. .. .fi ‘ 424.1,- :2 ‘ 4 3. Rania... ... ab... 21‘; u 0.. '5. ‘4 I». .9! .o 5.... 2.3103}! 1:330} LIE‘" ‘ "-‘Y Michi: . State University This is to certify that the dissertation entitled ACCESSING THE 2-AMINO-5,5-D|SUBST|TUTED-1H-IMIDAZOL-4-ONE SCAFFOLD FOR NATURAL PRODUCT SYNTHESIS AND EVALUATION FOR CHECKPOINT KINASE 2 INHIBITION presented by Christopher David Hupp has been accepted towards fulfillment of the requirements for the Ph.D. degree in Chemistry m \- Majorfiofessor’s Signature 6—Ol—oq] Date MSU is an Affirmative Action/Equal Opportunity Employer PLACE IN RETURN BOX to remove this checkout from your record. TO AVOID FINES return on or before date due. MAY BE RECALLED with earlier due date if requested. DATE DUE DATE DUE DATE DUE 5/08 K:/Prolecc&Pres/CIRC/DaleDue.indd ACCESSING THE 2-AMINO-5,5-DISUBSTITUTED-1H-IMIDAZOL—4-ONE SCAFFOLD FOR NATURAL PRODUCT SYNTHESIS AND EVALUATION FOR CHECKPOINT KINASE 2 INHIBITION By Christopher David Hupp A DISSERTATION Submitted to Michigan State University in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY Chemistry 2009 ~"I-Aa l ""‘v‘\. t“'* an a“. v a.“ A._ y, ””1 I I.‘H‘ h - VI“ (to (I'- N- 5 .:\\i (I, ABSTRACT ACCESSING THE 2—AMINO-5,5-DlSUBSTITUTED-1H-lMIDAZOL-4-ONE SCAFFOLD FOR NATURAL PRODUCT SYNTHESIS AND EVALUATION FOR CHECKPOINT KINASE 2 INHIBITION By Christopher David Hupp This dissertation is primarily focused on the development of a synthetic method for the preparation of 2-amino-5,5-disubstituted-1H-imidazol-4-ones through a novel rearrangement and the application toward the first total synthesis of a marine alkaloid. The first chapter identifies checkpoint kinase 2 (Chk2) as a viable drug target for adjuvant therapeutics. The second chapter elaborates on the development of a new rearrangement applicable for the synthesis of potential Chk2 inhibitors. The third chapter describes the total syntheses of a natural product and two analogs. Finally, the fourth chapter describes the biological evaluation of all synthesized compounds. Chemotherapy and radiation therapy offer effective methods to treat various forms of cancer. However, these procedures can often lead to negative side effects resulting in the destruction of healthy cells. Developing adjuvant drugs that inhibit Chk2, a kinase part of the DNA damage response network, is believed to help ameliorate these harmful side effects by desensitizing healthy cells toward treatments such as ionizing radiation. lndoloazepine, an analog of the natural product debromohymenialdisine, was found to be a potent inhibitor of Chk2. Additional literature evaluation led to the identification of a natural product, from the tunicate Dendrodoa grossu/aria, v N" ‘1‘ - v' R. uyA, l : U‘e ";\»1p. 5 I y ‘ “A"‘Br -F A “ In C m “c. " v»! T «ah ft "U‘ I“ 5“. ‘ ' Hz? N V‘ .U ’. ~;PP;F :- "‘ ,4 II‘ I’ which contained structural similarities to indoloazepine. The indole alkaloid contained a unique quaternary imidazolone scaffold that required the use of a novel transformation to attain. The traditional oxazole rearrangement was discovered by Prof. Steglich in 1975. Unfortunately, this transformation is often plagued by the inability to remove an N-acyl group resulting from the ring-opened oxazolone product, thus preventing any further chemical modifications. In order to apply an oxazole rearrangement to the synthesis of the natural product, a novel rearrangement was developed. Overall, a one-pot transformation from a thiourea to a quaternary hydantoin intermediate allowed access to the imidazolone scaffold needed to complete the first total synthesis of the natural product. The natural product was synthesized in 12 linear steps with an overall yield of 11.8%. The synthesis highlighted the use of the newly developed rearrangement and also allowed for analogs to be synthesized without a major modification of the synthetic pathway. Finally, biological evaluation of the natural product, analogs and other heterocycles revealed that all compounds prepared were inactive against the targets screened. Additionally, all compounds tested for cell cytotoxicity were found to be non-cytotoxic. This dissertation is dedicated to my parents, David and Sandra Hupp, who have always made my education a top priority. I told you I’d become a doctor someday! I‘n pl 1 «Jay ‘ ‘M u \ Vt ”it“ a PM» (1’ {v (I) ACKNOWLEDGMENTS I would first like to acknowledge my advisor, Prof. Jetze Tepe. It was truly a daunting experience to come into a large university from a small undergraduate college. Jetze showed patience (yes, he does Show that quality sometimes) throughout the first years of me learning the ropes in lab and encouraged me to not just do chemistry, but understand it. I believe he always had faith in me to complete my project even though at times the road was very bumpy and the outcome looked bleak. It is with his faith and my determination that pushed me to reach my goal. I truly learned a great deal about organic synthesis and medicinal chemistry but, overall, I believe the most valuable things I learned in his lab were the lessons on dealing with struggles in lab and how to approach a problem. For all of this, I thank you Jetze. I would also like to acknowledge Prof. Robert Maleczka, my second reader. By far, he is easily one of the best professors I have had for an Organic Chemistry course. Although his class was the hardest I have ever taken, it is also the class where I learned the most. I will always remember that class and what it has done for me on an intellectual level. I also want to acknowledge my other committee members, Professors William Wulff and Jim McCusker, whom have been an excellent source of information and guidance throughout my graduate career. I want to extend a sincere thanks to other staff at MSU including Bob Rasico, Melissa Parsons, Bill Flick, Dr. Daniel Holmes and Kermit Johnson. The Aq:~‘j » - ' L ‘1‘: ( Ii .. My U ‘ I. I" i) . "a ”A“ \ VL' i: "v la. .5.“ ~ - U ~11 E ,‘v- “u chemistry department is very lucky to have staff such as these people, who were always willing to lend a hand. MSU is truly blessed with such talented and thoughtful staff. The past and present Tepe group members have also shaped the type of scientist I have become and for that, I am very grateful. I wouldn’t be Where I am today without the help from all of the group members. It is not every day that co- workers and great friends coincide. I am blessed to have become friends with many of the people I worked with including Jason, Sam, Thu and Adam. It has made the whole process of graduate school much easier and I hope to stay in touch in the future. Lastly, but certainly not least, I would like to thank my wife Amber and my family. I am lucky to have such a wonderful and understanding wife as Amber. I am truly nobody without her. I want to thank her for being supportive for the late night study sessions, Saturday classes and every hoop we had to jump through for graduate school. I want to also thank my parents, Dave and Sandy, for giving me the means to achieve my goals and always believing in me. I also want to thank my in-Iaws, Tom and Jille, for all their support and encouragement over the years. They have treated me like a son and for that, I am grateful. It’s not everyday that you get two sets of great parents!! I would also like to thank my sister, Amy, who keeps me grounded by reminding me that I’m still JUST her big brother. Finally, I would like to extend a big thank you to all of my family who has been a source of comfort and relaxation throughout the past six years. I truly could not have done this without all of you. vi TABLE OF CONTENTS LIST OF TABLES ............................................................................................... ix LIST OF FIGURES ............................................................................................... x LIST OF SCHEMES ........................................................................................... xii LIST OF ABBREVIATIONS ............................................................................... xv CHAPTER I. CHECKPOINT KINASE 2 AS A TARGET FOR ADJUVANT CANCER THERAPEUTICS ......................................................... 1 LA. Introduction to cancer ......................................................................... 1 LB. Chemo and radiotherapy .................................................................... 3 LC. DNA damage response pathways ...................................................... 6 ID. Developing adjuvant therapeutics for cancer treatment ...................... 7 LE. Chk2 as drug target candidate ............................................................ 9 LP. Validation of Chk2 as drug target ..................................................... 15 LG. Purified drug target and X-ray crystal structure ................................ 17 I.H. Checkpoint kinase 2 inhibitors .......................................................... 19 Li. Chk2 binding pocket properties ........................................................ 23 I.J. Goal of project .................................................................................. 30 I.K. References ....................................................................................... 31 CHAPTER II. DEVELOPMENT OF A SYNTHETIC METHOD FOR THE PREPARATION OF 2-AMlNO-5,5-DISUBSTITUTED-1H- IMIDAZOL-4(5H)-ONES ............................................................ 43 "A. Nomenclature and numbering of imidazolones ................................. 43 ".8. Reactivity of imidazolones ................................................................ 45 MC. Natural products containing the imidazolone core ............................ 48 MD. Synthetic routes to imidazolones ...................................................... 53 II.D.1. Synthetic routes to imidazolones: Aza-wittig reaction ............... 54 ".02. Synthetic routes to imidazolones: Reaction of aziridinone and thiourea .................................................................................... 54 ".03 Synthetic routes to imidazolones: Oxazolone to imidazolone... 55 "DA. Synthetic routes to imidazolones: Oxidation of imidazoles ....... 56 II.D.5. Synthetic routes to imidazolones: Diketone rearrangement ..... 57 II.D.5.a. Synthetic routes to imidazolones: Using diketone rearrangement to produce imidazolone analogs of natural product ............................................................................... 61 II.D.6. Synthetic routes to imidazolones: Dehydration reactions ......... 65 "DJ. Synthetic routes to imidazolones: Cyclization with carbodiimide intermediates .............................. vii .............................................. 66 IIF. II. II 13. Hit III. CHAPTEI INA, IIIE lllc III) I: a, HIF. IIIG IIIH III I. CHAPTE IfA WE II”! C II.D.8. Synthetic routes to imidazolones: Cyclization reactions with nitriles ....................................................................................... 69 II.D.9. Synthetic routes to imidazolones: Cyclization reactions with N- cyanoguanidines ...................................................................... 71 NE. Synthesis of quaternary intermediates for the synthesis of the natural product ............................................................................................. 72 HF. Development of a novel EDCl-mediated oxazole rearrangement ..... 81 l|.F.1. Scope of rearrangement ........................................................... 84 lI.F.2. Proposed reaction mechanism ................................................. 91 ".6. General experimental information ..................................................... 95 "H. Experimental procedures and characterization ................................. 96 NJ. References ..................................................................................... 145 CHAPTER III. TOTAL SYNTHESIS OF AN INDOLE ALKALOID FROM ||l.A. l||.B. III.C. |||.D. III.E. |I|.F. ||I.G. III.H. l|I.|. THE TUNICATE DENDRODOA GROSSULARIA AND TWO ANALOGS ............................................................................... 157 Pharmacological value of marine compounds ................................ 157 Background on Dendrodoa grossu/aria .......................................... 158 Retrosynthetic analysis of indole alkaloid III-1 ................................ 162 Synthesis of indole alkaloid III-1 ..................................................... 163 Retrosynthetic analysis of analog 1 and 2 ...................................... 168 Synthesis of analogs 1 and 2 .......................................................... 169 General experimental information ................................................... 172 Experimental procedures and characterization ............................... 173 References ..................................................................................... 1 86 CHAPTER IV. BIOLOGICAL TESTING OF NATURAL PRODUCT, IV.A. NB. NC. ND. NE. IV.F. IV.G. IV.H. ANALOGS AND OTHER HETEROCYCLIC COMPOUNDS 189 Biological target of natural product and hypothesis for activity ....... 189 Kinase screen for natural product and analogs .............................. 190 Other biological testing for natural product and analogs ................. 191 Synthesis of additional heterocycles ............................................... 192 Biological testing for additional heterocycles .................................. 195 General experimental information ................................................... 196 Experimental procedures and characterization ............................... 197 References ..................................................................................... 209 viii n‘.’ Iazell. Table I-1 Table ”-1 Table "-2 Table ”—3 Table III-1 Table lV-1 Table IV-2 Table IV-3 LIST OF TABLES FDA-approved kinase inhibitors ................................................. 5 Oxazole rearrangement with various N-acyl groups ................. 79 Rearrangement containing various R groups ........................... 88 Rearrangement containing various allyl groups ........................ 91 Comparison of spectral data from synthesized natural product (III-1) and isolated natural product .......................................... 167 Results of kinase screen for III-1, Ill-16 and III-19 .................. 191 Results for cytotoxicity, NF-kB and lL-6 assays ..................... 192 Biological activity results for lV-1, lV-3, IV-6, IV-9 and lV-11.. 195 ’ l l . D pp |. Id v ' - D I F, .0 ‘u- I F . Ind u L I. .04 . Iii-fl div v . I -“'fl.l ~'« Idv'~ . I 5‘ A. .- ." 1.4. i n :A 5". ‘-..h :led . ~1p;'7 :" ., d a I u D La , 'A .I»EI~;I D -~'Al1 H :v«. O 55. -Pa 4 :‘W‘I . '“I M ‘ H 3" v '1 -. PM Figure l-1 Figure l-2 Figure I-3 Figure I-4 Figure l-5 Figure l-6 Figure l-7 Figure l-8 Figure I-9 Figure l-1O Figure H 1 Figure l-12 Figure l-13 Figure l-14 Figure l-15 Figure l-16 Figure I-17 Figure l-18 Figure l-19 Figure "-1 Figure "-2 LIST OF FIGURES Normal cell division .................................................................... 2 Cancer cell division .................................................................... 3 DNA damage response pathways .............................................. 7 Schematic for drug development ................................................ 9 Components of the damage response network ........................ 13 DNA damage pathway involving Chk2 ..................................... 16 Illustration of Chk2 and crystal structure of kinase domain ...... 18 Squaric acid derivatives and indazoles as Chk2 inhibitors ....... 19 Chk2 inhibitors from compound libraries .................................. 20 Benzimidazole and non-benzimidazole Chk2 inhibitors ........... 21 Natural products and analogs used as Chk2 inhibitors ............ 22 DBH inside the Chk2 active site ............................................... 23 Interactions between southern portion of DBH and Chk2 ......... 24 Indoloazepine modeled inside the Chk2 binding pocket ........... 25 New indole alkaloid and indoloazepine .................................... 26 Comparison of dihedral angles ................................................. 27 Hybridization of carbon connected to indole ring ...................... 28 New indole alkaloid modeled in Chk2 binding pocket ............... 29 Oxazolone chemistry affording various heterocycles ............... 30 Numbering of the 2-amino-imdazol-4-one ring system ............. 44 Representative structures of imidazolones ............................... 44 Figure “-3 Figure "-4 Figure "-5 Figure “-6 Figure "-7 Figure ”-8 Figure III-1 Figure III-2 Figure IV-1 Figure lV-2 Reactions of 2-aminoimidazolone ............................................ 45 Relative rates of deuterium exchange ...................................... 48 pKa’s of differently substituted 2-aminoimidazolones ............... 48 Natural products containing the 2-aminoimidazolone core ....... 50 Natural products containing an imidazolone core ..................... 51 Additional natural products containing 2-aminoimidazolone ..... 52 Alkaloids from Dendrodoa grossu/aria .................................... 158 X-ray crystal structure of indole alkaloid III-1 .......................... 168 The indole natural product III-1 and indoloazepine ................. 189 Kinase screen performed by Millipore .................................... 190 xi Scheme I-1 Scheme "-1 Scheme ”-2 Scheme "-3 Scheme "-4 Scheme ”-5 Scheme ”-6 Scheme "-7 Scheme "-8 Scheme "-9 Scheme "-10 Scheme "-1 1 Scheme "-12 Scheme "-13 Scheme "-14 Scheme "-15 Scheme "-16 Scheme "-17 Scheme "-18 Scheme "-19 Scheme "-20 LIST OF SCHEMES DNA damage by UV and IR ...................................................... 11 Alkylation of 2-aminoimidazoI-4-one ......................................... 46 Alkylation at C-5 of an imidazolone .......................................... 47 Aza-wittig reaction to produce imidazolones ............................ 54 Reaction of aziridinones and thiourea to form imidazolones 55 lmidazolones using oxazolones and thioureas ......................... 56 Oxidation of imidazoles to form imidazolones .......................... 57 lmidazolones from 1, 2-diketone and guanidine ....................... 58 Proposed mechanism for diketone rearrangement ................... 59 Alternative mechanism for diketone rearrangement ................. 60 Synthesis of imidazolone II-18 .................................................. 62 Synthesis of imidazolone II-23 .................................................. 64 lmidazolones produced by dehydration .................................... 65 Cyclization from diimide: urea to imidazolone .......................... 67 Cyclization from diimide: thiourea to imidazolone ..................... 67 Cyclization from diimide: azide to imidazolone ......................... 68 Intramolecular cyclization: thiourea to imidazolone .................. 69 Intramolecular cyclization: urea to imidazolone ........................ 69 Imidazolone through cyclization onto nitrile .............................. 70 Imidazolone through cyclization using N-cyanoguanidines ...... 71 Rearrangement developed by Steglich .................................... 73 xii .n-Mfl — ewwl A ‘FR~I .- vdv v \_ vvv V A . Annmfi' .- ‘ Henna ‘ _ Uri Uh ’ I .- .‘ :N‘Q v 5 Vi \ "_‘_m: V. \- I». I . \ ”:3“; In. , U I \"“'!-Al . I‘ v. ' U I. \.‘;Ma r. ' v 1V n \‘n-w‘ v. " Q ‘V A \"-~. c/ D t —P ",4 (D J ( I) (I) ") I Scheme "-21 Scheme "-22 Scheme ”-23 Scheme "-24 Scheme "-25 Scheme "-26 Scheme ”-27 Scheme "-28 Scheme ”-29 Scheme "-30 Scheme "-31 Scheme "-32 Scheme "-33 Scheme "-34 Scheme Ill-1 Scheme Ill-2 Scheme Ill-3 Scheme Ill-4 Scheme Ill-5 Scheme Ill-6 Scheme III-7 Scheme III-8 Scheme IV-1 Rearrangement leading to 4-fluoropyridines ............................ 74 Rearrangement affording d-trifluoromethyl a-amino acids ....... 75 Rearrangement leading to Il-26-lI-28 ....................................... 75 Rearrangement to synthesize C-glycosyl o-amino acids .......... 76 Unexpected reaction producing hydantoin II-41 ....................... 8O Unexpected reaction producing hydantoin Il-42 ....................... 81 Routes from hydantoin to imidazolone ..................................... 82 New rearrangement applied to the synthesis of III-1 ................ 84 General scheme for novel rearrangement ................................ 85 General synthesis of thioureas ll-63-ll-69 ................................ 86 Synthesis of thiourea lI-75 ........................................................ 87 Potential byproduct pathway .................................................... 89 Synthesis of higher substituted allylic thioureas ....................... 90 Proposed mechanism of EDCl-mediated rearrangement ......... 93 Synthesis of derivative Ill-4 .................................................... 161 Retrosynthesis of indole alkaloid Ill-1 ..................................... 163 Synthesis of thiourea III-9 ....................................................... 164 Synthesis of hydantoin III-1O .................................................. 165 Synthesis of alkaloid III-1 ....................................................... 166 Retrosynthesis of analog 1 (Ill-16) and analog 2 (Ill-19) ........ 169 Synthesis of analog 1 (III-16) .................................................. 170 Synthesis of analog 2 (III-19) ................................................. 171 Synthesis of IV-1 and lV-3 ...................................................... 193 xiii “Ema! ..., g V I! Scheme IV-2 Synthesis of hydantoin lV-6 .................................................... 193 Scheme IV-3 Synthesis of IV-11 .................................................................. 194 xiv LIST OF ABBREVIATIONS AIDS — acquired immune deficiency syndrome Asn — asparagine ATM - ataxia telangiectesia mutated ATP — adenosine triphosphate ATR - ataxia telangiectesia related Bn - benzyl Boc - tert-butyloxycarbonyl BOP - Benzotriazole-l-yl-oxy-tris-(dimethylamino)-phosphonium hexafluorophosphate Cbz - carbobenzyloxy Cdc — cell division cycle CDK — cyclin dependent kinase Chk# - checkpoint kinase # DBH - debromohymenialdisine DCC — dicyclohexylcarbodiimide DCM — dichloromethane DIPEA — diisopropylethyl amine DMAP — dimethylamino pyridine DMDO — dimethyldioxirane DMEM - Dulbecco's modified eagle medium DMF - dimethylformamide DMSO — dimethyl sulfoxide XV DNA — deoxyribonucleic acid E050 — half maximal effective concentration EDCI - 1-EthyI-3-(3'-dimethylaminopropyl)carbodiimide hydrochloride EDTA - ethylenediaminetetraacetic acid ELISA - enzyme-linked immunosorbent assay Et — ethyl FBS — fetal bovine serum FDA — food and drug administration Fmoc - 9H-fluoren-9-ylmethoxycarbonyl Glu - glutamic acid GSK - glycogen synthase kinase HMDS — hexamethyl disilazane HPLC - high performance liquid chromatography HRMS — high resolution mass spectrometry IBX - 2-lodoxybenzoic acid leo - half maximal inhibitory concentration IL - interleukin IR — ionizing radiation or infrared Law. Rgt. - Lawesson's reagent LCMS - liquid chromatography mass spectrometry Lys - lysine Me - methyl Met - methionine xvi 141:2 _ l a :"n - Ia... ‘I-I I"‘.;_, I'll.A A ‘II‘J-I ,4 III: II |'\"'A'\'I ‘V~V I \ Rh b ‘ - \- Ugl I I. n J v ‘ Y“ Q: u," 85 'a 3:: IA 0 ‘ L“ V ‘\ ~_I ‘\H| \t TI- 5 MOPS - 3-morpholinopropanesulfonic acid MS — mass spectrometry Napth. - naphthalene NF-KB — nuclear transcription factor — kappa B NMO — N-methylmorpholine-N-oxide NMR — nuclear magnetic resonance NOESY - nuclear overhauser enhancement spectroscopy OAc — acetate Ph — phenyl PIKK - phosphoinositide 3-kinase-like kinase ppm - parts per million psi — pounds per square inch Pyr. — pyridine RNA - ribonucleic acid rt — room temperature SAR — structure activity relationship Ser - serine tBu — tert butyl TEA — triethylamine TFA - trifluoroacetic acid TFAA — trifluoroacetic anhydride THF - tetrahydrofu ran Thr - threonine xvii TLC — thin layer chromatography TNF — tumor necrosis factor Ts - tosyl UV — ultraviolet xviii 5" Rip It"; ' 'VI LI I ”hung, L:.:l U -I H: Int IM ‘ ‘tTSEft HA I I M,‘ , ' i A,‘ “dyv .l ' ‘I PM “ ‘ i CHAPTER I CHECKPOINT KINASE 2 AS A TARGET FOR ADJUVANT CANCER THERAPEUTICS l.A Introduction to cancer Despite the increase in knowledge and therapeutic advances, cancer is currently one of the highest causes of mortality in the United States.1' 2 Even though the mortality rate of some forms of cancer (stomach, breast and rectum cancer) have slowly decreased, other forms of cancer (pancreatic, lung and bronchus) have seen little improvements over the last several years.2' 3 Cancer can be defined as an unregulated growth of tissue not governed by the regulations of normal cell growth.4 The main causes of human cancer include tobacco/tobacco products (30%), hormones (30%), diet (15%), viruses (10%), drugs, x-rays and UV light (10%) and occupational carcinogens (5%).4 Normal cell growth and division is systematic and necessary to replace the aging and dying cells or to repair injuries. Furthermore, normal cell growth and division can only be conducted in a manner that is cognisant of a neighboring cell’s growth to efficiently and correctly form tissues and organs.4 The process of division for a normal cell iS shown below in Figure H. A healthy cell will grow and divide into two new daughter cells, which can each then grow and divide themselves, and so on. In the event that a cell is damaged in the process or a mutation has occurred, the cell can either repair itself or initiate cell suicide (apoptosis). The apoptotic pathway is essential for eliminating damaged DNA and preventing mutated cells from passing on the damaged genetic information.4 /v—_:_>@ Form tissues /'@<:©_——> _, and/or organs O”@ /.@Repa.r Normal Mutation Cell suicide cell \ © @<: Damage Figure l-1. Normal cell division However, if cells stop cooperating with their neighboring cells, become autonomous in their growth and are mutated, they can form tumor cells, which could later become cancer cells. Tumors are generally classified into two categories: benign and malignant. Benign tumors are localized, do not spread to other parts of the body and are generally not lethal to their host. However, if a benign tumor is exerting pressure on a sensitive organ such as the brain, then complications can arise. Malignant tumors can destroy parts of the body from which they originate and can invade surrounding tissue. Furthermore, these tumors can form secondary tumors in new sites and eventually cause destruction of additional tissue and organs. This large difference in mobility distinguishes cancer from benign growths.4 Malignant tumor cells, or cancer cells, grow uncontrollably and divide in an unordered fashion. As shown in Figure I-2 a cancer cells division process proceeds in a non-systematic way leading to non-structured masses or tumors. These cells can then spread or metastasize toward other tissues and eventually cause damage at sites far from the original location.4 C§r\@/ @ cell \A @ ' ' " Uncontrolled and unordered growth Figure I-2. Cancer cell division LB Chemo and radiotherapy Although uncontrolled cancerous cell growth can lead to solid tumors (except in cases such as leukemias), cancer can be treated by a variety of methods including chemotherapeutics and/or radiation therapy.5'18 There are many different types of chemotherapeutics used to treat cancer such as alkylating agents, antimetabolites, topoisomerase inhibitors, mitotic inhibitors as well as other targeted therapies. Alkylating agents are used to directly damage DNA by alkylating the base pairs and ultimately preventing the cell from reproducing. Some of the more common types of alkylating agents include the nitrogen mustards‘o, mitomycin C12 and the powerful platinum drugs cis-platin and carboplatin.5 Antimetabolites are a class of drugs that interfere with the growth of DNA and RNA by essentially mimicking the natural metabolites used for the synthesis of DNA and RNA. Common modes of action for antimetabolites include inhibition of DNA synthesis enzymes or causing mismatched DNA synthesis, which could result in cell death. A few common antimetabolites that are used for chemotherapy include 5-fluorouracil8 and 6-mercaptopurine.15 II ..--A g . ‘~\—g \ ..wu V -.A-'unn 5 j-hvll I” J u)‘ ‘ ——.. w ’ “'9 i-' p l w: 5," «1:!- Raf, "5 U5“; TT’IQI‘W "“J~U Topoisomerase inhibitors are compounds that interact with the enzymes called topoisomerase I and topoisomerase II, which are responsible for assisting in the unwinding of DNA for the replication process. Topoisomerase I perfoms a reversible single strand cleavage of DNA to allow relaxation of the coil and then reseals the cleaved strand after unwinding. Topoisomerase l inhibitors include, camptothecin, topotecan and irinotecan.13 The topoisomerase II enzyme acts in a similar fashion as the topoisomerase I enzyme except that it completes a reversible double stranded DNA cleavage to aid in the unwinding of DNA for replication followed by a resealing of the cleaved strands. The most common topoisomerase II inhibitors include etoposide, teniposide, doxorubicin and daunorubicin.9' 11' 17' ‘8 Anthracyclines, such as doxorubicin, intercalate between the base pairs of DNA resulting in a distortion that prevents the DNA replication process. Funhermore, metabolism of specific anthracyclines, such as doxorubicin, leads to the formation of free radicals that oxidatively damage DNA and to reactive intermediates that form covalent adducts with DNA, inhibiting the overall replication process.18 Mitotic inhibitors interfere with mitosis and prevent the cell from reproducing. One of the more famous mitotic inhibitors include paclitaxel (Taxol), a natural product isolated from the pacific yew tree.14 Paclitaxel’s mode of action is unique from other mitotic inhibitors due to its effect on the polymerization of tubulin, the building block of microtubules. In general, most mitotic inhibitors induce the disassembly of microtubules, a component of cells responsible for aiding in the cell division process. However, paclitaxel was found to induce the ‘6 \I ~_‘ av; polymerization of tubulin, producing dysfunctional microtubules leading to the death of the cell.14 As researchers have come to understand cancer more thoroughly, new more specific therapies have developed. Kinases have become an interesting and challenging target for cancer drug discovery in the past decade. Although extensive research has been accomplished trying to develop kinase inhibitors, only a few have been FDA approved in the last ten years (Table M).7 The main complication that arises from targeted kinase chemotherapy is the formation of drug resistant tumors. Table l-1. FDA-approved Kinase inhibitors (adapted from ref. 13) U.S. brand Year Cancer . name approved type Company Target kinase Abl, c-Kit, Gleevec 2001 CML Novartis PDGFRa, PDGFRb lressa 2003 NSCLC AstraZeneca EGFR Tarceva 2004 NSCLC, P Gegesnltgm EGFR Raf, VEGFRZ, Nexavar 2005 HC, RCC Bayer, Onyx Vifizfs’ PDFGRb c-Kit, VEGFR, Sutent 2006 GIST, RCC Pfizer PDGFR, FLT3 Abl, c-Kit, Sprycel 2006 CML BMS PDGFR, Src Abl, c-Kit, Tasigna 2007 CML Novartis PDSBrERb’ Ephthrin Tykerb 2007 BC GSK EGFR, Her-2 CML = chronic myeloid leukemia; NSCLC = non-small-cell lung cancer; P = pancreatic; HC = hepatocellular carcinoma; RCC = renal cell carcinoma; GIST = gastrointestinal stromal tumor; BC = breast cancer h- at .'-fi1 i S n r'zn ,‘ 't ~U‘l Spa---. 3" 5'». .I~h . .l “7 " VII\ J P Radiation therapy also offers a highly sophisticated treatment of cancer. The radiation can be administered externally by a machine, internally by a catheter or taken orally. The radiation induces unsustainable DNA damage to targeted cells which results in cell death.6' 16 Overall, one of the impediments of chemotherapeutic and ionizing radiation therapy is that these treatments are often limited in efficacy by severe side effects to healthy tissue. Strategies that address these limitations may have the potential to enhance the efficacy of current treatments.3 I.C DNA damage response pathways On average, the induction of DNA damage appears to be the most common strategy in anticancer treatment and has dramatically increased the survival rate of patients.19’22 The anticancer effects are even better when these agents are used in combination with other drugs containing different modes of action.23 Upon treatment of cells with any of the chemotherapeutics described previously or ionizing radiation, two distinct DNA damage response pathways are induced (Figure I-3).24 One response includes the activation of the NF-KB pathway, which is largely responsible for antiapoptotic cell signaling. The second distinct response is the activation of cell cycle checkpoint kinases, which essentially mediate the induction of cell cycle arrest and apoptosis.”31 Activation of the NF- KB signaling pathway induces the expression of a wide range of genes involved in cell survival responses.”35 Activation of the DNA checkpoint kinases results in a complex network of Signaling pathways involved in the induction of apoptosis or cell cycle arrest, which allows for DNA repair and re-initiation of cell cycle A h 3 "RI“ fl , IIOI 4 IF A. I. 'JII A... progression.“ 36"” Unfortunately, the activation of these antiapoptotic signaling pathways limits the overall efficacy of cancer treatments.3 CHEMOTHERAPEUTICS Ol' IONIZING RADIATION I DNA damage Cell cycle checkpoint kinases 2 I - ACTIVATION Inhibition Cell cycle arrest of or apoptosis Apoptosis (cell survival) Figure I-3. DNA damage response pathways Efficacy of treatment is often limited by the narrow therapeutic window in which drugs are effective due to their inherent toxicity to the patient. Strategies of combining traditional chemotherapeutics with cell cycle checkpoint inhibitors to either sensitize cancerous cells or desensitize healthy cells, may widen the therapeutic window and improve the overall efficacy of the cancer treatment.3 I.D Developing adjuvant therapeutics for cancer treatment Adjuvant therapeutics offer a strategy to limit the negative side effects of current cancer treatment practices. It is hypothesized that by targeting a specific I ‘ ‘ - FR ”IT." Ii "1‘ I. .t-up'v“ leR§~ n‘ val I .v-J V ‘ E u ~¢C Bier dJut U‘ \ V ‘ T’ N .a :v UL . 'C “ h ”to.” find 'I the checkpoint kinase, healthy cells can be selectively desensitized to radiation treatment and allow a more effective procedure to manage cancer. Developing a drug to become an adjuvant therapeutic is a long process as illustrated in Figure l-4.41 This section (ID) is meant to briefly explain each major step in the process (Figure I—4) and will be complemented by the application of this process to my project in the following sections. As Shown in Figure I-4, the first step in developing therapeutics is to identify a drug target candidate. After initial studies determine if the candidate is a viable option, the target is validated. Subsequently, efforts are made to 1) purify the drug target so that additional information can be gathered and 2) compounds from a variety of areas are screened to identify potential hits. If a crystal structure is identified for the target, it can aid in the development of additional compounds to test and other potential drug candidates through a rational drug design approach. If a lead compound is designed or found, the process of optimizing the lead drug can begin through molecular modeling, medicinal chemistry and combinatorial chemistry techniques. Once optimized, the drug is subjected through many rounds of preclinical studies and finally clinical studies before being considered for approval to go on the market. Drug target candidate] I Validated druflrfil II , Purified I —> Natural ProductSI—> Screeningl/ drug target Assays , Combinatoriall / - l libraries X-ray crystal I Compound structure ’ collections l * Rational drug design I I Drug leads I Optimized drug Ieadsl I Preclinical studies then clInIcal trials Figure I-4. Schematic for drug development (adapted from ref. 40) LE Chk2 as drug target candidate The primary structure of DNA is continuously being altered by endogenous and exogenous stimuli, which cause abnormalities ranging from simple base changes to deletions. Despite these continuous DNA damaging events, nature has recruited reinforcements in the form of checkpoint pathways that maintain and monitor the integrity of the genome. These pathways were first observed when it was shown that damage induced by chemotherapeutic agents resulted in an inhibition of the cell cycle, allowing for cellular repair.“ 43 A Similar control of cell cycle progression was found in Saccharomyces cerevisiae and was then coined a ‘checkpoint’.44 It was originally thought that checkpoint pathways were operational for the sole purpose of regulating cell cycle transitions. Currently, it is generally accepted that the checkpoints are part of a cascade of signals that ultimately lead to DNA damage response processes.” 39 These response processes include removal of the DNA damaged sites and restoration of the DNA duplex. Temporary arrest of the cell cycle allows for repair and prevention of the duplication of damaged DNA or its transcriptional response. In the event of heavily damaged or seriously deregulated cells, induction of apoptosis or terminal cell cycle arrest occurs.” 45' 46 The DNA damage cell cycle checkpoints will repair, patch, and promote the overall survival of cells, even cancer cells, resulting in a reduction of the overall anticancer efficacy. However, if the induction of checkpoint pathways and DNA repair could be selectively inhibited in cancerous cells, the efficacy of the treatment may be enhanced. Alternatively, if DNA repair can be selectively induced in healthy cells, the therapeutic window of the treatment could be significantly widened, allowing for a more tolerable and effective anticancer therapy. Consequently, modulating the DNA checkpoint pathways using small molecules could potentially sensitize cancer cells and desensitize healthy cells to DNA damage induced by 2 chemotherapeutics or ionizing radiation.4 More specifically, altering the checkpoint pathways via modulation of two of the key checkpoint kinases, Cth 10 “u 'u g. and Chk2, has become an increasingly appealing approach to broaden the therapeutic window of conventional anticancer therapies.47 These checkpoint pathways, rather than being thought of as a molecular switch, represent a continuous process that is amplified in the presence of DNA damage, such as from ultraviolet (UV) and ionizing radiation (IR).37 Between UV and IR, UV radiation is lower in energy and the majority of DNA damage results from the formation of photoproducts, such as dimers of pyrimidine containing nucleotides (A, Scheme l-1). These dimmers can inhibit DNA transcription and replication machinery ultimately leading to cell death.“ 49 Ionizing radiation is higher in energy and can cause more substantial damage to DNA. IR is absorbed mostly by surrounding water molecules, which subsequently form highly reactive radicals that cause severe DNA damage. These radicals can cause nucleotide damage, crosslinking and strand breaks. Reaction B in Scheme I-1 illustrates an example of a strand cleavage reaction that could occur due to ionizing radiation.16 Scheme I-1. DNA damage by UV and IR O O O O HN I HN I A A A UV HN NH _,' cell death 0 N O N _’ ,2 ' 0%N NAO / \ / I F’O\k——7Base—.fl'+ PC)\:;7Basefi|i> POUasefi FIE—788% P= phosphate 11 ’50 BI I m vmnnnpf‘l W ilv- V ‘ p-Afiflr‘q't - . . uni-4 I... V '1!!! I I" . as 3 a 5: Oil-"I 3.5 l A v Uni ans The DNA damage checkpoint pathways consist of three different main components: sensors, signal transducers, and effectors (Figure I-5). The sensors recognize the damaged DNA and initiate subsequent events. Two of the main sensors are ATM and ATR, which belong to the phosphoinositide 3-kinase-like kinase (PIKK) family members.” 50 The first DNA damage sensor, ATM (ataxia telangiectesia mutated) exhibits kinase activity when activated by agents that induce double strand breaks, such as ionizing radiation.51 ATM will subsequently phosphorylate proteins such as Chk2 and p5351' 52 (among others) when activated after cells have been exposed to IR. A deficiency in ATM exhibits phenotypes such as cerebellar degeneration, immunodeficiency, genome instability, clinical radiosensitivity, and cancer predisposition.53 The second DNA damage sensor, ATR, has a sequence homology to both ATM and SpRad3, thus comes the name ATR (ATM and Rad3 related).37' 5‘ ATR is similar to ATM since it is a kinase that essentially phosphorylates the majority of the same substrates as ATM. However, ATR is activated in vivo by UV radiation rather than ionizing radiation.” 