V $5}? , 1...an “W15? :fifié I. . N — . é; . ,gmuvvum. m. 3. v pg? .\ 1:. I 0! 5 :3. ’v ‘25! _h ;. A.-. a 4.1. «is... was. umma 5:33.033“ L. . 91.3.51 ‘ 15.;xszlnvthfivw‘vdw‘3 .«mflaflmwv ‘ 3... ’Plitltvgr. n. wk. .. . h . .z. 1 . .313 gm: 1.3%.»)! brié its. 2...}... s: K. 3.: 153.2. ’15.. 1.1.3? 1 h I harms, 2 7.000 This is to certify that the dissertation entitled PREPARATION OF RARE SUGARS 8: ADVANCED DERIVATIVES FROM COMPLEX CARBOHYDRATES 8: CARBOHYDRATE POLYMERS presented by Changyou Yuan has been accepted towards fulfillment of the requirements for the Pd.D. : degree In Chemistry Extowk @th ., Major Rrofes jfi’or 3 Signature OV/li/O Date MSU is an Affinnative Action/Equal Opportunity Institution ”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 :JUN 1. 9 200'? 2/05 p:/ClRC/DateDue.indd-p.1 PREPARATION OF RARE SUGARS 8: ADVANCED DERIVATIVES FROM COMPLEX CARBOHYDRATES 8: CARBOHYDRATE POLYMERS By Changyou Yuan A DISSERTATION Submitted to Michigan State University in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY Department of Chemistry 2006 ABSTRACT PREPARATION OF RARE SUGARS & ADVANCED DERIVATIVES FROM COMPLEX CARBOHYDRATES & CARBOHYDRATE POLYMERS By Changyou Yuan Carbohydrates have the potential as the ultimate raw materials for highly functionalized, optically pure chemicals which were extensively used in pharmaceutical, agrichemical and biotech industry. However, these high potentials of carbohydrates remain nearly untouched due to the complex structure and ofien redundant hydroxyl groups of carbohydrates. The aim of this work is to develop strategies to overcome these difficulties and transform complex carbohydrates to advanced derivatives. A protocol for the transformation of glycosides to anhydroalditols was established using Me-D-glucoside as the model compound (Chapter 2). Transformation of the free hydroxyl groups to ally] ether followed by reductive cleavage of the glycosidic bond using triethylsilane gave the protected 1,5-anhydroglucitol. Cleavage of the allyl ether bond was achieved by employing PdClz/CuClz catalytic system. This Protection- Reduction-Deprotection protocol was successfully used in the transformation of complex carbohydrates, such as cellulose, starch and levan, to the corresponding anhydroalditols. The anhydroalditol, 1,5-anhydro-D-glucitol, was transformed to its 6-phosphate, and 2, 3- deoxy derivatives in Chapter 3. The carbohydrate derivated (S)-2-hydroxyl-butyrolactone and (S)-2-hydroxyl- tetrahydrofuran were transformed to their iodo derivatives in Chapter 4. By controlling the neighboring group effect, the mono and diiodo derivatives were selectively formed. Chapter 5 presents an efficient route to prepare cyclic nitrone from D-ribose. Optically pure iminosugar and isoxazolidines were prepared from the nitrone through stereoselective addition reactions and cycloaddition reactions. ACKNOWLEDEMENTS I would like to sincerely thank my advisor Dr. Rawle I. Hollingsworth for his precious advice, inspiring guidance and essential support throughout the whole period of my graduate work. I am grateful that he let me explore the carbohydrate chemistry world, full of challenges and opportunities. I am also grateful to my committee members, Dr. C. K. Chang, Dr. B. Borhan, and Dr. K. C. Hunt, for their valuable advice and help in my research. I would also like to thank the members of Dr. Hollingsworth’s group, for their company and help. Special thank goes to Xuezheng for his help in my thesis writing. I would also like to thank L. Lee, K. Johnson and Dr. D. Holmes from the NMR facility for their friendly help. Finally, I would like to thank my family members, my wife, my parents, my parents-in- law and my son for their support, understanding and the happy times we spend together. iv TABLE OF CONTENTS LIST OF TABLES ................................................................................ viii LIST OF FIGURES ............................................................................... vix Chapter 1. Literature Review: Bulky carbohydrates and their advanced derivatives 1.1. Introduction: Availability and structural features of commodity carbohydrates .................................................................................... 3 1.1.1. Polysaccharides: cellulose, starch, chitin and inulin ............................ 4 1.1.2. Oligosaccharides: cyclodextrins, maltodextrins ................................ 8 1.1.3. Monosaccharides and disaccharides: sucrose, lactose, maltose, fructose, xylose ................................................................................ 9 1.2. Progress in use of carbohydrates as general raw materials for organic synthesis ......................................................................................... 14 1.2.1. General difficulties in the transformation of carbohydrates to organic synthons ............................................................................. 17 1.2.2. Successes in the preparation of general synthons from carbohydrates. . .18 1.3. Some medicinally important synthesis targets from carbohydrates and carbohydrate derived synthons ........................................................ 21 1.3.1. From 3 or 4 carbon synthons .................................................... 22 1.3.2. Iminosugar derivatives .......................................................... 24 1.3.3. F uran/pyran derivatives .......................................................... 26 1.4. Conclusion ...................................................................................... 28 1.5. Reference ..................................................................................... 29 Chapter 2. Transformation of Commodity Carbohydrates to High Value Chemical Intermediates by Reductive Cleavage 2.1. Introduction .................................................................................... 36 2.1.1. Availability and structural features of commodity carbohydrates. ............ 36 2.1.2. Organosilane reduction ............................................................. 38 2.2. Results ........................................................................................... 40 2.2.1. Establishing the protocol for triethylsilane cleavage of the glycosidic linkage ................................................................................. 40 2.2.2. Practical and economic Palladium chloride catalyzed de-allylation of 1,5- anhydro-2,3,4,6-tetra-O-allyl-D-glucitol ......................................... 42 2.2.3. Application of the Allylation-Reduction-De-allylation protocol to other mono, disaccharides and complex carbohydrates ............................... 43 2.3. Discussion ...................................................................................... 49 2.3.1. Establishing the protocol for triethylsilane cleavage of the glycosidic linkage ................................................................................. 49 2.3.2. Palladium chloride catalyzed de-allylation of l,5-anhydro-2,3,4,6-tetra—O- allyl-D-glucitol ....................................................................... 56 2.3.3. Application of the Allylation—Reduction-De-allylation protocol to other mono, disaccharides and complex carbohydrates ................................ 60 2.3.4. Preparation of 1,5-anhydro-D-glucitol, 2,5-anhydro-D-glucitol and 2,5- anhydro-D-mannitol from sucrose ................................................. 64 2.4. Conclusion ..................................................................................... 68 2.5. Experimental ................................................................................... 70 2.6. Reference ....................................................................................... 82 Chapter 3. Advanced Derivatives of 1, 5-Anhydro-D-Glucitol 3.1. Introduction .................................................................................... 88 3.1.1. Glycolysis and 1,5~anhydro-D-glucitol-6-phosphate ........................... 88 3.1.2. Deoxy anhydroalditols: application in construction of biologically important molecules and chiral catalysts ......................................... 89 3.2. Preparation of 1,5-anhydro-D-glucitol-6-phosphate ................................... 93 3.3. Preparation of Deoxysugars from 1, 5 -anhydro-D-glucitol .......................... 94 3.3.1. Formation of 2,3-anhydro derivatives of 1,5-anhydro-D-glucitol. . . . . . . . ....94 3.3.2. Preparation of deoxyanhydrohexitols ............................................. 97 3.4. Conclusion ...................................................................................... 98 3.5. Experimental ................................................................................... 99 3.6. References .................................................................................... 107 Chapter 4. Iodo Derivatives of Advanced Carbohydrate Intermediates 4.1. Introduction .................................................................................. 1 1 1 4.2. Preparation of iodo derivatives from (S)-3-hydroxy-butyrolactone .............. 112 vi 4.3. Preparation of iodo derivatives by ring opening of (S)-3-hydroxyl- tetrahydrofuran with HI ................................................................... 116 4.4. Reactions of 1,5-anhydro-2,3,4,6-tetra-O-acetyl-D-glucitol with HI .............. 125 4.5. The application of iodide products in the preparation of advanced derivatives ..................................................................................... 127 4.6. Conclusion 129 4.7. Experimental ................................................................................. 130 4.8. References .................................................................................... 140 Chapter 5. Preparation of Ribose Derived Nitrone and its Application in the Preparation of Iminosugars 5.1. Introduction ................................................................................... 144 5.1.1. Iminosugars as glycosidases inhibitors ......................................... 144 5.1.2. Application of nitrones in the preparation of iminosugars and aza-C- glycosides ............................................................................ 145 5.2. An efficient route for the preparation of cyclic nitrone from ribose ............. 149 5.3. Steroeselective Addition of nitromethane to the chiral nitrone .................... 154 5.4. Cyclization reactions of the ribose derived nitrone with non-carbohydrate based alkenes .......................................................................................... 155 5.5. Cycloaddition reactions of the ribose derived nitrone with carbohydrate based alkenes .......................................................................................... 160 5.6. Conclusion .................................................................................... 164 5.7. Experimental ................................................................................. 165 5.8. References ..................................................................................... 178 Vii LIST OF TABLES Chapter 1. Table 1.1. Annual world production and prices of some commodity carbohydrates, carbohydrate derivatives and common solvents ................................................. 13 Chapter 2. Table 2.1. Reduction of methyl-2,3,4,6-tetra-O-acetyl-a-D-glucopyranoside ............. 41 Table 2.2. Deprotection of 2,3,4,6-tetra-O-allyl-1,5-anhydro-D-glucitol ................... 43 Table 2.3. Transformation of glycosides to anhydroalditols .................................. 68 Chapter 4. Table 4.1. Ring-opening reactions of (S)-3-hydroxy-butyrolactone with H1 in AcOH/AczO ......................................................................................... 113 Table 4.2. Ring-opening reactions of (S)-3-hydroxyl-butyrolactone with H1 in AcOH ................................................................................................ 114 Table 4.3. Reaction of (S)-3-hydroxy-butyrolactone with HI in acetic acid ............... 116 Table 4.4. Light and oxygen effects on the ring opening reaction of (S)-3-hydroxyl- tetrahydrofuran with HI ........................................................................... 1 19 Table 4.5. Reaction of 1,5-anhydro-2,3,4,6-tetra-O-acetyl-D-glucitol ..................... 126 Chapter 5. Table 5.1. Preparation of nitrone using tosylate as substrate ................................ 150 Table 5.2. Preparation of nitrone using mesylate as substrate ............................... 151 viii LIST OF FIGURES Chapter 1. Figure 1.1. Representative partial structures of complex carbohydrates. .................... 6 Figure 1.2. Representative partial structure of complex carbohydrate inulin. . . . . . . . . . .7 Figure 1.3. Structure of oc-cyclodextrin (a) and maltodextrins (b) ............................. 9 Figure 1.4. Structure of disaccharides (a) sucrose; (b) ot-lactose; (c) B-lactose ............ 10 Figure 1.5. Structure of (a) maltose; (b) fi'uctose; and (c) xylose ............................. 11 Figure 1.6. Sucrose derivatives of amines, uronic acids and crown ether analogues...... 14 Figure 1.7. Isomaltulose derivatives .............................................................. 15 Figure 1.8. Glucose derivatives glucosides, glucals, glycosyl bromides and enolone bromides .............................................................................................. 16 Figure 1.9. Application of carbohydrates in natural products ................................. 16 Figure 1.10. Structure of some important basic chemicals and D-glucose .................. 18 Figure 1.11. Transformation of starch to (S)—3-hydroxy-butyrolactone ..................... 19 Figure 1.12. Transformation of L-arabinose to (S)-3 -hydroxy-butyrolactone .............. 20 Figure 1.13. Preparation of 2,3-O-isopropylidene-D-glyceraldehyde from D-mannitol..20 Figure 1.14. Oxidation of 1,2-5,6-di-O-isopropylidene-D-mannitol with Sodium hypochlorite/Ruthenium chloride ................................................. 21 Figure 1.15. 2,3-O-isopropylidene-L-glyceraldehyde from L-ascorbic acid ............... 21 Figure 1.16. Preparation of Linezolid derivatives from (R)-glycidyl butyrate ............. 22 Figure 1.17. Preparation of (L)-carnitine derivatives ............................................ 23 ix Figure 1.18 Some of the medicinally important compounds derived from chiral 3-carbon synthons ............................................................................................... 24 Figure 1.19. Important iminosugars ............................................................. 25 Figure 1.20. Medicinally important furan and pyran derivatives ............................ 26 Figure 1.21. Preparation of isonucleoside from 1,4-anhydro-D-xylitol ..................... 27 Chapter 2. Figure 2.1. Repeating units in complex carbohydrates. (a) cellulose; (b) levan. . . . . . .37 Figure 2.2. Reduction of aldehydes using triethylsilane ....................................... 38 Figure 2.3. Reductive cleavage of polysaccharide by triethylsilane ......................... 39 Figure 2.4. Reaction of methyl 2,3,4,6-tetra-O-acetyl-or-D-glucopyranoside with Et3SiH ................................................................................................. 41 Figure 2.5. Reduction of methyl 2,3,4,6-tetra-O-allyl-a-D-glucopyranoside with Et3SiH ................................................................................................ 42 Figure 2.6. Deallylation of l,5-anhydro-2,3,4,6-tetra—O-allyl-D-glucitol with PdClz. ...42 Figure 2.7. Reductive cleavage of methyl 2,3,4,6-tetra-O-alIyl-D-mannopyranoside. . .44 Figure 2.8. Reductive cleavage of methyl 2,3,5-tri-O-allyl-D-ribofuranosides ............ 45 Figure 2.9. Reductive cleavage of Octa-O-allyl-sucrose ...................................... 46 Figure 2.10. Reductive cleavage of tri—O-allyl-cellulose ...................................... 47 Figure 2.11. Reductive cleavage of tri-O-allyl-starch .......................................... 48 Figure 2.12. Reductive cleavage of tri-O-allyl-levan .......................................... 49 Figure 2.13. Mechanism for reaction of methyl 2,3,4,6-tetra-O-acetyl-a-D- glucopyranoside with Et3SiH ...................................................................... 52 Figure 2.14. Mechanism for reaction of methyl 2,3,4,6-tetra-O-acetyl-oc-D- glucopyranoside with Et3SiH ...................................................................... 53 Figure 2.15. Mechanism for reaction of methyl 2,3,4,6-tetra-O-allyl-ot-D- glucopyranoside with Et3SiH ....................................................................... 56 Figure 2.16. Proposed mechanism for catalytic cleavage of allyl ether .................... 59 Figure 2.17. Mechanism for reduction of methyl a-D-glucoside and methyl a-D- mannoside ............................................................................................ 61 Figure 2.18. Relative acid-catalysed hydrolysis rates of glucoside, galactoside and guloside ............................................................................................... 61 Figure 2.19. Mechanism for reductive cleavage of Octa-O-allyl-sucrose ................... 65 Chapter 3. Figure 3.1. Anhydrosugars as substrate analogues in carbohydrate glycolysis ............. 86 Figure 3.2. Sialyl Lewis" analogues as potent E-selectin inhibitors .......................... 91 Figure 3.3. Anhydrohexitol Nucleosides ......................................................... 91 Figure 3.4. Deoxy anhydroalditols based chiral catalysts ..................................... 92 Figure 3.5. Preparation of 1,5-Anhydro-D-glucitol 6-phosphate ............................. 93 Figure 3.6. Preparation of 1,5-Anhydro-D-glucitol 6-phosphate using the direct phosphorylation method ............................................................................ 94 Figure 3.7. Preparation of 4,6-benzylidene-2,3-epoxide derivatives of 1,5-anhydro-D- glucitol ................................................................................................ 95 Figure 3.8. Mechanism for the formation of 2,3-anhydro derivatives under Mitsunobu conditions ............................................................................................ 96 Figure 3.9. Formation of 2,3-anhydro derivatives using NaH/Ist ........................ 97 xi Figure 3.10. LAH reduction of 2,3-anhydro derivatives to form deoxy anhydrohexitols ...................................................................................... 98 Chapter 4. Figure 4.1. Proposed reaction of(S)-3-hydroxy-butyrolactone with HI ................... 112 Figure 4.2. Neighboring acetyl group assisted ring Opening reaction of (S)-2-hydroxy-butyrolactone with HI ........................................................... 114 Figure 4.3. Formation of optically inactive 4-iodide butanoic acid ......................... 115 Figure 4.4. Reaction of (S)-3-hydroxy-butyrolactone with H1 in acetic acid at various temperatures ......................................................................................... 1 16 Figure 4.5. Possible iodo products from ring opening reaction of (S)-3-hydroxy- tetrahydrofuran with HI ........................................................................... 118 Figure 4.6. Ring opening reaction of (S)-3-hydroxy- tetrahydrofuran with H1 in water ................................................................................................. 118 Figure 4.7. Mechanism for iodine catalyzed ring opening iodination of (S)-3 -hydroxy- tetrahydrofuran .................................................................. 120 Figure 4.8. Proposed reaction of (S)-3-hydroxy1- tetrahydrofuran and H1 in Acetic acid ................................................................................................... 121 Figure 4.9. Reaction of (S)-3-hydroxy- tetrahydrofuran and H1 in Acetic acid .......... 121 Figure 4.10. Mechanism for reaction of (S)-3-hydroxy- tetrahydrofuran and H1 in acetic acid ................................................................................................... 122 Figure 4.11. Reaction of tetrahydrofuran and H1 in Acetic acid ............................ 123 Figure 4.12. Reaction of (S)-3-hydroxy-tetrahydrofiiran and H1 in trifluoroacetic acid ................................................................................................... 123 xii Figure 4.13. Reaction of (S)-3 -hydroxy-tetrahydrofi1ran and H1 in acetic acid/acetic anhydride ............................................................................................ 124 Figure 4.14. Reaction of (S)-3-hydroxy - tetrahydrofuran and HI under various conditions ........................................................................................... 1 25 Figure 4.15. Proposed reaction of H1 mediated ring opening reaction of 1,5-anhydro- 2,3,4,6-tetra-O-acetyl-D-glucitoI ................................................................. 126 Figure 4.16. Reaction of 1,5-anhydro-2,3,4,6-tetra-O-acetyl-D-glucitol .................. 126 Figure 4.17. Preparation of (S)-3-hydroxy-tetrahydrothiophene ........................... 127 Figure 4.18. Preparation of (S)-1-benzyl-3-hydroxy-pyrolidine ........................... 127 Figure 4.19. Preparation of (S)-diethyl-3-acetoxycyclopentane-1 ,1-dicarboxy1ate...... 128 Chapter 5. Figure 5.1. General structure of carbohydrate based acyclic and cyclic nitrones ........ 146 Figure 5.2. Carbohydrate based acyclic nitrones .............................................. 146 Figure 5.3. Strategies for the preparation of cyclic nitrones ................................. 147 Figure 5.4. Preparation of cyclic nitrone from L-xylose ..................................... 148 Figure 5.5. Preparation of nitrone using tosylate as substrate ............................... 150 Figure 5.6. Possible products from reaction of tosylate 10 with hydroxylamine under basic conditions ..................................................................................... 152 Figure 5.7. Preparation of nitrone using tosylate as substrate ............................... 153 Figure 5.8. Possible products from addition of nitromethane to nitrone ................... 154 Figure 5.9. Addition of nitromethane to nitrone .............................................. 155 Figure 5.10. Possible products from cyclization of nitrone with allyl alcohol ............ 157 Figure 5.11. Cycloaddition of nitrone with allyl alcohol .................................... 158 xiii Figure 5.12. Products from cycloaddition of nitrone with vinyl ethyl ether .............. 159 Figure 5.13. Reaction of nitrone with vinyl ethyl ether ...................................... 159 Figure 5.14. Preparation of carbohydrate derived alkenes ................................... 160 Figure 5.15. Cycloaddition of nitrone with carbohydrate derived alkene I ............... 162 Figure 5.16. Cycloaddition of nitrone with carbohydrate derived alkene II ............... 163 xiv Chapter 1 Literature Review: Commodity carbohydrates and their advanced derivatives Abstract Carbohydrates hold the promise as the general raw materials to substitute. fossil raw materials. They are renewable and available in large volume. However, the structural differences between carbohydrates and fossil based raw materials make the development of carbohydrate based chemistry challenging. At first, the structural features of carbohydrates available in large quantity are reviewed. These include polysaccharides, such as cyclodextrins and maltodextrins, monosaccharides and disaccharide, such as sucrose, lactose, maltose, fructose and xylose. In the last several decades, carbohydrates have been used extensively in the preparation of naturally occurring and medicinally important compounds. Recently, progresses have been made in the transformation of carbohydrates to general chiral 3 and 4 carbon synthons. These more general synthons have been used directly and more widely in the synthesis of medicinally important targets. 1.1 Introduction: Availability and structural features of commodity carbohydrates. From the beginning of the industrial revolution, more and more fossil resources- coal, oil, and natural gas, have been used to provide us energy. Fossil resources have also been used as raw materials to support our more and more luxury lives, from electricity and gasoline to bulk, intermediate and fine chemicals. One big disadvantage of fossil resources is that they are not renewable and we are near the peak of the fossil material production. For petroleum this will be around 2008, for natural gas around 2020 and for coal in a few decades'. With the depletion of the fossil raw materials, the development of a chemical industry based on renewable resources becomes inevitable and urgent" 2. Biomass had been used as the major raw materials before the industrial revolution It is expected that in the next few decades, or even in the next few years, the use of biomass as raw materials for chemical industry will become economically viable with the ever increasing prices of fossil materials, it is estimated that the oil price will reach $182 per barrel within the decade3. The widely available biomass is composed of carbohydrates, amino acids and lipids. Among them, the most important class is carbohydrates in term of volume production, which account for roughly 75% of the annually renewable biomass of about 200 billion tons. However, majority of the carbohydrate biomass decays and only a minor fraction (ca. 4 %) is used by human beings“. The majority of the annually renewable carbohydrate biomass is polysaccharides, these include cellulose, starch, chitin, inulin, xylan, etcs. Derived from these polysaccharides are some oligosaccharides, such as cyclodextrins and maltodextrins. Monosaccharides and disaccharides, such as sucrose, lactose, fructose, maltose and xylose, are also available in bulk. 1.1.1 Polysaccharides: cellulose, starch, chitin and inulin. Cellulose is the most abundant form of living terrestrial biomass, it is found in higher plants as the principle component of cell walls in the form of microfibrils (2-20 nm diameter and 100 - 40 000 nm long)6. It is a linear polymer of about 2,000-14,000 [3-(1-- >4)-D-glucopyranose units in 4C. conformation (Fig. 1.1 a). The usage of cellulose largely takes advantage of its ability to hold water. Uses include anticaking agents, emulsifiers, stabilizers, dispersing agents, thickeners, and gelling agents. Chitin, the second most abundant polysaccharide in nature, can be found in the covering layer of insects, crabs, and shrimps and in the cell walls of many fungi. Each year, at least 10 gigatons of chitin are synthesized and degraded in the biosphere. Chitin mainly consists of partially deacetylated aminosugar N-acetylglucosamine through [HI-)4)- linkage, the same as cellulose (Fig 1.1 d)7. The mostly deacetylated form of chitin is called chitosan. It is used for waste water clearing, for cosmetics and for medical and veterinary applications. Another abundant polysaccharide is starch; it is a major carbohydrate reserve in fruits, seeds, and tubers of plants. 