ABSTRACT

Design and Synthesis of Small-Molecule Inhibitors of L Bearing the Thiosemicarbazone Warhead as Potential Anti-Metastatic Agents

Erica N. Parker, Ph.D.

Mentor: Kevin G. Pinney, Ph.D.

Metastasis is a devastating component associated with tumor progression which claims the majority of lives lost to cancer. Upregulation of , a powerful cysteine capable of degrading components in the extracellular matrix, is correlated with increased invasiveness of cancer cells and poor prognosis. Consequently, cathepsin L has emerged as a target for the development of new anticancer agents designed to mitigate the invasion and metastasis of malignant cancer cells. The development of a focused, small library of thiosemicarbazone analogues was pursued in an effort to discover potent and selective inhibitors of cathepsin L, which possess the potential to bind to the of cathepsin L through a proposed covalent interaction with the catalytic amino acid residue Cys25. Incorporation of the thiosemicarbazone moiety with a variety of aryl based molecular scaffolds led to a functionally diverse series of thiosemicarbazone analogues that were evaluated, through collaborative efforts, for their ability to inhibit cathepsin L and thus to potentially function as anti-metastatic agents. Benzophenone, benzoylbenzophenone, di-2-naphthalenylmethanone, 1,3-

diphenylpropan-2-one, dibenzylideneacetone, and 1,5-diphenyl-3-pentanone aryl-based

molecular scaffolds were all explored. From the thirty-five compounds in this

thiosemicarbazone based library, fifteen analogues were determined to be potent

inhibitors of cathepsin L (IC50 < 10 μM). 1,3-Bis(2-fluorobenzoyl)-5-bromobenzene

thiosemicarbazone (KGP312) demonstrated the greatest inhibitory activity against

cathepsin L with an IC50 value of 8.1 nM. Furthermore, KGP312 inhibited the invasion

and migration of both MDA-MB-231 breast cancer cells and PC-3ML prostate cancer

cells. In addition to the design and synthesis of new thiosemicarbazone analogues,

improvement of the synthetic route for our previously discovered lead cathepsin L

inhibitor, KGP94, was achieved. Derivatization designed to increase aqueous solubility

and potentially oral bioavailability was performed and ultimately led to the preparation of

KGP420, the phosphate prodrug of KGP94. Owing to high cathepsin L inhibitory activity

and favorable outcomes in initial biological studies, on-going collaborative efforts have

been aimed towards the advancement of these preclinical candidates as potential anti- metastatic agents.

Design and Synthesis of Small-Molecule Inhibitors of Cathepsin L Bearing the Thiosemicarbazone Warhead as Potential Anti-Metastatic Agents

by

Erica N. Parker, B.S.

A Dissertation

Approved by the Department of Chemistry and Biochemistry

Patrick J. Farmer, Ph.D., Chairperson

Submitted to the Graduate Faculty of Baylor University in Partial Fulfillment of the Requirements for the Degree of Doctor of Philosophy

Approved by the Dissertation Committee

Kevin G. Pinney, Ph.D., Chairperson

Charles M. Garner, Ph.D.

Robert R. Kane, Ph.D.

Mary L. Trawick, Ph.D.

Bessie W. Kebaara, Ph.D.

Accepted by the Graduate School December 2015

J. Larry Lyon, Ph.D., Dean

Page bearing signatures is kept on file in the Graduate School.

Copyright © 2015 by Erica N. Parker All rights reserved

TABLE OF CONTENTS

LIST OF FIGURES ...... vii LIST OF SCHEMES...... ix LIST OF TABLES ...... x ACKNOWLEDGMENTS ...... xi DEDICATION ...... xiv CHAPTER ONE ...... 1 Introduction ...... 1 Cancer ...... 1 Structure of ...... 1 Normal Physiological Roles of Cathepsins ...... 5 Cathepsins and Cancer ...... 9 Inhibitors of Cathepsins ...... 11 Thiosemicarbazones ...... 18 CHAPTER TWO ...... 22 Synthesis and biochemical evaluation of benzoylbenzophenone thiosemicarbazone analogues as potent and selective inhibitors of cathepsin L ...... 22 1. Introduction ...... 24 2. Results and discussion ...... 27 2.1 Design and synthetic chemistry ...... 27 2.2 Cathepsin Inhibition Studies ...... 32 2.3 Molecular modeling studies ...... 37 2.4 Invasion and migration studies ...... 39 2.5 Growth Inhibition of Mammary Carcinoma ...... 41 2.6 Cytotoxicity ...... 42 3. Conclusions ...... 43 4. Experimental Section ...... 44 4.1 Chemistry ...... 44 4.2 Biology ...... 74 Acknowledgments ...... 79

v Supplementary data ...... 80 References ...... 80 CHAPTER THREE ...... 88 Synthesis and Biological Evaluation of a Water Soluble Phosphate Prodrug Salt and Structural Analogues of KGP94, a Lead Inhibitor of Cathepsin L ...... 88 Abstract ...... 88 Introduction ...... 89 Results and Discussion ...... 93 Biological evaluation ...... 99 Conclusion ...... 101 Experimental Section ...... 102 Acknowledgements ...... 125 CHAPTER FOUR ...... 126 Design and Synthesis of Cathepsin Inhibitors: Symmetrical Thiosemicarbazone Analogues ...... 126 Introduction ...... 126 Symmetrical Benzophenone Thiosemicarbazones ...... 126 Dibenzylideneacetone and 1,5-diphenyl-3-pentanone thiosemicarbazones ...... 131 General Synthetic Methods: Synthesis of Symmetrical Thiosemicarbazone Analogues ...... 136 CHAPTER FIVE ...... 162 Conclusions ...... 162 APPENDICES ...... 164 Appendix A ...... 166 Appendix B ...... 365 Appendix C ...... 504 REFERENCES ...... 668

vi

LIST OF FIGURES

Figure 1.1. Structural features of cathepsin L, a -like protease ...... 2

Figure 1.2. Catalytic mechanism of peptide hydrolysis in cysteine cathepsin……………………………………………..…………………………………………………………………...…...3

Figure 1.3. Schechter and Berger nomenclature: Defining subsites of ...... 4

Figure 1.4. Cathepsins and the MHC class II antigen-presentation pathway ...... 5

Figure 1.5. and bone resorption ...... 7

Figure 1.6. Involvement of cathepsins in invasion and migration of tumor cells ...... 11

Figure 1.7. Cathepsin inhibitors in clinical trials and advanced pre-clinical studies ...... 13

Figure 1.8. Cathepsin inhibitors from natural sources ...... 14

Figure 1.9. Inhibitors of cathepsins described in literature ...... 16

Figure 1.10. Thiosemicarbazone based inhibitors of cysteine proteases ...... 19

Figure 1.11. Proposed cathepsin L inhibition mechanism with thiosemicarbazone based inhibitors ...... 21

Figure 2.1 Overview of cysteine cathepsins in invasion and metastasis...... 24

Figure 2.2 Representative potent inhibitors of cathepsin L utilizing various warheads. ...26

Figure 2.3. Activity of selected benzophenone thiosemicarbazone analogues against cathepsin L ...... 27

Figure 2.4. Isomerization of thiosemicarbazone analogue 33 in DMSO-d6 ...... 32

Figure 2.5. Representative progress curves of cathepsin L (1 nM) activity using Z-FR-AMC (10 μM) as substrate and increasing concentrations of (A) inhibitor 1 and (B) inhibitor 32 (0–10 μM)...... 36

Figure 2.6. Molecular docking of analogues 1 and 32 within the active site of cathepsin L ...... 37

Figure 2.7. Effect of cathepsin L inhibitors on tumor cell invasion...... 39

vii Figure 2.8. Inhibition of invasion and migration of MDA-MB-231 breast cancer cells by thiosemicarbazone analogues 1, 8, and 32 ...... 40

Figure 2.9. 3-Benzoylbenzophenone thiosemicarbazone (1) effects on the growth of C3H mammary carcinomas implanted in the right rear foot of female CDF1 mice...... 41

Figure 3.1. Cathepsin inhibitors in the pharmaceutical pipeline...... 90

Figure 3.2 Representative sampling of potent covalent inhibitors of cathepsin L ...... 91

Figure 3.3. Sub-set of previously described thiosemicarbazone based inhibitors of cathepsin L ...... 92

Figure 3.4. ROESY and COSY correlations for analogue 13 as a representative example of the major isomer observed in DMSO-d6...... 97

Figure 3.5. KGP420 Incubated with 1.2 Units ALP...... 100

Figure 4.1. Previously synthesized benzophenone thiosemicarbazone analogues ...... 126

Figure 4.2 X-ray crystal structure of compound 37 ...... 135

viii

LIST OF SCHEMES

Scheme 2.1. Synthesis of benzoylbenzophenone thiosemicarbazone analogues...... 28

Scheme 2.2. Synthesis of benzoylbenzophenone thiosemicarbazone analogues utilizing Friedel-Crafts acylation...... 29

Scheme 2.3. Synthesis of benzoylbenzophenone thiosemicarbazone analogues utilizing isophthaloyl dichloride ...... 30

Scheme 2.4. Synthesis of benzoylbenzophenone thiosemicarbazone analogues utilizing 1,3,5-tribromobenzene...... 31

Scheme 3.1. Previously reported synthetic route towards KGP94 ...... 94

Scheme 3.2. Improved synthetic route towards KGP94 (11) and analogues 12-13...... 95

Scheme 3.3. Synthesis of dimethylresorcinol and resorcinol analogues 19-22 ...... 96

Scheme 3.4. Prodrug derivatization of KGP94 (11) to form water-soluble salt KGP420...... 97

Scheme 4.1. Synthesis of thiosemicarbazones with a symmetrical benzophenone scaffolds ...... 128

Scheme 4.2. Synthesis of the 1,3-diphenylpropan-2-one thiosemicarbazone ...... 129

Scheme 4.3. Synthesis of di(naphthalen-2-yl)methanone thiosemicarbazone ...... 129

Scheme 4.4. Synthesis of dibenzylideneacetone and 1,5-diphenyl-3-pentanone thiosemicarbazone analogues ...... 131

Scheme 4.5. Synthesis of (E)-7-(4-fluorobenzylidene)-3-(4-fluorophenyl)- 7a-hydroxy-3a,4,5,6,7,7a-hexahydro-1H-indazole-1-carbothioamide ...... 134

ix

LIST OF TABLES

Table 1.1 Cathepsin L processing of proneuropeptides ...... 8

Table 1.2 Cathepsin Inhibitors in the pharmaceutical pipeline ...... 12

Table 2.1 Inhibitory activity of benzoylbenzophenone thiosemicarbazone analogues...... 34

Table 2.2. Evaluation of cytotoxicity against HUVECs ...... 42

Table 3.1. Isomerization of benzophenone thiosemicarbazone analogues in

DMSO-d6 ...... 98

Table 3.2. Inhibitory activity of benzoylbenzophenone thiosemicarbazone Analogues ...... 99

Table 3.3. Cytotoxicity against HUVECs ...... 101

Table 4.1. Inhibitory activity of benzophenone symmetrical thiosemicarbazones and extended analogues 12 and 15...... 130

Table 4.2. Reduction of dibenzylideneacetone ...... 132

Table 4.3. Inhibitory activity of dibenzylideneacetone and 1,5-diphenyl-3-pentanone thiosemicarbazone analogues ...... 133

x

ACKNOWLEDGMENTS

I am grateful to many who have been influential in my life and helped mold the individual I have become. I am especially thankful to those who have in the past several

years offered so much support and encouragement during the journey towards my doctoral degree. I acknowledge, with gratitude, my debt of thanks to my advisor Dr.

Kevin G. Pinney for his mentorship and guidance throughout the time of my graduate career. I admire his immense passion for the development of anti-cancer therapeutics and greatly appreciate his unwavering patience as a leader, endless enthusiasm, and dedication. My experience as a graduate student in Dr. Pinney’s Laboratory has influenced me to pursue a career in medicinal chemistry.

I am thankful to Dr. Mary Lynn Trawick, an exceptional committee member and collaborator, who has offered encouragement over the years and kindly and enthusiastically strengthened my knowledge concerning the biological aspects of the cathepsin project through insightful discussions.

I am thankful to Dr. Charles M. Garner for his encouragement and appreciate his generosity, enthusiasm, and suggestions in helping me understand research based questions.

I express my appreciation and gratitude to my dissertation committee Dr. Kevin

G. Pinney, Dr. Charles M. Garner, Dr. Robert R. Kane, Dr. Mary Lynn Trawick, and Dr.

Bessie W. Kebaara who have given their valuable time in reviewing material for oral examinations and the dissertation defense as well as insightful comments and encouragement.

xi I would like to thank friends and colleagues in the Pinney Group and Trawick

Group. I would especially like to thank my classmates Chen-Ming and Laxman who have shared the same journey in pursuit of our doctoral degrees. We ardently encouraged each other along the way, provided valuable advice to each other, and I deeply cherish the friendship we have developed over the years.

I am thankful to Nancy Kallus, Adonna Cook, Virginia Hynek, and Andrea

Johnson for their hard work and help during these years. I am thankful to the Department of Chemistry and Biochemistry, Baylor University, and OXiGENE Inc. for their generous financial support.

I am extremely thankful to my family and friends for their love, encouragement, and support. From a very early age, my Mom encouraged my innate curiosity and emboldened me to pursue my dreams. Both my Mom and my Dad through pursuit of their own goals have demonstrated that hard-work, dedication, and perseverance are key elements for achieving personal dreams. For their infinite love and encouragement, I am thankful to my, Mom, Dad, Adele, Matt, Paw, Granny, and Meemaw. I am grateful to my sisters, Jessica and Julia, and truly treasured friends Anna, Sarah, Melissa, Kelly, and

Morgan for acceptance, understanding, laughter, consolation, motivation, and encouragement. I am thankful to my friend Anna who has been very much like a sister to me; I greatly appreciate her advice and honesty as well as her enduring encouragement in both social and intellectual realms. I express appreciation and gratitude to my friend

Sarah, a rock star in her own rights, who so often instills strength, courage, and a fiery passion in others through her encouragement. I am thankful to Vikas Kumar who has

xii provided me with vast amount of support, understanding, and encouragement over the past few years.

I am immensely privileged to have mentors, colleagues, family, and friends who

have given me an enormous amount of strength during this journey.

xiii

DEDICATION

To My Parents and Grandparents

xiv

CHAPTER ONE

Introduction

Cancer

Over 90% of fatalities resulting from cancer are attributed to metastasis. A number of changes occur within the tumor microenvironment which facilitates detachment of a tumor cell from the local, invasion into the surrounding tissue of the primary tumor site, migration to a distant location, and establishment of a secondary

tumor. The availability of anti-metastatic agents remains largely an unmet need.1,2

Inhibition of cysteine cathepsins has emerged in recent years as a potential anti-metastatic

target owing to their ability to degrade components of the extracellular matrix aiding in

cancer cell invasion migration and the correlation between their upregulation in various

cancers and overall poorer prognosis.

Structure of Cathepsins

Cathepsins are comprised of a diverse class of proteases including the serine

cathepsins A and G, the aspartyl cathepsins D and E, the metalloprotease cathepsin III,

and the cysteine cathepsins B, L, K, S, V, F, W, H, X, C, and O.3,4 The cysteine

cathepsins belong to the C1 family, also known as the papain family, of the Clan A

cysteine peptidases.5 Cysteine cathepsins, with the exception of , are

monomeric proteins comprised of a single polypeptide chain containing between 214 and

260 amino acids in their mature forms. Two domains, a left domain containing three α

helices and a right domain containing a β barrel motif, forming a V-shaped active site

cleft define the general tertiary structure of the mature (Figure 1.1). Hydrolysis

1 of peptide bonds are facilitated by the amino acid residues Cys25 (left domain), His159

(right domain), and Asn175 (right domain) which form the .6,7

Figure 1.1. Structural features of cathepsin L, a papain-like protease (Reprinted from Biochimica et Biophysica Acta (BBA)-Proteins and Proteomics, Vol 1824, Vito Turk, Veronika Stoka, Olga Vasiljeva, Miha Renko, Tao Sun, Boris Turk, Dusan Turk, Cysteine cathepsins: From structure, function and regulation to new frontiers, 66-88, 2012, with permission from Elsevier.)8

In papain-like enzymes, the Cys25 and His159 residues exist as a thiolate/imidazolium ion pair between the approximate pH range of 4-8.5.6,9 Analogous to the aspartic acid residue in serine proteases, the asparagine residue helps stabilize the active site thiolate/imidazolium ion pair through hydrogen bonding.6,10 The hydrogen bond between the His159 imidazolium NH and Asn175 side chain carbonyl is shielded from solvent molecules by the aromatic side chain of Trp177. Substrate proteolysis is initiated by the attack of the nucleophilic thiolate ion on the amide carbonyl carbon of the substrate to form a thiohemiketal intermediate (Figure 1.2).

2

Figure 1.2. Catalytic mechanism of peptide hydrolysis in cysteine cathepsins

The tetrahedral intermediate is stabilized in the through hydrogen bonding with the side chain NH of Gln19 and the backbone NH of Cys25.6,11 Proton

transfer from the imidazolium ion NH to the substrate NH followed by collapse of the

tetrahedral intermediate releases the C-terminus of the peptide and generates a thioester

intermediate. Deacylation of the is initiated by the nucleophilic attack of an

activated water molecule to form a tetrahedral intermediate which subsequently collapses

to release the N-terminus of the substrate and regenerate the enzyme active site.6

The active sites of proteases can be further described from the manner in which

amino acid residues of natural substrates bind within the active site of the enzyme prior to hydrolysis. According to Schechter and Berger nomenclature, 12,13 binding regions of the

3 enzyme are designated as subsites, S, and amino acid residues of the substrate are assigned positions, P. Numbering of the positions of the amino acid residues in the substrate begins with the cleaved peptide bond with the C-terminus residues labeled as primed and the N-terminus residues labeled as non-primed. Numbering of the enzyme binding regions, subsites, directly correlates to assigned P values for the amino acid residues of the substrate (Figure 1.3).

Figure 1.3. Schechter and Berger nomenclature: Defining subsites of proteases. (Reprinted by permission from Macmillan Publishers Ltd: [Nature Reviews Drug Discovery]14, copyright 2006)

The mature form of cathepsin L shares 79.5% sequence identity with cathepsin

V,15 the closest human homolog, 60% sequence identity with cathepsin K,16 and 57% sequence identity with .17 The S2 subsite of papain enzymes forms a binding pocket and is generally considered as the specificity binding pocket. The S2 pocket of cathepsin L, an endopeptidase, is large and hydrophobic and prefers amino acids with aromatic side chains. , an endopeptidase, showed preference for hydrophobic aromatic amino acid residues at the P2 position. Cathepsin S and Cathepsin K are both endopeptidases which prefer non-aromatic hydrophobic residues in the S2 binding pocket. , and endopeptidase as well as exopeptidase, prefers large aromatic residues as well as amino acids with basic side chains such as arginine in the S2 binding

4 pocket. Cathepsins L, V, K, S, and B prefer arginine, lysine and glycine at the S1 subsite.18,19

In addition to protein turnover, cysteine cathepsins have been implicated in specialized physiological roles such as antigen presentation, bone remodeling, neuropeptide and hormone processing. Cathepsins L and S play significant roles in the major histocompatibility complex (MHC) class II antigen presentation pathway (Figure

1.4). In antigen presenting cells, the invariant chain acts as a chaperone molecule of the

MHC class II αβ heterodimer as well as prevents premature binding of antigen peptides.20–23

Normal Physiological Roles of Cathepsins

Figure 1.4. Cathepsins and the MHC class II antigen-presentation pathway (Reprinted by permission from Macmillan Publishers Ltd: [Nature Reviews Immunology]20, copyright 2003)

5 Endosomal cathepsin L24 (present in cortical thymic epithelial cells and

macrophages) and cathepsin S25,26 (present in dendritic cells, B cells, and macrophages)

participate in the late-stage degradation of the invariant chain leaving a smaller peptide

fragment CLIP, which ultimately undergoes exchange with an antigen peptide. In

addition to processing of the invariant chain, cathepsins degrade antigens to generate

epitopes which can bind to the MHC class II αβ heterodimer.27,28 Once the antigen peptide is bound to the MHC class II αβ heterodimer and expressed on the cell surface, the MHC class II complex binds to the CD4+ protein on a T-helper cell allowing the

antigen to bind to the T-cell receptor which subsequently activates the adaptive immune

response.20

Discovery of a physiological role for cathepsin K emerged from the link between pycnodysostosis, a disease characterized by increased bone density resulting from

abnormal accumulation of collagen in osteoclasts, and a mutation in the cathepsin K

leading to deficient translation of the lysosomal protease.29,30 Physiological bone

remodeling involves bone resorption performed by osteoblasts followed by bone

formation performed by osteoclasts.31–33 Bone matrix consists of the inorganic bone

matrix which is primarily comprised of hydroxyapatite and calcium carbonate, and the organic bone matrix of which 90% is type I collagen.33–37 During bone resorption osteoclasts are recruited to the bone surface and acidify the extracellular lacunae which facilitate bone demineralization.38,39 Secretion of cathepsin K, a powerful collagenolytic

enzyme, into the resorption lacunae facilitates type I collagen degradation (Figure 1.5).40

Cathepsin K degrades collagen in the telopeptide region as well as the helical region

6 when in an oligomeric complex with glycosaminoglycans. The collagen fragments are endocytosed and subsequently released at the cell’s antiresorptive surface.41

Figure 1.5. Cathepsin K and bone resorption (Reprinted by permission from Macmillan Publishers Ltd: [IBMS BonekEy]31, copyright 2008)

Cathepsin L, also known as prohormone thiol protease (PTP), 42 is involved in processing proneuropeptides into neuropeptides and peptide hormones (Table 1.1).43

Cathepsin L knockout models revealed substantial dependence for the presence of this protease in secretory vesicles for processing a number of proneuropeptides including proenkephalin,42 proneuropeptide Y,44 proopiomelanocortin,45 prodynorphin,46 and prochlocystokinin.47 Cleavage of proneuropeptides by cathepsin L produces

intermediates which subsequently undergo removal of terminal basic amino acid residues

7 by the appropriate aminopeptidase or carboxypeptidase to generate the active neuropeptide. Cathepsin L facilitates processing of pro-enkephalin, pro- opiomelanocortin, and pro-dynorphin to generate Met-enkephalin, β-endorphin, and dynorphin A/B which are ligands for opioid receptors that mediate analgesic effects.48

Proteolytic processing of proneuropeptide Y generates neuropeptide Y which has implications in stress induced obesity49 and regulation of blood pressure.50

Cholecystokinin octapeptide (CCK8) generated from proteolytic processing of procholecystokinin suppresses hunger51 and induces anxiety.52

Table 1.1 Cathepsin L processing of proneuropeptides a

Proneuropeptide Neuropeptide/ Regulatory Percent hormone function reduction in CTSL KO model

Proenkephalin (Met)enkephalin Analgesia 50%42

Proneuropeptide Y Neuropeptide Y Blood pressure and 80-90% obesity

Proopiomelanocortin ACTH Steroid production 77%

Β-endorphin Analgesia 82%

α-MSH Skin pigmentation 93%

Prodynorphin Dynorphin A Analgesia 75%

Dynorphin B Analgesia 83%

α-Neoendorphin Analgesia 90% procholecystokinin CCK8 Anxiety, cognition, 75% analgesia a Adapted from L. Funkelstein et al43

8 Cathepsins and Cancer

Dysregulation of normal physiological processes contributes to numerous disease

states. For reviews discussing the involvement of cathepsins in cancer metastasis, see the

cited references.53–56 A strong association between cathepsins and cancer developed when

the major excreted protein (MEP) of transformed cells, which exhibited a 200-fold

increase in extracellular levels compared to non-transformed , was identified as cathepsin

L;57–60 moreover, variance of cathepsin expression of tumor cells coincides with the

metastatic state of the tumor cells.61,62

In A375SM melanoma cells, hypomethylation of a GpG island in the promoter

region of the gene encoding for cathepsin L leads to increased translation and subsequent

synthesis of the .63 Regulation of cathepsin proteolytic activity occurs

through auxiliary mechanisms beyond translation and protein synthesis. The prodomain

occupies the active site in a backwards binding orientation and prevents unwarranted proteolysis. Binding affinity of the pro-domain within the active sites of cathepsin are pH

dependent.64–66 Cathepsins are autocatalytically activated within endosomes or lysosomes

under low pH conditions.15,67,68 Acidosis of the tumor microenvironment contributes to

increased cathepsin activity through activation of proforms. Proteolytic potential of the

mature enzymes are governed by pH dependent activity and stability in which acidic tumor microenvironments provide favorable conditions.15 Additionally, hypoxic and

acidic conditions inherent in the tumor microenvironment lead to trafficking of lysosomes to the plasma membrane and subsequent exocytosis.69,70

Proteolytic activity of cathepsins are regulated by the endogenous proteins type 1

cystatins (stefins), type 2 cystatins, and type 3 cystatins (kininogens). Stefins A and B

9 primarily exhibit intracellular localization. Type 2 cystatins (C, D, E/M, F, S, SA, and

SN) contain a signal peptide for extracellular translocation which allows for the regulation of the proteolytic activity of secreted proteases. High molecular weight and low molecular weight kininogens, stefin A, stefin B, and cystatin C are potent inhibitors of cathepsins L, B, K, H, and S with Ki values in the picomolar to nanomolar ranges. 71

Cystatins D, F, SA, SN, L-kininogen, and H-kininogen strongly inhibit cathepsin L.71 In tumor cells, increased expression of cathepsins often coincides with downregulation of endogenous inhibitors.72

Upregulation of cathepsin activity occurs through increased expression, secretion

into the extracellular space, acidification of extracellular space, and downregulation of

endogenous inhibitors. The proteolytic potential of cathepsins in the degradation of extracellular matrix proteins including fibronectin,73–76 laminin,74–76 collagen70,75–80 and

participation in proteolytic cascades,81 facilitates invasion and migration of malignant

tumor cells (Figure 1.6).8

10

Figure 1.6. Involvement of cathepsins in invasion and migration of tumor cells. (Reprinted from Bioorganic and Medicinal Chemistry, Vol xx, Parker, E. N.; Song, J.; Kishore Kumar, G. D.; Odutola, S. O.; Chavarria, G. E.; Charlton-Sevcik, A. K.; Strecker, T. E.; Barnes, A. L.; Sudhan, D. R.; Wittenborn, T. R.; Siemann, D. W.; Horsman, M. R.; Chaplin, D. J.; Trawick, M. L.; Pinney, K. G. Synthesis and Biochemical Evaluation of Benzoylbenzophenone Thiosemicarbazone Analogues as Potent and Selective Inhibitors of Cathepsin L, 6974-6992, Copyright 2015, with permission from Elsevier).

Inhibitors of Cathepsins

Cathepsin inhibitors as promising candidates for FDA approval are currently in the pipeline of several pharmaceutical companies (Table 1.2). Recently, GSK-

2793660,82,83 a cathepsin C inhibitor, was in phase I clinical trials for the treatment of bronchiectasis. Past and present clinical trials involving inhibition of cathepsin K include

Odanacatib84,85 and MIV-71186 which are primarily aimed towards decreasing bone

11 resorption in individuals with osteoporosis. Cathepsin S inhibitors in clinical trials

include VBY-37687,88 for the treatment of liver fibrosis and therapeutic agents for the

mediation of overactive immune responses in psoriasis (VBY-89189,90 and RWJ-

44538091), rheumatoid arthritis (RWJ-445380)92, and neuropathic pain (VBY-03693,94 and

MIV-24795). Disclosed structures of cathepsin inhibitors in the pharmaceutical pipeline

are included in Figure 1.7.

Table 1.2. Cathepsin inhibitors in the pharmaceutical pipeline

Inhibitor Targeted Therapeutic Clinical Trial Pharmaceutical Cathepsin Indication Phase Company VBY-825 Pan-Cysteine Primary Biliary Pre-Clinical Virobay Cirrhosis GSK-2793660 Cathepsin C Bronchiectasis Phase I GlaxoSmithKline VBY-376 Cathepsin B Liver Fibrosis Phase I Virobay Odanacatib Cathepsin K Osteoporosis Phase III Merck MIV-711 Cathepsin K Osteoporosis Phase I Medivir VBY-891 Cathepsin S Psoriasis Phase I Virobay and LEO Pharma VBY-036 Cathepsin S Neuropathic Phase I Virobay Pain LY3000328 Cathepsin S Abdominal Phase I Eli Lily Aortic Aneurysm RWJ-445380 Cathepsin S Rheumatoid Phase II Johnson & Arthritis, Johnson psoriasis MIV-247 Cathepsin S Neuropathic Pre-Clinical Medivir Pain

12 VBY-82596,97 (Virobay) has been implicated for the treatment of primary biliary

cirrhosis and inhibits cathepsins S, L, V, B, K, and F with Ki values in the picomolar to

nanomolar range. VBY-825 contains a carbonyl electrophilic which reversibly interacts

with the catalytic cysteine residue via a hemiothioketal intermediate. Odanacatib84,85

(Merck), also known as MK-0822, targets osteoporosis through inhibition of cathepsin K.

The nitrile based inhibitor reversibly and selectively inhibits cathepsin K. LY300032898,99

(Eli Lily) targets the treatment of abdominal aortic aneurysm through potent and selective

inhibition of cathepsin S (IC50 = 7.7 nM). A co-crystal structure of cathepsin S with

LY3000328 revealed that inhibition occurred through non-covalent interactions. To date,

cathepsin L specific inhibitors have not yet entered clinical trials.

Figure 1.7. Cathepsin inhibitors in clinical trials and advanced pre-clinical studies

Small-molecule inhibitors of cysteine cathepsins incorporate electrophilic

warheads which form covalent bonds with the Cys25 thiolate in the catalytic triad.

Several natural products have been discovered to be inhibitors of cysteine proteases

(Figure 1.8). Nonpeptidic natural products 3-epiursolic acid (I),100 isolated from Myrcia

linga, and the prenylated dihydrochalcone (II),101 isolated from Metrodorea stipularis,

displayed reasonable activity towards cathepsin L with IC50 values in the low micromolar

range.

13

Figure 1.8. Cathepsin inhibitors from natural sources

E-64 (III),102–105 isolated from the mold Aspergillus japonicus, is an

epoxysuccinyl peptide which demonstrates specificity towards cysteine proteases

compared to serine proteases. E-64 (III) irreversibly inhibits several cysteine proteases

including actinidin, papain, cathepsin L, cathepsin B, and cathepsin K. Leupeptin

(IV),106–109 isolated from actinobacteria within the Streptomyces genus, displays broad specificity against serine, cysteine, and threonine proteases. An aldehyde electrophilic warhead lies within its argininal peptide molecular framework. Leupeptin (IV) reversibly inhibits cathepsins L and B with IC50 values of 6.6 nM and 9.4 nM, respectively.

Tokaramide A (V),110 another argininal peptide aldehyde inhibitor, was isolated from the

marine sponge Theonella aff. Mirabilis in 1999. Evaluation against cathepsin B revealed

Tokaramide A (V) to inhibit 50% of enzyme activity at 61 nM. Miraziridine A (VI),

14 isolated from the same species of marine sponge as Tokaramide A (V), irreversibly inhibits cathepsin L and cathepsin B. Incorporated into the framework are an aziridine ring, a statine, and an αβ-unsaturated ester as electrophilic centers available for interaction with nucleophilic amino acid residues within protease active sites. Analysis of truncated miraziridine A (VI)111,112 analogues suggested that the aziridine ring contributes

to the inhibitory activity against cathepsin B. Initial biological evaluation of gallinamide

A (VII),113,114 isolated from marine cyanobacteria from a Schizothrix species, showed

that the natural product possessed potent antimalarial activity against Plasmodium

falciparum, Leishmania donovani, and Trypanasoma cruzi. Gallinamide A (VII)

irreversibly inhibits cathepsin L with a low IC50 value of 5.0 nM (30 min pre-incubation

time). Additionally, inhibition is 28-fold more selective for cathepsin L than cathepsin V

(also referred as cathepsin L2) which share 79.5% sequence identity in their mature

forms.

Based on the natural product E-64, Katanuma et al incorporated the

epoxysuccinate warhead into a new series of cathepsin inhibitors named CLIK.115

Emerging from this series of inhibitors was CLIK-148 (VIII) which displayed selective inhibition of cathepsin L compared to cathepsins B, C, K, and S. Inhibition of cathepsin L by vinyl sulfone IX116 occurs irreversibly through formation of an irreversible thioether

bond with the major catalytic amino acid residue (Cys25) of cathepsin L (Figure 1.9).

Inhibition occurs with a second order rate constant of 181.420 s-1 M-1. Molecular

modeling suggests that the phenyl substituent extending from the sulfone resides in the

S2 pocket, the biphenyl group resides in the S1 subsite, and the 4-methylpiperazinyl

group extends towards the S2’ subsite.

15

Figure 1.9. Inhibitors of cathepsins described in literature

KD-1 (X),117a peptide based vinyl sulfonate, irreversibly inhibited cathepsin L with a second order rate constant of 4.3 x 106 M-1 s-1 and was selective for cathepsin L

compared to cathepsin S by 13-fold, cathepsin K by 100-fold, and cathepsin B by 44,000-

fold. KD-1 inhibited proteolytic activity of cathepsin L towards collagen type 1 as well as

intracellular cathepsin L activity of MDA-MB-231 breast cancer cells. The natural

product leupeptin inspired the design of potent peptidyl aldehyde cathepsin L and

cathepsin B inhibitors. Among the described inhibitors, the tryptophanal derivative XI109

showed selectivity towards cathepsin L compared to cathepsin B and inhibited in vivo

118 bone resorption. The peptidomimetic α-ketoamide XII inhibited cathepsin S (IC50 =

5.8 nM) and cell migration of CL1-5 lung cancer cells and Human umbilical vein endothelial cells (HUVECs). Azepanone based inhibitors of cathepsins L, K, S, and B first reported in 2001 by Marquis and co-workers119 interact with the catalytic cysteine

16 residue through formation of a reversible hemithioketal. Modification of the backbone

template to incorporate a β-naphthylalanine moiety in the P2 position and a naphthylene-

1-carboxamide moiety in the P3 position led to the design of XIII120 which selectively

inhibited cathepsin L (Ki, app = 0.43 nM) compared to cathepsin K (Ki, app = > 10,000 nM)

by >10,000-fold, cathepsin S (Ki, app = 15.6 nM) by 36-fold, and cathepsin B (Ki, app = 150 nM) by 348-fold. Comparison of crystallographic and molecular modeling studies of azepanone based inhibitors attributed the increase in selectivity for cathepsin L compared to cathepsin K to disruption of π- π stacking interactions of the P3 substituent with Tyr67 in the S3 subsite of cathepsin K and increased steric bulk of the P2 β-naphthylalanine moiety in the S2 pocket of cathepsin K which cathepsin L can more easily

120 121–123 accommodate. Thiocarbazate XVI potently inhibited cathepsin L (IC50 = 19 nM)

and possessed selectivity over cathepsins S (530 nM) and B (3250 nM). Molecular

modeling studies with similar analogues suggest that the indole ring occupies the S2

pocket while the thiocarbazate carbonyl interacts with Cys25. Triazine nitrile XV124 inhibits rhodesain (Ki = 13 nM), a cysteine protease found within Trypanosoma brucei,

and cathepsin L (Ki = 3 nM). Co-crystal structure of a triazine nitrile XV with cathepsin

L revealed that the cyclohexyl portion of the molecule resides deep within the S2 pocket and a thioimidate intermediate is formed between the Cys25 thiolate and nitrile of XV which is stabilized through hydrogen bonding with the side chain of Gln19 in the oxyanion hole. Optimization through interchanging substituents on the triazine ring lead to analogues with increased selectivity for rhodesain compared to cathepsin L.

125 Nonpeptidic cyanamide XVI inhibited cathepsin K (IC50 = 50 nM) and cathepsin L

17 (IC50 = 80 nM). Inhibitors of this class of molecules were found to be reversible time-

dependent inhibitors of cathepsin K.

Thiosemicarbazones

Thiosemicarbazone based inhibitors have been designed for a number of cysteine proteases of the papain family found in a number of species including Homo

sapiens,69,126–133,134 Trypanosoma cruzi and Trypanosoma brucei,135–140 Plasmodium

140–142 143 144 falciparum, Leishmania Mexicana, and Eimeria tenella (Figure 1.10). Prior to

the utilization of thiosemicarbazone based protease inhibitors closely related

semicarbazone analogues were shown to be bioisosteric replacements for dipeptides,145 and evaluated for inhibitory activity against cathepsin K.146 As an interesting note, semicarbazones are used as protecting groups for the purification of peptidyl aldehydes.147

In 2002, Du et al published a manuscript describing inhibitors of cruzain, a

cysteine protease in Trypanosoma cruzi which potentiates Chagas disease, with a variety

of scaffolds incorporating the thiosemicarbazone moiety including 3′-

bromopropiophenone thiosemicarbazone XVII (cruzain IC50 = 100 nM) and 5-(3-

chlorophenyl)-2-furancarboxaldehyde thiosemicarbazone XVIII (cruzain IC50 = 140

nM)136 (Figure 1.10). In 2004, Fujii et al described thiosemicarbazone based inhibitors of

cathepsin-L like cysteine proteases found in Trypanosoma brucei (rhodesain) and

Trypanosoma cruzi (cruzain) incorporating the 3,4-dichlorophenyl functionality on one

side of the imine bond and a variety of groups on the other side of the imine bond

139 including analogue XIX (cruzain IC50 = 40 nM, rhodesain IC50 = 38 nM).

18

Figure 1.10. Thiosemicarbazone based inhibitors of cysteine proteases

Drawing inspiration from these studies the Pinney Laboratory in collaboration with the Trawick Laboratory, described (in 2006) a series of thiosemicarbazone based cruzain inhibitors with tetrahydronaphthalene, benzophenone, and propiophenone molecular frameworks. 135 m-Bromo substituted benzophenone analogue XX (Cruzain

IC50 = 24 nM) and 7-bromo tetrahydronaphthalene analogue XXI (Cruzain IC50 = 17 nM)

emerged as lead inhibitors of cruzain.135 Selectivity screening for inhibitors of cruzain

against human cathepsins L, B, and K revealed analogues with potent inhibition against

these cysteine proteases. Owing to the upregulation of cathepsins in several diseases

including cancer, collaborative research in the Pinney Laboratory and the Trawick

Laboratory gradually channeled to the design and synthesis of cathepsin inhibitors as

anti-metastatic agents. Explored molecular frameworks (Pinney and Trawick

19 collaboration) incorporating the thiosemicarbazone warhead for cathepsin inhibition in

the include benzophenone, pyridine, thiophene, fluorene, thiochromanone, benzothiepine,

dihydroquinoline, and benzoylbenzophenone. 69,126–133 Potent thiosemicarbazone based

cathepsin L inhibitors of these series include (3-bromo-3'-hydroxybenzophenone)

128,129,69 thiosemicarbazone XXII (cathepsin L IC50 = 131.4 nM), (3-bromo-2'-

127 fluorobenzophenone) thiosemicarbazone XXIII (cathepsin L IC50 = 30.5 nM), and 6-

131 nitrothio-4-chromanone thiosemicarbazone XXIV (cathepsin L IC50 = 68 nM). In 2014,

Raghav et al described benzaldehyde thiosemicarbazone analogues including o-

chlororbenzadehyde thiosemicarbazone XXV, which inhibited cathepsin B with a Ki

value of 1.48 x10-5 M which was 50 times greater than the value observed for the

corresponding semicarbazone benzaldehyde derivative.134 The indole based

thiosemicarbazone XXVI discovered in a high throughput screening inhibited CPB, a

cathepsin L-like protease found in the protozoan Leishmania Mexicana, with an IC50 value of 60 nM.143 High throughput screening which led the identification of CPB

inhibitors also led to inhibitors of the cathepsin-B like protease EtCatB found in Eimeria

tenella. 144 Pryazole thiosemicarbazone analogue XXVII moderately inhibited EtCatB

(EtCatB IC50 = 940 nM) and showed selectivity against bovine cathepsin B (IC50 > 30).

Isatin based thiosemicarbazone analogues including XXVIII (falcipain-2 = 6070 nM)141 and dihydro-arteminsinin derivatives including XXIX (falcipain-2 = 520 nM)142

moderately inhibited the cysteine protease falcipain-2 found in the malaria causing

protozoan Plasmodium falciparum.

The proposed mechanism of inhibition, supported by molecular docking and

kinetic studies,129,133 of papain-like proteases with thiosemicarbazone inhibitors based on

20 the benzophenone and benzoylbenzophenone scaffolds occurs through formation of a

transient covalent bond between the thiocarbonyl of the inhibitor and the Cys25 thiolate

of the enzyme (Figure 1.11). Kinetic studies have shown that cathepsin inhibition with

these analogues is time-dependent and reversible.129,133

Figure 1.11. Proposed cathepsin L inhibition mechanism with thiosemicarbazone based inhibitors

21

CHAPTER TWO

Synthesis and biochemical evaluation of benzoylbenzophenone thiosemicarbazone analogues as potent and selective inhibitors of cathepsin L

This chapter is reprinted from Bioorganic and Medicinal Chemistry, Vol xx, Parker, E. N.; Song, J.; Kishore Kumar, G. D.; Odutola, S. O.; Chavarria, G. E.; Charlton-Sevcik, A. K.; Strecker, T. E.; Barnes, A. L.; Sudhan, D. R.; Wittenborn, T. R.; Siemann, D. W.; Horsman, M. R.; Chaplin, D. J.; Trawick, M. L.; Pinney, K. G. Synthesis and Biochemical Evaluation of Benzoylbenzophenone Thiosemicarbazone Analogues as Potent and Selective Inhibitors of Cathepsin L, 6974-6992, Copyright 2015, with permission from Elsevier. The author Erica N. Parker played a significant role in the preparation of the manuscript. Also, Erica N. Parker contributed to this manuscript through the synthesis, purification, and characterization of final analogues 1, 2, 4, 9, 11, 13, 20, 22, 31, 32, 33 and their corresponding intermediates. The author Jiangli Song contributed to this manuscript through the synthesis, purification, and characterization of final analogues 8, 14 and their corresponding intermediates. The author G.D.K. Kumar contributed to this manuscript through the synthesis, purification, and characterization of final analogues 1, 2, 8 and their corresponding intermediates. The author Ashleigh L. Barnes contributed to this manuscript through the synthesis, purification, and characterization of final analogue 10 and corresponding intermediates. Abbreviations used

HUVECs, human umbilical vein endothelial cells; ECM, extracellular matrix; TSC, thiosemicarbazide; SAR, structure-activity relationship; Z-FR-AMC, N-carbobenzyloxy- L-Phe-L-Arg-7-amino-4-methylcoumarin; FBS, fetal bovine serum; SRB, sulforhodamine B.

Keywords

Small-molecule synthesis, thiosemicarbazone warhead, cathepsin L inhibitors, inhibition of cancer cell invasion and migration, anti-metastatic agents

Note: Citations which appear outside of parenthesis correlate with the bibliography at the end of this chapter and references which appear inside of the parenthesis correlate with the bibliography at the end of the dissertation.

22 Abstract

Upregulation of cathepsin L in a variety of tumors and its ability to promote

cancer cell invasion and migration through degradation of the extracellular matrix

suggest that cathepsin L is a promising biological target for the development of anti-

metastatic agents. Based on encouraging results from studies on benzophenone

thiosemicarbazone cathepsin inhibitors, a series of fourteen benzoylbenzophenone

thiosemicarbazone analogues were designed, synthesized, and evaluated for their

inhibitory activity against cathepsins L and B. Thiosemicarbazone inhibitors 3-

benzoylbenzophenone thiosemicarbazone 1, 1,3-bis(4-fluorobenzoyl)benzene

thiosemicarbazone 8, and 1,3-bis(2-fluorobenzoyl)-5-bromobenzene thiosemicarbazone

32 displayed the greatest potency against cathepsin L with low IC50 values of 9.9 nM,

14.4 nM, and 8.1 nM, respectively. The benzoylbenzophenone thiosemicarbazone analogues evaluated were selective in their inhibition of cathepsin L compared to

cathepsin B. Thiosemicarbazone analogue 32 inhibited invasion through Matrigel of

MDA-MB-231 breast cancer cells by 70% at 10 µM. Thiosemicarbazone analogue 8

significantly inhibited the invasive potential of PC-3ML prostate cancer cells by 92% at 5

µM. The most active cathepsin L inhibitors from this benzoylbenzophenone

thiosemicarbazone series (1, 8, and 32) displayed low cytotoxicity toward normal

primary cells [in this case human umbilical vein endothelial cells (HUVECs)]. In an

initial in vivo study, 3-benzoylbenzophenone thiosemicarbazone (1) was well-tolerated in

a CDF1 mouse model bearing an implanted C3H mammary carcinoma, and showed

efficacy in tumor growth delay. Low cytotoxicity, inhibition of cell invasion, and in vivo

tolerability are desirable characteristics for anti-metastatic agents functioning through an

23 inhibition of cathepsin L. Active members of this structurally diverse group of

benzoylbenzophenone thiosemicarbazone cathepsin L inhibitors show promise as

potential anti-metastatic, pre-clinical drug candidates.

1. Introduction

In various human cancers cathepsins L, B, K, H, S, and X display increased

activity and play significant mechanistic roles in the invasion and migration of malignant

cells.1-5 (148,149,8,54,55) Cysteine protease cathepsins degrade proteolytic targets predominately within lysosomes under normal physiological conditions.1-5 (148,149,8,54,55)

Figure 2.1 Overview of cysteine cathepsins in invasion and metastasis. Compared to normal physiological conditions, certain cathepsins are upregulated in the tumor microenvironment. Additionally, changes occur within the tumor microenvironment such as downregulation of endogenous inhibitors including the cystatins, extracellular acidification increasing the proteolytic capability of cathepsins, and extracellular secretion of cathepsins. Degradation of the extracellular matrix through the direct proteolysis of matrix and cell-adhesion proteins such as laminin, fibronectin, collagen, E- cadherin facilitates tumor cell invasion from the primary tumor site and ultimately migration to a secondary tumor site. Small-molecule cathepsin inhibitors have the potential to inhibit the invasive nature of malignant cells and may provide increased effectiveness in combination with known chemotherapeutics.17 (151)

24 In cancer, cathepsins promote metastasis through phagocytosis as well as

extracellulary through multiple pathways including direct proteolysis of E-cadherin6 (150)

and components in the extracellular matrix (ECM) such as fibronectin,7-10 (73–76) laminin,8-

10 (74–76) collagen,9-15 (70,75–80) and indirectly through the activation of other proteases16 (81) resulting in the degradation of the ECM (Figure 2.1).

Elevated levels of cathepsin L, a ubiquitous endopeptidase, and cathepsin B, an endopeptidase as well as a carboxypeptidase, are associated with poor prognosis in breast cancer, colorectal cancer, lung cancer, brain tumors, melanoma, and head and neck cancer.18, 19 (152,153) Cathepsin K, an endopeptidase involved in bone resorption, shows

increased activity in breast cancer, cervical cancer, and lung cancer.20 (154) The cathepsin

K inhibitor Odanacatib (MK-0822) displayed promise in a recent clinical trial by

reducing bone resorption in individuals with metastatic bone disease.21-22 (155,156)

In addition to metastasis, cathepsins are involved in other pathological processes.

Drug candidates currently in the pipeline of pharmaceutical companies include the

cathepsin K inhibitors Odanacatib (Merck)21-24 (155,156,85,84) and MIV-711 (Medivir)25 (86)

for the treatment of osteoporosis, the cathepsin B inhibitor VBY-376 (Virobay)26-27 (87,88) and the pan cysteine protease inhibitor VBY-825 (Virobay)28-29 (96,97) for the treatment of

liver fibrosis, the cathepsin C inhibitor GSK2793660 (GlaxoSmithKline) for the

treatment of bronchiectasis,30-31 (82,83) and the cathepsin S inhibitor MIV-247 (Medivir)32

(157) for the treatment of neuropathic pain. Although selective inhibitors of cathepsin L have not yet reached clinical trials, several have been described in the literature (Figure

2.2).

25 Historically, molecules incorporating the thiosemicarbazone functionality have

been used as antiviral and antibacterial agents in the treatment of smallpox41-43 (161–163) and tuberculosis.44 (164)

Figure 2.2 Representative potent inhibitors of cathepsin L utilizing various warheads. Notable selective inhibitors of cathepsin L include the natural product gallinamide A I,33 (114) the epoxysuccinamide inhibitor Clik 148 II,34 (115) the oxocarbazate III,35 (158) the vinyl sulfonate IV,36 (117) the dipeptidyl nitrile V,37 (159) and the azepanone VI.38 (120) Potent but non-selective inhibitors of cathepsin L include the nonpeptidic cyanamide VII,39 (125) the azadipeptide nitrile VIII,40 (160) and the diketone VBY-825 IX.28-29 (96,97)

In the investigation of thiosemicarbazone inhibitors in our group45-50 (69,127–131) several cathepsin L inhibitors based on the benzophenone, thiochromanone, thiochromanone sulfone, and dihydroquinoline scaffolds including (3'-bromo-3- hydroxybenzophenone) thiosemicarbazone X,46-48 (69,128,129) (3'-bromo-2-

fluorobenzophenone) thiosemicarbazone XI, 45 (127) and 6-nitrothio-4-chromanone

thiosemicarbazone XVI49 (130) emerged as promising lead compounds (Figure 2.3).

26

Figure 2.3. Activity of selected benzophenone thiosemicarbazone analogues against cathepsin L

2. Results and discussion

2.1 Design and synthetic chemistry

Functional group considerations based on our initial benzophenone thiosemicarbazone inhibitors included the incorporation of a benzoyl moiety which, in

this study, led to the discovery that appropriately functionalized benzoylbenzophenone

thiosemicarbazone analogues demonstrated potent inhibition of cathepsin L. In addition

to incorporation of a benzoyl group, structural motifs previously identified in potent

benzophenone thiosemicarbazone inhibitors were integrated into the design of this new

series of benzoylbenzophenone thiosemicarbazone analogues.51

27 O O OH O c

R R R=H 3:R=H R=Bz

aorb a

S NH2 S NH2 NH NH O N OH N

R R 1:R=H 4:R=H 2: R=Bz

Scheme 2.1. Synthesis of benzoylbenzophenone thiosemicarbazone analogues. Reagents and conditions: (a) Thiosemicarbazide (TSC), TsOH, THF, reflux; (b) TSC, TsOH, o MeOH, reflux; (c) EtOH, NaBH4, 0 C.

Previously,45-46 (127,128) incorporation of the 3-bromo functionality in

benzophenone thiosemicarbazone analogues proved critical in providing analogues which

displayed potent inhibitory activity against cathepsin L as demonstrated by the large

differences in IC50 values found for X and XI compared to the non-brominated analogues

XIII and XIV, respectively (Figure 2.3). Additionally, the 4-bromo analogue XII displayed reduced activity towards cathepsin L compared to the 3-bromo analogue XI

further emphasizing the importance of the 3-bromo functionality. Drawing on previous

SAR studies based on the benzophenone scaffold,45-46 (127,128) extension of key structural

components of the potent cathepsin L benzophenone thiosemicarbazone inhibitors X and

XI led to the design of benzoylbenzophenone thiosemicarbazone inhibitors 31 and 32

which incorporated the 3-bromo functionality in the central aromatic ring. In order to

avoid potential complications that could arise due to the difficulty of separating

thiosemicarbazone regioisomers, only symmetrical diketones were utilized in the

28 synthesis of the new target compounds. Various synthetic methodologies (Schemes 2.1-

2.4) were employed to assemble each benzoylbenzophenone molecular scaffold.

Benzoylbenzophenone thiosemicarbazones 1, 2 and benzoylbenzhydrol

thiosemicarbazone 4 were prepared from commercially available diketones as illustrated

in Scheme 2.1.

Utilization of Friedel-Crafts acylation chemistry facilitated the synthesis of para

substituted benzoylbenzophenone analogues as depicted in Scheme 2.2. Reaction of

isophthaloyl dichloride and the appropriately substituted benzene ring with aluminum

chloride afforded para substituted 1,3-dibenzoylbenzene analogues 5-7 which underwent

condensation with thiosemicarbazide to yield target thiosemicarbazone analogues 8-11.

Demethylation of 1,3-bis(4-methoxybenzoyl)benzene 7 followed by condensation with

thiosemicarbazide afforded thiosemicarbazone 13 and reaction of diketone 12 with

isopropyl bromide followed by condensation with thiosemicarbazide afforded the p-

isopropoxy derivative 14.

Scheme 2.2. Synthesis of benzoylbenzophenone thiosemicarbazone analogues utilizing Friedel-Crafts acylation. Reagents and conditions: (a) AlCl3 (neat), reflux; (b) AlCl3, CH2Cl2, reflux; (c) TSC, TsOH, THF, microwave irradiation; (d) TSC, TsOH, THF, o reflux; (e) BF3·SMe2, CH2Cl2, rt; (f) isopropyl bromide, K2CO3, DMF, 90 C.

29 In order to incorporate meta substituents in the outermost rings of the benzoylbenzophenone scaffold (Scheme 2.3), isophthaloyl dichloride starting materials were used as precursors to Weinreb amides 16 and 17. Diketones 18 and 19 were synthesized from an intermediate organolithium reagent and Weinreb amides 16 and 17

(separately). Condensation of the resulting meta substituted 1,3-dibenzoylbenzene analogues with thiosemicarbazide and removal of any protecting groups afforded benzoylbenzophenone thiosemicarbazone analogues 20 and 22.

Scheme 2.3. Synthesis of benzoylbenzophenone thiosemicarbazone analogues utilizing isophthaloyl chloride. Reagents and conditions: (a) Me(OMe)NH ·HCl, NEt3, CH2Cl2, 0 oC -rt; (b) n-BuLi, THF, -78 oC; (c) THF, -78 oC; (d) TSC, TsOH, MeOH, reflux; (e) TSC, TsOH, THF, microwave irradiation, 90 oC; (f) TBAF, THF, rt.

In our previous studies, benzophenone thiosemicarbazone analogues45-48 (69,127–129)

containing a meta-bromo substituent were among those with the highest inhibitory activity against cathepsin L. Incorporation of meta-bromo substituents on the central aromatic ring in the benzoylbenzophenone scaffold was achieved by utilizing 1,3,5- tribromobenzene as a starting material. Halogen-metal exchange of 1,3,5- tribromobenzene with t-BuLi followed by the addition of two equivalents of either aldehyde 23 or Weinreb amides 24 and 25 (separately) allowed for the incorporation of a

30 meta-bromo substituent on the central ring of diketone analogues 27-29 (Scheme 2.4).

Subsequent condensation of the diketones 27-29 with thiosemicarbazide yielded the benzoylbenzophenone thiosemicarbazone analogues 30, 32-33, and deprotection of 30 afforded the m-hydroxy substituted analogue 31.

Scheme 2.4. Synthesis of benzoylbenzophenone thiosemicarbazone analogues utilizing 1,3,5-tribromobenzene. Reagents and conditions: (a) Me(OMe)NH ·HCl, NEt3, CH2Cl2, 0 oC -rt; (b) TBSCl, DMF, imidazole, 0 oC -rt; (c) (i) t-BuLi, ether, -78 oC; (ii) 23, 24, or 25 o o in ether, -78 C; (d) PCC, celite, CH2Cl2, 0 C -rt; (e) TSC, TsOH, THF, microwave o irradiation, 90 C; (f) TSC, TsOH, THF, reflux; (g) TSC, Ti(OiPr)4, THF, reflux (h) TBAF, THF, rt.

Moderate to low yields were observed for the condensation reaction to form the

mono thiosemicarbazone analogues due to the competing formation of the bis-

thiosemicarbazone products. For some of the desired mono-thiosemicarbazone products a

lower equivalency of thiosemicarbazide proved effective at minimizing formation of the

bis derivative and maximizing yield, but not in all cases. With the exception of 1,3-bis(2-

fluorobenzoyl)-5-bromobenzene thiosemicarbazone 32, which was isolated as the Z

isomer and did not isomerize after standing in acetone for one week, the final products

31 exist as an inseparable mixture of E/Z isomers in solution (As evidenced by 1H NMR).

Imines including thiosemicarbazones are well-known for their propensity to isomerize in solution under catalyzed62-63 (165,166) and non-catalyzed64-65 (167,168) conditions or from heating while in the solid state.66 (169)

Figure 2.4. Isomerization of thiosemicarbazone analogue 33 in DMSO-d6. The red ellipses indicate regions in the 1H NMR spectra where additional peaks emerge due to the presence of the other geometrical isomer. (a) 1H NMR of compound 33 after 0 hours in 1 1 DMSO-d6. (b) H NMR of compound 33 after 24 hours in DMSO-d6. (c) H NMR of compound 33 after 16 days in DMSO-d6.

As an example, benzoylbenzophenone thiosemicarbazone 33, after purification and drying, was isolated as a single isomer; however, the compound slowly isomerized in solution (Figure 2.4). After 16 days of standing in DMSO at room temperature, thiosemicarbazone 33 isomerized to a 1:1 mixture of E/Z isomers.

2.2 Cathepsin Inhibition Studies

The thiosemicarbazone derivative of commercially available 1,3- dibenzoylbenzene (3-benzoylbenzophenone thiosemicarbazone 1) displayed pronounced

32 inhibitory activity against cathepsin L with an IC50 value of 9.9 nM (Table 2.1).

Interestingly, the analogous benzophenone thiosemicarbazone XV (Figure 3) was

46 (127) inactive (IC50 >10000 nM) against cathepsin L. Since thiosemicarbazone 1

displayed pronounced activity against cathepsin L, several analogues were synthesized

including compounds which incorporated previously reported molecular scaffolds (Figure

2.3) known to be effective in terms of providing compounds with strong inhibitory

activity against cathepsin L.45-48 (69,127–129)

In addition, certain structural modifications of the unsubstituted analogue 1 such

as the incorporation of a structurally demanding benzoyl substituent on the central

aromatic ring, exemplified by tribenzoylbenzophenone thiosemicarbazone analogue 2

(IC50 = 56.0 nM), and the incorporation of a secondary alcohol in place of the carbonyl,

exemplified by 3-benzoylbenzhydrol thiosemicarbazone 4 (IC50 = 23.8 nM), were well

tolerated. Various para substituted analogues were evaluated against cathepsin L. The p-

fluoro analogue 8 had comparable activity to the extremely potent unsubstituted analogue

1. The activity against cathepsin L diminished with increasing steric hindrance in the

para-substituted series (p-hydroxy 13, p-methoxy 11, p-isopropoxy 14).

Thiosemicarbazone analogues 9, 22, and 33 each with p-bromo or m-bromo substituents on the outermost rings of the benzoylbenzophenone molecular template, although still active, showed a significant reduction in regard to inhibitory activity against cathepsin L compared to the other compounds. Additionally, the p-bromo substituted bis- thiosemicabazone analogue 10 showed no activity against cathepsin L at a concentration of 10,000 nM.

