throbber
COMBINATORIAL
`CHEMISTRY
`
`
`
`Synthesis and Application
`
`Edited by
`
`STEPHEN R. WILSON
`
`New York University
`
`ANTHONY W. CZARNIK
`
`IRORI Quantum Microchcmistry
`
`A Wiley-Interscience Publication
`
`JOHN WILEY & SONS, INC.
`
`New York 0 Chichester
`
`- Weinheim - Brisbane
`
`' Singapore
`
`- Toronto
`
`Page 1 of 27
`
`ILMN EXHIBIT 1037
`
`
`
`Page 1 of 27
`
`ILMN EXHIBIT 1037
`
`

`
`This text is printed on acid-free paper.
`
`Copyright © 199? by John Wiley & Sons, Inc.
`
`All rights reserved. Published simultaneously in Canada.
`
`Reproduction or translation of any part of this work beyond
`that permitted by Section 107 or 108 of the 1976 United
`States Copyright Act without the permission of the copyright
`owner is unlawful. Requests for permission or further
`information should be addressed to the Permissions Department.
`John Wiley 8'. Sons, Inc., 605 Third Avenue, New York, NY
`10l58-0012.
`
`Library of Congress Cataloging in Publication Data
`
`Combinatorial chemistry : synthesis and application I edited by
`Stephen R. Wilson and Anthony W. Czarnik.
`p.
`cm.
`Includes index.
`
`ISBN 0-4?-"l-1263?-X (cloth : alk. paper)
`I, Combinatorial chemistry.
`I. Wilson, Stephen R. (Stephen
`Ross), 1946-
`.
`II. Czarnik, Anthony W.. 1957-
`RS4l9.C666
`I99‘?
`6 I 5' . l9—dc20
`
`96-44718
`
`Printed in the United States of America
`
`I0 9 8 7 6 S 4 3 2
`
`Pae2of27
`
`
`
`Page 2 of 27
`
`

`
`CONTENTS
`
`List of Contributors
`
`Preface
`
`jl Introduction to Combinatorial Libraries: Concepts
`
`and Terms
`
`Stephen R. Wilson
`
`Parallel Organic Synthesis Using Parke-Davis Diversomer
`
`Technology
`Sheila Hobbs DeWi(t and Anthony W. Czarnik
`
`Polymer-Supported Synthesis of Organic Compounds
`and Libraries
`
`Mark J’. Kurt}:
`
`Macro Beads as Microreactors: New Solid—Phase Synthesis
`
`Methodology
`
`Wolfgang E. Rapp
`
`Combinatorial Libraries in Solution: Polyfunctionalized Core
`Molecules
`
`Edward A. Winmer and Juiius Rebek, Jr.
`
`Solid-Phase Methods in Combinatorial Chemistry
`
`Irving Smhoteiki
`
`Page 3 of 27
`
`
`
`Page 3 of 27
`
`

`
`CONTENTS
`
`Radiofrequency Encoding and Additional Techniques for the
`Structure Elucidation of Synthetic Combinatorial Libraries
`
`Xian-yl Xlao and Michael P. Nova
`
`Combinatorial Synthesis Exploiting Multiple-Component
`Condensations, Microchip Encoding, and Resin Capture
`Robert W. Armstrong, S. David Brown. Thomas A. Keating, and
`Paul A. Tempest
`
`Indexed Combinatorial Libraries: Nonoligomeric Chemical
`Diversity for the Discovery of Novel Enzyme Inhibitors
`Michael C Pirrzmg, Joseph H. -L. Chan, and Jrlung Chen
`
`Strategies for Combinatorial Libraries of Oligosaccharides
`
`Carol M. Taylor
`
`Soluble Combinatorial Libraries of Peptides, Peptidomimetics,
`and Organics: Fundamental Tools for Basic Research and Drug
`Discovery
`John M. Ostresh, Barbara Déirner, Sylvie E. Blondelle, and
`Richard A. Houglzlen
`
`Combinatorial Libraries of Peptides, Proteins, and Antibodies
`
`Using Biological Systems
`Stephen Benkovic, Grove P. Miller, Wenyan Zong, and Jefi’ Smiley
`
`Index
`
`Page 4 of 27
`
`
`
`Page 4 of 27
`
`

