throbber
Immunology 1988 65 329-335
`
`REVIEW PAPER
`
`Therapeutic strategies with monoclonal antibodies and
`immunoconjugates
`
`V. S. BYERS*t & R. W. BALDWIN* * Cancer Research Campaign Laboratories, University of Nottingham, Nottingham,
`U.K. and t Xoma Corporation, Berkeley, California, U.S.A.
`
`Acceptedfor publication I August 1988
`
`CONTENTS
`
`Therapeutic use of mAbs
`Immunotoxins
`Chemo-immunoconjugates
`Clinical trials
`Human anti-murine monoclonal antibody responses (HAMA)
`Clinical impact
`Approaches to abrogate anti-mouse immunoglobulin responses in patients
`Conclusions
`
`329
`330
`330
`331
`332
`332
`332
`333
`
`Introduction
`Monoclonal antibodies (mAbs) are emerging as a major
`modality for both detection and therapy ofvarious pathological
`conditions, including malignant disease. Murine monoclonal
`antibodies have been developed which react with tumour-
`associated antigens expressed on or near the cell surface, and
`present in high concentration on malignant tissues but in
`minimal amounts on normal tissues (Boyer et al., 1988). These
`include mAbs against colon cancer, ovarian cancer, breast
`cancer and neurological tumours, which have been used thera-
`peutically either alone or conjugated to cytotoxic agents
`(Baldwin & Byers, 1987) and diagnostically coupled to radio-
`isotopes for gamma camera imaging of patients (Chatal, 1988,
`Pimm, 1987). More recently investigators have taken advantage
`ofthe well-defined antigens on the surface oflymphoid cells, and
`mAbs directed against such antigens have been used therapeuti-
`cally against lymphoid malignancies and autoimmune diseases,
`including prevention of renal allograft rejection and treatment
`of graft-versus-host (GVH) disease (Goldstein et al., 1985;
`Byers, 1987; Byers et al., 1987a; Fahey et al., 1987; Youle &
`Colombatti, 1986).
`In spite of the encouraging clinical results, including the use
`of the anti-T-cell receptor mAb OKT3 in renal allograft
`rejection (Goldstein et al., 1985; Mayes et al., 1988), the central
`limitation in using these murine products has been the gene-
`ration of an immune response to murine immunoglobulin which
`usually precludes retreatment. Especially in solid tumour
`therapy, retreatment is viewed as a key factor in allowing this
`therapeutic modality to be optimized, and a variety of
`approaches to solve the problem are underway.
`These issues are reviewed in relation to the now quite
`extensive advances in designing immunoconjugates for therapy,
`particularly in malignant disease.
`Correspondence: Dr V. S. Byers, Cancer Research Campaign
`Laboratories, University of Nottingham, Nottingham NG7 2RD, U.K.
`
`Therapeutic use of mAbs
`Murine monoclonal antibodies that recognize tumour-asso-
`ciated antigens and localize in tumours, or that have well-
`defined reactivity against antigens on lymphocytes, are being
`evaluated for therapy (Baldwin & Byers, 1987). Unmodified
`antibodies have demonstrated efficacy in some cases. For
`example, in renal allograft transplantation OKT3 antibody
`directed against the T-cell receptor has been proven quite
`effective in reversing rejection, probably as a result of blocking
`lymphocyte-antigen interactions (Goldstein et al., 1985). Also,
`unconjugated antibodies are being used in cancers such as colon
`cancer (LoBuglio et al., 1986; Sears et al., 1985) and neuroblas-
`toma (Cheung et al., 1987). The rationale for their use is that
`host defence mechanisms such as antibody-dependent cell
`cytotoxicity (ADCC) can serve as the cytotoxic modality, but
`consideration is now being given to the view that murine
`monoclonal antibodies may be inducing anti-idiotypic anti-
`bodies that will react against the tumour (Viale et al., 1987;
`Nepom & Hellstrom, 1987; Herlyn et al., 1987a, b). These
`approaches have shown some encouraging results-for example
`in the case of antibodies against neuroblastoma-but most
`attention has turned to the use of mAbs for targeted therapy
`using cytotoxic agents, including radioisotopes and toxins.