33 Table 2.1. Inhibitory activity of benzoylbenzophenone thiosemicarbazone analogues

a IC50 Values (nM)

Cmpd R1 R2 R3 R4 X Cat L Cat B 1 H H H H C=O 9.9 >10000

2 H H H Bz C=O 56.0 >10000 4 H H H H CH(OH) 23.8 >10000 8 F H H H C=O 14.4 >10000 9 Br H H H C=O 1522 >10000

10 Br H H H C=NNHC(S)NH2 >10000 >10000

11 OCH3 H H H C=O 5117 >10000 13 OH H H H C=O 340 >10000

14 OCH(CH3)2 H H H C=O >10000 >10000

20 H CH3 H H C=O 654 >10000 22 H Br H OH C=O ~10000b >10000 31 H OH H Br C=O 71.6 >10000 32 H H F Br C=O 8.1 >10000 33 H Br H Br C=O 10347 >10000 aThese values are averages of a minimum of a triplicate of experiments. Each assay utilized 2% DMSO with a 5 min pre-incubation period. Standard error values can be found in the Supplementary data. b Compound 22 inhibited cathepsin L activity by 56.9% at 10000 nM.

Compared to the parent benzophenone thiosemicarbazone XI (Figure 2.3) with an

IC50 value of 30.5 nM, 1,3-bis(2-fluorobenzoyl)-5-bromobenzene thiosemicarbazone 32

exhibited a greater than three fold increase in activity against cathepsin L with an IC50

34 value of 8.1 nM. Incorporation of the benzophenone thiosemicarbazone X (Figure 2.3)

molecular design into the benzoylbenzophenone scaffold led to two thiosemicarbazone

analogues; compound 31 with meta-hydroxy substituents on the outermost rings and

compound 22 with meta-bromo substituents on the outermost rings. Thiosemicarbazone

31 showed a nearly two fold increase in activity against cathepsin L with an IC50 value of

71.6 nM compared to the parent benzophenone thiosemicarbazone X (IC50 = 131.4 nM)

while analogue 22 showed a significant decrease in activity against cathepsin L with an

IC50 value of ~10000 nM. Overall, increasing steric bulk on the outermost rings likely resulted in benzoylbenzophenone analogues with decreased inhibitory activity.

Interestingly, all active benzoylbenzophenone thiosemicarbazone analogues were selective against cathepsin L compared to cathepsin B and thiosemicarbazone analogues

(31 and 32) of the parent benzophenone thiosemicarbazones X and XI (which do not have bulky groups on the outermost rings) displayed increased activity against cathepsin

L. These studies have demonstrated that the benzoylbenzophenone scaffold coupled with the thiosemicarbazone electrophilic moiety represents an effective molecular design leading to low nM inhibitors of cathepsin L and additionally allows for a variety of structural modifications in the core scaffold while maintaining activity against cathepsin

L.

Kinetic analysis of the closely related thiosemicarbazone analogue X (Figure 2.3) showed that it was a time-dependent and slowly reversible inhibitor of cathepsin L.47 (129)

The reaction progress curves for benzoylbenzophenone thiosemicarbazone analogues 1 and 32 (Figure 2.5) were comparable to those previously observed for (3'-bromo-3- hydroxybenzophenone) thiosemicarbazone X. The cathepsin L catalyzed reaction was

35 carried out with N-carbobenzyloxy-L-Phe-L-Arg-7-amino-4-methylcoumarin (Z-FR-

AMC) as substrate in the presence of varying concentrations (0–10 μM) of

thiosemicarbazone analogues 1 and 32.

Figure 2.5. Representative progress curves of cathepsin L (1 nM) activity using Z-FR- AMC (10 μM) as substrate and increasing concentrations of (A) inhibitor 1 and (B) inhibitor 32 (0–10 μM). Reactions were initiated by the addition of enzyme with no pre- incubation with compounds. Release of the fluorescent product AMC was followed as a function of time.

The progress curves (Figure 2.5) demonstrated that these compounds are not classical, readily reversible inhibitors. The increase in inhibition as a function of time, as observed from a reduction in the slopes of the curves over time compared to untreated control, indicated that both compounds are time-dependent inhibitors of cathepsin L.

Separate experiments demonstrated that analogues 1 and 32 were slowly reversible inhibitors of cathepsin L (see Supplementary data).

36 2.3 Molecular modeling studies

Figure 2.6. Molecular docking of analogues 1 and 32 within the active site of cathepsin L. (A) Analogue 1 and (B) analogue 32 are shown in ball and stick mode (C, gray; H, white; O, red; N, blue; S, yellow; F, light blue; Br, brown). Cathepsin L is shown in ribbon mode with a transparent molecular surface (Green) and enzyme active site amino acid residues are labeled and shown in stick mode (C, gray; H, white; O, red; N, blue; S, yellow).

Thiosemicarbazone analogues 1 and 32 which displayed the most pronounced

activity against cathepsin L were selected for molecular docking studies. The co-crystal

structure of cathepsin L with a nitrile inhibitor (PDB: 2XU1)67 (170) within the active site

of cathepsin L was chosen as a starting model for the docking studies which were

performed with Discovery Studio 4.1 (Accelrys). A number of different binding modes

with similar interaction energies were obtained from docking analogues 1 and 32. In each

case, several of the top binding orientations placed the thiocarbonyl carbon atom of the

inhibitor in close proximity to the Cys25 thiolate in a similar manner to that observed

with benzophenone thiosemicarbazones X47 (129) and XI.45 (127) One of the top binding

orientations for each of benzoylbenzophenone thiosemicarbazone analogues 1 and 32 which positions the Cys25 thiolate within 5 Å of the thiocarbonyl carbon atom is illustrated in Figure 2.6. For analogue 1, the benzophenone fragment of the inhibitor

37 resides deep within the S2 pocket of cathepsin L which is considered an important subsite for binding among the papain family of cysteine proteases.67-68 (18,170) The remaining outermost ring of analogue 1 is oriented towards the S3 subsite. A hydrogen bond between the carbonyl oxygen of the inhibitor and the backbone NH of His163 and a hydrogen bond between the terminal NH2 of the thiosemicarbazone and the backbone carbonyl of Asp162 is observed for analogue 1. Molecular docking studies for analogue

32 orient the aryl rings of the inhibitor within the active site of cathepsin L in a similar fashion to analogue 1 with the benzophenone fragment occupying the S2 pocket and the opposing ring oriented toward the S3 subsite. For comparison, molecular docking studies were carried out with analogue 14, which was inactive against cathepsin L. Molecular modeling indicated that analogue 14 did not bind in a manner that would facilitate formation of a covalent bond. The benzophenone portion resides in the S2 pocket of cathepsin L in a similar fashion observed for active cathepsin L inhibitors 1 and 32.

However, the remaining outermost ring resides in the S1 subsite instead of being oriented towards the S3 subsite and the thiosemicarbazone moiety is oriented towards the solvent instead of residing in the S1 subsite. In each of the top binding orientations, the thiocarbonyl carbon atom of analogue 14 was not in close proximity to the Cys25 thiolate ion of cathepsin L. Active cathepsin L inhibitors 1 and 32 bind in such a manner to facilitate formation of a transient covalent bond between the enzyme and inhibitor. This comparison emphasizes that formation of a transient covalent bond is necessary for activity against cathepsin L in this series of compounds (see Supplementary data).

38 2.4 Invasion and migration studies

A hallmark of invasive cancer cells is their ability to invade through the ECM.

Cell invasion through Matrigel as a model for the ECM69 (171) of the highly metastatic

prostate cancer cell line PC-3ML48,70 (69,172) and the highly invasive breast cancer cell line

MDA-MB-23171-72 (173,174) was inhibited by selected benzoylbenzophenone

thiosemicarbazone inhibitors. Additionally, selected thiosemicarbazone analogues were

evaluated for their ability to inhibit cell migration. To separate anti-tumorigenic from

anti-metastatic effects, non-cytotoxic doses of evaluated thiosemicarbazones were used.

The results showed that treatment with thiosemicarbazone analogue 8 significantly

impaired the invasive capacities of prostate PC-3ML cancer cells by 59% and 92% at 1

µM and 5µM, respectively (Figure 2.7). The unsubstituted analogue 1 inhibited cell invasion of PC-3ML prostate cancer cells by 37% and 53% at 10 µM and 25 µM, respectively.

Figure 2.7. Effect of cathepsin L inhibitors on tumor cell invasion. PC-3ML prostate cancer cell invasion in the presence of cathepsin inhibitors compound 1 (A) or compound 8 (B). Data are mean and standard error values of 3-5 independent experiments.

39

Figure 2.8. Inhibition of invasion and migration of MDA-MB-231 breast cancer cells by thiosemicarbazone analogues 1, 8, and 32. (a) Inhibition of MDA-MB-231 breast cancer cell invasion through Matrigel at 10 µM concentration of inhibitor. (b) Inhibition of MDA-MB-231 breast cancer cell invasion through Matrigel at 25 µM concentration of inhibitor. (c) Inhibition of MDA-MB-231 breast cancer cell migration at 10 µM concentration of inhibitor. (d) Inhibition of MDA-MB-231 breast cancer cell migration at 25 µM concentration of inhibitor. All values are normalized to 2% DMSO vehicle and are from a minimum of a triplicate of experiments (* p < 0.05, ** p < 0.01, *** p < 0.001).

Thiosemicarbazone 32 displayed the greatest effect on inhibition of invasion of

MDA-MB-231 breast cancer cells with 70 % inhibition at 10 µM, slightly better than

60% inhibition found for the irreversible pan cysteine protease inhibitor E-6473 (175) which was used as a positive control for direct comparison of the effectiveness of the inhibitors

(Figure 2.8). A significant change in inhibition of cell invasion was not observed for analogue 32 at 25 µM compared to 10 µM. However, the p-fluoro analogue 8 demonstrated an increase from 15% to 60% inhibition of invasion of MDA-MB-231 cells at 10 µM compared to 25 µM. The unsubstituted analogue 1, which inhibited cell

40 invasion by 30% at 25 µM, did not impact invasion of MDA-MB-231 cells as significantly as analogues 8 and 32.

All three benzoylbenzophenone analogues when evaluated at 10 µM and 25 µM concentrations, inhibited MDA-MB-231 cell migration; 3-benzoylbenzophenone thiosemicarbazone 1 showed the greatest effect with 80% inhibition of migration at 25

µM which represented a greater than 2-fold increase in inhibition compared with the positive control E-64 (Figure 2.8). In addition, 1,3-bis(2-fluorobenzoyl)-5-bromobenzene thiosemicarbazone 32 and 1,3-bis(4-fluorobenzoyl)benzene thiosemicarbazone 8 also inhibited migration greater than 50% at 25 µM.

2.5 Growth Inhibition of Mammary Carcinoma

Figure 2.9. 3-Benzoylbenzophenone thiosemicarbazone (1) effects on the growth of C3H mammary carcinomas implanted in the right rear foot of female CDF1 mice. Mice were given 5 daily intraperitoneal injections from the day of tumour implant (as indicated by the arrows). Results show means + 1 S.E. from 5-8 mice/group. [Legend:  Control (10% Tween80)  Analogue 1 (20 mg/kg)]

41 To investigate the effect of 3-benzoylbenzophenone thiosemicarbazone (1) (20

mg/kg) on tumor initiation, animals were observed on a daily basis and tumor volume

determined, as described in Figure 2.9, when tumors were measureable. From these data

3 the endpoint of TGT500 (time taken in days for tumors to reach 500 mm ) was

determined. For control animals injected with vehicle (10% Tween80) for 5 days from

the time of tumor implant the mean TGT500 (+ 1 S.E.) was found to be 20.1 days (+ 0.6

days) and this was significantly (Student’s T-test; significance level of p<0.05) increased

to 22.1 days (+ 0.5 days) when mice were given 5 daily treatments with 3-

benzoylbenzophenone thiosemicarbazone (1). This initial in vivo study demonstrated that analogue 1 was well tolerated in this mouse model and showed modest, statistically relevant efficacy in tumor growth delay. Cathepsin L inhibitors, functioning as anti- metastatic agents, are not anticipated to impart dramatic decreases in tumor growth when utilized as single agents; however, it is anticipated that their full value will be realized in combination therapy with standard chemotherapy and/or radiation.17 (151)

2.6 Cytotoxicity

Table 2.2. Evaluation of cytotoxicity against HUVECs

Compound Doxorubicin Paclitaxel 1 8 31 32

Cytotoxicity GI50 0.0268 0.00148 53.3 >126 13.3 >85.9 (µM)

Benzoylbenzophenone thiosemicarbazone analogues 1, 8, 31, and 32 were

evaluated for cytotoxicity against normal endothelial cells (HUVECs). Compared to

known chemotherapeutics, doxorubicin and paclitaxel, none of the compounds displayed

42 significant cytotoxicity against HUVECs (Table 2.2). Compounds 8 and 32 were the least toxic against HUVECs with GI50 values greater than 126 µM and 85.9 µM, respectively.

Low cytotoxicity to normal cells (HUVECs in this case) is a desirable feature since the

development of cathepsin L inhibitors as anti-metastatic agents would likely involve

chronic dosing to achieve efficacy.

3. Conclusions

Several benzoylbenzophenone thiosemicarbazone analogues were synthesized and

evaluated for their inhibitory activity against cathepsins L and B. Thiosemicarbazone

inhibitors 1, 8 and 32 displayed the highest activity against cathepsin L with low IC50 values of 9.9 nM, 14.4 nM and 8.1 nM, respectively. The benzoylbenzophenone thiosemicarbazone analogues represent a class of potent inhibitors that can tolerate structural changes and maintain activity against cathepsin L. The high versatility of the core molecular scaffold offers potential for optimization of the benzoylbenzophenone thiosemicarbazone analogues as inhibitors of cathepsin L. Both thiosemicarbazone analogues 1 and 32 demonstrated time-dependent inhibition of cathepsin L. Molecular docking studies of analogues 1 and 32 revealed that the benzophenone fragment of the benzoylbenzophenone thiosemicarbazone inhibitors is capable of filling the lipophilic S2 pocket of cathepsin L thus placing the thiosemicarbazone moiety in close proximity to the Cys25 thiolate. In contrast, molecular modeling of analogue 14 (inactive against cathepsin L) indicated that it did not bind to the enzyme in a conformation that would promote formation of a covalent bond. Kinetic studies combined with molecular docking studies indicate that the formation of a covalent bond between the enzyme and the most potent of this subset of inhibitors represents an important contribution to their efficacy.

43 The most potent inhibitor of cathepsin L from this series, 1,3-bis(2-fluorobenzoyl)-5-

bromobenzene thiosemicarbazone 32 significantly inhibited the ability of MDA-MB-231

cells to invade through Matrigel by 70%. Additionally, the p-fluoro analogue 8 inhibited

cell invasion of PC-3ML cells by 92% at 5 µM. 3-Benzoylbenzophenone

thiosemicarbazone (1) delayed growth in vivo of recently implanted tumors in a C3H mouse mammary carcinoma model. Cathepsin L inhibitors 1, 8, 31, and 32 demonstrated very low cytotoxicity toward HUVECs. The high potency against cathepsin L and ability to inhibit the invasion of tumor cells in vitro coupled with the desirable property of low

cytotoxicity to normal cells positions these compounds as viable pre-clinical candidates

for further development.

4. Experimental Section

4.1 Chemistry

4.1.1 General Synthetic Protocols.

All reactions were carried out under an inert atmosphere of nitrogen unless

otherwise noted. Anhydrous solvents were used for carrying out reactions. Unless

otherwise noted all reagents and solvents were purchased from commercial suppliers and

used without further purification. Hexanes used for column chromatography were

distilled from 4 Å molecular sieves prior to use. Anhydrous tetrahydrofuran and

dichloromethane were purchased from commercial suppliers or dried using a VAC

(Vacuum Atmospheres Co.) solvent purification system. Reactions were monitored by

SiliaPlate™ silica gel thin layer chromatography (TLC) plates (250 μm, F-254, 60 Å).

Manual flash chromatography and automated flash chromatography was carried out with

silica gel purchased from either Silicycle Inc. (230-400 mesh) or Biotage (40-65

44 microns). Proton (1H), Carbon (13C), and Fluorine (19F) NMR spectra were recorded

using a Varian Inova 500 MHz NMR system, Bruker Avance III HD 600 MHz NMR

system, or a Bruker Avance III HD 400 MHz NMR system. Chemical shifts are reported

in ppm (δ), coupling constants (J) are reported in hertz (Hz), and peak patterns are

reported as broad (br), singlet (s), doublet (d), triplet (t), quartet (q) and multiplet (m).

High resolution mass spectra (HRMS) were obtained in the Baylor University Mass

Spectrometry Core Facility on a Thermo Scientific LTQ Orbitrap Discovery using

electrospray ionization (ESI). Purity of final compounds was analyzed using an Agilent

Technologies 1200 series HPLC system with a Diode Array and Multiple Wavelength

Detector SL, equipped with a Zorbax reliance cartridge guard-column; Agilent Eclipse

XDB-C18 column (4.6 mm ID X 250 mm, 5 μm particle size, 80 Å pore size); T = 25 oC; flow rate 1.0 mL/min; injection volume 20 µL; monitored at 254 nm, 300 nm and 320 nm. Two HPLC methods were used in the purity analysis of final compounds: Method A: water:acetonitrile, gradient 50:50 to 10:90 from 0-25 min and isocratic 10:90 from 25-30 min ; Method B: water:acetonitrile, gradient 70:30 to 10:90 from 0-25 min and isocratic

10:90 from 25-30 min.

4.1.2 3-Benzoylbenzophenone thiosemicarbazone (1).51 (132)

p-Toluenesulfonic acid monohydrate (0.026 g, 0.136 mmol) was added to a

solution of 1,3-dibenzoylbenzene (2.00 g, 6.98 mmol) in anhydrous methanol (20 mL).

After stirring at reflux for 10 min, thiosemicarbazide (0.476 g, 5.235 mmol) was added to

the flask. After 26 h, methanol was removed under reduced pressure and water (50 mL)

was then added. The products were extracted with ethyl acetate (2 x 50 mL) and the

combined organic phases were washed with brine, dried over anhydrous sodium sulfate,

45 and the solvent was removed under reduced pressure. Purification using flash

chromatography (silica gel, hexanes:ethyl acetate, gradient 90:10 to 52:48) afforded 3- benzoylbenzophenone thiosemicarbazone (0.829 g, 5.24 mmol, 44% yield) as a light

1 yellow solid. H NMR (500 MHz, CDCl3): δ 8.71 (1H, s), 8.00-7.99 (1H, m), 7.85-7.83

(1H, m), 7.77-7.67 (3H, m), 7.60-7.29 (10H, m), 6.61-6.57 (1H, m). 13C NMR (125 MHz,

CDCl3): δ 195.97, 195.29, 179.04, 178.99, 149.85, 149.70, 139.13, 137.95, 136.99,

136.94, 136.70, 136.01, 132.97, 132.81, 132.28, 131.84, 131.47, 131.45, 131.34, 130.68,

130.56, 130.48, 130.11, 130.09, 130.05, 128.86, 128.62, 128.58, 128.39, 128.34, 127.76.

+ + HRMS (ESI) calculated for C21H17N3OSH (M+H) 360.11651, found 360.11667. HPLC retention time (Method A): 7.50 min. (Obtained as a mixture of E/Z isomers)

4.1.3 1,3,5-Trisbenzoylbenzene thiosemicarbazone (2). 51 (132)

1,3,5–Tribenzoylbenzene (2.34 g, 6.00 mmol) (purified by flash chromatography

before using) and p-toluenesulfonic acid monohydrate (11.4 mg, 0.06 mmol) were

dissolved in anhydrous THF (20 mL) and heated to reflux. Thiosemicarbazide (546 mg,

6.00 mmol) was then added to the flask and the reaction mixture was stirred at reflux for

35 h. The reaction mixture was concentrated under reduced pressure and the products

were extracted from water (100 mL) with ethyl acetate (2 x 50 mL). The combined

organic phases were washed with brine, dried over anhydrous sodium sulfate, and the

solvent was removed under reduced pressure. Purification using flash chromatography

(silica gel, hexanes: dichloromethane: ethyl acetate, gradient 80:10:10 to 40:30:30)

afforded 1,3,5-trisbenzoylbenzene thiosemicarbazone (0.961 g, 2.07 mmol, 35% yield) as

1 a yellow solid. H NMR (500 MHz, acetone-d6): δ 9.40 (0.2H, s), 8.72 (0.8H, s), 8.33

(.2H, t, J = 1.5 Hz), 8.28 (1.6H, d, J = 1.5 Hz), 8.21 (0.8H, s), 8.13 (0.2H, s), 8.07 (0.8H,

46 t, J = 1.5 Hz), 8.05 (0.4H, d, J = 1.5 Hz), 7.96-7.95 (0.8H, m), 7.85-7.84 (3.2H, m), 7.78

13 (0.8H, s), 7.73-7.52 (10.4 H, m), 7.43-7.37 (0.8H, m). C NMR (125 MHz, acetone-d6): δ

194.47, 194.33, 180.08, 179.66, 147.62, 147.43, 139.25, 138.02, 137.94, 136.77, 136.74,

136.68, 133.68, 133.01, 132.99, 132.59, 131.83, 131.62, 131.37, 130.94, 130.47, 130.06,

130.05, 130.00, 129.99, 129.90, 128.67, 128.62, 128.53, 128.42, 127.85. X-ray

crystallographic data obtained for 1,3,5-trisbenzoylbenzene thiosemicarbazone has been

deposited in the Cambridge Crystallographic Data Centre and was allocated the

+ deposition number CDCC 1042567. HRMS (ESI) calculated for C28H21O2N3SH

(M+H)+ 464.14272, found 464.14280. HPLC retention time (Method A): 10.53, 10.81 min. (Obtained as a mixture of E/Z isomers)

[Note: The use of 0.8H and 0.2H for 1H NMR integral values relate to isomer ratios. This

system is use throughout.]

4.1.4 3-benzoylbenzyhdrol (3). 51,74 (132,176)

1,3-Dibenzoylbenzene (2.00 g, 6.98 mmol) was dissolved in anhydrous ethanol

(20 mL). The reaction mixture was cooled to 0 oC followed by the addition of sodium

borohydride (0.090g, 2.094 mmol). The reaction mixture was stirred for 4 h and

quenched by the addition of a small amount of water. The reaction mixture was

concentrated under reduced pressure and the products were extracted from water with

ethyl acetate (2 x 50 mL). The combined organic phases were washed with brine, dried

over anhydrous sodium sulfate, and the solvent was removed under reduced pressure.

Purification using flash chromatography (silica gel, hexanes: ethyl acetate, gradient 93:7

to 30:70) afforded 3-benzoylbenzhydrol (0.601 g, 2.084 mmol, 30% yield). 1H NMR

(500 MHz, acetone-d6): δ 7.90 (1H, t, J = 1.7 Hz), 7.77-7.74 (2H, m), 7.72-7.69 (1H, m),

47 7.67-7.62 (2H, m), 7.55-5.53 (2H, m), 7.51-7.48 (1H, m), 7.46-7.44 (2H, m), 7.34-7.30

(2H, m), 7.25-7.21 (1H, m), 5.95 (1H, s), 5.04 (1H, brs). 13C NMR (125 MHz, acetone- d6): δ 196.47, 146.94, 146.03, 138.59, 139.40, 133.24, 131.36, 130.58, 129.24, 129.23,

+ + 129.11, 128.54, 127.96, 127.36, 75.71. HRMS (ESI) calculated for C20 H16O2H (M+H)

289.12231, found 289.12266.

4.1.5 3-Benzoylbenzhydrol thiosemicarbazone (4). 51 (132)

p-Toluenesulfonic acid monohydrate (0.007 g, 0.03 mmol) was added to a

solution of 3-benzoylbenzyhdrol (0.396 g, 1.09 mmol) in anhydrous tetrahydrofuran (10

mL). After stirring at reflux for 10 min, thiosemicarbazide (0.199 g, 2.19 mmol) was

added to the reaction mixture. After 2 days, tetrahydrofuran was removed under reduced

pressure and water (50 mL) was added. The products were extracted with ethyl acetate (2 x 20 mL) and the combined organic phases were washed with brine, dried over anhydrous sodium sulfate, and the solvent was removed under reduced pressure.

Purification using flash chromatography (silica gel, hexanes:ethyl acetate, gradient 88:12 to 0:100) afforded 3-benzoylbenzhydrol thiosemicarbazone (0.228 g, 0.631 mmol, 57%

1 yield) as a white solid. H NMR (500 MHz, acetone-d6): δ 8.51 (0.5H, brs), 8.50 (0.5H,

brs), 8.08 (0.5H, brs), 8.00 (0.5H, brs), 7.82 (0.5H, t, J = 1.7 Hz), 7.74-7.61 (4.5H m),

7.49-7.19 (10H, m), 5.97 (0.5H, d, J = 3.9 Hz), 5.82 (0.5H, d, J = 3.9 Hz), 5.10 (0.5H, d,

13 J = 3.9 Hz), 4.87 (0.5H, d, J = 3.9 Hz). C NMR (125 MHz, acetone-d6): δ 179.44,

179.41, 149.48, 149.42, 147.57, 145.86, 145.21, 144.96, 136.76, 136.61, 131.66, 131.35,

130.05, 129.81, 129.72, 129.70, 128.46, 128.29, 128.28, 128.19, 128.07, 127.96, 127.61,

127.07, 126.89, 126.55, 126.39, 126.35, 126.24, 125.55, 74.99, 74.48. HRMS (ESI)

48 + + calculated for C21H19N3OSNa (M+Na) 384.11410, found 384.11447. HPLC retention time (Method B): 11.73, 12.32. (Obtained as a mixture of E/Z isomers)

4.1.6 1,3-Bis(4-fluorobenzoyl)benzene (5).51,75 (132,177)

Aluminum trichloride (6.53 g, 49.5 mmol) was added to a solution of

isophthaloyl dichloride (5.00 g, 24.8 mmol) in dichloromethane (100 mL). After heating

at reflux for 30 min, fluorobenzene (3.41 g, 37.3 mmol) was added. After stirring for 12

h, the reaction mixture was poured onto crushed ice, neutralized with 10% NaOH (100

mL), and extracted with dichloromethane (3 × 100 mL). The combined organic layers

were washed with water, followed by brine and dried over anhydrous sodium sulfate.

After the organic layer was concentrated under reduced pressure, purification using flash

chromatography (silica gel, hexanes: ethyl acetate, 60:40) afforded 1,3-bis-(4-

fluorobenzoyl)benzene (3.37 g, 10.5 mmol, 56% yield) as a white solid. 1H NMR (500

MHz, CDCl3): δ 8.13 (1H, t, J = 1.7 Hz), 8.00 (2H, dd, J = 7.7 Hz, 1.7 Hz), 7.87 (4H, dd,

13 J = 8.9 Hz, 5.4 Hz), 7.64 (1H, t, J = 7.7 Hz), 7.18 (4H, m). C NMR (125 MHz, CDCl3):

δ 194.24, 165.65 (d, J = 255 Hz), 137.81, 133.32, 133.17 (d, J = 3.2 Hz), 132.71 (d, J =

19 9.3 Hz), 130.79, 128.66, 155.75 (d, J = 22 Hz). F NMR (470 MHz, CDCl3): δ 104.91

+ + (1F, tt, J = 8.3 Hz, 5.4 Hz). HRMS (ESI) calculated for C20H12F2O2H (M+H)

323.08781, found 323.08817.

4.1.7 1,3-Bis(4-bromobenzoyl)benzene (6).51,76 (132,178)

To a flask containing aluminum trichloride (1.36 g, 10.2 mmol) under nitrogen

was added bromobenzene (15 mL, 142.8 mmol). Isophthaloyl dichloride (1.00 g, 4.88

mmol) was dissolved in a minimal amount of bromobenzene and added to the reaction

flask. The reaction stirred at reflux for 6 h, after which it was stirred at room temperature

49 for 12 h, followed by another 3 h at reflux. The reaction was quenched with water (20

mL). The resulting mixture was then added to 1M HCl (50 mL) cooled in an ice bath.

The mixture was extracted with ethyl acetate (3 × 10 mL) and the combined organic

layers were washed sequentially with deionized water, dilute HCl, deionized water, and

brine. Crude product crashed out of the organic layers as a white solid and was collected

via filtration. Recrystallization of the solid from ethyl acetate afforded 1,3-bis(4- bromobenzoyl)benzene (0.671 g, 1.51 mmol, 31% yield). 1H NMR (500 MHz, DMSO-

d6): δ 8.04 (2H, dd, J = 7.8 Hz, 1.7 Hz), 7.98 (t, 1H, J = 1.7 Hz), 7.82-7.78 (4H, d, J =

8.6 Hz), 7.77 (t, 1H, J = 7.7 Hz), 7.74-7.71 (4H, d, J = 8.6 Hz). 13C NMR (125 MHz,

DMSO-d6): δ 194.08, 136.78, 135.52, 133.51, 131.75, 131.72, 130.44, 129.24, 121.10.

+ + HRMS (ESI) calculated for C20H12Br2O2H (M+H) 442.92768, found 442.92792.

4.1.8 1,3-Bis(4-methoxybenzoyl)benzene (7).51,77 (132,179)

Aluminum trichloride (5.60 g, 42.0 mmol) was added to a solution of isophthaloyl

dichloride (4.00 g, 19.7 mmol) in dichloromethane (50 mL) and heated to reflux for 30

min. Anisole (2.58 mL, 23.7 mmol) was added and the reaction mixture was refluxed for

20 h. The reaction mixture was poured onto the crushed ice. The resulting solution was

neutralized with 10% NaOH (100 mL), and extracted with dichloromethane (3 × 100

mL). The combined organic layers were washed with water, followed by brine, and dried

over anhydrous sodium sulfate. After the organic layer was concentrated under reduced

pressure, the purification using flash chromatography (silica gel, hexanes: ethyl acetate,

60:40) afforded 1,3-bis-(4-methoxy benzoyl)benzene (2.60 g, 11.9 mmol, 63% yield) as

1 a white solid. H NMR (500 MHz, CDCl3): δ 8.09 (1H, td, J = 1.7 Hz, 0.4Hz), 7.95 (2H, dd, J = 7.7 Hz, 1.7 Hz), 7.84 (4H, d, J = 8.9 Hz), 7.61 (1H, td, J = 7.7 Hz, 0.4 Hz), 6.97

50 13 (4H, d, J = 8.9 Hz), 3.88 (s, 6H). C NMR (125 MHz, CDCl3): δ 194.71, 163.50, 138.36,

132.72, 132.60, 130.61, 129.62, 128.38, 113.74, 55.54. HRMS (ESI) calculated for

+ + C22H18O4H (M+H) 347.12779, found 347.12817.

4.1.9 1,3-Bis(4-fluorobenzoyl)benzene thiosemicarbazone (8).51(132)

1,3-Bis-(4-fluorobenzoyl)benzene (0.347 g, 1.08 mmol) was dissolved in

anhydrous methanol (50 mL). The solution was heated to reflux for 15 min followed by

the addition of thiosemicarbazide (0.049 g, 0.54 mmol) and a catalytic amount of p-

toluenesulfonic acid monohydrate (0.020 g, 0.11 mmol). After 10 h at reflux, the solution

was concentrated under reduced pressure. Purification using flash chromatography (silica

gel, hexanes: ethyl acetate, 70:30) afforded the desired product (0.110 g, 0. 278 mmol,

1 52% yield) as a white solid. H NMR (500 MHz, CDCl3): δ 8.66 (1H, brs), 7.99-7.97

(0.5H, m), 7.95 (0.5H, m), 7.90 (1H, dd, J = 8.9 Hz, 5.4 Hz), 7.83 (1H, dd, J = 8.9 Hz,

5.4 Hz), 7.77-7.73 (1H, m), 7.69-7.66 (1H, m), 7.54-7.52 (0.5H, m), 7.51-7.46 (1.5H, m),

7.38 (1H, brs), 7.32-7.30 (2H, m), 7.23-7.20 (1H, m), 7.18-7.14 (1H, m), 7.07-7.04 (1H,

13 m), 6.35 (1H, brs). C NMR (125 MHz, CDCl3): δ 194.44, 193.71, 179.16, 179.08,

165.74 (d, J = 256 Hz), 165.63 (d, J = 255 Hz), 164.16 (d, J = 252 Hz), 163.74 (d, J =

253 Hz), 148.67, 139.19, 138.02, 137.00, 133.31 (d, J = 3.0 Hz), 132.90 (d, J = 3.0 Hz),

132.81 (d, J = 9.2 Hz), 132.71 (d, J = 9.1 Hz), 132.28, 132.19 (d, J = 3.2 Hz), 131.83,

131.37, 131.26, 130.73 (d, J = 8.5 Hz ), 130.34, 129.82, 129.75 (d, J = 8.6 Hz), 128.59,

128.49, 126.44 (d, J = 3.7 Hz), 117.56 (d, J = 22 Hz), 115.84 (d, J = 22 Hz), 115.81 (d, J

19 = 22 Hz), 115.65 (d, J = 22 Hz). F NMR (565 MHz, CDCl3): δ -104.48 (0.5 F, tt, J =

8.3 Hz, 5.4 Hz), -104.80 (0.5 F, tt, J = 8.3 Hz, 5.4 Hz), -108.02 (0.5 F, tt, J = 8.0 Hz, 5.5

+ Hz), -109.13 (0.5 F, tt, J = 8.2 Hz, 5.4 Hz). HRMS (ESI) calculated for C21H15F2N3OSH

51 (M-H)+ 396.09767, found 396.09741. HPLC retention time (Method A): 8.09, 8.37 min.

(Obtained as a mixture of E/Z isomers)

4.1.10 1,3-Bis(4-bromobenzoyl)benzene thiosemicarbazone (9).

1,3-Bis(4-bromobenzoyl)benzene (106 mg, 0.238 mmol) was dissolved in

anhydrous ethanol (2 mL) followed by the addition of p-toluenesulfonic acid

monohydrate (1.0 mg, 0.005 mmol) and thiosemicarbazide (21.7 mg, 0.238 mmol). The

reaction was carried out at 100 oC for 30 min under microwave irradiation. The solvent

was removed under reduced pressure and the crude reaction mixture was purified using

flash chromatography (silica gel, hexanes: dichloromethane: ethyl acetate, gradient,

50:50:0 to 40:50:10) to afford 1,3-bis(4-bromobenzoyl)benzene thiosemicarbazone (17.5

1 mg, 0.034 mmol, 14% yield). H NMR (500 MHz, DMSO-d6): δ 9.45 (0.5H, s), 9.11

(0.5H, s), 8.58 (0.5H, s), 8.56 (0.5H, s), 8.37 (0.5H, s), 8.32 (0.5H, s), 8.05 (0.5H, d, J =

8.1 Hz), 7.91 (0.5H, m), 7.82-7.69 (5H, m), 7.66-7.53 (5H, m), 7.34-7.31 (1H, m). 13C

NMR (125 MHz, DMSO-d6): δ 194.37, 194.28, 146.81, 146.80, 137.78, 136.86, 136.82,

135.70, 135.68, 135.58, 132.98, 132.69, 131.90, 131.72, 131.66, 131.64, 131.51, 131.32,

131.28, 130.91, 130.76, 130.56, 130.45, 130.12, 130.00, 129.58, 128.70, 128.48, 127.04,

+ + 126.97, 123.52, 123.29. HRMS (ESI) calculated for C21H15Br2N3OSNa (M+Na)

537.91948, found 537.91980. HPLC retention time (Method A): 13.78, 14.50 min. The

purity for the mono thiosemicarbazone product (9) is less than 95%. The major impurity is the bis-thiosemicarbazone product (10) which was inactive against cathepsin L and B.

The combined mono and bis thiosemicarbazone products are 94.8% at 254 nm and

greater than 95% at 320 nm (254 nm: mono thiosemicarbazone (9) 88.21% and bis

52 thiosemicarbazone (10) 6.56%. 320 nm: mono thiosemicarbazone (9) 93.99% and bis thiosemicarbazone (10) 5.63%). (Obtained as a mixture of E/Z isomers)

4.1.11 1,3-Bis(4-bromobenzoyl)benzene bis-thiosemicarbazone (10).51 (132)

1,3-Bis(4-bromobenzoyl)benzene (0.550 g, 1.21 mmol) was dissolved in dry THF

(20 mL). The solution was heated at reflux for 15 min, and thiosemicarbazide (0.220 g,

2.42 mmol) and a catalytic amount of p-toluenesulfonic acid monohydrate (0.023 g,

0.121 mmol) were added to the reaction mixture. After 24 h at reflux, the result solution

was concentrated under reduced pressure and the residue was dissolved in

dichloromethane (25 mL). The organic layer was washed with deionized water (15 mL),

dried over anhydrous sodium sulfate, and concentrated. The crude product was purified

via column chromatography to give the double condensation product in <1% isolated

yield (6.7 mg, 11.3 µmol). The 1H NMR for the major isomer is reported. The 13C NMR for the isomer mixture is reported. Three isomers exist for the bis thiosemicarbazones.1H

NMR (500 MHz, DMSO-d6): δ 9.87 (2H, s), 8.46 (2H, s), 8.29 (2H, s), 7.84 (1H, t, J =

7.5 Hz), 7.64 (4H, d, J = 8.5 Hz), 7.56 (4H, d, J = 7.5 Hz), 7.48 (2H, dd, J = 7.5 Hz, 1.5

13 Hz), 7.20 (1H, s). C NMR (125 MHz, DMSO-d6): δ 178.73, 147.42, 146.67, 146.45,

137.67, 136.03, 135.55, 133.37, 132.76, 132.36, 131.25, 131.17, 130.90, 130.76, 130.25,

129.93, 129.86, 129.56, 129.50, 129.43, 129.38, 129.32, 126.59, 123.53, 123.27, 123.09.

X-ray crystallographic data obtained for 1,3,5-trisbenzoylbenzene thiosemicarbazone has been deposited in the Cambridge Crystallographic Data Centre and was allocated the

+ deposition number CDCC 1046061. HRMS (ESI) calculated for C22H18Br2N6S2Na

(M+Na)+ 610.92933, found 610.92822. HPLC retention time (Method A): 10.19, 11.76,

12.16 min. (Obtained as a mixture of isomers)

53 4.1.12 1,3-Bis(4-methoxybenzoyl)benzene thiosemicarbazone (11).

1,3-Bis(4-methoxybenzoyl)benzene (0.346 g, 1.00 mmol), thiosemicarbazide

(0.091 g, 1.00 mmol) and p-toluenesulfonic acid monohydrate (0.010 g, 0.050 mmol)

were dissolved in tetrahydrofuran (10 mL) and the reaction was carried out at 90 oC for

45 min under microwave irradiation. The solvent was removed under reduced pressure

and the crude reaction mixture was purified using flash chromatography (silica gel,

hexanes: ethyl acetate, gradient, 88:12 to 40:60) to afford 1,3-bis(4-

methoxybenzoyl)benzene thiosemicarbazone (0.189 mg, 0.410 mmol, 45% yield). 1H

NMR (500 MHz, CDCl3): δ 8.807 (0.6H, s), 8.667 (0.4H, s), 7.973-7.952 (1H, m), 7.869

(0.8H, d, J = 8.8 Hz), 7.813 (1.2H, d, J = 8.9 Hz), 7.741-7.700 (1H, m), 7.677-7.654 (1H, m), 7.501 (0.4H, dt, J = 7.7 Hz, 1.5 Hz), 7.460-7.393 (2.4H, m), 7.245 (1.2H, d, J = 8.8

Hz), 7.082 (1.2H, d, J = 8.8 Hz), 7.008 (0.8H, J = 8.9 Hz), 6.958 (1.2H, d, J = 8.9 Hz),

6.864 (0.8H, d, J = 8.9 Hz), 6.353 (1H, s), 3.897 (1.8H, s), 3.894 (3H, s), 3.827 (1.2H, s).

13 C NMR (125 MHz, CDCl3): δ 194.85, 194.11, 178.87, 178.69, 163.64, 163.50, 161.55,

161.11, 150.18, 149.93, 139.83, 138.65, 137.24, 132.68, 132.61, 131.80, 131.49, 131.33,

131.15, 131.10, 130.07, 130.02, 129.78, 129.61, 129.39, 129.31, 128.71, 128.56, 128.20,

122.39, 115.48, 114.00, 113.93, 113.65, 55.58, 55.54, 55.50, 55.43. HRMS (ESI)

+ + calculated for C23H21N3O3SNa (M+Na) 442.11958, found 442.11960. HPLC retention

time (Method A): 7.21, 7.62 min (Obtained as a mixture of E/Z isomers)

4.1.13 1,3-Bis(4-hydroxybenzoyl)benzene (12).51,78 (132,180)

To a well-stirred solution of 1,3-bis-(4-methoxybenzoyl)benzene (1.80 g, 5.20

mmol) in dichloromethane (85 mL) was added boron trifluoride dimethyl sulfide

complex (BF3·SMe2, 15 mL). The mixture was stirred for 27 h. After the reaction was

54 quenched by water, the mixture was extracted with ethyl acetate (3 × 80 mL). The

combined organic layer was washed with brine, dried over anhydrous sodium sulfate, and

concentrated under vacuum. The resulting solid was further purified using flash

chromatography (silica gel, hexanes: ethyl acetate, 50:50) to afford the pure product

1 (0.620 g, 1.95 mmol, 38% yield) as a white solid. H NMR (500 MHz, acetone-d6): δ

9.28 (2H, s), 8.04 (1H, td, J = 1.7 Hz, 0.4 Hz), 7.98 (2H, dd, J = 7.7 Hz, 1.7 Hz), 7.80

(4H, m), 7.73 (1H, td, J = 7.7 Hz, 0.4 Hz), 7.00 (4H, m). 13C NMR (125 MHz, acetone-

d6): δ 194.46, 162.74, 139.45, 133.49, 133.13, 130.96, 129.71, 128.38, 116.11. HRMS

+ + (ESI) calculated for C20H14O4H (M+H) 319.09649, found 319.09671.

4.1.14 1,3-Bis(4-hydroxybenzoyl)benzene thiosemicarbazone (13).

1,3-Bis(4-hydroxybenzoyl)benzene (0.318 g, 1.00 mmol) was dissolved in

anhydrous ethanol (10 mL) followed by the addition of p-toluenesulfonic acid

monohydrate (0.005 g, 0.025 mmol) and thiosemicarbazide (0.910 g, 1.00 mmol). The

reaction was carried out at 100 oC for 45 min under microwave irradiation. The solvent

was removed under reduced pressure and the crude reaction mixture was purified using

flash chromatography (silica gel, dichloromethane: ethyl acetate, gradient, 90:10 to

60:40) to afford 1,3-bis(4-hydroxybenzoyl)benzene thiosemicarbazone (0.160 mg, 0.410

1 mmol, 50% yield). H NMR (600 MHz, DMSO-d6): δ 10.49 (0.4H, s), 10.48 (0.6H, s),

10.08 (0.6H, s), 9.90 (0.4H, s), 9.03 (0.4H, s), 8.62 (0.6H, s), 8.56 (0.6H, s), 8.45 (0.4H,

s), 8.40 (0.6H, s), 8.17 (0.4H, s), 8.06 (0.6H, d, J = 7.9 Hz), 7.85 (0.4H, dt, J = 7.8 Hz,

1.4 Hz), 7.78-7.74 (1.8H, m), 7.65-7.63 (1.8H, m), 7.56-7.51 (1H, m), 7.49-7.46 (1.2H, m), 7.20 (1.2H, d, J = 8.5 Hz), 7.00 (1.2H, d, J = 8.5 Hz), 6.91 (0.8H, d, J = 8.7 Hz), 6.87

13 (1.2H, d, J = 8.7 Hz), 6.75 (0.8H, d, J = 8.8 Hz). C NMR (150 MHz, DMSO-d6): δ

55 193.87, 193.65, 177.88, 177.72, 162.23, 162.19, 159.27, 158.78, 148.80, 148.71, 139.04,

138.27, 137.11, 132.83, 132.66, 131.96, 130.50, 130.22, 130.10, 130.03, 129.84, 129.50,

129.33, 128.40, 128.27, 127.60, 127.59, 127.31, 120.89, 116.57, 115.39, 115.31, 115.2.

+ + HRMS (ESI) calculated for C21H17N3O3SNa (M+Na) 414.08828, found 414.08841.

HPLC retention time (Method B): 6.33, 6.75 min. Analysis by 1H NMR shows that

compound 13 contains 1.42 wt% ethyl acetate (6 mol% ethyl acetate). Ethyl acetate was found to have an IC50 value >>10000 nM. (Obtained as a mixture of E/Z isomers)

4.1.15 1,3-Bis(4-isopropoxybenzoyl)benzene (intermediate for compound 14).51 (132)

Reactions were conducted using a commercially available microwave reactor

(Biotage). In a microwave vial, 1,3-bis-(4-hydroxybenzoyl)benzene (0.310 g, 0.975

mmol), isopropyl bromide (0.851 g, 6.92 mmol) and potassium carbonate (0.955 g, 6.91

mmol) were added to DMF (10 mL), with a magnetic stir bar. The vial was capped tightly

and the reaction mixture was heated from r.t. to 90 oC for 2 h. After the reaction mixture

was cooled to room temperature, the vial was opened and the mixture was transferred to a round bottom flask. The mixture was quenched with water (50 mL) and extracted with ether (2 × 50 mL). The organic layer was washed with brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude product was purified

using flash chromatography (silica gel, hexanes: ethyl acetate, 50:50) to afford the pure

product (0.23 g, 0.57 mmol, 59% yield) as a white solid. 1H NMR (500 MHz, acetone-

d6): δ 8.07 (1H, s), 7.99 (2H, dd, J = 7.6 Hz, 1.4 Hz), 7.84 (4H, d, J = 8.7 Hz), 7.71 (1H, t, J = 7.6 Hz), 7.05 (4H, d, J = 8.7), 4.76 (2H, Septet, J = 6.0 Hz), 1.35 (12H, d, J = 6.0

13 Hz). C NMR (125 MHz, acetone-d6): δ 193.51, 162.03, 138.41, 132.40, 130.25, 129.22,

56 + + 128.57, 115.07, 69.96, 21.33. HRMS (ESI) calculated for C26H26O4H (M+H)

403.19039, found 403.19055.

4.1.16 1,3-Bis(4-isopropoxybenzoyl)benzene thiosemicarbazone (14).51 (132)

1,3-Bis-(4-isopropoxybenzoyl)benzene (0.14 g, 0.39 mmol) was dissolved in anhydrous methanol (20 mL). The solution was heated at reflux for 15 min followed by the addition of thiosemicarbazide (0.043 g, 0.47 mmol) and a catalytic amount of p- toluenesulfonic acid monohydrate. After 10 h at reflux, the solvent was removed under vacuum and the resulting solid was further purified using flash chromatography (silica gel, hexanes: ethyl acetate, 50:50) to afford the desired product as a white solid (0.050

1 g, 0. 105 mmol, 27% yield). H NMR (500 MHz, CDCl3): δ 8.84 (0.6H, brs), 8.66 (0.4H,

brs), 7.96-7.94 (1H, m), 7.84 (0.8H, d, J = 8.9 Hz), 7.78 (1.2H, d, J = 8.9 Hz), 7.74-7.65

(2H, m), 7.49 (0.4H, m), 7.45-7.40 (2.4H, m), 7.21 (1.2H, d, J = 8.8 Hz), 7.03 (1.2H, d, J

= 8.8 Hz), 6.98-6.95 (0.8H, d, J = 8.9 Hz), 6.91 (1.2H, d, J = 8.9 Hz), 6.83 (0.8H, d, J =

8.9 Hz), 6.46 (1H, brs), 4.69-4.54 (2H, m), 1.40 (3.6H, d, J = 6.1 Hz), 1.38-1.37 (6H, m),

13 1.33 (2.4H, d, J = 6.1 Hz). C NMR (125 MHz, CDCl3): δ 194.80, 194.08, 178.87,

178.66, 162.19, 162.04, 159.98, 159.59, 150.32, 150.04, 139.91, 138.71, 137.28, 132.74,

132.66, 131.72, 131.40, 131.34, 131.09, 131.03, 130.09, 129.97, 129.77, 129.41, 129.15,

128.84, 128.79, 128.17, 128.16, 121.94, 116.79, 115.53, 115.263, 114.98, 70.21, 70.19,

+ + 70.17, 70.03, 22.01, 21.91. HRMS (ESI) calculated for C27H29N3O3SH (M+H)

476.20024, found 476.20059. HPLC retention time (Method A): 14.82, 15.31 min

(Obtained as a mixture of E/Z isomers)

57 4.1.17 5-(tert-butyldimethylsilyloxy)isophthalic acid (intermediate for compound 15).79

(181)

3-hydroxyisophthalic acid (5.46 g, 30.0 mmol), tert-butyldimethylchlorosilane

(22.50 g, 150.0 mmol), and imidazole (12.24 g, 180.0 mmol) were dissolved in N,N-

dimethylformamide (150 mL) and the reaction mixture was stirred for 11 h between 50-

60 oC. The reaction mixture was diluted with water (150 mL) and extracted with hexanes

(4 x 100 mL). The organic phase was dried over sodium sulfate and the solvent was

removed under reduced pressure. The crude product was dissolved in tetrahydrofuran (30

mL) followed by the addition of glacial acetic acid (90 mL), and distilled water (30 mL).

After stirring for 3 h at room temperature, the reaction mixture was diluted with distilled

water (60 mL) and cooled to 0 oC. The precipitated product was filtered. The filtrate was

concentrated and cooled again to 0 oC. The precipitated product was filtered and the

combined solids were dried to yield 5-(tert-butyldimethylsilyloxy)isophthalic acid (6.55 g,

1 22.1 mmol ,74% yield). H NMR (600 MHz, DMSO-d6): δ 13.34 (2H, s), 8.10 (1H, t, J =

1.5 Hz), 7.56 (2H, d, J = 1.5 Hz), 0.96 (9H, s), 0.22 (6H, s). 13C NMR (150 MHz,

DMSO-d6): δ 166.28, 155.35, 132.80, 124.46, 123.21, 25.49, 17.98, -4.60. HRMS (ESI)

+ + calculated for C14H20O5SiNa (M+Na) 319.09722, found 319.09741.

4.1.18 5-(tert-butyldimethylsilyloxy)isophthaloyl chloride (15). 79 (181)

Oxalyl chloride (2.04 mL, 27.8 mmol) was added dropwise to a solution of 5-

(tert-butyldimethylsilyloxy)isophthalic acid (2.82 g, 9.51 mmol) in dichloromethane (90

mL). N,N-Dimethylformamide (0.073 mL, 0.095 mmol) was added dropwise. The

reaction mixture was allowed to stir until the solution was transparent. After 2 h, the

solvent was removed under reduced pressure. The product was dissolved in anhydrous

58 dichloromethane (20 mL) and the solvent was removed under reduced pressure. This was

repeated two times to afford 5-(tert-butyldimethylsilyloxy)isophthaloyl chloride as yellow

solid. The product was used immediately without further purification. 1H NMR (500

MHz, CDCl3): δ 8.46 (1H, t, J = 1.6 Hz), 7.82 (2H, d, J = 1.6 Hz), 1.02 (9H, s), 0.28 (6H,

13 s). C NMR (125 MHz, CDCl3): δ 167.21, 156.95, 135.55, 128.34, 126.72, 25.67, 18.39,-

4.30.

4.1.19 N1,N3-dimethoxy-N1,N3-dimethyl isophthalamide (16).51(132)

Triethylamine (5.61 mL, 40.0 mmol) was added dropwise to a solution of N,O- dimethylhydroxylamine hydrochloride (2.93 g, 30.0 mmol) in anhydrous dichloromethane (35 mL) at 0 oC. After stirring for 10 min, a solution of isophthaloyl

dichloride (2.03 g, 10 mmol) dissolved in anhydrous dichloromethane (6 mL) was added

dropwise. The reaction mixture was returned to room temperature and stirred for 5 h. The

reaction was quenched with water (50 mL) and the products were extracted with

dichloromethane (2 x 50 mL). The combined organic phases were washed with brine,

dried over anhydrous sodium sulfate, and the solvent was removed under reduced

pressure. Purification using flash chromatography (silica gel, dichloromethane:methanol,

95:5) afforded N1,N3-dimethoxy-N1,N3-dimethyl isophthalamide (2.47 g, 9.77 mmol, 97%

1 yield). H NMR (500 MHz, CDCl3): δ 7.98 (1H, s), 7.76 (2H, dd, J = 7.8 Hz, 1.6 Hz),

13 7.44 (1H, t, J = 7.8 Hz), 3.53 (6H, s), 3.35 (6H, s). C NMR (125 MHz, CDCl3): δ

169.15, 133.96, 130.55, 128.20, 128.05, 61.27, 33.79. HRMS (ESI) calculated for

+ + C12H16N2O4H (M+H) 253.11828, found 253.11859.

59 4.1.20 5-((tert-butyldimethylsilyl)oxy)-N1,N3-dimethoxy-N1,N3-dimethyl isophthalamide

(17).

Triethylamine (5.35 mL, 38.1 mmol) was added dropwise to a solution of N,O- dimethylhydroxylamine hydrochloride (2.78 g, 28.5 mmol) in dichloromethane (50 mL) at 0 oC. A solution of 5-(tert-butyldimethylsilyloxy)isophthaloyl chloride in

dichloromethane (10 mL) was added dropwise and the reaction was allowed to return to

room temperature. After 6 h, the reaction was quenched with water (90 mL) and extracted

with dichloromethane (3 x 60 mL). The combined organic phases were dried over

anhydrous sodium sulfate and the solvent was removed under reduced pressure.

Purification using flash chromatography (silica gel, dichloromethane:methanol, gradient,

100:0 to 90:10) afforded 5-((tert-butyldimethylsilyl)oxy)-N1,N3-dimethoxy-N1,N3-

dimethyl isophthalamide as a white solid (3.40 g, 8.89 mmol, 93% yield over two steps).

1 H NMR (500 MHz, DMSO-d6): δ 7.38 (1H, t, J = 1.5 Hz), 7.15 (2H, d, J = 1.5 Hz), 3.52

13 (6 H, s), 3.26 (6H, s), 0.96 (9H, s), 0.21 (6H, s). C NMR (125 MHz, DMSO-d6): δ

167.52, 154.16, 135.50, 120.98, 120.26, 60.80, 25.45, 17.94, -4.66. HRMS (ESI)

+ + calculated for C18H30 N2O5SiH (M+H) 383.19968, found 383.20117.

4.1.21 1,3-Bis(3-methylbenzoyl)benzene (18).51,80 (132,182)

3-Bromotoluene (6.684 g, 39.08 mmol) was dissolved in anhydrous tetrahydrofuran (30 mL). The solution was stirred for 5 min and cooled to -78 oC followed by the dropwise addition of n-butyllithium in hexanes (2.5 M, 13.6 mL). After stirring for 3.5 h, a solution of N1,N3-dimethoxy-N1,N3-dimethyl isophthalamide

tetrahydrofuran (5 mL) was added dropwise and the solution was stirred for 1 h at -78 oC.

The reaction mixture was quenched with 1 M HCl (40 mL) and the products were

60 extracted with dichloromethane (2 x 50 mL). The combined organic phases were washed

with aqueous saturated aqueous sodium bicarbonate, dried over anhydrous sodium

sulfate, and the solvent was removed under reduced pressure. Purification using flash

chromatography (silica gel, hexanes:ethyl acetate, gradient, 95:5 to 70:30) afforded 1,3- bis(3-methylbenzoyl)benzene (2.47 g, 9.77 mmol, 72% yield). 1H NMR (500 MHz,

CDCl3): δ 8.17-8.15 (1H, m), 8.02 (2H, dd, J = 7.7 Hz, 1.7 Hz), 7.66-7.61 (3H, m), 7.59

(2H, d, J = 7.5 Hz), 7.42 (2H, d, J = 7.5 Hz), 7.37 (2H, t, J = 7.5 Hz), 2.43 (6H, s). 13C

NMR (125 MHz, CDCl3): δ 192.22, 138.55, 138.06, 137.20, 133.79, 133.54, 131.30,

+ + 130.59, 128.63, 128.40, 127.55, 21.53. HRMS (ESI) calculated for C22H18O2H (M+H)

315.13796, found 315.13833.

4.1.22 1,3-Bis(3-bromobenzoyl)-5-tert-butyldimethylsilyloxybenzene (19).

n-Butyllithium in hexanes (2.5M, 4.0 mL, 10 mmol) was added dropwise to a

solution of 1,3-dibromobenzne (2.53 mL, 20.9 mmol) in tetrahydrofuran (50 mL) cooled

to -78 oC. After 30 min, a solution of 5-((tert-butyldimethylsilyl)oxy)-N1,N3-dimethoxy-

N1,N3-dimethyl isophthalamide (2.00g, 5.23 mmol) in tetrahydrofuran (10 mL) was added

dropwise to the reaction mixture. After stirring for 2 h, the reaction mixture was

quenched with 1 M HCl (30 mL). The product was extracted with ethyl acetate (3 x 50

mL). The combined organic phases were dried over anhydrous sodium sulfate and the

solvent was removed under reduced pressure. Purification using flash chromatography

(silica gel, hexane:ethyl acetate, gradient, 98:2 to 90:10) afforded 1,3-bis(3-

bromobenzoyl)-5-tert-butyldimethylsilyloxybenzene as a yellow oil (1.63 g, 2.84 mmol,

1 57% yield). H NMR (500 MHz, CDCl3): δ 7.95 (2H, t, J = 1.7 Hz), 7.75-7.70 (4H, m),

7.68 (1H, t, J = 1.5 Hz), 7.45 (2H, d, J = 1.5 Hz), 7.38 (2H, t, J = 7.8 Hz), 1.0 (9H, s),

61 13 0.26 (6H, s). C NMR (125 MHz, CDCl3): δ 193.76, 156.14, 138.75, 138.57, 135.77,

132.76, 130.04, 128.50, 125.17, 124.18, 122.79, 25.80, 18.24, -4.35. HRMS (ESI)

+ + calculated for C26H26Br2 O3SiH (M+H) 573.00907, found 573.00909.