`
`1 I
`
`NTRODUCTION TO
`COMBINATORIAL LIBRARIES:
`CONCEPTS AND TERMS
`
`STEPHEN R. WILSON
`
`Department of Chemistry, New York University, New York, New York E0003
`
`In the past few years, combinatorial chemistry has become the popular and often
`misunderstood “new wave” in drug discovery. In some cases, combinatorial chemis-
`try is presented as being in direct competition with rational, or computer-aided, drug
`design. Nothing could be further from the truth. Combinatorial chemistry encom-
`passes many strategies and processes for the rapid synthesis of large, organized
`collections of compounds called libraries. When planned intelligently, combina-
`torial methods produce collections of molecularly diverse compounds that can be
`used for rapidly screening for biological activity. Without planning,
`the GIGO
`(garbage in—garbage out) principle applies. Whether or not a library has in some
`way been designed or made more or less at random depends on the reasons for
`preparing the compounds. Combinatorial chemistry as a laboratory practice cannot
`replace computer modeling as an exercise in refining our basic understanding of
`molecular interactions. It is likely that both rational drug design and combinatorial
`chemistry will be used,
`in concert when appropriate, or directly applied to the
`problems best suited to each method.
`Combinatorial chemistry includes many research areas—new analytical meth-
`ods, new computer modeling and database—related challenges, new synthetic ap-
`proaches, new types of reagents, and new types of assays. Although this chapter
`will provide current leads into all these fields, the basic groundwork for combina-
`torial chemistry is still being laid. Many new research areas have yet to be explored.
`Combinatorial chemistry has its conceptual roots in the immune system. In the
`body, when a new antigen comes in contact with the preexisting large collection of
`antibodies, the antibody that binds best is selected and reproduced in large numbers
`
`1
`
`Page 5 of 27
`
`
`
`Page 5 of 27
`
`

`
`INTRODUCTION TO COMBINATORIAL LIBRARIES: CONCEPTS AND TERMS
`
`to effect the immune response. In a similar way, combinatorial chemistry involves
`the synthesis of a large number of compounds. (This collection can be a chemical
`mixture, a physical mixture, or individual pure components.) The collection is then
`tested for biological activity. Finally the active compound is identified and made in
`quantity as a single compound. This type of overall approach is not unlike a
`bioassay guided search for useful natural products.
`Thus, combinatorial chemistry approach has two phases: (1) making a library
`and (2) finding the active compound. Screening mixtures for biological activity has
`been compared to finding a needle in a haystack. This approach may seem unlikely
`to improve the likelihood of finding new drug leads. The fact that it appears to work
`has generated all the excitement.
`This introductory chapter will review several methods for haystack construction
`and needle searching. For example, the best way to prepare a mixture is in some
`type of array or coded form. Then, it is easy to locate and decode the structure of the
`active compound. Combinatorial chemistry has also prompted a resurgence of inter-
`est in rapid synthesis methods, particularly in the area of solid-phase synthesis.
`Many related topics such as automated synthesis, methods for identifying the struc-
`ture of the active compound on a small scale, and handling the massive amounts of
`data sometimes obtained (i.e., computer databases) are also discussed. The main
`portion of the book continues with specific topics written by experts in the field.
`
`1.] HISTORY
`
`The first report describing combinatorial chemistry appeared in 1984 in a study by
`Mario Geyson (1) titled, “Use of Peptide Synthesis to Probe Viral Antigens for
`Epitopes to a Resolution of a Single Amino Acid.” Another early pioneer was Furka
`(2) who introduced the commonly used pool-and-split methods. The 19808 was a
`period of rapid development in solid-phase peptide Syntl.'1csis—-Bruce Merrifield
`won the Nobel prize in chemistry in 1984 for his work on soIid—phase synthesis (3).
`During this time, automated peptide synthesizer technology was in its infancy, and
`the preparation of individual peptides was a challenge. ln 1985 Richard Houghten
`introduced the “tea bag” method for rapid multiple peptide synthesis (4). These and
`other advances in manual multiple-peptide synthesis (5) fed the beginnings of a
`wave of rapid bioassays based on the developing area of molecular biology. Mass
`screening of peptide ligands as a tool for drug discovery allowed the development of
`high-throughput bioassays (6,7).
`Because, historically, biological assays from natural products screening were
`often carried out on mixtures, early pioneers in combinatorial chemistry developed
`the concept of intentionally making mixtures for the purpose of testing. This ap-
`proach had the benefit of more rapid screening. For example, if a mixture showed
`no activity, then all compounds in the mixture could be assumed to be inactive.
`(This assumes that compound A in the mixture does not effect the assay of com-
`pound B or even react with compound B.) Instead of the usual method of individual
`peptide synthesis, it is possible to couple a mixture of all 20 amino acids to another
`
`Pae6of27
`
`
`
`Page 6 of 27
`
`

