throbber
Downloaded from
`
`http://science.sciencemag.org/
`
`
`
`on January 19, 2018
`
`Monoclonal Antibodies in
`Diagnosis and Therapy
`
`THOMAS A. WALDMANN
`
`Monoclonal antibodies have been applied clinically to the
`diagnosis and therapy of an array of human disorders,
`including cancer and infectious diseases, and have been
`used for the modulation of immune responses. Effective
`therapy using unmodified monoclonal antibodies has,
`however, been elusive. Recently, monoclonal antibody-
`mediated therapy has been revolutionized by advances
`such as the definition of cell-surface structures on abnor-
`mal cells as targets for effective monoclonal antibody
`action, genetic engineering to create less immunogemc
`and more effective monoclonal antibodies, and the arming
`of such antibodies with toxins or radionucides to en-
`hance their effector function.
`
`T HE DEVELOPMENT OF MONOCLONAL ANTIBODY TECHNOL-
`ogy by Kohler and Milstein (1) provided an enormous
`opportunity for examination of a range of previously elusive
`issues. For example, monoclonal antibodies are being used in
`radioimmunoassays, enzyme-linked immunosorbent assays, immu-
`nocytopathology, and flow cytometry for in vitro diagnosis, and in
`vivo for diagnosis and immunotherapy ofhuman disease. However,
`in the area of immunotherapy, monoclonal antibodies are just
`beginning to fulfill the promise inherent in their great specificity for
`recognizing and selectively binding to antigens on cells.
`Monoclonal antibodies have largely been applied clinically to the
`diagnosis and therapy of cancer and the modulation of the immune
`response to produce immunosuppression for treatment of autoim-
`mune and graft versus host diseases (GVHD) and for prevention of
`allograft rejection. Human monoclonal antibodies have also been
`applied clinically against cytomegalovirus, Varicella zoster virus, and
`the various specific serotypes of Pseudomonas aeruginosa, Escherichia
`coli, and Klebsiella pneumoniae. For example, in a multicenter
`clinical trial involving 200 patients with Gram-negative bacteremia,
`mortality was reduced in patients who received an immunoglobulin
`M (IgM) monoclonal antibody (HA-LA) that binds specifically to
`the lipid A domain of endotoxin (2). In other studies, monoclonal
`antibodies that are specific for leukocyte adhesion molecules, such as
`lymphocyte functional antigen-1 (LFA-1) or intercellular adhesion
`molecule-i (ICAM-1), the adhesion partner of LFA-1, have been
`used to inhibit accumulation of neutrophils and thereby reduce
`tissue damage in animal models of bacterial meningitis, hemorrhagic
`shock, and myocardial reprofusion injury (3).
`The use of monoclonal antibodies has also been proposed for
`therapy of myocardial infarction (4), for reversal of drug toxicity
`(digitalis intoxication), and for fertility control. Despite this wide-
`
`The author is in the Metabolism Branch, National Cancer Institute, National Institutes
`of Health, Bethesda, MD 20892.
`
`21 JUNE 1991
`
`ranging interest, the "magic bullet" of antibody therapy that has
`been the dream of immunotherapists since the time of Paul Ehrlich
`has proved to be elusive (5). Only one monoclonal antibody, OKT3,
`has been licensed for clinical use. Furthermore, the initial use of
`unmodified murine monoclonal antibodies in human patients with
`cancer was disappointing, with only 23 partial and 3 complete
`remissions reported among the initial 185 patients included in 25
`clinical trials (6). A number of factors explain the low therapeutic
`efficacy observed. Unmodified murine monoclonal antibodies are
`immunogenic and elicit a human immune response to the murine
`antibodies. Moreover, most mouse monoclonal antibodies are not
`cytocidal against neoplastic cells in humans because the antibodies
`do not participate in human complement or cell-mediated cytotox-
`icity. In most cases, the antibodies were not directed against a vital
`cell-surface structure such as a receptor for a growth factor required
`for tumor cell proliferation. In an attempt to circumvent these
`problems, researchers have developed human and humanized anti-
`bodies, prepared by genetic engineering, that are less immunogenic
`than murine antibodies. Cytotoxic action has been augmented by
`arming the antibodies with toxins or radionuclides. Finally, cell
`surface antigenic targets have been defined for effective monoclonal
`antibody action.