4.1.23 1,3-Bis(3-methylbenzoyl)benzene thiosemicarbazone (20).51(132)

p-Toluenesulfonic acid monohydrate (0.001 g, 0.01 mmol) was added to a

solution of 1,3-bis(3-methylbenzoyl)benzene (0.154 g, 0.480 mmol) in anhydrous methanol (10 mL). After stirring at reflux for 10 min, thiosemicarbazide (0.022 g, 0.24 mmol) was added to the reaction mixture and stirred for 7 h under an inert atmosphere of nitrogen gas. After 7 h, methanol was removed under reduced pressure and 10 mL of water was then added. The products were extracted with ethyl acetate and the combined

organic phases were washed with brine, dried over anhydrous sodium sulfate, and the

solvent was removed under reduced pressure. Purification using flash chromatography

(silica gel, hexanes:ethyl acetate, gradient 90:10 to 40:60) afforded 1,3-bis(3-

methylbenzoyl)benzene thiosemicarbazone (0.057 g, 0.015 mmol, 64% yield) as a

1 yellow solid. H NMR (500 MHz, DMSO-d6): δ 9.17 (0.4H, s), 8.66 (0.6H, s), 8.55

(0.4H, s), 8.48 (0.6H, s), 8.43 (0.6H, s), 8.27 (0.4H, s), 8.21 (0.6H, d, J = 7.8 Hz), 7.93

(0.4H, dt, J = 7.8 Hz, 1.5 Hz), 7.81 (0.4H, t, J = 7.6 Hz), 7.74-7.71 (1.2 H, m), 7.64-7.39

(7H, m), 7.33-7.30 (0.4 H, m), 7.27-7.17 (2H, m), 2.40 (1.8 H, s), 2.38 (1.2H, s), 2.36

13 (1.8H, s), 2.31 (1.2H, s). C NMR (125 MHz, DMSO-d6): δ 195.32, 195.31, 148.25,

148.14, 139.47, 138.18, 138.08, 137.98, 137.63, 137.27, 136.74, 136.70, 136.59, 136.34,

133.58, 133.51, 132.61, 131.91, 130.93, 130.78, 130.76, 130.64, 130.60, 130.46, 130.02,

129.99, 129.92, 129.75, 129.73, 128.66, 128.62, 128.50, 128.37, 128.21, 127.83, 127.18,

127.01, 125.27, 125.16, 20.97, 20.93, 20.88, 20.82. HRMS (ESI) calculated for

62 + + C23H21N3OSNa (M+Na) 410.12975, found 410.13013. HPLC retention time (Method

A): 11.38 min. (Obtained as a mixture of E/Z isomers)

4.1.24 1,3-Bis(3-bromobenzoyl)-5-tert-butyldimethylsilyloxybenzene thiosemicarbazone

(21).

1,3-Bis(3-bromobenzoyl)-5-tert-butyldimethylsilyloxybenzene (1.39 g, 2.43

mmol), thiosemicarbazide (0.166 g, 1.82 mmol), and p-toluenesulfonic acid monohydrate

(0.010 g, 0.05 mmol) were dissolved in tetrahydrofuran (15 mL). The reaction was carried out at 90 oC for 30 min under microwave irradiation. The solvent was removed

under reduced pressure and the crude reaction mixture was purified using flash

chromatography (silica gel, hexanes:ethyl acetate, gradient, 93:7 to 50:50) to afford 1,3-

bis(3-bromobenzoyl)-5-tert-butyldimethylsilyloxybenzene thiosemicarbazone (0.418 g,

1 0.650 mmol, 36% yield). H NMR (500 MHz, DMSO-d6): δ 9.35 (0.7H, s), 9.12 (0.3H,

s), 8.57 (1H, m), 8.44 (0.7H, s), 8.34 (0.3H, s), 8.04 (0.7H, t, J = 1.7 Hz), 7.94 (0.7H, t, J

= 1.7 Hz), 7.89-7.85 (1H, m), 7.83-7.80 (1H, m), 7.78 (0.3H, ddd, J = 8Hz, 2Hz, 1Hz),

7.70-7.68 (0.3H, m), 7.62-7.60 (0.3H, t, J = 1.7 Hz), 7.59-7.57 (1H, m), 7.56-7.48 (1.3H, m), 7.47-7.44 (0.7H, m), 7.39 (0.3H, dt, J = 7.6 Hz, 1.2 Hz), 7.33-7.29 (1.7H, m), 7.20

(0.7H, t, J = 1.5 Hz), 7.13-7.11 (1H, m), 0.96 (6H, s), 0.91 (3H, s), 0.24 (4H, s), 0.15(2H,

13 s). C NMR (125 MHz, DMSO-d6): δ 193.42, 193.38, 178.53, 155.99, 155.02, 146.05,

145.55, 139.32, 138.75, 138.66, 138.65, 138.44, 138.02, 135.57, 135.51, 133.66, 132.97,

132.90, 132.28, 132.02, 131.85, 131.69, 131.26, 130.80, 130.72, 130.36, 129.48, 128.90,

128.68, 127.68, 126.89, 124.47, 123.04, 122.84, 122.75, 122.12, 122.07, 121.85, 121.82,

121.80, 121.48, 25.52, 25.51, 18.00, 17.96, -4.59. HRMS (ESI) calculated for

+ + C27H29Br2N3O2SSiH (M+H) 646.01893, found 646.01995.

63 4.1.25 1,3-Bis(3-bromobenzoyl)-5-hydroxybenzene thiosemicarbazone (22).

1,3-Bis(3-bromobenzoyl)-5-hydroxybenzene (0.388 g, 0.600 mmol) was

dissolved in tetrahydrofuran (5 mL) followed by the dropwise addition of a 1M solution

of tetra-butylammonium fluoride in tetrahydrofuran (1.20 mL, 1.20 mmol) at room temperature. After 1 h, the reaction mixture was diluted with ethyl acetate (30 mL) and washed with brine (15 mL). The combined organic phases were dried over anhydrous sodium sulfate and the solvent was removed under reduced pressure. Purification using flash chromatography (silica gel, gradient, hexane:ethyl acetate: methanol, 77.5:20:2.5 to

57.5:40:2.5) afforded 1,3-bis(3-bromobenzoyl)-5-hydroxybenzene thiosemicarbazone as

1 a yellow solid (0.277 g, 0.520 mmol, 87% yield). H NMR (500 MHz, DMSO-d6): δ

10.39 (0.6H, s), 9.98 (0.4H, s), 9.24 (0.6H, s), 9.07 (0.4H, s), 8.62-8.54 (1H, m), 8.46

(0.6H, s), 8.28 (0.4H, s), 8.09 (0.6H, s), 7.94 (0.6H, t, J = 1.8 Hz), 7.88-7.84 (1H, m),

7.83-7.79 (1H, m), 7.77 (0.4H, ddd, J = 8.1 Hz, 2.0 Hz, 1.0 Hz), 7.69-7.66 (0.4H, m),

7.61-7.47 (2.4 H, m), 7.44 (0.6H, d, J = 7.8 Hz), 7.39-7.35 (0.8H, m), 7.34-7.27 (1.6H, m), 7.12 (0.4H, dd, J = 2.2 Hz, 1.0 Hz), 7.00-6.99 (0.6H, m), 6.97 (0.6H, dd, J = 2.2 Hz,

13 1.5 Hz). C NMR (125 MHz, DMSO-d6): δ 193.74, 193.73, 178.41, 158.46, 157.38,

146.50, 146.10, 139.12, 138.98, 138.91, 138.58, 138.16, 137.65, 135.43, 135.33, 133.81,

132.85, 132.59, 132.32, 131.91, 131.72, 132.71, 131.17, 130.75, 130.65, 130.40, 129.36,

128.86, 128.66, 127.71, 127.04, 122.82, 122.17, 121.87, 121.82, 120.40, 119.89, 119.52,

+ + 118.49, 117.60, 117.29. HRMS (ESI) calculated for C21H15Br2N3O2SNa (M+Na)

553.91439, found 553.91436. HPLC retention time (Method B): 15.75, 16.88 min.

(Obtained as a mixture of E/Z isomers)

64 4.1.26 3-(tert-butyldimethylsilyloxy)benzaldehyde (23).81 (182)

3-Hydroxybenzaldehyde (2.00 g, 16.4 mmol) was dissolved in N,N-

dimethylformamide (50 mL) followed by the addition of imidazole (2.23 g, 32.8 mmol).

The reaction mixture was cooled to 0 oC and tert-butyldimethylchlorosilane (3.69 g, 24.6

mmol) was added. The reaction mixture was returned to room temperature and stirred for

3 h. After reaction completion, the reaction mixture was quenched with saturated aqueous

sodium bicarbonate (50 mL) and the product was extracted with hexanes (2 X 50 mL).

The organic extracts were dried over anhydrous sodium sulfate and concentrated under

reduced pressure. The crude mixture was purified using flash chromatography (silica gel,

hexanes:ethyl acetate, gradient, 100:00 to 90:10) to afford 3-(tert-

butyldimethylsilyloxy)benzaldehyde (3.57, 15.1 mmol , 92% yield). 1H NMR (500 MHz,

CDCl3): δ 9.95 (1H, s), 7.47 (1H, dt, J = 7.3 Hz, 1.2 Hz), 7.40 (1H, t, J = 7.8 Hz), 7.34-

7.32 (1H, m), 7.12-7.09 (1H, m), 1.0 (9H, m), 0.23 (6H, m). 13C NMR (125 MHz,

CDCl3): δ 192.23, 156.54, 138.07, 130.21, 126.68, 123.69, 120.01, 25.76, 18.34, -4.28.

+ + HRMS (ESI) calculated for C13H20O2SiH (M+H) 237.13053, found 237.13066.

4.1.27 2-Fluoro-N-methoxy-N-methyl-benzamide (24).51,82 (132,183)

Triethylamine (5.32 mL, 37.8 mmol) was added dropwise to a solution of N,O- dimethylhydroxylamine hydrochloride (2.77 g, 28.4 mmol) in anhydrous dichloromethane (45 mL) at 0 oC. After stirring for 10 min, 2-flurobenzoyl chloride

(0.303 mL, 2.52 mmol) in anhydrous dichloromethane (15 mL) was added dropwise. The

reaction mixture was returned to room temperature and stirred for 5 h. The reaction

mixture was quenched with water (60 mL) and the products were extracted with

dichloromethane (2 x 60 mL). The combined organic phases were washed with brine,

65 dried over anhydrous sodium sulfate, and the solvent was removed under reduced

pressure. Purification using flash chromatography (silica gel, hexanes:ethyl acetate, gradient 93:7 to 60:40) afforded 2-Fluoro-N-methoxy-N-methyl-benzamide (3.12 g, 17.0

1 mmol, 90% yield). H NMR (500 MHz, CDCl3): δ 7.44-7.38 (2H, m), 7.19 (1H, t, J = 7.5

Hz), 7.10 (1H, t, J = 8.9 Hz), 3.55 (3H, brs), 3.35 (3H, brs). 13C NMR (125 MHz,

CDCl3): δ 166.40, 158.66, (d, J = 249 Hz), 131.50, 128.90, 124.11, 123.52 (d, J = 17 Hz),

19 - 115.69 (d, J = 21 Hz), 61.21, 32.31. F NMR (470 MHz, CDCl3): δ 114.04 (1F, s).

+ + HRMS (ESI) calculated for C9H10FNO2H (M+H) 184.07683, found 184.07702.

4.1.28 3-Bromo-N-methoxy-N-methyl-benzamide (25).83 (184)

To a well stirred suspension of N,O-dimethylhydroxylamine hydrochloride (5.33

g, 54.7 mmol) in dichloromethane (120 mL) was added triethylamine (10.2 mL, 72.9

mmol) dropwise at 0 oC. After stirring for a few minutes, 3-bromobenzoyl chloride (4.81

mL, 36.4 mmol) in dichloromethane (20 mL) was added dropwise. The reaction mixture

was allowed to warm to room temperature and stirred for 4 h. The reaction mixture was

quenched with water (150 mL) and extracted with dichloromethane (3 x 100 mL). The

combined organic phases were dried over anhydrous sodium sulfate and concentrated.

Purification using flash chromatography (silica gel, hexane: ethyl acetate, gradient, 90:10

to 60:40) afforded 3-bromo-N-methoxy-N-methyl-benzamide as a colorless oil (8.60 g,

1 35.2 mmol, 97% yield). H NMR (500 MHz, CDCl3): 7.83 (1H, t, J = 1.8 Hz), 7.61 (1H,

dt, J = 7.7 Hz, 1.3 Hz), 7.59 (1H, ddd, J = 8.0 Hz, 2.1 Hz, 1.1 Hz), 7.28 (1H, m), 3.35

13 (3H, s), 3.36 (3H, s). C NMR (125 MHz, CDCl3): 168.32, 136.06, 133.69, 131.36,

+ 129.74, 126.93, 122.14, 61.33, 33.71. HRMS (ESI) calculated for C9H10BrNO2H

(M+H)+ 243.99677, found 243.99704.

66 4.1.29 1,3-Bis[(3-tert-butyldimethylsilyloxyphenyl)hydroxymethyl]-5-bromobenzene (26).

Tert-butyllithium in pentane (1.7M, 11.76 mL) was added dropwise to a solution of 1,3,5 tribromobenzene (1.57 g, 5.00 mmol) in anhydrous diethyl ether (20 mL) cooled to -78 oC. The mixture was sonicated at every 30 min interval. After 2 h, a solution of 3-

(tert-butyldimethylsilyloxy)benzaldehyde (2.37 g, 10.0 mmol) in diethyl ether (10 mL) was added dropwise and the reaction mixture was allowed to slowly warm to room temperature over a period of 19 h. The reaction was quenched with 1 M HCl (30 mL) and extracted with diethyl ether (3 X 30 mL). The combined organic extracts were washed with saturated aqueous sodium bicarbonate, followed by brine and dried over anhydrous sodium sulfate. After removing the solvent under reduced pressure, the crude mixture was purified using flash chromatography (silica gel, hexanes:ethyl acetate, gradient,

100:00 to 40:60) to afford 1,3-bis[(3-tert-butyldimethylsilyloxyphenyl)hydroxymethyl]-

1 5-bromobenzene (1.16 g, 1.85 mmol, 37% yield). H NMR (600 MHz, CDCl3): δ 7.411-

7.390 (2H, m), 7.357 (0.5H, s), 7.327 (0.5H, s), 7.206-7.169 (2H, m), 6.905-6.881 (2H, m), 6.836-6.881 (2H, m), 6.768-6.741 (2H, m), 5.707-5.704 (2H, m), 2.196 (2H, s), 0.964

13 (9H, s), 0.963 (9H, s), 0.168 (6H, s), 0.166 (6H, s). C NMR (150 MHz, CDCl3): δ

155.89, 145.94, 145.89, 144.59, 144.56, 129.68, 129.67, 128.62, 128.51, 123.21, 122.71,

122.65 119.58, 119.56, 119.45, 119.43, 118.27, 118.23, 75.39, 25.68, 18.21, -4.40.

+ + HRMS (ESI) calculated for C32H45BrO4Si2Na (M+Na) 651.19320, found 651.19128.

4.1.30 1,3-Bis[(3-tert-butyldimethylsilyloxy)benzoyl]-5-bromobenzene (27).

1,3-Bis[(3-tert-butyldimethylsilyloxyphenyl)hydroxymethyl]-5-bromobenzene

(1.10 g, 1.75 mmol) in dichloromethane (5 mL) was added dropwise to a solution of pyridinium chlorochromate (1.13 g, 5.24 mmol), celite (1.00 g), and dichloromethane (5

67 mL) at 0 oC. The reaction mixture was returned to room temperature. After 4.5 h, the

reaction mixture was filtered over a pad of celite. The celite was rinsed several times with

dichloromethane and the filtrate was concentrated. The solvent was removed under

reduced pressure and the crude reaction mixture was purified using flash chromatography

(silica gel, hexanes:ethyl acetate, gradient, 90:08 to 20:80) to afford 1,3-bis[(3-tert-

butyldimethylsilyloxy)benzoyl]-5-bromobenzene (1.02 g, 1.63 mmol, 93% yield). 1H

NMR (500 MHz, CDCl3): δ 8.12 (2H, d, J = 1.5 Hz), 8.08 (1H, t, J = 1.5 Hz), 7.38-7.34

(4H, m), 7.26-7.25 (2H, m), 7.11-7.08 (2H, m), 0.98 (18H, s), 0.21 (12H, s). 13C NMR

(125 MHz, CDCl3): δ 193.91, 155.94, 139.51, 137.70, 135.96, 129.68, 129.51, 125.21,

123.25, 122.65, 121.14, 25.63, 18.22, -4.39. HRMS (ESI) calculated for

+ + C32H41BrO4Si2H (M+H) 625.17995, found 625.17969.

4.1.31 1,3-Bis(2-fluorobenzoyl)-5-bromobenzene (28).51 (132)

Tert-butyllithium in pentane (1.6M, 7.72 mL) was added dropwise to a solution of

1,3,5-tribromobenzene (0.972 g, 3.09 mmol) in anhydrous ether (30 mL) at -78 oC under a flow of nitrogen gas. After 2 h, 2-fluoro-N-methoxy-N-methyl-benzamide (1.132 g,

6.18 mmol) dissolved in anhydrous ether (5 mL) was added dropwise and the reaction

mixture was allowed to slowly come to room temperature and stirred for 24 h. After 24

h, the reaction mixture was quenched with water (30 mL) and the products were extracted

with ether (2 x 50 mL). The combined organic phases were washed with brine, dried

over anhydrous sodium sulfate, and the solvent was removed under reduced pressure.

Purification using flash chromatography (silica gel, hexanes:ethyl acetate, gradient 95:5

to 60:40) afforded 1,3-bis(3-fluorobenzoyl)-5-bromobenzene (0.617 g, 6.18 mmol, 50%

1 yield). H NMR (500 MHz, CDCl3): δ 8.16 (2H, m), 8.12 (1H, pentet, J = 1.4 Hz), 7.61-

68 7.56 (4H, m), 7.30 (2H, td, J = 7.5 Hz, 1.0Hz), 7.20-7.16 (2H, m). 13C NMR (125 MHz,

CDCl3): δ 191.23, 160.40 (d, J = 254 Hz), 139.67, 133.61, 134.29 (d, J = 8.6 Hz), 131.12

(d, J = 1.9 Hz), 129.31, 125.79 (d, J = 13.8 Hz), 124.79 (d, J = 3.8 Hz), 123.18, 116.70

19 - - (d, J = 21 Hz). F NMR (470 MHz, CDCl3): δ 109.73 - 109.78 (1F, m). HRMS (ESI)

+ + calculated for C20H11 BrF2O2H (M+H) 400.99833, found 400.99857.

4.1.32 1,3-Bis(3-bromobenzoyl)-5-bromobenzene (29).

Tert-butyllithium (1.7 M, 11.8 mL) was added dropwise to a solution of 1,3,5- tribromobenzene in diethyl ether (25 mL) at -78o C. The mixture was sonicated for 0.5

min at 20 min intervals to help dissolve the starting material. After 1 h, a solution of 3-

bromo-N-methoxy-N-methyl-benzamide (2.44 g, 10.0 mmol) in tetrahydrofuran (15 mL) was added to the reaction mixture and allowed to slowly come to room temperature overnight. The reaction was quenched with 1 M HCl (30 mL) and extracted with diethyl ether (3 x 30 mL). The combined organic phases were washed with saturated aqueous sodium bicarbonate and concentrated. Crystallization of the crude mixture in acetone followed by recrystallization in ethanol afforded 1,3-bis(3-bromobenzoyl)-5- bromobenzene as a white solid (1.77 g, 0.338 mmol, 68% yield). 1H NMR (500 MHz,

DMSO-d6): δ 8.17 (2H, d, J = 1.5 Hz), 7.96 (2H, t, J = 1.7 Hz), 7.92-7.98 (3H, m), 7.81-

13 7.79 (2H, m), 7.55 (2H, t, J = 7.9 Hz). C NMR (125 MHz, DMSO-d6): δ 192.28,

138.60, 138.18, 135.88, 135.54, 131.98, 130.86, 129.61, 128.93, 122.31, 122.04. HRMS

+ + (ESI) calculated for C20H11 Br3O2H (M+H) 520.83819, found 520.83820.

69 4.1.33 1,3-Bis[(3-tert-butyldimethylsilyloxy)benzoyl]-5-bromobenzene thiosemicarbazone

(30).

1,3-Bis[(3-tert-butyldimethylsilyloxy)benzoyl]-5-bromobenzene (0.890 g, 1.42

mmol ), thiosemicarbazide (0.970 g, 1.06 mmol) and p-toluenesulfonic acid monohydrate

(0.135 g, 0.0710 mmol) were dissolved in tetrahydrofuran (10 mL) and the reaction was

carried out at 90 oC for 30 min under microwave irradiation. The solvent was removed

under reduced pressure and the crude reaction mixture was purified using flash

chromatography (silica gel, hexanes:ethyl acetate, gradient, 90:10 to 20:80) to afford 1,3-

bis[(3-tert-butyldimethylsilyloxy)benzoyl]-5-bromobenzene thiosemicarbazone as a

1 yellow solid (0.329 g, 0.471 mmol , 33% yield). H NMR (500 MHz, CDCl3): δ 8.77

(1H, s), 7.87 (2H, d, J = 1.7 Hz), 7.83 (1H, t, J = 1.7 Hz), 7.48-7.44 (1H, m), 7.37-7.32

(2H, m), 7.30 (1H, dt, J = 7.6 Hz, 1.5 Hz), 7.23-7.22 (1H, m), 7.08 (1H, ddd, J = 7.8 Hz,

2.5 Hz, 1.5 Hz), 7.02 (1H, ddd, J = 8.3 Hz, 2.5 Hz, 1.0 Hz), 6.86 (1H, dt, J = 7.6 Hz, 1.0

Hz), 6.71-6.70 (1H, m), 6.44 (1H, s), 0.99 (9H, s), 0.98 (9H, s), 0.23 (6H, s), .22 (6H, s).

13 C NMR (125 MHz, CDCl3): δ 194.25, 179.26, 157.34, 156.02, 148.09, 139.76, 138.89,

137.90, 134.01, 133.88, 131.74, 131.25, 129.70, 127.55, 125.27, 123.35, 122.80, 122.70,

121.28, 120.99, 119.99, 29.84, 25.77, 18.35, -4.17, -4.24. HRMS (ESI) calculated for

+ + C33H44BrN3O3SSi2H (M+H) 698.18981, found 698.18915.

4.1.34 1,3-Bis(3-hydroxybenzoyl)-5-bromobenzene thiosemicarbazone (31).

1,3-Bis[(3-tert-butyldimethylsilyloxy)benzoyl]-5-bromobenzene

thiosemicarbazone (0.325 g, 0.465 mmol) was dissolved in tetrahydrofuran (10 mL)

followed by the addition of tetra-butylammonium fluoride trihydrate (0.604 g, 1.91 mmol) at room temperature. After 1.5 h, the reaction mixture was diluted with ethyl

70 acetate (50 mL), washed with brine (50 mL), and dried over anhydrous sodium sulfate.

After concentrating the reaction mixture under reduced pressure, the crude product was

purified using flash chromatography (silica gel, hexanes: dichloromethane, gradient,

50:50 to 0:100 followed by dichloromethane: ethyl acetate, gradient 100:0 to 60:40) to afford 1,3-bis(3-hydroxy-benzoyl)-5-bromobenzene thiosemicarbazone as a yellow solid

1 (0.158 mg, 0.336 mmol, 72% yield). H NMR (500 MHz, DMSO-d6) : δ 9.99 (0.8H, s),

9.92 (0.2H, s), 9.88 (0.2H, s), 9.86 (0.8H, s), 9.47 (0.2H, s), 8.72 (0.8H, s), 8.70 (0.8H, s),

8.53 (0.8H, s), 8.48 (0.8H, s), 8.45 (0.2H, s), 8.09 (0.2H,s), 7.98 (0.2H, t, J = 1.6 Hz),

7.81-7.79 (1H, m), 7.62 (0.8H, J = 1.5 Hz), 7.47-7.44 (1H, m), 7.37 (0.2H, t, J = 7.8 Hz),

7.34-7.31 (0.8H, m), 7.27-7.24 (0.4H, m), 7.17 (0.2H, t, J = 7.9 Hz), 7.13-7.04 (2.8H, m),

6.98 (0.8H, ddd, J = 8.3 Hz, 2.4 Hz, 0.65 Hz), 6.89 (0.2H, t, J = 2.0 Hz), 6.80 (0.2H, ddd,

J = 7.9 Hz, 2.4 Hz, 1.1 Hz), 6.78-6.76 (0.8H, m), 6.72-6.70 (0.8H, m). 13C NMR (125

MHz, DMSO-d6): δ 193.79, 193.70, 177.89, 158.50, 157.55, 157.48, 157.23, 146.51,

146.47, 140.20, 139.30, 138.72, 137.60, 137.38, 137.16, 134.93, 134.80, 132.63, 132.49,

132.46, 131.38, 131.25, 129.77, 129.70, 129.37, 128.84, 127.44, 122.68, 122.10, 121.38,

120.38, 120.50, 120.46, 188.48, 117.36, 116.81, 115.84, 115.79, 114.55. HRMS (ESI)

+ + calculated for C21H16BrN3O3SNa (M+Na) 491.99880, found 491.99903. HPLC

retention time (Method B): 9.97, 10.34 min. (Obtained as a mixture of E/Z isomers)

4.1.35 1,3-Bis(2-fluorobenzoyl)-5-bromobenzene thiosemicarbazone (32).51 (132)

p-Toluenesulfonic acid monohydrate (0.006 g, 0.03 mmol) was added to a

solution of 1,3-bis(3-fluorobenzoyl)-5-bromobenzene (0.190 g, 0.473 mmol) in

anhydrous tetrahydrofuran (15 mL). After stirring at reflux for 10 min, thiosemicarbazide

(0.088g, 0.97 mmol) was added to the reaction mixture and stirred for 28 h under an inert

71 atmosphere of nitrogen gas. After 28 h, tetrahydrofuran was removed under reduced

pressure and 10 mL of water was then added. The products were extracted with ethyl

acetate (2 x 50 mL) and the combined organic phases were washed with brine, dried over

anhydrous sodium sulfate, and the solvent was removed under reduced pressure.

Purification using flash chromatography (silica gel, hexanes:ethyl acetate, gradient 90:11

to 30:70) afforded 1,3-bis(3-fluorobenzoyl)-5-bromobenzene thiosemicarbazone (0.068

1 g, 0.143 mmol, 30% yield). H NMR (500 MHz, acetone-d6): δ 8.92 (1H, brs), 8.44 (1H, t, J = 1.8 Hz), 8.33 (1H, brs), 7.92 (1H, q, J = 1.5 Hz), 7.86 (1H, brs), 7.74-7.64 (3H, m),

7.59 (1H, td, J = 7.4 Hz, 1.8 Hz), 7.54-7.48 (2H, m), 7.46-7.41 (1H, m), 7.36 (1H, td, J =

7.5 Hz, 1.0 Hz), 7.27 (1H, ddd, J = 10 Hz, 8.4 Hz, 1.0 Hz). 1H NMR (400 MHz, acetone-

19 d6, F decoupled) 8.92 (1H, brs), 8.44 (1H, t, J = 1.8 Hz), 8.33 (1H, brs), 7.92 (1H, t, J =

1.7 Hz), 7.85 (1H, brs), 7.74-7.64 (3H, m), 7.59 (1H, dd, J = 7.7 Hz, 1.8 Hz), 7.57-7.47

(2H, m), 7.43 (1H, d, J = 8.4 Hz), 7.36 (1H, td, J = 7.5 Hz, 1.0 Hz), 7.27 (1H, dd, J = 8.4

13 Hz, 1.0 Hz). C NMR (125 MHz, acetone-d6): δ 191.49, 180.88, 160.87 (d, J = 251 Hz),

160.34 (d, J = 248 Hz), 141.81, 140.39, 140.01, 134.98 (d, J = 8.6), 134.13 (d, J = 8.3

Hz), 134.01, 133.79 (d, J = 1.4 Hz), 131.69 (d, J = 2.5 Hz), 131.59 (d, J = 3.1 Hz),

127.98 (d, J = 0.83 Hz), 126.83 (d, J = 3.5 Hz), 126.65 (d, J = 14 Hz), 125.59 (d, J = 3.5

Hz), 123.57, 118.89 (d, J = 17 Hz), 117.82 (d, J = 21 Hz), 117.11 (d, J = 22 Hz). ). 19F

- - - - NMR (470 MHz, acetone-d6): δ 112.74 - 112.79 (1F, m), 113.57 - 113.67 (1F, m).

+ + HRMS (ESI) calculated for C21H14BrF2 N3OSH (M+H) 474.00818, found 474.00845.

HPLC retention time (Method A): 10.19 min.

72 4.1.36 1,3-Bis(3-bromobenzoyl)-5-bromobenzene thiosemicarbazone (33).

1,3-Bis(3-bromobenzoyl)-5-bromobenzene (0.523 g, 1.00 mmol) was dissolved in

tetrahydrofuran (10 mL) followed by the dropwise addition of titanium isopropoxide

(1.18 mL, 4.00 mmol). The reaction was stirred for 10 min followed by the addition of

thiosemicarbazide (0.182 g, 2.00 mmol). After refluxing for 2 h, the reaction mixture was

quenched with 1 M HCl (20 mL) and extracted with ethyl acetate (3 x 40 mL). The combined organic phases were washed saturated aqueous sodium bicarbonate (20 mL) and dried over anhydrous sodium sulfate. Purification using flash chromatography (silica gel, hexane: ethyl acetate, gradient, 90:10 to 20:80) afforded 1,3-bis(3-bromobenzoyl)-5- bromobenzene thiosemicarbazone (0.140 g, mmol, 0.234 mmol, 23% yield). The product isomerizes in DMSO. The 1H NMR and 13C NMR for the isomer mixture is reported. 1H

NMR (500 MHz, DMSO-d6): δ 10.02 (0.5H, s), 9.14 (0.5H, s), 8.66 (0.5H, s), 8.60 (0.5H, s), 8.55-8.48 (1H, m), 8.40 (0.5H, s), 8.07 (0.5H, t, J = 1.7 Hz), 8.01 (0.5H, t, J = 1.7 Hz),

7.98 (0.5H, t, J = 1.7Hz), 7.91-7.84 (2.5H, m), 7.83 (0.5H, t, J = 1.7 Hz), 7.70 (0.5H, ddd,

J = 8.01 Hz, 2.0 Hz, 1.0 Hz), 7.67 (0.5H, ddd, J = 7.7 Hz, 1.6 Hz, 1.0 Hz), 7.65 (0.5H, t,

J = 1.7 Hz), 7.61-7.47 (3H, m), 7.42-7.36 (1H, m), 7.33-7.27 (0.5H, t, J = 8.0 Hz). 13C

NMR (125 MHz, DMSO-d6): δ 192.38, 178.49, 144.58, 138.87, 138.30, 138.14, 135.71,

133.08, 132.97, 132.92, 132.58, 131.83, 131.23, 130.71, 128.82, 127.85, 127.74, 122.96,

122.42, 121.94. X-ray crystallographic data obtained for 1,3-bis(3-bromobenzoyl)-5- bromobenzene thiosemicarbazone has been deposited in the Cambridge Crystallographic

Data Centre and was allocated the deposition number CDCC 1042568. HRMS (ESI)

+ + calculated for C21H14Br3N3OSH (M+H) 593.84805, found 593.84797. HPLC retention

time (Method A): 17.56. (Exist as a mixture of E/Z isomers in solution)

73 4.2 Biology

4.2.1 Cathepsin L Inhibition Assay

Enzyme assays for cathepsins L and B were modified from procedures originally described by Barrett and Kirschke.84 (185) Assays to determine the effects of inhibitors on human liver cathepsin L (Sigma Aldrich) activity were carried out using a fluorogenic peptide substrate N-carbobenzyloxy-L-Phe-L-Arg-7-amino-4-methylcoumarin (Z-FR-

AMC) (BACHEM). It should be noted that IC50 values were only determined for

compounds that, at a concentration of 10 µM, demonstrated greater than 50% inhibition

of enzyme activity. Cathepsin L was pre- incubated with inhibitors at various

concentrations (or with vehicle) for 5 min at 25˚C. The assay was initiated by the addition

of substrate Z-FR-AMC. The final assay conditions were 100 mM NaOAc buffer, pH 5.5,

1 mM EDTA, 3 mM DTT, 0.01% Brij 35 (Sigma), 1 nM cathepsin L, 2% DMSO

(Sigma) and 50 µM of Z-FR-AMC, in a total reaction volume of 200 μl. Inhibitors were

diluted to include a final concentration range of 10 µM to 10 pM. The release of AMC

from the substrate was monitored fluorometrically at 15 second intervals for 5 min at 25

oC using black 96 well Corning 3686 assay microplates with a Thermo Fluoroskan

Ascent FL microplate reader at excitation and emission filter wavelengths of 355 nm and

460 nm, respectively. Data were analyzed and IC50 values calculated utilizing GraphPad

Prism 5.0 software. IC50 +S.E (see Supplementary data) values represent the average

results from a minimum of three separate experiments.

4.2.2 Cathepsin B Inhibition Assay

Assays to determine the effects of inhibitors on cathepsin B activity were carried

out using the fluorogenic peptide substrate N-carbobenzyloxy-L-Arg-L-Arg-7-amino-4-

74 methylcoumarin (Z-RR-AMC), with IC50 values determined for compounds that demonstrated greater than 50% inhibition of enzyme activity at 10 µM of inhibitor concentration. Human liver Cathepsin B (EMD Millipore) was pre-incubated with inhibitors at various concentrations for 5 min at 37 oC. The assay was initiated by the addition of substrate Z-RR-AMC (EMD Millipore) and the final assay conditions were 1 nM cathepsin B, 60 µM Z-RR-AMC, 120 mM sodium potassium phosphate buffer pH

6.0, 1 mM EDTA, 3 mM DTT, 0.01% Brij 35, and 2% DMSO. Inhibitors were diluted to include a final concentration range of 10 µM to 10 pM. The reaction was monitored fluorometrically for 5 min at 37 oC using black 96 well Corning 3686 assay microplates with a Thermo Fluoroskan Ascent FL microplate reader at excitation and emission filter wavelengths of 355 nm and 460 nm, respectively. Data were analyzed and IC50 values calculated utilizing GraphPad Prism 5.0 software. Experiments were performed in triplicate.

4.2.3 Progress Curves

Various concentrations of compounds (final concentrations were 10, 5, 1, 0.5, 0.1,

0.01 and 0.005 μM) were mixed with Z-FR-AMC solution (final concentration 10 μM).

Reactions were initiated by the addition of cathepsin L in assay buffer with no pre- incubation time with inhibitor. Readings were taken every 30 sec for 50 min using a fluorescence microplate reader as described above.

4.2.4 Cell Culture

Human umbilical vein endothelial cell culture conditions:

Human umbilical vein endothelial cells (HUVECs) from pooled donors

(Invitrogen) were grown on rat tail collagen-1 coated flasks (CELLCOAT®) in M200

75 medium (Invitrogen) supplemented with 1% gentamycin sulfate (Teknova), 1% amphotericin B (Corning) and low serum growth factor supplement kit (Invitrogen).

HUVECs were passaged using Trypsin EDTA/Trypsin Neutralizer solutions (Invitrogen) as per manufacturer recommendations. Cells were maintained at 37 oC in a humidified atmosphere of 5% CO2. HUVECs were not used beyond passage 5 in these experiments.

PC-3ML cell culture conditions:

PC-3ML are highly metastatic sublines isolated from PC-3 cells.70(172) Cell were cultured in appropriate media supplemented with 10% FBS at 37 oC in a humidified atmosphere of

5% CO2 in air.

MDA-MB-231 cell culture conditions:

MDA-MB-231 cells (ATCC) were cultured in DMEM (Corning) supplemented with 10% fetal bovine serum (Gibco One Shot®) obtained from Invitrogen and 1% gentamycin sulfate, and passaged using trypsin solution (Mediatech) in T-75 culture flasks (Corning).

4.2.5 Cell Invasion Assay

PC-3ML

Invasion assays were performed using Matrigel coated invasion inserts from BD

Biosciences (354480). Cells suspended in serum free media were seeded into the inserts.

Media supplemented with 10% FBS was added to the bottom chamber. The cells were incubated under desired conditions and 24h later, cells that invaded to the underside of the membrane were stained and counted under a microscope.

76 MDA-MB-231

The effects of compounds 1, 8, and 32 on the invasive ability of MDA-MB-231 cells were analyzed using a cell invasion assay with BD Bioscience Matrigel™ invasion kits. MDA-MB-231 cells were cultured and passaged as previously described. Cells were trypsinized and removed from the culture flasks when they were 80% confluent. Cell density was determined with a Beckman Coulter Z-Coulter cell counter. Stock solutions of compounds 1, 8, and 32 in DMSO were prepared in addition to E-64, a known generic cysteine protease inhibitor which was used as a positive control. BD BioCoat Matrigel® invasion chambers, which contained 8 micron pore size PET membranes with a layer of

Matrigel, were used for the experiment. The experiment was initiated by adding DMEM supplemented with 10% FBS (which functions as a chemoattractant) and gentamicin to a

24-well microplate (i.e. lower chamber). Then, the inserts were carefully placed on top of the wells containing the chemoattractant. Cell and compound solutions were added such that the final conditions per well were: 2% DMSO, 50,000 cells in DMEM and 25 or 10

μM of the compounds. Final conditions for untreated (controls) cells were: 2% DMSO and 50,000 cells. The 24-well plates containing the invasion chambers were placed in an

o incubator with a 5% CO2 environment for 24 hours at 37 C. The experiments were terminated, invaded cells were fixed with methanol, stained using a Diff staining kit

(IMEB Inc), and rinsed with deionized water. Samples were air-dried and membranes were removed and placed on glass slides. Each sample was observed with a Zeiss

Axiovert 40 CFL inverted microscope to perform manual cell counting (ten fields were observed under a 40x objective). Experiments were performed in triplicate.

77 4.2.6 Cell Migration Assay

Inhibition of MDA-MB-231 cell migration was determined with an assay similar

to that described for the cell invasion assay. However, the membrane inserts had 8µm

pores but no Matrigel layer. Experiments were performed in triplicate.

4.2.7 Growth Inhibition of C3H Mammary Carcinoma

Experiments were performed using 10-14-week-old female CDF1 mice, in which

a C3H mammary carcinoma was implanted in the right rear foot. This tumor model is an

anaplastic adenocarcinoma that arose spontaneously in a C3H mouse at Aarhus

University Hospital and was originally designated as HB;85 (186) the name was changed to

C3H mammary carcinoma when it was grown in the more stable CDF1 mouse variant.86

(187) C3H mammary carcinomas do not grow in culture, thus experimental tumors were

produced following sterile dissection of large flank tumors as previously described.87 (188)

Basically, macroscopically viable tumor tissue was minced with scissors and 5-10 µl of this material implanted into the foot. Treatments were started at the time of tumor implantation; tumor volume was determined by the formula D1 x D2 x D3 x π/6 (where the D values represent three orthogonal diameters). All animal studies were conducted according to the animal welfare policy of Aarhus University (http://dyrefaciliteter.au.dk), with the Danish Animal Experiments Inspectorate’s approval. 3-Benzoylbenzophenone thiosemicarbazone (1) was supplied by Baylor University (Waco, Texas, USA). It was freshly prepared before each experiment by dissolving in Tween80 and then diluted to

10% using saline before injection. Stock solutions were kept cold and protected from light. 3-Benzoylbenzophenone thiosemicarbazone (1) was injected intraperitoneally (i.p.)

78 in a volume of 0.02 ml/g mouse body weight for 5 consecutive days starting either on the

day of tumor implantation or when tumors had reached 200 mm3.

4.2.8 Cytotoxicity Assay

The sulforhodamine B (SRB) assay was used to assess inhibition of human cell

line growth as previously described.88-90 (189–191) Briefly, HUVECs were passaged using

normal conditions and plated at 9,000 cells/well in 96-well plates (Corning) and

incubated for 24 h. Ten-fold serial dilutions of the compounds to be tested were then

added to the wells. After 48 h, treated and control cells were fixed with 10%

trichloroacetic acid, stained with 0.4% sulforhodamine B (Acid Red 52) (TKI) for 30 minutes, and subsequently washed 4 times with 1% acetic acid in water. The plates were air dried, and the SRB dye was solubilized with 200 µL of 10 mM Tris base and read at

540 nm with an automated Biotek Elx800 plate reader (Biotek). Values were normalized

91 (192) to 630 nm to account for background absorbance. A growth inhibition of 50% (GI50

or the drug concentration causing a 50% reduction in net protein absorbance relative to

controls) was calculated from a minimum of two replicates and averaged for a minimum

of three experiments with Excel software.

Acknowledgments

Financial support of this research was generously provided by OXiGENE, Inc.

(grant to KGP and MLT) and the NIH-United States (grant R01 CA 169300 to DWS).

The authors express their appreciation to Dr. Kevin K. Klausmeyer and Marissa Penney

for X-ray crystallography studies, Dr. Nicole Kruse (Applications Scientist at Bruker

BioSpin) for acquiring data on the 400 MHz NMR, Dr. Alejandro Ramirez (Mass

Spectrometry Core Facility, Baylor University) for assistance with mass spectrometry

79 analysis, Dr. Michelle Nemec (Director of the Molecular Biosciences Center at Baylor

University) for use of shared facilities, and Mr. Kahler Low for technical assistance. For

the in vivo experiments, the authors thank Ms. Dorthe Grand and Ms.Inger Marie

Horsman for technical assistance, and the Danish Cancer Society and the Danish Council

for Independent Research: Medical Sciences, for financial support.

Supplementary data

Supplementary data including X-ray crystallography, 1H NMR, 19F NMR, 13C

NMR, HRMS, HPLC analysis, molecular modeling studies, enzyme kinetic studies, and

Lipinski rule of five analysis associated with this article can be found, in the online version, at http://www.sciencedirect.com/science/article/pii/S0968089615300614.

References

1. Fonović, M.; Turk, B. Biochim. Biophys. Acta 2014, 1840, 2560-2570.

2. Mohamed, M. M.; Sloane, B.F. Nature Reviews Cancer 2006, 6, 764-775.

3. Gocheva, V.; Joyce, J. A. Cell Cycle 2007, 6, 60-64.

4. Lankelma, J. M.; Vorrend, D. M.; Barwari, T.; Koetsveld, J.; Van der Spek, A. H.; De Porto, A. P.; Van Rooijen, G.;Van Noorden, C. J. Life Sci. 2010, 86, 225- 233.

5. Brömme, D.; Wilson, S. Role of Cysteine Cathepsins in Extracellular Proteolysis. In Extracellular Matrix Degradation; Parks, W. C.; Mecham, R. P., Eds.; Biology of Extracellular Matrix 2; Springer Berlin Heidelberg, 2011; pp 23-51.

6. Gocheva, V.; Zeng, W.; Ke, D.; Klimstra, D.; Reinheckel, T.; Peters, C.; Hanahan, D.; Joyce, J. A. Dev. 2006, 543-546.

7. Blondeau, X.; Vidmar, S. L.; Emod, I.; Pagano, M.; Turk, V.; Keil-Dloua, V. Biol. Chem. Hoppe-Seyler 1993, 374, 651-656.

8. Ishidoh, K.; Kominami, E. Biochem. Biophys. Res. Commun. 1995, 217, 624-631.

80 9. Guinec, N.; Dalet-Fumeron, V.; Pagano, M. Biol. Chem. Hoppe-Seyler 1993, 374, 1135-1146.

10. Buck, M. R.; Karustis, D. G.; Day, N. A.; Honn, K.V.; Sloane, B. F. Biochem J. 1992, 282, 273-278.

11. Kirschke, H.; Kembhavi, A. A.; Bohley, P.; Barret, A. J. Biochem. J. 1982, 201, 367-372.

12. Maciewicz, R. A.; Wotton, S. F.; Etherington, D. J.; Duance, V. C. FEBS Lett. 1990, 269, 189-193.

13. Garnero, P.; Borel, O.; Byrjalsen, I.; Ferreras, M.; Drake, F. H.; McQueney, M. S.; Foged, N. T.; Delmas, P. D.; Delaissé, J.-M. J. Biol. Chem. 1998, 273, 32347- 32352.

14. Sameni, M.; Dosescu, J.; Moin, K.; Sloane, B. F. Mol. Imaging 2003, 2, 159-175.

15. Rothberg, J. M.; Bailey, K. M.; Wojtkowiak, J. W.; Ben-Nun, Y.; Bogyo, M.; Weber, E.; Moin, K.; Blum, G.; Mattingly, R. R.; Gillies, R. J.; Sloane, B. F. Neoplasia 2013, 15, 1125-1137.

16. Manson, S. D.; Joyce, J. A. Trends in Cell Biol. 2011, 21, 228-237.

17. Bell-McGuinn, K. M.; Garfall, A. L.; Bogyo, M.; Hanahan, D.; Joyce, J. A. Can. Res. 2007, 65, 7378-7385.

18. Berdowska, I. Clin. Chim. Acta 2004, 342, 41-69.

19. Ullah, M. F.; Aatif, M. Cancer Treat. Rev. 2009, 35, 193-200.

20. Chen, B.; Platt, M. O. J. Transl. Med. 2011, 9, 109-120.

21. Jensen, A. B.; Wynne, C.; Ramirez, G.; He, W.; Song, Y.; Berd, Y.; Wang, H.; Mehta, A.; Lombardi, A. Clin. Breast Cancer 2010, 10, 452-458.

22. Clezardin, P. Breast Cancer Res. 2011, 13, 207.

23. Stoch, S. A.; Zajic, S.; Stone, J. A.; Miller, D. L.; van Bortel, L.; Lasseter, K. C.; Pramanik, B.; Cilissen, C. ; Liu, Q.; Liu, L.; Boyd, B. S.; Paneblanco, D.; Ding, Yu, Gottesdiener, K.; Wagner, J. A. Br. J. Clin. Pharmacol. 2012, 75, 1240-1254.

24. Gauthier, J. Y.; Chauret, N.; Cromlish, W.; Desmarais, S.; Duong, L. T.; Falgueyret, J.-P.; Kimmel, D. B.; Lamontagne, S.; Léger, S.; LeRiche, T.; Li, C. S.; Massé, F.; McKay, D. J.; Nicoll-Griffith, D. A.; Oballa, R. M.; Palmer, J. T.; Percival, D.; Riendeau, D.; Robichaud, J.; Rodan, G. A.; Rodan, S. B.; Seto, C.;

81 Thérien, M.; Truong, V.-L.; Venuti, M. C.; Wesolowski, G.; Young, R. N.; Zamboni, R.; Black, W. C. Bioorg. Med. Chem. Lett. 2008, 18, 923-928.

25. Lindstrom, E.; Grabowska, U.; Jerling, M.; Edenius, C.; MIV-711, A Highly Selective Cathepsin K Inhibitor, Reduces Biomarkers of Bone Resorption and Cartilage Degradation in Healthy Subjects. Proceedings of the 2014 OASRI World Congress, Paris, France, April 24-27, 2014. Osteoarthritis and Cartilage, 2014, S197.

26. About Cathepsin B and Liver Disease. http://www.virobayinc.com/liver- fibrosis.php (Accessed on March 20, 2014).

27. Evaluation of Safety and Pharmacokinetics of Single Doses of VBY-376 in Healthy Adults. http://clinicaltrials.gov/ct2/show/NCT00557583 (Accessed on October 1, 2014).

28. Pan cathepsin inhibitor: VBY-825. http://www.virobayinc.com/VBY-825.php (Accessed on October 1, 2014)

29. Elie, B. T.; Gocheva, V.; Shree, T.; Darlrymple, S. A.; Holsinger, L. J.; Joyce, J. A. Biochimie 2010, 92, 1618-1624.

30. GSK Product development pipeline February 2014. http://www.gsk.com/media/280387/product-pipeline-2014.pdf (Accessed on September, 8, 2014)

31. A Study to Evaluate the Safety, Tolerability, Pharmacokinetics (PK), Pharmacodynamics (PD) and Food Effect of Single or Repeat Doses of GSK2793660 in Healthy Subjects. https://clinicaltrials.gov/ct2/show/NCT02058407 (Accessed on September, 8, 2014).

32. Rizoska, B; Edenius, C.; Göhlin, K.; Hewitt, E.; Lindström, E.; Malcangio, M.; Pitcher, T.; Tunblad, K. MIV-247 A Cathepsin Protease Inhibitor Developed as a Novel Therapy for Neuropathic Pain. Proceedings of the 15th World Congress on Pain 2014, Buenos Aires, Argentina, October 6-11, 2014.

33. Miller, B.; Friedman, A. J.; Choi, H.; Hogan, J.; McCammon, J. A.; Hook, V.; Gerwick, W. H. J. Nat. Prod. 2014, 77, 92-99.

34. Katunuma, N.; Murata, E.; Kakegawa, H.; Matsui, A.; Tsuzuki, H.; Tsuge, H.; Turk, D.; Turk, V.; Fukushima, M.; Yada, Y.; Asao, T. FEBS Lett. 1999, 458, 6- 10.

82 35. Shah, P. P.; Wang, T.; Kaletsky, R. L.; Myers, M. C.; Purvis, J. E.; Jing, H.; Huryn, D. M.; Greenbaum, D. C.; Smith III, A. B.; Bates, P.; Diamond, S. L. Mol. Pharmacol. 2010, 78, 319-324.

36. Dana, D.; De, S.; Rathod, P.; Davalos, A. R.; Novoa, D. A.; Paroly, S.; Torres, V. M. Afzal, N.; Lankalapalli, R. S.; Rotenberg, S. A.; Chang, E. J. Subramaniam, G.; Kumar, S. Chem. Commun. 2014, 50, 10875-10878.

37. Assad, N.; Bethel, P. A.; Coulson, M. D.; Dawson, J. E.; Ford, S. J.; Gerhardt, S.; Grist, M.; Hamlin, G. A.; James, M. J.; Jones, E. V.; Karoutchi, G. I.; Kenny, P. W.; Morley, A. D.; Oldham, K.; Rankine, N.; Ryan, D.; Wells, S. L.; Wood, L.; Augustin, M.; Krapp, S.; Simader, H.; Steinbacher, S. Bioorg. Med. Chem. Lett. 2009, 19, 4280-4283.

38. Marquis, R. W.; James, I.; Zeng, J.; Trout, R. E. L.; Thompson, S.; Rahman, A.; Yamashita, D. S.; Xie, R.; Ru, Y.; Gress, C. J.; Blake, S.; Lark, M. A.; Hwang, S.- M.; Tomaszek, T.; Offen, P.; Head, M. S.; Cummings, M. D.; Veber, D. F. J. Med. Chem. 2005, 48, 6870-6878.

39. Falgueyret, J.-P.; Oballa, R. M.; Okamoto, O.; Wesolowski, G.; Aubin, Y.; Rydzewski, R. M.; Prasit, P.; Riendeau, D.; Rodan, S. B.; Percival M. D. J. Med. Chem. 2001, 44, 94-104.

40. Frizler, M.; Lohr, F.; Furtmann, N.; Kläs, J.; Gütschow, M. J. Med. Chem. 2011, 54, 396-400.

41. Bauer, D. J. Clinical experience with the antiviral drug Marboran® (1-methylisatin 3-thiosemicarbazone) Ann. N. Y. Acad. Sci. 1965, 130, 110–117.

42. Bauer D. J.; St Vincent, L.; Kempe, C.; Young, P. A.; Downie, A.W. Am. J. Epidemiol.I 1969, 90, 130-145.

43. Weiss, M. M.; Weiss, P. D.; Mathisen, G.; Guze, P. Clin. Infect. Dis. 2004, 39, 1668-1673.

44. Caminero, J. A.; Sotgiu, G.; Zumla, A.; Migliori, G. B. Lancet Infect. Dis. 2010, 10, 621-629.

45. Kishore Kumar, G. D.; Chavarria, G. E.; Charlton-Sevcik, A. K.; Arispe, W. M.; MacDonough, M. T.; Strecker, T. E.; Chen, S.-E.; Siim, B. G.; Chaplin, D. J.; Trawick, M. L.; Pinney, K. G. Bioorg. Med. Chem. Lett. 2010, 20, 1415−1419.

46. Kishore Kumar, G. D.; Chavarria, G. E.; Charlton-Sevcik, A. K.; Yoo, G. K.; Song, J.; Strecker, T. E.; Siim, B. G.; Chaplin, D. J.; Trawick, M. L.; Pinney, K. G. Bioorg. Med. Chem. Lett. 2010, 20, 6610−6615.

83 47. Chavarria, G. E.; Horsman, M. R.; Arispe, W. M.; Kishore Kumar, G. D; Chen, S.-E.; Strecker, T. E.; Parker, E. N.; Chaplin, D. J.; Pinney, K.G.; Trawick, M.L. Eur. J. Med. Chem. 2012, 58, 568-572.

48. Sudhan, D. R.; Siemann, D. W. Clin. Exp. Metastasis 2013, 30, 891-902.

49. Song, J.; Jones, L. M.; Kishore Kumar, G. D.; Conner, Liela Bayeh; E. S.; Chavarria, G. E.; Charlton-Sevcik, A. K.; Shen-En Chen; Chaplin, D. J.; Trawick, M. L.; Pinney, K. G. ACS Med. Chem. Lett. 2012, 3,450-453.

50. Song, J.; Jones, L. M.; Chavarria, G. E.; Charlton-Sevcik, A.K.; Jantz, A.; Johansen, A.; Bayeh, L.; Soeung, V.; Snyder, L.K.; Lade, S.D.; Chaplin, J.; Trawick, M.L.; Pinney, K.G. Bioorganic and Medicinal Chemistry Letters 2013, 23, 2801-2807.

51. Chaplin, D. J.; Gaddale Devanna, K.K; Parker, E.; Pinney, K. G.; Jiangli, S.; Trawick, M. L. PCT Int. Appl., WO 2013142038 A2 2013092, September 26, 2013.

52. Raghav, N.; Kaur, R. Med. Chem. Res. 2014, 23, 4669-4679.

53. Siles, R.; Chen. S.-E.; Zhou, M.; Pinney, K. G.; Trawick, M. L. Bioorg. Med. Chem. Lett. 2006, 16, 4405-4409.

54. Du, X.; Guo, C.; Hansell, E.; Doyle, P. S.; Caffey, C. R.; Holler, T. P.; McKerrow, J. H.; J. Med. Chem. 2002, 45, 2695-2707.

55. Mallari, J. P.; Shelat, A.; Kosinski, A.; Caffrey, C. R.; Connelly, M.; Zhu, F.; McKerrow, J. H.; Guy, R. K. Bioorg. Med. Chem Lett. 2008, 18, 2883-2885.

56. Caputto, M. E.; Fabian, L. E.; Benítez, D.; Merlino, A.; Ríos, N.; Cerecetto, H.; Moltrasio, G. Y.; Moglioni, A. G.; González, M.; Finkielsztein, L. M. Bioorg. Med. Chem. 2011, 19, 6818-6826.

57. Greenbaum, D. C.; Mackey, Z.; Hansell, E.; Doyle, P.; Gut, J.; Caffrey, C. R.; Lehrman, J.; Rosenthal, P. J.; McKerrow, J. H.; Chibale, K. J. Med. Chem. 2004, 47, 3212-3219.

58. Chiyanzu, I.; Clarkson, C.; Smith, P. J.; Lehman, J.; Gut, J.; Rosenthal, P. J.; Chibale, K. Bioorg. Med. Chem. 2005, 13, 3249-3261.

59. Liu, Y.; Cui, K.; Lu, W.; Luo, W.; Wang, J.; Huang, J.; Guo, C. Molecules 2011, 16, 4527-4538.

60. Schröder, J.; Noack, S.; Marhöfer, R. J.; Mottram. J. C.; Coombs, G. H.; Selzer, P. M. PLoS ONE [Online] 2013, 8, e77460.

84 61. http://journals.plos.org/plosone/article?id=10.1371/journal.pone.0077460 (Accessed on September, 8, 2014).

62. Schaeffer, M.; Schroeder, J.; Heckeroth, A. R.; Noack, S.; Gassel, M.; Mottram, J. C.; Selzer, P. M.; Coombs, G. H. Antimicrob. Agents Chemother. 2012, 56, 1190- 1201.

63. Johnson, J. E.; Morales, N. M.; Gorczyca, A. M.; Dolliver, D. D.; McAllister, M. A. J. Org. Chem. 2001, 66, 7979-7985.

64. Karabatsos, G. J.; Vane, F. M.; Taller, R. A.; Hsi, N. J. Am. Chem. Soc. 1964, 86, 3351-3357.

65. Kerek, F.; Ostrogovich, G.; Simon, Z. J. Chem. Soc. B 1971, 541-544.

66. Ostrogovich, G.; Simon, Z.; Kerek, F. Revue Tomaine de Chimie, 1970, 15, 1453- 1459.

67. Curtin, D. Y.; Grubbs, E. J.; McCarty, C. G. J. Am. Chem. Soc. 1966, 88, 2775- 2786.

68. Hardegger, L. A.; Kuhn, B.; Spinnler, B.; Anselm, L.; Ecabert, R.; Stihle, M.; Gsell, B.; Thoma, R.; Diez, J.; Benz, J.; Plancher, J.-M.; Hartmann, G.; Banner, D. W.; Happ, W.; Diderich, F. Angew. Chem. Int. Ed. 2011, 50, 314-318.

69. Choe, Y.; Leonetti, F.; Greenbaum, D. C.; Lecaille, F.; Bogyo, M.; Bromme, D.; Ellman, J. A.; Craik, C. S. J. Biol. Chem. 2006, 281, 12824–12832.

70. Albini, A.; Iwamoto, Y.; Kleinman, H. K; Martin, G. R.; Aaronson, S. A.; Kozlowski, J. M.; McEwan, R. N. Cancer Res. 1987, 47, 3239-3245.

71. Wang, M.; Stearns, M. E. Differentiation 1991, 48, 115-125.

72. Neve, R. M,; Chin, K.; Fridlyand, J.; Yeh, J.; Baehner, F. L.; Fevr, T.; Clark, L.; Bayani, N.; Coppe, J.; Tong, F.; Speed, T.; Spellman, P. T.; DeVries, S.; Lapuk, A.; Wang, N.J.; Kuo, W.-L.; Stilwell, J. L.; Pinkel, D.; Albertson, D. G.; Waldman, F. M.; McCormick, F.; Dickson, R. B.; Johnson, M. D.; Lippman, M.; Ethier, S.; Gazdar, A.; Gray, J. W.. Cancer Cell 2006, 10, 515-527.

73. Zajc, I.; Sever, N.; Bervar, A.; Lah, T. T. Cancer. Lett. 2002, 187, 185-190.

74. Tamai, M.; Yokoo, C.; Murata, M.; Oguma, K.; Sota, K.; Sato, E.; Kanaoka, Y. Chem. Pharm. Bull. 1987, 35, 1098-1104.

85 75. Mitchell, D.; Lukeman, M.; Lehnherr, D.; Wan, P. Formal intramolecular photoredox chemistry of meta-substituted benzophenones. Org. Lett. 2005, 7, 3387-3389.

76. Zhao, W.; Carreira, E. M. Org. Lett. 2006, 8, 99-102.

77. Ye S.; Chen, J.; Di, C.; Liu, Y.; Lu, K.; Wu, W.; Du, C.; Liu, Y.; Shuai, Z.; Yu, G. J. Mater. Chem. 2010, 20, 3186-3194.

78. Sharghi, H.; Jokar, M.; Doroodmand, M. M.; Khalifeh, R. Adv. Synth. Catal. 2010, 352, 3031-3044.

79. Blick, F. F.; Patelski, R. A. J. Am. Chem. Soc. 1938, 60, 2283-2285.

80. Miller, T. M.; Kwock, E. W.; Neenan, T. X. Macromolecules 1992, 25, 3143- 3148.

81. Teki, Y.; Takui, T.; Itoh, K.; Iwamura, H.; Kobayashi, K. J. Am. Chem. Soc. 1986, 108, 2147-2156.

82. Ronald, S.; Michael, S. PCT Int. Appl. WO 9907336 A1 19990218, February 18, 1999.

83. Knudsen, S. M.; Pettersson, I.; Lau, J.; Behrens, C.; Petersen, A. K.; Garibay, P. W. PCT Int. Appl. WO 2006003096 A1 20060112, January 12, 2006.

