+ All Categories
Home > Documents > Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

Date post: 18-Dec-2016
Category:
Upload: derek-horton
View: 266 times
Download: 0 times
Share this document with a friend
415
Transcript
Page 1: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50
Page 2: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

Advances in Carbohydrate Chemistry and Biochemistry

Volume 50

Page 3: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

This Page Intentionally Left Blank

Page 4: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

Advances in Carbohydrate Chemistry

and Biochemistry

Editor DEREK HORTON

Board of Advisors LAURENS ANDERSON STEPHEN J. ANGYAL STEPHEN HANESSIAN HANS H. BAER BENGT LINDBERG CLINTON E. BALLOU HANS PAULSEN JOHN S. BRIMACOMBE NATHAN SHARON J. GRANT BUCHANAN ROY L. WHISTLER

GUY G. S. DUTTON

Volume 50

ACADEMIC PRESS, INC. San Diego New York Boston

London Sydney Tokyo Toronto

Page 5: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

This book is printed on acid-free paper. @

Copyright 0 1994 by ACADEMIC PRESS, INC. All Rights Reserved. No part of this publication may be reproduced or transmitted in any form or by any means, electronic or mechanical, including photocopy, recording, or any information storage and retrieval system, without permission in writing from the publisher.

Academic Press, Inc. 525 B Street, Suite 1900, San Diego, California 92101-4495

United Kingdom Edition published by Academic Press Limited 24-28 Oval Road, London NWl 7DX

International Standard Serial Number: 0065-23 18

International Standard Book Number: 0- 12-007250-5

PRINTED IN THE UNITED STATES OF AMERICA

94 9 5 9 6 91 98 99 BB 9 8 1 6 5 4 3 2 1

Page 6: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

CONTENTS

PREFACE ................................................................. ix

Robert Stuart Tipson, 1906 . 1991

D . HORTON

... Text .................................................................... x1u

How Emil Fischer Was Led to the Lock and Key Concept for Enzyme Specificity

RAYMOND U . LEMIEUX AND ULRIKE SPOHR

I . Introduction ....................................................... 1 I1 . Asymmetric Induction .............................................. 2

I11 . Yeast Fermentations and Enzymes .................................... 7 IV . TheLockandKey Concept .......................................... 9 V . Insights on Enzyme Specificity ........................................ 13

VI . Concluding Remarks ................................................ 15 References ........................................................ 19

Anomeric-Oxygen Activation for Glycoside Synthesis: The Tnchloroacetimidate Method

RICHARD R . SCHMIDT AND WILLY KINZY

I . General Introduction to Glycoside Synthesis: Activation through Anomerio OxygenExchangeReactions .......................................... 21

I1 . Anomeric-Oxygen Activation: Anomeric 0-Alkylation .................... 23 I11 . Anomeric-Oxygen Activation: The Trichloroacetimidate Method ........... 25 IV . Other Anomeric-Oxygen Activation Methods ........................... 114 V . Conclusions ....................................................... 116

References ........................................................ 117

Synthetic Reactions of AIdonoIactones

ROSA M . DE LEDERKREMER AND OSCAR VARELA

I . Introduction ....................................................... 125 I1 . Acetalation of Aldonolactones ........................................ 125

V

Page 7: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

CONTENTS vi

111 . IV . V . VI .

VII . VIII .

IX . X .

XI . XI1 .

Acylaton and Etherification of Aldonolactones .......................... 132 Reaction of Aldonolactones with Hydrogen Bromide ..................... 134 Chain Elongation through the Aldonolactone Carbonyl Group ............. 136 Reaction of Aldonolactones with Alcohols .............................. 148 Reaction of Aldonolactones with Ammonia and Related Nucleophiles ....... 151 Reduction of Aldonolactones ......................................... 157 p-Elimination Reactions ............................................. 162 Synthesis of Deoxy Sugars from Aldonolactones ......................... 170 Glycosylation of Aldonolactones ...................................... 179 Aldonolactones as Chiral Precursors for the Synthesis of Natural Products . . . . 181 References ........................................................ 201

Molecular Structure of Lipid A, The Endotoxic Center of Bacterial Lipopolysaccharides

ULRICH WHRINGER. BUKO LINDNER. AND ERNST TH . RIETSCHEL

I . Introduction ....................................................... 211 I1 . Lipid A Definition and General Properties ............................. 213

IV . Synthetic Lipid A .................................................. 252 V . Conformation of Lipid A ............................................ 252 VI . Endotoxicity of LPS and Lipid A ...................................... 256

VII . Serology of Lipid A ................................................. 258 Synopsis: The Structure. Activity. and Function of Lipid A ................

111 . Primary Structure of Lipid A: Backbone. Polar Substituents. and Fatty Acids . 214

VIII . 260 References ........................................................ 265

Developments in the Synthesis of Glycopeptides Containing Glycosyl L.Asparagine. L.Serine. and L-Threonine

HARI G . GARG. KARSTEN VON DEM BRUCH. AND HORST KUNZ

I . Introduction ....................................................... I1 . N-Glycopeptides ...................................................

111 . 3-O-Glycopeptides of L-Serine or L-Threonine ........................... IV . V . Solid-Phasesynthesis ...............................................

VI . Enzymes as Tools for Glycopeptide Synthesis ........................... VII . Addendum ........................................................

References ........................................................

Binding of Glycopeptides to Proteins ..................................

Physicochemical Analyses of Oligosaccharide Determinants of Glycoproteins

ELIZABETH F . HOUNSELL

217 278 287 298 299 303 306 307

I . Introduction ....................................................... 311 I1 . Methods of Structural Analysis ....................................... 311

Page 8: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

CONTENTS vii

I11 . Purification and Profiling ............................................ 314 IV . Backbones and Core Regions of N- and 0-Linked Chains of Secreted and Plasma

Membrane Glycoproteins ............................................ 315 V . 325

VI . The Conformations and Molecular Recognition of Carbohydrate Determinants 332

VII . The Conformations and Molecular Recognition of Carbohydrate Determinants 343

References ........................................................ 345

Peripheral Substitutions of N- and 0-Linked Chains of Glycoproteins .......

Distant from the Protein Oligosaccharide Core of Glycoproteins ............

Adjacent to the Protein Moiety of Glycoproteins .........................

AUTHOR INDEX FOR VOLUME 50 ........................................... SUBJECT INDEX FOR VOLUME 50 ........................................... CUMULATIVE AUTHOR INDEX FOR VOLUMES 46-50 ...........................

351 378 386

CUMULATIVE INDEX FOR VOLUMES 46.50 ................................... 389

Page 9: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

This Page Intentionally Left Blank

Page 10: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

PREFACE

With this fiftieth volume the Advances reaches its half century, during which time the carbohydrate field has evolved dramatically across a broad range of formal scientific disciplines, both fundamental and applied. Over the years the chapters in this series have chronicled these developments for the benefit of the general reader and have concurrently provided, for the specialist, important critical insight into gaps in our knowledge. The rich legacy of the early carbohydrate literature remains a fruitful resource in addressing new problems with today’s superior tools of research.

Lemieux and Spohr (Alberta) here trace our understanding of enzyme specificity in broad perspective as they assess Emil Fischer’s “lock and key” concept advanced a century ago in relation to current ideas of molecular recognition. It may be noted that the very first article in Volume 1 of Ad- vances, by Claude s. Hudson, was devoted to the Fischer cyanohydrin syn- thesis and the consequences of asymmetric induction.

The task of interpreting chemical transformations and the logical plan- ning of synthetic methods have been traditionally difficult with the carbohy- drates because of their polyfunctionality and complex stereochemical archi- tecture. The vast body of empirical literature is daunting to the newcomer to the field, and the synthesis of glycosides by endless permutations of the traditional Koenigs - Knorr synthesis presents especial difficulty. A major step forward has resulted from the insightful thinking of R. R. Schmidt (Konstanz) toward the rational design of practical and versatile methodol- ogy for glycoside synthesis. His trichloroacetimidate method, here surveyed in comprehensive detail in a chapter with his colleague W. Kinzy (Basel), constitutes one of the most imaginative approaches to an important problem in synthetic methodology. Their chapter will undoubtedly comprise a key reference source for numerous researchers for many years to come.

Although the use of abundant sugars as starting materials for chiral syn- thesis has received considerable attention, the ready availability of many aldonolactones is less well recognized by “mainstream” synthetic organic chemists. The chapter here contributed by de Lederkremer and Varela (Buenos Aires) provides a comprehensive overview of the practical potential of these cyclic esters and complements the more specialized contribution on gulonolactones by Crawford in Volume 38.

The biomedical importance of infections by gram-negative pathogens and the consequences of septic shock have drawn much attention to lipid A, the toxic subcomponent of the lipopolysaccharide endotoxin of these orga- nisms. A comprehensive account of the chemical structures and biological behavior of the lipid A structures is presented here by Zahrihnger, Lindner, and Rietschel. The chapter incorporates much of their own work from the

ix

Page 11: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

X PREFACE

Borstel laboratory where Westphal animated his pioneering work on bacte- rial lipopolysaccharides.

The sugar - amino acid linkage point in glycoproteins and proteoglycans, involving the side-chain nitrogen atom of L-asparagine or the hydroxyl group of L-serine or L-threonine, is a key structural region of these glycocon- jugates. Garg, von dem Bruch, and Kunz (Boston and Maim) now survey developments in the synthesis of glycopeptides containing these linkages, updating with significant new work the earlier reports in Volume 25 by Marshall and Neuberger and in Volume 43 by Jeanloz and Garg.

The final chapter, by Hounsell (London), also relates to an important aspect of glycoprotein structure, namely the structures and shapes, as deter- mined by physicochemical methods, of oligosaccharide determinants of glycoproteins that are antigens and targets for binding of adhesion mole- cules.

For most of its existence the Advances has been guided by the breadth of scientific insight and editorial expertise of two individuals, M. L. Wolfrom and R. S. Tipson. The obituary chapter in this volume records the life and scientific work of Robert Stuart Tipson, who contributed a chapter on the nucleic acids to Volume 1 and retired as Editor with the publication of Volume 48.

Washington, D. C. April, 1994

DEREK HORTON

Page 12: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

This Page Intentionally Left Blank

Page 13: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50
Page 14: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

ROBERT STUART TIPSON

1906- 1991

Bob Tipson made contributions to carbohydrate chemistry during more than six decades and exerted a far-reaching influence on the published litera- ture in the field. Although he was born and educated in England, his subse- quent career developed in North America. He was associated with Advances in Carbohydrate Chemistry since its inception in 1945 and served in an editorial capacity with the series until his death on July 13, 199 1, near his home in Kensington, Maryland.

When Robert was born on November 23,1906, his parents were living in a rural hamlet named Wadshelf in the county of Derbyshire. He was the first of their five children. His father, Herbert James Tipson, was a teacher, as was his mother, Mary Jane (nCe Stuart). Their son did not attend formal elemen- tary school and was tutored at home by his mother until the age of ten. Four daughters were subsequently born to Herbert and Mary Tipson between 1906 and 1920; the second died in early childhood, at which time the family moved to Coventry, a nearby city in the Midlands of England.

Herbert Tipson was an all-round academic with a flair for mathematics and the precise use of language, and he was also a gifted artist and a lover of gardening. For many years he taught at the Coventry Technical School, and his mind retained sharp focus on mathematical concepts even into his ninth decade. These attributes were clearly passed on to his son who, like his father, received his secondary education as a day pupil at the Bablake School in Coventry, a traditional and highly regarded English boys’ school, admission to which required early demonstration of high academic potential.

At Bablake School the young Robert Tipson followed the established rigorous and structured curriculum, which emphasized academic achieve- ment and leadership qualities. He proved to be an outstanding student and passed in sequence the School Certificate and Higher School Certificate with high marks and gained university matriculation in 1924. His keen interest in chemistry led him to enter the nearby University of Birmingham, where Professor W. N. (later Sir Norman) Haworth was shortly to bring his world- renowned school of carbohydrate chemistry, and Tipson received the B.Sc. degree with First Class Honours in 1927.

CopyriShlO 1994 by Academic Ress, Inc. AU rights of reproduction in any form reserved.

xiii

Page 15: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

XiV ROBERT STUART TIPSON

As the top student in his class, Tipson was awarded the Priestley Research Scholarship and began his research work on carbohydrates under the direc- tion of Professor Haworth. A year later, under support from a teaching scholarship, he served as a junior instructor in organic chemistry in Bir- mingham and associated with other future leaders in the carbohydrate field, notably Maurice Stacey, Fred Smith, and J. K. N. Jones, who were then progressing through their studies at Birmingham. Haworth was a hard and demanding taskmaster who insisted on long hours in the laboratory, but the young R. Stuart Tipson (he was proud to emphasize the Scottish ancestry on his mother’s side) nevertheless found time to enjoy sports and especially music. The drum set and piccolo of his school days had given way to the saxophone, and with a small group he played the popular music of the twenties at dances and other university functions. He also taught evening classes in chemistry and mathematics at the City of Coventry Technical College.

His research in Birmingham during 1927 - 1929 focused on fundamental studies on methylated mannopyranosides and the plant-storage fructan, inulin, and also with one of the earliest applied studies, sponsored by the British Empire Cancer Campaign, on the isolation of tobacco “tar” and the study of its constituents. However, he was eager to enlarge his horizons, and with the encouragement of Professor Haworth he left Birmingham in August 1929 for Montreal, Canada, to work with Haworth’s friend Professor Harold Hibbert in the Department of Industrial and Cellulose Chemistry at McGill University. He worked with Hibbert on the structure of another fructan, the bacterial polysaccharide levan, and the results led to his first publication, which appeared in 1930 in the Journal of the American Chemical Society, as well as a second report that appeared in the following year. While at McGill he also gave instruction on the technique of Pregl’s microanalysis, and for a short time he worked in Prince Rupert, British Columbia, conducting a study on fish oils for the Fisheries Experimental Station (Pacific).

After only a year in Canada, Tipson moved in August 1930 to New York to accept a position as a research assistant to P. A. Levene at the Rockefeller Institute for Medical Research, where he began a phase of his research career that was most productive and lasted for nine years. By 1932 his work with Levene had resulted in no fewer than eight publications, mostly in the Journal of Biological Chemistry. This series of reports established the correct ring structures of the methyl glycosides of D-ribose and demonstrated that the natural purine (and later pyrimidine) ribonucleosides have the sugar in the furanose ring form.

Tipson was able to assemble the research results from his Birmingham days, along with the work with Hibbert and that with Levene, into a pro- digious Ph.D. thesis “Studies in the Carbohydrate Group,” which he suc-

Page 16: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

ROBERT STUART TIPSON xv

cessfully defended in June 1932 before the Birmingham examiners, Profes- sor Haworth and Dr. E. L. (later Sir Edmund) Hirst, and the external examiner, Professor (later Sir Ian) Heilbron.

During Tipson’s return to New York from a trip to England the year before, he met on board ship a young lady, Constance Goodwin, from Asbury Park, New Jersey, who became his wife some months later. Her quiet and gentle personality was in sharp contrast to his forthright and outspoken demeanor, but she was nevertheless able to assert her influence in indirect ways, and she provided an excellent complement to the sometimes domi- neering mannerisms and quirky humor of her husband. The couple were devoted companions until Connie’s death in 1985; they had no children.

Levene, a physician by training, was by all accounts a true genius, a self-taught chemist and pioneer biochemist. The environment of the Rocke- feller Institute, with its provision for research opportunities for younger scientists, was a perfect situation for Tipson to exercise his experimental research talents to the fullest while absorbing the drive and dedication mani- fested by Levene in addressing immensely difficult problems of the chemis- try of life processes.

Tipson devoted most of his years in Levene’s laboratory accomplishing seminal work on the components of nucleic acids. To determine the ring forms of the ribose component of the ribonucleosides he applied Haworth’s methylation technique and established the furanoid structure for the sugar in adenosine, guanosine, uridine, and thymidine. He showed that formation of a monotrityl ether is not a reliable proof for the presence of a primary alcohol group in a nucleoside, whereas a tosyl ester that is readily displaced by iodide affords clear evidence that the ester is at the 5-position ofthe pentofuranose. Acetonation of ribonucleosides was shown to give the 2’,3’-O-isopropyl- idene derivatives, which were to become extensively used in nucleoside and nucleotide chemistry, and were utilized by Tipson in the first chemical preparation of a ribonucleotide, inosinic acid.

Structural work on the nucleic acids by the traditional techniques of the 1930s provided challenges of formidable complexity, especially as the pro- pensity of phosphate groups to migrate was not then recognized. Neverthe- less, as early as 1935 Levene and Tipson advanced the accepted backbone structure for DNA when they formulated an oligonucleotide ofdeoxyribonu- cleic acid as having 3’ - 5’ phosphate diester linkages between the furanose sugar components.

Over thirty publications resulted from Tipson’s work in Levene’s labora- tory. Along with the work on nucleic acid components, he also studied the structures of gum arabic and other plant gums, and conducted a range of synthetic investigations on sugars, with particular emphasis on uronic acids and 5-carbon ketoses. His 1939 observation that acetylated glycosyl halides

Page 17: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

xvi ROBERT STUART TIPSON

react with oxyanions to give products having the trans configuration of substituents at the anomeric position and the neighboring carbon atom was popularized two decades later by B. R. Baker as the “trans rule,” and Tipson made strenuous efforts in later literature to establish his own prior develop- ment of this concept.

Following the death of Levene in 1939, Tipson’s position as a research associate at the Rockefeller Institute came to an end. He had held a part-time appointment as a lecturer in advanced biochemistry at Brooklyn College during 1938 - 1939, but in August 1939 he accepted a position at the Mellon Institute, and the Tipsons moved from their home on Long Island to Pitts- burgh. He was affiliated with Dr. L. H. Cretcher, who headed the Depart- ment of Research in Pure Chemistry at the institute, which was then asso- ciated with the University of Pittsburgh. The work at the Mellon Institute was largely directed toward applied problems, including quinoline deriva- tives as antimalarial drugs, cinchona alkaloids as antipneumococcal agents, and the chemistry of alloxan.

Some 40 research articles resulted from Tipson’s 18 years at the Mellon Institute, and they demonstrate that he was able to sustain some of his interest in carbohydrate chemistry, and he continued to study the reactions of sulfonic esters with sodium iodide. In 1945 he compiled his published work into a senior thesis for the D.Sc. degree that was awarded by the University of Birmingham. However, a considerable proportion of the re- search at the Mellon Institute was never published because of patent restric- tions. This was particularly true for his work on carbohydrates and other organic compounds conducted after July 1952, when he was assigned to the Parke, Davis and Company Fellowship in Medicinal Chemistry to synthe- size potential antiviral and anticancer agents.

Tipson always enjoyed and took great pride in precise scientific writing. When the Advances in Carbohydrate Chemistry series was launched in 1945 under the editorship of W. W. Pigman and M. L. Wolfrom, he wrote a comprehensive article for Volume 1 on the chemistry of the nucleic acids that is to this day a model of historical accuracy, careful and economical use of language, and clear interpretation of experimental data based on thor- oughly characterized crystalline compounds.

Equally precise and meticulous as an experimentalist, he devoted time during his days at Pittsburgh to write comprehensive articles on such practi- cal techniques as crystallization, vacuum distillation, and sublimation, which were published in the Weissberger Techniques of Organic Chemistry series. His preoccupation with careful experimental techniques and their accurate recording in the literature remained with him always. He abhorred vague descriptions of procedures, speculative interpretations not based on

Page 18: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

ROBERT STUART TIPSON xvii

solid facts, failure to give proper credit to prior work, and any imperfections in the use of the English language.

Tipson’s long-standing interest in sulfonic esters led him to contribute a landmark article on carbohydrate sulfonates for Volume 8 of Advances in Carbohydrate Chemistry, which marked the start of his editorial involve- ment with the series; he joined as assistant editor to M. L. Wolfrom starting with Volume 9. This was the beginning of a long and fruitful association between Wolfrom and Tipson that assured researchers in the carbohydrate field of a regular series of authoritative articles on a wide range of topics, both fundamental and applied, related to carbohydrates.

The Wolfrom - Tipson team demanded standards of scientific accuracy and careful writing style that characterized their own works; all authors, however eminent, were subjected equally to the analytic judgment and revi- sion of the two editors. While Wolfrom focused on the balance of literature citation and scientific interpretation, Tipson took infinite pains to ensure the exact use of scientific nomenclature and clear presentation in grammatically correct English. Authors from all around the world became familiar with the Tipson treatment of their manuscripts, with line-by-line corrections in Tip- son’s fine and precise writing, rendered in inks of various colors and often with mild expletives or pungent comments in the margin. The revised manuscript would be accompanied by a long handwritten letter with point- by-point queries to be addressed by the author. The final manuscript would go to the typesetter with all of the exact copyediting directions marked by Tipson; he had no confidence in publishers’ copyeditors. Any unwarranted alterations by the publishers or erroneous “corrections” on the proofs by the authors would promptly elicit a terse and vexatious letter from Tipson.

Some authors took exception to this “interference” with their personal writing style, even when it involved the rectification of obvious solecisms, but few did not finally appreciate the significant improvement in their pub- lished articles. The readership was undoubtedly the ultimate beneficiary of Tipson’s painstaking work.

Although Bob Tipson was not a gregarious person and, in fact, became quite reclusive in his later years, he had a fine appreciation of the personal qualities of individuals he respected. This facet of his character is most evident in the sensitive and insightful articles he wrote for the Advances series on the life and work of two of his mentors, P. A. Levene and Harold Hibbert.

In August 1957, Dr. Tipson left Pittsburgh to accept a research appoint- ment in Washington, D.C., at the National Bureau of Standards, where he assumed a position in the prestigious research laboratory headed by Dr. Horace S. Isbell. Here he had an opportunity to devote his efforts full-time to

Page 19: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

xviii ROBERT STUART TIPSON

carbohydrate chemistry. He remained at the Bureau until his formal retire- ment in 1972. The Tipsons’ new home in Kensington, Maryland, close to the Capital Beltway that encircles Washington, became the office for all of Bob’s editorial work for more than 30 years.

While the broad mission of the National Bureau of Standards was con- cerned with standard reference materials, Dr. Isbell centered the work of his laboratory on his long interest in the carbohydrates and on the use of physical methods in their characterization. Infrared spectroscopy had shown promise in providing structural and conformational information on carbohydrates and their derivatives, and Isbell invited Tipson to conduct detailed infrared studies on the extensive collection of carbohydrate samples maintained by Isbell. The series of publications that rapidly resulted furnished a basis for assigning conformations to pyranoid sugars and their derivatives. Although this work was later to be overshadowed by application of the much more powerful technique of nuclear magnetic resonance spectroscopy, the Isbell - Tipson work helped to define the molecular shapes involved and the termi- nology required for their description.

In addition to these physical studies at the Bureau, Tipson was able to return to his synthetic interests, both alone and in collaboration with other staff members. He was especially pleased to prepare D-talose in crystalline form, an accomplishment that had eluded Emil Fischer. Pursuing his long- standing interest in the reaction of sulfonic esters with iodide and following an earlier observation that the tetratosyl ester of erythritol is converted into butadiene by the action of sodium iodide and zinc, he demonstrated (with A. Cohen) that nonterminal unsaturation may be conveniently introduced into alditol derivatives by reaction of contiguous secondary sulfonates with so- dium iodide and zinc dust in boiling N,N-dimethylformamide. This Tipson - Cohen reaction subsequently proved of great utility in other hands for the conversion of more complex carbohydrate structures into vicinal dideoxy derivatives.

While in government service, Tipson undertook special assignments from the U.S. Congress and in 1964- 1965 spent nine months as a consultant to the Surveys and Investigations Staff of the House Appropriations Commit- tee. For this work he received awards for outstanding service, although he did not enjoy the extensive travel that was required nor the dislocation from the editorial work that normally occupied a major part of each of his days at home. Following the retirement of Isbell from the National Bureau of Stan- dards in 1968, the orientation of the laboratory was directed progressively toward targets outside the carbohydrate area, and in 1972 Tipson elected to retire from the Bureau and concentrate his efforts full-time on writing and editorial work.

Page 20: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

ROBERT STUART TIPSON xix

Although Tipson set out with stellar credentials in his career as a chemist and sustained a remarkably productive scientific life, he was never able to establish himself as an independent academic who would lead a group of research students. He had the good fortune to associate with such towering figures in the carbohydrate field as Haworth, Levene, Wolfrom, and Isbell. However these long-standing liaisons with major leaders may at the same time have deprived him of the opportunity to exert independence and to develop as the animator of a research group of his own.

Tipson was a competent lecturer when he remained close to the technical material that he was presenting, and he could carry an argument forcefully in discussion, but he was much more comfortable with the written word when interacting with other scientists. He was a regular participant in major scien- tific meetings during much of his career and traveled at regular intervals to visit his native England, but he later tired of travel, especially after his consulting stint for Congress. Subsequent to that time he was rarely seen outside his Kensington home or the weekend cottage that he and Connie enjoyed and where they would happily receive friends and colleagues.

He was blunt and outspoken, had little interest in political finesse, and his written comments often projected a gruff and sometimes intolerant image. It is true that he had scant tolerance for fools, frauds, or pompous individuals, but those who had known him only through their “Tipsonized” manuscripts and the accompanying multicolored, handwritten letters were often sur- prised upon visiting him at home to find a kind and mild-mannered host who projected a friendly and engaging informality. A man of moderate stature and trim build, he enjoyed robust health for most of his life, despite his heavy smoking habit. He participated in a range ofindividual sports in his younger days, and later maintained his enjoyment of swimming and golf. His tastes were modest and his frugality sometimes reached the point of obsession, but he and Connie always delighted in extending the hospitality of their home to guests. The visitor would certainly be given a tour of the garden and also be shown Bob’s collection of memorabilia and possibly his exten- sive stamp collection. An overnight visitor might be aroused by an early serenade on the saxophone, generally as a prelude to Bob’s call to a copious English breakfast announced in his stentorian voice.

Throughout his career Tipson took a deep interest in organic nomencla- ture and played a leading role in the development of carbohydrate nomen- clature. As early as 1939 he was appointed to a standing committee of the American Chemical Society’s (ACS) Division of Sugar Chemistry that was in charge of standardizing terminology in the carbohydrate field. The nomen- clature recommendations developed by this committee were subsequently adopted by the ACS, and ongoing negotiations led subsequently to a revision

Page 21: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

xx ROBERT STUART TIPSON

that had joint British- American approval. Tipson continued for several years as head of the ACS committee and was involved with a joint commis- commission of the International Union of Pure and Applied Chemistry anc the International Union of Biochemistry in developing the first internationa set of recommendations for the nomenclature of carbohydrates. His enor. mous experience as a researcher and as an editor in the carbohydrate fielc provided an unmatched background of structural knowledge with which tc test proposals for new terminology.

He was one of the founding editors of the international journal Curbohy drute Research when it was established in 1964, and he remained active as ar editor of the journal for a quarter of a century. In this capacity he interactec with hundreds of researchers and became a familiar friend and unprejudicec critic to numerous scientists around the world, who benefited from hi! meticulous editing of their manuscripts, which he handled with astonishing swiftness. He was particularly insistent on the traditional literary use o punctuation in clarifying the text of complex material; he deplored tht journalistic style adopted by some authors. Members of the carbohydratt community coined the term “Tipsonized” for his treatment of their manu. scripts. His incisive revisions, often bearing marginal comments inspired bj the “Peanuts” cartoon series, bestowed impartially upon the mandarins o science as well as on the beginning investigator struggling with a language no his own, brought clarity and impact to countless texts that passed through hi! hands. The busy reader was the immediate beneficiary, but there are fea authors who did not ultimately recognize, after the initial trauma of retypini a heavily revised manuscript, the excellence in the presentation oftheir worl thanks to the unstinting efforts of Bob Tipson.

Tipson never lost touch with his early research on nucleosides and nucleo tides. In 1968 he joined with W. W. Zorbach in producing a manual, Syn thetic Procedures in Nucleic Acid Chemistry. This volume compiles detailec descriptions of key experimental procedures contributed by a selection o authors from around the world and includes a number of Tipson’s owl procedures. The success of this volume led 2 years later to the production of; second collection. Subsequently, with the collaboration of Leroy Townsenc as coeditor, a new and expanded series Nucleic Acid Chemistry was launchec and extended to four volumes, the final one being published in the last yea of Tipson’s life. Throughout these series the emphasis was on practicalit and thorough directions for guiding the novice experimentalist, as stressed ii the preface to the first volume: “The Editors have zealously avoided certaii detestable directions such as ‘processed in the usual manner’ . . . A re search worker with a reasonable skill in the art . . . may successfully repea the preparations without specialized training.”

Page 22: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

ROBERT STUART TIPSON xxi

The total scope of Bob Tipson’s many editorial and nomenclature activi- ties with house publications, professional society reports, and committee service is too extensive to list in detail. In addition to his long involvement with the journal Carbohydrate Research, his editorship of Advances in Car- bohydrate Chemistry and Biochemistry extended through Volume 48, pub- lished in 1990, and this completed a remarkable and profoundly influential 36 years of editorial activity with the series.

Dr. Tipson’s involvement in scientific societies included life memberships in the Royal Society of Chemistry, the American Society of Biological Chemists, and the American Chemical Society. He helped to organize the first Gordon Conference on Carbohydrates. He worked actively on behalf of the Carbohydrate Division of the ACS, serving a term as an officer of the division. In addition to his key role in the division’s Nomenclature Com- mittee, he edited for many years the abstracts of papers presented at the meetings of the division. His outstanding contributions to the carbohydrate field were recognized by the ACS in 197 1 with the conferring ofthe Claude S. Hudson Award, and, in 1986, the Carbohydrate Division honored him with the Melville L. Wolfrom Award. A special issue of the journal Carbohydrate Research was dedicated to Dr. Tipson on the occasion of his retirement as an editor of that journal.

Tipson’s remarkable capacity for work continued undiminished until very late in his life, even though he seldom ventured outside his home after the death of his wife. Physical impairment during his last two years finally curtailed the extent of the editorial activities from his celebrated mailbox, and he died in his 85th year after a short illness. He is survived by his sisters Joyce Kenward of Leamington Spa, England, and Jean McIlroy of Sedona, Arizona, as well as six nephews and four nieces.

Although memories of Bob Tipson will inevitably center on his insistence on perfection in the preparation of scientific manuscripts, the fundamental research contributions he made early in his long career to our knowledge of the structure of carbohydrates and especially of the nucleic acids merit equal recognition. His brusque and sometimes irascible outward manner con- cealed a kindly and concerned individual, a true gentleman and scholar.

DEREK HORTON

The author thanks the following persons for valuable discussions and information during the preparation of this article: J. S. Coombes, Joyce Kenward, Jean McIlroy, H. S. El Khadem, R. Schaffer, M. Stacey, and J. M. Webber.

Page 23: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

This Page Intentionally Left Blank

Page 24: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

ADVANCES IN CARBOHYDRATE CHEMISTRY AND BIOCHEMISTRY, VOL. 50

HOW EMIL FISCHER WAS LED TO THE LOCK AND KEY CONCEPT FOR ENZYME SPECIFICITY’

BY RAYMOND U. LEMIEUX AND ULRIKE SPOHR

Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada

I. INTRODUCTION

Emil Fischer’s genius was in the identification of important areas for research in the field of organic chemistry, which, as the name implies, was concerned with compounds derived from living organisms. Once the project was identified and engaged, he brought unsurpassed creativity for successful experimental involvement and logical interpretation of the results. He seemed to have had a global view of natural science, and his driving interest was to contribute to an understanding of the chemical processes of living organisms. He is quoted ( 1) as stating, in a letter to his mentor, Adolf Baeyer, that he wished to synthesize the first “artificial ferment” (enzyme activity) and that, with the achievement of this goal, he would consider his mission in life accomplished. Later, in the course of his research on polypeptides, he realized that he would not reach this goal. He, of course, could have no idea of what the synthesis of an enzyme would involve.

Our assessment of the literature suggests that Fischer’s motivation to enter the field of carbohydrate chemistry was the realization that knowledge of the relative configurations of the asymmetric carbon atoms of the sugars was an essential stepping stone for research in the biological systems, that area of inquiry central to his interest in a scientific career. His brilliant success in meeting this challenge has rightly earned him the title of father of carbohy- drate chemistry. This chapter demonstrates that a strong claim can be made that he is also a leading pioneer of biological chemistry.

Hudson (2) published a scholarly review of the monumental contribution

I Presented at the symposium “Emil Fischer: 100 Years of Carbohydrate Chemistry,” 203rd National Meeting of the American Chemical Society, Division of Carbohydrate Chemistry, San Francisco, California, April 5 - 10, 1992.

Copyright 0 1994 by Academic Press, Inc. Au nghts of reproduction in any form reserved. 1

Page 25: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

2 RAYMOND U. LEMIEUX AND ULRIKE SPOHR

of the relative configurations of the sugars- the feat for which Emil Fischer is best known. Indeed, this stellar accomplishment appears to have over- shadowed other major contributions, particularly the first insights into en- zyme specificity. We now attempt to present, with fidelity, how Emil Fischer was led to his “lock and key” concept for enzyme specificity. The objective is to produce a more widespread appreciation of the profound significance of this landmark contribution (3), which arose from his recognition of asym- metric induction. Although not always literal, our translations into English are expected not to be misleading. The nomenclature is retained essentially as used by Fischer.

Emil Fischer’s proof of the relative configurations of the four chiral centers in open-chain glucose (4) appeared in 189 1. The following three years saw a brilliant series of follow-up papers, based on the knowledge that certain sugars differ only in configuration. These publications mark the origin of our appreciation that molecular forces provide the stereochemical guidance necessary to living processes. Especially because of the present worldwide concern with molecular recognition, it seemed most appropriate to survey these classical contributions as a centenary commemoration to the first person in the field. One of his many students and a life-long friend, Karl Freudenberg, remarked in his excellent biography (5) of Emil Fischer:

Theoreticalquestions playeda minor role in his thoughts. . . . Emil Fischer was theclever tactician who proceeded on a broadfront. . . . There were many who were better read than he, but no-one who had more practical experience.

11. ASYMMETRIC INDUCTION Whereas the configurations of the sugars are truly a lasting monument to

Emil Fischer, the concept of asymmetric induction, also referred to as partial asymmetric synthesis, initiated a new era in chemical research that is still with us today.

The modem concept of asymmetric induction is illustrated by the formu- las in Fig. 1. As shown, the addition of hydrogen cyanide to the optically active aldehyde can lead to two diastereomers (1 and 2). If the process is under thermodynamic control, the formation of the more stable isomer will be favored; that is, that isomer for which the non-bonded interactions be- tween the newly formed cyano and the hydroxyl groups with the dissymme- tric R* group are weakest. On the other hand, the difference in the yields of 1 and 2 can be the result of kinetic control arising from a difference in the energies of the transition states- that state with the lower energy will form faster and lead to the product of higher yield. It is noteworthy that the tenets

Page 26: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

LOCK AND KEY CONCEF’T FOR ENZYME SPECIFICITY 3

CN CN

HOCH I I

I R’

I R’

Ill Ill

R’CHO + HCN - HCOH +

R R

1 2

E = Nonbonded interaction energy.

When R* is dissymmetric, El + E‘1 # E2 + E’2

:. 111 # 121

FIG. 1 . -Asymmetric induction-under thermodynamic control, 1 will form in higher yield if E, + E’, < E, + El2.

of conformational analysis first provided the theoretical base for the formu- lation in 1952 of an appreciation for the steric control of asymmetric induc- tion, which has become known as Cram’s rule (6).

Although organic chemistry was still at a primitive state of development, a strong foundation for Emil Fischer’s work had been laid by such great chem- ists as Berzelius, Wohler, Liebig, Baeyer, and Kiliani. Furthermore, Louis Pasteur had reported (7), as outlined in Fig. 2, the preferential metabolism of the dextro-enantiomer of tartaric acid about 15 years prior to Fischer’s doctoral studies with Adolf Baeyer at the University of Strasbourg. An un- derstanding of optical isomerism had been provided in 1874 by the van’t Hoff-Le Be1 theory of the asymmetric carbon atom (8) (see Fig. 3) and ball and stick three-dimensional molecular models were in use much as they are

COOH COOH COOH I I I

H-C-OH HO-C-H Penicilliwn HO-C-H I + I - I + Fermentation products

HO-C-H H-C-OH glaucwn H-C-OH I I I COOH COOH COOH Natural --------

dl-Tartaric acid I-Tartaric acid FIG. 2.-Louis Pasteur’s preparation of D-(kvo)-tartaric acid.

Page 27: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

4 RAYMOND U. LEMIEUX AND ULRIKE SPOHR

Planeof~ymmeby , ~ D i y m e h i c , O H COOH COOH COOH

. - . # 0 # *

. . 0 . H-C-OH - HO-C-H H-C-OH HO-C-H

H-C-OH HO-C-H HO-C-H H -C -OH

COOH COOH . 0 0

Meso Racemate

FIG. 3.-The basic assumption for Fischer’s research of the optical isomerism of sugars.

today. As Emil Fischer stated (4):

Allprevious observations in thesugar group are in such complete agreement with the van’t Hof - Le Be1 theory of the asymmetric carbon that the use of this theory seems justifiable.

The concept of the existence ofasymmetric forces in nature was not new to Fischer. Indeed, Louis Pasteur (9) was generalizing about asymmetry in 1874 when he prophesied:

I am convinced that life as we know it has arisen out of asymmetrical processes in the universe. The universe is asymmetric.

He was convinced that optical activity is a peculiarity of life and, therefore, his view was directed toward asymmetry at a cellular level rather than at the molecular level developed by Emil Fischer.

Thus, the stage was well set in 1882 when Emil Fischer, at the age of 30 years, was appointed Professor and Director of the Chemical Institute at the University of Erlangen and thereby gained full independence in the direction of research. He chose to study the carbohydrates, and his first publication in the field appeared in 1884. The work was concerned with the reaction of sugars with phenylhydrazine, a compound that he had discovered as a teach- ing assistant about 10 years earlier while helping one of Baeyer’s students. At that time, his father, Laurenz Fischer, who was a very successful business- man, added to his portfolio a brewery in Dortmund. His son, Emil, who was already a chemist, became involved and it is recorded (1) that he had advised his father to purchase a Linde ice-making machine to cool and store the beer. Also, Emil developed an interest in mycology, the science of lower plants, and he recommended that the brewery acquire a microscope to differentiate yeast species and to detect contaminants. He had actually become highly knowledgeable about yeasts some 6 years earlier while he was studying in Adolf Baeyer’s laboratory and, in the course of a brief stay in Strasbourg in 1876, through a viniculturist named Dr. Fritz, he became intensively in- volved in the study of lower plants at the Strasbourg-Botanical Institute.

Page 28: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

LOCK AND KEY CONCEPT FOR ENZYME SPECIFICITY 5

Fortunately for chemistry, he did not stay in Strasbourg because he wrote in his memoirs (lo),

I certainly would have done my research in thisfield (mycology) had I stayed longer in Strasbourg.

He was then 24 years of age. Organic chemistry in the late 19th century was focused on the chemistry of

natural products. The main opportunities, in terms of the techniques of the time, were offered by the major components of living tissues, that is, the carbohydrates, nucleic acids, lipids, tannins, and proteins. The idea that organic compounds could be synthesized only within living organisms had long been dispelled. Nevertheless, as already pointed out, Louis Pasteur (9) had quite recently expressed the opinion that there existed forces that could be exerted only within living cells. This idea was strenuously opposed by Liebig ( 1 I), who held that fermentation and similar processes were due to the action of chemical substances. The Pasteur - Liebig controversy ended when Buchner ( 12) succeeded in extracting a cell-free fermentation system from yeast that fermented glucose.

Van’t Hoff had predicted, in 1874, that there should exist 16 normal straight-chain optically isomeric aldohexoses (8,13). It was the awesome challenge to substantiate this prediction that Fischer accepted in the mid- 1880s, and that was met by his classical contribution in 189 1 entitled, “On the Configuration of Glucose and Related Compounds” (4).

He represented glucose as shown in formula 3 and indicated that it should be interpreted as seen for 4. Thus, the hydrogen atoms and the hydroxyl groups at the four asymmetric carbons are considered to project above the plane of the paper. The representation was later simplified to that shown for

COH HC=O HC=O

H-C-OH H-C-OH HYOH . I .

H O ~ H . . I - HO-C-H

H-C-OH H-C-OH HCPH - - - - HGC-H

-

0

H-C-OH H-C-OH H C ~ H . . I CH20H CH20H CH20H

3 4 5

dextro-G lucose

5 and became known as a Fischer projection formula (14). Fischer realized that an arbitrary assignment of absolute configuration was necessary to an orderly development of organic chemistry. Fortunately, his assignment of 3 to dextro-glucose proved correct.

Page 29: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

6 RAYMOND U. LEMIEUX AND ULRIKE SPOHR

The preparation of a-hydroxy acids by way of the cyanohydrin was estab- lished by Winckler (15) in 1832 by the synthesis of mandelic acid from benzaldehyde. As Emil Fischer emphasized, it was Kiliani (16) who first applied the well-known cyanohydrin synthesis of a-hydroxy acids from aldehydes and ketones to the building up of aldonic acids from aldoses. The reduction of these acids to fatty acids then provided Kiliani’s classical proof of the structures of glucose and fructose (17). Although he had used sodium amalgam for the reduction of sugars to alditols, it was Fischer who learned to reduce the aldonolactones to aldoses. The overall process of building aldoses to higher sugars has become known as Fischer- Kiliani synthesis.

We have no evidence that Kiliani was aware of the formation of dias- tereoisomeric aldonic acids in the course of his cyanohydrin reaction. Fischer knew that the readily available I-arabinose had been subjected to the cyanohydrin reaction by Kiliani and found to provide a substance he called “arabinocarbonic acid.” Indeed, Fischer offered the opinion that the reac- tion of sugars with hydrogen cyanide discovered by Kiliani in 1885 was the most important reaction in carbohydrate chemistry (1 8). Fischer character- ized this product as “I-mannonic acid” and realized that, likely, the com- pound could be epimerized to 1-gluconic acid by heating with quinoline. However, he felt this procedure was so tedious and inefficient that the acid would not have been prepared in this way unless it had first become available by some other method in order that its physical properties be known. He reported ( 18) that, in fact, a good quantity of I-gluconic acid formed along with the I-mannonic acid on the addition of hydrogen cyanide to l-arabinose (see Fig. 4). He found this strange and commented as follows:

The simultaneous formation of the two stereoisomeric products on addition of hydrogen cyanide to aldehydes, which was observed here for thefirst time, is quite remarkable in theoretical as well as in practical terms.

Also, in 1890 Fischer had proven that the reduction of fructose with sodium amalgam yields a mixture of mannitol and sorbitol and pointed out that this conformed with the van’t Hoff-Le Be1 theory (19). It seems, therefore, that the idea of asymmetric induction was clearly in a state of incubation prior to his publication of the relative configurations of the sugars in 189 1.

Fischer’s involvement with the relative configurations of the sugars re- quired the preparation of pure substances and he gradually accumulated experimental data (20-22), which required the formation of epimeric al- donic acids in unequal amounts (see Table I). Thus, he was able to write (23) in 1892:

A second question of general importance relates to the quantities in which the two stereo- isomeric products are obtained on the generation of a new asymmetric carbon atom. Starting with nonracemic optically inactive starting materials, only racemic products are

Page 30: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

LOCK AND KEY CONCEF'T FOR ENZYME SPECIFICITY

H-C=O COOH

H-C-OH H-C-OH . .

HCN H 2 0 . HGC-H - NH3 + H-C-OH t . H a C - H HCC-H

CH20H H a C - H .

CH2OH

COOH . HGC-H

H-C-OH

HGC-H

HGC-H . CHZOH

I-Arabinose I-mannonic acid I-gluconic acid (Kiliani's arabinocarbonic acid)

FIG. 4. -The discovery of asymmetric induction. The yield of the I-mannonic acid, under the conditions then used, was about three times greater than that of the ghco isomer (see Table I ) .

formed and that means the two stereoisomers are formed in equal quantities. In the case of the present syntheses (Kiliani cyanohydrin syntheses), where the sugars used as starting materials are already asymmetric systems, this rule does not apply.

Two years later, in 1894, he wrote as follows (3): To my knowledge, by these observations strictly experimental proof has been provided for thejirst time that in the case of asymmetric systems the further synthesis occurs in an asymmetric sense.

111. YEAST FERMENTATIONS AND ENZYMES Obviously, Fischer had conceived of the phenomenon we now refer to as

asymmetric induction and had become deeply interested in its relevance to biological processes. It was that year that he abandoned Wurzburg Univer- sity to accept the chair of chemistry at the University of Berlin, which was regarded as the highest position in the realm that could be achieved by a professor of chemistry. He was promised a large new institute and it appears that the design and financing of this laboratory met with considerable con- troversy. It seems probable that Fischer took advantage of this discontinuity in his research to write up much of the work he had done in Wurzburg on the

TABLE I Some Early Observations of Asymmetric Induction by Fischer

Yield (46) Reference

I-Arabinose (50 g) - Mannonic acid 34 20

d-Mannose (2 kg) - a-Mannoheptonic acid 87 21 d-Xylose (40 g) - Gluconic acid lactone 51 22

Ca-gluconate 1 1

Idonic acid 35

Page 31: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

8 RAYMOND U. LEMIEUX AND ULRIKE SPOHR

fermentation of sugars and the interpretations of the results, in a series of monumental publications in 1894.

The use ofthe enzyme system then known as invertin, which was extracted from beer yeast with water and precipitated from the aqueous solution, was available to Fischer when he began his classical studies of the enzymic hydrolysis of glucosides reported in 1894. The stage was also set by another enzyme known as emulsin, which Fischer purchased from E. Merck, Darm- stadt, and which was known to hydrolyze several natural aromatic glucosides such as salicin, coniferin, arbutin, and the synthetic phenyl glucoside. These aryl glucosides were already known to not be cleaved by invertin.

The lock and key concept for enzyme specificity appears to have gelled in Fischer’s mind in the course of using yeasts in his studies on the configura- tions of sugars. It is noteworthy in this connection that it was not until 1878 that the term “enzyme” was introduced by Kuhne (24). Until then, the substances responsible for these biological activities were referred to as fer- ments. In fact, enzyme is a Greek term that means “in yeast.” It is pertinent to note with regard to enzyme action that Pasteur’s opinion, that the fer- mentation process could not be separated from the living cell, did not take into account the observation, made in 1833 by Payen and Persoz (25), that starch was converted into reducing sugars by a thermolabile substance present in the precipitate that formed on adding alcohol to a cell-free aqueous extract of malt. They termed the substance a “diastase” and the “-ase” ending of this term became in time used to designate the protein catalysts that we now call enzymes. In an Emil Fischer memorial lecture, Forster (26) reported that, as early as 1837, Berzelius held the opinion “that in living plants and animals there take place thousands of catalytic processes between tissues and fluids.” It took Fischer to appreciate the significance of the diastase activity.

Fermentations using ordinary beer yeasts had played key roles in the investigations on the configurations of glucose performed in Wiirzburg, which Fischer published in 1891 (4). For example, in 1889, Fischer and Hirschberger (27) reported the fermentation of d-mannose, a sugar that they had obtained by oxidation of mannitol with nitric acid and found identical to an aldohexose of widespread occurrence in plants (28). In the following year, he reported that the fermentation of racemic mannose left the l-isomer intact (29). Similar observations were made with regard to the nonfermenta- bility of l-(dextro)-fructose, I-glucose, and l-galactose (29) (see Table 11). In addition, both the optically active isomeric guloses and various heptoses and octoses were found to resist fermentation. He saw these results as an essential extension of the older observation by Pasteur (7) that microorganisms alter only one of two enantiomers; that is, the fermentation of sugars depends on

Page 32: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

LOCK AND KEY CONCEPT FOR ENZYME SPECIFICITY 9

TABLE I1 Experiments That Demonstrated the Chemical Basis of Biology

Racemic mannose a I-mannose + CO, + ethanol

Similarly, d-glucose, I-fructose, or d-galactose

However, I-glucose, d-fructose, or I-galactose

CO, + ethanol

no fermentation

the total configuration and not only whether it is the dextro or lev0 form. He therefore concluded:

Thefermentabilityofhexoses is in close relationship to thegeometricshapeofthe molecule and can even be designated as a stereochemical question.

In a landmark paper (30) that he coauthored with Hans Thierfelder, a mycologist, the behavior of different sugars toward pure yeasts was de- scribed. In this connection, he realized that the yeasts he had used in his earlier investigations were mixtures and, therefore, the results could be mis- leading. For this reason, he turned his attention to the fermentation of sugars by 12 different pure yeast species. Furthermore, he realized that he was in a uniquely fortunate position to undertake these studies because the research on the relative configurations of the sugars had left him with a fine inventory of rare carbohydrate structures. It is interesting to note how the fermenta- tions were scaled down for the study of rare sugars as substrates (Fig. 5 ) . In this regard, he wrote:

Since the preparation of the artificial sugars is in part quite tedious and the experiments had to be modified repeatedly, we used a small fermentation tube, as shown below, in order to save material (30).

As seen in Table 111, all six yeasts rapidly fermented glucose, mannose, and fructose. However, three of the yeasts had difficulty in fermenting galactose. None could metabolize either the naturally occurring sugars I-arabinose and rhamnose or synthetic sugars including 1-glucose, sorbose, a-glucoheptose, and a-glucooctose. Thus, the data presented in Table I1 were confirmed with pure yeast cultures and Fischer proposed the generalization:

The same observation is likely to befound for other microorganisms as well as for other groups of organic compounds and perhaps a very great number of chemical processes occurring within an organism are influenced by the geometry of the cell.

IV. THE LOCK AND KEY CONCEPT

A number of glycosides were available to Fischer by way of the Koenigs- Knorr reaction and his own glycoside synthesis, which involves treatment of

Page 33: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

10 RAYMOND U. LEMIEUX AND ULRIKE SPOHR

Actual size FIG. 5.-Emil Fischer’s fermentations on a semi-micro scale using an apparatus of the size

shown. (a) -70 mg sugar, 0.35 ml water, 0.35 ml sterilized yeast extract, and 13 mg yeast species; (b) S-trap for evolved COz; (c) aqueous barium hydroxide.

the sugar in alcoholic hydrogen chloride solution. Consequently, the fer- mentation of a number of glycosides by different pure yeast species could be examined (30). The results, presented in Tables I11 and IV, showed that certain yeasts that avidly fermented glucose, fructose, and mannose only reluctantly fermented galactose and that a yeast that fermented sucrose and

TABLE 111 The Selective Fermentation of Natural Sugars by Pure Yeasts

sugar

Glucose Mannose Galactose

Yeast d 1 d I d I CFructosea d-Sorbose*

SpastorianusZ +++ - +++ - +++ - +++ - S. pastorianusZf +++ - +++ - ++ - +++ - S.pastorianusfff +++ - +++ - +++ - +++ - Brauereihefe +++ - +++ - +++ - +++ Brennereihefe +++ - +++ - + - +++ Milchzuckerhefe +++ - +++ - + - +++

- - -

a Used by Fischer to designate natural ~ h c t o s e . b Also negative were d-talose, I-gulose, I-arabinose, rhamnose, a-glucoheptose, and a-gIucooctose.

Page 34: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

LOCK AND KEY CONCEPT FOR ENZYME SPECIFICITY 11

TABLE IV The Fermentation of Glycosides by Different Pure Yeasts

Methyl Glucosyl Sucrose Maltose Lactose a-glucoside resorcinolb

~ ~

- + S. pastorianus Z +++ +++ -

+ Brauereihefe +++ +++ + Brennereihefe +++ +++ + S. productivus + +++

Milchzuckerhefe +++ - +++ -

- - - - - -

Not tested

a From Fischer synthesis. * From Koenigs-Knorr synthesis.

lactose did not ferment maltose. These observations led to his concern as to whether or not the different yeasts possessed different enzymes. Experiments were designed to answer this question, and it was soon established that, in fact, yeast contains at least two different enzymes. The procedure is outlined in Fig. 6, where it is seen that, whereas an extracellular enzyme of the Frohberg yeast could hydrolyze sucrose but not maltose, the cells contained an enzyme that ferments both the disaccharides. On this basis, it was con- cluded:

These present observations are undoubtedly in favor of the assumption that the yeast contains two direrent enzymes.

Of course, it is now established that the glycolysis of glucose to carbon dioxide and ethanol occurs by way of a complex pathway involving 10 different enzymes acting on a variety of sugar phosphate intermediates. The extracellular enzyme preparation that Fischer used was termed invertin, the origin of the term for the enzyme we now know as invertase. He termed the intracellular enzyme yeast-glucase (3 1) and this enzyme is of the type we now refer to as an amylase.

- Crushed Cells Ground Powdered glass

Frohberg Yeast

1 H20 1 H2O

Cell-free Extract I Cell-free Exiract I1

Invert sugar No hydrolysis Invert sugar Glucose

FIG. 6. -How the presence of different enzymes in a yeast cell was established.

Page 35: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

12 RAYMOND U. LEMIEUX AND ULRIKE SPOHR

Fischer then examined the lactose yeast in the same manner as he did the Frohberg yeast and found it to contain both an invertin-like enzyme and a lactose-cleaving enzyme, which he termed lactase. From these results he concluded that the first step in the fermentation of lactose, as for the fermen- tation of sucrose and maltose, is the hydrolysis of the disaccharide to mono- sacharide. From this observation, he drew the landmark conclusion that he considered it most unlikely that any polysaccharide (the term included di- saccharides) can be fermented without first being hydrolyzed to hexose (3 1).

The research with Thierfelder (30) had led to the hypothesis that the active chemical agents of yeast cells can react only with those sugars that are configurationally related. It was this stereochemical assessment of the fer- mentation process that, in turn, now led to the question (32):

Would similar diferences be found for the ferments that could be separated from the organism and termed “enzymes”?

To answer this question he turned to a study of the properties of two glucosi- dases, then known as invertin and emulsin (32). The substrates were to be the large number of artificial glucosides that he had synthesized from different sugars and alcohols. The results ofthese studies are presented in Table V. It is

TABLE V Effects of Structure and Configuration on

Enzymatic Hydrolysis

Crude enzyme preparation

Glycoside Invertin” Emulsinb

a-Glucosides Methyl Ethyl Sucrose Maltose

&Glucosides Methyl Phenyl Salicyl

Methyl

Methyl Lactose

a-Galactosidec

8-Galactosides

+ + + +

+ + +

+ +

Aqueous extract of air-dried beer yeast. Product of E. Merck, Darmstadt. Also, neither of the enzyme preparations hydrolyzed a-

glycosides of rhamnose or arabinose.

Page 36: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

LOCK AND KEY CONCEPT FOR ENZYME SPECIFICITY 13

to be noted that Fischer did not know the configurations of the anomeric carbon of glycosides. Furthermore, he presented these compounds as furan- oside structures in accordance with the prevailing notions on sugar struc- tures.

Fischer was intrigued by the fact that emulsin caused hydrolysis of both /--glucosides and /--galactosides but had no effect on either the a- or /--xylo- sides (33). Since, at the time, Fischer expected glycosides to be furanosides, he suggested that both the enzymes required the presence of a free hydroxyl group at position 5 of a hexoside.

At this point Fischer concluded that the enzymes, in terms of the configu- rations of the substrates, are as fastidious as yeast and other organisms. He then returned to the above-mentioned hypothesis that he and Thierfelder had proposed (30) and concluded (32) that the protein substances known as invertin and emulsin, like the substrates whose hydrolyses they effected, were asymmetrically formed molecules. On the basis of this consideration, he came to the momentous lock and key concept for enzyme activity and commented as follows:

The restricted effects of the enzymes may therefore be explained by the assumption that the approach of the molecules that cause the chemical process can occur only in the case of a similar geometric shape.

To use a picture, Z would like to say that enzyme andglucoside have tofit to each other like a lock and key in order to exert a chemical effect on each other.

v. INSIGHTS ON ENZYME SPECIFICITY

Emil Fischer developed a strong interest in the structural requirements for enzyme activity as the result of effects of changes in the structures of the a- and /--methyl glucosides on their properties as substrates for the enzymes invertin and emulsin, which, as we have seen, he had shown to be a- and /--glucosidases, respectively. As already mentioned, he was fascinated in 1895 by the fact that emulsin had no effect on either the a- or /--methyl xylosides (33). In a 1912 publication with Karl Zach (34), he reported that /--methyl6-deoxyglucoside was hydrolyzed by emulsin and wrote:

It appears to us very strange that the effect of the enzyme on the methoxylgroup at the other end of the carbon chain depends on the sixth carbon atom.

The following question, which appears to be the origin of the use of chemical synthesis to provide probes for the assessment of the structural requirements for complex formation, was asked:

How will the enzyme behave $there is a carbon richer alkyl at the end of the chain?

Soon after his death in 1919, his colleagues, as coauthors, reported the results presented in Table VI. Since methyl 2-deoxyglucoside had not been

Page 37: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

14 RAYMOND U. LEMIEUX AND ULRIKE SPOHR

TABLE VI The Probing by Fischer of the Active Site of an

Enzyme (Modem Formula)

Substituent

R R’ Hydrolysis by emulsin Reference

CH,OH OH + 32 35 33 H OH

36

CH,OH H -

CH, OH + 34 CH,Br OH -

-

hydrolyzed either by emulsin or by enzymes of yeast extract, it was con- cluded (35) that the presence of an hydroxyl group at the 2-position plays an essential role in the lock and key mechanism. Other collaborators reported (36) in 1920 that, although emulsin readily cleaved p-methyl6-deoxy-d-glu- coside (34), the enzyme had no effect on ~-methyl6-bromo-6-deoxy-d-glu- coside. Thus, it appears that among Emil Fischer’s last thoughts was the consideration of molecular recognition and how synthetic methodologies may provide the means to a precise understanding at the molecular level of his lock and key concept for enzyme activity. This idea was examined by many others, but a proper understanding had to await the development of synthetic methodologies for the synthesis of oligosaccharides and congeners thereof for the probing of protein combining sites.

As already stated, Fischer was deeply intrigued by the phenomenon of enzyme activity. He realized that the substances were proteins and this undoubtedly was why he next undertook the study of amino acids and peptides. He fully appreciated that the specificity of enzyme catalysis de- pended on the occurrence of a complementarity for interacting dissymmet- ric surfaces. In this regard, he wrote (3):

This example (the cyanohydrin reaction) appears to me to provide a simple solution for the natural asymmetric synthesis. The formation of the sugar, as the plant physiologists assume, occurs in the chlorophyll grain, which itserf is composed of optically active sub- stances. . . . Theprepared sugar is released and later on used by theplant, as is known, for thepreparation of other organic components. Their asymmetry is thus explainedfrom the nature of the building material. Of course, they also provide material for new chlorophyll

Page 38: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

LOCK AND KEY CONCEPT FOR ENZYME SPECIFICITY I5

grains, which again produce active sugars. In this way optical activity is passed on from molecule to molecule, such as Ige goes from cell to cell. Therefre it is not necessary to deduce the formation of optically active substances in the plant from asymmetric forces that reside outside the organisms, as Pasteur presumed.

In a biography prepared for the I966 issue of Advances in Carbohydrate Chemistry, Karl Freudenberg wrote (5):

Under his leadership, synthetic and theoretical chemistry was reunited with biochemistry, and a broad scientijc basis restored to organic chemistry.

Fischer never allowed synthesis to become an end in itself and thereby lose contact with general problems. In this regard, Freudenberg recorded ( 5 ) the following comment made by Fischer- probably incidentally -in 1904:

Only6 ofthe32 heptosesandonly2 ofthe128nonoseshavebeenprepared. But, sincethese compounds have not yet been found in nature and are, therefre, of only minor interest, their systematic elaboration may be Idt for a later period.

Freudenberg further commented (5): He never gave unbridled rein to his synthetic efforts, nor did hefall into the temptation of purposeless synthesis. He always remained a true scientist-a student of nature. . . . This great individual was a man ofinflexible veracity and simplicity. . . . Emil Fischers life was based on responsibility: a responsibilityfor the austerity andpurity of his work and its aims, responsibility for the university as an important organ of our cultural and eco- nomic Ige, and responsibility for each of his students.

Although few are endowed with comparable talent and energy, his career is surely a splendid example for all. Hopefully, like the passing of optical activity from molecule to molecule, this chapter will help induce some transfer of Emil Fischer’s way of thinking and actions to future generations of organic chemists.

VI. CONCLUDING REMARKS

It is appropriate to close this chapter with an illustration of how Emil Fischer’s lock and key concept has since been found to be relevant to enzyme specificity. Thus, it will be seen that, in fact, specific structural features of the substrate act somewhat like the wards of a key. That is, insertion of the substrate into the enzyme’s active site is strongly demanding in complemen- tarity as is inserting a key into the barrel of a lock. Formation of the complex brings the structural features of the substrate and the enzyme that are to interact into close proximity and proper orientation. Thus, the organization and thermal energy required to achieve the transition state are greatly di- minished and catalysis is effected. The hydrolysis of maltose and other a-linked glucosides by the commercial enzyme, which is most commonly

Page 39: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

16 RAYMOND U. LEMIEUX AND ULRIKE SPOHR

referred to as glucoamylase, but is also known as amyloglucosidase or simply AMG, serves well because not only is there much known about the lock and key characterization of the reaction pathway but also a consideration of this enzyme establishes a connection with Emil Fischer (37). It was in the course of an investigation of the effect of various enzyme preparations on cellobiose that the decision was made to examine an enzyme preparation derived from the fungus Aspergillus niger. It did not catalyze the hydrolysis of cellobiose, whereas emulsin did, and the conclusion was drawn that cellobiose must have a /%linkage.

As the name implies, the enzyme is an amylase. It is an em-hydrolase that releases glucose from the nonreducing ends of starch and dextrins. Along with alpha amylases, glucoamylases are fungal enzymes produced by a vari- ety of Aspergillus species that have found major industrial importance for the production of high-glucose syrups and related applications (38). Their use in the brewing of beer was likely the reason that Fischer examined the glucoamylase that is produced by the fungus A. niger.

The enzyme was examined by Fischer following earlier studies by Bour- quelot (39) in France. Bourquelot (39) reported that he had made known in 1883 that an extract from a culture ofA. niger hydrolyzed maltose. He now provided evidence that the solution also caused hydrolysis of trehalose. However, since this activity was lost on heating to 63 “C but that responsible for the hydrolysis of maltose was maintained up to 75 “C, he concluded that the fungus produced two different “ferments” -one a maltase, the other a trehalase.

Pazur and Ando (40) separated the glucoamylase from other carbohy- drases of A . niger by ion-exchange chromatography. They later found that the enzyme also hydrolyzes isomaltose but at a much lower rate than maltose (4 I). It was reasonable, therefore, to test whether or not the enzyme would hydrolyze a (1 + 3)-linked disaccharide of glucose. Consequently, nigerose was examined and found to hydrolyze at a rate intermediate to those of maltose and isomaltose. Thus, it became apparent that, although the enzyme has a high specificity for an a-D-glucopyranosyl disaccharide, the structure of the aglycon can be varied without total loss of activity. On the other hand, the enzyme was found to be quite ineffective for the hydrolysis of either methyl or phenyl a-D-glucopyranoside (42). The profound difference in rate was attributed to a difference in the conformations of the glucosyl units of maltose and methyl a-D-glucopyranoside. This postulation was made be- cause it had been suggested (43) that the glucosyl unit of maltose was in a boat conformation. Because methyl a-D-glucopyranoside was believed to favor a chair form, it was considered that the difference in rate of hydrolysis was related to the ease with which the glucosyl units in the compounds can transform into the conformation preferred by the enzyme (42). However,

Page 40: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

LOCK AND KEY CONCEPT FOR ENZYME SPECIFICITY 17

with the advent of high-resolution nuclear magnetic resonance it became evident (44) that the glucosyl units of maltose are held extensively in the same ‘C,(D) chair conformation as methyl a-D-glucopyranoside and, there- fore, another explanation had to be sought. The answer was provided by Bock and Pedersen (49, who studied the effect of deoxygenation at the various hydroxyl-group positions on rate of hydrolysis.

Lemieux (46) recently reviewed the studies with his coworkers on the origin of the specificity in the recognition of oligosaccharides by proteins. In the course of these studies, it became evident that hydroxyl groups are invariably involved in providing the stereochemical complementarity re- quired for the binding of an oligosaccharide by monoclonal antibodies and lectins. However, binding studies using monodeoxy derivatives revealed that only some of these hydroxyl groups establish essential polar interactions with the protein. These were termed the key hydroxyl groups. On applying this technique to the hydrolysis of maltose by the amyloglucosidase ofA. niger, Bock and Pedersen found that hydroxyl groups on both of the glucose units were essential for efficient catalysis. The key hydroxyl groups proved to be OH-3 of the reducing glucose unit and OH-4‘ and OH-6’ of the nonreducing glucose unit of maltose.

The two key hydroxyl groups ofthe nonreducing unit must establish polar interactions that tend to anchor this glucose unit in the enzyme’s active site. That methyl a-D-glucopyranoside is not a good substrate was no longer surprising because this compound cannot provide the key hydroxyl group of the aglycon. Since maltose, isomaltose, and nigerose are all substrates, it became apparent that the catalysis could entertain structural variations in the aglycon as long as it can project an hydroxyl group toward the active site in a manner similar to OH-3 of maltose. Indeed, as may be seen in Table VII, conformational analysis of these disaccharides indicates that in each case a similar disposition of the three key hydroxyl groups can be achieved with relative ease. In a sense, these hydroxyl groups perform as the wards of a key and thereby provide a fine illustration of Emil Fischer’s lock and key concept for enzyme specificity.

The turning of the key once the complex has formed is a separate issue. In this regard, Lemieux (47) has pointed out that rotation about the glycosidic bond must weaken the exo-anomeric effect and thereby importantly activate the anomeric carbon to nucleophilic attack. Therefore, it seems likely that the role of the key hydroxyl group of the aglycon is to accommodate the rotation prior to the attack by water to formp-D-glucopyranose, which is the first product of the reaction.

It appears that the overall mechanism for the hydrolysis of maltose by glucoamylase will soon be delineated. A brief summary of how this is being accomplished deserves comment.

Page 41: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

18 RAYMOND U. LEMIEUX AND ULRIKE SPOHR

TABLE VII The Three Key Hydroxyl Group# (Bold Letters) Necessary for Efficient Hydrolysis of an

a-D-Glucopyranoside by the Glucoamylase of AsprgZus niger

Structures

HO

Maltose4’ O* OH OH

Ho+

Nigerose4’ 0-g

Ho+

Ho+

Ho% HO

HO

OH o=oH HO OH

OH 0-

OH a,$-TrehaloseM

no

OH p - K o l i b i o ~ s ~ ~

Ho+&$-!$-F HO 0

HO Isornaliose4’

a Lemieux and Bock (44) have pointed out why the two-dimensional structural formulas used to represent the various disaccharides provide a useful approximation of the conformational preference.

Svensson and coworkers (48) were able to separate commercial A. niger glucoamylase into two catalytic components, which were termed G1 and G2. It is now established (49) that G2 is a proteolytic fragment derived from GI. Meagher and Reilly (50) showed the two forms to behave similarly and this finding appears common to all major variants of amyloglucosidase that contain the catalytic domain. The amino acid sequences of GI and G2 had been established by Boel et al. (5 1). Using glucoamylase-specific synthetic oligonucleotides and molecular cloning of the complementary DNA syn- thesized from A. niger, the primary structure of the mRNA for the GI

Page 42: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

LOCK AND KEY CONCEPT FOR ENZYME SPECIFICITY 19

enzyme was established. In vitro translations of the mRNA followed by immunoprecipitations with glucoamylase-specific antisera showed that both GI and G2 were in the culture medium. Thus, a goal contemplated by Emil Fischer, that is, the synthesis of an enzyme that he had examined (37), w as accomplished.

Aleshin and coworkers (49) have reported the X-ray crystal structure at 2.2-A resolution of a G2-type variant produced by Aspergillus awamori. Meanwhile, an attempt was made to determine the amino acid residues that participate in the substrate binding and catalysis provided by G2 of A. niger (52). The results of the chemical approach indicated that the Asp-176, Glu- 179, and Glu-180 form an acidic cluster crucial to the functioning of the enzyme. This conclusion was then tested by site-specific mutagenesis of these amino acid residues, which were replaced, one at a time, with Asn, Gln, and Gln, respectively (53). The substitution at Glu-179 provided an inactive protein. The other two substitutions affected the kinetic parameters but were not of crucial importance to the maintenance of activity. The crystal struc- ture (49) supports the conclusion that Glu- 179 functions as the catalytic acid but Asp- 176 does not appear to be a good candidate for provision of catalytic base. Thus, there still exists considerable uncertainty as to how the disaccha- ride is accepted into the combining site for hydrolysis. Nevertheless, the kind of scheme presented by Svensson and coworkers (52) almost surely prevails.

As already mentioned, the glucoamylase project was chosen to illustrate Emil Fischer’s lock and key concept for enzyme specificity. It is seen that his vision has become unequivocally established. Many other developments could have been chosen, as can be appreciated from recent reviews by Hehre (54) and by Svensson (55). Cornforth (56) provided a fine overview of asym- metry and enzyme action in his Nobel prize lecture. Noteworthy is the conclusion that “stereospecificity is something not just incidental, but es- sential to enzyme catalysis.’’ In other words, the key must fit the lock.

REFERENCES

1. K. Hoesch, Ber., 54 (1921) 3-480. 2. C. S. Hudson, J. Chem. Educ., 18 (1941) 353-357. 3. E. Fischer, Ber.. 27 (1894) 3189-3232. 4. E. Fischer, Ber., 24 (1891) 1836-1845,2683-2687. 5. K. Freudenberg, Adv. Carbohydr. Chem., 21 (1966) 2-38. 6. D. J. Cram, in J. L. Seeman (Ed.), From Design to Discovery, pp. 12-13. American

Chemical Society, Washington, D.C., 1990. 7. L. Pasteur, Cornpt. Rend., 51 (1860) 298-299. 8. F. G. Riddell and M. J. T. Robinson, Tetrahedron, 30 (1974) 2001 -2007. 9. (a) Renk Dubos, Pasteur andModern Science, p. 28. Springer-Verlag, New York, 1988; (b)

10. E. Fischer, Untersuchungen iiber Kohlenhydrate und Fermente (1884- 1908). J. Springer, J. W. Cornforth, Interdisciplinary Sci. Rev., 9 (1984) 107- 112.

Berlin, 1909.

Page 43: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

20 RAYMOND U. LEMIEUX AND ULRIKE SPOHR

1 1. J. Liebig, Ann. Chem. Phurm., 153 (1870) 1-47. 12. E. Buchner and R. Rapp, Ber., 30 (1897) 2668-2678. 13. G. M. Richardson, The Foundations of Stereo Chemistry, Scientific Memoirs Series.

14. C. S. Hudson, Adv. Curbohydr. Chem., 3 (1948) 1-21. 15. F. W. Winckler, Ann. Phurm., 4 (1832) 242-247. 16. H. Kiliani, Ber., 19 (1886) 767-772. 17. H. Kiliani, Ber., 22 (1889) 521-524. 18. E. Fischer, Ber., 23 (1890) 2114-2141. 19. E. Fischer, Ber., 23 (1890) 3684-3687. 20. E. Fischer, Ber., 23 (1890) 261 1-2624. 21. E. Fischer and F. Passmore, Ber., 23 (1890) 2226-2239. 22. E. Fischer and R. Stahel, Ber., 24 (1891) 528-539. 23. E. Fischer, Ann. Chem., 270 (1892) 64- 107. 24. W. Kiihne, Unters. Physiol. Instifut Univ. Heidelberg, 1 (1878) 291. 25. A. Payen and J. F. Persoz, Ann. Chim. (Phys.), 53 (1833) 73-92. 26. M. 0. Forster, J. Chem. Soc., 1 17 (1920) 1 157- 1201. 27. E. Fischer and J. Hirschberger, Ber., 22 (1889) 3218-3224. 28. E. Fischer and J. Hirschberger, Ber., 22 (1889) 365-376. 29. E. Fischer, Ber., 23 (1890) 370-394. 30. E. Fischer and H. Thierfelder, Ber., 27 (1894) 2031 -2037. 31. E. Fischer, Ber., 27 (1894) 3479-3483. 32. E. Fischer, Ber., 27 (1894) 2895-2993. 33. E. Fischer, Ber., 28 (1895) 1429- 1438. 34. E. Fischer and K. Zach, Ber., 45 (1912) 3761-3773. 35. E. Fischer, M. Bergmann, and H. Schotte, Ber., 53 (1920), 509-547. 36. E. Fischer, B. Helferich, and P. Ostmann, Ber., 53 (1920) 873-886. 37. E. Fischer and G. ZemplBn, Ann. Chem., 365 (1909) 1-6. 38. B. C. Saha and J. G. Zeikus, Starch/Starke, 41 (1989) 57-64. 39. E. Bourquelot, Compt. Rend. 116 (1893) 826-828. 40. J. H. Pazur and T. Ando, J. Biol. Chem., 234 (1959) 1966- 1970. 41. J. H. Pazur and T. Ando, Arch. Biochem. Biophys., 93 (1961) 43-49. 42. J. H. Pazur and K. Kleppe, J. Biol. Chem., 237 (1962) 1002- 1006. 43. R. E. Reeves, J. Am. Chem. SOC., 76 (1954) 4595-4598. 44. R. U. Lemieux and K. Bock, Arch. Biochem. Biophys., 22 1 (1983) 125 - 134. 45. K. Bock and H. Pedersen, Actu Chem. Scund., B41 (1987) 617-628. 46. R. U. Lemieux, Chem. SOC. Rev., 18 (1989) 347-374. 47. R. U. Lemieux, in J. I. Seeman (Ed.), Explorations with Sugars: How Sweet It Was, p. 100.

American Chemical Society, Washington, D.C., 1990. 48. B. Svensson, T. G. Pedersen, I. Svendsen, T. Sakai, and M. Ottesen, Carlsberg Res. Com-

mun., 47 (1982) 55-69. 49. A. Aleshin, A. Golubev, L. M. Firsov, and R. B. Honzatko, J. Biol. Chem., 267 (1992)

50. M. M. Meagher and P. J. Reilly, Biofechnol. Bioeng., 34 (1989) 689-693. 51. E. Boel, I. Hjort, B. Svensson, F. Norris, K. E. Norris, and N. P. Fiil, EMBO J., 3 (1984)

52. B. Svensson, A. J. Clarke, I. Svendsen, and H. Mraller, Eur. J. Biochem., 188 ( 1990) 29 - 38. 53. M. R. Sierks, C. Ford, P. J. Reilly and B. Svensson, Protein Eng., 3 (1990) 193- 198. 54. E. J. Hehre, Denpun Kagaku, 36 (1989) 197-205. 55. B. Svensson, Denpun Kugaku, 38 (1991) 125-135. 56. J. W. Cornforth, Science, 193 ( 1976) 121 - 125.

American Book Co., New York, 1901.

19291 - 19298.

1097- 1102.

Page 44: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

ADVANCES IN CARBOHYDRATE CHEMISTRY AND BIOCHEMISTRY, VOL. 50

ANOMERIC-OXYGEN ACTIVATION FOR GLYCOSIDE SYNTHESIS: THE TRICHLOROACETIMIDATE METHOD

BY RICHARD R. SCHMIDT*

*Fakultat fur Chemie, Universtitat Konstanz, 0-7750 Konstanz, Germany

WILLY KINZY~

t Zentrale Forschungslaboratorien, CIBA - GEIGY AG. CH-4002 Basel, Switzerland

I. GENERAL INTRODUCTION TO GLYCOSIDE SYNTHESIS: ACTIVATION THROUGH ANOMERIC OXYGEN-EXCHANGE REACTIONS

The biological significance of glycoconjugates has stimulated much syn- thetic activity in glycoside synthesis in the past years (1 -7). These efforts were initially concentrated mainly on improvements of the well-known Koenigs-Knorr method (8), introduced in 1901, which requires an ex- change of the anomeric hydroxylgroup by bromine or chlorineas the first step (generation o f the glycosyl-group donor). The second step involves glycosyl- group transfer to the glycosyl acceptor in the presence of a heavy metal ion promoter (Scheme 1, path B). Although this is the basis of a very valuable methodology that has been reviewed extensively (4,5,7), several inherent disadvantages make the Koenigs - Knorr method often experimentally de- manding and certainly not very suitable for large-scale preparations. For instance, the requirement of at least equimolar amounts of the heavy metal salt promoter, often incorrectly termed “catalyst,” is a limiting factor (1 - 3). Therefore, alternative methods are o f interest.

Other anomeric-oxygen exchange reactions have been recently investi- gated quite extensively. Closely related to the Koenigs- Knorr method is the introduction offluorine as the leaving group (Scheme 1, path B) (6,9 - 13). Because of the difference in halophilicity of this element as compared with bromine and chlorine, additional promoter systems besides silver salts were found useful as activators for glycosylation reactions (1 4 - 16). However,

Copyright 0 1994 by Academic preSr Inc. AU rights of reproduction in any form reserved. 21

Page 45: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

22 RICHARD R. SCHMIDT AND WILLY KINZY

Activation through Retention of the

Glycosyl Donors via Anomeric-Oxygen Exchange Reactions / \e Anomeric Oxygen

H

Hal-

*Ha, \H

@ Fischer-Helferich (Acid Activation)

@ Koenigs-Knorr; Br, CI, (I) Act.

@ Anomeric 0-Alkylation: Base Activation

@ Trichloroacetirnidate (Imidate Act.) F-Activation S02R - Act.

PO(OR)2-Act. RS-Activation

SCHEME 1 . -Synthesis of Glycosides and Saccharides.

because of the generally lower donor properties of glycosyl fluorides (1 7) these intermediates have not yet gained wide application in the synthesis of complex glycoconjugates.

Thioglycosides, where the anomeric oxygen atom is replaced by an alkyl or arylthio group, have recently attracted considerable attention as glycosyl donors (Scheme 1, path B) (5,18,19). They offer sufficient temporary protec- tion of the anomeric center and present several alternative possibilities for regioselective activation to generate glycosyl donor properties. Earlier meth- ods for activation include mainly mercury(II), copper(II), and lead(I1) salts (20 - 28). However, besides the requirement of generally more than equimo- lar amounts of heavy metal salts, relatively low glycosyl-donor properties were experienced with these systems. This problem was partly overcome by the use of heterocyclic thioglycosides (2 1,23,25 - 27). In addition to metal salts, bromonium and chloronium ions are also highly thiophilic and thus provide with counter-ions of bromide and chloride, respectively, the corre- sponding glycosyl halides for a subsequent Koenigs-Knorr type of reaction ( 18,19,29). If the counter-ion of the halonium ion is a poor nucleophile (for instance, succinimide from N-bromosuccinimide), then direct reaction with alcohols as competing nucleophiles is favored and thus leads to 0-glycosides. However, low a,/3 selectivities are frequently obtained for nonneighboring

Page 46: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

ANOMERIC-OXYGEN ACTIVATION FOR GLYCOSIDE SYNTHESIS 23

group-assisted reactions (23,30). Formation of sulfonium ions from thiogly- cosides by the action of methyl triflate was also successfully applied to 0-glycoside bond-formation (3 1 - 34). Disadvantages of this method include the low a,@ selectivity observed for nonparticipating 2-0-protective groups, the health hazard of methyl triflate, and the formation of methylation products in other side reactions. The recently introduced activator dimethyl(methy1thio)sulfonium triflate (DMTST) proved to be highly thio- philic and gives rise to faster glycosylation than does methyl triflate (35). However, again, with nonparticipating groups the a,@ selectivity is usually low (32). Radical activation of thioglycosides has also been recently re- ported, providing similar results in terms of yield and diastereoselectivity

The Fischer - Helferich method, as a direct anomeric-oxygen replacement reaction (Scheme 1, path A), has been very successfully applied for syntheses of simple alkyl glycosides. However, because of its reversibility, it has not gained general importance in the synthesis of complex oligosaccharides and glycoconjugates (1).

(36).

11. ANOMERIC-OXYGEN ACTIVATION: ANOMERIC 0-ALKYLATION

1. Introduction The requirements for glycoside syntheses, high chemical and stereochem-

ical yield, and applicability to large-scale preparations were not effectively met by any of the methods just described. However, it seems that the general strategy for glycoside synthesis is reasonable:

(i) The first step should consist of a sterically uniform activation of the anomeric center with formation of a stable glycosyl donor having either the a or the p configuration;

(ii) The second step should consist of a catalyzed, sterically uniform, irreversible glycosyl transfer to the acceptor, proceeding with either retention or inversion of configuration at the anomeric center in high chemical yield and without affecting other bonds.

Only simple means meeting these requirements will lead to a generally acceptable and useful methodology. Therefore, besides acid activation (Scheme 1, paths A and B), the simplest form of activation would be base activation generating first an anomeric alkoxide structure of a pyranose or furanose (Scheme 1, paths C and D). This approach is especially tempting because Nature has a similar approach for generating glycosyl donors, namely glycosyl phosphate formation [see Section IV.2 and Ref. (17)J.

Page 47: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

24 RICHARD R. SCHMIDT AND WILLY KINZY

2. Anomeric 0-Alkylation

The direct 0-alkylation of the anomeric center (Scheme 1, path C) by treatment of furanoses and pyranoses with base and then with simple alky- lating agents, for instance an excess of methyl iodide or dimethyl sulfate, has long been known (1,3). Surprisingly, no studies employing this simple method for syntheses of more-complex glycosides and saccharides have been reported prior to our work (1,37,38).

In the beginning, direct anomeric 0-alkylation seemed very unlikely to fulfill all of the requirements for glycoside and saccharide synthesis. Even when all remaining functional groups (generally hydroxyl groups) are blocked by protecting groups, the ring- chain tautomerism between the anomeric forms and the open-chain form (Scheme 2) already gives three

R O a 0- a R o e H ,

R O R O R O 7

R O O OR

I I 1

R O

X = mainly OTf

Fl-X I

SCHEME 2. - 1-0-Alkylation and 1 -0-Acylation (Irreversible Reactions).

possible sites for attack of the alkylating agent. In addition, base-catalyzed elimination in the open-chain form of the sugar could be a destructive side-reaction. Therefore, the yield, the regioselectivity, and the stereoselec- tivity of such direct anomeric 0-alkylation would not generally be expected to be outstanding. In any event, the process should be governed at least by the following factors:

(i) the stability of the deprotonated species; (ii) the ring-chain tautomeric equilibrium and its dynamics; and (iii) the relative reactivities (nucleophilicities) of the three O-deproton-

ated species.

Page 48: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

ANOMERIC-OXYGEN ACTIVATION FOR GLYCOSIDE SYNTHESIS 25

Because of the irreversibility of the 0-alkylation reaction, kinetic regio- and stereo-control is required for selective product-formation. Therefore, selective formation of either a or p product seemed to be unattainable.

The first experiments with iodide derivatives of carbohydrates revealed that better alkylating agents are required (37). However, excellent reactivity with corresponding trifluoromethanesulfonates (triflates) was observed, providing, for instance, with 2,3-0-isopropylidene-~-ribose and derivatives, depending on the reaction conditions, very high yields of either a- and P-linked disaccharides (37). Surprisingly, even partial 0-protection or, as recently discovered, 0-nonprotection was compatible with this reaction (39 -44). The stereocontrol could be effected by intramolecular metal-ion complexation, by steric effects, and by taking advantage of the increased nucleophilicity of the equatorial anomeric oxide over the axial anomeric oxide [kinetic anomeric effect (45,46)]. This method could even be em- ployed in selective formation of a-glycosides of Kdo (47,48). Thus, the direct anomeric 0-alkylation constitutes an especially simple procedure for glyco- side and saccharide synthesis, giving generally high yields and diastereose- lectivities. The limitation to primary triflates was a major drawback for the general use of this anomeric 0-alkylation in glycoside synthesis. However, this problem was recently overcome, at least in part, by modifying the reac- tion conditions (49).

111. ANOMERIC-OXYGEN ACTIVATION: THE TRICHLOROACETIMIDATE METHOD

1. Formation of 0-Glycosyl Trichloroacetimidates

Aside from direct anomeric 0-alkylation (Scheme I , path C), base-cata- lyzed transformation of the anomeric oxygen atom into a good leaving- group (Scheme 1, path D) should be easily readily effected. Therefore, it is not surprising that several approaches have been directed toward this goal, as will be discussed later (Section IV). However, stable and concomitantly reactive intermediates were never obtained for the separate anomers. Obvi- ously, for achievement of stereocontrolled activation of the anomeric oxy- gen atom, the anomerization of the anomeric hydroxyl group or the ano- meric oxide ion, respectively, has to be considered (Scheme 2). Thus, in a reversible activation process and with the help of kinetic and thermody- namic reaction-control, possibly both activated anomers should be ac- cessible.

These considerations led us to the conclusion that suitable triple-bond systems A=B (or compounds containing cumulative double-bond systems A=B=C) might be found that add pyranoses and furanoses under base

Page 49: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

26 RICHARD R. SCHMIDT AND WILLY KINZY

catalysis directly and, because of reversibility, in a stereocontrolled manner (Scheme 1, path D) (1 - 3); thus, both activated anomers may be obtainable at will. However, the instability of open-chain aldehydic intermediates in basic media and the insufficient or undifferentiated reactivities of the a- and p-anomeric oxides lowered the expectations for stereocontrolled anomeric 0-activations along these lines.

The desired formation of stable anomeric 0-activated intermediates via base catalysis requires a different catalytic system for reactivity in the subse- quent glycosylation step. Therefore, after base-promoted trapping of ano- meric 0-activated intermediates (first step), mild acid treatment in the pres- ence of acceptors, leading to the formation ofglycosides (namely, acetals and derivatives) in an irreversible manner (second step), would constitute the simple means of catalysis desired for a new and efficient glycosylation method. These demands have to be considered in the selection of A=B (or A=-). Thus, the stable intermediates obtained in the first step have to exhibit by appropriate choice of the centers A and B (or A, B, and C) good glycosyl-donor properties in the presence of strictly catalytic amounts of acid. The water liberated upon glycoside formation is then transferred in two separate steps to the activating agent A=B (or A=B=C), thus providing the driving force for the glycosylation reaction (Scheme 3). This concept fulfills

Base (cat.) , Ro& RO OH + A G E , RO

OR OR

Acid (cat.) If HoR'

h \

-H20 \ \

\ \

RO RO a OR'

OR

SCHEME 3. -Steps in the Glycosylation Reaction.

the requirements just given for an efficient glycosylation methodology: truly catalytic amounts of a simple base (first step) and of a simple acid (second step) are required for anomeric 0-activation and promotion of the glycosy- lation, respectively; liberated water will not compete with the glycosyl ac- ceptor for the glycosyl donor because it is concomitantly chemically bound

Page 50: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

ANOMERIC-OXYGEN ACTIVATION FOR GLYCOSIDE SYNTHESIS 27

to the activating species A=B (or A=B=C); and thus, reversibility in the first and irreversibility in the second step provide important means for controlling the yield and stereochemistry of the anomeric 0-activated inter- mediate and of glycoside-bond formation. The trichloroacetimidate method developed by us, and recent contributions from other laboratories to this methodology, have proved the validity of this concept ( 1 - 3).

Electron-deficient nitriles, such as for instance trichloroacetonitrile and trifluoroacetonitrile (AEB: A = N; B = CCCl,, CCF,), are known to undergo direct and reversible, base-catalyzed addition of alcohols providing 0-alkyl trichloroacetimidates (1,50). This imidate synthesis has the advan- tage that the free imidates can be isolated as stable adducts, which are less sensitive to hydrolysis than their corresponding salts.

A detailed study of the addition of trichloroacetonitrile to 2,3,4,6-tetra-O- benzyl-D-glucose (la, Scheme 4) revealed (1 - 3,45) that, from the equatorial

l a : R = B n l b : R = A C

OH RO

CCI3CN. Base It RO

RO

RooYNH CC!,

1 a-a 1 b-a

~ ~ ~ o y c c 1 3 RO RO

NH

m.p. [OC] 1 a-p 1 b-P

1 b-a

SCHEME 4.-Addition of CCl, CN at the Anomeric Position.

1 -oxide ion, the P-trichloroacetimidate la$ is generated preferentially or even exclusively in a very rapid and reversible addition-reaction (Schemes 2 and 3). However, this product anomerizes in a slow, base-catalyzed reaction (via retroreaction, anomerization of the 1 -oxide ion, and renewed trichloro- acetonitrile addition) to the a-trichloroacetimidate la- having the elec- tron-withdrawing 1-substituent in an axial disposition, as favored by the

Page 51: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

28 RICHARD R. SCHMIDT AND WILLY KINZY

thermodynamically operating anomeric effect. Thus, with different bases [for instance K2C03, Cs2C03 and NaH or 1,8-diazabicyclo[5,4,0]undec-7- ene (DBU)] both 0-activated anomers may be isolated in pure form and in high yields via kinetic and thermodynamic reaction-control. Both anomers are commonly thermally stable and may be stored easily. A similar result was

TABLE I Synthesis of Trichloroacetimidates of DGlucose

Anomeric config. Yield

Compound" Reaction conditions (a$) (%) Ref.

CHzCl2, NaH, CCl-,CN, ...-&$ En0 room temp.

1a-b Bno-oycci, CH2C12, K2CO3, CC13CN, En0 room temp. BnO

NH

AcO CH,Cl,, K,CO,, CCl,CN, 48 h, room temp.

lb-a

/OAC O Y CCI, NH

1b-P A c O ~ o y C C l 3 CHZCl,, K,COp, CC13CN, AcO 2 h room temp.

NH OAC

/

CH,Cl,, NaH, CC13CN, room temp.

O Y N H CCl,

CH2Cl2, K2CO3, CCl,CN, 6 h, room temp.

/OPiV

CH,Cl,, NaH, CC13CN, 1.5 h, room temp.

i d s a PivO

~

1 :o 78 65,66

0: 1 90 46,67,68

l : o 98 66

0: I 78 46

l : o 90 58a,61

I : 3 74 58a

1 :o 60 69,70

CCI,

r? Bn, benzyl; Piv, pivaloyl; Ac, acetyl.

Page 52: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

ANOMERIC-OXYGEN ACTIVATION FOR GLYCOSIDE SYNTHESIS 29

obtained for the less reactive 0-acetyl derivative 1 b of D-glucose, providing trichloroacetimidates lb-a and lb-p, respectively (see Table I).

The higher nucleophilicity of the p-oxide ion may be attributed to a steric factor in combination with a kinetically effective stereoelectronic effect that results from repulsions of lone electron pairs, dipole effects, or both (Scheme 5 ) (45,46). This effect should be more pronounced in anomericp-oxide ions

(a) Dipole-Dipole Interaction

- 0

(b) Lone-Pair Orbital Interaction

SCHEME 5 .-Enhanced Nucleophilicity of poxides (Kinetic Anomeric Effect).

than in p-pyranosides because of the difference in the number of oxygen lone-pair orbitals and the difference in their relative energies. In addition, this kinetic anomeric efect should be particularly efficient in the p-manno- pyranosyl oxide ion, where the thermodynamic anomeric effect, favoring the a-anomer, is also stronger. This expectation could be experimentally confirmed in the irreversible anomeric 0-alkylation of mannopyranose, which leads in nonpolar solvents preferentially to p-glycoside formation (see references in Section 11.2). However, in the reversible trichloroacetimidate formation, the stronger thermodynamic anomeric effect results in much faster generation of the a-trichloroacetimidate, and therefore trapping of the p-species becomes much more difficult. Thus, a distinction between the thermodynamic and the kinetic anomeric effect could be experimentally verified.

Page 53: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

30 RICHARD R. SCHMIDT AND WILLY KINZY

The stereoselective anomeric 0-activation of carbohydrates and their de- rivatives via 0-glycosyl trichloroacetimidate formation is capable of exten- sion to all important hexopyranoses (Glc, Gal, Man, Fuc, Rha, Qui, GlcN, GalN), hexofuranoses, pentopyranoses, and pentofuranoses, as well as to glucuronic acid, galacturonic acid, and muramic acid; to 2-deoxy-urubino- hexose derivatives; and to many di-, tri-, and oligo-saccharides (see Section 111.3). It commonly provides stable compounds in a stereocontrolled man- ner. Thus, the requirements put forward for the first step, namely, efficient stereocontrolled formation of stable glycosyl donors, are fulfilled (see Sec- tion 11.1).

Ultimately the significance of the 0-glycosyl trichloroacetimidates must be based solely on their glycosylation potential under mild acidic catalysis. This potential has indeed been confirmed overwhelmingly in various labo- ratories and is presented in comprehensive detail in this article.

2. Reaction with Brsnsted Acids

The trichloroacetimidate method for glycoside synthesis extended its ver- satility right at the outset ( 5 1,52a) by exhibiting an especially smooth reac- tion of 0-( glycosy1)trichloroacetimidates with Brnnsted acids. Without the addition of any catalyst, simple Brnnsted acids are able to substitute the trichloroacetimidate group at room temperature in high yields, as shown (1 7) for la-a in Scheme 6. Because of anomerization of possible p products

x = CI (90 %) CHzCIz, RT

X = F (88 O h ) (HX : Py ' HF) X = Na (90%)

la -a + HX (excess)

SCHEME 6. -Substitution of the Trichloroacetimidate Group by Simple Brmsted Acids.

formed at the beginning of the reaction, only a products are finally isolated in these instances.

Carboxylic acids, being weaker acids, react with la-a with inversion of configuration at the anomeric center to yield B-0-acyl compounds (1,53). This mild and convenient method for 1-0-acylation of carbohydrates is also useful for pharmacological drug modification (54) or for the resolution of carboxylic acids (53).

Accordingly, phosphoric acid mono- and di-esters permit uncatalyzed glycosyl transfer from 0-( glycosy1)trichloroacetimidates (52a,5 5 - 57,58a,58b). The reaction is thus very useful in the synthesis of glycophos- pholipids (1,55), which are important constituents of cell membranes (1). Commonly, direct phosphorylation at the anomeric hydroxyl group leads to

Page 54: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

RO RO % ~ ~ ~ o y c c l ; RO

OR N H la-p Possible Transition States

for Diastereospecific Phosphate Formation 1a.a Ro O Y N H CCI,

'4

, ,

CH,CI,, RT, 1 h

( 95 % )

Acid-catalyzed a-Anomerization F

0

SCHEME 7. -Reaction of a- and &Trichloroacetimidates with Dibenzyl Hydrogenphosphate.

Page 55: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

32 RICHARD R. SCHMIDT AND WILLY KINZY

low a,p-selectivity. However, with the 0-(glycosyl)trichloroacetimidates, a high a or p selectivity is observed and anomerization proceeds only in the presence of strong acids. Therefore, the generation of cyclic transition-states was proposed (56) (Scheme 7), which results in an S N ~ type of configura- tional inversion. Calculations on the basic structures of the a- and p-tri- chloroacetimidates, respectively, exhibit ground-state con formational pref- erence for conformers that support the intermediate generation of these cyclic transition states. Presumably, the cyclic transition-state is not a planar ring of eight atoms, because the calculated dihedral angles of the ground states deviate considerably from such a possibility, but rather resembles a chairlike transition-state with two long bonds constituting the 0 . . C - - SOandN. . H - - -0connections.

Thus, all systems having related A=B-C-H geometry may react via the cyclic transition-state proposed for phosphoric acid derivatives and there- fore exhibit high diastereoselectivity. Accordingly, in addition to the carbox- ylic acids and the phosphoric acids already mentioned, phosphonic (59) and phosphinic acids (59), monoalkylsulfuric acid (56,60), and even a-pyridone (17,56) exhibit the same reaction behavior. However, the 2-pyridyl p- glycoside thus obtained from la-a is subsequently transformed via the same kind of pyridone attack into the corresponding 2-pyridyl a-glycoside (1 7,6 1). This finding may also explain anomerization to the thermodynami- cally more stable product in formation of glycosyl phosphates (56).

Further development of this idea led to the proposal (56) that reactive B=C groups, for instance carbonyl systems, would be able to activate alco- hol acceptors AH by generating a related A-B-C-H intermediate (Scheme 8, path I). It seemed that chloral might act as a catalyst along these lines. However, it turned out that the rate of decay in the transition state is too low in all systems tested thus far. Therefore, the carbonyl compound is more or less a substitute for a Lewis acid catalyst, as indicated in Scheme 8, path 11. The high reactivity and diastereoselectivity in chloral-catalyzed re- actions is attributable to the nitriles used as solvents in these reactions [see Section III.3.b and Ref. (62)].

3. Alcohols and Sugars as 0-Nucleophiles

a. Introduction. -The synthesis of oligosaccharides is characterized, be- cause of the various connections, anomeric configuration, and branching, by a much larger number of possibilities for coupling than that of other natural biopolymers, such as peptides or proteins, and ribo- or deoxy-ribonucleo- tides. Comparison of the number of possible isomers with those of the corresponding peptides and nucleotides impressively illustrates this point as indicated earlier (1). The wide structural variety renders sugars and, in par-

Page 56: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

K'

33

Page 57: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

34 RICHARD R. SCHMIDT AND WLLY KINZY

ticular oligosaccharides, ideal as carriers of biological information, encoding considerably more information per building block than proteins and nucleic acids.

This great structural variety, however, complicates the specific biosynthe- sis of complex oligosaccharides. In general, the formation of each saccharide linkage requires specific enzymes (“one linkage -more than one enzyme”); and thus, in comparison with the enzymic synthesis of proteins and nucleic acids, much more effort is needed.

The chemical synthesis of oligosaccharides is also more complicated than the synthesis of other biopolymers, because the construction of each indi- vidual oligosaccharide poses a new challenge, requiring a knowledge of methods, together with experience and experimental skill. Thus, there are no universal methods available neither for biological in vivo nor for chemical in vitro syntheses.

In synthesis of a disaccharide, two polyfunctional sugar components must be specifically linked. Therefore, the reactivity and the disastereoselectivity of the glycosyl-donor species and the regioselectivity (that is, differentiation of the reactivities) at the glycosyl-acceptor species are important prerequi- sites for success. Protection strategies and suitable procedures for activation of the anomeric carbon atom are required; in addition, the coupling step must occur diastereoselectively with respect to formation of an a or P link- age. The high glycosylation potential of variously protected 0-glycosyl trichloroacetimidates, their excellent a/P diastereoselectivity generally found, and their high regioselectivity often observed with partially O-pro- tected sugar acceptors will be documented here.

b. 0-Glucosyl Trichloroacetimidates as Donors. - D-Glucose (63,64) plays a central role in the formation of plant polysaccharides (for instance, such homoglycans as cellulose and starch). Also, the heteroglycan repeating- units of many bacterial, plant, and animal polysaccharides contain glucose in a- and P-glycosidic linkage. As a constituent of the oligosaccharide moie- ties of glycosphingolipids and glycoproteins, D - ~ ~ U C O S ~ is less frequently en- countered. Glycosphingolipids contain D-glucose in the core region, where it is P-glycosidically linked to ceramide. In N-glycoproteins, a-linked ~-g lu- cose is a terminating signal in the biosynthesis of the complex oligosaccha- ride chains; fully developed glycoproteins do not contain glucose.

The synthesis and application of 0-glucosyl trichloroacetimidates is fo- cused on 0-benzyl- and 0-acetyl protected derivatives ( 1,52a) because these two protective groups have proven to be the most valuable in glycoside synthesis. Representative examples of trichloroacetimidate formation are collected in Table I (la- la). As already outlined (Section 111. I ) , the glucosyl trichloroacetimidates are obtained in high yields and the diastereoselectivi-

Page 58: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

ANOMERIC-OXYGEN ACTIVATION FOR GLYCOSIDE SYNTHESIS 35

ties observed for the base-catalyzed addition of the 1 -0-unprotected glucose derivatives to the electron-poor trichloroacetonitrile are remarkable, thus providing a- and p-glucosyl trichloroacetimidates, respectively, depending mainly on the reaction conditions. The reaction conditions have not yet been optimized in all examples described here and in subsequent sections; this is partly attributable to the fact that a,p-mixtures can be tolerated in glycosylation reactions when neighboring-group participation controls the diastereoselectivity in glycoside-bond formation.

For reaction as 0-nucleophiles with 0-glycosyl trichloroacetimidates, al- cohol components generally require the presence of an acid catalyst (1 - 3). Boron trifluoride etherate (BF3 * OEtJ at -40°C to room temperature in dichloromethane or dichloromethane - n-hexane as solvents and trimethyl- silyl trifluoromethanesulfonate (Me3SiOTf) at - 80°C to room temperature in ether or acetonitrile, respectively, as solvents have proved to be eminently suitable (52a,62). This is exemplified by the reactions of la-a and la$ with various acceptors (Tables I1 and 111). It should be noted that the results reported in the tables generally have not been optimized. Obviously, even at low temperatures, la-a exhibits high glycosyl-donor activity, thus providing generally the p-products in high yields and diastereoselectivities (Table 11, reactions with 2A-2M). The reaction of 2E exhibits that the (much less reactive) thioglycosides are not affected; therefore the products obtained may be used immediately for further glycosylations. However, glycosylation of 2E with the corresponding glucosyl fluoride as donor was not successful. The low diastereoselectivity found (71) for the reaction with acceptor 2D is rather unexpected. It may be due to the use of trifluoromethanesulfonic acid as catalyst, which as a Brnrnsted acid should interfere differently with the donor la-a. 0-Acyl-protected acceptors 25 and 2K, having 0-acetyl protec- tion vicinal to the accepting hydroxyl group, proved to be less reactive, and lower a,p selectivities were found in their glycosylation with la-a. 0-Acetyl protective groups at other positions did not affect the convenient p-glycoside formation.

Thus far, a-glucopyranoside formation has not been extensively investi- gated (67) because this connection is less frequently found in glycoconju- gates. However, it was observed that, with p-trichloroacetimidate la$ as donor, stronger catalyst systems, as for instance Me3SiOTf, favor formation of the thermodynamically more-stable product, especially when the reac- tions are performed in ethers as solvents (Table 111; reactions with 3 4 2G, and 2F) (67).

The influence of solvents in glycosylation reactions has been observed and discussed extensively already (1,4,74). For instance, the participation of ethers, when anomeric leaving-groups are removed under SN1 -type condi- tions, results [because of the reverse anomeric effect (75,76)] in the genera-

Page 59: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

TABLE I1

Reaction of the Benzyl-Protected Glucosyltrichloroacetimidate l a a with 0-Nucleophiles

Anomeric configuration Yield

Glycosyl acceptor Reaction conditions (%) Reference

CH,Cl, , BF, . OEt, , 1:13 78 - 18"C, 2.5 h

HO

CH2C12, BF, * OEt, , 1:19 85

28 Bno&,+, BnO CH,CN, Me,SiOTf, 1.16 89

OCH, CH,CH,CN, Me,SiOTf, 1.16 83

-4O"C, 2 h

-4O"C, 20 min

-4O"C, 20 min

-SOT, 15 min

2A &- CH,CH,CN, Me,SiOTf, 1:16 74

CH,Cl, , BF, . OEt, , 1:6 70 - 35°C

2c

BnO

OBn

/OH

51,52a

5 1,52a

62

62

62

65

2D BnO-,,, BnO

BnO

CHzCl,, CF3SO,H, - 20°C

1.2: 1 86 71

Page 60: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

w .I

2E

2F

2G

2H

21

2J

OBn /

Ho+-4?l BnO

0%

HO F? OBn

/

HO O\/CCI, BnO

AcO

HO BnO &Ov CCI,

AcO

/OBn

HO O\/CCl, AcO

Aco

CH,Cl,-n-hexane, 0: 1 80 72 BF,*OEtZ, - IOOC, 3 h

CHzClz, BF3 * OEtz , - 30°C. 2.5 h

1 :4 81 51,52a

CH3CH,CN, Me,SiOTf, 1:19 81 62 -SOT, 10 min

CHzCl,, BF, * OEt, , 1 : l O 90 71 -38"C, 1.5 h

CH,Cl,, BF3. OEtz , MS 4 A, - 70°C

CH,Cl,, BF3 - OEt, , MS 4 A, - 70°C

0: 1 96 73

0: 1 94 71

CH,Cl,, BF3. OEt, , 1 : 2.5 46 73 MS 4 A, -70°C

(continues)

Page 61: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

TABLE I1 (continue4

Glycosyl acceptor

Anomeric configuration Yield

Reaction conditions (a:B) (%) Reference

W m

2L

2K O\/CCI,

Aco

BnO F? OBn

CH,Cl,, BF, . OEt, , 1.8: 1 45 13 MS 4 A, -70°C

CHzCl,, BF, * OEt, , -35"C, 3.5 h

0: 1 32 51,52a

CH,CH,CN, Me,SiOTf, 1 :24 72 62 -8O"C, 10 min

Page 62: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

ANOMERIC-OXYGEN ACTIVATION FOR GLYCOSIDE SYNTHESIS 39

TABLE 111 Reaction of the Benzyl-Protected Glucosyl Trichloroacetimidate la+ with 0-Nucleophiles

Anomeric configuration Yield

Acceptor Reaction conditions (To) Reference

/OH

/OH

Rn 2G

HO

/OB"

OCH,

Et,O, Me,SiOTf, 8: 1 85 67

Et,O, Me,SiOTf, 5 : 1 89 67 room temp., 2 h

- 10°C, 1.25 h

CH,CH,CN, Me,SiOTf, 1 :24 74 62 - 8 0 T , 20 min

Et,O, Me,SiOTf, 3: 1 95 67 room temp., 5 h

Et,O, MeJiOTf, 3 : 1 72 67 room temp., 6 h

tion of equatorial oxonium ions ( p configuration in D-glucopyranose); these favor via invertive attack of the acceptor the formation of the thermody- namically more-stable axial products (a configuration in D-glucopyranose) (Scheme 9).

The dramatic effect of nitriles as participating solvents in glycosylation reactions was first observed in 0-glycosylations with 0-glucosyl trichloracet- imidates ( 5 1,53). This effect demonstrated that, independent of the configu- ration of the glucosyl donor, in the presence of a strong catalyst and at low temperatures, p-glucopyranoside formation is favored (see Tables I1 and 111; reactions with 2B, 2F, and 2M). The explanation (Scheme 9) that fast kinetic a-nitrilium - nitrile - conjugate formation providing the P-product precedes formation of the thermodynamically more-stable p-nitrilium - nitrile - conjugate, which then could also furnish a products as previously observed (77), was supported by several findings. Excellent leaving-group abilities even at low temperatures are required for the application of this methodol- ogy, and therefore, aside from trichloroacetimidates, not all leaving groups can be used in this highly useful reaction (78).

Page 63: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

P 0

Catalyst: Y R%%x + Acceptor:A-H

Ro \ SN2 or SN2-Type

(Nonpolar Solvents) S,2 or SN2-Type

(Nonpolar Solvents) [Polar (Donor) Solvents]

OR q @ A-H; @ =E@

J F l 4T " R e @ - - 7.

OR A

"Intramolecular" (B=C=Solvent) i

Me

XI- :

SCHEME 9.-Glycosylation Reaction courses.

Page 64: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

ANOMERIC-OXYGEN ACTIVATION FOR GLYCOSIDE SYNTHESIS 4 1

From these results there emerges a general picture of the reaction of trichloroacetimidate donors that is summarized in Scheme 9. In nonpolar solvents and with BF, - OEt, as catalyst at temperatures as low as possible S N ~ (or presumably it is better to say S~2-type) reactions (via a tight ion-pair) take place. With stronger catalysts, as for instance Me,SiOTf, a highly reac- tive carbenium-ion intermediate that favors kinetic attack from the a face is generated. However, with ethers and the result of reverse anomeric attack, fast transformation into a p-face shielded intermediate takes place, leading to formation of the a product; whereas with nitriles, on account of conjugate formation, a-face shielding remains efficient. A cyclic eight-membered transition state, leading to intramolecular glycoside-bond formation as shown in Scheme 9 may be hypothesized to explain the high reactivity and selectivity.

Oligosacharides as donors bearing glucose at the reducing end and having at least nonparticipating 2-0-protection provided essentially the same re- sults (Table IV) (66,79). For instance, trichloroacetimidate formation with NaH as base gave the donors 4a,b in high yields and with high a-selectivity (65). Their reaction with acceptor 2G furnished, under BF, * OEt, catalysis at low temperatures, exclusively /3 products (65). Consideration of recent findings (see foregoing) should lead to improved yields in these reactions (62).

The reaction of glucosyl trichloroacetimidates permitting neighboring- group participation through 2-0-acyl protection [see for instance, the tri- chloroacetimidates 1 b-a$ (Table I)] exhibits generally clean p-product for- mation regardless of the configuration of the starting material (1) (Table V). However, the examples clearly show that the donor reactivity is lowered by acyl protection. Therefore, good yields are still attainable with reactive ac- ceptors, but not as readily for acceptors of low reactivity. However, with Me,SiOTf as catalyst, very promising results even for less reactive acceptors were obtained (see Table V). In the Koenigs-Knorr reaction, orthoester formation was found to be a major drawback in these kinds of reactions (4). The mildly acidic nature of the trichloroacetimidate method decreases the problem of orthoester formation, thus leading to greatly improved glycosi- dation yields.

Because of the presence of a C = C double bond in the sphingosine moiety, 0-acyl protected glucosyl donors received general attention in the synthesis of glycosphingolipids (GSL). As a consequence of the many problems en- countered with direct glucosylation of ceramides, employing all known gly- cosylation procedures, the introduction of the “azidosphingosine glycosyla- tion” methodology, namely, glucosylation of azidosphingosine (70,84 - 95) (for instance, compounds 6A-6D, Table VI) and then attachment of the fatty acyl group to the amino group liberated from the azido function, led to

Page 65: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

TABLE IV Reaction of Glucosyl Trichloroacetimidates of Oligosaccharides with the Nucleophile 2G

Anomeric Trichloroacetimidate Reaction configuration Yield

Glycosyl donor formation conditions (a:B (Yo) Reference

4a-a,p NH CH,Cl, , NaH, CCl,CN, CH,Cl,, BF, . OEt, , 0: 1 5 1 65 room temp., 93%, -4O"C, 2 h

Bn0 oJL3 a:/9 11: 1 BnO & OBn

4b-a

CH,Cl, , NaH, CCl,CN, CH,Cl, , BF, - OEt, , 0: 1 40 65 room temp., 5 h; 96% -35"C, 5 h

OAC OBn

I CCI,

Page 66: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

TABLE V

P w

Glycosides and Saccharides from Acetylated Glucosyl Trichloroacetimidates

Anomeric configuration Yield

Glycosyl donor Glycosyl acceptor Reaction conditions b : B ) (Yo) Reference

OAc

CHZCl,, BF3 * OEtz , 0: 1 85 80 - 30°C

1 b-P CCI,

BnO 58

OCH,

CHZCl,, BF3 * OEt2, MS 4 A, -20°C

0: I 25 81

lb-P

CHZClz , BF3 * OEt, , 0: 1 67 51,52a H O e N O t 5C room temp., 2 h

H O e N O I 5 c

CHZCl2, BF3. OEtz , 0: 1 14 51,52a room temp., 2 h

CH2Cl,, BF3 * OEt, , 0: 1 64 51,52a room temp., 45 min

(continues)

Page 67: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

TABLE V (continued)

Anomeric configuration Yield

Glycosyl donor Glycosyl acceptop Reaction conditions (&:PI (YO) Reference

YOAC

O Y N H

5a-a

CCI,

'OAc 5b-u

HOCH2COOCH3 (5F)

HO(CHz),COOCH3 (5G)

A d OAC AdoYNH HO(CH2),COOCH, (5H)

ACO

CCI, 5c-a

HOCHzCOOCH2Ph (51)

HO(CH2),COOCH2Ph (5J)

0: 1 7 8 51 CH2C12, BF, . OEt, , room temp.