`In this article I summarize information on in vivo use of mono-
`clonal antibodies for diagnosis and therapy, with special reference to
`the most seminal discoveries and recent advances in (i) definition of
`cell-surface structures on abnormal cells as targets for effective
`monoclonal antibody action, (ii) development of genetic engineer-
`ing approaches for creating more effective agents, and (iii) develop-
`ment of techniques for arming monoclonal antibodies with radio-
`effector function.
`nucides or toxins and thereby increasing
`Comprehensive recent reviews provide more detail (7-9).
`
`Cell Surface Antigenic Targets
`The ideal immunosuppressive monoclonal antibody would be one
`that abrogates responses to a defined antigen and preserves respons-
`es to all others. However, the monoclonal antibody that is most
`widely used clinically (OKT3) is directed against the CD3 (duster of
`differentiation) antigen of the T cell receptor complex that is
`expressed on virtually all circulating T cells. The Food and Drug
`Administration licensed the use of OKT3 for the treatment of acute
`renal allograft rejection on the basis of randomized clinical trials
`showing the superiority of this treatment (93% reversal of acute
`rejection episodes) to more conventional, broad-spectrum immuno-
`suppressive agents (75% reversal) (10). There are, however, toxici-
`ties associated with the use of OKT3. Antibodies to CD3 initiate in
`vivo T cell activation that is accompanied by release of tumor
`necrosis factor, interferon -y, and, in some cases, interleukin-2
`(IL-2), which leads to an acute clinical syndrome that involves high
`fever, vomiting, diarrhea, and occasionally respiratory distress.
`1657
`
`ARTICLES
`
`Pfizer v. Genentech
`IPR2017-01488
`Genentech Exhibit 2054
`
`

`

`Downloaded from
`
`http://science.sciencemag.org/
`
`
`
`on January 19, 2018
`
`Furthermore, therapy with antibody to CD3 leads to broad immu-
`nosuppression that is associated with an increased incidence of
`infections and B cell neoplasms.
`Monoclonal antibodies have been used to target the polymorphic
`a and 1 subunits of the human T cell antigen receptor (TCR). In
`pilot studies, one monoclonal antibody (BMA 031) specific for the
`constant region of the TCR was used successfully in treatment of
`acute organ allograft rejection and GVHD (11). Monocdonal anti-
`bodies that specifically identify the hypervariable antigen-binding
`region of the TCR represent more antigen-specific agents. Janson
`and co-workers (12) prepared a murine monoclonal antibody reac-
`tive with an idiotypic determinant ofthe hypervariable region ofthe
`TCR expressed by a patient's malignant leukemic cells. There was an
`80% reduction in the number of leukemic cells after infusion of this
`monoclonal antibody. However, associated toxic side effects includ-
`ed fever, chills, nausea, vomiting, diarrhea, and shortness of breath.
`Monoclonal antibodies directed toward idiotypic determinants of
`the TCR may also be useful in the treatment of autoimmune disease
`because it has been observed that there is an extreme restriction in the
`TCR variable region usage in the T cells responsible for experimental
`allergic encephalomyelitis (EAE), a murine model of multiple sdero-
`sis. The majority of T cells specific for myelin basic protein that can
`transfer the disease express a specific V138 phenotype (13). Further-
`more, in vivo treatment with a monodonal antibody to V138 provided
`protection from disease transfer by encephalitogenic donal T cells.
`Evidence for such restricted usage ofthe TCR variable region is being
`sought in spontaneous human neurological diseases, induding multi-
`ple sderosis and tropical spastic paraparesis.