2850 million tones of starch is produced annually by photosynthesis. Corn is the largest source of starch; the other important sources include wheat, potato, tapioca and rice. Starch exists in the form of amylose (normally 20-30%) and amylopectin (normally 70-80%). Both amylose and amylopectin consist of polymers of ot-D-glucose units in the 4C1 conformation (Fig 1.1 a & b). In amylose, these units are linked in the form of 0t -(1-->4)- linkage, with the ring oxygen atoms all on the same side (Fig 1.1 b), whereas in amylopectin about one residue in every twenty units is linked in the form of a-(l-->6)- linkage to form branch-points (Fig 1.1 c). The ratio of amylose to amylopectin and the ot-(1-->6)-branch-points in amylopectin depend on the source of the starch, for example, amylomaizes contain over 50% amylose whereas 'waxy' maize has almost none (~3%). Starch is an important plant product; it is used as the major food resources for human beings throughout the history. Even today, starch still provides 80 % of man’s daily calorie intake in areas such as the Far East and Africas. With the excess production of starch, the non-food usage of starch increased during the last century. Like cellulose, the majority usage of starch depends on its ability to hold water, such as thickener, water binder, emulsion stabilizer and gelling agent. OH OH 3) 0 HO 0 o "o o H wow:mlo 0“ o o 0 OH OH :1 OH b) 0110 HO HO H hid n O O I.—T—-l OH c) ‘33: o 0 HO OH OH OH 0 o \ 0 35cm OH HO OH o 0 0 0 HO OH o 0 on HO OH 0 o O HO H or? d) o=( o=< "30 ml?" gfifiom «3'16le A0 OH Figure 1.1 Representative partial structures of complex carbohydrates. (a) cellulose; (b) amylose; (c) amylopectin; (d) chitin. Inulin, or fructose oligosaccharides, is present as plant carbohydrate storageg. High concentration of inulin can be found in dandelion, wild yam, Jerusalem artichokes, and chicory. Industrially, chicory has been used for the extraction of inulin. O I O (95/ O OH \OH Figure 1.2 Representative partial structure of complex carbohydrate inulin Inulin is a polydisperse B-(2-->1) fructan. The fructose units are linked through B-(2-->1) linkage, a glucose molecule typically resides at the end of each fructose chain and is linked by an (It—(192) bond. The chain lengths of these fructans range from 2 to 60 units, with an average DP of ~10"). The unique B—(2-91) bonds in inulin make it cannot be digested in the upper gastrointestinal tract as a typical carbohydrate and are responsible for its reduced caloric value and dietary fiber effectsg. The uptake of inulin does not lead to a rise in serum glucose level or stimulate insulin secretion. Therefore, inulin has been used to replace fat or sugar to reduce the calories of foods, such as dairy products, baked foods, etc. 1.1.2 Oligosaccharides: cyclodextrins, maltodextrins Recently, some oligosaccharides, such as cyclodextrins, maltodextrins, have been produced on an industrial scale1 1. Cyclodextrins are a family of three cyclic oligosaccharides composed of a-l,4- linked glucogan units. The a, B, y, cyclodextrin has 6, 7, 8 glucopyranose units respectively. In the 1970, cyclodextrins are only available as a rare fine chemical. Now, cyclodextrins are produced from partial hydrolysis of starch using the enzymes cyclodextrin gluconotransferases by choosing the type of the microorganism used, or or B cyclodextrin can be selectively produced”. Many factories are producing cyclodextrins at over 1000 tons/year scale. The usage of cyclodextrins largely depends on their ability to form host- 3 guest complexes with hydrophobic molecules. They have been used for drug release,I for environmental protection, food industry, cosmetics and personal care items, etc”. Hiya .. is i? w sass/a Figure 1.3 Structure of ot-cyclodextrin (a) and maltodextrins (b). OH 3-20 Maltodextrins are also produced from partial hydrolysis of starch using acid or enzyme”‘ '6. Unlike cyclodextrins, maltodextrins are linear a-1,4- linked glucogan units, with the chain length from 3 to 20 and average chain length 7. Maltodextrins are moderately sweet polysaccharides and are used as food additive. They are also used as coating agents in pharmaceutical industries and as water soluble glues among other uses”. 1.1.3 Monosaccharides and disaccharides: sucrose, lactose, maltose, fructose, xylose Sucrose, the table sugar, can be obtained from supermarkets in crystalline form in very high purity (>99%). It is the world’s single most abundant naturally occurring organic compound. In 2004, 144 million tons of sucrose was produced in the world's. Sucrose is a major carbohydrate reserve and energy resource in plants. Industrially, sucrose is extracted from sugar cane (20 % by weight) and sugar beet (15 % by weight). The structure of sucrose is shown in Figure 1.4 a. It is a non-reducing disaccharide, composed of one D-glucose and one-D-fructose unit linked through their anomeric carbon atoms by a, B-l—>2-glycosidic bond. About 95 % of the sucrose produced is used in the food industry as a sweetener. 3) OH O CHon HO 0 HO HO H 0 ° cazou OH b) HO OH HO 0 0 HO 2.0 OH 6) HO 0" OH 0 0 HO $10 0“ OH OH Figure 1.4 Structure of disaccharides (a) sucrose; (b) or-lactose; (c) B-lactose Lactose, also called milk sugar, is a unique carbohydrate only found in mammalian milk”. Lactose is obtained from whey, the by-product in the production of cheese, quark and casein. It is a disaccharide composed of one glucose and one galactose unit through [3-(1-)4) linkage by aldehyde group of D-galactose. Lactose exists in or and B isomeric forms, with difference in configuration of the hydroxyl moiety at C-1 of the glucose unit. Lactose is largely used in the food industry; it is also used in the pharmaceutical industry, infant nutrition and fermentation. 10 Naturally, the disaccharide maltose (Fig. 1.5 a) is present in germinating grain. Industrially, maltose is produced from the hydrolysis of starch'z‘ 20. The two D-glucose units in maltose are linked through (l—*4)-(X linkage. The glucose unit at the reducing end has the B configuration. The major usage of maltose is as a food sweetner in its reduced form maltitol. 3) OH O HO OH HO OH 0 0HO OH OH b) C) OH O OH O HO HO HO OH OH OH HO Figure 1.5 Structure of (a) maltose; (b) fructose; and (c) xylose. Fructose (Fig. 1.5 b) is a monosaccharide which can be found in many foods such as honey, tree fruits, berries, and beets, sweet potatoes etc”. It is one of the three most important blood sugars along with glucose and galactose. Fructose has been consumed by human beings for thousands of years at the amount of 16-20 grams per day. With the westernization of the diets, that number has increased significantly to 85-100 grams per day, mainly coming from the additives to foods and drinks in the form Of high fructose corn syrup. About 16 billion pounds of high fructose corn syrup was consumed each year 11 in the United States. The increased uptake of fructose has caused problems in human health, such as Obesity, diabetes, etc.” 23 . Xylose (Fig. 1.5 c) is the most abundant pentose; it can be easily Obtained from wood, straw-derived xylan24. Xylose is used in dyeing and tanning and also for diabetic diets. Besides the polysaccharides, oligosaccharides, disaccharides and monosaccharides discussed above, some other carbohydrates, such as isomaltoluse, L-sorbose, and carbohydrate derivatives, such as D-sorbitol, D-xylitol, D-gluconic acid are also available in large quantities and with the prices comparable to the common fossil based solvents, such as methanol, toluene. Table 1.1 listed the annual production and price of some common sugars, sugar derivatives and common solvents. 12 Table 1.1 Annual world production and prices of some commodity carbohydrates, carbohydrate derivatives and common solvents. World production Price (metric ton / year) ($/kg) Carbohydratesa Starch 2,729,000,000 0.34 Sucrose 144,000,000 0.22 glucose 304,073 0.43 Lactose 181,031 0.58 D-Fructose 91,863 0.69 Carbohydrate D-Sorbitol 650,000 1 .80 derivatesb D-Xylitol 30,000 5.00 D-Gluconic acid 60,000 1.40 Solvent?3 Acetone 3,200,000 0.55 Methanol 25,000,000 0.15 Toluene 6,500,000 0.25 8 Edited based on data from United States Department Of Agriculture Foreign Agricultural Service, for the year 2005. http://www.fas.usda.gov b Values are for the year 2003. Adapted from Table 1 in Lichtenthaler, F. W. Carbohydrates as Raw Materials for the Chemical Industry. Green Chemistry Series No. 1. Tundo, P. Ed., 3rd Ed., 2004, pp.105-127. 13 1.2 Progress in utilization of carbohydrates as general raw materials for organic synthesis In the last several decades, progress have been made in the usage of carbohydrates as starting materials for organic synthesis and several reviews are available on this subject. The chemistry for the transformations performed at the terminal positions of sucrose (C- 1’ and/or C-6 and/or C-6’) is reviewed by Jaroszzs. This review focus on the preparation of sucrose derivatives, such as amines, uronic acids, crown ether analogues, from the selectively protected sucrose 2,2’,3,3’,4-Penta-0—benzyl sucrose. Some of those derivatives were shown in Figure 1.6. R1 OBOM o /’\ BnO O O BnO BOO O 08 n 0 R; O o \ 9 OBD 1: R, . OH,OTBOPs. R, . OH,NH, Awfl o 2: R, - OH,NH,, R, - CH,OTBOPS AcO ,' ‘ 3: R, I R,- CH,NH, 0‘“ I"o\“‘ '. OAc 4: R, . CHZOTBDPS, R, a COOH AcO ’0‘“: 5: R, =- COOH, R, .. CHzOTBDPS s: R, - R, s coon 7 Figure 1.6 Sucrose derivatives of amines, uronic acids and crown ether analogues Isomaltulose, a disaccharide produced as the precursor to the sweetener isomalt®, is available in large quantities and its chemistry has been reviewed by Lichtenthaler's. Under acidic conditions, dehydration of the fructose portion Of isomaltulose afforded 5- l4 (a-D-glucosyl)-Oxymethyl—furfural (a-GMF). From oc-GMF, a variety of compounds have been made (Figure 1.7) OH 0 01130 I H+ OH H 0 3° -——-> "0% OH 0 OH 0 OH H 9 a - GMF 8 Isomaltulose 10: R I CHICHNOZ 11: R =- -CH=CI-ICOOH OH 12: R 3 -CH3CH2 H 0 13: R . cu fie 011043 14: R- -COOH o R 15: R . -CH2NH2 Figure 1.7 Isomaltulose derivatives The application of D-glucose as a starting material in organic synthesis has also been reviewed“‘ 5‘ '8‘ 26. The commonly used intermediates in the transformation of glucose to its derivatives are glucosides, glucals, hydroxyl glucals, glycosyl bromides, 2- oxoglycosyl bromides, enolone bromides, etc. (Fig. 1.8). The transformations of these intermediates have been demonstrated”. 15 820 OAc 0” O A 0 Ac 0 \ Ac AcO 820 16 1 7 1 8 08: OH OH o HO A" 320 / OMe 032 19 20 21 Figure 1.8 Glucose derivatives glucosides, glucals, glycosyl bromides and enolone bromides Carbohydrates were also often used in natural product synthesis. In the preparation of natural products, clinically important compounds, complex oligosaccharides and carbohydrate-based peptidomimetics, carbohydrate starting materials were extensively used in Nicolaou’s group”. Some representative structures were shown in Fig. 1.9 0 3,11. /CHO "MO; Ho'fioflo": .u R = 0 OR . ‘5‘ 0 MO 3 OOOCH,¢HMe, me“ O "on “‘0 22 Leucomycin A, 23 CarbomycInB Figure 1.9 Application of carbohydrates in natural products Leucomycin A3 22 and carbomycin B 23 are two clinically important 16-membered ring macrolide antibiotics. They were prepared starting from D-glucose. l6 The carbohydrates used in the above discussed applications are usually very specific, that is, the carbohydrates used have to be highly modified based on each synthetic targets. For this reason, the application of carbohydrates in organic synthesis is still limited compared to their potential. It is expected that if the more general synthons, such as 3, 4, or 5 carbon compounds with one or two stereocenters could be derived from carbohydrates, the carbohydrate based chemistry will be strong enough to challenge the position of fossil oil based chemistry. Here, the progress in the preparation of general raw materials for organic synthesis is reviewed. 1.2.1 General difficulties in the transformation of carbohydrates to organic synthons The transition from fossil based production to biomass carbohydrates based production is hampered by several factors. Fossil raw materials based chemicals are still more affordable at present and the technology for this transformation is well developed. Structurally, carbohydrates are very different from the commonly used fossil based chemicals. The commonly used industrial-scale fossil based raw materials are lipophilic low molecular weight products and their derivatives with fiinctional groups, such as —OH, COOH, C=C, C=O added (Figure 1.10). Compared to these compounds, carbohydrates are hydrophilic, have much higher molecular weight, are overly functionalized with hydroxyl groups and lack the C=C, C=O functional groups which are advantageous for chemical transformations. In the last decade, intense efforts have been made for the development of chemistries and technologies to obtain general raw organic materials from carbohydrates. 17 M/ H H H n-Hexane Ethylene Butadiene 1 PrOpanol O O JLOH /U\ Acetic acid Acetone HO mm H OH OH D-glucoee Figure 1.10 Structure of some important basic chemicals and D-glucose 1.2.2 Successes in the preparation of general synthons from carbohydrates Recently, 4-linked aldohexoses, such as lactose, maltose starch, maltodextrins, have been used in the preparation Of 3,4-dihydroxybutyric acid and its lactone28'30. Under basic conditions, the reducing end of the aldohexoses 24 is isomerized to ketose 25, base catalyzed B-elimination of the 4-alkoxy substituent gave the a-diketone 27 (Figure 1.11). Oxidative cleavage Of the diketone formed the dihydroxy butyric acid 28 and glycolic acid 29, cyclization of the dihydroxy acid gave the lactone 30. The chiral center in the lactone obtained come from OS of the D-aldohexoses and thus has (S) configuration. Similarly, the (R)-isomer 36 has been obtained from pentose (Figure 1.12). 18 OH 3‘ O OH OH o O Ho OH OH 0 HO OH A OH H OH OH 0 ' 24 HO \ 0.. ,5 OH ‘3 O O OH H H O °" 1%., 0" H0 25 /‘ H O OH OH OH‘ 1‘” O ‘0 0% OH OH 0 am 0" 0.. OH 0 0” OH HO \ + \ OH O HO OH 26 O 27 "202’ OH. O O OH £50 <—— <1 + mr HO HO o OH 30 28 29 Figure 1.11 Transformation of starch to (S)-3-hydroxy-butyrolactone 19 OH 0 O OH X X "mow +° ° r () 1::::=======:===:. () ‘------I> ()- ______. K1 OH \ OH OH 0 OH \ O 31 32 33 O O HO OH. OH OH O OH ..____ ‘OOH ‘ . \“‘ HO“ "O OH o (\0 3° 35 34 Figure 1.12 Transformation of L-arabinose to (R)-3-hydroxy-butyrolactone One of the oldest yet still widely used methods for the production of chiral 3-carbon synthons from carbohydrate is through the oxidative cleavage of protected monosaccharide“. Oxidation of l,2-5,6-di-O-isopropylidene-D-mannitol 38 with periodate or lead tetra-acetate gave the protected D-glyceraldehyde 39. Although satisfactory yields were obtained on small scales, the toxicity and high cost of the reagents prevents the application Of this method on industrial scale. 0 : OH Acetone / H1» We |04 or Pb(OAC)4 O . HO ; ———————> O ; a 6 OH OH %——6 OH ’ l 37 38 39 Figure 1.13 Preparation of 2,3-O-isopropylidene-D-glyceraldehyde from D-mannitol Recently, a more benign method was developed for the oxidation step. Using sodium hypochlorite as the oxidation reagent and ruthenium chloride as the catalyst, 1,2-5,6-di- O-isopropylidene-D-mannitol 38 was oxidized to isopropylidene-D-glyceric acid 39.32 20 OH 9% 0 WV NaOCI I RuCI3 Mk 0 O . OH C) - =T ; %O . 38 39 Figure 1.14 Oxidation of 1,2-5,6-di-O-isopropylidene-D-mannitol with Sodium hypochlorite/Ruthenium chloride To Obtain the L-isomer of glyceraldehydes, L-ascorbic acid, which is widely available, was used”. Oxidation of the protected L-ascorbic acid 40 with hydrogen peroxide gave 3,4-isopropylidene-D-erythronic acid 41 which was further oxidized by NaOCl to afford 2,3-isopropylidene-L-glyceraldehyde 42. °>< OH I O 7 o o 0 H202 / CECO3 o/WOH NaOCI — 7 1° ° ' OH OH 40 41 42 Figure 1.15 2,3-O-isopropylidene-L-glyceraldehyde from L-ascorbic acid 1.3 Some medicinally important synthesis targets from carbohydrates and carbohydrate derived synthons. Carbohydrates and carbohydrate derived 3 or 4 carbon synthons have been used extensively in natural product synthesis, in the preparation of medicinally important 34-36 targets 1.3.1 From 3 or 4 carbon synthons 21 The development of new antibiotics is always urgent and challenging with the emergence of multi-drug resistant organism. Linezolid, marketed by Pharrnacia and UpJOhn, is the newest antibiotic available for the treatment of infections caused by vancomycin-resistant Enterococcus faecium, hospital acquired pneumonia and methicillin—resistant Staphylococcus aureus37‘ 38. Chiral 3-carbon synthons are widely used in the preparation of linezolid and its derivatives3943. a-Qeg’lkocnfl + izxkfl/V __a_. Rfljikiofl 43 R==-M:::>) -M:::fi-4 uo'-d<:::H-{<::>-4:_fif_\N-:ifgrbn 40.0.... U 0 Figure 1.16 Preparation of Linezolid derivatives from (R)-g1ycidyl butyrate. NHAc (a) n-BuLi/hexane; (b) 4-Nitrobenzenesulfonyl chloride/Et3N; (c) NH3/CH3OH; (d) AczO Figure 1.16 lists one recent example for the preparation of Linezolid and its derivatives from of 3-carbon synthon‘“. Deprotonation of the carbamate 43 with n-BuLi followed by 22 addition of (R)—glycidyl butyrate 44 give the (5R)-(hydroxy methyl)-2-oxazolidinone 45, which is converted to the final Linezolid and its derivatives 47 in two more simple steps. (L)-Carnitine is a small molecule which was identified more than 100 years ago”. It can be found in all animals, in bacteria and in some plants. It is present in all cells and body fluids of human being“. (L)-carnitine is involved in the transfer of fatty acids into mitochondria. Mitochondria are potentially attractive targets of DNA damaging agents” 5 '. In cancer cells, the amount of mitochondria and the expression of carnitine transporter increased which makes the mimics of (L)—camitine potential antitumor agent552'53. The carnitine derivatives can be easily prepared from the 4-carbon synthons as shown in Figure 1.17“. o rpopsom 9R WCOO‘B“ 3._b. NNWCOOtBu rpopso 48 49 on 0 cu _Ev_d. c'D/VCOOH R =23 N/—/ Cl Rm 50 Figure 1.17 Preparation of (L)-carnitine derivatives. (3) bis-silylated amine, SiOz, 50 %; (b) Chloroambucil, Et3N, 52 %; (c) (i) TBAF/THF/imidazole; (ii) p-tosyl chloride, DMAP, Et3N, 23 %, 2 steps; (d) TFA, 87%. Ring opening of the chiral epoxide 48 by bis(tert-butyldiphenylsilyloxyethyl)amine followed by acylation using chloroambucil give the y-amino B-hydroxyester derivative 23 49. The free acid 50 is obtained after deprotection of 49 followed by transforming the hydroxyl groups to chlorides. Some other medicinally important compounds derived from 3 and 4 carbon synthons are listed in Figure 1.18. ° /\/\ k 0" )~~u 0.. H ° 3 u o "$er at“ -20 “Y 53 beta-3 Adrenerglc receptor 8 Pro ran 0' 0' antagonist for obesity, diabetes ( l' P (S)-Atenolol / \ (HO)2(O)PCH20 OH HO NHCH30H3NHCON O >_/ \_/ Wfluf/ ”43° F—t O 54 55 Xamotarol c f beta-1 Adrenocoptor partial agonist Ammrgnt Figure 1.18 Some of the medicinally important compounds derived from chiral 3- carbon synthons. 1.3.2 Iminosugar derivatives lmino-sugars, or aza sugars, are a class of sugar mimics with the ring oxygen substituted with nitrogen atom. They are important inhibitors for glycosidase and glycotransferase which are involved in many important biological processes, such as intestinal digestion, glycoprotein post-translation, glycoconjugate catabolism, and etc.55'57. They are potential drug candidates to treat diabetes, viral disease, lysosomal storage disease, cancer. and 24 etc.58'62 Because of the structural similarity between imino-sugars and carbohydrates. . . . 3-5 carbohydrates are often used for the preparation of immo-sugars6 6 . Figure 1.19 lists some of the important imino-sugars. Deoxynojirimycin 56 is a potent a- glucosidase inhibitor“. Its derivative, N-hydroxylethyl-deoxynojirimycin 57, is a potent a—glucosidases inhibitor with higher in vivo efficacy and has been approved for the treatment of type II diabetes (Miglitol)67. N-butyl-Deoxynojirimycin 58 (Zavesca) recently has been approved for the treatment of a glycosphingolipid lysosomal storage disease, type I Gaucher disease”. OH HO HO "0 N u/\/\ “ .0 " “lid HO HO HO HO OH OH OH 56 57 58 Deoxynojirimycin mgnw Zavesca Figure 1.19 Important iminosugars Many methods with different starting materials have been used in the preparation of imino-sugars. The obvious structural similarities between iminosugars and carbohydrates make carbohydrates the ideal starting point. Besides carbohydrates, other chiral pool materials. such as amino acids, tartaric acids have also been used“). Imino-sugars have also been synthesized from achiral precursors, such as olefins, through Sharpless O asymmetric epoxidation and dihydrolylaiton7. This subject has been extensively reviewed recentlym'n. 25 1.3.3 F uran/pyran derivatives Functionalized tetrahydrofuran and tetrahydropyran are common components of naturally . . . . 7 occurring and synthetic bloactlve compounds” 4. couug NH, /=( 4;l~\(cormz 99 %) and no further purification was needed. OH 0A" OAII b AIIO HO AIIO HO —"—’ AIIO A110 01" 0M3 OAII 0M9 5 5 7 CA" Figure 2.5 Reduction of methyl 2,3,4,6-tetra-O-allyl-a-D-glucopyranoside with Et3SiH (a) Allyl Bromide, DMSO/NaOH, 24 h, 85 %; (b) Et38iH/BF3OEt2, CF3SO3H, CHzClz, DOC-r.t, 6 h, 91 %. 2.2.2 Practical and economic Palladium chloride catalyzed de-allylation of 1,5- anhydro-2,3,4,6-tetra-O-allyl-D-glucitol. By refluxing allyl ether 7 in methanol in the presence of palladium(II) chloride for 12 h, the deprotected 1,5-anhydro-D-glucitol (8) was obtained in almost quantitative yield. OAII 0” o PdCIz, additives 0 AIIO = ”0 AIIO MeOH “0 OAII 0" 7 8 Figure 2.6 Deallylation of 1,5-anhydro-2,3,4,6-tetra-O-allyl-D-glucitol with PdClz Using Cqu as a co-catalyst and activated carbon as an additive, a real catalytic protocol 42 for the cleavage of allyl ether was developed. As shown in table 2.2, when Cqu was used as a co-catalyst, the product was obtained in 91% with only 1 mol % of PdClz (entry 3). The reaction runs smoothly at 0.1 gram scale. However, when the reaction was carried out on a 1.0 gram scale, only 15% of the fully de-allyllated product was obtained. When activated carbon was added to facilitate the distribution of the solid catalyst PdClz, the reaction can be performed on up to a 5.0 gram scale to yield the desired product in around 90% yield. Table 2.2 Deprotection of 2,3,4,6-tetra-O-allyl-D-1,5—anhydro-glucitol conditions Entry Allyl-glucitol (gram) C - PdClz (mol %) Yield (%) uCl2-2HzO Activated carbon 1 0.] none none 80 93 2 0.1 none none 4 10 3 0.1 1 eq none 1 91 4 1.0 1 eq none 1 15 5 1.0 1 eq 0.5 gram 1 90 6 3.0 leq 3.0 gram 1 91 7 5.0 1 eq 5.0 gram 1 90 2.2.3 Application of the Allylation-Reduction-De-allylation protocol to other mono, disaccharides and complex carbohydrates. Methyl D-mannopyranoside Under the same conditions as for the reduction of methyl 2,3,4,6-tetra-O-allyl-a-D- glucopyranoside, methyl 2,3,4,6-tetra-O-allyl-a-D-mannopyranoside 10 was reduced to 1,5-anhydro-2,3,4,6-tetra-O-allyl-D-mannitol 11 in 90 % yield (Figure 2.15), 43 DA" on on can ,0 HO ’0 __a___. At _L_. HO Al 0M0 0M0 9 i 10 OAII OAII c OH OH AIIO HO Al Ho 1 1 1 2 Figure 2.7 Reductive cleavage of methyl 2,3,4,6-tetra-O-allyl-D-mannopyranoside (a) Allyl Bromide, DMSO/NaOH, 24 h, 85 %; (b) Et3SiH/BF3OEt2, CF 3SO3H, CHzClz, 0°C, 5 min, 90 %; (c) PdClz/CuClz, activated carbon, MeOH, refulx, 12 h, 90 %. After refluxing l,5-anhydro-tetra—O-allyl-D-mannitol 11 in methanol for 12 h with 1 mol % of PdCIz, 1,5-anhydro-D-mannitol 12 was obtained in 91 % yield. Methyl D-ribofuranoside Methyl-D-riboside 13 (mixture of a/B anomers, ration 1/9) was first protected with allyl groups. The reduction of methyl 2,3,5-tri-O-allyl-D-riboside 14 under the same conditions as for the reduction of methyl-D-glucoside derivative was done within 3 minutes at 0°C and gave the reduced product 15 in 91 % yield. 44 HO A110 0 0 OMG a "9 b —_—___—.> —____> 0H 011 A110 0A" 13 14 H0 A110 0 0 C ————-> 0H 0H AIIO OAII 15 15 Figure 2.8 Reductive cleavage of methyl 2,3,5-tri-O-allyl-D-ribofuranosides (a) Allyl Bromide, DMSO/NaOH, 24 h, 87 %; (b) Et3SiH/BF3OEt2, CF3SO3H, CHzClz, 0°C, 3 min, 91 %; (c) PdClz/CuClz, activated carbon, MeOH, refulx, 12 h, 85 %. Sucrose Octa-O-allyl-sucrose 18 was prepared through allylation of sucrose 17 with allyl bromide and sodium hydride in DMF in 68 % yield. The reduction of 18 was done in 8 h, the D- glucopyranosyl group gave rise to a single anhydroalditol, namely, l,5-anhydro-2,3,4,6- tetra-O-allyl-D-glucitol 7 (45 %), and the D-fructofuranosyl group gave two anhydroalditols, identified as 2,5-anhydro-1,3,4,6-tetra-O-allyl-D-mannitol 22 (39 %) and 2,5-anhydro-1,3,4,6-tetra- O-allyl -D-glucitol 21 (7 %). 45 O H GA" 0 cnzon a 0 (”how HO o _, A110 0 HO HO AIIO AIIO OH 0 0A" O cnzoml cnzon OH OAII OAII email" 0 o b All 0 + AIIO + Alto A"° Anonzc cnzoau CHIOA" OAII can OAII 7 19 20 OH cnon o o c 0 HO _____. HO + “O + ”0 HOHzC cnzon ”2°” OH on on 8 21 22 Figure 2.9 Reductive cleavage of Octa-O-allyl-sucrose. (a) Allyl Bromide, DMSO/NaOH, 24 h, 68 %; (b) Et3SiH/BF3OEt2, CF3S03H, CHzClz, 0°C, 8 h, 91 %;(c) PdClZ/CuClz, activated carbon, MeOH, refulx, 12 h, 85 %. Cellulose For the preparation of tri-O-allyl-polysaccharides, the polysaccharides were first dissolved in hot DMSO (65°C), followed by addition of powered NaOH and then allyl bromide. In this way, tri-O-allyl-cellulose 24 was obtained in 85 % yield while tri-O- allyl-starch 26 and tri-O-allyl-levan 28 were obtained in 80 % and 78 % respectively. 46 OAII on can 0" 0 a A110 0 O HO 0 ___.. o 0 HO 0 AIIO O OH OAII OH n OAII n on b, c 0 ¢ 1'10 HO OH 8 Figure 2.10 Reductive cleavage of tri-O-allyl-cellulose. (a) Allyl Bromide, DMSO/NaOH, 24 h, 85 %; (b) Et3SiH/BF3OEt2, CF3SO3H, CHzClz, 0°C-r.t, 24 h; (c) PdClz/CuClz, activated carbon, MeOH, refulx, 66 %, 2 steps. Under similar conditions as for the reduction of methyl tetra-O-allyl-glucoside, tri-O- allyl-cellulose 24 was reduced, however, the reaction time was much longer, and it took 24 hours for the reaction to be completely done. After de-allylation, 1,5-anhydro-D- glucitol was obtained as the single product, and the yield for the two steps (reduction and de-protection) is 66%. Starch The reduction of tri-O-allyl-starch 26 was done in 24 hours, de-allylation of the mixture gave 1,5-anhydro-D-glucitol as the single product in 69 % yield. 47 OAII on o 0 OH a O HO AIIO OH 0 OAII 0 HO on n 25 26 Figure 2.11 Reductive cleavage of tri-O-allyl-starch (a) Allyl Bromide, DMSO/NaOH, 24 h, 80 %; (b) Et3SiH/BF3OEt2, CF 3SO3H, CHzClz, 0°C-r.t, 24 h; (c) PdClz/CuClz, activated carbon, MeOH, reflux, 69 %, 2 steps. Levan Using the same sequence as for reduction of tri-O-allyl-cellulose, reduction of tri-O-allyl- levan gave 2,5-anhydro-D-mannitol 22 and 2,5-anhydro-D-glucitol 21 as the two products in the ratio of 5 to l and in 65 % yield afler the reduction and de-protection steps. 48 HO O 0 OH A110 0 HO HO A110 a OH HO m OH AIIO All 0 HO 0 o AIIO o O HO HO A110 OH HO OH HO n A110 A110 “'0 A110 n 27 28 cnzou o o b,c HO _______ HO + Honzc cnzon OHIO” 21 22 Figure 2.12 Reductive cleavage of tri-O-allyl-levan (a) Allyl Bromide, DMSO/NaOH, 24 h, 78 %; (b) Et3SlH/BF3OE12, CF3SO3H, CH2C12, 0°C-r.t, 16 h; (C) PdCIz/CUClz, activated carbon, MeOH, reflux, 65 %, 2 steps. 