CH2C12, Me3SiOTf, - 20"C, 45 min

0: 1 81 82

CH,C12, Me,SiOTf, 0: 1 65 83

CH,Cl,, Me,SiOTf, 0: 1 13 83

CH,C12, Me,SiOTf, 0: 1 7 2 83

CH,Cl,, Me,SiOTf, 0: 1 71 83

CH2C1,, Me,SiOTf, 0: 1 67 83

- 15"C, 30 min

- W C , 30 min

- 1 5 T , 30 min

- 1 5 T , 30 min

- 1 5 T , 30 min

a Cbz, Z, benzyloxycarbonyl.

Page 68: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

TABLE VI Glycosylation of Azidosphingosine Derivatives with Trichloroacetimidates

Clycosyl acceptor'

OTBDMS OBZ OBZ

6A 68

R d o n conditionsb Refereace Clycosyl dwor

i k

1b-a

I d a

NH

u c c ,

6a-a OAc

Tricbloroacetimidate: Glycosylation:

Tricbloroacetimidate: Glycosylation: Trichloroacetimidate: Glycosylation:

Trichloroacetimidaw

Glycosylation:

See Table I 86 aC, CHzCIz, BFa'OEtz;

See Table I 87 6D See Table I 84 6B, C H z C l z , BF3.0Et2,

room temp.; 94% /3

80% B

C H z C l z , CCI,CN, DBU, 91

6 4 C H p , , Me3SiOTf; 88% a

51968

OAC 0

L3 AcO ACO

AcO 6b-a AcO

OAc

(continues)

Trichloroaoetimidate: CHzCIz, CCI,CN, DBU, 91 97% (I

Glycosylation: 6.4. C H z C I z , BF3.0Et2; 32% /3

Page 69: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

TABLE VI (continued)

P m

A.0- AcO

Ac:* AcO

Tririchloroacetimidate: (CH2C12h, CQCN, DBU, 91

Glycosyhtion: 6A, CH2Cl,, Me,SiOTf;

6Ll, CH2Cl,, Me,SiOTf;

-20°C; 88% (I

64% B

68% fl

Trichloroacetimidate: CH2Cl,, NaH, CC13CN, 84.85

Glywsylation: 68, CH2Cl,, BFl.OEtl; mom temp.; 52% (I

87%)

6d-a

Trichloroacetimidate: CH,C12, NaH, CC13cN, 85 66% (I

Glywsylation: 78%)

68, C H 2 C l 2 , BFl.OEh;

Ca,

Page 70: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

AcO v COOMe

AcO OAC

OAc

5

6h-a

0

Trichloroacehmidate: CHzaz, NaH, m3m, 92

Glycosylation: 92%,a:84:1

BF30Etz; 71%8 6B, CHzCIz-n-hexane,

Trichloroacetimidate: CHZa2, CCI&!N, 93

Glycosylation: 6B, C H z C I z , BFs'OEt,, 93 O'C, 2 h; 94%a

0°C. 4 h; 92%8

OAC AcO OAc Trichlodmidate: CHzClz, ccl,CN, DBU, 94

Glycosybn: 68, CHKlzCl,, BF,.OEt,; 94 87% (2

42% 8 AcO

61-a CCI, 0

(continues)

Page 71: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

P ca

TABLE VI (continued)

Reaction conditionsb Reference Clyeosyl dowr

OAc Trichloroacetimidate:

Glywsylation:

AcO

61-u

I CCI,

6k-u

TrichloroaCetimidate:

Glywsylation:

CH2CIz, CCllCN, DBU; 96

68, CH2Clz, MeaiOTf 96 87% a

(0.01 eq.), room temp.; 75% p

57

97

TBDMS, tert-butyldimethylsilyl. DBU, 1,8-diazabicyclo[5,4,0]undec-7ene.

Page 72: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

ANOMERIC-OXYGEN ACTIVATION FOR GLYCOSIDE SYNTHESIS 49

a major breakthrough in GSL synthesis. This is demonstrated in Table VI, which includes representative examples of this development. Thus, not only were simple glycosyl and lactosyl derivatives made readily accessible, but all major glycosphingolipid series were successfully synthesized, including gan- gliosides that contain neuraminic acid. The importance of tumor-associated antigens of glycosphingolipid nature also created interest in several L-fu- cose-containing glycosphingolipids, for instance, the Lewis X ( Lex) and Lewis Y (Ley) antigens. The acid sensitivity of the fucosyl anomeric bond generally requires special attention in glycoside-bond formation. However, it turned out that these 0-acetyl protected donors did not cause any prob- lems under the required reaction conditions. On account of the high accep- tor reactivity of the azidosphingosines, orthoester formation as a side reac- tion was encountered for the first time (85). In many instances a slightly higher catalyst concentration readily overcomes this problem. The problem may also be solved with the help of 2-0-acyl-protective groups, which for steric (2-0-pivaloyl) or electronic (2-0-benzoyl) reasons do not undergo orthoester formation as readily as do 2-0-acetyl groups (70,85).

As just discussed, “ceramide glycosylation” seemed to cause major prob- lems when the Koenigs-Knorr method was used (84). However, 0-acetyl protected glycosyl trichloroacetimidate donors also provided acceptable yields with glucosyl or the lactosyl trichloroacetimidates (Table VII) (84,98 - 108). For higher oligosaccharide donors the results were unsatisfac- tory. However, again by attaching the bulky 2-0-pivaloyl group to the glu- cose moiety, this drawback may be overcome (84,101), a result which rein- forces the search for further improvements in this most active field.

c. 0-Galactosyl Trichloroacetimidates as Donors. - D-Galactose (63,64) is a constituent of complex glycosphingolipids and glycoproteins, where it plays an important role. It is found as a terminal or subterminal building block in a variety of different connections. In glycosphingolipids, galactose is part of the lactosyl ceramide core-structure. Terminal and subterminal p- ( 1 + 4)-connection to 2-acetamido-2-deoxy-~-glucose (N-acetylglucos- amine) leads to the N-acetyllactosamine moiety, which is preferentially rep- resented in the lacto and the neolacto series. The a-( I + 4) and a-( 1 --* 3) connection determines the gala, globo, and isoglobo series. In glycoproteins, terminal galactose is a signal of the Ashwell receptor, whose function consists in the binding of galactosylated glycoproteins in the liver. In the asparaghe- connected glycan residues of N-glycoproteins, galactose is mainly found in N-acetyllactosamine, whereas in the serine- or threonine-connected 0-gly- coproteins, galactose is preferentially p-( 1 --* 3)-linked to 2-acetamido-2- deoxy-D-galactose (N-acetylgalactosamine). This connection is also met in the ganglio and the isoganglio series of glycosphingolipids.

Page 73: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

TABLE VII Glycosyhtion of Ceramides by Trichloroacetimidates

HNq (CH2)13CH3 HNK ( C H z ) t P h HNK (CHz)zzW H N A (W)zzCH3

0 0 0

\ (CH2)1ZCH3 HOW ( C H ~ ) ~ Z C H ~ HOW (CHz)izCH3

OBZ OTBDMS 7 (CHz)&H, How 5 OAc

HO = \

OBZ OBI 78 7c 7D 7A

Ttkhloro8ceimid.te Glyeasyhtion Yield (%) Ref- formatkllP eoDditioas Glymyl dwor

See Table 1 7A, C H z C l z , BF,.OEtz 57 86 7B, CH2Clz, BFl.OEt2, 70 98 See Table I

1b.iY l k

mom temp.

7 8 4 R = AC; C C I 3 C N , 7 c 13 100

ma: R = Piv; 52% 7C, Me,SiOTf 66 101 7c-a R = TMP 7D, Me,SiOTf 27 102

DBU, 79% L3 ACO AcO

OAC

NH OAc

7da . 83% OL 7C, Me,SOTf 31 101

Page 74: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

ACO

COOMe

AcO

OAC 0

OAc n r , / "nr I

NH

0 AcO 1

7 ~ : R = CH,; CCIlCN,

7fa: R = CH,OAc; CCIICN,

7D, BFi.OEt2,

7 4 BF,.OEt,, DBU, 92% (~P,),

DBU, 16% ( ~ Z ~ Z L

I o k ACO

7C, BF,.OEh

52 103

58 103

1 104

Page 75: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

52 RICHARD R. SCHMIDT AND WILLY KINZY

TABLE VIII Synthesis of Trichloroacetimidates of D-Galactose"

Trichloroacetimidate Reaction conditions Yield (To) Reference

En? ,OBn

CCl,CN, CH,CI, , NaH, 83 46 room temp. E n 0

I CCI,

E n 0 OBn

CC13CN, CH2C12, K2C03, 84 67,68

aa-p BnO ~ o y c c l , BnO room temp. NH

AcO OAc

CCI,CN, CH,Cl, , Na, 39 a -t 45 /3 66,104,109 AcO room temp.

CCI,

CCl,CN, CH,CI, , NaH, room temp., 1.5 h

60 85

LCI,

BnO OBn

BnO

CCI,

CCl,CN, CH,C12, DBU 80 I10

ACO oac

CCI,CN, CH2C12, DBU, I1 111 -5°C

&-a En0

CCI,

(continues)

Page 76: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

ANOMERIC-OXYGEN ACTIVATION FOR GLYCOSIDE SYNTHESIS 53

TABLE VIII (continued)

Yield (%) Reference Trichloroacetimidate Reaction conditions

To10 OTol

CCI,CN, CH,CI, , NaH, 76 112 ~ O ' C C I , MS 4 A, O"C, 2 h

8f-a

To'o To10

8g-a CCI,CN, CH,CI, , NaH, room temp.

77 113

CCI,

Tol, Toluyl; DBU, 1,8-diazabicyclo[5,4,O]undec-7-ene.

1 -0-Unprotected galactose derivatives may be readily transformed into the trichloroacetimidates 8a-8g, as shown in Table VIII. Again, as demon- strated for the 0-benzyl-protected compounds 8% either the a-trichloroace- timidate 8a-a or the P-trichloroacetimidate 8a-/l may be obtained highly selectively, depending on the base used for the catalysis of the addition to the trichloroacetonitrile.

Galactosylation with the 0-benzyl-protected donors 8a-a and 8a-P( Table IX) shows that conditions can be found for invertive product-formation in high yields. Thus, from 8a-P in ether, preferentially a products were formed, and from 8a-jl in the rather nonpolar solvent-mixture dichloromethane - n- hexane, mainly the /3 product was obtained. The higher tendency of galacto- syl donors to effect a-glycoside bond-formation compared with the corre- sponding glucosyl donors is well established in the literature (4) and is also observed here. This may be attributed to the generally higher reactivity ofthe galactosyl donor and to the axial 4-substituent.

2-0-Acyl-protected galactosyl donors readily provide j? products. The reactivity may be increased by having partial 0-benzyl protection, as exhib- ited (1 10,111) with donors 8d-a and 8e-a (Table X). The examples permit very successful 2-0-, 3-0-, and 4-0-connections, respectively. The high- yielding synthesis of the /%Gal-( 1 + 3)-GalNAc building-blocks [8f-a + 10H, Table X (1 12), and 8a-a + 9C, Table IX (1 15)] furnishes a convenient access to 0-glycoprotein moieties; for instance, selective removal of the l-O-ButMe,Si protective group in the 8a-9C P-product (Table IX) and subsequent P-trichloroacetimidate formation leads to the desired P-Gal-

Page 77: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

111 P

TABLE IX Glycosidation with Benzylated Galactosyl Trichloroacetimidates"

Anomeric Reaction Yield configuration

Trichloroacetimidate Glycosyl acceptor (W (a :B) Reference conditions

OH

OBn

Ho 6 BnO OCH, 2F

8 2G

OH OBn

,OBn

(C,H,),O, TBDMSOTf, 75 5 : 1 67 room temp., 0.75 h

(C2H,),0, Me,SiOTf, 65 8: 1 67 room temp., 5 h

(C,H5),0, Me,SiOTf, 66 3 6 : 1 67 room temp., 1 h

(C,H5),0, Me,SiOTf, 77 8: 1 67 room temp., 3.5 h

(C,H5),0, Me,SiOTf, 15 1 :o 114 -20°C

Page 78: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

8a-a

8a-a

8a-a

8a-a

8a-a

HO L$gyornDMs 9c

HO BnoG 9D

HO OTBDMS

OH

CH,Cl, - n-hexane, 84 1 : 7 115 BF, . OEt,

CH,Cl,- n-hexane, 80 1 : 4 116 Me,SiOTf, -25°C

CH,CH,CN, Me,SiOTf, 75 0: 1 72 -40°C

CH,Cl,- n-hexane, 83 1 : 3 12 BF3*OEt2, -25°C

CH,Cl,-n-hexane, 49 1 :o 117 Me,SiOTf, - 3 0 T , 2 h (2-0)

TBDMS, lei?-butyldimethylsilyl.

Page 79: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

TABLE X Glycosylation with Acetylated Galactopyranosyl Trichloroacetimidatese

An o m e ri c Trichloro- Reaction Yield configuration acetimidate Glycosyl acceptor conditions (%I (a:j?) Reference

'>( 8b-a o P

HO ! @ Y O T E D M S 9c

HO OEzl

N3 1 oc

CHCl,, BF,-OEt,, 33 0: 1 118 M S 3 A

31 0: 1 11s CHCl,, BF, . OEt,, M S 4 A

CH,CI, - n-hexane, 67 0: 1 117 BF, * OEt,, room temp., 1 h

CH,Cl,-n-hexane, 69 0: 1 117 BF3*OEt2, -2O"C, (3-0) 2 h

Page 80: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

En? ,OBn

1 OD N3

?CH,CH,

6en

CH,Cl,- n-hexane, BF3*OEtz, -2O"C, 2 h

CH,CI,, BF3. OEt,, M S 4 A

CHzCIz, Me,SiOTf, MS 4 A, - 3 0 T , 20 min

CH,CI,, Me,SiOTf, MS 4 A, - 30°C, 10 min

CH2C12, Me,SiOTf, lOT, 15 min

CH,Cl, - n-hexane,

room temp., 1 h BF3 * OEt,,

65

73

81

87

93

75

0: 1 117

0 : 1 I10

0: I 1 1 1

0 : 1 1 1 1

0: 1 112

0: 1 113

a TBDMS, terf-butyldimethylsilyl.

Page 81: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

58 RICHARD R. SCHMIDT AND WILLY KINZY

(1 4 3)-a-GalNAc-( 1 - 0)-Ser glycopeptide moiety (1 15). The reaction of 8e-a with thioglycoside (1 1 1) 10F demonstrates again that a thio group at the anomeric position is compatible with application ofthe trichloroacetimidate method.

Among the gangliosides, GM4 [a-NeuAc-(2 --* 3)-&Gal-( 1 - 0)-Cer] has a relatively simple chemical structure. It has been detected in human and chicken brain and also (1 19) as a major ganglioside of mouse erythrocytes, chicken-embryonic liver, and egg yolk. With the help of the azidosphingo- sine glycosylation it has been synthesized very efficiently from the neura- minic acid-containing galactosyl donor l la$ (Table XI) ( 120 - 122). Simi- larly the thio isomer was obtained from 11 b-8 and ( 120,123) the positional isomer from 1 lc-a.

d. 0-Mannopyranosyl Trichloroacetimidates as Donors. - D-Mannose (63,64) is less frequently encountered in glycosphingolipids (only in the arthro series); however, it is generally a constituent of N-glycoproteins: a central a-Man-( 1 ---* 6) [a-Man-( 1 -, 3)]Man trisaccharide moiety, which is p-( 1 + 4)-connected to a chitobiose unit, is part of the core structure.

Mannopyranosyl trichloroacetimidates that have been synthesized are compiled in Table XII. Because of the stronger anomeric effect (1,45,75), a-trichloroacetimidate formation is much faster than observed for corre- sponding glucose and galactose derivatives, and therefore the a-trichloro- acetimidates were generally isolated thus far. This was not regarded as a disadvantage because a-mannopyranoside formation, for instance from 12a-q should be readily achieved because of the stronger anomeric effect under thermodynamic reaction control. The examples in Table XIII show that this is indeed the case; selective a-product formation was observed even with BF, - OEt, as catalyst. However, clean p-product formation from the a anomer 12a-a under invertive conditions has not yet been achieved (5 1). Even the nitrile effect, as found recently, led to only partial success in this endeavor (62) (Table XIII). The ready formation of the a-mannopyranosyl linkage is also true for the 2-0-glycosylated 0-mannopyranosyl trichloroa- cetimidates 14a-a-14e-a (Table XIV). Use of Me,SiOTf as catalyst would be presumably superior in these reactions.

2-0-Acyl protection should lead, as a consequence of neighboring-group participation and the anomeric effect, exclusively to a products. This has been proved in many experiments (Table XV); with Me,SiOTf as catalyst excellent yields could be obtained in cases where all other methods essen- tially failed (129). It could be shown that at least some of the reactions proceed via rapid orthoester formation (129), and this intermediate then rearranges under Me,SiOTf catalysis to the desired reaction product.

Page 82: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

TABLE XI Glycosylation of Trichloroacetimidates of D-Galactose with Sphmgosine Derivative 6B

Glycosyl donor

~~

Yield Reaction Trichloroacetimidate formation conditions (%) Reference

&-a See Table VIII

NH CH,ClZ, CCl,CN, DBU, AcO COOMe

~ o @ o ) , c c , , f f :p OT, 1:8 2 h; 79-93%,

0 ACO 092 1la-B

NH ACO COOMe

CH2C12, CCI,CN, NaH, 0°C; a : P O : 1,85%

Acb 'OBz llb-P 0

CHzClz, CC1,CN a:p11:1

CHzClz, BFjOEtz, room temp.

CH2CI2, BFjOEt2, 0°C

CHzCl2, BFjOEtz, 0°C

CH& BFjOEtz

96 B 85

82P 120,121,122

70 P 123

78 P 120

Ca,

Page 83: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

60 RICHARD R. SCHMIDT AND WILLY KINZY

TABLE XI1 Trichloroacetimidates of D-Mannose

Anomeric config. Yield

Trichloroacetimidate Reaction conditions (a# (%) Ref.

BnO

CH2Cl,, CCl,CN, NaH, room temp., 0.5 h

I CCI,

CH30 I

1 :o 99 46,124

CC13CN, NaH

I I CCI,

ACO

CCI, BnO

CH,Cl,, CCl,CN, NaH, 0°C -room temp., 20 min

(ClCH,),, CCl,CN, DBU - 5°C

CHzClz, CC&CN, K2C03, la

1 :o 46 125

1 :o

1 :o

1 :o

n.n.

98

86

91,126

127,128

129

e. Trichloroacetimidates of Glucosamine Derivatives as Glycosyl Donors.-2-Acetamido-2-deoxy-~-glucose (N-acetylglucosamine) (63,64, 1 19) is an important constituent of all glycoconjugates. In the glycan chains of N-glycoproteins it is part of the core and of the glycan side-chains. In glycosphingolipids of the lucto and the lactoneo series, it is the main constit- uent. In proteoglycans, in bacterial lipopolysaccharides, in the murein of

Page 84: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

ANOMERIC-OXYGEN ACTIVATION FOR GLYCOSIDE SYNTHESIS 6 1

TABLE XI11 Reactions with Mannosyl Trichloroacetimidate 12a-a

Acceptor

Anomeric Reaction config. Yield

conditions @:/I) (%) Ref.

YJ' HO J3P

BnO BnO + OMe

,OBn

13A

28

2G

2M

CH2CI2, BF3.OEt2, - W C , 2 h

CH2Cl2, BF3. OEt2, 20°C. 5.5 h

CH& BF,. OEtz, 20°C, 20 h

CH3CH2CN, Me3SiOTf, - 8 0 T , 20 min

CH3CH2CN- n-hexane (1 : 4), Me3SiOTf, - 8 0 T , 20 min

CHZCl2, BF3. OEtz, 20°C 4 h

CH$H,CN, Me,SiOTf, -8O"C, 10 min

5 : 1

l : o

1 :o

1: l

3:2

1 :o

1: l

83

73

68

77

70

66

71

51

51

51

62

62

51

62

bacterial cell-walls, and as a polycondensation product in chitin it has wide distribution. In N-deacetylated form as free glucosamine it was identified as a constituent of glycosylphosphatidylinositols, which are membrane anchors for cell-surface glycoproteins (1 36).

This wide distribution is accompanied by a variety of different linkages, as compiled in Table XVI. Obviously, p-connection is generally favored.

(i) Glucosamine Donors. - The great number of trichloroacetimidates synthesized thus far underlines the fact that compounds displaying high reactivity and high diastereocontrol are required for the great variety of

Page 85: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

TABLE XIV Glycosides and Saccharides from Mannosy1 Tnchloroacetimidates

Trichloroacetimidate Reaction conditions Reference Trichloroacetimidatd formation

AcO OAc

I

O Y N H

CH2C12, CCl,CN, NaH; 14A, BF,-OEt,, 130,131 MS 4 A, 45% a 95% a

OYNH CCl,

AcO .OAc

CH2C12, CCl,CN, DBU, 14B; 10% a 133,134 O T , 30 min; 87% a! 14C, BFjOEt,,

14D, BF, . OEt,; CH2Cl,; 60% a

53% a-(l+ 6) 14G, 58% a

CCI,

Page 86: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

I

z )-$

%To'

SO

P

PO

0

20

Page 87: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

TABLE XIV (continued)

14A 14D

,OBn . .^. ..

NPMh t lnu -

140 14E

Page 88: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

OBn

'OBn 14C

0

14F

Page 89: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

TABLE XV

Glycosylation of Acetylated Trichloroacetimidates of DMannose

Trichloro- Reaction acetimidate Glycosyl acceptor conditions (%) Reference

Yield

CH,Cl,, Me3SiOTf, 59 135 15A MS 3 A, - 10°C a41 -P 6)

10 min AcO

12c-a 135

OH CH,Cl,, Me,SiOTf, 60

MS 3 A, -20°C CX-( 1 + 6) HO 20 min

AcF* ACO

I

158

88 a 127 CH2C12, Me3SiOTf, MS 4 A, - 30"C, 10 min

12c-a

15D ACO (3% HO

12c-a A c t d o CH,Cl,, Me,SiOTf, 92 a 126 -3O"C, 10 min

Page 90: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

m .I

12c-a

ACO HO

12c-a

15F

/-J ,o

15G

12c-a BnO

151 L n

OH

CH2C12, Me,SiOTf, 94 a MS 4 A, -3O"C, 10 min

CH,Cl,, Me,SiOTf, 15 a MS 3 A, room temp., 30 min

CH,Cl,, Me,SiOTf, 12c-a (2.3 q.),

86 a-( 1 + 3) a-( 1 4 6)

MS 3 A, room trisaccharide temp.

n.n. 90 a 3-012-0 1:3

CHzCIZ, Me,SiOTf, 81 a MS 3 A, room temp.,

10 min

CH,Cl,, Me,SiOTf, 82 MS 3 A, - 1O"C, CY-( 1 + 6 ) 10 min

126

135

135

91

135

135

Page 91: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

68 RICHARD R. SCHMIDT AND WILLY KINZY

TABLE XVI Naturally Occurring Glycosidic Linkages of N-Acetylglucosamine

Glycosidic linkage Acceptor Occurrence

p-( 1 + 4) GlcNAc Chitobiose core structure of N-glycoproteins 8-( 1 + 4) MurAc Part of murein of Gram-negative bacteria p-( 1 + 6) GlcNAc p-(l 6, GalNAc Part of core structures of the 0-glycoproteins

Disaccharide unit of lipid A (as in Salmonella minnesota)

p-( 1 4 3) GalNAc

'-(I + 3, Gal lacto- and neolacto-series of glycosphingolipids 8-(1 + 6) Gal p-(1 + 3) Man artho-series of glycosphingolipids a-( 1 - 6) GlcA Phosphoglycosphingolipids of tobacco leaves p-(1 -2) Man Phosphoglycosphingolipids

glycoside bond-formation processes encountered. Compounds capable of neighboring-group participation through N-acyl or N-phthaloyl groups (Table XVII) are readily obtained from glucosamine. Presumably on ac- count of the size of the N-phthaloyl group, only P-trichloroacetimidates (17c-P- 17f-P) were obtained (1 39 - 144). However, for the N-acyl-protected compounds 17a-a and 17b-a it could be shown that the glycosylation reac- tion proceeds via intermediate oxazolines (1 37); therefore, an advantage for application of the trichloroacetimidate procedure could not be established in these cases. The N-phthaloyl-protected trichloroacetimidates permitted an enormous improvement in terms of yield and diastereoselectivity. However, the removal of the N-substituent from the glycosides sometimes caused problems. Therefore, 2-azido-2-deoxyglucose derivatives, readily obtained from glucals via the azidonitration methodology of Lemieux and Ratcliffe (1 52), seemed to be ideal; various trichloroacetimidates were accordingly prepared (Table XVII). Careful investigation of trichloroacetimidate (17g) formation (145) led again to conditions for the selective formation of both anomers. Also noteworthy is the selective formation (149) of the 4-0- unprotected trichloroacetimidate 17i-P. Because the azido group is consid- ered a nonparticipating group, it remained to be shown that the a-trichlor- oacetimidates can be transformed cleanly into P-glycosides under S~2-type conditions.

Experiments (1 37,137a) with the N-phthaloyl-protected donor 17c-/l showed excellent glycosyl-donor properties, as indicated in Table XVIII. Various galactose- and galactosamine-derived acceptors underwent success- ful reaction. The 0-benzyl-N-phthaloyl-protected donors 17d,e, and f showed comparable properties ( 14 1,143).

Page 92: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

m

rn

c€* 13

- m

0

4

m

I-

- .. 0

hl

9

4

9

hl

m

4

.. 0

69

Page 93: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

TABLE XVII (continued)

4 0

Tnchloroacetimidate

Anomeric Reaction configuration Yield

conditions (a:8) (%) Reference

171-p

17e-P

17g-a

17h-0

ACO BnO &oycc, NPhth

NH OBn

/

BnO BnO -OuNH

CCI, N3

CCI,

(OB"

&a, YOTBDMS

171-a HO

CH2Cl2, CCl,CN, 0: 1 90 K2C0,, 20°C, 4 h

DME, CC13CN, NaH, 0°C 4 : 1 98

CHZClZ, CQCN, NaH, 1 :o 15 room temp.

CH2Cl2, CCl3CN, NaH, O'C, 12 h

CH2C12, CC13CN, NaH, room temp.

1 :o 15

141,142

145

53

5435

148

1 :o 98 149

CCI,

Page 94: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

OTBDMS /

17j-p

N3 NH

OTBDMS

17k-a

I /OAC CCI,

171-a ACok-

O Y N H CCI,

/OAC

NH

/OAC

NH

CH,Cl,, CC13CN, Kzco3, room temp., 4 h

CH,Cl,, CC13CN, KZCO,-NaH, room temp., 4.5 h

0 : 1 66b 149

1 :o 66b 150

CH,Cl,, CC13CN, 1 :o 44b 137,147 NaH, room temp., 1 h

CH,Cl,, CC13CN, Kzco3, room temp., 6 h

1:2 7 56 137

CHzCl,, CC13CN, 0: 1 n.n. 151 K2c03

a pMP, pmethoxyphenyl; pMBn, pmethoxybenzyl; DME, 1,2-dimethoxyethane; TBDMS, tea-butyldimethylsilyl. From an epimeric mixture.

Page 95: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

TABLE XVIII Reaction of 2-N-Phthaloyl Trichloroacetimidate 17c-B with Nucleophiles

An o m e ri c Reaction configuration Yield

Glycosyl acceptor conditions (am (To) Reference

CHjOH (1BA) BF, eOEt2, - 30°C 0: 1 65

OTBDMS HO 10H

N3 CH,O

HO Qo+ OCH,

OBn

BF3.0Et2, -20°C 0: 1 70

Me,SiOTf, -7O"C, 0: 1 93 5 min

BF3*OEt2, -20°C

AcO A c F ~ ~ o B & O C H 3

HO OH 1BD OAC

Me,SiOTf, OT, 15 min

0 : 1

0 : 1

71

68

Me,SiOTf, 5°C 0: 1 75 3,6-di-0-

1 BE glycosylation BzO

HO OH

140

140

138, 153

139

139

139

Page 96: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

ANOMERIC-OXYGEN ACTIVATION FOR GLYCOSIDE SYNTHESIS 73

The corresponding 2-azido derivatives revealed surprisingly similar re- sults: high reactivity was combined with extraordinary p selectivity. Table XIX lists many important glycoside-bond formation reactions ( 145) with the 0-benzyl-protected glycosyl donor 17g-a. Only the reaction with the sterically hindered muramic acid acceptor 19E led, in the presence of Me,SiOTf, to partial a-product formation (57); however, this problem could be readily overcome by replacing the bulky ButMe,Si protective group by the benzyl group, as shown (148) for 19F. Remarkable also are the reactions (149) of acceptor 17i-p with the partially protected acceptors 19B and 19G: regioselective reaction at the 6-position and clean P-product for- mation was observed. The high tendency for p-product formation with the a-trichloroacetimidate derivatives of azidoglucose as donors is also because of the fact that the solubility of the compounds permits the use of the rather nonpolar solvent-mixture dichlormethane - n-hexane, which favors S N ~ - type reactions in the presence of BF, * OEt, as catalyst and low tempera- tures.

An interesting example showing that a-product formation may be readily achieved by the use of a p-trichloroacetimidate and Me,SiOTf as catalyst is shown in Table XX: compounds 17n-j3 + 20A furnish exclusively the a-gly- coside in high yield (1 5 1).

(C) Lactosamine Donors. - The general importance of lactosamine in glycosphingolipid and glycopeptide synthesis is because of the frequent oc- currence of this building block (1 19). For instance, the branching of the pentasaccharide core of N-glycoproteins is determined by the connection with N-acetyllactosamine, which may occur in p-( 1 --* 3) linkage in long chains. In 0-glycoproteins, N-acetyllactosamine is part of the core of mucin-type oligosaccharides. Likewise, the core structure of the glycosphin- golipids of the luctoneo series is determined by N-acetyllactosamine.

The connection of these naturally occurring lactosamine units determines the protective-group pattern of the required building blocks. The general occurrence of the p linkage permits again the use of N-phthaloyl protection; however, azidolactose (1 5 3 , readily obtained from lactal, also should be very useful as a consequence of the advantages of this group already dis- cussed. Both of these types of trichloroacetimidates, having different protec- tive groups, have been very successfully prepared, as indicated in Table XXI. Again, as just discussed, with N-phthaloyl protection exclusively p-trichloro- acetimidates 21a-/I-21e-/l were obtained (130,139,140,143,144,156- 159). Both isomers may be selectively generated from the azidolactose derivatives, as shown (137, 160) for 21j. With sodium hydride as the base, a-tri- chloroacetimidates are obtained in very high yields. Some of these com-

Page 97: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

TABLE XIX Reaction of 2-Azido-2-deoxyglucopyranosyl Trichloroacetimidates with Nucleophiles

Anomeric Trichlora- Reaction configuration Yield

(%) Reference acetimidate Glycosyl acceptor. conditions (a$)

17g-c~

17g-a

4 P

HO OH

lgB 17g-c~

X H 3

17g-c~

N3 19c

CH,CI, - n-hexane, Me,SiOTf, - 50T, 10 min

CH,C12 - n-hexane, BF,*OEt,, -3O"C, 20 h

CH,Cl,- n-hexane, BF,*OEtz, -15°C

CH,Cl,- n-hexane, BF3*OEt2, -2O"C, 3 h

CH,Cl,- n-hexane, BF,.OEtz, - WC, 2 h

0: 1

0: I

0: 1

0: I

0 : 1

60

80

80

92

90

146

146

I50

145

154

N3 9c

Page 98: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

17g-a ,,XH

HO O&o& OTBDMS

10H N3

CH2Clz - n-hexane, 0: 1 60 154 BF3 * OEt2, - 18"C, 3 h

CH2Cl, - n-hexane, 0: 1 70 154 pMpXH

BF, OEt,, - 15 "C, 6 h O&o& OTBDMS HO

17g-a

19D N3

17g-a OTBDMS

1 : 1.6 90 57 CH2CI, - n-hexane, Me3SiOTf, - 1 5 "C,

Ho&OIBDMS I N3 5 h 19E

17g-a CH2C12- n-hexane, 0: 1 78 148 BF3OEt2, -2O"C,

N3 8 h 19F -.as" H A

CH3 COOMe

(continues)

Page 99: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

TABLE XIX (continued)

Anomeric Trichloro- Reaction confignration Yield

acetimidate Glycosyl acceptor' conditions (a$) (%) Reference

HO OH CH2C12 - n-hexane, 0: 1 85 149

BF30Et2 198

17i-/3

OCH,

CH2C12 - n-hexane, 0: 1 50 149 BF3 * OEt2 6 )

N3 19G

pMP, pmethoxyphenyl; TBDMS, ?err-butyldimethylsilyl.

Page 100: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

TABLE XX a-Selective Glycosidation of a j?-Trichloroacetirnidate"

Reactants Products

20A B"O 111.1"'

BnO 1711-p

a pMBn, pmethoxybenzyl.

Page 101: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

TABLE XXI Synthesis of Trichloroacetimidates of N-Acetyllactosamine

Anomeric Reaction configuration Yield

Trichloroacetimidate conditions (a:B (%I Reference

.OAc

A C O p o &‘y ‘“3

2la-p NPhth NH

AcO OAc

OBn

A C O e 0 & 0 y ‘“3 NPhth

NH 21 b-p

BnO OAc

-0MP

.OBn

21d-p AcO#o&O NPhth CCI,

NH Aco 06”

OAc

CH,CI,, CC13CN, DBU

CH,Cl,, CCl,CN, DBU, O”C, 3.5 h

CH& CC13CN, DBU

CH,Cl,, CC13CN, KZCO,

0: 1

0: 1

0: 1

0: 1

0: 1

12

92

87

12

66

130,139,140,156

151

143,114

158

159

Page 102: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

U

L

N

r

PI 00

- .. d 0

&

&

F;

f - - .. d

3qo 0

u

N

$

3

.. 3

0

79

Page 103: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

TABLE XXI (continued)

Tric hloroacetimidate

Anomeric Reaction configuration Yield

conditions (a : 8) (%I Reference

9: 1 5 3" 1 37,160

.OAc

21j-b A c o ~ o ACV- \- K I \

AcO OAC

1 :5 64" 137

From epimeric mixture.

Page 104: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

ANOMERIC-OXYGEN ACTIVATION FOR GLYCOSIDE SYNTHESIS 8 1

pounds are already suitable for further connections in the 3-,3’-, 4’-, and 6’-positions (1 14,115,137,155,160).

Several glycosylation reactions with N-phthaloyl-protected donor ( 140) 21a-j? have been very successfully performed, as indicated in Table XXII. The j?-selectivities and the yields are generally very good. The reaction (1 30) with 22C demonstrates that dilactosaminylation can also be successfully achieved. The reaction of the 3’,4-0-unprotected 0-benzyl-lactose deriva- tive (1 57) 22D led, contrary to the generally observed higher reactivity of the 3’-position, to reaction at both positions. This problem could be overcome by employing the 0-acyl protected lactosamine derivative 23A (Table XXIII). With donor 21e-j?, Veyrikres et al. (1 59) obtained tetrasaccharide 23B in high yield. This reaction could be repeated with the derived tetrasac- charides 23c as donor and 23D as acceptor, thus leading to the corresponding octasaccharide in good yield.

As already observed for azidoglucose-derived donors, glycosylations with azidolactose-derived donors (21f-a - 21 j-a, Table XXIV) also exhibited high reactivityand~selectivity(92,114,115,154,161,162). Withtheseresults in hand, excellent preconditions for successful syntheses of the Le” and L e y

antigens have been presented (1 64,165). Representative examples for the decisive glycoside-bond formations are compiled in Table XXV. Compari- son of the results of N-phthaloyl protection and of the azido group does not exhibit advantages for the use of N-phthaloyl derivatives.

(iii) Chitobiose Donors. - Only a few trichloroacetimidate-based chito- biose donors have been synthesized thus far, as indicated in Table XXVI. Their reaction with benzyl alcohol as acceptor demonstrates the potential usefulness of these donors in glycosylation reactions.

( iv ) Muramic Acid as Donor. -The cell-wall peptidoglycan of bacteria has a p-( 1 - 4)-linked glycan chain, consisting of alternating 2-acetamido- 2-deoxy-~-glucose and N-acylmuramic acid residues that are cross-linked by a peptide chain. The resulting peptidoglycan network (murein) and its frag- ments exhibit marked immunostimulatory and antitumor properties. The minimal structure for activity, the so-called Freund‘s complete adjuvant, is a “muramoyl dipeptide” (MDP). Many investigations have been directed toward the synthesis of derivatives of MDP, including glycosides and oligo- saccharides; the attachment of lipophilic groups is of special interest because of their potential in combined chemotherapy and immunotherapy ( 166,167).

The transformation of azidoglucose derivatives into muramic acid pre- cursors enabled the formation of trichloroacetimidates as muramic acid donors that could be very successfully employed in glycoside bond-forma-

Page 105: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

TABLE XXII Reaction of 2-N-Phthaloyl-2deoxytric~oroaceti1nidate 2la-p with Nucleophiles.

Anomeric Reaction configuration Yield

Glycosyl acceptor conditions (a$) (W Reference

0 m N

22c BnO

~~

0: 1 69 156 CH2C12, BF, *OEt2, - 8 T , 3.5 h

(CH2Cl)2, BF3.OEt2 0: 1

(CH2C1),, MS 4 A, 0 : 1 BF3*OEtz, - 15°C

CHZCI,, BF, * OEtz, 0 : 1 -2O'C

BF,*OEt,, -20°C

0: 1

73 132

67 140

75 139

Page 106: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

m W

22D

22E

22F

22G

Hoe n.n. 0: 1 88 157 /3-( 1 : 3’) pq1 : 4’)

2: 1

0: 1 87 157 (CH,Cl),, BF3*OEt, (2.0 eq.), room temp.

NPMh N3 BF,.OEt,, -20°C 0: 1 71 140

22 102

‘OBn

22H Me3SiOTf, -20°C 0: 1 80 139 6 ) ACO

OAC

0 Z, benzyloxywbonyl; pMBn, pmethoxybenzyk TBDMS, tm-butyldimethylsilyl; TMB, 2,4,6-trimethylbenzoyl.

Page 107: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

84 RICHARD R. SCHMIDT AND WILLY KINZY

TABLE XXIII Synthesis of Oligomers of Lactosamine (Ref. 159)

Reactions and products

R O &omy&o+o&x R'O

OAc NPMh RO OAC OAC

238 R = H. X = OBn. A'. R"= -C(CH$,

23D R = Ac, X- OBn. A', R"- H 23C R = Ac. X = OC(-NH)CCi,, R'. R"- -C(CH&

tion. Table XXVII shows that a- and /?-trichloroacetimidates 27a-a and -/? may be obtained directly and also that disaccharide donors were successfully prepared. These compounds can be used for selective /?- and a-glycoside bond-formations (57,97).

f. Trichloroacetimidates of Galactosamine Derivatives as Glycosyl Donors. - 2-Acetamido-2-deoxy-~-galactose (N-acetylgalactosamine) (63, 64) is a constituent of the core structure of mucin-type oligosaccharides; it is a-U-connected to serine and threonine. The derived U-glycoproteins con- stitute, along with the N-glycoproteins, a major class of glycoconjugates. In glycosphingolipids, N-acetylgalactosamine is mainly encountered in the globo, isoglobo, and ganglio series. Representative examples of these con- nections are compiled in Table XXVIII. Obviously, /?-( 1 + 3)-, /?-( 1 + 4)-, and a-( 1 + 3)-connections are most important and therefore N-phthaloyl protection is not appropriate for the production of versatile donors.

Table XXIX demonstrates that various protective-group patterns are compatible with trichloroacetimidate formation, and not only a but also /? derivatives may be generated highly selectively, as for instance 29b-/? (149)

Page 108: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

TABLE XXIV Reaction of 2-Azido-2-deoxytrichloroacetimidates of Lactosamine with Nucleophiles

Trichloro- Reaction Yield acetimidate Glycosyl acceptor. conditions (%) Reference

21f-a

2lf-a

2 1 g-a

21h-a

21i-a

CH,Cl, - n-hexane, BF, * OEt,, - 15°C

CH,Cl,- n-hexane, BF, * OEt,

CH,Cl, - n-hexane, BF, * OEt,, MS 4 A, -25°C

CH2C12 - n-hexane, Me,SiOTf, - 20 "C, 1 h

CH2Cl, - n-hexane, BF3 * OEt,, -15°C

115

154

92

114

162

a Z, benzyloxycarbonyl; TBDMS, tea-butyldimethylsilyl.

Page 109: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

TABLE XXV Synthesis of Oligosaccharides with W Determinants

Glycosyl donor Glycosyl acceptor

"&OY cc13 HO+o.&&OBn OBn

NPMh NH HO OBn 22D

'%$OBn 25a-P Trichbmacetimidate Formation: CHzC12. CC13CN, DBU; 67 % Glycosylatbn Condnbns: (CHzC1)z, BF30Elz; 67 % B (1-3)'04)

BnO

OBn OBn

I den BnO

OBn

BnO

Tkhbmacetimidate Formation: n. n. Glycosylatbn Condaions: (CHZCI)~. BF30Etz: 52 % B'")

Tkhbmacetimidate Formation: CHzCIz, CC13CN, DBU,

Glycosylatbn Conditions: CH,Cl.$bhexane. 1 l), MS 4 A. -25'C, 5 h, m m temp.: 94 %, a : j3 5 : 1

BF30Et2: 81 %p3Li

Page 110: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

OBn 25e-a

/OBn OH

(continues)

Page 111: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

m m

TABLE XXV (continued)

Glycosyl donor Glycosyl acceptor

ox eel, H o e 0 +oBn

25F

OBn Glycosylation Condiions: CH3CN, TMSOTI 0 02 eq.),

OBn

-40°C; 80 % pl'\

HO@ 0 & OBn

BnO OBn OPiv

25G

Glycosylation Condliins: CH CN TMSOTI 0 02 eq ), & ~ ; 74% &4 ' '

Page 112: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

m W

TABLE XXVI Glycosylation of Chitobiose Derivatives

Trichloroacetimidate Glycosyl Clycosylation Glycosyl donor. formation acceptor conditions References

NH

BnO

NPhth NPhth OAc OMP

CCl,CN, DBU (CICH,),, BF, . OEt,, 127 (CICH,),, -5"C, Ar; H O B

96% -2O"C, Ar; 87% (two steps) 26A

NH

BnO

NPMh NPMh OH OMP

CCl,CN, DBU (C1CH2),, BF, * OEt,, 127,142 -55"C, 30 min; -23"C, Ar; 84% 78.6% 26A

MP, pmethoxyphenyl.

Page 113: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

TABLE XXVII Synthesis of Glycosides of Muramic Acid

CH,Cl,, CCl,CN, NaH, 40°C; 90% (Ref 57)

CH,Cl,, CH,Cl,, K2C03, room temp.; 86% (Refs. 57,97) ** 0 +cia NH

A H na-p C W e

CH,Cl,, K,CO,/NaH, CCl,CN, room temp., 8 h 75%, a:p 6: 1 (Ref. 148)

ccb m

Anomeric Reaction Yield configuration G 1 y c o s y 1

donor

27a-a

G 1 y c o s y 1 acceptor conditions (%a) (a:j?) Reference

0 CH,Cl,, room 60 0: 1 57 HO-!'-(OBn) (27Al temp., 3 h

57 27a-o CHzC12, -2O"C, Me,SiOTf, 4 h

70 1 :o

Ho&mn

0

CH,Cl,, -2O"C, Me,SiOTf

71 3 : 1 57

Page 114: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

27a-a CH,Cl, - n-hexane 92 - lOT, BF3 * OEtz, 3 h

CH,Cl, - n-hexane 80 - lOT, BF, . OEt,, 30 min

CH2C1, - n-hexane 80 - S T , BF, * OEtZ, 6 h

27a-a

27a-P

27a-B

HNZ

Ho-lfoBn 0

EhO, MS 4 A, N,, 91 -2O"C, 2 h, Me3SiOTf

CHzCl,, MS 4 A, 3 h, BF3 * OEt,

85

0: 1 57

0: 1 51

1:4 57

1 :o 91

0: 1 97

CHZCl,, BF3 * OEh, 38 0: 1 97 room temp., 14 h

27a-a

BtlO "3

Page 115: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

TABLE XXVIII Structures of N-Acetylgalactosamine-Containing Glycosphingolipids

Gala-Series

Globo-Series a-GalNAc-( 1 + 3)-/3-GalNAc-( 1 - 3)-cu-Gal-( 1 + 4)-/3-Gal-( 1 + 0)-Cer

&GalNAc-( 1 + 3)a-Gal-( 1 + 4)-/?-Gal-( 1 + 4)-/?-Glc-( 1 + O)-Cer a-GalNAc-( 1 + 3)-/%GalNAc-( 1 - 3)-cuGal-( 1 + 4)-/?-Gal-( 1 - 4)-/%GlO( 1 + 0)-Cer D-Gal-( 1 - 3)-/?-GalNAc-( 1 + 3)-cr-Gal-( 1 + 4)-/?-Gal-( 1 - 4)-/?-Glc-( 1 -+ 0)Cer

BGalNAc-( 1 + 3)a-Gal-( 1 + 3)-/?-Gal-( 1 + 4)-/?-Glo( 1 - 0)-Cer

Fa lNAc-( 1 + 4)-&Gal-( 1 - 4)-/?-Glc-( 1 + 0)-Cer /3-Gal-( 1 - 3)-/?-GalNAc-( 1 + 4)-/?-Gal-( 1 + 4)-/?-Glc-( 1 + 0)-Cer

a-GalNAc-( 1 - 3)-/?-Gal-( 1 + 3)-/?-GlcNAc-( 1 - 3)-/?-Glc-( 1 - 4)-/?-Glc-( 1 - 0)-Cer

Globotetranosylceramide Forssman antigen Globopentaosylceramide

Isoglobotetraosylceramide

Gangliotriaos ylceramide Gangliotetraosylceramide

Isoglobo-Series

Ganglio-Series

Lacto-Series

2

a 1 Fuc t

Arthro-Series /?-GalNAc-( 1 - 4)-/%GlcNAc-( 1 - 3)-/?.-Man-( 1 + 4)-/?-Glc-( 1 + 0)-Cer a-GalNAc-( 1 + 4)-/?-GalNAc-( 1 - 4)-/3-GlcNAc-( 1 + 3)-&Man-( 1 + 4)-/?-Glc-( 1 + 0)-Cer

4-OMe-/?-Gal-( 1 + 3)-/%GalNAc-( 1 + 3)-a-Fuc-( 1 - 4)-pGlcNAc-( 1 + 2)-Man Phosphogl ycosphingolipids

(Fragment)

Page 116: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

TABLE XXIX Synthesis of Trichloroacetimidates of N-Acetylgalactosamine

Anomeric Yield configuration (W (a:B) Reference Reaction conditions Trichloroacetimidate

CH,Cl,, CCl,CN, NaH, 68 l h

1 :o 137 BnO

29a-a

O Y N H

CH,Cl,, CCl,CN, K,C03 88 0: 1 149

BnO ,OMEM

(CH,Cl),, CCl,CN, DBU room temp., 3 h

3: 1 169

BnO %

(CHZCI),, CCI,CN, DBU, room temp., 2 h

81 1 :o 169 29du

I CCI,

29e-0

(CHZCl),, CCl,CN, DBU, room temp.

79 1 :o 169 BnO

kCI3

(continues)

Page 117: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

TABLE XXIX (continued)

Trichloroacetimidate

Anomeric Yield configuration

Reaction conditions (%I Reference

W P

CCl,

AcO O k

29h-8 AcO $&oyccl,

NH N3

(CHKl),, CCI,CN, DBU, 81 1 :o 169 room temp., 2.5 h

DME, CCI,CN, NaH, 2 h

CH,Cl,, CCl,CN, K,CO,, 6 h

CH,CI,, CCl,CN, NaH, I h

64 1 :o 137

5 1" 0: 14 137,116,170

63" 1 :o" 137

Page 118: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

FI 4

d

00

FI d

c

5

4

0

d

I

z )-$

? w .-

? 6

N

? w

95

Page 119: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

TABLE XXIX (continued)

Trichloroacetimidate

Anomeric Yield configuration

Reaction conditions (No) (a:B) Reference

CCl,

CCI,

NH

CH2C12, CCl,CN, K,CO,/NaH, 95 1 :o room temp., 6 h

CH2Cl2, CCl,CN, K2CO3

CH2C12, CCl,CN, NaH, room temp.

76 0: I

75 1 :o

116

150

150

150

Page 120: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

5: 4

0

2

5: -

W

v)

.$ 0

9" m

0

-

0

0 P

d

0

91

Page 121: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

98 RICHARD R. SCHMIDT AND WILLY KINZY

and 29i-p (1 13). Useful building blocks for efficient oligosaccharide syn- theses are thus readily accessible. Trichloroacetimidates 29b-i ( 1 16, 137,149,169,170) are versatile building blocks for 3,6-branched core-struc- tures of mucin-type oligosaccharides. The selective formation of compound (1 37) 29g-a from the corresponding 1,3-0-unprotected azido-galactose de- rivative demonstrates again that only partial 0-protection may be required, because the anomeric hydroxylic group is more reactive toward trichloroa- cetonitde under basic conditions than the other hydroxyl groups. This aspect, which could decrease the number of protection and deprotection steps, has not yet been fully considered in the planning of complex of oligo- saccharide syntheses.

The first glycosylation experiments were carried out with donor (149) 29b-p, which with Me,SiOTf as catalyst exhibited high a selectivities; with the a-trichloroacetimidates (169) 29d-f and BF, * OEt, as catalyst in the nonpolar solvent toluene, excellent p selectivities were observed. In more recent glycosylations the a-connection to serine played a prominent role. Typical results with monosaccharide and oligosaccharide donors having azidogalactose at the reducing end vary (Table XXX). As expected, reac- tions with a-trichloroacetimidates, employing BF, - OEt, as catalyst, are not a selective. Obviously, p-trichloroacetimidates and Me,SiOTf at low temperatures are of advantage for attaining high a selectivity, as indicated in the reaction of donors 29b, g, h, m, and p, and serine and threonine acceptors 30A - E.

g. Trichloroacetimidates of Mannosamine Derivatives as Glycosyl Donors. -The relatively rare occurrence of 2-acetamido-2-deoxy-~-man- nose in Nature has consequently drawn little attention to its glycosylation reactions. The azido derivatives 31a-a and 3b-a (Table XXXI) have been successfully prepared. Reaction of 31 b-a has been successfully employed for phosphonate formation.

h. Trichloroacetimidates of 6-Deoxyhexoses: Fucose, Rhamnose, and Quinovose. - (9 0-Fucopyranosyl trichloracetimidates: Inverse Procedure for Glycosylation. - L-FUCOS~ is an important constituent of glycosphingo- lipids. Because most of the tumor-associated blood-group glycosphingolip- ids have been found to contain a-connected ~-fucose, for instance Le" and Ley, a-fucosylation constitutes an important task in glycosphingolipid syn- thesis ( 174). To this aim, the tri-0-benzylfucosyl donor (1 75,176) 32a (Table XXXII) has been prepared in high yield. Reaction with galactose acceptors led, with Me,SiOTf as catalyst in ether, to high yields of H-disaccharide ( 174), the determinant ofblood group 0. With the (less reactive) lactosamine derivatives as acceptors, lower yields were observed mainly because of de-

Page 122: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

TABLE XXX Glycosylation with Galactosamine Trichlomcetimidates

Trichloroacetimidate Glycosyl acceptor

Anomeric Reaction Yield con6guration

conditions (%I (a:n Reference

29b-/?

29g-a

29h-a

W W 29h-8

29h-8

2911-8

2911-8

HNZ

HO-lfOBn 0

HNBoc

0

HNBoc

30A CH,Cl,, Me,SiOTf, - 20T, 30 min

30A CH,Cl,, Me,SiOTf, -20°C

30A CH,Cl,, Me,SiOTf, - 15°C

30A CH,Cl,-n-hexane, - 30°C, Me,SiOTf

30B CH,Cl,-n-hexane, - 30°C, Me,SiOTf

3OC CH,Cl,- n-hexane, - 30T, Me,SiOTf

81 4: 1 116,150

43 1 :o 137

81 5: 1 171

86 1 :o 116

55 1 :o I16

60 1 :o 116

HNZ

30D CH,Cl,-n-hexane, - 30T, Me,SiOTf

80 1 :o 116

2911-8

HNFMOC

0

30E CH,Cl,-n-hexane, - 30T, Me,SiOTf

78 1 :o 116

(continues)

Page 123: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

TABLE XXX (continued)

Anomeric Yield configuration Reaction

Trichloroacetimidate Glycosyl acceptor conditions (YO) @:B) Reference

29m-p

29n-a

29p-a

29i-a

z

29j-a

29j-a

30A CH,Cl,-n-hexane, -2O"C, Me,SiOTf

30A CH,Cl,, Me,SiOTf, - 30°C

30A CH,Cl,, Me,SiOTf, -20°C

30F CH,Cl,, n-hexane, ZnCI,. OEt,, room temp., 15 h

Y O

HO o & o ~ O B n OBn

OBn

(oho@oB,l HO OBn 30G CH,CN, Me,SiOTf, - 40T, 15 min

OBn OBn

30H CH,CN, Me,SiOTf, Ho&oy - 40"C, 15 min

OBn Po COOMe OBn OBn

%,, OAc

AcO

85 l : o 116

86 2: 1 115

88 1 :o 154

81 1 : 1,2 113

46 0: 1 117

38 0: 1 117

Page 124: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

- .. 0 rn

v)

s

m c 0

m'

O&: m"0

0 6

0 2

r" m

N

m

2

0

-

0

L( 0

m

5

W

d

2

P R

c.(

101

Page 125: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

L

8

TABLE XXX (continued)

Anomeric Reaction Yield coafiguration

Trichloroacetiddate Glycosyl acceptor conditions (04 (a:8) Reference

CCI, COOMe

1 :2.7 121 30A (CH,Cl)t, BF, OEt2, n.n. MS 4 A, - 15°C

OAc 30 min

3OC-ff

Page 126: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

c

8

TABLE XXXI Glycosylation of Trichloroacetimidates of 2 - A z i d o - 2 d e o x y - ~ - m e Derivatives

~~

Tricbloroacetimidate Glycosyl acceptor Reaction conditions Reference

AcO AcO & O Y N H

31a-a “‘3

P(OCH,), Trichloroacetimidate formation: 147 31A CHzClZ, CCl3CN, NaH,

room temp. Glycosylation:

CHzClz, Me,SiOTf; 58%, a:/? 6: 1

0

150 II pivop /.Q$ HO-P(OBn), Trichloroacetimidate formation:

CHzCl2, CCl3CN, NaH; 60% aP OPiVo 31B PiVO opiv Glycosylation:

CHzC12, BF, * OEt,, - 10°C; 61% 31b-a O Y N H CCI,

~ ~ ~ ~~~~

From epimenc mixture.

Page 127: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

104 RICHARD R. SCHMIDT AND WILLY KINZY

TABLE XXXII Synthesis of Trichloroacetimidates of Fucose

Anomeric Yield configuration

Compound Reaction conditions (%) (a:n Reference

NH CH2Cl2, CCl,CN,

o ~ c c 1 3 K2C03, room temp. I

e o B n 32a CH2Cl2, CCI,CN, DBU, room temp.

En0

0 I c a 3 CH2C12, CCl,CN, I DBU. room temp.

NH CH,C12, CC13CN, o ~ c c 1 3 NaH, room temp.

CH3 CH,CN, CCl,CN, AcO OAc 32C K2C03, room temp.

BZO

CC13CN, K2C03 BZO

OBZ

32d

50 1 :o 175,58b

65 1 :o 162

79 1 :o 162

71

76

1 :o 58b

2:3 58b

90 1:1 178

composition of the highly reactive fucosyl donor 32a under the reaction conditions. Therefore, an alternative reaction procedure is required.

Glycosylations and also fucosylations are generally carried out as a for- mally termolecular reaction of donor (D), acceptor (A), and promotor or catalyst (C) (depending on the amount required) (1,4). Because of differ- ences in the affinities, the reaction course is expected to be first DC interac- tion, followed by interaction of the DC complex with A (Scheme 10, reaction course I). Obviously, for this sequence of interactions, donors and acceptors with matching reactivities are required. Therefore, acceptor and donor reac- tivities are often vaned by changing the protective-group pattern and, in addition, the donor reactivity is varied by the selection of leaving groups and

Page 128: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

ANOMERIC-OXYGEN ACTIVATION FOR GLYCOSIDE SYNTHESIS 105

D + C + A

SCHEME 10.- Postulated Reaction Courses.

catalysts (1,4). However, this strategy is less successful for very reactive glycosyl donors, which may decompose in the presence of the catalyst while awaiting reaction with the acceptor. Therefore, complexation of acceptor A with the catalyst C prior to interaction with the donor D (Scheme 10, reac- tion course 11) should overcome this problem.

The efficiency of this approach could be demonstrated in a-fucosylation with donor 32a and the acceptors 19C and 33A-D (Table XXXIII). Thus, with the help of this inverse procedure, the versatile building blocks for syntheses of the Lea, Le", Ley, and H antigen determinants are readily acces- sible (1 76). Presumably, this procedure may become of general importance when reactive glycosylating agents are employed. Alternatively, the reactiv- ity of the fucosyl donor could be decreased, as has been recently proven very successfully (177).

Acyl-protected fucosyl donors have also been generated very successfully (Table XXXII) (58b). Their reaction with acceptors led via neighboring- group participation to /3 products (58b, 178).

(Z) 0-Rhamnopyranosyl Trichloroacetimidates. - Rhamnosides are mainly found in plant heteroglycans (63,64). Some rather preliminary in- vestigations have been carried out with rhamnose derivatives. The trichloro- acetimidates obtained as rhamnopyranosyl donors are listed (1 24,178 - 18 1) in Table XXXIV. Their structural similarity to mannose explains the ready formation of a-glycosidic bonds.

(iii) 0-Quinovopyranosyl Trichloroacetimidates. - Quinovosides (6- deoxyglucosides) are found, for instance, as constituents of many saponins, which are composed of a carbohydrate portion attached to an aglycon that is a complex steroid in asterosaponins (182). Their dramatic biological effects have provided a motivation for structure elucidation and also for synthesis (183). The trichloroacetimidate donors 35ad prepared are listed in Table XXXV. They have been successfully used in oligosaccharide synthesis. Likewise, a 6-sulfoquinovosyl trichloroacetimidate has been successfully prepared (58a).

Page 129: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

TABLE XXXIII Reaction of Trichloroacetimidates of L-Fncose with U-Nucleophiles

Reaction Yield Glycosyl donor Glycosyl acceptor' conditions (%) Reference

NH

B " b 32a 19c

L

0 o\

32a

32a

33A

(C2H5),0, Me,SiOTf (0.01 eq.), room temp., IP

(C,H,),O, Me,SiOTf (0.01 eq.), room temp., IP

85 165

78 165

CH,CI,, Me,SiOTf (0.02 eq.), 72 177 room temp., IP, 1.5 eq. 32a (3-0)

CH,CI,, Me,OTf (0.02 eq.), 71 177 N3 room temp., IP, 4 eq. 32a (3,2' di-0)

338

Page 130: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

N

2

g3

F.? e

(0 I

9

"a O+

N

2 ?

-3

N" e

-.

n

zCI)

5 (0

0

m

0

ri 0

107

Page 131: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

TABLE XXXIV Glycosylation of L-Rhamnopyrmosyl Derivatives

Trichloroacetimidate Reaction formation Glycosyl acceptop conditions Reference

Glycosyl donor

NH I1

CHzCl,, ~ T s O H ; 124 E":S+ && OBn OBn 8 6 % ~ room temp., 30 min; 85% CCl,CN, NaH, CH,Cl,,

O A m ,

BnOBnO

34A OBn

34a-a

CH,Cl,, pTsOH; 124 ''& 9 6 % ~ ~

xo 34B OBn

OBn

CH,Cl,, pTsOH; 10% a E n 0

124

Page 132: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

d d

I" x

0 2

o\

I.

3

I

gq=

% 3

0

8

0

2

109

Page 133: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

110 RICHARD R. SCHMIDT AND WILLY KINZY

TABLE XXXV Synthesis of Trichloroacetimidates of D-Quinovose

~~

Anomeric Yield configuration

Glycosyl donoP Reaction conditions (%) (a:m Reference

CHzCl,, CCl,CN, DBU, room temp.

A:* AcO

KCCb 35a

NH

CH,Cl,, CCl,CN, A:* DBU, room temp.

35b oKcc13 NH

CH,Cl,, CCl,CN, DBU, room temp.

n NH

Me

CH,Cl,, CCl,CN, Bno,z95+) BnO DBU, room temp.

3% oKcc13 NH

92 1 :o 183

90

84

88

1 :o 183

6: 1 183

1 :o 183

* All, ally].

i. Trichloroacetimidates of 2-Deoxyhexoses: 2-Deoxy-~-arabino-hex- ose. - The presence of the 2-deoxy-~-~-arubino-hexopyranoside (“2- deoxy-/3-D-glucopyranoside”) moiety in natural products has stimulated various approaches for the selective synthesis of this glycosidic bond (1 84- 189). A temporary 2-phenylthio group as a neighboring group, generating an episulfonium-ion intermediate during glycoside-bond formation, seems to be advantageous because it is also readily removable by hydrogenation, affording the desired 2-deoxy sugar (1 88,189). Successful application of the trichloroacetimidate method to this problem required (i) a convenient syn- thesis of a 2-S-phenyl-2-thio-~-glucose derivative, subsequently (iz) a stable a-trichloroacetimidate, and finally (iii) high diastereoselection in the glyco-

Page 134: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

ANOMERIC-OXYGEN ACTIVATION FOR GLYCOSIDE SYNTHESIS 11 1

syl transfer. This could be accomplished starting from tri-0-benzyl-D-glucal, as shown in Table XXXVI (1 90). The 2-phenylthio-substituted trichloro- acetimidate 36-a! was readily obtained and it exhibited extraordinarily high reactivity; reactions with different acceptors were fast even at temperatures as low as - 95 "C, affording preferentially P-glycosides 36a - d in high yields. Transformation into the desired 2-deoxy derivatives 36A-D was readily achieved by treatment with Raney nickel (190).

Obviously, extension of this methodology to other 2-deoxy sugars and also to selective formation of a-glycoside bonds with 2-deoxy sugars should be feasible. The extension of this methodology to 3-deoxy-2-glyculosonates (for instance, Neu5Ac) is under investigation.

j. Trichloroacetimidates of Glucuronic Acid. - D-Glucosiduronate (glu- curonide) formation is an important means for detoxification in mammals and leads to soluble conjugates that can be excreted via the urine. Glycosides of D-glucuronic acid occur also in many microbial, plant, and animal poly- saccharides (for instance, in heparin) (1 9 1). Glycoside-bond formation with the help of trichloroacetimidates has been accomplished quite successfully (169,192,193). To this aim, donors 37a-c have been synthesized from the 1 -0-unprotected derivatives in high yields (Table XXXVII). Representative examples of their reaction with various acceptors are compiled in Table XXXVIII.

k. Trichloroacetimidates of Pentoses. -Thus far, there has been rela- tively little activity in the application of the trichloroacetimidate method to formation of pentopyranosides and pentofuranosides (46,183,194 - 199). The reported examples exhibit results similar to those already discussed, and thus special limitations are not expected.

1. Reactions of 0-Glycosyl Trichloroacetimidates with N-, S-, C-, and P-Acceptors. - Only a few studies with N-nucleophiles have been per- formed. Hydrazoic acid, as a strong acid, reacts with 0-glycosyl trichloro- acetimidates and readily gives the thermodynamically most stable glycosyl azide without any additional catalyst (53) (Scheme 6). Nitrogen heterocycles require an acid catalyst for reaction; thus, bis-(trimethylsilylated) uracil and thymine gave, with trichloroacetimidate la-a, exclusively the P-linked nu- cleosides at room temperature with boron trifluoride etherate as catalyst (1,53,200). Reactions in nitriles as solvent lead during workup to trapping of nitrilium adducts (53,78).

The strong interest in 1-thioaldoses and 1-thioglycosides (66,201) as a consequence of their recent use as anomeric protecting-groups, and con- comitantly for glycosyl transfer with the help of thiophilic activators, led to

Page 135: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

TABLE XXXVI Synthesis of 2-Deoxy-~~ar~bin~-hexopyranosides

OR EF3OEt2, Et,O/ Bno& CH2C12 BnO

HOR X

36a-d (X = SPh)

+

1. PhSCI. room temp.

E n 0 Na,C03, THF

2. CCI,CN. NaH,

36A-D (X = H)

room temp. (70 %)

36-a cCi3

Anomenc Yield configuration

Glycosyl acceptor Reaction conditions (To) (a :B) Reference # 20"C, lh 36a 90 8: 1 190

: : - . H H

HO H

2F -4O"C, 15min 36b 90 1 :o 190

OMe

2 6 -6O"C, 15min 36c 85 3: 1 190

OBn

190

HO 6 Bno%2J3 BnO -95"C, 15min 36d 83 1 :o

OMe

Page 136: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

ANOMERIC-OXYGEN ACTIVATION FOR GLYCOSIDE SYNTHESIS 1 13

TABLE XXXVII Synthesis of Trichloroacetimidates of D-Glucuronate

Compound Reaction conditions

COOMe

RoRXS+ 373 (R = Bn)

37b (R = Ac)

NaH, CC13CN, CH2C12, 15 min (98%) (Ref. 192)

DBU, CCl,CN, (CH2CI),, I h (92%) (Refs. 169,193) Ro 'YNH

CCI,

COOMe

37c K2C03, CCl,CN, CH2C12, 8 h (73%) (Ref. 193) BnO -&+ oKccI3

OBn NH

TABLE XXXVIII Synthesis of /?-D-Ghcosiduronates

~ ~~

Anomeric Yield configuration

Donor Acceptor Reaction conditions (%) (a! : B, Reference

37a

37a

37a

37b

37b

CH2CI2, BF3 . OEt2,

-25"C, 2 h

88 0: 1 I92

CH2C12, BF3 * OEt,, - 30°C, 2 h

CHZCl2, BF3 . OEt2, - 30°C 2 h

88 0: 1

82 0: 1

192

192

OMe HO

Toluene, Me,SiOTf, -20°C

Toluene, Me,SiOTf - 20°C

75 0: 1 169

72 0: 1 169

Page 137: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

114 RICHARD R. SCHMIDT AND WILLY KINZY

the study of the reactivity of 0-glycosyl trichloroacetimidates in the glycosy- lation of S-nucleophiles (66). In the examples investigated employing 0- acyl- and 0-benzyl protected donors, generally high reactivity was observed. Surprisingly, with the 0-benzyl protected trichloroacetimidate la-a in the presence of boron trifluoride etherate as catalyst, 1-thioglycosides of the a configuration are obtained exclusively. Because the anomeric effect in alkyl 1 -thioglycosides supposedly corresponds approximately to that in alkyl gly- cosides (202,203), under the reaction conditions kinetically controlled /%product formation was expected; under thermodynamic control, both anomers should be formed. Obviously, glycosyl transfer to the S-nucleo- philes in these cases occurs by a different mechanism. It was assumed, that, as in SNi reactions, the configuration is retained by intramolecular reaction via a tight ion-pair (66). Thiocarboxylic acids react again without the addi- tion of any acidic catalyst to provide 1-S-acetyl-1-thio sugars (66,68).

The great interest in C-glycosyl compounds is reflected in the extensive research in this field (204). Successful investigations with 0-glycosyl tri- chloroacetimidates as glycosyl donors and phenol ethers (1 99,207,208), silyl enol ethers (205,206), trimethylsilyl cyanide (205,206), and allyltrimethylsi- lane (206) as C-acceptors underline the wide scope of these highly reactive glycosyl donors.

The biological importance ofglycosyl phosphates prompted interest in the synthesis of glycosyl phosphates as structural analogues (209,2 10). Excellent examples for their synthesis were contributed by the reaction of trichloro- acetimidates 171-a, 29a-a, and 39a-a with trimethyl phosphite in presence of Me,SiOTf as catalyst (21 1) (Table XXXIX). Attack at phosphorus and subsequent 0-demethylation led in a Michaelis - Arbuzov type of reaction to the desired products. Obviously, various other elements or their deriva- tives are conceivable as glycosyl acceptors. These may react either directly as strong acids (as for instance hydrogen halides, see Scheme 6) or as good nucleophiles react in the presence of a catalyst with the highly reactive 0-glycosyl trichloroacetimidates as donors.

IV. OTHER ANOMERIC-OXYGEN ACTIVATION METHODS 1. Other Glycosyl Imidates, Glycosyl Carboxylates, and Glycosyl

Sulfonates

Base-catalyzed addition of glycosyl oxides for anomeric 0-activation has been extended meanwhile to trifluoroacetonitrile (see Scheme 9), to dichlor- oacetonitrile, to 1 -aryl- 1,l -dichloroacetonitriles, and to ketene imines (46,5 1,52). Also 24 glycosy1oxy)-pyridine and -pyrimidine derivatives were readily prepared from the corresponding 2-halo precursors (78). However,

Page 138: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

TABLE XXXIX Reaction of O-Glycosyl Trichloroacetimidates with P(OMe),

Anomeric Yield configuration

Donor Reaction conditions Product (W (a:B) Reference

CH,Cl,, room temp. 1 h, Me,SiOTf

A*-&+ AcO

171-a O Y N H CCl,

Bny

A&% 16 AcO

N3 P(OMe), II 0

l : o 21 1

211

CCI, .OAc

CH,Cl,, room temp. 1 h, Me,SiOTf

AcO

A c O G 59 6: 1

ACO ;;(OMei2 0

21 1

39a-a OYNH CCI3

Page 139: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

116 RICHARD R. SCHMIDT AND WILLY KINZY

none of the imidate donors thus obtained seems to exceed the 0-glycosyl trichloroacetimidates in terms of ease of formation, stability, and reactivity.

Acetimidate formation with N-methylacetamide and acylated glycosyl halides according to Sinay et a1 (2 12,2 13), using three equivalents of silver oxide as an activator, leads neither to particularly stable nor to reactive donors. Any other developments along these lines have already been sum- marized in previous reviews (1,3). The same is mainly true for anomeric 0-activations via 1 -0-acylation (1,2 14), including orthoester (1) formation, 1-0-alkylation ( 1,2 15) and -silylation ( l), and 1-0-sulfonylation (1).

2. Glycosyl Phosphates and Related Systems

One of the most important direct nucleophilic substitutions at activated carbon atoms carried out in nature is enzymic 0- and N-glycosyl bond-for- mation at the anomeric carbon atom (2 16). At this activated position, the leaving groups are phosphates, pyrophosphates, and their nucleoside and lipid ester derivatives, which are biosynthesized via anomeric O-phospho- rylation reactions. In vitro anomeric 0-phosphorylation readily furnishes dialkyl or diary1 glycosyl phosphates (2 17). These also exhibit, in inert sol- vents in the presence of boron trifluoride etherate or Me,SiOTf as catalysts, good glycosyl donor properties comparable to those of glycosyl fluorides and sulfides, respectively, as reported elsewhere (1 7). However, with A=B-C-H systems as acceptors (see Section 111.2), where a catalyst is not required, their reactivity is similar to that of the very reactive trichloroaceti- midate donors, as indicated by competition experiments ( 17). Thus, con- trary to a recent statement (2 18), not only in vivo but also in vitro nucleo- philic substitution at activated carbon atoms, as exemplified by the anomeric center, can be efficiently performed with glycosyl phosphates. This was recently demonstrated not only for glycosyl phosphates but also for such derivatives related to imidates as 0-P (=X)Y2, where X = 0 and Y =

NMe,, X = 0 and Y = Ph, X = S and Y = OMe, and X = NTs and Y =

Ph (219-223).

V. CONCLUSIONS

The requirements for new glycosylation methods outlined at the begin- ning of this chapter, namely convenient diastereocontrolled anomeric 0-ac- tivation (first step) and subsequent efficient diasterecontrolled glycosylation promoted by genuinely catalytic amounts of a catalyst (second step), are essentially completely fulfilled by the trichloroacetimidate method. This is clearly shown by the many examples and references given in this article. In terms of stability, reactivity, and applicability toward different acceptors, the

Page 140: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

ANOMERIC-OXYGEN ACTIVATION FOR GLYCOSIDE SYNTHESIS 1 17

0-glycosyl trichloroacetimidates have generally proven to be outstanding glycosyl donors, which resemble in various respects the natural nucleoside diphosphate sugar derivatives as glycosyl donors. Thus, base-catalyzed gen- eration of 0-glycosyl trichloroacetimidates and ensuing acid-catalyzed gly- cosylation have become a very competitive alternative to direct, often un- controlled acid-catalyzed transformation of sugars into glycosides (Fischer - Helferich method) or to glycosyl halide and glycosyl sulfide formation for the activation step, which requires at least equimolar amounts of a promoter system for the glycosylation step (Koenigs- Knorr method and variations). In addition, the trichloroacetimidate method may be readily adapted for large-scale preparations.

ACKNOWLEDGMENTS

Thanks are expressed to the Deutsche Forschungsgemeinschaft and to the Fonds der Che- mischen Industrie for financial support of our work reported in this chapter.

REFERENCES

1. R. R. Schmidt, Angew. Chem., 98 ( 1986) 2 13 - 236; Angew. Chem. Znt. Ed. Engl. 25 ( 1986)

2. R. R. Schmidt in W. Bartmann and K. B. Sharpless (Eds.), StereochemistryofOrganicand Bioorganic Transformations, Workshop Conferences Hoechst, Vol. 17, pp. 169- 189. VCH Verlagsgesellschaft GmbH, Weinheim, 1987.

3. R. R. Schmidt, PureAppl. Chem., 61 (1989) 1257- 1270; B. M. Trost, I. Fleming, and E. Winterfeldt (Eds.), Comprehensive Organic Synthesis, Vol. 6, pp. 33 -64. Pergamon Press, Oxford, 199 1.

4. H. Paulsen, Angew. Chem., 94 (1982) 184-201;Angew. Chem. Znt. Ed. Engl., 21 (1982)

5 . H. Paulsen, Angew. Chem., 102 (1990) 851 -867; Angew. Chem. Znt. Ed. Engl., 29 (1990)

6. H. Kunz, Angew. Chem., 99 (1987) 297-311; Angew. Chem. Znt. Ed. Engl., 26 (1987)

7. K. Krohn, Nachr. Chem. Technol. Lab., 36 (1988) 930-935. 8. W. Koenigs and E. Knorr, Ber., 34 (1901) 957-982. 9. T. Mukaiyama, Y. Murai, and S. Shoda, Chem. Lett. (1981) 431 -432.

212-235.

155- 173.

823-839.

294-308.

10. K. C. Nicolaou, A. Chucholowski, R. E. Dolle, and J. L. Randall, J. Chem. Sot. Chem.

I 1. S. Hashimoto, M. Hayashi, and R. Noyori, Tetrahedron Lett., 25 (1984) 1379- 1382. 12. H. Kunz and W. Sager, Helv. Chim. Acta, 68 (1985) 283-287. 13. M. Kreuzer and J. Thiem, Curbohydr. Res., 149 (1986) 347-361. 14. T. Matsumoto, H. Maeta, K. Suzuki, and G.4. Tsuchihashi, Tetrahedron Lett., 29 (1988)

15. K. Suzuki, H. Maeta, T. Matsumoto, and (3.4. Tsuchihashi, Tetrahedron Lett. 29 (1988)

16. K. Suzuki, H. Maeta, and T. Matsumoto, Tetrahedron Lett., 30 (1989) 4853-4856.

Commun. (1984) 1155-1156.

3567 - 3570.

3571-3574.

Page 141: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

118 RICHARD R. SCHMIDT AND WILLY KINZY

17. R. R. Schmidt in H. Ogura, A. Hasegawa, and T. Suami (Eds.), Carbohydrates-Synthetic Methods and Application in Medicinal Chemistry, pp. 66 - 88. VCH Verlagsgesellschafl mbH, Weinheim, 1992.

18. P. Fugedi, P. J. Garegg, H. LMnn, and T. Norberg, GIycoconjugateJ., 4 (1987) 97- 108. 19. T. Norberg and M. Walding, Glycoconjugate J., 5 (1988) 137 - 143. 20. R. J. Femer, R. W. Hay, and N. Vethaviyasar, Carbohydr. Res., 27 (1973) 55-61. 2 1. T. Mukaiyama, T. Nakatsuka, and S. Shoda, Chem. Lett. (1 979) 487 -490. 22. J. W. van Cleve, Carbohydr. Res., 70 (1979) 161 - 164. 23. S. Hanessian, C. Bacquet, and N. Lehong, Carbohydr. Res., 80 (1980) C17-C20. 24. P. J. Garegg, C. Henrichson, and T. Norberg, Carbohydr. Res., 116 (1983) 162- 165. 25. T. Y. R. Tsai, H. Jin, andK. Wiesner, Can. J. Chem., 62 (1984) 1403-1405. 26. K. Wiesner, T. Y. R. Tsai, and H. Jin, Helv. Chim. Acta, 68 (1985) 300-314. 27. R. B. Woodward [and 48 collaborators], J. Am. Chem. SOC., 103 (1981) 3215-3217. 28. P. G. M. Wuts and S. S. Bigelow, J. Org. Chem., 43 (1983) 3489-3493. 29. S. Koto, T. Uchida, and S. Zen, Chem. Lett. (1972) 1049- 1052. 30. K. C. Nicolaou, S. P. Seitz, and D. P. Papahatijs, J. Am. Chem. SOC., 105 (1983) 2430-

31. H. Lijnn, Carbohydr. Rex, 139 (1985) 105-113. 32. H. Ldnn, J. Curbohydr. Chem., 6 (1987) 301-306. 33. H. Liinn, Carbohydr. Res., 139(1985) 115- 121. 34. R. Eby and C. Schuerch, Carbohydr. Res., 39 (1975) 33-38. 35. P. Fugedi and P. J. Gareg, Carbohydr. Res., 149 (1986) C9-Cl2. 36. A. Marra, J.-M. Mallet, C. Amatore, and P. Sinay, Synlett (1990) 572-574. 37. R. R. Schmidt and M. Reichrath, Angew. Chem., 91 (1979)497-499; Angew. Chem. Int.

38. H. Bredereck, G. Hagelloch, and E. Hambsch, Chem. Ber., 87 (1954) 3< -37: thetrehalose

39. R. R. Schmidt, M. Reichrath, and U. Moering, Tetrahedron Lett., 2 1 (:, (30) 3561 -3564. 40. R. R. Schmidt, U. Moering, and M. Reichrath, Tetrahedron Lett., 21 ( 1 ~ 'Q, 356<-3568. 41. R. R. Schmidt, M. Reichrath, and U. Moering, Chem. Ber., 115 (1987 42. R. R. Schmidt, M. Reichrath, and U. Moering, J. Curbohydr. Chem., 3 (i2d.t) 67-84. 43. J. J. Oltvort, M. Klosterman, C. A. A. van Boeckel, and J. van Boom, Carbohydr. Res.,

44. R. R. Schmidt and W. Klotz, Synlett (1991) 168- 170. 45. R. R. Schmidt and J. Michel, Tetrahedron Lett., 25 (1984) 821 -824. 46. R. R. Schmidt, J. Michel, and M. Roos, Liebigs Ann. Chem. (1 984) 1343 - 1357. 47. R. R. Schmidtand A. Esswein, Angew. Chem., 100 (1988) 1234- 1236;Angew. Chem. Int.

48. A. Esswein, H. Rembold, and R. R. Schmidt, Carbohydr. Res., 200 (1990) 287-305. 49. J. Tsvetkov, W. Klotz, and R. R. Schmidt, Liebigs Ann. Chem. (1992) 371 -375. 50. J. U. Nef, Liebigs Ann. Chem., 287 (1895) 265-359. 5 1. J. Michel, Dissertation, University of Constance, 1983. 52. (a) R. R. Schmidt and J. Michel, Angew. Chem., 92 (1980) 783-784; Angew. Chem. Int.

Ed. Engl., 19 (1980) 73 1 - 732; (b) R. R. Schmidt, M. Stumpp, and J. Michel, Tetrahedron Lett., 23 (1982) 405-408.

2434.

Ed. Engl., 18 (1979) 466-468.

formation described in this paper may be interpreted accordingly.

1'9.

130 (1984) 147-163.

Ed. Engl., 27 (1988) 1178-1180.

53. R. R. Schmidt and J. Michel, J. Curbohydr. Chem., 4 (1985) 141 - 169. 54. J. E. Truelove, A. A. Hussain, and H. B. Kostenbander, J. Pharm. Sci., 69 (1980) 231 -

55. R. R. Schmidt and M. Stumpp, Liebigs Ann. Chem. (1984) 680-691. 56. R. R. Schmidt, H. Gaden, and H. Jatzke, Tetrahedron Lett., 31 (1990) 327-330. 57. W. Kinzy and R. R. Schmidt, Liebigs Ann. Chem. (1987) 407-415.

232.

Page 142: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

ANOMERIC-OXYGEN ACTIVATION FOR GLYCOSIDE SYNTHESIS 1 19

58. (a) M. Hoch, E. Heinz, and R. R. Schmidt, Curbohydr. Res. (1989) 21 -28; (b) R. R.

59. A. Esswein and R. R. Schmidt, Liebigs Ann. Chem. (1988) 675-678. 60. R. R. Schmidt and C. Regele, unpublished results. 6 1. H. Gaden, Dissertation, University of Constance, 1990. 62. R. R. Schmidt, M. Behrendt, and A. Toepfer, Synlett (1990) 694-696. 63. R. Schaffer in W. Pigman and D. Horton (Eds.), The Carbohydrates: Chemistry and

64. J. F. Kennedy, Curbohydrute Chemistry, Oxford Science Publications. Clarendon Press,

65. R. R. Schmidt and J. Michel, Angew. Chem., 94 (1982) 77-78; Angew. Chem. Int. Ed.

66. R. R. Schmidt and M. Stumpp, Liebigs Ann. Chem. (1983) 1249- 1256. 67. B. Wegmann and R. R. Schmidt, J. Curbohydr. Chem., 6 (1987) 357-375. 68. M. Schuhmacher, Dissertation, University of Constance, 1984. 69. R. Klaer, Dissertation, University of Constance, 1985. 70. P. Zimmermann, R. Bommer, T. Biir, and R. R. Schmidt, J. Curbohydr. Chem., 7 (1988)

71. P. Fugedi, P. Nanasi, and J. Szejtli, Curbohydr. Res., 175 (1988) 173- 181. 72. R. Schaubach and W. Kinzy, unpublished results. 73. T. Takano, F. Nakatsubo, and K. Murakami, Cell. Chem. Technol., 22 (1988) 135- 145. 74. G. WulffandG. Rohle, Angew. Chem., 86 (1974) 173- 187;Angew. Chem. Int. Ed. Engl.,

75. R. U. Lemieux, Pure Appl. Chem., 25 (1971) 527-548. 76. I. TvaroSka and T. Bleha, Adv. Curbohydr. Chem. Biochem., 47 (1989) 45- 123. 77. R. R. Schmidt and E. Rocker, Tetrahedron Lett., 21 (1980) 1421 - 1424. 78. Y. D. Vankar, P. S. Vankar, M. Behrendt, and R. R. Schmidt, Tetrahedron, 47 (1991)

79. G. 0. Aspinall, P. Capek, R. C. Carpenter, and J. Szafranek, Carbohydr. Res., 214 (1991)

80. P. B. Anzeveno, L. J. Creemer, J. K. Daniel, C. H. King, and P. S. Liu, J. Org. Chem., 54

81. N. A. Kraaijeveld and C. A. A. van Boeckel, Red. Truv. Chim. Puys-Bus., 108 (1989)

82. H. Paulsen, B. Helpap, and J.-P. Lorentzen, Curbohydr. Res., 179 (1988) 173-197. 83. T. Toyokuni, personal communication. 84. R. R. Schmidt and P. Zimmermann, Angew. Chem., 98 (1986) 722-723; Angew. Chem.

85. P. Zimmermann, Dissertation, University of Constance, 1988. 86. N. P. Singh and R. R. Schmidt, J. Carbohydr. Chem., 8 (1989) 199-216. 87. T. Biir and R. R. Schmidt, Liebigs Ann. Chem. (1988) 699-674. 89. R. R. Schmidt and T. Maier, Curbohydr. Res., 174 (1988) 169- 179. 90. R.R.Schmidt,T.Biir,andH.-J.Apell,Angew. Chem., 99(1987)813-814;Angew. Chem.

91. M. Masato, Y. Ito, and T. Ogawa, Curbohydr. Res., 195 (1990) 199-224. 92. R. Bommer and R. R. Schmidt, Liebigs Ann. Chem. ( 1989) 1 107 - 1 1 1 1. 93. T. Murase, H. Ishida, M. Kiso, and A. Hasegawa, Curbohydr. Res., 188 (1989) 71 -80. 94. A. Kameyama, H. Ishida, M. Kiso, and A. Hasegawa, Curbohydr. Res., 200 (1990)

95. H. Prabhanjan, H. Ishida, M. Kiso, and A. Hasegawa, Curbohydr. Res., 211 (1991)

Schmidt, B. Wegmann, and K.-H. Jung, Liebigs Ann. Chem. (1991) 121 - 124.

Biochemistry, 2nd ed., Vol. IA, p. 69. Academic Press, New York, 1972.

Oxford, 1988.

Engl., 21 (1982) 77-78; Angew. Chem. Suppl. (1982) 78-84.

435-452.

13 (1974) 157- 172.

9985 -9992.

95- 105.

(1989) 2539-2542.

39-50.

Int. Ed. Engl. (1986) 725.

Int. Ed. Engl., 26 (1987) 793-794.

269-285.

C1 -C5.

Page 143: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

120 RICHARD R. SCHMIDT AND WILLY KINZY

96. A. Toepfer, Dissertation, University of Constance, 1992. 97. A. Toepfer and R. R. Schmidt, Curbohydr. Res., 202 (1990) 193-205. 98. R. R. Schmidt and R. Klager, Angew. Chem., 97 (1985) 60-61; Angew. Chem. Znt. Ed.

99. R. Klager, Dissertation, University of Constance, 1985. Engl. (1985) 65.

100. K. Koike, M. Sugimoto, S. Sato, Y. Ito, Y. Nakahara, andT. Ogawa, Curbohydr. Res., 163

101. S . Sato, S. Nunomura, T. Nakano, Y. Ito, and T. Ogawa, Tetrahedron Lett., 29 (1988)

102. S. Nunomura and T. Ogawa, Tetruhedron Lett., 29 (1988) 5681 -5684. 103. M. Numata, M. Sugimoto, S. Shibayama, and T. Ogawa, Curbohydr. Res., 174 (1988)

104. S. Sato, Y. Ito, T. Nukuda, Y. Nakahara, and T. Ogawa, Curbohydr. Rex, 167 (1987)

(1987) 189-208.

4097 -4100.

73-85.

197-210. 105. 106. 107. 108.

109. 110. 111.

112. 113.

114. 115. 116. 117. 118.

119.

120.

121. 122.

123. 124. 125. 126.

127.

128. 129. 130.

K. Mori and Y. Funaki, Tetrahedron, 41 (1985) 2379-2386. K. Mori and T. Kinsho, Liebigs Ann. Chem. (1988) 807-814. K. Mori and H. Nishio, Liebigs Ann. Chem. (1991) 253-257. K. Ohashi, S. Kosai, M. Arizuka, T. Watanabe, Y. Yamagiwa, T. Kamikawa, and M. Kates, Tetrahedron, 45 (1989) 2557-2570. P. H. Amvam-Zollo and P. Sinay, Curbohydr. Res., 150 (1986) 199-212. K. Sakai, Y. Nakahara, and T. Ogawa, Tetrahedron Lett., 31 (1990) 3035-3038. A. M. P. van Steijn, M. Jetten, J. P. Kamerling, and J. F. G. Vliegenthart, Red. Truv. Chim. Puys-Bus, 108 (1989) 374-383. H. Paulsen, W. Rauwald, and U. Weichert, Liebigs Ann. Chem. (1988) 75-86. P. Zimmermann, U. Greilich, and R. R. Schmidt, Tetrahedron Lett., 31 (1990) 1849- 1852. R. Schaubach, J. Hemberger, and W. Kinzy, Liebigs Ann. Chem. (1991) 607-614. W. Kinzy and R. R. Schmidt, Curbohydr. Rex, 166 (1987) 265-276. W. Kinzy and R. Schaubach, J. Curbohydr. Chem., accepted for publication. U. Greilich, Dissertation, University of Constance, 1992. K. Koike, M. Sugimoto, Y. Nakahara, and T. Ogawa, Curbohydr. Res., 162 (1987)

H. Wiegandt, Glycolipids, New Comprehensive Biochemistry, Vol. 10. Elsevier, Amster- dam, 1985. T. Murase, A. Kameyama, K. P. R. Katha, H. Ishida, M. Kiso, and A. Hasegawa, J. Curbohydr. Chem., 8 (1989) 265-283. H. Iijima and T. Ogawa, Curbohydr. Res., 186 (1988) 107- 118. M. Numata, M. Sugimoto, K. Koike, and T. Ogawa, Curbohydr. Res., 163 (1987) 209- 225. Y. Ito, M. Kiso, and A. Hasegawa, J. Curbohydr. Chem., 8 (1989) 285-294. P. Fugedi, A. Liptik, P. NBnhi, and A. Neszmelyi, Curbohydr. Res., 107 (1982) C5-C8. V. A. Olah, J. Harangi, and A. Liptik, Curbohydr. Res., 174 (1988) 1 13- 120. J. Kerekgyarto, J. P. Kamerling, J. B. Bouwstra, J. F. G. Vliegenthart, and A. Liptik, Curbohydr. Rex, 186 (1989) 51-62. F. Yamazaki, S. Sato, T. Nukuda, Y. Ito, and T. Ogawa, Curbohydr. Rex, 201 (1990)

H. Paulsen and B. Helpap, Curbohydr. Res., 216 (1991) 289-313. H. Paulsen, F. Reck, and I. Brockhausen, Curbohydr. Rex, 236 (1992) 39-71. K. K. Sadozai, T. Nukada, Y. Ito, Y. Nakahara, T. Ogawa, and A. Kobata, Curbohydr. Rex, 157 (1986) 101-123.

237 -246.

31-50.

Page 144: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

ANOMERIC-OXYGEN ACTIVATION FOR GLYCOSIDE SYNTHESIS 12 1

13 1. K. K. Sadozai, T. Kitajima, Y. Nakahara, T. Ogawa, and A. Kobata, Carbohydr. Res.. 152

132. T. Ogawa, M. Sugimoto, T. Kitajima, K. K. Sadozai, and T. Nukada, Tetrahedron Lett.,

133. H. Paulsen, M. Heume, and H. Niirnberger, Carbohydr. Rex, 200 (1 990) I27 - 166. 134. J. J. Krepinsky, personal communication. 135. H. Paulsen, F. Reck, and I. Brockhausen, personal communication. 136. T. W. Rademacher, R. B. Parekh, and R. A. Dwek, Annu. Rev. Biochem., 57 (1988)

137. G. Grundler and R. R. Schmidt, Liebigs Ann. Chem. (1984) 1826- 1847. 137a. R. R. Schmidt and G. Grundler, Angew. Chem., 95 (1983) 805-806; Angew. Chem. Znt.

138. P. Tavecchia, M. Trumtel, A. Veyrikres, and P. Sinay, Tetrahedron Lett., 30 (1989)

139. A. Maranduba and A. Veyrikes, Curbohydr. Res., 151 (1986) 105-119. 140. G. Grundler and R. R. Schmidt, Carbohydr. Res., 135 (1985) 203-218. 141. F.Yamazaki,T. Kitajima,T.Nukada,Y.Ito, andT. Ogawa, Carbohydr. Res., 201 (1990)

142. F. Yamazaka, T. Nukada, Y. Ito, S. Sato, and T. Ogawa, Tetrahedron Lett., 30 (1989)

143. M. Kobayashi, F. Yamazaki, Y. Ito, and T. Ogawa, Curbohydr. Res., 201 (1990) 51 -67. 144. M. Kobayashi, F. Yamazaki, Y. Ito, and T. Ogawa, Tetrahedron Lett. 30 (1989) 4547-

145. W. Kinzy and R. R. Schmidt, Liebigs Ann. Chem. (1985) 1537-1545. 146. W. Kinzy, Diploma thesis, University of Constance, 1983. 147. K. Briner and A. Vasella, Helv. Chim. Actu, 70 (1987) 1341 - 1356. 148. A. Termin and R. R. Schmidt, Liebigs Ann. Chem. (1989) 789-795. 149. W. Kinzy and R. R. Schmidt, Tetrahedron Lett., 28 (1987) 1981 - 1984. 150. W. Kinzy, Dissertation, University of Constance, 1986. 15 1. G. J. P. H. Boons, M. Overhand, G. A. van der Marel, and J.-H. van Boom, Angew. Chem.,

152. R. U. Lemieux, and R. M. Ratcliffe, Can. J. Chem., 57 (1979) 1244-1251. 153. M. Trumtel, P. Tavecchia, A. Veyrieres, and P. Sinay, Carbohydr. Res.. 191 (1989)

154. W. Kinzy and R. R. Schmidt, Carbohydr. Res., 193 (1989) 33-47. 155. K.-H. Jung, M. Hoch, and R. R. Schmidt, Liebigs Ann. Chem. (1989) 1099- 1106. 156. H. Ammann and G. Dupuis, Can. J. Chem.. 66 (1988) 1651-1655. 157. Y. Ito and T. Ogawa, Agric. Biol. Chem., 50 (1986) 3231-3233. 158. S. Sato, Y. Ito, and T. Ogawa, Tetrahedron Lett., 29 (1988) 4759-4762. 159. J. Alais and A. Veyrieres, Tetrahedron Lett., 28 (1987) 3345-3348. 160. W. Kinzy, Proceedings of the 13th Symposium on Carbohydrate Chemistry, Stockholm,

16 1. R. Windmuller, Dissertation, University of Constance, submitted. 162. W. Kinzy and A. Low, Curbohydr. Res., 245 (1993) 193-218. 163. S. Sato, Y. Ito, and T. Ogawa, Tetrahedron Lett., 29 (1988) 5267-5270. 164. R. Bommer, W. Kinzy, and R. R. Schmidt, Liebigs Ann. Chem. (199 I ) 425 -433. 165. A. Toepfer and R. R. Schmidt, Tetrahedron Lett., 33 (1992) 5 161 -5 164. 166. P. Lefrancier and E. Lederer, Fortschr. Chem. Org. Naturst., 40 (1981) 1-47. 167. P. Lefrancier and E. Lederer, Pure Appl. Chem., 59 (1987) 449-454. 168. A. Termin, Dissertation, University of Constance, 199 1.

(1986) 173-182.

27 (1986) 5739-5742.

785-838.

Ed. Engl., 22 (1983) 776-777.

2533-2536.

15-30.

4417-4420.

4550.

101 (1989) 1538- 1539; Angew. Chem. Znt. Ed. Engl. (1989) 1504.

29-52.

1988. [Abstr.]

Page 145: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

122 RICHARD R. SCHMIDT AND WILLY KINZY

169. J.-C. Jacquinet, Carbohydr. Res., 199 (1990) 153- 181. 170. R. R. Schmidt and G. Grundler, Angew. Chem., 94 (1982) 790-79 1; Angew. Chem. Znt.

Ed. Engl. (1982); Angew. Chem. Suppl. (1982) 1707-1714. 171. T. Toyokuni, B. Dean, and S . 4 . Hakomori, Tetrahedron Lett., 31 (1990) 2673-2676. 172. H. Iijima and T. Ogawa, Carbohydr. Rex, 172 (1988) 183- 193. 173. H. Iijima and T. Ogawa, Carbohydr. Res., 186 (1989) 95- 106. 174. S.4. Hakomori, Annu. Rev. Biochem., 50 (1981) 733-764. 175. B. Wegmann and R. R. Schmidt, Carbohydr. Res., 184 (1988) 254-261. 176. R. R. Schmidt and A. Toepfer, Tetrahedron Lett., 32 (1991) 3353-3356. 177. R. Windmiiller, Dissertation, University of Constance, 1991. 178. I. Kitagawa, N. I. Baek, K. Ohashi, M. Sakagami, M. Yoshikawa, and H. Shibuya, Chem.

179. T. M. Slaghek, A. H. van Oijen, A. A. M. Maas, J. P. Kamerling, and J. F. G. Vliegenthart,

180. T. M. Slaghek, A. A. M. Maas, J. P. Kamerling, and J. F. G. Vliegenthart, Carbohydr. Rex,

18 1. G. 0. Aspinall, N. K. Khare, R. K. Sood, D. Chattejee, B. avoire, and P. J. Brennan,

182. L. Minale, C. Pizza, R. Riccio, and F. Zollo, Pure Appl. Chem., 54 (1982) 1935- 1950. 183. X.-B. Han, Dissertation, University of Constance, 1992; X.-B. Han and R. R. Schmidt,

184. K. Tatsuta, K. Fujimoto, M. Kinoshita, and S. Umezawa, Carbohydr. Rex, 54 (1977)

185. J. Thiem, H. Karl, and J. Schwentner, Synthesis (1978) 696-698. 186. G. Jaurand, J.-M. Beau, andP. Sinay, J. Chem. Soc. Chem. Commun. (1981)572-573. 187. K. Bock, I. Lundt, and C. Pedersen, Carbohydr. Res., 130 (1984) 125-134. 188. K. C. Nicolaou, T. Ladduwahetty, J. L. Randall, and A. Chucholowski, J. Am. Chem.

189. Y. It0 and T. Ogawa, Tetrahedron Lett., 28 (1987) 2723-2726. 190. R. Preuss and R. R. Schmidt, Synthesis (1988) 694-697. 191. G. J. Dutton, Glucuronic Acid: Free and Combined. Academic Press, New York, 1966. 192. R. R. Schmidt and G. Grundler, Synthesis(1981) 885-887. 193. G. Grundler, Dissertation, University of Constance, 1983. 194. S. Friedrich-Bochnitschek, H. Waldmann, and H. Kunz, J. Org. Chem., 54 (1989) 75 1 -

195. M. Masato, Y. Ito, and T. Ogawa, Carbohydr. Res., 195 (1990) 199-224. 196. H. Kunz and H. Waldmann, Angew. Chem., 90 (1984) 49-50; Angew. Chem. Znt. Ed.

197. R. R. Schmidt, M. Faas, and K.-H. Jung, Liebigs Ann. Chem. (1985) 1546- 1556. 198. R. R. Schmidt, W. Guilliard, D. Heermann, and M. H o h a n n , J. Heterocycl. Chem., 20

199. R. R. Schmidt and M. H o h a n n , Tetrahedron Lett., 23 (1982) 409-412. 200. M. Hoffmann, Diploma thesis, University of Constance, 198 1. 20 1. D. Horton and D. Hutson, Adv. Carbohydr. Chem. Biochem., 18 (1 963) 123 - 199. 202. E. L. Eliel and E. H. Juaristi, ACS Symp. Ser., 87 (1979) 95- 106. 203. A. J. Kirby, The Anomeric Effect and Related Stereoelectronic Efects at Oxygen, p. 10.

204. W. Frick and R. R. Schmidt, Liebigs Ann. Chem. (1989) 565-570, and references cited

Pharm. Bull, 37(1989) 1131-1133.

Carbohydr. Res., 207 (1990) 237-248.

211 (1991) 25-39.

Carbohydr. Rex, 216 (1991) 357-373.

Liebigs Ann. Chem. (1992) 817-823.

85-104.

SOC., 108 (1986) 2466-2467.

756.

Engl. (1984), 71.

(1983) 1447- 1451.

Springer, Berlin, 1983.

therein.

Page 146: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

ANOMERIC-OXYGEN ACTIVATION FOR GLYCOSIDE SYNTHESIS 123

205. R. R. Schmidt andM. Hoffmann,Angew. Chem., 95 (1983)417-418;Angew. Chem. Znt. Ed. Engl., 22 (1983) 406-407; Angew. Chem. Suppl. (1983) 543-544.

206. M. Hoffmann and R. R. Schmidt, Liebigs Ann. Chem. (1985) 2403-2419. 207. R. R. Schmidt and G. Effenberger, Curbohydr. Rex, 171 (1987) 59-79. 208. R. R. Schmidt and G. Effenberger, Liebigs Ann. Chem. (1987) 825-831. 209. H. Paulsen and J. Thiem, Chem. Ber., 106 (1973) 3850-3876. 210. R. Meuwly and A. Vasella, Helv. Chim. Actu, 68 (1985) 997- 1009. 2 1 1. K. Briner and A. Vasella, Helv. Chim. Actu, 70 ( 1987) 134 1 - 1356. 212. J. R. Pougny and P. Sinay, Tetrahedron Lett. (1976) 4073-4076. 2 13. J. R. Pougny, J. C. Jacquinet, M. Nassr, M. L. Milat, and P. Sinay, J. Am. Chem. Soc., 99

214. M. J. Ford and S. V. Ley, Synlett (1990) 255-256, and references cited therein. 215. B. Fraser-Reid, P. Konradsson, D. R. Mootoo, and R. E. Devitt, J. Chem. Soc., Chem.

216. M. L. Sinnott in M. I. Page and A. Williams (Eds.), EnzymeMechanisms, pp. 259-297.

217. M. Inage, H. Chaki, S. Kusumoto, and T. Shiba, Chem. Lett. (1982) 1281-1284, and

218. F. L. Westheimer, Science, 235 (1987) 1173-1178. 219. S. Hashimoto, T. Honda, and S. Ikegami, J. Chem. SOC. Chem. Commun. (1989) 685-

220. S. Hashimoto, T. Honda, and S. Ikegami, Chem. Phurm. Bull., 38 (1990) 2323-2325. 221. S. Hashimoto, T. Honda, and S. Ikegami, Tefrehedron Lett., 31 (1990) 4769-5772. 222. S. Hashimoto, T. Honda, and S. Ikegami, Heterocycles, 30 (1990) 775-778. 223. S. Hashimoto, T. Honda, and S. Ikegami, Tetrahedron Lett., 32 (1991) 1653- 1654.

(1977) 6762-6763.

Commun. (1990) 270-272.

The Royal Society of Chemistry, London, 1987.

references therein.

687.

Page 147: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

This Page Intentionally Left Blank

Page 148: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

ADVANCES IN CARBOHYDRATE CHEMISTRY AND BIOCHEMISTRY, VOL. 50

SYNTHETIC REACTIONS OF ALDONOLAmONES

BY ROSA M. DE LEDERKREMER AND OSCAR VARELA

Departamento de Quimica Orgcinica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, 1428 Buenos Aires, Argentina

I. INTRODUCTION Aldonolactones are useful starting materials for the synthesis of modified

sugars. They have also been used as chiral templates in synthesis of natural products. Some of them are inexpensive, commercially available products or they may be obtained readily from the respective monosaccharides. The purpose of this chapter is to survey the main reactions of aldonolactones. Previous reviews on the subject include articles on gulono- 1 ,Clactones (1) and D-ribonolactone (2). Methods of synthesis, conformational analysis, and biological properties are not discussed in this chapter.

11. ACETALATION AND ALDONOLACTONES

1. Reaction with Aldehydes

The hydroxyl groups of aldonolactones react with a variety of aldehydes and ketones to give the corresponding acetal derivatives. Treatment of the salts of aldonic acids with benzaldehyde and hydrochloric acid or zinc chlo- ride as catalysts give benzylidene derivatives of aldonic acids or aldonolac- tones (3).

The structures originally proposed (3) for the benzylidene derivatives of D-ribonolactone (1) have been revised (4). Reexamination of the 'H- and I3C-nuclear magnetic resonance (n.m.r.) data for the product of benzyliden- ation of 1 with benzaldehyde and hydrochloric acid, described (3,5,6) as 3,5-O-benzylidene-~-ribono- I ,4-lactone (2), and its acetylated and benzoy- lated derivatives, led to the conclusion (4) that these compounds had a 1,3-dioxolane ring (3a- c) rather than a 1,3-dioxane ring. To corroborate the assignments, the structure of 3c was determined by X-ray crystallography, which showed a central 1,5-1actone ring, in a boat conformation, cis-fused to a 1,3-dioxolane ring, having the R-configuration at the acetal carbon atom.

Copyright 0 1994 by Academic Press, Inc. AU rights of reproduction in any form memed. 125

Page 149: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

126 ROSA M. DE LEDERKREMER AND OSCAR VARELA

Reaction of 1 with benzaldehyde and zinc chloride gave a diastereomeric mixture (6) of R- and 92,3-0-benzylidene derivatives (4a and 4b). The former (4a) would be identical to the acetal described by Zinner et a!, (3) as “2,4-0-benzylidene-~-ribono- 1,5-lactone.” This structure was further es- tablished as 4a, by chemical and physical studies of the product obtained on reaction of 1 with benzaldehyde dimethyl acetal(7). The 2,3-0-benzylidene derivative, obtained by Garegg et al. (8) on reaction of 1 with &,a- dichlorotoluene in pyridine, has the same properties as compound 4a, which would indicate the R-configuration for the acetal carbon.

1 R = H 2 R=PhCH

3 a R = H 4 a R1 = H, R2 = Ph b R = A c b R1 = Ph. R 2 = H c R = B z

The physical constants of the reassigned structures of benzylidene deriva- tives of D-ribonolactone are shown in Table I.

Selective acetalation of gulono, galactono, idono, or talono- 1 ,Qlactones gave intermediates having a free hydroxyl group at either the 2- or 3-position, which were used in syntheses of ascorbic acid (9). Reaction of L-gulono- 1,4- lactone with benzaldehyde, catalyzed by concentrated hydrochloric acid (10) or hydrogen chloride (1 1 ), gave 3,5-0-benzylidene-~-gulono-1,4-lac- tone (5). Analogous derivatives of 5 with aromatic or aliphatic aldehydes have been reported (9). Treatment of L-gulono- 1 ,Clactone with benzalde- hyde diethyl acetal and concentrated hydrochloric acid gave ethyl 3,5 : 4,6- di-0-benzylidene-L-gulonate (6) in 84% yield (1 I).

Ph

CHzOH

HCOCHPh I

Ph

5 6

Page 150: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

SYNTHETIC REACTIONS OF ALDONOLACTONES 127

TABLE I Physical Constants for the Reassigned Structures of 0-Benzylidene

Derivatives of D-Ribonolactone (1)

Compound Melting point ("C) [aIn (solvent) Reference

3a 233-235.5 - 174.1" (HCONMe,) 3 230-231.5 - 177" (HCONMe,) 6

4a 163-164 -70" (HCONH,) 7 164- 166 - 85" (CHCl ,) 8

4b 87 - 88 - 40" (CHCl, ) 7

2. Reaction with Ketones

The hydroxyl groups of aldonolactones react with ketones to afford cyclic acetal derivatives. Thus, treatment of D-gulono- 1,4-1actone with acetone and such acid catalysts (1 2,13) as hydrochloric acid, sulfuric acid or zinc chloride gave 2,3 : 5,6-di-O-isopropylidene-~-gulono- 1 ,Clactone (7a). Se- lective hydrolysis of the di-U-isopropylidene derivative 7a gave the 2,3-0- isopropylidene derivative (7b) in satisfactory yields ( 12,13). Using copper sulfate as catalyst (12), the diacetal 7a (25% yield) was obtained, together with 5,6-U-isopropylidene-~-gulono- 1 ,Clactone (8a, 10% yield).

Selective isopropylidenation of aldonolactones has been achieved. For example, the 5,6-U-isopropylidene derivative 8a was the only product formed (95% yield) on treatment of D-gulono- 1 ,Clactone with 2-methoxy- propene ( 14). Similarly, D-galactono- 1 ,Clactone gave, under the same con- ditions, the expected monoisopropylidene derivative (8b) in 95% yield. Re- action of L-gulono- 1 ,Clactone with 2,2-dimethoxypropane - acetone, in the presence of tin( 11) chloride, produced the 5,6-O-isopropylidene derivative 9a in 60% yield. Similarly, ~-mannono-l,4-lactone gave 10a in 77% yield ( 15). The di-U-isopropylidene derivatives of ~-gulono-( 16) (9b, wrongly de- picted in Ref. 16) and D-mannono- 1 ,Clactones (1 7,18) (lob) have been reported. Acetonation of D-g2ycero-D-gulo-heptOnO- 1 ,Clactone with acetone-sulfuric acid yields - 80% of the 3,5 : 6,7-di-O-isopropylidene derivative (11) with a small proportion of the 2,3 : 5,6-diacetal(l9). Likewise 2,3 : 5,6-di-U-isopropylidene-~-g~ycero-~-gu~o-heptono- 1 ,Clactone (12) has been prepared (20).

Reaction of D-glucono- 1 ,4-lactone with 2,2-dimethoxypropane - tin( 11) chloride yields the 5,6-U-isopropylidene derivative 13, which on periodate oxidation afforded 2,3-U-isopropylidene-~-glyceraldehyde (2 1). However, the acid-catalyzed isopropylidenation of D-glucono- 1,5-lactone with 2,2-di- methoxypropane afforded methyl 3,4 : 5,6-di-U-isopropylidene-~-gluco- nate (14) as the main product (22). Reduction of the ester function gave

Page 151: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

128 ROSA M. DE LEDERKREMER AND OSCAR VARELA

HCOR I CH,OR

7 a R=CMe,

b R = H

1 0 a R = H b R=CMe,

8 a R’ =OH, R2 = H b R1 = H. R2 = OH

HCO-CMe, I

9 a R = H

b R=CMe,

HCO, f? HiO/CMe2

I ~H,OH

11 12

3,4 : 5,6-di-O-isopropylidene-~-glucitol, which was further oxidized to 2,3 : 4,5-di-O-isopropylidene-aldehydo-~-arabinose.

OCH,

OCH Me2.< I

Q0

OH

13

C0,Me I

I HCOH

OCH W / C M e 2

HCO

14

D-Ghcono- I ,5-lactone has been transformed (23) into trihydroxycarbox- ylates bearing a long-chain alkyl acetal group (15a,b). The synthesis involves the acetalation of HO-4 and HO-6 with a long-chain alkyl carbonyl com-

Page 152: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

SYNTHETIC REACTIONS OF ALDONOLACTONES 129

pound, followed by alkaline hydrolysis of the lactone group. These com- pounds may be utilized as a new type of cleavable surfactants.

C0,Na I I I

I \ I /

HCOH

HOCH

HCO

HCOH CR1R2

H,CO

15 a R1 = Me, R2 = (CHJn CH, [n = 7, 8. l o ] b R1 = H, R2 = (CH& CH,

D-Ribono- 1 ,Clactone (1) readily condenses with acetone, under acidic catalysis with mineral acids or anhydrous copper sulfate, to give 2,3-0- isopropylidene-D-ribono- 1 ,Clactone (16a), which was employed for the synthesis of 5-deoxy and 5-0-substituted derivatives of D-ribono- 1 +lac- tone and D-nbitol(24). Acid removal of the 1,3-dioxolane protecting group gave products having probable inhibitory activity of arabinose 5-phosphate isomerase (25). Other applications of 16a for the synthesis of natural prod- ucts will be discussed later.

Treatment of 1 with p-methoxyacetophenone dimethyl acetal or a,p-dimethoxystyrene, in the presence of pyridinium p-toluenesulfonate, gave the 2,3-acetal derivative. Although a new chiral center is generated in the product, a preference for a major isomer was observed (26).

Reaction of D-ribono- I ,blactone with cyclohexanone catalyzed by Am- berlite IR- 120 (H+) in refluxing benzene led to two products identified by their spectral data and chemical transformations as 2,3-0-cyclohexylidene- D-ribono- 1,4-lactone (16b) and 3,4-O-cyclohexylidene-~-ribono- 1 $lac- tone (17), obtained from the reaction mixture in 60 and 20% yields, respec- tively (27).