55 Unlike ATM, it does not appear that ATR is activated by double strand breaks.37 Thus, ATR is the main PIKK family member that initiates a signal transduction pathway after UV radiation damage.55 12 DNA Damage Sensors Signal Transducers Effectors W DNA DAMAGE / ATR I Chk1 I CD025 CDK Activation \ ATM I II Chk2 I ll ll p53 Pathway Cell cycle arrest (DNA repair & replication) fl Apoptosis (Cell death) Figure I-5. Components of the damage response network The Signal transducers are components of the DNA damage pathway that essentially receive the signal from the sensors. The two signal transducers that will be discussed here in detail are Chk1 and Chk2.56‘58 Chk1 and Chk2 are structurally unrelated but are essential checkpoint kinases downstream of the 13 It tTillslt 5390'853 320mg: 8386 Chen ' “I90: tel .eVed 'Efiajj‘ re DNA damage sensors and play a critical role in determining the cell’s fate.37' 59‘“ ATM is activated by double strand breaks and the signal is transduced by Chk2 52' 53, while the UV damage signal sensed by ATR is transduced by Chk1.55' 64 Chk1 is primarily responsible for cell arrest in response to DNA damage, allowing for the initiation of DNA repair65, whereas Chk2 has been implicated with the phosphorylation and activation of the apoptotic transcription factor p53.4°' 62' 65 It is important to note the drastic difference in Chk1 and Chk2 null mice. Chk1 (-/-) mice are not viable and exhibit embryonic lethality” 66 , whereas Chk2 (-/-) mice still illicit near normal checkpoint responses and are viable.67 The effector components, Cch5 and p53, are involved in the arrest of the cell cycle and apoptosis, respectively. Three phosphotyrosine phosphatases, Cch5a-c, are responsible for dephosphorylating cyclin-dependent kinases that ultimately affect proteins directly involved in cell cycle transitions?7 Essentially, the checkpoint kinases phosphorylate one or more of the Cdc25 proteins that results in their inactivation and degradation, thus preventing the Cd025 proteins to translocate into the nucleus and enable cell cycle progression'58 The tumor suppressor protein p53 is responsible for arresting the cell cycle and inducing apoptosis.69 Based on the described pathways it is possible that inhibition of these checkpoint kinases may be used to enhance the effects of chemotherapeutics by sensitizing cancer cells or desensitizing healthy cells.6°' 70‘" Inhibition of Chk1 is believed to sensitize tumor cells by blocking the cell’s ability to initiate DNA repair, resulting in unsustainable DNA damage.” 65' 73' 74 Inhibition of the G2 14 “It. al-m' ‘ V1 ‘v‘l It» :55 215 CI ~AAt—r 0‘ up a . .4. PM . .~ A “WAC. , «VCJ I DNA checkpoint Chk2 can selectively prevent apoptosis in p53 wild type cells (healthy cells), thus desensitizing them from cell death during chemotherapeutic treatment.” 75 Chk2 inhibitors are anticipated not to have an effect on apoptosis in cells containing mutated p53 (>50% of cancerous cells). We were interested in focusing our efforts on developing Chk2 inhibitors to desensitize healthy cells toward ionizing radiation to help widen the therapeutic window of this treatment. I.F Validation of Chk2 as drug target Activation of Chk2 is initiated by factors that cause DNA damage, such as ionizing radiation, chemotherapeutic agents, and telomere initiated senescence. 40' 76' 77 In response to IR induced DNA damage, signals are intercepted by ATM, which in turn directly phosphorylates Chk2 at Thr68 within the SQ/TQ rich domain.73' 79 After the initial phosphorylation and dimerization, multiple intermolecular phosphorylation events occur to complete the activation.” 80 The phosphorylation of Ser516 was recently found to be of great importance as this modification is required for full activation of Chk2.4°'81'82 A schematic of the DNA damage checkpoint pathway involving Chk2 is shown in Figure l-6. Once activated and all the phosphorylation events of Chk2 have transpired, Chk2 signals the effector proteins CchSa, Cd025c, and p53, among others (Figure l-6). The Cch5 family of phosphatases is responsible for the progression of the cell cycle by dephosphorylating cyclin dependent kinases cdk1 and cdk2 at specific sites.4°' “'86 Chk2 phosphorylates Cdc25a at Ser123 and consequently renders it inactive‘o' 84' 87, while Chk2 phosphorylates Cdc25c at Ser216. This results in the sequestering of Cdc25c in the cytoplasm, thus 15 preventing activation of the cdk1/Cyclin B complex, resulting in G2/M arrest.” 88‘ 90 Apoptosis I I G1-S phase checkpoint S-phase checkpoint G2-M phase checkpoint Figure l-6. DNA damage pathway involving Chk2 Another important downstream substrate of Chk2 is tumor suppressor protein p53. Although the exact relationship between p53 and Chk2 is still unclear, Chk2 is thought to be upstream of p53 in the reaction pathway that contains IR induced apoptosis.” 42' 91 It was reported that Chk2 phosphorylates p53 at Ser20 thereby regulating the amount of activated p53 in response to 16 ,‘cv- vdu' .3. ,.: .. d4 ‘. no u it r‘-. double strand breaks.” 62' ”'94 However, this point has been challenged based on a few observations. First, the p53 derived peptide containing the Chk2 targeted phosphorylation sites is a poor substrate for Chk2 and does not contain the characteristic sequence for directing a phosphorylation found in other Chk2 substrates. Second, the phosphorylation of p53 induced by DNA damage was only slightly affected, if at all, by down regulation or knockout experiments of Chk2.62' 91' 95"” Despite these arguments, most scientific analyses of the Chk2- p53 link strongly support the role of Chk2 as a p53 kinase.97 The role of Chk2 in DNA damage induced apoptosis is supported by Chk2 deficient mice, which Show an increased resistance to ionizing radiation and cellular defects in apoptosis.62' 67' 95' 96 Chk2 -/- mice survived significantly longer than the wild-type mice following whole body irradiation.96 The increased survival of those mice without Chk2 is attributed to resistance of several cell types to IR induced apoptosis.95' 96 Furthermore, it is important to remember that Chk2 (-/—) mice still illicit near normal checkpoint responses and are viable.67 The support given by the study where the Chk2 null mice survived whole body irradiation and the study which showed that Chk2 null mice were still viable offer validation for choosing Chk2 as the drug target. LG Purified drug target and X-ray crystal structure As illustrated in Figure l-4, it can be important to obtain information from the purified drug target to aid in the development Of inhibitors. Fortunately, Oliver et al. were able to crystallize the kinase domain of Chk2 (Figure I-7) and in effect 17 give an enormous amount of information to the aid in developing inhibitors of Chk2.99 1 12-175 220-486 1 543 N :26; [ FHA ]——-[ Kinase C Figure l-7. Illustration of Chk2 and crystal structure of kinase domain The top of Figure l-7 displays the linear representation of the whole checkpoint kinase 2 protein. As illustrated above, Chk2 is a 543 long amino acid protein that consists of three domains. The first domain is called a serine- glutamine/threonine-glutamine cluster domain (SCD), which is the substrate for 18 lg"! ,m I. .I. “F:- (t) ‘A ”I the upstream protein ATM, as shown in Figure l-6. The second domain is called the forkhead associated domain (FHA) which has been implicated to help modulate protein-protein interactions. The last domain, which was crystallized, is the kinase domain responsible for performing the major requirement of a kinase, that is, to phosphorylate its substrates.4°' 99 Overall, gaining access to this information helps in discovering new inhibitors of Chk2 and allows for the fonNard progress in obtaining an adjuvant drug for radiation therapy. l.H Checkpoint kinase 2 inhibitors Until recently, there were only a few reports of potent Chk2 inhibitors that show good Chk2 selectivity. Within the last decade there has been an increase into the amount of research being performed to develop checkpoint kinase 2 inhibitors. Some remain highly confidential like XL844, developed by Exelixis, which is one of the newest checkpoint kinase inhibitors to move into clinical trials.100 Others have been kept hidden in patents, like the indazole and squaric acid derivatives shown in Figure l-8, and have not been highly publicizedm'103 O o o R, HN R4 0 I / R HN / Fifi I: /R3 N\ \\ 3 \ HN-X X‘_©_S W/ Y /R n non-benzimidazole 2-arylbenzimidazole (2 nM) (GpM - 16 nM) Figure l-10. Benzimidazole and non-benzimidazole Chk2 inhibitors To help define the scope of the SAR study on the benzimidazole inhibitors, a series of non-benzimidazole Chk2 inhibitors were developed. The leo values for the more potent new inhibitors range from 5.8 pM to 16 nM.108 Further studies are needed to determine if these inhibitors are effective at potentiating the cytotoxic abilities of current cancer therapeutics.3 Natural products and analogs of natural products have also contributed to the number of Chk2 inhibitors currently known. Staurosporine (Figure M1) and analogs, such as UCN-01 (Figure l-11) have shown to be potent checkpoint inhibitors, however, the are not very selective for checkpoint kinases.1°6' 111' "2 Although UCN-O1 displays a potent inhibition of Chk2 (10 nM)24 and has shown to potentiate the anticancer activity of a variety of therapeutics such as cis-platin, 113-117 mitomycin and ionizing radiation and augments the cytotoxicity of temozolomide in human glioblastoma cells118, there are some concerns associated with UCN-O1 such as its promiscuity as a kinase inhibitor. In addition, 21 ,fll L'Vi‘tul SM 5, >85 UCN-01 contains some pharmacokinetic drawbacks including its strong binding to human plasma protein d1-acid glycoprotein and low bioavailability.“9'121 H O N U N N O . ,, N ; ”OCH, - 2 'I’OCH3 NHCH3 NHCH3 Natural Ems: Staurosporine (80 nM) ”CNN (10 nM) HZN /N HN 0 \ / I N H NH O W W lndoloazeplne (8 nM) DBH (183 nM) Figure I-11. Natural products and analogs used as Chk2 inhibitors Debromohymenialdisine (DBH) (Figure M1) is a natural product that displays moderate IC50 values for Chk2 inhibition (3.5 pM (cell culture), 183 nM in vitro).73' ‘22 However, a low selectivity for Chk2 over other kinases severely limits the use of debromohymenialdisine as a Chk2 inhibitor.3 A new analog of debromohymenialdisine, indoloazepine (Figure l-11), was developed in our laboratory and has been shown to be a potent and selective inhibitor of Chk2.122 It was illustrated that indoloazepine displayed an leo value of 8 nM for Chk2 kinase inhibition through an in vitro assay and was shown to be quite selective against a number of purified kinases including Chk1 (30 fold).3'122 22 Fl“ 5n“. ..5', .: :3 {0 Li Ch f F ui v'lv aJ 32:”lnar . - . ‘Vy ECI.ESE h. W2 It is with the success of indoloazepine as a potent checkpoint kinase 2 inhibitor that the foundation of my project is built upon. |.| Chk2 binding pocket properties With the success of the crystallization of the kinase domain of Chk2 by Oliver and co-workersQQ, information regarding the binding pocket can be examined and explored. Oliver et al. were also successful in crystallizing debromohymenialdisine inside of the binding pocket allowing a scrutinizing eye to identify possible residues important for binding purposes. It can be seen in Figure l-12 the important binding interactions between DBH and the Chk2 active site. Figure l-12. DBH inside the Chk2 active site DBH is shown in the center of Figure l-12 and the yellow dashed lines represent hydrogen bonding interactions with the side chains and main chain of Chk2. It is observed that the main interactions that make DBH a fairly potent 23 inhibitor include the imidazolone moiety, which shows interactions with threonine, arginine and glutamic acid residues. Furthermore, there are additional hydrogen bonding interactions that are present in the southern portion of DBH. Figure l-13 illustrates these interactions and it can be seen that the amide functional group of the seven—membered ring plays a large role in bonding to a glutamic acid and methionine residue along the main chain in Chk2. MET 304 Figure l-13. Interactions between southern portion of DBH and Chk2 Interpreting this information and applying it to the DBH analog, indoloazepine, it can be imagined that indoloazepine may have similar hydrogen bonding interactions within Chk2. In fact, according to Figure l—14, indoloazepine could potentially bind in a similar fashion as DBH. The hydrogen bonding interactions between the imidazolone ring of indoloazepine and the residues 24 -..,“ N l :4 av ‘- surrounding it could interact to help hold indoloazepine in the active site. Furthermore, Figure ”-14 also shows that indoloazepine could have the same interactions between the amide of the seven membered ring and the same residues that were proposed to hold DBH inside the active site. Figure l-14. lndoloazepine modeled inside the Chk2 binding pocket Although the exact nature of the binding of indoloazepine and the active site of Chk2 is not known, it can be hypothesized based on the known binding properties of a very closely related structure (DBH). Furthermore, we can use this information to rationalize why it is thought that a new indole alkaloid, the focal point of my research, could also be a potential Chk2 inhibitor. As shown previously in Figure l-4, the point at which drug leads have been identified can lead to a rational design approach, which feeds back into the natural product tank 25 .- fif‘fli‘ [‘ w a. "JV ' “map: F; a, ...‘UIJ a T1 . "en Men A A ‘- I. a I)- x. to uncover new natural products of interest as well as a new library of compounds. The success of indoloazepine as a Chk2 inhibitor has led to a reevaluation of another natural product, shown in Figure l-15. This indole alkaloid possesses some structural features that are similar to that of indoloazepine. As a result, it warranted further examination to determine its potential inhibition of Chk2. Indole alkaloid lndoloazepine Figure M5 New indole alkaloid and indoloazepine At first glance the structural similarities between the indole alkaloid and indoloazepine may not be obvious, besides the fact that they both contain an imidazolone ring and indole moiety. However, after some calculations using Spartan Pro®, it can be seen that there are some more important correlations. As illustrated in Figure I-16, the dihedral angle between the plane containing the imidazolone ring and the plane containing the indole ring for indoloazepine is calculated to be 50°. Furthermore, according to the crystal structure of the indoloazepine analog where the indole nitrogen is methylated, the dihedral angle is 56°. When examining the new indole alkaloid, it was determined that the dihedral angle between the imidazolone ring and indole is also very similar with an angle of 54°. It is important to remember that spatial arrangements inside the 26 binding pocket are very important when considering how the molecule fits into the active site. More importantly, the imidazolone moiety seemed to be very important to the binding of DBH and indoloazepine considering the number of hydrogen bonding interactions that were present. Dihedral angle = 54° Dihedral angle = 50° Figure I-16. Comparison of dihedral angles Further analysis of both compounds using Spartan Pro® revealed that the carbon connected to the indole ring (at position 3) for both compounds were surprisingly similar even though the two compounds contained different hybridizations at that carbon. It is shown in Figure l-17 that the hybridization of indoloazepine at carbon 3 of the indole ring, although technically sp2 hybridized, actually exhibits more of a sp3 hybridization, possibly due to the partial single bond character that the bond has as a result of the donating nature of the indole nitrogen’s lone pair of electrons. The angle at the indicated carbon of indoloazepine was calculated to be 111°, while the angle at the carbon on position 3 of the indole from the new indole alkaloid was calculated to be 110°. 27 3 Angle = 110.. Hybridization = sp2 Angle = 111° Hybridization = sp Figure l-17. Hybridization of carbon connected to indole ring Placing the new indole alkaloid inside the binding pocket of Chk2 (Figure l-18) reveals that there is potential for the imidazolone ring of the alkaloid to hydrogen bond to the same residues responsible for binding DBH and possibly indoloazepine. Although the indole alkaloid lacks the amide functional group that seems to play a role in the binding of DBH and indoloazepine, it does contain a ketone, which was thought to potentially interact with a lysine residue to form a Schiff base and result in a covalent bonding interaction, possibly allowing for a more potent inhibition. It is understood that these potential interactions and reason for possible potency are purely speculative. There is no definite way to know exactly which compounds are active and which are not. Our hypothesis is that this natural product has the potential and structural features that may allow it to be a checkpoint kinase 2 inhibitor. Thus, our hypothesis is the impetus for my project, which is focused on the new indole alkaloid and its potential biological activity. 28 LYS 249 Figure I-18. New indole alkaloid modeled in Chk2 binding pocket 29 l.J Goal of project The main goal of my project includes the synthesis of the natural product. Furthermore, it was thought that the synthesis of the natural product could be obtained through an oxazolone intermediate, a scaffold that our laboratory has explored quite extensively (Figure l-19).123‘135 It was thought that new methodology could be developed extending our chemistry to grant access to quaternary imidazolones, the core of the natural product. Additional goals were to synthesize analogs of the natural product, as well as other heterocyclic compounds that could be tested for Chk2 activity. The following chapters elaborate on the development of a new synthetic method that gives access to the quaternary imidazolone scaffold, the first total synthesis of the indole alkaloid and two analogs and the biological evaluation of the indole alkaloid, analogs and other heterocyclic compounds synthesized for testing. '34 HZNYH R4 R5 . R2 . _ Ix \ CO H NWRS / R1 N 2 0 Re 0R4 R1 0 T5 RI R2 \]|/ O R4/N /N 5 \ +—— N R _____ ... Y O o N C02” R2 3 HN72: H 3 R2 Oxazolone t 5\N/S;R2 F/ R4 )eN COZH R1 R2 Figure I-19. Oxazolone chemistry affording various heterocycles 30 |.K 10. 11. References Jemal, A.; Thun Michael, J.; Ries Lynn, A. G.; Howe Holly, L.; Weir Hannah, K.; Center Melissa, M.; Ward, E.; Wu, X.-C.; Eheman, C.; Anderson, R.; Ajani Umed, A.; Kohler, B.; Edwards Brenda, K., Annual report to the nation on the status of cancer, 1975-2005, featuring trends in lung cancer, tobacco use, and tobacco control. J Natl Cancer Inst 2008, 100, 1672-94. Greenlee, R. T.; Murray, T.; Bolden, S.; Wingo, P. A., Cancer statistics, 2000. CA CancerJ Clin 2000, 50, 7-33. Sharma, V.; HUpp, C. D.; Tepe, J. J., Enhancement of chemotherapeutic efficacy by small molecule inhibition of NF-kB and checkpoint kinases. Curr. Med. Chem. 2007, 14, 1061-1074. Warshawsky, D.; Landolph, J. R., Molecular Carcinogenesis and The Molecular Biology of Human Cancer. CRC Taylor & Francis: Boca Raton, FL, 2006. Aletras, V.; Hadjiliadis, D.; Hadjiliadis, N., On the Mechanism of Action of the Antitumor Drug cis-Platin (cis-DDP) and its Second Generation Derivatives. Met Based Drugs 1995, 2, 153-85. Bernier, J.; Hall, E. J.; Giaccia, A., Timeline: Radiation oncology: a century of achievements. Nat. Rev. Cancer 2004, 4, 737-747. Bikker, J. A.; Brooijmans, N.; Wissner, A.; Mansour, T. S., Kinase Domain Mutations in Cancer: Implications for Small Molecule Drug Design Strategies. J. Med. Chem. 2009, 52, 1493-1509. Ghoshal, K.; Jacob, S. T., An alternative molecular mechanism of action of 5-fluorouracil, a potent anticancer drug. Biochem. Pharmacol. 1997, 53, 1569—1575. Hande, K. R., Etoposide: four decades of development of a topoisomerase II inhibitor. Eur. J. Cancer 1998, 34, 1514-1521. Katsoulas, A.; Rachid, Z.; Brahimi, F.; McNamee, J.; Jean-Claude, B. J., Cytokinetics and mechanism of action of AKO4: a novel nitrogen mustard targeted to bcr-abl. Leuk. Res. 2005, 29, 565-572. Krohn, K.; Ed., Topics in Current Chemistry; Anthracycline Chemistry and Biology II; Mode of Action, Clinical Aspects and New Drugs. Springer- Verlag: 2008; Vol. 283, p 222 pp. 31 .. yw 5.). l\" —L R” K‘u'i and red R” i Uh Or: his Tie WC ail 201 to; 12. 13. 14. 15. 16. 17. 18. 19. 20. 21. 22. 23. 24. Kumar, G. S.; Lipman, R.; Cummings, J.; Tomasz, M., Mitomycin C-DNA adducts generated by DT-diaphorase. Revised mechanism of the enzymic reductive activation of mitomycin C. Biochemistry 1997, 36, 14128-14136. Rothenberg, M. L., Topoisomerase | inhibitors: review and update. Ann. Oncol. 1997, 8, 837-55. Rowinsky, E. K.; Donehower, R. C., Paclitaxel (taxol). N Engl J Med 1995, 332, 1004-14. Sahasranaman, 8.; Howard, D.; Roy, 8., Clinical pharmacology and pharmacogenetics of thiopurines. Eur. J. Clin. Pharmacol. 2008, 64, 753- 767. Sonntag, C. v., Free-Radical—lnduced DNA Damage and Its Repair. Springer-Verlag: 2006; p 523. Thorburn, A.; Frankel, A. E., Apoptosis and anthracycline cardiotoxicity. Mol. Cancer Ther. 2006, 5, 197-199. Trevino, A. V.; Woynarowska, B. A.; Herman, T. S.; Priebe, W.; Woynarowski, J. M., Enhanced topoisomerase ll targeting by annamycin and related 4-demethoxy anthracycline analogues. Mol. Cancer Ther. 2004, 3, 1403-1410. Foye, W. 0., Cancer Chemotherapeutic Agents. ACS: Washington, 1995. Pourquier, P.; Pommier, Y., Topoisomerase l-mediated DNA damage. Adv. Cancer Res. 2001, 80, 189-216. Sordet, O.; Khan, Q. A.; Kohn, K. W.; Pommier, Y., Apoptosis induced by topoisomerase inhibitors. Curr Med Chem Anticancer Agents 2003, 3, 271-90. Rajski, S. R.; Williams, R. M., DNA cross-linking agents as antitumor drugs. Chem. Rev. 1998, 98, 2723-2795. Hurley, L. H., DNA and its associated processes as targets for cancer therapy. Nat. Rev. Cancer 2002, 2, 188-200. Yu, 0.; La Rose, J.; Zhang, H.; Takemura, H.; Kohn, K. W.; Pommier, Y., Ucn-01 inhibits p53 up-regulation and abrogates g-radiation-induced G2-M checkpoint independently of p53 by targeting both of the checkpoint kinases, chk2 and chk1. Cancer Research 2002, 62, 5743—5748. 32 25. 26. 27. 28. 29. 30. 31. 32. 33. 35. 36. 37. 38. Janssens, S.; Tschopp, J., Signals from within: the DNA-damage-induced NF-kappaB response. Cell Death Differ 2006, 13, 773-84. Kastan, M. B.; Bartek, J., Cell-cycle checkpoints and cancer. Nature 2004, 432, 316-23. Webster, G. A.; Perkins, N. D., Transcriptional cross talk between NF- kappaB and p53. Mol. Cell Biol. 1999, 19, 3485-95. Ryan, K. M.; Ernst, M. K.; Rice, N. R.; Vousden, K. H., Role of NF-kappaB in p53-mediated programmed cell death. Nature 2000, 404, 892-7. Meek, D. W., The p53 response to DNA damage. DNA Repair (Amst) 2004, 3, 1049-56. Sherr, C. J., Cancer cell cycles. Science 1996, 274, 1672-7. Makin, G.; Hickman, J. A., Apoptosis and cancer chemotherapy. Cell Tissue Res 2000, 301, 143-52. Karin, M.; Lin, A., NF—kappaB at the crossroads of life and death. Nat. Immunol. 2002, 3, 221-7. Liu, 0.; Guntuku, S.; Cui, X.-S.; Matsuoka, S.; Cortez, D.; Tamai, K.; Luo, G.; Carattini-Rivera, S.; DeMayo, F.; Bradley, A.; Donehower, L. A.; Elledge, S. J., Chk1 is an essential kinase that is regulated by Atr and required for the G2/M DNA damage checkpoint. Genes Dev. 2000, 14, 1448-1459. Beg, A. A.; Baltimore, D., An essential role for NF-kappaB in preventing TNF-alpha-induced cell death. Science 1996, 274, 782-4. Boland, M. P., DNA damage signalling and NF-kappaB: implications for survival and death in mammalian cells. Biochem. Soc. Trans. 2001, 29, 674-8. Pommier, Y.; Weinstein, J. N.; Aladjem, M. l.; Kohn, K. W., Chk2 molecular interaction map and rationale for Chk2 inhibitors. Clin Cancer Res 2006, 12, 2657-61. Sancar, A.; Lindsey-Boltz, L. A.; Unsal-Kacmaz, K.; Linn, 8., Molecular Mechanisms of Mammalian DNA Repair and the DNA Damage Checkpoints. Annu. Rev. Biochem. 2004, 73, 39-85. Zhou, B. B.; Elledge, S. J., The DNA damage response: putting checkpoints in perspective. Nature 2000, 408, 433-9. 33 39. 40. 41. 42. 43. 44. 45. 46. 47. 48. 49. 50. 51. 52. Elledge, S. J., Cell cycle checkpoints: preventing an identity crisis. Science 1996, 274, 1664-72. Ahn, J.; Urist, M.; Prives, C., The Chk2 protein kinase. DNA Repair 2004, 3, 1039-1047. Liu, R.; Hsieh, C.-Y.; Lam, K. S., New approaches in identifying drugs to inactivate oncogene products. Semin. Cancer Biol. 2004, 14, 13-21. Zhou, B. B. S.; Bartek, J., Targeting the Checkpoint Kinases: Chemosensitization versus Chemoprotection. Nature Rev. 2004, 4, 1-10. Tobey, R. A., Different drugs arrest cells at a number of distinct stages in G2. Nature 1975, 254, 245-7. Weinert, T. A.; Hartwell, L. H., The RAD9 gene controls the cell cycle response to DNA damage in Saccharomyces cerevisiae. Science 1988, 241, 317-22. Zhou, B.-B. S.; Elledge, S. J., The DNA damage response: putting checkpoints in perspective. Nature 2000, 408, 433-439. Khanna, K. K.; Jackson, S. P., DNA double-strand breaks: signaling, repair and the cancer connection. Nat. Genet. 2001, 27, 247-254. Bartek, J.; Lukas, J., Chk1 and Chk2 kinases in checkpoint control and cancer. Cancer Cell 2003, 3, 421-9. Batista, L. F. 2.; Kaina, B.; Meneghini, R.; Menck, C. F. M., How DNA lesions are turned into powerful killing structures: Insights from UV- induced apoptosis. Mutat. Res., Rev. Mutat. Res. 2009, 681, 197-208. Cadet, J.; Sage, E.; Douki, T., Ultraviolet radiation-mediated damage to cellular DNA. Mutat. Res., Fundam. Mol. Mech. Mutagen. 2005, 571, 3-17. Durocher, D.; Jackson, S. P., DNA-PK, ATM and ATR as sensors of DNA damage: variations on a theme? Curr. Opin. Cell Biol. 2001, 13, 225-231. Banin, S.; Moyal, L.; Shieh, S. Y.; Taya, Y.; Anderson, C. W.; Chessa, L.; Smorodinsky, N. l.; Prives, C.; Reiss, Y.; Shiloh, Y.; Ziv, Y., Enhanced phosphorylation of p53 by ATM in response to DNA damage. Science 1998, 281, 1674-1677. Canman, C. E.; Lim, D.-S.; Cimprich, K. A.; Taya, Y.; Tamai, K.; Sakaguchi, K.; Appella, E.; Kastan, M. B.; Siliciano, J. D., Activation of the 34 J 17" (J) ‘1’.’ 53. 54. 55. 56. 57. 58. 59. 60. 61. 62. 63. 64. 65. ATM kinase by ionizing radiation and phosphorylation of p53. Science 1998, 281 , 1677-1679. Shiloh, Y., ATM and related protein kinases: safeguarding genome integrity. Nat. Rev. Cancer 2003, 3, 155-168. Cimprich, K. A.; Shin, T. B.; Keith, C. T.; Schreiber, S. L., cDNA cloning and gene mapping of a candidate human cell cycle checkpoint protein. Proc. Natl. Acad. Sci. U. S. A. 1996, 93, 2850-5. Abraham, R. T., Cell cycle checkpoint signaling through the ATM and ATR kinases. 2001; Vol. 15, p 2177-2196. Melo, J.; Toczyski, D., A unified view of the DNA-damage checkpoint. Curr. Opin. Cell Biol. 2002, 14, 237-245. Rhind, N.; Russell, P., Chk1 and Cds1: linchpins of the DNA damage and replication checkpoint pathways. J. Cell Sci. 2000, 113, 3889-3896. McGowan, C. H., Checking in on cds1 (chk2): a checkpoint kinase and tumor suppressor. BioEssays 2002, 24, 502-511. Zhou, B. B.; Bartek, J., Targeting the checkpoint kinases: Chemosensitization versus Chemoprotection. Nat Rev Cancer 2004, 4, 216-25. Zhou, B. B.; Anderson, H. J.; Roberge, M., Targeting DNA checkpoint kinases in cancer therapy. Cancer Biol Ther 2003, 2, 816-22. Blagden, S.; de Bono, J., Drugging cell cycle kinases in cancer therapy. Curr Drug Targets 2005, 6, 325-35. Hirao, A.; Kong, Y.-Y.; Matsuoka, S.; Wakeham, A.; Ruland, J.; Yoshida, H.; Liu, D.; Elledge, S. J.; Mak, T. W., DNA damage-induced activation of p53 by the checkpoint kinase Chk2. Science 2000, 287, 1824-1827. Matsuoka, S.; Rotman, G.; Ogawa, A.; Shiloh, Y.; Tamai, K.; Elledge, S. J., Ataxia telangiectasia-mutated phosphorylates Chk2 in vivo and in vitro. Proc. Natl. Acad. Sci. USA 2000, 97, 10389-10394. Zhao, H.; Piwnica-Worms, H., ATR-mediated checkpoint pathways regulate phosphorylation and activation of human Chk1. Mol. Cell. Biol. 2001, 21, 4129-4139. Zhou, B. B.; Sausville, E. A., Drug discovery targeting Chk1 and Chk2 kinases. Prog Cell Cycle Res 2003, 5, 413-21. 35 66. 67. 68. 69. 70. 71. 72. 73. 74. 75. Takai, H.; Tominaga, K.; Motoyama, N.; Minamishima, Y. A.; Nagahama, H.; Tsukiyama, T.; Ikeda, K.; Nakayama, K.; Nakanishi, M.; Nakayama, K.- l., Aberrant cell cycle checkpoint function and early embryonic death in Chk1-l- mice. Genes Dev. 2000, 14, 1439-1447. Jack, M. T.; Woo, R. A.; Hirao, A.; Cheung, A.; Mak, T. W.; Lee, P. W. K., Chk2 is dispensable for p53-mediated G1 arrest but is required for a latent p53-mediated apoptotic response. Proc. Natl. Acad. Sci. USA 2002, 99, 9825-9829. Bartek, J.; Lukas, J., Mammalian G1- and S-phase checkpoints in response to DNA damage. Curr. Opin. Cell Biol. 2001, 13, 738-747. Pietrancosta, N.; Garino, C.; Laras, Y.; Quelever, G.; Pierre, P.; Clavarino, G.; Kraus, J.-L., Are p53 inhibitors potentially useful therapeutics? Drug Dev. Res. 2005, 65, 43-49. Suganuma, M.; Kawabe, T.; Hori, H.; Funabiki, T.; Okamoto, T., Sensitization of cancer cells to DNA damage-induced cell death by specific cell cycle G2 checkpoint abrogation. Cancer Res 1999, 59, 5887- 91. Tenzer, A.; Pruschy, M., Potentiation of DNA-damage-induced cytotoxicity by G2 checkpoint abrogators. Curr Med Chem Anti-Canc Agents 2003, 3, 35—46. Sausville, E. A., Cyclin-dependent kinase modulators studied at the NCI: pre-clinical and clinical studies. Curr Med Chem Anti-Cane Agents 2003, 3, 47-56. Curman, D.; Cinel, B.; Williams, D. E.; Rundle, N.; Block, W. D.; Goodarzi, A. A.; Hutchins, J. R.; Clarke, P. R.; Zhou, B.-B.; Lees-Miller, S. P.; Andersen, R. J.; Roberge, M., Inhibition of the G2 DNA damage checkpoint and of protein kinases Chk1 and Chk2 by the marine sponge alkaloid debromohymenialdisine. J. Biol. Chem. 2001, 276, 17914-17919. Prudhomme, M., Novel checkpoint 1 inhibitors. Rec. Pat. Anti. Cancer Drug Discov. 2006, 1, 55-68. Lukas, C.; Bartkova, J.; Latella, L.; Falck, J.; Mailand, N.; Schroeder, T.; Sehested, M.; Lukas, J.; Bartek, J., DNA damage-activated kinase Chk2 is independent of proliferation or differentiation yet correlates with tissue biology. Cancer Res. 2001, 61, 4990-4993. 36 76. 77. 78. 79. 80. 81. 82. 83. 84. 85. 86. d'Adda di Fagagna, F.; Reaper, P. M.; CIay-Farrace, L.; Fiegler, H.; Carr, F.; von Zglinicki, T.; Saretzki, G.; Carter, N. R; Jackson, S. P., A DNA damage checkpoint response in telomere-initiated senescence. Nature 2003, 426, 194-198. Oh, H.; Wang, S. C.; Prahash, A.; Sano, M.; Moravec, C. S.; Taffet, G. E.; Michael, L. H.; Youker, K. A.; Entman, M. L.; Schneider, M. D., Telomere attrition and Chk2 activation in human heart failure. Proc. Natl. Acad. Sci. USA 2003, 100, 5378-5383. Ahn, J.-Y.; Schwarz, J. K.; Piwnica-Worms, H.; Canman, C. E., Threonine 68 phosphorylation by ataxia telangiectasia mutated is required for efficient activation of Chk2 in response to ionizing radiation. Cancer Res. 2000, 60, 5934-5936. Melchionna, R.; Chen, X.-B.; Blasina, A.; McGowan, C. H., Threonine 68 is required for radiation-induced phosphorylation and activation of Cds1. Nat. Cell Biol. 2000, 2, 762-765. Lee, C.-H.; Chung, J. H., The ths1 (Chk2)-FHA domain is essential for a chain of phosphorylation events on ths1 that is induced by ionizing radiation. J. Biol. Chem. 2001, 276, 30537-30541. Schwarz, J. K.; Lovly, C. M.; Piwnica-Worms, H., Regulation of the Chk2 protein kinase by oligomerization-mediated cis- and trans-phosphorylation. Mol. Cancer Res. 2003, 1, 598-609. Wu, X.; Chen, J., Autophosphorylation of Checkpoint Kinase 2 at Serine 516 Is Required for Radiation-induced Apoptosis. J. Biol. Chem. 2003, 278, 36163-36168. Matsuoka, S.; Huang, M.; Elledge, S. J., Linkage of ATM to cell cycle regulation by the Chk2 protein kinase. Science 1998, 282, 1893-1896. Falck, J.; Mailand, N.; Syljuasen, R. G.; Bartek, J.; Lukas, J., The ATM- Chk2-Cdc25a checkpoint pathway guards against radioresistant DNA synthesis. Nature 2001, 410, 842-847. Brown, A. L.; Lee, C.-H.; Schwarz, J. K.; Mitiku, N.; Piwnica-Worms, H.; Chung, J. H., A human Cds1-related kinase that functions downstream of ATM protein in the cellular response to DNA damage. Proc. Natl. Acad. Sci. USA 1999, 96, 3745-3750. Blasina, A.; Van de Weyer, |.; Laus, M. C.; Luyten, W. H. M. L.; Parker, A. E.; McGowan, C. H., A human homolog of the checkpoint kinase Cds1 directly inhibits Cd025 phosphatase. Curr. Biol. 1999, 9, 1-10. 37 ii Sowv LzKi phase raca‘ 247i Zfiou M Shl asoh ataxi; fl lu34 Pen; Worr had: 1501 Data local 6nd“ Ahn, Intt 204E Che Pho res; 137' Shh how- 053 300 Cra Elie Ehj file ma! 87. 88. 89. 90. 91. 92. 93. 94. 95. Sorensen, C. S.; Syljuasen, R. G.; Falck, J.; Schroeder, T.; Ronnstrand, L.; Khanna, K. K.; Zhou, B.-B.; Bartek, J.; Lukas, J., Chk1 regulates the S phase checkpoint by coupling the physiological turnover and ionizing radiation-induced accelerated proteolysis of Cdc25A. Cancer Cell 2003, 3, 247-258. Zhou, B.-B. S.; Chaturvedi, P.; Spring, K.; Scott, S. P.; Johanson, R. A.; Mishra, R.; Mattern, M. R.; Winkler, J. D.; Khanna, K. K., Caffeine abolishes the mammalian G2/M DNA damage checkpoint by inhibiting ataxia-telangiectasia-mutated kinase activity. J. Biol. Chem. 2000, 275, 1 0342-1 0348. Peng, C.-Y.; Graves, P. R.; Thoma, R. 8.; Wu, 2.; Shaw, A. S.; Piwnica- Worms, H., Mitotic and G2 checkpoint control: regulation of 14-3-3 protein binding by phosphorylation of CchSC on serine-216. Science 1997, 277, 1501-1505. Dalal, S. N.; Schweitzer, C. M.; Gan, J.; DeCaprio, J. A., Cytoplasmic localization of human cdc25C during interphase requires an intact 14-3-3 binding site. Mol. Cell. Biol. 1999, 19, 4465-4479. Ahn, J.; Urist, M.; Prives, C., Questioning the Role of Checkpoint Kinase 2 in the p53 DNA Damage Response. J. Biol. Chem. 2003, 278, 20480- 20489. Chehab, N. H.; Malikzay, A.; Stavridi, E. S.; Halazonetis, T. D., Phosphorylation of Ser—20 mediates stabilization of human p53 in response to DNA damage. Proc. Natl. Acad. Sci. U. S. A. 1999, 96, 1 3777-13782. Shieh, S.-Y.; Ahn, J.; Tamai, K.; Taya, Y.; Prives, C., The human homologs of checkpoint kinases Chk1 and Cds1 (Chk2) phosphorylate p53 at multiple DNA damage-inducible sites. Genes Dev. 2000, 14, 289- 300. Craig, A.; Scott, M.; Burch, L.; Smith, G.; Ball, K.; Hupp, T., Allosteric effects mediate CHK2 phosphorylation of the p53 transactivation domain. EMBO Rep. 2003, 4, 787-792. Hirao, A.; Cheung, A.; Duncan, G.; Girard, P.-M.; Elia, A. J.; Wakeham, A.; Okada, H.; Sarkissian, T.; Wong, J. A.; Sakai, T.; De Stanchina, E.; Bristow, R. G.; Suda, T.; Lowe, S. W.; Jeggo, P. A.; Elledge, S. J.; Mak, T. W., Chk2 is a tumor suppressor that regulates apoptosis in both an ataxia telangiectasia mutated (ATM)-dependent and an ATM-independent manner. Mol. Cell. Biol. 2002, 22, 6521-6532. 38 35' ”Di, -..L C) U? lake Andi sa‘V‘r tad}: 1 l Bari can SLI mmCUZID 0’ mmrnn 96. 97. 98. 99. 100. 101. 102. 103. 104. 105. Takai, H.; Naka, K.; Okada, Y.; Watanabe, M.; Harada, M; Saito, S. L; Anderson, C. W.; Appella, E.; Nakanishi, M.; Suzuki, H.; Nagashima, K.; Sawa, H.; Ikeda, K.; Motoyama, N., Chk2-deficient mice exhibit radioresistance and defective p53-mediated transcription. EMBO J. 2002, 21 , 5195-5205. Bartek, J.; Lukas, J., Chk1 and Chk2 kinases in checkpoint control and cancer. Cancer Cell 2003, 3, 421-429. Jallepalli, P. V.; Lengauer, C.; Vogelstein, B.; Bunz, F., The Chk2 Tumor Suppressor Is Not Required for p53 Responses in Human Cancer Cells. J. Biol. Chem. 2003, 278, 20475-20479. Oliver, A. W.; Paul, A.; Boxall, K. J.; Barrie, S. E.; Aherne, G. W.; Garrett, M. D.; Mittnacht, S.; Pearl, L. H., Trans-activation of the DNA-damage signalling protein kinase Chk2 by T-loop exchange. EMBO J. 2006, 25, 3179-3190. Matthews, D. J.; Yakes, F. M.; Chen, J.; Tadano, M.; Bornheim, L.; Clary, D. 0.; Tai, A.; Wagner, J. M.; Miller, N.; Kim, Y. D.; Robertson, 8.; Murray, L.; Karnitz, L. M., Pharmacological abrogation of S-phase checkpoint enhances the anti-tumor activity of gemcitabine in vivo. Cell Cycle 2007, 6, 104-110. Preparation of 3-aminoindazoles as CHK1/CHK2 kinase inhibitors. 2006- 1020060051791020060051 79, 20060206., 2007. Janetka, J. W.; Ashwell, S., Checkpoint kinase inhibitors: a review of the patent literature. Expert Opin. Ther. Pat. 2009, 19, 165-197. Mederski, W.; Gericke, R.; Klein, M.; Beier, N.; Lang, F. Preparation of 3- oxoindazolyl-substituted squaric acid derivatives as CHK1, CHK2 and/or SGK kinase inhibitors. 2006-EP76502007022858, 20060802., 2007. Arienti, K. L.; Brunmark, A.; Axe, F. U.; McClure, K.; Lee, A.; Blevitt, J.; Neff, D. K.; Huang, L.; Crawford, 8.; Pandit, C. R.; Karlsson, L.; Breitenbucher, J. G., Checkpoint Kinase Inhibitors: SAR and Radioprotective Properties of a Series of 2-Arylbenzimidazoles. J. Med. Chem. 2005, 48, 1873- 1885. Carlessi, L.; Buscemi, G.; Larson, G.; Hong, Z.; Wu, J. 2.; Delia, D., Biochemical and cellular characterization of VRX0466617, a novel and selective inhibitor for the checkpoint kinase Chk2. Mol. Cancer Ther. 2007, 6, 935-944. 39 106. 107. 108. 109. 110. 111. 112. 113. 114. Jobson, A. G.; Cardellina, J. H., II; Scudiero, D.; Kondapaka, S.; Zhang, H.; Kim, H.; Shoemaker, R.; Pommier, Y., Identification of a bis- guanylhydrazone [4,4'-diacetyldiphenylurea-bis(guanylhydrazone); NSC 109555] as a novel chemotype for inhibition of Chk2 kinase. Mol. Pharmacol. 2007, 72, 876-884. Larson, G.; Yan, S.; Chen, H.; Rong, F.; Hong, Z.; Wu, J. 2., Identification of novel, selective and potent Chk2 inhibitors. Bioorg. Med. Chem. Lett. 2007, 17, 172-175. McClure, K. J.; Huang, L.; Arienti, K. L.; Axe, F. U.; Brunmark, A.; Blevitt, J.; Guy Breitenbucher, J., Novel non-benzimidazole chk2 kinase inhibitors. Bioorg. Med. Chem. Lett. 2006, 16, 1924-1928. Neff, D. K.; Lee-Dutra, A.; Blevitt, J. M.; Axe, F. U.; Hack, M. D.; Buma, J. C.; Rynberg, R.; Brunmark, A.; Karlsson, L.; Breitenbucher, J. G., 2-Aryl benzimidazoles featuring alkyl-linked pendant alcohols and amines as inhibitors of checkpoint kinase Chk2. Bioorg. Med. Chem. Lett. 2007, 17, 6467-6471. Zabludoff, S. D.; Deng, C.; Grondine, M. R.; Sheehy, A. M.; Ashwell, S.; Caleb, B. L.; Green, S.; Haye, H. R.; Horn, C. L.; Janetka, J. W.; Liu, D.; Mouchet, E.; Ready, 8.; Rosenthal, J. L.; Queva, C.; Schwartz, G. K.; Taylor, K. J.; Tse, A. N.; Walker, G. E.; White, A. M., AZD7762, a novel checkpoint kinase inhibitor, drives checkpoint abrogation and potentiates DNA-targeted therapies. Mol. Cancer Ther. 2008, 7, 2955-2966. Kohn, E. A.; Yoo, C. J.; Eastman, A., The Protein Kinase C Inhibitor Goe6976 Is a Potent Inhibitor of DNA Damage-induced S and G2 Cell Cycle Checkpoints. Cancer Res. 2003, 63, 31-35. Yu, 0.; La Rose, J.; Zhang, H.; Takemura, H.; Kohn, K. W.; Pommier, Y., Ucn-01 inhibits p53 up-regulation and abrogates g-radiation-induced G2-M checkpoint independently of p53 by targeting both of the checkpoint kinases, chk2 and chk1. Cancer Res. 2002, 62, 5743-5748. Lara, P. M, Jr.; Mack, P. C.; Synold, T.; Frankel, P.; Longmate, J.; Gumerlock, P. H.; Doroshow, J. H.; Gandara, D. R., The Cyclin- Dependent Kinase Inhibitor UCN-01 Plus Cisplatin in Advanced Solid Tumors: A California Cancer Consortium Phase I Pharmacokinetic and Molecular Correlative Trial. Clin. Cancer Res. 2005, 11 , 4444—4450. Akinaga, S.; Nomura, K.; Gomi, K.; Okabe, M., Enhancement of antitumor activity of mitomycin C in vitro and in vivo by UCN-01, a selective inhibitor of protein kinase C. Cancer Chemo. Pharmacol. 1993, 32, 183-9. 40 115. 116. 117. 118. 119. 120. 121. 122. 123. 124. Pollack, I. F.; Kawecki, S.; Lazo, J. S., Blocking of glioma proliferation in vitro and in vivo and potentiating the effects of BCNU and Cisplatin: UCN- 01, a selective protein kinase C inhibitor. J. Neurosurg. 1996, 84, 1024- 1032. Wang, 0.; Fan, 8.; Eastman, A.; Worland, P. J.; Sausville, E. A.; O'Connor, P. M., UCN-01: A potent abrogator of G2 checkpoint function in cancer cells with disrupted p53. J. Natl. Cancer Inst. 1996, 88, 956-965. Hu, B.; Zhou, X. Y.; Wang, X.; Zeng, 2. C.; lliakis, G.; Wang, Y., The radioresistance to killing of A1-5 cells derives from activation of the Chk1 pathway. J. Biol. Chem. 2001, 276, 17693-8. Hirose, Y.; Berger, M. S.; Pieper, R. O., Abrogation of the Chk1-mediated G2 checkpoint pathway potentiates temozolomide-induced toxicity in a p53-independent manner in human glioblastoma cells. Cancer Res. 2001, 61 , 5843-5849. Fuse, E.; Tanii, H.; Kurata, N.; Kobayashi, H.; Shimada, Y.; Tamura, T.; Sasaki, Y.; Tanigawara, Y.; Lush, R. D.; Headlee, D.; Figg, W. D.; Arbuck, S. G.; Senderowicz, A. M.; Sausville, E. A.; Akinaga, S.; Kuwabara, T.; Kobayashi, S., Unpredicted clinical pharmacology of UCN-01 caused by specific binding to human a1-acid glycoprotein. Cancer Res. 1998, 58, 3248-3253. Sausville, E. A.; Arbuck, S. G.; Messmann, R.; Headlee, D.; Bauer, K. S.; Lush, R. M.; Murgo, A.; Figg, W. D.; Lahusen, T.; Jaken, S.; Jing, X.