`
`HISTORY
`
`mixture of 20 amino acids to produce 400 dipeptides. If these newly formed 400
`dipeptides are reacted with a mixture of 20 additional amino acids, 8,000 tripeptides
`are obtained. This process describes the original formulation of combinatorial
`chemistry. The mixture of 8,000 tripeptides would be called a combinatorial library.
`The combinatorial possibilities for the number of compounds is seen to increase
`exponentially as shown in Figure 1.1, that is, N reactions can produce N X N
`compounds.
`Although it is easy to see how the rapid production of many compounds in a
`mixture works, it is not so easy to see how to make this process work in drug
`discovery. There are several problems. For example, in the process shown in Fig-
`ure 1.l, what would happen i@ne or more of the amino acids does not react or
`reacts sluggishiy to give a low yield? The mixture would then not contain 8,000
`tripeptides but a lower number. The peptides
`uld not be present in the idealized
`statistical distribution (i.e., equimolar ratios
`dditionally, how does one find a
`single active peptide in a mixture with 7,999 inactive ones?
`The first problem is always a sticky one and will be discussed in more detail
`later. The second problem has the following solution, called. Imag-
`ine constructing the library not with all 20 amino acids but starting with only 19
`amino acids, leaving out one specific (known) residue. After subsequent coupling
`with 20 times 20 amino acids,
`this leads to a library of_ only 7,600 tripeptide
`compounds. If this library is now inactive, we have learned that position I of the
`
`Number at different peptides increases
`exponentially with length of molecule‘
`
`Number of
`amino acid
`residues
`
`Peptide
`
`.
`
`Number of
`distinct peptides
`
`NH2—x,x2-cooH
`
`I
`
`NH2-X1X2X3-CDOH
`
`NH2-X1X2X3X4-COOH
`
`NH2-X1X2X3X4X5-C00H
`
`400
`
`3,000
`
`160,000
`
`3,200,000
`
`NH2-X1X2X3X4X5)<fi-COOH
`
`64,000,000
`
`NH2-X1X2X3)(4X5X5X7-COOH
`
`1,230,000,000
`
`NH2-X1X2X3X4X5X5X7Xg-CODH
`
`25,600,000,000
`
`Xn represents individual amino acids residues.
`Numbers of distinct peptides are based on
`20 residues at each position
`
`Figure 1.1 Combinatorial peptide chemistry. The number of different peptides increases
`exponentially with length.
`
`Page 7 of 27
`
`
`
`Page 7 of 27
`
`

