

12.012.0

18.818.8

1.71.7

1.21.2

2.02.0

4.24.2

5.45.4

1.91.9

1.81.8

5.05.0

1.01.0

5.15.1

1.91.9

2.32.3

3.03.0

14.914.9

5.25.2

4.44.4

ppm0 0112233445566778-14.99

-0.0

2-0

.02

0.02

0.08

0.09

0.81

0.82

0.85

0.86

0.87

1.95

2.05

2.09

2.10

2.33

2.34

2.35

2.36

2.66

2.71

2.72

2.81

2.82

2.94

2.98

2.99

3.06

3.09

3.23

3.27

3.35

3.35

3.36

3.36

3.40

3.66

3.67

3.69

3.69

3.70

3.71

3.74

3.75

3.89

3.90

3.90

3.93

4.01

4.06

4.09

4.11

4.12

4.13

4.16

4.40

4.42

5.07

5.08

5.08

5.10

5.11

5.11

5.13

6.66

6.68

6.71

6.71

6.74

6.74

6.77

6.78

6.81

6.83

6.86

6.87

6.91

6.93

7.15

7.15

7.16

7.17

7.26

7.26

7.28

7.29

7.30

7.31

7.31

7.36

7.38

7.40

7.41

7.53

7.54

7.57

7.59

7.60

7.61

7.61

7.71

7.71

7.73

7.76

7.78

221

(3S,5aR,10bS,11aS)-3-(((tert-butyldimethylsilyl)oxy)methyl)-10b-(3-(((2S,5S)-5-

(((tert-butyldimethylsilyl)oxy)methyl)-1,4-dimethyl-3,6-dioxopiperazin-2-yl)methyl)-

1H-indol-1-yl)-2-methyl-2,3,5a,6,11,11a-hexahydro-1H-

pyrazino[1',2':1,5]pyrrolo[2,3-b]indole-1,4(10bH)-dione (3.90). Tetrapeptide 5.10 (300

mg, 0.227 mmol) and morpholine (2.3 mL) were dissolved in THF (9 mL) and heated to

35 °C. After stirring O/N, the reaction mixture was concentrated and purified by SiO2

chromatography (4% MeOH in CH2Cl2) to afford pure dioxopiperazine 3.90 (153 mg,

83% yield).

3.90A: [α]D = +56.6 (CH2Cl2); 1H-NMR (300 MHz; CDCl3): δ 7.51 (d, J = 7.9 Hz, 1H),

7.18 (t, J = 7.6 Hz, 1H), 7.05 (d, J = 11.2 Hz, 2H), 6.90 (t, J = 6.1 Hz, 2H), 6.69 (t, J =

7.5 Hz, 2H), 6.50 (d, J = 8.4 Hz, 1H), 5.70 (s, 1H), 4.80 (dd, J = 11.9, 5.9 Hz, 1H), 4.33-

4.09 (m, 4H), 3.76 (d, J = 10.4 Hz, 1H), 3.66 (dt, J = 9.9, 4.9 Hz, 2H), 3.50-3.43 (m, 2H),

3.37 (t, J = 4.8 Hz, 1H), 3.19 (dd, J = 14.8, 4.6 Hz, 1H), 3.10 (s, 3H), 3.02 (s, 3H), 2.31

(s, 3H), 0.82 (s, 9H), 0.74 (s, 9H), 0.04 (d, J = 4.6 Hz, 6H), -0.11 (d, J = 4.3 Hz, 6H); 13C-

NMR (101 MHz; CDCl3): δ 167.9, 166.5, 165.73, 165.62, 146.6, 135.6, 130.2, 129.5,

124.4, 123.1, 122.5, 120.2, 112.3, 110.0, 108.6, 82.9, 62.6, 62.2, 62.0, 60.5, 59.9, 56.9,

40.7, 32.5, 30.4, 30.0, 27.2, 25.64, 25.52, 18.1, 17.9, -5.60, -5.66, -5.69; IR (NaCl, film)

2930, 2857, 1662, 1461, 1256, 1116 cm-1; HRMS (ESI-APCI): [M+H]+ 815.4348 calcd

for C43H63N6O6Si2, found:815.4352.

NH

NNMe

O

OH

N

NMe

MeN

O

O

OTBS

OTBS

3.90

222

3.90B: [α]D = +80.0 (CH2Cl2); 1H-NMR (300 MHz; CDCl3): δ 7.60 (d, J = 7.9 Hz, 1H),

7.30 (s, 1H), 7.15 (td, J = 7.7, 1.0 Hz, 1H), 7.07 (t, J = 7.4 Hz, 1H), 6.99-6.94 (m, 1H),

6.88 (d, J = 7.4 Hz, 1H), 6.76 (d, J = 8.3 Hz, 1H), 6.69-6.63 (m, 2H), 5.91 (d, J = 2.6 Hz,

1H), 5.59 (d, J = 2.6 Hz, 1H), 4.57-4.52 (m, 1H), 4.26-4.19 (m, 2H), 4.03 (dd, J = 10.8,

2.7 Hz, 1H), 3.87-3.77 (m, 2H), 3.68-3.61 (m, 2H), 3.40 (dd, J = 9.4, 5.6 Hz, 2H), 3.26-

3.21 (m, 1H), 2.99 (d, J = 9.6 Hz, 6H), 2.71 (s, 3H), 2.60-2.55 (m, 1H), 0.84 (s, 9H), 0.74

(s, 9H), -0.01--0.08 (m, 12H); 13C-NMR (101 MHz; CDCl3): δ 166.0, 164.56, 164.55,

164.35, 146.7, 135.6, 130.1, 129.5, 123.1, 122.6, 120.4, 119.5, 112.1, 111.2, 109.7, 82.7,

65.1, 64.2, 63.7, 63.3, 60.6, 56.8, 41.5, 33.8, 33.1, 31.1, 30.7, 25.9, 25.5, 18.4, 18.0, -

5.38, -5.40, -5.71, -5.74; IR (NaCl, film) 2930, 2857, 1654, 1462, 1255, 1112 cm-1;

HRMS (ESI-APCI): [M+H]+ 815.4348 calcd for C43H63N6O6Si2, found:815.4349.

REF: TRW-III-301, TRW-III-315.

223

Figure 5.58a. 1H NMR spectrum of compound 3.90A.

Figure 5.58b. 13C NMR spectrum of compound 3.90A.

7.57.5

5.85.8

10.110.1

10.510.5

2.92.9

3.43.4

2.92.9

1.31.3

1.21.2

1.91.9

2.02.0

1.21.2

4.44.4

1.01.0

0.90.9

0.90.9

2.12.1

2.22.2

1.91.9

1.41.4

1.21.2

ppm0 01122334455667788-14.99

-0.1

2-0

.10

0.04

0.05

0.74

0.82

2.31

3.02

3.10

3.15

3.17

3.20

3.22

3.36

3.37

3.39

3.43

3.43

3.46

3.47

3.48

3.50

3.53

3.62

3.64

3.66

3.68

3.69

3.74

3.78

4.09

4.10

4.13

4.13

4.19

4.19

4.23

4.29

4.29

4.32

4.33

4.77

4.79

4.81

4.83

5.70

6.48

6.51

6.67

6.69

6.72

6.88

6.90

6.92

7.00

7.03

7.07

7.16

7.18

7.21

7.49

7.52

ppm0 05050100100150150-250.03

-5.6

9-5

.66

-5.6

0

17.9

218

.12

25.5

225

.64

27.2

230

.04

30.4

232

.51

40.6

8

56.9

459

.88

60.5

561

.98

62.2

262

.59

82.8

6

108.

6311

0.04

112.

2812

0.19

122.

4612

3.14

124.

4112

9.53

130.

1613

5.56

146.

56

165.

6216

5.73

166.

5016

7.87

224

Figure 5.59a. 1H NMR spectrum of compound 3.90B.

Figure 5.59b. 13C NMR spectrum of compound 3.90B.

10.710.7

8.48.4

9.29.2

1.11.1

2.72.7

5.85.8

1.31.3

2.12.1

2.12.1

2.42.4

1.01.0

1.71.7

0.80.8

0.80.8

0.80.8

1.81.8

1.11.1

1.01.0

1.01.0

1.01.0

1.31.3

0.90.9

1.01.0

ppm0 011223344556677-14.99

-0.0

8-0

.05

-0.0

1-0

.01

0.74

0.84

2.55

2.56

2.60

2.71

2.97

3.00

3.21

3.22

3.24

3.25

3.26

3.38

3.40

3.41

3.43

3.61

3.62

3.64

3.67

3.68

3.77

3.78

3.81

3.82

3.85

3.85

3.86

3.87

4.01

4.02

4.04

4.05

4.19

4.20

4.21

4.22

4.23

4.25

4.26

4.52

4.53

4.53

4.56

4.57

5.58

5.59

5.90

5.91

6.63

6.66

6.69

6.69

6.75

6.78

6.87

6.89

6.94

6.94

6.97

6.99

6.99

7.04

7.07

7.09

7.12

7.12

7.14

7.15

7.17

7.17

7.30

7.59

7.62

ppm0 05050100100150150-250.03

-5.7

4-5

.71

-5.4

0-5

.38

18.0

218

.35

25.5

525

.89

30.7

331

.13

33.0

533

.76

41.4

7

56.8

260

.59

63.3

463

.66

64.1

765

.06

82.6

5

109.

6811

1.21

112.

1511

9.52

120.

3812

2.57

123.

1512

9.46

130.

1213

5.60

146.

66

164.

3516

4.55

164.

5616

5.96

225

5-chlorovanillin (5.11). Vanillin (25 g, 164 mmol) was dissolved in AcOH (150 mL),

then chlorine gas bubbled through the solution. After a significant amount of white solid

had precipitated, the solution was filtered and the solid washed with hexanes. To the

AcOH filtrate was added an additional batch of vanillin (25 g), and chlorine gas again

bubbled through the solution. The resulting precipitate was filtered, washed with hexanes,

combined with the first yield, and dried to afford 5-chlorovanillin (39.30 g, 64% yield).

1H-NMR (400 MHz; acetone-d6): δ 9.83 (s, 1H), 7.58 (d, J = 1.7 Hz, 1H), 7.43 (d, J =

1.7 Hz, 1H), 3.98 (s, 3H); 13C-NMR (101 MHz; acetone): δ 189.5, 148.7, 129.2, 125.6,

120.0, 108.7, 56.0.

REF: TRW-II-413, TRW-II-463, TRW-II-479, TRW-III-076.

CHO

OMeHO

5.11

Cl

226

Figure 5.60a. 1H NMR spectrum of compound 5.11.

Figure 5.60b. 13C NMR spectrum of compound 5.11.

3.283.28

1.161.16

0.8280.828

1.011.01

ppm0 0224466881010-16.043.98

7.43

7.43

7.57

7.58

9.83

ppm0 05050100100150150200200-253.79

28.32

28.52

29.29

29.48

55.96

108.68

119.97

125.57

129.23

148.62

148.81

189.47

205.40

227

5-chlorovanillin-OAc (4.15). To a solution of 5-chlorovanillin (58.5 g, 313 mmol) in

THF (1000 mL) was added Ac2O (35.5 mL, 376 mmol), Et3N (65.5 mL, 470 mmol), and

DMAP (200 mg). The resulting solution was stirred O/N, then concentrated. The product

was extracted in CH2Cl2 from 1M HCl, the combined organic extracts washed with brine,

dried over Na2SO4, and concentrated to give 4.15 (71.45 g, >99% yield).

1H-NMR (400 MHz; acetone-d6): δ 9.98 (s, 1H), 7.67 (d, J = 1.7 Hz, 1H), 7.59 (d, J =

1.7 Hz, 1H), 3.97 (s, 3H), 2.36 (s, 3H); 13C-NMR (101 MHz; acetone): δ 190.0, 166.8,

153.6, 141.3, 135.4, 128.5, 123.2, 110.4, 56.2, 19.2; HRMS (ESI-APCI): [M-H]-

229.0268 calcd for C10H9ClO4, found: 229.9854.

REF: TRW-II-415, TRW-II-464, TRW-II-481, TRW-III-078.

CHO

OMeAcO

Cl

4.15

228

Figure 5.61a. 1H NMR spectrum of compound 4.15.

Figure 5.61b. 13C NMR spectrum of compound 4.15.

2.762.76

33

1.131.13

0.8580.858

11

ppm0 0224466881010-16.04

2.36

3.97

7.59

7.66

7.67

9.98

ppm0 05050100100150150200200-253.79

19.18

28.33

28.52

28.71

28.90

29.10

29.29

29.48

56.16

110.41

123.24

128.49

135.36

141.34

153.59

166.78

190.02

229

5-chloro-2-nitrovanillin (4.17). Nitric acid (fuming, 125) was chilled in an ethylene

glycol/dry ice bath, then 4.15 (28 g) added in small batches such that the internal

temperature did not rise above 5 °C. Following the addition, the reaction was stirred for 2

additional hours at -15 °C. The reaction mixture was carefully poured into ice water, and

the resulting bright orange precipitate filtered and rinsed with cold water until the

washings were pH neutral. This solid (combined from 2x28 g batches) was dissolved in

2M KOH (500 mL) and the solution heated to a boil for 10 min. After cooling to r.t., the

solution was poured into ice-cold concentrated HCl, and the resulting precipitate filtered,

washed with cold water, and dried to afford 4.17 (43.21 g, 76% yield).

1H-NMR (400 MHz; acetone-d6): δ 9.82 (s, 1H), 7.94 (s, 1H), 3.95 (s, 3H); 13C-NMR

(101 MHz; acetone): δ 186.0, 153.0, 140.9, 129.6, 123.2, 119.6, 98.5, 62.4; HRMS (-

APCI): [M-H]- 229.9856 calcd for C8H6ClNO5, found: 229.9860.

REF: TRW-II-417,419; TRW-II-424,425; TRW-II-484, TRW-III-080.

Nitric acid (fuming). Potassium nitrate (500 g) was added to concentrated sulfuric acid

(500 mL) in a flask equipped with a distillation column, and the resulting suspension

heated in an oil bath at 170 °C. Fuming nitric acid (75 mL) was collected over a 4 hour

period. This can be purchased commercially from VWR.

CHO

OMeHO

Cl

NO2

4.17

230

Figure 5.62a. 1H NMR spectrum of compound 4.17.

Figure 5.62b. 13C NMR spectrum of compound 4.17.

3.073.07

0.9080.908

11

ppm0 0224466881010-16.043.95

7.94

9.82

ppm0 05050100100150150200200-253.79

62.39

98.52

119.63

123.18

129.55

140.87

153.04

185.96

231

5-chloro-3,4-dimethoxy-2-nitrobenzaldehyde (4.18). Phenol 4.17 (15.0 g, 64.7 mmol)

was dissolved in absolute EtOH (26.3 mL), then Me2SO4 (26.3 mL, 278 mmol) added.

The resulting solution was immersed in an ice/brine bath before adding a solution of

NaOH (11.1 g, 278 mmol) in water (13.2 mL) dropwise. The reaction was heated to 50

°C for 3 hours, then cooled to r.t. and diluted with water. The product was extracted into

ether, and the organic extracts dried and concentrated to afford the title compound (15.9

g, >99% yield).

1H-NMR (400 MHz; CDCl3): δ 9.80 (d, J = 17.5 Hz, 1H), 7.73 (s, 1H), 4.05 (s, 3H), 4.01

(s, 3H); 13C-NMR (101 MHz; CDCl3): δ 184.7, 155.3, 151.4, 131.6, 126.7, 123.1, 120.3,

62.7, 61.4; HRMS (-APCI): [M-H]- 245.0091 calcd for C10H9ClN2O6, found: 245.0080.

4.19: 1H-NMR (300 MHz; CDCl3): δ 7.45 (s, 1H), 5.58 (s, 1H), 3.97 (s, 3H), 3.93 (s,

3H), 3.65-3.50 (m, 4H), 1.22 (t, J = 7.1 Hz, 6H).

REF: TRW-II-421, TRW-II-429, TRW-II-485, TRW-III-081.

CHO

OMeMeO

Cl

NO2

4.18

232

Figure 5.63a. 1H NMR spectrum of compound 4.18.

Figure 5.63b. 13C NMR spectrum of compound 4.18.

66

0.9710.971

1.111.11

ppm0 0224466881010-16.044.01

4.05

7.73

9.78

9.82

ppm0 05050100100150150200-253.79

61.42

61.53

62.71

120.35

123.05

126.73

131.57

151.41

155.26

184.72

233

Figure 5.64a. 1H NMR spectrum of compound 4.19.

6.16.1

4.14.1

3.33.3

3.33.3

0.90.9

1.01.0

ppm0 0224466881010-19.97

1.20

1.22

1.25

3.50

3.51

3.53

3.53

3.56

3.57

3.58

3.60

3.62

3.63

3.64

3.65

3.93

3.97

5.58

7.45

OMeMeO

Cl

NO2

OEt

OEt

4.19

234

Dinitrostyrene (4.7). Aldehyde 4.18 (13.64 g, 55.4 mmol) was suspended in EtOH (180

mL), and to it added nitromethane (4.4 mL, 77.6 mmol). The mixture was cooled to -15

°C, then a solution of KOH (13.3 g, 238 mmol) in water (18 mL) and EtOH (180 mL)

added dropwise over 2 hours. Upon complete addition of the KOH solution, the reaction

was acidified with concentrated HCl (34 mL), diluted with H2O (200 mL), and the

product extracted into chloroform. The organic extracts were dried and concentrated. To

the resulting oil was added 55 mL acetic anhydride and sodium acetate (5.5 g). The

solution was heated to reflux for 30 min., then poured into water and stirred. Water was

decanted from the brown paste then EtOH added and heated to dissolve the crude

product. Pure nitrostyrene crystals were recovered (10.3 g, 64% yield).

1H-NMR (400 MHz; DMSO-d6): δ 8.33 (d, J = 13.3 Hz, 1H), 8.15 (s, 1H), 7.63 (d, J =

13.4 Hz, 1H), 3.98 (s, 3H), 3.97 (s, 3H); 13C-NMR (101 MHz; DMSO): δ 152.3, 146.3,

145.0, 142.2, 131.5, 129.8, 124.9, 119.3, 63.1, 61.9; IR (NaCl, film) 1639, 1519 cm-1;

HRMS (-APCI): [M-H]- 288.0149 calcd for C10H9ClN2O6, found: 288.0141.

REF: TRW-II-426, TRW-II-431, TRW-II-489, TRW-III-003, TRW-III-011, TRW-III-

084.

OMeMeO

Cl

NO2

NO2

4.7

235

Figure 5.65a. 1H NMR spectrum of compound 4.7.

Figure 5.65b. 13C NMR spectrum of compound 4.7.

2.962.96

2.842.84

1.021.02

11

1.111.11ppm0 011223344556677889-16.04

3.97

3.98

7.61

7.65

8.15

8.31

8.34

ppm0 05050100100150150-253.79

61.8

863

.06

119.

25

124.

87

129.

8113

1.51

142.

2414

6.27

152.

31

236

5-chloro-6,7-dimethoxy-1H-indole (4.6). A suspension of 4.7 (380 mg, 1.3 mmol), SiO2

(3.8 g, 1000 wt%), Fe powder (1.37 g, 24.4 mmol), AcOH (7.6 mL), and PhCH3 (13 mL)

was heated to reflux for 3 h, then cooled to r.t. and diluted with CH2Cl2. The mixture was

filtered and the solid washed with CH2Cl2. The filtrate was washed with NaHSO3 (5%

aq), NaHCO3 (sat’d aq), and brine, then dried and concentrated. The resulting oil was

dissolved in CH2Cl2 (13 mL) and SiO2 added until the suspension was just fluid. After

stirring under Ar for 30 min, the suspension was filtered and the solid washed with

MeOH. The filtrate was concentrated and the product deemed sufficiently pure as crude

(250 mg, 91% yield).

1H-NMR (400 MHz; CDCl3): δ 8.40 (bs, 1H), 7.27 (s, 1H), 7.03 (t, J = 2.8 Hz, 1H), 6.32

(t, J = 2.6 Hz, 1H), 3.94 (s, 3H), 3.82 (s, 3H).; 13C-NMR (101 MHz; CDCl3): δ 191.9,

142.7, 139.1, 130.6, 128.8, 121.1, 115.8, 102.5, 61.4, 61.0; IR (NaCl, film) 3357, 2939,

2831, 1655 cm-1; HRMS (ESI-APCI+): [M+H]+ 212.0478 calcd for C10H11ClNO2, found:

212.0472.

REF: TRW-II-430, TRW-II-434, TRW-II-490, TRW-III-086.

OMeMeO

Cl

NH

4.6

237

Figure 5.66a. 1H NMR spectrum of compound 4.6.

Figure 5.66b. 13C NMR spectrum of compound 4.6.

3.23.2

3.23.2

1.01.0

1.01.0

0.80.8

1.01.0

ppm0 011223344556677889-16.04

3.82

3.94

6.31

6.32

6.32

7.02

7.03

7.04

7.27

8.40

ppm0 05050100100150150200-253.79

61.0

061

.41

102.

45

115.

80

121.

10

128.

7913

0.59

142.

72

191.

91

238

5-chloro-6,7-dimethoxy-1-methyl-1H-indole (5.12). To a solution of indole 4.6 (126

mg, 0.59 mmol) in DMF (1.5 mL) at 0 °C was slowly added NaH (47 mg). After stirring

30 minutes, iodomethane (0.044 mL, 0.71 mmol) was added dropwise, and the reaction

stirred an additional 30 minutes at 0 °C. The reaction was warmed to r.t. and stirred 5 h

before adding water. The product was extracted in EtOAc, washed with water (2x) and

brine, then dried and concentrated to afford the title compound, deemed sufficiently pure

as crude (133 mg, >99% yield).

1H-NMR (300 MHz; CDCl3): δ 7.35 (s, 1H), 6.93 (d, J = 3.1 Hz, 1H), 6.34 (d, J = 3.1

Hz, 1H), 4.02 (s, 3H), 3.98 (s, 3H), 3.93 (s, 3H); 13C-NMR (101 MHz; CDCl3): δ 143.7,

140.6, 131.0, 126.9, 120.6, 116.0, 115.7, 100.6, 61.8, 61.2, 35.4; IR (NaCl, film) 2928,

1048 cm-1; HRMS (ESI-APCI+): [M+H]+ 226.0635 calcd for C11H13ClNO2, found:

226.0618.

REF: TRW-II-433, TRW-II-437, TRW-III-087.

OMeMeO

Cl

NMe

5.12

239

Figure 5.67a. 1H NMR spectrum of compound 5.12.

Figure 5.67b. 13C NMR spectrum of compound 5.12.

3.23.2

3.33.3

3.33.3

1.01.0

1.01.0

1.01.0

ppm1 12233445566778-19.97

3.93

3.98

4.02

6.33

6.34

6.92

6.93

7.35

ppm0 05050100100150150-253.79

35.4

3

61.2

261

.83

100.

64

115.

7411

6.05

120.

62

126.

8913

1.03

140.

6214

3.68

240

5-chloro-6,7-dimethoxy-1-methyl-1H-indole-3-carbaldehyde (4.20). DMF (6.5 mL)

was cooled to 0 °C, then POCl3 (0.586 mL, 6.42 mmol) added dropwise. Indole 5.12

(1.32 g, 5.84 mmol) was added dropwise as a solution in DMF. After warming the

mixture to 35 °C and stirring an additional 1.5 h, ice water was added followed by 2M

NaOH (15 mL). The resulting solution was boiled for 10 min, then cooled and diluted

with EtOAc. The organic layer was washed with water and brine, then dried and

concentrated to give the aldehyde (1.38 g, 93% yield), used as crude in the next reaction.

1H-NMR (400 MHz; CDCl3): δ 9.76 (s, 1H), 7.96 (s, 1H), 7.44 (s, 1H), 3.95 (s, 3H), 3.94

(s, 3H), 3.82 (s, 3H); 13C-NMR (101 MHz; CDCl3): δ 183.9, 145.4, 140.92, 140.89,

129.6, 124.5, 123.1, 117.26, 117.20, 61.9, 61.1, 36.7; IR (NaCl, film) 2930, 2853, 1656,

1044 cm-1; HRMS (ESI-APCI+): [M+H]+ 254.0584 calcd for C12H13ClNO3, found:

254.0581.

REF: TRW-II-436, TRW-II-439, TRW-III-090.

OMeMeO

Cl

NMe

CHO

4.20

241

Figure 5.68a. 1H NMR spectrum of compound 4.20.

Figure 5.68b. 13C NMR spectrum of compound 4.20.

3.03.0

2.82.8

3.23.2

1.01.0

0.80.8

0.90.9

ppm0 011223344556677889910-16.043.

823.

943.

95

7.44

7.96

9.76

ppm0 05050100100150150200-253.79

36.6

7

61.1

261

.87

117.

2011

7.26

123.

1412

4.55

129.

64

140.

8914

0.92

145.

44

183.

93

242

(E)-methyl 3-(5-chloro-6,7-dimethoxy-1-methyl-1H-indol-3-yl)acrylate (4.5). To a

solution of aldehyde 4.20 (133 mg, 0.52 mmol) in toluene (1.8 mL) was added

methyl(triphenyl-phosphoranylidene)acetate (347 mg, 1.04 mmol). The resulting solution

was heated to reflux O/N, then concentrated and purified by SiO2 chromatography (9:1 –

4:1 – 2:1 hexanes:EtOAc) to afford olefin 4.5 (128 mg, 80% yield).

1H-NMR (400 MHz; CDCl3): δ 7.67 (d, J = 16.0 Hz, 1H), 7.51 (s, 1H), 7.11 (s, 1H), 6.20

(d, J = 16.0 Hz, 1H), 3.93 (s, 3H), 3.89 (s, 3H), 3.83 (s, 3H), 3.71 (s, 3H); 13C-NMR (101

MHz; CDCl3): δ 168.4, 144.7, 141.0, 137.2, 134.8, 129.9, 124.3, 122.8, 115.7, 112.5,

111.4, 61.8, 61.2, 51.4, 36.1; IR (NaCl, film) 2940, 1736, 1626, 1043 cm-1; HRMS (ESI-

APCI+): [M+H]+ 310.0846 calcd for C15H17ClNO4, found: 310.0840.

REF: TRW-II-441, TRW-II-443, TRW-III-091.

OMeMeO

Cl

NMe

CO2Me

4.5

243

Figure 5.69a. 1H NMR spectrum of compound 4.5.

Figure 5.69b. 13C NMR spectrum of compound 4.5.

2.82.8

3.13.1

2.92.9

2.92.9

1.11.1

1.01.0

1.01.0

1.01.0

ppm0 0112233445566778-16.04

3.71

3.83

3.89

3.93

6.18

6.22

7.11

7.51

7.65

7.69

ppm0 05050100100150150-253.79

36.0

6

51.3

6

61.1

761

.85

111.

4111

2.45

115.

72

122.

7812

4.25

129.

87

134.

8313

7.25

141.

0214

4.70

168.

38

244

(2S,3R)-methyl 2-(((benzyloxy)carbonyl)amino)-3-(5-chloro-6,7-dimethoxy-1-

methyl-1H-indol-3-yl)-3-hydroxypropanoate (4.4). To a solution of benzyl carbamate

(178 mg, 1.18 mmol) in 0.4 M NaOH (2.5 mL) and n-propanol (2 mL) in a flask covered

with foil was added fresh t-butyl hypochlorite (0.135 mL, 1.20 mmol). After stirring 5

min, (DHQD)2AQN (14 mg, 0.016 mmol) in n-propanol (0.5 mL), olefin 4.5 (100 mg,

0.32 mmol) in n-propanol (1 mL), and K2OsO2(OH)4 (5 mg, 0.013 mmol) in 0.4 M

NaOH (0.5 mL) were added successively, and the resulting solution stirred O/N. Sodium

bisulfite (0.2 g) was added and the product extracted into EtOAc. The organic layer was

washed with water, then brine, dried over Na2SO4, and concentrated. SiO2

chromatography (10:1 – 1:1 hexanes:EtOAc) gave pure tryptophan derivative 4.4 (54 mg,

35% yield).

[α]D = -20.9 (CH2Cl2); 1H-NMR (300 MHz; CDCl3): δ 7.40 (s, 1H), 7.32 (s, 5H), 6.92

(s, 1H), 5.77 (d, J = 9.2 Hz, 1H), 5.46-5.45 (bs, 1H), 5.06 (s, 2H), 4.69-4.65 (m, 1H), 3.98

(s, 3H), 3.89 (s, 3H), 3.88 (d, J = 9.3 Hz, 3H), 3.78 (s, 3H), 2.69 (bs, 1H); 13C-NMR (101

MHz; CDCl3): δ 166.1, 144.6, 140.9, 133.75, 133.73, 128.66, 128.60, 128.46, 128.43,

128.28, 128.18, 128.07, 126.3, 122.6, 114.2, 108.6, 72.9, 67.3, 66.9, 61.9, 61.2, 52.3,

36.2; IR (NaCl, film) 3346, 2952, 1723, 1465, 1218, 1040 cm-1; HRMS (ESI-APCI+):

[M+Na]+ 499.1248 calcd for C23H25ClN2NaO7, found: 499.1249.

REF: TRW-II-459, TRW-II-461, TRW-III-093.

OMeMeO

Cl

NMe

CO2MeHONHCbz

4.4

245

Figure 5.70a. 1H NMR spectrum of compound 4.4.

Figure 5.70b. 13C NMR spectrum of compound 4.4.

0.80.8

3.03.0

2.72.7

2.92.9

2.72.7

0.70.7

2.02.0

0.90.9

0.70.7

1.01.0

5.05.0

1.21.2

ppm0 01122334455667788-19.97

2.68

2.70

3.78

3.86

3.89

3.98

4.65

4.66

4.66

4.69

5.06

5.45

5.45

5.46

5.76

5.79

6.92

7.32

7.40

ppm0 05050100100150150-253.79

36.2

2

52.3

2

61.2

361

.87

66.9

267

.32

72.8

6

108.

57

114.

1612

2.57

126.

3412

8.07

128.

1812

8.28

128.

4312

8.46

128.

6012

8.66

133.

7313

3.75

140.

8614

4.57

166.

05

246

1,4-difluoroanthracene-9,10-dione (4.23). To a solution of phthalic anhydride (10 g,

67.6 mmol) in 1,4-difluorobenzene (68 mL) was added AlCl3 (36 g, 270 mmol). The

resulting solution was stirred at reflux for 48h before removing the solvent by distillation.

HCl (1M, 400 mL) was added and the product extracted into CH2Cl2 (3x). The combined

organic extracts were filtered through a cotton plug and concentrated. The crude residue

was taken up in chloroform (40 mL) and to it added hexanes (100 mL). The solution was

cooled to -20 °C then filtered to collect the resultant precipitate. After drying the

precipitate under vacuum, polyphosphoric acid (50 mL) was added and the mixture

heated to 140 °C for 2h. The mixture was poured over approximately 400 g ice and

allowed to warm to r.t. while stirring. Solid K2CO3 was added to neutralize the solution,

then the product was extracted into CH2Cl2. The combined organic extracts were dried

(Na2SO4) and concentrated. The residue was dissolved in CH2Cl2, filtered through basic

alumina using CH2Cl2 as the eluent, and concentrated to afford the title compound (920

mg).

1H-NMR (400 MHz; CDCl3): δ 8.21-8.16 (m, 2H), 7.76-7.72 (m, 2H), 7.43-7.39 (m,

2H); 13C-NMR (101 MHz; CDCl3): δ 134.3, 133.2, 127.0, 124.7, 124.48, 124.34.

REF: TRW-II-451, TRW-III-079.

O

O F

F4.23

247

Figure 5.71a. 1H NMR spectrum of compound 4.23.

Figure 5.71b. 13C NMR spectrum of compound 4.23.

2.12.1

2.122.12

22

ppm1 122334455667788-16.04

7.39

7.41

7.41

7.43

7.72

7.72

7.73

7.74

7.74

7.75

7.76

7.76

8.16

8.17

8.18

8.18

8.19

8.20

8.21

8.21

ppm20 20404060608080100100120120140140-253.79

76.6

476

.96

77.2

8

124.

3412

4.48

124.

6812

7.01

133.

2413

4.30

248

(DHQD)2AQN (4.24). To a solution of DHQD (3.1 g, 9.5 mmol) in THF (19 mL) at -50

°C was added nBuLi (1.6M in hexanes, 6.0 mL, 9.5 mmol) dropwise over 10 min. After

stirring 15 min at -50 °C, the reaction mixture was warmed to 0 °C and the difluoro

compound added (920 mg, 3.8 mmol). The reaction was allowed to warm to r.t. as it

stirred O/N under Ar, then warmed to 40 °C and stirred an additional 2h. EtOAc was

added and the reaction quenched by addition of sat’d NaHCO3. The product was

extracted in EtOAc, dried (Na2SO4), concentrated, and purified by SiO2 chromatography

(5% MeOH, 0.5% NH4OH, in CHCl3) to afford (DHQD)2AQN (2.55 g, 78% yield).

1H-NMR (300 MHz; CDCl3): δ 8.63 (d, J = 4.5 Hz, 2H), 8.22 (dd, J = 5.8, 3.3 Hz, 2H),

8.01 (d, J = 9.2 Hz, 2H), 7.79 (dd, J = 5.8, 3.3 Hz, 2H), 7.49 (d, J = 4.5 Hz, 3H), 7.37

(dd, J = 9.2, 2.5 Hz, 3H), 6.80 (bs, 2H), 5.28 (s, 2H), 3.95 (s, 6H), 3.28-3.26 (bs, 2H),

3.00-2.53 (broad, 10H), 1.83 (bs, 2H), 1.61-1.45 (broad, 12H), 0.84 (t, J = 7.3 Hz, 6H).

REF: TRW-II-457, TRW-III-002, TRW-III-082.

O

O DHQD

DHQD(DHQD)2AQN (4.24)

249

Figure 5.72a. 1H NMR spectrum of compound 4.24.

6.36.3

12.312.3

2.12.1

10.410.4

2.02.0

6.06.0

2.12.1

1.81.8

2.72.7

3.33.3

1.91.9

1.81.8

1.81.8

1.61.6

ppm0 0112233445566778899-19.97

0.82

0.84

0.86

1.45

1.46

1.46

1.46

1.47

1.54

1.55

1.55

1.56

1.56

1.58

1.59

1.59

1.61

1.83

1.83

2.53

2.53

2.76

2.77

2.88

2.88

2.88

2.88

2.89

2.91

2.91

2.91

2.91

2.92

3.00

3.26

3.26

3.27

3.27

3.27

3.27

3.27

3.28

3.28

3.95

5.28

6.80

6.80

7.35

7.36

7.38

7.39

7.48

7.50

7.78

7.79

7.80

7.81

8.00

8.03

8.21

8.22

8.23

8.24

8.62

8.64

250

(2S,3R)-methyl 2-(((benzyloxy)carbonyl)amino)-3-((tert-butyldimethylsilyl) oxy)-3-

(5-chloro-6,7-dimethoxy-1-methyl-1H-indol-3-yl)propanoate (4.25). Alcohol 4.4 (296

mg, 0.62 mmol) was dissolved in DMF (1.6 mL), and to it added imidazole (102 mg, 1.5

mmol) and TBSCl (242 mg, 1.6 mmol). After stirring O/N, the solution was diluted with

EtOAc, washed with water (3x), brine (1x), dried over Na2SO4, and concentrated to give

the title compound (366 mg, >99% yield).