2.3 Discussion 2.3.1 Establishing the protocol for triethylsilane cleavage of the glycosidic linkage. To avoid the decomposition and rearrangement of carbohydrates under acidic conditions, the free hydroxyl groups must be protected. To find the suitable protecting group for carbohydrates, especially the complex carbohydrates, such as cellulose, starch, levan, several factors must be considered. First, what kind of solvent should be used? Carbohydrates are not soluble in solvents with low dielectric constants, such as toluene, dichloromethane. The best solvent for carbohydrates is water, but water is not a good solvent for the protection of complex carbohydrates for several reasons. The first reason is that the hydroxyl group of water will compete with the hydroxyl groups of carbohydrates which make some reactions, e.g., acetylation of carbohydrates, impossible to be run in water. The second reason is that once the majority of the hydroxyl groups are 49 protected, the product tends to precipitate from water due to the hydrophobic properties of the protecting groups and thus makes it impossible for the protection of all the hydroxyl groups. The solvents with high dielectric constants, such as DMSO, DMF, can solve or partially solve the complex carbohydrates, and the protected carbohydrates are also soluble in them. However, one possible problem still exists: the partially protected carbohydrate chains tend to fold which makes some of the free hydroxyl groups are buried in the coil and hard to be functionalized. Using DMSO, DMSO/H20 and DMF, satisfactory results were obtained for both the monosaccharides and polysaccharides. Second, what kind of protecting group should be used? The installation and removal of the protecting group should be friendly and no harsh reaction conditions or toxic reagents should be used, also, costly reagents should be avoided. The protecting groups must survive the reduction process: since acid is used so no acid labile functional group should be used and no functional groups which are easily reduced by triethylsilane should be used either. The stereo and electronic effects of the installed functional groups on the reduction process are also crucial: the intermediate formed in the reduction process is oxocarbenium ion, electron withdrawing group at 2-position will retard or stop the formation of the oxocarbenium ion while electron giving groups will facilitate the process. Sterically demanding groups will make the access of the oxocarbenium ion by triethylsilane difficult, thus retard the reduction process. Methyl group is the best choice if no deprotection was needed for the reduced products. Methyl ethers are easy to install, stable to acid and silane, slightly electron giving, and relatively small in size. Indeed, methyl group was the mostly used protecting group in the 50 analysis of complex carbohydrates by the silane reduction method23'2°. However, the harsh conditions used in the cleavage of methyl ether bonds make it not a suitable choice since our purpose is to prepare the unprotected anhydroalditols. Acetyl groups are easy to install and remove, relatively small in size and it is one of the least costly protecting groups. One possible problem is that it is an electron withdrawing group, and participation of 2-acetyl group to the reduction process is also possible through formation of the dioxonium ion with C-l. However, similar reactions with saccharides containing propanoate groups at C-2 had been used with Me3SiOSOzCF3 as the catalyst25 . Thus acetyl groups were chosen as the protecting groups for evaluation of the reductive cleavage process. To simplify the reaction process, methyl a-D-glucopyranoside was chosen as a model compound. However, no reduction product was obtained from methyl 2,3,4,6-tetra-O-acetyl-a-D-glucopyranoside, the only product after workup was the hydrolyzed starting material, glucose (Table 2.2, Figure 2.13). A possible reaction pathway was shown in Figure 2.13. Once the carbocation 31 was formed, the acetyl group from C-2 apparently stabilized it and formed the dioxonium ion 32, thus making the hydride transfer to the anomeric carbon disfavored, so that no reduction product was formed. 00° 5 equiv Et3SlH OH O 5 equiv BF3_OEt AGO ¢ HR 0 Ac 0 acid HO OH 33 H9 1 MoOHIH-I- OAc 0“ OAC 4. ' c) o O A O AcO «éfl __. 1% L “3;, Ac '1 1 O (p O + O .,\ r r 30 31 32 Figure 2.13 Mechanism for reaction of methyl 2,3,4,6-tetra-O-acetyl-0t-D- glucopyranoside with Et3SiH Benzyl group will not participate in the reduction process; one possible problem is that under the action of hydride, the benzyl ether bond may be cleaved. However, if the cleavage of the glycosidic bond is faster than that of the benzyl ether bond, the cleavage of the benzyl ether bond will not be a problem. 0811 30 equiv EtasiH OH O 10 equiv BF3.OEt ”“32 . ; HO 0 mo 30 equnv TMSOTf H 0"“ CH2C'2 Ho one 4 0°C-r.t., 24hr 5 1 BF, I 08" can 1 ca mfl “£1 " 3"0 - BnO + ' F _ __. a 0 0 83; on. H 10‘ 0'“ g“;%\ ’ Et3SIH l 3‘2 0\ on. 51381 BF! C 'F 34 35 36 Figure 2.14 Mechanism for reaction of methyl 2,3,4,6-tetra-O-acetyl-0t-D- glucopyranoside with Et3SiH 52 No reduction of the glycosidic bond was observed even with excess Et3SiH (3O equiv) used. Instead, cleavage of the benzyl ether bond was observed. After workup, the product obtained was methyl a-D-glucoside 5. A mechanism was proposed in Figure 2.8. Coordination of BF3 with the benzyl ether oxygen formed complex 34; transfer of hydride from Et3SiH to the benzylic carbon gave the partially deprotected 36. Continued reductive cleavage of the benzyl ether bonds gave the deprotected 5. So, an alterative protecting group which will not participate during the reduction process and is more stable under acidic reductive conditions than benzyl ether is needed. And this protecting group should be easy to cleave. Allyl group seems to meet all the requirements for the reductive cleavage reaction: allyl ether is stable under acidic conditions; it will not participate in the reduction process as ester does, allyl group is relatively small in size, and the allylic hydrogens are also less reactive compared to benzylic hydrogens. The installation of allyl ether is relatively easy, and many methods have been reported for the cleavage of allyl ether. Allyl group was then chosen as the protecting group for the reductive cleavage reaction. For the formation of allyl ether, several methods are available. The most obvious and widely used method is through the reaction of alkali metal alkoxides with allyl halides. The reaction is best when carried out in polar solvent, such as DMF”. This method is good for the universal protection of all the hydroxyl groups in a certain compound. Another extensively used method for the formation of allyl ether is the tin method3°'32. The hydroxyl groups are first converted to tributylstannyl ethers or, in the case of diols, to dibutylstannylidene acetals and then alkylation of the alkoxy-tin derivatives give the 53 ally ether. An advantage of this method is that functional groups, such as azido groups, O-TBDMS groups and acetamido groups can be tolerated” 34. This method is mostly used when partial of the hydroxyl groups need to be protected selectively35‘37. The third widely used method is through palladium catalyzed decarboxylative rearrangement of mixed allyl alkyl carbonates. Alkyl allyl carbonates undergo decarboxylative rearrangement when heated at 50-70 0C in the presence of palladium catalysts”. The alcohols were first converted to allyl carbonates, and then palladium catalyzed extrusion of C02 give the allyl ether. This method has been successfully applied to base sensitive substrates, such as carbonate or 1,1,3,3-tetraisopropyldisiloxane-l,3-diyl (TIPS) derivatives of carbohydrates” 39. Alcohols can also be converted to allyl ethers through S40-42 d43, 44. their barium salt , or using the trichloracetamidate metho For the preparation of methyl 2,3,4,6-tetra-O-allyl-a-D-glucopyranoside 6, the most practical and economic method is through the Williamson synthesis. Using this method, methyl 2,3,4,6-tetra-O-allyl-a-D-glucopyranoside 6 was obtained in 85 % yield (Figure 2.9). The reduction of 6 gave the desired product 7 in high yield. At first, conditions used in the reductive cleavage of methyl protected carbohydrates were used23’ 24. With 30 equiv Et3SiH, 3O equiv TMSOTf, and 10 equiv BF3°OEt2, the reaction was done within 24 h at r.t., the desired product 1,5-anhydro-2,3,4,6-tetra-O-allyl-D-glucitol was obtained in 90% yield. Although these conditions work well for the reduction, the high equivalents of triethylsilane and TMSOTf used prevent the practical usage of these conditions for the preparation of 1,5-anhydro-D-glucitol in gram scale. Fortunately, we found that by using 54 catalytic amount of trifluoromethanesulfonic acid in dichloromethane, even without the expensive TMSOTf, a very good yield of the desired product can be obtained with much less triethylsilane and BF3-OEt2 used. Using only 2 equiv of triethylsilane, 3 equiv of trifluoroborane-etherate and catalytic amount of trifluoromethanesulfonic acid, the desired product was obtained in 91%, isolated yield. Furthermore, NMR shown the product was very pure (>99 %) and no further purification was needed. The proposed mechanism is shown in Figure 2.15. Coordination of boron trifluoride to the glycosidic oxygen forms the complex 37. At the assistance of the lone pair electrons from the ring oxygen, the glycosidic linkage is cleaved with the formation of the ring oxocarbenium ion and boron fluoride methanolate. The boron fluoride methanolate ion decomposes to form fluoride. and difluoromethoxylborane. At the assistance of fluoride, hydride transfers from triethylsilane to C-1 of 39 forms the reduced anhydroglucitol 7 and fluorotriethylsilane. All 4- A11 0‘" o + BFg-OMO A110 ---—> A110 A110 All 0‘" 0 A11 9 CA" 8152-0”. . \ 0“ 53/ _+ 6 37 38 F J 58351“ OAII GA" 4. 5:35“: + auo ° .— MIC 97 H AIIO AIIO U. CA" GA (31513 ' F 7 39 Figure 2.15 Mechanism for reaction of methyl 2,3,4,6-tetra—O-allyl-a-D- glucopyranoside with Et3SiH 55 2.3.2 Palladium chloride catalyzed de-allylation of 1,5-anhydro-2,3,4,6-tetra-O-allyl- D-glucitol. Many methods have been reported for the cleavage of allyl ether bond, most of them 4547 involve prior isomerization of the allyl ether, either base or transition metal catalysed,°°'50 to labile prop-l-enyl ethers which may then be cleaved under generally mild conditions. Several literature methods were evaluated. for the cleavage of the allyl ether. These included TMSCl/Nal5 ', NaBH4/1252, NBS and light53 and DDQ54. No satisfactory results were obtained either because the yield was too low (TMSCl/Nal, NaBH4/12, <5 %) or the recovery of the product was too problematic (N BS/light or DDQ ). Recently, palladium complexes have been used for the direct cleavage of allyl ether. PdC12(PhCN)2, used in stoichiometric amount, allows direct cleavage of allyl phenyl ethers to phenols in 85-95%yield55. Another powerful and selective method for cleavage of allyl ethers has found extensive use in carbohydrate chemistry5°’°3. In this method, the substrate is reacted with equimolar (or often excess) amounts of PdClz in aqueous acetic acid and in the presence of sodium acetate at temperatures ranging from 25°C to 70°C. The free alcohol is obtained directly. Although the above mentioned palladium mediated cleavage of allyl ether gave high yield of the deprotected product, stoichiometric amount of palladium usually is required. Kusama and co—workers devised a catalytic method for the deprotection of allyl glycosides“. Heating allyl glycosides in acetic acid in the presence of palladium tetrakis(triphenylphosphine) afforded the deprotected product. The amount of catalyst required is rather high (ca. 30 mol %). 56 For the deprotection of l,5-anhydro-2,3,4,6-tetra-O-allyl-D-glucitol, which has 4 allyl groups, 1.2 equivalent of palladium will be needed even 30 mol % of palladium per allyl group is used. Further more, the reported palladium based catalytic direct deprotection of allyl ether can only be used to cleave allyl ether as allyl glycosides“ 65. Thus, a more efficient, more general catalytic protocol must be developed for the deprotection of 1,5- anhydro-2,3,4,6-tetra-O-allyl-D-glucitol. We found that by refluxing the allyl ether solution in methanol in the presence of PdClz v for 12 h, the de-protected 1,5-anhydro-glucitol could be obtained in almost quantitative yield. In this case, catalytic amount of palladium (II) chloride (0.8 equiv or 0.2 equiv per allyl group) was used (entry 1, Table 2.2). When 0.04 equiv of PdClz was used, however, only 10 % of the product was obtained (entry 2, Table 2.2). The large amount of catalyst needed makes the development of a more efficient catalytic protocol necessary to make this reductive cleavage method practically viable. CA" CH PdClz, additives AIIO ° : o AIIO ”0 0A1! M90“ ”0 0H 7 8 Figure 2.6 Deallylation of l,S-anhydro-2,3,4,6-tetra—O-allyl-D-glucitol with PdClz One possible reason for the low efficiency of the palladium(II) chloride was that part of Pd(ll) was reduced to Pd(0) and thus lost the catalytic property. Cqu has the ability to oxidize Pd(0) to Pd(lI) and it is expected that by using Cqu as a co-catalyst, the Pd(0) will be recycled and thus only a very lower loading of PdClz was needed. Indeed, when 1 equiv of CuClz was used, with only 0.01 equiv of PdClz, the product was obtained in very high yield (91 %) (entry 3, Table 2.2). The reaction runs smoothly at 0.1 gram scale with the PdClz-CuClz catalytic system, however, when the reaction was carried out on a 1.0 gram scale, only 15% of the fully de-allyllated product was obtained (entry 3, Table 2.2). The possible reason was that at larger scale, as the volume of the solvent increased, the rate of the biphasic reaction decrease since PdC12 is not soluble in MeOH. A better disperse of the solid catalyst in the reaction system should be able to solve this problem. We were happy to find that with an equal amount of activated carbon used along with the substrate, the reaction can be performed on 5.0 gram scale without decreasing in yield (entry 7, Table 2.2). Although some palladium based catalysts for the deallylation of allyl ether have been developed, the mechanism is still not very clear°5'69 . A mechanism, based on anti- Markovnikov hydroxypalladation followed by B-alkoxy elemination, was proposed for cleavage of the allyl ether (Figure 2.16)7°. 58 H20 on fl 0" + %PdCl 42 '41 I—Pd612 HC' on 13% 40 ROH 43 pdCl §P0012 2 CUC12 Va Pdci OH NO + "<———Pd°" 2 /\5/ R= carbohydrate Figure 2.16 Proposed mechanism for catalytic cleavage of allyl ether Coordination of allyl-ether 40 with PdCl2 followed by attacking the terminal carbon of the olefin by H2O gives the intermediate 42, which loses ROH 43 (the free carbohydrate) and gives the allyl alcohol complex 44. Dissociation of PdCl2 with 44 gives free PdCl2 and finishes the catalytic cyc1e. The allyl alcohol formed in the process would reduce palladium(II) to palladium(0) with formation of oxidized by-products such as acrolein". Pd(0) can be oxidized back to Pd(II) by CuCl2 2.3.3 Application of the Allylation-Reduction-De-allylation protocol to other mono, disaccharides and complex carbohydrates. Once the Allylation-Reduction-De-allylation protocol was established, it was tested on the reduction of other glycosides and complex carbohydrates. It turns out that this protocol is very general; it can be used on other monosaccharides, such as mannose, 59 ribose, and disaccharide, such as sucrose, and on polysaccharides, such as starch, cellulose and levan. Methyl D-Mannopyranoside The stereochemistry at C-2'of aldohexapyranose has a great effect on the reactivity of C- 1. In this case, methyl ci-D-mannopyranoside was chosen to study the C-2 stereochemistry effect on the reductive-cleavage of the glycosidic linkage. For the reductive cleavage of the glycosidic linkage in methyl 2,3,4,6-tetra-O-allyl-a-D- mannopyranoside, the reaction was much faster than that of methyl 2,3,4,6-tetra-O-allyl- or-D-glucopyranoside, the reaction was done within 5 minutes at 0°C with 90 % yield (Figure 2.7) The only difference between the glucopyranoside derivatives and mannopyranoside derivatives is the stereochemistry at C-2: the hydroxyl (allyl ether for protected glycosides) group is equatorial for glucopyranoside while it is axial for mannopyranoside. It took 6 hours for the reduction to complete for glucopyranoside while only 5 minutes for mannopyranoside. What caused the big difference in the reaction rates between glucopyranoside and mannopyranoside derivatives? For the reduction of methyl 2,3,4,6-tetra-O-allyl-D-glucopyranoside and methyl 2,3,4,6- tetra-O-allyl-D-mannopyranoside, the mechanisms are the same (Figure 2.17): coordination of BF3 with the glycosidic oxygen forms the BF 3 - glycoside complexes 37 and 50, cleavage of the glycosidic bond forms the oxycarbenium ions 38 and 51, reduction of the oxycarbenium ion give the anhydroalditol derivatives 7 and 11. In this process, the rate determining step is the formation of the oxycarbenium ions 38 and 51, the same as in the hydrolysis of alkyl glycopyranosides72’73. 60 OAII 0“" ON. CA" slow + A110 0 L A" O _____, AIIO ‘2 fa" All 0 All A" OAII auo Et,sm All A110 0 - /o< can OAII \ F33 6 37 38 7 _ AIIO AIIO AIIO Auo OAII AIIO OAII 0‘0" BF; . 810W _—_.A"o' All All 0 _ + \ i=,a’o \ 1o 50 51 11 Figure 2.17 Mechanism for reduction of methyl a-D-glucoside and methyl a-D- mannoside. It has been known for many years that stereoisomeric glycosides hydrolyze with increasing rate depending on the number of axial hydroxyl groups”. OH HO OH HO 0“ HO O < O < 0 H0 H0 OHocn, . on OCH: OH 0" OCH, 5 ' 48 49 Figure 2.18 Relative acid-catalysed hydrolysis rates of glucoside, galactoside and guloside. An explanation for the hydrolysis rate difference between different glycopyranosides was based on the relief of steric strain. On the basis of available data and some assumptions on pyranoside reactant-state conformations and the mechanism of hydrolysis, Edward 75-78 gave an explanation to this phenomenon . The rationale as to why guloside 49 reacts faster than galactoside 48, which again reacts faster than glucoside 5 (Figure 2.18), was 61 based on the easiness for the relief of steric strain. Axial substituents would ease the rotation around the C2-C3 and C4-C5 bonds and thereby facilitate the transformation from reactant into intermediate, which possess a more flattened half-chair conformation. For the reduction of glycopyranoside derivatives, the rate determining step is also the formation of the oxocarbenium ion (Figure 2.17), the same rational can be used to explain the rate difference between glucopyranoside and mannopyranoside: the C-2 axial substitute in mannopyranoside eases the rotation around the C2-C3 and C4-C5 bonds and thereby facilitate the transformation from reactant into intermediate, which accounts for the big rate difference. The stereochemistry at C-1 of methyl 2,3,4,6-tetra-O-allyl-D-mannopyranoside does not affect the reaction rate much in the reduction process. No difference was observed for the or and [3 anomers. The de-allylation was also successful with the PdCl2-CuCl2-Activated- Carbon system. After refluxing 1,5-anhydro-tetra-O-allyl-D-mannitol in methanol for 12 h with 1 mol % of PdCl2, 1 equiv CuCl2 and activated carbon, 1,5-anhydro-D- mannitol was obtained in 91 % yield. Methyl D-ribofuranoside With the successful application of this reductive cleavage protocol on methyl D- mannopyranoside, the reductive cleavage of pentosides with this protocol should pose no problem. Ribosides were chosen as substrates to test this method. The reduction of methyl 2,3,5-tri-O-allyl-D-ribofiiranoside (mixture of a/B anomers. ration 1/9) under the same conditions as for the reduction of methyl D-glucopyranoside derivative was done 62 within 3 minutes at 0°C and the product was obtained in 91% isolated yield. In furanose, the ring carbon atoms are distorted from the tetrahedron structure and the substituents are eclipsed to each other. Thus, the furanose ring is highly strained. This strain is partially released when the oxocarbenium ion is formed. Compared to furanose, the pyranose ring is more flexible and the strains in pyranose were released through pucker of the ring. The structural difference between furanose and pyranose is the reason for the rate difference in the acid catalyzed hydrolysis of furanose and pyranose. For example, methyl a-D-mannofuranoside hydrolyzed 150 times faster than methyl a-D- mannopyranoside”. The same reason can be used to explain the rate difference in the triethylsilane reduction of methyl D-ribofuranoside and methyl D-glucopyranoside. In the silane reduction reactions, the 'rate determining step is also the formation of the oxocarbenium ion, as in the acid catalyzed hydrolysis of glycosides. The faster rate in the formation of the furan-oxocarbenium ion makes the reduction of methyl D- ribofuranoside faster than methyl D-glucopyranoside. 2.3.4 Preparation of 1,5-anhydro-D-glucitol, 2,5-anhydro-D-glucitol and 2,5- anhydro-D-mannitol from sucrose. Next, the disaccharide, sucrose, was used as the substrate for the reduction. Based on the results for reduction of monosaccharides, anhydroalditols are the expected products via the formation and reduction of oxocarbenium ions. The D-glucopyranosyl group was expected to give rise to a ‘single product, namely, 1,5-anhydro-2,3,4,6-tetra-O-allyl-D- glucitol The D-fructofuranosyl group can, however, give rise to two anhydroalditols, namely, 2,5-anhydro-1,3,4,6-tetra-O-allyl-D-mannitol and 2,5-anhydro-l,3,4,6-tetra-O- 63 allyl-D-glucitol. Compounds 2 and 3 are the result of net overall retention and inversion, respectively, of the stereochemistry at C-2 in the D-fructofuranosyl group. As expected, three products were obtained: 1, 5-anhydro-2,3,4,6-tetra-O-allyl-D-glucitol from the D-glucopyranosyl part of sucrose, 2,5-anhydro-l,3,4,6-tetra-O-allyl-D-mannitol and 2,5-anhydro-l,3,4,6-tetra-O-allyl-D-glucitol from the D-fructofuranosyl moiety of sucrose (Figure 2.21). A possible reaction pathway was shown in Figure 2.29. Coordination of trifluoroborane to the glycosidic oxygen forms the oxoniurn intermediate 52, which breaks down to form the glucopyranosyl derived oxocarbenium ion 38 and fructosyl derived oxocarbenium ion 53. Similar to the triethylsilane reduction of methyl glucopyranosides, transfer of hydride from triethylsilane to this oxocarbenium ion gives the allyl protected 1,5-anhydro-D- glucitol 7. 64 0A11 o cnzoau 0“" “"0 ‘8' BF, A" CH20AII A1 0 OAII o cnzoau + Allo OAII _? CHzoAll 3‘3 OAII 18 52 ON! OAII “’03" + AIIO All 0 L’s“ AIIO o + 0 mo A" \ | CH20AII can 0*" 'a's, OAII 7 38 F 53 , + AIIOHzc O o H o 5‘39"” anomc /AIIO + | ‘-——H (‘ “'0 CH20AII BF: ? CH20A11 OAII 3F, OAII 55 54 _ KN . F 3"“ CH20AII “’1 + O o AIIOH c /o ___._ “'0 “'0 2 “"0 Alionzc cnzoau t c.4202," CH2OAII can can cuzoau 56 19 20 Figure 2.19 Mechanism for reductive cleavage of Octa-O-allyl-sucrose Reduction of the oxocarbenium intermediate 55 gave products 19 and 20. The symmetrical 2,5-anhydro-D-mannitol derivatives 20 was formed as the major product (39 %) while the 2,5-anhydro-D-glucitol derivative 19, was obtained in 7 %. Cellulose The preparation of valuable chemicals from commodity carbohydrates, such as cellulose, starch, levan, etc., has very high economic potentials and is a great challenge for synthetic chemistry. The real test for the Allylation-Reduction-De-allylation proctol is 65 whether it can be used for the preparation of anhydroalditols from these commodity carbohydrates. Using this protocol, 1,5-anhydro-D-glucitol, 2,5-anhydro-D-glucitol and 2,5-anhydro-D-mannitol were prepared from complex carbohydrates, such as cellulose, starch and levan. The appropriate solvents for the allylation of polysaccharides should be able to dissolve or at least partially dissolve the polysaccharides and should also be able to solve the final products. Two polar, non-protonic solvents, DMSO and DMF, were chosen as the reaction solvents. When DMSO was used as solvent, the polysaccharides were first dissolved in hot DMSO (65°C), followed by addition of powered NaOH and then allyl bromide, the reaction was done in 10 h at 80°C. In this way, tri-O-allyl-cellulose was obtained in 85 % yield while tri-O-allyl-starch and tri-O-allyl-levan were obtained in 80 % and 78 % respectively (Figure 2.10). With DMF as solvent, NaH was used as the base, and the reaction was done in 24 h at r.t. A slightly higher yields were obtained with DMF as solvent (tri-O-allyl-cellulose, 92%, tri-O-allyl-starch, 85 %, tri-O-allyl-levan 85 %). For the reduction of these tri-O-allyl-polysaccharides, because all the hydroxyl groups are protected, no ring contraction or rearrangement should occur during the reduction process, and therefore, the only expected products are 1,5-anhydro-glucitol (from cellulose, starch), 2,5-anhydro-D-mannitol and 2,5-anhydro-D-glucitol (from levan). Under similar conditions as for the reduction of methyl tetra-O-allyl-glucoside, tri-O- allyl-cellulose was reduced, however, the reaction time was much longer, it took 24 hours for the reaction to be completely done. 13 C NMR analysis of the final product shown that 66 the anomeric carbon signals were totally disappeared. However, it also shown that the product was a complex mixture of several compounds. It is known that for the acetylation and alkylation of polysacchrides, it is hard to get the homogeneously functionalized products. Although no hydroxyl group signal (3100 cm") was detected by IR, it is still possible that part of the hydroxyl groups are un-allylated and the product obtained contain 1,5-anhydro-D-glucitol derivatives with different degree of allylation. If only 1,5-anhydro-D-glucitol derivatives present in the mixture, 1,5-anhydro-D-glucitol would be obtained as the single product after de-allylation. Indeed, afier de-allylation, 1,5-anhydro-D-glucitol was obtained as the single product, and the yield for the two steps (reduction and de-protection) is 66% (Figure 2.10). From tri-O-allyl-starch, 69 % of 1,5-anhydro-D-glucitol was obtained as the single product after the reduction-deprotection process (Figure 2.11). Levan is a D-fructan of high molecular weight that is comprised of a (2—6)-linked D- fructofuranose backbone which is branched at some of the O-l atoms. As for the reduction of sucrose, the expected products from the fructofuranosyl units are 2,5- anhydro-D-mannitol and 2.5-anhydro-D-glucitol. The same reaction sequences as for the reduction of tri-O-allyl-cellulose gave the two expected product 2,5-anhydro-D-mannitol and 2,5-anhydro-D-glucitol in the ratio of 5 to 1 and in 65 % yield after the reduction and de-protection steps (Figure 2.12). Table 2.3 listed the overall yields for the transformation of those commodity carbohydrates to the corresponding anhydroalditols. 67 Table 2.3 Transformation of glycosides to anhydroalditols. Yield Commodity carbohydrates Anhydroalditols (y) 0 Me-a-D-glucopyranoside l ,5-anhydro-D-glucitol 69.7 Me-a/B-D-mannopyranoside l ,5-anhydro-D-mannitol 70.5 Me-a/B-D-ribofuranosideide l ,4-anhydro-D-ribitol 72.9 1,5-anh dro-D- lucitol y g 38.3 Sucrose 2,5-anhydro-D-mannitol 33. 1 2,5-anhydro-D-glucitol 6.0 ‘ 1,5-anhydro-D-glucitol Cellulose 56.1 I ,5-anhydro-D-glucitol Starch 55.2 2.5-anhydro-D-mannitol 42.3 Levan 2,5-anhydro-D-glucitol 8.5 2.4 Conclusion The commercially viable transformation of commodity carbohydrates to high value added small chiral molecules and intermediates are imperative both environmentally and economically with the depleting of the fossil raw materials. An practical protocol for the preparation of anhydroalditols from commercially available carbohydrates was developed. The Allylation-Reduction-De-Allylation process transformed sucrose, cellulose, starch and levan to 1,5-anhydro-D-glucitol, 2,5-anhydro-D-mannitol and 2,5-anhydro-D- 68 glucitol in high yields and purity. The transformation was neat and clean, no column separation was needed for the preparation of 1,5-anhydro-D-glucitol from cellulose and starch. The catalytic cleavage of the allyl ether with the PdCl2-CuCl2-Activated-Carbon system makes these transformations more practical economically. 69 2.5 Experimental General procedures: lH, '3 C NMR spectra were recorded at 500, 125, MHz, respectively, with a Varian instrument at 293 K. The chemical shifts are given in ppm using CDCl3 residue as reference (87.24 ppm) for lH and relative to the central CDCl3 resonance (8: 77.00 ppm) for '3 C NMR unless otherwise specified. 