LIBRARY Michigan State University 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 6/01 c:/CIRC/DaIeDue.p65-p.15 LEWIS ACID MEDIATED IN SITU GENERATION OF MUNCHNONES AIMED AT DIVERSITY ORIENTED SYNTHESIS OF NITROGEN CONTAINING DIHYDROHETEROCYCLES By Satyamaheshwar Peddibhotla A DISSERTATION Submitted to Michigan State University in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY Department of Chemistry 2004 ABSTRACT LEWIS ACID MEDIATED IN SITU GENERATION OF MUNCHNONES AIMED AT DIVERSITY ORIENTED SYNTHESIS OF NITROGEN CONTAINING DIHYDROHETEROCYCLES By Satyamaheshwar Peddibhotla This dissertation describes the development of Lewis acid mediated/catalyzed generation of mesionic 5-oxazolones (mfinchnones) from amino acid derived azlactones. Milnchnones are highly reactive cyclic azomethine ylides, a class of 1, 3-dipoles used extensively for generation of five-membered nitrogen containing heterocycles. Our mild method allows for highly stereoselective cycloadditions with different dipolarophiles. Furthermore, it allows isolation of dihydroheterocyclic scaffolds without problems of decarboxylation, aromatization and isomerizations normally encountered in cycloadditions of traditionally used N-alkylated mitnchnones. The generation of diverse small molecular scaffolds with potential biological activity is an ongoing theme in our group. Five-membered nitrogen containing rings are at the core of many natural products and prospective libraries of drug discovery programs. On account of the structural rigidity and the three dimensional disposition of functional groups, dihydroheterocyclic five-membered rings that we have accessed are perfectly suited for the aim of drug discovery. They have a different shape profile from aromatic heterocycles generally encountered in drug discovery programs and as such might interact with entirely new therapeutic targets. Chapter I lays a strong foundation for the need of such molecules. Chapter II includes a survey of azlactones as a viable template for diversity oriented transformations. However, stereochemical diversity has not been efficiently achieved from azlactones. Stereochemical diversity is extremely important in screening for new ligands which can display functional groups for interactions with macromolecules that govern life as well as disease. Our proposed route to dihydroheterocycles from milnchnones is discussed in the context of other stereoselective methods that employ lewis acids in l, 3-dipolar cycloadditions of closely related nitrogen ylides. Discovery of mild in situ method of generation of mtinchnones (cyclic azomethine ylides) from azlactones using Lewis acids and their use for stereoselective synthesis of nitrogen containing dihydroheterocycles; 2- imidazolines and Al-pyrrolines are described in Chapter III and IV respectively. The scaffolds are obtained with high diastereoselectivity (stereochemical) and are amenable to diverse substitution (appendage diversity) and in many ways complement other methods towards these scaffolds. The dihydroheterocyclic scafi‘olds offer unexplored territory in terms of their structure (3-D disposition of functional groups) and their function (biological activity and catalysis). Chapter V describes selective and enhanced toxicity of the combination of imidazolines with chemotherapeutic drugs (camptothecin and cis-platin) in cancer cells. The mechanism and potential of this novel effect is being studied by other researchers in the group. The imidazoline scaffolds can catalyze a novel stereoselective reaction of ketenes with N-acylimines. We have also explored their use as analogs of amino alcohol ligands for diethyl zinc additions to electrophiles. To Vasudha iv ACKNOWLEDGEMENTS This dissertation would be incomplete without the mention of all those who contributed to my successful graduate career. Being his first student to graduate, both J etze and I have gone through times of learning and teaching. Jetze gave me a lot of independence to explore my own ideas, through out my graduate career in this group. His confidence in me has helped me achieve goals that might have seemed out of reach at that point of time. Whether I was lucky for his career or he for mine is a topic that is feverishly discussed from time to time. Time probably will be a better judge. I would like to thank Profs. Borhan, Wulff, and Dellapena for serving on my committee and their advise on several occasions. In fact, Babak (Dr. Borhan) is more of a fiend than a professor to me. I admire his ability to be able to ask questions ranging from specific to the more philosophical ones. I have great respect for Prof. Wulff, especially his composure and how he deals with people irrespective of who they are. Despite of his busy schedule Prof. Dellapenna has been extremely kind, accommodating and understanding. We had a very productive association while working on the biosynthesis of vitamin E. I want to thank Prof Hart for a summer fellowship in recognition of my graduate work. I also want to thank Prof. Maleczka for help with different things at different times and mainly for CEM 852. All the past and present group members have been good friends. .I in and Jayakurnar were good work colleagues. I enjoyed their company working late hours in the lab. They kept me in good humor through good and bad times. I especially want to thank Dr. Rui Huang for his expertise on X-ray crystallography. The confidence with which we could talk about the stereochemical issues throughout this work relied on the X-ray structures that Dr. Huang solved. Dr. Long Le, Dr. Kermit Johnson and Dr. Daniel Holmes have been helpful with the NMR spectroscopy. Nancy Lavrik, was ever helpful and kind soul and you can only hope to find more people like her through life. I would like to thank Sundar, Radha, RG, Pops, Malini, Manish, Param Edith, Mapitso, Ben and Chryssoula; friends who enriched my experience with help, support and encouragements to difi‘erent degrees and at different times during the last few years. I am grateful for the support of my parents and siblings, who have been crucial to my success. Their love and faith has helped me overcome all hurdles and pursue my goals tirelessly. Finally, I am grateful to god that I have Vasudha as the love of my life. Those long hours of lab work were made easy in her company. I am really fortunate that I can discuss chemistry and biology with Vasudha with the same ease as personal issues. She is a great critic as well as someone who encourages and helps all the way to the end. I can only hope that I have learnt from her “Never say I give up” attitude towards work and everything she does. I am really grateful to her for proof reading and help with formatting of this dissertation and the defense. vi TABLE OF CONTENTS (Images in this dissertation are presented in color) List of Tables ....................................................................... xi List of Schemes .................................................................... xii List of Figures ..................................................................... xv Key to Symbols and Abbreviations ............................................. xix CHAPTER 1 Small Heterocyclic Scaffolds and Drug Discovery ............................. l A. Drug discovery process ................................................ 1 B. A small molecule revolution .......................................... 2 C. Chemical genetics brings small molecules center stage .......... 4 D. Target-oriented synthesis (TOS) (Trial and Error method). . .7 E. Combinatorial chemistry (Trial and Selection method) ........... 9 F. Number game (HTS) vs. quality of libraries ....................... 12 G. Diversity deficit in combinatorial libraries vs. natural products ...................................................................................... 16 H. Rescuing Combinational Chemistry ................................. 19 1. Chemical diversity and biological space ............................. 22 J. Diversity oriented synthesis(DOS) ............................................. 25 1) Appendage Diversity ........................................... 28 2) Stereochemical Diversity ...................................... 28 3) Skeletal Diversity ............................................... 29 i. Differentiation ........................................... 29 ii. folding ................................................... 39 4) Integrated Forward-Synthetic Analysis for Generating Complexity and Diversity ................................................. 42 L. Small molecule heterocyclic scaffolds .................................... 43 M. Conformational constraint/rigid scaffolds ................................... 45 N. 1, 3-dipolar cycloadditions and confonnationally constrained heterocycles ............................................................... 45 0. References ..................................................................................... 48 CHAPTER II Azlactones to novel dihydroheterocyclic scaffolds ....................................... 56 A. Introduction ................................................................................... 56 1) Easily accessible starting materials .................................... 57 2) Pluripotency ....................................................................... 59 3) Skeletal diversity through azlactones ................................ 61 i. Differentiation/ Intermolecular reactions ................ 61 ii. Folding / Intramolecular reactions ........................ 63 vii B. 1, 3-Dipolar cycloadditions and stereochemical diversity ............. 66 C. Lack of stereochemical diversity in milnchnone cycloadditions...67 D. Potential of Lewis acid mediated generation of mtlnchnones & synthesis of novel dihydroheterocyclic scaffolds ......................... 70 E. Lewis acid mediated cycloadditions of nitrile ylides & azomethine ylides .......................................................................... 74 F. Nitrile ylides and Lewis acids ........................................................ 75 1) Synthesis of 2-oxazolines ................................................... 75 2) Synthesis of Az-pyrrolines .................................................. 76 3) Synthesis of 2-imidazolines ................................................ 77 G. Azomethine ylides and Lewis acids .............................................. 78 1) Synthesis of 2-imidazolines ................................................ 78 2) Synthesis of pyrrolidines .................................................... 79 3) Synthesis of dihydroheterocycles ....................................... 83 H. Current work .................................................................................. 84 1. Experimental Section ...................................................................... 86 J. References ....................................................................................... 93 CHAPTER III Silicon mediated diastereoselective multi-component synthesis of imidazolines ..................................................................................................... 99 A. Biological Importance of 2-Imidazolines and Imidazoles .............. 99 B. Chemical significance of imidazolines ........................................... 103 C. Lewis acid mediated synthesis of imidazolines .............................. 104 D. Lewis acids for in situ generation of milnchnones and Cycloadditions ................................................................................ 108 E. Possible mechanism for synthesis of imidazoline ........................... 112 F. Diversity .......................................................................................... 1 16 G. Proposed origin of Diastereoselectivity .......................................... 119 H. Synthetic methods for chiral imidazolines ...................................... 121 1) Resolution of enantiomers ................................................... 121 2) Use of Chiral auxillaries ...................................................... 123 i. Chiral acid chlorides ................................................. 123 ii. Chiral amino acid derived R1 .................................. 124 iii. Chiral amines as auxillaries .................................... 125 1. Synthesis of diamino acids and other scaffolds ............................... 127 1) 2-unsubstituted imidazolines ............................................... 128 2) 2-trifluoromethylimidazolines ............................................. 128 3) 2-amino-5-oxazolones ......................................................... 129 J. Ring opening of 2-phenyl imidazolines ........................................... 130 K. Synthesis of Imidazoles from 2-imidazoline-4-carboxylic acids...132 L. Experimental Procedure .................................................................. 134 M. References ...................................................................................... 160 viii CHAPTER IV Silver acetate catalyzed exo-selective synthesis of Al-pyrrolines and derived pyrrolidines ..................................................................................................... 165 A. Biological and chemical significance of A'pyrrolines .................. 165 B. Access to Al-pyrrolines using mttnchnone-alkene Cycloadditions .............................................................................. 166 C. TMSCI (Lewis acid) mediated mild protocol to A'-pyrroline-5- carboxylates .................................................................................. 169 D. Screening of additional Lewis acid for synthesis of AI -pyrrolines ................................................................................. 1 73 E. Diversity in AgOAc catalyzed synthesis of Al-pyrroline-S- carboxylates .................................................................................. 175 F. Probable origin of diastereoselectivity ......................................... 179 G. Expanding the diversity of A'pyrroline templates ........................ 182 H. Synthesis of pyrrolidines and DOS .............................................. 183 1. Importance of diastereoselective diversity-generating pyrrolidine syntheses .................................................................... 184 J. Extension of diastereocontrol and synthesis of exo pyrrolidines..187 K. Enantiocontrol in AI-pyrroline synthesis ...................................... 190 L. Experimental Section .................................................................... 191 M. References .................................................................................... 206 CHAPTER V Applications of Imidazoline Scaffolds in Therapy and Organocatalysis ....... 211 A. Sensitization of Tumor Cells towards Chemotherapy ................. 211 1) Enhancing the activity of Camptothecin by Novel Imidazolines ..................................................................... 21 1 2) Chemotherapy and chemoresistance — camptothecin & cis-platin ....................................................................... 212 3) NF-KB, the master key to cell growth, differentiation & apoptosis ............................................... 213 4) NF -KB as a target for drug design .................................... 215 5) Imidazoles, kinase inhibition and NF «B ........................ 216 6) From Imidazoles to Imidazolines ..................................... 218 7) Sensitization of leukemia cells toward CPT .................... 218 i. Imidazolines and Camptothecin ................................. 218 ii. Inhibition of camptothecin induced NF-KB -DNA binding by imidazoline l ............................................ 220 iii. Effect of imidazoline l on I-xBa phosphorylation ..... 223 8) Significance ...................................................................... 225 ix B. Catalytic Asymmetric Synthesis of 4, 5-dihydro-1, 3-oxazin -6-ones. Precursors to 0., B-dialkyl-B-amino acids using imidazoline-4-amides as novel organocatalysts .................................. 227 1) Introduction to B-amino acids ...................................... 227 2) Nucleophilic catalysts and stereocontrol in reaction of ketenes and imines ................................................... 228 3) Imidazolines as nucleophillic catalysts ........................ 231 4) Preparation of chiral imidazoline-4-amides catalysts ........................................................................ 234 5) Three component synthesis of 4, 5-dihydro-1, 3- oxazin-6-ones ............................................................... 235 6) Problems ....................................................................... 239 7) Modifications ............................................................... 240 8) Three component to a two-component synthesis of dihydroxozinones ..................................... 241 C. Chiral 4-hydroxymethyl Imidazolines as ligands for diethyl zinc addition ......................................................................................... 244 1) Introduction .................................................................. 244 2) Imidazolines as N-heterocyclic carbine ligands ........... 244 3) Imidazolines as nitrogen ligands .................................. 245 i. 2-hydroxymethylimidazolines ligands for diethyl zinc additions to aldehydes ............................. 248 ii. Our 4-hydroxymethylimidazolines as ligands for diethylzinc additions ............................................. 250 D. Experimental Section ..................................................................... 255 E. References ..................................................................................... 271 APPENDIX .................................................................................................... 281 Efficient two-step synthesis of methylphytylbenzo quinones: precursor intermediates in the biosynthesis of vitamin E .................... 282 LIST OF TABLES (Images in this dissertation are presented in color) Table III-1. Variation of R3 and R4 groups .................................................... 118 TableIII-2. Diversity of various groups on the imidazoline scaffold ............. 119 Table IV-l. TMSCI mediated synthesis of Al-pyrroline ................................ 172 Table IV~2. Lewis acid screen for synthesis of A'pyrrolines ......................... 174 Table IV-3. Cycloaddition of various alkenes ................................................ 176 Table IV-4. Variation of substituents at C2 and C5 in Al-pyrrolines ............... 179 Table V-l . Synthesis of dihydroxazinones using imidazoline based catalysts. ......................................................................................................... 237 Table V- 2. Chiral 4-hydroxymethy imidazoline ligands in diethyl zinc additions .................................................................................................. 254 xi LIST OF SCHEMES (Images in this dissertation are presented in color) Scheme [-1. Skeletal diversity from isoquinoline intermediate on solid phase....33 Scheme [-2. Squaric acid as a versatile precursor to various compound shapes..34 Scheme I-3. Allenes and transition metals: a diverging approach to Heterocycles ........................................................................................................ 35 Scheme [-4. A combinatorial scaffold approach based upon a multi-component reaction ................................................................................................................. 36 Scheme [-5. Diversity-oriented synthesis of azaspirocycles ................................ 37 Scheme 1-6. Cross-conjugated trienes for diene-transmissive cycloadditions ..... 37 Scheme [-7. Skeletal diversity via a branched pathway ....................................... 38 Scheme I-8. A skeletal diversity generating folding process ............................... 41 Scheme 1-9. Generation of skeletal complexity and diversity ............................. 43 Scheme 11-] . Synthesis of dihydroheterocycles from milnchnones ..................... 71 Scheme II-2. Lewis acid mediated insitu generation of mtlnchnones from azlactones l, 3 dipolar cycloaddition with dipolarophile .................................... 85 Scheme III-1. Addition of the tosylimines leads to loss of C02 and PhSOzH to gain aromaticity ............................................................................................... 1 Scheme III-2. Staudinger reaction imines and ketenes generated from Azlactones ........................................................................................................... 106 Scheme III-3. Lewis acid mediated generation of mttnchnone & cycloadditions ................................................................................................. 107 Scheme III-4. Lewis acid screen for the synthesis of Imidazolines ................... 112 Scheme III-5. Nitrilium ion intermediate is not involved in the formation of lrnidazolines .................................................................................................... 113 Scheme III-6. Generation of azomethine ylides by silatropy .............................. 114 Scheme III-7. Proposed mechanism for the formation of imidazolines via the xii 1, 3 dipolar cycloadditions .................................................................................. 115 Scheme III-8. Origin of Diastereoselectivity ...................................................... 120 Scheme III-9. Resolution of racemic imidazolines using (-) Menthol ................ 122 Scheme III-10. Synthesis of racemic imidazoline III-2-2, and (-) S, S- III-2-l and (+) R. R- [11-2-1 ............................................................................................ 124 Scheme III-11. Asymmetric induction by chiral acid chloride ........................... 125 Scheme III-12. Amino acid at R. position as a chiral template .......................... 125 Scheme III- 13. Use of chiral sulfinimine in milnchnone cycloaddition ............ 127 Scheme III-14. Extension of methodology for other templates .......................... 128 Scheme III-15. Synthesis of 2-aminoazlactones and 2-aminoimidazolines ........ 130 Scheme III-16. Tautomers of 2-arninoazlactones ............................................... 130 Scheme III-17. Reductive ring opening of imidazolines to diamino acids ......... 131 Scheme III-18. Synthesis of imidazoles from imidazolines ................................ 133 Scheme IV-l . Lewis acid mediated mild protocol to Al-pyrroline- 5-carboxylates. .................................................................................................... 169 Scheme IV-2. Isomerization of cis A'-pyrroline to trans Al-pyrroline .............. 178 Scheme IV-3.(a) proposed syn-dipole orientation for acyclic azomethine ylides derived from imines of a-amino acid esters ............................................. 180 Scheme IV-3. (b) munchnbne (cyclic azomethine ylide) locked as an anti-dipole ........................................................................................................... 180 Scheme IV-4. Ring opening and isomerization of cycloadducts ....................... 182 Scheme IV-S. Ring opening with different nucleophiles ................................... 183 Scheme IV-6. Extension of diastereocontrol to all four centers of Al-pyrroline ........................................................................................................ l 88 Scheme IV-7. Asymmetric synthesis A'-pyrroline ............................................ 19] Scheme V-l. Multicomponent synthesis of imidazolines ................................. 232 xiii Scheme V-2. Failed attempt to B-lactams using racemic imidazoline-4- carboxylates ........................................................................................................ 233 Scheme V-3. B-lactams from N-tosyl-a-iminoester using racemic imidazoline catalyst .......................................................................................... 234 Scheme V-4. Synthesis of chiral imidazoline-4-amide catalysts ..................... 234 Scheme V-S. Proposed mechanism for catalytic synthesis of Dihydrooxazinones ........................................................................................... 239 Scheme V-6. Synthesis of 2-aryl dihydrooxazinones from N-acylimines ....... 242 Scheme V-7. Synthesis of chiral 4-hydroxymethyl imidazolines from racemic imidazoline-4-carboxylic acid (III-la) ............................................. 252 xiv LIST OF FIGURES (Images in this dissertation are presented in color) Figure Ma. The drug discovery process ........................................................ 2 Figure I-lb. The drug discovery funnel .......................................................... 3 Figure I-2. Chemical Genetics ........................................................................ 4 Figure I-3a. Target Oriented Synthesis (TOS) ............................................... 8 Figure I-3b. Representation of a single target in chemical Space ................... 8 Figure I-4. High Throughput Screening (HTS) ............................................. 13 Figure I-5a. New drugs approved by FDA vs. R&D spending in billions ..... 15 Figure I-Sb. Number of US Food and Drug Administration (FDA) new drug approvals (N DA, blue), new molecular entities (N ME, red) and NME with significant improvements over existing products (green), between the years 1998 and 2002 ....................................................................................... 15 Figure I-6. Comparison of diversity ............................................................... 18 Figure [-7. Goal in medicinal and combinatorial chemistry.- .......................... 18 Figure I-8. Matching of biological and chemical structure spaces ................. 23 Figure I-9. Structural diversity is not identical to biological diversity ........... 23 Figure I-10. Structural Similarity is not identical with biological similarity..24 Figure [-1 1. Comparison of TOS, medicinal chemistry and DOS ................. 26 Figure I-12. Two general approaches for planning pathways to generate skeletal diversity ............................................................................................. 30 Figure I-13. Nature’s Synthesis of Secondary Metabolites ............................ 31 Figure I-l4. Flexible core structure libraries vary functionalities as well as orientation of display ...................................................................................... 32 Figure 11-1. Isomeric Oxazolones and structures ............................................ 56 Figure 11-2. Cyclodehydration of N-acyl-a-aminoacids to azlactones ............ 57 XV Figure 11-3. Syntheses of milnchnones from basic building blocks ................ 59 Figure 11-4. Diverse reactive sites and intermediates accessible from Azlactones ...................................................................................................... 60 Figure 11-5. Cyclocondensation reactions of azlactones to generate diverse heterocycles .................................................................................................... 61 Figure 11-6. Cycloadditions of mitnchnones to generate diverse heterocyclic skeletons. ...................................................................................................... 62 Figure 11-7. Alkenes as intramolecular diversity elemnts (6 elements) ......... 64 Figure 11- 8. Alkynes as intramolecular diversity elemnts (0’ elements) ........ 65 Figure 11- 9. Alkenes as intramolecular diversity elements in condensations (0 elements) ................................................................................................... 66 Figure II-lO. Synthesis of pyrroles from mfinchnones ................................... 68 Figure 11-1 1. Synthesis of imidazoles from mfinchnones ............................... 69 Figure II-12. Synthesis of bicyclic scaffolds fi'om cycloaddition of isomfinchnones ............................................................................................... 72 Figure 11-13. Synthesis of isoxazolidines as mimic of transcription regulators ........................................................................................................ 74 Figure II-14. Synthesis of oxazolines from oxazoles ..................................... 76 Figure II-15. Synthesis of Az-pyrrolines from isocyanoacetates .................... 77 Figure 11-16. Synthesis of 2-imidazolines from isocyanoacetates .................. 78 Figure 11-17. Synthesis of 2-imidazolines from azomethine ylides ................ 79 Figure 11-18. Synthesis of pyrrolidines from a—iminoesters ............................ 80 Figure II-l9. Synthesis of pyrrolidines from aziridines and electron-rich alkenes ............................................................................................................. 81 Figure II-20. Synthesis of dihydroheterocycles from soft enolization of imidates ............................................................................................................ 83 Figure III-1. Biological activity of molecules with 2-imidazoline nucleus ..... 100 xvi Figure III-2 (a & b). p53 bound to MDM2; key residues for binding ............. 102 Figure III-2 (c) .Overlay of p53 residues and nutlin ...................................... 102 Figure III-2 (d) The bromo phenyl moiety of nutlin deep in the groove of MDM2 ............................................................................................................ 102 Figure III-2 (e) Representative structure of a nutlin ....................................... 102 Figure III-3. Generation of imidazolium salt through Pd-catalysis ................ 108 Figure III-4. (a) 1H spectra for a representative B-lactam (III-A) and imidazoline (III-l-l) ....................................................................................... 109 Figure III-4. (b) '3 C spectra for a representative B-lactam (III-A) and imidazoline (III-l-l) ....................................................................................... 110 Figure III-4. (c)X-ray crystal structure of III-l-land 111-1 -7 .......................... 111 Figure III-5. Soft enolization vs. 1, 2-Prototropy ........................................... 115 Figure III-6. Chem3D models for minimized structures of 2, 4-dimethyl azlactone, 2-Methyl-4-phenyl azlactone, 4-methyl 2-phenyl azlactone .......... 121 Figure III-7. X-Ray crystal structure of (+) (R, R) - [11-2-1 ............................ 123 Figure III-8. Use of chiral sulphinimes in azomethine ylide cycloadditions...126 Figure III-9. Synthesis of vicinal diamine from imidazolines ......................... 128 Figure III-10. Biological activity of molecules with imidazole nucleus ......... 132 Figure IV-l. Biologically active pyrrolines and pyrrolidines .......................... 165 Figure IV-2. Dipolar cycloaddition reactions of N-alkylated mtlnchnones ..... 167 Figure IV-3. Primary cycloadducts isolated fiom intramolecular Cycloadditions ................................................................................................. 168 Figure IV-4. Primary cycloadducts isolated from intermolecular cycloadditions .................................................................................................. 168 Figure IV-S. Azomethine generation by N-silylation ...................................... 173 Figure IV- 6. X-ray crystal structure of IV-l-6 ............................................... 177 xvii Figure IV -7. Calculated electrostatic isopotential surfaces for exo- and endo- approach of dicyanocyclobutene ............................................. 181 Figure IV- 8. Synthesis of pyrrolidines from a-iminoesters .................... 185 Figure IV-9. Exo selectivity tuned using bulky phosphine ligands ................ 187 Figure IV-lOa. X-ray crystal structure of IV-l-S, IV-1-10 ............................ 189 Figure IV-lOb. X-ray crystal structure of IV-l-13 ......................................... 190 Figure V-l. General NF -KB activation pathway in eukaryotic cells .............. 214 Figure V-2. Inhibition of NF-KB might play a key role in the pathways involved in inflammation, diabetes and cancer ............................................. 217 Figure V-3. Imidazoline that showed maximum potentiation of apoptosis....218 Figure V-4. Sensitization of CEM cells to camptothecin by imidazoline 1, measured over 48 hours .................................................................................. 220 Figure V-5. Inhibition of CPT activated NF -I8 —"’ O Retro-synthetic Anubwis El —>CEI (a) ‘ descriptor 2 Ades/ptofi descriptor 1 (b) Figure [-3. (a) Target Oriented Synthesis (TOS); (b) Representation of a single target in chemical space. Adapted from Ref. 52 Prior to this, strategic solutions to the problems of synthesizing different target structures were developed on a case-by-case basis. Retro-synthetic concepts and thinking depend on the existence of a defined target structure. Synthesis pathways in TOS are linear and convergent, and they are planned in the reverse-synthetic direction by using retro-synthetic planning, which aims to move in the direction of complex to simple. This problem-solving technique involves the recognition of key structural elements in reaction products, rather than reaction substrates, that code for synthetic transformations. Repetitive application of this process allows a synthetic chemist to start with a structurally complex target and find a structurally simple compound that can be used to start a synthesis. The basic subunit of retro- synthetic planning is ‘to transform’, that is, the theoretical transformation of a product into a substrate by formally performing a chemical reaction in the reverse-synthetic direction. To make use of a transform in retro-synthetic analysis one must first identify the corresponding “retron”, that is, the enabling structural subunit (“keying element”) that permits its application, in the chemical target. Structurally simplifying transforms are critical in TOS when devising a retro- synthesis for a complex target structure, and iterative application of these transforms can lead to a plan for an efficient synthesis. Reactions that join together two different building blocks, called fragment-coupling reactions, are of great importance. Reactions that generate structural complexity stereoselectively (e.g. oxy-Cope and Diels-Alder reactions) are also of considerable value and are used widely in target-oriented organic synthesis. TOS does not share the aim of accessing diversity. E. Combinatorial chemistry (Trial and Selection method) Peptide libraries constituted the first modular arrays of functionality to be explored in a prospecting fashion, with the vision that peptide leads could be transformed into more pharmacologically active molecules. Solid phase peptide synthesis was first introduced to overcome the technical challenge of performing iterative couplings to yield long polypeptide chains. The nascent polypeptide chain is immobilized in this method, most commonly to spherical polystyrene beads, allowing coupling reagents to be added in high molar excess and by products (including the unused reagents) to be removed simply by washing the insoluble beads. This led to the generation of large combinatorial libraries of peptides'3 and oligonucleotides, '4 which were then screened against a receptor or enzyme to identify high-affinity ligands or potent inhibitors, respectively. These and the oligomeric systems that followed them, such as peptoids, '5 ' '7 were assembled readily, even robotically, on polycarbamates '6 and 'azatides solid-phase and their input materials were available in great variety.18 While these studies clearly demonstrated the power of library synthesis and screening strategies, peptides and oligonucleotides generally have poor oral activities and rapid in viva clearing times, and therefore their utility as pharmacological probes or therapeutic agents was often limited.'9 Moreover, the formidable challenge of turning a flexible ligand with modest affinity into a potent lead has limited their relevance. Although solid phase peptide synthesis was only recently adapted to non-peptidic small molecules (<600-700 molecular weight), which have favorable 2° simplification of the purification of synthetic pharmacokinetic properties, intermediates on the solid phase method has led to an increase in synthetic productivity. Again taking a lead from solid phase peptide (and oligonucleotide) synthesis, 2‘ solid phase syntheses have been performed in parallel '5’ 22 i. e., similar reactions are performed, but the structures of the building blocks in key fi'agment-coupling steps are varied. Solid phase, parallel synthesis is an example 10 of what is commonly referred to as combinatorial synthesis and is most commonly used by medicinal chemists in pharmaceutical companies and universities to synthesize a focused library of related compounds sharing structural features necessary for binding to a preselected protein target, allowing the general principles of retro-synthetic analysis to be applied readily. A second variation of solid phase synthesis, one that extends it beyond a mere purification technique, can provide a staggering increase in the ability of organic synthesis to produce collections of small molecules. This potential was realized originally in peptide synthesis with the invention of the split-and-pool (split-pool) strategy of synthesis.23 The strategy has more recently also been used in organic synthesis, resulting in structurally complex and diverse libraries of synthetic small molecules.24 In this method, a collection of beads is split into reaction vessels that subsequently each receive a unique set of reagents, for example, one of a collection of building blocks. Cycles of pooling, resplitting, and further chemistry then result in large collections of compounds that are spatially segregated on unique beads. Split-pool synthesis is referred to as the “one bead-one compound” approach, and it is analogous to genetic recombination. Encoding methods, which are analogous to the genetic code, have been developed that record the chemical history of the synthetic compounds, allowing the structures of compounds selected in screens to be inferred.25 Due to the extensive time period required for the identification of drug-like candidates, combinatorial chemistry is now firmly integrated in the drug discovery enterprise for both lead identification and lead optimization.26 By 11 applying combinatorial chemistry, it is possible to obtain large sets of compounds over short periods of time, by applying parallel high-throughput synthesis. " 4’ 27 The source of the starting or lead compounds can vary and may include a natural product, a known drug, or a rationally designed structure developed from a mechanistic hypothesis and/or a crystal structure of a macromolecule of interest (focused libraries) or simply exhibiting some intuitively appealing combination (prospecting libraries) of molecular weight, polarity, conformational constraint, novelty and so on.'8 Libraries of compounds that are deemed at the outset to be more 'drug-like' are more favored, comprising known pharmacological motifs. Library synthesis can be accomplished efficiently using solid-phase synthesis to append different sets of building blocks to a common molecular skeleton which facilitates binding to a preselected protein target.22 Retro-synthetic planning is used in this context to devise pathways to the target structure that permit the addition of diverse sets of building blocks during the actual synthesis. If this common skeleton contains multiple reactive sites with potential for orthogonal 23' 28 can be used functionalization, the powerful technique of split-pool synthesis to access all possible combinations of building blocks (namely, the complete matrix) efficiently. F. Number game (HTS) vs. quality of libraries Since the human genome sequence has been solved, the pharmaceutical industry has more than 3500 potential drug targets while previously only about 400 targets were pursued.29 The advent of genomic sciences, rapid DNA sequencing, combinatorial chemistry, cell-based assays, and automated high-throughput 12 screening (HTS) has led to a "new" concept of drug discovery (Figure I-4). In this new concept, the critical discourse between chemists and biologists and the quality of scientific reasoning are sometimes replaced by the magic of large numbers. Large numbers of hypothetical targets are incorporated into in vitro or cell-based assays and exposed to large numbers of compounds representing numerous variations on a few chemical themes or, more recently, fewer variations on a greater number of themes in high-throughput configurations. STOCK PLATE ADD COMPOUND INCUBATE CELLS DETECT CELLULAR CHANGE OF MICROPLATE WITH INTERESTPHENOTYPE CELLS ANTIBODY CYTOBLOT ASSAY MICROSCOPY REPORTER ASSAY Figure 1-4. High Throughput Screening (HTS).Adapted from Ref. 10 It was hoped that this experimental design would be suitable to identify many substances, which can modify the targets in question. Many such "hits"-- compounds that elicit a positive response in a particular assay--would then give rise to more leads, i.e., compounds that continue to Show the initial positive response in more complex models (cells, animals) in a dose-dependent manner. Eventually, the number of compounds also would increase. Data points generated by large screening programs at a pharmaceutical company have amounted to roughly 200,000 at the beginning of the 19905. Data points are screening results describing the effect of one compound at one concentration in a particular test. This Figure I- rose to 5 to 6 million at the middle of the decade and is presently approaching or even passing the 50-million mark.33 The goal for such an endeavor, in part, was to obviate the need for a rational approach completely. A large enough library with the right HTS assay promised the ultimate brute-force method for obtaining bioactive compounds without biologists and organic chemists getting in the way.30 However, the pharmaceutical industry is currently facing a major challenge (Figure 1- 5a) As measured by the number of new compounds entering the market place, the top 50 companies of the pharmaceutical industry collectively have not improved their productivity during the 19905.3| Two-thirds of the prescription drugs approved by the US Food and Drug Administration (FDA) between 1989 and 2002 were modified versions of existing medicines or identical to drugs already on the market (Figure I- 5b). Over the last five years the number of ’new molecular entities' (NME) approved for use as drugs by the FDA has steadily decreased. In 2002, only 17 NME were approved, and of those only 7 were classified as being significant improvements over existing products.” 32’ 33 14 1996 1997 1998 1999 2000 2001 - Total R&D spent + New Drug Approvals] Figure I-Sa. New drugs approved by FDA vs. R&D spending in billions. Adapted from www.mindfull .o GE/2004/Dru -I dust -FaIIS' Peter Landers /W all Street Journal 24 Feb 2004. 100 90 80 . 7o . 60 50 4o 30 1998 1999 2000 2001 2002 Figure I-5b. Number of US Food and Drug Administration (FDA) new drug approvals (NDA, blue), new molecular entities (NME, red) and NME with significant improvements over existing products (green), between the years 1998 and 2002. Adapted from Ref.29 G. Diversity deficit in combinatorial libraries vs. natural products Although the reasons underlying this apparent lack of productivity remain unclear, it may in part reflect significant deficiencies in the types of chemical structures generated using combinatorial approaches. Combinatorial chemistry allows for the synthesis of vast numbers of compounds; indeed, millions of compounds are realizable by one chemist on their own in a few weeks using split- pool combinatorial techniques.34 However, the limitation is that the compounds produced have limited structural diversity. Traditionally, natural products have been a major source of new drugs, and many successful drugs were originally synthesized to mimic the action of molecules found in nature.35 One of the unquestionable hallmarks of natural products is their exquisite level of three-dimensional sophistication. The diversity of ring skeletons, and the way in which they present functional groups topologically, and often provide highly specific biological activities. This follows from the proposition that essentially all natural products have some receptor binding capacity.36 Natural molecules, however, differ substantially from synthetic ones (Figure 1-6). These differences appear to be amplified when the products of combinatorial synthesis are considered. Although an important aim in combinatorial chemistry is to generate highly diverse libraries, the need for speed and automation introduces new structural idiosyncrasies into the method. Since Lipinski first published his "rule of five",37 describing statistically the optimal combination of properties of drug-like molecules, additional methods for identifying distinguishing features between drugs and other organic molecules 16 have been sought. Approaches have included the characterization of molecular frameworks and substituents, 38 the statistical analysis of different drug databases, 39 the application of artificial neural networks to differentiate drugs from reagents, 4° and the introduction of a drug-like index to rank compounds. 