1H-NMR (300 MHz; CDCl3): δ 7.36 (s, 1H), 7.32 (s, 5H), 6.83 (s, 1H), 5.49 (dd, J =

12.6, 2.3 Hz, 1H), 5.07 (s, 2H), 4.98 (s, 1H), 4.47 (dd, J = 9.6, 2.1 Hz, 1H), 4.00 (s, 3H),

3.91 (s, 3H), 3.89 (s, 3H), 3.79 (s, 3H), 0.86 (s, 9H), 0.09 (s, 6H); HRMS (ESI-APCI+):

[M+Na]+ 613.2113 calcd for C29H39ClN2NaO7Si, found: 613.2109.

REF: TRW-II-460, TRW-II-465, TRW-III-096.

OMeMeO

Cl

NMe

CO2MeTBSONHCbz

4.25

251

Figure 5.73a. 1H NMR spectrum of compound 4.25.

5.85.8

9.49.4

2.92.9

3.33.3

2.72.7

3.43.4

0.70.7

1.11.1

1.91.9

1.31.3

0.90.9

4.74.7

1.01.0

ppm0 011223344556677-19.97

0.09

0.86

3.79

3.89

3.91

4.00

4.45

4.45

4.48

4.49

4.98

5.07

5.46

5.47

5.50

5.51

6.83

7.32

7.36

252

(2S,3R)-methyl 2-amino-3-((tert-butyldimethylsilyl)oxy)-3-(5-chloro-6,7-dimethoxy-

1-methyl-1H-indol-3-yl)propanoate (4.26). To a suspension of 4.25 (366 mg, 0.61

mmol) and Pd/C (37 mg) in MeOH (3 mL) was added hydrogen gas. After stirring O/N,

the reaction was flushed with argon and filtered through celite to afford the free amine

(262 mg, 94% yield).

[α]D = +6.4 (CH2Cl2); 1H-NMR (300 MHz; CDCl3): δ 7.32 (s, 1H), 6.90 (s, 1H), 5.40 (d,

J = 1.8 Hz, 1H), 4.85-4.63 (m, 1H), 4.00 (s, 3H), 3.94 (s, 3H), 3.90 (s, 3H), 3.75 (s, 3H),

2.24-2.14 (bs, 2H), 0.88 (s, 9H), -0.01 (s, 3H), -0.15 (s, 3H); 13C-NMR (101 MHz;

CDCl3): δ 173.3, 143.8, 140.7, 129.3, 129.0, 123.9, 120.6, 114.8, 114.3, 70.4, 61.8, 61.1,

60.8, 52.0, 35.5, 25.66, 25.61, 25.52, 18.0, -4.6, -5.6; IR (NaCl, film) 3332, 2953, 2858,

1750, 1464, 1038 cm-1; HRMS (ESI-APCI+): [M+Na]+ 479.1745 calcd for

C21H33ClN2NaO5Si, found: 479.1746.

REF: TRW-II-462, TRW-II-468, TRW-III-098.

OMeMeO

Cl

NMe

CO2MeTBSONH2

4.26

253

Figure 5.74a. 1H NMR spectrum of compound 4.26.

Figure 5.74b. 13C NMR spectrum of compound 4.26.

2.72.7

2.92.9

9.09.0

2.02.0

2.92.9

2.82.8

2.92.9

3.43.4

1.31.3

0.90.9

0.80.8

1.01.0

ppm0 011223344556677-14.99

-0.1

5-0

.01

0.88

2.14

2.14

2.15

2.15

2.16

2.16

2.17

2.17

2.17

2.18

2.19

2.19

2.19

2.20

2.20

2.21

2.21

2.23

2.23

2.24

3.75

3.90

3.94

4.00

4.63

4.64

4.66

4.84

4.85

5.39

5.40

6.90

6.90

7.32

ppm0 05050100100150150-253.79

-5.6

2-4

.57

18.0

2

25.5

225

.61

25.6

6

35.5

0

51.9

8

60.7

661

.12

61.7

5

70.3

9

114.

2811

4.82

120.

5812

3.95

128.

9612

9.32

140.

6814

3.84

173.

34

254

(2S,3R)-methyl 2-((S)-2-((tert-butoxycarbonyl)amino)propanamido)-3-((tert-butyl-

dimethylsilyl)oxy)-3-(5-chloro-6,7-dimethoxy-1-methyl-1H-indol-3-yl)propanoate

(4.27). To a solution of amine 4.26 (262 mg, 0.57 mmol) in CH2Cl2 (3 mL) was added N-

Boc-ala-OH (108 mg, 0.57 mmol), iPr2NEt (0.1 mL, 0.57 mmol), and EDCI (110 mg,

0.57 mmol). The reaction was stirred at r.t. O/N, the concentrated and purified by SiO2

chromatography (10% - 25% - 50% EtOAc in hexanes) to afford 4.27 (210 mg, 59%

yield).

1H-NMR (300 MHz; CDCl3): δ 7.36 (s, 1H), 6.86 (s, 1H), 6.73-6.70 (bs, 1H), 5.53 (d, J

= 2.0 Hz, 1H), 4.97-4.96 (bs, 1H), 4.67 (dd, J = 9.1, 1.7 Hz, 1H), 4.19-4.15 (bs, 1H), 3.99

(s, 3H), 3.92 (s, 3H), 3.90 (s, 3H), 3.78 (s, 3H), 1.45 (s, 9H), 1.24 (dd, J = 7.1, 0.8 Hz,

3H), 0.89 (s, 9H), 0.01 (s, 3H), -0.15 (s, 3H).

REF: TRW-II-466, TRW-II-471.

NMe

Cl

MeOOMe

HNNHBoc

CO2Me

OMe

TBSO

4.27

255

Figure 5.75a. 1H NMR spectrum of compound 4.27.

2.72.7

2.62.6

9.19.1

3.03.0

8.98.9

2.72.7

2.92.9

2.92.9

2.92.9

1.21.2

0.90.9

0.90.9

0.90.9

0.70.7

0.90.9

1.01.0

ppm0 0112233445566778-19.97

-0.1

50.

01

0.89

1.23

1.23

1.25

1.26

1.45

3.78

3.90

3.92

3.99

4.15

4.15

4.17

4.17

4.18

4.18

4.18

4.18

4.18

4.19

4.65

4.65

4.68

4.68

4.96

4.96

4.96

4.97

4.97

4.97

5.52

5.53

6.70

6.70

6.70

6.71

6.72

6.72

6.72

6.72

6.73

6.86

7.36

256

(2S,3R)-methyl 2-((S)-2-aminopropanamido)-3-((tert-butyldimethylsilyl)oxy)-3-(5-

chloro-6,7-dimethoxy-1-methyl-1H-indol-3-yl)propanoate (4.28). Compound 4.27

(105 mg, 0.167 mmol) was dissolved in CH2Cl2 (1.7 mL), then 2,6-lutadine (0.039 mL,

0.334 mmol) added. The solution was cooled to 0 °C, then TMSOTf (0.045 mL, 0.251

mmol) added. After warming to r.t., the reaction was monitored by TLC and once

complete, quenched with sat’d NH4Cl. The product was extracted into CH2Cl2, the

organic extracts dried and concentrated, and the product purified by SiO2

chromatography to afford the title compound (52 mg, 59% yield).

1H-NMR (300 MHz; CDCl3): δ 7.36 (s, 1H), 6.92 (s, 1H), 5.53 (s, 1H), 4.79-4.77 (bs,

2H), 4.59 (d, J = 8.8 Hz, 1H), 3.97 (s, 3H), 3.90 (s, 3H), 3.88 (s, 3H), 3.82 (d, J = 7.4 Hz,

1H), 3.74 (s, 3H), 1.29 (d, J = 8.4 Hz, 3H), 0.87 (s, 9H), -0.01 (s, 3H), -0.16 (s, 3H).

REF: TRW-II-476; for TFA version, see TRW-II-474.

NMe

Cl

MeOOMe

HNNH2

CO2Me

OMe

TBSO

4.28

257

Figure 5.76a. 1H NMR spectrum of compound 4.28.

3.03.0

2.72.7

9.19.1

2.72.7

2.92.9

1.21.2

2.72.7

2.72.7

2.72.7

1.01.0

2.02.0

1.01.0

1.01.0

1.41.4

ppm0 0112233445566778-19.97

-0.1

6-0

.01

0.87

1.27

1.30

3.74

3.80

3.83

3.88

3.90

3.97

4.57

4.60

4.77

4.77

4.78

4.78

4.78

4.79

4.79

5.53

6.92

7.36

258

(S,Z)-3-((5-chloro-6,7-dimethoxy-1-methyl-1H-indol-3-yl)methylene)-6-

methylpiperazine-2,5-dione (4.29). To a solution of amine 4.28 (69 mg, 0.167 mmol) in

toluene (5 mL) was added a small amount of 2-OH-pyridine. The solution was heated to

reflux O/N, then cooled to r.t. The solvent was decanted off and the remaining solid

washed with toluene and dried to afford pure 4.29 (37 mg, 61% yield).

1H-NMR (300 MHz; DMSO-d6): δ 9.43 (s, 1H), 8.32 (s, 1H), 7.87 (s, 1H), 7.43 (s, 1H),

6.81 (s, 1H), 4.11 (q, J = 6.9 Hz, 1H), 3.96 (s, 3H), 3.93 (s, 3H), 3.81 (s, 3H), 1.32 (d, J =

6.8 Hz, 3H); 13C-NMR (101 MHz; DMSO): δ 167.9, 160.77, 160.73, 144.2, 141.0, 133.1,

128.4, 126.3, 123.8, 120.9, 114.2, 107.2, 106.2, 62.4, 61.4, 50.7, 36.0, 19.9; HRMS (ESI-

APCI+): [M+H]+ 364.1064 calcd for C17H19ClN3O4, found: 364.1075.

REF: TRW-II-475, TRW-II-477.

NMe

Cl

MeOOMe

HNNH

O

OMe

4.29

259

Figure 5.77a. 1H NMR spectrum of compound 4.29.

Figure 5.77b. 13C NMR spectrum of compound 4.29.

3.33.3

3.13.1

3.13.1

3.03.0

1.11.1

1.11.1

1.11.1

1.11.1

1.01.0

0.90.9

ppm0 0112233445566778899-19.97

1.31

1.33

3.81

3.93

3.96

4.08

4.10

4.12

4.15

6.81

7.43

7.87

8.32

9.43

ppm0 05050100100150150-253.79

19.8

6

35.9

7

50.7

3

61.4

262

.41

106.

1610

7.21

114.

17

120.

8712

3.79

126.

2612

8.40

133.

09

140.

9914

4.19

160.

7316

0.77

167.

85

260

(2S,3R)-2-(((benzyloxy)carbonyl)amino)-3-((tert-butyldimethylsilyl)oxy)-3-(5-chloro-

6,7-dimethoxy-1-methyl-1H-indol-3-yl)propanoic acid (4.30). To a solution of methyl

ester 4.25 (682 mg, 1.2 mmol) in THF/H2O (2:1, 2.6:1.3 mL) was added LiOH (144 mg,

6.0 mmol) and the resulting suspension stirred O/N. The reaction mixture was acidified

with 10% KHSO4, then the product extracted in EtOAc. The combined organic extracts

were washed with brine, dried over Na2SO4, and concentrated to afford 4.30 (545 mg,

82% yield).

1H-NMR (300 MHz; CDCl3): δ 7.37-7.28 (m, 6H), 6.87 (bs, 1H), 5.62-5.58 (broad, 1H),

5.10-5.00 (m, 2H), 4.70 (s, 1H), 4.51-4.49 (bs, 1H), 3.99-3.89 (broad, 9H), 0.91-0.86 (m,

9H), 0.10-0.01 (m, 6H).

REF: TRW-III-023.

OMeMeO

Cl

NMe

CO2HTBSONHCbz

4.30

261

Figure 5.78a. 1H NMR spectrum of compound 4.30.

6.06.0

9.09.0

9.49.4

1.01.0

0.60.6

2.12.1

1.41.4

1.01.0

6.56.5

ppm0 011223344556677-19.97

0.01

0.10

0.10

0.86

0.87

0.88

0.91

0.91

3.89

3.99

4.49

4.50

4.50

4.50

4.50

4.50

4.50

4.51

4.70

5.00

5.01

5.01

5.01

5.02

5.02

5.02

5.03

5.10

5.58

5.58

5.59

5.59

5.61

5.61

5.61

5.61

5.62

5.62

5.62

5.62

6.87

6.87

6.87

7.28

7.34

7.34

7.36

7.37

262

(S)-methyl 2-((2S,3R)-2-(((benzyloxy)carbonyl)amino)-3-((tert-

butyldimethylsilyl)oxy)-3-(5-chloro-6,7-dimethoxy-1-methyl-1H-indol-3-

yl)propanamido)propanoate (4.31). To a solution of acid 4.30 (100 mg, 0.173 mmol),

L-Ala-OMe·HCl (24 mg, 0.173 mmol), and iPr2NEt (0.075 mL, 0.433 mmol) in CH2Cl2

(2 mL) was added T3P (50 wt % in DMF, 127 mg of solution). The resulting solution

was stirred O/N under Ar at r.t., then quenched by addition of water. The product was

extracted in EtOAc and the combined organic layers dried over Na2SO4 and concentrated

to afford the crude material. Purification by SiO2 chromatography (10-40% EtOAc in

hexanes) gave 4.31 (39 mg, 34% yield).

1H-NMR (300 MHz; CDCl3): δ 7.58 (d, J = 7.4 Hz, 1H), 7.35 (d, J = 3.1 Hz, 5H), 7.04

(s, 1H), 5.56 (dd, J = 29.0, 5.4 Hz, 2H), 5.13-5.10 (broad, 3H), 4.76-4.74 (bs, 1H), 4.57-

4.50 (m, 1H), 3.98 (s, 3H), 3.89 (d, J = 3.7 Hz, 6H), 3.78 (s, 3H), 1.42 (d, J = 7.2 Hz,

3H), 0.94 (s, 9H), 0.06 (d, J = 48.3 Hz, 6H).

REF: TRW-III-026.

NMe

Cl

MeOOMe

NHCbz

TBSO O

NH

CO2Me

Me

4.31

263

Figure 5.79a. 1H NMR spectrum of compound 4.31.

6.36.3

9.19.1

2.82.8

3.03.0

6.16.1

3.23.2

1.31.3

1.01.0

2.82.8

1.91.9

0.80.8

7.57.5

0.90.9

ppm0 011223344556677-19.97

-0.0

20.

14

0.94

1.40

1.43

3.78

3.88

3.89

3.98

4.50

4.51

4.53

4.55

4.57

4.74

4.74

4.75

4.75

4.75

4.75

4.76

4.76

5.10

5.13

5.50

5.51

5.59

5.62

7.04

7.28

7.34

7.35

7.57

7.60

264

(2S,3R)-methyl 2-((S)-2-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)propan-

amido)-3-((tert-butyldimethylsilyl)oxy)-3-(5-chloro-6,7-dimethoxy-1-methyl-1H-

indol-3-yl)propanoate (4.32). To a solution of amine 4.26 (100 mg, 0.22 mmol) in

CH2Cl2 (2.2 mL) was added N-Fmoc-Ala (68 mg, 0.22 mmol), iPr2NEt (0.1 mL, 0.55

mmol), and T3P (160 mg 50 wt% solution in DMF, 0.25 mmol). The resulting solution

was allowed to stir O/N at r.t. under Ar, then quenched with water and the product

extracted in EtOAc. The combined organic layers were dried over Na2SO4, filtered, and

concentrated. Purification by SiO2 chromatography (1:1 hexanes:EtOAc) afforded 4.32 in

59% yield (97 mg).

[α]D = -3.9 (CH2Cl2); 1H-NMR (300 MHz; CDCl3): δ 7.76 (d, J = 7.5 Hz, 2H), 7.58 (d, J

= 7.4 Hz, 2H), 7.40 (d, J = 7.5 Hz, 2H), 7.36 (s, 1H), 7.31 (d, J = 7.4 Hz, 2H), 6.85 (s,

1H), 6.65 (s, 1H), 5.54 (d, J = 1.4 Hz, 1H), 5.41-5.38 (m, 1H), 4.68 (dd, J = 9.0, 1.7 Hz,

1H), 4.33-4.27 (m, 2H), 3.97 (s, 3H), 3.89 (s, 3H), 3.86 (s, 3H), 3.79 (s, 3H), 1.36 (d, J =

6.8 Hz, 3H), 0.89 (s, 9H), 0.02 (s, 3H), -0.13 (s, 3H); 13C-NMR (101 MHz; CDCl3): δ

171.4, 165.8, 156.4, 144.4, 143.7, 143.3, 141.27, 141.15, 140.9, 135.1, 128.3, 127.78,

127.70, 127.5, 127.10, 127.06, 126.5, 124.87, 124.82, 122.4, 119.98, 119.92, 117.9,

113.9, 108.1, 67.0, 61.8, 61.2, 52.3, 51.5, 47.1, 36.0, 25.61, 25.48, 25.3, 17.94, 17.91, -

3.6; IR (NaCl, film) 3302, 2953, 2857, 1714, 1462, 1254, 1040 cm-1; HRMS (ESI-

APCI+): [M+Na]+ 772.2797 calcd for C39H48ClN3NaO8Si, found: 772.2799.

REF: TRW-III-056, TRW-III-064, TRW-III-113.

NMe

Cl

MeOOMe

HNNHFmoc

CO2Me

OMe

TBSO

4.32

265

Figure 5.80a. 1H NMR spectrum of compound 4.32.

Figure 5.80b. 13C NMR spectrum of compound 4.32.

2.82.8

2.82.8

9.39.3

2.52.5

2.62.6

2.82.8

2.92.9

3.13.1

2.12.1

0.90.9

0.70.7

1.01.0

0.70.7

0.90.9

1.91.9

1.21.2

2.02.0

1.91.9

2.02.0

ppm0 01122334455667788-19.97

-0.1

30.

02

0.89

1.34

1.37

3.79

3.86

3.89

3.97

4.27

4.28

4.31

4.33

4.33

4.33

4.66

4.67

4.69

4.70

5.38

5.38

5.38

5.41

5.41

5.54

5.54

6.63

6.66

6.85

7.30

7.32

7.36

7.39

7.42

7.57

7.60

7.75

7.77

ppm0 05050100100150150-253.79

-3.6

1

17.9

117

.94

25.2

625

.48

25.6

1

35.9

8

47.1

151

.47

52.2

8

61.2

161

.83

67.0

4

108.

0511

3.90

117.

8711

9.92

119.

9812

2.45

124.

8212

4.87

126.

5012

7.06

127.

1012

7.48

127.

7012

7.78

128.

2613

5.08

140.

8714

1.15

141.

2714

3.32

143.

6814

4.44

156.

39

165.

75

171.

44

266

(6S)-3-((R)-((tert-butyldimethylsilyl)oxy)(5-chloro-6,7-dimethoxy-1-methyl-1H-

indol-3-yl)methyl)-6-methylpiperazine-2,5-dione (4.3). Dipeptide 4.32 (350 mg, 0.47

mmol) was dissolved in THF (20 mL), then morpholine (4.7 mL) added. The resulting

solution was stirred O/N at r.t., then concentrated. The crude residue was purified by SiO2

chromatography (10% methanol in CH2Cl2) to afford 181 mg of the title dioxopiperazine

(78% yield).

[α]D = -5.8 (CH2Cl2); 1H-NMR (300 MHz; CDCl3): δ 7.31 (s, 1H), 6.98 (s, 1H), 5.67 (s,

2H), 4.13 (q, J = 6.7 Hz, 1H), 4.05 (d, J = 1.5 Hz, 1H), 4.02 (s, 3H), 3.97 (s, 3H), 3.91 (s,

3H), 3.76 (s, 1H), 1.61 (d, J = 7.0 Hz, 3H), 0.88 (s, 9H), 0.03 (s, 3H), -0.03 (s, 3H); 13C-

NMR (101 MHz; CDCl3): δ 166.5, 160.4, 140.9, 136.1, 129.7, 127.4, 125.7, 122.7, 114.6,

108.6, 61.9, 61.2, 51.6, 51.3, 36.02, 36.00, 25.8, 25.6, 25.3, 20.4, 17.9, -1.6, -3.6; IR

(NaCl, film) 3225, 2932, 1681, 1449, 1259, 1042 cm-1; HRMS (ESI-APCI+): [M+Na]+

518.1854 calcd for C23H34ClN3NaO5Si, found: 518.1852.

REF: TRW-III-057, TRW-III-065.

NMe

Cl

MeOOMe

HNNH

O

OMe

TBSO

4.3

267

Figure 5.81a. 1H NMR spectrum of compound 4.3.

Figure 5.81b. 1H NMR spectrum of compound 4.3.

3.03.0

3.43.4

10.810.8

2.92.9

1.11.1

3.33.3

3.13.1

2.82.8

1.11.1

1.21.2

1.91.9

1.01.0

1.71.7

ppm0 0112233445566778-19.97

-0.0

30.

03

0.88

1.59

1.62

3.76

3.91

3.97

4.02

4.04

4.05

4.09

4.11

4.14

4.16

5.67

6.98

7.31

ppm0 05050100100150150-253.79

-3.6

2-1

.58

17.9

420

.43

25.2

625

.60

25.8

3

36.0

036

.02

51.3

351

.56

61.2

161

.85

108.

58

114.

65

122.

6712

5.72

127.

4112

9.73

136.

06

140.

86

160.

37

166.

53

268

(2S,3S,3aS,8aR)-1-benzyl 2-methyl 3-((tert-butyldimethylsilyl)oxy)-5-chloro-3a-

hydroxy-6,7-dimethoxy-8-methyl-3,3a,8,8a-tetrahydropyrrolo[2,3-b]indole-1,2(2H)-

dicarboxylate (4.40). Indole 4.25 (70 mg, 0.118 mmol) was dissolved in CH2Cl2 (6 mL)

and the resulting solution cooled to -78 °C. Freshly prepared DMDO (0.056 M in

acetone, 3 mL, 0.165 mmol) was added and the reaction mixture allowed to warm to r.t.

O/N. Sodium thiosulfate (sat’d aq) was added and the product extracted in CH2Cl2. The

combined organic extracts were washed with brine, dried over Na2SO4, and concentrated.

Purification by pTLC (40% EtOAc in hexanes) afforded pure 4.25 (6.2 mg, 9% yield).

1H-NMR (400 MHz; CDCl3): δ 7.46-7.40 (m, 5H), 7.05 (t, J = 9.1 Hz, 1H), 5.59 (s, 1H),

5.42-5.37 (m, 2H), 5.11-5.08 (m, 1H), 4.67-4.61 (m, 1H), 3.97-3.87 (m, 6H), 3.55 (s,

3H), 3.39 (s, 3H), 0.96 (s, 9H), 0.04 (d, J = 36.9 Hz, 6H); HRMS (ESI-APCI+): [M+H]+

607.2242 calcd for C29H40ClN2O8Si, found: 607.2240.

REF: TRW-III-107, TRW-III-110.

NMe

Cl

MeOOMe

NCbzH

HOTBSO

CO2Me

4.40

269

Figure 5.82a. 1H NMR spectrum of compound 4.40.

6.06.0

9.79.7

2.72.7

2.62.6

6.26.2

1.31.3

1.21.2

2.02.0

0.90.9

1.21.2

5.05.0

ppm0 011223344556677-16.04

0.00

0.09

0.96

3.39

3.55

3.87

3.89

3.92

3.95

3.97

4.61

4.64

4.64

4.65

4.67

5.08

5.11

5.11

5.37

5.38

5.40

5.41

5.42

5.59

7.03

7.05

7.07

7.40

7.40

7.40

7.40

7.41

7.42

7.42

7.43

7.44

7.44

7.45

7.45

7.46

270

(2S,3R)-methyl 3-acetoxy-2-(((benzyloxy)carbonyl)amino)-3-(5-chloro-6,7-

dimethoxy-1-methyl-1H-indol-3-yl)propanoate (4.47). To a solution of alcohol 4.4

(250 mg, 0.52 mmol) in THF (2.5 mL) was added Ac2O (0.059 mL, 0.62 mmol), Et3N

(0.109 mL, 0.78 mmol), and DMAP (small amt). The resulting solution was stirred at r.t.

O/N, then concentrated and 1M HCl added. The product was extracted in CH2Cl2,

washed with brine, dried over Na2SO4, and concentrated. The crude material was purified

by SiO2 chromatography (1:1 hexanes:EtOAc) to afford 4.47 (150 mg, 56% yield).

1H-NMR (300 MHz; CDCl3): δ 7.38-7.35 (broad, 6H), 6.92 (d, J = 38.1 Hz, 1H), 5.61-

5.54 (m, 1H), 5.18 (s, 1H), 5.09-5.06 (m, 2H), 4.82 (dd, J = 9.7, 3.7 Hz, 1H), 4.00-3.98

(m, 3H), 3.92-3.87 (m, 6H), 3.75 (d, J = 7.0 Hz, 3H), 2.17 (s, 3H).

REF: TRW-III-137.

OMeMeO

Cl

NMe

CO2MeAcONHCbz

4.47

271

Figure 5.83a. 1H NMR spectrum of compound 4.47.

2.92.9

3.13.1

7.17.1

3.23.2

0.50.5

1.81.8

0.90.9

1.01.0

0.80.8

7.07.0

ppm1 1223344556677-14.99

2.17

3.74

3.76

3.81

3.87

3.89

3.90

3.90

3.91

3.92

3.98

3.99

4.00

4.80

4.81

4.83

4.84

5.06

5.08

5.09

5.18

5.54

5.58

5.58

5.58

5.60

5.61

6.84

6.97

7.35

7.38

272

(2S,3R)-methyl 3-acetoxy-2-amino-3-(5-chloro-6,7-dimethoxy-1-methyl-1H-indol-3-

yl)propanoate (4.48). Compound 4.47 (150 mg, 0.29 mmol) was dissolved in MeOH

(1.5 mL) and to it added Pd/C (15 mg). The suspension was treated with H2 (atm) O/N,

then filtered through celite and concentrated to afford crude 4.48, used as crude in the

next step.

REF: TRW-III-141.

(3R)-methyl 2-((S)-2-((((9H-fluoren-9-

yl)methoxy)carbonyl)(methyl)amino)propanamido)-3-acetoxy-3-(5-chloro-6,7-

dimethoxy-1-methyl-1H-indol-3-yl)propanoate (4.49). Crude 4.48 (78 mg, 0.203

mmol) was dissolved in CH2Cl2 (2 mL) and to it added N-Me,Fmoc-Ala (66 mg, 0.203

mmol), iPr2NEt (0.088 mL, 0.233 mmol), and T3P (150 mg 50 wt% solution in DMF,

0.233 mmol). The resulting solution was allowed to stir O/N at r.t. under Ar, then

quenched with water and the product extracted in EtOAc. The combined organic layers

were dried over Na2SO4, filtered, and concentrated. Purification by SiO2 chromatography

afforded 4.49 in 18% yield (25 mg). A clean NMR could not be obtained for this sample.

REF: TRW-III-145.

OMeMeO

Cl

NMe

CO2MeAcONH2

4.48

NMe

Cl

MeOOMe

HN

AcO

NMeFmocCO2Me

OMe

4.49

273

(S,Z)-methyl 3-(5-chloro-6,7-dimethoxy-1-methyl-1H-indol-3-yl)-2-(2-

(methylamino)propanamido)acrylate (4.50). Dipeptide 4.49 (25 mg, 0.037 mmol) was

dissolved in THF (1.5 mL), then morpholine (0.37 mL) added. The resulting solution was

stirred O/N at r.t., then concentrated. The crude residue was purified by SiO2

chromatography (10% methanol in CH2Cl2) to afford the title product as a 1:1 mixture of

diasteromers (trace yield).

1H-NMR (400 MHz; CDCl3): δ 7.44 (d, J = 17.9 Hz, 2H), 6.86 (d, J = 4.3 Hz, 2H), 5.07

(dt, J = 15.5, 4.8 Hz, 2H), 4.84-4.77 (m, 2H), 3.98 (s, 3H), 3.96 (s, 3H), 3.90 (s, 3H), 3.89

(s, 3H), 3.77 (s, 3H), 3.71 (s, 2H), 3.66 (s, 3H), 3.32 (s, 3H), 3.27 (s, 3H), 2.40 (s, 3H),

2.29 (s, 3H), 1.18 (d, J = 6.9 Hz, 3H), 0.94 (d, J = 7.0 Hz, 3H); HRMS (ESI-APCI+):

[M+H]+ 410.1483 calcd for C19H25ClN3O5, found: 410.1471.

NMe

Cl

MeOOMe

HNNHMe

CO2Me

OMe

4.50

274

Figure 5.84a. 1H NMR spectrum of compound 4.50.

33

3.133.13

2.892.89

2.842.84

3.033.03

2.642.64

2.792.79

2.392.39

2.812.81

2.952.95

3.283.28

2.832.83

2.652.65

1.791.79

1.871.87

1.941.94

2.052.05

ppm0 011223344556677-16.04

0.93

0.95

1.17

1.19

2.29

2.40

3.27

3.32

3.66

3.71

3.77

3.89

3.90

3.96

3.98

4.77

4.78

4.80

4.80

4.82

4.84

5.04

5.05

5.07

5.08

5.09

5.10

6.85

6.86

7.42

7.46

275

(S)-(9H-fluoren-9-yl)methyl 4-methyl-5-oxooxazolidine-3-carboxylate (4.45). To a

solution of N-Fmoc-Ala (5.0 g, 16.1 mmol) in PhMe (50 mL) was added

paraformaldehyde (3.15 g, 105 mmol) and p-TsOH (247 mg, 1.3 mmol). The resulting

suspension was heated to reflux for 1 h with a Dean-Stark trap in place. Upon cooling,

EtOAc was added and the organic phase washed with sat’d NaHCO3, dried over Na2SO4,

and concentrated to afford 4.45, carried forward as crude.

1H-NMR (300 MHz; CDCl3): δ 7.78 (d, J = 7.3 Hz, 2H), 7.54 (d, J = 7.4 Hz, 2H), 7.38

(dt, J = 26.6, 7.4 Hz, 4H), 5.38-5.14 (broad, 3H), 4.61-4.60 (broad, 2H), 4.25 (t, J = 5.6

Hz, 1H), 1.51-1.14 (broad, 3H).

REF: TRW-III-125.

OFmocN

MeO

4.45

276

Figure 5.85a. 1H NMR spectrum of compound 4.45.

3.63.6

1.31.3

2.32.3

2.62.6

4.34.3

2.12.1

2.02.0

ppm1 12233445566778-14.99

1.14

1.14

1.14

1.15

1.15

1.16

1.16

1.16

1.17

1.17

1.49

1.50

1.50

1.50

1.50

1.51

1.51

1.51

4.23

4.25

4.26

4.60

4.60

4.60

4.61

4.61

4.61

5.14

5.14

5.15

5.32

5.33

5.34

5.34

5.34

5.35

5.35

5.35

5.36

5.36

5.37

5.37

5.37

5.37

5.38

7.31

7.33

7.36

7.40

7.42

7.44

7.53

7.55

7.77

7.79

277

N-Me,Fmoc-L-Ala-OH (4.46). Compound 4.45 (crude, directly from previous reaction)

was dissolved in CHCl3 (4.1 mL) and to the resulting solution added TFA (4.1 mL) and

Et3SiH (1.0 mL). After stirring 10 h, additional Et3SiH (3.0 mL) was added. The reaction

stirred another 24 h, then was concentrated and TFA removed by repeated azeotropic

distillations with toluene. The crude product was recrystallized from ether/EtOAc to

afford pure 4.46 (2.87 g, 55% yield over 2 steps).

1H-NMR (300 MHz; CDCl3): δ 7.77 (d, J = 7.2 Hz, 2H), 7.61-7.53 (m, 2H), 7.43-7.29

(m, 4H), 4.76 (dq, J = 84.6, 7.2 Hz, 1H), 4.44 (qd, J = 11.3, 9.0 Hz, 2H), 4.28 (t, J = 6.9

Hz, 1H), 2.92 (s, 3H), 1.42 (dd, J = 28.0, 7.2 Hz, 3H).

REF: TRW-III-126.

NMeFmoc

Me CO2H4.46

278

Figure 5.86a. 1H NMR spectrum of compound 4.46.

3.333.33

3.123.12

1.111.11

2.192.19

1.071.07

4.144.14

2.112.11

22

ppm0 01122334455667788-14.99

1.36

1.39

1.46

1.48

2.92

4.25

4.28

4.30

4.37

4.41

4.43

4.44

4.47

4.50

4.59

4.61

4.64

4.66

4.87

4.89

4.92

4.94

7.29

7.29

7.31

7.34

7.38

7.40

7.43

7.53

7.53

7.56

7.59

7.59

7.59

7.61

7.76

7.78

279

N-Bn-L-Ser-OH (5.13). To a solution of serine (14.7 g, 140 mmol) in 2M NaOH (70

mL) was added freshly distilled benzaldehyde (14.0 mL, 138 mmol). After stirring for 1 h

at r.t., the reaction mixture was cooled to 0 °C and NaBH4 (1.5 g, 40 mmol) added

portionwise such that the internal temperature did not exceed 10 °C. The resulting

solution was stirred 30 min at 5 °C, 1 h at r.t., then recooled to 0 °C. Additional NaBH4

(1.5 g, 40 mmol) was added portionwise as before. The reaction was allowed to warm to

r.t. as it stirred O/N. The crude reaction mixture was washed with ether (3x), then the

aqueous phase acidified to pH 5 with HCl. The resulting precipitate was filtered and

washed with water, then dried O/N to afford the crude product (6.55 g, 24% yield).

REF: TRW-III-288.

CO2H

NHBnHO

5.13

280

N-Me,Bn-L-Ser-OH (5.14). N-Bn-Ser (5.13, 2.49 g, 12.8 mmol) was dissolved in formic

acid (1.6 mL, 38.4 mmol) and formaldehyde (37% in H2O, 1.1 mL, 15.4 mmol) and

heated to 90 °C for 2 h. The reaction mixture was concentrated, then acetone added and a

solid triturated out of the solution. The suspension was concentrated, acetone added, and

the trituration process repeated 3x. Following the final concentration, acetone was again

added and the suspension allowed to stir 1 h at r.t, then 3 h at -20 °C. The solid was

filtered, washed with cold acetone, and dried to afford 5.14 (1.47 g, 55% yield).

1H-NMR (300 MHz; CD3OD): δ 7.58-7.45 (m, 5H), 4.52 (d, J = 12.6 Hz, 1H), 4.37 (d, J

= 12.7 Hz, 1H), 4.12 (qd, J = 11.8, 5.6 Hz, 2H), 3.73 (t, J = 3.5 Hz, 1H), 2.89 (s, 3H).

REF: TRW-III-289.

CO2H

NMeBnHO

5.14

281

Figure 5.87a. 1H NMR spectrum of compound 5.14.