HOCH, 0

Q RO OR

16 a R=>CMe2

b R = >C6H,,

17

Page 153: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

130 ROSA M. DE LEDERKREMER AND OSCAR VARELA

The chirality of compound 16b was completely inverted, to give 2,3-0- cyclohexylidene-L-ribonolactone (19), by means of a procedure (28) that involves oxidation of the hydroxymethyl group of 16b by ruthenium te- traoxide, followed by reduction of the lactone group. The resulting interme- diate 1 $lactone (18) underwent isomerization (with cyclohexylidene mi- gration) upon refluxing in xylene, in the presence of a catalytic amount of D-camphorsulfonic acid [in Ref. (28) the formulas for the series were errone- ously depicted].

16 b -

O-Q HO O/C6H10 A

i a 19

3. Use of Aldonolactones Acetals for the Synthesis of Sugar Derivatives

Acetal derivatives of aldonolactones are useful starting compounds for the synthesis of lower aldoses and different sugar-derived molecules. Many syn- theses of D- and L-erythrose from acetone derivatives of ribono- and gulono- lactones have been reported. For example (27), borohydride reduction of 16b followed by periodate degradation of the glycol and subsequent benzoy- lation gave l-0-benzoyl-2,3-di-O-cyclohexylidene-~-~-erythrofuranose. The same sequence of reactions applied to compound 17 gave the corre- sponding derivative of L-erythrose.

Lerner (29) reported a simple synthesis of L-erythrose that involves 2,3-di- 0-isopropylidene-D-gulono- 1 ,Clactone (7b) as a key intermediate. Reduc- tion of the lactone group of 7b with sodium borohydride, followed by periodate oxidation of the L-glucitol derivative, afforded 2,3-0-isopropy- lidene-L-erythrose. The free sugar may be readily obtained by acidic hydrol- ysis of the latter.

The borohydride reduction - periodate cleavage applied to 2,3-0-isopro- pylidene-D-ribono- 1 ,Clactone (16a) led to L-erythrose (30). The method was also employed (31) for the synthesis of D-erythrose, starting from an 0-benzylidene-D-ribonolactone. However, in this case, the structural as- signments for the intermediate compounds must be revised, as the starting material formulated as “3,5-0-benzylidene-~-ribono- 1 ,Clactone (2)” was, as discussed previously in this section, the 3,4-0-benzylidene-~-ribono- 1 3 - lactone (3a). Therefore, the correct structure for the product described as 3,5-0-benzylidene-~-nbitol(20, not isolated) would be 3,4-0-benzylidene-

Page 154: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

SYNTHETIC REACTIONS OF ALDONOLACTONES 131

D-ribitol (21), and the structure formulated as 2,4-O-benzylidene-~-er- ythrose (22), would be 2,3-O-benzylidene-~-erythrose (23). These reassign- ments are supported by comparison of the properties of the product described as 22 with data from the literature. Thus, an authentic sample of 22, obtained by a different route (32), had an optical rotation value of - 20°, which greatly differs from that found for the product formulated (31) as 22 (-65.2" + - 62.6'). The fact that mutarotation is observed, as well as the correspondence with the [a], value (-62") for 23, would indicate that the latter is the correct structure for the product described as 22. In any event, hydrolysis of the acetal function of both (22 and 23), leads to D-erythrose.

CH,OH CHzOH

HCO

HCO

I I

I

HCOH

HCO,

I I

I /

HCOH

HCO

I \ HAO/CHPh HCOH CHPh HCOH CHPh

I I \ I /

H,CO CH,OH H,CO

20 21 22 23

Sodium 2,3-O-isopropylidene-~-nbonate (24), obtained from 16a, was selectively cleaved between C-4 and C-5 by sodium periodate and the result- ing aldehyde group was reduced by sodium borohydride to give D-er- ythrono- 1 ,Clactone (25a) in 76% yield, with loss of the 0-isopropylidene group occurring during lactonization. New derivatives of 25a, including the 2,3-O-isopropylidene derivative (25b), were reported (33).

C0,Na

24 25 a R = H b R=CMe,

Periodate oxidation of 5,6-O-isopropylidene-~-gulono- 1 ,Clactone (9a) gave 2,3-O-isopropylidene-~-glyceraldehyde in 69% yield. This compound was used to prepare 2,3-O-isopropylidene-~-glycerol and it was also con- densed with amines and Wittig reagents (34).

Page 155: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

132 ROSA M. DE LEDERKREMER AND OSCAR VARELA

The synthesis of L-ribofuranose derivatives from 16a has been carried out by Walker and Hogenkamp (35). The procedure involves oxidation of the hydroxymethyl group of 16a with dimethyl sulfoxide -N,N’- dicyclohexylcarbodiimide and acid hydrolysis of the protecting group to give L-riburonic acid, which was converted into methyl (methyl a,P-ribofurano- sid)uronates (26). Reduction of 26 with sodium bis(2-methoxyethoxy)alu- minum hydride gave the chromatographically separable anomers of methyl L-ribofuranoside (27).

C0,Me CH,OH

26 27

Reactions of acetal derivatives of aldonolactones involving the lactone carbonyl group or used as chiral precursors in the synthesis of noncarbohy- drate natural products are discussed in later sections.

111. ACYLATION AND ETHERIFICATION OF ALDONOLACTONES

As acylation of aldonolactones is related to /%elimination reactions, the fully acylated aldonolactone derivatives are mentioned in Section IX (see also Sections VI and VIII). Only a few studies on selective acylation of aldonolactones have been described. The partially acylated derivatives have been employed as glycosylating agents or for the preparation of monometh- ylated sugars.

Partial benzoylation (36) of D-mannono or L-rhamnono- 1,4-1actone under controlled conditions gave the 3-unsubstituted derivatives 28a and 29a. Attempted methylation (37) of HO-3 with diazomethane- boron tri- fluoride etherate gave a very low yield of the 3-O-methyl derivatives (28b and 29b). Methylation by Purdie’s method caused benzoyl migration, and the 2-O-methyl fully benzoylated derivatives (28c and 29c) were obtained in good yield. Reduction of the lactone carbonyl group, followed by debenzoy- lation, afforded 2-O-methyl-~-mannose and 2-O-rnethyl-~-rhamnose. Ben- zoylation of D-galactono- 1 ,Clactone with 2.2 mol of benzoyl chloride at - 23 ’ gave crystalline, 2,6-di-O-benzoyl-~-galactono- 1 ,Clactone (30a) in 62% yield (38).

A partially acetylated galactonolactone, namely, 2,3,6-tri-O-acetyl-~-ga- lactono- 1 ,Clactone (30b), was obtained by an indirect method. Tritylation and further acetylation of D-galactono- 1,4-1actone gave 2,3,5-tri-O-acetyl-

Page 156: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

SYNTHETIC REACTIONS OF ALDONOLACTONES 133

6-0-trityl-~-gdactono- 1 ,Clactone (30c). Detritylation of 30c with F,B - OEt, caused 0-5 + 0-6 acetyl migration, affording 30b, which was used for the preparation of 5-O-methyl-~-galactofuranose (39).

I I CH20R4 CH3

28 a R1 = Bz, R2= H 29 a R’ = Bz, R2 = H 30 a Rf = R4 = Bz, R2 = R3 = H

b R’ = R2 = R4 = Ac, R3 E H c R1 = Me, R2 = Bz c R1 = Me, R2 = Bz c RT = R2 = R3 = Ac, R4 I Tr

R1 = Bz, R2 = Me b R1 = Bz, R2 = Me b

Aldonolactones having their hydroxyl groups protected as ethers (mainly benzylated and silylated derivatives) have been employed for chain elonga- tion through the carbonyl group, and therefore these type of derivatives are included in Section V.

Selective silylation of aldohexono- 1 ,Clactones with tert-butylchlorodi- methylsilane gave the 2,6-di-O-silylated derivatives as major products (40). However, D-glucono- 1,5-lactone underwent ring-contraction during silyla- tion to give 2,6-di-O-tert-butyldimethylsilyl-~-glucono-l,4-lactone (31a) together with its 5,6-di-O-silyl isomer (31b), in 65 and 18% yields, respec- tively.

Acylation and substitution reactions on the selectively silylated aldono- lactone derivatives have been performed.

FH20SiMe2But

I OR’

31 a R1 = SiMe,But, R2 = H

b R’ = H. R2 = SiMe2But

The 7-0-silyl and 2,7-di-O-silyl derivatives (55 and 18% yields, respec- tively) were obtained on treatment Of D-glycero-D-gulo-heptOnO- 1 ,blactone with 1.1 eq. of tert-butylchlorodiphenylsilane (20). Sugar lactone derivatives selectively substituted with different protecting groups, which were em-

Page 157: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

134 ROSA M. DE LEDERKREMER AND OSCAR VARELA

ployed as starting compounds for the synthesis of carbohydrate and non- carbohydrate natural products, are mentioned in later sections.

Iv. REACTION OF ALDONOLACTONES WITH HYDROGEN BROMIDE The reaction of aldonolactones with hydrogen bromide in acetic acid has

been extensively studied by Pedersen and coworkers. Treatment of D-galac- tono-1,blactone with HBr-AcOH for a few hours at room temperature followed by acetylation gave the acetylated 6-bromo-6-deoxy derivative 32 in good yield (4 1). On the other hand, reaction of D-mannono- 1 ,Clactone with the same reagents for 2 - 3 h gave a high yield of the dibromolactone 33a. D-Glucono- 1,4- or 1 $lactones both yielded 2,6-dibromo-2,6-dideoxy- ~-mannono-l,4-lactone (33b) when treated for 18 h with hydrogen bromide - acetic acid, followed by acetylation. Substitution by bromide at C-2 takes place with inversion of configuration. Thus, 2-bromo-2,6-di- deoxy-L-glucono- 1 ,Clactone (34) was obtained from L-rhamnono- 1,4-lac- tone (42).

CH2Br I fq A c o c i q p

OH HOCH

HCOAc OAc R’ I CH,Br

32 33 a R’ = Br, R2 = H b R’ = H, R2 = Br

I CH,

34

The authors postulated that substitution of the primary hydroxyl group by .bromide takes place through a 5,6-acetoxonium ion. Consequently, the reaction of pentono- 1 ,Clactones with HBr - AcOH for a few hours yielded the 2-bromo-2-deoxypentono- 1,4 lactones, whereas the 2,5-dibromo deriv- atives were formed only on prolonged treatment (43). In the case of ~ - r i - bono- I ,Clactone, a mixture of 2,5-dibromolactones was obtained. Substitu- tion by bromine at C-5 could take place via the 4,5-acetoxonium ion derived from the aldonic acid formed on opening of the lactone ring, thus explaining the formation of 2,5-dibromo-2,5-dideoxy-~-xylono- 1 ,Clactone together with the analogous D-arabinono- and D-ribono-dibromolactones.

In the heptonolactone series, D-g/ycero-D-gu/o-heptono- 1,4-lactone reacted with hydrogen bromide -acetic acid to afford (44) 2,7-dibromo-2,7- dideoxy-D-glycero-D-ido-heptono- 1 ,Clactone (35), together with two anhy-

Page 158: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

SYNTHETIC REACTIONS OF ALDONOLACTONES 135

drolactones. The latter, assumed to be 3,6 anhydro-L-glycero-D-gulo-hep- tono- 1 ,4-lactone (36) and 3,7-anhydro-~-glycero-~-gdo-heptono- I ,4-lac- tone (37), were not formed via the bromolactones; instead they were obtained on treatment of D-gl~Cero-D-gulo-heptOnOlaCtOne with one equiv- alent of formic or acetic acid in anhydrous hydrogen fluoride, with subse- quent deacylation. When an aqueous solution of the dibromolactone 35 was boiled, it gave 2,5 : 4,7-dianhydro-~-glycero-~-gub-heptonic acid.

0 0 po poH It II

HO

OH OH OH

HCOH HOCH, I

H ~ O H I

CH,Bi

35 36 37

The bromodeoxyaldonolactones have been used for the preparation of aminodeoxy aldonic acids and aminodeoxy sugars via azido derivatives (45,46). Likewise, a- and P-aminopolyhydroxy acids have been prepared by treatment of the bromodeoxyaldonolactones with liquid ammonia (47). Thus, 3-amino-3-deoxy-~-threonic acid and 3-amino-3-deoxy-~-arabin- onic acid (40b) were obtained from 2-bromo-2-deoxy-~-threono- or D-XY- lono- 1 ,4-lactone (38). It was shown that 2,3-epoxy carboxamides (namely, 39) are intermediates of the reaction. Heating at 90" for long periods led to the 3-amino-3-deoxyaldonamides, which upon acid hydrolysis yielded the corresponding aldonic acids.

COR I I I I

CONH,

Q - - Br CH,OH

HOCH HOCH, 0 I

HCNH,

HCOH

O / T ------ 'CH I I

HCOH

CH,OH

38 39 40 a R = NH2

b R = O H

In the case of the reaction of 2,5-dibromo-2,5-dideoxy-~-xylono- (41a) and D-lyxono- 1,4-lactones (41b) with liquid ammonia, the main product

Page 159: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

136 ROSA M. DE LEDERKREMER AND OSCAR VARELA

was the 2,5-diamino-2,5-dideoxy-~-xylono- 1,5-lactarn 43 formed via a cy- clic 2,3-epoxy-~-lyxonolactam (42), which opens exclusively at C-2. Simi- larly, 2-bromo-2-deoxylactones react with fluoride ions to afford the 2- fluoro derivatives, through a 2,3-epoxylactone intermediate (48).

R‘

41 a R’ = Br R2 = H

b R = H R ’ = B ~ 42

I NH2

43

V, CHAIN ELONGATION THROUGH THE ALDONOLACTONE CARBONYL GROUP

The addition of organometallic reagents to the carbonyl group of conve- niently substituted aldonolactones constitutes a viable chain-extension method. The reaction leads to the formation of hemiacetals of glyculoses, 1 -methylene sugars, and C-glycosyl compounds, which are precursors of, or occur as subunits of, a variety of natural products.

1. Reformatsky-Type Reactions

Carbohydrate lactones have been used as the carbonyl reagent in the Reformatsky reaction. Thus, 2,3 : 5,6-di-O-cyclohexylidene-~-mannono- 1 ,Clactone [44, obtained by oxidation of the mannofuranose derivative (49)] reacted with ethyl bromoacetate and zinc to give the protected 2- deoxy-3-octulosonic acid ethyl ester (454 in 69% yield (50). “Ketonic hy- drolysis” with potassium hydroxide in aqueous methanol, followed by aci- dification and heating, afforded the 1 -deoxyheptulose derivative 45b. Similarly, starting from compound 44, the 1 -C-substituted allyl and propar- gyl lactols were prepared on reaction with allyl or propargyl bromides in the presence of zinc (5 1).

44 45 a R = C0,Et

b R = H

Condensation of 2,3 : 5,6di-O-isopropylidene-~-gulono- 1 ,Clactone (7a) with ethyl bromoacetate in the presence of zinc also gave the expected

Page 160: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

SYNTHETIC REACTIONS OF ALDONOLACTONES 137

Reformatsky product (46). Treatment of 46 with methanol and an acid ion-exchange resin afforded the methyl glycoside 47a, the product of partial hydrolysis (47b), and the 3,8-anhydro sugar 48. Synthetic transformations were performed on these products (52).

CH,CO,Me 7a-v HCO I 'CMe, -

H,CO'

46

+ ;eC02Me ~ B;C02Me

OH HO o\ / o HO

HCOR I

H,COR

47 a R = CMe, 48

b R = H

Improvement to the Reformatsky reactions was achieved (53) by the use of a highly activated zinc- silver couple dispersed on the surface of graphite. Treatment ofprotected aldono- 1,Clactones 10b or 25b and 1,5-lactones 51a or 51b with a Reformatsky reagent prepared from a-haloesters or alkyl 2-(bromomethy1)acrylates resulted in the formation of the corresponding 3- or 4-glyculofuranos (or pyranos)onates 49a,b-50a,b, or 52a,b, respectively, under mild conditions (- 40" to 0" ) and in very good yields. Ethyl 2-deoxy- 2-fluoro (and 2-bromo)-a-~-manno-3,6-furanos-3-octulosonate derivatives were also obtained.

49 a R = CH2C02Et

b R = CH,-C=CH,

C02But I

BnOCH, BnOFH, I

50 a R = CH2C02Et

b R = CH2-C=CH2 I C02Et

51 a R1 = H, R2 = 0Bn b R' = OBn. R2 = H

52 a R' = H, R2 = OBn

b R1 = OBn. R2 = H

Page 161: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

138 ROSA M. DE LEDERKREMER AND OSCAR VARELA

Tetra-n-butylammonium ester enolates, assumed to be formed in the reaction of alkyl trimethylsilyl esters (or nitriles) with fluoride anion, show the same reactivity as the classical Reformatsky reagents. Protected aldono- lactones react with ethyl trimethylsilylacetate, 2-trimethylsilyloxypropan- oate, trimethylsilylacetate, methyl 2-trimethylsilylpropanoate, trimethylsi- lylacetonitrile, or alkyl2-(trimethylsilylmethyl)acrylates, in the presence of catalytic amounts of tetra-n-butylammonium fluoride to afford the chain- elongated alkyl glyc-3-ulosonates or -3-ulononitriles, in high yields (54). For example, reaction of 10b with Me,SiCH,CO,Et gave 49a, in 92% yield. When the reaction was conducted with anhydrous solution of Bu4NF, an a$-mixture of the 3-0-trimethylsilyl derivatives of the glyculosonate was obtained.

2. Reaction with Organomagnesium and Organolithium Reagents

Reactions of sugar lactones with organomagnesium and organolithium reagents have been described. For example, the Grignard reaction (55) of 2,3 : 5,6-di-O-isopropylidene-~-mannono- 1 ,Clactone (lob) with benzylox- ymethylmagnesium chloride afforded 1 -0-benzyl-3,4 : 6,7-di-O-isopropyli- dene-D-manno-heptofuranose, which on hydrogenolysis followed by acid- catalyzed hydrolysis gave D-manno-heptulose.

Addition of lithiated heterocycles to aldonolactones yields carbon-linked nucleosides (56). Thus, the reaction of 2,3 : 5,6-di-O-isopropylidene-~-gu- lono- 1 ,blactone (9b) or 2,3-0-isopropylidene-~-ribono- 1 ,blactone (16a) with various lithiated heterocycles gave gulofuranosyl derivatives 53a - g or ribofuranosyl derivatives 54b,c. Gulonolactols 53a - g and ribonolactols 54b,c were acetylated with acetic anhydride in pyridine to yield their acetyl derivatives. The stereochemistry of compounds 53a-g and 54b,c was dis- cussed in terms of the Cotton effect of circular-dichroism curves of the ring-opened alcohols formed upon reduction by sodium borohydride. The configuration at C-1 of 53g was proved by means of X-ray analysis (57,58).

53 a - g 54 b - c

a R = pyrid-2-yl b R = benzothiazol-2-yl c R = 1-benzylbenzimidazol-2-yl d R = benzirnidazol-2-yl e R = 1-benzylimidazol-2-yI f R = 1-benzylimidazol-5-yI g R = 1, 3-dithian-2-yl

Page 162: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

SYNTHETIC REACTIONS OF ALDONOLACTONES 139

Carbon-linked sugar- heterocycles were also obtained by reaction of the lithiated derivatives obtained from 2-bromopyridine, a-picoline, and ben- zo t hiazole with 4-O-benzyl-2,3 : 6,7-di-O-cyclohexylidene-~-g~ycero-~- gufo-heptono- 1,5-1actone (55). The corresponding D-gfpru-D-gufo-heptO- pyranose-substituted compounds 56a-c were isolated in 35 -43.5% yields. With other heterocycles (for example furan), 1 -disubstituted guloheptitols were obtained (59).

55 56 a R = 2-pyridyl b R = 2-pyridylmethyl c R = 2-benzothiazolyl

The hemiacetals obtained by nucleophilic addition of organometallic re- agents to the carbonyl group of aldonolactones may be reduced to the corre- sponding C-glycosyl compounds. For example, treatment of 2,3,4,6-tetra- 0-benzyl-D-glucono- 1,5-1actone (51a) with allylmagnesium bromide or with lithium ethyl acetate gave the corresponding hemiacetals, which were reduced with triethylsilane and boron trifluoride etherate to furnish stereo- selectively the /K'-glycosyl compounds 57a and 57b, respectively (60). Very good yields in the reaction were also obtained with benzyl derivatives of aldonolactones having the gulucto and malzno (51b) configurations.

The compatibility of the alcohol protecting-groups to the addition of organometallic reagents and to reduction with Et,SiH - BF, has been stud- ied (61). It was found that neither the methoxymethyl (MOM) groups, nor the 0-isopropylidene group was stable to the reduction conditions. How- ever, the benzylated derivative 51 a afforded stereoselectively the B-C-glyco- syl compounds 57c - h in good overall yields. The same sequence of reactions was performed on 0-benzyl-protected arabinono- 1 ,Clactone, providing a route to C-nucleosides of pentofuranosyl sugars (62).

Ethynyl compounds react with sugar lactones to give acetylenic lactols (16,63). Reaction of 2,3-O-isopropylidene-~-~bonolactone (Ma) with lith- ium acetylenic derivatives gave 1-(2-substituted ethynyl)-2,3-0-isopropyli- dene-D-ribofuranoses. Similarly, treatment of 2,3 : 5,6-di-O-isopropylidene- ~-gulono- 1 ,4-lactone (9b) with various lithium acetylenic reagents gave 1 -alkynyl-2,3 : 5,6-di-O-isopropylidene-~-gulofuranoses.

Reduction of ethynyl lactols obtained from 51a with Et,SiH-Lewis acid

Page 163: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

140 ROSA M. DE LEDERKREMER AND OSCAR VARELA

BnOCH,

1) RMgX or RLi

2) Et,SiH-F,B

51 a _____c

57 a - h

a R = CH,CH=CH,

b R=CH,CO,Et

c R = P h d R = 2-fury1 e R = 2-pyridyl f R = ethenyl g R = p-MeOC,H,

h R=mMeOC,H,

gave 1 -(P-D-glucopyranosy1)alkynes (64). These compounds are immediate precursors of 1 -(/?-D-glucopyranosyl) alkanes, of potential utility as new forms of nonionic detergents. Furthermore, the ethynyl derivatives could be selectively converted into the (E) and (Z)- 1 -(&D-glucosyl)ethenes. The se- quence ethylynation -reduction has been employed for the synthesis of a model fragment (58) of ambruticin.

58

Applying the procedure just described, Rouzaud and Sinay (65) described a stereospecific synthesis of a “C-disaccharide” in which a methylene group takes the place of the interunit oxygen atom. Starting from 51a and the dibromoalkene 59, the hemiacetal 60 was obtained. Reduction of 60, fol- lowed by catalytic hydrogenolysis, gave the /?-( 1 - 6)-“C-disaccharide7’ 61.

Likewise, the lactol62, obtained by addition ofmethyllithium to 2,3,5-tri- O-benzyl-D-arabinono- 1 ,Clactone, underwent a self-condensation reaction in the presence of BF3 - OEt, -MeCN, to afford an anomeric mixture of two C-disaccharides 63 and 64 in 93% yield (66).

The stereocontrolled synthesis of a chiral, polyhydroxy 177-dioxa- spiro[ 5.5lundecene from 2,3,4,6-tetra-O-benzyl-~-glucono- 1,5-lactone

Page 164: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

SYNTHETIC REACTIONS OF ALDONOLACTONES 141

OBn I

HC=CBr, CH,OBn

51 a + BnO Q OMe $ BnO Qf OBn OBn

60 59

1) E1,SiH J 2) H,/ Pd

CH,OH CH,-CH, I I I

, I OH OH

61

BnO

BnOCH, yx __

I BnO

62

BnOCH, 0 rn BnO y 2

BnOCH, Q CH2

I BnO

63

I BnO

64

(51a) has been described (67). This bicyclic system having a different pattern of substitution, can be found in a number of biologically important natural products, such as the avermectins and the milbemycins. Reaction of 51a with lithium 1 -trimethylsilyloxy-3-butyne, followed by treatment under mildly acidic conditions, afforded a mixture of anomers of the acetylenic lactol derivative 65, which was readily converted into the spiroacetals 66 and 67.

Page 165: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

142 ROSA M. DE LEDERKREMER AND OSCAR VARELA

1) LiC=C(CH2),0SiMe,

2) H+

51 a

OBn

65

CH,OBn I

CH20Bn I

66 67

The per(trimethylsily1) ether of D-glucono- 1,5-lactone (68) reacted (68) with 2-lithio-l,3-dithiane to give, after removal of the protecting groups, 1 -C-( 1,3-dithian-2-yl)-a-~-glucopyranose (69a) as a single isomer. Depend- ing on the conditions of acetylation of 69a, the tetraacetyl69b or pentaacetyl 69c derivatives were obtained. Desulfurization of 69b with Raney nickel gave the tetraacetate of 1 -deoxy-D-gluco-heptulose (70a), whereas similar desulhration of 69c was accompanied by removal of the tertiary acetoxyl group, providing stereospecific access to the C-p-D-glucosyl compound 70b.

LI

2) H,O, MeOH R20 OR’ AcO

OSiMe, OR2 OAc

68 69 a R’ = R2 = H 70 a R’ = O H

b Rf = H, R2= Ac b R ’ = H

c R’ = R2 = AC

Page 166: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

SYNTHETIC REACTIONS OF ALDONOLACTONES 143

Similar addition of other lithiated thio derivatives to substituted aldonolac- tones has been performed (69). Thus, reaction of tris(methy1thio)methyl- lithium to benzylidenated or isopropylidenated aldonolactones, followed by mercury( 11)-catalyzed desulfuration, led to derivatives of glyc-2-ulosonic acids.

3. Methylenation of Aldonolactones

Carbene-mediated methylenation of aldonolactones provides a direct route to 1-methylene sugars, which may be used as intermediates for the preparation of furanoid or pyranoid C-glycosyl compounds, or chiral pre- cursors for the synthesis of natural products.

The Tebbe reagent [p-chloro-bis(cyclopentadienyl)(dimethylalumin- ium)-p-methylenetitanium] in its pure, crystalline (70,7 1) or crude (72) forms has been employed for the methylenation of aldonolactones. Thus, D-ribono- 1,44actone derivatives 71a and 71 b reacted with Tebbe’s reagent to give (70,7 1) the exemethylene compounds 72a and 72b.

ButR$30CH2

R’O OR’ R’O Q= OR’

71 a R’ = CMe,, R2 = Me Cp = cyclopentadienyl 72 a R1 = CMe,, R2 = Me

b R’ = SiMe,, R2 = Ph b R‘ = SiMe,. R2 = Ph

Similarly, compound 73 was prepared from a persilylated D-galactono- 1,4-1actone precursor in 60 - 80% yield (7 1). However, when unpurified Tebbe’s reagent was employed, 1,4-lactones (for example 71a) gave a mix- ture of 72a and the product (74) of hydration of the double bond. The

iG)% ButMe,SiOCH, we 0 @ H 2

I CMe, CH,OSiMe,

OH RO

HCOSiMe, OSiMe, o\ /o OR

73 74 7 5 a R = B n b R - SiMe,

c R = M e

Page 167: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

144 ROSA M. DE LEDERKREMER AND OSCAR VARELA

proportion of the lactol 74 greatly increases during the chromatographic purification, even when slightly basic eluants were used, and the alkene 72a was isolated in only 6% yield (72). Reaction of Tebbe's reagent with fully benzylated (7 I ,72), silylated (7 I), and methylated (72) D-glucono-1 $lac- tone gave the corresponding 2,6-anhydroheptenitol derivatives 75a - c in high yields.

Another organotitanium reagent employed for the direct methylenation of aldonolactones was dicyclopentadienyldimethyltitanium (73). Reaction of the diacetone derivative of D-mannono-1 ,blactone (lob) with three equivalents of the reagent afforded 85% of the D-manno-hept- 1-enitol diace- tal. Analogous products were also obtained from 0-isopropylidene or 0-benzylidene derivatives of aldonolactones (such as, D-erythrono- 1,4- lactone, D-arabinono- 1 ,Clactone, D-glucono- 1,5-1actone, and D-mannono- 1,Slactone) in yields higher than 85% (except for the erythronolactone-der- ived 1-enitol, obtained in 64% yield). The lactols were by-products of the reaction.

The application of lithiumtrimethylsilyl acetate for the C- 1 elongation of aldonolactones has been examined (73). Although the reagent had been successfully used for the alkenation of lactone carbonyl groups (74), in the case of aldonolactones 10b or 25b only insignificant yields ofthe alkenes, but 30 - 40% of the lactols 49a or 50a, were obtained (73). However, these lactols, alternatively prepared in good yields by a Reformatsky-type reaction (53,54), were readily eliminated to the desired alkenes by simple treatment with methanesulfonyl chloride - triethylamine at 0". Thus, from 49a or 50a separable E,Z mixtures (76a and 76b, or 77a and 77b, respectively) were obtained in good yields (73).

I I o\ / o

CMe,

76 a R1 = H, R2 = C02E1 77 a R1 = H, R2 = C0,Et

b R1 = CO,Et, R2 = H b R1 = CO,Et, R2 = H

I-Methylene sugars are versatile starting compounds for the synthesis of aromatic and hydroxymethyl C-glycosyl compounds (7 1) and of double- ended C-glycosyl compounds (70). The double bond undergoes 1,3-dipolar cycloadditions to give isoxazoline derivatives (71), such as that used as a model for the synthesis of tunicamycin. Also, reaction with iodonium sym-

Page 168: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

SYNTHETIC REACTIONS OF ALDONOLACTONES 145

dicollidine perchlorate gave an easy access to 1-iodoheptuloses or 1 -iodo- methylene derivatives (75).

Dichloromethylenation and difluoromethylenation of aldonolactones have been achieved by concise sequences. Thus, aldonolactone derivatives react with tris(dimethy1amino)phosphine (hexamethylphosphorus tria- mide) and tetrachloromethane [( Me,N),P- CCl,] to give l-dichloromethy- lene sugars, which may be reduced to C-glycosyl compounds and chiral tetrahydrofurans (76). For example, dichloromethylenation of compound 71a gave the dichloroalkene 78, which was cleanly reduced by Raney nickel to the glycosylmethane 79. The same sequence, when applied to the 2,3 : 5,6- di-0-isopropylidene D-allono- 1 ,Clactone (80) led to the C-methyl com- pound 81 in 83% yield.

ButMezSiOCHz 0

Raney Ni CCI2 - Y)

(Me,N),P-CCI, 71 a +

I I o\ / o

CMe,

78 79

80 81

The presence of a dichloromethylene group at the anomeric center of 82 facilitates proton abstraction at C-3 by a strong base (77), affording the 4-deoxyglycos-3-ulose derivative 83. Reduction of the dichloromethylene group by Raney nickel gave a 1-C-methyl derivative with high stereospeci- ficity, which opens the way to a series of 2,5-anhydro- 1 -deoxyalditols. Com- pound 83 was the key intermediate for the synthesis (78) of tosyl L-(+)-epi- muscanne (84a) and tosyl L-(+)muscarine (84b).

Difluoromethylenation (79,80) of aldonolactones may be readily ac- complished by treatment of the lactone derivative with tris(dimethy1- amino)phosphine, dibromodifluoromethane, and zinc in refluxing tetrahy-

Page 169: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

146 ROSA M. DE LEDERKREMER AND OSCAR VARELA

82 83 84 a R1 = OH, R2 = H b R’ = H, R2 = OH

drofuran. The reaction was applied to 1,Clactones having the rib0 (71a), manno (lob), d o (80), and D-glycero-D-gulo configurations, and also to per(trimethylsily1)ated ~-glucono- I, 5-lactone (68), affording moderate yields (56 - 67%) of 1-difluoromethylene derivatives (for example, 85). Cata- lytic hydrogenation of these compounds gave the corresponding difluoro- methyl C-glycosyl derivatives (such as, 86) with excellent diastereofacial selectivity.

ButMe,SiOCH2 ButMe2SiOCH2 pF2 WF2 - H2/ Pd

o\ / o CMe,

(Me,N),P 71 a A

CF2Br, I Zn

o\ / o CMe,

85 86

4. Formylaminomethylenation of Aldonolactones

Sugars linked to amino acids via a carbon-carbon linkage are of interest as this type of structures occur in polyoxins. The formation of such a carbon - carbon bond was achieved by formylaminomethylenation of 2,3 : 5,6-di-U-isopropylidene-~-mannono- 1 ,Clactone (lob) with ethyl iso- cyanoacetate in the presence of potassium hydride (8 1). The E-isomer 87a was the major product of the condensation ( 5 1% yield); the 2-isomer 87b was isolated in 4% yield. Catalytic hydrogenation (Raney nickel) of 87a and 87b gave the N-formyl-mannofuranosylglycinates 88a and 88b, which on hydrolysis of the N-formyl group and deprotection of the sugar moiety led to the mannofuranosyl amino acids 89a and 89b. Selective hydrolysis of the 7,8-U-isopropylidene group of 88a, followed by periodate cleavage of the diol and reduction, gave the D-lyxofuranosyl amino acid 89c. Similar treat- ment on 88b gave 89d.

Page 170: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

SYNTHETIC REACTIONS OF ALDONOLACTONES 147

87 a R1 = CO,Et, R2 = NHCHO

b R1 = NHCHO. R2 = C0,Et

Me R3

89 a R’ = NH,, R2 = H, R3 = CH20H

b R1 = I-, R2 = NH,. R3 = CH,OH

d R1 = H, R2 = NH,. R3 = H

c R? = N H R 2 = R 3 = H

88 a R1 = NHCHO, R 2 = H

b R’ = H. R2 = NHCHO

Under basic conditions the D-erythro-L-gluco-oconate 88a underwent racemization at C-2 and “anomerization” at C-3 to give a mixture of four isomeric products. The configurations at C-2 and C-3 of these isomers were established by chemical and physical methods (82). Furthermore, alcohols and thiols, in the presence of a base, attack the double bond of the oct-2-en- onates (87b) affording isomeric oct-3-ulofuranosidonates.

Formylaminomethylenation of 2 : 3,5 : 6-di-O-isopropylidene-~-allono- 1,Clactone (80) gave the (E)alkene 90 in 59% yield. However, when the reaction was applied to the benzyl derivative of D-glucono- 1,5-lactone (51a),

CO,Et

NHCHO

o\ /o CMe,

C0,Et

HLOB~ I I I I

BnOCH

HCOBn

HCOH

CH20Bn

90 91

Page 171: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

148 ROSA M. DE LEDERKREMER AND OSCAR VARELA

the oxazole 91 (50% yield) was obtained (83). Use of potassium hydride instead of 1 ,5-diazabicyclo[4.3.O]non-5-ene (DBN) in the reaction of 10b or 80 with ethyl isocyanoacetate gave the corresponding sugar oxazole deriva- tives.

VI. REACTION OF ALDONOLACTONES WITH ALCOHOLS 1. Formation of Esters

Aldono- 1,5-1actones and free aldonic acids react with alcohols in the pres- ence of hydrogen chloride to give the corresponding alkyl aldonates (84). The reaction is slower with 1,4-1actones. Because esterification takes place very slowly in the absence of an acidic catalyst, aldonic acids and their lactones may be recrystallized from boiling alcohols without appreciable esterification (85). However, in some instances, alkyl esters are formed under these conditions. For example, essentially pure ethyl L-mannonate was isolated (6.490 yield) from the mother liquors of crystallization L-man- nono- 1 ,Clactone, obtained by Kiliani synthesis from L-arabinose (86). Sim- ilarly, repeated recrystallization from ethanol of crude 2,3,4,6-tetra-O- acetyl-D-glucono- 1,5-lactone afforded the corresponding ethyl gluconate derivative (87).

The tetra-0-benzoyl derivatives of ~-glucono- 1,5- and 1 ,Clactones un- derwent ring opening upon heating with methanol or ethanol at the reflux temperature, or under acid catalysis, to give methyl or ethyl tetra-0- benzoyl-D-ghconates having an unesterified hydroxyl group (88).

Ethanolysis of 2,4-di-0-benzoyl-3,6-dideoxy-~-arabino-hexono- 1,5-1ac- tone (92) afforded ethyl 2,4-di-O-benzoyl-3,6-dideoxy-~-arabino-hexonate (93a). On being kept, compound 93a was slowly converted into 2,5-di-0- benzoyl-3,6-dideoxy-~-arabino-hexono- 1,4-lactone (94). This transforma- tion involves 0-4 + 0-5 benzoyl migration, with formation of ethyl 2,5-di- O-benzoyl-3,6-dideoxy-~-arabino-hexonate (93b) as an intermediate (89).

CO, Et

HCOBz I I y 2 -

R’ OCH,

BzOCH OBz R d H 2

I CH3

OBz I BzoQ - CH3

92 93 a Rf = Bz, R2 = H 94

b R1 = H. R2 = Bz

Page 172: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

SYNTHETIC REACTIONS OF ALDONOLACTONES 149

The acid-catalyzed acetalation of aldonolactones with alkyl acetals of aldehydes or ketones takes place, in some instances, with esterification of the lactone group to give acetal derivatives of alkyl aldonates ( 1 1,22).

2. Formation of Orthoester Derivatives

Addition of alcohols to lactones results in the formation of orthoacid or orthoester derivatives. Thus, reaction of lactone 95a with potassium cyanide in ethanol led to displacement of the tosyl group by cyanide and addition of ethanol to the lactone carbonyl group, to give the orthoacid derivative 95b, which was isolated as its acetate 95c. Mild deacylation of95c led back to 95b, but under more vigorous reaction conditions the open-chain methyl aldon- ate was obtained (90).

CH, R4

R 3 0 L I

95 a R’ = RZ = 0, R3 = H, R4 = Ts b R’ = OEt, R2 = OH, R3 = H. R4 = CN c R’ = OEt, RZ = OAc, R3 = Ac. R4 = CN

Addition of diols to the carbonyl lactone group leads to the formation of cyclic orthoesters. Compounds containing a spiro, cyclic orthoester inter- linkage at the anomeric carbon atom are ofinterest, as this type of structure is found in the oligosaccharide antibiotics orthosomycins (9 I ) (such as ever- ninomicin, flambamycin, and avilamycin).

Reaction of 2,3,4,6-tetra-0-benzyl-~-glucono-1,5-lactone (51a) with epoxides or with diols afforded the corresponding orthoester derivatives (92,93). When two secondary hydroxyl groups were involved, low yields were obtained. The reaction conditions were optimized, and it was shown that condensation of lactone derivatives with bis-0-(trimethylsily1)- 1,2-diols in the presence of trimethylsilyl trifluoromethanesulfonate (Me,SiOTf) as catalyst gave good yields of the spiro, cyclic orthoesters (94). Thus, reaction of 51a with the trimethylsilyl derivatives of ethanediol, and cis- and trans- I ,2-cyclohexanediol gave the corresponding orthoesters in 9 1.7, 89.7, and 63.9% overall yields (95). Furthermore, condensation of 51a with conve-

Page 173: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

150 ROSA M. DE LEDERKREMER AND OSCAR VARELA

niently substituted derivatives of methyl a-D-glucopyranoside and methyl a-D-mannopyranoside gave the glucopyranosylidene derivatives 96 and 97. Compound 96 was isolated as a single isomer in 72% yield, and compound 97 was actually a mixture of isomers, obtained in 44.8 and 26.6% yields.

BnO QQOMe

BnO

OBn

96

CH,OBn I

97

Many other 2,3-O-~-glycopyranosylidene-a-~-mannopyranosides were obtained by the Me,SiOTf-promoted condensation (96) of D-glucono-, D-galactono-, and L-glycevo-D-gluco-heptono- 1,5-lactones, with methyl 2,3- di-O-trimethylsilyl-cu-D mannopyranosides having various substituents at C-4 and C-6. The absolute configuration of the orthoester carbon atom for one of the isomers of 2,3-~-~-glucopyranosylidene-cw-~-mannopyranosides (97), as the fully acetylated derivative, was established by X-ray analysis. The 'H and 13C n.m.r. data for the isomers were also reported (97). The dias- tereoisomer having the higher values for optical rotation and chemical shift for the orthoester carbon had the (S)-absolute configuration. On the basis of this observation, the configurations for various glycopyranosylidene deriva- tives were tentatively assigned. Hydrogenation of the benzyl derivatives afforded the free glycopyranosylydene, which showed antihelmintic activity against Ascaridia galli (98).

Page 174: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

SYNTHETIC REACTIONS OF ALDONOLACTONES 151

VII. REACTION OF ALDONOLACTONES WITH AMMONIA AND RELATED NUCLEOPHILES

1. Reaction of the Lactone Group with Ammonia and Amines

Aldonamides are readily prepared by reaction of lactones with liquid ammonia (86,99, loo), with ammonium hydroxide (101,102), or by bub- bling ammonia gas into alcoholic solutions of the sugar lactones (1 03 - 104). Aldonamides of the tetronic acids are stable in aqueous solution (lOS), but penton- or hexon-amides are hydrolyzed, as shown by the change of the optical rotation of the amide solutions (106). The hydrolysis is catalyzed by acids and bases, and the product was the ammonium salt of the aldonic acid.

Aldonolactones also react with a variety of amines, producing substituted amides. Thus, heating L-gulonolactone with pphenetidines led to N-substi- tuted gulonamides, products having antipyretic and analgesic activities ( 107). Condensation of aldonolactones with alkeneamines (such as, allyla- mine) or unsaturated urea derivatives afforded the alkeneamides and ureides of polyhydroxy aliphatic acids, suitable intermediates for the synthesis of mercurated diuretic compounds ( 108). Gluconolactone and methyl arabin- onate have been condensed with n-decylamine and higher homologues in a quest for synthetic emulsifying agents (109). The arabinonamides were only slightly soluble in water and had no emulsifying properties. The glucona- mides 98 were more soluble in water and were weak emulsifying agents.

98 (n = 11, 15, 17)

The aggregation behavior of the eight diastereomeric N-octylaldohexona- mides of the D series, three of the L-series, and the corresponding racemates has been investigated, mainly by electron microscopy (1 10). The amides were obtained by electrolytic oxidation of the corresponding hexoses to the lactones and amidation with octylamine. Depending on the disposition of the hydroxyl groups and on the chain conformation, different types of aggre- gates were observed.

Lactonic disaccharides (such as lactobiono- 1,5-1actone) reacted with long-chain primary amines to yield model glycolipids of a new type ( 1 1 1,112). For the formation of N-substituted aldonamides from aldonic acids and amines, N-N’-dicylohexylcarbodiimide was employed as the con- densing agent. However, no catalyst was needed in the case of the lactones.

Page 175: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

152 ROSA M. DE LEDERKREMER AND OSCAR VARELA

The synthetic glycolipids (for example 99) were obtained crystalline from ethanol.

CH,OH

~0QocH~ (CHOJL (cH2),w,

OH

99 (n= 7, 9, 11, 13, 15)

Malto-oligosaccharide aldonolactones react with ethylenediamine to give N-(2-aminoethyl)aldonamides ( 1 13 - 1 15), which have been successfully grafted onto carriers via amide linkages. The malto-oligosaccharides were produced by degradation of amylose with alpha amylase. After purification of the oligosaccharides, they were converted into the lactones by hypoiodite or electrolytic oxidation.

The high-pressure aminolysis of a variety of lactones, including uronic acid lactones, has been described (1 16). However, the procedure was not applied to aldonolactones.

Aromatic polyamines react with sugar lactones to give heterocyclic com- pounds having an attached open-chain polyhydroxyalkyl substituent. Thus, treatment of aldonolactones with o-phenylenediamine afforded ( 1 1 7) 2-polyhydroxyalkylimidazoles (100).

Condensation of ~-glucono- 1 $lactone with 4 : 5-diamino-6-diethyla- mino-2-methylpyrimidine afforded the 8-polyhydroxyalkylpurine 101, a C- nucleoside of an open-chain sugar ( 1 18). Some other pyrimidines gave anal- ogous products from either D-glucono- or D-nbono-lactones.

NEt, I

HCOH I I

HCOH

CH,OH

101

Page 176: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

SYNTHETIC REACTIONS OF ALDONOLACTONES 153

Similarly, open chain C-nucleosides of adenine have been prepared by condensing 4,5,6-triaminopyrimidine with aldonic acids (or aldonolac- tones) of various chain-lengths. The amides (102) first formed were ther- mally cyclized, affording the 8-(hydroxyalky1)adenines (103) in rather low yields ( 1 19).

102 103

N-Acyl indoles derived from amides have been employed for the conver- sion of lactones into protected hydroxyacids. Thus, (chloromethy1)alumi- num 2-(2-propenyl)anilide reacts (120) with 1,4- and 1,5-lactones, as for example per-0-tert-butyldimethylsilyl-D-ribono- 1,4-1actone (104), to afford hydroxyamides. After protection of the free hydroxyl group, these amides were converted by ozonolysis into N-acyl indoles, 105, which were readily saponified to the acid 106.

OR' 0

R20 -

OR2 dR' R1O

I I But Me2Si0 OSi Me2 But

104 105 R1 = SiMe2But, 106 R1 = SiMe2But.

R2 = CH,O(CW,OMe Rz = CH,O (CH3,O Me

The procedure was proved to be general for the preparation of protected hydroxy acids from lactones ( 12 1). This apparently trivial process is often difficult to carry out, as the attempted derivatization of y or 8-hydroxyacids frequently results in relactonization rather than hydroxyl protection. The method was applied to several aldonolactones to produce the correspond- ing intermediate hydroxyamides. Protection using [(2-trirnethylsilyl)- ethoxylmethyl chloride, methoxymethyl chloride, tert-butylchlorodimeth- ylsilane, or tert-butylchlorodiphenylsilane followed by ozonolysis gave the protected N-(y- or Ghydroxyacy1)indole derivatives. Mild saponification gave indole and the acetal- or silyl-protected hydroxy acids.

Page 177: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

154 ROSA M. DE LEDERKREMER AND OSCAR VARELA

2. Intramolecular Reaction of Aminolactones to Yield Lactams

Replacement of an hydroxyl group at C-4 or C-5 of an aldonolactone by amino leads to the formation of aldonolactams. For example, Fleet and coworkers (1 22) reported the synthesis of D- and L-mannonolactam from L- and D-gulonolactones, respectively. The synthesis involves the selective pro- tection of the primary hydroxyl group of the isopropylidene derivative 7b and sulfonylation of HO-5 to give 107a. Displacement of the triflate by sodium azide gave the corresponding derivative 107b, with inversion of configuration at C-5. Hydrogenation of the azide function produced an amine, which rearranged spontaneously to the Glactam 108. An identical sequence on L-gulonolactone led to D-mannonolactam (122,123).

CHzOR + I CH,OSi Me,But

107 a R’ = H, RZ = OSO,CF,

b R’ = N,. R 2 = H

108 R = SiMe,Bu‘

In an allegedly “facile synthesis of nojirimycin,” the intermediate nojiri- mycin d-lactam derivatives 109a and 109b were reported (124) to be ob- tained on treatment of 2,3,4,6-tetra-O-benzyl-~-glucono- 1 $lactone (51a) with aqueous ammonia at room temperature, or with benzylamine in re- fluxing toluene, and with catalytic amounts of ion-exchange resin. Hydro- genolysis of 109b with 5% Pd-C in acetic acid gave nojirimycin (110).

CH,OBn CHzOH I I

I I OB n OH

I 0 9 a R = H b R = B n

110

Difficulties were encountered ( 125) in repeating the previously described synthesis of nojirimycin. Thus, when the lactone 51a was treated with ben- zylamine under a number of conditions, the only product isolated was, as

Page 178: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

SYNTHETIC REACTIONS OF ALDONOLACTONES 155

expected, the amide 11 1. An authentic sample of 109b was obtained (126) by synthesis from the idofuranose derivative 112. Hydrogenation of 109b under the conditions reported (1 24) led to partial or complete debenzylation, but there was no evidence for the formation of nojirimycin (110) in the reaction mixture. The synthesis of p-lactams from aldonolactones or their 2,3-unsat- urated derivatives is discussed in Section XII.

CONHBn I I I I I

HCOBn

BnOCH

HCOBn

HCOH

CH,OBn

111 112

3. Reaction of the Lactone Group with Hydrazine and Derivatives

Sugar lactones react readily with hydrazine to give crystalline derivatives useful for isolation and identification (1 27). Thus, addition of hydrazine to a reaction mixture containing an aldonolactone facilitates isolation of the product. The lactone may be regenerated from the hydrazide by treatment with nitrous acid (128). The phenylhydrazides obtained on treatment of aldonolactones with phenylhydrazine are also useful for characterization (1 29,130).

Pentono- and hexono- 1 ,Clactones yielded substituted arylhydrazides on treatment with m- and p-tolyl-, m- and pmethoxyphenyl, p-bromophenyl, and p-ethoxycarbonylphenyl-hydrazines (1 3 1). It was found that the rate of hydrazinolysis depends on the configuration of the aldonolactone, as well as on the aryl substituent on the hydrazine.

A series of aryl- and aroyl-hydrazide derivatives was obtained from D-gly- cero-D-gubheptono- 1 ,Clactone (1 32). The reaction was faster with arylhy- drazines than with aroylhydrazines. The aroylhydrazides may be decom- posed by copper(I1) sulfate or nitrous acid to regenerate the precursor lactone.

D-Glucono- 1,5-1actone reacted with aroylhydrazines (1 33) to give the corresponding 1 -aroyl-2-~-gluconylhydrazides (1 13), which underwent cy- clization upon heating with triethyl orthoformate, affording 5-aryl-2-eth- oxy-3-~-gluconyl-2,3-dihydro-1,3,4-oxadiazoles (1 14).

Other heterocyclic compounds, precursors of C-nucleosides, were pre- pared by condensing D-glucono- 1,5-lactone with 1 -hydrazino-4-phen-

Page 179: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

156 ROSA M. DE LEDERKREMER AND OSCAR VARELA

CONHNHCOR I

I I I I

HCOH

HC(OEt), o\N/N + HOCH

I I I

HCOH

HCOH c=o

HCOH CH20H

HOqH

113 R = A r y l

HCOH I I

HCOH

CHzOH

114 R = A r y l

ylphthalazine ( 1 34). Cyclization of the intermediate hydrazide 115 by auto- dehydration, through the enolic form, gave 3-(~-gluc~-pentitol- 1 -yl)- 5-phenyl- 1,2,4-triazolo[3,4-a]phthalazine (1 16).

NH N-

HCOH

HCOH I I

\ / HCOH

HCOH Ph CH20H I Ph

CH20H

115 116

Aldonhydroximo-lactones (aldonolactone oximes) have been prepared by oxidation of monosaccharide or disaccharide oximes with activated MnO,, Hg(OAc),, or oxygen in the presence of Cu,Cl,-pyridine. The structure of the hydroximo-lactones depends both on the starting sugar and on the method of oxidation. Thus, D-glucose oxime gave the hydroximo- 1 ,Wac-

CH20H FH20H I

OH OH

117 I18

Page 180: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

SYNTHETIC REACTIONS OF ALDONOLACTONES 157

tone 117 by oxidation with MnO,, the 1,4-analog 118 upon oxidation with O2-Cu2Cl2-pyridine, and a mixture of 117 and 118 by treatment with Hg(OAc), . Hydroxyl-free and protected derivatives were prepared, and other approaches for their synthesis have been examined (1 35). Compound 117 is a competitive inhibitor of p-D-glucosidases (1 36).

VIII. REDUCTION OF ALDONOLACTONES 1. Reduction to Aldoses and Alditols

A common reagent for the reduction of lactones to aldoses is sodium amalgam, first described by Emil Fischer ( 137), in the course ofhis studies on the structure proof of aldoses. Fischer applied the cyanohydrin reaction to an aldose, obtaining the next higher pair of epimeric acids, which as lactones were reduced to aldoses with sodium amalgam. [For a current appreciation of the significance of this work. See Ref. (1 37a)l. This reagent was also used for the reduction of the monolactones of aldaric acids to afford the corre- sponding uronic acids (1 38 - 140), which may be further reduced to aldonic acids (141).

The reduction by sodium amalgam is strongly influenced by the pH of the reaction (142). For the optimum pH value (3 -4), the reaction is complete in 10 min. The pH must be carefully adjusted during the first 3 min of reaction, as rapid liberation of base and formation of the sugar take place during this period. At pH 2 - 3, the pH control is difficult because of the high rate of reaction, and at pH 4 - 5 the yields of sugar were 10 - 15% lower than those obtained at the optimum pH. Sulfuric acid, benzoic acid, sodium hydrogen- oxalate, and ion-exchange resins have been employed for maintaining the desired pH of the reaction (143).

Catalytic hydrogenation of 1 3 - and 1 ,4-aldonolactones in the presence of platinic oxide led, depending upon the reaction conditions, to aldoses or alditols (144). The yields are also dependent upon the configuration of the starting lactones. Thus, both lactones of D-mannonic acid were readily re- duced to D-mannose, whereas D-glucono- 1 ,Clactone gave 23.4% of the sugar and 24% of the alditol. Under the same conditions, D-glucono- I $lac- tone gave an 80% yield of D-glucose, with no alditol formation.

Various hydride reagents reduce aldonic acid lactones to aldoses or aldi- tols. For example, sodium borohydride has been widely used. D-glycero-D- gulo-Heptono- 1,4-1actone was reduced to the corresponding heptose by maintaining the pH (3 -4) through addition of sulfuric acid or to the alditol under alkaline conditions ( 145). Several aldonolactones have been reduced to the corresponding alditols with sodium borohydride by keeping the pH acidic during 1 h and then making it alkaline through addition of sodium

Page 181: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

158 ROSA M. DE LEDERKREMER AND OSCAR VARELA

hydroxide (146). The yield of alditol was always higher than 95%. Reduction of 1,4- or 1,5-1actones does not proceed readily in alkaline solutions because the formation of salts of aldonic acids prevent reaction with the reducing agent. Yields were improved by maintaining a low pH through buffering with boric acid, ion-exchange resins, or carbon dioxide (147). For example, under acidic conditions, D-gulono- 1 +lactone was reduced to D-gulose ( 148). Also, Pedersen and coworkers (42,45) have described high-yielding reductions of bromodeoxyaldonolactones with sodium borohydride in the presence of an acidic ion-exchange resin.

The reduction of 4,6-dideoxy-~-ribo-hexono- 1,5-lactone to the corre- sponding aldose with various reagents has been studied (149). For the reduc- tions in aqueous medium, ammonia-borane (NH, - BH,) and sodium borohydride were used, and for tetrahydrofuran solutions, sodium and lith- ium aluminum hydride were employed. The latter selectively gave the a-an- omer of the hexose, in 90% yield. The mixed hydrides AlC1,H and AICIH,, generated from LiAlH, and AlCl,, in molar ratios of 1 : 3 and 1 : 1, respec- tively, proved to be effective for the preparation of D-glucose or D-mannose from their aldonolactones ( 150). For these reductions, the O-tetrahydropyr- any1 derivatives were used, as the lactones themselves were unreactive. In some instances, relatively low yields of aldoses were obtained, as they were rapidly reduced to the alditols.

For the tota! synthesis of certain trideoxy sugars of biological significance [rhodinose (1 5 1) (2,3,6-trideoxy-~-threo-hexose, 120a) and DL-amicetose (1 52) (2,3,6-trideoxy-~~-erythro-hexose, 120b], diisobutylaluminum hy- dride was used in the stage of reduction of the corresponding rhodinonolac- tone (119a) and amicetonolactone (119b). The free trideoxy sugars were obtained as an anomeric mixture of furanoses and pyranoses, in 66 and 67% yields, respectively.

R3 FH3 CH3 I

Diisobutylaluminum hydride was also employed (1 53) for the reduction of 2,6-dideoxy-3-C-methyl-~-arubino-hexono- 1 ,Clactone (121) to an anti- biotic component, the branched-chain sugar evermicose (2,6-dideoxy-3-C- methyl-D-arubino-hexose, 122). The L-enantiomer of 122 (olivomycose, 125a), L-amicetose 125b, and 2,6-dideoxy-3-C-methyl-~-ribo-hexose (L-

Page 182: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

SYNTHETIC REACTIONS OF ALDONOLACTONES 159

mycarose, 12%) were also synthesized by a route that involves the diisobuty- laluminum hydride reduction of the corresponding lactones 123a, 123b, and 124, obtained from noncarbohydrate precursors (1 54).

121 122

@= HOCH

125 a R1 =OH, R2 = CH,

c R1 = CH,, R2 = OH

b R I = R Z = H

124

Borane has been used for the reduction of aldonolactones (1 50). The conversion of a lactone into the aldose seems to be rapid, and subsequent reduction to the alditol is slower. Employing a 10-fold excess of M diborane in tetrahydrofuran afforded isolated yields of D - ~ ~ U C O S ~ and D-mannose of 80 - 85 and 60 - 65%, respectively, from the corresponding lactones. Brown and Bigley (1 55) described the use of borane derivatives, namely diakylbor- anes, for the reduction of lactones to lactols. These reagents have proved highly effective for converting 0-acylated aldonolactones into the corre- sponding 0-acyl aldoses having HO- 1 free. Thus, 2,3,5,6-tetra-0-benzoyl- D-gulono- 1 ,Clactone was reduced with bis(3-methyl-2-butyl)borane (di- isoamylborane, disiamylborane) to 2,3,5,6-tetra-0-benzoyl-~-gulofuranose

Page 183: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

160 ROSA M. DE LEDERKREMER AND OSCAR VARELA

in 97% yield (1 56). The reaction was also applied to the tetra-0-benzoyl-al- dono- 1 ,Clactones of the ~-gulo, D-&, D - ~ u ~ o , and ~ - ~ l t r o configurations and to D-galactono- 1,4-1actone tetraacetate, furnishing the corresponding acylated tetraacylhexofuranoses in very high yields ( 1 57). Hydroboration reactions proceed slowly in most organic solvents, but they are catalyzed by ethers. The reaction may therefore be performed in ethyl ether, tetrahydro- furan, 2-methoxyethyl ether, and the like. Tetrahydrofuran is usually the solvent of choice because it readily dissolves the diakylborane and the per- 0-acyllactone. The reduction is complete after 14 - 20 h of stirring at room temperature and constitutes a general procedure for the preparation of fur- anose derivatives. Acylated aldono- l $lactones were also effectively re- duced by diisoamylborane (1 58). This reagent has been widely used in our laboratory for the synthesis of deoxy sugars from deoxyaldonolactones, as described in Section X. Dijong and coworkers (159,160) reported acetyl migration and ring expansion during the diisoamylborane reduction of ace- tylated aldono- 1 ,blactones. According to these authors, ring enlargement takes place only when the aglycon and the side chain are cis oriented in the intermediate 0-dialkylboryl furanoside and when the acetoxy groups at C-4 and C-5 have a threo-configuration. Thus, 5-0-acetyl-~-rhodinono- 1,4-lac- tone underwent ring expansion during reduction, whereas 5-0-acetyl-~- amicetono- 1,4-1actone did not. Furthermore, these authors could not repro- duce the yield reported (1 57) (83Yo) for the diisoamylborane reduction of 2,3,5,6-tetra-O-acetyl-~-galactono- 1 ,Clactone, and the highest yield ob- tained for the galactofuranose derivative was 14%, even under optimized conditions. Reduction of tetra-0-acetyl-D-glucono- 1 ,4-lactone led to de- composition products. However, in our hands, both the acetylated (16 1) and the benzoylated ( 162) derivatives of D-galactono- 1 ,Clactone gave high yields of furanoses, comparable to those reported by Kohn et al. (1 57).

Diisoamylborane has been employed for the reduction of free aldonolac- tones. Because diisoamylborane reacts with free hydroxyl groups, an excess of the reagent is required (163). By this means, D-galactono- 1 ,Clactone, D-glucono- 1 $lactone, and D-erythrono- 1,4-lactone could be reduced to the corresponding aldoses in 60 - 70% yield. In the synthesis of 2-deoxy-~~- and L-riboses ( 164) through a Reformatsky reaction from noncarbohydrate pre- cursors, the intermediate 2-deoxy-~~- and L-ribonolactones were reduced to the deoxy sugars by diisoamylborane. Other unprotected 2-deoxylactones have been successfully reduced by diisoamylborane (42,165). Although the reduction of acylated or free lactones to aldoses with diisoamylborane is eminently satisfactory, the preparation of the reagent is tedious, and solu- tions of diisoamylborane are difficult to manipulate. In order to find a reagent having more desirable handling properties, other alkylboranes, in- cluding bis(2,4,4-trimethylpentyl)borane, bis(2,3-dimethyl-2-butyl)borane,

Page 184: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

SYNTHETIC REACTIONS OF ALDONOLACTONES 161

and dicyclohexylborane, have been studied (166). It was found that, when the size of the alkyl group is increased, the extent of reduction decreases, and thus diisoamylborane is the most satisfactory reagent. On the other hand, diisocamphenylborane, which is more readily prepared and handled than diisoamylborane ( 167), was successfully employed for the reduction of 2,3 : 5,6-di-O-isopropylidene-~-mannonolactone (lob) to the correspond- ing mannofuranose derivative.

Polarographic (1 68) and electrochemical (1 69) procedures for the reduc- tion of D-ribono- 1,4-1actone have been developed, and the latter has been applied on a pilot-plant scale.

2. Isotopic Labeling and Substitution at the Anomeric Center of Aldoses by Reduction of Aldonolactones

Reduction of aldonolactones and their derivatives with isotopically modi- fied reducing agents leads to sugars labeled at the anomeric center. Glyco- sides substituted with deuterium or labeled with tritium are widely employed for kinetic isotope-effect measurements, mechanistic studies, isotope-trac- ing experiments, and so on.

D-( 1 -2H)Glucopyranose was prepared (1 70) by direct reduction of D-glu- cono- 1,5-1actone, in deuterium oxide solution, with sodium amalgam in the presence of phosphoric acid-d,,. or, alternatively, by reduction of the lactone tetrahydropyranyl derivative mth sodium borodeuteride in tetrahydrofuran (171).

A one-pot, high-yielding synthesis of 1,2,3,4,6-penta-O-acetyI-j?-~-( 1 - 2H)glucopyranose (127) from tetra-0-acetyl-D-glucono- 1,5-1actone (126) has been reported (1 72). Sodium borodeuteride reduction of 126, followed by in situ acetylation, gave the readily isolated and crystalline 127. The crystalline 2,4-dinitrophenyl 2,3,4,6-tetra-O-acetyl-j?-~-( 1 -2H)glucopyran- oside (128) was subsequently obtained from 127.

CH,OAc CH,OAc CH,OAc

AcO Qo - AcO Q C

AcO QH3(N02)2

OAc OAc OAc

126 127 128

The cyanohydrin synthesis of higher sugars, which involves intermediate aldonolactones, allows the introduction of a I4C label in the sugar chain. Thus, for example, ~-[5-'~C]arabinose was synthesized (12) from D-xylose, which was first converted, by addition of K 14CN and hydrolysis, into D-[ 1 -

Page 185: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

162 ROSA M. DE LEDERKREMER AND OSCAR VARELA

14C]gulono- 1 ,Clactone. The hydroxyl groups at C-2 and -3 were protected by isopropylidenation, and the 5,6-glycol was oxidized by sodium periodate. Treatment of the resulting syrupy product with methanolic hydrogen chlo- ride, followed by borohydride reduction and hydrolysis, afforded L - [ ~ - ' ~ C Jarabinose.

IX. /%ELIMINATION REACTIONS Because of the activating effect of the carbonyl group, aldonolactone

derivatives readily undergo /I-elimination reactions to yield unsaturated lac- tones.

1. /%Elimination in Aldono-1,5-lactones a. Enono-1,5-lactones.-One of the first reports on p-elimination in al-

donolactones was the observation by Haworth and Long ( 173) of the unex- pected formation of furan-Zcarboxylic acid upon heating an aqueous solu- tion of 2,3,4-tri-0-methyl-~-xylono- 1,5-lactone in the presence of pyridine. In 1944, Isbell (1 74) advanced a mechanism based on enolization and p-elimination of the starting lactone.

In the course of investigations ( 175) on the structure of evernitrose, the corresponding 1,5-lactone 129, obtained upon oxidation of the sugar, yielded the a,j-unsaturated derivative 130 when refluxed with methanolic potassium acetate. The same enonolactone 130 was obtained from L-mycar- ose (125c). Meov __t KOAc MeOH Meou 0

I Me

129

/ Me

130

Oxidation of evermicose (122) with bromine yielded a mixture of y- and 8-lactones, which was directly acetylated. Refluxing the acetate in benzene solution in the presence ofp-toluenesulfonic acid gave ( 176) a mixture of the unsaturated lactones 131 and 132. In related work, Ganguly and Saksena (1 77) obtained an enonolactone by oxidation of D-nogalose with Jones' reagent, followed by @-elimination promoted by piperidine. Similarly, L-no- galose gave the enantiomeric lactone.

a,fl-Unsaturated derivatives are readily formed on acylation of aldonolac- tones under alkaline conditions. Thus, Dijong and Wittkotter (178) found that acetylation of 4-0-benzyl-D-g/~cero-D-~/~heptono- I ,5-lactone (133a)

Page 186: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

SYNTHETIC REACTIONS OF ALDONOLACTONES 163

/ Me Me

131 132

or its 7-0-trityl derivative (133b) with pyridine-acetic anhydride led to formation of the unsaturated 1 $lactone derivatives 134a and 134b, respec- tively.

CH,OR I

CH,OR I

HOCH

13nOQo - BlrQ 0

OAc HO OH

133 a R = H b R = T r

134 a R = A c b R = T r

In the hexosamine series, Kuzuhara and Emoto (179) obtained 2-benza- mido-4,ii-O-benzylidene-2,3-dideoxy-~-erythro-hex-2-enono- 1,5-lactone (136a) by refluxing the hexosaminic acid derivative 135a with acetic anhydride - pyridine. The 2-benzyloxycarbonyl derivative (135b) also un- derwent &elimination (1 80) when acetylated in basic media, to give 136b. A similar result was obtained on acetylation of 2-amino-4,6-0-benzylidene-2- deoxy-D-gluconic acid (135c) with acetic anhydride and sodium acetate, which afforded ( 18 1) the 2-enono- 1,5-1actone 136c.

135 a R = B z b R=CO,Bn

c R = H

136 a R = B z b R = C0,Bn

c R = A c

Page 187: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

164 ROSA M. DE LEDERKREMER AND OSCAR VARELA

Benzoylation in an excess of pyridine also favors p-elimination. Thus, treatment of D-glucono- 1 $lactone with an excess of benzoyl chloride and pyridine for 16 h at room temperature gave crystalline 2,4,6-tri-O-benzoyl- 3-deoxy-~-eryrhro-hex-2-enono- 1 ,5-lactone (137a) in 97% yield ( 182). Under similar conditions L-rhamnono- 1,5-lactone afforded ( 158) 2,4-di-0- benzoyl-3,6-dideoxy-~-erythro-hex-2-enono- 1 $lactone (138). p-Elimina- tion is also the main reaction occurring when acylated sugars having HO- 1 free are oxidized in basic media. When 2,3,4,6-tetra-0-acetyl-a-~-glucopyr- anose was treated with methyl sulfoxide - pyridine - triethylamine - sulfur trioxide, oxidation followed by elimination took place readily to afford ( 183) 2,4,6-tri-0-acetyl-3-deoxy-~-erythro-hex-2-enono- 1 $lactone (137b). The same product was obtained starting from p-D-glucopyranose tetraacetate and also from the mannose analog (1 84). On the other hand, the per-0- benzyl derivative ofglucose afforded the saturated 1 ,5-lactone 51a under the same conditions; that is, the benzyloxy group was not eliminated. However, Tsuda er al. (1 85) reported the formation of 2,4,6-tri-O-benzy1-3-deoxy-~- erythro-hex-2-enono- 1,5-lactone (137c) upon treatment of 2,3,4,6-tetra-0- benzyl-D-glucono- 1,5-lactone (51a) with sodium methoxide.

137 a R = Bz b R = A c c R = B n

1 38

In the aldopentose series, 2,3,4-tri-O-acetyl-a-~-xylopyranose afforded the corresponding unsaturated 1,5-1actone by oxidation - elimination. Like- wise, hepta-0-acetylcellobiose gave, upon oxidation ( 1 84), the product of p-elimination at the reducing end. 2,3,4,6-Tetra-0-benzoyl-~-gluco- and D-mannopyranoses were not oxidized by the same reagents.

Studies on 2-acetamido-2-deoxyaldohexoses, fully acetylated except at 0-1, have been reported (186). Oxidation of the gluco isomer 139 with methyl sulfoxide - acetic anhydride afforded the unsaturated lactone 141 as the major product, along with the fully acetylated lactone 140. The manno isomer also gave 141, but in much lower yield. The 0-benzyl analog of 139 did not undergo p-elimination when oxidized with the same reagent. Oxida- tion of 4,6-isopropylidene or benzylidene acetals of 2-acetamido-2-deoxy- hexoses of the D-gluco and ma man no configurations by the Fktizon reagent

Page 188: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

SYNTHETIC REACTIONS OF ALDONOLACTONES I65

(187) led (188) to the a,p-unsaturated lactones (142 or 136c), together with the saturated lactones. Treatment of the latter with ptoluenesulfonyl chlo- ride in pyridine afforded the p-elimination products (142 or 136c). The free unsaturated lactone, 2-acetamido-2,3-dideoxy-~-erythro-hex-2-enono- 1,5- lactone (143), was prepared from 142 by hydrolysis of the isopropylidene group. In a related paper, Pokorny et al. (1 89) reported the preparation of 2-acetamido-2,3-dideoxy-~-threo-hex-2-enono- 1,5-1actone.

Me,SOAc,O Qo + ~

0 - AcO AcO AcO

NHAc NU4c NHAc

139 140 141

0-CH, CH,OH

Me2(oQo - HO Qo

NHAc NHAc

142 i 43

b. a-Pyrones. - Acylated aldono- 1,5-1actones yield pyran-2-one deriva- tives by a double elimination, but more vigorous conditions than those used for the formation of 2-enono-lactones are required. Nelson and Gratzl(l90) described the production of 3-acetoxy-6-acetoxymethylpyran-2-one (144a), in 92% yield, upon heating D-glucono-1 ,5-lactone with acetic anhydride- pyridine for 1 h at 80°C. When the reaction was conducted at room temper- ature for 30 h, a mixture of the monounsaturated lactone (137b) and the a-pyrone 144ain 5.5 : 26 ratio was obtained. Treatment of L-rhamnono-l,5- lactone with an excess of benzoyl chloride and pyridine for 20 h, with subse- quent sublimation of benzoic acid from the mixture at 120°C in vucuo, afforded 3-benzoyloxy-6-methylpyran-2-one (145) as the main product (19 1). The conversion ofthe 2-enono-lactones 137b or 138 into the 2-pyrone derivatives 145 or 144a, respectively, required both acid and base, suggesting a concerted process in which the 4-acyloxy group is removed by acid, with the formation of an incipient cation stabilized by allylic resonance. Simulta- neous abstraction of H-5 by a base results in the stable a-pyrone. In fact, the 3-benzoyloxy-2-pyrones 144b and 145, and the 3-acylamido-2-pyrone 146,

Page 189: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

166 ROSA M. DE LEDERKREMER AND OSCAR VARELA

have been obtained by reaction of the unsaturated sugar-lactone derivatives 137a, 138, and 141, respectively, with tin(1V) chloride (192). On prolonged reaction, the 2-enonolactone 137a afforded the halogenated pyrone 144c. The 3-acetamido 2-pyrone 146 was obtained on treatment of 136c with concentrated hydrochloric acid followed by acetylation.

~o ~o ~

0

- __ - OR1 OBz NHAc

$44 a R1 = Ac, Rz = OAc 145 b R1 = Bz, R2 = OBz

c R1 = Bz. R2 = CI

146

2. &Elimination in Aldono-1,4-lactones

2( 5H)-Furanones (A%utenolides). - Aldono- 1 ,44actones readily un- dergo P-elimination on acylation to yield furan-2-one derivatives. Le- derkremer and Litter (1 93) reported formation of the furanone derivative 147 on benzoylation of D-galactono- 1 ,Clactone with an excess of benzoyl chloride and pyridine for 16 h at room temperature. The configuration of the exocyclic double bond was identified (1 94) as 2. P-Elimination also could be accomplished by treatment of the acylated lactone, in chloroform solu- tion, with triethylamine, which gave 147 as an (E,Z) diastereomeric mixture ( 194). Similarly, 2,3,5-tri-O-benzoyl-6-O-trityl-~-galactono- 1 ,Clactone af- forded the furan-2-one derivative 148 in 78% yield (195). The precursor enonolactones, resulting from a single elimination, could not be isolated, probably because of a fast second elimination yielding the very stable furan- one. However, prolonged acetylation (48 h) of 6-O-trityl-~-galactono- 1,4- lactone gave the monounsaturated lactone 149, but only in 9% yield, with

147 148 149

2,3,5-tri-O-acetyl-6-O-trityl-~-galactono- I ,4-lactone being the major prod- uct ( 196). In contrast, 5,6-O-isopropylidene-~-galactono-, L-gulono-, and

Page 190: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

SYNTHETIC REACTIONS OF ALDONOLA~ONES 167

D-mannono 1,4-lactones (8b, 9a, and 10a) react readily with mesyl chloride in pyridine at 0 "C to produce (1 97) the corresponding hex-2-enono- 1,4-lac- tone 2-mesylates 150- 152.

150 I51 152

Benzoylation of D-glycero-D-gulo-heptono- 1 ,Clactone with an excess of benzoyl chloride and pyridine afforded the hept-2-enono- 1 ,Clactone as the main product (1 98). The di- and triunsaturated compounds were isolated in very low yield from the mother liquors (199). Higher yields of the di- and triunsaturated derivatives 153 and 154 were obtained when the p-elimina- tion reaction was performed with triethylamine on the previously synthe- sized per-0-benzoyl D-glycero-D-@lo-heptono- 1,4-1actone. Employing 10% triethylamine in chloroform, the lactone 153 was obtained as an E, Z dias- tereomeric mixture in 9: 11 ratio as determined by 'H n.m.r. When 20% triethylamine was used, the furanone 154 was obtained in 59% yield (200). Its structure was assigned, on the basis of 'H and I3C n.m.r. spectra, as 3 - benzoyloxy-(5Z)- [ ( Z ) - 3-benzoyloxy-2-propenyliden] -2(5H)-furanone. The stereochemistry of the exocyclic double bonds was established (20 1) by nuclear Overhauser effect spectroscopy (NOESY).

, OBZ

153 154

\ OBZ

The elimination of benzoic acid from benzoylated aldonolactones takes place in successive steps. The relative extent of breaking the C-H bond and the C - OBz bond depends on the ease of removal of the hydrogen as a proton and the fact that the benzoate is a poor leaving-group. The H-2 proton in aldonolactones is acidic and it is likely that an E , cb mechanism, through an

Page 191: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

168 ROSA M. DE LEDERKREMER AND OSCAR VARELA

intermediate stabilized carbanion, operates in the first elimination step. This hypothesis is supported by the fact that the same furanone (147) was ob- tained from D-galactono-, D-glucono-, or D-mannono- 1 ,Clactones ( 194), having a cis-orientation of leaving groups in the first two and trans-leaving groups in the last one.

A similar mechanism could explain the elimination of the second mole- cule of benzoic acid, but formation of the triunsaturated lactone 154, ob- tained from the D-glyCero-D-gulo-heptOnOlaCtOne perbenzoate, would re- quire a different intermediate. The driving force in this case is the stability of the resulting furanone. An acid-base catalyzed process, such as that pro- posed for the formation ofthe a-pyrone 144a, could explain the formation of 154.

Treatment of 2,3,5-tri-O-benzoyl-~-rhamnono- 1,4-lactone with triethy- lamine as just described afforded 3-benzoyloxy-5-ethylidene-2( 5H)-furan- one (155) in 75% yield (202). 1,8-Diazabicyclo[5.4.0]undec-7-ene (DBU) effectively causes p-elimination in aldonolactone derivatives. Thus, 2,3,5- tri-0-benzyl-D-ribono- 1 ,Clactone afforded 2,5-di-O-benzyl-~-glycero- pent-2-enono-l,4-lactone (156a) in 9 1% yield when treated with DBU in hexamethylphosphoramide (203). Similarly, Barrett and Sheth (204,205) treated 2,3,5-tri-O-acetyl-~-ribono- 1,Clactone with DBU in tetrahydro- furan for 3 h at - 20°C and obtained 3-acetoxy-5-methylen-2(5H)-furan- one (157) in 94% yield. Other bases were less efficient. The monounsaturated compound 156b was formed, together with the furanone 157, in 2: 1 ratio when the elimination reaction was conducted with DBU for 4 h at - 78°C. Similarly, the corresponding enono-lactones 156c and 156d were obtained from the 5-0-trityl and the 5-0-tert-butyldiphenylsilyl derivatives, respec- tively. In a related reaction, Komura et al. (206) obtained the furanone 156e by mesylation - p-elimination of 2,5-di-O-tert-butyldimethylsilyl-~-ribono- 1,44actone.

- Rzocu H2&Q -

OBz OR1 OAc

155 156 a R1 = R2= Bn 157 b R1 = R2 = Ac

c R1 = Ac. R2 = Tr

d R1 = Ac. R2 = SiPh,But

e R1 = R2 = Si Me,But

The lactone derivative 158 obtained from D-ribono- 1 ,Clactone afforded (-)-(R)-angelica lactone (159a) upon treatment with methanolic ammonia solution (6). In a similar way, Font and coworkers (207) synthesized (+)-(S)-

Page 192: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

SYNTHETIC REACTIONS OF ALDONOLACTONES 169

angelica lactone (159b), and Danilova and coworkers (208) obtained (S)-hy- droxymethyl-2(5H)-furanone (159c). The lactone 160 synthesized from 2- deoxy-D-erythro-pentose afforded the furanone 161 by mesylation - elimination (209).

BzO

I 5 8 159 a R1 = CH,, R2 = H

b R1 = H, R2 = CH,

c R1 = CH,OH, R2 = H

I AH,OSiMe,But CH20SiMe,But

160 161

A trans-acetoxy elimination was described (2 10) for the 2-bromodeoxyal- dono-l,4-lactone 33b, induced by NaHSO,, to afford the butenolide 162a. However, the C-2 epimeric bromolactone 33a, undergoes a trans-P-bromo- acetoxy elimination to produce (21 1) butenolide 162b.

CH, Br I

AcOCH 0 u R

162 a R = Br b R = H

2-Acylamido-hex-2-enono- 1,4- (164aJ64b) and 1,5-lactone derivatives (141,165) are obtained ( 18 1) by acylation of 2-amino-2-deoxy-~-gluconic acid (163) with the acyl chloride in pyridine, whereas reaction of 163 with hot acetic anhydride-sodium acetate afforded (1 8 1,2 12) the butenolides 166-E and 166-2, earlier wrongly formulated (2 13) as 146. The isomers 166-2 and 167-2 were obtained in high yield on treatment of furanones 164a or 164b

Page 193: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

170

co; I + I

I

I

I

HCNH,

HOCH

HCOH

HCOH

CH,OH

163

ROSA M. DE LEDERKREMER AND OSCAR VARELA

CYOR I

NHR

964 a R = A c b R = B z

NHAc

166 ~ E

CH,OR I

141 R = A c 165 R = Bz

+ NHR

1 6 6 - Z R = A c 1 6 7 - 2 R = B z

with DBU ( 1 8 1). The P-elimination reaction was extended to a lactonic disaccharide. Thus, treatment of 168 with triethylamine afforded the diun- saturated derivative 169 as an E, 2 mixture ( I 94).

HCOBz OBz

I

HdOBz b B z I CH,OBz

uocH2 HQ OBz

HCOBz OBz I CH,OBz

168 169

X. SYNTHESIS OF DEOXY SUGARS FROM ALDONOLACTONES

1. Catalytic Hydrogenation of Enonolactones

Catalytic hydrogenation of benzoylated 3-deoxy-2-enono- 1 $lactones is stereoselective and affords the 3-deoxyaldonolactone derivatives, useful in- termediates for the synthesis of deoxy sugars. Thus, 2,4,6-tri-O-benzoy1-3-

Page 194: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

SYNTHETIC REACTIONS OF ALDONOLACTONES 171

deoxy-~-erythro-hex-2-enono- 1,5-lactone 137a, obtained in 97% yield from the commercially available D-glucono- 1,5-lactone, was selectively hydro- genated ( 182) over palladium - charcoal to give the 3-deoxy-~-arabino-hex- ono-l,5-lactone derivative 170. Reduction and debenzoylation of 170 af- forded (2 14) 3-deoxy-~-arabino-hexose (171). On the other hand, oxidative degradation of 3-deoxy-~-arabino-hexono- 1,5-lactone with ceric sulfate in M sulfuric acid gave 2-deoxy-~-erythro-pentose (2 1 5). Similarly, 2-deoxy- D-fyxo-hexose and 2-deoxy-~-arabino-hexose were synthesized from 3- deoxy-D-gafacto-heptono- 1 ,44actone and 3-deoxy-~-gfuco-heptono- 1,4- lactone, respectively (216). The reaction may be controlled to yield from common aldonolactones the aldose having one fewer carbon atom (2 17,2 18).

FHzOBz CH20H

170 171

The sequence /%elimination -catalytic hydrogenation - diisoamylborane reduction was applied (1 56) for the synthesis of ascarylose (174a). Starting from L-rhamnono-1 ,5-lactone, compound 174a was obtained via the di- deoxylactone 172s and the sugar derivative 173a. When catalytic hydrogen- ation of the enonolactone 138 was performed with deuterium, stereospecific labeling took place (2 19) at C-3 and at the 2R position of ascarylose (174b).

138 - BZOQ - BZOQ OH - H o p

OBZ OBZ OH

1 7 2 a R = H 173 a R = H 174 a R = H

b R = D b R = D b R = D

When the benzoylated lactone 172a was debenzoylated with sodium methoxide and the product boiled in 1 ,Cdioxane, the more-stable 1,4-lac- tone 175 was isolated upon rebenzoylation. Reduction of the lactone carbonyl group afforded (220) the crystalline furanose derivative (176) of as- carylose. A similar procedure, performed on the benzoylated 3-deoxy-~- arabino-hexono- 1,5-lactone 170, gave (22 1) the 3-deoxy-~-arabino-hexo- furanose derivative.

Page 195: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

172 ROSA M. DE LEDERKREMER AND OSCAR VARELA

1 7 2 a - BzOCH R OBz - BzOCH Icy OBz

I I CH3 CH3

175 176

Catalytic hydrogenation (194) of butenolide 147, obtained from D-galac- tono-, D-glucono-, or D-mannono- 1 ,Clactones, afforded stereoselectively the 2,6-di-0-benzoyl-3,5-dideoxy-~,~-threo-hexono- 1 ,Clactone 177.

i 77

From the glycosylfuranone 169, obtained via p-elimination, a 1 : 1 dias- tereoisomeric mixture of the 2(S), 4(R)- and 2(R), 4(S)-3,5-dideoxylactones 178-(S,R) and 178-(R,S) was obtained (194). Therefore, a glycosylation reaction was employed for the resolution of the racemic dideoxylactone derivative 177.

178 (S , R) 178 (R, S )

Trideoxy sugars have also been prepared from aldono- 1 ,Clactones. Thus, 2-0-benzoyl-3,5,6-t~deoxy-cu-~,~-thveo-hexofuranose was obtained (202) from L-rhamnono- 1 ,Clactone via the furanone 155. L-Rhamnono- 1,5-lac-

Page 196: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

SYNTHETIC REACTIONS OF ALDONOLACTONES 173

tone yielded (222), through the intermediate 3-benzoyloxy-6-methylpyran- 2-one (145), 2-0-benzoyl-3,4,6-trideoxy-~,~-threo-hexopyranose (179), which on debenzoylation gave the free sugar 180. Stereospecific catalytic hydrogenation of furanone 154 afforded (200) 2,7-di-O-benzoyl-3,5,6-tri- deoxy-D,L-threo-heptono- 1 ,Clactone.

145 - Q OH ---+ NaMeO Q OH

OBz OH

179 180

A simple procedure based on the sequence /3-elimination - catalytic hy- drogenation of acylated aldonolactones was described by Pedersen et al. (223). Acetylated pentono- and hexono- 1 ,Clactones, when hydrogenated under pressure in the presence of triethylamine, afforded the acetylated 3-deoxy-aldono- 1 ,Clactones in high yield. The 3-deoxygenation of peracyl- ated D-gzyCero-D-gulo-heptOnO- 1 ,Clactone was also described (44,224). The perbenzoate of 3-deoxy-~-gluco-heptono- 1,4-lactone was used (224) for the preparation of crystalline 3-deoxy-~-gluco-heptose.

Hydrogenation of the lactonic disaccharide 168 in the presence of triethyl- amine and palladium afforded (194) the 3-deoxylactone derivative 181.

I HC!OBz OBz

I

HCOBZ OBZ I CH,OBz

181

2. Hydrogenolysis of Bromodeoxyaldono-l,4-lactones

Deoxyaldono- 1 ,Clactones have also been obtained from the respective bromo derivatives. Thus, abequose (3,6-dideoxy-~-xyZo-hexose, 182) was

Page 197: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

174 ROSA M. DE LEDERKREMER AND OSCAR VARELA

obtained (225) from D-galactono- 1,44actone (Scheme I), and 2,5-dideoxy- D-erythro-pentono- 1 ,Clactone from the 5-bromo percursor (6). The starting material for the preparation of the latter was compound 3 (erroneously formulated as 2) (see Section 11).

CH3

l a - Qo- " G O H

OH HCOBz OBz I

HCOBz OBZ I CH,R CH3

R = OCPh3

R = Br

SCHEME 1

182

The bromine atom of 38, 41a, and 183 may be replaced by hydrogen through catalytic hydrogenolysis in the presence of triethylamine (42,43), by treatment with iodide (41), or by treatment with hydrazine (226). When no acid acceptor is present, hydrogenolysis of 2-bromo-2-deoxyaldono- 1,4-lac- tones yields the corresponding 2,3-dideoxyaldonolactone with removal of the bromine atom and the C-3 hydroxyl group (227). Thus the 2-bromo-al- donolactone 183 gave the 2,3-dideoxyaldonolactone 184. The two isomeric dibromolactones 185a and 185b both yielded 186a, which on hydrogenoly- sis in the presence of triethylamine was converted into the trideoxylactone 186b. The enantiomer was obtained by hydrogenolysis of 34.

HOCH, 0

P HO - 183 184

185 a R1 = Br, R2 z H 186 a R' = Br

b R1 = H, R2= Br b R 1 = H

Page 198: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

SYNTHETIC REACTIONS OF ALDONOLACTONES 175

3. Base-Catalyzed Rearrangement of Bromodeoxy Aldonolactones

Treatment of 6-bromoaldohexono- or 7-bromoaldoheptono- 1 ,Clactones with strong base leads to rearrangement via epoxide migration. The reaction has been applied to the synthesis of deoxylactones (226). For example, 6- bromo-2,6-dideoxy-~-arabino-hexono- 1 ,Clactone (187) yields 2-deoxy-~- ribo-hexono- 1 ,Clactone (190) through formation of the 5,6-epoxide 188, which, in strong base, rearranges to the 4,5-epoxide 189. The conversion of 189 into 191 may take place by intramolecular attack of the carboxylate anion on C-4 with inversion to yield the 1 ,Clactone 190, which opens with the base to give 191.

C y B r I

187

F" y2

HOCH

HOCH

HOCH

I I I CH,OH

191

y2

HOCH

188

J

I CH,OH

189

H&H I CHzOH

190

The 2-deoxylactone 190 was used for a synthesis of L-digitoxose (193). Treatment of 190 with hydrogen bromide in acetic acid gave the 6-bromo- lactone 192a, which on hydrogenolysis yielded the 2,6-dideoxylactone 192b. Reduction of 192b with diisoamylborane afforded 2,6-dideoxy-~-ribo-hex- ose (193). When 192a was treated with aqueous potassium hydroxide, it gave the salt of 2-deoxy-~-arabino-hexonic acid, which, after acidification and acetylation, yielded the tnacetate of 2-deoxy-~-arabino-hexono- 1,4-1actone (194). This base-catalyzed rearrangement was also applied to 6-bromo-3,6- dideoxy-aldohexono- 1 ,Clactones (228).

Page 199: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

176 ROSA M. DE LEDERKREMER AND OSCAR VARELA

190 - 0-0 H O ~ H

I CH, R

192 a R = Br b R = H

CHzOAc

H O ~ H I CH3

193

194

The 6-bromo-3,6-didoxy-~-urubino- and D-xyb-hexono- 1,6lactones 195 and 200 were formed on treatment of the corresponding 3-deoxylac- tones with hydrogen bromide in acetic acid. Upon treatment of 195 or 200 with anhydrous potassium carbonate in acetone, the two bromodeoxylac- tones were converted into the 5,6-epoxides 196 and 201. Treatment of the epoxide 196 or the 6-bromolactone 195 with excess of aqueous potassium hydroxide afforded the 3-deoxy-~-ribo-hexonate 198 as the main product, which was characterized as 2,5,6-tri-0-acetyl-3-deoxy-~-ribo-hexono- 1,4- lactone (199). The rearrangement occurs, as monitored by 13C-n.m.r. spec- troscopy, through the intermediate 197. On the other hand, the 5,6-epoxide 201, having the xylo configuration, gave the 2,6- and 2,5-anhydrides as the major reaction products as determined from the l3C-n.m.r. spectra of the methyl esters 202 and 203.3-Deoxy-~-lyxo-hexonic acid was isolated as the acetylated 3-deoxylactone 204 in 15% yield. The 6-bromolactone 195 was converted by hydrogenolysis, followed by carbonyl reduction, into 3,6-di- deoxy-D-arubino-hexose (tyvelose).

The behavior of 7-bromo-2,3,7-t~deoxy-~-arab~no-heptono 1 ,Clactone (205) and 7-bromo-2,7-dideoxy- (208) and 7-bromo-3,7-dideoxy-~-gluco- heptono- 1,Clactone (211) toward aqueous base has also been studied (229). The 6,7-epoxide is formed by potassium carbonate treatment of each bro- modeoxy heptonolactone. With potassium hydroxide, epoxide migration occurs giving mixtures of epoxides that undergo intramolecular attack by the

Page 200: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

SYNTHETIC REACTIONS OF ALDONOLACTONES 177

195 196

I CH20H

197

J F”;

y 2

HOCH I

I ACOCH I

HOCH

HOCH

I CH20H CH20Ac

I 9 9 198

HCOH OH HC, OH

CH2Br I H 2 k 0

200 201

CH20Ac

I CH20Ac OAc

202 203 204

Page 201: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

178 ROSA M. DE LEDERKREMER AND OSCAR VARELA

carboxylate or by an alkoxide. Thus 205 is converted into 2,3-dideoxy-~-ar- abino-heptonic acid (206), which was isolated as the acetylated lactone 207b. Treatment of crude 207a with hydrogen bromide in acetic acid gave the 7-bromolactone (207c) enantiomer of 205. The 7-bromo-2,7-dideoxy (208) and the 7-bromo-3,7-dideoxy-~-gluco-heptono- 1 ,Clactone (21 1) gave an- hydro heptonic acids on treatment with base. Thus, reaction of 208 with potassium hydroxide led to the formation of 3,6-anhydro-2-deoxy-~-ido- heptonic acid 209 as the main product, as determined by I3C-n.m.r. spec- troscopy. Compound 209 was isolated as the isopropylidene derivative 210. On the other hand, compound 211 gave the 2,5-anhydride 212a as the major product, accompanied by a small proportion of the 2,6-anhydride 213a. These products were isolated by column chromatography as the benzoylated methyl esters 212b and 213b, respectively.

H ~ O H I I

HCOH

CH,Br

205

F" y2

y2 - HCOH I I I

HOCH

HOCH

CH,OH

206

CH,R~

Hr,,

HCOR' 0

Yt

0 II

OH,C I Me2C"

\

H ~ O H I I

HCOH

CH,Br

208

HOLH,

209 210

Page 202: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

SYNTHETIC REACTIONS OF ALDONOLACTONES 179 Ici o l + 0 CH,OR~

I I

L OR2 OR2

R20

R~OCH I

HCOH OH

HCOH CH,0R2

CH2Br

21 I

XI. GLYCOSYLATION OF ALDONOLACTONES Selectively substituted aldonolactones have been used as glycosylating

agents for monosaccharides. Glycosylaldono- 1 ,Clactones are useful precur- sors of disaccharides having the reducing end in a furanoid-ring structure. Condensation of 2,3,5-tri-O-benzoyl-6-O-trityl-~-galactono- 1 ,Clactone with either tetra-0-acetyl-a-D-glucopyranosyl bromide or D-glucopyranose pentaacetate, in the presence of silver trifluoromethanesulfonate or stannic chloride, respectively, afforded the 6-~-~-~-ghcopyranosy~-~-galactono- 1 ,Clactone derivative 214 in good yield (230). Diisoamylborane reduction of 214 gave the P-~-Glcp-( 1 + 6)-P-~-Galfderivative 215.

CH,OAc HCOBz OBZ CH,OAc HCOBz OBZ .;jl - .;; AcO AcO

OAc OAc

214 215

Galactofuranose disaccharides have been obtained by condensation of D-galactofuranose pentabenzoate (216) with a conveniently substituted D-

galactono- 1 ,Clactone. Thus the stannic chloride-catalyzed glycosylation of 216 with 2,3,5-tri-O-benzoyl-~-galactono- 1,4-lactone (217) or its 6-0-trityl derivative gave the benzoylated P-D-galactofuranosyl-( 1 + 6)-~-galactono-

Page 203: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

180 ROSA M. DE LEDERKREMER AND OSCAR VARELA

174-lactone (218). Reduction of 218 by diisoamylborane led to the disaccha- ride derivative, from which the methyl glycoside was prepared. Debenzoyla- tion gave methyl D-galactofuranosyl-( 1 - 6)-P-~-galactofuranoside (219). The free disaccharide P-~-Galf( 1 4 6)-~-Galp was also prepared (23 1).

I I CH20H CH,OBz

216 21 7 21 8

I

HLOH AH I CH,OH

219

A similar strategy was used for the preparation of the P-D-Galf( 1 - 5 ) - ~ - Galf( 221) immunogenic unit of extracellular polysaccharides of Penicillum and Aspergillus species (232). 2,6-Di-O-benzoyl-~-galactono- 1,44actone (220), readily obtained by partial benzoylation of D-galactono- 174-lactone, was used to glycosylate the galactofuranose benzoate 216. The HO-5 group of 220 reacted preferentially to give the P-glycosyllactone 221 in 70% yield; the trisaccharide derivative 222 was isolated as a by-product. Reduction of

Page 204: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

SYNTHETIC REACTIONS OF ALDONOLACTONES 181

the lactones 221 and 222 with diisoamylborane followed by debenzoylation

Galf (38). gave P-D-Gal$( I + 5)-~-Galjand j?-~-Gal$( I 4 3)-v-~-Gd$( 1 - 5)]-D-

+ I I I I

I CH,OBz

HCOBz OBz I

HCOH OBz

CH,OBz

220 221

OBZ

CH,OBz

HCOBz CHzOBz I 41l OBz

222

XII. ALDONOLACTONES AS CHIRAL PRECURSORS FOR THE SYNTHESIS OF NATURAL PRODUCTS

The regio- and stereo-selective functionalization of aldonolactones yields optically active lactones, which are important precursors in natural product synthesis. Concepts such as “chiral templates” and “chirons,” derived from carbohydrates, have been ingeniously and widely applied in synthesis (233). Among the commercially available aldonolactones, D-ribono- 1 ,Clactone is

Page 205: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

182 ROSA M. DE LEDERKREMER AND OSCAR VARELA

probably the most intensively used in organic synthesis to date. The chemis- try and utility of D-ribono- 1 ,Clactone have been reviewed (2). Butenolides derived from D-ribonolactone, having a single chiral carbon atom, have been employed for inducing asymmetry.

1. Synthetic Uses of y-Butenolides Derived from Aldonolactones

Numerous procedures for the preparation of butenolides have been devel- oped. Font and coworkers (234 - 236) prepared the 5-0-substituted deriva- tives 223a-c of D-ribono-1 ,Clactone. The &glycol system of 223a reacted with N,N-dimethylformamide dimethyl acetal and then with iodomethane to give the trimethylammonium methylidene intermediate 224. Pyrolysis of 224 gave the butenolide 225.

223 a R=CH,

b R = PhCH,

c R = Ph3C

224 225

Butenolides have also been prepared from aldohexono- 1 ,Clactones via trimethylammonium methylidene derivatives ( 1 5). 5,6-0-Isopropylidene- L-gulono- (9a) and D-mannono- 1,44actone (10a) were converted into 2-(di- methylamino)- 1,3-dioxolane derivatives, which on treatment with iodo- methane followed by thermal decomposition yielded compounds 226 and 227 respectively.

227 226

Cyclic orthoformates are useful intermediates for the synthesis of buteno- lides (235). Treatment of D-nbonolactone, or its 5-0-substituted derivatives, with one molar equivalent of ethyl orthoformate gave diastereoisomeric

Page 206: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

SYNTHETIC REACTIONS OF ALDONOLACTONES 183

mixtures of orthoesters (228a - c) in quantitative yield. Pyrolysis of the ortho- formates 228a-c produced the corresponding butenolides (159c, 229a, and 2298).

228 a R = n 159 c R = X

b R=CH, 229 a R=CH, c R-PhCH, b R=PhCH,

Butenolide 231 was obtained by a procedure that did not involve a pyroly- tic step (237). Thus, the 5-trityl ether 223c was converted into the thioxocar- bonate 230 upon treatment with N,N'-thiocarbonylbis(imidazo1e). Raney nickel effected the conversion of 230 into 231.

Ph,COCH, 0 Ph,COCH, 0

'cj- y, 223 c -

o\c/o II S

230 231

Regiospecific trans-/?-bromo - acetoxy elimination of readily accessible 2-bromodeoxyaldono- 1 ,Clactones With NaHSO, afforded (2 1 1) good yields of such butenolides as 159c and 162b. Enantiomerically pure 5,6-epoxides and 5,6-diols were obtained from 162b.

Butenolides have been employed as convenient chiral precursors of such molecules as the antileukemic lignan lactones (+)-trans-burseran, (-)-isos- tegane, and (+)-steganacin (2). Butenolide 159c was identical to the product obtained by enzymic hydrolysis of ranunculin, a glycoside present in Ran- unculaceae. A short and efficient synthesis of (-)-ranunculin, via 159c, has been described (236). The reactivity of butenolides with different nucleo- philes has been studied (238). A general methodology for the preparation of y-alkyl-a,/?-butenolides or y-alkylbutanolactones from ribonolactone has been developed (239). Some of these products have pheromonal activity in

Page 207: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

184 ROSA M. DE LEDERKREMER AND OSCAR VARELA

insects and are also used as fruit fragrances. Among the y-alkyl-2,3-buteno- lides, the readily available (6,207,238,240) angelicalactones (159a and 159b) have served as chiral starting compounds for the synthesis of asymmetric molecules. For example, angelica lactone was used for the synthesis of (+)- and (-)-blastmycinone (240,24 1). Although a$-butenolides are inert to epoxidation by peroxyacids, epoxidation may be accomplished by using sodium hypochlorite in pyridine. Thus, (+)-angelica lactone (159b) gave stereoselectively the epoxy derivative 232. Oxirane ring-opening by iodide produced the diastereoisomeric mixture of iodides 233, which was converted into the 2-deoxy derivative 234 by hydrogenolysis. The lithium enolate of 234 was alkylated at C-2 and then esterified with isovaleryl chloride, afford- ing (+)-blastmycinone (235).

232 233

1 1

I I I CH3 Bu CH3

235 234

The double bond of butenolides undergoes stereoselective Michael addi- tion of organometallic reagents, affording useful synthetic intermediates. Thus 1,Caddition of lithium dimethylcuprate to 231 gave 236 as a single isomer, which was employed (237) for the synthesis of the bromopentene derivative 237.

Similarly, 1 ,Cconjugated addition (242) of lithium divinylcyanocuprate to 238 gave the adduct 239, which on treatment with potassium carbonate in methanol produced the epoxide 240, a key intermediate for the synthesis of the carbocycline 241 through highly stereoselective processes.

Page 208: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

SYNTHETIC REACTIONS OF ALDONOLACTONES 185

Ph,COCH, 0

231- ++- 1.I; Me

236 237

238 239 240

241

A total synthesis of (3S,4R)-(+)-eldanolide (246), a sex attractant phero- mone, has been reported (243). Compound 246 was synthesized by two different routes, both involving the butenolide 245 as the key precursor. The higher-yielding sequence is described here. Treatment of the tosylate acetal 242 with methanolic sodium methoxide led, as previously described by Hoffman and Ladner (244), to the epoxide 243. Addition of lithium diiso- butenylcuprate to 243 afforded 244, which after successive hydrolysis of the isopropylidene group, treatment with triethyl orthoformate, and pyrolysis,

Page 209: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

186 ROSA M. DE LEDERKREMER AND OSCAR VARELA

gave 245. The Michael addition of Me,CuLi to the latter takes place with high stereocontrol, the chirality at C-4 being responsible for the asymmetry induced at C-3, and gives the aldanolide 246. The reaction of differently substituted butenolides with alkyl cuprates has also been investigated, in order to obtain molecules having the (4S,5R)-4,5-dialkyldihydro-2-(3H)- furanone type of constitution. Natural products possessing such a structure, as for example (+)-eldanolide (246) and (+)-trans-cognac lactone (247), have been synthesized (245).

TsOCH, ~ ~ L i i e 0

o\ / o 0, /o CMe2 CMe,

242 243

FH2CH=CMe,

bo I

Me

246

(Me,CCH),CuLi

o\ /o CMe,

244

CH,CH=CMe,

Me2CuLi

245

Many strategies for stereochemical control on butenolides have been ex- plored, and also reviewed (246), by Hanessian. For example, starting from butenolides having the 44s) or 44R) configuration (respectively obtained from L-glutamic acid or D-ribonolactone) the C-2 - C- 10 and C- 1 1 - C- 16 subunits, 252 and 253, respectively, of the ionophore antibiotic ionomycin, have been synthesized (247). Conjugate addition of lithiumtris(trimeth- y1thio)methane to 248, followed by C-2 hydroxylation and desulfurization led to a 3-C-methyl lactone, which was readily converted into the epoxide 249. The butenolide 250 was prepared from 249 through a four-step synthe- sis that involves a two-carbon homologation, a lactone replication process, and oxidative elimination. Stereocontrolled addition of LiMe,Cu to 250,

Page 210: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

SYNTHETIC REACTIONS OF ALDONOLACTONES 187

followed by selective silylation and deoxygenation, led to 251, which served as precursor for the synthesis of 252 and 253. A similar methodology was also employed for the synthesis of the C-1 -C-13 polyol system (248), and the C-14-C-20 and C-32 -C-38 segments (249) of the antibiotic amphotericin B, from readily available, single-chiral progenitors.

249

J J

250

251

J J

\ \

Ph02S

252 253

The double bond of butenolides reacts under Diels - Alder conditions and the resulting chiral bicycles have served as precursors of prostacycline ana- logs and chrysanthemic acids (250,25 l). The butenolide 248 was obtained by theproceduredescribedbyIrelandeta1. (237). Abicyclo[4.3.0]ringsystem (254) was prepared by Diels-Alder reaction of 248 with butadiene in the presence of aluminum trichloride. Reduction of 254 (LiBH,) yielded the

Page 211: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

188 ROSA M. DE LEDERKREMER AND OSCAR VARELA

diol255, which was converted into the substituted tetrahydrofuran 256 via the tosyl derivative. Cleavage of the double bond of 256, followed by esterifi- cation, provided the diester 257. A bicyclo[3.3.0] ring system (258) was obtained by Dieckmann cyclization and subsequent demethoxycarbonyla- tion. On the other hand, compound 248 reacted with diazopropane to pro- duce a mixture of diastereoisomeric pyrazolines, which was converted into the same bicyclo[3.1 .O] system (259) by irradiation.

254 R = Bu'PhzSi 255 R Bu'Ph,Si

259 R = ButPhzSi 258 R = ButPh,Si

LJ 256 R = Bu'Ph,Si

I

257 R = ButPhzSi

The pyrazoline derivative 260 was also the precursor for the synthesis (252) of the naturally occumng umbelactone. Reaction of butenolide 159c with diazomethane gave the pyrazoline 260, which was subjected to pyroly- sis to give (-)-(S)-umbelactone (261). As the natural umbelactone was de- scribed as being dextrorotatory, the synthesis of (+)-(R)-umbelactone from 159c was also performed.

Cycloadditions (253) of butenolides with isoprene afforded a 1 : 1 mixture of Diels- Alder regioisomers. The selectivity is increased by the use of alu- minum trichloride as catalyst. Although the butenolides studied did not react with furan, even in the presence of catalysts, they reacted smoothly with cyclopentadiene. For example, reaction of (-)-angelica lactone (159a) with

Page 212: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

SYNTHETIC REACTIONS OF ALDONOLACTONES 189

CH3

159 c 260 261

isoprene for 20 h at - 200°C afforded an equimolecular mixture of the regioisomers 262 and 263. However, in the presence of A1C1, (6 days, SOOC), the ratio of 262 and 263 was 85 : 15. Reaction of 159a with cyclopentadiene at 70- 100°C gave the tricyclic endo (264) and ex0 (265) adducts. The endo- ex0 selectivity has been shown to be temperature-dependent.

159 a

264 265

2-Acyloxybutenolides obtained by p-elimination from ribonolactone (see Section IX.2) have served as appropriate chiral intermediates in several synthesis of antibiotics. Barrett and Sheth (205) reported a seven-step syn- thesis of racemic tert-butyl-8-O-tert-butyldimethylsilylnonactate, a mono- meric moiety of the antibiotic nonactin, from 157. Also, (4S,6S)-4- dimethyl- tert- butylsilyloxy - 6 - [(dimethyl- tert- butylsilyloxy)methyl] - tetra-

Page 213: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

190 ROSA M. DE LEDERKREMER AND OSCAR VARELA

hydro-2H-pyran-2-one (266), a suitable precursor for the spiro-acetal unit that occurs in milbemycins and overmectins, was synthesized (254) from the 2-acetoxybutenolides 156b and 156d.

CH,OR

266 R = ButMezSi

2. Synthetic Uses of SButenolides

2,3-Dideoxyhex-2-enono- 1,5-lactone derivatives (penten-5-olides) have been prepared (255 - 258) and employed as starting compounds in synthesis. Thus, Michael addition of benzylhydroxylamine to racemic 6-0-acetyl- 2,3,4-trideoxy-~,~-g~ycero-hex-2-enono- 1,5-1actone (267) took place ster- eoselectively to give the unstable benzyloxyamino-2-pyne 268, which was readily converted into the j3-lactam derivative 269, a precursor of thienamy- cin (259). j3-Lactams were also obtained (260) by 1,3-dipolar cycloaddition of nitrone 270 to the unsaturated 1,5-1actone 267, followed by hydrogenoly- sis and subsequent cyclization to the j3-lactam 271, having a polyol side- chain at the C-3 position. bo CH,OAc Do CH,OAc f l R

OR

- NH

0

NHOCH,Ph

267 268 269 R = SiMe,But

0, ,C,H40Me H O Y H ~, ,,pc6H40Me

N=C

Ph' 'H

0 Ph

270 27i

Page 214: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

SYNTHETIC REACTIONS OF ALDONOLACTONES 191

Racemic (267) and chiral(272a and 272b) lactones have been employed as dipolarophiles in cycloadditions with differently substituted nitrones (26 1). The reaction generally furnished two diastereomeric compounds, such as 273 and 274, from 272b. The configuration of the pyranoid ring was estab- lished on the basis of ‘H-n.m.r. data. The transformation of cycloadduct 273 into the p-lactam 276 was achieved (262) through the intermediate hemiace- tal 275. However, other adducts having methyl or benzyl substituents on nitrogen did not yield p-lactams, and deamination was observed instead. Reaction of such unsaturated 1 ,5-lactones as 272b with a mixture of hydrox- ylamine and formaldehyde (formaldoxime), in the nitrone form, gave an 8-aza-3,7-dioxa-2-oxo-bicyclo[4.3.0]nonane derivative via a 1,3-dipolar cy- cloaddition. Alternatively, a stepwise process yielded the 1 -aza-3,9-dioxa-8- oxo-bicyclo[4.3.0]nonane derivative (263). Dipolar cycloaddition of ben- zonitrile oxide to 272a gave the bicyclic products 277 and 278 in 58 and 7% yields respectively (264). The structure of 278 was determined by X-ray analysis. Deacetylation of 277 took place with contraction of the lactone ring.

CH,OAc CH,OAc I I

.

272 a R1 = OAc, RZ = H

b R1 = H. R2 = OAc

273 R1 = H, R2 = C,H,OMe

274 R1 = C6H,0Me. R2 = H Roq ~, ,.pc6H40 CH3

0 Ph

I Ph

275 276 R = SiMe,Eut

Page 215: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

192 ROSA M. DE LEDERKREMER AND OSCAR VARELA

272 a - 0

277 278

3. Other Syntheses from ~-Ribono-l&lactone

D-Ribonolactone is a convenient source of chiral cyclopentenones, acyclic structures, and oxacyclic systems, useful intermediates for the synthesis of biologically important molecules. Cyclopentenones derived from ribono- lactone have been employed for the synthesis of prostanoids and carbocyclic nucleosides. The cyclopentenone 280 was synthesized (265) from 2,3-0- cyclohexylidene-D-ribono- 1,4-1actone (16b) by a threestep synthesis that involves successive periodate oxidation, glycosylation of the lactol with 2- propanol to give 279, and treatment of 279 with lithium dimethyl methyl- phosphonate. The enantiomer of 280 was prepared from D-mannose by converting it to the corresponding lactone, which was selectively protected at HO-2, HO-3 by acetalization. Likewise, the isopropylidene derivative 282 was obtained (266) via the intermediate unsaturated lactone 281, prepared from 16a. Reduction of 281 with di-tert-butoxy lithium aluminum hydride, followed by mesylation, gave 282.

LiCH,PO(OMe),

Me2c"oQo - Q 16 b

0, / o 0, /o C6H10

279 280

o\ /o CMe,

281

o\ /o CMe,

282

Page 216: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

SYNTHETIC REACTIONS OF ALDONOLACTONES 193

The 2-trifluoromethanesulfonates of the four diastereomeric 3,5-di-O- benzyl-pentono- 1 ,Clactones (such as D-ribono- 1,6lactone, 283) gave, upon treatment with potassium carbonate in methanol, the ring-contraction product, methyl oxetane-2-carboxylate (284). The stereochemistry at C-2 of the resulting oxetanes is determined by the configuration at C-3, rather than C-2, of the starting lactones (267).

I I I BnO OSOzCF3 OBn

283 284

Kandil and Slessor (268) conducted the synthesis of (+)-lineatin (287), the aggregation pheromone of Trypodendron lineaturn, starting from 2,3-0- isopropylidene-D-ribono- 1 ,Clactone (16a). Grignard reaction of 16a with an excess of methylmagnesium iodide, produced a triol, which was selec- tively protected by acetylation, and then treatment with [Z(trimethylsilyl)- ethoxylmethyl chloride (SEM chloride) and deacetylation gave the glycol 285. Periodate oxidation of 285 and condensation of the resulting aldehyde function with diethyl (3-cyano- 1,l -dimethoxypropyl)phosphonate pro- vided the a,P-unsaturated nitrile 286, from which (+)-lineatin (287) was obtained through a 10-step synthetic route (2.7% overall yield from 16a).

HOCH, OH OSEM w:: - o\ / o

CMe, o\ / o

CMe,

~e

Me

285 286 287

The mycotoxin (-)-citreoviridin and the fungal metabolite (+)-citreoviral (291) have been synthesized (269) from D-ribonolactone, via the interme- diate 288. Addition of methyllithium to the lactone 288 gave the corre- sponding lactol289a, which was converted into the methyl acetal289b. This compound was treated with trimethylallylsilane-ZnBr, to give the doubly alkylated C-glycosyl compound 290, a key intermediate for synthesis of the natural products.