`
`4
`
`INTRODUCTION TO COMBINATORIAI. LIBRARIES: CONCEPTS AND TERMS
`
`..._..--.__
`
`‘T’
`
`g‘-,1/ULi|¢I..
`
`tripeptide has the residue that was omitted from the synthesis. If you continue this
`process, methodically omitting one amino acid after another, a single combination
`still has the active compound. Thus a maximum of 20 experiments are needed to
`define position 1. Now, with the active first residue fixed, 20 more experiments are
`carried out to define the best residue in position 2. There are 20 + 20 + 20 = 60
`omission experiments required to define the complete tripeptide sequence of the
`active component in 8,000 compounds. This mathematical principle is the parallel-
`ism advantage: 20 + 20 + 20 = 60 (linear) versus 20 X 20 X 20 = 8000
`(exponential) that underlies the principle of combinatorial chemistry (8,9). This
`iterative synthesis and screening strategy has been discussed in detail by Dooley and
`Houghten (8). A careful analysis of methods for deconvolution of libraries and their
`intrinsic problems is described by Freier and co-workers (10).
`As you can see from this example, this strategy for combinatorial chemistry
`relies on two important points. First, the synthesis of the mixture must be fast and
`efficient because you need to prepare many variations of the libraries. Second, the
`testing must also be fast and easy, because you need to rapidly test many compound
`mixtures to find the active sequences. Because it is not always the case that both
`synthesis and testing are fast and efficient, these two key points return again and
`again. In addition, complications involving multiple active components (more than
`one needle in each haystack) often confuse the issue. On the other hand, the classic
`combinatorial situation in Figure 1.1 is a basic method with which one can Contrast
`other approaches.
`Fast and easy synthesis and testing suggests automation and robotics. Over the
`past few years, combinatorial chemistry has grown so quickly, in part because good
`automation exists in both areas. Section 1.7 on robotic instrumentation will review
`
`some advances in robotics that have enabled fast and easy preparation of com-
`pounds. A discussion of the automation of bioassays,
`that is, high-throughput
`screening,
`is beyond the scope of this book, and readers are referred to other
`reviews of advances in assay processes (6).
`
`1.2 REVIEW OF THE LITERATURE
`
`Combinatorial chemistry is an approach to synthetically produce molecular diver-
`sity. Traditionally, molecular diversity was primarily obtained from natural prod-
`ucts. Two early reviews contrast natural products diversity (11) with molecular
`diversity obtained from peptide combinatorial chemistry (12). Two other early re-
`views in 1993 on applications of combinatorial chemistry barely mention organic
`chemistry (13,14). It was not until 1994 that new comprehensive literature reviews
`of combinatorial chemistry were published covering both peptide combinatorial
`methods (15) and organic chemical applications (16). Since that time many other
`reviews have appeared directly addressing organic combinatorial chemistry (17-
`26). A comprehensive bibliography of combinatorial chemistry references is avail-
`able on the World Wide Web (http:Nvesta.pd.com) (27).
`
`Page 8 of 27
`
`
`
`Page 8 of 27
`
`

`
`SOLID-PHASE ORGANIC SYNTHESIS
`
`1.3 SOLID-PHASE ORGANIC SYNTHESIS
`
`Until the revolution in high-speed bioassays, the testing of new compounds was the
`rate-limiting step in the drug discovery process. While it might take a chemist one
`or two weeks to synthesize a single compound, it required a much longer time to
`carry out the biological assays. Now modern high-speed assays using robotic sarn-
`plers can screen more than 10,000 compounds per week. The biological assay has
`evolved from the rate-limiting step to the driving force in the need for large numbers
`of compounds, and thus the driving force of combinatorial chemistry. In some
`cases, where compounds are prepared attached to polymer supports, bioassays are
`still not very well adapted.
`In the area of high-speed automated synthesis, peptide chemistry has already
`arrived. In 1963, Merrifield reported the first examples of solid-phase synthesis of
`peptides using chloromethylated-polystyrene containing immobilized N-protected
`amino acid building blocks (3). This chemistry developed over the ensuing decade
`and became the basis for much of the progress in peptide chemistry.
`Figure 1.2 shows the Merritield approach. An insoluble polymer bead contain-
`ing—CH2Cl groups is prepared by chloromethylation of the copolymer of styrene
`and p-divinylbenzene. These chloromethyl groups can be esterified with N-pro-
`tected amino acid building blocks. The amino group is iteratively deprotected and
`coupled with new N-protected amino acids to build the growing chain. Merrifield
`used the rerr-butyloxycarbonyl protecting group (BOC group) to protect the free
`amino terminus during the coupling step. This allows depmtection with acid
`(CF3COOH) before coupling with the next N-protected residue. The cycle can be
`continued until the desired sequence is obtained. By-products and excess reagents
`are not bound by the resin and can be removed from the resin beads during washing
`cycles.
`An entire industry has developed to serve the peptide synthesis field. There are
`many instrument companies that provide automated peptide synthesizers, as well as
`other companies that provide peptide building blocks, reagents, and supplies (17).
`Solid-phase synthesis techniques do not, per se, mean fast synthesis. On the other
`hand, there are many years of experience in automating bead—based synthetic ap-
`proaches because it is easy to extend the principles to organic applications.
`Although peptide chemistry advanced using solid-phase techniques, applications
`of polystyrene bead-supported approaches to traditional organic molecules did not
`go far in the earlier days. Some work appeared in the 1970s, but an early review by
`Rapoport had a negative tone (28). Although later successes by Leznolf (29),
`Neckers (30), Frechet (31), and others clearly showed that polystyrene bead-
`supported organic chemistry worked quite well, interest in the field largely disap-
`peared (32). This might have been due to several factors. First, solid-phase chemis-
`try often produced only small quantities of compound. Pre-1980 nuclear magnetic
`resonance (NMR) instrumentation required much larger amounts of compound for
`characterization than is needed with today's high—field Fourier transform (FT) ma-
`chines. Second, the high-speed testing of small amounts of compound that launched
`
`Page 9 of 27
`
`
`
`Page 9 of 27
`
`