3.33.3

1.01.0

2.22.2

1.11.1

1.11.1

5.05.0

ppm0 01122334455667788-14.99

2.89

3.72

3.73

3.74

4.05

4.07

4.09

4.12

4.13

4.15

4.18

4.19

4.35

4.39

4.50

4.54

7.45

7.46

7.47

7.55

7.56

7.57

7.58

282

N-Me-L-Ser-OH (5.15). 5.14 (1.47 g, 7.0 mmol) was dissolved in MeOH (70 mL) and to

it added Pd(OH)2 (700 mg). The resulting suspension was treated with H2 (55 psi) for 40

h, then filtered through celite and concentrated to afford the title compound (660 mg,

79% yield), carried forward as crude.

REF: TRW-III-291.

N-Me-L-Ser(OTBS)-OH (5.16). To a solution of 5.15 (660 mg, 5.55 mmol) in DMF (12

mL) was added TBSCl (923 mg, 6.11 mmol) and imidazole (755 mg, 11.1 mmol). The

resulting solution was allowed to stir at r.t. O/N, then concentrated. To the crude residue

was added H2O and hexanes (1:1) and the resulting suspension stirred 4 h. The solid was

filtered, rinsed with hexanes, and dried to give 5.16 (685 mg, 53% yield).

1H-NMR (300 MHz; CD3OD): δ 4.09 (d, J = 4.1 Hz, 2H), 3.50 (t, J = 4.1 Hz, 1H), 2.72

(s, 3H), 0.92 (s, 9H), 0.12 (s, 6H).

REF: TRW-III-296.

CO2H

NHMeHO

5.15

CO2H

NHMeTBSO

5.16

283

Figure 5.88a. 1H NMR spectrum of compound 5.16.

5.35.3

9.09.0

3.03.0

1.01.0

2.02.0

ppm0 011223344-14.99

0.12

0.92

2.72

3.49

3.50

3.52

4.08

4.10

284

N-Me,Fmoc-L-Ser(OTBS)-OH (5.17). To a suspension of 5.16 (375 mg, 1.61 mmol) in

Na2CO3 (aq, 10%, 3.2 mL) and dioxane (1.3 mL) at 0 °C was added dropwise FmocOSu

(570 mg, 1.69 mmol) as a solution in dioxane (2.1 mL). The resulting solution was

allowed to warm to r.t. as it stirred O/N. After concentration, the crude mixture was

diluted with water, acidified to pH 3-4 with 10% KHSO4, and the product extracted in

EtOAc. The combined organic layer was washed with brine, dried over Na2SO4, and

concentrated to afford 5.17 (663 mg, 90% yield).

1H-NMR (300 MHz; CDCl3): δ 7.77 (d, J = 7.6 Hz, 2H), 7.61 (d, J = 7.2 Hz, 2H), 7.36

(dt, J = 27.2, 7.4 Hz, 4H), 4.82 (t, J = 5.9 Hz, 1H), 4.49-4.40 (m, 2H), 4.30-4.22 (m, 1H),

4.14-4.05 (m, 2H), 3.88 (t, J = 5.1 Hz, 1H), 3.04 (d, J = 24.1 Hz, 3H), 0.86 (s, 9H), 0.08-

0.03 (m, 6H); HRMS (ESI-APCI+): [M+H]+ 456.2206 calcd for C25H34NO5Si, found

456.2200.

REF: TRW-III-298.

CO2H

NMeFmocTBSO

5.17

285

Figure 5.89a. 1H NMR spectrum of compound 5.17.

5.55.5

9.09.0

3.03.0

0.70.7

1.71.7

1.01.0

2.02.0

0.70.7

4.34.3

2.12.1

2.22.2

ppm0 0112233445566778-14.99

0.03

0.07

0.08

0.86

2.99

3.07

3.86

3.88

3.89

4.05

4.07

4.08

4.10

4.11

4.14

4.14

4.22

4.28

4.30

4.40

4.43

4.45

4.48

4.49

4.80

4.82

4.84

7.26

7.26

7.29

7.31

7.34

7.38

7.40

7.43

7.60

7.62

7.76

7.78

286

N-Me,Bn-L-Trp-OMe (5.18). To a solution of L-Trp-OMe·HCl (500 mg, 2 mmol) in

MeOH (20 mL) and Et3N (1.67 mL) was added PhCHO (212 µL, 2.1 mmol). The

resulting mixture was stirred at r.t. for 1 h, then NaBH3CN (132 mg, 2.1 mmol) added.

The reaction mixture was stirred overnight at r.t., at which time paraformaldehyde (180

mg, 2 mmol) was added. After full dissolution (ca. 5 h), PhCHO (132 mg, 2.1 mmol) was

added and the reaction stirred overnight. The crude reaction mixture was concentrated

and the residue dissolved in EtOAc, filtered through celite, and concentrated. Purification

by SiO2 chromatography (5-30% EtOAc in hexanes) afforded diprotected amine 5.18

(300 mg, 47% yield).

1H-NMR (300 MHz; CDCl3): δ 8.11 (s, 1H), 7.55 (d, J = 7.9 Hz, 1H), 7.35 (t, J = 4.3 Hz,

6H), 7.23 (t, J = 6.9 Hz, 1H), 7.14 (t, J = 6.9 Hz, 1H), 7.02 (d, J = 2.2 Hz, 1H), 3.92 (d, J

= 13.5 Hz, 1H), 3.82 (dd, J = 9.1, 5.8 Hz, 1H), 3.72-3.68 (m, 4H), 3.42 (dd, J = 14.4, 9.1

Hz, 1H), 3.17 (ddd, J = 14.3, 5.8, 0.6 Hz, 1H), 2.44 (s, 3H).

REF: TRW-I-278, TRW-I-284.

NH

CO2Me

NMeBn

5.18

287

Figure 5.90a. 1H NMR spectrum of compound 5.18.

3.33.3

1.21.2

1.21.2

4.44.4

1.31.3

1.21.2

1.11.1

1.21.2

1.31.3

6.16.1

1.21.2

1.01.0

ppm1 122334455667788-19.97

2.44

3.14

3.14

3.16

3.16

3.19

3.19

3.21

3.21

3.38

3.41

3.43

3.46

3.68

3.70

3.72

3.79

3.81

3.82

3.84

3.90

3.95

7.01

7.02

7.12

7.14

7.16

7.20

7.23

7.25

7.33

7.35

7.36

7.53

7.56

8.11

288

N-Me-L-Trp-OMe (5.19). To a degassed solution of 5.18 (300 mg, 0.93 mmol) in

MeOH (9 mL) was added Pd(OH)2/C (150 mg). The resulting suspension was treated

with H2 (55 psi) for 18 h, then filtered through celite and concentrated to afford

methylamine 5.19 (215 mg, 99.5% yield), deemed sufficiently pure to carry forward

without further purification.

1H-NMR (300 MHz; CDCl3): δ 8.26 (s, 1H), 7.61 (d, J = 7.7 Hz, 1H), 7.36-7.33 (m, 1H),

7.15 (tdd, J = 14.4, 6.5, 1.2 Hz, 2H), 7.04 (d, J = 2.3 Hz, 1H), 3.66 (s, 3H), 3.57 (t, J =

6.6 Hz, 1H), 3.16 (qd, J = 13.0, 6.6 Hz, 2H), 2.38 (s, 3H), 2.05-1.99 (bs, 1H).

REF: TRW-I-282.

NH

CO2Me

NHMe

5.19

289

Figure 5.91a. 1H NMR spectrum of compound 5.19.

1.41.4

3.423.42

2.492.49

1.141.14

2.982.98

11

2.42.4

1.281.28

1.061.06

0.950.95

ppm1 1223344556677889-14.99

1.99

2.00

2.00

2.01

2.01

2.02

2.02

2.02

2.03

2.03

2.04

2.04

2.05

2.05

2.38

3.09

3.11

3.13

3.16

3.17

3.19

3.22

3.24

3.55

3.57

3.59

3.66

7.04

7.04

7.09

7.10

7.12

7.12

7.14

7.15

7.16

7.17

7.19

7.19

7.21

7.22

7.33

7.33

7.36

7.60

7.62

8.26

290

N-Me,Boc-L-Trp-OMe (5.20). To a solution of 5.19 (6.40 g, 27.6 mmol) in

water/dioxane (1:1, 110 mL) was added NaOH (1.10 g, 27.6 mmol) and Boc anhydride

(7.22 g, 33.1 mmol). The resulting solution was allowed to stir O/N at r.t., then acidified

with HCl to pH 6-7. The product was extracted in CHCl3 and the organic phase dried

over Na2SO4 and concentrated. Crude material was purified by SiO2 chromatography

(25% EtOAc in hexanes) to afford 5.20 (4.50 g, 49% yield).

1H-NMR (300 MHz; CDCl3): δ 8.07-8.06 (m, 1H), 7.61 (d, J = 7.8 Hz, 1H), 7.36 (d, J =

7.5 Hz, 1H), 7.16 (dt, J = 21.1, 7.3 Hz, 2H), 7.06-7.00 (broad, 1H), 5.07-4.73 (m, 1H),

3.76 (s, 3H), 3.48-3.40 (m, 1H), 3.27-3.11 (m, 1H), 2.75 (d, J = 14.1 Hz, 3H), 1.29 (d, J =

61.0 Hz, 9H).

REF: TRW-III-209.

NH

CO2Me

NMeBoc

5.20

291

Figure 5.92a. 1H NMR spectrum of compound 5.20.

11.411.4

3.53.5

1.31.3

1.31.3

3.43.4

1.21.2

1.11.1

2.32.3

1.21.2

1.11.1

1.01.0

ppm1 12233445566778-14.99

1.21

1.41

2.73

2.77

3.11

3.14

3.16

3.18

3.19

3.22

3.22

3.23

3.23

3.23

3.23

3.24

3.24

3.27

3.40

3.42

3.43

3.47

3.48

3.48

3.48

3.76

4.73

4.75

4.77

4.78

5.01

5.02

5.02

5.03

5.03

5.04

5.04

5.04

5.05

5.05

5.06

5.06

5.06

5.07

5.07

7.00

7.00

7.06

7.06

7.10

7.12

7.15

7.17

7.19

7.22

7.34

7.37

7.60

7.62

8.06

8.06

8.06

8.07

8.07

292

L-Ser-OMe (5.21). To a solution of L-serine (50.0 g, 0.48 mol) in MeOH (450 mL) at 0

°C was added SOCl2 (35 mL, 0.48 mol). The reaction was allowed to warm to r.t. as it

stirred O/N. The resulting solution was concentrated, taken up in ether, filtered, and the

solid washed with ether. The crude solid material was recrystallized in MeOH/ether to

afford pure crystalline methyl ester 5.21 as the HCl salt.

REF: TRW-II-118.

CO2MeH2N

OH

5.21

293

N-Bn,Me-L-Ser-OMe (5.22). To a solution of L-Ser-OMe·HCl (10.0 g, 64.1 mmol) in

MeOH (500 mL) and Et3N (50.0 mL, 385 mmol) was added PhCHO (6.8 mL, 67.3

mmol). The resulting mixture was stirred at r.t. for 1 h, then NaBH3CN (4.24 g, 67.3

mmol) added. The reaction mixture was stirred overnight at r.t., at which time

paraformaldehyde (5.78 g, 64.1 mmol) was added. After full dissolution (ca. 5 h),

NaBH3CN (4.24 g, 67.3 mmol) was added and the reaction stirred overnight. The crude

reaction mixture was concentrated and the residue dissolved in EtOAc, filtered through

celite, and concentrated. Purification by SiO2 chromatography (2:1 hexanes:EtOAc)

afforded diprotected amine 5.22 (7.41 g, 52% yield).

1H-NMR (300 MHz; CDCl3): δ 7.37-7.29 (m, 5H), 4.69 (d, J = 5.9 Hz, 1H), 3.79-3.71

(m, 5H), 3.54 (dd, J = 8.8, 6.6 Hz, 1H), 2.78 (dd, J = 8.6, 3.0 Hz, 1H), 2.33 (s, 3H).

REF: TRW-II-019, TRW-II-120, TRW-III-208.

CO2MeBnMeN

OH

5.22

294

Figure 5.93a. 1H NMR spectrum of compound 5.22.

3.03.0

0.80.8

1.01.0

5.65.6

0.70.7

6.46.4

ppm0 0112233445566778-14.99

2.33

2.76

2.77

2.79

2.80

3.52

3.54

3.55

3.57

3.71

3.72

3.73

3.73

3.74

3.75

3.76

3.79

3.84

3.88

4.68

4.70

7.29

7.31

7.33

7.33

7.36

7.37

295

N-Me-L-Ser-OMe (5.23). To a degassed solution of 5.22 (3.7 g, 16.6 mmol) in MeOH

(140 mL) was added Pd(OH)2 (1.85 g). The resulting suspension was treated with H2 (55

psi) for 18 h, then filtered through celite and concentrated to afford methylamine 5.23

(99.5% yield), deemed sufficiently pure to carry forward without further purification.

1H-NMR (300 MHz; CDCl3): δ 3.82-3.77 (m, 4H), 3.63 (dd, J = 10.9, 6.1 Hz, 1H), 3.28

(t, J = 5.2 Hz, 1H), 2.43 (s, 3H).

REF: TRW-II-020, TRW-II-123.

CO2MeMeHN

OH

5.23

296

Figure 5.94a. 1H NMR spectrum of compound 5.23.

3.33.3

1.01.0

1.11.1

4.14.1

ppm0 011223344556677-14.99

2.43

3.26

3.28

3.30

3.60

3.62

3.64

3.66

3.77

3.79

3.81

3.82

297

3-(1H-indol-3-yl)-2-(methylamino)propan-1-ol (5.24). To a suspension of LAH (813

mg, 21.4 mmol) in THF (20 mL) at 0°C was added methyl ester 5.19 (1.70 g, 5.35 mmol)

dropwise. The resulting mixture was heated to reflux and stirred O/N. After cooling to

0°C, 15% NaOH (8 mL) was added and the reaction stirred an additional 1 h at r.t. The

suspension was filtered through celite and the filtrate dried over Na2SO4 before

concentrating to afford 1.09 g (100% yield) of methylamine alcohol 5.24.

1H-NMR (300 MHz; CDCl3): δ 8.76 (s, 1H), 7.62 (d, J = 7.7 Hz, 1H), 7.34 (d, J = 8.0

Hz, 1H), 7.23-7.10 (m, 2H), 6.92 (s, 1H), 3.71 (dd, J = 11.5, 2.1 Hz, 1H), 3.47 (dd, J =

10.9, 4.3 Hz, 1H), 2.98-2.90 (m, 3H), 2.41 (s, 3H).

REF: TRW-I-294, TRW-I-310, TRW-I-323, TRW-I-327.

NH

NHMe

5.24

OH

298

Figure 5.95a. 1H NMR spectrum of compound 5.24.

2.82.8

3.33.3

1.01.0

1.01.0

1.11.1

2.32.3

1.11.1

1.01.0

1.01.0

ppm0 011223344556677889-19.97

2.41

2.86

2.90

2.91

2.93

2.96

2.98

3.45

3.46

3.48

3.50

3.69

3.69

3.72

3.73

6.92

7.10

7.10

7.12

7.13

7.15

7.15

7.18

7.18

7.20

7.21

7.23

7.23

7.33

7.35

7.61

7.63

8.76

299

N-Cbz-L-Ser-OH (5.25). To a solution of L-Ser (10.0 g, 95 mmol) in saturated aqueous

NaHCO3 (380 mL) was added CbzCl (15 mL, 100 mmol). The resulting solution was

stirred O/N at r.t., then acidified to ph 4 with 1M HCl. The product was extracted in

EtOAc (3x), and the combined organic layers dried and concentrated to afford N-Cbz-L-

Ser-OH (21.78 g, 96% yield).

1H-NMR (300 MHz; CD3OD): δ 7.38-7.28 (m, 5H), 5.10 (s, 2H), 4.27 (t, J = 4.3 Hz,

1H), 3.91-3.80 (m, 2H).

REF: TRW-II-337, TRW-II-398.

CO2HCbzHN

OH

5.25

300

Figure 5.96a. 1H NMR spectrum of compound 5.25.

2.22.2

1.01.0

2.12.1

5.05.0

ppm0 0112233445566778-19.97

3.80

3.82

3.84

3.86

3.87

3.90

3.91

4.26

4.27

4.29

5.10

7.28

7.30

7.31

7.33

7.36

7.38

301

N-Cbz-L-Ser(OTBS)-OH (5.26). N-Cbz-L-Ser-OH (10.0 g, 41.8 mmol), TBSCl (6.95 g,

46.0 mmol), and imidazole (5.68 g, 83.6 mmol) were dissolved in DMF (100 mL). The

resulting solution was stirred at r.t. under Ar for 48 h before concentrating. The residue

was suspended in hexanes and the product extracted into 5% aqueous NaHCO3. KHSO4

(1M) was added to acidify the aqueous layer to pH 3, from which the product was

extracted into EtOAc. The combined organic extracts were washed with brine, dried over

Na2SO4, and concentrated to afford N-Cbz-L-Ser(OTBS)-OH (10.04 g, 68% yield).

1H-NMR (300 MHz; CDCl3): δ 7.33-7.30 (m, 5H), 5.08 (d, J = 3.4 Hz, 2H), 4.33-4.31

(m, 1H), 4.05 (dd, J = 10.2, 2.8 Hz, 1H), 3.82 (dd, J = 10.2, 2.8 Hz, 1H), 0.82 (d, J = 2.8

Hz, 9H), -0.02 (s, 6H).

REF: TRW-II-198, TRW-II-338, TRW-II-399.

CO2HCbzHN

OTBS

5.26

302

Figure 5.97a. 1H NMR spectrum of compound 5.26.

6.06.0

8.98.9

1.31.3

1.11.1

1.11.1

2.42.4

5.15.1

ppm0 011223344556677-19.97

-0.0

2

0.81

0.82

3.80

3.81

3.83

3.84

4.03

4.04

4.06

4.07

4.31

4.32

4.32

4.33

5.07

5.08

7.26

7.30

7.31

7.32

7.33

303

L-Trp-OMe·HCl (5.27). To a solution of L-tryptophan (25 g, 123 mmol) in MeOH (250

mL) at 0 °C was added SOCl2 (8.9 mL, 123 mmol) dropwise. The reaction was allowed

to warm to r.t. as it stirred O/N. Upon completion, the reaction was concentrated, then

MeOH (2x) and ether (3x) separately added and the mixture concentrated to thoroughly

dry the crude product. Pure L-tryptophan-OMe·HCl crystals were recovered following

recrystallization in MeOH/ether (25.08 g, 80% yield).

1H-NMR (300 MHz; CD3OD): δ 10.62 (bs, 1H), 7.53 (d, J = 7.8 Hz, 1H), 7.39 (d, J =

8.1 Hz, 1H), 7.20 (d, J = 2.3 Hz, 1H), 7.17-7.03 (m, 2H), 4.33 (dd, J = 7.4, 5.5 Hz, 1H),

3.79 (s, 3H), 3.42 (dq, J = 24.1, 7.9 Hz, 2H).

REF: TRW-II-391.

NH

CO2Me

NH2

5.27

304

Figure 5.98a. 1H NMR spectrum of compound 5.27.

2.02.0

3.03.0

1.01.0

2.12.1

0.90.9

1.01.0

1.01.0

0.80.8

ppm0 0224466881010-19.97

3.30

3.34

3.36

3.39

3.42

3.44

3.47

3.49

3.79

4.30

4.32

4.33

4.35

4.91

7.03

7.04

7.06

7.06

7.08

7.09

7.11

7.12

7.14

7.14

7.16

7.17

7.20

7.20

7.38

7.40

7.52

7.55

10.6

210

.62

10.6

210

.63

10.6

3

305

N-Cbz-L-Trp-OMe (5.28). To a suspension of L-tryptophan-OMe·HCl (15 g, 58.8

mmol) in dioxane (75 mL) at 0 °C was added 1M NaOH (60 mL). CbzCl (9.7 mL, 64.7

mmol) was added, followed by a second portion of 1M NaOH (60 mL). The reaction

mixture was allowed to stir for 20 minutes at 0 °C, then diluted with EtOAc. The organic

layer was separated, washed with 1M HCl and brine, then dried and concentrated to

afford the title compound.

1H-NMR (300 MHz; CDCl3): δ 8.22 (bs, 1H), 7.53 (d, J = 7.9 Hz, 1H), 7.34 (bs, 6H),

7.26-7.17 (m, 2H), 7.13-7.08 (m, 1H), 6.93 (s, 1H), 5.39 (d, J = 8.1 Hz, 1H), 5.11 (d, J =

4.4 Hz, 2H), 4.77-4.70 (m, 1H), 3.68 (s, 3H), 3.32 (d, J = 5.3 Hz, 2H).

REF: TRW-II-448.

NH

CO2Me

NHCbz

5.28

306

Figure 5.99a. 1H NMR spectrum of compound 5.28.

2.12.1

3.13.1

1.01.0

2.22.2

1.21.2

1.01.0

1.31.3

2.12.1

6.26.2

1.11.1

1.01.0

ppm0 0112233445566778899-19.973.

313.

33

3.68

4.70

4.72

4.73

4.74

4.74

4.74

4.75

4.76

4.77

5.10

5.12

5.37

5.40

6.93

7.08

7.08

7.10

7.13

7.13

7.17

7.19

7.19

7.21

7.22

7.26

7.34

7.52

7.55

8.22

8.22

8.22

307

N’-SO2Ph-N-Cbz-L-Trp-OMe (5.29). Phenylsulfonyl chloride (4.4 mL, 34.1 mmol) was

added dropwise to a suspension of N-Cbz-L-Trp-OMe (10 g, 28.4 mmol), NaOH (3.4 g,

85.2 mmol), and Bu4NHSO4 (483 mg, 1.42 mmol) in CH2Cl2 (140 mL). The reaction was

allowed to stir O/N at r.t., then to it added concentrated NH4Cl and EtOAc. The organic

layer was removed and the product further extracted into EtOAc (2x), then the combined

organic extracts dried and concentrated to afford the title compound (13.31 g, 95% yield).

1H-NMR (300 MHz; CDCl3): δ 8.05 (d, J = 8.0 Hz, 1H), 7.97 (d, J = 8.3 Hz, 1H), 7.81

(d, J = 7.8 Hz, 2H), 7.64 (d, J = 8.0 Hz, 1H), 7.37 (sextet, J = 7.7 Hz, 9H), 7.20 (d, J =

7.4 Hz, 1H), 5.31 (d, J = 7.9 Hz, 1H), 5.16-5.06 (m, 2H), 4.70 (q, J = 6.6 Hz, 1H), 3.62

(s, 3H), 3.22 (t, J = 5.6 Hz, 2H).

REF: TRW-II-452.

NSO2Ph

CO2Me

NHCbz

5.29

308

Figure 5.100a. 1H NMR spectrum of compound 5.29.

2.02.0

2.82.8

0.90.9

2.12.1

0.80.8

0.80.8

9.39.3

0.90.9

1.91.9

1.11.1

1.01.0

ppm0 0112233445566778899-19.97

3.20

3.22

3.24

3.62

4.67

4.69

4.71

4.73

5.06

5.10

5.12

5.16

5.30

5.32

7.19

7.21

7.31

7.33

7.35

7.38

7.41

7.43

7.47

7.49

7.63

7.65

7.80

7.82

7.95

7.98

8.04

8.07

309

Bromopyrroloindoline (5.30). N’-SO2Ph-N-Cbz-L-Trp-OMe (13.31 g, 27 mmol) was

dissolved in CH2Cl2 (400 mL), and to the resulting solution added NBS (4.81 g, 27

mmol) and PPTS (6.78 g, 27 mmol). The reaction was allowed to stir O/N at r.t. NaHCO3

(10% aq.) and Na2S2O4 (10% aq.) were added in a 1:1 mixture, the organic layer

removed, dried, and purified (SiO2 chromatography, 1:1 H:E) to afford the title

compound (11.46 g, 74% yield).

1H-NMR (300 MHz; CDCl3): δ 7.63 (d, J = 8.1 Hz, 1H), 7.50-7.45 (m, 3H), 7.36-7.34

(m, 8H), 7.26-7.25 (m, 2H), 7.19 (d, J = 7.4 Hz, 1H), 6.36-6.29 (bs, 1H), 5.42-5.22

(broad, 2H), 3.90-3.84 (m, 1H), 3.69-3.52 (broad, 3H), 3.09 (dd, J = 12.8, 6.0 Hz, 1H),

2.82-2.74 (m, 1H).

REF: TRW-II-453.

NSO2Ph

NCbzCO2Me

Br

H

5.30

310

Figure 5.101a. 1H NMR spectrum of compound 5.30.

1.31.3

1.41.4

2.62.6

1.11.1

2.12.1

1.01.0

1.31.3

2.42.4

7.97.9

3.13.1

1.31.3

ppm0 0112233445566778-19.97

2.74

2.76

2.77

2.78

2.78

2.82

3.06

3.08

3.10

3.12

3.52

3.52

3.52

3.53

3.53

3.53

3.53

3.68

3.69

3.69

3.69

3.69

3.84

3.84

3.84

3.88

3.88

3.89

3.89

3.90

3.90

3.90

5.22

5.22

5.22

5.22

5.22

5.23

5.23

5.37

5.37

5.37

5.38

5.38

5.40

5.41

5.41

5.41

5.41

5.42

6.29

6.29

6.29

6.29

6.31

6.31

6.32

6.36

7.18

7.20

7.25

7.25

7.26

7.34

7.34

7.35

7.35

7.36

7.45

7.45

7.47

7.47

7.47

7.50

7.50

7.61

7.64

311

Methyl 3-bromo-2-(tert-butoxycarbonylamino)propanoate (5.31). To a solution of N-

Boc-Ser-OMe (2.0 g, 9.1 mmol) in THF (50 mL) at 0 °C was added CBr4 (4.54 g, 13.7

mmol) and PPh3 (3.59 g, 13.7 mmol). The reaction was stirred at 0 °C for 10 minutes,

then warmed to r.t. and stirred 4 h under Ar. The crude mixture was filtered through a bed

of celite, diluted with ether, and washed with water and brine. The organic phase was

dried over Na2SO4, filtered, and concentrated. The crude residue was purified by SiO2

chromatography (3:1 hexanes:EtOAC) to afford bromide 5.31 (1.64 g, 64% yield).

1H-NMR (300 MHz; CDCl3): δ 5.42-5.39 (m, 1H), 4.78-4.73 (m, 1H), 3.81 (s, 3H), 3.71

(dd, J = 10.4, 3.4 Hz, 1H), 1.46 (s, 9H).

REF: TRW-I-191.

CO2MeBocHN

Br

5.31

312

Figure 5.102a. 1H NMR spectrum of compound 5.31.

8.88.8

1.01.0

3.03.0

0.80.8

0.70.7

ppm0 011223344556677-19.97

1.46

3.69

3.70

3.72

3.73

3.81

4.73

4.74

4.74

4.75

4.76

4.76

4.76

4.77

4.78

4.78

5.39

5.39

5.39

5.40

5.40

5.40

5.41

5.41

5.41

5.41

5.42

5.42

313

Methyl 2-(benzyloxycarbonylamino)-3-hydroxypropanoate (5.32). To a solution of

Ser-OMe·HCl (5.0 g, 32.1 mmol) in MeOH:NaHCO3 (1:1, 120 mL) was added CbzCl

(5.75 g, 33.7 mmol). The reaction mixture was stirred at r.t. for 48 h, then 1 M HCl

added. The crude mixture was concentrated and the resulting residue dissolved in EtOAc.

The organic phase was washed with 1 M NaOH, dried over Na2SO4, filtered, and

concentrated under reduced pressure to afford Cbz-amine 5.32 (8.12 g, 100% yield) as a

clear oil, carried forward without additional purification.

1H-NMR (300 MHz; CDCl3): δ 7.37-7.34 (m, 5H), 5.72-5.67 (m, 1H), 5.13 (s, 2H), 4.47-

4.43 (m, 1H), 4.01-3.93 (m, 2H), 3.79 (s, 3H).

REF: TRW-I-256.

CO2MeCbzHN

OH

5.32

314

Figure 5.103a. 1H NMR spectrum of compound 5.32.

3.03.0

2.12.1

0.90.9

1.91.9

0.80.8

4.74.7

ppm0 011223344556677-19.97

3.79

3.93

3.93

3.93

3.93

3.93

3.94

3.94

3.94

3.95

3.95

3.95

3.96

3.96

3.96

3.97

3.97

3.97

3.97

3.97

3.97

3.99

3.99

4.01

4.43

4.44

4.44

4.44

4.44

4.45

4.45

4.46

4.46

4.47

4.47

5.13

5.67

5.67

5.68

5.68

5.68

5.68

5.68

5.69

5.69

5.69

5.69

5.69

5.70

5.70

5.70

5.70

5.70

5.71

5.71

5.71

5.72

7.34

7.34

7.35

7.36

7.37

7.37

315

3-benzyl 4-methyl 2,2-dimethyloxazolidine-3,4-dicarboxylate (5.33). To a solution of

Cbz-Ser-OMe (7.56 g, 30.0 mmol) in dry acetone (100 mL) was added 2,2-

dimethoxypropane (40 mL), BF3·OEt2 (3.8 mL, 30.0 mmol), and Na2SO4 (10 g). The

reaction mixture stirred overnight at r.t. under Ar, and then Et3N (12 mL) was added. The

resulting suspension was filtered and the filtrate concentrated under reduced pressure.

The product was extracted with ether (3 x 75 mL), washed with NaHCO3, washed with

brine, dried over Na2SO4, and concentrated. The crude oil was purified by SiO2

chromatography (5-50% EtOAc in hexanes) to afford 5.33 (7.20 g, 82% yield) as a

mixture of rotamers. Crude oil was likely pure enough to carry forward without

purification.

1H-NMR (300 MHz; CDCl3): δ 7.36-7.30 (m, 5H), 5.20-5.02 (m, 2H), 4.52 (ddd, J =

23.2, 6.6, 2.7 Hz, 1H), 4.19-4.07 (m, 2H), 3.69 (d, J = 40.4 Hz, 3H), 1.68 (d, J = 20.1 Hz,

3H), 1.58 (d, J = 9.5 Hz, 3H).

REF: TRW-I-257.

O

CbzN

CO2Me

MeMe

5.33

316

Figure 5.104a. 1H NMR spectrum of compound 5.33.

3.13.1

3.33.3

3.33.3

2.42.4

1.11.1

2.22.2

5.05.0

ppm1 1223344556677-19.97

1.49

1.56

1.59

1.64

1.71

3.64

3.77

4.07

4.08

4.10

4.11

4.12

4.13

4.14

4.14

4.16

4.16

4.17

4.19

4.46

4.47

4.48

4.49

4.54

4.55

4.56

4.57

5.02

5.06

5.15

5.18

5.19

5.20

7.30

7.31

7.32

7.32

7.34

7.35

7.36

317

Benzyl 4-(hydroxymethyl)-2,2-dimethyloxazolidine-3-carboxylate (5.34). To a

solution of ester 5.33 (1.0 g, 3.4 mmol) in THF (12 mL) at -10 °C was added NaBH4 (517

mg, 13.6 mmol). The resulting mixture was stirred for 30 minutes at -10 °C, then MeOH

(5 mL) added dropwise. The reaction was allowed to warm to r.t. while it stirred

overnight under Ar. Water was added and the resulting suspension stirred for 30 minutes,

concentrated under reduced pressure, diluted with brine, and the product extracted with

EtOAc (3 x 15 mL). The combined organic layers were washed with brine, dried over

Na2SO4, and concentrated to afford alcohol 5.34 (900 mg, 100% yield), deemed

sufficiently pure to carry forward without additional purification.

1H-NMR (300 MHz; CDCl3): δ 7.36 (s, 5H), 5.17-5.12 (m, 2H), 4.15-4.13 (m, 1H), 4.04-

3.98 (m, 2H), 3.83-3.59 (m, 3H), 3.39-3.36 (bs, 1H), 1.65-1.47 (m, 6H).

REF: TRW-I-260, TRW-I-263, TRW-I-267.

O

CbzN

MeMe

5.34OH

318

Figure 5.105a. 1H NMR spectrum of compound 5.34.

7.17.1

0.60.6

3.03.0

2.12.1

0.90.9

2.12.1

5.05.0

ppm0 011223344556677-19.97

1.47

1.54

1.62

1.65

3.36

3.36

3.36

3.37

3.37

3.37

3.37

3.38

3.38

3.39

3.39

3.59

3.59

3.59

3.70

3.70

3.70

3.71

3.79

3.79

3.80

3.82

3.82

3.83

3.98

3.98

3.99

4.00

4.01

4.03

4.03

4.03

4.04

4.13

4.14

4.14

4.14

4.14

4.14

4.15

4.15

4.15

5.12

5.12

5.13

5.13

5.17

7.36

319

Benzyl 4-(iodomethyl)-2,2-dimethyloxazolidine-3-carboxylate (5.35). To a solution of

Ph3P (550 mg, 2.1 mmol) and imidazole (143 mg, 2.1 mmol) in CH2Cl2 (10 mL) at 0 °C

was added I2 (533 mg, 2.1 mmol) in 3 portions. The reaction mixture was warmed to r.t.

for 10 minutes, then cooled to 0 °C before adding dropwise a solution of alcohol 5.34

(450 mg, 1.7 mmol) in 5 mL CH2Cl2. The resulting solution was stirred 1 h at 0 °C, then

allowed to warm to r.t. as it stirred an additional 1.5 h. The crude reaction mixture was

filtered through a plug of silica with 1:1 EtOAC/Hexanes. The filtrate was concentrated

under reduced pressure and purified by SiO2 chromatography (5-20% EtOAc in hexanes)

to afford iodide 5.35 (30 mg, 8% yield).

1H-NMR (300 MHz; CDCl3): δ 7.36 (s, 5H), 5.15 (s, 2H), 4.25-4.19 (m, 1H), 4.08-4.02

(m, 2H), 3.54-3.35 (m, 1H), 3.21-3.10 (m, 1H), 1.62 (d, J = 20.1 Hz, 3H), 1.47 (d, J =

19.3 Hz, 3H); 13C-NMR (75 MHz; CDCl3): δ 128.9, 128.0, 67.69, 67.58, 67.0, 59.5, 58.9,

28.1, 27.2, 24.7, 23.2, 6.8, 6.5.

REF: TRW-I-261, TRW-I-266.

O

CbzN

MeMe

5.35I

320

Figure 5.106a. 1H NMR spectrum of compound 5.35.

Figure 5.106b. 13C NMR spectrum of compound 5.35.

3.33.3

3.13.1

1.31.3

1.11.1

2.22.2

1.01.0

2.12.1

5.05.0

ppm0 011223344556677-19.97

1.43

1.49

1.58

1.64

3.10

3.13

3.14

3.16

3.17

3.18

3.19

3.21

3.21

3.21

3.35

3.38

3.38

3.39

3.51

3.51

3.53

3.54

3.54

3.54

4.02

4.04

4.05

4.08

4.19

4.20

4.22

4.23

4.23

4.24

4.25

5.15

7.36

ppm0 05050100100150150200-303.86

6.46

6.77

23.2

424

.67

27.2

128

.08

58.8

959

.46

67.0

367

.58

67.6

9

127.