4' The differences between three different compound classes, natural products, molecules from combinatorial synthesis, and drug molecules, were investigated (Figure I-6). The major structural differences between natural and combinatorial compounds originate mainly from properties introduced to make combinatorial synthesis more efficient. These include the number of chiral centers, the prevalence of aromatic rings, the introduction of complex ring systems, and the degree of the saturation of the molecule as well as the number and ratios of different heteroatoms. Natural products generally have high binding affinities for specific receptor systems and their biological action is ofien highly selective. In view of these facts, it is interesting to consider that the search for the replacement of natural compounds with synthetic ones is usually based on exactly these kinds of 'unfavorable' modifications. Given the stereospecificity of most biological targets, it is likely that many non-stereospecific synthetic analogues, created, for example by the introduction of aromatic rings, represent suboptimal compromises, especially in terms of selectivity. This situation appears to be amplified in the case of combinatorially synthesized compounds. In addition, the greater flexibility of combinatorial products is likely to have detrimental entropic consequences for the binding of these compounds; it may also adversely affect their ability to induce conformational changes in the receptor required for biological function. 17 .5 Combinatorial Products compounds .5” Natural Figure [-6. Comparison of diversity: the plot of the first two principal components, obtained from a database containing a) a random selection of combinatorial compounds (n = 13 506), b) natural products (n = 3287), and c) drugs (n = 10 968). For clarity, the data points from the three databases are plotted separately but on the same axes. The Figure [-6 shows that combinatorial compounds cover a well-defined area in the diversity space given by these principal components. In contrast, natural products and drugs cover almost all of this space as well as a much larger additional volume. Drugs and natural products have approximately the same coverage of this space. Adapted from Ref. 43 descriptor 2 Libraries based on a defined target descriptor 3 descriptor 1 Figure [-7. The goal in medicinal and combinatorial chemistry is to synthesize a collection of analogues (blue spheres) of a target structure (lead) having known or predicted properties (red sphere). Adapted from Ref. 52 As well, the process of producing synthetic analogues radically alters the numbers and ratios of different atom types, such as nitrogen, oxygen, sulfur, and halogens. These distributions in turn have a direct impact on the patterns of donors and acceptors available to complement receptor surface properties. We can view the production of unique bioactive libraries by plants and organisms as analogous and complementary to the combinatorial generation of synthetic libraries, each approach providing access to very different types of lead compounds.I " 42 However, there is a significant difference between these processes. The generation of combinatorial libraries has been constrained primarily by the availability of reagents and suitable reactions. In contrast, the generation of natural product diversity has occurred not only within the constraints of available biosynthetic reactions and precursors but also in the context of biological utility. Most natural products have a function, and the synthetic routes generating these compounds have coevolved with the requirements of ligand functionality. This suggests that combinatorial synthesis must also evolve beyond synthetic feasibility to explicitly address the creation of compounds with proper biological function.43 H. Rescuing Combinational Chemistry (Combichem) The question arises how to improve the diversity of synthetic and especially combinatorial compounds. Some features that are being focused on to rescue combinational chemistry are as follows: 19 o Chirality is an essential issue. As chiral separation is rather expensive, the number of chiral centers could be somewhat increased by using readily available chiral reagents more often in combinatorial syntheses. e Attempting to change the ratios of different heteroatoms is another promising way of increasing diversity in combinatorial products. This will also impact donor/acceptor properties as well as lipophilicities and hence raise the diversity of designs. 0 Drug-like filters, such as the Lipinski rules, are very helpful in removing likely problem molecules. However, overly strict adherence to it can have the adverse effect of restricting diversity and also reducing similarity to natural products. 0 A large proportion of natural products is biologically active and has favorable ADMET properties, despite the fact that they often do not satisfy 'drug- like' criteria. It is likely that by mimicking certain distribution properties of natural compounds, a substantially more diverse set of combinatorial products might be made that could also have greater biological relevance. Analogously to different 'drug-like' filters, the concept of 'natural-product-like' filters can be introduced based on the statistical analysis of different properties described above. Other methods that might help to bring some of the properties of combinatorial compounds closer to those of natural products, e. g. 0 application of biocatalysis and biotransformations in the synthesis44a or using natural product templates in library design, 44” have been suggested. 20 0 Dixon and Villar45 have shown that a protein can bind a set of structurally diverse molecules with very similar affinities in the nanomolar range, whereas a number of analogs closely related to one of the good binders display only weak affinities (>2.5 mM). The design and sampling of compound libraries should, therefore, be guided not only by structural descriptors, but also by descriptors of biological activity. 0 In ‘click chemistry’ 46" developed by Sharpless and co-workers, a biological receptor such as an enzyme is permitted to select the best fitting partial ligands (ligands that don’t occupy the entire binding site) from a range of modules that are capable of reacting with one another. After the modules bind to the site their reactivity towards each other induces them to combine forming a compound that blocks and thus inhibits the entire catalytic site. 0 Dynamic combinatorial chemistry (DCC) 46” involves a target receptor which is exposed to a library of potential ligands. Each ligand is formed by reversible combinations of small building blocks in an equilibrium solution. Selective binding of one of the ligands to the target lowers the ligands concentration in solution, causing the equilibrium towards the formation of the ligand. Thus, the system enhances the amount of strong binding ligand and discriminates against poorly binding ones. 0 The majority of drug targets cluster into densely populated target families. Numerous lead compounds from a multi-purpose ‘privileged structure’ libraries are generated, which can address a variety of targets in a single cluster irrespective of a therapeutic area."6c For e.g. a single library built around a 5,5- 21 trans- fused lactarn as a core scaffold generated inhibitors for different serine proteases ( a single family of proteases) involved in different diseases.46c I. Chemical diversity and biological Space Combinatorial chemistry aims to access diversity to some degree, and usually involves synthesizing analogues of a given target structure i.e. to explore a dense region of chemical space in proximity to a precise or dense region (occupied by a lead) known to have useful properties (Figure I-8).47a Since the structural diversity of the products was only due to the building blocks and starting scaffold the resulting molecular framework is the same in every case. If the protein binding sites for potential ligands are defined as a biological structure space, the task is to find compounds that are “complementary" to this structure space, that is, to correlate the biological structure space with a chemical structure space. The real question was neglected: how biologically relevant is the diversity created in chemistry? Even the most beautiful molecules synthesized using the most elegant methods are useless if they do not reach a biological target. Even experts fail to agree on how diversity should be defined (Figure I-9 & 10). We use the term “diversity” in the sense of structural variety on the basis of molecular scaffolds and substitution patterns. Despite the sequencing of the human genome, the biological structure space is largely unknown (Figure I-10). However, it is clear that it cannot be filled by using the substances that exist today. In theory, it is conceivable that the entire chemical structure space of all the possible drug molecules could be synthesized and hence filled. HTS would then automatically identify the suitable molecules. In practice, this approach is not 22 feasible: theoretical considerations have shown that the universe does not contain enough atoms to synthesize even one copy of every conceivable molecule."7b The number of ‘drug-like' molecules possible has been estimated to be astronomic (l 062 to 10200)_48, 49 BIOLOGICAL l— STRUCTURE SPACE —-r-» *CHEMICAL STRUCTURE SPACE MISMATCHED OPTIMAL CHEMICAL gggmzuiicgggmtl BIOLOGICAL AND DIVERSE LIBRARY SPACES CHEMICAL SPACES Figure [-8. Matching of biological & chemical structure spaces. Adapted from Ref. 47a. OH Chloramphenicol b. - 'OH N\ \ O ': QH HN O HOII'QOH NH2 01L o 3/ \MO‘ ”Jj’c' N\ O\ Clindamycin CC-Puromycin Erythromycin Figure I-9. Structural diversity is not identical to biological diversity. The four antibiotics Chloramphenicol, clindamycin, erythromycin and puromycin are structurally very diverse. They are macrocyclic, heterocyclic or homocyclic. They bear different fimctional groups such as nitro, ester, keto, hydroxy, thioether, tertiary amine, or Chloro. Nevertheless, their modes of action are the same. They all bind to the bacterial peptidyl transferase. They even have overlapping binding sites at the enzyme cavity Adapted from Ref. 47c. 23 Ratj adone 0 Leptomycin A HOOC \ \ \ \ Anguinomycin A Kazusamycin \ a W 0 Pironectin Garnahomolide A \ \ OH 0503 \ \ \ 9H HNa \ \ \ O \ ,5 \ 0 Ho 9 F ostriecin Ebelactone A Figure I-10. Structural similarity is not identical with biological similarity and vice versa. A collection of similar natural products exerting a diversity of biological activities. Members of the highly bioactive family of pentenolide natural products and relatives. They contain a pentenolide head and a long (C6— C23), often branched unsaturated lipophilic tail with a spare ornamentation of hydroxy or keto functionalities: antifungal Ratjadone, the cytostatic Leptomycin B inhibitor of the nuclear export receptor CRM-l, Anguinomycin A cytotoxic against transformed cells, antibiotic Kazusamycin, immunosuppressant Pironetin, cytotoxic Leptofitranin A acting on retinoblastoma protein, antileukemic CI 920, antifungal Garnahomolide A, gap phase specific inhibitor of the mammalian cell 24 cycle Leptolstatin (formula not shown), tubulin inhibitor Discodermolide (formula not shown), and enzyme inhibitor Ebelactone.Adapted from Ref. 470. As a comparison there are approximately 105 1 atoms on earth, so every 'drug- like' molecule (let alone ones not considered ‘drug-like'); cannot be made and one must be selective. Fortunately, there is hope; there is more than one ‘answer' to biological ‘problems’ (e. g. the HMG CoA reductase inhibitors: lovastatin, pravastatin, simvastatin, fluvastatin, atorvastatin, cerivastatin. . .) so we don't need to make and screen everything. In fact, biological activity is not a rare chemical property; the reality is that all small molecules are active biologically in some way or another, even ethanol. In terms of lead generation it is quality (structural diversity) and, but not just, quantity (number of compounds) that counts. J. Diversity Oriented Synthesis (DOS) Diversity-oriented synthesis aims to pick up where traditional combinatorial chemistry left ofl... Stu Borman, C&EN Washington.46b Are the regions of chemistry space defined by natural products and known drugs, the best or most fertile regions for discovering small molecules that modulate macromolecular function in useful ways? DOS aims to answer this question. In contrast to target-oriented synthesis (TOS) and combinatorial chemistry, which aim to access precise or dense regions of chemistry space, diversity-oriented synthesis (DOS) populates chemical space broadly with small-molecules having diverse structures. The synthesis effort in DOS aims to create a broad distribution of compounds in chemistry space, including currently poorly populated (or even vacuous) space, and in the future, in space found empirically to correlate best with desired properties (Figure H l). 25 descriptor 2 descriptor 2 Libraries based on a defined 6590me 3 descriptor I C escriptor 3 A descriptor I Figure M 1. Comparison of TOS (A), medicinal and combinatorial chemistry (B), and DOS (C). Each three-dimensional plot is meant to represent the chemical product or collection of products derived from a single synthesis pathway. Each axis plots a calculable or measurable property of a small molecule (for example, molecular weight, solubility). A) The aim in TOS is to synthesize a single target structure having known or predicted properties (red sphere). B) The goal in medicinal and combinatorial chemistry is to synthesize a collection of analogues (blue spheres) of a target structure having known or predicted properties (red Sphere). C) The aim in DOS is to populate chemistry space broadly with complex and diverse structures having unknown properties (blue spheres) as a first step in the small molecule discovery process. In some ways, these three approaches to synthesizing small-molecules represent points along a continuum. Adapted from Ref. 52 The structures and functions of natural products suggest that structural complexity may be positively correlated with macromolecule-perturbing function and specificity of action. This correlation is particularly striking in small molecules known to disrupt protein—protein interactions. Hence, in contrast to the relatively flat molecular skeletons ofien used in medicinal and combinatorial chemistry that tend to project appendages outward along the perimeter of a circle, The goals of DOS include the development of pathways leading to the efficient (three- to five- step) synthesis of collections of small molecules having skeletal and stereochemical diversity with defined coordinates in chemical space. Ideally, these pathways also yield more globular or spherical molecular skeletons to which substituents can be potentially appended site- and stereo-selectively during a post- screening, optimization stage. The diverse skeletons and stereochemistry ensure 26 that the appendages can be positioned in multiple orientations about the surface of the molecules. Small molecules used in chemical genetics0 screens can be synthesized with guidance from compounds known to have biological activity5 I or with the desire to distribute the products in chemical descriptor space. In the latter case, it may be advantageous to maximize the representation of different functional groups and conformations in a screen, since in most cases the nature of the small-molecule-target interaction can not be foreseen. 1' ’ 5 2 In DOS, where the structural complexity of the individual compounds and the structural diversity of the overall collection are maximized, synthesis pathways are branched and divergent, and they are planned in the forward-synthetic direction52 by using forward-synthetic analysis. Forward-synthetic planning aims to move in the direction of simple and similar to complex and diverse. Forward- synthetic planning aimed at accessing these diversity elements relies on the use of diversity generating processes, which are defined as the transformation of a collection of relatively similar substrates into a collection of more diverse products. In an ideal DOS pathway all of the products of one diversity-generating process are substrates for another, thus making it possible to use split-pool synthesis to access combinatorially matrices of building blocks, stereochemical isomers, and even molecular skeletons. To maximize efficiency and to be compatible with one-bead/one-stock solution technology platforms, synthesis pathways in DOS should be no more than three to five steps (which leaves no room for protective- group manipulations). The products of one diversity- generating process should share some common inherent chemical reactivity. This 27 common reactivity serves as a keying element that makes the products collective substrates for a subsequent diversity-generating process. The goal of achieving diversity can be simplified by considering three distinct diversity elements: appendages, stereochemistry, and skeletons; l) Appendage Diversity (Building Block) The simplest diversity-generating process is the central feature of combinatorial chemistry and involves the use of coupling reactions to attach different appendages to a common molecular skeleton. In forward-synthetic analysis these are referred to as appending processes. If a molecular skeleton has multiple reactive sites with potential for orthogonal functionalization, then the technique of split-pool synthesis can be used to harness the power of combinatorics (a multiplicative increase in the number of products with an additive increase in the number of reaction conditions), and thereby generate all possible combinations of appendages (that is, the complete matrix) efficiently. The core of all the compounds in a library is same. 2) Stereochemical Diversity Stereochemical diversity increases the number of relative orientations of potential macromolecule-interacting elements in small molecules. It can best be achieved by using stereospecific reactions that proceed with enantio— or diastereoselectivity. The collective transformation of chiral substrates into products having increased stereochemical diversity (namely, diastereoselective diversity-generating processes) requires powerful reagents that can override substrate bias and deliver diastereomeric products with very high selectivity.” It 28 is conceivable that these types of stereochemical diversity generating transformations could be carried out with spatially segregated, pooled substrates under common reaction conditions. Forward-synthetic planning that incorporates multiple stereochemical diversity-generating processes into a single pathway should also make it possible to generate stereochemical diversity in a combinatorial fashion, analogous to the ability of appending processes to generate building block diversity in a combinatorial manner. 3) Skeletal Diversity Although efficient pathways leading to families of structurally complex small molecules are being reported with increasing frequency, 5" pathways leading to structurally diverse products, with many different skeletal arrays, are rare. The primary reason for this gap in our planning capabilities is that efficient and effective “branching" pathways remain elusive. Such pathways require the identification of a substrate or class of substrates that can be differentially manipulated, in single transformations, to afford products containing distinct skeletal arrays of connecting atoms.55 There are, at present, two different strategies for planning DOS pathways that generate skeletal diversity. i) Differentiation: The first strategy involves using different reagents to transform a common pluripotent substrate with the potential for diverse reactivity into a collection of products having distinct molecular skeletons (Figure I-12). 29 . Reagent - based Substrate - based differentiation approach folding approach reagent X = 0 common reagents D ...... O [1.330 reagent Z> Figure I-12. General approaches for planning pathways to generate skeletal diversity. Adapted from Ref. 11. To address the diversity of the core scaffold, some approaches involve the use of reaction-based templatess6 or reaction intermediates57 that can be converted in subsequent steps into a highly diverse set of structures. This approach is analogous to the natural process of cell differentiation in which a pluripotent stem cell is transformed into different cell types on exposure to distinct differentiation factors. These reagent based skeletal diversity-generating transformations are, therefore, also referred to as differentiating processes. Nature has made extensive use of this diversification strategy, for example, enzyme-catalyzed construction of the various sesquiterpene carbon skeletons of secondary metabolites from one common precursor, famesyl pyrophosphate (Figure I-13).58 AW... __. e0 caryophylline farnesyl pyrophosphate guaiazul‘ * l . '2' B—cadinene Figure I-13. Nature’s Synthesis of Secondary Metabolites. Adapted from Ref. 58. 30 Several versatile molecules that were key intermediates used by traditional medicinal chemists, for example, Corey's lactone, the Wieland-Miescher ketone, and the Prelog-Djerassi lactone, could lead to a variety of apparently unrelated pharmacophoric frameworks. These starting materials are small molecules equipped with many reactive sites and functionalities, which can undergo both skeletal rearrangements and functional group interconversions. The use of such a polymorphic compound as the cornerstone of a library would be of great advantage since: Many known synthetic key intermediates lead to medicinally important or “privileged” scaffolds. They possess high potential for both structural and functional diversification. Their chemical transformations and modifications in solution are well- documented. The pharmacological activity of their descending structures is well- precedented. Finally, while the compounds generated so far are new and will be tested for biological activity, it is the unique reactivity profile of each scaffold and its potential to be differentially functionalized that is deemed to be the most useful feature of this approach.58 Varying the core scaffold as well as the substituents further increases the flexibility of small-molecule libraries (Figure I-l4). 31 V E] single core ;0 % En El flexible core > g) (E Q) Figure I-14. Flexible core structure libraries vary functionalities as well as orientation of display. Adapted from Ref. 61. Some prominent examples for reagent based differentiation approach from recent times are presented below. 1) Schreiber and co-workers report a three-step diversity-oriented synthetic pathway (Scheme I-l) based on reactive dihydroisoquinoline and dihydropyridine intermediates, which undergo a variety of transformations to generate skeletally diverse alkaloid—like compounds. The synthetic strategy presented herein enables the synthesis of skeletally diverse alkaloid-like compounds in only three steps. Key to the synthesis is the generation and isolation of reactive dihydropyridines and dihydroisoquinolines, which are useful substrates for a variety of skeleton- determining transformations. 32 Pri. IPr R‘ — MS Si?" ,N. (OPh) 500 um macrobeads I? 100 nmollbead N R0 N ()Ph NaCNEH3 3 (I34 )2 Reduction 3+2 cycloaddition Br F O‘N GEE/Q Br :j: 1. s dipolar COzMe c. cycloaddition /¢YIIUOI‘I I’I TEA 2+2 cycloaddition Ph N ~ ...... ee 0 _ COOM E \ Bf I e / Bf RC) N Br: Scheme [-1. Skeletal diversity from isoquinoline intermediate on solid phase. This synthetic sequence leads to twelve distinct Skeletons, all of which contain a hydroxy group, which is important for the microarray technology used in protein- binding assays, 59 and all with high purity (80 % purity on average by LC-MS). The modularity of the synthetic pathway allows for the efficient incorporation of building blocks at a number of key sites on the skeletons. Efforts are currently underway to develop a library of alkaloid-like compounds through this synthetic pathway. 60 2) Armstrong and co-workers have identified the squaric acid derived synthesis of fused aromatic and heteroaromatic cores (Scheme I-2) as a reaction platform to generate multiple core structure libraries (MCSLs). For example, quinones, 61b 61c phenols, naphthylfurans, 6" pyrones, indoles, quinolizin- 4-ones, and 61d,6lc multicyclic systems containing identical substituents can be generated from a common precursor. Transformations involving squaric acid derivatives can 33 provide libraries containing two variable sites prior to commitment to a final core structure. Subsequent manipulation of the differentiated hydroxyls or the use of substituted nucleophiles readily provides a library with up to six variable sites stereochemical aspects were neglected. An alkynyl allene has been converted to heterocycles possessing an u-alkylidene cyclopentenone, a 4-alkylidene cyclopentenone, or a cross-conjugated triene. Thus, a common intermediate has been converted to three structurally unique compounds by changing only the reaction conditions and, therefore, controlling various reaction pathways.62 0 Rtfi‘er R o 2 o R. o 9“ R “I ~/\ R2lIo / Scheme I-2. Squaric acid as a versatile precursor to various compound shapes. Adapted from Ref. 61. 3) By taking advantage of competing reaction pathways available via transition metal-catalyzed carbocyclization processes, Brummond and co-workers58 have shown the potential generality of converting a common intermediate to at least 34 three structurally unique and thus unknown hetereocycles (Scheme I-3). While the compounds generated thus far are new and will be tested for biological activity, it is the unique reactivity profile of each scaffold (a-alkylidene cyclopentenone, 4- alkylidene cyclopentenone, and cross-conjugated triene) and its potential to be differentially functionalized that are deemed to be the most useful feature of this 0 pathway a <:/i pathway C (I MO(CO)3 \ [Rh(CO)2C|]2 \ I .... D I toluene. N2 approach. pathway b [RhtCOhC'lz PPhg. AgBF4,CO Geo Scheme [-3. Allenes and Transition metals: A Diverging Approach to Heterocycles. Adapted from Ref. 62. 4) Olsson63 reports a conceptually distinct methodology of combinatorial scaffolding built upon first generating the three necessary pharrnacophore elements followed by constructing the central core unit as a fourth diversity point. This fourth diversity point is mainly the diverse spatial arrangement of the pharrnacophore elements. A practical and efficient Mglz multi-component synthesis of or, B-enones incorporating three diversity elements was carried out using inexpensive starting materials. Branching into different cores (fourth diversity element) was achieved by stepwise cyclization (Scheme I-4). 35 o 0 CH0 (:7). M912 ' + N Cl Cl 1) EtNH2. Et2AlI (mum >9911) 2) KOt-Bu . .. Q (antizsyn >9921) (antizsyn 3:1) Scheme 14. A combinatorial scaffold approach based upon a multicomponent reaction. Adapted from Ref. 63. 5) Wipf and co-workers64 have extended the multicomponent condensation of alkenylzirconocenes with N-diphenylphosphinoyl imines and diiodomethane to the direct cyclopropanation—rearrangement of readily available alkynyl phosphinamides (Scheme 1-5). The resulting C-[l-(2-arylallyl) cyclopropyl] alkylamines provided valuable starting points for the diversity-oriented synthesis of heterocycles. These novel building blocks were converted into heterocyclic 5- azaspiro[2.4]heptanes, 5-azaspiro-[2.5]octanes, and 5—azaspiro[2.6]nonanes by means of selective ring-closing metathesis, epoxide opening, or reductive amination. The resulting functionalized pyrrolidines, piperidines, and azepines are easily accessible in 2-3 steps in good overall yields and are of considerable relevance for chemistry-driven drug discovery. 36 R fig. N // Prom” /¢ —> R' n=0,1 immolph, \\ x x--- H OH R . n mRI .3. Scheme I-S. Diversity-Oriented Synthesis of Azaspirocycles. 6) F allis and co-workers established that the organoindium reagent derived from S-bromo-l, 3-pentadiene and indium metal reacts with excellent regioselectivity with a variety of aldehydes and ketones to afford the nonconjugated, (y- pentadienylation) diene products in respectable yields (Scheme I-6). In addition, the trienes derived from dehydration of the condensation products afford rapid entry to complex multicyclic skeletons fi'om tandem [4 + 2] cycloadditions.‘55 Scheme 1-6. Cross-Conjugated Trienes for Diene-Transmissive Cycloadditions 37 Schreiber and co-workers‘f”5 have reported a branching DOS pathway that builds on the report by Fallis and co-workers on the use of consecutive Diels-Alder reactions. The pathway yielded 29,400 discrete compounds comprising 10 distinct polycyclic skeletons. The six-step, stereoselective synthesis, which affords products having a central skeleton with between two and four rings and up to six stereocenters, was achieved using an inexpensive and accessible, “one bead-one stock solution” technology platform. Scheme I-7. Skeletal diversity via a branched pathway. Adapted from Ref. 66. 38 Schreiber has adapted the above triene synthesis and subsequent complexity- generating reactions to phenolic aldehyde-loaded macrobeads and discovered a set of dienophiles that react only once with the Fallis-type trienes. The latter observation provides a branch point to the pathway, where diene products are formed from a single Diels-Alder cycloaddition, and monoene products are formed from consecutive Diels-Alder reactions involving either the same or different dienophiles (Scheme I-7). An important feature of the branched pathway is that the diastereoselection observed in the original report has been extended to reaction sequences involving different dienophiles. In contrast to appending processes, these differentiating processes have not (as of yet) been used to generate skeletal diversity combinatorially. Doing so will require the identification of differentiating processes having the products-equals- substrates relationship, that is, all of the skeletally distinct products of one differentiating process must share a common chemical reactivity that makes them all potential substrates for another differentiating process. This type of forward- synthetic planning is challenging, and will require nonmutually exclusive approaches to the two, potentially conflicting, goals of maximizing structural diversity and maintaining common reactivity. ii) Folding: An alternative synthesis strategy circumvents this potential conflict. In this case, diverse skeletons of small molecules can be accessed combinatorially by transforming a collection of substrates having different appendages that pre- encode skeletal information (called 0 elements) into a collection of products having distinct molecular skeletons using common reaction conditions (Figure I- 39 12).67 This strategy is analogous to the natural process of protein folding,68 in which different structural information pre-encoded in primary amino acid sequences is transformed into structurally diverse macromolecules using a common folding buffer. Thus, these substrate-based skeletal diversity-generating transformations are referred to as folding processes in forward-synthetic analysis. An advantage of this approach is that sets of 0 elements can be identified that act in combination, that is, a matrix of 6 elements can pre-encode all combinations of distinct skeletal outcomes. These folding processes can be planned by first identifying a relatively unreactive core structure that can be transformed into a more reactive intermediate upon treatment with mild reagents. Distinct skeletal outcomes can then be pre-encoded into a collection of substrates by attaching to this common core different appendages (6 elements) having complementary reactivity with the latent, reactive intermediate. Mild conditions can then be used to liberate the reactive intermediate and to realize the pre-encoded, complementary reactivity, thus resulting in the formation of different skeletons. The aromatic furan ring, for example, is a relatively unreactive core structure that, upon treatment with a mild oxidant, can be transformed into a more reactive, electrophilic cis-enedione intermediate.69 As shown in Scheme I-8, by appending three distinct two-carbon side chains containing two, one, or zero nucleophilic hydroxy groups to a common furan core, it was possible to transform three structurally similar substrates into three products having distinct molecular skeletons through a common set of oxidative and acidic reaction conditions (NBS and PPTS, respectively). 40 Scheme I-8. Skeletal diversity generating folding process. Adapted from Ref. 67. Furan derivative with a side chain containing two nucleophilic hydroxy groups, underwent NBS-mediated oxidative ring expansion and subsequent ketalization70 to yield the [3.2.1]bicyclic ketal (I-8a). Alternatively, the Evans aldol product, with a single hydroxy group on its side chain, underwent oxidative ring expansion and acid-catalyzed dehydration to yield the alkylidene pyran-3-one (I-8b). Finally, treatment of furan derivative, with no nucleophilic hydroxy groups on its two-carbon side chain, under the same reaction conditions resulted in oxidative Opening of the furan ring followed by olefin isomerization71 to yield the trans- enedione (I-8c). The use of this substrate-based approach to generate skeletal diversity combinatorially (namely, achieving a multiplicative increase in skeletons with an additive increase in appendages) requires at least two sets of 0 elements that can be appended at different sites and function in combination to pre-encode a matrix of distinct skeletal outcomes. In contrast to the one-synthesis/one—skeleton 41 approach (which typically involves forming a single molecular skeleton early in a synthesis), a folding process can be used to generate new skeletons at the end of a synthesis pathway. This approach facilitates the generation of functionalized skeletons that might otherwise be difficult to access, such as those having building blocks coupled through carbon-carbon bonds at stereogenic quaternary carbon centers and/or potentially unstable structural elements. Additionally, 0’ elements can be attached to a common molecular skeleton by using appending processes (similar to the way building blocks are appended in the one- synthesis/one skeleton approach). The maintenance of structural similarity and, therefore, common reactivity until late in the synthesis pathway facilitates the realization of this approach using the split-pool technique. 4) Integrated Forward-Synthetic Analysis for Generating Both Complexity and Diversity (Simple and Similar to Complex and Diverse) Achieving high levels of both complexity and diversity in the context of a single DOS pathway will require integrated forward-synthetic planning. One logical approach is to incorporate complexity generating reactions into stereochemical and skeletal diversity-generating processes. 42 OHC U Ugi 4CC-IMDA R2. ,R NC ; = “2M 2 R1’ (sequential use of complexity- _. generatIng reactions) R ,HNOC EtOOC\/\COOH \ O \/\ H ’Rz /—\ ,N Mes’N N‘Mes R1 0 CI., I'll“; Y3P Ph ° 0 \ K‘efo R "‘12:, (complexity-generating reactions 0 O in the context of a skeletal diversity R ‘ H R. O generating folding process) ‘ N H M beta R3 (R) "’R3 3"» alpha R3 (8) / fig Scheme I-9. Integrated skeletal complexity and diversity. Adapted from Ref. 72. Recently, the potential of linking stereochemical control, to the challenging problem of skeletal diversity has been successfully demonstrated72 by using 0 element where stereochemistry of the G-element and not its constitution controls the outcome of the pathway selected. L. Small molecule heterocyclic scaffolds Heterocyclic compounds are important structural motifs present both in natural and synthetic compounds. It is estimated that far more than half of the published chemical literature concerns heterocyclic structures. Small polyfunctionalized heterocyclic compounds play important roles in the drug discovery process 38" 73 and in isolation and structural identification of biological macromolecules.74 Substituted heterocyclic compounds generally offer a high degree of structural diversity and have had a substantial impact when used as therapeutic agents. Indeed, analysis of drugs in late development or on the market shows that 68% of 43 30" Therefore, the cost-effective creation of new and them are heterocycles. versatile heterocyclic core structures and the approach to them are vital for many innovative drug discovery programs. It is hence not surprising that research in the field of combinatorial synthesis of heterocycles has received special attention.” One striking structural feature common to cyclic molecules and inherent to heterocycles, which continues to be exploited to great advantage by the drug industry, lies in their ability to manifest substituents around a core scaffold in a defined three-dimensional representation, thereby allowing for far less degrees of conformational freedom than the corresponding conceivable acyclic structures. In addition, as a result of the presence of heteroatoms such as O, N, and S, heterocycles often exhibit altered absorption, distribution, metabolism, and excretion properties.76 The design of rigid scaffolds which allow the incorporation of a variety of substituents is an attractive approach for the preparation of combinatorial libraries, especially rigid molecules that position amino acid functionality in 3-dimensional mimicry of peptide secondary structure, such as B- tums." Small heterocycles, in particular, are used as rigid, highly functionalized molecular scaffolds and are biologically very interesting.78 Although libraries of peptides and other linear oligoarnides continue to play an important role in 15, 79 bioorganic and medicinal chemistry, more compact, conforrnationally 78, 80 constrained heterocyclic scaffolds hold greater appeal for lead discovery 11, 18, 45 prospecting libraries and represent a greater challenge for synthetic design.81 44 M. Conformational constraint/rigid scaffolds Studies on conformationally constrained molecular structures indicated that rigidification of ligand molecules can potentially influence their biological activity either by modulating their potencies and selectivities for the target receptors. From the thermodynamic point of view, a conformationally constrained molecule with correct bioactive conformation should exhibit enhanced affinity as the reduction in flexibilities reduces loss of entropy upon binding normally occurring when a rotationally free compound becomes more restricted upon alignment with the binding site.82"' 82" In addition, such transformation is also a powerful tool to illustrate possible bioactive conformation of a flexible parent molecule. As a result of such transformations, it might improve their overall pharmacological properties such as the ability to cross blood-brain barrier. Structurally constrained versions of these molecules might produce interesting properties as they might display altered pharmacological and pharmacokinetic properties.” '85 N. l, 3-dipolar cycloadditions and conformationally constrained heterocycles Compact structures which adopt specific conformations are often more valuable as leads than linear and very flexible molecules. The goals of DOS concur with above aim as it includes the development of pathways leading to the efficient (three- to five-step) synthesis of collections of small molecules having skeletal and stereochemical diversity with defined coordinates in chemical space. The development of solid-phase synthetic approaches to small molecules, particularly those which embrace polyfunctional heterocyclic targets is hence of great interest 45 in DOS.86 Structural constraint can be introduced by cyclization more efficiently compared to sequential reactions, but reaction sequences are longer if rings are introduced one step at a time. Reactions that generate multiple linkages in one transformation are extraordinarily powerful in introducing complexity quickly. As a consequence, cycloaddition and cyclocondensation reactions, which generate more than one bond in a single step, are particularly attractive in diversity oriented synthetic design since a premium is placed on reducing the total number of transformations, to ensure adequate yield and purity. Cycloaddition and its intramolecular variants have hence become indispensable for the construction of mono- and polycyclic systems as a consequence of being able to deliver complex cyclic molecules with many positions where variation or substitution is possible from relatively simple and accessible precursors. Inter- and intramolecular Diels-Alder reaction has been used by itself or in sequence with MCR’s in atom economical approaches to conformationally constrained tricyclic nitrogen heterocycles.87 Like the Diels-Alder reaction, 1, 3- dipolar cycloadditions are fundamental processes in organic chemistry for the synthesis of diverse heterocyclic compounds. The reaction of azomethine ylides with various dipolarophiles results in highly substituted five-membered nitrogen heterocycles. l, 3 - dipolar cycloadditions are reactions of considerable interest in diversity oriented synthesis, since they can provide access to these polyfunctional molecules with overall good control of the relative configuration of several consecutive asymmetric centers under relatively simple and scalable experimental procedures. In addition to being versatile, it is also an atom economical process. 46 One of the most important classes of l, 3-dipolar cycloadditions involves azomethine ylides. The recent explosion of interest in azomethine ylides is well justified when considering their synthetic utility in the construction of heterocycles and polycyclic systems. Not only does the wealth of ylide generation protocols permit application to a range of heterocyclic structures, but the advent of chiral control over reaction products and intramolecular protocols allows for azomethine ylide cycloadditions to be applied to challenging synthetic problems, particularly those developed by natural product synthesis. Generation of the ylide, usually in situ, followed by dipolarophile attack affords the heterocycle with operational simplicity. A noteworthy example is l, 3-dipolar cycloaddition of azomethine ylides with electronic-deficient olefins which is of considerable interest in DOS because its stereospecificity enables stereochemical diversification of up to four tetrahedral centers on pyrrolidine rings. Achieving diastereoselectivity and enantioselectivity in this reaction is presently a field of intense scientific investigation. Although established methods for azomethine ylide generation have proven to be both general and efficient, new procedures are constantly being divulged. These new methods are not only for ylide generation, but are also for new chemical equivalents and by careful selection; the synthetic chemist has a wealth of technologies available at his disposal. 47 10. ll. 12. 13. 14. References Ellman, J. A. Acc. Chem. Res. 1996, 29, 132 -143. Verkman, A. S. Am. J. Physiol. Cell Physiol. 2004, 286, C465-C474. Lipsky, M. S.; Sharp, L. K. J. Am. Board Fam. Pract. 2001, 14, 362-367. Carr, R.; Harm, M. Modern drug discovery 2002, 5 (4), 45-48. Pagliaro, L.; Felding, J.; Audouze, K.; Neilson, S. J.; Terry, R. B.; Krog- Jensen, C.; Butcher, S. Curr. Opin. Chem. Biol. 2004, 8, 442-449. Justice, M. J .; Noveroske, J. K.; Weber, J. S.; Zheng, 3.; Bradley, A. Hum. Mol. Genet. 1999, 8, 1955-1963. Schena, M.; Shalon, D.; Heller, R.; Chai, A.; Brown, P. 0.; Davis, R. W. Proc. Natl. Acad. Sci. USA 1996, 93, 10614-10619. Agrawal, S.; Zhao, Q. Curr. Opin. Chem. Biol. 1998, 2, 519-528. Dervan, P. B.; Burli, R. W. Curr. Opin. Chem. Biol. 1999, 3, 688-693. Stockwell, B. R. Nature Reviews Genetics 2000, 1, 116-125. Schreiber, S. L. Science, 2000, 287, 1964-1969. Ding, 8.; Gray, N. S. Wu, Xu; Ding, Q.; Schultz, P. G. J. Am. Chem. Soc. 2002, 124, 1594 -1596. (a) Gallop, M. A.; Barrett, R. W.; Dower, W. J.; Fodor, S. P. A.; Gordon, E. M. J. Med. Chem. 1994, 37, 1233-1251. (b) Pinilla, C.; Appel, J.; Blondelle, S.; Dooley, C.; Domer, B.; Eichler, J .; Ostresh, J .; Houghten, R. A. Biopolymers (Pept. Sci.) 1995, 37, 221-240. (c) Pavia, M. R.; Sawyer, T. K.; Moos, W. H. BioMed. Chem. Lett. 1993, 3, 387-396. ((1) Jung, G.; Becksickinger, A. G. Angew. Chem, Int. Ed. Engl. 1992, 31, 367-383. (C) Dower, W. J.; Fodor, S. P. A. Annu. Rep. Med. Chem. 1991, 26, 271-280. (a) Gold, L.; Polisky, B.; Uhlenbeck, 0.; Yarus, M. Annu. Rev. Biochem. 1995, 64, 763-797. (b) Ecker, D. J.; Vickers, T. A.; Hanecak, R.; Driver, V.; Anderson, K. Nucleic Acids Res. 1993, 21, 1853-1856. (c) Bock, L. C.; Griffin, L. C.; Latham, J. A.; Verrnaas, E. H.; Toole, J. J. Nature 1992, 355, 564-566. 