84. Chang, L. L. PCT Int. Appl. WO 9822108 A1 19980528, May 28, 1998.

85. Barrett, A. J.; Kirschke, H. Method. Enzymol. 1981, 80, Pt C, 535-561.

86. Overgaard, K.; Overgaard, J. Eur. J. Cancer 1972, 8, 65-78.

87. Overgaard, J. Int. J. Radiat. Oncol. Biol. Phys. 1980, 6, 1507-1515.

88. Horsman, M.R.; Murata, R.; Breidahl, T.; Nielsen, F.U.; Maxwell, R.J.; Stødkilde- Jørgensen, H.; Overgaard, J. Adv. Exp. Med. Biol. 2000, 476, 311-323.

89. Monks, A.; Scudiero, D.; Skehan, P.; Shoemaker, R.; Paull, K.; Vistica, D.; Hose, C.; Langley, J.; Cronise, P.; Vaigro-Wolff, A.; Gray-Goodrich, M.; Campbell, H.; Mayo, J.; Boyd, M. J. Natl. Cancer Inst. 1991, 83, 757-766.

90. Vichai, V.; Kirtikara, K. Nat. Protoc. 2006, 1, 1112-1116.

91. MacDonough, M. T.; Strecker, T. E.; Hamel, E.; Hall, J. J.; Chaplin, D. J.; Trawick, M. L.; Pinney, K. G. Bioorg. Med. Chem. 2013, 21, 6831-6843.

86 92. Skehan, P.; Storeng, R.; Scudiero, D.; Monks, A.; McMahon, J.; Vistica, D.; Warren, J. T.; Bokesch, H.; Kenney, S.; Boyd, M. R. J. Natl. Cancer Inst. 1990, 82, 1107-1112.

87

CHAPTER THREE Synthesis and Biological Evaluation of a Water Soluble Phosphate Prodrug Salt and Structural Analogues of KGP94, a Lead Inhibitor of Cathepsin L

Erica N. Parker †, Samuel O. Odutola‡, Yifan Wang†, Gustavo E. Chavarria†, Tracy E. Strecker†, David J. Chaplin§, Mary Lynn Trawick†, and Kevin G. Pinney*,‡,† †Department of Chemistry and Biochemistry Baylor University One Bear Place #97348 Waco, TX 76798-7348 ‡Institute of Biomedical Studies Baylor University One Bear Place #97224 Waco, TX 76798-7224 §OXiGENE, Inc. 701 Gateway Blvd, Suite 210 South San Francisco, CA 94080

Note: The present chapter is formulated as a manuscript draft in preparation for

submission to a medicinal chemistry journal.

Abstract

The degree of expression of cathepsin L, often unregulated in the tumor

microenvironment, reciprocally parallels with the invasive and metastatic nature of

certain cancer cell lines. Inhibition of cathepsin L represents and emerging strategy for

the treatment of metastatic cancer. KGP94 ([(3-bromophenyl)-(3-hydroxyphenyl)-ketone]

thiosemicarbazone), a potent inhibitor of cathepsin L, was synthesized by an improved

route in order to eliminate the replacement of bromine by hydrogen as observed in the

previous synthesis. Additional analogues containing the KGP94 motif were synthesized

and evaluated for inhibitory activity against cathepsin L. Due to limited aqueous

solubility of KGP94, a water-soluble phosphate salt (KGP420) was prepared. Enzymatic

hydrolysis assays with alkaline phosphatase demonstrated that the phosphate prodrug,

KGP420, was readily converted to the parent compound, KGP94.

88 Introduction

The discovery and development of effective therapeutic regimens that target cancer metastasis remains an urgent and largely unmet need. Metastasis is a significant contributing factor in over ninety percent of deaths attributed to cancer.1 The sequence of steps involved in the metastatic process associated with tumor cells includes invasion of these cells from the primary tumor into the surrounding tissue, intravasation into the circulatory system, extravasation from the circulatory system, and establishment of a secondary tumor.1,193,194 One promising strategy towards the development of anti- metastatic agents involves targeting one or more members of the Papain family of cysteine cathepsin proteases (comprised of 11 members: B, C, F, H, K, L, O, S, V, W, and X/Z) with small-molecule inhibitors.195,53,54,8,148 Cathepsins aid in the invasion and migration of tumor cells through the degradation of proteins comprising the extracellular matrix including collagen,70,75–80 fibronectin, 73–76 and laminin74–76. Elevated levels of cathepsins L, B, H, X, and S have been detected in several cancer types including breast, prostate, brain, colorectal, and lung cancers. Moreover, increased expression of these cathepsins correlates inversely to overall prognosis in breast, ovarian, colorectal, brain, lung, and head and neck cancers.152,153 Decreased tumor volume and increased survival rates were observed in a mouse model with inclusion of a pan-cathepsin inhibitor with cyclophosphamide, a known chemotherapeutic, as a cancer treatment regimen.196

In addition to the role that these enzymes play in metastasis and migration of cancer cells through degradation of components of the extracellular matrix, cysteine cathepsin proteases have been implicated as targets for osteoporosis, rheumatoid arthritis, atherosclerosis, and diseases of the immune system.195 Cathepsin inhibitors as drug

89 candidates for the treatment of various diseases in the pharmaceutical pipeline include

VBY-825 (Virobay), 96,97 Odanacatib (Merck), 84,85 LY3000328 (Eli Lily), 98,99 (Figure

3.1).

Figure 3.1. Cathepsin inhibitors in the pharmaceutical pipeline.

VBY-825 (Virobay), a pan cysteine protease inhibitor targeting the treatment of

liver fibrosis, incorporates a diketone warhead into a peptide-like backbone allowing for

reversible inhibition of targeted cathepsins. Odanacatib (Merck), a nitrile based inhibitor, targets cathepsin K for suppression of bone resorption in osteoporosis. LY3000328 (Eli

Lily), a noncovalent cathepsin S inhibitor with an IC50 value in the low nM range, targets

the treatment of abdominal aortic aneurysm. Although significant progress has been

achieved towards the development of cathepsin inhibitors as therapeutic treatment

options, no FDA approved drugs currently exist in this pharmaceutical field.

Additionally, small molecular entities that specifically target the inhibition of cathepsin L

for the treatment of pathological processes in clinical trials remains an unmapped frontier

open to exploration.

90

Figure 3.2 Representative sampling of potent covalent inhibitors of cathepsin L

Small-molecule inhibitors of cathepsin L have been synthesized that incorporate a variety of electrophilic moieties (warheads) capable of interacting with the catalytic site residue Cys-25 (Figure 3.2). Warheads which undergo covalent bonding with the Cys25 thiolate of cathepsin L include the epoxide in Clik 148 (I),158 the carbonyl of thiocarbazate II,123 the nitrile in the purine analogue III,197 the cyclic carbonyl in azepanone IV,120 the nitrile in the triazine analogue V,124 the α,β-unsaturated amide of gallinamide A (VI), 113,114 and the aldehyde of the N-(1-naphthalenylsulfonyl) peptide derivative VII109 (Figure 3.2). Cruzain, is a cysteine protease found in Trypanosoma cruzi, and is targeted for the treatment of Chagas’ disease. Initial studies towards the development of cruzain135,136 inhibitors led to the discovery of cathepsin L inhibitors bearing the thiosemicarbazone moiety which contains an electrophilic thiocarbonyl warhead. Building on these results, we embarked on a structure-activity relationship

(SAR) guided program designed to incorporate the thiosemicarbazone moiety within

91 appropriately functionalized benzophenone, pyridine, thiophene, fluorene,

thiochromanone, benzothiepine, and dihydroquinoline molecular scaffolds. A focused

small library of cathepsin inhibitors resulted from these studies, from which a sub-set of

69,126– molecules (greater than 35) demonstrated IC50 values below 500 nM (Figure 3.3).

133

Figure 3.3. Sub-set of previously described thiosemicarbazone based inhibitors of cathepsin L

A lead compound (referred to as KGP94), which is a slowly reversible, time- dependent inhibitor of cathepsin L, emerged from this small library of structurally diverse

thiosemicarbazone analogues.128,129,69 Low cytotoxicity against human umbilical vein

endothelial cells (HUVECs) and the ability to inhibit the invasive and migratory potential

of both PC-3ML (prostate cancer cell line) and MDA-MB-231 (breast cancer cell line)

has led to the identification of KGP94 as a pre-clinical candidate for potential

development as an anti-metastatic agent, functioning through a potent inhibition of

cathepsin L.69,129 Since KGP94 demonstrates limited solubility in water, it proved

92 desirable to prepare a water-soluble prodrug salt to further the pre-clinical development

of this promising agent. Fortunately, KGP94 bears a phenolic hydroxyl group which

provides a convenient molecular handle for the introduction of a bioreversible promoiety in order to improve aqueous solubility and potentially enhance ADME (adsorption, distribution, metabolism, and excretion) pharmacokinetic properties. Installation of a phosphate prodrug salt for the purpose of increasing aqueous solubility of FDA approved drugs intended for oral or parental administration has been successfully demonstrated by fosfpropofol, fosamprenavir, and fosfluconazole.198

Results and Discussion

Design and Synthetic Chemistry

In addition to the synthesis of a water-soluble phosphate prodrug of KGP94, several analogues of this parent, lead compound were also prepared. Previous structure- activity relationship (SAR) studies related to thiosemicarbazone inhibitors based on the benzophenone molecular scaffold highlighted the importance of the 3-bromophenyl moiety. 127,128The extended series of benzophenone-based thiosemicarbazone inhibitors

incorporate both m-hydroxy and m-bromo substituents. Our initial synthetic route128 to

KGP94 (Scheme 3.1) utilized the addition of 3-bromophenylmagnesium bromide to the corresponding Weinreb amide to afford (3-bromophenyl)-(3-hydroxyphenyl)-methanone, which was condensed with thiosemicarbazide followed by deprotection of the silyl ether.

However, HPLC analysis of the final product revealed that a trace amount of the bromine atom had been replaced by a hydrogen.128 Replacement of bromine by hydrogen likely occurred during the halogen-metal exchange reaction since excess magnesium was used to prepare benzophenone (3-bromophenyl)-(3-hydroxyphenyl)-methanone.

93

Scheme 3.1. Previously reported synthetic route towards KGP94128

In an effort to avoid these trace impurities, KGP94 and analogues were synthesized utilizing a revised route (Scheme 3.2). Instead of using 1,3- dibromobenzene as the precursor to the organometallic reagent for the synthesis of KGP94 (11), a protected m-bromophenol 1 was reacted with n-butyllithium to form the intermediate organolithium reagent, which was reacted with Weinreb amide 4 to afford the desired functionalized benzophenone 7. Benzophenone intermediates 8 and 10 were synthesized in a similar manner by reacting the appropriately substituted aromatic ring with n- butyllithium followed by the addition of Weinreb amide 5 to afford ketone 8 or the addition of aldehyde 6 followed by oxidation with PCC to afford ketone 10.

Condensation of benzophenones intermediates 7, 8, and 10 (separately) with thiosemicarbazide followed by desilylation with TBAF afforded target thiosemicarbazone analogues KGP94 (11), 12, and 13. HPLC analysis indicated no trace amount of the impurity in which bromine was replaced by hydrogen in the final product for KGP94

(11).

94 Scheme 3.2. Improved synthetic route towards KGP94 (11) and analogues 12-13.

Synthesis of dimethylresorcinol and resorcinol analogues 19-22 utilized

commercially available 1-bromo-3,5-dimethoxy benzene as a starting material to form an

intermediate organolithium reagent which upon reaction with Weinreb 4 or 14afforded

ketones 15 and 16, respectively (Scheme 3.3). Demethylation of these 3,5-

dimethoxybenzophenone intermediates 15 and 16 with boron tribromide afforded 3,5- dihydroxybenzophenones 17 and 18. Subsequent condensation of ketones 15-18 with

thiosemicarbazide (separately) under microwave irradiation generated target

thiosemicarbazone analogues 19-22.

95

Scheme 3.3. Synthesis of dimethylresorcinol and resorcinol analogues 19-22.

In order to increase the aqueous solubility and possibly the bioavailability of

KGP94 (11), a water-soluble phosphate prodrug salt was prepared through phosphorylation of the phenolic moiety. Thus, 3-bromophenyl-3-hydroxyphenyl methanone 23 was phosphorylated with dibenzyl chlorophosphate (prepared in situ)199 to afford the corresponding dibenzyl phosphate ester 24 (Scheme 3.4). Subsequent deprotection of the benzyl groups with 33% HBr in AcOH generated phosphoric acid ester 25. Successful completion of the synthesis of the phosphate salt of benzophenone

thiosemicarbazone KGP420 (27) was accomplished by the condensation of phosphoric

acid ester 25 with thiosemicarbazide to afford benzophenone thiosemicarbazone 26, which upon reaction with sodium carbonate generated the desired disodium phosphate salt KGP420 (27).

96

Scheme 3.4. Prodrug derivatization of KGP94 (11) to form water-soluble salt KGP420.

Isomerization

Figure 3.4. ROESY and COSY correlations for analogue 13 as a representative example of the major isomer observed in DMSO-d6.

One challenge inherent to the synthesis of thiosemicarbazone based inhibitors is the propensity for isomerization about the imine bond.166,200,201 As a notable example, 2-

formylpyridine-4’,4’-dimethyl thiosemicarbazone, isolated in the Z configuration,

isomerized in solution generating varying Z/E equilibrium isomeric ratios which were

97 dependent upon on the capability of the particular solvent to disrupt the intramolecular

hydrogen bonding interaction between the pyridine nitrogen and N-H hydrogen of the

thiosemicarbazone.202 Benzophenone thiosemicarbazone analogues reported herein, except KGP420 (27, 60:40 ratio in D2O), exist predominately in the E configuration in

solution (Figure 3.4). Isomerization of KGP94 (11) and benzophenone

thiosemicarbazone analogues in DMSO-d6 occurred over a period of time (Table 3.1).

After standing for one week in DMSO-d6 at room temperature, KGP94 isomerized by the

greatest extent to an equilibrium ratio of 25%. Analogue 22 isomerized the least with

only 9% of the minor isomer present after standing in DMSO-d6 for one week.

Table 3.1. Isomerization of benzophenone thiosemicarbazone analogues in DMSO-d6

Percent Z isomer present a

Compound 0 hours 48 hours 1 week

11 5% 25%b

12 NAc NA NA

13 1.5% 17% 18%

19 4% 12% 20%

20 2% 10% 14%

21 11% 13% 13%

22 0.2% 8% 9%

a Isomerization of KGP94 and analogues were monitored by H NMR in DMSO-d6 as solvent. bReported percent of Z isomer present observed at 24 h cNot applicable

98 Biological Evaluation

Table 3.2. Inhibitory activity of benzoylbenzophenone thiosemicarbazone analogues

a IC50 Values (nM) Percent Inhibition (10 µM) Cmpd R1 R2 R3 R4 CatL CatB CatL

11 H Br H OH 131b >10000 b

12 OH Br OH Br >10000 NDc 40%

13 Br Br H OH 202 ND 94%

19 H Br OCH3 OCH3 >10000 ND 46%

20 Br Br OCH3 OCH3 >10000 ND 23%

21 H Br OH OH ~10000 ND 52%

22 Br Br OH OH >10000 ND 44%

27 H Br H O-P(O)O2Na2 >10000 ND ND

a These values are averages of a minimum of a triplicate of experiments. Each assay utilized 2% DMSO with a 5 min pre-incubation period. b Previously reported203 c Not Determined

(3,5-Dibromophenyl)-(3-hyrdroxyphenyl) ketone thiosemicarbazone (13) exhibited comparable activity to KGP94 (11) against cathepsin L with an IC50 value of

202 nM (Table 3.2). With cathepsin L inhibition of 23% - 52% at 10 μM, activity of the symmetrical, dimethylresorcinol, and resorcinol thiosemicarbazone analogues19-22

99 borders the cutoff threshold (percent inhibition ≤ 50% at 10 μM). While having one m-

hydroxyl group on the aromatic ring opposing the 3-bromophenyl substituent in the

thiosemicarbazone analogues is important for activity against cathepsin L, for this group

of compounds, the presence of two m-hydroxyl or two m-dimethoxy substituents impairs

inhibitory activity against cathepsin L.

Chemical and Enzymatic Hydrolysis

Chromatogram of 18 hours ALP-treated KGP420 Rate Study of KGP420 (from an old batch) with 2% DMSO 2.5 30 2 KGP420, 1.564 min R = 0.9168 2.0

KGP94, 5. 598 min 20 Control 1.5 ALP-treated w/ 2% DMSO 1.0 10 Area (320nm) 0.5

Absorbance (mau, 320nm) (mau, Absorbance 0 0.0 0 2 4 6 8 10 1 2 3 4 5 6 7 Time (min) Time (min)

Figure 3.5. Incubation of KGP420 with 1.2 Units ALP.

The stability of phosphate prodrug KGP420 (27) in aqueous solution was

evaluated with and without the presence of alkaline phosphatase. Significant self-

hydrolysis was not observed for prodrug KGP420 (27) after a 48 hour incubation at 37 oC in glycine buffer solution (pH 8.6) (Figure 3.5). Additionally, KGP420 was not hydrolyzed when stored as a stock solution for one week at 4 oC. Enzymatic cleavage of

prodrug KGP420 (27) in alkaline phosphatase solution occurred with nearly 100%

conversion to the anticipated parent drug KGP94 (11) over the course of 18 hours.

KGP94 (11) was formed at a rate of 0.0832 μM/min (100 µM of KGP420 (27), 0.005

100 units of alkaline phosphatase) and the specific activity of alkaline phosphatase toward

KGP420 was determined to be 16.6 µM/ min.

Cytotoxicity

Table 3.3. Cytotoxicity against HUVECs

Compound Doxorubicin Paclitaxel KGP94 (11) KGP420 (27)

* * a Cytotoxicity GI50 (µM) 0.0268 0.00148 >50 >55.9 a Previously reported204

KGP94 (11) and the corresponding prodrug KGP420 (27) were evaluated for cytotoxicity toward normal primary cells. HUVECs were used a model for normal primary cells. Both KGP94 (11) and KGP420 (27) did not exhibit significant cytotoxicity against HUVECs especially compared to FDA approved cancer therapeutics doxorubicin and paclitaxel (Table 3.3).

Conclusion

Improved methodology resulted in an alternative synthesis of KGP94 which circumvented unwanted byproduct formation. In an effort to prepare thiosemicarbazone based inhibitors which exhibit a lower threshold for isomerization, new analogues with

KGP94 as the core structure were prepared and evaluated for inhibitory activity against cathepsin L. The most potent of these, [(3,5-dibromophenyl)(3-hydroxyphenyl) ketone] thiosemicarbazone exhibited comparable activity to KGP94 with and IC50 value of 202 nM. Advancement of the pre-clinical candidate, KGP94, through phosphate prodrug derivatization to generate KGP420 resulted in a water soluble analogue which is desirable for potentially increasing oral bioavailability. In vitro studies demonstrated that KGP420

101 is hydrolyzed to the parent compound, KGP94, in the presence of alkaline phosphatase.

Additionally, KGP420 favorably displayed low cytotoxicity to HUVECs which were

used as a model for normal cells.

Experimental Section

Chemistry

General Synthetic Protocols

All reactions were performed under inert atmosphere using nitrogen gas unless

otherwise specified. Chemicals and reagents used in the synthetic procedures were

purchased from commercial suppliers and used without further purification. Anhydrous

tetrahydrofuran and dichloromethane were purchased from commercial suppliers or dried

using a VAC (Vacuum Atmospheres Co.) solvent purification system. Reactions were

monitored by normal phase thin layer chromatography (TLC) plates SiliaPlateTM (silica

gel, 250 µM, F-254, 60 Å) and with reverse phase EMD Chemicals Inc. TLC plates (C18,

200-270 µM, F-254, 60 Å). Normal phase chromatographic purification was performed

using manual flash chromatography and automated flash chromatography which was carried out with silica gel purchased from either Silicycle Inc (230–400 mesh) or Biotage

(40–65 microns). Reverse phase chromatographic purification was performed using automated flash chromatography with a C-18 column purchased from Biotage (30g,

SNAP C18-KP-HS). Intermediates and products synthesized were characterized using a

Varian Inova 500 MHz NMR system and/or Bruker Avance III HD 600 MHz NMR system. Deuterated CDCl3 (with 0.03 % TMS as internal standard), acetone-d6, DMSO-

d6, and D2O were used as solvents for recording the NMR. All the chemical shifts are

102 expressed in ppm (δ), coupling constants (J, Hz) and peak patterns are reported as broad

(br), singlet (s), doublet (d), triplet (t), quartet (q) and multiplet (m). High resolution mass spectra (HRMS) were obtained in the Baylor University Mass Spectrometry Core Facility on a Thermo Scientific LTQ Orbitrap Discovery using electrospray ionization ESI. Purity of the final compounds was analyzed using an Agilent Technologies 1200 series HPLC system with a Diode Array and Multiple Wavelength Detector SL, equipped with an

Agilent Eclipse XDB-C18 column (4.6 mm ID x 250 mm, 5 μm particle size, 80 Å pore size). HPLC method parameters: T = 25 oC; 1.0 mL/min; injection volume 20 μL;

monitored at 254 nm, 300 nm, 320 nm. Four different HPLC gradients were used for purity analysis; Method A: acetonitrile/water, gradient 30:70 to 70:30 from 0 to 20 min,

70:30 to 100:0 from 20 to 25 min, isocratic 100:00 from 25 to 30 min, gradient 100:00 to

30:70 from 30-32 min, and isocratic 30:70 from 32 to 40 min; Method B:

water/acetonitrile, gradient 30:70 to 90:10 from 0 to 25 min and isocratic 90:10 from 25

to 30 min; Method C: water/acetonitrile, gradient 50:50 to 90:10 from 0 to 25 min and

isocratic 90:10 from 25 to 30 min; Method D: 0.05 % trifluoroacetic acid in

water/acetonitrile, isocratic 10:90 from 0 to 5 min, gradient 10:90 to 100:0 from 5 to 25

min and isocratic 100:00 from 25 to 30 min.

(3-Bromophenoxy)-tert-butyl-dimethyl-silane (1).

Tert-butyl dimethylsilyl chloride (3.150 g, 21.00 mmol) was added to a solution

of imidazole (1.900 g, 27.94 mmol) and 3-bromophenol (1.520 mL, 14.01 mmol) in

anhydrous DMF (40 mL) at 0o C. The reaction mixture was stirred for 6 hrs. Upon

completion of the reaction, 5% aqueous NaHCO3 (20 ml) was added to the reaction

mixture. The products were extracted with hexanes (2 x 50 mL) and concentrated under

103 reduced pressure. Purification by flash column chromatography (silica gel, hexanes

100%) afforded (3-bromo-phenoxy)-tert-butyl-dimethyl-silane (3.905 g, 13.59 mmol,

1 97% yield) as a colorless oil. H NMR (500 MHz, CDCl3) δ 7.10-7.06 (2H, m), 7.01-7.00

13 (1H, m), 6.78-6.74 (1H, m), 0.98 (9H, s), 0.20 (6H, s). C NMR (125 MHz, CDCl3)

δ156.67, 130.55, 124.61, 123.66, 122.61, 118.96, 25.75, 18.33, -4.32.

(3,5-Dibromophenoxy)-tert-butyldimethylsilane (2).

3,5-dibromophenol (3.78 g, 15.0 mmol) was dissolved in N,N-dimethylformamide

(45 mL) followed by the addition of imidazole (2.04 g, 30.0 mmol). The reaction mixture

was cooled to 0 oC and tert-butyldimethylchlorosilane (3.37 g, 22.5 mmol) was added.

The reaction mixture was returned to room temperature and stirred for 4 h. After reaction

completion, the reaction mixture was quenched with saturated aqueous sodium

bicarbonate (50 mL) and the product was extracted with hexanes (3 X 50 mL). The

organic extracts were dried over anhydrous sodium sulfate and concentrated under

reduced pressure. The crude mixture was purified using flash chromatography (silica gel,

hexanes) to afford (3,5-dibromophenoxy)-tert-butyldimethylsilane (5.38 g, 14.7 mmol ,

1 98%). H NMR (600 MHz, CDCl3): δ 7.26 (1H, t, J = 1.7 Hz), 6.93 (2H, d, J = 1.7 Hz),

13 0.97 (9H, s), 0.21 (6H, s). C NMR (150 MHz, CDCl3): δ 156.98, 127.12, 122.76,

122.35, 25.49, 18.12, -4.53. tert-Butyldimethylsilyl 3-bromo-5-((tert-butyldimethylsilyl)oxy)benzoate (Precursor for

Compound 3).

3-Bromo-5-hydroxybenzoic acid (1.960 g, 9.031 mmol) and imidazole (4.064,

27.09 mmol) were dissolved in anhydrous dichloromethane (35 mL) followed by the

addition of tert-butyldimethylchlorosilane (3.684, 54.18 mmol) and the reaction mixture

104 was refluxed for 5 h. The reaction mixture was allowed to cool to room temperature and

was quenched with water (50 mL) and the product was extracted with dichloromethane (3

X 50 mL). The organic extracts were dried over sodium sulfate and concentrated. . The

crude mixture was purified using flash chromatography (silica gel, hexanes:ethyl acetate,

gradient, 98:02 to 70:30) to afford tert-butyldimethylsilyl 3-bromo-5-((tert-

butyldimethylsilyl)oxy)benzoate (1.587 g, 3.562 mmol, 39%). 1H NMR (600 MHz,

CDCl3): δ 7.84 (1H, t, J = 1.6 Hz), 7.47 (1H, dd, J = 2.3 Hz, 1.4 Hz), 7.23 (1H, dd, J =

2.3 Hz, 1.8 Hz), 0.99 (9H, s), 0.91 (9H, s), 0.24 (6H, s), 0.11 (6H, s). 13C NMR (150

MHz, CDCl3): δ 169.89, 156.54, 131.71, 128.66, 126.11, 122.56, 120.35, 25.62, 25.54,

18.17, 17.98, -3.60, -4.49.

3-Bromo-5-((tert-butyldimethylsilyl)oxy)benzoyl chloride (3).

Oxalyl chloride (359 μL, 4.19 mmol) was added dropwise to a solution of tert-

butyldimethylsilyl 3-bromo-5-((tert-butyldimethylsilyl)oxy)benzoate (1.494 g, 3.353

mmol) in dichloromethane (10 mL). A catalytic amount of DMF (2.6 μL, 0.034 mmol)

was added dropwise and the reaction mixture was stirred for 12 h at room temperature.

After concentration, the reaction mixture was dissolved in dichloromethane and concentrated and repeated to afford 3-bromo-5-((tert-butyldimethylsilyl)oxy)benzoyl

chloride. The product was used immediately without further purification. 1H NMR (600

MHz, CDCl3): δ 7.85 (1H, t, J = 1.7 Hz), 7.47 (1H, dd, J = 2.3 Hz, 1.7 Hz), 7.29 (1H, dd,

13 J = 2.3 Hz, 1.7 Hz), 0.99 (9H, s), 0.25 (6H, s). C NMR (150 MHz, CDCl3): δ 167.01,

156.77, 135.53, 130.03, 127.05, 122.99, 121.21, 25.50, 18.18, -4.49.

105 3-Bromo-N-methoxy-N-methylbenzamide (4).

To a well stirred suspension of N,O-dimethylhydroxylamine hydrochloride (5.33

g, 54.7 mmol) in dichloromethane (120 mL) was added triethylamine (10.2 mL, 72.9

mmol) dropwise at 0 oC. After stirring for a few minutes, 3-bromobenzoyl chloride (4.81

mL, 36.4 mmol) in dichloromethane (20 mL) was added dropwise. The reaction mixture

was allowed to warm to room temperature and stirred for 4 h. The reaction mixture was

quenched with water (150 mL) and extracted with dichloromethane (3 x 100 mL). The

combined organic phases were dried over anhydrous sodium sulfate and concentrated.

Purification using flash chromatography (silica gel, hexane: ethyl acetate, gradient, 90:10

to 60:40) afforded 3-bromo-N-methoxy-N-methyl-benzamide as a colorless oil (8.604 g,

1 35.25 mmol, 97% yield). H NMR (500 MHz, CDCl3): 7.83 (1H, t, J = 1.8 Hz), 7.61 (1H,

dt, J = 7.7 Hz, 1.3 Hz), 7.59 (1H, ddd, J = 8.0 Hz, 2.1 Hz, 1.1 Hz), 7.28 (1H, m), 3.35

13 (3H, s), 3.36 (3H, s). C NMR (125 MHz, CDCl3): 168.32, 136.06, 133.69, 131.36,

129.74, 126.93, 122.14, 61.33, 33.71.

3-Bromo-5-((tert-butyldimethylsilyl)oxy)-N-methoxy-N-methylbenzamide (5).

Triethylamine (0.941 mL, 6.70 mmol) was added dropwise to a solution of N,O- dimethylhydroxylamine hydrochloride ( 0.512 g, 5.03 mmol) in dichloromethane (12 mL) at 0 oC. A solution of 3-bromo-5-((tert-butyldimethylsilyl)oxy)benzoyl chloride

(11.7 g, 3.35 mmol) in dichloromethane (3 mL) was added dropwise to the reaction

mixture and the ice bath was removed. After 3 h, the reaction was quenched with water

(50 mL) and the product was extracted using dichloromethane (3 x 20 mL). The

combined organic phases were dried over sodium sulfate and concentrated. Purification

using flash chromatography (silica gel, hexanes:ethyl acetate, gradient 90:10 to 40:60)

106 afforded 3-bromo-5-((tert-butyldimethylsilyl)oxy)-N-methoxy-N-methylbenzamide (1.10

1 g, 2.94 mmol, 88% yield over two steps). H NMR (600 MHz, CDCl3): δ 7.40 (1H, t, J =

1.6 Hz), 7.08 (1H, dd, J = 2.3 Hz, 1.8 Hz), 7.07-7.06 (1H, m), 3.55 (3H, s), 3.34 (3H, s),

13 0.98 (9H, s), 0.21 (6H, s). C NMR (150 MHz, CDCl3): δ 167.91, 156.00, 136.44,

125.45, 124.15, 122.06, 118.71, 61.21, 25.55, 18.16, -4.49.

3-((tert-Butyldimethylsilyl)oxy) benzaldehyde (6).

3-hydroxybenzaldehyde (2.000 g, 16.38 mmol) was dissolved in N,N-

dimethylformamide (50 mL) followed by the addition of imidazole (2.227 g, 32.75

mmol). The reaction mixture was cooled to 0 oC and tert-butyldimethylchlorosilane

(3.684 g, 24.56 mmol) was added. The reaction mixture was returned to room

temperature and stirred for 4 h. After reaction completion, the reaction mixture was

quenched with saturated aqueous sodium bicarbonate (50 mL) and the product was

extracted with hexanes (2 x 50 mL). The organic extracts were dried over anhydrous

sodium sulfate and concentrated under reduced pressure. The crude mixture was purified

using flash chromatography (silica gel, hexanes:ethyl acetate, gradient, 100:00 to 90:10)

to afford 3-((tert-Butyldimethylsilyl)oxy) benzaldehyde (3.568 g, 15.09 mmol , 92%

1 yield). H NMR (500 MHz, CDCl3): δ 9.95 (1H, s), 7.47 (1H, dt, J = 7.3 Hz, 1.2 Hz),

7.40 (1H, t, J = 7.8 Hz), 7.34-7.32 (1H, m), 7.12-7.09 (1H, m), 1.0 (9H, m), 0.23 (6H, m).

13 C NMR (125 MHz, CDCl3): δ 192.23, 156.54, 138.07, 130.21, 126.68, 123.69, 120.01,

25.76, 18.34, -4.28.

[3-(t-Butyldimethylsilyl)oxyphenyl]-(3-bromophenyl) methanone (7).

The (4-bromophenoxy)-tert-butyl-dimethyl-silane (3.905 g, 13.59 mmol) was dissolved in THF (20 mL) and stirred for 5 min. The solution was cooled to -78 oC and

107 stirred for an additional 10 min before dropwise addition of n-butyllithium (2.96 mL, 6.8

mmol). The solution was allowed to stir for 1 hour and 20 minutes. A solution of 3-

Bromo-N-methoxy-N-methylbenzamide (1.508 g, 6.17 mmol) in 5 mL THF was added

and allowed to stir for 2 hours at -78 oC. The ice bath was removed and the reaction mixture was allowed to stir for 30 minutes. The reaction mixture was quenched with 20

mL of 1 M HCl, and the organic phase was extracted with chloroform (2 x 50 mL). The

organic phase was washed three times with saturated sodium bicarbonate. The organic phase was separated, dried over sodium sulfate, and concentrated under reduced pressure.

Purification by flash column chromatography (silica gel, hexanes:EtOAc, gradient 100:0

to 85:15) afforded [3-(t-Butyldimethylsilyl)oxyphenyl]-(3-bromophenyl)methanone

1 (2.184 g, 5.58 mmol, 91% yield). H NMR (600 MHz, DMSO-d6) δ 7.89 (1H, ddd, J =

8.0 Hz, 2.1 Hz, 1.0 Hz), 7.83 (1H, t, J = 1.8 Hz), 7.71 (1H, ddd, J = 7.7 Hz, 1.6 Hz, 1.0

Hz), 7.53 (1H, t, J = 7.9 Hz), 7.47 (1H, t, J = 7.9 Hz), 7.34 (1H, ddd, J = 7.6 Hz, 1.6 Hz,

1.0 Hz), 7.20 (1H, ddd, J = 8.1 Hz, 2.6 Hz, 1.0 Hz), 7.14 (1H, dd, J = 2.5 Hz, 1.6 Hz),

13 0.95 (s, 9H), 0.20 (s, 6H). C NMR (150 MHz, DMSO-d6) δ 193.90, 155.11, 139.16,

137.84, 135.32, 131.84, 130.82, 130.19, 128.56, 124.80, 123.09, 121.80, 120.57, 25.55,

18.01, -4.54. Note: The above reaction was run several times on the scale above to generate the required amount of intermediate used for the synthesis of final compounds

11 and 27.

Bis(3-bromo-5-((tert-butyldimethylsilyl)oxy)phenyl) methanone (8).

(3,5-Dibromophenoxy)-tert-butyldimethylsilane (1.864 g, 5.091 mmol) was

dissolved in THF (12 mL) followed by the addition of n-butyllithium (1.6 M, 1.43 mL) at

-78 oC. After 1 h, a solution of 3-bromo-5-((tert-butyldimethylsilyl)oxy)-N-methoxy-N-

108 methylbenzamide (0.953 g, 2.55 mmol) in THF (15 mL) was added dropwise to the

reaction mixture. After 3 h, the reaction mixture was quenched with hydrochloric acid

(1M, 50mL) and the products were extracted with dichloromethane (3 x 50 mL). The

combined organic phases were washed with sodium bicarbonate (50 mL), dried over

anhydrous sodium sulfate, and concentrated. Purification using flash chromatography

(silica gel, hexanes: ethyl acetate, gradient 97:3 to 90:10) afforded bis(3-bromo-5-((tert- butyldimethylsilyl)oxy)phenyl) methanone (0.930 g, 1.55 mmol, 67%). 1H NMR (600

MHz, CDCl3): δ 7.48 (2H, t, J = 1.6 Hz), 7.23 (2H, dd, J = 2.3 Hz, 1.8 Hz), 7.13 (2H, dd,

13 J = 2.3 Hz, 1.4 Hz), 0.98 (18H, s), 0.23 (12H, s). C NMR (150 MHz, CDCl3): δ 192.24,

155.59, 138.47, 126.72, 124.95, 121.82, 119.24, 76.37, 76.16, 75.95, 24.70, 17.33, -5.28.

3-((t-Butyldimethylsilyl)oxy)phenyl)-(3,5-dibromophenyl) methanol (9).

tert-Butyllithium (1.7 M, 13.24 mL) was added dropwise to a solution of 1,3,5-

tribromobenzene (1.77 g, 17.88 mmol) in diethyl ether (100 mL) at -78 oC. The reaction

mixture was sonicated for 1 min at 30 min intervals. After 1.5 h, a solution of (2.660 g,

11.25 mmol) in diethyl ether (10 mL) was added dropwise to the reaction mixture and

stirred at -78 oC. After 2 h, the dry ice bath was removed and the reaction mixture was

stirred for 18 h. The reaction was quenched with 100 mL of water and extracted with

ethyl acetate (2 X 50 mL) followed by dichloromethane (2 X 50 mL). The combined

organic extracts were dried over sodium sulfate and concentrated. Purification using flash chromatography (silica gel, hexanes:ethyl acetate, gradient 90:10 to 40:60) afforded (3-

Bromophenyl)-(3,5-methoxyphenyl) methanol (4.047 g, 8.57 mmol) in a 76% yield. 1H

NMR (600 MHz, CDCl3): δ 7.55 (1H, t, J = 1.8 Hz), 7.46 (2H, dd, J = 1.8, 0.7 Hz), 7.22

(1H, t, J = 7.8 ), 6.91 (1H, m), 6.82 (1H, t, J = 2.1 Hz), 6.78 (1H, ddd, J = 8.1, 2.5, 1.0 Hz

109 13 ), 5.69 (1H, s), 2.26 (1H, d, J = 3.1 Hz). C NMR (150 MHz, CDCl3): δ 156.03, 147.36,

144.10, 132.97, 129.87, 128.21, 122.93, 119.90, 119.45, 188.29, 74.85, 25.66, 18.23, -

4.40.

3-((t-Butyldimethylsilyl)oxy)phenyl)-(3,5-dibromophenyl) methanone (10).

A solution of 3-(t-butyldimethylsilyl)oxyphenyl)-(3,5-dibromophenyl) methanol

(3.624 g, 7.673 mmol) in dichloromethane (10 mL) was added dropwise to a suspension

of pyridinium chlorochromate (2.474 g, 11.51 mmol) and celite (2.5 g) in anhydrous

dichloromethane (40 mL) at 0 oC. The ice bath was removed and the reaction was stirred

at room temperature. After 5 h, the reaction mixture was filtered over a pad a celite and

rinsed with dichloromethane (5 X 30 mL). The solvent was removed under reduced

pressure and purification using flash chromatography (silica gel, hexanes:ethyl acetate,

gradient 100:00 to 90:10) afforded 3-(t-Butyldimethylsilyl)oxyphenyl)-(3,5-

dibromophenyl) methanone (3.523 g, 7.491 mmol) in a 98% yield. 1H NMR (500 MHz,

CDCl3): δ 7.88 (1H, t, J = 1.8 Hz), 7.84 (1H, d, J = 1.8 Hz), 7.37 (1H, td, J = 7.7, 0.5 Hz),

7.33 (1H, dt, J = 7.7, 1.5 Hz), 7.22 (1H, ddd, J = 2.5, 1.5, 0.5 Hz), 7.10 (1H, ddd, J = 7.7,

13 2.5, 1.5 Hz), 1.00 (s, 9H), 0.23 (s, 6H). C NMR (125 MHz, CDCl3): δ 193.24, 155.89,

140.67, 137.61, 137.55, 131.47, 129.72, 125.15, 123.03, 121.15, 25.64, 18.24, -4.37.

[[3-(t-Butyldimethylsilyl)oxyphenyl]-(3-bromophenyl)-ketone] thiosemicarbazone

(Intermediate between compound 7 and 11).

[3-(t-Butyldimethylsilyl)oxyphenyl]-(3-bromophenyl) methanone (2.011 g, 5.16

mmol) was dissolved in anhydrous methanol (25 mL), followed by the addition of p-

toluenesulfonic acid monohydrate (0.020 g, .105 mmol) and thiosemicarbazone (0.939 g,

10.32 mmol). The reaction mixture was heated to reflux and stirred under an inert

110 atmosphere of nitrogen for 9 h. After reaction completion, methanol was removed under

reduced pressure. Products were extracted into EtOAc (2 x 100 mL) from 100 mL of

water. The combined organic phases were washed with brine, dried over anhydrous

Na2SO4, and concentrated under reduced pressure. Purification by flash column chromatography (silica gel, hexanes:EtOAc, gradient 90:10 to 70:30) afforded [[3-(t-

Butyldimethylsilyl)oxyphenyl]-(3-bromophenyl)-ketone] thiosemicarbazone (1.764 g,

1 3.80 mmol, 73 % yield) as a light yellow solid. H-NMR (DMSO-d6, 600 MHz) δ 8.70

(1H, s), 8.57 (1H, s), 8.45 (1H, s), 8.04 (1H, s), 7.59 (1H, ddd, J = 8.0 Hz, 2.0 Hz, 1.0

Hz), 7.55 (1H, t, J = 7.9 Hz), 7.46 (1H, ddd, J = 8.0 Hz, 1.7 Hz, 1.0 Hz ) 7.31 (1H, t, J =

8.0 Hz), 7.10 (1H, ddd, J = 8.3 Hz, 2.5 Hz, 1.0 Hz), 6.94 (1H, ddd, J = 7.5 Hz, 1.5 Hz,

1.0 Hz), 6.81 (1H, dd, J = 2.5 Hz, 1.5 Hz), 0.945 (9H, s), 0.21 (6H, s). 13C-NMR

(DMSO-d6, 150 MHz) δ 177.87, 156.21, 146.80, 138.57, 132.36, 132.03, 131.59, 130.43,

129.46, 126.81, 122.18, 121.87, 121.27, 119.73, 25.59, 18.05, -4.51.

[(3-Bromophenyl)-(3-hydroxyphenyl)-ketone] thiosemicarbazone (11).

[3-(t-Butyldimethylsilyl)oxyphenyl]-(3-bromophenyl) methanone (1.764 g, 3.80

mmol) was dissolved in 30 mL of tetrahydrofuran and tetra-n-butyl ammonium fluoride

trihydrate (2.396 g, 7.60 mmol) was added. The reaction mixture was stirred at room

temperature under an inert atmosphere of nitrogen gas for 1.5 hrs. After reaction

completion, the reaction mixture was diluted with ethyl acetate and washed with brine.

The combined organic phases were washed with brine, dried over anhydrous Na2SO4, and concentrated under reduced pressure. Purification by flash column chromatography

(silica gel, hexanes:EtOAc, gradient 90:10 to 20:80) afforded [(3-Bromophenyl)-(3- hydroxyphenyl)-ketone] thiosemicarbazone (1.070 g, 3.05 mmol, 80 % yield) as a light

111 1 yellow solid. H-NMR (DMSO-d6, 600 MHz) δ 10.00 (1H, s), 8.70 (1H, s), 8.57 (1H, s),

8.40 (1H, s), 8.10 (1H, s), 7.59 (1H, ddd, J = 7.9 Hz, 2.0 Hz, 1.0 Hz), 7.46 (1H, t, J = 7.8

Hz), 7.44 (1H, ddd, J = 7.9 Hz, 1.7 Hz, 1.0 Hz), 7.31 (1H, t, J = 7.9 Hz), 7.00 (1H, ddd, J

= 8.3 Hz, 2.5 Hz, 1.0 Hz), 6.73 (1H, ddd, J = 7.4 Hz, 1.5 Hz, 1.0 Hz), 6.65 (1H, dd, J =

13 2.5 Hz, 1.5 Hz). C-NMR (DMSO-d6, 150 MHz) δ 177.76, 158.53, 147.38, 138.51,

132.38, 131.71, 131.42, 130.46, 129.33, 126.98, 122.21, 118.41, 117.24, 114.53. HPLC

retention time (Method A): 12.07 min for major isomer, 10.75 min for minor isomer.

[Bis(3-bromo-5-((tert-butyldimethylsilyl)oxy)phenyl) ketone] thiosemicarbazone

(Intermediate between compound 8 and 12).

Bis(3-bromo-5-((tert-butyldimethylsilyl)oxy)phenyl) methanone (0.150 g, 0.250

mmol), thiosemicarbazide (0.0455 g, 0.500 mmol), and p-toluenesulfonic acid

monohydrate (0.006 g, 0.03 mmol) were dissolved in anhydrous tetrahydrofuran (5.0 mL)

and refluxed for 27 h. The solvent was removed under reduced pressure. Purification

using flash chromatography (silica gel, hexanes:ethyl acetate, gradient 99:01 to 80:20)

afforded [bis(3-bromo-5-((tert-butyldimethylsilyl)oxy)phenyl) ketone]

1 thiosemicarbazone (0.151 g, 0.224 mmol) in a 90% yield. H NMR (600 MHz, CDCl3):

δ 8.82 (1H, s), 8.21 (1H, s), 7.79 (1H, s), 7.73 (1H, t, J = 1.6 Hz), 7.31 (1H, t, J = 2.0 Hz),

7.22 (1H, t, J = 1.6 Hz), 7.10 (1H, t, J = 2.0 Hz), 7.00 (1H, t, J = 1.8 Hz), 6.79 (1H, t, J =

1.8 Hz), 1.01 (9H, s), 0.94 (9H, s), 0.30 (6H, s), 0.17 (6H, s). 13C NMR (150 MHz,

CDCl3): δ 180.63, 158.63, 157.30, 146.00, 140.40, 135.16, 125.87, 125.21, 124.89,

124.68, 123.44, 123.32, 120.21, 119.87, 25.94, 25.93, 18.82, 18.80, -4.37, -4.39.

112 [Bis(3-bromo-5-hydroxy) ketone] thiosemicarbazone (12).

Tetra-n-butylammonium fluoride (0.232 g, 0.736 mmol) was added to a solution

of [bis(3-bromo-5-((tert-butyldimethylsilyl)oxy)phenyl) ketone] thiosemicarbazone

(0.124 g, 0.184 mmol) in tetrahydrofuran (3 mL) and the reaction mixture was allowed to

stir for 50 min. The solvent was removed under reduced pressure and the product was

extracted from water (10 mL) with ethyl acetate (3 x 10 mL). The combined organic

phases were dried over sodium sulfate and concentrated. Purification using flash

chromatography (silica gel, dichloromethane:methanol, gradient 98:02 to 85:15) afforded

[bis(3-bromo-5-hydroxy) ketone] thiosemicarbazone (0.0703 g, 0.158 mmol) in a 86%

1 yield. H NMR (600 MHz, acetone-d6): δ 9.14 (2H, brs), 8.75 (1H, brs), 8.24 (1H, brs),

7.80 (1H, brs), 7.50 (1H, t, J = 1.6 Hz), 7.26 (1H, t, J = 2.0 Hz), 7.06-7.04 (2H, m), 6.89

(1H, dd, J = 2.3 Hz, 1.5 Hz), 6.85 (1H, dd, J = 2.2 Hz, 1.3 Hz). 13C NMR (150 MHz,

acetone-d6): δ 180.55, 160.39, 159.08, 146.66, 140.37, 135.21, 124.76, 123.32, 122.87,

121.72, 121.07, 120.26, 115.35, 115.17. HPLC retention time (Method B): 10.25 min.

- - HRMS (ESI) calculated for C14H10Br2N3O2S [M-H] 441.88660, found 441.88740.

[3-((t-Butyldimethylsilyl)oxy)phenyl)-(3,5-dibromophenyl)-ketone] thiosemicarbazone

(Intermediate between compound 10 and 13).

(3,5-Dibromophenyl)-(3-methoxyphenyl) methanone (0.470 g, 1.00 mmol),

thiosemicarbazide (0.455 g, 5.00 mmol), and p-toluenesulfonic acid monohydrate (0.0038

g, 0.020 mmol) were dissolved in anhydrous ethanol (3.0 mL). The reaction was carried

out at 100 oC for 2 h under microwave irradiation. The solvent was removed under

reduced pressure. Purification using flash chromatography (silica gel, hexanes:ethyl

acetate, gradient 95:05 to 60:40) afforded [3-(t-Butyldimethylsilyl)oxyphenyl)-(3,5-

113 dibromophenyl)-ketone] thiosemicarbazone (0.154 g, 0.283 mmol) in a 28% yield. 1H

NMR (600 MHz, acetone-d6): δ 9.11 (0.15H, s), 8.63 (0.85H, s), 8.37 (0.85H, s), 8.03

(0.15H, s), 7.99 (0.15H, t, J = 1.8 Hz), 7.85 (0.85H, s), 7.81 (1.7H, d, J = 1.8 Hz), 7.78

(0.85H, t, J = 1.8 Hz), 7.73 (0.15H, s), 7.65 (0.3H, d, J = 1.8 Hz), 7.62-7.59 (0.85H, m),

7.34-7.31 (0.15H, m), 7.28 (0.15H, td, J = 7.9 Hz, 0.5 Hz), 7.17 (0.85 H, ddd, J = 8.3 Hz,

2.5 Hz, 1.0 Hz), 7.03 (0.85H, ddd, J = 7.5 Hz, 1.5 Hz, 1.0 Hz), 6.98-6.96 (1.0H, m), 6.95

(0.15H, ddd, J = 7.9 Hz, 2.5 Hz, 1.1 Hz), 1.00 (7.65H, s), 0.95 (1.35H, s), 0.27 (5.1H, s),

13 0.17 (0.9H, s). C NMR (150 MHz, acetone-d6): δ 180.45, 157.94, 146.41, 141.60,

135.23, 132.58, 132.52, 129.94, 123.60, 123.28, 122.17, 120.92, 26.02, 18.85, -4.27.

[(3,5-Dibromophenyl)-(3-hyrdroxyphenyl) ketone] thiosemicarbazone (13).

Tetra-n-butylammonium fluoride ( 1.0 M in THF, 1.25 mL) was added dropwise

to a solution of [3-(t-butyldimethylsilyl)oxyphenyl)-(3,5-dibromophenyl)-ketone]

thiosemicarbazone (0.134 g, 0.246 mmol) in tetrahydrofuran (2 mL) and the reaction

mixture was allowed to stir for 1.5 h. The solvent was removed under reduced pressure

and the product was extracted from water (10 mL) with ethyl acetate(3 x 10 mL). The

combined organic phases were dried over sodium sulfate and concentrated. Purification

using flash chromatography (silica gel, hexanes:ethyl acetate, gradient 90:10 to 40:60)

afforded [(3-Bromophenyl)-(3,5-methoxyphenyl) ketone] thiosemicarbazone (0.089 g,

1 0.21 mmol) in a 85% yield. H NMR (600 MHz, DMSO-d6): δ 10.04 (1H, s), 8.77 (1H,

s), 8.71 (1H, s), 8.44 (1H, s), 7.87 (1H, t, J = 1.8 Hz), 7.83 (2H, m), 7.48 (1H, t, J = 7.9

Hz), 7.01 (1H, ddd, J = 8.4, 2.5, 1.0 Hz), 6.74 (1H, dt, J = 7.5, 1.3 Hz), 6.68 (1H, t, J =

13 2.0 Hz). C NMR (150 MHz, DMSO-d6): δ 177.84, 158.58, 145.83, 140.22, 134.17,

131.56, 131.09, 128.92, 122.77, 118.44, 117.47, 114.54. HPLC retention time (Method

114 C): 7.54 min for major isomer, 5.84 min for minor isomer. HRMS (ESI) calculated for

+ + C14H11Br2N3OSH [M+H] 427.90623, found 427.90685.

3,5-Dibromobenzoyl chloride (Precursor for Compound 14).

Oxalyl Chloride (1.10 mL, 12.9 mmol) was added dropwise to a solution of 3,5-

dibromobenzoic acid in anhydrous dichloromethane (50 mL). After 10 min, a catalytic

amount of N,N-dimethylformamide (0.0066 μL, 0.086 mmol) was added to the reaction

mixture. After 3.5 h, the reaction was concentrated. The product was dissolved in

anhydrous dichloromethane (20 mL) and the solvent was removed under reduced

pressure. The crude product was used immediately without further purification. 1H NMR

(600 MHz, CDCl3): δ 8.18 (2H, d, J = 1.8Hz), 7.98 (1H, t, J =1.8 Hz).

3,5-Dibromo-N-methoxy-N-methyl-benzamide (14).

Triethylamine (1.73 mL, 17.1 mmol) was added dropwise to a solution of N,O- dimethylhydroxylamine hydrochloride ( 1.30 g, 12.9 mmol) in dichloromethane (50 mL) at 0 oC. A solution of 3,5-dibromobenzoyl chloride in dichloromethane (10 mL) was

added dropwise to the reaction mixture and the ice bath was removed. After 3 h, the

reaction was quenched with water (100 mL) and the product was extracted using

dichloromethane (3 x 50 mL). The combined organic phases were dried over sodium

sulfate and concentrated. Purification using flash chromatography (silica gel,

hexanes:ethyl acetate, gradient 90:10 to 20:80) afforded 3,5-dibromo-N-methoxy-N-

methyl-benzamide (2.39 g,7.40 mmol) in a 86% yield over two steps. 1H NMR (600

13 MHz, CDCl3): δ 7.77-7.75 (3H, m), 3.56 (3H, s), 3.36 (3H, s). C NMR (150 MHz,

CDCl3): δ 166.59, 137.06, 136.00, 130.02, 122.54, 61.39, 33.30.

115 (3-Bromophenyl)-(3,5-dimethoxyphenyl) methanone (15).

n-Butyllithium in hexanes (2.5 M, 2.88 mL) was added dropwise to a solution of

1-bromo-3,5-dimethoxybenzene (2.60 g, 12.0 mmol) in THF (33 mL) cooled to -78 oC.

After 30 minutes a solution of 3-bromo-N-methoxy-N-methyl-benzamide (1.96 g, 8 mmol) in tetrahydrofuran (7 mL) was added to the reaction mixture and allowed to stir for 2 h at -78 oC. After 2 h, the reaction mixture was quenched with water (50 mL) and extracted with dichloromethane (3 x 50 mL). The combined organic phases were dried over sodium sulfate and concentrated. Purification using flash chromatography (silica gel, hexanes:ethyl acetate, gradient 95:05 to 20:80) (3-bromophenyl)-(3,5-dimethoxyphenyl) methanone (1.97 g, 6.13 mmol) in a 85% yield as a yellow solid. 1H NMR (600 MHz,

CDCl3): δ 7.95 (1H, t, J = 1.8 Hz), 7.71 (2H, dd, J = 1.8 Hz), 7.36 (1H, t, J = 7.8 Hz),

6.90 (2H, d, J = 2.3 Hz), 6.69 (1H, t, J = 2.3 Hz), 3.83 (6H, s). 13C NMR (150 MHz,

CDCl3): δ 194.84, 160.63, 139.37, 138.71, 135.31, 132.73, 129.81, 128.52, 122.55,

107.84, 105.10, 55.64.

(3,5-Dibromophenyl)-(3,5-dimethoxyphenyl) methanone (16).

To a solution of 1-bromo-3,5-dimethoxybenzene (2.28 g, 10.5 mmol) in

tetrahydrofuran (33 mL) cooled to -78 oC was added n-butyllithium in hexanes (2.5 M,

2.52 mL) dropwise. After 40 min, a solution of 3,5-dibromo-N-methoxy-N-methyl-

benzamide (2.26 g, 8.7 mmol) in tetrahydrofuran (7 mL) was added dropwise and the

reaction mixture was allowed to stir for 1.5 h. The reaction was quenched with 1 M HCl

(50 mL) and extracted with dichloromethane (3 x 50 mL). The combined organic phases were dried over sodium sulfate and concentrated. Purification using flash chromatography (silica gel, hexanes:ethyl acetate, gradient 98:2 to 80:20) afforded (3,5-

116 dibromophenyl)-(3,5-dimethoxyphenyl) methanone (0.935 g, 2.34 mmol) in a 37% yield

1 as a white solid. H NMR (600 MHz, CDCl3): δ 7.88 (1H, t, J = 1.8 Hz), 7.84 (2H, d, J =

1.8 Hz), 6.87 (2H,d, J = 2.3 Hz), 6.71 (1H, t, J = 2.3 Hz), 3.84 (6H, s). 13C NMR (150

MHz, CDCl3): δ 193.39, 160.74, 140.59, 138.06, 137.59, 131.41, 123.02, 107.80, 105.44,

55.68.

(3-Bromophenyl)-(3,5-dihydroxyphenyl) methanone (17).

(3-Bromophenyl)-(3,5-dimethoxyphenyl) methanone ( 1.45 g, 4.51mmol) was dissolved in anhydrous dichloromethane (20 mL) and cooled to 0 oC in an ice bath. Boron

tribromide in dichloromethane (1 M, 9.93 mL) was added dropwise to the reaction

mixture and the ice bath was removed. After 7 h, boron tribromide in dichloromethane (1

M, 5 mL) was added to the reaction mixture. After an additional 17 h, the reaction

mixture was quenched with hydrochloric acid (1M, 40 mL) and the products were

extracted with ethyl acetate (3 x 40 mL). The combined organic phases were washed with

sodium bicarbonate (50 mL), dried over anhydrous sodium sulfate, and concentrated.

Purification using flash chromatography (silica gel, hexanes: ethyl acetate, gradient 95:5

to 90:10) afforded (3-bromophenyl)-(3,5-dihydroxyphenyl) methanone (0.955 g, 3.26

1 mmol) in a 72% yield. H NMR (600 MHz, acetone-d6): δ 8.70 (2H, s), 7.90 (1H, t, J =

1.8 Hz), 7.83 (1H, ddd, J = 8.0 Hz, 2.1 Hz, 1.0 Hz), 7.75 (1H, dt, J = 7.7 Hz, 1.3 Hz) 7.54

(1H, t, J = 7.8 Hz), 6.74 (2H, d, J = 2.2 Hz), 7.64 (1H, t, J = 2.2 Hz). 13C NMR (150

MHz, acetone-d6): δ 193.89, 158.59, 140.01, 138.91, 134.93, 132.02, 130.34, 128.44,

121.88, 108.29, 106.95.

117 (3,5-Dibromophenyl)-(3,5-dihydroxyphenyl) methanone (18).

(3,5-Dibromophenyl)-(3,5-dimethoxyphenyl) methanone (0.606 g, 1.51 mmol)

was dissolved in anhydrous dichloromethane (5 mL) and cooled to 0 oC. Boron

tribromide in dichloromethane (1 M, 3.3 mL) was added dropwise to the reaction mixture

and the ice bath was removed. After 7 h, boron tribromide in dichloromethane (1 M, 3.3

mL) was added dropwise to the reaction mixture. After an additional 17 h, the reaction

was quenched with 1 M HCl (20 mL) and the product was extracted with ethyl acetate (3

x 25 mL). The combined organic extracts were washed with sodium bicarbonate, dried

over sodium sulfate and concentrated. Purification using flash chromatography (silica gel,

hexanes:ethyl acetate, gradient 88:12 to 0:100) afforded (3,5-dibromophenyl)-(3,5-

dihydroxyphenyl) methanone (0.254 g, 0.683 mmol) in a 45% yield. 1H NMR (600 MHz, acetone-d6): δ 8.76 (2H, s), 8.04 (1H, t, J = 1.8 Hz), 7.87 (2H, d, J = 1.8 Hz), 6.75 (2H, d,

13 J = 2.2 Hz) 6.65 (1H, t, J = 2.2 Hz). C NMR (150 MHz, acetone-d6): δ 193.35, 159.56,

142.29, 139.12, 137.79, 131.99, 123.47, 109.20, 180.21.

[(3-Bromophenyl)-(3,5-dimethoxyphenyl) ketone] thiosemicarbazide (19).