9712

8.87

321

Benzyl 4-(bromomethyl)-2,2-dimethyloxazolidine-3-carboxylate (5.36). To a solution

of alcohol 5.34 (450 mg, 1.7 mmol) in THF (9 mL) at 0 °C was added CBr4 (862 mg, 2.6

mmol) and PPh3 (681 mg, 2.6 mmol). The reaction was stirred at 0 °C for 10 minutes,

then warmed to r.t. and stirred 4 h under Ar. The crude mixture was concentrated, diluted

with ether, filtered through a bed of celite, and concentrated. The crude residue was

purified by SiO2 chromatography (5-20% EtOAc in hexanes) to afford bromide 5.36 (460

mg, 82% yield).

1H-NMR (300 MHz; CDCl3): δ 7.35 (s, 5H), 5.15 (s, 2H), 4.20-4.15 (m, 1H), 4.12-4.07

(m, 1H), 3.99 (ddd, J = 7.0, 5.1, 2.3 Hz, 1H), 3.68-3.49 (m, 1H), 3.30 (dt, J = 15.3, 10.0

Hz, 1H), 1.60 (d, J = 20.2 Hz, 3H), 1.48 (d, J = 19.8 Hz, 3H).

REF: TRW-I-262, TRW-I-269.

O

CbzN

MeMe

5.36Br

322

Figure 5.107a. 1H NMR spectrum of compound 5.36.

3.23.2

3.33.3

1.11.1

1.11.1

1.11.1

1.11.1

1.11.1

2.02.0

5.05.0

ppm1 1223344556677-19.97

1.44

1.51

1.56

1.63

3.25

3.28

3.30

3.31

3.33

3.36

3.49

3.50

3.50

3.52

3.53

3.53

3.54

3.65

3.65

3.68

3.68

3.97

3.98

3.98

3.99

4.00

4.01

4.02

4.07

4.08

4.10

4.10

4.10

4.11

4.12

4.15

4.16

4.18

4.19

4.21

5.15

7.35

323

3,5,6-trichloroindole (5.37). NCS (1.75 g, 13.2 mmol) was added in one portion to a

solution of 5,6-dichloroindole (2.45 g, 13.2 mmol) in DMF (65 mL) at rt. The reaction

was stirred for 3 h at rt, and brine (50 mL) was added. The mixture was extracted with

EtOAc (3 x 50 mL). The combined organic layers were washed with water (100 mL),

brine (100 mL), dried (Na2SO4), and concentrated under reduced pressure. The residue

was purified by flash chromatography eluting with hexanes/EtOAc (9:1) to give 1.95 g

(67%) of 5.37 as a brown solid.

1H NMR (300 MHz, CDCl3) δ 8.11 (bs, 1 H), 7.71 (s, 1 H), 7.49 (s, 1 H), 7.20 (d, J = 2.6

Hz, 1 H); 13C NMR (100 MHz, CDCl3) δ 133.7, 127.5, 125.3, 125.1, 122.9, 119.7, 113.2,

106.5; IR (neat) 3442, 1448,1266, 1011 cm-1; HRMS (ES/ApCl) calcd for C8H3NCl3 (M-

H) 217.9337, found 217.9347.

NH

Cl

Cl

Cl

5.37

324

Figure 5.108a. 1H NMR spectrum of compound 5.37.

Figure 5.108b. 13C NMR spectrum of compound 5.37.

325

5,6-dichloro-2-(2-methylbut-3-en-2-yl)-1H-indole (5.38). Solid 5.37 (1.95 g, 8.84

mmol) was added in one portion to a solution of freshly prepared prenyl-9-BBN (26.52

mmol) and Et3N (3.12 g, 30.94 mmol, 4.30 mL) in THF (53 mL) at rt. The reaction was

stirred for 3 h, and then quenched with sat. NaHCO3 (50 mL). The organic layer was

separated and the aqueous layer was extracted with Et2O (3 x 50 mL). The combined

organic layers were washed with H2O (100 mL), brine (100 mL), and dried (Na2SO4),

and concentrated under reduced pressure. The residue was purified by flash

chromatography eluting with hexanes/EtOAc (99:1) to give 1.80 g (80%) of 5.38 as a

yellow oil.

1H NMR (300 MHz, CDCl3) δ 7.87 (bs, 1 H), 7.59 (s, 1 H), 7.38 (s, 1 H), 6.23 (m, 1 H),

6.01 (dd, J = 17.3, 10.6 Hz, 1 H), 5.15 (d, J = 10.6 Hz, 1 H), 5.09 (d, J = 17.3 Hz, 1 H),

1.46 (s, 6 H); 13C NMR (75 MHz, CDCl3) δ 148.2, 145.6, 134.9, 128.5, 125.0, 123.7,

121.1, 113.0, 112.0, 97.6, 38.5, 27.5; IR (neat) 3231, 2927, 1453, 1102 cm-1; HRMS

(FAB) calcd for C13H13NCl2 (M+) 253.0425, found 253.0428.

NH

Cl

Cl MeMe

5.38

326

Figure 5.109a. 1H NMR spectrum of compound 5.38.

Figure 5.109b. 13C NMR spectrum of compound 5.38.

327

1-(5,6-dichloro-2-(2-methylbut-3-en-2-yl)-1H-indol-3-yl)-N,Ndimethylmethanamine

(5.39). A solution of Me2NH (0.78 mL, 40% solution in H2O, 6.00 mmol) and a solution

of CH2O (0.47 mL, 37% solution in H2O, 6.00 mmol) were sequentially added to AcOH

(2 mL) and the reaction stirred for 1 h. Neat 5.38 (1.45 g, 5.71 mmol) was added, and the

reaction was stirred for 12 h at rt. 2N NaOH was added until pH ≈ 12. The reaction was

extracted with Et2O (3 x 20 mL). The combined organic layers were washed with H2O

(50 mL), brine (50 mL), and dried (Na2SO4), and concentrated under reduced pressure to

give 1.66 g (90%) of 5.39 as a yellow oil which was used without further purification.

1H NMR (400 MHz, CDCl3) δ 7.86 (bs, 1 H), 7.76 (s, 1 H), 7.32 (s, 1 H), 6.08 (dd, J =

17.6, 10.0 Hz, 1 H), 5.14 (d, J = 10.0 Hz, 1 H), 5.12 (d, J = 17.6 Hz, 1 H), 3.49 (s, 2 H),

2.16 (s, 6 H), 1.52 (s, 6 H); 13C NMR (100 MHz, CDCl3) δ 145.7, 143.4, 132.9, 130.4,

124.9, 123.3, 120.6, 112.7, 111.8, 109.0, 54.1, 45.5, 39.5, 27.2; IR (neat) 3320, 2928,

1463, 1016 cm-1; HRMS (+TOF) calcd for C16H21N2Cl2 (M+H) 311.1082, found

311.1020.

NH

Cl

Cl MeMe

NMe2

5.39

328

Figure 5.110a. 1H NMR spectrum of compound 5.39.

Figure 5.110b. 13C NMR spectrum of compound 5.39.

329

Ethyl 2-amino-3-(5,6-dichloro-2-(2-methylbut-3-en-2-yl)-1H-indol-3-yl)–propanoate

(5.40). PBu3 (346 mg, 1.72 mmol, 0.43 mL) was added to a solution of 5.39 (1.0 g, 3.43

mmol) and N-(Diphenylmethylene)glycine ethyl ester (832 mg, 3.12 mmol) in CH3CN

(30 mL) at rt. The reaction was heated to reflux and was stirred under Ar for 12 h. The

reaction was cooled to rt and concentrated under reduced pressure. The residue was

purified by flash chromatography eluting with hexanes/EtOAc (4:1) to give 1.19 g of a

yellow oil which was dissolved in CH2Cl2 (25 mL). 1N HCl was added and the reaction

was vigorously stirred for 12 h at rt. The organic layer was separated and the aqueous

layer was extracted with CH2Cl2 (2 x 25 mL). The combined organic layers were

concentrated under reduced pressure. The residue was purified by flash chromatography

eluting with MeOH/CH2Cl2 (1:99-5:95) to give 848 mg (67%) of 5.40 as a yellow oil.

1H NMR (400 MHz, CDCl3) δ 7.89 (bs, 1 H), 7.59 (s, 1 H), 7.34 (s, 1 H), 6.08 (dd, J =

17.2, 10.8 Hz, 1 H), 5.16 (d, J = 10.8 Hz, 1 H), 5.15 (d, J = 17.2 Hz, 1 H), 4.09 (comp, 2

H), 3.73 (m, 1 H), 3.20 (dd, J = 14.8, 5.6 Hz, 1 H), 2.96 (dd, J = 14.8, 8.8 Hz, 1 H), 1.53

(s, 6 H), 1.16 (t, J = 7.2 Hz, 3 H); 13C NMR (75 MHz, CDCl3) δ 170.6, 145.6, 142.7,

133.1, 129.9, 125.3, 123.6, 119.9, 112.9, 112.0, 107.3, 61.2, 56.0, 39.5, 31.0, 27.8, 14.3;

IR (neat) 3368, 2973, 2927, 1729, 1463, 1199 cm-1; HRMS (TOF-) calcd for

C18H21N2O2Cl2 (M-H) 367.0986, found 367.0975.

NH

Cl

Cl Me Me

NH2

CO2Et

5.40

330

Figure 5.111a. 1H NMR spectrum of compound 5.40.

Figure 5.111b. 13C NMR spectrum of compound 5.40.

331

2-(tert-butoxycarbonylamino)-3-(5,6-dichloro-2-(2-methylbut-3-en-2-yl)-1Hindol-3-

yl)propanoic acid (5.41). 0.5M NaOH (4.5 mL, 2.25 mmol) and Boc2O (586 mg, 2.69

mmol) were sequentially added to a solution of 5.40 (848 mg, 2.24 mmol) in dioxane (5

mL). The reaction was stirred at rt for 3 h, and then concentrated under reduced pressure

to remove dioxane. 10% KHSO4 was added until pH ≈ 2, and the solution was extracted

with EtOAc (3 x 25 mL). The combined organic layers were dried (Na2SO4) and

concentrated under reduced pressure. The residue was dissolved in 2:1 THF/H2O (18

mL), and LiOH (280 mg, 11.25 mmol) was added in one portion. The reaction stirred at rt

for 12 h. 10% KHSO4 was added until pH ≈ 2, and the solution was extracted with

EtOAc (3 x 25 mL). The combined organic layers were dried (Na2SO4) and concentrated

under reduced pressure to afford 902 mg (91%) of a yellow solid which was used without

further purification.

1H NMR (300 MHz, CDCl3) δ 11.0 (bs, 1 H), 8.19 (m, 1 H), 6.63 (m, 1 H), 7.31 (m, 1 H),

6.80 (bs, 1 H), 6.08 (m, 1 H), 5.18 (comp, 2 H), 4.56 (bs, 1 H), 3.37 (bs, 1 H), 3.14 (m, 1

H), 1.52 (s, 6 H), 1.28-1.02 (comp, 9 H); 13C NMR (75 MHz, CDCl3) δ 177.3, 176.3,

170.4, 156.6, 155.3, 145.4, 142.9, 133.0, 129.9, 129.7, 128.4, 125.0, 124.9, 123.5, 123.3,

119.7, 119.5, 112.9, 112.8, 106.6, 106.0, 81.4, 80.3, 55.4, 54.7, 39.2, 29.8, 28.6, 28.2,

27.8, 27.5; IR (neat) 3335, 2976, 2932, 1697, 1461 cm-1; HRMS (TOF-) calcd for

C21H25N2O4Cl2 (M-H) 439.1197, found 439.1185.

NH

Cl

Cl Me Me

NHBoc

CO2H

5.41

332

Figure 5.112a. 1H NMR spectrum of compound 5.41.

Figure 5.112b. 13C NMR spectrum of compound 5.41.

333

(2S,3R)-ethyl-1-(2-(tert-butoxycarbonylamino)-3-(5,6-dichloro-2-(2-methylbut-3-en-

2-yl)-1H-indol-3-yl)propanoyl)-3-hydroxypyrrolidine-2-carboxylate (5.42). To a

solution of 5.41 (280 mg, 0.63 mmol) in CH3CN (6 mL) was sequentially added HATU

(361 mg, 0.95 mmol), iPr2NEt (327 mg, 2.54 mmol), and cis-3-hydroxy-L-proline ethyl

ester HCl (217 mg, 1.11 mmol). The reaction was stirred at rt for 3 h, and 2N HCl (10

mL) was added. The mixture was extracted with CH2Cl2 (3 x 25 mL), and the combined

organic layers were dried (Na2SO4) and concentrated under reduced pressure. The residue

was purified by flash chromatography eluting with hexanes/EtOAc (1:1) to give 252 mg

(68%) of 5.42 as a colorless oil.

1H NMR (300 MHz, CDCl3) δ 8.73- 8.47 (m, 1 H), 7.58-7.32 (comp, 2 H), 6.11 (m, 1 H),

5.50 (m, 1 H), 5.17 (comp, 2 H), 4.63-3.04 (comp, 9 H), 1.57-1.14 (comp, 18 H); 13C

NMR (75 MHz, CDCl3) δ 172.1, 169.3, 169.1, 169.0, 155.0, 154.8, 154.3, 145.7, 144.8,

144.7, 143.7, 143.3, 142.9, 133.1, 132.7, 129.9, 129.5, 129.2, 124.9, 124.7, 123.1, 119.6,

112.9, 112.6, 112.0, 105.9, 105.6, 79.6, 79.3, 71.7, 70.4, 70.1, 63.8, 63.3, 62.0, 61.4, 60.6,

53.0, 52.4, 45.2, 44.4, 44.3, 39.2, 33.5, 32.6, 30.8, 30.5, 30.2, 28.3, 28.0, 27.7, 21.1, 18.4,

14.2, 14.0; IR (neat) 3359, 2978, 2932, 1694, 1633, 1500, 1455 cm-1; HRMS (TOF-)

calcd for C28H36N3O6Cl2 (M-H) 580.1987, found 580.1976.

NHCl

NHBoc

NO

Me Me

Cl

EtO2C OH

5.42

334

Figure 5.113a. 1H NMR spectrum of compound 5.42.

Figure 5.113b. 13C NMR spectrum of compound 5.42.

335

(8R,8aS)-3-((5,6-dichloro-2-(2-methylbut-3-en-2-yl)-1H-indol-3-yl)methyl)-8-

hydroxyhexahydropyrrolo[1,2-a]pyrazine-1,4-dione (5.43). TFA (0.86 mL, 11.16

mmol) was added to a solution of 5.42 (252 mg, 0.43 mmol) in CH2Cl2 (9 mL). The

reaction was stirred for 3 h, and sat. NaHCO3 (30 mL) was added. The mixture was

extracted with EtOAc (3 x 20 mL), and the combined organic layers were dried (Na2SO4)

and concentrated under reduced pressure. The residue was dissolved in toluene (9 mL)

and 2-hydroxypyridine (9 mg, 0.09 mmol) was added. The reaction was refluxed under

Ar for 12 h, cooled to rt, and concentrated under reduced pressure. The residue was

dissolved in CH2Cl2 (30 mL) and washed with 1N HCl (30 mL). The organic layer was

dried (Na2SO4) and concentrated under reduced pressure to afford 156 mg (85%) of a

beige solid which was used without further purification.

1H NMR (300 MHz, CDCl3) δ 8.14 (s, 1 H), 7.54 (s, 1 H), 7.43 (s, 1 H), 6.10 (dd, J =

17.4, 10.6 Hz, 1 H), 5.72 (bs, 1 H), 5.22 (d, J = 10.6 Hz, 1 H), 5.17 (d, J = 17.4 Hz, 1 H),

4.71-2.10 (comp, 9 H), 1.55 (s, 6 H); 13C NMR (75 MHz, CDCl3) δ 167.7, 165.5, 145.0,

144.1, 133.2, 128.8, 126.1, 124.3, 119.0, 113.6, 112.6, 104.5, 71.0, 64.7, 54.5, 44.2, 39.1,

30.3, 27.8, 26.3; IR (neat) 3352, 2973, 1647, 1459 cm-1; HRMS (TOF+) calcd for

C21H24N3O3Cl2 (M+) 436.1189, found 436.1167.

NHCl

NH

NHO

O

Me Me

Cl

OHH

5.43

336

Figure 5.114a. 1H NMR spectrum of compound 5.43.

Figure 5.114b. 13C NMR spectrum of compound 5.43.

337

3-((5,6-dichloro-2-(2-methylbut-3-en-2-yl)-1H-indol-3-yl)methyl)-2,3,6,7-

tetrahydropyrrolo[1,2-a]pyrazine-1,4-dione (5.44). DEAD (419 mg soln., 40% in

toluene, 0.44 mL, 0.96 mmol) was added to a solution of 5.43 (140 mg, 0.32 mmol) in

CH2Cl2 (10 mL). The reaction was stirred for 5 min, and PBu3 (194 mg, 0.96 mmol) was

added. The reaction was stirred for 3 h, and then concentrated under reduced pressure.

The residue was purified by flash chromatography eluting with hexanes/EtOAc (1:1) to

give 86 mg (64%) of 5.44 as a pale yellow oil.

1H NMR (300 MHz, CDCl3) δ 8.72 (s, 1 H), 7.52 (s, 1 H), 7.33 (s, 1 H), 6.05 (comp, 3

H), 5.09 (d, J = 17.3 Hz, 1 H), 5.09 (d, J = 10.6 Hz, 1 H), 4.42 (m, 1 H), 4.01 (comp, 2

H), 3.57 (dd, J = 14.6, 4.0 Hz, 1 H), 3.15 (dd, J = 14.6, 10.6 Hz, 1 H), 2.73 (comp, 2 H),

1.46 (s, 6 H); 13C NMR (75 MHz, CDCl3) δ 162.4, 156.8, 145.4, 144.3, 133.4, 132.8,

128.7, 125.5, 123.7, 119.4, 119.2, 112.6, 112.4, 104.3, 60.5, 57.2, 45.7, 39.2, 30.7, 27.8;

IR (neat) 3323, 2971, 1681, 1644, 1446 cm-1; HRMS (TOF+) calcd for C21H22N3O2Cl2

(M+H) 418.1084, found 418.1089.

NHCl

NH

NHO

O

Me Me

Cl

5.44

338

Figure 5.115a. 1H NMR spectrum of compound 5.44.

Figure 5.115b. 13C NMR spectrum of compound 5.44.

339

Preparation of Cycloadducts 5.45a and 5.45b. To a solution of 5.44 (86 mg, 0.21

mmol) in MeOH (16 mL) at 0 °C was added 20% aqueous KOH (4 mL). The reaction

was warmed to room temperature (rt) and was stirred for 12 h. The reaction was

quenched with sat. NH4Cl (30 mL) and extracted with CH2Cl2 (3x30 mL). The combined

organic layers were dried (Na2SO4) and concentrated under reduced pressure. The residue

was triturated with CHCl3 (30 mL), and the suspension was filtered to provide 53 mg

(60%) of 5.45a as a white amorphous solid. Concentration of the filtrate gave 25 mg

(29%) of 5.45b as a white amorphous solid.

Data for major isomer 5.45a: 1H NMR (300 MHz, CD3OD) δ 7.55 (s, 1 H), 7.40 (s, 1 H),

3.57 (d, J ) 15.5 Hz, 1 H), 3.47 (m, 1 H), 2.74 (d, J ) 15.5 Hz, 1 H), 2.60 (m, 1 H), 2.20-

1.96 (comp, 6 H), 1.35 (s, 3 H), 1.11 (s, 3 H); 13C NMR (75 MHz, CD3OD) δ 175.7,

171.3, 144.3, 137.2, 128.1, 125.4, 123.3, 119.8, 113.1, 104.8, 68.3, 61.6, 50.8, 45.2, 36.2,

31.7, 30.1, 28.6, 25.4, 24.9, 22.3; IR (neat) 1663, 1428 cm-1; HRMS (TOF+) calcd for

C21H22N3O2Cl2 (M + H) 418.1084, found 418.1084.

Data for minor isomer 5.45b: 1H NMR (300 MHz, CDCl3) δ 9.78 (s, 1 H), 7.49 (s, 1 H),

7.33 (s, 1 H), 3.71 (d, J ) 17.8 Hz, 1 H), 3.47 (comp, 2 H), 3.25 (s, 1 H), 2.78 (d, J ) 17.8

Hz, 1 H), 2.67 (m, 1 H), 2.18- 1.78 (comp, 6 H), 1.24 (s, 3H), 1.16 (s, 3 H); 13C NMR (75

MHz, CD3OD /CDCl3 (1:9)) δ 173.7, 169.6, 142.6, 135.7, 127.1, 125.0, 122.9, 119.1,

112.4, 102.9 67.2, 61.5, 45.8, 44.3, 34.8, 32.6, 29.8, 29.1, 28.4, 24.5, 23.2; IR (neat)

1676, 1453 cm-1; HRMS (TOF+) calcd for C21H22N3O2Cl2 (M + H) 418.1084, found

418.1079.

OH

N N

HNMe Me Cl

H Cl

O

OH

N N

HNMe Me Cl

H Cl

O5.45a 5.45b

340

Figure 5.116a. 1H NMR spectrum of compound 5.45a.

Figure 5.116b. 13C NMR spectrum of compound 5.45a.

341

Figure 5.117a. 1H NMR spectrum of compound 5.45b.

Figure 5.117b. 13C NMR spectrum of compound 5.45b.

342

Synthesis of Malbrancheamide (5.46). DIBAL-H (0.70 mL, 1 M in toluene, 0.70 mmol)

was added to a suspension of 5.45a (15 mg, 0.036 mmol) in toluene (7 mL) at rt. The

reaction was stirred at rt for 12 h, whereupon finely powdered Na2SO4-10H2O was added

until bubbling ceased. The mixture was filtered with a medium porosity fritted funnel

washing with EtOAc (50 mL) and MeOH (50 mL), and the filtrate was concentrated

under reduced pressure. The residue was purified by flash chromatography eluting with

MeOH/ CH2Cl2 (2:98) to give 12 mg (80%) of 5.46 as a white amorphous solid.

1H NMR (400 MHz, CD3OD) δ 7.48 (s, 1 H), 7.40 (s, 1 H), 3.43 (d, J ) 10.3 Hz, 1 H),

3.06 (m, 1 H), 2.85 (comp, 2 H), 2.54 (m, 1 H), 2.27 (dd, J ) 10.2, 1.5 Hz, 1 H), 2.20-1.18

(comp, 6 H), 1.43 (s, 3 H), 1.34 (s, 3 H); 13C NMR (100 MHz, CD3OD) δ 176.6, 145.1,

137.3, 128.2, 125.3, 123.2, 119.6, 113.1, 104.7, 66.1, 59.4, 57.4, 55.4, 48.5, 35.5, 32.4,

30.6, 30.0, 28.1, 24.2, 23.5; IR (neat) 3226, 1658, 1460 cm-1; HRMS (TOF+) calcd for

C21H24N3OCl2 (M + H) 404.1291, found 404.1290.

OH

N N

HNMe Me Cl

H Cl

5.46

343

Figure 5.118a. 1H NMR spectrum of compound 5.46.

Figure 5.118b. 13C NMR spectrum of compound 5.46.

344

Chetomin (1.12). Authentic sample purchased from Santa Cruz Biotechnology.

1H-NMR (400 MHz; CDCl3): δ 7.66 (dd, J = 6.1, 3.0 Hz, 1H), 7.35-7.33 (m, 1H), 7.31-

7.27 (m, 2H), 7.23-7.22 (m, 2H), 7.18 (s, 1H), 6.95 (t, J = 7.4 Hz, 1H), 6.80 (d, J = 8.0

Hz, 1H), 6.21 (s, 1H), 4.43 (s, 1H), 4.39 (d, J = 4.7 Hz, 1H), 4.35 (s, 1H), 4.31 (d, J = 3.5

Hz, 1H), 4.28 (s, 1H), 3.87 (d, J = 15.6 Hz, 1H), 3.71 (d, J = 15.4 Hz, 1H), 3.20 (s, 3H),

3.16 (s, 3H), 3.10 (s, 1H), 2.96 (s, 3H); 13C-NMR (101 MHz; CDCl3): δ 166.8, 165.51,

165.48, 163.2, 148.3, 134.0, 131.4, 130.4, 127.2, 126.5, 125.0, 122.8, 120.60, 120.40,

119.2, 111.4, 111.2, 107.7, 80.0, 76.5, 76.1, 74.7, 73.7, 73.5, 61.3, 60.7, 42.6, 28.2, 27.44,

27.34, 27.1.

FOR NMR FILES, SEE Tim/NMR/robot/trw-III-chetomin.

See also (for characterization data): Fujimoto, H.; Sumino, M.; Okuyama, E.; Ishibashi,

M. Immunomodulatory Constituents from an Ascomycete, Chaetomium Seminudum. J.

Nat. Prod. 2004, 67, 98-102.

chetomin (1.12)

NNMe

O

O

SS

NH

HOH

MeNNMe

O

O

SS

NOH

345

Figure 5.119a. 1H NMR spectrum of compound 1.12.

Figure 5.119b. 13C NMR spectrum of compound 1.12.

2.92.9

0.90.9

3.03.0

3.23.2

1.41.4

1.41.4

0.40.4

1.11.1

1.61.6

1.21.2

1.01.0

0.60.6

1.01.0

1.31.3

1.31.3

1.11.1

1.81.8

2.12.1

1.11.1

1.01.0

ppm0 011223344556677-16.042.

963.

103.

163.

20

3.69

3.73

3.85

3.89

4.25

4.28

4.31

4.32

4.35

4.38

4.39

4.43

6.21

6.79

6.81

6.93

6.95

6.97

7.18

7.22

7.23

7.23

7.23

7.24

7.27

7.28

7.28

7.29

7.29

7.29

7.30

7.31

7.31

7.33

7.33

7.35

7.65

7.66

7.67

7.67

ppm0 05050100100150150-253.79

27.0

927

.34

27.4

428

.22

42.6

2

60.6

861

.25

73.5

173

.74

74.7

476

.05

76.5

180

.03

107.

6611

1.17

111.

3811

9.16

120.

4012

0.60

122.

8212

5.05

126.

5012

7.25

130.

4013

1.43

134.

00

148.

31

163.

2516

5.48

165.

5116

6.83

117

4Biomimetic Synthesis of Alkaloids Derivedfrom Tryptophan: Dioxopiperazine AlkaloidsTimothy R. Welch and Robert M. Williams

4.1Introduction

Countless secondary metabolic indole alkaloids produced in both marine and ter-restrial fungi are derived from tryptophan. Our crude attempts to synthesize someof the vast array of structurally diverse natural alkaloids only serves to showcase theefficiency and elegance with which Nature is able assemble the same molecules.Still, we strive to mimic and in turn better understand the mechanisms inside thecell that are able to produce molecular architecture of such synthetic complexity.

Moreover, it has often been found advantageous to exploit Nature’s evolutionarycreative design of alkaloids in search of new compounds of therapeutic potential.The rich subclass of biologically active, tryptophan-derived dioxopiperazines foundin nature has inspired medicinal chemists to use the dioxopiperazine core indrug design efforts to mimic the interactions of natural peptides while reducingsusceptibility to metabolic amide bond cleavage. Furthermore, a dioxopiperazineshould pay a small entropic penalty upon binding to a target in comparison to ananalogous peptide, as a direct consequence of the reduced conformational mobilityinherent in the dioxopiperazine ring. In this chapter, we present a brief review of aselect group of (partly) biomimetic syntheses of tryptophan-derived dioxopiperazinealkaloids (Figure 4.1). In most of these syntheses, a single step or key transformationhas been deemed to constitute the ‘‘biomimetic’’ aspect of that particular work. Asthe actual biosynthetic pathways to most, if not all, of the alkaloid natural productscovered here are either unknown or known only in part we have attempted, whereappropriate, to point out the particular biomimetic step or transformation.

4.2Prenylated Indole Alkaloids

Birch, Wright, and Russell first isolated brevianamide A from Penicillium brevi-compactum in 1969 [1–3]. Several years later, Birch and coworkers determinedthat brevianamide A was biosynthetically derived from tryptophan, proline, and

Biomimetic Organic Synthesis, First Edition. Edited by Erwan Poupon and Bastien Nay.! 2011 Wiley-VCH Verlag GmbH & Co. KGaA. Published 2011 by Wiley-VCH Verlag GmbH & Co. KGaA.

346

APPENDIX I Publications

118 4 Biomimetic Synthesis of Alkaloids Derived from Tryptophan: Dioxopiperazine Alkaloids

NH

NH

O N

OH H

brevianamide F

NN

O

OMe

Me

HN

O

spirotryprostatin B

N

Me MeNH

NOOH

H

NMe Me

HOH

okaramine N

NN

NH

HN

O

O

Me

Me

Me

Me

HO

OH

H

H

gypsetin

O

H

NN

HNMe Me Cl

H Cl

malbrancheamide (+)-versicolamide B

O

NHO

NN

OMe

Me

MeMe

O

H

H

NHN N

OO

Me

NH

O

ent-alantrypinone

NN

O

O MeSSHO H

OH(+)-gliotoxin

Section 4.2.1

Section 4.2.2

Section 4.2.3 Section 4.3 Section 4.3.1

Figure 4.1 Representative molecules discussed in this chapter.

mevalonic acid through feeding experiments (Scheme 4.1) [4]. Furthermore, Birchshowed that radiolabeled brevianamide F was incorporated into 5. It was postu-lated at the time and later supported with experimental evidence by Williams andcoworkers that deoxybrevianamide E was also a biosynthetic precursor [5].

NH

CO2HH3HN

H

14CH2

NH2

CO2H

14CH2

O

OHMe

O

[2-14C]-D,L-tryptophan(1)

[5-3H]-L-proline(2)

[2-14C]-D,L-mevalonicacid lactone (3)

NH

14CH2NH

O N

OH H

3H

brevianamide F (4)

O

H

NNH

O

(+)-brevianamide A (5)

NH

OMe(14C)

Me

NH

NH

O N

OH H

MeMe

3H3H

deoxybrevianamide E (6)

Scheme 4.1 Proposed biosynthesis of the brevianamides.

Since these early studies, Williams has developed a proposal for the biosyn-thesis of the brevianamides, with that of brevianamide E shown in Scheme 4.2.Derived from tryptophan and proline, 6 was proposed to undergo oxidation tohydroxyindolenine 7. Irreversible nucleophilic ring closure was supposed to leadto brevianamide E (8), supported by incorporation of [8-3H2]6 into 8 in significantradiochemical yield [5].

347

4.2 Prenylated Indole Alkaloids 119

NH

NH

O N

OH H

MeMe

3H3H

deoxybrevianamide E (6)

[ox]

N

NH

O N

OH H

MeMe

HOnucleophilic

additionN

N

NH O

O

Me

Me

H

H

brevianamide E (8)

HO

7

Scheme 4.2 Proposed biosynthesis of brevianamide E.

4.2.1Dioxopiperazines Derived from Tryptophan and Proline

Brevianamide F (12) was first isolated in 1972 and is one of the simplesttryptophan-derived dioxopiperazine natural products. It is readily synthesizedthrough amino acid coupling of N-Boc-tryptophan (9) to proline ethyl ester (10),Boc-deprotection, and ring closure, in modest overall yield (Scheme 4.3) (R.M.Williams, unpublished results).

NH

CO2HH

NH

EtO2CHATU, iPr2NEt

MeCN+

NH

NHBoc

O N 1. TFA, CH2Cl2

2. 2-OH-pyridinePhCH3

EtO2C

NH

NH

O N

OH H

brevianamide F (12)

NHBoc

9 10

11

62%

1:1 = cis:trans

56%

Scheme 4.3 Biomimetic synthesis of brevianamide F.

Brevianamide F lacks only the reverse prenyl group found in deoxybrevianamideE, which has been synthesized by Kametani and coworkers en route tobrevianamide E (Scheme 4.4) [6]. N-Benzyloxycarbonyl-l-proline (13) wassubjected to Schotten–Baumann conditions with dimethyl aminomalonateto give amide 14. Debenzyloxycarbonylation of 14 followed by heating withcatalytic 2-hydroxypyridine effected cyclization to dioxopiperazine 15 in 93%yield. Condensation with indole 16 gave a separable mixture of diastereomers,individually hydrolyzed to the corresponding free acids (17). Heating of the desireddiastereomer in dioxane gave deoxybrevianamide E (18) and its epimer (19) in 29and 55% yield, respectively. Irradiation of methanolic 18, containing Rose Bengalin the presence of oxygen, followed by addition of dimethyl sulfide, resulted in thebiomimetic hydroxylation of deoxybrevianamide E, furnishing brevianamide E (8)and 20 as a separable mixture of diastereomers.

Nineteen years later, a more efficient synthesis of brevianamide E was com-pleted by Danishefsky and coworkers [7]. The synthesis commenced with C3chlorination of the known phthaloylated tryptophan derivative 21, followed byaddition of fresh prenyl-9-borabicyclo[3.3.1]nonane (prenyl-9-BBN) to the resultant3-chloroindolenine (Scheme 4.5). Hydrazinolysis in ethanol provided amino ester

348

120 4 Biomimetic Synthesis of Alkaloids Derived from Tryptophan: Dioxopiperazine Alkaloids

CO2BzNHO2C

H2NCO2Me

CO2Me 1. Pd/C, H2

2. 70 °C, 2-OH-pyridine (cat)

HNN

O

O

CO2MeH

H

HN

NO

CO2MeCO2Me

H

H

CO2Bz

NH

NMe2

MeMe

+

1. NaH, "HNHNMe

Me

N

O

OH

HO2C

2. NaOH HNHNMe

Me

N

O

OH

HNHNMe

Me

N

O

OH

+

H Hdioxane, "

deoxybrevianamide E (18)

HNHNMe

Me

N

O

OH

HRose Bengal

deoxybrevianamide E (18)

hn, O2; Me2SN

N

NH O

O

Me

Me

HOH

H

brevianamide E (8)

13 14 15 16

17 19

NN

NH O

O

Me

Me

HOH

H

bis(epi)brevianamide E (20)

+

69%

93%

22%

84%

18:19 = 1:1.9

63%8:20 = 2:1

Scheme 4.4 Kametani’s total synthesis of deoxybrevianamide E and brevianamide E.

NH2HN

H

MeMe

1. Boc-proline,BOPCl

52%

2. TFA; NH3, MeOHHNHN

H

MeMe

N

O

OH

CH2Cl2, acetone

76%

NN

NH O

O

Me

Me

HOH

H

NN

NH O

O

Me

Me

HOH

H

brevianamide E (8)

bis(epi)brevianamide E (20)

deoxybrevianamide E (18)

NPhth

CO2Me

CO2Me

CO2Me

NH

H1. tBuOCl, Et3N

THF, #78 °C

NPhthHN

H

MeMe

95%

N2H4, EtOH, rt

65%

21 22

23

Me

Me

9-BBN2.

DMDO

Scheme 4.5 Danishefsky’s total synthesis of brevianamide E.

23 in 65% yield, which was coupled to N-Boc-l-proline, deprotected, and cyclizedto afford deoxybrevianamide E (18) in 52% yield. Compound 18 was elaboratedto brevianamide E (8) and bis(epi)brevianamide E (20) in a ratio of !1 : 5 upontreatment with dimethyldioxirane (DMDO) in a biomimetic oxidative cyclizationsequence reminiscent of the original Kametani work discussed above.