-x.; Roberge, M.; Fuse, E.; Kuwabara, T.; Senderowicz, A. M., Phase I trial of 72-hour continuous infusion UCN-01 in patients with refractory neoplasms. J. Clin. Oncol. 2001, 19, 2319-2333. Fuse, E.; Kuwabara, T.; Sparreboom, A.; Sausville, E. A.; Figg, W. D., Review of UCN-01 development: a lesson in the importance of clinical pharmacology. J Clin Pharmacol 2005, 45, 394-403. Sharma, V.; Tepe, J. J., Potent inhibition of checkpoint kinase activity by a hymenialdisine-derived indoloazepine. Bioorg. Med. Chem. Lett. 2004, 14, 4319-4321. Mosey, R. A.; Tepe, J. J., New synthetic route to access (+-)- salinosporamide A via an oxazolone-mediated ene-type reaction. Tetrahedron Lett. 2009, 50, 295-297. Mosey, R. A.; Fisk, J. S.; Friebe, T. L.; Tepe, J. J., Synthesis of tert-Alkyl Amino Hydroxy Carboxylic Esters via an Intermolecular Ene-Type Reaction of Oxazolones and Enol Ethers. Org. Lett. 2008, 10, 825-828. 41 125. 126. 127. 128. 129. 130. 131. 132. 133. 134. 135. Fisk, J. S.; Mosey, R. A.; Tepe, J. J., The diverse chemistry of oxazol-5- (4H)-ones. Chem. Soc. Rev. 2007, 36, 1432-1440. Fisk, J. S.; Tepe, J. J., Intermolecular Ene Reactions Utilizing Oxazolones and Enol Ethers. J. Am. Chem. Soc. 2007, 129, 3058-3059. Sharma, V.; Peddibhotla, S.; Tepe, J. J., Sensitization of Cancer Cells to DNA Damaging Agents by Imidazolines. J. Am. Chem. Soc. 2006, 128, 9137-9143. Sharma, V.; Tepe, J. J., Diastereochemical diversity of imidazoline scaffolds via substrate controlled TMSCl-mediated cycloaddition of azlactones. Org. Lett. 2005, 7, 5091-5094. Keni, M.; Tepe, J. J., One-Pot Friedel-Crafts/Robinson-Gabriel Synthesis of Oxazoles Using Oxazolone Templates. J. Org. Chem. 2005, 70, 4211- 4213. Sharma, V.; Lansdell, T. A.; Peddibhotla, S.; Tepe, J. J., Sensitization of Tumor Cells toward Chemotherapy: Enhancing the Efficacy of Camptothecin with Imidazolines. Chem. Biol. 2004, 11 , 1689-1699. Peddibhotla, S.; Tepe, J. J., Stereoselective Synthesis of Highly Substituted Delta 1-Pyrrolines: exo—Selective 1,3-Dipolar Cycloaddition Reactions with Azlactones. J. Am. Chem. Soc. 2004, 126, 12776-12777. Sharma, V.; Tepe, J. J., Potent inhibition of checkpoint kinase activity by a hymenialdisine-derived indoloazepine. Bioorg. Med. Chem. Lett. 2004, 14, 4319-4321. Sharma, V.; Lansdell, T. A.; Jin, G.; Tepe, J. J., Inhibition of Cytokine Production by Hymenialdisine Derivatives. J. Med. Chem. 2004, 47, 3700- 3703. Peddibhotla, S.; Tepe, J. J., Multicomponent synthesis of highly substituted imidazolines via a silicon mediated 1,3-dipolar cycloaddition. Synthesis 2003, 1433-1440. Peddibhotla, S.; Jayakumar, S.; Tepe, J. J., Highly Diastereoselective Multicomponent Synthesis of Unsymmetrical Imidazolines. Org. Lett. 2002, 4, 3533-3535. 42 l 1 ‘Wfi I 5;" Saul; Tel. '19 I ‘5’7'; for Ci‘E'Eht "31W Li88 Va lidazol CHAPTER II DEVELOPMENT OF A SYNTHETIC METHOD FOR THE PREPARATION OF 2-AMINO-5, 5-DISUBSTITUTED-1H-lMlDAZOL-4(5H)-ONES II.A Nomenclature and numbering of imidazolones The general structure for a 2-amino-imidazolone is shown in Figure “-1. It can be observed that there are actually three tautomeric forms for a 2-amino- imidazolone where the double bond is switched between the three nitrogens making up the guanidine portion of the heterocycle. The preferred tautomeric form for the imidazolone was shown by Matsumoto and co-workers1 and in a different set of experiments by Kenyon and co-workers2 to be tautomer "-2. Traditional nomenclature for these types of molecules encountered in literature has varied,3 with names such as 2-amino-imidazoI-4-ones (2-amino- imidazolone), glycocyamidines and 2-iminohydantoins being some of the more recent common terms. The numbering of the ring system varies depending on the tautomer specified as shown in Figure "-1.3 When the double bond is exocyclic as in the case of "-1 or when the double bond is endocyclic and in conjugation with the carbonyl (II-2), the numbering of the ring begins with the nitrogen furthest from the carbonyl and is given position 1. Numbering the ring clockwise from that point will result in the second nitrogen contained in the ring being in position 3, the carbonyl carbon in position 4 and the methylene carbon in position 5.3 43 HI \ l - \v-I- fps v, ' I ‘aZfi. HN "-1 "-2 "-3 Figure "-1. Numbering of the 2-amino-imdazoI-4-one ring system However, when the double bond is endocyclic and not in conjugation with the carbonyl (II-3), the numbering of the ring system starts with the opposite nitrogen in the ring. A counter-clockwise numbering scheme follows which puts the second nitrogen in position 3, the methylene in position 4 and the carbonyl carbon in position 5.3 When an amino group is not present in the 2 position, the structure is still considered an imidazolone, however, it simply contains a different substitution at the 2 position. For the purposes of consistency, the term imidazolone will be used for the nomenclature of the general ring structure. When substitution of the imidazolone ring is present, the numbering scheme will follow that for structures "-1 and "-2, which is the most commonly accepted numbering format.3 In addition, the name of the substituent will follow the number at which it is found. For example, structure "-4 in Figure "-2, would have the name 2- aminoimidazolone where structure "-5 (also in Figure "-2) would have the name 2, 5, 5-trimethylimidazolone. HN "-4 “-5 Figure "-2. Representative structures of imidazolones 44 vet: "J's a later a fiDldi 01 ".3 Reactivity of imidazolones The 2—aminoimidazolone ring system can undergo a number of reactions in a variety of locations such as at the carbonyl carbon, alpha to the carbonyl and at the guanidine nitrogens (Figure Il-3).3'7 H2N N Wig: o HZN HN/N NH Oxidation JL OH Reduction H N N 2 H/\[()]/ or H drol sis , Alkylation H2 N VCOZH < y y HZNJ/f / H2N N or RX 7 11:):0 H O YN O Condensation Acylation HN HZNHNKHORJLH :fSI/fo R R Figure "-3. Reactions of 2-aminoimidazolone Alkylation of imidazolones using alkyl halides such as methyl iodide occurs at N-3 primarily, although if N-1 and N-3 are already alkylated, N-2 can be alkylated.2 It was shown by Kenyon and co-workers that the nitrogen in position 3 is more reactive and the alkylated product can undergo a reversible rearrangement to put the double bond back into conjugation with the carbonyl, affording N-2 bearing the alkyl group (Scheme "-1).2 45 Scheme "-1. Alkylation of 2-aminoimidazol-4-one Mel, EtOH _ H2 HZN /N N N/ Hl/fo 90% If f0 'lMeOH J1 J. \ U n be *— 0 1r, \ Further support for N-3 being the most basic nitrogen, and hence most nucleophilic, comes from a study by Reddick and co-workers where they identified via 15N-NMR that the nitrogen in position 3 is primarily protonated at a low pH.8 Acylation reactions occur in a similar fashion?" 5 lmidazolones can also undergo a condensation reaction with aldehydes producing a 5-arylidene imidazolone (Figure ll-3).3' 4' 9'12 The success of the reaction is dependent on the nucleophilic character of the imidazolone and the electrophilic character of the aldehyde. Furthermore, the reaction often produces a mixture of E/Z isomers, some of which can isomerizes to one other depending on the product and conditions.3' 4 Alkylation is also possible at carbon 5 of an 2-aminoimidazolone as shown by Danishefsky and co-workers (Scheme ”-2).13 The alkylation was completed using a imidazolone core similar to "-1 (Figure “-1) and a benzyl bromide giving the quaternary imidazolone in 91% yield.13 46 Scheme "-2. Alkylation at C-5 of an imidazolone OMe OMe /N LiHMDS, THF, 91% /N O 03” o Hydrolysis of the 2-aminoimidazolone ring can lead to a variety of products depending on the reaction conditions (Figure II-3).3' 4' 6 Cleavage of the amide bond of the heterocyclic ring can lead to products that resemble a glycine derivative with a guanidine moiety instead of a primary amine. It has also been observed that under certain conditions, the imidazolone ring can hydrolyze at a different position and form urea and glycine.3 Lastly, hydrolysis can also transform the 2-aminoimidazolone into the hydantoin analog under basic conditions.4' 6 Oxidation of the methylene group at position 5 of the heterocyclic ring is possible and the resulting dicarbonyl compound is formed (Figure ”-3). However, these reactions are often run under alkali conditions and the resulting dicarbonyl heterocycle undergoes further chemical modifications and is known to hydrolyze giving the oxalate and derivatives of guanidine.3 Reduction of the carbonyl at position 5 can occur using hydrogenation under a platinum catalyst. The typical reaction conditions include 50 mol% of a platinum catalyst and a hydrogen pressure of 15-35 psi. The result of this reaction is the formation of cyclic guanidine structures (Figure "-3).7 The acidity of imidazolones have been studied by a few different groups." 3' ‘4 Chandrasekhar and co-workers compared the relative rates of a base 47 catalyzed deuterium exchange for an imidazolone, oxazolone and pyrrolone. It was found that the rate of deuterium exchange was greatest for the imidazolone and worst for the pyrrolone (Figure ”-4),14 leading to conclude that based on these results the imidazolone is more acidic than the other analogs tested. N N lmidazolone Oxazolone Pyrrolone Figure "-4. Relative rates of deuterium exchange The pKa’s of multiple 2-aminoimidazolone hydrochloride compounds have been identified initially by Matsumoto and co-workers and then to a greater extent by Kenyon and co-workers.1' 2 It was found that the pKa’s of the protonated imidazolones range from 4.48-9.01 (in H20) depending on the substitution on the heterocyclic ring. There is a clear distinction between the pKa’s of imidazolones that contain the double bond in conjugation with the carbonyl group and those that do not, such as between "-8 and "-9 (Figure ”-5).2 - r 1 F '1 r 1 HCI- HCI- I HCI- I HCI- / HZN\I4\N)=OW /NYN/ /N\l./’N /N\j/N O | “N Mfo HN\)=O fo ll-6 _ "-7 a _ II-8 _ L "-9 _ 9K3 = 4-30 pKa = 7.57 pKa = 4.48 pKa = 9-01 Figure "-5. pKa’s of differently substituted 2-aminoimidazolones ll.C Natural products containing the imidazolone core There are many accounts of natural products that contain the imidazolone core in a variety of different forms.”28 One of the most common imidazolones is an end product of nitrogen metabolism in the human body, a compound called 48 creatinine (Figure ”-6).8 Hymenialdisine is a natural product that contains the 2- aminoimidazolone core with an alkenic substitution at position 5 of the heterocyclic core (Figure ”-6). The compound comes from the sponges AxineI/a verrucosa and Acanthella aurantiaca and was isolated in 198218 and first synthesized in 1995.29 De-bromohymenialdisine, an analog of hymenialdisine without the bromine on the pyrrole ring, also contains the imidazolone core. Dispacamide 1 is a novel bromo pyrrole alkaloid that was isolated from a Caribbean sponge in 199616 and was synthesized in 1997.30 Debromodispacamides B and D were isolated from the marine sponge Age/as mauritiana and synthesized in 200827 , while the related compounds polyandrocarpamines A and B were isolated from the ascidian Polyandrocarpa sp. and synthesized in 2002.19 All of the dispacamide and polyandrocarpamine compounds exhibit the same type of imidazolone core that hymenialdisine possesses, the 2-aminoimidazoI-4-one core with an alkenic substitution pattern at carbon 5 (Figure "-6). The last members of natural products that contain this type of heterocyclic core are the Ieucettamines B and C and Ieucettamidine (Figure ”-6). Leucettamine B and Ieucettamidine were isolated from the sponge Leucetta microraphis in 1993,17 while Ieucettamine C was isolated from Leucetta avocado in 2003.31 To date, only Ieucettamine B has been synthesized with the first synthesis reported in 1994.32 49 an N: L t :1 HCH‘ ‘ vvuvULUK My“ Jv 5 In... 3d pk v dflfias; HZNYffO \ N / Br / \ Creatinine N NH H O Hymenialdisine O \ O / O n 1 .— N \ N Br yNH H \ / HZN N o HzN . . ) Dispacamide 1 Br Leucettamine B 0 Figure "-6. Natural products containing the 2-aminoimidazolone core Natural products that contain the imidazolone core without the 2-amino substitution have also been reported.15' 23' 25 Rhopaladins A-D (Figure "-7) contain a unique imidazolone core consisting of an alkenic substitution at carbon 5 and an acyl group at carbon 2. These natural products come from the Okinawan tunicate Rhopalaea sp. and were isolated in 1998.25 The first synthesis of rhopaladin D was completed in 200033, while the total synthesis of rhopaladins A-C were completed shortly after in 2002.34 Kottamides A-D (Figure "-7) are structurally unusual due to the quaternary substitution pattern on carbon 2. These natural products are optically active, were isolated from the New Zealand ascidian Pycnoclavella kottae in 2002 and have not been synthesized to date.15 The last example of a natural product that has the imidazolone core but lacking the 2-amino functionality is a novel bis(indole) alkaloid that was isolated from the sponge Dragmacidon sp. in 1996. The structure of this compound (Figure "-7) is very unique with a quaternary 50 center at carbon 5 and has also not been synthesized to date. Due to the light absorbing tendencies of the compound in solution, chiroptical data was hard to establish.23 HO Kottamide A Novel bis(lndole) alkaloid Figure "-7. Natural products containing an imidazolone core The next group of natural products contains the imidazol—4-one heterocyclic core and also the 2-amino functionality. However, these compounds do not contain the alkenic substitution pattern at carbon 5 as those in Figure "-8. Instead, most of these natural products contain a quaternary center in the imidazolone cycle. Compound 2096A (Figure "-8) and an analog 20963, a diastereomer of 2096A, were isolated in 2000 from extracts of Streptomyces sp.28 It is found that these two compounds can interconvert due to the acidity of the proton at the quaternary center of the bicyclic heterocycle and as a result, the stereochemistry is not defined. Additionally, the compounds were found to decompose readily in aqueous solutions making the synthesis of these 51 “0qu Iva- NIP J 3an let's; Fl compounds a challenge.28 To date, there are no reported syntheses of 2096A or B. Oxysceptrin (Figure "-8) and related alkaloids have been isolated from the sponge Age/as conifera in 1991 .35' 36 Oxysceptrin contains an unique cyclobutane core flanked by imidazole, pyrrole and imidazolone appendages. The synthesis of oxysceptrin as well as other alkaloids from the same family was completed by O’Malley and co-workers in 2007.37 2096A and B Indole alkaloid (:l:)-CalcaridineA (III-1) kN O H2N Mauritlamine Figure "-8. Additional natural products containing 2-aminoimidazolone Another natural product related to oxysceptrin is mauritiamine, a compound with a quaternary imidazolone containing imidazole and pyrrole appendages at position 5 (Figure ”-8). The natural product comes from the marine sponge Age/as mauritiana and was isolated in 1996. Furthermore, the 52 natural product contains a quaternary center but is optically inactive and was isolated at a racemate.26 The first racemic synthesis of mauritiamine was completed by Olofson and co-workers in 1997 utilizing an oxidative dimerization of two imidazole containing intermediates.38 Calcaridine A (Figure ”-8) was isolated from a Fijian sponge Leucetta sp. in 2003.20 The natural product contains a uniquely substituted 2-aminoimidazol-4- one core with a quaternary center at carbon 5. The natural product is optically active although the absolute stereochemistry is not yet defined.20 The first racemic total synthesis of calcaridine A was completed by Koswatta and co- workers in 2008 using a novel rearrangement to afford the quaternary stereocenter.39 The last natural product that contains the 2-aminoimidazol-4-one core and also a quaternary center at carbon 5 is the indole alkaloid (Ill-1) shown in Figure ”-8. It was isolated from the tunicate Dendrodoa grossularia in 1998 and the stereochemistry at the quaternary carbon is also not defined.22 Further detail on this molecule is given in chapter Ill. The synthesis of this natural product is the goal of my project and thus a synthetic route to form the uniquely substituted quaternary imidazolone is needed. ".0 Synthetic routes to imidazolones lmidazolones with and without a quaternary center at position 5 have been synthesized in a variety of ways.”‘73 The following section illustrates the various routes that have been used previously to obtain imidazolones as well as the routes that were attempted to create quaternary imidazolones for synthesizing 53 the indole alkaloid, the focal point of my project. Finally, the new synthetic method developed for gaining access to the quaternary imidazolone scaffold needed for the total synthesis of the indole alkaloid will be discussed. II.D.1 Synthetic routes to imidazolones: Aza-wittig reaction The first route that will be discussed is the synthesis of imidazolones via an intramolecular aza-wittig reaction (Scheme ”-3). Takeuchi and co-workers developed an efficient aza-wittig reaction which reacts an azide and triphenylphosphine to obtain an intermediate that attacks the carbonyl of the imide to produce imidazolones in good yields.69 They were able to use alkyl, aryl and heteroaryl groups at R1; alkyl and aryl groups in the R2 position; and hydrogen and aryl groups in the R3 position. The reaction proceeds at room temperature in two hours and produces yields ranging from 70-99%.69 Scheme "-3. Aza-wittig reaction to produce imidazolones *- 1 R O 0 PPh )3: O cPPh3 0 hi 2 1 l benzene, 1 I />‘R1 R2 R3 rt R2 R3 R3 N 70-99% ".02 Synthetic routes to imidazolones: Reaction of aziridinone and thiourea Talaty and co-workers use a reaction between an aziridinone and a thiourea to produce imidazolones in yields up to 55% (Scheme ”—4).70 The aziridinones were substituted at the R1 and R2 position with either a tert-butyl or adamantly substituent and reacted with a thiourea to attack the carbonyl of the aziridinone. After the ring opening of the aziridinone it is proposed that the reaction eventually produces a guanidine thioester intermediate.7O After a final 54 attack of the guanidine on the thioester, the imidazolone product is proposed to be formed.70 Scheme “-4. Reaction of aziridinones and thiourea to form imidazolones -I o f o 69NH2 0 / N~ THF reflux r ,N R2 R1 . R1/ No R1 Nl H‘ngz O 0 O sz/lL RBKKIN L R2 —§NH N i: N ‘ N ; R/ R? NH2 R’1 NH 1 Y II.D.3 Synthetic routes to imidazolones: Oxazolone to imidazolone Oxazolones have been used to produce imidazolones using a thiourea as well (A, Scheme “-5).” 64' 65 Tepe and co-workers found that by treating oxazolones with a thiouronium salt under basic conditions, they were able to isolate 28% of the corresponding imidazolone.“ 65 Flygare and co-workers developed a solid-phase synthesis of imidazolones using the same type of chemistry giving yields ranging from 46-91% yields (B, Scheme "-5).49 The oxazolone consisted of aromatic R1 substituents while the thiourea contained alkyl and benzyl R2 substituents.49 55 Scheme "-5. lmidazolones using oxazolones and thioureas N O s’Bn H2N~ x ,,> H H2N N H B 0 NH2 00 HN/ngo H20 HN/lggk'N HNékfi 215:0 HNéLfi {3H 71% II.D.5 Synthetic routes to imidazolones: Diketone rearrangement Chalcones, a,B-unsaturated ketones, o,B—epoxy ketones and 1,2- diketones have been reported to undergo a rearrangement with guanidine and substituted guanidines to form imidazolones.4°'43' 62' 7‘ Darvas and co-workers reported that a, B-unsaturated ketones (R1 and R2 = aryl or heteroaryl) rearrange with guanidine to form imidazolones in yields ranging from 29-87% (A, Scheme "-7).71 Dhar et al. reported that a similar reaction occurs with a, B-epoxy ketones (R1 and R2 = aryl) under basic conditions with yields between 40-83% (B, Scheme “-7).43 The rearrangement of 1,2-diketones with guanidines to form imidazolones has been studied quite thoroughly.4°42' 62' 63' 75' 76 Nishimura reported the formation of imidazolones from the reaction of diketones with guanidine and 1, 1-disubstituted guanidines with yields ranging from 70-92% (C, 57 Scheme "-7).62 Like the previous examples, R1 and R2 were mostly aryl.62 However, when R1 = methyl the rearrangement failed to produce the imidazolone and made a deep colored solution, which was thought to have gone through a 62. 75 completely different reaction. Scheme "-7. lmidazolones from 1, 2-diketone and guanidine O G 'd' H o HZNYN / uanI me, 2 2 _ / O A R1/U\/\R2 o r HN KOH, EtOH, 80 c 2n,29-87% R1 R2 0 H2N M Guanidine ‘ YN O 3 R1 0 R2 NaH, THF, reflux ”N7: 4-12 h, 40-83% R, R 2 o H2N MR2 Guanidine _ YN O C R1 ROH,A,2-4h, ”N72; 0 70-92% R1 The proposed mechanism of the reaction between o,B-unsaturated ketones, a,B-epoxy ketones and 1,2-diketones with guanidine is related to each other based on the intermediates proposed to be formed throughout the mechanism.“ 62' 71 It is proposed that when the starting material is an 0,8- unsaturated ketone, the mechanism starts with the formation of an o,j3-epoxy ketone using hydrogen peroxide. Next, a base mediated rearrangement occurs from the epoxy ketone to afford a 1,2-diketone.77'81 Subsequently, the guanidine then reacts with a diketone to produce intermediates that rearrange to the final imidazolone (Scheme "-8).“ 62' 71 58 Scheme "-8. Proposed mechanism for diketone rearrangement I- -I o - o 1 o / H202 0 Base 0 O O O O 0 D Guanidine HZNYN )N\HZ NHZ HN O N \N o N \N o \ CO or O \ O When the starting material is an (LB-epoxy ketone, the mechanism is truncated and begins with the rearrangement of the epoxy ketone to a 1,2- diketone and continues on as shown in Scheme “-8. Furthermore, when starting with the 1,2-diketone, the rearrangement occurs after the guanidine forms the heterocyclic intermediate with the diketone. In summary, whether the starting material is an a,j3-unsaturated ketone, (LB-epoxy ketone or 1,2-diketone, the reaction mechanism consists of similar intermediates and simply begins at different points in the mechanism shown in Scheme ”-8. Alternatively, it is proposed by Nishimura that when an alkyl group is used in the rearrangement (R1 = methyl; see C, Scheme "-7) a different reaction occurs (Scheme "-9).” 75 It is thought that instead of rearranging to the imidazolone, the methyl group is deprotonated under the basic conditions leading to intermediate "-10 (Scheme "-9). After addition of water to form "-12, it is surmised that the imidazole is transformed into a diimidazole compound (ll-13) 59 similar to the Voges-Proskauer pigment, which is red in color.75 Although the exact mechanism of this transformation is not fully understood, it offers an explanation to the red color observed when Nishimura attempted the rearrangement with a methyl substituent. Scheme "-9. Alternative mechanism for diketone rearrangement '- '7 R2 HO R2 Ho 9 R2 CH3 Aqueous H042 H / e / ——. N N /N Base N /N N / Y Y Y "-10 R1 R1 R1 R R 2 2 R2 R2 NWN H_ 7—1— ‘— N Ne :>\/NH N\50 2.uw, 10-15 min, 77-98% R R3 3 Liu used microwave technology to speed up the dehydration process and produce imidazolones in 10-15 min,72 where Gillman’s reactions took 2 h.53 Liu produced a diamide intermediate in a different fashion as Gillman and used the energy of microwaves to effect the dehydration and produce quaternary imidazolones (Scheme Il-12). R1 and R2 for the majority of the examples Liu illustrated were connected through a cyclic alkane producing spiro-imidazolones 65 as the final product in yields ranging from 77-98%.72 There were a few cases were R1 and R2 were alkyl and not tethered together and those yields were between 85-95%. Additionally, R3 and R4 were varied between alkyl, phenyl and benzyl and afforded imidazolones in yields between 77-98%. The yields reported were determined by HPLC from LC-MS results of the reaction mixture. The isolated yields of the imidazolones greatly decreased and were reported to be between 36-78%.72 II.D.7 Synthetic routes to imidazolones: Cyclization with carbodiimide intermediates The main type of reactions used to produce imidazolones are cyclizations where a carbodiimide intermediate is attacked internally or externally by a nucleophile, which causes a subsequent formation of the imidazolone product.“ 46' 54' 55' 6° The main differences in most of the examples that use this type of ring closure is in the nucleophile used or if similar nucleophiles are used, such as amines to produce a guanidine moiety, then the difference is in the formation of that guanidine moiety. Lee et al. and Lange et al. have used the dehydration of ureas with triphenylphosphine/Bromine and Burgess’ reagent, respectively, to produce in situ a carbodiimide intermediate, which is attacked by an external nucleophile.55' 6° Lee used alkyl and benzyl substituents for R2; phenyl and heteroaryl grignards for the nucleophile (R3); and benzyl and phenyl substituents for R4 (Scheme II- 13). Yields ranged from 56-99% and reactions took about 2 h to complete. There were no reports on producing any quaternary imidazolones.55 On the other hand, Lange used a solid phase technique where R1 was connected to a solid support, 66 which helped in the isolation of the imidazolones (Scheme ll-13).60 Benzyl and alkyl substituents were used for R2, while R3 tended to be various amines giving products that were a variety of 2-aminoimidazolones. R4 was strictly confined to aryl groups and the yields ranged from 47-93°/o.60 Scheme "-13. Cyclization from diimide: urea to imidazolone - 2 0 ,R4 1.dehydrating ,R4 R2 '1 agent _ O R2 1 -R1OH R2 N’R4 RI/OVN O 2. Nucleo. (R3) R1/ Wj/kN R3 solvent, rt, NZ< O H O H R3 Drewry and co-workers used a thiourea instead of a urea to form the guanidine moiety. A desulfurization of the thiourea using Mukaiyama’s reagent led to a carbodiimide intermediate which was attacked by various amines to produce 2-aminoimidazolones.46 Like Lange, Drewry used a solid support connected at R1 allowing an easier isolation of the resulting imidazolones. Furthermore, R2 was typically a benzyl group; R3 was a range of primary and secondary amines; and R4 was primarily aryl groups (Scheme ll-14). The reactions took longer than solution phase reactions with times around the 24 h mark and yields ranged from 34-94%.46 Scheme “-14. Cyclization from diimide: thiourea to imidazolone ' ' 0 ,R4 1.desulfurinzing ,R4 R2 HN agent R7- )NL -R1OH R2 N’R4 O k = /O ——» “1% R1’ N S 2. amine (R3) RI ” R3 solvent, rt, 0 H 0 34-94% R3 Aza-Wittig reactions have also been used to produce a carbodiimide intermediate, which ultimately gets attacked by a nucleophile to produce the 67 guanidine moiety (Scheme ll-15).45' 54 Villalgordo used the aza-Wittig reaction between an azide and an isocyanate to form a highly reactive carbodiimide intermediate. Subsequently, alkyl and benzyl amines, as well as, secondary amines were used to attack the carbodiimide, form the guanidine and cyclize on an ethyl ester to produce the 2-aminoimidazolone products. R2 was typically an alkyl or aryl substituent, while R4 was always aryl substituents. The yields for this reaction were between 72-89°/o and reaction times varied between 6-15 h.54 Ding used a similar technique to produce the guanidine and yields for the 2- aminoimidazolones ranged from 40-86%. Similar substituents to Villalgordo were used, i.e. primary and secondary amines, Aryl R4 substituents and alkyl and hydrogen at R245 Scheme "-15. Cyclization from diimide: azide to imidazolone R2 1. PPh3, DCM F R2 N’th O BOW/\N rt' 8 h = EtO i 'EtOH RZTQLN’R“ 3 2. isocyanate (R4) \ll/kfi R3 solvent, rt, N=< 0 DCM, rt, 2 h 0 R3 3. amine (R3) ‘ ‘ Alternatively, Batey and co-workers used an opposite approach to those described above. Batey utilized the reactivity of a thiourea to form a carbodiimide intermediate in situ using HgCl2, which was subsequently attacked internally by an amide, closing up to form an imidazolone (Scheme ”46).” 52 Multiple amino acid amides were used as starting materials allowing R2 to correspond to the appropriate amino acid. Peptides could be used in this reaction, allowing R1 to be a long peptidic chain, as well as linked to a solid phase support ameliorating the isolation process. R3 was typically aryl groups and yields ranged from 68-99%.47 68 Scheme "-16. Intramolecular cyclization: thiourea to imidazolone _ 2 O O O ,N. H H HgCl 20’ R3 R2¥LWR1 R1\ )KKN N. ___2_, R1\ )KrN : _ 1:1 \fl/ R3 n solvent, rt, N R2 3 R2 18h [NH _ _. R3 68-99% Frutos and co—workers used a dehydration of a urea to mediate the cyclization to an imidazolone product (Scheme ll-17).48 The urea was proposed to be transformed into a carbodiimide intermediate, which was spontaneously attacked intramolecularly by the secondary amide present in the molecule. As a result, the dehydration mediated by triphenylphosphine and carbon tetrachloride afforded the substituted imidazolone in a 81% yield.48 The quaternary substitution pattern at position 5 consisted of R1 and R2 being a benzyl and methyl group, while R3 was an aryl group.48 Scheme "-17. Intramolecular cyclization: urea to imidazolone 0 0 R2 R R R1 N, 3 PPh;,, col4 22(1LN'R3 H = R TEA, rt, 12 h, 1 — HMro 81% N—(NH HNVCOZEt EtOzC—/ II.D.8 Synthetic routes to imidazolones: Cyclization reactions with nitriles A second type of ring closure to form imidazolones involves the intramolecular cyclization of an amine or amide on a nitrile (Scheme ll-18). Nagasawa and Kwon describe a facile synthesis of imidazolones using a ring closure between an amine and nitrile using primarily heat although sometimes a little acid was used (A, Scheme “48).” 59 The amine moiety was protected with a carbamate group (Cbz) and the R2 and R3 substituents were varied between 69 hydrogen, alkyl, benzyl and phenyl. Quaternary imidazolones have been successfully prepared and yields for all imidazolones produced using this method ranged from 20-50%.58 The reactive intermediate for the reaction is not clear due to IR bands observed at 2140 cm'1 and 2260 cm", depending on the pH.58 A band at 2140 cm'1 hints at a carbodiimide intermediate, while the band at 2260 cm‘1 suggests more of a nitrile intermediate. The tautomeric relationship between the acylcyanamide and carbodiimide makes it hard to specify which intermediate is responsible for the cyclization. Regardless, the overall product in the reaction is variously substituted 2-aminoimidazolones. Lempert and co-workers report a similar cyclization where an amide attacks a nitrile and forms an imidazolone product (B, Scheme ll-18).66 Lempert successfully produces a variety of imidazolones, where R = alkyl, benzyl and differently substituted aryl groups, in yields ranging from 90-98%. It is also possible to remove the tert-butyl protecting group using aqueous HCI to provide t.66 the free 2-aminoimidazolone as the final produc Scheme "-18. lmidazolone through cyclization onto nitrile 0 R2 R3 H 3 A CBz\N)Kn/N\\ 1.solvent, A, 20-50%_ R Xk NH ' 2 I) H o \N 2. H2/Pd HN NH 0 5‘ 0 J4 P“ J< B //N>(1LN TEA , PhXLN N/ ph Ph H solvent, A, 90-98% N R NH 70 ll.D.9 Syn ryanogua siren-me seen-me m fiat, m ital lmic fi'aazolo were 789-2 Schem II.D.9 Synthetic routes to imidazolones: Cyclization reactions with N- cyanoguanidines The last type of cyclization reaction to produce imidazolones involves a rarely seen cyclization between an N-cyanoguanidine and a carboxylic acid (Scheme lI-19).51 Garratt proposes a reaction mechanism where under acidic conditions, a N-cyanoguanidine is attacked by a carboxylic acid to form the seven-membered ring intermediate (ll-24). Subsequent rearrangement of the seven-membered ring to the five-membered ring (imidazolone lI-25), followed by elimination of the amide protecting group on N-3 results in the formation of the final imidazolone product (Scheme ll-19).51 There were only a couple imidazolones made through this route where R = ethyl and benzyl and the yields were 78% and 79%, respectively. Scheme "-19. lmidazolone through cyclization using N-cyanoguanidines G N o OTf filing o HE’S: N I TFA N/ff) Bn‘N’LkN R Bnt A f0 l H N) N l H R ”-24 OTf N BgN/ C)>\‘~NH Bn’ YN n \\ (:1 2 o HN HN$O R R ”-25 78% To sum up thus far, many different types of reactions have been used to create imidazolones. Many have used unique routes to obtain quaternary (5, 5- 71 disubstitution on the imidazolone ring) and non-quaternary imidazolones. However, the entirety of the cyclization reactions that produced quaternary 2- aminoimidazolones contained the quaternary stereocenter before the cyclization. With this in mind, it was thought that the best route to obtain the desired quaternary imidazolone for the natural product (Ill-1) would be to first set the quaternary center and then cyclize to form the imidazolone. II.E Synthesis of quaternary intermediates for the synthesis of the natural product Steglich and co-workers first described a rearrangement in 1975 where an allyl ester of an N-acyl amino acid was converted into a quaternary oxazolone using a variety of dehydrating reagents (Scheme ll-20).84' 85 The proposed mechanism is shown below in Scheme "-20 and starts with a cyclodehydration of an allyl ester to afford an oxazole intermediate, which upon a Claisen-type reaction produces an oxazolone.“ 85 The oxazole intermediate had not been isolated in the original reports, but is presumed based on the reaction of alkyl esters of N-acyl amino acids.” 87 Two acyl groups were used for the rearrangement, with R = phenyl and isopropyl, while R1 varied between aryl and alkyl substituents. The substitution on the allyl group (R2 and R3) varied between hydrogen, alkyl and aryl groups with yields for the quaternary oxazolone ranging from 26-820/o.84’ 85 Like the Claisen rearrangement, it was observed that the oxazole rearrangement produced only one diastereomer in cases where two stereocenters were able to be formed.88 Steglich discusses an additional hetero- Cope rearrangement that occurred (when R2 or R3 at H) after the formation of the quaternary oxazolone to afford oxazolin-5-one products in yields ranging from 72 12-100%. Depending on the substitution of R2 and R3, the hetero-cope rearrangement sometimes needed additional heating to complete.“ 85 Scheme "-20. Rearrangement developed by Steglich r— I- H 0 R2 R 0 n RTNW/KOMRs Reagent F\/8~O’E 0 R1 WR3 _ _ oxazole intermediate [Claisen R3 R 0 o R \ / 2 O O A hetero-Cope my I R=phenyl,alkyl R‘, R2R3 R N R oxazolone 12-100% 26-82% Steglich’s oxazole rearrangement is a useful tool to create a quaternary stereocenter and has been used to produce a variety of different compounds.”97 Haufe et al. used the oxazole rearrangement to lead to 4-fluoropyridines (Scheme ll-21).97 Fluoro allyl esters of N-acyl amino acids were cyclodehydrated using triphenylphosphine and carbon tetrachloride to produce the corresponding quaternary oxazolones in yields ranging from 98-100%. Subsequently, Haufe exposed the oxazolones to decahydronaphthalene and air for 20 h at 160°C which transformed the oxazolones to the corresponding 4-fluoropyridines in yields between 21-55% (Scheme ll-21).97 73 Sct Bu. oxazole l "alltl‘tls e Vifime “a l», , ~eir|l€e “ta .‘Vrme Scheme "-21. Rearrangement leading to 4-fluoropyridines R 0 Ph F c /U\ )YO Ph3P,CCI4,TEA_ WNT o R Q CH3CN,rt,16h, 0 R1 98-100% Ph Ph F 0 R1 "r -co2 o F R R1 R decahydronaphthalene air, 160°C, 20 h l Ph F F / ——’ —" \ R1 R Ph N R Ph N/ R 21 -55% Burger et al. described a unique method for the formation of the allyl oxazole intermediate by coupling the fluorinated oxazole starting material with various alcohols (Scheme ll-22).89'91 Subsequently, the oxazole rearrangement produced a variety of a-trifiuoromethyl q-amino acids (Scheme ll-22) in yields between 40-69%.90 Interestingly, Burger also identified that the allyl oxazole intermediate could be spectroscopically characterized when R = (CH3)3Si- CH=CH-CH2OH. This is the first example of the allylic oxazole intermediate being observable spectroscopically.90 Additionally, Burger showed that benzyl alcohols and hydroxymethylheterocycles, such as 2-hydroxymethylthiophene, also are compatible in the oxazole rearrangement.”91 Furthermore, Burger along with Krantz, illustrated that propargyl alcohols could be used in place of allylic alcohols to produce q-allenic a-amino acids.” 90' 92- 93 74 Scheme "-22. Rearrangement affording a-trifluoromethyl a-amino acids CF3 R1 CF3/\)\ NiF HO/VRR NaH or KOH “Ito/J ph/ko\ Ph/ko\ oxazole F3C Ph CO H O NHCOCSHS N R, R R1 F3C R \ 40-69% The oxazole rearrangement has been utilized in the synthesis of a-benzyl v-lactam derivatives, a-benzyl 6-Iactam derivatives and d-benzylproline derivatives (Scheme "-23, "-26, "-27, "-28, respectively).96 Holladay and co- workers used phosgene to carry out the oxazole rearrangement to afford a quaternary oxazolone that is quite unstable. By treating the oxazolone formed in situ with ammonium hydroxide, a primary amide was formed. Subsequent treatment of the amide with a combination of reagents led to each of the desired heterocycles (Scheme ll-23).96 Scheme “-23. Rearrangement leading to lI-26-ll-28 Ph Ph / 0 o F CAN/KNOW 1'C'ZCO'TEA : JL NH2 —> lI-26-il-28 3 H 2. NH4OH, 70% F30 fl ' 0 0 Ph NH RHN o NH2 "-26 "-27 “-23 75 Colombo et al. applied the oxazole rearrangement to the synthesis of C- glycosyl a-amino acids, and q-D-C-mannosyl-(R)-a|anine.94' 95 It was found that the cyclodehydration/rearrangement reaction afforded a 2.6:1 mixture of two diastereomeric oxazolones (Scheme ll-24). It was proposed that the major isomer is produced through a boat-like transition state during the oxazole rearrangement possibly due to steric interactions that would accompany a chair- Iike transition state for this specific case.95 Scheme "-24. Rearrangement to synthesize C-glycosyl a-amino acids *0 )VO 0 o 0 / Ph3P, ccu, TEA _ 0 )fi/ rt, 12 h, 86% N \ / o /N 04 >’ Ph Ph The majority of the examples of the oxazole rearrangement only produced quaternary oxazolones with one stereocenter.“ 85' 91' 96' 97 However, it was reported that a mixture of diastereomers were observed in the examples that had the potential to produce them, as in the case of Burger and Krantz with the 89, 90, 92, 93 94, 95 In allenic oxazolones and Colombo with the glycosyl oxazolones. the case of the allenic oxazolones, it was described that having a substitution on the methylene of the propargyl alcohol could influence the diastereomeric ratio depending on the size of the substituent.88 Moreover, in the glycosyl oxazolone cases, the stereochemistry of the sugar moiety helps dictate the stereochemical outcome of the rearrangement.“ 95 76 We proposed that this oxazole rearrangement is a unique and efficient way to create a quaternary center and would be excellent to utilize the rearrangement in the synthesis of the natural product (III-1). Furthermore, it seemed to augment and highlight the theme of our group’s work dealing with the development of oxazolone chemistry?8 The oxazole rearrangement was attempted with various N-acyl groups as shown in Table ”-1. The dehydrating reagents varied depending on the N-acyl group used. Trifluoroacetic anhydride worked the best when R = phenyl (Table ”-1) affording the oxazolone (ll-36) in a 76% yield. POC|3 was the dehydrating reagent used when R = methyl and dimethylamino to give the quaternary oxazolones "-37 and "-38 in a 69% and 86% yield, respectively. However, when the N-acyl derivative was used, a different isomer was observed as the product (Table ”-1). The product isolated was actually the lactone (lsomer B, Table "-1) in which a hetero-Cope rearrangement occurred after the initial Claisen rearrangement, which had been seen before in Steglich’s initial study.85 The physical evidence that supports this structural determination comes from the absorptions seen in the IR for oxazolone "-37 compared to those reported for the oxazolone product (lsomer A) and lactone (lsomer B). For "-37, the signals observed in the carbonyl region are at 1779 cm'1 and 1627 cm'1, which match very closely the reported lR signals for the lactone product (1780- 1770 cm" and 1645-1610 cm'1)84' 85. The IR signals associated with the oxazolone product tend to be in the range of 1850-1810 cm'1 and 1660-1650 cm‘ 1.84' 85 It is also noteworthy to mention that when the dimethylamino urea (II-31) 77 was used in the rearrangement, the resulting oxazolone was so labile that isolation could only occur after treatment with methanol. This resulted in the isolation of the quaternary ring-opened methyl ester (Structure C, Table ”-1). Electron withdrawing acyl groups attached to the nitrogen, such as the trifiuoromethyl acetyl group and 2-ethoxy-2-oxoacetyl group (compounds "-32 and "-33, respectively) require a different dehydrating reagent altogether. TFAA, POCI3, PCI3, PC|5 and PPhg/CCI4 have all been evaluated and failed to effect the rearrangement. It was found that phosgene was the only reagent that could produce the corresponding quaternary products (ll-39 and "-40, Table ”-1) in yields of 75 and 55%, respectively. However, due to the lability of the resulting oxazolones, they would readily ring-open at the carbonyl carbon even on silica gel. As a result, treatment of the quaternary oxazolones immediately with a nucleophile was necessary to be able to isolate the corresponding ring-opened products (Structure C, Table "-1). A carbonate and pivaloyl group were also attempted in the rearrangement (entries "-34 and II-35) but failed to produce the quaternary oxazolone using any of the previously described dehydrating reagents. It was observed that when a carbonate was used as the acyl group, the reaction gave back starting material after heating for 24 h with each dehydrating reagent. Conversely, when the pivaloyl group was used as the acyl group, the starting material seemed to decompose in almost every reaction condition, presumably due to the acidic nature of the reagents. 