`The initial selection ofantibody for use as a targeting agent is
`primarily based upon its reactivity with appropriate target cells
`in comparison with binding to normal cells, since this latter
`reactivity will be translated in vivo into 'side-effects'. This is still
`a major limitation, although there are only a few antibodies
`which meet specificity requirements. For example, with solid
`tumours, preclinical studies indicate that antibodies which react
`with malignant melanoma (Spitler et al., 1987) and colorectal
`cancer (Baldwin & Byers, 1988; Byers et al., 1988a, b) had
`minimal normal tissue reactivity, and immunotoxins made from
`these monoclonal antibodies were well tolerated in clinical
`studies. However, other potentially useful antibodies, such as
`
`329
`
`IMMUNOGEN 2137, pg. 1
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`330
`
`V. S. Byers & R. W. Baldwin
`
`those binding to epidermal growth factor receptor (Griffin et al.,
`1987; FitzGerald et al., 1987) and to breast/ovarian cancer-
`associated mucins (Price, 1988a, b), exhibit quite widespread
`normal tissue reactivity. Immunotoxins constructed with these
`antibodies may exhibit normal tissue toxicity, thus limiting their
`clinical use.
`The mechanism of action of the different cytotoxic agents
`influences to some extent the choice of antibody. With radio-
`isotopes, the monoclonal antibody must be targeted to the
`tumour, but internalization into the cell may be detrimental
`since it can result in deconjugation of the isotopes from the
`monoclonal antibody, and exocytosis, facilitating its removal
`from the tumour (Press et al., 1988). On the other hand, most
`cytotoxic drugs, e.g. methotrexate, are bound to the antibody by
`labile bonds and endocytosis into an acidic intracellular com-
`partment maximizes activity through drug release (Garnett et
`al., 1985; Embleton & Garnett, 1986). Antibody targeting with
`these agents is intended primarily to reduce uptake by normal
`tissues, so lowering toxicity, although it may convert resistant
`cells to susceptible cells in cases where resistance is due to
`defective drug uptake. In contrast, ricin toxin A chain (RTA) is
`inactive extracellularly and non-toxic as the free agent. Anti-
`body conjugation targets the toxin moiety to the target cell,
`allowing it to be internalized by endocytosis (Baldwin et al.,
`1988; Byers et al., 1988a).
`
`Immunotoxins
`Toxins of plant and bacterial origin represent a class of highly
`cytotoxic agents for linking to monoclonal antibodies to form
`immunotoxins (Vitetta et al., 1987; Baldwin & Byers, 1987;
`Blakey & Thorpe, 1987). One type of toxin, typified by ricin
`produced from beans of the plant Ricinus Communis, is
`composed of two disulphide-linked subunits. The intact toxin
`binds to mammalian cells through galactose residues on the B
`subunit and is endocytosed (Van Deurs et al., 1988). The A
`chain, which is inactive extracellularly, after introduction into
`the cell catalytically inactivates the 60s ribosomal subunit of
`eukaryotic cells by modifying one or two nucleoside residues of
`the 28s ribosomal RNA (Endo et al., 1987). If the bond between
`the A and B chains is broken and an antibody substituted for the
`B chain, the A chain can now be directed to specific target cells.
`This approach has now been used for several A-B chain toxins,
`including pseudomonas exotoxin, diphtheria toxin and abrin
`(Vitetta et al., 1987; Baldwin & Byers, 1987).
`Much work has been done with RTA immunotoxins
`containing ricin A chain conjugated to antibody through
`the cross-linker N-succinimidyl-3-(2-pyridyldithio) propionate
`(SPDP). This contains a disulphide bond, and it is generally
`assumed that this is cleaved intracellularly to allow the A chain
`to translocate through the cytoplasm to the Golgi apparatus.