The structural similarities between deoxybrevianamide E and the then newlyisolated natural products tryprostatin A and B did not escape notice of Danishefsky

349

4.2 Prenylated Indole Alkaloids 121

and coworkers. While prenylation failed in attempts to use a reverse prenylboranenucleophile directly as in the method used to synthesize 22 above, a solutionwas found in treating chloroindolenine 24 with tri(n-butyl)prenylstannane andBCl3 to afford the desired prenyl functionality at C2 in excellent yield (Scheme 4.6).Phthalimide deprotection, peptide coupling, Boc-deprotection, and cyclization wereachieved to afford tryprostatin B (30) in 43% overall yield [7].

NPhth

CO2Me

CO2Me

HN

H tBuOCl,CCl4

NPhth

CO2MeCO2Me CO2Me

N

HCl

n-Bu3Sn

Me

Me+ BCl3

MeMe

BCl2

NPhthHN

H

MeMe

N2H4 H2O

82%

NH2HN

H

MeMe

N-Boc-L-Pro-FHNHN

H

MeMe

ONBoc

1. TMSI, MeCN

67%94%

HNHN

H

MeMe

N

O

O

H

tryprostatin B (30)

CH2Cl2

21 24

25 26

27 28

29

Et3N

MeOH, CH2Cl2

CH2Cl2, NaHCO3 2. NH3, MeOH

83%

Scheme 4.6 Biomimetic total synthesis of tryprostatin B.

Danishefsky and coworkers also completed the total synthesis of the spirooxin-dole spirotryprostatin B [8]. l-Tryptophan methyl ester was converted into theoxindole derivative 32, followed by addition of prenyl aldehyde under basicconditions to afford an inseparable four-component mixture of spirooxindoles(33–36, Scheme 4.7). Peptide coupling and subsequent treatment of the mix-ture with lithium bis(trimethylsilyl)amide (LHMDS) followed by selenylationpresumably gave phenyl selenide mixture 38. Oxidative elimination produceda mixture from which 39 was separated and elaborated to spirotryprostatin B (40)via Boc-deprotection and base-induced cyclization.

Danishefsky took a markedly different approach in the synthesis of spirotrypro-statin A [9]. A potentially biomimetic Pictet–Spengler reaction of tryptophanderivative 42 with thioaldehyde 41 as a masked isoprene equivalent gave thedesired cis-tetrahydrocarboline (43) with marginal selectivity (Scheme 4.8).N-Bromosuccinimide (NBS)-mediated oxidative rearrangement proceeded viaintermediate 44 to the oxindole was followed by deprotection of the carbamate togive amine 45. The modest yield of the sequence (57%) reflects the susceptibility ofthe oxindole to electrophilic aromatic bromination under the spiro-rearrangementconditions. Peptide coupling and Troc-deprotection resulted in cyclization to thedioxopiperazine, after which oxidation and sulfoxide elimination revealed theprenyl group to afford selectively spirotryprostatin A (48).

The synthesis of notoamide J was completed by Williams and coworkers, startingwith the Boc-protection of 7-hydroxyindole (Scheme 4.9) [10]. Chlorination at C3was followed by reverse prenylation of the resultant 3-chloroindolenine 51 to afford

350

122 4 Biomimetic Synthesis of Alkaloids Derived from Tryptophan: Dioxopiperazine Alkaloids

NH

CO2Me CO2Me

CO2Me

CO2MeCO2Me

CO2Me

CO2MeCO2Me CO2Me

NH2•HCl NH2•HCl

H

DMSO, 12N HCl

NH

H

O

Me

Me

CHO

Et3N, py.

NHHN

O

Me

Me

NHHN

O

Me

Me

H H

NHHN

O

Me

Me

H

NHHN

O

Me

Me

H

NBoc

CO2HH

BOPCl

73% (2 steps)2:3:3:3

NN

O

OMe

Me

HN

O

NHN

O

Me

H

OMe

NBoc

LHMDS NHN

O

Me

PhSe

OMe

NBoc

DMDO

NHN

O

Me

OMe

NBoc

1. TFA, CH2Cl2

2. Et3N

7% (5 steps)

31 32

33 34 35 36

37 38

39 spirotryprostatin B (40)

AcOH, PhOH

+ + +

PhSeCl

Scheme 4.7 Danishefsky’s synthesis of spirotryprostatin B.

52. The corresponding gramine was prepared by treating 52 with formaldehyde anddimethylamine, and subsequent Somei–Kametani coupling and imine hydrolysisgave tryptophan derivative 54 in good yield. Protection of the free amine as theBoc-carbamate and ester hydrolysis gave 55, which was coupled to proline ethylester in the presence of O-(7-azabenzotriazol-1-yl)-N,N,N",N"-tetramethyluroniumhexafluorophosphate (HATU) to afford amide 56. Cyclization to dioxopiperazine57 was followed by a biomimetic oxidation sequence accompanied by pinacol-typerearrangement to the oxindoles notoamide J (58) and 3-epi-notoamide J (59) in a2 : 1 separable mixture.

4.2.2Dioxopiperazine Derived from Tryptophan and Amino Acids other than Proline

Corey and coworkers designed a succinct synthesis of okaramine N, featuringa Pd-promoted dihydroindoloazocine formation [11]. Readily available trypto-phan derivative 60 was reduced to indoline 61 and subsequently prenylatedvia copper(I)-catalyzed alkylation with butyne 62 (Scheme 4.10). Treatment with

351

4.2 Prenylated Indole Alkaloids 123

PhS

H

Me

Me

O N H

CO

2Me

CO

2Me

CO

2Me

NH

2

MeO

H

1. T

FA

, 4Å

CH

2Cl 2

2. (

Boc

) 2O

MeC

N, E

t 3N

+N H

NB

oc

MeO

Me

Me

SP

h

H

74%

57%

(2

step

s)

NH

HNO

SP

h

Me

Me

CO

2Me

MeO

H

H

Tro

cN

ClO

CH

+1.

CH

2Cl 2

, Et 3

N

2. Z

no , TH

F, M

eOH

3.N

aIO

4, M

eOH

H2O

4. P

hCH

3,"

57%

(4

step

s)

NH

N

O

Me

MeO

H H

N

O OH

RhC

l 3. 3

H2O

EtO

H,"

45%

NH

N

O Me

Me

MeO

H H

N

O OH

spiro

tryp

rost

atin

A (

48)

N H

NB

oc

MeO

Me

Me

SP

h

HB

r

HO

H

NB

S, T

HF

AcO

H, H

2O

TF

A, C

H2C

l 2

2:1,

cis:

trans

41

4243

44

45

46

47

Sche

me

4.8

Dan

ishe

fsky

’ssy

nthe

sis

ofsp

irotr

ypro

stat

inA

.

352

124 4 Biomimetic Synthesis of Alkaloids Derived from Tryptophan: Dioxopiperazine Alkaloids

N HH

O

Boc

2O, D

MA

P

MeC

NN H

Boc

O

NC

S

DM

FN H

Boc

O

Cl

Et 3

N, T

HF

N HB

ocO

Me M

e

HN

Me 2

, CH

2O

AcO

HN H

Me M

e

NM

e 2NC

O2E

t Ph Ph

1.

PB

u 3, M

eCN

,2.

1 M

HC

l, T

HF

N H

Me

Me

NH

2

CO

2Et

Boc

OB

ocO

1. B

oc2O

, 1M

NaO

Hdi

oxan

e

2. L

iOH

, TH

F/H

2O

N H

Me

Me

NH

Boc

CO

2H

Boc

O

Me

Me

9-B

BN

4950

5152

5354

55

N H

Me

Me

NH

Boc

ON

Boc

O

1. T

FA

, CH

2Cl 2

2. 2

-OH

-pyr

idin

eP

hCH

3N H

EtO

2C

EtO

2C

56

1. L

iOH

, TH

F/H

2O

2.

N H

Me

Me

N H

ON

OH

H

HO

57

N H

N H

ON

O

H

O

MeMe

HO

SNO

nB

u OO

H

noto

amid

e J

(58)

N H

N H

ON

O

H

O

MeMe

HO

H

59

+

70%

83%

48%

84%

76%

83%

59%

53%

cis:

trans

= 1

:1

CH

2Cl 2

47%

58:5

9 =

2:1

HA

TU

,iP

r 2N

Et

MeC

N

Sche

me

4.9

Will

iam

s’bi

omim

etic

synt

hesi

sof

noto

amid

eJ.

353

4.2 Prenylated Indole Alkaloids 125

CO2Me

CO2Me

CO2Me

NHBocNH

NaBH3CN,AcOH

60% NHBocNH

1. CuCl, iPr2NEt

87%

2. DDQ; H2,quinoline, Pd/C

MeOAcMe

NHBocN81%Me

Me

1. SOCl2

2. LiOH; Fmoc-Cl

CO2H

NHFmocNMe

Me64

60 61

63

62

Scheme 4.10 Synthesis of the N-reverse-prenylated tryptophan derivative.

2,3-dichloro-5,6-dicyano-1,4-benzoquinone (DDQ) effected the dehydrogenation ofthe indoline, and the resulting tryptophan derivative 63 was deprotected, saponified,and reprotected as the N-Fmoc derivative (64).

The synthesis of okaramine N was completed through reductive amination of3-methyl-buten-2-al onto l-tryptophan methyl ester, followed by coupling of theresultant product (65) with acid 64 to form the desired tetracycle (Scheme 4.11).Treatment of 66 with Pd(OAc)2 provided the eight-membered ring (67) in modestyield (38%). The free amine obtained upon Fmoc cleavage underwent cyclization toform dioxopiperazine 68 in 95% yield. A potentially biomimetic oxidative cyclizationwas effected by treating 68 with N-methyltriazolinedione (MTAD), which reactedselectively with the N-unsubstituted indole subunit, and after photooxidation and

CO2Me

NH2NH

CO2Me

HNNH Me

Me

MeMe

CHODCM;

NaBH4, MeOH 70%

BOPCl,64

NH

NMeO2C

MeMe

O

FmocHN

N

MeMe

38%

Pd(OAc)2, O2

N O

FmocHN

N

MeMe

MeO2C

Me MeNH

95%

Et2NH

N

N

MeMe

Me MeNH

HNO

OH

H

70%

MTAD; O2,sunlamp,

methylene blue;

N

Me MeNH

NOO

HHMe2S; "

NMe Me

HOH

okaramine N (69)

31 65

66

67 68

Scheme 4.11 Completion of the total synthesis of okaramine N.

354

126 4 Biomimetic Synthesis of Alkaloids Derived from Tryptophan: Dioxopiperazine Alkaloids

CO2Me

NH2NH

NaOH, Boc2O,

(n-Bu)4NHSO4

91%CO2Me

NHBocBocN

N-PSP,PPTS

93% BocN NBoc

CO2Me

SePh

BocN NBoc

CO2Me

SePh

H

+

71 (major) exo 72 (minor) endo

60%

MeOTf,2,6-di(t-

butyl)pyridine

Sn(n-Bu)3 BocN NBoc

CO2R

Me Me

18:1 (a :b)

83%

TMSI

NH NH

CO2Me

Me Me

NaOHTHF/MeOH/H2O

73 (a) R=Me

74 (a) R=H98%

1. 74, BOPCl,Et3N NH N

Me Me

2. TMSI34%

N NH

O

O H

H

MeMeamauromine (76)

31 70

75

Scheme 4.12 Danishefsky’s total synthesis of amauromine.

then reduction formed a hydroxylated octacycle that was directly converted intookaramine N (69) via thermolysis in 70% yield.

Amauromine was synthesized as shown in Scheme 4.12, starting with thebis(Boc) protection of l-tryptophan [12]. Conversion into the selenide by re-action with N-phenylselenophthalimide (N-PSP) in the presence of pyridiniump-toluenesulfonate (PPTS) afforded a mixture of 71 and 72 (9 : 1). Photolysis ofthe mixture in the presence of prenyltri(n-butyl)tin produced a mixture of reverseprenylated pyrrolodinoindolines, the desired major diastereomer of which wasseparated by crystallization from hexanes. The Boc groups were globally cleavedusing iodotrimethylsilane (TMSI) to afford the ester (75). Coupling of 75 withacid 74 gave the amide, which readily cyclized to the desired dioxopiperazine (76,amauromine) upon treatment with TMSI.

Danishefsky and coworkers completed a total synthesis of gypsetin in the samefashion as their effort on brevianamide E [7]. The reverse prenylated amine, 23,was synthesized as shown above (Scheme 4.5). Boc protection and cleavage of themethyl ester afforded acid 77, which was coupled to amine 23, deprotected, andcyclized to dioxopiperazine 78 (Scheme 4.13). Treatment with DMDO effected thebiomimetic oxidative conversion into the natural product gypsetin (79).

4.2.3Bicyclo[2.2.2]diazaoctanes

In 1970, Sammes proposed a hetero-Diels–Alder cycloaddition to be the biosyn-thetic origin of the bicyclo[2.2.2]diazaoctane core found in brevianamides A and B(Scheme 4.14) [13].

Support for this proposal was observed upon treating dihydroxypyrazine 82 withdimethyl acetylenedicarboxylate (83) or with norbornadiene (84) to give cycloadducts85 or 86, respectively (Scheme 4.15) [13].

355

4.2 Prenylated Indole Alkaloids 127

NPhth

CO2Me

NH

H1. t BuOCl, Et3N

THF, #78 °C

2. Me

Me 9-BBNNPhth

CO2Me

HN

H

MeMe

95%

N2H4, EtOH, rt

65% NH2

CO2Me

HN

H

MeMe

1. (Boc)2O, Et3N THF2. LiOH, MeOH, THF

94%

NHBoc

CO2H

HN

H

MeMe

1. BOPCl, CH2Cl2

2. TFA, CH2Cl23. NH3MeOH

87%

HNHN

H

MeMe

NH

O

O

NHMe

Me

HCH2Cl2,acetone

NN

NH

HN

O

O

Me

Me

Me

Me

HO

OH

H

H

gypsetin (79)40%

(+ 18% all syn-isomer + 20% double anti -isomer)

DMDO

2122

23

77

78

Scheme 4.13 Synthesis of gypsetin.

NN

O

OH

MeMe

O

NNH

O

MeMe

80 81

Scheme 4.14 Proposed hetero-Diels–Alder formation of bicyclo[2.2.2]diazaoctanes.

N

N OH

MeHO

PhH2CNH

HN O

O

MeO2C

CO2Me

Me

CH2Ph

NH

HN

O

OCH2Ph

Me

MeO2C CO2Me

DMF, r.t.

82

85

86

84

83

Scheme 4.15 Sammes’ model study of proposed cycloaddition.

Williams and coworkers expanded on the pioneering work of Sammes withthe following biosynthetic proposal for the brevianamides [5]. DeoxybrevianamideE (18) was thought to undergo oxidation to hydroxyindolenine 7, which couldundergo a pinacol-type rearrangement to indoxyl 87 (Scheme 4.16). Subsequenttwo-electron oxidation and enolization to azadiene 88, followed by an intramolecularhetero-Diels–Alder reaction was, following from the original proposal of Sammes,envisioned to give the natural products (+)-brevianamide A and (+)-brevianamide

356

128 4 Biomimetic Synthesis of Alkaloids Derived from Tryptophan: Dioxopiperazine Alkaloids

NH

NH

O N

OH H

MeMe

deoxybrevianamide E (18)

[ox]

N

NH

O N

OH H

MeMe

HOrearrangement N

H

O N

OH H

MeMe NH

O

2e# ox

NH+

O N

OMeMe NH

OFe3+

enolization

NNH+O

O#

MeMe

H

HN O

N

H+N

Me

H

HNO

#OMe

O

O

H

NNH

O(+)-brevianamide A (89)

NH

OMe

Me

O

H

NNH

O(+)-brevianamide B (90)

N

O

Me MeH

7 87

8888a 88b

Scheme 4.16 Biosynthetic proposal for the brevianamides.

B. The pseudo-enantiomorphic relationship between the two natural productswas envisaged to arise from the equilibrium between conformers 88a and 88b,which undergo cycloaddition to give 89 and 90, respectively. Ab initio studiesof the two transition states demonstrated that 88a is the more stable of thetwo, which is consistent with the observed product ratios of 89 and 90. Thetheoretical insights published by Domingo et al. lend support to the proposalof a biosynthetic intramolecular Diels–Alder reaction of intermediate 88 [14].Unfortunately, experimental support for this pathway has been elusive to secureand thus it remains a speculative biogenetic construction.

The total synthesis of d,l-brevianamide B demonstrated the first congru-ent application of a biomimetic Diels–Alder reaction used to form the bicy-clo[2.2.2]diazaoctane core common to the series of prenylated dioxopiperazines tobe discussed in this section [15]. 9-Epi-deoxybrevianamide E (19) was convertedinto the lactim ether (91) and oxidized to give the Diels–Alder precursor 92(Scheme 4.17). Treatment with aqueous methanolic KOH induced tautomeriza-tion to azadiene 93, which underwent a potentially biomimetic intramolecularDiels–Alder cycloaddition to give a mixture of diastereomers (94 and 95, 2 : 1).Oxidation, pinacol-type rearrangement, and lactim ether deprotection of the minordiastereomer (95) afforded brevianamide B (90) in 65% overall yield from 96. Thisstudy was one of the first to experimentally support the biogenetic origin of thecore bicyclo[2.2.2] ring system as arising via a dioxopiperazine that undergoes a nettwo-electron oxidation to an azadiene moiety.

357

4.2 Prenylated Indole Alkaloids 129

NH

MeMe

NHN

O

OH

NH

MeMe

NN

O

OMeH

NH

MeMe

NN

O

OMe

NH

MeMe

NN

O

OMe

MeOH

NN

MeMe

O 95

HN

MeO

H

NN

MeMe

O94

HN

MeOH

NN

MeMe

O

96

N

HO O

H

NNH

O(±)-brevianamide B (90)

N

O

MeMeH

Me3OBF4KOH

MeOH

H2O

9119 92 93

60%94:95 = 2:1

+

1. mCPBA

2. HCl, 99%

1. NaOMe

2. HCl, 65%

69%

DDQ

40%

Scheme 4.17 Application of the proposed biomimetic Diels–Alder reaction.

Williams and coworkers also applied the intramolecular Diels–Alder reaction tothe racemic synthesis of VM55599 [16]. The reverse-prenylated tryptophan deriva-tive 97 was coupled to !-methyl-!-hydroxyproline ethyl ester 98 to afford dipeptide99 (Scheme 4.18). N-Boc deprotection and cyclization afforded dioxopiperazine 100,and elimination with thionyl chloride gave enamide 101. Formation of the lactimether (102) and treatment with aqueous KOH gave azadiene 103, which sponta-neously suffered intramolecular Diels–Alder reaction to give a separable mixture ofall four possible racemic diastereomers (104–107, 2.6 : 3.7 : 1.0 : 1.6, respectively).Cleavage of the lactim ether and diisobutylaluminum hydride (DIBAL-H) reductiongave VM55599 in 73% yield from 105.

The synthesis of VM55599 allowed for assignment of the absolute stereochem-istry of the molecule, which places the methyl group at the !-position of theproline residue syn- to the bridging isoprene moiety [16]. In stark contrast, theanalogous methyl group of paraherquamide A is anti to the bridging isopreneunit. Scheme 4.19 outlines a possible unified biosynthesis for VM55599 and para-herquamide A, arising from dimethylallyl pyrophosphate (DMAPP), l-isoleucine,and l-tryptophan. If a Diels–Alder cycloaddition is to be invoked, approach of theisoprene moiety must occur from the same face as the methyl group on the prolinering for synthesis of VM55599, and from the opposite face to the methyl group togive paraherquamide A. The diastereofacial selectivity of the Diels–Alder reactiongave a preponderance of the syn-relative stereochemistry alpha to the gem-dimethylgroup in both molecules. As VM55599 is a very minor metabolite of Penicilliumsp. IMI332995, it is plausible that cyclization of 112#114 is preferred and furthermetabolization gives paraherquamide A, whereas the minor cycloaddition via 113produces VM55599 as a dead-end shunt metabolite. As shown in Scheme 4.18

358

130 4 Biomimetic Synthesis of Alkaloids Derived from Tryptophan: Dioxopiperazine Alkaloids

NH

CO2HBocHN

MeMe

BOPCl

TFA$HN

EtO2CMeHO

98

83%NH

BocHN

Me Me

ON

CO2Et

MeOH

1. TFA

2. 2-OH-pyr.", 95%

97 99

NH

MeMe

NHN

O

O

100

MeOH

SO2Cl, py.

75%

NH Me

Me

NHN

O

O

101

Me

Me3OBF4

72%

NH

MeMe

NN

O

OMe

102

Me

KOH

NH

MeMe

NN

O

OMe

103

Me

MeO

NN

HN MeMe

O

HH

Me

MeO

NN

HN MeMe

O

HH

Me

MeO

NN

HN MeMe

O

MeH

H

MeO

NN

HN MeMe

O

MeH

H

O

H

NN

HN Me

Me

HMe

H

VM55599 (108)

1. HCl2. PhCH3, "

3. DIBAL-H73%104

106

105

107

104:105:106:107 = 2.6 : 3.7 : 1.0 : 1.6

Scheme 4.18 Williams’ biomimetic total synthesis of VM55599.

HN

HO2C

H2N

Me

Me

PPO

CO2H

NH2

Me

Me

DMAPP (109)

L-Ile (110) L-Trp (111)

OH

NN

HNMe Me

HMe

O

H

NN

HN MeMe

HMe

H

X

HO

NN

HN MeMe

MeH

X

VM55599 (108)minor

O

H

NN

HNMe

Me

HH

Me

X

syn-selective [4+2]

OMe

NHO

NN

MeH

HMeMe

O

O

MeMe

[ox]SAM

DMAPP

114major

paraherquamide A (115)

112 113

Scheme 4.19 Proposed biosynthesis of paraherquamide A and VM55599.

359

4.2 Prenylated Indole Alkaloids 131

above, the intrinsic diastereofacial bias of the Diels–Alder reaction is modest atbest, giving a slight excess (1.47 : 1) of cycloaddition from the same face as themethyl group, and favoring the syn-relative stereochemistry to the extent of 2.4 : 1.Such observations suggest that the biosynthesis may rely on protein organizationof the precyclization conformers to stereoselectively produce the syn-isomers.

A report from Liebscher and coworkers showed promise in terms of improvingthe diastereoselectivity of the Diels–Alder cycloaddition, using neutral conditionsto prepare the azadiene in contrast to the basic conditions employed by Williams[17]. Compound 118 was prepared by a Horner–Wadsworth–Emmons reactionof aldehyde 116 with phosphonate 117 (Scheme 4.20). Treatment of 118 withneat acetyl chloride for 20 days gave the Diels–Alder product 119 as a singlediastereomer in 48% yield.

NMOM

CHO

MeMe

HNN

O

OP

(MeO)2

O

H117

KOtBuN

NH

NO

O

MeMe

118

H

116

AcCl

20 d O

H

NN

O

HN MeMe

H

119one diastereomer

MOM48%

Scheme 4.20 Liebscher’s Diels–Alder work.

The precedent set by Liebscher’s work was applied to an asymmetric totalsynthesis of VM55599 [18]. The loss of stereochemistry observed at the prolinemethyl group in 101 above led Williams to employ a dehydrotryptophan derivativeas the Diels–Alder azadiene precursor, allowing for the preparation of VM55599 inan enantioselective fashion. Williams has demonstrated that the !-methylprolineresidue of paraherquamide A and VM55599 are biosynthetically derived from l-Ile.In a biomimetic construct, l-Ile was converted into optically pure !-methylproline120 using Hoffman–Loffler–Freytag conditions in 45% yield (Scheme 4.21).

MeMe

CO2HH2NNBoc

Me

CO2Et

45%L-Ile (110) 120

S

S SS LiOH

96%NBoc

Me

CO2H

121

HNN

O

O

Me

122

1. EDCI, HOBtEt3N

ClH3N CO2Me

2. TFA; Et3N3. PhCH3, "

88%

NaH,MeSCH2Cl

62%

NN

O

O

Me

123

SMe N

CHO

MeMe

NaHMDS87%

116

NMe Me

124

NN

MeO

O

MeSHO

H

NH

MeMe

125

N

NH

OO

Me

1. MeI, NaHCO3, 92%

2. 50% aq.

MOM

MOMHCO2H, 70%

Scheme 4.21 Key dioxopiperazine synthesis.

360

132 4 Biomimetic Synthesis of Alkaloids Derived from Tryptophan: Dioxopiperazine Alkaloids

NH

MeMe

125

N

NH

OO

Me

AcCl

14 days

NH

MeMe

126

N

NAcO

O

MeH

NH Me

Me

127

N

NAcO

O

Me

H+

O

H

NN

O

HN Me

Me

HMe

H

O

H

NN

O

HN MeMe

HMe

H

S

SO

H

NN

O

HNMe

Me

HH

Me

O

H

NN

O

HNMe

Me

HH

Me

S

S129 (35%)

131 (15%)

128 (10%)

130 (0%)

O

H

NN

HN Me

Me

HMe

HS

(–)VM55599 (108)

DIBAL-H

68%

Scheme 4.22 Completion of the asymmetric total synthesis of ($)-VM55599.

Cleavage of the ethyl ester, peptide coupling to glycine methyl ester hydrochloride,Boc deprotection, and cyclization gave dioxopiperazine 122 in good overall yield.Protection of the secondary amide and subsequent condensation with aldehyde 116gave an epimeric mixture of dioxopiperazines, which gave selectively (Z)-isomer125 following deprotection and dehydration.

Following Liebscher’s protocol, 125 was treated with acetyl chloride for 14 days,yielding a mixture of three diastereomers (Scheme 4.22). The reaction is thoughtto proceed by initial acylation to the O-acyl lactim 126, tautomerization to azadiene127, which suffers intramolecular Diels–Alder reaction from three of the four pos-sible diastereomeric transition states, followed by loss of acetate to give compounds128–131. The major diastereomer 129 was treated with excess DIBAL-H to effectreduction to ($)-VM55599 (108). Interestingly, cycloadduct 130 was not observedfrom the cycloaddition reaction. Penicillium sp. produces paraherquamide A inlarge excess over VM55599 (>600 : 1), so it is surprising that this provocative bio-genetic precursor to paraherquamide A is not observed in laboratory cycloadditionreactions. Despite the structural differences between the laboratory and biologicalDiels–Alder precursors, the intrinsic facial selectivity of the cyclization does notappear to mimic the bias toward paraherquamide stereochemistry one wouldexpect given the observed product ratios of the fungal metabolites.

As many of the bicyclo[2.2.2]diazaoctane natural products differ only at thesubstitution of the indole ring, a convergent approach utilizing a Fischer indolesynthesis was undertaken in an alternative racemic synthesis of brevianamideB [19]. This work had the objective of validating other possible biosynthetic pathways

361

4.2 Prenylated Indole Alkaloids 133

O

Me Me

S S

EtO2C H

nBuLi, CuI

76%132

O

Me Me

134

EtO2CS S

LiOH

99%

O

Me Me

135

HO2CS S

NH

CONH2

H

BOPCl, iPrNEt

77%

136

O

Me Me

137

S SN

O

O NH2H

NCS, AgNO3

88%

138:139 = 45:43

O

Me Me

138

N

O

O NH2H

O

AlCl3

EtOAc

48%

NNH

O

O O

Me Me

H

+

139

O

H

NN

Me Me

H

O

O

140

1. PhNHNH2

2. ZnCl2, 58%O

H

NN

Me Me

H

O141

HN

1. mCPBA

2. NaOH, 16%O

H

NNH

O(±)-brevianamide B (90)

N

O

Me MeH

133

Scheme 4.23 Concise synthesis of brevianamide B.

to reach the oxidation state of the azadiene. In the present instance, the "-ketoamidespecies (138) was posed to serve as a surrogate for the possible biosynthetic oxidativedeamination of the tryptophan moiety and coupling to a proline amide species(Scheme 4.23). Conjugate addition of carboxylate 133 to ketone 132 gave ester 134in 76% yield. Saponification, peptide coupling with l-proline amide, and dithianedeprotection gave a mixture of the uncyclized amide 138 and dioxopiperazine139. Aluminum trichloride was added to the mixture to give the Diels–Aldercycloadduct (140) in exclusively the anti-configuration, whereas mixtures of boththe syn- and anti-cycloadducts were observed in previous syntheses of VM55599and brevianamide B discussed above. A Fischer indole synthesis was completed bytreatment of 140 with phenyl hydrazine followed by ZnCl2, affording 141 in goodyield. Oxidation and pinacol-type rearrangement of this known intermediate gavebrevianamide B. While an appealing convergent approach towards the synthesisof other related bicyclo[2.2.2]diazaoctanes, the utility of this strategy is limited tothose natural products containing the anti-stereochemistry observed in formationof compound 140, the remainder of which must be able to withstand the harshconditions of the Fischer indole synthesis.

The same biomimetic Diels–Alder disconnection was exploited in the total syn-thesis of stephacidin A [20]. Starting with reverse prenylated tryptophan derivative142 (prepared in eleven steps from 6-hydroxyproline), dipeptide 144 was preparedthrough bis(2-oxo-3-oxazolindinyl)phosphinic chloride (BOPCl) mediated coupling

362

134 4 Biomimetic Synthesis of Alkaloids Derived from Tryptophan: Dioxopiperazine Alkaloids

step

haci

din

A (

150)

OH

NN

O

H NM

eM

eO

Me

Me

N HO

Me M

e

N H

N

HHO

O

OH

Me

Me

145

N HO

Me M

e

N H

NHO

O

Me

Me

146

N HO

Me M

e

N

NHO

OM

e

Me

Me

147

MeOH

NN

O

H NM

eM

eO

Me

Me

N HO

Me M

e

NH

Fm

oc

NHO M

eM

e

144

EtO

2CO

H

N HO

Me M

e

NH

Fm

oc

CO

2H

Me

Me

142

N H·H

Cl

EtO

2CHO

143

BO

PC

l,iP

rNE

t 254

%

N HO 95%

PB

u 3, D

EA

D

91%

Me 3

OB

F4

Cs 2

CO

3

81%

KO

H, M

eOH

86%

148:

149

= 1:

2.4

148

MeOH

NN

O

H NM

eM

eO

Me

Me

149

+H

HC

l

96%

Sche

me

4.24

Synt

hesi

sof

step

haci

din

A.

363

4.2 Prenylated Indole Alkaloids 135

with cis-3-hydroxyproline ethyl ester (143, Scheme 4.24). Fmoc deprotection resultedin the cyclization to dioxopiperazine 145, which upon treatment with tributyl phos-phine and diethyl azodicarboxylate (DEAD) underwent Mitsunobu dehydration togive enamide 146. Formation of the lactim ether (147) was followed by intramolec-ular Diels–Alder reaction to give a mixture of epimers enriched with the syn-isomer(149, 2.4 : 1). Deprotection of 149 gave stephacidin A (150) in excellent yield.

Williams found that the bicyclo[2.2.2]diazaoctane core could be accessed directlyfrom compound 145 by treating it with excess PBu3 and DEAD, effecting the dehy-dration, tautomerization, and Diels–Alder reaction in one pot to afford stephacidinA and its epimer (Scheme 4.25) [21].

NHO

MeMe

NH

N

HHO

O

OH

Me Me

145

PBu3

DEAD

40 °C64%

stephacidin A (150)

O

H

NN

O

HN MeMeO

Me

Me

H

+ epi

150:epi = 2.4:1

Scheme 4.25 Improved biomimetic synthesis of stephacidin A.

Myers and coworkers, in the course of their total synthesis of avrainvillamide,discovered that synthetic ($)-152 spontaneously dimerized to stephacidin B underseveral mild conditions, including addition of triethylamine or exposure to silica gel[22, 23]. Baran and coworkers later determined that stephacidin A could be readilyconverted into avrainvillamide through reduction to indoline 151 followed bySomei oxidation (Scheme 4.26). In accord with Myers’ observations, dimerizationto stephacidin B occurred readily upon exposure to silica gel, triethylamine,or on evaporation from dimethyl sulfoxide (DMSO) [24–26]. Myers has furtherdemonstrated that the observed biological activity of stephacidin B may be due tothe formation of 152 from 153 in vivo [22].

Stephacidin A is of particular biogenetic interest, as both enantiomers havebeen isolated in nature: (+)-stephacidin A from Aspergillus ochraceus [27] andfrom a marine-derived Aspergillus sp. [28] and ($)-stephacidin A from terrestrialAspergillus versicolor [29–31]. Operating under the assumption that the biosynthe-sis of stephacidin A proceeds through a common, achiral intermediate, Williamsand coworkers have proposed notoamide S (154) as the point of divergence in thetwo biosyntheses [32]. Oxidation of 154 could give achiral azadiene 155, whichis postulated to undergo intramolecular Diels–Alder reaction to give either (+)-or ($)-stephacidin A depending on the enantiofacial selectivity of the reaction(Scheme 4.27).

Tryptophan derivative 142 used in the above synthesis of stephacidin A was alsoemployed in the biomimetic total synthesis of marcfortine C (164) [33]. Pipecolicacid derivative 156 was coupled to acid 142 to form amide 157, which underwentcyclization to the dioxopiperazine following Fmoc-deprotection to give 158 as

364

136 4 Biomimetic Synthesis of Alkaloids Derived from Tryptophan: Dioxopiperazine Alkaloids

stephacidin A (150)

O

H

NN

O

HN MeMeO

MeMe

H

N O

MeMe

Me Me

NNO

O

H

N

O MeMe

MeMe O

OH

H

HNO

N O

H

O

H

NN

O

HN MeMeO

MeMe

H

O

H

NN

O

N MeMeO

MeMe

H

O

avrainvillamide (152)

stephacidin B (153)

NaBH3CN

AcOH

SeO2, H2O2

SiO2

151

Scheme 4.26 Biomimetic conversion of stephacidin A into stephacidin B.

(#)-stephacidin A, (#)-150

O

H

NN

O

HN MeMeO

Me

Me

H

(+)-stephacidin A, (+)-150

O

H

NN

O

HNMe

Me O

Me

Me

H

NH

NH

NO

OH

H

Me MeHO

MeMe

NH

N

NO

OH

Me MeHO

MeMe

notoamide S (154)

[ox]

Aspergillusversicolor

Aspergillussp.

IMDA

[ox]

155(achiral)

Scheme 4.27 Proposed biosynthesis of (+)- and ($)-stephacidin A through notoamide S.

an inconsequential mixture of diastereomers (Scheme 4.28). The biomimeticDiels–Alder reaction preferred the syn-diastereomer 160 in 2.4-fold excess, asexpected. Excess DIBAL-H selectively reduced the tertiary amide of 160, and aminesalt formation followed by a biomimetic oxidative rearrangement gave marcfortineC (164).

