throbber
1 1 8
`
`Applications and Engineering of Monoclonal Antibodies
`
`of PMN recruitment. to the site. Up-regulation of the selectins allows rolling of the Pl\1N
`along the endothelial cells and up-regulation ofintercellular adhesion molecule-I (ICAM-
`1) allows the PMN to become firmly attached to the endothelial cell and then to migrate
`into the interstitium. Imaging sites of inflammation by antibodies to these endothelial cell
`adhesion molecules may therefore give additional selectivity. 1 1 1In-labelled antibodies to
`both E-selectin and ICAM- 1 have been investigated in animal models with promising
`results (Keelan et al., 1 994; Sasso et al. , 1 9 96).
`The abundance of CD4 on inflammatory cells present in arthritic joints has led to
`interest in targeting CD4 to image arthritic joints. Antibodies to CD4 have been used to
`assess targeting in animal models of arthritis. Design of the reagent is crucial for success,
`as intact IgG demonstrated no benefit over a non-specific antibody, whereas a 99mTc­
`labelled Fab' led to improved imaging (Kinne et al. , 1 995). Anti-CD4 reagents may also
`be useful in monitoring the distribution of CD4 positive lymphocytes, which could be
`useful in assessing patients with HIV infection (Rubin et al., 1 996). RAID has also been
`investigated for detection of atherosc!erotic plaques using an antibody towards proliferat­
`ing smooth muscle cells (Narula et al., 19 95) and lesions in Alzheimer's disease using a
`cationised antibody towards beta A4 protein (Bickel et al., 1 994) . Cationisation through
`chemical modification of the antibody with charged groups is believed to aid transport of
`antibodies through the blood:brain barrier, which is obviously helpful to image brain
`lesions in Alzheimer' s disease.
`
`HOSPIRA EX. 1029
`Page 1 26
`
`
`
`126 of 254
`
`Celltrion, Inc. 1195
`Celltrion v. Genentech
`IPR2017-01374
`
`

`

`4
`
`Monoclonal Antibodies in
`Therapeutic Applications
`
`4.1
`
`I ntroduction
`
`The idea of targeted therapy using antibodies dates from the beginning of the twentieth
`century when it was proposed by Paul Ehrlich, and indeed antibodies have been used in
`humans for many years as polyclonal antisera, particularly for passive immunisation.
`Since the discovery of the technology for production of MAbs there has been intense
`interest in their development as therapeutic agents for human disease. However, until
`recently progression of many MAb-based therapeutics was limited by the human immune
`response generated by the administration of murine or rat antibodies. Since the introduc­
`tion ofrecombinant chimeric and humanised antibodies, and ultimately human antibodies,
`interest in developing therapeutics has been revived. There are now five antibody-based
`products licensed for human therapeutic use, and many more under clinical investigation
`(Table 4. 1).
`MAbs can be used in several different modes, depending on the required therapeutic
`effect. In some cases a simple blocking or neutralising effect may be required, for example
`in the neutralisation of an inflammatory cytokine or the blocking of a specific receptor.
`In other applications therapy may require an active role for the antibody by the targeting
`of an effector function. Therapeutics can utilise natural antibody effector functions such
`as complement activation, phagocytosis or ADCC, or entirely novel functions can be
`introduced, as in the case of targeting radioisotopes, drugs or toxins to kill tumour cells.
`In some cases antibodies may also find a role through their ability to mediate signal
`transduction from binding to cell surface receptors (Vitetta and Uhr, 1 994). Available
`mechanisms for generation of therapeutic effects can be considered as a range of anti­
`body effector functions (Table 4.2). Many of these effector functions can be designed
`into, or out of, antibody molecules for specific therapeutic purposes as described in
`Chapter 2. In this chapter the basis of targeted therapy is illustrated using examples of
`the various modes of antibody-based therapy in different disease states. The emphasis is
`placed on the design of antibody-based therapeutics rather than a catalogue of antibodies
`used in therapeutic studies.
`
`HOSPIRA EX. 1 029
`Page 1 27
`
`
`
`127 of 254
`
`Celltrion, Inc. 1195
`Celltrion v. Genentech
`IPR2017-01374
`
`