`The pharmacokinetics of the clearance of the antibody moiety
`of the immunotoxins is markedly changed by the addition of the
`RTA (Byers et al., 1987b; Bourrie et al., 1986; Blakey et al.,
`1987) since this protein contains mannose-terminating oligosac-
`charides that are recognized by reticuloendothelial cells, includ-
`ing Kupffer cells in the liver (Blakey et al., 1987). This results in
`rapid liver clearance and reduces the blood half-life from days to
`hours. Increasing the blood half-life improves localization of
`immunotoxin in the tumours, so various strategies are being
`devised to eliminate the mannose residues on the RTA without
`affecting activity. These include deglycosylation of RTA
`
`(Blakey et al., 1988), synthesis of recombinant RTA, and use of
`a subfraction of RTA with reduced oligosaccharide content
`(Fulton et al., 1986). The increased circulation time increases
`localization at the disease site; this must be balanced against the
`possibility of increased or altered toxicity.
`Endocytosis and intracellular processing are essential for
`immunotoxins to exert their effect (Vitetta et al., 1987; Baldwin
`et al., 1988). Since the mechanism of intracellular trafficking is
`poorly understood, immunotoxins are selected by screening
`them for specific cytotoxicity. This is usually expressed as the
`concentration of immunotoxin necessary to inhibit 50% of
`target cell growth in vitro (IC50) and, in most cases, the difference
`in potency between immunotoxin and free RTA is in the order
`of 103-104-fold. One immunotoxin against the CD22 antigen on
`B lymphomas actually has a lower ICo than whole ricin (May et
`al., 1986).
`As additional information of the mechanism of internaliza-
`tion and intracellular trafficking of immunotoxins is obtained,
`construction of potent immunotoxins should become more
`efficient. Both the rate of endocytosis and the intracellular
`compartment into which the immunotoxin is moved depends in
`some part on the antigen to which the monoclonal antibody
`binds. Receptor-mediated endocytosis is more efficient than
`pinocytosis, for example the rate of endocytosis of a gelonin-
`containing immunotoxin which reacts with the C3b receptor on
`human B lymphocytes can be markedly increased by infection
`with Epstein-Barr virus, which is the preferred ligand for the
`receptor (Tedder et al., 1986). Also, recent studies by Sandvig et
`al. (1987) demonstrated that under conditions in which recep-
`tor-mediated endocytosis through coated pits is blocked, inter-
`nalization of transferrin and epidermal growth factor does not
`occur, but internalization and intoxication by ricin continues
`essentially unhindered. These and other data have been taken to
`suggest that at least two independent mechanisms exist for the
`internalization of surface-bound ligands, one via coated pits,
`and the other via 'smooth' pits which lack a clathrin coat. This
`may influence the cellular compartment into which the immuno-
`toxin is directed. After endocytosis, the endocytotic vacuole is
`acidified and, in contrast to chemoimmunoconjugates and
`immunotoxins such as diphtheria toxin, RTA immunotoxins
`are inactivated by acidic environments. Thus for these conju-
`gates, any inhibition of intracellular acidification increases
`activity. The potent potentiating agents for many immunotox-
`ins, monensin and ammonium chloride, are postulated to work
`by decreasing the rate of intracellular acidification, thereby
`prolonging intracellular activity of the immunotoxins.
`
`Chemo-immunoconjugates
`The design of chemo-immunoconjugates is developing along
`similar lines to those established with immunotoxins, and
`conjugates have been produced with several classes ofcytotoxic
`drugs, including alkylating agents and antimetabolites (Bald-
`win, 1985; Baldwin et al., 1986; Ghose & Blair, 1987). These are
`produced by conjugating drug to antibody through a specific
`functional group such as amino, hydroxyl or sulphydryl
`residues. In this case, the selected conjugation site should not be
`required for drug action or, if so, should become available
`following intracellular release of the drug. Cytotoxic drugs are
`generally less active than toxins and it is necessary to introduce
`the maximum number of drug residues compatible with reten-
`tion of antibody reactivity. High drug substitution ratios have
`
`IMMUNOGEN 2137, pg. 2
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`Therapeutic strategies
`
`331
`
`been reported, but with monoclonal antibodies of the IgG
`subclass, substitution of more than 10 drug residues per
`antibody molecule may produce an unacceptable level of
`antibody damage. Consequently drug carrier molecules such as
`human serum albumin and dextrans are being used to yield
`antibody conjugates with much higher molar ratios of drug to
`antibody (Garnett & Baldwin, 1986; Endo et al.,
`1988).