Malbrancheamide and malbrancheamide B were synthesized via the biomimeticDiels–Alder reaction of enamides 165 and 166 to afford both syn-cycloadducts 167

365

4.2 Prenylated Indole Alkaloids 137

N H

CO

2H

NH

Fm

oc

OM

eM

e

Me

Me

N HC

O2E

t

OH

N H

NH

Fm

oc

ON

EtO

2C

OM

eM

e

Me

Me

OH

N HO

Me

Me

Me

Me

N H

NO

OHH

OH

Bu 3

P, D

EA

D

40°C

60%

159:

160

= 1:

2.4

+OH

NN

O

H NM

eM

eO

Me M

e

OH

NN

O

H NM

eM

eO

Me M

e

HH

OH

NN

H NM

eM

eO

Me M

e

H

NS

OnB

u OO

OH

NN

H NM

eM

eO

Me M

e

H

H OT

s

+m

arcf

ortin

e C

(16

4)

OH NNH

NH

Me

Me

O

OM

e

Me

142

156

157

158

159

160

BO

P,i

Pr 2

NE

t77

%94

%O N H

DIB

AL-

H

89%

PP

TS

77%

161

162

163

Sche

me

4.28

Tota

lsy

nthe

sis

ofm

arcf

ortin

eC

.

366

138 4 Biomimetic Synthesis of Alkaloids Derived from Tryptophan: Dioxopiperazine Alkaloids

NHCl

NH

NHO

O

Me Me

X

165: X = Cl166: X = H

O

H

NN

HNMe Me Cl

H Cl

malbrancheamide B (172)

O

H

NN

HNMe Me Cl

H

O

H

NN

HNMe Me Cl

H Cl

O

H

NN

HNMe Me Cl

H

O

O

DIBAL-H

80%

DIBAL-H

74%

X = Cl

89%167:168 = 2:1

167

+ epi (168)

169

+ epi (170)

X = H

86%169:170 = 1.8:1

KOH

MeOH

malbrancheamide (171)

Scheme 4.29 Completion of the total syntheses of the malbrancheamides.

and 169, as well as the anti-epimers 168 and 170 (Scheme 4.29) [34]. Treatmentof the syn-cycloadducts with excess DIBAL-H gave either malbrancheamide ormalbrancheamide B.

As the malbrancheamides were the first of this family of prenylated indolealkaloids to possess a halogenated indole ring, Williams and coworkers probed thebiosynthesis to establish the timing of the chlorination event [35]. Malbrancheamideis proposed to arise from tryptophan, proline, and dimethylallyl diphosphate,leading to deoxybrevianamide E (18, Scheme 4.30). Oxidation of 18 could giveintermediate 174, which is expected to suffer intramolecular Diels–Alder reaction

NH

CO2H

NH2

Me

Me

OPP

HN

HO2C

NH

NH

NO

OH

H

Me Me NH

N

NX

OH

Me Me

[ox]

O

H

NN

HNMe Me

H

X

IMDA

174, X = O175, X = H2

176, X = O

redO

H

NN

HNMe

Me

Hhalogenase

O

H

NN

HNMe Me

H

Cl

malbrancheamide B (172)

halogenaseO

H

NN

HNMe Me

H

Cl

malbrancheamide (171)

Cl

111

173

109 18

177

Scheme 4.30 Proposed biosynthesis of the malbrancheamides.

367

4.2 Prenylated Indole Alkaloids 139

ON H

OM

e Me

HN

NO

O

Me

Me

H

OH

H

178

ON H

OM

e Me

HN

NO

O

Me

Me

H

180

ON H

O

Me M

e

HN

NO

O

Me

Me

H

OH

H

179

ON H

O

Me M

e

HN

NO

O

Me

Me

H

181

+

CH

2Cl 2

80%

178:

179

= 3:

1

DE

AD

, PB

u 3

CH

2Cl 2

71%

DE

AD

, PB

u 3

CH

2Cl 2

78%

163S

N

nBu

O

OO

N HO

Me M

e

N H

N

HHO

O

OH

Me

Me

145

Sche

me

4.31

Synt

hesi

sof

key

oxin

dole

s.

368

140 4 Biomimetic Synthesis of Alkaloids Derived from Tryptophan: Dioxopiperazine Alkaloids

(IMDA) to cycloadduct 176. Reduction of the tertiary amide would provide premal-brancheamide (177). Alternatively, 18 could suffer reduction of the tertiary amideto 175, providing 177 directly upon cycloaddition. Premalbrancheamide is pro-posed to undergo subsequent halogenation events to give both malbrancheamideB and malbrancheamide. In feeding studies, labeled dioxopiperazine 176 andpremalbrancheamide (177) were added to separate cultures of Malbranchea au-rantiaca, but interestingly only premalbrancheamide 177 was incorporated intomalbrancheamide B. This suggests that reduction of the tertiary amide mustprecede Diels–Alder construction of the bicyclo[2.2.2]diazaoctane core through anintermediate analogous to monooxopiperazine 175.

Williams has reported that–like stephacidin A and notoamide B, which areproduced in Nature as distinct enantiomers–the minor metabolite versicolamideB is likewise produced as distinct enantiomers in different strains of Aspergillus sp.The asymmetric syntheses of both (+)- and ($)-versicolamide B (182) have recentlybeen accomplished by deploying compound 145, previously used in the synthesisof stephacidin A (Scheme 4.31). Oxaziridine oxidation of 145 and pinacol-typerearrangement consequent to oxidation of the indole gave a 3 : 1 separable mixtureof 178 and 179 [36]. As previously reported, treatment with tributyl phosphine andDEAD induced Mitsunobu dehydration to give enamides 180 and 181.

Treatment of 180 and 181 individually with potassium hydroxide in methanolinduced the intramolecular Diels–Alder reaction to afford either (+)- or($)-versicolamide B (182), along with the minor anti-diastereomers (+)-183 or($)-183 (Scheme 4.32). As previous biomimetic Diels–Alder reactions containingindole-based azadienes display syn-selectivity (typically !2.5 : 1 syn : anti), theexclusive selectivity for the anti-products in the versicolamide B syntheses is ofparticular interest. The authors suggest that the anti-preference stems from astable transition state leading to the anti-cycloadduct when an oxindolic azadieneis employed, whereas the syn- and anti-transition states arising from an indolicazadiene are of roughly equal stability [14, 37].

O NH

OMe

Me

HN

NO

O

MeMe H

180

O NH

O

Me

Me

HN

NO

O

MeMe H

181

KOH

MeOH75%

182:183 = 1.4:1

KOH

MeOH72%

182:183 = 1.4:1

(+)-versicolamideB, (+)-182

O

NHO

NN

OMe

Me

MeMe

O

O

HNO

NN

OMe

Me

MeMe

O

(+)-183

H H+

H H

(#)-versicolamide B, (#)-182 (#)-183

O

HNO

NNO

MeMe

MeMe

O

O

NHO

NN O

MeMe

MeMe

O

H H+

H H

Scheme 4.32 Completion of the asymmetric syntheses of the versicolamides.

369

4.3 Non-prenylated Indole Alkaloids 141

While the above syntheses have differed in the approach to the azadiene, allshare a common biomimetic intramolecular Diels–Alder reaction to give thebicyclo[2.2.2]diazaoctane core featured in the molecules presented in this section.

4.3Non-prenylated Indole Alkaloids

Hart and coworkers have expressed interest in the biosynthesis of the fumiquina-zoline family of alkaloid natural products, and reported a biomimetic synthesisof ent-alantrypinone as the initial effort at a synthetic program to access morecomplex, related substances [38]. l-Tryptophan methyl ester was coupled to isatoicanhydride to afford amide 184 in good yield (Scheme 4.33). Acylation of 184 withacyl chloride 186 (prepared in two steps from S-methyl-l-cysteine, 185) underSchotten–Baumann conditions furnished diamide 187, which underwent cyclode-hydration to iminobenzoxazine 188. Treatment with excess Li[Me3AlSPh] effectedthe rearrangement to quinazolinone 189, and Fmoc deprotection was accompaniedby amide formation to give 190. Oxidation of 190 provided the sulfoxide, which wasconverted into enamide 191 upon heating in benzene with triphenylphosphine.Trifluoroacetic acid induced the conversion into bicycle 192, presumably throughintramolecular electrophilic attack of an intermediate N-acyliminium ion ontoindole. Conversion into the oxindole proceeded via oxidative rearrangement of 192with NBS to give the polybrominated indolinone, which was hydrogenolyzedover platinum on carbon to give ent-alantrypinone (193), along with ent-17-epi-alantrypinone (194).

Three dimeric tryptophan-derived dioxopiperazines have succumbed tobiomimetic total syntheses, all completed by Movassaghi and coworkers, namely,(+)-WIN 64821, ($)-ditryptophenaline, and (+)-11,11"-dideoxyverticillin A [39, 40].Syntheses of the former two began with cleavage of the Boc carbamate to effect thecyclization to dioxopiperazine 196 (Scheme 4.34) [40]. Treatment with brominethen gave a separable mixture of two diastereomers, endo-(+)-197 and exo-($)-198.endo-Bromide (+)-197 was carried on to (+)-WIN 64821 (201) by, first, treatmentwith tris(triphenylphosphine)cobalt chloride to afford the dimerized product 199,which was globally deprotected upon exposure to samarium diiodide. (+)-WIN64821 was thus obtained in 75% yield. Similarly, ($)-ditryptophenaline (202) wassynthesized following methylation of exo-($)-198, dimerization, and deprotectionto give the product in 79% yield.

4.3.1Epidithiodioxopiperazines

(+)-11,11"-Dideoxyverticillin A (211) differs from dimers 201 and 202 in that it islikely derived from l-tryptophan and l-alanine, rather than from l-phenylalanine.Additionally, the dioxopiperazines are bridged by a disulfide, adding a difficultchallenge to the synthetic construction of the molecule. Similar to their previous

370

142 4 Biomimetic Synthesis of Alkaloids Derived from Tryptophan: Dioxopiperazine Alkaloids

isat

oic

anhy

drid

e

97%

CO

2Me

NH

2N

H"

96%

NH

N H

MeO

2CO

NH

2

NH

N H

MeO

2CO

HN

NH

Fm

ocSM

e

HO

ON

H2

SM

e1.

Fm

ocC

l

89%

2. S

OC

l 2C

l

ON

HF

moc

SM

e

DC

M

H2O

, NaH

CO

3

80%

Ph 3

P, I

2, D

CM

76%

Li[M

e 3A

lSP

h]

NH

N

MeO

2CN

O

Fm

ocH

NS

Me

NH

N

MeO

2C

N

O

Fm

ocH

NS

Me

94%

pipe

ridin

e

NH

NN

O

H NS

Me

O

79%

1.m

CP

BA

2. P

h 3P

,"

NH

NN

O

H NO

89%

TF

A

NH

NN

OO

Me

NH

74%

193:

194

= 1:

1.5

1. N

BS

, TH

F,

TF

A, H

2O

NH

NN

OO

Me

2. H

2, P

t/C

NH

NN

OO

Me

HN

O

NH

O+

ent-1

7-ep

iala

ntry

pino

ne (

194)

ent-a

lant

rypi

none

(19

3)

O

3118

4

185

186

187

188

189

190

191

192

iPr 2

NE

t

Sche

me

4.33

Synt

hesi

sof

ent-a

lant

rypi

none

.

371

4.3 Non-prenylated Indole Alkaloids 143

NH

BocO

HN

PhS

O2N

TF

A;

mor

phol

ine

80%

PhS

O2N

86%

HN

NH

O O

Br 2

N SO

2Ph

SO

2PhN

NH

Br

H

O O

48%

CoC

l(PP

h 3) 3

OM

eO

N SO

2PhN

NH

H

O O

SO

2Ph

NN

HN

H

OO

75%

Sm

I 2, N

MP

N H

NN

H

H

O O

H NN

HN

H

OO (+)-

WIN

648

21(2

01)

NN

NH

Br

H

O O

197

48%

2. C

oCl(P

Ph 3

) 3

NN

NM

e

H

O O

SO

2Ph SO

2Ph

NN

MeN

H

OO

79%

Sm

I 2, N

MP

N H

NN

Me

H

O O

H NN

MeN

H

OO

(#)-

ditr

ypto

phen

alin

e (2

02)

198

1. M

eI

195

196

197

198

199

200

Sche

me

4.34

Con

cise

tota

lsy

nthe

ses

of(+

)-W

IN64

821

and

($)-

ditr

ypto

phen

alin

e.

372

144 4 Biomimetic Synthesis of Alkaloids Derived from Tryptophan: Dioxopiperazine Alkaloids

NHBocO

HN

Me

CO2Me

PhSO2N

TFA;morpholine

84%PhSO2N

59%

HNNH

O

OMe

1. Br2

NSO2Ph

NNMeBr

H

O

OMe2. MeI

46%

CoCl(PPh3)3

NSO2Ph

NNMe

H

O

OMe

SO2PhN

NMeN

H

O

OMe

63%

Py2AgMnO4

NSO2Ph

NNMe

H

O

OMe

SO2PhN

NMeN

H

O

OMe

HO

OHHO

OH55%

TBSCl

NSO2Ph

NNMe

H

O

OMe

SO2PhN

NMeN

H

O

OMe

TBSO

OHHO

OTBS

PPY

87%

Na(Hg)

NH

NNMe

H

O

OMe

HN

NMeN

H

O

OMe

TBSO

OHHO

OTBS 56%

1. K2CS3, TFA

2. ethanolamine

NH

NNMe

H

O

OMe

HN

NMeN

H

O

OMe

HS

SHHS

SH 87%

KI3

pyridine

NH

NNMe

H

O

O

HN

NMeN

H

O

OMe

Me

SS

SS

(+)-11,11%-dideoxyverticillin A (211)

203 204 205

206 207

208 209

210

Scheme 4.35 Biomimetic total synthesis of (+)-11,11"-dideoxyverticillin A.

work, Movassaghi and coworkers showed that cleavage of the N-Boc carbamate wasaccompanied by cyclization to dioxopiperazine 204 (Scheme 4.35) [39]. Exposure of204 to bromine produced the 3-bromopyrroloindoline, and the amides were sub-sequently methylated upon treatment with iodomethane. Reductive dimerizationwith the cobalt(I) complex as before gave the desired dimeric intermediate 206. Thedimer was oxidized with bis(pyridine)-silver(I) permanganate to octacycle 207, andexposure to Fu’s (R)-(+)-4-pyrrolidinopyridinyl(pentamethylcyclopentadienyl)iron(PPY) catalyst with t-butyldimethylsilyl chloride (TBSCl) gave selectively thealanine-derived protected hemiaminals of 208. Removal of the benzenesulfonylgroups with sodium amalgam revealed diaminodiol 209. Treatment of 209 withK2CS3 followed by ethanolamine gave diaminotetrathiol 210, which readily oxi-dized to (+)-11,11"-dideoxyverticillin A (211) when partitioned between aqueoushydrochloric acid and dichloromethane and treated with potassium triiodide.

373

4.3 Non-prenylated Indole Alkaloids 145

The total synthesis of sporidesmin A was completed by Kishi and coworkersin 1973. In a series of communications, Kishi described a novel strategy for thesynthesis of epidithiodioxopiperazines using a dithioacetal moiety as a protectinggroup for the disulfide bridge [41–43]. Thus protected, the dithioacetal is stableto acidic, basic, and reducing conditions, allowing for the introduction of thiolgroups at an early stage in a total synthesis. Synthesis of the sporidesmins be-gan with the treatment of dioxopiperazine 212 with the dithiane derivative ofp-anisaldehyde in the presence of acid to afford dithioacetal-protected dioxopiper-azine 213 (Scheme 4.36) [43]. Condensation with acid chloride 214 and subsequentmethoxymethyl deprotection gave compound 215. Treatment of ketone 215 withDIBAL-H at $78 %C resulted in stereoselective reduction to the alcohol, which wasthen converted into acetate 216 in 80% yield. Cyclization to the diacetate (217)proceeded upon addition of iodosobenzene diacetate, and hydrolysis of the acetatesgave the corresponding diol. Treatment of the diol with m-chloroperbenzoic acid(mCPBA) afforded an intermediate sulfoxide, which decomposed to the disulfideupon exposure to strong Lewis acid, revealing (±)-sporidesmin A (218).

NNMe

O

O

MeO

AcS

p-MeOC6H4CH2S2H2,H+

NMe

COClCl

MeOOMe

+

50%

1. BuLi2. HCl, THF

3. NaOH

NMe

Cl

MeOOMe

O

HNNMe

O

OMe

SS Ar

NNMe

O

OMe

SS ArMeO

80%

1. DIBAL-H2. Ac2O

NMe

Cl

MeOOMe

HNNMe

O

OMe

SS Ar

AcO

30%

iodosobenzenediacetate

NMe

Cl

MeOOMe

N

25%

1. NaOH, MeOH2. mCPBA

H

AcOAcO

3. BF3$Et2O

(±)-sporidesmin A (218)

212 213 214

215 216

217

NMe

O

OMe

SS Ar

NMe

Cl

MeOOMe

NH

HOHO

NMe

O

OMe

SS

Scheme 4.36 Total synthesis of (±)-sporidesmin A.

The biosynthesis of gliotoxin is believed to proceed through the intramolecularnucleophilic ring-opening of a phenylalanine-derived arene oxide and has beenthe subject of considerable speculation and interest. Kishi and coworkers drewinspiration from these biogenetic hypotheses in devising a brilliant total synthesisof gliotoxin. The total synthesis of (±)-gliotoxin was completed in 1976 utilizingthe same disulfide protecting strategy as deployed above for the sporidesmins,and was re-engineered in 1981 by the same route starting from optically pure

374

146 4 Biomimetic Synthesis of Alkaloids Derived from Tryptophan: Dioxopiperazine Alkaloids

HNN

O Triton B

O Me

SS Ar

O

O

OtBu+N

N

O

O Me

SS Ar

OOtBu

HO NN

O

O Me

SS Ar

OOtBu

HO+

1. Ac2O2. TFA

3. ClCO2Et4. NaBH2

77%

98%

NN

O

O Me

SS Ar

OH

AcO

1. MsCl2. LiCl

3. NaOMe90%

NN

O

O Me

SS Ar

Cl

HO

PhLi,PhCH2OCH2Cl

53%

NN

O

O Me

SS ArHO H

OCH2OPh

1. BCl32. mCPBA

35%

NN

O

O Me

SSHO H

OH

3. HClO4

219 220221 222

221:222 = 2:1

223 224

225 (+)-gliotoxin (226)

Scheme 4.37 Kishi’s total synthesis of (±)- and (+)-gliotoxin.

dithioacetal 219 obtained from resolution (Scheme 4.37) [44, 45]. Coupling of 219with t-butoxy arene oxide 220 in the presence of triton B afforded 221 and 222 in a2 : 1 ratio. Acylation, deprotection, mixed anhydride formation, and reduction gavealcohol 223 in 77% yield. Alcohol 223 was converted into the chloride followingmesylation, and then deprotected to reveal alcohol 224. The key stereoselectivecyclization–alkylation reaction was achieved upon addition of phenyllithium to 224and phenoxymethyl chloride, affording cycloadduct 225 in modest yield (53%). Theprimary alcohol was revealed upon removal of the benzyl ether, and the thioacetaloxidatively removed to afford either (±)- or (+)-gliotoxin (226).

4.4Conclusion

Dioxopiperazine alkaloids cover an astonishing array of molecular architectureand, with that, corresponding synthetic challenges to construct such substances.The biosynthesis of many of the natural substances touched on in this chapter has,in some instances, been studied going back several decades, and many workershave sought to exploit insights from Nature’s strategic bond constructions in asynthetic laboratory context. Advances in whole genome sequencing have broughtnew and invigorated interest in elucidating the biosynthesis of structurally intrigu-ing and biomedically relevant secondary metabolites. The insights to be gainedfrom educated guess work on what specific compounds might lie along a pos-sible biosynthetic pathway, traditionally accomplished by isolation and structural

375

References 147

elucidation to map metabolite co-occurrence in conjunction with isotopically la-beled precursor incorporation experiments, is in the process of giving way to amuch higher resolution picture of secondary metabolism in microorganisms andplants. With the advent of powerful new genomics and proteomics tools to studyand manipulate secondary metabolite production, advances in our understandingof Nature’s creative synthetic palette will surely explode in the coming years.The fruits of these insights will undoubtedly be extensively exploited by syntheticchemists working at the forefront of complex molecule synthesis. In addition, manynew natural products–the biosynthetic intermediates themselves, often isolated intrace amounts if at all–will provide and constitute worthy new synthetic targetsand substrates for various important applications.

Acknowledgment

R.M.W. is grateful to the National Institutes of Health (GM068011; CA70375;CA85419) for financial support.

References

1. Birch, A.J. and Russell, R.A. (1972)Tetrahedron, 28, 2999–3008.

2. Birch, A.J. and Wright, J.J. (1969)J. Chem. Soc., Chem. Commun.,644–645.

3. Birch, A.J. and Wright, J.J. (1970) Tetra-hedron, 26, 2329–2344.

4. Baldas, J., Birch, A.J., and Russell, R.A.(1974) J. Chem. Soc., Perkin Trans. 1,50–52.

5. Sanz-Cervera, J.F., Glinka, T., andWilliams, R.M. (1993) Tetrahedron, 49,8471–8482.

6. Kametani, T., Kanaya, N., and Ihara,M. (1980) J. Am. Chem. Soc., 102,3972–3975.

7. Schkeryantz, J.M., Woo, J.C.G.,Siliphaivanh, P., Depew, K.M., andDanishefsky, S.J. (1999) J. Am. Chem.Soc., 121, 11964–11975.

8. von Nussbaum, F. and Danishefsky,S.J. (2000) Angew. Chem., Int. Ed., 39,2175–2178.

9. Edmondson, S., Danishefsky, S.J.,Sepp-Lorenzino, L., and Rosen, N.(1999) J. Am. Chem. Soc., 121,2147–2155.

10. Finefield, J.M. and Williams, R.M.(2010) J. Org. Chem., 75, 2785–2789.

11. Baran, P.S., Guerrero, C.A., and Corey,E.J. (2003) J. Am. Chem. Soc., 125,5628–5629.

12. Depew, K.M., Marsden, S.P., Zatorska,D., Zatorski, A., Bornmann, W.G., andDanishefsky, S.J. (1999) J. Am. Chem.Soc., 121, 11953–11963.

13. Porter, A.E.A. and Sammes, P.G. (1970)J. Chem. Soc., Chem. Commun., 1103.

14. Domingo, L.R., Sanz-Cervera, J.F.,Williams, R.M., Picher, M.T., andMarco, J.A. (1997) J. Org. Chem., 62,1662–1667.

15. Williams, R.M., Sanz-Cervera, J.F.,Sancenon, F., Marco, J.A., and Halligan,K. (1998) J. Am. Chem. Soc., 120,1090–1091.

16. Stocking, E.M., Sanz-Cervera, J.F., andWilliams, R.M. (2000) J. Am. Chem. Soc.,122, 1675–1683.

17. Jin, S.D., Wessig, P., and Liebscher, J.(2001) J. Org. Chem., 66, 3984–3997.

18. Sanz-Cervera, J.F. and Williams,R.M. (2002) J. Am. Chem. Soc., 124,2556–2559.

19. Adams, L.A., Valente, M.W.N., andWilliams, R.M. (2006) Tetrahedron, 62,5195–5200.

376

148 4 Biomimetic Synthesis of Alkaloids Derived from Tryptophan: Dioxopiperazine Alkaloids

20. Greshock, T.J., Grubbs, A.W.,Tsukamoto, S., and Williams, R.M.(2007) Angew. Chem. Int. Ed., 46,2262–2265.

21. Greshock, T.J. and Williams, R.M.(2007) Org. Lett., 9, 4255–4258.

22. Herzon, S.B. and Myers, A.G. (2005)J. Am. Chem. Soc., 127, 5342–5344.

23. Myers, A.G. and Herzon, S.B. (2003)J. Am. Chem. Soc., 125, 12080–12081.

24. Baran, P.S., Guerrero, C.A.,Ambhaikar, N.B., and Hafensteiner,B.D. (2005) Angew. Chem. Int. Ed., 44,606–609.

25. Baran, P.S., Guerrero, C.A.,Hafensteiner, B.D., and Ambhaikar,N.B. (2005) Angew. Chem. Int. Ed., 44,3892–3895.

26. Baran, P.S., Hafensteiner, B.D.,Ambhaikar, N.B., Guerrero, C.A., andGallagher, J.D. (2006) J. Am. Chem. Soc.,128, 8678–8693.

27. Qian-Cutrone, J., Huang, S., Shu, Y.Z.,Vyas, D., Fairchild, C., Menendez, A.,Krampitz, K., Dalterio, R., Klohr, S.E.,and Gao, Q. (2002) J. Am. Chem. Soc.,124, 14556–14557.

28. Kato, H., Yoshida, T., Tokue, T., Nojiri,Y., Hirota, H., Ohta, T., Williams, R.M.,and Tsukamoto, S. (2007) Angew. Chem.Int. Ed., 46, 2254–2256.

29. Deyrup, S.T., Swenson, D.C., Gloer, J.B.,and Wicklow, D.T. (2006) J. Nat. Prod.,69, 608–611.

30. Mudur, S.V., Gloer, J.B., andWicklow, D.T. (2006) J. Antibiot., 59,500–506.

31. Shim, S.H., Swenson, D.C., Gloer, J.B.,Dowd, P.F., and Wicklow, D.T. (2006)Org. Lett., 8, 1225–1228.

32. McAfoos, T.J., Li, S., Tsukamoto, S.,Sherman, D.H., and Williams, R.M.(2010) Heterocycles, 82, 461–472.

33. Greshock, T.J., Grubbs, A.W., andWilliams, R.M. (2007) Tetrahedron, 63,6124–6130.

34. Miller, K.A., Welch, T.R., Greshock,T.J., Ding, Y.S., Sherman, D.H., andWilliams, R.M. (2008) J. Org. Chem., 73,3116–3119.

35. Ding, Y.S., Greshock, T.J., Miller, K.A.,Sherman, D.H., and Williams, R.M.(2008) Org. Lett., 10, 4863–4866.

36. Miller, K.A., Tsukamoto, S., andWilliams, R.M. (2009) Nat. Chem., 1,63–68.

37. Domingo, L.R., Zaragoza, R.J., andWilliams, R.M. (2003) J. Org. Chem., 68,2895–2902.

38. Hart, D.J. and Magomedov, N.A. (2001)J. Am. Chem. Soc., 123, 5892–5899.

39. Kim, J., Ashenhurst, J.A., andMovassaghi, M. (2009) Science, 324,238–241.

40. Movassaghi, M., Schmidt, M.A., andAshenhurst, J.A. (2008) Angew. Chem.Int. Ed., 47, 1485–1487.

41. Kishi, Y., Fukuyama, T., and Nakatsuk,S. (1973) J. Am. Chem. Soc., 95,6492–6493.

42. Kishi, Y., Fukuyama, T., and Nakatsuk,S. (1973) J. Am. Chem. Soc., 95,6490–6492.

43. Kishi, Y., Nakatsuk, S., Fukuyama, T.,and Havel, M. (1973) J. Am. Chem. Soc.,95, 6493–6495.

44. Fukuyama, T. and Kishi, Y. (1976) J.Am. Chem. Soc., 98, 6723–6724.

45. Fukuyama, T., Nakatsuka, S., and Kishi,Y. (1981) Tetrahedron, 37, 2045–2078.

377

Biomimetic Total Synthesis of Malbrancheamide andMalbrancheamide B

Kenneth A. Miller,† Timothy R. Welch,† Thomas J. Greshock,† Yousong Ding,‡David H. Sherman,‡ and Robert M. Williams*,†,§

Department of Chemistry, Colorado State UniVersity, Fort Collins, Colorado 80523-1872, UniVersity ofColorado Cancer Center, Aurora, Colorado, 80045, and Life Sciences Institute, and Departments of

Medicinal Chemistry, Chemistry, Microbiology & Immunology, The UniVersity of Michigan,210 Washtenaw AVenue, Ann Arbor, Michigan 48109-2216

[email protected]

ReceiVed January 17, 2008

The biomimetic total syntheses of both malbrancheamide and malbrancheamide B are reported. Thesynthesis of the two monochloro species enabled the structure of malbrancheamide B to be unambiguouslyassigned. The syntheses each feature an intramolecular Diels-Alder reaction of a 5-hydroxypyrazin-2(1H)-one to construct the bicyclo[2.2.2]diazaoctane core, which has also been proposed as the biosyntheticroute to these compounds.

Introduction

Our research group has exhibited a long-standing interest inthe synthesis and biosynthetic study of a number of uniqueprenylated indole alkaloids containing a characteristic bicyclo-[2.2.2]diazaoctane core.1 This class of natural products includessuch highly biologically active fungal metabolites as theparaherquamides,2 brevianamides,3 notamides,4 and stephaci-dins,5 among others, which we have shown all arise biogeneti-cally from tryptophan, mevalonate-derived isoprene units, and

proline or derivatives of proline.1 Sammes originally proposedthat the bicyclo[2.2.2]diazaoctane core common to all of thesenatural products arises in Nature via an intramolecular hetero-Diels-Alder reaction of a 5-hydroxypyrazin-2(1H)-one,6 andwork from this laboratory has extensively supported such aproposal.1 In fact, we have applied such a [4+2] hetero-Diels-Alder cycloaddition strategy to the total synthesis of several ofthese prenylated indole alkaloids, including stephacidin A,7brevianamide B,8 marcfortine C,9 notoamide B,7b and VM55599.10

Malbrancheamide (1)11 and malbrancheamide B (2) wererecently isolated from Malbranchea aurantiaca RRC1813, a† Colorado State University.

‡ The University of Michigan.§ University of Colorado Cancer Center.(1) (a) Williams, R. M.; Cox, R. J. Acc. Chem. Res. 2003, 36, 127. (b)

Williams, R. M. Chem. Pharm. Bull. 2002, 50, 711. (c) Williams, R. M.;Stocking, E. M.; Sanz-Cevera, J. F. Top. Curr. Chem. 2000, 209, 98.

(2) (a) Yamazaki, M.; Okuyama, E. Tetrahedron Lett. 1981, 22, 135.(b) Ondeyka, J. G.; Goegelman, R. T.; Schaeffer, J. M.; Kelemen, L.; Zitano,L. J. Antibiot. 1990, 43, 1375. (c) Liesch, J. M.; Wichmann, C. F. J. Antibiot.1990, 43, 1380. (d) Banks, R. M.; Blanchflower, S. E.; Everett, J. R.; Manfer,B. R.; Reading, C. J. J. Antibiot. 1997, 50, 840.

(3) (a) Birch, A. J.; Wright, J. J. J. Chem. Soc. Chem. Commun. 1969,644. (b) Birch, A. J.; Wright, J. J. Tetrahedron 1970, 26, 2329. (c) Birch,A. J.; Russell, R. A. Tetrahedron 1972, 28, 2999.

(4) Kato, H.; Yoshida, T.; Tokue, T.; Nojiri, Y.; Hirota, H.; Ohta, T.;Williams, R. M.; Tsukamoto, S. Angew. Chem., Int. Ed. 2007, 46, 2254.

(5) (a) Qian-Cutrone, J.; Huang, S.; Shu, Y.-Z.; Vyas, D.; Fairchild, C.;Menendez, A.; Krampitz, K.; Dalterio, R.; Klohr, S. E.; Gao, Q. J. Am.Chem. Soc. 2002, 124, 14556. (b) Qian-Cutrone, J.; Krampitz, K.; Shu,Y.-Z.; Chang, L. P. U.S. Patent 6,291,461, 2001.

(6) (a) Porter, A. E. A.; Sammes, P. G. J. Chem. Soc. Chem. Commun.1970, 1103. (b) Baldas, J.; Birch, A. J.; Russell, R. A. J. Chem. Soc., PerkinTrans I 1974, 50.

(7) (a) Greshock, T. J.; Williams, R. W. Org. Lett. 2007, 9, 4255. (b)Greshock, T. J.; Grubbs, A. W; Tsukamoto, S.; Williams, R. W. Angew.Chem., Int. Ed. 2007, 46, 2262.

(8) (a) Williams, R. M.; Sanz-Cervera, J. F.; Sancenon, F.; Marco, J.A.; Halligan, K. J. Am. Chem. Soc. 1998, 120, 1090. (b) Williams, R. M.;Sanz-Cervera, J. F.; Sancenon, F.; Marco, J. A.; Halligan, K. Bioorg. Med.Chem. 1998, 6, 1233. (c) Sanz-Cervera, J. F.; Williams, R. M.; Marco, J.A.; Lopez-Sanchez, J. M.; Gonzalez, F.; Martınez, M. E.; Sancenon, F.Tetrahedron 2000, 56, 6345. (d) Adams, L. A.; Valente, M. W. N.; Williams,R. M. Tetrahedron 2006, 62, 5195.

(9) Greshock, T. J.; Grubbs, A. W.; Williams, R. M. Tetrahedron 2007,63, 6124.

(10) (a) Stocking, E. M.; Sanz-Cervera, J. F.; Williams, R. M. J. Am.Chem. Soc. 2000, 122, 1675. (b) Sanz-Cervera, J. F.; Williams, R. M. J.Am. Chem. Soc. 2002, 124, 2556.

3116 J. Org. Chem. 2008, 73, 3116-311910.1021/jo800116y CCC: $40.75 © 2008 American Chemical Society

Published on Web 03/18/2008

378

fungus collected on bat detritus collected in a cave in Mexicoby Mata and co-workers. These new substances are the firstalkaloids in this class of prenylated indole alkaloids to containa halogenated indole ring (Figure 1). The lack of a tertiary amidein the bicyclo[2.2.2]diazaoctane core also serves to characterizethe malbrancheamides. In addition to these notable structuralfeatures, malbrancheamide has been shown to be a calmodulin(CaM) antagonist that inhibits the activity of CaM-dependentphosphodiesterase (PDE1) in a concentration dependent man-ner.11 The chemotherapeutic potential of PDE1 inhibitorsincludes applications in the treatment of neurodegenerativediseases, cancers, and vascular diseases, due to the effect onintracellular cAMP and cGMP concentrations.12 New pharma-cological properties of malbrancheamide may be discoveredthrough the study of malbrancheamide, malbrancheamide B, andother analogs, as specific PDE1 inhibitors are scarce and theexact function of the enzyme has not been fully characterized.

Although compelling spectroscopic evidence indicated thatthe structure of 1 was as shown,11 the precise structure ofmalbrancheamide B (2) was less certain. Isolation and structuralcharacterization of 2 indicated the presence of a single chlorineon the indole ring, and further, preliminary biosynthetic experi-ments indicated that malbrancheamide B (2) is a putativebiosynthetic precursor to 1,13 which is thought to arise bysequential halogenation events. However, the question as towhether malbrancheamide B (2) was constituted as the 5-chloroor the 6-chloro derivative was unclear from the preliminarycharacterization data due to the scarce supply of the naturalmaterial. With these issues at the forefront, we undertook thesynthesis of both natural substances to determine the exactidentity of malbrancheamide B. To this end, we envisioned thatmalbrancheamide (1) and malbrancheamide B (2) would arisefrom the aforementioned hetero-IMDA of the 5-hydroxypyrazin-2(1H)-one 3, which we could access by enolization andtautomerization of the enamide 4 (Scheme 1). Using chemistrypreviously established in our laboratory,7a we planned onassembling the enamide 4 from a reverse prenylated tryptophan5, which could be obtained in a few steps from the correspond-ing chlorinated indole 6.