`

`Table 4.1 Com mercially ava i lable monoclonal antibody therapeutics
`
`1 20
`
`Applications and Engineering of Monoclonal A ntibodies
`
`Therapeutic agent Antibody
`
`Disease
`
`Orthoclone OKT3 Murine OKT3
`
`ReoPro
`
`Chimeric 7E3 Fab
`
`Acute transplant rej ection
`(kidney, liver and heart)
`
`Complications of post-coronary
`angioplasty
`
`Panorex
`
`Rituxan
`
`Zenapax
`
`Table 4.2
`
`Murine 1 7- l A
`
`Colorectal cancer
`
`Chimeric 288
`
`Lymphoma
`
`Humanised anti-tac Acute kidney transplant rejection
`
`Effector functions for antibody targeted therapy
`
`Company
`
`Ortho Biotech
`
`Centocor
`
`Centocor
`
`IDEC/Genentech
`
`Protein Design
`Labs/ Roche
`
`Blocking/neutralising
`
`Natural - Fe mediated effects
`
`Cell signalling
`
`Natural inunune responses
`
`Artificial effectors
`
`B ifunctional
`
`4.2 Cancer
`
`Antigen binding
`
`Complement fixation
`ADCC
`Phagocytosis
`
`Receptor cross-linking
`
`Generation of anti-idiotype response
`Other 'vaccination' approaches
`
`Radioisotopes
`Toxins - bacterial
`- plant
`Cytotoxic drugs
`Cytokines
`Enzymes - prodrug activation
`- direct toxicity
`
`Cross-linking cytotoxic effector cells
`Two-step targeting strategies for
`radioisotopes, toxins, etc.
`
`One of the maj or targets of antibody-based therapeutics has been in the development of
`anti-cancer agents. Initially MAbs to tumour-associated antigens were raised and invest­
`igated without modification, in attempts to target natural antibody effector mechanisms to
`tumour cells. Results of such studies were initially disappointing in many cases, particu­
`larly with the conunon solid tumour types. However, beneficial effects were seen when
`treating patients with minimal disease and in treating lymphomas and leukemias, suggest­
`ing that further research to investigate the role of antibodies as anti-cancer agents may
`lead to improved results. In addition, the use of antibodies to target traditional anti-cancer
`agents such as cytotoxic drugs and radioisotopes has made steady progress, particularly
`since the introduction of recombinant antibodies and improved methods of coupling
`
`HOSPIRA EX. 1 029
`Page 128
`
`
`
`128 of 254
`
`Celltrion, Inc. 1195
`Celltrion v. Genentech
`IPR2017-01374
`
`

`

`Monoclonal antibodies in therapeutic applications
`
`1 2 1
`
`cytotoxic agents (Chapter 2). A s described in Chapter 3, many tumour-associated anti­
`gens representative of different tumour types are now characterised (see Table 3 .5) which
`allow therapeutic molecules to be designed.
`
`4.2.1 Cancer therapy with unmodifed (naked) antibodies
`
`The use of umnodified, or naked, antibodies for tumour therapy initially resulted from
`attempts to harness the immune system through the natural antibody effector fonctions,
`such as ADCC or complement-mediated lysis. More recently it has become apparent that
`cell signalling mechanisms may be involved in many of the anti-tumour effects observed,
`through arrest of the cell cycle or inducing apoptosis. Design of antibodies to elicit
`natural effector functions has largely been a process of choosing the optimal isotype to
`harness human ADCC and complement effects, although as described in Section 2.6. 1 the
`selection of a suitable isotype alone is not sufficient to ensure good cell killing, and
`suitable antigen targets must be individually tested. Mouse IgG2a is the best of the
`murine isotypes for eliciting human ADCC, with rat IgG2b also potent. Of the human
`isotypes, IgG 1 and IgG3 are the most potent in cell killing studies. Of these IgG 1 is most
`commonly used for construction of humanised antibodies, partly because IgG3 antibodies
`are more difficult to purify and to handle in vitro due to a tendency to aggregate. The
`preparation of chimeric or humanised antibodies from rodent antibodies thus has the dual
`benefit of reduced immunogenicity and the ability to use a constant region best suited to
`recruitment of human effector functions. Similarly human antibodies isolated from phage
`display, or other means, can be reconstructed and expressed with the desired isotype
`constant regions. Expression of the antibody in mammalian cells is required as the
`glycosylation of the Cm region is required for maintainence of the ability to elicit
`effector functions. However, cell killing effects are dependent not only on the constant
`region, but also on the disposition of antigenic sites and other poorly understood mech­
`anisms. Many cells also have protective mechanisms against attack from the immune
`system, and thus the in vivo effects of antibodies capable of eliciting cell killing in vitro
`are often difficult or impossible to predict.
`Several alternative mechanisms have also been suggested for tumour cell killing by
`umnodified antibodies. Antibodies against cell surface markers on many types of tumour
`cell can act as ligands eliciting anti-tumour effects by signal transduction (Vitetta and
`Uhr, 1 9 94). Signal transduction can result in arrest of the cell cycle, hence preventing
`tumour cell growth, or in some cases the induction of programmed cell death known as
`apoptosis. In addition, antibodies to growth factors or their receptors may exert anti­
`tumour effects through blocking binding of growth factors needed for tumour cell growth.
`Growth factors such as EGF (epidern1al growth factor) and IL-6 have been implicated in
`the growth of a number of tumour types, and target tumour cells overexpress large
`numbers of molecules of the growth factor receptors. Receptor blocking antibodies pre­
`vent interaction with the ligand, and can lead to down-regulation of the number of
`receptor molecules present on the tumour cell such that growth is inhibited. However,
`unravelling the contribution of each of these factors to effective tumour cell killing is not
`straightforward and often several mechanisms may operate concurrently. Future research
`may allow the selection of antibodies based on an improved understanding of the import­
`ance of eliciting each of these anti-tumour effects. For example, it may be possible to
`select combinations of MAbs capable of different mechanisms of cell killing.
`
`I
`
`HOSPIRA EX. 1 029
`Page 1 29
`
`
`
`129 of 254
`
`Celltrion, Inc. 1195
`Celltrion v. Genentech
`IPR2017-01374
`
`