48 15. 16. 17. l8. 19. 20. 21. 22. 23. 24. 25. Simon, R. J.; Kania, R. S.; Zuckerrnann, R. N.; Huebner, V. D.; Jewell, D. A.; Banville, S.; Ng, S.; Wang, L.; Rosenberg, S.; Marlowe, C. K.; Spellmeyer, D. C. Tan, R.; Frankel, A. D.; Santi, D. V.; Cohen, F. E.; Bartlett , P. A. Proc. Natl. Acad. Sci. USA 1992, 89, 9367—9371. Cho, C. Y.; Moran, E. J.; Cherry, S. R.; Stephans, J. C.; Fodor, S. P. A.; Adams, C. L.; Sundararn, A.; Jacobs, A. W.; Schultz, P. G. Science 1993, 261, 1303—1305. Han, H.; Janda, K. D. J. Am. Chem. Soc. 1996, 118, 2539-2544. Spaller, M. R.; Burger, M. T.; Fardis, M.; Bartlett, P. A. Curr. Opin. Chem. Biol. 1997, 1, 47-53. Smith, A. B.; Hirschmann, R.; Pasternak, A.; Akaishi, R.; Guzman, M. C.; Jones, D. R.; Keenan, T. P.; Sprengeler, P. A.; Darke, P. L.; Emini, E. A.; Holloway, M. K.; Schleif, W. A. J. Med. Chem. 1994, 37, 215-218. Kim, E. E.; Baker, C. T.; Dwyer, M. D.; Murcko, M. A.; Rao, B. G.; Tung, R. D.; Navia, M. A. J. Am. Chem. Soc. 1995, 117, 1181-1182. (a) Frank, R.; Heikens, W.; Heisterberg-Moutsis, G.; Blbcker, H. Nucleic Acids Res. 1983, 11, 4365-4377. (b) Geyson, H. M.; Meloen, R. H.; Barteling, S. J. Proc. Natl. Acad. Sci. U.S.A. 1984, 81, 3998-4002. (c) Houghton, R. A. Proc. Natl. Acad. Sci. U.S.A. 1985, 82, 5131-5135. (a) Bunin, B. A.; Ellman, J. A. J. Am. Chem. Soc. 1992, 114, 10997- 10998. (b) DeWitt, S. H.; Kiely, J. S.; Stankovic, C. J.; Schroeder, M. C.; Cody, D. M. R.; Pavia M. R. Proc. Natl. Acad. Sci. U.S.A. 1993, 90, 6909- 6913. Furka, A.; Sebestye'n, F.; Asgedom, M.; Dibo, G. Int. J. Peptide Prot. Res. 1991, 3 7, 487-493. (a) Herrnkens, P. H. H.; Ottenheijm, H. C. J .; Rees, D. Tetrahedron 1996, 52, 4527-4554. (b) Herrnkens, P. H. H.; Ottenheijm, H. C. J.; Rees, D. Tetrahedron 1997, 53, 5643-5678. (c) Dolle R. E.; Nelson, K. H. Jr., J. Comb. Chem. 1999, 1, 235-282. (d) Tan, D. S.; Foley, M. A.; Shair, M. D.; Schreiber, S. L. J. Am. Chem. Soc. 1998, 120, 8565-8566. (a) Kerr, J. M.; Banville, S. C.; Zuckermann, R. N. .1. Am. Chem. Soc. 1993, 115, 2529-2531. (b) Nikolaiev, V.; Stierandova, A.; Krchnak, V.; Seligrnann, B.; Lam K. S.; Salmon, S. E.; Lebl, M. Peptide Res. 1993, 6, 161-170. (c) Ohlmeyer, M. H. J.; Swanson, R. N.; Dillard, L. W.; Reader, 49 26. 27. 28. 29. 30. 31. 32. 33. 34. 35. 36. J. C.; Asouline, G.; Kobayashi, R.; Wigler, M.; Still, W. C. Proc. Natl. Acad. Sci. U.S.A. 1993, 90, 10922-10926. Arlens, E. J.; Beld, A. J.; Rodrigues, d. M. J. F.; Simonis, A. M. In The Receptors: A Comprehensive Treatise; O’Brien, R. D., Ed.; 1979, 1, 33- 41. Arya, B; Chou, D. T. H.; Back, M. -Y. Angew. Chem. Int. Ed. 2001, 40 (2), 339—346. (a) Houghten, R. A.; Pinilla, C.; Blondelle, S. E.; Appel, J. R.; Dooley, C. T.; Cuervo, J. H. Nature 1991, 354, 84-86. (b) Lam, K. S.; Salmon, S. E.; Hersh, E. M.; Hruby, V. J.; Kazmierski, W.M.; Knapp R. J. Nature 1991, 354, 82-84. Spring, D. R. Org. Biomol. Chem. 2003, 1 (22), 3867 -— 3870. (a) Lesney, M. S. Modern drug discovery 2002, 5(2), 26—30. (b) Janvier, P.; Sun, X.; Bienaymé, H.; Zhu, J. J. Am. Chem. Soc., 2002, 124 (11), 2560 -2567. (a) Drews, J .; Ryser, S. Drug Inf. J. 1996, 30, 97-108. (b) Drews, J. Drug Discovery Today 1998, 3, 491-494. There are many reasons for this trend, for example, the drug discovery process has become much more expensive ($231 million in 1990 to $897 million in 2000; a 388% increase); and although investment in pharmaceutical research and development has also increased dramatically, it has not increased as much as drug discovery costs ($10 billion in 1990 to >$25 billion in 2000; a 250% increase). The Tufts Center for the Study of Drug Development, News Release: 13 May 2003: http://csdd.tufis.edu/NewsEvents/ $231 million in 1990 is equivalent to $318 million in year 2000 dollars. Drews, J. Science 2000, 28 7, 1960-1964. Terrett, N. K. Combinatorial Chemistry, Oxford University Press, 1998. Kingston, D. G. 1. Natural Products as Pharmaceuticals and Sources for Lead Structures. In The Practice of Medicinal Chemistry; Wermuth, C. G., Ed.; Academic Press: London, 1996. Verdine, G. L. Nature, 1996, 384, 11-13. 50 37. 38. 39. 40. 41. 42. 43. 44. 45. 46. 47. 48. 49. 50. Lipinski, C. A.; Lombardo, F .; Dominy, B. W.; Feeney, P. J. Adv. Drug Del. Rev. 1997, 23, 3-25. (a) Bemis, G. W.; Murcko, M. A. J. Med. Chem. 1996, 39, 2887-2993. (b) Bemis, G. W.; Murcko, M. A. J. Med. Chem. 1999, 42, 5095-5099. (a) Oprea, T. I. J. Comput.-Aided M01. Des. 2000, 14, 251-264. (b) Simon, R. J.; Kania, R. S.; Zuckermann, R. N.; Huebner, V. D.; Jewell, D. A.; Banville, S.; Ng, S.; Wang, L.; Rosenberg, 8.; Marlowe, C. K.; Spellmeyer, D. C. Tan, R.; Frankel, A. D.; Santi, D. V.; Cohen, F. E.; Bartlett , P. A. Proc. Natl. Acad. Sci. USA 1992, 89, 9367—9371. (a) Ajay; Walters, W. P.; Murcko, M. A. J. Med. Chem. 1998, 41, 3314- 3324.(b) Sadowski, J .; Kubinyi, H. J. Med. Chem. 1998, 41, 3325-3329.(c) Frimurer, T. M.; Bywater, R.; Naerum, L.; Lauritsen, L. N.; Brunak, S. J. Chem. Inf Comput. Sci. 2000, 40, 1315-1324. Xu, J .; Stevenson, J.; J. Chem. Inf Comput. Sci. 2000, 40, 1177-1187. Ortholand, J. — Y.; Ganesan, A. 2004, 8, 271-280. Feher, M.; Schmidt, J. M. J. Chem. Inf Comput. Sci, 2003, 43 (1), 218 - 227. (a) Zaks, A.; Dodds, D. R. Drug Design Discovery 1997, 2, 513-531. (b) Hall, D. G.; Manku, S.; Wang, F. J. Comb. Chem. 2001, 3, 125-150. Dixon, S. L.; Villar, H. O. J. Chem. Inf Comp. Sci. 1998, 38, 1192-1203. (a) Borman, S. Chem. Eng. News 2002, 80(6), 29-34. (b) Borman, S. Chem. Eng. News 2004, 82(40), 32-40. (c) Miiller, G. Drug Discovery Today, 2003, 8(15), 681-691. (a) Wess, G.; Urrnann, M.; Sickenberger, B. Angew. Chem. Int. Ed. 2001, 40(18), 3341-3350. (b) Fenniri, H. Curr. Med. Chem. 1996, 3, 343-378. (c) DOmling, A. Curr. Opin. Chem. Biol. 2002, 6, 306-313. Bohacek, R. S.; McMartin, C.; Guida, W. C. Med. Res. Rev., 1996, 16, 3- 50. Owen, M. J . Biotech Advantage 2002, 6, l 7. http://www.biotecplatfomgguk/newsletter/ Schreiber S. L. Chem. Eng. News 2003, 81, 51-61. 51 51. 52. 53. 54. 55. 56. 57. 58. 59. 60. 61. Breinbauer, R.; Vetter, I. R.; Waldmann, H. Angew. Chem. 2002, 114, 3002-3015. Burke, M. D.; Schreiber, S. L. Angew. Chem. Int. Ed. 2004, 43, 46-58. (a) Sharpless, K. B. Chem. Scr. 1985, 25, 71-75. (b) Masamune, S.; Choy, W.; Petersen, J. S.; Sita, L. R. Angew. Chem. Int. Ed. Engl. 1985, 24, l- 30. (c) Masamune, S. Heterocycles 1984, 21, 107-136. (d) KO, S. Y.; Lee, A. W. M.; Masamune, S.; Reed, L. A., III; Sharpless, K. 8.; Walker, F. J. Science 1983, 220, 949-951 (e) Harrison, B. A.; Verdine, G. L. Org. Lett. 2001, 3, 2157-2159. (1) Gierasch, T. M.; Shi, Z.; Verdine, G. L. Org. Lett. 2003, 5, 621-624. (a) Xu, R.; Greiveldinger, G.; Marenus, L. E.; Cooper, A.; Ellman, J. A. J. Am. Chem. Soc. 1999, 121, 4898-4899. (b) Lee, D.; Sello, J. K.; Schreiber, S. L. J. Am. Chem. Soc. 1999, 121, 10648-10649. (0) Spring, D. R.; Krishnan, S.; Schreiber, S. L. J. Am. Chem. Soc. 2000, 122, 5656-5657. (d) Lee, D.; Sello, J. K.; Schreiber, S. L. Org. Lett. 2000, 2, 709-712. (e) Pelish, H. E.; Westwood, N. J.; Feng, Y.; Kirchhausen, T.; Shair, M. D. J Am. Chem. Soc. 2001, 123, 6740-6741. Micalizio, G. C.; Schreiber, S. L. Angew. Chem. Int. Ed. 2002, 41, 152- 154. Keating, T. A.; Armstrong, R. W. J. Am. Chem. Soc. 1996, 118, 2574- 2583. Gordon, E. M.; Gallop, M. A.; Patel, D. V. Acc. Chem. Res. 1996, 29, 144- 154. Brummond, K. M.; Mitasev, B. Org. Lett. 2004, 6 (13), 2245 -2248. (a) Hergenrother, P. J .; Depew, K. M.; Schreiber, S. L. J. Am. Chem. Soc. 2000, 122, 7849-7850; (b) Barnes-Seeman, D.; Park, S. B.; Koehler, A. N.; Schreiber, S. L. Angew. Chem. Int. Ed. 2003, 42, 2376-2379. Taylor, S. J.; Taylor, A. M.; Schreiber, S. L. Angew. Chem. Int. Ed. 2004, 43(13), 1681-1685. (a) Turnbull, P.; Heileman, M. J.; Moore, H. W. J. Org. Chem. 1996, 61, 2584-2585.(b) Xiang, Y.; Xia, H.; Moore, H. W. J. Org. Chem. 1995, 60, 6460-6467. (c) Birchler, A. G.; Liu, F .; Liebeskind, L. S. J. Org. Chem. 1994, 59, 7737-7745.(d) Paquette, L. A.; Doyon, J .; Kuo, L. H. Tetrahedron Lett. 1996, 37, 3299-3302. (e) Paquette, L. A.; Morwick, T. M.; Negri, J. T.; Rogers, R. D. Tetrahedron 1996, 52, 3075-3094. 52 62. 63. 64. 65. 66. 67. 68. 69. 70. 71. 72. 73. 74. 75. 76. Tempest, P. A.; Armstrong, R. W. J. Am. Chem. Soc. 1997, 119, 7607- 7608. Bertozzi, F .; Gundersen, B. V.; Gustafsson, M.; Olsson, R. Org. Lett. 2003, 5, 1551-1554. Wipf, P.; Stephenson, C. R. J.; Walczak, M. A. A. Org. Lett. 2004, 6, 3009-3012. W00, 8.; Squires, N.; Fallis, A. G. Org. Lett. 1999, 1(4), 573 -576. Kwon, 0.; Park, S. B.; Schreiber, S. L. J. Am. Chem. Soc. 2002, 124, 13402- 13404. Burke, M. D.; Berger, E. M.; Schreiber, S. L. Science 2003, 302, 613-618. Anfinsen, C. B. Science 1973, 181, 223-230. (a) Clauson-Kaas, N.; Dietrich, P.; Nielson, J. T. Acta Chem. Scand. 1953, 7, 845-848. (b) Elming, N. Advances Org. Chem. Methods results 1960, 2, 67-115. (a) Achmatowicz, 0. Jr.; Bukowski, P.; Szechner, B.; Zwierzchowska, Z.; Zamojski, A. Tetrahedron 1971, 27, 1973-1996. (b) Harris, J. M.; O'Doherty, G. A. Tetrahedron Lett. 2000, 41 , 183-187. Kobayashi, Y.; Nakano, M.; Kumar, G. B.; Kishihara, K. J. Org. Chem. 1998, 63 (21), 7505 -7515. Sello, J. K.; Andreana, P. R.; Lee, D.; Schreiber, S. L. Org. Lett. 2003, 5, 4125-4127. De Laet, A.; Hehenkamp, J. J. J .; Wife, R. L. J. Heterocycl. Chem. 2000, 37, 669-674. (a) Hinterding, K.; Alons-Diaz, D.; Waldmann, H. Angew. Chem, Int. Ed. Engl. 1998, 3 7, 688-749. (b) Hung, D. T.; Jamison, T. F .; Schreiber, S. L. Chem. Biol. 1996, 3, 623-639.(c) Mitchison, T. J. Chem. Biol. 1994, 1, 3- 6. (d) Schreiber, S. L. Bioorg. Med Chem. 1998, 6, 1127-1152. (a) Nefzi, A.; Ostresh, J. M.; Houghten, R. A. Chem. Rev. 1997, 97, 449- 472. (b) Franzen, R. G. J. Comb. Chem. 2000, 2, 198-214. (c) Dolle, R, E. J. Comb. Chem. 2000, 2, 383-433. Beck, B.; Picard, A.; Herdtweck, E.; DOmling, A. Org. Lett. 2004, 6 (1), 39-42. 53 77. 78. 79. 80. 81. 82. 83. 84. 85. 86. (a) Souers, A. J.; Ellman, J. A. Tetrahedron 2001, 57, 7431-7448. (b) Wallace, 0. B.; Whitehouse, D. L.; Dodd, D. S. Combinatorial Synthesis of Peptidomimetics; Abell, A., Ed.; JAI Press Inc.: Stamford CT, 1999, 2, 1-51. (c) J. Comb. Chem. 2003, 5 (3), 278 -284. Franzen, R. G. J. Comb. Chem. 2000, 2, 195-214. (a) Zuckermann, R. N.; Kerr, J. M.; Kent, S. B. H.; Moos, W. H. J. Am. Chem. Soc. 1992, 114, 10646-10647. (b) Hagihara, M.; Anthony, N. J.; Stout, T. J.; Clardy, J.; Schreiber, S. L. J. Am. Chem. Soc. 1992, 114, 6568-6570. (a)AndreS, C. J.; Denhart, D. J .; Deshpande, M. S.; Gillman, K. W. Comb. Chem. High Throughput Screening 1999, 2, 191-210. (b) Dolle, R. E. J. Comb. Chem. 2001, 3, 477-517. Oliver, S. F.; Abell, C. Curr. Opin. Chem. Biol. 1999, 3, 299-306. (a) Hruby, V. J. Life Sci. 1982, 31 , 189-199. (b) Avbelj, F.; Hadzi, D Mol. Pharmacol. 1985, 27, 466-470. (c) Kolhatkar, R. B.; Ghorai, S. K.; George, C.; Reith, M. E. A.; Dutta, A. K. J. Med. Chem. 2003, 46, 2205- 2215. Hanessian, S.; McNaughton-Smith, G.; Lombart, H.-G.; Lubell, W. D. Tetrahedron 1997, 53, 12789-12854. (a) Wheal, H. V.; Thomson, A. M., Eds. Excitatory Amino Acids and Synaptic Transmissions; Academic Press: London, 1995. (b) Brauner- Osborne, H.; Egebjerg, J .; Nielsen, E. 0.; Madsen, U.; Krogsgaard-Larsen, P. J. Med. Chem. 2000, 43, 2609-2645. (c) Ozawa, S.; Kamiya, H.; Tsuzuki, K. Prog. Neurobiol. 1998, 54, 581-618. (a) Conti, P.; De Amici, M.; di Ventimiglia, S. J.; Stenstl, T. B.; Madsen, U.; Brauner-Osbome, H.; Russo, E.; De Sarro, G.; Bruno, G.; De Micheli, C. J. Med. Chem. 2003, 46 (14), 3102 -3108.(b) Clerici, F.; Gelmi, M. L.; Gambini, A. J. Org. Chem. 1999,64, 5764-5767. (c) Souers, A. J .; Rosenquist, A.; Jarvie, E. M.; Ladlow, M.; Feniuk, W.; Ellman, J. A. Bioorg. Med. Chem. Lett. 2000, 10(24), 2731-2733.(d) Lewis, R. T.; Macleod, A. M.; Merchant, K. J .; Kelleher, F .; Sanderson, 1.; Herbert, R. H.; Cascieri, M. A.; Sadowski, S.; Ball, R. G.; Hoogsteerr, K. J. Med. Chem. 1995, 38, 923-933. (a) Gordon, B. M.; Barrett, R. W.; Dower, W. J .; Fodor, S. P. A.; Gallop, M. A. J. Med Chem. 1994, 37, 1385-1401. (b) Terrett, N. K.; Gardner, M.; Gordon, D. W.; Kobylecki, R. J.; Steele, J. Tetrahedron 1995, 51, 54 87. 8135-8173. (c) Thompson, L. A.; Ellman, J. A. Chem. Rev. 1996, 96, 555- 600. (d) Gong, Y. -D.; Najdi, S.; Olmstead, M. M.; Kurth, M. J. J. Org. Chem. 1998, 63 (9), 3081 -3086. Paulvannan, K. J. Org. Chem. 2004, 69, 1207-1214. 55 CHAPTER II AZLACT ONES TO NOVEL DIHYDROHETEROCYCLIC SCAFFOLDS. Oxazolones represent an immensely versatile family of heterocyclic compound for fixture exploitation by synthetic chemists... .A. 1. Myersl A. Introduction In our quest for small molecule heterocyclic scaffolds with novel biological activities, we focused on the azlactone template (5-oxazolone) on account of its versatility as a template for synthesis of other heterocycles. Oxazolones can exist in five isomeric forms all of which are known (Figure II-1)2. RI 0 R1 0 R1 0 0 o 0 o Will/$0 I§2=O \INl’We—Rz III/\fm T§k R1 R2 R1 0 R2 R2 5(4H)-oxazolone 5(2H)-oxazolone 4(5H)-oxazolone 2(3H)-oxazolone 2(5H)-oxazolone or or or azlactone pseudoxazolone isoxazolone Figure II-l. Isomeric Oxazolones and structures. Azlactones or 2-oxazolin-5-ones (5(4H)—oxazolone) are cyclic esters of N—acyl-OI- amino acids. In 1883 Plbchl first discovered them by condensation of benzaldehyde and hippuric acid in acetic anhydride, but it was Erlenmeyer (1993) who rationalized the structure and christened them as “azlactones”.3 Because he failed to realize the ease of hydrolysis of saturated azlactones, it was in 1908-1910 that Mohr and co-workers were successful in preparing several of them by using acetic anhydride as dehydrating agent.3 Azlactones gained prominence as the intermediates primarily responsible for racemization when N—acyl-a-amino acids 56 or N-protected peptides are coupled with nucleophiles (amino acids, amines, alcohols, etc.) using DCC or other reagents during peptide synthesis.’ Since then the chemistry of azlactones has evinced great interest mainly as synthons for peptide synthesis and other heterocycles.6 Efficient multi-component reactions permit the incorporation of several elements of diversity in a single step and are ideal for application to the diversity-oriented synthesis of the libraries of complex molecules.5 For our efforts, to generate natural product like libraries for chemical genetics, we decided to focus on azlactones as template/fluid core7 for diversity oriented synthesis of other heterocycles. The following features of azlactones guided us in our choice; 0% 0 0 R/IK‘. NJYO Carboxyl acid activator R 6 "I9 R (OH @NR' R2 R=H alkyl or acyl munchnone ICarboxyl acid activator: (RCO)20. DCC. EDCIJ Figure II-2. Cyclodehydration of N-acyl-a-aminoacids to azlactones/munchnones. 1) Easily accessible starting materials. As a source of input materials for the synthesis of azlactones, o-amino acids occupy a special position. They remain unmatched as a source of diverse starting materials: densely functionalized, commercially available in protected form, as pure enantiomers, as the natural as well as a myriad of "unnatur " derivatives.8 A variety of N-acyl-ot-amino acid can be obtained under Schotten-Baumann conditions or simple acylation of or-arnino acid esters followed by mild basic 57 hydrolysis.9 Traditionally the cyclodehydration of N-acyl-or-amino acid to azlactones has been carried out by refluxing in acetic anhydride.lo This method suffers from side reactions, and it is hard to get rid of the acid impurity which is detrimental to sensitive azlactones. Milder versions involving trifluoroacetic anhydride (stoichiometric amounts) or mixed anhydrides have also been used routinely.ll Although high yields are obtained by using DCC, removal of urea byproduct completely is not trivial.12 Use of water soluble carbodirnides like EDCI have gained popularity because of the high purity of final azlactones (which can be used without further purification for subsequent reactions) and also because they minimize racemization of optically pure products (Figure II-2).'2 Any discussion of azlactones is incomplete without the tautomeric mesionic 1,3- oxazolium-S-oxides (mitnchnones), formed during the preparation of azlactones with acetic anhydride or other acid anhydrides.'3’ " Azlactones can also be trapped in mesionic form by N-alkylation.ls A new palladium-catalyzed route to prepare milnchnones directly fiom basic building blocks imine, carbon monoxide, and acid chloride has been developed recently providing an efficient way to add diverse substituents to this versatile template (Figure II-3).'6 Use of 1- isocyanocyclohexene as “universal isocyanide” enables postcondensation modification of Ugi four-component condensation products via munchnone that reacts with many external and internal nucleophiles to yield a variety of products. '7 58 [Pd2(dba)3] (5 "101 %) 0 R2 0 PO—toll 15mol°/ 0 HE"; A +co ‘ ”3‘ °’ - ...—«I. R, CI 3.5, EtN‘Prz, 65 °C 6) I" R2 Amdtsen's method R1 R1-COOH q 0 R2 RNH2 Ugi )Lc. Lb 0 0 —-—> R1 N \ ———> R1—<\ e RZCHO ' @N II NC Armstrong '3 method Figure II-3. Syntheses of milnchnones from basic building blocks. 2) Pluripotency Azlactones afford a host of diverse products with different reagents (Figure II-4) and the reaction may involve retention or collapse of the heterocycle itself. The azlactone is an activated internal ester (lactone) of the corresponding N-acyl-or- amino acid and as such can undergo nucleophilic attack by host of nucleophiles including alcohols, amines, carboxylates, etc.2’ '8 It also undergoes reactions of activated carbonyls like Friedel-Crafis arylationl9 and Wittig olefination.20 59 R1 / Nucleophiles Y° /~' 0 :9 Electrophiles Nucleophiles R2 saturated azlactone R1YO 1.3-dipole | &7 GIN e 0 R A , . \\ I? R2 dipolarohlles B R = H alkyl or acyl mu'nchnone II Nucleophiles 2Y0 0% )oL l: / Nucleophiles NR Nucleophiles R1 N SD R2 ketene unsaturated azlactone Figure II-4. Diverse reactive sites and intermediates accessible from azlactones. Unsaturated azlactones (prepared by condensation with aldehydes or ketones) present a uniquely reactive exocyclic double bond for additional reactions with nucleophiles.18 Monosubstituted azlactones at C4 can react with a variety of electrophiles due to acidity of C4-H bond. Alkylations, arylation, acylation, Claisen rearrangement, addition of azine N-oxides, addition of diazonium salts and Michael additions have been well documented.'8 Azadienes first undergo electrophilic addition at C4 followed by a nucleophilc attack of nitrogen at C2 of azlactones to afford 2-pyridones.2' Miinchnones and their a-amide substituted ketene tautomers, are an important family of substrates for the synthesis of biologically relevant molecules.22 Miinchnones are cyclic azomethine ylides and hence versatile 1,3-dipolar addition substrates and have proven key to the construction of a range of natural products and pharrnacologically relevant 22,23a,b 23c,d heterocycles, including pyrroles, imidazoles, pyrrolines,23¢ and indole 23f derivatives. Alternatively, the less stable ketene tautomer can serve as a precursor to III-amino acid and peptide derivatives, via reaction with alcohols or N- 60 terminated peptides?“ and their cycloaddition with imines provides access to peptide- based [3-1actarns.2‘""c 3) Skeletal diversity through azlactones Ar k” R,COHN R2 0 4 R4 If N R4 B-Iactem Rs R‘IKIOH 1) EtsN l TMSCI l < R1 N’ R2 2) R/—\R‘ 4-hydroxypyn‘dine 3 O o , . .. M l R4 R3 \ A R OCHN OCR‘ ‘ R2 3 CHIOR),, Aep, A PM /\e R \ 002m 1) / PPhaBr 1 N R t . 2 2 N OMe pyrrolIne ) a Rf." R2NHCOR1 \ R3 2 R3. -pyndone WZNMN'R‘ R7." IiihHCOR. A R3 N Nlllle2 pyrimidinone 1)ArH/AICI3 R'TW/ N 2) POClg. A R2 oxazole 0 Mail I R3X R11 R > 3 R2 disubstituted azlactone Figure II-5. Cyclocondensation reactions of azlactones to generate diverse heterocycles. Azlactones are extremely versatile and reactive templates/intermediates and are well-suited for the synthesis of diverse heterocycles by both reagent/reaction condition based differentiating processes and substrate controlled folding (intramolecular) procedures.25 i. Differentiation] Intermolecular reactions. The azlactone template can be diversified into hetrocyclic skeletonsls like 26.27 oxazoles,l9 pyridines, pyridones,” 61 pyrimidinones,28 pyrazolones, imidazolones, isoxazolidinones, isoquinolines, tetrazoles, etc by short cyclocondensation sequences.2J8 R R Ph “El/Ce — R. h [>35 R’O R2 1 Pllozs.IN Q...” Ni: CIR kAl’ o :0) OZT’R“ R76 0 R1/4N\ R2 Z—Ar r R2 N of Ph N'R R4 R3 ( R2: cyclic amino acid / \ R . ‘ R _ R 7 I 2 R4 — s n Figure II-6. Cycloadditions of mtinchnones to generate diverse heterocyclic skeletons. Oxazoles, pyrananones, simple pyrrolines and quartemary amino acids can be accessed with diverse substituents and such diversity of substituents is hard to be rivaled by other methods (Figure II-5).2"8 1,3-Dipolar cycloaddition reactions of N-methylated mesoionic oxazolones or munchnone (Figure II-6), provides a general route for the syntheses of a host of heterocycles with diverse skeletons, 62 for example pyrroles, pyrrolines, azepines and imidazoles, etc.”"’ 23“ 36 By N- methylation, the azlactones are locked in the mesoionic form and are extremely reactive dipoles, which afford the heterocycles in very high yields. Strained rings like cyclopropenes and benazetes also participate in cycloadditions with mfinchnones to afford 4-pyridones and indoles respectively.2 Gelmi and co- workers used vinyl phosphonium salts to overwhelm the effect of polarization of the vinyl group with the strong interaction of phosphonium group and carbonyl group.29 Trisubstituted pyrrole can thus be accessed with high regiocontrol. Gribble and co-workers have shown mfinchnones can react in tandem fashion with cyclooctadiene to give caged compounds.30 Tandem addition of maleimides to the mesionic Al-pyrroline after decarboxylation occurs in exo fashion compared to the first endo addition resulting in complex tricyclic heterocycles.l3 Munchnones derived from cyclic amino acids like proline, can be used for efficient construction of pyrrolizidine and indolizidine skeletons, which form the core of several natural products and biologically active compounds.3| ii. Folding I Intramolecular reactions The development of effective approaches to bridged heterocyclic ring systems is an important topic in organic synthesis. Because of the ease of constructing two rings simultaneously, intramolecular 1,3-dipolar cycloaddition strategies have emerged as an important tool for the synthesis of structurally complex fused heterocyclic ring systems.32 Particularly, several bicyclic and polycyclic fused pyrrolidine ring systems are synthesized by the intramolecular cycloadditions of stabilized acyclic azomethine ylides with tethered dipolarophiles (0 elements)” 63 Combination of the intramolecular 1,3-dipolar cycloaddition with other reactions offers a number of interesting synthetic possibilities. In spite of copious literature dealing with bimolecular cycloaddition reactions of mesionic heterocycles, intramolecular examples have received only a minimum of attention. R1 m0 E 1lea 6:? °©f§¥° OH \ Na J\CF3 N CF3 n H Figure II-7. Alkenes as intramolecular diversity elemnts (0 elements). An attractive feature associated with the internal cycloaddition of mesionic compounds is the opportunity to control the stereochemistry of the products at several centers. In systems where the dipole and dipolarophile are linked by several atoms, the highly ordered transition state can also induce useful regiochemical control.40 Padwa and co-workers utilized the intramolecular mfinchnone-alkene cycloadditions to craft a series of caged compounds with quinoline and isoquinoline cores (Figure II-7).41 B-carbolines skeletons found frequently in alkaloids were generated by a Pictet-Spengler reaction of an intramolecular azlactone derived from tryptophan.33 Alkynes tethered to N—acyl- a-amino acids have been shown to undergo intramolecular cycloadditions when refluxed in acetic anhydride via the miinchnones to afford benzopyrano[4,3-b] pyrrole and lH-pyrrolo[l,2-c] thiazole derivativesm’ Martinelli and co-workcrs have developed a new approach to octahydroindoles via intramolecular cycloadditions of milnchnones with tethered alkynes (Figure II--8).34 ITIOS 0 o Ac20 \ 0 up > 0' .2 \ eR2 A N o". R 0 / 0' #0 \\ sr—fkb SYeNfiko Ph Ph Figure 11- 8. Alkynes as intramolecular diversity elemnts (0 elements). The approach is simple, convergent, widely applicable and uses both natural and unnatural amino acids. It also provides quick entry to mitomycin skeleton and other medicinally important polycyclic heterocyclic compounds. The only limitation is the substituent on nitrogen needed for formation of munchnone intermediate. 65 0 NHCOR1 3 \ ,2 ire/<0 O 3Q 0 \ o o \ R2 A R2 / N‘~ N\ I R1 R1 R3 R0 0 R3 o “5%? 0 ”Y N o \ R1 Y E Figure 11- 9. Alkenes as intramolecular diversity elements in condensations(o elements). Oxazinones, pyridines and pyrones are obtained after stepwise intramolecular cycloadditions (Figure II-9) depending on the 0 element (reactive group that decides the skeleton of the product) on the C4 carbon of the azlactones.22a B. l, 3-Dipolar cycloadditions and stereochemical diversity 25’ 35 Stereochemical diversity increases the number of relative orientations of potential macromolecule-interacting elements in small molecules. It can best be achieved by using stereospecific reactions that proceed with enantio— or diastereoselectivity. Since diversity-generating processes involve the transformation of a collection of substrates into a collection of products, it is critical that the processes used to generate new stereogenic centers are both selective and general. Many 1, 3-dipolar cycloadditions have the ability to generate rings (and functionality derived from the transformations of such rings) 66 containing several contiguous stereocenters in one synthetic operation. The configurations of these new stereocenters arise from the geometry of the dipole and dipolarophile as well as the topography (endo or exo) of the cycloaddition. Additional stereospecificity is possible when the reactive n faces of either of the cycloaddends are made diastereotopic by the use of chiral auxillaries or chiral complexes with external agents. Since the diastereoselectivity of 1,3-dipolar cycloaddition is under a powerful substrate control it is challenging to generate the opposite diastereomeric product. Double-diastereoselective reagents that can override the face selectivity of both coupling partners, for example, to achieve exo versus endo selectivity would be highly valuable and the discovery of these types of powerful reagents is critical to achieving stereochemical diversity in DOS. F orward-synthetic planning that incorporates multiple stereochemical diversity generating processes into a single pathway should also make it possible to generate stereochemical diversity in a combinatorial fashion, analogous to the ability of appending processes to generate building-block diversity in a combinatorial manner. C. Lack of stereochemical diversity in munchnone cycloadditions It has been well documented that instead of 1, 3-dipolar cycloaddition azlactones undergo a Staudinger-type reaction when heated with imines in toluene yielding B-lactams. The azlactone, which is in equilibrium with valence tautomeric ketene intermediate, undergoes a [2+2] cycloaddition reaction with the imine (Scheme II- 3). It can be concluded that either mesoionic oxazolium oxide, which has the 67 nitrogen atom unsubstituted, is not in reasonable concentration and/or the imine nitrogen is not nucleophilic enough. Hence formation of ketene takes predominance, which reacts readily with the imines to afford the B-lactam.6’ 36' 37 In 1970, Huisgen had reported that a moderate equilibrium concentration of the mesoionic tautomer is essential for the 1, 3-dipolar cycloaddition leading to heterocycle formation (Figure II-lO).l3 By N-alkylation or acylation, the azlactones are locked in the mesoionic form and are extremely reactive dipoles, which afford the pyrroles and other hetereocycles in very high yields.15 002Me ll'ne Ph 0 09 H Ph N 002'“ M9020 002Me \ / 002Me o -002 / \ ,N -———> 002Me ——> Ph R Me® R1 0 R1 'i‘ 1 Me Figure II-10. Synthesis of pyrroles from mfinchnones. The analogous reaction of a munchnone with a nitrile to provide an imidazole is not viable due to the low reactivity of nitriles. Imidazoles (Figure [H 1) can be generated by treatment of the mtlnchnones with tosylimines. Addition of the tosylimines results in an unstable bicyclic adduct that loses carbon dioxide and benzenesulphinic acid to gain aromaticity.23°""""38 The phenylsulphonyl residue as leaving group enhances the tendency to aromatize (Figure IL] 1). Milnchnones with different substituents, at the 2 and 4-positions lead regioselectively to products in which a bond is always formed between the 2-carbon atom of the dipole and the imine nitrogen atom. This attack was explained on the basis of a dipole-dipole interaction between partially negative nitrogen of the imine and the 68 electrophilic C-2 atom of the munchnone. The yield reported for these reactions are low at least partly due to self-condensation of milnchnones.23d The problem of self-condensation has been suppressed by using a solid-phase approach towards imidazoles. Bilodeau et 0123 ° used a standard reductive amination protocol to hook an amino acid ester onto a aldehyde group on a commercially available resin. This path of attachment also served to alkylate the nitrogen atom of the azlactone that was made by subsequent cyclodehydration of the amino acid, which was condensed with tosylamines to afford the imidazoles in good yields and purities. | ”Y e ”W" Ph " G?" N “2 NH “2 .ofix; __-_<_=°_2, P, If / "“369 R,” -PhSOzH I}! R1 Figure II-ll. Synthesis of imidazoles from mfinchnones. 1, 3-dipolar cycloadditions of mfinchnones are simple thermal processes, unaided by external reagents. The products resulting from cycloadditions of mtlnchnones with various dipolarophiles are aromatic and carry little or no stereochemical information. In intermolecular reactions, primary cycloadducts have generally not been isolated and identified because they readily eliminate carbon dioxide resulting in aromatization.39 Double bond isomerization (in case of alkenes to A2- pyrrolines) and multiple cycloadditions have been observed after decarboxylation, especially in case of addition of alkenes.40 This results in loss of stereogenic centers from the cycloadditions in the final products. Although, the decarboxylation seems to be driven by the stability gained by aromatization, it is 69 not a concerted process and the harsh conditions often employed might play a major role. Maryanoff and co-workers have described one example of the isolation of a Al-pyrroline-S-carboxylic acid from reaction of l, 2- dicyanocyclobutene from the intermolecular cycloaddition of a munchnone.“ A notable exception was reported by Padwa and co-workers who successfully isolated and characterized the primary adducts from intramolecular cycloadditions of mfinchnones to terminal alkenes.“ ‘3 In the above reactions, expulsion of carbon dioxide is presumably impeded by the severe structural constraints in the intermediate polycycle. They were also able to show that the cycloadditions occurs with exo stereochemistry for the substituents on alkenes and concluded that it is reflective of the steric constraints imposed by a unimolecular transition state. The polycyclic heterocyclic compounds oxazoloquinolines and oxazoloisoquinoline products are potential precursors to amaryllidaceae alkaloids. Padwa and co-workers further diversified these heterocyclic systems by addition of organometallic reagents/reduction with LAH to tricyclic lactols. These lactols undergo ring opening to benzazepine scaffolds which are valuable peptidomimetics displaying diverse biological activities. 42’ 43 D. Potential of Lewis acid mediated generation of miinchnones and synthesis of novel dihydroheterocyclic scaffolds. We envisioned that coordination of a Lewis acid to nitrogen atom of the azlactone, would increase the equilibrium concentration of munchnone intermediate and accelerate the azomethine ylide cycloaddition reaction (Scheme II-l).44 Moreover if milder protocols could be developed by Lewis acid 70 mediation, then it might be possible to isolate either the bicyclic cycloadduct or the dihydroheterocycles (Scheme II-l) without any decarboxylation or 45, 46 rearrangement. [3+2] cycloadditions in which heterodipolarophiles are employed to intercept reactive azomethine ylides represent a useful entry into 45"" In case of milnchnones, functionalized, protected amino acids. dihydroheterocycles isolated would be masked a,a-disubstituted (quartemary) amino acids. Interest in the synthesis of biologically active products containing quaternary carbon atoms has increased notably over the last decade.48 The interesting properties of a- and B-amino acids with conformational rigidity, in particular , the a,[3-disubstituted series have recently attracted the attention of numerous research groups since the incorporation of these products into peptides can drastically modify their properties. .134 HX R.—<\:I° éj R‘fiNrj Ce L. R; L’® ® (OH >9 O J R2 Y RF’X R1-<\ 1R2 /N I R3 N '- R4 '0‘ v = o, N, -CH Scheme II-l. Synthesis of dihydroheterocycles from mfinchnones. During their efforts to utilize the l, 3-dipolar cycloadditions in the diversity oriented synthesis of biomimetic probes, Austin and co-workers49 discovered that the cycloadditions of vinyl ethers with isomilnchnones proceeds with complete endo diastereoselectivity. Removal of chiral auxillary through an unusually facile 71 ester aminolysis resulted in efficient construction of bicyclic [2.2.1] scaffold (Figure II- 12). The hetero-bicyclic [2.2.1] scaffolds allows for the diversity both in terms of position and type of functionality incorporated and were used to interact with the HIV-1 Tat/T AR system. RN A-protein interactions constitute a rapidly growmg area of research, especially because of their prevalent role in many cellular processes. 0 RKanH o o R 1) condensation i + , ; R1 [was 0 0 2) diazotransfar R2 N2 HOMOR3 0R4 P e 0R3 ‘ RFDVOR‘ R5 R6 Re 0 R0: I Rh(ll) R14 f0 3 *0 N o I o O N .. .1 R1 ‘R2 bicyclic [2.2.1] scaffold Figure II-12. Synthesis of bicyclic scaffolds from cycloaddition of isomunchnones. Dihydro— and completely reduced five-membered heterocycles represent an important addition to the repertoire of small molecules that are being used to control complex biological processes. Mapp and co-workers50 have described the design and synthesis of isoxazolidines, the very first examples of synthetic small molecule heterocycles that activate transcription at levels comparable to those of a natural activation domain. Transcriptional activators play an essential role in the regulatory network that controls gene-specific transcription (Figure II—l 3).5 1The misregulation of this complex event cascade is correlated with a growing number 72 of human diseases,52 and thus interest in developing artificial transcriptional activators has intensified.53 To identify a minimal fimctional unit for a small molecule-based activation domain, a series of isoxazolidines containing functional groups typically found in endogenous activation domains were designed (Figure II-13). The isoxazolidine scaffold was chosen due to the relative ease with which diverse functional groups could be incorporated in a stereocontrolled manner onto the conformationally constrained ring, 50’ 54 thus displaying those groups in a three-dimensional array. Remarkably, one isoxazolidine (Figure II-13) was nearly as active as the positive control ATF14 (natural ligand) despite a considerable difference in size (MW 290 versus 1674). Similar to natural activation domains such as ATF14, a balance of hydrophobicity and polarity was found to be important for overall potency.” 55 Two factors were suggested to play a role in the isoxazolidine being nearly as active as ATF14 despite the size difference. As an organic molecule, the isoxazolidine ring is resistant to proteolytic degradation and thus probably has a longer lifetime. In addition, the isoxazolidine molecule likely populates conformations more closely related to the final bound state due to structural constraints imposed by the ring. 73 “0 g , __ MN 0 [3+2] hi— 1) 14,98, N— OH > A OH Dihydrofolate receptor (DHFR) " u transcriptlbn Lax A Lax A Figure II-l3. Synthesis of isoxazolidines as mimic of transcription regulators. E. Lewis acid mediated cycloadditions of nitrile ylides and azomethine ylides.“ Control of the stereo-, regio-, and enantioselectivity of l, 3-dipolar cycloadditions with Lewis acids is a relatively new area and has been examined with only a limited number of dipoles. A common difficulty is the competition between the dipole and the dipolarophile for the external reagent when it is a Lewis acid. When one tries to activate electrophilic dipolarophiles with Lewis acids, binding to the 1, 3-dipole can predominate, resulting in a slower reaction. Such strong binding can also cause decreased reactivity of the l, 3-dipole towards nucleophilic dipolarophiles. Strong binding of 1, 3-dipoles to a Lewis acid is often not avoidable because of the electronic structure of the 1, 3-dipoles and the typical 74 prescence of Lewis basic sites. Accordingly, Lewis acidic reagents may combine with the basic sites on certain I, 3-dipoles, making relatively stable salt-like complexes. The co-ordinating power of external reagents must be carefully adjusted in order to be successful in attaining high rate enhancements. The nature of the ligands on the Lewis acid and the lability and exchangeability of these ligands are typical issues to be considered. F. Nitrile ylides and Lewis acids i. Syntheis of 2-oxazolines Suga et al reported that 5-alkoxyoxazoles react with aldehydes giving 4,5-cis-2- oxazoline-4-carboxylates stereoselectively when activated with a stoichiometric amount of methylaluminium B-binapthoxide as the Lewis acid (Figure II-l4).57 This reaction has recently been extended to a catalytically enantioselective version using an enantiopure methylaluminium [3-binapthoxide.’8 Although the actual reacting species were not assigned, 5-alkoxyoxazoles are known to behave as nitrile ylide 1, 3-dipole equivalents in Lewis acid catalyzed reactions with aldehydes. Ito et 01 developed new catalyzed asymmetric cycloadditions reactions of formal nitrile ylides. Thus, methyl a-isocyanoalkanoates were allowed to react with benzaldehyde or acetaldehyde in the prescence of 0.5-] mol % of a chiral (aminoalkyl)ferrocenylphosphine-gold(l) complex to give optically active methyl 2-oxazoline-4-carboxylates with high enantoselectivity in quantitative yield. 75 0\ 0’ AlMe CO ‘(COzMe N \ 30 mol °/o N I1 ’ I 3. ' : : R1/ko OMe RZCHO 111/40 'R2 ”wags" ‘7 Suga's approach: chHO écone [Au(o-CBH11NC)3]*BF4' N1 CNCH co Me _ / only trans 2 2 7 k0 R2 97 %ea N/\/NEt2 l Fe :3th Ito's approach Figure II-l4. Synthesis of oxazolines from oxazoles. The oxazolines were converted into optically active B-hydroxy-or-amino acid methyl esters. The selective formation of cis-isomers of 2-oxazoline-4- carboxylates in suga’s reactions is synthetically complimentry to Ito’s method.59 ii. Synthesis of Az-pyrrolines. Grigg et. a]. recently extended Ito’s reaction to the Ag(I) catalyzed 1,3-dipolar cycloadditions of methyl isocyanate with a,B-unsaturated carbonyl compounds."0 The rate of this reaction can be enhanced effectively with a catalytic amount of silver(I) acetate (1 mol%). Grigg et al proposed a reaction mechanism in which silver(l) acetate adds to the carbon atom of isocyano group to form an organometallic intermediate that is subsequently deprotonated to generate a nitrile 76 ylide 1, 3-dipole. After the cycloadditions, double-bond migration results from the imine-enamine tautomerism to afford Az-pyrrolines (Figure II-15).6o ROZC C02R CNCHZCOZMe ca" A90“ ; 2/ S Roc COR H Gn'gg's approach major: trans C e0A0 /=\ H e 9 R020 002R A 9// Ag ROZC C02R R02¢ C02R U , Z/‘j N COZMe a 002Me Figure II-15. Synthesis of AZ-pyrrolines from isocyanoacetates. iii. Synthesis of 2-imidazolines. Although mechanistically suggested to proceed through aldol reaction, 2- imidazolines (Figure II-l6) have been synthesized from isocyanoacetate esters using Lewis acid catalysis. Hayashi and Ito et a] reported a diastereoselective reaction of N-sulfonylimines with methyl isocyanoacetate catalyzed by An (I) complexes, which provide efficient access to erythro 2, 3-diamino acids." 77 MeZSAuCI N . CN H M __ / casztrans 90:10 C 2C02 e r (N R2 99 q i l Tosyl «RAND F! e PPh2 Lin's approach Figure II-l6. Synthesis of 2-imidazolines from isocyanoacetates. Lin et al have extended the reaction to enantioselective version using catalytic chiral ferrocenylphosphine Au(I) complex.62 G. Azomethine ylides and Lewis acids i. Synthesis of 2-imidazolines. Viso et al reported the very first examples of a highly diastereoselective 1,3- dipolar cycloadditions of azomethine ylides derived from or-iminoestcrs (Figure II-l7) with chiral sulfinimines to produce N-sulfinyl imidazolidines and transformation into non-symmetrical vicinal diarnines.63 Dipole generation under standard conditions using LiBr, Et3N, MeCN or AgOAc, DBU, toluene had failed. Use of LDA allowed for successful cycloadditions with a high degree of endo stereocontrol. Addition of BF 3.0Et2 results in abrogation of cycloadditions pathway and the reaction proceeds with opposite diastereoselectivity via a step- wise mechanism.64 78 Ph ”31> THF. 20h + ‘ . R ' _ o N’ ‘0 O 78 to4 C )l OMe Viso's approach Ar 8 Ph’:'\‘N ;\ OMe 0 Ph 5 I! II : Lr-O s. 1 LAH NH .K 10: t' ——> N4 ) . 2 NH2 5C! r "H 2)TFA N "'R N:\ Ar 'rrR 0 Ar 002Me H0 endo approach major enantiomer Figure II-l7. Synthesis of 2-imidazolines from azomethine ylides. ii. Synthesis of pyrrolidines: The reaction of azomethine ylide l, 3-dipoles with olefinic dipolarophiles to form highly substituted pyrrolidines is a reaction of intense research interest.‘55 It has been applied towards synthesis of substituted prolines, which can serve as catalysts and serve as important motifs in many biologically active molecules. Among the different versions of this reaction, the most practical approach has been the interaction between stabilized N-metalated azomethine ylides derived from a-imino glycinate esters and n—deficient alkenes (Figure II-18). The reaction proceeds under mild conditions and with a high degree of diastereocontrol. Silver (1) and Lithium (1) metal cations (usually stoichiometric amounts) are most commonly used along with an excess of tertiary amine base. Enantiocontrol has recently been extended to this reaction raising its stock for the generation of complex heterocyclic molecules. While Jorgensen er al66 used chiral oxazoline complexes of Zn, Zhang and co-workers65 employed Ag(I) complex with FAP ligands for above cycloadditions. 79 CO Me M60 C a 2 :21 A _ Ph N/\fl/ ' Ph 002MB 0 fi ‘PerEt. 0 °C. Eth. -20 °C. AgOAc. 3 mol % Zn(OTf)2. 10 mol % °7§95 % yield 60%ee, 90 % yield J¢rgenson's approach Zhang's approach COZ‘Bu ‘Buozc, '_—_'/ . PhANJWrW" e D ' Ph N "'cone H o ‘PerEt, -20 °C. AgOAc, 10 mol % O \ /N CO PPM I S-QUINAP Schreiber's approach Figure II-l8. Synthesis of pyrrolidines from a—iminoesters. Schreiber and co-workers further improved on Zhang’s protocol by expanding the substrate scope and increasing the enantioselectivities achieved by employing the QUINAP ligand.67 An important highlight of this work is that they were able to generate quartenary stereogenic centers by employing substituted amino acids. Recently, the normal endo selectivity of the above cycloadditions has been 80 reversed by Komatsu et 01 by a opportune selection of bulky phosphine ligands in the cycloadditions of N-substituted maleimides and a-imino glycinate.68 Reversing the diastereoselectivity which is under substrate control by using external reagents (Lewis acids) is highly desirable in a diversity oriented synthesis program.25 Johnson and co-workers”' document reaction conditions that achieve the first productive cycloadditions of azomethine ylides obtained from Lewis acid catalyzed carbon-carbon bond cleavage of aziridines (Figure II-19). in i" N /\ 0M8 A Ph \ N’fil’ = ph N 002m H O EI3N, 40 °C. 00 6”” Pth CO S-QUINAP Komatsu's approach OMe Ph ifh A Ph t3 0025! A0023 > meager Ph 002Et ZnClz, toluene, 23 °C 5 OMe 76“ J d. r. : 1.4:1 0V?“ 0 69 %yiald G /g \ C? o \O'an'" Johnson '3 approach Figure II-l9. Synthesis of pyrrolidines from aziridines and electron-rich alkenes. 81 The constitution of the cycloadduct (a [4+2] stepwise adduct or 3 [3+2] cycloadduct) obtained was further shown to be strongly dependent on the identity of the alkene trap employed. The activation barrier for electrocyclic carbon- carbon bond cleavage in a prototypical N-aryl-2,3-diester aziridine was measured by Huisgen to be ca. 29 kcal/mol, which translates to rather forcing conditions in thermal dipolar cycloadditions.45a Johnson et a! hypothesized that the metal- coordinated ylide is extremely electron poor and thus belongs to type III in the Sustrnann classification system of 1, 3-dipolar cycloadditions and that rate acceleration should be observed with electron rich dipolarophiles due to the correct HOMO/LUMO match. Accordingly, in presence of catalytic ZnClz acyclic enol ethers intercept the metal-coordinated ylide via the [3+2] pathway to afford pyrrolidines in good yields and with modest diastereocontrol. It is important to note that we have been unable to effectively perform these cycloadditions in the absence of Lewis acid. These experiments indicate that Lewis acid promotion is essential and provide additional support for the type III classification of this family of cycloadditions. 