Results and Discussion

Installation of the reverse prenyl group at the indole 2-positionwas carried out using a two-step protocol developed byDanishefsky and co-workers.14 Chlorination at the 3-position

of indoles 6a-c15 using NCS in DMF gave 7a-c (Scheme 2),which were treated with prenyl-9-BBN in the presence of Et3Nto afford the reverse prenylated indoles 8a-c.14 The corre-sponding gramines 9a-c were prepared by treating 8a-c withformaldehyde and dimethylamine, and subsequent Somei-Kametani coupling16 and imine hydrolysis gave the tryptophanderivatives 5a-c in good yields.

The free amine moieties in 5a-c were protected as thecorresponding BOC-carbamates followed by ester hydrolysisunder standard conditions to yield acids 10a-c (Scheme 3).Coupling of cis-3-hydroxyproline ethyl ester with the tryptophanderivatives 10a-c in the presence of HATU delivered theamides 11a-c as inseparable mixtures of diastereomers. Treat-ment of 11a-c with TFA led to carbamate deprotection andthe resulting amino esters were immediately cyclized to thecorresponding diketopiperazines 12a-c after refluxing with2-hydroxypyridine. Dehydration of 12a-c under Mitsunobuconditions gave the enamides 4a-c, which would serve as therespective IMDA substrates.17

Treatment of the enamides 4a-c with aqueous KOH inMeOH gave intermediate hydroxy-azadienes by enolization andtautomerizaion, and subsequent IMDA gave mixtures of 13a-cand 14a-c favoring the desired syn-isomers 13a-c in ratios of(2-1.6):1 (Scheme 4). The observed preference for the IMDAto provide the syn-isomers 13a-c as the major products mirrors

(11) (a) Martinez-Luis, S.; Rodriguez, R.; Acevedo, L.; Gonzalez, M.C.; Lira-Rocha, A.; Mata, R. Tetrahedron 2006, 62, 1817. (b) Figueroa,M.; del Carmen Gonzalez, M.; Mata, R. Unpublished results.

(12) (a) Zhu, H. J.; Wang, J. S.; Guo, Q. L.; Jiang, Y.; Liu, G. Q. Biol.Pharm. Bull. 2005, 28, 1974. (b) Leisner, T. M.; Liu, M. J.; Jaffer, Z. M.;Chernoff, J.; Parise, L. V. J. Cell Biol. 2005, 170, 465.

(13) Ding, Y.; Sherman, D. H.; Williams, R. M. Unpublished results.(14) Schkeryantz, J. M.; Woo, J. C. G.; Siliphaivanh, P.; Depew, K. M.;

Danishefsky, S. J. J. Am. Chem. Soc. 1999, 121, 11964.

(15) For the preparation of 5,6-dichloroindole see: Bromidge, S. M.; etal. J. Med. Chem. 1998, 41, 1598.

(16) (a) Somei, M.; Karasawa, Y.; Kaneko, C. Heterocycles 1981, 16,941. (b) Kametani, T.; Kanaya, N.; Ihara, M. J. Chem. Soc., Perkin Trans.1 1981, 959.

(17) Curiously, attempts to effect the one-step dehydration/IMDA reactionsequence from 12a-c directly to 13a-c + 14a-c failed under the sameconditions used successfully for stephacidin A (ref 7a) and marcfortine C(ref 9).

FIGURE 1. Malbrancheamide and malbrancheamide B.

SCHEME 1. Retrosynthetic Plan

SCHEME 2. Reverse Prenylated Tryptophan Derivatives

Synthesis of Malbrancheamide and Malbrancheamide B

J. Org. Chem, Vol. 73, No. 8, 2008 3117

379

the preference we have noted for this cycloaddition in thepast.7-9 Considering that previous optimization efforts toimprove the syn:anti ratio in related IMDAs were not productivewhen a variety of solvents and temperatures were studied, weelected to simply separate the major isomers 13a-c and continuethe total syntheses without further optimization.7-9

Completion of the syntheses required selective reduction ofthe tertiary amide in the presence of the secondary amide, andto that end, 13a-c were treated with excess DIBAL-H,18 whichcleanly provided malbrancheamide (1) from 12a (80%) andmalbrancheamide B (2) from 12c (74%) (Scheme 5). Synthetic1 was identical in all respects (1H, 13C, HRMS) to the naturalproduct.11 Comparison of the 1H NMR spectrum of 15, the5-chloro derivative, to that of natural malbrancheamide Brevealed significant differences in the aromatic region revealingthat the correct structure contained a 6-chloroindole ring.

Gratifyingly, synthetic 2 was identical in all respects (1H,HRMS) to natural malbrancheamide B.

It is striking that the initial halogenation of the indole ringduring the biosynthesis of malbrancheamide B, occurs at theless-activated 6-position as opposed to the more electron-rich5-position. Studies to clone and express the putative halogenasefrom M. aurantiaca are currently under investigation in theselaboratories.

In summary, the first total synthesis of malbrancheamide (1)and malbrancheamide B (2) have been completed in twelve stepsin 5.3% and 8.2% overall yield, respectively. In addition, thestructure of malbrancheamide B (2) was confirmed though thesynthesis of both the 5-chloro and the 6-chloro regioisomers.Experiments to establish the biosynthetic relationship between1 and 2 and their putative progenitors are in progress and willbe reported in due course.

Experimental Section

Representative Procedure for the Hetero-Diels-Alder Reac-tion of Enamides 4. Preparation of Cycloadducts 13a and 14a.To a solution of 4a (86 mg, 0.21 mmol) in MeOH (16 mL) at 0 °Cwas added 20% aqueous KOH (4 mL). The reaction was warmedto room temperature (rt) and was stirred for 12 h. The reactionwas quenched with sat. NH4Cl (30 mL) and extracted with CH2-Cl2 (3 ! 30 mL). The combined organic layers were dried (Na2-SO4) and concentrated under reduced pressure. The residue wastriturated with CHCl3 (30 mL), and the suspension was filtered toprovide 53 mg (60%) of 13a as a white amorphous solid.Concentration of the filtrate gave 25 mg (29%) of 14a as a whiteamorphous solid. Data for major isomer 13a: 1H NMR (300 MHz,CD3OD) ! 7.55 (s, 1 H), 7.40 (s, 1 H), 3.57 (d, J ) 15.5 Hz, 1 H),3.47 (m, 1 H), 2.74 (d, J ) 15.5 Hz, 1 H), 2.60 (m, 1 H), 2.20-1.96 (comp, 6 H), 1.35 (s, 3 H), 1.11 (s, 3 H); 13C NMR (75 MHz,CD3OD) ! 175.7, 171.3, 144.3, 137.2, 128.1, 125.4, 123.3, 119.8,113.1, 104.8, 68.3, 61.6, 50.8, 45.2, 36.2, 31.7, 30.1, 28.6, 25.4,24.9, 22.3; IR (neat) 1663, 1428 cm-1; HRMS (TOF+) calcd forC21H22N3O2Cl2 (M + H) 418.1084, found 418.1084. Data for minorisomer 14a: 1H NMR (300 MHz, CDCl3) ! 9.78 (s, 1 H), 7.49 (s,1 H), 7.33 (s, 1 H), 3.71 (d, J ) 17.8 Hz, 1 H), 3.47 (comp, 2 H),3.25 (s, 1 H), 2.78 (d, J ) 17.8 Hz, 1 H), 2.67 (m, 1 H), 2.18-1.78 (comp, 6 H), 1.24 (s, 3H), 1.16 (s, 3 H); 13C NMR (75 MHz,CD3OD /CDCl3 (1:9)) ! 173.7, 169.6, 142.6, 135.7, 127.1, 125.0,122.9, 119.1, 112.4, 102.9 67.2, 61.5, 45.8, 44.3, 34.8, 32.6, 29.8,29.1, 28.4, 24.5, 23.2; IR (neat) 1676, 1453 cm-1; HRMS (TOF+)calcd for C21H22N3O2Cl2 (M + H) 418.1084, found 418.1079.

Cycloadducts 13b and 14b. Prepared from 4b in to give 43%of 13b as a white amorphous solid and 27% of 14b as a whiteamorphous solid according to the representative procedure describedabove for 13a and 14a. Data for major isomer 13b: 1H NMR (300MHz, CD3OD) ! 7.54 (s, 1 H), 7.44 (s, 1 H), 7.22 (d, J ) 8.6 Hz,1 H), 7.04 (d, J ) 8.6 Hz, 1 H), 3.68 (d, J ) 15.4 Hz, 1 H), 3.55-3.40 (comp, 2 H), 2.77 (d, J ) 15.4 Hz, 1 H), 2.76 (m, 1 H), 2.61(m, 1 H), 2.25-1.92 (comp, 5 H), 1.37 (s, 3 H), 1.12 (s, 3 H); 13CNMR (100 MHz, CD3OD) ! 175.8, 171.4, 143.4, 136.8, 129.2,125.2, 122.1, 118.2, 112.8, 104.5, 68.3, 61.6, 50.8, 45.2, 36.2, 31.7,30.1, 28.7, 25.5, 25.0, 22.4; IR (neat) 1678, 1441 cm-1; HRMS(18) Fukuyama, T.; Liu, G. J. Am. Chem. Soc. 1996, 118, 7426-7427.

SCHEME 3. Enamide Diels-Alder Precursors

SCHEME 4. Hetero-IMDA Reactions

SCHEME 5. Amide Reductions

Miller et al.

3118 J. Org. Chem., Vol. 73, No. 8, 2008

380

(TOF+) calcd for C21H23N3O2Cl (M + H) 384.1473, found384.1460. Data for minor isomer 14b: 1H NMR (300 MHz, CD3-OD) ! 7.91 (s, 1 H), 7.41 (d, J ) 2.0 Hz, 1 H), 7.23 (d, J ) 8.6Hz, 1 H), 7.01 (dd, J ) 8.6, 2.0 Hz, 1H), 3.71 (d, J ) 17.6 Hz,1H), 3.53 (comp, 2 H), 2.90 (d, J ) 17.6 Hz, 1 H), 2.70 (m, 1 H),2.23-1.89 (comp, 6 H), 1.34 (s, 3 H), 1.27 (s, 3 H); 13C NMR(100 MHz, CD3OD) ! 175.4, 171.7, 143.4, 136.8, 129.7, 125.3,122.1, 118.1, 112.8, 103.8, 68.6, 62.7, 47.3, 45.2, 35.9, 33.3, 30.8,29.9, 28.7, 25.4, 24.8, 23.8; IR (neat) 1675, 1461 cm-1; HRMS(TOF+) calcd for C21H23N3O2Cl (M + H) 384.1473, found384.1468.

Cycloadducts 13c and 14c. Prepared from 4c in to give 55%of 13c as a white amorphous solid and 31% of 14c as a whiteamorphous solid according to the representative procedure describedabove for 13a and 14a; data for major isomer 13c: 1H NMR (400MHz, DMSO-d6) ! 8.75 (s, 1 H), 7.40-6.98 (comp, 3 H), 3.42 (d,J ) 15.3 Hz, 1 H), 3.30 (comp, 2 H), 2.68 (d, J ) 15.3 Hz, 1 H),2.50 (comp, 1 H), 2.10-1.70 (comp, 6 H), 1.27 (s, 3 H), 0.99 (s,3 H); 13C NMR (100 MHz, DMSO-d6) ! 173.0, 168.4, 142.0, 136.8,125.3, 125.2, 118.9, 118.5, 110.4, 103.8, 66.0, 59.6, 48.9, 43.6,34.6, 30.0, 28.7, 27.9, 24.0, 23.7, 21.6; IR (neat) 1671, 1409 cm-1;HRMS (TOF+) calcd for C21H23N3O2Cl (M+H) 384.1473, found384.1470. Data for minor isomer 14c: 1H NMR (400 MHz, CD3-OD/CDCl3 (1:9)) ! 9.58 (s, 1 H), 7.33 (d, J ) 8.4 Hz, 1 H), 7.23(d, J ) 1.8 Hz, 1 H), 6.97 (dd, J ) 8.4, 1.8 Hz, 1 H), 3.75 (d, J )17.8 Hz, 1 H), 3.47 (comp, 2 H), 3.30 (bs, 1 H), 2.84 (d, J ) 17.8Hz, 1 H), 2.70 (m, 1 H), 2.23-1.77 (comp, 6 H), 1.25 (s, 3 H),1.18 (s, 3 H); 13C NMR (100 MHz, DMSO-d6) ! 172.43, 169.0,142.0, 136.8, 125.8, 125.3, 119.0, 118.5, 110.4, 103.1, 79.2, 66.4,60.5, 45.4, 43.7, 34.2, 31.6, 28.6, 27.7, 24.0, 22.5; IR (neat) 1672,1410 cm-1; HRMS (TOF+) calcd for C21H23N3O2Cl (M + H)384.1473, found 384.1468.

Representative Procedure for the Selective Reduction ofTertiary Amides with Excess DIBAL-H. Synthesis of Mal-brancheamide (1). DIBAL-H (0.70 mL, 1 M in toluene, 0.70mmol) was added to a suspension of 13a (15 mg, 0.036 mmol) intoluene (7 mL) at rt. The reaction was stirred at rt for 12 h,whereupon finely powdered Na2SO4‚10H2O was added untilbubbling ceased. The mixture was filtered with a medium porosityfritted funnel washing with EtOAc (50 mL) and MeOH (50 mL),and the filtrate was concentrated under reduced pressure. Theresidue was purified by flash chromatography eluting with MeOH/CH2Cl2 (2:98) to give 12 mg (80%) of 1 as a white amorphoussolid: 1H NMR (400 MHz, CD3OD) ! 7.48 (s, 1 H), 7.40 (s, 1 H),3.43 (d, J ) 10.3 Hz, 1 H), 3.06 (m, 1 H), 2.85 (comp, 2 H), 2.54(m, 1 H), 2.27 (dd, J ) 10.2, 1.5 Hz, 1 H), 2.20-1.18 (comp, 6H), 1.43 (s, 3 H), 1.34 (s, 3 H); 13C NMR (100 MHz, CD3OD) !176.6, 145.1, 137.3, 128.2, 125.3, 123.2, 119.6, 113.1, 104.7, 66.1,59.4, 57.4, 55.4, 48.5, 35.5, 32.4, 30.6, 30.0, 28.1, 24.2, 23.5; IR

(neat) 3226, 1658, 1460 cm-1; HRMS (TOF+) calcd for C21H24N3-OCl2 (M + H) 404.1291, found 404.1290.

Isomalbrancheamide B (15). Prepared from 13b in 68% yieldas a white amorphous solid according to the representativeprocedure described above for 1: 1H NMR (400 MHz, DMSO-d6)! 8.39 (s, 1 H), 7.33 (s, 1 H), 7.26 (d, J ) 8.5 Hz, 1 H), 7.01 (d,J ) 8.5 Hz, 1 H), 3.33 (d, J ) 7.2 Hz, 1 H), 3.26 (d, J ) 9.9 Hz,1 H), 2.76 (s, 2 H), 2.42 (m, 1 H), 2.15-1.73 (comp, 7 H), 1.33 (s,3 H), 1.27 (s, 3 H); 13C NMR (100 MHz, DMSO-d6) ! 173.1. 143.4,134.9, 127.7, 122.8, 120.4, 116.7, 112.1, 103.4, 64.1, 58.6, 55.3,53.9, 47.0, 34.0, 31.1, 30.0, 28.7, 26.6, 23.7, 22.5; IR (neat) 3311,1637, 1458 cm-1; HRMS (TOF+) calcd for C21H25N3OCl (M +H) 370.1680, found 370.1675.

Malbrancheamide B (2). Prepared from 13c in 74% yield as awhite amorphous solid according to the representative proceduredescribed above for 1: 1H NMR (400 MHz, DMSO-d6) ! 8.41 (s,1 H), 7.32 (d, J ) 8.4 Hz, 1 H), 7.27 (d, J ) 1.7 Hz, 1 H), 6.95(dd, J ) 8.4, 1.7 Hz, 1 H), 3.36 (s, 1 H), 3.27 (d, J ) 10.0 Hz, 1H), 2.95 (m, 1 H), 2.76 (s, 2 H), 2.43 (m, 1 H), 2.13 (d, J ) 9.9Hz, 1 H), 2.10-1.70 (comp, 6 H), 1.32 (s, 3 H), 1.26 (s, 3 H); 13CNMR (100 MHz, DMSO-d6) ! 173.1, 142.6, 136.8, 125.3, 125.2,118.7, 118.5, 110.3, 103.7, 64.1, 58.5, 55.3, 53.9, 47.0, 34.0, 31.1,30.0, 28.7, 26.6, 23.7, 22.5; IR (neat) 3297, 1652, 1457 cm-1;HRMS (TOF+) calcd for C21H25N3OCl (M + H) 370.1680, found370.1670.

Acknowledgment. Financial support from the NationalInstitutes of Health (CA70375 to R.M.W) and the Hans W.Vahlteich Professorship (to D.H.S.) is gratefully acknowledged.We are grateful to Prof. Rachel Mata and Dr. Marıa del CarmenGonzalez of the Departamento de Farmacia, Facultad deQuımica, Universidad Nacional Autonoma de Mexico forproviding 1H NMR spectra of natural malbrancheamide andmalbrancheamide B as well as an authentic specimen ofmalbrancheamide. We also acknowledge Prof. Mata for sharinga pre-print of their manuscript describing the structural elucida-tion of malbrancheamide B (ref 11b). We thank Dr. AnthonyE. Glenn of the USDA for providing M. aurantiaca RRC1813strain (originally obtained from Dr. Marıa del Carmen Gonza-lez). Mass spectra were obtained on instruments supported bythe NIH Shared Instrumentation Grant GM49631.

Supporting Information Available: Spectroscopic data andexperimental details for the preparation of all new compounds aswell as copies of 1H NMR and 13C NMR spectra. This material isavailable free of charge via the Internet at http://pubs.acs.org.

JO800116Y

Synthesis of Malbrancheamide and Malbrancheamide B

J. Org. Chem, Vol. 73, No. 8, 2008 3119

381

Principal Investigator: Welch, Timothy R. 6.4

Postdoctoral Fellowship Application 2012

Plans for Work Under Fellowship

B. Research Plan: Targeting STAT5 in Prostate Cancer Cells with DNA Binding Polyamides

I. Specific Aims Prostate cancer was responsible for approximately 33720 deaths in 2011.1 While patients with metastatic prostate cancer typically respond to surgical or medicinal androgen deprivation initially, all will eventually progress to an androgen independent state termed castration-resistant prostate cancer (CRPC). The progression of prostate cancer cells to the androgen independent phenotype occurs concomitantly with an increase in the constitutive activation of the transcription factors androgen receptor (AR) and signal transducer and activator of transcription 5 (STAT5).2 AR is established as a relevant target for the treatment of CRPC, and several therapies targeting the AR signaling pathway are available or in development.3,4 While the exact role of STAT5 in CRPC is not as well defined, it appears to be equally important to prostate cancer cell growth and survival, as evidenced by the following: (1) STAT5 is overactive in 95% of CRPCs,5 (2) STAT5 activity is associated with highly aggressive (high Gleason grade) prostate cancers,6,7 and (3) inhibition of STAT5 by siRNA results in massive apoptotic cell death in human prostate cancer cell lines.8 STAT5 represents a promising target for the treatment of CRPC, a disease for which no effective therapy currently exists. To date, no small molecule inhibitors of STAT5 transcriptional activity have been reported. Our long term goal is to develop small molecule inhibitors of STAT5 function for the treatment of malignancies displaying aberrant STAT5 activity. The overall objective of this proposal is to determine the therapeutic potential of DNA binding pyrrole-imidazole polyamides that target STAT5 for the treatment of CRPC. We hypothesize that polyamides can be designed to downregulate the expression of a subset of genes required for CRPC cell survival. Polyamides containing N-methylpyrrole (Py) and N-methylimidazole (Im) comprise the only known class of programmable small molecules capable of binding specific DNA sequences.9,10 Previous studies from the Dervan group have shown that polyamides are cell permeable,11-13 access nuclear chromatin,14-16 inhibit specific protein–DNA interfaces,17 and modulate gene expression in cell culture.18-23 Polyamides represent a promising approach toward the pursuit of small molecule inhibitors of STAT5 transcription for the treatment of CRPC. The specific aims proposed below will determine the suitability of this approach while contributing to the overall understanding of STAT5 signaling in prostate cancer.

AIM 1. Synthesize a series of Py/Im polyamides designed to target the STAT5 response element and evaluate their DNA binding interactions in vitro. Polyamides designed to target the consensus STAT5 binding sequence 5’-TTCNNNGAA-3’† will be synthesized by solid phase methodology. DNA binding interactions of the polyamides will be evaluated in vitro using melting temperature analysis and electrophoretic mobility shift assay (EMSA).

AIM 2. Examine nuclear localization and characterize gene regulatory potency of polyamides.

Laser-scanning confocal microscopy will be used to examine cellular uptake and localization of high-affinity polyamides in several cancer cell lines. The expression of STAT5 regulated oncogenes BCL-2,24-27 BCL-XL,28,29 and CYCLIN-D12,8,30 in prostate cancer cells will be examined following polyamide treatment using quantitative real-time RT-PCR (qRT-PCR). The global effects of polyamides that are shown to modify gene expression will then be studied

† Abbreviations for degenerate nucleotides: N = any base; W = A or T.

382

APPENDIX II Research Proposal

Principal Investigator: Welch, Timothy R. 6.5

Postdoctoral Fellowship Application 2012

with chromatin immunoprecipitation coupled with sequencing (ChIP-seq) and mRNA sequencing (RNA-seq).

AIM 3. Investigate polyamide induced apoptosis in prostate cancer cell lines and in mice

bearing xenograft tumors. General cell death will be analyzed following polyamide treatment in both normal and prostate cancer cell lines. Polyamide effects on C4-2 tumor growth will be studied in a murine model at the Caltech Animal Facility.

The aims in this proposal represent a new approach toward the development of inhibitors of STAT5 regulated gene expression. The proposed experiments will provide thorough and systematic understanding of the activity, mechanism, and specificity of polyamides in prostate cancer. If successful, the proposed research could produce the first known specific inhibitors of STAT5 regulated gene expression for the treatment of CRPC. II. Background and Significance A limited set of transcription factors has been implicated in a large number of diverse oncogenic signaling pathways.31 Selective inhibition of a single transcription factor would thus influence the expression of numerous upstream oncogenes, many of which are currently individually targeted by discrete inhibitors. Signal transducer and activator of transcription (STAT) proteins are overactive in a variety of cancers, promote cancer cell survival and proliferation, and are susceptible to inhibition, making them ideal targets for cancer therapy.32-34 STATs are latent cytoplasmic proteins that, once activated by tyrosine kinase phosphorylation, dimerize, traffic to the nucleus, and bind specific response elements of target gene promoters (Figure 1).32,35-37 In normal cells, STAT activation has a role in cell survival, angiogenesis, immune function, and in controlling cell growth.38,39 However, disregulation of this activity can contribute to tumor cell growth and proliferation.32,40 Persistent activity of STAT5, a member of the STAT family of proteins, is associated with prostate cancer,5,6,8,41,42 lung, head, and neck cancers,43 and several types of leukemia44-47 and lymphomas.35,48 The link to prostate cancer is particularly strong, with 95% of castration-resistant prostate cancers (CRPC) displaying constitutive STAT5 activity.5 Active STAT5 in prostate cancer predicts early disease recurrence and is associated with high Gleason grade prostate cancers characterized by an aggressive phenotype and poor patient outcome.6,7 Furthermore, active STAT5 has been shown to increase the motility of prostate cancer cells, enhancing the intrinsic ability of the disease to metastasize.49 STAT5 is a generic term for two highly homologous protein isoforms, 94-kDa STAT5a and 92-kDa STAT5b.50 Both isoforms share a conserved DNA binding domain that permits binding of STAT5 to consensus motifs (5’-TTCNNNGAA-3’) found in target gene promoters, including the oncogenes encoding anti-apoptotic (BCL-XL, BCL-2) and pro-proliferative (Cyclin-D1) proteins.2,8,24-30,51 STAT5 binding to this consensus motif

Figure 1. Canonical STAT5 signaling pathway.

383

Principal Investigator: Welch, Timothy R. 6.6

Postdoctoral Fellowship Application 2012

found at non-canonical recognition sites has additionally been shown to activate several novel and potentially oncogenic genes, including TNFRSF13b, MKP-1, and C3ar1.52 Numerous studies have shown that inhibition of STAT5 results in massive apoptotic cell death in human prostate cancer cell lines, regardless of the inhibitory method employed (adenoviral expression of dominant-negative mutants of STAT5, siRNA, or antisense oligonucleotides).2,8,41,53 STAT5 inhibition by antisense oligonucleotides has specifically been shown to suppress the expression of Cyclin-D1 and BCL-XL in LNCaP, C4-2, and DU145 prostate cancer cell lines at both the mRNA and protein levels, resulting in increased apoptotic death and decreased cell growth.2,8 In mouse xenograft models, the progression of CRPC was delayed significantly by STAT5 inhibition with antisense oligonucleotides.2 These findings imply prostate cancers depend on persistent STAT5 activation for cell growth and survival. Chemotherapeutic small molecules targeting STAT5 signaling could thus have a substantial impact on the treatment of CRPC, a disease for which no effective pharmacological therapy currently exists. STAT5 inhibitors published to date indiscriminately downregulate STAT5 regulated transcription and lack practical therapeutic potential. Herein, we describe a new, promising approach for the selective inhibition of STAT5 mediated gene expression in prostate cancer, using DNA binding polyamides to disrupt the STAT5–DNA interface. Polyamides containing N-methylpyrrole (Py) and N-methylimidazole (Im) comprise a class of programmable small molecules that can be designed to bind specific DNA sequences with affinities

Figure 2. Molecular recognition of the minor groove of DNA by a hairpin polyamide. (A) Model of an eight ring hairpin polyamide (ImImPyPy-(R)H2N-!-PyPyPyPy-"-IPA) bound to a 5’-TGGAAA-3’ sequence. Putative hydrogen bonds are represented by dashed lines. (B) X-ray crystal structure of a cyclic polyamide (blue) in complex with dsDNA (0.95Å resolution). (C) Ball-and-stick model of the polyamide in 2A. Closed and open circles represent N-methylimidazole and N-methylpyrrole monomers, respectively. Diamonds, "-alanine; triangle with positive charge, triamine linker; curved line substituted with positive charge, #-amino-substituted !-turn. The isophthalic acid derived tail is represented by a hexagon.

!

NN

O

N H

NN

NO H

N

O NH

N NO

H

5' 3'

T

G

A

A

A

G

A

T

T

T

C

C

N

ONH

N

N OH

N

ONH

N

N O

H

NH

O

NH3

H

H

NH

NH

IPA + +

W G G W W W

W C C W W W

5' -

3' -

- 3'

- 5'

A B

C

!-turntargetsW"W

Py/PytargetsW"W

Im/Pytargets

G"C

#-alatargetsW"W

O

-O

O

384

Principal Investigator: Welch, Timothy R. 6.7

Postdoctoral Fellowship Application 2012

comparable to native DNA binding proteins.9 Sequence specificity of Py/Im polyamides is determined by side-by-side pairing of the heterocyclic amino acids to form distinct hydrogen bonds to nucleotides in the minor groove of DNA (Figure 2).54 Im/Py pairs distinguish G!C from C!G, whereas Py/Py pairs degenerately bind A!T and T!A.10 Hairpin polyamides designed as such have been shown to bind DNA with high affinities,55 inhibit interactions with DNA binding proteins,17 bind chromatin,14-16 and traffic to the nucleus in a variety of cell types.11-13 Recently, analyses have shown that 8-ring cyclic polyamides allosterically perturb the DNA helix through a 4Å widening of the minor groove and consequent major groove compression.56 This significant change provides a mechanistic basis for disruption of protein–DNA interfaces by hairpin polyamides. To evaluate the specific inhibition of STAT5 mediated gene expression by cell permeable, sequence specific DNA binding polyamides, we propose the synthesis, biochemical evaluation, and biological study of a small library of Py/Im polyamides designed to specifically disrupt the interactions of STAT5 with regulatory regions of pro-proliferative and anti-apoptotic genes. III. Preliminary Studies This proposal builds upon known chemical and biological techniques developed in the Dervan group. The study highlighted here shows precedence for the use of polyamides in the modulation of endogenous gene expression (Figure 3). In the study, hypoxia inducible factor (HIF-1#) binding to the sequence 5’-WWWCGW-3’ found in the vascular endothelial growth factor (VEGF) hypoxia response element (HRE) was perturbed by a hairpin polyamide.21,22 The polyamide reduced occupancy of HIF-1# at the VEGF HRE and suppressed VEGF mRNA expression. Only a subset of genes induced by deferoxamine (DFO)—a small molecule used to mimic hypoxia in cells—was inhibited by polyamide, whereas HIF-1# siRNA and the DNA-binding natural product echinomycin inhibited most of the DFO induced transcripts. This demonstrated the relative selectivity of Py/Im polyamides compared to other methods used to perturb HIF-1# activity. A second investigation key to the design of this proposal was detailed in a series of publications from the Dervan group in which the nuclear localization of a library of over 120 polyamide-fluorophore conjugates was described.11-13 Cellular uptake profiles of the polyamides were assayed in 13 cell lines using confocal microscopy. The reported nuclear uptake trends suggest the following structural features to be conducive to uptake: an eight-ring hairpin or cyclic core, a positive charge on the amino substituent of the !-aminobutyric acid (GABA) turn, and either a conjugated fluorescein fluorophore (FITC) or isophthalic acid (IPA) moiety on the hairpin tail. Polyamides targeting 5’-WGWWGW-3’ and 5’-WGGWWW-3’ similar to those proposed below were shown to have strong nuclear localization in PC-3 and LNCaP prostate cancer cell lines.11,12 The transition from cell culture work to murine models is an important next step in Py/Im polyamide research that is focused on biomedical applications of the technology. Recent studies undertaken in the Dervan laboratory have provided basic pharmacokinetic analyses of polyamides in C57/BL6 wild type mice.57 Micromolar blood levels of Py/Im polyamides were achieved for multiple hours following a single injection of the compound subcutaneously or intraperitoneally. An example is shown in Figure 4a (a representative HPLC trace) and 4B (a representative pharmacokinetic profile for a hairpin Py/Im polyamide). Decline in hairpin polyamide concentration was observed over the course of multiple hours,

Figure 3. Polyamide mediated inhibition of HIF-1 DNA binding to the HRE suppresses VEGF expression. (The square symbol represents chlorothiophene.)

! !" HIF-1

IPA + +

G C A T A C G T G G5' -

3' -

- 3'

- 5'C G T A T G C A C C

HRE

VEGFtranscription

XX

385

Principal Investigator: Welch, Timothy R. 6.8

Postdoctoral Fellowship Application 2012

with a peak concentration noted between 1-3h depending on route of administration. Compound plasma levels were below detection level 24h after the injection. The polyamide was well tolerated by the animals. Multiple administrations (i.p. or s.c. at 120 nmol per injection per animal) did not result in any significant reduction in body weight. Weight loss is a commonly used criterion for health deterioration in mice (Figure 4C, D).57 The Dervan group has recently gained broad experience with xenograft experimentation in immunocompromised mice, encompassing the engraftment of human cancer cell lines A549, LNCaP, U251, and T47D. FITC-labeled polyamides injected subcutaneously on the flank opposite the tumor were shown by confocal microscopy to access the tumor derived cell nuclei. Moreover, gene expression changes have been observed in engrafted A549 tumors following polyamide treatment.58 The studies outlined above demonstrate the validity of targeting the transcription factor–DNA interface as a means of modulating gene expression. Furthermore, evaluation of nuclear uptake trends allows for the rational design of new polyamides that are structurally optimized for nuclear localization. The Dervan group has also recently developed a gram-scale synthesis of hairpin polyamides and shown that polyamides are bioavailable in mice, eliminating two major obstacles in the evaluation of polyamides in animal models.59,60 Tumor engraftment protocols, established tolerable dosing conditions, and general animal care and handling expertise found in the Dervan group will be invaluable during in vivo testing of the proposed STAT5-targeting polyamides. We are confident the methods established by these studies can be extended to the proposed research and will increase the probability of success. IV. Research Design and Methods An estimated timeline for completion of the specific aims is given in Figure 5. The proficiency of the Dervan group with these techniques coupled with the outstanding equipment, facilities, and expertise found at Caltech will provide strong support for the success of this research project. We are confident that the proposed objectives will be thoroughly pursued during the planned three year period.

A B

C D

Figure 4. Pharmacokinetic data for hairpin polyamide 1 in C57/BL6 wild type mice. (A) HPLC analysis of a blood sample collected retro-orbitally 1.5 h post intraperitoneal (i.p.) injection of 1. Plasma was isolated by centrifugation and pre-cleared from protein by methanol precipitation. (4 = internal reference standard). (B) Plasma levels of 1 as a percent of reference standard 4 following i.p. or subcutaneous (s.c.) injection. (C, D) Weight of four individual mice in response to polyamide dosing regimen. 120 nmol doses were administered either i.p. (C) or s.c. (D) at intervals denoted by arrows in the plot. The solid red line indicates the 15% weight loss mark considered an endpoint criterion for the mice.

386

Principal Investigator: Welch, Timothy R. 6.9

Postdoctoral Fellowship Application 2012

AIM 1. Synthesize a series of Py/Im polyamides designed to target the STAT5 response element

and evaluate their DNA binding interactions in vitro. Three oncogenes shown in the literature to be functionally activated by STAT5 and to contain a STAT5 binding site characterized at the sequence level were selected as targets for polyamide inhibition (Table 1). The three degenerate base pairs found in the middle of the consensus STAT5 binding sequence (5’-TTCNNNGAA-3’) represent the great degree of diversity of the binding sequences on target genes, allowing for the design of polyamides with greater DNA site sequence specificity than the native transcription factor. Three polyamides (Figure 6) are proposed to bind the sequences 5’-WGWWGW-3’ (1, found in BCL-XL), 5’-WWCGGW-3’ (2, found in BCL-XL), and 5’-WGGWWW-3’ (3, found in BCL-2 and CYCLIN-D1).27,30 Polyamide 3 is also expected to bind non-canonical STAT5 binding sites found in MKP-1, TNFRSF13b, and C3ar1.52 Mismatch controls will be prepared for each compound by substituting a Py for an Im (structures not shown). Polyamides 1-3 and the three mismatch controls will be synthesized using solid-phase methodology established in the Dervan laboratory.18,61,62 Preparative reverse-phase high performance liquid chromatography (RP-HPLC) will be used to purify crude reaction products. Chemical purity of polyamides will be determined by analytical HPLC and the chemical composition of each confirmed by matrix-assisted laser desorption ionization time-of-flight mass spectrometry (MALDI-TOF).

The ability of synthesized polyamides 1-3 to bind DNA will be determined using melting temperature (Tm) analysis. The degree to which both match and mismatch polyamides stabilize 14 base pair DNA duplexes containing the appropriate match sequence will be measured. Previous results have shown a correlation between the increase in Tm of DNA duplexes and DNA binding affinity.63-65 Polyamides

Gene Function STAT5 Binding Sequence BCL-XL anti-apoptotic 5’-GCATTTCGGAGAAGACG-3’ BCL-2 anti-apoptotic 5’-CAAGTTCCAGGAAAGCG-3’

CYCLIN-D1 pro-proliferative 5’-GGCGTTCTTGGAAATGC-3’

Table 1. Promoter sequences for selected genes demonstrated to be STAT5 dependent. The consensus sequence is specified in bold.