Page 217: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

194 ROSA M. DE LEDERKREMER AND OSCAR VARELA

Me I

288 289 a R = H b R = M e

J

I I

o\ / o CMe,

290

Me OH

291

A number of enantiomerically pure chiral building-blocks, such as 292 - 294, have been prepared (270,27 1 ) by zinc-copper cleavage of 5-bromo-5- deoxy-2,3-O-isopropylidene-~-ribono- 1 ,Clactone, followed by reduction. Similarly, from the 5-iodo lactone analogue the enoic acid 295 was obtained by reaction with zinc/silver-graphite (272).

292 293

294 295

Page 218: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

SYNTHETIC REACTIONS OF ALDONOLACTONES 195

Asymmetric synthesis of the C- 1 - C-5 segment 298 of a bis-normaytasin- oid was developed by Barton and coworkers (273) from the ditosyl derivative (296) of ribonolactone. Compound 296 was transformed into the dibro- mide, benzylated, and reduced to the 5-bromoaldose 297. Compound 298 was obtained in five steps from the intermediate 297.

296 297 298

4. Synthesis of Nucleosides, Amino Acids, and Other N-Containing Products from Aldonolactones

Aldonolactones have been employed as starting compounds for the syn- thesis of nucleosides. The first approaches to such syntheses were based on the reduction of aldono- 1 ,Clactone derivatives to the corresponding furan- oses, which were then coupled to purines or pyrimidines. Thus, 2-C-methyl- D-ribono- I ,Clactone (274) was reduced with diisoamylborane, the resulting HO-1 group was benzoylated, converted into the glycosyl chloride by HC1 in ether, and condensed with 9-chloromercuri-6-benzamidopunne. Removal of the benzoyl blocking groups gave 2’-C-methyladenosine (299). A similar procedure was employed (275) for the preparation of 9-fi-~-gulofuranosyl- adenine (300) from tetra-0-benzoyl-D-gulono- 1 ,Clactone. Compound 300 and 9-a-~-lyxofuranosyladenine (301) were also synthesized (276) from 2,3 : 5,6-di-O-isopropylidene-~-gulono- 1 ,Clactone (7a).

HO OH OH OH HO OH

HCOH I

CH,OH

299 300 301

Page 219: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

196 ROSA M. DE LEDERKREMER AND OSCAR VARELA

Borchardt and coworkers (265) have employed the chiral cyclopenten- ones derived from aldonolactones for the synthesis of the analogue 302a of neplanocin A. Neplanocin A (302b) and aristeromycin (303), carbocyclic analogs of adenosine having antiviral and antitumor activities, have also been synthesized (277,278).

302 a R = H b R = CH,OH

303

Methyl oxetane-2-carboxylate derivatives (e.g., 284), obtained by ring contraction of aldonolactones, have been employed for the synthesis (279) of the nucleoside P-noroxetanocin [9-(P-~-erythro-oxetanosyl)adenine, 3041 and its a-anomer via an a-chloride obtained by a modified Hunsdiecker reaction. Displacement of chloride by adenine and debenzylation gave 304. The threo isomer of 304, P-epinoroxetanocin (305), was likewise synthesized from D-lyxono- 1,4-1actone. The oxetane nucleosides display potent antiviral activity against the human immunodeficiency virus (HIV).

OH

304 Ad = Adenine 305

Such dideoxynucleosides as CNT (306) and the potent anti-HIV drug ddC (307) have been obtained (280), respectively, from the butenolide 248 and from its saturated analogue. Thus, conjugate addition of cyanide to 248, followed by reduction of the lactone group, acetylation of HO- 1 , and cou- pling with silylated thymine, afforded, after deprotection, compound 306.

Page 220: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

SYNTHETIC REACTIONS OF ALDONOLACTONES 197

0

Me+--L I 0

HOCH, 0 u CLo I

HOCH, 0 k2 CN

306 307

A general approach to the synthesis of polyfunctionalized amino acids from sugar lactones is exemplified by the synthesis (28 1,282) of the D-amino acid (2RY3S,4R)-3,4-dihydroxyproline (310) from 3,4-0-benzylidene-~-ri- bono-l,5-lactone(3). The C-2 hydroxyl group of 3 was derivatized as the trifluoromethanesulfonate to afford 308a. Nucleophilic displacement of the triflate by sodium azide took place, unexpectedly, with retention of configu- ration at C-2 to give the azide derivative 308b. Debenzylidenation followed by sulfonylation of the primary hydroxyl group gave the mesylate 309a. Hydrogenation of the azide group and treatment of the resulting amino mesyl lactone 309b with sodium hydroxide gave the proline derivative 310, whose structure was determined by X-ray crystallography to ensure that no epimerization had occurred at C-2.

308 a R = OSO,CF, 309 a R’ = N,, R2 = OS0,Me

b R=N, b R1 = NH,, R2 = OS0,Me

310

Other polyhydroxylated prolines have been also synthesized (283) from 0-isopropylidene derivatives of D-gulonolactone (7a) and D-galactonolac- tone (8b). Thus, starting from 7a, the pyrrolidine derivatives 311a and 311b were prepared by successive reduction of the lactone, mesylation of the resulting diol, cyclization with benzylamine, and removal of one or both 0-isopropylidene groups and the N-benzyl substituent. Oxidation of the diol 31 l a (after prior protection of the amino group) afforded the dihydroxypro-

Page 221: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

198 ROSA M. DE LEDERKREMER AND OSCAR VARELA

line 312, the enantiomer of 310. 1 ,4-Dideoxy-l,4-imino-~-glucitol, an ana- logue of 31 l b, was likewise synthesized from 8b.

7 a 5 steps -

311 a R = CMe,

b R = H

312

The pyrrolidine derivative 314, a skeletal analog of the antitumor antibi- otic anisomycin, was synthesized from the acetal derivative 16b. The 5-OH group of 16b was tosylated and then substituted with sodium azide. Reduc- tion (sodium borohydride) of the lactone group afforded an open-chain derivative, which was selectively protected to give 313. Hydrogenation of the wide function, followed by ptoluenesulfonylation, led to 314 by an intra- molecular nucleophilic displacement (284).

TS CH20CH2Ph I I

HCOTs

HCOH HCOCH,Ph I I C;"-..CH,Ph

PhCH,O OH I

CH2N3

- 0, / o

C6HVl

16 b 313 314

The d-lactam 108, obtained from D-gulonolactone (1 22), was employed for the synthesis of L-6-epicastanospermine (318a) and L- 1,6-diepicastano- spermine (318b). The natural enantiomer of 318a was similarly synthesized from L-gulonolactone (285). Compound 108 was benzylated and the lactam function reduced ( LiA1H4-A1Cl3) to the corresponding tertiary amine 315. Removal of the silyl protecting group, oxidation of the alcohol to the alde- hyde, and addition of vinylmagnesium bromide gave the epimeric alcohols, which were protected by silylation (316). After hydroboration of the alkene, the diastereomers were separated, and on treatment with mesyl chloride- Et3N, the quaternary ammonium salts 317a and 317b were obtained. Hy- drogenolysis and then acid hydrolysis of the protecting groups led to 318a and 318b, respectively.

Page 222: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

SYNTHETIC REACTIONS OF ALDONOLACTONES 199

315 R' = Bu'Me,Si 316 317 a R2 = H, R3= OR'

b R2 = OR'. R3 = H

318 a R2= H, R3=OH

b R2 = OH, R3 = H

Fleet and coworkers (20) also reported two different routes for the synthe- sis of the tetrahydropyrrolizidine 321, starting from D-glycero-D-gulo-hep- tono- 1 ,Clactone derivative 12. One approach involves the introduction of an azido group at C-7 via cleavage of the silyl ether, sulfonylation of HO-7, and substitution by azide. Reduction of the lactone (NaBH,) gave the azido- diol319a, which was mesylated. Hydrogenation of the azidomesylate 319b,

4 steps 12 - 6 steps

c- OS02Me 2 steps c-- 12

CH2N3

319 a R = H

b R = S0,Me

321 320

Page 223: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

200 ROSA M. DE LEDERKREMER AND OSCAR VARELA

followed by heating in ethanol in the presence of sodium acetate, led directly to the di-0-isopropylidene derivative of the pyrrolizidine. Removal of the acetals by acid hydrolysis gave compound 321. The alternative synthesis of 321 starts by reduction of the lactone group of 12 and then mesylation to afford 320. Nitrogen was introduced by treatment of 320 with benzylamine to give a monocyclic pyrrolidine. The formation of the second ring was achieved by removal of the silyl group, followed by mesylation. The resulting mesylate was unstable and spontaneously closed to give the N-benzyl pyrro- lizidinium salt, which was readily converted into 321.

Similar strategies have been used for the synthesis (286) of the tetrahydro- pyrrolizidine 323 from 2,3 : 5,6-di-O-isopropylidene-~-glycero-~-talohep- tono- 1 ,Clactone (322). These polyhydroxylated pyrrolidines and pyrrolizi- dines are potential specific inhibitors of glycosidases. The stereochemistry of the hydroxyl groups have a profound effect on the selectivity of the inhibi- tion (286-288).

322

Enantiomerically pure 4,5,6-trihydroxy-norleucins (for instance 325) were obtained ( 197) from the hex-2-enono- 1,4-1actone-2-mesylates (such as 152). These butenolides were stereoselectively hydrogenated to afford, upon treatment with sodium azide, the C-2-inverted derivatives, such as 324. Reduction of the azide function and hydrolysis of the acetal group gave the amino acids (namely 325), which were converted into lactones in acid media.

N3

324 325

Page 224: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

SYNTHETIC REACTIONS OF ALDONOLACTONES 20 1

2,3 : 5,6-Di-O-cyclohexylidene-~-gulono- I ,Clactone (326) was the start- ing compound for a synthesis (289) of (+)-negamycin (329). Reduction of 326 with DIBAL gave the gulofuranose derivative, which was converted into the corresponding oxime. The nitrone 327, generated in situ by reaction of the oxime with methyl glyoxylate, reacted in a highly enantioselective 1,3- dipolar cycloaddition with the benzyloxycarbonyl derivative of allylamine, to produce 3R, 5R-trans (328a) and 3S, 5R-cis (328b) adducts. The two new asymmetric centers created in 328a were elaborated to the 3R, 5R stereo- chemistry of 329.

HCO, H,~O/C6Hi0

HCO,

H2~o/c6H10

326 327

w N H C O , C H , P h I

I - 4- H, N

H

HCO I ‘CH

H,CO’ ‘ ’O 329 328 a R’ = H, Rz = C0,Me

b R’ = C0,Me. R2 = H

REFERENCES

1 . T. C. Crawford, Adv. Carbohydr. Chem. Biochem., 38 (1981) 287-321. 2. K. L. Bhat, S.-Y. Chen, and M. M. Joullit, Heterocycles, 23 (1985) 691-729. 3. A. H. Zinner, H. Voigt, and J. Voigt, Carbohydr. Res., 7 (1968) 38-55.

Page 225: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

202 ROSA M. DE LEDERKREMER AND OSCAR VARELA

4. N. Baggett, J. G. Buchanan, M. Y. Fatah, C. H. Lachut, K. J. McCullough, and J. M.

5. S. Y. Chen and M. M. Joullib, Tetrahedron Lett., 24 (1983) 5027-5030. 6. S. Y. Chen and M. M. Joullib, J. Org. Chem., 49 (1984) 2168-2174. 7. G. J. F. Chittenden and H. Regeling, Recl. Truv. Chim. Pays-Bus, 105 (1986) 186- 187. 8. P. J. Garegg, L. Maron, and C. G. Swahn, Actu Chem. Scund., 26 (1972) 5 18-522. 9. T. C. Crawford, U.S. Patent No. 4,111,958 (1978); Chem. Abstr., 90 (1979) 138150~.

10. M. Matsui, M. Saito, M. Okada, and M. Ishidate, Chem. Phurm. Bull., 16 (1968) 1294-

11. T. C. Crawford and R. Breitenbach, J. Chem. SOC. Chem. Commun. (1979) 388-389. 12. R. K. Hulyalkarand J. K. N. Jones, Can. J. Chem., 41 (1963) 1898-1904. 13. L. M. Lemer, B. D. Kohn, andP. Kohn, J. Org. Chem., 33 (1968) 1780-1783. 14. C. Copeland and R. V. Stick, Austr. J. Chem., 31 (1978) 1371-1374. 15. J. A. J. M. Vekemans, J. Boerekamp, E. F. Godefroi, and G. Chittenden, Red. Truv. Chim

16. H. Ogura, H. Takahashi, and T. Itoh, J. Org. Chem., 37 (1972) 72-75. 17. K. Ohle and G. Berend, Ber., 58 (1925) 2590-2592. 18. 0. T. Schmidt, C. C. Weber-Molster, and H. Hauss, Ber., 76 (1943) 339-344. 19. J. S. Brimacombe and L. C. N. Tucker, Curbohydr. Res., 2 (1966) 341 -348. 20. W. F. Collin, G. W. J. Fleet, M. H d d s s o n , I. C. di Bello, and B. Winchester, Curbohydr.

21. G. J. F. Chittenden, Recl. Truv. Chim. Pays-Bus., 107 (1988) 455-458. 22. H. Regeling, E. De Rouville, and G. J. F. Chittenden, Recl. Truv. Chim. Puys-Bus., 106

23. T. Kida, A. Masuyama, and M. Okahara, Tetrahedron Lett., 31 (1990) 5939-5942. 24. L. Hough, J. K. N. Jones and D. L. Mitchell, Can. J. Chem., 36 (1958) 1720- 1728. 25. E. C. Bigham, C. E. Gragg, W. R. Hall, J. E. Kelsey, W. R. Mallory, D. C. Richardson, C.

26. B. H. Lipshutz and M. C. Morey, J. Org. Chem., 46 (1981) 2419-2423. 27. A. M. Sepulchre, A. Gateau, and S. Gero, C.R. Acud. Sci. Paris. C, 269 (1969) 1312-

28. S. Takano, K. Inomata, and K. Ogasawara, Heterocycles, 27 (1988) 2413-2415. 29. L. M. Lerner, Curbohydr. Rex, 9 (1969) 1-4. 30. R. H. Shah, Carbohydr. Res., 155 (1986) 212-216. 31. R. H. Shah, J. Curbohydr. Chem., 5 (1986) 139-146. 32. N. Baggett, K. W. Buck, A. B. Foster, B. H. Rees, and J. M. Webber, J. Chem. SOC. C.

33. D. L. Mitchell, Can. J. Chem., 41 (1963) 214-217. 34. C. Hubschwerlen, Synthesis (1986) 962-964. 35. T. E. Walker and H. P. C. Hogenkamp, Carbohydr. Res., 32 (1974) 413-417. 36. A. Fernindez Cirelli, M. Sznaidman, L. Jeroncic, and R. M. de Lederkremer, J. Carbo-

37. L. 0. Jeroncic, M. L. Sznaidman, A. Fernandez Cirelli, and R. M. de Lederkremer,

38. R. M. de Lederkremer, C. Marino, and 0. Varela, Curbohydr. Res., 200 (1990) 227-235. 39. M. L. Sznaidman, A. FernPndez Cirelli, and R. M. de Lederkremer, Curbohydr. Res., 146

40. E. Mark and E. Zbiral, Monutsh. Chem., 112 (1981) 215-239. 4 1. K. Bock, I. Lundt, and C. Pedersen, Curbohydr. Res., 68 (1979) 3 13 - 3 19. 42. K. Bock, I. Lundt, and C. Pedersen, Carbohydr. Res., 90 (1981) 7 - 16.

Webber, J. Chem. SOC. Chem. Commun., (1985) 1826- 1827.

1299.

Pu~s-Bus., 104 (1985) 266-272.

Res., 202 (1990) 105- 116.

(1987) 461 -464.

Benedict, and P. H. Ray, J. Med. Chem., 27 (1984) 717-726.

1314.

(1966) 212-215.

hydr. Chem., 2 (1983) 167- 176.

Curbohydr. Res., 191 (1989) 130-137.

(1986) 233-340.

Page 226: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

SYNTHETIC REACTIONS OF ALDONOLACTONES 203

43. K. Bock, I. Lundt, and C. Pedersen, Curbohydr. Res., 90 (1981) 17-26. 44. K. Bock, 1. Lundt, C. Pedersen, and R. Sonnichsen, Curbohydr. Res., 174 (1988) 331 -

45. K. Bock, I. Lundt, and C. Pedersen, Actu Chem. Scund. B, 41 (1987) 435-441. 46. M. Bols and I. Lundt, Actu Chem. Scund B, 42 (1988) 67-74. 47. M. Bols and I. Lundt, Acut Chem. Scund., 45 (1991) 280-284. 48. M. Bols and I. Lundt, Actu Chem. Scund., 44 (1990) 252-256. 49. R. D. Guthrie and J. Honeyman, J. Chem. SOC., (1959) 853-854. 50. Y. A. Zhdanov, Y. E. Alexeev, and C. A. Khourdanov, Curbohydr. Res., 14 (1970)

5 1. Y. A. Zhdanov, Y. E. Alexeev, and V. A. Tyumenev, Dokl. AkadNuuk. SSSR., 226 (1976)

52. V. H. Shrivastava and L. M. Lemer, J. Org. Chem., 44 (1979) 3368-3373. 53. R. Csuk and B. I. GEnzer, J. Curbohydr. Chem., 9 (1990) 797-807. 54. R. Csuk and B. I. Glanzer, J. Curbohydr. Chem., 9 (1990) 809-822. 55. A. Kampf and E. Dimant, Curbohydr. Res., 32 (1974) 380-382. 56. H. Ogura and H. Takahashi, J. Org. Chem., 39 (1974) 1374- 1379. 57. H. Ogura, K. Furuhata, and H. Takahashi, Nucleic Acids Res., Spec. Publ. 3 (1977)

58. H. Ogura, K. Furuhata, H. Takahashi, and Y. Iitaka, Chem. Phurm. Bull., 26 (1978)

59. Y. A. Zhdanov, V. G. Alekseeva, E. L. Korol, and V. A. Polenov, Dokl. Akad Nuuk.

60. M. D. Lewis, J. K. Cha, andY. Kishi, J. Am. Chem. Soc., 104(1982) 4976-4978. 61. G. A. Krauss and M. T. Molina, J. Org. Chem., 53 (1988) 752-753. 62. S. Czemecki and G. Ville, J. Org. Chem., 54 (1989) 610-612. 63. H. Ogura, H. Takahashi, Synth. Commun., 3 (1973) 135- 143. 64. J. M. Lancelin, P. H. A. Zollo, and P. Sinay, Tetrahedron Lett., 24 (1983) 4833-

65. D. Rouzaud and P. Sinay, J. Chem. SOC. Chem. Commun. (1983) 1353- 1354. 66. A. Boschetti, F. Nicotra, L. P a m , G. R u m , and L. Zucchelli, J. Chem. SOC. Chem.

67. S. Hanessian and A. Ugolini, Curbohydr. Rex, 130 (1984) 261 -269. 68. D. Horton and W. Priebe, Curbohydr. Rex, 94 (1981) 27-41. 69. J. E. Hengeveld, V. Grief, J. Tadanier, C. M. Lee, D. Riley, and P. A. Lartey, Tetrahedron

70. C. S. Wilcox, G. W. Long and H. Sugh, Tetrahedron Lett., 25 (1984) 395-398. 71. T. V. RajanBabu and G. S. Reddy, J. Org. Chem., 5 1 (1986) 5458-5461. 72. M. H. Ah, P. M. Collins, and W. G. Overend, Curbohydr. Res., 205 (1990) 428-434. 73. R. Csuk, and B. I. Glanzer, Tetrahedron, 47 (1991) 1655- 1664. 74. A. Takahashi, Y. Kirio, M. Sodeoka, H. Sasai, and M. Shibasaki, J. Am. Chem. Soc., 1 I 1

75. D. Noort, G. H. Veeneman, G-J. P. H. Boons, G. A. van der Marel, G. J. Mulder, and J. H.

76. Y. Chapleur, Chem. Commun. (1984) 449-450. 77. A. Bandzouzi and Y. Chapleur, Curbohydr. Res., 171 (1987) 13-24. 78. A. Bandzouzi and Y. Chapleur, J. Chem. SOC. Perkin Trans., I (1987) 661 -664. 79. W. B. Motherwell, M. J. Tozer, and B. C. Ross, J. Chem. SOC. Chem. Commun. (1989)

80. W. B. Motherwell, B. C. Ross, and M. J. Tozer, Synlett. (1989) 68-69.

340.

422 -423.

1334-1336; Chem. Abstr., 85 (1976) 33285e.

23-26; Chem. Abstr., 89 (1978) 75429t.

2782-2787.

SSSR., 230 (1976) 856-859; Chem. Abstr., 86 (1977) 90157j.

4836.

Commun. (1989) 1085-1086.

Lett., 25 (1984) 4075-4078.

(1989) 643-647.

van Boom, Synlett (1990) 205-206.

1437-1439.

Page 227: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

204 ROSA M. DE LEDERKREMER AND OSCAR VARELA

81. K. Bischofberger, R. H. Hall, and A. Jordaan, Chem. Commun. (1975) 806-807. 82. R. H. Hall, K. Bischofberger, S. J. Eitelman, and A. Jordaan, J. Chem. SOC. Perkin Trans.,

83. S. J. Eitelman, R. H. Hall, and A. Jordaan, Chem. Commun. (1976) 923-924. 84. 0. F. Hedenburg, J. Am. Chem. SOC.. 37 (1915) 345-372. 85. K. Rehorst, Ber., 63 (1930) 2279-2292. 86. M. L. Wolfrom and H. B. Wood, J. Am. Chem. Soc., 73 (1951) 730-733. 87. A. M. Dempsey and L. Hough, Curbohydr. Res., 16 (1971) 449-451. 88. R. M. de Lederkremer, A. Femindez Cirelli, and J. 0. Deferrari, Carbohydr. Res., 13

89. 0. Varela, A. Fernindez Cirelli, and R. M. de Lederkremer, Anales Asoc. Quim. Argen-

90. I. Dijong and U. Wittkhtter, Chem. Ber., 104 (1971) 2806-2812. 91. D. E. Wright, Tetrahedron, 35 (1979) 1207-1237. 92. J. Yoshimura and M. Tamaru, Carbohydr. Res., 72 (1979) C9-Cll. 93. M. Tamaru, S. Horito, and J. Yoshimura, Bull. Chem. SOC. Jpn., 53 (1980) 3687-3690. 94. J. Yoshimura, S. Horito, and H. Hashimoto, Chem. Lett. (1982) 701 -703. 95. S. Horito, K. Asano, K. Umemura, H. Hashimoto, and J. Yoshimura, Carbohydr. Res.,

96. J. Yoshimura, K. Asano, K. Umemura, S. Horito, and H. Hashimoto, Carbohydr. Res.,

97. K. Asano, S. Horito, A. Saito, J. Yoshimura, and K. Ueno, Carbohydr. Rex, 136 (1985)

98. K. Asano, S. Horito, J. Yoshimura, T. Nakazawa, Z.-I. Ohya, and T. Watanabe, Curbo-

99. J. W. E. Glattfeld and D. MacMillan, J. Am. Chem. Soc., 56 (1934) 2481 -2482.

I(1977) 743-753.

(1970) 9-13.

tina, 69 (1981) 161-170.

121 (1983) 175-185.

121 (1983) 187-204.

1-11.

hydr. Res., 138 (1985) 325-328.

100. G. B. Robbins and F. W. Upson, J. Am. Chem. Soc., 60 (1938) 1788- 1789. 101. C.S.HudsonandS.Komatzu,J.Am. Chem.Soc.,41 (1919) 1141-1147. 102. J. W. W. Morgan and M. L. Wolfrom, J. Am. Chem. Soc., 78 (1956) 1897- 1899. 103. R. A. Weerman, Rec. Trav. Chim. Pays-Bas, 37 (1917) 52-66. 104. A. G. Renfrew and L. H. Cretcher, J. Am. Chem. Soc., 54 (1932) 4402-4404. 105. M. L. Wolfrom and R. B. Bennett, J. Org. Chem., 30 (1965) 458-462. 106. M. L. Wolfrom, R. B. Bennett, and J. D. Crum, J. Am. Chem. Soc., 80 (1958) 944-946. 107. Y. Nakajima and Y. Hoshi, Japanese Patent No. 2 1,240 (1964); Chem. Abstr., 62 (1965)

108. D. L. Tabem, U.S. Patent No. 2,084,626 (1937); Chem. Abstr., 31 (1937) 5949*. 109. M. Fieser, L. F. Fieser, E. Toromanoff, Y. Hirata, H. Heymann, M. T e a , and S. Bhatta-

1 10. J.-H. Fuhrhop, P. Schneider, E. Boekema, and W. Helfrich,J. Am. Chem. Soc., 1 10( 1988)

11 1. T. J. Williams, N. R. Plessas, and I. J. Goldstein, Carbohydr. Res., 67 (1978) C1 -C3. 112. T. J. Williams, N. R. Plessas, and I. J. Goldstein, Arch. Biochem. Biophys., 195 (1979)

113. W. N. Emmerling and B. Pfannemiiller, Carbohydr. Res., 86 (1980) 321 -324. 114. V. Zabel, A. Miiller-Fahmow, R. Hilgenfeld, W. Saenger, B. Pfannemiiller, V. Enkel-

115. G. Ziegast and B. Pfannemuller, Carbohydr. Rex, 160 (1987) 185-204. 116. K. Matsumoto, S. Hashimoto, T. Uchida, T. Okamoto, and S. Otani, Bull. Chem. SOC.

117. N. K. Richtmyer, Adv. Carbohydr. Chem., 6 (1951) 175-203.

11,900d.

chanya, J. Am. Chem. Soc., 78 (1956) 2825-2832.

2861 -2867.

145 - 15 1.

mann, and W. Welte, Chem. Phys. Lipids., 39 (1986) 313-327.

Jpn., 62 (1989) 3138-3142.

Page 228: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

SYNTHETIC REACTIONS OF ALDONOLACTONES 205

118. R. Hull, J. Chem. SOC. (1958) 4069-4073. 119. H. S. El Khadem and R. Sindric, Curbohydr. Res., 34 (1974) 203-207. 120. A. C. M. Barrett and D. Dhanak, Tetrahedron Lett., 28 (1987) 3327-3330. 12 1. A. G. M. Barrett, B. C. B. Bezuidenhoudt, D. Dhanak, A. F. Gasiecki, A. R. Howell, A. C.

Lee, and M. A. Russell, J. Org. Chem., 54 (1989) 3321 -3324. 122. G. W. J. Fleet, N. G. Ramsden, and R. Witty, Tetrahedron Lett., 29 (1988) 2871 -2874. 123. T. K. M. Shing, J. Chem. SOC. Chem. Commun. (1988) 1221. 124. B. Rajanikanth and R. Seshadri, Tetrahedron Lett., 30 (1989) 755-758. 125. G. W. J. Fleet, N. G. Ramsden, N. M. Carpenter, S. Petursson, and R. T. Aplin, Tetruhe-

126. G. W. J. Fleet, N. M. Carpenter, S. Petursson, and N. G. Ramsden, Tetrahedron Lett., 3 1

127. T. W. J. van Marle, Rec. Truv. Chim. Puys-Bus., 39 (1920) 562-565. 128. A. Thompson and M. L. Wolfrom, J. Am. Chem. SOC., 68 (1946) 1509- 1510. 129. L. Maquenne, Bull. SOC. Chim. Paris, 48 (1887) 719-723. 130. E. Fischer and F. Passmore, Ber., 22 (1889) 2728-2736. 131. H. H. Stroh and D. Henning, Chem. Ber., 100 (1967) 388-394. 132. S.M. Sharaf,M.A.E.Sallam,andH.A.ElShenawy,Curbohydr. Res., 91 (1981)39-48. 133. M. A. M. Nassr, M. A. M. Taha, and M. A. E. Shaban, Curbohydr. Res., 121 (1983)

134. M. A. E. Shaban, M. A. M. Nassr, and M. A. M. Taha, Curbohydr. Res., 113 (1983)

135. D. Beer and A. Vasella, Helv. Chim. Actu, 68 (1985) 2254-2274. 136. D. Beer and A. Vasella, Helv. Chim. Actu, 69 (1986) 267-270. 137. E. Fischer, Ber., 22 (1889) 2204-2205; 23 (1890) 930-938. 137a. R. U. Lemieux and U. Spohr, Adv. Curbohydr. Chem. Biochem., 50 (1994) 1-20. 138. E. Fischer and 0. Piloty, Ber., 24 (1891) 521-528. 139. C. Niemann, S. Kajala, and K. P. Link, J. Biol. Chem., 104 (1934) 189- 194. 140. M. Sutter and T. Reichstein, Helv. Chim. Actu., 21 (1938) 1210- 1218. 141. L. A. Mai, U.S.S.R. Patent No. 102,627 (1956); Chem. Abstr., 52 (1958) 4682i. 142. N. Sperber, H. E. Zaugg, and W. M. Sandstrom, J. Am. Chem. Soc., 69 (1947) 9 15 -920. 143. H. S. Isbell, J. V. Karabinos, H. L. Frush, N. B. Holt, A. Schwebel, andT. T. Galkowski, J.

144. J. W. E. GlattfeldandG. W. Schimpff,J. Am. Chem. Soc., 57 (1935) 2204-2208. 145. M. L. Wolfrom and H. B. Wood, J. Am. Chem. SOC., 73 (1951) 2933-2934. 146. H. L. Frush and H. S. Isbell, J. Am. Chem. Soc., 78 (1956) 2844-2846. 147. M. L. Wolfrom and A. Thompson, Methods Curbohydr. Chem., 2 (1963) 65-68. 148. L. M. Lerner, Curbohydr. Res., 44 (1975) 116- 120. 149. J. Nemek, and J. Jary, Collect. Czech. Chem. Commun., 33 (1968) 843-848. 150. S. S. Bhattacharjee, J. A. Schwarcz, and A. S. Perlin, Curbohydr. Hes., 42 (1975) 259-266. 151. R. Knollmann and I. Dyong, Chem. Ber., 108 (1975) 2021-2029. 152. I. Dijong and N. Jersch, Chem. Ber., 109 (1976) 896-900. 153. I. Dijong and D. Glittenberg, Chem. Ber., 110 (1977) 2721 -2728. 154. C. Fuganti and P. Grasselli, J. Chem. SOC. Chem. Commun., 7 (1978) 299-300. 155. H. C. Brown and D. B. Bigley, J. Am. Chem. SOC., 83 (1961) 486. 156. P. Kohn, R. H. Samaritano, andL. M. Lerner,J. Am. Chem. SOC., 86 (1964) 1457- 1458. 157. P. Kohn, R. H. Samaritano, andL. M. Lerner, J. Am. Chem. SOC., 87 (1965) 5475-5480. 158. 0. Varela, A. Fernandez Cirelli, and R. M. de Lederkremer, Curbohydr. Rex, 70 (1979)

159. R. Knollmann, N. Jersch, and I. Dijong. Chem. Ber., 110 (1977) 2729-2743.

dron Lett., 3 1 ( 1990) 405 -408.

(1990) 409-412.

119-124.

C16-C17.

Res. Nutl. Bur. Stand., 48 (1952) 163-171.

27-35.

Page 229: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

206 ROSA M. DE LEDERKREMER AND OSCAR VARELA

160. I. Dijong, L. Baumeister, and H. Bendlin, Chem. Ber., 112 (1979) 161- 174. 161. 0. Varela and R. M. de Lederkremer, unpublished results. 162. 0. Varela, C. Marino, and R. M. de Lederkremer, Curbohydr. Res., 155(1986) 247-251. 163. T. A. Giudici and A. L. Fluharty, J. Org. Chem., 32 (1967) 2043-2045. 164. G. Nakaminami, M. Nakagawa, S. Shioi, Y. Sugiyama, S. Isemura, and M. Shibuya,

165. G. Nakaminami, S. Shioi, Y. Sugiyama, S. Isemura, M. Shibuya, and M. Nakagawa, BUN.

166. P. Kohn, L. M. Lemer, A. Chan, Jr., S. D. Ginocchio, andC. A. Zitnn, Curbohydr. Res., 7

167. J. B. Lee and T. J. Nolan, Tetrahedron, 23 (1967) 2789-2794. 168. V. G. Mairanovskii, J. Swiderski, and A. M. Ponomarev, Kim.-Furm. Zh., 13 (1979)

62-65; Chem. Abstr., 91 (1979) 5425f. 169. A. Korczynski, L. Piszczek, J. Swiderski, and A. Doniec, Zesz. Nuuk. Politech. Slusk.

Chem., 631 (1979) 85-96; Chem. Abstr. 93 (1980) 114855. 170. M. L. Sinnott and I. J. L. Souchard, Biochem. J., 133 (1973) 89-98. 171. L. Hosie and M. L. Sinnott, Biochem. J., 226 (1985) 437-446. 172. L. A. Berven and S. G. Wethers, Curbohydr. Res., 156 (1986) 282-285. 173. W. N. Haworth and C. W. Long, J. Chem. SOC. (1929) 345-350. 174. H. S. Isbell, J. Res. Nutl. Bur. Stand., 32 (1944) 45-59. 175. A. K. Ganguly, 0. Z. Sarre, and H. Reimann, J. Am. Chem. Soc., 90(1968) 7129-7130. 176. A. K. Ganguly and 0. Z. Sarre, J. Chem. Soc. Chem. Commun. (1969) 1149- 1150. 177. A. K. Ganguly and A. K. Saksena, J. Chem. SOC. Chem. Commun. (1973) 531 -532. 178. I. Dijong and U. Wittkoetter, Tetrahedron Lett. (1969) 3793-3795. 179. H. Kuzuhara and S. Emoto, Agric. Biol. Chem. (Tokyo), 26 (1962) 334-340; Chem.

180. G. Deak, K. Gall-Istok, and P. Sohar, Acfu Chim. Acud. Sci. Hung., 75 (1973) 189- 191;

18 1. D. Horton, J. K. Thomson, 0. Varela, A. Nin, and R. M. de Lederkremer, Curbohydr.

182. R. M. de Lederkremer, M. I. Litter, and L. F. Sala, Curbohydr. Res., 36 (1974) 185- 187. 183. G.M.Cree,D.M.Mackie,andA.S.Perlin,Cun. J. Chem.,47(1969)511-512. 184. D. M. Mackie and A. S. Perlin, Curbohydr. Res., 24 (1972) 67-85. 185. Y. Tsuda, T. Nunozawa, and K. Yoshimoto, Chem. Phurm. Bull (Tokyo), 28 (1980)

186. N. PravdiE and H. G. Fletcher, Jr., Curbohydr. Res., 19 (1971) 353-364. 187. M. Fetizon and M. Golfier, Compt. Rend. C, 267 (1968) 900-903. 188. N. PravdiE, B. Danilov, andH. G. Fletcher, Jr., Curbohydr. Res., 36 (1974) 167-180. 189. M. Pokomy, E. Zissis, H. G. Fletcher, Jr., and N. PravdiE, Curbohydr. Res., 43 (1975)

190. C. R. Nelson and J. S. Gratzl, Curbohydr. Res., 60 (1978) 267-273. 191. 0. Varela, A. Fernandez Cirelli, and R. M. de Lederkremer, Curbohydr. Res., 79 (1980)

192. A. Nin, 0. Varela and R. M. de Lederkremer, Synthesis (1991) 73-74. 193. R. M. de Lederkremer and M. I. Litter, Curbohydr. Res., 20 (1971) 442-444. 194. C. Marino, 0. Varela, and R. M. de Lederkremer, Curbohydr. Res., 220 (1991) 145- 153. 195. R. M. de Lederkremer, C. Du Mortier, and 0. Varela, Rev. Latinoam. Quim., 17 (1986)

196. M. L. Sznaidman, A. Femandez Cirelli, and R. M. de Lederkremer, Curbohydr. Res., 146

Tetrahedron. Lett. (1967) 3983-3987.

Chem. SOC. Jpn., 45 (1972) 2624-2634.

(1968) 21-26.

Abstr., 58 (1963) 4638h.

Chem. Abstr.. 78 (1973) 97934.

Res., 193 (1989) 49-60.

3223-3231; Chem. Abstr., 94 (1981) 19259511.

345 - 354.

2 19-224.

16- 19.

(1986) 233-240.

Page 230: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

SYNTHETIC REACTIONS OF ALDONOLACT'ONES 207

197. J.A. J. M. Vekemans,R.G.M.deBruyn,R.C.H.M.Caris,A. J.P.M.Kokx,J. J.H.G. Konings, E. F. Godefroi, and G. J. F. Chittenden, J. Org. Chem., 52 (1987) 1093- 1099.

198. M. I. Litter and R. M. de Lederkremer, Curbohydr. Rex, 26 (1973) 431 -434. 199. M. I. Litter and R. M. de Lederkremer, Andes Asoc. Quim. Argentina, 62 (1974) 147-

200. L. 0. Jeroncic, 0. J. Varela, A. Fernandez Cirelli, and R. M. de Lederkremer, Tetruhe-

20 1. L. 0. Jeroncic, Doctorate Thesis, University of Buenos Aires, 1988. 202. 0. Varela, A. Fernandez Cirelli, and R. M. de Lederkremer, Curbohydr. Res., 100 (1982)

203. W. Timpe, K. Dax, N. Wolf, and H. Weidmann, Curbohydr. Res., 39 (1975) 53-60. 204. A. G. M. Barrett and H. G. Sheth. J. Org. Chem., 48 (1983) 5017-5022. 205. A. G. M. Barrett and H. G. Sheth, Chem. Commun. (1982) 170- 171. 206. H. Komura, T. Iwashita, H. Naoki, and K. Nakanishi, J. Am. Chem. Soc., 105 (1983)

207. R. M. Ortuiio, D. Alonso, and 5. Font, Tetrahedron Lett., 27 (1986) 1079- 1082. 208. G. A. Danilova, V. I. Mel'nikova, and K. K. Pivnitsky, Tetrahedron Lett., 27 (1986)

209. K. Tadano, Y. Iimura, T. Ohmori, Y. Ueno, and T. Suami, J. Curbohydr. Chem., 5 (1986)

210. C. Pedersen, K. Bock, and I. Lundt, PureAppl. Chem., 50 (1978) 1385-1400. 2 1 1. J. A. J. M. Vekemans, G. A. M. Franken, C. W. M. Dapperens, E. F. Godefroi, and G. J. F.

212. C. T. Clarke, J. H. Jones, and R. Walker. J. Chem. SOC. Perkin Trans., Z (1976) 1001 -

213. M. Bergmann, L. Zervas, and E. Silberkweit, Ber., 64 (1931) 2428-2436. 214. C. Du Mortier and R. M. de Lederkremer, J. Carbohydr. Chem., 3 (1984) 219-228. 215. R. M. de Lederkremer and L. F. Sala, Curbohydr. Res., 40 (1975) 385-386. 216. L. F. Sala, A. Fernandez Cielli, and R. M. de Lederkremer, Curbohydr. Res., 78 (1980)

2 17. L. F. m a , A. Fernlndez Cirelli, and R. M. de Lederkremer, J. Chem. Soc. Perkin Trans.,

2 18. L. F. Sala, A. Fernindez Cirelli, and R. M. de Lederkremer, Andes Asoc. Quim. Argen-

2 19. R. N. Russell, T. W. Weigel, 0. Han, and H. Liu, Curbohydr. Res., 201 ( 1990) 95 - 1 14. 220. 0. Varela, A. Fedndez Cirelli, and R. M. de Lederkremer, Curbohydr. Res., 85 (1980)

22 1. C. Du Mortier and R. M. de Lederkremer, Curbohydr. Rex, 139 ( 1985) 47- 54. 222. M. Sznaidman, A. Fernindez Cirelli, and R. M. de Lederkremer, Andes Asoc. Quim.

223. K. Bock, I. Lundt, and C. Pedersen, Actu Chem. Scund. B, 35 (1981) 155- 162. 224. L. 0. Jeroncic, A. Fernlndez Cirelli, and R. M. de Lederkremer. Curbohydr. Rex, 167

225. 0. Moradei, C. Du Mortier, 0. Varela, and R. M. de Lederkremer, J. Curbohydr. Chem.,

226. K. Bock, I. Lundt, and C. Pedersen, Actu. Chem. Scund. B, 38 (1984) 555-561. 227. I. Lundt and C. Pedersen, Synthesis (1986) 1052- 1054. 228. K. Bock, I. Lundt, andC. Pedersen, Actu. Chem. ScundB, 40 (1986) 163-171. 229. K. Bock, I. Lundt, and C. Pedersen, Curbohydr. Rex, 179 (1988) 87-96. 230. C. Du Mortier, 0. Varela, and R. M. de Lederkremer, Curbohydr. Res., 189 (1989) 79- 86.

150.

dron, 40 (1984) 1425- 1430.

424-430.

5 164- 5 165.

2489-2492.

423 -435.

Chittenden, J. Org. Chem., 53 (1988) 627-633.

1003.

61 -66.

ZI(1977) 685-688.

tina, 66 (1978) 57-63.

130-135.

Argent., 70 (1982) 341-348.

(1987) 175-186.

10 (1991) 469-479.

Page 231: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

208 ROSA M. DE LEDERKREMER AND OSCAR VARELA

231. C. Marino, 0. Varela, and R. M. de Lederkremer, Carbohydr. Res., 190 (1989) 65-76. 232. S. Notermans, G. H. Veeneman, C. W. E. M. van Zuylen, P. Hoogerhout, and J. H. van

233. S. Hanessian Total synthesis of natural products: The chiron approach. Pergamon, 1983. 234. P. Camps, J. Font, and 0. Ponsati, Tetrahedron Lett., 22 (1981) 1471-1472. 235. J. Cardellach, C. Estopa, J. Font, M. Moreno-Mafias, R. M. Ortuiio, F. Sinchez-Ferrando,

236. P. Camps, J. Cardellach, J. Font, R. M. Ortuiio, and 0. Ponsati, Tetrahedron, 38 (1982)

237. R. E. Ireland, R. G. Anderson, R. Badoud, B. J. Fitzsimmons, G. J. McGarvey, S.

238. P. Camps, J. Cardellach, J. Corbera, J. Font, R. M. Ortufio, and 0. Ponsati, Tetrahedron

239. J. Cardelach, J. Font, and R. M. Ortuiio, J. Heterocyclic Chem., 21 (1984) 327-331. 240. R. M. Ortuiio, D. Alonso, J. Cardellach, and J. Font, Tetrahedron, 43 (1987)2192-2198. 241. J. Cardellach, J. Font, and R. M. Ortu60, Tetrahedron Lett., 26 (1985) 2815-2819. 242. P. Magnus and D. P. Becker, J. Am Chem. Soc., 109 (1987) 7495-7498. 243. R. M. Ortuiio, R. Merck and J. Font, Tetrahedron Lett., 27 (1986) 2519-2522. 244. R. W. Hoffmann and W. Ladner, Chem. Ber., 116 (1983) 1631 - 1642. 245. R. M. Ortuiio, R. Merck, and J. Font, Tetrahedron, 43 (1987) 4497-4506. 246. S. Hanessian, Aldrichim Acta, 22 ( 1989) 3 - 15. 247. S. Hanessian and P. Murray, Can. J. Chem., 64 (1986) 2231 -2234. 248. S. Hanessian, S. P. Sahoo, and M. Botta, Tetrahedron Lett., 28 (1987) 1147- 1150. 249. S. Hanessian and M. Botta, Tetrahedron Lett., 28 (1987) 1 15 1 - 1 154. 250. J. Mann and A. Thomas, Chem. Commun. (1985) 737-738. 251. M. G. B. Drew, J. Mann, and A. Thomas, J. Chem. SOC. Perkin Trans., I(1986) 2279-

252. R. M. Ortuiio, J. Bigorra, and J. Font, Tetrahedron, 43 (1987) 2199-2202. 253. R. M. Ortufio, R. Batllori, M. Ballesteros, M. Monsalvatje, J. Corbera, F. Sinchez-

254. S. V. Attwood and A. G. M. Barren, J. Chem. SOC. Perkin Trans., I (1984) 1315- 1322. 255. J. Mieczkowski, J. Jurczak, M. Chmielewski, and A. Zamojski, Carbohydr. Res., 56

256. P. Rollin and P. Sinay, Carbohydr. Res., 98 (1981) 139- 142. 257. P. J. Card, Carbohydr. Rex, 104 (1982) 338-340. 258. P. Jarglis and F. W. Lichtenthaler, Tetrahedron Lett., 23 (1982) 3781 -3784. 259. M. Chmielewski and S. Maciejewski, Carbohydr. Res., 157 (1986) Cl -C3. 260. I. Panfil, M. Chmielewski, and C. Belzecki, Heterocycles, 24 (1986) 1609- 1617. 261. I. Panil and M. Chmielewski, Tetrahedron, 41 (1985) 4713-4716. 262. I. Panfil, C. Belzecki, and M. Chmielewski, J. Carbohydr. Chem., 6 (1987) 463-470. 263. I. Panfil, C. Belzecki, M. Chmielewski, and K. Suwinska, Tetrahedron, 45 (1989) 233-

264. H. Gnichtel, L. Autenrieth-Ansoge, and J. Dachman, J. Carbohydr. Chem., 6 (1987)

265. D. R. Borcherding, S. A. Scholtz, and R. T. Borchardt, J. Org. Chem., 52 (1987) 5457-

266. P. Belanger and P. Prasit, Tetrahedron Lett., 29 (1988) 5521 -5524. 267. D. R. Witty, G. W. J. Fleet, K. Vogt, F. X. Wilson,Y. Wang, R. Storer, P. L. Myers, andC.

268. A. A. Kandil and K. N. Slessor, J. Org. Chem., 50 (1985) 5649-5655.

Boom, Mol. Imrnunol., 25 (1988) 975-979.

S. Valle, and L. Vilamajo. Tetrahedron, 38 (1982) 2377-2394.

2395-2402.

Thaisrivongs, and C. S. Wilcox, J. Am. Chem. SOC., 105 (1983) 1988-2006.

Lett., 39 (1983) 395-400.

2285.

Ferrando, and J. Font, Tetrahderon Lett., 28 (1987) 3405-3408.

(1977) 180-182.

238.

673-683.

546 1.

J. Wallis, Tetrahedron Lett.. 3 1 (1990) 4787-4790.

Page 232: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

SYNTHETIC REACTIONS OF ALDONOLACTONES 209

269. H. Suh and C. Wilcox, J. Am. Chem. SOC., 110 (1988) 470-481. 270. B. Hafele, D. Schroter, and V. JQer, Angew. Chem. Znt. Ed. Engl., 25 (1986) 87-89. 271. V. Jager and B. HSele, Synthesis(1987) 801-806. 272. A. Furstner and H. Weidmann, J. Org. Chem., 54 (1989) 2307-231 1. 273. D. H. R. Barton, M. Btnkchie, F. Khuong-Huu, P. Potier, andV. Reyna-Pinedo, Tetrahe-

274. E. Walton, S. R. Jenkins, R. F. Nutt, M. Zimmerman, and F. W. Holly, J. Am. Chem.

275. P. Kohn, R. H. Samaritano, and L. M. Lerner, J. Org. Chem., 31 (1966) 1503- 1506. 276. L. M. Lerner, B. D. Kohn, and P. Kohn, J. Org. Chem., 33 (1968) 1780-1783. 277. M. S. Wolfe, D. R. Borcherding, and R. T. Borchardt, Tetrahedron Lett., 30 (1989)

278. M. S. Wolfe, B. L. Anderson, D. R. Borcherding, and R. T. Borchardt, J. Org. Chem., 55

279. F. X. Wilson, G. W. J. Fleet, D. R. Witty, K. Vogt, Y. Wang, R. Storer, P. L. Myers, and C.

280. M. Okabe, R-C Sun, S. Y. -K. Tam, L. J. Todaro, and D. L. Coffen. J. Org. Chem., 53

28 1. J. C. Dho, G. W. J. Fleet, J. M. Peach, K. Prout, and P. W. Smith, Tetrahedron Lett., 27

282. P. D. Baird, J. C. Dho, G. W. J. Fleet, J. M. Peach, K. Prout, and P. W. Smith, J. Chem.

283. G. W. J. Fleet and J. C. Son, Tetrahedron, 44 (1988) 2637-2647. 284. A. M. Sepulchre, A. Gateau, A. Gaudemer, and S. D. Gero, J. Chem. SOC. Chem. Com-

285. G. W. J. Fleet, N. G. Ramsden, R. J. Molyneux, and G. S. Jacob, Tetrahedron Lett., 29

286. A. J. Fairbanks, G. W. J. Fleet, A, H. Jones, I. Bruce, S. A. Dahler, I. Cenci di Bello, and B.

287. S. A. Dahler, G. W. J. Fleet, S. K. Namgoong, and B. Winchester, Biochem. J., 258 (1989)

288. I. Cenci di Bello, G. W. J. Fleet, S. K. Namgoong, K. I. Tadano, and B. Winchester,

289. H. Iida, K. Kasahara, and C. Kibayashi, J. Am. Chem. Soc., 108 (1986) 4647-4648.

dron Lett., 23 (1982) 651 -654.

SOC., 88 (1966) 4524-4525.

1453- 1456.

(1990) 4712-4717.

J. Wallis, Tetrahedron: Asymmetry, 1 (1990) 525-526.

(1988) 4780-4786.

(1986) 5203-5206.

SOC. Perkin, Trans. Z(1987) 1785-1791.

mum. (1970) 759-760.

(1988) 3603-3606.

Winchester, Tetrahedron, 47 ( 199 1) 13 1 - 138.

6 13 -6 15.

Biochem. J., 259 (1989) 855-861.

Page 233: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

This Page Intentionally Left Blank

Page 234: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

ADVANCES IN CARBOHYDRATE CHEMISTRY AND BIOCHEMISTRY, VOL. 50

MOLECULAR STRUCTURE OF LIPID A, THE ENDOTOXIC CENTER OF BACTERIAL LIPOPOLYSACCHARIDES'

BY ULRICH ZAHRINGER, BUKO LINDNER, AND ERNST TH. RIETSCHEL

Department of Immunochemistry and Biochemical Microbiology, Forschungsinstitut Borstel, Institut fur Experimentelle Biologie und Medizin, 0-23845 Borstel, Germany

I. INTRODUCTION Gram-negative bacteria such as the Enterobacteriaceae, Neisseriaceae,

and Pseudomonadaceae express at their outer membrane amphiphilic mac- romolecules termed lipopolysaccharides (LPS, endotoxin), which are of considerable microbiological, genetical, chemical, pharmacological, and, in particular, biomedical interest ( 1 - 9b). LPS participate in various physiolog- ical membrane functions essential for growth and survival of Gram-negative bacteria. LPS also constitute the heat-stable 0-antigens of the bacteria and, thus, serve to identify the multiplicity of 0-serotypes. Finally, LPS are en- dowed with an overwhelming spectrum of biological activities, expressed either in vitro or after injection into experimental animals. LPS constitute potent immunostimulators and adjuvants (lo), but have also been recog- nized as playing a key role in the pathogenesis and manifestations of Gram- negative infection in general and of septic shock in particular. Thus, LPS are among the most potent agents capable of inducing local or generalized inflammatory reactions in both humans and experimental animals.

In the past, much emphasis has been placed on identifying structural principles of LPS and on elucidating their chemical structures with the aim, among other reasons, of defining the substructures that determine the various biological effects of LPS. This objective has been achieved for a number of such activities. These results, in turn, have stimulated the chemi- cal synthesis of biologically active LPS substructures and chemical ana- logues (1 l - 19). At the same time, the mechanisms of biological action of endotoxin are being studied at the molecular level, providing a rational basis

I This article is dedicated to Professor Dr. Dr. med. h.c. Otto Westphal on the occasion of his 80th birthday (February lst, 1993).

Copyright 0 1994 by Academic press Inc. AU rights of reproduction in any form reserved. 21 1

Page 235: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

212 ULRICH ZAHRINGER et al.

- 0-Speci f ic Outer Inner Chain Core Core

for strategies aimed at the immunological or pharmacological prevention and treatment of endotoxicosis (20,2 1).

Structurally, LPS is comprised of three genetically, chemically and anti- genically distinct regions, namely the 0-specific chain, the core oligosaccha- ride, and lipid A (Fig. 1). The latter serves to anchor LPS to the bacterial membrane (3). The 0-specijk chain is a heteropolymeric polysaccharide and responsible for the 0-antigenic properties of LPS, which denote the induction and binding of species-specific antibodies. It is composed of up tg 60 repeating units that, themselves, are made up of two to six glycose resi- dues. The 0-specific chain is characteristic for a bacterial species and shows, on comparison of different Gram-negative bacteria, an enormous structural variability. In a given LPS preparation it exhibits heterogeneity because of the presence of various molecules differing in the number of repeating units. In the LPS of some nonenterobacterial pathogens, such as Neisseria and Haemophilus, the 0-specific chain is absent (22). The core oligosaccharide (23) may be divided into two regions, namely the outer core and the inner core. The outer core region of enterobacterial LPS, contains such neutral sugars as D-galactose (Galp), D-glucose (Glcp), 2-acetamido-2-deoxy-~-glu- cose (GlcpNAc), and 2-acetamido-2-deoxy-~-galactose (GalpNAc) and is therefore also called the hexose region. The inner core is mainly composed of the rare glycose residues L-glycero-D-manno-heptose (L,D-Hep) and 3- deoxy-D-manno-octulosonic acid (“2-keto-3-deoxy-~-manno-octonic acid,” Kdo), which often carry phosphate (6,23) or 2-aminoethyl- (pyro)phosphate [Etn-(P)P] (24). The structure of the core-oligosaccharide of some enterobacterial LPS [Salmonella minnesota, Escherichia coli, Pro- teus mirabilis, Citrobacterfreundii, and other bacterial genera (23)] has been elucidated and shown to be of moderate interbacterial variability.

Lipid A constitutes the covalently bound lipid component and the least variable component of LPS (25). It anchors LPS to the bacterial cell by hydrophobic and electrostatic forces and mediates or contributes to many of the functions and activities that LPS exerts in prokaryotic and eukaryotic organisms. In the following sections, the primary structure of lipid A of different Gram-negative bacteria is described, together with some of its characteristic biological properties. Furthermore, this article describes some of the principal methods that have been used for the structural analysis of lipid A and discusses their merits and limitations.

Lipid A

FIG. 1. -Schematic architecture of an enterobacterial lipopolysaccharide.

Page 236: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

MOLECULAR STRUCTURE OF LIPID A 213

11. LIPID A: DEFINITION AND GENERAL PROPERTIES Lipid A may be obtained by subjecting an LPS preparation to mild acid

hydrolysis, by which treatment the ketosidic linkage between the inner core oligosaccharide and lipid A, namely, between Kdo and the distal D - ~ ~ U C O S ~ - mine residue [GlcN (11)] of the lipid A backbone, is cleaved. Acid hydrolysis must be used, because enzymes that split the ketosidic bond, or mutants that synthesize oligosaccharide-free lipid A are known only for a few cases (26). The acid lability of the linkage between lipid A and the polysaccharide portion was first detected by Boivin and coworkers (27) in 1933, but the term lipid A was introduced only in 1954 by Westphal and Luderitz (28) to describe the hydrophobic and chloroform-soluble precipitate obtained after acid treatment of LPS and to distinguish it from another lipid (lipid B), investigated earlier by Morgan and Partridge (29). This lipid B turned out to be a noncovalently bound phospholipid that could be extracted into the organic layer from the LPS complex using ether or petroleum ether (28,30). Later, the term lipid A was also used for the designation of the bound lipid component as it is present in LPS. In order to differentiate between bound and isolated lipid A, it was proposed to restrict the term lipid A to the lipid component as it is present in LPS, and to use the term free lipid A for polysaccharide-deprived (or isolated) lipid A (2,4). Neither term, however, describes a homogeneous material having a chemically defined structure. As it is now known, lipid A harbors a certain structural heterogeneity that is either due to incomplete biosynthesis or generated by chemical degradation during the preparation of free lipid A by acid hydrolysis (3 1). Therefore, the term lipid A designates, rather, a class of structurally related molecules that are constructed according to a common architectural principle.

Despite the enormous progress made in the analytical and synthetic chemistry of lipid A, the complete structural elucidation of a lipid A prepa- ration still represents a challenge for analytical chemists. The reason for the difficulties encountered in analysis of lipid A are due, in part, to its amphi- philic nature, i.e., the presence of hydrophilic and hydrophobic regions fa- voring the formation of aggregates in either organic nonpolar or aqueous solution. This factor has, in the past, greatly limited the application of such advanced chromatographic purification procedures as adsorption-, ion-ex- change-, or reverse-phase high-performance liquid chromatography (h.p.1.c.). Moreover, molecular aggregates of lipid A and LPS preparations also limit the application of modem proton (lH), carbon ( 13C), and phos- phorus ( 31P) nuclear magnetic resonance (n.m.r.) spectroscopy. Further, the amphoteric character of lipid A, resulting from the simultaneous presence of negatively and positively charged groups as well as its compositional and structural microheterogeneity, has added to the problems associated with the chemical and physical analysis of lipid A.

Page 237: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

214 ULRICH ZAHRINGER et al.

Nevertheless, in the past decade, considerable progress has been made in this field. The most successful approach toward the isolation of homoge- neous lipid A and derivatives was the application of controlled degradation reactions and the introduction of specific protecting groups in combination with the development of various h.p.1.c. purification procedures (32 - 35). In addition, improvements in soft desorption ionization techniques, such as laser-desorption mass spectrometry (1.d. -m.s.) (36 - 40), fast-atom bom- bardment mass spectrometry (f.a.b. - m.s.) (32,33,4 l), and ZszCf-plasma de- sorption mass spectrometry (42 -44b), which have been found suitable also for nonderivatized macromolecules, have deepened our knowledge of the lipid A structure. Moreover, nondestructive IH-, I3C-, and 31P-n.m.r. spec- troscopy contributed tremendously to the progress made in the structural elucidation of various types of lipid A. Modifications of derivatization reac- tions for methylation analysis, in combination with coupled gas - liquid chromatography- mass spectrometry (g.1.c. - m.s.) or h.p.1.c. - m.s. have been of great help. Finally, the recent availability of various synthetic lipid A reference compounds and partial structures thereof has allowed, in most cases, an unequivocal interpretation of the results obtained with these em- pirical analytical procedures.

111. PRIMARY STRUCTURE OF LIPID A : BACKBONE, POLAR SUBSTITUENTS, AND FATTY ACIDS

Comparison of endotoxic lipid A of different Gram-negative genera and families reveals that they not only share chemical constituents but that they are also made up according to a similar architectural principle, which is exemplified by the well-studied lipid A of the Escherichia coli Re mutant strain F5 15 (Fig. 2, for literature compare Ref. 25). This E. coli lipid A is composed of a 2-amino-2-deoxy-6-0-(2-amino-2-deoxy-~-~-glucopyrano- sy1)-a-D-glucopyranose disaccharide [p-~-GlcpN-( 1 + 6)-a-~-GlcpN], which carries two phosphate groups: one in position 4’ [of the distal GlcpN residue, GlcN(II)] and one in the a-glycosylic position 1 [of the reducing GlcpN residue, GlcN( I)]. Of the hydroxyl groups present in this hydrophilic lipid A backbone, those in positions 4 and 6’ are not substituted. The latter primary hydroxyl group is only unsubstituted in free lipid A, because in LPS it represents the attachment site of Kdo, that is, the polysaccharide compo- nent (compare Fig. 2). Positions 3’, 2’, 3, and 2 of the backbone carry a total of four (R)-3-hydroxytetradecanoic acid residues [ 14:0(3-OH)]. As these are directly linked to the GlcpN disaccharide, they are termed primary acyl groups (45). The hydroxyl groups of the two 14 : O(3-OH) residues bound to GlcN(I1) are acylated by secondary fatty acids, that at position 2 by dode- canoic acid ( 12 : 0) and that at position 3’ by tetradecanoic acid ( 14 : 0). The

Page 238: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

MOLECULAR STRUCTURE OF LIPID A 215

- 0

P HO ’ 0, II ,o

FIG. 2.-Chemical structure of lipid A of the Escherichiu coli Re mutant strain F5 15. The hydroxyl group at position 6’ constitutes the attachment site of Kdo. The numbers in circles indicate the number of carbon atoms present in the fatty acyl chains. The 14 : 0(3-OH) residues possess the (R)-configuration. The glycosylic phosphate group may be substituted by a phos- phate group (see Table I) (46,65,69).

glycosylic phosphate at C- 1 may carry a further phosphate group in nonstoi- chiometric amounts, as is the case in E. coli K-12 (46). In addition to the main structure shown in Fig. 2, molecules having a smaller number of acyl groups are also present (47,48). The same structure as that of E. coli lipid A has been identified in S. typhimurium (32). The lipid A of E. coli was the first of a series of lipid A preparations that have also been obtained by total chemical synthesis (1 4,15).

The structural components encountered in E. coli are also present in lipid A of other bacterial sources. Thus, a survey of the structures analyzed shows that lipid A, in general, contain two gluco-configured and pyranosidic D- hexosamine residues (2-amino-2-deoxy-~-~ucose, GlcpN, or 2,3-diamino- 2,3-dideoxy-~-ghcose, GlcpN3N, also termed “DAG’ (49,50)], which are present as ap-( 1 + 6)-linked disaccharide (monosaccharide backbones have also been identified, but the respective lipid A lack endotoxic activity). The disaccharide is phosphorylated by one or, in most cases, two phosphate

Page 239: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

216 ULRICH ZAHRINGER et al.

residues, one in a-glycosylic linkage at position 1 and a second ester-bound to position 4’ of HexN( 11). Both phosphate groups may carry substituents that often contain (positively charged) amino groups. Primary fatty acids attached to the lipid A backbone are amide- and ester-linked and comprise, in general, medium- to long-chain (R)-3-hydroxy fatty acids (C Both ester- and amide-linked (R)-3-hydroxy fatty acids may be acylated at their 3-hydroxyl group to constitute (R)-3-acyloxyacyl residues.

As these results and Fig. 2 show, three structural components may be defined in lipid A: (i) the lipid A backbone consisting of a pyranosidic HexN disaccharide and phosphate groups, (ii) substituents of the backbone phos- phate residues (polar head groups), and (iii) fatty acids. Therefore, lipid A of different bacteria may be classified according to the nature of the backbone constituents (GlcpN or GlcpN3N), the type and nature of the polar head groups, and features of the acylation pattern. In a few instances, other back- bone substituents have been encountered. These will be described later in conjunction with individual lipid A forms.

In the following paragraphs, our present knowledge of these structural components and their significance for the lipid A architecture will be dis- cussed. With few exceptions, the chemical structure of these lipid A forms was determined by analyzing LPS derived from rough R-mutant bacteria. In contrast to wild-type (S) bacteria, these mutants form, because of defects in the synthesis, transfer, epimerization, or phosphorylation of glycosyl resi- dues, incomplete LPS (R-form LPS), consisting of lipid A and a truncated polysaccharide region.

1. The Lipid A Backbone

a. General Principles and Structures. - The lipid A backbone constitutes the hydrophilic portion of the amphiphilic lipid A. Structurally, the lipid A backbone is a bisphosphorylated p-( 1 6)-interlinked HexN disaccharide, the HexN residues being always present in the D-glucopyranosidic form. Two types of 2-amino-2-deoxy-~-glucopyranoses have thus far been en- countered as backbone constituents, namely GlcpN and GlcpN3N. Two phosphate groups are directly attached to the lipid A backbone, one in position 4’ [of HexN( II)] and one in the a-glycosidic position 1 [of HexN( I)]. As will be shown here, various lipid A backbone structures, isolated from taxonomically related but also remote bacteria, possess this common struc- tural principle, which comprises the most conservative region of the entire LPS molecule. The p-( 1 - 6)-interlinked HexpN disaccharide structure has thus far not been identified in other biomolecules such as glycoproteins, glycolipids, or glucans (5 1) and is thus unique to lipid A.

Page 240: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

MOLECULAR STRUCTURE OF LIPID A 217

b. Structural Elucidation of the Lipid A Backbone.-The presence of GlcpN and GlcpN phosphate in lipid A was demonstrated in studies (1,28,30,52) on enterobacterial LPS. In 1964, Burton and Carter (53) pro- vided evidence that free lipid A of E. coli contains a glycosidically linked GlcpN (phosphate) disaccharide. Gmeiner et al. (54) were the first to show, by using chemical degradation procedures (periodate oxidation), in combi- nation with chemical and enzymic analysis, that the lipid A backbone of a S. minnesota Re mutant (strain R595) was a (1 + 6) interlinked GlcpN disac- charide that had been previously synthesized by Foster and Horton (55). The first methylation analysis of the lipid A backbone was carried out by Adams and Singh (56), studying lipid A of Serratia marcescens. Also in this case, the GlcpN glycosidic linkage was found to be 1 + 6, and most probably p. A more-systematic degradation scheme for the isolation of the GlcpN disac- charide and application of methylation analysis, including g.1.c. - m.s., was first introduced and described by Hase and Rietschel(57,58). Theg.1.c. -m.s. analysis was further improved by Jensen et al. (59) with the help of synthetic reference compounds. Upon g.1.c. - m.s., the permethylated B-D-G~C~NAC- (1 + 6)-~-GlcNAc-ol could be distinguished from the a-anomeric isomer a-~-GlcpNAc-( 1 + 6)-~-GlcNAc-ol by its g.1.c. retention time. Moreover, the electron-impact (e.i.) fragmentation pattern of permethylated p-D- GlcpNAc-( 1 + 6)-~-GlcNAc-ol was characteristic for a 6-substituted

The first successful lH-n.m.r. analysis of the lipid A backbone was achieved with a purified dimethyl monophosphoryl lipid A derivative iso- lated from the E. coli Re mutant strain F5 15 (35). By using preparative thin-layer chromatography (t.1.c.) for the isolation of the lipid A structures, the authors also obtained evidence for heterogeneity relating to the degree of acylation of the lipid A backbone. Qureshi et al. (32) were the first to intro- duce the h.p.1.c. methodology for isolating homogeneous monophosphated lipid A derivatives from LPS of a S. typhimurium Re mutant strain (G30/ C2 1). H-N.m.r. analysis of a dimethyl pentatrimethylsilyl monophosphoryl lipid A derivative (60) revealed structural identity with E. coli lipid A (35).

A disadvantage of the derivatization procedure used in the case of S. typhimurium (60) was that the trimethylsilylated dimethyl monophosphoryl lipid A derivative was not suitable for the characterization of the (free) hydroxyl group in position 4. This identification could be achieved, how- ever, by the approach of Imoto et al. (33, who investigated the dimethyl monophosphoryl lipid A of E. coli and its per-0-acetylated derivative to compare the chemical shifts before and after acetylation. From the differ- ences in the chemical shift observed in lH-n.m.r., the authors could clearly demonstrate that, in their preparation, position 4 of GlcN(1) was free. All

GlcNAc-01.

Page 241: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

218 ULRICH ZAHRINGER et al.

other backbone hydroxyl groups showed no significant chemical-shift dif- ferences, indicating that the hydroxyl group at C-4 was the only free one in E. coli monophosphated lipid A. (The chemical shift of the primary hydroxyl group at C-6’ was not assigned and it could, therefore, not be determined as being free.)

The first 13C-n.m.r. analysis of the p-(1 -6) GlcpN disaccharide was camed out on the lipid A backbone ofE. coli (6 1). However, the assignments in this study were different from those obtained by Krasikova et al. (62) who synthesized the disaccharide p-~-GlcpN-( 1 - 6)-~-GlcpN and used it as a reference compound. I3C-N.m.r. was also used to identify the lipid A back- bone of S. typhimurium (63), S. minnesota (64,64a), E. coli (34,64a, 65,66,66a), Proteus mirabilis (67), and Haemophilus injluenzae (68).

c. Glycosyl Region of the Backbone.-The lipid A backbone contains phosphate and glycosyl residues, the latter composed of the gluco-configured D-hexosamines GlcpN and GlcpN3N. In the following, types of lipid A backbones are described that are classified according to the nature of the constituent hexosamine are described.

(9 Lipid A Containing a GlcpN- GlcpN Disaccharide Backbone. - In addition to its presence in LPS of E. coli (57,69), S. minnesota (54,57,70), and S. typhimurium (60), thep-D-GlcpN-( 1 + 6)-~-GlcpN disaccharide was also identified as the lipid A backbone structure in many other enteric and nonenteric Gram-negative bacteria. These include the Enterobacteriaceae Yersinia enterocolytica (7 I ) , Y. pseudotuberculosis (62), S. marcescens (56), P. mirabilis (67,72), Shigella jlexneri (57) and Enterobacter agglomerans (72a,72b); the Neisseriaceae Neisseria gonorrhoeae (33), N. meningitidis (73,74), and Acinetobacter calcoaceticus (75); the Pseudomonadaceae Pseu- domonas aeruginosa (76,77a) and Xanthomonas sinensis (78); the Bacteroi- daceae Bacteroides fragilis (79), Fusobacterium nucleatum (80) and Por- phyromonas (Bacteroides) gingivalis (80a); the Rhizobiazeae Rhizobium trifolii (8 1) and R. meliloti (82), and members of families such as Haemophi- lus injluenzae (68), Vibrio parahaemolyticus (83), Rhodocyclus gelatinosus (84), Chromobacterium violaceum (85), Vibrio cholerae (86), Rhodopseudo- monas gelatinosa, Rhodobacter (formerly Rhodopseudomonas) sphaeroides (87,88), Rhodobacter capsulatus (89), Rhodospirillum tenue (90), Rhodomi- crobium vannielii (9 I), Sphaerotilus natans (92), Bordetella pertussis (93), Actinobacillus actinomycetemcomitans (94), Erwinia carotovora (95) and others ( 1,4). Moreover, the p-( 1 - 6) linked GlcpN disaccharide has been identified in LPS of wild-type strains (S-form LPS) of, for example, S. min- nesota, E. coli 086, E. coli 0 1 1 1, and x. sinensis, indicating that this lipid A backbone structure is present (57) in both S-form and R-form LPS.

In addition to a p-( 1 - 6) GlcpN disaccharide, an a-( 1 + 6) GlcpN disac-

Page 242: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

MOLECULAR STRUCTURE OF LIPID A 219

charide backbone has been postulated (on the basis of g.1.c. -m.s. investiga- tions) as also being present in an R-mutant (R4) of P. mirabilis (67), although the p-( 1 + 6)-linked GlcpN backbone predominated over the a-( 1 - 6)-linked GlcpN in the molar ratio of 8 : 1. Thus far, no other bacte- ria, including other P. mirabilis strains, have been identified as containing an a-( 1 - 6) GlcpN disaccharide as a lipid A backbone constituent. It, there- fore, remains to be established whether this backbone structure is unique to this particular mutant, whether it has been overlooked in other bacteria, or whether it constitutes an artifact formed during degradation or derivatiza- tion procedures.

(ii) Lipid A Containing a GlcpN3N-GlcpN Disaccharide Backbone. - The diaminohexose 2,3-diamino-2,3-dideoxy-~-glucopyranose (GlcpN3N) was not known to occur naturally until 1975, when it was identified as a constituent of the lipid A of Rhodopseudomonas viridis and Rhodopseudo- monas palustris LPS (96). The GlcpN3N component replaces completely (see later) or partially the GlcpN residues of the backbone, as has been suggested by analytical data (49). Lipid A containing both GlcpN and GlcpN3N have previously been termed “mixed lipid A” (49,50).

Figure 3 shows this glycose residue as a component of the backbone of Campylobacter jejuni lipid A, which thus far is the most thoroughly investi- gated representative of a GlcpN3N-containing, disaccharidic lipid A back- bone (97). In the major lipid A component of C. jejuni, GlcpN3N position- ally replaces the GlcN( 11) residue of the classical p-~-GlcpN-( 1 - 6 ) - ~ - GlcpN structure discussed in the previous section. The HexN disaccharide is p-( 1 - 6)-linked, and similarities to the E. coli lipid A structure with regard to the location of phosphate and acyl groups are also evident (Fig. 2). The lipid A backbone of C. jejuni, however, is heterogeneous with respect to the number of GlcpN and GlcpN3N residues present in the disaccharide struc- ture. Three types out of the four theoretically possible combinations were identified: ( i ) the classical p-~-GlcpN-( 1 - 6)-~-GlcpN lipid A backbone disaccharide, (ii) a p-~-GlcpN3N-( 1 - 6)-~-GlcpN disaccharide, and (iii) a p-~-GlcpN3N-( 1 - 6)-~-GlcpN3N disaccharide. These structures are present in a molar ratio of approximately 1 : 6 : 1, the GlcpN3N-GlcpN hy- brid backbone, therefore, predominates (97). This type of hybrid backbone is also present in Brucella and in such phototrophic microorganisms as the Chromatiaceae (98). Thus far, a lipid A in which the backbone disaccharide contains GlcpN3N at the reducing and GlcpN at the nonreducing position has not been identified. A lipid A composed of GlcpN3N and GlcpN was also identified in Thiobacillus ferrooxidans and T. thiooxidans (99). In the case of P. carboxydovoruns (loo), which contains GlcpN3N, the lipid A structure was not elaborated [for further details, see also Ref. (lol)].

Page 243: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

220 ULRICH ZAHRINGER et al.

FIG. 3. -Chemical structure of the major component of Campylobacter jejuni lipid A. For details see the text. See also the legend to Fig. 2. For substituents of the phosphate groups see Table I. The a-anomenc phosphate has been tentatively assigned (97).

(iii) Lipid A Containing a GlcpN3N-GlcpN3N Disaccharide Backbone. -In Pseudomonas diminuta lipid A (102), a GlcpN3N disaccharide was identified as constituting the lipid A backbone (103). The nature of the glycosidic linkage of the disaccharide has been postulated to 8, but only on the basis of serological cross-reactivity with Salmonella lipid A. It was, thus, concluded that the disaccharide wasp-( 1 + 6)-linked and that at position 4’ of the distal GlcpN3N residue a phosphate group is present, whereas the nature of the substituent at the glycosidic position was not identified. As noted before in C. jejuni lipid A (Fig. 3), a minor fraction contains a p-( 1 - 6)-linked GlcpN3N disaccharide (97). In the lipid A backbone of Caulo- bacter crescentus GlcN could not be detected. It is possible, however, that a GlcpN3N-GlcpN3N disaccharide is present ( 103a).

( iv ) Lipid A Containing a GlcpN3N Monosaccharide Backbone.- As a constituent of lipid A, GlcpN3N was first detected in R. viridis (96,104), where it is present as a nonphosphated bis-N,N-acylated monosaccharide. A monosaccharidic GlcpN3N residue forms also the lipid A backbone in R.

Page 244: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

MOLECULAR STRUCTURE OF LIPID A 22 1

palustris (96), Pseudomonas diminuta (102), and Phenylobacterium immo- bile (105). Lipid A containing exclusively a GlcpN monosaccharide has not been identified thus far. In this context, it should be pointed out that the p-glycosidic linkage of the N-acylated lipid A backbone lacking the 4’-phos- phate is very susceptible toward the acid hydrolysis that is employed to isolate lipid A (69). The possibility must, therefore, be kept in mind that lipid-A-derived monomeric HexN structures may also result from acid treatment of LPS.

( v ) Lipid A Containing Other Glycosyl Backbones.-In a recent study (1 06) it was claimed that, in R. trifolii, D-glucosaminuronic acid (GlcpAN), carrying amide-linked 27-hydroxyoctacosanoic acid [28 : 0(27-OH)], com- prises the lipid A backbone and not a p-( 1 + 6)-linked GlcpN disaccharide as had been found previously (81). Whether the reported GlcpAN residue constitutes the lipid A backbone or whether it is present in addition to the GlcpN-containing disaccharide remains to be elaborated. Recently, Bhat et al. (107) reported the existence of three different types of lipid A backbones in the LPS of members of the family Rhizobiazeae, these being those con- taining only GlcpN (R. meliloti and R. fredii), a second type containing a GlcpN-GalA disaccharide (R. leguminosarum), and a third type containing GlcpN3N either in the form of a monosaccharide or as a hybrid disaccharide together with GlcpN (Bradirhizobium spp.). The detailed structures of these lipid A backbones await further analysis.

( v f i Conclusions.-In summary, the general structure of the lipid A backbone disaccharide is represented by a bisphosphated p-( 1 + 6)- interlinked HexpN disaccharide having GlcpN or GlcpN3N as glycosyl components. GlcpN disaccharides having a p-glycosidic linkage are also present in other natural molecules such as glycoproteins (5 I), the bacterial murein (peptidoglycan), and chitin (108). In these examples, however, the disaccharide linkage isp-( 1 - 4). To our best knowledge, ap-( 1 + 6) linked GlcpN or GlcpN3N linkage is not present in other biomolecules, and conse- quently, this structure appears to be unique and characteristic for one, namely, lipid A. Lipid A containing monosaccharidic GlcpN3N (but not GlcpN) has also been encountered. In these instances, however, further structural studies are indicated to establish details of these lipid A structures.

d. Phosphate Groups of the Backbone. - The backbone of disaccharidic lipid A contains, in general, two phosphate groups. Of these, one is a-linked to the glycosylic hydroxyl group at C- 1, and the other is ester-bound to the hydroxyl group at position C-4’ of GlcN( 11).

(i) The Glycosylic Phosphate (la-P).-As investigations of the lipid A

Page 245: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

222 ULRICH ZAHRINGER et al.

structure were often carried out using monophosphoryl lipid A, which lacks the glycosylic phosphate, the anomeric configuration of GlcN( I) remained unknown for a long time. In 1982, however, Redmond and colleagues (109) showed by 'H- and 31P-n.m.r. spectroscopy that, in the Re LPS ofS. minne- sotu R595, the phosphate residue attached to GlcN(1) is a-glycosylically linked (Fig. 4). This was also shown by I3C-n.m.r. spectroscopy to be true for an Re mutant of E. coli (66), for an 0-deacylated and per-0-acetylated derivative of Re LPS of E. coli F5 15 (34) and for the E. coli mutant D3 1 m4 (1 10). Indirect evidence for the a- 1 -phosphate in B. pertussis was obtained by Caroff et al. (1 1 l), who synthesized the 2-deoxy-2-[(R)-3-hydroxytetra- decanamidol-a- and P-D-glucopyranose 1 -phosphates. By comparing the (different) rates of phosphate release during acid-catalyzed hydrolysis of the synthetic a- and P-glycosylically bound phosphate on the one hand and bacterial lipid A on the other, the anomeric configuration of GlcN(1) in B. pertussis lipid A was determined to be a. In later investigations dealing with lipid A of C. jejuni (97), the 1-phosphate was determined by a similar method also to be a. Further, in lipid A of H. influenzue, the glycosylic 1-phosphate was found to be a, as determined by 'H- and '3C-n.m.r. spectroscopy (68).

FIG. 4. -Chemical structure of lipid A of the Salmonella minnesota Re mutant strain R595. For details see the text. See also the legend to Fig. 2. For substituents ofthe phosphate groups see Table I.

Page 246: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

MOLECULAR STRUCTURE OF LIPID A 223

(ig The Non-Glycosylic Phosphate (4 ’4 . -A nonglycosylic (ester- bound) phosphate, linked to GlcN(II), was detected in acid hydrolyzates of lipid A (52). From a study on deacylated S. minnesota Re LPS, it was determined (1 12) that phosphate was attached to the hydroxyl group at C-4’ of the lipid A backbone disaccharide (Fig. 4). This investigation showed that hydrazinolysis of S. rninnesota Re LPS followed by mild acid hydrolysis yielded the phosphorylated oligosaccharide P-4’-GlcpN-P-( 1 - 6)-GlcpN and, after N-acetylation, P-4’-GlcpNAc-P-( 1 - 6)-GlcpNAc as well as the nonphosphated GlcpN-P-( 1 4 6)-GlcpN and GlcpNAc-P-( 1 + 6)- GlcpNAc disaccharides, respectively, which were purified and isolated by preparative high-voltage paper electrophoresis (h.v.p.e.) ( 1 12). Subsequent to periodate oxidation of the lipid A backbone derivatives, the nonreducing GlcpN in P-4’-GlcpNAc-p-( 1 - 6)-GlcpNAc was found to be resistant to periodate oxidation, indicating that the phosphate group was linked to either position 3’ or position 4’. When the non-N-acetylated P-GlcpN-P-( 1 - 6)- GlcpN and GlcpN-p-( 1 - 6)-GlcpN disaccharides were similarly treated, GlcpN (and P-GlcpN) was quantitatively oxidized and no longer detectable, strongly indicating that position 4‘ of GlcN(I1) was the attachment site of phosphate. The first ”P-n.m.r. data (1 13) of various chemically degraded LPS from S. minnesota R595 were in accordance with these results. Per- iodate resistance of GlcN( 11) in nonmodified and N-acetylated P-4’-GlcpN- GlcpN was also shown for the lipid A disaccharide ofR. sphaeroides (87) and was explained on the basis of a phosphate group being present at position 4’. An elegant chemical approach toward the determination of the position of the ester-bound phosphate in a Re mutant of E. coli K-12 (69) comprised oxidation of 0-deacylated lipid A with Me,SO/Ac,O, whereby the primary hydroxyl group at C-6’ is oxidized to the aldehyde (hydroxyl groups at C-3’ and C-3 are also oxidized to keto groups). Alkaline conditions then caused the release of the phosphate (in the form of inorganic phosphate) from position 4’ via a p-elimination reaction . The application of ‘H- and 31P- n.m.r. spectroscopy, measuring selective proton - proton coupling constants by polarization transfer from the proton (H-4’) to phosphorus, also proved the location of phosphate at position 4’ in the lipid A of S. minnesota R595 (1 09).

Another approach was devised by Helander et al. (68): LPS of adeep rough mutant of H. inJluenzae (strain 1-69 Rd-/b+) was methanolyzed (2 MHCI- MeOH, 2 h,86 “C) and permethylated. The main products incorporating both GlcpN and phosphate were identified by g.1.c. - m.s. analysis as pyrano- sidic GlcpN 4’-phosphate carrying a 3-0-methyltetradecanoic acid residue by the characteristic fragmentation pattern in the electron impact mass spectrum (e.i. - m.s.). The same procedure was successfully applied to P. aeruginosa (mutant PAC605), which also contains a phosphate group at

Page 247: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

224 ULRICH ZAHFUNGER et al.

FIG. 5. -Chemical structure of the main component of lipid A of Bacteroides fragilis NCTC 9434 LPS. For substituents ofthe phosphate groups see Table I. The anomeric configuration of the GlcN(1) has been tentatively assigned as (Y (79).

position 4’ of GlcN( 11) (77). As an advantage of this latter approach, not only the position of phosphate but also the nature of fatty acid, amide-bound to the nonreducing GlcpN, may be determined in the same experiment.

For the determination of the phosphate group a new procedure was estab- lished (64a) using a two-step deacylation of the rough form LPS (E. coli F5 15 and S. minnesota R595) and h.p.1.c. purification of the intact core-backbone oligosaccharide. Data obtained by lH-, 13C-, and 31P-n.m.r. spectroscopy as well as f.a.b.-m.s. analysis unequivocally showed the presence of 4’-phos- phate at GlcN (11). A similar procedure was also applied to E. coli J5 LPS (66a).

Although the phosphate group bound to position 4’ of HexN(I1) is a general lipid A constituent, examples are known where it is lacking. Thus, in B. fragilis (79) (Fig. 5.) , the hydroxyl group in position 4’ is free and in R. vannielii (9 l), the phosphate group in position 4’ is positionally replaced by a P-linked D-mannose residue. The ester-linked phosphate group is also lack- ing in other lipids A, including Chromatiurn vinosum (1 14), Selenomonas ruminantium (1 1 9 , and Thiobacillus species (99) (for further examples, compare Refs. 49 and 50).

Page 248: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

MOLECULAR STRUCTURE OF LIPID A 225

(iii) Conclusions. -The p-( 1 + 6)-linked HexpN disaccharide of lipid A carries, in general, two phosphate groups. The glycosylic phosphate group is a-linked to C- 1 of HexN(1) and the nonglycosylic phosphate group is ester- bound to position 4' of HexN(I1). All disaccharide-containing lipid A carry the a-glycoslyic phosphate residue, whereas the nonglycoslyic phosphate is lacking in lipid A of certain Gram-negative bacteria. These latter prepara- tions are generally of low endotoxic activity (9 1,l I6 - I 18)

2. Phosphate Substitutents

Both the glycosylic and the nonglycosylic phosphate residues may be substituted, in general, by nonacylated, charged glycosyl or nonglycosyl residues. These polar head-groups are often not present in stoichiometric amounts, leading to a certain intrinsic heterogeneity of lipid A. It should be noted that, in Legionellu pneurnophilu (1 19) and H. influenzue (120), glyc- erol phosphate was identified as a constituent of LPS or lipid A, the location of which, however, was not studied. Some of the substituents have been structurally elucidated and their nature and location are summarized in Table I.

TABLE I Phosphate-Linked Substituents of the Lipid A Backbone

(Y-P-FD-GlcpN-(l+ 6>a-D-Gl~pN-l-P)

Substituent of backbone phosphate group at

Organism C-4' c-1 Reference

Escherichia coli Salmonella minnesota Proteus mirabilis Yersinia pestis Vibrio cholerae Neisseria meningitidis Chromobacterium violaceurn Campylobacter jejuni Pseudomonas aeruginosa Haemophilus influenzae Rhodospirillum tenue Rhodobacter capsulatus Sphaerotilus natans Moraxella catarrhalis

- L-Arap4N" ~-Arap4N L-Arap4N

P-Etna ~-Arap4N P-Etn"

~-Arap4N P-Etn* P-Etn P

pa .b

P-Etna.= - D-Araf" P-Etn P-Etna GlcpN P-Etn

- D-Anf P-Etna

P-Etna

4 4 4

121 86 73 85 97 77 68 90 89 92

193b

a Nonstoichiometric.

cEtn, 2-aminoethanol. P, phosphate.

Page 249: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

226 ULRICH ZAHRINGER et al.

a. Substituents at the Non-Glycosylic 4’-Phosphate. - Substitution of the 4’-phosphate was, for the first time, identified in lipid A of C. violaceum (8 5), where 4-amino-4-deoxy-~-arabinopyranose (~-Arap4N) quantitatively substitutes the 4’-phosphate of GlcN( 11). This is also the case in lipid A of P. mirabilis (72) and Y. pestis (121). In S. minnesota Re LPS (Fig. 4), the 4’-phosphate of GlcN( 11) is substituted by this aminopentose, however, in a nonstoichiometric manner (85,113). The L-Arap4N linked to the phosphate at position 4’ is released from (hydrazine-treated) LPS under mild alkaline conditions (0.2 MNaOH, 1 h, l0O’C;) in the form of its 1-phosphate (85), and concomitantly with the release of P-~-Arap4N 1-phosphate, the 4’- phosphate group is removed from the lipid A backbone. In S. minnesota Re LPS, the glycoslyic linkage of ~-Arap4N to lipid A was determined, by IH-n.m.r. (109), to be p. In Rhodobacter capsulatus (89), C. jejuni (97), S. natans (92), and N. meningitidis (73), 2-aminoethyl phosphate (Etn-P) is bound to the nonglycosylic phosphate residue (Table I). In the case of N. meningitidis, the substitution is nonstoichiometric. As Fig. 6 shows, out of four conceivable lipid A species, the major one carries 2-aminoethyl pyro- phosphate at positions 4’ and 1, and a minor one contains monophosphate groups at these positions.

Studying a lipid A precursor isolated from a temperature-sensitive mutant of S. typhimurium (STiSO), Strain et al. (63), by applying ‘H- and 31P-n.m.r., found that L-Arap4N was linked to the glycosylic phosphate at C-1 and 2-aminoethyl phosphate (Etn-P) was linked to position 4’ ofGlcN( 11). Thus, the polar head-groups in the lipid A precursor produced by this mutant are at opposite locations, as previously found for a structurally similar (“neutral”) precursor (1 22) and for lipid A of, for instance, S. minnesota (109,113). At present it is not known whether this discrepancy is due to differences in the methodology used or whether the substitution pattern in the precursor of S. typhimurium is different from that of mature lipid A of the bacterial strains investigated previously.

b. Substituents of the Glycosyl a-Phosphate. - Either the substituents of the phosphate glycosylically linked to GlcN(1) are neutral or they carry

FIG. 6.-Chemical structure of the lipid A backbone of Neisseria meningitidis M986 LPS having polar substituents. The nonglycosylic and glycosylic phosphate groups are substituted by Etn-P to a similar degree (85 vs SO%, w/w). The anomeric configuration of the phosphate has been assigned tentatively as a. R’ = 12:0(3-OH), RZ = 14-0[12:0(3-OH)] (73).

Page 250: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

MOLECULAR STRUCTURE OF LIPID A 227

positive or negative charges. A neutral substituent is present in R. tenue (90), where D-arabinofuranose (D-Araf ) is glycosylically linked to the phosphate group at C- 1. In E. coli, phosphate may be substituted the glycosylic phos- phate, resulting in a pyrophosphate group at C- 1 (69). As a positively charged group, nonacylated GlcpN was identified in C. violaceum (85) as being linked to the glycosylic phosphate. In C. jejuni, an Etn group, and in S. minnesota (Fig. 4), V. cholerae, R. capsulatus, and N. meningitidis (Fig. 6) an Etn-P group are linked to the C-1 phosphate (Table I). The location of these residues at the glycosylic phosphate has also been studied in a precursor of lipid A biosynthesis, however, as just discussed, with discrepant results.

c. Conclusions.-Both phosphate groups of lipid A at C-1 and C-4’ may carry (polar) substituents, although examples in which the phosphate groups are free are known. The substitution of phosphate groups is often nonstoi- chiometric, as exemplified with N. meningitidis lipid A (Fig. 6). In this interesting example the glycosylic position [GlcN( I)] and the hydroxyl group at C-4’ [GlcN( 11)] both carry Etn-PP. This substitution pattern is symmetric, and careful chemical and 31P-n.m.r. analyses have shown (73) that, in the lipid A part of this LPS, both phosphate groups at positions 4’ and 1 are nonstoichiometrically substituted by Etn-P to a similar degree (85 vs 80%). In lipid A produced with sodium acetate buffer (0.1 M NaOAc, pH 4.5, 1 h, 100°C) the degree of Etn-P substitution at position 4’ (73%, w/w) is slightly lower, whereas substitution at C-1 is decreased to one half (45%, w/w) com- pared with native LPS (80%, w/w). This indicates that the pyrophosphate group of Etn-PP at C-4’ is resistant to mild acid hydrolysis, whereas that of the a-glycosylically linked Etn-PP group at C- 1 is susceptible to even mild acid hydrolysis.

The extent of phosphate substitution is influenced by, for instance, the growth conditions ( 122 - 124). It is possible that bacteria, depending on their physiological demands, are able to add or omit ionic head-groups and thereby regulate their net surface charge (1 13). It is noteworthy that most of the substituents on the phosphate, such as Etn, L-Arap4N, and GlcpN carry, at neutral pH, a positively charged amino group. Their presence in the neighborhood of phosphate residues (or of Kdo in LPS) may be considered as a regulating factor, controlling the electrostatic interaction of negatively charged residues (phosphate) with such bivalent cations as CaZ+ or MgZ+ of the medium. The Ca2+ and Mgz+ ions may neutralize intermolecularly negative charges of lipid A. These interactions, therefore, appear to be of great importance for the stability and function of the bacterial outer mem- brane. Moreover, it has been postulated that positively charged groups in lipid A, such as L-Arap4N, significantly contribute to the resistance ofcertain Gram-negative bacteria (for instance P. mirabilis) to such antibiotics as polymyxin B ( 125). In accordance with this concept, P. mirabilis mutants lacking ~-Arap4N are highly sensitive toward polymyxin B (126).

Page 251: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

TABLE I1 Distribution of Acyl and 3-Acyloxyacyl Residues between Hydroxyl and Amino Groups of the Classical Lipid A Backbone

I&&lcN(II)-(l- 6)-&lcN (I)] of Various Bacteria

Nature and linkage type of acyl sobstitwots of the lipid A backbone bound to

GlcN (II) at position GIcN (I) at position

Bacteria 3‘ (ester) 2’ (nmide) 3 (ester) 2 (amide) Reference

Escherichia coli Salmonella typhimurium Salmonella minnesota

Envinia carotovora Proteus mirabilis

Haemophilus infuenzae Neisseria gonorrhmae Neisseria meningitidis Pseudomonas aeruginosa

Bacteroides fragilis

Rhodocylclus gelatinosus

Rhodobacter sphaeroides

Rhodobacter capsulaus Sphaerotilus natans Actinobacillus

actinomycetemcomitans Moraxella caiarrhalis Porphyromonas gingivalis Enterobacter agglommans

14 : 0[3-q14 : O)] 14 : 0[3-q 14 : O)]

14 : 0[3-q 14 : O)] 14:0[3-q14:0)]

14 : 013-q 14 : O)]

14 : 0[3-0(14 :O)p 14:0[3-0[14:0(2-0H)]’]

12 : 0(3-OH) 12 : 0(3-OH) 10:0(3-0H)

16:0(3-0H)” 15:0(3-OH)” 10 :0(3-OH)

10 : 0(3-0H)

10 : 0[3-0(A5- 12 : I)] 10:0(3-OH)

14 : 0[3-q14 : O)] 12:0(3-0H) H 140[3-o[14:0(2-0H)]]d

14 :q3-q12 : O)] 14 : 0[3-q 12 : O)] 14 : 0[3-q 12 : O)]

14 : 0[3-q12 : O)] 14 : 0[3-q14 : O)]

14 : 0[3-q 14 : O)] 14 : 0[3-q 12 : O)] 14 : O[ 3 - q 12 : O)] 12 : 0[3-0(12 : 0)]’ 12 : 0[3-OIl2: 0(2-0H)])” 15-Msl6 : 0[3-O[ I3-Me-14:0]]’ 16 : 0[3-0( 13-Met14 : 0]’ 10: 0[3-q12 : O)]

14:0[3-0(A7-14: 1)p 14 : 0[3-0( 14 : 0)]’

10: 0[3-0(12 : 0)p 14 : o(3-0~0)

14 : 0[3-q 14 : O)] n o [ 3 - q 100))

140[3-q120)] 15-Me-16:0[3-0(15:0)]

14 : 0(3-0H) 14:0(3-0H) 14 :0(3-OH)

14 : O(3-0H) 14 : 0(3-0H)

14: 0(3-0H) 12 : q3-OH) 12 : 0(3-OH)

14:0(3-0(16 :O)lb

10 : 0(3-OH)b

15:0(3-OH)”

10 : O(3-OH)

10:0(3-0H)

10:0(3-OH) 10:0(3-OH)

14: 0(3-0H)

H 140(3-OH)

16:0(3-0H)”

120[3-q loo)]

14: 0(3-0H) 14 : 0(3-0H) 14 : 0(3-0H)

14 : 0(3-OH) 14 : 0(3-0H) 14: 0[3-0(16 : O)]’ 14 : 0(3-OH) 14 : 0[3-q 12 : O)] 14 : 0[3-0(12 : O)] 12 : 0[3-0( 12 : 0)p

16: 0(3-OH)” 15-Me-I6 : 0(3-OH)” 10: 0[3-0( 12 : 0)p 10:0[3-0(14 :O)p 14 : o(3-0~0) 14: 0(3-0H)

12 :0[3-0[12:0(2-0H)]p

14:0(3-0XO) 10 : 0[3-0(12 : 0)p

14 : 0(3-0H)

15-Ma16 :0(3-OH) 12:0[3-q 12:0)]

14 : O[ 3-o( 16 : O)]

4 32,60

37,181 95 12

68 33 35 71

19

84

87,88

89 92

94 196a 80a 51a

a Fatty acids in one column add up to 1 mol eq of lipid A. b b n t in nonstoichiomctric amounts.

”Position of the hydroxy p u p (2-0H or 3-0H) was not daermined; intrinsic hetnogcneity. I40 (2-0H) may be nplaced by 140. Intrinsinc h-neity, I2 :O may be replaced by 10:0,10: I, 12 : 1, 14:0,14: 1 ester linked to 10:0(3-0H).

Page 252: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

MOLECULAR STRUCTURE OF LIPID A 229

3. Fatty Acids

a. General. -As a characteristic feature, lipid A contains saturated me- dium- to long-chain ( 10 - 22 carbon atoms) fatty acids in ester and amide linkages. These fatty acids comprise nonhydroxylated and hydroxylated ones ofwhich 60 - 75% are 3-hydroxylated. Thus, 3-hydroxylated fatty acids usually preponderate within the fatty acid profile of lipid A. The fatty acid profile of lipid A differs significantly from that of other bacterial phospho- lipids (1 27). Fatty acids may be linked directly to hydroxyl and amino groups of the lipid A backbone. These are termedprimary (45) acyl groups and they include 3-hydroxy and 3-keto fatty acids. Fatty acids may also be linked to the 3-hydroxyl groups of the ester and amide-linked 3-hydroxy fatty acids. These are termed secondary acyl residues, and they include nonhydroxy- lated (normal) and 2-hydroxylated fatty acids.

b. Nature of Fatty Acids. - (9 (R)-3-Hydroxylated Fatty Acids. - 3-Hydroxylated fatty acids constitute common and obligatory constituents of LPS, namely, their lipid A component. The 3-hydroxy (or P-hydroxy) fatty acids of lipid A possess, without exception, the (R)-configuration (1 28,129). The (R)-3-hydroxy fatty acids of different bacteria, either amide- or ester-bound, have been found generally to be saturated straight-chain and even-numbered and to have a number of carbon atoms between 10 [e.g., in P. aeruginosa lipid A (77,130,13 l)] and 22 [e.g., in Chlamydia trachornatis (1 32) and C. psittaci (1 33) lipid A]. In Veillonella, however, and some other bacteria, even-numbered 3-hydroxy fatty acids are present ( 134). Of these 3-OH acyl groups, (R)-3-hydroxytetradecanoic acid [ 14 : 0(3-OH)] was identified in about two thirds of all bacteria investigated so far (127). As a general rule, (R)-3-hydroxy fatty acids are directly bound to the amino or hydroxyl groups of the lipid A backbone disaccharide (positions 3’,2’,3, and 2) and are, therefore, designated as primary lipid A fatty acids (45). 3-Hy- droxylated fatty acids as substituents of an amide-bound 3-hydroxy fatty acid (3-acyloxyacyl residue in amide linkage) have not been identified thus far (38). However, an ester-bound 3-acyloxyacyl residue in which both fatty acids are (R)-3-hydroxylated [ V. cholerae (86)] was identified. In general, as is the case in all Enterobacteriaceae, and in Helicobacter pylori (1 35,136), only one type of 3-hydroxy fatty acid is involved in amide linkage (Table 11, Figs. 2,4), but in a few instances (Bacteroides, Xanthomonas, Veillonella, and Myxobacteria) two different 3-hydroxy acids are amide-bound to the lipid A backbone. In B.fragilis, the acyl groups having the higher number of carbon atoms are linked to GlcN(I1) (79). In LPS of a number of intracellular Gram-negative bacteria such as Coxiella burnetii (1 37) and L. pneumophila ( 1 19), a large number ofdifferent amide-bound (R)-3-hydroxy fatty acids are present exclusively in amide-linkage, however, in unknown location.

Page 253: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

230 ULRICH ZAHRINGER et a1

In a few cases, 3-hydroxy fatty acids that contain additional functional groups were characterized. Thus (R)-3-hydroxydodec-(5Z)-enoic acid [A5- 12 : 1(3-OH)] was identified in ester linkage in lipid A of the chloridazon-de- grading bacterium P. immobile (105). Unsaturated 3-hydroxy fatty acids are also present in R. trifolii (81) and R. meliloti (82). In an extract of L. pneu- mophila, 2,3-&hydroxy- 12-methyltridecanoic acid [ 12-Me- 13 : 0(2,3- diOH)] and 2,3-dihydroxytetradecanoic acid [ 14 : 0(2,3-diOH)] were identi- fied in amide-linkage ( 138). It remains, however, to be established, whether these 2,3-dihydroxylated fatty acids are lipid A constituents. Small amounts of 2-methyl-3-hydroxy-fatty acids have been detected in B. pertussis (1 39).

The determination of (R)-3-hydroxylated fatty acids is of considerable chemotaxonomic value (30,127), and lipid A-linked 3-hydroxylated fatty acids are used as diagnostic markers for the qualitative and approximate quantitative determination of endotoxins in biological samples (140). For the analysis of 3-hydroxy fatty acids in very low concentration, they are first liberated by hydrolysis of LPS or lipid A, followed by modification of the carboxyl groups to methyl esters and of the 3-hydroxyl groups to the tri- methylsilyl ethers. Using g.1.c. -m.s., this approach was found to be suitable for the determination of endotoxin concentrations in the serum with a detection limit of 200 fmol3-hydroxy fatty acid/mL (140). The concept of using 3-hydroxylated fatty acids for the detection of endotoxin in serum (140), peritoneal fluid and plasma (141), or organs of higher animals is further justified by the fact that the amount and type of @)-3-hydroxylated fatty acids present in lipid A are not altered by changing the growth condi- tions (temperature, medium, and aeration), as has been shown for P. mira- bilis, S. minnesota, E. coli (37,142,143), P. aeruginosa (144), and Y. entero- litica (145). Moreover, some bacteria, such as B. frasilis, express characteristic and diagnostic hydroxy fatty acids [ 15-Me-16 : 0(3-OH)]. The presence of (R)-3-hydroxylated fatty acids in all lipid A investigated thus far shows that they constitute a highly conserved structural feature of LPS.

It should be mentioned that, although very characteristic, (R)-3-hydroxyl- ated fatty acids are not present exclusively in lipid A. They have also been identified in such other lipids as the ornithine-containing lipids of various Gram-negative bacteria. Ornithine lipids having (R)-3-hydroxylated fatty acids of a chain length similar to that of lipid A-linked (R)-3-hydroxy fatty acids (C L4-C were found, for example, in B. pertussis, Gluconobacter, Acetobacter, Brucella, and Francisella (for details see Refs. 127, 146, and 147). (R)-3-Hydroxylated fatty acids are also present in the rhamnolipid of P. aeruginosa ( 148), in the fish poison pahutoxin ( 149), and in glycolipids of Rhodotorula (1 50).

(iq (S)-2-Hydroxylated Fatty Acids. - The 2-hydroxylated (a-hydrox- ylated) fatty acids encountered in lipid A possess, without exception, the

Page 254: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

MOLECULAR STRUCTURE OF LIPID A 23 1

(S)-configuration ( 129,15 1). They are not as common as 3-hydroxy fatty acids and are never substituted at their 2-hydroxyl group. Among the (S)-2- hydroxy fatty acids identified, (S)-2-hydroxydodecanoic [ 12 : 0(2-OH)] and (S)-2-hydroxytetradecanoic acid [ 14 : 0(2-OH)] predominate. Thus, 14 : O(2-OH) was found to be present in lipid A of Salmonella (Fig.4), Kleb- siella, and Serratia, but to be absent from LPS of Escherichia, Proteus, Enterobacter, Shigella, and Yersinia strains (15 1). On the other hand, 12 : O(2-OH) was identified in Pseudomonas (77,152,153), Acinetobacter (1 54), Bordetella (1 39), Chromobacterium (85) , and other genera.

Unlike 3-hydroxylated acyl groups, 2-hydroxy fatty acids are not bound directly to the lipid A backbone. They are present exclusively as secondary ester-linked fatty acids, attached to the hydroxyl group of an (R)-3-hydrox- ylated acyl residue, thus forming an acyloxyacyl group. These 2-hydroxy fatty acid-containing 3-acyloxyacyl groups are either amide- (C. violaceurn, P. aeruginosa, X. sinensis, and A. calcoaceticus) or ester-linked [S. minne- sota and A. calcoaceticus (38)J to the lipid A backbone.

In several instances (such as S. minnesota and P. aeruginosa), the amount of the 2-hydroxylated fatty acid and that of the nonhydroxylated fatty acid of the same chain length adds up to 1 mol per molecule of lipid A. [thus, in S. minnesota 14 : O(2-OH) and 14 : 0; in P. aeruginosa 12 : O(2-OH) and 12 : 01. As the 2-hydroxylated fatty acid occupies the same position in lipid A as the corresponding nonhydroxylated fatty acid, it appears that a-hydroxylation takes place at the level of fully acylated lipid A. As a consequence of partial a-hydroxylation, these lipid A express heterogeneity in that they contain at least two species, one containing the 2-hydroxylated and one containing the corresponding nonhydroxylated acyl group (26,77,15 1) (compare Fig. 4).

In P. aeruginosa PAO, the degree of hydroxylation, that is, lipid A hetero- geneity with respect to hydroxylated [ 12 : 0(2-OH)] and nonhydroxylated ( 12 : 0) fatty acids, was found to be mainly influenced by the growth temper- ature. With increasing temperature, the proportion of the nonhydroxylated fatty acids ( 12 : 0 and 16 : 0) was decreased at the expense of such hydroxyl- ated ones as 12 : O(2-OH) (1 44). A similar finding was made with Salmonella species ( 142).

It has previously been reported (1 5 5 ) that the lipid A of Pseudomonas paucimobilis (now termed Sphingomonas paucimobilis) contains an un- usual lipid A-type glycolipid having 14 : O(2-OH) as the only amide-linked fatty acid. A recent reinvestigation of this compound ( 156), however, re- vealed that it constitutes a glycosphingolipid containing (in addition to GlcpA, Galp, Manp, and GlcpN) 14 : O(2-OH) in amide linkage to the dihy- drosphingosine base. As our recent analyses show, S. paucimobilis bacteria are completely devoid of 3-hydroxylated fatty acids, and hence we hypothe- size, that, in this case, LPS is replaced by this very glycosphingolipid, which, interestingly, lacks endotoxic activity. Thus, S. paucirnobilis, together with

Page 255: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

232 ULRICH ZAHRINGER et al.

Borrelia burgdorferi (1 57), appears to constitute a group of microorganisms, which do not, in contrast to the majority of Gram-negative bacteria, require a typical LPS for growth and multiplication.

(iii) Other Hydroxylated Fatty Acids. - P. diminuta, P. vesicularis (1 58), and L. pneumophila (1 38) contain, in addition to 3-hydroxylated fatty acids, 5,9-dihydroxytetradecanoic acid. It is likely, but not unequivocally proven, that this fatty acid constitutes a lipid A component. 27-Hydroxyoc- tacosanoic acid [28 : 0(27-OH)] has been identified as a major fatty acid in LPS of R. trifolii (106), R. meliloti (82), and L. pneumophila (1 59). Using 1.d. - m.s. analysis of dephosphated lipid A, in combination with chemical analysis, 28 : O(27-OH) was determined to be ester-bound to the 3’-hydroxyl group of GlcN( 11) in R. meliloti. The 28 : O(27-OH) residue was also found in R. viridis, R. palustris, Nitrobacter winogratsky, Nitrobacter hamburgensis, Pseudomonas carboxydovorans, Bradirhizobium spp., Agrobacterium spp., and Thiobacillus spp., all being members (10 1,160) of the 16s rRNA sub- group a-2. Moreover, two 28 : O(27-OH) homologues, namely 26 : O(25-OH) and 29 : 0(28-OH), were identified in Bradirhizobium lupini (101) and R. palustris le5 (1 60), respectively, the latter having a branched structure. In L. pneumophila such (0-1)-0x0- and 1,o-dioic long-chain fatty acids as 27- oxooctacosanoic acid [28 : O(27-0xo)l and heptacosan- 1,27-&0ic acid [27 : O(dioic)] and their homologues 29-oxotriacontanoic acid [30 : O(29- oxo)] and nonacosan- 1,29-dioic acid [29 : O( 1,29)dioic] were identified (1 59). In other Legionella species, such as L. jordanis, L. rneceacherinii, and L. micdadei, such a-hydroxylated long-chain fatty acids as 2-hydroxy-27- 0x0-octacosanoic acid [28 : 0(2-OH,27-oxo)], 2-hydroxy-29-0x0-triacon- tanoic acid [ 30 : 0(2-OH,29-oxo)], 2-hydroxyheptacosane- 1,27-dioic acid [27 : O(2-OH)dioic], and 2-hydroxynonacosan-1,29-dioic acid [29 : O(2- OH)dioic] have been analyzed ( 16 1) in the phenol- chloroform - petroleum ether (PCP) extracts, indicating that they are constituents of lipopolysac- charide.

( iv) 3-0x0 (3-Keto) Fatty Acids.-As lipid A constituents, 3-ox0 fatty acids were first encountered in Mbrio anguillarum (D. Shaw, personal com- munication). Later it was shown that R. capsulatus contains 3-ketotetrade- canoic acid [ 14 : O(3-0xo)l in amide linkage (Fig. 7). As shown in Fig. 8, R. sphaeroides also carries at GlcN(1) a 14: o(3-0x0) group in amide linkage whereas 14:0(3-OH) is amide-bound to GlcN(I1) (50,87,88,162). As a pe- culiar feature, this lipid A contains the unsaturated fatty acid (Z)-A7-tetra- decenoic acid (A7-14 : l), which is present as a 3-(7-tetradecenoyl)oxytetra- decanoic acid (14 : O[ 3-O(A7- 14 : l)]) group, amide-bound to GlcN( 11) (87,88,162a). Unsaturated and 3-keto fatty acids are rarely encountered in

Page 256: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

0 -0,II 0

P’ HO’

0

.P II ,o-

‘OH

FIG. 7. -Chemical structure of lipid A of Rhodobucter capsulatus. Dashed lines indicate nonstoichiometric substitution. The anomeric a! configuration of phosphates and the configu- ration(ZorE)ofthedoublebondinA5-12: 1 areassignedonly tentatively(89). Forsubstituents of the phosphates see Table I.

0 II,o-

OH p \

FIG. 8.-Chemical structure of lipid A of Rhodobucter sphueroides (87,88,164). For details see the text and the legend to Fig. 6.

233

Page 257: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

234 ULRICH ZAHRINGER et al.

lipid A, and the acylation pattern of R. capsulatus and R. sphaeroides has, therefore, to be considered unusual. A 14 : O(3-0x0) residue was also found in the lipid A of Paracoccus denitrificans (163). Interestingly, those lipid A containing 3-ox0 fatty acids (R. sphaeroides, R. capsulatus, and P. denitrifi- cans) also contain unsaturated ( 12 : 1 or 14 : 1) acyl groups. Thus far, 14 : O( 3- 0x0) is the only 3-keto fatty acid identified in lipid A, where it is always present in amide linkage (87,88,164).

Lipid A of R. sphaeroides exhibits, compared with enterobacterial lipid A, very low toxicity (87,162,164- 166), and it was speculated that this was related to the presence of 3-0x0 and unsaturated fatty acids. In order to evaluate the possible role of these groups in endotoxin activity, Qureshi et al. ( 162) subjected lipid A of R. sphaeroides to hydrogenation treatment in the presence of Pt( 1V)oxide hydrate, whereby 14 : O(3-0x0) and A'- 14 : 1 were reduced to the racemic (R,S)-3-hydroxy and the saturated fatty acid, respectively. Biological analysis of the reduced material, however, revealed that the chemical treatment had not changed, that is, augmented, the low endotoxicity of R. sphaeroides lipid A, indicating that other factors (namely the size of acyl groups) are responsible for the lack of endotoxic activity ( 162).

