throbber
ANNUAL
`REPORTS IN
`MEDICINAL
`CHEMISTRY
`Volume 29
`
`Sponsored by the Division of Medicinal Chemistry
`of the American Chemical Society
`
`EDITOR-IN-CHIEF
`JAMES A. BRISTOL
`PARKE-DAVIS PHARMACEUTICAL RESEARCH
`DIVISION OF WARNER-LAMBERT COMPANY
`ANN ARBOR, MICHIGAN
`
`SECTION EDITORS
`WILLIAM K HAGMANN • JOHN C. LEE • JOHN M. MCCALL
`JACOB J. PLATTNER • DAVID W. ROBERTSON • MICHAEL C. VENUTI
`
`EDITORIAL ASSISTANT
`LISA GREGORY
`
`(k
`
`ACADEMIC PRESS, INC.
`San Diego New York Boston London Sydney Tokyo Toronto
`
`eonard Presta, Ph.Dj
`/1/2O18
`
`j
`
`Repored iy Chrote Lacey
`APR, GSA S14224
`
`PFIZER and SAMSUNG v. GENENTECH
`IPR2017-01488
`PFIZER EX. 1196, Page 1
`
`

`

`Academic Press Rapid Manuscript Reproduction
`
`This book is printed on acid-free paper.
`
`Copyright © 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 informat4on
`storage and retrieval system, without permission in writing from the publisher.
`
`Academic Press, Inc.
`A Division of Harcourt Brace & Company
`525 B Street, Suite 1900, San Diego, California 9210J-4495
`
`United Kingdom Edition published h
`Academic Press Limited
`24-28 Oval Road, London NW 7DX
`
`International Standard Serial Number: 0065-7743
`
`International Standard Book Number: 0-12-040529-6
`
`PRINTED IN THE UNITED STATES OF AMERICA
`96 97 98 99 Q 987654321
`94
`95
`
`PFIZER and SAMSUNG v. GENENTECH
`IPR2017-01488
`PFIZER EX. 1196, Page 2
`
`

`

`Chapter 32. Humanized Monoclonal Antibodies
`
`Leonard Presta
`Genentech, Inc., 460 Point San Bruno Blvd.
`South San Francisco, CA 94080
`
`-
`
`Introduction - Almost two decades ago, the development of monoclonal hybridoma
`technology -
`the ability to make cultured cells produce antibodies of predefined specificity
`promised a new weapon in the arsenal of molecules able to combat disease (1).
`Antibodies against a specific antigen (or target molecule) could be generated, incorporated
`into a hybridoma for production, and then used in diagnosis or therapy. However, only
`rodent monoclonal antibodies could be made due to technological limitations. In most
`clinical applications, these rodent monoclonal antibodies exhibit properties which severly
`limit their utility. First, they may induce an immunogenic response in humans, referred to as
`HAMA (human anti-mouse antibodies), when the human immune system recognizes them as
`foreign substances. Second, the therapeutic efficacy of the rodent antibody may also be
`reduced because a rodent antibody is cleared from serum more rapidly than human ones.
`Third, in humans, rodent antibodies generally exhibit only weak recruitment of effector
`Junctions, such as antibody-dependent cell-mediated cytotoxicity and complement fixation,
`which may be requisite for the function of the antibody. Use of a human antibody would
`circumvent these three problems.
`
`With advances in molecular biology and mammalian tissue culture, it became
`possible to obtain useful amounts of any antibody, thereby surmounting the limitations of
`hybridoma technology and use of only rodent antibodies. But one problem still existed --
`how does one obtain human antibodies against a particular human antigen? Even if one
`could ethically use a human subject as the biological factory, the human immune system, in
`general, does not produce antibodies against human proteins. Two techiques have been
`developed to address this problem. Earliest of these was the construction of chimeric
`antibodies in which entire antigen-binding domains from a rodent antibody are substituted
`for those of a human antibody (Fig. 1) (2). In many cases, these molecules still exhibited
`some, albeit reduced, HAMA. The next step was generation of humanized antibodies in
`which a significantly reduced number of rodent residues are incorporated into a human
`antibody such that the humanized antibody has the same binding characteristics and
`specificity as the original rodent antibody (3-5). In order to understand the technique of
`humanization and the difference between a chimeric and humanized antibody, the structure
`of antibodies must be appreciated.
`
`two identical light
`Antibody Structure - An antibody consists of four peptide chains --
`which form a 'V' shape. The functions of the
`chains and two identical heavy chains -
`antibody reside in different domains. Antigen binding occurs at the ends of the arms of the
`'V', each arm being referred to as a Fab or antigen binding fragment. Hence each antibody
`can bind two antigen molecules. The effector functions reside in the base of the 'Y',
`referred to as the Fc portion. Each light chain consists of one variable domain and one
`constant domain; each heavy chain consists of one variable domain and three constant
`domains (Fig. 1). The constant' notation refers to the fact that for a particular species and
`immunog!obulin class (e.g. lgG, IgE, lgA) the amino acid sequence for the constant domain
`
`ANNUAL REPORTS IN MEDICINAL CHEMISTRY-29
`
`M
`
`Copyright (D 1994 by Academic Press, Inc.
`All rghLs of reproduction In any form reserved.
`
`PFIZER and SAMSUNG v. GENENTECH
`IPR2017-01488
`PFIZER EX. 1196, Page 3
`
`