`

`1 22
`
`Applications and Engineering of Monoclonal Antibodies
`
`As described in Chapter 2, dimeric or polymeric antibodies have been produced either
`by chemical cross-linking or by recombinant means which have increased activity for
`both complement activation and ADCC. Chemically cross-linked constructs comprising
`Fab fragments linked to two Fe regions FabFc2 have also been produced in attempts to
`increase therapeutic efficacy through effector functions. However, studies comparing
`such constructs with a humanised antibody for immunotherapy of multiple myeloma
`showed no advantage in complement activation or ADCC (Ellis et al., 1 995).
`Homodimerisation of IgG has also been reported to be able to confer signalling activ­
`ity, leading to cell cycle arrest or apoptosis, to antibodies which are unable to do so in
`monomeric form (Ghctie et al., I 997a). Chemically cross-linked IgG homodimers were
`produced for several antibodies and shown to be capable of specific anti-tumour effects,
`presumably due to cross-linking the antigen and/or a lower dissociation rate. In the case
`of an anti-CD 1 9 antibody the Fe region was not required for anti-tumour activity, leading
`to the possibility that small multimeric fragments may be capable of potent anti-tumour
`effects.
`Although many studies with unmodified antibodies for cancer therapy have been
`disappointing, with no evidence of anti-tumour effects, several antibodies have shown
`promising effects in the clinic. It is only in the past few years that recombinant chimeric
`and humanised antibodies have reached clinical evaluation. In these studies it has been
`apparent that the ability of the human constant regions to interact with the human im­
`mune system plus the ability to re-treat without the generation of a prohibitive immune
`response is leading to more effective therapeutic agents.
`The first humanised (CDR-graftcd) antibody to be used clinically, CAMPATH-I H,
`recognises the CAMPATH-1 antigen, also known as CDw52, which is present on most
`human lymphocytes and monocytes but not stem cells. CAMPATH- l H has been invest­
`igated as a potential therapeutic agent in non-Hodgkin lymphoma (B-ce!l lymphoma) as
`well as in several inflammatory diseases (Section 4.5.3). Although only limited studies
`have been reported, use of CAMPA TH-I H for the treatment of B-ccll lymphoma has Jed
`to tumour remissions in some patients (Hale et al., 1 988). Studies with different isotypes
`of the rat parent antibody suggest that human effector cells arc involved, as the IgM and
`rat IgG2a versions, which can activate complement but not ADCC, led to only transient
`falls in blood cell counts. In contrast, the rat IgG2b version which could bind to and
`
`activate human effector cells led to much more efficient depletion of tumour cells in vivo
`
`(Dyer et al., 1 989).
`Another antibody, which has shown promising effects in B-cell lymphoma, recognises
`CD20, a phosphoprotein present on the surface of 8 cells. A chimeric IgG 1 version of the
`antibody 288, tenned C288, has been shown in in vitro assays to be an effective media­
`tor of both complement-mediated effects and ADCC using human effector cells (Reff
`et al., 1 994). In addition, this antibody has been shown to induce transmembrane signal­
`ling leading to cell cycle arrest and occasionally apoptosis of CD20 positive cells, and
`thus several mechanisms may be important in the anti-tumour effects observed (Demiden
`et al., 1 995). Phase II and III clinical studies for relapsed B-cell lymphoma have shown
`tumour shrinkage of 50% or more in approximately half of the patients studied (Maloney
`et al., 1 997). In addition, C2B8 was found to sensitise resistant lymphoma cells to certain
`cytotoxic drugs, which may lead to synergistic effects in combination therapy. In a phase
`II clinical study of C2B8 with standard chemotherapeutic treatment, responses were seen
`in all patients studied (Czuczman et al., 1 995). Although as a single agent, tumour
`responses were not as impressive as those observed with radiolabelled antibodies to Co20
`(Section 4.2.5), treatment with this unmodified antibody was much less toxic and is more
`
`HOSPIRA EX. 1 029
`Page 1 30
`
`
`
`130 of 254
`
`Celltrion, Inc. 1195
`Celltrion v. Genentech
`IPR2017-01374
`
`

