throbber

`
`
`
`
`
`-
`
`Hospira v. Genentech
`Hospira v. Genentech
`IPR2017-00737
`IPR2017—00737
`Genentech Exhibit 2031
`
`Genentech Exhibit 2031
`
`

`

`
`
`Cancer Chemotherapy and Biotherapy:
`Principles and Practice
`
`
`
`
`
`
`
`

`

`
`
`Cancer Chemotherapy
`and Biotherapy:
`Principles and Practice
`
`SECOND EDITION
`
`
`
`EDITED BY
`
`Bruce A. Chabner, M.D.
`Chief, Hematology/Oncology
`Clinical Director
`
`Massachusetts General Hospital Cancer Center
`Boston, Massachusetts
`
`Dan L. Longo, M.D.
`Director, Biological Response Modifiers Program
`Division of Cancer Treatment
`National Cancer Institute
`
`Frederick, Maryland
`
`
`
`
`n Lippincott - Raven
`
`
`
`Philadelphia - New York
`
`_It______
`
`

`

`
`
`Copyright © 1996 by Lippincott—Raven Publishers. All rights reserved. This book is protected by copy-
`right. No part of it may be reproduced, stored in a retrieval system, or transmitted, in any form or by
`any means—electronic, mechanical, photocopy, recording, or otherwise—without the prior written
`permission ofthe publisher, except for brief quotations embodied in critical articles and reviews. Printed
`in the United States of America. For information write Lippincott—Raven Publishers, 227 East Wash—
`ington Square, Philadelphia, PA 19106.
`
`Library of Congress Cataloging-in-Publications Data
`
`Cancer chemotherapy and biotherapy : principles and practice / edited
`by Bruce A. Chabner, Dan L. Longo -—- 2nd ed.
`p.
`cm.
`Rev. ed. of: Cancer chemotherapy.
`Includes bibliographical references and index.
`ISBN 0—397-514184 (hard : alk. paper)
`1. Cancer—Chemotherapy.
`2. Cancer—Immunotherapy.
`(Dan Louis), 1949—
`.
`II. Cancer chemotherapy.
`[DNLM: 1. Neoplasms—drug therapy.
`2. Biological Products—therapeutic use.
`3. Antineoplastic Agents—therapeutic use. 4. Chabner, Bruce. QZ 267 C21515 1996]
`RC271.CSC32219
`1996
`616.99’4061—dc20
`DNLM/DLC
`for Library of Congress
`
`I. Longo, Dan L
`
`95-38920
`CIP
`
`The material contained in this volume was submitted as previously unpublished material, except in
`the instances in which credit has been given to the source fi'om which some of the illustrative material
`was derived.
`Great care has been taken to maintain the accuracy ofthe information contained in the volume. How—
`ever, neither Lippincott—Raven Publishers nor the editors can be held responsible for errors or for any
`consequences arising from the use of the information herein.
`The authors and publisher have exerted every effort to ensure that drug selection and dosage set forth
`in this text are in accord with current recommendations and practice at the time of publication. How-
`ever, in view of ongoing research, changes in government regulations, and the constant flow of infor-
`mation relating to drug therapy and drug reactions, the reader is urged to check the package insert for
`each drug for any change in indications and dosage and for added warnings and precautions. This is
`particularly important when the recommended agent is a new or infrequently employed drug.
`Materials appearing in this book prepared by individuals as part of their official duties as US.
`Government employees are not covered by the above-mentioned copyright.
`
`987654321
`
`

`

`Cancer Chemotherapy and Biothcrapy, second ulitim,
`edited by Bruce A. Chabner and Dan L. Longo.
`Lippincott—Raven Publishers, Philadelphia ©1996
`
`
`CHAPTER
`
`Antibody-Based lmmunotherapies
`for Cancer
`
`
`
`.
`‘
`
`Richard P. Junghons, George Sgouros,
`and David A. Scheinberg
`
`Monoclonal antibodies (mAbs) are remarkably versatile
`agents with potential therapeutic applications in a num—
`ber of human diseases, including cancer. mAbs have long
`promised to offer a safe, specific approach to therapy.
`More than a decade of preclinical evaluation and human
`clinical trials have identified new strategies for the use of
`mAbs, as well as a number of difl‘icult obstacles to their
`effective application. Although the use of antibodies as
`targeting agents dates to the 19503,1 it was not until
`methods for production of mAbs appeared in the late
`19705,2 whereby reproducible lots of a defined molecule
`could be produced in quantities adequate for clinical
`study, that the properties of antibodies as therapeutic
`agents for cancer could be studied appropriately.
`Five approaches to therapy are used in the application
`of mAbs in humans in vivo. First, mAbs can be used to
`focus an inflammatory response against a target cell. Bind-
`ing of a mAb to a target cell can result in fixation of
`complement, yielding cell lysis or can result in opsoniza—
`tion, which marks the cell for lysis by various effector cells
`such as natural killer (NK) cells, neutrophils, or monocytes.
`Second, mAbs may be used as carriers to deliver another
`small molecule, atom, radionuclide, peptide, or protein to
`a specific site in vivo. Third, mAbs may be directed at crit—
`ical hormones, growth factors, interleukins, or other regu—
`latory molecules or their receptors in order to control
`growth or other cell fimctions. Fourth, anti-idiotypic mAbs
`may be used as vaccines to generate an active immune re—
`sponse. Finally, mAbs may be used to speed the clearance
`of other drugs or toxins or can fundamentally alter the
`pharmacokinetic properties of other therapeutic agents.
`For example, mAbs may be fused to drugs or factors to in-
`crease their plasma half-life, change their biodistribution,
`or render them multivalent. Alternatively, mAbs may be
`used to clear previously infused mAbs from the circulation.
`Yet despite the diversity of approaches, significant
`problems remain that are peculiar to mAbs. mAbs are
`large, immunogenic proteins, often of rodent origin, that
`rapidly generate neutralizing immune responses in pa-
`tients within days to weeks after their first injection. The
`sheer size ofmAbs, 150 kDa for IgG to 900 kDa for IgM,
`100 times larger than typical drugs, makes their pharma-
`cology (particularly diffusion into bulky tumors or other
`
`extravascular areas) problematic for effective use. Many
`early mAbs or mAb constructs were either poorly cy—
`totoxic or relatively nonspecific, rendering them ineffec—
`tive. Moreover, the high degree of mAb specificity that is
`routinely achievable now can work against mAbs, since
`tumor cells that do not bear the specific antigen target
`may escape from cytotoxic effects.
`Within this context, it is still clear that mAbs have great
`potential to be safe and effective anticancer agents; recent
`clinical investigations have highlighted several areas where
`mAbs can be eEective, either alone or in combination with
`other, more conventional agents.
`This chapter reviews the basic biochemical and biologic
`properties of mAbs and the most commonly used deriva-
`tives
`(immunotoxins,
`radioimmunoconjugates, mAb
`fragments), discusses the pharrnacologic issues peculiar to
`mAbs, and outlines some of the important clinical results
`with mAbs. Potential solutions to the most difficult issues
`
`in the use of mAbs will be presented. Since mAbs and con—
`jugates ofmAbs represent many different drugs, with char-
`acteristics that result from their origin (rodent or human),
`their isotypes, their structure, or the various conjugated
`toxic agents, generalizations about the prOperties of mAbs
`often may not be possible. mAb therapy of cancer is a new
`and rapidly changing field, and readers are encouraged to
`consult other reviews for more comprehensive discussions
`of individual areas?‘8
`
`IMMUNOGLOBULIN (lg) CLASSES
`
`Immunoglobulins are separated into five classes or iso-
`types based on structure and biologic properties: IgM,
`IgD, IgE, IgA, and IgG. For reasons discussed under
`“Ontogeny” (below), IgM is the primordial antibody
`Whose expression by the B cell on its surface represents the
`commitment of that cell to a particular but broad recog-
`nition space that subsequently narrows as part of the mat-
`uration response induced by antigen interactions? IgD is
`normally coexpressed with IgM on B cells and may play a
`signaling role in B—cell development. IgE, IgA, and IgG
`are mature immunoglobulins that are expressed after mat-
`uration of the response and class switch have occurred.
`
`655
`
`th-
`
`

`

`
`
`656 .
`
`CANCER CHEMOTHERAPY AND BIOTHERAPY
`
`Each of these antibodies parn'cipates in specialized func-
`tions: IgE in immediate-type hypersensitivity reactions
`and parasite immunity, IgA in mucosal immunity, and
`IgG in humoral immunity. In some cases, the antibodies
`interact with specialized receptors that link their action to
`host cellular defenses; in others, the antibody interacts
`with the humoral complement system. IgG is further
`divided into four subclasses and IgA into two subclasses.
`Heritable deficiencies
`in individual
`immunoglobulin
`classes or IgG subclasses are associated with susceptibility
`to particular infections and autoimmune disorders.10
`Table 28—1 summarizes various features of the antibodies
`that will be discussed in this section.
`
`STRUCTURE
`
`The fundamental structural elements of all antibodies are
`
`indicated by size as heavy and light chains of 55-75 and
`22 kDa (Fig. 28—1). Light chains are either kappa (K) or
`lambda (A) and are each distributed among all
`irn—
`munoglobulin subclasses. Overall, kappa comprises 60%
`of light chain in humans versus 95% of light chain in
`mouse. Heavy chains are u, 8, y, e, and a, corresponding
`
`
`
`Table 28-1. Proper’ries of Antibody Classes.
`
`to IgM, D, G, E, and A, and conferring the biologic char-
`acteristics of each antibody class. Each chain is comp0sed
`of so-called irnmunoglobulin (Ig)—like domains of an-
`tiparallel beta-pleated sheets, two for light chain and four
`such domains for heavy chain, excepting IgM and. IgE,
`which have five. The amino-terminal domain of each
`chain is the variable (VI-I or VL) region that mediates
`antigen recognition; the remaining domains are constant
`regions designated CL for light chain and CH1, CH2,
`and CH3 for heavy chain (and CH4 for p, and 6). Be-
`tween CH1 and CH2 is the so-called hinge region, which
`confers flexibility on the antibody “arms” and suscep-
`tibility to proteases (below) excepting IgM and IgE in
`which the CH2 domain itself serves this role.
`Heavy (H) and light (L) chains are normally paired 1 :1
`with each other, but the smallest stable unit is a four-
`chain (HL); structure (see Fig. 28-1), for a nominal total
`mass of 150 to 160 kDa for IgG and higher for other iso-
`types (see Table 28—1). While isolated light chain (Bence
`Jones protein) exists in small amounts as monomers or
`dimers in normal individuals, the isolated heavy chain is
`stable only in association with another heavy chain to
`mask the hydrophobic surface on the carboxy-terminal
`CH3 domain (CH4 in IgM) and to generate a high-
`affinity noncovalent interaction between the molecular
`
`
`PROPERTY
`
`IgG
`
`IgA
`
`IgM
`
`IgD
`
`IgE
`
`Usual molecular form
`Molecular formula
`
`Monomer
`
`-y2K2 or 72x2
`
` Pentamer
`
`Monomer, dimer, etc.
`(a2K2)n or (u2k2)n
`
`Monomer
`
`Monomer
`
`(u2K2)5 or (u2)\2)5
`
`82nd or 82m did or 52x2
`
`V,CH1-3
`—
`
`V,CH1 -4
`. ..
`
`—
`190,000
`88
`13%
`0.03
`0.003
`
`2.5
`2
`
`— F
`
`ceR—I,FceR—II
`
`Mast cells
`
`—
`175,000
`78
`9%
`4
`0.3
`
`2.8
`1 or 2
`—
`
`H chain domains
`Other chains
`Subclasses
`
`Heavy chain allotypes
`Molecular weight
`Sedimentation constant
`Carbohydrate content
`Serum level (mg/100 ml)
`Percentage of total
`serum lg
`Half-life (days)
`Antibody valence
`Complement fixation
`(classic)
`Fc receptors
`
`Binding to cells
`
`V,CH1-3
`—
`IgG1,IgG2,
`IgG3,IgG4
`Gm (ca. 30)
`150,000
`668
`3%
`1250 i 300
`75—85
`
`23 (IgGS 7d)
`2
`+ (IgG1,2,3)
`
`V,CHl-3
`I chain, 8 piece
`IgA1,IgA2
`
`Am (2)
`160,000
`7S,9S,1 18,148
`7%
`210 i 50
`7—15
`
`5.8
`2,4,6, .
`—
`
`.
`
`.
`
`Fc'yR-I,Fc'yR-II,
`Fc-yR-III
`Monocyte
`macrophages,
`neutrophils,
`LGLs
`
`—
`
`V,CH1~4
`I chain
`
`Min (2)
`950,000
`19S
`10%
`125 1- 50
`5-—10
`
`5.1
`10
`+ +
`
`P
`
`Other biologic properties
`
`Secondary Ab
`response;
`placental
`transfer
`
`Secretory anfibody
`
`Primary Ab response;
`B-cell surface Ig,
`rheumatoid factor
`
`B—cell
`surface 1g
`
`Homocytotropic
`Ab; anaphylaxis;
`allergy
`
`
`

`:fi‘l7gig
`
`'is
`
`_-sdsééE-‘Jf
`
`
`Sigma"-'‘
`
`

`

`
`
`
`
`|_
`
`Antibody-Based Immunotherapies for Cancer
`
`Figure 28-1. Antibody structure. The structural relationships and Functions of domains of IgG. (Reprinted with permission From Wasser-
`man RL, and Capra JD, Immunoglohulins. ln Horowitz MI, Pigman W, eds, The glycoconjugates. NY: Academic Press, 1977: 323.)
`
`
`
`
`
`
`
`LIGHT CHAIN ___
`HYPERVARIABLE{
`REGIONS
`
`LIGHT CHAIN
`
`HEAVY CHAIN
`
`HEAVY CHAIN
`HYPERVARIABLE
`REGIONS
`
`CARBOHYDRATE
`
`
`
`ANTIGEN
`(BINDING) F°b
`
`BIOLOGICAL
`ACTIVITY Fc
`MEDIATION
`
`
`
`INTERCHAIN
`L DISULFIDE
`I
`BONDS
`
`INTRACHAIN
`DISULFI DE
`BONDS
`
`VL AND VH1VARIABLE REGIONS
`CL AND CH: CONSTANT REGIONS
`
`
`
`halves.n It is notable that the inter—heavy chain disulfides
`and heavy—light chain disulfides are not required for as-
`sembly, which is mediated through primary noncovalent
`interchain interactions. IgE and IgG are composed of a
`single (HL); unit, Whereas IgM exists as a pentamer of
`(HL); units joined by disulfide bonding with a third I-
`chain component. IgA exists mainly as a monomer in
`serum but in secretions exists primarily as a dimer plus
`trirner and higher forms in which the oligomers are linked
`by I chain as well as the fi'agment of secretory chain (se-
`cretory piece) that is involved in the mucosal transport.
`The V region itself is composed of subdomains: rela—
`tively conserved framework regions interdigitated with
`the
`so-called
`complementarity—determining regions
`(CDRs) [also termed hypermriable segments (HVSs)] that
`make primary contact with antigen”,12 (see Fig. 28-1).
`There are three CDRs in each heavy and light chain that
`may participate in antigen binding. The V regions should
`be seen as juxtaposed three-finger gloves, with the CDRs
`covering the tips (Fig. 28—2), arrayed in a broad contact
`surface with antigen (Fig. 28-3).
`Antibodies are glycoproteins. Glycosylation ofproteins
`plays various roles including solubility, transport, confor-
`mation, function and stability. Carbohydrate is located
`mainly in antibody constant domains, with a lower fre—
`quency in V regions (see data on M195 below).13 IgG
`contains a major conserved glycosylation site in CH;
`which contributes to the conformation ofthis domain that
`is crucial to the functional ability to bind to complement
`and to Pay receptors.
`The IgG antibody “unit” has been defined in terms of
`susceptibility to proteases that cleave in the exposed, non-
`
`_I
`
`folded regions of the antibody (see Fig. 28-1). A tabula-
`tion of anu'body firagments and engineered or synthetic
`products is presented in Table 28-2. Fab contains the
`V region and first constant domain ofthe heavy chain (VH
`+ CH1 = Pd) and the entire light chain (L); Fab’ includes
`additionally a portion ofthe H chain hinge region and one
`or more flee cyteines (Fd'); Fab’2 is a dimer ofFab’ linked
`through hinge disulfide(s); and Fv is a semistable antibody
`fragment that includes only VH + VL, the smallest anti-
`gen-binding unit. Fe is the C-terminal crystallizable frag-
`ment that includes the complement and Fe receptor—bind-
`ing domains (below). Genetically engineered products
`include the ACHZ constructs, lacking the second constant
`domain ofheavy chain, which behave like a Fab’2, with bi-
`valency, abbreviated survival, and lack of interacu'on with
`host effector systems, but which do not require enzymic
`processing.13a st is Fv with a peptide linkage engineered
`to join the C-terminus of one chain to the N-terminus of
`the other for improved stability. More advanced products
`have been designed that conceptually represent the anti-
`gen—binding domain in a single peptide productm’; this is
`not related structurally to an antibody and is therefore
`considered an antibody mimic.
`
`ONTOGENY
`
`Antibodies represent the most strikingly evolved, adaptive
`system possibly in all of biology. It is both an ancient and
`evolved system, present
`in mammals, birds, reptiles,
`amphibians, teleosts, elasmobranchs (sharks), and possi-
`bly cyclostomes (hagfish, lampreys), which, if true, would
`
`

`

`658
`
`
`
`CANCER CHEMOTHERAPY AND BIOTHERAPY
`
`
`Figure 28-2. Space-filling model of human [96] antibody
`with CDRs in color representing anti-Tac-H; human myeloma
`protein Eu with CDRs grafted from murine anti-Toe. (Photo
`provided courtesy of Dr. C. Queen.) (See Color Plate 1.)
`
`I
`
`
`
`include all chordates.14 Its most diverse representation of
`classes and functions is found in mammalia. The power of
`antigen recognition begins with an inherited array of du-
`plicated and diversified germ—line V genes, a random mu-
`tational process that creates novel CDRs, a combinatorial
`selection process that amplifies the germ-line capabilities,
`and a controlled and directed mutational process that
`hones the specificity and matures the antibody into a
`high-affinity, antigen-specific reagent.
`The biologic expression of antibody begins with the
`B-cell progenitor, which undergoes a series ofmaturation
`steps that begins with V gene selection for heavy chain
`followed by light chain V selection that yields surface
`expression and secretion by the mature B cell. Upon in-
`teraction with antigen, the B cells are activated to prolif-
`erate, secrete antibody and undergo CDR mutagenesis
`and affinity maturation, and finally to undergo chain
`switch and plasma cell conversion. Plasma cells remain in
`
`tissues, spleen, or lymph nodes and secrete large quanti-
`ties of antibody, the sole functiOn of this terminally dif.
`ferentiated cell.9
`.
`The genes of heavy and light chains share important
`features of structure and maturation. Each gene locus
`contains widely separated variable, constant, and so-called
`minigene domains that are placed into juxtaposition by
`DNA recombination mechanisms. The minigenes~
`diversity (D) and joining (I) regions for heavy chain and
`I regions for light chain—contribute to or constitute,
`with modifications, the CDR3.15 The kappa and lambda
`light chain loci are located on chromosomes 2 and 22,
`respectively, but all heavy chains are contained within a
`single massive locus on chromosome 14.
`To understand the nature of the generation of the
`antibody repertoire, it is instructive to recapitulate what
`is known about germ—line diversity. On the heavy chain
`locus, there are an estimated 80 functional VH genes, 12
`D regions, and 6 I regions for a potential of 6000 com-
`binationsm‘19 (Fig. 28-4). There are roughly 80 V kappa
`[VK] light chain and 5 I kappa [In] domains, which, ran-
`domly associated, can generate 400 combinations (the
`lambda locus contains a smaller number of distinct V
`genes). A simple arithmetic calculation suggests that
`VKVH combinations alone could generate a diversity of
`approximately 2 X 106. Yet even this number is conserv-
`ative, because this diversity is amplified in turn by errors
`in recombination and processes called N and P nucleotide
`addition in CDR3 which add enormously to the poten-
`tial complexity, in theory exceeding the total lifetime B—
`cell output by several orders of magnitude.” However,
`many authors have cautioned that the mathematical di-
`versity does not allow for the redundancy in configura-
`tions that could provide equivalent binding domains; in
`terms of antigen binding, the practical diversity is proba-
`bly in the 1 to 10 X 10‘5 range. The smallest “complete”
`immune system is that of the young tadpole with 106 B
`cells, which suggests that repertoires of 105 to 10‘5 con-
`stitute a sufficiently complete topologic set for meaning-
`fully diverse, if not exhaustive, antigen recognition.21
`V gene selection is based on random expression
`followed by specific amplification. It has been argued on
`physicochemical grounds that 105 diEerent antibody
`molecules are sufficient to create a topologic set that
`recognizes any antigen surface with an affinity of 105 t0
`10'5M‘1,22 a weak but biologically important number that
`corresponds to recognition affinities of naive antibody-
`antigen contacts that are often broadly polyreactive-
`B cells express antibody, principally IgM and IgD, on
`their membranes. On contacting antigen, these cells are
`stimulated to divide and undergo CDR mutations. Sub-
`sequent binding and stimulation are in proportion to the
`strength ofthe binding reaction; hence there is an in ViVO
`selection for mutations that enhances the affinity of the
`antibody for the antigen, a process termed afi‘inity 14W“
`“ration.23 Simultaneous with this increased affinity is a
`narrowing of the specificity, with the antibody shedding
`its early polyreactive phenotype. The cells then undergo
`
`“class switch” to one ofthe mature antibodies (IgG, 13A?
`
`-a
`
`Wyn-cs
`rOenigma:
`"Fm3”
`
`.....Inm-"_I___.__._w
`
`.1"
`
`

`

`Antibody-Based lmmunotherapies For Cancer
`
`
`Figure 28-3. Antigen-antibody binding surtace luxtaposition. The V region (Fv) oi antibody (right) binds to influenza virus protein
`neuraminidase (left) in the top panel. The VH (red) and VL (blue) are separately colored to show their respective binding contribu-
`tions. The bottom panel ottsets the two molecules by 8
`to show the complementarity of surfaces that promotes the binding interac-
`tion. The stippled surface of the neuraminidase defines the antigen
`"epitope." (Photo provided courtesy of Drs. P.M. Colman and W.R.
`Tulip, CSIRO Australia.) (See Color Plate 2.)
`
`l
`
`
`
`
`
`IgE) by deleting out DNA between the VD] region and
`the new C region of the heavy chain, which brings this
`new constant domain in juxtaposition with the V region
`(see Fig. 28—4). (Light chain is unchanged.) Some time
`after commitment to a mature antibody, the cell will cease
`its CDR mutagenesis, affinity maturation will have been
`completed, and the B cell will undergo morphogenesis to
`a tissue-resident plasma cell.9
`
`ANTIGEN-ANTIBODY INTERACTIONS
`
`Afi‘inity is a quantitative measure of the strength of the
`interaction between antibody and its cognate antigen and
`
`is intended in the same sense as the equilibrium constant
`in the chemical mass action equation:
`
`[AB] = KalA] [B]
`
`(28—1)
`
`The equilibrium or aflinity constant is represented in
`units of M‘l. In most instances studied by x-ray crystal-
`lography, contacts between antibody and protein antigen
`are dominated by noncovalent hydrogen bonds (0—H),
`with a lower frequency ofsalt bridges (COO'— + H3N),
`with a total of about 15 to 20 contacts. The effect of
`adding a new H- bond can be estimated from the free
`energy gain (0.5 to l kcal/mol-°C) and from AG =
`—RTln Ka to yield affinity increases of approximately
`
`

`

`660
`CANCER CHEMOTHERAPY AND BlOTHERAPY
`
` L Table 28-2. Antibody Fragment Definitions
`
`Ji
`
`DESCRIPTION
`
`Complete IgG
`
`Papain digest; Pd + L
`
`Pepsin digest monomer; Fd’ + L
`
`Pepsin digest dimer
`
`which antigen is 50% saturated; if the antibody is in large
`
`
`V region digestion fragment; VH + VL
`
`C region digestion fragment; crystallizable fragment
`
`Smaller fragments of PC
`
`Deleted CH2 domain; dimer of V — CH1 - CH3 + L-
`
`Single-chain Fv; VH and VL joined by peptide linker
`
`Antigen-binding unit; peptide mimic
`
`___J
`
`Although affinity and K, directly express the binding
`potential of the antibody and are the most suitable mea—
`sures for comparing affinities, the inverse of the K,”
`termed K; or dissociation constant, is expressed in molar
`units and indicates the concentration that is the middle of
`the range for the biologic action of the antibody:
`
`K4 = 1
`
`(28-2)
`
`That is, the K4 is the concentration of free antibody at
`
`DESIGNATION
`
`REPRESENTATION
`
`Fat
`
`\, /
`
`Enzyme-generated products
`
`Fd
`
`\\L
`
`Fd’
`k
`
`VFW
`
`/4 C
`
`H2
`
`CH3
`
`
`
`
`
`IICH2
`llCHs
`
`\ /
`CH3
`
`\Ia
`¢‘1»
`
`w
`
`Fab
`
`Fab’
`
`Fab’2
`
`Fv
`
`Fc(och')
`
`F (
`F )
`p c orp c'
`
`Genetically engineered products
`
`delta CH2
`
`st
`
`Synthetic products
`
`ABU
`
`
`
`3- to 10—fold. Therefore, the affinity maturation that
`takes place (or affinity that may be lost in antibody engi-
`neering) changes quickly with a relatively small change in
`the number ofbonds. That is, creating as few as three new
`hydrogen bonds may generate an aflinity enhancement of
`more than 100—fold. This has been borne out by affinity
`changes that accompanied productive amino acid substi-
`tutions in V region engineering (below). It is notable that
`antibody affinities for protein antigens are generally much
`higher than for carbohydrate antigens, which may have
`less opportunity for hydrogen-bonding interactions (but
`are also “T-independent” antigens).
`
`_‘aM‘Aghiis“5;?I
`
`
`
`

`

`
`
`Antibody-Based Immunotherapies for Cancer
`
`Figure 28-4. Generation of diversity. VJ and VDJ ioining occur in Lchain and H chain by excision of intervening DNA in the genome.
`Class switch involves deletion of intervening constant (C) domains (C, Cd, etc.) and transcription through the new proximal C region.
`C is Finally joined to the V gene by splicing of the mRNA. (Reprinted with permission from Cooper MD, Current concepts: b lympho-
`cytes-normal development. N Engl J Med 31721452, 1987.)
`
`A. Heavy chain genes on chromosome 14 p32
`
`C 3
`
`am
`
`vi>50l
`
`D(> 20)
`
`J
`
`3. Kappa light chain genes
`on chromosome 2 p11
`Vl>50l
`
`J
`
`C. Class switching
`
`by DNA deletion VDJ
`
`S};
`
`m
`
`CPL
`08
`- -
`‘L
`
`J
`Histones
`
`
`
`
`
`
`
`CY:
`
`‘l’c a!
`
`O O
`
`VDJ
`VA?
`
`5].].
`
`Syj
`05
`op
`- - I”
`
`Cy3
`
`
`
`Recombinase
`\L
`3/4 37: Cr:
`VDJ
`
`
`excess, the input antibody concentration approximates
`the free concentration. The Kd is a frequently used term,
`but its relationship to aflinity must always be borne
`in mind: i.e., low affinity = high Kd; high affinity = 10w
`Kd. For example, a IQ of 2 X 109 M‘1 implies a IQ of
`0.5 X 10“9 M(0.5 nM), or about 0.1 (Lg/ml antibody
`concentration for IgG. If antigen is in the picomolar
`(10‘12 M) range, this concentration of antibody will have
`half of antigen saturated and half of antigen will remain
`“free.” At lO-fold higher antibody concentration (1 ug/ml,
`10 X IQ), antigen will be 90% saturated and 10% free,
`
`and at 100-fold higher concentration (10 (Lg/ml, 100 X
`IQ), antigen Will be 99% saturated and only 1% unbound.
`It is a key point ofunderstanding that the ratio ofantibody
`over antigen has very little impact on the degree ofantigen
`saturation where antibody is in excess. If antibody concen—
`tration is 1 nM with a Kd of 1 nM, it does not matter
`whether antigen is 0.1 nM at the IQ, 0.1 pM, or 0.1 HM;
`antigen in each case is 50% bound, although the ratio of
`antibody to antigen is 10, 104, and 107. It is only the re-
`lation offree antibody to its K4 that determines the degree
`ofantigen saturation.
`
`

`

`662
`
`CANCER CHEMOTHERAPY AND BIOTHERAPY
`
`The afl‘inity constant K, is itself composed of two
`terms that describe the on (forward; units ofM‘l-s‘l) and
`off (back; units of 5“) rates of the reaction:
`
`K. = ikl,
`
`(28-3)
`
`To a first approximation, the forward rate is diffusion
`limited and comparable for many antibodies reacting
`with macromolecules or cell-bound structures; reactions '
`of antibodies with haptens and other small molecules in
`solution will be dominated by the faster linear and rota—
`tional difliision rates of the smaller component.24 For ex-
`ample, 0.1 nM of DNP-lysine (0.1 ng/ml) or 0.1 nM
`cell-bound HIA—AZ (50 ng/ml) mixed with specific IgG
`antibody at 10 pig/ml (65 nM) requires 0.1 second to
`react with 50% of the antigen for the hapten but requires
`4 minutes for the surface protein. Yet they have virtually
`the same affinity constant.“ This is due to the fact that
`the fast association rate is balanced by a fast dissociation
`rate for the hapten (t1/2 = 0.7 5) versus a longer stability
`for the protein antigen (1.1/2 = 6 min).
`While there are exceptions, the on rates of antibodies
`to protein and cell—bound antigens are primarily in this
`range and inversely proportional to antibody concentra-
`tion for antibody in excess of antigen (i.e., at 1 ug/ml,
`the 50% on time would be of the order of 0.5 to 1 hour).
`Accordingly, diflerences in aflinity between antibodies to
`the same cellular antigen will in many instances be seen to
`be reflective of the off rate (kg). For most such purposes,
`an antibody is generally considered of “good” affinity if
`its K, 2 109 M", Where off rate 271/2 values of an hour or
`more at 4°C are common. Association and dissociation
`times at 37°C are both accelerated relative to 4°C, on the
`order of 5 or more, frequently with a net decrease in
`antibody affinity of 2- to 10-fold. Thismust be explicitly
`tested, however, since there are instances of protein—
`ligand affinities that are enhanced by higher temperature.25
`The foregoing expresses basic principles of binding
`processes. A further important feature ofantibodies is em—
`bodied in their multivalent structures. While the on rates
`for monovalent Fab and bivalent Fab’2 constructs are
`comparable, the bivalent 01ftimes may be 10-fold or more
`longer than the monovalent constructs, yielding affinities
`that are similarly enhanced.24 To discriminate the affinity
`that is intrinsic to the V region antigen interaction from
`the effective aflinity in a bi- or multivalent interaction, the
`latter is often referred to as avidity. For monovalent inter-
`actions, avidity : affinity; for multivalent interactions,
`avidity 2 affinity. Theory predicts avidity enhancements
`that vastly exceed observed numbers, but structural con-
`straints undoubtedly restrain the energy advantage ofmul-
`tivalent binding.”27 In its extreme, steric factors constrain
`some bivalent antibodies (e.g., anti-Tacza) to bind only
`monovalently to antigens on cell surfaces but which will
`bind bivalently to antigen in solution. Yet even where anti-
`gen on the surface is not bivalently bound by antibody,
`and for all solution interactions, careful treatment ofthese
`settings will note the molarity of binding site rather than
`
`antibody in comparing Fab with higher-valency homo-
`logues. When antigens are presented multivalently on sur-
`faces of cells, viruses, or other pathogens, even the low-
`affinity IgM interactions can yield "a high-avidity, stable
`binding to such targets in vivo.
`
`humans, and is dependent largely on kidney filtration
`
`PHARMACOKINETICS/PHARMACODYNAMICS
`
`Metabolism of immunoglobulins determines the duration
`of usefulness in vivo of antibodies. Under normal condi-
`tions, the serum levels of endogenous immunoglobulins
`are determined by a balance between synthetic and cata-
`bolic rates.29 When antibodies are administered as thera—
`peutics, these catabolic rates efi'ectively specify the dose
`and schedule necessary to maintain therapeutic blood lev-
`els where steady-state exposures are targeted. Table 28-1
`lists the half-lives ofhuman antibody survivals in humans,
`with IgG having the longest survival, 23 days (IgG1,2,4;
`IgG3 survival is 7 days.) Autologous IgG survivals are
`correlated with animal size, with IgG survival in mouse of
`4 days, in dog 8 days, in baboon 12 days, in cow 21 days,
`etc.21 Of interest, however, is the observation of survivals
`in heterologous systems in which the shorter of the sur-
`vival of the IgG in the host or in the donor is dominant.
`That is, murine IgG will survive the same in humans or in
`mice, whereas human IgG will survive shorter in mice at
`a rate compatible with their own catabolism ofIgG.21 The
`site of catabolism of intact IgG remains controversial, al—
`though recent work With slowly mobilized radioisotope
`conjugates suggests that the reticuloendothelium (RE)
`system is a prominent site of this catabolism."’°’3°a
`A substantial body of knowledge exists on the metab-
`olism of immunoglobulins in various disease states.
`Conditions of protein wasting (enteropathies, vascular
`leak syndromes, burns), febrile states, hyperthyroidism,
`hypergammaglobulinemia, and inflammatory disorders
`are accompanied by significant acceleration of immuno-
`globulin catabolism.29 This information is of importance
`to understanding in vivo survival data in various clinical
`applications. In fact, the controlled conditions of testing
`immunoglobulin metabolism are rarely duplicated in
`practice, with antibody survivals typically shorter than
`suggested by the numbers above. Typically, murine anti-
`body survival t1/2 values are in the range of less than 1 to
`3 days, and antibodies with human gamma Fc domains
`(chimeric or humanized) have t1/2 values in the range of
`1 to 15 days. Some of this acceleration in clearance is
`clearly due to disease-associated catabolic factors and to
`antigen binding in vivo, but subtle changes in the drug
`structure during product preparation may have a role in
`this acceleration as well. Influence on antibody clearance
`by antigen expression in vivo is considered below.
`Antibody fragments have been studied because oftheir
`abbreviated survival and because small size may translate
`into better tissue penetration. Fab and Fab’Z have survivalS
`in vivo of 2 to 5 hours in mice, with comparable values in
`
`55
`
`
`
`__...m.n1".—
`
`R .
`
`
`
`
`
`mutt_'!'."'""2.'!'
`
`

`

`Antibody-Based lmmunotherapies for Cancer
`
`Soluble versus Surface Antigen
`
`mechanisms.29 This is not based on size alone, because the
`Fc fragment, which is comparable in size to Fab, is not fil—
`tered and has an in vivo survival of 10 days in humans.
`These rapidly catabolized fragments,
`like other

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