`

`Z51 8
`
`Section Vi-Topos ua Drug Design and Discovery
`
`Venuti, Ed.
`
`is, for our purposes, invariant. The 'variable' notation refers to the fact that certain portions
`of variable domains differ extensively in sequence among antibodies and are used in the
`binding and specificity of each particular antibody for its particular antigen. These portions
`are referred to as complementarity-determining regions (CDRs). Both light and heavy chain
`variable domains contain three CDRs to give a total of six CDRs involved in binding antigen.
`Each CDR is a contiguous sequence of amino acids which form a loop connecting two 3-
`strands of the framework. Note though that the amino acid sequence of the non-CDR
`portion of the 'variable' domain is relatively invariant and is used to categorize these
`domains into subgroups (6).
`
`o
`
`Mouse
`
`Chimera
`
`\,
`
`U
`Humanized
`Human
`Figure 1. Chimeric and Humanized Antibodies. CDRs are represented by 'fingers'
`on variable domains of the light chain (VO and heavy chain (VH). Note that only the mouse,
`chimera and humanized antibodies bind antigen. Disulfide bonds between the two heavy
`chains and between the light and heavy chains are denoted by thicker, dark lines.
`
`In one type of chimeric antibody all of the variable domains from a rodent antibody
`are fused onto the constant domains of a human antibody (Fig. 1). This type of chimeric
`antibody is comprised of four rodent domains and eight human domains and consequently
`retains approximately 33% rodent residues. In the corresponding humanized antibody, only
`the six rodent CDRs replace the six human CDRs (Fig. 1). The resulting antibody contains
`only about 5-10% rodent residues and is still chimeric in that it has residues derived from
`different species. From the perspective of the human immune system, the humanized
`antibody is the more human of the two types as it contains fewer rodent residues.
`Moreover one must appreciate that the amino acids in CDRs are highly variable and this
`variance is independent of species. The nature of the CDRs is dependent upon the target,
`whereas the framework is species dependent. Hence even though the humanized antibody
`still retains about 5-10% rodent residues, these residues would also be variant in human as
`
`PFIZER and SAMSUNG v. GENENTECH
`IPR2017-01488
`PFIZER EX. 1196, Page 4
`
`