78 Table "-1. Oxazole rearrangement with various N-acyl groups o ' R TFAA, POCI3, Y0 R | O H 0 55-86% R. N‘ R'= N-tosyl indole A B R' ll-29-II-35 |so| ted Dehydrat. 0 Compound R Strui'ture Reagent Yield Product R—( O HN OMe "-29 A TFAA 76% "-36 R- c "-30 )1, B POC|3 69% 11.37 "-31 \N)" c POCI3 86% "-38 11.32 F3C5‘t c cocr2 75% "-39 EtO 11.33 c cocr2 55% "-40 o "-34 C|3CAO/1LL NR NA NA NA "-35 NA NA NA NA As stated before, the majority of the techniques used to create quaternary imidazolones involved the synthesis of the stereocenter prior to heterocycle formation. This technique was thought to be employed in the synthesis of the natural product. Therefore, since the quaternary center has now been achieved through the oxazole rearrangement, the next step was to form the imidazolone heterocycle. The quaternary oxazolone products (ll-36 and ll-39) were subjected to different reaction conditions to open the quaternary oxazolone and remove the resulting acyl group on the nitrogen to move ahead in the synthesis of the natural 79 product. However, basic reaction conditions usually only opened the oxazolone and/or removed the tosyl protecting group, while acidic reaction conditions resulted in degradation of the starting material and/or formation of multiple by- products. Surprisingly, attempts to manipulate the urea quaternary product (II-38) provided some interesting results. When treated with reagents intended to activate the urea carbonyl and subsequently substitute with an aminegg, an unexpected result was observed (Scheme ll-25). Instead of forming the intended guanidine, the final product corresponded to the quaternary hydantoin "-41 which was isolated in good yields (71 %). Scheme "-25. Unexpected reaction producing hydantoin II-41 Bn\ 0 | N / e O \ NH I OM BOP DBU /N—<\N OMe HN 0 ' 96* 0 BnNHz, CH3CN, \ \ reflux, 16 h \ N N N +3 . “-38 +3 "-41 Ts 71% An even more unusual reaction was observed when the amine was left out of the reaction and water was added instead. it was believed that the unexpected reaction might have proceeded through an isocyanate intermediate and the water was intended to hydrolyze the intermediate and upon basic workup, afford an amine (Scheme ll-26). However, a quaternary hydantoin with an incorporation of DBU was observed ih a 55% yield (Scheme ll-26). 80 Scheme "-26. Unexpected reaction producing hydantoin Il-42 l / N O OMe o ”N o BOP, DBU V O: HN O . 2. NaOMe (excess) . R,NH R R Natural Product (Ill-1) ll.F.1 Scope of rearrangement It is hypothesized that the new rearrangement initiates after the thiourea is activated and converted into a carbodiimide, which undergoes a 5-exo—dig cyclization to form an oxazole intermediate (based on data discussed in the mechanism section). There are a variety of reagents that have been reported to transform a thiourea into a carbodiimide intermediate and subsequently allow for attack by an external or internal nucleophile.1°3'107 We screened the typical reagents chosen for this type of chemistry including HgCl2,1°4' 108' "’9 Mukaiyama’s reagent,1°7' “0 and EDCI105' "1 and found that the success of the reaction was highly dependent on the reagent choice. Mercuric chloride did provide a reasonable yield of the quaternary hydantoin (49%), while Mukaiyama’s reagent failed to produce any desired hydantoin. EDCI was the most effective at yielding the quaternary hydantoin in good yields (70%). Furthermore, the hazard of mercury waste removal and the ease of workup with EDCI solidified the reason to use EDCI as the reactant for the rest of the study. A more cost efficient carbodiimide, DCC, was also attempted in the rearrangement 84 but failed to provide any reasonable yields (<5%). Polymer supported EDCI was also used although longer reaction times were necessary. A brief solvent screen revealed that dichloromethane provided the best results (70%), while solvents such as acetonitrile, benzene, tetrahydrofuran, and dichloroethane all yielded 102 poorer results (50%, 30%, 47%, and 12%, respectively). Scheme "-29. General scheme for novel rearrangement 5 Kg: R' 80% 0 H , NaOMe Y O EDCI ”YO (excess) Y O 0 NH R HN TEA R R' The first structural aspect we investigated in the rearrangement was the different groups compatible at the R position (Scheme ll-29). The different thiourea starting materials were synthesized through standard amino acid chemistry (Scheme ll-30). The Boc protected amino acids (ll-43-lI-48) (R = u 114 methyl,112 benzyl,113 pheny, l "5 pOMe-pheny, | “6 pF-pheny , and napthyl,117 respectively) were esterified with the appropriate allylic alcohol using dicyclohexylcarbodiimide (DCC). Deprotection of the Boc group with a mixture of TFA and DCM (1:1) led to the TFA salt of the amino allylic esters (ll-56-Il-62). Upon treatment of the amine salts with ethyl isothiocyanatoformate under basic conditions the desired thioureas (ll-63-Il-69) were produced.102 85 Scheme "-30. General synthesis of thioureas ll-63-Il-69 R = Me; R' = allyl, "-63 R = Bn; R' = allyl, "-64 R = Ph; R' = allyl, "-65 R = pOMe-Ph; R' = allyl, “-66 R = pF-Ph; R' = allyl, "-67 R = Napth.; R' = allyl, "-68 n O H O ' Boc’ OH R'OH. DCC V Boc’N\(u\O’R R DMAP, DCM, rt R R = Me, "-43 R = Me; R' = allyl, "-49 R = Bn, "-44 R = Bn; R' = allyl, "-50 R = Ph, "-45 R = Ph; R' = allyl, "-51 R = pOMe-Ph. "-46 R = pOMe-Ph; R' = allyl, "-52 R = pF-Ph, "-47 R = pF-Ph; R' = allyl, "-53 R = Napth.. "-48 R = Napth.; R' = allyl, "-54 R = Ph; R' = 1,1-disub. allyl, ll-55 TFA/DCM (1 :1 ), rt 0 O as ii H R- JL 'TFA O \n, T \l/‘LO’ ‘L EtO NCS HzN 0’ R' o s R TEA, DCM, rt R R = Me; R' = allyl, "-56 R = Bn; R' = allyl, "-57 R = Ph; R' = allyl, "-58 R = pOMe-Ph; R' = allyl, "-59 R = pF-Ph; R' = allyl, "-60 R = Napth.; R' = allyl, "-61 R = Ph; R' = 1,1-disub. allyl, "-62 R = Ph; R' = 1,1-disub. allyl, "-69 The synthesis of thiourea (ll-75, when R = N-tosylindole) followed a different synthetic pathway (Scheme ll-31). 2-(1H-indol-3-yl)-2-oxoacetyl chloridem' “9 (ll-70) was treated with allyl alcohol in CH3CN to yield keto-ester (ll-71) in a 95% yield. Protection of the indolic nitrogen with p-toluene sulfonyl chloride under basic conditions afforded keto-ester (ll-72). The transformation of the ketone functional group in (ll-72) to oxime (ll-73) was completed by refluxing the starting ketone with hydroxyl amine and pyridine in dioxane. The resulting oxime was a mixture of isomers and both were reduced to amine (ll-74) using zinc and acetic acid and subsequently reacted with TFA to produce the TFA salt. Finally, treatment of the amine salt with ethyl isothiocyanatoformate led to the desired thiourea (ll-75).102 86 Scheme "-31. Synthesis of thiourea ll-75 N CH3CN, 95% N “41 H 11.70 H TsCl,DlPEA DMAP, DCM, 86% H0vN\ o O \ o < NHZOH-HZOmyr. \ O N "73 Dioxane/H2O, 93% N "-72 ‘rs ' ‘Ts 1.Zn,AcOH 2. TFA, 85% EtO H N o ‘— 2 EtO NOS 0 8%NH 0 \ O TEA, DCM, 89% \ o N, "-74 N "-75 Ts Ts The results of the rearrangement with the various R groups are illustrated in Table “-2. It is shown that an alkyl group such as a methyl provided no product, while a benzyl only performed slightly better by producing the quaternary hydantoin (ll-76) in low yields (19%). When R = aryl (ll-65-lI-68), the rearrangement occurred in good yields (Table "-2) except for when R = napthyl, which could be attributed to the steric bulk of the napthyl group. The rearrangement was also successful using a heterocycle in the R position with the N-tosyl indole thiourea (ll-75) providing the corresponding hydantoin (ll-81) in 87 good yields. This particular example is important for the synthesis of indole alkaloid (Ill-1).102 Table "-2. Rearrangement containing various R groups H H O o H VOTNTNfl/Ko/m 1.EDCI,TEA,DCM,O°C : Ifi O S R 2. NaOMe, MeOH,rt R ll-63-Il-68, "-75 \ ll-76-lI-81 entry R Yield (%) Product "-63 Me 0 NA "-64 Bn 19 "-76 "-65 Ph 7o "-77 ll-66 p-OMe-Ph 67 "-78 ll-67 p-F-Ph 57 "-79 "-68 Napth 31 "-80 "-75 N-tosyl-lndole 7o "-81 It was observed that when R = methyl and benzyl, there appeared to be the formation of byproducts. It is surmised that a potential reason for the formation of byproducts and low (if any) yield of product could be an elimination of the activated thiourea affording an intermediate that could potentially go through side reactions (Scheme Il-32). Likewise, when the rearrangement was first attempted with R = indole (no nitrogen protection), the rearrangement failed to give the desired quaternary hydantoin and produced multiple byproducts. As shown in Scheme ”-32, it is postulated that a similar elimination reaction could occur when R = indole giving rise to possible side reactions and byproducts. Protection of the indole nitrogen with an electron-withdrawing group (tosyl) appeared to hinder the side reactions and allow the desired rearrangement to 88 occur. Overall, it is believed that the reaction works well under conditions where a potential elimination reaction cannot occur and when the aromatic oxazole intermediate (discussed in the mechanism section) is stabilized by aromatic R groups. Scheme "-32. Potential byproduct pathway H s N EDC|\ H o 0 Y O/\|l EDCI 693‘ N o A 0 NH R ———» UT” 3 /\|| _. o TEA 0 NH H I | OEt TB R ‘3. R When R = methyl, benzyl _ B _ o H COZEt O n 3323 o (\O N\n/NH EDCI (\O VY O/\l B I S TEA Q (3‘9 I l \ j EDCI \ / NH '3’ N ._ H .— We then examined the different allyl groups compatible with the rearrangement. The synthesis of the thiourea with a 1, 1-disubstituted allylic moiety (ll-69) followed the synthetic route that is outlined in Scheme "-30. However, when trisubstituted allylic thioureas were synthesized using the corresponding trisubstituted allylic alcohols, a different route was adopted. It was found that the higher substituted allylic esters would decompose to the corresponding carboxylic acid when treated with TFA.120 As a result, instead of using the Boc protecting group, the Fmoc group was used for the synthesis of those thioureas (Scheme ll-33). Using Fmoc phenyl glycine121 as the starting material, the allylic esters (II-83, ll-84) were produced under the same conditions as before using DCC and DMAP. Deprotection of the Fmoc group from the amine 89 was completed using piperidine to afford the free amines (ll-85, ll-86). Upon treatment of the amines with ethyl isothiocyanatoformate, the corresponding thioureas (II-87, II-88) were formed in moderate yields.102 Scheme "-33. Synthesis of higher substituted allylic thioureas H O 0 N H R Fmoc OH ROH, DCC t Fmoc O DCM, DMAP, rt "-82 , "-83 m Piperidine, CH3CN 0°C to rt R=m,l|87 Rszl-85 R: M , "-88 R: M , "-86 The rearrangement was successful with all of the differently substituted allylic esters that were synthesized (Table "-3). When the 1, 1-disubstituted allyl group was used (entry ll-69) the rearrangement gave about the same yield of corresponding quaternary hydantoin as the allyl group (entry II-65). Entry "-87, which had a 1, 1, 2~trisubstitution pattern on the allylic group was also as successful as the other entries. However, unlike the two previous entries, the product from the rearrangement (ll-90) contained two stereocenters allowing for 90 db. il' .‘v we. u'J 1., J v"- - I4 the formation of diastereomers. It was observed by 1H NMR and 13c NMR that two unseparatable diastereomers were formed from the reaction in about a 121.3 ratio. The last entry in Table "-3 includes an allyl moiety containing a 1, 2, 2- trisubstitution pattern. It is hypothesized that the lower yield resulting from this rearrangement could be attributed to the steric bulk of this particular trisubstituted allyl group. An electron withdrawing group (ethyl carbamate) on the thiourea was only used since it was previously found that electron withdrawing groups accelerate the reaction of a thiourea and desulfurizing reagent and increase the 102, 122 reactivity of the corresponding carbodiimide toward a nucleophile. Table "-3. Rearrangement containing various allyl groups 0 H H O CY“ \/ \n/Nm/NEERle 1.EDCl,TEA,DCM,0°C HN o o s T 2. NaOMe, MeOH, rt R1 "-65. ll-69, "-87, "-88 "-77, "-89 . o H entry R1 Yield (%) Product H.,N o HN "-65 m 7o "-75 O . "-69 W 66 "-89 "-90 H CY” "-87 m 66 "-90 o HN \ "-88 q 47 "-91 "-91 "£2 Proposed reaction mechanism The proposed mechanism is illustrated below in Scheme ”-34. The first step is thought to be the transformation of the starting thiourea to a carbodiimide 91 rzerlledlate (ll-92 artermentally, 'lt' Sereral reports sr sch as EDCl, Hg 'lermediale.‘6' 1 aiacled by an e rill various 5le :ferenl heleroc rlcleoph'lle on Batey and co-“ tlaloccured aft 31 all intern; resullur'lzallon arlllolm‘ldazol 'easonab‘ll C( image” of a PEHCllVe, lhu mobselllable leactlv'lty‘ lh E the GarbOdlir 34)?” intermediate (II-92). Although this intermediate has not been isolated or observed experimentally, it is presumed to be formed based on previous studies.122 Several reports suggest that thioureas undergo a desulfurization with reagents such as EDCI, HgCl2, Mukaiyama’s reagent, and others to afford a carbodiimide 122' ‘23 Subsequently, this transient carbodiimide could be intermediate.46' attacked by an external nucleophile, such as an amine, to produce guanidines with various substitutions.1°3'1°6' 1084‘" 122' 123 Furthermore, cyclizations to form different heterocycles have also been shown to occur through an attack of a nucleophile on a carbodiimide formed from the desulfurization of a thiourea. Batey and co—workers produced various iminohydantoins through a cyclization that occured after a carbodiimide intermediate, formed using HgCl2, was attacked by an internal amide.47 Additionally, Drewry and co-workers utilized a desulfurization of a thiourea with Mukaiyama’s reagent to form 2- aminoimidazolinones.46 These examples serve as supportive evidence to reasonably conclude that the initial step in the rearrangement involves the formation of a carbodiimide intermediate. The carbodiimide is believed to be very reactive, thus giving reason to a short lifespan and consequently being unobservable spectroscopically. As a consequence of the carbodiimide’s high reactivity, the carbonyl of the ester is believed to be nucleophilic enough to attack the carbodiimide, in a similar fashion as the amide in the report by Batey,47 to cause a cyclization reaction affording an oxazole intermediate (ll-93) (Scheme l|- 34)_102 92 Scheme “-34. Proposed mechanism of EDCI-mediated rearrangement H H O O>_ O EtO N N EDCI N==N \ll/ \ll/ \l/lLO/Y —’ EtO j/lLo/Y o s R R carbodiimide intermediate (II-92) EtO PEtoH _ O 0 _ \n/N “‘Ifi/W NaOMe R\'/ (excess) oxazolone ("_94) oxazole intermediate (II-93) ll 0 H H N N OEt H EtO\n/ \ll/ OMe iffo 0 N O O R HN HN 0 "-95 R "-96 After oxazole (ll-93) undergoes a Claisen rearrangement, oxazolone intermediate (ll-94) is formed (Scheme Il-34). This intermediate was isolated and characterized with 1HNMR, 13CNMR and IR, which showed the characteristic absorption bands for the oxazolone (see experimental section for spectral information). It is proposed that from the oxazolone intermediate (ll-94), the mechanism of the novel rearrangement continues with the nucleophilic opening of the oxazolone by sodium methoxide to yield urea (ll-95). As a consequence of having excess sodium methoxide in the reaction mixture, urea (ll-95) is deprotonated and cyclizes to form hydantoin (ll-96), which is further modified by 93 sodium methoxide to yield the final quaternary hydantoin upon removal of the carbamate group (Scheme ll-34). The step in which the quaternary stereocenter was formed illustrates an oxazole rearrangement first described by Steglich and co-workers.84' 85 Steglich discusses an additional hetero-Cope rearrangement that occurs with the oxazolone products to form oxazolin-5-ones, depending on the substitution on the allyl ester.“ 85 However, it is noteworthy to mention that this additional hetero-Cope rearrangement was not observed in our rearrangement. Overall, a novel rearrangement was developed that allows access to the scaffold needed for the synthesis of the natural product. The success of the N- tosyl indole and the 1, 1-disubstituted allylic moiety in the rearrangement ensures that the quaternary hydantoin formed from the pairing of those two structural features will lead to an intermediate capable of transforming into the natural product (Ill-1). The next chapter will discuss, in depth, the marine organism from which the natural product comes from as well as the first total synthesis of the natural product and two analogs. 94 ll.G General experimental information Reactions were carried out in flame-dried glassware under nitrogen atmosphere. All reactions were magnetically stirred and monitored by TLC with 0.25 pm pre-coated silica gel plates using either UV light or iodine to visualize the compounds. Column chromatography was carried out on Silica Gel 60 (230-400 mesh) supplied by EM Science. Yields refer to chromatographically and spectroscopically pure compounds unless otherwise noted. Infrared spectra were recorded on a Nicolet IR/42 spectrometer. 1H and 13C NMR spectra were recorded on a Varian Unity Plus-500 spectrometer or a Varian lnova-300, as noted in the experimental for each compound. Chemical shifts are reported relative to the residue peaks of the solvent (CDCI3: 7.24 ppm for 1H and 77.0 ppm for 13C) (Acetone-d5: 2.04 ppm for 1H and 29.8 ppm for 13C) (DMSO-d5: 2.49 ppm for 1H and 39.5 ppm for 13C). The following abbreviations are used to denote the multiplicities: s = singlet, d = doublet, dd = doublet of doublets, t = triplet, and m = multiplet. HRMS were obtained with a Micromass Q-ToF Ultima API LC-MS/MS mass spectrometer. Elemental analysis data were obtained on a Perkin Elmer 2400 Series II CHNS/O analyzer. Purity of compounds, whose elemental analyses were above the ACS tolerated 0.4% deviation, were confirmed by 1H NMR and 13C NMR. Melting points were obtained using an Electrothermal® capillary melting point apparatus and are uncorrected. Reagents and solvents were purchased from commercial suppliers and used without further purification. Anhydrous methylene chloride and toluene were 95 dispensed from a delivery system which passes the solvents through a column packed with dry neutral alumina. ll.H Experimental procedures and characterization (ll-14).124 To a flame dried 25 mL round bottom flask was added dry dioxane (10.0 mL) and KOH (0.352 g, 6.29 mmol). Then acetyl indole (1.00 g, 6.29 mmol) and benzaldehyde (1.27 mL, 12.6 mmol) were added and the mixture refluxed for 24 h under nitrogen. The precipitate was filtered off and recrystallized from EtOH to give yellow solid crystals. Yield (1.00 g, 65.0%). 1H NMR (500 MHz), d-Acetone: 6 7.20-7.30 (m, 2H), 7.38-7.45 (m, 3H), 7.50-7.55 (m, 1H), 7.75-7.80 (m, 4H), 8.50 (m, 1H), 8.57 (s, 1H), 11.00 (bs, 1H); 13C NMR (125 MHz), d-Acetone: 8 112.6, 119.4, 122.7, 123.2, 124.1, 125.3, 127.2, 129.0, 129.6, 130.4, 133.9, 136.6, 138.0, 140.6, 184.5. ms: calculated for C17H13NO (M+) = 247.1 and found (M+) = 247.1. (ll-15). To a 50 mL round bottom flask was added "-14 (0.700 g, 2.83 mmol) and MeOH (20.0 mL). Then NaOH (1.95 mL of a 10% solution) was added to the reaction followed by H2O2 (1.95 mL, 17.0 mmol). The reaction refluxed for about 5 h under nitrogen. The reaction mixture was reduced to about 5.00 mL and then H2O (20.0 mL) was added and the solid product precipitated out of solution and was filtered off. The product was recrystallized from EtOH. Yield (0.483 g, 65.0%). 1H NMR (500 MHz), DMSO: 8 4.20 (d, J = 1.8 Hz, 1H), 4.49 (d, J = 1.9 Hz, 1H), 7.24 (m, 2H), 7.40 (m, 5H), 7.51 (m, 1H), 8.22 (dd, J = 1.2, 6.6 Hz. 1H), 8.65 (s, 1H), 12.20 (s, 1H); 13c NMR (125MHz), DMSO: 8 57.9, 59.9, 112-2, 115.2, 121.2, 122.1, 123.3, 125.3, 126.2, 128.4, 128.5, 135.1, 136.2, 96 136.4, 187.3. lR (NaCl) 3190 cm'1, 1625 cm”, 1612 cm". M.S: calculated for C17H13NO2 (M+) = 263.2 and found (M+) = 263.4. Melting Point = 232-234°C. (II-16). To a 100 mL round bottom flask was added EtOH (30.0 mL) and ll- 15 (0.250 g, 0.950 mmol). Then 10% Pd/C (1 scoop) was added and the round bottom was fitted with a hydrogen balloon and the mixture stirred at room temperature for 30 min. The mixture was filtered over celite and the filtrate concentrated. The crude material was purified by column chromatography (silica gel, 50% EtOAc; 50°/o hexane) affording the product as an oil. Yield (0.225 g, 89.0%). 1H NMR (500 MHz), acetone: 8 2.95 (dd, J = 7.9, 13.9 Hz, 1H), 3.22 (dd, J = 4.2, 13.9 Hz, 1H), 4.24 (d, J = 6.6 Hz, 1H), 5.06 (s, 1H), 7.14-7.28 (m, 7H), 7.53 (m, 1H), 8.33 (m, 1H), 8.38 (s, 1H), 11.10 (s, 1H); 13c NMR (125 MHz), Acetone: 8 43.9, 75.5, 112.8, 114.5, 122.7, 122.9, 124.1, 126.9, 127.0, 128.7, 130.4, 134.5, 137.6, 139.2, 196.6. IR (NaCl) 3389 cm", 3261 cm", 1633 cm". HRMS: [M + H]+ = 266.1181, calculated for C17H16NO2, 266.1181. (II-17). To a 50 mL round bottom flask was added "-16 (0.169 g, 0.640 mmol) and EtOAc (20.0 mL). Then IBX (0.357 g, 1.28 mmol, synthesized according to Frigerio‘zs) was added and the mixture refluxed overnight under nitrogen. The yellow mixture was filtered over celite and the filtrate concentrated. The crude material was purified by column chromatography (silica gel, 30% EtOAc; 70% hexane) affording the product as an oil. Yield (0.147 g, 88.0%). 1H NMR (500 MHz), CDCI3: 8 4.30 (s, 2H), 7.24-7.40 (m, 8H), 8.28 (d, J = 3.2 Hz, 1H), 8.43 (dd, J = 1.1, 8.6 Hz, 1H), 8.78 (s, 1H); 13c NMR (125 MHz), cocr3: 8 44.1 , 111.5, 112.7, 122.4, 123.4, 124.3, 126.1, 127.0, 128.6, 129.8, 133.2, 135.8, 97 136.4, 183.7, 199.6. lR (NaCl) 3273 cm'1, 1718 cm", 1602 cm". HRMS: [M + H]+ = 264.1027, calculated for C17H14NO2, 264.1025. (ll-18). To a flame dried 25 mL round bottom flask was added "-17 (0.0740 g, 0.280 mmol) and dry THF (5.00 mL). Then dimethylguanidine hydrogen sulfate (0.114 g, 0.420 mmol) was added followed by NaH (0.0390 mg, 0.980 mmol). The reaction stirred at room temperature for 30 min and then at reflux for 2-3 h and again at room temperature overnight all under nitrogen. A precipitate was filtered off and washed with water and extracted with EtOAc (10 x 20.0 mL). The organics were combined and dried using anhydrous sodium sulfate and concentrated to give the product as a solid. Yield (0.0490 g, 53.0%). 1H NMR (500 MHz), DMSO: 8 2.85 (s, 6H), 3.28 (d, J = 13.2 Hz, 1H), 3.46 (d, J = 12.9 Hz, 1H), 6.94 (t, J = 7.4 Hz, 1H), 7.07 (t, J = 7.3 Hz, 1H), 7.15-7.25 (m, 5H), 7.37 (d, J = 8.2 Hz, 1H), 7.41 (d, J = 7.9 Hz, 1H), 7.46 (s, 1H); 13C NMR (125 MHz), DMSO: 8 37.8, 41.9, 68.8, 111.7, 115.4, 118.8, 119.9, 121.3, 123.4, 125.3, 126.5, 127.6, 130.6, 136.0, 136.8, 169.7, 187.6. IR (KBr) 3279 cm", 1681 cm", 1615 cm'1. M.S: calculated for C20H20N4O (M+) = 332.4 and found (M+) = 332.2. Melting Point = 298-300°C decomposed. (II-19). To a flame dried 25 mL round bottom flask was added isoamyltriphenylphosphonium bromide (0.690 g, 1.67 mmol) and anhydrous THF (6.00 mL). The mixture was cooled to -78°C and then NaHMDS (1.25 mL, 1.25 mmol) was added slowly and the resulting mixture stirred at -78°C for 1 h under nitrogen. Then a solution of N-tosyl-1H-indole-3-carboxaldehyde126 (0.250 g, 0-836 mmol) in anhydrous THF (6.00 mL) was added and the mixture stirred at - 98 Tinor 3 h ar Hazel (5.00 it "moved and ' rainwater (l ) arrjdrous 50 05mm chr0l :‘iliact as ar =55 Hz. 6H 3H) 5.86 8.1 Hz. 2H), 7H). 7.58 (s liZSlile) 123.3. 124 ”“1 (weak ("-2 ”1100,30 and 030: Wemlglit Strifile SO ‘v‘u’ere W8 sodium 1 99 001/0). 78°C for 3 h and then warmed to room temperature overnight all under nitrogen. Water (5.00 mL) was then added and stirred for 30 min. The solvent was removed and the residue was extracted with DCM (2 x 20.0 mL) and washed with water (1 x 20.0 mL) and brine (1 x 20.0 mL). The organics were dried using anhydrous sodium sulfate and concentrated. The crude residue was purified by column chromatography (silica gel, 50°/o ether; 50°/o hexane) affording the product as an oil. Yield (0.264 g, 89.0%). 1H NMR (500 MHz), CDC13: 8 1.00 (d, J = 6.5 Hz, 6H), 1.80 (sep, J = 6.7 Hz, 1H), 2.26 (dt, J = 1.8 Hz, 6.8 Hz, 2H), 2.31 (s, 3H), 5.86-5.92 (dt, J = 7.0 Hz, 11.5 Hz, 1H), 6.46-6.49 (m, 1H), 7.20 (d, J = 8.1 Hz, 2H), 7.27 (t, J = 7.4 Hz, 1H), 7.36 (d, J = 8.3 Hz, 1H), 7.55 (d, J = 7.6 Hz, 1H), 7.58 (s, 1H), 7.80 (d, J = 8.5 Hz, 2H), 8.06 (d, J = 8.3 Hz, 1H); 13c NMR (125MHz), CDCl;,: 8 21.3, 22.4, 28.6, 38.6, 113.5, 117.9, 119.3, 119.4, 123.1, 123.3, 124.7, 126.6, 129.7, 130.8, 133.6, 134.5, 135.0, 144.7. IR: (NaCl) 1610 cm'1 (weak). HRMS: [M + HT = 354.1536, calculated for C21H24NO2S, 354.1528. (ll-20). To a 50 mL round bottom flask was added "-19 (0.264 g, 0.748 mmol), acetone (18.0 mL) and water (2.00 mL). Then NMO (0.131 g, 1.12 mmol) and 0504 (0.760 mL, 0.0748 mmol) were added and the mixture stirred at RT overnight under nitrogen. The mixture was quenched with a saturated potassium sulfite solution (10.0 mL) and extracted with EtOAc ( 2 x 60.0 mL). The organics were washed with a brine solution (1 x 60.0 mL) and dried using anhydrous sodium sulfate and concentrated to give the product as a solid. Yield (0.288 g, 99.0%). 1H NMR (500MHz), acetone: 8 0.75 (d, J = 6.6 Hz, 3H), 0.84 (d, J = 6.6 Hz. 3H), 1.24 (m, 1H), 1.32 (m, 1H), 1.80 (m, 1H), 2.32 (s, 3H), 3.68 (s, 1H), 3.98 99 (s, 1H), 4.32 (s, 1H), 4.84 (d, J = 4.4 Hz, 1H), 7.21 (t, J = 8.1 Hz, 1H), 7.28-7.35 (m, 3H), 7.64 (s, 1H), 7.71 (d, J = 7.8 Hz, 1H), 7.82 (d, J = 8.3 Hz, 2H), 7.97 (d, J = 8.5 Hz, 1H); 13C NMR (125MHz), acetone: 8 21.3, 21.8, 24.2, 25.1, 41.9, 72.2, 72.8, 114.3, 121.9, 123.8, 124.9, 125.2, 125.4, 127.6, 130.7, 131.0, 136.13, 136.17, 146.1. IR: (NaCl) 3425 (br) cm". HRMS: [M + H]+ = 370.1482, calculated for C21H24N038, 370.1477 (ll-20 minus H2O). Melting Point = 136-138°C. (ll-21). To a 50 mL round bottom flask was added "-20 (0.288 g, 0.744 mmol), EtOAc (20.0 mL) and IBX (0.833 g, 2.98 mmol). The mixture was refluxed for 18 h under nitrogen and then the IBX byproduct was filtered off. The filtrate was concentrated and the crude residue was purified by column chromatography (silica gel, 30% EtOAc; 70% hexane) affording the product as an oil. Yield (0.163 g, 57.0%). 1H NMR (500 MHz), CDCI3: 8 0.99 (d, J = 6.7 Hz , 6H), 2.23 (m, 1H), 2.33 (s, 3H), 2.80 (d, J = 6.5 Hz, 2H), 7.24-7.26 (m, 2H), 7.34-7.37 (m, 2H), 7.83- 7.85 (m, 2H), 7.93-7.95 (m, 1H), 8.32-8.34 (m, 1H), 8.74 (s, 1H); 13C NMR (125 MHz), 00013: 8 21.5, 22.6, 24.3, 45.9, 113.1, 115.2, 122.8, 125.0, 125.9, 127.2, 127.9, 130.2, 134.3, 134.4, 136.4, 146.0, 184.9, 201.1. IR: (KBr) 3175 cm", 1712 cm‘1, 1651 cm‘1. HRMS: [M + H]+ = 384.1273, calculated for C21H22NO4S, 384.1270. (ll-22). To a 25 mL round bottom flask was added "-21 (0.0540 g, 0.141 mmol) and MeOH (10.0 mL). Then K2CO3 (0.0490 g, 0.352 mmol) was added and the mixture stirred at RT for 1.5 h under nitrogen. The MeOH was taken off and the residue was put into solution with EtOAc (30.0 mL) and washed with 1%HCl (1 x 10.0 mL), water (1 x 10.0 mL) and brine (1 x 10.0 mL). The organics 100 were dried using anhydrous sodium sulfate and concentrated. The crude residue was purified by column chromatography (silica gel, 30% EtOAc; 70% Hexane) affording the product as a yellow solid. Yield (0.0170 g, 53.0%). 1H NMR (500 MHz), CDCl3: 8 0.97 (d, J = 6.6 Hz, 6H), 2.24 (m, 1H), 2.83 (d, J = 7.0 Hz, 2H), 7.29-7.42 (m, 3H), 8.31 (d, J = 3.2 Hz, 1H), 8.42 (m, 1H), 9.15 (bs, 1H); 13C NMR (125 MHz), CDC13: 8 22.6, 24.4, 46.3, 111.5, 112.5, 122.4, 123.3, 124.3, 126.1, 136.0, 136.1, 185.0, 203.1. IR (NaCl) 3200 cm'1, 1712 cm", 1608 cm", 1584 cm' 1. M.S: calculated for C14H15NO2 (M+) = 229.1 and found (M+) = 229.1. Melting Point = 130-132’C. (II-23). To a flame dried 10 mL round bottom flask was added "-22 (0.0750 g, 0.328 mmol) and dry THF (5.00 mL). Then dimethylguanidine hydrogen sulfate (0.134 g, 0.491 mmol) was added followed by NaH (0.0460 g, 1.15 mmol). The reaction stirred at room temperature for 30 min and then refluxed for 2-3 h and again at room temperature overnight all under nitrogen. A precipitate was filtered off and washed with water and extracted with EtOAc (10 x 20.0 mL). The filtrate was concentrated and the residue was extracted with EtOAc (3 x 20.0 mL) and washed with water (1 x 10.0 mL) and brine (1 x 10.0 mL). The organics were combined and dried using anhydrous sodium sulfate and concentrated. The crude residue was purified by column chromatography (silica gel, 90% DCM; 10% MeOH) affording the product as a solid. Yield (0.0190 g, 19.0%). 1H NMR (500 MHz), CDC|32 8 0.87 (d, J = 6.6 Hz, 3H), 0.89 (d, J = 6.6 Hz, 3H), 1.65 (m, 1H), 2.01 (dd, J = 5.6 Hz, 11.1 Hz, 1H), 2.07 (dd, J = 5.6 Hz, 11 1Hz, 1H), 3.00-3.08 (bs, 3H), 3.09-3.14 (bs, 3H), 6.91 (t, J = 7.1 Hz, 1H), 7.03 (t, 101 J = 7.1 Hz, 1H), 7.24 (d, J = 2.7 Hz, 1H), 7.32 (d, J = 8.0 Hz, 1H), 7.45 (d, J = 7.8 Hz, 1H), 8.26 (s, 1H), 10.9 (s, 1H); 13c NMR (125 MHz), c003: 8 23.7, 24.0, 24.5, 36.1, 38.3, 44.5, 67.7, 111.3, 115.7, 118.3, 119.8, 120.8, 122.5, 124.8, 136.6, 169.7, 188.5. IR: (KBr) 3304 cm", 1681 cm", 1608 cm'1. HRMS: [M + H]+ = 299.1879, calculated for C17H23N4O, 299.1872. Melting Point = 274-276°C decomposes. (ll-29). To a flame dried 50 mL round bottom flask was added Ill-8 (0.104 g, 0.261 mmol) and anhydrous DCM (20.0 mL). Then TEA (0.0700 mL, 0.522 mmol) was added and the reaction was cooled to 0°C. Then benzoyl chloride (0.0400 mL, 0.392 mmol) was added slowly and the mixture warmed up to RT while stirring overnight under nitrogen. The mixture was washed with saturated NaHC03 (1 x 10.0 mL) and brine (1 x 10.0 mL) and the organics were dried using anhydrous sodium sulfate and concentrated. The crude material was purified using column chromatography (silica gel, 7:3 hexanes/ethyl acetate) to yield a solid (0.111 g, 85.0%). 1H NMR (500 MHz), 00013: 8 1.57 (s, 3H), 2.30 (s, 3H), 4.59 (q, J = 13.1 Hz, 2H), 4.85 (d, J = 7.1 Hz, 2H), 6.08 (d, J = 7.4 Hz, 1H), 6.96 (d, J = 7.3 Hz, 1H), 7.18-7.25 (m, 3H), 7.30-7.5 (m, 4H), 7.64-7.77 (m, 6H), 7.95 (d, J = 7.7 Hz, 1H) ; 13C NMR (125 MHz), CDCI3: 8 19.3, 21.5, 49.4, 69.2, 113.7, 113.9, 117.7, 119.9, 123.6, 124.9, 125.2, 126.9, 127.1, 128.5, 128.6, 129.9, 131.9, 133.4, 134.9, 135.1, 138.9, 145.2, 166.8, 170.1. IR: (NaCI) 3374 cm'1 , 1748 cm", 1657 cm". HRMS: [M + H]"’ = 503.1642, calculated for C23H27N205S, 503.1641. Melting Point = 118-120°C. 102 (ll-30). To a flame dried 50 mL round bottom flask was added Ill-8 (0.250 g, 0.628 mmol) and anhydrous DCM (20.0 mL). Then TEA (0.200 mL, 1.57 mmol) was added and the reaction was cooled to 0°C. Then acetyl chloride (0.0900 mL, 1.26 mmol) was added slowly and the mixture warmed up to RT while stirring overnight under nitrogen. The mixture was washed with saturated NaHCOa (1 x 10.0 mL) and brine (1 x 10.0 mL) and the organics were dried using anhydrous sodium sulfate and concentrated. The crude residue was purified by column chromatography (silica gel, 80% DCM; 20% EtOAc) affording the product as a solid. Yield (0.264 g, 95.0%). 1H NMR (500 MHz), CDCI3: 8 1.58 (s, 3H), 2.02 (s, 3H), 2.34 (s, 3H), 4.55 (d, J = 12.9, 1H), 4.59 (d, J = 13.0 Hz, 1H), 4.85 (s, 2H), 5.91 (d, J = 7.6 Hz, 1H), 6.40 (d, J = 6.6 Hz, 1H), 7.20-7.36 (m, 4H), 7.61 (s, 1H), 7.63 (d, J = 8.0 Hz, 1H), 7.75 (d, J = 8.3 Hz, 2H), 7.96 (d, J = 8.3 Hz, 1H); 13C NMR (125 MHz), CDCI3: 8 19.2, 21.5, 22.9, 48.9, 69.0, 113.6, 113.7, 117.6, 119.9, 123.5, 124.8, 125.2, 126.8, 128.4, 129.9, 134.9, 135.0, 138.9, 145.1, 169.4, 170.1. lR: (NaCI) 3291 cm", 1748 cm", 1651 cm". HRMS: [M + H]" = 441.1486, calculated for C23H25N2058, 441.1484. Melting Point = 118-120°C. (ll-31). To a flame dried 25 mL round bottom flask was added Ill-8 (1.48 g, 3.73 mmol) and anhydrous DCM (50.0 mL). Then TEA (3.29 mL, 23.8 mmol) was added and the reaction was cooled to 0°C. Then dimethylcarbamoyl chloride (1.50 mL, 16.4 mmol) was added slowly and the mixture warmed to RT overnight under nitrogen. The mixture was washed with saturated NaHC03 (1 x 10.0 mL) and brine (1 x 10.0 mL) and the organics were dried using anhydrous sodium sulfate and concentrated. The crude residue was purified by column 103 chromatography (silica gel, 70% DCM; 30% EtOAc) affording the product as an oil. Yield (1.38 g, 79.0%). 1H NMR (500 MHz), CDC|3I 8 1.55 (s, 3H), 2.29 (s, 3H), 2.86 (s, 6H), 4.48 (d, J = 13.1 Hz, 1H), 4.57 (d, J = 13.0 Hz, 1H), 4.78 (d, J = 10.5 Hz, 2H), 5.30 (d, J = 7.5 Hz, 1H), 5.77 (d, J = 7.5 Hz, 1H), 7.15 (m, 4H), 7.55 (s, 1H), 7.65 (d, J = 8.2 Hz, 1H), 7.70 (d, J = 8.1 Hz, 2H), 7.91 (d, J = 8.5 Hz, 1H); 13C NMR (125 MHz), CDCI3: 8 19.1, 21.3, 36.0, 50.4, 68.6, 113.3, 113.4, 118.5, 120.0, 123.3, 124.4, 124.9, 126.7, 128.7, 129.7, 134.8, 134.9, 139.0, 144.9, 157.1, 171.1. IR: (NaCI) 3425 cm", 3334 cm", 1748 cm", 1645 cm". HRMS: [M + H]+ = 470.1750, calculated for C24H28N3058, 470.1750. (II-32). To a 250 mL flame dried round bottom flask was added Ill-8 (2.00 g, 5.03 mmol) and anhydrous DCM (0.100 L). Then anhydrous TEA (1.60 mL, 11.6 mmol) was added and the mixture was cooled to 0°C. Trifluoroacetic anhydride (1.06 mL, 7.54 mmol) was then added dropwise and the reaction mixture was warmed to room temperature while stirring under nitrogen overnight. Saturated NaHC03 was added to quench the excess TFAA. The organics were separated from the aqueous solution and then washed with brine (1 x 50.0 mL). The organics were dried using anhydrous sodium sulfate and concentrated. The crude residue was purified by column chromatography (silica gel, 80% hexanes; 20% EtOAc) affording the product as a solid. Yield (2.00 g, 80.0%). 1H NMR (500 MHz), CDCl3: 8 1.55 (s, 3H), 2.32 (s, 3H), 4.58 (d, J = 12.8 Hz, 1H), 4.64 (d, J = 12.9 Hz, 1H), 4.85 (s, 2H), 5.92 (d, J = 7.2 Hz, 1H), 7.22 ( d, J = 8.7 Hz, 2H), 7.27 (t, J = 8.1 Hz, 1H), 7.36 (t, J = 8.3 Hz, 1H), 7.60 (d, J = 7.9 Hz, 1H), 7.63 (d, J = 7.2 Hz, 1H), 7.74 (s, 1H), 7.76 (d, J = 8.5 Hz, 2H), 7.98 (d, J = 8.5 Hz, 1H); 104 13C NMR (125 MHz), CDC13: 8 19.0, 21.3, 49.2, 69.6, 112.0, 113.6, 114.0, 114.3, 115.6, 116.5, 118.8, 119.4, 123.7, 125.3, 125.5, 126.7, 127.8, 129.8, 134.5, 134.8, 138.4, 145.3, 156.0, 156.3, 156.6, 156.9, 168.5. lR: (NaCI) 3346 cm”, 1742 cm", 1718 cm". HRMS: [M + H]° = 495.1206, calculated for C23H22N205SF3, 495.1202. Melting Point = 119-121°C. (ll-33). To a flame dried 50 mL round bottom flask was added Ill-8 (0.250 g, 0.628 mmol) and anhydrous DCM (20.0 mL). Then TEA (0.170 mL, 1.26 mmol) was added and the reaction was cooled to 0°C. Then ethyl (chlorocarbonyl)formate (0.100 mL, 0.942 mmol) was added slowly and the mixture stirred at 0°C for 1.5 h under nitrogen. Water (5.00 mL) was added and the organics were separated, washed with brine (1 x 20.0 mL), dried using anhydrous sodium sulfate and concentrated. The crude residue was purified by column chromatography (silica gel, 70% hexane; 30% EtOAc) affording the product as an oil. Yield (0.172 g, 55.0%). 1H NMR (500 MHz), CDC13: 8 1.32 (t, J = 7.3 Hz, 3H), 1.54 (s, 3H), 2.29 (s, 3H), 4.29 (q, J = 7.5 Hz, 2H), 4.55 (m, 2H), 4.82 (m, 2H), 5.87 (d, J = 8.1 Hz, 1H), 7.19 (d, J = 8.2 Hz, 1H), 7.21-7.31 (m, 3H), 7.59 (m, 1H), 7.64 (s, 1H), 7.74 (m, 2H), 7.93 (m, 2H); 130 NMR (125 MHz), CDCI3: 8 13.8, 19.1, 21.4, 49.2, 63.3, 69.2, 113.6, 113.9, 116.3, 119.7, 123.5, 125.24, 125.26, 126.8, 128.0, 129.9, 134.8, 134.9, 138.6, 145.1, 155.8, 159.6, 168.8. IR: (NaCI) 3360 cm“, 1750 cm", 1615 cm". HRMS: [M + H]+ = 499.1544, calculated for C25H27N2078, 499.1539. (II-34). To a 100 mL flame dried round bottom flask was added Ill-8 (0.311 g. 0.781 mmol) and anhydrous DCM (50.0 mL). Then DIPEA (0.270 mL, 1.56 105 mmol) was added and the mixture was cooled to 0°C. Then Troc-Cl (0.160 mL, 1.17 mmol) was then added dropwise and the reaction mixture was warmed to room temperature while stirring under nitrogen overnight. The solvent was removed and the residue was resolvated in ether (50.0 mL) and washed with 1% HCI (30.0 mL). The organics were separated and washed with brine (1 x 30.0 mL). The organics were dried using anhydrous sodium sulfate and concentrated. The crude residue was purified by column chromatography (silica gel, 80% hexanes; 20% EtOAc) affording the product as a solid. Yield (0.264 g, 59.0%). 1H NMR (500 MHz), CDCI3: 8 1.57 (s, 3H), 2.34 (s, 3H), 4.57 (d, J = 12.7 Hz, 1H), 4.61 (d, J = 13.0 Hz, 1H), 4.72 (d, J = 12.2 Hz, 1H), 4.76 ( d, J = 11.9 Hz, 1H), 4.85 (s, 2H), 5.70 (d, J = 7.8 Hz, 1H), 6.11 (d, J = 7.5 Hz, 1H), 7.20-7.28 (m, 3H), 7.34 (t, J = 8.3 Hz, 1H), 7.64 (d, J = 8.1 Hz, 1H), 7.66 (s, 1H), 7.76 (d, J = 8.3 Hz, 2H), 7.98 (d, J = 8.3 Hz, 1H); 13C NMR (125 MHz), CDCI3: 8 19.1, 21.4, 50.9, 69.2, 74.6, 95.1, 113.65, 113.69, 113.8, 117.1, 119.8, 123.5, 124.9, 125.2, 126.7, 126.81, 126.88, 128.1, 129.84, 129.89, 129.94, 134.8, 135.0, 138.7, 145.1, 153.6, 169.5. lR: (NaCI) 3370 cm", 1742 cm". HRMS: [M + H]+ = 573.0430, calculated for C24H24N2058Cl3, 573.0421. Melting Point = 84-87°C. (ll-35). To a flame dried 50 mL round bottom flask was added Ill-8 (0.250 g, 0.628 mmol) and anhydrous DCM (20.0 mL). Then TEA (0.200 mL, 1.57 mmol) was added and the reaction was cooled to 0°C. Then pivaloyl chloride (0.150 mL, 1.26 mmol) was added slowly and the mixture stirred at 0°C for 1.5 h under nitrogen. The mixture was washed with saturated NaHC03 (1 x 10.0 mL) and brine (1 x 10.0 mL) and the organics were dried using anhydrous sodium 106 sulfate and concentrated. The crude residue was purified by column chromatography (silica gel, 70% hexane; 30% EtOAc) affording the product as a solid. Yield (0.244 g, 80.0%). 1H NMR (500MHz), CD013: 8 1.20 (s, 9H), 1.57 (s, 3H), 2.32 (s, 3H), 4.53 (d, J = 13.0 Hz, 1H), 4.62 (d, J = 13.0 Hz, 1H), 4.84 (m, 2H), 5.87 (d, J = 7.1 Hz, 1H), 6.55 (d, J = 7.2 Hz, 1H), 7.20-7.34 (m, 4H), 7.6- 7.62 (m, 2H), 7.75 (d, J = 8.5 Hz, 2H), 7.96 (d, J = 8.5 Hz, 1H); 13C NMR (125MHz), CDC13: 8 19.1, 21.4, 26.9, 27.2, 38.5, 49.0, 68.9, 113.6, 117.8, 119.7, 123.4, 124.6, 125.1, 126.7, 128.4, 129.8, 134.8, 135.0, 138.8, 145.0, 170.0, 177.8. IR: (NaCI) 3352 cm" , 1748 cm", 1663 cm". HRMS: [M + H]° = 483.1961, calculated for C26H31N2058, 483.1954. Melting Point = 114-116°C. (ll-36). To a 100 mL round bottom flask was added "-29 (0.492 g, 0.980 mmol), dichloroethane (60.0 mL), and trifluoroacetic anhydride (1.37 mL, 9.80 mmol). The yellowish solution stirred at room temperature under nitrogen for 24 h and then was brought to reflux for 6 h and then stirred overnight at room temperature. The organics were washed with sodium bicarbonate (2 x 20.0 mL) and brine (2 x 10.0 mL), combined, dried using anhydrous sodium sulfate, and concentrated. The crude material was purified using column chromatography (silica gel, 8:2 hexanes/ethyl acetate) to yield a yellowish oil (0.360 g, 76.0%). 1H NMR (500MHz), CDCI3: 8 1.74 (s, 3H), 2.31 (s, 3H), 2.93 (dd, J = 5.8, 13.4 Hz, 2H), 4.80 (d, J = 17.8 Hz, 2H), 7.20-7.32 (m, 5H), 7.49-7.59 (m, 3H), 7.72 (s, 1H), 7.75 (d, J = 8.3 Hz, 2H), 7.94 (d, J = 9 Hz, 1H), 8.04-8.06 (m, 2H) ; 13C NMR (125MHz), CDCI3: 8 21.5, 24.3, 46.1, 72.8, 113.6, 116.6, 119.3, 122.0, 123.3, 123.5, 124.9, 125.7, 126.9, 127.8, 128.1, 128.8, 129.9, 132.9, 135.1, 135.4, 107 139.2, 145.1 935.1542, 02 (ll-37 g. 0.227 ml [.794 mmol solvent was :3 slasher :8 organic 3336 resid EtOAc 139.2, 145.1, 160.7, 177.6. lR: (NaCI) 1821 cm", 1657 cm". HRMS: [M + H]° = 485.1542, calculated for C28H25N2O4S, 485.1535. (II-37). To a flame dried 25 mL round bottom flask was added "-30 (0.100 g, 0.227 mmol) and anhydrous benzene (12.0 mL). Then POC13 (0.0600 mL, 0.704 mmol) was added and the mixture refluxed overnight under nitrogen. The solvent was removed and the residue was put into solution with EtOAc (20.0 mL) and washed with saturated NaHCO3 (1 x 10.0 mL) and brine (1 x 10.0 mL) and the organics were dried using anhydrous sodium sulfate and concentrated. The crude residue was purified by column chromatography (silica gel, 80% Hexane; 20% EtOAc) affording the product as a solid. Yield (0.0690 mg, 72.0%). 