( v ) Nonhydroxylated, Iso-, and Ante-Iso Fatty Acids. - Nonhydrox- ylated fatty acids are, like 3-hydroxy fatty acids, common and obligatory constituents of lipid A. In general, they are saturated and are composed of straight-chain and even-numbered acyl residues having carbon atom num- bers ranging from 12 to 18. In Veillonella, odd-numbered acyl groups are present (1 34). Iso- and ante-iso fatty acids were identified in lipid A of such Bacteroides strains as B. fragilis NCTC 9343 (38,79,117) (Fig. 5), B. gingi- valis, B. distasonis, and B. ovatus (1 27). In Bacteroides strains, 13-Me- 14 : 0 preponderates and, because it does not occur in other lipids A, it may be considered as a diagnostic marker for Bacteroides LPS (80a, 1 17). In LPS of the plant pathogen X. sinensis (78), 9-methyldecanoic acid (9-Me-10: 0), and its &-oxidation product 9-Me-10 : O(2-OH) in addition to 10 : 0(3-OH), 12 : 0(3-OH), and the corresponding iso-branched 1 1-Me- 12 : O(3-OH) have been identified. Iso- and ante-iso branched fatty acids have also been en- countered in other Gram-negative bacteria including Flexibacter, Myxococ- cus, and Cystobacter ( 127). Cyclopropyl fatty acids have previously been postulated to occur as ester-linked constituents of lipid A of Neisseria sicca [cyclo- 13 : 01 and A. calcoaceticus (formerly Micrococcus aceticus). How- ever, subsequent studies on purified lipid A preparations revealed that these are devoid of cyclic (cyclopropyl) nonhydroxylated fatty acids, and, there- fore, the previously encountered lipid A-associated cyclopropyl acyl groups are likely to be derived from other, contaminating bacterial lipids.

Page 258: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

MOLECULAR STRUCTURE OF LIPID A 235

( v i ) Unsaturated (Alkenic) Fatty Acids. -As a general rule, unsaturated fatty acids are absent from lipid A. Nevertheless, R. sphaeroides ATCC 17023 (87,88,162,164) incorporates into its lipid A an unsaturated fatty acid (A7-14: 1)in molaramounts(Fig. 8). In thisexample, 14:0[3-0[(A7-14: l)]] was unequivocally identified by g.1.c.-m.s. (87), f.a.b. -m.s. (88), and 'H- n.m.r. ( 162) analysis as the amide-linked acyloxyacyl residue at GlcN( 11) (Fig. 7). In R. capsulutus lipid A, A5-12: 1 is linked to the 10:0(3-OH) residue bound to C-3 of GlcN(I1) (Fig. 7). In R. vanielii, AI4-22 : 1 was identified in ester linkage; however, its position at the lipid A backbone was not determined (9 1).

Also Enterobacteria are able to synthesize unsaturated fatty acids and to incorporate these into the lipid A component. Thus, when grown at low temperature (10- 15OC) E. coli (143), Salmonella spp. (142), P. mirubilis (37), and Y. enterocolitica (145) are incorporated into the lipid A component unsaturated fatty acids that are not present in LPS ofbacteria grown at 37 "C. For E. coli and Salmonella strains grown at low temperatures, it was found that (2)-A9-hexadecenoic acid (A9-16 : 1) was incorporated at the expense of 12 : 0 (142,143), however, not quantitatively. Further investigations of these lipid A by 1.d.-m.s. revealed that the unsaturated fatty acid specifically replaced the 12 : 0 residue in 14 : 0[3-0( 12 : O)] that is bound to GlcN( 11) (37). A similar effect of thermoadaptation, resulting in the formation of amide- bound 14 : 0[3-O(A9-16 : l)], was detected in P. mirabilis and Y. enterocoli- tica (145).

Based on the foregoing examples, it may be concluded that unsaturated fatty acids present in lipid A are always bound to the hydroxyl group of the amide-linked 3-hydroxy fatty acid residue at GlcN(I1) (position 2'). Inter- estingly, the substitution pattern of this particular hydroxyl group, which, in all lipid A studied, cames a fatty acid, varies with the growth temperature and within bacterial genera. This specific site of the hydrophobic region of lipid A, therefore, expresses a relatively high degree of variability.

c. Linkage of Fatty Acids to the Lipid A Backbone. - The lipid A back- bone of E.coli (Fig. 2), S. minnesota (Fig. 4), and S. typhirnurium carries approximately 4 mol eq. of (R)-3-hydroxy fatty acids [ 14 : 0(3-OH)], 2 of which occupy amino functions (amide linkage at positions 2 and 2') and 2 of which are linked to backbone hydroxyl groups (ester linkage at positions 3 and 3'). In C. jejuni lipid A (Fig. 3), 3 mol eq. of (R)-3-hydroxy fatty acids are arnide-linked, and in P. diminuta the four primary fatty acids are exclusively amide-bound. Both amide- and ester-bound (R)-3-hydroxy fatty acids carry, at their 3-hydroxyl groups, secondary fatty acids in ester linkage, forming 3-acyloxyacyl groups. These 0- and N-linked 3-acyloxylacyl groups are characteristic for lipid A and are found in taxonomically remote groups of Gram-negative bacteria.

Page 259: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

236 ULRICH ZAHRINGER et al.

( i ) Ester-Bound Fatty Acids.-It is known that the ester-bound fatty acids, that is, their number, size, and type, play an essential role in the expression of endotoxic activity both in vitro and in vivo (5,118,162). As an example, not only 0-deacylated lipid A, but also the disaccharide precursor molecule (tetraacyl precursor Ia) containing four 14 : O( 3-OH) residues (Fig. 9A) is inactive in inducing interleukin- 1 (IL- 1 ), interleukin-6 (IL-6), and tumor necrosis factor a (TNFa) in vitro (1 67 - 169) or in eliciting the local Shwartzman phenomenon (1 70). This is in contrast to bacterial ( 17 1) and synthetic (1 72) pentaacyl precursor Ib (Fig. 9B) containing, in addition, 16 : 0 bound to 14 : O(3-OH) at GlcN(1) (1 73), which expresses significant endotoxicity.

Ester-linked fatty acids are bound to two types of hydroxyl groups in lipid A, namely, (i) the hydroxyl groups of the backbone at position 3 and 3’, which carry either 3-hydroxyacyl or 3-acyloxyacyl groups, or (ii) the hy- droxyl groups of the backbone-linked 3-hydroxy fatty acids, which carry, in general, nonhydroxylated acyl groups. In order to obtain selectively O-de- acylated derivatives of lipid A of defined chemical structure and, thus, to elucidate the biological significance of ester-bound acyl groups, many at- tempts, including selective chemical degradation or application of specific esterases, have been made. One successful approach toward selective O-de- acylation of S. minnesota Re mutant R595 (128) LPS led to the discovery of ester-bound 14 : 0[3-O( 14 : O)]. Under mild alkaline conditions (0.25 M NaOH, 5 min, 56“C), the 14 : 0[3-O( 14: O)] residue, attached to position 3’ of GlcN(I1) (Figs. 2 and 4), is released. On the other hand, treatment of 4’-monophosphated lipid A of S. minnesota with triethylamine [3% (v/v), pH 9.41 at 100°C (1 74) causes rapid liberation of the 14 : O(3-OH) residue at position 3 of GlcN(1) and only slow release of 14 : O[ 3-O( 14 : O)] (1 74). Simi- lar observations were made with the monophosphated lipid A of S. typhi- murium (32,41), S. minnesota R595, and E. coli J5 (1 75). The reason for the different behavior of 14 : O(3-OH) at position 3 and of 14 : 0[3-O( 14 : O)] at position 3’ is currently not understood. It is likely that the free j3-hydroxyl group of 14 : O(3-OH) contributes to its lability toward alkaline saponifica- tion (175).

Hydroxylaminolysis, treatment with stronger alkali (0.5 M NaOH, 2 h, 100°C) and alkaline methanolysis (0.25 M NaOMe, 1 h, 5OoC) lead to com- plete 0-deacylation of LPS and lipid A (176). Particularly in the case of alkaline methanolysis, ester-linked 3-acyloxyacyl residues undergo, in addi- tion to transmethylation, a j3-elimination reaction, whereby the (R)-3-hy- droxy fatty acid ester is first transformed into the a,P-unsaturated and then into the (S,R)-3-methoxy fatty acid methyl ester. The acyl substituent, on the other hand, is eliminated in the form of the free fatty acid (176). In fact, the presence of a 3-methoxyacyl derivative in the fatty acid spectrum of a given LPS is a strong indication for the presence of an ester-bound 3-acyloxyacyl

Page 260: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

MOLECULAR STRUCTURE OF LIPID A 237

A

-0,

HO'

0

"OH

I I ,o-

B

0 -0,II 0

P' /

HO

FIG. 9.-Chemical structure of precursor Ia (A) and precursor Ib (B).

residue. This P-elimination reaction, therefore, is useful in the structural elucidation of lipid A.

Furthermore, enzymes that quantitatively liberate ester-bound fatty acids of lipid A are known. Such esterases are present in the amoebae Dictyoste- lium discoideum (1 77 - 1 79) and Acanthamoeba castellanii (1 78). Selective

Page 261: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

238 ULRICH ZAHRINGER et al.

liberation of exclusively secondary fatty acids is elegantly achieved by appli- cation of the enzyme 3-acyloxyacylhydrolase (AOAH). This enzyme, which is present in granulocytes and macrophages (45,180) and which is likely to be involved in the biodegradation of LPS, cleaves nonhydroxylated and 2-hy- droxylated secondary fatty acids present in 0- and N-linked 3-acyloxyacyl groups, that is, regardless of their structural context in the lipid A molecule (181).

Lipid A partial structures, exclusively lacking secondary fatty acids, have recently been found to constitute, like lipid A of R. sphueroides (1 82) and R. cupsulutus (89), strong endotoxin antagonists and to inhibit the LPS-in- duced production of TNFa, IL-1, and IG6 in human monocytes (1 67,169,183 - 185a). In view of these and previous (1 86) findings, partially 0-deacylated lipid A preparations currently occupy a central place in endo- toxin research. Therefore, oligoacyl lipid A structures constitute attractive targets for future chemical synthesis, and the development of new methods for selective 0-deacylation will, most likely, prove to be highly rewarding.

(ii) Amide-Bound Fatty Acids.-In contrast to the wide spectrum of different types of fatty acids involved in ester linkage, only two types of acyl groups are amide-bound, namely, (R)-3-hydroxy and 3-0x0 fatty acids. The amide linkage is relatively resistant toward alkaline or acidic hydrolysis and prolonged treatment may lead to degradation and condensation reactions of 3-hydroxy or 3-0x0 fatty acids. Therefore, other degradation reactions have been developed for the liberation and identification of amide-bound acyl groups (176).

For the analysis of fatty acids amide-linked to GlcN(I), several chemical degradation procedures are available. One comprises periodate oxidation of 0-deacylated and 2H-reduced lipid A (lipid A-OH,). Following permethyl- ation, g.1.c. - m.s. analysis revealed a 2-deoxy- 1-deuterio- 1,3-di-O-methyl- 2-(N-methyl-3-methoxyacylamido)glycerol derivative in which the amide- linked fatty acid at GlcN(1) was identified as 14: O(3-OH) in B. pertussis (93), and as 16 : O(3-OH) and 18 : O(3-OH) in R. trifolii (8 1).

A second method uses permethylation of the dephosphated (48% aqueous HF, 48 h, 4°C) and 2H-reduced lipid A. This approach allowed the assign- ment of amide-bound fatty acids linked to GlcN(1) and GlcN(II), as well as the identification of the backbone structure as a HexpN disaccharide (85). Mass-spectrometric analysis of the products was performed by using either a short g.1.c. column (0.3 X 5 cm) or by direct insertion-probe analysis (87). In the case of C. violuceum (85 ) , the mass spectra obtained from the permethyl- ated HexpN disaccharide bearing attached N-methylacyl residues revealed unequivocally that both amino groups camed 12 : O(3-OH).

The third approach to analyze amide-linked fatty acids in lipid A (1 87) is

Page 262: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

MOLECULAR STRUCTURE! OF LIPID A 239

based on selective methylation, that is, the formation of a methyl imidate in the presence of methyl iodide and silver salts as catalyst ( 188). The interme- diate acyl imidate (a Schiff s base) is highly susceptible to mild acid hydroly- sis and, at pH 2.0 (20 min, 20°C), the formerly N-bound acyl or acyloxyacyl group is rapidly released as the methyl ester, whereas all ester linkages remain intact. The liberated acyl methyl esters can be analyzed directly by g.1.c. - m.s.. Using this procedure, the presence of amide-bound 14 : 0[3-O( 12 : O)] and 14:0[3-O( 16 : O)] was, for the first time, demonstrated in lipid A of S. minnesota (1 87). Later, it was shown that amide-linked 3-acyloxyacyl resi- dues are also present in lipid A of other Gram-negative bacteria (38). A disadvantage of this procedure is, however, the fact that only the amide- linked 3-acyloxyacyl of /3-glycosides are liberated to a satisfactory extent (1 88). The low yields obtained in the case of a-anomeric GlcN(1) are most likely attributable to hindrance of an association with the catalyst (Ag+).

Finally, amidases from the slime mould D. discoideum (1 79) were found to cleave the amide-bound long chain fatty acids of S. london LPS. Interest- ingly, 0- and N-linked acetyl groups in the oligosaccharide region of LPS were not affected. Khorana and coworkers (1 89,190) isolated and purified two different acyl amidases from D. discoideum. The two enzymes expressed distinct substrate specificity, one liberating specifically the N-acyl residue attached to GlcN( I) and the other the amide-bound acyl group at GlcN( 11).

It should be pointed out that modem spectrometric techniques, such as f.a.b. - m.s., 1.d. - m.s., or even n.m.r. if applied alone, do not allow to the elucidation of the nature of amide-linked acyl groups. This problem can be solved only by using, in addition, chemical or enzymic methodologies such as the ones just described.

d. Location of Fatty Acids at the Lipid A Backbone.-The chemical methods described thus far provide a great deal of information as to the type of linkage and, therefore, the possible location of acyl groups in lipid A. However, for unequivocal allocation of the fatty acids to individual hydroxyl and amino groups of the lipid A backbone, the information gained by chem- ical analysis must be supplemented by other (physical) methods such as n.m.r., f.a.b.-m.s., Z5zCf-p.d.-m.s., and 1.d.-m.s..

The first f.a.b.-m.s. investigations were performed on highly purified mono- and bis-phosphate lipid A preparations of S. typhimurium (41). F.a.b. - m.s. permitted the determination of the molecular weight ofboth bis- ([M + H]+ = 1798) and monophosphated ([M + H]+ = 1700) lipid A. Furthermore, cleavage at the C-1'-0-C-6 glycosidic bond gave an oxon- ium ion fragment (m/z = 1087) that corresponded to GlcN( 11) carrying one (dimethyl) phosphate, two 14 : 0(3-OH), one 14 : 0, and one 12 : 0 residue (Figs. 2 and 4). Fragments derived from the reducing end of lipid A [GlcN( I)]

Page 263: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

240 ULRICH ZAHRINGER et al.

were not observed in the positive f.a.b. m a s spectrum but could be calcu- lated as the difference between the molecular mass ([ M + H]+ = 1700) and the fragment of the oxonium ion (m/z = 1087). Thus, in this case, GlcN(1) carried two 14: O(3-OH) fatty acyl groups, which, however, could not be assigned to specific positions 2, 3, or 4 of GlcN(1) or to one of the hydroxyl groups of the two 14 : O(3-OH) residues. Nevertheless, the f.a.b. -m.s. analy- sis not only confirmed the presence of acyloxyacyl residues as earlier sug- gested by chemical analysis (187), but also allowed, for the first time, the assignment of the two acyloxyacyl residues to the distal GlcN (11) in lipid A of S. typhimurium. It was realized in consequence that, in S. typhimurium lipid A, GlcN(I1) camed four and GlcN(1) only two acyl groups; that is, the fatty acid distribution over the two GlcN residues was asymmetrical (see later).

Laser-desorption mass spectrometry (1.d. - m.s.) of dephosphated and nonpurified lipid A was introduced in studies on the acylation pattern of lipid A of S, minnesota, E. coli, and P. mirabilis (37). Although these earlier studies did not establish whether some mass peaks originated from laser-in- duced fragmentation or from smaller, intact molecular species (namely monomeric subunits formed during acidic hydrolysis), the first 1.d. -m.s. study, taken in combination with chemical results (38,187), revealed the location of the individual fatty acids in S. minnesota, E. coli, and P. mirabi- lis. The success of 1.d. - m.s. for locating acyl groups was demonstrated with P. mirabilis lipid A isolated from cells grown at physiological and low tem- peratures (37). Cultivation of P. mirabilis at 12°C caused the incorporation of A9-16 : 1 at the expense of 12 : 0. Using 1.d. -m.s. in combination with chemical analysis, it was possible to determine precisely the position of the A9-1 6 : 1. It was found (37) that the unsaturated fatty acid positionally re- placed 12:O at the 3-hydroxyl group of amide-linked 14:0(3-OH) at GlcN( 11). Furthermore, the study confirmed earlier analytical investigations on the fatty acid distribution in Salmonella (187). An advantage of the 1.d. - m.s. method lies in the possibility of controlling the degree of fragmen- tation by the amount and kind of alkali salts (NaI, CsI) added to the sample for cationization. In this way, both the fragmentation pattern and the het- erogeneity of a preparation may be studied in detail (39).

It has recently been demonstrated ( 19 1) that the nature and location of lipid A primary fatty acids is determined by the specificity of the enzymes UDP-GlcpNAc-0-acyltransferase and UDP-3-0-[(R)-hydroxyacyl]- GlcpN-N-acyltransferase for acyl- acyl camer protein (acyl ACP). The anal- ysis of the acyl ACP specificity of these 0- and N-acyltransferases should, therefore, constitute a biochemical approach for elucidation of the location of primary fatty acids in lipid A ( 19 1).

Page 264: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

MOLECULAR STRUCTURE! OF LIPID A 24 1

e. Lipid A Differing in the Number and Location of Fatty Acids. -Lipid A derived from various bacterial sources may vary in the number of fatty acids present and their location. These variations have been elucidated in lipid A derived from R-form LPS. On the other hand enterobacterial S-form LPS is heterogeneous and that fraction containing a long polysaccharide component, namely, the 0-specific side chain, contains acyl-deficient lipid A, the structure of which has not been studied thus far(47). Described here are selected but representative examples of lipids A that are classified ac- cording to the number of acyl groups they carry and to their distribution over the HexpN residues of the backbone.

( i ) Hexaacyl Lipid A.-Most lipid A studied contain up to six acyl groups. Hexaacyl lipid A is present in Enterobacteriaceae (such as E. coli), but also in such taxonomically remote bacterial families as the Neisseria- ceae, and the Pseudomonadaceae (for instance, C. violaceurn). These lipid A carry the four primary 3-hydroxy fatty acids and two additional acyl groups linked to hydroxyl groups of primary fatty acids. With respect to the location of the secondary acyl groups, two types of lipid A may be distinguished, namely, lipid A having an asymmetric (E. coli type) and symmetric (C. violaceurn type) acylation pattern. As noted previously (92), bacteria pos- sessing a symmetric acylation pattern belong to the P-subclass of Protobac- teria ( 192), whereas the Enterobacteriaceae, exhibiting an asymmetric acy- lation of lipid A, are members of the y-subclass.

(a). Asymmetrical Acylation Pattern of H e x w y l Lipid A. The classical representative of an asymmetrically acylated hexaacyl lipid A is that of E. coli Re LPS (strain F5 15). As described, this lipid A carries at positions 3’,2’,3, and 2 of the backbone, four 14: O(3-OH) residues (Fig. 2) with the hydroxyl groups of the two 14 : O(3-OH) residues bound to GlcN( 11) being acylated by I2 : 0 and 14 : 0. Therefore, a total of six fatty acids is present, four of which are associated with GlcN(I1) and two with GlcN(1). Thus, the overall acylation pattern is asymmetric (4 + 2). A number of other hexaacyl lipid A were identified as being asymmetrically substituted by fatty acids as in E. coli, and these include S. typhirnuriurn (4 l), the hexaacyl species of S. rninnesota (37), P. rnirabilis (37), H. injluenzae (68), Providencia rettgeri ( 193), A. actinornycetemcornitans (94) and probably Pasteurella rnultocida (1 93a). The lipid A of C. jejuni also belongs to the class of asymmetrically acylated hexaacyl species. It should be noted, however, that GlcN(I1) is replaced here by GlcpN3N (97).

In asymmetrically acylated lipid A, the four primary hydroxy-fatty acids possess the same chain length of 14 carbon atoms [thus far, with one excep- tion ( 193b), only 14 : O( 3-OH) was identified].

Page 265: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

242 ULRICH ZAHRINGER et al.

- 0, II ,o II ,o- P

HO'

0

FIG. 10.-Chemical structure of lipid A of Chromobacterium violaceurn. The dashed line indicates nonstoichiometric a-hydroxylation (85) of 12 : 0. For details, see the text, and for substitutents of phosphate residues, see Table I.

(b). Symmetrical Acylation Pattern of Hexaacyl Lipid A. In the case of C. violaceurn (Fig. lo), the lipid A backbone also carries four primary acyl groups [O-linked 10 : O(3-OH) and N-linked 12 : 0(3-OH)]. The secondary fatty acids present [approximately 2 mol of 12:O plus 12:0(2-OH)] are linked to the hydroxyl groups of the two 12 : O( 3-OH) residues of which one is associated with GlcN(1) and one with GlcN(I1) (38,85). In this instance, therefore, GlcN( I) and GlcN(I1) each carry three fatty acids, resulting in a symmetric acylation pattern (3 + 3).

Other lipid A examples possessing a symmetrically acylated backbone are N. gonorrhoeae (33), N. rneningitidis (73), the hexaacyl species of P. aerugi- nosa (77), R. gelatinosus (84), R. gelatinosa (194), S. natans (92), and, most likely, B. pertussis ( 139) and X . sinensis (38). In symmetrically acylated lipid A, the two ester-linked and the two amide-linked primary 3-hydroxy fatty acids possess (with the exceptions of R. gelatinosus, R. gelatinosa, and S. natans) different chain lengths, the 0-bound acyl group having 10 or 12 and the N-bound acyl group possessing 10, 12 or maximally 14 carbon atoms. It should be noted that, in C. violaceurn lipid A, the amide-linked 12 : O(3-OH) at GlcN( 11) carries either 12 : 0 or 12 : 0(2-OH), yielding two molecular spe- cies of lipid A (Fig. 10).

Page 266: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

MOLECULAR STRUCTURE OF LIPID A 243

(ii) Heptaacyl Lipid A.-Lipid A species carrying seven fatty acids have been encountered in Re LPS of S. minnesota R595 (174,187), S. typhimur- ium (195), E. carotovora (99, and P. mirabilis R45, (37,72) but also in a temperature-sensitive mutant of E. coli (195a). These heptaacyl forms ex- hibit the hexaacyl E. coli type of acylation pattern but have, in addition, one 16 : 0 residue bound to the 3-hydroxyl group of 14 : 0(3-OH), which is amide-linked to GlcN(1) (Fig. 4). It must be noted, however, that the hep- taacyl species often represents a minor (20 - 3096, w/w) lipid A component, whereas the major species is a hexaacyl form resembling, in its acylation pattern, E. coli type lipid A. Heptaacyl S. minnesota lipid A has been chemi- cally synthesized (196).

In lipid A of Moraxella catarrhalis an unusual lipid A was identified possessing an asymmetrical (3 + 4) acylation pattern with four 12:0(3-OH) residues as primary fatty acids, three of which are acylated to form acyloxya- cyl residues [ 12:0(3-O( 10:0)] at positions 2’ and 3 and [ 12:0(3-0(12:0)] at position 2 (193b).

(iii) Pentaacyl Lipid A. -Among the pentaacyl lipid A structures studied thus far are those in which only one of the four primary acyl groups is 3-0-acylated [B.fragilis (79), R. sphaeroides (87,88), R. capsulatus(89), and precursor Ib (173)l. On the other hand, lipid A are now known in which a major species lacks one of the primary fatty acids (P. aeruginosa) (77).

In the major component of B. fragilis lipid A, five comparatively long- chain and, in part, iso-branched fatty acids are present (79). Their location is shown in Fig. 5. It is evident that the only secondary acyl group present is bound to the 3-hydroxy fatty acid at position 2’ [GlcN(II)]. It is a peculiar feature of B.fragilis lipid A that the hydroxyl group at position 4’ ofGlcN( 11) is free; that is, that the nonglycosylic phosphate group is absent.

A pentaacyl lipid A is also found in R. sphaeroides (Fig. 7) (88). This lipid A has the same distribution of fatty acids as B. fragilis (79) but, as a notable feature, it contains two rare types of fatty acids, namely, A’-14 : 1 and 14 : O(~-OXO), the former being present as a 3-acyloxytetradecanoic acid group amide-linked to GlcN (11), the latter being amide-bound to GlcN(1).

Furthermore in R. capsulatus (Fig. 7) a pentaacyl lipid A having two 14 : O(3-0x0) groups in amide linkage constitutes a main component (89). Two residues of 10 : 0(3-OH), of which the one bound to GlcN(I1) carries dodecenoic acid (A5-12 : 1) at its 3-hydroxyl group in nonstoichiometric amounts, are ester linked. The location of R. capsulatus fatty acids, however, differs significantly from that of the B. fragilis and R. sphaeroides pentaacyl lipid A in that the secondary fatty acid (1 2 : 1) is bound to the ester-linked 3-hydroxy fatty acid at position 3‘. Another fraction of R. capsulatus lipid A lacks 12 : 1 and, thus, exemplifies a tetraacyl lipid A species.

Page 267: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

244 ULRICH ZAHRINGER et a1

A representative of a pentaacyl lipid A, in which the only secondary fatty acid present is associated with GlcN(I), is precursor Ib (Fig. 9 B). This compound was isolated from a temperature-sensitive mutant of S. typhi- murium (mutant Ts5) (173). The only 3-acyloxyacyl residue in precursor Ib is 14 : 0[3-0( 16 : O)], which is amide-linked to GlcN (I) (position 2).

A pentaacyl lipid A is also present in P. aeruginosa (77) (Fig. 1 1A). Here, the main lipid A species contains a total of five fatty acids, and a minor hexaacyl species (Fig. 1 1B) corresponds structurally to lipid A of C. violu- ceum (Fig. 10). The prominent pentaacyl component, which makes up approximately 75% (w/w) of P. aeruginosa lipid A, encompasses three struc- tural forms that all possess the same p-( 1 + 6)-linked GlcpN backbone, but with only three (primary) 3-hydroxy fatty acids attached to positions 3’, 2’, 3, and 2 (Fig. 11A). These structural forms differ from each other by the 3-0-acylation of each of the two amide-linked 12 : O(3-OH) residues by the secondary acyl groups 12 : 0 or 12 : 0(2-OH), as indicated by the dashed lines. Of the four conceivable structural types, the one bearing two 12 : O(2-OH) residues is not present.

( i v ) Tetraacyl Lipid A. - Tetraacyl lipid A constitute a minor species of, for example, enterobacterial lipid A preparations (32). They may be the result of incomplete biosynthesis or arise from the acid treatment applied to LPS for the preparation of free lipid A. In this sense, mature tetraacyl lipid A does not exist. However, a tetraacyl lipid A partial structure (Fig. 9 A) is produced by a S. typhimurium mutant having a conditional defect in the biosynthesis of Kdo (197). This lipid A partial structure, constituting a precursor in lipid A biosynthesis, was first termed acidic precursor ( 122,123) and (around 1982) precursor Ia. Unfortunately, the same molecule, also isolated from a S. typhimurium mutant, was later termed precursor IV or lipid IV (63,198). In order to distinguish the tetraacyl compound from an associated and I6 : 0-containing pentaacyl form (the latter is called precursor Ib or lipid IVB), the terms precursor Ia (199) and lipid IV, (63) were intro- duced and used synonymously from 1985 on. In view of the fact that it was the first introduced into the literature, the term precursor I (with the distinc- tion between precursor Ia and precursor Ib) will be used in this article. Precursor Ia is composed of a p-~-GlcpN-( 1 - 6)-a-~-GlcpN 1,4’-bisphos- phate carrying only primary acyl groups, namely, four 14 : O( 3-OH) residues, two in amide (2’ and 2) and two in ester linkage (3’ and 3), respectively. Lipid A precursor Ia has been chemically synthesized (compound 406 or LA-14- PP) (200).

f. Concluding Remarks. -Fatty acids constitute important components of lipid A and confer the hydrophobic properties characteristic of the mole-

Page 268: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

MOLECULAR STRUCTURE OF LIPID A 245

A

0 - 0 II

HO

‘ P’ /

B

0

P -0 , II ,o

HO’

FIG. 1 1 . -Chemical structure of the two preponderant lipid A forms of Psatdomonus ueru- ginosu. (A) Pentaacyl lipid A (major lipid A fraction; 75%, w/w). (B) Hexaacyl lipid A (minor lipid A fraction, 2596, w/w). Dashed lines indicate nonstoichiometric a-hydroxylation of 12 : 0. A lipid A species having 2 mol 12 : 0(2-OH)/moI lipid A was not detected (77).

Page 269: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

246 ULRICH ZAHRINGER et al.

cule. With regard to the nature, linkage type, size, and number of the fatty acids as well as with regard to their location in lipid A, the following general- izations may be made.

Lipid A contains primary fatty acids directly linked to hydroxyl and amino groups of the backbone, and secondary fatty acids bound to hydroxyl groups provided by the primary acyl residues. The number of carbon atoms of primary and secondary fatty acids is, in the majority of lipid A studied, in the range of 10 to 18. They are, in general, saturated, even-numbered, and straight-chain fatty acids and in only few cases, are unsaturated, odd-num- bered, and iso- and ante-iso branched derivatives present in molar amounts.

The primary fatty acids are (R)-configured 3-hydroxy fatty acids, but 3-OX0 fatty acids [thus far only 14 : O(3-0xo)l have also been identified in lipid A. The primary 3-hydroxyacyl groups can be involved in both ester and amide linkage, whereas 14 : O(3-0x0) is only amide-bound. 3-Hydroxy fatty acids are present exclusively as primary acyl groups, the only exception known so far being K cholerae (86) lipid A. Often, only one type of 3-hy- droxy fatty acid is present [ 14 : O(3-OH) in Enterobacteriaceae], but exam- ples in which lipid A contains two or more different 3-hydroxy fatty acids (C. violaceurn, and B. fragilis) are also known. If there are several homologous 3-hydroxylated acyl groups, the one having the longest chain is amide-bound (4), the other@) being engaged in ester linkage (P. aeruginosa, and N. gon- orrhoeae). In general, the chain lengths of the amide-bound 3-hydroxy fatty acids at HexN(1) and HexN(I1) are identical (E. coli), but they may also differ as in B. ji-agilis (79), X . sinensis (78), M. fulvus (201), and R. trifolii (202). If the amide-bound fatty acids associated with HexN(1) and HexN(I1) differ in the number of carbon atoms, the one having the longest chain is linked to HexN(1) [for example, GlcN(1) in B.fragilis (79)].

The secondary fatty acids are particularly composed of nonhydroxylated fatty acids, but 2-hydroxylated fatty acids are also found. The 2-hydroxy fatty acids always possess the (3-configuration and, together with the non- hydroxylated fatty acids of the same chain length, they often add up to 1 mol eq. (as in S. rninnesota). Up to three secondary fatty acids may be linked to the 3-hydroxy fatty acids at positions 2,2’, and 3’. With one exception (193b) the 3-hydroxy fatty acid at position 3 [HexN(I)] is never 3-0-acylated. In lipid A possessing two secondary acyl groups, these are linked either to the 3-hydroxy fatty acid at position 2’and 3’ (E. coli type asymmetric acylation) or to those at positions 2’ and 2 (C. violaceurn type symmetric acylation). If only one secondary acyl group is present, it may be linked to the 3-hydroxy fatty acid at position 3‘ (R. capsulatus), position 2’ ( B. ji-agilis and R. sphaeroides) or position 2 (precursor Ib). Secondary fatty acids are absent from the tetraacyl lipid A partial structure precursor la.

Among the lipid A-associated fatty acids, 28 : O(27-OH) or 29 : O(28-OH) and 26 : O(25-OH) deserve special mention, as they deviate considerably

Page 270: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

MOLECULAR STRUCTURE OF LIPID A 241

from the former in chain length and position of hydroxylation. These fatty acids possess twice the number of carbon atoms as the usual fatty acids present in, for example, enterobacterial lipid A. They extend completely through the outer membrane and may be important for a stable bilayer arrangement. The penultimately located hydroxyl group may be substituted by a secondary acyl group, thus contributing to the rigid organization of the outer membrane system. These long chain fatty acids have only recently been identified. Further studies are necessary to see whether they adhere to the general rules presented here, to identify their stereochemistry, and to learn more about their taxonomic, biological and physicochemical signifi- cance. Interestingly, they are associated with members ofthe a-2 subgroup of bacteria based on homologies of their 16s rRNA (203). The presence and estimation of 28 : 0(27-OH), therefore, is of great taxonomical value (10 1,160).

4. Hydroxyl Group in Position 4 of the Lipid A Backbone

The presence of a free, that is, unsubstituted, hydroxyl group at the lipid A backbone was indirectly postulated after the discovery of amide-linked 14:0[3-0(12:0)] and 14:0[3-0(16:0)] in S. minnesota R595 lipid A (4,187). It was known that phosphate was bound to the hydroxyl group at position 4’, and that Kdo occupied one hydroxyl group, and because 1 mol of each 14 : 0[3-O( 14 : O)] and 14 : O(3-OH) was known to be ester-linked, it was obvious that one out of the three available hydroxyl groups was not substituted. However, the question as to which hydroxyl group of the lipid A backbone was free remained open for some time. The elucidation of the enterobacterial lipid A biosynthesis and the identification of lipid X “2- deoxy-2-[(R)-3-hydroxytetradecanamido]-3-0-[(~)-3-hy~oxytetradecan- oyll-a-D-glucopyranosylphosphate, that is a-~-GlcpN 1-phosphate acyl- ated at positions 2 and 31 as a precursor of lipid A biosynthesis rendered the hydroxyl group at position 4 of GlcN( I) a likely candidate. That indeed this hydroxyl group was free was unequivocally shown by Imoto et al. (35), who isolated from E. coli Re LPS (strain F5 15) a monophosphated lipid A, part of which, after esterifying the phosphate groups with diazomethane, was sub- jected to two-dimensional ‘H-n.m.r. spectroscopy (‘H,’H-COSY). A fur- ther sample was, in addition, peracetylated (whereby five acetyl groups were introduced into the molecule) and also analyzed by H-n.m.r. spectroscopy. Comparison of the ‘H-n.m.r. data of both lipid A derivatives revealed a significant downfield shift of about 1.5 ppm for the H-4 signal (3.55 vs. 5 .O 1 ppm) before and after 0-acetylation, whereas all other signals remained unchanged. In this way, for the first time, the authors could clearly demon- strate that position 3 of GlcN(1) [and position 3’ of GlcN(II)] was acylated, whereas the hydroxyl group at C-4 of GlcN( I) was free. Unfortunately, the expected shift of the primary hydroxyl group in position 6’ (compare Section

Page 271: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

248 ULRICH ZAHRINGER et al.

111.5) was not observed by this approach, as the signals of H-6a’ and H-6b’ could not be unequivocally assigned because of the overlapping with other signals in the region of 3.7-4.2 ppm.

Takayama and coworkers (60) introduced the h.p.1.c. separation tech- nique for such amphiphilic molecules as lipid A, and in earlier experiments they applied paired-ion reverse-phase h.p.1.c. for the preparation of homoge- neous fractions deriving from 4’-monophosphated lipid A of S. typhimur- iurn. The purified preparations obtained were suitable for f.a.b. - m.s. analy- sis. However, monophosphated lipid A isolated in this way expressed a considerable heterogeneity with respect to the number and location of 0- acyl residues (60). In order to further improve the purification procedure, as well as to obtain lipid A derivatives suitable for n.m.r. spectroscopy, Qureshi et al. (174) prepared the dimethyl phosphate derivative of S. minnesota (R595) lipid A, which, after purification by reverse-phase h.p.1.c. (C,8), could be analyzed by H-n.m.r. The n.m.r. spectrum of, for example, the heptaacyl lipid A dimethyl monophosphate fraction, unequivocally revealed 0-acyl substitution [ 14 : O(3-OH)J at position 3 and a free hydroxyl group at posi- tion 4 of GlcN(1).

The problem of determining unsubstituted free hydroxyl group(s) located at the lipid A backbone still remains a challenge in analytical chemistry. This is mainly because of the fact that this problem cannot be solved by classical methylation analysis (Hakomori procedure), because the alkaline pH of the reaction conditions causes removal of 0-acyl residues and, thereby, artifi- cially creates free hydroxyl groups. Therefore, a new procedure to determine free hydroxyl group(s) in lipid A, using methylation analysis, has been re- cently introduced by our laboratory (77). The method is based on a methyla- tion procedure originally described by Fugedi and Kovacz (204). By this method, free hydroxyl groups are transformed into their methyl ethers via electrophilic substitution with trimethyloxonium tetrafluoroborate [Me,O+ BF,-]. The reaction is performed in dichloromethane for 16 h at 20°C in the presence of a sterically hindered base (2,5-di-tert-butyl-4-methyl-pyridine). In contrast to other methylation procedures (such as Hakomori methyla- tion), these conditions allow free hydroxyl groups to be transformed into methyl ether derivatives without affecting or cleaving ester bonds. On apply- ing this method to monophosphated lipid A of S. rninnesota (R595) and E. coli (F5 1 5), followed by acetolysis, NaBH,-reduction, and per-0-acetyla- tion, g.1.c. - m.s. analysis revealed 1,3,5,6-tetra-O-acety1-2-deoxy-4-0- methyl-2-(N-methylacetamido)-~-glucitol to be the only derivative of GlcN(1). This result shows that, in the lipid A preparations studied, the hydroxyl group in position 4 of GlcN(1) was free. This procedure was also applied to lipid A of P. aeruginosa (77) and N. rneningitidis (73), in which cases the hydroxyl group at C-4 was also found to be unsubstituted.

Another procedure for determining free hydroxyl groups applies diazo-

Page 272: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

MOLECULAR STRUCTURE OF LIPID A 249

methane (CH,N,) with SiO, as a catalyst (205,206). This method was used to demonstrate the presence of free hydroxyl groups at position 4 (and 6’; compare Section 111.5) in, for instance, lipid A of R. sphaeroides ATCC 17023 (87). This convenient procedure, however, suffers from the great disadvantage of not giving quantitative yields of methyl ethers. It, therefore, does not allow the characterization of the degree of substitution of hydroxyl groups of the lipid A backbone and determination of whether the hydroxyl groups are free.

One example in which the C-4 hydroxyl group of lipid A is substituted is known. Thus, in R. tenue (90), nonacylated GlcpN is glycosidically linked to position 4 of GlcN( I).

It must be emphasized that the problem of unsubstituted hydroxyl groups is usually studied employing free lipid A prepared by treatment of LPS with acid. The demonstration of a free hydroxyl group at C-4 in monophosphated lipid A therefore does not exclude the possibility that, in LPS, a substituent, present in acid-labile linkage, could be bound to 0-4 of lipid A. This possi- bility has thus far been excluded for E. coli Re LPS which was analyzed by ‘H- and I3C-n.m.r. and shown to contain an unsubstituted hydroxyl group at

5. Hydroxyl Group in Position 6’ as the Attachment Site of Kdo

Based on results obtained by periodate oxidation of deacylated S. minne- sota Re (R595) LPS, it was proposed (1 12) in 197 1 that the hydroxyl group at position 3’ of GlcN( 11) served as the attachment site of Kdo to lipid A. This structural proposal was corroborated by other groups, who investigated the core-backbone structure by various degradation procedures (69) and by ‘H- and 31P-n.m.r. spectroscopy (109). However, methylation analysis of the deacylated core (Kdo) - lipid A backbone oligosaccharide obtained from a P. mirabilis Re mutant (strain R45), in combination with g.1.c. -m.s. of par- tially methylated glucosaminitol acetates (72), revealed l75,6-tri-0-acetyl-2- deoxy-3,4,5-tri-0-methyl-2-(N-methylacetamido)-~-gluc~tol as the major product deriving from GlcN(I1). The presence of an acetyl residue at the primary hydroxyl group at position 6’ of (the reduced) GlcN(II), as well as the presence of a methyl group in position 3, clearly showed that an acid-la- bile substituent, namely, Kdo, had been linked to position 6’ and not to position 3’ as previously assumed. Also in 1983, ‘H- and I3C-n.m.r. spectro- scopic investigations on LPS of a Re mutant of E. coli LPS (35,66) suggested that Kdo was linked to the hydroxyl group at position 6’ of GlcN( 11) of the lipid A backbone. Again at approximately the same time it was discovered that lipid X represented a monomeric precursor of lipid A biosynthesis(207). The known location of lipid A acyl groups (positions 3’, 2’, 3 and 2) and phosphate residues (positions 4’ and 1) suggested that Kdo was linked to the primary hydrsxyl group at position 6’. Unger and coworkers (70) investi-

C-4 of GkN( I) (34,110).

Page 273: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

250 ULRICH ZAHRINGER et al

gated the structure of a (Kdo), - (GlcpN), tetrasaccharide derived from S. minnesota Re (R595) LPS by hydrazinolysis and h.p.1.c. purification. Based on the 13C-n.m.r. data, it was suggested that Kdo was linked to 0-6’ of GlcN(II), although the site of attachment of Kdo to the backbone could not be determined unequivocally. This is because a-ketosidically linked Kdo residues give rise to only a small and nondiagnostic glycosidic shift in the range of only 1 - 2 ppm (glycosidic shifts are usually 8 - 10 ppm downfield) of the C-6’ signal (64). In addition, such model compounds as methyl a- and P-ketosides of Kdo exhibit nondiagnostic chemical-shift differences for the determination of the anomeric linkage at (2-2, these differences being less than 1 ppm (208).

In summary, these investigations strongly indicated that Kdo was linked to 0-6’ of GlcN( 11) of lipid A, but final experimental proofwas lacking. Also, the anomeric configuration of the lipid A-proximal Kdo residue was un- known. This is mainly because, in contrast to normal aldoses, the anomeric linkage of Kdo to lipid A cannot be determined by simple ‘H-n.m.r. analysis because of the lack of a proton at the anomeric center. This problem was first addressed (65) in E. coli (mutant D31m4) by applying W-n.m.r. In the proton-coupled 13C-n.m.r. spectrum of Kdo, the heteronuclear coupling constant 3JC1,H.3ax was determined to be = 5 Hz for the p-D anomer (209), whereas it appeared as a sharp singlet (3JG,,H-3an) < 1 Hz when the a-D anomer was present (64). This results from the stereochemical arrangement of the carboxyl group at C- 1 and the methylene protons at C-3 exhibiting a trans-relationship for the 8-D (3J~1,~.3ax = 5 Hz) and a gauche orientation for the a-D anomer (3JCI,H-3ax < 1 Hz). The proton-coupled 13C-n.m.r. spectrum of E. cdi Re LPS (triethylammonium salt) showed a 3JG1,H-3ax value of = 3 Hz (after subtraction of computer-generated line broadening), which nevertheless led the authors to conclude that the Kdo linkage was a (65).

Unequivocal proof for the attachment site to lipid A and the configuration of Kdo was provided in 1985 by investigations on an 0-deacylated, per-0- acetylated, carboxy- and phosphate-methylated, and highly purified deriva- tives isolated from LPS of an Re mutant (F5 1 5) of E. cali (34) (Fig. 12). This compound was subjected to chemical analysis, 1.d.-m.s., and ‘H- and I3C- n.m.r. spectroscopy. From the chemical-shift values of the H3ax and H-3eq protons in ‘H n.m.r., as well as from the observation of large differences in the chemical shift of the methylene protons (H-8a and H-8b) of the individ- ual Kdo residues [which are known to be characteristic for a-ketosides (210)], and from the signals in the 13C-n.m.r. spectrum, the structure of the Kdo-lipid A backbone was completely elaborated as shown in Fig. 12. According to this study, Kdo is in a-D-ketosidic linkage attached to the hydroxyl group in position 6’ of GlcN(I1) of the lipid A backbone. This

Page 274: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

MOLECULAR STRUCTURE OF LIPID A 25 1

OMe

0

p’ AcO AcO

AcO

Me0,II 0

/ Me0

I 0 R

I R

,OMe

\ 0 Me

0 QAc

R =

FIG. 12. -Chemical structure of an 0-acetylated N,N’-bis(3-acetoxytetradecanoyl) tetrasac- charide tetramethyl phosphate isolated from the E. coli Re mutant F5 15 (34). [For details see text.]

structural arrangement was later confirmed by Qureshi et al., who purified and investigated the hexaacyl LPS species of the E. coli Re mutant D3 1 m4. (1 10).

An interesting case concerning the core -lipid A linkage-region was en- countered in the LPS ofA. calcoaceticus (75). Here, in part of LPS, the link between the polysaccharide region and lipid A is not mediated by Kdo. Instead, the isosteric ~-glycero-~-talo-2-octulosonic acid ( ~ g ~ t - K o ) is present, which, like Kdo, is a-ketosidically linked to C-6’ of GlcN( 11) of lipid A. Interestingly, the a-ketosidic linkage of DgDt-KO to lipid A is highly resistant to acid hydrolysis, a fact most likely related to the presence (75) of the axial hydroxyl group at C-3. DgDt-KO has now been identified as being linked to the lipid A - proximal Kdo residue in Pseudomonas pseudomallei (21 1) and P. cepacia (2 12).

In conclusion, only in few LPS has the Kdo-lipid A linkage been eluci- dated unequivocally. In these examples, Kdo is a-ketosidically linked to 0-6’ of HexN( 11) of the lipid A backbone [E. coli (34,65), H. influenzae (68), an S. minnesota R595 recombinant carrying the genus-specific antigen of Chlamydia spp. (213), and S. minnesota (70)]. The same type of linkage, however, is very likely to exist also in other Gram-negative bacteria, includ- ing P. mirabilis (72), B. fragilis(79), P. aeruginosa (77), V. parahaemolyticus (83), N. meningitidis (73), Rhodobacter, Rhodocyclus, and others. It is note-

Page 275: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

252 ULRICH ZAHRINGER et al.

worthy, that also in lipid A containing a GlcpN3N disaccharide [C. jejuni (97)] or a GlcpN3N monosaccharide backbone [P. immobile (lOS)], the hydroxyl group at position 6’ serves as the attachment site of Kdo.

IV. SYNTHETIC LIPID A

Based on the results of chemical analysis, free lipid A has been chemically synthesized [compare (1 2 - 15,17)]. The first fully synthetic lipid A molecule (preparation 506 or LA-15-PP) corresponds in structure to E. coli lipid A (Fig. 2) carrying a glycosylic (and nonglycosylic) monophosphate. Later, other lipid A and lipid A partial structures were prepared that contain a p-( 1 4 6)-linked GlcpN disaccharide, but that differ in the acylation and phosphorylation pattern. These preparations include the heptaacyl species of S. minnesota lipid A (compound 516 or LA-l6-PP, Ref. 196) and P. mirabilis lipid A (compound A-403, Ref. 2 14), the pentaacyl precursor Ib (LA-2O-PP, Ref. 172, Fig. 9B), and the tetraacyl precursor Ia (406 or LA- 14- PP, Ref. 215, Fig. 9A). In addition, the 4’-monophosphate (for example, compound 504 or LA- 15-PH) and the 1-monophosphate (505 or LA- 15- HP) partial structures of E. coli lipid A, and such acyl-deficient compounds as preparation LA-19-PP, corresponding to 0-deacylated E. coli lipid A, were prepared. Further, C. violuceum-type lipid A (LA-22-PP) containing 0-linked 14 : O( 3-OH) and N-linked 14 : O[ 3-O( 14 : O)] in a symmetrical dis- tribution, and partial structures thereof, was synthesized (1 5) . Also, such lipid A disaccharide analogues as the pentaacyl compound 3 16, having an acylation pattern different from that of lipid A, have been prepared by chemical synthesis (2 16). A, phosphonooxyethyl ( 1 5) compound, a 3-ether (2 17), and fluorinated (2 18) analogues have also been synthesized. Finally, a great number of monosaccharide partial structures having different acyl- ation and phosphorylation patterns have been prepared by several groups (compare see Refs. 1 1,13 - 16,19). These compounds include synthetic counterparts of lipid A-related bacterial products, such as lipid X and lipid Y , as well as other partial structures and analogues, corresponding to either the reducing or the distal HexpN unit of the lipid A backbone. The 4’-mono- phosphate partial structure of bacterial and synthetic precursor Ia (2 15) and Ib (1 72), E. coli hexaacyl lipid A (33, and S. rninnesota heptaacyl lipid A (196) were compared by ‘H-n.m.r., t.l.c., and other methods (2 19), and were found to be structurally identical.

V. CONFORMATION OF LIPID A

In parallel with the increasing knowledge on the primary structure of lipid A, calculations and physical investigations have been performed in order to

Page 276: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

MOLECULAR STRUCTURE OF LIPID A 253

gain insight into the molecular shape and the three-dimensional arrange- ment of lipid A and LPS.

1. Molecular Shape

Single crystals of free lipid A or LPS are as yet not available. Therefore, the most promising approach to obtain molecular models is to perform theoret- ical calculations. After the chemical structures of enterobacterial lipid A had been elucidated, this methodology was successfully applied with heptaacyl 5'. minnesota lipid A (220) and hexaacyl E. coli Re LPS (221). As an example, Fig. 13 shows the atomic model of the E. coli lipid A molecule, as calculated by Kastowsky et al. (22 1) using energy-minimization techniques.

The most striking property of the model is that the lipid A backbone is not orientated perpendicular to the direction of the parallel-aligned fatty acids, but that it displays an angle tilt in the range of 53 O & 7 '. This tilt leads to an energetically favored close packing of the acyl chains and to the consequence that the fatty acids do not end in one plane (perpendicular to the z-axis) as is the case for phospholipids. As a further result, the fatty acids may be inter- digitated, as proposed from neutron-scattering data (222). It should be men-

u A

FIG. 13. -Ball-and-stick (A) and space-filling (B) atomic models ofE. culi lipid A. One out of several possible conformers is shown. The ring plane (right) of GlcN(I1) almost coincides with the plane of the paper (221). Courtesy of Dr. Manfred Kastowsky, University of Berlin.

Page 277: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

254 ULRICH ZAHRINGER et al.

tioned that a similar tilt of the lipid A backbone had also been calculated for free lipid A of S. minnesota (220). Furthermore, the primary hydroxyl group at position 6’ of GlcN( TI), which carries in LPS the polysaccharide region, is exposed on the upper surface of the molecule.

The close packing of the acyl groups associated with the inclination of the lipid A backbone with respect to the fatty acid orientation seems to consti- tute a common and characteristic feature of the lipid A conformation. This specific (endotoxic) conformation is very likely to influence greatly the tend- ency of the amphiphilic lipid A to adopt peculiar supramolecular structures.

2. Three-Dimensional Organization

LPS and free lipid A are amphiphiles, and, therefore, they are not present as individual molecules in aqueous solution above the critical micellar con- centration, but rather as aggregates that may adopt different three-dimen- sional structures. Among the supramolecular arrangements that may be assumed by amphiphiles are micellar, lamellar, hexagonal, and nonlamellar cubic, as well as inverted-hexagonal, structures (223). Factors that influence the formation of aggregates comprise intrinsic parameters such as the pri- mary chemical structure, and extrinsic ones such as temperature, pH, and the concentration of such bivalent cations as Mg2+ and Ca2+. On elevation of the temperature, within one type of structure, transitions can take place at a phase transition temperature (T,) from the gel (p) to the liquid-crystalline (a) state of the acyl chains, reflecting an increase of the fluidity hydrophobic region of the amphiphiles. The /3 - Q phase transition may be associated with a transition between different three-dimensional structures.

In order to learn about the phase states adopted by LPS and lipid A, Fourier-transform infrared spectroscopy, differential scanning calorimetry, and X-ray small-angle diffraction with CuK, or synchrotron radiation have been applied. In the following section, some recent results are summarized.

a. Three-Dimensional Structure. - For free lipid A of S. minnesota and E. coli, complete phase diagrams were established; that is, the dependence of the structural polymorphism on temperature, water content, and Mg2+ con- centrations was determined (224 - 226). It was found, as depicted schemati- cally in Fig. 14, that the - a acyl chain-melting transition is in these studies, under all conditions, connected with a change in the supramolecular structure of the lipid assembly. Within the transition range, different phases usually coexist. In the gel (p) state, cubic phases (Q) preponderate at high water content (> 60 %) and at high [lipid A] : [Mg2+] ratios. In the range of low water concentration, the lamellar phase (L) is the exclusive structure at all Mg2+ concentrations. In the liquid-crystalline (a) state, the hexagonal H, phase is predominant. Its contribution, however, is weak at low Mg2+ con-

Page 278: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

MOLECULAR STRUCTURE OF LIPID A 255

I I I lamellar

I

TC Temperature

t

FIG. 14.-Phase diagram (227) of free lipid A.The phase diagram was established using S. minnesota Re LPS-derived lipid A.

centration and high water content, but becomes prominent at higher Mg2+ concentration and lower water content. Most importantly, under near-phy- siological conditions of water content, divalent cation concentration, and temperature, the lipid A assemblies adopt cubic structures almost exclu- sively (226). A similar polymorphism was observed for Re mutant LPS (227), and for this as well as for free lipid A, the /3 - a chain-melting transition and the transitions between different structures are reversible with temperature.

In other studies applying small-range X-ray or neutron scattering, only lamellar structures were found for free lipid A (2 19,228). The pentaacyl lipid A analogue 3 16, which cames two 14 : O(3-OH) residues in position 2 and 2’, and 14 : 0 groups in position 3,4, and 6’, was found to have its acyl chains in an a (liquid)-type arrangement and to adopt a nonlamellar inverted (H,) structure (2 19). The monosaccharidic lipid X, on the other hand, appears to aggregate in micellar structures, with its two acyl groups [ 14 : 0(3-OH)] being very mobile and in the a-phase (229). Further details of the three- dimensional structure of lipid A are comprehensively reviewed by Seydel and Brandenburg (227a) and Naumann et al. (228).

A clear correlation between the supramolecular structure and endotoxic activity has been found recently using synchrotron radiation X-ray diffrac- tion and Fourier-transform infrared spectroscopy (229a). LPS or lipid A expressing lamellar structures were biologically inactive, whereas lipid A expressing a strong tendency to form nonlamellar (cubic or hexagonal) structures exhibited strong endotoxicity. It should be noted that at present it cannot be decided whether bioactivity of endotoxin is expressed by the aggregate described (229b) or by a single endotoxin molecule having a con-

Page 279: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

256 ULRICH ZAHRINGER et al.

formation which leads - at higher concentration - to the supramolecular structure observed. Recent studies appear to favor the concept that mono- meric structures are involved in the mediation of endotoxicity (229c,229d). b. Phase States (Fluidity).-We have previously shown that, in the S.

minnesota S- and R-form LPS series, the values of T, are lowest for Re mutant preparations (around 30°C) and that they increase with the length of the polysaccharide portion, being very high for S-form LPS, which demon- strates the T, around 37 -40°C (228,230). Surprisingly, free lipid A possesses the highest T, value (approximately 45°C).

The phase transition temperatures of the various mutant LPS and lipid A are significantly higher than those of the phospholipid mixture composing the inner leaflet of the outer membrane. At physiological temperature (37°C) membranes made of this mixture are exclusively in the a-state and, thus, show a high fluidity. In contrast, the phase-transition temperature of Re mutant LPS, S-form LPS, and lipid A are just below, in the range, or even above this temperature, respectively, leading to a low fluidity in membrane systems made up from these compounds. Therefore, it should be expected that the exclusive presence of LPS in the outer leaflet of the asymmetric outer membrane leads to a lower fluidity, thus contributing to its permeability- barrier function.

Addition of Mg2+ or Ca2+ and lowering of the pH cause a significant rigidification of the acyl chains of free lipid A and LPS preparations and lead to an increase in T,. Under alkaline conditions (pH > 7), a fluidization of the LPS and lipid A acyl chains, and a decrease in T,, takes place. Free lipid A and all R-mutant LPS preparations investigated exhibit an extremely strong lyotropic behavior, that is a strong dependence of the p - cx chain-melting transition on the water content. For example, the lipid A phase-transition is clearly observed (23 1) only at water concentrations higher than 50 - 60%.

VI. ENDOTOXOCITY OF LPS AND LIPID A LPS are endowed with an overwhelming spectrum of biological activities

expressed either in vivo after injection into mammalians or in humoral or cellular in vitro systems ( 1,3,4,6,20,118). In fact, LPS has been postulated as playing a central role in the pathogenesis and manifestations of Gram-nega- tive infection, in general, and of septic shock, in particular. Therefore, one of the driving forces in LPS research has been the search for that region and structure of the macromolecule that is responsible for the induction of the endotoxic reactions.

1. Lipid A, the Endotoxic Center of LPS Almost 40 years ago, Westphal and Luderitz postulated that the lipid A

component constitutes the endotoxic principle that is responsible for the

Page 280: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

MOLECULAR STRUCTURE OF LIPID A 257

induction of the manifold pathophysiological effects of LPS (28). This hy- pothesis was based, in part, on the finding that all LPS, independent of their bacterial origin, showed comparable endotoxic activity and that they all, despite their different polysaccharide compositions, contained lipid A. Dur- idg the past 20 years a large body of data confirming this idea has been accumulated, mainly through the work of Galanos et al., who showed, for example, that water-soluble, polysaccharide-free, free lipid A exhibits potent endotoxic activity (1,3 - 6,232). The successful chemical synthesis of lipid A and partial structures then offered the possibility of confirming this concept. By studying the biological activity of such synthetic preparations in compar- ison to natural products in various in vitro and in vivo systems, relationships between the chemical structure and biological activity could be established at the molecular level. Lethal toxicity, local Shwartzman reactivity, adju- vanticity, induction of interferon, tumor necrosis, endotoxin (cross) toler- ance, and, in particular, fever-inducing capacity (pyrogenicity) were mea- sured as typical in vivo endotoxin activities. The effectiveness of the preparations was tested in vitro in humoral systems (such as complement activation and gelation of the Limulus amebocyte lysate), and in cellular ones including murine B-lymphocyte mitogenicity and, in particular, acti- vation of macrophages associated with the release of such bioactive media- tors as prostaglandins, and leukotrienes, IL-l, IL-6, and TNFa (167,168,230,233).

The results from all biological assays performed showed that chemically synthesized E. coli lipid A (compound 506 or LA-15-PP) expresses, with similar doses, the same spectrum of endotoxic effects as bacterial (E. coli) free lipid A (5,234-237). Thus, lipid A constitutes the lethal, pyrogenic, leukopenic, and mediator-inducing, that is, the endotoxically essential re- gion of LPS, its endotoxic properties being embedded in a molecule having the structure shown in Fig. 2.

The lipid A of E. coli possesses a complex structure and the question arose as to which of its constituents is important for, or contributes to, bioactivity. To answer this question a great number of lipid A partial structures were analyzed, again in various in vivo and in vitro systems, for endotoxic activity. The results of the in vivo studies, with emphasis on fever-producing capacity, may be summarized as follows (1 16,118,235,238). HexpN monosaccharide compounds largely lack in vivo endotoxic activity [ pyrogenicity (rabbit) and lethal toxicity (mouse)]. GlcpN disaccharide structures carrying two phos- phate groups and seven (LA- 16-PP), five (LA-20-PP) or four (LA- 14-PP) fatty acids exhibit pyrogenic activity that, however, is weaker than that of E. coli lipid A (containing six fatty acids in asymmetric distribution). Com- pounds having only two fatty acids (LA- 19-PP) are completely inactive. Glucosamine disaccharide preparations carrying only one phosphate group (at either position 1 or 4’, see Fig. 2) are also less pyrogenic than E. colilipid A (containing two phosphate residues).

Page 281: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

258 ULRICH ZAHRINGER et al.

In vitro, the capacity of LPS, lipid A, and partial structures to induce the production of IL-1, IG6, and TNFa in human peripheral monocytes was analyzed. The results of these studies (167,168,183,239) were, in principle, similar to those obtained in vivo. Thus, hexosamine (GlcpN, GlcpN3N) monosaccharide structures were completely inactive. Glucosamine disac- charide compounds carrying two phosphate groups and seven (LA- 16-PP) or five (LA-20-PP)fatty acids exhibit lower activity than E. coli lipid A (six fatty acids), The importance of the location of fatty acids became obvious on analyzing hexaacyl compound LA-22-PP. This preparation contains, like E. coli lipid A, a p-( 1 + 6)-linked GlcpN-disaccharide and four primary 14 : O(3-OH) residues. As secondary fatty acids, 14 : 0 residues are linked to the 3-hydroxyl fatty acids at C-2’ and C-2, resulting in a C. violaceurn-type symmetric acylation pattern. Compound LA-22-PP, having six fatty acids with 14 carbon atoms was less active by a factor of lo4 in inducing IL-1 in peripheral monocytes ( 167). This result shows that not only the number of acyl groups but also their location (keeping the chain length constant) is an important factor in the expression of endotoxic activity. This has also been found to be true for the anomeric configuration of GlcN(1) as observed on investigating 2-phosphonooxyethyl derivatives of lipid A ( 1 5). The synthetic structures analyzed correspond to E. coIi lipid A except that, instead of the two 14 : O( 3-OH) residues at GlcN( I) two 14 : 0 groups are present and the glycosidically linked phosphate is replaced by an a- orp-linked 2-phosphono- oxyethyl group. As our preliminary results show (1 85) the a-anomeric com- pound was a potent inducer of TNFa, being comparable in activity to synthetic E. coli lipid A (compound 506). The p anomer, however, was less active by a factor of lo3. These data show that (i) the phosphate does not have to be directly linked to GlcN( I) for lipid A bioactivity (1 5 ) and, most impor- tant, (ii) that the a-anomeric configuration at GlcN(1) is essential for the expression of endotoxic activity of lipid A (1 85). The importance of the chain length of acyl groups in this respect has to be demonstrated.

Most interestingly, the tetraacyl precursor Ia (LA- 14-PP), as compounds having only two fatty acids (LA- 19-PP), completely lacked mediator-induc- ing activity. Monophosphated E. coli lipid A partial structures were also significantly less active than the corresponding bisphosphate compound LA- 15-PP. As a further point of interest, LPS, including LPS of enterobac- terial Re mutants was, in general, more active (by approximately a factor of 10) than free lipid A, suggesting that Kdo contributes to the induction of mediators (1 16,118,240).

VII. SEROLOGY OF LIPID A In the early seventies, Galanos et al. (241) showed that lipid A may be

immunogenic, and acid-treated bacteria (exposing free lipid A on their sur-

Page 282: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

MOLECULAR STRUCTURE OF LIPID A 259

face) or free lipid A complexed to suitable carriers were found to be good immunogens for the preparation of anti-lipid A antibodies. Such antisera could be raised with lipid A preparations derived from various Gram-nega- tive bacteria, provided they contained the E. coli- or C. violaceum-type of structure.

The specificity of lipid A antibodies was, however, not clearly defined until recently when Brade et al. (6,242 - 244b) could characterize epitopes present in E. coli-type lipid A by using bacterial and synthetic lipid A antigens and polyclonal anti-lipid A antisera. Thus far, eight different antigenic determi- nants have been defined, all residing in the hydrophilic lipid A backbone region. One epitope is composed of the 1,4’-bisphosphorylated p-( I - 6)- linked GlcpN disaccharide, a second of the 4’-phosphorylated, and a third of the 1 -phosphorylated disaccharide. The other two immunoreactive deter- minants are located in acylated D-gluco-HexpN 1 - and 4-phosphate mono- saccharides, respectively. In the latter instances, the specificity is also ex- pressed by acylated and phosphorylated preparations differing in the nature of the glycosyl residue (Glcp, GlcpN, GlcpN3N, and Glcp3N). The specific- ity of monosaccharide and disaccharide lipid A antibodies is dependent on the presence of phosphate groups but independent of the acylation pattern; that is, fatty acids are not part of the epitopes (243). Acyl groups, however, by influencing the three-dimensional organization of lipid A, may modulate the exposure of the epitopes.

In recent years, the field of research on lipid A antibodies has been rapidly expanding (6,245 -249). The interest in lipid A antibodies is based on the significance of LPS and lipid A in Gram-negative septic shock and the expectation that lipid A antibodies, being directed against a structure com- mon to all Gram-negative bacteria, which, at the same time, harbors the toxic principle, may be widely cross-reactive and possibly cross-protective. In fact, polyclonal anti-E. coli lipid A antibodies cross-react with a large variety of free lipid A of distinct bacterial origin, namely, those lipid A containing ap-( 1 4 6)-linked HexpN disaccharide bisphosphate. It must be emphasized, however, that these lipid A antibodies do not cross-react, at least in vitro, with LPS, that is, with the lipid A component carrying the saccharide portion. Thus, in LPS either the lipid A epitopes are not expressed (that is, free lipid A constitutes a neoantigen exposing structural or confor- mational determinants not present in LPS) or the epitopes are cryptic (that is, Kdo or other LPS constituents sterically hinder the binding of antibodies to lipid A). There are, however, reports in the literature of mouse and human monoclonal antibodies, in particular of the IgM class, that appear to be directed against lipid A and that are reported to cross-react with different LPS (250-25 la). Their reactivity was elucidated in the ELISA assay system, but it is known that ELISA determinations, in particular of such amphiphilic molecules as lipid A and of sticky antibodies such as those of the IgM class,

Page 283: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

260 ULRICH ZAHRINGER et al.

may, unless certain precautions are taken, lead to false positive results simu- lating cross-reactivity (252). These IgM antibodies are reported to exhibit protective activity in septicemic states (250,25 1) but one of these studies has received critical comments (244a,253). It is evident, therefore, that further studies are required in order to understand their mechanism ofaction and, in particular, to define unequivocally the epitopes that they recognize.

VIII. SYNOPSIS: THE STRUCTURE, ACTIVITY, AND FUNCTION OF LIPID A

As the preceding sections show, the term lipid A does not denote a defined molecular entity but rather a family of structurally closely related but not identical phosphoglycolipids. Their structures may differ in the type and number (one or two) of HexpN residues present in the backbone; by the number, location, nature, size, and linkage of acyl residues; by the nature of phosphate substituents; and finally by the degree of phosphorylation of the backbone. Lipid A components of different bacterial origin are classified in the present article on the basis of structural variations.

Despite these variations, lipid A of different origin not only share many identical constituents, but also are constructed according to a similar archi- tectural principle (Fig. 15). Setting aside those few preparations containing a monosaccharide backbone, lipids A contain ap-( 1 + 6)-linked disaccharide having the D-gluco configuration and pyranosidic D-hexosamine residues, which carry an a-glycosidic and, at position C-4’, a nonglycosidic phosphate group. This lipid A backbone is substituted at the amino and hydroxyl groups in positions 3’, 2’, 3, and 2 by (R)-3-hydroxy fatty acids, up to three of which are acylated at their 3-hydroxyl group by secondary fatty acids. Lipid A expressing strongest endotoxicity contains six fatty acids, and, in this case, the secondary fatty acids are bound to the 3-hydroxy fatty acid either at position 2‘ and 3‘ as in E. coli, or at position 2’ and 2, as in C. violaceurn. In the latter instance, however, the chain length of the primary fatty acid is lower than that in E. coli. Figure 15 shows the structure of the two hexaacyl lipid A types that are the most abundant among lipid A preparations studied thus far. The two lipid A types differ only in the location of one secondary fatty acid (designated R1 or R2 in Fig. 15) and in the chain length of the primary fatty acids. Attachment of one acyl group to the 3-hydroxy fatty acid at position 2 [GlcN(I)] yields heptaacyl lipid A (see Fig. 4), whereas penta- acyl lipid A (Figs. 5,7, 8, and 9B) formally result from the omission of any one of the secondary acyl groups of the structures shown in Figs. 2 and 10. As comparison of the structures shows, the 3-hydroxyl group of the primary fatty acid at position 3 is never acylated, whereas that of the 3-hydroxy fatty acid at position 2’ is generally acylated. Other lipid A structures are created by replacing GlcpN by GlcpN3N, or by omitting the 4’-phosphate group.

Page 284: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

MOLECULAR STRUCTURE OF LIPID A 26 1

HO -OH

FIG. 15. -Structure of the Kdo-lipid A domain. The domain is structurally highly conserved and constitutes the biologically active region of LPS. R’, RZ = secondary fatty acids: In hexa- acyl E. coli-type lipid A (m = n = 14), R2 = fatty acid, R‘ = H; in hexaacyl C. violaceum-type lipid A (m = 10 or 12, n = 12 or 14), RZ = H, R’ =fatty acid. R3, R4 = H or substituent of backbone phosphate groups (see Table I). R5 = negatively charged groups: Kdo [in Salmonella (70) and E. coli (34)], P03Hz (in Haemophilus injluenzae (68), Bordetella pertussis, Vibrio parahaemolyticus (83), Bacteroidesgingivalis (268), Vibrio cholerae (269) GalpA [in Rhodocy- clus gelatinosus Dr2 (270)], and GlcpA [in Rhizobium sphaeroides (165)l. R6 = core region: L-glycerm-D-manno-Hep (Enterobacteriaceae, Neisseriaceae, and other bacteria). D-glycero- a-D-mannc-Hepp [Rhodocyclus gelatinosus Dr2(270)], a-~-Glcp 4-P03H, [Acinetobacter cal- coaceticus (75)], and a-D-Man [Rhizobium trifolii ANU 843 (271)].

The hydroxyl group at C-4 [GlcN( I)] is generally not substituted, whereas in almost all Gram-negative bacteria studied in detail, the primary 6’-hy- droxyl group cames Kdo (or the isosteric DgDt-KO) in a-ketosidic linkage (Fig. 15). The universal presence and the structural conservation of the Kdo and lipid A components led us earlier to propose that this LPS domain should not be viewed separately but rather as one structural and functional molecular entity ( 1 18). Thus, the Kdo - lipid A domain constitutes the mini- mal structure that bacteria require for growth and multiplication and that expresses full endotoxic activity. It should be realized that the separate dis- cussion of the lipid A and Kdo regions is justified merely by one chemical feature of LPS, namely, the peculiar susceptibility of the Kdo-lipid A link-

Page 285: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

262 ULRICH ZAHRINGER et al.

age to acid hydrolysis and, thus, the ready dissociation ofthe Kdo-containing core region from the lipid A component, but not by genetic, biological or other properties. In Enterobacteriaceae, the Kdo-lipid A domain also forms biosynthetically one molecular entity. As elucidated in E. coli and Salmo- nella, UDP-N-acetyl-D-GlcpN is, after addition of two 14 : O(3-OH) residues (1 9 1,254), condensed with 2,3-bisacyl-~-GlcpN 1-phosphate to form a te- traacyl disaccharide 1-phosphate (a reaction catalyzed by the enzyme lipid A synthase). This molecule is then phosphorylated at position 4’ to yield pre- cursor Ia of lipid A biosynthesis (255). To this precursor, Kdo is transferred before the completion of the lipid A structure by addition of 12 : 0 and 14 : 0 [In Pseudomonas, however, mature lipid A is formed first, and Kdo is then added in a subsequent step (26)]. The lipid A synthase also accepts UDP-ac- tivated and acylated GlcpN3N. It is, therefore, understandable that back- bone structures containing a &( 1 - 6)-linked GlcpN3N disaccharide or hybrid disaccharides such as GlcpN3N-GlcpN are likewise formed in vitro ( 13) and in vivo (see Section 111,l .c.iii).

Lipid A having Kdo attached at 0-6’ comprises a domain of LPS that exhibits a remarkable structural conservatism, as far as the principal archi- tecture is concerned (Fig. 15). This type of structure is found in most Gram- negative bacteria. It is not present in other biomolecules and is thus unique to Gram-negative bacteria and their LPS. It contains those structures that are required for (i) bacterial viability and (ii) for optimal expression of endotoxic activity.

Thus far, viable (that is growing and multiplying) Gram-negative bacteria lacking LPS have not been isolated in nature, nor have they been produced in the test tube. This observation indeed suggests that LPS is essential for the viability of this class of microbes. In the bacterial cell, LPS is located in the outer membrane and it may be assumed that the vital significance of LPS is related to the successful assembly, architecture, and function of this outer membrane. As discussed, isolated LPS in aqueous solution may self-organ- ize to a membrane structure that, as revealed by electron microscopy (256,257), resembles fragments of the bacterial membrane. Artificially cre- ated LPS membranes may, therefore, serve as objects for studying the archi- tecture and function of the outer membrane of Gram-negative bacteria. As X-ray diffraction studies have shown, lipid A tends to be organized in large domains comprising of up to 1200 molecules, which are partly arranged in parallel and which persist for long periods of time (220). As conformational calculations have revealed, the lipid A fatty acids are tightly packed in a dense two-dimensional hexagonal lattice, the formation of which is favored by the absence of unsaturated fatty acids (220,22 1,228). In this respect, LPS membranes differ significantly from other biological membranes, which, at physiological temperature, are more fluid and in which the phospholipids

Page 286: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

MOLECULAR STRUCTURE OF LIPID A 263

possess rotational freedom. The high degree of order of LPS assemblies and the nearly crystalline nature of the lipid A fatty acid arrangement yields a well-ordered and rigid membrane system that may prevent hydrophobic molecules from diffusing into or through an LPS layer system as is present in the outer leaflet of the Gram-negative outer membrane (258). Indeed it is known that the outer membrane of enteric bacteria is relatively imperme- able for certain hydrophobic antibiotics, detergents, hydrophobic dyes, and bile acids (259,260). Thus, the outer membrane serves as a permeation barrier and, thereby, protects the bacterial cell from external toxic mole- cules. It is probable that the highly ordered LPS molecule, and, in particular, the lipid A component, is largely responsible for the permeation-barrier properties of the outer membrane (258). The Kdo residues carrying negative charges and the phosphate groups of lipid A are of importance for the organization and function of the outer membrane in that they constitute high- and low-affinity binding-sites for Mg2+ and Ca2+ ions (65). The Kdo- lipid A domain thus facilitates the accumulation ofbivalent ions essential for the stability and proper function of the membrane, as well as probably for the activity ofenzymes and integral proteins (such as porines) present in the cell or the outer membrane, respectively.

On the other hand, the negatively charged Kdo -lipid A domain consti- tutes a target for binding of certain antibiotics, such as polymyxin B (125) and basic peptides derived from host cells such as the bactericidal/perme- ability-increasing protein (BPI) (261). In this respect the Kdo-lipid A do- main constitutes a weak site of the outer membrane, rendering bacteria vulnerable to the action of cationic agents.

Endotoxic activity (including pyrogenicity and mediator induction) is, according to present knowledge, optimally expressed by a molecule contain- ing two j3-( 1 + 6)-linked HexpN residues of the D-gluco configuration, two phosphate groups, and six fatty acids (saturated and, in part, 3-hydroxyl- ated), including 3-acyloxyacyl groups with the chain length, in the linkages and the locations present in E. coli- (Fig. 2) or C. violaceurn-type lipid A (Fig. 6) and exemplified in Fig. 15. Most interestingly, none of the synthetic compounds thus far available exhibits stronger endotoxicity than natural or synthetic E. coli-type lipid A. Partial structures lacking one constituent, or molecules having a different distribution of constituents, are endotoxically less active or inactive an exception being the 2-phosphonooxyethyl deriva- tive ( 15,185) of compound 506. Thus, slight modifications at any site of the E. coli lipid A architecture result in a significant decrease of biological activity, showing that endotoxicity is not dependent on one single lipid A constituent or a toxophore group. The role of fatty acids appears to be of particular importance. As discussed, the number of acyl groups is a decisive factor for endotoxic activity, six fatty acids being optimal. However, the

Page 287: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

264 ULRICH ZAHRINGER et al

chain length of (the primary) fatty acids also plays an important role, as indicated from biological analyses of E. coli and C. violaceurn lipid A and also of lipid A having particularly long acyl chains [such as C. psittaci (1 33) and B. fragilis (79)], and shorter ones [P. aeruginosa (77) and reduced R. sphaeroides (162)J. It appears that the expression of endotoxic activity of lipid A requires a certain equilibration of hydrophilic (bisphosphated, p- ( 1 ---* 6)-linked gluco-configured HexpN backbone) and hydrophobic (de- fined number of fatty acids having a specific chain length) regions. The peculiar arrangement of lipid A constitutents, generating a unique chemical architecture, results in the formation of a peculiar conformation (endotoxic conformation) and a particular supramolecular organization, which allows the expression of endotoxic activity. This conformation is likely to be in- fluenced by Kdo, as the up-regulating effect of the negatively charged inner core region on lipid A bioactivity shows.

It is most remarkable that precursor Ia (Fig. 9 A) which differs from E. coli lipid A (Fig. 2) only by the absence of two secondary acyl residues ( 12 : 0 and 14 : 0), thus lacking 3-acyloxyacyl groups, is completely inactive in stimulat- ing human mononuclear cells to release bioactive mediators. This observa- tion points to an extreme structural specificity ofthe expression of endotoxic activity by lipid A. In turn, this factor also strongly suggests that a specific binding-protein or a specific receptor is involved in the interaction between LPS or lipid A and such responsive target cells as monocytes. The existence of lipid-A-binding and monocyte/macrophage-associated proteins has been demonstrated (262 - 264). In accord with this concept, we and others found that the tetraacyl precursor Ia strongly competes with the specific binding of labeled LPS to monocytes and that it inhibits, as does R. sphaeroides ( 182) and R. capsulatus (265) LPS (probably via an antagonistic mechanism), the production of LPS or free lipid A-induced mediator in human mono- cytes( 167,169,183,184,266). Also the presence of a Kdo-specific receptor on LPS target cells has been postulated (240,267).

Studies along these lines appear important, as they may lead to new strategies for pharmacological control of endotoxin effects and because they provide deeper insight into the mechanisms involved in the interaction of endotoxins with the host. It should be mentioned that, on biodegradation of E. coli lipid A by macrophages and granulocytes, the two secondary fatty acids 12 : 0 and 14 : 0 are removed by the enzyme 3-acyloxyacylhydrolase, yielding (1 8 1 ) a structural counterpart of precursor Ia. As discussed, this compound not only lacks endotoxic activity in the human system, but also constitutes a potent endotoxin antagonist. Thus, on enzymic degradation of lipid A, a molecule (precursor Ia) is formed, which not only is detoxified, but also counteracts endotoxin bioactivity ( 1 8 1).

Page 288: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

MOLECULAR STRUCTURE OF LIPID A 265

Bacterial endotoxins, and, in particular, their lipid A component, because of their complex structure, their significance for bacterial viability, and their important role in Gram-negative infection and sepsis, have fascinated re- searchers in the field of chemistry, physics, biology, immunology, genetics, microbiology, pharmacology, and clinical medicine for several decades. It was through the efforts of these scientists that extensive molecular knowl- edge on various aspects on the chemistry and biology of lipid A was acquired. It is hoped that in the future advantage will be taken of this knowledge in application to the immunological and also pharmacological control of Gram-negative sepsis and associated local and generalized inflammatory reactions caused by endotoxins and their lipid A component.

ACKNOWLEDGMENTS

The financial support of the Deutsche Forschungs Gemeinschaft (SFB 367, project B2) and Fonds der Chemie (EThR) is greatly appreciated. We thank M. Lohs, F. Richter, B. Kohler and I. Stegelmann-Muller for preparing drawings and photographs. We are particularly grateful to our friends and colleagues H. Brade, E. Coats, H.-D. Grimmecke, 0. Holst, M. Kastowsky, S. Kusumoto, H. Labischinski, 0. Liideritz, H. Mayer, G. Seltmann, and U. Seydel for valuable suggestions.

REFERENCES

1 . C. Galanos, 0. Liideritz, E. Th. Rietschel, and 0. Westphal in T. W. Goodwin. (Ed.), International Review ofBiochemistry, Vol. 14, Biochemistry ofLipids 11: Newer Aspects of the Chemistry and Biology of Bacterial Lipopolysaccharides, with Special Reference to Their Lipid A Component, p. 239. University Park Press, Baltimore, 1977.

2. 0. Westphal, 0. Liideritz, E. Th. Rietschel, and C. Galanos, Biochem. Rev., 9 (1981)

3. 0. Liideritz, M. A. Freudenberg, C. Galanos, V. Lehmann, E. Th. Rietschel, and D. H. Shaw, Curr. Top. Membr. Tramp.. 17 (1982) 79-151.

4. E. Th. Rietschel, C. Galanos, 0. Luderitz, and 0. Westphal, in D. R. Webb. (Ed.), Immunopharmacology and the Regulation of Leukocyte Function: The Chemistry and Biology of Lipopolysaccharides and Their Lipid A Component, p. 183. Dekker, New York/Basel, 1982.

5. E. Th. Rietschel, H. Brade, L. Brade, K. Brandenburg, U. F. Schade, U. Seydel, U. Ziihringer, C. Galanos, 0. Liideritz, 0. Westphal, H. Labischinski, S. Kusumoto, and T. Shiba, Prog. Clin. Biol. Rex, 231 (1987) 25-53.

191-195.

6. H. Brade, L. Brade, and E. Th. Rietschel, Zbl. Bakt. Hyg., A268 (1 988) 15 1 - 179. 7. C. R. H. Raetz, Annu. Rev. Biochem., 59 (1990) 129-170. 8. E. Th. Rietschel and H. Brade, Sci. Am., 267 (1992) 54-61. 9. E. Th. Rietschel, L. Brade, B. Lindner, and U. Zilhringer, in D. C. Morrison and J. L.

Ryan. (Eds.), Bacterial Endotoxic Lipopolysaccharides: Biochemistry of Lipopolysaccha- rides, p. 3. CRC Press, Boca Raton, FL, 1992.

9a. E. Th. Rietschel, T. Kirikae, F. U. Schade, U. Mamat, G. Schmidt, H. Loppnow, A. J. Ulmer, U. Zahringer, U. Seydei, F. Dipadova, M. Schreier, and H. Brade, FASEB J., 8 (1994) 217-225.

Page 289: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

266 ULRICH ZAHRINGER et al.

9b. E. Th. Rietschel, T. Kirikae, F. U. Schade, A. J. Ulmer, 0. Holst, H. Brade, G. Schmidt, U. Mamat, H.-D. Grimmecke, S. Kusumoto, and U. Zaringer, Immunobiol., 187 (1993) 169- 190.

10. C.R. Alving,J. Immunol.Meth., 140(1991) 1-13. 11. M. Kiso, S . Tanaka, M. Tanahashi, Y. Fujishima, Y. Ogawa, and A. Hasegawa, Carbo-

12. P. Westerduin, J. H. van Boom, C. A. A. van Boeckel, and T. Beetz, Carbohydr. Rex, 137

13. P. Stiitz, H. Aschauer, J. Hildebrandt, C. Lam, H. Loibner, I. Macher, D. Scholz, E. Schiitze, and H. Vyplel, in A. Nowotny (Ed.), Endotoxin Research Series, Vol. 1, Cel- lular and Molecular Aspects of Endotoxin Reactions: Chemical Synthesis of Endotoxin Analogs and Some Structure Activity Relationships, p. 129. Elsevier, Amsterdam, 1990.

14. S. Kusumoto, N. Kusunose, M. Imoto, T. Shimamoto, T. Kamikawa, H. Takada, S. Kotani, E. Th. Rietschel, and T. Shiba, Pure Appl. Chem., 6 1 (1989) 46 1 -464.

15. S. Kusumoto, T. Kamikawa, M. Kurosawa, K. Fukase, T. Shiba, T. Kusama, T. Soga, E. Shioya, K. Nakayama, H. Nakayima, Y. Osada, and Y. Ono, in A. Nowotny, J. J. Spitzer, and E. J. Ziegler (Eds.), Endotoxin Research Series, Vol. 1, Cellular and Molecular Aspects of Endotoxin Reactions: New Results in the Synthesis of Lipid A and Endotoxins, p. 41. Elsevier Science Publishers, Amsterdam, 1990.

16. M. Kiso, S. Tanaka, M. Fujita, Y. Fujishima, Y. Ogawa, H. Ishida, and A. Hasegawa, Carbohyr. Res., 162 (1987) 127-140.

17. D. Charon, C. Diolez, M. Mondange, S. R. Sarfati, L. Szabo, P. Szab6, and F. Trigalo, Am. Chem. SOC. Symp. Ser., 231 (1983) 301-316.

18. T. Shimizu, Y. Ohtsuka, Y. Yanagihara, I. Hajime, S. -I. Nakamoto, and K. Achiwa, Znt. J. Immunopharmacol., 13 (1991) 605-611.

19. S . Ikeda, M. Matsuura, M. Nakatsuka, J. Y. Homma, M. Kiso, A. Hasegawa, and C. Nishimura, J. Clin. Lab. Immunol., 32 (1990) 177- 181.

20. D. C. Momson and J. L. Ryan, Annu. Rev. Med., 38 (1987) 417-432. 21. M. Pollack, in G. L. Mandell, R. G. Douglas, and J. E. Bennet (Eds.), Principles and

Practice of Infection Diseases, Update 8, New Therapeutic Strategies in Gram-Negative Sepsis and Septic Shock Based on Molecular Mechanism of Pathogenesis, p. 1. Churchill Livington, New York, 1990.

22. J. M. Griffiss, H. Schneider, R. E. Mandrell, R. Yamasaki, G. A. Jarvis, J. J. Kim, B. W. Gibson, R. Hamadeh, and M. A. Apicella, Rev. Inf Dis., 10 (1988) s287-s295.

23. 0. Holst and H. Brade, in D. C. Momson and J. L. Ryan (Eds.), Bacterial Endotoxic Lipopolysaccharides: Chemical Structure of the Core Region of Lipopolysaccharides. p. 135. CRC Press, Bow Raton, FL, 1992.

24. 0. Holst, E. Rohrscheidt-Andrzejewski, H. Brade, and D. Charon, Carbohydr. Res., 204

25. E. Th. Rietschel, H. -W. Wollenweber, H. Brade, U. Dhringer, B. Lindner, U. Seydel, H. Bradaczek, G. Barnickel, H. Labischinski, and P. Giesbrecht, in E. Th. Rietschel, (Ed.), Handbook of Endotoxin, Vol. 1, Chemistry of Endotoxin: Structure and Conformation of the Lipid A Component of Lipopolysaccharides, p. 187. Elsevier, Amsterdam, 1984.

26. R. C. Goldman, C. C. Doran, S. K. Kadam, and J. 0. Capobianco, J. Biol. Chem., 263

27. A. Boivin, J. Mesrobeanu, and L. Mesrobeanu, Compt. Rend. Sci. Biol., 114 (1933)

28. 0. Westphal and 0. Liideritz, Agnew. Chem., 66 (1954) 407-417. 29. W. T. J. Morgan and S. M. Partridge, Biochem. J., 35 (1941) 1140- 1163. 30. 0. Liideritz, K. Jann, and R. Wheat, in M. Florkin and E. H. Stotz (Eds.), Comprehensive

hydr. Res., 148 (1986) 221-234.

(1985) ~ 4 - ~ 7 .

(1990) 93-102.

(1988) 5217-5223.

307- 3 10.

Page 290: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

MOLECULAR STRUCTURE OF LIPID A 267

Biochemistry, Vol. 26A, Somatic and Capsular Antigens of Gram-Negative Bacteria, p. 105. Elsevier, Amsterdam, 1968.

31. R. S. Johnson, G.-R. Her, J. Grabarek, J. Hawiger, and V. N. Reinhold, J. Biol. Chem.,

32. N. Qureshi, K. Takayama, and E. E. Ribi, J. Biol. Chem., 257 (1982) 1 1808 - 1 18 15. 33. K. Takayama, N. Qureshi, K. Hyver, J. Honovich, R. J. Cotter, P. Mascagni, and H.

34. U. Zahringer, B. Lindner, U. Seydel, E. Th. Rietschel, H. Naoki, F. M. Unger, M. Imoto,

35. M. Imoto, T. Shiba, H. Naoki, T. Iwashita, E. Th. Rietschel, H. -W. Wollenweber, C.

36. R. J. Cotter, J. Honovich, N. Qureshi, and K. Takayama, Biomed. Mass Spectrom., 14

37. U. Seydel, B. Lindner, H. -W. Wollenweber, and E. Th. Rietschel, Eur. J. Biochem., 145

38. H.-W. Wollenweber, U. Seydel, B. Lindner, 0. Luderitz, and E. Th. Rietschel, Eur. J. Biochem., 145 (1984) 265-272.

39. B. Lindner, U. ahringer, E. Th. Rietschel, and U. Seydel, in A. Fox, S. L. Morgan, L. Larsson, and G. Odham (Eds.), Analytical Microbiology Methods: Structural Elucidation of Lipopolysaccharides and Their Lipid A Component: Application of Soft Ionization Mass Spectrometry, p. 149. Plenum, New YorkfLondon, 1990.

40. U. Seydel, M. Haas, E. Th. Rietschel, and B. Lindner, J. Microb. Meth., 15 (1992)

41. N. Qureshi, K. Takayama, D. Heller, and C. Fenselau, J. Biol. Chem., 258 (1983) 12947-

42. M. Caroff, C. Deprun, D. Karibian, and L. Szab6, J. Biol. Chem., 266 (1991) 18543-

43. R. Wang, L. Chen, R. J. Cotter, N. Qureshi, and K. Takayama, J. Microb. Meth., 15

44. D. Karibian, C. Deprun, L. Szabb, Y. Le Beyec, and M. Caroff, Int. J. Mass Spectrom. Ion

45. A. L. Erwin and R. S. Munford, Jr., J. Biol. Chem., 265 (1990) 16444- 16449. 46. M. R. Rosner, H. G. Khorana, and A. C. Satterthwait, J. Biol. Chem., 254 (1979) 5918-

47. B. Jiao, M. A. Freudenberg, and C. Galanos, Eur. J. Biochem.. 180 (1989) 5 15 - 5 18. 48. W. Fischer, Eur. J. Biochem., 194 (1990) 655-661. 49. J. Weckesser and H. Mayer, FEMSMicrobiol. Lett., 54 (1988) 143-154. 50. H. Mayer, J. H. Krauss, A. Yokota, and J. Weckesser, in H. Friedman, T. W. Klein, M.

Nakano, and A. Nowotny (Eds.), Endotoxin: Natural Variants ofLipid A, p. 45. Plenum, New York, 1990.

265 (1990) 8108-8116.

Schneider, J. Biol. Chem., 261 (1986) 10624-10631.

S. Kusumoto, and T. Shiba, Tetrahedron Lett., 26 (1985) 6321 -6324.

Galanos, and 0. Liideritz, Tetrahedron Lett., 24 (1983) 4017-4020.

(1987) 591 -598.

(1984) 505-509.

167-183.

12951.

18549.

(1992) 151-166.

Processes, 11 I (1991) 273-286.

5925.

51. J. Montreuil, Adv. Curbohydr. Chem. Biochem., 37 (1980) 157-223. 52. A. Nowotny, J. Am. Chem. Soc., 83 (1961) 501-503. 53. A. J. Burton and H. E. Carter, Biochemistry, 3 (1964) 41 1-418. 54. J. Gmeiner, 0. Liideritz ,and 0. Westphal, Eur. J. Biochem., 7 (1969) 370-379. 55. A. B. Foster and D. Horton, J. Chem. SOC., 384 (1958) 1890- 1894. 56. G. A. Adams and P. P. Singh, Biochim. Biophys. Acta. 187 (1969) 457-459. 57. S. Hase and E. Th. Rietschel, Eur. J. Biochem., 63 (1976) 101-107. 58. S. Hase and E. Th. Rietschel, Eur. J. Biochem., 63 (1976) 93-99. 59. M. Jensen, D. Borowiak, H. Paulsen, and E. Th. Rietschel, Biomed. Mass Spectrom., 6

60. K. Takayama, N. Qureshi, and P. Mascagni, J. Biol. Chem., 258 (1983) 12801- 12803. (1979) 559-565.

Page 291: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

268 ULRICH ZAHRINGER et al.

61. D. Blache, M. Bruneteau, and G. Michel, Biochimie (Paris), 62 (1980) 191 - 194. 62. I. N. Krasikova, V. I. Gorbach, V. V. Isakov, T. F. Solov'eva, and Y. S. Ovodov, Eur. J.

63. S . M. Strain, I. M. Armitage, L. Anderson, K. Takayama, N. Qureshi, andC. R. H. Raetz,

64. A. Neszmelyi, P. Kosma, R. Christian, G. Schulz, and F. M. Unger, Carbohydr. Res., 139

64a. U. Ziihringer, V. Sinnwell, J. Peter Katalinic, E. Th. Rietschel, and C. Galanos, Tetrahe-

65. S. M. Strain, S. W. Fesik,andI. M. Armitage,J. Biol. Chem., 258(1983) 13466-13477. 66. S. M. Strain, S. W. Fesik, and I. M. Armitage, J. Biol. Chem., 258 (1983) 2906-2910. 66a. 0. Holst, S. Miiller-Loennies, B. Lindner, and H. Brade, Eur. J. Biochem., 214 (1993)

67. J. Radziejewska-Lebrecht, U. R. Bhat, H. Brade, and H. Mayer, Eur. J. Biochem., 172

68. I. M. Helander, B. Lindner, H. Brade, K. Altmann, A. A. Lindberg, E. Th. Rietschel, and

69. M. R. Rosner, J. Tang, I. Barzilay, and H. G. Khorana, J. Biol. Chem., 254 (1979)

70. R. Christian, G. Schulz, P. Waldstiitten, and F. M. Unger, Tetrahedron Lett., 25 (1984)

7 1. M. Jensen, Strukturanalyse der Lipoid A Komponente von Lipopolysacchariden aus Yer-

72. Z. Sidorczyk, U. mringer , and E. Th. Rietschel, Eur. J. Biochem., 137 (1983) 15-22. 72a. A. K. Harrata, L. N. Domelsmith, and R. B. Cole, Biological Mass Spectrometry, 22

72b. R. B. Cole, L. N. Domelsmith, C. M. David, R. A. Laine, and A. J. DeLucca, Rapid

73. V. A. Kulshin, U. ahringer, B. Lindner, C. E. Frasch, C. -M. Tsai, B. A. Dmitriev, and E.

74. V. L. L'vov, I. K. Verner, L. Y. Musina, A. V. Rodionov, A. V. Ignatenko, and A. S.

75. K. Kawahara, H. Brade, E. Th. Rietschel, andU. ahringer, Eur. J. Biochem., 163 (1987)

76. D. T. Drewry, J. A. Lomax, G. W. Gray, and S. G. Wilkinson, J. Biochem. (Tokyo), 133

77. V. A. Kulshin, U. ahringer, B. Lindner, K. -E. Jeer , B. A. Dmitriev, and E. Th.

77a. D. N. Kurunaratne, J. C. Richards, and R. E. W. Hancock, Arch. Biochem. Biophys. 299

78. E. Th. Rietschel, 0. Liideritz, and W. A. Volk, J. Bacteriol., 122 (1975) 1180- 1188. 79. A. Weintraub, U. ZAhringer, H. -W. Wollenweber, U. Seydel, and E. Th. Rietschel, Eur. J.

80. S. Haw, T. Hofstad, and E. Th. Rietschel, J. Bacteriol., 129 (1977) 9- 14. 80a. T. Ogawa, FEBS Lett., 332 (1993) 197-201. 81. R. Russa, 0. Liideritz, and E. Th. Rietschel, Arch. Microbiol., 141 (1985) 284-289. 82. T. Urbanik-Sypniewska, U. Seydel, M. Greck, J. Weckesser, and H. Mayer, Arch. Micro-

83. S. Kondo, U. ZAhringer, U. Seydel, K. Hisatsune, and E. Th. Rietschel, Eur. J. Biochem.,

Biochem., 126 (1982) 349-351.

J. Biol. Chem., 260 (1985) 16089-16098.

(1985) 13-22.

dron, 49 (1993) 4193-4200.

695 -701.

(1988) 535-541.

U. Z&ringer, Eur. J. Biochem., 177 (1988) 483-492.

5906-5017.

3433 - 3436.

sinia enterocolitica, Doctoral Thesis, University of Freiburg, 1980,

(1993) 59-67.

Communications in Mass Spectrometry, 6 (1992) 6 16-622.

Th. Rietschel, J. Bacteriol., 174 (1992) 1793- 1800.

Shashkov, Arch. Microbiol., 157 (1 992) 13 1 - 134.

489-495.

(1973) 563-572.

Rietschel, Eur. J. Biochem., 198 (1991) 697-704.

(1992) 368-376.

Biochem., 183 (1989) 425-431.

biol., 152 (1989) 527-532.

200 (1991) 689-698.

Page 292: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

MOLECULAR STRUCTURE OF LIPID A 269

84. H. Masoud, B. Lindner, J. Weckesser, and H. Mayer, System. Appl. Microbiol., 13 (1990)

85. S. Hase and E. Th. Rietschel, Eur. J. Biochem., 75 (1977) 23-34. 86. K. W. Broady, E. Th. Rietschel, and0. Liideritz, Eur. J. Biochem., 115 (1981)463-468. 87. P. V. Salimath, J. Weckesser, W. Strittmatter, andH. Mayer, Eur. J. Biochem., 136 (1983)

88. N. Qureshi, J. Honovich, H. Hara, R. J. Cotter, and K. Takayama, J. Biol. Chem., 263

89. J. H. Krauss, U. Seydel, J. Weckesser, and H. Mayer, Eur. J. Biochem., 180 (1989)

90. R. N. Tharanathan, J. Weckesser, and H. Mayer, Eur. J. Biochem., 84 (1978) 385-394.

227-233.

195 - 200.

(1988) 5502-5504.

5 19 - 526.

91. 0. Holst, D. Borowiak, J. Weckesser, and H. Mayer, Eur. J. Biochem., 137 (1983) 325 - 332.

92. H. Masoud, T. Urbanik-Sypniewska, B. Lindner, J. Weckesser, and H. Mayer, Arch.

93. M. Caroff and L. SzabB, Carbohydr. Rex, I14 (1983) 95- 102. 94. H. Masoud, S. T. Weintraub, R. Wang, R. Cotter, and S. C. Holt, Eur. J. Biochem., 200

95. S . Fukuoka, H. Kamishima,Y. Nagawa, H. Nakanishi,K. Ishikawa,Y. Niwa, E. Tamiya,

96. J. Roppel, H. Mayer, and J. Weckesser, Carbohydr. Res., 40 (1975) 31 -40. 97. A. P. Moran, U. e n g e r , U. Seydel, D. Scholz, P. Stutz, and E. Th. Rietschel, Eur. J.

Biochem., 198 (1991) 459-469. 98. H. Mayer, U. R. Bhat, H. Masoud, J. Radziejewska-Lebrecht, C. Widemann, and J. H.

Krauss, Pure Appl. Chem., 61 (1989) 1271 - 1282. 99. A. Yokota, M. Rodriguez, Y. Yamada, K. Imai, D. Borowiak, and H. Mayer, Arch.

Microbiol., 149 (1987) 106-111. 100. H. Mayer, J. H. Krauss, T. Urbanik-Sypniewska, V. Puvanesarajah, G. Stacey, andG.

Auling, Arch. Microbiol., 151 (1989) I 1 1 - 116. 101. H. Mayer, S. A. Campos-Portuguez, M. Busch, T. Urbanik-Sypniewska, and U. R. Bhat,

in A. Nowotny, J. J. Spitzer, and E. J. Ziegler (Eds.), Endotoxin Research Series, Vol. 1, Cellular and Molecular Aspects of Endotoxin Reactions: Lipid A Variants- Or, How Constant Are the Constant Regions in Lipopolysaccharides?p. 1 1 I . Elsevier, Amsterdam, 1990.

Microbiol., 156 (1991) 167-175.

(1991) 775-779.

andI. Karube, Arch. Microbiol., 157 (1992) 311-318.

102. S. G. Wilkinson and D. P. Taylor, J. Gen. Microbiol., 109 (1978) 367-370. 103. N. Kasai, S. Arata, J. -I. Mashimo, Y. Akiyama, C. Tanaka, K. Egawa, and S. Tanaka,

103a. N. Ravenscroft, S. G. Walker, G. G. S. Dutton, and J. Smith, j . Bacteriol., 174 (1992)

104. G. Keilich, J. Roppel, and H. Mayer, Carbohydr. Res., 5 1 (1976) 129- 134. 105. R. Weisshaarand F. Lingens, Eur. J. Biochem., 137 (1983) 155-161. 106. R. I. Hollingsworth and R. W. Carlson, J. Biol. Chem., 264 (1989) 9300-9303. 107. U. R. Bhat, H. Mayer, A. Yokota, R. I. Hollingsworth, and R. W. Carlson, J. Bacteriol.,

108. Biological Properties of Peptidoglycan, P. H. Seidl and K. H. Schleifes (Eds.), Walter de

109. M. Batley, N. H. Packer, and J. W. Redmond, Biochemistry, 21 (1982) 6580-6586. 1 10. N. Qureshi, K. Takayama, P. Mascagni, J. Honovich, R. Wong, and R. J. Cotter, J. Biol.

1 1 1. M. Caroff, A. Tacken, and L. Szab6, Carbohydr. Res., 175 (1988) 273-282.

Biochem. Biophys. Res. Commun., 142 (1987) 972-978.

7595-7605.

173 (1991) 2155-2159.

Gruyter, Berlinmew York, 1986.

Chem., 263 (1988) 11971-11976.

Page 293: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

270 ULRICH ZAHRINGER et al.

112. J. Gmeiner, M. Simon, and 0. Luderitz, Eur. J. Biochem., 21 (1971) 355-356. 113. P. F. Miihlradt, V. Wray, and V. Lehmann, Eur. J. Biochem., 81 (1977) 193-203. 114. R. E. Hurlbert, J. Weckesser, H. Mayer, and I. Fromme, Eur. J. Biochem., 68 (1976)

1 15. Y. Kamio, K. C. Kim, and H. Takahashi, J. Biochem. (Tokyo), 70 (197 1) 187 - 19 1. 116. E. Th. Rietschel, L. Brade, 0. Holst, V. A. Kulshin, B. Lindner, A. P. Moran, U. F. Schade,

U. Zahringer, and H. Brade, in A. Nowotny, J. J. Spitzer, and E. J. Ziegler (Eds.), Endo- toxin Research Series, Ed., Vol. I , Cellular and Molecular Aspects of Endotoxin Reac- tions: Molecular Structure of Bacterial Endotoxin in Relation to Bioactivity, p. 15. Else- vier, Amsterdam, 1990.

117. A. A. Lindberg, A. Weintraub, U. Zilhringer, andE. Th. Rietschel, Rev. Infect. Dis., 12 (2)

1 18. E. Th. Rietschel, L. Brade, U. F. Schade, U. Seydel, U. ZAhringer, S. Kusumoto, and H. Brade, in E. Schrinner, M. Richmond, G. Seibert, and U. Schwartz (Eds.), Surface Struc- tures of Microorganisms and Their Interaction with the Mammalian Host: Bacterial Endotoxins: Properties and Structure of Biologically Active Domains, p. 1. Verlag Chemie, Weinheim, 1988.

119. A. Sonesson, E. Jantzen, K. Bryn, L. Larsson, and J. Eng, Arch. Microbiol., 153 (1989)

120. S. E. Zamze, M. A. J. Ferguson, E. R. Moxon, R. A. Dwek, and T. W. Rademacher,

121. N. Dalla Venezia, S. Minka, M. Bruneteau, H. Mayer, and G. Michel, Eur. J. Biochem.,

122. V. Lehmann and E. Rupprecht, Eur. J. Biochem., 81 (1977) 443-452. 123. V. Lehmann, J. W. Redmond, A. Egan, and I. Minner, Eur. J. Biochem., 86 (1978)

124. S. K. B. Lakshmi, U. R. Bhat, K. Wartenberg, S. Schlecht, and H. Mayer, FEMS Micro-

125. M. Vaara, T. Vaara, M. Jensen, I. M. Helander, M. Nurminen, E. Th. Rietschel, and P. H.

126. W. Kaca, J. Radziejewska-Lebrecht, and R. Bhat, Microbios, 6 I (1990) 23-32. 127. S. G. Wilkinson, in C. Ratledge and S. G. Wilkinson (Eds.,) Microbial Lipids, Vol. 1,

128. E. Th. Rietschel, H. Gottert, 0. Liideritz, and 0. Westphal, Eur. J. Biochem., 28 (1972)

129. E. Th. Rietschel, Eur. J. Biochem., 64 (1976) 423-428. 130. D. Horton, D. A. Riley, S. Samreth, and M. G. Schweitzer, Am. Chem. SOC. Symp. Ser.,

131. S. G. Wilkinson, Rev. Infect. Dis., 5 (1983) 941-949. 132. M. Nurminen, E. Th. Rietschel, and H. Brade, Infect. Immun., 48 (1985) 573-575. 133. L. Brade, S. Schramek, U. Schade, and H. Brade, Infect. Immun., 54 (1986) 568-574. 134. M. J. Hewett, K. W. Knox, and D. G. Bishop, Eur. J. Biochem., 19 (1971) 169-175. 135. I. Mattsby-Baltzer, Z. Mielniczuk, L. Larsson, K. Lindgren, and St. Goodwin, Infect.

136. A. P. Moran, I. M. Helander, and T. U. Kosunen, J. Bacteriol., 174 (1992) 1370- 1377. 137. H.-W. Wollenweber, S. Schramek, H. Moll, and E. Th. Rietschel, Arch. Microbiol., 142

138. W. R. Mayberry, J. Bacteriol., 147 (1981) 373-381. 139. N. Haeffner, R. Chaby, and L. Szab6, Eur. J. Biochem., 77 (1977) 535-544. 140. S. K. Maitra, M. C. Schotz, T. T. Yoshikawa, and L. B. Guze, Proc. Natl. Acad. Sci. USA,

365-371.

(1 990) S133 - S 14 1.

72-78.

Biochem. J., 245 (1987) 583-587.

151 (1985) 399-404.

487-496.

biol. Lett., 60 (1989) 317-322.

MZikelB, FEBSLett., 129 (1981) 145-149.

Gram-Negative Bacteria, p. 299. Academic Press, London/San Diego, 1988.

166-173.

231 (1983) 21-47.

Immun., 60 (1992) 4383-4387.

(1985) 6- 1 1.

75 (1978) 3993-3997.

Page 294: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

MOLECULAR STRUCTURE OF LIPID A 27 1

141. A. Sonesson, L. Larsson, R. Anderson, N. Adner, and K. -G. Tranberg, J. Clin. Micro-

142. H.-W. Wollenweber, S. Schlecht, 0. Liideritz, andE. Th. Rietschel, Eur. J. Biochem., 130

143. L. van Alphen, B. Lugtenberg, E. T. Rietschel, and C. Mombers, Eur. J. Biochem., 101

144. A. M. B. Kropinski, V. Lewis, and D. Berry, J. Bucteriol., 169 (1987) 1960- 1966. 145. K. Wartenberg, W. Knapp, N. M. Ahamed, C. Widemann, and H. Mayer, Zbl. Bnkt.

146. Y. Kawai and I. Yano, Eur. J. Biochem., 136 (1983) 531 -538. 147. 0. W. Thiele, J. Oulevey, and D. H. Hunnemann, Eur. J. Biochem., I39 (1984) 13 1 - 135. 148. J. R. Edwards and J. A. Hayashi, Arch. Biochem. Biophys., I I 1 (1965) 415-421. 149. D. B. Boylan and P. J. Scheuer, Science, 155 (1967) 52-56. 150. A. P. Tulloch and J. F. T. Spencer, Can. J. Chem., 42 (1964) 830-837. I5 1. K. Bryn and E. Th. Rietschel, Eur. J. Biochem., 86 (1978) 3 1 I - 3 15. 152. U. R. Bhat, A. Man, C. Galanos, and R. S. Conrad, J. Bucteriol., 172 (1990) 663 I -

153. S. E. Zamze, A. R. W. Smith, and R. C. Hignett, J. Gen. Microbiol., 13 I (1 985) 194 1 -

154. H. Brade and C. Galanos, Eur. J. Biochem., 122 (1982) 233-237. 155. K. Kawahara, K. Uchida, and K. Aida, Biochim. Biophys. Actu, 712 (1982) 571 -575. 156. K. Kawahara, U. Seydel, M. Matsuura, H. Danbara, E. Th. Rietschel, and U. ahringer,

157. K. Takayama, R. J. Rothenberg, and A. G. Barbour, Infict. Immun., 55 (1987) 231 1 - 158. S . Arata, T. Hirayama, N. Kasai, T. Itoh, and A. Ohsawa, FEMS Microbiol. Lett., 60

159. H. Moll, A. Sonesson, E. Jantzen, R. Marre, and U. Z&ringer, FEMSMicrobiol. Lett.. 97

160. U. R. Bhat, R. W. Carlson, M. Busch, and H. Mayer, In?. J. Syst. BuderioZ., 41 (1991)

161. A. Sonesson, H. Moll, E. Jantzen, and U. W n g e r , FEMS Microbiol. Lett., 106 (1993)

162. N. Qureshi, K. Takayama, K. C. Meyer, T. N. Kirkland, C. A. Bush, L. Chen, R. Wang,

162a. C. R. H. Raetz, J. Bucteriol., 175 (1993) 5745-5753. 163. B. J. Wilkinson, M. S. Hindahl, L. Galbraith, and S. G. Wilkinson, FEMS Microbiol.

164. W. Strittmatter, J. Weckesser, P. V. Salimath, and C. Galanos, J. Bucteriol., 155 (1983)

165. P. V. Salimath, R. N. Tharanathan, J. Weckesser, and H. Mayer, Eur. J. Biochem., 144

166. N. Qureshi, K. Takayama, and R. Kurtz, Infect. Immun., 59 (1991) 441-444. 167. H. Loppnow, H. Brade, I. Diirrbaum,C. A. Dinarello, S. Kusumoto, E. Th. Rietsche1,and

H.-D. Flad, J. Zmmunol., 142 (1989) 3229-3238. 168. W. Feist, A. J. Ulmer, J. Musehold, H. Brade, S. Kusumoto, and H.-D. Had, Immuno-

biol., 179 (1989) 293-307. 169. M. H. Wang, H.-D. Had, W. Feist, J. Musehold, S. Kusumoto, H. Brade, J. Gerdes, E. Th.

Rietschel, and A. J. Ulmer, Lymphokine Cytokine Rex, 1 1 (1 99 1) 23 - 3 1. 170. C. Galanos, V. Lehmann, 0. Luderitz, E. Th. Rietschel, 0. Westphal, H. Brade, L. Brade,

M. A. Freudenberg, T. Hansen-Hagge, T. Liideritz, G. McKenzie, U. F. Schade, W.

biol., 28 (1990) 1163-1168.

(1983) 167-171.

(1979) 571-579.

Hyg.., 253 (1983) 523-530.

6636.

1950.

FEBS Lett., 292 (1991) 107-110.

2313.

(1989) 219-222.

(1992) 1-6.

213-217.

3 15 -320.

and R. J. Cotter, J. Biol. Chem., 266 (1991) 6532-6538.

Lett., 37 (1986) 63-67.

153-158.

(1984) 227-232.

Page 295: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

272 ULRICH ZAHNNGER et al.

Strittmatter, K. -I. Tanamoto, U. ahringer, M. Imoto, M. Yamamoto, T. Shimamoto, S. Kusumoto, and T. Shiba, Eur. J. Biochem., 140 (1984) 221 -227.

171. C. Galanos, T. Hansen-Hagge, V. Lehmann, and 0. Liideritz, Infict. Immun., 48 (1985)

172. E. Th. Rietschel, L. Brade, U. F. Schade, C. Galanos, M. A. Freudenberg, 0. Liideritz, S.

173. T. Hansen-Hagge, V. Lehmann, U. Seydel, B. Lindner, and U. Zduinger, Arch. Micro-

174. N. Qureshi, P. Mascagni, E. E. Ribi, and K. Takayama, J. Biol. Chem., 260 (1985)

175. K. R. Myers, A. T. Truchot, J. Ward, Y. Hudson, and J. T. Ulrich, in A. Nowotny, J. J. Spitzer, and E. Ziegler (Eds.), Endotoxin Research Series, Vol. 1, Cellular and Molecular Aspects of Endotoxin Reactions: A Critical Determinant ofLipid A Endotoxic Activity, p. 145. Excerpta Medica, Amsterdam/New York/ Oxford, 1990.