82 iii. Synthesis of dihydroheterocycles. H X=( oer R N dipolarophile YN CO: E \r’ 0 = xfcoza OEt M902. 5 MOI %. 25 °C 30 _ R if OEt /g 9 \Q 051 \ ,M Cl BO 0 g 2 Johnson '3 approach dipolarophile product % yield YN COZEt PhCHO x COzEt 89 Ph Ph025\ /N C023 N=\ N cogs: so Ph PhOZS’ ph °2N\_ /N CozEt Ph .- CozEt 64 02" Ph Figure II-20. Synthesis of dihydroheterocycles from sofi enolization of imidates. Johnson et a1 45 have investigated N-metalated azomethine ylides formed via “soft enolization” of (imino)glycine esters as alternatives to ylides generated fi'om the same compounds in the classic thermal [1, 2]-prototropy manifold which require a weak base working in concert with a metal salt.45b An evaluation of simple Lewis acids revealed that N-malonylimidates undergo catalyzed [3+2] cycloadditions reactions with aldehydes, imines, and activated olefins to form oxazolines, imidazolines, and pyrrolines, respectively (Figure II-20). The reactions proceed 83 optimally at ambient temperature with the addition of 5 mol % of Mng in CH3CN. Based on experiments the authors suggest that the Mg Lewis acid promotes a 1, 2-prototropic shift to give a metal-coordinated azomethine ylide, rather than ionization and proton transfer to give a nitrile ylide. The use of a scalemic metal complex to catalyze the cycloadditions further opens up the possibility of enantioselective variants that are not possible therrnolytically. H. Current work With the awareness that five-membered nitrogen containing skeletons make up the core of several natural products and prospective libraries, the generation of diverse small molecular scaffolds with potential biological activity is an ongoing theme in our group. Dihydroheterocyclic scaffolds on account of their structural rigidity and the three dimensional disposition of functional groups are perfectly suited for this aim. They have a different shape profile from aromatic heterocycles generally encountered in drug discovery programs and as such might access entirely new therapeutic target. There are very few reports in literature involving short and efficient syntheses of dihydroheterocycles, which would also result in appendage and stereochemical diversity. During my graduate work, I discovered a mild in situ method of generation of milnchnones (cyclic azomethine ylides) from azlactones using Lewis acids.” 1 have applied this condition for establishing new stereoselective methods to synthesize N-containing dihydroheterocycles; 2- imidazolines‘m’ and A1-pyrrolines.44c The scaffolds are obtained with high diastereoselectivity and are amenable to diverse substitution (Scheme 11-2). 84 Rs LA = TMSCI N R3 : R j: / ,NVRg 1_<\N C0211 R2 _ _ R4 imine ° 0 Lewis acid 0 o 2-imidazolina R1__<\ = R1_.<\ Io N R2 N R2 - LA l LA=AgOAc R4 R azlactone mllnchnone \ or TMSCI R a 3 4v 1 \ ,cozn R4Ifl\/R3 N 9R2 alkene A1-pyrrolina Scheme II-2. Lewis acid mediated insitu generation of mtin'chnones from azlactones l, 3 dipolar cycloaddition with dipolarophile. These scaffolds offer unexplored territory in terms of their structure (3 -D disposition of functional groups) and their function (biological activity and catalysis). In addition to the novel development of these molecules, we have been able to establish that the imidazoline scaffolds can catalyze a novel stereoselective reaction of ketenes with N-acylimines derived from glycine esters. This reaction is currently under investigation in the group. Most of my work has been published in peer reviewed reputed journals and in light of the potential therapeutic value of the compounds as inhibitors of activation of NF-KB and enhancers of chemotherapeutic potential of anti-cancer drugs; 69 we have been able to obtain patents for the above work.70 The leads are being now carried through cellular and animal studies in active collaborations with other researchers and industry. In the future, enantiocontrol and application of the above mild method of generation of mtlnchnones to other scaffolds, and natural products will be pursued. 85 1. Experimental procedure General procedure for synthesis of azlactones”. 0 R2 EDCI. HCI O O R /U\ *COOH CHZCIZ N 1” R2 A solution of benzoyl amino acid (2 mmol) and EDCI.HC1 (2 mmol) in dichloromethane (20 mL) was stirred at 0°C for 1h till the starting material disappeared. The reaction mixture was washed successively with cold (containing ice) water, aqueous NaHCO3 (10 mL) and water (lOmL). The solution was dried over anhydrous magnesium sulfate, filtered, and the solvent evaporated to dryness in vacuo giving the oxazolones as solids or oils. 2-phenyl-4-mcthyl-4H-oxazolin-S-one II-l: Cyclodehydration of N-benzoyl-L- alanine (2 g, 10 mmol) with EDCI.HC1 (1.92 g, 10 mmol.) gave 1.8lg of IL] in 90 % yield. 'H NMR (300 MHz) (CDCl3): 6 1.6 (3H, d, J = 7.4 Hz), 4.46 (1H, q, J= 7.5 Hz), 7.47 (3H, m), 8.0 (2H, d); l3C NMR (75 MHz) (CDCl;): 816.1, 60.3, 125.3, 127.2, 128.2, 132, 160.7, 178.2; IR (neat): 1828 cm", 1653 cm". 86 o o Flt—(\I N Ph ll-2 2, 4-diphenyl—4H-oxazolin-5-one Il-2: Cyclodehydration of N-benzoyl-L- phenylglycine (2 g, 7.8 mmol) with EDCI.HC1 (1.5 g, 7.8 mmol.) gave 1.3g of II- 2 in 70 % yield. 1H NMR (300 MHz) (CDC13)Z 8 5.5 (1H, s), 7.35-7.65 (8H, m), 8.05-8.15 (2H, m); l3c NMR (75MHz) (CDC13): 8 176. 4, 162.8, 133.9, 133.2, 129.2, 129.18, 129.1, 129, 128.99, 128.96, 128.9, 128.7, 128.4, 127.1, 126.1, 68.4; IR (neat) 1825 cm", 1655 cm". 0 o 911% NH N / Il-3 4-(lH-Indol-3-ylmethyl)-2-phenyl-4H-oxazol-5-one II-3: Cyclodehydration of N-benzoyl-L-tryptophan (2 g, 6.5 mmol) with EDCI.HC1 (1.24 g, 6.5 mmol.) gave 1.5 g of [LB in 80 % yield. 1H NMR (300 MHz) (CDC13): 8 3.5 (2H, ddd, J, = 5 Hz, J2 = 15 Hz, J = 50 Hz), 3.5 (2H, t, J = 5 Hz), 7.05-8.0 (9H, m), 8.18 (1H, b); ”CNMR (75MHz) (013013): 8 178.1, 161.7, 135.9, 132.6, 128.6, 127.8, 127.3, 125.8, 123, 122, 119.5, 119.1, 1 11. 1, 109.5, 66.6, 27.2; IR (neat) 3416.4 cm", 1815 cm", 1653 cm". 87 0 o Ph’QniAFO /0 ll-4 3-(5-Oxo-2-phenyl-4,5-dihydro-oxazol-4-yl)—propionic acid methyl ester [14: Cyclodehydration of N-benzoyl-glutamic acid-S-metyl ester (4 g, 15 mmol) with EDCI.HCl (3.17g, 16.5 mmol.) gave 2.6g of II-4 in 70 % yield. IH NMR (300 MHz) (CDCl3): 8 2.16 (1H, sextet, J = 6.6 Hz), 2.4 (1H, sextet, # 6.5 Hz), 2.4 (3H, t, J = 9 Hz), 3.7 (3H, s), 4.54 ( 1H, t, J = 9 Hz), 7.4-7.6 (6H, m), 8.0 (2H, d, J = 9.9 Hz), 8.2 (2H, d, J = 9.9 Hz); 13C NMR (75 MHz) (CDCI3) 8 177.8, 172.6, 162.2, 161.9, 134.5, 132.8, 130.4, 129.9, 129.8, 128.8, 128.7, 128.5, 128.2, 127. 8, 125.6, 51.7, 29.6, 26.6; IR (neat) cm", 1828 cm", 1651 cm". 0 Me—(SI Me Il-5 2, 4-dimethyl-4H-oxazolin-5—one II-S: Cyclodehydration of N-benzoyl-glutarnic acid-S-metyl ester (4 g, 30 mmol) with EDCI.HC1 (6.3g, 33 mmol.) gave 2.0g of [LS in 60 % yield. lH NMR (300 MHz) (CDCl3): 8 1.37 (3H, t, J = 8 Hz), 2.05 (3H, s), 4.0-4.16 (1H, m); ”C NMR (75 MHz) (CDC13) 8 179.2, 162.6, 60.4, 16.4, 15.1; IR (neat) cm", 1826 cm", 1686 cm". 88 o o Ph/-<\ N:/(p,1 ll-B 2-Benzyl-4-phenyl-4H-oxazol-5-one II-6: Cyclodehydration of N- phenacetylphenylglycine (2 g, 10 mmol) with EDCI.HC1 (1.9g, 10 mmol.) gave 1.64g of II-6 in 90 % yield. 1H NMR (300 MHz) (CDCl;;): 8 3.96 (2H, s), 5.35 (1H, s), 7.2-7.6 (10H, m); 13C NMR (75 MHz) (CDCll) 8 176.2,165.5,133.0, 132.7, 129.3, 128.9, 128.7, 128.5, 128.4, 128.3, 127.7, 127.4, 126.6, 125.42; IR (neat) cm", 1829 cm", 1674 cm". ll-7 4-Methyl-4H-oxazol-5-one Il-7: Cyclodehydration of N-formyl-alanine (2 g, 17 mmol) with EDCI.HC1 (3.3g, 17 mmol.) gave 1.2g of II-7 in 70 % yield. lH NMR (300 MHz) (CDC13): 8 1.46 (3H, t, J = 7.5 Hz), 4.13 (1H, q, J= 7.5Hz), 7.52 (1H, s); l3C NMR (75 MHz) (CDC13) 178.2, 152.8, 58.3, 16.2; IR (neat) cm", 1817 O 0 F3C\< N ll-8 2-trifluoromethyl-4-Methyl-4H-oxazol-5-one 11-8: Cyclodehydration of N- cm", 1676 cm". tn'fluoromethyl alanine (2 g, 10.8 mmol) with EDCI.HC1(2.06g, 10.8 mmol.) 89 gave II-8 in 28 % yield. Partial data: IR (neat) cm", 1840 cm"; Pseudooxazolone: O O .../3.1 IR (neat) cm" 1809 cm". Partial data: 2-Benzyl-4-methyl-4H-oxazol-5-one II-9: Cyclodehydration of N- phenacetyl alanine (4 g, 19.3 mmol) with EDCI.HC1 (3.71 g, 19.3 mmol) gave 3.1g of II-9 as viscous oil in 82 % yield. 1H NMR (300 MHz) (CDCl;): 8 1.47 (3H, d, J = 6.9 Hz), 4.35-4.5 (2H, m), 4.77 (1H, q, J = 7.5 Hz), 7.38 (2H, t, J = 7.2 Hz), 7.48 (1H, t, J = 7.2 Hz, 7.72 (2H, d, J= 7.8 Hz); IR (neat) cm", 1824 cm", 1676 cm". ")—<° O O \ I >—NH N Me 0 1140 Ph/— Partial data: Benzyl-(S)—1-(4,5-dihydro-4-methyl-5-oxooxazol-2-yl)—2-methyl propyl carbamate II-10: Cyclodehydration of N-benzoyl-L-Val-alanine (1 g, 3.25 mmol) with EDCI.HC1 (0.63 g, 3.26 mmol.) gave 0.79 g of II-10 as viscous oil in 84 % yield. 1H NMR (300 MHz) (CDC13): 8 0.77 and 0.79( 3H, 2d, J =4.5 Hz), 0.859 (3H, dd, J, = 2.1 Hz, J; = 6.6 Hz), 1.3 (3H, d, J: 7.5 Hz), 2.03 (1H, m), 4.065 (1H, q, J = 7.2 Hz), 4.4 (1H, q, J = 6 Hz), 4.97 (2H, m), 5.23 (1H, bs), 7.18 (5H, bs); IR (neat): 1828 cm", 1722 cm", 1672 cm". 90 0 O 643—3 I N Me ll-11 Partial data: 2-(4-methoxyphenylH-methyl-4H-oxazolin-S-one II-ll: Cyclodehydration of N-(4—methoxybenzoyl) alanine (3 g, 13 mmol) with EDCI.HC1 (2.5 g, 13 mmol) gave 2g of [H] as a pale yellow oil in 75 % yield. 'H NMR (300 MHz) (CDC13): 8 1.547 (3H, d, J = 7.5 Hz), 3.85 (3H, s), 4.402 (1H, q, J= 7.8 Hz), 6.92-6.96 (2H, m), 7.88-7.91 (2H, m); IR (neat): 1824 cm'l, 1653 cm". Ph—<\ N ll-1 2 Partial data: 2-pheny1-4-isopropyl-4H-oxazolin-5-one II-12: Cyclodehydration of N-benzoyl-L-alanine (6 g, 27 mmol) with EDCI.HC1 (5.2 g 27 mmol.) gave 4.93 g of 11-12 as a low melting solid in 90 % yield. 'H NMR (300 MHz) (CDC13): 8 1.055 (3H, d, J = 6.9 Hz), 1.12 (3H, d, J = 6.9 Hz), 2.3-2.41 (1H, m), 4.29 (1H, d, J: 4.5 Hz), 7.47 (3H, t, J = 7 Hz), 7.56 (3H, t, J = 7.2 Hz), 7.99 (2H, d, J = 7.8 Hz); 13C NMR (75 MHz) (CDC13): 8 17.18, 18.33, 70.13, 125.38, 127.61, 128.11, 128.47, 132.5, 161.7, 177.13; IR (neat): 1826 cm", 1657 cm". 91 O O Ph—QNI/Ph "4 3 2-phenyl«-4-benzyl—4H-oxazolin-5-one II-l3: Cyclodehydration of N-benzoyl—L- phenylalanine (2 g, 7.38 mmol) with EDCI.HC1 (1.42 g, 10 mmol.) gave 1.77g of [L] as a white solid in 95 % yield. 1H NMR (300 MHz) (CDC13): 8 3.17 (1H, dd, J1= 6.6 Hz, J2 = 13.8 Hz), 3.35 (1H, dd, J, = 5.1 Hz, J2 = 13.8 Hz), 4.67 (1H, dd, J, = 5.1 Hz, J2 = 6.6 Hz), 7.17-7.25 (5H, m), 7.39-7.45 (2H, m), 7.5-7.55 (1H, m), 7.86-7.9 (2H, m); 13C NMR (75 MHz) (CDC13)Z 8 37.3, 66.5, 125.7, 127.2, 127.8, 128.4, 128.7, 129.6, 132.7, 135.2, 161.7, 177.6; IR (neat): 1823 cm'l, 1648 cm". 92 _ 0 9° 10. ll. 12. 13. 14. References Meyers, A. 1., In Palmer, D. C., Ed., Oxazoles: Synthesis, Reactions and Spectroscopy, Vol. 60: Part B, The Chemistry of Heterocyclic Compounds; Jon Wiley and Sons, INC., New Jersey, 2002, foreword. Rao, Y. S.; Filler, R., In Turchi, I. 1., Ed., Oxazoles: Synthesis, Reactions and Spectroscopy, Vol. 45, The Chemistry of Heterocyclic Compounds; Jon Wiley and Sons, INC., New York, 1986, 363-691. Carter, H. E. Organic Reactions 3, Jon Wiley and Sons, INC., New York, 1947, 198-239. Comforth, J. W. In Clarke, H. T., Johnson, J., Robinson, R., Eds, Chemistry of Penicillin; Princeton University Press, Princeton, N. J., 1948, 688-848. (a) VonWangelin, A. J.; Neumann, H.; G8rdes, D.; Klaus, S.; Strilbing, D.; Beller, M. Chem. Eur. J. 2003, 9, 4286 - 4294. (b) deling, A. Curr. Opin. Chem. Biol. 2002, 6, 306-313. (c) Démling, A. and Ugi, I. Angew. Chem. Int. Ed. 2000, 39, 3168-3210. ((1) Armstrong, R. W.; Combs, A. P.; Tempest, P. A.; Brown, D. S.; Keating T. A. Acc. Chem. Res. 1996, 29, 123-131. (e) Andreana, P. R.; Liu, C. C.; Schreiber, S. L. Org. Lett. 2004, 6(23), 4231-4233. Mukeljee, A. K. Heterocycles 1987, 26, 1077-1097 Tempest, P. A.; Armstrong, R. W. J. Am. Chem. Soc. 1997, 119, 7607- 7608. (a) Trump, R. P.; Bartlett, P. A. J. Comb. Chem. 2003, 5(3), 285-291. (b) Lewis, J. G.; Bartlett, P. A. J. Comb. Chem. 2003, 5(3), 278-284. Georg, G. I.; Boge, T. C.; Cheruvallath, Z. S.; Harriman, G. C. B.; Hepperleand, M.; Park, H. Bioorg. Med. Chem. Lett. 1994, 4, 335-338. Boyd, G. V., In Potts, K. T., Ed., Comprehensive Heterocyclic Chemistry, 6, Pergamon Press, Oxford, UK, 1984, 177-234. (a) Bergman, J.; Lidgreen, G. Tetrahedron Lett. 1989, 30, 4597-4600; (b) Benoiton, N. L. Int. J. Protein. Res. 1991, 38, 285-286. Chen, F. M. P.; Kuroda, K.; Benoiton, N. L. Synthesis 1970, 230. Gothardt, H.; Huisgen, R.; Bayer, H. O. J. Am. Chem. Soc. 1970, 103, 4340. (a) Padwa, A., In Taylor, E. C.; Weissberger, A., Eds., 1, 3-Dipolar Cycloaddition Chemistry, General Heterocyclic Chemistry Series, Jon 93 15. 16. 17. 18. 19. 20. 21. 22. 23. 24. 25. Wiley and Sons, INC., New Jersey, 1984 .(b) Gribble, G. W., In Palmer, D. C., Ed., Oxazoles: Synthesis, Reactions and Spectroscopy, Volume 60 Part A, The Chemistry of Heterocyclic Compounds; Jon Wiley and Sons, INC., New Jersey, 2002; 473-576. Hershenson F. M.; Pavia, M. R. Synthesis, 1988, 999-1001. Dhawan, R.; Dghaym, R. D.; Amdtsen B. A. J. Am. Chem. Soc. 2003, 125, 1474-1475. Keating, T. A. and Armstrong, R. W. J. Am. Chem. Soc. 1996, 118, 2574- 2583. Cativiela, C.; Diaz-de-Villegas, M. D., In Palmer, D. C., Ed., Oxazoles: Synthesis, Reactions and Spectroscopy, Volume 60: Part B, The Chemistry of Heterocyclic Compounds; Jon Wiley and Sons, INC., New Jersey, 2002, 130-299. (a) Balaban, A. T.; Bally, 1.; Frangopol, P. T.; Bacescu, M.; Cioranescu, B.; Birladeanu, L. Tetrahedron, 1963, I9, 169.; (b) Frangopol, P. T.; Balaban, A. T.; Birladeanu, L. Cioranescu, B.; Tetrahedron, 1961, I6, 59. Erba, E.; Gehni, M. L.; Pocar, D. Chem. Ber. 1988, 121, 1519-1524. Sain, B.; Baruah, J. N.; Sandhu, J. S. J. Chem. Soc. Perkin Trans. 1. 1985, 773-777. (a) Gingrich, H. L.; Baum, J. S. In Oxazoles; Turchi, I. J., Ed.; Wiley: New York, 1986; Vol. 45, p 731. (b) Potts, K. T. In 1.3-Dipolar Cycloaddition Chemistry; Padwa, A., Ed.; Wiley: New York, 1984; Vol. 2, p 1. (a) Keating, T. A.; Armstrong, R. W. J. Am. Chem. Soc. 1996, 118, 2574. (b) Santiago, B.; Dalton, C. R.; Huber, E. W.; Kane, J. M. J. Org. Chem. 1995, 60, 4947. (c) Bilodeau, M. T.; Cunningham, A. M. J. Org. Chem. 1998, 63, 2800. (d) Consonni, R.; Dalla Croce, P.; Ferraccioli, R.; La Rosa, C. J. Chem. Res, Synop. 1991, 7, 188. (e) Huisgen, R.; Gotthardt, H.; Bayer, H. 0. Chem. Ber. 1970, 103, 2368. (f) Nayyar, N. K.; Hutchison, D. R.; Martinelli, M. J. J. Org. Chem. 1997, 62, 982. (a) Bayer, H. 0.; Huisgen, R.; Knorr, R.; Schaefer, F. C. Chem. Ber. 1970, 103, 2581. (b) Funke, B.; Huisgen, R. Chem. Ber. 1971, 104, 3222. (c) Prajapati, D.; Mahajan, A. R.; Sandhu, J. S. J. Chem. Soc. Perkin Trans. 1. 1992, 734. Martin D. Burke and Stuart L. Schreiber Angew. Chem. Int. Ed. 2004, 43, 46—58. 94 26. 27. 28. 29. 30. 31. 32. 33. 34. 35. 36. 37. 38. 39. 40. 41. Takagaki, H.; Yasuda, N.; Asaoka, M.; Take, H. Chem. Lett. 1979, 183. Gotze, S.; Kubel, B.; Steglich, W. Chem. Ber. 1976, 109, 2331. Konwar, D.; Prajapati, D.; Sandhu, J. S.; Karnetani, T.; Honda, T. Heterocycles 1984, 22, 2483-2486. Clerici, F.; Gelmi, M. L.; Trimarco, P. Tetrahedron 1998, 54, 5763-5774. Gribble, G. W.; Sponholtz, W. R 111; Switzer, F. L.; D’Amato, F. J.; Bym, M. P. Chem. Commun. 1997, 993-994. Anderson, W. K.; Mach, R. H. J. Med. Chem. 1987, 30, 2109-2115. Pandey, G.; Sahoo, A. K.; Bagul, T. D.; Org. Lett., 2000, 2(15), 2299 -2301. Condie, G. C.; Bergman, J. Eur. J. Org. Chem. 2004, 1286-1297. Nayyar, N. K.; Hutchison, D. R.; Martinelli, M. J. J. Org. Chem. 1997, 62, 982-991. Padwa, A.; Pearson, W. H., In Padwa, A. Pearson, W. H., Eds., Synthetic Applications of 1, 3-Dipolar Cycloaddition Chemistry Towards Heterocycles and Natural Products, Vol. 59: The Chemistry of Heterocyclic Compounds; Jon Wiley and Sons, INC., New Jersey, 2002, Preface. Avalos, M.; Babiano, R.; Cintas, P.; Hursthouse, M. B.; Jimenez, J. L.; Light, M. B.; Lopez, 1.; Palacios, J. C.; Silvero, G. Chemistry 2001, 7, 3033. Prasad, K. K.; Petrzilka, T. Helv. Chim. Acta 1975, 58, 2504. Croce, P. D.; Ferraccioli, R.; La Rosa, C. Tetrahedron 1999, 55, 201. Padwa, A.; Pearson, W. Synthetic applications of dipolar cycloaddition chemistry towards heterocyclic and natural products; Wiley-VCH: Weinheim, 2002. (a) Huisgen, R.; Gotthardt, H.; Bayer, H. 0. Chem. Ber. 1970, 103, 2368- 2387; (b) Gotthardt, H.; Huisgen, R.; Schaefer, F. C. Tetrahedron Lett. 1964, 10, 487-491; (c) Huisgen, R.; Gotthardt, H.; Bayer, H. O. Tetrahedron Lett. 1964, 9, 481-485. Maryanoff, C.; Karash, C. B.; Turchi, I. J. J. Org. Chem. 1989, 54, 3790- 3792. 95 42. 43. 45. 46. 47. 48. 49. 50. 51. 52. Padwa, A.; Gingrich, H. L.; Lim, R. J. Org. Chem. 1982, 47, 2447-2456. Padwa, A.; Lim, R.; MacDonald, J. G. J. Org. Chem. 1985, 50, 3816-3823. (a) Peddibhotla, S.; Jayakumar, S.; Tepe, J. J. Org. Lett. 2002, 4(20), 3533- 3535. (b) Peddibhotla, S.; Tepe, J. J. Synthesis 2003, 9, 1433-1440. (c)Peddibhotla, S.; Tepe, J. J. J. Am. Chem. Soc. 2004, 126(40), 12776- 12777. (a) Pohlhaus, P. D.; Bowman, R. K.; Johnson, J. S. J. Am. Chem. Soc. 2004, 126(8), 2294-2295. (b) Bowman, R. K.; Johnson, J. S. J. Org. Chem 2004 ASAP Article. (a) Grigg, R.; Kemp, J.; Sheldrick, G.; Trotter, J. J. Chem. Soc., Chem. Commun. 1978, 109-111. (b) Joucla, M.; Hamelin, J. Tetrahedron Lett. 1978, 2885-2888. (a) Pearson, W. H.; Stoy, P.; Mi, Y. J. Org. Chem. 2004, 69, 1919-1939. (b) Harwood, L. M.; Macro, J.; Watkin, D.; Williams, C. B.; Wong, L. F. Tetrahedron: Asymmetry 1992, 3, 1127-1130. Denissova, I., Barriault, L. Tetrahedron 2003,5961), 10105-10146. (a)McKenzie, K. M.; Austin, D. J .; Department of Chemistry, Yale University, New Haven, CT, USA.; Abstracts of Papers, 223rd ACS National Meeting, Orlando, FL, United States, April 7-11, 2002. (b) Savizky, R. M.; Savinov, S. N.; Nathan, D.; Crothers, D. M.; Austin, D. J.; Department of Chemistry, Yale University, New Haven, CT, USA.; Abstracts of Papers, 224th ACS National Meeting, Boston, MA, United States, August 18-22, 2002. (C) Savizky, R. M.; Savinov, S. N.; Nathan, D.; Crothers, D. M.; Austin, D. J.; Department of Chemistry, Yale University, New Haven, CT, USA.; Abstracts of Papers, 226th ACS National Meeting, New York, NY, United States, September 7-11, 2003. (d) Savinov, S.N.; Austin, D. J. Org. Lett. 2002, 4(9), 1415-1418. (e) Savinov, S.N.; Austin, D. J. Org. Lett. 2002, 4(9), 1419-1422. (1) Savinov, S.N.; Austin, D. J. Chem. Commun. 1999, 1813—1814. Minter, A. R.; Brennan, B. B.; Mapp, A. K. J. Am. Chem. Soc. 2004, 126(34), 10504 -10505. Ptashne, M.; Gann, A. Genes & Signals; Cold Spring Harbor Laboratory: New York, 2001. (a) Darnell, J. E. Nat. Rev. Cancer 2002, 2, 740-749. (b) Duncan, 8. A.; Navas, M. A.; Dufort, D.; Rossant, J .; Stoffel, M. Science 1998, 281, 692- 695. (c) Pandolfi, P. P. Oncogene 2001, 20, 3116-3127. 96 53. 54. 55. 56. 57. 58. 59. 60. 61. 62. 63. (a) Ansari, A. Z.; Mapp, A. K. Curr. Opin. Chem. Biol. 2002, 6, 765-772. (b) Mapp, A. K. Org. Biomol. Chem. 2003, I, 2217-2220. (c) Weatherman, R. V. Org. Biomol. Chem. 2003, 1, 3257-3260. (a) Kanemasa, S.; Nishiuchi, M.; Kamimura, A.; Hori, K. J. Am. Chem. Soc. 1994, 116, 2324-2339. (b) Bode, J. W.; Fraefel, N.; Muri, D.; Carreira, E. M. Angew. Chem, Int. Ed 2001, 40, 2082-2085. (c) Minter, A. R.; Fuller, A. A.; Mapp, A. K. J. Am. Chem. Soc. 2003, 125, 6846-6847. (a) Drysdale, C. M.; Duenas, B.; Jackson, B. M.; Reusser, U.; Braus, G. H.; Hinnebusch, A. G. Mol. Cell. Biol. 1995, 15, 1220-1233. (b) Regier, J. L.; Shen, F.; Triezenberg, S. J. Proc. Natl. Acad. Sci. U.S.A. 1993, 90, 883-887. (c) Lin, J. Y.; Chen, J. D.; Elenbaas, B.; Levine, A. J. Genes Dev. 1994, 8, 1235-1246. ((1) Sullivan, S. M.; Horn, P. J.; Olson, V. A.; Koop, A. H.; Niu, W.; Ebright, R. H.; Triezenberg, S. J. Nucleic Acids Res. 1998, 26, 4487- 4496. Kanemasa, S., In Padwa, A. Pearson, W. H., Eds., Synthetic Applications of 1,3-Dipolar Cycloaddition Chemistry Towards Heterocycles and Natural Products, Vol. 59: The Chemistry of Heterocyclic Compounds; Jon Wiley and Sons, INC., New Jersey, 2002, 757-804. (a) Suga, H.; Shi, X.; Ibata, T. J. Org. Chem. 1993, 58, 7397. (b) Suga, H.; Fujieda, H.; Hirotsu, Y.; Ibata, T. J. Org. Chem. 1994, 59, 3359. (a) Suga, H.; Ikai, K.; Ibata, T. Tetrahedron Lett. 1998, 39, 869. (a) Suga, H.; Ikai, K.; Ibata, T. J. Org. Chem. 1999, 64, 7040. (a) Ito, Y.; Sawamura, M.; Hayashi, T. J. Am. Chem. Soc. 1986, 108, 6405. (b) Ito, Y.; Sawamura, M.; Hayashi, T. Tetrahedron Lett. 1987, 28, 6215. (c) Ito, Y.; Sawamura, M.; Hayashi, T. Tetrahedron Lett. 1988, 29, 235. (d) Ito, Y.; Sawamura, M.; Shirakawa, K.; Hayashizaki, K.; Hayashi, T. Tetrahedron Lett. 1988, 28, 239. (d) Ito, Y.; Sawamura, M.; Shirakawa, K.; Hayashizaki, K.; Hayashi, T. Tetrahedron 1988, 44, 5253. Grigg, R.; Landsdell, M. 1.; Thornton-Pett, M. Tetrahedron 1999, 55, 2025- 2044. Hayashi, T.; Kishi, B.; Soloshonok, V. A.; Uozumi, Y. Tetrahedron Lett. 1996, 37(28), 4969-4972. Zhou, X-T.; Lin, Y-R.; Dai, L-X. Tetrahedron Asymm. 1999, 10, 855-862. Viso, A.; Fernandez De La Pradilla, R.; Guerrero-Strachan, C.; Alonso, M.; Martinez Ripoll, M.; Andre, I. I. J. Org. Chem. 1997, 62, 2316-2317. 97 64. 65. 66. 67. 68. 69. 70. Viso, A.; Fernandez De La Pradilla, R.; Garcia, A.; Guerrero-Strachan, C.; Alonso, M.; Tortosa, M.; Flores, A.; Martinez-Ripoll, M.; Fonseca, 1.; Andre, I.; Rodriguez, A. Chem. Eur. J. 2003, 9, 2867-2876. Longmire, J. M.; Wang, B.; Zhang X.; J. Am. Chem. Soc. 2002, 124, 13400- 13401. Gothelf, A. S.; Gothelf, K. V.; Hazel], R. G.; Jorgensen, K. A.; Angew. Chem. Int. Ed 2002, 41 (22), 4236-4238. Chen, C.; Li, X.; Schreiber, S. L. J. Am. Chem. Soc. 2003, 125, 10174- 10175. Oderaotoshi, Y.; Cheng, W.; Fujitomi, S.; Kasano, Y.; Minakata, S.; Komatsu, M. Org. Lett. 2003, 5, 5043-5046. Shanna, V.; Lansdell, T. A.; Peddibhotla, S.; Tepe, J. J. Chem. Biol. 2004 (in print). Tepe, Jetze J .; Peddibhotla, Satyamaheshwar. PCT Int. Appl, WO 2003101969. 98 CHAPTER III SILICON MEDIATED DIASTEREOSELECTIVE MULTI-COMPONENT SYNTHESIS OF IMIDAZOLINES. A. Biological Importance of 2-Imidazolines and Imidazoles Derivatives of 2-imidazoline and 2-imidazole have attracted substantial interest due to their interesting biological activities.1 Imidazolines are known to exhibit a wide range of pharmacological activitiesz, including a-receptor stimulation; vasodepressor activity; a-adrenergic inhibition; and syrnpathomirnetic, antihistaminic, histamine-like, and cholinomimetic activity. Imidazoline derivatives have also been found to exhibit anti-inflammatory], anti-nociceptive, immuno-modulating, antioxidant activities‘. 4,5-Dihydro-lH-imidazoles (2- imidazolines) are useful intermediates for the synthesis of molecules with pharmacological activities such as anti-hypercholesterolemic, anti-diabetic, anti- hypertensive, or anti-cancer activity.3' 5 Cyclic ureas and cyclic thioureas are reported to be potent inhibitors of human immunodeficiency virus (HIV) protease and HIV replication.6 2-imidazolines are also convenient building blocks for the synthesis of pharrnaceutically relevant molecules such as azapenams, dioxocyclarns, and diazapinones.7 Also, suitably functionalized 2-imidazolines are easily converted to 2, 3-diamino acids, which are incorporated in a wide range of antibiotics and other biologically active compounds.8 Streptolidine lactam9 which incorporates a y-hydroxy-a, B-diaminoacid moiety, is at the core of streptothricin antibiotics, which due to their high antibacterial activity have drawn a great deal of synthetic attention (Figure 111-] ). The structures of imidazolines and imidazoles 99 are analogous to those of oxazolines and oxazoles, or thiazolines, and thiazoles, respectively. The latter heterocycles are found in numerous macrolactams isolated from marine sources.lo Imidazolines have been easily converted to imidazoles and incorporated into macrolactam analogues of bistratamide H without loss of stereochemical integrity. H020) H02C rodent central nervous system 1" V5110 @"fi'hYPOfQ'YOBMiC cholecystokinin inhibitor actiwtv In rats NH N\ N~R1 HNJLNH HI 0 . ” Hot-o OJ NH R streptolidine lactam core of streptothricin antibiotics 59131001" 790091“ 89005818 Figure III-1. Biological activity of molecules with 2-imidazoline nucleus. Recent studies have also revealed that the imidazoline ring is a required pharmacophore for certain potent anti-hyperglycemic properties. Imidazoline derivatives, such as midaglizole, deriglidole, and efaroxan, have been identified as promising anti-hyperglycemic agents.4 An imidazoline moiety has previously been incorporated into a known antihypertensive agent, clonidine.‘l During the past decade, the concept of imidazoline (1) receptors has been developed and gained consensus.‘2 Findings from different laboratories have shown that they are widely distributed in different tissues and species and may participate in the 100 regulation of various physiological functions. Moreover, recent evidence suggests their involvement in diverse pathologies, and therefore a more definite knowledge of the structure and function of this receptor system could help in the search for therapeutic agents useful for treating efficaciously a variety of disorders such as hypertension, diabetes mellitus, gastric ulcer, endogenous depression, and 13 stroke. Imidazoline derivatives have been studied as a2-adrenoceptor or 3b’ I" '4' hnidazoline rings with appropriate estrogen receptor modulators. substituents can serve as amino acid replacement in a peptide”, making it less susceptible to proteolytic cleavage and hence improve the stability and therapeutic utility peptides. In fact, the imidazoline scaffold like other rigid five-membered heterocycles can mimic functions of natural peptides and hence are of great therapeutic value."5 Developing small-molecule inhibitors of non-enzyme protein-protein interactions has been a difficult task because binding pockets on protein surfaces involved are shallow and dynamic as compared to enzymes which have well defined and rigid binding pockets or active sites.17 However, the crystal structure of MDM2 bound to a peptide from the transactivation domain of p53 has revealed that MDM2 possesses a relatively deep hydrophobic pocket that is filled primarily by three side chains from the helical region of the peptide.18 The existence of such a well- defined pocket on the MDM2 molecule raised the expectation that compounds with low molecular weights could be found that would block the interaction of MDM2 with p53. One such class was a series of cis-irnidazoline analogs named 101 HO 1m \\/N , 1° 0 / ~<° Om Figure III-2 (a&b): p53 bound to MDM2; key residues for binding (c) Overlay of p53 residues and nutlin (d) The bromo phenyl moiety of nutlin deep in the groove of MDM2 (e) Representative structure of a nutlin. Adapted from Ref. 18 and 19. 102 Nutlins (Figure 111-2).19 The imidazoline scaffold thus replaces the helical backbone of the p53 peptide and is able to direct, in a fairly rigid fashion, the projection of three groups into the pockets normally occupied by Phe”, Trp23, and Len26 of p53. By binding MDM2 in the p53-binding pocket the cis-imidazolines activate the p53 pathway in cancer cells, leading to cell cycle arrest, apoptosis, and growth inhibition of human tumor xenografis in nude mice. This example has served as a paradigm for the study and development of other small molecule inhibitors of protein-protein interactions (SMPPI’s).19 B. Chemical significance of imidazolines The most direct chemical utility of imidazolines has been their conversion to chiral diamineszo, which are widely preferred group of ligands for asymmetric synthesis. There is definitely a need for more synthetic methods, which can provide optically active imidazolines with greater choice of substituents. Hegedus et al have efficiently reacted optically active imidazolines photochemically with a chromium carbene complex to obtain azapenams2| in good yield and high stereoselectivity. C2 Symmetric imidazolines have been shown to induce excellent to modest acyclic diastereoselectivity to form quartemary benzylic centers”, which to date remains a synthetic challenge. MacMillan et al have reported a very 23, as catalysts for highly enantioselective impressive use of irnidazolidinones indole alkylations and Diels-Alder reactions. This represents an important example in the fast growing field of organocatalysis. Furthermore, chiral 2- imidazolines have attracted considerable interest as templates for asymmetric synthesis 24 and as chiral ligands for asymmetric catalysiszs’ 26 and have found 103 wide application as potent N-heterocyclic carbene ligands in organometallic catalysis.27 N-Heterocyclic “carbene” ligands derived from imidazole were recently reported as alternatives for phosphines in organometallic catalysis. The authors claim that unlike the phosphines, the new imidazole ligands do not dissociate from the transition metals and hence provide sturdy catalysts.” C. Lewis acid mediated synthesis of imidazolines The synthesis of stereodefined imidazolines has been scarcely reported despite their usefulness. Methodologies are limited by the difficult availability of the precursors, and for this reason the variety of groups bonded at C4 or C-5 of the ring is quite limited. Most of the imidazolines described are only 4-suhstitutcd28 or 4, 5-disubstituted with the same group (generally phenyl).29 Only a few examples of syntheses of 4, 5-disubstituted imidazolidines (containing a range of groups) have been reported?“ 28¢ 30 As part of our program to develop small molecular weight scaffolds containing a high degree of diversity, we focused on developing a stereoselective synthesis of substituted imidazolines. 1, 3-Dipolar cycloaddition reactions utilizing N- methylated mesoionic oxazolones (or milnchnones) provide a general route for the syntheses of pyrroles and imidazoles.”33 A moderate equilibrium concentration of the mesoionic tautomer is essential for the 1, 3-dipolar cycloaddition leading to heterocycle formation.34 By N-methylation, the azlactones are locked in the mesoionic form, which is extremely reactive with dipoles and affords the heterocycles in very high yields. Imidazoles can be generated by treatment of the mtlnchnones with tosylimines. Addition of the tosylimines results in an unstable 104 bicyclic adduct that loses carbon dioxide and benzenesulphinic acid to gain aromaticity.32’ 35 The phenylsulphonyl residue as leaving group enhances the tendency to aromatize (Scheme III-1). Munchnones with different substituents, at the 2 and 4-positions lead regioselectively to products in which a bond is always formed between the 2-carbon atom of the dipole and the imine nitrogen atom. This attack was explained on the basis of a dipole-dipole interaction between partially negative nitrogen of the imine and the electrophilic C-2 atom of the munchnone. The yield reported for these reactions are low at least partly due to self-condensation of mllnchnones.32 The problem of self-condensation can be suppressed by using a solid-phase approach towards the preparation of imidazoles.33 The munchnone generated on solid phase was condensed with tosylamines to afford the imidazoles in good yields and purities. | N-SO Ph P“ o e ’11 2 Ph N CPO?“ N R2 Y / 0 R2 0% .N 3 - 002 ’41 Me ,N ———> U H ——-> ph N R1 Scheme III-1. Addition of the tosylimines leads to loss of CO2 and PhSO2H to gain aromatrcrty. Surprisingly, the utilization of oxazolones (or azlactones) has not yet resulted in an efficient entry into a stereoselective highly diverse class of imidazoline scaffolds (Refer Chapter 2). It has been well documented that azlactones undergo a Staudinger-type reaction when heated with imines in toluene yielding B- lactams.”38 The azlactone, which is in equilibrium with valence tautomeric 105 ketene intermediate, undergoes a [2+2] cycloaddition reaction with the imine (Scheme III-2). It can be concluded that either mesoionic oxazolium oxide, which has the nitrogen atom unsubstituted, is not in reasonable concentration and/or the imine nitrogen is not nucleophilic enough. Hence formation of ketene takes predominance, which reacts readily with the imines to afford the B-lactam.”38 11 Ph NH H ’1: Ph 0 ph NA toluene ~l ‘ H :(O + OPhJNI Ph * J ... Ph/ILN NvPh reflux ,._~ / s H o CH2Ph 0 ll-1 lll-I-‘l O Ill-A Ill-B Ph Ph CH Ph NH 1 2 O p PHCO H N Ph ’ /\ H30 11 H \( C + N| PD ....... N—< P11) N6) CH3 Ph 90 CHzPh 0 methyl outward or exo lite l Ill-B Ill-A Scheme III-2. Staudinger reaction imines and ketenes generated from azlactones. The S-oxazolones (azlactones) in this study were prepared from different N-acyl a-aminoacids by EDCI-mediated dehydration to provide the pure oxazolones in high yields.” 4-methyl-2-phenyl-2-oxazolin-5-one (II-1) synthesized from N- benzoylalanine, when heated with N-benzilidene-benzyl amine in toluene under N2 atmosphere to reflux, gave B-lactam III-A and the amide III-B.4o The azlactone is in equilibrium with valence tautomeric ketene intermediate, which then undergoes a ketene-imine or the Staudinger“1 type reaction to form the B- lactam (III-A). The amide III-B is obtained by hydrolysis of the uncyclized 106 zwitterionic intermediate (Scheme III-2). Theoretical studies have predicted that torquo-electronic effects play a dominant role over steric effects in the cycloaddition of the zwitterionic intermediate to the B-lactam III-1a. 42 The imine preferably adopts the more stable (E) geometry because of the bulky aromatic substituents and the methyl group (positive inductive effect) on the ketene adopts an “outward” or exo position to the direction of attack by the imine. Electron withdrawing benzamido substituents adopts an endo or “outwar ” position in accordance with the predicted model for these cycloadditions. A conrotatory electrocyclic ring closure affords B-lactam with cis orientation for the methyl and phenyl groups in 30 % yield as reported before.“l RJ C,0 _>R1/u—NH fig .1 w" Toluene R2 ‘R4 heat R3 ISO-50% o 0 R1‘<\NI R2 R J’Rt R. R 1 o 3 L» tree . H :I Lit/G) R2 CO2” Scheme III-3. Lewis acid mediated generation of munchnone and cycloaddition We envisioned that a Lewis acid coordination to nitrogen of the azlactone would increase the equilibrium concentration of munchnone intermediate and mediate the azomethine ylide-imine cycloaddition reaction. The choice of the imine was important as unactivated or electron rich imines have rarely been used with mllnchnones before (tosylimines lead to enhanced aromatization). Only one 107 example where N-benzylidine-benzylamine (III-I-l) was used as the imine is by Arndtsen and co-workers.43 They used Pd-catalysis to couple basic building blocks like an imine, acid chloride and carbon monoxide to generate N-alkylated munchnone in situ. Mild acidic conditions were suggested to accelerate the reaction to afford the products as N-alkylated imidazolium salts. The main shortcoming for this method is that only symmetrical imidazolines can be obtained as two equivalents of the imine are involved in the synthesis, one for generation of munchnone and other for cycloadditions. Also, only electron rich aromatic aldehydes can be used. Electron deficient imines and phosphine ligands, both stop the cycloadditions completely. One advantage was that the imidazolium salts were easily isolated from the reaction mixtures by precipitation fiom ethereal solutions. 55 °c [Pd2(dba)3J (5 mol %) Ph R2 0 2, 2'-bipyridine (10 mol %) Rt-(aékwla, )_.___N\ + /U\ + co > H R1 R3 Cl CH3CN Rz/thco? Figure III-3. Generation of imidazolium salt through Pd-catalysis. D. Lewis acids for in situ generation of miinchnones and cycloaddition With one equivalent of SnCl4, TiCl4, ZnClz, silver triflate only degradation was observed in the reaction of 4-methyl-2-phenyl-2-oxazolin-5-one (II-1) and N- benzilidene-benzyl amine (III-I-l) in toluene. Harder lewis acids such as TiCl4 and BF3-OEt2 or protic acids such as camphor sulfonic acid (CSA) and methyl sulfonic acid (MSA) did not promote any product formation (Scheme III-4). Cu 108 and Al based lewis acids or lanthanides were not encouraging either. Only with TMSCl, on refluxing in toluene a White precipitate separated from the reaction mixture in 75 % yield, and was isolated and characterized as the imidazoline—4- carboxylic acid (III-l-l, Figure III-4). fl-lactam (in CDClg N H ,tPh Z d— s o CHzPh III-A Imidazoline (in DMSO-dd Bn 3.; Ph Ph—(\ IMe N cozH lll-1-1 ITTTT[TYYTIrTTYl'YYYYIYYYTTITYY IIVIIYIYVUfTYIYI T T Y Y 10 9 8 7 6 5 4 3 2 I Figure III-4. (a) 1H spectra for a representative B-lactam (III-A) and imidazoline (III-l-l). 109 B-Iactam: IR (car‘) 1751, 1654. 0 JL Ph NH I’r,'I ‘\\ Ph I i 4 o ‘CHzph Ill-A MM -lil--l 2i ww w 'v - w I I I I I I I I I I I I I T I I I I I I I I I I I I I 200 175 150 125 100 75 50 25 Imidazoline: IR (cm'1) broad 3600-2500, 1734, 1610. 3'! N Ph Ph—<\ |,.Me N 00214 lll-‘l-1 1‘ ‘T ‘ 5V. ‘3, “AC. ‘ ,w _.. ,f w “ ,2,in W Z, ___L .7. ,‘,_w ,, lw‘ LQAVV LJWJYP‘T‘ ‘22 I I I I I I I I I I j I I I I I I I I I I I j T I I I I I I I I I I I I I I I 200 175 150 125 100 75 50 25 Figure III-4. (b) 13C spectra for a representative B-lactam (III-A; in C130,) and imidazoline(III-1-1; in DMSO-dg). 110 c1151 o: o 0 o III-1-7 Figure III-4. (c)cht: X-ray crystal structure of III-l-l. Right: X-Ray crystal structure of III-l-7. The relative stereochemistry has been established unambiguously by X-ray crystals structure of III-l-l and III-1-7 (Figure 111-4). The remaining filtrate is made up of 15-20 % of the B-lactam (Scheme 111-2, 111- la). Subsequently, the same results were obtained in THF and dichloromethane as solvents. We were encouraged that TMSCl promotes the reaction of azlactones and imines to afford the imidazoline-4-carboxylate scaffold in very good yields as 111 a single diastereomer, with trans- relation between C4 and C5 substituents (Scheme III-4). Ill-H Ph N. Ph 0 V 80 F"‘_\ 11" LA N N —-—> Pit—s . 0112012 N 111 C02“ ' 111-1-11- LA 96 yield LA % yleld “Ch 0 msc1 70 811014 0 ch12 0 1 eq.TMSC|/1 eq. 5th 0 EtzAlCI 0 2 eq. TMSCI I 1 eq. £th 0 CuCl 0 T5801 70 31°Phl3 0 PhasiCl 45 A90" 0 CHgCOCl 55 “3'3 ° CSA 0 ZrlO-Prlt 0 TMSOTf 0 Scheme III-4. Lewis acid screen for the synthesis of imidazolines. E. Possible mechanism for synthesis of imidazoline While the complete mechanistic details of this process are still under investigation, the reaction does not seem to proceed via a ring-opened nitrilium ion intermediate (Scheme III-5).“ ‘5 A common method to trap nitrilium ions is by nucleophiles like water or methanol. After addition of TMSCI, the reaction mixture was refluxed with saturated sodium acetate solution. The azlactone was obtained intact and not even a trace of O-acetylimidate was observed. The possibility of a step-wise Michael-type addition (via the formation of the nitrilium 112 ion) and subsequent cycloaddition was investigated by first preparing the silyl enol ether46 with TMSCl (1.0 equiv) and TEA (1.0 equiv) followed by the addition of the imine and an additional one equivalent of TMSCl.‘7'5° R1 0 0 TMSCI 0W5 N=\ R1 \ _IEA_. R:lt—<\ +fi3 ~(N CHzclz R R2 2 TMSCI R4 R TMS-N R2 H o N H R3 _ 2 Fit-=87???“ ——» R...- I R3 cozms N COzH nitrilium ion Scheme III-5. Nitrilium ion intermediate is not involved in the formation of Imidazolines This resulted merely in isolation of starting materials. Excess of triethylarnine also halted the reaction suggesting that acidic conditions were required. TMSOTf (likely O-silylation) also did not result in any product formation. This indicates the requirement of a nucleophilic counterion to establish possible equilibrium between O-silation and N-silation and a probable C-silation of the azlactone (Scheme 111-6). The role of counterion in establishing such equilibrium has been reported by Wilde in his work with N-acyl munchnones." The generation of azomethine ylides by “silatropy " has recently also been reported by Komatsu and co-workers.”'54 The authors report the formation of an azomethine ylide after a 1, 2-silatropic shift of or-silylimines resulting in the formation of pyrrolidines as a mixture of stereoisomers. 113 A §1M93 MfiSiYNVRZ = GrgWRz R1 1,2-311atropy R1 ot-silyl imine azomethine ylide Komatsu's approach 0 OSiMe3 o_§\ M63$iCI O \ __‘ ... R1/k\ R2 R1/A\N R2 ”0' Me38iCl 1 M93810“ 0 0 M SiCl ° ’g-KQ + H01 4:; O SlMea+ HCI R1 9 '1‘ R2 Ri/Q R2 SiMeg N N-silyl munchnone Our approach Scheme III-6. Generation of azomethine ylides by silatropy Johnson et al have investigated N-metalated azomethine ylides formed via “soft enolization” of (imino)glycine esters as alternatives to ylides generated from the same compounds in the classic thermal [1, 2]-prototropy manifold (Figure III-5), which require a weak base working in concert with a metal salt.44 In light of these findings, we proposed that the reaction proceeds via a 1, 3-dipolar cycloaddition (Scheme III-7). While N-silyation results in reactive munchnone, acidic conditions, during the reaction activate the imine. This double activation should result in accelerated 1, 3-dipolar cycloadditions affording the bicyclic intermediate. NMR studies in CDC13 at elevated temperatures (not shown) indeed confirm the rapid consumption of azlactone in presence of the dipolarophile. The azlactone does not show any appreciable change in presence of TMSCl, if not heated with dipolarophile. 114 Lewisacid . R Mlld base I sz N YCOZR‘ = R2V$®COQR1 H via 1,2-prototropy R = H, Metal General approach H OMe RZYNYCOZMG MQCIZ Rng 9 \0 0151 002m: 31:10 \0’ M9012 Johnson's approach Figure III-5. Sofi enolisation vs. 1, 2-Prototropy Other silyl chlorides such as triphenyl silyl chloride and triethyl silyl chloride also provided the product as a single diastereomer, in notably longer reaction times and lower yields (Scheme 111-4, 45% and 70%, respectively). Additional support of the proposed mechanism is shown in Scheme III-7. We found that one equivalent of acetyl chloride resulted in the formation of the imidazoline as well, albeit in somewhat lower yields (Scheme Ill- 4, 55%)." o R" ° f o TMSCI R1 1) R1‘<\ I = N R R3 N R2 ms 2 - R4 TC? . H RI; E" R3 H20 N E R3 Rt-<\ . R ‘— R‘_<(3 "R2 o\ 2 ’ N 00211 ms 602” c? Cl Scheme III-7. Proposed mechanism for the formation of imidazolines via the l, 3 dipolar cycloaddition 115 F. Diversity The cycloaddition reactions proceeded well with a wide variety of imines at slightly elevated temperatures (dichloromethane, reflux) to provide the highly substituted imidazolines in very good yields. Only the trans diastereomers (with respect to R2 and R3) of the imidazolines were observed in almost all cases, as determined by NOE experiments and X-ray crystallography (Table 111-1). A range of structurally diverse imidazolines were prepared and are listed in Table III-l and -2. We have recently reported this highly diastereoselective multi-component one- pot synthesis of substituted imidazolines.” 56 These low molecular weight imidazoline scaffolds contain four point diversity applicable to alkyl, aryl, acyl, and heterocyclic substitution (Table III-1) R4 groups/amines: 4-methyl-2-phenylazlactone (II-1) derived from N-benzoyl alanine was reacted with a host of imines (lII-I-l - III-I-14) to afford imidazolines in moderate to good yields. Benzyl, aryl and aminoacid ester derived imines of benzaldehyde could be used with good yields of imidazolines. Electron deficient 4-fluoroaniline as well as electron rich 4-methoxyaniline could be used successfully. However N-benzhydryl group is too sterically encumbered to afford any imidazoline (entry III-l-6, Table III-1). R3 group/aldehydes: Although aromatic aldehydes generally afforded cleaner reactions and higher yields, it is probably a reflection on the purity of imines derived from other aldehydes and not the cycloadditions. Benzaldehyde, p- anisaldehyde (4-methoxybenzaldehyde) derived imines reacted in very good yields. Electron deficient ethyl glyoxalate and 2-nitrobenzaldehyde can also be 116 used and afford good yields for imidazolines. Among the heterocyclic aldehydes, pyridine 4-carboxaldehyde was a good substrate, while with unprotected pyrrole- 2-carboxaldehyde or indole-3-carboxa1dehyde, imidazolines were not obtained at all. Aliphatic aldehydes could not be used because the imines suffered from possible enamine formation were not obtained in sufficient purity. We have also been able to extend the method to 5-unsbstituted imidazolines with the use of formaldehyde imines formed in situ by Lewis acid mediated cracking of triazines [data not shown]. R2 group/amino acids: The diversity in R2 substituents result from the use of various amino acids. Both natural and unnatural amino acids which are suitably protected can be utilized for the formation of azlactone component of this cycloadditions. Glycine could not be used because the corresponding 2-phenyl-5- oxazolone does not enolize under the current conditions. Simple or-alkyl amino acids; alanine, valine, isoleucine and phenylalanine can be used (Table 111-2) and their corresponding 2-phenyl-4-a1kyl-5-oxazolones result in excellent yield of imidazolines with N-benzyllidine-benzylamine (III-I-l). Phenylglycine (unnatural amino acid), tryptophan, and glutamate (acidic) have also been used with good results. R1 group/acyl halides: Phenyl and substituted phenyls, alkyl and benzyl groups could be introduced. Heterocyclic groups like 2-furyl and 2-pyridy1 groups could not be used as the corresponding N-acyl-ol-amino acids could not be cyclized to the azlactones. 