`
`-»
`
`INTRODUCTION TO COMBINATORIAL LIBRARIES: CONCEPTS AND TERMS
`
`I
`
`R1
`
`R‘
`
`O
`
`3oc\
`
`N
`H
`T--pr
`
`GOOH
`

`
`NHBOC
`CF-_-,COOH
`----——I-
`
`O
`
`*3
`
`mi:
`
`rmeoe
`
`R:
`
`CF3COOH
`T».
`
`D
`
`O
`
`R
`
`1
`
`N
`H
`
`0
`
`R2
`
`""2
`
`P
`
`R,
`
`O
`
`°\")\N
`
`H
`
`‘3
`
`P
`
`Figure 1.2 Merrifield solid-phase peptide synthesis (3).
`
`the field of combinatorial chemistry had not emerged. The late 1970s and 19803 was
`a period of tremendous growth in the development of selective reagents and meth-
`ods for solution-phase organic synthesis. There were plenty of new targets and new
`synthesis methodology to occupy the attention of organic chemists. Thus, while
`solid-phase peptide methods rapidly matured and led to instrumentation for automa-
`tion of chemistry, solid-phase methods for organic synthesis all but faded away.
`Until 1992, the combinatorial library field was exclusively the domain of peptide
`and oligonucleotide-based chemistry.
`The situation changed in 1992 with the report by Bunin and Ellman of the
`preparation of combinatorial libraries of organic molecules (33). The report of solid-
`phase synthesis of Diversomers by the Parke-Davis group (34) shortly thereafter
`stimulated considerable interest in reexamination of solid-phase organic synthesis.
`Within the past few years, a number of reports have appeared that point to great
`potential for solid-phase organic synthesis. An excellent recent review of the litera-
`ture of solid-phase organic synthesis has recently been published (35).
`Bunin and Ellman’s (33) solid-phase synthesis of 1,4-benzodiazopines is an
`excellent illustration of the process of combinatorial library production using organ-
`ic templates (Fig. 1.3). The chemistry is very similar to the Parke—Davis Diversomer
`
`Page 10 of 27
`
`
`
`Page 10 of 27
`
`

`
`SOLID-PHASE ORGANIC SYNTHESIS
`
`Figure 1.3 Ellman solid-phase l,4-benzodiazcpine synthesis.
`
`approach (34) detailed in Chapter 2, except that Bunin and Ellman’s compounds
`were not cleaved from the solid support. 1,4-Benzodiazepines are constructed from
`three components: 2-aminobenzophenones, N-protected amino acids, and alkylat-
`ing agents. As one can see in Figure 1.3, the preparation of a combinatorial library
`on solid—phase synthesis beads requires several things. First, a selection of building
`blocks are needed with R1, R2, R3, and R4 that can be permuted. Second, a reaction
`scheme that is suitable for interative conversion, in a similar manner to peptide
`synthesis, is required. Third, a method for linking one of the building blocks to a
`solid support and conditions for cleaving the compound(s) off the support at the end
`of the cycle must be found.
`The variation of groups Ry-R4 give a matrix of different structures (Fig. 1.4).
`These compounds are the combinatorial library and the chemistry is sometimes
`called matrix chemistry. Besides chemistry issues, keeping track of a large collec-
`tion of structures is often difficult. Section 1.8 discusses some of these points as
`well as software solutions that attempt to deal with multidimensional collections of
`pharmacophores.
`From only a few studies in 1992, the number of publications reporting applica-
`tions of the use of solid-phase organic chemistry has increased dramatically. So
`many new applications of organic reactions on a solid support have been reported
`that only a partial list of the types of reactions carried out on a solid support can be
`provided here (Table 1.1).
`Besides the reaction itself, so1id—phase synthesis methods also require consider-
`ation of several new technical issues. Table 1.2 reports some characteristics of the
`
`Page 11 of 27
`
`
`
`Page 11 of 27
`
`