`Most anti-idiotypic antibodies utilized in treatment of human
`lymphoid malignancies are directed toward B cell immunoglobulin
`idiotypes. Idiotypic monodonal antibodies to immunoglobulin
`were used (14) for treatment ofpatients with B cell lymphomas. The
`results were encouraging; one patient manifested an uninterrupted
`remission for more than 4 years after therapy. A partial response was
`observed in approximately halfofthe remaining patients. However,
`there were certain difficulties. During maturation, B cells use somatic
`hypermutation to increase their diversity. Thus, there is a tremendous
`idiotypic heterogeneity within the B cell population, which permits
`some neoplastic B cells to escape the attack of an antibody directed
`toward an individual idiotype. The TCR does not undergo somatic
`hypermutation and is less susceptible to this problem.
`Monoclonal antibodies that recognize surface molecules that
`facilitate cell-cell interactions are also effective immunosuppressive
`agents. For example, antibodies specific for LFA-1 or its adhesion
`partner, ICAM-1, inhibit a series of T cell functions and thereby
`inhibit allograft rejection and GVHD (15). The CD4 and CD8
`proteins, which are important in T cell activation, recognition of
`major histocompatibility complex gene products, and cooperative
`cellular interactions, are also useful targets. In animal models,
`antibodies to CD4 or CD8 suppressed allograft rejection, GVHD,
`and autoimmune reactions (16). Certain antibodies to CD4 are of
`special interest because they induce immunological tolerance in mice
`to simultaneously administered human, rat, and rabbit immuno-
`globulins. This form of tolerance was induced in T helper cells but
`not in B cells (17), and thus CD4 cells may be made tolerant when
`simultaneously confronted with certain antigens and monoclonal
`antibodies to CD4. Effective CD4 monoclonal antibody tolerance
`therapy is not confined to special protein antigens but is exploitable
`in transplantation. For example, short-term administration ofa mono-
`donal antibody to CD4 induced cellular depletion and led to indefi-
`nite survival of adult pancreatic islet cell transplants in mice without
`subsequent immunotherapy (18). Furthermore, administration of
`antibodies to CD4 ameliorated Type II collagen-induced arthritis in
`rats, murine EAE, and autoimmune myasthenia gravis (16).
`
`1658
`
`The monodonal antibodies to primate CD4 examined to date are
`not effective in tolerance induction in subhuman primate models.
`Despite this limitation, the administration of monoclonal antibodies
`to CD4 has led to improvement in preliminary clinical trials
`involving patients with severe rheumatoid or psoriatic arthritis (19).
`The inducible a chain of the interleukin-2 receptor (IL-2Ra)
`expressed on the surface of activated or abnormal T cells is also a
`target for effective monoclonal antibody immunotherapy (20).
`Interleukin-2 receptor (IL-2R) is a useful target because resting cells
`do not express the IL-2Ra, whereas the T cells participating in
`allograft rejection and abnormal T cells in certain autoimmune
`disorders express this receptor. To exploit this difference in expres-
`sion, we used anti-Tac, a monoclonal antibody that blocks the
`binding of IL-2 to IL-2Ra, in the treatment of patients with human
`T cell leukemia/lymphoma virus-I (HTLV-I)-associated, IL-2Ra-
`expressing adult T cell leukemia (ATL) (20). The 20 patients treated
`in this study did not suffer any untoward reactions. Of the 20 treated
`patients, seven had remissions, three of these partial, one mixed, and
`three complete, lasting from 1 to more than 17 months after
`anti-Tac therapy. Unmodified anti-Tac has also been used for
`prevention of early allograft rejection episodes in patients receiving
`renal allografts. The IL-2/IL-2R system offers a variety of other
`possibilities for relatively specific immune intervention strategies
`that will be considered below.