The (3-Bromophenyl)-(3,5-dimethoxyphenyl) methanone (0.204 g, 0.64 mmol), thiosemicarbazide (0.114 g, 1.25 mmol), p-toluene sulfonic acid monohydrate (0.0059 g,

0.031mmol) were dissolved in anhydrous methanol (1 mL), sonicated for 30 s, and the reaction was carried out at 90 Ԩ for 1 h under microwave irradiation. The solvent was removed under reduced pressure and the product was extracted from water (5 mL) with ethyl acetate (3 x 5 mL). The combined organic phases were dried over sodium sulfate and concentrated. Purification using flash chromatography (silica gel, hexanes:ethyl acetate, gradient 90:10 to 40:60) afforded [(3-Bromophenyl)-(3,5-dimethoxyphenyl)

118 ketone] thiosemicarbazone (0.207 g, 0.525 mmol) in a 84% yield. 1H NMR (600 MHz,

DMSO-d6): δ 8.70 (1H, s), 8.59 (1H, s), 8.44 (1H, s), 8.14 (1H, s), 7.59 (1H, ddd, J = 7.9

Hz, 2.0 Hz, 1.0 Hz), 7.45 (1H, ddd, J = 7.9 Hz, 1.6 Hz, 1.0 Hz), 7.31 (1H, t, J = 8.0 Hz),

6.72 (1H, t, J = 2.3 Hz), 6.47 (2H, d, J = 2.3 Hz), 3.79 (6H, s). 13C NMR (150 MHz,

DMSO-d6): δ 177.82, 161.67, 147.06, 138.35, 132.51, 132.35, 130.45, 129.25, 126.99,

122.21, 105.89, 101.51, 55.59. HPLC retention time (Method C): 9.17 min for major

+ isomer, 8.10 min for minor isomer. HRMS (ESI) calculated for C16H15Br2N3O2SNa

[M+Na]+ 416.00388, found 416.00403.

[(3,5-Dibromophenyl)-(3,5-dimethoxyphenyl) ketone] thiosemicarbazone (20).

(3,5-Dibromophenyl)-(3,5-dimethoxyphenyl) methanone (0.150 g, 0.375 mmol),

thiosemicarbazide (0.0683 g, 0.750 mmol), and p-toluenesulfonic acid monohydrate

(0.0035 g, 0.018 mmol) were dissolved in anhydrous methanol (1.0 mL). The reaction was carried out at 90 oC for 1 h under microwave irradiation. The solvent was removed

under reduced pressure and the product was extracted from water (15 mL) with ethyl

acetate (3 x 10 mL). The combined organic phases were dried over sodium sulfate and

concentrated. Purification using flash chromatography (silica gel, hexanes:ethyl acetate, gradient 93:07 to 50:50) afforded [(3,5-dibromophenyl)-(3,5-dimethoxyphenyl) ketone] thiosemicarbazone (0.120 g, 0.254 mmol) in a 68% yield. 1H NMR (600 MHz, DMSO-

d6): δ 8.76 (1H, s), 8.72 (1H, s), 8.48 (1H, s), 7.87-7.84 (3H, m), 6.73 (1H, t, J = 2.3 Hz),

13 6.50 (2H, t, J = 2.3 Hz), 3.80 (6H, s). C NMR (150 MHz, DMSO-d6): δ 177.91, 161.73,

145.55, 140.10, 134.14, 131.88, 128.89, 122.76, 105.93, 101.67, 55.62. HPLC retention

time (Method C): 13.57 min for major isomer, 11.12 min for minor isomer. HRMS (ESI)

+ + calculated for C16H15Br2N3O2SH [M+H] 471.93245, found 471.93283.

119 [(3-Bromophenyl)-(3,5-dihydroxyphenyl) ketone] thiosemicarbazide (21).

The (3-bromophenyl)-(3,5-dihydroxyphenyl) methanone (0.153 g, 0.52 mmol),

thiosemicarbazide ( 0.0937 mg, 1.03 mmol), p-toluene sulfonic acid monohydrate

(0.0054 g, 0.028 mmol ) were dissolved in anhydrous methanol (1 mL), sonicated for 30

s, and the reaction was carried out at 90 oC for 30 min under microwave irradiation. The

solvent was removed under reduced pressure and the product was extracted from water (5

mL) with ethyl acetate (3 x 5 mL). The combined organic phases were dried over sodium

sulfate and concentrated. Purification using flash chromatography (silica gel,

hexanes:ethyl acetate, gradient 80:20 to 40:60) afforded [(3-bromophenyl)-(3,5-

dihydroxyphenyl) ketone] thiosemicarbazone (0.110 g, 0.300 mmol) in a 58% yield. 1H

NMR (600 MHz, DMSO-d6): δ 9.85 (2H, s), 8.70 (1H, s), 8.56 (1H, s), 8.43 (1H, s), 8.11

(1H, s), 7.59 (1H, ddd, J = 8.0, 2.0, 1.0 Hz), 7.49 (1H, ddd, J = 8.0, 1.7, 1.0 Hz), 7.32

(1H, t, J = 8.0 Hz), 6.41 (1H, t, J = 2.2 Hz), 6.08 (2H, d, J = 2.2 Hz). 13C NMR (150

MHz, DMSO-d6): δ 177.65, 159.90, 147.54, 138.31, 132.38, 132.08, 130.46, 129.27,

126.99, 122.17, 105.44, 103.99. HPLC retention time (Method B): 6.15 min for major

+ isomer, 7.51 min for minor isomer. HRMS (ESI) calculated for C14H12BrN3O2SH

[M+H]+359.99064, found 365.99097.

[(3,5-Dibromophenyl)-(3,5-dihydroxyphenyl) ketone] thiosemicarbazone (22).

(3,5-Dibromophenyl)-(3,5-dihydroxyphenyl) methanone (0.194 g, 0.523 mmol), thiosemicarbazide (0.100 g, 1.10 mmol), and p-toluenesulfonic acid monohydrate (0.049 g, 0.026 mmol) were dissolved in anhydrous methanol (1.5 mL). The reaction was carried out at 90 oC for 30 min under microwave irradiation. The solvent was removed under

reduced pressure and the product was extracted from water (5 mL) with ethyl acetate (3 x

120 5 mL). The combined organic phases were dried over sodium sulfate and concentrated.

Purification using flash chromatography (silica gel, hexanes:ethyl acetate, gradient 80:20

to 40:60) afforded [(3,5-dibromophenyl)-(3,5-dihydroxyphenyl) ketone]

thiosemicarbazone (0.163 g, 0.366 mmol) in a 70% yield. 1H NMR (600 MHz, DMSO-

d6): δ 9.89 (2H, s), 8.76 (1H, s), 8.70 (1H, s), 8.45 (1H, s), 7.90-7.85 (3H, m), 6.43 (1H, t,

13 J = 2.2 Hz), 6.09 (2H, d, J =2.2 Hz). C NMR (150 MHz, DMSO-d6): δ 177.74, 159.98,

145.99, 140.01, 134.17, 131.47, 128.88, 122.76, 105.44, 104.23. HPLC retention time

(Method B): 10.57 min for major isomer, 8.65 min for minor isomer. HRMS (ESI)

+ + calculated for C14H11Br2N3O2SH [M+H] 443.90115, found 443.90132.

(3-Bromophenyl)-(3-hydroxyphenyl) methanone (23).

[3-(t-Butyldimethylsilyl)oxyphenyl]-(3-bromophenyl) methanone (4.857 g, 12.42 mmol) was dissolved in tetrahydrofuran (40 mL) and solution of tetra-n-butyl ammonium fluoride trihydrate (6.598 g, 16.87 mmol) in THF (10 mL) was added dropwise. The reaction mixture was stirred at room temperature for 45 min. After reaction completion,

the reaction mixture was concentrated and extracted with ethyl acetate (3 x 100 mL) from

water (100 mL). The combined organic extracts were dried over Na2SO4 and

concentrated. Purification by flash column chromatography (silica gel, hexanes:EtOAc,

gradient 90:10 to 60:40) afforded (3-bromophenyl)-(3-hydroxyphenyl) methanone (4.07

1 g, 14.69 mmol, 87 % yield) as a light yellow solid. H-NMR (DMSO-d6, 500 MHz) δ

9.88 (1H, s), 7.87 (1H, ddd, J = 8.0 Hz, 2.1 Hz, 1.0 Hz), 7.83 (1H, t, J = 1.7 Hz, ArH),

7.69 (1H, ddd, J = 7.7 Hz, 1.5 Hz, 1.1 Hz), 7.52 (1H, t, J = 7.8 Hz), 7.37 (1H, t, J = 7.8

13 Hz), 7.12-7.15 (2H, m), 7.08 (1H, ddd, J = 8.1 Hz, 2.5 Hz, 1.1 Hz). C-NMR (DMSO-d6,

121 125 MHz) δ 194.24, 157.46, 139.44, 137.57, 135.10, 131.67, 130.73, 129.84, 128.50,

121.79, 120.68, 120.27, 115.96.

3-(3-Bromobenzoyl)phenyl dibenzyl phosphate (24).

(3-Bromophenyl)-(3-hydroxyphenyl) methanone (3.75 g, 13.65 mmol) was

dissolved in 50 mL of anhydrous acetonitrile. The flask was cooled to -40o C using a dry

ice/acetonitrile bath followed by the dropwise addition of carbon tetrachloride (6.5 mL,

68.25 mmol) and stirred for 5 min. 4-Dimethylaminopyridine (0.153 g, 1.36 mmol), N,N-

diisopropylethylamine (5.00 mL, 28.68 mmol), and dibenzyl phosphite (4.38 mL, 19.79

mmol) were added dropwise. The solution was allowed to slowly come to room

temperature and stirred for 1 h 20 min. After reaction completion, water (100 mL) was

added to the reaction mixture and products were extracted into EtOAc (3 x 100 mL). The

combined organic phases were dried over anhydrous Na2SO4 and concentrated under

reduced pressure. Purification by flash column chromatography (silica gel,

hexanes:EtOAc, gradient 90:10 to 30:70) afforded 3-(3-bromobenzoyl)phenyl dibenzyl

1 phosphate (6.698 g, 12.47 mmol, 91 % yield) as a yellow oil. H-NMR (DMSO-d6, 500

MHz) δ 7.91 (1H, ddd, J = 8.0 Hz, J = 2.1 Hz, J = 1.0 Hz), 7.85 (1H, t, J = 1.8 Hz), 7.67

(1H, ddd, J = 7.7 Hz, 1.6 Hz, 1.0 Hz), 7.61-7.56 (2H,m), 7.53-7.47 (3H, m), 7.36-7.33

13 (10H, m), 5.19 (4H, d, J = 8.8 Hz). C NMR (DMSO-d6, 125 Hz) δ 193.15, 150.10 (d, J

= 6.4 Hz), 138.68, 137.93, 135.50, 135.46 (d, J = 6.4 Hz), 131.78, 130.79, 130.45,

128.60, 128.54, 128.46, 128.01, 126.58, 124.60 (d, J = 4.8 Hz), 121.93, 120.65 (d, J = 4.6

Hz), 69.63, 69.58, 31P-NMR (DMSO, decoupled, 202 MHz) δ -6.39. 31P-NMR (DMSO,

85 % phosphoric acid as external standard, decoupled, 243 MHz) δ -5.35.

122 3-(3-Bromobenzoyl)phenyl dihydrogen phosphate (25).

The phosphate ester (6.58 g, 12.25 mmol) was dissolved 33% HBr in AcOH (25

mL) and the reaction mixture was stirred under air. After 1 h, water (75 mL) was added

to the flask (30 mL) and the resulting mixture was washed with hexanes (5 x 50 mL) at

which point the product precipitated out of solution. The aqueous layer was cooled to 0

oC and the solid was filtered and rinsed with ice cold water (15 mL). The solid was

allowed to dry overnight in the vacuum filter flask to yield 3-(3-bromobenzoyl)phenyl

dihydrogen phosphate (3.839 g, 10.75 mmol, 88%) as a white solid. 1H NMR (600 MHz,

DMSO) δ 11.98 (2H, brs), 7.90 (1H, ddd, J = 8.0 Hz, 2.1 Hz, 1.0 Hz), 7.88-7.87 (1H, m),

7.72 (1H, ddd, J = 7.7 Hz, 1.6 Hz, 1.0 Hz), 7.58-7.56 (2H, m), 7.54 (1H, t, J = 7.84),

7.52-7.50 (1H, m), 7.49-7.46 (1H, m). 13C NMR (150 MHz, DMSO) δ 193.59, 151.63 (d,

J = 6.2 Hz), 138.96, 137.60, 135.46, 131.83, 130.85, 130.10, 128.69, 125.58, 125.00 (d, J

= 5.2 Hz), 121.98, 120.85 (d, J = 4.5 Hz). 31P-NMR (DMSO, 85 % phosphoric acid as

external standard, decoupled, 243 MHz) δ -5.19.

[(3-bromophenyl)-(3-phosphophenyl)ketone] thiosemicarbazone (26).

A solution of 3-(3-Bromobenzoyl)phenyl dihydrogen phosphate (1.00 g, 2.80

mmol) and thiosemicarbazide (0.510 g, 5.60 mmol) in THF (15 mL) was refluxed for 6 h.

After reaction completion (determined from 1H NMR), the reaction mixture was cooled

to rt then to 0 0C and the THF layer was carefully transferred to another flask leaving the

solid behind. The filtrate was concentrated under a stream of N2 and further dried under

vacuum. The crude product was used without further purification.

123 [Disodium (3-bromophenyl)-(3-phosphophenyl)ketone] thiosemicarbazone (27).

Sodium carbonate (0.445 g, 4.20 mmol) was added to a suspension of crude 3-(3- bromobenzoyl)phenyl phosphate thiosemicarbazone in water (5 mL) and allowed to stir for 10 min. The reaction mixture was washed with EtOAc (3 x 10 mL) and the aqueous layer was concentrated to 2-3 mL using a stream of N2 gas. After purification by flash column chromatography (C-18,water:acetonitrile, 90:10), the eluent was concentrated under a stream of N2 gas to 15 mL followed by lyophilization to afford disodium (3-

bromophenyl)-(3-phosphophenyl)ketone] thiosemicarbazone (0.956 g, 2.02 mmol, 72%

1 yield over two steps) as a yellow solid. H NMR (600 MHz, D2O) δ 7.82 (0.6 H, t, J = 1.9

Hz), 7.73 (0.4 H, ddd, J = 8.1 Hz, 2.0 Hz, 1.0 Hz), 7.59-7.54 (2H, m), 7.51-7.47 (1H, m),

7.39 (0.6H, ddt, J = 8.3 Hz, 2.3 Hz, 1.1 Hz), 7.34 (0.4H, ddd, J = 7.7 Hz, 1.6 Hz, 1.0 Hz),

7.28-7.23 (1.4H, m), 7.07-7.06 (0.6 H, m), 7.00-6.98 (0.4 H, m), 6.97-6.95 (0.6H, m). 13C

NMR (150 MHz, D2O) δ 176.96, 176.76, 154.66 (d, J = 6.1 Hz, 153.94 (d, J = 6.1 Hz),

152.00, 151.83, 138.35, 137.04, 133.47, 133.16, 133.03, 131.99, 131.37, 131.14, 130.54,

130.42, 130.03, 129.21, 127.49, 126.82, 122.95, 122.86, 122.68 (d, J = 4.1 Hz), 122.39,

122.06 (d, J = 4.0 Hz), 122.00, 120.46 (d, J = 5.2 Hz), 118.74 (d, J = 4.8 Hz). 31P-NMR

(DMSO, 85 % phosphoric acid as external standard, decoupled, 243 MHz) δ 0.77, 0.65.

HPLC retention time (Method D): 12.57 min. HRMS (ESI) calculated for

+ C14H11BrN3O4PSNa2 [M+H] 473.92594, found 473.92602; HRMS (ESI) calculated for

+ C14H11BrN3O4PSNa2 [M+Na] 495.90789, found 495.90744.

AUTHOR INFORMATION

Corresponding Author

*Tel: 254-710-4117. Fax: 254-710-4272. E-mail: [email protected].

124 Funding

We express our appreciation to OXiGENE Inc. (grants to K.G.P. and M.L.T.) for their

financial support of this project and to the National Science Foundation for funding both

the Varian 500 MHz NMR spectrometer (Grant No. CHE- 0420802) and the Bruker X8

APEX diffractometer (Grant CHE-0321214).

Notes

The authors declare the following competing financial interest(s):The corresponding author (KGP) discloses a potential conflict of interest in regard to a paid consulting relationship with OXiGENE Inc.

Acknowledgements

We are grateful to Dr. Kevin K. Klausmeyer for his valuable assistance with the X-ray crystallographic study. We also thank Dr. Alejandro Ramirez (Mass Spectrometry Core

Facility, Baylor University) for mass spectroscopic analysis, Dr. Craig Moehnke for assistance with NMR studies, Dr. James Karban and Dr. Michelle Nemec (Director) for use of the shared Molecular Biosciences Center at Baylor University, and Amy Boylan for technical assistance.

125

CHAPTER FOUR

Design and Synthesis of Cathepsin Inhibitors: Symmetrical Thiosemicarbazone Analogues

Introduction

The propensity of the imine bond of the thiosemicarbazone moiety to isomerize is well known.166,200,202 Isomerization is potentially problematic since the E and Z geometrical isomers will likely exhibit different biological activity. To circumvent the possibility of generating different geometrical isomers arising from isomerization of the imine bond, symmetrical ketones were synthesized and utilized as base scaffolds for installation of the thiosemicarbazone moiety.

Symmetrical Benzophenone Thiosemicarbazones

Figure 4.1. Previously synthesized benzophenone thiosemicarbazone analogues

126 Previous structural activity relationship (SAR) studies based on the benzophenone

scaffold included brominated, hydroxylated, and fluorinated analogues.127,128 It was

discovered that incorporation of a 3-bromo substituent provided analogues with potent

inhibitory activity against cathepsin L. Interestingly, 3-bromobenzophenone

thiosemicarbazone (I) did not exhibit inhibitory activity against cathepsin L (Figure

4.1).205 Bis(3-bromophenyl)methanone thiosemicarbazone (III), a potent cathepsin L

inhibitor previously prepared in our laboratory, was resynthesized in preparation for

future derivatization of the precursor ketone to incorporate other electrophilic

warheads.205 (3-Bromopheneyl)(3-hydroxylphenyl) (V)128 thiosemicarbazone emerged as a lead inhibitor owing to the presence of a hydroxyl substituent which potentially could be utilized as a molecular handle for optimization of pharmacodynamics properties.

Incorporation of hydroxyl substituents onto a symmetrical benzophenone scaffold led to the design of analogue 9. Previous SAR studies revealed that incorporation of fluoro substituents in the ortho position compared to the meta or para positions resulted in analogues with greater inhibitory activity against cathepsin L demonstrated by the subsets of fluorinated analogues VII, VIII, IX and X, XI, XII.127 Extending this observed trend

to symmetrical analogues led to the design of analogue 10. Previously both the meta and para symmetrical bromo benzophenone thiosemicarbazone analogues were prepared and evaluated against cathepsin L. Incorporation of bromine substituents in the ortho

position led to analogues with reduced activity in previous studies. Although, bis(2-

bromophenyl)methanone thiosemicarbazone (11) was not expected to be as active, the

ortho analogue was prepared in order to expand the benzophenone thiosemicarbazone

SAR study.

127 Additionally, unsubstituted symmetrical thiosemicarbazones based on the 1,3- diphenylacetone and di(naphthalen-2-yl)methanone molecular frameworks were

prepared.

Scheme 4.1. Synthesis of thiosemicarbazones with a symmetrical benzophenone scaffolds

Synthesis of benzophenone thiosemicarbazones 8-11 (Scheme 4.1) and 2- naphthalenylmethanone thiosemicarbazone (15, Scheme 4.3) proceeded using similar synthetic methodology. Halogen-metal exchange with the appropriate aryl bromide with n-butyllithium generated an intermediate organolithium which was reacted with the corresponding Weinreb amide to afford ketones 4, 5, 7, and 14. Ketones 4, 6, 7, 14 and commercially available ketones bis(2-bromophenyl)methanone and 1,3-diphenylpropan-

2-one were condensed with thiosemicarbazide under various reaction conditions to afford final thiosemicarbazone analogues 8-12, and 15 (Schemes 4.1-4.3).

128

Scheme 4.2. Synthesis of the 1,3-diphenylpropan-2-one thiosemicarbazone

O

OH

NH(CH3)(OCH3)•HCl o Et3N, CH2Cl2, 0 C-rt 49%

O OMe N CH 13 3 O Br n-BuLi Li

THF, -78 oC THF, N ,-78oC 2 14 33%

TsOH, THF S reflux NH2 H2N N 13% H

S NH2 NH N

15 Scheme 4.3. Synthesis of di(naphthalen-2-yl)methanone thiosemicarbazone

The meta-hydroxy analogue 9 with 10% inhibition at 10 μM did not significantly impact cathepsin L inhibitory activity. The ortho-fluoro analogue 10 inhibited cathepsin

L by 72% at 10 μM. The ortho-bromo 11 analogue, as expected, did not inhibit cathepsin

L significantly (14% inhibition at 10 μM). 1,3-diphenylpropan-2-one thiosemicarbazone

(12) inhibited cathepsin L (30% at 10 μM) by a greater extent than the unsubstituted 1,5- diphenyl-3-pentanone thiosemicarbazone 27 (Scheme 4). Di(naphthalen-2-yl)methanone

129 thiosemicarbazone, a more rigid structure that the corresponding benzophenone

thiosemicarbazone, displayed inhibitory activity against cathepsin L with an IC50 value of

1826 nM which was less than the corresponding unsubstituted benzoylbenzophenone analogue but more than the unsubstituted benzophenone, dibenzylideneacetone (23) , and

1,5-diphenyl-3-pentanone (33) analogues.

Table 4.1. Inhibitory activity of benzophenone symmetrical thiosemicarbazones and extended analogues 12 and 15. % inhibition at 10 μM Cmpd Structure Cat L

8 * (IC50 = 16.7)

9 10%

10 72%

11 14%

30% 12

75% 15

(IC50 = 1826±126 nM) *Inhibition for compound 8 was previously determined prior to synthesis of current batch described. 205

130 Dibenzylideneacetone and 1,5-diphenyl-3-pentanone thiosemicarbazones

Preceding this work, potent cathepsin L inhibitors emerged from a library of

benzoylbenzophenone thiosemicarbazones. To explore the scope of effective inhibitors

based on the thiosemicarbazone warhead with incorporation of phenyl substituents which

spatially extended in a similar range to the benzoylbenzophenone thiosemicarbazones, inhibitors based on the di-2-naphthalenylmethanone (Scheme 4.3) dibenzylideneacetone

(Scheme 4.4), and 1,5-diphenyl-3-pentanone (Scheme 4.4) scaffolds were investigated.

Scheme 4.4. Synthesis of dibenzylideneacetone and 1,5-diphenyl-3-pentanone thiosemicarbazone analogues

Aldol condensation of acetone with the requisite benzaldehyde under basic conditions afforded dibenzylideneacetones 16-21. Several products are possible during

131 Pd/C-catalyzed hydrogenolysis of dibenzylideneacetone derivatives due to competing reduction of the alkene to alkane and ketone to alcohol or methylene products. Reported

methods for chemoselective Pd/C-catalyzed hydrogenolysis of the olefin bond in α,β-

unsaturated ketones include the addition of a catalyst poison206 and the hydrogen

source207. Attempts to reduce the double bonds of the unsubstituted dibenzylideneacetone

derivative 16 to afford the corresponding 1,5-diphenyl-3-pentanone 22 resulted in

multiple products. Reaction conditions explored included the use diphenyl sulfide as a catalyst poison, Hantzch ester as a hydrogen source, and cyclohexadiene as a hydrogen source (Table 4.2).

Table 4.2. Reduction of dibenzylideneacetone

Entry wt% of 10% Solvent Reaction Poison Hydrogen Yield Pd/C Conditions Source

1 10% MeOH Room Ph2S H2 gas 37% temperature balloon pressure 2 10% EtOH reflux None Hantzcsh Ester 58%

3 20% EtOH 100 oC None Cyclohexadiene 87% M.W. irradiation

The condensation of dibenzylideneacetone analogues 16-21 generated the desired

thiosemicarbazone analogues 26-31. Final thiosemicarbazone analogs 32-35 were prepared by reduction of the corresponding dibenzylideneacetone derivative by catalytic hydrogenation using palladium on carbon and cyclohexadiene followed by condensation with thiosemicarbazide.

132 Table 4.3. Inhibitory activity of dibenzylideneacetone and 1,5-diphenyl-3-pentanone thiosemicarbazone analogues

S NH2 R NH R 1 N 1

R2 R4 R4 R2 R3 R3

% inhibition at 10 μM

Cmpd R1 R2 R3 R4 Cat L 26 H H H -C=C- 31% 27 H Br H -C=C- 23% 28 H Cl H -C=C- <1% 29 H OH H -C=C- 29% 30 F H H -C=C- 38%

31 H H F -C=C- 7% 32 H H H -C-C- 23% 33 H OH H -C-C- 9% 34 F H H -C-C- 28% 35 H H F -C-C- 3%

Initially, percent inhibition of cathepsin L was determined at a concentration of 10

μM and compounds exceeding 50% inhibition were then further evaluated to obtain the

IC50 value. Although, cathepsin L inhibitory activity of thiosemicarbazone analogues based on the dibenzylideneacetone and 1,5-diphenyl-3-pentanone core scaffolds evaluated to date has not exceeded 50% inhibition at 10 μM, insight regarding cathepsin

L activity in relationship to molecular architecture was obtained. The observation that dibenzylideneacetone thiosemicarbazone derivatives 26, 29-31 display greater inhibitory activity against cathepsin L compared to their 1,5-diphenyl-3-pentanone

133 thiosemicarbazones counterparts, analogues 32-35, indicates that the presence of the double bond is valuable for the effective design of cathepsin L inhibitors based on this general scaffold (Table 4.3). The unsubstituted analogue 26 with 31% percent inhibition at 10 μM possessed greater activity than analogues incorporating ortho or meta substituents. The p-fluoro dibenzylideneacetone analogue 30 exhibited the greatest activity against cathepsin L with 38% inhibition at 10 μM. The overall structure of

(1E,4E)-1,5-bis(4-fluorophenyl)penta-1,4-dien-3-one thiosemicarbazone (36) was incorporated into the design of a new analogue which incorporated a cyclohexane ring in the center.

Scheme 4.5. Synthesis of (E)-7-(4-fluorobenzylidene)-3-(4-fluorophenyl)-7a-hydroxy- 3a,4,5,6,7,7a-hexahydro-1H-indazole-1-carbothioamide

Aldol condensation of p-fluorobenzaldehyde with cyclohexanone under basic

conditions afforded 2,6-bis((E)-4-fluorobenzylidene)cyclohexan-1-one (Scheme 4.5).

Attempted synthesis of the requisite thiosemicarbazone through condensation with

thiosemicarbazide using p-toluene sulfonic acid monohydrate as catalyst under

microwave irradiate, sonication, and reflux conditions generated zero to little product

formation as mainly starting material remained (observed by TLC). Replacing p-

134 toluenesulfonic acid monohydrate with titanium (IV) isopropoxide as catalyst resulted in

multiple products. The dominant product was isolated through purification using flash

chromatography and recrystallized from dichloromethane/pentane. X-ray crystallographic

analysis revealed the structure as (E)-7-(4-fluorobenzylidene)-3-(4-fluorophenyl)-7a-

hydroxy-3a,4,5,6,7,7a-hexahydro-1H-indazole-1-carbothioamide (37) (Figure 4.2).

Figure 4.2 X-ray crystal structure of compound 37

Although analogue 37 was not the desired product, the new analogue contained a thiocarbonyl bond, an electrophilic moiety capable of forming a covalent bond with cysteine proteases, within its molecular framework. Interestingly, biochemical evaluation of the new analogue inhibited cathepsin L activity by 70% at 10 μM.

135 General Synthetic Methods: Synthesis of Symmetrical Thiosemicarbazone Analogues

All reactions were performed under inert atmosphere using nitrogen gas unless otherwise specified. Chemicals and reagents used in the synthetic procedures were purchased from commercial suppliers and used without further purification. Anhydrous tetrahydrofuran and dichloromethane were purchased from commercial suppliers or dried using a VAC (Vacuum Atmospheres Co.) solvent purification system. Reactions were monitored by normal phase thin layer chromatography (TLC) plates SiliaPlateTM (silica

gel, 250 µM, F-254, 60 Å). Purification was performed using manual flash

chromatography and automated flash chromatography which was carried out with silica

gel purchased from either Silicycle Inc (230–400 mesh) or Biotage (40–65 microns).

Intermediates and products synthesized were characterized using a Varian Inova 500

MHz NMR system and/or Bruker Avance III HD 600 MHz NMR system. Deuterated

CDCl3 (with 0.03 % TMS as internal standard), acetone-d6, and DMSO-d6 were used as

solvents for recording the NMR. All the chemical shifts are expressed in ppm (δ),

coupling constants (J, Hz) and peak patterns are reported as broad (br), singlet (s),

doublet (d), triplet (t), quartet (q) and multiplet (m). High resolution mass spectra

(HRMS) were obtained in the Baylor University Mass Spectrometry Core Facility on a

Thermo Scientific LTQ Orbitrap Discovery using electrospray ionization ESI. Purity of

the final compounds was analyzed using an Agilent Technologies 1200 series HPLC

system with a Diode Array and Multiple Wavelength Detector SL, equipped with an

Agilent Eclipse XDB-C18 column (4.6 mm ID x 250 mm, 5 μm particle size, 80 Å pore

size). HPLC method parameters: T = 25 oC; 1.0 mL/min; injection volume 20 μL;

monitored at 254 nm, 300 nm, 320 nm. Four different HPLC gradients were used for

136 purity analysis; Method A: water/acetonitrile, gradient 50:50 to 90:10 from 0 to 25 min

and isocratic 90:10 from 25 to 30 min; Method B: acetonitrile/water, gradient 50:50 to

100:00 from 0 to 25 min, isocratic 100:0 from 25 to 30 min, gradient 100:00 to 50:50

from 30-32 min, and isocratic 50:50 from 32 to 40 min; Method C: acetonitrile/water,

gradient 30:70 to 100:00 from 0 to 25 min, isocratic 100:0 from 25 to 30 min, gradient

100:00 to 30:70 from 30-32 min, and isocratic 30:70 from 32 to 40 min.

3-Bromo-N-methoxy-N-methylbenzamide (1)

Triethylamine (1919 mL, 13.66 mmol) was added dropwise to a solution of N,O dimethylhydroxylamine hydrochloride (1.000 g, 10.25 mmol) in anhydrous dichloromethane (20 mL) at 0oC. After 10 min of stirring, a solution of 3-bromobenzoyl

chloride (902 mL, 6.83 mmol) dichloromethane (5 mL) was added dropwise and the

reaction mixture was returned to room temperature. After 4.5 h of stirring, the reaction mixture was quenched with 35 mL of water. The products were extracted with dichloromethane (2 x 25 mL) and the combined organic phases were dried over anhydrous sodium sulfate and concentrated under reduced pressure. Purification by flash column chromatography (silica gel, hexanes: ethyl acetate, gradient 90:10 to 70:30) afforded 3-bromo-N-methoxy-N-methylbenzamide (1.549 g, 6.83 mmol, 93% yield) as a

1 light yellow oil. H NMR (500 MHz, CDCl3) δ 7.83 (1H, t, J = 1.8 Hz, 1H), 7.62 (1H, dt,

J = 7.7 Hz, 1.3 Hz), 7.59 (1H, ddd, J = 8.0 Hz, 2.0 Hz, 1.0 Hz), 7.28 (1H, t, J = 7.9 Hz),

13 3.55 (3H, s), 3.36 (3H, s). C NMR (125 MHz, CDCl3) δ 168.19, 135.93, 133.56,

131.22, 129.61, 126.79, 122.01, 61.20, 33.57.

137 3-Methoxy-N-methoxy-N-methylbenzamide (2)

Triethylamine (2.47 mL, 17.6 mmol) was added dropwise to a solution of N,O-

dimethylhydroxylamine hydrochloride (1.286 g, 13.18 mmol) in dichloromethane (25

mL) at 0 oC. A solution of 3-methoxybenzoyl chloride (1.24 mL, 8.82 mmol) in

dichloromethane (10 mL) was added dropwise to the reaction mixture and the ice bath

was removed. After 4 h, the reaction was quenched with water (50 mL) and the product

was extracted using ethyl acetate (2 X 25 mL). The combined organic phases were dried

over sodium sulfate and concentrated. Purification using flash chromatography (silica gel,

hexanes:ethyl acetate, gradient 80:20 to 60:40) afforded 3-methoxy-N-methoxy-N-

1 methyl-benzamide (1.524 g, 7.807 mmol) in a 88% yield. H NMR (600 MHz, CDCl3): δ

7.31 (1H, t, J = 7.9 Hz), 7.24 (1H, d, J = 7.6 Hz), 7.20 (1H, s), 6.99 (1H, dd, J = 8.4, 2.4

13 Hz), 3.83 (3H, s), 3.58 (3H, s), 3.35 (3H, s). C NMR (150 MHz, CDCl3): δ 169.72,

159.19, 135.39, 129.10, 120.35, 116.58, 113.31, 77.25, 77.04, 76.83, 61.10, 55.37, 33.92.

2-Fluoro-N-methoxy-N-methylbenzamide (3)

Triethylamine (2.66 mL, 18.9 mmol) was added dropwise to a solution of N,O- dimethylhydroxylamine hydrochloride (1.384 g, 14.19 mmol) in dichloromethane (25 mL) at 0 oC. A solution of 2-fluorobenzoyl chloride (1.11 mL, 9.46 mmol) in

dichloromethane (10 mL) was added dropwise to the reaction mixture and the ice bath

was removed. After 6 h, the reaction was quenched with water (100 mL) and the product

was extracted using dichloromethane. The combined organic phases were dried over

sodium sulfate and concentrated. Purification using flash chromatography (silica gel,

hexanes:ethyl acetate, gradient 95:05 to 40:60) afforded 2-fluoro-N-methoxy-N-methyl-

1 benzamide (1.551 g, 8.467 mmol) in a 90% yield. H NMR (500 MHz, CDCl3): δ 7.44-

138 7.38 (2H, m), 7.19 (1H, t, J = 7.5 Hz), 7.10 (1H, t, J = 8.9 Hz), 3.55 (3H, brs), 3.35 (3H,

13 brs). C NMR (125 MHz, CDCl3): δ 166.40, 158.66, (d, J = 249 Hz), 131.50, 128.90,

124.11, 123.52 (d, J = 17 Hz), 115.69 (d, J = 21 Hz), 61.21, 32.31. 19F NMR (470 MHz,

- CDCl3): δ 114.04 (1F, s).

Bis(3-bromophenyl) methanone (4)

n-Butyllithium in hexanes (2.5 M, 1.97 mL) was added dropwise to a solution of

1,2-dibromobenzene (1.20 mL, 9.95 mmol) in THF (15 mL) at -78 oC. After 30 min., a

solution of 3-bromo-N-methoxy-N-methylbenzamide (1.215 g, 4.977 mmol) in THF (15 mL) was added dropwise and the reaction mixture was allowed to stir for 1 h. The reaction mixture was quenched with 1 M HCl (24 mL) and extracted with chloroform (2

x 40 mL). The combined organic phases were dried over sodium sulfate and

concentrated. Purification using flash chromatography (silica gel, hexanes:ethyl acetate, gradient 99:01 to 90:10) afforded bis(3-bromophenyl) methanone (0.368 g, 3.40 mmol,

1 22%) as a white solid. H NMR (500 MHz, CDCl3): δ 7.93 (2H, t, J = 1.8 Hz), 7.74 (2H,

ddd, J = 8.0, 2.1, 1.1 Hz), 7.69 (2H, dt, J = 7.7, 1.4 Hz), 7.38 (2H, t, J = 7.8 Hz). 13C

NMR (125 MHz, CDCl3): δ 193.60, 138.78, 135.69, 132.71, 130.00, 128.49, 122.80.

Bis(3-methoxyphenyl) methanone (5)

n-Butyllithium in hexanes (2.5 M, 2.93 mL) was added dropwise to a solution of

3-bromoanisole (1.03 mL, 8.14 mmol) in THF (15 mL) at -78 oC. After 30 min., a

solution of 3-methoxy-N-methoxy-N-methylbenzamide (1.444 g, 7.916 mmol) in THF

(10 mL) was added dropwise and the reaction mixture was allowed to stir for 1 h. The

reaction mixture was quenched with 1 M HCl (24 mL) and extracted with EtOAc (2 x 30

mL). The combined organic phases were dried over sodium sulfate and concentrated.

139 Purification using flash chromatography (silica gel, hexanes:ethyl acetate, gradient 98:02

to 80:20) afforded bis(3-methoxyphenyl) methanone (0.824 g, 3.40 mmol, 46%) as a

1 yellow oil. H NMR (600 MHz, CDCl3): δ 7.41 – 7.32 (6H , m), 7.13 (2H, ddd, J = 7.9,

13 2.7, 1.3 Hz), 3.86 (6H, s). C NMR (150 MHz, CDCl3): δ196.28, 159.54, 138.90,

129.18, 122.84, 118.89, 114.28, 55.48.

Bis(3-hydroxyphenyl) methanone (6)

Bis(3-dimethoxyphenyl) methanone ( 0.320 g, 1.32 mmol) was dissolved in

anhydrous dichloromethane (20 mL) and cooled to 0 oC in an ice bath. Boron tribromide

in dichloromethane (1 M, 2.65 mL) was added dropwise to the reaction mixture and the

ice bath was removed. After 7 h, the reaction mixture was poured over ice and 3 M

hydrochloric acid (5 mL) was added. The products were extracted with ethyl acetate (3 x

20 mL) and the combined organic phases were washed with sodium bicarbonate (20 mL), dried over anhydrous sodium sulfate, and concentrated. Purification using flash

chromatography (silica gel, hexanes: ethyl acetate, gradient 70:30 to 60:40) afforded

bis(3-hydroxyphenyl) methanone (0.228 g, 1.06 mmol) in an 80% yield. 1H NMR (600

MHz, acetone-d6): 8.71 (2H, s), 7.37 (2H, t, J = 7.8 Hz), 7.27 – 7.20 (4H, m), 7.11 (2H,

13 ddd, J = 8.1, 2.5, 1.1 Hz) C NMR (150 MHz, acetone-d6): δ 196.15, 158.17, 140.07,

130.31, 121.93, 120.23, 116.97.

Bis(2-fluorophenyl) methanone (7)

n-Butyllithium in hexanes (2.5 M, 3.128 mL) was added dropwise to a solution of

2-fluorobromobenzene (0.950 mL, 8.69 mmol) in THF (25 mL) at -78 oC. After 30 min.,

a solution of 2-fluoro-N-methoxy-N-methylbenzamide (1.450 g, 7.916 mmol) in THF (10

mL) was added dropwise and the reaction mixture was allowed to stir for 1 h. The

140 reaction mixture was quenched with 1 M HCl (30 mL) and extracted with EtOAc (2 x 30

mL). The combined organic phases were dried over sodium sulfate and concentrated.

Purification using flash chromatography (silica gel, hexanes:ethyl acetate, gradient 98:02

to 80:20) afforded bis(2-fluorophenyl) methanone (0.836 g, 3.83 mmol, 49%) as a

1 yellow oil. H NMR (600 MHz, CDCl3): δ 7.71 (2H, td, J = 7.5, 1.9 Hz), 7.59 – 7.51 (2H,

m), 7.27 (2H, td, J = 7.5, 1.0 Hz), 7.12 (2H, ddd, J = 10.6, 8.3, 1.1 Hz). 13C NMR (150

MHz, CDCl3) δ 189.74 , 161.05 (dd, J = 255.0, 1.6 Hz), 134.22 (d, J = 9.5 Hz), 130.86 ,

127.55 (d, J = 12.2 Hz), 124.39 – 124.24 (m), 116.22 (d, J = 21.8 Hz). 19F NMR (565

MHz, CDCl3) δ -112.16 – -112.22 (2F, m).

[Bis(3-bromophenyl) ketone] thiosemicarbazone (8)

Bis(3-bromophenyl) methanone ( 0.100 g, 0.294 mmol), thiosemicarbazide (0.053

g, 0.59 mmol), and p-toluene sulfonic acid monohydrate (0.006 g, 0.03 mmol) were

dissolved in anhydrous tetrahydrofuran (10 mL) and the reaction was refluxed overnight.

The reaction mixture was concentrated and the product was extracted with ethyl acetate from water. The combined organic phases were dried over sodium sulfate and concentrated. Purification using flash chromatography (silica gel, hexanes:ethyl acetate, gradient 93:07 to 40:60) afforded [bis(3-bromophenyl) ketone] thiosemicarbazone (0.050

1 g, 0.121 mmol) in a 41% yield. H NMR (500 MHz, DMSO-d6): δ 8.94 (1H, s), 8.64

(1H, s), 8.52 (1H, s), 8.08 (1H, s), 7.80 (1H, ddd, J = 8.2, 2.1, 1.0 Hz), 7.63 – 7.56 (3H ,

13 m), 7.37 – 7.33 (2H, m), 7.30 (1H , t, J = 7.8 Hz). C NMR (125 MHz, DMSO-d6): δ

178.32, 145.62, 138.60, 133.41, 132.98, 132.35, 131.89, 131.13, 130.44, 129.28, 127.69,

126.96, 122.98, 122.21. HPLC (Method A) 10.37 min.

141 [Bis(3-hydroxyphenyl) ketone] thiosemicarbazone (9)

Bis(3-hydroxyphenyl) methanone (0.054 g, 0.25 mmol), thiosemicarbazide (0.046 g, 0.50 mmol), and p -toluene sulfonic acid monohydrate (0.0034 g, 0.018 mmol ) were dissolved in ethanol (2 mL) and the reaction was carried out at 100 oC for 1 h under microwave irradiation. The reaction mixture was concentrated and purification using flash chromatography (silica gel, hexanes:ethyl acetate, gradient 80:20 to 40:60) afforded

[bis(3-hydroxyphenyl) ketone] thiosemicarbazone (0.0549 g, 0.191 mmol) in a 76%

1 yield. H NMR (500 MHz, DMSO-d6): δ 9.95 (1H, s), 9.45 (1H, s), 8.61 (1H, s), 8.35

(1H, s), 8.26 (1H, s), 7.46 (1H, t, J = 7.8 Hz), 7.26 (1H, d, J = 7.8 Hz), 7.17 (1H, t, J =

7.9 Hz), 6.98 (1H, ddd, J = 8.3 Hz, 2.5 Hz, 1.0 Hz), 6.92 (1H, t, J = 2.0 Hz), 6.79 (1H, ddd, J = 7.5 Hz, 1.3 Hz), 6.70 (1H, dt, J = 8.0 Hz, 2.5 Hz, 1.0 Hz), 6.63-6.61 (1H, m). 13C

NMR (125 MHz, DMSO-d6): δ 177.62, 158.37, 157.22, 149.28, 137.45, 132.52, 131.16,

129.29, 118.39, 118.28, 116.92, 116.75, 114.82, 114.54. HPLC (Method B) 2.69 min.

+ + HRMS (ESI) calculated for C14H13N3O2SH [M+H] 288.08012, found 288.08020.

[Bis(2-fluorophenyl) ketone] thiosemicarbazone (10)

Bis(2-fluorophenyl) methanone ( 0.273 g, 1.25 mmol), thiosemicarbazide (0.455

g, 5.00 mmol), and p-toluene sulfonic acid monohydrate (0.009 g, 0.028 mmol) were

dissolved in anhydrous ethanol (3 mL) and the reaction was carried out at 100 oC for 2 h

under microwave irradiation. The reaction mixture was concentrated and the product was

extracted with ethyl acetate from water. The combined organic phases were dried over

sodium sulfate and concentrated. Purification using flash chromatography (silica gel,

hexanes:ethyl acetate, gradient 93:07 to 40:60) afforded [bis(2-fluorophenyl) ketone]

thiosemicarbazone (0.0862 g, 0.296 mmol) in a 23% yield. 1H NMR (500 MHz, acetone-

142 d6): δ 8.90 (1H, s), 7.99 (1H, s), 7.80 (1H, s), 7.77 (1H, td, J = 7.9 Hz, 1.8 Hz), 7.67-7.61

(1H m), 7.49 (1H, dddd, J = 8.3 Hz, 7.4 Hz, 5.0 Hz, 1.8 Hz), 7.46-7.41 (2H, m), 7.41-

7.36 (1H, m), 7.26 (1H, td, J = 7.6 Hz, 1.1 Hz), 7.14 (1H, ddd, J = 11.6 Hz, 8.3 Hz, 1.1

13 Hz). C NMR (125 MHz, acetone-d6): δ 180.90, 161.74 (d, J = 252 Hz), 160.04 (d, J =

248 Hz), 140.82 (d, J = 1.8 Hz), 133.48 (d, J = 8.2 Hz), 132.57 (d, J = 8.6 Hz), 131.28 (d,

J = 2.1 Hz), 130.91 (dd, J = 3.4 Hz, 1.4 Hz), 126.43 (d, J = 3.4 Hz), 125.86 (d, J = 9.5

Hz), 125.29 (d, J = 3.7 Hz), 121.29 (dd, J = 17 Hz, 1.5 Hz), 117.45 (d, J = 21 Hz), 117.18

19 - - - - (d, J = 22 Hz). F NMR (470 MHz, acetone-d6): δ 113.92- 113.96 (1F, m), 114.86-

114.91 (1F, m). HPLC (Method B) 4.64 min. HRMS (ESI) calculated for

+ + C14H11F2N3SH [M+H] 314.05340, found 314.05349.

[Bis(2-bromophenyl) ketone] thiosemicarbazone (11)

Bis(2-bromophenyl) methanone (0.500 g, 1.47 mmol), thiosemicarbazide (0.535

g, 5.88 mmol), and p -toluene sulfonic acid monohydrate (0.0139 g, 0.0735 mmol ) were

dissolved in anhydrous dimethyl sulfoxide (1.5 mL) and the reaction was carried out at

100 oC for 2 h under microwave irradiation. The reaction mixture was diluted with water

(25 mL) and the product was extracted with ethyl acetate (2 x1 5 mL). The combined organic phases were dried over sodium sulfate and concentrated. Purification using flash

chromatography (silica gel, hexanes:ethyl acetate, gradient 93:07 to 40:60) afforded

[bis(2-bromophenyl) ketone] thiosemicarbazone (0.0074 g, 0.018 mmol) in a 1.2% yield.

1 H NMR (500 MHz, acetone-d6) δ 8.87 (1H, s), 7.90 (1H, s), 7.84 (1H, dd, J = 8.1, 0.7

Hz), 7.80 (1H, s), 7.73 (1H, dd, J = 8.0, 0.8 Hz), 7.62 (1H, td, J = 7.5, 1.0 Hz), 7.64-7.60

13 (1H, m), 7.48 – 7.40 (3H, m), 7.36 – 7.32 (1H, m). C NMR (125 MHz, acetone-d6): δ

143 181.02, 146.28, 137.70, 135.36, 134.89, 134.32, 133.11, 132.87, 132.14, 131.52, 129.62,

128.42, 123.10, 122.59. HPLC (Method B) 6.96 min.

1,3-diphenylacetone thiosemicarbazone (12)

1,3-diphenylacetone (500 mg, 2.38 mmol) was dissolved in anhydrous THF (10

mL) followed by the addition of p-toluenesulfonic acid (9.13 mg, 0.048 mmol). After 10

min. of refluxing, thiosemicarbarbazide (433 mg, 4.76 mmol) was added and the reaction

mixture was refluxed for 1 day. The reaction mixture was concentrated and products were extracted using dichloromethane from water. The combined organic phases were washed with brine and dried over anhydrous sodium sulfate, and concentrated under reduced

pressure. The crude product was purified by flash chromatography (silica gel,

dichloromethane:EtOAc, gradient 100:0 to 98:2) to afford 1,3-diphenylacetone

thiosemicarbazone (0.577 g, 2.04 mmol, 85% yield) as a white solid. 1H NMR (500

MHz, acetone-d6): δ 9.36 (1H, s), 7.66 (1H, s), 7.42 (1H, s), 7.34-7.30 (4H, m), 7.27-7.23

13 (4H, m), 7.17-7.15 (2H, m), 3.82 (2H, s), 3.60 (2H, s). C NMR (125 MHz, acetone-d6):

δ 181.22, 152.66, 137.91, 136.23, 130.11, 129.71, 129.65, 129.35, 127.69, 127.54, 43.86,

+ + 35.32. HPLC (Method A) 6.11 min. HRMS (ESI) calculated for C16H17N3SH [M+H]

284.12159, found 284.12288.

N-Methoxy-N-methyl-2-naphthamide (13)

Triethylamine (2.95 mL, 21.0 mmol) was added dropwise to a solution of N,O- dimethylhydroxylamine hydrochloride (1.535 g, 15.73 mmol) in dichloromethane (30 mL) at 0 oC. A solution of 2-naphtoyl chloride (2.00 g, 10.5 mmol) in dichloromethane

(10 mL) was added dropwise to the reaction mixture and the ice bath was removed and

the reaction mixture was stirred overnight. The reaction was quenched with water and the

144 product was extracted with dichloromethane. The combined organic phases were dried

over sodium sulfate and concentrated. Purification using flash chromatography (silica gel,

hexanes:ethyl acetate, gradient 95:05 to 60:40) afforded N-methoxy-N-methyl-2-

1 naphthamide (1.096 g, 5.092 mmol) in a 49% yield. H NMR (500 MHz, CDCl3): δ 8.23

(1H, s), 7.91-7.89 (1H, m), 7.87-7.85 (2H, m), 7.76 (1H, dd, J = 8.5 Hz, 1.7 Hz), 7.58-

13 7.50 (2H, m), 3.56 (3H, s), 3.42 (3H, s). C NMR (125 MHz, CDCl3): δ 169.90, 134.22,

132.49, 131.40, 128.83, 128.68, 127.69, 127.61, 127.37, 126.46, 125.06, 61.12, 33.87.

Bis-(naphthalen-2-yl) methanone (14)

n-Butyllithium in hexanes (2.5 M, 1.84 mL) was added dropwise to a solution of

3-naphthyl bromide (1.080 g, 5.110 mmol) in THF (20 mL) at -78 oC. After 1 h, a solution of N-methoxy-N-methyl-2-naphthamide (1.006 g, 4.645 mmol) in THF (10 mL) was added dropwise and the reaction mixture was allowed to stir for 2 h. The reaction mixture was quenched with 1 M HCl (40 mL) and extracted with CH2Cl2 (2 x 40 mL).

The combined organic phases were washed with saturated sodium bicarbonate and dried

over sodium sulfate and concentrated. Purification using flash chromatography (silica gel,

hexanes:ethyl acetate, gradient 99:01 to 90:10) afforded bis-(naphthalen-2-yl) methanone

1 (0.427 g, 1.51 mmol, 33%) as a white solid. H NMR (600 MHz, CDCl3): δ 8.33 (2H, s),

8.00 (2H, dd, J = 8.5, 1.6 Hz), 7.98 (2H, d, J = 8.8 Hz), 7.97 – 7.90 (4H, m), 7.63 (2H,

ddd, J = 8.2, 6.8, 1.3 Hz), 7.57 (2H, ddd, J = 8.1, 6.8, 1.3 Hz). 13C NMR (150 MHz,

CDCl3): δ 196.80, 135.26, 135.16, 132.29, 131.81, 129.43, 128.35, 128.31, 127.85,

126.83, 125.88.

145 Bis-(naphthalen-2-yl) ketone thiosemicarbazone (15)

Bis-(naphthalen-2-yl) methanone (150 mg, 0.531 mmol) was dissolved in

anhydrous THF (10 mL) followed by the addition of p-toluenesulfonic acid (2.00 mg,

0.011 mmol). After 10 min. of refluxing, thiosemicarbarbazide (0.967 g, 1.06 mmol) was

added and the reaction mixture was stirred for 1 day. The reaction mixture was

concentrated and products were extracted with dichloromethane from water. The

combined organic phases were washed with brine and dried over anhydrous sodium

sulfate, and concentrated under reduced pressure. The crude product was purified by

flash chromatography (silica gel, hexane:EtOAc, gradient 93:7 to 40:60) to afford bis-

(naphthalen-2-yl) ketone thiosemicarbazone (0.025 g, 0.070 mmol, 13% yield) as a

1 yellow solid. H NMR (500 MHz, acetone-d6) δ 8.76 (1H, brs, NH), 8.28 (1H, dd, J=

8.8 and 2.0 Hz, ArH), 8.24 (1H, d, J= 8.3 Hz, ArH), 8.21 (1H, brs, NH), 8.12-8.07 (3H, m, ArH), 7.82-7.66 (5H, m, ArH, H), 7.55-7.51 (2H, m, ArH), 7.49-7.45 (1H, m, ArH);

13 C NMR (125 MHz, acetone-d6) δ 180.53, 150.32, 135.54, 134.99, 134.74, 134.47,

133.99, 130.80, 130.03, 129.92, 129.50, 129.49, 129.43, 128.99, 128.93, 128.48, 128.45,

128.05, 128.00, 127.40, 126.39, 124.85. HPLC retention time (Method A) 12.73 min.

+ + HRMS (ESI) calculated for C22H17N3SH [M+H] 356.12159, found 356.12176.

(1E,4E)-1,5-Diphenylpenta-1,4-dien-3-one (16)

Benzaldehyde (3.23 mL, 32.0 mmol) dissolved in acetone (1.17 mL, 16.0 mmol) was added dropwise to a solution of NaOH (6 g), ethanol (30 mL), and water (45 mL) at

0 oC. After stirring at room temperature for 5 hrs, the reaction mixture was filtered and

washed with water (3 x 30 mL). The crude product was recrystallized from ethanol to

afford (1E,4E)-1,5-diphenylpenta-1,4-dien-3-one (2.680 g, 11.44 mmol, 71% yield) as a

146 1 yellow solid. H NMR (500 MHz, CDCl3): δ 7.75 (2H, d, J = 16 Hz), 7.65-7.61 (2H, m),

13 7.45-7.40 (6H, m), 7.09 (2H, d, J = 16 Hz). C NMR (125 MHz, CDCl3): δ 188.91,

143.31, 134.80, 130.49, 128.96, 128.39, 125.43.

(1E,4E)-1,5-Bis(3-bromophenyl)penta-1,4-dien-3-one (17)

3-bromobenzaldehyde (1.87 mL, 16.0 mmol) dissolved in acetone (0.587 mL, 8.0 mmol) was added dropwise to a solution of NaOH (4 g), ethanol (20 mL), and water (30 mL) at 0 oC. After stirring at room temperature at 2 h, the reaction mixture was filtered

and washed with water (2 x 25 mL). The crude product was recrystallized from ethyl

acetate/methanol to afford (1E,4E)-1,5-bis(3-bromophenyl)lpenta-1,4-dien-3-one (1.846

1 g, 4.71 mmol, 59% yield) as a yellow solid. H NMR (500 MHz, CDCl3): δ 7.76 (2H, t, J

= 1.8 Hz ), 7.65 (2H, d, J = 16 Hz ), 7.54-7.50 (4H, m), 7.29 (2H, t, J = 7.8 Hz), 7.05

13 (2H, d, J = 16 Hz). C NMR (125 MHz, CDCl3) δ 188.11, 141.88, 136.78, 133.34,

130.89, 130.49, 127.15, 126.38, 123.12.

(1E,4E)-1,5-Bis(3-chlorophenyl)penta-1,4-dien-3-one (18)

3-chlorobenzaldehyde (1.81 mL, 16.0 mmol) dissolved in acetone (0.587 mL, 8.0 mmol) was added dropwise to a solution of NaOH (4 g), ethanol (20 mL), and water (30 mL) at 0 oC. After stirring at room temperature at 2 h, the reaction mixture was filtered

and washed with water (2 x 20 mL). The crude product was recrystallized from ethanol

to afford (1E,4E)-1,5-bis(3-chlorophenyl)lpenta-1,4-dien-3-one (1.035 g, 3.414 mmol,

1 43% yield) as a yellow solid. H NMR (500 MHz, CDCl3) δ 7.67 (2H, d, J = 16 Hz), 7.61

(2H, t, J = 1.9 Hz), 7.48 (2H, dt, J = 7.3Hz, 1.7 Hz), 7.39 (2H, dt, J = 8.0 Hz, 1.7 Hz),

13 7.37-7.34 (2H, m), 7.06 (2H, d, J = 16 Hz). C NMR (125 MHz, CDCl3) δ 188.18,

141.98, 136.51, 135.03, 130.43, 130.23, 127.96, 126.70, 126.37.

147 (1E,4E)-1,5-Bis(3-hydroxyphenyl)penta-1,4-dien-3-one (19)

A solution of sodium hydroxide (4 g) in water (10 mL) was added dropwise to a

mixture of 3-hydroxybenzaldehyde (2.442 g, 20.0 mmol) and acetone (0.734 mL, 16.0

mmol) in ethanol (15 mL) at 0 oC. After stirring at room temperature at 1.5 h, the reaction

mixture was neutralized to pH 7 with 3 M HCl, filtered and washed with water.

Purification using flash chromatography (silica gel, hexanes:ethyl acetate, gradient 85:15

to 60:40) afforded (1E,4E)-1,5-bis(3-hydroxyphenyl)lpenta-1,4-dien-3-one (0.826 g, 3.10

1 mmol, 31% yield) as a yellow solid. H NMR (600 MHz, acetone-d6): δ 8.56 (2H, s), 7.71

(2H, d, J = 16.0 Hz), 7.28 (2H, t, J = 7.8 Hz), 7.25 – 7.19 (6H, m), 6.93 (2H, ddd, J = 8.0,

13 2.5, 1.1 Hz). C NMR (150 MHz, acetone-d6): δ 188.90, 158.70, 143.47, 137.46, 130.86,

126.52, 120.82, 118.37,115.58.

(1E,4E)-1,5-bis(4-fluorophenyl)penta-1,4-dien-3-one (20)

4-Fluorobenzaldehyde (1.72 mL, 16.0 mmol) dissolved in acetone (0.587 mL, 8.0

mmol) was added dropwise to a solution of NaOH (4 g), ethanol (20 mL), and water (30

mL) at 0 oC. After stirring at room temperature at 2 h, the reaction mixture was filtered

and washed with water (2 x 20 mL). The crude product was recrystallized from ethanol

to afford (1E,4E)-1,5-bis(4-fluorophenyl)lpenta-1,4-dien-3-one (1.520 g, 5.62 mmol,

1 70% yield) as a yellow solid. H NMR (600 MHz, CDCl3): δ 7.70 (2H, d, J = 15.9 Hz),

7.60 (4H, dd, J = 8.5, 5.4 Hz), 7.11 (4H, t, J = 8.5 Hz), 6.99 (2H, d, J = 15.9 Hz). 13C

NMR (150 MHz, CDCl3): δ 188.44, 164.06 (d, J = 252.0 Hz), 142.08, 130.99 (d, J = 3.3

Hz), 130.28 (d, J = 8.4 Hz), 125.09 (d, J = 2.4 Hz), 116.16 (d, J = 22.0 Hz. 19F NMR (565

MHz, CDCl3) δ -108.99 – -109.08 (2F, m).