`Conjugates of large molecular size may have inappropriate
`pharmacokinetic properties and are less able to penetrate
`tumour tissue. Currently, therefore, attention is being given to
`the chemistry of drug-antibody conjugation so as to improve
`the potency of chemo-immunoconjugates. This includes the
`selection of more cytotoxic analogues of the drugs used for
`antibody conjugation, and design of new drug carrier molecules
`to replace currently used polymers, as well as modification ofthe
`drug conjugates, particularly the overall charge in relation to
`their pharmacokinetics.
`The use of hybrid antibodies is being considered as an
`alternative to chemical conjugation for in vivo targeting drugs to
`tumours. Hybrid-hybrid monoclonal antibodies recognizing
`both a tumour-associated antigen and the drugs are being
`produced either by fusion of two existing hybridomas or by
`fusion of one existing hybridoma with spleen cells from donor
`mice immunized against the other antigen. Studies in this field
`have been reported by Corvalan et al. (1987a, b) in which a
`hybrid antibody reactive with a carcinoembryonic antigen
`(CEA) epitope and vinca alkaloid was produced by fusion of an
`anti-CEA antibody-producing hybridoma with spleen cells
`from mice immunized with vindesine-protein conjugate. The
`IgG antibody had the yl heavy chain from the parental anti-
`CEA antibody and a y2a from the anti-vinca alkaloid donor
`lymphocyte. This antibody localized in an appropriate CEA
`producing colon carcinoma xenograft. The antibody also
`induced profound changes in the biodistribution of 3H-labelled
`vinblastine, resulting in specific localization of the drug to
`tumour tissue and so producing a therapeutic response.
`It is feasible to prepare monoclonal antibodies to other
`drugs and therefore there is the possibility of constructing a
`range of hybrid antibodies. For example, Pimm et al., (1987)
`with a hybridoma producing monoclonal antibody to metho-
`trexate showed that complexing antibody with drug profoundly
`altered the biodistribution of the drug, prolonging blood
`survival and reducing liver uptake, and clearly hybridomas such
`as this are candidates for constructing hybrid-hybrid antibodies
`with anti-tumour antibodies.
`
`Clinical trials
`Clinical trials are in progress with murine monoclonal anti-
`bodies alone and as immunoconjugates. Antibodies specific for
`the idiotype of the surface immunoglobulin of B-cell lympho-
`mas have been used successfully in inducing tumour regression,
`but the clinical outcome has been widely variable ranging from
`prolonged remission to no effect (Lowder et al., 1987). Factors
`involved include antigen modulation, which may be of particu-
`lar concern with lymphoid tumours, and generation of anti-
`mouse antibody responses which markedly alter antibody
`pharmacokinetics. Another approach with B-cell tumours
`involves treatment with murine monoclonal antibodies directed
`against normal CD20 antigens. Treatment of four patients with
`refractory B-cell lymphomas with an anti-CD20 antibody
`
`produced a 90% elimination ofmalignant cells from the blood in
`two patients and a reduction in lymph node disease in one
`patient, but the duration of remission was shortlasting (Press et
`al., 1987). Antibodies specific for tumour-associated ganglio-
`sides which activate human complement and are active in
`ADCC are in clinical trials with neuroblastoma (Cheung et al.,
`1987). The antigen recognized by this antibody is also expressed
`on neurons and peripheral brain fibres. Because of pain during
`infusion, patients must be anaesthetized and maintained on
`analgesics for several weeks afterwards. However, complete
`remission of two neuroblastomas have been reported, suggest-
`ing that, with appropriate medical support, substantial reacti-
`vity with normal tissues can be tolerated.