`
`Genetically Engineered Antibodies
`Although murmne antibodies are of value in therapy of human
`diseases, their effectiveness is limited because rodent monoclonal
`antibodies have a short survival time in humans and induce an
`immune response that neutralizes their therapeutic effect. Further-
`more, the responses induced by murine antibodies are limited
`because they only weakly recruit human effector elements and are
`relatively ineffective as cytocidal agents. To circumvent these diffi-
`culties, genetically engineered antibody variants were produced that
`combine the rodent variable or hypervariable regions with the
`human constant or constant and variable framework regions (21-
`25). The ability to genetically engineer antibodies represents a
`quantum leap in immune intervention that is comparable to the
`immunological revolution initiated by the introduction ofmonoclo-
`nal antibodies.
`After the demonstration that lymphoid cells can express cloned
`transfected immunoglobulin genes, mouse-human chimeric mono-
`clonal antibodies to tumors were generated with specificities direct-
`ed toward antigens expressed by colorectal, mammary, pancreatic,
`and B cell and T cell malignancies (21, 22). Jones et al. (23) proposed
`that because V domains represent a framework of P sheets topped
`with antigen-binding loops and because 1-framework structures of
`most crystallized antibodies are nearly invariant, the specificity ofthe
`antibody combining site might be independent of the framework
`region. Thus, to further reduce the immunogenicity of rodent
`elements, Winter and colleagues generated humanized antibodies
`that retained only the antigen-binding complementarity-determin-
`ing regions (CDRs) from the parent rodent monoclonal antibody in
`association with human framework regions (23, 24). Unfortunately,
`in some cases humanized antibodies produced by this approach have
`reduced binding affinity for antigen when compared to the original
`rodent antibody. Queen and co-workers (25) addressed this prob-
`lem in two ways. First, the human framework was chosen to be as
`homologous as possible to the original mouse antibody in order to
`reduce deformation of the transplanted mouse CDRs. Second,
`computer modeling was used to identify several amino acids in the
`mouse antibody framework that, although outside the CDRs, were
`SCIENCE, VOL. 252
`
`

`

`Downloaded from
`
`http://science.sciencemag.org/
`
`
`
`on January 19, 2018
`
`likely to interact with the CDRs or antigen. These specific amino
`acids were retained in the humanized antibody.
`One of the goals in the generation of humanized antibodies is the
`reduction oftheir immunogenscity. Although humanized antibodies
`are less immunogenic than their murine counterparts (26-28), the
`idiotypic element may be immunogenic. Furthermore, the presence
`of allotypes on the human immunoglobulin G (IgG) framework
`may provide foreign carrier determinants, thereby enhancing im-
`mune responses to the idiotypic element (29). In the case of
`anti-Tac, the humanized version was dramatically less immunogenic
`than the parent murine monoclonal antibody when administered to
`cynomolgus monkeys (26). Similarly, in the first clinical trial of a
`chimeric antibody, a murine human chimeric IgGI antibody specific
`for a gastrointestinal tumor antigen elicited an antibody response
`against the chimeric antibody in only one of ten patients studied
`(27). Furthermore, no anti-monoclonal antibody response was
`detected in two patients with non-Hodgkin's lymphoma studied
`who received CAMPATH-1, an antibody recognizing an antigen
`expressed by human lymphocytes (28). The pharmacokinetics of an
`antibody molecule may also be altered genetically. The survival time
`of a monoclonal antibody can be altered to increase its period of
`effective action or, alternatively, to accelerate its clearance. For
`example, the pharmacokinetics of radiolabeled humanized anti-Tac
`differed substantially from that of murine anti-Tac when adminis-
`tered to normal cynomolgus monkeys, with a prolongation of the
`survival half-time of humanized anti-Tac to 103 hours, as compared
`to 38 hours for murine anti-Tac (26). Prolonged survival of
`humanized monoclonal antibodies in humans was observed by
`LoBuglio and co-workers (27). The catabolic rate of an immuno-
`globulin is determined by the CH2 domain of the Fc region of the
`immunoglobulin. Thus, the longer survival of humanized antibodies
`observed probably reflects replacement of the murine IgG CH2
`domain with the human IgG CH2 domain (30, 31).