117 2‘2" R4 NHz Ph\(° Rcho :Phxz‘ Ph\«N ZR; TMSCI (1.3 eq. ) COZH Me DCM ICOZH azlactone ”flux tren cis s 1H Ill-1a Ill-1b 96 Yield Ratlo Entry R3 R‘ a mi: lll-1-1 Ph Bn 75 >95:5 III-1-2 4-methoxyphenyl Bn 78 >95:5 Ill-1-3 Ph 4-fluorophenyl 74 >95:5 Ill-14 4-pyridinyl Bn 76 >95:5 Ill-1-5 -COzEt 4-fluorophenyl 72 >95:5 111-1-6 Ph benzhydryl 0 — Ill-1-7 Ph -CH2C02Me 7O >95:5 lll-1-8 Ph -CH(CH3)C02Me 66 >95:5 Ill-1-9 Ph -CHZCH2C02Me 51 >95:5 Ill-1-10 Ph Bn 75 ethyl ester 111-1-11 4-pyridiny1 Bn 76 ethyl ester Ill-1-12 Ph Bn 79 alcohol III-143 Ph H 70 >95:5 Ill-144 Ph 4-methoxy phenyl 50 >95:5 Table III-1. Variation of R3 and R4 groups 118 Derivatives: The imidazolines can also be suitably derivatized into esters, alcohols or deprotected to afford N-unsubstituted imidazolines in excellent yields (entries III-1-10, III-l-l 1, III-1-12, lII-l-l3, Table III-1). 13.-NH: 2‘ R1\«OR RacHO :R1\«E‘IZC::H R1\« R3 TMSCI(1.3eq.) N_Z R2 reflux OCOZH azlactone trans2 053 II Illa lllb Entry R1 R2 R, R. 11 Y1eld(ll) $6 Yleld Ratlo (Illa) (atb) 111-1 -1 Ph Me Pb Bn 75 75 >95:5 Ill-2-1 Ph Ph Ph Bn 70 65 >95:5 111-24 Ph Ph 4-pyridiny1 Bn 75 55 >95:5 "1-2-5 Ph Ph £0261 4-fluorophenyl 75 68 >95:5 Ill-34 Ph indolyl-S-methyl Ph Bn 80 68 >95:5 lII-4-1 Ph -CH2CH2002Me Ph Bn 69 60 >95:5 111-5-1 Me Me Ph Bn 60 74 50:50 Ill-6-1 Bn Ph Ph Bn 90 30 75:25 Ill-9.1 Bn Me Ph 8n 76 15 67:33 4-methoxy . 111-11-1 pm“ Me Ph Bn 75 62 >955 lll-12-1 Ph iso-Propyl Ph Bn 90 71 >95:5 Table 111-2. Diversity of various groups on the imidazoline scaffold G. Proposed origin of Diastereoselectivity The diastereoselectivity appears to primarily arise from the steric interaction of the bulky silyl group of the azlactone and the R3 moiety of the imine (Scheme III- 8). This results in the imine approaching from endo type of orientation (with 119 respect to R3) of the racemic azlactone resulting in the trans-isomer as the major/sole product. The electronic effects of the R. position also appear to play a significant role in the reactivity of the azlactone as well as the diastereoselectivity of the reaction. SK H R2 N. C02H Major (”R.; R1_(N (”KO .\‘R3 \Si :\I\R3\ R2 M. COZH lnor Scheme III-8. Origin of diastereoselectivity Reducing the resonance of the stabilized carbocation of the dipolarophile by changing R1 from a phenyl to a methyl or benzyl group resulted in a significant decrease in diastereoselectivity (Table 111-2). With R1 was a benzyl group, there was significant erosion of diastereoselectivity. But it is clear that substituents R2 still controls the outcome. Bulky phenyl as R2 results in a 3:1 ratio in favor of the trans-imidazoline, while when R2 is methyl, a 3:2 ratio of diastereomers was observed. The more reactive and unstable 2, 4-dimethylazlactone provided a 1:1 mixture of the cis- and trans-products. Complete switching of diastereoselectivity to the cis-imidazoline was seen when the positions of the methyl and phenyl groups were exchanged at C2 and C; (data not shown). Figure III-6 shows the Chem 3D minimized structures of azlactones with Me and Ph groups at C2 and C4 positions. It is clear that the steric volume needed by a C2 methyl results in 120 blocking the approach of E-imine required for the trans product. In comparison, the phenyl group can be rotated out of plane and hence is more directional and makes C2 less hindered for approach of imine to result in trans (major) product. Figure III-6. Chem3D models for minimized structures of TOP (lefi) :2, 4 dimethylazlactone, TOP(right): 2-Methyl—4-phenyl azlactone, BOTTOM: 4- methyl 2-pheny1 azlactone H. Synthetic methods for chiral imidazolines In contrast with more traditional lewis acids, the application of organosilicon compounds as lewis acids in selective organic synthesis has a relatively brief history.57 Chiral silicon lewis acids are rarer to find in literature because of difficulty associated with their generation. Hence we decided to follow some conventional methods to get our hands on enantiomerically pure imidazolines. 1) Resolution of Enatiomers. 121 In view of the biological importance of imidazolines, it was important to evaluate bio-activity of the separate enantiomers (chapter V). In order to expedite this study the diastereomeric menthyl esters of imidazoline III-l-l were prepared (Scheme 111-9) and separated by column chromatography. Although the Rf values for the diastereomers are very close, one of the diastereomers was separated clean in very small amount from the column and was subjected to hydrolysis with LiOH to explore the feasibility of this step (data not shown). The hydrolysis did not undergo as planned with the mild base and use of menthol was abandoned to screen for a chiral alcohol that would allow a better separation of diastereomers by column chromatography. R‘N R‘N R3 EDCI. HCIIDMAP R1_<\ REFER: > R‘—<::§; + R‘—<‘ :lR: COOH fiat-I £0 7:0 O O\Z%Z separate and d e (1R. ZS. 5R) -(-)-Menthol R: 1 R4 W W N R3 N 3: R‘—<‘~ ...R, R.—<\ :La, 0 7:0 H0 H0 Scheme III-9. Resolution of racemic imidazolines using (-) Menthol The use of (+) R and (-) S-l-phenylethanol resulted in better separation of diastereomers (chapter V) and racemic imidazoline (III-2-l) was resolved using (+) R-methyl benzyl alcohol. EDCI.HC1 mediated esterification of the racemic imidazoline III-Z-l and the auxilliary resulted in a mixture of diastereomers, which were separated using column chromatography. Hydrolysis of the diastereomers resulted only under reflux with 2N NaOH to give enantiopure 122 imidazolines (-)III-2-l and (+)IlI-2-l. Compound (+)III-2-l was successfully crystallized in CHClg/octane and the x-ray structure is shown in Figure III-7.58 Even though we were able to successfully resolve the racemic imidazolines by converting to diastereomeric esters using chiral alcohols and subsequent hydrolysis, getting the cycloaddition would be more desirable because of the application for the imidazolines (refer chapter V). 1) Me Ph W Ph Ho’\© PhW Phj Ph_ '3th Ph N pr.) CHZClz, reflux N -.,’ 0025' ".1 Ill-I-1 2) (00002. EtOH (i) III-1.10 Scheme III-l l. Asymmetric induction by chiral acid chloride ii. Chiral amino acid derived R1 We decided to use an amino acid as a chiral auxillary to influence enantiocontrol via the azlactone. Coupling of N-cbz-L-valine with L-alanine methyl ester and subsequent hydrolysis with 2N NaOH resulted in the dipeptide N-cbz-L-val-L- ala-CO2H. The dipeptide acid was then cyclized with EDCI.HC1 to obtain the corresponding azlactone (II-l0), which was allowed to react with N- benzylidinebenzylamine in the usual conditions. One diastereomer has been isolated from the reaction mixture and characterized, demonstrating that this is a feasible method. Removal of the chiral auxillary is not possible easily. However, 124 the product is an imidazoline based amino acid and Kelly and co-workers have used similar strategy to develop enantiopure imidazoline-amino acids and incorporated them into biologically relevant macrolactams.lo /\OJL ONIKOO EDCI. HCI Ph/\ /\O/u\ OLOO Y‘LOH 78 DC% «40 rah/\NIk ——"n'."s"éi(1§" , Ph°i1r§n . eq. DCM. reflux C02” III-104 Scheme III-12. Amino acid at R. position as a chiral template iii. Chiral amines as auxillaries Viso et a! reported the very first examples of a highly diastereoselective 1,3- dipolar cycloadditions of azomethine ylides derived from a-iminoesters with chiral sulfinimines59 to produce N-sulfmyl imidazolidines (Figure III-8) and transformation into non-symmetrical vicinal diamines.49 Dipole generation under standard conditions using LiBr, Et3N, MeCN or AgOAc, DBU, toluene had failed. Use of LDA allowed for successful cycloadditions with a high degree of endo stereocontrol. 125 Ph ”@N’) THF. 20h + _ N’S“o 0 -78 to4 °c )I OMe Viso's appmach N R Ph’Ng "\ OMe o Li'—O . : , 1 LAH NH &57 I / —>/©::J\( (P‘P'LNA ) . 2 NHz N=\Ar MR 2) TFA Ar "'R COzMe H0 endo approach major en a I' Figure III-8. Use of chiral sulphinimes in azomethine ylide cycloaddition Addition of BF3.OEt2 results in abrogation of cycloadditions pathway and the reaction proceeds with opposite diastereoselectivity via a step-wise mechanism.so We realized that the above observations by Viso and co-workers would perfectly suit our purpose of generating additional reagent control on stereochemical diversity of the imidazoline scaffolds. The chiral sulfinimines were readily prepared from commercially available chiral p-toluenesulfinamides using reported procedure.49 Starting from S-(+)-p-toluene sulfonarnide and benzaldehyde, the imine (III-I-l4) was prepared and used for cycloaddition with 2-phenyl-4-methylazlactone (II-1). It was surprising, that the sulfinyl group was lost under the acidic reaction conditions and we isolated N- unsubstituted imidazoline (III-l-l3) in 30 % yield afler precipitation from DCM/ether (Scheme III-13). Since, it was not clear whether the chiral auxillary falls off during or afier the cycloadditions, the optical rotation was measured and was not very promising. 126 Q's". PhCHO Q's " ‘NHZ TI(OEt)4 (4 eq.); ‘N DCM, reflux ' 86 % Ph S(+)-p-toluenesulfinamide "”43 O a 8’; W? TMSCI (1. 3 eq.:) :30ng DCM, reflux "'1 [a1200=+ 3.0 (c= 5, MeOH) ""143 Scheme III-13. Use of chiral sulfmimine in munchnone cycloaddition. l-Phenyl ethylamine was also tried as a chiral auxillary“ but in accordance with the stereochemical model proposed above for these reactions, very poor conversions were seen. Moreover, azlactone (II-l) resulted in 3:2 mixture of diastereomers (data not shown) which were difficult to separate. The difficulty of separation combined with the difficulty in selective removal of the chiral 1- phenylethyl group in presence of other benzylic sites made us abandon this approach. 1. Synthesis of diamino acids and other scaffolds The most direct chemical utility of imidazolines has been their conversion to chiral diamines,20 which are widely preferred group of ligands for asymmetric synthesis. Efforts towards exploitation of the in-built diversity for synthesis of chiral diamines and introduction of heteroatom functional groups directly on the imidazoline ring to afford cyclic ureas (2-oxoimidazolines), 2-aminoimidazolines and 2-thioimidazolines have been planned as extension of the methodology we have developed for imidazolines (Scheme III-l4). This “libraries from libraries” concept has been frequently in combinatorial chemistry and drugsdesign.62 127 R1HN 2-aminoimidazoline YICROZHR‘ '34 / HN Ph R3 \(ICROZH ”2" .9 C02H \ R2 * Gig-rm 2-oxomidazoline RCOZH imidazoline diamino aCId R1S\(N NICRozH 2-mIoimidazoII'ne R2 Scheme III-l4. Extension of methodology for other templates Kohn and Jung” have developed a stereospecific route to vicinal diamines from imidazolines. F ormamidine was obtained by catalytic hydrogenation of cynamide under acidic conditions with palladium on charcoal. Cyclization to imidazoline by NaOMe followed by mild alkaline hydrolysis with alcoholic aqueous NaOH afforded the diamines from starting alkenes in good yields (Figure III-9). a, 3 NHch 1% PdlC Br .3 o—.‘ : ‘\\‘ l \ N35 5; :lHCN—> AcOH-MeOi-I NH HN=/ MeOH ; \w III : "HM NV” 0.25 M “2" NH: NaOH Figure III-9. Synthesis of vicinal diamine from imidazolines l) 2-unsubstituted imidazolines. N-formyl-alanine was cyclized to 4-methylazlactone (II-7) using EDCI.HCl and 2-unsubstituted imidazoline (111-7-1); (1:1 transzcis, with respect to R2:R3) was 128 made from 4-methylazlactone and N-benzylidinebenzylamine(III-I-l) using the established cycloaddition conditions. Normal silica-gel chromatography was ineffective in separating the diastereomers. Even derivatization to ethyl esters did not yield good separation. 2) 2-trifluoromethylimidazolines. Woodbum and J ohnson64 have reported that when trifluoroactonitrile is reacted with ethylene diamine, 2-trifluoromethylimidazoline is obtained. When they tried to isolate the hydrochloride salt of the imidazoline from a non-anhydrousrous ether solution, they isolated the diamine which shows the lability of the trifluoromethyl group to hydrolysis. With this precedent we set out to make 2- trifluoromethyl imidazolines using our route. N-trifluoroacetylalanine was made from alanine using reported procedure65 in 90 % yield. Cyclodehydration of N- trifluoroacetylalanine using EDCI.HC1 afforded the 2-trifluoromethyl-4-methyl-5- oxazolone (II-8). But the oxazolone is in equilibrium with the pseudooxazolone. The equlibrium is in favour of the pseudooxazolone.66 3) 2-amino-S-oxazolones. At the onset, we tried to establish a common method to get to 2-amino-5- oxazolones with different substituents. The starting materials, the ureidoamino acids could be made by coupling an amine with isocyanate of an amino acid ester. Alanine methyl ester gave a volatile isocyanate, which was not practical to use. Hence the synthesis began with phenyl glycine methyl ester, which when treated with phosgene67 in a biphasic CH2Cl2/sat.NaI-ICO3 (aq.) system yielded the pure isocyanate in almost quantitative yields. This isocyanate was then treated with 129 various amines and amides. The ureidoacids were then cyclodehydrated with EDCI.HCl to obtain the 2-aminoazlactones (Scheme III-15). The azlactones were condensed with imines in the presence of TMSCl under standard protocol and results are listed in Table III-1. Ph COCI; Ph 0 Ph sat. Mal-1003: 0\\ ‘ )\ R1NH2 JL )\ HzN COzMe DCM ‘N 002M9J v R1HN ” C02Me 0 /\ A 8‘ R HN Ph \ N EDCI.HCI _ ‘ \E‘Z‘o Ill-:1 Pb R‘HNW g :03 H T .............. ’ 00” Ph TMSCI N Ph 2 . DCM . . . . 2-amlnoazlactone reflux 2-amIn0ImIdazolIne Scheme III-15. Synthesis of 2-aminoazlactones and 2-aminoimidazolines R,HN\«O o R1NY0 0 Ph Ph R1NH2 oxazolone 2-amlnolmldazollnc all”? 1844 cm-1 not Isolated O G/KNHZ, 1840 cm-1 not isolated 0 (Dim-l 1844 cm-1 not isolated 0 Scheme III-l6. Tautomers of 2-aminoazlactones The main problem encountered was isolation of these azlactones, because on concentration they led to multitude of products. Tautomerism with the iminooxazolidinones (Scheme III-16) can be expected to make the aminoazlactones highly unstable and moisture sensitive. It is possible that they 130 hydrolyse to the N-carboxy anhydrousrides and hence do not participate in cycloadditions.68 J. Ring opening of 2-phenyl imidazolines. The imidazolines resisted direct reduction with sodium borohydride and with stronger hydride reducing agents like LAH, the 4-carboxylic acid group was reduced to the corresponding alcohol. Thus imidazoline III-l-l on reduction with LAH (0.3 eq) in THF resulted in 90 % yield of alcohol (111-1-12). This suggested that C2 position in imidazolines is resonance stabilized and not sufficiently electrophilic. Zhou and co-workers have reported successful ring opening of simple 2-arylimidazolines to the corresponding diamimes.69 They first alkylated the iminic nitrogen with methyl iodide to obtain N—methyl-Z-arylimidazolium iodides, which then underwent a smooth reduction ring opening with sodium borohydride in ethanol to afford the diamines in high yields (Scheme III-17). P11 P" Ph anhydrous N Ph Ph K co NaBH4 (5 eq.) HN 1’" Ph~250 °C (dec.). HRMS (El): m/z calcd for C32H26N302 [M — H], 484.2025; found, 484.2011; IR (neat): 3420 cm", 1741 cm". Ph 3 Ph N P11 00211 III-44 002Me dl-(4S,5S)-1-Benzyl-4-(2-methoxycarbonyl-ethyl)—2,5-diphenyl- 4,5-dihydro- lH-imidazole-4-carboxylic Acid (2r) A solution of benzaldehyde (0.252 g, 2.4 mmol), benzylamine (0.258 g, 2.4 mmol) in anhydrous CH2C12 (100 mL) was refluxed under nitrogen for 2 h. 3-(5- Oxo-2-phenyl-4,5- dihydro-oxazol-4-yl)-propionic acid methyl ester II-4 (0.5 g, 2 mmol) and chlorotrimethylsilane (0.282 g, 2.6 mmol) were added and the mixture was refluxed under nitrogen for 6 h and then stirred overnight at r.t. The reaction mixture was evaporated to dryness under vacuum. The product was precipitated using CH2C12/ hexanes (1:1); yield: 60% (0.54 11); white solid. 'H NMR (300MHz) DMSO'd6: 8 2.05—2.25 (2 H, m), 2.30—2.50 (2 H, m), 3.55 (3 H, s), 4.38 (2 H, ddd, J1 = 4 Hz, J2 = 9 Hz, J3 = 25 Hz), 4.86 (1 H, q, J= 3.3), 7.10-7.60 (12 H, m), 7.70—7.90 (4 H, m) '30 NMR (75MHz) DMSO—d;: 8 27.6, 30.1, 43.3, 51.6, 52.7, 127.1, 127.2, 127.3, 128.2, 128.3, 131.5, 131.6, 133.3, 137.8, 167.5, 150 171.4, 173.6; HRMS (FAB): m/z calcd for C27H27N204 [M + H], 443.1893; found, 443.1971; IR(neat): 1734 cm", 1653 cm". Ph W N Ph BnAg—ZCOgl-l Ph Ill-G-‘I dI-(4S,5S)-l,2-dibenzyl-4,5 diphenyl-4,S-dihydro—lH-imidazole— 4-carboxylic Acid (2s) A solution of benzaldehyde (0.6 g, 5.7 mmol), benzylamine (0.61 g, 5.7 mmol) in anhydrous CH2C12 (120 mL) was refluxed under nitrogen for 2 h. 2- Benzyl-4-phenyl-4H-oxazolin-5-one (1.43 g, 5.7 mmol) and chlorotrimethylsilane (0.8 g, 7.4 mmol) were added and the mixture was refluxed under nitrogen for 6 h and then stirred overnight at r.t. The product was a mixture of diastereomers (3:1 ratio); yield: 60%. The above trans-diastereomer was obtained by repeated precipitation from MeOH—Et20. ‘H NMR (300MHz) DMSO-d;: 8 3.47 (2 H, d, J = 15.6 Hz), 4.31 (2 H, d, J = 15.6 Hz), 5.80 (l H, s), 6.40—7.40 (20 H, m) l3C NMR (75MHz) DMSO-d;: 8 30.7, 47.5, 72.1, 100.2, 126.2, 126.6, 126.8, 127.3, 127.7, 127.9, 128.0, 128.5, 128.6, 129.0, 131.4, 132.5, 133.4, 136.4, 164.4, 171.6a HRMS (FAB): m/z calcd for C30H27N2O2 [M + H] 447.2010; found, 447.2072; IR(neat): 3350 cm", 1704 cm". 151 Ph 3 N P11 C02H lIl-9-1a dl-(4S,5.S')-l,2-Dibenzyl-4-methyl S-phenyl4,5-dihydro-lH-imidazole- 4- carboxylic Acid III-9-1a and dl-(4S,5R)-1,2 Dibenzyl-4- methyl-5-phenyl-4,5- dihydro-lH-imidazole-4-carboxylic Acid II-9-lb: A solution of benzaldehyde (1.13g, 10.58 mmol), benzylamine (1.13g, 10.58 mmol) in anhydrous CH2C12 (250 mL) was refluxed under nitrogen for 2 h. 2-Benzyl-4-methyl-4H oxazolin-S- one (2g, 10.58 mmol) and chlorotrimethylsilane (1.48 g, 10.58 mmol) were added and the mixture was refluxed under nitrogen for 6 h and then stirred overnight at r.t. The reaction mixture was evaporated to dryness under vacuum. The product cis isomer III-9-lb was precipitated using CH2C12-Et20; yield: 30% (0.6 g); white solid. The trans-isomer III-9-la was then precipitated out from the mother liquor. A small amount was then reprecipitated to remove traces of III-9-lb; yield: 17% (0.15g). III-9-1a: 1H NMR (300MHz) DMSO-d6: 8 1.74 (3 H, s), 3.67 (1 H, d, J= 15.3 Hz), 4.11 (1 H, d, J= 14.7 Hz), 4.38 (1 H, s), 4.46 (1 H, d, J= 14.7 Hz) 4.59 (1 H, d, J = 15.3 Hz), 6.77 (2 H, d, J = 7 Hz), 7.00—7.60 (13 H, m) 13C NMR (75MHz) DMSO-d;: 8 22.1, 32.4, 48.1, 70.6, 71.1, 127.6, 128.3, 128.6, 128.9, 129.1, 129.2, 129.7, 132.2, 132.8, 133.5, 164.8, 175.1; HRMS (FAB): m/z calcd for C25H25N2O2 [M + H], 385.1898; found, 385.1916; IR (neat): 3350 cm", 1624 cm". 152 Ph 1 Bn/\« Ph N "“COzH Ill-94b 111-9-111: 1H NMR (300MHz) DMSO—d;: 8 1.14 (3 H, s), 3.94 (1 H, d, J = 15.6 Hz), 4.24 (2 H, q, J: 8.7 Hz), 4.56 (1 H, d, J= 15 Hz), 5.74 (1 H, s), 6.65 (2 H, d, J = 7.5 Hz), 7.00—7.40 (13 H, m) l3c NMR (75MHz) DMSO-d;: 8 27.1, 38.7, 48.6, 71.5, 73.5, 127.9, 128.3, 128.5, 128.7, 128.9, 129, 129.3, 129.4, 129.5, 129.8, 120.9, 132.2, 133.5, 134.2, 165.6, 170.6 ; HRMS (FAB): m/z calcd for C25H25N2O2 [M + H], 385.1898; found, 385.1917; IR (neat): 3350 cm", 1738 cm' c0211 PhAO/ILNIWN Ph lll-10-1 Benzyl (S)-1-(1-Benzyl-4-methyl 2, S-diphenyl-4,5-dihydro-IH-imidazolyl- 4- carboxylate)-2-methylpropyl carbamate (III-10-l): A solution of benzaldehyde (0.34 g, 3.3 mmol), benzylamine (0.35 g, 3.3 mmol) in anhydrous CH2C12 (15 mL) was refluxed under nitrogen for 2h. 2-Phenyl-4-methyl-4H- oxazolin-S-one (l g, 3.3 mmol) and chlorotrimethylsilane (0.53 g, 4.93 mmol) were added and the mixture was refluxed under nitrogen for 6 h and then stirred overnight at r.t. The solvent was evaporated under vacuum. The product was precipitated using CH2C12/hexanes (1:1); yield: 25% (0.411 g); white foam; 1H NMR (300MHz) DMSO-d;: 8 0.4 (3H, d, J = 6.9 Hz), 0.54 (3H, d, J = 6.9 Hz), 153 1.58 (3H, s), 1.8 (1H, s), 3.94 (1H, m), 3.64 (1H, d, 14.7 Hz), 4.25 (1H, s), 4.59 (1H, d, J =9 Hz), 4.76 (1H, d, J = 15.3 Hz), 4.916 (2H, s), 6.36 (1H, s), 6.96-7.22 (14H, m); 13C NMR (75MHz) DMSO-d6: 8 17.5, 17.9, 18.9, 30.6, 30.9, 43.2, 48.6, 60.5, 66.8, 127.2, 127.3, 127.5, 127.8, 128.0, 128.3, 128.4, 135.8, 137.7, 156.3, 171.1, 171.3, 171.6; HRMS (BI): m/z calcd. for C30H33N3O4 [M +H], 499.6007; found, 5002.; IR (neat): 3302 cm", 1759 cm", 1672 cm". Ill-11-1 9” d1- (45', 53).]-Benzyl-4-methyl-2(4-methoxyphenyl), 5-diphenyl-4,5—dihydro- lH-imidazole- 4-carboxylic Acid (III-ll-l): A solution of benzaldehyde (1.026 g, 9.75 mmol), benzylamine (1.043 g, 9.75 mmol) in anhydrous CH2C12 (15 mL) was refluxed under nitrogen for 2h. 2-(4-methoxyphenyl)-4-methyl-4H-oxazolin- S-one (2 g, 9.75 mmol) and chlorotrimethylsilane (1.58 g, 14.63 mmol) were added and the mixture was refluxed under nitrogen for 6 h and then stirred overnight at r.t. The solvent was evaporated under vacuum. The product was precipitated using CH2C12/hexanes (1:1); yield: 62% (2.42 g); white solid (light sensitive in solution!); 'H NMR (300MHz) DMSO-dgz 8 1.692 (3H, 3), 3.851 (3H, s), 4.11 (1H, d, J = 15.9 Hz), 4.92 (2H, d, J = 15 Hz + s), 6.99-7.02 (2H, m), 7.22 (2H, d, J = 9 Hz), 7.27-7.29 (4H, m), 7.31-7.41 (4H, m), 7.9 (2H, d, J = 8.7 Hz); I3C NMR (75MHz) DMSO—d6: 8 24.9, 48.6, 55.7, 69.7, 72.9, 113.5, 114.8, 127.4, 128.0, 128.2, 128.6, 128.8, 129.4, 131.3, 132.3, 133.1, 163.4, 165.4, 169.3; 154 LRMS (El): m/z calcd. for C25H24N203 [M+], 400.4697; found, 400.6; IR (neat): 3350 cm", 1738cm '1. Ph ) Ph N Ph 111-12-1 F Partial data dl- (4S, 5S)-1-Benzyl-4-isopropyl 2, 5-diphenyl-4,5-dihydro—1H- imidazole- 4-carboxylic Acid (III-12-l): A solution of benzaldehyde (1.58 g, 15 mmol), benzylamine (1.605 g, 15 mmol) in anhydrous CH2C12 (15 mL) was refluxed under nitrogen for 2h. 2-Phenyl-4-methyl-4H-oxazolin-5-one (3 g, 15 mmol) and chlorotrimethylsilane (2.43 g, 22.5 mmol) were added and the mixture was refluxed under nitrogen for 6 h and then stirred overnight at r.t. The solvent was evaporated under vacuum. The product was precipitated using CH2C12/hexanes (1:1); yield: 71% (4.25 g); white solid; 'H NMR (500MHz) DMSO-d6: 6 0.8 (3H, d, J = 7 Hz), 0.93 (3H, d, 7 Hz), 2.35 (1H, q, J = 6.5 Hz), 3.97 (1H, d, J =15 Hz), 4.78 (1H, d, J =15.5 Hz), 5.06 (1H, s), 6.88 (2H, d, J = 7 Hz), 7.265-7.527 (9H, m), 7.75 (1H, t, J =7.5 Hz), 7.847 (1H, t, J =7.5 Hz), 7.95 (2H, d, J = 7 Hz) 13C NMR (125MHz) CD301): 8 16.1, 18.1, 36.4, 42.8, 49.5, 70.5, 78.2, 122.5, 128.9, 129.1, 129.5, 129.54, 129.6, 129.7, 130, 130.2, 130.3, 132.9, 134, 134.6, 134.8, 166.5, 169.6; IR (neat): 3345 cm", l732cm ". 155 Ph ) Ph N Ph Gang—Ecozl'l (Pb Ill-I145 dl- (4S, 5S)-l,3-Dibenzyl-4-methyl-2,5-diphenyl-4,5-dihydro-1H- imidazolinium- 4-carboxylic Acid. Chloride salt (III-l-lS): In a flame dried flask under nitrogen atmosphere, imidazoline III-l-l (0.9 g, 2.42 mmol) was suspended in 200 ml dry benzene. Solid anhydrous K2C03 (4.5 g, 26 mmol) was weighed in followed by addition of benzyl chloride (0.367 g, 2.9 mmol) was added and the mixture was refluxed under nitrogen atmosphere overnight. After all the starting material was consumed, the solvent was removed under vaccum. The residue was dissolved in dichloromethane and the product imidazolium chloride was precipitated from dichloromethane/hexanes (1 :1) mixture in 77 % yield (0.926 g) as white foam; 1H NMR (300MHz) CDC13: 8 1.44 (3H, 8), 3.658 (1H, d, J = 15.6), 4.17 (1H, s), 4.27 (1H, d, J = 12.3 Hz), 4.48 (1H, d, J = 12.3 Hz), 4.56 (1H, d, J = 15.6 Hz), 6.77-6.8 (2H, m), 6.91-6.93 (2H, m), 7.05-7.13 (10H, m) 7.32-7.34 (2H, m), 7.57-7.6 (2H, m); l3C NMR (75MHz) CDC13: 8 26.5, 48.6, 66.0, 72.9, 77.6, 127.4, 127.4, 127.5, 127.7, 127.9, 128.1, 128.4, 129.9, 130.6, 135.3, 136.2, 136.5, 165.9, 171.5; LRMS (EI): m/z calcd. for C31H29N202 [M+], 461.2224; found, 461.2; IR (neat): 3350 cm", 1736 cm ". 156 Ph HN I?“ HN CozH Ph dl—(2S,38)-2,3-bis(benzylamino)—2-methyl-3-phenylpropanoic acid III-1-16: In a round bottom flask equipped with a stirrer III-l-lS (0.5g, 1 mmol) was suspended in 25 mL of MeOH. NaBH4 (0.114 g, 3 mmol) was added in three portions and the mixture was stirred rapidly overnight. After all the starting material was consumed, MeOH was evaporated off. The residue was dissolved in ethyl acetate (50 mL) and extracted with saturated ammonium chloride solution. The organic layer was dried over anhydrous sodium sulfate, filtered and the solvent removed under vaccum. The residue was dissolved in dichloromethane and the product diamino acid was precipitated from ether/dichoromethane mixture a solid foam in 65 % yield (0.24g); 'H NMR (300MHz) (CDCl; + 2 drops D20): 8 1.19 (3H, s), 2.87 (1H, d, J= 12 Hz), 2.98 (1H, d, J= 12 Hz), 3.72 (1H, d, J=15 Hz), 4.10 (1H, s), 4.57 (1H, d, J= 15 Hz), 6.71-7.6 (15H, m); l3C NMR (75MHz) CDC13: 8 26.0, 48.8, 67.2, 71.6, 71.9, 127.7, 127.9, 128.0, 128.6, 128.6, 128.8, 130.3, 130.5, 135.9, 136.0, 165.3; LRMS (E1) m/z calcd. for C24H25N20 [ M]+, 357.1; found, 357.3 ; HRMS (El): m/z calcd. for C24H25N20 [M] +, 357.1966; found, 357.1967. IR (neat): 3182om", 3063 cm", 2924 cm", 2858.9 cm", 1616 cm", 1593.4 cm", 1452 cm °'. 157 9,. @786. 1 lll-1-17 l-Benzyl-4-methyl-2, S-diphenyl-lH-imidazole III-l-l7: A solution of III-l-l (0.5 g, 0.1.35 mmol) in acetic anhydride (15 mL) was heated to reflux for 6h. The acetic anhydride was evaporated to dryness in vacuo. The residue was chromatographed on silica-gel with 3:2 Diethyl ether / Hexanes to afford 0.1 g of III-l-l7 in 23 % yield. Partial data: IH NMR (300 MHz) (CDC13)1 8 2.3 (3H, s), 5.18 (1H, s), 6.7 —6.8 (2H, m), 7.1 —7.65 (13H, m); 13c NMR (75 MHz) (CDC13) 8; 13.0, 48.5, 125.9, 127.3, 127.9, 127.9, 128.4, 128.5, 128.7, 128.8, 129.7, 130.1, 130.2, 130.3, 130.6, 131.9, 135.4, 137.6, 147.4; MS(EI): calculated for C23H20N2 (m/z) 324.4 and observed (m/z) 324. 1‘" <> ,N / \ N lll-1-18 4-(3-Benzyl-5-methyl-Z-phenyl-3H-imidazol-4-yl)—pyridine III-l-l8: A solution of III-l-ll (0.1 g, 0.27 mmol) in acetic anhydride (15 mL) was heated to reflux for 6h. The acetic anhydride was evaporated to dryness in vacuo. The residue was chromatographed on silica-gel with ethyl acetate to afford 0.02 g of III-l-18 in 22 % yield. Partial data: 1H NMR (300 MHz) (CDC13): 8 2.3 (3H, s), . 5.18 (1H, s), 6.7 —6.8 (2H, m), 7.1 -7.65 (12H, m); 13c: NMR (75 MHz) (CDC13): 158 813.5, 29.6, 50.1, 117.6, 126.5, 127.7, 128.4, 128.7, 128.9, 130.3, 137.1, 137.7, 147.4. 159 10. 11. References (a) Li, H. -Y.; Drummond, S.; DeLucca, 1.; Boswell, G. A. Tetrahedron 1996, 52, 11153. (b) Ueno, M.; Imaizumi, K.; Sugita, T.; Takata, 1.; Takeshita, M. Int. J. Immunopharm. 1995, l 7, 597. (c) Rondu, F.; LeBihan, G.; Wang, X.; Lamouri, A.; Touboul, B.; Dive, 6.; Bellahsene, B.; Renard, P.; Guardiola-Lema’l‘tre, B.; Manéchez, D.; Pénicaud, L.; Ktorza, A.; Godfroid, J. -J. J. Med. Chem. 1997, 40, 3793-3803. ((1) Bousquet, P.; Feldman, J. Drugs 1999, 58, 799.(e) Schann, S.; Bruban, V.; Pompermayer, K.; Feldman, J .; Pfeiffer, B.; Renard, P.; Scalbert, B.; Bousquet, P.; Ehrhardt, J. -D. J. Med. Chem. 2001, 44, 1588. (t) Gust, R.; Keilitz, R.; Schmidt, K.; von Ranch, M. J. Med. Chem. 2002, 45, 3356. Acharya, A. N.; Ostresh, J. M; Houghten, R. A. J. Comb. Chem. 2001, 3, 612. Concellon, J. M.; Riego, B.; Suarez, J. R.; Garcia-Granda, S.; Rosario Diaz, M. Org. Lett. 2004, 6, 4499 Acharya, A. N.; Ostresh, J. M; Houghten, R. A. J. Org. Chem. 2001, 66, 8673. Chem, J. -W.; Liaw, Y. -C.; Chen, C. -S.; Rong, J. -G.; Huang, C. -L.; Chan, C. -H.; Wang, A. H. -J. Heterocycles 1993, 36, 1091-1103. De Lucca, G. V. J. Org. Chem. 1998, 63, 4755-4766. (a) Bon, R. S.; Hong, C.; Bouma, M. J.; Schmitz, R. F.; de Kanter, F. J. J.; Lutz, M.; Spek, A. L.; Orru, R. V. A. Org. Lett. 2003, 5 (20), 3759 -3762. (b) Gentili, F.; Bousquet, P.; Brasili, L.; Dontenwill, M.; Feldman, J.; Ghelfi, F.; Gianella, M.; Piergentili, A.; Quaglia, W.; Pigini, M. J. Med Chem. 2003, 46, 2169-2176. (a) Dunn, P. J.; Haner, R.; Rapoport, H. J. Org. Chem. 1990, 55, 5017- 5025. (b) Han, H.; Yoon, J.; Janda, K. D. .1. Or . Chem. 1998, 63, 2045- 2048. Femandez-Megia, B.; Sardina, F. J. Tetrahedron Lett. 1997, 38(4), 673. You, S.-L.; Kelly, J. W. Org. Lett. 2004, 6(10), 1681 -1683. Munk, S. A.; Harcourt, D.; Ambrus, G.; Denys, L.; Gluchowski, C.; Burke, J. A.; Kharlamb, A. B.; Manplapaz, C. A.; Padillo, E. U.; Runde, B.; Williams, L.; Wheeler, L. A.; Garst, M. E. J. Med. Chem. 1996, 39, 3533. 160 12. 13. 14. 15. 16. 17. 18. 19. 20. 21. 22. 23. 24. 25. 26. Regunathan, S., Feinstein, D. L., Reis, D. J. Ann. N. Y. Acad Sci. 1999 881, 410-419. Dardonville, C.; Rozas, 1. Med. Res. Rev. 2004, 5, 639-661. (a) Schfifer, U.; Burgdorf, C.; Engelhardt, A.; Kurz, T.; Richardt, G. J. Pharmacol. Exp. T her. 2002, 303, 1163-1170. (b) Gust, R.; Keilitz, R.; Schmidt, K. J. Med Chem. 2001, 44, 1963-1970. (a) Gilbert, 1. H.; Rees, D. C.; Crockett, A. K.; Jones, R. C. F. Tetrahedron 1995, 51, 6315. (b) Jones, R. C. F.; Dickson, J. J. Peptide Sci. 2000, 6, 621-624. Minter, A. R.; Brennan, B. B.; Mapp, A. K. J. Am. Chem. Soc. 2004, 126(34), 10504 -10505. Arkin, M. R.; Wells, J. A. Nature Rev. Drug Discov. 2004, 3(4), 301-317. Kussie, P. H.; Gorina, S.; Marechal, V.; Elenbaas, B.; Moreau, J .; Levine, A. J .; Pavletich, N. P. Science, 1996, 274, 948-953. Vassilev, L. T.; Vu, B. T.; Graves, B.; Carvajal, D.; Podlaski, F.; Filipovic, Z.; Kong, N.; Kammlott, U.; Lukacs, C.; Klein, C.; Fotouhi, N.; Lin B. A. Science, 2004, 303 , 844-848. (a) Dai, L.; Lin, Y.; Hou, X.; Zhou, Y. Pure. Appl. Chem. 1999, 71(6), 1033. (b) Lucet, D.; Le Gall, T.; Mioskowski, C. Angew. Chem. Int. Ed. 1998, 37, 2580. (c) Zhou, X.; Lin, Y.; Dai, L.; Sun, J.; Xia, L.; Tang, M. J. Org. Chem. 1999, 64, 1331. Hsiao, Y.; Hegedus, L. S. J. Org. Chem. 1997, 62, 3586. (a) Dalko, P. 1.; Langlois, Y. J. Org. Chem. 1998, 63, 8107 (b) Austin J. F .; MacMillan, D. W. C. J. Am. Chem. Soc. 2002, 124(7), 1172. (a) Jones, R. C. F.; Howard, K. J .; Snaith, J. S. Tetrahedron Lett. 1996, 37, 1707-1710. (b) Jones, R. C. F.; Howard, K. J.; Snaith, J. S. Tetrahedron Lett. 1996, 3 7, 1711-1714. (c) Dalko, P. 1.; Langlois, Y. Chem. Commun. 1998, 331-332. Menges, F.; Neuburger, M.; Pfaltz, A. Org. Lett. 2002, 4, 4713-4716. Herrmann, W. A. Angew. Chem, Int. Ed. 2002, 41 , 1290-1309. Hermann, W. A.; Goossen, L. J .; Speigler, M. Organometallics. 1998, 17, 1172. 161 27. 28. 29. 30. 31. 32. 33. 34. 35. 36. 37. 38. 39. 40. (a) Botteghi, C.; Schionato, A.; Chelucci, G.; Brunner, H.; Ktlrzinger, A.; Obermann, U. J. Organomet. Chem. 1989, 370, 17-31. (b) Betschart, C.; Hegedus, L. S. J. Am. Chem. Soc. 1992, 114, 5010-5017. (c) Jones, R. C. F.; Turner, 1.; Howard, K. J. Tetrahedron Lett. 1993, 34, 6329-6332.(d) Boland, N. A.; Casey, M.; Hynes, S. J.; Matthews, J. W.; Smyth, M. P. J. Org. Chem. 2002, 67, 3919-3922. (a) Botteghi, C.; Schionato, A.; Chelucci, G.; Brunner, H.; Ku”rzinger, A.; Obermann, U. J. Organomet. Chem. 1989, 370, 17-31. (b) Dauwe, C.; Buddrus, J. Synthesis 1995, 171-172. (c) Dalko, P. 1.; Langlois, Y. J. Org. Chem. 1998, 63, 8107-8117. ((1) Davenport, A. J .; Davies, D. L.; Fawcett, J.; Rusell, D. R. J. Chem. Soc., Perkin Trans. 2001, 1500-1503. (e) Dupont, J.; Ebeling, G.; Delgado, M. R.; Consorti, C. S.; Burrow, R.; Farrar, D. H.; Lough, A. J. Inorg. Chem. Commun 2001, 4, 471-474. (1) Oi, R.; Sharpless, K. B. Tetrahedron Lett. 1991, 32, 999-1002 (a) Legters, J.; Willems, J. G. H.; Thijs, L.; Zwanenburg, B. Rec. TraV. Chim. Pays-Bas 1992, 111, 59-68. Hershenson, F. M.; Pavia, M. R. Synthesis 1988, 999-1001. Consonni, R.; Croce, P. D.; Ferraccioli, R.; La Rosa, C. J. Chem. Res, Synop. 1991, 188-189. Bilodeau, M. T.; Cunningham, A. M. J. Org. Chem. 1998, 63, 2800-2801. Gotthardt, H.; Huisgen, R.; Bayer, H. O. J. Am. Chem. Soc. 1970, 92, 4340. Croce, P. D.; Ferraccioli, R.; La Rosa, C. Tetrahedron 1999, 55, 201. Mukerjee, A. K. Heterocycles 1987, 26, 1077. Avalos, M.; Babiano, R.; Cintas, P.; Hursthouse, M. B.; Jimenez, J. L.; Light, M. B.; Lopez, 1.; Palacios, J. C.; Silvero, G. Chemistry 2001, 7, 3033. Prasad, K. K.; Petrzilka, T. Helv. Chim. Acta 1975, 58, 2504. Chen, F. M. F.; Kuroda, K.; Benoiton, N. L. Synthesis 1979, 230. Palomo, C.; Aizpurua, J. M.; Ganboa, 1.; Oiarbide, M. Eur. J. Org. Chem. 1999, 309, 3223. Sain, B.; Sandhu, J. S. Heterocycles. 1985, 23, 1611. 162 41. 42. 43. 45. 46. 47. 48. 49. 50. 51. 52. 53. 54. 55. 56. Cossio, F. P.; Ugalde, J. M.; Lopez, X.; Lecea, B.; Palomo, C. J. Am. Chem. Soc. 1993, 115, 995. Dghaym, R. D.; Dhawan, R.; Arndtsen, B. A. Angew. Chem. Int. Ed. 2001, 40, 3228. Bowman, R. K.; Johnson, J. S. J. Org. Chem. 2004, 69(24), 8537-8540. Roberts, R. M.; Higgins, T. D.; Noyes, P. R. J. Am. Chem. Soc. 1955, 77, 3801-3805. Takagaki, H.; Yasuda, N.; Asaoka, M.; Take, H. Chem. Lett. 1979, 183. Hayashi, T.; Kishi, E.; Soloshonok, V. A.; Uozumi, Y. Tetrahedron Lett. 1996, 37(28), 4969-4972. Zhou, X. -T.; Lin, Y. -R.; Dai, L. -X. Tetrahedron Asymm. 1999, 10, 855- 862. Viso, A.; Fernandez De La Pradilla, R.; Guerrero-Strachan, C.; Alonso, M.; Martinez-Ripoll, M.; Andre, I. I. J. Org. Chem. 1997, 62, 2316-2317. Viso, A.; Fernandez De La Pradilla, R.; Garcia, A.; Guerrero-Strachan, C.; Alonso, M.; Tortosa, M.; Flores, A.; Martinez-Ripoll, M.; Fonseca, 1.; Andre, 1.; Rodriguez, A. Chem. Eur. J. 2003, 9, 2867-2876. Wilde, R. G. Tetrahedron Lett. 1988, 29(17), 2027-2030. Okada, H.; Akaki, T.; Oderaotoshi, Y.; Minakata, S.; Komatsu, M. Tetrahedron 2003, 59, 197. Komatsu, M.; Okada, H.; Yokoi, S.; Minakata, S. Tetrahedron Lett. 2003, 44, 1603. Komatsu, M.; Okada, H.; Akaki, T.; Oderaotoshi, Y.; Minakata, S. Org. Lett. 2002, 4, 3505. Peddibhotla, S.; Jayakurnar, S.; Tepe, J. J. Org. Lett. 2002, 4, 3533-3535. Peddibhotla, S.; Tepe, J. J. Synthesis 2003, 9, 1433-1440. Oishi, Masataka. Eds.: Yamamoto, Hisashi. Lewis Acids in Organic Synthesis (2000), 1 355-393. Publisher: Wiley-VCH Verlag GmbH, Weinheim, Germany. 163 57. 58. 59. 60. 61. 62. 63. 65. 66. 67. 68. 69. 70. Shanna, V.; Tepe, J. J. Enhancing efficacy of camptothecin using chiral imidazolines selectively in cancer cells. J. Am. Chem. Soc. 2004 (in preparation) Zhou, P.; Chen B. -C.; Davis F. A. Tetrahedron 2004, 60, 8003-8030. Juaristi, E.; Leon-Romo, J. L.; Reyes, A.; Escalante, J. Tetrahedron Asymm. 1999, 10, 2441—2495. Nefzi, A.; Ostresh, J. M.; Yu, J.; Houghten R. A. J. Org. Chem. 2004, 69(11), 3603 -3609. (a) Jung, S. -H.; Kohn, H. J. Am. Chem. Soc. 1985, 107, 2931. (b) Jung, S- H.; Kohn, H. Tetrahedron Lett. 1985, 25(4), 399. Johnson, R. N.; Woodbum, H. M. J. Org. Chem. 1962, 40, 3958. Chambers, J. J.; Kurrasch-Orbaugh, D. M.; Parker, M. A.; Nichols, D. E. J. Med. Chem. 2001, 44, 1010. Johnson, M. A.; Sousa, L. R. Synthesis. 1978, 43, 317. Nowick, J. S.; Holmes, D. L.; Noronha, G.; Smith, E. M.; Nguyen, T. M.; Huang, S. -L. J. Org. Chem. 1996, 61 , 3929. Perrin C. L.; Young. D. B. J. Am. Chem. Soc. 2001, 12309), 4446-4450. Xia, C.; Hao, J.; Tang, Y.; Ni, Y.; Zhou, P. Syn. Commun. 2002, 32(9), 1457-1464. Chang, L. A.; Sidler, K. L.; Cascieri, M. A.; de Laszlo, S.; Koch, G.; Li, B.; MacCoss, M.; Mantlo, N.; O’keefe, A.; Pang, M.; Rolando, A.; Hagmann, W. K. Bioorg. Med Chem. Lett. 2001, 11, 2549. Slee, D. H.; Romano, S. J.; Yu, J.; Nguyen, T. U.; John, J. K.; Raheja, N. K.; Axe, F. U.; Jones, T. K.; Ripka, W. C. J. Med. Chem. 2001, 44, 2094. Tasdemir, D.; Mallon, R.; Greenstein, M.; Feldberg, L. R.; Kim, S. C.; Collins, K.; Wojciechowicz, D.; Mangalindan, G. C.; Concepcion, G. P.; Harper, M. K.; Ireland, C. M. J. Med. Chem. 2002, 45, 529. 164 CHAPTER IV SILVER ACETATE CATALYZED Exo-SELECTIVE SYNTHESIS OF A'- PYRROLINES AND DERIVED PYRROLIDIN ES A. Biological and chemical significance of A'pyrrolines A'-pyrrolines can be found in nature as biosynthetic intermediates, and as a part of pheromones, and alkaloids, steroids, hemes and chlorophylls (Figure IV-l)." 2 Myosmine, amathaspiramide E, dysinosin A, broussonetine U1, solamalidine, veracintine and lanopylins are some recently isolated biologically active alkaloids which contain a pyrroline unit.3 Moreover, these scaffolds have been shown to possess insecticidal, fungicidal, anti-viral, anti-tumor and immunoactivity. 3°’ 4 Cl C02” H020 M NH; 5,, 00,11 Amathaspiramide E Kaitooephalin Y 0% ’2, 90H 0 H o 0 HO Spirotryostatin A Lactacystin Figure IV-l. Biologically active pyrrolines and pyrrolidines In addition to wide ranging biological activity, A'-pyrrolines are important synthetic intermediates, as they have three contiguous stereocenters, and one 165 prochiral center as part of a cyclic imine, which is amenable to further synthetic manipulation such as addition of nucleophiles with stereocontrol." 5 Hence, they are of immense value for the synthesis of pyrrolidines and complex natural products containing highly functionalized five-membered N containing rings.“ 7 Nitrones generated from A'-pyrrolines have been employed in [3+2] cycloadditions to generate polycyclic heterocycles like pyrrolizidines, indolizidines and aza sugars.8 Synthesis of 01,01-disubstituted amino acids (especially prolines) and their analogs is a topic of intense synthetic interest because of their application in the field of peptidomimetics, catalysis and synthesis of natural products.9 A stereoselective and efficient preparation of a general 131-pyrroline template would provide rapid access to a range of biologically active natural products including the myosmines, amathaspirarnides, lactacystin and kaitocephalin type alkaloids.lo B. Access to A'-pyrrolines using mfinchnone-alkene cycloaddition Although, there are some reports11 for the synthesis of AI-pyrrolines cycloaddition of milnchnones to alkenes has not been fruitful as a synthetic method to access these compounds.”'3 Extensive work by Huisgen and others has established 1,3- dipolar cycloaddition reactions of N-alkylated mtlnchnones (Figure IV-2), as a general route for the syntheses of pyrroles, Al-pyrrolines, azepines and imidazoles." Synthesis of Al-pyrrolines from intermolecular 1, 3- dipolar cycloaddition of mtlnchnones to alkenes is fraught with following difficulties. '5 Under the conditions generally used (reflux in acetic anhydride) the reaction 166 cannot be controlled at the primary adduct stage as the decarboxylation of bicyclic intermediates is facile, leading to cyclic azomethine ylide/Al-pyrroline.“5 R o 9 "_ 3 E R4 R‘ R3 9 / —* \ IN 0 R1 N R2 R2 0 R2 R 4:0/ R4 R3 R4 R3 R1 Ng R2 R1 Ne R2 l 1 R4 R R R 3 l j____/R3 l R1/Z/;j(COZI-l R4 R 1 R2 R4 R3 R N R4 R A 184% AZ-pyrroline-Z-earboxylate R‘ N R2 R3 is R3 R2 PYrrole 7-azabicyclo [2.2.1] heptane Figure IV-2. Dipolar cycloaddition reactions of N-alkylated mfinchnones This cyclic azomethine ylide then undergoes the following transformations: 1‘ 1. Isomerization of double bond leads to formation of Az-pyrrolines and partial loss of stereochemistry generated during cycloadditions. 2. The position of double bond in AZ-pyrrolines is determined by the C3 and C4 substituents and often results in mixture if unsymmetrically substituted. This results in complex inseparable mixtures. 3. Aromatization to pyrroles and complete loss of stereochemical information. 4. A second cycloaddition with excess alkene to afford 7-azabicyclo [2.2.1] heptarle derivative. 167 5. a-olefins (monosubstituted alkenes) have been shown not to participate in the cycloaddition. 6. Even, if the decarboxylation is prevented when reactions are run at lower temperatures, isomerization to Az-pyrrolines is inevitable.'7 / R1 A A020 R1 @N O I‘BflUX N/ O 1329 0 R2 0 l R2<9 0 R2 R1 0 @N A020 k 0 reflux N 0 R1 Figure IV-3. Primary cycloadducts isolated from intramolecular cycloadditions However, Padwa and co-workers have been successful in isolating and characterizing primary adducts of intramolecular cycloadditions of milnchnones to terminal alkenes (Figure IV-3).18 Steric constraints (violation of Bredt’s rule) on the transition state prevent the elimination of carbon dioxide from these cycloadducts and also result in the observed stereochemistry. o 0 CI 0 _<9 _A°1_, reflux H Figure IV-4. Primary cycloadducts isolated from intermolecular cycloadditions 168 Turchi and co-workers described the isolation of a Al-pyrroline-S-carboxylic acid from the intermolecular cycloaddition of 1, 2-dicyanocyclobutene to a munchnone in refluxing acetic anhydride (Figure IV-4).l5 Decarboxylation and hydride migration are presumably prevented by structural constraints of the bicyclic product. The authors were able to decarboxylate the intermediate by thermolysis in refluxing decalin. Exclusive exo selectivity was observed in the primary cycloadduct. c. TMSCI (Lewis acid) mediated mild protocol to Al-pyrroline-S- carboxylates Re INVR3 N R3 R4 \ : R1—'<\ ICOzH O O / imine N '3 0 0 R2 TMSCI R1—<\ I wa—h R,-<\ :E, 2—imidazoline—4-earboxylate N R . N R4 2 I R2 LA \ KWR" ‘ ‘dRa azlactone munchnone amene R1 \N -. COZH D‘-pyrroline-5:28rboxylate Scheme IV-l. Lewis acid mediated mild protocol to Al-pyrroline-S-carboxylates We had reported that coordination of a trimethylsilyl chloride to nitrogen atom of the azlactone, can increase the equilibrium concentration of munchnone intermediate and accelerate the cycloaddition reaction with imines (Scheme III- 7).'9 Moreover, Lewis acid mediation leads to milder protocol (reflux in dichloromethane compared to acetic anhydride) and it was possible to isolate the 2-imidazoline-4-carboxy1ic acid without any decarboxylation to imidazole and in 169 excellent yields. [3+2] cycloadditions in which heterodipolarophiles are employed to intercept reactive azomethine ylides represent a useful entry into functionalized, protected amino acids.20 In case of mfinchnones, dihydroheterocycles isolated are masked 01, a-disubstituted (quarternary) amino acids. In case of munchnone-imine cycloadditions we obtained quarternary imidazoline-4-carboxylic acids. These scaffolds were formed with high diastereoselctivity. The scaffolds exhibit four-point diversity, are amenable to alkyl, aryl, acyl and heteroalkyl substitutions and display novel, interesting and important biological activity.” 2' Buoyed by our own results and the isolation of Al-pyrroline-S-carboxylic acid by Maryanoff and co-workers, we decided to apply the mild TMSCl mediated condition for generation of mttnchnones to cycloadditions with alkenes.ls Azlactones were synthesized by cyclodehydration of N-acyl-a-amino acids with EDCI.HC1.22 2-Phenyl, 4-methyl azlactone (II-1) was refluxed in THF along with maleic anhydride in presence of 1.5 equivalents of TMSCl overnight. The residue afier acid-base workup and extraction into ethyl acetate afforded crude product, which was shown to have pyrroline in substantial amount. We were gratified that the reaction did indeed occur, but unfortunately the product A'-pyrroline could not be isolated pure (data not shown). Diethyl fumarate, after same extraction procedure resulted in a complex mixture of diastereomers of Al-pyrroline-Z- carboxylic acids and N-benzoylalanine (probably from unreacted azlactone). The mixture of acids was methylated with diazomethane. Column chromatography of the mixture resulted in 50 % combined yield of Al-pyrrolines in a 2.5:] 170 diastereomeric ratio (Table IV-l, entry IV-l-2). Reaction of 2,4-dimethyl azlactone with t-butyl acrylate resulted in only 5 % isolated yield of diastereomer of Al-pyrroline after methylation and column. Most of the N-benzoylalanine was recovered from unreacted azlactone (Table IV-l , entry IV-l-3). Methyl acrylate only resulted in 25 % yield of A'-pyrroline-2-carboxylic acid, which was directly precipitated from the crude product using a mixture of ether and methanol. It is evident that the conversion of azlactone to product is not complete in THF for the reaction times and temperature, which were used. Moreover alpha olefms, which are reported not to react with mtlnchnones, seem to be poor dipolarophiles. It also occurred to us that TMSCI might not be the ideal Lewis acid for this reaction. 