1H NMR (300MHz), CDCI3: 8 1.72 (s, 3H), 1.79 (s, 3H), 2.39 (s, 3H), 2.73 (dd, J = 14.1 Hz, 22.5 Hz, 2H), 4.82 (s, 1H), 4.97 (s, 1H), 7.28-7.46 (m, 4H), 7.91 (d, J = 8.7 Hz, 2H), 8.06-8.09 (m, 1H), 8.37-8.40 (m, 1H), 8.92 (s, 1H); 13C NMR (125MHz), CDCI3: 8 21.5, 24.0, 24.7, 46.8, 106.6, 111.1, 113.3, 117.4, 122.5, 124.4, 125.7, 127.1, 127.7, 130.0, 131.5, 134.6, 134.7, 138.5, 145.6, 151.5, 164.7. IR: (NaCI) 1779 cm", 1627 cm". HRMS: [M + H]+ = 423.1379, calculated for C23H23N2O4S, 423.1379. Melting Point = 121-123°C. (ll-38). To a flame dried 50 mL round bottom flask was added "-31 (0.150 g, 0.320 mmol) and anhydrous benzene (20.0 mL). Then POCI3 (0.0900 mL, 0.991 mmol) was added and the mixture was refluxed overnight under nitrogen. The solvent was then taken off and the residue was put into solution with EtOAc (20.0 mL) and washed with saturated NaHC03 (1 x 10.0 mL) and brine (1 x 10.0 mL) and the organics were dried using anhydrous sodium sulfate and 108 {133080173161 159011 (5.01 Cree gas E 335 purifiel afardng tr ,5. 3H), 3.7 m» “blame concentrated. The residue was then resuspended in benzene (20.0 mL) and MeOH (5.00 mL) and TMSCHN2 (0.480 mL, 0.960 mmol) was added slowly. Once gas evolution ceased, the solvents were taken off and the crude residue was purified by column chromatography (silica gel, 90% DCM; 10% EtOAc) affording the product as an oil. Yield (0.134 g, 86.0%). 1H NMR (500 MHz), CDC13: 8 1.63 (s, 3H), 2.27 (s, 3H), 2.84 (s, 6H), 3.10 (d, J = 13.5 Hz, 1H), 3.61 (s, 3H), 3.76 (d, J = 13.0 Hz, 1H), 4.71 (s, 1H), 4.90 (s, 1H), 5.96 (s, 1H), 7.12- 7.22 (m, 4H), 7.49 (d, J = 9.1 Hz, 1H), 7.70-7.72 (m, 3H), 7.85 (d, J = 9.5 Hz, 1H); 130 NMR (125 MHz), coca: 8 21.4, 23.4, 36.0, 41.3, 52.9, 61.3, 113.6, 115.8, 120.1, 122.1, 123.1, 124.2, 125.1, 126.8, 128.4, 129.7, 135.1, 140.7, 144.6, 155.8, 173.4. lR: (NaCI) 3443 cm", 1736 cm", 1663 cm". HRMS: [M + H]° = 484.1915, calculated for C25H30N3058, 484.1906. (ll-39). To a flame dried 25 mL round bottom flask was added "-32 (0.100 g, 0.202 mmol) and anhydrous acetonitrile (10.0 mL). Then anhydrous TEA (0.140 mL, 1.03 mmol) was added and the mixture was cooled down to 0°C. Then phosgene (0.240 mL, 0.465 mmol) was added and the reaction turned yellow and stirred at 0°C for 2 h. The reaction was quenched by being poured into an ice water mix (10.0 mL). EtOAc was added (3 x 20.0 mL) to extract the organics, which were dried and concentrated. The crude residue was resolvated into a basic methanolic solution and stirred for 20 min. The reaction was neutralized with a 1% HCI solution and the solvent was removed. The crude residue was purified was purified by column chromatography (silica gel, 90% hexanes; 10% EtOAc) affording the product as a solid. Yield (0.0530 mg, 55.0%). 109 '11 111R (5001 3.3 is. 3H). 1 arrow Hm1H)‘ EHJBQJ 1299. 134.8. WOMM C;:H;.-.Nzoss m4m "0,3860 mg, 7611,0120 iC.Then pl reiow and t in"0 an ice \ 03816913) (Rainy Rwflnd residue Wa reranes; 41 :H NMR (51 RU83H ”213, J = 3J=95 1H NMR (500MHz), CDC13: 8 1.71 (s, 3H), 2.30 (s, 3H), 3.20 (d, J = 13.4 Hz, 1H), 3.68 (s, 3H), 3.78 (d, J = 13.6 Hz, 1H), 4.81 (s, 1H), 4.99 (s, 1H), 7.14-7.27 (m, 4H), 7.32 (d, J = 8.0 Hz, 1H), 7.71 (d, J = 8.3 Hz, 2H), 7.79 (s, 1H), 7.87 (d, J = 9.3 Hz, 1H), 7.91 (s, 1H); 13C NMR (125MHz), CDCI3: 8 21.4, 22.8, 40.2, 53.7, 61.3, 113.9, 114.1, 116.3, 117.3, 118.9, 119.2, 123.6, 124.9, 126.0, 126.8, 127.4, 129.9, 134.8, 135.1, 138.6, 145.1, 154.8, 155.1, 172.0. IR: (NaCI) 3389 cm", 1730 (with shoulder) cm". HRMS: [M + H]+ = 509.1358, calculated for C24H24N2058F3, 509.1358. Melting Point = 166-168°C. (ll-40). To a flame dried 25 mL round bottom flask was added "-33 (0.0860 mg, 0.173 mmol) and anhydrous acetonitrile (10.0 mL). Then anhydrous TEA (0.120 mL, 0.882 mmol) was added and the mixture was cooled down to 0°C. Then phosgene (0.210 mL, 0.397 mmol) was added and the reaction turned yellow and stirred at 0°C for 2 h. The reaction was quenched by being poured into an ice water mix (10.0 mL). EtOAc was added (3 x 20.0 mL) to extract the organics, which were dried and concentrated. The crude residue was resolvated into a basic methanolic solution and stirred for 20 min. The reaction was neutralized with a 1% HCI solution and the solvent was removed. The crude residue was purified was purified by column chromatography (silica gel, 60% hexanes; 40% EtOAc) affording the product as a solid. Yield (0.0470 mg, 55.0%). 1H NMR (500MHz), CDCI3: 8 1.68 (s, 3H), 2.30 (s, 3H), 3.18 (d, J = 13.4 Hz, 1H), 3.67 (s, 3H), 3.79 (d, J = 11.9 Hz, 1H), 3.80 (s, 3H), 4.78 (s, 1H), 4.93 (s, 1H), 7.12 (d, J = 8.0 Hz, 1H), 7.21 (d, J = 9.5 Hz, 2H), 7.39 (d, J = 8.1 Hz, 1H), 7.72 (d. J = 9.5 Hz, 2H), 7.77 (s, 1H), 7.84 (d, J = 8.3 Hz, 1H), 8.61 (s, 1H); 13c NMR 110 12531112)" 034.124 .5 171 4&15459 0L4 133409' 0.118 mm‘ brsrmn. were it removed 2 ramr(10 sodium 8 ea'o'rlatol ail. Yield 3H12190 1H)117E 3H), 7.95 412.63. 1285,12 3258 cm CzeHzeN: (ll {005005 (125MHz), CDCI3: 8 21.5, 23.0, 40.2, 53.4, 53.5, 61.3, 113.7, 116.8, 119.6, 119.7, 123.4, 124.7, 125.6, 126.8, 127.7, 129.9, 134.9, 135.0, 139.2, 145.0, 154.3. 160.5, 171.9. lR: (NaCI) 3388 cm", 1742 cm", 1712 cm". HRMS: [M + H]° = 499.1545, calculated for C25H27N2078, 499.1539. Melting Point = 180-182°C. (ll-41). To a flame dried 25 mL round bottom flask was added "-38 (0.0440 9, 0.0910 mmol) and anhydrous CH3CN (10.0 mL). Then BOP (0.0520 g, 0.118 mmol) and DBU (0.200 mL, 1.37 mmol) were added and the mixture stirred for 5 min. Then benzyl amine (0.0100 mL, 0.137 mmol) was added and the mixture refluxed overnight under a nitrogen atmosphere. The solvent was removed and the residue was resolvated with EtOAc (20.0 mL) and washed with water (10.0 mL) and brine (10.0 mL). The organics were dried using anhydrous sodium sulfate and concentrated. The crude residue was purified by column chromatography (silica gel, 4% MeOH; 96% DCM) affording the product as an oil. Yield (0.0330 g, 71.0%). 1H NMR (500MHz), CDC13: 8 1.41 (s, 3H), 2.31 (s, 3H), 2.90 (m, 2H), 4.61 (d, J = 14.5 Hz, 1H), 4.67 (d, J = 14.5 Hz, 1H), 4.70 (s, 1H), 4.76 (s, 1H), 6.75 (s, 1H), 7.16-7.38 (m, 9H), 7.63 (s, 1H), 7.71-7.78 (m, 3H), 7.95 (d, J = 9.0 Hz, 1H); 13c NMR (125MHz), coch: 8 21.5, 23.7, 42.5, 44.2, 63.8, 113.7, 117.3, 120.2, 121.5, 123.5, 123.9, 125.1, 126.8, 127.4, 127.8, 128.5, 128.6, 129.9, 134.9, 135.5, 135.6, 138.3, 145.2, 156.6, 173.1. IR: (NaCI) 3258 cm", 1774 cm“, 1715 cm". HRMS: [M + Hr = 514.1807, calculated for C29H28N304S, 514.1801. (ll-42). To a flame dried 25 mL round bottom flask was added "-38 (0.0500 g, 0.104 mmol) and anhydrous CH3CN (10.0 mL). Then BOP (0.0590 g, 111 3.135 mmo' stredforS 'efluxed 0vr re'esidue mL) and b s.iiate an verrematogl 01. Yield (C arr 6H), 1. 3J:13; 1H), 4.92 hiz, 2H), 7 kai5. 1140‘ 11 1359. 13: Cm“. 162 577.2485 (Il- 9-529 m mW) ar brOught c was am)“ The Whit 0.135 mmol) and DBU (0.0230 mL, 0.156 mmol) were added and the mixture stirred for 5 min. Then water (0.400 mL, 22.2 mmol) was added and the mixture refluxed overnight under a nitrogen atmosphere. The solvent was removed and the residue was resolvated with EtOAc (20.0 mL) and washed with water (10.0 mL) and brine (10.0 mL). The organics were dried using anhydrous sodium sulfate and concentrated. The crude residue was purified by column chromatography (silica gel, 4% MeOH; 96% DCM) affording the product as an oil. Yield (0.0330 g, 55.0%). 1H NMR (500MHz), CDCI3: 8 1.58 (s, 3H), 1.59-1.73 (m, 6H), 1.84 (m, 2H), 2.35 (s, 3H), 2.50 (m, 2H), 2.93 (d, J = 13.7 Hz, 1H), 2.99 (d, J = 13.7 Hz, 1H), 3.28 (m, 2H), 3.41 (m, 2H), 3.53 (t, J = 7.1 Hz, 2H), 4.82 (s, 1H), 4.92 (s, 1H), 6.96 (s, 1H), 7.22-7.36 (m, 4H), 7.67 (s, 1H), 7.78 (d, J = 8.5 Hz, 2H), 7.87 (d, J = 7.8 Hz, 1H), 7.97 (d, J = 8.3 Hz, 1H); 13C NMR (125MHz), CDC13: 8 21.8, 23.6, 24.3, 26.9, 28.8, 30.1, 37.0, 37.4, 44.4, 45.8, 49.7, 64.0, 114.0, 117.4, 120.5, 121.8, 123.8, 124.2, 125.3, 127.1, 127.7, 130.2, 135.1, 135.9, 138.9, 145.5, 157.0, 173.6, 176.1. lR: (NaCI) 3243 cm", 1779 cm", 1718 cm", 1621 cm". HRMS: [M + H]+ = 577.2490, calculated for C31H37N4058, 577.2485. (II-49). To a flame dried 100 mL round bottom flask was added "-43 (1.00 g, 5.29 mmol) and anhydrous DCM (50.0 mL). Then allyl alcohol (0.720 mL, 10.6 mmol) and DMAP (0.0650 g, 0.529 mmol) were added and the mixture was brought down to 0°C. DCC (1.63 g, 7.94 mmol) was then added and the mixture was allowed to warm to room temperature overnight while stirring under nitrogen. The white precipitate that formed was filtered off and the DCM filtrate was 112 washed with brine (1 x 20.0 mL), dried using anhydrous sodium sulfate and concentrated. The crude residue was purified by column chromatography (silica gel, 20% EtOAc; 80% Hexane) affording the product as an oil. Yield (1.09 g, 90.0%). 1H NMR (500 MHz), CDCI3: 8 1.33 (d, J = 7.1 Hz, 3H), 1.37 (s, 9H), 4.26 (bs, 1H), 4.56 (m, 2H), 5.08 (bs, 1H), 5.17-5.27 (m, 2H), 5.81-5.87 (m, 1H); 13c NMR (125 MHz), CDCI3: 8 18.4, 28.2, 49.1, 65.6, 79.6, 118.4, 131.6, 155.0, 172.9. IR: (NaCI) 3380 cm", 1750 cm“, 1710 cm". HRMS: [M + H1" = 230.1401, calculated for C11H20NO4, 230.1392. Anal. Calcd. For C11H19NO4: C, 57.62; H, 8.35; N, 6.11. Found: C, 58.89; H, 8.22; N, 5.90. (ll-50). To a flame dried 100 mL round bottom flask was added "-44 (1.38 g, 5.23 mmol) and anhydrous DCM (50.0 mL). Then allyl alcohol (0.710 mL, 10.5 mmol) and DMAP (0.0640 g, 0.523 mmol) were added and the mixture was brought down to 0°C. DCC (1.62 g, 7.85 mmol) was then added and the mixture was allowed to warm to room temperature overnight while stirring under nitrogen. The white precipitate that formed was filtered off and the DCM filtrate was washed with brine (1 x 20.0 mL), dried using anhydrous sodium sulfate and concentrated. The crude residue was purified by column chromatography (silica gel, 20% EtOAc; 80% Hexane) affording the product as a whitish solid. Yield (1.34 g, 84.0%). 1H NMR (500MHz), CDCI3: 8 1.39 (s, 9H), 3.02-3.14 (m, 2H), 4.58 (d, J = 6.6 Hz, 2H), 4.96 (bs, 1H), 5.20-5.30 (m, 2H), 5.80-5.88 (m, 1H), 7.12 (m, 2H), 7.20-7.29 (m, 3H); 13c NMR (125MHz), cocra: 8 28.2, 38.3, 54.4, 65.8, 79.8, 118.8, 126.9, 128.4, 129.3, 131.5, 135.9, 155.0, 171.5. IR: (NaCI) 3380 cm“ 1 , 1750 cm", 1710 cm". HRMS: [M + H]* = 306.1703, calculated for C17H24NO4, 113 336.1705. 7 53.29: H, 7. (ll-5‘ 5.97 mm 3301) and 33.ng do was allowe "re white trashed w :ercentrat Get, 20% 11.49 g, 8 5.12-5.17 7.26-7.36 127, 123: 710 cm" Anal. Calc “1.4.78, 1). (Il- 170992 9, mL, 7-06 was brou ”Entire W 306.1705. Anal. Calcd. For C17H23NO4: C, 66.86; H, 7.59; N, 4.59. Found: C, 68.29; H, 7.41; N, 4.62. Melting Point = 62-64°C. (ll-51). To a flame dried 100 mL round bottom flask was added "-45 (1.50 g, 5.97 mmol) and anhydrous DCM (50.0 mL). Then allyl alcohol (0.820 mL, 12.0 mmol) and DMAP (0.0730 g, 0.597 mmol) were added and the mixture was brought down to 0°C. DCC (1.85 g, 8.96 mmol) was then added and the mixture was allowed to warm to room temperature overnight while stirring under nitrogen. The white precipitate that formed was filtered off and the DCM filtrate was washed with brine (1 x 20.0 mL), dried using anhydrous sodium sulfate and concentrated. The crude residue was purified by column chromatography (silica gel, 20% EtOAc; 80% Hexane) affording the product as a whitish solid. Yield (1.49 g, 86.0%). 1H NMR (500MHz), CDCI3: 8 1.41 (s, 9H), 4.58-4.60 (m, 2H), 5.12-5.17 (m, 2H), 5.32 (d, J = 7.3 Hz, 1H), 5.56 (bs, 1H), 5.75-5.83 (m, 1H), 7.26-7.36 (m, 5H); 13c NMR (125MHz), CDC13: 8 28.2, 57.6, 65.9, 80.0, 118.3, 127, 128.3, 128.7, 131.3, 136.8, 154.7, 170.7. lR: (NaCI) 3390 cm“ , 1750 cm", 1710 cm". HRMS: [M + H]°° = 292.1559, calculated for C16H22NO4, 292.1549. Anal. Calcd. For C16H21NO4: C, 65.96; H, 7.27; N, 4.81. Found: C, 66.78; H, 7.14; N, 4.78. Melting Point = 40-42°C. (II-52). To a flame dried 100 mL round bottom flask was added "-46 (0.992 g, 3.53 mmol) and anhydrous DCM (50.0 mL). Then allyl alcohol (0.480 mL, 7.06 mmol) and DMAP (0.0430 g, 0.353 mmol) were added and the mixture was brought down to 0°C. DCC (1.09 g, 5.30 mmol) was then added and the mixture was allowed to warm to room temperature overnight while stirring under 114 733383. The res washed :errerltratec 331.30% Et $07g.94l .1: 5.3 Hz, 8.7112, 2H 567, 65.4. 180‘) 339 RTCRHEJ FOUHd; C, (“-1 {3.625 g, 11‘4.65 was bfoL “Emits l ”13880 was alga. when. '36], 20 37.0% 1 1013 21:4 nitrogen. The white precipitate that formed was filtered off and the DCM filtrate was washed with brine (1 x 20.0 mL), dried using anhydrous sodium sulfate and concentrated. The crude residue was purified by column chromatography (silica gel, 30% EtOAc; 70% hexane) affording the product as a whitish solid. Yield (1.07 g, 94.0%). 1H NMR (500MHz), CDC13: 8 1.32 (s, 9H), 3.63 (s, 3H), 4.48 (d, J = 5.3 Hz, 2H), 5.14 (m, 2H), 5.19 (d, J = 7.5 Hz, 1H), 5.69 (m, 2H), 6.74 (d, J = 8.7 Hz, 2H), 7.18 (d, J = 8.4 Hz, 2H); 13c NMR (125MHz), CDC13: 8 27.9, 54.7, 56.7, 65.4, 79.4, 113.8, 117.8, 128.0, 128.5, 131.1, 154.5, 159.2, 170.6. IR: (NaCI) 3395 cm", 1743 cm", 1711 cm". HRMS: [M + H]'° = 322.1653, calculated for C17H24N05, 322.1654. Anal. Calcd. For C17H23N05: C, 63.54; H, 7.21; N, 4.36. Found: C, 63.69; H, 7.14; N, 4.61. Melting Point = 60-62'C. (ll-53). To a flame dried 100 mL round bottom flask was added "-47 (0.625 g, 2.32 mmol) and anhydrous DCM (50.0 mL). Then allyl alcohol (0.320 mL, 4.65 mmol) and DMAP (0.0280 g, 0.232 mmol) were added and the mixture was brought down to 0°C. DCC (0.717 g, 3.48 mmol) was then added and the mixture was allowed to warm to room temperature overnight while stirring under nitrogen. The white precipitate that formed was filtered off and the DCM filtrate was washed with brine (1 x 20.0 mL), dried using anhydrous sodium sulfate and concentrated. The crude residue was purified by column chromatography (silica gel, 20% EtOAc; 80% hexane) affording the product as an oil. Yield (0.626 g, 87.0%). 1H NMR (500MHz), CDCI3: 8 1.40 (s, 9H), 4.59 (d, J = 5.5 Hz, 2H), 5.15 (m, 2H), 5.29 (d, J = 6.9 Hz, 1H), 5.57 (s, 1H), 5.79 (m, 1H), 6.09-7.02 (m, 2H), 7.31-7.33 (m, 2H); 13c NMR (125MHz), coca: 8 28.2, 56.9, 66.1, 80.2, 115.7 (d, 115 J = 11.7 Hz), 118.6,,128.83 (d, J = 8.3 Hz), 131.1, 132.8, 154.7, 162.6 (d, J = 247.5 Hz), 170.5. IR: (NaCI) 3383 cm'1 , 1749 cm", 1711 cm". HRMS: [M + H]* = 310.1463, calculated for C15H21FNO4, 310.1455. Anal. Calcd. For C15H20FNO4: C, 62.12; H, 6.52; N, 4.53. Found: C, 61.93; H, 6.29; N, 4.49. (ll-54). To a flame dried 100 mL round bottom flask was added "-48 (1.30 g, 4.32 mmol) and anhydrous DCM (50.0 mL). Then allyl alcohol (0.590 mL, 8.64 mmol) and DMAP (0.0530 g, 0.432 mmol) were added and the mixture was brought down to 0°C. DCC (1.33 g, 6.48 mmol) was then added and the mixture was allowed to warm to room temperature overnight while stirring under nitrogen. The white precipitate that formed was filtered off and the DCM filtrate was washed with brine (1 x 20.0 mL), dried using anhydrous sodium sulfate and concentrated. The crude residue was purified by column chromatography (silica gel, 20% EtOAc; 80% hexane) affording the product as an oil. Yield (1.13 g, 77.0%). 1H NMR (500MHz), CDC13: 8 1.42 (s, 9H), 4.61-4.64 (m, 2H), 5.08—5.15 (m, 2H), 5.49 (d, J = 7.5 Hz, 1H), 5.77 (m, 1H), 6.09 (d, J = 7.8 Hz, 1H), 7.40- 7.58 (m, 4H), 7.82 (m, 1H), 7.86 (d, J = 8.7 Hz, 1H), 8.17 (d, J = 8.4 Hz, 1H); 13C NMR (125MHz), CDC|32 8 28.2, 54.7, 66.1, 80.2, 118.4, 123.3, 125.2, 125.4, 126.0, 126.9, 128.8, 129.3, 131.0, 131.3, 132.8, 134.0, 155.0, 171.5. IR: (NaCI) 3389 cm'1 , 1749 cm", 1711 cm". HRMS: [M + Hr = 342.1706, calculated for C20H24NO4, 342.1705. Anal. Calcd. For C20H23NO4: C, 70.36; H, 6.79; N, 4.10. Found: C, 67.48; H, 6.43; N, 3.85. (ll-55). To a flame dried 100 mL round bottom flask was added "-45 (1.50 g, 5.97 mmol) and anhydrous DCM (50.0 mL). Then 2-methyI-2-propen-1-ol (1 .00 116 mL, 12.0 mmol) and DMAP (0.0730 g, 0.597 mmol) were added and the mixture was brought down to 0°C. DCC (1.85 g, 8.96 mmol) was then added and the mixture was allowed to warm to room temperature overnight while stirring under nitrogen. The white precipitate that formed was filtered off and the DCM filtrate was washed with brine (1 x 20.0 mL), dried using anhydrous sodium sulfate and concentrated. The crude residue was purified by column chromatography (silica gel, 20% EtOAc; 80% hexane) affording the product as a whitish solid. Yield (1.39 g, 76.0%). %). 1H NMR (500MHz), CDCI3: 8 1.41 (s, 9H), 1.58 (s, 3H), 4.46-4.58 (m, 2H), 4.78 (s, 1H), 4.81 (s, 1H), 5.33 (d, J = 7.0 Hz, 1H), 5.56 (s, 1H), 7.27-7.37 (m, 5H); 13c NMR (125MHz), cocrgz 8 19.1, 28.2, 57.7, 68.6, 80.0, 113.2, 127.1, 128.3, 128.8, 136.9, 139.1, 154.7, 170.8. lR: (NaCI) 3389 cm‘ 1 , 1743 cm", 1711 cm". HRMS: [M + H]" = 306.1705, calculated for C17H24NO4, 306.1705. Anal. Calcd. For C17H23NO4: C, 66.86; H, 7.59; N, 4.59. Found: C, 66.95; H, 7.66; N, 4.64. Melting Point = 38-40°C. (II-56). To a 100 mL round bottom flask was added "-49 (1.32 g, 4.32 mmol), DCM (2.50 mL) and TFA (2.50 mL). The resulting solution stirred for 30 min. The excess TFA and DCM were removed and CHC|3 (3 x 15.0 mL) was added and subsequently taken off to remove any residual TFA. The crude material (oil) was taken on without further purification. Yield (1.00 g, 88.0%). 1H NMR (500MHz), CDCI3: 8 1.56 (d, J = 7.3 Hz, 3H), 4.10 (q, J = 7.1 Hz, 1H), 4.63 (m, 2H), 5.25-5.32 (m, 2H), 5.8-5.9 (m, 1H), 8.24 (s, 3H); 13C NMR (125MHz), CDCI3: 6 15.6, 49.2, 67.1, 115.8 (0, J = 290.9 Hz), 119.7, 130.5, 161.8 (q, J = 117 359112111 #390872 (ll-5' moi), DC Ti'r, The I 33:93 am areeipitatt lit-'educt E 3.34-3.1E :m“_ (.4 Calcd. 1.31, r 36.9 Hz), 169.9. IR: (KBr) 3100 (br) cm°1, 1748cm", 1675 cm". HRMS: [M + Hr = 130.0872, calculated for C6H11NO2, 130.0868. (ll-57). To a 100 mL round bottom flask was added “-50 (1.32 g, 4.32 mmol), DCM (2.50 mL) and TFA (2.50 mL). The resulting solution stirred for 30 min. The excess TFA and DCM were removed and CHCI3 (3 x 15.0 mL) was added and subsequently taken off to remove any residual TFA. The product was precipitated out of the crude residue using ether/petroleum ether to afford the product as a white solid. Yield (1.27 g, 92.0%). 1H NMR (500MHz), DMSO: 8 3.04-3.19 (m, 2H), 4.32 (t, J = 6.1 Hz, 1H), 4.58 (m, 2H), 5.18-5.26 (m, 2H), 5.73- 5.81 (m, 1H), 7.20-7.36 (m, 5H), 8.64 (bs, 3H); 13c NMR (125MHz), DMSO: 8 36.1, 53.2, 65.8, 117.1 (q, J = 299.2 Hz), 118.6, 127.2, 128.5, 129.3, 131.4, 134.6, 158.4 (q, J = 31.3 Hz), 168.7. IR: (KBr) 3100 (br) cm'1, 1745 cm", 1660 cm". HRMS: [M + H]’° = 206.1188, calculated for C12H15NO2, 206.1181. Anal. Calcd. For C14H16F3NO4: C, 52.67; H, 5.05; N, 4.39. Found: C, 52.69; H, 4.91; N, 4.31. Melting Point = 88-90°C. (ll-58). To a 100 mL round bottom flask was added "-51 (1.44 g, 4.95 mmol), DCM (2.50 mL) and TFA (2.50 mL). The resulting solution stirred for 30 min. The excess TFA and DCM were removed and CHCl3 (3 x 15.0 mL) was added and subsequently taken off to remove any residual TFA. The product was precipitated out of the crude residue using ether/petroleum ether to afford the product as a white solid. Yield (1.48 g, 98.0%).1H NMR (500MHz), DMSO: 8 4.66 (m, 2H), 5.12-5.16 (m, 2H), 5.33 (s, 1H), 5.80 (m, 1H), 7.44-7.50 (m, 5H), 9.07 (bs, 3H); 13c NMR (125MHz), DMSO: 8 55.4, 66.0, 117.1 (q, J = 299.7 Hz), 118 ((3.1, 128.1 31511310fr 0.1131102, 91.30205 (ll-SS mmol). DC 31:3. The t aaeed anr precipitate era-duet a 3.75 (8,? 1": 8.9 DMSO: 131.5, ‘ 1650 c Anal. 1 4.64; I 118.1, 128.1, 129.0, 129.5, 131.5, 132.6, 158.3 (q, J = 31.3 Hz), 168.1. IR: (KBr) 3150 (br) cm", 1745 cm", 1675 cm°1. HRMS: [M + H]° = 192.1027, calculated for C11H14NO2, 192.1025. Anal. Calcd. For C13H14F3NO4: C, 51.15; H, 4.62; N, 4.59. Found: C, 50.95; H, 4.51; N, 4.45. Melting Point = 96-98°C. (II-59). To a 100 mL round bottom flask was added "-52 (1.02 g, 3.17 mmol), DCM (2.00 mL) and TFA (2.00 mL). The resulting solution stirred for 30 min. The excess TFA and DCM were removed and CHC13 (3 x 15.0 mL) was added and subsequently taken off to remove any residual TFA. The product was precipitated out of the crude residue using ether/petroleum ether to afford the product as a white solid. Yield (0.953 g, 90.0%). 1H NMR (500MHz), DMSO: 8 3.76 (s, 3H), 4.66 (m, 2H), 5.14-5.18 (m, 2H), 5.26 (s, 1H), 5.82 (m, 1H), 7.01 (d, J = 8.9 Hz, 2H), 7.41 (d, J = 8.8 Hz, 2H), 8.91 (s, 3H); 13C NMR (125MHz), DMSO: 8 54.8, 55.2, 65.9, 114.3, 117.2 (d, J = 300.3 Hz), 118.2, 124.4, 129.6, 131.5, 158.1 (q, J = 30.6 Hz), 160.0, 168.4. IR: (KBr) 3088 (br) cm", 1749 cm", 1680 cm'1. HRMS: [M + H]° = 222.1140, calculated for C14H15N03, 222.1130. Anal. Calcd. For C14H16F3N05: C, 50.15; H, 4.81; N, 4.18. Found: C, 50.29; H, 4.64; N, 4.19. Melting Point = 88-90°C. (ll-60). To a 100 mL round bottom flask was added "-53 (0.600 g, 1.94 mmol), DCM (2.00 mL) and TFA (2.00 mL). The resulting solution stirred for 30 min. The excess TFA and DCM were removed and CHC13 (3 x 15.0 mL) was added and subsequently taken off to remove any residual TFA. The product was precipitated out of the crude residue using ether/petroleum ether to afford the product as a white solid. Yield (0.424 g, 68.0%). 1H NMR (500MHz), DMSO: 8 119 4.67 (m, 2H), 5.13-5.19 (m, 2H), 5.40 (s, 1H), 5.82 (m, 1H), 7.33 (m, 2H), 7.56 (m, 2H), 8.98 (s, 3H); 13C NMR (125MHz), DMSO: 8 54.6, 66.1, 115.9 (d, J = 22.1 Hz), 117.2 (q, J = 300.1 Hz), 118.2, 128.9 (d, J = 3.2 Hz), 130.6 (d, J = 8.8 Hz), 131.4, 158.0 (q, J = 30.9 Hz), 162.6 (d, J = 246.2 Hz), 168.0. IR: (KBr) 3100 (br) em", 1749 cm", 1680 cm". HRMS: [M + H]+ = 210.0933, calculated for C11H13FNO2, 210.0930. Anal. Calcd. For C13H13F4NO4: C, 48.30; H, 4.05; N, 4.33. Found: C, 48.33; H, 3.77; N, 4.23. Melting Point = 68-70°C. (ll-61). To a 100 mL round bottom flask was added "-54 (1.11 g, 3.26 mmol), DCM (2.00 mL) and TFA (2.00 mL). The resulting solution stirred for 30 min. The excess TFA and DCM were removed and CHC|3 (3 x 15.0 mL) was added and subsequently taken off to remove any residual TFA. The product was precipitated out of the crude residue using ether/petroleum ether to afford the product as a white solid. Yield (0.962 g, 83.0%). 1H NMR (500MHz), DMSO: 8 4.65 (m, 2H), 5.02-5.09 (m, 2H), 5.75 (m, 1H), 6.19 (s, 1H), 7.58-7.72 (m, 4H), 8.04 (t, J = 9.0 Hz, 2H), 8.31 (d, J = 8.6 Hz, 1H), 9.15 (s, 1H); 13C NMR (125MHz), DMSO: 8 51.3, 66.1, 117.2 (q, J = 300.3 Hz), 118.0, 123.3, 125.3, 125.9, 126.5, 127.2, 128.8, 129.1, 130.2, 130.5, 131.4, 133.5, 158.2 (q, J = 31.1 Hz), 168.5. IR: (KBr) 3090 (br) cm", 1730 cm", 1667 cm". HRMS: [M + Hr = 242.1192, calculated for C15H15NO2, 242.1181. Anal. Calcd. For C17H16F3NO4: C, 57.47; H, 4.54; N, 3.94. Found: C, 57.33; H, 4.47; N, 3.87. Melting Point = 112- 114°C. (ll-62). To a 100 mL round bottom flask was added "-55 (1.39 g, 4.56 mmol), DCM (3.00 mL) and TFA (3.00 mL). The resulting solution stirred for 30 120 min. The excess TFA and DCM were removed and CHCI3 (3 x 15.0 mL) was added and subsequently taken off to remove any residual TFA. The product was precipitated out of the crude residue using ether to afford the product as a white solid. Yield (1.32 g, 91.0%). 1H NMR (500MHz), DMSO: 8 1.53 (s, 3H), 4.58 (dd, J = 13.4, 33.9 Hz, 2H), 4.76 (s, 1H), 4.82 (s, 1H), 5.35 (s, 1H), 7.42-7.53 (m, 5H), 9.00 (s, 3H); 130 NMR (125MHz), DMSO: 8 18.7, 55.3, 68.4, 112.9, 117.2 (q, J = 300.1 Hz), 128.0, 129.0, 129.5, 132.6, 139.0, 158.0 (q, J = 31.1 Hz), 168.1. IR: (KBr) 3150 (br) cm'1, 1743 cm“, 1680 cm". HRMS: [M + H]+ = 206.1182, calculated for C12H16NO2, 206.1181. Anal. Calcd. For C14H16F3NO4: C, 52.67; H, 5.05; N, 4.39. Found: C, 52.63; H, 4.99; N, 4.34. Melting Point = 142-144°C. (II-63). To a flame dried 100 mL round bottom flask was added "-56 (1 .00 g, 4.11 mmol) and anhydrous DCM (50.0 mL). Then the solution was brought down to 0°C and anhydrous TEA (0.630 mL, 4.52 mmol) was added followed by dropwise addition of ethoxycarbonyl isothiocyanate (0.560 mL, 4.93 mmol). The solution was allowed to warm to room temperature overnight while stirring under a nitrogen atmosphere. The solvent was removed and ether (30.0 mL) was added to the crude residue and was washed with sat. sodium bicarbonate. The organics were dried using anhydrous sodium sulfate and concentrated. The crude residue was purified by column chromatography (silica gel, 30% EtOAc; 70% hexane) affording the product as a whitish solid. Yield (1.04 g, 97.0%). 1H NMR (500MHz), CDC13: 8 1.28 (t, J = 7.1 Hz, 3H), 1.54 (d, J = 7.2 Hz, 3H), 4.21 (q, J = 7.1 Hz, 2H), 4.65 (m, 2H), 5.00 (p, J = 7.2 Hz, 1H), 5.22-5.26 (m, 1H), 5.30-5.35 (m, 1H), 5.89 (m, 1H), 8.15 (s, 1H), 10.14 (d, J = 5.9 Hz, 1H); 13c NMR 121 droowis solutior (125MHz), CDCI3: 8 14.0, 17.5, 53.6, 62.7, 66.0, 118.7, 131.3, 152.5, 171.4, 178.8. IR: (KBr) 3295 cm", 3240 cm", 1750 cm", 1725 cm". HRMS: [M + Hr =261.0920, calculated for C10H17N2O4S, 261.0909. Anal. Calcd. For C10H15N2O4S: C, 46.14; H, 6.20; N, 10.76. Found: C, 46.88; H, 6.06; N, 10.80. Melting Point = 43-45°C. (ll-64). To a flame dried 100 mL round bottom flask was added "-57 (1 .27 g, 3.98 mmol) and anhydrous DCM (50.0 mL). Then the solution was brought down to 0°C and anhydrous TEA (0.610 mL, 4.38 mmol) was added followed by dropwise addition of ethoxycarbonyl isothiocyanate (0.540 mL, 4.78 mmol). The solution was allowed to warm to room temperature overnight while stirring under a nitrogen atmosphere. The solvent was removed and ether (30.0 mL) was added to the crude residue and was washed with sat. sodium bicarbonate. The organics were dried using anhydrous sodium sulfate and concentrated. The product was recrystallized from the crude residue with EtOAc/hexanes affording the product as a whitish solid. Yield (1.08 g, 81.0%). 1H NMR (500MHz), CDC13: 8 1.27 (t, J = 7.1 Hz, 3H), 3.20-3.34 (m, 2H), 4.19 (q, J = 7.1 Hz, 2H), 4.60 (m, 2H), 5.24 (m, 3H), 5.82 (m, 1H), 7.15-7.30 (m, 5H), 8.10 (s, 1H), 10.08 (d, J = 5.5 Hz, 1H); 13C NMR (125MHz), CDC13: 8 14.0, 37.2, 59.2, 62.8, 66.1, 118.9, 127.1, 128.5, 129.2, 131.2, 135.3, 152.3, 170.0, 179.1. IR: (KBr) 3290 cm'1 , 3225 cm", 1730 cm'1(with shoulder). HRMS: [M + H]’° =337.1227, calculated for C16H21N2O4S, 337.1222. Anal. Calcd. For C16H20N2O4S: C, 57.12; H, 5.99; N, 8.33. Found: C, 55.80; H, 5.80; N, 8.30. Melting Point = 79-81°C. 122 0 g. 4.85 33310 :mpws solution amrog abedt eqaoc emde r 79%lm r1119 (1 (ll-65). To a flame dried 100 mL round bottom flask was added "-58 (1.48 g, 4.85 mmol) and anhydrous DCM (50.0 mL). Then the solution was brought down to 0°C and anhydrous TEA (0.740 mL, 5.33 mmol) was added followed by dropwise addition of ethoxycarbonyl isothiocyanate (0.660 mL, 5.82 mmol). The solution was allowed to warm to room temperature overnight while stirring under a nitrogen atmosphere. The solvent was removed and ether (30.0 mL) was added to the crude residue and was washed with sat. sodium bicarbonate. The organics were dried using anhydrous sodium sulfate and concentrated. The crude residue was purified by column chromatography (silica gel, 30% EtOAc; 70% hexane) affording the product as a whitish solid. Yield (1.39 g, 89.0%). 1H NMR (500MHz), CDC13: 8 1.29 (t, J = 7.1 Hz, 3H), 4.23 (m, 2H), 4.64 (m, 2H), 5.14-5.22 (m, 2H), 5.81 (m, 1H), 5.98 (d, J = 6.9 Hz, 1H), 7.31-7.45 (m, 5H), 8.06 (s, 1H), 10.59 (d, J = 6.4 Hz, 1H); 13c NMR (125MHz), CDCI3: 8 14.0, 61.8, 62.8, 66.2, 118.5, 127.5, 128.7, 128.9, 131.1, 135.0, 152.5, 169.3, 178.9. IR: (KBr) 3290 cm" , 3225 cm", 1750 cm", 1720 cm". HRMS: [M + H]+ =323.1070, calculated for C15H19N2O4S, 323.1066. Anal. Calcd. For C15H13N2O4S: C, 55.88; H, 5.63; N, 8.69. Found: C, 55.46; H, 5.51; N, 8.68. Melting Point = 44-46’C. (II-66). To a flame dried 100 mL round bottom flask was added "-59 (0.900 g, 2.69 mmol) and anhydrous DCM (50.0 mL). Then the solution was brought down to 0°C and anhydrous TEA (0.410 mL, 2.96 mmol) was added followed by dropwise addition of ethoxycarbonyl isothiocyanate (0.360 mL, 3.22 mmol). The solution was allowed to warm to room temperature overnight while stirring under a nitrogen atmosphere. The solvent was removed and ether (30.0 123 mL) was added to the crude residue and was washed with sat. sodium bicarbonate. The organics were dried using anhydrous sodium sulfate and concentrated. The crude residue was purified by column chromatography (silica gel, 30% EtOAc; 70% hexane) affording the product as an oil. Yield (0.917 g, 97.0%). 1H NMR (500MHz), CDCl3: 8 1.31 (t, J = 7.2 Hz, 3H), 3.80 (s, 3H), 4.22- 4.26 (m, 2H), 4.60-4.72 (m, 2H), 5.19-5.25 (m, 2H), 5.85 (m, 1H), 5.93 (d, J = 6.8 Hz, 1H), 6.90 (d, J = 8.8 Hz, 2H), 7.36 (d, J = 8.5 Hz, 2H), 8.20 (s, 1H), 10.55 (d, J = 6.5 Hz, 1H); 13C NMR (125MHz), CDCI3: 8 14.0, 55.2, 61.2, 62.8, 66.2, 114.3, 118.5, 127.1, 128.8, 131.2, 152.5, 159.8, 169.5, 178.7. IR: (KBr) 3289 cm'1 , 3226 cm", 1751 cm", 1724 cm". HRMS: [M + Hr = 353.1180, calculated for C16H21N2O5S, 353.1171. Anal. Calcd. For C15H20N205S: C, 54.53; H, 5.72; N, 7.95. Found: C, 52.68; H, 5.45; N, 7.55. (II-67). To a flame dried 50 mL round bottom flask was added "-60 (0.394 g, 1.22 mmol) and anhydrous DCM (20.0 mL). Then the solution was brought down to 0°C and anhydrous TEA (0.190 mL, 1.34 mmol) was added followed by dropwise addition of ethoxycarbonyl isothiocyanate (0.160 mL, 1.46 mmol). The solution was allowed to warm to room temperature overnight while stirring under a nitrogen atmosphere. The solvent was removed and ether (30.0 mL) was added to the crude residue and was washed with sat. sodium bicarbonate. The organics were dried using anhydrous sodium sulfate and concentrated. The crude residue was purified by column chromatography (silica gel, 30% EtOAc; 70% hexane) affording the product as an oil. Yield (0.383 g, 92.0%). 1H NMR (500MHz), cocr: 8 1.29 (t, J = 7.1 Hz, 3H), 4.20-4.26 (m, 2H), 4.59-4.69 (m, 124 2H), 5.1 7.3-87.4 CDClgz 1 Hz}. 13‘ IR: (KB 2H), 5.15-5.25 (m, 2H), 5.81 (m, 1H), 5.96 (d, J = 6.8 Hz, 1H), 7.02-7.07 (m, 2H), 7.38-7.42 (m, 2H), 8.10 (s, 1H), 10.61 (d, J = 6.3 Hz, 1H); 13C NMR (125MHz), CDCI3: 8 14.1, 61.0, 62.9, 66.4, 115.9 (d, J = 22.1 Hz), 118.8, 129.3 (d, J = 8.8 Hz), 131.07 (d, J = 2.3 Hz), 131.1, 152.6, 162.8 (d, J = 248.1 Hz), 169.2, 178.9. IR: (KBr) 3282 cm'1 , 3232 cm", 1751 cm", 1724 cm". HRMS: [M + H]+ = 341.0974, calculated for C15H13FN2O4S, 341.0971. Anal. Calcd. For C15H17FN2O4S: C, 52.93; H, 5.03; N, 8.23. Found: C, 51.76; H, 4.89; N, 8.07. (II-68). To a flame dried 100 mL round bottom flask was added “-61 (0.902 g, 2.54 mmol) and anhydrous DCM (50.0 mL). Then the solution was brought down to 0°C and anhydrous TEA (0.390 mL, 2.79 mmol) was added followed by dropwise addition of ethoxycarbonyl isothiocyanate (0.340 mL, 3.05 mmol). The solution was allowed to warm to room temperature overnight while stirring under a nitrogen atmosphere. The solvent was removed and ether (30.0 mL) was added to the crude residue and was washed with sat. sodium bicarbonate. The organics were dried using anhydrous sodium sulfate and concentrated. The crude residue was purified by column chromatography (silica gel, 30% EtOAc; 70% hexane) affording the product as an oil. Yield (0.897 g, 95.0%). 1H NMR (500MHz), CDCI3: 8 1.24 (t, J = 7.1 Hz, 3H), 4.12-4.22 (m, 2H), 4.58-4.72 (m, 2H), 5.10-5.18 (m, 2H), 5.78 (m, 1H), 6.80 (d, J = 7.3 Hz, 1H), 7.42-7.60 (m, 4H), 7.85 (t, J = 8.6 Hz, 2H), 8.20 (m, 2H), 10.57 (d, J = 7.1 Hz, 1H); 13c NMR (125MHz), coca: 8 14.0, 58.9, 62.8, 66.3, 118.5, 123.2, 125.2, 126.0, 126.1, 127.0, 128.8, 129.7, 131.0, 131.1, 131.2, 134.0, 152.5, 169.7, 179.2. IR: (KBr) 3289 cm" , 3226 cm", 1749 cm“, 1718 cm". HRMS: [M + Hr = 125 373.1222. C. 61.27; I (ll-I 3 3.13 rr torn 10C dropwise solution v a nitroge added 10 organics product . the prod (51.28 (1 5.8 Hz, : 5-5 H; 127.5, . 3232 Cl CliHati 8.33, F, 373.1222, calculated for C19H21N204S, 373.1222. Anal. Calcd. For C19H20N204S: C, 61.27; H, 5.41; N, 7.52. Found: C, 58.71; H, 4.85; N, 7.15. (II-69). To a flame dried 100 mL round bottom flask was added "-62 (1 .00 g, 3.13 mmol) and anhydrous DCM (50.0 mL). Then the solution was brought down to 0°C and anhydrous TEA (0.480 mL, 3.45 mmol) was added followed by dropwise addition of ethoxycarbonyl isothiocyanate (0.420 mL, 3.76 mmol). The solution was allowed to warm to room temperature overnight while stirring under a nitrogen atmosphere. The solvent was removed and ether (30.0 mL) was added to the crude residue and was washed with sat. sodium bicarbonate. The organics were dried using anhydrous sodium sulfate and concentrated. The product was recrystallized from the crude residue with EtOAc/hexanes affording the product as a whitish solid. Yield (0.960 g, 91 .0%). 1H NMR (500MHz), CDCI3: 5 1.28 (t, J = 7.2 Hz, 3H), 1.59 (s, 3H), 4.23 (m, 2H), 4.55 (m, 2H), 4.83 (d, J = 5.8 Hz, 2H), 5.98 (d, J = 6.9 Hz, 1H), 7.31-7.44 (m, 5H), 8.08 (s, 1H), 10.62 (d, J = 6.6 Hz, 1H); 13C NMR (125MHz), CDCI3: 5 14.1, 19.1, 61.9, 62.9, 68.9, 113.5, 127.5, 128.8, 128.9, 135.1, 139.0, 152.5, 169.4, 178.9. IR: (KBr) 3289 cm‘1 , 3232 cm“, 1749 cm‘1, 1724 cm". HRMS: [M + H]+ = 337.1230, calculated for C16H21N204S, 337.1222. Anal. Calcd. For C16H20N204S: C, 57.12; H, 5.99; N, 8.33. Found: C, 55.96; H, 5.84; N, 8.03. Melting Point = 62-64’C. (ll-71). To a flame dried 50 mL round bottom flask was added "-70118'119 (1.55 g, 7.49 mmol) and anhydrous CH3CN. Then allyl alcohol (2.56 mL, 37.5 mmol) was added and the mixture stirred at room temperature under nitrogen overnight. The solvent was removed and the residue was put into solution with 126 EtOAc (0.200 L) and washed with sat. sodium bicarbonate (1 x 0.100 L). The organics were dried using anhydrous sodium sulfate and concentrated to give pure product as a solid. Yield (1.70 g, 99.0%). 1H NMR (500MHz), acetone: 5 4.85 (m, 2H), 5.27-5.31 (m, 1H), 5.42-5.48 (m, 1H), 6.07 (m, 1H), 7.27-7.32 (m, 2H), 7.56-7.60 (m, 1H), 8.32 (m, 1H), 8.46 (s, 1H), 11.35 (s, 1H); 13C NMR (125MHz), Acetone: 5 66.5, 113.2, 114.2, 119.0, 122.5, 123.6, 124.7, 126.8, 132.8, 137.7, 138.2, 164.0, 179.5. IR: (KBr) 3200om'1 , 1743om", 1620 cm". HRMS: [M + H]+ =230.0819, calculated for C13H12NO3, 230.0817. Anal. Calcd. For C13H11N03: C, 68.11; H, 4.84; N, 6.11. Found: C, 66.14; H, 4.80; N, 5.93. Melting Point = 159-161°C. (ll-72). To a flame dried 250 mL round bottom flask was added "-71 (1.70 g, 7.42 mmol) and anhydrous DCM (0.100 L). Then TsCl (2.82 g, 14.8 mmol), DMAP (2.26 g, 18.6 mmol), and DIPEA (3.20 mL, 18.6 mmol) were added and the mixture stirred at room temperature overnight under a nitrogen atmosphere. The resulting brown solution was washed with 5% HCI (1 x 30.0 mL) and brine (1 x 30.0 mL) and the organics were dried using anhydrous sodium sulfate and concentrated. The crude residue was purified by column chromatography (silica gel, 30% EtOAc; 70% Hexanes) and recrystallized with EtOAc/hexanes to afford the product as a solid. Yield (2.46 g, 86.0%). 1H NMR (500MHz), CDCI3: 8 2.35 (s, 3H), 4.86 (m, 2H), 5.35 (m, 1H), 5.46 (m, 1H), 6.03 (m, 1H), 7.27 (d, J = 8.0 Hz, 2H), 7.37 (m, 2H), 7.85 (d, J = 7.4 Hz, 2H), 7.94 (m, 1H), 8.34 (m, 1H), 8.82 (s, 1H); 13C NMR (125MHz), CDCI3: 6 21.6, 66.9, 113.1, 116.9, 120.0, 122.9, 125.2, 126.1, 127.3, 127.6, 130.3, 130.7, 134.2, 134.4, 136.8, 146.2, 161.2, 127 178.3. IR: (KBr) 1730cm", 1674 cm". HRMS: [M + H]+ = 384.0914, calculated for C20H18N058, 383.0906. Anal. Calcd. For ConnNOsS: C, 62.65; H, 4.47; N, 3.65. Found: C, 62.47; H, 4.48; N, 3.64. Melting Point = 104-106°C. (II-73). To a 250 mL round bottom flask was added "-72 (2.30 g, 6.01 mmol) and dioxane (0.100 L). Then hydroxylamine hydrochloride (1.24 g, 18.0 mmol) and pyridine (1.55 mL, 19.2 mmol) were added and the mixture refluxed under nitrogen overnight (enough water to dissolve the hydroxylamine salt was added). The solvent was taken off and the residue was put into solution with EtOAc (0.100 L). The organics were washed with 1% HCI (1 x 30.0 mL) and brine (1 x 30.0 mL), then dried using anhydrous sodium sulfate and concentrated. The crude residue was purified by column chromatography (silica gel, 30% EtOAc; 70% Hexanes) affording the product as an oil (mixture of isomers). Yield (2.22 g, 93.0%). Isomer A: 1H NMR (500MHz), CDCI3: 6 2.32 (s, 3H), 4.83 (d, J = 5.9 Hz, 2H), 5.28 (d, J = 10.5 Hz, 1H), 5.38 (d, J = 17.2 Hz, 1H), 5.96 (m, 1H), 7.21 (d, J = 8.5 Hz, 2H), 7.25 (t, J = 8.0 Hz, 1H), 7.35 (t, J = 8.0 Hz, 1H), 7.52 (d, J = 8.1 Hz, 1H), 7.83 (d, J = 8.4 Hz, 2H), 8.01 (d, J = 8.3 Hz, 1H), 8.25 (s, 1H), 10.59 (s, 1H); 13C NMR (125MHz), CDCI3: 8 21.4, 66.8, 109.5, 113.2, 119.6, 122.4, 123.3, 124.8, 126.9, 128.0, 129.9, 130.1, 130.9, 133.9, 134.7, 143.1, 145.3, 162.7. Isomer B: 1H NMR (500MHz), CDCI3: 5 2.25 (s, 3H), 4.91 (d, J = 5.9 Hz, 2H), 5.33 (d, J = 10.3 Hz, 1H), 5.45 (d, J = 17.2 Hz, 1H), 6.02 (m, 1H), 7.14-7.21 (m, 3H), 7.32 (t, J = 7.5 Hz, 1H), 7.74 (d, J = 8.4 Hz, 2H), 7.76 (s, 1H), 7.94 (d, J = 8.5 Hz, 1H), 8.00 (d, J = 8.1 Hz, 1H), 8.99 (s, 1H); 13C NMR (125MHz), CDCI3: 5 21.3, 66.5, 113.2, 113.6, 119.7, 122.9, 124.1, 125.6, 126.8, 128 126.9, 127.3, 129.9, 130.7, 134.4, 135.0, 145.4, 146.5, 162.4. IR: (NaCI) 3276 cm", 17300m'1. HRMS: [M + H]+ = 399.1017, calculated for C20H19N2058, 399.1015. Anal. Calcd. For C20H18N205SI C, 60.29; H, 4.55; N, 7.03. Found: C, 58.91; H, 4.30; N, 6.82. (ll-74). To a 250 mL round bottom flask was added water (55.0 mL) and AcOH (55.0 mL). Then "-73 (2.12 g, 5.33 mmol) was dissolved in THF (25.0 mL) and was added to the aqueous acid. The mixture was brought down to 0°C and zinc (3.46 g, 53.3 mmol) was then slowly added in small portions over 20 min. The suspension stirred at 0°C for 2 h. The solid was filtered off and the filtrate was reduced and then brought to a pH of 8 using concentrated ammonium hydroxide. The amine was extracted into ethyl acetate (4 x 50.0 mL), dried using anhydrous sodium sulfate and concentrated. The crude residue was then put into solution with DCM (5.00 mL) and TFA (3.00 mL) was added. The mixture stirred for 10 min and then the solvent and excess TFA was removed. CHCI3 (3 x 15.0 mL) was added and subsequently taken off to remove any residual TFA. The product was precipitated out of the crude residue using ether/petroleum ether to afford the product as a white solid. Yield (2.25 g, 85.0%). 1H NMR (500MHz), DMSO: 6 2.31 (s, 3H), 4.66 (m, 2H), 5.06 (m, 1H), 5.09 (m, 1H), 5.71 (s, 1H), 5.75 (m, 1H), 7.33 (t, J = 8.1 Hz, 1H), 7.40 (m, 3H), 7.75 (d, J = 8.1 Hz, 1H), 7.85 (d, J = 7.5 Hz, 2H), 7.94 (d, J = 7.3 Hz, 1H), 8.09 (s, 1H), 9.12 (s, 3H); 130 NMR (125MHz), DMSO: 5 20.9, 47.8, 66.2, 113.1, 114.1, 117.2 (q, J = 299.2 Hz), 118.1, 120.3, 123.6, 125.5, 126.7, 126.8, 127.8, 130.3, 131.3, 133.7, 133.8. 145.9, 158.3 (q. J = 31.3 Hz), 167.5. IR: (KBr) 3100 (br)cm'1, 1736 cm", 1574 129 cm". HRMS: [M + H]+ = 385.1235, calculated for C20H21N204S, 385.1222. Anal. Calcd. For C22H21F3N2068: C, 53.01; H, 4.25; N, 5.62. Found: C, 52.95; H, 4.05; N, 5.50. Melting Point = 170-172°C. (ll-75). To a flame dried 100 mL round bottom flask was added "-74 (1.30 g, 2.61 mmol) and anhydrous DCM (50.0 mL). Then the solution was brought down to 0°C and anhydrous TEA (0.400 mL, 2.87 mmol) was added followed by dropwise addition of ethoxycarbonyl isothiocyanate (0.350 mL, 3.13 mmol). The solution was allowed to warm to room temperature overnight while stirring under a nitrogen atmosphere. The solvent was removed and ether (30.0 mL) was added to the crude residue and was washed with sat. sodium bicarbonate. The organics were dried using anhydrous sodium sulfate and concentrated. The product was recrystallized from the crude residue with EtOAc/Hexanes affording a whitish solid. Yield (1.19 g, 89.0%). 1H NMR (500MHz), CDCI3: 5 1.26 (t, J = 7.2 Hz, 3H), 2.31 (s, 3H), 4.20 (m, 2H), 4.64 (m, 2H), 5.13-5.22 (m, 2H), 5.78 (m, 1H), 6.26 (d, J = 7.9 Hz, 1H), 7.19-7.25 (m, 3H), 7.30 (t, J = 8.3 Hz, 1H), 7.63 (d, J = 7.5 Hz, 1H), 7.72 (s, 1H), 7.74 (d, J = 8.4 Hz, 2H), 7.93 (d, J = 7.3 Hz, 1H), 8.04 (s, 1H), 10.59 (d, J = 6.9 Hz, 1H); 13c NMR (125MHz), CDCI3: 8 14.1, 21.5, 54.7, 62.9, 66.6, 113.7, 115.9, 119.0, 119.9, 123.6, 125.1, 126.2, 126.9, 128.2, 129.9, 131.0, 134.8, 135.0, 145.1, 152.4, 168.7, 179.0. IR: (KBr) 3282 cm", 3232 cm", 1749 cm", 1724 cm". HRMS: [M + H]* = 516.1273, calculated for C24H25N30582, 516.1263. Anal. Calcd. For C24H25N30682: C, 55.91; H, 4.89; N, 8.15. Found: C, 55.91; H, 4.79; N, 8.09. Melting Point = 117-119°C. 130 g. 0.45 1.34 W hen re the so 938% and til. then tr {3 x 3 sodium chrom; Whitish = 7.4, 3.06 (c 5H). 