`The most successful therapeutic use of an unconjugated
`antibody has been the use of OKT3 in renal allograft rejection
`(Goldstein et al., 1985). This murine monoclonal antibody is
`directed against the CD3 (T-cell receptor) antigen on mature
`human T cells and blocks their function. When given over a 14-
`day period as initial therapy in patients experiencing acute
`rejection, it will reverse 94% of the rejections, and significantly
`improve I year graft survival to 62%. This is now an approved
`drug for that indication.
`Clinical evaluation of radioisotope-labelled monoclonal
`antibodies is in progress, using 131-iodine (gamma and B
`emission), 90-yttrium (gamma emission) and 21 1-astadine (a-
`emission) (Humm, 1986; Order et al., 1988; Sands, 1988). It has
`been argued that the minimum requirement for effective therapy
`using radioimmunoconjugates is accumulation of 10 times more
`conjugate in tumour compared with blood (Vaughan et al.,
`1987) and this cannot be attained by intravenous administra-
`tion. Therefore, recent clinical applications have focused upon
`intraperitoneal injection of 131 -iodine-labelled antibody in
`patients with ovarian and colon cancer and intrathecal injection
`in patients with malignant melanoma. In one trial in ovarian
`cancer (Stewart et al., 1988) patients were treated with a range of
`antibodies. There were complete responses in 3/6 patients with
`microscopic disease and partial response in 2/15 patients with
`nodules smaller than 2 cm.
`The most impressive response to immunotoxin therapy so
`far has been in the treatment of acute graft versus host (GVH)
`disease developing following bone marrow transplantation
`(Kernan et al., 1988; Byers et al., 1987b). The immunotoxin used
`contains RTA conjugated to a monoclonal antibody reacting
`with the CD5 antigen on mature T lymphocytes. In 12/25
`evaluable patients, progression of disease was reversed after the
`first seven doses of immunotoxin; in another five it was
`stabilized. Overall these patients continued to attain complete
`responses as late as 60 days after therapy, whereas most of the
`non-responders had died by Day 40. Responses were seen in all
`three involved organs, skin, gut and liver. Pharmacokinetic
`studies indicated that the immunotoxin was cleared rapidly
`from blood. Even so, the circulating T lymphocytes, as mea-
`sured by CD3 and CD5 expression, dropped by Day 6 of
`treatment to less than 20% of the initial value and remained low
`for several weeks to months thereafter.
`Clinical trials with ricin A chain immunotoxins also include
`phase I/II trials in malignant melanoma with antibody directed
`against the high molecular weight (> 200,000) melanoma
`antigen (Spitler et al., 1987) and a phase I trial in colorectal
`carcinoma with monoclonal antibody 791T/36 which reacts
`with a 72,000 molecular weight tumour membrane glycoprotein
`
`IMMUNOGEN 2137, pg. 3
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`332
`
`V. S. Byers & R. W. Baldwin
`
`(gp72) (Byers et al., 1988b). These immunotoxins are well
`tolerated in terms of side-effects. These include reversible
`hypoalbuminemia, and myalgias, which are generic for the
`immunotoxins, and proteinuria which is associated with the
`immunotoxin constructed with monoclonal antibody 791T/36.
`In the melanoma study, approximately 50% of the patients
`showed some degree of response, with one patient having
`complete remission. In the colorectal cancer study, three
`patients with hepatic metastases had resolution of the smaller
`lesions. Although encouraging, most of the clinical responses
`have not yet reached the requirement for categorization as
`response by the WHO criteria. For this to be achieved it is likely
`that retreatment schedules will be necessary and for this the
`development of anti-mouse antibody responses in immunotoxin
`patients must be controlled.
`
`Human anti-murine monoclonal antibody responses (HAMA)
`Clinical impact
`Almost all of the murine monoclonal antibodies being used
`clinically provoke antibody responses (HAMA) in patients.