`

`Chap. 32
`
`Humanized Monoclonal Antibodies
`
`Presta
`
`319
`
`well as rodent and thus the humanized antibody could well be very similar to a human
`antibody directed against the same site on the same target as the original rodent antibody.
`
`Humanized Antibody Construction - Al first glance constructing a humanized antibody is
`rather simple and straightforward. One chooses a human antibody, clips off the six human
`CDRs and replaces them with their structurally analogous rodent CORs. Indeed some
`humanized antibodies have approached this simplicity in method (7,8). However, there are
`both theoretical and technical difficulties. Among the former is the exact definition of a
`CDR, i.e. exactly which residues comprise the CDRs and which comprise the framework.
`Two definitions have been proposed. The first relies solely on analyses of the amino acid
`sequences of a multitude of antibodies (6). The variance of amino acid types at each
`residue position of antibodies was evaluated and those positions which exhibited a
`relatively large variance were included in a CDR as long as they were contiguous to other
`positions with high variance. Residue positions showing relatively low variance were termed
`framework. The second definition was derived from structures of antibodies (9). When the
`two definitions are compared some striking differences are apparent. While the sequence-
`based CDRs are generally larger than, and encompass, the structure-based CDRs, a
`notable discrepancy occurs in the definition of CDR-H1 (i.e. the first CDR loop in the heavy
`chain sequence) where the two definitions overlap by only two residues. Generally, the
`sequence-based definition of CDRs has been used when an antibody is humanized.
`
`The technical difficulties fall into three categories. First, the choice of constant
`domains must be made. To date almost all antibodies humanized have been those of the
`lgG class. In humans there are four recognized subclasses, denoted lgG 1, lgG2, lgG3 and
`lgG4, the classification of which is based on the sequence of their constant domains. Each
`subclass also exhibits differences in effector, or biological, functions mediated by the
`constant domains. The intended therapeutic use of the antibody must be considered when
`choosing the lgG subclass, e.g. is complement fixation an advantage or disadvantage for
`the particular application?
`
`Second, the choice of human variable domains, both light and heavy, must be
`made. In the homology or 'best-fit' method, the sequence of the rodent variable domain is
`screened against the entire library of known human variable domain sequences. The human
`sequence which is closest to that of the rodent is then accepted as the human framework
`(8,10). Another method uses a particular framework derived from the consensus sequence
`of all human antibodies of a particular subgroup of light or heavy chains. The same
`framework may be used for several different humanized antibodies (11,12). Remembering
`that the purpose of humanization is to trick the human immune system into recognizing the
`humanized antibody as one of its own, is the 'best-fit' or the consensus framework better for
`this trickery? Unfortunately there are very limited in vivo data on the reaction of the human
`immune system to humanized antibodies and no comparative study has been performed on
`the two types. In the final analysis both may function well with regard to acceptance by the
`human immune system, with perhaps an occasional aberration.
`
`designing the humanized
`Third is the most imposing technical difficulty --
`antibody such that it has the same binding affinity and specificity as the original rodent
`antibody. When the rodent CDRs are simply grafted onto a human framework the resultant
`humanized antibody may exhibit signficiantly reduced binding compared to the original
`rodent antibody (11,12). This reduction in binding may be caused by certain buried
`framework residues. Each CDR is a loop and these loops may be anchored to the
`framework not only covalently at their ends but through noncovalent interactions with the
`sidechains of buried framework residues. For example, in CDR-L1 the sidechain at position
`29 (residue numbering is according to ref. 6) is usually hydrophobic, buried and contacts
`hydrophobic sidechains at framework positions 2, 25, 33, 71 and 90 (13). If an incorrect
`amino acid is chosen for any of these framework positions the packing of CDR-L1 against
`the protein might be altered, resulting in an incorrect presentation of the exposed CDR-L1
`
`PFIZER and SAMSUNG v. GENENTECH
`IPR2017-01488
`PFIZER EX. 1196, Page 5
`
`