1H and '3C are assigned on the basis of 2D IH COSY and 1H-13 C chemical-shift correlated experiments. Melting points were determined on a Fisher-Johns melting point apparatus (uncorrected). Optical rotations were measured on a Jasco P1010 polarimeter at 20°C. IR spectra (wave numbers in cm") were recorded on 3 FT IR Nicolet 740 spectrometer in CHC13 solutions or KBr pellets. All chemicals were purchased from .Aldrich Chemical Co. and used without further purification. Methyl-2,3,4,6-tetra-0-ally1-a-D-glucopyranoside (6) To a stirred solution of methyl-a-D-glucopyranoside (1.94g, lOmmol) in DMSO (50ml), was added powered NaOH (2.72 g, 68 mmol). After stirring at r.t. for 30 min, allyl bromide (68 mmol) was drop-wise added during a 30 min periond. Stirring was continued for 24 h at r.t., the reaction mixture was then poured into ice-water (100ml) and the stirring was continued for another 30 min. The mixture was extracted with Ethyl ether(3x50ml), dried over anhydrous Na2SO4. concentrated under reduced pressure, and the product was purified by column chromatography on silica gel. The product methyl- 2,3,4,6-tetra-allyl-ci-D-glucopyranoside 6 was obtained as a colorless oil (3.0 g, 85 % ). [(11020 +940 (c 1, CHCl3); 'H NMR(CDC13): 5 3.34 (s, 3H, OCH3), 3.35 (dd, J = 3.4, 9.0 Hz, H-2), 3.40 (m, 1H, H-4), 3.57-3.60 (m, 3 H, H-S, H-6a, H-6b), 3.65 (t, J = 9.5 Hz, 1H, 70 H-3), 3.91-4.33 (m, 8H, 4 CH2 O-allyl), 4.70 (d, J = 3.4, 1H, H-l), 5.06-5.24 (m, 8H, 4 CH2 vinyl), 5.78-5.97 (m, 4H, 4 CH vinyl); l3C NMR(CDC13): 8 54.9 (OCH3), 68.3 (C-6), 69.8, 72.3, 72.4, 73.7 (4 CH2 O-allyl), 74.1, 77.2, 79.2, 81.3, 98.2 (C-l), 116.2, 116.5, 117.0, 117.4 (4 CH2 vinyl), 134.4, 134.7, 134.8, 135.2 (4 CH vinyl); IR umax (CHCl3) (cm") 3080, 2982, 2910, 2844, 1645, 1460, 1422, 1384, 1351, 1200, 1150, 1083, 1052, 980, 920. 1,5-anhydro-2,3,4,6-tetra-0-allyl-D-glucitol (7) To a solution of methyl-2,3,4,6-tetra-O-al1y1-a-D-glucopyranoside(2.83 g, 8.0 mmol) in CH2C12(40 ml) at 0°C, were added Et3SiH(2.68 ml, 16 mmol), BF3OEt2(3.48 ml, 24 mmol), and CF3SO3H(50 DI), and the reaction mixture was stirred at room temperature for 12 h. The reaction was then quenched with saturated aqueous NaHCO3(50 ml) and extracted with CH2C12(3 x 50 ml). The combined dichloromethane extracts were washed with water and brine, dried over anhydrous MgSO4, and concentrated under reduced pressure. The product 1,5-anhydro-2,3,4,6-tetra-O-allyl-D-glucitol 7 was obtained as colorless oil (2.36 g, 91 %): [(11920 +587 (c 1, CHC13); 'H NMR (CDC13): 5 3.07 (dd, J=- 10.3Hz, 11.0Hz, 1H, H-l-b), 3.23 (m, 1H, H-5), 3.28-3.31 (m, 2H, H3, H4), 3.35 (m, 1H, H-2), 3.60 (dd, J=2.0Hz, 10.5Hz, 1H, H-6a), 3.94 (m, 1H, H-la), 3.97-4.08 (m, 5H, CH2 O-allyl), 4.23-4.30 (m, 3H, CH2 O-allyl), 5.08-5.26 (m, 8H, CH2 vinyl), 5.76-5.96 (m, 4H, CH vinyl); l3C NMR(CDC13): 8 67.8(C-1), 68.7 (C-6), 71.7, 72.1, 73.5, 73.8 ( 4 CH2- allyl), 77.2 (C-3 or C-4), 77.6 (C-3 or C-4), 79.0 (C-S), 85.5 (C-2), 115.9, 116.2, 116.6, 116.8 (4 CH2 vinyl), 134.3, 134.6, 134.7, 135.1 (4 CH vinyl); umax (CHC13) 3083, 3008, 2924, 2867, 1672, 1462, 1423, 1350, 1243, 1151, 1087, 997, 933 cm’1 . 1,5-anhydro-D-glucitol (8) (a) With PdCl2 only as the catalyst: To a solution of 1,5-anhydro-2,3,4,6-tetra-O-allyl- D-glucitol(0.l g, 0.31 mmol) in methanol(7 ml) was added PdCl2(38.5 mg, 0.22 mmol), and the reaction mixture was stirred at 60°C for 8 h. After remove the solid and the solvent, the product 1,5-anhydro-D-glucitol 8 was obtained quantitatively (45 mg, 100 %). (b) With PdCl2-CuC12 catalyst system: To a solution of 1,5-anhydro-2,3,4,6-tetra-O- allyl-D-glucitol (0.1 g, 0.31 mmol) in methanol (7 ml) was added PdCl2 (385mg, 0.22 mmol), and CuC12 (0.54 mg, 0.0031 mmol), the reaction mixture was stirred at 60°C for 8 h. After remove the solid and the solvent, 1,5-anhydro-D-glucitol 8 was in 91 % yield. (c) With PdCl2-CuCI2-Activated Carbon catalyst system: To a solution of 1,5- anhydro-2,3,4,6—tetra-O-allyl-D-glucitol(5.0 g, 15.5 mmol) in methanol(350 ml) was added PdCl2(27.0 mg, 0.155mmol), and CuC12 (2.1 g, 15.5 mmol), the reaction mixture was stirred at 60°C for 8 h. After remove the solid and the solvent, ,5-anhydro-D-glucitol 8 was in 90 % yield. The crude 1,5-anhydro-D-glucitol was crystallized from EtOH: mp 142-143 °C; [a]DZ° +405 (c 1, H2O); 'H NMR (1)20): 3.83(dd, 1H, J=11.2Hz, 2.6Hz,H-1’),3.73(dd, 1H, J=13.7Hz, 1.0Hz, H-6), 3.53(ddd, 1H, J= 12.5, 4.2, 1.6, H-6’), 3.43(m, 1H, 1+2), 3.28(m, 1H, 11-3), 3.21(m, 2H, 11-4, 5), 3.12(dd, 1H, 11:12:11.0, H-l-b); 13C NMR: 60.1(C-6), 68.0(C-1), 68.5(C-2), 68.8(C-4), 76.6(C-3), 79.4(C-5). 1,3,4,6-Tetra-O-acety1-2,5-anhydro-D-glucito1. The 1,5-anhydro-D-g1ucitol product obtained was acetylated using AC2O/Pyr for further structural identification. 1H NMR (CDC13): 6 2.08, 2.10, 2.11, 2.12 (fours, 12 H, OAc), 4.06 (ddd, l H, H-2), 4.22-4.40 (complex, 4 H, H-1,1',6,6'), 5.00 (dd, J = 1.5, J = 3.5, 4.8, 6.3 Hz, 1 H, H-5), 4.16 (ddd, J= 3.7, 6.4, 11.3 Hz, 3.5 Hz, 1 H, H-4), 5.32 (dd, J= 1.5, 3.7 Hz, 1 H, H-3). The Allylation, reduction and deprotection of other monosaccharides are the same as the ones for glucoside, except that the reaction time varied. Methyl 2,3,4,6-tetra-0-ally1-a-D-mannopyranoside (10) The same procedure as for the preparation of methyl 2,3,4,6-tetra-O-allyl-a-D- mannopyranoside gave methyl 2,3,4,6-tetra-O-allyl-or-D-mannopyranoside 10 as a colorless oil (87 %): lH NMR(500 M Hz, CD3Cl): 5 3.25 (s, OMe), 3.53-3.56 (m, 1H, H-S), 3.57-3.59 (m, 4H, H-2, H-4, H-6a, H-6b), 3.91-4.08 (m, 8H, 4 CH2 O-allyl), 4.26 (dd, J = 5.7, 11.4 Hz, 1H, H-3), 4.63 (s, 1H, H-l), 5.02-5.23 (m, 8H, 4 CH2 vinyl), 5,78- 5.88 (m, 4H, 4 CH vinyl); 13C NMR ( 125M Hz, CDC13) 8 54.5 (OMe), 69.2 (C-6), 70.9, 71.2, 71.8, 72.2 (4 CH2O-a11yl), 73.7, 74.5, 74.6, 79.3, 98.9 (C-l), 116.3, 116.4, 116.6, 117.2 (4 CH2 vinyl), 134.7, 134.8, 134.9, 135.0 (4 CH vinyl). 1,5-anhydro-2,3,4,6-tetra-0-ally1-D-mannitol (11) The product was obtained as colorless oil (2.36g, 91 %): 1H NMR (CDC13): 3.29 (dd, J = 12.8, 1.1 Hz, 1H, H-lb), 3.32 (ddd, J = 9.5, 6.3, 2.0 Hz, 1H, H-5), 3.40 (dd, J = 9.3 Hz, 1H, H-3), 3.57(t, J = 9.7 Hz, 1H, H-4), 3.61(dd, J = 10.5, 6.3 Hz, 1H, H-6b); 3.70(dd, 73 J=10.5 Hz, 2.0 Hz, 1H, H-6a); 3.71(m, 1H, H-2); 3.98-4.06(m, 6H, H-la, CH2 O-allyl); 4.14-4.19(m, 2H, CH2 O-allyl), 4.34(m, 1H, CH2 O-allyl), 5.10-5.18(m, 4H, CH2 vinyl), 5.20-5.30(m, 4H, CH2 vinyl), 5.85-5.96(m, 4H, 4 CH vinly); '3 C NMR(CDC13): 67.1(C- l), 69.8(C-6), 70.6, 70.7, 72.5 (3 carbon, 3 CH2 O-allyl), 72.7 (C-2), 74.1 (CH2 O-allyl), 75.2(C-4), 79.6 (C-5), 82.3 (C-3), 116.6, 116.9, 117.2, 117.5 (4 CH2 vinyl), 134.8, 134.9, 135.0, 135.1 (4 CH2 vinyl); umax (CHC13) cm'I 3080, 3010, 2928, 2865, 1674, 1459, 1424, 1355, 1244, 1157, 1087, 995, 930 cm'l . 1,5-Anhydro-D-mannitol (12) To a solution of 1,5-anhydro-2,3,4,6-tetra-O-allyl-D-mannitol (1.0 g, 3.1 mmol) in methanol(70 ml) was added PdCl2(6.0 mg, 0.031 mmol), and CuCl2 (0.42 g, 3.1 mmol), the reaction mixture was stirred at 60°C for 8 h. After remove the solid and the solvent, 1,5-anhydro-D-mannitol 9 was in 92 % yield (0.47 g): mp 154-155 °C; [a]DZ°- 50.5 (c 1, H2O); lH NMR (D20): 8 3.86 (br s, 1H), 3.80 (dd, J = 12.5, 1.5 Hz, 1H), 3.77 (dd, J = 12.5, 2.5, Hz, 1H), 3.57 (dd, J = 12.5, 6.4 Hz, 1H), 3.55-3.48 (m, 2 H), 3.46 (d, J = 9.5 Hz, 1H), 3.18 (ddd, J = 9.5, 6.5, 2.5 Hz, 1H); l3C NMR (D20): 8 82.21, 75.20, 71.48, 70.69, 68.93, 62.85. 1,S-anhydro-Z,3,4,6-tetra-O-acetyl-D-mannitol. lH NMR (CDC13): 2.01, 2.05, 2.11, 2.17 (4 s, 12 H, OAc), 3.59 (ddd, H-la), 4.07 (dd, J = 2.1, 13.2 Hz, 1 H, H-le), 4.14 (dd, J= 2.4, J: 2.4, 5.4, 9.9 Hz, 1 H, H-S), 3.67 (dd, J= 1.3, 13.2 Hz, 1 H, 12.3 Hz, 1 H, H-6), 4.24 (dd, J = 5.4, 12.3 Hz, 1 H, H-6), 5.06 (dd, J = 3.5, 10.0 Hz, 1 H, H-3), 5.28 (t, J = 10.0 Hz, 1 H, H-4), 5.32 (complex, 1 H, H-2). 74 For methyl D-ribofuranosides, a 01/8 mixture was used as the starting material for reduction. The tri-O-allyl products were partially separated, and methyl 2,3,5-tri-O-ally1- B-D-ribofuranoside was obtained while the a anomer was obtained as its mixture with the B anomer. Me-2,3,5-tri-0-alIyl-a-D-ribofuranoside. 13C NMR (CDC13): 8 55.0 (OMe), 70.0 (C-S), 71.2, 71.3, 71.5 ( 3 CH2 0-allyl), 75.2(C-), 77.9 (C-), 81.6 (C-), 102.0 (C-l), 116.7, 117.1, 117.3 (3 CH2 vinyl), 134.3, 134.4, 134.7 (3 CH vinyl). Me-2,3,5-tri-0-allyl-B-D-ribofuranoside lH-NMR 8 3.26 (s, 3H, OMe), 3.42 (dd, J = 5.9, 10.8 Hz, 1H, H-Sa), 3.50 (dd, J = 4.0, 10.7 Hz, 1H, H-Sb), 3.72 (d, J = 4.7’Hz, 1H, H-2), 3.86 (dd, J = 4.5, 7.5 Hz, 1H, H-3), 3.95-4.00 (m, 4H, 2 CH2 0-ally1),4.03-4.06(m, 2 H, CH2 O-allyl), 4.12 (ddd, J = 4.3, 6.0, 10.7 Hz, 1H, H-4), 4.79 (s, 1H, H-l). 5.05-5.24(m, 6H, 3 CH2 vinyl), 5.74-5.89(m, 3H, (3 CH vinyl); l3C NMR(CDC13): 8 54.7 (OMe), 71.2 (3 carbon, 3 CH2 O-allyl), 71.9 (C-5), 78.1, 79.5, 80.1, 106.1 (C-l), 116.5, 117.1, 117.2 (3 CH2 vinyl), 134.1, 134.2, 134.5(3 CH vinyl). l,4-anhydro-2,3,5-tri-0-allyl-D-ribitol (15) Reduction of pure methyl-2,3,5-tri-O-allyl-B-D-ribofuranoside and a mixture of 01/13 anomers gave the same product 1,4-anhydro-2,3,5-tri-O-a11yl-D-ribitol 15 as a 75 colorless oil in 90 % yield: lH-NMR 8 3.44 (dd, J = 4.4, 10.8 Hz, 1H, H-Sa), 3.54 (dd, J = 3.5, 10.7 Hz, 1H, H-Sb), 3.81 (t, J = 5.6 Hz, 2H, H-), 3.92-4.03 (m, 8H, 3 CH2 0- allyl), 4.08 (m, 1H, H-S), 5.08-5.26(m, 6H, 3 CH2 vinyl), 5.78-5.91(m, 3H, 3 CH vinyl); l3C NMR(CDC13): 8 69.9, 70.4, 70.8, 71.1, 72.2, 76.4, 78.1, 80.1, 116.7, 117.1, 117.2 (3 CH2 vinyl), 134.3, 134.4, 134.4(3 CH vinyl). 1,4-anhydro-D-ribitol (16) De-allylation of l,4-anhydro-2,3,5-tri-O-ally1-D-ribitol gave 1,4-anhydro-D-ribitol 16 in 89 % yield: mp 101-102 °c ; [011920 +63.0; 'H-NMR (1320) a 3.45 (m, 2 H, 115, H-S’), 3,63 (m 3 H), 3.89 (m, 2 H), 4.09 (m, l H); l3C NMR (D20) 8 61.5, 71.2, 71.7, 72.4, 81.7. 1,4-anhydro-2,3,5-Tri-0-acetyl-D-ribitol. lH NMR (CDC13): 8 2.08, 2.09, 2.10( 3 s, 9 H, OAc), 3.87 (dd, J = 3.9, 10.3 Hz, 1 H, H-la), 4.12 (dd, J = 5.0, 11.3 Hz, 1 H, H-S), 4.16 (complex, 1 H, H-4), 4.23 (dd, J = 5.2, 10.3 Hz, 1 H, H-lb), 4.33 (complex, 1 H, H-S'), 5.13 (complex, 1 H, H-3), 5.37 (dt, J = 3.9, 5.3 Hz, 1 H, H-2); l3C NMR(CDC13): 8 19.9, 20.0, 20.0 (3 Me from Ac), 61.0, 61.2 (C-1 and C-5), 67.4(C-2), 68.5, 68.6 (C-3 and C-4), 169.0, 169.2, 169.6(3 Ac) Octa-O-allyl-sucrose (18) In a 250 m1 round bottom flash was added NaH (60 % in mineral oil, 6.7 g, 140 mmol), after washing with hexanes (3 x 20 ml), DMSO (150 ml) was added, followed by addition of a solution of sucrose (3.0 g, 8.77 mmol)1 in DMSO (30 ml). Water bath was used to control the temperature of the reaction mixture below 45°C. After stirring for 1 h, allyl bromide (9.2 ml, 105 mmol) was drop wised added over 30 min period, with water bath to control the temperature below 50°C. After stirring at 45°C for 12 h, the reaction mixture was poured into cold water, and extracted with diethyl ether (4 x 70 ml). The combined organic layer was washed with water (3 x 100 ml), brine (3 x 100ml) and then dried over anhydrous sodium sulfate. After filtration and removal of the solvent under reduced pressure, a colorless oil (5.9 g) was obtained. NMR shown the product was the desired octa-O-allylsucrose with > 96 % purity (85 % yield). Small amount the raw product was further purified by flash column (Hexanes/Ethyl Acetate: 1/9) for characterization purpose. [00020 + 58.0° (c 1.00, EtOAc); lH-NMR 8 3.25 (dd, J = 3.9, 9.7 Hz, 1H, H-2), 3.34 (dd, J = 6.4, 9.1 Hz, 1H, H-4), 3.41 (s, 1H, H-lb’ ), 3.52-3.62 (m, 6 H, H-3, H-6a, H-6b, H-l’a, H-6’a, H-6’b), 3.87 (m, 1H, H-5’), 3.91-3.96(m, 5 H, H-5, 2 CH2 O-allyl), 3.98 (m, 2 H, CH2 O-allyl), 4.00(m, 3 H, H-4’, CH2 O-allyl), 4.04-4.06(m, 4 H, 2 CH2 O-allyl), 4.11-4.18(m, 4 H, 2 CH2 0-ally1), 4.22-4.30(m, 1H, H-3’), 5.14-5.30 and 4.98-5.13 (m, 16H, CH2 vinyl), 5.49(d, J = 3.8 Hz, 1H, H-l), 5.74-5.94 (m, 8H, CH2 vinyl); 13C NMR(CDCI3): 8 68.4 (C-6), 70.2 (C-5), 70.6 (C-l’), 71.2 (CH2-3’ 0- allyl), 71.5 (CH2-4’ 0-allyl), 71.5 (CH2-2 O-allyl), 71.9 (C-6’), 71.9 (CH2-6’ O-allyl), 72.2 (CH2-6 O-allyl), 72.2 (CH2-1’ O-allyl), 73.5 (CH2-4 O-allyl), 73.9 (CH2-3 O-allyl), 76.9 (C-4), 79.2 (C-2), 79.4 (C-5’), 81.1 (C-3), 82.2 (C-4’), 83.3 (C-3’), 89.9 (C-l), 104.3 (C-2‘), 116.0, 116.2, 116.2, 116.5, 116.5, 116.6, 116.7, 116.8, (8 CH2 vinyl), 134.4, 134.5, 134.6 (3 carbon), 134.8, 135.0, 135.4, (s CH vinyl). Reduction of Octa-O-allyl-sucorse To a solution of Octa-O-al'lyl-sucrose 18 (3.31 g, 5.0 mmol) in CH2C12 (40 ml) at 0°C, were added Et3SiH (3.35 ml, 20 mmol), BF30Et2 (4.35 ml, 30 mmol), and CF3SO3H (60 77 01), and the reaction mixture was stirred at room temperature for 12 h. The reaction was then quenched with saturated aqueous NaHC03 (50 ml) and extracted with CH2Cl2 (3 x 50 ml). The combined dichloromethane extracts were washed with water and brine, dried over anhydrous MgS04, and concentrated under reduced pressure. Flash column chromatography gave three products 7, 19 and 20. l,5-anhydro-2,3,4,6-tetra-0-allyl-D-glucitol (7): 45 % yield, gave the same spectra data as the product from reduction of methyl 2,3,4,6-tetra-O-allyl-a-D—glucopyranoside. 2,5-anhydro-l,3,4,6-tetra-0-allyl D-mannitol 20: 39% yield, [91620 + 26.4° ( c 1.00, CHC13); lH-NMR 8 3.48 (dd, J = 4.3, 5.8 Hz, 4H, H-la, H-lb, H-6a, H-6b), 3.88 (dd, J = 1.4, 2.5 Hz, 2H, H-3, H-4), 3.95-3.99 (m, 8H, 4 CH2 O-allyl), 4.03-4.06 (m, 2H, H-2, H-S), 5.08-5.12 (m, 4H, CH2 vinly), 5.18-5.24 (m, 4H, CH2 vinly), 5.78-5.86 (m, 4H, CH vinly); l3C NMR(CDC13): 8 70.0 (2 carbon, C-1, C-6), 70.5 (2 carbon, CH2 O-allyl), 72.1 (2 carbon, CH2 O-allyl), 81.4 (2 carbon, C-2, C-5), 84.6 (2 carbon, C-3, C-4), 116.7 (2 carbon, 2 CH2 vinyl), 116.8 (2 carbon, 2 CH2 vinyl), 134.2 (2 carbon, 2 CH vinyl), 134.5 (2 carbon, 2 CH2 vinyl); Anal. Calcd for C18H2805: C, 66.64; H, 8.70. Found: C, 66.56; H, 8.62. 2,5-anhydro-D-mannito1 (22): [61020 +522 °( c 1.00, H20); M.p. 100-102 °C; 'H NMR (D20): 8 3.68 (dd, J1 = J2 = 5.6 Hz, 2H. H-lb, H-6b), 3.78 (dd, J = 3.1, 12.4 Hz, 78 2H, H-la, H-6a), 3.90(m, 2H, 11-2, 11-5), 4.05(m, 2H, H-3, H-4); l3c NMR(CDC13): 8 61.3 (2 carbon, C-1, C-6), 76.6 (2 carbon, C-3, C-4), 82.4 (2 carbon, C-2, C-5). 1,3,4,6-Tetra-0-acetyl-2,5-anhydro-D-mannitol: [a][22° + 26.5 °( c 1.00, CHC13); 1H NMR (CDCl3): 8 2.11 (s, 12 H, 4 OAc), 4.25 (s, 6 H, H-la,1H-1b, H-2, H-5, H-6a, H- 6b), 5.16 (ss, 2 H, H-3, H-4); ”C NMR(CDC13): 8 21.2 (4 carbon, CH3 OAc), 63.4 (2 carbon, C-1, C-6), 78.5 (2 carbon, c3. 04), 81.4 (2 carbon, c.2, 05), 170.3 and 171.0 (2 carbon, C=O Ac). 2,5-anhydro-l,3,4,6-tetra-0-allyl D-glucitol (19) 7 % yield, lH-NMR 8 3.46 (m, 4 H, H-la, H-lb, H-6a, H-6b), 3.90 (m, 2H, H-3, H-4), 3.93-3.99 (m, 8H, 4 CH2 O-allyl), 4.00-4.05 (m, 2H, H-2, H-S), 5.07-5.12 (m, 4H, CH2 vinly), 5.19- 5.25 (m, 4H, CH2 vinly), 5.79—5.86 (m, 4H, CH vinly); l3C-NMR 8 70.2( C-l or C-6), 70.4 (C-l or C-6), 70.5, 70.9, 72.0, 72.9 (4 CH2 0 allyl), 75.3, 79.0, 83.1, 83.7, 116.1, 1 16.6, 116.7, 117.0 (4 CH2 vinyl), 134.4, 134.7, 134.9, 135.3 (4 CH vinyl). 2,5-Anhydro-D-glucitol (21) lH NMR (D20): 8 3.68 (dd, J = 12.1, 6.0 Hz, 1H, H-l), 3.73 (dd, J = 7.0, 11.8 Hz, 1H, H- 6), 3.77 (dd, J = 3.8, 12.1 Hz, 1H, H-l’), 3.82 (dd, J = 4.3, 11.8 Hz, 1H, H6’), 3.84 (ddd, J = 3.7, 4.3, 6.0 Hz, 1H, H-2), 4.01 (dd. J —- 2.4, 4.3 Hz, 1H, H-3), 4.12 (dt, J = 4.4, 7.0 Hz, 1H, H-5), 4.17 (dd, J = 2.5, 4.3 Hz, 1H, H-4); l3C-NMR(D20): 8 60.7, 62.3 (C-l, C- 6), 77.5, 78.6, 81.5, 85.2 (C-2, C-3, C-4, C-5). 79 General method for the preparation of tri-O-allyl-polysaccharides: Unprotected polysaccharide(1.0 gram) was dissolved in DMSO (50ml) at 60 °C, NaH (60 % in mineral oil, 5.84g, 122 mol, 5 mol/mol hydroxlyl group) was added to this after washing with hexanes (3 x 20 ml). Afier stirring at r.t for 1 h under nitrogen atmosphere, allyl bromide (10.7 ml, 122 mmol) was drop wised added over 30 min period, with water bath to control the temperature below 50°C. After stirring at r.t. for 12 h, the reaction mixture was poured into cold water, and extracted with chloroform (4 x 50 ml). The combined organic layer was washed with water (3 x 100 ml), brine (3 x 100ml) and then dried over anhydrous sodium sulfate. After filtration and removal of the solvent under reduced pressure, the tri-O-allyl-polysaccharides were obtained as a syrup. General method for the reduction of tri-O-allyl-polysaccharides: The methods for the reduction of tri-O-allyl-polysaccharides are the same as for monosaccharides, except longer times (24 h) were needed for the reduction to complete. Tri-O-allyl-cellulose (24) . 85 % yield for allylation: 1H NMR (CDCl;): 8 3.14 (br, 1H), 3.54 (br, 1H), 3.84-4.00 (br, 2H), 4.12 (br,lH ), 4.16-4.30 (br, 1H), 4.49-4.64 (br, 8H, 4 CH2 O-allyl ), 4.93-5.27 (br, 9H, H-1, 4 CH2 vinyl), 5.66-5.93 (br, 4H, 4 CH vinyl); '3 C NMR(CDCl3): 8 69.2 (br, C-6), 73.3 (br, C-S), 75.2-76.0 (br, CH2 O-allyl), 76.1 (br. C-3), 82.3 (br, 02), 84.2 (br, C-4), 102.9 (br, C-l), 116.3-117.0 (br, CH2 vinyl), 134.9-135.5 (br, CH vinyl); Reduction-deprotection steps gave 1,5-anhydro-D-glucitol in 66 % for 2 steps. Tri-O-allyl-starch (26) 80 85 % yield for allylation: : IH NMR (CDC13): 8 3.32 (br, 1H), 3.55 (br, 1H), 3.65 (br, 1H), 3.85-4.28 (br, 12 H, H-1, 4 CH2 O-allyl ), 5.00-5.23 (br, 8H, 4 CH2 vinyl), 5.60— 5.85 (br, 4H, 4 CH vinyl); l3C NMR(CDC13): 8 68.2(br, C-6), 70.0(br, C-5), 72.0-72.4 (br, 4 CH2 0-allyl), 73.1 (br, C-3), 79.5 (br, C-4), 81.4 (br, C-2), 116.2-117.0 (br, CH2 vinyl), 134.7-135.8 (br, CH vinyl); Reduction-deprotection steps gave 1,5-anhydro-D-glucitol in 65 % for 2 steps. Tri-O-allyl-levan (28) 78 % yield for allylation: 'H NMR (CDC13): 8 3.42-3.58 (m, 2H, H-6a, H-6b), 3.79- 3.94(m, 2H, H-3, H-4), 4.02-4.44(m, 11H, H-la, H-lb, H-5, 4 CH2 O-allyl), 5.18-5.55(m, 8H, CH2 vinyl ), 5.84-6.12(m,' 4H, CH vinyl); 13C NMR(CDC13): 8 62.2(br, C-6), 71.0(br), 71.1(br), 72.2(br), 72.3(br, 4 CH2 O-allyl), 72.3(br, C-5), 78.0(br, C-l), 82.1(br, C-4), 83.9(br, C-3), 103.8(br, C-2), 116.4(2 CH2 vinyl ), 116.7(2 carbon CH2 vinyl), 134.5, 134.7, 134.8, 135.0(4 CH vinyl). Reduction-deprotection gave 2,5-anhydro-D-mannitol (54.2 %), 2,5-anhydro-D-glucitol (10.8 %). 81 2.6 Reference [11 [2] [3] [41 [5] [6] [7] [8] [9] [10] [111 [12] [13] [14] [15] [16] [17] [18] [19] Morris, D.; Ahmed, I.. The carbohydrate economy: making chemicals and industrial materials from plant matter. 1992. Descotes, G., Editior. Carbohydrates as Organic Raw materials II. 1992. Bekkum, H. van; Roper, H.; Voragen, F. Editiors. Carbohydrates as Organic Raw Materials III. 1994. Lichtenthaler, F.W.; Peters, 8.. C. R. Chimie 2004, 7, 65. Kunz, M. From Sucrose to Semisynthetical Polymers. In Carbohydrates as Organic Raw Materials 11. Descotes, G. Ed. New York, 1993, pp135. Heinze, T. J.; Glasser, W. J. Editor. Cellulose derivatives: modification, characterization, and nanostructures. 1996. Nathan, S. Complex carbohydrates, their chemistry, biosynthesis, and functions: a set of lecture notes, 1975. Gilman, H.; Diehl, J. .1. Am. Chem. SOC. 1961, 83, 4817. Doyle, M. P. DeBruyn, D. J .; Kooistra, D. A. J. Amer. Chem. Soc., 1972, 94, 3659. West, C. T.; Donnelly, S. J.; Kooistra, D. A.; Doyle, M. P. J. Org. Chem, 1973, 38,2675. Doyle, M. P.; West, C. T. J. Org. Chem. 1975, 40, 3835. Doyle, M. P.; West. C. T. West. J. Org. Chem. 1975, 40, 3830. Doyle, M. P.; West, C. T.; Donnelly, S. J.; McOsker, C. C. J. Organomet. Chem, 1976, 117, 129. Doyle, M. P.; McOsker, C. C.; Ball, N.; West, C. T. J. Org. Chem, 1977, 42, 1922. Janzen, A. F.; Willis, C. J. Can. J. Chem, 1965. 43, 3083. Calas, R.; Frainnet, E.; Bonastre, J. C. R. Hebd. Seances Acad. Sci. 1960, 251 , 2987. Lapkin, 1. 1.; Povarnitsyna, T. N.; Anvarova, G. Y. Zh.Obshch. Khim, 1965, 35, 1835. Glushkova, N. E.; Kharitonov, N. P. Izv. Akad. Nauk SSSR, Ser. Khim, 1967, 88. Lapkin, I. 1.; Povamitsyna, T. N. Zh. Obshch. Khim, 1968, 38, 643. 82 [20] [21] [22] 1231 1241 1251 1261 1271 [28] [29] [30] [31] [32] [33] [341 [35] [36] [37] [38] Lapkin, I. 1.; Povarnitsyna, T. N. ; Kostareva, L. A. Zh. Obshch. Khim, 1968, 38, 1578. Fry, J. L.; Orfanopoulos, M.; Adlington, M. G.; Dittman, W. R. Jr., Silverman, S. B. J. Org. Chem, 1978, 43, 374. Doyle, M. P.; DeBram, D. J.; Donnelly, S. J.; Kooistra, D. A.; Odubela. A. A.; West, C. T.; Zonnebelt, S. M. J. Org. Chem. 1974, 39, 2740. Rolf, D.; Gary, G. R. Carbohydrate Research, 1984, 131, 17. D'Ambra, A. G.; Gray, G. R. Carbohydrate Research, 1994, 251, 115. Sherman, J. S.; Gray, G. R. Carbohydr. Res. 1992, 231, 221-35. Mischnick, P.; De Ruiter, G. A. Carbohydrate Polymers 1994, 23, 5. Kiwitt-Haschemie, K.; Renger, A.; Steinhart, H. Carbohydrate Polymers, 1996, 30, 31. Antonio, M.; Antonio, E.; Nicola, 1.; Rosa, Emiliano, Michelangelo, P. Eur. J. Org. Chem 2001, 927, 2931. Gill, J.; Gigg, R.; Payne, 3.; Conant, R. J. Chem. Soc. Perkin Trans. I 1987, 423- 429. David, 3; Hanessian, s. Tetrahedron 1985, 41,643-66. Blunden, S. J.; Cusak, P. A.; Smith, P. J. J. Organomet. Chem. 1987, 325, 141- 152; . . Pereyre, M.; Quintard, .J. P.; Rahm, A. Tin in Organic Synthesis, Butterworths, London, 1987, pp. 261-285. Boons, G. 1.; Castle, G. H.; Clase, J. A.; Grice, P.; Ley, S. V.; Pinel, C. Synlett 1993, 913-914. Nashed, M. A.; El-Sokhary, R. 1.; Ratib, L.; AnderSon, L. Carbohydr. Res. 1984, 131, 47-52. ‘ , Alais, J.; Veyrires, A. J. Chem. Soc. Perkin Trans. I. 1981, 377-381. Ogawa, T.; Kaburagi, T. Carbohydr. Res. 1982, 103, 53-64. Dasgupta, F.; Garegg, P. J. Synthesis 1994, 1121-1123. Guibb, F.; Mleux, Y. S. Tetrahedron Lett. 1981, 22, 3591-3594. 83 [39] [40] [41] [42] [43] [44] [45] [46] [47] [48] [49] [50] [51] [52] [53] [54] [551 [56] [57] [58] Oltvoort, J. J.; Klosterman, M.; Van Boom J. H. Reel. Trav. Chim. Pays-Bas 1983, 102, 501-505. Hingsdaul, 0.; Norberg, T.; Pendu, J. L.; Lemieux, R. U. Carbohydr. Res. 1982, 109, 109-142; Nashed, M. A.; El-Sokhary, R. 1.; Rateb, L. Carbohydr. Res. 1984, 131, 47-52. Eby, R.; Schuerch, C. Carbohydr. Res. 1982, 100, C41-C43. Iversen, T.; Bundle, D. R. .1. Chem. Soc. Chem. Commun. 1981, 1240-1241. Wessel, H. P.; Iversen, T.; Bundle, D. R. J. Chem. Soc. Perkin Trans. 1 1985, 2247-2250. Hubert, A. J .; Reimligen, W. Synthesis, 1969, 97. Prosser, T. J. J. Am. Chem. Soc. 1961, 83, 1701. Price, C. C.; Snyder, W. H. J. Am. Chem. Soc. 1961, 83, 1773. Crabtree, R. H. The Organometallic Chemistry of the Transition Metals, John Wiley, 1988, chap. 9, pp.185-190. Parshall, G. W. Homogeneous Catalysis, John Wiley, 1980, pp. 31-35. Hubert, A. J .; Reimlinger, H. Synthesis 1970, 405-430. Kama], A.; Laxman, E.; Rao, N.V. Tetrahedron lett. 1999, 40, 371-372. Thomas, R. M.; Mohan, G. H.; Iyengar, D. S. Tetrahedron Lett. 1997, 38, 4721- 4724. Diaz, R. R.; Melagatejo, C. R.; Espinosa, M. T. P. L.; Cubero, I. I. J. Org. Chem. 1994, 59, 7928. Yadav. K. S.; Chandrasekhar, S.; Sumitra, G; Kacke, R. Tetrahedron Lett. 1996, 37, 6603-6606. Bruce, M.; Roshan-Ali, Y. J. Chem. Res. S. 1981, 193. Ogawa, T.; Kitajima, T.; Nukada, T. Carbohydr. Res. 1983, 123, C5-C7. Smith, A. N.; Rivero,lR. A.; Hale, K. J.; Vaccaro, H. A. J. Am. Chem. Soc. 1991, 113, 2092-2112. Gyrggdeak, E. Liebigs Ann. Chem. 1991, 1291-1300. 84 [59] [60] [61 l [621 [63] [64] [65] [66] [67] [63] [69] [70] [71] [72] [73] [74] [75] [76] [77] [78] [79] Nukada, T.; Kitajima, T.; Nakahara, Y.; Ogawa, T. Carbohydr. Res. 1992, 228, 157-170. Ogawa, T.; Nakabayasi, S.; Kitajima. T. Carbohydr. Res. 1983, 114, 225-236. Ogawa, T.; Horisaki, T. Carbohydr. Res. 1983, 123, C1-C4. Iijima, H.; Ogawa, T. Carbohydr. Res. 1988, 172, 183-193. Iijima, H.; Ogawa, T. Carbohydr. Res. 1989, 186, 107-118. Nakayama, K.; Uoto, K.; Higashi, K.; Soga, T.; Kusama, T. Chem. Pharm. Bull. 1992, 40, 1718-1720. Meryala, H.B.; Lingannagaru, S.R. Tetrahedron. 1997, 53, 17501-17512. Moil, M.; Ban, Y. Chem. Pharm. Bull. 1976, 24, 1992-1999. Pietropaolo, R.; Faxaone, P.; Sergi, S.; Pietropaolo, D. J. Organomet. Chem. 1972, 42, 177-182. Tsuji, J. Organic Synthesis with Palladium Compounds, Springer Verlag, Berlin, 1980, pp. 37-50 and 125-134. Wenzel, T. T. J. Chem. Soc. Chem. Commun. 1993, 862-864. Heumann, A.; Jens, K.-J.; Rglier, M. Progress in Inorganic Chemistry, 1994, 42,483-576. Bennet, A. J.; Kitos, T. E. J. Chem. Soc, Perkin Trans. 2 2002, 1207-1222. Capon, B. Chem. Rev. 1969, 69, 407-498. Feather, M. S.; Harris,J. F.J. Org. Chem. 1965, 30, 153-157. Armstrong, 11. E.; Glover, W. H. Proc. R. Soc. London B. 1908, 80, 312-331. Edward, J. T. Chem. Ind. (London) 1955, 1102-1104. Thatcher, G. R. J ., Ed. The A nomeric Eflect and Associated Stereoelectronic Eflects; ACS Symposium Series 539; American Chemical Society: Washington, DC, 1993. Wolfe, S.; Shi, Z. Isr. J. Chem. 2000, 40, 343-355. Wolfe, S.; Rauk, A. Tel, L. M.; Csizmadia, I. G. J. Chem. Soc. B. 1971, 136-145. Bemiller, J. N. Acid-catalyzed Hydrolysis of Glycosides in Adv. Carbohydr. Chem. Wolfrom, M. L.; Tipson, R. S. Eds. New York. 1967. pp 25-108. 85 Chapter 3 Advanced Derivatives of 1, 5-Anhydro-D-Glucitol 86 Abstract Anhydroalditols and their derivatives are considered metabolically inert for lacking the C1 hydroxyl group. The glucose 6-phophate analogue 1,5-Anhydro-D-glucitol 6- phosphate is widely used as a hexokinase inhibitor in the study of enzyme mechanisms and as a probe to study carbohydrate metabolic pathways. Recently, deoxy anhydroalditols have been used in the construction of biologically important molecules, such as anhydrohexitol nucleosides, which showed activities against herpes simplex virus, Sialyl Lewisx analogues as potent E-selectin inhibitors. They have also been used in the preparation of catalysts for enantioselective cyanation of ketones. With the easily available anhydroalditols through the reductive cleavage of complex carbohydrates, these anhydroalditols and deoxy anhydroalditols can be easily prepared. From 1,5—anhydro-D- glucitol, l,5-Anhydro-D-glucitol—6-phosphate was prepared from 1,5-anhydro-D-glucitol in two steps in over 60 % yield. The important deoxy-anhydrohexitols, which are components of E-selectin inhibitors, scaffolds of anhydrohexitol nucleosides and chiral catalysts, were also prepared from 1,5-anhydro-D-glucitol efficiently. 87 3.1 Introduction 3.1.1 Glycolysis and l,S-anhydro-D-glucitol-6-phosphate Glycolysis is a catabolic pathway in the cytoplasm that is found in almost all organisms- irrespective of whether they live aerobically or anaerobically]. During glycolysis, glucose is oxidized to either lactate or pyruvate. Figure 3.1 lists the substrates and enzymes for the first several steps of glycolysis and the corresponding anhydrosugars as the substrate analogues. The first step of glycolysis is the ATP-dependent phosphorylation of glucose to form glucose 6-phosphate catalyzed by the isoenzymes known as hexokinases. The phosphorylation of glucose accomplishes two goals: First, nonionic glucose was converted into glucose 6-phosphate (G6P), an anion that is trapped in the cell, since cells lack transport systems for phosphorylated sugars. Second, the otherwise biologically inert glucose becomes activated into a labile form capable of being further metabolized. The second reaction of glycolysis is an isomerization, in which G6P is converted to fructose 6-phosphate (F6P). The enzyme catalyzing this reaction is glucose 6-phosphate isomerase. The reaction is freely reversible at normal cellular concentrations of the two hexose phosphates and thus catalyzes this interconversion during glycolytic carbon flow and during gluconeogenesis. 88 P\ 0" hexokinase O'P gulcose-S—phophate O 0 OH H O glucoklnase isomerase HO —-—D 910 11% 0 fi’ OH OH OH O 0” 0H 0H 1 2 3 ’P 0H 0 0 o = .