`Antibodies or their fragments can also be genetically engineered
`to have more rapid clearance. This might be desirable when a
`monoclonal antibody is conjugated to a radionuclide for use in
`radioimmunoscanning. For example, antigen-binding fragment
`(Fab), F(ab')2, or single chain Fv fragments of monoclonal anti-
`bodies have survival half-lives of less than 5 hours. Rapid turnover
`can also be accomplished by the deletion of the CH2 domain as
`demonstrated for an antibody reactive with the disaloganglioside
`GD2 expressed on human tumors of neuroectodermal origin (32).
`Effector functions can be improved by introduction of human
`constant regions that impart biological activity to a murine antibody
`that lacks effector function but has the desired binding specificity. The
`human IgG subclasses differ in their antitumor activity and in their
`capacity to induce complement or antibody-dependent cell-mediated
`cytotoxicity. The IgGl subclass appears to be superior to the other
`subclasses in most functions (33). The human IgGl versions of the
`CAMPATH-1 antibody and the L6 monodonal antibody to a carci-
`noma-associated antigen were more effective in antibody-dependent
`cell-mediated cytotoxicity (ADCC) than the parent rodent antibodies
`(8, 24). Furthermore, the humanized version of anti-Tac induces
`ADCC with human mononudear cells, a function absent from the
`original mouse monoclonal antibody (34).
`
`Human Monoclonal Antibodies
`One solution to the problems of immunogenicity and poor
`recruitment of effector functions characteristic of rodent monoclon-
`als is to produce human monoclonal antibodies. Human antibodies
`of appropriate specificity and of high affinity have been difficult to
`isolate. The use of mouse myeloma cells as fusion partners for
`
`21 JUNE 1991
`
`human cells often leads to preferential loss of human chromosomes
`and instability of the hybrids. For ethical reasons one cannot
`immunize humans with certain tumor antigens. An alternative, the
`immortalization of human cells by Epstein-Barr virus, often gener-
`ates lines that produce only low amounts of IgM-type antibodies.
`Human antibodies may be produced more easily with the use of
`SCID-hu mice (35), that is, immunodeficient mice reconstituted by
`human peripheral blood or human fetal thymus, bone marrow, and
`lymph nodes. When peripheral blood is used, antibodies may be
`produced if the donor has already been primed with antigen. Such
`antibodies have not yet been used in therapeutic trials.
`An alternative approach to the production of human monoclonal
`antibodies has been reported that bypasses hybridoma technology
`(7, 36, 37). The immunoglobulin V-region genes from B cells were
`cloned with the use of the polymerase chain reaction technique. The
`antibody derivatives were then expressed in E. coli and screened for
`ability to bind antigen. Initially, heavy chain V regions were
`expressed alone or with an irrelevant light chain V region. However,
`a large combinatorial library of the immunoglobulin repertoire of
`the mouse in phage lambda has now been generated (36). Heavy
`and light chain libraries were prepared in phage lambda and used to
`generate a large array of random heavy plus light chain pairs
`expressed in bacteria in the form of Fab molecules. The screening for
`binding of antigens to hapten was rapid and permitted the analysis
`of many monospecific Fab-producing clones. In similar studies,
`Mullinax and co-workers (37) identified human antibody fragment
`clones specific for tetanus toxoid in a bacteriophage lambda immu-
`noexpression combinatorial library prepared using messenger RNA
`derived from human peripheral blood lymphocytes. The bacterio-
`phage clones are directly amenable to genetic manipulation for
`preparing complete immunoglobulins of the desired isotype. Fur-
`ther studies will be required to determine whether this approach will
`allow the efficient identification of human monoclonal antibodies of
`sufficiently high affinity for clinical studies.