`

`Section VI—Topics in Drug Design and Discovery
`
`Venuti, E4.
`
`sidechains and consequent reduction in binding to antigen. Such a case may occur when
`the sidechains at position 29 in the rodent CDR and positions 2, 25, 33, 71 and 90 in the
`rodent framework differ from those in the human antibody. In order to retain proper binding
`the offending framework residues must be altered from human to murine so that the CDR
`packs against the protein properly. Alternatively, the buried sidechain in the CDR itself,
`such as at position 29 in CDR-L1, might be changed from murine to human to evaluate
`whether proper CDR conformation can be attained. This regimen must be evaluated for
`each of the six CDRs because if just one CDR has an improper conformation it may reduce
`the binding of the humanized antibody to the point that it is useless as a therapeutic
`replacement for the rodent antibody. While buried framework residues can affect the
`binding of the humanized antibody through an indirect mechanism, i.e. by influencing the
`conformation of a CDR, occassionally an exposed framework residue is involved in direct
`binding to the antigen and must be included in the humanized form (11,14).
`
`While performing this regimen for six CDRs may seem a daunting task, especially
`since crystal structures of the rodent and human antibodies are usually not available,
`nature has provided some guidance. Even though CDRs vary widely in sequence and to a
`lesser extent in size, analysis of crystal structures of Fab fragments shows that for each
`CDR (except CDR-H3) only a limited number of conformations may be utilized (13). These
`canonical conformations are dependent on size and sequence of the CDR. Hence if one
`inspects the sequence and size of the particular rodent CDR the canonical conformation
`can then be assigned. This provides information as to which CDR residues will be exposed,
`which will be buried and which framework residues may play a role in stabilizing the CDR
`conformation.
`
`How has this technique fared regarding the ability of a humanized antibody to
`reproduce the binding affinity of its parent rodent antibody? The first humanization involved
`transferring only the CDRs of the heavy chain from a murine to human antibody. When
`expressed with the murine light chain, the humanized heavy chain exhibited binding less
`than 2-fold down from the parent murine (15). Two years later another two humanizations
`were published in which both the light and heavy chain CDRs were transferred. In the first, a
`humanized anti-lysozyme antibody, binding was reduced by 10-fold (16). The second was
`more interesting since it was potentially clinically relevant, underscored the importance of a
`single buried residue for maintaining proper CDR conformation, and noted the difference
`between the sequence-based and structure-based CDR definitions (17). Directed against
`the CAMPATH-1 antigen on human lymphocytes and monocytes, the initial humanized
`version was 40-fold down in binding. Changing the human Ser27 to murine Phe27 in the
`heavy chain resulted in a version which exhibited only a 3-fold reduction in affinity. Was
`this a framework or a CDR residue? According to the structure-based definition of CDRs
`used to design the humanized anti-CAMPATH-1 antibody, heavy chain position 27 was part
`of the framework while according to the sequence-based definition position 27 was part of
`the CDR. Regardless of the definition, the marked improvement in binding when the murine
`Phe was substituted for the human Ser emphasized the need to pay attention to framework,
`as well as CDR, sequence in future humanizations.
`
`Since 1988 the technique has developed and become increasingly utilized for
`clinically relevant antibodies. The 'best-tit method, used first in 1989 (10), has remained
`the more popular method for designing the sequence of the humanized antibody than the
`later consensus method (11). More recently, in the design of an anti-IgE humanized
`antibody, both methods were evaluated (14). The study concluded that although the
`consensus sequence showed the best overall binding, no clear advantage in binding was
`evident for the consensus antibody versus the best-fit' antibody. Another humanization of
`an anti-IgE antibody, utilizing only the consensus method, was reported earlier that year
`(12).
`
`PFIZER and SAMSUNG v. GENENTECH
`IPR2017-01488
`PFIZER EX. 1196, Page 6
`
`