`
`INTRODUCTION TO COMBINATORIAL LIBRARIES: CONCEPTS AND TERMS
`
`Template
`
`R,
`
`OH
`
`OH
`
`OH
`
`OH
`
`OH
`
`OH
`
`R,
`
`R,
`
`Cl
`
`CH,
`
`CH, CH,
`
`CH, CH, CH,
`
`CH,
`
`CH,
`
`CH, CH,
`
`C]
`
`C]
`
`CH, CH,
`
`CH, CH,
`
`CH, CH,
`
`CH, CH, CH,
`
`Figure 1.4 Typical molecular spread sheet of benzodiazepine derivatives.
`
`most common form of resin bead: crosslinked polystyrene. In planning a synthesis,
`one must consider loading, the level of substitution on the beads and swelling
`characteristics of the resin. As can be seen in Table 1.2, crosslinked polystyrene
`beads swell quite a bit in volume with different solvents. The physical size of the
`beads used in a solid-phase synthesis, of course, also effects the weight of each bead
`
`TABLE 1.1 Organic Reactions on Polystyrene
`Solid Supports (35)
`
`Reaction
`
`Reference
`
`Dieckman cyclization
`Diels-Alder reaction
`Micheal reaction
`Aldol condensation
`
`Mitsunobu reaction
`
`Pausen-Khand Cycloaddilion
`Organometallic additions
`Stille reaction
`
`Suzuki reaction
`Heck reaction
`
`Suzuki coupling
`Urea synthesis
`Thiazoline synthesis
`Solid-phase Steroid synthesis
`Wittig reaction
`
`28
`36
`37
`38
`
`39
`
`40
`41
`42
`
`43
`44 and 45
`
`4-6
`47
`48
`49
`37
`
`Pae12of27
`
`.._ -
`
`
`
`Page 12 of 27
`
`

`
`SOLID—PHASE ORGANIC SYNTHESIS
`
`TABLE 1.2 Data for Polystyrene-Bead Solid Support (50, 51)
`
`Styrenewdivinylbenzene copolymer:
`
`l—2% crosslinked; 1 g of 200-400 mesh resin has ~4-
`IO X 105 beads
`
`l05—l30°C
`Thermal stability:
`Bead sizes: 100 mesh (212 um), 140 mesh (150 pm), 200 mesh (106 um), 325 mesh
`(75 pm), 400 mesh (45 um)
`Swelling: DMF (3.5 mLfg), TI-IF (5.1 mLfg), CH30H (1.5 mug), H20 (1.5 n1LIg),
`CI-IECI2 (5.3 mug), CHCI3 (5.9 mI..;’g)
`Loading: 0.1-0.4 rnmollg resin, ~I00—400 pmol per head
`
`and therefore the amount of product on each head. Although crosslinl-ted poly-
`styrene resin beads (Merrifield-type resin) are the most common for historical
`reasons, a popular new material called TentaGel is now coming into wide use (52).
`This new material contains a polystyrene core with polyethylene glycol spacer arms
`(PS—PEG). TentaGel (although physiomechanically less robust than polystyrene)
`has very desirable characteristics for synthesis because the attached reacting groups
`project out in solution rather than being anchored close to the polymer back-
`bone. This provides for better reactivity and for reactivity more closely paralleling
`
`R1"CH3,R;=H
`K1=t-Bu,R2=H
`R]=Ph, R1'='H
`R1=CH3»R2=CH3
`R1"t-Bu.R2='CH3
`R1=Fh. Rz"CH3
`R1"'CH3,R1“"Cl
`R]‘='I,'B|),R2=Cl
`R]‘=Pl'l, Rz=‘CI
`
`Figure 1.5 Solid-phase synthesis of small molecule combinatorial libraries (37).
`
`Page 13 of 27
`
`
`
`Page 13 of 27
`
`