148 (1E,4E)-1,5-bis(2-fluorophenyl)penta-1,4-dien-3-one (21)

2-Fluorobenzaldehyde (1.69 mL, 16.0 mmol) dissolved in acetone (0.587 mL, 8.0

mmol) was added dropwise to a solution of NaOH (4 g), ethanol (20 mL), and water (30

mL) at 0 oC. After stirring at room temperature at 1.5 h, the reaction mixture was diluted

with water (100 mL) and extracted with dichloromethane (2 x 50 mL). The combined

organic phases were washed with brine and dried over sodium sulfate. Purification using

flash chromatography (silica gel, hexanes:ethyl acetate, gradient 95:05 to 60:40) afforded

(1E,4E)-1,5-bis(2-fluorophenyl)penta-1,4-dien-3-one (0.903 g, 0.198 mmol, 42% yield)

1 as a yellow solid. H NMR (500 MHz, CDCl3): δ 7.86 (2H, d, J = 16.2 Hz), 7.63 (2H, td,

J = 7.6 Hz, 1.7 Hz), 7.38 (2H, dddd, J = 8.2 Hz, 7.2 Hz, 5.2 Hz, 1.7 Hz), 7.22 – 7.16 (4H,

13 m), 7.13 (2H, ddd, J = 10.7 Hz, 8.3 Hz, 1.2 Hz). C NMR (125 MHz, CDCl3): δ 189.08,

161.77 (d, J = 254.3 Hz), 136.22 (d, J = 2.8 Hz), 132.04 (d, J = 9.0 Hz), 129.50 (d, J =

3.1 Hz), 127.75 (d, J = 6.4 Hz), 124.66 (d, J = 3.7 Hz), 122.99 (d, J = 11.5 Hz), 116.40

19 (d, J = 22.0 Hz). -113.79 (ddd, J = 10.9, 7.4, 5.3 Hz) F NMR (470 MHz, CDCl3) -

113.79 (ddd, J = 10.9, 7.4, 5.3 Hz).

1,5-Diphenyl-3-pentanone (22)

Entry 1: Diphenyl sulfide (7.2 μL, 0.043 mmol) was added to a solution of 10%

palladium on carbon (0.100 g) and (1E,4E)-1,5-diphenylpenta-1,4-dien-3-one (1.00 g,

4.27 mmol) in methanol (20 mL). The reaction mixture was flushed with nitrogen gas, evacuated and carried out under hydrogen gas (balloon pressure). After 3 h, the reaction mixture was filtered through celite and concentrated. Purification using flash chromatography (silica gel, hexanes:ethyl acetate, gradient 99:01 to 85:15) afforded 1,5- diphenyl-3-pentanone (0.380 g, 1.59 mmol) in a 37% yield. 1H NMR (500 MHz,

149 acetone-d6): δ 7.27-7.23 (4H,m), 7.20-7.18 (4H, m), 7.17-7.14 (2H, m), 2.86-2.82 (4H,

13 m), 2.78-2.75 (4H, m). C NMR (125 MHz, acetone-d6): δ 208.74, 142.44, 129.17,

129.15, 126.68, 44.66, 30.33.

Entry 2: 10% Palladium on Carbon (0.47 mg) was added to a solution of (1E,4E)-1,5- diphenylpenta-1,4-dien-3-one (0.234 g, 1.00 mmol) and diethyl 1,4-dihydro-2,6- dimethyl-3,5-pyridinedicarboxylate (0.532g, 2.10 mmol) in anhydrous ethanol (50 mL).

The reaction mixture was refluxed for 1 h followed by filtration through celite.

Purification using flash chromatography (silica gel, hexanes:ethyl acetate, gradient 99:01 to 70:30) afforded 1,5-diphenyl-3-pentanone (0.138 g, 0.579 mmol) in a 58% yield.

Entry 3: 10% Palladium on Carbon (0.27 mg) was added to a solution of (1E,4E)-1,5- diphenylpenta-1,4-dien-3-one (0.117 g, 0.500 mmol) and cyclohexadiene (472 µL, 5.00 mmol) in anhydrous ethanol (4 mL). The reaction mixture was carried out at 100 oC for

20 min under microwave irradiation. The reaction mixture was filtered through celite and

solvent was removed under reduced pressure. Purification using flash chromatography

(silica gel, hexanes:ethyl acetate, gradient 99:01 to 70:30) afforded 1,5-diphenyl-3- pentanone (0.104 g, 0.436 mmol) in a 87% yield.

1,5-Bis(3-hydroxyphenyl)-3-pentanone (23)

1E,4E)-1,5-bis(3-hydroxyphenyl)lpenta-1,4-dien-3-one (0.399 g, 1.50 mmol), 1,4- cyclohexadiene (1.42 mL, 15.0 mmol), and 10% palladium on carbon (79 mg, 0.075 mmol) were dissolved in ethanol (18 mL) and the reaction was carried out at 100 oC for

20 min under microwave irradiation. The reaction mixture was filtered through celite and

solvent was removed under reduced pressure. Purification using flash chromatography

(silica gel, hexanes:ethyl acetate, gradient 90:10 to 50:50) afforded 1,5-Bis-3-

150 hydroxphenyl-3-pentanone (0.238 g, 0.880 mmol, 59% yield). 1H NMR (500 MHz,

acetone-d6): δ 8.13 (2H, s), 7.07 (2H, t, J = 7.8 Hz), 6.69-6.63 (8H, m), 2.79-2.71 (8H,

13 m). C NMR (125 MHz, acetone-d6): δ 207.89, 157.28, 142.97, 129.16, 119.24, 119.24,

115.10, 112.70, 43.61, 29.32.

1,5-Bis-4-fluorophenyl-3-pentanone (24)

(1E,4E)-1,5-bis(4-fluorophenyl)lpenta-1,4-dien-3-one (0.271 g, 1.00 mmol), 1,4-

cyclohexadiene (0.94 mL, 10 mmol), and 10% palladium on carbon (54 mg, 0.050

mmol) were dissolved in ethanol (12 mL) and the reaction was carried out at 100 oC for

20 min under microwave irradiation. The reaction mixture was filtered through silica and

solvent was removed under reduced pressure. Purification using flash chromatography

(silica gel, hexanes:ethyl acetate, gradient 97:03 to 70:30) afforded 1,5-Bis-4-

fluorophenyl-3-pentanone (0.156 g, 0.569 mmol) in a 57% yield. 1H NMR (500 MHz,

CDCl3): δ 7.10 (4H, dd, J = 8.3 Hz, 5.6 Hz), 6.94 (4H, t, J = 8.7 Hz), 2.85 (4H, t, J = 7.5

13 Hz), 2.67 (4H, t, J = 7.5 Hz). C NMR (125 MHz, CDCl3): δ 208.78 , 161.48 (d, J =

244.1 Hz), 136.68 (d, J = 3.3 Hz), 129.83 (d, J = 7.7 Hz), 115.34 (d, J = 21.0 Hz), 44.66

19 (d, J = 0.9 Hz), 28.94. F NMR (470 MHz, CDCl3): δ -117.23 (2F, tt, J = 8.9, 5.4 Hz).

1,5-Bis-2-fluorophenyl-3-pentanone (25)

(1E,4E)-1,5-bis(2-fluorophenyl)lpenta-1,4-dien-3-one (0.410 g, 1.52 mmol), 1,4-

cyclohexadiene (1.42 mL, 10 mmol), and 10% palladium on carbon (78 mg, 0.074

mmol) were dissolved in ethanol (12 mL) and the reaction was carried out at 100 oC for

20 min under microwave irradiation. The reaction mixture was filtered through silica and

solvent was removed under reduced pressure. Purification using flash chromatography

(silica gel, hexanes:ethyl acetate, gradient 95:05 to 80:20) afforded 1,5-Bis-2-

151 fluorophenyl-3-pentanone (0.336 g, 1.22 mmol, 80% yield). 1H NMR (500 MHz,

CDCl3): δ 7.19 – 7.15 (4H, m), 7.05 – 6.97 (4H, m), 2.91 (4H, t, J = 7.6 Hz), 2.72 (4H, t,

13 J = 7.6 Hz). C NMR (150 MHz, CDCl3): δ 208.61, 161.27 (d, J = 245.0 Hz), 130.88 (d,

J = 4.8 Hz), 128.05 (d, J = 8.1 Hz), 127.88 (d, J = 15.4 Hz), 124.20 (d, J = 3.7 Hz),

115.40 (d, J = 21.9 Hz), 42.90 (d, J = 1.4 Hz), 23.56 (d, J = 2.8 Hz). 19F NMR (565 MHz,

CDCl3) δ -114.37 – -124.62 (m).

(1E,4E)-1,5-Diphenylpenta-1,4-dien-3-one thiosemicarbazone (26)

Thiosemicarbazide (0.116 mg, 1.28 mmol), p -toluene sulfonic acid monohydrate

(0.0025 g, .013 mmol), and (1E,4E)-1,5-diphenylpenta-1,4-dien-3-one (0.150 g, 0.64

mmol) were dissolved in ethanol (2 mL) and the reaction was carried out at 100 oC for 30

min under microwave irradiation. The solvent was removed under reduced pressure and products were extracted using ethyl acetate (2 x 25 mL) from water (25 mL). The

combined organic phases were dried over sodium sulfate and concentrated. Purification

using flash chromatography (silica gel, hexanes:ethyl acetate, gradient 80:20 to 70:30)

afforded (1E,4E)-1,5-diphenylpenta-1,4-dien-3-one thiosemicarbazone (0.146 g, 0.475

1 mmol, 74% yield). H NMR (500 MHz, DMSO-d6): δ 11.05 (1H, s), 8.36 (1H, s), 8.00

(1H, s), 7.84-7.80 (2H, m), 7.70-7.62 (3H, m), 7.49 (1H, d, J = 16 Hz), 7.47-7.29 (8H,

13 m), 7.21 (1H, d, J = 16 Hz). C NMR (125 MHz, DMSO-d6): δ 178.53, 143.91, 137.20,

136.63, 136.26, 133.63, 129.10, 128.65, 128.59, 128.30, 127.93, 127.17, 123.19, 118.29.

+ + HPLC (Method A) 9.34 min. HRMS (ESI) calculated for C18H17N3SNa [M+Na]

330.10354, found 330.10376.

152 (1E,4E)-1,5-Bis(3-bromophenyl)penta-1,4-dien-3-one thiosemicarbazone (27)

Thiosemicarbazide (0.182 g, 2.00 mmol), p -toluene sulfonic acid monohydrate

(0.0039 g, 0.021 mmol), and 1,5-bis-3-bromophenyl-3-pentanone (0.392 g, 1.00 mmol)

were dissolved in in anhydrous ethanol (3 mL) and the reaction was carried out at 100 oC for 30 min under microwave irradiation. The reaction mixture was concentrated and purification using flash chromatography (silica gel, hexanes:ethyl acetate, gradient 80:20 to 50:50) afforded 1,5-bis(3-chlorophenyl)-3-pentanone thiosemicarbazone (0.368 g,

1 0.791 mmol 79 % yield) as a yellow solid. H NMR (600 MHz, DMSO-d6): δ 11.14 (1H,

s), 8.45 (1H, s), 8.07 (1H, t, J = 1.8 Hz), 8.07 (1H, s), 7.82 – 7.78 (1H, m), 7.71 – 7.65

(2H, m), 7.56 (1H, ddd, J = 8.0, 2.0, 1.0 Hz), 7.54 – 7.48 (2H, m), 7.42 – 7.34 (3H, m),

13 7.27 (1H, d, J = 16.0 Hz). C NMR (150 MHz, DMSO-d6): δ 178.66, 142.62, 139.24,

138.85, 135.51, 131.88, 131.59, 130.78, 130.71, 129.90, 129.57, 127.23, 126.14, 124.28,

122.19, 119.46. HPLC (Method B) 14.82 min. HRMS (ESI) calculated for

+ + C18H15N3SBr2H [M+H] 463.94262, found 463.94299.

(1E,4E)-1,5-Bis(3-chlorophenyl)penta-1,4-dien-3-one thiosemicarbazone (28)

Thiosemicarbazide (0.154 g, 1.68 mmol), p -toluene sulfonic acid monohydrate

(0.0039 g, 0.021 mmol), and 1,5-bis-3-chlorophenyl-3-pentanone (0.127 g, 0.419 mmol)

were dissolved in in anhydrous tetrahydrofuran (2 mL) and the reaction was carried out at

90 oC for 30 min under microwave irradiation. The reaction mixture was concentrated

and purification using flash chromatography (silica gel, hexanes:ethyl acetate, gradient

93:07 to 40:60) afforded 1,5-bis(3-chlorophenyl)-3-pentanone thiosemicarbazone (0.062

1 g, 0.016 mmol 38 % yield) as a yellow solid. H NMR (600 MHz, DMSO-d6): δ 11.15

(1H, s), 8.45 (1H, s), 8.07 (1H, s), 7.96 (1H, t, J = 1.8 Hz), 7.79 (1H, t, J = 1.8 Hz), 7.75

153 (dt, J = 7.7, 1.5 Hz, 1H), 7.70 (1H, d, J = 16.2 Hz), 7.67 – 7.62 (2H, m), 7.53 (1H, d, J =

16.0 Hz), 7.47 (1H, t, J = 7.8 Hz), 7.46 – 7.40 (2H, m), 7.39 (1H, d, J = 16.2 Hz), 7.37

(1H, ddd, J = 7.9, 2.1, 1.0 Hz), 7.29 (1H, d, J = 16.0 H). 13C NMR (150 MHz, DMSO-

d6): δ 178.69, 142.60, 138.98, 138.60, 135.59, 133.58, 133.56, 131.95, 130.46, 128.72,

127.90, 126.96, 126.63, 125.88, 124.30, 119.49. HPLC (Method A) 14.82 min. HRMS

+ + (ESI) calculated for C18H15Cl2N3SH [M+H] 376.04365, found 376.04376.

(1E,4E)-1,5-Bis(3-hydroxyphenyl)penta-1,4-dien-3-one thiosemicarbazone (29)

Thiosemicarbazide (0.102 g, 1.13 mmol), p -toluene sulfonic acid monohydrate

(0.0025 g, 0.013 mmol), and (1E,4E)-1,5-bis(3-hydroxyphenyl)penta-1,4-dien-3-one

(0.150 g, 0.563 mmol) were dissolved in in anhydrous tetrahydrofuran (10 mL) and the

reaction was refluxed for 22 h. The reaction mixture was concentrated and products were

extracted with ethyl acetate (2 x 40 mL) from water (80 mL) and dried over sodium

sulfate. Purification using flash chromatography (silica gel, dichloromethane: methanol,

gradient 97.5:2.5 to 95:05) afforded (1E,4E)-1,5-bis(3-hydroxyphenyl)penta-1,4-dien-3-

one thiosemicarbazone (0.144 g, 0.425 mmol 75 % yield) as a yellow solid. 1H NMR

(500 MHz, DMSO-d6): δ 11.01 (1H, s), 9.48 (1H, s), 9.43 (1H, s), 8.31 (1H, s), 7.99 (1H, s), 7.53 (1H, d, J = 16.2 Hz), 7.36 (1H, d, J = 16.0 Hz), 7.23-7.16 (5H, m), 7.11-7..07

(2H, m), 7.03 (1H, t, J = 2.1 Hz), 6.80-6.77 (1H, m), 6.73 (1H, ddd, J = 8.0 Hz, 2.4 Hz,

13 1.0 Hz). C NMR (125 MHz, DMSO-d6): δ 178.51, 157.57, 157.55, 144.36, 137.93,

137.57, 137.41, 133.94, 129.56, 129.47, 123.20, 119.21, 118.22, 116.30, 115.51, 114.30,

+ 113.76. HPLC (Method C) 7.35 min. HRMS (ESI) calculated for C18H17OH2N3SH

[M+H]+ 340.11142, found 340.11175.

154 (1E,4E)-1,5-bis(4-fluorophenyl)penta-1,4-dien-3-one thiosemicarbazone (30)

Thiosemicarbazide (0.375 g, 4.00 mmol) and (1E,4E)-1,5-bis(4-

fluorophenyl)penta-1,4-dien-3-one (0.270 g, 1.00 mmol) were dissolved in

tetrahydrofuran (15 mL) and sonicated for 15 h. The solvent was removed under reduced

pressure and products were extracted with dichloromethane from water. The combined

organic phases were washed with brine, dried over sodium sulfate and concentrated.

Purification using flash chromatography (silica gel, hexanes:ethyl acetate, gradient 90:10

to 20:80) afforded (1E,4E)-1,5-bis(4-fluorophenyl)penta-1,4-dien-3-one

thiosemicarbazone (0.214 g, 0.624 mmol, 62% yield) as an orange solid. 1H NMR (500

MHz, acetone-d6) δ 9.84 (1H, s), 7.88 (1H, s), 7.82 (2H, dd, J= 8.8 Hz, 5.4 Hz), 7.68

(2H, dd, J= 8.8 Hz, 5.4 Hz), 7.55 (1H, s), 7.39 (1H, d, J = 16.4 Hz ), 7.38 (1H, d, J =

16.1 Hz), 7.31 (1H, d, J= 16.4 Hz), 7.20 (1H, t, J= 8.8 Hz), 7.16 (1H, t, J= 8.8 Hz), 7.11

13 (1H, d, J= 16.1 Hz); C NMR (125 MHz, acetone-d6) δ 180.75, 164.14 (d, J = 248 Hz),

163.66 (d, J = 247 Hz), 146.61, 138.46, 134.59, 134.19 (d, J = 3.3 Hz), 133.47 (d, J = 3.3

Hz), 130.66 (d, J = 8.6 Hz), 129.91 (d, J = 8.1 Hz), 125.65 (d, J = 2.4 Hz), 118.05 (d, J =

2.4 Hz), 116.45 (d, J = 21.9 Hz), 116.42(d, J = 21.9 Hz). 19F NMR (470 MHz, acetone- d6): δ 113.41 (1F, tt, J = 9.1 Hz, 5.6 Hz), 114.56 (1F, tt, J = 8.9 Hz, 5.5 Hz). HPLC

+ + (Method B) 9.19 min. HRMS (ESI) calculated for C18H15F2N3SH [M+H] 344.10275,

found 344.10307.

(1E,4E)-1,5-bis(2-fluorophenyl)penta-1,4-dien-3-one thiosemicarbazone (31)

Thiosemicarbazide (0.279 g, 3.07 mmol) and (1E,4E)-1,5-bis(2-

fluorophenyl)penta-1,4-dien-3-one (0.207 g, 0.766 mmol) were dissolved in

tetrahydrofuran (15 mL) and sonicated for 10 h. After 10 h, no reaction was observed

155 and the reaction mixture was refluxed overnight. The solvent was removed under

reduced pressure and products were extracted with ethyl acetate (2 x 25 mL) from water

(50 mL). The combined organic phases were dried over sodium sulfate and concentrated.

Purification using flash chromatography (silica gel, hexanes:ethyl acetate, gradient 85:15

to 50:50) afforded (1E,4E)-1,5-bis(2-fluorophenyl)penta-1,4-dien-3-one

1 thiosemicarbazone (0.068 g, 0.198 mmol, 26% yield). H NMR (500 MHz, DMSO-d6): δ

11.08 (1H, s), 8.39 (1H, s), 8.10 (1H, td, J = 7.8 Hz, 1.7 Hz), 8.03 (1H, s), 7.81 (1H, td, J

= 7.7 Hz, 1.7 Hz), 7.62 (1H, d, J = 16.3 Hz), 7.46-7.35 (3H, m), 7.31-7.20 (6H, m). 13C

NMR (125 MHz, DMSO-d6): δ 178.79 , 160.05 (d, J = 249.6 Hz), 160.03 (d, J = 249.0

Hz), 144.19 , 131.16 (d, J = 8.5 Hz), 130.23 (d, J = 8.4 Hz), 128.43 (d, J = 3.3 Hz),

128.36 (d, J = 4.6 Hz), 128.11 (d, J = 2.6 Hz), 126.75 (d, J = 5.5 Hz), 126.32 (d, J = 2.8

Hz), 124.77 (d, J = 20.9 Hz), 124.74 (d, J = 20.9 Hz), 124.07 (d, J = 11.9 Hz), 123.64 (d,

J = 11.5 Hz), 120.54 (d, J = 2.7 Hz), 115.92 (d, J = 21.8 Hz), 115.81 (d, J = 21.6 Hz).19F

- - - - NMR (470 MHz, DMSO-d6) δ 117.11- 117.16 (1F, m), 117.97- 118.02 (1F, m). HPLC

+ + (Method B) 9.29 min. HRMS (ESI) calculated for C18H15N3SF2H [M+H] 344.1028,

found 344.1027.

1,5-Diphenyl-3-pentanone thiosemicarbazone (32)

1,3-Diphenyl-3-pentanone (0.340 g, 1.43 mmol) was dissolved in tetrahydrofuran

(10 mL) followed by the addition of p -toluene sulfonic acid monohydrate (0.0053 g,

0.028 mmol) and thiosemicarbazide (0.260 g, 2.86 mmol). The solvent was removed

under reduced pressure and products were extracted with dichloromethane from water.

The combined organic phases were dried over sodium sulfate and concentrated.

Purification using flash chromatography (silica gel, hexanes:ethyl acetate, gradient 90:10

156 to 20:80) afforded 1,5-diphenyl-3-pentanone thiosemicarbazone (0.258 g, 0.828 mmol,

1 58% yield) as a white solid. H NMR (500 MHz, CDCl3): δ 8.59 (1H, s), 7.34-7.24 (5H,

m), 7.21-7.18 (1H, m), 7.16-7.11 (4H, m), 7.08 (1H, s), 6.42 (1H, s). 13C NMR (125

MHz, CDCl3): δ 179.14, 155.08, 140.99, 139.52, 128.98, 128.61, 128.26, 128.20, 127.00,

126.28, 38.32, 32.12, 31.94, 31.43. HPLC (Method A) 8.22 min.

1,5-Bis(3-hydroxyphenyl)-3-pentanone thiosemicarbazone (33)

Thiosemicarbazide (0.103 g, 1.13 mmol), p -toluene sulfonic acid monohydrate

(0.0021 g, 0.011 mmol), and 1,5-bis-3-hydroxyphenyl-3-pentanone (0.154 g, 0.569

mmol) were dissolved in in anhydrous tetrahydrofuran (10 mL) and the reaction was

refluxed for 11 h. The reaction mixture was concentrated, products were extracted with

ethyl acetate (2 x 30 mL) from water (60 mL), and combined organic phases were dried

over sodium sulfate. Purification using flash chromatography (silica gel,

dichloromethane: methanol, gradient 97.5:2.5 to 95:05) afforded 1,5-bis(3-

hydroxyphenyl)-3-pentanone thiosemicarbazone (0.172 g, 0.501 mmol 88 % yield) as a

1 white solid. H NMR (500 MHz, DMSO-d6): δ 10.25 (1H, s), 9.24 (1H, s), 9.20 (1H, s),

8.05 (1H, s), 7.45 (1H, s), 7.08-7.02 (2H, m), 6.69-6.67 (2H, m), 6.62-6.54 (5H, m), 2.74-

13 2.71 (2H, m), 2.68-2.60 (4H, m), 2.46-2.43 (2H, m). C NMR (125 MHz, DMSO-d6): δ

78.62, 157.25, 157.24, 155.56, 143.07, 142.26, 129.11, 129.09, 118.91, 118.86, 115.27,

115.11, 112.97, 112.68, 37.63, 31.54, 31.13, 30.52. HPLC (Method B) 2.48 min. HRMS

+ + (ESI) calculated for C18H21N3O 2SNa [M+Na] 366.1247, found 366.1246.

1,5-Bis(4-fluorophenyl)-3-pentanone thiosemicarbazone (34)

Thiosemicarbazide (0.910 g, 1.00 mmol), p -toluene sulfonic acid monohydrate

(0.0019 g, 0.010 mmol) and 1,5-Bis-4-fluorophenyl-3-pentanone (0.135 g, 0.493 mmol)

157 were dissolved in tetrahydrofuran (10 mL) and refluxed for 3.5 h. The solvent was

removed under reduced pressure and products were extracted with dichloromethane from

water. The combined organic phases were dried over sodium sulfate and concentrated.

Purification using flash chromatography (silica gel, hexanes:ethyl acetate, gradient 88:12

to 00:100) afforded 1,5-Bis-4-fluorophenyl-3-pentanone thiosemicarbazone (0.167 g,

1 0.481 mmol, 98% yield). H NMR (600 MHz, DMSO-d6): δ 10.30 (1H, s), 8.07 (1H, s),

7.46 (1H, s), 7.32 (2H, dd, J = 8.5, 5.7 Hz), 7.25 (2H, dd, J = 8.5, 5.7 Hz), 7.11 (2H, t, J =

8.9 Hz), 7.08 (2H, t, J = 8.9Hz), 2.83 (2H, t, J = 7.8 Hz), 2.75 – 2.63 (4H, m), 2.50 –

13 2.46 (2H, m). C NMR (150 MHz, DMSO-d6): δ 178.62, 160.73 (d, J = 241 Hz), 159.55

(d, J = 241 Hz), 154.81, 137.81 (d, J = 3.1 Hz), 136.97 (d, J = 3.0 Hz), 130.14 (d, J = 8.0

Hz), 129.98 (d, J = 7.9 Hz), 114.84 (d, J = 21 Hz), 114.83 (d, J = 21 Hz), 37.56, 31.31,

19 30.50, 29.67. F NMR (565 MHz, DMSO-d6) δ -117.24 (tt, J = 9.3, 5.7 Hz), -117.60 – -

- 117.68 (m). HPLC (Method A) 8.41 min. HRMS (ESI) calculated for C18H18N3SF2 [M-

H]-346.1195, found 346.1195.

1,5-Bis(2-fluorophenyl)-3-pentanone thiosemicarbazone (35)

Thiosemicarbazide (0.103 g, 1.13 mmol), p -toluene sulfonic acid monohydrate

(0.0033 g, 0.017 mmol) and 1,5-Bis-2-fluorophenyl-3-pentanone (0.155 g, 0.565 mmol) were dissolved in tetrahydrofuran (10 mL) and refluxed for 9 h. The solvent was removed under reduced pressure and products were extracted with ethyl acetate (2 x 25 mL) from water (50 mL). The combined organic phases were dried over sodium sulfate and concentrated. Purification using flash chromatography (silica gel, hexanes:ethyl acetate, gradient 90:10 to 50:50) afforded 1,5-Bis-2-fluorophenyl-3-pentanone thiosemicarbazone

1 (0.172 g, 0.495 mmol, 88% yield). H NMR (500 MHz, DMSO-d6): δ 10.25 (1H, s), 8.09

158 (1H, s), 7.46 (1H, s), 7.35 (1H, td, J = 7.7 Hz, 1.7 Hz), 7.30-7.21 (3H, m), 7.16-7.09 (4H, m) 2.86 (2H,t, J = 7.8 Hz), 2.77-2.74 (2H, m), 2.71-2.67 (2H, m), 2.49-2.47 (2H, m). 13C

NMR (125 MHz, DMSO-d6): δ 178.71, 160.48 (d, J = 243 Hz), 160.37 (d, J = 243 Hz),

154.33 , 130.76 (d, J = 4.8 Hz), 130.65 (d, J = 4.8 Hz), 128.24-128.05 (2C, m), 127.91 (d,

J = 8.1 Hz), 127.32 (d, J = 15.7 Hz), 124.29 (d, J = 3.3 Hz), 115.02 (d, J = 21.9 Hz),

115.00 (d, J = 21.9 Hz), 36.18 , 29.72 , 24.64 (d, J = 2.4 Hz), 23.62 (d, J = 2.4 Hz). 19F

- NMR (470 MHz, DMSO-d6) δ -118.89- 118.99 (2F, m). HPLC (Method B) 8.05 min.

+ + HRMS (ESI) calculated for C18H19N3SF2H [M+H] 348.1341, found 348.1342.

2,6-Bis((E)-4-fluorobenzylidene)cyclohexan-1-one (36)

4-Fluorobenzaldehyde (1.72 mL, 16.0 mmol) dissolved in cyclohexanone (0.828 mL, 8.0 mmol) was added dropwise to a solution of NaOH (4 g), ethanol (20 mL), and

water (30 mL) at 0 oC. After stirring at room temperature at 3 h, the reaction mixture was

filtered and washed with water (6 x 10 mL). The crude product was recrystallized from

ethanol and ethyl acetate to afford (1E,4E)-1,5-bis(4-fluorophenyl)lpenta-1,4-dien-3-one

1 (1.828 g, 5.62 mmol, 74% yield) as a yellow solid. H NMR (500 MHz, acetone-d6): δ

7.67 (2H, d, J = 2.1 Hz), 7.60 (4H, dd, J = 8.9, 5.5 Hz), 7.23 (4H, t, J = 8.8 Hz), 2.95

13 (4H, td, J = 6.4, 2.1 Hz), 1.80 (2H, p, J = 6.2 Hz). C NMR (125 MHz, acetone-d6): δ

189.27, 163.47 (d, J = 248.0 Hz), 137.13 (d, J = 2.1 Hz), 135.51 , 133.38 (d, J = 8.5 Hz),

133.24 (d, J = 3.4 Hz), 116.25 (d, J = 21.9 Hz), 28.96 , 23.62. 19F NMR (470 MHz, acetone-d6) δ -113.61 (tt, J = 8.8, 5.5 Hz).

159 (E)-7-(4-fluorobenzylidene)-3-(4-fluorophenyl)-7a-hydroxy-3a,4,5,6,7,7a-hexahydro-1H-

indazole-1-carbothioamide (37)

Titanium (IV) ispropoxide (0.592 mL, 2 mmol) was added dropwise to a solution

of 2,6-bis((E)-4-fluorobenzylidene)cyclohexan-1-one (0.310 g, 1.00 mmol) in

tetrahydrofuran (5 mL). After 10 min, thiosemicarbazide (0.182 g, 2.00 mmol) was

added and the reaction mixture was stirred at room temperature for 1h followed by reflux

for 20 h. The reaction was quenched with 1 M HCl (10 mL) and extracted with ethyl

acetate (2 x 20 mL). The combined organic phases were washed with saturated sodium

bicarbonate (20 mL), dried over sodium sulfate and concentrated. Purification using flash

chromatography (silica gel, hexanes:ethyl acetate, gradient 85:15 to 50:50) followed by

recrystallization from dichloromethane and pentane afforded (E)-7-(4-

fluorobenzylidene)-3-(4-fluorophenyl)-7a-hydroxy-3a,4,5,6,7,7a-hexahydro-1H-

indazole-1-carbothioamide (0.035 g,0.088 mmol, 9% yield) as beige crystals.

1 H NMR (500 MHz, acetone-d6): δ 8.02 (2H, dd, J = 9.0, 5.4 Hz), 7.95 (1H, s), 7.50 (1H,

s), 7.33 (2H, ddd, J = 8.8, 5.5, 0.7 Hz), 7.26 (2H, t, J = 8.8 Hz), 7.14 (2H, t, J = 8.9 Hz),

7.01 (1H, d, J = 2.2 Hz), 5.96 (1H, s), 3.88 (1H, dd, J = 6.9, 2.7 Hz), 2.74 (1H, ddd, J =

15.2, 9.8, 7.8 Hz), 2.33 (1H, ddt, J = 14.9, 9.8, 2.5 Hz), 1.74 (1H, tdd, J = 13.6, 6.7, 3.4

13 Hz), 1.67 – 1.48 (2H, m), 1.41 – 1.25 (1H, m). C NMR (125 MHz, acetone-d6): δ

178.69 , 164.83 (d, J = 249.0 Hz), 162.42 (d, J = 244.0 Hz), 155.62 , 138.94 , 134.77 (d, J

= 3.2 Hz), 131.80 (d, J = 7.8 Hz), 130.51 (d, J = 8.6 Hz), 127.74 (d, J = 3.2 Hz), 127.48 ,

116.73 (d, J = 22.0 Hz), 115.70 (d, J = 21.5 Hz), 96.93 , 56.89 , 25.48 , 24.48 , 18.59. 19F

NMR (470 MHz, acetone-d6) δ -111.31 (tt, J = 8.9, 5.4 Hz), -117.24 (tt, J = 9.0, 5.5 Hz).

160 - - HPLC (Method A) 12.23 min. HRMS (ESI) calculated for C21H18F2N3OSH [M-H]

398.11441, found 398.11502.

161

CHAPTER FIVE

Conclusions

Encouraged by efforts towards the development of cathepsin inhibitors

incorporating the thiosemicarbazone moiety in the Pinney and Trawick laboratories, an

extended library of aryl based thiosemicarbazones have been designed, synthesized, and

evaluated through collaboration for inhibitory activity against cathepsins L and B. From

an inhibitory design aspect, the aryl substituents can potentially bind subsites within the

cathepsin L active site through noncovalent interactions in a manner which positions the

thiocarbonyl of the thiosemicarbazone moiety within proximity to the Cys25 thiolate of

cathepsin L allowing for formation of a covalent bond between the inhibitor and enzyme.

Thirty-five thiosemicarbazone analogues which incorporated benzophenone, benzoylbenzophenone, di-2-naphthalenylmethanone, 1,3-diphenylpropan-2-one, dibenzylideneacetone, and 1,5-diphenyl-3-pentanone aryl-based scaffolds were

synthesized. Biochemical evaluation through collaboration with the Trawick laboratory

revealed fifteen analogues which potently inhibited cathepsin L (IC50 ≤ 10 µM). Among

the explored molecular frameworks, the benzoylbenzophenone thiosemicarbazone

analogues provided the greatest number of inhibitors which significantly impacted

cathepsin L proteolytic activity. The most potent cathepsin L inhibitor from this library,

1,3-bis(2-fluorobenzoyl)-5-bromobenzene thiosemicarbazone (KGP312; IC50 = 8.12 nM),

exerted greater effect against cathepsin L proteolytic potential in comparison to

previously published inhibitors in the Pinney and Trawick laboratories.

162 Improvement of the synthetic methodology for KGP94led to a route in which a difficult to purify byproduct was circumvented. Additionally, an effort to further the pre- clinical development of KGP94 has led to the synthesis of the water soluble phosphate prodrug KGP420.

Throughout the past decade, the evolution of thiosemicarbazone based cysteine protease inhibitors has led to analogues exhibiting great inhibitory effect against cathepsins. On-going collaborative efforts have been aimed towards the advancement of thiosemicarbazone based inhibitors as potential anti-metastatic agents. Expansion of the present work through the design and synthesis of an extended library could potentially lead to the development of cathepsin L inhibitors which exhibit greater potency, enhanced oral bioavailability, and/or are devoid of isomerization.

163

APPENDICES

164

APPENDICES

Appendix A ...... 166

Appendix B ...... 365

Appendix C ...... 504

165

APPENDIX A

Supplementary Data: Synthesis and biochemical evaluation of benzoylbenzophenone thiosemicarbazone analogues as potent and selective inhibitors of cathepsin L

This appendix is reprinted from Supplementary Data associated with Bioorganic and Medicinal Chemistry, Vol xx, Parker, E. N.; Song, J.; Kishore Kumar, G. D.; Odutola, S. O.; Chavarria, G. E.; Charlton-Sevcik, A. K.; Strecker, T. E.; Barnes, A. L.; Sudhan, D. R.; Wittenborn, T. R.; Siemann, D. W.; Horsman, M. R.; Chaplin, D. J.; Trawick, M. L.; Pinney, K. G. Synthesis and Biochemical Evaluation of Benzoylbenzophenone Thiosemicarbazone Analogues as Potent and Selective Inhibitors of Cathepsin L, 6974- 6992, Copyright 2015, with permission from Elsevier.

The author Erica N. Parker played a significant role in the preparation of the manuscript. Also, Erica N Parker contributed to this manuscript through the synthesis, purification, and characterization of final analogues 1, 2, 4, 9, 11, 13, 20, 22, 31, 32, 33 and their corresponding intermediates. The author Jiangli Song contributed to this manuscript through the synthesis, purification, and characterization of final analogues 8, 14 and their corresponding intermediates. The author G.D.K. Kumar contributed to this manuscript through the synthesis, purification, and characterization of final analogues 1, 2, 8 and their corresponding intermediates. The author Ashleigh L. Barnes contributed to this manuscript through the synthesis, purification, and characterization of final analogue 10 and corresponding intermediates.

166 167 168 169 170 171 172 173 174 175 176 177 178 179 180 181 182 183 184 185 186 187 188 189 190 191 192 193 194 195 196 197 198 199 200 201 202 203 204 205 206 207 208 209 210 211 212 213 214 215 216 217 218 219 220 221 222 223 224

225 226 227 228

229 230 231 232 233

234

235 236 237 238 239 240 241 242 243 244 245 246 247 248 249 250 251 252 253 254 255 256 257 258 259 260 261 262 263 264 265 266 267 268 269 270 271 272 273 274 275 276 277 278 279 280 281 282 283 284 285 286 287 288 289 290 291 292 293 294 295 296 297 298 299 300 301 302 303 304 305 306 307 308 309 310 311 312 313 314 315 316 317 318 319 320 321 322 323 324 325 326 327 328 329 330 331 332 333 334 335 336 337 338 339 340

341

342 343 344 345 346 347 348 349 350 351 352 353 354 355 356 357 358 359 360 361 362

363 364

APPENDIX B

Supplementary Data: Synthesis and Biological Evaluation of a Water Soluble Phosphate Prodrug Salt and Structural Analogues of KGP94, a Lead Inhibitor of Cathepsin L

This appendix will be published as supplementary data; : Parker, E. N.; Odutola, S. O.; Wang, Y.; Chavarria, G. E.; Strecker, T. E.; Chaplin, D. J.; Trawick, M. L.; Pinney, K. G. Synthesis and Biological Evaluation of a Water Soluble Phosphate Prodrug Salt and Structural Analogues of KGP94, a Lead Inhibitor of Cathepsin L. Journal to be Determined. (In Progress).

365

Supporting Information

Synthesis and Biological Evaluation of a Water Soluble Phosphate Prodrug Salt and

Structural Analogues of KGP94, a Lead Inhibitor of Cathepsin L

Erica N. Parker †, Samuel O. Odutola‡, Yifan Wang†, Gustavo E. Chavarria†, Tracy E.

Strecker†, David J. Chaplin§, Mary Lynn Trawick†, and Kevin G. Pinney*,‡,†

†Department of Chemistry and Biochemistry Baylor University One Bear Place #97348

Waco, TX 76798-7348

‡Institute of Biomedical Studies Baylor University One Bear Place #97224 Waco, TX

76798-7224

§OXiGENE, Inc. 701 Gateway Blvd, Suite 210 South San Francisco, CA 94080

366

Table of Contents

1 H NMR (500 MHz, CDCl3) of Compound 1 ...... 372

13 C NMR (125 MHz, CDCl3) of Compound 1 ...... 373

1 H NMR (600 MHz, CDCl3) of Compound 2 ...... 374

13 C NMR (150 MHz, CDCl3) of Compound 2 ...... 375

1 H NMR (600 MHz, CDCl3) of Precursor for Compound 3 ...... 376

13 C NMR (150 MHz, CDCl3) of Precursor for Compound 3 ...... 377

1 H NMR (600 MHz, CDCl3) of Compound 3 ...... 378

13 C NMR (150 MHz, CDCl3) of Compound 3 ...... 379

1 H NMR (500 MHz, CDCl3) of Compound 4 ...... 380

13 C NMR (125 MHz, CDCl3) of Compound 4 ...... 381

1 H NMR (600 MHz, CDCl3) of Compound 5 ...... 382

13 C NMR (150 MHz, CDCl3) of Compound 5 ...... 383

1 H NMR (500 MHz, CDCl3) of Compound 6 ...... 384

13 C NMR (125 MHz, CDCl3) of Compound 6 ...... 385

1 H NMR (600 MHz, CDCl3) of Compound 7 ...... 386

13 C NMR (150 MHz, CDCl3) of Compound 7 ...... 387

1 H NMR (600 MHz, CDCl3) of Compound 8 ...... 388

13 C NMR (150 MHz, CDCl3) of Compound 8 ...... 389

1 H NMR (600 MHz, CDCl3) of Compound 9 ...... 390

13 C NMR (150 MHz, CDCl3) of Compound 9 ...... 391

1 H NMR (500 MHz, CDCl3) of Compound 10 ...... 392

13 C NMR (125 MHz, CDCl3) of Compound 10 ...... 393

367

1 H NMR (DMSO-d6,, 600 MHz) of Precursor for Compound 11 ...... 394

13 C-NMR (DMSO-d6, 150 MHz) of Precursor for Compound 11 ...... 395

1 H NMR (DMSO-d6,, 600 MHz) of Compound 11 ...... 396

13 C-NMR (DMSO-d6, 150 MHz) of Compound 11 ...... 397

COSY (DMSO-d6, 600 MHz) of Compound 11 ...... 398

Expansion COSY (DMSO-d6, 600 MHz) of Compound 11 ...... 399

ROESY (DMSO-d6, 600 MHz) of Compound 11 ...... 400

HPLC Trace of Compound 11 ...... 401

1 H NMR (600 MHz, CDCl3) of Precursor for Compound 12 ...... 404

13 C NMR (150 MHz, CDCl3) of Precursor for Compound 12 ...... 405

1 H NMR (600 MHz, acetone-d6) of Compound 12 ...... 406

13 C NMR (150 MHz, acetone-d6) of Compound 12 ...... 407

Mass Spec of Compound 12 ...... 408

HPLC Trace of Compound 12 ...... 409

H NMR (600 MHz, acetone-d6) of Precursor for Compound 13 ...... 412

13 C NMR (150 MHz, acetone-d6) of Precursor for Compound 13 ...... 413

1 H NMR (600 MHz, DMSO-d6) of Compound 13 ...... 414

13 C NMR (150 MHz, DMSO-d6) of Compound 13 ...... 415

COSY of Compound 13 ...... 416

ROESY of Compound 13 ...... 417

Isomerization of Compound 13 ...... 418

Mass Spec of Compound 13 ...... 419

HPLC Trace of Compound 13 ...... 420

368

1 H NMR (600 MHz, CDCl3) of Precursor for Compound 14 ...... 426

1 H NMR (600 MHz, CDCl3) of Compound 14 ...... 427

13 C NMR (150 MHz, CDCl3) of Compound 14 ...... 428

1 H NMR (600 MHz, CDCl3) of Compound 15 ...... 429

13 C NMR (150 MHz, CDCl3) of Compound 15 ...... 430

1 H NMR (600 MHz, CDCl3) of Compound 16 ...... 431

13 C NMR (150 MHz, CDCl3) of Compound 16 ...... 432

1 H NMR (600 MHz, acetone-d6) of Compound 17 ...... 433

13 C NMR (150 MHz, acetone-d6) of Compound 17 ...... 434

1 H NMR (600 MHz, acetone-d6) of Compound 18 ...... 435

13 C NMR (150 MHz, acetone-d6) of Compound 18 ...... 436

1 H NMR (600 MHz, DMSO-d6) of Compound 19 ...... 437

13 C NMR (150 MHz, DMSO-d6) of Compound 19 ...... 438

COSY of Compound 19 ...... 439

ROESY of Compound 19 ...... 440

Expanded ROESY of Compound 19 ...... 441

Isomerization of Compound 19 ...... 442

Mass Spec of Compound 19 ...... 444

HPLC Trace of Compound 19 ...... 445

1 H NMR (600 MHz, DMSO-d6) of Compound 20 ...... 451

13 C NMR (150 MHz, DMSO-d6) of Compound 20 ...... 452

COSY of Compound 20 ...... 453

ROESY of Compound 20 ...... 454

369

Isomerization of Compound 20 ...... 455

Mass Spec of Compound 20 ...... 457

HPLC Trace of Compound 20 ...... 458

1 H NMR (600 MHz, DMSO-d6) of Compound 21 ...... 464

13 C NMR (150 MHz, DMSO-d6) of Compound 21 ...... 465

COSY of Compound 21 ...... 466

ROESY of Compound 21 ...... 467

Isomerization of Compound 21 ...... 468

Mass Spec of Compound 21 ...... 469

HPLC Trace of Compound 21 ...... 470

1 H NMR (600 MHz, DMSO-d6) of Compound 22 ...... 476

13 C NMR (150 MHz, DMSO-d6) of Compound 22 ...... 477

COSY of Compound 22 ...... 478

ROESY of Compound 22 ...... 479

Isomerization of Compound 22 ...... 480

Mass Spec of Compound 22 ...... 481

HPLC Trace of Compound 22 ...... 482

1 H-NMR (DMSO-d6, 500 MHz) of Compound 23 ...... 488

13 C-NMR (DMSO-d6, 125 MHz) Compound 23 ...... 489

1 H-NMR (DMSO-d6, 500 MHz) of Compound 24 ...... 490

13 C-NMR (DMSO-d6, 125 MHz) Compound 24 ...... 491

31P-NMR (DMSO, 85 % phosphoric acid as external standard, decoupled, 243 MHz) of Compound 24 ...... 492

370

1 H-NMR (DMSO-d6, 600 MHz) of Compound 25 ...... 493

13 C-NMR (DMSO-d6, 150 MHz) Compound of 25 ...... 494

31P-NMR (DMSO, 85 % phosphoric acid as external standard, decoupled, 243 MHz) of Compound 25 ...... 495

1 H-NMR (D2O, 600 MHz) of Compound 27 ...... 496

13 C-NMR (D2O, 150 MHz) Compound of 27 ...... 497

31 P-NMR (D2O, 85 % phosphoric acid as external standard, decoupled, 243 MHz) of

Compound 27 ...... 498

Mass Spec of Compound 27 ...... 499

HPLC Trace of Compound 27 ...... 501

371

1 H NMR (500 MHz, CDCl3) of Compound 372

13 C NMR (125 MHz, CDCl3) of Compound 1 373

1 H NMR (600 MHz, CDCl3) of Compound 2 374

13 C NMR (150 MHz, CDCl3) of Compound 2 375

1 H NMR (600 MHz, CDCl3) of Precursor for Compound 3 376

13 C NMR (150 MHz, CDCl3) of Precursor for Compound 3 377

1 H NMR (600 MHz, CDCl3) of Compound 3 378

13 C NMR (150 MHz, CDCl3) of Compound 3 379

1 H NMR (500 MHz, CDCl3) of Compound 4 380

13 C NMR (125 MHz, CDCl3) of Compound 4 381

1 H NMR (600 MHz, CDCl3) of Compound 5 382

13 C NMR (150 MHz, CDCl3) of Compound 5 383

1 H NMR (500 MHz, CDCl3) of Compound 6 384

13 C NMR (125 MHz, CDCl3) of Compound 6 385

1 H NMR (600 MHz, CDCl3) of Compound 7 386

13 C NMR (150 MHz, CDCl3) of Compound 7 387

1 H NMR (600 MHz, CDCl3) of Compound 8 388

13 C NMR (150 MHz, CDCl3) of Compound 8 389

1 H NMR (600 MHz, CDCl3) of Compound 9 390

13 C NMR (150 MHz, CDCl3) of Compound 9 391

1 H NMR (500 MHz, CDCl3) of Compound 10 392

13 C NMR (125 MHz, CDCl3) of Compound 10 393

1 H NMR (DMSO-d6,, 600 MHz) of Precursor for Compound 11 394

13 C-NMR (DMSO-d6, 150 MHz) of Precursor for Compound 11 395

1 H NMR (DMSO-d6,, 600 MHz) of Compound 11 396

13 C-NMR (DMSO-d6, 150 MHz) of Compound 11 397

COSY (DMSO-d6, 600 MHz) of Compound 11

398

Expansion COSY (DMSO-d6, 600 MHz) of Compound 11 399

ROESY (DMSO-d6, 600 MHz) of Compound 11 400

HPLC Trace of Compound 11

401

HPLC Trace of Compound 11

402

HPLC Trace of Compound 11 .

403

1 H NMR (600 MHz, CDCl3) of Precursor for Compound 12 404

13 C NMR (150 MHz, CDCl3) of Precursor for Compound 12 405

1 H NMR (600 MHz, acetone-d6) of Compound 12 406

13 C NMR (150 MHz, acetone-d6) of Compound 12 407

Mass Spec of Compound 12

C:\Xcalibur\...\ENP_VI_49____Orbi_-ESI 5/7/2015 11:46:12 PM

ENP_VI_49____Orbi_-ESI #2-7 RT: 0.02-0.09 AV: 6 NL: 1.80E6 T: FTMS - p ESI Full ms [100.00-1000.00] 443.8844 100

90

80

70

60 445.8815 441.8874 50

40 446.8840

Relative Abundance 30

20

479.8607 541.8514 10 325.1840 384.9014 426.8581 539.8538 756.0608 148.1855 195.9401 265.1477 297.1528 563.8334 631.8217 683.7824 710.0395 784.0926 825.0758 0 408 150 200 250 300 350 400 450 500 550 600 650 700 750 800 m/z

443.88444 NL: 100 1.80E6 80 ENP_VI_49____Orbi_- ESI#2-7 RT: 0.02-0.09 AV: 6 T: FTMS - p ESI 60 441.88740 445.88153 Full ms [100.00-1000.00] 40 445.71627 446.04591 442.04947 443.72053 20 444.88691 Relative Abundance Relative 442.89032 446.88403 441.72466 444.04738 447.87645 448.87995 451.23103 0 443.88455 NL: 100 4.01E5

80 C14 H11Br2 N3 O2 S+H: C14 H10 Br2 N3 O2 S1 60 pa Chrg -1 441.88660 445.88250 40

20 444.88790 442.88995 446.88586 447.87830 448.88165 449.88501 450.88087 451.88422 0 438 439 440 441 442 443 444 445 446 447 448 449 450 451 452 m/z

HPLC Trace of Compound 12

409

HPLC Trace of Compound 12

410

HPLC Trace of Compound 12

411

H NMR (600 MHz, acetone-d6) of Precursor for Compound 13 412

13 C NMR (150 MHz, acetone-d6) of Precursor for Compound 13 413

1 H NMR (600 MHz, DMSO-d6) of Compound 13 414

13 C NMR (150 MHz, DMSO-d6) of Compound 13 415

COSY of Compound 13 416

ROESY of Compound 13 417

Isomerization of Compound 13

0 h in DMSO 418

Mass Spec of Compound 13

C:\Xcalibur\...\ENP_V_78_Orbi_+ESI 7/30/2014 9:10:22 PM

ENP_V_78_Orbi_+ESI #1-10 RT: 0.00-0.09 AV: 10 NL: 6.70E6 T: FTMS + p ESI Full ms [100.00-2000.00] 429.9044 100

90

80

70

60 427.9069 431.9021 50

40

Relative Abundance 30

20 451.8859 417.2093 10 313.1435 355.9103 453.8837 391.2842 576.9213 142.9478 162.9686 205.0859 240.9250 276.0019 338.9009 491.8864 548.5034 639.8506 654.3310 669.8224 716.3455 0

419 150 200 250 300 350 400 450 500 550 600 650 700 m/z

429.90443 NL: 100 6.70E6 80 ENP_V_78_Orbi_+ ESI#1-10 RT: 430.06003 0.00-0.09 AV: 10 T: 60 427.90685 429.75002 431.90206 FTMS + p ESI Full ms 40 [100.00-2000.00]

20 430.90729 Relative Abundance 428.90983 432.90479 425.88972 427.37902 433.89704 434.90082 436.87786 437.90469 439.90617 441.29646 441.90394 0 429.90419 NL: 100 4.02E5

80 C14 H11Br2 N3 OS +H: C14 H12 Br2 N3 O1S1 60 pa Chrg 1 427.90623 431.90214 40

20 430.90754 428.90959 432.90550 433.89794 434.90129 435.90465 436.90051 437.90386 0 425 426 427 428 429 430 431 432 433 434 435 436 437 438 439 440 441 442 m/z

HPLC Trace of Compound 13

420

HPLC Trace of Compound 13

421

HPLC Trace of Compound 13

422

HPLC Trace of Compound 13 (1h in ACN/Water)

423

HPLC Trace of Compound 13 (1h in ACN/Water)

424

HPLC Trace of Compound 13 (1h in ACN/Water)

425

1 H NMR (600 MHz, CDCl3) of Precursor for Compound 14 426

1 H NMR (600 MHz, CDCl3) of Compound 14 427

13 C NMR (150 MHz, CDCl3) of Compound 14 428

1 H NMR (600 MHz, CDCl3) of Compound 15 429

13 C NMR (150 MHz, CDCl3) of Compound 15 430

1 H NMR (600 MHz, CDCl3) of Compound 16 431

13 C NMR (150 MHz, CDCl3) of Compound 16 432

1 H NMR (600 MHz, acetone-d6) of Compound 17 433

13 C NMR (150 MHz, acetone-d6) of Compound 17 434

1 H NMR (600 MHz, acetone-d6) of Compound 18 435

13 C NMR (150 MHz, acetone-d6) of Compound 18 436

1 H NMR (600 MHz, DMSO-d6) of Compound 19 437

13 C NMR (150 MHz, DMSO-d6) of Compound 19 438

COSY of Compound 19 439

ROESY of Compound 19 440

Expanded ROESY of Compound 19 441

Isomerization of Compound 19

0 h in DMSO 442

Isomerization of Compound 19

0 h in DMSO 443

Mass Spec of Compound 19

C:\Xcalibur\...\ENPVI16_Orbi_+ESI 12/12/2014 2:23:16 PM

ENPVI16_Orbi_+ESI #12 RT: 0.10 AV: 1 NL: 1.81E7 T: FTMS + p ESI Full ms [120.00-1000.00] 396.0200 100 z=1 90 80 70 60 50 40 418.0017 z=1 Relative Abundance 30 283.0789 20 z=1 149.0120 207.0629 253.0708 477.9743433.9756 610.5015 850.9481 10 318.0127 550.6284 665.0971 753.0292 888.9222 954.3491 997.5206 z=? z=1 z=1 z=1z=1 z=2 z=1 0 z=1 z=? z=? z=1 z=1 z=? z=?

444 581.1449 811.0157 150 200 250 300 350 376.9955 400 450 500 550 600 650 700 750 800 850 900 950 1000 z=1 z=? z=1 m/z

416.00403 418.00171 NL: 416.44632 7.93E6 100 z=1 z=1 418.44745z=? ENPVI16_Orbi_+ 80 z=? ESI#12 RT: 0.10 AV: 60 1 T: FTMS + p ESI Full ms [120.00-1000.00] 40 20 413.26617 413.99460 415.49588 417.00729 419.00482 421.00079 421.74930424.03229 426.02960 427.99142 Relative Abundance z=? 0 z=? z=? z=1 z=1 419.99725z=1 z=?z=1 z=1 z=? z=1 416.00388 418.00183 100 NL: 16 H16 BrN 3 O2 S+Na: 80 3.98E516CH16 Br 1N3 O2 S 1Na 1 pa Chrg 1 60 C 40 20 417.00724 419.00519 423.00020 424.00355 425.00445 426.00780 0 412 413 414 415 416 417 418 419419.99763 421.00099420 422.00434421 422 423 424 425 426 427 428 m/z

HPLC Trace of Compound 19

445

HPLC Trace of Compound 19

446

HPLC Trace of Compound 19

447

HPLC Trace of Compound 19 (1 h in ACN/Water)

448

HPLC Trace of Compound 19 (1 h in ACN/Water)

449

HPLC Trace of Compound 19 (1 h in ACN/Water)

450

1 H NMR (600 MHz, DMSO-d6) of Compound 20 451

13 C NMR (150 MHz, DMSO-d6) of Compound 20 452

COSY of Compound 20 453

ROESY of Compound 20 454

Isomerization of Compound 20

0 h in DMSO 455

Isomerization of Compound 20

0 h in DMSO 456

Mass Spec of Compound 20

C:\Xcalibur\...\ENPVI15_Orbi_+ESI 12/12/2014 2:09:45 PM

ENPVI15_Orbi_+ESI #12 RT: 0.11 AV: 1 NL: 6.29E6 T: FTMS + p ESI Full ms [120.00-1000.00] 473.9305 100 z=1 90 80 70 60 50 40

Relative Abundance 30 397.9209 495.9121 z=1 z=1 20 207.0627 456.9037 603.1266 968.8342 10 129.0301 283.0789 335.0737 511.8866 729.3650665.0970 910.8491796.8489 870.8431 z=1 z=1 z=1 z=2 z=? z=? z=? z=1 z=2z=1 z=1 z=1 0 z=? 457 150 200 250 300 350 400 450 500 550 600 650 700 750 800 850 900 950 1000 m/z

473.93045 NL: 6.29E6 100 z=1 ENPVI15_Orbi_+ 80 ESI#12 RT: 0.11 AV: 60 471.93283 475.92804 1 T: FTMS + p ESI Full z=1 ms [120.00-1000.00] 40 z=? 20 469.32877 471.10550 474.93317 478.92566 481.96509 483.12106

Relative Abundance 472.93582 476.93076 0 z=? z=? z=1 477.92224z=1 z=? z=? z=? z=1 473.93040 z=1 100 NL: 16 H15 Br 2 N3 O2 S+H: 80 3.92E516CH16 Br 2 N3 O2 S 1 pa Chrg 1 60 C 471.93245 40 475.92836 20 474.93376 478.92751 480.92672 481.93008 482.93097 0 472.93580 476.93171 469 470 471 472 473 474 475 476 477477.92415 478 479 480 481 482 483 484 485 486 487 m/z

HPLC Trace of Compound 20

458

HPLC Trace of Compound 20

459

HPLC Trace of Compound 20

460

HPLC Trace of Compound 20 (3 h in ACN/Water)

461

HPLC Trace of Compound 20 (3 h in ACN/Water)

462

HPLC Trace of Compound 20 (3 h in ACN/Water)

463

1 H NMR (600 MHz, DMSO-d6) of Compound 21 464

13 C NMR (150 MHz, DMSO-d6) of Compound 21 465

COSY of Compound 21 466

ROESY of Compound 21 467

Isomerization of Compound 21

0 h in DMSO 468

Mass Spec of Compound 21

C:\Xcalibur\...\ENPVI12_run3_Orbi_+ESI 12/12/2014 2:51:33 PM

ENPVI12_run3_Orbi_+ESI #10 RT: 0.09 AV: 1 NL: 8.44E6 T: FTMS + p ESI Full ms [120.00-1000.00] 367.9886 100 z=1 90 80 70 60 50 40

Relative Abundance 30 389.9704 289.9812 20 z=1 413.2661 603.1268 207.0627 z=1 451.9407 754.9535 796.9417 10 129.0301 253.0708 z=1505.0901 550.6275640.0009 z=1698.9848 840.9058 887.3853 934.4388 973.5638 z=1 z=1 z=1 z=? z=1 z=1 z=1 z=? z=1 z=1 z=1 0 350.9620 z=3 z=2 z=?