`

`Chap. 32
`
`Humanized Monoclonal Arntnbodiee
`
`Prosta
`
`N a
`
`Both of the anti-IgE humanizations used another recent technology in the design
`of the antibody - molecular modeling. As the database of antibody crystal structures
`grows (which consists predominanly of structures of Fab fragments), information
`accumulates on the three-dimensional structure of variable domains and the relationship
`between CORs and framework. Since the structure of the rodent antibody of interest will
`usually not be available, a computer graphical model based on crystal structures can be
`generated and used to determine which framework residues might influence CDR
`conformation. Many of the humanizations published of late include use of molecular
`modeling or crystal structure information (7,12,14,18,19). Only recently, however, have
`crystal structures of humanized antibodies been determined and compared to the molecular
`model used to design the humanized antibody (20,21). In this case, the model correctly
`predicted the conformation of the framework and five of the CDR loops. The conformation of
`the sixth CDR in the model differed from that in the crystal structure, but this same CDR also
`varied in conformation among the crystal structures of three closely related humanized
`variants (21).
`
`Though some humanizations have been successful without any alteration of the
`chosen human framework (7,8), other studies have found it necessary to alter some human
`framework residues to murine in order to attain binding equivalent to the parent murine
`antibody.
`In both of the aforementioned anti-lgE humanizations, murine framework
`In humanization of the Mikj1 antibody, eighteen
`residues were important (12,14).
`framework changes from the 'best-fit' sequence were incorporated (18). Another version of
`Mik1 with just six changes bound only about 24old less well than the version with eighteen
`changes. However the latter was superior in biological activity and it was noted that the
`bioactivities of these two versions could not be predicted solely from their binding affinities
`to the target molecule, in this case the 1L2 receptor n-chain. Such a lack of correlation
`between binding affinity and bioactivity had been noted previously for another humanized
`antibody (11). The importance of a particular framework residue may vary from antibody to
`antibody. For example, among six humanizations reported very recently, two retained the
`human amino acid at heavy chain position 71 (7,8), three changed to the murine amino acid
`(14,18,19) and in the last the murine and human consensus residues at position were
`identical and no change was required (12). This emphasizes the importance of considering
`each murine antibody as idiosyncratic, i.e. each antibody may possess its own unique CDR-
`framework interactions which are sensitive to changes in amino acid constitution. As more
`humanizations are performed and antibody crystal structures become increasing available,
`perhaps nature's rules for determining these characteristic packing interactions may
`become more evident so that they can be ascertained during the initial design of the
`humanized antibody. Some effort in this area has already been accomplished (13,22,23).
`
`While buried framework residues have been shown to be important, less attention
`has been focused on exposed (or semi-exposed) framework residues. Crystal structures of
`Fab-antigen complexes have shown that on occassion a framework residue will be involved
`in direct binding to the antigen (24). The first mention of an exposed framework residue
`important for retention of binding and bioactivity in a humanized antibody was for the anti-
`p185HER2 antibody (11). The murine kg at position 66 in the light chain, predominantly Gly
`in human kappa light chains, was shown to improve binding by 4-fold and improve
`bioactivity. More recently, having the murine amino acids at light chain residues 1 and 3
`were shown to enhance binding (14). Though the involvement of exposed framework
`residues in binding antigen may not be common, they deserve consideration when
`attempting to further improve the binding of a less than competent humanized antibody.
`
`The focus of humanized antibodies has been on the variable domains since these
`bind antigen, but some studies have begun to concentrate on the rest of the humanized
`molecule, namely the Fc portion which possesses the effector functions. While some
`humanized antibodies have exhibited the ability to effect antibody-dependent cell-mediated
`
`PFIZER and SAMSUNG v. GENENTECH
`IPR2017-01488
`PFIZER EX. 1196, Page 7
`
`