- "iifiK/(a "1334 " ”3”: 0H 0H _ 1 .5-anhydro-D-glucitol 1 ,5-anhydro-D—glucitol 6—phosphate 4 5 Figure 3.1 Anhydrosugars as substrate analogues in carbohydrate glycolysis. Substrate analogues are useful tools in the study of enzyme catalyzed reaction mechanisms. Such analogues could help in the identification of the active substrate conformation or configuration or in the stabilization of reaction intermediates. Thus changes of the substrates during the enzyme catalyzed reactions could be identified at molecular level. Some sugar metabolizing enzymes have been studied using this methodz' 4. 1,5-Anhydro-D-glucitol 4 and 1,5-anhydro-D-glucitol 6-phosphate 5 are the analogue of D-glucose l and D-glucose-6-phosphate 2. Because 4 and 5 lack a hydroxyl group at the C1 position of the pyranose ring, they are considered to be metabolically inert. 1,5- Anhydro-D-glucitol 6-phosphate 5 is widely used as a hexokinase inhibitor in the study of enzyme mechanisms and as a probe to study carbohydrate metabolic pathwayss'w. 3.1.2 Deoxy anhydroalditols: application in construction of biologically important molecules and chiral catalysts 89 Deoxysugars are carbohydrates in which one or more of the normally occurring oxygen atoms are deleted (i.e., replaced by hydrogen atoms) or replaced by any other heteroatom or heteroatomic group, such as sulfur (thiosugars), halogen, nitrogen (aminosugars) or NO,( (nitro- and nitrososugars)I '. Many of those naturally occurring deoxysugars have been shown to serve as ligands for cell-cell interactions or as targets for toxins, antibodies, and microorganisms'z'w. The replacement of one or more hydroxyl groups in these sugars by various functionalities generally induces fundamental changes in the chemical properties of the resulting monosaccharides and, therefore, has a direct bearing on the wide range of their biological activities”. Deoxysugars are also frequently found in the secondary metabolites of microorganisms and plants, such as cardioglycosides, antibiotics, and anticancer agents”. These sugar residues play crucial roles in conferring optimal biological activity for many natural products. Their removal often results in the loss of all biological activity of the parent compounds”. Recently, another class of deoxysugars, the deoxy 1-deoxy sugars, or deoxy anhydrosugars, has been used in the construction of molecules which have important biological functionsZO'23 . 90 C02Na Sialy Lewis" 0 0“” relative 1050 (E-Sel) = 1.0 W00 0 1 .. 2 _ _ OH 6 R - Me R — H 1c50 — 0.020 0“ no 7 1::1 = H R2 = H 1050 = 0.305 R1 0H 8 R1 = Me R2 = Bz IC5O = 0.010 9 R1 = H R2 = 82 1050 = 0.090 COOH k on 0W0 go 10 R=H IC50>5 0H %J Ho 11 R =32 1c50=0290 0 OH OH OH Figure 3.2 Sialyl Lewis" analogues as potent E-selectin inhibitors. Sialyl Lewis" is a tetrasaccharide which is a weak E-selectin inhibitor (KD = 1060 uM), it is a lead structure in the identification of simplified but more-potent selectin antagonistszo'zz. Thoma and co-workers prepared the simplified Sialyl Lewis" analogues 6-11, which contain a 1,5-anhydro-2-deoxy-D-glucitol building block (Figure 3.2)20'22. The much simplified trisaccharides showed enhanced inhibitory properties against E- selectin. The most potent compound is 8, which is lOO-fold more potent than Sialyl Lewis" in the binding assay and has an ICso value of 1-2 UM in a cell-based in vitro flow assay. 0 R ""JE/ 12 R=l 15 R= J.,-é )\ 0%" 13 R=Cl S o 1‘ R=CF3 17 R: -§-cscn 15 R: g—CH2CH2 HO _ - _. : _ Ho 16 R- E C—0 CH3 Figure 3.3 Anhydrohexitol Nucleosides 91 Recently, a series of anhydrohexitol nucleosides (compounds 12-18, Figure 3.3) are prepared and tested against herpes simplex virus (HSV)23. The trifluoromethyl, vinyl, and propynyl analogues 14,. 15 and 18 showed potent activity against HSV. The selectivity index for 15 is large than 16000 against HSV-l and larger than 1000 against HSV-2. The deoxy anhydroalditols have also been used as scaffold for the construction of catalyst for enantioselective cyanosilylation of ketones. High yields and ee’s were obtained for 24,25 we, . ...{y <5 aryl ketones (Figure 3. 4) 19 O 19 0 )L + TMSCN THF. -20 c.0611= 1113ch Ph CH: 92 0/0 YIBId Ph CH; 94 % ee Figure 3.4 Deoxy anhydroalditols based chiral catalysts Although these anhydroalditols derivatives have been used extensively, no general route is available for their preparation. With the easily available anhydroalditols through our reduction method as discussed in chapter 2, these anhydroalditols derivatives can be easily prepared. 92 3.2 Preparation of 1,5-anhydro-D-glucitol-6-phosphate With 1,5-anhydro-D-glucitol in hand, 1,5-Anhydro-D-glucitol 6-phosphate was easily prepared in several steps (Figure 3.5). on 01101091112 011 C b 110 o a Pin/\Bo o H O _, Ho 0 H PIVO Pl ”V 0Plv 0H OPIV OPIV 4 20 21 22 ll 0, (0.. ii 1 0H "$3 “—8—" HH‘%L ”fir P H0 OPIV Figure 3.5 Preparation of 1,5-Anhydro-D-glucitol 6-phosphate (a) l). PhCH(OMe)2, p-TsOH, DMF; 2). PivCOCl, Pyr, 83 %, 2 steps; (b). EtOH-H2O- CF3C00H, 85%; (c). (Ph0)2P0Cl/Pyr, 62%; ((1). H2, Pt02, MeOH, 100%; (e). NaOMe, MeOH, 83%; (f). NH3, 100%. The 4,6-hydroxy groups were first protected with benzylidene by treatment of 1,5- anhydro-D-glucitol 4 with benzaldehyde dimethyl acetal in DMF. The 2 and 3 hydroxy groups were then protected with pivolyl groups to afford the fully protected 1,5-anhydro- D-glucitol derivative 20. Treatment of 20 with EtOH-H20-CF3C00H (60:8:1) gave compound 21 with 4, 6 hydroxyl groups freed. Phosphorylation of 21 using (PhO)2P(0)C1 in pyridine was selective among the primary and secondary hydroxyl groups and gave the protected phosphate ester 22. The final product 19 was obtained 93 from 22 by first removing the Ph group using H2/Pt02 and then removing the pivolayl group using NaOMe/MeOH. Because 19 is a strong acid, it was converted to its ammonia salt 24 by treatment with ammonia. o 1' 0.. 0H 01:0(01311)2 0’ \OH 1'10 0 a 1'10 0 0 H0 0 1'10 H0 1'10 0H OH OH 4 25 19 Figure 3.6 Preparation of 1,5-Anhydro-D-glucitol 6-phosphate using the direct phosphorylation method. (a) (PhO)2P0Cl/Pyr; (b) H2, Pt02, MeOH. Because (PhO)2P(0) is a sterically demanding reagent, it is expected that under controlled conditions, the selective mono-functionalization of 4 is possible. Thus, the direct transformation of 4 to 25 was attempted (Figure 3.6). Treatment of 4 with 1 equivalent of (PhO)2P(0)Cl in pyridine gave the protected 6-phosphate 25 in 60 % isolated yield. Deprotection of 25 using H2/Pt02 gave 19 directly in quantitative yield. Compared to the method in figure 3.5, this method is more straightforward, and 1,5- anhydro-D-glucitol-6-phosphate 19 was prepared in two steps in 60 % overall yield from 1,5-anhydro-D-glucitol 4. 3.3 Preparation of Deoxysugars from 1, 5 —anhydro-D-g1ucitol 3.3.1 Formation of 2,3-anhydro derivatives of 1,5-anhydro-D-glucitol 94 The 2,3-anhydro derivatives 27 and 28 can be easily prepared from 26 since the 2, 3 hydroxyl groups are anti to each other. Transforming one of the hydroxyl groups to a good leaving group followed by an intramolecular attack from the other free hydroxyl group will form the 2,3-anhydro derivatiesz°'29.' 0H PIC/m 3 Ph 09% b 27 H0 0 HO OH Fri/29% 4 25 Minor 0 28 Figure 3.7 Preparation of 4,6-benzylidene-2,3-epoxide derivatives of 1,5-anhydro-D- glucitol (a) PhCH(OMe)2, stOH, DMF, r.t. 12 h, 82 %; (b.) Ph3P, DIAD, THF, 60°C, 12 h, 72 %, 27/28 2.1/1 Reaction of 4 with benzaldehyde dimethyl acetal in DMF with catalytic amount of p- Ts0H gave 4,6-0-benzylidene protected derivative 26 in 82 % yield. Treatment of 26 under Mitsunobu conditions (Ph3P, DIAD) at room temperature for 24 hour gave no product. Raising the reaction temperature to 60°C, the reaction was done in 12 hour. Compound 27 with manno configuration was formed as the major product in 48.8 % and compound 28 with allo configuration was formed as the minor product in 23.2 % (Figure 3.7). The mechanism for formation of 127 and 28 is shown in figure 3.8. Formation of the triphenylphosphonium intermediate 29 followed by intramolecular attach by 3-0H gave 27, while formation of the triphenylphosphonium intermediate 30 at 3-0H followed by intramolecular attach by 2-0H gave 28. In intermediate 30, there is a very severe steric 95 interaction between the triphenylphosphonium ion and the 4,6-0-benzylidene ring, while in 29, this steric interaction is greatly reduced. n’?0 ——> °g°4 " ' P" 59% Meter H o O 27 +1396, Ph’voo o Ph3P,D|AD 29 Ho 0 911’ Y‘ 0” Ph’VS’A/o _. 0 ° 26 o w Minor 9 OH O Ph3Pi’ K/ 28 30 Figure 3.8 Mechanism for the formation of 2,3-anhydro derivatives under Mitsunobu conditions. (p-Tolylsulfonyl)imidazole (Ist) has also been used in the construction of 1,2 anhydro sugars from 1,2 diols in carbohydrates. With Ist, the separation of the products from the reaction mixture is easier compared to Ph3P/D1AD3°'33. For substrate 26, another advantage using Ist is that formation of epoxides 27 and 28 can be controlled by the reaction conditions. If Ist was added immediately after adding NaH (2.2 equivalent), a mixture of 27 and 28 was obtained in 67 % yield (27/28 3/1). While if 26 was treated with NaH (2.2 equivalent) for 2 hour and then adding Ist, only 27 was obtained and the yield is very high (86 %). The different results came from the formation of either monoalkoxide or dialkoxide derivative of 26. After adding NaH, the C-2 or C-3 monoalkoxide (31 or 32) was formed at first. If adding Ist at this time, a mixture of 02 or C-3 tosylate was formed and thus the final products were obtained as mixture of 27 and 28. However, if adding Ist after the dialkoxide 33 was formed, the formation of C-2 or C-3 tosylate was controlled by the 96 steric effect, and the more sterically favored C-2 tosylate was formed. Once the C-2 tosylate was formed, it immediately transformed to product 27 since the C-3 alkoxide is already formed. PH’E§%§_. lst "(29% 27 lib/29m NaH _’ Flt/YrfiA—fi lst ”(39% O 0 H OH 28 ——’ "Tea ”freq 27 28 Figure 3.9 F orrnation of 2.3-anhydro derivatives using NaH/Ist. 3.3.2 Preparation of deoxyanhydrohexitols From the 4,6-0-benzylidene-2,3-anhydro derivatives, the 2 or 3 deoxy derivatives 34 and 35 were prepared. Reduction of 27 with LAH gave compound 4,6-0-benzylidene-3- deoxy-1,5-anhydro-D-mannitol 34 exclusively in 96 % yield with the delivery of the hydride to the axial position. OH Pn/Yfifi LAH, E120 2 "(3% H2. Pd/C ”%E 7 HO r, 1 n r.t 12 h o o 100 % 27 '6 /° 34 36 o LAH, E! 0 H2. Pd/C H Ph 0% 2 : Ph 00 o —-—> "o O r.t 12 h o r.t1 h 95 % 10° % 0H 28 35 37 97 Figure 3.10 LAH reduction of 2,3-anhydro derivatives to form deoxy anhydrohexitols Similarly, 1,5-anhydro-4,6-O-benzylidene-2-deoxy-D-allitol 35 was prepared in 95 % yield from 28 (Figure 3. 10). Deprotection of the protecting group in 34 and 35 gave the free deoxy anhydrohexitols 36 and 37 in quantitative yield. 3.4 Conclusion Important anhydroalditol derivatives were prepared efficiently from the anhydroalditols. 1,5-Anhydro-D-glucitol —6-phosphate, the inert analogue of D-glucose-6-phosphate, was prepared from 1,5-anhydro-D-glucitol in two steps in over 60 % yield. The important deoxy-anhydrohexitols, which are components of E-selectin inhibitors, scaffolds of anhydrohexitol nucleosides and chiral catalysts, were also prepared from 1,5-anhydro-D- glucitol efficiently. 98 3.5 Experimental General Procedures lH, '3 C NMR spectra were recorded at 500, 125, 121 MHz, respectively, with a Varian instrument at 293 K. The chemical shifts are given in ppm using CDC13 residue as reference (57.24p) for 1H and relative to the central CDCl3 resonance (5: 77.00p) for 13C NMR unless otherwise specified. 31P NMR chemical shifts are given in ppm with H3PO4 as an external reference. 1H and '3C are assigned on the basis of 2D lH COSY and 1H- '3 C chemical-shift correlated experiments. Melting points were determined on a melting point apparatus (uncorrected). Optical rotations were measured on a Jasco P1010 polarimeter at 20°C. IR spectra (wave numbers in cm") were recorded on a FT IR Nicolet 740 spectrometer in CHC13 solutions or KBr pellets. All chemicals were purchased from Aldrich Chemical Co. and used without further purification. 1,5-Anhydro-2,3-dipivolyl-4,6-O-benzylidene-D-glucitol (20) To a solution of 1,5-anhydro-glucitol 4 (0.984g, 6.0mmol) in anhydrous DMF, were added benzaldehyde dimethyl acetal( 1.65ml, 12.0mmol), sulfuric acid(98%, 45ul). The mixture was stirred overnight under N2 atmosphere. After adding anhydrous pyridine (1.0ml) and stirring for 10 minutes, methanol was removed under reduced pressure. The mixture was cooled to 0°C, then anhydrous pyridine (9.7ml, 120mmol), and pivaloyl chloride (2.22ml, 60mmol) were added. After stirring at room temperature for 24hr, water and chloroform were added, the organic layer was separated, and the aqueous layer was dried over anhydrous Na2804. After remove the solvent, the residue was crystallized from Hexane-Ethyl acetate to give 20 (2.1g, 83%): [(11020 +192 (c l, CHC13); lH NMR 99 (500 MHz, CDC13): 6 l.14(s, 9H, tBu); 1.16(s, 9H, tBu), 3.36(dd, 1H, J1=J2=10.7 Hz, H- l-b), 3.47(dd, 1H, J=9.7 Hz, 4.9 Hz, H-6-b); 3.65(dd, 1H, J1=J2=9.7 Hz, H-4), 3.69(dd, 1H, J1=J2=10.4 Hz, H-6-a), 4.09(dd, 1H, J=8.4 Hz, 5.9 Hz, H-l-a), 4.33(dd, 1H, J=7.7 Hz, 5.0 Hz, H-5), 5.03(ddd, 1H, J1=J2=8.2Hz, J3=5.6Hz, H-2), 5.35(dd, 1H, J=9.7Hz, 9.7Hz, H-3), 5.50(s, 1H, PhCHO), 7.40(m, 5H, H-arom); 13C NMR: 27.1, 38.8, 67.5(C- 1), 68.6(C-5), 69.3(C-2), 71.3(C-4), 71.9(C-3), 79.1(C-6), 101.0, 125.8, 1289.1, 128.8, 136.9, 177.2,177.4; omax (KBr) 3072, 2973, 2935, 2873, 1731, 1602, 1585, 1481, 1454, 1284, 1168, 1101, 709. Anal. Calcd for C23H3207: C, 65.70; H, 7.67. Found: C, 65.50; H, 7.49 1,S-Anhydro-2,3-dipivolyl-D-glucitol (21) A solution of 20 (1.68g, 4.0mmol) in EtOH-H20-CF3COOH (60:8:1, 50ml) was stirred at room temperature for 24hr. The mixture was concentrated. Column chromatography of the residue on silica gel using EtOAC-Hexane 1:1 as eluant afforded the desired product 1,5-anhydro-2,3-dipivolyl-glucitol 21 (1.11 g, 85%): mp 84-85°C; MD” +493 (0 1, CHC13); 1H NMR (500 MHz, CDC13): 6 1.13(s, 9H); 1.17(s, 9H), 3.26(dd, J=10.5 Hz, 1H, H-l-a), 3.33(ddd, J1=12=9.6 Hz, J3=4.5 Hz, 1H, H-5), 3.65(dd, J 1=J2=9.0 Hz, 1H, H-1-4), 3.78(dd, J=4.5 Hz, 12.0 Hz, .1 H, H-6-b), 3.90(dd, J=3.0 Hz, 12.0 Hz, 1H, H-6-a), 4.05(dd, J=5.4 Hz, 10.8 Hz, 1H, H-l-b), 4.94(ddd, J 1=JZ=11.4 Hz, J3=5.7, 1H, H-2), 5.04(dd, J1=J2=9.6Hz, 1H, H-3); 13C: 27.1, 38.8, 39.0, 62.2, 66.8, 68.8, 69.9, 76.6, 80.4; omax (KBr) 3380, 2976, 2973, 2873, 1739, 1286, 1174, 1153; Anal. Calcd for C16H2807: C, 57.82; H, 8.49. Found: C, 67.30; H, 8.10. 100 1,5-Anhydro-2,3-dipivolyl-6-diphenylphosphate-D-glucitol (22) To a solution of 21 (0.664g, 2.0mmol) in CHzClz, pyridine (0.9ml) and diphenylchlorophosphate (0.62ml, 3.0mmol) were added at 0°C. After stirring at room temperature for 1.5hr, water and CHzClz were added. The organic layer was separated, and the aqueous layer was extracted with CHzClz. The combined organic layer was washed with 1M HCl, saturated NaHCO3, and water, then dried over anhydrous NaZSO4. After remove the solvent under reduced pressure and purify by column chromatography on silica gel using CH2C12-CH3OH(13:1) as an eluant afforded the product 22 ( 0.701g, 62%): mp 75-76 °C; [011.320 +123 (c 1. CHC13); ‘H NMR (500 MHz,CDCl3):51.11(s, 9H, tBu); l.14(s, 9H. tBu), 3.18(dd, J1=12=10.2 Hz, 1H, H-l-b), 3.37(m, 2H, H-4,5), 3.97(dd, J 1=10.8 Hz, JZ=J3=5.4 Hz, 1H, H-l-a), 4.42(dd, J 1=J2=11.7 Hz, 1H, H-6-b), 4.50(ddd, J 1 =12 =9.8 Hz, J3, 5 =3.5 Hz, H-6-a), 4.76(ddd, J l=10.2 Hz, J2=J3=5.7 Hz, 1H, H-2), 5.06(dd, J=9.3Hz, 1H, H-3), 7.25(m, 10H, H-arom); l3C NMR (CDC13): 526.9, 38.5, 38.6, 66.4(C-1), 67.4(C-6), 67.9(C-5), 68.5(C-2), 75.0(C-3), 78.9(C-4), 120.1, 125.6, 129.0, 150.1, 177.1, 178.1; 3'P NMR (CDCl3): 6 -9.77; umax (KBr) 3399, 2971, 2873, 1735, 1590, 1487, 1280, 1184, 1166, 1056, 962, 773, 690; Anal. Calcd for C28H37010P: C, 59.57; H, 6.61. Found: C, 59.27; H, 6.10. l,S-Anhydro-2,3-dipivolyl-6-phosphate-D-glucitol (23) To a solution of 22 (0.564g, 1.0mmol) in methanol, Pt02 (0.113g, 0.50mmol) was added. After stirring vigorously at room temperature under H2 atmosphere for 6 hr, the solid was removed by filtration. The product 23 was obtained after removing the solvent under reduced pressure as syrup (0.412g, 100%): 1H NMR (500 MHz, CDC13): 5 1.03(s, 9H, 101 tBu); 1.08(s, 9H, tBu), 3.30(dd, 11:12:10.2 Hz, 1H, H-lb), 3.45(m, 1H, H-5), 3.67(dd, J1=JZ=9.8 Hz, 1H, H-4), 4.03(dd, J1=JZ=11.0 Hz, 6.1 Hz, 1H, H-6b), 4.20(m, 2H, H-la, H-6a), 4.88(ddd, 11:-9.0 Hz, 12=J3=6.1 Hz, 1H, H-2), 5.16(dd,J1=12=9.8Hz, 1H, H-3); l3C NMR: 8 26.8, 38.5, 38.6, 65.3, 66.3, 68.0, 68.8, 75.1, 79.0, 177.4, 178.2; 3'P NMR (CD3CN): 5 1.97 1,S-Anhydro-D-glucitol-6-phosphate (5) To a solution of 23 (0.247 g, 0.60 mmol) in methanol, 95% powered NaOMe (65.0 mg, 1.2 mmol) was added. After stirring at 40°C for 10 hr, the solution was passed through a cation ion-exchange resin to remove the salt. Removal of the solvent gave the free acid 5 as a syrup (0.123 g, 83 %). [(1)020 +295 (c 1, D20); lH NMR(D20): 8 2.85(dd, 11:12:95 Hz, 1H, H-l-b); 3.02(m, 2H, H-l-a, H-S), 3.04(m, 1H, H-4; 3.16(m, 1H, H-3), 3.53(dd, J 1=5.7 Hz, 12=11.0 Hz, 1H, H-2), 3.69(ddd, J1=9.0 Hz, JZ=J3=4.5 Hz, 1H, H-6-b), 3.80(ddd, J1=9.0Hz, J2=7.1Hz, J3=3.9 Hz, 1H, H-6-a); l3C NMR(D20): 8 65.2, 68.8, 69.9, 69.1, 77.2, 78.5; 3'P NMR (D20): 0.96; 0mm (KBr) 3382, 2925, 2898, 1203, 1097, 1051; l,S-Anhydro-D-glucitol-6-phosphate monoammonium salt (24) In the preparation of 5, the elute from cation ion-exchange was condensation to about 2 ml, ammonia (0.5 M in 1,4-dioxane, 10ml) was then added to the solution. After stirring at room temperature for 1hr, the product was precipitated as white solid. Filtration gave 24 in quantitative yield: mp. 105-107°C; 'H NMR(D20): 8 3.10(t, J=10.9Hz, H-l-b), 3.20(m, 1H, H-2), 3.27(d, J=9.0Hz, H-3 or H-4), 3.33(d, J=9.3Hz, H-3 or H-4), 3.42(m, 102 1H, H-5), 3.76-3.81(m, 3H, H-6-a, H-6-b, H-l-a); l3c NMR(D20): 8 63.8 (C-6), 67.9(C- 1), 68.1(C-3 or C-4), 68.3(C-3 or G4), 76.3(C-2), 77.9(c-5); 3'? NMR(DZO):4.30;1)mam (KBr) 3411, 3222, 2919, 2867, 1631, 1459, 1097, 1054. 1,5-anhydro-D-Glucitol-6-diphenolphosphate (25) To a solution of 4 (0.328g, 2.0 mmol) in dry pyridine (5m1) at 0°C diphenylchlorophosphate (0.43 ml, 2.1 mmol) was drop-wise added. After stirring at 0°C for 1.5 h and then 0.5 h at room temperature, MeOH (2.0ml) was added to quench the reaction. Removal of the solvent followed by flash chromatography (10:1 CHC13: MeOH) afford 25 as a crystal (0.46g, 60%): mp 113-114°C; [61020 +221 (c 1, CH3OH); ‘H NMR(DMSO): 8 2.99(t, J=9.6Hz, H-l-b), 3.06(t, J=8.8Hz, H-l-a), 3.12(t, J=8.8Hz, H-4), 3.22-3.30(m, 2H, H3 and H5), 3.71(dd, J 1=10.8Hz, 12:4.9Hz, 1H, H-2), 4.24(m, 1H, H- 6-b), 4.47(m, 1H, H-6-a), 4.96(d, J=3.6Hz, 1H, OH), 5.02(sb, 1H, OH), 5.20(d, J=4.9Hz, 1H, OH), 7.24(m, 5H, H-arom), 7.42(m, 4H, H-arom); l3C NMR(DMSO): 5 64.7(C-6), 65.2(3 C. C-1, C-2, C-3), 73.6(C-4), 74.4(C-5), 115.7(C-arom), 121.3(C-arom), 125.8(C- arom), 145.7(C-arom); 3'? NMR (DMSO): -10.65; umax (KBr) 3382, 3282, 3066, 2971, 2904, 2877, 2852, 1590, 1488, 1454, 1390, 1253, 1224, 1190, 1101, 1083, 1043, 943, 771, 688, 520 cm'I ; Anal. Calcd for C18H2108P: C, 54.55; H, 5.34. Found: C, 54.21; H, 5.12. Formation of epoxides 27, 28 from 4,6—O-benzylidene-1,5-anhydro-D-glucitol: NaH-Ist method: 103 In a 100ml flask, Sodium hydride (50% in mineral oil, 0.525 g, 10.5 mmol) was washed free of oil with hexane, anhydrous DMF (20 ml) was added followed by addition of 4,6- O-benzyliden-l,5-anhydro-D-glucitol (26) (1.01g, 4.0 mmol). After stirring at r.t. under N2 for 0.5 h, Ist (1.16 g, 5.24 mmol, in 5 ml DMF) was added. The reaction mixture was let to stir for another 4 h at r.t., and then EtOAc (30 m1) and brine (30 ml) was added. After separation, the aqueous phase was extracted with EtOAc (2x30ml), the combined organic phase washed with water (30ml), brine(3‘0m1), and dried over anhydrous MgSO4. Evaporation of the solvent under reduced pressure gave a syrup. Flash column (Hexane/EtOAc 5/1) gave 27 (0.337 g, 46.5 %) and 28 (0.160 g, 22.1 %). 1,5:2,3-Dianhydro-4,6-O-benzylidene-D-mannitol (27) [61920 -1932 (c 1, CHC13); ‘H NMR (500 MHz, CDC13) 8 3.40(m, 1H, H-2); 3.54 (d, J =- 4.4 Hz, 1H, H-3), 3.63 (td, J = 10.3, 1.5 Hz, 1H, H-6’), 3.72 (ddd, J = 9.8, 4.9, 1.5 Hz, 1H, H-S), 4.00 (d, J = 9.8 Hz, H'-4), 4.06 (m, 2H, H-l, H-l’), 4.21 (dd, J = 10.3, 4.8Hz, 1H, H-6), 5.57 (s, 1H, PhCI-I),-7.36 (m, 3H, Ph-H), 7.50 (m, 2H, Ph-H); 13c NMR (125 M Hz, CDC13) 8 51.0 (02); 53.1 (03), 64.4 (C-l or OS), 64.5 (01 or 05), 68.8 (C-6), 78.1 (04), 102.5 (PhCH), 126.2, 128.2, 129.1, 137.1; Anal. Calcd for C13H1404: C, 66.66; H, 6.02. Found: C, 66.54; H, 6.10. 1,5:2,3-Dianhydro-4,6-O-benzylidene-D-Allitol (28) [(1][)20 -4.22 (c 1, CHC13); 1H NMR (500 MHz, CDC13) 8 3.14 (td, J = 9.8, 4.7Hz, 1H, H- 5), 3.19(dd, J = 4.0, 1.2 Hz, 1H, H-2), 3.48(d, J = 4.0Hz, 1H, H-3), 3.66 (d, J = 10.0Hz, 1H, H-4), 3.68 (t, J = 10.3Hz, 1H, H-6), 3.91 (dd, J = 13.7, 1.3 Hz, lH,H-1’), 4.21 (d, J = 104 13.7 Hz, 1H, H-l), 4.27 (dd, J= 10.5, 4.6Hz, 1H, H-6), 5.56 (s, 1H, PhCH), 7.38 (m, 3H, Ph-H),7.50(m,2H,Ph-H);13C NMR (125M Hz, CDC13) 8 49.8 (C-2), 53.3 (03), 65.2 (C-l), 69.2 (OS), 69.3 (C-6), 75.3 (04), 102.1 (PhCH), 126.0, 128.2,129.1, 137.0; Anal. Calcd for C13H1404: C, 66.66; H, 6.02. Found: C, 66.40; H, 5.92. 1,5-Anhydro-3-deoxy-4,6-O-benzylidene-D-arabino-hexitol (34) To a stirred solution of 27 (0.117 g, 0.5 mmol) in 320 (5 ml) was added LAH (37 mg, 1.0 mmol). After stirring under nitrogen atmosphere for 4 h, ethanol (1 ml) was added. Stirring was continued for another 30 min. Filtration followed by removal of the solvent gave the crude product as a syrup. Column chromatography (CH2C12:MeOH 30:1) gave the pure 34 (113 mg, 96 %): [61920 -30 (c 1, CHCl3); m.p 101-103°C; ‘H NMR (500 MHz, CDC13) 5 7.34-7.51 (m, 5 H, Ar-H), 5.56 (s, 1H, PhCH), 4.31-4.27 (m, 1H), 3.98- 3.95 (m, 1 H), 3.89-3.83 (m, 1H), 3.70 (t, J = 10.2 Hz, 1H), 3.57-3.52 (m, 1H), 3.42 (t, J = 10.2 Hz, 1H), 3.40-3.31 (m, 1H), 2.65 (br s, 1H, OH), 2.03-1.98 (m, 1H), 1.84-1.75 (m, 1H); 137.3, 129.2, 128.3, 126.2, 101.9, 83.9, 71.1, 69.5, 68.8, 66.3, 33.2; Anal. Calcd for C13H1604: C, 66.09, H, 6.83. Found C: 66.01; H, 6.89. 1,5-Anhydro-2-deoxy-4,6-O-benzylidene—D-ribo-hexitol (35) Using the same procedure as for the preparation of 34 from 27, 35 was obtained from 28 in 95 %. [61920 - 23.7 (c 1, CHC13); 'H NMR (500 MHz, CDC13) 8 1.86 (ddd, J = 3.1, 11.8, 13.0 Hz, 1H, H-2), 2.12 (br s, 1H, OH), 2.33 (m, 1H, H-2’), 3.42 (ddd, J = 5.0, 9.2, 10.1 Hz, 1H, H-6), 3.71 (dd, J = 1.5, 2.4 Hz, 1H), 3.81 (t, J = 10.3 Hz, 1H), 3.96 (ddd, J = 1.7, 2.1, 12.4 Hz, 1H), 4.04 (ddd, J = 4.6, 9.2, 11.9 Hz, 1H), 4.14 Our 5, 1H), 4.33 (dd, J = 105 4.9, 10.4 Hz, 1H, H-4), 5.63 (s, 1H), 7.38-7.54 (m, 5H, Ar-H); l3C NMR (125M Hz, CDC13) 835.6, 67.0, 69.0, 72.5, 74.2, 74.3, 101.9, 126.1, 128.3, 129.0, 137.4; Anal. Calcd for C13H1604: C, 66.09, H, 6.83. Found C: 66.05; H, 6.78. 1,S-Anhydro-Z-deoxy-D-ribo-hexitol (37) A solution of 35 (100 mg, 0.42 mmol) in methanol (3 ml) with Pd/C (10 %, 40 mg) was stirred under hydrogen atmosphere over night. After filtration through celite, the solvent was removed under vacuum. Water (1 ml) was added to the flask and the solution was washed with toluene (2 x 1 m1). Removal of the solvent gave 37 in quantitative yield (62 mg, 100 %); 1H NMR (500 MHz, D20) 5 3.97—3.95 (m, 1H), 3.79 (dd, J = 2.4, 12.2 Hz, 1H), 3.72 (td, J = 2.1, 12.2 Hz, 1H), 3.68 (dddd, J = 0.5, 4.8, 6.8, 9.6 Hz, 1H), 3.56 (dd, J = 6.8, 12.2 Hz, 1H), 3.48 (dd, J = 0.5, 12.5 Hz, 1H), 3.17 (ddd, J = 2.2, 6.9, 9.6 Hz, 1H), 2.09 (m, 1H), 1.59 (ddd, J = 3.2, 11.4, 14.6 Hz, 1H); 13C NMR (125M Hz, CDC13)5 82.2, 71.0, 66.3, 62.5, 61.5, 37.5; Anal. Calcd for C6H1204: C, 48.64, H, 8.16. Found C: 48.52; H, 8.09. 106 3.6 References [1] Koolman, J.; Roehm, K. H. Color Atlas of Biochemistry, 2004, 2nd ed. Pp150- 160 [2] Yamanouchi, T.; Tachibana, Y.; Sekino, N.; Akanuma, H.; Akaoka, I.; Miyashita, H. J. Biol. Chem. 1994, 269, 9664. [3] Yamanouchi, T.; Akaoka, 1.; Akanuma, Y.; Akanuma, H.; Miyashita, H.. Am. J. Physiol. 1990, 258, E423. [4] Pitkaen, E.; Pitkaen, O.M. Pflugers Arch. 1992, 420, 367— 375. [5] Ferrari, R.A.; Mandelstam, P.; Crane, R.K. Arch. Biochem. Biophys. 1959, 80, 372. [6] Losow, D. P.; Rose., 1. A. J. Biol. Chem. 1968, 243, 3623. [7] Fang, T. Y.; Alcchina, O.; Aldshin. A. E.; Fromm, H. J.; Honzatko, R.B. J. Biol Chem. 1998, 273, 19548. [8] Shiga, Y.; Mizuno, H.; Akanuma, H. J. Bacteriol. 1993, 175, 7138. [9] Yamanouchi, T.; Inoue, T.; Ichiyanagi, K.; Sakai, T.; Ogata, N. Biochim. Biophys. Acta. 2003, 1623, 82. [10] Yamanouchi, T.; Ogata, N.; Yoshimura, T.; Inoue, T.; Ogata, E.; Kawasaki, T.; Kashiwabara, A.; Muraoka, H. Biochim. Biophys. Acta. 2000, 1474, 291. [11] Kirschning, A.; Bechthold, A. F. W.; Rohr. J. Chemical and Biochemical Aspects of Deoxysugars and Deoxysugar Oligosaccharides. 1n Topics in Current Chemistry. Rohr, J. (eds) 1997, I88, pp1-84. [12] von Ahsen, U.; Noller, H. F. Science 1993, 260, 1500. [13] Hendrix, M.; Alper, P. B.; Priestley, E. S.; Wong, C. H. Angew. Chem. Int. Ed. Engl. 1997, 36, 95. [14] Van Dyke, M.; Dervan, P. B. Biochemistry 1983, 22, 2373. [15] Tronchet, J. M. J.; Tronchet, J. F. Helv. Chim. Acta 1981, 64, 425. [16] Varki, A. Glycobiology 1993, 3, 97. [17] Hecht, S. M. ed. Bioorganic Chem-istry: Carbohydrates. 1999, New York:Oxford Univ. Press. [18] Weymouth—Wilson A. C. Nat. Prod. Rep. 1997, I4. 99.. 107 [19] Hecht S. M. Ed. Bioorganic Chemistry: Carbohydrates. New York, 1999. [20] Thoma, G.; Kinzy, W.; Bruns, C.; Patton, J. T.; Magnani, J. L.; Btmnteli, R. J. Med. Chem. 1999, 42, 4909. [21] Hanessian, S.; Mascitti, V.; Rogel, O. J. Org. Chem. 2002, 67, 3346. [22] Thoma, G.; Schwarzenbach F. Helv Chim Acta, 2003, 86, 855. [23] Ostrowski, T.; Wroblowski, B.; Busson, R.; Rozenski, J.; De Clercq, E.; Bennett, M. S.; Champness, J. N.; Summers, W. C.; Sanderson, M. R.; Herdewijn, P. J. Med. Chem. 1998, 41, 4343. [24] Hamashima, Y.; Kanai, M.; Shibasaki, M. Tetrahedron Lett. 2001, 42, 691. [25] Masumoto, S.; Yabu, K.; Kanaia, M..: Shibasakia, M. Tetrahedron Lett. 2002, 43, 2919. [26] Ermolenko, M. S.; Shekharam, T.; Lukacs, G.; Potier, P. Tetrahedron Lett. 1995, 36, 2461-2464. [27] Paquette, L. A.; Arbit, R. M.; Funel, J.-A.; Bolshakov, 8. Synthesis, 2002, 14, 2105-2109. [28] Baer, H. H.; Mekarska, M.; Bouchard, F. Carbohydr. Res. 1985, 136, 335-45. [29] Sunay, U.; Fraser-Reid, B. Tetrahedron Lett. 1986, 27, 5335-533 8. [30] Sato, K.; Miyama, D.; Akai, S. Tetrahedron Lett. 2004, 45, 1523—1525. [31] Keck, G. E.; Kachensky, D. F. J. Org. Chem. 1986, 51, 2487-2493. [32] Magnusson, G.; Ahlfors, S.; Dahmn, J.; Jansson, K.; Nilsson, U.; Noori, G.; Stenval, K.; Tjornebot, A. J. Org. Chem. 1990, 55, 3932-3946. [33] Pasetto, P.; Franck, R. W. J. Org. Chem. 2003, 68, 8042-8060. 108 Chapter 4 Iodo Derivatives of Advanced Carbohydrate Intermediates 109 Abstract Using hydriodic acid as an economic and general reagent, the general 4-carbon synthons derived from carbohydrates, (S)-3-hydroxy-butyrolactone and (S)-3-hydroxy- tetrahydrofuran, were successfully transformed to the acyclic iodo derivatives. From (S)- 3-hydroxy-tetrahydrofuran, all the mono and diiodo derivatives were prepared selectively by taking advantage of the electron withdrawing property of the 3-hydroxy group and controlling the neighboring group effect of the 3-acetyl group. The application of these iodo derivatives was demonstrated in the preparation (S)-3-hydroxyl cylopentane, (S)-3- hydroxyl tetrahydrothiphene and (S)-3-hydroxyl-pyrolidien derivatives. 