`
`Biflmctional Antibodies
`Antibodies with two distinct binding activities have been gener-
`ated to deliver radionuclides, toxins, cytotoxic drugs, or host
`cytotoxic cells to specific cellular targets (38). Their predominant use
`has been to direct cytotoxic cells to target and lyse cells that they
`normally would not lyse. Such bifunctional antibodies have been
`prepared by chemical cross-linking, disulfide exchange, or the
`production of hybrid-hybridomas (quadromas). Bispecific antibod-
`ies have also been produced by introduction of two sets of immu-
`noglobulin heavy and light chains into myeloma cells or by con-
`struction of single-peptide bispecific antibodies with the use of
`peptide linkers between the variable domains of two distinct mono-
`clonal antibodies. To be effective, the bispecific antibody must
`retarget the cytotoxic cell from its natural ligand to the ligand
`identified by the monoclonal antibody. Furthermore, the antibody
`must activate the cytotoxic cells into functional effectors without the
`normal major histocompatibility complex- and antigen-specific re-
`strictions. The most effective bispecific monoclonal antibodies have
`the CD3 antigen on cytotoxic T cells or the CD16 Fc y R III
`receptor on natural killer cells as their nonantigen specificity.
`Bispecific antibodies with specificity against both tumor targets and
`CD3 or CD16 effector cells are effective in mediating the killing of
`tumor cells in vitro and in vivo. For example, murine anti-Tac does
`not participate in ADCC with human mononuclear cells. In con-
`trast, murine anti-Tac-anti-CD3 and anti-Tac-anti-CD16 bifunc-
`tional agents used in conjunction with peripheral blood mononu-
`clear cells killed targets that express IL-2 receptors. A universal
`1659
`ARTICLES
`
`

`

`Downloaded from
`
`http://science.sciencemag.org/
`
`
`
`on January 19, 2018
`
`bispecific antibody for retargeting effector cells to tumor cells can be
`generated with the use of a bispecific hybrid antibody with dual
`specificities for CD3 and for a rat immunoglobulin light chain
`allotype (39). This bispecific antibody mediates retargeting of
`effector cells to a range of tumor cells, each coated with rat
`monoclonal antibodies bound to surface antigens.
`Human T cells that had been retargeted by bifunctional antibodies
`were used to treat established human ovarian carcinoma in a nude
`mouse model (40). Peripheral blood lymphocytes from patients
`were incubated overnight with IL-2 and treated with heteroconju-
`gates containing antibody to CD3 cross-linked to an appropriate
`antibody to tumor and were injected interperitoneally into tumor-
`bearing mice. Tumor growth was inhibited. Bispecific antibodies
`have also been used in nontumor systems. For example, thrombol-
`ysis was enhanced by targeting oftissue plasminogen activator (tPA)
`by bispecific antibodies to tPA and fibrin (4). Thus, although
`problems associated with their manufacture remain to be resolved,
`bispecific antibodies show great promise as therapeutic agents.
`
`Monoclonal Antibody-Cytotoxic Agent
`Conjugates
`The limited efficacy of many unmodified monoclonal antibodies
`led to an alternative approach, the use of these agents as carriers of
`cytotoxic substances. An array of toxins of bacterial and plant origin
`have been coupled to monoclonal antibodies for production of
`immunotoxins (8, 41). The strategy is to select from nature a toxic
`protein and then to modify the toxin so that it will no longer
`indiscriminately bind and kill normal cells but will instead kill only
`the cells expressing the antigen identified by the monoclonal anti-
`body. The majority of toxins targeted to cell surfaces by immuno-
`conjugates act in the cytoplasm, where they inhibit protein synthe-
`sis. After binding to cell surface antigens, immunotoxins are taken
`up by endocytosis and delivered to endosomes. Fragments of some
`toxins (for example, diphtheria toxin) are then translocated across
`the membrane of this organelle. Other immunotoxins (for example,
`ricin) are routed further to the trans-Golgi network, where a
`minority undergo translocation to the cytoplasm. Unfortunately,
`most are routed to lysosomes, where they are degraded. In the
`cytoplasm, the toxins used clinically act either to adenosine diphos-
`phate (ADP)-ribosylate elongation factor 2 (for example, Pseudo-
`monas exotoxin) or to inactivate the 60S ribosomal subunit so that
`it has a decreased capacity to bind elongation factor 2 (for example,
`ricin). Less than ten toxin molecules in the cytoplasm are sufficient
`to kill the cell; however, more must bind to the cell surface to
`compensate for the inefficiencies in internalization and translocation.