`These include antibody responses against the framework, the
`isotype, and the idiotype of the murine IgG antibodies. It is rare
`that anaphylaxis or serum sickness occurs in patients with
`HAMA when retreated with mAb, although anaphylactoid
`reactions are not uncommon and primarily consist ofperiorbital
`oedema or urticaria. But HAMA responses lead to altered
`pharmacokinetics of the injected monoclonal antibody. Anti-
`melanoma immunotoxin is rapidly cleared from serum with
`reduced drug levels being attained (LoBuglio et al., 1988a).
`Immunoscintigraphy of tumours with radiolabelled antibody is
`also seriously compromised in patients developing HAMA, and
`has resulted in negative imaging of tumour with radiolabelled
`product (immune complexes) being localized in liver and spleen
`(Pimm et al., 1985; Perkins, Pimm & Powell, 1988). The extent to
`which HAMA influences therapy clinically is still unclear. One
`study of patients treated with OKT3 (Jaffres et al., 1986)
`reported that of 21 patients given OKT3 60% made anti-Id
`antibodies, which in some cases interfered with its therapeutic
`effectiveness, while another report indicated that low levels of
`blocking antibodies did not significantly compromise access to
`OKT3 for treatment of subsequent rejection episodes (Mayes et
`al., 1988). Possibly the more important finding is the fact that
`even though these patients were immunosuppressed with azath-
`ioprine, prednisone, and in some cases cyclosporine, they still
`were able to generate HAMA with anti-Ids as a prominent
`feature of the response, even though this must constitute a
`relatively minor portion of the murine monoclonal antibody.
`The actual generation of HAMA will probably depend in some
`part on the underlying disease as well as the concomitant
`immunosuppression; for example patients with cutaneous T-cell
`lymphoma have a higher incidence of HAMA to the anti-CD5
`mAb T1O1 than do patients with CLL, where almost no patients
`produce an immune response (Dillman et al., 1984). Also,
`patients receiving an immunotoxin composed of an anti-CD5
`mAb coupled to RTA, who are suffering from GVH disease and
`therefore are immunosuppressed both exogenously and endoge-
`nously, have very little HAMA (V. S. Byers, R. P. Mischak, N.
`Kernan and P. Scannon, unpublished findings). In some
`situations, a single course of mAb therapy may be sufficient to
`cure the disease, analagous to the experience with anti-thymo-
`
`cyte globulin in which 4-10 days of therapy is sufficent to cure
`aplastic anaemia (Young et al., 1988). Overall, however, most
`investigators take the position that if multiple courses of mAb
`are to be used therapeutically, reliable methods for abrogating
`the immune response must be devised.
`
`Approaches to abrogate anti-mouse immunoglobulin responses in
`patients
`Concomitant treatment with immunosuppressive medication is
`one method being developed, and in one study the primary
`HAMA response was blunted following pretreatment with
`azathioprine/prednisone for 8 weeks (LoBuglio et al., 1988). A
`significant inhibition of the primary HAMA response has been
`achieved with cyclosporin (CsA) pretreatment for 6 days,
`although CsA had no effect on abrogation of response in
`patients with pre-existing anti-murine antibodies (Lederman et
`al., 1988). The problems with such regimens is non-specific
`immunosuppression, and toxicity of the additional drugs which
`makes such therapy with mAbs more hazardous than with mAb
`given as a single agent. An alternative approach is to produce a
`human antibody or 'humanized' version of murine monoclonal
`antibodies.