`
`I0
`
`INTRODUCFION TO COMBINATORIAL LIBRARIES: CONCEPTS AND TERMS
`
`solution-phase chemistry. Applications of TentaGeI in combinatorial chemistry is
`the subject of Chapter 4.
`A good example to illustrate the use of well-known organic reactions is Chen and
`co—workers’ report of the preparation of a small collection of compounds from a trityl
`resin-linked alcohol (Fig. 1.5) (37). The diol was linked to trityl polystyrene and then
`oxidized with S03!pyridine to the aldehyde. The resulting polystyrene-linked al-
`dehyde could be reacted with three different Homer-Emmons reagents (R, = Me,
`I-Bu, and Ph). Using the split-mix method, these enones were combined in equal
`amounts, split into three separate reaction vessels, and reacted in a Michael addition
`reaction with three different R2-SH compounds. Nine different compounds were
`formed, a very small but prototypical combinatorial library. After product cleavage,
`each of the nine compounds could be isolated and completely characterized. In
`addition, the use of Fourier transform—infrared (FT-LR) and gas chromatography-
`mass spectroscopy (GC—MS) for analysis of products attached to beads was demon-
`strated. This work and its extensions are more fully described in Chapter 3.
`
`1.4 ANALYTICAL TECHNIQUES FOR SOLID-PHASE SYNTHESIS
`
`The principle of solid-phase synthesis requires that the product remain attached to
`the resin head. A disadvantage of the technique is that it is more difficult to
`characterize the product at intermediate stages of the synthesis. Methods used to
`identify reaction products from solid-phase synthesis typically involve elemental
`analysis, titration of reactive groups, or simply weight gain (35). Sometimes, a
`small amount of the intermediate resin beads are removed, and the attached product
`cleaved and analyzed in the usual manner. For product identification, organic chem-
`ists usually use spectroscopic methods such as IR, NMR, or MS. Scheme 3. i5 in
`Chapter 3 shows the FT-IR of starting material and product in a typical solid-phase
`reaction;
`the conversion to product can be clearly observed by changes in the
`spectrum.
`
`is NMR. Unfortunately,
`The mainstay of organic structural analysis, however,
`the heterogeneous environment of resin—attached compounds leads to very broad
`‘H-NMR lines. Recent work suggests that the use of magic—angle spinning ‘H-NMR
`(53,134) and '3C—NMR (34) with polystyrene resin-bound compounds may be use-
`ful. The use of ‘3C-NMR with synthetic compounds bound to TentaGel resin beads
`(PS—PEG) was shown to give excellent line widths for compounds where no signal
`could be observed for the same compounds directly attached to polystyrene (55).
`The use of 13C-enriched building blocks also provided high-quality spectra with less
`interference from the polymer and solvent.
`Finally, the use of mass spectroscopy for evaluating solid-phase synthesis has
`been explored. Methods usually require cleavage of the product from the resin and
`analysis of the resulting microsamplc in the usual manner. Several examples have
`been reported involving direct GC—MS of the product from a single head. A single
`100-200 mesh polystyrene “Merrifield” bead weighs approximately 0.1-0.2 p.g.
`At a usual loading, each head contains r-400 pmol of compound, more than enough
`for a mass spectrum. Figure 3.20 in Chapter 3 shows a typical single-bead result.
`
`a
`
`, Pae
`
`
`
`Page 14 of 27
`
`

`
`ENCODING OF BEADS
`
`11
`
`Several reports have also appeared using direct matrix-assisted laser desorption
`(MALDI) of combinatorial library beads (56-58). The most elegant approach to
`mass spectral analysis of synthesis beads was the report of the molecular weight
`imaging of individual beads using time-of-flight secondary ion mass spectrometry.
`The structure of small peptides could be seen directly from individual 30-60 um
`polystyrene beads after cleaving the resin—peptide link with trifluoroacetic acid
`(TFA) (59).
`The combination of afiinity selection for active receptor ligands with electro-
`spray mass spectroscopy shows promise for the rapid identification of active com-
`pounds in a combinatorial library (Fig. 1.6) (60). A small molecule combinatorial
`library containing 600 compounds was incubated with purified receptor and then
`separated by size-exclusion chromatography. The receptor containing bound ligand
`was collected, and the ligand was released and analyzed by electrospray mass
`spectroscopy. Figure 1.6 shows the low-molecular-weight (MW) region of the spec-
`trum reveals a tight-binding ligand from the library at MW = 708. Specific binding
`of this compound can be determined by evaluating its competition with a known
`receptor ligand. It is likely that further developments of this “affinity selection”
`technique could speed up isolation and identification of leads from combinatorial
`libraries.
`
`1.5 ENCODING OF BEADS
`
`The combinatorial method for producing mixtures of thousands or millions of
`compounds has always presented an awkward deconvolution process. In principle, a
`better strategy would be to prepare the compounds in pure form, in separate, labeled
`bottles. What if each compound was attached to a separate bead—-one compound
`per head (61)? The mixture of beads then represents a physically separable collec-
`tion of compounds. This approach has been widely used and only requires a means
`to determine what is attached to a single bead. Instead of direct structure determina-
`tion of the compound attached to a single head, the synthesis history of the head is
`recorded on the bead—-encoding. Each head can be derivatized with a “tag” that
`contains information on the structure attached to the bead. Therefore, instead of
`identifying the compound, one identifies the code. This concept was first described
`for beads encoded with a genetic DNA tag (62). This elegant approach allows
`amplification of the encoded “signal” by polymerase chain reaction (PCR) for
`analysis.
`A more practical approach for organic chemistry is the tagging strategy devel-
`oped by Ohlmeyer and co-workers (63). The use of the split-synthesis method
`provides a final library wherein each head has a single compound attached. This
`method (Fig. 1.7) involves attachment (at the level of a few percent of the real
`compound) of a binary code of chlorophenyl groups at each synthesis step. Using
`electron-capture GC analysis, 0.1 pmol of code attached to a single 50-80 pm head
`is more than enough for detection (64). The presence or absence of one of the four
`GC separable chlorophenyl derivatives (Fig. 1.7, n = l—4) gives a binary code of 0
`or 1. Thus the binary codes such as 0000, 0001, 0010, 0100, and so forth can be
`
`Page 15 of 27
`
`
`
`Page 15 of 27
`
`