110 4.1 Introduction Organic halides, especially organic iodides, are important intermediates in organic synthesis; they are often involved in the formation of C-C bond via radical1 or ionic reactionsz. They are also important indispensable intermediates in substitution reactions, rearrangement reaction and elimination reactions3’4’5. Iodides are the most reactive among halides; therefore, many methods have been developed for the preparation of organic iodides“8 . The most used method is converting the corresponding alcohol to the iodides, and a number of methods for this transformation have been reported. The classical method is through a two-step procedure — the alcohol is first converted to the tosylate and then a SNZ substitution with iodide ion gives the organic iodideg. Recently, many new methods have been reported, such as BF3-EtzO/KI'0, P.;/12”, Clst-DMF/KIIZ, Mg12'3, Me3SiCl/Na1”, Ph3P/12'5, Ph3P/DDQ/R4N+I' ‘6. The reported methods suffer from one or the other problems. Using BF3-EtzO/KI, Only allylic or benzylic alcohols can be used as substrates"). Other methods involve the usage of expensive reagents (PdClz/Et3SiH)”, give low yields (lg/petroleum ether)”, need long reaction time (Clst-DMF/KI)8, and require tedious workup procedures (Ph3P/DDQ/R4N+I')‘6. Using hydriodic acid as an economic and general reagent, we successfully transformed the general 4-carbon synthons derived from carbohydrates, (S)-3-hydroxy-butyrolactone and (8)-3-hydroxy-tetrahydrofuran'9’20, to the acyclic iodo derivatives. The application of 111 these iodide derivatives was demonstrated in the preparation of the S and N containing heterocycles. 4.2 Preparation of iodo derivatives from (S)-3-hydroxy-butyrolactone The ring-opening reaction of (S)-3-hydroxy-butyrolactone l with HI seemed to be very straight forward: under acidic conditions, iodide attacks C-4 position to give the acyclic 4-iodo product 3 (Figure 4.1). Figure 4.1 Proposed reaction of (8)-3-hydroxy-butyrolactone with HI Reaction of (S)-3-hydroxy-butyrolactone l with HI was very slow when the reaction was run in water. No reaction product was observed after treating compound 1 with 5 equivalent of H1 in water for 24 h. To accelerate the reaction, the free hydroxyl group was acetylated. It is known that acyl groups can participate in reactions involving carbocations through formation of 1,3- dioxonium ions and thus stabilize the carbocation formedmz. For acetylated compound 1, with the assistance of the acetyl group, under acidic conditions, the ring opening reaction of 1 would be much faster. 112 To avoid the hydrolysis of the acetate, the reaction was run in AcOI-I/AczO. The results are shown in Table 4.1 (Figure 4.2) Table 4.1 Ring-opening reactions of (S)-3-hydroxy-butyrolactone with H1. in AcOH/AczO Iodo Product Entry AczO (%) Tlme (h) 4 (%) l 50 20 0 2 50 44 < 1 3 50 68 1.9 4 5 20 21.5 5 5 44 35.0 6 5 68 40.0 * 5 equiv HI **AczO concentration in AcOH No product was observed after treating (S)-3-hydroxy1-butyrolactone 1 with 5 equiv H1 in acetic acid containing 50 % AczO. Even after 68 h, only 1.9 % of the desired product 4 was observed. The yield of 4 increased with the decreased concentration of AczO. With only 5 % AczO in acetic acid solution, 21.5 % of 4 was formed after 20 h, after 68 h, 40 % of 4 was formed. The increased yield of product 4 with the decreased concentration of acetic anhydride is related to the dissociation of H1 in the solution. H1 is a weaker acid in acetic anhydride than in acetic acid. With the increasing concentration of acetic anhydride, less free iodide and proton is available and thus the rate for the ring-opening reaction of 5 decreased. It is 113 expected then that with no AczO in the reaction system, the iodide product should be formed much faster. Table 4.2 listed the results for ring-opening reactions of (S)-2-hydroxyl-butyrolactone 1 with H1 in AcOH without AczO. Table 4.2 Ring-opening reactions of (8)-3-hydroxy1-butyrolactone with H1 in AcOH Temp Time Iodo product my (’0 (h) 4(%> 1 25 0.5 16.5 2 25 6 58.1 3 25 22 61.5 4 25 48 10 The reaction rate was highly accelerated without AczO. After only 0.5 h, 16.5 % of the desired iodide product 4 was formed, 61.5 % of 4 was formed after 22 h. (ofo HI In AcOH '/\/\[]/°H a 5M: 0 Figure 4.2 Neighboring acetyl group assisted ring opening reaction of (S)-2-hydroxy-butyrolactone with H1 114 However, the yield of the iodide product cannot be further improved by simply prolonging the reaction time. Product 4 was obtained in only 10 % yield after stirring the reaction solution at room temperature for 48 h (Figure 4.3). Two new products were formed. The but-3-enoic acid 9 was formed in 5 % and 3-iodo butanoic acid 8 was formed in 85 % yield. Compound 8 is optically inactive. 5A0 O I O 4 8 l .. 1" A on \ OH 9 4 Figure 4.3 Formation of optically inactive 4-iodide butanoic acid It is known that iodide can reduce organic iodo compounds”. In this case, once the 4- iodo product 4 was formed, the free iodide in the reaction system attacks the primary iodide. Elimination of the 3-acetate group gave the olefin 9 (Figure 4.3). Addition of H1 to 9 formed the secondary iodo compound 3-iodo butanoic acid 8. The optical activity was lost in the elimination step. Without excess iodide ion in the system, the byproducts would be greatly diminished. Thus, when the reaction was run with only 1 equivalent of H1 for every equivalent of lactone, no elimination product 8 or 9 was observed. 115 Acé 0 o 8 HI in AcOH 4 5 ———> oli OH \ 0H 1 W + W 1 o 0 a 9 Figure 4.4 Reaction of (S)-3-hydroxy-butyrolactone with H1 in acetic acid at various temperatures Table 4.3 Reaction of (S)-3—hydroxy-butyrolactone with H1 in acetic acid‘ Temp Time Products (%) Entry (°C) (h) 1 4 5 8 + 9 1 4 12 6 20 74 0 2 4 36 4.2 59.5 34.1 2.2 3 30 4 0 2.4 97.6 0 4 30 6 0 70.6 29.4 5 30 12 0 81.0 16.4 2.6 6 30 22 0 90.5 4.7 4.8 7 40 6 0 70.6 22.0 7.4 8 4O 22 0 73.5 15.1 11.4 9 60 6 O 89.5 0 10.5 10 60 22 0 47.6 0 52.4 * 1 equivalent of H1 used. At lower temperature (4°C), the desired iodo product 4 was formed in about 60 % after 36 h (Table 4.3, entry 2), along with 34 % of the cyclic lactones (1 + 5), and small amount (2.2 %) of the elimination products (Table 4.3, entry 2). At higher temperature 116 (30, 40, and 60 °C), product 4 was formed in more than 70 % yield after 6 h (Table 4.3, entry 4, 7, 9). However, substantial amount of elimination products were formed when the reaction was run at 40 °C and 60°C, especially with longer reaction time (Table 4.3, entry 8, 10). The best results were obtained when the reaction was run at 30°C for 22 h. The desired iodo product 4 was obtained in 90.5 % yield with a fraction of elimination products (4.8 %) and acelyted starting lactone (4.5 %) left (Table 4.3, entry 6). The optical purity of compound 4 was greater than 98 % determined by optical rotation. 4.3 Preparation of iodo derivatives by ring opening of (S)-3-hydroxyl- tetrahydrofuran with H] After successful preparation of (8)-3-acetoxy-4-iodobutanoic acid 4 through the ring opening reaction of (S)-3-hydroxy-butyrolactone with HI, (S)-3-hydroxyl- tetrahydrofuran was used as the substrate for the ring opening reaction with HI. Unlike the lactone l, which can only cleave the C4-O bond in the ring opening reactions and thus can only get the 4-iodo product, the cyclic ether 10 has 2 possible sites for iodination and thus 3 iodo products can be obtained: the monoiodo products 11, 12 and the diiodo product 13. 117 10 HI i eon :011 :9" .fLen Hrs—FL. ._/_\_. 11 12 13 Figure 4.5 Possible iodo products from ring opening reaction of (S)-3-hydroxy- tetrahydrofuran with HI Water as solvent When water was used as solvent, the mono iodide product 11 was obtained. However, the yields varied if the reaction was run in an open flask. Two factors can affect the yield in this reaction: light and oxygen. Light can catalyze the formation of iodine from HI; oxygen can oxidize HI to iodine.24 Table 4.4 listed the light and oxygen effects on the ring opening reaction of (S)-3-hydroxyl- tetrahydrofuran 10 with HI. 3°” 1.2 oqulv HI :0“ 0 H20, 60°C ' 5 l—'/__\—OH 0 Conditions 10 1 1 Figure 4.6 Ring opening reaction of (S)-3-hydroxy- tetrahydrofuran with H1 in water 118 Table 4.4 Light and oxygen effects on the ring opening reaction of (S)-3-hydroxy1- tetrahydrofuran with HI . Yield (%) Entry condit1ons 1.5 h 3 h 6 h 1 - 35.5 43.5 25 2 hv 44.1 40.2 15.6 3 02 93.5 68.0 18.6 4 hv + 02 95.2 78.1 13.0 These results indicate that light and oxygen can accelerate the reaction. Without light or oxygen, the highest yield obtained was 43 %, with light slightly higher yield was obtained, while with oxygen, 93 % of the iodide compound was obtained in 1.5 h. The effects of light and oxygen were additive: with both light and oxygen, 95 % of the iodo product was obtained. It is noted that during the first 1.5 h of the reaction process, the color of the reaction solution (which came from H13) increased in the order of Without light/oxygen < light < oxygen < light and oxygen the same order as for the formation of the iodide compound. It has been reported that iodine can catalyze the ring opening polymerization of THFZS, a similar iodine catalyzed ring opening iodination of (S)-3-hydroxy- tetrahydrofuran was proposed in Figure 4.7. 119 9H 9H '1' fl '—_’ I OH / :. OOH—L. @0 14 11 \ OH 9” a , l 5 1" _/—\__ 5+ ——-> HO 1 9 L I. l'—'—l 14 12 Figure 4.7 Mechanism for iodine catalyzed ring opening iodination of (S)-3-hydroxy- tetrahydrofuran Reaction of H1 with oxygen forms iodine, light can catalyze this process. Coordination of iodine with the ring oxygen forms the iodine complex. Iodide can attack the complex in two possible pathways: from path A iodide attacks C-5 to form the 4-iodo 1,2 diol product 11, from path B iodide attacks C-2 to form the 4-iodo 1,3-diol product 12. The exclusive formation of 1,2 diol product 11 is understandable as the electron withdrawing 2-hydroxyl group makes C2 of 14 less reactive toward the nucleophile iodide compared to C5 of 14, and thus the cleavage of C5-O bond is much easier than that of C2-O bond. Acetic acid as solvent If the HI mediated ring opening iodination of (S)-3-hydroxy- tetrahydrofuran was run in acetic acid, it is expected that the acetyl group at C-3 position would participate in the ring opening process to form the five membered 1,3-dioxolanium followed by iodide attacking C-l to form the 4-iodo 1,3-diol derivative 16 as shown in Figure 4.8. 120 10 17 18 16 Figure 4.8 Proposed reaction of (S)-3-hydroxyl- tetrahydrofuran and HI in Acetic acid When 2 equiv of H1 was used, the diiodo product 20 was obtained in 90 % with 10 % of acetylated starting material 19 lefi, while no expected mono iodide product was formed. Reducing the amount of H1 to 1 equiv, still, only diiodo product 20 was obtained, even though 55 % of 19 remained in the reaction system. 59“ 59A° QAc 19 : 20 CH3COOHIHI : I Z 2 = Z 9 "’ ,/\/\/ Zequiv HI 10: 90 0 r.t.. 1 h 0 tequiv H1 55 I 45 1o 19 20 Figure 4.9 Reaction of (S)-3-hydroxy- tetrahydrofuran and H1 in Acetic acid The formation of only diiodo product indicated that the monoiodo intermediate was very reactive. Once it was formed, it quickly transformed to the diiodo product. A mechanism based on this observation is shown in Figure 4.10. 121 39H :‘Ojl/ \‘0 Z S ' l 1 04- HI 3 20 22 21 Figure 4.10 Mechanism for reaction of (S)-3-hydroxy- tetrahydrofuran and H1 in Acetic acid Once the monoiodo compound 21 was formed, the 4-hydroxyl group attacks the acetyl carbonyl carbon to form the six-membered orthoester intermediate 22, iodide attacked C- 4 to cleave the orthoester and give the diiodo product 20. In this process, the rate determining step is the formation of the 1,3-dioxolanium 18 from 17. To form 18, a highly strained fused bicyclic ring transition state must be formed. Once 18 was formed, it was quickly transformed to 20 through 21 and 22. If no participation group exists, the opening of the THF ring is expected to be slower and the monoiodo product should be formed predominately. Indeed, treating THF with 2 equiv H1 in acetic acid at room temperature for 3 hour gave 64 % monoiodo product 4- iodo butyl acetate 24 with 36 % THF remaining, no diiodo product was observed (Figure 4.11). 122 2 equiv HI, CH3000H /\/\/OAC = I Z 5 r.t 3 h 0 64 % 23 24 Figure 4.11 Reaction of tetrahydrofuran and H1 in Acetic acid Trifluoroacetic acid (TFA) as solvent If the reaction of (8)-3-hydroxy-tetrahydrofuran 10 with HI was run in a non- participation solvent, such as TF A, the monoiodo product should be formed exclusively without the formation of the diiodo product. Also, without participation in the opening of the THF ring, the 1,2-diol derivative should be obtained because the position of the electron withdrawing hydroxyl group. 911 00(0)“, 2 equiv HI, cracoorl Momma, Z 5 t I O 60°C, 2 h 8.4 % 10 25 Figure 4.12 Reaction of (S)-3-hydroxy-tetrahydrofuran and H1 in trifluoroacetic acid Reaction of (S)-3-hydroxy-tetrahydrofi1ran 10 with 2 equiv H1 at room temperature for 5 hour gave 3.2 % of the expected 4-iodo-1,2-diol derivative 25 without formation of the 1,3 diol or diiodide product (Figure 4.12). However, the reactions in TFA were very slow. After 64 h at room temperature, only 12 % of the product 25 was obtained. Raising 123 the reaction temperature to 60 °C did not give satisfactory results either. Product 25 was obtained in 4.7 % and 6.8 % after 2 and 5 hour respectively. Acetic Acid/Acetic Anhydride as solvent If extra acetic anhydride existed in the reaction system with acetic acid as solvent, then once the ring was opened, the free hydroxyl group will be acetylated and thus the monoiodo 1,2 diol product should be obtained. pH 2equiv HI OAc OAc . AcOH/Aczo CL A020 WI Z S g = AcO l 0 60°C 3 h OH 77.8 % 10 26 16 Figure 4.13 Reaction of (S)-3-hydroxy-tetrahydrofuran and H1 in acetic acid/acetic anhydride Reaction of (S)-3-hydroxy-tetrahydrofuran 10 with 2 equiv HI and 5 equiv AczO in AcOH at 60°C for 3 h gave 77.8 % of the expected 4-iodo-1,3 diol derivative 16 without formation of the 1,2 diol or diiodo product (Figure 4.13). As discussed in Chapter 4.2, the existence of AczO in the reaction system lowered the reaction rate (without AczO, 90 % of the diiodo product was obtained at room temperature for 1 hour, Figure 4.9). The results for the reaction of (S)-3-hydroxy-tetrahydrofuran with HI under different conditions are shown in Figure 4.14. The regio selectivity is realized by taking advantage 124 of the electron withdrawing property of the 3-hydroxy group and controlling the neighboring group effect of the 3-acetyl group. 9H ./\/\/0H 11 H20 911 ; AcOH/ACZO 3 Aeo/VV' ‘ l 2 9A1: O 16 10 TFA QCIOICFs I momma: 25 20 Figure 4.14 Reaction of (S)-3-hydroxy - tetrahydrofuran and HI under various conditions 4.4 Reactions of 1,S-anhydro-2,3,4,6-tetra-O-acetyl-D-glucitol with HI Under similar conditions as for the reaction of (S)-3-hydroxy-tetrahydrofi1ran with H1 in acetic acid, the reaction of the tetrahydropyran derivative 1,5-anhydro-2,3,4,6-tetra-O- acetyl-D-glucitol 27 should give the open chained 6-iodo product 30 as proposed in Figure 4.15. Protonation of the ring oxygen followed by 6-acetyl group assisted cleavage of the ring C5-O bond should give the open chained dioxonium ion 29, which should be opened by iodide at O6 to give the iodo product 30. H1 OAC AcO O ......... .. AcO OAc A OH O c 37 on c 30 = t t : r )2 5 7 0 ° 036‘ '9' ------- -> AcO AcO OH AcO AcO 0A6 0A6 L. .4 28 29 Figure 4.15 Proposed reaction of H1 mediated ring opening reaction of 1,5-anhydro- 2,3,4,6-tetra-O-acetyl-D-glucitol The results for reaction of 1,5-anhydro-2,3,4,6-tetra-O-acetyl-D-glucitol with HI are listed in table 4.5 (Figure 4.16). ON: I o 10equiv HI 0 AcO AcO AcO AcO A00“ AcO AcO O 4. OM: OM: OM: 27 31 32 Figure 4.16 Reaction of 1,5-anhydro-2,3,4,6-tetra-O-acetyl-D-glucitol Table 4.5 Reaction of 1,5-anhydro-2,3,4,6-tetra-O-acetyl-D-glucitol Entry Temp (°C) Time (h) Product(s) 1 25 18 27 (95 %) 2 55 18 27 (95%) 27 (30 %) 3 95 18 31 (30 %) 32 (10 %) 4 1 10 . 7 Decomposed At lower temperature (25 °C, 55 °C) after 18 h (Table 4.5, entry 1, 2), no product is obtained while 95 % of the starting 27 is recovered. Raising the temperature to 95 °C for 126 18 h, the cyclic 6-iodo product 31 was obtained in 30 % yield along with 10 % olefin product 32. 27 is recovered in ~30% and no open chained iodo product was observed. At higher temperature (110 °C), no significant amount of any product or starting material can be isolated. It is known that the cleavage of tetrahydropyran (THP) is much slower than that of THF, as the six-membered cyclic THP is more stable than the five-membered cyclic THF. As for l,5-anhydro-2,3,4,6-tetra-O-acetyl-D-glucitol, the 4 electron withdrawing groups make the THP ring more electron deficient and harder to be cleaved. Under the conditions used, no ring-cleavage product can be formed. 4.5 The application of iodide products in the preparation of advanced derivatives The chiral iodide products prepared can easily be converted to other heteroatom containing compounds, such as S, N heterocyclic compounds and carbocyclic compounds. Refluxing 1,4-diiodo-2-butanol-acetate 20 with sodium sulfide in ethanol for 5 h gave (8)-3-hydroxy-tetrahydrothiophene 33 in quantitative yield. The optical purity of 33 is higher than 99 %.2° OH QAC N328.9H20 3‘ l/\/\/ ' 7‘ l ) EtOH. reflux, 5h 8 100 % 20 33 Figure 4.17 Preparation of (S)-3-hydroxy-tetrahydrothiophene 127 Reaction of the diiodo compound with benzyl amine in toluene at the existence of potassium carbonate for 5 h under reflux followed by hydrolysis of the acetyl gave the desired (8)-l-benzyl-3-hydroxy-pyrolidine 34 in 60 % over 2 steps. The optical purity of 31 is larger than 99 %.27 9H 9A0 ( 1 T | a. b N l/\/\/ V 20 34 Figure 4.18 Preparation of (S)-1-benzy1-3-hydroxy-pyrolidine Reaction conditions: a). BnBHz, KzCOy, Toluene, reflux, 5 h; b) K2CO3, H20 2 steps, 60 %. Compound 20 is also a good intermediate for the construction of carbocyclic compounds. Reaction of 20 with diethyl malonate in the presence of t-butoxyl potassium gave the chiral cyclopentane derivative 35 in 65 % yield. 9A1: gm: 0 O tBuOK / omso /\/\/ I M = ' + 51° 05‘ Etozc c0251 20 35 Figure 4.19 Preparation of (S)-diethy1-3-acetoxycyclopentane-l ,l-dicarboxylate 4.6 Conclusion Using HI as an economic and general iodide source, the important 4-carbon iodide synthons, (S)-3-hydroxy-4-iodo-butanoic acid, (S)-4-iodo-1,3-butanediol, (S)-4-iodo- 1,2-butanediol, (S)-1,4-diiodo-2-butanol and their acetylated derivatives are prepared. The usefulness of the iodide synthons were demonstrated through the preparation of the heterocyclic compounds (S)-3-hydroxy-tetrahydrothiophene and (S)-l-benzyl-3-hydroxy- pyrolidine and also the carbocyclic compound (S)-diethyl-3-acetoxycyclopentane-1,1- dicarboxylate. 129 4.7 Experimental General procedures: '11, '3 C NMR spectra were recorded at 500, 125, MHz, respectively, with a Varian instrument at 293 K. The chemical shifts are given in ppm using CDC13 residue as reference (5 7.24 p) for 1H and relative to the central CDC13 resonance (5= 77.00p) for I3C NMR unless otherwise specified. Optical rotations were measured on a Jasco P1010 polarimeter at 20°C. IR spectra (wave numbers in cm") were recorded on a FT IR Nicolet 740 spectrometer in CHC13 solutions or KBr pellets. All chemicals were purchased from Aldrich Chemical Co. and used without further purification. The H1 in acetic acid, acetic acid/acetic anhydride, trifluoroacetic acid solution was prepared as needed. For the preparation of H1 in acetic acid solution, 47 % H1 in H20 (6.16 g) was cooled to 0°C with ice-bath, AczO (17.16 ml) was added drop-wised with the control of the solution temperature no higher than 40°C. Stirring was continued for another 30 min at r.t. after the addition was done, the solution prepared contain 11 % HI. For the preparation of H1 in acetic acid/acetic anhydride, trifluoroacetic acid, the same protocol is followed with the addition of the amount of anhydrides needed. For HI mediated reactions, the progress of the reaction was monitored with 1H and '3C No-Deuterium NMR. The signal monitored for the ring opening reaction of (S)-3- hydroxy-butyrolactone is H-3 and C-4. For (8)-3- hydroxy-tetrahydrofuran, the signal monitored are H-3 and C-1 or 04 (whichever is iodinated). (S)-3-acetoxy-butyrolactone (5) To a stirred solution of AC2O (4.7 ml) in dry pyridine (15 ml) at r.t was added (8)-3- hydroxy-butyrolactone (1.02 g, 10mmol). After stirring at r.t overnight, most of the solvent was removed under high vacuum, diethyl ether (50 ml) was added and washed successively with cold water, saturated aqueous NaHC03, and brine (30 ml each) and dried over anhydrous Na2SO4. Removal of the solvent gave compound 5 as a colorless oil (1.32 g, 94 %). [(11200 -455 (e 1, CHC13); 'H NMR(CDC13): 8 2.03 (s, 3H, CH3 Ac), 2.48-2.54(m, 1H, H-2a), 2.82(dd, J = 6.7, 18.4 Hz, 1H, H-2b), 4.27—4.30(m, 1H, H-4a), 4.45 (dd, J = 5.0, 11.1 Hz, 1H, H-4b), 5.36 (m, 1H, H-3); l3C NMR(CDC13): 5 20.5, 34.2 (02), 69.6 ((3-3), 72.7 (04), 170.0, 174.5. Reaction of (S)-3-hydroxy-butyrolactone 1 with HI: H20 as solvent: (8)-3-hydroxy-butyrolactone (0.102 g, 1 mmol) was added to a solution of H1 in H20 (47%, 1.36 g), after stirring at r.t for 24 h, no reaction product was observed from NMR. AcOH, AcOH/Ac20 as solvent: (S)-3-hydroxy-butyrolactone (0.102 g, 1 mmol) was added to a solution of H1 in AcOH with excess AC2O (50 %, 5 % or 0 %), the samples were stirred at the desired temperature, and were tested using NMR for the formation of products at the time listed in table 4.1, 4.2. 131 For workup: after removing most of AcOH under vacuum, EtOAc (20 ml) was added followed by addition of saturated aqueous NaHCO3, after separation, the aqueous layer was extracted with EtOAc(2 x 20 ml), the combined organic layer was washed with brine (20 ml) and dried over anhydrous Na2SO4. Removal of the solvent gave a colorless oil. Column chromatography (if needed) gave 3-acetoxy-4-iodo-butyric acid .3: 1H NMR (CDCI3): 5 2.04(s, 3H, CH3 Ac), 2.76(dd, J = 6.7, 7.0 Hz, 2H, H-2), 3.34 (dd, J = 4.5, 11.0 Hz, 1H, H-4a), 3.39 (dd, J = 5.2, 11.0 Hz, 1H, H-4b), 4.99(m, 1H, H-3); 13 C NMR(CDC13): 5 6.8 (C-4), 20.8, 38.6 (C-2), 68.3 (C-3), 170.1, 175.6. Ethyl 3-(S)-hydroxy-4-iodo-butyrate For the preparation of Ethyl 3-(S)-acetoxy1-4-iodo-butyrate, ethanol (30ml) was added to the condensed reaction solution (~2 ml) of (S)-3-hydroxy-butyrolactone (0.102 g) with H1 in acetic acid along with 2 drops of concentrated sulfuric acid. After stirring at r.t. for 12 h, the reaction solution was concentrated, EtOAc (20 ml) and saturated aqueous NaHCO3 (20 ml) was added, after separation, the aqueous layer was extracted with EtOAc(20 ml), the combined aqueous layer was washed with brine (20 m1), dried over anhydrous Na2SO.; and condensed. Column chromatography gave Ethyl (S)-3-hydroxy- 4-iodo-butyrate as a colorless oil (0.21 g, 81 %), [01]2°D -9.7 (c 1, CHC13); 1H NMR (CDC13): 5 1.16 (t, J = 7.2 Hz, 3 H, CH3 0 ethyl), 2.47 (dd, J = 4.2, 16.6 Hz, 1 H), 2.58 (dd, J = 4.1, 16.5 Hz, 1H), 3.20-3.36 (m, 2H, ), 3.86-4.00 (m, 1H, H-3), 4.05 (q, J = 7.0 Hz, 2 H, 0CH2); l3C NMR(CDC13): 5 12.0 (C-4), 13.8 (CH; O-ethyl), 40.7 (C-2), 60.8 (CH2 O-ethyl), 67.3 (C-3), 171.4 (C=O). 132 (S)-3-Tetrahydrofuryl-acetate (l9) [a]20[) -16.75 (c 1, CHC13); 'H NMR (coc13): 81.89 (m, 1H, H-4), 1.95 (s, 3H, COCHy), 2.06 (m, 1H, H-4’), 3.69-3.83 (m, 4H, H-2, H-5), 5.17 (m, 1H, H-3); l3C NMR(CDC13): 8 20.8 (COCHy), 32.4 (04), 66.7 (05), 72.8 (02), 74.5 (C-3), 170.5 (C0CH3). 4-Iodo-(S)-2-hydroxy-butan-l-o| (11)29 General method for the reaction of (S)-3-hydroxy-tetrahydrofuran with H1 in water: (S)- 3-hydroxy-tetrahydrofuran (0.088 g, 1 mmol) was added to a solution of H1 in water (47 %, 0.33 g, 1.2 mol) at 60°C, under the conditions listed as in table 4.4 (for the conditions with no light, no oxygen, the reactions were run in a deep-dark colored flask under argon atmosphere; for the conditions with light only, the reactions were run in a ordinary flash under argon atmosphere; for the conditions with both light and oxygen, the reactions were run with ordinary flask and air was bubbled to the reaction solution through the process). For workup: Sodium thiosulfate (~ 0.05 g) was added to the stirred solution until the Iodine color disappear, then brine (10 ml) was added, the mixture was extracted with THF (4 x 15 ml), the organic layers were combined, dried and condensed to give a colorless oil, column chromatography gave the pure product 11. [01200 'H NMR (CDCI3): 5 1.81-1.88 (m, 1H, H-3), 1.93-2.00(m, 1H, H-3’), 3.23-3.32 (m, 2H, H-l, H-l’), 3.46 (dd, J = 6.6, 11.6 Hz, 1H, H-4), 3.56 (dd, J = 3.8, 11.6 Hz, 1H, H-4’), 3.76 (m, 1H, H-2); l3c NMR(CDClg): 5 1.28 (C-4), 34.0 (C-3), 63.0 (C-l), 69.7 (C-2). 133 (S)—4-(2-iodoethyl)-2,2-dimethyl- 1,3-Dioxolane A solution of 4-Iodo-(S)-2-hydroxy-butan-1-ol (0.216 g, Immol) in acetone with catalytic amount of concentrated sulfuric acid was stirred at r.t for 5 h and then solid sodium bicarbonate (0.5 g) was added and stirring was continued for 1 h. After filtration, concentration, the residue was dissolved in diethyl ether and washed successively with saturated aqueous sodium bicarbonate, water and brine. Evaporation of the solvent gave the product as a colorless oil (0.235 g, 92 %).[oz]2°p (c l, CHC13); 1H NMR (CDC13): 5 1.35 (s, 3H, CH3), 1.40 (s, 3H, CH3), 2.01-2.14 (m, 2H, H-), 3.15-3.30 (m, 2H, H-), 3.55 (dd, J = 6.2, 7.9 Hz, 1H, H—), 4.07 (dd, J = 6.2, 7.9 Hz, 1H, H-), 4.14-4.19 (m, 1H, H-); 13C NMR (CDC13): 5 1.51 (C-4), 25.5, 27.0, 37.8, 68.7, 75.7 1, 4—diiodo-(S)-2-bntanol-acetate (20)29 To a stirred solution of H1 in water (47 %, 3.1 g, 11.4 mmol) under ice-water bath was added drop-wised acetic anhydride (8.6 ml). After stirring another 10min, the ice-water bath was removed and (S)-hydroxytetrahydrofuran (0.50 g, 5.7 mmol) was added. After stirring at r.t. for 1 h, the reaction solution was cooled to r.t. and then concentrated to about 3 ml under vacuum at r.t. Diethyl ether and saturated aqueous NaHCO3 (30 ml each) were added to the reaction solution. After separation, the aqueous layer was extracted with EtOAc, the combined organic layer was washed with 5 % aqueous sodium thiosulfate, brine and dried over anhydrous Na2SO4. Removal of the solvent gave 20 a colorless oil ( 1.88 g, 90 %).[a]2°D -35.9 (c 1, CHC13); 1H NMR (CDC13): 5 4.56(m, 1H, H-2), 3.23(dd, 1H, J=10.7 Hz, 5.3 Hz, li-l.a), 3.13(dd, 1H, J= 10.7 Hz, 4.8 Hz, H-lb), 134 2.94-3.04(m, 2H, H-4), 2.03-2.10(m, 2H, H-3), 1.92(s, 3H, CH3CO); 13C NMR (CDC13): 8 -020 (C-4), 7.53 (C-l), 20.77 (CH3CO), 37.41(C-3), 71.42(c-2), 169.1 (CH3CO); Anal. Calcd for C6H101202: C, 19.59; H, 2.74. Found: C, 19.69; H, 2.62. Triflnoroacetic acid as solvent for the reaction of (S)-3-hydroxy-tetrahydrofuran with HI: To a stirred solution of H1 in water (47 %, 1.55 g, 5.7 mmol) under ice-water bath was added drop-wised trifluoroacetic anhydride (6.3 ml). After stirring another 10 min, the ice-water bath was removed and (S)-hydroxytetrahydrofuran (0.50 g, 5.7 mmol) was added. After stirring at 60°C for 2 h, NMR shown 8.4 % of 4-Iodo-l,2-butanediol — di- trifluoroacetate was formed. The reaction solution was cooled to r.t. and then concentrated to about 3 ml under vacuum at r.t, brine (10 ml) was added, followed by addition of solid sodium thiosulfate until the Iodine color disappear, the mixture was then extracted with THF (4 x 15 ml), the combined organic layer was dried, condensed. 