469 150 200 250 300 z=1350 400 450 500 550 600 650 700 750 800 850 900 950 1000 m/z

NL: 8.44E6 365.99097 367.98856 ENPVI12_run3_Orbi_+ 100 z=1 z=1 ESI#10 RT: 0.09 AV: 1 T: FTMS + p ESI 80 Full ms 60 [120.00-1000.00] 40

20 365.26608 366.99402 368.99164 373.99304 375.25003 385.29196 Relative Abundance z=1 z=? z=1 377.99054 379.98840 z=? 0 362.05023 363.25046 z=1 z=1 370.98715 382.00394 383.98349 NL: z=1 z=1 z=? z=1 365.99064 z=1 z=1 z=1 4.07E5 100 367.98859 C 14 H12 BrN 3 O2 S+H: 80 14CH13 Br 1N3 O2 S 1 pa Chrg 1 60 40 20 0 366.99399 368.99194 362 364 366 368 370.98774370 372.98695372 374.99120374 376 378 380 382 384 386 m/z

HPLC Trace of Compound 21

470

HPLC Trace of Compound 21

471

HPLC Trace of Compound 21

472

HPLC Trace of Compound 21 (1 h in ACN/Water)

473

HPLC Trace of Compound 21 (1 h in ACN/Water)

474

HPLC Trace of Compound 21 (1 h in ACN/Water)

475

1 H NMR (600 MHz, DMSO-d6) of Compound 22 476

13 C NMR (150 MHz, DMSO-d6) of Compound 22 477

COSY of Compound 22 478

ROESY of Compound 22 479

Isomerization of Compound 22

0 h in DMSO 480

Mass Spec of Compound 22

C:\Xcalibur\...\ENPVI13_Orbi_+ESI 12/12/2014 1:46:12 PM

ENPVI13_Orbi_+ESI #2-9 RT: 0.01-0.08 AV: 8 NL: 2.64E6 T: FTMS + p ESI Full ms [120.00-1000.00] 445.8990 100

90

80

70

60 443.9013 447.8966 50

40 448.9000 603.1266

Relative Abundance 207.0628 30 389.0818 581.1447 183.0262 20 367.0998 149.0120 605.1426 253.0707 283.0788 529.8511 325.0894 413.2661 505.0900 10 531.8486 928.7365 619.1005 954.7017 665.0968 742.4948 803.5421 831.5735 890.7913 0 481 150 200 250 300 350 400 450 500 550 600 650 700 750 800 850 900 950 1000 m/z

445.89903 NL: 100 2.64E6 80 ENPVI13_Orbi_+ ESI#2-9 RT: 0.01-0.08 AV: 8 T: FTMS + p ESI 60 447.89663 443.90132 Full ms 40 [120.00-1000.00]

20 446.90217 Relative Abundance Relative 444.90460 448.90000 451.05190 439.08364 441.29750 443.17712 453.09870 454.10258 455.31273 457.27079 0 445.89910 NL: 100 4.01E5

80 C14 H11Br2 N3 O2 S+H: C14 H12 Br2 N3 O2 S1 60 pa Chrg 1 443.90115 447.89706 40

20 446.90246 444.90450 448.90041 449.89285 450.89621 452.89542 453.89878 454.89967 0 438 439 440 441 442 443 444 445 446 447 448 449 450 451 452 453 454 455 456 457 458 m/z

HPLC Trace of Compound 22

482

HPLC Trace of Compound 22

483

HPLC Trace of Compound 22

484

HPLC Trace of Compound 22 (1 h in ACN/Water)

485

HPLC Trace of Compound 22 (1 h in ACN/Water)

486

HPLC Trace of Compound 22 (1 h in ACN/Water)

487

1 H-NMR (DMSO-d6, 500 MHz) of Compound 23 488

13 C-NMR (DMSO-d6, 125 MHz) Compound 23 489

1 H-NMR (DMSO-d6, 500 MHz) of Compound 24 490

13 C-NMR (DMSO-d6, 125 MHz) Compound 24 491

31P-NMR (DMSO, 85 % phosphoric acid as external standard, decoupled, 243 MHz) of Compound 24 492

1 H-NMR (DMSO-d6, 600 MHz) of Compound 25 493

13 C-NMR (DMSO-d6, 150 MHz) Compound of 25 494

31P-NMR (DMSO, 85 % phosphoric acid as external standard, decoupled, 243 MHz) of Compound 25 495

1 H-NMR (D2O, 600 MHz) of Compound 27 496

13 C-NMR (D2O, 150 MHz) Compound of 27 497

31 P-NMR (D2O, 85 % phosphoric acid as external standard, decoupled, 243 MHz) of Compound 27 498

Mass Spec of Compound 27

C:\Xcalibur\...\ENP_VI_53C4_Orbi_+ESI 5/7/2015 10:47:37 PM

ENP_VI_53C4_Orbi_+ESI #1-11 RT: 0.00-0.09 AV: 11 NL: 1.47E7 T: FTMS + p ESI Full ms [100.00-1000.00] 475.9236 100

90

80

70

60

50

40

Relative Abundance 30

20 413.2662 476.9265 451.9438 10 218.9793 495.9074 249.9614 311.2945 537.3942 124.9373 156.9635 349.9957 398.9242 573.4330 617.4593 661.4848 696.6492 749.5374 793.5640 859.5377 0

499 150 200 250 300 350 400 450 500 550 600 650 700 750 800 850 m/z

473.92602 475.92360 NL: 100 1.47E7 80 ENP_VI_53C4_Orbi_+ ESI#1-11 RT: 0.00-0.09 60 AV: 11 T: FTMS + p ESI Full ms [100.00-1000.00] 40

20 474.92903 476.92651 Relative Abundance 470.33251471.36545 473.34542 477.91795 479.40629 481.31285483.34282 485.38072 487.36053 489.42776 0 NL: 473.92594 475.92389 100 4.05E5

80 C14 H11BrN3 Na 2 O4 PS +H: C14 H12 Br1N3 Na 2 O4 P1S1 60 pa Chrg 1

40

20 474.92930 476.92725 477.91969 478.92305 480.92729 481.93065 482.92651 0 470 471 472 473 474 475 476 477 478 479 480 481 482 483 484 485 486 487 488 489 m/z

Mass Spec of Compound 27

C:\Xcalibur\...\ENP_VI_53C4_Orbi_+ESI 5/7/2015 10:47:37 PM

ENP_VI_53C4_Orbi_+ESI #1-11 RT: 0.00-0.09 AV: 11 NL: 1.47E7 T: FTMS + p ESI Full ms [100.00-1000.00] 475.9236 100

90

80

70

60

50

40

Relative Abundance 30

20 413.2662 476.9265 451.9438 10 218.9793 495.9074 249.9614 311.2945 537.3942 124.9373 156.9635 349.9957 398.9242 573.4330 617.4593 661.4848 696.6492 749.5374 793.5640 859.5377 0

500 100 150 200 250 300 350 400 450 500 550 600 650 700 750 800 850 m/z

495.90744 497.90525 NL: 100 1.01E6 80 ENP_VI_53C4_Orbi_+ ESI#1-11 RT: 0.00-0.09 AV: 60 11 T: FTMS + p ESI Full ms [100.00-1000.00] 40 497.35616 20 496.91112 498.90883 504.01706 Relative Abundance 502.01921 501.37578 494.42584 495.32891 496.33422 498.35842 499.89997 503.36829 505.42212 506.42535 507.43807 0 495.90789 497.90584 NL: 100 4.05E5

80 C14 H11BrN3 Na 2 O4 PS +Na: C14 H11Br1N3 Na 3 O4 P1S1 60 pa Chrg 1

40

20 496.91124 498.90919 499.90164 500.90499 501.90835 502.90924 503.91259 504.90845 505.91181 0 494 495 496 497 498 499 500 501 502 503 504 505 506 507 m/z

HPLC Trace of Compound 27

501

HPLC Trace of Compound 27

502

HPLC Trace of Compound 27

503

APPENDIX C

Design and Synthesis of Cathepsin Inhibitors: Symmetrical Thiosemicarbazones

Table of Contents

1 H NMR (500 MHz, CDCl3) of Compound 1 ...... 512

13 C NMR (125 MHz, CDCl3) of Compound 1 ...... 513

1 H NMR (600 MHz, CDCl3) of Compound 2 ...... 514

13 C NMR (150 MHz, CDCl3) of Compound 2 ...... 515

1 H NMR (500 MHz, CDCl3) of Compound 3 ...... 516

13 C NMR (125 MHz, CDCl3) of Compound 3 ...... 517

19 F NMR (470 MHz, CDCl3) of Compound 3 ...... 518

1 H NMR (500 MHz, CDCl3) of Compound 4 ...... 519

13 C NMR (125 MHz, CDCl3) of Compound 4 ...... 520

1 H NMR (600 MHz, CDCl3) of Compound 5 ...... 521

13 C NMR (150 MHz, CDCl3) of Compound 5 ...... 522

1 H NMR (500 MHz, DMSO-d6) of Compound 6 ...... 523

13 C NMR (125 MHz, DMSO-d6) of Compound 6 ...... 524

1 H NMR (600 MHz, CDCl3) of Compound 7 ...... 525

13 C NMR (150 MHz, CDCl3) of Compound 7 ...... 526

504

19 F NMR (565 MHz, CDCl3) of Compound 7 ...... 527

1 H NMR (500 MHz, DMSO-d6) of Compound 8 ...... 528

13 C NMR (125 MHz, DMSO-d6) of Compound 8 ...... 529

HPLC Trace of Compound 8 ...... 530

HPLC Trace of Compound 8 ...... 531

HPLC Trace of Compound 8 ...... 532

1 H NMR (500 MHz, DMSO-d6) of Compound 9 ...... 533

13 C NMR (125 MHz, DMSO-d6) of Compound 9 ...... 534

Mass Spec of Compound 9 ...... 535

HPLC Trace of Compound 9 ...... 536

HPLC Trace of Compound 9 ...... 537

HPLC Trace of Compound 9 ...... 538

1 H NMR (500 MHz, acetone-d6) of Compound 10 ...... 539

13 C NMR (125 MHz, acetone-d6) of Compound 10 ...... 540

19 F NMR (470 MHz, CDCl3) of Compound 10 ...... 541

Mass Spec of Compound 10 ...... 542

HPLC Trace of Compound 10 ...... 543

HPLC Trace of Compound 10 ...... 544

HPLC Trace of Compound 10 ...... 545

1 H NMR (500 MHz, acetone-d6) of Compound 11 ...... 546

505

13 C NMR (125 MHz, acetone-d6) of Compound 11 ...... 547

HPLC Trace of Compound 11 ...... 548

HPLC Trace of Compound 11 ...... 549

HPLC Trace of Compound 11 ...... 550

1 H NMR (500 MHz, acetone-d6) of Compound 12 ...... 551

13 C NMR (125 MHz, acetone-d6) of Compound 12 ...... 552

Mass Spec of Compound 12 ...... 553

HPLC Trace of Compound 12 ...... 554

HPLC Trace of Compound 12 ...... 555

HPLC Trace of Compound 12 ...... 556

1 H NMR (500 MHz, CDCl3) of Compound 13 ...... 557

13 C NMR (125 MHz, CDCl3) of Compound 13 ...... 558

1 H NMR (600 MHz, CDCl3) of Compound 14 ...... 559

13 C NMR (150 MHz, CDCl3) of Compound 14 ...... 560

1 H NMR (500 MHz, acetone-d6) of Compound 15 ...... 561

13 C NMR (125 MHz, acetone-d6) of Compound 15 ...... 562

Mass Spec of Compound 15 ...... 563

HPLC Trace of Compound 15 ...... 564

HPLC Trace of Compound 15 ...... 565

HPLC Trace of Compound 15 ...... 566

506

1 H NMR (500 MHz, CDCl3) of Compound 16 ...... 567

13 C NMR (125 MHz, CDCl3) of Compound 16 ...... 568

1 H NMR (500 MHz, CDCl3) of Compound 17 ...... 569

13 C NMR (125 MHz, CDCl3) of Compound 17 ...... 570

1 H NMR (500 MHz, CDCl3) of Compound 18 ...... 571

13 C NMR (125 MHz, CDCl3) of Compound 18 ...... 572

1 H NMR (500 MHz, acetone-d6) of Compound 19 ...... 573

13 C NMR (150 MHz, acetone-d6) of Compound 19 ...... 574

1 H NMR (600 MHz, CDCl3) of Compound 20 ...... 575

13 C NMR (150 MHz, CDCl3) of Compound 20 ...... 576

19 F NMR (565 MHz, CDCl3) of Compound 20 ...... 577

1 H NMR (500 MHz, CDCl3) of Compound 21 ...... 578

13 C NMR (125 MHz, CDCl3) of Compound 21 ...... 579

19 F NMR (470 MHz, CDCl3) of Compound 21 ...... 580

1 H NMR (500 MHz, acetone-d6) of Compound 22 ...... 581

13 C NMR (125 MHz, acetone-d6) of Compound 22 ...... 582

1 H NMR (500 MHz, acetone-d6) of Compound 23 ...... 583

13 C NMR (125 MHz, acetone-d6) of Compound 23 ...... 584

1 H NMR (500 MHz, CDCl3) of Compound 24 ...... 585

13 C NMR (125 MHz, CDCl3) of Compound 24 ...... 586

507

19 F NMR (470 MHz, CDCl3) of Compound 24 ...... 587

1 H NMR (500 MHz, CDCl3) of Compound 25 ...... 588

13 C NMR (125 MHz, CDCl3) of Compound 25 ...... 589

19 F NMR (565 MHz, CDCl3) of Compound 25 ...... 590

1 H NMR (500 MHz, DMSO-d6) of Compound 26 ...... 591

13 C NMR (125 MHz, DMSO-d6) of Compound 26 ...... 592

Mass Spectra of Compound 26 ...... 593

HPLC Trace of Compound 26 ...... 594

HPLC Trace of Compound 26 ...... 595

HPLC Trace of Compound 26 ...... 596

1 H NMR (600 MHz, DMSO-d6) of Compound 27 ...... 597

13 C NMR (150 MHz, DMSO-d6) Compound 27 ...... 598

Mass Spectra of Compound 27 ...... 599

HPLC Trace of Compound 27 ...... 600

HPLC Trace of Compound 27 ...... 601

HPLC Trace of Compound 27 ...... 602

1 H NMR (600 MHz, DMSO-d6) of Compound 28 ...... 603

13 C NMR (150 MHz, DMSO-d6) Compound 28 ...... 604

Mass Spectra of Compound 28 ...... 605

HPLC Trace of Compound 28 ...... 606

508

HPLC Trace of Compound 28 ...... 607

HPLC Trace of Compound 28 ...... 608

1 H NMR (500 MHz, DMSO-d6) of Compound 29 ...... 609

13 C NMR (125 MHz, DMSO-d6) Compound 29 ...... 610

Mass Spectra of Compound 29 ...... 611

HPLC Trace of Compound 29 ...... 613

HPLC Trace of Compound 29 ...... 614

HPLC Trace of Compound 29 ...... 615

1 H NMR (500 MHz, acetone-d6) of Compound 30 ...... 616

13 C NMR (125 MHz, acetone-d6) of Compound 30 ...... 617

19 F NMR (470 MHz, acetone-d6) of Compound 30 ...... 618

Mass Spectra of Compound 30 ...... 619

HPLC Trace of Compound 30 ...... 620

HPLC Trace of Compound 30 ...... 621

HPLC Trace of Compound 30 ...... 622

1 H NMR (500 MHz, DMSO-d6) of Compound 31 ...... 623

13 C NMR (125 MHz, DMSO-d6) of Compound 31 ...... 624

19 F NMR (470 MHz, DMSO-d6) of Compound 31 ...... 625

Mass Spectra of Compound 31 ...... 626

HPLC Trace of Compound 31 ...... 627

509

HPLC Trace of Compound 31 ...... 628

HPLC Trace of Compound 31 ...... 629

HPLC Trace of Compound 31 ...... 630

1 H NMR (500 MHz, DMSO-d6) of Compound 32 ...... 631

13 C NMR (125 MHz, DMSO-d6) of Compound 32 ...... 632

HPLC Trace of Compound 32 ...... 633

HPLC Trace of Compound 32 ...... 634

HPLC Trace of Compound 32 ...... 635

1 H NMR (500 MHz, DMSO-d6) of Compound 33 ...... 636

13 C NMR (150 MHz, DMSO-d6) of Compound 33 ...... 637

Mass Spectra of Compound 33 ...... 638

HPLC Trace of Compound 33 ...... 639

HPLC Trace of Compound 33 ...... 640

HPLC Trace of Compound 33 ...... 641

1 H NMR (600 MHz, DMSO-d6) of Compound 34 ...... 642

13 C NMR (150 MHz, DMSO-d6) of Compound 34 ...... 643

19 F NMR (565 MHz, DMSO-d6) of Compound 34 ...... 644

Mass Spectra of Compound 34 ...... 645

HPLC Trace HPLC Trace of Compound 34 ...... 646

HPLC Trace of Compound 34 ...... 647

510

HPLC Trace of Compound 34 ...... 648

1 H NMR (500 MHz, DMSO-d6) of Compound 35 ...... 649

13 C NMR (125 MHz, DMSO-d6) Compound 35 ...... 650

19 F NMR (470 MHz, DMSO-d6) of Compound 35 ...... 651

Mass Spectra of Compound 35 ...... 652

Mass Spectra of Compound 35 ...... 653

Mass Spectra of Compound 35 ...... 654

HPLC Trace of Compound 35 ...... 655

HPLC Trace of Compound 35 ...... 656

HPLC Trace of Compound 35 ...... 657

1 H NMR (500 MHz, acetone-d6) of Compound 36 ...... 658

13 C NMR (125 MHz, acetone-d6) of Compound 36 ...... 659

19 F NMR (470 MHz, acetone-d6) of Compound 36 ...... 660

1 H NMR (500 MHz, acetone-d6) of Compound 37 ...... 661

13 C NMR (125 MHz, acetone-d6) of Compound 37 ...... 662

19 F NMR (470 MHz, acetone-d6) δ of Compound 37 ...... 663

Mass Spectra of Compound 37 ...... 664

HPLC Trace of Compound 37 ...... 665

511

1 H NMR (500 MHz, CDCl3) of Compound 1 512

13 C NMR (125 MHz, CDCl3) of Compound 1 513

1 H NMR (600 MHz, CDCl3) of Compound 2 514

13 C NMR (150 MHz, CDCl3) of Compound 2 515

1 H NMR (500 MHz, CDCl3) of Compound 3 516

13 C NMR (125 MHz, CDCl3) of Compound 3 517

19 F NMR (470 MHz, CDCl3) of Compound 3 518

1 H NMR (500 MHz, CDCl3) of Compound 4 519

13 C NMR (125 MHz, CDCl3) of Compound 4 520

1 H NMR (600 MHz, CDCl3) of Compound 5 521

13 C NMR (150 MHz, CDCl3) of Compound 5 522

1 H NMR (600 MHz, acetone-d6) of Compound 6 523

13 C NMR (150 MHz, acetone-d6) of Compound 6 524

1 H NMR (600 MHz, CDCl3) of Compound 7 525

13 C NMR (150 MHz, CDCl3) of Compound 7 526

19 F NMR (565 MHz, CDCl3) of Compound 7 527

1 H NMR (500 MHz, DMSO-d6) of Compound 8 528

13 C NMR (125 MHz, DMSO-d6) of Compound 8 529

HPLC Trace of Compound 8

530

HPLC Trace of Compound 8

531

HPLC Trace of Compound 8

532

1 H NMR (500 MHz, DMSO-d6) of Compound 9 533

13 C NMR (125 MHz, DMSO-d6) of Compound 9 534

Mass Spec of Compound 9

C:\Xcalibur\...\ENP_IV_57_+ESI_Orbi 7/22/2013 10:07:58 PM ENP_IV_57

ENP_IV_57_+ESI_Orbi #1 RT: 0.00 AV: 1 NL: 1.05E8 T: FTMS + p ESI Full ms [150.00-2000.00] 288.08020 100 310.06216 90 80 70 60 50 40 597.13483 Relative Abundance 30

20 323.14639 613.09912 10 413.26611 725.40179 212.07048 573.13715449.35986 884.20752 1025.734501171.28064 1315.81702 1458.85681 1603.891851745.42932 1883.71631 0 535 200 300 400 500 600 700 800 900 1000 1100 1200 1300 1400 1500 1600 1700 1800 1900 2000 m/z

288.08020 NL: 100 310.06216 1.05E8 ENP_IV_57_+ESI_Orbi#1 50 RT: 0.00 AV: 1 T: FTMS 311.06537 + p ESI Full ms 286.06464289.08334 293.20859 295.18802 301.14102297.23987304.07501 309.20389 307.22452 313.14313 318.09055316.11127 321.23993 0 [150.00-2000.00] 288.08012 100 NL: 8.02E514 H13 N3 O2 S+H: 50 14CH14 N3 O2 S 1 paC Chrg 1 289.08348 291.07927 293.07849 296.08609 0 Relative Abundance 310.06207 100 NL: 8.02E5 14 H13 N3 O2 S+Na: 50 C14CH13 N3 O2 S 1Na 1 pa Chrg 1 311.06542 313.06122 315.06043 317.07464 0 286 288 290 292 294 296 298 300 302 304 306 308 310 312 314 316 318 320 m/z

HPLC Trace of Compound 9

536

HPLC Trace of Compound 9

537

HPLC Trace of Compound 9

538

1 H NMR (500 MHz, acetone-d6) of Compound 10 539

13 C NMR (125 MHz, acetone-d6) of Compound 10 540

19 F NMR (470 MHz, CDCl3) of Compound 10 541

Mass Spec of Compound 10

C:\Xcalibur\...\ENP_IV_31_+ESI_Orbi 7/22/2013 9:04:59 PM ENP_IV_31

ENP_IV_31_+ESI_Orbi #1-15 RT: 0.01-0.11 AV: 15 NL: 3.24E8 T: FTMS + p ESI Full ms [100.00-2000.00] 292.0717 100 294.0670 90 314.0535 80 70 60 50 40

Relative Abundance 30 20 10 218.0775 275.0450 324.0435 547.1322 116.9858 130.1589 157.0833 203.0666 260.0994 365.1359 376.0238 413.2662 441.2974456.5770 490.1649 513.3224 0 216.0619 315.0565 542 120 140 160 180 200 220 240 260 280 300 320 340 360 380 400 420 440 460 480 500 520 540 m/z

292.07166 NL: 100 314.05349 3.24E8 ENP_IV_31_+ESI_Orbi#1-15 50 RT: 0.01-0.11 AV: 15 T: 313.14367 308.06653 315.05653 FTMS + p ESI Full ms 295.07024 298.07568 301.14105303.14875 306.05082 311.04524 317.05220 319.26060322.08191 0 293.07468 [100.00-2000.00] 292.07145 100 NL: 8.06E514 H11 N3 F2 S+H: 50 14CH12 N3 F2 S 1 paC Chrg 1 293.07481 295.07060 297.06982 299.07653 0 Relative Abundance 314.05340 100 NL: 8.07E5 14 H11 N3 F2 S+Na: 50 C14CH11 N3 F2 S 1Na 1 pa Chrg 1 315.05675 317.05255 319.05176 321.05847 0 292 294 296 298 300 302 304 306 308 310 312 314 316 318 320 322 m/z

HPLC Trace of Compound 10

543

HPLC Trace of Compound 10

544

HPLC Trace of Compound 10

545

1 H NMR (500 MHz, acetone-d6) of Compound 11 546

13 C NMR (125 MHz, acetone-d6) of Compound 11 547

HPLC Trace of Compound 11

548

HPLC Trace of Compound 11

549

HPLC Trace of Compound 11

550

1 H NMR (500 MHz, acetone-d6) of Compound 12 551

13 C NMR (125 MHz, acetone-d6) of Compound 12 552

Mass Spec of Compound 12

C:\Xcalibur\...\ENP-III-83_orbi_+ESI 11/30/2012 12:18:17 AM ENP-III-83

ENP-III-83_orbi_+ESI #2-10 RT: 0.01-0.09 AV: 9 NL: 6.37E7 T: FTMS + p ESI Full ms [100.00-2000.00] 284.12288 306.10486 100 90 80 70 60 50 40

Relative Abundance 30 20 125.98671 346.03725 10 589.22035 149.01260 208.11289 267.09613 322.07867368.07546 537.39699 181.10180 226.95241 413.26788 441.29933469.33068 511.36290 0 553 120 140 160 180 200 220 240 260 280 300 320 340 360 380 400 420 440 460 480 500 520 540 560 580 600 620 m/z

284.12288 306.10486 NL: 100 6.37E7 ENP-III-83_orbi_+ESI#2-10 50 RT: 0.01-0.09 AV: 9 T: 283.11366 307.10794 FTMS + p ESI Full ms 280.11341 285.12593 289.07797 291.19464 295.12810 297.24132 301.14249 304.30149309.10351 312.15390 314.13329 0 [100.00-2000.00] 284.12159 100 NL: 7.89E516 H17 N3 S+H: 50 1C6 H18 N3 S 1 paC Chrg 1 285.12495 287.12075 289.11996 291.12667 0 Relative Abundance 306.10354 100 NL: 7.89E5 16 H17 N3 S+Na: 50 C1C6 H17 N3 S 1Na 1 pa Chrg 1 307.10689 309.10269 311.10190 313.10861 0 280 282 284 286 288 290 292 294 296 298 300 302 304 306 308 310 312 314 316 m/z

HPLC Trace of Compound 12

554

HPLC Trace of Compound 12

555

HPLC Trace of Compound 12

556

1 H NMR (500 MHz, CDCl3) of Compound 13 557

13 C NMR (125 MHz, CDCl3) of Compound 13 558

1 H NMR (600 MHz, CDCl3) of Compound 14 559

13 C NMR (150 MHz, CDCl3) of Compound 14 560

1 H NMR (500 MHz, acetone-d6) of Compound 15 561

13 C NMR (125 MHz, acetone-d6) of Compound 15 562

Mass Spec of Compound 15

C:\Xcalibur\...\ENP-III-61_orbi_+ESI 11/29/2012 11:16:59 PM ENP-III-61

ENP-III-61_orbi_+ESI #2-14 RT: 0.01-0.11 AV: 13 NL: 9.72E7 T: FTMS + p ESI Full ms [100.00-2000.00] 356.12176 100 90 80 280.11216 70 60 50 40

Relative Abundance 30 318.96012324.14965 337.10479 20 365.13587 388.09368 10 618.26544 144.05682 191.04426 163.07521233.09131295.12174 261.10978 413.26620 454.19483488.15577507.32949 550.62852 595.38136 645.27630 0 563 150 200 250 300 350 400 450 500 550 600 650 m/z

NL: 9.72E7 356.12176 100 ENP-III-61_orbi_+ ESI#2-14 RT: 0.01-0.11 80 AV: 13 T: FTMS + p 60 ESI Full ms [100.00-2000.00] 40 356.08755 20

Relative Abundance 357.12480 355.77233 356.82290 357.16844 360.24374 361.23481 0 356.12159 358.11723 NL: 100 356.16602 359.12041 7.39E5 80 C 22 H17 N3 S+H: 22C H18 N3 S 1 60 pa Chrg 1 40

20 357.12495 0 355.5 356.0 356.5 357.0 357.5 358.11739358.0 358.5359.12075 359.0 360.12410359.5 360.0 361.12746360.5 361.0 361.5 m/z

HPLC Trace of Compound 15

564

HPLC Trace of Compound 15

565

HPLC Trace of Compound 15

566

1 H NMR (500 MHz, CDCl3) of Compound 16 567

13 C NMR (125 MHz, CDCl3) of Compound 16 568

1 H NMR (500 MHz, CDCl3) of Compound 17 569

13 C NMR (125 MHz, CDCl3) of Compound 17 570

1 H NMR (500 MHz, CDCl3) of Compound 18 571

13 C NMR (125 MHz, CDCl3) of Compound 18 572

1 H NMR (600 MHz, acetone-d6) of Compound 19 573

13 C NMR (150 MHz, acetone-d6) of Compound 19 574

1 H NMR (600 MHz, CDCl3) of Compound 20 575

13 C NMR (150 MHz, CDCl3) of Compound 20 576

19 F NMR (565 MHz, CDCl3) of Compound 20 577

1 H NMR (500 MHz, CDCl3) of Compound 21 578

13 C NMR (125 MHz, CDCl3) of Compound 21 579

19 F NMR (470 MHz, CDCl3) of Compound 21 580

1 H NMR (500 MHz, acetone-d6) of Compound 22 581

13 C NMR (125 MHz, acetone-d6) of Compound 22 582

1 H NMR (500 MHz, acetone-d6) of Compound 23 583

13 C NMR (125 MHz, acetone-d6) of Compound 23 584

1 H NMR (500 MHz, CDCl3) of Compound 24 585

13 C NMR (125 MHz, CDCl3) of Compound 24 586

19 F NMR (470 MHz, CDCl3) of Compound 24 587

1 H NMR (500 MHz, CDCl3) of Compound 25 588

13 C NMR (125 MHz, CDCl3) of Compound 25 589

19 F NMR (565 MHz, CDCl3) of Compound 25 590

1 H NMR (500 MHz, DMSO-d6) of Compound 26 591

13 C NMR (125 MHz, DMSO-d6) of Compound 26 592

Mass Spectra of Compound 26

C:\Xcalibur\...\ENPIV62_Orbi_+ESI 7/7/2014 9:42:45 PM

ENPIV62_Orbi_+ESI #12 RT: 0.09 AV: 1 NL: 8.71E6 T: FTMS + p ESI Full ms [150.00-2000.00] 330.10376 100 z=1 90 80 308.12180 70 z=1 60 50 637.21790 z=1 40 392.07397 232.11215 z=1 Relative Abundance 30 z=1 20 480.65250 615.23584 788.26740 944.41864 z=2 1251.44714 1406.00305 1559.56067 1764.304931867.67578 10 z=1 677.14911 z=2 z=1 1095.38037 z=2 z=? z=2 z=? z=3 z=3 0 z=1 593 200 300 400 500 600 700 800 900 1000 1100 1200 1300 1400 1500 1600 1700 1800 1900 2000 m/z

330.10376 NL: 8.71E6 100 308.12180 z=1 z=1 329.01971 ENPIV62_Orbi_+ESI#12 50 309.12500 z=? RT: 0.09 AV: 1 T: FTMS 306.10669 311.25519315.19299 322.13770 324.11649 327.09540 333.10257331.10696 335.10303 337.23483 z=1 + p ESI Full ms 0 z=? z=? z=? z=? z=? z=? z=? z=1 z=? z=? [150.00-2000.00] 308.12159 100 NL: 7.72E518 H17 N3 S+H: C18 H18 N3 S 1 50 paC Chrg 1 309.12495 311.12075 313.12746 315.12667 0

Relative Abundance NL: 330.10354 100 7.72E5 18 H17 N3 S+Na: CC18 H17 N3 S 1Na 1 50 pa Chrg 1 331.10689 333.10269 335.10940 337.10861 0 306 308 310 312 314 316 318 320 322 324 326 328 330 332 334 336 m/z

HPLC Trace of Compound 26

594

HPLC Trace of Compound 26

595

HPLC Trace of Compound 26

596

1 H NMR (600 MHz, DMSO-d6) of Compound 27 597

13 C NMR (150 MHz, DMSO-d6) Compound 27 598

Mass Spectra of Compound 27

C:\Xcalibur\...\ENP_IV_86_Orbi_+ESI 5/7/2014 8:47:05 PM

ENP_IV_86_Orbi_+ESI #13 RT: 0.11 AV: 1 NL: 1.02E7 T: FTMS + p ESI Full ms [150.00-2000.00] 465.9406 100 z=1 90 80 70 60 50 40

Relative Abundance 30 20 10 299.1103 719.0928 778.0764 1102.23801184.3168 1308.2202 1417.7855 1493.6945 1717.21921604.1896 1883.8346 z=? z=1 z=1 z=1 z=? z=? z=2 z=1 z=?z=? z=? 0 406.9577 930.8757 599 226.9515 200 300 400z=1 495.9509500 600 700 800 z=1 900 1000 1100 1200 1300 1400 1500 1600 1700 1800 1900 2000 z=? z=1 m/z NL: 1.02E7 465.94061 ENP_IV_86_Orbi_+ 100 z=1 ESI#13 RT: 0.11 AV: 1 T: FTMS + p 80 ESI Full ms 60 463.94299 467.93811 [150.00-2000.00] 40 z=? z=1 20 462.88498 463.67322 464.11853 465.76471 466.94363 467.76123 468.11563 Relative Abundance z=? z=? z=? 464.94617 z=? z=1 z=? z=?468.94119 0 z=? 466.11758 z=1 469.93341 470.93738 NL: 465.94057z=? z=1 z=1 3.86E5 100 C 18 H15 Br 2 N3 S+H: 80 18CH16 Br 2 N3 S 1 pa Chrg 1 60 463.94262 40 467.93853 20 466.94393 0 464.94597 468.94188 462 463 464 465 466 467 468 469469.93432 470.93768470 471 m/z

HPLC Trace of Compound 27

600

HPLC Trace of Compound 27

601

HPLC Trace of Compound 27

602

1 H NMR (600 MHz, DMSO-d6) of Compound 28 603

13 C NMR (150 MHz, DMSO-d6) Compound 28 604

Mass Spectra of Compound 28

C:\Xcalibur\...\ENP_V_84_run3_Orbi_+ESI 7/30/2014 11:08:27 PM

ENP_V_84_run3_Orbi_+ESI #1-12 RT: 0.01-0.10 AV: 12 NL: 5.03E6 T: FTMS + p ESI Full ms [150.00-2000.00] 376.04376 100 90 80 70 378.04054 60 50 40

Relative Abundance 30 344.07163 398.02545 20 330.89427 349.99563 262.90685 406.05403 10 300.03408 164.92967 232.91743 702.04806 421.23236 480.84325 502.82621 524.79514550.62807 600.80463 622.78683 659.28654 0 605 200 250 300 350 400 450 500 550 600 650 700 m/z

376.04376 NL: 100 5.03E6 378.04054 ENP_V_84_run3_Orbi_+ 50 373.97571 ESI#1-12 RT: 0.01-0.10 AV: 379.04373 406.05403 12 T: FTMS + p ESI Full ms 371.97772 382.86477 386.02784 388.02467 392.03820 398.02545394.03533 413.26601408.05104 0 400.02242 [150.00-2000.00] 376.04365 100 NL: 378.04070 4.43E518 H15 Cl 2 N3 S+H: 50 18CH16 Cl 2 N3 S 1 paC Chrg 1 382.03355 385.04361 387.04282 0 379.04405 Relative Abundance 398.02559 100 NL: 4.43E5 400.02264 18 H15 Cl 2 N3 S+Na: 50 C18CH15 Cl 2 N3 S 1Na 1 pa Chrg 1 404.01549 407.02556 409.02477 0 401.02600 370 375 380 385 390 395 400 405 410 415 m/z

HPLC Trace of Compound 28

606

HPLC Trace of Compound 28

607

HPLC Trace of Compound 28

608

1 H NMR (500 MHz, DMSO-d6) of Compound 29 609

13 C NMR (125 MHz, DMSO-d6) Compound 29 610

Mass Spectra of Compound 29

C:\Xcalibur\...\ENP_IV_36_run2_+ESI_Orbi 7/23/2013 8:05:08 PM ENP_IV_36_run2 Did not lock in previous run ENP_IV_36_run2_+ESI_Orbi #2-14 RT: 0.01-0.11 AV: 13 NL: 5.31E7 T: FTMS + p ESI Full ms [150.00-2000.00] 340.11175 100 342.10739 90 80 70 60 50 40

Relative Abundance 30 362.09368 20 301.14125 10 279.11298 323.08512 413.26640 164.01630 225.10983 241.17747 308.13957 441.29759 513.32257463.37590 528.63731 606.20618 586.24527 643.21240 0 611 160 180 200 220 240 260 266.11770280 300 320 340 360370.12215380 400 420 440 460 480 500 520 540 560 580 600 620 640 m/z

NL: 5.31E7 340.11175 100 ENP_IV_36_run2_+ ESI_Orbi#2-14 RT: 80 0.01-0.11 AV: 13 T: 60 340.00217 FTMS + p ESI Full ms [150.00-2000.00] 40 20 Relative Abundance 340.22106339.36261341.11493 344.11455 345.06678 346.12866 348.29564 0 340.11142 342.10739 NL: 100 341.26605 343.11058 347.29215 7.68E5 80 C 18 H17 N3 O2 S+H: 18CH18 N3 O2 S 1 60 pa Chrg 1 40 20 341.11478 0 340 341 342.10722342 343.11057 343 344.11393344 345.11728 346.11314345 347.11650346 348.11739347 348 m/z

Mass Spectra of Compound 29

C:\Xcalibur\...\ENP_IV_36_run2_+ESI_Orbi 7/23/2013 8:05:08 PM ENP_IV_36_run2 Did not lock in previous run ENP_IV_36_run2_+ESI_Orbi #2-14 RT: 0.01-0.11 AV: 13 NL: 5.31E7 T: FTMS + p ESI Full ms [150.00-2000.00] 340.11175 100 343.11058 90 80 70 60 50 40

Relative Abundance 30 362.09368 20 301.14125 10 279.11298 323.08512 413.26640 164.01630 225.10983 241.17747308.13957 463.37590441.29759 513.32257528.63731 586.24527 606.20618 643.21240 0 612 160 180 200 220 240 260 266.11770280 300 320 340 360370.12215380 400 420 440 460 480 500 520 540 560 580 600 620 640 m/z

NL: 1.85E7 362.09368 100 ENP_IV_36_run2_+ ESI_Orbi#2-14 RT: 80 0.01-0.11 AV: 13 T: 60 FTMS + p ESI Full ms [150.00-2000.00] 40 361.97401 20 Relative Abundance 360.14474 361.30796363.09697 363.28699362.31168 365.13606 367.24550368.42521 369.29780 0 362.09337 NL: 364.08939 365.26635 366.13937 100 7.68E5 80 C 18 H17 N3 O2 S+Na: 18CH17 N3 O2 S 1Na 1 60 pa Chrg 1 40 20 363.09672 0 360 361 362 363 364.08916364 365.09252 365 366.09587366 367.09923 368.09509367 369.09844368 369 370 m/z

HPLC Trace of Compound 29

613

HPLC Trace of Compound 29

614

HPLC Trace of Compound 29

615

1 H NMR (500 MHz, acetone-d6) of Compound 30 616

13 C NMR (125 MHz, acetone-d6) of Compound 30 617

19 F NMR (470 MHz, acetone-d6) of Compound 30 618

Mass Spectra of Compound 30

C:\Xcalibur\...\ENP-III-100_orbi_+ESI 11/30/2012 2:52:28 AM ENP-III-100

ENP-III-100_orbi_+ESI #1-11 RT: 0.00-0.09 AV: 11 NL: 2.61E7 T: FTMS + p ESI Full ms [100.00-2000.00] 344.10307 100 90 80 70 60 50 40

Relative Abundance 30 20 116.98596 268.09336 10 166.01209 413.26632 537.39489 749.11224 685.18296 1092.20842 865.62299 953.67505 1173.80666 1395.372541556.80021 1739.46158 1883.72338 0 366.08484 709.18019 619 200 300 400 500 600 700 800 900 1000 1100 1200 1300 1400 1500 1600 1700 1800 1900 2000 m/z

344.10307 NL: 100 2.61E7 ENP-III-100_orbi_+ESI#1-11 50 RT: 0.00-0.09 AV: 11 T: 366.08484 FTMS + p ESI Full ms 345.10608 347.10164 352.07142353.26622359.24048 362.92653 368.42517 370.29513 374.11336 0 342.08773 356.12174 [100.00-2000.00] 344.10275 100 NL: 7.72E518 H15 F2 N3 S+H: 50 18CH16 F2 N3 S 1 paC Chrg 1 345.10611 347.10190 349.10861 351.10783 0 Relative Abundance 366.08470 100 NL: 7.72E5 18 H15 F2 N3 S+Na: 50 C18CH15 F2 N3 S 1Na 1 pa Chrg 1 367.08805 369.08385 371.09056 373.08977 0 342 344 346 348 350 352 354 356 358 360 362 364 366 368 370 372 374 m/z

HPLC Trace of Compound 30

620

HPLC Trace of Compound 30

621

HPLC Trace of Compound 30

622

1 H NMR (500 MHz, DMSO-d6) of Compound 31 623

13 C NMR (125 MHz, DMSO-d6) of Compound 31 624

19 F NMR (470 MHz, DMSO-d6) of Compound 31 625

Mass Spectra of Compound 31

C:\Xcalibur\...\ENP_IV_20_+ESI_Orbi 7/23/2013 8:53:01 PM ENP_IV_20

ENP_IV_20_+ESI_Orbi #13 RT: 0.10 AV: 1 NL: 1.05E8 T: FTMS + p ESI Full ms [150.00-2000.00] 344.1027 100 90 80 70 60 50 40

Relative Abundance 30 20 685.1818 10 268.0931 413.2659 241.1772 513.3220 651.1946 709.1793 803.5428 1006.5481 909.65301092.2091 1173.8121 1382.1987 1453.3672 1603.3324 1754.3010 1883.8427 0 360.0975 626 200 300 400 500 600 700 800 900 1000 1100 1200 1300 1400 1500 1600 1700 1800 1900 2000 m/z

NL: 1.05E8 344.1027 100 ENP_IV_20_+ESI_Orbi#13 RT: 0.10 AV: 1 T: FTMS + p 80 ESI Full ms [150.00-2000.00] 60 40 20 Relative Abundance 343.9926 344.2148 345.1057 344.6357345.2380 348.2941 0 344.1028 346.0981 NL: 100 343.0899 347.1014 349.2708 1.81E4 80 C 18 H15 F2 N3 S+H: 18CH16 F2 N3 S 1 60 p (gss, s /p:40) Chrg 1 R: 30000 Res .Pwr . @FWHM 40 20 345.1057 0 346.1018 343.0 343.5 344.0 344.5 345.0 345.5 346.0 346.5347.1029 347.0 348.1039347.5 348.0 349.1050 348.5 349.0 349.5 m/z

HPLC Trace of Compound 31

627

HPLC Trace of Compound 31

628

HPLC Trace of Compound 31

629

HPLC Trace of Compound 31

630

1 H NMR (500 MHz, DMSO-d6) of Compound 32 631

13 C NMR (125 MHz, DMSO-d6) of Compound 32 632

HPLC Trace of Compound 32

633

HPLC Trace of Compound 32

634

HPLC Trace of Compound 32

635

1 H NMR (500 MHz, DMSO-d6) of Compound 33 636

13 C NMR (150 MHz, DMSO-d6) of Compound 33 637

Mass Spectra of Compound 33

C:\Xcalibur\...\ENP_IV_22_+ESI_Orbi 7/22/2013 10:35:37 PM ENP_IV_22

ENP_IV_22_+ESI_Orbi #15 RT: 0.12 AV: 1 NL: 1.48E8 T: FTMS + p ESI Full ms [150.00-2000.00] 344.1428 100 90 80 70 709.2594 60 50 366.1246 40

Relative Abundance 30 20 270.1487 687.2775 1052.3944 10 413.2659 725.2335 241.1773 488.2001 613.2839831.5762 1030.41281074.3763 1221.4557 1395.5311 1564.59141739.6694 1883.7814 0 638 200 300 400 500 600 700 800 900 1000 1100 1200 1300 1400 1500 1600 1700 1800 1900 2000 m/z

NL: 7.63E7 366.1246 100 ENP_IV_22_+ESI_Orbi#15 RT: 0.12 AV: 1 T: FTMS + p 80 ESI Full ms [150.00-2000.00] 60 40 20 Relative Abundance 366.0023 366.2474 367.1278 372.1928374.1530 373.3065 0 366.1247 368.1203 NL: 100 367.2451 369.1235 371.3155 1.80E4 80 C 18 H21 N3 O2 S+Na: 18CH21 N3 O2 S 1Na 1 60 p (gss, s /p:40) Chrg 1 R: 30000 Res .Pwr . @FWHM 40 20 367.1276 0 368.1241 366.0 366.5 367.0 367.5 368.0 368.5369.1254 369.0 370.1263369.5 370.0 371.1276370.5 371.0 372.1292371.5 373.1309372.0 372.5 374.1328373.0 373.5 374.0 374.5 m/z

HPLC Trace of Compound 33

639

HPLC Trace of Compound 33

640

HPLC Trace of Compound 33

641

1 H NMR (600 MHz, DMSO-d6) of Compound 34 642

13 C NMR (150 MHz, DMSO-d6) of Compound 34 643

19 F NMR (565 MHz, DMSO-d6) of Compound 34 644

Mass Spectra of Compound 34

C:\Xcalibur\...\ENP-III-99_orbi_negESI 11/30/2012 1:31:01 AM ENP-III-99

ENP-III-99_orbi_negESI #13 RT: 0.11 AV: 1 NL: 5.76E7 T: FTMS - p ESI Full ms [100.00-2000.00] 346.1195 100

80

60

40 Relative Abundance Relative 20 715.2271 204.0880 297.1528 392.1245 444.0862 602.3587 672.3105 791.1354 881.18211004.3687 1102.2859 1160.3215 1263.9237 1334.3945 1508.44901660.1316 1736.6204 1824.3275 1882.8542 0 312.1317 693.2457 100 200 300 400 500 600 700 800 900 1000 1100 1200 1300 1400 1500 1600 1700 1800 1900 2000 m/z NL: 5.76E7 346.1195 100 ENP-III- 99_orbi_negESI#13 RT: 0.11 AV: 1 T: 50 FTMS - p ESI Ful l ms

645 [100.00-2000.00] 346.2008 346.6816345.5604347.1213 347.2114 350.1198350.8903 351.2193 353.1987 Relative Abundance Relative 0 NL: 346.1195 348.1146 100 346.0340 349.1166 7.72E5 H N SF +H: C 18 19 3 2 18CH18 N3 S 1 F2 50 pa Chrg -1

347.1229 0 348.1153 344.5 345.0 345.5 346.0 346.5 347.0 347.5 348.0 348.5349.1186349.0 350.1220349.5 350.0 351.1254350.5 351.0 352.1212351.5 353.1246352.0 352.5 353.0 353.5 m/z NL: 7.18E6 693.2457 100 ENP-III-99_orbi_negESI#13 RT: 0.11 AV: 1 T: FTMS - p ESI Full 50 694.2485 ms [100.00-2000.00] 715.2271 700.4677 702.4114 714.4826 719.2300 NL: 0 695.2440 692.2341693.2463 697.2480 705.2086709.2403 711.2416 717.2240 725.2349 727.2275 728.4992 5.96E5 100 C 50 36 H38 N6 S 2 F 4 +H: 694.2496 36CH37 N6 S 2 F 4 pa Chrg -1 0 695.2421 697.2488 699.2446 701.2513 703.2505 715.2282 NL:

Relative Abundance Relative 100 5.96E5 36 H36 N6 S 2 F 4 Na: 50 C 716.2316 36CH36 N6 S 2 F 4 Na 1 717.2240 pa Chrg -1 0 719.2307 721.2265 723.2332 725.2324 692 694 696 698 700 702 704 706 708 710 712 714 716 718 720 722 724 726 728 730 m/z

HPLC Trace HPLC Trace of Compound 34

646

HPLC Trace of Compound 34

647

HPLC Trace of Compound 34

648

1 H NMR (500 MHz, DMSO-d6) of Compound 35 649

13 C NMR (125 MHz, DMSO-d6) Compound 35 650

19 F NMR (470 MHz, DMSO-d6) of Compound 35 651

Mass Spectra of Compound 35

C:\Xcalibur\...\ENP_IV_17_+ESI_Orbi 7/22/2013 10:50:45 PM ENP_IV_17

ENP_IV_17_+ESI_Orbi #15 RT: 0.12 AV: 1 NL: 3.16E8 T: FTMS + p ESI Full ms [150.00-2000.00] 348.1342 100 90 80 70 60 50 40

Relative Abundance 30 20 695.2606 10 386.0896 602.2897 241.1774 545.2573 717.2421 739.2241 887.7978 1010.5792 1104.3005 1264.73801411.49831495.9916 1616.4448 1743.4790 1883.8468 1999.0612 0 652 200 300 400 500 600 700 800 900 1000 1100 1200 1300 1400 1500 1600 1700 1800 1900 2000 330.1435 m/z

NL: 3.16E8 348.1342 100 ENP_IV_17_+ESI_Orbi#15 RT: 0.12 AV: 1 T: FTMS + p 80 ESI Full ms [150.00-2000.00] 60 40 20 Relative Abundance 347.1216 347.7953 349.1371 355.1248 358.3670 0 348.1341 350.1296 352.1250 NL: 100 346.1185 348.7852 351.1326 353.1284 354.1209 1.81E4 80 C 18 H19 F2 N3 S+H: 18CH20 F2 N3 S 1 60 p (gss, s /p:40) Chrg 1 R: 30000 Res .Pwr . @FWHM 40 20 349.1370 0 350.1331 346 347 348 349 350 351.1342 351 352.1353352 353.1363 354.1378353 355.1397354 355 356 357 358 m/z

Mass Spectra of Compound 35

C:\Xcalibur\...\ENP_IV_17_+ESI_Orbi 7/22/2013 10:50:45 PM ENP_IV_17

ENP_IV_17_+ESI_Orbi #15 RT: 0.12 AV: 1 NL: 3.16E8 T: FTMS + p ESI Full ms [150.00-2000.00] 348.1342 100 90 80 70 60 50 40

Relative Abundance 30 20 695.2606 10 386.0896 602.2897 241.1774 545.2573 717.2421 739.2241 887.7978 1010.5792 1104.3005 1264.73801411.49831495.9916 1616.4448 1743.4790 1883.8468 1999.0612 0 653 200 300 400 500 600 700 800 900 1000 1100 1200 1300 1400 1500 1600 1700 1800 1900 2000 330.1435 m/z

NL: 3.04E8 370.1160 100 ENP_IV_17_+ESI_Orbi#15 RT: 0.12 AV: 1 T: FTMS + p 80 ESI Full ms [150.00-2000.00] 60 40 20 Relative Abundance 370.8303371.1188371.2427 374.1176375.3229 376.1669377.3019 0 370.1160 372.1115 NL: 100 370.2403 373.1145 1.81E4 80 C 18 H19 F2 N3 S+Na: 18CH19 F2 N3 S 1Na 1 60 p (gss, s /p:40) Chrg 1 R: 30000 Res .Pwr . @FWHM 40 20 371.1189 0 372.1151 370.0 370.5 371.0 371.5 372.0 372.5373.1162 373.0 374.1172373.5 374.0 375.1182374.5 375.0 376.1197375.5 377.1216376.0 376.5 377.0 377.5 m/z

Mass Spectra of Compound 35

C:\Xcalibur\...\ENP_IV_17_+ESI_Orbi 7/22/2013 10:50:45 PM ENP_IV_17

ENP_IV_17_+ESI_Orbi #15 RT: 0.12 AV: 1 NL: 3.16E8 T: FTMS + p ESI Full ms [150.00-2000.00] 348.1342 100 90 80 70 60 50 40

Relative Abundance 30 20 695.2606 10 386.0896 602.2897 241.1774 545.2573 717.2421 739.2241 887.7978 1010.5792 1104.3005 1264.73801411.49831495.9916 1616.4448 1743.4790 1883.8468 1999.0612 0 654 200 300 400 500 600 700 800 900 1000 1100 1200 1300 1400 1500 1600 1700 1800 1900 2000 330.1435 m/z

NL: 5.74E7 717.2421 100 ENP_IV_17_+ESI_Orbi#15 RT: 0.12 AV: 1 T: FTMS + p 80 ESI Full ms [150.00-2000.00] 60

40 718.2451 20 Relative Abundance 726.2206 726.7186 727.7114 0 717.2428 NL: 719.2377 720.2395 100 717.0921 721.2286 725.4027 1.40E4 80 C 36 H38 F 4 N6 S 2 +Na: 36CH38 F 4 N6 S 2 Na 1 60 p (gss, s /p:40) Chrg 1 R: 30000 Res .Pwr . @FWHM 40 718.2457 20 719.2436 0 717 718 719 720.2440720 721.2436 721 722.2435722 723.2439 724.2446723 725.2455724 726.2466725 727.2471 726 727 m/z

HPLC Trace of Compound 35

655

HPLC Trace of Compound 35

656

HPLC Trace of Compound 35

657

1 H NMR (500 MHz, acetone-d6) of Compound 36 658

13 C NMR (125 MHz, acetone-d6) of Compound 36 659

19 F NMR (470 MHz, acetone-d6) of Compound 36 660

1 H NMR (500 MHz, acetone-d6) of Compound 37 661

13 C NMR (125 MHz, acetone-d6) of Compound 37 662

19 F NMR (470 MHz, acetone-d6) δ of Compound 37 663

Mass Spectra of Compound 37

C:\Xcalibur\...\ENP_V_90A_Orbi_-ESI 8/18/2014 3:33:27 PM

ENP_V_90A_Orbi_-ESI #20 RT: 0.27 AV: 1 NL: 3.03E6 T: FTMS - p ESI Full ms [150.00-2000.00] 398.11502 100 90 80 70 60 50 40

Relative Abundance 30 20 10 321.12109 254.91695 434.09113 518.06415681.29718 859.156921034.41357 1250.277711185.28064 1316.30786 1500.44775 1600.70215 1717.44312 1883.28003 0 364.12711 664 200 300 400 500 600 700 800 900 1000 1100 1200 1300 1400 1500 1600 1700 1800 1900 2000 m/z

NL: 3.03E6 398.11502 100 ENP_V_90A_Orbi_- ESI#20 RT: 0.27 80 AV: 1 T: FTMS - p 60 ESI Full ms [150.00-2000.00] 40 20

Relative Abundance 399.11673 397.97812 398.25473 402.11737403.39133 406.86566 0 398.11441 400.11057 NL: 100 396.10062 401.11273 7.46E5 80 C 21 H19 F2 N3 OS +H: 21CH18 F2 N3 O1S 1 60 pa Chrg -1 40 20 399.11777 0 396 397 398 399 400.11021400 401.11356 401 402.11692402 403.12027 404.11613403 405.11949404 406.12284405 406 407 m/z

HPLC Trace of Compound 37

665

HPLC Trace of Compound 37

666

HPLC Trace of Compound 37

667

REFERENCES

(1) Iiizumi, M.; Liu, W.; Pai, S. K.; Furuta, E.; Watabe, K. Drug Development against Metastasis-Related Genes and Their Pathways: A Rationale for Cancer Therapy. Biochim. Biophys. Acta BBA - Rev. Cancer 2008, 1786 (2), 87–104.

(2) Steeg, P. S. Tumor Metastasis: Mechanistic Insights and Clinical Challenges. Nat. Med. 2006, 12 (8), 895–904.

(3) McGrath, M. E. THE LYSOSOMAL CYSTEINE PROTEASES. Annu. Rev. Biophys. Biomol. Struct. 1999, 28 (1), 181–204.

(4) Rossi, A.; Deveraux, Q.; Turk, B.; Sali, A. Comprehensive Search for Cysteine Cathepsins in the . Biol. Chem. 2004, 385 (5).

(5) Rawlings, N. D.; Barrett, A. J. [32] Families of Cysteine Peptidases. In Methods in Enzymology; Elsevier, 1994; Vol. 244, pp 461–486.

(6) Rawlings, N. D.; Salvesen, G. Handbook of Proteolytic Enzymes; 2013.

(7) Polgár, L.; Halász, P. Current Problems in Mechanistic Studies of Serine and Cysteine Proteinases. Biochem. J. 1982, 207 (1), 1–10.

(8) Gocheva, V.; Joyce, J. A. Cysteine Cathepsins and the Cutting Edge of Cancer Invasion. Cell Cycle 2007, 6 (1), 60–64.

(9) Polgár, L. The Mechanism of Action of Thiolenzymes. Int. J. Biochem. 1977, 8 (3), 171– 176.

(10) Vernet, T.; Tessier, D. C.; Chatellier, J.; Plouffe, C.; Lee, T. S.; Thomas, D. Y.; Storer, A. C.; Menard, R. Structural and Functional Roles of Asparagine 175 in the Cysteine Protease Papain. J. Biol. Chem. 1995, 270 (28), 16645–16652.

(11) Menard, R.; Carriere, J.; Laflamme, P.; Plouffe, C.; Khouri, H. E.; Vernet, T.; Tessier, D. C.; Thomas, D. Y.; Storer, A. C. Contribution of the Glutamine 19 Side Chain to Transition-State Stabilization in the Oxyanion Hole of Papain. Biochemistry (Mosc.) 1991, 30 (37), 8924–8928.

(12) Schechter, I.; Berger, A. On the Size of the Active Site in Proteases. I. Papain. Biochem. Biophys. Res. Commun. 1967, 27 (2), 157–162.

(13) Berger, A.; Schechter, I. Mapping the Active Site of Papain with the Aid of Peptide Substrates and Inhibitors. Philos. Trans. R. Soc. B Biol. Sci. 1970, 257 (813), 249–264.

668

(14) Turk, B. Targeting Proteases: Successes, Failures and Future Prospects. Nat. Rev. Drug Discov. 2006, 5 (9), 785–799.

(15) Brömme, D.; Li, Z.; Barnes, M.; Mehler, E. Human Cathepsin V Functional Expression, Tissue Distribution, Electrostatic Surface Potential, Enzymatic Characterization, and Chromosomal Localization ‡. Biochemistry (Mosc.) 1999, 38 (8), 2377–2385.

(16) Bossard, M. J.; Tomaszek, T. A.; Thompson, S. K.; Amegadzie, B. Y.; Hanning, C. R.; Jones, C.; Kurdyla, J. T.; McNulty, D. E.; Drake, F. H.; Gowen, M.; Levy, M. A. Proteolytic Activity of Human Osteoclast Cathepsin K: EXPRESSION, PURIFICATION, ACTIVATION, AND SUBSTRATE IDENTIFICATION. J. Biol. Chem. 1996, 271 (21), 12517–12524.

(17) Mcgrath, M. E.; Palmer, J. T.; Brömme, D.; Somoza, J. R. Crystal Structure of Human Cathepsin S. Protein Sci. 1998, 7 (6), 1294–1302.

(18) Choe, Y.; Leonetti, F.; Greenbaum, D. C.; Lecaille, F.; Bogyo, M.; Bromme, D.; Ellman, J. A.; Craik, C. S. Substrate Profiling of Cysteine Proteases Using a Combinatorial Peptide Library Identifies Functionally Unique Specificities. J. Biol. Chem. 2006, 281 (18), 12824–12832.

(19) Biniossek, M. L.; Nägler, D. K.; Becker-Pauly, C.; Schilling, O. Proteomic Identification of Protease Cleavage Sites Characterizes Prime and Non-Prime Specificity of Cysteine Cathepsins B, L, and S. J. Proteome Res. 2011, 10 (12), 5363–5373.

(20) Honey, K.; Rudensky, A. Y. Lysosomal Cysteine Proteases Regulate Antigen Presentation. Nat. Rev. Immunol. 2003, 3 (6), 472–482.

(21) Ghosh, P.; Amaya, M.; Mellins, E.; Wiley, D. C. The Structure of an Intermediate in Class II MHC Maturation: CLIP Bound to HLA-DR3. Nature 1995, 378 (6556), 457– 462.

(22) Busch, R.; Cloutier, I.; Sékaly, R. P.; Hämmerling, G. J. Invariant Chain Protects Class II Histocompatibility Antigens from Binding Intact Polypeptides in the Endoplasmic Reticulum. EMBO J. 1996, 15 (2), 418–428.