9. (II-76). To a flame dried 50 mL round bottom flask was added "-64 (0.150 g, 0.446 mmol) and anhydrous DCM (15.0 mL). Then anhydrous TEA (0.200 mL, 1.34 mmol) was added and the mixture was cooled to 0°C. EDCI (0.189 g, 0.982 mmol) was then added and the mixture stirred at 0°C under nitrogen for 1 h and then refluxed for 15 h. After the first step was completed, (as indicated by TLC), the solution was cooled to 0°C and a solution of NaH (0.0890 g, 2.33 mmol) in MeOH (5.00 mL) was added dropwise. The cloudy mixture stirred at 0°C for 1 h and then at room temperature for 2 h. The mixture was acidified with 5% HCl and then the solvents were removed. The aqueous mixture was extracted with EtOAc (3 x 30.0 mL) and then the organics were combined, dried using anhydrous sodium sulfate and concentrated. The crude residue was purified by column chromatography (silica gel, 20% EtOAc; 80% DCM) affording the product as a whitish solid. Yield (0.0200 g, 19.0%). 1H NMR (500MHz), Acetone: 5 2.51 (dd, J = 7.4, 13.8 Hz, 1H), 2.61 (dd, J = 7.3, 13.9 Hz, 1H), 2.90 (d, J = 13.7 Hz, 1H), 3.06 (d, J = 13.7 Hz, 1H), 5.15 (m, 2H), 5.79 (m, 1H), 7.07 (s, 1H), 7.20-7.28 (m, 5H), 9.28 (s, 1H); 13C NMR (125MHz), Acetone: 5 42.1, 42.9, 67.9, 120.2, 127.6, 128.8, 131.1, 132.3, 138.0, 158.5, 178.7. IR: (KBr) 3220 cm'1 (broad), 1761cm'1, 1711 cm". HRMS: [M + H]+ = 231.1141, calculated for C13H15N202, 231.1134. Anal. Calcd. For C13H14N202: C, 67.81; H, 6.13; N, 12.17. Found: C, 66.59; H, 6.14; N, 12.09. Melting Point = 203-205‘C. (ll-77). To a flame dried 50 mL round bottom flask was added "-65 (0.150 g, 0.466 mmol) and anhydrous DCM (15.0 mL). Then anhydrous TEA (0.200 mL, 1.40 mmol) was added and the mixture was cooled to 0°C. EDCI (0.197 g, 1.02 131 mmol) was then added and the mixture stirred at 0°C under nitrogen for 1 h and then refluxed for 15 h. After the first step was completed, (as indicated by TLC), the solution was cooled to 0°C and a solution of NaH (0.0930 g, 2.33 mmol) in MeOH (5.00 mL) was added dropwise. The cloudy mixture stirred at 0°C for 1 h and then at room temperature for 2 h. The mixture was acidified with 5% HCI and then the solvents were removed. The aqueous mixture was extracted with EtOAc (3 x 30 mL) and then the organics were combined, dried using anhydrous sodium sulfate and concentrated. The crude residue was purified by column chromatography (silica gel, 20% EtOAc; 80% DCM) affording the product as a whitish solid. Yield (0.0700 g, 70.0%). 1H NMR (500MHz), acetone: 5 2.78 (dd, J = 7.4, 14.0 Hz, 1H), 2.95 (dd, J = 7.3, 13.9 Hz, 1H), 5.13 (d, J = 10.1 Hz, 1H), 5.21 (d, J = 17.1 Hz, 1H), 5.73 (m, 1H), 7.33 (t, J = 7.4 Hz, 1H), 7.40 (t, J = 8.2 Hz, 2H), 7.64 (d, J = 7.9 Hz, 2H), 7.68 (s, 1H), 9.65 (s, 1H); 13C NMR (125MHz), acetone: 5 43.5, 68.6, 120.8, 126.3, 128.7, 129.3, 132.0, 139.7, 157.0, 175.9. IR: (KBr) 3245 cm'1 (broad), 1774cm'1, 1724 cm". HRMS: [M + H]+ = 217.0979, calculated for C12H13N202, 217.0977. Anal. Calcd. For C12H12N202: C, 66.65; H, 5.59; N, 12.96. Found: C, 66.43; H, 5.49; N, 12.90. Melting Point = 172-174°C. (ll-78). To a flame dried 50 mL round bottom flask was added "-66 (0.155 g, 0.440 mmol) and anhydrous DCM (15.0 mL). Then anhydrous TEA (0.180 mL, 1.32 mmol) was added and the mixture was cooled to 0°C. EDCI (0.186 g, 0.968 mmol) was then added and the mixture stirred at 0°C under nitrogen for 1 h and then refluxed for 15 h. After the first step was completed, (as indicated by TLC), the solution was cooled to 0°C and a solution of NaH (0.0880 g, 2.20 mmol) in 132 MeOH (5.00 mL) was added dropwise. The cloudy mixture stirred at 0°C for 1 h and then at room temperature for 2 h. The mixture was acidified with 5% HCI and then the solvents were removed. The aqueous mixture was extracted with EtOAc (3 x 30.0 mL) and then the organics were combined, dried using anhydrous sodium sulfate and concentrated. The crude residue was purified by column chromatography (silica gel, 30% EtOAc; 70% DCM) affording the product as a whitish solid. Yield (0.0720 mg, 67.0%). 1H NMR (500MHz), acetone: 5 2.74 (dd, J = 7.3, 14.1 Hz, 1H), 2.92 (dd, J = 7.9, 14.1 Hz, 1H), 3.78 (s, 3H), 5.12 (m, 1H), 5.20 (m, 1H), 5.72 (m, 1H), 6.94 (d, J = 9.0 Hz, 2H), 7.52 (d, J = 9.0 Hz, 2H), 7.64 (s, 1H), 9.63 (s, 1H); 13C NMR (125MHz), Acetone: 5 43.4, 55.5, 68.2, 114.6, 120.6, 127.6, 131.7, 132.2, 156.9, 160.3, 176.1. IR: (KBr) 3251 cm'1 (broad), 1774cm“, 1724cm". HRMS: [M + H]* = 247.1088, calculated for C13H15N203, 247.1083. Anal. Calcd. For C13H14N203: C, 63.40; H, 5.73; N, 11.38. Found: C, 62.74; H, 5.53; N, 11.35. Melting Point = 166-168°C. (ll-79). To a flame dried 50 mL round bottom flask was added "-67 (0.151 g, 0.444 mmol) and anhydrous DCM (15.0 mL). Then anhydrous TEA (0.180 mL, 1.33 mmol) was added and the mixture was cooled to 0°C. EDCI (0.188 g, 0.977 mmol) was then added and the mixture stirred at 0°C under nitrogen for 1 h and then refluxed for 15 h. After the first step was completed, (as indicated by TLC), the solution was cooled to 0°C and a solution of NaH (0.0880 g, 2.20 mmol) in MeOH (5.00 mL) was added dropwise. The cloudy mixture stirred at 0°C for 1 h and then at room temperature for 2 h. The mixture was acidified with 5% HCI and then the solvents were removed. The aqueous mixture was extracted with EtOAc 133 (3 x 30.0 mL) and then the organics were combined, dried using anhydrous sodium sulfate and concentrated. The crude residue was purified by column chromatography (silica gel, 30% EtOAc; 70% DCM) affording the product as a whitish solid. Yield (0.0600 g, 57.0%). 1H NMR (500MHz), acetone: 5 2.76 (dd, J = 7.3, 14.0 Hz, 1H), 2.95 (dd, J = 7.2, 14.1 Hz, 1H), 5.14 (dd, J = 2.1, 10.1 Hz, 1H), 5.21 (dd, J = 2.1, 10.2 Hz, 1H), 5.72 (m, 1H), 7.16 (m, 2H), 7.67 (m, 2H), 7.75 (s, 1H), 9.74 (s, 1H); 13C NMR (125MHz), Acetone: 5 43.6, 68.2, 115.9 (d, J = 21.7 Hz), 121.0, 128.6 (d, J = 8.2 Hz), 131.8, 135.9 (d, J = 3.1 Hz), 156.8, 183.2 (d, J = 245.4 Hz), 175.8. IR: (KBr) 3220 cm" (broad), 1780cm", 1730cm". HRMS: [M + H]+ = 235.0876, calculated for C12H12FN202, 235.0883. Anal. Calcd. For C12H11FN202: C, 61.53; H, 4.73; N, 11.96. Found: C, 61.48; H, 4.61; N, 11.81. Melting Point = 178-180’C. (ll—80). To a flame dried 50 mL round bottom flask was added "-68 (0.156 g, 0.419 mmol) and anhydrous DCM (15.0 mL). Then anhydrous TEA (0.170 mL, 1.26 mmol) was added and the mixture was cooled to 0°C. EDCI (0.177 g, 0.922 mmol) was then added and the mixture stirred at 0°C under nitrogen for 1 h and then refluxed for 15 h. After the first step was completed, (as indicated by TLC), the solution was cooled to 0 °C and a solution of NaH (0.0840 g, 2.09 mmol) in MeOH (5.00 mL) was added dropwise. The cloudy mixture stirred at 0°C for 1 h and then at room temperature for 2 h. The mixture was acidified with 5% HCI and then the solvents were removed. The aqueous mixture was extracted with EtOAc (3 x 30.0 mL) and then the organics were combined, dried using anhydrous sodium sulfate and concentrated. The crude residue was purified by column 134 chromatography (silica gel, 30% EtOAc; 70% DCM) affording the product as an oil. Yield (0.0340 g, 31.0%). 1H NMR (500MHz), CDCI3: 5 3.07 (dd, J = 7.5, 14.2 Hz, 1H), 3.26 (dd, J = 6.9, 14.1 Hz, 1H), 5.12 (d, J = 11.1 Hz, 1H), 5.21 (d, J = 18.0 Hz, 1H), 5.71 (m, 1H), 7.33 (t, J = 7.7 Hz, 1H), 7.42-7.53 (m, 3H), 7.67 (d, J = 7.3 Hz, 1H), 7.78 (d, J = 8.2 Hz, 1H), 7.84 (d, J = 8.1 Hz, 1H), 8.22 (d, J = 7.6 Hz, 1H), 9.68 (s, 1H); 13C NMR (125MHz), CDCI3: 5 42.1, 69.7, 121.4, 124.4, 124.8, 125.6, 126.5, 129.6, 130.05, 130.08, 130.14, 132.3, 134.8, 157.6, 175.3. IR: (KBr) 3245 cm" (broad), 1774cm'1, 1724cm". HRMS: [M + H]* = 287.1134, calculated for C16H15N202, 267.1134. (ll-81). To a flame dried 50 mL round bottom flask was added "-75 (0.150 g, 0.291 mmol) and anhydrous DCM (15.0 mL). Then anhydrous TEA (0.120 mL, 0.874 mmol) was added and the mixture was cooled to 0°C. EDCI (0.123 g, 0.640 mmol) was then added and the mixture stirred at 0°C under nitrogen for 1 h and then refluxed for 15 h. After the first step was completed, (as indicated by TLC), the solution was cooled to 0°C and a solution of NaH (0.0580 g, 1.46 mmol) in MeOH (5.00 mL) was added dropwise. The cloudy mixture stirred at 0°C for 1 h and then at room temperature for 2 h. The mixture was acidified with 5% HCI and then the solvents were removed. The aqueous mixture was extracted with EtOAc (3 x 30.0 mL) and then the organics were combined, dried using anhydrous sodium sulfate and concentrated. The crude residue was purified by column chromatography (silica gel, 20% EtOAc; 80% DCM) affording the product as a whitish solid. Yield (0.0830 g, 69.0%). 1H NMR (500 MHz), acetone: 5 2.33 (s, 3H), 2.99 (dd, J = 7.2, 14.1 Hz, 1H), 3.05 (dd, J = 7.3, 14.2 135 Hz, 1H), 5.15 (d, J = 10.1 Hz, 1H), 5.22 (d, J = 17.5 Hz, 1H), 5.75 (m, 1H), 7.27 (t, J = 8.0 Hz, 1H), 7.36 (m, 3H), 7.65 (s, 1H), 7.83 (s, 1H), 7.88 (d, J = 8.3Hz. 3H), 8.01 (d, J = 8.5 Hz, 1H), 9.83 (s, 1H); 13C NMR (125 MHz), Acetone: 5 21.3, 41.5, 65.7, 114.4, 121.1, 121.8, 122.4, 124.2, 125.4, 125.7, 127.8, 128.7, 130.9, 131.4, 135.6, 136.3, 146.5, 156.7, 175.0. IR: (KBr) 3232 cm“, 17800m'1, 1730 cm". HRMS: [M + H]+ = 410.1178, calculated for C21H20N3O4S, 410.1175. Anal. Calcd. For C21H19N304S: C, 61.60; H, 4.68; N, 10.26. Found: C, 61.59; H, 4.60; N, 9.80. Melting Point = 226-228°C. (II-83). To a flame dried 100 mL round bottom flask was added "-82 (1.21 g, 3.24 mmol) and anhydrous DCM (50.0 mL). Then cyclopent-1-enylmetnanol127 (0.477 g, 4.87 mmol) and DMAP (0.0400 g, 0.324 mmol) were added and the mixture was brought down to 0°C. DCC (1 .00 g, 4.87 mmol) was then added and the mixture was allowed to warm to room temperature overnight while stirring under nitrogen. The white precipitate that formed was filtered off and the DCM filtrate was washed with brine (1 x 20.0 mL), dried using anhydrous sodium sulfate and concentrated. The crude residue was purified by column chromatography (silica gel, 20% EtOAc; 80% hexane) affording the product as a whitish solid. Yield (0.997 g, 68.0%). 1H NMR (500MHz), CDCl3: 5 1.82 (p, J = 7.5 Hz, 2H), 2.13 (s, 2H), 2.27 (s, 2H), 4.20 (t, J = 6.6 Hz, 1H), 4.38 (m, 2H), 4.70 (m, 2H), 5.40 (d, J = 7.4 Hz, 1H), 5.52 (s, 1H), 5.89 (d, J = 6.9 Hz, 1H), 7.28-7.40 (m, 9H), 7.57 (d, J = 7.3 Hz, 2H), 7.74 (d, J = 7.5 Hz, 2H); 13c NMR (125MHz), CDCI3: 5 23.1, 32.3, 32.5, 47.1, 58.0, 64.4, 67.1, 119.9, 125.0, 127.0, 127.1. 127.6, 128.5, 128.8, 129.1, 136.6, 138.1, 141.2, 143.7, 143.8, 155.3, 170.6. IR: 136 (NaCI) 3350 cm'1 , 1725 cm'1 (broad). HRMS: [M + H]+ = 454.2020, calculated for C29H28NO4, 454.2018. Anal. Calcd. For C29H27NO4: C, 76.80; H, 6.00; N, 3.09. Found: C, 76.97; H, 5.84; N, 3.12. Melting Point = 96-98°C. (ll-84). To a flame dried 100 mL round bottom flask was added "-82 (1.25 g, 3.35 mmol) and anhydrous DCM (50.0 mL). Then 3-methyl-2-buten-1-ol (0.690 mL, 6.70 mmol) and DMAP (0.0410 g, 0.335 mmol) were added and the mixture was brought down to 0°C. DCC (1.04 g, 5.03 mmol) was then added and the mixture was allowed to warm to room temperature overnight while stirring under nitrogen. The white precipitate that formed was filtered off and the DCM filtrate was washed with brine (1 x 20.0 mL), dried using anhydrous sodium sulfate and concentrated. The crude residue was purified by column chromatography (silica gel, 20% EtOAc; 80% hexane) affording the product as a whitish solid. Yield (1.15 g, 77.0%). 1H NMR (500MHz), CDCI3: 5 1.62 (s, 3H), 1.71 (s, 3H), 4.21 (t, J = 7.1 Hz, 1H), 4.35-4.44 (m, 2H), 4.54-4.72 (m, 2H), 5.27 (m, 1H), 5.40 (d, J = 7.6 Hz, 1H), 5.91 (d, J = 7.3 Hz, 1H), 7.27-7.41 (m, 9H), 7.58 (d, J = 7.3 Hz, 2H), 7.75 (d, J = 7.6 Hz, 2H); 13C NMR (125MHz), CDCI3: 5 17.9, 25.6, 47.1, 57.9, 62.7, 67.0, 117.7, 119.9, 125.0, 127.0, 127.1, 127.6, 128.4, 128.8, 136.7, 140.0, 141.2, 143.7, 143.8, 155.3, 170.7. IR: (NaCI) 3351 cm", 1724 cm’1 (broad). HRMS: [M + H]+ = 442.2019, calculated for C23H28NO4, 442.2018. Anal. Calcd. For C23H27NO4: C, 76.17; H, 6.16; N, 3.17. Found: C, 75.87; H, 6.10; N, 3.19. Melting Point = 108-110°C. (II-85). To a 100 mL round bottom flask was added “-83 (0.947 g, 2.09 mmol) and DCM (8.00 mL). The solution was brought down to 0°C and then 137 3385033 330gen empvec wthsat 333g 8 pudfied 90% DC lst G 222-21 13C N13" ”3000 pipertd Whoge rem0v wth S; Umng Dufifie 90$; [ Chara piperidine (2.00 mL, 20.9 mmol) was added and the mixture stirred under nitrogen for 1 h and then 1 h at room temperature. The solvent was then removed and the crude residue was taken up in EtOAc (40.0 mL) and washed with sat. NH4CI (1 x 10.0 mL) and brine (1 x 10.0 mL). The organics were dried using anhydrous sodium sulfate and concentrated. The crude residue was purified by column chromatography (silica gel, 30% EtOAc; 70% hexane, then 90% DCM 10% MeOH) affording the product as an oil. Yield (0.434 g, 90.0%).1H NMR (500MHz), CDCI3: 5 1.77-1.83 (m, 2H), 1.91 (s, 2H), 2.10-2.15 (m, 2H), 2.22-2.28 (m, 2H), 4.59 (s, 1H), 4.63 (m, 2H), 5.49 (m, 1H), 7.24-7.37 (m, 5H); 13c NMR (125MHz), c0013: 5 23.4, 32.5, 32.9, 59.0, 84.1, 127.0, 128.1, 128.9, 138.8, 140.6, 174.0. IR: (KBr) 3385 (br)cm'1, 3321 cm", 1733 cm". HRMS: [M + H]+ = 232.1341, calculated for C14H18N02, 232.1338. Anal. Calcd. For Cr4H17N02: C, 72.70; H, 7.41; N, 6.06. Found: C, 72.05; H, 7.30; N, 6.09. (ll-86). To a 100 mL round bottom flask was added “-84 (1.15 g, 2.61 mmol) and DCM (10.0 mL). The solution was brought down to 0°C and then piperidine (2.60 mL, 26.1 mmol) was added and the mixture stirred under nitrogen for 1 h and then 1 h at room temperature. The solvent was then removed and the crude residue was taken up in EtOAc (50.0 mL) and washed with sat. NH4C| (1 x 20.0 mL) and brine (1 x 20.0 mL). The organics were dried using anhydrous sodium sulfate and concentrated. The crude residue was purified by column chromatography (silica gel, 30% EtOAc; 70% hexane, then 90% DCM 10% MeOH) affording the product as an oil. Yield (0.488 g, 85.0%). Characterization is for the TFA salt. 1H NMR (500MHz), DMSO: 5 1.58 (s, 3H), 138 1.66 (s, 3H), 4.60 (dd, J = 7.3, 12.2 Hz, 1H), 4.67 (dd, J = 7.0, 11.7 Hz, 1H), 5.22 (m, 1H), 5.26 (s, 1H), 7.45 (m, 5H), 8.89 (s, 3H); 13C NMR (125MHz), DMSO: 5 17.7, 25.2, 55.3, 62.6, 117.2 (q, J = 300.5 Hz), 117.5, 128.1, 128.9, 129.5, 132.5, 139.8, 157.9 (q, J = 30.9 Hz), 168.3. IR: (KBr) 3164 (br) cm", 1736 cm", 1675 cm". HRMS: [M + H]+ = 220.1341, calculated for C13H13N02, 220.1338. Anal. Calcd. For C15H18F3NO4: C, 54.05; H, 5.44; N, 4.20. Found: C, 53.92; H, 5.26; N, 4.18. Melting Point = 118-120°C. (II-87). To a flame dried 100 mL round bottom flask was added "-85 (0.406 g, 1.76 mmol) and anhydrous DCM (50.0 mL). Then the solution was brought down to 0°C and ethoxycarbonyl isothiocyanate (0.240 mL, 2.11 mmol) was added dropwise. The solution was allowed to warm to room temperature overnight while stirring under a nitrogen atmosphere. The solvent was removed and ether (30.0 mL) was added to the crude residue and was washed with sat. sodium bicarbonate. The organics were dried using anhydrous sodium sulfate and concentrated. The crude residue was purified by column chromatography (silica gel, 20% EtOAc; 80% hexane) and the product was recrystallized with EtOAc/hexanes affording the product as a white solid. Yield (0.518 g, 81.0%). 1H NMR (500MHz), CDCI3: 5 1.29 (t, J = 7.1 Hz, 3H), 1.81 (m, 2H), 2.13 (m, 2H), 2.26 (m, 2H), 4.23 (m, 2H), 4.69 (s, 2H), 5.53 (s, 1H), 5.97 (d, J = 6.9 Hz, 1H), 7.30-7.42 (m, 5H), 8.08 (s, 1H), 10.62 (d, J = 6.5 Hz, 1H); 13C NMR (125MHz), CDCI3: 5 14.1, 23.1, 32.3, 32.5, 61.8, 62.8, 64.5, 127.5, 128.7, 128.9, 129.1. 135.2, 138.1, 152.5, 169.4, 178.8. IR: (KBr) 3289 cm'1, 3226 cm", 1743cm‘1, 1724 cm". HRMS: [M + H]+ = 383.1387, calculated for C13H23N204S, 383.1379. 139 Anal. Calc 5.78: N, 7 (Il- 10428 g. Drought c .185 add 31813190 and ethe sodium and con lislllca gr 9. 99.00.. is. 3H), 910.44 mlnot) Anal. Calcd. For C18H22N204S: C, 59.65; H, 6.12; N, 7.73. Found: C, 59.36; H, 5.78; N, 7.69. Melting Point = 58-60°C. (II-88). To a flame dried 100 mL round bottom flask was added "-86 (0.428 g, 1.95 mmol) and anhydrous DCM (50.0 mL). Then the solution was brought down to 0°C and ethoxycarbonyl isothiocyanate (0.260 mL, 2.35 mmol) was added dropwise. The solution was allowed to warm to room temperature overnight while stirring under a nitrogen atmosphere. The solvent was removed and ether (30.0 mL) was added to the crude residue and was washed with sat. sodium bicarbonate. The organics were dried using anhydrous sodium sulfate and concentrated. The crude residue was purified by column chromatography (silica gel, 20% EtOAc; 80% hexane) affording the product as an oil. Yield (0.678 g, 99.0%).1H NMR (500MHz), CDCI3: 5 1.29 (t, J = 7.1 Hz, 3H), 1.60 (s, 3H), 1.69 (s, 3H), 4.23 (m, 2H), 4.55 (dd, J = 7.3, 12.4 Hz, 1H), 4.68 (dd, J = 7.1, 12.2 Hz, 1H), 5.26 (m, 1H), 5.95 (d, J = 6.8 Hz, 1H), 7.30-7.44 (m, 5H), 7.96 (s, 1H), 10.59 (d, J = 6.6 Hz, 1H); 13C NMR (125MHz), CDCI3: 5 14.1, 18.0, 25.6, 61.9, 62.88, 62.89, 117.7, 127.5, 128.6, 128.9, 135.3, 140.0, 152.5, 169.6, 178.8. IR: (KBr) 3282 cm" , 3232 cm", 1724 cm'1(with shoulder). HRMS: [M + H]* = 351.1380, calculated for C17H23N204S, 351.1379. Anal. Calcd. For C17H22N204S: C, 58.27; H, 6.33; N, 7.99. Found: C, 54.58; H, 5.86; N, 7.46. (II-89). To a flame dried 50 mL round bottom flask was added "-69 (0.150 g, 0.446 mmol) and anhydrous DCM (15.0 mL). Then anhydrous TEA (0.190 mL, 1.34 mmol) was added and the mixture was cooled to 0°C. EDCI (0.188 g, 0.981 mmol) was then added and the mixture stirred at 0°C under nitrogen for 1 h and 140 Immref fiesoh MeOHl then refluxed for 15 h. After the first step was completed, (as indicated by TLC), the solution was cooled to 0°C and a solution of NaH (0.0890 g, 2.23 mmol) in MeOH (5.00 mL) was added dropwise. The cloudy mixture stirred at 0°C for 1 h and then at room temperature for 2 h. The mixture was acidified with 5% HCI and then the solvents were removed. The aqueous mixture was extracted with EtOAc (3 x 30.0 mL) and then the organics were combined, dried using anhydrous sodium sulfate and concentrated. The crude residue was purified by column chromatography (silica gel, 10% acetone; 90% DCM) affording the product as a whitish solid. Yield (0.0680 g, 66.0%). 1H NMR (500MHz), acetone: 5 1.70 (s, 3H), 2.70 (d, J = 13.2 Hz, 1H), 2.99 (d, J = 13.7 Hz, 1H), 4.85 (m, 1H), 4.91 (m, 1H), 7.30-7.42 (m, 3H), 7.66 (m, 2H), 7.69 (s, 1H), 9.64 (s, 1H); 13C NMR (125MHz), acetone: 5 24.1, 46.8, 68.7, 116.7, 126.3, 128.7, 129.2, 140.4, 140.7, 158.8, 178.1. IR: (KBr) 3243 cm", 1772 cm'1, 1724 cm". HRMS: [M + Hr = 231.1139, calculated for C13H15N202, 231.1134. Anal. Calcd. For C13H14N202: C, 67.81; H, 6.13; N, 12.17. Found: C, 66.99; H, 5.86; N, 11.81. Melting Point = 169- 171°C. (lI-90). To a flame dried 50 mL round bottom flask was added "-87 (0.150 g, 0.414 mmol) and anhydrous DCM (15.0 mL). Then anhydrous TEA (0.170 mL, 1.24 mmol) was added and the mixture was cooled to 0°C. EDCI (0.175 g, 0.911 mmol) was then added and the mixture stirred at 0°C under nitrogen for 1 h and then refluxed for 15 h. After the first step was completed, (as indicated by TLC), the solution was cooled to 0°C and a solution of NaH (0.0830 g, 2.07 mmol) in MeOH (5.00 mL) was added dropwise. The cloudy mixture stirred at 0°C for 1 h 141 arc-then. tre'ltlles 3X3OC 333m 8 mromato .nsepara "01.3) 1 3.45 (t, J (II 9. 0.429 129 mlr m”‘01) VI. thenrefi the sol” IlleOH ( and ther and then at room temperature for 2 h. The mixture was acidified with 5% HCI and then the solvents were removed. The aqueous mixture was extracted with EtOAc (3 x 30.0 mL) and then the organics were combined, dried using anhydrous sodium sulfate and concentrated. The crude residue was purified by column chromatography (silica gel, 10% acetone; 90% DCM) affording the two unseparatable diastereomers (1.321 ratio) as a whitish solid. Yield (0.0700 g, 66.0%). 1H NMR (500MHz), acetone: 5 1.28-1.96 (m, 8H), 2.26-2.38 (m, 4H), 3.45 (t, J = 7.4 Hz, 1H), 3.56 (t, J = 7.6 Hz, 1H), 4.03 (s, 1H), 4.73 (s, 1H), 4.94 (s, 1H), 5.03 (s, 1H), 7.32-7.43 (m, 6H), 7.55 (s, 1H), 7.64-7.71 (m, 4H), 7.87 (s, 1H), 9.66 (s, 1H); 13C NMR (125MHz), acetone: 5 24.9, 25.2, 35.9, 36.6, 49.9, 50.0, 71.2, 72.3, 108.4, 109.2, 126.7, 127.0, 128.63, 128.69, 129.21, 129.22, 139.7, 139.8, 150.7, 151.7, 157.2, 157.4, 175.7, 176.3. IR: (KBr) 3289 cm“, 1774 cm", 1718 cm'1. HRMS: [M + H]+ = 257.1295, calculated for C15H17N202, 257.1290. Anal. Calcd. For C15H16N202: C, 70.29; H, 6.29; N, 10.93. Found: C, 68.27; H, 6.37; N, 10.48. Melting Point = 234-236°C. (II-91). To a flame dried 50 mL round bottom flask was added "-88 (0.150 g, 0.429 mmol) and anhydrous DCM (15.0 mL). Then anhydrous TEA (0.180 mL, 1.29 mmol) was added and the mixture was cooled to 0°C. EDCI (0.181 g, 0.944 mmol) was then added and the mixture stirred at 0°C under nitrogen for 1 h and then refluxed for 15 h. After the first step was completed, (as indicated by TLC), the solution was cooled to 0°C and a solution of NaH (0.0860 g, 2.15 mmol) in MeOH (5.00 mL) was added dropwise. The cloudy mixture stirred at 0°C for 1 h and then at room temperature for 2 h. The mixture was acidified with 5% HCI and 142 renthe 13x 30 $3031 mmmd wodsh HRMS ForC. 91125 0874 0.640 and u by Tl Ol'gar Conce 98L 1 OH,‘} 3H). then the solvents were removed. The aqueous mixture was extracted with EtOAc (3 x 30.0 mL) and then the organics were combined, dried using anhydrous sodium sulfate and concentrated. The crude residue was purified by column chromatography (silica gel, 10% acetone; 90% DCM) affording the product as a whitish solid. Yield (0.0490 g, 47.0%). 1H NMR (500MHz), acetone: 5 1.09 (s, 3H), 1.14 (s, 3H), 4.99 (dd, J = 1.0, 17.3 Hz, 1H), 5.07 (dd, J = 1.2, 11.0 Hz, 1H), 6.02 (dd, J = 10.7, 17.3 Hz, 1H), 7.35 (m, 3H), 7.75 (m, 2H), 7.95 (s, 1H), 9.70 (s, 1H); 13C NMR (125MHz), acetone: 5 22.1, 22.8, 44.6, 72.7, 115.3, 128.1, 128.3, 128.6, 136.8, 142.9, 156.6, 175.2. IR: (KBr) 3243 cm“, 1772 cm", 1718 cm'1. HRMS: [M + H]+ = 245.1292, calculated for C14H17N202, 245.1290. Anal. Calcd. For C14H16N202: C, 68.83; H, 6.60; N, 11.47. Found: C, 68.67; H, 6.39; N, 11.26. Melting Point = 229-231’C. (II-94). To a flame dried 50 mL round bottom flask was added "-75 (0.150 g, 0.291 mmol) and anhydrous DCM (15.0 mL). Then anhydrous TEA (0.120 mL, 0.874 mmol) was added and the mixture was cooled to 0°C. EDCI (0.123 g, 0.640 mmol) was then added and the mixture stirred at 0°C under nitrogen for 1 h and then refluxed for 15 h. After the rearrangement was completed (as indicated by TLC), the reaction was quenched by addition of ice water (10.0 mL). The organic layer was separated, dried using anhydrous sodium sulfate and concentrated. The crude residue was purified by column chromatography (silica gel, 10% Ethyl acetate; 90% DCM; ARr = 0.69) affording the product as a thick oil. 1H NMR (500MHz), CDCI3: 5 1.38 (t, J = 7.2 Hz, 3H), 1.52 (s, 3H), 2.36 (s, 3H), 3.00 (d, J = 13.9 Hz, 1H), 3.04 (d, J = 13.8 Hz, 1H), 4.42 (dq, J = 2.8, 7.1 143 Hz. 2t ti‘fR Hz, 2H), 4.85 (s, 1H), 4.98 (m, 1H), 8.48 (s, 1H), 7.24-7.38 (m, 4H), 7.64 (s, 1H), 7.79 (d, J = 8.5 Hz, 2H), 7.91 (d, J = 8.9 Hz, 1H), 8.00 (d, J = 8.3 Hz, 1H); 130 NMR (125MHz), once: 8 13.9, 21.5, 23.8, 44.4, 83.2, 84.5, 113.7, 118.0, 119.3, 121.6, 123.7, 124.1, 125.3, 127.0, 127.1, 130.0, 134.7, 135.7, 138.2, 145.4, 147.5, 151.4, 189.4. IR: (KBr) 3340 cm", 1812 cm", 1770 cm“, 1728 cm". HRMS: [M + H]+ = 496.1548, calculated for C25H26N3068, 496.1542. 144 g) 11. 10. 11. References Matsumoto, K.; Rapoport, H., Preparation and properties of some acyl- guanidines. J. Org. Chem. 1968, 33, 552-8. Kenyon, G. L.; Rowley, G. L., Tautomeric preferences among glycocyamidines. J. Am. Chem. Soc. 1971, 93, 5552-60. Lempert, C., The chemistry of the glycocyamidines. Chem. Rev. 1959, 59, 667-736. Mukerjee, A. K.; Joseph, K.; Homami, S. S.; Tikdari, A. M., Triethylamine, ethanol-mediated disciplined reactions of S-benzylisothiouronium chloride with unsaturated 2-oxazolin-5-ones: synthesis of (Z)-2-amino-4- arylmethylene-2-imidazolin-5-ones, 5-benzoylamino-2-benzylthio-6-oxo- 4,4-spirocyclohexyl-1,4,5,6-tetrahydropyrimidine, and their structures. Heterocycles 1991, 32, 1317-25. Prager, R. H.; Tsopelas, C., Approaches to the synthesis of 5- benzylidene-2-imidazolin-4-ones. Aust. J. Chem. 1990, 43, 367-74. Tikdari, A. M.; Tripathy, P. K.; Mukerjee, A. K., Reactions of some 1,3- diamino nucleophiles with azlactones. J. Chem. Soc., Perkin Trans. 1 1988, 1659-62. Wegner, M. M.; Rapoport, H., Catalytic hydrogenation of some acylguanidines. J. Org. Chem. 1978, 43, 3840-4. Reddick, R. E.; Kenyon, G. L., Syntheses and NMR studies of specifically labeled [2-15N]phosphocreatine, [2-15N]creatinine, and related 15N- labeled compounds. J. Am. Chem. Soc. 1987, 109, 4380-7. Guella, G.; Mancini, I.; Zibrowius, H.; Pietra, F., Aplysinopsin-type alkaloids from Dendrophyllia sp., a scleractinian coral of the family Dendrophylliidae of the Philippines. Facile photochemical (Z/E) photoisomerization and thermal reversal. Helv. Chim. Acta 1989, 72, 1444-50. Hu, B.; Malamas, M.; Ellingboe, J.; Largis, E.; Han, S.; Mulvey, R.; Tillett, J., New oxadiazolidinedione derivatives as potent and selective human beta 3 agonists. Bioorg. Med. Chem. Lett. 2001, 11, 981-984. Pardasani, R. T.; Pardasani, P.; Jain, A.; Kohli, 8., Synthesis and semiempirical calculations of thiazolidinone and imidazolidinone derivatives of alpha -diones. Phos., Sulf. Sil. Relat. Elem. 2004, 179, 1569-1575. 145 19. 20. 21 12. 13. 14. 15. 16. 17. 18. 19. 20. 21. Ryabukhin, S. V.; Plaskon, A. S.; Volochnyuk, D. M.; Pipko, S. E.; Tolmachev, A. A., Chlorotrimethylsilane mediated synthesis of 5-(2- hydroxybenzoyl)pyrimidines from 3-formylchromones. Heterocycles 2008, 75, 583-597. Li, C.; Danishefsky, S. J., Studies directed toward the synthesis of the guanidine alkaloid, spiroleucettadine: some observations at the level of structure. Tetrahedron Lett. 2005, 47, 385-387. Chandrasekhar, S.; Karri, P., Aromaticity in azlactone anions and its significance for the Erlenmeyer synthesis. Tetrahedron Lett. 2006, 47, 5763-5766. Appleton, D. R.; Page, M. J.; Lambert, G.; Berridge, M. V.; Copp, B. R., Kottamides A-D: novel bioactive imidazolone-containing alkaloids from the New Zealand ascidian Pycnoclavella kottae. J. Org. Chem. 2002, 67, 5402-5404. Cafieri, F.; Gattorusso, E.; Mangoni, A.; TaglialateIa-Scafati, 0., Dispacamides, anti-histamine alkaloids from Caribbean Agelas sponges. Tetrahedron Lett. 1996, 37, 3587-3590. Chan, G. W.; Mong, S.; Hemling, M. E.; Freyer, A. J.; Offen, P. H.; DeBrosse, C. W.; Sarau, H. M.; Westley, J. W., New leukotriene B4 receptor antagonist: Ieucettamine A and related imidazole alkaloids from the marine sponge Leucetta microraphis. J. Nat. Prod. 1993, 56, 116-21. Cimino, G.; De Rosa, 8.; De Stefano, S.; Mazzarella, L.; Puliti, R.; Sodano, G., Isolation and x-ray crystal structure of a novel bromo compound from two marine sponges. Tetrahedron Lett. 1982, 23, 767-8. Davis, R. A.; Aalbersberg, W.; Meo, S.; Moreira da Rocha, R.; Ireland, C. M., The isolation and synthesis of polyandrocarpamines A and B. Two new 2-aminoimidazolone compounds from the Fijian ascidian, Polyandrocarpa sp. Tetrahedron 2002, 58, 3263-3269. Edrada, R. A.; Stessman, C. C.; Crews, P., Uniquely modified imidazole alkaloids from a calcareous Leucetta sponge. J. Nat. Prod. 2003, 66, 939- 942. Keenan, M.; Jones, K.; Hibbert, F., Highly efficient and flexible total synthesis of coelenterazine. Chem. Commun. (Cambridge) 1997, 323- 324. 146 25. 27. 28. 29. 30. 22. 23. 24. 25. 26. 27. 28. 29. 30. 31. Loukaci, A.; Guyot, M.; Chiaroni, A.; Riche, C., A new indole alkaloid from the marine tunicate Dendrodoa grossularia. J. Nat. Prod. 1998, 61 , 519- 522. Mancini, l.; Guella, G.; Debitus, C.; Dubet, D.; Pietra, F., lmidazolone and imidazolidinone artifacts of a pivotal imidazolthione, zyzzin, from the poecilosclerid sponge Zyzzya massalis from the Coral Sea. The first thermochromic systems of marine origin. Helv. Chim. Acta 1994, 77, 1886-94. Olofson, A.; Yakushijin, K.; Horne, D. A., Unusually Large 13C NMR Chemical Shift Differences between Neutral and Protonated Glycocyamidines. New Insights on Previously Reported Chemical Shift Assignments and Chemical Properties. J. Org. Chem. 1998, 63, 5787- 5790. Sato, H.; Tsuda, M.; Watanabe, K.; Kobayashi, J. i., Rhopaladins A-D, new indole alkaloids, from marine tunicate Rhopalaea sp. Tetrahedron 1998, 54, 8687-8690. Tsukamoto, S.; Kato, H.; Hirota, H.; Fusetani, N., Mauritiamine, a New Antifouling Oroidin Dimer from the Marine Sponge Agelas mauritiana. J. Nat. Prod. 1996, 59, 501-3. Vergne, C.; Appenzeller, J.; Ratinaud, C.; Martin, M.-T.; Debitus, C.; Zaparucha, A.; Al-Mourabit, A., Debromodispacamides B and D: Isolation from the Marine Sponge Agelas mauritiana and Stereoselective Synthesis Using a Biomimetic Proline Route. Org. Lett. 2008, 10, 493—496. Wang, H.; Lim, K. L.; Yeo, S. L.; Xu, X.; Sim, M. M.; Ting, A. B; Wang, Y.; Yee, 8.; Tan, Y. H.; Pallen, C. J., Isolation of a Novel Protein Tyrosine Phosphatase Inhibitor, 2-MethyI-Fervenulone, and Its Precursors from Streptomyces. J. Nat. Prod. 2000, 63, 1641-1646. Annoura, H.; Tatsuoka, T., Total syntheses of hymenialdisine and debromohymenialdisine: stereospecific construction of the 2-amino-4-oxo- 2-imidazolin-5(Z)-disubstituted ylidene ring system. Tetrahedron Lett. 1995, 36, 413-16. Lindel, T.; Hoffmann, H., Synthesis of dispacamide from the marine sponge Agelas dispar. Tetrahedron Lett. 1997, 38, 8935-8938. Crews, P.; Clark, D. P.; Tenney, K., Variation in the alkaloids among lndo- Pacific Leucetta sponges. J. Nat. Prod. 2003, 66, 177-182. 147 32. R 30. 39. 40. 41. 42. 43. 32. 33. 34. 35. 36. 37. 38. 39. 40. 41. 42. 43. Molina, P.; Almendros, P.; Fresneda, P. M., An iminophosphorane- mediated efficient synthesis of the alkaloid Ieucettamine B of marine origin. Tetrahedron Lett. 1994, 35, 2235-6. Fresneda, P. M.; Molina, P.; Sanz, M. A., The first synthesis of the bis(indole) marine alkaloid rhopaladin D. Synlett 2000, 1190-1192. Janosik, T.; Johnson, A.-L.; Bergman, J., Synthesis of the marine alkaloids Rhopaladins A, B, C and D. Tetrahedron 2002, 58, 2813-2819. Keifer, P. A.; Schwartz, R. E.; Koker, M. E. S.; Hughes, R. G., Jr.; Rittschof, D.; Rinehart, K. L., Bioactive bromopyrrole metabolites from the Caribbean sponge Agelas conifera. J. Org. Chem. 1991, 56, 2965-75. Kobayashi, J.; Tsuda, M.; Ohizumi, Y., A potent actomyosin ATPase activator from the Okinawan marine sponge Agelas cf. nemoechinata. Experientia 1991, 47, 301-4. O'Malley, D. R; Li, K.; Maue, M.; Zografos, A. L.; Baran, P. 8., Total Synthesis of Dimeric Pyrrole-lmidazole Alkaloids: Sceptrin, Ageliferin, Nagelamide E, Oxysceptrin, Nakamuric Acid, and the Axinellamine Carbon Skeleton. J. Am. Chem. Soc. 2007, 129, 4762-4775. Olofson, A.; Yakushijin, K.; Horne, D. A., Synthesis of Mauritiamine. J. Org. Chem. 1997, 62, 7918-7919. Koswatta, P. B.; Sivappa, R.; Dias, H. V. R.; Lovely, C. J., Total Synthesis of (+-)—Calcaridine A and (+-)-epi-Calcaridine A. Org. Lett. 2008, 10, 5055- 5058. Anzai, K., alpha -Diketone rearrangement. Reaction of 1,2- cyclohexanedione with arginine and with guanidine. Bull. Chem. Soc. Jap. 1969, 42, 3314-17. Atwood, J. L.; Barbour, L. J.; Heaven, M. W.; Raston, C. L., Synthesis of 2-imino-5-phenylimidazolidin-4-one and the structure of its trifluoroacetate salt. J. Chem. Crystallogr. 2003, 33, 175-179. Bengelsdorf, I. S., A reaction of guanidine with glyoxals in aqueous solution. The preparation of glycocyamidines. J. Am. Chem. Soc. 1953, 75, 3138-40. Bhat, B. A.; Dhar, K. L.; Puri, S. C.; Spiteller, M., A novel one-pot rearrangement reaction of 2,3-epoxydiaryl ketones: synthesis of (+-)-5,5- disubstituted imidazolones and 5,5-disubstituted hydantoins. Synlett 2006, 2723-2726. 148 48. 49. 50. 51. 52. 53. 54. 44. 45. 46. 47. 48. 49. 50. 51. 52. 53. 54. Chu, Y.; Lynch, V.; Iverson, B. L., Synthesis and DNA binding studies of bis-intercalators with a novel spiro-cyclic linker. Tetrahedron 2006, 62, 5536-5548. Ding, M.-W.; Tu, H.-Y.; Liu, Z.—J., Application of carbodiimide in heterocyclic synthesis: new facile synthesis of 2-aminoimidazolinone derivatives. Synth. Commun. 1997, 27, 3657-3662. Drewry, D. H.; Ghiron, C., Solid-phase synthesis of 2- aminoimidazolinones. Tetrahedron Lett. 2000, 41, 6989-6992. Evindar, G.; Batey, R. A., Peptide Heterocycle Conjugates: A Diverted Edman Degradation Protocol for the Synthesis of N-Terminal 2- lminohydantoins. Org. Lett. 2003, 5, 1201-1204. Frutos, R. P.; Johnson, M., Regiocontrolled synthesis of highly- functionalized fused imidazoles: a novel synthesis of second generation LFA-1 inhibitors. Tetrahedron Lett. 2003, 44, 6509-6511. Fu, M.; Fernandez, M.; Smith, M. L.; Flygare, J. A., Solid-Phase Synthesis of 2-Aminoimidazolones. Org. Lett. 1999, 1, 1351-1353. Gadwood, R. C.; Kamdar, B. V.; Dubray, L. A. C.; Wolfe, M. L.; Smith, M. P.; Watt, W.; Mizsak, S. A.; Groppi, V. E., Synthesis and biological activity of spirocyclic benzopyran imidazolone potassium channel openers. J. Med. Chem. 1993, 36, 1480-7. Garratt, P. J.; Thorn, S. N.; Wrigglesworth, R., Preparation of imidazolones from N-cyano-N'-methylcarboxyguanidines. An unusual carbon-nitrogen bond formation in the hydrogenolysis of a benzyl ester in an attempted synthesis of an inhibitor of carboxypeptidase A. Tetrahedron 1993, 49, 6885-98. Gavrilyuk, J. I.; Evindar, G.; Batey, R. A., Peptide-Heterocycle Hybrid Molecules: Solid-Phase Synthesis of a 400-Member Library of N-Terminal 2-lminohydantoin Peptides. J. Comb. Chem. 2006, 8, 237-246. Gillman, K. W.; Higgins, M. A.; Poindexter, G. S.; Browning, M.; Clarke, W. J.; Flowers, 8.; Grace, J. E.; Hogan, J. B.; McGovern, R. T.; lben, L. G.; Mattson, G. K.; Ortiz, A.; Rassnick, 8.; Russell, J. W.; AntaI-Zimanyi, |., Synthesis and evaluation of 5,5-diphenylimidazolones as potent human neuropeptide Y5 receptor antagonists. Bioorg. Med. Chem. 2006, 14, 5517-5526. Heras, M.; Ventura, M.; Linden, A.; Villalgordo, J. M., Reaction of a- iminomethylene amino esters with mono- and bidentate nucleophiles: a 149 55. 56. 57. 58. 59. 60. 62. 63. 64. 65. 86. 55. 56. 57. 58. 59. 60. 61. 62. 63. 64. 65. 66. straightforward route to 2-amino-1H-5-imidazolones. Tetrahedron 2001, 57, 4371—4388. Kang, J. H.; Moon, J. T.; Kim, J.; Joo, D. J.; Lee, J. Y., Synthesis of 2- arylsubstituted imidazolone derivatives. Bull. Korean Chem. Soc. 2007, 28, 913-914. Khodair, A. l.; Bertrand, P., A new approach to the synthesis of substituted 4-imidazolidinones as potential antiviral and antitumor agents. Tetrahedron 1998, 54, 4859-4872. Kiec-Kononowicz, K.; Zejc, A., Syntheses of some 2-amino-5,5- diphenylimidazolin-4-one derivatives. Pol. J. Chem. 1980, 54, 2217-24. Kwon, C. H.; Iqbal, M. T.; Wurpel, J. N. D., Synthesis and anticonvulsant activity of 2-iminohydantoins. J. Med. Chem. 1991, 34, 1845-9. Kwon, C. H.; Nagasawa, H. T., Facile synthesis of substituted 2- iminohydantoins. Synth. Commun. 1987, 17, 1677-82. Lange, U. E. W., Solid-phase synthesis of 2,3,5-trisubstituted 4H- imidazolones. Tetrahedron Lett. 2002, 43, 6857-6860. Mueller, H.; Carell, T., A carbocyclic analog of the oxidatively generated DNA lesion spiroiminodihydantoin. Eur. J. Org. Chem. 2007, 1438-1445. Nishimura, T.; Kitajima, K., Reaction of guanidines with alpha -diketones. Syntheses of 4,5-disubstituted-Z-aminoimidazoles and 2,6- unsymmetrically substituted imidazo[4,5-d]imidazoles. J. Org. Chem. 1979, 44, 818-24. Nishimura, T.; Nakano, K.; Shibamoto, S.; Kitajima, K., Novel synthesis of 2-(disubstituted amino)-5(4)-phenylimidazoles. J. Heterocycl. Chem. 1975, 12, 471-6. Sharma, V.; Lansdell, T. A.; Jin, G.; Tepe, J. J., Inhibition of Cytokine Production by Hymenialdisine Derivatives. J. Med. Chem. 2004, 47, 3700- 3703. Sharma, V.; Tepe, J. J., Potent inhibition of checkpoint kinase activity by a hymenialdisine-derived indoloazepine. Bioorg. Med. Chem. Lett. 2004, 14, 4319-4321. Simig, G.; Lempert, K.; Tamas, J.; Czira, G., Hydantoins, thiohydantoins, and glycocyamidines. 41. Reaction of N-cyano amines with 1-(tert-butyl)- 3,3-diphenylaziridinone. General method for the synthesis of 1-alkyl-, 1- 150 69. 71. 72. 73. 74. 75. 76. 77. 67. 68. 69. 70. 71. 72. 73. 74. 75. 76. 77. aralkyI-, and 1-aryl-5,5-diphenyl hydantoins and -g|ycocyamidines. Tetrahedron 1975, 31, 1195-200. Slade, P. G.; Priestley, N. D.; Sugden, K. D., Spiroiminodihydantoin as an Oxo—Atom Transfer Product of 8-Oxo-2'-deoxyguanosine Oxidation by Chromium(V). Org. Lett. 2007, 9, 4411-4414. Stover, J. S.; Ciobanu, M.; Cliffel, D. E.; Rizzo, C. J., Chemical and Electrochemical Oxidation of C8-Arylamine Adducts of 2'- Deoxyguanosine. J. Am. Chem. Soc. 2007, 129, 2074-2081. Takeuchi, H.; Hagiwara, S.; Eguchi, S., A new efficient synthesis of imidazolinones and quinazolinone by intramolecular aza-Wittig reaction. Tetrahedron 1989, 45, 6375-86. Talaty, E. R.; Gomez, J. A.; Dillon, J. A.; Palomino, E.; Agho, M. 0.; Batt, B. C.; Gleason, K. J.; Park, S.; Hernandez, J. R., Reaction of aziridinones with thiourea: a novel synthesis of specifically substituted glycocyamidines and hydantoins. Synth. Commun. 1987, 17, 1063-70. Varga, L.; Nagy, T.; Kovesdi, |.; Benet-Buchholz, J.; Dorman, G.; Urge, L.; Darvas, F., Solution-phase parallel synthesis of 4,6-diaryI-pyrimidine-2- ylamines and 2-amino-5,5-disubstituted-3,5-dihydro-imidazol-4-ones via a rearrangement. Tetrahedron 2003, 59, 655-662. Ye, P.; Sargent, K.; Stewart, E.; Liu, J.-F.; Yohannes, D.; Yu, L., Novel and Expeditious Microwave-Assisted Three-Component Reactions for the Synthesis of Spiroimidazolin-4-ones. J. Org. Chem. 2006, 71, 3137-3140. Ye, W.; Sangaiah, R.; Degen, D. E.; Gold, A.; Jayaraj, K.; Koshlap, K. M.; Boysen, G.; Williams, J.; Tomer, K. B.; Ball, L. M., A 2-Iminohydantoin from the Oxidation of Guanine. Chem. Res. Toxicol. 2006, 19, 506-510. Lovely, C. J.; Du, H.; He, Y.; Dias, H. V. R., Oxidative rearrangement of imidazoles with dimethyldioxirane. Org. Lett. 2004, 6, 735-738. Nishimura, T.; Toku, H.; Ueno, T.; Shibamoto, 8.; Imai, T., Mechanism of Voges-Proskauer reaction. Chem. Lett. 1972, 649-52. Toi, K.; Bynum, F.; Norris, E.; ltano, H. A., J. Biol. Chem. 1967, 242, 1036. Bach, R. D.; Domagala, J. M., The effect of Lewis acid and solvent on concerted 1,2-acyl migration. J. Org. Chem. 1984, 49, 4181-8. 151 .78. 80. 82. 83. 85. 86. 87. 88. 89. 78. 79. 80. 81. 82. 83. 84. 85. 86. 87. 88. 89. Chang, C.-L.; Kumar, M. P.; Liu, R.-S., A Highly Efficient Ruthenium- Catalyzed Rearrangement of alpha ,beta -Epoxyketones to 1,2-Diketones. J. Org. Chem. 2004, 69, 2793-2796. House, H. O.; Ryerson, G. D., The rearrangement of alpha ,beta -epoxy ketones. VIII. Effect of substituents on the rate of rearrangement. J. Am. Chem. Soc. 1961, 83, 979-83. House, H. 0.; Wasson, R. L., Rearrangement of alpha ,beta - epoxyketones. V. Rearrangements resulting in ring contraction. J. Am. Chem. Soc. 1957, 79, 1488-92. Rao, T. B.; Rao, J. M., Silica gel catalyzed rearrangement of alpha -epoxy ketones to 1,2-diketones. Synth. Commun. 1993, 23, 1527-33. Bajaj, K.; Srivastava, V. K.; Lata, S.; Chandra, R.; Kumar, A., Synthesis of some new benzothiazepinyl- and benzoxazepinyl indoles as antipsychotic agents. Indian J. Chem, Sect. B: Org. Chem. Incl. Med. Chem. 2003, 423, 1723-1728. Martinez, R.; Ramon, D. J.; Yus, M., Easy alpha -a|kylation of ketones with alcohols through a hydrogen autotransfer process catalyzed by RuCI2(DMSO)4. Tetrahedron 2006, 62, 8988-9001. Engel, N.; Kubel, B.; Steglich, W., C-C Linkage of Carboxylic Acids with Allyl Alcohols Using 2-Phenylglycine as Vehicle. Angew. Chem. Int. Ed. 1977, 16, 394-396. Kubel, B.; Hofle, G.; Steglich, W., Hetero Cope Rearrangements in the Cyclization of Allyl and Propargyl Esters of N-Acyl Amino Acids to Oxazolin-5-ones. Angew. Chem. Int. Ed. 1975, 14, 58-59. Firestone, R. A.; Harris, E. E.; Reuter, W., Synthesis of pyridoxine by Diels-Alder reactions with 4-methyl-5-alkoxyoxazoles. Tetrahedron 1967, 23, 943-55. Maeda, |.; Takehara, M.; Togo, K.; Asai, S.; Yoshida, R., Synthetic intermediate of pyridoxine. l. A novel synthesis of 5-alkoxy-2-carboxy-4- methyloxazole. Bull. Chem. Soc. Jap. 1969, 42, 1435-7. Fischer, J.; Kilpert, G; Klein, U.; Steglich, W., Stereochemistry of [3.3]- sigmatropic rearrangements in the oxazole series. Tetrahedron 1986, 42, 2063-74. Burger, K.; Geith, K.; Gaa, K., A Simple Access to 2-Substituted 3,3,3- Trifluoroalanine Derivatives. Angew. Chem. Int. Ed. 1988, 27, 848-849. 152 90. Burger, K.; Hennig, L.; Tsouker, P.; Spengler, J.; Albericio, F.; Koksch, B., Domino reactions with fluorinated five-membered heterocycles. alpha - Trifluoromethyl alpha -amino acids with unsaturated side-chains. Amino Acids 2006, 31, 427-433. 91. Burger, K.; Hennig, L.; Tsouker, P.; Spengler, J.; Albericio, F.; Koksch, 8., Synthesis of alpha -trifluoromethyl alpha -amino acids with aromatic, heteroaromatic and ferrocenyl subunits in the side chain. Amino Acids 2006, 31 , 55-62. 