`Initially, intensive efforts were directed toward making
`human mAbs, but products generated have been unsatisfactory
`(Campbell et al., 1987; James & Bell, 1987). Lymphocytes from
`human peripheral blood or from draining nodes from human
`tumours were fused with either human or murine myeloma
`partners, and the mAbs generated were of the IgM subclass and
`usually directed against intracellular antigens. IgM antibodies
`do not function well as imaging agents, probably because they
`are too large to marginate from the peripheral blood into the
`tissues, and would also be expected to be poor therapeutic
`agents for that same reason. The problem in finding mAbs
`directed against human cell membrane antigens may relate to
`humans being tolerant to such antigens, but the reason for the
`difficulty in producing human mAbs of the IgG subclass is still
`not understood. Meanwhile, however, methods are being
`devised by which the variable region of the murine mAb may be
`engineered onto a human constant region. A chimeric antibody
`composed of the variable regions of murine monoclonal
`antibody 17.1A (IgG2aK), which recognizes a glycoprotein
`present on human colorectal cells, has been coupled with the
`constant region of human IgG3. This chimeric 17.1A mAb has
`the same reactivity against colon tumour cells as the native
`antibody, and both have identical capacity to inhibit radiola-
`belled native 17. 1A binding to tumour cells (Shaw et al., 1987).
`Native 17.1A has been used extensively in clinical trials in
`colorectal cancer patients, and it elicits a very pronounced
`HAMA response, altering its pharmacokinetics (LoBuglio et
`al., 1988b; Khazael et al., 1988). In contrast, antibody responses
`have not been detected in patients receiving multiple treatments
`with the chimeric 17.1A, and the pharmacokinetics of the
`chimeric 17.1A was not modified, as additional evidence of lack
`of an antibody response (Shaw, Khazaeli & LoBuglio, 1988).
`This result was unexpected since this construct still has the
`whole of the variable region as murine derived, and since the
`idiotypic component of the immune response forms a prominent
`proportion of the HAMA response. Several groups are, there-
`fore, in the process of 'humanizing' murine mAbs, using various
`techniques such as the chimeric antibody constructed from rat
`antibody CAMPATH I by introducing only the six hypervari-
`
`IMMUNOGEN 2137, pg. 4
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`Therapeutic strategies
`
`333
`
`able regions from the heavy- and light-chain domains into a
`human IgGl molecule (Riechmann et al., 1988). This chimera
`was as effective as the native mAb in complement-mediated lysis
`of B-cell lymphocytic leukaemia cells.
`Control of the immune response to immunoconjugates will
`probably be more difficult, especially where the conjugated
`moiety itself is immunogenic, e.g. ricin A chain. The immuno-
`conjugates themselves should theoretically exert a cytotoxic
`effect not only against their target cell, but also against the
`antigen-specific cells of the immune system that are capable of
`producing antibody responses against them. Thus antigen-
`specific B lymphocytes having clonally distributed specific
`surface immunoglobulin receptors should react with immuno-
`conjugates. Such recognition should lead to endocytosis of the
`cytotoxic conjugate and, following intracellular release, the
`cytotoxic moiety should cause cell death. The HAMA seen with
`certain immunoconjugates, such as RTA-containing immuno-
`toxins, indicates that for various reasons they fail to adequately
`exert a cytotoxic effect against B lymphocytes. Immunoconju-
`gates have been constructed, however, which suppress anti-
`mouse antibody responses. Conjugates constructed with mono-
`clonal antibody 791T/36 and cis-aconityl-substituted daunomy-
`cin render rats immunologically unresponsive to subsequent
`repeated immunization with the unconjugated antibody (Dur-
`rant et al., 1988). However, rats still produced HAMA in
`response to challenge with the RTA immunotoxin made from
`791T/36 antibody. This probably means that RTA itself is
`immunogenic and serves as an effective antigen-presenting
`molecule, possibly through its avid uptake by macrophages.
`Supporting this view is the finding that murine monoclonal
`antibody 791 T/36 coupled to ricin A chain produces strong anti-
`idiotypic antibody responses in mice (Pimm, Durrant & Bald-
`win, 1988). Further manipulation may be necessary, therefore,
`to tolerize the host against the cytotoxic moieties such as ricin A
`chain prior to therapy with immunoconjugates constructed with
`humanized antibodies. Benjamin et al. (1988) showed that mice
`made tolerant to rat immunoglobulin constant region determi-
`nants were, in some instances, capable ofinducing only minimal
`reactivity to the idiotypic region of the antibody and the same
`approach may be used to suppress specific responses to
`immunoconjugates. If, however, there is a non-specific enhance-
`ment of antigen processing by components attached to huma-
`nized murine monoclonal antibodies, the pathway would be to
`generate cytotoxic analogues to eliminate this component. For
`the moment, therefore, investigators using immunoconjugates
`continue to utilize concomitant therapy with immunosuppres-
`sive agents such as CsA.