171 alkene R. o 9 Rf‘V'“ AR; R1_‘<\N -—> R1 \N . COzH R2 TMSCI (1.3 eq.) R THF, reflux 2 azlactone Dl-pyrroline-S-carboxylate entry alkene 96 yield product Ph Rh 0 N' N 0 lV-1-1 0 0 - Ph COzEt lV-1-2 _/ .nCOzEt E1020 MCOzMe t-BUOzC IV-1-3 t-Buozc a % \= P" \ .nC02Me N MeOzc IV-1-4 MOOZC 27% E Ph N .nCOZMe Table IV-l. TMSCl mediated synthesis of A'-pyrroline. Several manipulations of reaction conditions with respect to solvent, time of reaction and stoichiometry of alkene used were tried, without any success (data not shown). This led us to search for a different Lewis acid. Related to our work, Komatsu and co-workers had described in situ generation of N-silylated azomethine ylides carrying an alkoxy substituent by treatment of a-silylimidates with trifluorophenyl silane.23 The resulting azomethine ylide is a nitrile ylide equivalent as it eliminates the alkoxy group after cycloaddition to alkenes, to afford A'-pyrrolines in good yields (Figure lV-S). Although the configuration of 172 starting alkene (cis- or trans-) is retained in the cycloadditions, no diastereoselctivity was observed. This is presumably because of the interconversion of syn and anti forms of the dipole as is well known for acyclic 7n systems. fi’ho N 0v 0 OR DCM rt 48" OPhH; TMS OR - PhSIF e _2, AQA (1: 1 endo: exo) R1 NARZ -TMSF R1 0' R2 cozue nitrile ylide equivalent M9020 > 2 ,- 2M9 DCM, rt, 48h Ph N’ Ph (1 :1 endozexo) Figure IV-S. Azomethine generation by N-silylation. D. Screening of additional Lewis acid for synthesis of 151-pyrrolines Afier screening a variety of Lewis acids we found that silver (I) acetate successfully catalyzed the cycloaddition reaction of 2-phenyl-4-methyl azlactone with N-phenyl maleimide generating the bicyclic Al-pyrroline in excellent yield without isomerization to the Az-pyrrolines or decarboxylation (Table IV-2).8‘ '3 The Al-pyrroline was isolated as methyl ester by in situ methylation with excess trimethylsilyl diazomethane. It was surprising to note that neither TMSCl nor TMSOAc resulted in any cycloadditions product. Both Cu (1) and Cu (11) both did catalyze the cycloadditions but Cu (I) definitely resulted in cleaner reaction and higher yields. The role of the counter anion seems to be important to favor equilibration to the N-metallated munchnone. Komatsu had concluded that the 173 exact number of fluorine ligands were important in their work with N-silylated azomethine ylides (Figure IV-S).23 I'Dh ‘81:)” Ph 0 o 1)LA(10mol%) 085%,: o THF, rt, dark ' -‘ P"_<‘~ = tau—<1, ': COzMe 2) TMSCHNZ N a 11.1 was entry LA 96 yleld 1 AgOAc‘ 78 2 CuOAc 5O 3 Cu(OAc)2 30 4 TMSCI' 0 5 M9 Br zEtzO 0 6 mom; 0 7 ch12 o a LiCl 0 $ No significant pyrrole fomation was detected; ‘TMSCI was used in 1.3 eq.; # Yb(OTf)3 resulted in polymerization Table IV-2. Lewis acid screen for synthesis of A'pyrrolines. Grigg et al have recently reported24 highly efficient synthesis of Azpyrrolines from glycine derived isocyanate esters and alkenes using silver acetate as Lewis acid. The use of acetate has been suggested to arise from specific need to have a mild base like acetate ion to deprotonate a-position of the isocyanate ester to generate a highly reactive azomethine ylide for the 1, 3-dipolar cycloaddition. The products isolated, AZ-pyrrolines were obtained by in situ hydride migration of A'- pyrrolines. Hydride migration was either blocked only by a-substitution of the isocyanoacetate or by constraining the B-position to the iminic N in the A'- 174 pyrrolines. It is important to note that a mixture of diastereomers was seen, again highlighting the difficulty of achieving complete diastereocontrol because of equilibration between syn and anti-dipole geometry." E. Diversity in AgOAc catalyzed synthesis of Al-pyrroline-S-carboxylates The cycloaddition reactions proceed well with electron deficient alkenes and 10 mol % silver acetate in THF at room temperature to provide the highly substituted Al-pyrrolines, ofien in good yields. Only the exo adducts of the A'-pyrrolines were observed with cis-olefins as determined by NOE experiments and X-ray crystallography (Figure IV-6 and Table IV-3). 175 1) AgOAc (10 mol %) R4 0 ° THF rt dark '- R _<\N alkene : Ph—G— \ R 2) TMSCHN2 N a 002R azlactone 1 % Y'O'd A -pyrrollne E ntry R alkene (azb) a 1) F11 Ph 0V“ ' >=o — P“ \N , cozm Me Iv-1-5 EtOzc EtO co Et @0023 2 Me 2 2 75 C\=/ Ph \N '. cone Me IV-1-6 Etozc BO 0 co Et doom 3 Ph 2 2 15 E’ P" \N ._ cognac 13h lV-2-6 61020 5102 doom 002151 4 Me i; 75 (2:1) ph 1:1—(j: co 25, econe econe (v.1 .7; IV-1 -7b M9020 COzMe 5 Me M$020 953121th— ”141 \= ( ) eCOZMe N :on" lV-1-8a IV-1-8b Table IV-3. Cycloaddition of various alkenes 176 ”a? f?" 01221 / ‘2’) (1 "1 11 J .1112; f ‘23) (N, 0491.1, a R 01121 x 9 C(45)? ., - ‘3‘” . l: . . " .C14l 8"“ ... 80121 c? . 61411 f ‘ _ / 83 . \' :-'- ‘\ I \ ’ - V 7 C111) crux-t . 9 N C113) 87’ O 2‘, r - LI'IHIf J42. 01111 Figure IV- 6. X-ray crystal structure of IV-l-6. Alkenes (R3 and R4 substituents). Electron deficient cis-alkenes (N-phenylmaleimide and diethyl maleate) as well as trans alkene (diethyl fumarate) resulted in very high yields for Al-pyrrolines (entries 1, 2 and 4, Table IV-3). However, trans-diethyl firmarate resulted in 2:1 ratio of exo/endo-diastereomers (entry 4, Table IV-3). Diethylfumarate and diethylmaleate provided both a 3, 4-trans relationship of the ethoxycarbonyl groups. This is likely the result of the isomerization (Scheme IV-2) of the cis- substituted A'-pyrroline to the thermodynamically more stable trans-product (entries 2 versus 4, Table 1V-3). This is in accordance with previous observation by other researchers. '4 177 e — r o EtOZQ EtO Etc 3: AmCCbEl £00251 BMe ECChEt _.._.‘ = P" \N a cozue ‘ P" \N , C02Me P“ ‘N , 002Me Me _ Me _ Me lv-t-e Scheme IV-2. Isomerization of cis Al-pyrroline to trans Al-pyrroline. A main highlight is that, monosubstituted alkenes (methyl acryalate) which were previously not known to participate in intermolecular cycloadditions of mllnchnones also afforded very high yield for the A1-pyrroline products.18 The lack of regioselectivity is consistent with reported literature on intermolecular cycloadditions of mfinchnones with monosubstituted dipolarophiles.26 Efforts to introduce alkyl or aryl substituents in 4-position with crotonic or cinnamic esters were not successful (entries IV-2-5 and IV-l3-5, Table IV-4). Electron rich alkenes such as vinyl trifluoroacetate (entry IV-3-5, Table IV -4) were also unreactive in the cycloaddition with mfinchnones in accordance with the 27 Sustrnann classification for these dipoles. 178 O 1) AgOAc (10 mol %) §=0 R _<\ THF, rt, dark 7‘ 1 N * Rl‘acozm R 2) TMSCHN; N '— 2 R 2 azlactone en 96 leld try R1 R2 y lV-2-5 ph pr; 0 lV-13-5 Ph Bn 67 lV-3-5 Ph 3-indolylmethyl 7O lV-5-5 Me Me 59 lV-9-5 Bn Me 75 Table IV-4. Variation of substituents at C2 and C5 in Al-pyrrolines. Amino Acids and acid chlorides (R; and R; substituents). Very good yields were obtained with benzyl and indolylrnethyl substituted azlactones (entries IV-13-5 and IV-3-5, Table IV-4). The phenyl glycine derived azlactone reacted with diethyl maleate to afford the Al-pyrroline in a modest 15% yield (entry IV-2-5, Table IV-4) and was unreactive towards maleimide. The 2- position on Al-pyrroline scaffold is also amenable to difl‘erent substituents including the 2-methy1 and 2-benzyl (entries IV-5-5, and IV-5-5, Table IV-4). F. Probable origin of diastereoselectivity Only the exo adducts of the Al-pyrrolines were observed as major products determined by NOE experiments and X-ray crystallography (Figure IV-6). The exo-preference is in contrast to the endo-preference observed in the synthesis of . . . . . 2829-30 . . . pyrrolldmes from acycllc azomethlne ylldes. A notable exceptlon to thls 15 31 the ligand induced exo-selectivity described by Komatsu. The exo-selectivity 179 observed by Padwa was reported to reflect the influence of steric factors in a o o o o o I 32 unimolecular transrtton state of an intramolecular cycloaddltlon. (I) (b) (c) acyclic azomethine ylide Munchnone Munchnone syn-dipole anti-dipole anti-dipole R R L/ “ R L’Ag"" “ L’Ag"‘ \ \ Ph-l’ g / [3th :00 L pthl ‘.q~l Ph-“ “0 L F)h‘N” i '/ O N /‘ , :/ H N- H H endol lexo endo %( Ph Ph Ph I l I O N O OVNVO O N O 0MB 2. _: H'“ .uH H H OH H'“ .nH OH Ph 2 Ph ‘ -. \ ‘ a \ u R O N R 0 Ph N R 0 Scheme IV-3. (a) proposed syn-dipole orientation for acyclic azomethine ylides derived from imines of ct-amino acid esters; (b) munchnone (cyclic azomethine ylide) locked as an anti-dipole. Orientation of alkene is same and differing outcome from (a) and (b) could be a result of different dipole geometry. Acyclic azomethine ylides derived from a-iminoesters have been proposed to adopt a preferred syn orientation in presence of Lewis acids due to chelation with the carboxyl oxygen. 28‘ 29‘ 30 With silver (I) Lewis acids the inter-conversion to the anti-orientation is minimum and hence the cycloadditions are highly endo selective.7n The milnchnones are azomethine ylides locked in anti-orientation. This could be a possible basis for obtaining the opposite exo diastereoselectivity in the resulting product for the same orientation of alkene (Scheme IV-3). 180 Figure IV-7. Calculated electrostatic isopotential surfaces for exo- and endo- approach of dicyanocyclobutene Turchi and co-workers have carried out extensive MO calculations to rationalize the stereochemical outcome.33 AMI calculations for additions of 1, 2- dicyanocyclobutene to miinchnones indicated that the cycloaddition is a concerted but nonsynchronous process. Semi-empirical calculations (FMO) have not been successful in explaining regiochemical and stereochemical outcome of cycloadditions of milnchnones. Electronic effects were suggested to play a dominant role in favouring the exo-product in intermolecular cycloaddition reaction.33 Inspection of electrostatic isopotential surfaces revealed that the exo transition state shows attractive stabilizing interaction between the CH-NH-CH portion of dipole and the nitrile groups. In fact, it was found that the exo-adduct is also electrostatically favoured over the endo-adduct. This may provide a rationale for the formation of the exo-cycloadduct with this in situ generated munchnone- alkene cycloaddition reaction. endo transition state for reaction with munchnone is proposed to be disfavourcd due to repulsive destabilizing interactions between 181 O-C=O region of munchnone and the carbonyls of N-phenyl maleimide (anti- orientation of carbonyl dipoles expected to be more stable).25 G. Expanding the diversity of A'pyrroline templates25 During isolation the bicyclic cis Al-pyrroline-S-carboxylate by methylation using trimethyl diazomethane in protic solvents like methanol, we observed isomerization and equilibration to the thermodynamically more stable ring opened trans product. Of the various bases screened to convert IV-l-S to IV—1—9, DMAP resulted in clean conversion and afforded a 50:50 mixture of IV-l-S and IV-1-9 in quantitative yields. Treatment of the isolated 3, 4-trans A'- pyrroline methyl ester (IV-l-9) with DMAP in methanol reestablished the 1:1 ratio of IV -1-5 and IV-l- 9, indicating that the interconversion of IV-l-S and IV-1-9 most likely proceeds via a common ring opened ketene intermediate (Scheme IV- 4). 0 Ph ’ - Ph N u , ' V }=O DMAP c HN Ph ”“9020 ”1 Ph \ ‘ co Me MeOH ph J§o MN“ Ph \ ‘ ° N 2 .222. h com ~ ...... u . M8 lV-1-5 Me ‘ lV-1-9 o F" .Ph _ _ M6020 HN “ Ph YNEO Ago DMAP HN ° 6 Ph \ -—'> wk "" PTO—002m + ”'1'9 N E. COzMe MBOH Ph \ O N E. Me $3. N a 002m M9 Iv-1-s ‘ Me ‘ lV-1-5 Scheme IV -4. Ring opening and isomerization of cycloadducts. We were successful in selectively trapping the ketene intermediate with benzylamine to afford the trans amide in good yield (entry IV-l-10, Scheme IV- 182 5). This selective ring-opening reaction provides an opportunity to expand on the stereochemical diversity of the scaffolds. Ph :5 ° 0 0% \th Nucleophile R PNHPh ' ' DMAP ' PhQCOZ’Me THF > Ph \ 'v 002R N Me N Me Entry Nucleophile R % Yield Iv-1-s MeOH OMe 50 % IV-1-10 NHan NHBn 76% Scheme IV-S. Ring opening with different nucleophiles Thus, we have developed a highly diastereoselective and efficient synthesis of Al- pyrroline-S-carboxylic acid scaffolds via a Ag-(I) catalyzed generation of mtlnchnones and subsequent [3+2] cycloaddition to alkenes. The exo-selectivity complements the endo-selective cycloaddition of related acyclic azomethine ylides very well. Diastereocontrol can be extended to the three stereocenters by appropriate choice of substrates and isolation conditions. H. Synthesis of pyrrolidines and DOS As a continuing theme of developing new diversity oriented synthetic methods for small molecule scaffolds with potential biological activity, we focused on developing stereoselective synthesis for novel A'-pyrrolines and pyrrolidines derived from them. Stereochemical diversity is desirable as it increases the number of relative orientations of potential macromolecule-interacting elements in small molecules. It can best be achieved by using stereospecific reactions that proceed with enantio- or diastereoselectivity. 183 The dipolar cycloadditions of an azomethine ylide to an alkene is a very useful synthetic method for the synthesis of pyrrolidines. Two carbon-carbon bonds are formed in a single operation, usually with a a high degree of regioselectivity, while upto four new stereocenters are created, often in a highly stereoselective manner. Therefore, this method is of considerable interest in DOS because its stereospecificity enables stereochemical diversification of up to four tetrahedral 33 Achieving diastereoselectivity and centers on pyrrolidine rings.” enantionselectivity in this reaction has hence attracted intense scientific investigation. I. Importance of diastereoselective diversity-generating pyrrolidine syntheses 3‘ involve the transformation of a collection Since diversity-generating processes of substrates into a collection of products, it is critical that the processes used to generate new stereogenic centers be both selective and general. Greater stereochemical diversity requires powerful reagents that can override substrate bias and deliver diastereomeric products with very high selectivity. Double- diastereoselective reagents that can override the face selectivity of both coupling partners, for example, to achieve exo versus endo selectivity are highly desirable and the discovery of these types of powerful reagents is critical to achieving stereochemical diversity in DOS.34 Although established methods for azomethine ylide generation have proven to be both general and efficient, new procedures are constantly being divulged. These new methods are not only for ylide generation, but are also for new chemical 184 equivalents and by careful selection; the synthetic chemist has a wealth of technologies available at his disposal. 002Me M6020 o ' Ph i3: 002m ‘PerEt. 0 °C. Et3N. -20 °C. AgOAc. 3 mol % Zn(OTf)2, 10 mol % 0 "v. 0 Q QQ 1 Lo) NH H'N F9 N N ' Ar P tBu 1Bu F8 PAI’ 2 tBu-BOX @2 XV'YFFAP 78 %ee, >95 % yield 60%. 90 % yield Jwgensons approach Zhang's approach COz‘Bu ‘Buozc, =/ A 0M8 ¥ . Ph NJ\y0( v ph'Q'I’cozm H ‘PerEt. -20 °C. AgOAc, 10 mol % S-QUINAP Schreiber's approach Figure IV- 8. Synthesis of pyrrolidines from a-iminoesters. However, most 1, 3-dipolar cycloadditions of azomethine ylides catalyzed by chiral metal complexes have shown only endo-selectivity. Because it is important that any of the desired diastereomers of cycloadducts can be synthesized selectively, a study of methods of achieving exo-selectivity has attracted attention of researchers in recent times.3 ' 185 Among the different versions of this reaction, the most practical approach has been the interaction between stabilized N-metalated azomethine ylides derived from a-imino glycinate esters and n-deficient alkenes (Figure IV- 8). The reaction proceeds under mild conditions and with a high degree of diastereocontrol. Silver (1) and lithium (1) metal cations (usually stoichiometric amounts) are most commonly used along with an excess of tertiary amine base. Enantiocontrol has recently been extended to this reaction raising its stock for the generation of complex heterocyclic molecules. While Jorgensen et 0129 used chiral oxazoline complexes of Zn, Zhang and co-workers28 employed Ag (1) complex with FAP ligands for above cycloadditions. Schreiber and co-workers further improved on Zhang’s protocol by expanding the substrate scope and increasing the enantioselectivities achieved by employing the QUINAP ligand.30 An important highlight of their work is that they were able to generate quartenary stereogenic centers by employing substituted amino acids. 186 Ph Ifh N O O 0y y0 A OMe U A Ph NW > Ph COzMe O Et3N, 40 °C. u CU(0Tf)2. 2 "'0' % exo: >95 °/o 0O 64 %ee Pth I i Pth Komatsu's approach Figure IV-9. Exo selectivity tuned using bulky phosphine ligands Recently, the normal endo selectivity of the above cycloadditions has been reversed by Komatsu et 01 by a opportune selection of bulky phosphine ligands in the cycloadditions of N-substituted maleimides and a-imino glycinate (Figure IV- 9).31 Reversing the diastereoselectivity which is under substrate control by using external reagents (Lewis acids) is highly desirable in a diversity oriented synthesis program.34 J. Extension of diastereocontrol and synthesis of exo pyrrolidines Our ultimate aim to develop stereoselective methodology for Al-pyrrolines was to diversify these templates to other heterocycles.“ 7’ 8' 9 Foremost among these aims, was reduction to pyrrolidines. 0t, a-disubstituted a-amino acids like lactacystin (proteosome inhibitor), kaitocephalin (Glutamate receptor antagonist), dysibetaine (glutamate agonist) have generated considerable interest due to their biological activities.'0 187 Unusual regioselection and stereochemical outcome resulted in the reaction of these Al-pyrroline scaffolds (having competing electrophilic centers) with various nucleophiles.” By a judicious choice of conditions we are able to extend diastereocontrol to all four centers of these scaffolds (Scheme IV-6). O o Pia—(I N R control of two stereocenters I AgOAc Ph gantrol of ' rec stereocenters 0:3“?0 Nu o 0}»:th Dcozm DMAP' N" = bcone P" \N " Ph ‘N ’R R 50—75 % R-Ih. lV-1-5 NuIMcOl-l, Iv-1-s “'3'“ ”'1“ Nu- NH,3n,Iv-1-1o NaBH4 65 % MeOH control of NaCNBH3 four stereocenters MeOHzAcOH 80 % Ph 0 O Nvo ,1; Oi—NHPh O §-NHPh 0“ : Bas NU "I ' NU " . . CO M ---N'u-% . COZMe . COZNU Phi“): 2 ° Ph N 'R Ph‘" N ’R R H H H m, IV-1-11 R-Ilo. Nu-Nflan. IV-1-12 Scheme IV-6. Extension of diastereocontrol to all four centers of A'-pyrroline. Thus, bicyclic Al-pyrroline (exo, 3, 4-cis) on treatment with DMAP and nucleophiles (MeOH) resulted in ring opened Al-pyrroline IV-l-9 (exo, 3, 4- trans). Reduction of bicyclic Al-pyrroline with NaBH.. in MeOH resulted in bicyclic pyrrolidine IV-l-ll(exo, 3, 4-cis, 2, 3-trans). Surprisingly, reduction of ring opened Al-pyrroline with NaBH4/ MeOH resulted in reduction of the 5- 188 carboxylic acid (appendage diversity; data not shown). Afier generation of iminium ion in 1:1 acetic acid/MeOH was facile with NaCNBH; and led to monocyclic pyrrolidine IV-l-12 (exo, 2, 3, 4-all trans) diastereomer. Ring opening of bicyclic pyrrolidine was attempted under various conditions to obtain ring opened pyrrolidine (exo, 3, 4-trans, 2, 3-cis) without success. All the above structures have been corroborated with X-ray crystallographic data (Fig. IV-10). Figure IV-10a. X-ray crystal structure of TOP: IV-l-S, BOTTOM: IV-l-9 189 Figure IV-10b. X-ray crystal structure of IV-l-12. K. Enantiocontrol in Al-pyrroline synthesis Moderate enantiomeric excesses have also been achieved in this silver catalyzed process using chiral P, N-ligands, namely (S)-QUINAP.30 Thus azlactone IL] was reacted with N-phenylmaleimide, in presence of 10 mol % AgOAc and S- QUINAP (preformed complex) in THF at -40 °C for 48h. After methylation, purification the Al-pyrroline isolated was analyzed by 'H NMR in presence and absence of Eu (III) shift reagent. 190 Ph '1 1) '1‘ O 0 0% yo :0 0 U = =‘ AgOAc, 10 mol % , 0 THF, 40 C, lV-1-5 O \ 25 96 ee / N i i 9th S-QUINAP 2) TMSCHNZ Z-V Scheme IV-7. Asymmetric synthesis Al-pyrroline The enantiomeric excess estimated from the chemical shift was 25 %. Achieving practically useful enantioselectivity will be actively pursued in near future. L. Experimental Section Reactions were carried out in flame-dried glassware under nitrogen or argon atmosphere, unless otherwise noted. Commercial solvents and reagents were used as received. Anhydrous solvents were dispensed from a delivery system which passes the solvents through packed columns (tetrahydrofuran, methylene chloride: dry neutral alumina). All reactions were magnetically stirred and monitored by TLC with 0.25 pm pre-coated silica gel plates and column chromatography was carried out on Silica Gel 60 (230—400mesh) supplied by EM Science. Yields refer to chromatographically and spectroscopically pure compounds unless otherwise stated. Melting points were determined on a Mel-Temp (Laboratory devices) apparatus with a microscope attachment. Infrared spectra were recorded on a Nicolet IR/42 spectrometer. 'H and ”C NMR spectra were recorded on a Varian Gemini-300 spectrometer, Varian VXR-SOO spectrometer and Unity+-500 191 spectrometer. Chemical shifts are reported relative to the residue peaks of the solvent CDC13 (7.24 ppm for 1H and 77.0 ppm for 13C). HRMS were obtained at the Mass Spectrometry Facility of the Michigan State University with a JEOL J MS HX-l 10 mass spectrometer. Gas chromatography/ low resolution mass spectra were recorded on a Hewlet—Packard 5890 Series II gas chromatograph connected to a TRIO-1 EI mass spectrometer. Combustion analysis was carried out on Perkin Elmer Series II 2400 CHNS/O Analyzer. General Procedure 1) Method A. Synthesis of A'-Pyrroline-2-carboxylic acid methyl ester: A solution of azlactone (Immol), AgOAc (0.1 mmol) and alkene (2 to 3 mmol) in dry THF (50 mL) was stirred under argon in a flame-dried flask in dark for the requisite amount of time as monitored by TLC. Excess trimethylsilyl diazomethane was added and the mixture was stirred for 2 h. Completion of esterification was monitored by TLC (20 % MeOH / EtOAc). The solvent was removed under vacuum and residue was chromatographed on silica gel with an ether / hexanes mixture to isolate A'-pyrroline—2-carboxylic acid methyl esters. 1,3a,4,5,6,6a-hexahydro-l-methyl-4,6-dioxo-3,5-diphenylpyrrolo[3,4- clpyrrole-l-carboxylate methyl ester (IV-l-S): Nh12% >= H3 .~ ‘ H 2-phenyloxazol-5(4H)—one (0.4g, 2.28 mmol), Me(H1) AgOAc (0.038g, 0.228 mmol) and N- According to general procedure using 4-methyl- 192 phenylmaleimide (0.474g, 2.74 mol) at room temperature for 48 h and methylation with excess trimethylsilyl diazomethane, 0.62 g of pyrroline IV-l-S was obtained (75 % yield) as a white solid after silica gel column chromatography (45 % ether / hexanes). 1H NMR (500 MHz), CDC13: 8 1.75 (s, 3H), 3.81 (s, 3H), 4.35 (d, J = 9 Hz, 1H), 4.91 (d, J = 9 Hz, 1H), 7.21-7.22 (m, 2H), 7.36-7.37 (m, 1H), 7.41-7.42 (m, 4H), 7.44-7.45 (m, 1H), 8.16-8.18 (m, 2H); NOESYID: (HI - > H2) 6 %, (H1 -> H3) 1 %, (H2 -> H3) 12 % , (H3 -> H2) 10 % (see X-ray data; crystallized from dichloromethane loctane); l3C NMR + DEPT ( 125MHz) CDC13: 22.16 (-CH3), 50.30 (-CH), 53.62 (l—COZCH3), 57.61 (-CH), 82.14 (quatemary C), 126.58, 128.65, 129.13, 129.42, 130.16, 132.17 (aromatic CH), 131.59, 131.66 (aromatic quaternary 1C), 167.08, 171.58, 172.90, 173.73; (IR (cm'l): 3070, 1780.5, 1718, 1614; M. P. = 186-188 °C. HRMS (FAB): m/z calcd for C21H13N204 [M + H], 363.1345; found, 363.1348; Anal Calcd. for C zthsNzO4: C, 69.60; H, 5.01; N, 7.73. Found: C, 69.06; H, 4.98; N, 7.56. 3,4-diethyl 2-methyl 3,4-dihydro-2-methyl-S-phenyl-2H-pyrrole-2,3,4- tricarboxylate (IV -l-6): According to general procedure using 4-methyl-2-phenyloxazol-5(4H)-one (0.2g, 1.14 mmol), AgOAc (0.019g, 0.114 mmol) and diethyl maleate (0.37ml, 2.28 mol) at room temperature for 48 h and methylation with excess trimethylsilyl diazomethane, 0.31 g of pyrroline IV-l-6 was obtained (75 % yield) as a white solid afier silica gel column chromatography (30 % ether / hexanes). lH NMR (500 MHz), CDC13: 8 1.1 (t, J = 7 Hz, 3H), 1.27-1.29 (m, 3H), 1.48 (s, 3H), 193 3.82 (d, J = 1 Hz, 3H), 4.04-4.12 (m, 2H), 4.16-4.26 (m, 3H), 4.73 (d, J = 6.5 Hz), 7.38 (t, J = 7 Hz, 2H), 7.43 (t, J = 6.5 Hz, 1H), 7.82 (d, J = 8H2, 2H);NOESY1D: (H1 -> HZ) 1 %; (H2 -> H3) 1 %;(Hl -> H3) 1 % (see X-ray crystallographic data; crystallized from ether); 13C NMR + DEPT (125 MHz) coca: 13.73 (- COZCH3) 14.06 (-C02CH3) 20.61 (-CH3), 52.85 (—CO2CH3), 53.94 (~CH), 56.80 (-CH), 61.37 (-CH2), 61.56 (-CH2), 80.51 (quaternary 1C), 128.13, 128.27, 131.12 (aromatic CH), 132.51 (aromatic quaternary 1C), 169.13, 169.89, 170.39, 172.73 (IR (cm'l): 2853, 1738.2, 1622.3; M. P. = 100-103 °C; HRMS (FAB): m/z calcd for C19H23NO6 [M + H], 362.1604; found, 362.1602; Anal Calcd. For C19H23N06: C, 63.15; H, 6.41; N, 3.88. Found: C, 62.99; H, 6.37; N, 3.89. 3,4-Diethyl 2-methyl 3,4-dihydro-2,S-diphenyl-2H-pyrrole-2,3,4- tricarboxylate (IV-2-6) According to general O EtOZC 2:: procedure using 4-methyl-2-phenyloxazol-5(4H)- ”2"... PhQ:/"'"C02Et one (0.2g, 0.84 mmol), AgOAc (0.014g, 0.084 N/g‘COzMe . 13h mmol) and dlethyl maleate (0.273ml, 2.28 mol) at room temperature for 60 h and methylation with excess trimethylsilyl diazomethane, 0.053 g of pyrroline IV-2-6 was obtained (15 % yield) as a yellow oil after silica gel column chromatography (30 % ether / hexanes) which solidified in ether as a low melting pale yellow solid. 1H NMR (500 MHz), CDC13: 8 1.19-1.23 (m, 6H), 3.46 (d, J = 8.5 Hz, 1H), 4.1-4.16 (m, 2H), 4.2-4.3 (m, 5H), 3.53 (d, J = 8.5 Hz, 1H), 7.27-7.31 (m, 1H), 7.32-7.46 (m, 7H), 7.92 -7.8 (m, 2H); NOESYlD: (H1 -> H2) 2 %; 13C NMR + DEPT (125MHz) CDC13: 8 13.83 (-C02CH3), 14.05 (-C02CH3), 61.33 (-CH2), 63.40 194 (-CH2), 65.90 (-COOCH3), 73.08 (-CH), 89.17 (quaternary 1C), 127.37, 127.67, 128.12, 128.47, 128.55, 131.05 (aromatic CH), 132.65, 141.88 (aromatic quaternary 1C), 168.85, 169.67, 171.99; (IR (cm'l):2853, 1734.2, 1618.5; M. P. = 58-60 ° C; LRMS (El): m/z calcd for C24stNOe [M —C02], 363.2; found, 363.3. Anal calcd. for C24stNOs: C, 68.07; H, 5.95; N, 3.31. Found: C, 69.36; H, 6.17; N, 3.34; 3,4-Diethyl-2-methyl-3,4-dihydro-2-methyl-S-phenyl-ZH-pyrrole-2,3,4 tricarboxylate (IV-1-7b) According to general 3 % procedure using 4-methy1-2-phenyloxazol-5(4H)- 151020.,”3002 t phdnz 5% one (021;, 1.14 mmol), AgOAc (0.019g, 0.114 \ N R1381? mmol) and diethyl fumarate (0.37 mL, 2.28 mol.) at room temperature for 48 h and methylation with excess trimethylsilyl diazomethane, 0.216 g (50 % yield) of (IV-1-7a) / 2) was obtained as a white solid after silica gel column chromatography (30 % ether / hexanes). 0.108 g of pyrroline (lV-l-7b) was obtained (25 % yield) colourless viscous liquid in later fractions. 1H NMR (500 MHz), CDC13: 8 1.13 (t, J = 7.2 Hz, 3H), 1.24 (t, J = 7.2 Hz, 3H), 1.86 (s, 3H), 3.52 (d, J = 9 Hz, 1H), 3.62 (s, 3H), 4.08-4.18 (m, 4H), 4.86 (d, J = 9 Hz, 1H), 7.34-7.43 (m, 3H), 7.79-7.82 (m, 2H);NOESY1D: (H2 -> H1) 5 %; (H2 -> H3) 3 %;(H3 -> H2) 2.5 %; 13C NMR + DEPT (125MHz) CDC13: 14.11 (-C02CH2CH3), 14.25 (-C02CH2CH3), 25.42 (-CH3), 52.68 (- COZCH3), 58.51 (-C02CH2CH3), 58.74 (-C02CH2CH3), 61.62 (-CH), 61.90 (- CH), 81.80 (-quaternary C), 128.03, 128.32, 131.17 (aromatic CH) 132.72 195 (aromatic quaternary C), 170.12, 170.8, 171.0, 171.5; HRMS (FAB): m/z calcd for C19H23N06 [M+H] = 361.1604 and found [M+H] = 361.1606. Dimethyl 3,4-dihydro-2-methyl-S-phenyl-2H-pyrrole-2,4-dicarboxylate (IV - 1-8a): According to general procedure using 4- 14 % methyl-2-phenyloxazol-5(4H)-one (0.2g, 1.14 [.3529 H2a 30 % mmol.), AgOAc (0.019g, 0.114 mmol.) and Ph \N ancazMe 2 % methyl acrylate (0.294 g, 3.42 mol.) at room Me (“11 temperature for 60 h, 0.172 g of pyrroline IV-l- 8a was obtained (55% yield) as a yellow viscous material after silica gel column chromatography (25 % ether / hexanes) lH NMR (500 MHz), CDCl3: 6 1.41 (s, 3H), 3.27 (dd, J1= 17 Hz, J2= 9.75 Hz, 1H), 3.53 (dd, J, = 17.5 Hz, J2= 8.25 Hz, 1H), 3.73 (s, 3H), 3.79 (s, 3H), 3.86 (dd, J1= 9.5 Hz, J; = 8.5 Hz, 1H), 7.38-7.41 (m, 2H), 7.43-7.46 (m, 1H), 7.82 -7.83 (m, 2H); NOESYID: (IN -> H2b) 2 % , (H2a -> H2b) 30 % , (H2a -> H3) 14 % (H3 -> H23) 6 %; l3C NMR + DEPT (125MHz) CDC13: 19.81 (-CH3), 38.19 (-CH2), 48.76 (-CH), 52.11 (—C02CH3), 52.90 (-C02CH3), 81.20 (quaternary 1C), 128.01, 128.49, 131.20 (aromatic CH), 133.32 (aromatic quaternary 1C), 172.31, 172.53, 173.63; (IR (cm'l): 2953.4, 1738, 1612.7; HRMS (FAB): m/z calcd for C15H17NO4 [M + H], 276.1236; found, 276.1235. 196 Dimethyl 3,4-dihydro-2-methyl-5-phenyl-2H-pyrrole-2,3-dicarboxylate (IV- 1-8b): additional fractions afforded 0.142 g of product IV-l-8b (45 % yield) as a mixture of two stereoisomers. No further attempts to COzMe separate the two products were successful. 1H NMR P“ ‘ co M N a 2 e \| (500 MHz), CDC13: 6 1.71 (s, 3H), 1.72 (s, 3H), 2.19 (d, J= 14 Hz, 1H), 2.44 (d, J: 14.5 Hz, 1H), 2.75 (d, J= 14 Hz, ll-I), 3.12 (d, J =14 Hz, 1H), 3.72 (s, 3H), 3.74 (s, 3H), 3.78 (s, 3H), 3.79 (s, 3H), 3.93 (s, 1H), 4.36 (s, 1H), 7.36 (t, J = Hz, 3H), 7.43 (t, J = Hz, 3H), 7.81-7.83 (m, 2H), 7.87-7.88 (m, 211); 13C NMR (125MHz) CDC13: 8 24.91, 27.20, 29.67, 48.68, 49.62, 52.60, 53.13, 53.42, 53.72, 86.48, 87.48, 128.03, 128.12, 128.5, 130.99, 131.05, 131.51, 169.0, 170.89, 173.55, 174.45, 174.6, 175.04. l-benzyl-l,3a,4,5,6,6a-hexahydro-4,6-dioxo-3,5-diphenylpyrrolo[3,4- c]pyrrolc-l-carboxylate methyl ester (IV-13-5): According to general procedure using 4-benzyl-2-phenyloxazol- 5(4H)-one (0.2g, 0.8 mmol), AgOAc (0.013g, 0.08 mmol) and N-phenylmaleimide (0.165g, 0.956 mol) at room temperature for 60 h, 0.24 g of pyrroline IV-l3-5 was obtained (68 % yield) as a off-white solid afier silica gel column chromatography (55 % ether / hexanes). 1H NMR (500 MHz), CDC13: 6 3.09 (d, J = 14 Hz, 1H), 3.73 (s, 3H), 3.97 (d, J= 15 Hz, 1H), 4.29 (d, J= 9 Hz, 1H), 4.88 (d, J= 8.5 Hz, 197 1H), 7.02 (d, J = 7 Hz, 2H), 7.21-7.22 (m, 3H), 7.32-7.34 (m, 2H), 7.36-7.41 (m, 4H), 7.49-7.55 (m, 4H), 8.24-8.25 (m, 2H);NOESY1D: (H1 -> H1) 58 °/o, (H2 -> H3) 12 % , (H3 -> H2) 16 % ; l3C NMR + DEPT (125 MHz) CDC13: 41.4 (- PhCH2), 50.74 (-CH), 53.35 (1—C02CH3), 56.87 (-CH), 85.51 (quaternary 1C), 126.64, 127.15, 127.2, 127.35, 128.35, 128.49, 128.71, 129.02, 129.15, 129.22, 129.42, 130.23, 131.21, 131.33, 132.22 (aromatic CH), 131.43, 131.68, 135.58 (aromatic quaternary 1C), 167.11, 171.08, 172.75, 173.19; (IR (cm'l): 2980, 1780.5, 1718.8, 1614.6; M. P. = 165-168 °C. HRMS (FAB): m/z calcd for C27H22N204 [M + H], 439.1658; found, 439.1655. Anal calcd. for C27H22N204: C, 73.96; H, 5.06; N, 6.39. Found: C, 73.3; H, 5.14; N, 6.40. l-(lH-indol-3-yl)methyl)-l,3a,4,5,6,6a-hexahydro-4,6-dion-3,5 diphenylpyrrolol3,4-c] pyrrole-l-carboxylate methyl ester (IV-3-5): According to general procedure using 4-((1H- indol-3 -y1)methyl)-2-phenyloxazol-5(4H)-one (0.2g, 0.69 mmol), AgOAc (0.0123, 0.069 mmol) and N-phenylmaleimide (0.143 g, 0.826 mol) at room temperature for 60h, 0.23 g of pyrroline IV-3-5 was obtained (70 % yield) as a pale brown solid afier silica gel column chromatography (80 % ether / hexanes) and precipitation from dichloromethane / ether mixture. 1H NMR (500 MHz), CDC]; + DMSO-daz 8 3.55 (d, J = 15 Hz, 1H), 3.72 (s, 3H), 3.86 (d, J = 15 Hz, 1H), 4.23 (d, J= 9 Hz, 1H), 4.79 (d, J= 9.5 Hz, 1H), 6.29 (d, J= 8 Hz, 2H), 6.91 (m, 1H), 6.95 (t, J = 7.5 Hz, 1H), 7.0-7.02 (m, 3H), 7.07 (t, J = 7 Hz, 1H), 198 7.38 (t,J=7Hz,2Hz), 7.43 (t,J= 7Hz,1H), 7.55 (d,J= 8 Hz, 1H), 8.l7(d,J= 8 Hz, 2H), 9.03 (broad s, 1H);NOESY1D: (Hl -> H1) 42.5 % , (H1 -> H2) 0.5 % , (HZ -> H3) 7.5 % , (H3 -> H2) 9 % ; 13C NMR + DEPT (125 MHz) CDC13 + DMSO-da: 28.86 (-IndolleH2), 48.86 (-CH), 52.74 (1—C02CH3), 56.73 (-CH), 85.35 (quaternary 1C), 110. 66, 118.48, 119.03, 121.07, 123.97, 125.46, 125.55, 127.93, 128.02, 129.57, 131.3 (aromatic CH), 108.05, 130.67, 131.11, 135.27 (aromatic quaternary 1C), 167.08, 171.58, 172.9, 173.73; (IR (cm‘l): 3402, 1747.7, 1709.15, 1614.6; M. P. = 230-232 °C. HRMS (FAB): m/z calcd for C29H23N3O4 [M + H], 478.1767; found, 478.1768.; Anal Calcd. for C29H23N3O4: C, 72.94; H, 4.85; N, 8.8. Found: C, 71.4; H, 4.84; N, 8.44. l,3a,4,5,6,6a-hexahydro-l,3-dimethyl-4,6-dioxo-S-phenylpyrrolo[3,4-c] pyrrole-l-carboxylate methyl ester (IV-S-S): According to general procedure using 2,4- H §=O . 2]?" d1methyloxazol-5(4H)-one (0.2g, 1.77 mmol), AgOAc 2 \ ; OzMe - (0.029g, 0.177 mmol.) and N-phenylmaleimide (0.37g, M6011) 2.12 mol) at room temperature for 48 h, 0.31 g of pyrroline IV-S-S was obtained (59 % yield) as a white solid after silica gel column chromatography (80 °/o ether/ hexanes) and precipitation from dichloromethane / ether mixture. 1H NMR (500 MHz), CDC13: 8 1.65 (s, 3H), 2.3 (s, 3H), 3.8 (s, 3H), 4.17 (d, J = 8.5 Hz, 1H), 4.22 (d, J = 9 Hz, 1H), 7.21-7.23 (m, 2H), 7.4 (d, J = 6.5 Hz, 1H), 7.44-7.47 (m, 2H);NOESY1D: (H1 -> H2) 1 %, (H1 -> H3) 0.4 % (H2 and H3 are coupled and have very close chemical shifls, hence no value for NOE could be obtained); '3C 199 NMR + DEPT (125 MHz) CDC13: 18.56 (1-CH3), 21.96 (3-CH3), 49.47 (-CH), 53.34 (1—COZCH3), 60.58 (-CH), 82.33 (quaternary 1C), 126.19, 126.3, 128.97, 129.28 (aromatic CH), 131.28 (aromatic quaternary C), 169.0, 171.57, 172.7, 173.61; (IR (cm"): 2955, 1780.5, 1716.8, 1653.2; M. P. = 156-158 °C. HRMS (FAB): m/z calcd for C16H16N204 [M + H], 301.1188; found, 301.1189. 3-Benzyl-1,3a,4,5,6,6a-hexahydro-l-methyl-4,6-dioxo-5-phenylpyrrolo[3,4-c] pyrrole-l-carboxylate methyl ester (IV -9-5): According to general procedure using 2-benzy1-4-methyloxazol-5(4H)-one (0.2g, 3 % 0 Ph 1.057 mmol), AgOAc (0.018g, 0.106 mmol) and N- /H—32I 0 phenylmaleimide (0.22g, 1.26 mol) at room H2 ph \N M ((383048 temperature for 48 h, 0.31 g of pyrroline IV-9-5 was e 1 obtained (75 % yield) as a hydroscopic white solid foam after silica gel column chromatography (65 % ether / hexanes). 1H NMR (500 MHz), CDC13: 6 1.62 (s, 3H), 3.79 (s, 3H), 3.93-4.01 (m (d + q), 3H), 4.16 (d, J = 8.5 Hz, 1H), 7.18-7.2 (m, 2H), 7.27 (d, J = 7 Hz, 1H), 7.31-7.41 (m, 5H), 7.44-7.47 (m, 2H);NOESY1D: (H1 -> H2) 2 %, (H2 -> H3) 3 % ((H2 and H; are coupled and have very close chemical shifts, hence dtflicult to get a value for NOE)); 13C NMR + DEPT (125 MHz) CDC13: 21.77 (-CH3), 38.47 (PhCH2-), 49.37 (-CH), 53.27 (1—C02CH3), 57.81 (-CH), 82.17 (quaternary C), 126.28, 127.29, 128.95, 128.99, 129.28, 129.51 (aromatic CH), 131.26, 134.76 (aromatic quaternary 1C), 170.8, 171.65, 172.51, 173.5; (IR (cm'l): 2920, 1780.5, 1738, 200 1722.6, 1643.5; M. P. = 46-48 °C. HRMS (FAB): m/z calcd for C22H20N204 [M + H], 377.1501; found, 377.1500. 2) Method B) Two step synthesis of Al-Pyrroline-Z-carboxylic methyl esters using silver acetate catalyst: A solution of azlactone (1.0 mmol), AgOAc (0.1 mmol.) and alkene (2 to 3 mmol) in dry THF (50mL) was stirred under argon in a flame-dried flask in dark for the requisite amount of time. The crude residue was dissolved in 9:1 benzene / methanol mixture (50 mL). Assuming quantitative conversion from azlactone to the A‘-pyrro1ine-2-carboxylic acid, the equivalent amount of trimethylsilyl diazomethane was added and mixture stirred for 2 h and completion of methylation monitored by TLC. The solvent was removed under vacuum and residue was chromatographed on silica with ether / hexanes mixture to isolate AI-pyrroline-Z-carboxylic acid methyl esters. l,3a,4,5,6,6a-hexahydro-l-methyl-4,6-dioxo-3,5-diphenylpyrrolo[3,4- clpyrrole-l-carboxylate methyl ester (IV-l-S): Using 4-methyl-2- phenyloxazol-5(4H)-one (0.4g, 2.28 mmol), N h 12 % AgOAc (0.038g, 0.228 mmol) and N- 11321: phenylmaleimide (0.474g, 2.74 mmol) 0.2 g of Ph \ N M 0(2er 6 % pyrroline IV-l-S was obtained (50 % yield) as a e 1 white solid after silica gel column chromatography (45 % ether / hexanes) and precipitation from dichloromethane / ether mixture. Characterization was identical to method A. 201 Dimethyl 3-(phenylcarbamoyl)-3, 4-dihydro-Z-methyI-S-phenyl-ZH-pyrrole— 2, 4-dicarboxylate (IV-l-9): After all IV-l-S was precipitated, the filtrate on 3 % evaporation afforded 0.15 g of pyrroline IV-l-9 (35 % .Ph MeO N _ 1 yleld). H NMR (500 MHz), CDCl;: 6 1.5 (s, 1H), 03% P , ”2 3.69 (s, 3H), 3.99 (s, 3H), 4.11 (d, J= 9.5 Hz, 1H), 7 % 0.919, M 5.09 (d, J = 9.5 Hz, 1H), 7.14 (t, J = 7 Hz, 1H), 7.36 (t, J = 8 Hz, 1H), 7.42 (t, J = 8 Hz, 2H), 7.48 (m, 1H), 7.61 (d, J = 8.5 Hz, 2H), 7.83 (d, J = 8 Hz, 2H), 8.98 (s, 1H);NOESY1D: (H1 -> H2) 3 %, (H1 -> H3) 7 % (H2 -> H3) 3 % , (H3 -> H2) 3 % (see X-ray data; crystallized from dichloromethane /octane); 13C NMR + DEPT (125 MHz) CDC13: 21.94 (1-CH3), 52.81 (4-C02CH3), 53.67 (1—C02CH3), 55.17 (-CH), 56.32 (-CH), 80.36 (quaternary 1C), 119.63, 124.39, 126.55, 127.82, 127.97, 128.54, 129.04, 131.34 (aromatic CH), 132.61, 137.78 (aromatic quaternary 1C), 166.26, 169.1, 171.42, 177.04; (IR (cm’l): 3346, 2953, 1738 (broad), 1680.2, 1599.1; M. P. = 155-157 ° C; HRMS (FAB): m/z calcd for C22szNzOs [M + H], 395.1607; found, 395.1608; Anal Calcd. for C221-122N205: C, 66.99; H, 5.62; N, 7.1. Found: C, 67.16; H, 5.48; N, 7.13. 3) Synthesis of (IV-l-9) from (IV-l-S) in methanol using DMAP: Compound (IV -1-5) (0.02g, 0.055 mmol) was suspended in 20 m1 of methanol in a glass vial 202 having a screw cap. DMAP (0.007g, 0.06 mmol) was added and reaction mixture stirred at room temperature. The reaction was monitored by TLC (l: R; = 0.38 and 1b: Rf = 0.35, 50 % ether / hexanes) and aliquots were taken out and the solvent was evaporated and the residue dried under vaccum for lHNMR. After determination of ratio of 1:11 by 1H NMR, the residue was chromatographed as outlined above in procedure 2. 4) Synthesis of (IV-l-lo) from (IV-l-S): Compound (IV-l-S) (0.05g, 0.138 mmol) was suspended in 20 mL of benzylamine in a flame-dried flask under nitrogen and DMAP (0.016g, 0.14 mmol) was added and mixture stirred at room temperature for 36 hours. The mixture was dissolved in 50 mL dichloromethane and extracted with 10 % HCl solution (3x, 30 mL). The dichloromethane layer was dried over anhydrous sodium sulphate and evaporated under vacuum. The residue was chromatographed with 45 % ether / hexanes to afford the product as a white solid foam (0.057g, 76 % yield). (ZS, 3S, 4S)-N2,N‘-dibenzyl-3,4-dihydro-2-methyl-Na,5-diphenyl-2H-pyrrole- 2,3,4-tricarboxamide (IV-l-10): 1H NMR (500 MHz), CDC13: 8 1.62 (s, 1H), 3.84 (d, J =10 Hz, 1H), 4.31 (dd, 11 = 4.5 Hz, 12 = Ph RN ,0, .Ph 1 % 14.5 Hz, 2H), 4.45 (dd, J1 = 4.5 Hz, J2 = 14.5 Hz, H3». . 0 Ph 2 2H), 4.59 (dd, J1 = 6 Hz, 12 = 15 Hz, 2H), 4.75 (dd, "Me (H1) 2 % NH 11 = 6.5 Hz, 12 = 14.5 Hz, 2H), 4.84 (d, J = 10 Hz, P“ 1H), 6.98 (broad s, 1H), 7.15 (t, J = 7 Hz, 11-1), 7.2- 203 7.52 (m, 14H), 7.65 (d, J = 8 Hz, 2H), 7.75 (d, J = 6.5 Hz, 2H), 10.92 (s, 1H); NOESYID: (H1 -> H2) 1% , (HI -> H3) 2 % (H2 -> H3)1% , (H3 -> H2)1%; 13C NMR + DEPT (125 MHz) CDC13: 22.5 (1-CH3), 43.77 (~CONHCH2Ph), 43.9 (—CONHCH2Ph), 56.01 (-CH), 57.5 (-CH), 78.54 (quaternary 1C), 120.21, 124.55, 127.18, 127.76, 127.82, 127.93, 128.02, 128.2, 128.78, 128.87, 128.95, 129.07, 129.18, 129.24 (aromatic CH), 131.54, 133.13, 137.5, 138.27, 138.37 (aromatic quaternary 1C), 167.93, 170.21, 170.62, 176.75; (IR (cm'l): 3325, 2925, 1676,1660,1649.3, 1599.2; M. P. = 80-83 ° C; HRMS (FAB): m/z calcd for C34H32N4O3 [M + H], 545.2552; found, 545.2554; Anal Calcd. for C34H32N4O3: C, 74.98; H, 5.92; N, 10.29. Found: C, 74.16; H, 5.88; N, 10.13. 4) Synthesis of (IV-l-ll) from (IV-l-S): Compound (IV-l-S) (0.1g, 0.275 mmol) was suspended in 20 mL of MeOH in a flame-dried flask under nitrogen Ph and NaBH4 (0.020 g, 0.55 mmol) was added N 2.4 % . . H z 3: and mixture strrred at room temperature for 3 3 = _.- @1186 I‘D 0 5 0/ hours. After all the starting material had ‘ -. 2 e . 0 H N - 1% “H Meth reacted, MeOH was evaporated off. The mixture was dissolved in 50 mL dichloromethane and extracted with 5 % HCl solution (3x, 30 mL). The dichloromethane layer was dried over anhydrous sodium sulphate and evaporated under vacuum. The residue was chromatographed with 80 % ether / hexanes to afford the product as white solid foam (0.076g, 76 % yield). 204 1H NMR (500 MHz), coca: 8 1.71 (s, 3H), 3.72 (s, 3H), 3.97 (d, J = 15.5 Hz, 1H), 4.19 (d, J: 15.5 Hz, 1H), 5.52(d, J = 10 Hz, 1H), 7.18-7.48 (m, 8H), 7.94- 7.97 (m, 2H);NOESY1D: (H1 -> H2) 0.5 %, (H2 -> H3) 2.4 %, (H3 -> H4) 1 % ; ”C NMR + DEPT (125MHz) CDC13: 22.19 (-CH3), 50.95 (-CH), 53.25 (1— coch3), 57.84 (-CH), 82.93 (quaternary C), 85.93(-CHPh), 124.49, 127.22, 128.94, 129.03, 129.16, 131.88 (aromatic CH), 131.96, 136.40 (aromatic quaternary 1C), 171.24, 171.78, 173.51; (IR (cm'l): 3360, 2928.3, 1738.1, 1680.2; HRMS (FAB+): m/z calcd for C21H20N204 [M + H], 364.3945; found, 364.1413. 4) Synthesis of (IV-l-12) from (IV-1-5): Compound (IV-l-S) (0.05g, 0.091 mmol) was suspended in 20 mL of MeOH/AcOH (1:1) in a flame-dried flask under nitrogen and NaBHr (0.016g, 0.14 mmol) was added and mixture stirred at room temperature for 3 hours. After all the starting material had reacted, MeOH was evaporated off. The mixture was dissolved in 50 mL dichloromethane and extracted with saturated ammonium chloride (3x, 30 mL). The dichloromethane layer was dried over anhydrous sodium sulphate and evaporated under vacuum. The residue was chromatographed with 50 % Ether/hexanes to afford the product as white solid foam (0.032 g, 66 % yield). 'H NMR (500 MHz), CDC13: 8 1.53 (s, 1H), 3.3.43 (t, J = 11 Hz, 1H), 3.74 (d, J = 12 Hz, 1H), 4.292-4.631 (complex multiplet , 5H), 6.6 (t, J = 6 Hz, 1H), 7.03 (t, J = 3.5 Hz, 2H), 7.12-7.39 (m, 15H), 7.62 (d, J = 8 Hz, 2H), 8.43 (t, J = 5.5 Hz, 1H), 10.334 (broad 205 singlet, 1H); NOESYlD: (H1 -> H3) 4 % , (H1 -> H2) 2 %, (H2 -> H3) 1 %, (H3 -> H2) 1 °/o, (H2 -> H4) 6 % , (H3 -> H4) 2 %; (see X—ray data; crystallized fi'om dichloromethane /octane); '3 C NMR + DEPT (125 MHz) CDC13: 24.1 (1-CH3), 43.47 (-CONHCH2Ph), 43.73 (—CONHCH2Ph), 55.47 (-CH), 58.4 (-CH), 64.47 (-CHPh), 66.75 (quaternary 1C), 120.12, 124.41, 127.14, 127.35, 127.73, 127.94, 128.72, 129.14, 129.29 (aromatic CH), 137.83, 138. 36,138.38, 138.88 (aromatic quaternary 1C), 169.0, 170.66, 178.2. 206 References. (a) Shvekhgeimer, M.-G. A. Chemistry of Heterocyclic Compounds 2003, 39(4), 405. (b) Miltyk, W.; Palka, J. A. Comparative Biochemistry and Physiology Part A 2000, 125, 265-271. (c) Beame, S. L.; Hekmat, O.; MacDonnell, J. E. Biochem. J. 2001, 356, 223-232. ((1) Beame, S. L.; Wolfenden, R. W. Biochemistry 1995, 34, 11515-11520. (e) Stapon, A.; Li, R.; Townsend, C. A. J. Am. Chem. Soc. 2003, 125, 8486-8493. (a)Leusch, H.; Hoffinann, D.; Hevel, J. M.; Becker, J. E.; Golakoti, T.; Moore, R. E. J. Org. Chem. 2002, 68, 83-91. (b) Stocking, E. M.; Martinez, R. A.; Silks, L. A.; Sanz-Cervera, J. F .; Williams, R. M. J. Am. Chem. Soc. 2001, 123, 3391-3392. (c) Helms, G. L.; Moore, R. B.; Niemczura, W. P.; Patterson, G. M. L. J. Org. Chem. 1988, 53, 1298- 1307. (d) Kuo, M. S.; Yurek, D. A.; Coats, J. H.; Chung, S. T.; Li, G. P. J. Antibiot. 1992, 45, 1773-1777. (a) Kleinsasser, N. H.; Wallner, B. C.; Harreus, U. A. Zwickenpflug, W.; Richter, E. Toxicology 2003, 192, 171-177. (b) Ohkura, H.; Berbasov, D. O.; Soloshonok, V. A. Tetrahedron Letters 2003, 44, 2417-2420. (c) Morris, B. D.; Prinsep, M. R. J.Natl. Prod. 1999, 62, 688-693. ((1) Hughes, C. C.; Trauner, D. Angew. Chem. Int. Ed. 2002, 41 , 4556-4559.(e) Carroll, A. R.; Pierens, G. K.; Fechner, G.; de Almeida Leone, P.; Ngo, A.; Simpson, M.; Hyde, B.; Hooper, J. N. A.; Bostrom, S. L.; Musil, D.; Quinn, R. J. J. Am. Chem. Soc. 2002, 124, 13340-13341. (t) Tsukamoto, D.; Shibano, M.; Kusano, G. Chem. Pharm. Bull. 2001, 49, 1487-1491. (g) Usubillaga, A.; Zabel, V.; Watson, W. H. Acta Crystallographica Section C-crytstal structure communications 1982, B38, 966-969. (h) Tomko, J .; Brazdova, V.; Voticky, Z. Tetrahedron Lett. 1971, 32, 3041-3043-3044. (i) Yuichi, S.; Masaki, S.; Atsuko, M.; Takahashi, Y.; Hiroshi, T.; Satoshi, O.; Yutaka, E. J. Antibiot. 2003, 56, 817-826. (a) Moya, P.; Cantin, A.; Castillo, M.; Prino, J.; Miranda, M. A.; Primo- Yufera, E. J. Org. Chem. 1998, 63, 8530-8535.; (b) Birouk, M.; Harraga, S.; Panouse-Pen'in, J .; Robert, J. F .; Damelincourt, M.; Theobald, F.; Mercier, R.; Pannouse, J. J. European Journal of Medicinal Chemistry 1991, 26, 91-99.; (c) Fuska, J.; Fuskova, A.; Vassova, A.; Voticky, Z. Neoplasma 1981, 28, 709-714. (a) Dannhardt, G.; Keifer, W. Arch. Pharm. Pharm. Med Chem. 2001, 334, 183.; (b) Goti, A.; Cicchi, S.; Mannucci, V.; Cardona, F.; Guama, F.; Merino, P.; Tejero, T. Org. Lett. 2003, 5(22), 4235.; (c) Lombardo, M.; Fabbroni, S.; Trombini, C. J. Org. Chem. 2001, 66, 1264.; (d) Ballini, R.; Marcantoni, B.; Petrini, M. J. Org. Chem. 1992, 5 7, 1316.; (e) Clerici, F.; Gehni, M. L.; Pocar, D.; Rondena, R. Tetrahedron 1995, 51 (36), 9985.; (f) 207 Goti, A.; Cicchi, S.; Mannucci, V.; Cardona, F.; Cordero, F. M.; Brandi, A. Org. Lett. 2001, 3(9), 1367. (a) Fehn, S.; Burger, K. Tet. Asymm. 1997, 8(12), 2001.; (b) Sakai, R.; Oiwa, C.; Takaishi, K.; Karniya, H.; Tagawa, M. Tet. Lett. 1999, 40, 6941.; (c) Okue, M.; Kobayashi, H.; Shin-ya, K.; Furihata, K.; Hayakawa, Y.; Seto, H.; Watanabe, H.; Kitahara, T. Tet. Lett. 2002, 43, 857.; (d) Watanabe, H.; Okue, M.; Kobayashi, H.;Kitahara, T. Tet. Lett. 2002, 43, 861.; (c) Sun, P.; Sun, C.; Weinreb, S. M. Org. Lett. 2001, 3, 3507-3510. (a) Tehrani, K. A.; D'hooghe, M.; De Kirnpe, N. Tetrahedron 2003, 59, 3099-3108.; (b) Peroche, S.; Remuson, R.; Gelas-Mialhe, Y.; Grarnain, J. C. Tetrahedron Lett. 2001, 42, 4617-4619.; (b) Xia, Q.; Ganem, B. Tetrahedron Lett. 2002, 43, 1597-1598.; (c) Pearson, W. H.; Stevens, E. P. J. Org. Chem. 1998, 63, 9812-9827.; ((1) Graf v. Keyserlingk, N.; Martens, J. Tetrahedron .‘Asymmetry 2001, 12, 2213-2222.; (e) Yamago, S.; Miyazoe, H.; Nakayarna, T.; Miyoshi, M.; Yoshida, J. Angew. Chem. Int. Ed 2003, 42, 117-120.; (f) Ohkura, H.; Berbasov, D. 0.; Soloshonok, V. A. Tetrahedron Letters 2003, 44, 2417-2420.;(g) Yu, M.; Pagenkopf, B. L. J. Am. Chem. Soc. 2003, 125, 8122-8123.; (h) Ballini, R.; Marcantoni, E.; Petrini, M. J. Org. Chem. 1992, 57, 1316-1318.; (i) Cordero, F. M.; Valenza, S.; Machetti, F.; Brandi, A. Chem Commun. 2001, 1590-1591.; (1) Shin-Ya, K.; Kim, J.