`

`322
`
`Section VI—Toptos in Drug Design and Discovery
`
`Venuti, Ed
`
`cytolysis (11,18,25,26) others have not (8). in other studies the differences between the
`various classes of lgG have been evaluated (27) and, recently, the removal of conserved
`carbohydrate in the Fc has been investigated (28). As humanizations become more
`commonplace (or are replaced by human antibodies) and clinical data become available, the
`necessity for understanding and tailoring the Fc for a desired effector function will increase.
`
`Therapeutic Use of Chimeric and Humanized Antibodies - By the mid-1980's, it was
`recognized that rodent antibodies used as therapeutics would be problematic in that they
`may elicit a HAMA response, are more rapidly cleared from serum and less efficacious at
`eliciting effector functions (29). The HAMA response was also categorized into at least two
`types of anti-antibodies: those directed against the CDRs, referred to as anti-idiotypic
`antibodies, and those directed against the remainder of the molecule, referred to as non-
`anti-idiotypic antibodies (30). Initially chimeric antibodies were thought be the solution. in
`one interesting study the immunogenicity of chimeric antibodies was evaluated by
`introducing human antibodies and human/murine chimeric antibodies into mice (31). The
`murine response to fully human antibodies was robust, with 90% of the anti-human
`antibodies directed against the constant domains and 10% directed against the variable
`domains. The chimeric antibodies also elicited a response directed against the human
`variable domains, whereas fully murine antibodies gave no response or only an anti-
`ailotypic response. The latter arise from variation in the sequence of constant (and
`sometimes variable) domains among individuals and is akin to blood group antigens (A, B,
`AS, 0). In humans, chimeric antibodies have given mixed results. For example, the first
`chimeric antibody in a clinical trial, directed against a surface antigen on adenocarcinoma
`cells of gastrointestinal origin, elicited a detectable but modest HAMA response in only 1 of
`10 patients (32). in contrast, the same investigators later reported that a different chimeric
`antibody, directed against a different surface antigen on adenocarcinoma cells, elicited a
`HAMA response in 7 of 12 patients after only one infusion (33). For both antibodies the
`HAMA response was directed against the murine variable region.
`
`The first humanized antibody used in humans, CAMPATH-1H, induced remission in
`two patients with non-Hodgkin lymphoma and showed no detectable HAMA (34). CAMPATH-
`iN was evaluated a few years later for rheumatoid arthritis (35). In this study, 7 of 8
`patients showed significant clinical benefit and all 8 patients exhibited no HAMA after one
`course of treatment. However upon retreatment, 3 of 4 patients did show a response. Of
`these, two had only an anti-idiotypic response while the third had both an anti-idiotypic and
`anti-allotypic response. Another antibody which was among the earliest humanized, the
`anti-Tac antibody (10), has been shown to be less immunogenic in cynomolgus monkeys
`than the parent murine antibody (36). As expected the response in the cynomolgus
`monkeys to the humanized antibody was anti-idiotypic while that against the murine was
`directed against the rest of the immunoglobulin. These anti-idiotypic antibodies have
`recently been shown to be directed primarily against combinations of CDRs Hi, H2 and L3
`rather than against a single CDR (37). In a similar study, murine and humanized OKT4A
`antibodies were evaluated in cynomolgus monkeys (38). All 17 monkeys which received
`murine OKT4A showed a response which was both idiotypic and non-idiotypic. While all 8
`monkeys which received the humanized OKT4A also showed a response, it was only anti-
`idiotypic, In addition, higher serum levels of the humanized antibody were maintained for a
`longer period following treatment than were serum levels of the murine OKT4A.
`
`Finally, during the past year a study was published in which mice injected with
`murine antibodies were used as a model for estimating potential patient sensitization
`against humanized antibodies (39). As in a similar earlier study (31), anti-idiotypic and anti-
`altotypic antibodies were elicited. That both types of anti-antibodies have been found
`underscores the need to determine the biological and therapeutic consequences of each.
`Anti-idiotypic antibodies directed against the CDRs may prove to be a necessary evil when
`humanized antibodies are used as therapeutics since the CDRs are the part of the
`immunoglobulin which bind antigen and therefore cannot be altered. Anti-idiotypic
`
`PFIZER and SAMSUNG v. GENENTECH
`IPR2017-01488
`PFIZER EX. 1196, Page 8
`
`