`

`lvfonoclonal antibodies in therapeutic applications
`
`1 23
`
`straightforward than handling large amounts of radioisotope. Therefore, it is likely that this
`agent will find clinical application alongside chemotherapy for B-cell lymphoma treatment.
`The HER2 gene (also known as neu or c-erb-B2) is a proto-oncogcnc which encodes
`a transmembrane growth factor receptor (pl85 HER2) which is ovcrexprcsscd in 25-30% of
`patients with breast cancer, particularly those with a poor prognosis. A humanised anti­
`body to p l 85 11ER1 has been developed which has shown significant growth inhibition of
`cells overexpressing this receptor (Carter et al., 1 992b). The humanised antibody was
`also shown to be more efficient at mediating ADCC with human effector cells than the
`parent murine antibody. In a phase II clinical study in patients with metastatic breast
`cancer that overcxpressed HER2, objective responses were seen in 5 of 43 patients
`including one complete remission and 4 partial remissions (Baselga et al., 1 996). The
`mechanisms of action of this agent are also not clear at present. The antibody induced a
`clear down-regulation of the growth factor receptor which may reverse the malignant
`phenotype. This antibody is also known to be capable of activation of a signal transduction
`pathway that leads to inhibition of tumour cell proliferation and possibly cell death, and
`also elicits ADCC.
`
`4.2.2 Anti-idiotype antibodies
`
`MAbs may also be used as vaccines to generate an anti-idiotype response. The idotype of
`an antibody is made up of a cluster of cpitopes at the antigen-binding site. As each MAb
`has a different antigen-binding site, the idiotype of each MAb is unique. Antibodies can
`be raised through an immune response to the idiotype which have a conformation that
`resembles the antigen of the original antibody (Figure 4.1 ). These anti-idiotype antibodies
`arc also known as anti-id or ab2 antibodies. Immunisation with an ab2 antibody can in
`tum lead to the elicitation of an anti-idiotype to the ab2 antibody, an anti-anti-id, known
`as ab3, a proportion of which will have the same or overlapping specificity as the original
`ab 1 antibody (Figure 4. 1 ). Structural studies suggest that anti-idiotype antibodies carry an
`' internal image' of the original antigen which allows the development of ab3 responses
`that recognise the original antigen. The crystallographic structure of an anti-idiotype
`antibody in complex to an antibody raised against lysozyme showed that the same antigen­
`binding residues used to bind lysozyme were used for binding to the ab2, and that the ab2
`mimicked lysozyme (Fields et al., 1 995).
`Vaccination with ab2 has been investigated in attempts to raise a human ab3 response
`to target antigens which may be poorly immunogenic or difficult to prepare or use in
`immunisation. ab2 may also be able to raise antibodies not raised to the original antigen
`through breaking immunological tolerance and also give rise to T cell responses which
`may have therapeutic significance. Many studies, both preclinical and clinical, have been
`conducted in attempts to achieve therapeutic effects with ab2 vaccines for cancer (re­
`viewed by Herlyn et al., 1 996). In several cases antigen-specific immune responses have
`been achieved, including both antibody and cellular responses. For example, the antibody
`1 0 5AD7 is an anti-idiotype antibody that mimics a colorectal tumour-associated antigen.
`Phase I clinical studies demonstrated that a T cell response to tumour was induced on
`treatment with I 05AD7 and this resulted in delayed tumour growth and an increase in
`survival time compared with patients at the same stage of disease that did not receive the
`antibody (Buckley et al., 1 995).
`It is, of course, also possible for antibodies to tumour-associated antigens administered
`for tumour therapy (ab l ) to elicit anti-idiotype responses. The murine MAb 1 7- l A has
`
`HOSPIRA EX. 1 029
`Page 1 3 1
`
`
`
`131 of 254
`
`Celltrion, Inc. 1195
`Celltrion v. Genentech
`IPR2017-01374
`
`