-S.; Furihata, K.; Hayakawa, Y.; Seto, H. Tetrahedron Lett. 1997, 38, 7079-7092.; (k) Watanabe, H.; Okue, M.; Kobayashi, H.; Kitahara, T. Tetrahedron Lett. 2002, 43, 861-864.; (1) Okue, M.; Kobayashi, H.; Shin-Ya, K.; Furihata, K.; Hayakawa, Y.; Seto, H.; Watanabe, H.; Kitahara, T. Tetrahedron Lett. 2002, 43, 857-860.; (m) Plehiers, M.; Heilpom, S.; Ekehnans, D.; Leclercq, S.; Sangermano, M.; Braekman, J. C.; Daloze, D. Can J. Chem. 2000, 78, 1030-1034. (n) Grigg, R.; Millington, E. L.; Thornton-Pett, M. Tetrahedron Lett. 2002, 43, 2605-2608. (a) Goti, A.; Cicchi, S.; Mannucci, V.; Cardona, F.; Guama, F.; Merino, P.; Tejero, T. Org. Lett. 2003, 5, 4235-4238. (b) Chapman, T. M.; Courtney, S.; Hay, P.; Davis, B. G. Chem. Eur. J. 2003, 9, 3397-3414. (c) Pisaneschi, F.; Monica, C. D.; Cordero, F. M.; Brandi, A. Tetrahedron Letters 2002, 43, 5711-5714. ((1) Goti, A.; Cacciarini, M.; Cardona, F.; Cordero, F. M.; Brandi, A. Org. Lett. 2001, 3, 1367-1369. (e) Lombardo, M.; Fabbroni, S.; Trombini, C. J. Org. Chem. 2001, 66, 1264-1268. (0 Black, D. S.; Edwards, G. L.; Evans, R. H.; Keller, P. A.; Laaman, S. M. Tetrahedron 2000, 56, 1889-1897. (a) Belokon, Y. N.; Bhave, D.; D'Addario, D.; Groaz, B.; Maleev, V.; North, M.; Pertrosyan, A. Tetrahedron Lett. 2003, 44, 2045-2048. (b) Humphrey, J. M.; Charnberlin, A. R. Chem. Rev. 1997, 97, 2243-2266. (c) Dennisova, 1.; Barriault, L. Tetrahedron 2003, 59, 10105-10146. ((1) 208 10. 11. 12. 13. 14. 15. 16. 17. 18. 19. Manthrope, J. M.; Gleason, J. L. Angew. Chem. Int. Ed. 2002, 41, 2338- 2340. (e) Fenteany, G.; Standaert, R. F.; Soongyu Choi, W. S.; Corey, E. J.; Schreiber, S. L. Science 1995, 268, 726. (1) Corey, E. J.; Guzrnan- Perez, A. Angew. Chem. Int. Ed. 1998, 37, 388-401. (a) Kleinsasser, N. H.; Wallner, B. C.; Harreus, U. A.; Zwickenpflug, W.; Richter, E. Toxicology 2003, 192, 171-177; (b) Hughes, C. C.; Trauner, D. Angew. Chem. Int. Ed 2002, 41 , 4556-4559; (c) Ma, D.; Yang, J. J. Am. Chem. Soc. 2001, 123, 9706-9707. (a) Kercher, T.; Livinghouse, T. J. Org. Chem. 1997, 62, 805-812; (b) Campos, P. J .; Soldevilla, A.; Sampedro, D.; Rodriguez, M. A. Org. Lett. 2001, 3, 4087-4089; (c) Pearson, W. H.; Stevens, E. P. J. Org. Chem. 1998, 63, 9812-9827; ((1) Harwood, L. M.; Vickers, R. J. Chemistry of Heterocyclic Compounds 2002, 59, 169-252. (e) Uchiyarna, K.; Yoshida, M.; Hayashi, Y.; Narasaka, K. Chem. Lett. 1988, 607-608. (a) Padwa, A. 1,3-Dipolar Cycloaddition Chemistry, 1984; (b) Gribble, G. W. Mesoionic ring systems. The Chemistry of Heterocyclic Compounds, Volume 60, Part A; Jon Wiley and Sons, 2002; pp 681-753. Padwa A.; Pearson, W. H., Synthetic applications of dipolar cycloaddition chemistry towards heterocyclic and natural products; Wiley-VCH: Weinheim, 2002. (a) Huisgen, R.; Gotthardt, H. Chem. Ber. 1970, 103, 2625-2632; (b) Huisgen, R.; Gotthardt, H.; Bayer, H. 0. Chem. Ber. 1970, 103, 2368- 2387; (c) Gotthardt, H.; Huisgen, R.; Schaefer, F. C. Tetrahedron Lett. 1964, 10, 487-491; ((1) Huisgen, R.; Gotthardt, H.; Bayer, H. O. Tetrahedron Lett. 1964, 9, 481-485; (e) Grigg, R.; Lansdell, M. I.; Thornton-Pett, M. Tetrahedron 1999, 55, 2025-2044. Maryanoff, C. A.; Karash, C. B.; Turchi, l. J. J. Org. Chem. 1989, 54, 3790-3792. Gingrich, H. L.; Baum, J. S. In Oxazoles; Turchi, I. J., Ed.; Wiley: New York, 1986; Vol. 45,731. Friedrichsen, W.; Sch8er, W.-D. Tetrahedron Lett. 1977, 1603-1605. Padwa, A.; Gingrich, H. L.; Lim, R. J. Org. Chem. 1982, 47, 2447-2456. (a) Peddibhotla, S.; Jayakumar, S.; Tepe, J. J. Org. Lett. 2002, 4, 3533- 3535; (b) Peddibhotla, S.; Tepe, J. J. Synthesis 2003, 9, 1433-1440. 209 20. 21. 22. 23. 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. 34. 35. (a) Pearson, W. H.; Stoy, P.; Mi, Y. J. Org. Chem. 2004, 69, 1919-1939. (b) Harwood, L. M.; Macro, J.; Watkin, D.; Williams, C. E.; Wong, L. F. Tetrahedron: Asymmetry 1992, 3, 1127-1130. Shanna, V.; Lansdell, T.; Peddibhotla, S.; Tepe, J. J. Chem. Biol. 2004, in print. Chen, F. M. F.; Kuroda, K.; Benoiton, N. L. Synthesis 1970, 230. Washizuka, K.; Minakata, S.; Ryu, I.; Komatsu, M. Tetrahedron 1999, 55, 12969-12976. Grigg, R.; Lansdell, M. 1.; Thornton-Pett, M. Tetrahedron 1999, 55, 2025- 2044. Peddibhotla, S.; Tepe, J. J. J. Am. Chem. Soc. 2004, 126(40), 12776- 12777. Blazey, C. M.; Heathcock, C. H. J. Org. Chem. 2002, 6 7, 298-300. Sustmann, R. Pure Appl. Chem. 1974, 40, 569-593. Longmire, J. M.; Wang, B.; Zhang X.; J. Am. Chem. Soc. 2002, 124, 13400-13401. Gothelf, A. S.; Gothelf, K. V.; Hazel], R. G.; J Drgensen, K. A.; Angew. Chem. Int. Ed 2002, 41 (22), 4236-4238. Chen, C.; Li, X.; Schreiber, S. L.; J. Am. Chem. Soc. 2003, 125, 10174- 10175. Oderaotoshi, Y.; Cheng, W.; Fujitomi, S.; Kasano, Y.; Minakata, S.; Komatsu, M. Org. Lett. 2003, 5, 5043-5046. Padwa, A.; Lim, R.; MacDonald, J. G. J. Org. Chem. 1985, 50, 3816- 3823. Galley, G.; Liebscher, J .; Patzel, M. J. Org. Chem. 1995, 60, 5005-5010. Martin D. Burke and Stuart L. Schreiber Angew. Chem. Int. Ed 2004, 43, 46—58. Gong, Y.-D.; Kim, S.-K.; Yoo, S.; Kurth, M. J. Bull. Chim. Soc. Jpn. 2002, 75, 2477-2480. 210 CHAPTER V APPLICATIONS OF IMIDAZOLINE SCAFFOLDS IN THERAPY AND ORGANOCATALYSIS. A. Sensitization of Tumor Cells towards Chemotherapy. l) Enhancing the activity of Camptothecin by Novel Imidazolines Apoptosis or programmed cell death is a cellular mechanism that maintains the cell number and eliminates damaged or mutated cells." 2 Alterations in apoptotic pathways can disrupt the delicate balance between cell proliferation and cell death leading to a variety of diseases?” 4 In many cancers, apoptosis is abnormally down-regulated, either by the mutation of pro-apoptotic proteins or by the up- regulation of anti-apoptotic proteins.5 Aberrant apoptosis provides an intrinsic survival advantage to cancer cells causing growth and expansion of the tumor, and resistance to pro-apoptotic signals including chemotherapeutic agents." 7 Chemotherapeutic agents may also induce anti-apoptotic factors thereby adding to this intrinsic resistance to chemotherapy.8 The combination of these anti-apoptotic mechanisms has resulted in an increased dose intensity of chemotherapeutics, often without the anticipated improved therapeutic results.9 The search for new chemotherapeutic strategies has therefore shifted to small molecules that can selectively induce apoptosis in cancer cells or retard the cellular chemoresistance.lo Strategies using combinations of inducers of apoptosis and/or inhibitors of anti-apoptotic factors and traditional chemotherapeutic drugs may provide an improved alternative to conventional chemotherapy. 'Ob' ” 211 2) Chemotherapy and chemoresistance - camptothecin and cis-platin The antitumor agent camptothecin (CPT) is an alkaloid isolated from the extracts of the fruit of Camptotheca acuminata and was identified as a topoisomerase 1 inhibitor.“ ‘3 CPT-11 and several water soluble analogues including topotecan have successfully passed clinical trials in the United States.14 Camptothecin exhibits its antitumor activity via the formation of a stable ternary topoisomerase I - DNA cleavable complex.“ '5 Stabilization of this cleaved DNA complex initiates an apoptotic signaling pathway, ultimately resulting in cell death.“ '6 Concomitant with the initiation of this apoptotic cell signal, these agents induce anti-apoptotic signaling pathways, which have compromised their efficacy in the clinic.” 27" This cellular resistance has been attributed to the activation of anti- apoptotic signaling pathways mediated by several transcription factors, in d’ 18’ 2" cDNA microarrays particular the nuclear transcription factor, NF-icB.8 using all annotated human genes have been used to establish the association between characteristic gene expression patterns in response to drug treatment by the topoisomerase poison camptothecin.l9 These studies in HeLa cells provided striking evidence that administration of camptothecin resulted in up-regulation in a large number of genes controlled by NF-KB.'9’ NF -1cB mediated anti-apoptotic response induced by DNA damaging agents may also result from the NF -1 4 , A )1 —v—Cr'r+1-sr-4-84 g E O as” :2 r 0 I I I I T I 1 Figure V-7. Decrease in the tumor size in RIF-1 tumors. Tumor volume (y-axis) vs. Days (x-axis) to ‘four times’ volume. Chemotherapeutic drug: 8 mg/ 1kg; Imidazolines: 100 mg/ 1kg.” 8) Significance Antitumor drugs exert their antitumor activity via the initiation of apoptotic signaling pathways ultimately resulting in cell death. Concomitant with the initiation of this apoptotic cell signal, many chemotherapeutic agents induce anti- apoptotic signaling pathways, which have compromised the efficacy of these antitumor agents in the clinic. Inhibition of this anti-apoptotic transcription factor retards this cellular resistance. The chemosensitizing agent 1 was found to sensitize leukemia cells at nanomolar concentrations to camptothecin and drastically enhance the level of camptothecin mediated apoptosis. Combination therapy of CEM cells with camptothecin and the imidazoline 1, provided ~75 fold enhancement of efficacy after 48 hours. The ability of the imidazoline to enhance the apoptotic events initiated by camptothecin is postulated to be via the 225 inhibition of the NF-KB signaling pathway. These studies indicate that combination therapy of classical anticancer agents with small molecule NF-ch inhibitors has the potential to target this chemoresistance and provides a new strategy to treat chemoresistant tumors. Work towards a detailed molecular mechanism and the clinical potential of these agents is currently under investigation in our laboratory. 226 B. Catalytic Asymmetric Synthesis of 4, 5-dihydro-l, 3-oxazin-6-ones. Precursors to a, ll-dialkyl-B-amino acids using Imidazoline-4-amides as Novel Organocatalysts 1) Introduction to [l-amino acids. During the last decade, stereoselective synthesis of B-amino acids has gained considerable attention due to their structural and important pharmacological 50 They exhibit powerful anti-bacterial properties and are key properties. constituents of naturally occurring peptides, terpenes, alkaloids, and macrolides, and as potential precursors for fl-Iactams.5 " 52 It is well known that the B-amino, a-hydroxyphenylpropionic acid side chain in taxol53 and related terpenoids is crucial for their anti-cancer activity. Substituted aspartic acid derivatives are currently of interest medicinally for use as inhibitors of L-asparagine syntlretase,54 key constituents of several proteins involved in blood-clotting cascade55 and for their ability to act as non-transportable glutamate transporter blockers.56 Although less abundant than the (1.-amino acids, B-amino acids occur in nature in both free and bound form in peptides. They tend to be stronger bases and weaker acids compared to 11.-amino acids. B-amino acids are hence used as a-amino acid surrogates for the construction of peptides which, because of different skeletal pattern possess interesting folding patterns and conformational properties and hence enhanced activity and metabolic stability.57 The main approaches58 available till date for the stereoselective synthesis of 8- amino acids include Amdt-Eistert homologation of a-amino acids, enzymatic resolution, alkylation of enolates derived from perhydropyrimidin-4-ones and 227 aspartic acid derived oxazolidinones and butanolides, hydride reduction of chiral aziridine carboxylate esters and subsequent oxidation of product alcohols”, addition of enolates or their equivalents to imines, Curtius rearrangement, Michael addition of nitrogen nucleophiles to 01,13-unsaturated esters or imides with or without trapping of intermediate enolates with electrophiles at the a-position, hydrogenation, amino hydroxylation and B-lactam synthesis. Although a plethora of synthetic methods use chiral auxillaries or substrate control for achieving stereoselectivity, catalytic asymmetric synthesis of B-amino acids is just coming of age. Jacobsenboand Miller“ 62 have exploited the sequential azidation/ reduction of a, B-unsaturated carbonyl compounds to this end. A different approach recently reported by Boger utilizes the Sharpless asymmetric amino hydroxylation of cinnarnate esters.63 2) Nucleophilic catalysts and stereocontrol in reaction of ketenes and imines Chemists have long used ketenes as useful intermediates in the synthesis of organic molecules. Since Staudinger’s original discovery, numerous research groups have made advances both in the generation and reactions of ketenes. Catalytic stereoselective transformations of ketenes via [2+2] and [4+2] cycloadditions are at the forefront of research in present times."4 228 O é': Ts.N 10 mol %, catalyst O at“ JL "' k ’ .R; * R1 R; H R toluene, rt t . R 1. ,1 3 75-98 % ix R2 3 RzN 80-98 % ee m \N e. 2% Fu’s catalyst Figure V-8. Planar chiral heterocycles for enantioselective additions to ketenes Fu and co-workers have used the planar-chiral heterocycles (Figure V-8) as catalysts for the enantioselective addition of alcohols to ketenes and anhydrides.65 They have also extended the utility of these catalysts in the enantioselective synthesis of B-lactam derivatives.66 The reaction is mechanically distinct /inverse from the usual [2+2] cycloaddition or the Staudinger reaction between ketenes and imines. General mechanism for a nucleophilic catalyst mediated stereocontrol in fi-lactam synthesis is shown in Figure V-9. Addition of a tertiary amine to a ketene presumably generates ammonium enolate A. This enolate can react with an electrophilic imine generating zwitterionic intermediate B. Cyclization of the intermediate B generates the B-lactam and regenerates the catalyst. Asymmetric induction is imparted at the intermediate A, where the catalyst blocks one n-face of the ammonium enolate, thereby setting the or-stereogenic center. The catalyst further controls the orientation of the approaching electrophile (imine) through non-bonding interactions, thereby setting the B-stereogenic center. 229 Nu: Nucleophilic catalyst O ,T 11 R RKILR 1R2R 2 G O Na NU&R‘ NUVR R; B A Ts}i H R Figure V-9. Mechanism for stereocontrol by a nucleophilic catalyst Lectka67 and co-workers have reported a remarkable catalytic system consisting (Figure V-10) of a chiral organic base and a Lewis acid. They have used 10 mol % benzoquuinine (Cinchona alkaloid) as chiral nucleophilc or organocatalyst. Addition of 10 mol % In (111) triflate resulted in dramatic enhancement of reaction rates and chemical yields. In fact, the same catalyst performs additional roles of generating the imine in situ and also ring-opening by alcohols amines and amino acids to provide B-amino acid derivatives. 230 82‘ O 60 % O H.N,Bz 10 mol % catalyst Tj MeOH BZ‘NH 0 R\/u\01 + :Etozc“'|\/u\oule EtOZCXCI 9'9“)" $90099 EtOZC“. "'R catalyst |n(OTf)3 R toluene — 95 % ee -78 °C to rt t O 82 N C ‘N JL” 'k R H c0251 Lackta's approach Figure V-10. Use of Cinchona alkaloid derivative as a multifunctional nucleophilic catalyst 3) Imidazolines as nucleophilic catalysts 68' 69 reported a diastereoselective multi-component synthesis of We recently highly functionalized imidazolines (Scheme V-l). A silicon mediated 1, 3 dipolar cycloaddition of the in situ generated munchnone with an imine resulted in the formation of highly substituted imidazolines. The imidazolines contain four-point diversity and two stereocenters and the cycloaddition reaction is applicable to aryl, alkyl, acyl and heterocyclic substitutions. The most widely used organocatalysts have a five membered heterocyclic moiety, proline, Kagans’s prolinol, MacMillian’s imidazolidinone catalyst.70 We realized that above imidazolines, on account of unique arrangement of groups around a rigid five- membered ring had a accessible face comprising of the iminic nitrogen and carboxyl group and a hindered face. Hence they seem to have a pocket or catalytic core with requisite groups and could possibly be used as organocatalysts. Thus, the present studies focused on the synthesis of enantiopure imidazoline derivatives and their use as chiral organic bases / catalysts for the reaction of ketenes with imines. 231 R4-NH2 0 o '3‘ R 1mf R3CHO = R1\«N R3 R2 TMSCI (1.3 eq.)/ CHZCIZ NI COzH ’R2 reflux azlactone trans (major) 0 0 C(10) cm) . " g 0 Cl2ll 2’ 4t 5) o _ o ‘ 35’8"“.- Ct3l o' ‘ 91‘. 0‘25 :-'* “23’ ms: 0 . "\"lb 0 . “(9 om: 0" cl. Scheme V-l. TOP: Multi-component synthesis of imidazolines. BOTTOM: X-ray structure of III-7-l. In order to achieve imidazoline catalyzed enantioselective B-lactam synthesis few reactions were attempted. They have resulted in the formation of the amide probably formed from the intermediate acyliminium ion (Scheme V-2). It has been found in the literature that only N-tosyl iminoesters7| (electron deficient) have been used in the asymmetric synthesis using nucleophilic catalysts. But there is no report concerning the use of nucleophilic imines in the catalytic synthesis of B-lactam. Initial attempts to achieve the B-lactam synthesis using N—benzylidene- benzylamine (III-I-l) in presence of racemic imidazoline-4-carboxylic acid and 232 esters were not successful and resulted in the formation of the corresponding amide (Scheme V-2). Ph \\N Ph Ph—<\ I N 002Et O 0 CI to ,-78°Ct rt T uene ot PhAuJK’Ph Ph PhANAPh 09 ® PhA hikr Ph PhH Scheme V-2. Failed attempt to B-lactams using racemic imidazoline-4- carboxylates. N-tosyl imine of ethyl glyoxylate resulted in the formation of trans-B-lactam in moderate yield, using non nucleophilic base (proton sponge) and racemic ethyl imidazoline-4-carboxylate. Whereas the synthesis of B-lactam using only DMAP and TEA resulted in the formation of mixture of cis and trans (1:4) B-lactam in good yields (Scheme V-3).72 233 ¢ r: Nov 0: S—0 benzene 0: 8:0 NC|3 reflux ACOOEt N“ 9% Ph > N Ph he Cl 5- COOEt Ts‘Ni/(O ‘ Ill-1 -10 m .. O: S: O “ ,1. NW; NMe2 5‘0“: ”A... toluene -78°C to rt Scheme V-3. B-lactams from N-tosyl-a-iminoester using racemic imidazoline catalyst 4) Preparation of chiral imidazoline-4-amides catalysts NH2 ....H Me Ph~\ Phfi Ph\\N N Ph single enantiomer N Ph Ph ph—<\ 1"“ > Ph—(x at} Me + Ph—(\ :91)" H N N -' '1 COOH EDCI. DMAP 5 Y n’ ”Ye CH2CI2 O P“ P" (t) ""14 +I- V-amlde-1 separated by column chromatography Scheme V-4. Synthesis of chiral imidazoline—4-amide catalysts Resolution of 1-benzyl-4-methyl-2,5-diphenyl-4,5-dihydro-lH-imidazole-4- carboxylic acid with R-(+)-1-phenyl-ethylamine using EDCI and DMAP in DCM resulted in the diastereomeric mixture which was separated by column chromatography, providing enantiopure amides (Scheme V-4). Similarly S-(-)-1- phenyl-ethylamine also resulted in the formation of diastereomers separable by 234 column chromatography.72 We decided to use imidazoline 4-amide as catalysts for following reasons; i) they could be obtained in enantiomerically pure form. ii) the amide side chain may form a hydrogen bond with the enolate (zwitterionic species formed with ketene) and thus stabilize the enolate. iii) it may enhance the steric block on a n-face of the enolate and achieve high amount of stereocontrol. The absolute configurations are unknown for the amides because in the x-ray structure the a-methyl group was missing in the amide side chain and hence had to be discarded. 5) Three component synthesis of 4, 5-dihydro-l, 3-oxazin-6-ones (Toluene-4-sulfonylimino)-acetic acid ethyl ester (1 mmol) and phenyl ketene generated in presence. of proton sponge (1 mmol) and the catalyst (+)-V-amide-l (30 mol %) were allowed to react at -78°C for 12 h and two products, which could not be separated by column chromatography were obtained (Entry V-l, Table V- 1). The reaction was carried out over a longer time (-78°C, 36h) (Entry V-4, Table V-l) and the 4, S-dihydro, 1, 3-oxazin-6-one was formed in moderate yields after some optimization (by crude spectra) and was purified by column chromatography. The mass spectra of the product indicated the addition of two molecules of ketene to (toluene-4-sulfonylimino)-acetic acid ethyl ester. The lH NMR spectra and '3 C NMR spectra also supported the addition of two molecules of ketene to (toluene-4-sulfonylimino)-acetic acid ethyl ester. The structure of the product 2-benzylidene-6-oxo-5-phenyl-3-(toluene-4-sulfonyl)-[l, 3] oxazinane-4- 235 carboxylic acid ethyl ester V-6a was unequivocally established by X-ray crystallographic data (Figure V-l 1). Figure V-l 1. X-ray structure showing trans geometry of dihydrooxazinone. The reaction was repeated using 7 equivalents of ketene (Entry V-7, Table V-l) and 60 mol % of catalyst (Entry V—6, Table V-l) but no significant increase in the yield was achieved. Use of stoichiometric catalyst led to complex reaction mixture.72 236 NM°2NM92 0 0 Ph 0 2eq. Ph\)\ CI 0‘ _ Ph‘E‘ko + If ' \ Ph N, + 23M. -78°C, 36h R? ”k R1 T03 1 N R1 W TOS . eq. TOS’ V N catalyst optically active racemic Ill-I-15 Ph\« Ph V-6a V-6b N : XYPh - 0 R2 Entry R1 R, x Catalyst 3320:":gdn3: % yleld(a) % yleld(b) v.1 -COzEt Me N (+)-V4mlde-1, 0.3 eq. 1eq. products not separable“ V-2 -COzEt Me N (+)-V-amlde-1, 0.3 eq. 1eq. products not separable‘ V-3 -COzEt Me N (+)-V4mlde-1. 0.3 eq. 1eq. poor yields V4 -COzEt Me N (+)-V-amlde-1, 0.3 eq. 2.5 eq. 30 % - V6 -C02Et Me N (-)-V-amlde-1 0.3 eq. 2.5 eq. 30 % - V-6 -COzEt Me N (+)-V4mlde-1 0.6 eq. 2.5 eq. 37% 10 % V-7 -C02Et Me N (+)-V4mlde-106 eq. 7 eq. 37% 10 % V-8 -C02Et Me N (+)-V-amlde-1,1eq. 2.5 eq. messy reaction V-9 -Ph Me N (+)-V4mlde-1, 0.3 eq. 2.5 eq. 20% 10% V-10 -Ph Me N (+)-V4mlde-1, 0.6 eq. 2.5 eq. 35% 10% V-11 -Ph Me O (-)V-ester-2, 0.6 eq. 2.5 eq. 0% 50% V42 -Ph H N (+I-)V.amlde-2, 0.6 eq. 2.5 eq. 0% 0% d. toluene, -78 °C, 12h; e. DCM. -78°C, 12h Ph Ph W 1 Ph N Ph \ H N . 0 Ph N = "Y P" i Y ' 0 R2 0 R2 +I- V-amlde-1 +I- V-ester-z Table V-l. Synthesis of dihydrooxazinones using imidazoline based catalysts 237 Several reports in the 1970’s indicated the formation of the racemic form of the above dihydrooxazinone.”77 One report from Furukawa et 0178 described the asymmetric synthesis of B-amino-a, cl-dimethyl-B-phenylpropionic acid via the asymmetric synthesis of the dihydrooxazinones by using a chiral imine auxillary. To our knowledge, this is the first example of a catalytically induced asymmetric multi-component synthesis of dihydrooxazinones. These dihydrooxazinones can simply be hydrolyzed to the chiral a,B-substituted,B-amino acids.” The proposed mechanism for the formation of the dihydrooxazinones is depicted in the following Scheme V-S.72 Acid chloride or ketene can react with imidazolines-amide leading to the formation of intermediate A (see also Figure V- 2). It is possible intermediate A, the ammonium enolate is stabilized because of the hydrogen bond with the amide -NH. Intermediate A on reaction with imine leads to another intermediate B (only one n-face of enolate blocked by catalyst). Intermediate B collapses to form B-lactam in cases where other catalysts are used. In our case steric block (possibly the cl-methyl of amide side chain) prevents C-N bond rotation and cyclization. Intermediate B hence reacts with another molecule of ketene and leads to the formation of intermediate C which on intramolecular ring closure and elimination of catalyst affords the 2-alkylidene-4, 5-dihydro-l,3- oxazin-6-one (Scheme V-6). The above proposed mechanism seems very likely as, the dihydrooxazinone was obtained as a single diastereomer and in 70 % enantiomeric excess. 238 dihydrooxazinone] HIPhO l HV /2 9 EtOOéh P“ N i HN Ph H imidazoline- 4- amide >..... Ph P AN Ph”\ Ph h )tj O N .u“ Ph Ph \ Ph “(9 NH ph 0 Ph COOEt WAQQ Ts—N H A we Ph H C Ph"\ ph Tst 9 N . a C Ph \PQM N H 0008 Scheme V-S. Proposed mechanism for catalytic synthesis of dihydrooxazinones 6) Problems so far72 1. Importance of the 4—amide side chain, need to be ascertained clearly. 2. The preparation of (toluene-4-sulfonylimino)-acetic acid ethyl ester is not trivial and the imine has to be used immediately because it decomposes very fast. 3. The highest yield obtained is 30 % and this results in very difficult separation problems. The product has to be subjected to column chromatography at least three times to get analytically pure material. 239 4. The absolute stereochemistry of catalyst imidazoline-4-amides is not known as the x-ray structure misses the a-methyl group from the l-phenylethyl side chain. This observation could not be rationalized by simple reasoning. 5. The enantiomeric excess need to be determined with more confidence. The racemic dihydrooxazinone is needed for this purpose. To this end, resolution by aminolytic ring opening with optically active amino acid has been attempted but without any success. 7) Modifications to methodology 1. (Toluene-4-sulfonylimino)-acetic acid ethyl ester is highly unstable and the preparation is neither trivial nor reliable in terms of yields or purity. Hence, we decided to try aromatic aldimines, which are more stable and hence allow several manipulations of the reaction condition and optimization of product yields and enantiomeric excess. Under identical conditions as used before, catalyst (+)- V-amide-l (30 mol %) resulted in 20 % yield with N-benzylidinetosylamine (III- I-lS) (Entry V-9, Table V-l). Doubling the amount of catalyst increased the yields to 35 %, indicating that aryl aldimines are less reactive than a-iminoesters. However, they are a lot easier to prepare and are stable molecules which have long shelf life. Even the multi-component reaction is much cleaner than with the tosylimino ester. 2. Replacing the 4-amide side chain with the ester, catalyst V-ester-l (60 mol %) afforded the B-lactam in 50 % yield. No dihydrooxazinone was observed (Entry V-ll, Table V-l). 240 3. Racemic benzylamide V-amide-Z did not result in a productive reaction (Entry V-12, Table V-l). 8) Three component to a two-component synthesis of dihydrooxazinones Continuing with our pursuit for a more reactive imine than N-benzylidine tosylamine(llI-I-15) and more stable than the tosyliminoesters, made us focus on the improved procedure by Lectka and co-workers79 for synthesis of B-amino esters with Cinchona alkaloid derivatives. They generated N-acylimines by in situ dehydrohalogenation of N-acyl—a-chloroaminoesters. The imines were reactive but again unstable; hence preparation of imines in situ was preferred. Kobayashi80 have reported an asymmetric Mannich reaction using N-acylimines generated by using polymer bound bases. We decided to pursue the use of Kobayashi’s method for the generation of N-acylimines from N-acyl-a-chlorogylcinate ethyl ester using polystyrene bound piperidine as the base of choice (Scheme V-6).80 We realized an inherent advantage in using the N-acylimines, not only because they are they reactive but they present an internal nucleophilc after enolization. i. In essence, we would have achieved tethering of the second equivalent of ketene involved in the three component synthesis of dihydrooxazinones. ii. The internal nucleophilc (enolic —OH) would probably lead to higher rates of dihydrooxazinone synthesis vs. the background non-catalyzed B-lactam synthesis. iii. More diversity can be introduced, because of replacement of second equivalent of ketene with the acyl group from the imine. 241 iv. The product can be expected to be more reactive than the 2-exo- substituted dihydrooxazinones that were obtained before. Ph N CO Et NH 2 )0" j; : PhYNVCOZEt 0 Ph . / k EtO c“ N Ph ”RAG T t 2 v 13 CHZCIZ, -78°C—rt, 4o % _" (e: Proton sponge ; Nu: Ph Cat Ph\\N i (nucleophile) Ph 0 EtOsz/k Ph—<\ :KN NYMG Ph NU Ph’k o i 0 Ph a tethered nucleophilc catalyst Etozc NH TS B-amino acid and derivatives Scheme V-6. Synthesis of 2-aryl dihydrooxazinones from N-acylimines N-benzoylimino ethyl glycinate was made by dehydrohalogenation of N-benzoyl- a-chloro ethylglycinate in a separate vessel using polystyrene bound piperidine. The imine was then transferred to a separate reaction vessel and reacted with phenylacetyl chloride using (+) V-amide-l and proton sponge, using the same conditions as before (Scheme V-6). Afier acid-base workup and a short silica-gel column, the product was precipitated using DCM/ether/hexanes mixture. Although, both proton NMR and EIMS indicated the proposed dihydrooxazinone, the material could not be obtained analytically pure. Dihydrooxazinones like V- 13, six membered analogues of azlactones have been reported as intermediates during Amdt-Eistert reaction of peptides.8| They are extremely short-lived and prone to polymerization.82 242 Though disappointing, the chemistry is possible and with appropriate substitution it should be possible to isolate pure 2-aryldihdyrooxazinones. It would be interesting to see their use as templates for synthesis of heterocycles in comparison to azlactones. 243 C. Chiral 4-hydroxymethyl imidazolines as ligands for diethylzinc addition. 1) Introduction The synthesis of organic compounds is ultimately dependent on the formation of carbon-carbon bonds. It is, therefore, clear that the most expeditious route to chiral compounds is one in which a carbon-carbon bond and a stereocenter are formed in a single step with high enantioselectivity.83 The use of catalytic quantities of metal complexes containing chiral ligands has proven to be a particularly fruitful approach to asymmetric catalysis.84 Generally, the emphasis in designing ligands has been on maximizing the difference in steric effects between the alternative pathways in the enantio discriminating step. However, in recent times the important role of electronic effects in influencing enantioselectivity is being recognized and exploited, and some examples of strong electronic effects have been reported, mostly involving phosphorus ligands. 85 2) Imidazolines as N-heterocyclic carbene ligands 2-Imidazolines have found wide application as potent N-heterocyclic carbene ligands in organometallic catalysis.86 N-Heterocyclic carbenes (NHCs) are being used increasingly in organometallic chemistry as neutral two-electron-donor ligands, commonly replacing phosphines in that role. Some attractive features87 of NHCs are: i. The strong o-donating capability and low level of n-acidity of these ligands provide their metal complexes with electronic properties that are often quite different from those with phosphines or other traditional neutral ligands.88 This change can sometimes enhance the reactivity of metal-based catalysts that feature 244 NHCs. Examples of this improved reactivity include ruthenium-based olefin metathesis catalysts89 and palladium-based catalysts for C-C and C-N coupling reactions.90 ii. The possible diversity in the substituents on the nitrogen atoms, which allows 92‘ 93 and electronic features. for a wide range of steric, 9' asymmetric, iii. It is possible to functionalize these nitrogen substituents in such a way as to make ligands capable of chelation.” 95 Such variability has led to the synthesis of NHC analogues of many traditional ligands. iv) In contrast, to the corresponding phosphine complexes, ligand dissociation has never been observed so that these catalysts normally do not depend on an excess of ligand.86 These properties make them suitable for chiral modifications. 3) Imidazolines as nitrogen ligands Nitrogen ligands are widely used in asymmetric catalysis and, while relatively few examples of electronic tuning have been reported pronounced effects have been noted.84 Oxazolines and amines are the most widely used chiral nitrogen ligands, but unfortunately there is no straightforward approach to electronic tuning of either of these types of donor. Although ligands carrying an imidazoline unit are structurally very similar to oxazoline ligands, they have not received much attention.94 4-Substituted 2-imidazolines are potentially useful as catalysts and ligands, especially since they have the same shape as oxazolines and should therefore give comparable enantioselectivity. Phosphino-oxazolines 2 (PHOX ligands) are versatile chiral ligands that have found a wide range of applications in asymmetric catalysis.95 Cationic iridium complexes derived from these ligands 245 have proven to be highly effective catalysts for the enantioselective hydrogenation of imines and olefins, including unfunctionalized alkenes.96 Crystal structures of PHIM (Phosphino-imidazoline) complex and the corresponding PHOX complex, bearing the same substituents at the phosphorus atom (o-Tol) and the stereogenic center (tBu), closely resemble each other (Figure V-12). However, the torsion angles (1:) between the central phenyl ring and the five-membered heterocycle differ by 20 ° (C-C-C-O angle = -11° vs C-C-C-N angle = -31°). The Ir-P distances (2.3009(12) in li vs 2.3055(17) A in Ir-2) and the Ir-N distances (2.080(5) vs. 2.106 (3) A) are almost identical, indicating that the electron densities on the coordinating nitrogen atoms are similar.94 + . Q31 o-Tok I, l . o-Tok. D . o-Tol’ ‘2) PF6 o-Tol’ PF“ MN MW Figure V-12. PHIM and PHOX ligands in solid state. Anions and COD’s not shown. 246 However there are some important differences between the two ring systems: 84' 94 i. Imidazolines are much stronger bases than oxazolines. As a result they are stronger donors, which may be an advantage in some reactions. ii. The additional nitrogen atom provides a handle for tuning the electronic and conformational properties of the ligand by proper choice of the R3 group. iii. R3 group could also serve as a linker for attaching the ligand to a solid support. iv. Importantly, changes in R3 substituents do not significantly affect the chiral environment created by the group at the 4-position, i.e. the electronic and steric properties can be varied independently of each other. Despite this potential, there are few reports of their use as chiral ligands. Brunner made a brief reference to their use in Rh-catalyzed hydrosilation reactions97 and Morimoto showed that imidazolines bearing an additional sulfur donor gave excellent enantioselectivity in Pd—catalyzed allylations.98 Recently, Davies used ruthenium complexes of pyridyl imidazolines as Lewis acids.99 Dupont used biiimidazolones in Pd-catalyzed hydroarylations.loo Claver reported that pyridyl imidazolines are useful ligands for Pd-catalyzed alkene/CO copolymerizations and noted some electronic effects'o' Brusacca found that the nature of the 1- substituent of 2-(phosphinoaryl)imidazoline ligands had a substantial effect on the Yield and enantioselectivity of an intramolecular Heck reaction.‘02 247 i. 2-hydroxymethylimidazolines ligands for diethylzinc additions to aldehydes Substantial effort has been put forth to develop methods to promote the asymmetric addition of alkyl groups to aldehydes and ketones since 50 years.83 For many years, these investigations were largely unsuccessful due to the highly reactive nature of the organomagnesium and organolithiurn reagents employed. The breakthrough did not come until 1984, when Oguni103 discovered that organozinc reagents added enantioselectively to aldehydes in the presence of chiral amino alcoholsm' '05 Extensive mechanistic studies, primarily from the Noyori group with DAIB (Figure V-12),'°6‘“° have greatly contributed to the present understanding of the mechanism of reaction. The design of catalysts for the asymmetric addition of organozinc reagents to aldehydes to give chiral secondary alcohols has become the focus of intensive research in present times.'°4’ '05' ‘08 A large number of catalysts for this reaction have been developed that rely on either Lewis basic or Lewis acidic for catalyzing the reaction.lll The addition of diethylzinc to aldehydes has been studied exhaustively and amino alcohol catalysts are available that give excellent enantioselectivity for a wide range of aldehydes. While there is no pressing need to develop new catalysts, the surprising and dramatic electronic effects described above with imidazoline are deserving of further study in their own right. Casey et al prepared a series of chiral 2-(hydroxyalkyl)imidazolines from enantiopure amino alcohols.”ll2 These imidazolines were expected to be similar to B-aminoalcohols in electronic properties, and were used as catalysts for the 248 addition of diethylzinc to aldehydes (Figure V-13). The study was designed mainly to observe the effect of ligand electronics on enantioselctivity as this aspect has rarely been studied so far.84 Et2Zn O - OH /U\ 5 % ligand /'\/ P" H toluene, rt P“ aldehyde R1 R2 % ee, configuration Bn CF3 1 5%, R ipr CF3 62%, S iPr Me 21%. R iPr OMe 87%, S tBu OMe 26%, S \ ‘8 0 Zn\ R2 H ”J ”a \ 1 ,’ HO N .,’R E /Zn Et Ph . 1 t 1493’” transition state Figure V-13. 2-hydroxymethylimidazoline ligands for diethyl zinc addition. The following trends were observed by Casey et (I!84 i. Diastereomer with C4 substituent on opposite face of carbinol group as shown in Figure V-l3, was the only active isomer. This indicated that the stereochemistry at both centers and especially at carbinol center was important. ii. The predominant formation of (S)-alcohols from (S)-carbinol ligands was consistent with the observed trend for amino alcohol catalysts. 249 iii. Isopropyl substituent at C4, gave the best result, but further increase in size to t-butyl was detrimental. iv. The predominant formation of (S)-alcohol has been explained based on accepted models as shown in Figure V-13. v. Most striking feature of the para substituents on l-aryl group (R2), are that groups with stronger inductive effects gave higher % ee’s. vi. The electron donating 4-methoxy substituent results in R—alcohol like the electron withdrawing 4-trifluoromethyl group. This was suggested to be because of coordination of diethyl zinc, which makes 4-methoxy sucbstituent to be electron withdrawing. The results reported above emphasize the importance of the concept of ligand designs that readily permit ‘orthogonal’ tuning of steric and electronic effects, and illustrate the potential of imidazolines in asymmetric catalysis, especially as electronically tunable alternatives to oxazolines. ii. Our 4-hydroxymethylimidazolines as ligands for diethylzinc additions To gain quick access to a diversity of ligands, a modular system comprised of simple components is essential. Utilizing amino acids from the chiral pool allows for the easy incorporation of Chirality into the ligands and opens the way for a number of straightforward chemical modifications. Amino acids have been used as the basis for a variety of catalyst systems, and ligands that are based on proline, valine, tyrosine, tryptophan, serine, and leucine have been reported.”l We have recently reported highly diastereoselective multicomponent one-pot synthesis of substituted imidazolines from amino acid based azlactones.”3‘ ”4 These low 250 molecular weight imidazoline scaffolds contain four point diversity applicable to alkyl, aryl, acyl, and heterocyclic substitution. In light of the exciting work by Casey et a1 and the use of modular ligands based on amino acids for diethyl zinc additions to aldehydes, we decided to explore our chiral 4-hydroxymethyl imidazolines as ligands. For this purpose racemic imidazoline-4-carboxylic acids derived from the cycloadditions were converted to diastereomeric esters using R (+)-l- phenylethanol. The diastereomeric esters were separated by column chromatography. Reduction with LAH, afforded the chiral 4- hydroxymethylimidazolines in excellent yields (Scheme V-7). The absolute configuration was determined for one pair of enantiomeric 4-hydroxymethyl imidazolines (Figure V-14). The 4-hydroxymethyl imidazolines tested proved to be moderately good ligands for addition of diethyl zinc to benzaldehyde. The following trend could be seen from the limited number of experiments performed (Table V-2). i. The S, S-carbinol imidazolines resulted in S-configuration 1- phenylpropanol. ii. The 1-(4-methoxyphenyl) substituted ligand showed reverse preference for configuration of l-phenylpropanol as was seen by Casey and co- workers. iii. Use of stoichiometric quantity of additive Lewis acid, Titanium isopropoxide following the method of Walsh and co-workers (Method B, 251 Table V-2) also resulted in opposite configuration of l-phenylpropanol. There was no improvement in observed ee values. QH RY“? R,""_, R R2 EDCI. MAHCI COZH DCMP Ill-1a racemic R4 R R A . R2 "j; 3 1 N r0 R1’<\N 1 R2 0 \FPh _ ; H0 1) separated by + RRR-ester column chromatography RR-alcohol 60—65 °/o 2) Reduction R4 R3 LAH, THF. 0°C N 55—65 % _ R14” -“R2 H0 SSR-ester SS-aloohol Ester R1 R2 R3 R4 alcohol V-1-14 Ph Me Ph 8n V-1-17 v-1-15 Ph Me Ph 4'32";ng v.1-1a _ _ 4~methoxy _ _ V1116 phenyl Me Ph Bn V1119 Scheme V-7. Synthesis of chiral 4-hydroxymethyl imidazolines from racemic imidazoline-4-carboxylic acid (III-la) 252 N N Ph .leh Ph Ph \« \6 N "'I\ N 5 OH = OH R, R S, S Figure V- 14. X-ray structures of chiral 4-i1yd1ux ,ruetl. y" '4 " (R, R) -V- 1-17 and (S, S) -V-1-17 as ViewerLite 3D models. iv. Use of chiral amides (of R-l-phenylethylamine), as Seto and co-workers have successfully employed, did not work in this case. v. 4-isopropyl ligands could not be obtained as the corresponding esters could not be separated by column chromatography. 253 R9 éph R9 Ph R149: R‘AN ' ““ HO/ EtZZn 0 10 mol °/o ligand 0“ H0 PNJLH M thod AIB Ph e RR-aloohol SS-aloohol Ligands R, a" common Method at yield at on, configuration Ph Bn SS A 60 29%. S Ph Bn SS 8 80 0% Ph Bn RR A 89 27%. R Ph Bn RR B 95 35%. S“ 4-methoxy Ph phenyl SS A 50 30%. R 4-methoxy phenyl Bn RR A 65 32%, R Method A: toluene. it Method B: Ti(0‘Pr)4 (1.2 eq.), toluene. rt * only 15 mol °/oTi(O‘Pr)4 Table V- 2. Chiral 4-hydroxymethy imidazoline ligands in diethyl zinc additions. Although, additional experiments are needed to establish synthetic utility of the quarternary 4-methyl-4-hydroxymethylimidazolines as ligands for diethyl zinc additions their performance is not all too disappointing in comparison to the 2- hydroxymethylimidazolines.84 254 D. Experimental Section General: Melting points were determined by open capillary method using Electrothermal Melting Point apparatus (Melt-Temp) and are uncorrected. IR spectra were recorded on a Nicolet spectrophotometer. lH NMR spectra were recorded with Varian [(Inova or Gemini) 300MHz] spectrometers. Chemical shifi values are expressed as 6-(ppm ) and J values are in Hz. Splitting patterns are indicated as s: singlet, d: doublet, t: triplet, m: multiplet, q: quartet and br: broad peak. l3C-NMR spectra were also recorded on Varian [(Inova or Gemini) 75.MHz] spectrometers. The symbols q, t, d and s indicate the number of protons (3, 2, 1, and 0 respectively) attached to each carbon atom, as determined by the '3C DEPT technique. Mass spectra were recorded on Perkin Elmer mass spectrometer. Column chromatography was performed on a silica gel (60-120) mesh. THF/diethyl ether were dried over sodiumbenzopenone ketyl and distilled under nitrogen. Dichloro methane dried over calcium hydride distilled under nitrogen. All manipulations were conducted over a nitrogen atmosphere by use of standard Schlenck techniques. Synthesis of l-Benzyl-4-methyl-2,5-diphenyl-4,5-dihydro-lH-imidazole-4- carboxylic acid (l-phenyl-ethylyamide V-amide-l from l-Benzyl-4-methyl- 2,5-diphenyl-4,5-dihydro-lH-imidazoIe-4-carboxylic acid: To a well-stirred suspension of 1-Benzyl-4-methyl-2,5—diphenyl-4,S-dihydro—1H-imidazole-4- carboxylic acid III-l-1( 1.0g, 0.27 mmol) in dry methylene chloride (25mL), (R)- 255 (+)-l-Phenyl-ethylamine (0.36g, 29mmol) was added EDCI.HC1 (0.57g, 29mmol), after five minutes added a solution of DMAP (.35 gm, 29 mmol) in methylene chloride (lOmL) and stirred for 5-6 hrs. The reaction mixtures was washed with water (2x 10 mL), saturated sodium bicarbonate (20 mL), water (20mL), 2N HCl (20 mL) and then with water (30mL). The organic layer dried over sodium sulfate and evaporated under reduced pressure. The crude product was purified by column silica-gel chromatography using ethyl acetate hexane mixture (1:1). (+)-V-amide-l: Yield: (0.26 g, 66.6%). {[a]D= +41 .5°)} 1H NMR (300MHz) : 5 1.02 (d, J = 6.9, 3H), 1.56 (s, 3H), 3.85 (d, J = 15.6, 1H), 4.40 (s, 1H), 4.66(d, J =15.6 ,lH), 4.72 (t, J= 6.9, 1H), 7.07-7.09 (m, 2 H), 7.17-7.55(m, 16 H), 7.69- 7.73 (m, 2H).; l3C NMR (75MHz): 21.39, 27.56, 48.09, 48.73, 72.66, 126.52, 127.24, 127.71, 127.99, 128.42, 128.57, 128.67, 128.95, 129.01, 129.14, 130.75, 130.82, 137.38, 137.60, 143.29, 165.44, 171.61. IR (neat) 1874 cm-, 1498 cm-; m/z= 473(M+) (-)-V-amide-l: (0.24 g, 61%). {[ot]D= -37.7°)} 'H NMR (300 MHz) : 8 1.40 (d, J = 7.2, 3H), 1.61 (s, 3H), 3.77 (d, J = 15.6, 1H), 4.37 (s, 1H), 4.60(d, J =15.6 ,1H), 4.75 (t, J= 7.5, 1H), 6.922-7.090 (m, 2H), 7.11-7.22 (m, 13H), 7.507-7.529 (m,3H), 7.651-7.682(m, 211).: '3c NMR (75MHz): 21.58, 28.08, 47.97, 48.59, 72.62, 126.66, 126.99, 127.200, 127.69, 127.96, 128.21, 128.51, 128.58, 128.64, 129.13, 129.122, 130.70, 130.83, 137.184, 137.22, 143.28, 165.35, 171.62. IR (neat) 1872 cm-, 1498 cm-; m/z=325 (M+-CO-NH-CH(CH3)Ph). 256 . Synthesis of l-BenzyI-4-methyl-2,5-diphenyl-4,5~dihydro-lH-imidazole-4- carboxylic acid benzylamide from 1-Benzyl-4-methyI-2,5—diphenyI-4,5- dihydro-lH-imidazole-4-carboxylic acid V-12:To a well-stirred suspension of 1 -Benzyl-4-methyl-2,5-diphenyl-4,5-dihydro-1H-imidazole—4-carboxylic acid (0.5g, 1.34 mmol) in dry methylene chloride (150 mL) at 0 °C, was added EDCI.HCL (0.31 g, 1.608 mmol), after five minutes DMAP (0.2 g, 1.6 mmol) and stirred for 20 minutes. Benzyl amine (0.17 g, 1.6 mmol) was then added dropwise. The reaction mixture was then stirred overnight at room temperature. The reaction mixtures was washed 2N HCl (2 x 50 mL), saturated sodium bicarbonate (2 x so mL),, and then with brine (50mL). The organic layer was dried over anhydrous sodium sulfate and evaporated under reduced pressure to afford the amide product as a white foam V-12: Yield: (0.5 g, 66.6%). 