82 % (0.41 g) of the starting (S)-2-hydroxy-tetrahydrofuran was recovered through column chromatography, 4-Iodo-(S)-2-hydroxy-butan-1-ol 11 was obtained as the hydrolyzed product (67.5 mg, 5 %), which gave the same spectra as the product from reaction in water. (S)-4-Iodo-l,3-butanediol—diacetate (16)::9 To a stirred solution of H1 in water (47 %, 1.55 g, 5.7 mmol) under ice-water bath was added drop-wised acetic anhydride (9.1 ml). After stirring another 10min, the ice-water bath was removed and (8)-hydroxytetrahydrofuran (0.50 g, 5.7 mmol) was added. After 135 stirring at 60°C for 3 h, the reaction solution was cooled to r.t. and then concentrated to about 3 ml under vacuum at r.t. Diethyl ether and saturated aqueous NaHCO3 (30 ml each) were added to the reaction solution. After separation, the aqueous layer was extracted with diethyl ether, the combined organic layer was washed with cold 5 % aqueous sodium thiosulfate, brine and dried over anhydrous Na2SO4. Removal of the solvent gave 16 as a colorless oil (1.32 g, 77.8 %): 1H NMR (CDC13): 5 4.42-4.51 (m, 1H, H-3), 4.11 (t, J = 6.2 Hz, 1H), 3.22-3.43 (m, 2H), 2.13-2.2.25 (m, 2H), 2.09, 2.03 (2 s, 3H each, 2 OAc); ”C NMR (CDC13): 5 171.7, 171.0, 69.4, 60.3,34.8, 21.1, 21.0, 7.8. Reactions of 2,3,4,6-tetra-O-Acetyl-1.5-Anhydro-D-glucitol (27) with HI A solution of H1 in AcOH (11 % w/w, 23.3 g, 20 mmol) containing compound 27 (0.664 g, 2.0 mmol) was stirred at 95°C for 18 h. Afler removing the solvent under vacuum, EtOAc and saturated aqueous NaHCO3 (30 ml each) were added to the reaction solution. After separation, the aqueous layer was extracted with EtOAc (2 x 30 ml), the combined organic layer was washed with, brine and dried over anhydrous Na2S04. Removal of the solvent followed by column chromatography gave compound 27 (30 %), 31 (30 %) and 32 (10 %). 1,5-anhydro-6-deoxy-6-iodo-D-glucitol (31) 'H NMR (CDCI3): 5 5.18 (t, J = 9.4 Hz, 1H), 4.98 (ddd, J = 5.8, 9.6, 10.6 Hz, 1H, H-), 4.86 (t, J = 9.3 Hz, 1H), 4,16 (dd, J = 5.7, 10.3 Hz, 1H), 3.36-3.26 (m, 3H, H-6, H-6’, H- 1), 3.10 (dd, J = 7.6, 11.7 Hz, 1H, H-l’), 2.04, 2.01, 2.00 (s, 3 H each, 3 OAc); l3c NMR(CDC13): 5 170.18, 169.60, 169.35 (3 OAc), 77.27. 73.16, 72.22, 68.94, 66.56, 20.62, 20.59, 20.59 (3 OAc), 3.55 (06). 136 2,6-anhydro-l-deoxy-3,4,5-tri-O-acetate-L-xylo-Hex-l-enitol (32) [(1]ZOD + 9.00 (c 1 CHC13); 'H NMR (CDC13): 8 2.12, 2.10, 2.09 (3 s, 9 H, 3 x OAc), 3.45 (dd, J = 8.3, 11.5 Hz, 1H, H-6), 4.07 (dd, J = 4.9, 11.5 Hz, 1H, H-6’), 4.35 (t, J = 1.5 Hz, 1H, H-l), 4.60 (t, J = 1.5 Hz, 1H, H-l’), 4.93 (m, 1H, H-S), 4.99 (t, J = 7.8 Hz, 1H, H-4), 5.47 (d, J = 7.8 Hz, 1H, H-3); l3C NMR(CDC13): 8 20.3, 20.3, 20.6 (3 Carbon, 3 CH3), 66.7 (C-6), 68.5 (2 Carbon) 72.0 (03, 4, 5), 95.4 (C-2), 153.5 (C-l), 168.9, 169.4, 169.4 (3 x OAc). (S)-3-Hydroxy-tetrahydrothiophene (33)” 31 To a stirred solution of 1, 4—diiodo-2-butanol- acetate (0.368 g, 1.0 mmol) in ethanol (6.0 ml) was added Sodium sulfide nonahydrate (0.360 g, 1.5 mmol). After stirring under reflux for 5 h, EtOAc (20 ml) was added. After filtration and condensation, 33 was obtained as colorless oil (1.04g, 100%) [a]2°D -14.1 (c 1 CHC13); 1H NMR (CDC13): 5 1.75-1.84 (m, 1H, H-4a), 2.07-2.14 (m, 1H, H-4b), 2.13 (s, 1H, C(3) 0H), 2.76-2.98 (m, 4H, 2 H-2, 2 111-5), 4.56(m, 1H, H-3); l3C NMR(CDC13): 5 28.1 (C-5), 37.9 (C-4), 39.7 (02), 74.4 (GB). l-Benzyl-3-(S)-hydroxypyrrolidine(34)°2’°° To a mixture of benzylamine (0.54 g, 5.0 mmol) and potassium carbonate (1.4 g, 10.0 mmol) in acetonitrile (30 mL) was added 1,4-diiodo-(S)-2-butanol-acetate (1.84 g, 5.0 mmol). After refluxing for 12 h, the solvent was removed under vacuum, water (2.0 g) and ethanol (10 ml) were added. The solution was then stirred under reflux forl h. After 137 removing the solvent, water and ethyl acetate (30 mL each) were added to the residue. After separation and extracting the aqueous layer with ethyl acetate (2 x 30 mL), the organic layers were combined. dried over Mg804 and concentrated under vacuum. Purification using column chromatography on silica gel gave 34 as a colorless oil (0.53 g, 60%); [a]20[) -3.6 (c 1 CHC13); 'H NMR(CDC13): 8 1.62-1.77 (m, 1H), 207235 (in, 2 H), 2.50 (br, 1H), 2.52 (dd, J = 5.1, 10.0 Hz, 1H), 2.65 (dd, J = 2.2, 10.0 Hz, 1H), 2.82 (ddd, J = 4.1, 8.3, 8.3 Hz, 1H), 3.60 (s, 2H), 4.31 (m, 1H), 7.29 (m, 5H); I3C NMR(CDC13): 5 35.0 (C-4), 52.6 (C-S), 60.3 (C-2), 63.1 (PhCH2), 70.7 (C-3), 127.0, 128.1, 128.8, 138.6. 3-Acetoxy-cyclopentane-1,l-dicarbohylic acid diethyl ester (35) To a mixture of dimethyl malonate (0.66 g, 5.0 mmol) and KOBut in DMSO (15 ml) was added a solution of 1,4-diiodo-(S)-2-butanol-acetate (1.84 g, 5.0 mmol) in DMSO(5 ml). After stirring at r.t. for 48 h, brine and ethyl acetate (30 ml each) were added to the residue. After separation and extracting the aqueous layer with ethyl acetate (2 x 30 mL), the organic layers were combined, dried over MgSOa and concentrated under vacuum. Purification using column chromatography on silica gel gave 3-Acetoxy-cyclopentane- 1,1-dicarbohy1ic acid diethyl ester 35 as a colorless oil (0.88 g, 65 %); [01]2°D -2.26 (c 1 CHC13); IH NMR (CDC13): 5 1.07-1.15 (m, 6 H, 2 0CH2CH3), 1.66-1.72 (m, 1H, H-5), 1.84 (s, 3H, OAc), 1.92 (dd, J = 1.1, 22.1 Hz, 1H, H-4), 2.03 (dddd, J = 4.8, 8.3, 13.4 Hz, 1H, H-4’), 2.23 (m, 1H, H-2), 2.30 (dt, J = 8.3, 13.4 Hz, 1H, H-5’), 2.44 (dd, J = 6.1, 14.9 Hz, 1H, H- 2’), 4.01-4.07 (1n, 4H, 2 OCH2CH3), 5.00-5.04 (m, 1H, H-3); I3C NMR(CDC13): 513.67, 13.71 ( 2 OCH2CHy), 20.78 (OAc-CH3), 31.58 (C-4 or C-S), 138 31.63 (C-4 or C-5), 39.88 (C-2), 58.77 (C-l), 61.14, 61.28 (2 OCH2CH3), 75.33 (C-3), 170.13, 171.16, 171.59 (3 CH3CO). 139 4.8 References [l] Giese B. Radicals in Organic Synthesis: Formation of Carbon—Carbon Bonds, Pergamon Press, Oxford, 1986. [2] Wakefield, B. J. Organolithium Methods; Academic: London, 1988. [3] Yang, L.M.; Huang, L.F.; Luh, T.Y.. Org. Lett. 2004, 6, 1461. [4] Bohlmann, R. In ComprehensiVe Organic Synthesis; Trost, B. M., Fleming, 1., Ed.; Pergamon Press: Oxford, 1991, Vol 6, pp 203. [5] Jeropoulos, S.; Smith, E. H. J. Chem. Soc, Chem. Comm. 1986, 21,1621. [6] Rydon, H. N.; Tonge, B. L. J. Chem. Soc. 1956, 3043. [7] Verheyden, J. P. H.; Moffatt, J. G. J. Org. Chem. 1972, 37, 2289. [8] Garegg, P. J.; Johansson, R.; Orthega, C.; Samuelson, B. J. Chem. Soc., Perkin Trans. 1 1982, 681. [9] Harrison, G. C.; Diehl, H. Organic Synthesis, Wiley: New York, 1953; Collect. Vol. 111. pp 370. [10] Bandgar, B. P.; Sadavarte, V. S.; Uppalla, L. S. Tetrahedron Lett.200l, 42. 951. [11] Jung, M.E.; Ornstein, P. L. Tetrahedron Lett. 1977, 31, 2659. [12] Fernandez, 1.; Garcia, B.; Munoz, 8.; Pedro, J. R.; Salud, R. Synlett 1993, 489. [13] Martinez, A. G.; Alvarez, R. M.; Vilar, E. T.; Fraile, A. G.; Barnica, J. 0.; Hanack, M.; Subramanian, L. R. Tetrahedron Lett. 1987, 28, 6441. [14] Olah, G. A.; Gupta, B. G.; Malhotre, R.; Narang, S. C. J. Org. Chem. 1980, 45, 1638. [15] Wiley, G. A.; Hershkowitz, R. L.; Rein, B. M.; Chung, B. C. J. Am. Chem. Soc. 1964, 86, 964. [16] Iranpoor, N.; Firouzabadi, H.; Aghapour, G.; Vaez zadeh, A. R. Tetrahedron 2002, 58, 8689. [17] Ferreri, C.; Costantino, C.; Chatgilialoglu, C.; Boukherroub, R.; Manuel, G. J. Organometallic. Chem. 1998, 554, 135. [18] Joseph, R.; Pallan, P.; Sudalai, A.; Ravindranathan, T. Tetrahedron Lett. 1995, 36, 609. 140 [19] Hollingsworth, R 1.; J. Org. Chem. 1999, 64, 7633. [20] Hollingsworth, R. 1.; Wang, G. J. Chem. Rev. 2000, 100, 4267. [21] Bowden, K. Adv. Phys. Org. Chem. 1993, 28, 171. [22] Bowden, K. Chem. Soc. Rev. 1995, 24, 431. [23] Gervay, J .; Gregar, T. Q. Tetrahedron Lett. 1997, 38, 5921. [24] Wiberg, N.; Holleman, A. F .; Wiberg, E. Inorganic Chemistry, Academic Press, 2001,pp432. [25] Cataldo, F. Eur. Polym. J. 1996,32, 1297. [26] Brown, H. C.; Prasad, J. V. N.; Vara, R. B. W. J. Am. Chem. Soc. 1986, 108, 2049. [27] Bhat, K. L.; Flanagan, D. M.; Joullie, M. M. Synth Comm.1985, 15, 587. [28] Hoye T. R.; Eklov B. M.; Ryba T. D.; Voloshin M.; Yao L. J. Org. Lett. 2004, 6, 953. [29] Mimer, P.; Saluzzo, C.; Amouroux, R. Synth. Comm. 1995, 25, 613-627. [30] Zhang, X.; Taketomi, T.; Yoshizumi, T.; Kumobayashi, H.; Akutagawa, S.; Mashima, K.; Takaya, H. J. Am. Chem. Soc. 1993, 115, 3318-3319. [31] Brown, H. C.; Prasad, J. V. N. V.; Zee, S. H. J. Org. Chem. 1985, 50, 1582. [32] Bhat, K. L.; Flanagan, D. M.; Joullie, M. M. Synth Comm. 1985, 15, 587-598. [33] Mehta, A.; Gupta, J. B.; Sarma, P. K. S. Preparation of 1-substituted-3- pyrrolidine derivatives as muscarinic receptor antagonists. PCT Int. Appl. WO 2004056767 (2004). 141 Chapter 5 Preparation of Ribose Derived Nitrone and its Application in the Preparation of Iminosugars 142 Abstract Starting from D-ribose, a six-membered cyclic nitrone was prepared in 3 steps. The nucleophilic reaction and 1,3-dipolar cycloaddition reaction of the nitrone prepared were explored. Employing nitromethane as the nucleophile, a 2-aminomethyl-3,4,5- piperidinetriol derivative was prepared without using the toxic TMSCN reagent. Isoxazolidines were prepared by the 1,3-dipolar cycloaddition of the nitrone with allyl alcohol and vinyl ethyl ether. Spiro-isoxazolidines were also prepared by employing carbohydrate derived alkenes. The cycloaddition reactions of nitrone and carbohydrate derived alkenes are highly regio and diastereoselective, only one product was obtained from each alkene. 143 5.1 Introduction 5.1.1 Iminosuars as glycosidases inhibitors Glycosidases catalyze the hydrolysis of glycosidic bonds in carbohydrates, glycoproteins and glycolipids" 2. Glycosidases are involved in the biosynthesis of the oligosaccharide chains and quality control mechanisms in the endoplasmic reticulum of the N-linked glycoproteins. Inhibition of these glycosidases can have prbfound effects on quality control, maturation, transport, and secretion of glycoproteins and can alter cell-cell or cell-virus recognition processesz' 3 . In 1966 nojirimycin was discovered as the first glucose analog with the nitrogen atom in place of the ring oxygen (iminosugars)4. Nojirimycin was first described as an antibiotic produced by Streptomyces roseochromogenes R-468 and S. lavendulae SF-425 and was shown to be a potent inhibitor of 01- and B-glucosidases from various sourcess. Since then, over 100 iminosugars have been isolated from plants and microorganisms and many of those natural occurring glycosidase inhibitors and their derivatives or analogues have been synthesized”. In recent years, the biological activities of these iminosugars have been extended to the 10. ll 13 inhibition of glycosyltransferases , of nucleosidase'z‘ and 14 glycogenphosphorylases , and of sugar nucleotide mutase's‘ '°. These remarkable properties of iminosugar give tremendous opportunities in the development of iminosugar 17,18 19,20 based medicines for a wide range of diseases, such as diabetes , viral infections , and tumor metastasis” 22. 144 The strong therapeutic potential of iminosugars has generated a huge interest in their synthesis and structural modification and has stimulated many groups to develop short and stereoselective routes for their synthesis. Many recent syntheses use readily available 2 26-28 and inexpensive chiral-pool starting materials such as carbohydrate323' 5, amino acids , and tartaric acids29‘ 3°. Sharpless asymmetric epoxidation and dihydroxylation reactions have found successful applications in the chiral synthesis of azasugars3 "33. 5.1.2 Application of nitrones in the preparation of iminosugars and aza-C- glycosides. For the preparation of the iminosugars and aza-C-glycosides from carbohydrates23'25, usually, the nitrogen functionalities were installed at very late stage of the synthesis and thus make the preparation of libraries of nitrogen containing carbohydrate mimics very tedious. If the nitrogen functionality can be introduced at the early stage, or carbohydrate derivated synthons containing nitrogen functionality can be prepared, the preparation of the iminosugar libraries will be much easier. Recently, carbohydrate derivated nitrones have been used as the nitrogen containing chiral starting materials for the preparation of iminosugars34'38. The carbohydrate derivated nitrones can be classified into two categories based on the position of the nitrone functionality: the acyclic nitrones and the cyclic nitrones (Figure 5.1). 145 '1' 2—0 '1’ PO / §\ 11 GP Figure 5.1 General structure of carbohydrate based acyclic and cyclic nitrones. The acyclic nitrones can be prepared very efficiently by condensation of the carbonyl groups, usually aldehydes. with hydroxyl amines”. Figure 5.2 list some of the acyclic nitrones. Figure 5.2 Carbohydrate based acyclic nitrones. For the preparation of cyclic nitrones, usually, the protected carbohydrates are first condensed with protected hydroxylamines to form the protected oximes. Transforming one of the hydroxyl groups to a good leaving group followed by deprotection of the oxime give the cyclic nitrones (Figure 5.3 I). 146 op ”WM... OP Deprotection Cyclization O' I Ni ii) POWO n-‘l fl LG formation OP PO 114 LG Condensation fl Cyclization 2—0 + \ OP Figure 5.3 Strategies for the preparation of cyclic nitrones. Recently, Goti et al reported the preparation of the cyclic nitrone 8 from L-xylose (Figure 5.4)“. 147 BnO OBn “0 9°” a, b, c 3110 pan (1 g b "“ “ —* HO O "II/OH HO O .,,’/oan 5n...— u,,/OBn THPO "0 4 5 6 8110 QBn f g Bnobf: e 3 . —-———. -——-—> \ OBn éN....——-f—>'I”/OB'1 H N THPO "8° 1') 7 8 Figure 5.4 Preparation of cyclic nitrone from L-xylose (a) MeOH/H2SO4, anhydr. Na2SO4, rt, 21 h; (b) BnCl/KOH, Na2SO4, reflux, 8 h; (c) 6N HCl, CH3COOH, 60—70°C (50% over three steps); (d) NH2OTHP, no solvent, rt, 6 d, 100%; (e) MsCl, TEA, CH2C12, rt, 24 h, 50%; (DDOWEX 50W X8, MeOH, rt, 24 h, 96%; (g) 0.1 M NaOH, dioxane, 0°C, 2 h, 55%. From these acyclic and cyclic nitrones, a variety of iminosugar derivatives have been prepared through the addition reaction and 1,3-dipolar cycloaddition reactions. For recent developments in the nucleophilic addition reactions of chiral nitrones, Merino’s review is a good resource“. in another review, the application of 1,3-dipolar cycloaddition reactions of nitrones in the construction of carbohydrate mimics were discussed”. Although progresses have been made in the preparation of carbohydrate derived nitrones, these processes are not very efficient, especially for the preparation of cyclic nitrones. Usually, the expensive THPONH243 or TBDPSONH244 is needed or the oxime intermediates formed have to be protected” . 148 In this chapter, a different method was used for the preparation of cyclic nitrone. In this approach, the good leaving group is installed at first. The following one-pot condensation-cyclization reaction gives the cyclic nitrone directly. No protected hydroxylamines or extra steps to protect-deprotect the oxime is needed (Figure 5.3 II). In the addition reactions of nitrones, for the introduction of the aminomethyl group, TMSCN as a nucleophile gave good results4°’ 47. However, the high toxicity of TMSCN prohibits its practical application. For the cycloaddition reactions of nitrones, usually, the alkenes used were not derivated from carbohydrates. In the rare cases where the alkenes were derivated from carbohydrates, the olefin functionalities are usually not connected to the carbohydrate ring“. Here, our efforts for the development of a more efficient and economic method for the preparation of carbohydrate derived nitrone and its application in the addition and cycloaddition reactions involving carbohydrate derived alkenes are discussed. 5.2 An efficient route for the preparation of cyclic nitrone from ribose. Reaction of D-ribose with acetone using concentrated H2804 as the catalyst gives 2,3- O-isopropylidene-D-ribofuranose in quantitative yield. Reaction of 2,3-O-isopropylidene- D-ribofuranose with tosyl chloride in pyridine at 0°C affords 5-O-tosy1-2,3-O- isopropylidene-D-ribofuranose 10 in 78 %. 10 is unstable at room temperature and is 149 used directly in the next step without further purification. (At -17 °C, tosylate 10 can be stored for several weeks without obvious change). 1'(DH Ct (P- (3 8 2 + O HOH2C O OH C N\ C) (”1 ——3LE—a» “""“_" + W “0‘ ' "o O O ; O O on 01-1 x o-fi X 9 1o 11 12 Figure 5.5 Preparation of nitrone using tosylate as substrate. (a) acetone, conc. H2804, quantitative; (b) tosyl chloride. pyridine, 78 %; (c) H2NOH.HC1, base, various yields. For the condensation-cyclization step, various bases and solvents are used. The results are listed in Table 5.1. Table 5.1 Preparation of nitrone using tosylate as substrate. Entry Base Solvent Time (h) Yield (%) l NaHCO; MeOH 24 6 2 NaHCO; MeOH/H2O 12 1 1 3 NaHCO; i-PrOH 30 5 4 NaHCO; i-PrOH/ H20 12 ~7 5 NaHCO; EtOH 24 ~ 5 6 NaHCO3 EtOH/H2O 12 ~ 6 7 Pyridine MeOH 24 - 8 Et3N MeOH 24 12 9 Et3N EtOH 24 15 * Reactions are run at room temperature, TLC are used to monitor the reactions. 150 The base used is crucial to this reaction. If pyridine is used, no nitrone 11 can be formed. Changing the base to NaHCO3, nitrone 11 is formed in various solvent. The best result is obtained in MeOH/ H2O with 11 % of nitrone 11 formed (Table 5.1, entry 2). A better result was obtained by using Et3N as the base and ethanol as solvent (15 %, Table 5.1, entry 9). For all the reaction conditions listed in table 5.1, except entry 7, along with the nitrone, a small amount of the 2,3-O-isopropyliden-l,4-anhydro-D-ribopyranose was formed (4-10 % yield). The other components obtained were complex mixture of compounds. The possible products from the reaction of tosylate 10 with hydroxylamine under basic conditions are shown in Figure 5.6. No further efforts are made to separate and characterize these compounds except 11 and 12. 151 9- TOO /\ N" kl \ °" -u*°” —. O HO‘ L ’o o 0 °“{\ X 11 no OQNmH T30 T80 OH NHOH ' N H2N0H ""0" U ——* —+ ‘ ‘ . 0’ \ o’"‘ 0’ \ %‘OH I K) ' 'OX 4» T.o\\\‘ do 0 o 8.9 6t x Figure 5.6 Possible products from reaction of tosylate 10 with hydroxylamine under basic conditions 152 Fortunately, much better results are obtained by switching tosylate 10 to mesylate 14 (Figure 5.7, Table 5.2). Figure 5.7 Preparation of nitrone using tosylate as substrate. ' (a) Mesyl chloride, pyridine, 75 %; (b) H2NOH.HC1. Table 5.2 Preparation of nitrone using mesylate as substrate. Entry Base Solvent Time (h) Yield (%) l NaHCO; MeOH 24 14 2 NaHCO3 MeOH/H2O 12 18 3 NaHC03 EtOH 24 l 5 4 NaHCO; EtOH/H20 12 22 5 Et3N MeOH 24 25 6 Et3N EtOH 24 62 * Reactions are run at room temperature. TLC is used to monitor the reaction. The best result is obtained with Et3N as the base and EtOH as solvent (62 %, entry 6, Table 5.2). Although the yield is still not very high, considering the short steps and the cheap reagents used, this is still a practical route for the preparation of nitrone from D- ribose. 153 5.3 Steroeselective addition of nitromethane to the chiral nitrone. To search for a substitute of TMSCN as the reagent for the introduction of an aminomethyl group to the nitrone, nitromethane is chosen since it is much less toxic compared to TMSCN and also it has been used extensively for Michael Reactions,”52 Henry Reaciton553’ 54. I '1 N —K . N02 6~$ 15 15 OH N «NO: I 1' N (R) /N02 1” Figure 5.8 Possible products from addition of nitromethane to nitrone Two possible transition states were shown in figure 5.8 for the nucleophilic addition of nitromethane to nitrone. Addition of nitromethane from the Re face (TS 15) of the nitrone will give the 8 product 16 while addition of nitromethane from the Si face (TS 17) of the nitrone will give the R product 18. In TS 15, the incoming nitromethane is anti to the isopropylidene group, while in TS 17, it is syn to the isopropylidene group. The strong steric interaction between the nitromethane group and the isopropylidene in TS 17 will prohibit the Si face approach and thus it is expected that only the Re face attack will happen and only the S product 16 can be obtained. 154 fl NH2 0 I I O \“ ‘O’o \\ a \\‘ HO HO H0 11 16 19 Figure 5.9 Addition of nitromethane to nitrone. (a) MeN02, MeONa, MeOH, r.t, 12 h, 82 %; (b) H2, Pd/C (10 %), MeOH, 100 %. The reaction of nitrone 11 with nitromethane is very efficient. Using MeONa as the base, with 2 equivalent of MeN02, the reaction is completed in 12 h with a high yield (82 %). NMR showed only one product is formed. The stereochemistry of the nitro compound was confirmed by the NOE. No NOE is observed between H3a and H4, which suggests that they are anti to each other as in structure 16. Treatment of the nitro compound 16 with H2 using Pd/C as catalyst give the reduced product 19 in quantitative yield (Figure 5.9). 5.4 Cyclization reactions of the ribose derived nitrone with non-carbohydrate based alkenes For the 1,3-dipolar cycloadditions of nitrones to akenes, usually, the 5-substituted isoxazolidines are obtained from mono-substituted and 1,1-disubstituted alkenes with electron-donating and moderate electron-withdrawing groupsss’ 5°. For olefins with strong electron-withdrawing groups, the 4-substituted isoxazolidines are forrned57’58. If 1,2- disubstituted alkenes are used, mixtures of regioisomers are often formed. Usually, the isomer with the less electron rich substituent at 4-position is formed as the major product59'°' . 155 To control the stereochemistry outcome of the 1,3-dipolarcycloaddtion, many methods have been developed, for example, Lewis acids activation of the dipolarophiles, using chiral auxiliaries, employing enantioselective catalysts (metal or organo-catalysts), etc°2’ °3. For chiral nitrones, the steric hindrance plays an important role in control the approaching of the alkenes toward the nitrones, which makes it possible to achieve high regio and stereo control for cycloaddition reactions of chiral nitrones without the assistance of Lewis acids, chiral auxiliaries or catalysts“. A model for the cycloaddition of the chiral nitrone 11 with allyl alcohol is shown in figure 5.10. As in the nucleophilic addition reactions of the chiral nitrone (Figure 5.8), allyl alcohol can only approach nitrone 11 from the Re face to form the anti product. In figure 5.10, only the transition states with Re attack of the alkenes to the nitrone are shown. 156 1' HO‘ ‘ ‘ L_:’,’/—OH HO‘ v 110‘“ I 24 , 140‘" ll 270 Figure 5.10 Possible products from cyclization of nitrone with allyl alcohol 26 There are 4 possible products from the 1,3-dipolar cycloaddition of nitrone toward allyl alcohol: the exo-4 (4 substituted isoxazolidines 21), endo-4 (23), exo-S (5 substituted isoxazolidines 25) and endo-S (27). Since no strong electron withdrawing group present in allyl alcohol, the cycloaddition reaction should show high region-selectivity with only the 5 substituted isoxazolidines 25, 27 formed. As for 25 and 27, the exo-S product 25 is expected to be formed as the major product for steric reason. It is reported that for the 157 cycloaddition of 6-member-ring nitrone, 3-(tert-butyldiphenylsi1yl)oxy-l-piperidine-l- oxide with allyl alcohol, a 90:10 exo/endo-selectivity, 74:26 diastereofacial selectivity was observed“. For the cycloaddition of 5-member—ring nitrone, 3-(tert- butyldimethylsilyl)oxy-l-pyrroline-l-oxide with allyl alcohol, a 85:15 exo/endo- selectivity, 82:18 diastereofacial selectivity was observed in the favorite formation of the exo product65 . Refluxing nitrone 11 with allyl alcohol in toluene for 4 h give the cycloaddition products in 92 % yield (Figure 5.11). The reaction is highly regio-selective as no endo-4 or exo-4 product 21 or 23 is observed. For the two exo-5 and endo-5 products 25 and 27, 25 is formed as the major product (89:11). The stereochemistry was confirmed by NOESYID. No NOE between H-2 and H-3a is observed for the major product 25 while a 13 % NOE is observed between H-2 and H-3a in the minor product 27. + _ 0 KO” 110“" . "/0 toluene, reflux 3 4n92% , O$ HO\\‘ 11 25 89:11 2—0 Figure 5.11 Cycloaddition of nitrone with allyl alcohol Next, the cycloaddition of nitrone 11 with vinyl ethyl ether is studied. Similarly, no syn product should be formed as the vinyl ethyl ether can only attack the nitrone from the Si face. Vinyl ether type alkenes always give the 5-substituted isoxazolidines in 158 cycloaddition reaction with nitrones°°88. So the expected products are only the exo-5 and the endo-5 products 29 and 31 (Figure 5.12). ..——--0' 1+ ““Q OX0 I i 1 CD 1 X 1 O l or l I l l _ 110“" /—0 28 OH 0 . + f , endo 5 (?_N\ O7< ------------------ -> x0 : ’1 \\" '2 \ I _ I H0 '. I 30 WK 31 Figure 5.12 Products from cycloaddition of nitrone with vinyl ethyl ether Heating a mixture of nitrone 11 and vinyl ethyl ether in chloroform at 62°C in a sealed flask for 4 h give the cycloaddition products in 94 % yield (Figure 5.13). As expected, no endo-4 or exo-4 product is observed. For the two exo-5 and endo-5 products, exo-5 is formed as the major product (68:32). The stereochemistry is confirmed by NOESYID. No NOE between H-2 and H-3a is observed for the major product while 11 % NOE was observed between H-2 and H-3a in the minor product. O I —— it: \o / Ho“" , "/0 chloroform, 62°C H0“ H0 6‘0 4h, 94% 11 2° 68 :32 Figure 5.13 Reaction of nitrone with vinyl ethyl ether 159 5.5 Cycloaddition reactions of the ribose derived nitrone with carbohydrate based alkenes After the successful cycloaddition reactions of the nitrone with non-carbohydrate based alkenes, the carbohydrate based alkenes are used in the 1,3-dipolar cycloaddition reactions. Alkenes 34 and 37, derived from methyl-D-glucopyranoside and 1,5-anhydro- D-glucitol, are used. OH c HO O a! b A60 0 ACAO O O HO ————’ A60 c OH R 32R'OMO 33R'OMO 32:35“ 35RBH 38R=H Figure 5.14 Preparation of carbohydrate derived alkenes. (a) Ph3P/I2, imidazole, toluene, 70°C, 3 h; (b). AC2O/pyridine, r.t, 12 h, 2 steps, 68 % for 33, 65 % for 36. (c) DBU, DMF, 751151, 3 h, 75 % for 34, 76 % for 37. Conversion of the primary hydroxyl groups to iodides followed by elimination of H1 under strong basic conditions give the carbohydrate derived alkenes 34 and 37 in modest yields (~52 %, Figure 5.14). As discussed above, alkenes only approach the nitrone 11 from the Si face. For the vinyl ether type alkenes, the 5—substituted isoxazolidines are formed with modest to high exo/endo selectivity. Similar results are expected for the 1,3-dipolar cycloaddition of the carbohydrate derived vinyl ether type alkenes 34 and 37. However, higher exo/endo selectivity is expected since the chiral alkenes 34 and 37 are used. 160 Refluxing nitrone 11 and alkene 34 in toluene for 12 h give only one product in a very high yield (96 %, Figure 5.15). No NOE was observed between H3’ and H301, which suggests the structure of the product is the spiro-isoxazolidine 39. The high diastereoselectivity is not surprising when the two possible transition states 38 and 40 are considered. In the two transition states, the one with fewer groups which are endo to the nitrone ring and at the same time syn to the isopropylidene group will be favored. In transition state 38, the 3- and 4-acetyl groups are endo to the nitrone ring while only 3- acetyl group is syn to the isopropylidene group. In transition state 40, only the 1- methoxyl group is endo to nitrone the nitrone ring and at the same time syn to the isopropylidene group. Although both the 3-acetyl group in 38 and l-methoxyl group in 40 are at [5 position to the alkene, the shorter C-O bond makes the l-methoxyl group in 40 more sterically hindered than the 3-acetyl group in 38. From the sterically favored transition state 38, the product 39 is formed exclusively. 161 3’ .3 .3 0‘" toluene, reflux 12 h, 96 % O I > o 10 AcO U -----o - l I Egg :- O 0 cl 0 -----o- | 2+ {2‘2 o>€ Aco‘“. 0% I 8. 8 D O 01:. O > 0 OAc OM AcO e 110“" 39 41 96 % not observed Figure 5.15 Cycloaddition of nitrone with carbohydrate derived alkene I From the above analysis, if the 1-methoxyl group was removed, as shown in figure 5.16, transition state 44 should be favored and the product 45 should be formed as the major product. 162 ‘1’ N Ac 0 \ AcO + ,. . Ho“ . ’I OAc ; o“/\ 37 1 1 toluene. reflux 10 h. 89 % AGO " I be AcO‘w 0 110“" not observed Figure 5.16 Cycloaddition of nitrone with carbohydrate derived alkene 11 Only one product was obtained by the cycloaddition reaction of nitrone 11 and alkene 37 (89 %, Figure 5.16). A high NOE between H3’ and H301 was observed (NOE: H301, H30 30 %, H301, H3’ 15 %), which means the structure is the spiro-isoxazolidine 45 from transition state 44. 