92. Castelhano, A. L.; Home, 8.; Taylor, G. J.; Billedeau, R.; Krantz, A., Synthesis of alpha-amino acids with beta, gamma-unsaturated side chains. Tetrahedron 1988, 44, 5451-5466. 93. Castelhano, A. L.; Pliura, D. H.; Taylor, G. J.; Hsieh, K. C.; Krantz, A., Allenic suicide substrates. New inhibitors of vitamin B6 linked decarboxylases. J. Am. Chem. Soc. 1984, 106, 2734-5. 94. Colombo, L.; Casiraghi, G.; Pittalis, A., Stereoselective Synthesis of C- Glycosyl alpha-Amino Acids. J. Org. Chem. 1991, 56, 3897-3900. 95. Colombo, L.; di Giacomo, M.; Ciceri, P., Practical stereoselective synthesis of alpha -D-C-mannosyl-(R)-alanine. Tetrahedron 2002, 58, 9381-9386. 96. Holladay, M. W.; Nadzan, A. M., Synthesis of alpha-Benzyl gamma- Lactam, alpha-Benzyl delta-Lactam, and alphaBenzylproline Derivatives as Conformationally Restricted Analogues of Phenylalaninamide. J. Org. Chem. 1991, 56, 3900-3905. 97. Wittmann, U.; Tranel, F.; Froehlich, R.; Haufe, 6., Novel synthesis of 4- fluoropyridines based on 2-fluoroallylic alcohols by succeeding Ireland- Claisen and aza-Cope rearrangements as key steps. Synthesis 2006, 2085-2096. 98. Fisk, J. S.; Mosey, R. A.; Tepe, J. J., The diverse chemistry of oxazol-5- (4H)-ones. Chem. Soc. Rev. 2007, 36, 1432-1440. 99. Wan, Z.-K.; Wacharasindhu, S.; Binnun, E.; Mansour, T., An efficient direct amination of cyclic amides and cyclic ureas. Org. Lett. 2006, 8, 2425-2428. 100. Bartlett, P. A.; Tanzella, D. J.; Barstow, J. F., Ester-Enolate Claisen Rearrangement of Lactic Acid Derivatives. J. Org. Chem. 1982, 47. 153 101. 102. 103. 104. 105. 106. 107. 108. 109. 110. 111. 112. Overman, L. E.; Rogers, B. N.; Tellew, J. E.; Trenkle, W. C., Stereocontrolled Synthesis of the Tetracyclic Core of the Bisguanidine Alkaloids Palau'amine and Styloguanidine. J. Am. Chem. Soc. 1997, 119, 7159-7160. Hupp, C. D.; Tepe, J. J., 1-EthyI-3-(3-dimethylaminopropyl)carbodiimide Hydrochloride-Mediated Oxazole Rearrangement: Gaining Access to a Unique Marine Alkaloid Scaffold. J. Org. Chem. 2009, 74, 3406-3413. Kent, D. R.; Cody, W. L.; Doherty, A. M., Two new reagents for the guanylation of primary, secondary and aryl amines. Tetrahedron Lett. 1996, 37, 8711-8714. Kim, K. S.; Qian, L., Improved Method for the Preparation of Guanidines. Tetrahedron Left. 1993, 34, 7677-80. Linton, B. R.; Carr, A. J.; Orner, B. P.; Hamilton, A. D., A Versatile One- Pot Synthesis of 1,3-Substituted Guanidines from Carbamoyl Isothiocyanates. J. Org. Chem. 2000, 65, 1566-1568. Ramadas, K.; Srinivasan, N., An expedient synthesis of substituted guanidines. Tetrahedron Lett. 1995, 36, 2841-4. Shibanuma, T.; Shiono, M.; Mukaiyama, T., A convenient method for the preparation of carbodiimides using 2-chloropyridinium salt. Chem. Lett. 1977, 575-6. Levallet, C.; Lerpiniere, J.; Ko, S. Y., The HgCI2-promoted guanylation reaction: the scope and limitations. Tetrahedron 1997, 53, 5291-5304. Yu, Y.; Ostresh, J. M; Houghten, R. A., Solid-Phase Synthesis of 1,5- Disubstituted 2-Aryliminoimidazolidines. J. Org. Chem. 2002, 67, 3138- 3141. Yong, Y. F.; Kowalski, J. A.; Lipton, M. A., Facile and Efficient Guanylation of Amines Using Thioureas and Mukaiyama's Reagent. J. Org. Chem. 1997, 62, 1540-1542. Manimala, J. C.; Anslyn, E. V., A highly efficient method for the synthesis of guanidinium derivatives. Tetrahedron Lett. 2002, 43, 565-567. Alvarado, C.; Diaz, E.; Guzman, A., Total synthesis of ulongamide A, a cyclic depsipeptide isolated from marine cyanobacteria Lyngbya sp. Tetrahedron Lett. 2007, 48, 603-607. 154 118. 119. 120. 1.22. 123. I24. 113. 114. 115. 116. 117. 118. 119. 120. 121. 122. 123. 124. Kojima, S.; Chabayashi, T.; Umeda, Y.; lwamoto, A.; Tanabe, K.; Ohkata, K., Stereoselective synthesis of a novel chiral piperazine. Heterocycles 2008, 75, 1493-1501. Shimizu, M.; Kume, K.; Fujisawa, T., Switchover of the diastereoselectivity induced by a simple selection of titanium(lV) halides in the addition reaction of silyl ketene acetals to a chiral imine. Tetrahedron Left. 1995, 36, 5227-30. Oertqvist, P.; Peterson, S. D.; Aakerblom, E.; Gossas, T.; Sabnis, Y. A.; Fransson, R.; Lindeberg, G.; Danielson, U. H.; Karlen, A.; Sandstroem, A., Phenylglycine as a novel P2 scaffold in hepatitis C virus NS3 protease inhibitors. Bioorg. Med. Chem. 2007, 15, 1448-1474. Yan, S.; Larson, 6.; Wu, J. 2.; Appleby, T.; Ding, Y.; Hamatake, R.; Hong, 2.; Yao, N., Novel thiazolones as HCV NSSB polymerase allosteric inhibitors: Further designs, SAR, and X-ray complex structure. Bioorg. Med. Chem. Left. 2007, 17, 63-67. Barberis, C.; Voyer, N.; Roby, J.; Chenard, S.; Tremblay, M.; Labrie, P., Rapid access to N-Boc phenylglycine derivatives via benzylic lithiation reactions. Tetrahedron 2001, 57, 2965—2972. Garg, N. K.; Sarpong, R.; Stoltz, B. M., The First Total Synthesis of Dragmacidin D. J. Am. Chem. Soc. 2002, 124, 13179-13184. Kharasch, M. 8.; Kane, S. S.; Brown, H. 0., Presence of indole in \"practical\" a-methylnaphthalene. J. Am. Chem. Soc. 1940, 62, 2242-3. Nishizawa, M.; Yamamoto, H.; Seo, K.; Imagawa, H.; Sugihara, T., TMS Triflate-Catalyzed Cleavage of Prenyl (3-Methylbut-2-enyl) Esters. Org. Lett. 2002, 4, 1947-1949. Geistlinger, T. R.; Guy, R. K., Novel Selective Inhibitors of the Interaction of Individual Nuclear Hormone Receptors with a Mutually Shared Steroid Receptor Coactivator 2. J. Am. Chem. Soc. 2003, 125, 6852-6853. Katritzky, A. R.; Rogovoy, B. V., Recent developments in guanylating agents. ARKIVOC (Gainesville, FL, United States) 2005, 49-87. Martin, N. I.; Woodward, J. J.; Marletta, M. A., NG-Hydroxyguanidines from Primary Amines. Org. Left. 2006, 8, 4035-4038. Rani, P.; Srivastava, V. K.; Kumar, A., Synthesis and anti-inflammatory activity of heterocyclic indole derivatives. Eur. J. Med. Chem. 2004, 39, 449-452. 155 125. Frigerio, M.; Santagostino, M.; Sputore, S., A user-friendly entry to 2- iodoxybenzoic acid (IBX). J. Org. Chem. 1999, 64, 4537-4538. 126. Yang, L.; Deng, G.; Wang, D.-X.; Huang, Z.-T.; Zhu, J.-P.; Wang, M.-X., Highly Efficient and Stereoselective N-Vinylation of Oxiranecarboxamides and Unprecedented 8-endo-Epoxy-arene Cyclization: Expedient and Biomimetic Synthesis of Some Clausena Alkaloids. Org. Lett. 2007, 9, 1387-1390. 127. Kim, D. D.; Lee, S. J.; Beak, P., Asymmetric Lithiation-Substitution Sequences of Substituted Allylamines. J. Org. Chem. 2005, 70, 5376- 5386. 156 CHAPTER III TOTAL SYNTHESIS OF AN INDOLE ALKALOID FROM THE TUNICATE DENDRODOA GROSSULARIA AND TWO ANALOGS III.A Pharmacological value of marine compounds The biodiversity found in the marine environment offers a wealth of organisms each containing unique metabolites and secondary metabolites with chemically diverse structures.“ 2 Efforts to explore this rich and diverse set of compounds have led to the opening of a “marine pipeline,” which allows for the flow of these compounds into the pharmaceutical arena. The pharmaceutical industry has benefited from the study of these organisms and the introduction of the marine natural products they contain in many areas of research. Compounds coming from marine organisms have shown to possess biological activity for diseases such as cancer, arthritis, AIDS and have potential as anti-viral, anti- microbial, anti-parasitic, analgesic and anti-inflammatory agentsf"5 The extraordinary biological properties that compounds from natural products possess justify the isolation and harvesting of the sources for these compounds. Unfortunately, these secondary metablites found in the marine organisms are usually present in trace amounts making acquisition of an appropriate amount of these compounds for testing very difficult. In order to determine the biological potential of a particular compound, there are two options commonly used: 1) massive recollection of the organism and reisolation of the molecule or 2) development of a concise synthetic route to the molecule.6 Factors concerning the availablility of the marine organism and ease of isolation 157 will determine which method will access the desired compound the easiest and fastest. III.B Background on Dendrodoa grossularia Invertebrate marine animals such as tunicates, which include sea squirts and sea cucumbers, have been a prosperous source of structurally unique and biologically interesting secondary metabolites. In particular, these marine organisms are known to contain indole alkaloids, nitrogen containing compounds that also posses an indole structural moiety.7 Furthermore, the novel frameworks that these indole alkaloids possess often attracts the attention of chemists interested in developing synthetic routes to the scaffolds. The tunicate Dendrodoa grossularia (baked bean sea squirt), a red marine organism that grows along the coasts of Brittany and in the Baltic and North Seas, contains some heterocyclic alkaloids with unique scaffolds (Figure III-1).8'13 \ ‘N/ 0 N / N y \ N O N \ k S’N o \ \ \ N N H H H Dendrodoine . 3-IndonI-4H-imidazoI-4-one “bomon \N/ N2\NH __‘_ O / O \ N R N H R = Indole (Grossularine 1) Indole Alkaloid R = pOH-phenyl (Grossularine 2) III-1 Figure III-1. Alkaloids from Dendrodoa grossularia 158 Dendrodoine was isolated by Heitz and coworkers in 198010 and first synthesized in 198414 and is a unique molecule that exhibited the first example of a divalent sulfur-nitrogen bond found in a natural product, coming from its 1, 2, 4- thiadiazole heterocyclic nucleus.12 Results from a brief biological study on the compound revealed moderate cytotoxicity for L1210 leukemia cells with an ID50 of 10 pg/mL.13 An additional report suggests that dendrodoine inhibits the synthesis of DNA in leukemia cells through preventing the incoporation of thymidine into DNA.15 3-lndonI-4H-imidazoI-4-one was isolated and then synthesized in 1986 by Guyot and coworkers.9 The compound was found to be light sensitive in solution which resulted in the rearrangment of the molecule to yield additional byproducts.9 Furthermore, a cytotoxicity assay revealed that the compound was void of activity.13 Alboinon isolated in 1997 by Steffan and coworkers contains a rare 1, 3, 5-oxadiazin-2-one scaffold and represents the first natural product bearing this moiety.8 The synthesis of alboinon was attempted by the same group using a protecting group on the indolic nitrogen, however, all attempts to remove the protecting group resulted in the attack on the oxadiazinone ring. Fortunately, it was found that oxidation of 3-lndonI-4H-imidazol-4-one with m- chloroperbenzoic acid produced alboinon in a good yield.8 This result offers insight into a potential biosynthetic route to alboinon suggesting that perhaps it is synthesized in the tunicate by oxidation of 3-IndolyI-4H-imidazoI-4-one. Grossularines 1 and 2 were isolated in 1989 and were originally given an alternative structure. X-ray studies and the availability of larger amounts of the 159 compounds allowed the definite determination of the physical structure.“ 13' 16 Furthermore, these compounds represent the first natural products to contain an o-carboline skeleton.13 The first total synthesis of both grossularine 1 and 2 was achieved in 1995 using a thermal electrocyclic reaction to obtain the heterocyclic core of both compounds.17 Grossularines 1 and 2 were found to be cytotoxic toward L1210 leukemia cells displaying ID50 values of 6 and 4 pg/mL, respectively. A closer investigation revealed that these compounds caused accumulation of these cells in the G1 phase at concentrations of 10 ug/mL (grossularine 1) and 1.5 pg/mL (grossularine 2).13 The mode of action for grossularine 2’s cytotoxicity was examined and it was found that grossularine 2 intercalates into the DNA and also inhibits the incorporation of thymidine into DNA. Grossularine 1 was found to have a somewhat more ambiguous mode of action by giving results reminiscent of drugs that show intercalation as well as alkylation.15 Furthermore, they were found even more cytotoxic toward the solid tumor cell lines WiDr (colon) and MCF7 (breast) showing activity in the 10 ng/mL range.13 The latest indole alkaloid (III-1) to come from Dendrodoa grossularia was isolated in 1998 by Guyot and coworkers and contains a unique quaternary imidazolone core.11 The structure was determined by 1D and 2D NMR techniques as well as by information given by the X-ray structure of a derivative of the indole alkaloid (Ill-4), obtained by treating the natural product with acetic anhydride and pyridine (Scheme III-1). After an initial acetylation of the indolic nitrogen and nitrogen contained within the imidazolone ring of III-1 affords III-2, it 160 is proposed that an aldol reaction occurs to give III-3. The derivative III-4 is finally formed after a Bayliss-Hillman reaction occurs between III-3 and acetic anhydride (Scheme Ill-1).11 Scheme III-1. Synthesis of derivative III-4 O O /U\O/U\ pyridine Indole Alkaloid Derivative III-4 Acetylation Bayliss-Hillman reaction III-2 III-3 The crystal structure of III-4 was ineffective at identifying the absolute stereochemistry at the only stereogenic center in the natural product due to the racemic nature of the crystal. It is suggested that the optical rotation observed for the indole alkaloid III-1 and derivative III-4 ([010 = -15 and -12, respectively) is due to the presence of an excess of one enantiomer. Furthermore, indole alkaloid III-1 could be considered to be derived from 3-Indolyl-4H—imidazoI-4-one another alkaloid from the same tunicate.11 As a result of the known biological properties of other alkaloids from Dendrodoa grossularia and the physical 161 similarities the new indole alkaloid has with the Chk2 inhibitor, indoloazepine, a total synthesis for the indole alkaloid was developed. III.C Retrosynthetic analysis of indole alkaloid III-1 As a continuation of our laboratory’s focus on the development of new heterocyclic methodologies for the syntheses of pharmacologically significant scaffolds,18 the racemic total synthesis of indole alkaloid Ill-1 was completed along with two additional analogs. The key step in forming the quaternary stereocenter in the alkaloid and analogs utilizes a novel oxazole rearrangement19 (discussed in Chapter II) producing an oxazolone intermediate, ultimately leading to a quaternary hydantoin. Scheme III-2 illustrates our retrosynthetic strategy for the total synthesis of indole alkaloid III-1. It was envisioned that the imidazolone moiety of the natural product could be accessed from the hydantoin intermediate III-10.2“ 2‘ Through a newly developed EDCI-mediated oxazole rearrangement, hydantoin III-10 was thought to be formed from thiourea III-919' 22 In turn, thiourea III-9 could be obtained from keto allyl ester III-5 after a few functional group manipulations. 162 Scheme III-2. Retrosynthesis of indole alkaloid III-1 Oxazole Rearrangement o H 0 DY .- OYNH - / \ OEt /N n III-5 Ts III-9 III.D Synthesis of indole alklaloid III-1 Keto allyl ester III-5 (Scheme III-3) was synthesized in excellent yields (94%) through an esterification of 2-(1H-indol-3-yI)-2-oxoacetyl chloride, which is the product of a known reaction between indole and oxalyl chloride,23' 24 with 2- methyl-Z-propen-1-ol at room temperature for 16 h. Subsequent protection of the indolic nitrogen with p-toluene sulfonyl chloride under basic conditions for 16 h gave keto ester III-6 in a 77% yield, which when treated with hydroxylamine and pyridine in dioxane, produced oxime III-7 in a 96% yield as a mixture of E and Z isomers. Each isomer was isolated and both were carried on in the synthesis by reducing the oximes using zinc powder and aqueous acetic acid at 0°C for 2 h to amine III-8 in a 84% yield. Thiourea III-9 was formed in a 87% yield after treating amine III-8 with commercially available ethoxy carbonyl isothiocyanate for 16 h at room temperature (Scheme III-3).22 163 Scheme III-3. Synthesis of thiourea III-9 O O O O HO Cl /\H/ o/\H/ \ EtOAc, rt, 18 h, \ N 94% N III-5 H H TsCl, DMAP, DIPEA, DCM rt, 16 h, 77% OH 2 o O O N \ O/T A NHZOH-HCI (VT \ Dioxane/HZO, \ N III-7 Pyr, reflux N III-6 . 16 h, 96% Ts Ts Zn, AcOH 0 °C, 2 h, 84% H N o s H 0 2 0 N CY A 0 Y O/\"/ \ EtO NCS = NH / O N Ill-8 DCM. rt, 16 h, 87 /o OEt /N Ts T5 Ill-9 Subsequent treatment of thiourea III-9 with EDCI followed by sodium methoxide yielded hydantoin III-10 in a 71% yield, resulting from the EDCI- mediated oxazole rearrangement (Scheme III-4). The abbreviated proposed mechanism for the formation of hydantoin III-10 is also highlighted in Scheme III- 4. After the thiourea moiety was converted into a carbodiimide intermediate using EDCI, a 5-exo-dig cyclization occured between the carbonyl oxygen of the ester and the carbodiimide to form a transient oxazole intermediate.22 164 Scheme III-4. Synthesis of hydantoin III-10 H H 0 OT” SYN O/\H/ 1.EDCI, TEA, DCM, HN O 0°C to reflux,9h 0 NH s Y / 2. NaOMe, MeOH, \ OEt /N rt,4h,71% Ts N III-9 TS III-10 S R’ 1’0 G \ N. , TS TS —- oxazolone oxazole intermediate— Through a Claisen-type rearrangement, the oxazole was transformed into a quaternary oxazolone intermediate. Upon completion of the rearrangement, as indicated by TLC, a solution of sodium methoxide in methanol was added to complete the transformation from the oxazolone to hydantoin III-1t).22 Treatment of hydantoin III-10 with Lawesson’s reagent25 for 24 h under refluxing conditions produced thiohydantoin III-11 (Scheme Ill-5) in a 82% yield, which was selectively methylated at the thiocarbonyl using methyl iodide, DMAP, and diisopropylethylamine at room temperature for 2 h to furnish imidazolone Ill-12 in a 83% yield. After heating imidazolone III-12 with a THF solution of dimethylamine in a sealed tube at 75 °C for 14 h, imidazolone III-13 was produced in a 94% yield.22 165 Scheme III-5. Synthesis of alkaloid III-1 H H 08/“ Sm“ O L . O HN awesson S HN Rgt \ toluene, reflux, \ 24 h, 82% "l‘ III-10 "l' III-11 Ts Ts Mel, DMAP, DIPEA, DCM f1, 2 h, 83% \ NH / in THF sealed tube N 75 °C, 14 h, 94% \ . Ts II|13 KOEt, EtOH reflux, 24 h, 86% 1. 0804, NMO, rt, 4 h 2. NalO4, 0 °C, 2 h, 62% N H III-14 ""1 The final two steps for the total synthesis of III-1 began with the deprotection of the indole nitrogen of III-13 using potassium ethoxide and ethanol under refluxing conditions to produce imidazolone III-14 in an 86% yield. Finally, oxidation of the terminal alkene of III-14 was achieved through a two-step/one- pot modified Johnson-Lemieux26 reaction to give indole alkaloid Ill-1 in a 62% yield (Scheme Ill-5). A tabulated spectral comparison of the synthesized alkaloid (III-1) and the isolated natural product is given below in Table III-1. Fortunately, a 166 suitable crystal of III-1 was able to be formed and allowed 3 X-ray crystal structure of III-1 to be determined (Figure III-2), which also confirms the structure of the natural produc t22 Table III-1. Comparison of spectral data from synthesized natural product (III-1) and isolated natural product Spectrum Indole Alkaloid (III-1) Isolated Natural Product A5 2.18 (s, 3H) 2.18 (s, 3H) 0.00 3.05 (s, 3H) 3.04 (s, 3H) 0.01 3.23 (s, 3H) 3.22 (s, 3H) 0.01 3.48 (dd, J =18, 37 Hz, 2H) 3.45 (dd, J =16, 8 Hz, 2H) 0.01 1H NMR 8.97 (t, J = 7 Hz, 1H) 6.9 (t, J = 8 Hz, 1H) 0.07 (ppm in 7.09 (t, J = 7 Hz, 1H) 7.1 (t, J = 8 Hz, 1H) 0.01 00300) 7.28 (s, 1H) 7.28 (s, 1H) 0.00 7.33 (d, J = 8 Hz, 1H) 7.33 (d, J = 8 Hz, 1H) 0.00 7.45 (d, J = 8 Hz, 1H) 7.48 (d, J = 8 Hz, 1H) 0.01 8.24 (s, 1H, DMSO-d5) 8.2 (s, 1H, DMSO-d5) 0.04 11.01 (s, 1H, DMSO-d6) 11.01 (s, 1H, DMSO-d6) 0.00 31.0 31 0.0 38.9 37 0.1 38.8 38.8 0.0 49.0 48 1.0 87.0 87 0.0 112.8 112.6 0.0 114.5 114.8 0.1 13.31: :33: 133-3 9.3 CD300) 122.7 122.7 0.0 124.1 124.1 0.0 125.8 125.8 0.0 138.8 138.6 0.0 170.8 170.8 0.0 191.7 191.7 0.0 207.3 207.3 0.0 IR (cm") 3234, 1720, 1703, 1811 3200, 1710, 1840 NA HRMS [Mm]. 299.1508 299.1483 NA 167 Figure III-2. X-ray crystal structure of indole alkaloid III-1 III.E Retrosynthetic analysis of analogs 1 and 2 The versatility of the synthetic pathway developed for the natural product was illustrated by the synthesis of 2 analogs that required no major modification of the original synthetic route. Hypothetically, a number of analogs could be developed with the synthetic route as a result of the diversity allowed in the 168 rearrangement and the variety of amines that could be used to create the final imidazolone. Our retrosynthetic strategy for the total synthesis of analog 1 and analog 2 is illustrated in Scheme III-6. It was envisioned that the imidazolone moiety of the analog 2 (Ill-19) could be accessed from the hydantoin intermediate Ill-10 utilizing classical chemical modifications.” 21 Hydantoin III-1O could be formed from thiourea III-9 using the protocol developed for the EDCI-mediated rearrangement. Analog 1 (Ill-16) was also envisioned to be formed from thiourea III-9 by slightly modifying the rearrangement to include a removal of the tosyl protecting group. Scheme III-6. Retrosynthesis of analog 1 (III-16) and analog 2 (III-19) N H Ill-19 III.F Synthesis of analogs 1 and 2 The first analog, hydantoin Ill-16 shown in Scheme III-7, was synthesized using the same thiourea (III-9) used to produce the natural product. Instead of carefully monitoring the second step of the rearrangement to avoid removal of 169 the tosyl protecting group, a mixture of potassium ethoxide and ethanol was used and refluxed overnight to yield hydantoin III-15 in a 83% yield. Using the same procedure as the natural product synthesis, the terminal olefin on hydantoin III-15 was oxidized to the ketone in a 61% yield affording hydantoin III-16 (analog 1).19 Scheme III-7. Synthesis of analog 1 (III-16) H O SYN O/T 1.EDCI, TEA, DCM, O NH / 0 °C to reflux, 9 h : 2. KOEt, EtOH, OEt N reflux, 83% / Ts Ill-9 1. 0804, NMO, rt 2. NaIO4, 0°c, 81% The second analog shown in Scheme III-8, imidazolone III-19, took advantage of the synthetic handle that S-methylimidazolone III-12 provided due to its reactivity and tendency to be substituted with a nucleophile. In this instance, the S-methyl group was replaced using ammonium hydroxide to give imidazolone III-17 in a 72% yield. The deprotection method previously described with the natural product synthesis was also used to afford imidazolone III-18 in a 47% yield. Finally, the synthesis of analog 2 (Ill-19) was completed after the terminal alkene of III-18 was oxidized under previously determined conditions. Unfortunately, due to the difficult nature of isolating the product, only 19% of imidazolone Ill-19 was recovered although the overall conversion was higher.19 In general, the free amino imidazolones at every step were very hard to isolate 170 due to their tendency to stay in the aqueous phase during extractions, even after using n-butanol as an extracting solvent. Scheme III-8. Synthesis of analog 2 (III-19) NH4OH, THF, _ sealed tube, 90°C,72% KOEt, EtOH reflux, 47% 1. OsO4, NMO, rt ‘ 2. NalO4, 0°c, 19% 171 III.G General experimental information Reactions were carried out in flame-dried glassware under nitrogen atmosphere. All reactions were magnetically stirred and monitored by TLC with 0.25 pm pre-coated silica gel plates using either UV light or iodine to visualize the compounds. Column chromatography was carried out on Silica Gel 60 (230-400 mesh) supplied by EM Science. Yields refer to chromatographically and spectroscopically pure compounds unless otherwise noted. Infrared spectra were recorded on a Nicolet IR/42 spectrometer. 1H and 13C NMR spectra were recorded on a Varian Unity Plus-500 spectrometer or a Varian lnova-300, as noted in the experimental for each compound. Chemical shifts are reported relative to the residue peaks of the solvent (CDCI3: 7.24 ppm for 1H and 77.0 ppm for 13C) (Acetone-d5: 2.04 ppm for 1H and 29.8 ppm for 13C) (DMSO-d5: 2.49 ppm for 1H and 39.5 ppm for 13C). The following abbreviations are used to denote the multiplicities: s = singlet, d = doublet, dd = doublet of doublets, t = triplet, and m = multiplet. Low resolution mass spectra were recorded on a Hewlet—Packard 5890 Series II gas chromatograph connected to a TRIO-1 EI mass spectrometer. HRMS were obtained with a Micromass Q-ToF Ultima API LC-MS/MS mass spectrometer. Elemental analysis data were obtained on a Perkin Elmer 2400 Series II CHNS/O analyzer. Purity of compounds, whose elemental analyses were above the ACS tolerated 0.4% deviation, were confirmed by 1H NMR and 13C NMR. Melting points were obtained using an Electrothermal® capillary melting point apparatus and are uncorrected. Reagents and solvents were purchased from commercial suppliers and used 172 without further purification. Anhydrous methylene chloride and toluene were dispensed from a delivery system which passes the solvents through a column packed with dry neutral alumina. III.H Experimental procedures and characterization (III-5). To a flame dried 100 mL round bottom flask was added EtOAc (50.0 mL) and 2-(1H-indol-3-yI)-2-oxoacetyl chloride23' 2“ (2.00 g, 9.88 mmol). The yellow mixture was put under an N2 atmosphere. Then 2-methyI-2-propene- 1-ol (4.00 mL, 48.3 mmol) was added. Within minutes the yellow cloudy mixture became an orange clear solution. The solution stirred overnight at room temperature under N2. The EtOAc was evaporated by rotary evaporation and the solid was re-dissolved in EtOAc and washed with brine (2 x 30.0 mL). The aqueous layer was extracted with EtOAc (2 x 30.0 mL) and the organics were combined, dried using anhydrous sodium sulfate, and concentrated to give a the product as a dark reddish solid. Yield: (2.21 g, 94.0%). 1H NMR (300MHz), Acetone: 5 1.82 (s, 3H), 4.78, (s, 2H), 4.99 (s, 1H,), 5.10 (s, 1H), 7.26-7.33 (m, 2H), 7.55-7.59 (m, 1H), 8.30-8.33 (m, 1H), 8.43 (s, 1H), 11.33 (s, 1H); 13C NMR (75MHz), Acetone: 5 19.6, 69.0, 113.2, 113.7, 114.2, 122.5, 123.7, 124.8, 126.9, 137.8, 138.1, 140.7, 164.1, 179.6. IR: (NaCI) 1610 cm", 1740 cm'1, 3190 (broad) cm". M.S: calculated for C14H13N03 [M+] = 243 and found [M+] = 243.0; Anal. Calcd. For C14H13N03: C, 69.12; H, 5.39; N, 5.76. Found: C, 67.78; H, 5.32; N, 5.26. Melting Point = 122-124°C. (Ill-6). To a 500 mL flame dried round bottom flask was added III-5 (10.7 g, 44.0 mmol) and anhydrous DCM (0.250 L). Then TsCl (16.7 g, 88.1 mmol), 173 re W (I! In DMAP (13.4 g, 0.110 mol) and DIPEA (19.2 mL, 0.110 mol) were added and the mixture stirred at room temperature under nitrogen overnight. The solvent was removed and diethyl ether (0.200 L) was added to the residue and was washed with 1% HCI (2 x 80.0 mL) and brine (1 x 80.0 mL). The organics dried with anhydrous sodium sulfate and concentrated. The crude residue was purified by column chromatography (silica gel, 30% EtOAc; 70% Hexane) affording the product as a whitish solid. Yield (13.6 g, 77.0%). 1H NMR (500MHz), CDCI3: 5 1.84 (s, 3H), 2.34, (s, 3H), 4.78 (s, 2H), 5.03 (s, 1H), 5.11 (s, 1H), 7.27 (d, J = 8.0 Hz, 2H), 7.36 (m, 2H), 7.84 (d, J = 8.3 Hz, 2H), 7.94 (d, J = 7.3 Hz, 1H), 8.33 (d, J = 7.33 Hz, 1H), 8.80 (s, 1H); 13C NMR (125MHz), CDCI3: 5 19.5, 21.6, 69.4, 113.1, 114.5, 117.0, 122.9, 125.2, 126.1, 127.2, 127.6, 130.3, 134.2, 134.5, 136.7, 138.6, 146.1, 161.3, 178.4. IR: (NaCI) 1670 cm", 1732 cm". HRMS: [M + H]+ = 398.1075, calculated for 021H20N058, 398.1062. Anal. Calcd. For C21H19N058: C, 63.46; H, 4.82; N, 3.52. Found: C, 63.46; H, 5.00; N, 3.39. Melting Point = 86-88°C. (III-7). To a 250 mL round bottom flask was added III-6 (2.10 g, 5.29 mmol) and dioxane (0.100 L). Then hydroxylamine hydrochloride (1.09 g, 15.9 mmol) was added along with a little water (5.00 mL). Then pyridine (1.36 mL, 16.9 mmol) was added and the mixture refluxed overnight under nitrogen. The solvents were removed and diethyl ether (0.100 L) was added to the residue and was washed with brine (1 x 50.0 mL). The organics were dried with anhydrous sodium sulfate and concentrated. The crude residue was purified by column chromatography (silica gel, 30% EtOAc; 70% Hexane) affording the product as a 174 thick clear oil. Yield (2.10 g, 96.0%). Isomer A: 1H NMR (500MHz), CDCI3: 5 1.83 (s, 3H), 2.32 (s, 3H), 4.82 (s, 2H), 5.02 (s, 1H), 5.15 (s, 1H), 7.14 (d, J = 8.5 Hz, 2H), 7.17 (t, J = 8.0 Hz, 1H), 7.32 (t, J = 8.0 Hz, 1H), 7.73 (d, J = 8.5 Hz, 2H), 7.77 (s, 1H), 7.96 (d, J = 8.0 Hz, 1H), 8.01 (d, J = 8.0 Hz, 1H), 9.14 (s, 1H); 13C NMR (125MHz), CDCI3: 5 19.4, 21.3, 69.1, 113.1, 113.7, 114.5, 123.0, 124.1, 125.6, 126.8, 126.9, 127.2, 129.9, 134.4, 135.0, 138.7, 145.4, 146.5, 162.5. Isomer B: 1H NMR (500MHz), CDCI3: 5 1.78 (s, 3H), 2.33 (s, 3H), 4.75 (s, 2H), 4.97 (s, 1H), 5.04 (s, 1H), 7.18 (d, J = 8.5 Hz, 2H), 7.25 (t, J = 8.0 Hz, 1H), 7.35 (t, J = 8.0 Hz, 1H), 7.54 (d, J = 8.0 Hz, 1H), 7.82 (d, J = 8.5 Hz, 2H), 8.02 (d, J = 8.0 Hz, 1H), 8.28 (s, 1H), 9.78 (s, 1H); 130 NMR (125MHz), c0013: 8 19.3, 21.3, 69.4, 109.6, 113.1, 114.2, 122.4, 123.2, 124.7, 126.8, 128.0, 129.8, 130.0, 133.9, 134.6, 138.8, 143.0, 145.3, 162.8. IR: (NaCI) 3460 cm", 1736 cm'1. HRMS: [M + H]+ = 413.1167, calculated for C21H21N2058, 413.1171. Anal. Calcd. For 021H20N2058: C, 61.15; H, 4.89; N, 6.79. Found: C, 60.17; H, 4.87; N, 6.56. (III-8). To a 1 L round bottom flask was added water (0.165 L) and AcOH (0.165 L). Then III-7 (13.6 g, 33.0 mmol) was dissolved in THF (0.100 L) and was added to the aqueous acid. The mixture was brought down to 0°C and zinc (21.5 g, 0.330 mol) was then slowly added in small portions over 20 min. The suspension stirred at 0°C for 2 h. The solid was filtered off and the filtrate was reduced and then brought to a pH of 8 using concentrated ammonium hydroxide. The amine was extracted into ethyl acetate (4 x 0.150 L), dried using anhydrous sodium sulfate and concentrated. The crude residue was purified by column chromatography (silica gel, 30% EtOAc; 70% DCM) affording the product as a 175 thick oil. Yield (11.1 g, 84.0%). 1H NMR (500MHz), CDCI3: 5 1.51 (s, 3H), 1.93 (s, 2H), 2.30 (s, 3H), 4.51 (dd, J = 14.0, 19.2 Hz, 2H), 4.77 (m, 2H), 4.85 (s, 1H), 7.16-7.32 (m, 4H), 7.58 (s, 1H), 7.63 (d, J = 8.0 Hz, 1H), 7.73 (d, J = 8.0 Hz, 2H), 7.94 (d, J = 8.0 Hz, 1H); 13c NMR (125MHz), once: 8 19.1, 21.4, 51.8, 88.5, 113.3, 113.6, 120.1, 121.5, 123.2, 123.8, 124.9, 126.7, 128.7, 129.8, 135.1, 135.2, 139.2, 144.9, 172.9. IR: (NaCI) 3389 cm“, 3350 cm‘, 1740 cm". HRMS: [M + H]+ = 399.1369, calculated for C21H23N204S, 399.1379. Anal. Calcd. For C21H22N204S: C, 63.30; H, 5.56; N, 7.03. Found: C, 63.18; H, 5.70; N, 6.87. (III-9). To a flame dried 500 mL round bottom flask was added III-8 (10.4 g, 26.1 mmol) and anhydrous DCM (0.250 L). The solution was brought down to 0°C and ethoxycarbonyl isothiocyanate (3.54 mL, 31.4 mmol) was added dropwise. The solution was warmed to room temperature and stirred overnight under nitrogen. The solvents were removed and the product was recrystallized from EtOAc/Hexanes to give a white powder. Yield (12.1 g, 87.0%). 1H NMR (500MHz), CDCI3: 5 1.27 (t, J = 7.1 Hz, 3H), 1.52 (s, 3H), 2.31 (s, 3H), 4.21 (m, 2H), 4.55 (s, 2H), 4.81 (d, J = 8.0 Hz, 2H), 6.26 (d, J = 7.1 Hz, 1H), 7.18-7.33 (m, 4H), 7.62 (d, J = 7.9 Hz, 1H), 7.72-7.76 (m, 3H), 7.92 (d, J = 8.3 Hz, 1H), 8.02 (s, 1H), 10.60 (d, J = 7.1 Hz, 1H); 13C NMR (125MHz), CDCI3: 5 14.1, 19.1, 21.5, 54.8, 62.9, 69.3, 113.7, 113.8, 116.0, 119.9, 123.6, 125.1, 126.1, 126.9, 128.2, 129.9, 134.9, 135.1, 138.8, 145.1, 152.4, 168.8, 179.0. IR: (NaCI) 3250 cm", 3227 cm", 1740 cm", 1724 cm", 1527 cm". HRMS: [M + H]: = 530.1575, calculated for C25H28N30582, 530.1420. Anal. Calcd. For C25H27N30682: C, 56.69; H, 5.14; N, 7.93. Found: C, 56.22; H, 5.13; N, 7.80. Melting Point = 138-140°C. 176 (III-10). To a flame dried 50 mL round bottom flask was added III-9 (0.265 g, 0.501 mmol), anhydrous DCM (20.0 mL), and anhydrous TEA (0.210 mL, 1.50 mmol). The solution was cooled to 0°C and then EDCI (0.212 g, 1.10 mmol) was added and the mixture stirred at 0°C for 1 h and then refluxed until disappearance of the starting material, as indicated on TLC. A solution of NaH (0.100 g, 2.51 mmol) in MeOH (10.0 mL) was then added to the mixture and stirred at room temperature for 2 h. The reaction was washed with 1% HCI (1 x 10.0 mL) and brine (1 x 10.0 mL) and the organics were dried using anhydrous sodium sulfate and concentrated. The crude residue was purified by column chromatography (silica gel, 20% EtOAc; 80% DCM) affording the product as a white solid. Yield (0.151 g, 71.0%). 1H NMR (500MHz), Acetone: 5 1.69 (s, 3H), 2.32 (s, 3H), 2.92 (d, J = 14.7 Hz, 1H), 3.10 (d, J = 14.7 Hz, 1H), 4.85 (s, 1H), 4.92 (s, 1H), 7.27 (m, 1H), 7.36 (m, 3H), 7.66 (s, 1H), 7.83 (s, 1H), 7.87 (d, J = 8.3 Hz, 2H), 7.94 (d, J = 7.8 Hz, 1H), 8.01 (d, J = 8.3 Hz, 1H), 9.81 (s, 1H);13C NMR (125MHz), Acetone: 5 21.3, 24.2, 44.6, 66.0, 114.4, 117.1, 122.6, 122.8, 124.2, 125.2, 125.7, 127.8, 128.8, 130.9, 135.7, 136.4, 140.2, 146.5, 156.6, 175.2. IR: (NaCI) 3390 cm“, 3283 cm'1, 1778 cm", 1728 cm". HRMS: [M + H]+ = 424.1301, calculated for C22H22N304S, 424.1331. Anal. Calcd. For C22H21N304S: C, 62.40; H, 5.00; N, 9.92. Found: C, 62.08; H, 5.15; N, 9.67. Melting Point = 236-238°C. (III-11). To a 100 mL flame dried round bottom flask was added III-10 (0.595 g, 1.41 mmol) and anhydrous toluene (50.0 mL). Then Lawesson’s reagent (0.341 g, 0.844 mmol) was added and the mixture was refluxed for 24 h. 177 The toll mL). T annyd colon as a 2.32 4.96 21 .z 128 176 44C 60. The toluene was then taken off and the crude residue was put into EtOAc (50.0 mL). The organic solution was then washed with brine (1 x 20.0 mL), dried using anhydrous sodium sulfate and concentrated. The crude residue was purified by column chromatography (silica gel, 4% EtOAc; 96% DCM) affording the product as a solid. Yield (0.506 g, 82.0%). 1H NMR (500MHz), Acetone: 5 1.77 (s, 3H), 2.32 (s, 3H), 3.01 (d, J = 13.6 Hz, 1H), 3.21 (d, J = 13.5 Hz, 1H), 4.90 (s, 1H), 4.96 (s, 1H), 7.26-7.40 (m, 4H), 7.76 (d, J = 8.0 Hz, 1H), 7.88-7.92 (m, 3H), 8.03 (d, J = 8.4 Hz, 1H), 9.46 (s, 1H), 10.80 (s, 1H); 13C NMR (125MHz), CDCI3: 5 21.4, 24.1, 43.8, 68.8, 114.4, 117.4, 120.8, 121.8, 124.4, 125.4, 125.9, 127.8, 128.3, 130.9, 135.5, 136.1, 139.6, 146.6, 175.5, 183.0. IR: (NaCI) 3190 cm", 1783 cm", 1895 cm". HRMS: [M + H]* = 440.1167, calculated for C22H22N3O382, 440.1103. Anal. Calcd. For C22H21N30382: C, 60.11; H, 4.82; N, 9.56. Found: C, 60.07; H, 4.75; N, 9.58. Melting Point = 224-226°C. (III-12). To a flame dried 25 mL round bottom flask was added III-11 (0.105 g, 0.239 mmol), anhydrous DCM (10.0 mL), DMAP (0.00300 9, 0.0239 mmol) and DIPEA (0.210 mL, 1.20 mmol). Then Mel (0.0450 mL, 0.717 mmol) was added and the solution stirred at room temperature under nitrogen for 2 h. The DCM was taken off and diethyl ether (30.0 mL) was added to the residue. The organic solution was washed with 1% HCI (1 x 10.0 mL) and brine (1 x 10.0 mL), dried using anhydrous sodium sulfate and concentrated. The crude residue was purified by column chromatography (silica gel, 5% EtOAc; 95% DCM) affording the product as an oil. Yield (0.0900 g, 83.0%). 1H NMR (500MHz), Acetone: 5 1.76 (s, 3H), 2.31 (S, 3H), 2.60 (8, 3H), 2.77 (d, J = 13.2 Hz, 1H), 2.89 178 (d, J = 13.2 Hz, 1H), 4.66 (s, 1H), 4.72 (s, 1H), 7.22-7.35 (m, 4H), 7.70 (s, 1H), 7.83 (d, J = 8.3 Hz, 2H), 7.98 (d, J = 8.5 Hz, 1H), 8.10 (d, J = 8.0 Hz, 1H), 10.27 (s, 1H); 13c NMR (125MHz), CDCI3: 8 12.3, 21.3, 24.8, 48.2, 75.1, 114.2, 115.4, 122.3, 123.5, 123.8, 124.0, 125.4, 127.6, 129.7, 130.8, 135.8, 136.2, 141.3, 148.3, 180.8, 183.0. IR: (NaCI) 3239 cm’1 (broad), 1743 cm", 1701 cm". HRMS: [M + H]+ = 454.1276, calculated for C23H24N30382, 454.1259. Anal. Calcd. For 023H23N30382: C, 60.90; H, 5.11; N, 9.26. Found: C, 59.91; H, 5.19; N, 8.90. (III-13). To a flame dried 50 mL sealed tube was added III-12 (0.0900 g, 0.199 mmol) and dimethylamine (10.0 mL of a 2.0M solution in THF). The tube was sealed and heated to an external sand bath temperature of 75°-90° for 14 h. Once cooled, the tube was opened and nitrogen was purged into the tube to expel any methane thiol that was formed. The whitish product was filtered off and the filtrate was concentrated and the residue was triturated with diethyl ether to obtain additional product, which was then filtered off. Yield (0.0810 g, 91 .0%). 1H NMR (500MHz), DMSO: 5 1.65 (s, 3H), 2.31 (s, 3H), 2.79 (dd, J = 13.1, 26.0 Hz, 2H), 3.00 (s, 3H), 3.08 (s, 3H), 4.76 (s, 1H), 4.8 (s, 1H), 7.22 (t, J = 8.1 Hz, 1H), 7.32 (t, J = 8.2 Hz, 1H), 7.39 (d, J = 8.5 Hz, 2H), 7.58 (d, J = 8.0 Hz, 1H), 7.75 (s, 1H), 7.87-7.92 (m, 3H), 8.33 (s, 1H); 13C NMR (125MHz), DMSO: 5 20.9, 23.9, 36.0, 38.0, 43.2, 67.1, 113.0, 115.3, 121.5, 122.8, 123.0, 123.5, 124.6, 126.7, 127.9, 130.2, 134.1, 134.5, 139.7, 145.5, 169.8, 186.2. lR: (NaCI) 3210 cm'1 (broad), 1715 cm“, 1814 cm". HRMS: [M + H]+ = 451.1818, calculated for C24H27N403S, 451.1804. Anal. Calcd. For C24H25N4038: C, 63.98; H, 5.82; N, 12.44. Found: C, 62.82; H, 5.96; N, 11.76. Melting Point = 142-144°C. 179 (III-14). To a 50 mL round bottom flask was added III-13 (0.200 g, 0.444 mmol) and EtOH (20.0 mL). Then KOEt (0.186 g, 2.22 mmol) was added and the solution refluxed overnight. The solvent was taken off and EtOAc (30.0 mL) was added to the residue. Then 1% HCI (30.0 mL) was added to acidity the solution. The EtOAc layer was discarded and the acidic aqueous layer was extracted again with EtOAc (1 x 10.0 mL) and that organic layer was also discarded. The acidic aqueous layer was neutralized with solid NaHC03 and then extracted with EtOAc (2 x 50.0 mL) and n-BuOH ( 2 x 50.0 mL). The organics were then dried using anhydrous sodium sulfate and concentrated. The crude solid was washed with acetone (10.0 mL) to remove a colored impurity to leave a white solid as product. Yield (0.113 g, 86.0%). 1H NMR (500MHz), CD3OD: 5 1.80 (s, 3H), 2.96 (d, J = 13.2 Hz, 1H), 3.03 (s, 3H), 3.07 (d, J = 13.2 Hz, 1H), 3.20 (s, 3H), 4.85 (s, 1H, hidden by MeOH), 4.90 (s, 1H), 6.96 (t, J = 8.0 Hz, 1H), 7.08 (t, J = 8.0 Hz, 1H), 7.30 (s, 1H), 7.33 (d, J = 8.0 Hz, 1H), 7.39 (d, J = 8.1 Hz, 1H); 13C NMR (125MHz), CD30D: 5 24.6, 36.9, 38.7, 44.4, 69.7, 112.5, 115.2, 116.2, 120.1, 122.6, 124.2, 126.2, 138.5, 141.4, 170.1, 192.1. IR: (NaCI) 3321 cm"1 (broad), 3284 cm'1 (broad), 1884 cm", 1611 cm‘l. HRMS: [M + H]+ = 297.1723, calculated for C17H21N4O, 297.1715. Melting Point = 267-269°C. (III-1). To a 25 mL round bottom flask was added III-14 (0.0500 g, 0.169 mmol), DMF (3.00 mL), THF (4.00 mL) and water (1 .00 mL). Then NMO (0.0290 g, 0.253 mmol) and OsO4 (0.171 mL of a 0.0980 M solution in toluene, 0.0169 mmol). The solution stirred at room temperature for 4 h and then was cooled to 0°C before a solution of NalO4 (0.108 g, 0.507 mmol) in water (2.00 mL) was 180 added a mL)lNa lnkflne aqueot waec BuOH gwe a the n (500k 37411 (d,J 36£L 170, (s, 3 H2, 1th 111 018 299 642 248 9.1 added and stirred for 2 h at 0°C. The solvents were removed and EtOAc (10.0 mL) was added along with a sat. solution of K2803 (10.0 mL). This biphasic mixture stirred for 10 min and then the organic layer was separated and the aqueous layer was extracted with EtOAc (10.0 mL) again. The EtOAc layers were combined, and discarded. The aqueous layer was then extracted with n- BuOH (3 x 20.0 mL), dried using anhydrous sodium sulfate and concentrated to give a whitish powder. The crude product was recrystallized from EtOH to give the natural product as a white powder. Yield (0.0310 g, 62.0%). 1H NMR (500MHz), CDgOD: 5 2.16 (s, 3H), 3.05 (s, 3H), 3.23 (s, 3H), 3.46 (dd, J = 16.6, 37.4 Hz, 2H), 6.97 (t, J = 7.0 Hz, 1H), 7.09 (t, J = 7.1 Hz, 1H), 7.26 (s, 1H), 7.33 (d, J = 8.2 Hz, 1H), 7.45 (d, J = 8.1 Hz, 1H); 13C NMR (125MHz), CD300: 5 31.0, 36.9, 38.8, 49.0, 67.0, 112.6, 114.5, 120.2, 120.3, 122.7, 124.1, 125.8, 138.6, 170.8, 191.7, 207.3. 1H NMR (500MHz), DMSO: 5 2.08 (s, 3H), 2.99 (s, 3H), 3.11 (s, 3H), 3.24 (dd, J = 15.8, 38.3 Hz, 2H), 6.93 (t, J = 8.0 Hz, 1H), 7.05 (t, J = 8.0 Hz, 1H), 7.27 (s, 1H), 7.33 (d, J = 8.2 Hz, 1H), 7.42 (d, J = 7.7 Hz, 1H), 8.24 (s, 1H), 11.01 (s, 1H); 13C NMR (125MHz), DMSO: 5 30.9, 36.0, 37.9, 48.7, 65.2, 111.5, 114.3, 118.5, 119.6, 121.0, 123.0, 124.5, 136.6, 170.1, 187.6, 205.1. IR: (NaCI) 3234 cm1 (broad), 1720 cm", 1703 cm", 1611 cm". HRMS: [M + H]+ = 299.1508, calculated for C16H19N402, 299.1508. Anal. Calcd. For C15H13N402: C, 64.41; H, 6.08; N, 18.78. Found: C, 64.33; H, 6.22; N, 18.42. Melting Point = 246- 248°C. (III-15). To a flame dried 250 mL round bottom flask was added Ill-9 (1 .00 g, 1.89 mmol), anhydrous DCM (75.0 mL), and anhydrous TEA (0.780 mL, 5.67 181 mn adt dis ITII’ W E on Oil 8C mmol). The solution was cooled to 0°C and then EDCI (0.798 g, 4.16 mmol) was added and the mixture stirred at 0°C for 1 h and then refluxed until disappearance of the starting material, as indicated on TLC. A solution of NaH (0.750 g, 18.9 mmol) in MeOH (70.0 mL) was then added to the mixture and refluxed for 8 h. Since the de-tosylation was sluggish with NaOMe the solvent was removed and ethanol (0.120 L) was added followed by KOEt (0.793 g, 9.45 mmol) and the mixture refluxed overnight. The solvent was removed and residue was acidified using 1% HCI and extracted using EtOAc (3 x 50.0 mL). The organics were dried using anhydrous sodium sulfate and concentrated. The crude residue was purified by column chromatography (silica gel, 20% Acetone; 80% DCM) affording the product as a white solid. Yield (0.426 g, 83.0%). 