`Although immunotoxins are simple in concept, the first-genera-
`tion immunotoxins were relatively ineffective. Several requirements
`must be fulfilled for an immunotoxin to be effective (41). In
`particular, (i) the immunoconjugate should be specific and should
`not react with normal tissues. Binding to tissues that do not express
`antigen can be reduced by removal of the nonspecific natural
`cell-binding subunits or domains of the toxin. Furthermore, be-
`cause plant glycoprotein toxins contain mannose oligosaccharides
`that bind to cells of the reticuloendothelial system and, in some
`cases, also contain fucose residues that are recognized by the
`receptors on hepatocytes, deglycosylation of plant toxins may be
`required to avoid rapid clearance and potential cytotoxic effects on
`these cells. (ii) The linkage of the toxin to the antibody should not
`impair the capacity of the antibody to bind antigen. (iii) The
`immunotoxin must be internalized into endosomic vesicles. Thus,
`toxins directed by monoclonal antibodies to surface receptors that
`are normally internalized may be more active than those directed
`
`1660
`
`toward noninternalizing cell surface molecules. (iv) The active
`component of the toxin must translocate into the cytoplasm. These
`various goals can be in conflict; thus, the removal of the B chain of
`ricin reduces nonspecific binding but also reduces the capacity of the
`residual A-chain monoclonal antibody to translocate across the
`endosomic vesicle membrane. (v) For in vivo therapy, the linkage
`must be sufficiently stable to remain intact while the immunotoxin
`passes through the tissues of the patient to its cellular site of action.
`The first generation of heterobifunctional cross-linkers used to bind
`the toxin to the monoclonal antibody generated disulfide bonds that
`were unstable in vivo. This problem was solved in part by the
`synthesis of more stable cross-linkers, which used phenyl or methyl
`groups, or both, adjacent to the disulfide bond to restrict access to
`the bond.
`The development by Pastan and co-workers (41) of IL-2R-
`directed Pseudomonas exotoxin (PE) conjugates for the treatment of
`IL-2R-expressing ATL demonstrates recent progress in the devel-
`opment of effective immunotoxins. PE, chemically coupled to
`anti-Tac, showed specificity in vitro (42). However, only a few
`milligrams of this agent could be given to patients without produc-
`ing liver damage because the toxin had not been sufficiently changed
`so that it would no longer bind to normal liver cells. Functional
`analysis of deletion mutants of the 66-ld PE (43) showed that
`Domain III was responsible for ADP-ribosylation of elongation
`factor 2; Domain II helped in translocation of the toxin to the
`cytosol, whereas Domain I was responsible for unwanted ubiquitous
`cell binding. A PE molecule from which the 26-d) Domain I had
`been deleted (PE40) had full ADP-ribosylating activity but extreme-
`ly low cell-killing activity when used alone. PE40 conjugated to
`anti-Tac inhibited protein synthesis in T cell lines expressing Tac but
`not in lines not expressing the IL-2R. However, immunotoxins
`made by chemical attachment of this truncated PE to anti-Tac
`yielded a product that was heterogeneous. Active single-chain Fv
`fragments of antibodies have been produced in E. coli by linking of
`the light and heavy chain variable domains with a peptide linker
`(44). Chaudhary and co-workers (45) used this genetic engineering
`approach to produce a single-chain antibody toxin fusion protein
`[anti-Tac (Fv)-PE40] in which the variable regions of anti-Tac were
`joined in peptide linkage to PE40 to generate an immunotoxin that
`was cytotoxic to human cell lines bearing IL-2R and to freshly
`obtained ATL cells but not to receptor-negative cells.