`

`1 24
`
`Applications and Engineering of lvfonoclonal Antibodies
`
`(a)
`
`(b)
`
`(c)
`
`Figure 4.1 Representation of anti-idiotype antibodies: (a) in itial antibodies (abl ) recognise
`
`the antigen to which they were raised; (b) anti-id iotype antibod ies (ab2) recognise the
`antigen-b inding region of ab1 and thus can be considered as contain ing an 'internal
`image' of the antigen; (c) anti-anti-idiotype anti bodies (ab3) raised against ab2 have
`similar specificity to the original antibody (ab 1 ) and can thus recogn ise the same antigen
`
`been extensively studied. It recognises the epithelial membrane antigen (EMA) present
`on the majority of colorectal tumour cells. Originally developed for its ability as a murine
`IgG2a to elicit ADCC in humans, early clinical studies revealed occasional responses in
`patients with advanced disease which were delayed following antibody administration,
`implying an active immune response. Most patients developed an anti-idiotype response
`to 1 7-lA which may have led to the observed therapeutic effects (Fagerberg et al., 1 996).
`In a phase III trial of 1 7-l A for the treatment of minimal residual disease in colorectal
`cancer patients following surgical removal of the primary tumour, 1 7- 1 A treatment re­
`sulted in a statistically significant improval in survival (Riethmuller et al., 1 994 ). After
`following patients for five years, 17-1 A therapy reduced the overall death rate by 30%
`and decreased the recurrence rate by 27%. These results led to the approval of 1 7- l A as
`a therapeutic agent, sold under the name ' Panorex' (Table 4. 1). Analysis has identified
`patients with high levels of ab3 as those that live longer, suggesting that the induction of
`an anti-idiotype response is important in the mechanism of the therapeutic effect of this
`antibody (Fagerberg et al., 1 996).
`
`HOSPIRA EX. 1029
`Page 1 32
`
`
`
`132 of 254
`
`Celltrion, Inc. 1195
`Celltrion v. Genentech
`IPR2017-01374
`
`

`

`Monoclonal antibodies in therapeutic applications
`
`1 25
`
`(a)
`
`(b)
`
`figure 4.2 Mechanisms of targeting effector cells to tumours: (a) act ive isotypes which
`can bind to Fe receptors on the effector cel l can be used to target the cel l to a tumou r­
`
`associated antigen; (b) alternatively, bispeci fic a ntibodies may be used which bind to both
`the tumour cell and the effector cel l through the antigen-binding arms of the antibody
`(for further details see text)
`
`4.2.3 Bispecific antibody-mediated effector cell targeting
`
`The limited success that has been achieved in using antibodies to elicit ADCC responses
`has prompted the search for other approaches to recruit effector cells, such as cytotoxic T
`cells, to the tumour site. One such approach is to use bispecific antibodies with one
`specificity for a tumour-associated antigen and one for the effector cell such that the
`effector cell is linked directly to the tumour cell (Figure 4.2). A surface 'trigger molecule'
`is used as the antigen on the effector cell such that the cell is activated to lead to a
`cytotoxic response. Several different types of effector cell and trigger molecule have been
`investigated, including Fey receptors and molecules of the CD3ff cell receptor complex
`(Table 4.3). The advantages and disadvantages of each of these have been reviewed
`(Renner and Pfreundschuh, 1 995). In addition to requiring triggering, cytotoxic effector
`mechanisms may also require the involvement of accessory molecules and activation via
`cytokine release.
`On monocytes, macrophages, eosinophils and polymorphonuclear neutrophils (PMNs),
`activation is achieved through linking to Fey receptors. Natural killer cells are activated
`through FcyRIII and CD2, whereas activation of T cells is usually achieved through the
`
`I
`
`HOSPIRA EX. I 029
`Page 1 33
`
`
`
`133 of 254
`
`Celltrion, Inc. 1195
`Celltrion v. Genentech
`IPR2017-01374
`
`