`
`Conclusions
`One should not underestimate the complexities of the problems
`still to be resolved in producing monoclonal antibodies and
`immunoconjugates for clinical use, especially in solid tumours.
`Outstanding problems include improving the design of the
`immunoconjugates themselves, particularly refining techniques
`for directing the conjugation site away from the active site of the
`antibody and conjugating adequate amounts of cytotoxic
`agents. More effective conjugation procedures are also impor-
`tant, particularly with small hydrophobic molecules. Second
`generation immunoconjugates, one anticipates, will be pro-
`duced with even better therapeutic efficacy.
`
`Enhanced target tissue accessibility is very important with
`solid tumours, since these are notorious for poor vascularity.
`Longer serum half-life will increase tumour localization, but it
`probably will be necessary to develop agents with a direct effect
`on tumour vascularity for adequate penetration. Antigenic
`heterogeneity is also an issue that is being addressed by use of
`cocktails of monoclonal antibodies, and it is still under discus-
`sion as to whether the cytotoxic molecules bound to the different
`monoclonal antibodies should also be different. More impor-
`tant is the issue ofantibody responses to the immunoconjugates.
`If these agents are viewed by the same criteria as conventional
`cytotoxic agents, it will be critical to retreat patients with more
`than one course of immunoconjugate, and possibly with
`immunoconjugates of different specificities. The antibody re-
`sponse seen with almost all monoclonal antibodies and immu-
`noconjugates at present effectively restricts treatment to a single
`short cycle. Several strategies have been identified to overcome
`this, including the design of human or humanized monoclonal
`antibodies. Whether or not this will restrict the generation of
`anti-idiotypic antibodies is not yet known, although studies with
`monoclonal antibody 17-1A suggest that in some cases this may
`be achieved. Even so it seems that immunoconjugates may still
`produce anti-idiotypic antibodies, and so in these cases immu-
`nosuppressive procedures may be needed. This may be achieved
`with immunosuppressive drug treatment, but with the asso-
`ciated toxicities, particularly in treatment with cytotoxic immu-
`noconjugates as envisaged in cancer patients, this is not
`desirable. An alternative approach is to devise methods for
`specific abrogation ofantibody responses along the lines already
`reported with daunomycin-antibody conjugates or by some
`other form of immune intervention.
`Considerable advances are being made in the clinical use of
`monoclonal antibodies and immunoconjugates. These include
`immunoscintigraphy with radioisotope-labelled antibodies for
`tumour detection, and monoclonal antibodies can be viewed as
`a new class of immunosuppressive agents in transplantation.
`Promising results are also being obtained in solid tumour
`therapy with immunoconjugates and one anticipates that this
`approach to cancer treatment will be even more effective with
`improvements in conjugate design.
`
`REFERENCES
`BALDWIN R.W. (1985) Design and development of drug-monoclonal
`antibody 791T/36 for cancer therapy. In: Monoclonal antibody
`therapy of human cancer (eds K. Foon and A.C. Morgan), p. 23.
`Martinus Nijoff Publishing, Boston, MA.
`BALDWIN R.W. & BnERs V.S. (1987) Monoclonal antibodies and
`immunoconjugates for cancer treatment. In: Cancer Chemotherapy
`and Biological Response Modifiers Annual 9 (eds H.M. Pinedo, D.L.
`Longo and B.A. Chabner), p. 409. Elsevier Science Publishers,
`Amsterdam.
`BALDWIN R.W. & BYERS V.S. (1988) Monoclonal antibodies in colo
`rectal cancer dia

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