(23) Cresswell, P. Invariant Chain Structure and MHC Class II Function. Cell 1996, 84 (4), 505–507.

(24) Nakagawa, T. Cathepsin L: Critical Role in Ii Degradation and CD4 T Cell Selection in the Thymus. Science 1998, 280 (5362), 450–453.

(25) Nakagawa, T. Y.; Brissette, W. H.; Lira, P. D.; Griffiths, R. J.; Petrushova, N.; Stock, J.; McNeish, J. D.; Eastman, S. E.; Howard, E. D.; Clarke, S. R. .; Rosloniec, E. F.; Elliott, E. A.; Rudensky, A. Y. Impaired Invariant Chain Degradation and Antigen Presentation and Diminished Collagen-Induced Arthritis in Cathepsin S Null Mice. Immunity 1999, 10 (2), 207–217.

669

(26) Riese, R. J.; Wolf, P. R.; Brömme, D.; Natkin, L. R.; Villadangos, J. A.; Ploegh, H. L.; Chapman, H. A. Essential Role for Cathepsin S in MHC Class II–Associated Invariant Chain Processing and Peptide Loading. Immunity 1996, 4 (4), 357–366.

(27) Honey, K. Cathepsin L Regulates CD4+ T Cell Selection Independently of Its Effect on Invariant Chain: A Role in the Generation of Positively Selecting Peptide Ligands. J. Exp. Med. 2002, 195 (10), 1349–1358.

(28) Hsieh, C.-S.; deRoos, P.; Honey, K.; Beers, C.; Rudensky, A. Y. A Role for Cathepsin L and Cathepsin S in Peptide Generation for MHC Class II Presentation. J. Immunol. Baltim. Md 1950 2002, 168 (6), 2618–2625.

(29) Gelb, B. D.; Shi, G.-P.; Chapman, H. A.; Desnick, R. J. Pycnodysostosis, a Lysosomal Disease Caused by Cathepsin K Deficiency. Science 1996, 273 (5279), 1236–1238.

(30) Johnson, M. R.; Polymeropoulos, M. H.; Vos, H. L.; Ortiz de Luna, R. I.; Francomano, C. A. A Nonsense Mutation in the Cathepsin K Gene Observed in a Family with Pycnodysostosis. Genome Res. 1996, 6 (11), 1050–1055.

(31) Rodan, S. B.; Duong, L. T. Cathepsin K – A New Molecular Target for Osteoporosis. IBMS BoneKEy 2008, 5 (1), 16–24.

(32) Teitelbaum, S. L. Bone Resorption by Osteoclasts. Science 2000, 289 (5484), 1504– 1508.

(33) Kini, U.; Nandeesh, B. N. Physiology of Bone Formation, Remodeling, and Metabolism. In Radionuclide and Hybrid Bone Imaging; Fogelman, I., Gnanasegaran, G., van der Wall, H., Eds.; Springer Berlin Heidelberg: Berlin, Heidelberg, 2012; pp 29–57.

(34) Neuman, W. F.; Neuman, M. W. The Nature of the Mineral Phase of Bone. Chem. Rev. 1953, 53 (1), 1–45.

(35) Eastoe, J. E.; Eastoe, B. The Organic Constituents of Mammalian Compact Bone. Biochem. J. 1954, 57 (3), 453–459.

(36) Herring, G. M.; Ashton, B. A.; Chipperfield, A. R. The Isolation of Soluble Proteins, Glycoproteins, and Proteoglycans from Bone. Prep. Biochem. 1974, 4 (2), 179–200.

(37) Herring, G. M. A Comparison of Bone Matrix and Tendon with Particular Reference to Glycoprotein Content. Biochem. J. 1976, 159 (3), 749–755.

(38) Blair, H. C.; Kahn, A. J.; Crouch, E. C.; Jeffrey, J. J.; Teitelbaum, S. L. Isolated Osteoclasts Resorb the Organic and Inorganic Components of Bone. J. Cell Biol. 1986, 102 (4), 1164–1172.

(39) Blair, H. C.; Teitelbaum, S. L.; Ghiselli, R.; Gluck, S. Osteoclastic Bone Resorption by a Polarized Vacuolar Proton Pump. Science 1989, 245 (4920), 855–857.

670

(40) Gowen, M.; Lazner, F.; Dodds, R.; Kapadia, R.; Feild, J.; Tavaria, M.; Bertoncello, I.; Drake, F.; Zavarselk, S.; Tellis, I.; Hertzog, P.; Debouck, C.; Kola, I. Cathepsin K Knockout Mice Develop Osteopetrosis Due to a Deficit in Matrix Degradation but Not Demineralization. J. Bone Miner. Res. 1999, 14 (10), 1654–1663.

(41) Nesbitt, S. A. Trafficking of Matrix Collagens Through Bone-Resorbing Osteoclasts. Science 1997, 276 (5310), 266–269.

(42) Yasothornsrikul, S.; Greenbaum, D.; Medzihradszky, K. F.; Toneff, T.; Bundey, R.; Miller, R.; Schilling, B.; Petermann, I.; Dehnert, J.; Logvinova, A.; Goldsmith, P.; Neveu, J. M.; Lane, W. S.; Gibson, B.; Reinheckel, T.; Peters, C.; Bogyo, M.; Hook, V. Cathepsin L in Secretory Vesicles Functions as a Prohormone-Processing Enzyme for Production of the Enkephalin Peptide Neurotransmitter. Proc. Natl. Acad. Sci. U. S. A. 2003, 100 (16), 9590–9595.

(43) Funkelstein, L.; Beinfeld, M.; Minokadeh, A.; Zadina, J.; Hook, V. Unique Biological Function of Cathepsin L in Secretory Vesicles for Biosynthesis of Neuropeptides. Neuropeptides 2010, 44 (6), 457–466.

(44) Funkelstein, L.; Toneff, T.; Hwang, S.-R.; Reinheckel, T.; Peters, C.; Hook, V. Cathepsin L Participates in the Production of Neuropeptide Y in Secretory Vesicles, Demonstrated by Protease Gene Knockout and Expression. J. Neurochem. 2008, 106 (1), 384–391.

(45) Funkelstein, L.; Toneff, T.; Mosier, C.; Hwang, S.-R.; Beuschlein, F.; Lichtenauer, U. D.; Reinheckel, T.; Peters, C.; Hook, V. Major Role of Cathepsin L for Producing the Peptide Hormones ACTH, -Endorphin, and -MSH, Illustrated by Protease Gene Knockout and Expression. J. Biol. Chem. 2008, 283 (51), 35652–35659.

(46) Minokadeh, A.; Funkelstein, L.; Toneff, T.; Hwang, S.-R.; Beinfeld, M.; Reinheckel, T.; Peters, C.; Zadina, J.; Hook, V. Cathepsin L Participates in Dynorphin Production in Brain Cortex, Illustrated by Protease Gene Knockout and Expression. Mol. Cell. Neurosci. 2010, 43 (1), 98–107.

(47) Beinfeld, M. C.; Funkelstein, L.; Foulon, T.; Cadel, S.; Kitagawa, K.; Toneff, T.; Reinheckel, T.; Peters, C.; Hook, V. Cathepsin L Plays a Major Role in Cholecystokinin Production in Mouse Brain Cortex and in Pituitary AtT-20 Cells: Protease Gene Knockout and Inhibitor Studies. Peptides 2009, 30 (10), 1882–1891.

(48) Janecka, A.; Fichna, J.; Janecki, T. Opioid Receptors and Their Ligands. Curr. Top. Med. Chem. 2004, 4 (1), 1–17.

(49) Kuo, L. E.; Kitlinska, J. B.; Tilan, J. U.; Li, L.; Baker, S. B.; Johnson, M. D.; Lee, E. W.; Burnett, M. S.; Fricke, S. T.; Kvetnansky, R.; Herzog, H.; Zukowska, Z. Neuropeptide Y Acts Directly in the Periphery on Fat Tissue and Mediates Stress-Induced Obesity and Metabolic Syndrome. Nat. Med. 2007, 13 (7), 803–811.

(50) Walker, P.; Grouzmann, E.; Burnier, M.; Waeber, B. The Role of Neuropeptide Y in Cardiovascular Regulation. Trends Pharmacol. Sci. 1991, 12, 111–115. 671

(51) Little, T. J.; Horowitz, M.; Feinle-Bisset, C. Role of Cholecystokinin in Appetite Control and Body Weight Regulation. Obes. Rev. 2005, 6 (4), 297–306.

(52) Hernandez-Gómez, A. M.; Aguilar-Roblero, R.; Pérez de la Mora, M. Role of Cholecystokinin-A and Cholecystokinin-B Receptors in Anxiety. Amino Acids 2002, 23 (1-3), 283–290.

(53) Sudhan, D. R.; Siemann, D. W. Cathepsin L Targeting in Cancer Treatment. Pharmacol. Ther. 2015.

(54) Lankelma, J. M.; Voorend, D. M.; Barwari, T.; Koetsveld, J.; Van der Spek, A. H.; De Porto, A. P. N. A.; Van Rooijen, G.; Van Noorden, C. J. F. Cathepsin L, Target in Cancer Treatment? Life Sci. 2010, 86 (7-8), 225–233.

(55) Brömme, D.; Wilson, S. Role of Cysteine Cathepsins in Extracellular Proteolysis. In Extracellular Matrix Degradation; Parks, W. C., Mecham, R. P., Eds.; Springer Berlin Heidelberg: Berlin, Heidelberg, 2011; pp 23–51.

(56) Platt, M.; Dumas, J. Cathepsins and Other Proteases in Tumor Angiogenesis. In Tumor Angiogenesis Regulators; Gonzalez-Perez, R., Rueda, B., Eds.; CRC Press, 2013; pp 297–339.

(57) Dong, J. M.; Prence, E. M.; Sahagian, G. G. Mechanism for Selective Secretion of a Lysosomal Protease by Transformed Mouse Fibroblasts. J. Biol. Chem. 1989, 264 (13), 7377–7383.

(58) Gottesman, M. M. Transformation-Dependent Secretion of a Low Molecular Weight Protein by Murine Fibroblasts. Proc. Natl. Acad. Sci. U. S. A. 1978, 75 (6), 2767–2771.

(59) Gal, S.; Gottesman, M. M. Isolation and Sequence of a cDNA for Human pro-(cathepsin L). Biochem. J. 1988, 253 (1), 303–306.

(60) Troen, B. R.; Gal, S.; Gottesman, M. M. Sequence and Expression of the cDNA for MEP (major Excreted Protein), a Transformation-Regulated Secreted Cathepsin. Biochem. J. 1987, 246 (3), 731–735.

(61) Chen, Q.; Chen, L.; Zhao, R.; Yang, X.; Imran, K.; Xing, C. Microarray Analyses Reveal Liver Metastasis-Related Genes in Metastatic Colorectal Cancer Cell Model. J. Cancer Res. Clin. Oncol. 2013, 139 (7), 1169–1178.

(62) Kedinger, V.; Sansregret, L.; Harada, R.; Vadnais, C.; Cadieux, C.; Fathers, K.; Park, M.; Nepveu, A. p110 CUX1 Homeodomain Protein Stimulates Cell Migration and Invasion in Part through a Regulatory Cascade Culminating in the Repression of E-Cadherin and Occludin. J. Biol. Chem. 2009, 284 (40), 27701–27711.

(63) Jean, D.; Rousselet, N.; Frade, R. Expression of Cathepsin L in Human Tumor Cells Is under the Control of Distinct Regulatory Mechanisms. Oncogene 2006, 25 (10), 1474– 1484. 672

(64) Carmona, E.; Dufour, É.; Plouffe, C.; Takebe, S.; Mason, P.; Mort, J. S.; Ménard, R. Potency and Selectivity of the Cathepsin L Propeptide as an Inhibitor of Cysteine Proteases †. Biochemistry (Mosc.) 1996, 35 (25), 8149–8157.

(65) Maubach, G.; Schilling, K.; Rommerskirch, W.; Wenz, I.; Schultz, J. E.; Weber, E.; Wiederanders, B. The Inhibition of Cathepsin S by Its Propeptide--Specificity and Mechanism of Action. Eur. J. Biochem. FEBS 1997, 250 (3), 745–750.

(66) Burden, R. E.; Snoddy, P.; Jefferies, C. A.; Walker, B.; Scott, C. J. Inhibition of Cathepsin L-like Proteases by Cathepsin V Propeptide. Biol. Chem. 2007, 388 (5).

(67) Bromme, D. Production and Activation of Recombinant Papain-like Cysteine Proteases. Methods 2004, 32 (2), 199–206.

(68) McQueney, M. S. Autocatalytic Activation of Human Cathepsin K. J. Biol. Chem. 1997, 272 (21), 13955–13960.

(69) Sudhan, D. R.; Siemann, D. W. Cathepsin L Inhibition by the Small Molecule KGP94 Suppresses Tumor Microenvironment Enhanced Metastasis Associated Cell Functions of Prostate and Breast Cancer Cells. Clin. Exp. Metastasis 2013, 30 (7), 891–902.

(70) Rothberg, J. M.; Bailey, K. M.; Wojtkowiak, J. W.; Ben-Nun, Y.; Bogyo, M.; Weber, E.; Moin, K.; Blum, G.; Mattingly, R. R.; Gillies, R. J.; Sloane, B. F. Acid-Mediated Tumor Proteolysis: Contribution of Cysteine Cathepsins. Neoplasia N. Y. N 2013, 15 (10), 1125–1137.

(71) Turk, V.; Turk, B. Lysosomal Cysteine Proteases and Their Protein Inhibitors: Recent Developments. Acta Chim. Slov. 2008, 55 (4), 727–738.

(72) Cox, J. L. Cystatins and Cancer. Front. Biosci. Landmark Ed. 2009, 14, 463–474.

(73) Blondeau, X.; Vidmar, S. L.; Emod, I.; Pagano, M.; Turk, V.; Keil-Dlouha, V. Generation of Matrix-Degrading Proteolytic System from Fibronectin by Cathepsins B, G, H and L. Biol. Chem. Hoppe. Seyler 1993, 374 (8), 651–656.

(74) Ishidoh, K.; Kominami, E. Procathepsin L Degrades Extracellular Matrix Proteins in the Presence of Glycosaminoglycans in Vitro. Biochem. Biophys. Res. Commun. 1995, 217 (2), 624–631.

(75) Guinec, N.; Dalet-Fumeron, V.; Pagano, M. “In Vitro” Study of Basement Membrane Degradation by the Cysteine Proteinases, Cathepsins B, B-like and L. Digestion of Collagen IV, Laminin, Fibronectin, and Release of Gelatinase Activities from Basement Membrane Fibronectin. Biol. Chem. Hoppe. Seyler 1993, 374 (12), 1135–1146.

(76) Buck, M. R.; Karustis, D. G.; Day, N. A.; Honn, K. V.; Sloane, B. F. Degradation of Extracellular-Matrix Proteins by Human Cathepsin B from Normal and Tumour Tissues. Biochem. J. 1992, 282 ( Pt 1), 273–278.

673

(77) Kirschke, H.; Kembhavi, A. A.; Bohley, P.; Barrett, A. J. Action of Rat Liver Cathepsin L on Collagen and Other Substrates. Biochem. J. 1982, 201 (2), 367–372.

(78) Maciewicz, R. A.; Wotton, S. F.; Etherington, D. J.; Duance, V. C. Susceptibility of the Cartilage Collagens Types II, IX and XI to Degradation by the Cysteine Proteinases, Cathepsins B and L. FEBS Lett. 1990, 269 (1), 189–193.

(79) Garnero, P.; Borel, O.; Byrjalsen, I.; Ferreras, M.; Drake, F. H.; McQueney, M. S.; Foged, N. T.; Delmas, P. D.; Delaissé, J. M. The Collagenolytic Activity of Cathepsin K Is Unique among Mammalian Proteinases. J. Biol. Chem. 1998, 273 (48), 32347–32352.

(80) Sameni, M.; Dosescu, J.; Moin, K.; Sloane, B. F. Functional Imaging of Proteolysis: Stromal and Inflammatory Cells Increase Tumor Proteolysis. Mol. Imaging 2003, 2 (3), 159–175.

(81) Mason, S. D.; Joyce, J. A. Proteolytic Networks in Cancer. Trends Cell Biol. 2011, 21 (4), 228–237.

(82) GSK Product Development Pipeline February 2014. http://www.gsk.com/media/280387/product-Pipeline-2014.pdf (Accessed on September, 2, 2015).

(83) A Study to Evaluate the Safety, Tolerability, Pharmacokinetics (PK), Pharmacodynamics (PD) and Food Effect of Single or Repeat Doses of GSK2793660 in Healthy Subjects. https://clinicaltrials.gov/ct2/show/NCT02058407 (accessed Sep 2, 2015).

(84) Gauthier, J. Y.; Chauret, N.; Cromlish, W.; Desmarais, S.; Duong, L. T.; Falgueyret, J.- P.; Kimmel, D. B.; Lamontagne, S.; Léger, S.; LeRiche, T.; Li, C. S.; Massé, F.; McKay, D. J.; Nicoll-Griffith, D. A.; Oballa, R. M.; Palmer, J. T.; Percival, M. D.; Riendeau, D.; Robichaud, J.; Rodan, G. A.; Rodan, S. B.; Seto, C.; Thérien, M.; Truong, V.-L.; Venuti, M. C.; Wesolowski, G.; Young, R. N.; Zamboni, R.; Black, W. C. The Discovery of Odanacatib (MK-0822), a Selective Inhibitor of Cathepsin K. Bioorg. Med. Chem. Lett. 2008, 18 (3), 923–928.

(85) Stoch, S. A.; Zajic, S.; Stone, J. A.; Miller, D. L.; van Bortel, L.; Lasseter, K. C.; Pramanik, B.; Cilissen, C.; Liu, Q.; Liu, L.; Scott, B. B.; Panebianco, D.; Ding, Y.; Gottesdiener, K.; Wagner, J. A. Odanacatib, a Selective Cathepsin K Inhibitor to Treat Osteoporosis: Safety, Tolerability, Pharmacokinetics and Pharmacodynamics - Results from Single Oral Dose Studies in Healthy Volunteers: Odanacatib Pharmacokinetics and Pharmacodynamics. Br. J. Clin. Pharmacol. 2013, 75 (5), 1240–1254.

(86) Lindstrom, E.; Grabowska, U.; Jerling, M.; Edenius, C. MIV-711, a Highly Selective Cathepsin K Inhibitor, Reduces Biomarkers of Bone Resorption and Cartilage Degradation in Healthy Subjects. Osteoarthritis Cartilage 2014, 22, S197.

(87) About Cathepsin B and Liver Disease. http://www.virobayinc.com/liver-fibrosis.php (accessed Sep 2, 2015).

674

(88) Evaluation of Safety and Pharmacokinetics of Single Doses of VBY-376 in Healthy Adults. https://clinicaltrials.gov/ct2/show/NCT00557583 (accessed Sep 2, 2015).

(89) Cathepsin S inhibitor: VBY-891 for Psoriasis. http://www.virobayinc.com/VBY-891.php (accessed Sep 2, 2015).

(90) Safety Study of VBY-891 in Healthy Volunteers After Single or Multiple (7 Days) of Oral Dosing (VBY891P1). https://clinicaltrials.gov/ct2/show/NCT01947738 (accessed Sep 2, 2015).

(91) Study to Investigate the Safety, Tolerability, Absorption, Distribution, Metabolism, and Elimination of RWJ-445380 Administered to Patients With Plaque Psoriasis. https://clinicaltrials.gov/ct2/show/NCT00396422 (accessed Sep 2, 2015).

(92) Safety and Effectiveness Study of RWJ-445380 Cathepsin-S Inhibitor in Patients With Active Rheumatoid Arthritis Despite Methotrexate Therapy. https://clinicaltrials.gov/ct2/show/NCT00425321 (accessed Sep 2, 2015).

(93) Cathepsin S inhibitor: VBY-036 for Neuropathic Pain and Crohn’s Disease. http://www.virobayinc.com/VBY-036.php (accessed Sep 2, 2015).

(94) Safety Study of VBY-036 in Healthy Volunteers After 7 Days of Oral Dosing. https://clinicaltrials.gov/ct2/show/NCT01892891 (accessed Sep 2, 2015).

(95) Rizoska, B; Edenius, C.; Göhlin, K.; Hewitt, E.; Lindström, E.; Malcangio, M.; Pitcher, T.; Tunblad, K. MIV-247 A Cathepsin Protease Inhibitor Developed as a Novel Therapy for Neuropathic Pain. Proceedings of the 15th World Congress on Pain 2014, Buenos Aires, Argentina, October 6-11, 2014.

(96) Pan cathepsin inhibitor: VBY-825. http://www.virobayinc.com/VBY-825.php (accessed Sep 2, 2015).

(97) Elie, B. T.; Gocheva, V.; Shree, T.; Dalrymple, S. A.; Holsinger, L. J.; Joyce, J. A. Identification and Pre-Clinical Testing of a Reversible Cathepsin Protease Inhibitor Reveals Anti-Tumor Efficacy in a Pancreatic Cancer Model. Biochimie 2010, 92 (11), 1618–1624.

(98) Jadhav, P. K.; Schiffler, M. A.; Gavardinas, K.; Kim, E. J.; Matthews, D. P.; Staszak, M. A.; Coffey, D. S.; Shaw, B. W.; Cassidy, K. C.; Brier, R. A.; Zhang, Y.; Christie, R. M.; Matter, W. F.; Qing, K.; Durbin, J. D.; Wang, Y.; Deng, G. G. Discovery of Cathepsin S Inhibitor LY3000328 for the Treatment of Abdominal Aortic Aneurysm. ACS Med. Chem. Lett. 2014, 5 (10), 1138–1142.

(99) Payne, C. D.; Deeg, M. A.; Chan, M.; Tan, L. H.; LaBell, E. S.; Shen, T.; DeBrota, D. J. Pharmacokinetics and Pharmacodynamics of the Cathepsin S Inhibitor, LY3000328, in Healthy Subjects: PK/PD and Safety and Tolerability of Cathepsin S Inhibitor. Br. J. Clin. Pharmacol. 2014, 78 (6), 1334–1342.

675

(100) Ramalho, S. D.; De Sousa, L. R. F.; Nebo, L.; Maganhi, S. H.; Caracelli, I.; Zukerman- Schpector, J.; Lima, M. I. S.; Alves, M. F. M.; Da Silva, M. F. das G. F.; Fernandes, J. B.; Vieira, P. C. Triterpenoids as Novel Natural Inhibitors of Human Cathepsin L. Chem. Biodivers. 2014, 11 (9), 1354–1363.

(101) Burger, M. C. de M.; Fernandes, J. B.; da Silva, M. F. das G. F.; Escalante, A.; Prudhomme, J.; Le Roch, K. G.; Izidoro, M. A.; Vieira, P. C. Structures and Bioactivities of Dihydrochalcones from Metrodorea Stipularis . J. Nat. Prod. 2014, 77 (11), 2418–2422.

(102) Hanada, K.; Tamai, M.; Ohmura, S.; Sawada, J.; Seki, T.; Tanaka, I. Structure and Synthesis of E–64, a New Thiol Protease Inhibitor. Agric. Biol. Chem. 1978, 42 (3), 529– 536.

(103) Inaba, T.; Hirayama, Y.; Fujinaga, N. Inhibition of Cathepsin B1 by E-64, a Thiol Proteinase Inhibitor, and Its Derivatives. Agric. Biol. Chem. 1979, 43 (3), 655–656.

(104) Matsumoto, K.; Mizoue, K.; Kitamura, K.; Tse, W. C.; Huber, C. P.; Ishida, T. Structural Basis of Inhibition of Cysteine Proteases by E-64 and Its Derivatives. Biopolymers 1999, 51 (1), 99–107.

(105) Barrett, A. J.; Kembhavi, A. A.; Brown, M. A.; Kirschke, H.; Knight, C. G.; Tamai, M.; Hanada, K. L-Trans-Epoxysuccinyl-leucylamido(4-Guanidino)butane (E-64) and Its Analogues as Inhibitors of Cysteine Proteinases Including Cathepsins B, H and L. Biochem. J. 1982, 201 (1), 189–198.

(106) Kondo, S. I.; Kawamura, K.; Iwanaga, J.; Hamada, M.; Aoyagi, T. Isolation and Characterization of Leupeptins Produced by Actinomycetes. Chem. Pharm. Bull. (Tokyo) 1969, 17 (9), 1896–1901.

(107) Aoyagi, T.; Takeuchi, T.; Matsuzaki, A.; Kawamura, K.; Kondo, S. Leupeptins, New Protease Inhibitors from Actinomycetes. J. Antibiot. (Tokyo) 1969, 22 (6), 283–286.

(108) Baici, A.; Gyger-Marazzi, M. The Slow, Tight-Binding Inhibition of Cathepsin B by Leupeptin. A Hysteretic Effect. Eur. J. Biochem. FEBS 1982, 129 (1), 33–41.

(109) Yasuma, T.; Oi, S.; Choh, N.; Nomura, T.; Furuyama, N.; Nishimura, A.; Fujisawa, Y.; Sohda, T. Synthesis of Peptide Aldehyde Derivatives as Selective Inhibitors of Human Cathepsin L and Their Inhibitory Effect on Bone Resorption 1. J. Med. Chem. 1998, 41 (22), 4301–4308.

(110) Fusetani, N.; Fujita, M.; Nakao, Y.; Matsunaga, S.; van Soest, R. W. M. Tokaramide A,a New Cathepsin B Inhibitor from the Marine Sponge Theonella Aff. Mirabilis. Bioorg. Med. Chem. Lett. 1999, 9 (24), 3397–3402.

(111) Konno, H.; Kubo, K.; Makabe, H.; Toshiro, E.; Hinoda, N.; Nosaka, K.; Akaji, K. Total Synthesis of Miraziridine A and Identification of Its Major Reaction Site for Cathepsin B. Tetrahedron 2007, 63 (38), 9502–9513. 676

(112) Schaschke, N. Miraziridine A: Natures Blueprint towards Protease Class-Spanning Inhibitors. Bioorg. Med. Chem. Lett. 2004, 14 (4), 855–857.

(113) Linington, R. G.; Clark, B. R.; Trimble, E. E.; Almanza, A.; Ureña, L.-D.; Kyle, D. E.; Gerwick, W. H. Antimalarial Peptides from Marine Cyanobacteria: Isolation and Structural Elucidation of Gallinamide A. J. Nat. Prod. 2009, 72 (1), 14–17.

(114) Miller, B.; Friedman, A. J.; Choi, H.; Hogan, J.; McCammon, J. A.; Hook, V.; Gerwick, W. H. The Marine Cyanobacterial Metabolite Gallinamide A Is a Potent and Selective Inhibitor of Human Cathepsin L. J. Nat. Prod. 2014, 77 (1), 92–99.

(115) Katunuma, N.; Murata, E.; Kakegawa, H.; Matsui, A.; Tsuzuki, H.; Tsuge, H.; Turk, D.; Turk, V.; Fukushima, M.; Tada, Y.; Asao, T. Structure Based Development of Novel Specific Inhibitors for Cathepsin L and Cathepsin S in Vitro and in Vivo. FEBS Lett. 1999, 458 (1), 6–10.

(116) Mendieta, L.; Picó, A.; Tarragó, T.; Teixidó, M.; Castillo, M.; Rafecas, L.; Moyano, A.; Giralt, E. Novel Peptidyl Aryl Vinyl Sulfones as Highly Potent and Selective Inhibitors of Cathepsins L and B. ChemMedChem 2010, 5 (9), 1556–1567.

(117) Dana, D.; De, S.; Rathod, P.; Davalos, A. R.; Novoa, D. A.; Paroly, S.; Torres, V. M.; Afzal, N.; Lankalapalli, R. S.; Rotenberg, S. A.; Chang, E. J.; Subramaniam, G.; Kumar, S. Development of a Highly Potent, Selective, and Cell-Active Inhibitor of Cysteine Cathepsin L–A Hybrid Design Approach. Chem. Commun. 2014, 50 (74), 10875.

(118) Chen, J.-C.; Uang, B.-J.; Lyu, P.-C.; Chang, J.-Y.; Liu, K.-J.; Kuo, C.-C.; Hsieh, H.-P.; Wang, H.-C.; Cheng, C.-S.; Chang, Y.-H.; Chang, M. D.-T.; Chang, W.-S. W.; Lin, C.- C. Design and Synthesis of α-Ketoamides as Cathepsin S Inhibitors with Potential Applications against Tumor Invasion and Angiogenesis. J. Med. Chem. 2010, 53 (11), 4545–4549.

(119) Marquis, R. W.; Ru, Y.; LoCastro, S. M.; Zeng, J.; Yamashita, D. S.; Oh, H.-J.; Erhard, K. F.; Davis, L. D.; Tomaszek, T. A.; Tew, D.; Salyers, K.; Proksch, J.; Ward, K.; Smith, B.; Levy, M.; Cummings, M. D.; Haltiwanger, R. C.; Trescher, G.; Wang, B.; Hemling, M. E.; Quinn, C. J.; Cheng, H.-Y.; Lin, F.; Smith, W. W.; Janson, C. A.; Zhao, B.; McQueney, M. S.; D’Alessio, K.; Lee, C.-P.; Marzulli, A.; Dodds, R. A.; Blake, S.; James, I. E.; Gress, C. J.; Bradley, B. R.; Lark, M. W.; Gowen, M.; Veber, D. F. Azepanone-Based Inhibitors of Human and Rat Cathepsin K. J. Med. Chem. 2001, 44 (9), 1380–1395.

(120) Marquis, R. W.; James, I.; Zeng, J.; Trout, R. E. L.; Thompson, S.; Rahman, A.; Yamashita, D. S.; Xie, R.; Ru, Y.; Gress, C. J.; Blake, S.; Lark, M. A.; Hwang, S.-M.; Tomaszek, T.; Offen, P.; Head, M. S.; Cummings, M. D.; Veber, D. F. Azepanone-Based Inhibitors of Human Cathepsin L. J. Med. Chem. 2005, 48 (22), 6870–6878.

677

(121) Myers, M. C.; Shah, P. P.; Diamond, S. L.; Huryn, D. M.; Smith, A. B. Identification and Synthesis of a Unique Thiocarbazate Cathepsin L Inhibitor. Bioorg. Med. Chem. Lett. 2008, 18 (1), 210–214.

(122) Myers, M. C.; Shah, P. P.; Beavers, M. P.; Napper, A. D.; Diamond, S. L.; Smith, A. B.; Huryn, D. M. Design, Synthesis, and Evaluation of Inhibitors of Cathepsin L: Exploiting a Unique Thiocarbazate Chemotype. Bioorg. Med. Chem. Lett. 2008, 18 (12), 3646– 3651.

(123) Liu, Z.; Myers, M. C.; Shah, P. P.; Beavers, M. P.; Benedetti, P. A.; Diamond, S. L.; Smith, A. B.; Huryn, D. M. Design, Synthesis and Biological Evaluation of a Library of Thiocarbazates and Their Activity as Cysteine Protease Inhibitors. Comb. Chem. High Throughput Screen. 2010, 13 (4), 337–351.

(124) Ehmke, V.; Winkler, E.; Banner, D. W.; Haap, W.; Schweizer, W. B.; Rottmann, M.; Kaiser, M.; Freymond, C.; Schirmeister, T.; Diederich, F. Optimization of Triazine Nitriles as Rhodesain Inhibitors: Structure-Activity Relationships, Bioisosteric Imidazopyridine Nitriles, and X-Ray Crystal Structure Analysis with Human Cathepsin L. ChemMedChem 2013, 8 (6), 967–975.

(125) Falgueyret, J. P.; Oballa, R. M.; Okamoto, O.; Wesolowski, G.; Aubin, Y.; Rydzewski, R. M.; Prasit, P.; Riendeau, D.; Rodan, S. B.; Percival, M. D. Novel, Nonpeptidic Cyanamides as Potent and Reversible Inhibitors of Human Cathepsins K and L. J. Med. Chem. 2001, 44 (1), 94–104.

(126) Siles, R.; Zhou, M.; Ackley, J. F.; Pinney, K. G.; Chen, S.-E.; Arispe-Angulo, W. M.; Trawick, M. L. Inhibitors of Cysteine Proteases and Methods of Use Thereof. US8173696 B2, May 8, 2012.

(127) Kishore Kumar, G. D.; Chavarria, G. E.; Charlton-Sevcik, A. K.; Arispe, W. M.; MacDonough, M. T.; Strecker, T. E.; Chen, S.-E.; Siim, B. G.; Chaplin, D. J.; Trawick, M. L.; Pinney, K. G. Design, Synthesis, and Biological Evaluation of Potent Thiosemicarbazone Based Cathepsin L Inhibitors. Bioorg. Med. Chem. Lett. 2010, 20 (4), 1415–1419.

(128) Kumar, G. D. K.; Chavarria, G. E.; Charlton-Sevcik, A. K.; Yoo, G. K.; Song, J.; Strecker, T. E.; Siim, B. G.; Chaplin, D. J.; Trawick, M. L.; Pinney, K. G. Functionalized Benzophenone, Thiophene, Pyridine, and Fluorene Thiosemicarbazone Derivatives as Inhibitors of Cathepsin L. Bioorg. Med. Chem. Lett. 2010, 20 (22), 6610–6615.

(129) Chavarria, G. E.; Horsman, M. R.; Arispe, W. M.; Kumar, G. D. K.; Chen, S.-E.; Strecker, T. E.; Parker, E. N.; Chaplin, D. J.; Pinney, K. G.; Trawick, M. L. Initial Evaluation of the Antitumour Activity of KGP94, a Functionalized Benzophenone Thiosemicarbazone Inhibitor of Cathepsin L. Eur. J. Med. Chem. 2012, 58, 568–572.

678

(130) Song, J.; Jones, L. M.; Kumar, G. D. K.; Conner, E. S.; Bayeh, L.; Chavarria, G. E.; Charlton-Sevcik, A. K.; Chen, S.-E.; Chaplin, D. J.; Trawick, M. L.; Pinney, K. G. Synthesis and Biochemical Evaluation of Thiochromanone Thiosemicarbazone Analogues as Inhibitors of Cathepsin L. ACS Med. Chem. Lett. 2012, 3 (6), 450–453.

(131) Song, J.; Jones, L. M.; Chavarria, G. E.; Charlton-Sevcik, A. K.; Jantz, A.; Johansen, A.; Bayeh, L.; Soeung, V.; Snyder, L. K.; Lade, S. D.; Chaplin, D. J.; Trawick, M. L.; Pinney, K. G. Small-Molecule Inhibitors of Cathepsin L Incorporating Functionalized Ring-Fused Molecular Frameworks. Bioorg. Med. Chem. Lett. 2013, 23 (9), 2801–2807.

(132) Chaplin, D. J.; Devanna, K. K. G.; Parker, E.; Pinney, K. G.; Song, J.; Trawick, M. L. Compositions and Methods for Inhibition of Cathepsins. US8877967 B2, November 4, 2014.

(133) Parker, E. N.; Song, J.; Kishore Kumar, G. D.; Odutola, S. O.; Chavarria, G. E.; Charlton-Sevcik, A. K.; Strecker, T. E.; Barnes, A. L.; Sudhan, D. R.; Wittenborn, T. R.; Siemann, D. W.; Horsman, M. R.; Chaplin, D. J.; Trawick, M. L.; Pinney, K. G. Synthesis and Biochemical Evaluation of Benzoylbenzophenone Thiosemicarbazone Analogues as Potent and Selective Inhibitors of Cathepsin L. Bioorg. Med. Chem. 2015, 21, 6974-6992.

(134) Raghav, N.; Kaur, R. Synthesis and Evaluation of Some Semicarbazone- and Thiosemicarbazone-Based Cathepsin B Inhibitors. Med. Chem. Res. 2014, 23 (11), 4669– 4679.

(135) Siles, R.; Chen, S.-E.; Zhou, M.; Pinney, K. G.; Trawick, M. L. Design, Synthesis, and Biochemical Evaluation of Novel Cruzain Inhibitors with Potential Application in the Treatment of Chagas’ Disease. Bioorg. Med. Chem. Lett. 2006, 16 (16), 4405–4409.

(136) Du, X.; Guo, C.; Hansell, E.; Doyle, P. S.; Caffrey, C. R.; Holler, T. P.; McKerrow, J. H.; Cohen, F. E. Synthesis and Structure−Activity Relationship Study of Potent Trypanocidal Thio Semicarbazone Inhibitors of the Trypanosomal Cysteine Protease Cruzain. J. Med. Chem. 2002, 45 (13), 2695–2707.

(137) Mallari, J. P.; Shelat, A.; Kosinski, A.; Caffrey, C. R.; Connelly, M.; Zhu, F.; McKerrow, J. H.; Guy, R. K. Discovery of Trypanocidal Thiosemicarbazone Inhibitors of Rhodesain and TbcatB. Bioorg. Med. Chem. Lett. 2008, 18 (9), 2883–2885.

(138) Caputto, M. E.; Fabian, L. E.; Benítez, D.; Merlino, A.; Ríos, N.; Cerecetto, H.; Moltrasio, G. Y.; Moglioni, A. G.; González, M.; Finkielsztein, L. M. Thiosemicarbazones Derived from 1-Indanones as New Anti-Trypanosoma Cruzi Agents. Bioorg. Med. Chem. 2011, 19 (22), 6818–6826.

(139) Fujii, N.; Mallari, J. P.; Hansell, E. J.; Mackey, Z.; Doyle, P.; Zhou, Y. M.; Gut, J.; Rosenthal, P. J.; McKerrow, J. H.; Guy, R. K. Discovery of Potent Thiosemicarbazone Inhibitors of Rhodesain and Cruzain. Bioorg. Med. Chem. Lett. 2005, 15 (1), 121–123.

679

(140) Greenbaum, D. C.; Mackey, Z.; Hansell, E.; Doyle, P.; Gut, J.; Caffrey, C. R.; Lehrman, J.; Rosenthal, P. J.; McKerrow, J. H.; Chibale, K. Synthesis and Structure−Activity Relationships of Parasiticidal Thiosemicarbazone Cysteine Protease Inhibitors against Plasmodium Falciparum , Trypanosoma Brucei , and Trypanosoma Cruzi. J. Med. Chem. 2004, 47 (12), 3212–3219.

(141) Chiyanzu, I.; Clarkson, C.; Smith, P. J.; Lehman, J.; Gut, J.; Rosenthal, P. J.; Chibale, K. Design, Synthesis and Anti-Plasmodial Evaluation in Vitro of New 4-Aminoquinoline Isatin Derivatives. Bioorg. Med. Chem. 2005, 13 (9), 3249–3261.

(142) Liu, Y.; Cui, K.; Lu, W.; Luo, W.; Wang, J.; Huang, J.; Guo, C. Synthesis and Antimalarial Activity of Novel Dihydro-Artemisinin Derivatives. Molecules 2011, 16 (12), 4527–4538.

(143) Schröder, J.; Noack, S.; Marhöfer, R. J.; Mottram, J. C.; Coombs, G. H.; Selzer, P. M. Identification of Semicarbazones, Thiosemicarbazones and Triazine Nitriles as Inhibitors of Leishmania Mexicana Cysteine Protease CPB. PloS One 2013, 8 (10), e77460.

(144) Schaeffer, M.; Schroeder, J.; Heckeroth, A. R.; Noack, S.; Gassel, M.; Mottram, J. C.; Selzer, P. M.; Coombs, G. H. Identification of Lead Compounds Targeting the Cathepsin B-Like Enzyme of Eimeria Tenella. Antimicrob. Agents Chemother. 2012, 56 (3), 1190– 1201.

(145) Limal, D.; Grand, V.; Vanderesse, R.; Marraud, M.; Aubry, A. The Semicarbazone Peptidomimetic Group in Imino Aza Peptides. Tetrahedron Lett. 1994, 35 (22), 3711– 3714.

(146) Adkison, K. K.; Barrett, D. G.; Deaton, D. N.; Gampe, R. T.; Hassell, A. M.; Long, S. T.; McFadyen, R. B.; Miller, A. B.; Miller, L. R.; Payne, J. A.; Shewchuk, L. M.; Wells- Knecht, K. J.; Willard, D. H.; Wright, L. L. Semicarbazone-Based Inhibitors of Cathepsin K, Are They Prodrugs for Aldehyde Inhibitors? Bioorg. Med. Chem. Lett. 2006, 16 (4), 978–983.

(147) Ito, A.; Takahashi, R.; Baba, Y. A New Method to Synthesize .ALPHA.- Aminoaldehydes. Chem. Pharm. Bull. (Tokyo) 1975, 23 (12), 3081–3087.

(148) Fonović, M.; Turk, B. Cysteine Cathepsins and Extracellular Matrix Degradation. Biochim. Biophys. Acta BBA - Gen. Subj. 2014, 1840 (8), 2560–2570.

(149) Mohamed, M. M.; Sloane, B. F. Cysteine Cathepsins: Multifunctional Enzymes in Cancer. Nat. Rev. Cancer 2006, 6 (10), 764–775.

(150) Gocheva, V. Distinct Roles for Cysteine Cathepsin Genes in Multistage Tumorigenesis. Genes Dev. 2006, 20 (5), 543–556.

680

(151) Bell-McGuinn, K. M.; Garfall, A. L.; Bogyo, M.; Hanahan, D.; Joyce, J. A. Inhibition of Cysteine Cathepsin Protease Activity Enhances Chemotherapy Regimens by Decreasing Tumor Growth and Invasiveness in a Mouse Model of Multistage Cancer. Cancer Res. 2007, 67 (15), 7378–7385.

(152) Berdowska, I. Cysteine Proteases as Disease Markers. Clin. Chim. Acta 2004, 342 (1-2), 41–69.

(153) Ullah, M. F.; Aatif, M. The Footprints of Cancer Development: Cancer Biomarkers. Cancer Treat. Rev. 2009, 35 (3), 193–200.

(154) Chen, B.; Platt, M. O. Multiplex Zymography Captures Stage-Specific Activity Profiles of Cathepsins K, L, and S in Human Breast, Lung, and Cervical Cancer. J. Transl. Med. 2011, 9 (1), 109.

(155) Jensen, A. B.; Wynne, C.; Ramirez, G.; He, W.; Song, Y.; Berd, Y.; Wang, H.; Mehta, A.; Lombardi, A. The Cathepsin K Inhibitor Odanacatib Suppresses Bone Resorption in Women With Breast Cancer and Established Bone Metastases: Results of a 4-Week, Double-Blind, Randomized, Controlled Trial. Clin. Breast Cancer 2010, 10 (6), 452– 458.

(156) Clézardin, P. Therapeutic Targets for Bone Metastases in Breast Cancer. Breast Cancer Res. 2011, 13 (2), 207.

(157) Rizoska, B.; Edenius, H.; Göhlin, K.; Hewitt, E.; Lindström, E.; Malcangio, M.; Pitcher, T.; Tunblad, K. MIV-247 A Cathepsin Protease Inhibitor Developed as a Novel Therapy for Neuropathic Pain, 2014.

(158) Shah, P. P.; Wang, T.; Kaletsky, R. L.; Myers, M. C.; Purvis, J. E.; Jing, H.; Huryn, D. M.; Greenbaum, D. C.; Smith, A. B.; Bates, P.; Diamond, S. L. A Small-Molecule Oxocarbazate Inhibitor of Human Cathepsin L Blocks Severe Acute Respiratory Syndrome and Ebola Pseudotype Virus Infection into Human Embryonic Kidney 293T Cells. Mol. Pharmacol. 2010, 78 (2), 319–324.

(159) Asaad, N.; Bethel, P. A.; Coulson, M. D.; Dawson, J. E.; Ford, S. J.; Gerhardt, S.; Grist, M.; Hamlin, G. A.; James, M. J.; Jones, E. V.; Karoutchi, G. I.; Kenny, P. W.; Morley, A. D.; Oldham, K.; Rankine, N.; Ryan, D.; Wells, S. L.; Wood, L.; Augustin, M.; Krapp, S.; Simader, H.; Steinbacher, S. Dipeptidyl Nitrile Inhibitors of Cathepsin L. Bioorg. Med. Chem. Lett. 2009, 19 (15), 4280–4283.

(160) Frizler, M.; Lohr, F.; Furtmann, N.; Kläs, J.; Gütschow, M. Structural Optimization of Azadipeptide Nitriles Strongly Increases Association Rates and Allows the Development of Selective Cathepsin Inhibitors. J. Med. Chem. 2011, 54 (1), 396–400.

(161) Bauer, D. J. Clinical Experience with the Antiviral Drug Marboran (1-Methylisatin 3- Thiosemicarbazone). Ann. N. Y. Acad. Sci. 1965, 130 (1), 110–117.

681

(162) Bauer, D. J.; St Vincent, L.; Kempe, C. H.; Young, P. A.; Downie, A. W. Prophylaxis of Smallpox with Methisazone. Am. J. Epidemiol. 1969, 90 (2), 130–145.

(163) Weiss, M. M.; Weiss, P. D.; Mathisen, G.; Guze, P. Rethinking Smallpox. Clin. Infect. Dis. 2004, 39 (11), 1668–1673.

(164) Caminero, J. A.; Sotgiu, G.; Zumla, A.; Migliori, G. B. Best Drug Treatment for Multidrug-Resistant and Extensively Drug-Resistant Tuberculosis. Lancet Infect. Dis. 2010, 10 (9), 621–629.

(165) Johnson, J. E.; Morales, N. M.; Gorczyca, A. M.; Dolliver, D. D.; McAllister, M. A. Mechanisms of Acid-Catalyzed Z / E Isomerization of Imines. J. Org. Chem. 2001, 66 (24), 7979–7985.

(166) Karabatsos, G. J.; Vane, F. M.; Taller, R. A.; Hsi, N. Structural Studies by Nuclear Magnetic Resonance. VIII. Ring-Substituted Phenylhydrazones, Semicarbazones, and Thiosemicarbazones. J. Am. Chem. Soc. 1964, 86 (16), 3351–3357.

(167) Kerek, F.; Ostrogovich, G.; Simon, Z. Mechanism of the Uncatalysed Syn?anti- Isomerization of Imine Systems. Part IV. A Theoretical Study of the Influence of Substituents. J. Chem. Soc. B Phys. Org. 1971, 541.

(168) Ostrogovich, G.; Simon, Z.; Kerek, F. Stereochemistry of Imine Systems. III. Mechanism of Their Uncatalysed Activation Energy. Rev. Roum. Chim. 1970, 15, 1453–1469.

(169) Curtin, D. Y.; Grubbs, E. J.; McCarty, C. G. Uncatalyzed Syn-Anti Isomerization of Imines, Oxime Ethers, and Haloimines 1. J. Am. Chem. Soc. 1966, 88 (12), 2775–2786.

(170) Hardegger, L. A.; Kuhn, B.; Spinnler, B.; Anselm, L.; Ecabert, R.; Stihle, M.; Gsell, B.; Thoma, R.; Diez, J.; Benz, J.; Plancher, J.-M.; Hartmann, G.; Banner, D. W.; Haap, W.; Diederich, F. Systematic Investigation of Halogen Bonding in Protein-Ligand Interactions. Angew. Chem. Int. Ed. 2011, 50 (1), 314–318.

(171) Albini, A.; Iwamoto, Y.; Kleinman, H. K.; Martin, G. R.; Aaronson, S. A.; Kozlowski, J. M.; McEwan, R. N. A Rapid in Vitro Assay for Quantitating the Invasive Potential of Tumor Cells. Cancer Res. 1987, 47 (12), 3239–3245.

(172) Wang, M.; Stearns, M. E. Isolation and Characterization of PC-3 Human Prostatic Tumor Sublines Which Preferentially Metastasize to Select Organs in S.C.I.D. Mice. Differ. Res. Biol. Divers. 1991, 48 (2), 115–125.

(173) Neve, R. M.; Chin, K.; Fridlyand, J.; Yeh, J.; Baehner, F. L.; Fevr, T.; Clark, L.; Bayani, N.; Coppe, J.-P.; Tong, F.; Speed, T.; Spellman, P. T.; DeVries, S.; Lapuk, A.; Wang, N. J.; Kuo, W.-L.; Stilwell, J. L.; Pinkel, D.; Albertson, D. G.; Waldman, F. M.; McCormick, F.; Dickson, R. B.; Johnson, M. D.; Lippman, M.; Ethier, S.; Gazdar, A.; Gray, J. W. A Collection of Breast Cancer Cell Lines for the Study of Functionally Distinct Cancer Subtypes. Cancer Cell 2006, 10 (6), 515–527.

682

(174) Zajc, I.; Sever, N.; Bervar, A.; Lah, T. T. Expression of Cysteine Peptidase Cathepsin L and Its Inhibitors Stefins A and B in Relation to Tumorigenicity of Breast Cancer Cell Lines. Cancer Lett. 2002, 187 (1-2), 185–190.

(175) Tamai, M.; Yokoo, C.; Murata, M.; Oguma, K.; Sota, K.; Sato, E.; Kanaoka, Y. Efficient Synthetic Method for Ethyl (+)-(2S,3S)-3-[(S)-3-Methyl- 1-(3- Methylbutylcarbamoyl)butylcarbamoyl]-2-Oxiranecarb Oxylate (EST), a New Inhibitor of Cysteine Proteinases. Chem. Pharm. Bull. (Tokyo) 1987, 35 (3), 1098–1104.

(176) Mitchell, D.; Lukeman, M.; Lehnherr, D.; Wan, P. Formal Intramolecular Photoredox Chemistry of Meta-Substituted Benzophenones. Org. Lett. 2005, 7 (15), 3387–3389.

(177) Zhao, W.; Carreira, E. M. Synthesis and Photochromism of Novel Phenylene-Linked Photochromic Bispyrans. Org. Lett. 2006, 8 (1), 99–102.

(178) Ye, S.; Chen, J.; Di, C.; Liu, Y.; Lu, K.; Wu, W.; Du, C.; Liu, Y.; Shuai, Z.; Yu, G. Phenyl-Substituted Fluorene-Dimer Cored Anthracene Derivatives: Highly Fluorescent and Stable Materials for High Performance Organic Blue- and White-Light-Emitting Diodes. J. Mater. Chem. 2010, 20 (16), 3186.

(179) Sharghi, H.; Jokar, M.; Doroodmand, M. M.; Khalifeh, R. Catalytic Friedel-Crafts Acylation and Benzoylation of Aromatic Compounds Using Activated Hematite as a Novel Heterogeneous Catalyst. Adv. Synth. Catal. 2010, 352 (17), 3031–3044.

(180) Blicke, F. F.; Patelski, R. A. The Action of Aluminum Chloride on the Diphenyl Ester of Isophthalic, Terephthalic and Naphthalic Acids. J. Am. Chem. Soc. 1938, 60 (10), 2283– 2285.

(181) Miller, T. M.; Kwock, E. W.; Neenan, T. X. Synthesis of Four Generations of Monodisperse Aryl Ester Dendrimers Based on 1,3,5-Benzenetricarboxylic Acid. Macromolecules 1992, 25 (12), 3143–3148.

(182) S, R. R.; Michael, S. Photoresponsive Sunscreen Compositions. WO1999007336 A1, February 18, 1999.

(183) KNUDSEN, S. M.; Pettersson, I.; Behrens, C.; Petersen, A. K.; Lau, J.; Garibay, P. W. Condensed Thiophene Derivatives and Their Use as Cyclic Glp-1 Agonists. EP1765814 A1, March 28, 2007.

(184) Chang, L. L. Triaryl Substituted Imidazoles and Methods of Use. WO1998022108 A1, May 28, 1998.

(185) Barrett, A. J.; Kirschke, H. [41] Cathepsin B, , and Cathepsin L. In Methods in Enzymology; Elsevier, 1981; Vol. 80, pp 535–561.

(186) Overgaard, K.; Overgaard, J. Investigations on the Possibility of a Thermic Tumour therapy—I. Eur. J. Cancer 1965 1972, 8 (1), 65–78.

683

(187) Overgaard, J. Simultaneous and Sequential Hyperthermia and Radiation Treatment of an Experimental Tumor and Its Surrounding Normal Tissue in Vivo. Int. J. Radiat. Oncol. Biol. Phys. 1980, 6 (11), 1507–1517.

(188) Horsman, M. R.; Murata, R.; Breidahl, T.; Nielsen, F. U.; Maxwell, R. J.; Stødkiled- Jørgensen, H.; Overgaard., J. Combretastatins Novel Vascular Targeting Drugs for Improving Anticancer Therapy. In Angiogenesis; Maragoudakis, M. E., Ed.; Springer US: Boston, MA, 2000; Vol. 476, pp 311–323.

(189) Monks, A.; Scudiero, D.; Skehan, P.; Shoemaker, R.; Paull, K.; Vistica, D.; Hose, C.; Langley, J.; Cronise, P.; Vaigro-Wolff, A.; Gray-Goodrich, M.; Campbell, H.; Mayo, J.; Boyd, M. Feasibility of a High-Flux Anticancer Drug Screen Using a Diverse Panel of Cultured Human Tumor Cell Lines. JNCI J. Natl. Cancer Inst. 1991, 83 (11), 757–766.

(190) Vichai, V.; Kirtikara, K. Sulforhodamine B Colorimetric Assay for Cytotoxicity Screening. Nat. Protoc. 2006, 1 (3), 1112–1116.

(191) MacDonough, M. T.; Strecker, T. E.; Hamel, E.; Hall, J. J.; Chaplin, D. J.; Trawick, M. L.; Pinney, K. G. Synthesis and Biological Evaluation of Indole-Based, Anti-Cancer Agents Inspired by the Vascular Disrupting Agent 2-(3′-Hydroxy-4′-Methoxyphenyl)-3- (3″,4″,5″-Trimethoxybenzoyl)-6-Methoxyindole (OXi8006). Bioorg. Med. Chem. 2013, 21 (21), 6831–6843.

(192) Skehan, P.; Storeng, R.; Scudiero, D.; Monks, A.; McMahon, J.; Vistica, D.; Warren, J. T.; Bokesch, H.; Kenney, S.; Boyd, M. R. New Colorimetric Cytotoxicity Assay for Anticancer-Drug Screening. JNCI J. Natl. Cancer Inst. 1990, 82 (13), 1107–1112.

(193) Sleeman, J.; Steeg, P. S. Cancer Metastasis as a Therapeutic Target. Eur. J. Cancer 2010, 46 (7), 1177–1180.

(194) Steeg, P. S. Tumor Metastasis: Mechanistic Insights and Clinical Challenges. Nat. Med. 2006, 12 (8), 895–904.

(195) Lah, T. T.; Durán Alonso, M. B.; Van Noorden, C. J. F. Antiprotease Therapy in Cancer: Hot or Not? Expert Opin. Biol. Ther. 2006, 6 (3), 257–279.

(196) Bell-McGuinn, K. M.; Garfall, A. L.; Bogyo, M.; Hanahan, D.; Joyce, J. A. Inhibition of Cysteine Cathepsin Protease Activity Enhances Chemotherapy Regimens by Decreasing Tumor Growth and Invasiveness in a Mouse Model of Multistage Cancer. Cancer Res. 2007, 67 (15), 7378–7385.

(197) Teno, N.; Miyake, T.; Ehara, T.; Irie, O.; Sakaki, J.; Ohmori, O.; Gunji, H.; Matsuura, N.; Masuya, K.; Hitomi, Y.; Nonomura, K.; Horiuchi, M.; Gohda, K.; Iwasaki, A.; Umemura, I.; Tada, S.; Kometani, M.; Iwasaki, G.; Cowan-Jacob, S. W.; Missbach, M.; Lattmann, R.; Betschart, C. Novel Scaffold for Cathepsin K Inhibitors. Bioorg. Med. Chem. Lett. 2007, 17 (22), 6096–6100.

684

(198) Stella, V. J. Prodrugs: Some Thoughts and Current Issues. J. Pharm. Sci. 2010, 99 (12), 4755–4765.

(199) Silverberg, L. J.; Dillon, J. L.; Vemishetti, P. A Simple, Rapid and Efficient Protocol for the Selective Phosphorylation of Phenols with Dibenzyl Phosphite. Tetrahedron Lett. 1996, 37 (6), 771–774.

(200) Carradori, S.; Cirilli, R.; Dei Cicchi, S.; Ferretti, R.; Menta, S.; Pierini, M.; Secci, D. 3- Methylcyclohexanone Thiosemicarbazone: Determination of E/Z Isomerization Barrier by Dynamic High-Performance Liquid Chromatography, Configuration Assignment and Theoretical Study of the Mechanisms Involved by the Spontaneous, Acid and Base Catalyzed Processes. J. Chromatogr. A 2012, 1269, 168–177.

(201) Tenchiu (Deleanu), A.-C.; Kostas, I. D.; Kovala-Demertzi, D.; Terzis, A. Synthesis and Characterization of New Aromatic Aldehyde/ketone 4-(β-D- Glucopyranosyl)thiosemicarbazones. Carbohydr. Res. 2009, 344 (11), 1352–1364.

(202) Pessôa, M. M. .; Andrade, G. F. .; Paoli Monteiro, V. R.; Temperini, M. L. . 2- Formylpyridinethiosemicarbazone and Methyl Derivatives: Spectroscopic Studies. Polyhedron 2001, 20 (26-27), 3133–3141.

(203) Kumar, G. D. K.; Chavarria, G. E.; Charlton-Sevcik, A. K.; Yoo, G. K.; Song, J.; Strecker, T. E.; Siim, B. G.; Chaplin, D. J.; Trawick, M. L.; Pinney, K. G. Functionalized Benzophenone, Thiophene, Pyridine, and Fluorene Thiosemicarbazone Derivatives as Inhibitors of Cathepsin L. Bioorg. Med. Chem. Lett. 2010, 20 (22), 6610–6615.

(204) Chavarria, G. E.; Horsman, M. R.; Arispe, W. M.; Kumar, G. D. K.; Chen, S.-E.; Strecker, T. E.; Parker, E. N.; Chaplin, D. J.; Pinney, K. G.; Trawick, M. L. Initial Evaluation of the Antitumour Activity of KGP94, a Functionalized Benzophenone Thiosemicarbazone Inhibitor of Cathepsin L. Eur. J. Med. Chem. 2012, 58, 568–572.

(205) Chavarria, G. E. Kinetic Characterization of Thiosemicarbazones as Cysteine Protease Inhibitors and Their Potential Use as Therapeutic Agents against Metastatic Cancer and Chagas’ Disease. Ph.D. Dissertation, Baylor University, Waco, TX, 2012.

(206) Mori, A.; Mizusaki, T.; Miyakawa, Y.; Ohashi, E.; Haga, T.; Maegawa, T.; Monguchi, Y.; Sajiki, H. Chemoselective Hydrogenation Method Catalyzed by Pd/C Using Diphenylsulfide as a Reasonable Catalyst Poison. Tetrahedron 2006, 62 (51), 11925– 11932.

(207) Liu, Q.; Li, J.; Shen, X.-X.; Xing, R.-G.; Yang, J.; Liu, Z.; Zhou, B. Hydrogenation of Olefins Using Hantzsch Ester Catalyzed by Palladium on Carbon. Tetrahedron Lett. 2009, 50 (9), 1026–1028.

685