335 - 358.

Kusumoto, and T. Shiba, Eur. J. Biochem., 169 ( 1987) 27 - 3 1.

biol., 141 (1985) 353-358.

5271-5278.

176. H.-W. Wollenweber and E. Th. Rietschel, J. Microb. Meth., 1 1 (1990) 195-21 1 . 177. W. Drozanski, A. E. A. Moustafa, 0. Liideritz, and 0. Westphal, Carbohydr. Res., 178

178. W. Drozanski, C. Galanos, S. Schlecht, and 0. Liideritz, Eur. J. Biochem., 155 (1986)

179. D. Malchow, 0. Liideritz, B. Kickhofen, 0. Westphal, and G. Gerisch, Eur. J. Biochem., 7

180. R. S. Munford, Jr., and C. L. Hall, Infect. Immun., 48 (1985) 464-473. 181. R. S. Munford,Jr., L. Eidels, and E. J. Hansen in A. Nowotny, J. J. Spitzer, and E. J.

Ziegler (Eds.), Endotoxin Research Series, Vol. I, Cellular and Molecular Aspects of Endotoxin Reactions: Purifrcation ofHuman Acyloxyacyl Hydrolase UsingAfinity Chro- matography, p. 305. Medica, Amsterdam/New York/Oxford, 1990.

182. K. Takayama, N. Qureshi, B. Beutler and T. N. Kirkland, Infect. Immun., 57 (1989)

183. M.H. Wang, W.Feist,H. Herzbeck,H.Brade,S.Kusumoto,E.Th.Rietschel,H.-D.Flad, and A. J. Ulmer, FEMSMicrob. Immunol., 64 (1990) 179- 186.

184. D. T. Golenbock, R. Y. Hampton, N. Qureshi, K. Takayama, and C. R. H. Raetz, J. Biol. Chem., 266 (1991) 19490- 19498.

185. A. J. Ulmer, H. Heine, W. Feist, S. Kusumoto, T. Kusama, H. Brade, U. Schade, E. T. Rietschel, and H. -D. Had, Infect. Immun., 60 (1992) 3309-3314.

185a. H. Loppnow, E. Th. Rietschel, H. Brade, U. Schdnbeck, P. Libby, M.-H. Wang, H. Heine, W. Feist, 1. Diirrbaum-Landmann, M. Emst, E. Brandt, H. Grage-Griebenow, A. J. Ulmer, S. Campos Portuguez, U. Schade, R. Kirikae, S. Kusumoto, J. Krauss, H. Mayer, and H.-D. Had, Vol. 2, Bacterial endotoxin: Recognition and eflector mechanisms. (J. Levin, C. R. Alving, R. S. Munford, and P. L. Stiitz, eds.) Excerpta Medica, Elsevier, Amsterdam, 1993, p. 337.

186. T.H.Pohlmann,R. S.Munford,andJ.M. Harlan,J. Exp.Med., 165(1987) 1393- 1402. 187. H.-W. Wollenweber, K. W. Broady, 0. Liideritz, and E. Th. Rietschel, Eur. .I. Biochem.,

188. U. Kraska, J. R. Pougny, and P. Sinay, Curbohyd. Rex, 50 (1976) 18 1 - 190. 189. R. C. Verret, M. R. Rosner,andH.G. Khorana,J. Biol. Chem., 257(1982) 10222- 10227. 190. R.C. Verret,M.R. Rosner,andH.G. Khorana,J. Biol. Chem.,257(1982) 10338- 10234. 191. J. M. Williamson, M. S. Anderson, and C. R. H. Raetz, J. Bacteriol., 173 (1991) 3591-

192. E. Stackebrandt, R. G. E. Murray, and H. G. Triiper, Znt. J. Syst. Bacteriol., 38 (1988)

(1988) 225-232.

433-437.

(1969) 239-246.

1336-1338.

124 (1982) 191-198.

3596.

321-325.

Page 296: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

MOLECULAR STRUCTURE OF LIPID A 273

193. S. Basu, J. Radziejewska-Lebrecht, and H. Mayer, Arch. Microbiol.. 144 (1986) 213-2 18. 193a. W. Erles, H. Feist, and K. D. Feossmann, Arch. Exp. Veterinurmed., 31 (1977) 203-

193b. H. Masoud, M. B. Perry, and J. C. Richards, Eur. J. Biochem. 220 (1994) 209-216. 194. R. N. Tharanathan, P. V. Salimath, J. Weckesser, and H. Mayer, Arch. Microbiol., 141

195. I. M. Helander, L. Hirvas, J. Tuominen, and M. Vaara, Eur. J. Biochem., 204 (1992)

195a. I. M. Helander, B. Lindner, U. Seydel, and M. Vaara, Eur. J. Biochem., 212 (1993)

196. C. Galanos, 0. Luderitz, M. A. Freudenberg, L. Brade, U. F. Schade, E. Th. Rietschel, S.

197. V. Lehmann, E. Rupprecht, and M. J. Osborn, Eur. J. Biochem., 76 (1977) 41-49. 198. C. R. H. Raetz, S. Purcell, M. V. Meyer, N. Qureshi, and K. Takayama, J. Biol. Chem.,

199. T. Hansen-Hagge, V. Lehmann and 0. Ludentz, Eur. J. Biochem., 148 (1985) 21 -27. 200. M. Imoto, H. Yoshimura, N. Sakaguchi, S. Kusumoto, and T. Shiba, Tetrahedron Lett.,

201. G. Rosenfelder, 0. Luderitz, and 0. Westphal, Eur. J. Biochem., 44 (1974) 41 1-420. 202. R. Russa and Z. Lorkiewicz, J. Bucteriol.. 119 (1974) 771-775. 203. C. R. Woese, E. Stackebrandt, W. G. Weisburg, B. J. Paster, M. T. Madigan, V. J. Fowler,

C. M. Hahn, P. Blanz, R. Gupta, K. H. Nealson, and G. E. Fox, Syst. Appl. Microbiol., 5

209.

(1985) 279-283.

1101-1 106.

363-369.

Kusumoto, and T. Shiba, Eur. J. Biochem., 160 (1986) 55-59.

260 (1985) 16080- 16088.

26 (1985) 1545-1548.

(1984) 315-326. 204. P. Fiigedi and J. Kovacs, Fifth Euro. Symp. Curbohydr., (1989) A-69. [Abstr.] 205. K. Ohno, H. Nishiyama, and H. Nagase, Tetrahedron Lett., 45 (1979) 4405-4406. 206. Y. Inoue and K. Nagasawa, Curbohyd. Res., 168 (1987) 181-190. 207. K. Takayama, N. Qureshi, P. Mascagni, M. A. Nashed, L. Anderson, and C. R. H. Raetz,

208. A. K. Bhattacharjee, H. J. Jennings, and C. P. Kenny, Biochemistry, 17 (1978) 645 -65 1. 209. A. Neszmelyi, K. Jann, P. Messner, and F. M. Unger, J. Chem. SOC., Chem. Commun.

2 10. F. M. Unger, D. Stix, and G. Schulz, Curbohydr. Rex, 80 (1 980) 19 1 - 195. 21 1. K. Kawahara, H. Moll, P. Kosma, S. Dejsirilert, T. Ezaki, and U. Ziihringer, in Second

Conference of thelnternutionul Endotoxin Society, Vienna, August 17 - 20,1990. [Abstr.] 2 t2. U. Zannger, K. Kawahara, H. Moll, and P. Kosma, unpublished results. 213. 0. Holst, L. Brade, P. Kosma, and H. Brade, J. Bucteriol., 173 (1991) 1862- 1866. 214. K. Ikeda, T. Takahachi, H. Kondo, and K. Achiwa, Chem. Phurm. Bull., 35 (1987)

215. M. Imoto, H. Yoshimura, S. Kusumoto, and T. Shiba, Proc. Jupun Acud., 60 (1984)

2 16. S. Kusumoto, M. Inage, H. Chaki, M. Imoto, T. Shimamoto, and T. Shiba, Am. Chem.

217. M. Shiozaki, Y. Kobayashi, M. Arai, N. Ishida, T. Hiraoka, M. Nishijima, S. Kuge, T.

218. H. Vyplel, D. Scholz, H. Loibner, M. Kern, K. Bednarik, and H. Schaller, Tetrahedron

219. H. Labischinski, D. Naumann, C. Schultz, S. Kusumoto, T. Shiba, E. Th. Rietschel, and

220. H. Labischinski, G. Barnickel, H. Bradaczek, D. Naumann, E. Th. Rietschel and P.

J. Biol. Chem., 258 (1983) 7379-7385.

(1982) 1017-1019.

13 11 - 13 14.

285-288.

SOC. Symp. Ser., 231 (1983) 237-254.

Otsuka, and Y. Akamatsu, Curbohydr. Res., 222 (1991) 69-82.

Lett., 33 (1992) 1261-1264.

P. Giesbrecht, Eur. J. Biochem., 179 (1989) 659-665.

Giesbrecht, J. Bucteriol., 162 (1985) 9-20.

Page 297: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

274 ULRICH ZAHRINGER et al.

221. M. Kastowsky, A. Sabisch, T. Gutberlet, and H. Bradaczek, Eur. J. Biochem., 197 (1991)

222. H. Labischinski, E. Vorgel, W. Uebach, R. P. May, and H. Bradaczek, Eur. J. Biochem.,

223. V. Luzzati, in D. Chapman (Ed.), Biological Membranes: Physical Fact and Function, X-Ray Diffraction Studies of Lipid- Water Systems, Academic Press, London, 1968.

224. U. Seydel, K. Brandenburg, M. H. J. Koch, and E. Th. Rietschel, Eur. J. Biochem., 186 (1989) 325-332.

225. U. Seydel and K. Brandenburg, in A. Nowotny, J. I. Spitzer, and E. J. Ziegler (Eds.), Endotoxin Research Series, Vol. 1 , Cellular and Molecular Aspects of Endotoxin Reac- tions: Conformations of Endotoxin and Their Relationship to Biological Activity, p. 6 1. Excerpta Medica, Elsevier, Amsterdam, 1990.

707- 7 16.

190 (1990) 359-363.

226. K. Brandenburg, M. H. J. Koch, and U. Seydel, J. Struct. Biol., 105 (1991) 1 1-21. 227. U. Seydel and K. Brandenburg, in D. C. Momson and J. L. Ryan (Eds.), Bacterial

Endotoxic Lipopolysaccharides: Supramolecular Structure of Lipopolysaccharides and Lipid A, p. 225. CRC Press, Boca Raton, FL, 1992.

227a. K. Brandenburg, Biophys. J., 64 (1993) 1215-1231. 228. D. Naumann, C. Schultz, A. Sabisch, M. Kastowsky, and H. Labischinski, J. Mol. Struct.,

229. G. Lipka, R. A. Demel, and H. Hauser, Chem. Phys. Lipids, 48 (1988) 267-280. 229a. K. Brandenburg,H. Mayer,M. H. J. Koch, J. Weckesser,E. Th. Rietschel, andU. Seydel,

229b. A. Shnyra, K. Hultenby, and A. A. Lindberg, Infect. Immun. 61 (1993) 5351-5360. 229c. K. Takayama, D. H. Mitchell, Z. Z. Din, P. Mukerjee, C. Li, and D. L. Coleman,

J. Biol. Chem. 269 (1994) 2241-2244. 229d. T. Kirikae, F. U. Schade, U. Zihringer, F. Kirikae, H. Brade, S. Kusu-

moto, T. Kusama, and E. Th. Rietschel, FEMS Immunol. Med. Microbiol. 8 (1994)

230. T. Liideritz, K. Brandenburg, U. Seydel, A. Roth, C. Galanos, andE. Th. Rietschel, Eur. J.

231. K. Brandenburg and U. Seydel, Eur. J. Biochem., 191 (1990) 229-236. 232. 0. Westphal, 0. Liideritz, C. Galanos, H. Mayer, andE. Th. Rietschel, inL. Chedid, J. W.

Hadden, F. Spreafico, P. Dukor, and D. WiIloughby (Eds.), Advances in Immunopharma- cology: The Story of Bacterial Endotoxin, p. 13. Pergamon Press, Oxford, 1985.

233. K.4. Tanamoto, U. F. Schade, E. Th. Rietschel, S. Kusumoto, and T. Shiba, Infect. Immun., 58 (1990) 217-221.

234. C. Galanos, 0. Liideritz, E. Th. Rietschel, 0. Westphal, H. Brade, L. Brade, M. A. Freudenberg, U. F. Schade, M. Imoto, S. Yoshimura, S. Kusumoto, and T. Shiba, Eur. J. Biochem., 148 (1985) 1-5.

235. J. Y. Homma, M. Matsuura, and Y. Kumazawa, Adv. Exp. Med. Biol., 256 (1990)

236. S. Kotani, H. Takada, M. Tsujimoto, T. Ogawa, I. Takahashi, T. Ikeda, K. Otsuka, H. Shimauchi, N. Kasai, J.4. Mashimo, S. Nagao, A. Tanaka, S. Tanaka, K. Harada, K. Nagaki, H. Kitamura, T. Shiba, S. Kusumoto, M. Imoto, and H. Yoshimura, Infect. Immun., 49 (1985) 225-237.

237. S. Kanegasaki, K.4. Tanamoto, T. Yasuda, J. Y. Homma, M. Matsuura, M. Nakatsuka, Y. Kumazawa, A. Yamamoto, T. Shiba, S. Kusumoto, M. Imoto, H. Yoshimura, and T. Shimamoto, J. Biochem. (Tokyo), 99 (1986) 1203- 1210.

214 (1989) 213-246.

Eur. J. Biochem., 218 (1993) 555-563.

13-26.

Biochem., 179 (1989) 1 1 - 16.

101- 119.

238. S. Kotani and H. Takada, Adv. Exp. Med. Biol., 256 (1990) 13-43.

Page 298: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

MOLECULAR STRUCTURE OF LIPID A 275

239. H. Loppnow, L. Brade, H. Brade, E. Th. Rietschel, S. Kusumoto, r. Shiba, and H. -D.

240. J. M. Cavaillon and N. Haeffner-Cavaillon, Cytokine, 2 ( 1990) 3 13 - 329. 24 1 . C. Galanos, M. A. Freudenberg, F. Jay, D. Nerkar, K. Veleva, H. Brade, and W. Strittmat-

242. L. Brade, 0. Holst, and H. Brade, Infect. Immun., 61 (1993) 4514-4517. 243. L. Brade, K. Brandenburg, H.-M. Kuhn, S. Kusumoto, I. Macher, E. Th. Rietschel, and

244. H.-M. Kuhn, L. Brade, B. J. Appelmelk, S. Kusumoto, E. Th. Rietschel, and H. Brade,

244a. H.-M. Kuhn, L. Brade, B. J. Appelmelk, S. Kusumoto, E. Th. Rietschel, and H. Brade,

244b. H.-M. Kuhn, Infect. Immun. 61 (1993) 680-688. 245. W. C. Bogard, Jr., D. L. Dunn, K. Abernethy, C. Kilgarriff, and P. C. Kung, Infect.

246. R. Chaby, D. Charon, T. Pedron, and R. Girard, Biochim. Biophys. Res. Commun.. 143

247. N. Kasai, S. Arata, J.4. Mashimo, M. Ohmori, T. Mizutani, and K. Egawa, in A. Nowotny (Ed.), Endotoxin Research Series, Vol. 1 , Cellular and Molecular Aspects of Endotoxin Reactions, p. 121. Elsevier, Amsterdam, 1990.

Flad, Eur. J. Imrnunol., 16 (1986) 1263- 1267.

ter, Rev. Infect. Dis., 6 (1984) 546-552.

H. Brade, Infect. Immun.. 55 (1987) 2636-2644.

Infect. Immun., 60 (1992) 2201 -2210.

Immunbiol. 189 (1993) 457-471.

Immun., 55 (1987) 899-908.

(1987) 723-731.

248. T. N. Kirkland, N. Qureshi, and K. Takayama, Infect. Imrnun., 59 (1991) 131 - 136. 249. M. Pollack, K. Oishi, J. Chia, M. E. Evans, G. Guelde, and N. C. Koles, Adv. Exp. Med.

Biol., 256 (1990) 331-340. 250. E. J. Ziegler, C. J. Fisher, C. L. Sprung, R. C. Straube, J. C. Sadoff, G. E. Foulke, C. H.

Wortel, M. P. Fink, R. P. Dellinger, N. N. H. Teng, I. E. Allen, H. J. Berger, G. L. Knatterud, A. F. LoBuglio, C. R. Smith, and the HA- 1A sepsis study group, New Engl. J. Med., 324 (1991) 429-436.

251. K. Gorelick, P. J. Scannon, J. Hannigan, N. Wedel, and S. K. Ackerman, in C. A. K. Borrebaeck and J. W. Lamck (Eds.), Therapeutic Monoclonal Antibodies: Randomized Placebo-Controlled Study ofE.5 Monoclonal Antiendotoxin Antibody, p. 16. M. Stockton, New York, 1990.

251a. Y. Fujihara, M.-G. Lei, andD. C. Momson, Infect. Zmmun., 61 (1993)910-918. 252. M. A. Freudenberg, A. Fomsgaard, I. Mitov, and C. Galanos, Infection, 17 (1989) 322-

253. J. -D. Baumgartner, Infect. Dis. Clin. N. Am., 5 (1991) 915-927. 253a. H. S. Warren, R. L. Damus, and R. S. Munford, New Engl. J. Med., 326 (1993) 1 1 53-

1157. 254, C. R. H. Raetz, in Methods in Enzymology, Vol. 209, pp. 455-466. Academic Press, San

Diego, 1992. 255. C. R. H. Raetz, in C. Neidhardt, J. 1. Ingraham, K. Brooks Low, B. Magasanik, M.

Schaechter, and H. E. Umbarger (Eds.), Escherichia coli and Salmonella typhimurium: Cellular and Molecular Biology: Structure and Biosynthesis ofLipid A, p. 498. Amencan Society of Microbiologists, Washington DC, 1987.

256. J. W. Shands, Jr., J. Infect. Dis., 128 (1973) 189- 193. 257. J. W. Shands, Jr., in S. Kadis, G. Weinbaum, and J. S. Ajl (Eds.), Microbial Toxins, Vol.

IV, Bacterial Endotoxins: The Physical Structure of Bacterial Lipopolysaccharides, p. 127. Academic Press, New York, 197 1 .

258. H. Nikaido and R. E. W. Hancock, in The Bacteria, Vol. X, Outer Membrane Permeabil- ity ofpseudomonas aeruginosa, p. 146. Academic Press, New York, 1986.

329.

Page 299: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

276 ULRICH ZAHRINGER et al.

259. G. Seltmann, in Die bakterielle Zellwand. G. Fischer Verlag, Stuttgart, Germany, 1982. 260. R. Vuorio and M. Vaara, Antimicrob. Agents Chemother., 36 (1992) 826-829. 261. C. E. Ooi, J. Weiss, M. E. Doerfler, and P. Elsbach, J. Exp. Med., 174 (1991) 649-

262. D. C. Momson, Microb. Pathogen., 7 (1989) 389-398. 263. S. D. Wright, R. A. Ramos, P. S. Tobias, R. J. Ulevitch, and J. C. Mathison, Science, 249

264. T. Kirikae, F. Kirikae,F. U. Schade, M. Yoshida, S. Kondo, K. Hisatsune, S. LNishikawa, and E. Th. Rietschel, FEMSMicrobiol. Immunol., 76 (1991) 327-336.

265. H. Loppnow, P. Libby, M. A. Freudenberg, J. H. Krauss, J. Weckesser, and H. Mayer, Infect. Immun., 58 (1990) 3743-3750.

266. W. Feist, A. J. Ulmer, M.-H. Wang, J. Musehold, C. Schliiter, J. Gerdes, H. Herzbeck, H. Brade, S. Kusumoto, T. Diamantstein, E. T. Rietschel, and H.-D. Had, FEMSMicrobiol. Immunol., 89 ( 1 992) 73 - 90.

266a. B. E. Henricson, P. Y. Perera, N. Qureshi, K. Takayama, and S. Vogel, Infect. Immun., 60 (1992) 4285-4290.

267. J. B. Parent, J. Biol. Chem., 265 (1990) 3455-3461. 268. H. Kumada, K. Watanabe, T. Umemoto, Y. Haishima, S. Kondo, and K. Hisatsune,

269. H. Brade, J. Bacteriol., 161 (1985) 795-798. 270. H. Masoud, H. Mayer, T. Kontrohr, 0. Holst, and J. Weckesser, System. Appl. Microbiol.,

27 1. R. I. Hollingsworth, R. W. Carlson, F. Garcia, and D. A. Gage, J. Biol. Chem., 265 (1990)

655.

(1990) 1431-1433.

FEMS Microbiol. Lett., 51 (1988) 77-80.

14 (1991) 222-227.

12752.

Page 300: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

ADVANCES IN CARBOHYLlRATE CHEMISTRY AND BIOCHEMISTRY, VOL. 50

DEVELOPMENTS IN THE SYNTHESIS OF GLYCOPEPTIDES CONTAINING GLYCOSYL L-ASPARAGINE, L-SERINE, AND

L-THREONINE

BY HARI G. GARG,* KARSTEN VON DEM BRUCH,? AND HORST KUNZ-~

*Pulmonary and Critical Care Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 021 14; and flnstitut f i r Organische Chemie, Johannes Gutenberg-Universitat. 0-55128 Mainz, Germany

I. INTRODUCTION In 1932, the first synthesis of a D-glucopyranosylamine derivative (1)

substituted on the nitrogen atom by a glycine residue was achieved by Berg-

H,N-CH-COOH

I I

CHZ

HO*oH HO H O H ~ E ~ o

RHN AcHN

1 R=COCHzNH2 3

2 R = COCHR'NH2

mann and Zervas (1). Twenty-one years later, Link and coworkers (2), utilizing this method, prepared a series of N-acyl derivatives of 2. Neuberger and associates (3) proposed a 2-acetamido-N-(~-aspart-4-oyl)-2-deoxy-j?-~- glucopyranosyl linkage between carbohydrate and protein in egg albumin. In order to obtain a direct proof for the proposed carbohydrate-protein linkage, Marks and Neuberger (4) in 196 1 reported the synthesis of deriva- tive 3. Since that time, a number ofglycoproteins have been found to contain this or other types of carbohydrate-protein linkages. A selection ( 5 ) is given in Table I.

The afore-mentioned results have stimulated an interest in the synthesis of glycopeptides for the purpose of supplying structurally well-defined deriva- tives as models for biochemical and immunochemical studies. Synthetic

Copyright 0 1994 by Academic Press, Inc. AU rights of reproduction in any form reserved. 277

Page 301: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

278 HARI G. GARG el al.

TABLE In The Amino Acid and Carbohydrate Residues in Carbohydrate-Protein

Linkages of Mammalian Origin. ~~

Anomeric Amino acid Corresponding carbohydrate residue configuration

L- Asparagine 2-Acetamido-2-deoxy-~-glycose P L-Serine/L-threonine 2-Acetamido-2deoxy-~-galactose (Y

D-XylOSe B

5-Hydroxy-~-lysine D - G d a ct o se P D-Mannose (Y

(1 In plants a linkage between Chydroxy-bproline and L-arabinose and in halobacteria a linkage between L-asparagine and DGlucose/2-acetamid~2deoxy-D-galactose has been reported.

strategies for glycopeptides are more complex and difficult compared with the synthesis either of oligopeptides or of oligosaccharides. Glycopeptide synthesis requires the protection of amino and carboxyl termini of amino acids and peptides and also the protection of hydroxyl groups of carbohy- drate residues. In addition, for stereoselective synthesis of specific anomers, participating or nonparticipating groups, respectively, at C-2 of the carbohy- drate moiety are needed. The respective glycosyl donors have to be coupled with amino acid derivatives to give the desired isomer. Furthermore, condi- tions are required throughout the synthesis that permit the deprotection of only one functional group without attacking the other protection and also without affecting the anomeric configuration or the breaking of an oligosac- charide bond. For this purpose, several new amino-protecting groups of the urethane type, new carboxyl-protecting groups, and new methodologies have been developed since the first article appeared in this series (6). This chapter focuses mainly on the advances made in glycopeptide synthesis that have been published between 1984 and 1993. Several reviews describing different aspects of synthetic glycopeptides have been published during this

11. N-GLYCOPEPTIDES 1. Formation of the 2-Acetarnido-2-deoxy-~-~-glucosyl- L-

Asparagine Linkage

a. Anomeric Azides as Precursors. - The reduction of a glycosyl azide derivative (4) to glycosylamine (5), followed by a coupling with the 4-car- boxyl group of L-aspartic acid (6) in the presence of dicyclohexylcarbodii- mide (DCC), was first employed by Marks and Neuberger ( 5 ) for synthesis of 2-acetamido-N-(~-aspart-4-oyl)-2-deoxy-~-~-glucopyranosyl (3). Since

period (7 - 11).

Page 302: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

GLYCOPEPTIDE SYNTHESIS 279

2-HN-CH-COOBzl 11 DCc

Ad* HZ ~ I 2) depmtenon

NH, + 'HZ - 3

I AcO N3

AcHN AcHN COOH

4 5 6

&I I PhCH2 2 .PhCHfxn

then, this method of synthesis has been extended to the preparation of a number of derivatives (6) and different condensation methods have been compared (1 2).

In order to construct the L-asparagine derivative of per-0-acetylated 0-( 2-acetamido-2-deoxy-/3-~-glucopyranosyl( 1 + 4)-2-acetamido-2-deoxy- D-glucose (7), Spinola and Jeanloz (1 3) used (the sensitive) silver azide for conversion of the a-chloro anomer of the chitobiose derivative 8 into the /3-azido derivative 9. Kunz and associates ( 14,15) have accomplished the conversion of the a-chloro anomer of 8 into 9 using sodium azide in the presence of tri-n-octyl-methylammonium chloride as a phase-transfer cata- lyst in chloroform water.

ACHN AcHh

k 7 R - H, R' = Boc-L-Asn-OAll

8 R=CI. R ' = H

9 R - H , R ' = N s

10 R = H, R' = NHz

Boc-HN- CH-COO-All

10 + CHZ

I COOH

1 1

Boc = (CH~JICOCO All -CHZ;CHGY

Hydrogenolysis of the azide 9 furnished the amino derivative 10, which was subsequently coupled with N-tat-butyloxycarbonylaspartate ( 16) (1 1) in the presence of ethyl 2-ethoxy- 1,2-dihydroquinoline- 1 -carboxylate W D Q ) ( 17).

Warren and associates ( I 8) have prepared a glycosyl azide derivative (15) of a heptasaccharide. This glycosyl azide was obtained from 0-a-D-manno- pyranosyl-( 1 -6)-O-[a-~-mannopyranosyl-( I -3)-O-a-~-mannopyrano syl - ( 1 + 6) - 0- a - D - mannopyranosyl - ( 1 - 3)] - 0-/3- D - mannopyranosyl- ( 1 -4) - 0 - (2 - acetamido - 2 - deoxy - /3 - D - glucopyranosyl) - ( 1 4 4 ) - 2 - acetamido-2-deoxy-~-glucopyranose (12) by treatment of its peracetylated derivative 13 with trimethylsilyl trifuoromethanesulfonate, followed by re- action of the intermediary oxazoline 14 with trimethylsilyl azide. Reduction of the glycosyl azide 15 in the presence of Lindlar catalyst gave the glycosyla- mine derivative 16. The condensation of 16 with I-tert-butyl N(9-fluoren-

Page 303: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

280 HARI G. GARG et al.

Frnoc~HN- CH- COOtBu

I RO AcO -.&lo+ NH, + CH2 I

AcHN COOH

16 R - X ' 1 7

IBu =(CHIIIC

R'-HN- CH-COOF

0.O-dtelhyl I phosphowlqanide

c I

RO AcO * ; = O NH

AcHN

18 R = X', R = Frnlx, R" = tBu 19 R = X ' R ' = R " = H

ylmethoxycarbony1)-L-aspartate (17) in the presence of 0,O-diethylphos- phoryl cyanide gave the corresponding glycosylasparagine derivative 18, which was partially deprotected to furnish derivative 19.

b. In Situ Anomerization Procedure for Attachment of CX-L-FUCOS~~ Units. - The synthesis of the 0-protected trisaccharide azide 20 containing an a-L-fucosyl residue was achieved ( 19c) by using a chitobiosyl azide deriv- ative containing a free hydroxyl group at C-6. The coupling was performed under in situ anomerization conditions (20) with benzyl-protected a-fuco- pyranosyl bromide ( 19b). Selective hydrogenation employing neutral (!) washed Raney nickel gave the protected trisaccharide glycosylamine deriva- tive 21. Derivative 21, when coupled with 1 -tert-butyl N-allyloxycarbonyl- L-aspartate (22) in the presence of EEDQ, gave (1 9) the protected fucosyl- chitobiose-L-asparagine derivative 23. Similarly, the per-0-acetylated

Page 304: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

GLYCOPEFT'IDE SYNTHESIS 28 1

BzlO

H3C

,OAc Aloc-HN- CH-COOIBu

20 R=N,

21 R=NH2

Aloc-HN- CH- COOfBu

2 2

A l a - CH+H-CHzCCC IBu =(CH3I3C

trisaccharide azide having the 4-0-(a-~-fucopyranosyl)lactosamine struc- ture was synthesized by reaction of 2,3,4-tri-0-(4-methoxybenzyl)-a-Lfu- copyranosyl chloride with a lactosamine azide derivative, subsequent oxida- tive removal of the 4-methoxybenzyl (Mpm) protecting groups, and acetylation of the liberated hydroxyl groups. Reduction of the azido function and condensation of the resulting amine with 1 -ally1 N-trichloroethoxycar- bonyl-L-aspartate afforded the 4-0-(a-fucosyl)lactosamine glycopeptide having the Lewis" antigen structure (21).

c. 1-Isocyanates as Precursors. - Per-0-acetylglycosyl isothiocyanate derivatives of monosaccharides were first coupled with L-aspartate deriva- tive 6 by Khorlin and coworkers (22) to form l-N-(~-aspart-4-oyl)glycosyl derivatives. This procedure was modified and has successfully been applied for the synthesis of the P-D-mannosyl-chitobiosyl-L-asparaghe glycopeptide 24 via reaction (23) of the per-0-acetyl derivative of 25 with L-aspartate derivative 22. It is noteworthy that attempts to prepare the 1 -azido derivative

H,N-CH-COOH

I H O ~ o H ~ o & / ~ o &HZ

HO

HO AcHN

AcHN

2 4

.OH& HO

HO N=C=S

AcHN AcHN

2 5

Page 305: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

282 HAM G. GARG et d.

26

Trimsfhylsllyl ulde I SKI,

I CH3

ACO NR AcO

AcHN AcHN

27 R=N2

28 R = H?

(26) of this trisaccharide by treatment of oxazoline 27 with trimethylsilyl a ide and trimethylsilyl trifluoromethanesulfonate or SnCl,, under condi- tions described by Warren and associates (1 8), with the goal of synthesizing the corresponding 1 -amino trisaccharide derivative 28, were unsuccessful.

d. Reactions of 1-Hydroxy-D-glycosamines with Ammonium Hydrogen Carbonate. - 2-Acetamido-2-deoxy-~-glucose (24,25) (29) and per-O-ace- tylated chitobiose (26,27) (30) have been reported to react directly with ammonium hydrogen carbonate (NH,HCO,) to give the 1 -amino deriva- tives 31 and 32, respectively. These derivatives were coupled with L-aspar- tate derivative 33 to give the corresponding glycosyl- L-asparagine deriva- tives 34 and 35. Derivatives 34 and 35 are useful intermediates for solid- phase synthesis of glycopeptides (described in Section V).

-../.g+ HO AcHN OH or Ac:cO*o& AcHN AcO AcHN OH

2 9 30

3 1 32

Page 306: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

GLYCOPEFTIDE SYNTHESIS 283

Fmw-HN -CH -COOtBu Fmoc-HN -CH -COOtBu I I I

&I,

I C Hz

I Fmoc-HN -CH -COOtBu

I c=o

or AcO NH , .. . &OOPIp HCtlN

AcHN 3 3

3 4 PIP =CeF5-

IBU = (CH313CC

Fmac- = CH,GCO- I

3 5

2. Selective Deprotection and Peptide-Chain Enlongation Glycopeptides (3) containing a 2-acetamido-N-( ~-aspart-4-oyl)-2-deoxy-

/3-D-glucopyranosyl residue were obtained, together with their regioisomers, by using the benzyloxycarbonyl (Z)-protected L-aspartic anhydride and gly- cosylamine 5. After separation of the regioisomers, elongation of the peptide chain was performed in 3 in the direction of the carboxyl terminus by Garg and Jeanloz (28 - 30) and in the direction of the amino terminus by Shaban and Jeanloz (31). In the past several years Kunz and associates (14- 16,19,2 1,32 - 37,39) have developed different protective groups and protect- ing-group combinations of orthogonal stability (that is, the alternative sta- bility and cleavability of two or more protecting groups) for the amino and carboxyl termini of such glycopeptides as 36 and have used them extensively for elongation of the peptide chains of glycopeptides. The allyl ester as a temporary protecting group (32) for protection of the L-aspartic acid residue of the chitobiose glycopeptide 36 was introduced (14) for elongation of the peptide chain. The allyl ester group may be removed either by rhodium( 1)-catalyzed (1 6) or by palladium(0)-catalyzed (32) cleavage to give ( 14,15) the carboxyl-deprotected glycopeptide derivative 37. Chain lengthening at the C terminus with peptide allyl esters in the presence of EEDQ ( 17) gave derivatives 38 and 39, respectively. The free glycopeptide

Boc-HN- CH -COOAll Boc-HN-CH-COOH I I

36 37

Page 307: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

284 HARI G. GARG et al.

Boc HN-CH-COOR

I CH,

A:c* 2c& I + peplide ally1 esfers (H (Xaa),-OAll: EEDa -

AcHN AcHN

38 R = L-Phe-L-Thr-OAll

39 R = L-Ala-L-Thr~L Ala~L~Ser OAll

amide 40 was obtained by treatment of 38 with methanolic ammonia (1 5,33). Treatment of39 with trifluoroacetic acid resulted (1 5,33) in removal

Boc-HN- CH - COO-L-Phe~L-Thr-NH,

I CHZ

HO HO *o&i=o NH

HO AcHN

AcHN

4 0

of the tert-butyloxycarbonyl (Boc) group to give 41. The protecting-group combination used in these syntheses offers the possibility of extension of the peptide chain in both directions.

CF,COOH H,N-CH -COO-L-Ala L-Thr-L-Ala-L-Ser OAll

I CHZ

I /OAC c=o

39 CF3COOH -

A r U h l \

4 1

The noteworthy advantages of the allyl ester are (a) it is readily introduced into amino acids; (b) after isomerization (to 1-propenyl) by a palladium(0) catalyst it may be removed under weakly acidic or basic and neural condi- tions (32), even if sulfur-containing amino acids are present (34); (c) it shows orthogonal stability to the tert-butyloxycarbonyl and 9-fluorenylmethoxy- carbonyl groups (lo); and (d) it is not affected by the hydrogen fluoride- pyridine complex (35).

The allyl protecting group has also been demonstrated to be useful for blocking of side-chain functionalities (36). Thus, the palladium(0)-catalyzed

Page 308: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

GLYCOPEPTIDE SYNTHESIS 285

removal of the P-ally1 ester protection from aspartic acid peptides, even from Asp - Gly and Asp - Ser sequences, was achieved without transpeptidation (36).

The combination of the N-terminal allyloxycarbonyl (Aloc) group (34) with the tert-butyl ester proved to be a particularly efficient tool in the synthesis of complex glycopeptides, as has been demonstrated in the synthe- sis of sensitive fucosyl chitobiose glycopeptides (19). The Aloc group was

0 v O C C " H II ,COOtBU

\ I CH

I OAc

H & h o A c CHz

H-NH CoOtBu

..dN

4 3

4 2

Aloc-Ala-Leu- NH , ,CWH Aloc-Ala-Leu-NH c o o t ~ u \ , \ , 'CH cn

OAc Aloc- Ala-Leu-OH

Me2N(CH2)3N=C=NE~ IDAECI H,C-OAc {HZ CFiCOOH

I 1 ,OAc n c=o

4 4 4 5 /

J

4 6

AcHN

removed from the trisaccharide - asparagine conjugate 42 with complete chemoselectivity by palladium(0)-catalyzed ally1 transfer to N,N-dimethyl- barbituric acid as the allyl-trapping nucleophile, to give the N-deprotected conjugate 43 almost quantitatively. Chain extension using a water-soluble carbodiimide furnished 44.

Page 309: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

286 HARI G. GARG et al.

Removal of the tert-butyl ester protection from 44 proceeded quantita- tively to give 45 by the action of trifluoroacetic acid. Because of the indirect protection by the 0-acetyl groups, the sensitive fucoside bond remained absolutely stable. It is noteworthy, that the fucoside bond in the analogous fucosyl chitobiose derivative 23, partially protected by benzyl ether groups, was completely decomposed upon treatment with trifluoroacetic acid. Chain extension on 45 furnished the trisaccharide hexapeptide 46, which is a partial sequence of an envelope glycoprotein of a leukemia virus ( 19). Com- plete deprotection and the binding of the synthetic glycopeptide to bovine serum albumine has been achieved (19). The synthesis of the totally pro- tected 2-acetamido- 1 -N-(~-aspart-4-oyl)-2-deoxy-~-~-ghcopyranosy~ de- rivative 47 containing the I ,3-dithian-2-ylmethyl (Dim) ester group has been described (37). Selective C-terminal deprotection to give derivative 48 was carried out by oxidation to the disulfone and its immediate /I-elimina- tion. These cleavage conditions do not affect the Boc group, the 0-acetyl

Boc-HN- CH- COODim Boc-HN-CH-COOH

I CH,

AcHN

48

groups, or the glycoside bond. In this way, the synthesis of glyco-tri-peptides and -tetrapeptides by amino- and carboxyl-terminal elongation of the pep- tide chain has been achieved (37).

Using the 2,2,2-trichloroethoxycarbonyl (Teoc) group (38) in combina- tion with the ally1 ester protecting group, Kunz and Marz (39) have synthe- sized glycosylated peptide T derivatives, which are partial structures of the -glycoprotein gp 120 of the human immunodeficiency virus HIV- I . In this context, the N-Teoc group was selectively removed from the per-0- acetylated Lewisa antigen trisaccharide - asparagine conjugate 49 by using zinc in acetic acid. Subsequent chain-extension furnished the trisaccharide

R-HN-CH-COOAll

I A A J * m c T o H y 9 OAc CH2 I

AcHN AcHN

49 R = C13C-CH2-O-C0 (Tcoc)

50 R = Boc-Ala-Ser(1Bu)-Thr(lBu)-Thr(tBu)-Thr(tBu)

Page 310: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

GLYCOPEPTIDE SYNTHESIS 287

hexapeptide allyl ester 50, which was carboxyl-deblocked via palladium(0)-catalyzed cleavage and extended to the N-glycosyl peptide T derivative. Complete deblocking of the glycosylated HIV- 1 structure has been achieved (39). Analogous glycosylated peptide T derivatives have also been synthesized by solid-phase glycopeptide syntheses employing allylic anchoring groups (40).

111. 3-0-GLYCOPEPTIDES OF L-SERINE OR L-THREONINE 1. Formation of the 3-0-Glycosidic Linkage

a. Koenigs - Knorr Methods. - Jones and coworkers (4 1 ) prepared the 3-0-(2-acetamido-2-deoxy-/3-~-glucopyranosyl)-~-se~ne derivative 51 by treating 2-acetamido-3,4,6-tri-~-acetyl-2-deoxy-~-~-glucopyranosyl chlo- ride with N-(benzyloxycarbony1)-L-serine methyl ester in the presence of silver carbonate. Bencomo and Sinay (42) used the N-benzyloxycarbonyl (Z) group in combination with the tert-butyl ester for the carboxyl protection of L-serine. The tert-butyl group was removed with trifluoroacetic acid without cleaving the 0-glycosidic bond. Nevertheless, glycopeptides involving 0-glycosylated L-serine/L-threonine are more or less acid-sensitive (hydrol- ysis and anomerization) and alkali-labile @-elimination). This holds true, in particular, for the /3-xylose - L-serine bond.

2-HN-CH- COOMe 2-HN- CH- COOMe

I

ACO

Silver carbonale

I AcO

OH ACO Ace++ AcHN CI iHz

ACHN

2 = C,H,CH,OCO. 51

To overcome these difficulties in the selective deprotection and chain extension, several carboxyl-protecting groups, namely, allyl ( 16,32), benzyl (43,44), tert-butyl(42), 2-bromoethyl(45), 2-chloroethyl(45), heptyl(46), 4-nitrophenyl(47,48), and pentafluorophenyl(49) for L-serine/L-threonine have been introduced or applied. Similarly, amino-protecting groups for L-serine/L-threonine that have proved useful for the synthesis of glyco- peptides are tert-butyloxycarbonyl (50), 9-fluorenylmethoxycarbonyl (43,44,48), 2-(2-pyridyl)ethoxycarbonyl (5 1 ), 2-(4-pyridyl)ethoxycarbonyl (44,52), and 2-triphenylphosphonioethoxycarbonyl(53). Some applications of these groups have been discussed in earlier reviews (7- 1 1).

Schultheiss-Reimann and Kunz(43) applied silver triflate as the promot- ing reagent for the reaction of 2,3,4-tri-0-benzyl-c-~-glucopyranosyl bro- mide (52) with N-(9-fluorenylmethoxycarbonyl)-~-serine benzyl ester to

Page 311: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

288 HAM G. GARG et al.

furnish the 3-0-(j?-glucopyranosyl)-~-serine derivative 53. The protecting- group combination Fmoc/OBzl of 53 allows versatile and selective depro- tection and chain extension for the synthesis of glycopeptides. Removal of the Fmoc group from 53 was achieved selectively and quantitatively by using morpholine (43,44). Under these conditions, the 0-glycosyl - serine (- threonine) linkage is unaffected. Peptide condensation gave the glycotri- peptide 54, which was completely deprotected by subsequent application of morpholine to remove the Fmoc group, hydrogenolysis of the benzyl ester, and methanolysis of the carbohydrate ester groups catalyzed by the weak base hydrazine, to give (43-45) 55. This methodology (4334) proved to be

Fmm-NH-CH-C00Bz1 I

1. MOpholine 1. Maphollne Fmm-Asn-Leu -NH-CH -COOEzl

* 2 H*/P&C 2. Fmc-Asn-Leu-OH

H-Asn-Leu-NH-CH-CWH

I (OH FHZ

of quite general applicability for glycopeptide syntheses, as in the synthesis of tumor-associated T,- and T-antigen glycopeptides (44) or in solid-phase synthesis (see Refs. 25, 26, 55, and 56; see Section V).

Kenne and associates (57) have applied this procedure for the synthesis of a-D-mannopyranosyl derivatives linked to L-serine/L-threonine (59 and 60). Compounds 59 and 60 were obtained by coupling 2,3,4,6-tetra-O-acetyl-~- mannopyranosyl chloride (56) with Fmoc-L-serine benzyl ester or Fmoc-L- threonine benzyl ester in the presence of silver triflate and 4-A molecular sieves, followed by deprotection (57).

Paulsen and associates (5 8) synthesized mono- and di-D-galactopyranosyl derivatives of L-serine/L-threonine. Condensation of the 2-azido-2-deoxy- glycosyl halides 61 and 62 with the benzyl or tert-butyl esters of N-benzyl- oxycarbonyl (Z)-protected L-serine, L-threonine, and L-leucyl-L-serine (63 - 65) in the presence of silver carbonate, silver perchlorate, Drierite, and molecular sieves gave (59) the corresponding 0-glycopeptides 67 -69. The free glycopeptides were obtained after total deprotection.

Page 312: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

GLYCOPEPTIDE SYNTHESIS 289

Fmoc-HN-CH- COOBzl

I I

HC-R Fmoc-HN- CH- COOBrl

511YBilrillae * I nlol&"lor SiBYBS

I 57 R = H

* A c 6 c O e o 58 R=CH3

HC-R

OH A c O

OAc AcO

56 R = H, or R = CH,

a) Morphaline b) HydrOgBndySiSIPdC c) Hydmine hydrate

H,N- CH- COOH

HC-R I I

59 R = H 60 R=CH3 HO

2-L-Leu-HN- CH-COOBzl I

0 ,OAc +

.

I 6 1

R'-HN-CH-COOR

I I

w + 2-A

HC-R

ACO AcO &:+ A C O F O & ~

AcO OAc OAc N3 OAc

Br AcO 6 2

63 A = L Leu-L-Ser-OBzl 67 R = H, R' = 2. R" = IBU

68 R =H, R'=Z-L-LeU, R " = Bzl 64 A = L-Ser-OlBu

65 A = L-Thr-OlBu 69 R = CH3, R' = 2, R" = IBU

b. Trichloroacetimidate Method. - Kunz and Waldmann (32) applied the trichloroacetimidate method (60) to couple the tri-0-benzylated xylosyl residue of 70 to N-(benzyloxycarbony1)-L-serine ally1 ester (71) to give the xylosyl-L-serine derivatives 72 and 73 (ratio ofp to a, 6 : 1).

In order to obtain the biologically interesting sialyl T, antigen glyco- peptides, N-(benzyloxycarbonyl)-O-[methyl(5-acetamido-4,7,8,9-tetra-O- acetyl-3,5-d~deoxy-~-g~ycero-a-~-gu~uc~~2-nonu~opyranosy~)-onate-(~ -

Page 313: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

290 HAM G. GARG et al.

2-HN-CH - COOAll Z-HN- CH- COOAll

I + I

CHZ I Z HN-CH-COOAll

0F3. - 3 P C - I

BZlO OH

7 0 7 1 7 2 II

NH 7 3

3) - 0 - (2,4,6 - tri - 0 - acetyl -p - D - galactopyranosyl)] - ( 1 --j 3) - 0 - L - seryl- [methyl( 5 -acetamid0 - 4,7,8,9 - tetra- 0-acetyl - 3,5 -dideoxy-~-glycero-a- D - gulucto-2-nonulopyranosyl)onate[(2 + 6)-0-(2-acetamido-4-0-acetyl-2- deoxy-a-~-galactopyranosyl)]-( 1 - 3)-~-serine benzyl ester has been syn- thesized by employing the corresponding trichloroacetimidate derivative and N-(benzyloxycarbony1)-L-serine benzyl ester. The ratio of the a- and p-glycosides formed (6 1) was 2 : 5.

c. Glycosyl Fluorides.-The synthesis of a mixture of a and p-D-xylo- pyranosyl-L-serine derivatives (72 and 73) and the 2,3,4,6-tetra-O-pivaloyl- p-D-glucopyranosyl-L-serine derivative 76 from the glycosyl fluorides 74 and 75 and N-(benzyloxycarbony1)-L-serine ally1 ester (71) in the presence of boron trifluoride etherate has been achieved (62). The stereoselective forma-

,OPlV

F 7 4 7 5

Z-HN-CH- COOAll

7 2 I 73 + or plvo+~Hz PWO 0

PlVO

tion of derivative 76, the stability ofglycosyl fluorides, and the homogeneous reaction proceeding without heavy-metal promotion make this procedure an interesting alternative of Koenig- Knorr methods.

d. 1-Thioglycosides. - Thioglycosides have been introduced as efficient glycosyl donors (63). The ethyl 1-thio mono (77 and 78)-, di (79 and 80)-, and tri (81)-saccharides were condensed with N-( benzyloxycarbony1)-L-serine benzyl ester (82) in the presence of dimethyl (methy1thio)sulfoniumtrifluor- omethanesulfonate (DMTST) (63) as promoter to give (64) the correspond- ing a-0-glycopeptides (83 -87). Derivative 87, on reduction with hydrogen sulfide and acetic anhydride gave the 2-acetamido-2-deoxy derivative 88, which was completely deprotected by reduction and the action of metha- nolic ammonia to give (64) 89. Thioacetic acid (65) has also been employed for converting the azido group of carbohydrate residues to an acetamido

Page 314: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

GLYCOPEPTIDE SYNTHESIS 29 1

group. An alternative direct transformation of 2-azido-2-deoxyglycosyl pep- tide derivatives to the corresponding 2-acetamido derivatives has been achieved by applying a Staudinger reaction with tri-ocylphosphine in dry acetic acid (67,68).

TI R = A c

78 R = Erl

RO Z-HN- CH- COOEzl

SC2Hg + 'HZ - I

RO

N, OH OR

82

AcO

OEzl

81

Z-HN-CH-CCOEzI I

2-HN-CH- CoOBzl

I y 2

I

- RO&O

RO OR

ZHN-CH-CCOBzl

I CH, I

+ RO@o&'

R'O OR OR

RO

2-HN- CH- C00Er l I CH,

EzlO I

Ad& AcO OAc

87

H2N-CH-CCOH

I

OH

HO

89

Page 315: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

292 HAM G. GARG et al.

a-D-Mannopyranosyl-( 1 + 2)-a-~-mannopyranosyl derivatives of L-ser- ine and L-threonine glycopeptides (95 and 96) were prepared by treating ethyl 2,3,4,6-tetra-O-benzyl- 1-thio-a-D-mannopyranoside (90) with bro- mine, followed by treatment with ethyl 3,4,6-tri-O-benzyl- 1-thio-a-D-man- nopyranoside (91) to give the ethyl 1-thio derivative of the disaccharide 92. Treatment of 92 with Fmoc-L-serine or L-threonine benzyl ester gave 93 and 94, respectively. On total deprotection, the freeglycopeptides 95 and 96 were obtained (66).

BZlO

Fmoc-HN-CH- COOBzl

I I

HC-R

I

H,N - CH- COOH

HC-R I I

2+0

Ho I 95 R = H 66 R=CH,

93 R - H 94 R-CH,

Using the ethyl 1-thio derivative (97) of 2,3,4-tri-O-benzoyl-~-xylose, the fully protected and free 0-glycopeptides 99 and 100, having the N-terminal amino acid sequence 3 to 6 (98) of the protein core of a proteodermatan sulfate have been prepared (69).

R"-L-Ala-L-Ser-Gly-L-IleOFI

2-L-Ala-L-Ser-Gly-L-Ile-OBzl I - DMTST ROR-b B z ~ z ~ S C 2 H 5 0 0 2 + OH OR

97 Bz=C,H5C0 98 99 R = Bz, R' = C6H&H2, R" = C6H,CH20CC 100 R = R ' = R " = H

e. 1-0-Acetyl Activation. -Glycopeptides 101 and 102, containing the A3 - A, sequence of proteodermatan sulfate and xylose, have been synthe-

Page 316: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

GLYCOPEPTIDE SYNTHESIS 293

sized alternatively by activation of the 1 -0-acetyl group of the xylose deriva- tive 103 with trimethylsilyl triflate (69,70). Thus, compound 103 reacted with 104 and 105 in the presence of trimethylsilyl triflate to give the pro- tected glycopeptides 106 and 107, respectively, which on deprotection yielded glycopeptides 101 and 102.

R-L-Ser-Gly-L I le~OR ii~meihyliilyl

R-L-Ser-Giy I -L-lle-OBzl - rrdla,e y&+b A ~ a ~ O A c + OH OAC

OAc

103 104 R = CeH50C0

105 R = C,H,OCO-L-Ala

106 R - C,H,CH,OCO

107 R - C,H,OCO-L-Ala

R - L-Ser -Gly - L-lle-OH

101 R = H

102 R = L-Ala

f. Electrophile-Induced Lactonization of Glycosyl 4-Pentenoates. - Carbohydrates having an anomeric pent-4-enoic acyl group have been used as glycosyl donors to prepare glycopeptides (7 1). Thus, condensation of the glucopyranose derivatives 108 and 109 with pent-4-tenoic acid in the pres- ence of N,N'-diisopropylcarbodiimide and cuprous chloride gave the pro- tected glucopyranosyl4-pentenoates 110 and 111, respectively. Activation

0

4penlenoic acid. CuCI.

R3 + N.N'diisoptopyl carbodiimide R3 R'O

1W R' = Bzl. R2 = R3 = OBzl

~

OH R'O

R2

110 R' = BzI. R2 - R3 = OBzl

Frnoc-HN--CH-COOBzl Frnoc-HN-CH-COOBzl I CH, NIS

OH lC%CF$%H BzlO 0

NIS = N-lodasucontmlde

Page 317: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

294 HARI G. GARG ef a1

0

Teoc-HN - CH- C - L-Ala-OtBu

111 + Teoc- L-Ser OH I - L-Ala- OIBu

A**/cH2 I " 113 Tecc = CI&CH,OCO AcO

'% 114 (aP,O100)

of 110 and 11 1 with soft electrophiles, such as iodonium compounds, [sym- collidine,I]C10, or N-iodosuccinmide (NIS), trifluoromethanesulfonic acid, or l73-dithian-2-y1ium tetrafluoroborate in the presence of 3-A molec- ular sieves or Drierite and treatment with Fmoc L-serine benzyl ester or the serine peptide 113 gave glycopeptides 112 and 114, respectively. Condensa- tion of 11 1, having a participating group at C-2, proceeds with complete stereoselectivity. Because of the mild reaction conditions, this method is of potential interest for the glycosylation of preformed peptides.

2. Methods of Selective Deprotection, Followed by Peptide-Chain Elongation

Selective methods for deprotecting glycopeptides either at the amino or at the carboxyl terminus have been developed during the past decade by intro- duction and application of different combinations of protecting groups (Table 11).

The protected 0-xylosyl derivative (1 15) of L-serine (43) B-D-gdactopy-

TABLE I1 Different Amino and Carboxyl Protecting-Group Combinations of L-Serine/L-Threonine

Used for Glycopeptide Chain-Lengthening

Terminus

-NHz -COOH Reference

Benz ylox ycarbon yl Ally1 32,68

9-Fluorenylmethoxycarbonyl MY1 67,72 ferf-Butyl 42

Benzyl 43,44,56,13

4-Nitrophenyl 47,48,15 14

Pentachlorophen yl 75 Pentafluorophenyl 49 Phenacyl 66

2-(4-Pyridyl-ethoxycarbonyl Benzyl 44,73 Trichloroethoxycarbonyl MY1 39 All ylox ycarbon yl ferf-Butyl 19,33,76

Page 318: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

GLYCOPEPTIDE SYNTHESIS 295

ranosyl - (1 + 3) - 2 - acetamido - 2 - deoxy - a - D - galactopyranosyl - L - serine (44,73,74) (1 16), and the analogous L-threonine compound (44,73,74) 117 having the Fmoc group at the amino terminus and the benzyl group at the carboxyl terminus were used successfully for the synthesis of 0-glycopeptides. As was first demonstrated for 0-glucosyl and 0-xylosyl serine derivatives (43), this protecting group combination allows selective deprotection of either the amino terminus by using morpholine (43,44) or

FrnooHN- CH- COOBzl Frnoc-HN - CH - COOBzl I I CHZ I 0

L O B Z

I HC-R I 0

116 R = H

117 R=CH,

1 1 5

H I

B-D-Galp(l-13)-a-D-GalpNAc-(l -to) -L-Ser

P-D-Galp(l-13)-a-D-GalpNAc-(l +O) - L-Ser

~-D-Galp(l+3)-a-D-GalpNAc-(l -+O) -L-Ser

I I

AH 1 1 8

the carboxyl terminus by hydrogenation (8). Beginning at the carboxyl end and in the presence of EEDQ as the coupling reagent, these derivatives were used (74) as a step-by-step synthesis of the free glycosylated tripeptide 118.

Another combination of orthogonally stable protecting groups, which proved very useful in glycopeptide synthesis, consists of the Fmoc group together with the ally1 ester (67). Removal of the Fmoc group from glyco-

Frnoc-HN-CH-COOAll I

HC-R I I

119 R - H

120 R=CH,

H2N- CH-COOAll

I

morphdrm I HC-R

AcHN 0 .

12l R=H.

122 R=CH3

Fmoc-HN-CH-CCOH I I

HC-R

A c O e '

123 R - H

124 R -CH, AcO

Page 319: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

296 HAM G. GARG et al.

Frnoc Fmoc

a-D.AclGalpNAC-(l+O) -L-Thr a-D Ac,GalpNAc-[l+O) -L-Ser

u-D-Ac,GalpNAc-(l+O) - L-Ser a-D-Ac~GGalpNAc-(l+O) -L-Thr

OAI

I I I or I

I I

121 + 123 l lD0 or ___)

122 + 124

OAll

1 2 6 1 2 5

peptides 119 and 120 by morpholine gave the amino-deprotected derivatives 121 and 122. Selective removal of the ally1 ester group from 119 and 120 was performed by a pallidium (0)-catalyzed allyl-transfer reaction. However, as the Fmoc group is sensitive to morpholine, N-methylaniline a very weak base was applied as the allyl-trapping nucleophile. Under these conditions, the selectively deprotected compounds 123 and 124 were obtained in high yields and without side reactions. Coupling of 121 -124 in the presence of IIDQ furnished the protected diglycosyl peptide derivatives of 125 and 126, which have the partial structure of human glycophorin (67). Similarly, other glycopeptide derivatives containing T-antigen disaccharide units (1 27 and 128) have been synthesized by elongation of the peptide chain (67).

p-D-Ac,Galp(l-+3)-a-D-Bz~GalpYAc.(l+O) -Xaa

p-o-Ac,Galp(l~3)-a-o-B~2GalpNAc-(1+0) -Xaa 126 L-Thr

I 12' Xaa=L-Ser

I OAll

The Fmoc/OAll protecting-group combination has also successfully been applied in the synthesis of glycopeptides constituting the linkage-region structures of proteoglycans (72). Using Fmoc amino protection and the phenacyl ester of L-serine, the glycopeptides 129 and 130 have been obtained (66).

H I

H I

a-D-NeuFsAc-(2+6)-a-D-G~lpNAc-(l+O) -L-Ser

o.D-NeugAc-(2+6)-a-D-Gal~Ac-(l +0) -L.Ser

a-D-NsupsAc.(2+6)-a-D-G~l~Ac-( l -0) -L-Ser

I L-Val

OH

I I I

a-D-NeupsAc-(2-6)-a-D-GaiFNAc.(l+o) - M e ,

a-D-NeugAc-(2+6)-a-~-GalFNAc-~l+O) -L.Ser

L-Val

I I

I 1 3 0 1 2 9 OH

Synthesis of a number of 0-glycosides (such as 131 - 134) having different chain lengths between amino acids A, and Alo, which are part of interleu- kin-2, was accomplished by application of the Fmoc group in combination with the tert-butyl ester (77,78).

Page 320: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

GLYCOPEFTIDE SYNTHESIS 291

H L-Ala-L-Pro-L-Thr -L-Ser-L Ser-OH

I p - ~ ~ G a l p ( l --t3)-u-D~Ac3GalpNAc

1 3 1

H-L-Ala-L~Pra-L-Thr -L-Ser-L-Ser OH

I (1~D Ac,Gal@Ac

1 3 2

The synthesis were effected with the appropriate glycosylated derivatives of L-threonine (135 and 136). Selective removal of the ally1 ester derivative

Frnoc-L-Thr -01Bu Z- L-Ala- L-Pro -L-Thr - O!Bu

I I a-D-Ac,GalpNAc u-D-AclGalpNAc

1 3 5 1 3 6

137 by using [(Ph,P),Pd(O)] catalyst to give 138, followed by coupling with dipeptide 139, gave (68) the C-terminally extended glycopeptide 140.

2-L~Ser-L-Ala-L-Ala- OH 2-L-Ser -L-Ala-L-Ala- OAll

llPh3Pl.Pdnl. morpholine

AcO 1 3 7 AcO 1 3 8

2-L-Ser -L-Ala-L-Ala-Gly-L-Ala-OAll AcHN I

Tumor-associated T, and T-antigen type 0-glycopeptides (144 and 145) have been obtained by starting with the Fmoc/Bzl protecting-group combi- nation. Continuing with condensation of the amino-deblocked component with 2-(4-pyridyl)ethoxycarbonyl(Pyoc-~-serine yielded the glycopeptide derivative 141. Hydrogenolysis to 142 and coupling with 143 promoted by EEDQ gave the TN-antigen glycopeptide 144. In the selective deprotection of the carboxyl terminus, the Pyoc group has an advantage as it is stable to hydrogenolysis. The T-antigen glycopeptide derivative 145 was obtained similarly (44,73).

P y x - L-Ser -L Ssi- OBd Pyoc-L-Ser -L-Ser -OH

ACO@ hydropnolysia ~ AcO@

AcO 1 4 1 AcO 1 4 2

Page 321: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

298 HARI G. GARG et al.

L-Thr -0Bzl

Pyoc-L-Ser-L-Ser-L-Thr -0B21

EEDO I I Y Y

A& 1 4 3

AcO AcO

145 Y = AcO

AcO% OAc AcHN

144 Y =

The Pyoc group is stable to acids and bases, but may be selectively re- moved after conversion into the N-methylpyridinium form and subsequent treatment with morpholine, to give 146. Treatment of 146 with acetic anhy- dride in pyridine gave 147. Hydrogenolysis of the benzyl ester group of 147 followed by methanolysis catalyzed by hydrazine afforded (44,73) the de- protected derivative 148.

a-D-Ac,Gal@Ac a-D-R,GalENAc AC& i I 1 C Y l I pyrrdlne

144 H-L-Ser-L.Ser-L-Thr-oB~1 - Ac-L-Sar-L.Ser-L-Thr-OR

I I I I R a-D-R,Gal@Ac

2 mOrphOi,ne

H ~-D-Ac,G.~&NAc

147 R = Ac. R = OBzl 1. H P d - C 2 hydrazine. MeOH L 148 R = R. =

146

IV. BINDING OF GLYCOPEPTIDES TO PROTEINS

As proteins are insoluble in organic solvents, their coupling with glyco- peptide derivatives having a definite structure has to be effected in water solution. Thus, the carboxyl-deprotected glycopeptide derivative 148 has been coupled with bovine serum albumin (BSA) (about 25 glycopeptide molecules per protein molecule), presumably at the 6-amino group of L-ly- sine residues, to give (44,73) the conjugate 149.

148

a-D-GalmAc

I 1 -~hyl-~(3dimethylimnopmyl) o M l m d e h y d m h l a n d s (DAEC)

I -hydroxybenrbna2ole (WBI) + BSA - Ac - L.Ser -L-SaI - L-Thr-NH-ESA

I a-D-Gal@Ac

1 4 9

A glycopeptide - BSA conjugate (150) containing the tumor-associated T-antigen structure has been constructed (44,73).

P-D-Galp(1-3)- a-0-GalpAc

I Ac-L-Sel-L-Sei-L-Thr-NH-BSA

I P-DCalF(l--t3)- a-D-GalpNAc

1 5 0

Page 322: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

GLYCOPEPT'IDE SYNTHESIS 299

The trisaccharide - hexapeptide partial structure (151) of a leukemia-virus envelope glycoprotein, which was obtained from 46 (see Section II,2) via saccharide and carboxyl deprotection, has also been coupled to BSA to furnish a synthetic glycoprotein 152 containing 12% of carbohydrate in. form of the trisaccharide (1 9).

HO

CAla

the

NOC

L-Na

L - L a I

I I

1 5 2 "i'" BSA

V. SOLID-PHASE SYNTHESIS

The solid-phase synthesis of glycopeptides was first realized by applying the polymeric benzyl ester principle of Merrifield. According to this meth- odology, Lavielle and associates (50) used N-(tert-butyloxycarbony1)-0- glycosyl serine derivative 153 for condensation with resin-linked alanine 154.

H-Gly-L-Ser-L-Ala-OH 1) coupling with

Boc-HN-CH-COOH I I CH3

155

I H2N-CH

1 5 4

2) N-depmleclion 31 colpling with Boc-glycine 4) deproleclion wilh dry HF

AcHN

AcO

153

The N-(tert-butyloxycarbonyl) protecting group was removed by trifluo- roacetic acid. Subsequent coupling to N-(tert-butyloxycarbony1)glycine and final treatment with hydrogen fluoride gave the glycopeptide 155. In this instance, the success of the release of the glycopeptide from the resin through cleaving of the benzyl ester depends upon total exclusion of moisture from the HF. The glycosylated somatostatin 156 was synthesized similarly, start- ing from resin-linked cysteine (49).

I L-Ala-Gly -L-Cys - L-Lys -L-Asn- L-Phe -L-Phe - L-Trp- L-Lys - L - l l r - L-Phe- L-Thr -L-Ser - L-Cys

I p-GlcpNAc

156

Page 323: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

300 HAM G. GARG et al.

A new allylic anchoring technique for elongation at the N terminus of peptides linked to a polymer support has been developed (79,80). The an- choring allylic ester is formed by reactin of the cesium salt of the N-protected amino acid, for instance, tert-butyloxycarbonyl-L-alanine, with 2-(4- bromo-2-butenamid0)methyl-polystyrene (157) (HYCRAM", trademark of Orpegen, Germany). The allylic ester linkage is stable to the acids and bases required for amino deprotection. Removal of the N-Boc group of 158 gave derivative 159. This product could be extended by condensation of the 0-glycosylserine derivative 160 to furnish 161.

Hp I

R- L - A h - 0 W C / N

II

L J v 158 RIBOC HycrarnTM

CF3CMH L 159 R =ti

161 R=FmOC

wh*'"e L162 R = H

I.Z-Ala-OHIoIc. H O B 1

Z-L-AI~-HN-CH-CO-L-AI~.OH I

Ez0-Y €310

163 BZO

Removal of the Fmoc group was achieved by treatment with morpholine - dichloromethane, leaving the allylic anchor and the O-glyco-

Page 324: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

GLYCOPEPTIDE SYNTHESIS 30 1

1 6 4

sylserine linkage unaffected. The resulting resin-linked glycopeptide 162 was extended with Z-alanine. Finally, the glycotripeptide 163 was detached by palladium(0)-catalyzed allyl ester cleavage (32,79). Under the prevailing, practically neutral conditions, the other protecting groups and the O-glyco- sidic bond remained untouched (79).

The allylic HYCRAM derivative was subsequently modified by insertion of a standard amino acid between the aminomethyl resin and the hydroxy butenoic acid moiety. Using this allylic anchor, the resin-linked, glycosyl- ated HIV peptide T-derivative 164 was synthesized by application of Fmoc amino protection and sidechain protection with tert-butyl groups. The lac- tosamine peptide T (165) could be released from the resin by application of the palladium(0)-catalyzed allyl-transfer reaction to N-methyl aniline as the allyl acceptor.

(Ph3P),Pdo, cat. Ph-NHMe I DMSO

A& 165

166

h 0

Page 325: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

302 HAlU G. GARG et al.

Similarly, the glycosylated peptide T derivative 166, carrying one N-gly- cosyldically and two 0-glycosylically linked saccharide side-chains was suc- cessfully synthesized (40,8 1).

Paulsen and associates (55) described the synthesis of the glycopeptide 167, having a partial sequence of porcine glycophorine, on resins having acid-sensitive alkoxylbenzyl ester anchoring-groups. The application of this technique is based on use of the Fmoc group for temporary amino protection and its selective removal with morpholine. The N-terminal tevt-butyloxy- carbonyl group and the alkoxylbenzyl anchor were simultaneously cleaved from 167 with 95% trifluoroacetic acid, and subsequent base-catalyzed methanolysis gave 168.

a-0-Gal@Ac

I L-AIa-L-Thr-LL-Val- L.Thr-L-Ala-GIy -OH

I a-D-Gal@JAc

1 6 6

Luning and coworkers (56) also used the Fmoc group for temporary amino protection and employed a commercial resin (SASRIN", trademark of Bachem, Switzerland) having a dialkoxybenzyl anchoring group. The synthesized glycopeptide could be cleaved from the resin with 1% trifluoro- acetic acid in dichloromethane.

Meldal and Jensen (49) utilized pentafluorophenyl esters and performed solid-phase glycopeptide synthesis, again based on temporary Fmoc protec- tion of glycosyl - amino acid components. Per-0-acetylated derivatives, for instance 169, were used as building blocks, as in the synthesis of the N-glyco- peptide (25) H - L- Thr - L - His- L - Ala - L - Ser - L - Asn(G1cNAc) - Gly - L - Ser - L - Met - L - Ser - L - Gly - OH. Applying this technique, O-glycosyl- ated derivatives of the insulin-like growth factor 1 (Ref. 82) and porcine submaxillary gland mucin (83) have been synthesized successfully, The method was shown to be also useful in multiple-column syntheses (83).

Otvos et a/. (27) adopted the Fmoc-pentafluorophenyl ester methodol- ogy and have applied 0-deacetylated glycopeptide intermediates (170 and 171) for solid-phase synthesis of glycopeptides. In this way, the N-glycopep- tide (26) H -Gly - L - Lys- L- Ala- L- Tyr- L- Thr- L- Ile - L- Phe- L -

Page 326: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

GLYCOPEFITDE SYNTHESIS 303

Fmoc-HN- CH--COOP@ Fmoc-HN-CH-CDOPlp I I ..a'=" Y

RO NH AcHN

AcHN AcHN

169 R=Ac

170 R = H

171

Asn(G1cNAc) - L - Lys - L - Thr - L - Leu - L - Met - NH, and glycosylated derivatives of peptide T have been obtained (84).

VI. ENZYMES AS TOOLS FOR GLYCOPEPTIDE SYNTHESIS

The high specificity and stereoselectivity of enzymes, as well as the mild conditions under which they react, make enzyme-catalyzed reactions versa- tile tools in the synthesis of glycoconjugates. In some instances, an enzymic one-step transformation affords higher yields then the conventional and more-complex chemical synthesis. The application of enzymes in glycopep- tide synthesis is under active development for selective deprotection and glycosylation purposes.

1. Selective Enzymic Deprotection

In the synthesis of glycopeptides of biological interest, the selective depro- tection of various protected groups often requires neutral conditions. En- zymes operate at neutral, weakly acidic, or weakly basic conditions and in many instances combine these advantages with high selectivity. Some of them catalyze the processes with broad substrate specificity. The application of biocatalysts to effect the removal or introduction of protecting groups offers viable alternatives to chemical methods used (85 -87). Thus, the re- moval of the heptyl ester from the glycopeptide derivatives 172- 174 has been achieved, to give the carboxyl-deblocked glycopeptides 175 - 177, re- spectively, in high yield (46).

During this process, the amino protection, the azido group, the base-labile acetyl groups, and glycosidic linkages remain totally unaffected. This proce- dure is both efficient and experimentally simple. The scope of enzymic protecting-group techniques has been demonstrated by the selective cleav- age of glycopeptide tert-butyl esters by action of the enzyme termitase (87).

2. Glycosylation Reactions Transfer of glycosyl residues to saccahride acceptors under catalysis by

glycosyltransferases has been studied in recent years (85,88,89). In contrast to chemical glycosylation, enzymic extensions of the saccharide chain of

Page 327: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

304 HARI G. GARG et al.

2-HN-CH-CCOHep

I I

CH2

AcoE+ AcO OAc

172

2-HN- CH- CCOH

I

Ace# AcO OAc

175

Fmoc-HN- CH-COOHep

I I CH2

Aco#o

AcO OAC

173

Fmoc-HN-CH-COOH

I CHz

Ace@ AcO OAc

176

2-HN-CH-Cmep

I

ACU A c b r N,

174

177

glycopeptides occur not only under neutral conditions but without extensive protecting-group manipulations. As a rule, the glycosyltransferase-pro- moted glycosyltransfer reactions proceed with high regio- and stereoselec- tivity. Complex saccharide structures difficult to construct by chemical methods, such as sialoglycosides, become efficiently accessible by glycosyl- transferase reactions (89). Paulson et al. (90) have reported the synthesis of the N-acetylneuraminyl -1actosamine-L-asparagine derivatives 180 and 181.

Starting from the 2-acetamido-2-deoxy-~-~-glucopyranosyl-~-aspara~ne derivatives 178 and 179, the (1 + 4-P-~-galactosyl and (2 + 6)a-NeuSAc moieties, respectively, were coupled by two subsequent enzymic glycosyla- tions in a "one-pot" procedure. In both glycosyltransferase reactions, alka- line phosphatase was used to decompose nucleotide diphosphate in order to prevent product-induced inhibition generated in the course of reaction.

R-HN-CH-COR I f".

Ho&$o Ho

H O I R -HN-

.NH

180 R L-Ssr-L-Thr-L IleOH. R - AbC-L-Phe

181 A - GlyGlyOH. R = H-Glq-Glq

Page 328: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

GLYCOPEFT'IDE SYNTHESIS 305

Enzymic synthesis of the trisaccharide asparagine conjugate a-D- (186) NeupAc-(2 + 6)-P-~-Galp-( 1 + 4)-/3-~-GlcpNAc-( 1 + 4N)-~-Asn

has been reported by AugC and coworkers (9 1).

R'-HN - CH-COR R'-HN-CH-COR I I CH2 I

E C 2 4 1 9 0 = HO &o*i=o CH,

NH HO

HO NH HO

+5J=O AcHN AcHN OH

HO

182 R = R . = H

183 R I Me. R' = AC

184 R = R ' = H

185 R=Me. R = A c

H,N-CH-COOH

AcHN I EC 2 498 1

____L

NH HO

OH AcHN

186

Two enzymic steps are involved in the synthesis, starting from the 2- acetamido-2-deoxy-~-glucose - L-asparaghe derivative 182: (i) galactosyla- tion catalyzed by immobilized N-acetyllactosamine synthase (EC 2.4.1.90) to give 184 and (ii) sialylation catalyzed by soluble CMP-N-acetylneur- aminate [ P-D-galactopyranosyl-( 1 - 4)-2-acetamido-2-deoxy-~-~-gluco- pyranose-a-(2 + 6) sialyltransferase (EC 2.4.99. l)] to give glycopeptide 186.

Similarly Thiem and Wiemann (92) conducted the galactosylation of the 2-acetamido-2-deoxy-~-glucose - L-asparagine derivative 183 and the chitobiose- L-asparagine derivative 187 to furnish the di- and trisaccharide derivatives 185 and 188, respectively, by using galactosyltransferase.

AC-HN-CH-CQOCH, I

187

I CH2

no

n o ~ o H ~ o ~ ~ = o no NH

AcHN AcHN

1 8 8

Likewise, the galactosylation of 0-glycopeptide derivatives 189 - 191 to give 0-lactosamine- L-serine derivatives (192 - 194) using ( 1 * 4)-/?-~-

Page 329: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

306 HARI G. GARG et al.

galactosylatransferase has been accomplished (93). This reaction illustrates that both N- and O-glycopeptides can be subjected to enzymic elongation of the saccharide chains.

R-L-Ser -L-Ala-OlBu

I

OH

189 R F C6H&HZCC0

190 R = CCI&H&O

191 R = C6HSCH$xX-L-Ala

R-L-S~~-L-AI~-OIEU

I HO &gNHAC

HO

OH

192 R = CsH&H2CC0

183 R = CCI,CH@O

194 R = C.H5CH2CC&L-Ala

Although the limitations of the enzymic reactions lie in the substrate selectivity and in the availability of enzymes and appropriate nucleotide sugar derivatives, further developments in this field will offer an interesting and efficient potential for the synthetic construction of glycopeptides of biological interest.

VII. ADDENDUM A number of publications on glycopeptide chemistry have appeared since

this manuscript was submitted. Cohen-Anisfeld and Lansbury (94) reported on a convergent synthesis ofglycopeptides. Peptides containing aspartic acid unblocked at the P-carboxy group were synthesized and, subsequently, con- densed with glycosylamines.

The optimized solid-phase synthesis of glycopeptides using Fmoc pro- tected glycosyl amino acids activated as pentafluorophenyl esters was dem- onstrated by Meldal et al. in a number of examples including glycopeptide .T-cell epitopes (93, mucin glycopeptides (96), mannose-6-phosphate con- taining glycopeptides (97), N-glycopeptides (98), and O-glycopeptide struc- tures of a blood clotting factor (99).

Lewis” antigen glycopeptides carrying clustered saccharide side chains were synthesized according to the Boc/OAll strategy (1 00). After complete deprotection these glycopeptides were coupled to BSA and KLH (keyhole limpet hemocyanine) to give neoglycoproteins. A new glycosylation method consisting in the electrophile-induced activation of ally1 carbamates was applied to the synthesis of O-glycopeptides (10 1). The formation of glyco- peptides was achieved by a direct glycosylation of pre-formed peptides ( 102).

Lipase-catalyzed cleavage of glycopeptide n-heptyl esters was successfully used in syntheses of multiple glycosylated O-glycopeptides (103). Surpris-

Page 330: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

GLYCOPEFTIDE SYNTHESIS 307

ingly, polar 2-(N-morpholino)ethyl esters of glycopeptides that enhance the water-solubility revealed them to be efficiently hydrolyzed by lipases under neutral conditions (104).

An enzymatic peptide bond formation catalyzed by a gene-technologi- cally produced thiosubtilisin was used for the synthesis of glycopeptide amides (1 05). Oligosacharyl transferases from yeast were applied to form the N-glycosidic bonds between asparagine peptides and oligosaccharides (106,107). The saccharide portions were first transformed to their dolichyl diphosphates. Saccaride chain extension was camed out on glucosamine 0-glycopeptides by means of a galactosyltransferase-catalyzed reaction (1 08).

REFERENCES

1. M. Bergmann andL. Zervas, Chem. Ber., 65 (1932) 1201-1205. 2. D. G. Doherty, E. A. Popenoe, and K. P. Link, J. Am. Chem. Soc., 75 (1953) 3466-3468. 3. P. G. Johansen, R. D. Marshall, and A. Neuberger, Biochem. J., 78 (1961) 518-527. 4. G. S. Marks and A. Neuberger, J. Chem. SOC. (1961) 4872-4879. 5. R. D. Marshall and A. Neuberger, Adv. Curbohydr. Chem. Biochem., 25 (1970) 407-478. 6. H. G. Gargand R. W. Jeanloz, Adv. Curbohydr. Chem. Biochem.. 43 (1985) 135-201. 7. H. Kunz, Angew. Chem. Int. Ed. Engl., 26 (1981) 294-308. 8 . H. Paulsen, Angew. Chem. Int. Ed. Engl., 29 (1990) 823-839. 9. H. Waldmann, Nuchr. Chem. Tech. Lab., 39 (1991) 615-682.

10. H. Kunz and W. Brill, Trends Glycosci. and Glycotech. (Jupun), 4 ( 1992) 7 1 - 82. 1 1. H. Kunz, W. Brill, and K. von dem Bruch, in P. Pr&e (Eds.), Juhrbuch Biotechnologie,

12. S. T. Anisfeld and P. Lansbury, Jr., J. Org. Chem., 55 (1 190) 5560-5562. 13. M. Spinola and R. W. Jeanloz, J. Biol. Chem., 245 (1970) 4158-4162. 14. H. Kunz and H. Waldmann, Angew. Chem. Znt. Ed. Engl., 24 (1985) 883-885. 15. H. Kunz, H. Waldmann, and J. Man, Liebigs Ann. Chem., (1 989) 45 -49. 16. H. Waldmann and H. Kunz, Liebigs Ann. Chem. (1983) 1712- 1725. 17. B. Belleau and G. Malek, J. Am. Chem. Soc., 90 (1968) 165 1 - 1652. 18. SNakabayashi, C. D. Warren,andR. W. Jeanloz, Curbohydr. Res., 174(1988)219-289. 19a. H. Kunz and C. Unverzagt, Angew. Chem. Int. Ed. Engl., 27 (1988) 1697- 1699. 19b. C. Unverzagt and H. Kunz, J. prukt. Chem., 334 (1992) 570-578. 19c. H. Kunz and C. Unverzagt, J. prukt. Chem., 334 (1992) 579-583. 19d. C. Unvenagt and H. Kunz, Bioorg. Med. Chem. 1994, in press. 20. R. U. Lemieux, K. B. Hendriks, R. V. Stick, K. James, J. Am. Chem. Soc., 91 (1975)

21. J. MIrz and H. Kunz, Synlett (1992) 589-590. 22. A. Y. Khorlin, S. E. Zurabyan, and R. G. Macharadze, Curbohydr. Rex, 85 (1980)

23. W. Gunther and H. Kunz, Angew. Chem. Znt. Ed. Engl., 29 (199) 1050- 1051. 24. L. M. Likhoshersov, 0. S. Novikova, V. A. Dervitskaya, and N. K. Kochetkov, Curbo-

25. M. Meldal and K. Bock, Tetrahedron Lett., 31 (1990) 6897-6990. 26. L. Otvos, Jr., L. Urge, M. Hollosi, K. Wroblewski, G. Graczyk, G. D. Frasman, and J.

Vol. 4, pp. 103- 123. Hanser Verlag, Munich, 1992.

4056 -4062.

201-208.

hydr. Res., 146 (1986)Cl-C5.

Thurin, Tetrahedron Lett., 31 (1990) 5889-5892.

Page 331: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

308 HARI G. GARG et al.

27. L. Urge, E. Kollat, M. Hollosi, I. Lackzo, K. Wroblewski, J. Thurin, and L. Otvos, Jr.,

28. H. G. Garg and R. W. Jeanloz, Carbohydr. Res., 32 (1974) 37-46. 29. H. G. Garg and R. W. Jeanloz, J. Org. Chem., 41 (1976) 2480-2484. 30. H. G. Garg and R. W. Jeanloz, Carohydr. Res., 86 (1980) 69-68. 31. M. A. E. Shaban and R. W. Jeanloz, Carbohydr. Res.. 43 (1975) 281 -291. 32. H. Kunz and H. Waldmann, Agnew. Chem. Int. Ed. Engl., 23 (1984) 71 -72. 33. H. Waldmann, J. M h , and H. Kunz, Carbohydr. Res., 196 (1990) 75-93. 34. H. Kunz and C. Unverzagt, Angew. Chem. Int. Ed. Engl., 23 (1984) 436-437. 35. H. Kunz and H. Waldmann, J. Chem. SOC. Chem. Commun. (1985) 638-640. 36. H. Kunz and H. Waldmann, and C. Unvenagt, Int. J. Peptide Protein Res., 26 (1 985)

37. H. Waldmann and H. Kunz, J. Org. Chem., 53 (1988) 4172-4175. 38. T. B. Windholz and D. B. R. Johnston, Tetrahedron Lett (1967) 2555-2559. 39. H. Kunz and J. M&, Synlett (1992) 591-593. 40. H. Kunz, W. Kosch, and J. Man, in J. A. Smith and J. Rivier (Eds.), Peptides: Chemistry

41. J. K. N. Jones, M. B. Perry, B. Shelton, and D. T. Walton, Can. J. Chem. 39 (1961)

42a. V. Bencomo and P. Sinay, Carbohydr. Res., 116 (1983) C9-CI2. 42b. V. Bencomo and P. Shy, Glycoconjugate J., 1 (1984) 5-8. 43. P. Schultheiss-Reimann and H. Kunz, Angew. Chem. Znt. Ed. Engl., 22 (1983) 62-63. 44. H. Kunz and S. Birnbach, Angew. Chem. Int. Ed. Engl., 25 (1986) 360-362. 45. M. Buchholz and H. Kunz, Liebigs Ann. Chem. (1983) 1859- 1885. 45a. H. Kunz and M. Buchholz, Angew. Chem. Int. Ed. Engl., 21 (1981), 894-895. 46. P. Braun, H. Waldmann, and H. Kunz, Synlett (1992) 39-40. 47. B. Ferrari and A. A. Pavia, Int. J. Peptide Protein Rex, 22 (1983) 549-559. 48. J. M. Lacombe and A. A. Pavia, J. Org. Chem., 48 (1983) 2557-2563. 49. M. Meldal and K. J. Jensen, J. Chem. SOC. Chem. Commun. (1990) 483-485. 50. S. Lavielle, N. C. Ling, R. Saltman, and R. Guilleman, Curbohydr. Res., 89 (1981)

51. H. Kunz and R. Barthels, Angew. Chem. Int. Ed. Engl., 22 (1983) 783-784. 52. H. Kunz and S. Birnbach, Tetrahedron Lett.. 25 (1984) 3567-3570. 53. H. Kunz and H. Kauth, Angew. Chem. Int. Ed. Engl., 20 (1981) 895-896. 54. P. Schultheiss-Reimann and H. Kunz, Angew. Chem. Suppl. (1982) 39-46. 5 5 . H. Paulsen, G. Merz, and U. Weichert, Angew. Chem. Int. Ed. Engl., 27 (1988) 1365-

56. B. Luning, T. Norberg, and J. Tejbrant, J. Chem. SOC. Chem. Commun. (1989) 1267-

57. A. Helander, L. Kenne, S. Oscarson, T. Peters, and J.-R. Brisson, Carbohydr. Rex, 230

58. H. Paulsen, M. Schultz, J.-D. Klamann, B. Walter, and M. Paal, Liebigs Ann. Chem.

59. H. Paulsen and M. Paal, Carbohydr. Rex, 135 (1984) 53-69. 60. R. R. Schmidt and J. Michel, Angew. Chem. Int. Ed. Engl., 19 (1980) 731-732; R. R.

61. H. Ijima and T. Ogawa, Carbohydr. Res., 186 (1989) 107-118. 62. H. Kunz and W. Sager, Helv. Chim. Acta, 68 (1985) 283-285. 63a. H. Lijnn, Curbohydr. Rex, 139 (1985) 115- 121. 63b. P. Fugedi and P. J. Garegg, Carbohydr. Res., 149 (1986) C9-CI2.

Tetruhedron Lett., 32 (1991) 3445-3448.

493 -497.

and Biology, pp. 502 - 504. ESCOM, Leiden, The Netherlands, 1992.

1005- 1016.

229 -236.

1367.

1268.

(1992) 299-318.

(1985) 2028-2048.

Schmidt and W. Kinzy, Adv. Curbohydr. Chem. Biochem., 50 (1994).

Page 332: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

GLYCOPEPTIDE SYNTHESIS 309

64. H. Paulsen, W. Rauwald, and U. Weichert, Liebigs Ann. Chem. (1988) 75-86. 65. T. Rosen, I. M. Lico, andD. T. W. Chu,J. Org. Chem., 53 (1986) 1580-1582. 66. N. Nakahara, H. Ijima, S. Siba, and T. Ogawa, Tetrahedron Lett., 31 (1990) 6897-6900. 67. M. Ciommer and H. Kunz, Synlett (1991) 593-595. 68. S. Friedrich-Bochnitschek, H. Waldmann, and H. Kunz, J. Org. Chem., 54 (1 989) 75 1 -

69. H. Paulsen and M. Brenken, Liebigs Ann. Chem. (1988) 649-654. 70. H. G. Garg, T. Hasenkamp, and H. Paulsen, Carbohydr. Res., 151 (1986) 225-232. 71. H. Kunz, P. Wernig, and M. Schultz, Synlett (1990) 631 -632. 72. S. Rio, J.-M. Beau, and J.-C. Jacquinet, Carbohydr. Rex, 219 (1991) 71-90. 73. H. Kunz, S. Birnbach, and P. Wernig, Carbohydr. Res., 202 (1990) 207-223. 74. H. Paulsen and M. Schultz, LiebigAnn. Chem. (1986) 1435-1447. 75. B. Ferrari and A. A. Pavia, Tetrahedron, 41 (1985) 1939- 1944. 76. H. Paulsen, G. Men, and I. Brockhausen, Liebigs Ann. Chem. (1990) 7 19 -739 and 1272. 77. K. Paulsen and K. Aderman, Liebigs. Ann. Chem. (1989) 751 -769. 78. K. Paulsen and K. Adermann, Liebigs Ann. Chem. (1989) 771 -780. 79. H. Kunz, B. Dombo, and W. Koxh, Proc. Europ. Peptide Symp., 20 ( I 988) 154 - 156. 80. H. Kunz and B. Dombo, Angew. Chem. Int. Ed. Engl., 27 (1988) 71 1-713. 81. H. Kunz, W. Kosch, J. M h , M. Ciommer, W. Gtinther, and C. Unverzagt, in R. Epton

(Ed.), Innovations and Perspectives in Solid Phase Synthesis, pp. 17 1 - 178. Intercept, Andover, U. K., 1992.

82. A.M. Janson, M. Meldal, andK. Bock, J. Chem. SOC. Perk. Trans. Z(1992) 1699-1707. 83. S. Peters, T. Bielefeldt, M. Meldal, K. Bock, and H. Paulsen, J. Chem. SOC. Perkin TransZ

84a. I. Laczko, M. Hollosi, L. Urge, K. E. Ugen, D. B. Weiner, H. H. Mautsch, J. Thurin, and

84b. L. Urge, L. Gorbies, and L. Otvos, Jr., Biochem. Biophys. Res. Comm., 184 (1992)

85. H. Waldmann, Nachr. Chem. Tech. Lab., 40 (1992) 828-834. 86. P. Braun, H. Waldmann, W. Vogt, and H. Kunz, Synlett (1990) 105- 107. 87. M. Schultz, P. Hermann, and H. Kunz, Synlett (1992) 37-38. 88. E. J. Toone, E. S. Simon, M. D. Bednarski, and G. M. Whitesides, Tetrahedron, 47 (1989)

89. S. Sabesan, and J. C. Paulson, J. Am. Chem. SOC., 108 (1986) 2068-2080. 90. C. Unverzagt, H. Kunz, and J. C. Paulson, J. Am. Chem. Soc., 112 (1990) 9308-9309. 91. C. Augk, C. Gautheron, and H. Pora, Carbohydr. Res., 193 (1989) 288-293. 92. J. Thiem and T. Wiemann, Angew. Chem. Znt. Ed. Engl., 29 (1990) 80-82. 93. M. Schultz and H. Kunz, Tetrahedron Lett., 33 (1992) 5319-5322. 94. S.T.Cohen-AnisfeldandP.T.LansburyJr.,J.Am. Chem. SOC. I15(1993) 10531-10537. 95. S. Peters, T. Bielefeldt, M. Meldal, K. Bock, and H. Paulsen, Tetrahedron Lett. 33 (1992)

96. M. Meldal, S. Mountsen, and K. Bock, Am. Chem. SOC., Symp. ser. (1993) pp. 19-33. 97. M. Christensen, M. Meldal, and K. Bock, J. Chem. SOC. Perkin Trans I (1993) 1453-

98. I. Christiansen-Brams, M. Meldal, and K. Bock, J. Chem. SOC. Perkin Trans I (1993)

99. K. B. Reimer, M. Meldal, S. Kusumoto, K. Fukase and K. Bock, J. Chem. SOC. Perkin

756.

(1992) 1163- 1171.

L. Otvos, Jr., Biochemistry, 21 (1990) 4282-4288.

1125- 1132.

5365-5422.

4126-4135.

1460.

146 1 - 147 I.

Trans l(1993) 925-931. 100. K. von dem Bruch and H. Kunz, Angew. Chem. Int. Ed. Engl. 33 (1994) 101 - 103. 101. H. Kunz and J. Zimmer, Tetrahedron Lett. 34 (1993) 2907-2910.

Page 333: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

310 HARI G. GARG et al.

102. C. D. Warren and H. G. Garg, Glycobiology 3 (1993) 540. 103. P. Braun, H. Waldmann, and H. Kunz, Bioorg. Med. Chem. 1 (1993) 197-207. 104. G. Braum, P. Braun, D. Kowalczyk, and H. Kunz, Tetrahedron Lett. 34 (1993) 31 11 -

105. C.-H. Won& M. Schuster, P. Wan& and P. Sears, J. Am. Chem. Soc. 115 (1993) 5893-

106, J. Lee and J. K. Coward, J. Org. Chem. 57 (2992) 4126-4135. 107. J. Lee and J. K. Coward, Biochem. 32 (1993) 67694-6801. 108. M. Schultz and H. Kunz, Tetrahedron Asymmetry 4 (1993) 1205- 1220.

31 14.

5901.

Page 334: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

ADVANCES IN CARBOHYDRATE CHEMISTRY AND BIOCHEMISTRY, VOL. 50

PHYSICOCHEMICAL ANALYSES OF OLIGOSACCHARIDE DETERMINANTS OF GLYCOPROTEINS

BY ELIZABETH F. HOUNSELL

MRC Glycoprotein Structure/Function Group, Department of Biochemistry and Molecular Biology, University College London, London, WCl E 6BT, England

I. INTRODUCTION

A large number of oligosaccharide structures of glycoproteins have now been unequivocally characterized by using a combination of chromatogra- phy, mass spectrometry (m.s.), and nuclear magnetic resonance (n.m.r.) spectroscopy. Information from these physicochemical techniques, together with that obtained from circular dichroism and X-ray diffraction of mono- and polysaccharides [ previously discussed in this series ( 1 ,2)], is providing data for incorporation into computer-graphics molecular models of oligo- saccharides and glycopeptides in order to visualise oligosaccharide se- quences important in molecular recognition. This chapter surveys the diver- sity of structures and shapes of oligosaccharide determinants of glycoproteins that are antigens and targets for binding of adhesion mole- cules.

11. METHODS OF STRUCTURAL ANALYSIS

Four major techniques (summarized in Table I) provide the basis of our current structural-analysis stratagem for oligosaccharides of glycoconju- gates. Historically, the first of these was methylation analysis. The desire to delineate the linkages through which monosaccharides are coupled led to the idea of labeling the free hydroxyl groups by permethylation. The low molec- ular weight of the methyl group is an important factor in subsequent mass- spectroscopic analysis, and the hydrophobicity imparted to the oligosaccha- ride permits efficient purification, increased chromatographic resolution, and good m.s. ionization properties. The method of permethylation using dimethylsulfinyl anion introduced by Hakomori (43) for complete derivati- zation of quite large oligosaccharides became universally adopted (6 - 1 I). A

Copyright 8 1994 by Academic Press, Inc. All rights of reproduction in any form reserved. 31 1

Page 335: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

312 ELIZABETH F. HOUNSELL

TABLE I A General Stratagem for Oligosaccharide Physicochemical Analysis

Method applied Sequence of applied techniques

Methylation analysis

Chromatographic analysis

F.a.bm.s. or 1.s.i.-m.s

N.m.r. spectroscopy

Permethylation of intact oligosaccharides Hydrolysis of glycosidic linkages Reduction to monosaccharide alditols Acetylation of free. hydroxyl groups previously involved in link-

G.1.c. separation of substituent, partially methylated alditol ace-

On-line e.i.- or c.i.-m.s. analysis G.1.c. analysis after sequential hydrolysis, reduction, and acetyla-

G.1.c. analysis after methanolysis and trimethylsilylation H.p.a.e. chromatography after hydrolysis H.p.1.c. -h.p.a.e. chromatographic profiling of reducing oligo-

Native oligosaccharides placed on the m.s. probe-tip with a ma-

Bombardment with atoms (f.a.b.-m.s.) or ions (1.s.i.m.s.) MA. analysis in negative-ion mode Permethylated or acetylated oligosaccharides analyzed in posi-

tive-ion mode Reducing oligosaccharides or periodate-oxidized alditols deriva-

tized by reductive amination and analyzed in positive-ion mode

T.1.c.-m.s. analysis of oligosaccharides coupled to a lipid amine (neogl ycolipids)

IH n.m.r. spectrum in D20 after exchange of free protons with deuterium

Experiments conducted at 295 K, with acetone as the internal standard (set at 2.225 p.p.m. from 4,4-dimethyl-4-silapentane- 1-sulfonate)

Results compared, to within +0.005 p.p.m. (laboratory-to-la- boratory variation) of data in the literature

Conformational studies by n.0.e. experiments Nat~ral-abundance-l~C analysis Chemical-shift assignment by 2D IH-IH and lH-l3C n.m.r.

ages

tates

tion

saccharides, alditols, or organic amine derivatives

trix

spectroscopy

Note. Abbreviations: g.l.c., gas-liquid chromatography; e.i.-m.s., electron-impact mass spectrometry; c.i.-m.s., chemical-ionization mass spectrometry; h.p.l.c., high-performance liquid chromatography; h.p.a.e., high pH anionexchange; f.a.b.-m.s., fast-atom-bombardment mass spectrometry; 1.s.i.-m.s., liquid second- ary-ion mass spectrometry; n.O.e., nuclear Overhauser enhancement. Details of these methodologies are given in Ref. (3).

Page 336: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

OLIGOSACCHARIDE DETERMINANTS OF GLYCOPROTEINS 313

more facile method (1 2) involving the use of sodium hydroxide in dimethyl sulfoxide, based on the original Kuhn (1 3) procedure, is now largely used. The combination of hydrolysis, reduction, acetylation, and g.1.c. (now capil- lary g.1.c.) remains the best method for analysis of the large number of closely related, partially methylated monosaccharide derivatives formed, although separation by high-performance liquid chromatography (h.p.1.c.) has also been investigated ( 14). Electron-impact mass spectrometry (e.i.-m.s.) is the most generally applied robust technique for detection and characterization, although chemical-ionization (c.i.)-ms., in particular, of trifluoroacetylated, methylated alditols gives increased sensitivity and a greater abundance of the molecular ion. Gas-liquid chromatography (g.1.c.)-m.s. analysis of intact permethylated oligosaccharides ( 15) can also be achieved.

As is general in biochemistry, improvements in chromatographic tech- niques have been a major factor in research advances. This is particularly true in the carbohydrate field, in which separation of the large number of molecules that are structurally closely related is a major challenge. Thin- layer chromatography (t.1.c.) and paper, gas-liquid, high-performance liq- uid, and supercritical fluid ( 16) chromatographies, together with electropho- resis (1 7,18), have their application in oligosaccharide analysis and purification.

The introduction of g.1.c. for oligosaccharide analysis constituted a major breakthrough in the field. In addition to strategies for accurate and sensitive quantitation of monosaccharide type (19,20), chiral procedures may be adopted for enantiomeric (D and L) determination (21). The sensitivity of h.p.a.e. chromatography with pulsed amperometric detection now provides an alternative to g.1.c. for oligosaccharide compositional analysis (22).

The use of m.s. in oligosaccharide analysis was advanced greatly by the advent of high-field magnets and the technique of f.a.b. -m.s. (23,24). This enabled analysis of larger oligosaccharides in their intact state, allowing for sequence determination at the nanomolar level. Analysis of native oligosac- charides in this way, and by the similar technique of 1.s.i. -m.s., gives data on acyl substitution of hydroxyl groups, substituents which may be lost during derivatization procedures. Derivatization (for example, acetylation, methylation, and reductive amination) gives further sensitivity and se- quence information (23 - 26). F.a.b.-m.s. of permethylated periodate-oxi- dized (27) oligosaccharides and t.1.c.-1.s.i.-m.s. of periodate-oxidized alditols coupled to lipidamines (28) provide additional strategies for gaining se- quence and linkage information. Coupling to lipidamines (neoglycolipids) allows for in situ analysis of oligosaccharides on t.1.c. by m.s. and overlay with carbohydrate binding proteins in studies of receptor and antibody specificities, as introduced by Feizi and colleagues (29).

Page 337: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

3 14 ELIZABETH F. HOUNSELL

The application of proton ('H) n.m.r. spectroscopy to oligosaccharide characterization has rapidly increased since the early days when low-field instruments were used in the characterization of synthetic products and first applied to complex oligosaccharides of glycoproteins (30 - 32); high-field n.m.r. is now a relatively routine tool for providing a first-line profile of a purified oligosaccharide (32,33). The application has been diversified into providing conformational information on oligosaccharides and glycopep- tides in conjunction with 2D methods. Now available are large databanks of chemical shifts that can be readily searched to give structural intrepretation (32 - 38). The purpose of the present article is not to supply a large volume of data, but rather to categorize the structures found in mammalian glycopro- teins by using 'H n.m.r. spectroscopy and mass spectrometry and to discuss the conformational information obtained.

111. PURIFICATION AND PROFILING

In addition to studies uniquely on oligosaccharide structure, the carbohy- drate moiety of glycoproteins is now being viewed in the context of the protein skeleton, in order both to characterize the complete glycoprotein molecule and to elucidate the biosynthetic controls that determine site-spe- cific glycosylation patterns. The modern methods of purification and profi- ling reflect this philosophy. For example, commercial kits are available for the sensitive detection of glycosylated proteins; glycoprotein-specific degra- dative enzymes are being purified and cloned; h.p.l.c., n.m.r. spectroscopy, and circular dichroism (c.d.) are being used for glycopeptide analysis; and glycoproteins produced by recombinant DNA technology are increasingly being screened for variations in glycosylation patterns (39 -42).

Profiling methods originally involved the analysis of N-linked glycosyla- tion by hydrazinolysis, followed by size-exclusion on columns of Biogel P4 before and after specific cleavage by exoglycosidases (43). Release of oligo- saccharide chains by endoglycosidases (44) and h.p.1.c. analysis (reviewed in Ref. 45) have now been added to the stratagem (see Scheme 1). Both chro- matography on Biogel P4 and h.p.1.c. have been perfected for purification of oligosaccharides prior to n.m.r.-spectral analysis and biological studies. Here, the h.p.a.e. technique is not generally applicable, owing to the presence of a high salt content and the lower loading and yield compared with those encountered on h.p.1.c. and Biogel P4 chromatography in volatile buffers. Initial separation by size exclusion is advantageous, because retention be- havior in h.p.1.c. is affected by both composition and linkage, and, therefore, it is much easier to separate each size subset first and then to purify isomers by h.p.1.c. Reverse phase using octadecylsilyl (0.d.s.) or porous graphitized carbon (p.g.c.) columns, normal phase using amine-bonded silica (for ex- ample, a.p.s.), and anion-exchange (a.e.) chromatographies in volatile

Page 338: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

OLIGOSACCHARIDE DETERMINANTS OF GLYCOPROTEINS 315

Glywprotein

1 Protiase Mild hydrolysis O(carboxymethy1)ation digestion with acid for and protease digestion

sialic acid 4 determination 1.s.i.rn.s. 4

h.p.1.c.

t GI ywpeptides Peptides

1.s.i.m.s. Endo H. peptide or N-terminal N-glycanase F and

analysis mild alkali treatment

t

+ 1-e Mild a ~ t a l ~ d ~ o ~

I Peptides 1 Oligosaccharides

~

Reductive amination and h.p.a.e. chromatography Methylation

e-7 h.p.1.c. ort.1.c.-m.s. (of neoglycolipids)

1.s.i.rn.s. Methylation analysis

SCHEME 1 . -A Flow Chart Outlining the Methodologies for Profiling Protein Glycosylation Patterns.

buffers-organic phase are very reliable and efficient methods (reviewed in Ref. 46).

It is important to integrate more than one method of purification and analysis for the unambiguous characterization of mammalian glycoconju- gates. Detailed studies of glycoproteins frequently reveal new structures, in either monosaccharide type, acyl substitution, or linkage mode. In addition, the variation in glycosylation pattern from proteins translated in different cell types can be quite subtle and may be readily missed on bulk analysis. In order to alert the molecular biologist to the wide diversity of posttransla- tional changes available in the repertoire employed by nature, it is therefore necessary to resort to detailed studies, ideally using a combination of m.s. and n.m.r. spectroscopy and using high-resolution chromatographic tech- niques.

IV. BACKBONES AND CORE REGIONS OF N- AND 0-LINKED CHAINS OF

In a series of elegant studies in the 1980s, the groups of Montreuil(30), Kobata (47), and Vliegenthart (3 1,32) began to explore the structural diver-

SECRETED A N D PLASMA MEMBRANE GLYCOPROTEINS

Page 339: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

316 ELIZABETH F. HOUNSELL

sity of N-linked glycosylation of serum glycoproteins. This followed the extensive work of Morgan (48), Watkins (49), and &bat (50) in characteriz- ing the blood-group antigens and mucin structure. It became obvious that, in many instances, oligosaccharide sequences forming the periphery and back- bone could be present on different protein and lipid core-regions, as de- scribed in reviews by Feizi ( 5 1) and Hakomori (52). From their studies, the concept of oligosaccharides as oncodevelopmental antigens began to be appreciated, that is, the concept that oligosaccharide structures change dur- ing development and may be reexpressed as tumor antigens in the adult. These antigens are found on glycolipids, on 0-linked chains of mucins, and on the 0- and N-linked chains of serum and cell-membrane glycoproteins.

Mucins have provided a large-scale source of oligosaccharide sequences, important not only as oncodevelopmental antigens, but also as representa- tive structures present in the peripheral and backbone regions (53) of N- linked chains of serum glycoproteins, which are generally of more restricted availability. Several series of detailed studies of the multiple oligosaccharide chains of these species having very high molecular weight have been initiated for the foregoing reasons and also because of the desire to correlate their oligosaccharide structure with (i) the known physicochemical properties important in maintaining an effective viscoelastic bamer, (ii) their role as a bacterial interface, (iii) regulation of biosynthesis, and (iv) functional molec- ular recognition.

1. Mucin Oligosaccharide Structure

The majority of the initial studies of mucin structure were carried out on animal gastrointestinal mucins and ovarian-cyst blood-group substances of humans. These gave a good grounding to the diversity to be expected, as described in two reviews (5334). High-field 'H-n.m.r. (270-600 MHz) and m.s. studies then began to dissect this diversity. Oligosaccharides from mucins of ovarian-cyst fluids (55 - 58) were among the first to be investigated by these new techniques, following earlier work on this material using chem- ical and immunochemical methods (50). A map of ovarian mucin structure (shown opposite) from these early studies (59) stands today as a tribute to the elegance of the early techniques and the accuracy of the understanding of oligosaccharide structure, variation, and antigenicity.

In the 1980s, several structural studies of human mucin were initiated, their overall aim being to describe the oligosaccharide sequences of bron- chial mucins (60-68), adult gastric (69,70) and colonic (7 1,72) mucins, and fetal gastrointestinal mucins (73 - 76). The first study sought to understand the changes in oligosaccharide structure in bronchial mucins important in cystic fibrosis and bronchiectesis, in addition to the role of bacterial growth in defining much status. The second set of studies set out to establish oligosaccharide patterns in the normal and neoplastic adult mucosae. The

Page 340: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

OLIGOSACCHARIDE DETERMINANTS OF GLYCOPROTEINS 317

New gene interaction product [now known as Y or Ley]

A or B gene products A h

H gene New gene [now known as X, SSEA-I or L e x ]

a-L-FUC cu - L - F u c D-D-Gal-( 1 + 4)-p-~GlcNAc 1 1 1 .1 1 1

a-DGalNAc-( I - 3)- 2 4 6

or a-D-Gal-( 1 - 3)- 1 3 1 t 6 1

3 t

a-DGalNAc-( 1 + 3)- 1

or a-D-Gal-( 1 + 3)- 2 3 t t 1 I

CU-L-FUC a-L-FUC - - H gene Leagene -

Leb gene interaction product

&D-GaI-( I - ~)-P-D-GIcNAc a-DGalNAc

b-D-Gal-( 1 + 3)-/%~-GlcNAc-( 1 - 3)-p-~-Gal

p-D-Gal-( 1 -, ~)-/%D-GIcNAc

last study was begun in order to identify novel oligosaccharide sequences present in the bacteria-free environment of the fetal gut that constitute mucin oligosaccharides synthesized in early development and that might reoccur in the adult as tumor antigens (oncodevelopmental antigens). From the extensive information on mucin structure obtained from these studies, and several additional studies on adult mucins (77 - 8 I), it is pertinent to point out the differences found. Table I1 displays the various backbone and core regions. Although it cannot be concluded that structures found in one study are not present as minor components in another, the relative amounts detected in these systematic studies demonstrated differences in each of the particular biological scenarios. The results of these studies underscore sev- eral salient points, as follows. (i) Specific carbohydrate tumor-antigens are unlikely to occur (5 I), and, therefore, quantitative differences will have to be exploited for any future tumor diagnosis or therapy. (ii) Biosynthetic control is often reflected in the amount of an enzyme produced rather than its total absence from the gene repertoire for a particular cell. (iii) Homogeneous cell-population studies will be required in order to dissect further the diver- sity of oligosaccharide patterns. (iv) Outside factors will affect endogenous structure, for example, the influence of bacterial degradative enzymes.

One additional aspect of studies on the core and backbone variation in

Page 341: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

W

00 e

TABLE I1 The Backbone and Core Sequences Found in H u m Mucins.

Yield from Core type” meconiume (%)

CY-G~~NAC 13

I 4 &Gal( 1 -D 3)a-GalNAc I 10

[B-Gal-( 1 -D 3/4)wlcNAc-( 1 -D 3)-],,, ,,-&Gal( 1 + 3)e-GalNAc

B-Gal-( 1 -D 4)-p-GlcNAc

i -

6

3

1

B-Gal-( 1 + 4)-wlcNAc-( 1 -D 3)-wal( 1 -D 3)a-GalNA~ I

T &Gal-( 1 + 4)-wlcNAc Adult gastric mucins only

&Gal-( 1 + 4)-&GlcNAc ! .1 6

3 &Gal-( 1 -+ 3)e-GalNAc Ib

t 1

&Gal-( 1 4 4)-&GlcNAc &Gal-( 1 -D 4)-&GlcNAc

1 1 6

t

wf-( 1 + 3)a-GalNA~ Ib

0.5

1

Ovarian cyst only /%Gal-( I + 3)-fl-GlcNAc

Page 342: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

2 /?-Gal-( 1 4 4)-&GlcNAc

! 6

a-GalNAc 3 t 1

&Gal W-Gal-( 1 4 3/4)-&GlcNAc-( 1 - 3)-]Gal-( 1 + 4)-&GlcNAc

i 6

a-GalNAc 3

1 t

W-Gal-( 1 4 3/4)-b-GlcNA0( 1 + 3)]-/?-Gal [Gal-( 1 4 4)-/?-GlcNAc-( 1 4 6)],-b-Gal-( 1 4 4)-&GlcNAc

L Human skimmed milk only 6

a-GalNAc 3

1 t

&Gal P-Gal-( 1 4 4)-&GlcNAc

1

23

1 6

a-GalNAc

s i

6)-/?-Gal 3 T 1

Gal-( 1 + 4)-/?-GlcNAc-( 1

_+ F a l l - ( 1 4 4)-&GlcNAc] Linear GlcNAc 1-6 sequence in meconium only

I1

I1

IIb

1

2 (3 : 1 ratio of branch

extension on the lower branch)

Continued

Page 343: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

TABLE I1 (continued)

W h) 0

Yield from Core type, meconium' (TO)

/3-Gal-( 1 4 4)+GlcNAc

i 6

&Gal-( 1 + 4)-&GlcNAc 1

- I1 3

1 T

/3-G:-( 1 4 4)-/3-GlcNAc-( 1 + 3)-&Gal

t 1

pGal-( I 4 4)-WlcNAc Adult gastric mucin only

&GlCNAC-( 1 + 3M-GalNAc 111

I11 &Gal-( 1 4 3/4)-/3-GlCNAc-( 1 4 3)-/%Gal-( 1 4 3)-&GkNAo( 1 + 3)e-GalNAc Human colonic much is reported to have a &Gal-( 1 + 3)-j?-GlcNAc sequence linked at 0-6 of internal Gal, giving a branched structure

Wal-( 1 -+ 4)-WlcNAc

i 6

3

1 &GlCNAC

a-GalNAc IV

t

Bronchial mucins only

10

0.2

Page 344: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

B-Gal-( 1 + ~)-B-GICNAC

i ? 6

a-GalNAc

1 /%Gal-( 1 - 3/4)-/?-GlcNAc

cu-GalNA~-( 1 -+ 3)-c~-GalNAc

wid-( 1 - 4)-wlcNAc-( 1 + 6W-GalNAc Meconium and Smith degraded ovarian cyst only

a-GalNAc-( 1 + 6)-~t-GalNAc

Identified in bovine submaxillary mucin only, not in human &Gal-( -* 4)-PGlcNAc-( 1 -+ 3)-,!?-Gal

1

GlcNAc !