`The antitumor activity of immunotoxins has been evaluated in
`animal models since their introduction (46). Treatment usually
`delayed the appearance of tumors and prolonged the lifespan of the
`animals. In a few cases, there was complete regression of the tumor.
`Several in vivo clinical trials in humans have involved the paren-
`teral administration of immunotoxins. In certain cases, for example,
`with monoclonal antibody-toxin conjugates directed toward breast
`or ovarian cancers, severe neurological toxicity was observed because
`of an unanticipated crossreactivity of the monoclonal antibody with
`an antigen in the central nervous system (47). The most common
`toxicity observed was capillary leakage that resulted in hypoalbu-
`minemia, edema, fatigue, and myalgia.
`The results of in vivo clinical trials in patients with cancer with
`first-generation immunotoxins did not fulfill the hopes engendered
`by in vitro and animal model studies. There were only two complete
`remissions and eight partial remissions among the 127 patients
`treated in ten clinical trials with toxin-conjugated monoclonal
`antibodies directed toward ovarian, breast, colorectal, and lymphoid
`neoplastic cells. More encouraging results were obtained when
`benign diseases were treated with immunotoxins or when modified
`immunotoxins were used in therapy of patients with cancer. For
`example, 9 of 22 patients manifested a mixed or partial response,
`and one had a complete remission after treatment of a refractory B
`SCIENCE, VOL. 252
`
`

`

`Downloaded from
`
`http://science.sciencemag.org/
`
`
`
`on January 19, 2018
`
`cell leukemia-lymphoma with a monoclonal antibody to CD19
`conjugated to a modified ricin toxin that had galactose binding sites
`blocked sterically (48). Furthermore, 22 of 32 evaluable patients
`with GVHD had a favorable early response in at least one organ
`after therapy with monoclonal antibody to CD5 conjugated to ricin
`A (49).
`An alternative approach for the delivery of cytotoxic agents to
`cancer cells involves the use of monoclonal antibodies as carriers for
`enzymes to tumor cell surfaces (50). The enzymes are chosen for
`their ability to convert drug precursors injected parenterally into
`active antineoplastic drugs. The active cytotoxic agents formed can
`then penetrate nearby tumor cells and cause the death of these cells.
`A number of prodrugs (drugs in an inactive form that can be
`transformed at the tumor into active anticancer drugs by antibody-
`enzyme conjugates) have been developed. The antibody-enzymes
`conjugates were shown to localize to tumors through the activity of
`the monoclonal antibodies that bind to tumor-associated antigens.
`In vivo studies showed that prodrug administration after monoclo-
`nal antibody-enzyme infusion can result in antitumor activities
`significantly greater than the activities of the prodrugs, drugs, or
`monoclonal antibodies given alone.
`
`Radiolabeled Monoclonal Antibodies
`Toxin conjugates do not pass easily from the endosome to the
`cytosol. Furthermore, the toxins are immunogenic and thus provide
`only a short therapeutic window before the development of anti-
`bodies directed toward the toxin. Radiolabeled monoclonal anti-
`bodies have been developed as alternative immunoconjugates for
`delivery of a cytotoxic effector to target cells and for radioimaging
`(8, 51). Radioimmunodetection with the use of radiolabeled mono-
`clonal antibodies, most often with monoclonal antibodies to carci-
`noembryonic antigen, is widely used to complement other ap-
`proaches for tumor detection. Although intact IgG antibodies are
`retained better by tumors and thus appear to be better for therapy,
`F(ab')2 and Fab fragments are preferred for imaging because both
`targeting and blood clearance are more rapid, which reduces the
`background. Tumors as small as 0.5 cm, which are sometimes
`missed by other radiological methods, can be imaged with antibod-
`ies or antibody fragments labeled with suitable radionuclides.
`One advantage in the use of radiolabeled monoclonal antibody
`conjuga

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