1H NMR (500MHz), DMSO: 5 1.75 (s, 3H), 2.72 (d, J = 13.4 Hz, 1H), 3.01 (d, J = 13.5 Hz, 1H), 4.82 (s, 1H), 4.91 (s, 1H), 6.99 (t, J = 6.8 Hz, 1H), 7.09 (t, J = 7.1 Hz, 1H), 7.37 (m, 2H), 7.57 (d, J = 8.1 Hz, 1H), 8.38 (s, 1H), 10.70 (s, 1H), 11.13 (s, 1H); 13C NMR (125MHz), Acetone-d6: 5 23.9, 44.1, 65.9, 111.9, 114.7, 115.9, 119.5, 120.4, 121.9, 123.1, 125.2, 137.6, 140.3, 156.6, 175.8. IR: (NaCI) 3300 cm“, 1790 cm“, 1720 cm". HRMS: [M + H]+ = 270.1255, calculated for C15H16N302, 270.1243. Anal. Calcd. For C15H15N302: C, 66.90; H, 5.61; N, 15.60. Found: C, 66.38; H, 5.69; N, 15.11. Melting Point = 238-240°C. (III-16). To a 25 mL round bottom flask was added III-15 (0.172 g, 0.639 mmol), THF (8.00 mL) and water (1.00 mL). Then NMO (0.112 g, 0.959 mmol) and 0804 (0.650 mL of a 0.0980 M solution in THF, 0.0639 mmol) were added. The solution stirred at room temperature for 2 h and then was cooled to 0°C 182 before a solution of Na|O4 (0.410 g, 1.92 mmol) in water (3.00 mL) was added and stirred at room temperature overnight. The solvents were removed and EtOAc (10.0 mL) was added along with a sat. solution of K2803 (10.0 mL). This biphasic mixture stirred for 10 min and then the organic layer was separated and the aqueous layer was extracted with n-BuOH (3 x 10.0 mL). The EtOAc layer and nBuOH layers were combined, dried using anhydrous sodium sulfate and concentrated. The crude residue was purified by column chromatography (silica gel, 10% MeOH; 90% DCM) affording the product as an off white solid. Yield (0.107 g, 61.0%). Product was recrystallized over 1 week with EtOAc. 1H NMR (500MHz), CDgOD: 5 2.15 (s, 3H), 3.49 (d, J = 7.9 Hz, 1H), 3.59 (d, J = 7.9 Hz, 1H), 7.02 (t, J = 8.0 Hz, 1H), 7.11 (t, J = 8.2 Hz, 1H), 7.27 (s, 1H), 7.35 (d, J = 8.1 Hz, 1H), 7.69 (d, J = 8.1 Hz, 1H); 13C NMR (125MHz), CD3OD: 5 30.5, 49.2, 64.1, 112.7, 113.9, 120.5, 120.6, 122.9, 123.9, 125.6, 138.8, 160.0, 179.0. IR: (KBr) 3364 (br) cm'1, 1774 cm", 1711 cm“. HRMS: [M + H]+ = 272.1055, calculated for C14H14N303, 272.1035. Anal. Calcd. For C14H13N303: C, 61.99; H, 4.83; N, 15.49. Found: C, 61.10; H, 4.82; N, 15.38. Melting Point = 234-236°C. (III-17). To a sealed tube was added III-12 (0.850 g, 1.88 mmol) in THF (5.00 mL) and NH4OH (15.0 mL). The mixture was heated at 90°C until the disappearance of the starting material, as indicated by TLC. The precipitate that formed was filtered, acidified with 5% HCI and extracted with nBuOH (3 x 20.0 mL), dried using anhydrous sodium sulfate and concentrated. The crude residue was purified by column chromatography (silica gel, 10% MeOH; 90% DCM) affording the product as an off white solid. Yield (0.575 g, 72.0%). 1H NMR 183 (500MHz), DMSO: 5 1.62 (s, 3H), 2.30 (s, 3H), 2.72 (d, J = 13.5 Hz, 1H), 2.81 (d, J = 13.5 Hz, 1H), 4.72 (s, 1H), 4.77 (s, 1H), 7.22 (t, J = 8.3 Hz, 1H), 7.32 (t, J = 8.3 Hz, 1H), 7.37 (d, J = 9.5 Hz, 2H), 7.64 (d, J = 8.1 Hz, 1H), 7.69 (s, 1H), 7.85 (d, J = 9.5 Hz, 2H), 7.90 (d, J = 8.5 Hz, 1H), 8.25 (s, 1H); 13C NMR (125MHz), DMSO: 5 20.9, 23.7, 43.1, 66.0, 113.0, 115.2, 121.5, 123.0, 123.3, 124.7, 126.7, 128.0, 130.2, 134.0, 134.5, 139.9, 145.5, 170.9, 187.5. IR: (KBr) 3470 cm", 3351 cm“, 3307 cm", 3088 cm'1, 1707 cm‘1, 1857 cm‘1. HRMS: [M + H]+ = 423.1494, calculated for C22H23N4038, 423.1491. Anal. Calcd. For C22H22N4038: C, 62.54; H, 5.25; N, 13.25. Found: C, 61.21; H, 5.17; N, 13.05. Melting Point = 273-275’C. (III-18). To a 100 mL round bottom flask was added III-17 (0.500 g, 1.18 mmol) and EtOH (60.0 mL). Then KOEt (0.991 g, 11.8 mmol) was added and the mixture refluxed for 24 h. The EtOH was taken off and the pH of an aqueous mixture of the crude residue was adjusted to 8. The aqueous mixture was then extracted with nBuOH (3 x 40.0 mL). The organics were combined, dried using anhydrous sodium sulfate and concentrated. The crude residue was purified by column chromatography (silica gel, 20% IVleOH; 90% DCM) affording the product as an off white solid. The product was recrystallized from EtOH. Yield (0.150 g, 47.0%). 1H NMR (500MHz), DMSO: 5 1.67 (s, 3H), 2.75 (d, J = 13.4 Hz, 1H), 2.81 (d, J = 13.4 Hz, 1H), 4.75 (s, 1H), 4.78 (s, 1H), 6.92 (t, J = 7.1 Hz, 1H), 7.04 (t, J = 7.0 Hz, 1H), 7.26 (d, J = 2.4 Hz, 1H), 7.33 (d, J = 8.2 Hz, 1H), 7.48 (d, J = 8.0 Hz, 1H), 8.03 (s, 1H), 10.96 (s, 1H); 13C NMR (125MHz), DMSO: 5 23.9, 43.4, 66.3, 111.4, 114.7, 115.3, 118.3, 119.7, 120.9, 122.6, 124.8, 136.5, 140.7, 170.8, 189.1. IR: (KBr) 3470 cm'l, 3390 (br) cm'1, 3200 (br) cm’1, 1892 cm", 184 1650 cm'i. HRMS: [M + H]* = 269.1405, calculated for C15H17N4O, 269.1402. Anal. Calcd. For C15H15N4O: C, 67.15; H, 6.01; N, 20.88. Found: C, 63.98; H, 5.89; N, 19.84. Melting Point = 264-266°C. (III-19). To a 25 mL round bottom flask was added III-18 (0.0990 g, 0.366 mmol), DMF (7.00 mL), THF (1.00 mL) and water (1.00 mL). Then NMO (0.0640 g, 0.549 mmol) and OsO4 (0.370 mL of a 0.0980 M solution in THF, 0.0366 mmol) were added. The solution stirred at room temperature for 3 h and then was cooled to 0°C before a solution of NaIO4 (0.235 g, 1.10 mmol) in water (2.00 mL) was added and stirred at room temperature overnight. The solvents were removed and EtOAc (10.0 mL) was added along with a sat. solution of K2803 (10.0 mL). This biphasic mixture stirred for 10 min and then the organic layer was separated and the aqueous layer was extracted with n-BuOH (6 x 10.0 mL). The EtOAc layer and nBuOH layers were combined, dried using anhydrous sodium sulfate and concentrated. The crude residue was purified by column chromatography (silica gel, 20% MeOH; 90% DCM) affording the product as an off white solid. The product was recrystallized from EtOH. Yield (0.0190 g, 19.0%). 1H NMR (500MHz), CD3OD: 5 2.18 (s, 3H), 3.19 (d, J = 7.3 Hz, 1H), 3.64 (d, J = 7.1 Hz, 1H), 6.97 (t, J = 8.0 Hz, 1H), 7.08 (t, J = 8.0 Hz, 1H), 7.20 (s, 1H), 7.33 (d, J = 8.2 Hz, 1H), 7.51 (d, J = 8.2 Hz, 1H); 13C NMR (125MHz), CD3OD: 5 30.9, 66.5, 112.5, 114.1, 120.28, 120.29, 122.7, 124.0, 125.9, 138.5, 171.7, 192.5, 207.7. IR: (KBr) 3470 cm", 3420 cm'l, 3270 (br) cm", 3178 (br) cm", 1720 cm“, 1890 cm‘1, 1850 cm". HRMS: [M + H]: = 271.1191, calculated for C14H15N402, 271.1195. Melting Point = 283-2850C. 185 10. 11. 12. References Baker, D. D.; Alvi, K. A., Small-molecule natural products: new structures, new activities. Curr. Opin. Biotechnol. 2004, 15, 576-583. Hill, R. A., Marine natural products. Annu. Rep. Prog. Chem, Sect. B: Org. Chem. 2005, 101, 124-136. Jha, R. K.; Xu, Z.-r., Biomedical compounds from marine organisms. Mar. Drugs 2004, 2, 123-146. Molinski, T. F.; Dalisay, D. S.; Lievens, S. L.; Saludes, J. P., Drug development from marine natural products. Nat. Rev. Drug Discovery 2009, 8, 69-85. Simmons, T. L.; Andrianasolo, E.; McPhail, K.; Flatt, P.; Gerwick, W. H., Marine natural products as anticancer drugs. Mol. Cancer Ther. 2005, 4, 333-342. Barsby, T., Drug discovery and sea hares: bigger is better. Trends Biotechnol. 2006, 24, 1-3. Gul, W.; Hamann, M. T., Indole alkaloid marine natural products: An established source of cancer drug leads with considerable promise for the control of parasitic, neurological and other diseases. Life Sci. 2005, 78, 442-453. Bergmann, T.; Schories, D.; Steffan, B., Alboinon, an oxadiazinone alkaloid from the ascidian Dendrodoa grossularia. Tetrahedron 1997, 53, 2055-2060. Guyot, M.; Meyer, M., An 3-indonI-4H-imidazol-4-one from the tunicate Dendrodoa grossularia. Tetrahedron Left. 1986, 27, 2621-2. Heitz, S.; Durgeat, M.; Guyot, M.; Brassy, C.; Bachet, B., New indolic derivative of 1,2,4-thiadiazole, isolated from a tunicate (Dendrodoa grossular). Tetrahedron Left. 1980, 21 , 1457-8. Loukaci, A.; Guyot, M.; Chiaroni, A.; Riche, C., A new indole alkaloid from the marine tunicate Dendrodoa grossularia. J. Nat. Prod. 1998, 61, 519- 522. Moquin, C.; Guyot, M., Grossularine, a novel indole derivative from the marine tunicate, Dendrodoa grossularia. Tetrahedron Left. 1984, 25, 5047-8. 186 13. 14. 15. 16. 17. 18. 19. 20. 21. 22. 23. 24. 25. Moquin-Pattey, C.; Guyot, M., Grossularine-1 and grossularine-2, cytotoxic a-carbolines from the tunicate Dendrodoa grossularia. Tetrahedron 1989, 45, 3445-50. Hogan, I. T.; Sainsbury, M., The synthesis of dendrodoine, 5-[3-(N,N- dimethylamino-1,2,4-thiadiazolyI]-3-indolylmethanone, a metabolite of the marine tunicate Dendroda grossular. Tetrahedron FIELD Full Journal Tif/e:Tetrahedron 1984, 40, 681-2. Helbecque, N.; Moquin, C.; Bernier, J. L.; Morel, E.; Guyot, M.; Henichart, J. P., Grossularine-1 and grossularine-2, a-carbolines from Dendrodoa grossularia, as possible intercalative agents. Cancer Biochem. Biophys. 1987, 9, 271-9. Loukaci, A.; Guyot, M., Revised assignments of the 13C NMR spectra of grossularine-1 and -2 using 2D heteronuclear 1H-1BC correlations. Magn. Reson. Chem. 1996, 34, 143-5. Choshi, T.; Yamada, S.; Sugino, E.; Kuwada, T.; Hibino, 8., Total synthesis of grossularines-1 and -2. J. Org. Chem. 1995, 60, 5899-904. Fisk, J. S.; Mosey, R. A.; Tepe, J. J., The diverse chemistry of oxazol-5- (4H)-ones. Chem. Soc. Rev. 2007, 36, 1432-1440. Hupp, C. D.; Tepe, J. J., 1-Ethyl-3-(3-dimethylaminopropyl)carbodiimide Hydrochloride-Mediated Oxazole Rearrangement: Gaining Access to a Unique Marine Alkaloid Scaffold. J. Org. Chem. 2009, 74, 3406-3413. Gadwood, R. C.; Kamdar, B. V.; Dubray, L. A. C.; Wolfe, M. L.; Smith, M. P.; Watt, W.; Mizsak, S. A.; Groppi, V. E., Synthesis and biological activity of spirocyclic benzopyran imidazolone potassium channel openers. J. Med. Chem. 1993, 36, 1480-7. Kiec-Kononowicz, K.; Zejc, A., Syntheses of some 2-amino—5,5- diphenylimidazolin-4-one derivatives. Pol. J. Chem. 1980, 54, 2217-24. Hupp, C. D.; Tepe, J. J., Total Synthesis of a Marine Alkaloid from the Tunicate Dendrodoa grossularia. Org. Left. 2008, 10, 3737-3739. Garg, N. K.; Sarpong, R.; Stoltz, B. M., The First Total Synthesis of Dragmacidin D. J. Am. Chem. Soc. 2002, 124, 13179-13184. Kharasch, M. 8.; Kane, S. S.; Brown, H. C., Presence of indole in \"practical\" a-methylnaphthalene. J. Am. Chem. Soc. 1940, 62, 2242-3. Scheibye, S.; Pedersen, B. S.; Lawesson, S. 0., Studies on organophosphorus compounds. XXI. The dimer of p- 187 26. methoxyphenylthionophosphine sulfide as thiation reagent. A new route to thiocarboxamides. Bull. Soc. Chim. Belg. 1978, 87, 229-38. Pappo, R.; Allen, D. 8., Jr.; Lemieux, R. U.; Johnson, W. S., Osmium tetroxide-catalyzed periodate oxidation of olefinic bonds. J. Org. Chem. 1956, 21 , 478-9. 188 CHAPTER IV BIOLOGICAL TESTING OF NATURAL PRODUCT, ANALOGS AND OTHER HETEROCYCLIC COMPOUNDS IV.A Biological target of natural product and hypothesis for activity The biological target from the start of the project was checkpoint kinase 2 (Chk2), as discussed in chapter I. This kinase is responsible for helping to maintain the integrity of the genome by interpreting certain cellular distress signals and activating other proteins downstream to initiate a response. These responses can include cell cycle abrogation and allow for cellular repair or it can include programmed cell death, or apoptosis.1 It was hypothesized that the natural product (III-1) had some structural characteristics similar to that of a known Chk2 inhibitor, indoloazepine (Figure IV- 1). Furthermore, the majority of the hydrogen bonding contacts thought to be important for indoloazepine’s activity was present in the natural product. As a result, the curiosity of elucidating potential biological activity of this uniquely substituted imidazolone (III-1) prompted an endeavor to determine if there was any similarity in biological properties to indoloazepine, specifically, as a checkpoint kinase 2 inhibitor. lndoloazepine Figure IV-1. The indole natural product III-1 and indoloazepine 189 IV.B Kinase screen for natural product and analogs The natural product (III-1) and the two analogs synthesized were sent to a kinase screening company (Millipore). The compounds were screened for more than just Chk2 inhibition so that in the event of activity, insight into the degree of selectivity against other kinases could be evaluated. The kinases screened included Chk1, Chk2, GSK3B, IKKd and IKKB. A general illustration of how the kinase screen works is shown in Figure IV-2. Essentially, a known substrate for the kinase is incubated with a compound, such as III-1, along with radio-labelled ATP. The amount of radio-activity in the end is directly related with the incorporation of 33P, which can be correlated to how well the compound is at inhibiting the kinase. For example, if the compound is a good inhibitor of the kinase, there will be very little radio-activity in the end. Filter Scintillation counfing Substrate Substrate Figure IV-2. Kinase screen performed by Millipore The results from the kinase screen are shown below in Table IV-1. The numerical value in each of the cells of the table is the percent activity of that kinase after being incubated with the corresponding compound. The lower the percent activity, the better of an inhibitor the compound is. The natural product and analogs were tested at 50 (M to identify it there was any activity of kinase 190 inhibition. As shown in Table lV-1, all three compounds were shown to be inactive for all five kinases tested. Taking into account the experimental error, all compounds essentially resulted in 100% activity for all kinases. Although these results are disappointing, it doesn’t unequivocally prove that the compounds are inactive. Table IV-1. Results of kinase screen for Ill-1, III-16 and Ill-19 OT“ HN O Kinase \ 0 fl III-16 Chk1 1 12 103 107 Chk2 102 106 101 GSK3fi 100 94 99 IKKa 103 103 105 IKKB 96 93 102 Biological . . . . . . result Inactive Inactive Inactive IV.C Other biological testing for natural product and analogs The natural product and two analogs were also tested in our laboratory in other assays to determine any biological activity. Compounds III-1, III-16 and III- 19 were tested in a cell proliferation assay to determine if they were cytotoxic to MCF-7 cells, a breast cancer cell line. Furthermore, they were tested in a Iuciferase and whole blood assay for inhibition of NF-KB and IL-6 production, respectively, which are other biological targets of our laboratory. The two other quaternary imidazolones synthesized via the diketone rearrangement, compounds "-18 and "-23, were also tested for NF-KB inhibition. Table IV-2 191 shows the results of all compounds for these assays. It can be seen that all compounds tested are inactive for the inhibition of NF-KB, inactive for the inhibition of IL-6 production and non-cyctotoxic for the MCF-7 cancer cell line. Table lV-2. Results for cytotoxicity, NF-KB and IL-6 assays Compound Target NF-KB lL-6 MCF-7 cells III-1 inactive inactive non-cytotoxic III-16 -- inactive non-cflotoxic III-19 -- inactive non-cytotoxic ll-18 inactive -- -- lI-23 inactive -- -- IV.D Synthesis of additional heterocycles Throughout the process of developing a synthetic method to gain access to the imidazolone scaffold found in the natural product, other heterocycles were synthesized and were subsequently tested for biological activity. The majority of the heterocycles formed contained the indole and hydantoin moiety in various constitutions. Additionally, an indoloazepine analog was also synthesized. The following section describes the synthesis of these heterocycles. Heterocycles such as indoles substituted in the 3-position by a hydantoin moiety were synthesized through a reaction between an aldehyde, ammonium carbonate and potassium cyanide (Scheme IV-1).2 Hydantoin IV-13 was synthesized in a 36% yield through a reaction between indole-3-carboxaldehyde and the reagents described above, while a N-tosyl analog was synthesized in a 18% yield through a similar reaction. After the indolic nitrogen of indole-3- carboxaldehyde was protected with a tosyl group, the reaction using ammonium carbonate and potassium cyanide was used to produce the final hydantoin (IV-3). 192 Scheme IV-1. Synthesis of lV-1 and IV-3 O O H HN/Q \ (NH4)ZCO3, KCN _ N” H 0/ EtOH ' N 2 i 0 R l‘l IV-2: R = Ts R IV-1: R = H, 38% IV-3: R = Ts, 18% A unique compound was synthesized using a hydantoin phosphonate reagent developed by Meanwell4 (Scheme lV-2). Protection of the indolic nitrogen of lV-4 with p-toluene sulfonyl chloride gave keto ester lV-5 in a 90% yield. A subsequent Horner—Wadsworth-Emmons reaction produced hydantoin lV-6 in about an 8:1 mixture of E and Z isomers in a 91% yield (Scheme IV-2). Geometric isomers were identified using NOESY experiments (Figure IV-9). Scheme IV-2. Synthesis of hydantoin IV-6 O O O O OEt TsCI, DMAP _ OEt \ DIPEA DCM? \ H 'V'4 90% N IV-5 Ts O Phosphonate, HN o NaOEt, EtOH, rt Phosphonate= 04 PI, 91%,~8:1(E22) HEtd OEt H o N o HN \ O OEt \ N IV-6 Ts Since indoloazepine was found to be a potent checkpoint kinase 2 inhibitor,5 it was thought that synthesizing an analog could help identify the 193 structural features necessary for activity. It was decided to synthesize an analog that did not contain the seven membered ring between the indole and imidazolone moieties. To synthesize this analog, lndole-2-carboxylic acid was coupled to methylamine using EDCI to afford IV-7 in a 81% yield. Following a Vilsmeier-Haacka' 7 reaction to produce aldehyde IV-8, a condensation between thiohydantoin and IV-8 produced thiohydantoin IV-9 in a 86% yield. Subsequently, after an S-methylation, the final imidazolone, IV-11, was produced in a 26% yield by replacing the S-methyl moiety with an NH2 group using ammonium hydroxide. Scheme IV-3. Synthesis of IV-11 . O N EDCI, DMAP, _ H DCM, 0°c to rt, N ”N 81% IV-7 (COC|)2. DMF, DCM H 0°C to rt, 75% N S O H :h 88’” O H \ O ‘ piperidine, EtOH, N HN— N 86% H H HN— IV-8 N-9 Mel, NaOH, MeOH, 96% NH4OH THF, sealed 7 tube, 26% 194 IV.E Biological testing for additional heterocycles The biological results for compounds IV-1, IV-3, IV-6, IV-9 and lV-11 are illustrated in Table lV-3. Unfortunately, all the compounds tested for kinase inhibition were found to be inactive. Additionally, none of the compounds exhibited any propensity for inhibition or abrogation of lL-6 production. Further testing is needed to determine if any of the compounds synthesized have any biological value. Table IV-3. Biological activity results for IV-1, IV-3, IV-6, IV-9 and lV-11 Cmp d Tagget ' Chk1 Chk2 GSK3B IKKa IKKB NF-KB IL-6 IV-1 inactive inactive inactive inactive inactive inactive inactive IV-3 -- -- -- -- -- inactive inactive IV-6 -- -- -- -- -- inactive -- IV-9 -- -- -- -- -- inactive -- IV-1 1 inactive inactive -- -- -- -- -- 195 IV.F General experimental information Reactions were carried out in flame-dried glassware under nitrogen atmosphere. All reactions were magnetically stirred and monitored by TLC with 0.25 pm pre-coated silica gel plates using either UV light or iodine to visualize the compounds. Column chromatography was carried out on Silica Gel 60 (230-400 mesh) supplied by EM Science. Yields refer to chromatographically and spectroscopically pure compounds unless otherwise noted. Infrared spectra were recorded on a Nicolet IR/42 spectrometer. 1H and 13C NMR spectra were recorded on a Varian Unity Plus-500 spectrometer or a Varian lnova-300, as noted in the experimental for each compound. Chemical shifts are reported relative to the residue peaks of the solvent (CDCI3: 7.24 ppm for 1H and 77.0 ppm for 13C) (Acetone-d5: 2.04 ppm for 1H and 29.8 ppm for ‘30) (DMSO-de: 2.49 ppm for 1H and 39.5 ppm for 13C). The following abbreviations are used to denote the multiplicities: s = singlet, d = doublet, dd = doublet of doublets, t = triplet, and m = multiplet. HRMS were obtained with a Micromass Q-ToF Ultima API LC-MS/MS mass spectrometer. Elemental analysis data were obtained on a Perkin Elmer 2400 Series II CHNS/O analyzer. Purity of compounds, whose elemental analyses were above the ACS tolerated 0.4% deviation, were confirmed by 1H NMR and 13C NMR. Melting points were obtained using an Electrothermal® capillary melting point apparatus and are uncorrected. Reagents and solvents were purchased from commercial suppliers and used without further purification. Anhydrous methylene chloride and toluene were 196 dispensed from a delivery system which passes the solvents through a column packed with dry neutral alumina. IV.G Experimental procedures and characterization Cell Proliferation Assay The cell proliferation study was performed by Thu Nguyen and all detailed experimental information is found within her notebook. The general procedure is as follows. Adherent cells were seeded with 500 uL of approximately 2.5 x 105 cells/mL in a 24-well plate. When cells were near 80% confluent, cells were treated with the appropriate drug in various doses. Cells were incubated for 12, 24 and 48 hours. At each time point, cells were released from the plate with 100 pL of 0.25% trypsin/EDTA and diluted to 500 pL, which was then counted via a cell counter (Beckman Coulter Z1 Coulter Particle Counter). Luciferase Assay The Iuciferase assay was performed by Thu Nguyen, Teri Lansdell and/or Behnaz Shafii. All detailed experimental information is located within their respective notebooks. The general experimental procedure is as follows. HeLa NF-KB cell line was cultured in DMEM, complemented with 10% Fetal Bovine Serum, 1 mM sodium pyruvate, 0.1 mg/mL Hygromycin, 2mM-L-glutamine, 1mM sodium pyruvate, 100 units/mL penicillin, 4.5 g/L D-glucose, and no phenol red. The day before the Iuciferase assay, the medium was switched to DMEM with 2% FBS in addition to 2mM-L-glutamine, 1mM sodium pyruvate, and 100 units/mL penicillin. The day of the experiment, the medium utilized was serum free DMEM with 2mM-L-glutamine, 1mM sodium pyruvate, and 100 units/mL 197 ex prr 110 CH 001 010 Ser biOI tEst hog 200 penicillin. Cells were propagated at 37°C with 5% C02, and ambient oxygen. Cells were seeded in a 96-well plate between 35,000-50,000 cells per well with DMEM 2% FBS. The outside rows and columns of the plate were not used. Cell were allowed to adhere and grow overnight at 37°C with 5% CO2, and ambient oxygen. 0n the day of the experiment, the media was removed and replaced with serum free DMEM. Cells were pretreated with 2 (in 1 [JL DMSO/well) for 30 minutes before activation with 25 ng/mL TNF-o and incubated an additional 8 hours. Steady glo reagent (Promega) was added to each well and data was collected using a Iuminometer. Whole Blood IL-6 Assay The whole blood assay was performed by Teri Lansdell and all detailed experimental information is located in her notebooks. The general experimental procedure is as follows. After obtaining the appropriate approval for de-identified human cell lines, human whole blood was obtained through the Jasper Research Clinic, Kalamazoo, MI, from a single healthy, fasted volunteer and was collected in glass citrated tubes by venipuncture. Only samples with a white blood cell count falling within the normal range (4800-10,800 per liter) were used. The blood was diluted 1:10 in RPMI-1640 media supplemented with 10% fetal bovine serum, 100 U/mL penicillin, and 100 pg/mL streptomycin. Aliquots of diluted blood (1 mL) were preincubated with vehicle (0.1% DMSO, final concentration) or test agent (final concentrations were 20, 10, 5, 2.5, 1.25 and 0.625 pM) for two hours at 37 °C, 5 % C02. lL-1B (Roche) was added to a final concentration of 200 U/mL and the samples were further incubated for 18 hours at 37 °C, 5 % 198 C02. At the end of the incubation period, the blood samples were centrifuged at 3000 X g, 4 °C, for 10 minutes. The plasma was removed, snap frozen and stored at -80 C. IL-6 levels were determined by ELISA (R & D Systems). Kinase Profiler (Millipore) The kinase testing was performed by Millipore. All detailed experimental information can be found on their website.8 The general experimental procedure for each kinase is described below. CHK1 (h) CHK1 (h) is incubated with 8 mM MOPS pH 7.0, 0.2 mM EDTA, 200 (M KKKVSRSGLYRSPSMPENLNRPR, 10 mM MgAcetate and [v-33P-ATP] (specific activity approx. 500 cpm/pmol, concentration as required). The reaction is initiated by the addition of the MgATP mix. After incubation for 40 minutes at room temperature, the reaction is stopped by the addition of 3% phosphoric acid solution. 10 pL of the reaction is then spotted onto a P30 filtermat and washed three times for 5 minutes in 75 mM phosphoric acid and once in methanol prior to drying and scintillation counting. CHK2 (h) CHK2 (h) is incubated with 8 mM MOPS pH 7.0, 0.2 mM EDTA, 200 pM KKKVSRSGLYRSPSMPENLNRPR, 10 mM MgAcetate and [v-33P-ATP] (specific activity approx. 500 cpm/pmol, concentration as required). The reaction is initiated by the addition of the MgATP mix. After incubation for 40 minutes at room temperature, the reaction is stopped by the addition of 3% phosphoric acid solution. 10 pL of the reaction is then spotted onto a P30 filtermat and washed 199 three times for 5 minutes in 75 mM phosphoric acid and once in methanol prior to drying and scintillation counting. GSK3B (h) GSKBB (h) is incubated with 8 mM MOPS pH 7.0, 0.2 mM EDTA, 20 uM YRRAAVPPSPSLSRHSSPHQS( p) EDEEE (phospho G82 peptide), 10 mM MgAcetate and [v-33P-ATP] (specific activity approx. 500 cpm/pmol, concentration as required). The reaction is initiated by the addition of the MgATP mix. After incubation for 40 minutes at room temperature, the reaction is stopped by the addition of 3% phosphoric acid solution. 10 pL of the reaction is then spotted onto a P30 filtermat and washed three times for 5 minutes in 50 mM phosphoric acid and once in methanol prior to drying and scintillation counting. IKKo (h) IKKa (h) is incubated with 8 mM MOPS pH 7.0, 0.2 mM EDTA, 200 pM peptide, 10 mM MgAcetate and [v-33PATP] (specific activity approx. 500 cpm/pmol, concentration as required). The reaction is initiated by the addition of the MgATP mix. After incubation for 40 minutes at room temperature, the reaction is stopped by the addition of 3% phosphoric acid solution. 10 pL of the reaction is then spotted onto a P30 filtermat and washed three times for 5 minutes in 75 mM phosphoric acid and once in methanol prior to drying and scintillation counting. IKKB (h) IKKB (h) is incubated with 8 mM MOPS pH 7.0, 0.2 mM EDTA, 100 pM peptide, 10 mM MgAcetate and [v-33P-ATP] (specific activity approx. 500 200 cpm/pmol, concentration as required). The reaction is initiated by the addition of the MgATP mix. After incubation for 40 minutes at room temperature, the reaction is stopped by the addition of 3% phosphoric acid solution. 10 pL of the reaction is then spotted onto a P30 filtermat and washed three times for 5 minutes in 75 mM phosphoric acid and once in methanol prior to drying and scintillation counting. Experimentals for compounds IV-1-IV-11 (IV-1). To a flame dried 250 mL round bottom flask was added indole-3- carboxaldehyde (1.00 g, 6.90 mmol) and ethanol (70.0 mL). Then ammonium carbonate (2.00 g, 20.7 mmol) and water (70.0 mL) were added and the mixture was heated in an oil bath to 50°C. Once everything was in solution, KCN (0.583 g, 8.97 mmol in 10.0 mL H2O) was added over 25 min. The light orange solution stirred at 60°C overnight and became a darker orange color. The ethanol was taken off by rotary evaporation and the solid that precipitated out was filtered off. The filtrate was neutralized with 1% HCI (aq.) and extracted with ethyl acetate (3 x 50.0 mL). The organics were dried using anhydrous sodium sulfate and concentrated to give a yellowish oil. The crude material was purified by column chromatography (silica gel, 4:1 ethyl acetate/hexanes) affording the product as an off white powder. Yield (0.537 g, 36.0%). 1H NMR (500 MHz), Acetone: 5 5.47 (s, 1H), 7.03-7.06 (m, 1H), 7.12-7.16 (m, 1H), 7.32 (bs, 1H), 7.42-7.44 (m, 2H), 7.57-7.59 (m, 1H), 9.80 (bs, 1H), 10.30 (bs, 1H); 13c NMR (125 MHz), Acetone: 8 57.1, 110.8, 112.6, 119.7, 120.2, 122.7, 125.4, 126.5, 137.9, 157.9, 174.8; IR (NaCI) 1710 cm", 1770 cm'1, 3300 cm". M.S: calculated for C11H9N302 [M+] = 201 215 and found [M+] = 214.9. Anal. Calcd. For C11H9N302: C, 61.40; H, 4.20; N, 19.50. Found: C, 60.90; H, 3.60; N, 18.90. Melting Point = 220-222°C. (IV-2).9 To a flame dried 500 mL round bottom flask was added indole-3- carboxaldehyde (5.00 g, 34.5 mmol), dry DCM (0.300 L), TsCl (13.1 g, 69.0 mmol), DMAP (10.5 g, 86.2 mmol), and DIPEA (15.0 mL, 86.2 mmol). The solution stirred at room temperature overnight under nitrogen. The reaction was quenched and washed with 1% HCI (aq.) and the organics were dried using anhydrous sodium sulfate and concentrated. A silica plug was done to isolate product (silica gel, 100% ethyl acetate). Yield (10.3 g, 99.0%). 1H NMR (500 MHz), CDCI3: 5 2.35 (s, 3H), 7.26-7.41 (m, 4H), 7.83 (d, J = 8.9 Hz, 2H), 7.93 (d, J = 8.1 Hz, 1H), 8.21 (s, 1H), 8.23 (d, J = 8.9 Hz, 1H), 10.10 (s, 1H); 13C NMR (125 MHz), CDCI3: 5 21.6, 113.2, 122.3, 122.5, 124.9, 126.2, 127.1, 130.2, 134.2, 135.1, 136.1, 148.1, 185.2; IR (NaCI) 1710 cm“, 2750 cm", 2850 cm". (IV-3). To a flame dried 250 mL round bottom flask was added IV-2 (3.30 g, 11.0 mmol) and ethanol (75.0 mL). Then ammonium carbonate (3.18 g, 33.1 mmol) and water (75.0 mL) were added and the mixture was heated in an oil bath to 50°C. Once everything was in solution, KCN (1.08 g, 16.6 mmol in 10.0 mL H20) was added over 25 min. The light orange solution stirred at 60°C overnight and became a darker orange color. The ethanol was taken off by rotary evaporation and the solid that precipitated out was filtered off. The filtrate was neutralized with 1% HCI (aq.) and extracted with ethyl acetate (3 x 50.0 mL). The organics were dried using anhydrous sodium sulfate and concentrated. The crude material was purified by column chromatography (silica gel, 3:2 ethyl 202 acetate/hexanes) affording the product as an off white powder. Yield (0.717 g, 18.0%). 1H NMR (500 MHz), Acetone: 5 2.34 (s, 3H), 5.53 (s, 1H), 7.27 (t, J = 7.3 Hz, 1H), 7.35-7.40 (m, 4H), 7.64 (d, J = 7.8 Hz, 1H), 7.84 (s, 1H), 7.89 (d, J = 8.3 Hz, 2H), 8.01 (d, J = 8.3 Hz, 1H), 9.78 (s, 1H); 13C NMR (125 MHz), Acetone: 5 21.4, 56.3, 114.4, 118.5, 121.3, 124.2, 125.9, 126.0, 127.9, 129.3, 131.0, 135.8, 138.2, 148.8, 157.5, 173.3; IR (NaCI) 3249 cm", 1772 cm", 1724 cm". M.S: calculated for C18H15N304S [M+] = 369 and found [M+] = 369.0. Anal. Calcd. For C13H15N304S: C, 58.53; H, 4.09; N, 11.38. Found: C, 57.68; H, 4.16; N, 11.25. Melting Point = 226-228 °C. (IV-5). To a 500 mL flame dried round bottom flask was added IV-4 (4.47 g, 20.6 mmol), dry dichloromethane (0.225 L), TsCI (7.83 g, 41.2 mmol), DMAP (6.28 g, 51.5 mmol), and DIPEA (9.00 mL, 51.5 mmol), The reaction stirred under nitrogen overnight. The reaction was washed with sat. NaHC03 ( 2 x 50.0 mL) and brine (2 x 50.0 mL) and the organic layer was dried using anhydrous sodium sulfate and concentrated. The crude material was purified by column chromatography (silica gel, DCM) affording the product as a solid. Yield (6.80 g, 90.0%). 1H NMR (500 MHz), CDCI3: 5 1.43 (t, J = 7.2 Hz, 3H), 2.34 (s, 3H), 4.43 (q, J = 7.0 Hz, 2H), 7.26-7.41 (m, 4H), 7.85 (d. J = 8.5 Hz, 2H), 7.94 (m, 1H), 8.34 (m, 1H), 8.83 (s, 1H); 13C NMR (125 MHz), CDCI3: 5 14.0, 21.6, 62.5, 113.1, 116.9, 122.9, 125.2, 126.1, 127.2, 127.6, 130.3, 134.2, 134.4, 136.7, 146.1, 181.8, 178.7. IR (NaCI) 1725 cm", 1870 cm". HRMS: [M + H]+ = 372.0907, calculated for C19H18NO5S, 372.0906. Melting Point = 106-108°C. 203 (IV-6). To a flame dried 50 mL round bottom flask was added anhydrous EtOH (20.0 mL) and Na° (0.0740 g, 3.23 mmol). Once all the sodium metal reacted and the formation of bubbles ceased, diethyl 2,5-dioxoimidazolidin-4- ylphosphonate4 (0.763 g, 3.23 mmol, borrowed from Thu Nguyen) was added and stirred at room temperature for 30 min. Then III-5 (1.00 g, 2.70 mmol) was added to the mixture and stirred for 18 h at room temperature under a nitrogen atmosphere. The solvent was then removed and the crude residue was neutralized with 1% HCI and extracted with EtOAc (2 x 40.0 mL). The organics were washed with brine, dried using anhydrous sodium sulfate and concentrated. The crude material was purified by column chromatography (silica gel, 96% DCM; 4% MeOH) affording the product as an oil (mixture of diastereomers (8:1; E:Z)). Yield (1.11 g, 91.0%). 1H NMR (500 MHz), CDCI3: (Isomer B) 5 1.28 (t, J = 7.2 Hz, 3 H), 2.28 (s, 3H), 4.35 (q, J = 7.1 Hz, 2H), 7.16-7.22 (m, 3H), 7.29 (t, J = 7.3 Hz, 1H), 7.49 (d, J = 8.1 Hz, 1H), 7.78 (d, J = 8.5 Hz, 2H), 7.87 (s, 1H), 7.89 (d, J = 8.5 Hz, 1H), 8.32 (bs, 1H), 9.01 (bs, 1H); 13C NMR (125 MHz), CDCI3: 5 13.7, 21.5, 62.4, 111.7, 113.0, 113.5, 120.4, 123.9, 125.5, 126.7, 127.0, 127.6, 127.9, 130.0, 134.5, 134.7, 145.5, 153.9, 161.6, 165.7. (Isomer A) 1H NMR (500 MHz), CDCI3: 5 1.07 (t, J = 7.0 Hz, 3H), 2.28 (s, 3H), 4.15 (q, J = 7.0 Hz, 2H), 7.10-7.22 (m, 5H), 7.58 (s, 1H), 7.75 (d, J = 8.3 Hz, 2H), 7.83 (d, J = 8.2 Hz, 1H), 9.70 (bs, 1H); 130 NMR (125 MHz), CDC13: 8 13.8, 21.4, 81.8, 103.1, 112.4, 113.3, 120.0, 123.0, 124.3, 126.9, 127.3, 129.7, 130.5, 134.3, 135.0, 137.6, 144.7, 152.9, 181.9, 187.2. IR (NaCI) 3281 cm", 1779 cm", 1738 cm", 1657 cm' 1. HRMS: [M + H]+ = 454.1078, calculated for C22H20N3053, 454.1073. 204 (IV-7).10 To a flame dried 100 mL round bottom flask was added methylamine hydrochloride (1.00 g, 14.9 mmol) and dry DCM (55.0 mL). The mixture was cooled to 0°C and then DMAP (3.79 g, 31.1 mmol), indole-2- carboxylic acid (2.00 g, 12.4 mmol) and EDCI (2.86 g, 14.9 mmol) were added. The mixture stirred at 0°C under nitrogen for 4 h and then at room temperature overnight. The clear brown solution was washed with sat. sodium bicarbonate solution (1 x 60.0 mL), 1% HCI (1 x 60.0 mL) and brine (1 x 60.0 mL). The organics were combined, dried using anhydrous sodium sulfate and concentrated. A small amount of DCM was then added and the product precipitated out of solution to give an off white solid. Yield (1.57 g, 73.0%).‘H NMR (500 MHz), Acetone: 5 2.94 (dd, J = 1.3, 4.7 Hz, 3H), 7.05 (t, J = 7.5 Hz, 2H), 7.20 (dd, J = 7.1, 8.3 Hz, 1H), 7.56-7.61 (m, 2H), 7.81 (s, 1H), 11.01 (s, 1H); 13C NMR (125 MHz), Acetone: 5 26.2, 102.5, 113.0, 120.7, 122.3, 124.3, 128.7, 132.8, 137.6, 162.8. M.S: calculated for C10H10N20 (M+) = 174.2 and found (M+) = 174.3. (IV-8). To a flame dried 50 mL round bottom flask was added anhydrous DMF (3.50 mL) and dry DCM (25.0 mL). The solution was cooled to 0°C and then oxalyl chloride (0.280 mL, 3.16 mmol) was added and the solution became a thick white mixture. IV-7 (0.500 g, 2.87 mmol) was then added and the mixture turned from yellow to red. The solution stirred at room temperature for 8 h under nitrogen. The precipitate that formed was filtered off and washed with water (20- 30.0 mL) and extracted with EtOAc (5 x 30.0 mL). The organics were combined and dried using anhydrous sodium sulfate and concentrated to give the product 205 as a solid. Yield (0.398 g, 69.0%). 1H NMR (500 MHz), DMSO: 5 2.91 (d, J = 4.7 Hz, 3H), 7.28 (t, J = 7.1 Hz, 1H), 7.34 (t, J = 7.1 Hz, 1H), 7.55 (d, J = 8.0 Hz, 1H), 8.19 (d, J = 8.8 Hz, 1H), 9.82 (s, 1H), 10.30 (s, 1H), 12.77 (s, 1H); 130 NMR (125 MHz), DMSO: 5 26.1, 113.0, 113.9, 120.5, 122.9, 124.7, 126.5, 134.9, 138.0, 159.9, 186.5. IR (NaCI) 3182 cm", 1651 cm“, 1590 cm". M.S: calculated for C11H10N202 (M+) = 202.2 and found (M+) = 202.4. Anal. Calcd. For C11H10N202: C, 65.34; H, 4.98; N, 13.85. Found: C, 64.08; H, 4.80; N, 13.58. Melting Point = 257-259 °C. (IV-9). To a 25 mL round bottom flask was added IV-8 (0.306 g, 1.51 mmol) and EtOH (8.00 mL). Then thiohydantoin (0.193 g, 1.66 mmol) and piperidine (0.900 mL, 9.10 mmol) were added and the mixture was stirred under nitrogen at room temperature overnight. The mixture was filtered off and the precipitate was stirred in acetone for 5 min and then filtered. The precipitate was collected to give the product as a solid. Yield (0.390 g, 86.0%). 1H NMR (500 MHz), DMSO: 5 2.83 (d, J = 4.6 Hz, 3H), 6.95 (s, 1H), 7.17 (t, J = 7.4 Hz, 1H), 7.29 (t, J = 7.5 Hz, 1H), 7.47 (d, J = 8.3 Hz, 1H), 7.58 (d, J = 8.2 Hz, 1H), 8.14 (bs, 1H), 11.71 (bs, 1H), 11.98 (bs, 1H); 13C NMR (125 MHz), DMSO: 5 26.2, 105.5, 109.5, 112.5, 120.6, 120.8, 124.1, 126.1, 128.8, 131.1, 135.5, 161.6, 185.4, 177.6. IR (KBr) 3370 cm", 3194 cm", 1730 cm", 1883 cm", 1815 cm". M.S: calculated for C14H12N402S (M+) = 300.3 and found (M+) = 300.3. Anal. Calcd. For C14H12N402S: C, 55.99; H, 4.03; N, 18.65. Found: C, 54.91; H, 4.16; N, 17.95. Melting Point = 380 °C decomposes. 206 (IV-10). To a 25 mL round bottom flask was added lV-9 (0.250 g, 0.833 mmol), MeOH (5.00 mL) and a solution of NaOH (0.0370 g, 0.917 mmol in 0.300 mL H20). Then Mel (0.0600 mL, 0.917 mmol) was added and the reaction stirred under nitrogen over night at room temperature. The product precipitated out of solution after stirring overnight and the orange solid was filtered off. The crude product was recrystallized in MeOH to give pure product as a solid. Yield (0.252 g, 96.0%). 1H NMR (500 MHz), DMSO: 5 2.66 (s, 3H), 2.84 (d, J = 4.7 Hz, 3H), 7.14 (t, J = 8.1 Hz, 1H), 7.26 (t, J = 8.1 Hz, 1H), 7.42 (d, J = 8.0 Hz, 1H), 7.55 (s, 1H), 8.42 (d, J = 5.7 Hz, 1H), 9.11 (d, J = 8.0 Hz, 1H), 11.66 (s, 1H), 12.06 (s, 1H); 13C NMR (125 MHz), DMSO: 5 12.2, 26.1, 112.0, 112.5, 116.7, 120.6, 124.2, 125.1, 125.5, 135.2, 135.6, 135.9, 160.0, 161.8, 170.7. IR (KBr) 3267 cm' I, 1708 cm", 1827 cm", 1802 cm". HRMS: [M + H]+ = 315.0918, calculated for C15H15N402S, 315.0916. Anal. Calcd. For C15H14N402S: C, 57.31; H, 4.49; N, 17.82. Found: C, 57.10; H, 4.29; N, 17.58. Melting Point = 268-270°C decomposes. (IV-11). To a sealed tube was added IV-10 (0.420 g, 1.34 mmol) and THF (4.00 mL). Then ammonium hydroxide (4.00 mL) was added and the mixture was sealed and heated to 90°C for 16 h. The solution was cooled and the solvent was removed. Methanol (15.0 mL) was added and the product precipitated out. Yield (0.100 g, 26.0%). 1H NMR (500 MHz), DMSO: 5 2.79-2.95 (m, 3H), 6.90 (bs, 1H), 7.22 (t, J = 8.0 Hz, 1H), 7.43 (t, J = 8.2 Hz, 1H), 7.54 (d, J = 8.3 Hz, 1H), 7.74- 7.80 (m, 1H), 7.90-8.18 (m, 2H), 8.32-8.72 (bs, 1H), 10.42 (bs, 1H), 12.25 (bs, 1H); 13c NMR (125 MHz), DMSO: 8 27.5, 101.2, 101.5, 112.6, 120.13, 120.16, 207 121.3, 122.5, 123.8, 126.5, 129.6, 129.7, 132.70, 132.76, 139.4, 153.8, 161.9, 182.3, 188.2, 169.0. IR (KBr) 3488 cm'l, 3304 cm", 3218 cm", 1839 cm", 1808 cm", 1578 ml. HRMS: [M + H]‘“ = 284.1150, calculated for C14H14N502, 284.1147. Anal. Calcd. For C14H13N502: C, 59.36; H, 4.63; N, 24.72. Found: C, 57.30; H, 4.08; N, 24.33. Melting Point = 382-386°C decomposes. 208 IV.H References 1. 10. Sharma, V.; Hupp. C. D.; Tepe, J. J., Enhancement of chemotherapeutic efficacy by small molecule inhibition of NF-kB and checkpoint kinases. Curr. Med. Chem. 2007, 14, 1061-1074. Arnusch, C. J.; Pieters, R. J., Solid phase synthesis of vancomycin mimics. Eur. J. Org. Chem. 2003, 3131-3138. Mancini, |.; Guella, G.; Debitus, C.; Dubet, D.; Pietra, F., lmidazolone and imidazolidinone artifacts of a pivotal imidazolthione, zyzzin, from the poecilosclerid sponge Zyzzya massalis from the Coral Sea. The first thermochromic systems of marine origin. Helvefica Chimica Acta 1994, 77, 1886-94. Meanwell, N. A.; Roth, H. R.; Smith, E. C. R.; Wedding, D. L.; Wright, J. J. K., Diethyl 2,4-dioxoimidazolidine-5-phosphonates: Homer-Wadsworth- Emmons reagents for the mild and efficient preparation of C-5 unsaturated hydantoin derivatives. J. Org. Chem. 1991, 56, 6897-904. Sharma, V.; Tepe, J. J., Potent inhibition of checkpoint kinase activity by a hymenialdisine-derived indoloazepine. Bioorg. Med. Chem. Lett. 2004, 14, 4319-4321. Chacun-Lefevre, L.; Beneteau, V.; Joseph, B.; Merour, J.-Y., Ring closure metathesis of indole 2-carboxylic acid allylamide derivatives. Tetrahedron 2002,58, 10181-10188. Vilsmeier, A.; Haack, A., Action of phosphorus halides on alkylformanilides. A new method for the preparation of secondary and tertiary p-aIkylaminonobenzaldehydes. Ber. Dtsch. Chem. Ges. 1927, 608, 119-22. http://www.millipore.com/druqdiscovery/dd3/KinaseProfiler. Yang, L.; Deng, G.; Wang, D.-X.; I-Iuang, Z.-T.; Zhu, J.-P.; Wang, M.-X., Highly Efficient and Stereoselective N-Vinylation of Oxiranecarboxamides and Unprecedented 8-endo-Epoxy-arene Cyclization: Expedient and Biomimetic Synthesis of Some Clausena Alkaloids. Org. Lett. 2007, 9, 1387-1390. Dutcher, J. D.; Kjaer, A., The C11H8NZOS degradation product of gliotoxin. J. Am. Chem. Soc. 1951, 73, 4139-41. 209 67 ml M“ B" Uiii I Hi 8” u ”it lill I “3 03062 78 129 lllllllllllllllllllll