1H NMR (300MHz), CDC13 : 8 1.53 (s, 3H), 3.77 (d, J= 15.3, 1H), 3.91 (dd, J, = 14.7 Hz, J2= 5.1 Hz, 1H ), 4.21 (dd, J, = 15 Hz, J2= 6 Hz, 1H ), 4.35 (s, 1H), 4.60 (d, J=15.6 ,1H), 6.81-7.64 (m, 20 H).; l3C NMR (75MHz), CDC13: 27.71, 42.77, 48.45, 72.18, 76.57, 126.89, 127.46, 127.7, 127.79, 128.03, 128.26, 128.37, 128.4, 128.73, 128.81, 130.39, 130.46, 136.99, 137.17, 138.28, 165.02, 172.26.; EIMS: m/z= 459.3 (M+) Synthesis of Z-Benzylidene-6-oxo-5-phenyl-3—(toluene-4-sulfonyl)- [l,3]oxazinane-4-carboxylic acid ethyl ester from (Toluene-4-sulfonylimino)- acetic acid ethyl ester v.6 using l-Benzyl-4-methyl-2,5—dipheny1-4,5-dihydro- 1H-imidazole-4-carboxylic acid (l-phenyl—ethylyamide (+)-V-amide-l 257 To a well stirred solution of phenylacetyl chloride (0.3 mL, 2.1 mmol) in dry DCM (4 mL) at -78°C added proton sponge (0.4 gm, 2.1 mmol), after 5-10 min the color of the solution changes to pale yellow then added a solution of 1-benzyl- 4-methyl-2,5-dipheny1-4,5-dihydro-1 H-imidazole-4-carboxylic acid (1 -phenyl- ethyl)-amide (V-amide—l) (0.28 gm, 0.6 mmol or 0.14 gm, 0.3 mmol) in 3 mL of dry DCM. The reaction allowed to stir for 5—10 min on which the color changes to deep yellow then added a solution of (toluene-4-sulfonylimino)-acetic acid ethyl ester (0.25 gm, 1 mmol) in dry DCM (3mL). The reaction mixture allowed to stir for 36 hrs at —78 °C and then allowed to come to room temperature. The reaction mixture was passed through a silica gel bed to remove unreacted acid chloride and washed with ethyl acetate (20 mL). The solution was subjected to column chrotography using 1:3 ethyl acetate: hexanes to isolate pure 2-benzylidene-6- oxo-5-phenyl-3-(toluene-4-sulfonyl)-[1, 3]oxazinane-4-carboxylic acid ethyl ester (V -6a). Further elution with same solvent give mixture of 2-benzylidene-6-oxo-5- phenyl-3-(toluene-4-sulfonyl)-[1,3]oxazinane-4-carboxylic acid ethyl ester and B- lactam V-6b, and further pure B-lactam. o 0 Ph ”KAN 1 002 E1 3:0 11 0 O V-6a 2-Benzylidene-6-oxo-5-phenyl-3-(toluene-4-sulfonyI)—[1,3]oxazinane-4- carboxylic acid ethyl ester V-6a: Yield: 0.17 gm, 34.7%, 'H NMR (300MHz) : 6 0.98 (t, J = 7.5, 3H), 2.49 (s, 3H), 4.05 (q, J=7.5, 2H), 4.16 (d, J= 11.4, 1H), 258 4.50 (d, J =11.4, 2H), 6.45 (s, 1H), 7.10-7.13 (m, 2H), 7.27-7.41 (m, 8H), 7.60 (dd, # 0.6 and 7.5, 2H), 7.80 (d, J= 8.4, 2H); ”‘0 NMR (300MHz) : 8 13.65, 21.74, 49.72, 61.05, 62.15, 115.34, 127.77, 128.56, 128.67, 128.92, 129.02, 129.34, 129.55, 129.86, 130.27, 130.53, 131.75, 134.12, 136.71, 145.50, 165.66, 168.26. IR (neat); 1794, 1749, 1670, 1369; MS; m/z=491.2 o OJIPh ”RAB: Ph :0 |\ 0° V-10a Synthesis of 2-Benzylidene—4, S-diphenyl—3(toluene-4-sulfonyl)-l1, 3]oxazinan-6-one V-10a. To a well stirred solution of phenylacetyl chloride (0.3 mL, 2.1 mmol) in dry DCM (4 mL) at -78°C added proton sponge (0.4 gm, 2.1 mmol), after 5-10 min the color of the solution changes to pale yellow then added a solution of 1- Benzyl-4-methyl-2,5-diphenyl-4,5-dihydro-1 H-imidazole-4-carboxylic acid (1 - phenyl-ethyl)-amide (0.28 gm, 0.6 mmol or 0.14 gm, 0.3 mmol) in 3 mL of dry DCM. The reaction allowed to stir for 5-10 min on which the color changes to deep yellow then added a solution of N-benzylidine-(Toluene-4-sulfonyl)amine (0.25 gm, 1 mmol) in dry DCM (3mL). The reaction mixture allowed to stir for 36 hrs at -78 0C and then allowed to come to room temperature. The reaction mixture was passed through a silica gel bed to remove unreacted acid chloride and washed with ethyl acetate (20 mL). The solution was subjected to column chromatography using 1:4 ether : hexanes to isolate pure 2-Benzylidene-4,5- 259 diphenyl-3(toluene-4-su1fonyl)-[1,3]oxazinan-6-one V-10a. Further elution with same solvent give mixture of 2-Benzylidene-4, 5-diphenyl-3(toluene-4-sulfonyl)- [1, 3]oxazinan-6-one and B-lactam dl-(3R, 4R)-Diphenyl-1-(toluene-4-sulfonyl)- azetidin-2-one V-10b, and further pure B-lactam. (3R, 4S)-2-Benzylidene-4,5-diphenyl-3(toluene-4-sulfony1)—[1,3]oxazinan-6- one V-10a. Yield: 0.17 g, 35 %, 'H NMR (500MHz) : 6 2.47 (s, 3H), 3.95 (d, J= 11.5 Hz, 1H), 5.05 (d, J =11.0 Hz, 1H), 6.41 (s, 1H), 6.8496867 (dd, 2H, J 1 = 7 Hz, J2 = 1.5 Hz, 2H), 7.09-7.1 (dd, 2H, J 1 = 7 Hz, J2 = 1.5 Hz, 2H), 7.2-7.24 (m, 6H), 7.32-7.36 (t, 2H, J = 8.5 Hz, 3H), 7.38-7.41 (t, 2H, J = 8.5 Hz, 3H), 7.63- 7.64 (d, J =7.5 Hz, 2H), 7.72-7.73 (d, J =7.5 Hz, 2H); I3C NMR (125 MHz) : 6 21.84, 29.86, 55.3, 64.36, 115.46, 126.77, 128.07, 128.53, 128.71, 128.71, 128.82, 128.9, 129.72, 129.79, 130.23, 132.26, 132.29, 135.46, 138.53, 139. 63, 145.19, 166.06; IR (neat); 1738, 1666; EIMS; m/z=495.2. dI-(3R, 4R)-Diphenyl-l-(toluene-4-sulfonyI)-azetidin-2-one [V-10b]. Yield 0.037 g, 10 %, 'H NMR (300MHz) : 6 2.43 (s, 3H), 4.25 (t, J= 3.3 Hz, 1H), 4.96 (d, J =3 Hz, 1H), 7.03-7.3 (m, 15H), 7.67-7.7 (m, 2H); 13C NMR (75 MHz) : 8 21.68, 64.17, 65.78, 126.5, 127.23, 127.55, 128.07, 128.36, 128.6, 128.93, 129.05, 129.13, 129.66, 129.83, 132.79, 135.66, 135.93, 145.29, 165.37; IR (neat); 1795,; MS; m/z=377.9. 260 General procedure for diethyl zinc reduction: 0 OH /lL———> Ph H ph Method A: According to general procedure by Soai et a1, a 1M solution of diethyl zinc in hexane (2.2 mL, 2.2 mmol) was added to a solution of the chiral ligand (10 mol %) and the aldehydes (1mmol) in toluene (5 mL). The mixture was stirred under alkylation was complete under argon atmosphere in a sealed flask. The reaction mixture was diluted with dichloromethane (50 mL) and extracted with 10 % HCl solution. The organic layer was dried over anhydrous sodium sulfate, filtered and evaporated to obtain an oily residue. The residue was chromatographed with 20 % ether/hexanes to obtain the l-phenyl propanol. The enantiomeric excess was found by comparison to literature values. [Soai, K.; Yokayama, S.; Hayasaka, T. J. Org. Chem. 1991, 56, 4264.] Method B: Stoichiometn'c or catalytic Ti-isopropoxide was added was added to a solution of the chiral ligand (10 mol %) in toluene (5 mL) and stirred for 20 minutes followed by 1M solution of diethyl zinc in hexane (2.2 mL, 2.2 mmol) the aldehydes (1mmol). The mixture was stirred under alkylation was complete under argon atmosphere in a sealed flask. The reaction mixture was diluted with dichloromethane (50 mL) and extracted with 10 % HCl solution. The organic layer was dried over anhydrous sodium sulfate, filtered and evaporated to obtain an oily residue. The residue was chromatographed with 20 % ether/hexanes to obtain the 1-pheny1 propanol. The enantiomeric excess was found by comparison to literature values [Walsh P. J. Acc. Chem. Res., 36 (10), 739 -749, 2003.] 261 OH Ph lH NMR (300MHz), CDCl3: 8 0.89 (t, J = 7.5 Hz, 3H), 1.67-1.85 (m, 2H), 1.97 (broad singlet, 1H), 4.561 (t, J = 6.6 Hz, 1H), 7.23-7.35 (m, 5H); l3C NMR (75MHz), CDC13: 5 10.07, 31.75, 75.87, 125.92, 127.35, 128. 28, 144.53. Synthesis of l-Benzyl-4-methyl-2, 5-diphenyl-4, 5-dihydro-lH—imidazole-4- carboxylic acid l-phenyl-ethyl ester V-l-l4. A well—stirred suspension of 1-Benzyl-4-methy1-2,5-diphenyl-4,5-dihydro-1H- imidazole-4-carboxylic acid III-l-l ( 1.0g, 2.7 mmol) in dry methylene chloride (150mL) was made in a flame dried flask under nitrogen atmosphere and cooled in an ice-bath to 0 °C. To this mixture was added EDCI.HCl (0.57g, 2.9mmol), followed by DMAP (0.35 gm, 2.9 mmol) and stirred for 20 minutes. (R)-(+)-l- phenyl-ethanol (0.72g, 2 eq., 5.8 mmol) was added and mixture stirred overnight at room temperature. The reaction mixtures was washed, 2N HCl (2 x 50 mL), saturated sodium bicarbonate (2 x 50 mL), and then with brine (50mL). The organic layer dried over sodium sulfate and evaporated under reduced pressure. The crude product was purified by column silica-gel chromatography using 70 % ether / hexane mixture. 262 (211’. N <82...© NWO a? O .\“ (rm (-)v-1-14 P" (-)- (RRR) v-1-14. Yield: (0.34 g, 61 %). [(Ih): -118 °, c = 1.2, CHC13; 'H NMR (500MHz), coca: 8 1.236 (d, J = 6.5, 3H), 1.62 (s, 3H), 3.83 (d, J = 15.5 Hz, 1H), 4.34 (s, 1H), 4.70 (d, J=15.5 Hz,1H), 5.32 (q, J: 6.5, 1H), 6.94-7.16 (m, 4 H), 7.17-7.28 (m, 11H), 7.48-7.49 (t, J = 3.5 Hz, 3H) 7.76-7.77 (m, 2H).; I3C NMR (125MHz) coch: 21.8, 26.95, 48.88, 72.81, 73.27, 77.61, 125.87, 127.2, 127.57, 127.7, 127.77, 128.0, 128.27, 128.55, 128.64, 130.08, 131.12, 136.59, 136.64, 141.35, 165.92, 171.08.; EIMS: m/z = 474.1(M+). 3H7,“ (2) | S N S O O a“ - - - 08' (+) V1 14 Ph (+)- (SSR) v-1-14: (0.38 g, 66 %). [61],,= +113 °, c = 1.2, CHC13; 'H NMR (500MHz), CDC13: 8: 0.957 (d, J = 6.5, 3H), 1.61 (s, 3H), 3.77 (d, J = 15.5 Hz, 1H), 4.33 (s, 1H), 4.66 (d, J =15.5 Hz,1H), 5.33 (q, J= 6.5, 1H), 6.92-6.94 (m, 2 H), 7.17-7.29 (m, 13H), 7.47-7.48 (m, 3H) 7.73-7.75 (m, 2H).; l3c NMR (125MHz), CDC13: 21.44, 26.85, 48.83, 72.74, 73.57, 77.58, 126.0, 127.45, 263 127.54, 127.79, 127.9, 128.0, 128.14, 128.37, 128.51, 128.53, 128.62, 130.03, 131.15, 136.52, 137.06, 141.77, 165.94, 170.86.; EIMS: m/z = 474.2 (M+). Ph\' Ph\«‘Z.\OH lPh (-)V-1-17 [(-)(4R, 5R)-1-benzyl-4,5-dihydro-4-methyI-2,5-diphenyl-lH-imidazol-4-yl] methanol V-l-17: Lithium aluminum hydride (0.04 g, 1.06 mmol) was suspended in 100 mL of dry THF in a flame dried flask under nitrogen atmosphere. The suspension was cooled to 0 °C in an ice-bath. (-)«RRR ester V-l- 14 (0.34 g, 0.71 mmol) solution in dry THF (10 mL) was added dropwise. The mixture was stirred at 0 °C and monitored with TLC for complete disappearance of the ester spot. The reaction mixture was diluted with ethyl acetate (100 mL) and organic layer was extracted with 5 % HCl solution (3x, 100 mL). The organic layer was dried over anhydrous sodium sulfate, filtered and evaporated under vacuum. The residue was chromatographed with 20 % MeOH/ethyl acetate to afford the (-) RR alcohol V-l-l7 as colourless oil (0.2 g, 79 % yield) which solidified on standing under high vacuum. [a]D= -98 °, c = 1.0, CHC13; lH NMR (500MHz), CDC13: 5: 1.35 (s, 3H), 3.08 (d, J = 11.7 Hz, 1H), 3.17(d, J = 11.7 Hz, 1H), 3.58 (broad singlet, 1H), 3.89 (d, J = 15.6 Hz, 1H), 4.28 (s, 1H), 4.72 (d, J = 15.3 Hz, 1H), 6.83-6.86 (m, 2H), 7.2-7.72 (m, 13H). 264 MeO N 0/'\ Ph V-1 -1 5 4, S-dihydro-l-(4-methoxyphenyl)-4-methyl-2, S-diphenyl-lfl imidazole-4- carboxylic acid l-phenylethyl ester V-l-15. A well-stirred suspension of 1- Benzyl-4-methy1-2,5~diphenyl-4,5-dihydro-1H-imidazole-4-carboxylic acid III-l- 1 (1 .0g, 2.6 mmol) in dry methylene chloride (150mL) was made in a flame dried flask under nitrogen atmosphere and cooled in an ice-bath to 0 °C. To this mixture was added EDCI.HCl (0.6g, 3.1 mmol), followed by DMAP (0.38 gm, 3.1 mmol) and stirred for 20 minutes. (R)- (+)-1-phenyl-ethanol (0.72g, 2 eq., 58 mmol) was added and mixture stirred overnight at room temperature. The reaction mixtures was washed, 2N HCl (2 x 50 mL), saturated sodium bicarbonate (2 x 50 mL), and then with brine (50mL). The organic layer dried over sodium sulfate and evaporated under reduced pressure. The crude product was purified by column silica-gel chromatography using 50 % ether / hexane mixture. 266 OMe (+)-v-1-15 (8811): (0.2 g, 31%). [a]D= +138 °, c = 1.0, CHC13; 'H NMR (300MHz), (213013: 8: 1.2 (d, J = 6.3, 3H), 1.785 (s, 3H), 3.63 (s, 3H), 4.73 (s, 1H), 5.28 (q, J= 6.3, 1H), 6.56 (d, J = 9H2, 1H), 6.58-6.66 (m, 2H), 7.02-7.35 (m, 13H), 7.62 (d, J = 9 Hz, 2H); l3C NMR (125MHz), CDC13: 21.44, 26.85, 56.83, 72.74, 73.57, 77.58, 126.0,127.45, 127.54,127.79, 127.9, 128.0, 128.14, 128.37, 128.51, 128.53, 128.62, 130.03, 131.15, 136.52, 137.06, 141.77, 165.94, 170.86. OMe N Ph 8 Ph (36 t N (Ska OH [(+)(4S,SS)-4,5-dihydro-l-(4-methoxyphenylH-methyl-Zfi-diphenyl-1H- imidazol-4-yl]methanol V-l-18: Lithium aluminum hydride (0.03 g, 0.78 mmol) was suspended in 100 mL of dry THF in a flame dried flask under nitrogen atmosphere. The suspension was cooled to 0 °C in an ice-bath. (+)-SSR ester V-l- 15 (0.2 g, 0.52 mmol) solution in dry THF (10 mL) was added dropwise. The mixture was stirred at 0 °C and monitored with TLC for complete disappearance 267 of the ester spot. The reaction mixture was diluted with ethyl acetate (100 mL) and organic layer was extracted with 5 % HCl solution (3x, 100 mL). The organic layer was dried over anhydrous sodium sulfate, filtered and evaporated under vacuum. The residue was chromatographed with 10 % MeOH/ethyl acetate to afford the (+)-SS alcohol V-1-18 as colourless oil (0.130 g, 65 % yield). (+)-SS alcohol V-l-l8 [ct]D= +100 °, c = 1.0, CHC13; 1H NMR (500MHz), CDC13: 6: 1.51 (s, 3H), 2.97 (broad s, 1H), 3.14 (d, J = 11.1 Hz, 1H), 3.21(d, J = 11.7 Hz, 1H), 3.63 (s, 3H), 4.61 (s, 1H), 6.554 (d, J = 9 Hz, 1H), 6.65 (d, J = 9 Hz, 1H), 7.21-7.35 (m, 8H), 7.52 (d, J = 7.2 Hz, 2H), 13C NMR (125MHz), CDC13: 6 21.7, 64.19, 65.74, 65.8, 77.42, 126.52, 127.08, 127.25, 127.57, 127.75, 128.09, 128.21, 128.37, 128.62, 128.87, 128.95, 129.07, 129.15, 129.53, 129.68, 129.85, 130.48, 130.67, 132.81, 135.67, 135.95, 145.31, 165.39. Ph l-benzyl-4,5-dihydro-4-methyl-2, S-diphenyl-lH-imidazole-4-carboxylic acid l-phenylethyl ester V-11-16: A well-stirred suspension of 1-Benzyl-4-methyl- 2,5-diphenyl-4,5-dihydro-1H-imidazole-4-carboxylic acid (1.0g, 2.5 mmol) in dry methylene chloride (150mL) was made in a flame dried flask under nitrogen atmosphere and cooled in an ice-bath to 0 °C. To this mixture was added EDCI.HCL (0.57g, 3.0 mmol), followed by DMAP (0.36 gm, 3.0 mmol) and 268 stirred for 20 minutes. (R)-(+)-1-Phenyl-ethanol (0.67g, 2 eq., 5.5 mmol) was added and mixture stirred overnight at room temperature. The reaction mixtures was washed, 2N HCl (2 x 50 mL), saturated sodium bicarbonate (2 x 50 mL), and then with brine (50mL). The organic layer dried over sodium sulfate and evaporated under reduced pressure. The crude product was purified by column silica-gel chromatography using 60 % ether / hexane mixture. (+)- (SSR) V-11-16: (0.27 g, 46%). [a]D= +130°, c = 1.2, CHC13; lH NMR (300MHz), CDC13: 6: 1.17 (d, J = 6.3, 3H), 1.55 (s, 3H), 3.78 (d, J = 15.9 Hz, 1H), 3.83 (s, 3H), 4.27 (s, 1H), 4.72 (d, J =15.3 Hz, 1H), 5.23 (q, J= 6.6, 1H), 6.91-6.98 (m, 6H), 7.15-7.28 (m, 12H), 7.68 (d, J = 8.6 Hz, 2H); I3C NMR (125MHz), CDC13 21.6, 26.74, 48.63, 55.02, 72.46, 72.74, 77.27, 113.66, 122.9, 125.59, 126.94, 127.3, 127.37, 127. 46, 127.7, 127.74, 128.0, 128.3, 129.88, 136.41, 136.46, 141.0, 160.74, 165.46, 171.0. Ph Meo W (2) \ N N BllPh .nI|\ 1; OH [(-)(4R,5R)-1-benzyl-4,5-dihydro-2-(4-methoxyphenylH-methyl-S-phenyl- 1H-imidazol-4-yl]methanol V-ll-19: Lithium aluminum hydride (0.03 g, 0.78 mmol) was suspended in 100 mL of dry THF in a flame dried flask under nitrogen 269 atmosphere. The suspension was cooled to O 0C in an ice-bath. (-)-RRRester V- 11-16 (0.28 g, 0.55 mmol) solution in dry THF (10 mL) was added dropwise. The mixture was stirred at 0 °C and monitored with TLC for complete disappearance of the ester spot. The reaction mixture was diluted with ethyl acetate (100 mL) and organic layer was extracted with 5 % HCl solution (3x, 100 mL). The organic layer was dried over anhydrous sodium sulfate, filtered and evaporated under vacuum. The residue was chromatographed with 10 % MeOH/ethyl acetate to afford the (-)-RR alcohol V-ll-19 as colourless oil (0.144 g, 68% yield). [a]D= +114 0, c = 1.0, CHC13; lH NMR (300MHz), CDC13: 6: 1.317 (s, 3H), 2.98 (d, J = 12.0 Hz, 1H), 3.11(d, J = 11.4 Hz, 1H), 3.42 (broad s, 1H), 3.86 (s, 3H), 3.87 (d, J = 15.0 Hz, 1H), 4.23 (s, 1H), 4.78 (d, J = 15.6 Hz, 1H), 6.83-6.86 (m, 2H), 7.0 (d, J = 8.7 Hz, 2H), 7.22-7.65 (m, 9H), 7.67 (d, J = 9 Hz); l3C NMR (75MHz), CDC13: 6 25.7, 48.57, 66.84, 71.4, 71.7, 127.37, 127.58, 127.65, 128.18, 128.26, 128.4, 128.46, 130.08, 135.65, 135.76, 164.79. 270 E. 10. 11. 12. 13. References Green, D. R. Cell 1998, 94, 695-698. Hengartner, M. 0. Nature 2000, 407, 770-776. Fesik, S. W. Cell 2000, 103, 273-282. Thompson, C. B. Science 1995, 26 7, 1456-1462. Hanahan, D.; Weinberg, R. A. Cell 2000, 100, 57-70. Johnstone, R. W.; Ruefli, A. A.; Lowe, S. W. Cell 2002, 108, 153-164. Lowe, S. W.; Lin, A. W. Carcinogenesis 2000, 21 , 485-495. (a) Pommier, Y.; Sordet, 0.; Antony, S.; Hayward, R. L.; Kohn, K. W. Oncogene 2004, 23, 2934-2949. (b) Mayo, M. W.; Baldwin, A. S. Biochim. Biophys. Acta 2000, 14 70, M55-62. (c) Wang, C. Y.; Mayo, M. W.; Komeluk, R. G.; Goeddel, D. V.; Baldwin, A. S., Jr. Science 1998, 281, 1680-1683. ((1) Wang, C. Y.; Mayo, M. W.; Baldwin, A. S., Jr. Science 1996, 274, 784-787. Makin, G.; Hickman, J. A. Cell Tissue Res. 2000, 301, 143-152. (a)Nesterenko, V.; Putt, K. S.; Hergenrother, P. J. J. Am. Chem. Soc. 2003, 125, 14672-14673.(b) Nicholson, D.W. Nature 2000, 407, 810-816.(c) Hayakawa, Y.; Kim, J. W.; Adachi, H.; Shin-ya, K.; Fujita, K. -I.; Seto, H. J. Am. Chem. Soc. 1998, 120, 3524-3525. (a)Mitsiades, N.; Mitsiades, C. S.; Richardson, P. G.; Poulaki, V.; Tai, Y. T.; Chauhan, D.; Fanourakis, G.; Gu, X.; Bailey, C.; Joseph, M.; Libermann, T. A.; Schlossman, R.; Munshi, N. C.; Hideshima, T.; Anderson, K. C. Blood 2003 101, 2377-2380. (b) Weaver Kyle, D.; Yeyeodu, S.; James, C., Jr.; Baldwin, A. S., Jr.; Ewend, M. G. Journal of Neuro-Oncology 2003, 61, 187-196. (c) Chen, L.; Agrawal, S.; Zhou, W.; Zhang, R.; Chen, J. Proc. Natl . Acad. Sci. USA 1998, 95, 195-200. Pommier, Y.; Pourquier, P.; Fan, Y.; Strumberg, D. Biochim. Biophys. Acta 1998, 1400, 83-105. Wall, M. B.; Wani, M. C. Camptothecin and Analogues. In Cancer Chemotherapeutic Agents; Foye, W. 0., Ed.; (Washington: ACS) 1995, 293-310. Wall, M. E. Med. Res. Rev. 1998, 18, 299-314. 271 15. l6. 17. 18. 19. 20. 21. 22. 23. 24. 25. 26. 27. Macdonald, T. L.; Labroli, M. A.; Tepe, J. J. DNA topoisomerase II inhibitors. In Comprehensive natural products chemistry; Vol. 7, Kool, E. T., Ed.; Amsterdam: Elsevier-North Holland, 1999, 593-614. Pourquier, P.; Pommier, Y. Adv Cancer Res 2001, 80, 189-216. (a) Huang, T. T.; Wuerzberger—Davis, S. M.; Seufzer, B. J .; Shumway, S. D.; Kurama, T.; Boothman, D. A.; Miyamoto, S. J. Biol. Chem. 2000, 275, 9501-9509. (b) Piret, B.; Piette, J. Nucleic Acids Res. 1996, 24, 4242- 4248. (c) Xu, Y.; Villalona-Calero, M. A. Annals of Oncology 2002, 13, 1841-1851. (d) Sodhi, A.; Singh, R. A. K. Immunology Letters 1998, 63, 9-17. (e) Sénchez-Pérez, I.; Benitah, S. A.; Martinez- Gomariz, M.; Lacal, J. C.; Perona, R. Mol. Biol. Cell 2002, 13, 2933-2945. Cusack, J. C., Jr.; Liu, R.; Baldwin, A. S. Drug Resist. 1999 Update 2, 271- 273. (a) Carson, J. P.; Zhang, N., Frampton, G. M.; Gerry, N. P.; Lenburg, M. B.; Christman, M. F. Cancer Res. 2004, 64, 2096-2104. (b) Zhou, Y.; Gwadry, F. G.; Reinhold, W. C.; Miller, L. D.; Smith, L. H.; Scherf, U.; Lin, B. T.; Kohn, K. W.; Pommier, Y.; Weinstein, J. N. Cancer Res. 2002, 62, 1688-1695. Boland, M. P. Biochem. Soc. Trans. 2001, 29, 674-678. Cusack, J. C., Jr.; Liu, R.; Baldwin, A. S., Jr. Cancer Res. 2000, 60, 2323- 2330. Guttridge, D. C.; Albanese, C.; Reuther, J. Y.; Pestell, R. 6.; Baldwin, A. S., Jr. Mol. Cell Biol. 1999, 19, 5785-5799. Baeuerle, P. A.; Henkel, T. Annu. Rev. Immunol. 2000, 12, 141-179. Baldwin, A. S., Jr. Annu. Rev. Immunol. 1996, 14, 649-683. Perkins, N. D. Int. J. Biochem. Cell Biol. 1997, 29, 1433-1448. Webster, G. A.; Perkins, N. D. Mol. Cell Biol. 1999, 19, 3485-3495. (a) Das, K. C.; White, C. W. J. Biol. Chem. 1997, 272, 14914-14920.(b) Baldwin, A. S. J. Clin. Invest. 2001, 107, 241-246. (c) Hideshima, T.; Chauhan, D.; Richardson, P.; Mitsiades, C.; Mitsiades, N.; Hayashi, T.; Munshi, N.; Dang, L.; Castro, A.; Palombella, V.; Adams, J.; Anderson, K. C. J. Biol. Chem. 2002, 277, 16639-16647.(d) Bottero, V.; Busuttil, V.; Loubat, A.; Magne, N.; F ischel, J. L.; Milano, G.; Peyron, J. F. Cancer 272 28. 29. 30. 31. 32. 33. 34. Res. 2001, 61, 7785-7791. (e) Um, J. H.; Kang, C. D.; Lee, B. G.; Kim, D. W.; Chung, B. S.; Kim, S. H. Oncogene 2001, 20, 6048-6056. (1) Weldon, C. B.; Burow, M. B.; Rolfe, K. W.; Clayton, J. L.; Jaffe, B. M.; Beckman, B. S. Surgery 2001, 130, 143-150. (g) Arlt, A.; Vomdamm, J.; Breitenbroich, M.; Folsch, U. R.; Kalthoff, H.; Schmidt, W. E.; Schafer, H. Oncogene 2001, 20, 859-868. (h) Liu, S.; Yu, Y.; Zhang, M.; Wang, W.; Cao, X. J. Immunol. 2001, 166, 5407-5415. (i) Kim, J. Y.; Lee, S.; Hwangbo, B.; Lee, C. T.; Kim, Y. W.; Han, S. K.; Shim, Y. S.; Yoo, C. G. Biochem. Biophys. Res. Commun. 2000, 273, 140-146. Lin, Y. C.; Brown, K.; Siebenlist, U. Proc. Natl. Acad. Sci. U S A 1995, 92, 552-556. (a) Karin, M.; Lin, A. Not. Immunol. 2002, 3, 221-227. (b)Zhong, H.; SuYang, H.; Erdjument-Bromage, H.; Tempst, P.; Ghosh, S. Cell 1997, 89, 413-424. (c) Sizemore, N.; Leung, S.; Stark, G. R. Mol. Cell Biol. 1999, 19, 4798-4805. ((1) Madrid, L. V.; Wang, C. Y.; Guttridge, D. C.; Schottelius, A. J.; Baldwin, A. S., Jr.; Mayo, M. W. Mol. Cell Biol. 2000, 20, 1626-1638. Wu, M. X.; Ao, Z.; Prasad, K. V.; Wu, R.; Schlossman, S. F. Science 1988, 281, 998-1001. (a) Jones, D. R.; Broad, R. M.; Madrid, L. V.; Baldwin, A. S., Jr.; Mayo, M. W. Ann. T horac Surg. 2000, 70, 930-936; discussion 936-937. (b) Cusack, J. C., Jr.; Liu, R.; Houston, M.; Abendroth, K.; Elliott, P. J.; Adams, J.; Baldwin, A. S., Jr. Cancer Res. 2001, 61, 3535-3540. Habraken, Y.; Piret, B.; Piette, J. Biochemical Pharmacol. 2001, 62, 603- 616. Epinat, J. C. ; Gilmore, T. D. Oncogene 1999, 18, 6896-6909. (a) Cuzzocrea, S., Chatterjee, P. K.; Mazzon, E.; Dugo, L.; Serraino, 1.; Britti, D.; Mazzullo, G.; Caputi, A. P.; Thiemermann, C. Br. J. Pharmacol , 2002, 135, 496-510. (b) Roshak, A.; Jackson, J. R.; Chabot-Fletcher, M.;Marshall, L. A. J. Pharmacol. Exp. Ther. 1997, 283, 955-961. (c) Breton, J. J .; Chabot-Fletcher, M. C. J. Pharmacol. Exp. Ther. 1997, 282, 459-466. ((1) Sharma, V.; Tepe, J. J. Bioorg. Med. Chem. Lett. 2004, 14, 4319-4321. (e) Meijer, L.; Thunnissen, A. M.; White, A. W.; Gamier, M.; Nikolic, M.; Tsai, L. H.; Walter, J .; Cleverley, K. E.; Salinas, P. C.; Wu, Y. Z.; Biemat, J .; Mandelkow, E. M.; Kim, S. H.; Pettit, G. R. Chem. Biol. 2000, 7, 51-63. (1) Wan, Y.; Hur, W.; Cho, C. Y.; Liu, Y.; Adrian, F. J.; Lozach, 0.; Bach, S.; Mayer, T.; Fabbro, D.; Meijer, L.; Gray, N. S. Chem. Biol. 2004, 11, 247-259. (g) Kishore, N.; Sommers, C.; Mathialagan, S.; Guzova, J.; Yao, M.; Hauser, S.; Huynh, K.; Bonar, S.; 273 35. 36. 37. 38. 39. 40. Mielke, C.; Albee, L.; Weier, R.; Graneto, M.; Hanau, C.; Perry, T.; Tripp, C. S. J. Biol. Chem. 2003, 278, 32861-32871. (11) Grisharn, M. B., Palombella, V. J.; Elliott, P. J.; Conner, E. M.; Brand, S.; Wong, H. L.; Pien, C.; Mazzola, L. M.; Destree, A.; Parent, L.; Adams, J. Methods Enzymol. 1999, 300, 345-363. (i) Nemeth, Z. H.; Wong, H. R.; Odoms, K.; Deitch, E. A.; Szabo, C.; Vizi, E. S.; Hasko, G. Mol. Pharmacol. 2004, 65, 342-349. (j) Fenteany, G.; Standaert, R. F.; Soongyu Choi, W. 8.; Corey, E. J.; Schreiber, S. L. Science 1995, 268, 726. (k) Teicher, B. A.; Ara, G.; Herbst, R.; Palombella, V. J.; Adams, J. Clin. Cancer Res. 1999, 5, 2638-2645. (1) Kwok, B. H.; Koh, B.; Ndubuisi, M. 1.; Elofsson, M.; Crews, C. M. Chem. Biol. 2001, 8, 759-766. Hehner, S. P.; Hofmann, T. G.; Droge, W.; Schmitz, M. L. J. Immunol. 1999, 163, 5617-5623. Shah, S. A.; Potter, M. W.; McDade, T. P.; Ricciardi, R.; Perugini, R. A.; Elliott, P. J.; Adams, J.; Callery, M. P. J. Cell. Biochem. 2001, 82, 110- 122. Ma, M. H.; Yang, H. H.; Parker, K.; Manyak, S.; Friedman, J. M.; Altamirano, C.; Wu, Z. Q.; Borad, M. J.; Frantzen, M.; Roussos, B.; Neeser, J .; Mikail, A.; Adams, J .; Sjak-Shie, N.; Vescio, R. A.; Berenson, J. R. Clin. Cancer Res. 2003, 9, 1136-1144. (a) Laufer, S. A.; Striegel, H. - G.; Wagner, G. K. J. Med. Chem. 2002 45(21), 4695-4705. (b) Mc Kenna, J. M.; Halley, F .; Souness, J. E.; McLay, I. M.; Pickett, S. D.; Collis, A. J.; Page, K. M.; Ahmed, I. 2002, 45, 2173- 2184. (c) Collis, A. J .; Foster, M. L.; Halley, F.; Maslen, C.; McLay, I. M.; Page, K. M.; Redford, E. J .; Souness, J. E.; Wilsher, N. E. Bioorg. Med. Chem. Lett. 2001, 11, 693-696. (a) Chang, L. A.; Sidler, K. L.; Cascieri, M. A.; de Laszlo, S.; Koch, G.; Li, B.; MacCoss, M.; Mantlo, N.; O'keefe, A.; Pang, M.; Rolando, A.; Hagmann, W. K.; Bioorg. Med. Chem. Lett. 2001, 11, 2549. (b) Liverton, N. J.; Butcher, J. W.; Claiborne, C. F.; Claremon, D. A.; Libby, B. B.; Nguyen, K. T.; Pitzenberger, S. M.; Selnick, H. G.; Smith, G. R.; Tebben, A.; Vacca, J. P.; Varga, S. L.; Agarwal, L.; Dancheck, K.; Forsyth, A. J.; Fletcher, D. S.; Frantz, B.; Hanlon, W. A.; Harper, C. F.; Hofsess, S. J.; Kostura, M.; Lin, J.; Luell, S.; O'Neill, E. A.; Orevillo, C. J.; Pang, M.; Parsons, J.; Rolando, A.; Sahly, Y.; Visco, D. M.; O'Keefe, S. J. J. Med. Chem. 1999, 42(12), 2180-2190. (a) Slee, D. H.; Romano, S. J.; Yu, J.; Nguyen, T. U.; John, J. K.; Raheja, N. K.; Axe, F. U.; Jones, T. K.; Ripka, W. C. J. Med. Chem. 2001, 44, 2094. (b) Boyle, E. M., Jr.; Kovacich, J. C.; Canty, T. G., Jr.; Morgan, E. 274 41. 42. 43. 44. 45. 46. 47. 48. 49. 50. 51. N.; Chi, E.; Verrier, E. D.; Pohlman, T. H. Circulation, 1998, 98, 11282- 11288. (a) Tasdemir, D.; Mallon, R.; Greenstein, M.; Feldberg, L. R.; Kim, S. C.; Collins, K.; Wojciechowicz, D.; Mangalindan, G. C.; Concepcion, G. P.; Harper, M. K.; Ireland, C. M. J Med Chem 2002, 45, 529-532. (b) Sharma, V.; Lansdell. T.; Jin, G. Y.; Tepe, J. J. J Med Chem 2004, 47(14), 3700- 3703. (c) Shanna, V.; Tepe, J. J. Bioorg. Med. Chem. Lett. 2004, 14 (16), 4319-4321. Sharma, V.; Lansdell, T.; Peddibhotla, S.; Tepe, J. J. Chem. Biol. 2004 (in print) Lansiaux, A.; Facompre, M.; Wattez, N.; Hildebrand, M. P.; Bal, C.; Demarquay, D.; Lavergne, 0.; Bigg, D. C.; Bailly, C. Mol. Pharmacol. 2001, 60, 450-461. (a) Nicholson, D. W.; Thomberry, N. A. Science 2003, 299, 214-215. (b) Nicholson, D. W.; Thomberry, N. A. Trends Biochem. Sci. 1997, 22, 299- 306. (c) Thomberry, N. A. Chem. Biol. 1998, 5, R97-103. Johnson, N.; Ng, T. T. C.; Parkin, J. M. Leukemia Research 1997, 21 , 961- 972. (a) Delhase, M.; Hayakawa, M.; Chen, Y.; Karin, M. Science 1999, 284, 309-313. (b) Karin, M. Oncogene 1999, 18, 6867-6874. Thanos, D.; Maniatis, T. Cell 1995, 80, 529-532. (a)Tozawa, K.;Okamoto, T.; Hayashi, Y.; Sasaki, S.; Kawai, N.; Kohri, K. Urological Res. 2002, 30, 53-58. (b) Schow, S. R.; Joly, A. Cell Immunol. 1997, 175, 199-202. Tepe, J. J .; Shanna, V.; Lansdell, T.; Peddibhotla, S.; Michaels, S.; Brown, D. M. 2003, Abstracts of Papers, 225th ACS National Meeting, New Orleans, LA, United States, March 23-27, 2003. Liu, M.; Sibi, M. P. Tetrahedron 2002, 7991-8035. (a) Sivakumar, A. V.; Babu, G. S.; Bhat, S. V. Tetrahedron: Asymmetry 2001, 1095-1099. (b) Waisvisz, J .M.; van der Hoeven, M.G.;te Nijenhuis, B. J. Am. Chem. Soc. 1957, 4524-4527. (c) Helms, G. L.; Moore, R. E.; Niemczura, W. P.; Patterson, G. M. L.; Tomer, K.B.; Gross, M.L.; J. Org. Chem. 1988, 1298-1307. (d) Hechi, S.M. Acc. Chem. Res. 1986, 383-391. 275 52. 53. 54. 55. 56. 57. 58. 59. 60. 61. 62 (a) Salzmann, T. N.; Ratcliffe, R. W.; Christensen, B. G.; Boufi‘ard, F.A. J. Am. Chem. Soc. 1980, 6161-6163. (b) Okano, K.; Izawa, T.; Ohno, M. Tetrhedron Lett. 1983, 217. (c) Huang, H.; Iwasawa, N.; Mukaiyama, T. Chem. Lett. 984, 1465. (d) Kim, S.;Chang, S. B.; Lee, P. H. Tetrahedron Lett. 1987, 2735. (e) Kim, S.; Lee, P. H.; Lee, T. A. J. Chem. Soc., Chem Commun. 1988, 1242. (1) Kunieda, T.; Nagamatsu, T; Higuchi, T.; Hirobe, M. Tetrahedron Lett. 1988, 2203. (g) Tanner, D.; Somfai, P. Tetrahedron 1988, 613. (a) J .D. Winker Tetrahedron 48 (1992), p. 6953-6969. (b) Nicolaou, K.C.; Dai, W. M.; Guy, R. K. Angew. Chem, Int. Ed Engl. 1994, 33, 15-17. (a) Mokotofi‘, M.; Bagaglio, J. F .; Parikh, B. S. J. Med Chem. 1975, 18, 354-358. (b) Hansson, T. G.; Kihlberg, J. O. J. Org. Chem. 1986, 51, 4490-4492.(c) Wagner, R.; Tilley, J. W. J. Org. Chem. 1990, 55, 6289- 6291. (d) Wagner, R.; Tilley, J. W.; Lovey, K. Synthesis 1990, 785-786. Shimamoto, K.; Shigeri, Y.; Yasuda-Kamatani, Y.; Lebrun, B.; Yumoto, N.; Nakajima, T. Bioorg. Med. Chem. Lett. 2000, 10, 2407-2410. (a) von Riniker, B.; Schwyzer, R. Helv. Chim. Acta 1964, 4 7, 2357. (b) Ondetti, M.; Engel, S. L. J. Med. Chem. 1975, 18, 761. (c) Stachowiak, K.; Khosla, M. C.; Plucinska, K.; Khairallah, P. A.; Bumpus, F. M. J. Med. Chem. 1979, 22, 1128 (d) Hintermann, T.; Seebach, D. Chimia 1997, 50, 244. Sibi, M. P.; Liu, M. Tetrahedron 2002, 58, 7991-8035. (a) Enantioselective Synthesis of ,B-Amino Acids; Juaristi, E., Ed.; Wiley- VCH: New York, 1997. (b) Rzasa, R. M.; Shea, H. A.; Romo, D. J. Am. Chem. Soc. 1998, 120, 591-592. (c) Cole, D. C. Tetrahedron 1994, 50, 9517-9582 (d) Kobayashi, S.; Nagayama, S. J. Am. Chem. Soc. 1997, 119, 10049-10053 (e) Ferraris, D.; Young, B.; Dudding, T.; Lectka, T. J. Am. Chem. Soc. 1998, 120, 4548-4549 (1) Sibi, M. P.; Shay, J. J.; Liu, M.; Jasperse, C. P. J. Am. Chem. Soc. 1998, 120, 6615-6616. (g) Liu, M.; Sibi, M. P. Tetrahedron 2002, 58, 7991-8035. Davis, F. A.; Reddy, G. V.; Liang, C. H. Tetrahedron Lett. 1997, 38, 5139- 5142. Myers, J. K.; Jacobsen, E. N. J. Am. Chem. Soc. 1999, 121, 8959-8960. Horstmann, T. B.; Guerin, D. J .; Miller, S. J. Angew. Chem, Int. Ed 2000, 39, 3635-3638. Guerin, D. J.; Miller, S. J. J. Am. Chem. Soc. 2002, 124, 2134-2136. 276 63. 64. 65. 66. 67. 68. 69. 70. 71. 72. 73. 74. 75. 76. 77. 78. Boger, D. L.; Lee, R. J .; Bounaud, P.-Y.; Meier, P. J. Org. Chem. 2000, 65, 6770-6772. Orr, R. K.; Calter, M. A. Tetrahedron 2003, 3545-3565. F u, G.C. Acc. Chem. Res. 2000, 33, 412 and references there in. Hodous, B. L.; Fu, G. C. J. Am. Chem. Soc. 2002, 124, 1578-1579. (a) Taggi, A.E.; Hafez, A. M.; Wack, H.; Young, B.; Ferraris, D; Lectka, T. J. Am. Chem. Soc. 2002, 124, 6626-6635.(b) Taggi, A. E.; Wack, H.; Hafez, A. M.; France, S.; Lectka, T. Org. Lett. 2002, 4, 627-629. (c) Dudding, T.; Hafez, A. M.; Taggi, A. E.; Wagerle, T. R.; Lectka, T. Org. Lett. 2002, 4, 387-390. ((1) Hafez, A. M.; Taggi, A. E.; Dudding, T.; Lectka, T. J. Am. Chem. Soc. 2001, 123, 10853-10859. (e) Taggi, A. E.; Hafez, A. M.; Wack, H.; Young, B.; Drury, W. J., 111; Lectka, T. J. Am. Chem. Soc. 2000, 122, 7831-7832 and references therein. Peddibhotla, S.; Jayakumar, S.; Tepe, J. J. Org. Lett. 2002, 4, 3533-3535. Peddibhotla, S.; Tepe, J. J. Synthesis 2003, 9, 1433-1440. Brochu, M. P.; Brown, S. P.; MacMillan, D. W. C. J. Am. Chem. Soc. 2004, 4108-4109. Tschaen, D. M.; Turos, B.; Weinreb, S. M. J. Org. Chem. 1984, 49, 5058 — 5064. Jayakumar, S.; Peddibhotla, S.; Tepe, J. J. unpublished results. Satsumabayashi, S. J. Org. Chem. 1976, 41, 156-157. Martin, J. C.; Brannock, K. C.; Burpitt, R. D.; Gott, P. G.; Hoyle, Jr. V. A. J. Org. Chem. 1971, 36, 2211-2215. Davis, B. A.; Masanobu, M.; Angew. Chem. Int. Ed Engl. 1968, 7, 826- 827. Martin, J. C.; Hoyle, Jr. V. A.; Brannock, K. C.; Kent, C. Tet. Lett. 1965, 40, 3589-3594. Schaumann, B.; Mrotzek, H. Chem. Ber. 1978, 111, 661-672. Furukawa, M.; Okawara, T.; Noguchi, H.; Terawaki, Y. Heterocycles 1977, 6, 1323-1328. 277 79. 80. 81. 82. 83. 84. 85. 86. 87. 88. 89. 90. 91. 92. Hafez, A. M.; Dudding, T.; Wagerle, T. R.; Shah, M. H.; Taggi, A. E.; Lectka, T. J. Org. Chem. 2003, 5819-5825. Kobayashi, S.; Matsubara, R.; Kitagawa, H. Org. Lett. 2002, 143-145. Podlech, J .; Seebach, D.; Angew. Chem. Int. Ed Engl. 1995, 471-472. Kobayashi, S.; Tsukamoto, Y.; Saegusa, T. Macromolecules 1990, 2609- 2612. Walsh P. J. Acc. Chem. Res., 36 (10), 739 -749, 2003. Casey, M.; Smyth, M. P. Synlett. 2003, 102. (a) Inoguchi, K.; Sakuraba, S.; Achiwa, K. Synlett. 1992, 169. (b) Mashima, K.; Kusano, K. H.; Sato, N.; Matsumara, Y.; Nozaki, K.; Kumobayashi, H.; Sayo, N.; Hori, Y.; Ishizaki, T.; Akutagawa, S.; Takaya, H. J. Org. Chem. 1994, 59, 3064. (c) Clyne, D. S.; Mennet- Bouvier, Y. C.; Nomura, N.; Rajan Babu, T. V. J. Org. Chem. 1999, 64, 7601. (d) Gambs, C.; Consiglio, G.; Togni, A. Helv. Chim. Acta 2001, 84, 3105. Hermann, W. A.; Goossen, L. J .; Speigler, M. Organometallics 1998, 17, 1172. Waltman, A. W.; Grubbs, R. H. Organometallics 2004, 23(13), 3105 - 3107. (a) Scholl, M.; Ding, S.; Lee, C. W.; Grubbs, R. H. Org. Lett. 1999, I, 953-956. (b) Morgan, J. P.; Grubbs, R. H. Org. Lett. 2000, 2, 3153-3155. (a) Viciu, M. H.; Kissling, R. M.; Stevens, E. D.; Nolan, S. P. Org. Lett. 2002, 4, 2229-2231. (b) Viciu, M. S.; Germaneau, R. F.; Nolan, 8. P. Org. Lett. 2002, 4, 4053-4056. (c) Herrmann, W. A. Angew. Chem, Int. Ed 2002, 41, 1290-1309. (d) McGuinness, D. S.; Cavell, K. J. Organometallics 1999, 18, 1596-1605. (e) McGuinness, D. S.; Cavell, K. J. Organometallics 2000, 19, 741-748. Ma, Y.; Song, C.; Jiang, W.; Wu, Q.; Wang, Y.; Liu, X.; Andrus, M. B. Org. Lett. 2003, 5, 3317-3319. Seiders, T. J.; Ward, D. W.; Grubbs, R. H. Org. Lett. 2001, 3, 3225-3228. Van Veldhuizen, J. J .; Garber, S. B.; Kingsbury, J. S.; Hoveyda, A. H. J. Am. Chem. Soc. 2002, 124, 4954-4955. 278 93. (a) Younkin, T. R.; Connor, E. F.; Henderson, J. 1.; Friedrich, S. K.; Grubbs, R. H.; Bansleben, D. A. Science 2000, 287, 460-462. (b) Connor, E. F.; Younkin, T. R.; Henderson, J. 1.; Waltman, A. W.; Grubbs, R. H. Chem. Commun. 2003, 2272-22. (c) Matsui, S.; Mitani, M.; Saito, J .; Tohi, Y.; Makio, H.; Matsukawa, N.; Takagi, Y.; Kazutaka, T.; Nitabaru, M.; Nakano, T.; Tankaka, H.; Kashiwa, N.; Fujita, T. J. Am. Chem. Soc. 2001, 123, 6847-6856 ((1) Tian, J.; Hustad, P. D.; Coates, G. W. J. Am. Chem. Soc. 2001, 123, 5134-5135.(e) Gibson, V. C.; Mastroianni, S.; Newton, C.; Redshaw, C.; Solan, G. A.; White, A. J. P.; Williams, D. J. Dalton 2000, 1969-1971. 94. Menges, F .; Neuburger,M.; Pfaltz, A. Org. Lett., 2002, 4 (26), 4713 -4716. 95. Helmchen, G.; Pfaltz, A. Acc. Chem. Res. 2000, 33, 336-345. 96. (a) Schnider, P.; Koch, G.; Prét6t, R.; Wang, G.; Bohnen, F. M.; Kruger, C.; Pfaltz, A. Chem. Eur. J. 1997, 3, 887-892. (b) Lightfoot, A.; Schnider, P.; Pfaltz, A. Angew. Chem, Int. Ed. 1998, 37, 2897-2899. (c)Blackmond, D. G.; Lightfoot, A.; Pfaltz, A.; Rosner, T.; Schnider, P.; Zimmennann, N. Chirality 2000, 12, 442-449. 97. Botteghi, C.; Schionato, A.; Chelucci, G.; Brunner, H.; Kiirzinger, A. Oberman, U.; J. Organomet. Chem. 1989, 370, 17. 98. Morimoto, T.; Tachibana, K.; Achiwa, K. Synlett 1997, 783. 99. Davenport, A. J .; Davies, D. L.; Fawcett, J .; Russell, D. R.; J. Chem. Soc., Perkin Trans. 1 2001, 1500. 100. Dupont, J.; Ebeling, G.; Delgado, M. R.; Consorti, C. S.; Burrow, R.; Farrar, D. H.; Lough, A. J. Inorg. Chem. Commun. 2001, 4, 471. 101. Bastero, A.; Ruiz, A.; Claver, C.; Castillon, S. Eur. J. Inorg. Chem. 2001, 3009. 102. Bussaca, C. A. US Patent 6316620, 2001. 103. Oguni, N.; Omi, T. Tetrahedron Lett. 1984, 25, 2823-2824. 104. Soai, K.; Niwa, S. Chem. Rev. 1992, 92, 833-856. 105. Pu, L.; Yu, H.-B. Chem. Rev. 2001, 101, 757-824. 106. Kitamura, M.; Suga, S.; Oka, H.; Noyori, R. J. Am. Chem. Soc. 1998, 120, 9800-9809. 279 107. 108. 109. 110. 111. 112. 113. 114. Kitamura, M.; Okada, S.; Suga, S.; Noyori, R. J. Am. Chem. Soc. 1989, 111, 4028-4036. Noyori, R.; Kitamura, M. Angew. Chem, Int. Ed Engl. 1991, 30, 49-69. Chen, Y. K.; Costa, A. M.; Walsh, P. J. J. Am. Chem. Soc. 2001, 123, 5378-5379. Buono, F.; Walsh, P. J .; Blackmond, D. G. J. Am. Chem. Soc. 2002, 124, 13652-13653. Richmond, M. L.; Seto, C. T. J. Org. Chem. 2003; 68(19); 7505-7508. Boland, N. A.; Casey, M.; Hynes, S. J.; Matthews, J. W.; Smyth, M. P. J. Org. Chem. 2002; 67(11); 3919-3922. Peddibhotla, S.; Jayakumar, S.; Tepe, J. J. Org. Lett. 2002, 4, 3533-3535. Peddibhotla, S.; Tepe, J. J. Synthesis 2003, 9, 1433-1440. 280 APPENDIX 281 EFFICIENT TWO-STEP SYNTHESIS OF METHYLPHYTYLBENZO QUINONES: PRECURSOR INTERMEDIATES IN THE BIOSYNTI-IESIS OF VITAMIN E A. Introduction to Tocopherols. Tocopherols are a class of lipid-soluble antioxidants synthesized in higher plants and other oxygenic photosynthetic organisms such as cyanobacteria.I The four tocopherols synthesized (01, B, 6, and y-tocopherol) differ only in the number and position of methyl groups on the chroman head group. Tocopherols are important in limiting lipid peroxidation in a variety of organisms and are an essential component of mammalian diets as vitamin E (2R,4'R,8'R-[01]-tocopherol).2' 3 The production of tocopherols for animal nutrition and various industrial purposes has sparked renewed interest in the synthesis and enzymology of the tocopherol biosynthetic enzymes.2’ 4'6 Although the tocopherol pathway has been studied for many years,7 it is only recently that pathway enzymes have been cloned, providing new opportunities to study the substrate specificity and kinetics of the reactions. 5' 6' 8' 9 B. Biosynthesis of a-tocopherol. The pathway leading to the synthesis of til-tocopherol is shown in Figure VI-l. The committed intermediate in the tocopherol biosynthetic pathway is 2-methyl- 6-phytyl-1,4-benzoquinone (MPBQ), the product of the condensation of the aromatic compound homogentisate (HGA) and the 20-carbon isoprenoid derived compound, phytyl pyrophosphate.9’ '0 MPBQ methyltransferase catalyzes the first 282 methylation reaction in tocopherol synthesis and the methylation of the same ring position of a related compound in plastoquinone synthesis. 1" '2 11 0” PP. + co; ,t — o o J 1'10sz 0\ ”/0\ 1.;/OW” OH ' ' Homogentisate Phytyl pyrophosphate SAM SAM SAH V > W H v OH 3 SAH OH 2-methyl-6-phytyl-1 .4-benzoquinol \ OH cyclisatlon HO H o H 3 3 17 11 2.3-dimethyl-6-phytyl-1 .4-benzoquinol y-tocopherol SAUH Ho > O H 3 tat-tocopherol Figure VI-l. Biosynthesis of a-tocopherol. Access to a variety of methylated phytylbenzoquinone precursors and intermediates is required in order to elucidate the regulation and the activity of MPBQ/MSBQ methyltransferase. 11 Previously reported methods provide some of these substrates in relatively low yields and as multiple isomers. '2'” The development of a short and efficient synthetic method applicable to generating a wide range of these substrates has therefore become an integral part of elucidation of the biosynthetic pathway. We developed an efficient two-step synthesis of 283 methylphytylbenzoquinones (MPBQs) to study the kinetics and substrate specificity of MPBQ methyltransferase. The earlier reported chemical synthesis of MPBQ involved oxidation of commercially available methylated phenols to methyl quinones followed by coupling of the phytyl side chain in the presence of BF3'OE12 resulting in a mixture of positional isomers. '0 Oxidation of the resulting quinols with AgzO provided a mixture of isomers, which were separated by repeated thin-layer chromatography. Unfortunately, due to the limited availability of the required isophytol side chain, the generation of wide mixtures of isomers, and corresponding low yielding reactions, this procedure was quite limited as a routine method for the large scale preparation of the many different tocopherol precursors. We discovered an alternative preparation of MPBQ derivatives from the naturally available tocopherols to provide more efficient access to these biosynthetic precursors. C. Synthesis of required methylphytlbenzoquinone. Oxidative cleavage of a chroman ring with cerium sulfate affords the quinone alcohol. '5 The well established FeCl; oxidation as described by Cohen has also successfully been used in the conversion of (1.-tocopherol to a- tocopheroquinones.l6 Cerium sulfate oxidation of 6-tocopherol resulted in a mixture of products from which the quinone alcohol VI-l was readily isolated in 30% yield at multigram scale. The quinone alcohol VI-l was subjected to 284 dehydration with Burgess reagent under argon atmosphere to afford a mixture of methylphytqulrinones v1-2 — VI-6 (Scheme v1-1)”:"3 HO 06(30412 H H2804(eonc) 3 H20. MeOH ' 2 hours. r.t. 30 % Vl-1 EtaN 04590 NYC 0 Burgess reagent + *7 \ THF. Argon. 2h, rt 0 42 % “'2 : H / 3 Vl-3 natural substrate V14 M: ...MH 3 VI-6 Scheme VI-l. Synthesis of methylphytylbenzoquinone. The positional and geometric isomers VI-2 - VI-6 were isolated by flash chromatography, followed by two rounds of HPLC purification using a silica column with hexane/isopropyl ether (992:8) as the eluant. Compounds VI-2 - VI- 6 were isolated in a relative ratio of 1:2:1.1:1.5:2.2. The positional and geometric isomers from the dehydration will be further used as competitive inhibitors for 285 evaluating the reaction mechanism and kinetic parameters of the MPBQ methyltransferase.l l D. Experimental Section Synthesis of VI-2 — VI-6: The quinone alcohol VI-l (0.4 g, 9 mmol) was weighed into a flame dried flask under argon atmosphere and dry THF (50 mL) was added. To this solution was added Burgess reagent (0.4 g, 1.8 mmol) and the mixture stirred at room temperature ovemight. The reaction mixture was then diluted with ether (200 mL) and extracted twice with water and the organic layer was dried over anhydrous sodium sulfate and the ether layer was evaporated to obtain a yellow residue. The residue was flash chromatographed on silica-gel (5% etherzhexane) to obtain the methylphytylbenzoquinone as a mixture of five isomers (0.16 g, 42%) They were further separated by HPLC. Spectral data: V1- 3 1H NMR (500 MHz, acetone-d6) 6 6.6 ( 1H, td, J.=1.5 Hz, J2=4.5 Hz), 6.46 (1H, td, J1=1.5 Hz, J2=3.5 Hz), 5.22 (1H, qt, Jl=1.5 Hz, J2=7.5 Hz), 3.14 (2H, d, J=7.5 Hz), 2.06—2.07 (3H, m), 2.02 (2H, d, J=1.5 Hz), 1.66 (3H, s), 1.0—1.6 (18H, m), 0.85—0.9 (12H, m); 13C NMR (125 MHz, acetone-d6) 6187.61, 148.56, 146.07, 139.55, 133.01, 132.04, 118.87, 39.85, 39.41, 37.43, 37.42, 37.31, 36.56, 32.81, 32.66, 28.82, 28.67, 27.99, 27.48, 25.25, 24.8, 24.43, 22.29, 22.2, 19.4, 15.36, 15.15; IR (neat): 1670 cm“', 1600 cm". MS (EI): calculated for C27H4402 [M]+ 400.64, found [M]+ 400.33. Purification of MPBQ and its isomers: The mixture of isomeric methylphytylbenzoquinones was injected on a ReliaSil Silica, 5 pm, 250><4.6 mm 286 normal phase column (Column Engineering, Ontario, CA) with a guard column containing the same matrix. Chromatography was performed at 30°C with a mobile phase of 992:8 (v/v) hexanezisopropyl ether flowing at 1.0 mL/min. For large-scale purification, a preparative column of the same matrix (Column Engineering, Ontario, CA) was used. The collected fractions were dried down under vacuum before a small portion was injected onto the analytical column to monitor the purity. The natural substrate VI-3 was eluted as the second HPLC peak. Enzyme Assay: Assays of MPBQ methyltransferase activity were carried out in a 100 11L reaction containing 10 uL of solubilized E. coli extract (50 and 75 pg protein) expressing the MPBQ methyltransferase of Synechocystis PCC6803, " 50 mM Tris pH 8.0, 10 mM DTT, 80 11M [l4C-methyl]S-adenosy1methionine (56 mCi/mmol), 40 11M unlabeled S-adenosylmethioine, and 100 11M each of the individual MPBQ compounds VI-2 — VI-6. After incubation at 30°C for 2 h, the reactions were stopped by the addition of 200 11L of 0.9% (w:v) NaCl, extracted with 450 uL of 2:1 (vzv) methanol:chlorofonn, centrifuged, and the organic phase collected and dried under a stream of nitrogen gas. The resulting residue was resuspended in hexane and fractionated on 250 1160A KF6 silica gel TLC plates (Whatman, Clifton, NJ, USA) developed with 3:7 (vzv) diethyletherzpetroleum ether. The TLC plates were exposed on a Molecular Dynamics low energy phosphor screen (Amersham) to determine the incorporation of MC into the products. 287 Interestingly, MPBQ methyltransferase was found to utilize all five MPBQ isomers VI-2 — VI-6 however the activity with the genuine substrate VI-3 (Figure VI-2) results in specific activity that is 10—100 times higher than those of its isomers. The method described in this report has also been successfully applied to large-scale preparation of phytquuinone precursors of (1.-tocopherol (data not shown) and is applicable to a wide range of methylphytylbenzoquinone precursors. 288 4o _=52> 2:22: LQLQCL 30 I 20 10 o m-- o s!’~%\ e-eseu-suumuse-ceueueueweaum ‘ Efficiency relative to genuine substrate 8 i l l VI-2 Vl-3 VI-4 VI-5 Vl-6 HPLC Fractions Figure VI-2. TOP (left): HPLC trace of VI-2 — VI-6.TOP(right): Efficacy of isomers VI-2 — VI-6 compared to natural MPBQ as substrates monitored on TLC using l4C-adenosyl methionine. BOTTOM: Efficacy of isomers VI-2 — VI-6 compared to natural MPBQ as substrates for methyltransferase activity. The percent efficacy of the isomers was based on the average of three independent assays. 289 E. References 1. 2. 10. ll. 12. 13. 14. 15. 16. 17. 18. Kamal-Eldin, A.; Appelqvist, L. A. Lipids 1996, 31, 671—701 . Fryer, M. J. Plant Cell Environ. 1992, 15, 381—392. Topinka, J .; Binkova, B.; Sram, R. J.; Erin. A. N. Mutat. Res. 1989, 225, 131—136. Weber, P.; Bendich, A.; Machlin, L. J. Nutrition 1997, 13, 450—460. Grusak, M. A.; DellaPenna,D.Annu. Rev. Physiol. Plant Mol. Biol. 1999, 50, 133—161. DellaPenna, D. Science 1999, 285, 375—379. Trelfall, D. R.; Whistance, G. R. Aspects of Terpenoid Chemistry and Biochemistry, Academic Press, London 1971. Garcia, 1.; Rodgers, M.; Lenne, C.; Rolland, A.; Sailland A.; Matringe, M. Biochem. J. 1997, 325, 761—769. Shintani, D.; DellaPenna, D. Science 1998, 282, 2098—2100. Soll, J .; Schultz, G. Phytochemistry 1980, 19, 215-218. Shintani, D. K.; Cheng, Z.; DellaPenna, D. FEBS Lett. 2002, 511,1—5. Chenevert, R.; Courchesne, G. Tetrahedron Lett. 2002, 43,7971—7973. Soll, J. Methods Enzymol. 1987, 248, 383-387. Henry, A.; Powls, R.; Pennock, J. F. Biochem. J. 1987, 242, 367-373. Cohen, N.; Lopresti R. J .; Saucy, G. J. Am. Chem. Soc. 1979, 101, 6710— 6716. Cohen, N.; Lopresti, R. J.; Neukom, C. J. Org. Chem. 1981, 46, 2445-— 2450. Burgess, E.M.; Penton, H. R. J. ; Taylor, E. A. J. Org. Chem. 1973, 38, 26—31. Peddibhotla, S.; Cheng, Z.; Dellapena, D.; Tepe, J. J. Tetrahedron Lett. 2003, 4 7, 237-239. 290 YYYYYYYYYY 1111111211111111111111 L 06 A 1111 l 1‘11