`

`Chap. 32
`
`Humanized Monoclonal Antibodies
`
`Presta
`
`323
`
`antibodies could present a problem in that they would block the function of the humanized
`antibody after a repeated application, thereby requiring an increased dose and/or
`significantly reducing the efficacy of the humanized antibody. If this is the only problem
`caused by anti-idiotypic antibodies, then one possible solution would be to have several
`bioactive humanized antibodies which bind the same antigen but have different CDRs. After
`a course of therapy with the first, the second (or even third) could then be used if a chronic
`therapy was required. Anti-allotypic antibodies, directed against idiosyncratic sequences
`of an individual, will be more problematic. Chronic therapy may require matching of a
`recipient's allotype prior to therapy (39). If the allotype of the recipient differs from that of
`the available humanized antibody then treatment might have to be abandoned or,
`alternatively, the constant portion of the humanized antibody would have to be redesigned
`for the patient's allotype. While such a tailored-to-fit' antibody is technically feasible, it
`would undoubtedly significantly increase the cost of treatment. Since no studies have been
`published on the biological consequences of either anti-idiotypic or anti-allotypic antibodies
`in humans, these questions await further clinical trial.
`
`Conclusion - Although several successful humanizations of antibodies have been
`published, clinical data at this point in time are scarce due to the relatively recent
`development of the technique. What is available supports the contention that antibodies
`hold great promise as therapeutics (3,4). Most likely, however, humanization as a
`technique will be replaced by novel methods of generating human antibodies. Two such
`technologies are the production of human antibody repertoires in transgenic mice (40) and
`from phage display libraries (41 42). Though it is destined for replacement, humanization of
`antibodies has stimulated research on the structure-function of antibodies and provided an
`important intermediary step in utilization of monoclonal antibodies as therapeutics.
`
`1. G. Kohler and C. Milstein, Nature, 2a 52 (1975).
`2. S.L. Morrison, M.J. Johnson, LA. Hevzenberg and V.T. 01, P,oc.Natt.Acad.Sci.USA, 5t 6851 (1984).
`3. G. Winter and W.J. Harris, Trends Ptiarmacol.Sci., 14, 139 (1993).
`4. L.K. Jolliffe, lntRev.lmmunol., 10, 241 (1993).
`5. L.G. Presta, Carr.0p4nn Struct.Biol., 2, 593(1992).
`6. E.A. Kabat T.T. Wu, H.M. Pony, K.S. Gottesman and C. Froefler, 'Sequences of Proteins of
`Immunological Interest', Vol. 1, 51h Ed., NIH Publ. No. 91-3242 (1991)
`7. M.E. Verhoeyen, J.A. Saunders, M.R. Pnce, J.D. Marugg, S. Briggs, E.L. Brodenck S.J. Eida, A.T.A.
`Mooren and R.A. Badley, Immunology, Z. 364(1993).
`8. M.J. Sims, D.G. Hassat, S. Brett, W. Rowan, M.J. Lockyor, A. Angel, A.P. Lewis, G. Hale, H. Waldmann
`and S.J. Crowe, Jimmunol., 151, 2296 (1993).
`9. C. Chothia and AM, Lesk, J.MoLBiol.,.196,901 (1987).
`10. C. Queen, W.P. Schneider, H.E. Selick, P.W. Payne, N.F. Landolfi, J.F. Duncan, N.M. Avdalovic, M.
`Levitt, R.P. Junghans and T.A. Waldmann, Proc.Nati.Acad.Sci.USA, 86, 10029(1989).
`11 P. Carter, L. Presta, C.M. Gorman, J.B.B. Ridgway, D. Henner, W.L.T. Wong, A.M. Rowland, C. Kotts,
`M.E. Carver and H.M. Shepard, Pn,c.Natl.Acad.Sci.USA, 8, 4285(1992).
`12.L.G. Presta, S.J. Lahr, R.L. Shields, J.P. Porter, C.M. Gorman, B.M. Fendly and P.M. Jardleu,
`J.Immunol,, J-51,2623 (1993).
`13. C. Chothia, A.M. Lesk, A. Tremontano, M. Levitt, S.J. Smith-Gill, G. Air, S. Sheriff, E.A. Padlan, 0.
`Davies, W.A.Tulip, P.M. Colman, S. Spinelli, P.M. Aizari and R.J. Poljak, Nature, 3AZ 877(1989),
`14. F. Kolbinger, J. Saldanha, N. Hardman and M.M. Bendlg, Protein Eng., 1 971 (1993).
`15. P.T. Jones, P.H. Dear, J. Foote, M.S. Neuberger and G. Winter, Nature,
`j.. 522(1986).
`16, M. Verhooyen, C. Milstein and G. Winter, Science, 22. 1534 (1988).
`323(1988).
`17. L. Roichmann, M. Clark, H. Waldmann and G. Winter, Nature, =
`18. J. Haicimi, V.C. Ha, P. Lin, E. Campbell, M.K. Gately, M. Tsudo, P.W. Payne, T.A. Waldmann, A.J.
`Grant, W.-H. Ten and W.P. Schneider, J.Immunol., .11, 1075(1993).
`19. K. Sato, M. Tsuchiya, J, Saldanha, V. Koishihara, V. ohsugi, T. Klshimoto and M.M. Bendig, Cancer
`, 851 (1993).
`Res.,
`, 969(1993).
`20. C. Eigenbrot, M. Randal, L Presta and A.A, Kossiakoff, J.Mot.Biot.,
`21. C. Ei9enbrot, T. Gonzalez, J. Mayeda, P. Carter, W. Werther, T. Hotaling, J. Fox and J. Kessler,
`Proteins, ,j.. 49(1994).
`22. J. Foote and G. Winter, J.Mol.8io1., '21487 (1992).
`23. A. Tramontano, C. Chothia and A.M. Lesk, J.Mol.Biol., 215,175 (1990).
`
`PFIZER and SAMSUNG v. GENENTECH
`IPR2017-01488
`PFIZER EX. 1196, Page 9
`
`