`
`won3
`
`____.._._._...._____.____..___.__._.____O
`
`mdmm
`
`
`
`
`
`
`
`....dam:EE.mamm....._..........mwa......nmmvN53.33"an
`
`
`
`
`
`
`
`Tan.TamNmmm.1
`
`...m.mENd?Nmmmcm"1.
`nmnm3%_mumGa2.9:1
`
`mmq
`
`rr
`an
`
`(N
`IN
`
`r-4
`-4
`
`O
`
`‘*5!Ln
`N
`
`“E0'‘!
`.—I
`
`(9%) "WI '|3H
`
`(96) ‘W1 '|9H
`
`
`
`Page 16 of 27
`
`
`

`
`ENCODING OF BEADS
`
`polymer
`
`eleavable linker
`
`Figure 1.7 Still bead encoding method from combinatorial libraries (41). The synthesis
`polymer is derivitized with a photocleavablc linker and a tag containing a choloroaryl group.
`The tags can be removed from the beads, the resulting alcohols silylatcd and then detected
`by GC.
`
`used to define the synthetic steps applied to the head. The compound synthesis
`proceeds on the beads as usual. After each step a coding reaction is carried out on
`the bead at a trace level (~1%). Usually the code is linked using a photocleavable
`linker so that the code can be removed without damaging the valuable (perhaps
`active) compound attached to that bead. Details of the use of an encoded library to
`locate ligands for a synthetic (unnatural) receptor molecule has been reported (65).
`Further development and commercialization of this coding method is now in the
`hands of Pharmacopoeia (66) and a review of the Still coding method has just
`appeared (67).
`Another coding method uses peptide sequences for a reaction code (68). Two
`orthogonally protected points of extension allow the primary peptide (or organic)
`molecule to be formed at one point and a coding sequence built at the second. The
`code can be “read" by selective cleavage of the coding peptide, followed by se-
`quencing. The coding sequence was claimed not to effect binding of the primary
`structure .
`
`(a) Electrospray MS of a 600—compound small molecule library. (b) Library
`Figure 1.6
`from (a) after affinity selection with an excess of receptor. Note the strong enrichment and
`selection of the compound at MW == 708 (MI-l+). (c) Mixture after afiinity selection in the
`presence of a strong competitive ligand. This figure shows that the compound at 708 MH+ {Z
`can be competed off the receptor by this peptide ligand.
`
`Page 17 of 27
`
`
`
`Page 17 of 27
`
`

`
`14
`
`INTRODUCTION TO COMBINATORIAL LIBRARIES: CONCEPTS AND TERMS
`
`Finally, an intriguing new coding method has just appeared. TentaGel polymer
`beads containing a memory storage device are used in synthesis. The synthesis code
`can be written into the memory chip with radiofrequency pulses (69). The memory
`chips are similar to EPROM chips (erasable, programmable, read-only memory.) A
`more comprehensive discussion of this technology is covered in Chapters 7 and 8.
`
`1.6 POSITIONALLY ADDRESSABLE SPATIAL ARRAYS
`
`Another approach to separationfidentification of combinatorial library collections is
`based on preparing each compoun

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket