throbber
A D V A N C E S IN IhlMUNOI.OCY, VOL 5fi
`Chemoimmunoconjugates for the Treatment of Cancer
`
`GEOFFREY A. PIETERSZ, APRIL ROWLAND, MARK J. SMYTH, AND
`IAN F. C. MCKENflE
`
`Austin Research Institute, Austin Hospital, Studley Road, Heidelberg 3084, Victoria, Australia
`
`1. Introduction: Concept of Targeted Chemotherapy
`
`Targeted chemotherapy involves the specific carrier-mediated deliv-
`ery of chemotherapeutic agents to tumors or other target tissues. This
`approach presumes the existence of some molecular, genetic or meta-
`bolic characteristic that differs between target and nontarget cells such
`as a structural membrane protein, a cell-surface receptor, an intracellu-
`lar enzyme, or an altered sequence in the genome. Until recently, a
`problem existed in establishing a discrete and accessible difference
`between neoplastic and normal cells; however, the isolation of some
`oncogenes and their products and the production of monoclonal and
`polyclonal antibodies to tumor-associated antigens indicate that it is
`possible to biochemically distinguish normal and tumor cells (1, 2).
`In parallel with definition of differences between normal and neoplas-
`tic cells is the development ofreagents with a high degree of selectivity
`for targets on the surface and within neoplastic cells. Over the past
`20 years, considerable interest has been focused on targeting systems
`designed to permit selective delivery of drugs, radioisotopes, and tox-
`ins to tumors for both diagnosis and therapy and a great deal of this
`research has been performed utilizing antibodies as carriers (Table I).
`As vehicles for carrying cytotoxic agents to tumors, antibodies have
`the greatest potential; however, a number of other possible carriers
`have been investigated (Table 11). The advantages of antibodies and
`other carriers include: (i) the selective delivery of the cytotoxic agent
`to the tumor cells; (ii) the slow release of the cytotoxic agent from
`the conjugate enabling prolonged exposure of the tumor cells to the
`cytotoxic agent; (iii) the preferential uptake of the cytotoxic agent-car-
`rier conjugate by tumor cells; (iv) the use of extremely cytotoxic agents
`which cannot be used alone because of toxicity; (v) the binding of
`cytotoxic agents to carriers, which may protect the agent from enzy-
`matic degradation and rapid excretion. Evidently then, the use of
`carriers to target cytotoxic agents is an attractive and provocative area
`of research; however, for the drug-targeting concept to succeed, both
`the cytotoxic agent and the carrier when conjugated must retain their
`
`301
`
`Copyright 0 1YY4 hy AcadriiilL P r a % I n <
`All rights of reproduction in m y form rebrrvrd
`
`IMMUNOGEN 2140, pg. 1
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`302
`
`GEOFFREY A. PIETERSZ ET AL.
`
`TABLE I
`AGENTS CONJUGATED TO MONOCLONAL ANTIBODIES
`
`Agent
`
`Examples
`
`Toxins (3)
`
`Anticancer drugs
`Enzymes (5)
`
`Chemotactic factors (6)
`Cytokines (7)
`Isotopes (8)
`Radiosensitizers (9)
`Photosensitizers (10)
`Liposomes (1 1)
`Nuclear magnetic resonance
`contrast agents (12)
`Plasminogenactivators (13)
`Carborane cages (14)
`Iron oxide particles (15)
`Other (16)
`
`Ricin, Pseudomonas
`exotoxin
`see Table 111
`Cytosine deaminase,
`carboxypeptidase
`fMLP
`IL-2
`Wy 1311
`Misonidazole
`Chlorin-e
`
`Gadolinium
`
`Muramyl dipeptide
`
`Note. References are in parentheses.
`
`activity in uiuo. For this and many other reasons outlined below, the
`development of the hybridoma technique to produce monoclonal anti-
`bodies (MAbs) has led to the production of more refined cytotoxic
`agent-carrier conjugates (33).
`As indicated in Tables I and 11, there are many carriers and many
`“bullets” which could be targeted. This review focuses on drug-anti-
`body conjugates; the use of toxins, isotopes, and enzymes are exten-
`reference to them is included
`sively reviewed elsewhere (34)-some
`for comparative purposes.
`
`II. Monoclonal Antibodies as Carriers
`A. DEVELOPMENT
`The use of antibodies as carriers for cytotoxic agents has been under
`consideration since the first recorded suggestions for targeting (35).
`The earliest studies made use of antisera raised by immunizing mice,
`rabbits, sheep, horses, and goats with tumor cells or their subcellular
`fractions (36-38). Antibodies reacting with normal tissue antigens
`were removed by absorption with normal tissue homogenates, thereby
`rendering the antisera relatively “tumor specific.” These approaches
`
`IMMUNOGEN 2140, pg. 2
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`CHEMOIMMUNOCONJUGATES IN CANCER TREATMENT
`
`303
`
`TABLE I1
`NONANTIBODY CARRIERS FOR CYTOTOXIC DRUGS,
`TOXINS AND RADIOISOTOPES
`
`Macromolecules
`DNA (17)
`Bovine serum albumin (18)
`Polyamino acid carriers (19)
`Dextrans (20)
`Lectins
`Concanavalin A (21)
`Hormones
`Insulin (22)
`Melanotropin (23)
`Thyrotropin (24)
`Microparticulate carriers
`Liposomes (25)
`Cells (26)
`Microspheres (27)
`Genetically engineered cytokines
`IL2-PE (28)
`IL6-PE (28)
`IL4-PE (28)
`TCFa-PE (28)
`ICF-PE (28)
`CD4-PE (29)
`IL2-DAB4M (30)
`Miscellaneous
`Arachidonic acid (31)
`Epidermal growth factor (32)
`
`Note. References are in parentheses
`
`were limited, principally because the reagents still lacked specificity
`for tumor antigens; however, many preparations were of value in formu-
`lating procedures for coupling antibodies to cytotoxic drugs (38-40).
`The desire for monospecific antibody reagents and some of the earlier
`difficulties with cell-mediated immunity to detect human tumor anti-
`gens provided some of the impetus for developing MAbs (4 1) and the
`advent of the hybridoma technology and MAbs represented a real
`advance in the field of tumor immunology (33). As a result of this
`technology, the production of many MAbs and the subsequent identifi-
`cation of tumor-associated antigens have considerably extended the
`possibilities of targeting cytotoxic agents to tumors. MAbs, by virtue
`of their unique specificity, the ability to select for the desired affinity,
`and ease of production, have surpassed polyclonal preparations as
`carriers for targeted delivery of cytotoxic agents to tumors. Indeed,
`
`IMMUNOGEN 2140, pg. 3
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`304
`
`GEOFFREY A. PIETERSZ ET AL
`
`the prospect of using antibodies as vehicles for isotopes, drugs, and
`toxins only became a reality with the development of MAbs with some
`degree of specificity for tumors. Reexploration of this approach using
`MAbs has been strengthened by studies which demonstrated that xeno-
`geneic MAbs could not only be safely administered to patients and
`localize in tumors (42) but could also have a therapeutic effect of their
`own in xenograft models (43, 44) and in patients with leukemia and
`lymphoma (45, 46). Although therapeutic effects against tumors have
`been obtained using MAbs alone, and these responses have involved
`complement-mediated effects or modulation of effector macrophages
`and natural killer cells (48), clinical responses to serotherapy have
`been variable (49,50), and animal studies indicate there are limitations
`to this approach (51). The variable antitumor effects of MAbs, however,
`may well be improved by conjugation to cytotoxic agents, given that
`the cytotoxic potential and mechanism of action of many drugs and
`toxins are well understood as many have already been used in the
`clinic.
`
`B. ANTIBODIES ALONE
`Why not antibodies alone? They clearly function in vivo after active
`or passive immunisation, particularly for infectious disease. In prac-
`tice, the use of passively administered antibodies, in cancer, has rarely
`been successful. With regard to antibodies only OKT3 (52) and Cam-
`path 1 (53) appear to be active in transplantation (both) and in lym-
`phoma-leukemia (Campath-1). The reasons have been discussed else-
`where, but essentially there are three major problems: (a) amount of
`antibody bound; (b) poor mobilization of effector mechanisms by
`mouse antibodies; and (c) the development of immune responses to the
`foreign immunoglobulin-refered
`to as human antimouse antibodies
`(HAMA). Recombinant monoclonal antibodies consisting of murine
`variable sequences and human constant domains are now available
`and some have been tested in Phase I clinical trials (54). These recombi-
`nant antibodies where the variable domains of the mouse antibodies
`are engineered onto human constant domains, binds complement and
`have antibody-dependent cellular cytotoxicity (ADCC) activity and
`therefore may activate effector function in man. Alternatively, antibody
`constant domains have been modified (e.g., altered hinged region)
`to improve various functional activities (55) for improved therapy in
`humans. How these modifications effect the HAMA response is dis-
`cussed below. More recently, additional approaches to increase the
`antitumor activity of monoclonal antibodies in uivo have been studied
`by administering biological response modifiers such as interferons (56),
`interleukins (57), and colony-stimulating factors (58).
`
`IMMUNOGEN 2140, pg. 4
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`CHEMOIMMUNOCONJUCATES IN CANCER TREATMENT
`
`305
`
`C. SPECIFICITY AND LOCALIZATION
`1. Targets
`
`To increase the selective targeting of cytotoxic agents to neoplastic
`cells, it is desirable to have clearly defined targets which ideally are
`expressed on the cell surface of tumor cells but not on normal cells.
`Despite the repertoire of murine MAbs reacting with antigens associ-
`ated with human tumors (59), there is no conclusive evidence for
`the existence of human tumor-specific antigens detected by murine
`MAbs-with
`the possible exception of the idiotype of surface immuno-
`globulin on B cell lymphomas (60). While the search continues for
`specific antitumor MAbs produced by murine and more by human
`hybridomas, the targeting of cytotoxic agents with MAbs of absolute
`specificity may not be necessary. For example, an antigen which has
`a higher expression on tumor than normal cells or is absent on vital
`normal cells (e.g., hemopoietic stem cells) may be a suitable target for
`the delivery of cytotoxic agents. Many potential antigens have been
`found to be highly tumor-associated, three of the best known examples
`being a-fetoprotein (AFP) (61), carcinoembryonic antigen (CEA) (62),
`and common acute lymphoblastic leukemia antigen (CALLA) (63).
`A better definition of the known tumor-associated antigens, such as
`CEA and AFP, has been possible using MAbs recognizing different
`epitopes (64-66). CEA is representative of many tumor-associated
`antigens and is one of the most widely studied tumor markers. It is
`immunologically a complex macromolecule, expressing both protein
`and carbohydrate determinants on colon carcinoma cells (67) and has
`been reported to be cross-reactive with NCA-1 (68), NCA-2 (69), nor-
`mal biliary glycoprotein (70), and some circulating cells (71). These
`types of cross-reactivities with normal tissues, displayed by many
`MAbs-binding tumor-associated antigens, make it necessary to clearly
`define the properties of the MAbs both biochemically and by immuno-
`histochemical techniques before they are used as carriers for cytotoxic
`agents. Epitope analysis and immunohistology has allowed a number
`of CEA-specific and cross-reactive antibodies to be identified, prvvid-
`ing the opportunity of using different mixtures of antibodies to over-
`come heterogeneity of CEA epitope expression found within individ-
`ual tumors and between different patients (72). The isolation and
`characterization of cDNA clones encoding CEA reveal a highly con-
`served repeating structure (73). Antibodies to various parts of the CEA
`molecule have been made as an effort to obtain more specific antibod-
`ies (74). MAbs against CEA have proved to be of value for the radioim-
`munoassay of human circulating CEA (75), for the radioimmunolocal-
`ization of tumors (76), and as carriers of cytotoxic drugs such as
`
`IMMUNOGEN 2140, pg. 5
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`306
`
`GEOFFREY A. PIETERSZ ET AL.
`
`vindesine (VDS) (77). Studies using a VDS-anti-CEA MAb conjugate
`demonstrated that the in vivo efficacy against lung and colorectal carci-
`noma xenografts in nude mice was dependent on target antigen expres-
`sion on the tumor xenograft cells. This not only demonstrated the
`specificity of MAbs as carriers for cytotoxic agents, but also showed
`that the amount of antibody binding the tumor cells was limited by
`the number of antigen receptors expressed.
`With the production and characterization of MAbs, there has been
`an identification of many other tumor-associated membrane markers
`of potential value in the diagnosis and therapy of tumors such as
`melanoma (78), lung carcinoma (79), breast carcinoma (80), leukemia,
`and lymphoma (81-83). Some of the antigens identified are carbohy-
`drate structures present on glycoproteins and glycolipids (84), while
`other MAbs have been shown to recognize noncarbohydrate deter-
`minants on glycoproteins (85). Indeed, it has been demonstrated
`that a protooncogene HER-e/neu, an epidermal growth factor (EGF)
`receptor-like member of the tyrosine-specific protein kinase family,
`was amplified in 30% of subjects with human breast cancer, and the
`presence of HER-2 was a significant bad prognostic factor (86). The
`cell-surface membrane protein encoded by this protooncogene is the
`type of target that may be a good candidate for targeted chemotherapy,
`particularly as the target antigen has an integral function in the growth
`of tumor cell. To date, however, MAbs to protooncogene products do
`not appear to be suitable vectors for targeting cytotoxic agents such
`as methotrexate (MTX) to tumor cells (87), possibly due to their poor
`access to oncoproteins in tumor cells as most are expressed intracellu-
`larly and not on the surface (88). However, totally intracellular and
`intranuclear oncogene proteins have been found to be expressed on
`the cell surface as small peptides-in
`association with MHC class I
`molecules (89). In the future, it may be possible to make antibodies
`to these peptides to enable cell-surface detection. At present, they are
`only detected by cytotoxic T cells.
`The cDNAs coding for several other tumor-associated antigens have
`been cloned, CALLA (go), the protein core of the human milk fat
`globule antigen (HMFG) (91) and human prostatic acid phosphatase
`(PAP) (92). cDNAs have been characterized and have led to the produc-
`tion of second-generation antibodies raised to peptide sequences
`based on translation of cDNA (93). These antibodies could be more
`specific than monoclonal antibodies.
`
`2. LocalizationlHeterogeneity
`While highly expressing tumor antigens are required for the specific
`targeting of cytotoxic agents with MAbs, other important factors are
`
`IMMUNOGEN 2140, pg. 6
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`CHEMOIMMUNOCONJUGATES IN CANCER TREATMENT
`
`307
`
`the localization of MAbs to tumor cells in uiuo and their uniform
`penetration into the tumor. Quantitative analysis of the biodistribution
`of radiolabeled antibodies in patients is required as this provides an
`assessment of their tumor : normal tissue localization ratio and ade-
`quately defines the potential of that MAb to localize a particular cyto-
`toxic agent to a particular tumor. MAbs that have been shown to local-
`ize to tumors in uiuo include anti-CEA antibodies to colorectal cancer
`(76,94), anti-PAP antibodies to prostatic cancer (95), anti-HMFG anti-
`bodies to breast cancer (96), MAb p97 to melanoma (97), and 791T/
`36 to bone and soft tissue sarcomas (98). However, radioimmunolocal-
`ization has revealed marked variation in the localization of antitumor
`antibodies in animal and human tumors (99, loo), some MAbs localiz-
`ing in tumors at concentrations fivefold greater than that of normal
`tissues, others failing to selectively localize tumors at all in vivo.
`It is clearly important to coat many antigen-binding sites on the
`tumor with the drug-antibody conjugate and therefore deliver the
`maximum dose of drug to the tumor. Within a given tumor mass not
`all tumor cells are alike and individual cells may not express every
`tumor antigen and result in antigenic heterogeneity. Antitumor MAbs
`have not been demonstrated to bind to all the individual tumor cells
`in human tumor xenografts or patients, and autoradiography of tissue
`sections after localization of MAb 791T/36 in human osteogenic sar-
`coma xenografts revealed peripheral localization and low levels of
`penetration (98). The nonuniform distribution of anti-CEA antibodies
`and their F(ab‘), in colon carcinoma xenografts has been noted (94),
`and in patients l3lI-1abeled 791T/36 did not bind many colon carci-
`noma cells, but rather bound to tumor-pseudoacini and stroma (101).
`Thus the limitation ofany one MAb not only is due to the heterogeneity
`of antigen expression, but is also a reflection of its inability to bind
`every tumor cell due to poor access. Poor tumor vascularity and the
`inefficient transport of MAb across the capillary endothelium into the
`tumor may prevent MAbs from reaching every neoplastic cell in a
`solid tumor. Whether a mixture of several antitumor MAbs can over-
`come these difficulties is yet to be determined, although “cocktails”
`of several MAbs should reduce the more fundamental problem of
`antigen heterogeneity. In this regard, immunohistological staining and
`flow cytometry with MAbs have demonstrated the heterogeneity of
`tumor cell populations (both with regard to antigen density as well as
`the presence or absence of antigen) and emphasized that mixtures of
`several anticolon carcinoma antibodies can react with almost all colon
`carcinomas (102, 103). Of relevance is the study by Ceriani et al.
`which demonstrated an increased therapeutic effect of a cocktail of 1311-
`labeled monoclonal antibodies against a breast xenograft compared
`
`IMMUNOGEN 2140, pg. 7
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`308
`
`GEOFFREY A. PIETERSZ ET AL
`
`to individual labeled antibodies (104). The use of such radionuclide
`antibody conjugates may overcome tumor heterogeneity by killing
`bystander tumor cells lacking antigen and provides a clear advantage
`for using isotopes in immunoconjugates rather than toxin or drugs.
`Cocktails of drug-antibody conjugates have also been used to investi-
`gate the potential of overcoming problems due to antigenic heterogene-
`ity. A cocktail of up to three antibodies conjugated to Idarubicin was
`used against a human colon cancer xenograft in nude mice and it
`was found that combinations of the Ida-MAb conjugates were more
`effective antitumor agents than each conjugate alone (105).
`
`D. INTERNALIZATION
`AND MODULATION
`Once localized at the tumor cell surface an immunoconjugate may
`exert its cytotoxic effect in several ways. First, the cytotoxic agent
`need not be released from the MAb to act; in macromolecular form it
`may exert a cytotoxic action either at the plasma membrane (e.g.,
`phospholipase C) or following internalization. Alternatively, the drug
`or active drug-containing fragment of the conjugate may have to be
`released to be active. This dissociation could occur in the extracellular
`space, at the cell surface, or intracellularly due to degradation by
`lysosomal enzymes. The latter mechanism of action requiring internal-
`ization of the drug-MAb conjugate and delivery to the lysosomes
`constitutes the “lysosomotropic” approach to drug targeting (106).
`The present status of MAbs, as carriers of cytotoxic agents, suggests
`that these reagents should ideally be internalized by tumor cells (100,
`107). The pathways that MAbs use to enter different cell types have
`not been extensively studied, although the internalization of MAbs
`by leukemic, melanoma, and breast carcinoma cells has been examined
`(108-1 11). The fact that some toxin-MAb conjugates have poor cytotox-
`icity (112-114) may be due to the inability of certain antibodies to be
`endocytosed, either due to a low binding affinity or because the target
`antigen is not readily internalized. Indeed, a toxin-F(ab’)2 conjugate
`has been shown to be more cytotoxic than the corresponding Fab’
`conjugate in uitro, suggesting that crosslinking of MAb-antigen com-
`plexes and endocytosis was important for this conjugate’s cytotoxic
`effect (115). The cell type may also influence internalization, as MAbs
`directed against different epitopes of the same target antigen on differ-
`ent cells induce different internalization responses (116). Also given
`that the A-chain fragments of toxin molecules such as ricin require
`translocation from an endosomal compartment to avoid deactivation
`by lysosomal enzymes, the proximity of the MAb-bound epitope to
`
`IMMUNOGEN 2140, pg. 8
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`CHEMOIMMUNOCONJUGATES IN CANCER TREATMENT
`
`309
`
`the plasma membrane may have some bearing on the cytotoxic effect
`of some A-chain toxin-MAb conjugates.
`The availability of large panels of antitumor MAbs to different
`epitopes on target antigens is no guarantee of successful targeted
`chemotherapy, as antigenic modulation and/or immunoselection of
`antigen-negative tumor cells may prevent effective therapy. Antigenic
`modulation is defined as the redistribution of surface antigen after
`binding of antibody (117) and may involve internalization and degrada-
`tion of the antigen or shedding of the antibody-antigen complex from
`the cell surface. The factors regulating antigenic modulation are not
`clear. Some antigens modulate rapidly, slowly, or not at all, others
`modulate when exposed to multiple antibodies directed against unre-
`lated antigens (118). Loss ofantigen from the cell surface by internaliza-
`tion has posed problems for the use of’ MAbs aIone in clinical therapy;
`however, the toxicity of some agents and the intracellular nature of
`their mechanism of action suggest that MAb-targeted cytotoxic agents
`could eradicate tumor cells which modulate by antigen internalization
`(1 19). In targeting surface antigens that are modulated by shedding,
`effective therapy may require that the cytotoxic agent be released from
`the surface-bound antibody before shedding occurs and that the agent
`be cytotoxic extracellularly, or intracellularly independent ofthe MAb.
`Selectivity may be compromised, however, and, therefore, target anti-
`gens that are modulated by cell-surface shedding are not good candi-
`dates for MAb-targeted chemotherapy. Immunoconjugates prepared
`using a noninternalizing antibody have also shown antitumor effects
`in mice. These conjugates used a vinca alkaloid derivative linked to
`antibody via a hydrazide linkage and antitumor efficacy was attributed
`to the release of drug at the tumor site (120).
`Another possible drawback for therapy by immunoconjugates is the
`presence of circulating tumor-associated antigens resulting from cell-
`surface shedding or tumor cell destruction. These bind conjugates in
`the serum before they have an opportunity to eradicate tumor cells.
`As an example, the presence of serum CEA has made MAbs against
`CEA difficult to use as diagnostic agents in vivo (121). Similarly, in B
`cell tumors with circulating monoclonal globulins and where the MAb
`is commonly directed against the idiotype of the antibody secreted by
`the tumor cell little may reach the tumor; some reduction in circulating
`antigen can be obtained by plasmaphoresis but the reduction is short-
`lived (122).
`Several novel strategies for targeting have been described and are
`based on the enzymatic activation of “prodrugs,” i.e., inactive forms
`of the drug which become active only when the drug is released. In
`
`IMMUNOGEN 2140, pg. 9
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`310
`
`GEOFFREY A. PIETERSZ ET AL.
`
`these studies enzymes such as alkaline phosphatase (123), or cytidine
`deaminase (124), can be coupled to monoclonal antibody and, after
`the complex localizes to the surface of tumor cells, a nontoxic prodrug
`is administered. These prodrugs are activated at the target site and
`diffuse into the tumor or act on the surface. For such therapy, MAbs
`to noninternalizing antigens are required.
`
`E. SIZE
`One of the factors that controls the uptake of MAb by the tumor is
`the permeability across the capillary wall of tumor blood vessels which
`may have varying types of endothelium, depending on the site and
`origin of the tumor. The capillaries in tumors have been shown to be
`“leaky,” at least to proteins, and it is therefore assumed that they are
`relatively permeable to MAb; this may not apply to all parts of all
`tumors, and therefore measures which increase permeability, reduce
`the size of the cytotoxic agent-MAb conjugate, and define its shape
`and charge must all be considered. Some emphasis has been placed
`on reducing conjugate size by using antibody fragments: F(ab’), (MW
`-110 kDa) and Fab’ (MW -55 kDa) are smaller than intact immuno-
`globulin IgG (MW -150 kDa) and therefore may permeate into tumors
`more easily. Radioimmunolocalization studies demonstrate that anti-
`body fragments can give earlier and superior localization to intact
`MAbs (125); however, this is probably due to faster clearance of frag-
`ments from the blood, thereby reducing the background rather than
`giving absolutely higher levels of antibody in the tumor. In addition,
`cleaving the Fc portion from the antibody molecule should decrease
`nonspecific binding to nontumor cells possessing Fc receptors and
`reduce the immunogenicity of xenogeneic MAbs. Not only are frag-
`ments cleared more rapidly than intact IgG (126), but the lower affinity
`associated with univalent Fab’ binding reduced cytotoxicity of
`toxin-Fab’ conjugates compared with their corresponding divalent
`F(ab’), and intact IgG conjugates in uitro (127). The preparation of
`fully active fragments by current techniques (128) is not always easy,
`each antibody requiring optimization of fragmentation and activity
`testing in uitro and in uiuo. Therefore MAbs will vary with respect to
`the in uivo localization of their Fab’ and F(ab’), fragments depending
`on the particular behavior of the MAb after fragmentation or coupling
`to cytotoxic agents. Using a nontoxic derivative of the alkylating agent
`melphalan N-acetylmelphalan (N-AcMEL), fewer molecules of N-
`AcMeL were coupled to F(ab’)z than intact MAb and the antitumor
`activity against a murine thymoma was only marginally better (129,
`130). The use of F(ab‘), immunoconjugates may be useful and neces-
`
`IMMUNOGEN 2140, pg. 10
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`CHEMOIMMUNOCONJUGATES IN CANCER TREATMENT
`
`31 1
`
`sary for drug conjugates of methotrexate, aminopterin, and other ex-
`tremely toxic drugs. Immunoconjugates are sometimes more toxic than
`fragments are used as carriers (131, 132).
`unbound drug unless F ( a l ~ ’ ) ~
`Advances in the production of genetically engineered antibodies and
`antibody fragments may reduce some of the problems associated with
`antibody fragments produced by enzymatic methods (133). Fab and
`F(ab’), fragments have been expressed in transfected cells and more
`importantly chimeric fragments produced in this way may decrease
`the immunogenicity (see below) associated with the administration of
`murine antibody in humans (134, 135). Single-chain antigen-binding
`proteins (SCA) have also been engineered. These proteins consist of
`regions linked via a peptide (136). The SCAs
`antibody V, and V,
`retain full antigen-binding capacity with some decrease in binding
`affinity (137). In a study by Yokota et d., the penetration of various
`immunoglobulin forms, IgG, F(ab‘),, Fab’, and sFv were compared in
`uiuo using autoradiography (138). The main conclusion from this study
`is that maximum penetration for sFv was at 0.5 hr while IgG reached
`maximum at 48-96 hr postinjection. The percentage injected dose per
`gram for IgG, F(ab‘)2, Fab’, and sFv were 27.2, 19.2, 3.7, and 1.7,
`respectively. Furthermore, this study showed that sFv penetrated
`more deeply (more distal from blood vessels) into the tumour than
`intact IgG. The smaller size and as a result the rapid clearance from
`the blood make sFv’s exceptional for coupling radioisotopes for immu-
`noscintigraphy (139); their use as carriers of drugs still remains to be
`examined.
`
`F. IMMUNOGENICITY AND TOXICITY OF IMMUNOCONJUGATES
`
`An antibody response develops in humans after the administration
`of mouse or rat MAb to tumors (140, 141), resulting in the presence
`of circulating human anti-immunoglobulin, HAMA. These human anti-
`bodies may, on repeated exposure to the originally administered xeno-
`geneic antibodies, form immune complexes leading to hypersensitivity
`reactions (142), which preclude further immunotherapy. Surprisingly,
`however, a large amount (1.5-3 g) of foreign MAb has been injected
`into human subjects without signs of toxicity (50, 122). Immunogenic
`responses have been noted in more than half the patients receiving
`mouse antibodies, but few instances of a severe anaphylactic reaction
`have been recorded. Other relevant observations include the follow-
`ing: in one patient the therapeutic effect of the MAb was neutralized
`(143) and patients initially developed antibodies against the Fc deter-
`minants on mouse MAbs (142). In a Phase I study where a cocktail of
`three N-AcMEL-MAbs were administered using hepatic artery infu-
`
`IMMUNOGEN 2140, pg. 11
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`312
`
`GEOFFREY A. PIETERSZ ET AL.
`
`sion to patients with colorectal metastases, all patients produced a
`human anti-mouse response (144). Xenogeneic antibodies given to
`patients may have a serious limitation to the duration and effectiveness
`of targeted chemotherapy.
`Different measures have been used to overcome this problem includ-
`ing: chemical engineering or murine monoclonal antibodies by adding
`polyethylene glycol residues (145); the development of immunosup-
`pressive treatment schedules with drugs or anti-T cell antibodies (146);
`the use of genetic engineering to produce stable and functional anti-
`body from the fusion of mouse variable and human constant regions
`of immunoglobulin genes (147-149) or CDR grafting onto human
`framework regions (150); the prior administration of irrelevant MAbs
`to direct antiglobulin responses away from antibodies used to target
`the cytotoxic agent; and the use oftotally human MAbs. The production
`of human MAbs has had some success (151) and improved techniques
`should bring more encouraging results (152-154). Human MAbs
`should not elicit the same intensity of immune response as murine
`antibodies as there will be no protein from a foreign species. However,
`if patients receive (say) 1-2 g of antibody of a unique idiotype, then
`antiidiotype responses may develop. Nevertheless such antibodies
`should have enhanced therapeutic potential as carriers of cytotoxic
`agents .
`
`111. Conjugation Chemistry
`A. INTRODUCTION
`While MAbs with the appropriate immunologic specificity are neces-
`sary for successful targeting of cytotoxic agents to tumors, equally
`important is the method of conjugation (155,156). Prior to conjugating
`cytotoxic agents to MAbs, it is essential to know the available reactive
`groups on both the MAb and the cytotoxic agent. Antineoplastic drugs
`are of many different types and include antimetabolites, alkylating
`agents, DNA intercalators, and antimitotic agents, all of which have
`structures that can interfere with cancer cells, leading to cell death
`(157) (Table 111). The cytotoxic part of anticancer drugs is not usually
`amenable to chemical modification due to its binding to the target,
`and therefore close attention must be focused on the structure-activity
`of cytotoxic drugs when coupling them to MAbs. Reactive groups found
`on drug molecules that can be used to link to antibodies include amino
`groups, keto groups, vicinal hydroxyl groups, phenols, free hydroxyl
`groups, and side-chain carboxyl groups (Table IV). MAbs and interme-
`
`IMMUNOGEN 2140, pg. 12
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`CHEMOIMMUNOCONJUGATES IN CANCER TREATMENT
`
`313
`
`TABLE 111
`ANTINEOPLASTIC DRUGS USED IN
`DRUG-ANTIBOVY IMMUNOCONJUCATES
`
`Antimetabolites
`
`Alkylating agents
`
`Antimitotic agents
`
`DNA intercalating agents
`
`Miscellaneous agents
`
`Methotrexate (158)
`5-Fluorouracil (159)
`Cytosine arabinoside (159)
`Aminopterin (160)
`5-Fluoro-2’-deoxyuridine (161)
`Chlorambucil (162)
`Melphalan (129)
`Mitoinycin C (163)
`Cisplatinum (164)
`Trenimon (165)
`Phenylenediamine mustard (166)
`Verrucarin (167)
`T2-toxin (168)
`Vinca alkaloids (169)
`Deacetyl colchicine (170)
`Podophyllotoxin (171)
`Daunomycin

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