`

`M
`
`Section VI- Topics in Drug Design axici Discovery
`
`Venut,, Ed.
`
`24. I.S. Mian, A.R. Bradwell and A,J. Olson, J.Mol.BoL, 2E, 133(1991).
`25. R.P. Junans, T.A. Waldmann, N.F. Landolfi, N.M. Avdaks,nc, W.P. Schneider and C. Queen, Cancer
`Res., 5Q, 1495(1989).
`26. P.C. Caron, M.S. Co, M.K. Bull, N.M. Avdalovic, C. Queen and D.A. Stheinberg, Cancer Res., 2. 6761
`(1992).
`, 1098(1993).
`27. J. Greenwood, M. Clark and H. Waldmann, Eur. J.lmmunoi,
`28. S. Bolt, E. Routledgo, I. Lloyd, L Chatonoud, H. Pope, S.D. Gorman and H. Waldmann, Eur.J.lmmunol.,
`403(1993).
`29. R.J. Benjamin, S.P. Cobbold, M.R. Clark and H. Waldmann, J.Exp.Med., j, 1539(1986).
`30. G.J. Jailers, T.C. Fuller, A.B. Cosimi, P.S. Russell, H.J. Winn and R.B. Colvin, Transplantation, 41, 572
`(1986).
`31. M. Brueggemann, G. Winter, H. Waldmann and M.S. Neuberger, J.Exp.Med., IZQ, 2163(1989).
`32, A.F. LoBugilo, R.H. Wheeler, J. Trang, A Haynes, K Rogers, E.B. Harvey, L Sun, J. Ghrayeb and M.B.
`. 4220(1989).
`Khazaeh, Proc.NatLAcad.Sci.USA,
`33. M.B. Khazaeli, M.N. Saleh, T.P. Uu, R.F. Meredith, R.H. Wheeler, T.S. Baker, D. King, D. Secher, L
`5461 (1991).
`Allen, K Rogers, D. Colcher, J. Schiorn, D. Shochat and A.F. LoBugho, Cancer Res.,
`34. G. Hale, M.R. Clark, P. Marcus, G. Winter, M.J.S. Dyer, J.M. Philips, L. Reichmann and H. Waldmann,
`Lancet,2, 1394(1988).
`35, J.D. Isaacs, R.A. Watts, B.L. Hazleman, G. Hale, M.T. Keogan, S.P. Cobbold and H. Waldmann, Lancet
`74.8(1992).
`36. J. Hakimi, R. Chizzonite, D.R. Luke, P.C. Familleth, P. Bailori, J.A. Kondas, P.S. Pilson, P. Lin, D.V.
`Weber, C. Spence, U. Mondini, W.-H. Tsien, J.L Levin, V.H. Gallati, L Kom, T.A. Wakthiann, C.
`Queen and W.R. Benjamin, J.Immunol.,,147,1352 (1991).
`37. W.P. Schneider, S.M. Glaser, J.A. Kondas and J. Hakimi, J.lmmunot, 150,3086 (1993).
`38. F.L. Delmonico, A.B. Cosimi, T. Kawai, D. Cavender, W.

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