5-

2 6

GalNAc

IV

V

VI

VII

7 (2:1ratioof1-+4

and 1 + 3 on lower branch)

4

1

4 I 6 1

/%Gal-( 1 -* 4)-&GlcNAc-( 1 -, 3)-&Gal

? 1 m-( 1 + 4)-/?-GlcNAc

Also, related structures reported in adult gastric much

a From references (60-8 1); a-GalNAc-(l+ 6)-GalNAc is included for completeness, although so far this has been found only in bovine submaxillary mucin (82). All monosaccharides have the ~-~~uligurat ion; when the structure. has been identified in only one study, the source is identified.

b Cores have been named historically (53,83), but appear in the following semilogical order Gal at 0-3 in the absence or presence ofGlcNAc at 0-6; GlcNAc at 0-3 in the absence or presence ofGlcNAc at 0-6; W A C at 0-3; GlcNAc at 0-6; GalNAc at 0-6; Gal at 0-6 in the presence of Gal at 0-3.

c The percentage yields for the various oligosacchatides found in the systematic study of the backbone and core moieties of fetal, gastrointestinal-tract mucins found in blocd-group H-active, meconium glycoproteins depleted of Ii antigenic activity (5% of the hexose retained on an anti-I immunoaflinity column) (73 - 75) are given in parenthem. The remaining 2 1% oligosaccharides not accounted for are a heterogeneous mixture larger than heptasaccharide..

Page 345: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

322 ELIZABETH F. HOUNSELL

mucins is our ignorance of which factors in the protein skeleton that are distant from the glycosylation site influence biosynthesis and whether the diversity of the carbohydrate structures of mucins will also be found in the 0-linked chains of serum and cell-membrane glycoproteins. Originally, it was thought that 0-linked chains of these nonmucin and, by comparison, relatively low-molecular-weight glycoproteins were only of the following class:

f a-D-NeuAc 2

6

3

1 a-~-GalNAc-( 1 - )Ser/Thr

1 f a-~-NeuAc-(2 - ~ ) - D - D - G ~

Additional sequences have now been found, for example, those shown in Table 111. Early studies (87) of the T and B cell-membrane antigen T200 (now called CD45) also suggested the presence of 0-linked oligosaccharides larger than tetrasaccharides and provided evidence that cell-membrane gly- coproteins can have clustered 0-glycosylation sites similar to those found in mucins. This has now been shown for a series of serum and cell glycopro- teins, discussed in Section VII below, where multiple 0-linked chains affect protein conformation and function. The interaction of oligosaccharides with the protein backbone can lead to joint protein-carbohydrate epitopes, and therefore at a conformational level it is important to analyze glycopeptides rather than only the released oligosaccharides. However, in order to study the diversity of oligosaccharide sequences, the 0-linked, much-type chains of glycoproteins have in general been analyzed after release from protein by treatment with mild alkali - borohydride according to the conditions origi- nally described by Carlson (88).

2. ‘H-n.m.r. - Spectral and Mass-Spectrometric Analysis of Mucin Oligosaccharide Chains

The oligosaccharide-alditols released from mucins have 2-acetamido-2- deoxy-D-galactit01 (GalNAc-01) at the reduced end, which may readily be detected by H-n.m.r. spectroscopy from the characteristic, multiplet pat- tern for protons attached at C-2 and C-5 in the regions 4.40-4.24 and 4.28-3.75 ppm, respectively. The chemical shifts for these multiplets and for protons at G I , C-3, C-4, and C-6 are primarily dependent on the differ- ent substitution patterns at C-3 and C-6 as described in Table IV. However, certain substituents on the residues linked to GalNAc-ol can significantly (greater than k 0.03 ppm) affect the chemical shifts of GalNAc-01, in partic- ular, the presence of CX-L-FUC at 0 - 3 or 0-4 of p-D-GlcNAc linked to Gal-

Page 346: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

OLIGOSACCHARIDE DETERMINANTS OF GLYCOPROTEINS 323

NAc-ol and CX-L-FUC-( 1 + 2), CX-D-G~CNAC-( 1 - 4), or a-D-GalNAc-( 1 - 3) linked to the p-~-Gal that is linked to GalNAc-o 1. Thus, in the database used for computer-assisted interpretation of H-n.m.r. spectra (33), the chemical shifts of the “core regions” for mucin-type alditols shown in Table IV have been defined to a tolerance of f0 .03 ppm. Unlike the situation obtaining with 13C-n.m.r. spectroscopy, no empirical rules can be deduced for predicting the effect of substitution of a hydroxyl group at a particular carbon atom on the chemical shift of the attached proton (except for sulfa- tion, as discussed in Section Vl). Therefore, each type of linkage has to be specified in the databases used for structural interpretation.

For novel oligosaccharides for which ‘H-n.m.r. data have not previously been documented in the literature, it is necessary to supplement the ‘H- n.m.r. data with composition, sequence, and linkage information provided by chromatographic and m.s. analyses. An illustrative example is the char- acterization of the following oligosaccharide of the glycopeptides of fetal gastrointestinal mucins of meconium (74):

p-~-Gal-( 1 4 4)-/3-~-GlcNAc 1 1 6

G ~ ~ N A C - O ~ 3

1 t

p-D-Gd-( 1 4 ~)-~-D-G~cNAc-( 1 -.* 6)-p-D-Gd

There were significant differences in the chemical shifts of the GalNAc-ol protons in this oligosaccharide compared with the tetrasaccharide lacking the p-~-Gal-( 1 + 4)-p-~-GlcNAc-( 1 + 6) sequence on the lower branch (see Table V) and oligosaccharides having a P-D-Gal-( 1 - 4)-p-~-GlcNAc- (1 - 3)-p-~-Gal sequence linked at 0-3 of GalNAc-ol in the presence or absence of a p-D-Gal-( 1 + 4)-p-~-GlcNAc-( 1 + 6) branch (see Table IV). In addition, the signals for protons at C-5 and C-6 ofp-D-Gal residues could not be readily distinguished. Thus, from ‘H n.m.r. alone, it was difficult to determine the structure of this oligosaccharide, and m.s. studies were neces- sary in order to permit the interpretation that the p-D-Gal residue linked (1 - 3) to GalNAc-ol was itself linked at 0-6. Methylation analysis of this oligosaccharide showed the presence of terminal Gal, GlcNAc linked at 0-4, Gal linked at 0-6, and GalNAc-ol linked at 0-6 and 0-3. Sequence infor- mation from 1.s.i.-ms. of permethylated oligosaccharides showed abundant fragment-ions of m/z 464 and 432, characteristic of terminal Gal-( 1 + 4)- GlcNAc sequences. Analysis of PPEADP-derivatives formed after periodate oxidation of the C-4-C-5 bond and reductive amination (28) showed the distribution of tri- and disaccharides at 0-3 and 0-6, respectively, of Gal- NAc-01. Together, these data gave an unambiguous assignment for the oli- gosaccharide structure. Thus, it was established that P-D-G~CNAC-( 1 - 6)-

Page 347: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

324 ELIZABETH F. HOUNSELL

TABLE 111

0-Linked Oligosaccharide Chains of Serum and Cell-Membrane Glycoproteins

Glycoprotein type Oligosaccharide structurfl ~

Erythrocyte glycophorin (84)

Human chorionic gonad- otropin (85)

f [a-NeuAc-(2 - 8)I-a-NeuAc 2

6 .1

a-GalNAc 3 t i

f [a-NeuAc-(2 + S)]-cy-NeuAc-(2 + 3)-B-Gal a-Neu Ac

2

6 a-GalNAc

3

1 &GalNAc-( 1 + 4)-PGal

3

2 a-NeuAc

1

t

t

f [a-NeuAc-(2 + 3)-/?-Gal-( 1 + 4)-/.-G 1 cNAc]

f 6

a-GalNAc 3 t i

a-NeuAc-(2 -+ 3)-P-Gal Immunoglobulin A (86) a-Fuc-( 1 - 2)-&Gal-( 1 - 3)+-GlcNAc-( 1 + 3)-P-Gal-( 1 -+ 4)-GlcNAc

1 1 6

a- Gal N A c 3 t i

a-NeuAc-(2 - 3)-/3-Gal Also, related sequences of the mucin type

a All of the monosaccharides shown have the D configuration except for LFUC.

P - D - G ~ can occur as a linear sequence in the absence of&D-GlcNAc linked at 0-3 ofp-D-Gal [this has since also been established for oligosaccharides of human-milk mucins (8 l)]. Similarly, it was shown by characterization ofthe trisaccharide P-D-Gal-( 1 - 4)-p-~-GlcNAc-( 1 + 6)-GalNAc-ol in the mucins of fetal gastrointestinal tract (meconium) (73,89) that the (1 + 6) linkage can occur in the absence of a (1 - 3) linkage in the core of mucin- type oligosaccharides. This had previously been found in Smith-degraded

Page 348: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

OLIGOSACCHARIDE DETERMINANTS OF GLYCOPROTEINS 325

mucins of human ovarian cyst (57), but is more likely in that case to be formed during the degradation procedure than by specific biosynthesis. In either event, characterization of these novel sequences has increased the diversity of the mucin-oligosaccharide structure now expected and it under- scores the necessity for stringent physiochemical parameters for structural studies.

An early pointer to the potential diversity of mucin glycosylation was the identification of core regions in addition to P-D-Gal-( 1 + 3)-a-~-GalNAc and P-D-G~cNAc-( 1 + 6)-v-~-Gal-( 1 + 3)]-a-~-GalNAc found in the early classical studies. The first “new” core region was the trihexosamine core P-D-G~cNAc-( 1 + 6)-u-~-GlcNAc( 1 + 3)]-a-~-GalNAc found ini- tially in sheep gastric mucins (90) and later substantiated by n.m.r. spectros- copy as present in human bronchial mucins (61), ovarian cyst fluid (57), porcine gastric mucins (9 l), and fetal gastrointestinal mucins (75,76). Addi- tional core regions, having linear P-D-G~cNAc-( 1 -+ 3)-a-~-GalNAc, /3-D- GlcNAc-( 1 + 6)-a-~-GalNAc, a-D-GalNAc-( 1 + 3)-a-~-GalNAc, and a- D-GalNAc-( 1 + 6)-a-~-GalNAc, have also been found [e.g., Refs. (73,82); Table 111. The substantiation of the core (Table 11) p-~-Gal-( 1 + 6)-@-~- Gal-( 1 + 3)]-a-~-GalNAc awaits relevant n.m.r. data, and the other core that completes this series, namely, linearj?-D-Gal-( 1 + 6)-a-~-GalNAc, has not been identified.

V. PERIPHERAL SUBSTITUTIONS OF N- AND 0-LINKED CHAINS OF GLYCOPROTEINS

Considerable structural information and n.m.r. data are available on the core and backbone regions of N- and 0-linked chains of glycoproteins and proteoglycans as already described for mucins and well known for N-linked chains (30-32,39-47) and proteoglycans (92). The backbone regions of oligosaccharide chains of glycoproteins and proteoglycans consist of the following disaccharide units: p-~-Gal-( 1 + 3/4)-p-~-GlcNAc or a-D-Man- (1 - 2/6)-a-~-Man in glycoproteins, /~-D-G~cNAc-( 1 + 4)-p-~-GlcA- (1 + 3) in hyaluronate, p-~-GalNAc-( 1 + 4)-p-~-GlcA-( 1 + 3) in chon- droitin sulfate (CS), and a-~-GlcNS0,-( 1 -+ 4)-p-~-GlcA-( 1 - 4) in hepa- ran sulfate (HS). The P-D-G~cA residues of CS and HS can then be epimerized to a-L-IdoA (CS is then called dermatan sulfate and HS, hepa- rin). The chains are further modified by 0-sulfation and, for heparin/he- paran sulfate, N-deacetylation followed by N-sulfation.

Peripheral to the Gal-GlcNAc sequences of glycoproteins, there can occur an array of blood-group-related glycosylations, anionic substitutions (sialy- lation, 0-sulfation, and 0-phosphorylation), or both. Chains of the high- mannose type may also bear phosphate groups. Table VI reviews the pat-

Page 349: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

TABLE IV Averaged ’H n.m.r. Chemical Shifts‘ from Data in the Literature for 0-Linked

Chain-Core Region Substitutions of GalNAc-ol’

GalNAc4 chemical shifts

Sequence H-1 H-1’ H-2 H 3 H-4 H-5 H d Hd’ NAc

/3-Gal-( 1 + 3)-GalNAGOl 3.74 3.61 4.39 a-GalNAc

3

2

1

/?-GlcNAc

6

3 t

f - - 4.30 /?-Gal-( 1 + 3)-GalNAc-01

t a-FUC

f - - 4.39 B-Gal-( 1 + 3)GalNAc-ol

4.06

4.10

3.99

3.41 4.16 3.69 3.65 2.050

- 2.050 3.60 4.13 -

3.51 4.14 - - 2.050

i B-GlcNAc a-NeuAc

2

6

3

1

1

T B-Gd

G al N A c -01 3.86 3.15 4.38 4.06 3.53 4.20 3.85 3.41 2.044

Page 350: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

B-GlcNAc 1

w t:

.1

t

6

3 GalNA001 - - 4.39 4.06 3.46 4.28 3.93 - 2.067

i /%GlCNAC-( 1 - 4)-Gal

WlcNAc

f 6

3

1

GalNAc-01 -

t (Y-Fu~-( 1 - 2)-Wal WlcNAo( 1 - 3)-GalNAc-ol - WlcNAc

t .1 6

GalNAc-01

; i

WlcNAc (Y-G~NAG( 1 L-, 3)-GalNAool 3.8 1 B-CICNAC-( 1 6)-GalNAc-01 3.73 a-NeuAc-(2 - 6)-GalNAc-ol 3.73

-

-

-

-

3.72 3.67 3.66

4.40

4.40

4.28

4.28

4.39 4.25 4.24

4.08

4.08

4.00

3.99

3.89 3.84 3.84

3.57

3.49

3.56

3.51

3.68 3.38 3.41

4.26 3.95

4.25 -

4.13 3.65

4.23 3.91

3.75 3.65 4.03 3.93 - 3.84

-

-

-

-

3.67 3.71 3.53

2.054

2.054

2.034

2.044

2.049 2.044 2.056

0 Given to f0.03 p.p.m. (or f0.005 p.p.m. for NAc) from 4-4-dimethyl4silapentane-l-sulfonate at 295 K * The data are those used in a computer program for spectral interpretation [as described in Refs. (33) and (34)].

Page 351: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

328 ELIZABETH F. HOUNSELL

TABLE V Comparison of the 'H N.m.r. Chemical Shifts (p.p.m. from 4,4-Dimethyl-4-

silapentane-1-sulfonate at 295 K) of a Tetrasaccharide and Related Hexasaccharide from Meconium Glycoproteins (73-75)

Proton Tetrasaccharide He xa saccharide

GalNAcol H- 1 H-1' H-2 H-3 H-4 H-5 H-6 H-6'

GlcNAc H- 1 H-2 H-3 H-4 H-5 H-6 H-6'

p-Gal-( 1 -, 4) H- 1 H-2 H-3 H-4

B-Gal-( 1 + 3) H- 1 H-2 H-3 H-4

NAc

/%Gal-( 1 + ~)-B-GICNAC &Gal-( 1 -+ 4)-p-GlcNAc

! 6 6

3 3

1 1 p-Gal-( 1 + 4)+-GlcNAc-( 1 + 6)-j?-Gal

! GalNAc-01 GalNAc-01

t t /%Gal

- -

4.400 4.063 3.451 4.289 - -

4.558 3.171 3.725 3.692 3.614 4.000 3.840

4.410 3.536 3.668 3.923

4.464 3.561 3.673 3.897 2.068, 2065

3.768 3.76 4.389 4.0 13 3.416 4.235 3.946 3.10

4.599,4.556 3.76 3.13 3.71 1 3.602, 3.593 4.002, 3.998 3.858, 3.842

4.470 3.537 3.668 3.922

4.470 3.547 3.612 3.882 2.066, 2.055

2.046

Page 352: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

TABLE VI Patterns of Peripheral Anionic Glycosylation Documented in 0- and N-Linked

Chains of Mammalian Glycoproteins.

Sequence Position found where uncommon

a-NeuAc-(2 + 6)-a-GalNAc-( 1 +)-Ser/Thr a-NeuAc-( 2 -+ 3)-P-Gal-( 1 - a-NeuAc-(2 + 6)-P-Gal-( 1

a-NeuAc-(2 + 8@-NeuAc-(2-

a-NeuAc-(2 + 6)$-GlcNAc-( 1 - a-NeuAc

2

6 P-GlcNAc

1 P-Gal a-NeuAc

2 .1 3

P-Gal 4 t 1

P-GalNAc

.1

?

a-NeuAc-(2 + 6)-/?-GalNAc-( 1- a-NeuGc-(2 + 3)+Gal-( 1- a-NeuGc-(2 - 6)-(~-GalNAc-( 1 +)-Ser/Thr

S0;-6-/l-Gal-( 1-

SOT-~-P-G~CNAC-( 1 -

a-NeuAc-(2 - 3)-[HS03-6]-P-Gal-( 1-

SO,-3-P-Gal-( 1 -

SO;-3 or 4-P-GlcNAc-( 1-

PO;-6-cy-Man-( 1 - SO;-4-P-GalNAc-( 1 -

Human colon (71), swine trachea (93), and rat sublingual mucins (54), in ad- dition to N-linked chains

Glycophorin (84), neural cell adhesion molecule (NCAM) (94)

Bovine submaxillary mucins (54)

Fetuin (93, rat plasma hemopexin (96) Rat serotransferrin (97)

Human Tamm Horsfall glycoprotein, erythrocytes, and urinary mucins (98)

Human lutropin (99) Porcine submaxillary mucins (100) Porcine ( 100) and bovine (10 1) submax-

iUary mucins Keratan sulfate (102- 104), tracheo-

bronchal mucins (64) Keratan sulfate ( 102 - 104), hen ovomu-

cin (105), rat gastric mucins (106), porcine thyroglobulin (107)

Recombinant tissue plasminogen activator expressed in mouse epithe- lial cells (108)

Porcine thyroglobulin (107), meconium

Canine submaxillary mucins (54) Glycopeptide hormones (99,109) Lysosomal hydrolases (1 10)

(76)

a Gal, GlcNAc, and GalNAc have the D configuration. Only the broad type of sialic acid is specified as 5-N-acetyl-(NeuAc) and 5-N-glycolyl (NmGc) neuraminic acid. The large family of sialic acids varying in acylation patterns has to be reviewed as a separate topic ( I 1 I), and indeed classical studies involving release of chains by hydrazinolysis or alkaline-borohydride degradation have largely ignored these substitutions. In more-recent studies (1 12), in which enzymic release of N-Linked chains has been adopted, patterns of 0-acetylation and 0-sulfation are beginning to be established.

329

Page 353: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

330 ELIZABETH F. HOUNSELL

terns of anionic substitution found for N- and 0-linked chains of glycoproteins. Short stretches of the sulfated sequences of proteoglycans can also be found on 0- and N-linked chains of serum and cell-membrane glycoproteins. Thus, it is pertinent to look for sulfated sequences on all types ofglycoconjugate. Both mass-spectrometric and n.m.r.-spectral methods are ideal for their identification, and these are treated next.

1. lH-n.m.r. - spectral and Mass-Spectrometric Analysis of Sulfated Oligosaccharide Chains of Glycoproteins

The presence of sulfate groups can be readily detected by f.a.b.- or 1.s.i.- m.s. because of the characteristic mass and fragmentation pattern given by successive loss of sulfate, usually as the Na+ or K+ salt. This was well exem- plified by the characterization of oligosaccharides of keratan sulfate (102) carried out by negative-ion f.a.b.-m.s. of native oligosaccharides at the 5- nmol level. In this study, no fragmentation of the underlying oligosaccharide sequence was seen, and, therefore, no indication was given of the position of the sulfate groups, but only the number of them present. The former infor- mation was obtained (103) by 'H-n.m.r. spectroscopy, and another study ( 104) has documented 13C-n.m.r.-spectral parameters for keratan sulfate. The characterization of monosaccharide sequence, linkage pattern, and sul- fate location of di- to decasaccharides of keratan sulfate was achieved by 'H-n.m.r. spectroscopy by comparison of the chemical shifts with those of the nonsulfated disaccharide. Table VII shows this comparison for p-D- GlcNAc-( 1 + 3)-~-Gal and HS03(-, 6)-p-~-GlcNAc-( 1 -+ 3)-~-Gal. Large, downfield-shift differences of 0.44 and 0.46 ppm were found for the protons at C-6, and progressively lesser downfield-shift differences were found for protons at C-5, C-4, C-3, and C-2. For the larger oligosaccharides of this series, 'H-n.m.r. correlated spectroscopy (COSY) gave the chemical- shift assignments of the majority of protons, which showed downfield-shift differences compatible with 0-6 sulfation of all but the reducing-end sugar, as in the following.

SOT 1

SOT 1

so; 1

6 6 6 p-~-GlcNAc-( 1 -D 3)-p-D-Gd-( 1 -.) ~)-~-D-G~cNAc-( 1 + 3)-D-Gd

Although data on other sulfated oligosaccharides are not so extensive, it has been shown, for example, that sulfation at 0-3 of galactose in 0-linked chains of meconium (76) and N-linked chains ofporcine thyroglobulin (107) increases the chemical shift of H-1 of Gal by f 0.1 16 ppm (kO.00 1 ppm).

Together, the published data show characteristic changes in chemical shift, caused by sulfation, which may be used as an indicator in elucidating

Page 354: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

OLIGOSACCHARIDE DETERMINANTS OF GLYCOPROTEINS 33 1

TABLE VII Comparison of the 'H n.m.r. Chemical Shifts (p.p.m. from 4,4-Dimethyl-4-silapentane-l-

sulfonate at 295 K) for the Disaccharide &~-GlcNAc-(l - 3)-a/&~-Gal and Its Analogue Sulfated at 0-6 of GlcNAc

Sulfated disaccharide

&~-GlcNAc-(l+ 3)-cu/BD-h1 6

Disaccharide t

Proton a P a P

&~-GlcNAc-(l+ 3)u/&~-Gal so,

GlcNAc H- 1 H-2 H-3 H-4 H-5 H-6 H-6' NAc

Gal H- 1 H-2 H-3 H-4 H-5 H-6 H-6'

4.7 17

3.561 3.49 1 3.44

5.226 3.849 3.900 4.198 4.086

4.695

3.565 3.472 3.45

3.752

3.897 3.763 2.038

4.560 3.529 3.699 4.142

3.8-3.6 3.8-3.6

4.727

3.576 3.521 3.66 4.335 4.228

5.227 3.852 3.895 4.233 4.086

4.707

3.580 3.512 3.55 4.337 4.219

3.781

2.037

4.563 3.531 3.695 4.176

3.8-3.6 3.8-3.6

the sulfation patterns of a wide range of oligosaccharides. As shown by the results of studies (1 13) of heparin binding to antithrombin 111, sulfation patterns have significant effects on molecular recognition by virtue of their influence on charge density and oligosaccharide topography. For keratan sulfate, there has been characterized a series of monoclonal antibodies that are specific for polysulfated sequences (102,103). The n.m.r. data were con- sistent with the X-ray crystallographic data showing sulfate residues pointing away from the Gal-GlcNAc backbone and defining a tetrasulfated tetrasac- charide unit involved in molecular recognition.

2. 'H-n.m.r. Analysis of Sialic Acid Substitution Patterns

The linkage of sialic acid to Gal, GlcNAc, or GalNAc may be determined on the microscale by methylation analysis before and after treatment with

Page 355: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

332 ELIZABETH F. HOUNSELL

neuraminidase or mild acid (0.01 -0.1 M, 80°C) (3). When sufficient mate- rial is available, 'H n.m.r. permits identification of the type of sialic acid present and the substitution pattern. Characteristic chemical shifts for the sialic acid of various sialylated sequences are compared in Table VIII. It may be seen that at an accuracy of k 0.03 ppm (33), it is possible to distinguish the major types of sialic acid linkages shown from the H-3 axial and equatorial signals alone. Additional differences in chemical shift are brought about by the presence of 0-acyl groups as described by Vliegenthart et al. in Ref. 1 1 1 , in particular large downfield shifts of protons on 0-acetylated carbon atoms and on carbon atoms adjacent to those 0-acetylated. More-subtle differ- ences in the chemical shifts of the two H-3 protons and of the H-4-H-9 of sialic acids occur in different oligosaccharide sequences, including those varying at the monosaccharide adjacent to that linked by the sialic acid; for example, the chemical shifts for NeuAc linked (2 --.* 3) to Gal are dependent on the linkage of Gal (1 + 3) to GlcNAc, (1 -+ 4)-GlcNAc, or (1 ---* 3)- GalNAc-ol and on the presence of NeuAc (2 -, 6), Fuc (1 ---* 3), for Fuc (1 + 4) linked to the GlcNAc. As with fucosylated sequences (1 14- 1 16), these differences may be interpreted in terms of conformational information additional to that obtained from nuclear Overhauser enhancement (n.0.e.) studies.

VI. THE CONFORMATIONS AND MOLECULAR RECOGNITION OF CARBOHYDRATE DETERMINANTS DISTANT FROM THE PROTEIN

OLIGOSACCHARIDE CORE OF GLYCOPROTEINS To a certain extent, the peripheral moieties of oligosaccharide chains at a

distance from the protein skeleton may be viewed as independent confor- mational entities. This is exemplified by the fact that the n.m.r. data from a particular structural determinant can be translated to the same determinant on a different core, as discussed previously (33). Considerable use has been made of this situation when viewing oligosaccharides as antigens related to the blood-group structures in that glycoproteins, glycolipids, and the free oligosaccharides of milk, urine, and feces have provided an extensive library for structural and immunochemical studies (as reviewed in Refs. 33 and 5 1 and discussed next). 'H-n.m.r.-spectroscopy of oligosaccharides and glycoli- pids have provided near-complete chemical-shift assignments and some n.0.e. data on through-space interactions, that have been interpreted in terms of 3D determinants. Conformational analysis by 'H-n.m.r. spectros- copy of free oligosaccharides is discussed next.

Page 356: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

OLIGOSACCHARIDE DETERMINANTS OF GLYCOPROTEINS 333

TABLE VIII 'H N.m.r. Chemical Shifts" (p.p.m. from 4,4-Dimethyl-4-silapntane-l-

sulfonate at 295 K) of the H-3 Axial and H-3 Equatorial Protons of Oligosaccharides Containing N-Acetylneuraminic Acid and N-

Glycolylneuraminic Acid

Resonance

Structure H-3ar H-3eq ~~

a-NeuAc-(2 + 3)-/3-~-Gd a-NeuAc-(2 -B 3)-p-~-GalNAc-( 1 -+ 4)-]B-~-Gal a-NeuAc-(2 + 6)-B-~-Gal or -p~-GalNAc c~-NeuAc(COAc)-(2 -P 6)-/3-~-Gal a-NeuAc-(2 -, 6)-/3-~-GlcNAc c~-NeuAc-(2 -, 6)-~-GalNAc-ol a-NeuGc-(2 -B 3)-/3-~-Gal a-NeuGc-(2 --* 6)-GalNAc-ol

1.80 2.16 1.93 2.66 1 I1 2.67 1.85 2.68 1.70 2.14 1.69 2.12 1.82 2.19 1.71 2.15

Chemicalshiftsareaveragedvalues(~.03 p.p.m.)(33)fromourownstudiesand thosefrom Refs. 31-33,62,66,86,96-101, 111,and 112.

1. Blood-Group-Related Antigens Lemieux (1 14) has discussed the recognition of the blood-group-type oli-

gosaccharide determinants by antibodies and lectins based on (i) a series of pioneering studies on the n.m.r.-spectral analysis of chemically synthesized oligosaccharides and (iz] molecular modeling. Earlier studies reviewed by Durette and Horton ( 1 17) had established monosaccharide conformational analysis, in particular defining the ring stereochemistry and the importance of the exoanomeric effect. The use of the hard-sphere exoanomeric algo- rithm (HSEA) to predict qb,y angles around the glycosidic bonds, together with data from n.m.r.-spectral studies [which, for example, permits detec- tion of strong deshielding of protons in close proximity to glycosidic-bond and ring-oxygen atoms (1 15)], was applied to the analysis of a series of oligosaccharide determinants recognized by lectins and anti-blood-group antisera ( 1 18 - 12 1). Similar studies were performed on oligosaccharides obtained from milk (1 16,122). The extensive purification and structural studies (123- 125) of Kuhn, Kobata, Yamashita, Ginsburg, and colleagues established the diversity of the oligosaccharide molecules in milk. New structures continue to be found ( 126) and similar oligosaccharides in feces (1 27 - 129), and urine ( 130 - 132) have provided other sources for chemical- shift assignment by COSY, relayed COSY, and long-range COSY experi- ments. Additional n.m.r. parameters from I3C- and 'H-n.m.r.-spectral ex-

Page 357: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

334 ELIZABETH F. HOUNSELL wo HO e H , o H

OH OH

(P 4) (3

FIG. 1. -The disaccharide&D-Gal-( 1 -P 4)-/.%~-GlcNAc, showing the 4 , ~ angles defined by IUPAC a~ 0'-C'-0-C,, C'-O-C,-C~,~~~.

periments leading to conformational information on these oligosaccharide sequences [for example, analysis of spin-relaxation time, n.0.e. effects, and 13C- 'H long-range coupling constants (1 33 - 136)] have provided extensive data that substantiate the modeling approach, as have molecular-dynamic simulations (1 35,137,138).

In several cases, these data have been incorporated into molecular models built with the aid of information from molecular-mechanics algorithms (e.g., HSEA, GSEA, MM2CARB) on energetically favorable 4 , ~ torsion angles (see Fig. 1) around the glycosidic bonds. The models are made using commercial software packages, the forcefields of which (e.g., AMBER, CVFF, TRIPOS, and CHARMm) are parameterized to take account of specific oligosaccharide factors such as charge and the anomeric effect (137,138). To date, these calculations have mostly been based on simula- tions in a vacuum, although other studies have examined the effect of water on energy minimization and molecular dynamics or have approximated the

TABLE IX Calculation of the 4, yl Angles of Minimum-Energy Conformers

by the HSEA Algorithm"

Angles

Sequence 6 Structure(Fig. 2)

a-Fuc-( 1 + 2) to Galb -70 135 a ~-Fuc-( 1 - 3) to GIcNAc -70 -95 b ~-Fuc-( 1 -+ 4) to G~cNAc -65 145 C

/%Gal-( 1 - 3) to GlcNAc - 60 - 1 10 d B-Gal-( I -4) to GlCNAc -65 130 e p-GlcNAC-( 1 + 3) to Gal -66 150 f c~-Gal-( 1 - 3) to Gal 55 -170 8

a Data taken from Lemieux ef al. and standardized to IUPAC format (Fig. 1).

All monosaccharides are in the D form except for L-FUC.

Page 358: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

OLIGOSACCHANDE DETERMINANTS OF GLYCOPROTEINS 335

presence of water by use of a dielectric constant of 1 - 80 keV. The 4,vangles are shown in Table IX for the series of blood-group-related disaccharides at minimum-energy conformations (Fig. 2) predicted by the HSEA algorithm. These disaccharides serve as building units for oligosaccharides, which can then be refined by using computer-graphics molecular mechanics programs parameterized for oligosaccharides. Different methods of calculation often give similar contour patterns of energetically favorable conformations, but find different energy minima. As the probability of finding the molecule in the energetic minimum is low, conformational space around several low- energy states should be explored ( 139). Where through-space distances given by n.m.r.-n.0.e. experiments are known and where interpretation of immunological - biological data is involved, models incorporating solution conformations within the low-energy wells may be of great help in visual- izing oligosaccharide determinants of activity. N.m.r.-spectral data are cru- cial for any model building, as the relative flexibility and heterogeneity of the oligosaccharide moieties of glycoproteins has thus far largely prevented their visualization by X-ray crystallography. It should be remembered, however, that the molecules could sample 3D conformational space at a higher rate than can be detected by n.m.r. relaxation and, furthermore, that, in molecu- lar recognition of oligosaccharides, energetically unfavorable solution- conformations could be bound. With respect to the blood-group-related antigens, these generally form structures that are more stereochemically restricted and for which modeling incorporating n.0.e. data may be more appropriate than for flexible backbones. This factor may also contribute to their immunogenicity, as determined by their recognition by polyclonal serum antibodies and monoclonal antibodies raised in vitro.

The first oligosaccharide determinant for a hybridoma antibody charac- terized in detail was the 3-fucosyl-N-acetyllactosamine (W, Table X) se- quence P-D-Gal-( 1 + 4)-[a-~-Fuc( 1 + 3)]$-~-GlcNAc. The initial anti- body was raised against a stage-specific embryonic antigen (SSEA-1) of mouse (140) and characterized by using oligosaccharides obtained from human milk ( 140,14 1). Other antibodies raised against a promyelomonocy- tic leukemia cell line (HL60) and against the receptor for epidermal growth- factor were also found to recognize the same or a related structure (142,143). Determination of the specificity of other monoclonal antibodies recognizing oligosaccharides of glycoproteins soon followed [as reviewed in Ref. (5 l)]. Studies described therein established, for example, that W, W, and com- bined difucosylated W/W are tumor-associated antigens of milk fat- globule membrane glycoproteins (1 16) and that the ALeb and ALeY (Table X) sequence is present on the N-linked chains of the receptor for epidermal growth factor on A43 1 cells (143). Structural studies on human, small intes- tinal, epithelial cells ( 144) have since revealed the presence of Ley and ALeY

Page 359: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

336

a

ELIZABETH F. HOUNSELL

C

e b

FIG. 2. -Conformations of oligosaccharides having the 4 , ~ angles shown in Table IX for minimum-energy conformations predicted by the HSEA algorithm for use as building units in molecular modeling. The molecules are shown without the anomeric hydroxyl group.

Page 360: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

OLIGOSACCHARIDE DETERMINANTS OF GLYCOPROTEINS 337

FIG. 2. (Continued)

sequences on N-linked chains. As with the 3-fucosyl-N-acetyllactosamine and poly-N-acetyllactosamine (i antigenic structure) sequences, these were detected first on N-linked chains by antibody reactivity before they were established by structural characterization (5 1). Figure 3 shows predicted solution conformers for oligosaccharide determinants related to Le" and Le". These models may be used to propose molecular features recognized by the series of monoclonal antibodies raised against these structures. They can also be used to interpret the molecular recognition of other carbohydrate-binding proteins of nonimmune origin in plants and mammals. As discussed next, related structures have been shown to be important in the interactions of lymphocytes, platelets, and endothelial cells ( 145).

2. Sialylated Oligosaccharide Determinants

The first conformational information on the large family of sialic acids (1 1 1) of mammalian glycoproteins came from the comprehensive study ( 146) of the following glycopeptides,

a-NeuAc-(2 - 3)-/?-~-Gd-( 1 4 4)-P-~-GlcNAo(l~ Am)

a-NeuAe(2 6)-/?-DGd-( 1 + 4)-/?-~-GlcNAc-(l+ Am),

and the sialylated Le" antigen Ca 19.9, which is a tumor-associated antigen associated with several gastrointestinal malignancies ( 147):

a-NeuAc-(2 - 3)-/?-~-Gal-( 1 - 3)-a-[~-Fuc-( 1 - 4)]-~-GlcNAc.

The data for these oligosaccharides have been incorporated into the molecu- lar models shown in Figs. 3 and 4. The conformation and molecular recog- nition of sialylated oligosaccharides are an important area for future investi-

Page 361: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

338 ELIZABETH F. HOUNSELL

TABLE X The Oligosaccharide Sequences of Blood-Group and Related

Determinants Shown in Figs. 3 and 4

Determinant Sequence

L e a

Le"

Sialyl Le"

Sialyl Le"

ALeb

ALeY

LY-L-FUC

i

f

i

f

4

gal

4

P-D-Gal-( 1 - ~)-~D-G~cNAc

LY-L-FUC-( 1 - 3)-p-D-&NAc a-L-FUC

4 a-~-NeuAc-(2 - 3)-j.-~-Gd-( 1 + 3)-/3-~-GkNAc

a-~-NeuAc-(2 - 3)-b-~-Gal

4 (Y-L-Fuc-( 1 - ~)-~-D-G~cNAc

1 1 LY-D-G~NAc (Y-L-FUC

1 4 3

LY-L-FUC-( 1 + 2)-p-~-Gd-( 1 - 3)-/3-~-GlcNAc (Y-D-G~~NAc

1

3 LY-L-FUC-( 1 - 2)-p-~-Gd

4 i

LY-L-FUC-( I - 3)-/3-D-GlcNAC

gation because of the perceived central importance of these prominent chain-terminating sequences in cellular interactions. There is strong evi- dence that the Lea and Lex determinants having a-NeuAc-(2 +- 3)-P-~-Gal substitution are involved in the specific recognition of lymphocytes and neutrophils with endothelial cells via carbohydrate-binding proteins called selectins [reviewed in Ref. (1431. Both sialylated and sulfated oligosaccha- ride sequences have been implicated in trafficking of lymphocytes to high endothelial cell venules. These findings build on earlier studies on mamma- lian carbohydrate recognition, which showed a binding lectin of the liver ( 1 48) and a Man-6-PO,-binding lectin of lysosomal membranes ( 1 10,149), both involved in glycoprotein trafficking.

Page 362: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

OLIGOSACCHARIDE DETERMINANTS OF GLYCOPROTEINS 339

Additional conformational studies have been carried out on the [a- NeuAc-(2 + s)], homopolymers that make up the type-specific antigens of meningococcus (1 50,15 1). These same sequences have been identified on the human neural cell-adhesion molecule (NCAM) as components of N- linked glycoprotein chains (94) and are strongly implicated in the etiology of meningococcal disease, because they occur primarily in the young, who are susceptible, but are not expressed on adult NCAM. The importance of sialic acid in infection was first recognized for the cellular receptor of influenza virus hemagglutinin by Gottschalk (1 52). Further conformational studies ( 153) have identified the molecular features recognized by the hemaggluti- nin binding-site, which appears to be largely restricted to functional groups on the a-NeuAc-(2 + 3/6) moiety itself rather than internal residues of the glycoprotein or glycolipid chains that the virus binds.

Sialylated sequences are also the targets for bacterial adhesion and bacte- rial toxins. The specificity for binding of Mycoplasma pneumoniae was shown ( 154) to involve polylactosamine sequences terminating in a- NeuAc-(2 + 3)-P-~-Gal. There is a transient rise in anti-1 titer directed against the backbone [p-D-Gal-( 1 - 4)-p-~-GlcNAc-( 1 - 3/6)], sequence experienced after infection by M. pneumoniae. The i and I antigens were initially characterized as, respectively, linear and branched poly-N-acetylac- tosamine sequences recognized by naturally occurring monoclonal cold- agglutinin autoantibodies (5 1). In addition, among the erythrocyte autoan- tibodies associated with cold-agglutinin disease of man, several are known to be monoclonal antibodies directed against sialic acid-containing determi- nants (1 55,156). The antibodies designated anti-Pr, anti-Gd, Fl, and Sa appear to react primarily with the sequence a-NeuAc-(2 - 3)-p-~-Gal- (1 - 4)-p-GlcNAc, although there is some variability in activity with a-D- NeuAc-(2 - 3) or c~-NeuAc-(2 - 6) on different backbones (156). Early data (1 57,158) on the conformations of the backbone antigens i and I are incorporated into the molecular modeling shown in Fig. 4, together with sialylated analogues.

Another antigen of importance in medicine is the sequence (Fig. 4) a- NeuGc-(2 - 3)-p-~-Gal-. This is the Hanganutziu- Diecher (H-D) antigen originally recognized as being an antigen giving rise to serum sickness caused by transfusion of horse serum into humans ( 159). The sequence is present on horse erythrocyte glycoproteins, but is not biosynthesized in human cells, except as a minor component on tumor glycolipids. However, in the new era of recombinant DNA technology, this sequence has been detected on glyco- proteins destined for human use that have been grown in Chinese hamster ovary (CHO) cells. This finding underlines the importance of the character- ization of oligosaccharide determinants that are central factors involved in infection and immunity.

Page 363: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

340

Page 364: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

FIG. 3-Possible solution conformers of oligosaccharide sequences related to the Len and Leb antigens ( Lea, Lex, sialyl Lea, sialyl Lex, ALeb, ALey ). As described in the text, oligosaccharides in solution sample 3D space in local energy-minima wells, so that the conformations shown are approximate only. The models stand only to show the stereochemical hindrance to movement in these branched oligosaccharides. The molecules are shown without the anomeric hydroxyl group.

Page 365: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

Sialyl i Sialyl I

FIG. 4-A comparison of the H-D antigenic component and the receptor for influenza virus hemagglutinin, which are a-NeuGe(2 + 3)-/3-Gal and a-NeuAc-(2 - 3)-/3-Gal, respectively, on the backbones pD-Gal-( 1 +

4)-p~-GlcNAc-( 1 + 3)-&~-Gal-( 1 - 4)-~-GlcNAc (sialyl i), /3-~-Gal-( 1 + 4)-&~-GlcNAc-( 1 + 6)-~-Gal (sialyl I). The CH, of the acetamido group of N-acetylneuraminic acid, which is a CH,OH group in N-glycolyl- neuraminic acid. is shown.

Page 366: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

OLIGOSACCHARIDE DETERMINANTS OF GLYCOPROTEINS 343

VII. THE CONFORMATIONS AND MOLECULAR RECOGNITION

MOIETY OF GLYCOPROTEINS The oligosaccharide sequences just discussed are linked to glycoproteins

through the following (generalized) N- and 0-linked cores (to asparagine and serine or threonine, respectively).

OF CARBOHYDRATE DETERMINANTS ADJACENT TO THE PROTEIN

u-D-Gd-( 1 - 4/3)-&~-GkNAc-( 1 + 3)In- 1 - 6/4/2-c~-~-Man-( 1 + 6/3)- &&Man-( 1 - 4)-&~-GlcNAc-( 1 + 4)-8-~-GlcNAc-( 1 + Am)

p-~-Gal-( 1 + 4/3)-/3-~-GlcNAc-( 1 - 6/3)-],-fi~-Gd-( 1 -+ 3)-a-~-GdNAc-(l-+ Ser/Thr)

Numerous detailed studies have been performed on the conformations of D-Man-containing sequences of glycoproteins ( 137 - 139,160 - 168). The re- sults show that there is considerable flexibility and make it difficult to sum- marize the solution conformations that oligosaccharide chains containing these sequences are likely to adopt. The general consensus is, as predicted by Montreuil(30), that many allowed conformations are possible. For opera- tional reasons, many of the studies have been performed on isolated oligo- saccharides and have not taken into account (i) the restrictions of degrees of freedom imposed by the hydrodynamic volume of complete oligosaccharide chains in solution and (iz] the effect ofinteraction with the protein backbone.

For N-linked chains, it is pertinent to discuss the latter aspect, for which some constraints on degrees of freedom have been established. For 0-linked chains, a picture in which the chains can be said to be in intimate contact with the protein, imposing conformational constraints and allowing for joint oligosaccharide - protein determinants, is emerging.

p-D-Gd-( 1 -D 4/3)-fiD-GlCNAC-( 1 -D ~/~)-],-~Y-D-G~NAC-(I -P Ser/Thr) or

1. N-Linked Chains

There is an extensive databank on the chemical shifts of free oligosaccha- rides and those linked to one or two amino acids. Several n.m.r. and model- ing studies have also been performed on glycopeptides and glycoproteins ( 169 - 172). The conclusion from these latter studies is that the asparagine - GlcNAc bond extends away from the protein backbone, so that only small conformational changes take place upon elongation of the oligosaccharide side-chains, and attachment to protein does not affect the solution confor- mation of the oligosaccharide.

Where X-ray crystallographic data for glycoproteins are available, results show that the N-linked oligosaccharides either can extend away from the protein [as suggested (1 73), for example, for the single chain at Asn 86 in the human lymphocyte antigen HLA A2], or may interact with the rest of the molecule [as shown (42,174), for example, for the two chains of the CH,

Page 367: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

344 ELIZABETH F. HOUNSELL

domains of the Fc region of immunoglobulin GI. In the former case, it appears that there is no significant interaction of the protein and carbohy- drate, and therefore it may be envisaged that the oligosaccharide determi- nants at the periphery of the chains are themselves functional units that have a unique accessibility at the surface of glycoproteins and cells.

2. 0-Linked Chains

Initial conformational studies suggest that, for 0-glycosylation, the glyco- sylated amino acid will be in a p-turn in order to accommodate the &-Gal- NAc residue and that there is more conformational flexibility for GalNAc- Ser than GalNAc-Thr, but both of these 0-a-glycosidic linkages are substantially more rigid than those in the p-GlcNAc-Asn case (1 70,174).

An additional factor to be considered with respect to 0-glycosylation is that multiple glycosylation sites are often present, for example, in glyco- phorin (175 - 177), the antifreeze glycoprotein of fish (178), and mucins (1 79 - 180). Multiple 0-glycosylation sites have also been shown on a variety of cell-membrane and serum glycoproteins (Table XI). Their effects on protein conformation are now being ascribed specific biological roles ( 18 1) in addition to their recognition as antigens. Because of the close interaction of 0-linked oligosaccharides with the peptide backbone, each moiety may affect the antigenicity of the other, for example, sialic acid residues on the disaccharide p-D-Gal-( 1 +. 3)-a-~-GalNAc-( 1 -)-Ser/Thr are necessary for binding to some blood-group M and N antisera ( I 75 - 177). The Thomsen- Friedenreich (T) antigen, having the unsialylated sequence p-D-Gal-( I - 3)-a-~-GalNAc-( 1 -+Ser/Thr, may be detected, for example, as an onco- developmental antigen in fetal and neoplastic colonic mucosa but not in the normal adult mucosa (175). Likewise a single-a-GalNAc linked to a Thr residue of human fibronectin forms a specific oncodevelopmental epitope

TABLE XI Examples of Serum and Membrane

O-Glycosylation Sites Glycoproteins Having Multiple

Lymphocyte antigen CD45 Human chorionic gonadotropin Lipoprotein (a) Low-density lipoprotein (LDL) receptor Human lymphocyte antigen CD8 Interleukin-2 receptor Rat renal a-glutamyltranspeptidase Sucrase and isomaltase

Page 368: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

OLIGOSACCHARIDE DETERMINANTS OF GLYCOPROTEINS 345

recognized by a monoclonal antibody (1 82). Last, a single N-acetylglucosa- mine group /I-linked to Ser or Thr occurs in cytoplasmic glycoproteins ( 183,184) and appears to be important in their intracellular trafficking; it has been proposed ( 17) that this may be a reciprocal signal to 0-phosphorylation of proteins. The overall view that can be obtained to date is that oligosaccha- ride sequences have many and diverse roles that we are now beginning to dissect, starting from a base of detailed physicochemical characterization.

ACKNOWLEDGMENTS

The author is extremely grateful to Miss E. White for her patient typing, to Mr. D. Renouf for his collaboration in the molecular-modeling studies, and to Dr. T. Feizi for helpful discussions.

REFERENCES

1. D. A. Rees, Adv. Carbohydr. Chem. Biochem., 24 (1969) 267-332. 2. W. C. Johnson, Adv. Carbohydr, Chem. Biochem., 45 (1987) 73- 124. 3. E. F. Hounsell (Ed.), Methods in Molecular Biology, Vol. 14, Glycoprotein Analysis in

4. S . Hakomori, J. Biochem. (Tokyo), 5 5 (1964) 205-207. 5. K. Stellner, H. Saito, and S. Hakomori, Arch. Biochem. Biophys., 155 (1975) 464-472. 6. P. A. Sandford and H. E. Conrad, Biochemistry, 5 ( 1966) 1508 - I5 12. 7. D. M. W. Anderson and G. M. Cree, Carbohydr. Res., 2 (1966) 162- 167. 8. J. S. Brimacombe, B. D. Jones, M. Stacey, and J. J. Willard, Carbohydr. Rex, 2 (1966)

9. H. Bjomdal, C. G. Hellerqvist, B. Lindberg, and S. Svensson, Angew. Chem. Int. Ed.

Biomedicine. Humana Press, 1993.

167- 174.

Engl., 9 (1970) 610-619. 10. G. G. S. Dutton, Adv. Curbohydr. Chem. Biochem., 30 (1974) 10- 110. 1 1. K. A. Karlsson, I. Pascher, W. Pimlott, and B. E. Samuelsson, Biomed. Mass Spectrom., 1

12. I. Ciucanu and F. Kerek, Carbohydr. Res., 131 (1984) 209-217. 13. R. Kuhn and H. Egge, Chem. Ber., 96 (1963) 3338-3348. 14. S. Honda, Anal. Biochem., 140 (1984) 1-47. 15. G. C. Hansson, I. Carlstedt, and H. Karlsson, Biochemistry, 28 (1989) 6672-6678. 16. Y. Leroy, J. Lemoine, G. Ricart, J.-C. Michalski, J. Montreuil, and B. Foumet, Anal.

17. K. Kakehi and S. Honda, J. Chromatogr., 379 (1986) 27-55. 18. P. Jackson, Biochem. SOC. Trans. 21 (1993) 121 - 125. 19. C. C. Sweeley, R. Bentley, M. Makita, and W. W. Wells, J. Am. Chem. Soc., 85 (1963)

20. T. Bhatti, R. E. Chambers, and J. R. Clamp, Biochim. Biophys. Acta., 222 (1970) 339-

2 1. G. J. Genvig, J. P. Kamerling, and J. F. G. Vliegenthart, Carbohydr. Rex, 77 ( I 979) 1 - 7. 22. M. R. Hardy, in Methods in Enzymolom, Vol. 179, pp. 76-82. Academic Press, San

23. A. Dell, Adv. Carbohydr. Chem. Biochem., 45 (1987) 19-72. 24. A. L. Burlingame, D. Maltby, D. H. Russell, and P. T. Holland, Anal. Chem., 60 (1988)

(1974) 49-56.

Biochem., 184 (1990) 235-243.

2497-2507.

347.

Diego, 1989.

294R-342R.

Page 369: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

346 ELIZABETH F. HOUNSELL

25. E. F. Hounsell, M. J. Madigan, and A. M. Lawson, Biochem. J., 219 (1984) 947-952. 26. A. M. Lawson, W. Chai, G. C. Cashmore, M. S. Stoll, E. F. Hounsell, and T. Feizi,

27. A. S. Angel and B. Nilsson, in Methods in Enzymology, Vol. 193, pp. 587-607. Academic

28. M. S. Stoll, E. F. Hounsell, A. M. Lawson, W. Chai, and T. Feizi, Eur. J. Biochem., 189

29. T . Feizi, in Carbohydrate Recognition in Cellular Function, Ciba Foundation Sympo-

30. J. Montreuil, Adv. Carbohydr. Chem. Biochem., 37 (1980) 157-223. 31. J. F. G. Vliegenthart, H. van Halbeek, and L. Dorland, Pure Appl. Chem., 53 (1981)

32. J. F. G. Vliegenthart, L. Dorland, and H. van Halbeek, Adv. Carbohydr. Chem. Biochem.,

33. E. F. Hounsell and D. J. Wright, Carbohydr. Res., 205, (1990) 19-29. 34. K. Bock, C. Pederson, and H. Pederson, Adv. Carbohydr. Chem. Biochem., 42 (1984)

35. K. Dill, E. Berman, and A. A. Pavia, Adv. Carbohydr. Chem. Biochem., 43 (1985) 1-49. 36. J. A. van Kuik, K. Hird, and J. F. G. Vliegenthart, Carbohydr. Res. 235 (1992) 53-68. 37. K. Hermansson, P.-E. Jansson, L. Kenne, G. Widmalm, and F. Lindh, Carbohydr. Rex

38. G. M. Lipkind, A. S. Shashkov, N. E. Nifant’ev, and N. K. Kochetkov, Carbohydr. Res.,

39. M. W. Spellman, Anal. Chem., 62 (1990) 1714- 1722. 40. G. Pfeiffer, H. Geyer, I. Kalsner, P. Wendorf, and R. Geyer, Biomed. Chrornatogr., 4

4 1. T. Mizuochi, T. J. Matthews, M. Kato, J. Hamako, K. Titani, J. Solomon, and T. Feizi, J.

42. T. W. Rademacher, R. B. Parekh, and R. A. Dwek, Annu. Rev. Biochem., 57 (1988)

43. K. Yamashita, T. Mizuochi, and A. Kobata, in Methods in Enzymolou, Vol. 83, pp.

44. F. Maley, R. B. Trimble, A. L. Tarentino, and T. H. Plummer, Jr., Anal. Biochem., 180

45. K. D. Smith, A.-M. Harbin, R. A. Carruthers, A. M. Lawson, andE. F. Hounsell, Biomed.

46. M. J. Davies, K. D. Smith, R. A. Carruthers, W. Chai, A. M. Lawson and E. F. Hounsell, J.

47. A. Kobata, in V. Ginsburg and P. W. Robbins (Eds.), Biology of Carbohydrates, Vol. 2, p.

48. W. T. G. Morgan, Proc. R. SOC. London Ser. B., 151 (1960) 308-347. 49. W. M. Watkins, Biochem. SOC. Trans., 15 (1987) 620-624. 50. E. A. Kabat, in Carbohydrates in Solution, Advances in Chemistry Series No. 117, pp.

51. T. Feizi, Nature, 314(1985) 53-57. 52. S. Hakomori, Sci. Am., 254(5) (1986) 32-41. 53. E. F. Hounsell and T. Feizi, Med Biol., 60 (1982) 227-236. 54. A. Herp, A. M. Wu, and J. Moschera, Mol. Cell Biochem., 23 (1979) 27-44. 55. A. M. Wu, E. A. Kabat, B. Nilsson, D. A. Zopf, F. G. Gruezo, and J. Liao, J. Biol. Chem.,

56. B. N. N. Rao, V. K. Dua, and C. A. Bush, Biopolymers, 24 (1985) 2207-2229

Carbohydr. Rex, 200 (1990) 47-57.

Press, San Diego, 1990.

(1990) 499-507.

sium 145, pp. 62-79. Ciba Foundation, 1989.

45-77.

41 (1983) 209-374.

193 -225.

235 (1992) 69-81.

237 (1992) 11-22.

(1990) 193- 199.

Biol. Chem., 265 (1990) 8519-8524.

785-838.

105- 126. Academic Press, San Diego, 1982.

(1989) 195-204.

Chromatogr., 4 (1 990) 26 1-266.

Chromatography 646 (1993) 317-326.

87. Wiley, New York, 1984.

334-361. American Chemical Society, Washington, D.C., 1972.

259 (1984) 7178-7186.

Page 370: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

OLIGOSACCHARIDE DETERMINANTS OF GLYCOPROTEINS 347

57. J. H. G. M. Mutsaers, H. van Halbeek, J. F. G. Vliegenthart, A. M. Wu, and E. A. Kabat,

58. V. K. Dua, B. N.N. Rao, S.-S. Wu, V. E. Dube,andC.A. Bush, J. Biol. Chem., 261 (1986)

59. T. Feizi, E. A. Kabat, G. Vicari, B. Anderson, and W. L. Marsh, J. Imrnunol., 106 (1971)

60. G. Lamblin, M. Lhermitte, A. Boersma, P. Roussel, and V. Reinhold, J. Biol. Chem., 255

6 1 . H. van Halbek, L. Dorland, J. F. G. Vliegenthart, W. E. Hull, G. Lamblin, M. Lhermitte,

62. G. Lamblin, A. Boersma, A. Klein, P. Roussel, H. van Halbeek, and J. F. G. Vliegenthart,

63. G. Lamblin, A. Boersma, M. Lhermitte, P. Roussel, J. H. G. M. Mutsaers, H. van

64. T. P. Mawhinney, E. Adelstein, D. A. Moms, A. M. Mawhinney, and G. J. Barbero, J.

65. J. Breg, H. van Halbeek, J. F. G. Vliegenthart, G. Lamblin, M.-C. Houvenaghel, and P.

66. H. van Halbeek, J. Breg, J. F. G. Vliegenthart, A. Klein, G. Lamblin, and P. Roussel, Eur.

67. J. Breg, H. van Halbeek, J. F. G. Vliegenthart, A. Klein, G. Lamblin, and P. Roussel, Eur.

68. A. Klein, G. Lamblin, M. Lhermitte, P. Roussel, J. Breg, H. van Halbeek, and J. F. G.

69. B. L. Slomiany, E. Zdebska, and A. Slomiany, J. Biol. Chem., 259 (1984) 2863-2869. 70. A. Slomiany, E. Zbedska,andB. L. Slomiany, J. Biol. Chem., 259(1984) 14,743- 14,749. 71. D. K. Podolsky, J. Biol. Chem., 260(1985) 15,510-15,515. 72. D. K. Podolsky, J. Biol. Chem., 260 (1985) 8262-8271. 73. E. F. Hounsell, A. M. Lawson, J. Feeney, H. C. Gooi, N. J. Pickering, M. S. Stoll, S. C. Lui,

and T. Feizi, Eur. J. Biochem., 148 (1985) 367-377. 74. E. F. Hounsell, A. M. Lawson, J. Feeney, G. C. Cashmore, D. P. Kane, M. Stoll, and T.

Feizi, Biochem. J., 256 (1988) 397-401. 75. E. F. Hounsell, A. M. Lawson, M. S. Stoll, D. P. Kane, G. C. Cashmore, R. A. Carruthers,

J. Feeney, and T. Feizi, Eur. J. Biochem., 186 (1989) 597-610. 76. C. Capon, Y. Lertoy, J.-M. Wieruszeski, G. Ricart, G. Strecker, J. Montreuil, and B.

Fournet, Eur. J. Biochem., 182 (1989) 139- 152. 77. A. Kurosaka, H. Nakajima, I. Funakoshi, M. Matsuyama, T. Nagayo, and I. Yamashina,

J. Biol. Chem., 258 (1983) 11,594-11,598. 78. F.-G. Hanisch, H. Egge, J. Peter-Katalinic, G. Uhlenbruck, C. Dienst, and R. Fangmann,

Eur. J. Biochem., 152 (1985) 343-351. 79. E. C. Yurewicz, F. Matsuura, and K. S. Moghissi, J. Biol. Chem., 262 (1987) 4733-4739. 80. T. Saito, T. Itoh, and S. Adachi, Biochim. Biophys. Actu, 964 (1988) 213-220. 81. F.-G. Hanisch, G. Uhlenbruck, J. Peter-Katalinic, H. Egge, J. Dabrowski, and U. Da-

82. W. Chai, E. F. Hounsell, G. C. Cashmore, J. R. Rosankiewicz, C. J. Bauer, J. Feeney, T.

83. H. Schachter, Biochem. Cell. Biol., 64(1986) 163-181. 84. M. Fukuda, M. Lauffenburger, H. Sasaki, M. E. Rogers, and A. Dell, J. Biol. Chem., 262

85. L. A. Cole, S. Birken, and F. Perini, Biochem. Biophys. Res. Commun., 126 (1985)

Eur. J. Biochem., 157 (1986) 139-146.

1599- 1608.

1578- 1592.

(1980) 4595-4598.

A. Boersma, and P. Roussel, Eur. J. Biochem., 127 (1982) 7-20.

J. Biol. Chem., 259 (1984) 905 1-9058.

Halbeek, and J. F. G. Vliegenthart, Eur. J. Biochem., 143 (1984) 227-236.

Biol. Chem., 262 (1987) 2994-3001.

Roussel, Eur. J. Biochem., 168 (1987) 57-68.

J. Biochem., 177 (1988) 443-460.

J. Biochem., 171 (1988) 643-654.

Vliegenthart, Eur. J. Biochem., 171 (1988) 631-642.

browski, J. Biol. Chem., 264 (1989) 872-883.

Feizi, and A. M. Lawson, Eur. J. Biochem., 203 (1992) 257-268.

(1987) 11,952-11,957.

333-339.

Page 371: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

348 ELIZABETH F. HOUNSELL

86. A. Pierce-Crktel, J.-M. Wieruszeski, G. Strecker, J. Montreuil, and G. Spik, Eur. J.

87. R. A. Childs, R. Dalchau, P. Scudder, E. F. Hounsell, J. W. Fabre, and T. Feizi, Biochem.

88. R. N. Iyer and D. M. Carlson, Arch. Biochem. Biophys., 142 (1971) 101 - 105. 89. J. Feeney, T. A. Frenkiel, and E. F. Hounsell, Curbohydr. Res., 152 (1986) 63-72. 90. E. F. Hounsell, M. Fukuda, M. E. Powell, T. Feizi, and S. Hakomori, Biochem. Biophys.

9 1. H. van Halbeek, L. Dorland, J. F. G. Vliegenthart, N. K. Kochetkov, N. P. Arbatsky, and

92. A. R. Poole, Biochem. J., 236 (1986) 1 - 14. 93. S. S. Rana, E. V. Chandrasekaran, and J. Mendicino, J. Biol. Chem., 262 (1987) 3654-

94. J. Finne, V. Finne, H. Deagostini-Bazin, and C. Goridis, Biochem. Biophys. Res. Com-

95. E. D. Green, G. Adelt, J. U. Baenziger, S. Wilson, and H. van Halbeek, J. Biol. Chem., 263

96. N. Bernard, R. Engler, G. Strecker, J. Montreuil, H. van Halbeek, and J. F. G. Vliegenth-

97. G. Spik, B. Coddeville, G. Strecker, J. Montreuil, E. Regoeczi, P. A. Chindemi, and J. R.

98. C. P. C. Soh, A. S. R. Donald, J. Feeney, W. R. J. Morgan, and W. M. Watkins, Glycocon-

99. G. Weisshaar, J. Hiyama, G. C. Renwick, and M. Nimtz, Eur. J. Biochem., 195 (1991)

100. A. V. Savage, C. M. Donoghue, S. M. DArcy, C. A. M. Koeleman, and D. H. van den Eijnden, Eur. J. Biochem., 192 (1990) 427-432.

101. A. V. Savage, P. L. Koppen, W. E. C. M. Schiphorst, L. A. W. Trippelvitz, H. vanHalbeek, J. F. G. Vliegenthart, and D. H. van den Eijnden, Eur. J. Biochem., 160 (1986) 123- 129.

102. P. Scudder, P. W. Tang, E. F. Hounsell, A. M. Lawson, H. Mehmet, and T. Feizi, Eur. J. Biochem., 157 (1986) 365-373.

103. E. F. Hounsell, J. Feeney, P. Scudder, P. W. Tang, and T. Feizi, Eur. J. Biochem., 157

104. G. H. Cockin, T. N. Huckerby, and I. A. Nieduszynski, Biochem. J., 236 (1986) 921 -924. 105. G. Strecker, J.-M. Wieruszeski, C. Martel, and J. Montreuil, Glycoconjugute J., 4 (1987)

106. Y. Goso and K. Hotta, Biochem. J., 264 (1989) 805-812. 107. J. P. Kamerling, I. Rijkse, A. A. M. Maas, J. A. vanKuik, and J. F. G. Vliegenthart, FEES,

108. G. Pfeiffer, M. Schmidt, K.-H. Strube, and R. Geyer, Eur. J. Biochem., 186 (1989)

109. G. Weisshaar, J. Hiyama, and A. G. C. Renwick, Eur. J. Biochem., 192 (1990) 741 -751. 110. S. Kornfeld, Biochem. SOC. Trans., 18 (1990) 367-374. 11 I . R. Schauer (Ed.), The Siulic Acids. Springer-Verlag, New York, 1982. 112. J. B. L. Damm, H. Voshol, K. Hard, J. P. Kamerling, and J. F. G. Vliegenthart, Eur. J.

I 13. D. H. Atha, J.-C. Lormeau, M. Petitou, R. D. Rosenberg, and J. Choay, Biochemistry, 26

1 14. R. U. Lemieux, in K. H. Laidler (Ed.), Frontiers of Chemistry, pp. 3 - 24. Pergamon, New

115. R. U. Lemiuux and K. Bock, Arch. Biochem. Biophys., 221 (1983) 125- 134.

Biochem., 182 (1989) 457-476.

Biophys. Res. Commun.. 110 (1983) 424-431.

Res. Commun., 92 (1980) 1143- 1150.

V. A. Derevitskaya, Eur. J. Biochem., 127 (1987) 21 -29.

3659.

mun., 112 (1983) 482-487.

(1988) 18,253- 18,268.

art, Glycoconjugutes, 1 (1984) 123 - 140.

Rudolph, Eur. J. Biochem., 195 (1991) 397-405.

jugute J., 6 (1989) 319-332.

257-268.

(1986) 375-384.

329-337.

241 (1988) 246-250.

273-286.

Biochem., 180(1989) 101-110.

(1987) 6454-6461.

York, 198 I.

Page 372: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

OLIGOSACCHARIDE DETERMINANTS OF GLYCOPROTEINS 349

116. E. F. Hounsell, N. J. Jones, H. C. Gooi, T. Feizi, A. S. R. Donald, and J. Feeney,

1 17. P. L. Durette and D. Horton, Adv. Curbohydr. Chem. Biochem., 26 (1971) 49- 125. 11 8. H. Thprgersen, R. U. Lemieux, K. Bock, and B. Meyer, Can. J. Chem., 60 (1982) 44-57. 1 19. R. U. Lemieux, T. C. Wong, J. Liao, and E. A. Kabat, Mol. Zmmunol., 2 1 ( 1984) 75 1 - 759. 120. U. Spohr, 0. Hindsgaul, and R. U. Lemieux, Can. J. Chem., 63 (1985) 2644-2658. 121. U. Spohr and R. U. Lemeiux, Curbohydr. Res., 174 (1988) 21 1-237. 122. M. Biswas and V. S. R. Rao, Int. J. Quantum Chem., 20 (1981) 99- 121. 123. A. Kobata and V. Ginsburg, J. Biol. Chem., 244 (1969) 5496-5502. 124. D. F. Smith, D. A. Zopf, and V. Ginsburg, in Methods in Enzymology, vol. 50, pp.

125. A. Kobata, K. Yamashita, and Y. Tachibana, in Methods in Enzymology, Vol. 50, pp.

126. A. S. R. Donald and J. Feeney, Curbohydr. Res., 178 (1988) 79-91. 127. H. Sabharwal, B. Nilsson, M. A. Chester, S. Sjoblad, and A. Lundblad, Mol. Zmmunol., 2 I

128. H. Sabharwal, B. Nilsson, M. A. Chester, F. Lindh, G. Gronberg, S. Sjoblad, and A.

129. H. Sabharwal, B. Nilsson, G. Gronberg, M. A. Chester, J. Dakour, S. Sjoblad, and A.

130. G. Strecker, J.-M. Wieruszeski, J.-C. Michalski, and J. Montreuil, Glycoconjugute. J., 6

13 1. J.-M. Wieruszeski, J.-C. Michalski, J. Montreuil, and G. Strecker, Glycoconjugute, J., 7

132. N. Platzer, D. Davoust, M. Lhermitte, C. Bauvy, D. M. Meyer, and C. Derappe, Curbo-

133. P. R. Rosevear, H. A. Nunez, and R. Barker, Biochemistry, 21 (1982) 1421- 1431. 134. K. Bock, PureAppl. Chem., 55 (1983) 605-622. 135. Z.-Y. Yan and C. A. Bush, Biopolymers, 29 (1990) 799-811. 136. J. Breg, D. Romijn, J. F. G. Vliegenthart, G. Strecker, and J. Montreuil, Curbohydr. Res.,

137. A. D. French and J. W. Brady (Eds.), Computer Modelling of Carbohydrate Molecules, ACS Symposium Series No. 430. American Chemical Society, Washington, D.C., 1990.

138. S. W. Homans, Biochemistry, 29 (1990) 9100-91 18. 139. D. A. Cumming and J. P. Carver, Biochemistry, 26 (1987) 6664-6676. 140. H. C. Gooi, T. Feizi, A. Kapadia, B. B. Knowles, D. Solter, and M. J. Evens, Nature

I4 I . E. F. Hounsell, H. C. Gooi, and T. Feizi, FEBS Lett., 13 1 (1 98 1) 279 - 282. 142. H. C. Gooi, S. J. Thorpe, E. F. Hounsell, H. Rumpold, D. Kraft, 0. Forster, and T. Feizi,

143. H., C. Gooi, E. F. Hounsell, I. Lax, R. M. Kris, T. A. Libermann, J. J. Schlessinger, J. D.

144. J. Finne, M. E. Breimer, G. C. Hansson, L A . Karlsson, J. Leffler, J. F. G Vliegenthart,

145. T. Feizi, Trends Biochem. Sci., 16 (1991) 84-86. 146. J. Breg, L. M. J. Kroon-Batenburg, G. Strecker, J. Montreuil, and J. F. G. Vliegenthart,

147. B. Bechtel, A. J. Wand, K. Wroblewski, H. Koprowski, and J. Thurin, J. Biol. Chem., 265

148. A. G. Morrell, G. Gregoriades, I. H. Scheinberg, J. Hickman, and G. Ashwell, J. Biol.

149. E. F. Neufeld and M. J. Cantz, Ann. N.Y. Acud. Sci., 179 (1971) 580-587.

Curbohydr. Res., 178 (1988) 67-78.

221 -226. Academic Press, San Diego, 1978.

2 16 - 220. Academic Press, San Diego, 1978.

(1984) 1105-1 112.

Lundblad, Curbohydr. Res. 178 (1988) 145- 154.

Lundblad, Arch. Biochem. Biophys., 265 (1988) 390-406.

(1989) 271-284.

(1990) 13-26.

hydr. Rex, 191 (1989) 191-207.

183 (1988) 19-34.

(London), 292 (1981) 156- 158.

Eur. J. Zmmunol.. 13 (1983) 306-312.

Sato, T. Kawamoto, J. Mendelsohn, and T. Feizi, Biosci. Rep., 5 (1985) 83-94.

and H. van Halbeek, J. Biol. Chem., 264 (1989) 5720-5735.

Eur. J. Biochem., 178 (1989) 727-739.

(1990) 2028-2037.

Chem., 246 (1971) 1461-4.

Page 373: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

350 ELIZABETH F. HOUNSELL

150. F. Michon, J.-R. Brisson, and H. J. Jennings, Biochemistry, 26 1987) 8399-8405. 151. R. Yamasaki, Biochem. Biophys. Res. Commun., 154 (1988) 159-164. 152. A. Gottschalk, G. Belyavin, and F. Biddle, in A. Gottschalk (Ed.), Glycoproteins: Their

Composition, Structure and Function, BBA Library Vol. 5, Part B, pp. 1082-1096. Elsevier, Amsterdam, 1972.

153. N. K. Sauter, M. D. Bednarski, B. A. Wunburg, J. E. Hansson, G. M. Whitesides, J. J. Skehel, and D. C. Wiley, Biochemistry, 28 (1989) 8388-8396.

154. L. M. Loomes, K.4. Uemura, R. A. Childs, J. C. Paulson, G. N. Rogers, P. R. Scudder, J.-C. Michalski, E. F. Hounsell, D. Taylor-Robinson, and T. Feizi, Nature, 307 (1984) 560-563.

155. D. Roelcke, R. Brossmer, and W. Riesen, Scand. J. Zmmunol., 8 (1978) 179- 185. 156. K. Uemura, D. Roelcke, Y. Nagai, and T. Feizi, Biochem. J., 219 (1984) 865-874. 157. H. Niemann, K. Watanabe, S. Hakomori, R. A. Childs, and T. Feizi, Biochem. Biophys.

158. E. A. Kabat, J. Liao, M. H. Burzynska, C. T. Wong, H. Thngerson, and R. U. Lemieux,

159. H. Higashi, M. Naiki, S. Matuo, and K. Okouchi, Eiochem. Biophys. Res. Commun., 79

160. J.-R. Brisson and J. P. Carver, Biochemistry, 22 (1983) 3671-3680. 161. J.-R. Brisson and J. P. Carver, Biochemistry, 22 (1983) 3680-3686. 162. D. A. Cumming, D. S. Dime, A. A. Grey, J. J. Krepinsky, and J. P. Carver, J. Biol. Chem.,

163. D. A. Cumming, R. N. Shah, J. J. Krepinsky, A. A. Grey, and J. P. Carver, Biochemistry,

164. S. W.Homans,R.A.Dwek,andT. W. Rademacher,Biochemistry, 26(1987)6553-6560. 165. S. W. Homans, Biochemistry 28 (1990) 91 10. 166. A. Imberty, S. Gerber, V. Tran, and S. Perez, Glycoconjugate J., 7 (1990) 27-54. 167. C. J. Edge, U. C. Singh, R. Bazzo, G. L. Taylor, R. A. Dwek, and T. W. Rademacher,

168. M. Hricovini, R. N. Shah, and J. P. Carver, Biochemistry 31 (1992) 10018. 169. C. A. Bush, V. K. Dua, S. Ralapati, C. D. Warren, G. Spik, G. Strecker, and J. Montreuil,

170. A. J. Duben and C. A. Bush, Arch. Biochem. Biophys., 225 (1983) 1 - 15. 17 1. E. Berman, U. Dabrowski, and J. Dabrowski, 176 ( 1988) 1 - 15. 172. R. L. Brockbank and H. J. Vogel, Biochemistry, 29 (1990) 5574-5583. 173. P. J. Bjorkman, M. A. Shaper, B. Samraoui, W. S. Bennett, J. L. Strominger, and D. C.

174. H. Paulsen, Angew. Chem. Znt. Ed. Engl., 29 (1990) 823-839. 175. R Prohaska, T. A. W. Koerner, Jr., I. M. Armitage, and H. Furthmayr, J. Bid. Chem., 256

176. E. J. Welsh, D. Thom, E. R. Moms, and D. A. Rees, Biopolymers, 24 (1985) 2301 -2332. 177. G. Pepe, Y. Oddon, D. Sin, J. P. Reboul, and A. A. Pavia, Carbohydr. Rex, 209 (1991)

178. A. Bush and R. E. Feeney, Int. J. Peptide Protein Res. 28 (1986) 386-397. 179. N. J. Ringler, R. Selvakumar, H. D. Woodward, I. M. Simet, V. P. Bhavanandan, and

180. T. A. Gerken, K. J. Butenhof, and R. Shogren, Eiochemistry, 28 (1989) 5536-5543. 18 1. M. Kuwano, T. Seguchi, and M. Ono, J. Cell Science, 98 ( 199 1) 13 1 - 134. 182. H. Matsuura, T. Greene, and S. Hakomori, J. Biol. Chem., 264 (1989) 10,472- 10,476. 183. G. W. Hart, R. S. Haltiwanger, G. D. Holt, and W. G. Kelly, Annu. Rev. Biochem., 58

184. I. A. King and E. F. Hounsell, J. Biol. Chem., 264 (1989) 14022- 14028.

Res. Commun., 81 (1978) 1286-1293.

Mol. Zmmunol., 18 (1981) 873-881.

(1977) 388-395.

261 (1986) 3208-3213.

26 (1987) 6655-6663.

Biochemistry, 29 (1990) 1971 - 1974.

J. Biol. Chem., 257 (1982) 8199-8204.

Wiley, Nature, 329 (1987) 506-518.

(198 1) 578 1-5791.

67-81.

E. A. Davidson, Biochemistry, 26 (1987) 5322-5328.

(1989) 841-74.

Page 374: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

AUTHOR INDEX FOR VOLUME 50

Numbers in parentheses are reference citation numbers. Complete references are listed in numerical order at the end of each chapter in which they are cited.

A

Abernethy, K., 259(245) Achiwa, K., 21 1(18), 252(214) Ackerman, S. K., 259-260(251) Adachi, S., 317(80), 321(80) Adams, G. A., 217(56) Adelstein, E., 316(64), 321(64), 329(64) Adelt, G., 329(95) Adermann, K., 296(77-78) Adner, N., 230(141) Ahamed, N. M., 230(145), 235(145) Aida, K., 231(155) Akamatsu, Y., 252(217) Akiyama, Y., 220( 103) Alais, J., 73(159), 78(159), 81(159), 84(159) Alekseeva, V. G., 139(59) Aleshin, A., 18 - 19(49) Alexeev, Y. E., 136(50-51) Ali, M. H., 143- 144(72) Men, I. E., 259-260(250) Alonso, D., 168(207), 184(207,240) Altmann, K., 218(68), 222-223(68),

225(68), 228(68), 241(68), 251(68), 261(68)

Alving, C. R., 21 l(10) Amatore, C., 23(36) Ammann, H., 73( 156), 78( 156), 82( 156) Amvam-Zollo, P. H., 52(109) Anderson, B., 316(59) Anderson, B. L., 196(278) Anderson, D. M. W., 3 1 l(7) Anderson, L., 218(63), 226(63), 244(63),

Anderson, M. S., 240(191), 262(191) Anderson, R. G., 183- 184(237), 187(237) Anderson, R., 230(141) Ando, T., 16(40,41) Angel, A. S., 313(27) Anisfeld, S. T., 279( 12) Anzeveno, P. B., 43(80)

Apicella, M. A., 212(22) Aplin, R. T., 154(125) Appelmelk, B. J., 259(244, 244a) Arai, M., 252(217)

249(207)

ApU, H.-J., 41(90)

Arata, S., 220( 103), 232( 158), 259(247) Arbatsky, N. P., 325(91) Arizuka, M., 49(108) Armitage, I. M., 215(65), 218(63, 65, 66),

222(66), 226(63), 244(63), 249(66), 250-251(65), 263(65), 344(175)

Asano, K., 149(95), 150(96-98) Axhauer, H., 211(13), 252(13), 262(13) Ashwell, G., 338(148) Aspinall, G. O., 41(79), 105(181) Atha, D. H., 331(113) Attwood, S. V., 190(254) Auge, C., 305(91) Auling, G., 219(100) Autenrieth-Ansoge, L., 191(264)

B

Bacquet, C., 22-23(23) Badoud, R., 183- 184(237), 187(237) Baek, N. I., 104-105(178), 109(178) Baenziger, J. U., 329(95) Baggett, N., 125(4), 13 l(32) Baird, P. D., 197(282) Ballesteros, M., 188(253) Bandzouzi, A., 145(77-78) B2r, T., 28(70), 41(70,87,90), 43(87), 49(70) Barbero, G. J., 316(64), 321(64), 329(64) Barbour, A. G., 232(157) Barker, R., 334( 133) Barnickel, G., 212(25), 214(25), 253-

254(220), 262(220) Barrett, A. C. M., 153(120) Barrett, A. G. M., 153(121), 168(204-205),

189(205), 190(254) Barthels, R., 287(51) Barton, D. H. R., 195(273) Barzilay, I., 215(69), 218(69), 221(69),

223(69), 227(69), 249(69) Basu, S., 241(193) Batley, M., 222-223(109), 226(109) Batllori, R., 188(253) Bauer, C. J., 321(82), 325(82) Baumeister, L., 160( 160) Baumgartner, J.-D., 260(253) Bauvy, C., 333( 132)

35 1

Page 375: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

352 AUTHOR INDEX, VOLUME 50

Bazzo, R., 343( 167) Beau, J.-M., 110(186), 294(72), 296(72) Bechtel, B., 337( 149) Becker, D. P., 184(242) Bednarik, K., 252(218) Bednarski, M. D., 303(88), 339(153) Beer, D., 157(135-136) Beetz, T., 211(12), 252(12) Behrendt, M., 32(62), 35-36(62), 38(62),

39(78), 41(62), 58(62), 61(62), 11 1(78), 114(78)

Belanger, P., 192(266) Belleau, B., 279(17), 283(17) Belyavin, G., 339( 152) Belzecki, C., 190(260), 191(261-263) Bencomo, V., 287(42a, 42b) Bendlin, H., 160( 160) Enkchie, M., 195(273) Benedict, C., 129(25) Bennett, R. B., 151(105-106) Bennett, W. S., 343( 173) Bentley, R., 313(19) Berend, G., 127( 17) Berger, H. J., 259-260(250) Bergmann, I. M., 277( 1) Bergmann, M., 14(35), 169(213) Berman, E., 314(35), 343(171) Bernard, N., 329(96), 333(96) Berry, D., 230-231(144) Berven, L. A., 161(172) Beutler, B., 238( 182), 264(182) Bezuidenhoudt, B. C. B., 153(121) Bhat, K. L., 125(2), 182(2) Bhat, U. R., 218(67), 219(67, 98, IOl),

221(107), 227(124, 126), 231(152), 232(101, 160), 247(101, 160)

Bhattachanya, S., 151(109) Bhattachajee, A. K., 250(208) Bhattacharjee, S. S., 158- 159(150) Bhatti, T., 3 13(20) Bhavanandan, V. P., 344(179) Biddle, F., 339( 152) Bielefeldt, T., 302(83), 306(95) Bigelow, S. S., 22(28) Bigham, E. C., 129(25) Bigley, D. B., 159( 155) Bigorra, J., 188(252) Birken, S., 324(85) Bimbach, S., 294-295(73), 297-298(73)

Bischofberger, K., 146(81), 147(82) Bishop, D. G., 229(134), 234(134) Biswas, M., 333(122) Bjorkman, P. J., 343( 173) Bjomdal, H., 31 l(9) Blache, D., 218(61) Blanz, P., 247(203) Bleha, T., 35(76) Bock, K., 17(44,45), 18(44), 110(187),

134(41-44), 135(45), 158(42, 43), 160(42), 169(210), 173(44,223), 174(41-43,226), 175(226,228), 176(229), 282(25), 288(25), 302(25,

332(115), 333(115, 118), 334(134) 82-83), 306(95-99), 314(34), 327(34),

Boekema, E., 151(110) Boel, E., 18(51) Boerekamp, J., 127(15), 182(15) Boersma, A., 316(60-63), 321(60-63),

Bogard, W. C., Jr., 259(245) Boivin, A., 213(27) Bols, M., 135(46, 47), 136(48) Bommer, R., 28(70), 41(70,92), 47(92),

49(70), 81(92, 164), 85(92) Boons,G. J. P. H., 71(151),73(151),

144(75) Borchardt, R. T., 192(265), 196(265,277,

278) Borcherding, D. R., 192(265), 196(265,

Borowiak, D., 217(59), 218(91), 219(99),

Boschetti, A., 140(66)

Bourquelot, E., 16(39) Bouwstra, J. B., 60(126), 63(126), 66,67(126) Boylan, D. B., 230(149) Bradaczek, H., 212(25), 214(25), 253(220-

Brade, H., 211(5-6, 8, 9a, 9b), 212(6, 23- 25), 214(25), 218(66a, 67,68,75), 2 19(67), 222,223(68), 224(66a), 225(68, 1 16, 1 18), 228(68), 229( 132,

238(167, 169, 183, 185), 241(68), 251(68,75,213), 256(6, l18,229d), 257(5, 116, 118, 167, 168,234,239), 258(116, 167-168, 183, 185, 185a,

333(62)

277-278)

224(91,99), 225(91), 235(91)

Botta, M., 187(248-249)

222), 262(220-221)

133), 231(154), 236(5, 118, 167-170),

Page 376: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

AUTHOR INDEX, VOLUME 50 353

241), 259(6,242-244,244a), 261(68, 75, 118,269), 263(185), 264(133, 167, 169, 183,266)

Brade, L., 21 l(5, 6,9), 212(6), 225(118), 229(133), 236(5, 118, 170, 172), 243(196), 251(213), 252(196), 256(6, 118), 257(5-6, 116, 118,234,239), 258(116, 118, 172), 259(6,242-244, 244a), 26 1( 1 18), 264( 133)

Brady, J. W., 334( 137), 343( 137) Brandenburg, K., 21 1(5), 236(5), 254(224-

226), 257(5) Brandt, E., 238( 185a) Braum, G., 307(104) Braun, P., 287(36), 303(46, 86), 307(103-

Bredereck, H., 24(38) Breg, J., 316(65-68), 321(65-68), 334(136),

Breimer, M. E., 335( 144) Breitenbach, R., 126(1 I), 149(11) Brenken, M., 292-293(69) Brennan, P. J., 105(181)

Brimacombe, J. S., 127(19), 311(8) Brinbach, S., 287(44, 52), 288(44), 294,

Briner, K., 70( 147), 114(21 I), 115(211) Brisson, J.-R., 288(57), 339(150), 343( 160,

Broady, K. W., 218(86), 225(86), 228(187),

104)

337( 146)

Brill, W., 278(10- 1 I), 284(10), 286(10, 11)

295(44), 297-298(44)

161)

229(86), 238-240(187), 243(187), 246(86), 247( 187)

Brockbank, R. L., 343(172) Brockhausen, I., 58( 129), 60( 129), 66,

Brossmer, R., 339(155) Brown, H. C., 159(155) Bruce, I., 200(286) Bruneteau, M., 218(61), 225-226(121) Bryn, K., 225(119), 229(119), 231(151) Buchanan, J. G., 125(4) Buchholz, M., 287(45,45a), 288(45) Buchner, E., 5(12) Buck, K. W., 131(32) Bulusu, A. R. C. M. Burlingame, A. L., 3 13(24) Burton, A. J., 217(53) Burzynska, M. H., 339(158)

67(135), 294(76)

Busch, M., 219(101), 232(101, 160), 247(101, 160)

Bush, A., 344(178) Bush, C. A., 232(162), 235-236(162),

264(162), 316(56, 58), 334(135), 343( 169- 170), 344(170)

Butenhof, K. J., 344( 180)

C

Campos-Portuguez, S. A., 219( 101), 232(101), 238(185a), 247(101)

Camps, P., 182(234,236), 183(236, 238), 184(238)

Cantz, M. J., 338(149) Capek, P., 41(79) Capobianco, J. O., 2 13(26), 262(26) Capon, C., 316(76), 321(76), 325(76), 329-

Card, P. J., 190(257) Cardellach, J., 182(235,236), 183(236,

Cans, R.C. H. M., 167(197),200(197) Carlson, D. M., 322(88) Carlson, R. W., 221(106, 107), 232(106,

Carlstedt, I., 3 13( 15) Caroff, M., 214(42,44), 218(93), 222(11 I),

Carpenter, N. M., 154(125), 155(126) Carpenter, R. C., 41(79) Carruthers, R. A., 3 14(45), 3 15(46),

330(76)

238-239), 184(238,240,241)

160), 247(160), 261(271)

238(93)

316(75), 321(75), 325(45,46,75), 328(75)

Carter, H. E., 217(53) Carver, J. P., 335( 139), 343( 139, 160- 163,

Cashmore, G. C., 313(26), 316(74, 75), 168)

321(74, 75, 82), 323(74), 325(75, 82), 328(74,75)

Cavaillon, J. M., 258(240), 264(240) Cenci di Bello, I., 200(286, 288) Cha, J. K., 139(60) Chaby, R., 230-231(139), 242(139),

259(246) Chai, W., 313(26,28), 315(46), 321(82),

323(28), 325(46, 82) Chaki, H., 116(217), 252(216)

Page 377: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

354 AUTHOR INDEX. VOLUME 50

Chambers, R. E., 313(20) Chan, A., Jr., 161(166) Chandrasekaran, E. V., 329(93) Chapleur, Y., 145(76-78) Charon, D., 211(17), 212(24), 252(17),

Chattejee, D., 105(181) Chen, L., 214(43), 232(162), 235-236(162),

264( 162) Chen, S.-Y., 125(2, 5,6), 126-127(6),

168(6), 174(6), 182(2), 184(6) Chester, M. A., 333(127- 129) Chia, J., 259(249) Childs, R. A., 322(87), 339(154, 157) Chindemi, P. A., 329(97), 333(97) Chittenden, G. J. F., 126(7), 127(7, 15, 21,

22), 149(22), 167(197), 169(21 I), 182(15), 183(21 I), 200(197)

Chmielewski, M., 190(255,259,260),

Choay, J., 33 I( 1 13) Christensen, M., 306(97) Christensen-Brams, I., 306(98) Christian, R., 218(64,70), 249(70), 250(64),

Chu, D. T. W., 290(65) Chucholowski, A., 2 1( lo), 1 10( 188) Ciommer, M., 291(67), 294-296(67),

Ciucanu, I., 313(12) Clamp, J. R., 3 13(20) Clarke, A. J., 19(52) Clarke, C. T., 169(2 12) Cockin, G. H., 329-330(104) Coddeville, B., 329(97), 333(97) Coffen, D. L., 196(280) Cohen-Anisfeld, S. T., 306(94) Cole, L. A., 324(85) Cole, R. B., 218(57a, 57b), 228(57a) Coleman, D. L., 256(229c) Collin, W. R., 127(20), 133(20), 199(20) Collins, P. M., 143- 144(72) Conrad, H. E., 31 l(6) Conrad, R. S., 23 I( 152) Copeland, C., 127(14) Corbera, J., 183- 184(238), 188(253) Cornforth, J. W., 4-5(9b), 19(56) Cotter, R., 218(94), 228(94), 241(94) Cotter, R. J., 214(33, 36, 43), 218(88),

259(246)

191(261-263)

251(70), 261(70)

302(8 1)

222(1 lo), 228(33, 88), 232(88, 162),

234(88), 235(88, 162), 236(162), 242(33), 243(88), 249( 1 lo), 25 1( 1 lo), 264( 162)

Coward, J. K., 307(106, 107) Cram, D. J., 3(6) Crane, A.M., 105(181) Crawford, T. C., 125(1), 126(9, 1 l), 149(11) Cree, G. M., 164(183), 311(7) Creemer, L. J., 43(80) Cretcher, L. H., 151(104) Crum, J. D., 151(106) Csuk, R., 137(53), 138(54), 144(53, 54) Cumming, D. A., 335( 139), 343( 139),

Czuk, R., 144(73) 343( 162 - 163)

D

Dabrowski, J., 317(81), 321(81), 324(81),

Dabrowski, U., 317(81), 321(81), 324(81),

Dachman, J., 19 l(264) Dahler, S. A., 200(286-287) Dakour, J., 333(129) Dalchau, R., 322(87) Dalla Venezia, N., 225-226(121) Damm, J. B. L., 329(112), 333(112) Danbara, H., 231(156) Daniel, J. K., 43(80) Danilov, B., 165(188) Danilova, G. A., 169(208) Dapperens, C. W. M., 169(21 I), 183(211) David, C. M., 218(57b) Davidson, D. A., 344(179) Davies, M. J., 3 15(46), 325(46) Davoust, D., 333(132) Dax, K., 168(203) de Bello, I. C., 127(20), 133(20), 199(20) de Bruyn, R. G. M., 167(197), 200(197) de Lederkremer, R. M., 148(88,89),

343(171)

343( 17 1)

160(158, 161, 162), 163(181), 164(182),

200), 168(194, 202), 169(181), 170(181,

172(194, 202), 173(194, 200, 222, 224), 174(225), 179(230), 180(231)

De Rouville, E., 127(22), 149(22)

165(191), 166(192- 196), 167(198-

194), 171(182,214-218,220,221),

Page 378: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

AUTHOR INDEX, VOLUME 50 355

Deagostini-Bazin, H., 329(94) Deak, G., 163(180) Dean, B., 99(171) Deferrari, J. O., 148(88) Dejsirilert, S., 25 l(2 1 1) Dell, A., 3 13(23), 324(84), 329(84) Dellinger, R. P., 259-260(250) DeLucca, A. J., 218(57b) Demel, R. A., 255(229) Dempsey, A. M., 148(87) Deprun, C., 214(42,44) Derappe, C., 333( 132) Dervitskaya, V. A., 282(24), 325(91) Devitt, R. E., 116(215) Dhanak, D., 153(120, 121) Dho, J. C., 197(281,282) Diamantstein, T., 264(266) Dienst, C., 317(78), 321(78) Dijong, I., 149(90) Dill, K., 314(35) Dimant, E., 138(55) Dime, D. S., 343( 162) Din, Z. Z., 256(229c) Dinarello, C. A., 236(167), 238(167), 257-

258(167), 264(167) Diolez, C., 211(17), 252(17) Dipadova, F., 2 1 l(9a) Dmitriev, B. A., 218(73, 77), 224(77),

225(73, 77), 226-227(73), 228- 229(77), 231(77), 242(73, 77), 243- 245(77), 248(73, 77), 251(73, 77), 264(77)

Doerfler, M. E., 263(261) Doherty, D. G., 277(2) Dolle, R. E., 21(10) Dombo, B., 300(79-80), 301(79) Domelsmith, L. N., 218(57a, 57b), 228(57a) Donald, A. S. R., 329(98), 332( 116),

333(98, 116, 126), 335(116) Doniec, A,, 161(169) Doran, C. C., 213(26), 262(26) Dorland, L., 314-315(31, 32), 316(61),

Draft, D., 335( 142) Drew, M. G. B., 187(25 1) Drewry, D. T., 218(76) Drozanski, W., 237(177, 178) Du Mortier, C., 166(195), 171(214, 221),

174(225), 179(230) Dua, V. K., 3 16(56, 58), 343( 169)

321(61), 325(31, 32,91), 333(31, 32)

Dube, V. E., 316(58) Duben, A. J., 343 - 344( 170) Dubos, R., 4-5(9a) Dunn, D. L., 259(245) Dupuis, G., 73(156), 78(156), 82(156) Durette, P. L., 333(117) Diirrbaum, I., 236(167), 238(167), 257-

258(167), 264(167) Diirrbaum-Landmann, I., 238( 185a) Dutton, G. G. S., 31 I( 10) Dutton, G. J., 11 l(191) Dwek, R. A., 61(136), 225(120), 314(42),

Dyong, I., 158(151-153), 160(159, 160), 325(42), 343(42, 164, 167)

162( 178)

E

Eby, R., 23(34) Edge, C. J., 343(167) Edwards, J. R., 230(148) Effenberger, G., 114(207,208) Egan, A., 227( 123), 244( 123) Egawa, K., 220( 103), 259(247) Egge, H., 313(13), 317(78, 81), 321(78, 81),

Eidels, L., 238(181), 264(181) Eitelman, S. J., 147(82), 148(83) El Khadem, H. S., 153(119) El Shenawy, H. A., 155(132) Eliel, E. L., 114(202) Elsbach, P., 263(261) Emmerling, W. N., 152( 1 13) Emoto, S., 163(179) Eng, J., 225(119), 229(119) Engler, R., 329(96), 333(96) Enkelmann, V., 152(114) Emst, M., 238(185a) Erwin, A. L., 214(45), 229(45), 238(45) Esswein, A., 25(47,48), 32(59) Estopa, C., 182(235) Eustache, R. J. Evans, M. E., 259(249) Evens, M. J., 335( 140) Ezaki, T., 251(211)

324(81)

F

Faas, M., 111(197) Fabre, J. W., 322(87) Fairbanks, A. J., 200(286)

Page 379: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

356 AUTHOR INDEX. VOLUME 50

Fangmann, R., 317(78), 321(78) Fatah, M. Y., 125(4) Feeney, J., 316(73-75), 321(73-75,82),

323(74), 324(73, 89), 325(73, 75, 82),

331(103), 332(116), 333(98, 116, 126), 335( 1 16)

328(73-75), 329(98, 103), 330-

Feeney, R. E., 344(178) Feist, W., 236(168, 169), 238(183, 185,

185a), 257(168), 258(168, 183, 185), 263( 185), 264( 169, 183,266)

Feizi, T., 313(26, 28,29), 314(41), 316(51,

75, 82), 322(87), 323(74), 324(73),

331(102, 103), 332(51, 116), 333(116),

338(145), 339(51, 154, 156, 157), 343(41)

241(41)

53,59, 73-75), 317(51), 321(53,73-

325(41, 73,75, 90), 328(73-75), 329-

335(51, 116, 140-143), 337(51, 145),

Fenselau, C., 214(41), 236(41), 239(41),

Ferguson, M. A. J., 225(120) Fernhndez Cirelli, A., 132(36, 37), 133(39),

148(88, 89), 160(158), 164(158), 165( 19 l), 166( 196), 167(200),

172(202), 173(200, 222, 224) Ferrari, B., 287(47), 294(47), 294(75) Femer, R. J., 22(20) Fesik, S. W., 215(65), 218(65,66), 222(66),

Fktizon, M., 165(187) Feudenberg, M. A., 236( 170, 172) Fieser, L. F., 151(109) Fieser, M., 151(109) Fiil, N. P., 18(51) Fink, M. P., 259-260(250) Finne, J., 329(94), 335(144) Finne, V., 329(94) Firsov, L. M., 18 - 19(49) Fischer, E., 2(3,4), 4(4), 5(4, lo), 6(18-23),

168(202), 17 l(216 - 2 18,220),

249(66), 250-251(65), 263(65)

7(3,20-22), 8(4,27-29), 9- 10(30), 11(31), 12(30-32), 13(30, 32-34), 14(3, 32-36), 16(37), 19(37), 155(130), 157(137, 138)

Fischer, W., 215(48) Fisher, C. J., 259-260(250) Fitzsimmons, B. J., 183- 184(237), 187(237) Flad, H.-D., 236(167- 169), 238(167, 169,

183, 185, 185a), 257(167, 168),

258(167, 168, 183, 185,239), 263(185), 264(167, 169,266)

125), 155(126), 193(267), 196(279), Fleet, G. W. J., 127(20), 133(20), 154(122,

197(281-283), 198(285), 198( 122), 199(20), 200(286 -288)

Fletcher, H. G., Jr., 164( 186), 165( 188, 189) Fluharty, A. L., 160(163) Fomsgaard, A., 259-260(252) Font, J., 168(207), 182(234-236), 183(236,

238-239), 184(207,238,240-241), 185(243), 186(245), 188(252-253)

Ford, C., 19(53) Ford, M. J., 116(214) Foridis, C., 329(94) Forster, M. O., 8(26) Forster, O., 335(142) Foster, A. B., 131(32), 217(55) Foulke, G. E., 259-260(250) Fournet, B., 313(16), 316(76), 321(76),

Fowler, V. J., 247(203) Fox, G. E., 247(203) Fracsch, C. E., 218(73), 225-227(73),

Franken, G. A. M., 169(2 1 l), 183(2 1 1) Fraser-Reid, B., 116(215) Frasman, G. D., 282(26), 288(26), 302(26) French, A. D., 334( 137), 343( 137) Frenkiel, T. A., 324(89) Freudenberg, K., 2(5), 15(5) Freudenberg, M. A., 21 1(3), 215(47),

325(76), 329-330(76)

242(73), 248(73), 251(73)

241(47), 243( 196), 244(3), 247(234), 252(172, 196), 256-257(3), 258(241), 259-260(252), 264(265)

Frick, W., 114(204) Friedrich-Bochnitschek, S., 11 I( 194),

Friffiss, J. M., 2 12(22) Fromme, I., 224( 114) Frush, H. L., 157(143), 158(146) Fuganti, C., 159( 154) Fugedi, P., 22(18), 23(35), 35-37(71),

291(68), 294(68), 297(68)

60( 124), 105( 124), 108( 124), 248(204), 290(63b)

Fuhrhop, J.-H., 15 1 ( 1 10) Fujimoto, K., 1 lO(184) Fujishima, Y., 211(11, 16), 252(11, 16) Fujita, M., 211(16), 252(16) Fukase, K., 211(15), 215(15), 252(15),

Page 380: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

AUTHOR INDEX, VOLUME 50 357

258( 15), 263( 15), 306(99) Fukuda, M., 324(84), 325(90), 329(84) Fukuoka, S., 218(95), 228(95), 243(95) Funaki, Y., 49(105) Funakoshi, I., 317(77), 321(77) Fiirstner, A., 194(272) Furthmayr, H., 344( 175) Furuhata, K., 138(56-57)

G

Gaden, H., 28(61), 30(56), 32(56, 61) Gage, D. A., 261(271) Galanos, C., 21 l(1-5), 213(2,4), 214(35),

2 15(47), 2 17( 1, 35), 2 18( 1,4,64a), 224(64a), 225(4), 228(4, 35), 231(152), 231(154), 233(164), 234(1, 164),

241(47), 243( 196), 244(3), 246(4), 247(4, 35), 249(35), 252(35, 172, 196),

235(164), 236(5, 170-172), 237(178),

256(1, 3,4,230), 257(1, 3-5,230,232, 234), 258(241), 259-260(262)

Galbraith, L., 234(163) Galkowski, T. T., 157( 143)

Gammon, D. W., 105(181) Ganguly, A. K., 162(175-177) Garcia, F., 261(271) Garegg, P. J., 22(18, 24), 23(35), 126-

Gall-Istok, K., 163( 180)

127(8), 290(63b)

293(70), 306( 102) G-, H. G., 278-279(6), 283(28-30),

Gasiecki, A. F., 153(121) Gateau, A., 129- 130(27), 198(284) Gaudemer, A., 198(284) Gautheron, C., 305(91) Gerber, S., 343(166) Gerdes, J., 236( 169), 238(169), 264(169,266) Gerisch, G., 237(179) Gerken, T. A., 344( 180) Gero, S. D., 129- 130(27), 198(284) Gerwig, G. J., 3 13(2 1) Geyer, H., 314(40), 325(40) Geyer, R., 314(40), 325(40), 329(108) Gibson, B. W., 212(22) Giesbrecht, P., 212(25), 214(25), 252(219),

253(220), 254(2 19,220), 262(220) Ginocchio, S. D., 161(166) Ginsburg, V., 333(123- 124)

Girard, R., 259(246) Giudici, T. A., 160(163) GIBnzer, B. I., 137(53), 138(54), 144(53, 54,

Glattfeld, J. W. E., 147(144), 151(99) Glittenberg, D., 158(153) Gmeiner, J., 217-218(54), 223(112),

Gnichtel, H., 191(264) Godefroi, E. F., 127(15), 167(197),

73)

249( 112)

169(211), 182(15), 183(211), 200( 197)

Goldman, R. C., 213(26), 262(26) Goldstein, I. J., 151(111, 112) Golenbock, D. T., 238( 184), 264( 184) Golfier, M., 164(185), 165(187)

Goodwin, S., 229(135) Gooi, H. C., 316(73), 321(73), 324-

Golubev, A., 18- 19(49)

325(73), 328(73), 332-333(116), 335(116, 140- 143)

Gorbach, V. I., 218(62) Gorbies, L., 303(84b) Gorelick, K., 259-260(251) Goso, Y., 329(106) Gottert, H., 229(128), 236(128) Gottschalk, A., 339(152) Grabarek, J., 213(31) Graczyk, G., 282(26), 288(26), 302(26) Grage-Griebenow, H., 238(185a) Gragg, C. E., 129(25) Grasselli, P., 159( 154) Gratzl, J. S., 165(190) Gray, G. W., 218(76) Greck, M., 218(82), 230(82), 232(82) Green, E. D., 329(95) Greene, T., 345(182) Gregoriades, G., 338( 148) Greilich, U., 53(113), 55-56(117), 57(113,

117),95(113, 117), lOO(113, 117), 101( 117)

Grey, A. A., 343(162, 163) Grief, V., 143(69) Grimmecke, H.-D., 21 l(9b) Gronberg, G., 333(128, 129) Gruezo, F. G., 316(55) Grundler, G., 68-69(137, 137a, 140),

71(137), 72(140), 73(137, 140),

83( 140), 93( 137), 94( 137, 170), 78(140), 80(137), 81(137, 140), 82-

Page 381: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

358 AUTHOR INDEX, VOLUME 50

98(137), 99(137, 170), lll(192, 193), 113(192, 193)

Guelde, G., 259(249) Guilleman, R., 287(50), 299(50) Guilliard, W., 11 l(198) Giinther, W., 281(23), 302(81) Gupta, R., 247(203) Gutberlet, T., 253(221), 262(221) Guthrie, R. D., 136(49) Guze, L. B., 230(140) Gzernecki, S., 139(62)

H

Haas, M., 214(40) Haeffner, N., 230-231(139), 242(139) Haeffner-Cavaillon, N., 258(240), 264(240) Hafele, B., 194(270-271) Hagelloch, G., 24(38) Hahn, C. M., 247(203) Haishima, Y., 261(268) Hajime, I., 21 l(18) Hakomori, S., 98(174), 99(171), 31 l(4-5),

Hall, C. L., 238( 180) Hall, R. H., 146(81), 147(82), 148(83) Hall, W. R., 129(25) Haltiwanger, R. S., 345( 183) Hamadeh, R., 2 12(22) Hamako, J., 314(41), 325(41) Hambsch, E., 24-25(38) Hampton, R. Y., 238(184), 264(184) Han, O., 171(219) Han, X.-B., 105(183), 110-lll(183) Hancock, R. E. W., 263(258) Hanessian, S., 22-23(23), 141(67),

Hanisch, F.-G., 317(78, 81), 321(78, 81),

Hannigan, J., 259-260(251) Hansen, E. J., 238(181), 264(181) Hansen-Hagge, T., 236(170- 171, 173),

243(173), 244(173, 199) Hansson, G. C., 313(15), 335(144) Hansson, J. E., 339(153) Hara, H., 218(88), 228(88), 232-235(88),

Harada, K., 257(236)

316(52), 325(90), 339(157), 345(182)

181(233), 186(246, 247), 187(248,249)

324(81)

243(88)

Harangi, J., 60(125) Harbin, A.-M., 314(45), 325(45) Hard, K., 314(36), 329(112), 333(112) Hardy, M. R., 3 13(22) Harlan, J. M., 238(186) Harrata, A. K., 218(57a), 228(57a) Hart, G. W., 345( 183) Hw, S., 217(57-58), 218(57, 80,85), 225-

227(85), 231(85)

58(120, 123),211(11, 16, 19),252(11, 16, 19)

Hasegawa, A., 41(93-95), 47(93, 94),

Hasenkamp, T., 293(70) Hashimoto, H., 149(94,95), 150(96) Hashimoto, S., 21(1 I), 116(220-223),

152(116) Hauser, H., 255(229) Haus, H., 127( 18) Hawiger, J., 213(31) Haworth, W. N., 162(173) Hay, R. W., 22(20) Hayashi, J. A., 230(148) Hayashi, M., 21( 1 I ) Hedenburg, 0. F., 148(84) Heermann, D., 11 l(198) Hehre, E. J., 19(54) Heine, H., 238(185, 185a), 258(185),

263( 185) Heinz, E., 28(58), 30(58), 105(58a) Helander, A., 288(57) Helander, I. M., 2 18(68), 222-223(68),

225(68), 227( 125), 228(68), 229( 136), 241(68), 243(195, 195a), 251(68), 26 l(68)

Helferich, B., 14(36) Helfrich, W., 151(110) Heller, D., 214(41), 236(41), 239(41),

Hellerqvist, C. G., 3 1 l(9) Helpap, B., 44(82), 60(128) Hemberger, J., 54(114), 78-79(114),

81(114), 85(114) Hendriks, K. B., 280(20) Hengeveld, J. E., 143(69) Henning, D., I55( 13 1) Henrichson, C., 22(24) Henricson, B. E. Her, G.-R., 213(31) Hermann, P., 303(87)

24 l(4 1)

Haraldsson, M., 127(20), 133(20), 199(20) Hermansson, K., 314(37)

Page 382: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

AUTHOR INDEX, VOLUME 50 359

Herp, A., 316(54), 329(54) Herzbeck, H., 238(183), 258(183), 264(183,

Heume, M., 62(133) Hewett, M. J., 229( 134), 234( 134) Heymann, H., 15 1( 109) Hickman, J., 338(148) Higashi, H., 339( 159) Hignett, R. C., 23 1( 153) Hildebrandt, J., 211(13), 252(13), 262(13) Hilgenfeld, R., 152(114) Hindahl, M. S., 234( 163) Hindsgaul, O., 333(120) Hiraoka, T., 252(217) Hirata, Y., 151(109) Hirayama, T., 232(158) Himhberger, J., 8(27,28) Hirvas, L., 243( 195) Hisatsune, K., 218(83), 251(83), 261(83,

268), 264(264) Hiyama, J., 329(99, 109) Hjort, I., 18(5 1) Hoch, M., 28(58a), 30(58a), 73(155),

79( 155), 81( 155), 105(58a) Hoesch, K., 1(1), 4(1) Hoffmann, M., 1 1 I ( 198 -200), 1 14( 199,

Hoffmann, R. W., 185(244) Hofstad, T., 2 18(80) Hogenkamp, P. C., 132(35) Holland, P. T., 313(24) Hollingsworth, R. I., 221(106, 107),

232( 106), 261(271) Hollosi, M., 282(26,27), 288(26), 302(26,

27), 303(84a) Holly, F. W., 195(274) Holst, O., 211(9b), 212(23, 24), 218(66a,

91), 224(66a, 91), 225(91, 116),

261(270)

266)

205 -206)

235(91), 251(213), 257-258(116),

Holt, G. D., 345(183) Holt, N. B., 157(143) Holt, S. C., 218(94), 228(94), 241(94) Homans, S. W., 334(138), 343(138,164,165) Homma, J. Y., 211(19), 252(19), 257(235,

Honda, S., 313(14, 17), 345(17) Honda, T., 116(220-223) Honeyman, J., 136(49) Honovich, J., 214(33, 36), 218(88),

237)

222(1 lo), 228(33, 88), 232-235(88), 242(33), 243(88), 249(1 lo), 251(110)

Honzatko, R. B., 18 - 19(49) Hoogerhout, P., 180(232)

Horton, D., 11 1(201), 142(68), 163(181), Horito, S., 149(94-95), 150(96-98)

169- 170(181), 217(55), 229(130), 333(117)

Hoshi, Y., 151(107) Hosie, L., 161(171) Hotta, K., 329( 106) Hough, L., 129(24), 148(87) Hounsell, E. F., 312(3), 313(25,26,28),

314(33, 45), 315(46), 316(53, 73-75), 321(53,73-75, 82), 322(87), 323(28, 33, 74), 324(73,89), 325(45,46,73,75,

331(102, 103), 332(3, 33, 116), 333(33,

345( 184)

82,90), 327(33), 328(73-75), 329-

116), 335(116, 141-143), 339(154),

Houvenaghel, M.-C., 316(65), 321(65) Howell, A. R., 153(121) Hricovini, M., 343(168) Hubschwerlen, C., 131(34) Huckerby, T. N., 329-330( 104) Hudson, C. S., 1(2), 5(14), 151(101) Hudson, Y., 236( 175) Hull, R., 152( 1 18) Hull, W. E., 316(61), 321(61) Hultenby, K., 255(229b) Hulyalkar, R. K., 127(12), 161(12) Hunnemann, D. H., 230(147) Hurlbert, R. E., 224( 1 14) Husaain, A. A., 30(54), 70(54) Hutson, D., 11 l(201) Hyver, K., 214(33), 228(33), 242(33)

P

Ibrahim, I. H., 105(181) Ignatenko, A. V., 218(74) Iida, H., 201(289) Iijima, H., 58(121), lOl(172, 173), 102(121) Iimura, Y., 169(209) Iitaka, Y., 138(58) Ijima, H., 290(6 l), 292(66), 294(66), 296(66) Ikeda, K., 252(214) Ikeda, S., 211(19), 252(19) Ikeda, T., 257(236) Ikegami, S., 116(220-223)

Page 383: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

360 AUTHOR INDEX, VOLUME 50

Ilancock, R. E. W., 218(77a) Imai, K., 219(99), 224(99) Imberty, A., 343(166) Imoto, M., 21 1(14), 214(34, 35),215(14),

217(35), 218(34), 228(35), 236(170), 244(200), 247(35), 249(34,35), 250(34), 251(34), 252(14,215,216), 257(234,236,237), 261(34)

Inage, M., 116(217), 252(216) Inomata, K., 130(28) Inoue, Y., 249(206) Ireland, R. E., 183- 184(237), 187(237) Isakov, V. V., 218(62) Isbell, H. S., 157(143), 158(146), 162(174) Isemura, S., 160(164- 165)

21 1(16), 252(16) Ishida, N., 252(217) Ishidate, M., 126(10) Ishikawa, K., 21 8(95), 228(95), 243(95) Ito, Y.,41(91), 43(91), 46(91),49(100, 101,

I~hida, H., 41(93-95), 47(93,94), 58(120),

104), 50(100, IOl), 51-52(104), 58(123), 60(91, 127), 62-63(130), 66( 127), 67(9 I), 68 - 69( 14 1 - 144), 70(141-142), 73(130, 143, 144, 157, 158), 78(130, 143, 157, 158), 81(130, 157), 82(130), 83(157), 89(127, 142), 110(189), 1 ll(195)

317(80), 321(80)

228(35), 247(35), 249(35), 252(35)