163 5.6 Conclusion Start from D-ribose, a six-membered cyclic nitrone was prepared in 3 steps. The nucleophilic reaction and 1,3-dipolar cycloaddition reaction of the nitrone prepared were explored. Employing nitromethane as the nucleophile, a 2-aminomethyl-3,4,5- piperidinetriol derivative was prepared without using the toxic TMSCN reagent. Isoxazolidines and Spiro-isoxazolidines were prepared by the 1,3-dipolar cycloaddition of the nitrone with achiral and chiral alkenes. For the chiral carbohydrate derived alkenes, the cycloaddition reaction is highly diastereoselective, only one product was obtained from each alkene. 164 5.7 Experimental General procedures: lH, l3C NMR spectra were recorded at 500, 125, MHz, respectively, with a Varian instrument at 293 K. The chemical shifts are given in ppm using CDC13 residue as reference (5 7.24 ppm) for lH and relative to the central CDC13 resonance (5= 77.00 ppm) for '3 C NMR unless otherwise specified. lH and 13 C are assigned on the basis of 2D lH COSY and IH-BC chemical-shift correlated experiments. For the 1,3 cyclization products, all the NMR were done at 100°C with DMSO-d6 as solvent. Melting points were determined on a Fisher-Johns melting point apparatus (uncorrected). Optical rotations were measured on a Jasco P1010 polarimeter at 20°C. IR spectra (wave numbers in cm") were recorded on a PT IR Nicolet 740 spectrometer in CHC13 solutions or KBr pellets. All chemicals were purchased from Aldrich Chemical Co. and used without further purification. 2,3-O-isopropylidene-D-ribofuranose (13) In a 250 ml flask containing 100 m1 acetone, D-Ribose (10.0 gram, 66.6 mmol) was added followed by the addition of cone. H2804 (1.0 ml). After stirring at r.t. for 4 h, NaHCO3 (4.0 gram) was added. Stirring was continued until the pH of the solution is 7. Filtration followed by removal of the solvent gave the product 13 as a syrup (12.6 g, 100 %). The product was used without further purification. lH NMR(CDC13): 5.43(s, 1H, H- 1); 4.59(d, J=5.9 Hz, 1H, H-2); 4.85(d, J = 5.9 Hz, 1H, H-3), (br, 1H, H-4), 3.81-3.66 (m, 2H, H-5, H-5’); 1.58(s, 3H, CH3); 1.41 (s, 3H, lH NMR (CDC13): 5.43(s, 1H, H-l); 4.59(d, J=5.9 Hz, 1H, H-2); 4.85(d, J = 5.9 Hz, 1H, H-3), (br, 1H, H-4), 3.81-3.66 (m, 2H, H-5, H-S’); 1.58(s, 3H, CH3); 1.41 (s, 3H, CH3); CH3); l3C NMR(CDC13): 112.06 ( 165 C(CH3)2 ); 103.07(C-1); 87.82 (C-4); 86.89 (C-2); 81.68 (C-3); 63.65 (C-5); 24.74, 26.28 ( 2 C(CH3)2 ); 2,3-0-isopropylidene-S-O-Tosyl-I)-ribofuranose (10) To a stirred solution of 2,3-O-isopropylidene-D-ribofuranose 13 (6.3 g, 33.3 mmol) in pyridine (100 ml) at 0°C was added Tosyl chloride (6.35 g, 33.3 mmol) in 3 portions at 30 minutes apart. The reaction solution was warmed to r.t gradually and stirred for another 12 h. Ethanol (5 ml) was added to quench the reaction. After removal of the pyridine, the remaining syrup was partitioned between CHC13 and saturated aqueous NaHCO3 (60 ml each). The aqueous phase was extracted with CHCl3 (30 ml) once and the combined organic phases were washed with water, brine, dried over MgSO4 and condensed to gave 10 as a syrup (8.9 g, 78 %). The product 10 is not stable at r.t and it is used directly for the next step without further purification. A small amount of the product was purified through column chromatography for characterization purpose. mp. 94-95 (lit 92.5-94 )69 'H NMR (CDC13): 8 7.73 (d, J = 18.2 Hz, 2H), 7.30 (d, J = 18.2 Hz, 2H), 5.36 (d, J = 2.6 Hz, 1H, H-l), 4.58 (dd, J = 0.9, 5.8 Hz, 1H, H-3), 4.51 (d, J = 5.9 Hz, 1H, H-2), 4.25 (m, 1H, H-4), 4.02-4.06 (m, 2H, H-5, H-S’), 3.69 (br s, 1H, OH), 2.39 (s, ArCH3), 1.39, 1.23 (s, 2 C(CH3); l3C NMR(CDC13): 5145.07, 132.37, 129.95, 127.84, 112.52, 102.82, 85.44, 83.55, 81.41, 69.87, 26.17, 24.65, 21.50. 2,3-O-Isopropylidene-S-O-methanesulfonyl-B-D-ribofuranose (14) To a stirred solution of 2,3-0-isopropylidene-D-ribose 13 (5.0 g, 26.3 mmol) in pyridine (50 m1) at 10251 was drop-wise added methanesulfonyl chloride (3.10 g, 26.3 mmol). The reaction mixture was gradually warmed to r.t and stirring was continued for another 4 h. 166 Ethanol (5 ml) was added to quench the reaction. After removal of the pyridine under vacuum, CH2Cl2 (30 ml) and water (30 ml) were added. After extraction the aqueous phase with CH2C12 (30 ml), the organic phases were combined, washed with cold 3 M HCl (20 ml), saturated aqueous NaHCO3, and dried over MgSO4. Removal of the solvent gave 14 as a white solid (5.4 g, 76.6 %). Part of the product was recrystalized from CH2Cl2/Et20 for characterization purpose. M.p. 112-113°C (lit 112-114)70 'H NMR (CD3CN): 5 5.35 (s, 1H, H-I), 4.68 (d, J = 5.9 Hz, 1H, H-3), 4.55 (d, J = 5.9 Hz, 1H, H- 2), 4.18-4.27 (m, 3H, H-4, 11-6, H-6’), 3.09 (s, MeSO2), 1.49, 1.34 (s, 2 C(CH3); l3C NMR(CD3CN): 5 112.18, 102.74, 85.93, 83.46, 81.58, 71.13, 37.01, 26.10, 24.44. (3aS, 7R, 7aR)-3a, 6, 7, 7a-tetrahydro-2, 2-dimethyl-5-oxide-1, 3-Dioxolo [4, 5-c] pyridin-7-ol (1 1) Method 1: From 2,3-O-isopropylidene-5-O-Tosyl-D-ribofuranose To 5 ml water was added NaHCO3 (1.26 g, 15 mmol) and H2NOH.HCI (1.04 g, 15 mmol), the mixture was stirred until no bubble come out. This solution was then transferred to a flask containing a solution of 2,3-O-isopropylidene-5-O-Tosyl-D- ribofuranose (1.03 g, 3 mmol) in methanol (15 ml). After stirring at r.t. for 24 h, the solution was then condensed under vacuum with the bath temperature at no higher than 25°C to a syrup. Column chromatography of this syrup gave the desired nitrone 11 in 24 % yield. Method 2: From 2,3-O-Isopropy1idene-S-O-methanesulfonyl-B-D-ribofuranose 167 To 5 ml water was added NaHC03 (1.26 g, 15 mmol) and H2N0H.HCI (1.04 g, 15 mmol), the mixture was stirred until no bubble come out. This solution was then transferred to a flask containing a solution of 2,3-O-isopropylidene—5-0- methanesulfonyl-D-ribofitranose (0.805 g, 3 mmol) in methanol (15 ml). After stirring at r.t. for 24 h, the solution was then condensed under vacuum with the bath temperature at no higher than 25°C to a syrup. Column chromatography of this syrup gave the desired nitrone 11 in 30 % yield. Method 3: From 2,3-O-Isopropylidene-5-O-methanesulfonyl-B-D-ribofuranose with Et3N as base To 10 ml ethanol was added Et3N (1.52 g, 15 mmol) and H2NOH.HC1 ( 1.04 g, 15 mmol), the mixture was stirred until the entire solid dissolved. This solution was then transferred to a flask containing a solution of 2,3-0-isopropylidene-S-O-methanesulfonyl-B—D- ribofuranose ( 1.03 g, 3 mmol) in ethanol (15 ml). After stirring at r.t. for 24 h, the solution was then condensed under vacuum with the bath temperature at no higher than 25°C to a syrup. Column chromatography (Hexane/EtOAc/EtOH 1:1:0 to 0:1 :1) of this syrup gave the desired nitrone in 11 62 % yield. [61200 -148.75 (c 1 CHC13); 'H NMR (CDC13): 1.41 (s, 3H, ), 1.44 (s, 3H), 2.15 (s, 1H, 0H), 3.88 (dd, J = 4.8, 14.3 Hz, 1H, H-6a), 3.96 (ddt, J = 1.5, 11.3, 14.3 Hz, 1H, H-6b), 4.14 (ddd, J = 3.1, 4.8, 9.2 Hz, 1H, H-7), 4.50 (dd, J = 3.0, 6.4 Hz, 1H, H-7a), 4.85 (dd, J = 3.5, 6.4 Hz, 1H, H-3a), 7.05 (dd, J = 1.8, 3.5 Hz, 1H, H-4); I3C NMR (CDC13): 134.3 (C-4), 110.9, 72.2, 70.6, 64.1, 58.1 (06), 26.8, 25.4. Calculated for C8H13N04 C, 51.33, H, 7.00, N, 7.48; Found C 51.30, H, 6.94, N, 7.50. 168 1,5-anhydro-2,3-O-(l-methylethylidene} B-D-Ribofuranose (12)70 [61200 -72 (c 1 CHC13); 'H NMR(CDC13): 8 5.43 (s, 1H, H-l), 4.69 (d, J = 3.7 Hz, 1H, H-4), 4.27 (d, J = 5.5 Hz, 1H, H-2), 3.42 (dd, J = 3.8, 7.2 Hz, 1H, H-5a), 3.30 (d, J = 7.2 Hz, 1H, H-Sb), 1.46, 1.29 (2 s, 3 H each, C(Me)2 ); 13C NMR (CDC13): 112.0, 99.7 (C-l), 81.1 (C-2), 79.3 (C-3), 77.4 (C-4), 63.0 (C-5), 25.8, 25.1 (CMe2). (3aS, 4S, 7R, 7aR)-2,2-Dimethyl-4-nitromethyl-tetrahydro-[1,3]dioxolo[4,5- c]pyridine-5,7-diol (16) To a stirred solution of nitrone 11 (46.7 mg, 0.25 mmol) in methanol (2.0 ml) was added nitromethane (61 mg, 1.0 mmol) and MeONa (13.0 mg, 0.25 mmol). After stirring at r.t for 8 h, the reaction mixture was cooled to 0°C and 3 N HCl was added to adjust the solution pH to 7. After condensation, ethyl acetate and water (10 ml each) were added to the flash. The aqueous layer was separated and extracted with ethyl acetate twice (2 x 10 ml). The combined organic extracts were washed with water, brine, dried over MgSO4. Condensation followed by column chromatography (chloroform : methanol 25 : 1) gave 16 as a white solid (50.8 mg, 82 %). [6120.) -352 (c 1 CHC13); 1H NMR (DMSO): 8 7.87 (br s, 1H, N-OH), 4.63 (dd, J =. 7.7, 13.0 Hz, 1H, CH2N02), 4.56 (dd, J = 3.6, 13.0 Hz, 1H, CH2NO2), 4.32 (t, J = 4.2 Hz, 1H, H-7a), 4.09 (ddd, J = 3.9, 5.0, 11.3 Hz, 1H, H-7), 4.01 (dd, J = 4.5, 9.1 Hz, 1H, H-3a), 3.23; (ddd, J = 3.5, 7.7, 11.2 Hz, 1H, H-4), 3.08 (dd, J = 5.1, 11.3 Hz, 1H, H-6), 2.73 (t, J = 11.3 Hz, 1H, H-6’), 1.50, 1.34 (2 s, 3 H each, CMe2); l3C NMR(DMSO): 5108.59, 75.47, 74.73, 72.77, 64.10, 61.95, 57.70, 27.37, 25.68; NOESYID: no NOE observed between H-4 and H-3a; 169 (3aS, 4S, 7R, 7aR)-4-Aminomethyl-2,2-dimethyl-hexahydro-[l,3]dioxolo[4,5- c]pyridine-7-ol (19) To a solution of 16 (0.128 g, 0.5 mmol) in methanol (5 ml) was added Pd/C (10 %, 40 mg), the solution was stirred under H2 atmosphere (1 atm) for 8 h. Filtration of the solution through celite followed by removal of the solvent gave the hydrogenated product 19 in quantitative yield ( 0.101 g, 100 %). [01200 (c 1 CHCl3); lH NMR (DMSO): 5 4.61 (dd, J = 4.6, 11.3 Hz, 1H), 4.34 (t, J = 4.2 Hz, 1H), 4.27 (t, J = 4.2 Hz, 1H), 4.07 (ddd, J = 4.2, 4.9, 11.3 Hz, 1H), 4.00 (dd, J = 4.6, 9.4 Hz, 1H), 3.81-3.84 (m, 1H), 3.67-3.74 (m, 1H), 3.06-3.12 (m 1H), 3.03 (dd, J = 5.5, 9.8 Hz, 1H), 2.90-2.98 (m, 1H), 2.77 (dd, J = 6.5, 11.5 Hz, 1H), 1.47, 1.33 (2 s, 3 H each, 2 CMe2); '3 C NMR(DMSO): 5 74.83, 74.40, 62.37, 57.75, 57.77, 53.50, 27.49, 25.80. Cycloaddition of nitrone (11) with allyl alcohol: To a solution of nitrone (93.5 mg, 0.5 mmol) in toluene (5.0 ml) was added allyl alcohol (145 mg, 2.5 mmol). The reaction mixture was stirred under reflux for 6 h. Removal of the solvent gave a syrup (112 mg, 92 %, 89 : 11 by NMR), which is a diastereomeric mixture of the cyclization products and can not be separated by column chromatography. Crystallization from chloroform gave the 2S anomer 25 as white crystals (75 mg, 61 %). ' Upon standing at —17°C for 48 h, the 2R anomer 27 was obtained as a crystal from the mother liquid (14 mg, 11 %). (ZS, 3aS, 48, SR, 6R) 2-methanol-4,5-0-isopropylidene-6-hydroxyl-hexahydro-2H- Isoxazolo [2,3a] pyridine (25) 170 1H NMR (DMSO): 5 4.28-4.42 (br, 2 H, 2 OH), 4.23 (t, J = 4.3 Hz, 1H, H-5), 3.98-4.03 (m, 3 H, H-2, H-4, H-6), 3.40-3.42 (m, 2 H, CH2OH), 3.16 (dd, J = 4.9, 10.1 Hz, 1H, H- 7), 2.65-2.72 (m, 2 H, H-3a, H-7), 2.08-2.14 (m, 1H, H-3), 1.98 (ddd, J = 8.4, 11.2, 18.0 Hz, 1H, H-3’). 1.45, 1.32 (2 s, 3 H each, 2 CMe2); 13C NMR (DMSO): 5 108.02, 76.63, 75.81, 74.59, 63.88, 63.70, 62.61, 53.95, 34.91, 27.13, 25.36; Calculated for C11H19N05 C, 53.87, H, 7.81, N, 5.71; Found C 53.60, H, 7.65, N, 5.86. (2R, 3aS, 4S, 5R, 6R) 2-methanol-4,5-0-isopropylidene-6-hydroxyl-hexahydro-2H- Isoxazolo [2,3a] pyridine (27) 1H NMR (DMSO): 5 4.49 (d, J = 2.1 Hz, 1H, C-6 0H), 4.27 (t, J = 5.5 Hz, 1H, CH2OH), 4.23 (t, J = 4.2 Hz, 1H, H-S), 3.98-4.02 (m, 3 H, H-2, H-4, H-6), 3.37-3.43 (m, 2 H, CH2OH), 3.16 (dd, J = 5.7, 10.2 Hz, 1H, H-7), 2.65-2.71 (m, 2H, H-3a, H-7’), 2.12 (ddd, J = 4.9, 7.0, 16.8 Hz, 1H, H-3), 1.98 (ddd, J = 8.5, 8.5, 16.8 Hz, 1H, H-3’), 1.46, 1.32 (2 s, 3 H each, 2 CMe2); 13C NMR (DMSO): 5 76.59, 75.78, 74.59, 63.87, 63.62, 62.57, 53.95, 34.88, 27.13, 25.36; NOESYID: no NOE observed between H-2 and H-3a; Calculated for C11H19N05 C, 53.87, H, 7.81, N, 5.71; Found C 53.89, H, 7.88, N, 5.79. Cycloaddition of Nitrone 11 with Vinyl ethyl ether To a solution of nitrone 11 (93.5 mg, 0.5 mmol) in chloroform (5.0 ml) was added ethyl vinyl ether (180 mg, 2.5 mmol). After sealing the flask, the reaction mixture was stirred at 62°C 4 h. Removal of the solvent gave a syrup, which is a diastereomeric mixture of the cyclization products (121.8 mg, 94 %). The diastereomeric mixture was separated by 171 column chromatography gave the pure 2R anomer 29 (58.3 mg, 45 %) and the 2S anomer 31 (40.1 mg, 31 %) (2S, 3aS, 48, SR, 6R) 2-ethoxy-4,5-0-isopropylidene—6-hydroxyl-hexahydro-ZH- Isoxazolo [2,3a] pyridine (29) [(11200 + 57.53 (c 1 CHC13); lH NMR (DMSO): 5 5.10 (t, J = 4.3 Hz, 1H, H-2), 4.26 (t, J = 4.8 Hz, 1H, H-5), 4.05 (t, J = 6.1 Hz, 1H, H-4), 3.99 (dt, J = 3.9, 9.9 Hz, 1H, H-6), 3.56-3.66 (m, 1H, OCH2CH3), 3.38-3.49 (m, 1H, OCH2CH3), 3.06-3.17 (m, 2 H, H-3a, H-7), 2.94 (dd, J = 9.9, 11.1 Hz, 1H, H—7’), 2.16-2.20 (m, 2 H, H-3, H-3’), 1.44, 1.31 (s, 3 H each, , 2 C(CH3)2), 1.12 (t, J = 7.0 Hz, 3H, OCH2CH3); 13C NMR (DMSO): 5 107.91 (C(CH3)2), 99.86(C-2), 75.56, 74.15, 62.07 , 61.68, 60.33, 52.33, 40.29 (C-3), 26.72, 25.00 (2 C(CH3)2), 14.31; NOESYID: no NOE observed between H-2 and H-3a; Calcd. for C21H31NO4 489.1846, found 489.1846; Calculated for C12H21N05 C, 55.58, H, 8.16, N, 5.40; Found C 55.70, H, 8.12, N, 5.10. (2R, 3aS, 48, SR, 6R) 2-ethoxy-4,5-O-isopropylidene-6-hydroxyl-hexahydro-ZH- Isoxazolo [2,3a] pyridine (31) [a]200 -105.56 (c 1 CHC13); 'H NMR (DMSO): 8 5.11 (t, J = 3.1 Hz, 1 H, H-2), 4.24 (t, J = 3.9 Hz, 1H, H-S), 4.04 (t, J = 5.4 Hz, 1H, H-4), 3.99 (m, 1H, H-6), 3.57-3.72 (m, 1H, 0CH2CH3), 3.40-3.53 (m, 1H, OCH2CH3), 3.25 (dd, J = 5.1, 9.4 Hz, 1H, H-7), 2.53-2.65 (m, 2 H, H3a, H-7’), 2.17-2.21 (m, 1H, H-3), 1.79-1.86 (m, 1H, H-3’), 1.45, 1.32 (2 s, 3 H each, 2 CMe2), 1.13 (t, J = 6.9 Hz, 3H, OCH2CH3); 108.09, 100.70, 75.24, 74.71, 65.15, 64.33, 62.22, 54.99, 40.38, 27.24, 25.38, 14.27; NOESYID: 11 % NOE observed 172 between H-2 and H-3a; Calculated for C12H21N05 C, 55.58, H, 8.16, N, 5.40; Found C 55.68, H, 8.19, N, 5.49. l,5-anhydro-2,3,4-tri-O-acetyl-6-deoxy-6-iodo-D-glucitol (36) To a solution of toluene (25 ml) containing triphenylphosphine (3.05 g, 12.0 mmol), iodine (2.98 g, 12.0 mmol) and imidazole (2.45 g, 36.0 mmol) were added 1,5-anhydro- D-glucitol (1.64 g, 10 mmol). The reaction mixture was stirred at 701151 for 3 h and then cooled to r.t. After adding water (30 ml), the mixture was stirred vigorously for 30 min. After separation, the toluene phase was extracted with water (3 x 20 ml). The combined water extracts were condensed and dried under high vacuum. Pyridine (40 ml) was added to dissolve the residue, followed by the addition of acetic anhydride (20 ml). The mixture was stirred at r.t. for 12 h. After concentration of the reaction mixture, EtOAc (50 ml) and aqueous saturated NaHCO3 (50 ml) were added to the flask. The aqueous phase was separated and extracted with EtOAc (30 ml) once. The combined organic phase was washed with water, brine, and dried over MgSO4. Column chromatography gave the iodo product 36 (2.60 g, 65 %). 1H NMR (CDC13): 5 5.18 (t, J = 9.4 Hz, 1H), 4.98 (ddd, J = 5.8, 9.6, 10.6 Hz, 1H, H-), 4.86 (t, J = 9.3 Hz, 1H), 4,16 (dd, J = 5.7, 10.3 Hz, 1H), 3.36- 3.26 (m, 3H, H-6, H-6’, H-l), 3.10 (dd, J = 7.6, 11.7 Hz, 1H, H-l’), 2.04, 2.01, 2.00 (s, 3 H each, 3 OAc); l3C NMR(CDC13): 5 170.18, 169.60, 169.35 (3 OAc), 77.27, 73.16, 72.22, 68.94, 66.56, 20.62, 20.59, 20.59 (3 OAc), 3.55 (C-6). MethyI-6-deoxy-6-iodo-2,3,4-tri-O-acetyl-a-D-glucopyranoside (33) Methyl-6-deoxy-6-iodo-2,3,4-tri-O-acetyl-a-D-glucopyranoside was prepared from methyl-a-D-glucopyranoside using the same sequences as for the preparation of 1,5- 173 anhydro-2,3,4-tri-O-acetyl-6-deoxy-6-iodo-D-g1ucitol (68 %). [a]20[) + 113.5 (c 1 CHC13); 'H NMR (CDC13): 8 5.45 (dd, J = 9.4, 10.0 Hz, 1H, H-3), 4.94 (d, J = 3.7 Hz, 1H, H-l), 4.87 (dd, J = 3.7, 10.1 Hz, 1H, H-2), 4.86 (t, J = 10.1 Hz, 1H, H-4), 3.77 (ddd, J = 2.4, 9.5, 8.3 Hz, 1H, H-5), 3.46 (s, 3H, OMe), 3.28 (dd, J = 2.4, 10.9 Hz, 1H, H-6), 3.12 (dd, J = 8.3, 10.9 Hz, 1H, H-6’), 2.05, 2.03, 1.98 (s, 3 H each, 3 OAc); ”C NMR(CDC13): 8 170.1, 170.0, 169.6 (3 OAc), 96.69 (C-1 ), 72.45, 70.89, 69.65, 68.62, 55.74, 20.69, 20.67, 20.64 (3 OAc), 3.59 (C-6). 2,6-anhydro-l-deoxy-3,4,5-tri-O-acetate-L-xylo-Hex—l-enitol (37) To a solution of 1,5-anhydro-2,3,4-tri-0-acetyl-6-deoxy-6-iodo-D-glucitol (2.0 g, 5.0 mmol) in anhydrous DMF (20 ml) was added DBU (4.8 ml, 30 mmol) and the mixture was stirred at 75°C for 3, h. After cooling to r.t, water and ethyl acetate (30 ml each) were added to the reaction mixture. The aqueous layer was separated and extracted with ethyl acetate twice (2 x 20 ml). The combined organic extracts were washed with saturated aqueous NaHCOg, water, brine, dried over MgS04. Condensation followed by column chromatography (ethyl acetate: hexane l : 5) gave 37 as a white solid (1.03 g, 76 %). [011200 + 9.00 (c 1 CHC13); 1H NMR (CDC13): 5 5.47 (d, J = 7.8 Hz, 1H, H-3), 4.99 (t, J = 7.8 Hz, 1H, H-4), 4.93 (m, 1H, H-5), 4.60 (t, J = 1.5 Hz, 1H, H-l’), 4.35 (t, J = 1.5 Hz, 1H, H-l), 4.07 (dd, J = 4.9, 11.5 Hz, 1H, H-6’), 3.45 (dd, J = 8.3, 11.5 Hz, 1H, H-6), 2.12, 2.10, 2.09 (3 s, 9 H, 3 x OAc), l3C NMR(CDC13): 5 168.9, 169.4, 169.4 (3 x OAc), 153.5 (C-l), 95.4 (C-2), 72.0 68.5 (2 Carbon) (C-3, 4, 5), 66.7 (C-6), 20.3, 20.3, 20.6 (3 Carbon, 3 CH3). Methyl 2,3,4 tri-O-acetyl 6-deoxy-or—D-xylohex-5-enopyranoside (34) 174 Methyl 2,3,4 tri-O-acetyl 6-deoxy-01—D-xylohex-5-en0pyranoside was prepared from Methyl-6-deoxy-6-iodo-2,3,4-tri-O-acetyl-01-D-glucopyranoside using the same sequences as for the preparation of 2,6-anhydro-1-deoxy-3,4,5-tri-O-acetate-L-xylo-Hex- l-enitol (75 %). [a]200 (c 1 CHC13); 'H NMR (CDC13): 8 5.42-5.48 (m, 2H, H-6, 11-1), 4.95-4.98 (m, 2H, H-6’, H-3), 4.76 (t, J = 1.8 Hz, 1H, H-4), 4.58 (t, J = 1.8 Hz, 1H, H-2), 3.42 (s, 3 H, OMe), 2.09, 2.05, 2.00 (3 s, 3 OAc); l3C WR(CDC13): 5 170.1, 169.8, 169.5 (3 OAc), 97.74, 97.46, 70.63, 69.68, 69.37, 55.53 (OMe), 20.67, 20.66, 20.64 (3 OAc). (3aR, 3’S, 4R, 4’R, S’R, 6’S, 9aS, 9bS)-4-hydroxy-6'-methoxy-2,2- dimethyldecahydrospirol1,3-dioxolo[4,5-c]isoxazolo[2,3-a]pyridine-8,2'-pyran]- 3',4',5'-triyl triacetate (39) A solution of toluene (10.0ml) containing nitrone (93.5 mg, 0.5 mmol) and methyl 6- deoxy-a—D-xylo-hex-S-enopyranoside (151 mg, 0.5 mmol) was stirred under reflux for 12 h. Removal of the solvent followed by column chromatography gave the cyclization product 39 (235 mg, 96 %). [6:120D +3741 (c 1 CHC13); 'H NMR (DMSO): 8 8.12 (s, 1H, 0H), 5.17 (t, J = 8.1 Hz, 1H, H-4), 4.99 (d, J = 3.5 Hz, 1H, H-2), 4.98(d, J = 8.0 Hz, 1H, H-5, ), 4.94 (dd, J = 3.5, 8.2 Hz, 1H, H-3), 4.30(dd, J = 4.4, 5.4 Hz, 1H, H12), 4.26 (t, J = 5.9 Hz, 1H, H-1 1), 3.92 (dt, J -'= 4.4, 10.0 Hz, 1H, H-l3), 3.43 (s, 3H, OMe), 3.31 (m, 1H, H-9), 3.12 (dd, J = 4.2, 11.5 Hz, 1H, H-l4), 2.90 (t, J = 10.8 Hz, 1H, H-14), 2.60 (dd, J = 6.2, 12.8 Hz, 1H, H-lO), 2.23 (dd, J= 10.7. 12.8 Hz, 1H, H-IO’), 2.05, 2.01, 2.00 (s, 3 OAc), 1.46, 1.32 (s, 2 Me); l3C NMR (DMSO): 5168.56, 168.17, 167.82 (3 OAc), 175 108.04, 104.49 (C-6), 95.98 (C-2), 78.30, 74.71, 73.88, 69.22, 68.65, 67.82, 62.60, 61.28, 55.49, 53.51, 40.21(C-10), 26.57, 24.78, 19.44, 19.38, 19.38; NOESYID: NOE H10 - H10’ 25 %, H10 — HS’ < 1 %; HRFABMS; Calcd. for C21H31N04 489.1846, found 489.1846; Calculated for C21H31N012 C, 51.53, H, 6.38, N, 2.86; Found C 51.50, H, 6.44, N, 3.10. (3aR, 3’S, 4R, 4’R, 5’R, 9aS, 9bS)-4-hydroxy-2,2-dimethyldecahydrospiro[l,3- dioxolo[4,5-c]isoxazolo[2,3-a]pyridine-8,2'-pyran]-3',4',5'-triyl triacetate (39) To a stirred solution of nitrone (93.5 mg, 0.5 mmol) in toluene (5.0 ml) was added 2,6- anhydro-l-deoxy-3,4,5-tri-O-acetate-L-xylo-Hex-l-enitol (163 mg, 0.6 mmol). The reaction mixture was stirred under reflux for 10 h. Removal of the solvent followed by column chromatography gave the cyclization product as a single diastereomer 45 (204 mg, 89 %). 6' s 0r>-OA0 [61209 -12220 (c 1 CHC13); 'H NMR (CDC13): 8 5.13 (t, J = 31 "" "’OAc 9.8Hz, 1H, H-4), 5.02 (dd, J = 1.0, 10.2 Hz, 1H, H-S), 4.87- 4.95(m, 1H, H-3), 4.22 (t, J = 4.3 Hz, 1H, H-12), 4.05 (dd, J = 4.9, 8.3 Hz, 1H, H—1 1), 4.02 (t, J = 4.9 Hz, 1H, H-3, 2H), 3.81 (dd, J = 6.1, 11.0 Hz, 1H, H-2), 3.65 (t, J = 10.9 Hz, 1H, H-2’), 3.34 (dd, J = 5.2, 9.0 Hz, 1H, H-l4), 2.96 (br s, 1H), 2.69 (t, J = 9.5 Hz, 1H, 11-14:), 2.54- 2.62 (m, 1H, H-9), 2.40 (dd, J —-= 6.6, 13.0 Hz, 1H, H-10), 2.08 (dd, J = 10.3, 13.0 Hz, 1H, H-10’), 2.03, 1.97, 1.95 (3 OAc), 1.30, 1.42 (s, 3 H each, 2 CH3); l3C NMR(DMSO): 8 168.75, 168.61, 168.55 (3 OAc), 108.28, 103.57, 78.50, 74.78, 74.75, 70.34, 68.23, 65.99, 64.31, 56.63, 55.22; 42.11, 27.25. 25.38, 19.53, 19.47, 19.44; NOESYID: NOE 176 H3 — H3’ 30 %, H3 — H5” 15 %. Calculated for C20H29N011 C, 52.28, H, 6.36, N, 3.05; Found C 52.20, H, 6.42, N, 3.26. 177 5.8 References [1] Spiro, R. G. Glycobiology 2002, 12, 43R-56R. [2] Rye, C. S., Withers, S. G. Curr Opin Chem Biol 2000, 4, 573-580. [3] Vasella, A.; Davies, G. J.; Bohm, M. Curr. Opin. Chem. Biol. 2002, 6, 619-629. [4] Inoue, S.; Tsuruoka, T.; Niida, T. J. Antibiot. 1966, 19, 288-292. [5] Niwa, T.; Inoue, S.; Tsuruoka, T.; Koaze, Y.; Niida, T. Agric. Biol. Chem. 1970, 34, 966-968. [6] Karpas, A.; Fleet, G. W. J .; Dwek, R. A.; Petursson, S.; Narngoong, S. K.; Ramsden, N. G.; Jacob, G. S.; Rademacher, T. W. Proc. Natl. Acad. Sci. USA 1988, 85, 9229. [7] Dimitriadis, G. D.; Tessari, P.; Go, V. L. W.; Gerich, J. E. Metabolism 1985, 34, 261. [8] Humphries, M. J .; Matsumoto, K.; White, S. L.; Olden, K. Cancer Res. 1986, 46, 5215. [9] Goss, P. E.; Baker, M. A.; Carver, J. P.; Dennis, J. w. Clin. Cancer Res. 1995, 1, 935. [10] Compain, P.; Martin, O. R. Bioorg. Med. Chem. 2001, 9, 3077. [11] Bastida, A.; Fernandez-Mayoralas, A.; Arrayas, R. G.; Iradier, F.; Carretero, J. C.; Garcia-Junceda, E. Chem. Eur. J. 2001, 7, 2390. [12] Fedorov, A.; Shi, W.; Kicska, G.; Fedorov, E.; Tyler, P. C.; Fumeaux, R. H.; Hanson, J. C.; Gainsford, G. J.; Larese, J. Z.; Schramm, V. L.; Almo, S. C. Biochemistry 2001, 40, 853. [13] Miles, R. W.; Tyler, P. C.; Fumeaux, R. H.; Bagdassarian, C. K.; Schramm, V. L. Biochemistry 1998, 3 7, 8615. [14] Jakobsen, P.; Lundbeck, J. M.; Kristiansen, M.; Breinholt, J .; Demuth, H.; Pawlas, J.; Candela, M. P. T.; Andersen, B.; Westergaard, N.; Lundgren, K.; Asano, N. Bioorg. Med. Chem. 2001, 9, 733. [15] Lee, R. E.; Smith, M. D.; Pickering, L.; Fleet, G. W. J. Tetrahedron Lett. 1999, 40, 8689. [16] Lee, R. E.; Smith, M. D.; Nash, R. J.; Griffiths, R. C.; McNeil, M.; Grewal, R. K.; Yan, W.; Besra, G. S.; Brennan, P. J .; Fleet, G. W. J. Tetrahedron Lett. 1997, 38, 6733. 178 [17] Bollen, M.; Stalmans, w. Eur. J.Biochem. 1989, 181, 775. [18] Andersen, B.; Rassov, A.; Westergaard, N.; Lundgren, K. Biochem. J. 1999, 342, 545. [19] Hausler, H.; Kawakami, P.; Mlaker, E.; Severn, W. B.; Wrodnigg, T. M.; Stutz, A. E. J. Carbohydr. Chem. 2000, I9, 435. [20] Durantel, D.; Branza-Nichita, N.; Carrouee-Durantel, S.; Butters, T.D.; Dwek, R. A.; Zitzmann, N. J. Virol. 2001, 75, 8987. [21] Goss, P. E.; Baker, M. A.; Carver, J. P.; Dennis, J. W. Clin. Cancer Res. 1995, I, 935. [22] Nishimura, Y.; Satoh, T.; Adachi, H.; Kondo, S.; Takeuchi, T.; Azetaka, M.; Fukuyasu, H.; Lizuka, Y. J. Med. Chem. 1997, 40, 2626. [23] Goujon, J. Y.; Gueyrard, D.; Compain, P.; Martin, O. R.; Asano, N. Tetrahedron: Asymmetry 2003, I 4. 1969—1972. [24] Godin, G.; Compain, P.; Masson, G.; Martin, O. R. J. Org. Chem. 2002, 7, 6960— 6970. [25] O’Brien, J. L.; Tosin, M.; Murphy, P. V. Org. Lett. 2001, 3, 3353—3356. [26] Liang, X.; Andersch, J.; 3015, M.; J. Chem. Soc., Perkin Trans. 1 2001, 2136—2157. [27] Banba, Y.; Abe, C.; Nemoto, H.; Kato, A.; Adachi, 1.; Takahata, H. Tetrahedron: Asymmetry 2001, 12, 817—819. [28] Takahata, H.; Banba, Y.; Ouchi, H.; Nemoto, H. Org. Lett. 2003, 5, 2527—2529. [29] Ghosh, A. K.; Koltun, E. S.; Bilcer, G. Synthesis 2001, 1281—1301. [30] Kummeter, M.; Kazmaiér, U. Eur. J. Org. Chem. 2003, 3330—3334. [31] Kolb, H. C.; Van Nieuwenhze, M. S.; Sharpless, K. B. Chem. Rev. 1994, 94, 2483— 2547. [32] Li, G.; Chang, H-T; Sharpless, K. B. Angew. Chem. Int. EdEngl. 1996, 35, 451— 454; [33] Kolb, H. C.; Sharpless. K. B. in Asymmetric Aminohydroxylation, in Transition Metals for Organic Synthesis: Building Blocks and Fine Chemicals (Eds. M. Beller, C. Bolm), Wiley-VCH, Weinheim, 1998. [34] Leon A.G.M. van den Broek. Tetrahedron, 1996, 52, 4467-4478. 179 [3 5] Chiacchio, U.; Saita, M. G.; Crispino, L.; Gumina, G.; Mangiafico, S.; Pistara, V.; Romeo, G.; Pipemo, A.; De Clercq, E. Tetrahedron 2006, 62, 1171-1181. [36] Bashiardes, G.; Cano, C.; Mauze, B. Synlett 2005, 9, 1425-1428. [37] Evans, G. B.; F umeaux, R. H.; Hausler, H.; Larsen, J. S.; Tyler, P. C. J. Org. Chem. 2004, 69, 2217-2220. [38] Shing, T. K. M.; Fung, W. C.; Wong, C. H. J. Chem. Soc., Chem. Comm. 1994, 4, 449-50 ‘ [39] Merino, P.; Tejero, T. Molecules 1999, 4, 169-179. [40] Cardona, F.; Faggi, E.; Liguori, F.; Cacciarini M.; Goti, A. Tetrahedron Lett. 2003, 44, 2315—2318 [41] Merino, P.; Franco, S.; Merchan, F. L.: Tejero, T. Synlett 2000, 4, 442—454. [42] Koumbis, A. E.; Gallos, J. K. Curr. Org. Chem. 2003, 7, 585-628. [43] Cardona, F.; Faggi, E.; Liguori, E.; Cacciarini, M.; Goti, A. Tetrahedron Lett. 2003, 44, 2315—2318 [44] Peer, A.; Vasella, A. Helv. Chim. Acta 1999, 82, 1044. [45] Chevrier, C.; LeNouen, D.; Neuburger, M.; Defoina, A.; Tamusa, C. Tetrahedron Lett. 2004, 45, 5363—5366. [46] Marradi, M.; Cicchi, S.; Ignacio Delso, J .; Rosi, L.; Tejero, T.; Merinob, P.; Gotia, A. Tetrahedron Lett. 2005, 46, 1287—1290. [47] Merino, P.; Tejero, T.; Revuelta, J .; Romero, P.; Cicchi, S.; Mannucci, V.; Brandi A.; Goti A. Tetrahedron: Asymmetry 2003, 14, 367—379. [48] Duff, F. J.; Vivien, V.; Wightrnan, R. H. Chem. Commun. 2000, 21, 2127-2128. [49] Bergbreiter, D. E.; Lalonde, J. J. J. Org. Chem. 1987, 52, 1601. [50] Takashi, 0.; Shingo, F .; Keiji, M. J. Am. Chem. Soc. 2004, 126, 11790-11791. [51] Chetia, A.; Saikia, C. J .; Lekhok, K. C.; Boruah, R. C. Tetrahedron Lett. 2004, 45, 2649-2651. [52] Kanemasa, S.; Ito, K. Eur. J. Org. Chem. 2004,23, 4741-4753. [53] Palomo, C.; Oiarbide, M.; Mielgo, A. Angewandte Chemie, International Edition (2004), 43(41), 5442-5444. 180 [54] Westermann, Bernhard. Faculty of Natural Sciences, University of Paderbom, Paderbom, Germany. Angew. Chem, Inter. Ed. 2003, 422, 151-153. [55] Gothelf, K. V.; Jorgensen, K. A. Chem. Rev. 1998, 98, 863—909. [56] Kobayashi. S.; Jorgensen, K. A. Cycloaddition Reactions in Organic Synthesis, Wiley-VCH, Weinheim, 2002. [57] Silva, M. A.; Goodman, J. M. Tetrahedron, 2002, 58, 3667-3671. [58] Riick-Braun, K.; Freysoldt, T. H. E.; Wierschem, F. Chem. Soc. Rev. 2005, 34, 507- 516. [59] Herrera, R.; Nagarajan, A.; Morales, M. A.; Mendez, F.; Jimenez-Vazquez, H. A.; Zepeda, L. G.; Tamariz, J. J. Org. Chem. 2001, 66, 1252-1263. [60] Alibes, R.; Busque, F.; March, P. D.; F igueredo, M.; Font, J .; Parella, T. Tetrahedron 1998, 54, 10857-10878. [61] Ishar, M. P. S.; Singh, G.; Kumar, K.; Singh, R. Tetrahedron, 2000, 56, 7817-7828. [62] Frederickson, M. Tetrahedron. 1997, 53, 403-425. [63] Gothelf, K. V.; Jergensen, K. M. Chem. Rev. 1998,98, 8631909 [64] Ooi, H.; Urushibara, A.; Esumi, T.; Iwabuchi, Y.; Hatakeyama, S. Org. Lett. 2001, 3, 953-955. [65] Goti, A.; Cicci, S.; Fedi, V.; Nannelli, L.; Brandi, A. J. Org. Chem. 1997, 62, 3119. [66] DeShong, P.; Dicken, C. M.; Leginus, J. M.; Whittle, R. R. J. Am. Chem. SOC. 1984, 106, 5598-5602. [67] Libes, R.; Blanco, P.; de March P.; Figueredo, M.; Font, J .; Alvarez-Larenab, A.; Piniellab, J. F. Tetrahedron Lett. 2003, 44, 523—525. [68] Tamura, 0.; Gotanda, K.; Yoshino, J .; Morita, Y.; Terashima, R.; Kikuchi, M.; Miyawaki, T.; Mita, N.; Yamashita, M.; Ishibashi, H.; Sakamoto, M. J. Org. Chem. 2000, 65, 8544-8551 [69] Chan, M. F.; Hsiao, C.-N. Tetrahedron Lett. 1992, 33, 3567-3570. [70] F leetwood, A.; Hughes, N. A. Carbohyr. Res. 1999, 317, 204—209. 181 llllliljliltglill111111