Figure 6. Proposed library of polyamide inhibitors of STAT5. Structural abbreviations are defined in Figure 2.

IPA + +

W G G W W W

W C C W W WIPA + + IPA + +

W W C G G W

W W G C C W

W G W W G W

W C W W C W3'

3'

5'

5'

1 2

3'

3'

5'

5'

3'

3'

5'

5'

3

Found in promoters of: BCL-XL Found in promoters of: BCL-XL Found in promoters of: BCL-2CYCLIN-D1

Figure 5. Predicted timetable for the proposed research.

Synthesis (2 months)

Characterization of DNA binding (6 months)

Confocal microscopy (2 months)

qRT-PCR (6 months)

ChIP-seq and RNA-seq (13 months)

Apoptosis assay

(6 months)

Tumor xenograft studies

(18 months)

Year 1 Year 2 Year 3

387

Principal Investigator: Welch, Timothy R. 6.10

Postdoctoral Fellowship Application 2012

identified to bind DNA with high affinity will be subjected to electrophoretic mobility shift assay (EMSA) to evaluate their ability to disrupt STAT5 binding to their cognate sequences. Published protocols from the Dervan laboratory will be followed using commercially available STAT5 and 32P-labelled oligonucleotides purchased from Abcam and Integrated DNA Technologies, respectively.19-21,63

AIM 2. Examine nuclear localization and characterize gene regulatory potency of polyamides. Fluorescent (FITC) derivatives of polyamides 1-3 will be evaluated in cell culture to examine their uptake and localization properties. Cells treated with varying concentrations of fluorescent polyamides will be examined using confocal microscopy as previously described.11-13 Several prostate cancer cell lines will be studied to establish general cellular uptake trends, including LNCaP, C4-2, DU145, and PC-3 (Table 2). All have been previously used in the Dervan laboratory or are commercially available and will be cultured and propagated according to recommended American Type Culture Collection (ATCC) procedures. Polyamides will then be screened for the ability to suppress gene expression in cells by quantitative real-time RT-PCR (qRT-PCR).22 Specifically, we will examine the mRNA expression profiles of STAT5 regulated genes BCL-XL, BCL-2, and CYCLIN-D1 in the presence of polyamides following induction of STAT5 activity by interleukin 3 (IL-3) in four different prostate cancer cell lines. A reduction in IL-3 induced expression is expected if a polyamide disrupts STAT5 activation of the gene under investigation. Mismatch polyamides will serve as a negative control, while STAT5 siRNA known to inhibit expression of all three target genes will be used as a positive control for comparison.8 Since constitutive STAT5 activation is most prevalent in cells expressing an androgen independent phenotype, cell lines for this experiment were selected based on androgen dependence (Table 2). Polyamide binding is expected to impart a greater change on gene expression in androgen independent cell lines C4-2 and DU145 than on androgen sensitive LNCaP cells. PC-3 cells do not express STAT5 and are included as a negative control for STAT5 regulated gene expression. The global effects of polyamide treatment on prostate cancer cells will be investigated through chromatin-immunoprecipitation coupled with sequencing (ChIP-seq) and cellular mRNA sequencing (RNA-seq). To probe the mechanism of polyamide induced gene suppression in prostate cancer cells, ChIP-seq will be conducted to assess STAT5 occupancy in nuclear chromatin. In the absence of polyamide or in the presence of the mismatched control, amplification of DNA isolated from anti-STAT5 antibody (purchased from Santa Cruz) pull-down is expected, demonstrating normal STAT5–DNA binding. A decrease in pull-down efficiency from polyamide treated cells would support an inhibitory mechanism in which displacement of STAT5 is attributable to polyamide binding to gene promoters. Isolated DNA will be submitted to high throughput sequencing using the Illumina Genome Analyzer at Caltech. Sequencing results mapped to the human genome will be compared between the untreated, mismatch control, and match polyamide experiments to identify specific genomic sites susceptible to polyamide-mediated STAT5 displacement. A consensus polyamide binding site will also be characterized, providing valuable insight as to the sequence specificity of the polyamide. The global effect of polyamides on gene expression can be evaluated using RNA-seq. STAT5 activity will be induced in cells with IL-3, then either (1) left untreated, or treated with (2) mismatch polyamide, (3) polyamide, or (4) STAT5 siRNA. Total mRNA will be isolated from each experiment, sequenced, and scanned for a 2-fold or greater alteration in expression. Our hypothesis that polyamides can be designed to selectively inhibit

Cell Line

Androgen Dependence

STAT5 (+/-)

Commercial source

LNCaP sensitive + ATCC C4-2 independent + UroCor Labs

DU145 independent + ATCC PC-3 independent - ATCC

Table 2. Proposed prostate cancer cell lines.

388

Principal Investigator: Welch, Timothy R. 6.11

Postdoctoral Fellowship Application 2012

a subset of STAT5 regulated genes will be supported if fewer IL-3 induced transcripts are affected by polyamide than by siRNA. A sequence specific mechanism of polyamide mediated perturbation is also supported if the mismatch control suppresses fewer induced gene transcripts than the matched polyamide. AIM 3. Investigate polyamide induced apoptosis in prostate cancer cell lines and in mice bearing

xenograft tumors. The effect on cell apoptosis of the proposed polyamides will be evaluated in each of the prostate cancer cell lines mentioned above. Cells will be incubated with varying concentrations of polyamide, then the effect on cell cycle population analyzed by flow cytometry, caspase-3 activity, and cleaved poly ADP ribose polymerase (PARP) expression.2,53 Polyamide induced apoptosis would be characterized by dose-dependent increases in the fraction of cells in the sub-G0/G1 phases of the cell cycle, caspase-3 activity, and in the expression of cleaved PARP. STAT5-negative PC-3 cells will serve as a necessary negative control. The selectivity of polyamide induction of apoptosis in cancerous versus normal cell lines will be determined by comparing these results to those found in an analogous experiment on HUVEC and NHDF cell lines. Polyamide effects on the C4-2 cell line will be further studied in an in vivo murine model. All experiments are to be conducted at the Caltech Animal Facility following the experimental procedures outlined in the IACUC protocol of the Dervan laboratory (#1638-11, date of approval: 9/21/11). Male immunocompromised mice will be injected subcutaneously (s.c.) with up to 107 C4-2 cells in PBS. Following a 1-2 week tumor engraftment period, Py/Im polyamides will be injected intraperitoneally (i.p.) at doses not exceeding 40 mg/kg over the course of up to 12 weeks. The animals will be injected with polyamide up to three times a week with a resting period of at least onea day between injections. A vehicle-treated reference control group will be maintained throughout the experiment. Recent investigations conducted in the Dervan laboratory showed that micromolar concentrations of polyamide in murine blood could be achieved following i.p. injection of polyamides with varying sequences.57,58 Tumor volume will be monitored at least once a week using caliper measurements. Retro-orbital blood samples will be collected weekly and levels of tumor-specific secreted factor PSA measured by enzyme-linked immunosorbent assay (ELISA). After four weeks of polyamide treatment, the mice will be sacrificed by asphyxiation (CO2 chamber, 60%). Tumors will be excised and sectioned for histological blood vessel quantification. RNA-seq experiments will further be conducted to determine global polyamide effects on the tumor transcriptome in comparison to the vehicle treated group. Potential Problems with this Proposal Three possible problems with this proposal are presented and addressed. (i) The proposed polyamides are not sufficiently specific to be considered therapeutic candidates for the

selective inhibition of STAT5 in the context of the human genome. The human genome likely contains over a million potential binding sites for a compound that targets six base pair sequences. However, many of these sites are inaccessible in the context of nuclear chromatin. Boyle and coworkers have shown using DNase I hypersensitivity maps that only 2.1% of the genome is accessible in CD4+ T-cells.66 Moreover, polyamide binding at non-promoter sequences of accessible DNA does not affect transcription, as RNA polymerase dislodges polyamides from DNA during transcription.67 Despite the prevalence of six base pair sequences targeted by 8-ring polyamides in the genome, the HIF-1 studied mentioned above shows that polyamides have a relatively small effect on global gene expression.

389

Principal Investigator: Welch, Timothy R. 6.12

Postdoctoral Fellowship Application 2012

(ii) The STAT family of proteins share a similar DNA binding domain, so polyamides designed to target a six base pair STAT5 sequence will invariably target other members of the family of transcription factors. STAT5 and STAT3 site sequences share the most

homology (5’-TTCNNNGAA-3’ and 5’-TTCCNGGAA-3’, respectively) among the STAT family.68 Polyamides 1 and 2 target STAT consensus sequences with at least two mismatched sites and are expected to be selective for STAT5 over STAT3. Polyamide 3 matches the STAT3 consensus when N=W and will likely inhibit some binding of both STAT3 and STAT5 to the cognate sequence. Comparison of the gene regulatory activity of polyamides 1-3 will allow us to analyze selectivity of the proposed polyamides. Binding specificity could be improved by expanding the Py/Im core to target seven or eight base pair sequences (Figure 7). However, disruption of STAT3 activated gene expression would not necessarily be an undesired consequence of polyamide binding, as constitutive STAT3 activity has also been implicated in the malignant phenotype of numerous cancers.32 For this reason, polyamide 3 was deliberately included in this proposal.

(iii) Inhibition of STAT5 target gene expression is not selective for cancer cells. CRPC tumors are dependent upon persistent STAT5 activity for malignant cell growth and survival. It has been suggested that even partial inhibition of STAT5 mediated transcription would be fatal to tumor cells as a consequence of this increased STAT5 dependence.32 Normal cells could plausibly survive with low levels of active STAT5 or utilize other survival pathways. Indeed, even STAT5 null mice remain viable, despite impaired mammary gland development and abnormalities in hematopoiesis.36,69 Conversely, disruption of STAT5 signaling in tumor cells causes massive apoptosis. Aim 3 will provide a direct comparison of polyamide treatment on normal versus cancerous cells. Closing Remarks The primary aim of this proposal is the development of a small library of polyamides designed to inhibit the binding of STAT5 to its cognate promoter site sequences. We hypothesize that the disruption of STAT5 binding events should suppress gene expression necessary for tumor cell growth and survival, resulting in apoptosis of CRPC cells. Inhibition of STAT5 transcriptional activity at the protein–DNA interface represents a new approach toward the treatment of CRPC. Accordingly, extensive experimentation has been proposed to fully elucidate the binding affinities and specificities of polyamides 1-3 for the cognate DNA sequences, to determine if the polyamides are able to traffic to the nucleus, and to quantify the effect of polyamide treatment on gene expression in prostate cancer cells. The ability of polyamides to reduce the viability of cancer cells will then be examined in cell lines and in mice bearing xenograft tumors. Results of these studies will reveal the therapeutic potential of polyamides in the treatment of CRPC and contribute to the understanding of STAT5 signaling in prostate cancer cells.

Figure 7. Polyamide modifications: extended hairpins targeting 7 bp sequences of CYCLIN-D1 promoter.

IPA + +

IPA + +

G W W C W W G G W W

C W W G W W C C W W

4

3'

3'

5'

5'

W C W W G G W W W W

W G W W C C W W W W

5

3'

3'

5'

5'

390

Principal Investigator: Welch, Timothy R. 6.13

Postdoctoral Fellowship Application 2012

Literature Cited 1. Siegel, R.; Ward, E.; Brawley, O.; Jemal, A. Cancer statistics, 2011: the impact of eliminating

socioeconomic and racial disparities on premature cancer deaths. CA Cancer J Clin 2011, 61, 212-236.

2. Thomas, C.; Zoubeidi, A.; Kuruma, H.; Fazli, L.; Lamoureux, F.; Beraldi, E.; Monia, B. P.; MacLeod, A. R.; Thuroff, J. W.; Gleave, M. E. Transcription factor Stat5 knockdown enhances androgen receptor degradation and delays castration-resistant prostate cancer progression in vivo. Mol Cancer Ther 2011, 10, 347-359.

3. Antonarakis, E. S.; Carducci, M. A.; Eisenberger, M. A. Novel targeted therapeutics for metastatic castration-resistant prostate cancer. Cancer Lett 2010, 291, 1-13.

4. Ryan, C. J.; Tindall, D. J. Androgen receptor rediscovered: the new biology and targeting the androgen receptor therapeutically. J Clin Oncol 2011, 29, 3651-3658.

5. Tan, S. H.; Dagvadorj, A.; Shen, F.; Gu, L.; Liao, Z.; Abdulghani, J.; Zhang, Y.; Gelmann, E. P.; Zellweger, T.; Culig, Z.; Visakorpi, T.; Bubendorf, L.; Kirken, R. A.; Karras, J.; Nevalainen, M. T. Transcription factor Stat5 synergizes with androgen receptor in prostate cancer cells. Cancer Res 2008, 68, 236-248.

6. Li, H.; Ahonen, T. J.; Alanen, K.; Xie, J.; LeBaron, M. J.; Pretlow, T. G.; Ealley, E. L.; Zhang, Y.; Nurmi, M.; Singh, B.; Martikainen, P. M.; Nevalainen, M. T. Activation of signal transducer and activator of transcription 5 in human prostate cancer is associated with high histological grade. Cancer Res 2004, 64, 4774-4782.

7. Li, H.; Zhang, Y.; Glass, A.; Zellweger, T.; Gehan, E.; Bubendorf, L.; Gelmann, E. P.; Nevalainen, M. T. Activation of signal transducer and activator of transcription-5 in prostate cancer predicts early recurrence. Clin Cancer Res 2005, 11, 5863-5868.

8. Dagvadorj, A.; Kirken, R. A.; Leiby, B.; Karras, J.; Nevalainen, M. T. Transcription factor signal transducer and activator of transcription 5 promotes growth of human prostate cancer cells in vivo. Clin Cancer Res 2008, 14, 1317-1324.

9. Dervan, P. B.; Edelson, B. S. Recognition of the DNA minor groove by pyrrole-imidazole polyamides. Curr Opin Struct Biol 2003, 13, 284-299.

10. Trauger, J. W.; Baird, E. E.; Dervan, P. B. Recognition of DNA by designed ligands at subnanomolar concentrations. Nature 1996, 382, 559-561.

11. Best, T. P.; Edelson, B. S.; Nickols, N. G.; Dervan, P. B. Nuclear localization of pyrrole-imidazole polyamide-fluorescein conjugates in cell culture. Proc Natl Acad Sci U S A 2003, 100, 12063-12068.

12. Edelson, B. S.; Best, T. P.; Olenyuk, B.; Nickols, N. G.; Doss, R. M.; Foister, S.; Heckel, A.; Dervan, P. B. Influence of structural variation on nuclear localization of DNA-binding polyamide-fluorophore conjugates. Nucleic Acids Res 2004, 32, 2802-2818.

13. Nickols, N. G.; Jacobs, C. S.; Farkas, M. E.; Dervan, P. B. Improved nuclear localization of DNA-binding polyamides. Nucleic Acids Res 2007, 35, 363-370.

14. Dudouet, B.; Burnett, R.; Dickinson, L. A.; Wood, M. R.; Melander, C.; Belitsky, J. M.; Edelson, B.; Wurtz, N.; Briehn, C.; Dervan, P. B.; Gottesfeld, J. M. Accessibility of nuclear chromatin by DNA binding polyamides. Chem Biol 2003, 10, 859-867.

15. Suto, R. K.; Edayathumangalam, R. S.; White, C. L.; Melander, C.; Gottesfeld, J. M.; Dervan, P. B.; Luger, K. Crystal structures of nucleosome core particles in complex with minor groove DNA-binding ligands. J Mol Biol 2003, 326, 371-380.

16. Edayathumangalam, R. S.; Weyermann, P.; Gottesfeld, J. M.; Dervan, P. B.; Luger, K. Molecular recognition of the nucleosomal "supergroove". Proc Natl Acad Sci U S A 2004, 101, 6864-6869.

17. Gottesfeld, J. M.; Neely, L.; Trauger, J. W.; Baird, E. E.; Dervan, P. B. Regulation of gene expression by small molecules. Nature 1997, 387, 202-205.

391

Principal Investigator: Welch, Timothy R. 6.14

Postdoctoral Fellowship Application 2012

18. Chenoweth, D. M.; Harki, D. A.; Phillips, J. W.; Dose, C.; Dervan, P. B. Cyclic pyrrole-imidazole polyamides targeted to the androgen response element. J Am Chem Soc 2009, 131, 7182-7188.

19. Muzikar, K. A.; Nickols, N. G.; Dervan, P. B. Repression of DNA-binding dependent glucocorticoid receptor-mediated gene expression. Proc Natl Acad Sci U S A 2009, 106, 16598-16603.

20. Nickols, N. G.; Dervan, P. B. Suppression of androgen receptor-mediated gene expression by a sequence-specific DNA-binding polyamide. Proc Natl Acad Sci U S A 2007, 104, 10418-10423.

21. Nickols, N. G.; Jacobs, C. S.; Farkas, M. E.; Dervan, P. B. Modulating hypoxia-inducible transcription by disrupting the HIF-1-DNA interface. ACS Chem Biol 2007, 2, 561-571.

22. Olenyuk, B. Z.; Zhang, G. J.; Klco, J. M.; Nickols, N. G.; Kaelin, W. G., Jr.; Dervan, P. B. Inhibition of vascular endothelial growth factor with a sequence-specific hypoxia response element antagonist. Proc Natl Acad Sci U S A 2004, 101, 16768-16773.

23. Dickinson, L. A.; Burnett, R.; Melander, C.; Edelson, B. S.; Arora, P. S.; Dervan, P. B.; Gottesfeld, J. M. Arresting cancer proliferation by small-molecule gene regulation. Chem Biol 2004, 11, 1583-1594.

24. Buitenhuis, M.; Baltus, B.; Lammers, J. W.; Coffer, P. J.; Koenderman, L. Signal transducer and activator of transcription 5a (STAT5a) is required for eosinophil differentiation of human cord blood-derived CD34+ cells. Blood 2003, 101, 134-142.

25. Wierenga, A. T.; Vellenga, E.; Schuringa, J. J. Maximal STAT5-induced proliferation and self-renewal at intermediate STAT5 activity levels. Mol Cell Biol 2008, 28, 6668-6680.

26. Weber-Nordt, R. M.; Egen, C.; Wehinger, J.; Ludwig, W.; Gouilleux-Gruart, V.; Mertelsmann, R.; Finke, J. Constitutive activation of STAT proteins in primary lymphoid and myeloid leukemia cells and in Epstein-Barr virus (EBV)-related lymphoma cell lines. Blood 1996, 88, 809-816.

27. Li, G.; Miskimen, K. L.; Wang, Z.; Xie, X. Y.; Brenzovich, J.; Ryan, J. J.; Tse, W.; Moriggl, R.; Bunting, K. D. STAT5 requires the N-domain for suppression of miR15/16, induction of bcl-2, and survival signaling in myeloproliferative disease. Blood 2010, 115, 1416-1424.

28. Gesbert, F.; Griffin, J. D. Bcr/Abl activates transcription of the Bcl-X gene through STAT5. Blood 2000, 96, 2269-2276.

29. Horita, M.; Andreu, E. J.; Benito, A.; Arbona, C.; Sanz, C.; Benet, I.; Prosper, F.; Fernandez-Luna, J. L. Blockade of the Bcr-Abl kinase activity induces apoptosis of chronic myelogenous leukemia cells by suppressing signal transducer and activator of transcription 5-dependent expression of Bcl-xL. J Exp Med 2000, 191, 977-984.

30. Ehret, G. B.; Reichenbach, P.; Schindler, U.; Horvath, C. M.; Fritz, S.; Nabholz, M.; Bucher, P. DNA binding specificity of different STAT proteins. Comparison of in vitro specificity with natural target sites. J Biol Chem 2001, 276, 6675-6688.

31. Darnell, J. E. Transcription factors as targets for cancer therapy. Nature Reviews Cancer 2002, 2, 740-749.

32. Yu, H.; Jove, R. The STATs of cancer--new molecular targets come of age. Nat Rev Cancer 2004, 4, 97-105.

33. Frank, D. A. STAT signaling in cancer: insights into pathogenesis and treatment strategies. Cancer Treat Res 2003, 115, 267-291.

34. Turkson, J.; Jove, R. STAT proteins: novel molecular targets for cancer drug discovery. Oncogene 2000, 19, 6613-6626.

35. Ferbeyre, G.; Moriggl, R. The role of Stat5 transcription factors as tumor suppressors or oncogenes. Biochim Biophys Acta 2011, 1815, 104-114.

36. Levy, D. E.; Darnell, J. E., Jr. Stats: transcriptional control and biological impact. Nat Rev Mol Cell Biol 2002, 3, 651-662.

37. Darnell, J. E., Jr. STATs and gene regulation. Science 1997, 277, 1630-1635.

392

Principal Investigator: Welch, Timothy R. 6.15

Postdoctoral Fellowship Application 2012

38. Akira, S. Roles of STAT3 defined by tissue-specific gene targeting. Oncogene 2000, 19, 2607-2611.

39. Nevalainen, M. T.; Ahonen, T. J.; Yamashita, H.; Chandrashekar, V.; Bartke, A.; Grimley, P. M.; Robinson, G. W.; Hennighausen, L.; Rui, H. Epithelial defect in prostates of Stat5a-null mice. Lab Invest 2000, 80, 993-1006.

40. Libermann, T. A.; Zerbini, L. F. Targeting transcription factors for cancer gene therapy. Curr Gene Ther 2006, 6, 17-33.

41. Ahonen, T. J.; Xie, J.; LeBaron, M. J.; Zhu, J.; Nurmi, M.; Alanen, K.; Rui, H.; Nevalainen, M. T. Inhibition of transcription factor Stat5 induces cell death of human prostate cancer cells. J Biol Chem 2003, 278, 27287-27292.

42. Dagvadorj, A.; Collins, S.; Jomain, J. B.; Abdulghani, J.; Karras, J.; Zellweger, T.; Li, H.; Nurmi, M.; Alanen, K.; Mirtti, T.; Visakorpi, T.; Bubendorf, L.; Goffin, V.; Nevalainen, M. T. Autocrine prolactin promotes prostate cancer cell growth via Janus kinase-2-signal transducer and activator of transcription-5a/b signaling pathway. Endocrinology 2007, 148, 3089-3101.

43. Xi, S.; Zhang, Q.; Dyer, K. F.; Lerner, E. C.; Smithgall, T. E.; Gooding, W. E.; Kamens, J.; Grandis, J. R. Src kinases mediate STAT growth pathways in squamous cell carcinoma of the head and neck. J Biol Chem 2003, 278, 31574-31583.

44. Lin, T. S.; Mahajan, S.; Frank, D. A. STAT signaling in the pathogenesis and treatment of leukemias. Oncogene 2000, 19, 2496-2504.

45. Schwaller, J.; Parganas, E.; Wang, D.; Cain, D.; Aster, J. C.; Williams, I. R.; Lee, C. K.; Gerthner, R.; Kitamura, T.; Frantsve, J.; Anastasiadou, E.; Loh, M. L.; Levy, D. E.; Ihle, J. N.; Gilliland, D. G. Stat5 is essential for the myelo- and lymphoproliferative disease induced by TEL/JAK2. Mol Cell 2000, 6, 693-704.

46. Huang, M.; Dorsey, J. F.; Epling-Burnette, P. K.; Nimmanapalli, R.; Landowski, T. H.; Mora, L. B.; Niu, G.; Sinibaldi, D.; Bai, F.; Kraker, A.; Yu, H.; Moscinski, L.; Wei, S.; Djeu, J.; Dalton, W. S.; Bhalla, K.; Loughran, T. P.; Wu, J.; Jove, R. Inhibition of Bcr-Abl kinase activity by PD180970 blocks constitutive activation of Stat5 and growth of CML cells. Oncogene 2002, 21, 8804-8816.

47. Levis, M.; Allebach, J.; Tse, K. F.; Zheng, R.; Baldwin, B. R.; Smith, B. D.; Jones-Bolin, S.; Ruggeri, B.; Dionne, C.; Small, D. A FLT3-targeted tyrosine kinase inhibitor is cytotoxic to leukemia cells in vitro and in vivo. Blood 2002, 99, 3885-3891.

48. Kelly, J. A.; Spolski, R.; Kovanen, P. E.; Suzuki, T.; Bollenbacher, J.; Pise-Masison, C. A.; Radonovich, M. F.; Lee, S.; Jenkins, N. A.; Copeland, N. G.; Morse, H. C., 3rd; Leonard, W. J. Stat5 synergizes with T cell receptor/antigen stimulation in the development of lymphoblastic lymphoma. J Exp Med 2003, 198, 79-89.

49. Gu, L.; Vogiatzi, P.; Puhr, M.; Dagvadorj, A.; Lutz, J.; Ryder, A.; Addya, S.; Fortina, P.; Cooper, C.; Leiby, B.; Dasgupta, A.; Hyslop, T.; Bubendorf, L.; Alanen, K.; Mirtti, T.; Nevalainen, M. T. Stat5 promotes metastatic behavior of human prostate cancer cells in vitro and in vivo. Endocr Relat Cancer 2010, 17, 481-493.

50. Liu, X.; Robinson, G. W.; Gouilleux, F.; Groner, B.; Hennighausen, L. Cloning and expression of Stat5 and an additional homologue (Stat5b) involved in prolactin signal transduction in mouse mammary tissue. Proc Natl Acad Sci U S A 1995, 92, 8831-8835.

51. Horvath, C. M.; Wen, Z.; Darnell, J. E., Jr. A STAT protein domain that determines DNA sequence recognition suggests a novel DNA-binding domain. Genes Dev 1995, 9, 984-994.

52. Basham, B.; Sathe, M.; Grein, J.; McClanahan, T.; D'Andrea, A.; Lees, E.; Rascle, A. In vivo identification of novel STAT5 target genes. Nucleic Acids Res 2008, 36, 3802-3818.

53. Gu, L.; Dagvadorj, A.; Lutz, J.; Leiby, B.; Bonuccelli, G.; Lisanti, M. P.; Addya, S.; Fortina, P.; Dasgupta, A.; Hyslop, T.; Bubendorf, L.; Nevalainen, M. T. Transcription factor Stat3 stimulates metastatic behavior of human prostate cancer cells in vivo, whereas Stat5b has a preferential

393

Principal Investigator: Welch, Timothy R. 6.16

Postdoctoral Fellowship Application 2012

role in the promotion of prostate cancer cell viability and tumor growth. Am J Pathol 2010, 176, 1959-1972.

54. Chenoweth, D. M.; Dervan, P. B. Structural basis for cyclic Py-Im polyamide allosteric inhibition of nuclear receptor binding. J Am Chem Soc 2010, 132, 14521-14529.

55. Hsu, C. F.; Phillips, J. W.; Trauger, J. W.; Farkas, M. E.; Belitsky, J. M.; Heckel, A.; Olenyuk, B. Z.; Puckett, J. W.; Wang, C. C.; Dervan, P. B. Completion of a Programmable DNA-Binding Small Molecule Library. Tetrahedron 2007, 63, 6146-6151.

56. Chenoweth, D. M.; Dervan, P. B. Allosteric modulation of DNA by small molecules. Proc Natl Acad Sci U S A 2009, 106, 13175-13179.

57. Raskatov, J. A.; Hargrove, A. E.; So, A. Y.; Dervan, P. B. Pharmacokinetics of Py-Im Polyamides Depend on Architecture: Cyclic versus Linear. J Am Chem Soc 2012, Submitted.

58. Raskatov, J. A.; Nickols, N. G.; Dervan, P. B. Unpublished results, 2012. 59. Chenoweth, D. M.; Harki, D. A.; Dervan, P. B. Solution-phase synthesis of pyrrole-imidazole

polyamides. J Am Chem Soc 2009, 131, 7175-7181. 60. Harki, D. A.; Satyamurthy, N.; Stout, D. B.; Phelps, M. E.; Dervan, P. B. In vivo imaging of

pyrrole-imidazole polyamides with positron emission tomography. Proc Natl Acad Sci U S A 2008, 105, 13039-13044.

61. Baird, E. E.; Dervan, P. B. Solid phase synthesis of polyamides containing imidazole and pyrrole amino acids. J Am Chem Soc 1996, 118, 6141-6146.

62. Belitsky, J. M.; Nguyen, D. H.; Wurtz, N. R.; Dervan, P. B. Solid-phase synthesis of DNA binding polyamides on oxime resin. Bioorg Med Chem 2002, 10, 2767-2774.

63. Dose, C.; Farkas, M. E.; Chenoweth, D. M.; Dervan, P. B. Next generation hairpin polyamides with (R)-3,4-diaminobutyric acid turn unit. J Am Chem Soc 2008, 130, 6859-6866.

64. Pilch, D. S.; Poklar, N.; Baird, E. E.; Dervan, P. B.; Breslauer, K. J. The thermodynamics of polyamide-DNA recognition: hairpin polyamide binding in the minor groove of duplex DNA. Biochemistry 1999, 38, 2143-2151.

65. Muzikar, K. A.; Meier, J. L.; Gubler, D. A.; Raskatov, J. A.; Dervan, P. B. Expanding the Repertoire of Natural Product-Inspired Ring Pairs for Molecular Recognition of DNA. Org Lett 2011, ASAP.

66. Boyle, A. P.; Davis, S.; Shulha, H. P.; Meltzer, P.; Margulies, E. H.; Weng, Z.; Furey, T. S.; Crawford, G. E. High-resolution mapping and characterization of open chromatin across the genome. Cell 2008, 132, 311-322.

67. Gottesfeld, J. M.; Belitsky, J. M.; Melander, C.; Dervan, P. B.; Luger, K. Blocking transcription through a nucleosome with synthetic DNA ligands. J Mol Biol 2002, 321, 249-263.

68. Chen, X.; Xu, H.; Yuan, P.; Fang, F.; Huss, M.; Vega, V. B.; Wong, E.; Orlov, Y. L.; Zhang, W.; Jiang, J.; Loh, Y. H.; Yeo, H. C.; Yeo, Z. X.; Narang, V.; Govindarajan, K. R.; Leong, B.; Shahab, A.; Ruan, Y.; Bourque, G.; Sung, W. K.; Clarke, N. D.; Wei, C. L.; Ng, H. H. Integration of external signaling pathways with the core transcriptional network in embryonic stem cells. Cell 2008, 133, 1106-1117.

69. Teglund, S.; McKay, C.; Schuetz, E.; van Deursen, J. M.; Stravopodis, D.; Wang, D.; Brown, M.; Bodner, S.; Grosveld, G.; Ihle, J. N. Stat5a and Stat5b proteins have essential and nonessential, or redundant, roles in cytokine responses. Cell 1998, 93, 841-850.

394

  395

LIST OF ABBREVIATIONS

Ac2O Acetic anhydride

AcCl Acetyl Chloride

AcOH Acetic acid

AIBN Azobisisobutyronitrile

AQN Anthraquinone

ATP Adenosine-5’-triphosphate

9-BBN 9-Borabicyclo[3.3.1]nonane

Bn Benzyl

BnBr Benzyl bromide

Boc tert-Butoxycarbonyl

Boc2O Di-tert-butyldicarbonate

BOMCl Benzyloxymethyl chloride

BOP (Benzotriazol-1-yloxy)tris(dimethylamino) phophonium

hexafluorophosphate

CbzCl Benzyl chloroformate

cod 1,5-Cyclooctadiene

CREB cAMP response element-binding

d Day

Davis’s oxaziridine 2-(Phenylsulfonyl)-3-phenyloxaziridine

DEAD Diethyl azodicarboxylate

Dess-Martin or DMP Triacetoxy o-iodoxybenzoic acid

DBU 1,8-Diazabicyclo[5.4.0]undec-7-ene

DHQD Dihydroquinidine

DIBAL-H Diisobutylaluminium hydride

DMAP 4-(Dimethylamino)-pyridine

  396

DMDO Dimethyldioxirane

DME Dimethoxyethane

DMF Dimethylformamide

DMS Dimethylsulfide

DMSO Dimethyl sulfoxide

dppf 1,1’-Bis(diphenylphosphino)ferrocene

DTBMP 2,6-di-tert-butyl-4-methylpyridine

EDCI N-(3-dimethylaminopropyl)-N’-ethylcarbodiimide

ee Enantiomeric excess

e.r. Enantiomeric ratio

Et3N Triethylamine

EtOAc Ethyl Acetate

Et2O Diethyl ether

EtOH Ethanol

Fmoc Fluorenylmethyloxycarbonyl

FmocOSu 9-Fluorenylmethyl N-succinimidyl carbonate, N-(9-

fluorenylmethoxycarbonyloxy)succinimide

GSH Glutathione

h Hour

HATU O-(7-Azabenzotriazol-1-yl)-N,N,N’,N’-tetramethyluronium

hexafluorophosphate

HIF-1 Hypoxia-inducible factor 1

hν Irradiation with light

iBu Isobutyl

iPr Isopropyl

iPr2NEt N,N-Diisopropylethylamine

Imid Imidazole

KOtBu Potassium tert-butoxide

LDA Lithium N,N-diisopropylamide

LHMDS Lithium bis(trimethylsilyl)amide

mCPBA m-Chloroperbenzoic acid

  397

Me Methyl

MeCN Acetonitrile

MeI Methyl iodide

MeOH Methanol

min Minute

MOMCl Methyl chloromethyl ether

MRSA Methicillin-resistant Staphylococcus aureus

MsCl Methanesulfonyl chloride

NADPH Nicotinamide adenine dinucleotide phosphate

NaHMDS Sodium bis(trimethylsilyl)amide

NBS N-Bromosuccinimide

NCS N-Chlorosuccinimide

NIS N-Iodosuccinimide

nBuLi n-Butyllithium

N-PSP Diphenyl diselenide

O/N Overnight

Pd(OAc)2 Palladium(II) acetate

Pd/C Palladium on carbon

Ph Phenyl

PhI(OAc)2 Iodobenzene diacetate

PhMe Toluene

PPTS Pyridinium p-toluenesulfonate

PPY (R)-(+)-4-pyrrolidinopyridinyl(pentamethylcyclo-

pentadienyl)iron

Py. or Pyr. Pyridine

No Rxn No reaction was observed

RT or r.t. Room temperature

Sat’d Saturated aqueous solution

T3P Propane phosphonic acid anhydride

TBAF Tetrabutyl ammonium fluoride

TBDMSCl or TBSCl tert-Butyldimethylsilyl chloride

  398

TBDPS tert-Butyldiphenylsilyl

TBSOTf tert-Butyldimethylsilyl trifluoromethanesulfonate

Teoc 2-(trimethylsilyl)ethoxycarbonyl

TES Triethylsilyl

Tf Trifluoromethanesulfonate

TFA Trifluoroacetic acid

TFAA Trifluoroacetic anhydride

Tf2O Trifluoromethanesulfonyl anhydride

THF Tetrahydrofuran

TIPS Triisopropylsilyl

TLC Thin layer chromatography

TMS Trimethylsilyl

TMSOTf Trimethylsilyl trifluoromethylsulfonate

Tr Trityl (triphenylmethyl)

Triton B Benzyltrimethylammonium hydroxide

pTsOH or pTSA p-Toluenesulfonic acid

µw Microwave irradiation

V-70 2,2’-Azobis(4-methoxy-2,4-dimethyl valeronitrile)

VRE Vancomycin-resistant Enterococcus