Itoh, T., 127(16), 139(16), 232(158),

Iwashita, T., 168(206), 214(35), 217(35),

Iyer, R. N., 322(88)

J

Jackson, P., 313(18) Jacob, G. S., 198(285) Jacquinet, J. C., 93 - 94( 169), 98( 169),

1 1 I ( 169), 1 13( 169), I 16(213), 294(72), 296(72)

Jager, K.-E., 218(77), 224-225(77), 228- 229(77), 231(77), 242-245(77), 248(77), 25 1(77), 264(77)

Jager, V., 194(270,271) James, K., 280(20) Jann, K., 213(30), 217(30), 230(30), 250(209) Janson, A. M., 302(82) Jansson, P.-E., 3 14(37)

Jantzen, E., 225(119), 229(119), 232(159,

Jarglis, P., 190(258) Jarvis, G. A., 212(22) Ja@, J., 158( 149) Jatzke, H., 30(56), 32(56) Jaurand, G., 1 lO(186) Jay, F., 258(241) Jeanloz, R. W., 278-279(6), 279(13, 18),

Jenkins, S. R., 195(274) Jennings, H. J., 250(208), 339( 150) Jensen, K. J., 287(49), 294(49), 299(49),

Jensen, M., 217(59), 218(71), 227(125) Jeroncic, L. O., 132(36-37), 167(200, 201),

Jersch, N., I58( 152), 160( 159) Jetten, M., 52-53( 1 I l), 57-58( 11 1) Jiao, B., 215(47), 241(47) Jin, H., 22(25,26) Johansen, P. G., 277(3) Johnson, R. S., 213(31) Johnson, W. C., 3 I l(2) Johnston, D. B. R., 286(38) Jones, A. H., 200(286) Jones, B. D., 31 l(8) Jones, J. H., 169(212) Jones, J. K. N., 127(12), 129(24), 161(12),

Jones, N. J., 332-333(116), 335(116) Jordaan, A,, 146(81), 147(82), 148(83) Joullit, M. M., 125(2, 5 , 6), 126-127(6),

168(6), 174(6), 182(2), 184(6) Juaristi, E., 114(202) Jung, K.-H., 73(155), 79(155), 81(155),

Jurczak, J., 190(255)

161)

282( 18), 283(28-3 1)

302(49)

173(200,224)

287(41)

111(197)

K

Kabat, E. A., 316(37, 50, 55, 59), 325(57),

Kaca, W., 227( 126) Kadam, S. K., 2 13(26), 262(26) Kakehi, K., 3 13( I7), 345( 17) Kalamnn, J.-D., 288(58) Kalsner, I., 3 14(40), 325(40) Kamerling, J. P., 52-53(111), 57-58(111),

333(119), 339(158)

60( 126), 63( 126), 66-67( 126), 105( 179,

Page 384: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

AUTHOR INDEX, VOLUME 50 36 1

180), 109(179, 180), 313(21), 329(107, 112), 330(107), 333(112)

Kameyama, A., 58(120) Kamikawa, T., 49( 108), 2 1 I ( 14, 15),

215(14, 15), 252(14-15), 258(15), 263( 15)

Kamio, Y ., 224( 1 15) Kamishima, H., 218(95), 228(95), 243(95) Kampf, A., 138(55) Kandil, A. A., 193(268) Kane, D. P., 316(74-79, 321(74-75),

Kanegasaki, S., 257(237) Kapadia, A., 335(140) Karabinos, J. V., 157( 143) Karibian, D., 214(42,44) Karjala, S., 157(139) Karl, H., 1 lO(185) Karlsson, H., 3 13( 15) Karlsson, K. A., 3 1 I( 1 I), 335( 144) Karube, I., 218(95), 228(95), 243(95) Kasahara, K., 201(289) Kasai, N., 220(103), 232(158), 257(236),

323(74), 325(75), 328(74-75)

259(247)

262(221, 228) Kates, M., 49( 108) Katha, K. P. R., 58(120) Kato, M., 314(41), 325(41) Kauth, H., 287(53) Kawahara, K., 218(75), 231(155, 156),

Kawai, Y., 230(146) Kawamoto, T., 333143) Keilich, G., 220( 104) Kelly, W. G., 345(183) Kelsey, J. E., 129(25) Kenne, L., 288(57), 314(37) Kennedy, J. F., 34(64), 49(64), 58(64),

60(64), 84(64), 105(64) Kenny, C. P., 250(208) Kerek, F., 313(12) Kerekgyarto, J., 60(126), 63(126), 66-

67(126) Kern, M., 252(218) Khare, N. K., 105(181) Khorana, H. G., 215(46,69), 2 18(69),

Kastowsky, M., 253(221), 255-256(228),

251(75,211,212), 261(75)

221(69), 223(69), 227(69), 239(189, 190), 249(69)

Khorlin, A. Y., 281(22)

Khourdanov, C. A., 136(50) Khuong-Huu, F., 195(273) Kibayashi, C., 201(289) Kickhofen, B., 237( 179) Kida, T., 128(23) Kilganiff, C., 259(245) Kiliani, H., 6( 16, 17) Kim, J. J., 212(22) Kim, K. C., 224( I 15) King, C. H., 43(80) King, I. A., 345(184) Kinoshita, M., I lO(184) Kinsho, T., 49( 106) Kinzy, W., 30(57), 37(72), 48(57), 54( 114),

55(72, 115, 116), 58(115), 68(145, 149),

149, 160), 74(145, 146, 150, 154), 75(57), 76(149), 78(114), 79(114, 115), 80(160), 81(114, 115, 154, 160, 162, 164), 84(57, 149), 85(114, 115, 154,

97(150), 98(116, 149), 99(116, 150), lOO(115, 116, 154), 103(I50), 104(162), 107(162), 288(60)

Kirby, A. J., 114(203) Kirikae, F., 256(229d), 264(264) Kirikae, R., 238(185a) Kirikae, T., 21 1(9a, 9b), 256(229d), 264(264) Kirio, Y., 144(74) Kirkland, T. N., 232(162), 235-236(162),

238(182), 259(248), 264(162, 182) Kishi, Y., 139(60)

70(145, 149), 71(149-150), 73(145,

162), 90-91(57), 93(149), 94(116), 96-

=SO, M., 41(93-95), 47(93-94), 58(120, 123), 21 l (11 , 16, 19), 252(11, 16, 19)

Kitagawa, I., 104-105(178), 109(178) Kitajima, T., 62(131), 63(132), 68-70(141),

Kitamura, H., 257(236) Klager, R., 28(69), 49(98-99), 50(98),

52(69), 114(69) Klein, A., 316(62, 66-68), 321(62, 66-68),

3 3 3( 62) Kleppe, K., 16(42) Klosterman, M., 25(43) Klotz, W., 25(44, 49) Knapp, W., 230( 145), 235(145) Knatterud, G. L., 259-260(250) Knollmann, R., 158( 15 I), 160( 159) Knorr, E., 21(8) Knowles, B. B., 335(140)

82( 132)

Page 385: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

362 AUTHOR INDEX, VOLUME 50

Knox, K. W., 229(134), 234(134) Kobata, A., 62(130- 131), 63(130), 73(130),

78( 130), 8 1 - 82( 130), 3 14(43), 3 15(47), 325(43,47), 333(123, 125)

Kobayashi, M., 68-69(143-144), 73(143, 144), 78(143)

Kobayashi, Y., 252(217) Koch, M. H. J., 254(224,226), 255(229a) Kochetkov, N. K., 282(24), 314(38), 325(91) Koenigs, W., 21(8) Koerner, T. A. W., Jr., 344( 175) Kohn, B. D., 127(13), 195(276) Kohn, P., 127(13), 160(156, 157), 161(166),

Koike, K., 49-50(100), 56(118), 58(122) Kokx, A. J. P. M., 167(197), 200(197) Koles, N. C., 259(249) Kollat, E., 282(27), 302(27) Komatzu, S., 151(101) Komura, H., 168(206) Kondo, H., 252(2 14) Kondo, S., 218(83), 251(83), 261(83,268),

Konings, J. J. H. G., 167(197), 200(197) Konradsson, P., 1 16(2 15) Kontrohr, T., 261(270) Koppen, P. L., 329( loo), 333( 100) Koprowski, H., 337(147) Korczynski, A., 1 6 1 ( 169) Kornfeld, S., 329( 1 lo), 338( 1 10) Korol, E. L., 139(59) KO&, S., 49(108)

171(156), 195(275,276)

264(264)

KoXh, W., 287(40), 300-301(79), 302(40, 81)

Kosma, P., 218(64), 250(64), 251(211-213) Kostenbander, H. B., 30(54), 70(54) Kosunen, T. U., 229( 136) Kotani, S., 211(14), 215(14), 252(14),

Koto, S., 22(29) Kovacs, J., 248(204) Kowalczyk, D., 307( 104) Kraaijeveld, N. A., 43(81) Krasikova, I. N., 2 18(62) Kraska, U., 239(188) Krauss, G. A., 139(61) Krauss, J. H., 215(50), 218(89), 219(50,98,

257(236,238)

loo), 225 -226(89), 228(89), 232(50), 233(89), 238(89, 185a), 243(89), 264(265)

Krepinsky, J. J., 62(134), 343(162, 163) Kreuzer, M., 2 1 ( 13) Kris, R. M., 335(143) Krohn, K., 21(7) Kroon-Batenburg, L. M. J., 337( 146) Kropinski, A. M. B., 230-231(144) Kuge, S., 252(217) Kuhn, H.-M., 259(243,244,244a, 244b) Kuhn, R., 3 13( 13) Kiihne, W., 8(24) Kulshin, V. A., 218(73, 77), 224(77),

225(116), 225(73, 77), 226-227(73), 228-229(77), 231(77), 242(73, 77), 243-245(77), 248(73, 77), 25 l(73, 77), 257-258(116), 264(77)

Kumada, H., 261(268) Kumazawa, Y., 257(235,237) Kundt, I., 173(223) Kung, P. C., 259(245) Kunz, H.,21(6, 12), lll(194, 196),278(7,

10-11), 279(7, 14-16), 280(19a-l9c), 281(21,23), 283(14-16, 19a-l9c, 32- 37, 39), 284(10, 15, 32-36), 285(19a- 19c, 34, 36), 286(7, 10, 11, 19a-l9c, 37, 39), 287(16, 39-40,43-45,45a, 46,

290(62), 291(67,68), 293(71), 294(19a- 19c, 32, 33, 39, 43, 44, 67, 68, 73), 295(67, 73), 296(67), 297(44,68, 73), 298(44, 73), 299( 19a-l9c), 300(79,80),

87), 304(90), 306(93, 100, 101), 307(103, 104, 108)

51 -53), 288(43-45, 54), 289(32),

301(32,79), 302(40, 81), 303(46,86-

Kurosaka, A., 317(77), 321(77) Kurosawa, M., 2 1 1 (1 5), 2 15( 15), 252( 15),

Kurtz, R., 234( 166) Kurunaraine, D. N., 218(77a) Kusama, T., 21 1(15), 215(15), 238(185),

258(15), 263(15)

252(15), 256(229d), 258(15, 185), 263( 15), 264( 185)

Kusumoto, S., 116(217), 21 1(5,9b, 14, 15), 214(34), 215(14, 15), 218(34),

238(167, 169, 183, 185, 185a), 243( 196), 244(200), 249(34), 250(34), 251(34), 252(14, 15, 172, 196,215, 216,219), 254(219), 256(118,229d),

237), 258(15, 118, 167, 168, 183, 185,

225(118), 236(5, 118, 167- 170, 172),

257(5, 118, 167-168,233,234,236,

Page 386: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

AUTHOR INDEX, VOLUME 50 363

239), 259(243,244,244a), 261(34, 118), 263(185), 263(15), 264(167, 169, 183,266), 306(99)

Kusunose, N., 21 1(14), 215(14), 252(14) Kuwano, M., 344( 181) Kuzuhara, H., 163(179)

L

Labischinski, H., 21 1(5), 212(25), 214(25), 236(5), 252(219), 253(220,222),

262(220,228) Lachut, C. H., 125(4) Lackzo, I., 282(27), 302(27) Lacombe, J. M., 287(48), 294(48) Laczko, I., 303(84a) Ladduwahetty, T., 110( 188) Ladner, W., 185(244) Laine, R. A., 218(57b) Lakshmi, S. K. B., 227( 124) Lam, C., 211(13), 252(13), 262(13) Lamblin, G., 316(60-63,65-68), 321(60-

Lancelin, J. M., 139(64) Lansbury, P. T., Jr., 279( 12), 306(94) Larsson, L., 225(119), 229(119, 135),

Lartey, P. A., 143(69) Lauffenburger, M., 324(84), 329(84) Lavielle, S., 287(50), 299(50) Lawson, A. M., 313(25-26,28), 314(45),

323(28,74), 324(73), 325(45,46,73,

254(219-220), 255 -256(228), 257(5),

63,65-68), 333(62)

230(141)

315(46), 316(73-75), 321(73-75, 82),

75, 82), 328(73-75), 329-331(102) Lax, I., 335(143) LeBeyec, Y., 214(44) Lederer, E., 81(166, 167) Lederkremer, R. M., 132(36-38), 133(39),

Lee, A. C., 153(121) Lee, C. M., 143(69) Lee, J., 307( 106, 107) Lee, J. B., 161(167) Leffler, J., 335(144) Lefrancier, P., 81(166, 167) Lehmann, V., 21 1(3), 223(113), 226(113,

164( 158), 18 l(38)

Lehong, N., 22-23(23) Lemieux, R. U., 17(44,46,47), 18(44),

35(75), 58(75, 152), 157(137a), 280(20),

121), 339(158) 332(114-115), 333(114, 115, 118-

Lemoine, J., 3 13( 16) Lerner, L. M., 127(13), 130(29), 137(52),

158(148), 160(156, 157), 161(166), 171(156), 195(275,276)

Leroy, Y., 313(16), 316(76), 321(76),

Lewis, M. D., 139(60)

Ley, S. V., 116(214) Lhermitte, M., 316(60-61, 63, 68), 321(60,

Li, C., 256(229c) Liao, J., 316(55), 333(119), 339(158) Libby, P., 238(185a), 264(265) Libermann, T. A., 335( 143) Lichtenthaler, F. W., 190(258) Lico, I. M., 290(65) Liebig, J., 5( 11) Likhoshersov, L. M., 282(24) Lindberg, A. A., 218(68), 222-223(68),

325(76), 329-330(76)

Lewis, V., 230-231(144)

61,63,68), 333(132)

225(68, 117), 228(68), 234(117), 241(68), 251(68), 255(229b), 261(68)

Lindberg, B., 31 l(9) Lindgren, K., 229( 135) Lindh, F., 314(37), 333(128) Lindner, B., 21 1(9), 212(25), 214(25, 34,

37-40), 218(34, 66a, 68, 73, 77, 84, 92), 222-223(68), 224(66a, 771, 225(68, 77,92), 226(73, 92), 228(37, 68, 73, 77, 84,92), 229(38, 77), 230(37), 231(38, 77), 234(38), 235(37),

68,92), 242(92), 242(38,77, 84,92), 243(37, 77, 173, 195a), 244(77, 173), 245(77), 248(77), 249(34, 73), 250(34), 251(34,68,73,77), 261(34,68), 264(77)

236(173), 239(38), 240(37-39), 241(37,

Ling, N. C., 287(50), 299(50) Lingens, F., 221( l05), 230(105), 252(105) Link, K. P., 157(139), 277(2) Lipka, G., 255(229) Lipkind, G. M., 314(38)

122), 227(113, 122, 123), 236(170, 171, 173), 243(173), 244(3, 122, 123, 173, 197, 199), 256-257(3)

Lipshutz, B. H., 129(26) Liptik, A., 60(124- 126), 63(126), 66-

67(126), 105(124), 108(124)

Page 387: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

364 AUTHOR INDEX, VOLUME 50

Litter, M. I., 164( 182), 166( 193), 167( 198-

Liu, H., 171(219) Liu, P. S., 43(80) LoBuglio, A. F., 259-260(250) Loibner, H., 211(13), 252(13,218), 262(13) Lomax, J. A., 218(76) Long, C. W., 162(173) Long, G. W., 143- 144(70) Lijnn, H., 22(18), 23(31-33), 290(63a) homes, L. M., 339( 154) Loppnow, H., 21 1(9a), 236(167), 238(167,

185a), 257(167), 258(167,239), 264( 167,265)

Loretzen, J.-P., 44(82) Lorkiewicz, Z., 246(202) Lormeau, J.-C., 331(113) Liiw, A., 81(162), 85(162), 104(162),

107(162) Liideritz,0.,211(1-5),213(2,4,28, 30),

214(35, 38), 217(1,28, 30,35, 54), 218(1,4, 54, 78, 81, 86), 221(81), 223(112), 225(4, 86), 228(4, 187), 229(38, 86, 128), 230(30,81, 142), 231(38, 142), 234(1, 38, 78), 235(142),

199), 171(182)

236(5, 128, 170- 172), 237(177- 179), 238(81, 187), 239-240(38, 187), 242(38), 243( 187, 196), 244(3, 199), 246(4, 78,86,201), 247(4, 35, 187), 249(35, 112), 252(35, 172, 196), 256(1,

234) 3-4,230), 257(1, 3-5,28,230, 232,

Liideritz, T., 236(170) Lugtenberg B., 230( 143), 235( 143) Lui, S. C., 316(73), 321(73), 324-325(73),

Lundblad, A., 333( 127- 129) Lundt, I., 110(187), 134(41-44), 135(45-

47), 136(48), 158(42,43), 160(42),

227), 175(226,228), 176(229)

328(73)

169(210), 173(44), 174(41-43,226-

Liining, B., 288(56), 294(56) Luzzati, V., 254(223), 255(227,227a, 229a),

L'vov, V. L., 218(74) 256(230-23 I), 257(230), 259(243)

M

Maas, A. A.M., lOS(179, 180), 109(179, 180), 329 - 330( 107)

Macharadze, R. G., 281(22) Macher, I., 21 1(13), 252(13), 259(243),

Maciejewski, S., 190(259) Mackie, D. M., 164( 183) MacMillan, D., 151(99) Madigan, M. J., 313(25) Madigan, M. T., 247(203) Maeta, H., 21(14-16) Magnus, P., 184(242) Mai, L. A., 157(141) Maier, T., 41(89) Mairanovskii, V. G., 161(168) Maitra, S. K., 230(140) Makela, P. H., 227(125) Malcita, M., 313(19) Malchow, D., 237(179) Malek, G., 279(17), 283( 17) Maley, F., 3 14(44), 325(44) Mallet, J.-M., 23(36) Mallow, W. R., 129(25) Maltby, D., 3 13(24) Mamat, U., 21 1(9a, 9b) Mandrell, R. E., 212(22) Mann, J., 187(250,25 1) Maquenne, L., 144( 129) Marino, C., 132(38), 160(162), 166(194),

262( 13)

168(194), 170(194), 172-173(194), 180(23 l), 18 l(38)

Mark, E., 133(40) Marks, G. S., 277(4) Maron, L., 126- 127(8) Mama, A., 23(36) Marre, R., 232( 159) Marsh, W. L., 316(59) Marshall, R. D., 277(3, 5), 278(5) Martel, C., 329(105) Marx, A., 23 1( 152) Man, J., 279(15), 281(21), 283(15, 33, 39),

284(15, 33), 286(39), 287(39,40), 294(39), 297(33), 302(40, 8 1)

Masato, M., 41(91), 43(91), 46(91), 60(91), 67(91), 11 1( 195)

Mascagni, P., 214(33), 217(60), 222(1 lo), 228(33,60), 236(172), 242(33), 243(172), 248(60, 172), 249(110,207), 251( 110)

Mashimo, J.-L,220( 103), 257(236), 259(247) Masoud, H., 2 1 8(84,92,94), 2 19(98), 225 -

226(92), 228(84,92,94), 241(92,94),

Page 388: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

AUTHOR INDEX, VOLUME 50 365

242(84,92), 26 l(270) Masuyama, A., 128(23) Mathison, J. C., 264(263) Matsui, M., 126( 10) Matsumoto, K., 152( 1 16) Matsumoto, T., 21(14-16) Matsuura, F., 317(79), 321(79) Matsuura, H., 345(182) Matsuura, M., 211(19), 231(156), 252(19),

257(235,237) Matsuyama, M., 317(77), 321(77) Matthews, T. J., 314(41), 325(41) Mattsby-Baltzer, I., 229( 135) Matuo, S., 339(159) Mautsch, H. H., 303(84a) Mawhinney, A. W., 316(64), 321(64), 329(64) Mawhinney, T. P., 316(64), 321(64), 329(64) May, R. P., 253(222) Mayberry, W. R., 230( 138), 232( 138) Mayer, H., 215(49, 50), 218(82, 84,87, 89-

104), 221(96, 107), 223(87), 224(91,99,

121), 227(90, 124), 228(84, 87,89,92), 230(82, 145), 232(50, 82, 87, 101, 160), 233(87,89), 234(87, 165), 235(87,91, 145), 238(87,89, 185a), 241(92, 193), 242(84, 92, 194), 243(87,89), 247(101, 160), 249(87, 90), 255(229a), 257(232), 261(165,270), 264(265)

92), 219(49-50,96,98-101), 220(96,

114), 225(89-91, 121), 226(89,92,

McCullough, K. J., 125(4) McGarvey, G. J., 183-184(237), 187(237) McKenzie, G., 236( 170) Meagher, M. M., 18(50) Mehmet, H., 329-331(102) Meldal, M., 282(25), 287(49), 288(25),

294(49), 299(49), 302(25, 82,83,49), 306(95 - 99)

Mel'nikova, V. I., 169(208) Mendelsohn, J., 335(143) Mendicino, J., 329(93) Meranduba, A., 68-69(139), 72-73(139),

Merck, R., 185(243), 186(245) Men, G., 288(55), 294(76), 302(55) Mesrobeanu, J., 2 13(27) Mesrobeanu, L., 2 13(27) Messner, P., 250(209) Meuwly, R., 114(210) Meyer, B., 333( 11 8)

78( 139), 82-83( 139)

Meyer, D. M., 333(132) Meyer, K. C., 232(162), 235-236(162),

Meyer, M. V., 244( 198) Michalski, J.-C., 313(16), 333(130, 131),

Michel, G., 218(61), 225-226(121) Michel, J., 25(45-46), 27(45), 28(46, 65),

264( 162)

339( 154)

29(45-46, 30(51, 52a, 53), 34-35(52a), 36(51, 52a, 65), 37-38(51, 52a), 39(51, 53), 41-42(65), 43(51,52a), 44(51), 52(46), 58(45,51), 60(46), 61(51), 70(53), 11 l(46, 53), 114(46, 51, 52a, 52b), 289(60)

Michon, F., 339( 150) Mieczkowski, J., 190(255) Mielniczuk, Z., 229( 135) Milat, M. L., 116(213) Minale, L., 105(182) Minka, S., 225-226(121) Minner, I., 227(123), 244(123) Mitchell, D. H., 256(229c) Mitchell, D. L., 129(24), 13 l(33) Mitov, I., 259-260(252) Mizuochi, T., 314(41,43), 325(41,43) Mizutani, T., 259(247) Moenng, U., 25(39-42) Moghissi, K. S., 317(79), 321(79) Molina, M. T., 139(61) Moll, H., 229(137), 232(159, 161),

Meller, H., 19(52) Molyneux, R. J., 198(285) Mombers, C., 230(143), 235(143) Mondange, M., 21 1(17), 252(17) Monsalvatje, M., 188(253) Montreuil, J., 216(51), 221(51), 313(16),

3 14-3 15(30), 3 16(76), 32 1(76), 324(86), 325(30, 76), 329(76,96,97,

131), 334(136), 337(146), 343(30, 169)

251(211-212)

105), 330(76), 333(86,96-97, 130,

Mootoo, D. R., 116(215) Moradei, O., 174(225) Moran, A. P., 219-220(97), 222(97),

225(979, 116), 226(97), 229(136), 241(97), 252(97), 257-258(116)

Moreno-Mafias, M., 182(235) Morey, M. C., 129(26) Morgan, J. W. W., 151(102) Morgan, W. R. J., 329(98), 333(98)

Page 389: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

366 AUTHOR INDEX, VOLUME 50

Morgan, W. T. G., 316(48) Morgan, W. T. J., 213(29)

Morrell, A. G., 338( 148) Moms, D. A., 3 16(64), 32 1 (64), 329(64) Moms, E. R., 344( 176) Momson, D. C., 212(20), 256(20), 264(262) Motherwell, W. B., 145(79, 80) Mountsen, S., 306(96) Moustafa, A. E. A., 237(177) Moxon, E. R., 225(120) Miihlradt, P. F., 223(113), 226-227(113) Mukaiyama, T., 21(9), 22(21) Mukerjee, P., 256(229c) Mulder, G. J., 144(75) Miiler-Fahmow, A., 152( 114) Miiler-Loennies, S., 2 18(66a), 224(66a) Munford, R. S., Jr., 214(45), 229(45),

Murai, Y., 21(9) Murakami, K., 37-38(73) Murase, T., 41(93), 47(93), 58(120) Murray, P., 186(247) Murray, R. G. E., 241(192)

M o ~ , K., 49( 105 - 107)

238(45, 180- 181, 186), 264(181)

Musehold, J., 236(168, 169), 257-258(168), 264( 169,266)

Musina, L. Y., 218(74) Mutsaers, J. H. G. M., 316(57,63), 321(63),

Myers, K. R., 236( 175) Myers, P. L., 193(267), 196(279)

325(57)

N

Nagai, Y., 339(156) Nagaki, K., 257(236) Nagao, S., 257(236) Nagasawa, K., 249(206) Nagase, H., 249(205) Nagawa, Y., 218(95), 228(95), 243(95) Nagayo, T., 317(77), 321(77) Naiki, M., 339(159) Nakabayashi, S., 279( 18), 283( 18) Nakagawa, M., 160(164, 165) Nakahara, N., 292(66), 294(66), 296(66) Nakahara, Y., 49(100, 104), 50(100),

51(104), 52(104, 110), 53(110), 56(118), 57(110), 62(130, 131), 63( 130), 73( 130), 78( 130), 8 1 - 82( 130)

Nakajima, H., 317(77), 321(77) Nakajima, Y., 151(107) Nakaminami, G., 160(164, 165) Nakamoto, S.-I., 21 l(18) Nakanishi, H., 218(95), 228(95), 243(95) Nakanishi, K., 168(206)

Nakatsuka, M., 21 1(19), 252(19), 257(237) Nakatsuka, T., 22(21) Nakayama, K., 211(15), 215(15), 252(15),

258(15), 263(15) Nakayima, H., 211(15), 215(15), 252(15),

258( 15),263( 15) Nakazawa, T., 150(98) Namgoong, S. K., 200(287,288) Nbnhi, P., 35-37(71), 60(124), 105(124),

108( 124) Naoki, H., 168(206), 214(34, 35), 217(35),

218(34), 228(35), 247(35), 249(34, 35), 250(34), 251(34), 252(35), 261(34)

Nakatsubo, F., 37-38(73)

Nashed, M. A., 249(207) Nassr,M.A. M., 116(213), 155(133),

156( 134) Naumann, D., 252(219), 253(219,220),

255-256(228), 262(220,228) Nealson, K. H., 247(203) Nef, J. U., 27(50) Nelson, C. R., 165(190) Nemek, J., 158(149) Nerkar, D., 258(241) Neszmelyi, A., 60(124), 105(124), 108(124),

2 18(64), 250(64,209) Neuberger, A., 277(3-5), 278(5) Neufeld, E. F., 338(149) Nicolaou, K. C., 21(10), 23(30), llO(188) Nicotra, F., 140(66) Nieduszynski, I. A., 329-330( 104) Niemann, C., 157( 139) Niemann, H., 339(157) Nifant’ev, N. E., 314(38) Nikaido, H., 263(258) Nilsson, B., 313(27), 316(55), 333(127-129) Nimtz, M., 329(99), 333(99) Nin, A., 163(181), 166(192), 169-170(181) Nishijima, M., 252(217) Nishikawa, S. I., 264(264) Nishimura, C., 211(19), 252(19) Nishio, H., 49(107) Nishiyama, H., 249(205) Niwa, Y., 218(95), 228(95), 243(95)

Page 390: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

AUTHOR INDEX, VOLUME 50 367

Nolan, T. J., 161(167) Noort, D., 144(75) Norberg, T., 22(18, 19,24), 294(56) Noms, F., 18(51) Noms, K. E., 18(51) Notermans, S., 180(232) Novikova, 0. S., 282(24) Nowotny, A., 217(52), 223(52) Noyori, R., 21(11) Nukada, T., 68-70(141, 142), 89(142)

62( 130), 63( 130, 132), 66( 127),

132), 89( 127) Numata, M., 49(103), 51(103), 58(122) Nunez, H. A., 334(133) Nunomura, S., 49-50(101, 102) Nunozawa, T., 164( 185) Nurminen, M., 227(125), 229(132) Niirnberger, H., 62( 133) Nutt, R. F., 195(274)

Nukuda, T., 49(104), 51-52(104), 60(127),

73(130), 78(130), 81(130), 82(130-

0

Oddon, Y., 344(177) Ogasawara, K., 130(28) Ogawa, T., 41(91), 43(91), 46(91), 49(100-

104), 50(100- 102), 51(103, 104), 52(104, IIO), 53(110), 56(118), 57(1 lo), 58(121, 122), 60(91, 127), 62( 130, 13 I), 63( 130, 132), 66( 127),

142), 73(130, 143, 144, 157, 158), 78(130, 143), 81(130, 157), 82(130, 132), 83(157), 89(127, 142), lOl(172, 173), 102(121), 110(189), 111(195), 218(80a), 228(80a), 257(236), 290(61), 292(66), 294(66), 296(66)

Ogawa,Y.,211(11, 16),252(11, 16) Ogura, H., 127( 16), 138(56, 57), 139( 16,63) Ohashi, K., 49(108), 104- 105(178), 109(178) Ohle, K., 127( 17) Ohmori, M., 259(247) Ohmori, T., 169(209) Ohno, K., 249(205) Ohsawa, A., 232( 158) Ohtsuka, Y., 211(18) Ohya, Z.-I., 150(98) Oishi, K., 259(249) Okabe, M., 196(280)

67(9 I), 68 -69( 141 - 144), 70( 141 -

Okada, M., 126(10) Okahara, M., 128(23) Okamoto, T., 152(116) Okouchi, K., 339(159) Olah, V. A., 60( 125) Oltvort, J. J., 25(43) Ono, M., 344( 18 1) Ono, Y., 21 1(15), 215(15), 252(15), 258(15),

Ooi, C. E., 263(261) Ortufio, R. M., 168(207), 182(235,236,

263( 15)

238-239), 184(207,238,240, 241), 185(243), 186(244), 188(252,253)

Osada, Y., 211(15), 215(15), 252(15), 258(15), 263(15)

Osborn, M. J., 244( 197) Oscarson, S., 288(57) Ostmann, P., 14(36) Otani, S., 152(116) Otsuka, K., 257(236) Otsuka, T., 252(217) Ottesen, M., 18(48) Otvos, L., Jr., 282(26,27), 288(26), 302(26-

Oulevey, J., 230( 147) Overend, W. G., 143- 144(72) Overhand, M., 71(151), 73(151) Ovodov, Y. S., 218(62)

27), 303(84a, 84b)

P

Paal, M., 288(58-59) Packer, N. H., 222-223(109), 226(109) Panfil, I., 190(260), 191(261-263) Panza, L., 140(66) Papahatijs, D. P., 23(30) Parekh, R. B., 61(136), 314(42), 325(42),

Parent, J. B., 264(267) Partridge, S. M., 213(29) Pascher, I., 311(11) Passmore, F., 6-7(21), 155(130) Paster, B. J., 247(203) Pasteur, L., 3(7), 8(7) Paulsen, H., 21(4-5), 22(5), 35(4), 41(4),

44(82), 53(4, 112), 57(112), 58(129),

104- 105(4), 114(209), 217(59), 278(8),

343(42)

60(128-129), 62(133), 66-67(135),

286(8), 288(55, 58-59), 290(64), 292(69), 293(69-70), 294(74, 76),

Page 391: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

368 AUTHOR INDEX. VOLUME 50

295(8,74), 302(55,83), 306(95), 343- 344( 174)

Paulsen, K., 296(77,78) Paulson, J. C., 303(89), 304(89-90),

Pavia, A. A., 287(47,48), 294(47,48,75),

Payen, A., 8(25) Pazur, J. H., 16(40-42) Peach, J. M., 197(281-282) Pedersen, C., 110(187), 134(41-44),

339( 154)

314(35), 344(177)

135(45), 158(42-43), 160(42), 169(210), 173(44, 223), 174(41, 42, 226,227), 175(226,228), 176(229)

Pedersen, H., 17(45) Pedersen, T. G., 18(48) Pederson, C., 314(34), 325’34) Pederson, H., 327(34) Pedron, T., 259(246) Pepe, G., 344( 177) Perera, P. Y. Pkrez, S., 343(166) Perini, F., 324(85) Perlin, A. S., 158-159(150), 164(184) Perry, M. B., 287(41) Persoz, J. F., 8(25) Peter-Katalinic, J., 2 18(64a), 224(64a),

Peters, S., 302(83), 306(95) Peters, T., 288(57) Petitou, M., 33 1( 1 13) Petursson, S., 154(125), 155(126) Pfannemuller, B., 152( 1 13, 1 15) Pfeiffer, G., 314(40), 325(40), 329(108) Pickering, N. J., 316(73), 321(73), 324,

Pierce-Crktel, A., 324(86), 333(86) Piloty, O., 157(138) Pimlott, W., 31 l(11) Piszczek, L., 161(169) Pivnitsky, K. K., 169(208) Pizza, C., 105(182) Platzer, N., 333(132) Plessas,N.R., 151(111, 112) Plummer, T. H., Jr., 314(44), 325(44) Podolsky, D. K., 316(71, 72), 321(71, 72),

Pohlmann, T. H., 238(186) Pokorny, M., 165(189) Polenov, V. A., 139(59)

317(78, 81), 321(78, 81), 324(81)

325(73), 328(73)

329(71)

Pollack, M., 212(21), 259(249) Ponomarev, A. M., 161(168) Ponsati, O., 182(234,236), 183(236,238),

Poole, A. R., 325(92) Popenoe, E. A., 277(2) Pora, H., 305(91) Potier, P., 195(273) Pougny, J. R., 116(212-213), 239(188) Powell, M. E., 325(90) Prasit, P., 192(266) Pravdic, N., 164(186), 165(188, 189) Preuss, R., 1 1 1 - 112( 190) Priebe, W., 142(68) Prohaska, R., 344( 175) Prout, K., 197(281-282) Purcell, S., 244( 198) Puvanesarajah, V., 2 19( 100)

184(238)

Q Qureshi, N., 214(32, 33, 36,41,43),

215(32), 217(32,60), 218(63, 88), 222( 1 lo), 228(32, 33,60, 88), 232(88, 162), 233(88), 234(88, 166), 235(88, 162), 236(32,41, 162, 174), 238(182, 184), 239(41), 241(41), 242(33), 243(63, 88, 174, 198), 244(32,63), 248(60, 174), 249(110,207), 251(1 lo), 259(248), 264(162, 182, 184)

R

Rademacher, T. W., 61(136), 225(120), 314(42), 325(42), 343(42, 164, 167)

Radziejewska-Lebrecht, J., 2 18(67), 2 19(67, 98), 227(126), 241(193)

Radzyner-Vyplel, H. Raetz, C. R. H., 21 1(7), 218(63), 226(63),

232(162a), 238(184), 240(191), 244(63,

264( 184) 198), 249(207), 262(191,254-255),

RajanBabu, T. V., 143- 144(71) Rajanikanth, B., 154- 155(124) Ralapati, S., 343( 169) Ramos, R. A., 264(263) Ramsden, N. G., 154(122, 125), 155(126),

198(122,285) Rana, S. S., 329(93) Randall, J. L., 21(10), llO(188) Rao, B. N. N., 316(56, 58)

Page 392: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

AUTHOR INDEX, VOLUME 50 369

Rao, V. S. R., 333( 122)

Ratcliffe, R. M., 58( 152) Rauwald, W., 53( 1 12), 57( 112), 290(64) Ray, P. H., 129(25) Reboul, J. P., 344(177) Reck, F., 58(129), 60(129), 66-67(135) Reddy, G. S., 143-144(71) Redmond, J. W., 222-223(109), 226(109),

227(123), 244(123) Rees, B. H., 13 l(32) Rees, D. A., 311(1), 344(176) Reeves, R. E., 16(43) Regele, C., 32(60) Regeling, H., 126(7), 127(7, 22), 149(22) Regoeczi, E., 329(97), 333(97) Rehorst, K., 148(85) Reichrath, M., 24(37), 25(37, 39-42) Reichstein, T., 157( 140) Reilly, P. J., 18(50), 19(53) Reimann, H., 162(175) Reimer, K. B., 306(99) Reinhold, V., 316(60), 321(60) Reinhold, V. N., 213(31) Rembold, H., 25(48) Renfrew, A. G., 151(104) Renwick, G. C., 329(99, 109), 333(99) Reyna-Pinedo, V., 195(273) Ribi, E. E., 214-215(32), 217(32), 228(32),

Rapp, R., 5 (W

236(36, 172), 243(172), 244(32), 248( 172)

Ricart, G., 313(16), 316(76), 321(76),

Riccio, R., 105(182) Richards, J. C., 218(77a) Richardson, D. C., 129(25) Richardson, G. M., 5(13) Richtmyer, N. K., 152( 1 17) Riddell, F. G., 3(8), 5(8) Riesen, W., 339( 155) Rietschel, E. T.,211(1-6, 8,9,9a,9b, 14),

325(76), 329-330(76)

212(25), 213(2,4), 214(25, 34-35, 37- 40), 215(14), 217(1,35, 57-59), 218(1,

83), 218(85,86), 219-220(97), 221(81), 4, 34, 57, 64a, 68, 72, 73, 75, 77-81,

222(68,97), 223(68), 224(64a, 77, 79), 225(4,68, 73,77, 85, 86,97, 116- 118), 226(72,73, 85,97), 227(73,85, 125), 228(4, 35, 37, 68, 72, 77, 79, 187), 229(38, 77,79,86, 128, 129, 132, 137),

230(37, 81, 142, 143), 231(38, 77, 85,

117), 235(37, 142, 143), 236(5, 118, 128, 167, 169, 170, 172, 176), 238(81, 85, 167, 169, 176, 183, 185, 185% 187),

68, 97), 242(38, 73,77,85), 243(37,77, 79, 187, 196), 244(3, 77), 245(77), 246(4,78, 79, 86), 247(4, 35, 187),

251(34, 68, 72, 73, 75, 77, 79, 83)), 252(14,35,97, 172, 196,219), 253(220), 254(219,220,224), 255(229a), 256(1, 3-5,6, 118,2294

232-234), 258(116, 118, 167, 183, 185, 239), 259(6,243, 244,244a), 261(34, 68,75, 83, 118), 262(220), 263(185), 264(77,79, 167, 169, 183,264,266)

129, 142, 151, 156), 234(1, 38, 78-79,

239(38, 187), 240(37-39, 187), 241(37,

248(73,77), 249(34-35,72), 250(34),

230),257(1, 3-6, 116, 118, 167,230,

Rijkse, I., 329-330(107) Riley, D. A., 143(69), 229( 130) angler, N. J., 344( 179) Rio, S., 294(72), 296(72) Rivoire, B., 105( 18 1) Robbins, G. B., 151(100) Robinson, M. J. T., 3(8), 5(8) Rodionov, A. V., 2 18(74) Rodriguez, M., 219(99), 224(99) Roelcke, D., 339( 155, 156) Rogers, G. N., 339(154) Rogers, M. E., 324(84), 329(84) Rohle, G., 35(74) Rohrscheidt-Andrzejewski, E., 2 12(24) Rollin, P., 190(256) Romijn, D., 334(136) ROOS, M., 25(46), 28-29(46), 52(46),

60(46), 11 1(46), 114(46) Roppel, J., 219(96), 220(96, 104),

221(96) Rosankiewicz, J. R., 321(82), 325(82) Rosen, T., 290(65) Rosenberg, R. D., 33 1( 1 13) Rosenfelder, G., 246(20 1) Rosevear, P. R., 334(133) Rosner, M. R., 2 15(46,69), 2 18(69),

221(69), 223(69), 227(69), 239(189, 190), 249(69)

Ross, B. C., 145(79, 80)

Rothenberg, R. J., 232( 157) Roth, A., 256-257(230)

Page 393: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

370 AUTHOR INDEX, VOLUME 50

Rousd, P., 316(60-63,65-68), 321(60- 63,65-68), 333(62)

Rouzaud, D., 140(65) Riicker, E., 39(77) Rudolph, J. R., 329(97), 333(97) Rumpold, H., 335(142) Rupprecht, E., 226-227( 122), 244( 122, 197) Russa, R., 218(81), 221(81), 230(81),

238(81), 246(202) Russell, D. H., 313(24) Russell, M. A., 153(121) Russell, R. N., 171(219) Russo, G., 140(66) Ryan, J. L., 212(20), 256(20)

S

Sabesan, S., 303-304(89) Sabharwal, H., 333(127- 129) Sabisch, A., 253(221), 255-256(228),

Sadoff, J. C., 259-260(250) Sadozai, K. K., 62(130, 131), 63(130, 132),

Saenger, W., 152(114) Sager, W., 21(12), 290(62) Saha, B. C., 16(38) Sahoo, S. P., 187(248) Saito, A., 150(97) Saito, H., 3 1 l(5) Saito, M., 126(10) Saito, T., 317(80), 321(80) Sakagami, M., 104- 105(178), 109(178) Sakaguchi, N., 244(200) Sakai, K., 52-53(110), 57(110) Sakai, T., 18(48) Saksena, A. K., 162(177) Sala, L. F., 164(182), 171(182,215-218) Salimath, P. V., 218(87), 223(87), 228(87),

233(87, 164), 234(87, 164, 165), 235(87, 164), 238(87), 242(194), 243(87), 249(87)

Sallam, M. A. E., 155(132) Saltman, R., 287(50), 299(50) Samantano, R. H., 160(156, 157), 171(156),

Samraoui, B., 343(173) Samreth, S., 229( 130) Samuelsson, B. E., 31 I( 11) Sinchez-Ferrando, F., 182(235), 188(253)

262(221,228)

73(130), 78(130), 81(130), 82(130, 132)

195(275)

Sandford, P. A., 31 l(6) Sandstrom, W. M., 157(142) Sarfati, S. R., 21 1(17), 252(17) Sane, 0. Z., 162(175-176) S a d , H., 144(74) Sash, H., 324(84), 329(84) Sato, J. D., 335( 143) Sato, S., 49(100-101, 104), SO(l00, 101),

51-52(104), 60(127), 66(127), 68- 70(142), 73(158), 78(158), 89(127, 142)

Satterthwait, A. C., 214(46) Sauter, N. K., 339(153) Savage, A. V., 329( loo), 333( 100) Scannon, P. J., 259-260(251) Schachter, H., 321(83) Schade, F. U., 21 1(9a, 9b), 256(229d),

264(264) Schade, U., 229(133), 238(185, 185a),

258(185), 263(185), 264(133) Schade,U.F.,211(5),225(116, 118), 236(5,

118, 170, 172), 243(196), 252(172, 196), 256(118), 257(5, 116, 118,233, 234), 258(116, 118), 261(118)

84(63), 105(63) Schaffer, R., 34(63), 49(63), 58(63), 60(63),

Schaller, H., 252(218) Schaubach, R., 37(72), 54(114), 55(72, 116),

78-79( 1 14), 8 1( 1 14), 85( 1 14), 94( 1 16), 98- 100( 1 16)

Schauer, R., 329( 1 1 l), 332-333( 11 l),

Scheinberg, I. H., 338(148) Scheuer, P. J., 230(149) Schimpff, G. W., 147(144) Schiphorst, E. C. M., 329(100) Schlecht, S., 227(124), 230-231(142),

235(142), 237(178) Schlessinger, J. J., 335(143) Schliiter, C., 264(266) Schmidt, G., 21 1(9a, 9b) Schmidt, M., 329( 108) Schmidt, 0. T., 127(18) Schmidt, R. R., 21(1-3), 22-23(17), 24(1,

337( 1 1 1)

3, 37), 25(37, 39-42,44-49), 26(1-3), 27(1-3,45), 28(46, 58a, 65-67, 70), 29(45-46), 30(1, 17, 52a, 53, 55-57, 58a, 58b), 32(1, 17, 56, 59-60, 62), 34(1, 52a), 35(1-3, 52a, 62, 67), 36(52a, 62,65), 37(52a), 38(52a, 62), 39(53, 67, 77, 78), 41(1, 62, 65, 66, 70,

Page 394: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

AUTHOR INDEX, VOLUME 50 37 1

84, 86-90, 92), 42(65), 43(52a, 84, 86, 87), 46(84), 47(92), 48(57,97), 49(70, 84,98), 50(86,98), 52(46,67), 53((113, 115), 54(67), 55(115), 57(113), 58(1, 45,62, 115), 60(46), 61(62), 68(137, 137a, 140, 145, 149), 69(137, 137a, 140), 70(53, 55, 145, 148, 149), 71(137, 149), 72(140), 73(137, 140, 145, 148, 149, 155), 74(145, 154), 75(57, 148, 154), 76(149), 78(140), 79(115, 155), 80(137), 81(92, 115, 137, 140, 154, 155, 164, 165), 82,83(140), 84(57,97, 149), 85(92, 115, 154), 90(57, 148), 91(57, 97), 93(137, 149), 94(137, 170), 95(113), 98(137, 149, 170, 175, 176), 99(137), lOO(113, 115, 154), 104(1, 58b, 175), 105(1,58a, 58b, 176), 106(165), 107(176), 111(1,46, 53,66,

113(192), 114(46, 52a, 52b, 78, 199,

Schneider, H., 212(22), 214(33), 228(33),

Schneider, P., 151(110) Scholtz, S. A., 192(265), 196(265)

78, 190, 192, 197-199), 112(190),

204- 208), 1 16( 1, 3, 17), 289(60)

242(33)

Scholz, D., 211(13), 219-220(97), 222(97), 225-226(97), 241(97), 252(13, 97, 2 18), 262( 13)

Schonbeck, U., 238( 185a) Schotte, H., 14(35) Schotz, M. C., 230(140) Schramek, S., 229(133, 137), 264(133) Schreier, M., 21 l(9a) Schroter, D., 194(270) Schuerch, C., 23(34) Schuhmacher, M., 28(68), 52(68), 1 14(68) Schultheiss-Reimann, P., 287(43), 288(43,

54), 294(43)

256(228), 262(228)

303(87), 306(93), 307(108)

210), 251(70), 261(70)

Schultz, C., 252(219), 254(219), 255-

Schultz, M., 288(58), 293(71), 294-295(74),

Schulz, G., 218(64, 70), 249(70), 250(64,

Schuster, M., 307(105) Schiitze, E., 211(13), 252(13), 262(13)

Schwebel, A., 157( 143) Schweitzer, M. G., 229( 130) Schwentner, J., 1 lO(185)

Schwarcz, J. A., 158-159(150)

Scudder, P., 322(87), 329-331(102, 103) Scudder, P. R., 339(154) Sears, P., 307( 105) Seguchi, T., 344( 18 1) Seitz, S. P., 23(30) Seltmann, G., 263(259) Selvakumar, R., 344(179) Sepulchre, A. M., 129(25,27), 130(27),

Seshadri, R., 154-155(124) Seydel, U., 21 1(5,9a), 212(25), 214(25, 34,

219(97), 220(97), 222(97), 224(79), 225(89, 97, 118), 226(89,97), 228(37, 79, 89), 229(38,79), 230(37, 82), 231(38, 156), 232(82), 233(89), 234(38, 79), 235(37), 236(5, 118, 173), 238(89),

242(38), 243(89, 173, 195a), 244(173), 246(79), 249(34), 250(34), 251(34, 79,

229a), 256(118,230,231), 257(5, 118, 230), 26 1(34,83, 1 1 8), 264(79)

283(31)

198(284)

37-40), 218(34, 79, 82,83, 89),

239(38), 240(37-39), 241(37,97),

83), 252(97), 254(224-226), 255(227,

Shaban, M. A. E., 155(133), 156(134),

Shah, R. H., 130(30-31), 131(31) Shah, R. N., 343(163, 168) Shands, J. W., Jr., 262(256,257) Shaper, M. A., 343(173) Sharaf, S. M., 155(132) Shashkov, A. S., 218(74), 314(38) Shaw, D. H., 21 1(3), 244(3), 256-257(3) Shelton, B., 287(41) Sheth, H. G., 168(204-205), 189(205) Shiba, T., 116(217), 211(5, 15), 214(34-35),

215(15), 217(35), 218(34), 228(35), 236(5, 170, 172), 243(196), 244(200), 247(35), 249(34, 35), 250(34), 251(34), 252(15,35, 172, 196,215,216,219), 254(219), 257(5, 233,234,236,237), 258(15,239), 261(34), 263(15)

Shibasaki, M., 144(74) Shibayama, S., 49(103), 51(103) Shibuya, H., 104-105(178), 109(178) Shibuya, M., 160(164, 165) Shimamoto, T., 211(14), 215(14), 236(170),

252(14, 216), 257(237) Shimauchi, H., 257(236) Shimizu, T., 21 l(18) Shing, T. K. M., 154( 123)

Page 395: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

372 AUTHOR INDEX, VOLUME 50

Shioi, S., 160(164, 165) Shioya, E., 211(15), 215(15), 252(15),

Shiozaki, M., 252(217) Shnyra, A., 255(229b) Shoda, S., 21(9), 22(21) Shogren, R., 344( 180) Shrivastava, V. H., 137(52) Siba, S., 292(66), 294(66), 296(66) Sidorczyk, Z., 21 8(72), 226(72), 228(72),

249(72), 251(72) Sierks, M. R., 19(53) Silberkweit, E., 169(213) Simet, I. M., 344( 179) Simon, E. S., 303(88) Simon, M., 223(112), 249( 112) Sinay, P., 23(36), 52( 109), 69(138), 72(138,

65), 190(256), 239( 188), 287(42a, 42b)

258( I5), 263( 15)

I53), 1 10( 186), 1 16(2 12-2 13), 140(64,

Sindric, R., 153( 1 19) Singh, N. P., 41(86), 43(86), 50(86) Singh, P. P., 2 17(56) Singh, U. C., 343(167) Sinnott, M. L., 116(216), 161(170, 171) Sinnwell, V., 2 19(64a), 224(64a) Sin, D., 344(177) Sjoblad, S., 333(127- 129) Skehel, J. J., 339(153) Slaghek, T. M., 105(179, 180), 109(179, 180) Slessor, K. N., 193(268) Slomiany, A., 316(69,70), 321(69,70) Slomiany, B. L., 316(69,70), 321(69,70) Smith, A. R. W., 231(153) Smith, C. R., 259-260(250) Smith, D. F., 333( 124) Smith, K. D., 314(45), 315(46), 325(45,46) Smith, P. W., 197(281,282) Sodeoka, M., 144(74) Soga, T., 211(15), 215(15), 252(15), 258(15) Soh, C. P. C., 329(98), 333(98) Sohar, P., 163(180) Solomon, J., 314(41), 325(41) Solov’eva, T. F., 218(62) Solter, D., 335( 140) Son, J. C., 197(283) Sonesson, A., 225( 119), 229(119), 230(141),

Sonnichsen, R., 134(44), 173(44) Souchard, I. J. L., 161(170)

232(159, 161)

Spellman, M. W., 314(39), 325(39) Spencer, J. F. T., 230( 150) Sperber, N., 147(142) Spik, G., 324(86), 329(97), 333(86,97),

Spinola, M., 279( 13) Spohr, U., 157(137a), 333(120, 121) Sprung, C. L., 259-260(250) Stacey, G., 219(100) Stacey, M., 31 l(8) Stackebrandt, E., 241(192), 247(203) Stahel, R., 6-7(22) Stellner, K., 31 l(5) Stick, R. V., 127(14), 280(20) Stix, D., 25q210)

343( 169)

Stoll, M. S., 313(26,28), 316(73-75), 321(73-75), 323(28, 74), 324(73), 325(73,75), 328(73-75)

Storer, R., 193(267), 196(279) Strain, S. M., 215(65), 218(63,65,66),

222(66), 226(63), 244(63), 249(66), 250-25 1(65), 263(65)

Stmube, R. C., 259-260(250) Strecker, G., 316(76), 321(76), 324(86),

325(76), 329(76,96,97, 105), 330(76), 333(86,96-97, 130, 131), 334(136), 337(146), 343(169)

233-235(87, 164), 236(170), 238(87), 243(87), 249(87), 258(241)

Strittmatter, W., 2 18(87), 223(87), 228(87),

Stroh, H. H., 155(131) Strominger, J. L., 343(173)

Stumpp, M., 28(66), 30(55), 41(66), 52(66), Strube, K.-H., 329(108)

70(55), 1 I1(66), 114(52b, 66)

225-226(97), 241(97), 252(13,97), 262( 13)

Stiitz, P., 21 1(13), 219-220(97), 222(97),

Stiitz, P. L. Suami, T., 169(209) S ~ g h , H., 143- 144(70) Sugimoto, M., 49-50( loo), 49( 103),

51(103), 56(118), 58(122), 63(132), 82( 132)

Sugiyama, S., 160(164, 165) Suh, H., 193(269) Sun, R.-C., 196(280) Sutter, M., 157(140) Suwinska, K., 191(263)

Page 396: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

AUTHOR INDEX, VOLUME 50 373

Suzuki, K., 21(14, 15) Svendsen, I., 18(48), 19(52) Svensson, B., 18(48, 51), 19(52-53, 55) Svensson, S., 31 l(9) Swahn, C. G., 126- 127(8) Sweeley, C. C., 3 13( 19) Swiderski, J., 161(168, 169) Szab6, L., 211(17), 214(42,44), 218(?3),

222(11 l), 230(139), 232(139), 238(93), 252( 17)

Szab6, P., 211(17), 252(17) Szafranek, J., 41(79) Szejtli, J., 25-27(71) Sznaidman, M., 132(36-37), 133(39),

166(196), 173(222)

T

Tabern, D. L., 151(108) Tachibana, Y., 333( 125) Tacken, A., 222( 1 1 1) Tadanier, J., 143(69) Tadano, K., 169(209) Tadano, K. I., 200(288) Taha, M. A. M., 155( 133), 156( 134) Takada, H., 21 1(14), 215(14), 252(14),

257(236,238) Takahachi, T., 252(214) Takahashi, A., 144(74) Takahashi, H., 127(16), 138(57-58),

Takahashi, I., 257(236) Takano, S., 130(28) Takano, T., 37-38(73) Takayama, K., 214(33, 36, 41, 43), 217(60),

218(63, 88), 222(1 lo), 226(63), 228(33, 60, 88), 232(88, 157, 162), 233(88), 234(88, 166), 235(88, 162), 236(41, 162, 174), 238(182, 184), 239(41), 241(41), 242(33), 243(88, 174), 244(63, 198), 248(60, 174), 249( 110,207), 25 I( 1 lo), 256(229c), 259(248), 264(162, 182, 184)

139( 16,63), 224( 1 15)

Tam, S. Y.-K., 196(280) Tamaru, M., 149(92-93) Tamiya, E., 218(95), 228(95), 243(95) Tanahashi, M., 2 1 1( 1 I), 252( 1 1) Tanaka, A., 257(236) Tanaka, C., 220(103)

Tanaka, S., 211(11, 16), 220(103), 252(11,

Tanamoto, K . 4 , 236( 170), 257(233, 237) Tang, J., 215(69), 218(69), 221(69), 223(69),

Tang, P. W., 329-331(102, 103) Tarentino, A. L., 314(44), 325(44) Tatsuta, K., 1 10( 184) Tavecchia, P., 69(138), 72(138, 153) Taylor, D. P., 220-221(102) Taylor, G. L., 343(167) Taylor-Robinson, D., 339( 154) Tefft, M., 151(109) Tejbrant, J., 288(56), 294(56) Teng, N. N. H., 259-260(250) Termin, A., 70(148), 73(148), 75(148),

Thaisrivongs, S., 183- 184(237), 187(237) Tharanathan, R. N., 218(90), 225(90),

227(90), 234( 165), 242(194), 249(90), 261( 165)

16), 257(236)

227(69), 249(69)

81(168), 90(148)

Thiele, 0. W., 230(147) Thiem, J., 21(13), 110(185), 114(209),

Thierfelder, H., 9- 10(30), 12- 13(30) Thergersen, H., 333(118), 339(158) Thorn, D., 344( 176) Thomas, A., 187(250, 251) Thompson, A., 155(128), 158(147) Thomson, J. K., 163(181), 169- 170(181) Thorpe, S. J., 335(142) Thurin, J., 282(26-27), 288(26), 302(26,

27), 303(84a), 337(147) Timpe, W., 168(203) Titani, K., 314(41), 325(41) Tobias, P. S., 264(263) Todaro, L. J., 196(280) Toepfer, A., 32(62), 35 -36(62), 38(62),

305(92)

41(62), 48(96-97), 58(62), 61(62), 81(165), 84(97), 91(97), 98(176), 105( 176), 106( 165), 107( 176)

Toone, E. J., 303(88) Toromanoff, E., 15 1 ( 109) Toyokuni, T., 44(83), 99(171) Tozer, M. J., 145(79, 80) Tran, V., 343(166) Tranberg, K.-G., 230(141) Trigalo, F., 21 1(17), 252(17) Trimble, R. B., 314(44), 325(44)

Page 397: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

374 AUTHOR INDEX, VOLUME 50

Trippelvitz, L. A. W., 329(100), 333(100) Truchot, A. T., 236( 175) Truelove, J. E., 30(54), 70(54) Trumtel, M., 69(138), 72(138, 153) Triiper, H. G., 241(192)

248(73), 25 l(73) Tsai, T. Y. R., 22(25,26) Tsuchihashi, G.-I., 21(14, 15) Tsuda, Y., 164(185) Tsujimoto, M., 257(236) Tsvetkov, J., 25(49) Tucker, L. C. N., 127(19) Tulloch, A. P., 230( 150) Tuominen, J., 243( 195) Tvaroska, I., 35(76) Tyumenev, V. A., 136(51)

Tsai, C.-M., 218(73), 225-227(73), 242(73),

U

Uchida, K., 231(155) Uchida, T., 22(29), 152(116) Uebach, W., 253(222) Uemura, K.-L,339( 154, 156) Ueno, K., 150(97) Ueno, Y., 169(209) Ugen, K. E., 303(84a) Ugolini, A., 141(67) Uhlenbruck, G., 317(78, 81), 321(78, 81),

Ulevitch, R. J., 264(263) Ulmer, A. J., 21 1(9a, 9b), 236(168, 169),

238(183, 185, 185a), 257(168), 258(168, 183, 185), 263(185), 264(169, 183,266)

324(8 1)

Ulrich, J. T., 236(175) Umemoto, T., 261(268) Umemura, K., 149(95), 150(96) Umezawa, S., 1 lO(184) Unger, F. M., 214(34), 218(34,64, 70),

249(34, 70), 250(34,64,209,210), 251(34, 70), 261(34, 70)

Unverzagt, C., 280( 19a- 1 9c), 283( 19a, 19c, 34,46), 284(34, 36), 285(19a, 19c, 34, 36), 286(19a, 19c), 294(19a, 19c), 299(19a, 19c), 302(81), 303(90)

Upson, F. W., 151(100) Urbanik-Sypniewska, T., 2 18(82,92),

219(100, 101), 225-226(92), 228(84,

92), 230(82), 232(82, IOl), 241- 242(92), 247(101)

303(84a, 84b) Urge, L., 282(26-27), 288(26), 302(26,27),

V

Vaara, M., 227(125), 243(195, 195a), 263(260)

Vaara, T., 227(125) Valle, S., 182(235) van Alphen, L., 230(143), 235(143) van Boeckel, C. A. A., 25(43), 43(81),

211(12), 252(12) van Boom, J., 25(43) van Boom, J. H., 71(151), 73(151), 144(75),

van Cleve, J. W., 22(22) van den Eijnden, D. H., 329(100), 333(100) van der Marel, G. A., 71(151), 73(151),

van Halbeek, H., 314-315(31, 32), 316(57,

180(232), 211(12), 252(12)

144(75)

61-63,65-68), 321(61-63, 65-68), 325(31-32, 57,91), 329(95, 96, 101), 333(31-32,62,96, 101), 335(144)

van Kuik, J. A., 314(36), 329-330(107) van Marle, T. W. J., 155(127) van Oijen, A. H., 105(179), 109(179) van Steijn, A. M., 52-53(11 I), 57-58(111) van Zuylen, C. W. E. M., 180(232) Vankar, P. S., 39(78), 11 1(78), 114(78) Vankar, Y. D., 39(78), 1 I1(78), 114(78) Varela, O., 132(38), 148(89), 160(158, 161-

162), 163( 18 l), 164( 158), 165( 19 I),

168(199?, 202), 169(181), 170(181, 194), 171(220), 172(194,202), 173(194, 200), 174(225), 179(230), 180(231), 18 l(38)

115(211), 157(135, 136)

166( 192, 194- 195), 167(200),

Vasella, A., 70(147), 114(210, 21 l),

Veeneman, G. H., 144(75), 180(232) Vekemans, J. A. J. M., 127(15), 167(197),

169(211), 182(15), 183(211), 200(197) Veleva, K., 258(241) Verner, I. K., 2 18(74) Verret, R. C., 239(189, 190) Vethaviyasar, N., 22(20) Veyritres, A., 68( 139), 69( 138, 139),

72(138-139, 153), 73(139, 159),

Page 398: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

AUTHOR INDEX, VOLUME 50 375

78(139, 159), 81(159), 82-83(139), 84( 159)

Vicari, G., 316(59) Vilamjo, L., 182(235) Ville, G., 139(62) Vliegenthart, J. F. G., 52-53(111), 57-

58(1 l l) , 60(126), 63(126), 66-67(126), 105(179, 180), 109(179, 180), 313(21), 314(31,32, 36), 315(31, 32), 316(57,

325(31, 32, 57,91), 329(96, 101, 107, 112), 330(107), 333(31, 32,62,96, 101, 112), 334(136), 335(144), 337(146)

61-63,65-68), 321(61-63,65-68),

Vogel, H. J., 343(172) Vogel, S. Vogt, K., 193(267), 196(279) Voigt, H., 125- 127(3) Voigt, J., 125-127(3) Volk, W. A., 218(78), 234(78), 246(78) von dem Bruch, K., 278( 1 l), 286( 1 I),

Vorgel, E., 253(222) Voshol, H., 329(112), 333(112) Vuorio, R., 263(260) Vyplel, H., 211(13), 252(13, 218), 262(13)

306(100)

W

Walding, M., 22(19) Waldmann, H., 11 l(194, 196), 278(9),

279(14-16), 283(14-16, 32, 33, 35- 37), 284(15, 32, 33, 35, 36), 285(35- 36), 286(9, 37), 287(16,46), 289(32), 291(68), 294(32, 33,68), 297(68), 301(32), 303(46, 85, 86), 307(103)

Waldstiitten, P., 218(70), 249(70), 251(70), 261(70)

Walker, R., 169(212) Walker, T. E., 132(35) Wallis, C. J., 193(267), 196(279) Walter, B., 288(58) Walton, D. T., 287(41) Walton, E., 195(274) Wand, A. J., 337(149) Wang, M.-H., 238(183, 185a), 258(183),

264(183,266) Wang, P., 307( 105) Wang, R., 214(43), 218(94), 228(94),

232( 162), 235-236(162), 241(94), 264( 162)

Wang, Y., 193(267), 196(279) Ward, J., 236(175) Warren, C. D., 279(18), 283(18), 306(102),

343( 169) Wartenberg, K., 227(124), 230(145),

235( 145) Watanabe, K., 261(268), 339(157) Watanabe, T., 49(108), 150(98) Watkins, W. M., 316(49), 329(98), 333(98) Webber, J. M., 125(4), 131(32) Weber-Molster, C. C., 127( 18) Weckesser, J., 215(49, 50), 218(82, 84, 87,

89-92)), 219(49, 50,96), 220-221(96), 223(87), 224(91, 114), 225(89-92), 226(89,92), 227(90), 228(84, 87, 89, 92), 230(82), 232(50,82), 233(87, 89,

164), 238(87,89), 241(92), 242(84,92, 194), 243(87, 89), 249(87, 90), 255(229a), 261(165,270), 264(265)

164), 234(87, 164- 165), 235(87,91,

Wedel, N., 259-260(251) Weerman, R. A., 15 I ( 103) Wegmann, B., 28(67), 30(58b), 35(67),

39(67), 52(67), 54(67), 98(175), 104- 105(58b), 104(175)

290(64), 302(55) Weichert, U., 53(112), 57(112), 288(55),

Weidmann, H., 168(203), 194(272) Weigandt, H., 58( I 19), 60( 1 19), 73( 1 19) Weigel, T. W., 171(219) Weiner, D. B., 303(84a) Weintraub, A., 218(79), 224(79), 225(117),

228-229(79), 234(79, 117), 243(79), 246(79), 251(79), 264(79)

Weintraub, S. T., 218(94), 228(94), 241(94) Weisburg, W. G., 247(203) Weiss, J., 263(261) Weisshaar, G., 329(99, 109), 333(99) Weisshaar, R., 221( 105), 230(105), 252(105) Wells, W. W., 313(19) Welsh, E. J., 344(176) Welte, W., 152(114) Wendorf, P., 314(40), 325(40) Wernig, P., 293(71), 294-295(73), 297-

Westerduin, P., 2 1 1( 12), 252( 12) Westheimer, F. L., 1 16(2 18) Westphal, O., 211(1,2,4-5), 213(2,4, 28),

298(73)

217(1, 28, 54), 218(1, 4, 54), 225(4), 228(4), 229(128), 234(1), 236(5, 128,

Page 399: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

376 AUTHOR INDEX, VOLUME 50

170), 237(177, 179), 246(4,201), 247(4), 256(1,4), 257(1,4, 5,28,232, 234)

Wethers, S. G., 161(172) Wheat, R., 213(30), 217(30), 230(30) Whitesides, G. M., 303(88), 339(153) Widemann, C., 219(98), 230( 145), 235(145) Widmalm, G., 314(37) Wiemann, T., 305(92) Wieruszeski, J.-M., 3 16(76), 32 1(76),

324(86), 325(76), 329(76, 105), 330(76), 333(86, 130- 131)

Wiesner, K., 22(25-26) Wilcox, C., 193(269) W~~COX, C. S., 143- 144(70), 183- 184(237),

187(237) Wiley, D. C., 339(153), 343(173) Wilkinson, B. J., 234( 163) Wilkinson, S. G., 218(76), 220-221(102),

Willard, J. J., 31 l(8) Williams,T.J., 151(111, 112) Williamson, J. M., 240(191), 262(191) Wilson, F. X., 193(267), 196(279) Wilson, S., 329(95) Winchester, B., 127(20), 133(20), 199(20),

Winckler, F. W., 6(15) Windholz, T. B., 286(38) Windmuler, R., 81(161), 83(161), 105,

Wittkiietter, U., 149(90), 162( 178) Witty, D. R., 193(267), 196(279) Witty, R., 154(122), 198(122) Woese, C. R., 247(203) Wolf, N., 168(203) Wolfe, M. S., 196(277, 278) Wolfrom, M. L., 148(86), 151(86, 102,

Wollenweber, H.-W., 212(25), 214(25, 35,

229(127, 131), 230(127), 234(127, 163)

200( 286 - 288)

106(177)

105-106), 155(128), 157(145), 158(147)

37-38), 217(35), 218(79), 224(79), 228(35, 37,79, 187), 229(38, 79, 137), 230(37, 142), 231(38), 234(38, 79), 235(37, 142), 236(176), 238(176, 187), 239(38, 187), 240(37, 38, 187), 241(37), 242(38), 243(37,79, 187), 246(79), 247(35, 187), 249(35), 251(79), 252(35), 264(79)

Wong, C.-H., 307( 105) Wong, C. T., 339(158)

Wong, R., 222(1 lo), 249(1 lo), 251(110) Wong, T. C., 333( 119) Wood, H. B., 148(86), 151(86), 157(145) Woodward, H. D., 344(179) Woodward, R. B., 22(27) Wortel, C. H., 259-260(250) Wray, V., 223(113), 226-227(113) Wright, D. E., 149(91) Wright, D. J., 314(33), 323(33), 327(33),

Wright, S. D., 264(263) Wroblewski, K., 282(26,27), 288(26),

Wu, A. M., 316(54, 57), 325(57), 329(54)

Wulff, G., 35(74) Wurzburg, B. A., 339(153) Wuts, P. G. M., 22(28)

Y

332-333(33)

302(26-27), 337(147)

WU, S.-S., 316(58)

Yamada, Y., 219(99), 224(99) Yamagiwa, Y., 49( 108) Yamamoto, A., 257(237) Yamamoto, M., 236(170) Yamasaki, R., 212(22), 339(151) Yamashina, I., 317(77), 321(77) Yamashita, K., 314(43), 325(43), 333(125) Yamazaki, F., 60(127), 66(127), 68-

69( 141 - 144), 70( 141 - 142), 73( 143, 144), 89(127, 142-143)

Yan, Z.-Y., 334( 135) Yanaghara, Y., 21 l(18) Yano, I., 230(146) Yasuda, T., 257(237) Yokota, A., 215(50), 219(50,99), 221(107),

224(99), 232(50) Yoshida, M., 264(264) Yoshikawa, M., 104-105(178), 109(178) Yoshikawa, T. T., 230(140) Yoshimoto, K., 164(185) Yoshimura, H., 244(200), 252(2 15),

Yoshimura, J., 149(92-95), 150(97,98) Yoshimura, S., 257(234) Yurewicz, E. C., 317(79), 321(79)

257(236-237)

Z

Zabel, V., 152( 1 14) Zach, K., 13- 14(34)

Page 400: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

AUTHOR INDEX, VOLUME 50 377

Zahringer, U., 21 l(9, 9a, 9b), 212(25), 214(25, 34, 39), 218(34,64a, 68, 72,

222(68,97), 223(68), 224(64a, 77, 79), 73, 75, 77, 79, 83), 219-220(97),

224(77,79), 225(68,73, 77,97, 116- 118), 226(72-73,97), 227(73), 228(68, 72,77,79), 229(77,79), 231(77, 156), 232(159, 161), 234(79, 117), 236(118, 170), 240(39), 241(68, 97), 242(77), 242(73, 77), 243(77,79, 173), 244(77, 173), 245(77), 246(79), 248(73, 77), 249(34, 72), 250(34), 251(34,68, 72, 73, 75, 77, 79,83,211,212), 252(97), 256( 1 l8,229d), 257( 1 16, 1 18), 258(116, 118),261(34,68, 75, 83, 118), 264(77, 79)

Zamojski, A., 190(255) Zamze, S. E., 225(120), 231(153) Zaugg, H. E., 147(142) Zbedska, E., 316(69-70), 321(69,70) Zbiral, E., 133(40)

Zeikus, J. G., 16(38) Zempkn, G., 16(37), 19(37) Zen, S., 22(29) Zervas, L., 169(213), 277(1) Zhdanov, Y. A., 136(50, 51), 139(59) Ziegast, G., 152( 115) Ziegler, E. J., 259-260(250) Zimmer, J., 306(101) Zimmerman, M., 195(274) Zimmermann, P., 28(70), 41(70, 84,85),

43(84), 46(84-85), 49(70, 84, 85), 52(85), 53(113), 57(113), 95(113), 100( 113)

Zinner, A. H., 125-127(3) Zissis, E., 165(189) Zitrin, C. A., 161(166) Zollo, F., 105(182) Zollo, P. H. A., 139(64) Zopf, D. A., 316(55), 333(124) Zucchelli, L., 140(66) Zurabyan, S. E., 28 l(22)

Page 401: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

SUBJECT INDEX

A

Abequose, 173-174 Acetalation, 125 - 126 2-Acetamido-2-deoxyaldohexoses, 164- 165 2-Acetamido-2deoxy-~~-glucosyl- a as par-

agine, linkage with N-glycopeptides,

anomeric azides as precursors, 278-280 1 -hydroxy-D-glycosamines reactions with

ammonium hydrogencarbonate,

278-283

282-283 in situ anomektion, 280-281 1-isocyanates as precursors, 28 1-282

3-Acetoxy-6-acetoxymethylpyran-2-one,

3-Acetoxy-5-methylen-2(5H)-furanone, 168 1 -0-Acetyl, activation, 3-0-glycosidic link-

N-Acetylgalactosamine, trichloroacetimi-

N-Acetylglucosamine, naturally occurring

N-Acetyllactosamine, trichloroacetimidates,

2-Acylamido-hex-2-enonolactones, 169 - 170 3-Acylamido-2-pyrone, 165 - 166 Acylation, aldonolactones, 132 - 134 Alcohols

165-166

age formation, 292-293

dates, synthesis, 84, 93-98

glycosidic linkages, 6 1,68

synthesis, 73, 78 - 80

as 0-nucleophiles, see 0-Nucleophiles reaction with aldonolactones, 149- 15 1

Aldehydes, reaction with aldonolactones,

Alditols, aldonolactone reduction to, 157 -

Aldohydroximo-lactones, 156 - 157 Aldonamides, preparation, 15 1 Aldonolactones, 125 - 201

125-127

161

acylation and etherification, 132 - 134 benzoylated, benzoic acid elimination,

p-elimination, 162- 166 carbonyl group, chain elongation, 136 -

167- 168

148 formylaminomethylenation of aldono-

methylenation of aldonolactones, 143 -

reaction with organomagnesium and

lactones, 146- 148

146

organolithium reagents, 138- 143

FOR VOLUME 50

Reformatsky-type reactions, 136 - 138 as chiral precursors for synthesis of natu-

ral products, 1 8 1 - 20 1 y-butenolides, 182- 190 Gbutenolides, 190- 192 nucleosides, amino acids, and other N-

containing products, 195 -20 1 from ~-ribono-l,4-lactone, 192- 195

glycosylation, 179 - 18 1 intramolecular reaction yielding lactams,

lactone group, reaction with 154-155

ammonia and amines, 15 1 - 153 hydrazine and derivatives, 155 - 157

methylenation, 143- 146 reaction with

alcohols, 149 - 15 1 aldehydes, 125-127 hydrogen bromide, 134- 136 ketones, 127- 130

to aldoses and alditols, 157 - 16 1 by borane, 159- 160 isotopic labeling and substitution at an-

omeric center of aldoses, 16 1 - 162 by sodium amalgam, 157

reduction

synthesis of deoxy sugars, 170- 179 use of acetals for sugar derivative synthe-

Aldoses, aldonolactone reduction to, 157 -

0-Alkylation, anomeric, 23 -25 y-Alkylbutanolactones, synthesis, 183 - 184 y-Alkyl-a,pbutenolides, synthesis, 183 - 184 Allyloxycarbonyl group, N-terminal, with

Amicetose, 158 - 159 Amines, lactone group reaction with, 15 1 -

Amino acids, synthesis from aldonolac-

N-(2-Aminoethyl)aldonamides, 152 Aminolactones, intramolecular reaction,

lactam preparation, 154- 155 Ammonia, lactone group reaction with,

Ammonium hydrogencarbonate, reactions with 1-hydroxy-D-glycosamines, 282 - 283

sis, 130- 132

161

tert-butyl ester, 285-286

153

tones, 195-201

15 1 - 153

378

Page 402: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

SUBJECT INDEX. VOLUME 50 379

Amylase, 1 1, 16 Amyloglucosidase, I6 - 17 Angelica lactone

as chiral starting compound, 184 reaction with isoprene, 188 - 189

Anisomycin, 198 Anomeric oxygen-exchange reactions, 2 1 -

1 17; see also Trichloroacetimidate method

0-alkylation, 23-25 Anomerization, in situ, attachment of a-L-

Antibodies, lipid A, 259-260 Antigens, blood-grouprelated, 333 - 337 Aristeromycin, 196 I-Aroyl-2-~-gluconylhydrazides, 155 - 156 5-Aryl-2-ethoxy-3-~-gluconyl-2,3d~hydro-

Ascarylose, 17 1 - 172 Aspergillus niger, glucoamylase, I6 - 1 8 Azides, anomeric, as precursors, 278-280 2-Azido-2-deoxyglucopyranosyl trichloroa-

fucosyl units, 280-28 I

1,3,4-oxadizoles, 155- 156

cetimidates, reaction with nucleophiles, 73-76

2-Azido-2-deoxy-~-mannose derivatives, trichloroacetimidates, glycosylation, 98, 103

Azidosphingosine, derivatives, glycosylation with trichloroacetimidates, 41,45-48

B

Bacteria, gram-negative, lacking lipopoly-

Base-catalyzed rearrangement, bromodeox y

2-Benzamido-4,6-0-benzylidene-2,3-

saccharides, 262-263

aldonolactones, 175- 179

dideoxy-~-erythro-hex-2 -enono- 1,5- lactone, 163

Benzoylation, in excess of pyndine, 164 3-Benzoyloxy-5-ethylidene-2( 5SI)-furanone,

168 3-Benzoyloxy-6-methyIpyran-2-one, 165 -

166 Biology, chemical basis, experiments dem-

onstrating, 8-9 Bis-normaytasinoid, 195 (+)-Blastmycinone, 184 Bromodeoxy aldonolactones

base-catalyzed rearrangement, I75 - 179

hydrogenolysis, 173 - 174 Bromodeoxyaldonolactones, preparation of

aminodeoxy aldonic acids and sugars, 135

0-(glycosyl)trichloroacetimidates, 30- 32

Brernsted acids, reaction with

Butenolide, 169 d-Butenolides, synthetic uses, 190 - I9 1 y-Butenolides, synthetic uses, 182 - 190

double bond reaction, 187 - 188

C

Carbohydrate -protein linkages, amino acid and carbohydrate residues, 277 - 278

Ceramides, glycosylation by trichloroaceti- midates, 49 - 5 1

Chitobiose derivatives, glycosylation, 8 1, 89 as donor, 8 1,89

Chromatography, oligosaccharide physico-

Chromobacterium violaceum, lipid A, 242 (+)-Citreoviral, 193- 194

(+)-trans-Cognac lactone, 186 2,3-O-Cyclohexylidene-~-ribono- 1,4-lac-

chemical analysis, 3 12 - 3 13

CNT, 196- 197

tone, 129-130

D

ddC, 196-197 2-Deoxy-~-~-arabino-hexopyranosides,

3-Deoxy-~-arabino-hexose, I7 I 2-Deoxyhexoses, trichloroacetimidates,

Deoxy sugars, synthesis from aldonolac-

110- I12

110-112

tones, 170- 179 base-catalyzed rearrangement of bromo-

deoxy aldonolactones, 175 - 179 bromodeoxyaldono- 1 $-lactone hydrogen-

olysis, 173- 174 enonolactone catalytic hydrogenation,

170- 173 Deprotection, selective

enzymic, glycopeptide synthesis, 303 N-glycopeptides, 283-287 methods, 294-298

Page 403: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

380 SUBJECT INDEX, VOLUME 50

2,4-Di-O-benzoyl-3,6-dideoxy-~-erythro-

2,6-Di-O-benzoy1-3,5-dideoxy-~,~-threo- 1,4-

Dibenzyl hydrogenphosphate, reaction with

2,5-Di-O-benzyl-~-glycero-pent-2-enono-

Dichloromethylenation, aldonolactones, 145 2,3-Dideoxyhex-2-enono- 1,5-1actone deriva-

1 ,4-Dideoxy- 1,4-imino-~-glucitol, 198 Dideoxylactones, 172 L-1 ,6-Diepicastanospermine, 198 - 199 Difluoromethylenation, aldonolactones,

(2R,35',4R)-3,4-Dihydroxyproline, 197 Diisoamylborane, reduction of aldonolac-

tones, 160- 161 (4S,6S)-4-Dimethyl-tert-butylsilyloxy-6-

[(dimethyl-tert-butylsilyloxy)methyl]-te- trahydro-2H-pyran-2-one, 1 90

hex-2-enono- 1,5-1actone, 164

lactone, 172

a and 8-trichloroacetimidates, 30- 32

1,4-lactone, 168

tives, 190

' 145- 146

E

(+)-Eldanolide, 185 - 186 8-Elimination

aldono- 1 ,Clactones, 166 - 170 aldono-l,5-lactones, 162- 166

following selective deprotection, 294- 298 N-glycopeptides, 283 -287

effect on glucosides, 1 3 - 14 lack of effect on methyl xylosides, I3

/3-elimination, 162- 165 catalytic hydrogenation, 170- 173

active site probing, 13 - 14 introduction of term, 8 specificity, 13 - 15

Elongation, peptide chain

Emulsin, 8, 12

Enonolactones

Enzymes

~-6-Epicastanospermine, 198 - 199 /3-Epinoroxetanocin, 196 ~-Erythrono-l,4-lactone, 13 1 L-Erythrose, synthesis, 130- 13 1 Escherichiu coli, lipid A chemical structure,

Esters, formation, aldonolactones reaction 214-215

with alcohols, 148- 149

Etherification, aldonolactones, 132- 134 Ethynyl compounds, reaction with sugar

lactones, 139- 140

F

Fatty acids, 228-247 amide-bound, 238-239 ester-bound, 236-238 hexaacyl lipid A, 24 1 - 242 (S)-2-hydroxylated, 230- 232 (R)-3-hydroxylated, 229 - 230 linkage to lipid A backbone, 235-239 location at lipid A backbone, 239-240 nonhydroxylated, iso-, and anti-iso, 234 other hydroxylated, 232

primary and secondary, in lipid A, 246 unsaturated, 235

glycosides, 10- 1 1

3-0X0,232-234

Fermentation

SUW, 8- 10 Fischer, Emil

asymmetric induction concept, 2 - 7 probing of enzyme active site, 13- 14 yeast fermentations, 7 -9

Fischer-Helferich method, 23 Fischer - Kiliani synthesis, 6 Fluorine, as leaving group, 21 -22 Formylaminomethylenation, aldonolac-

0-Fucopyranosyl trichloroacetimidates, in- tones, 146 - 148

verse procedure for glycosylation, 98, 104- 107

Fucose, trichloroacetimidates reaction with N-nucleophiles, 105- 107 synthesis, 98, 104

~ - L - F u c o s ~ ~ units, attachment, in situ ano-

2( 5H)-Furanones, &elimination, 166 - 170 merization, 280-281

G

Galactosamine trichloroacetimidates glycosylation, 98- 102 as glycosyl donors, 84,92- 102

O-Galactosyl trichloroacetimidates acetylated, glycosylation with, 53, 56-57 benzylated, glycosidation, 53 - 55 as donors, 49, 52 - 59

Page 404: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

SUBJECT INDEX, VOLUME 50 38 1

glycosylation with sphingosine deriva- tives, 58-59

synthesis, 52-53 &~-Galf-( 1 - 5)-D-Galf; 1 80 - 1 8 I Glycosylation, 0-fucopyranosyl trichloro-

acetimidates, inverse procedure, 98, 104- 107

Glucoamylase, 16 Gluconamides, 15 1 a-D-Glucopyranoside, hydrolysis by gluco-

Glucosamine amylase, 17 - 18

as donors, 61,68-77 trichloroacetimidates, as glucosyl donors,

60-61,68-91 chitobiose donors, 8 1, 89 glucosamine donors, 6 1,68 - 77 lactusamine donors, 73,78-88 muramic acid donors, 8 1, 84,90 - 9 1

thesis, 68 - 7 1 D-Glucosamine, trichloroacetimidates, syn-

dextro-Glucose, structure, 5 D-Glucose, trichloroacetimidates, synthesis,

Glucosides 27 - 29

emulsin effect, 13- 14 synthesis, 2 1 - 1 17

anomeric 0-alkylation, 23 - 25 P-D-Glucosiduronates, synthesis, 1 1 1, 1 13 0-clucosyl trichloroacetimidates

acetylated, glycosides and saccharides from, 4 1,43 -44

benzyl-protected, 35 - 39 as donors, 34-49

acid catalyst, 35 glycosylation, 39-40

azidosphingosine derivatives, 4 1,45 -

ceramides, 49 - 5 1 solvent effects, 35, 39

48

synthesis, 34-35 Glucuronic acid, trichloroacetimidates, 1 1 1,

Glycoconjugates, biological significance, 2 1 Glycolipids, synthesis, 15 1 - 152 Glycopeptide n-heptyl esters, lipase-cata-

lyzed cleavage, 306-307 Glycopeptides, 277 - 307

binding to proteins, 298-299 synthesis, 277- 307

113

enzymes as tools, 303 - 306 lactosamine importance, 73 solid-phase, 299 - 303, 306

N-Glycopeptides, 278 -287 2-acetamido-2-deoxy-~~-glucosyl- L-

asparagine linkage, 278-283 selective deprotection and peptide-chain

elongation, 283-287 3-O-Glycopeptides, L-serine or L-threonine,

selective deprotection methods, 294 - 298 3-0-glycosidic linkage formation, 287 -

287-298

294 G1 ycoproteins

with multiple 0-glycosylation sites, 344 N- and 0-linked chains, peripheral substi-

oligosaccharide determinants, 3 1 1 - 345 tutions, 325,239-332

backbones and core regions N- and 0-linked chains, secreted and

plasma membrane glycoprotein,

'H-NMR-spectral and mass-spec- trometric analysis, 322- 328

mucin oligosaccharide structure,

315-325

3 16-322 conformations and molecular recogni-

determinants adjacent to protein

determinants distant from core, 332-

blood-group-related antigens, 333 -

sialylated oligosaccharide determi-

tion

moiety, 343 - 345

342

337

nants, 337 - 342 purification and proliling, 3 14 - 3 1 5 structural analysis methods, 3 1 1 - 3 14

sulfated oligosaccharide chains, 'H- NMR - spectral and mass-spectro- metric analysis, 330-331

Glycosidation, benzylated 0-galactosyl trichloroacetimidates, 53 - 55

G1 ycosides from acetylated glucosyl trichloroacetimi-

dates, 41,43-44 fermentation, 10- 1 1 from mannosyl trichloroacetimidates, 58,

62-65 0-Glycosides, synthesis, 296 -297

Page 405: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

382 SUBJECT INDEX. VOLUME 50

3-0-Glycosidic linkage formation, 287 -294 1 -0-acetyl activation, 292 -293 electrophile-induced lactonization of gly-

glycosyl fluorides, 290 Koenigs-Knorr methods, 287-289 1-thioglycosides, 290-292 trichloroacetimidate method, 289 -290

N-acetylgalactosamine-containing, struc-

synthesis, lactosamine importance, 73

acetylated 0-galactosyl trichloroacetimi-

acetylated 0-mannopyranosyl trichloro-

aldonolactones, 1 79 - 18 1 azidosphingosine derivatives, with tri-

chloroacetimidates, 4 1,45 -48 ceramides, by trichloroacetimidates, 49 -

51 chitobiose derivatives, 8 1, 89 galactosamine trichloroaetimidates, 98 -

102 0-galactosyl trichloroacetimidates, with

sphingosine derivatives, 58 -59 0-glucosyl trichloroaetimidates, 39 -40 glycopeptide synthesis, 303 -306 solvent effects, 35,39 steps, 26 trichloroacetimidates of 2-azido-2deoxy-

D-mannose derivatives, 98, 103 Glycosyl carboxylates, base-catalyzed addi-

Glycosyl fluorides, 3-0-glycosidic linkage

Glycosylic phosphate, lipid A backbone,

Glycosyl imidates, base-catalyzed addition,

Glycosyl oxides, base-catalyzed addition, 1 14 Glycosyl4-pentenoates, electrophile-in-

duced lactonization, 293 - 294 Glycosyl phosphates, anomeric-oxygen acti-

vation, 116 Glycosyl region, lipid A backbone, 2 18 -22 1 Glycosyl sulfonates, base-catalyzed addition,

cosy1 4-pentenoates, 293 -294

GI ycosphingolipids

tures, 84, 92

GI ycosylation

dates, 53, 56-57

acetimidates, 58,66-67

tion, I16

formation, 290

221 -222

114, 116

116

0-Glycosyl trichloroacetimidates, reactions with N-, S-, C-, and P-acceptors, 1 1 1, 114- 115

Glycoyl a-phosphate, lipid A substituents, 226-227

9-/3-~-Gulofuranosyladenine, 195

H

Hanganutziu-Diecher antigen, 339,342 Hard-sphere exoanomeric algorithm, 333 -

Hept-2-enono- 1,4-1actone, 167 Heterocycles, lithiated, addition to aldono-

Hex-2-enono- I ,4-lactone 2-mesylates, 167 Hydrazine, lactone group reaction with,

Hydrogenation, catalytic, enonolactones,

Hydrogen bromide, reaction with aldono-

Hydrogen01 ysis, bromodeoxyaldono- 1,4-lac-

Hydrolysis

335

lactones, 138 - 139

155-157

170-173

lactones, 134- 136

tones, 173 - 174

enzymic, structure and configuration ef-

a-D-glucopyranoside by glucoamylase,

a-Hydroxy acids, cyanohydrin synthesis, 6 8-(Hydroxyalkyl)adenines, 153 1 -Hydroxy-D-glycosamines, reactions with

ammonium hydrogencarbonate, 282 - 283

fects, 12

17- 18

Hydroxyl group, lipid A backbone position 4', 247-249 position 6', 249-252

I

Induction, asymmetric, 2-7

Invertase, 11 Invertin, 8, 12 1-Isocyanates, as precursors, 28 1-282 Isopropylidenation, aldonolactones, 127 2,3-~-Isopropylidene-~-ribono- I ,Cladone,

discovery, 6 - 7

129

Page 406: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

SUBJECT INDEX, VOLUME 50 383

K

Ketones, aldonolactones, 127 - 130 Kinetic anomeric effect, 29

Koenigs-Knom method, 21 3-0-glycosidic linkage formation, 287-

289

L

Lactams, from aldonolactone intramolecu-

Lactase, 12 Lactone group, reaction with hydrazine and

Lactonization, electrophile-induced, glyco-

Lactosamine

lar reaction, 154- 155

derivatives, 155 - 157

syl-4-pentenoates, 293 - 294

2-azido-2-deoxytrichloroacetimidates, re- action with nucleophiles, 8 1, 85

as donors, 73,78-88 oligomers, synthesis, 8 1, 84

(+)-Lineatin, synthesis, 193 Lipid A, 2 1 1 - 265

amphiphilic nature, 2 13 amphoteric nature, 2 13 antibodies, 259-260 backbone, 2 16 - 225,260

glycosyl region, 2 18 -22 1 hydroxyl group at position 4’, 247 -

hydroxyl group at position 6‘, 249-

phosphate groups, 221 -225 principles and structures, 216 structural elucidation, 2 17 -2 18

249,26 1 -262

252,262

conformation, 252-256 definition, 2 13 endotoxic activity, 263 - 264 fatty acids, see Fatty acids heptaacyl, 243 hexaacyl, 241 -242 as lipopolysaccharide endotoxic center,

256-258 molecular shape, 253-254 pentaacyl, 243-245 phase states, 258 phosphate substituents, 225 -227 properties, 2 1 3 - 2 14 serology, 258-260

structure, 260-26 1 chemical, 214-215 primary, 214-216

synthetic, 252 tetraacyl, 244 three-dimensional organization, 256-258

Lipopolysaccharides, 21 1-265, see also Lipid A

core oligosaccharide, 2 12 lipid A as endotoxic center, 256-258 0-specific chain, 2 12 structural principles, 2 1 1 - 2 12

Lithium divinylcyanocuprate, 1 ,Cconjugate

Lithiumtrimethylsilyl acetate, C- 1 elonga-

Lock and key concept, 9- 13 9-a-~-Lyxofuranosyladenine, I 95

addition, 184- 185

tion of aldonolactones, 144

M

0-Mannopyranosyl trichloroacetimidates acetylated, glycosylation, 58,66 -67 as donors, 58,60-67

glycosides and saccharides from, 58,

reactions with, 58,6 1

glycosyl donors, 98, 103

62-65

Mannosamines, trichloroacetimidates, as

Mass spectrometry mucin oligosaccharide chains, 322-328 oligosaccharide physicochemical analysis,

312-313 2’-C-Methyladenosine, 195 Methylation, oligosaccharide of glycoconju-

Methylenation, aldonolactones, 143 - I46 Methyl D-gdactofuranosyl-( 1 -+ 6)-/?-~-

Methyl a-D-glucopyranoside, 16 - 17 Methyl oxetane-2-carboxylate, 193 Mucins

gates analysis, 3 1 1 - 3 13

galactofuranoside, 180

backbone and core sequences, 3 16 - 322 oligosaccharide structure, 3 I6 - 322

IH-NMR- spectral and mass-spectro- metric analysis, 322 - 328

Muramic acid, glycosides, synthesis, 8 1, 84,

Muramic acid, as donor, 8 1,84,90-9 1 90-91

Page 407: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

384 SUBJECT INDEX, VOLUME 50

L-MYCXO~~, 158 - 159 Mycoplasm pneumoniae, binding specificity,

339

N

(+)-Negamycin, 201 Neplanocin A, 196 Nojirimycin, 154- 155 Nonglycosylic phosphate, lipid A backbone,

j?-Noroxetan&n, 196 'H-Nuclear magnetic resonance, oligosac-

223 - 224

charide physicochemical analysis, 3 12, 314

mucin oligosaccharide chains, 322-328 sulfated oligosaccharide chains, 330,333

0-Nucleophiles, alcohols and sugars as, 32- 1 14; see also O-Glucosyl trichloro- acetimidates, as donors

0-galactosyl trichloroacetimidates, 49,

trichloroacetimidates 52-59

of 6-deoxyhexoses, 98, 104- 110 of galactosamine derivatives, 84,92 -

102 of glucosamine derivatives, see Gluco-

samines, trichloroacetimidates of mannosamine derivatives, 98, 103

0-mannopyranosyl trichloroacetimidates, 58,60-67

Nucleophilicity, enhanced, /.?-oxides, 29 Nucleosides, synthesis from aldonolactones,

195-201

0

Oligosaccharide chains, 0-linked, glycopro-

Oligosaccharides teins, 322, 324

conformations, 335 - 337 glucosyl trichloroacetimidates, reaction,

with Le" determinants, synthesis, 8 1, 86 -

much structure, 3 16 - 322

41 -42

88

'H-NMR-spectral and mass-spectro- metric analysis, 322 - 328

physiochemical analysis, 3 1 1 - 3 14 recognition specificity of proteins, 17

sequences related to Le" and Leb antigens,

sialylated, determinants, 337 - 342 Olivomycose, 158 - 159 Organic chemistry, in 19th century, 5 Organolithium reagents, reaction with al-

donolactones, 138- 143 Organomagnesium reagents, reaction with

aldonolactones, I38 - 143 Orthoesters, derivatives, formation, aldono-

lactones reaction with alcohols, 149- 15 1 /.?-Oxides, enhanced nucleophilicity, 29

337,340-341

P

Pasteur, Louis, tartaric acid preparation, 3 Penten-5-olides, 190 3-(~-gluco-Pentitol- 1 -yl)-Sphenyl- 1,2,4-tra-

zol[3,4-a]phthalazine, 156 Pentoses, trichloroacetimidates, 1 1 1 Peptide chain, elongation

following selective deprotection, 294-298 N-glycopeptides, 283 -287

groups, lipid A backbone, 22 1 -225 substituents, lipid A, 225-227

Phosphate

4'-Phosphate, non-glycosylic, lipid A sub- stituents, 226

2-N-Phthaloyl-2deoxytrichloroacetimidate, reaction with nucleophiles, 8 1 - 83

2-N-Phthaloyl trichloroacetimidate, reaction with nucleophiles, 68,72-76

2-Polyhydroxyalkylimidazoles, 152 Proteins

glycopeptide binding, 298-299 oligosaccharide recognition, specificity, 17

Pseudomonas aeruginosa, lipid A, 244- 245 cdyrones, /.?-elimination, 165 - 166

Q 0-Quinovopyranosyl trichloroacetimidates,

105, 110 Quinovosides, 105

R

Ranunculin, synthesis, 183 Reformatsky-type reactions, aldonolactone

carbonyl group, 136- 138

Page 408: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

SUBJECT INDEX, VOLUME 50 385

0-Rhamnopyranosyl trichloroacetimidates,

Rhamnosides, 105 Rhodinose, 158 Rhodobacter capsulatus, lipid A, 232-234 Rhodobacter sphaeroides, lipid A, 232 - 234 L-Ribofuranose, derivatives, synthesis, 132 Ribonolactone, p-elimination, I89 D-Ribonolactone

105, 108-109

0-benzylidene derivatives, physical con- stants for reassigned structures, 126- 127

syntheses from, 192- 195

S

from acetylated glucosyl trichloroacetimi-

from mannosyl trichloroacetimidates, 58,

Saccharides, synthesis, 21 -22

dates, 4 1,43 - 44

62-65 Salmonella typhimurium, lipid A, 217 Serology, lipid A, 258-260 Sialic acid, substitution patterns, 'H-NMR

analysis, 331 -332 Sodium amalgam, aldonolactone reduction,

I57 Somatostatin, glycosylated, 299 Sphingosines, derivatives, trichloroacetimi-

date glycosylation with, 58-59 swFs

derivatives, synthesis, aldonolactones ace-

fermentation, 8 - 10 labeled at anomeric center by aldonolac-

tones, 161 - 162 as 0-nucleophiles, see 0-Nucleophiles relative configurations, proof by Fisher, 2

tals use, 130-132

T

Thioglycosides, as glycosyl donors, 22

1 -Thioglycosides, 3-0-glycosidic linkage for-

2,5,6-Tri-0-acetyl-3-deoxy-~-rib~hexono-

Tri-0-benzylfucosyl donor, 98, 104 Trichloroacetimidate, 0-glycosyl, forma-

PTrichloroacetirnidate, a-selective glycosi-

Trichloroacetimidate method, 25 - 1 15

mation, 290-292

1 ,4-lactone, 176

tion, 25 - 30

dation, 13, 77

alcohols and sugars as 0-nucleophiles, see 0-Nucleophiles

kinetic anomeric effect, 29 3-0-glycosidic linkage formation, 289 -

0-glycosyl trichloroacetimidate forma-

reaction with Brernsted acids, 30-32

290

tion, 25 - 30

2,2,2-Trichloroethoxycarbonyl group, 286- 287

U Umbelactone, 188 - 189

V

van't Hoff-Le Be1 theory, asymmetric car- bon atom, 3 -4

X

Xylosides, methyl, lack of effect by emulsin, 13

Y

Yeast enzyme presence, 11 fermentations, 7 - 9

Page 409: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

CUMULATIVE AUTHOR INDEX FOR VOLS. 46-50

A

ANGYAL, STEPHEN J., Complexes of Metal Cations with Carbohydrates in Solu- tion, 47, 1-43; The Composition of Reducing Sugars in Solution: Current Aspects, 49, 19 - 35

AuGB, CLAUDINE. See David, Serge

B

BIERMANN, CHRISTOPHER J., Hydrolysis and Other Cleavages of Glycosidic Linkages in Polysaccharides, 46,25 1 - 27 1

BLEHA, TOMAS. See TvaroSka, Igor

C

CARTER, DOUGLAS R. See Dill, Kilian CLARKE, RONALD J., COATES, JOHN H.,

and LINCOLN, STEPHEN F., Inclusion Complexes of the Cyclomalto-oligosac- charides (Cyclodextrins), 46,205 - 249; Addendum, 46, 333-335

COATES, JOHN H. See Clarke, Ronald J. CSUK, RENB, and GLANZER, BRIGITTE I.,

N.M.R. Spectroscopy of Fluorinated Monosaccharides, 46,73 - 177; Adden- dum, 46,331 -332

D

DAVID, SERGE, AuGB, CLAUDINE, and GAUTHERON, CHRISTINE, Enzymic Methods in Preparative Carbohydrate Chemistry, 49, 175-231

EDUARDO G., [Obituary of] Venancio DE LEDERKREMER, ROSA M., and GROS,

Ions with Carbohydrates: Use of Relax- ation Probes, 47, 125- 166

F

FERRIER, ROBERT J. See Somdk, Liszl6

G

GARG, HARI G., and LYON, NANCY B., Structure of Collagen Fibril-Associated, Small Proteoglycans of Mammalian Origin, 49,239-261

STEN, and KUNZ, HORST, Develop- ments in the Synthesis of Glycopetides Containing Glycosyl L-Asparagine, L-

Serine, and L-Threonine, 50,271 - 3 10 GAUTHERON, CHRISTINE. See David, Serge GLANZER, BRIGITTE I. See Csuk, Rent GROS, EDUARDO G. See de Lederkremer,

GARG, HARI G., VON DEM BRUCH, KAR-

Rosa M.

H

HARDING, STEPHEN E., The Macrostructure of Mucus Glycoproteins in Solution, 47,345-381

HEMMER, REINHARD. See Stoss, Peter HICKS, KEVIN B., High-Performance Liquid

Chromatography of Carbohydrates, 46, 17 - 72; Addendum, 46,327 - 329

HORTON, DEREK., [Obituary of] R. Stuart Tipson, 50, xiii-xxi; see also Tsuchiya, Tsutomu

HOUNSELL, ELIZABETH F., Physicochemical Analyses of Oligosaccharide Determi- nants of Glycoproteins, 50,3 1 1 - 350

K

KASHIMURA, NAOKI. See Komano, Tohru KINZY, WILLY. See Schmidt, Richard R. KNIREL, YURIY A,, VINOGRADOV, EVGENY

V., and MORT, ANDREW J., Applica- tion of Anhydrous Hydrogen Fluoride

Deulofeu, 46, 1 1 - 15 DE LEDERKREMER, ROSA M., and VARELA,

OSCAR, Synthetic Reactions of Aldono- lactones, 50, 125 -209

')C-Nuclear Magnetic Resonance-Spec- tral Studies of the Interactions of Metal

DILL, KILIAN, and CARTER, R. DOUGLAS,

* Starting with Volume 30, a Cumulative Author Index covering the previous 5 volumes has been published in every 5th volume. That listing the authors of chapters in Volumes 1 - 29 may be found in Volume 29.

386

Page 410: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

CUMULATIVE AUTHOR INDEX FOR VOLS. 46-50 387

in the Structural Analysis of Polysac- charides, 47, 161-202

NAOKI, [Obituary ofj Konoshin Ono- dera, 46, 1 -9

KOMANO, TOHRU, and KASHIMURA,

KUNZ, HORST. See Garg, Hari G.

L

LEGLER, GUNTER, Glycoside Hydrolases: Mechanistic Information from Studies with Reversible and Irreversible Inhibi- tors, 48, 3 19 - 384

LEMIEUX, R. U., and SPOHR, U., How Emil Fischer Was Led to the Lock and Key Concept for Enzyme Specificity, 50, 1 - 20

LINCOLN, STEPHEN F. See Clarke, Ronald J. LINDBERG, BENGT, Components of Bacte-

rial Polysaccharides, 48,219 - 3 18 LINDNER, BUKO. See Ziihringer, Ulrich LIPTAK, ANDRAS, Ninfisi, Pa, and Sztarics-

kai, Ferenc, [Obituary of] Rezsd Bogn- k, 49,3-9

LYON, NANCY B. See Garg, Hari G.

M

MAEDA, KENJI. See Tsuchiya, Tsutomu MORT, ANDREW J. See Knirel, Yuriy A.

N

NANAsI, PAL. See Liptik, Andris NELSON, DAVID A. See Theander, Olof

0

OGAWA, SEIICHIRO. See Suami, Tetsuo

P

PALASINSKI, MIECZYS~AW. See Tomasik,

PERCHERON, FRANCOIS, [Obituary of] Jean Piotr

Emile Courtois, 49, 1 1 - 18

R

RIETSCHEL, ERNST TH. See Ziihringer, Ulrich

S

SCHMIDT, RICHARD R., and KINZY, WILLY, Anomeric-Oxygen Activation for Glycoside Synthesis: The Trichlor- oacetimidate Method, 50,2 1 - 123

SOMSAK, LAsz~6, and FERRIER, ROBERT J., Radical-Mediated Brominations at Ring Positions of Carbohyrdrates, 49, 37-92

SPOHR, U. See Lemieux, R. U. STOSS, PETER, and HEMMER, REINHARD,

SUAMI, TETSUO, and OGAWA, SEIICHIRO, 1,4:3,6-Dianhydrohexitols, 49,93- 173

Chemistry of Carba-Sugars (Pseudo- Sugars) and Their Derivatives, 48,2 1 - 90

SZTARICSKAI, FERENC. See Liptik, Andris.

T

THEANDER, OLOF, and NELSON, DAVID A. Aqueous, High-Temperature Transfor- mation of Carbohydrates Relative to Utilization of Biomass, 46,273 - 326

TOMASIK, PIOTR, PALASINSKI, MIECZY- SLAW, and WIEJAK, STANISLAW, The Thermal Decomposition of Carbohy- drates. Part I. The Decomposition of Mono-, Di-, and Oligosaccharides, 47, 203-278

TOMASIK, PIOTR, WIEJAK, STANISLAW, and PALASINSKI, MIECZYS~AW, The Ther- mal Decomposition of Carbohydrates. Part 11. The Decomposition of Starch,

TSUCHIYA, TSUTOMU, Chemistry and De- 47,219-343

velopments of Fluorinated Carbohy- drates, 48, 91 -271.

HORTON, DEREK, [Obituary of] Hamao Umezawa, 48, 1-20

TVAROSKA, IGOR, and BLEHA, TOMAS, An- omeric and Exo-anomeric Effects in Carbohydrate Chemistry, 47,45- 123

TSUCHIYA, TSUTOMU, MAEDA, KENJI, and

Page 411: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

388 CUMULATIVE AUTHOR INDEX FOR VOLS. 46-50

V

VARELA, OSCAR. See de Lederkremer,

VINOGRADOV, EVGENY V. See Knirel,

VON DEM BRI-H, KARSTEN. See Garg,

Rosa M.

Yuriy A.

Hari G.

W

WEIJAK, STANISLAW. See Tomasik, Piotr

Z

Z~~HRINGER, ULRICH, LINDNER, BUKO, and RIETSCHEL, ERNST TH., Molecular Structure of Lipid A, the Endotoxic Center of Bacterial Lipopolysaccha- rides, 50,2 1 1 - 276

tive Protecting Groups in Carbohydrate Chemistry, 46, I79 -204

ZEHAVI, URI, Applications of Photosensi-

Page 412: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

CUMULATIVE SUBJECT INDEX FOR VOLS. 46-50

A

Aldonolactones, synthetic reactions of, 50,

Anhydrous hydrogen fluoride, application 125-209

of for the structural analysis of polysac- charides, 47, 167 -202

Anomeric and exo-anomeric effects in car- bohydrate chemistry, 47,45 - 123

Anomeric-oxygen activation for glycoside synthesis, trichloroacetimidate method, 50,21- 123

B

Bacterial polysaccharides, components of,

Bognir, RezsB, obituary of, 49, 3 - 9 48,279-318

C

Carba-sugars (pseudo-sugars) and their de- rivatives, chemistry of, 48,21-90

Carbohydrate chemistry anomeric and exo-anomeric effects in, 47,

applications of photosensitive protecting 45- 123

groups in, 46, 179 -204 Carbohydrates

aqueous, high-temperature transforma- tion of relative to utilization of bio- mass, 46,273 - 326

with complexes of metal cations in solu- tion, 47, 1-43

"C-nuclear magnetic resonance-spectral studies of the interactions of metal ions with, 47, 125- 166

high-performance liquid chromatography of, 46, 17-72

addendum to 46,327 - 329

sitions of, 49, 37-92 radical-mediated brominations at ring po-

thermal decomposition of, 47,203-278 '3C-nuclear magnetic resonance-spectral

studies of the interactions of metal ions with carbohydrates, 47, 125- 166

Collagen fibril-associated small proteoglycans of mammalian origin, structure of, 49,

Complexes of metal cations with carbohy-

Courtois, Jean Emile, obituary of, 49, 1 1 - 18 Cyclomalto-oligosaccharides (cyclodextrins),

inclusion complexes of, 46,205 -249

239-261

drates in solution, 47, 1-43

addendum to, 46,333-33s

D

Decomposition of mono-, di-, and oligosac-

Decomposition of starch, 47,279-343 Deulofeu, Venancio, obituary of, 46, 1 1 - 15 1,4:3,6-Dianhydrohexitols, 49, 93- 173 Disaccharides, decomposition of, 47,203 -

charides, 47,203-278

278

E

Enzymic methods in preparative carbohy-

Enzyme specificity, how Emil Fischer was drate chemistry, 49, 175-237

led to the lock and key concept for, 50, 1-20

F

Fluorinated carbohydrates, chemistry and

Fluorinated monosaccharides, N.M.R. spec- developments of, 48,91-277

troscopy of, 46,73 - 177 addendum to, 46,33 1-332

G

Glycopeptides containing glycosyl L-aspara- gine, L-serine, and L-threonine, devel- opments in, 50,277 - 3 10

Glycoside hydrolases, mechanistic informa- tion from studies with reversible and irreversible inhibitors, 48,3 19 - 384

Glycoside synthesis, anomeric-oxygen acti- vation for (trichloroacetimidate method), 50,2 1 - 123

* Starting with Volume 30, a Cumulative Subject Index covering the previous 5 volumes has been published in every 5th volume. That listing the chapters in Volumes 1 -29 maybe found in Volume 29.

389

Page 413: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

390 CUMULATIVE SUBJECT INDEX FOR VOLS. 46-50

H

High-performance liquid chromatography of carbohydrates, 46, 17 - 72

addendum to, 46,327 - 329

Inclusion complexes of cyclomaltc-oligosac- charides (cyclodextrins), 46,205 -249

addendum to, 333-335

L Lipid A, molecular structure of, 50,2 1 1 -276 Liquid chromatography, high-performance,

of carbohydrates, 46, 17-72 addendum to, 46,327- 329

Lock and key concept for enzyme specific- ity, how Emil Fischer was led to, 50, 1 - 20

M

Macrostructure of mucus glycoproteins in

Metal cations, complexes of, with carbohy-

Molecular structure of lipid A, 50,211-276 Monosaccharides, decomposition of, 47,

Mucus glycoproteins, macrostructure of, in

solution, 47,345 - 38 1

drates in solution, 47, 1-43

203-278

solution, 47,345 - 38 1

N

N.M.R. spectroscopy of fluorinated mono- saccharides, 46,73 - 177

addendum to, 46,33 1 - 332

0

Oligosaccharide determinants of glycopro- teins, physicochemical analyses of, 50, 311-350

Oligosaccharides, decomposition of, 47,

Onodera, Konoshin, obituary of, 46, 1 - 9 203-278

P

Photosensitive protecting groups in carbo- hydrate chemistry, applications of, 46, 179 -204

Physicochemical analyses of oligosaccharide determinants of glycoproteins, 50,3 1 1 - 350

Polysaccharides application of anhydrous hydrogen fluo-

ride for the structural analysis of, 47, 167 -202

hydrolysis and other cleavages of glyco- sidic linkages in, 46,25 l -27 l

Preparative carbohydrate chemistry, enzy- mic methods in, 49, 175 - 237

R

Radical-mediated brominations, at ring po- sitions of carbohydrates, 49,37-92

Reducing sugars in solution, composition of, 49, 19-35

S

Starch, decomposition of, 47,279-343 Synthetic reactions of aldonolactones, 50,

125 -209

T

Thermal decomposition of carbohydrates,

Tipson, R. Stuart, obituary of, 50, xiii-xxi Trichloroacetimidate method, anomeric-ox-

ygen activation for glycoside synthesis,

47,203-278

50.2 1 - 123

U

Umezawa, Hamao, obituary of, 48, 1-20

Page 414: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

This Page Intentionally Left Blank

Page 415: Advances in Carbohydrate Chemistry and Biochemistry, Volume 50

I S B N 0-12-007250-5

9 0 0 5 1


Recommended