`

`1 26
`
`Table 4.3 Cytotoxic effector cells and trigger molecules
`
`Applications and Engineering of Monoclonal Antibodies
`
`Effector cell
`
`T cells
`
`Natural killer cells
`
`Monocytes/macrophages
`
`Granulocytes ( eosinophils, PMNs)
`
`PMNs activated with interferon-y,
`GM-CSF or G-CSF
`
`Trigger molecules
`
`CD3
`T cell receptor (y/o)
`CD2
`CD28
`
`Fey receptor III (CD 1 6)
`CD2
`
`F cy receptor I ( CD64)
`Fey receptor II (CD32)
`F cy receptor III (CD 1 6)
`
`Fey receptor II (CD32)
`
`Fey receptor I (CD64)
`
`CD3/T-cell receptor complex. The CD3/T-cell receptor complex is normally responsible
`for antigen-specific T cell responses, but the use of bispecific antibodies allows activation
`through this complex which can be redirected to the antigen recognised by the other arm
`of the bispecific antibody. Activation of effector cells is greatly increased by cytokines
`such as interleukin-2 or interlcukin-7, or by co-stimulatory signals. CD2 and CD28 arc
`co-stimulatory molecules which when stimulated increase the activation of T cells. For
`example, combinations of triggering CD3 and CD28 allow particularly potent anti­
`tumour effects. Cell killing by a bispecific antibody cross-linking the Hodgkin lymphoma
`antigen CD30 to CD3 resulted in relatively little tumour cell killing unless co-administered
`with CF30-CD28 bispecific antibody (Renner et al., 1994). The combination was also far
`more potent in treatment of xenografted human tumour in immunodeficient mice
`repopulated with human T cells. Cures of established tumours could only be achieved
`with the combination of both bispecific molecules (Renner et al., 1 994 ). Similar effects
`can be achieved by co-administration of anti-CD28 IgG with anti-CD3/anti-tumour
`
`bispecific antibodies (Demanet et al., 1996). An alternative approach has been the use of
`interesting results, although these have been limited by the need to activate T cells ex vivo
`
`a trispecific antibody, made by cross-linking three Fab' fragments, with specificity for
`tumour antigen as well as two different T cell antigens (Tutt et al., 1 99 1 b ).
`Several phase I clinical studies have been carried out with bispecific antibodies with
`
`and re-infuse to the patient at the time of bispecific antibody therapy. IL-2 activated cells
`targeted by an anti-CD3/anti-tumour bispecific antibody administered intracranially to
`glioma patients resulted in survival of 76% of patients over two years, compared to 33%
`of those treated with activated cells alone (Nitta et al., 1990). Anti-FcyRIII/anti-tumour
`antibodies designed to activate natural killer cells and mononuclear phagocytic cells have
`also been used clinically, with minor responses observed (Weiner et al., 1 995).
`The production of bispecific antibodies can be achieved using a variety of approaches,
`including the production of hybrid hybridomas, chemical cross-linking and recombinant
`approaches (Section 2.5). The affinity of the antibody for the triggering molecule may
`also be important. Humanised Fab' fragments to the breast tumour antigen pl 85HER2 and
`
`HOSPIRA EX. 1029
`Page 1 34
`
`
`
`134 of 254
`
`Celltrion, Inc. 1195
`Celltrion v. Genentech
`IPR2017-01374
`
`

`

`Monoclonal antibodies in therapeutic applications
`
`127
`
`to CD3 have been expressed and cross-linked to form a bispecific antibody capable of
`efficient lysis of tumour cells (Zhu et al. , 1 995). In these studies a high affinity version of
`the humanised anti-CD3 Fab' was found to be more effective at tumour cell lysis than a
`lower affinity version. The same antibody specificities have been used to produce a
`bispecific diabody which was equally active in tumour cell lysis but was much simpler to
`produce as it was directly expressed by secretion from E. coli at high yield (Zhu et al.,
`1 996). Under some circumstances diabodies may be more effective than bispecific IgG at
`achieving tumour cell lysis, possibly due to the smaller size of the diabody bringing
`target and effector cell closer together (Holliger et al., 1 996).
`
`4.2.4 Other approaches to recruit the immune system using MAbs
`
`Superantigens are bacterial or viral proteins, such as the exotoxin produced by
`Staphylococcus aureus, that stimulate T cells by cross-linking the T cell receptor to MHC
`class II molecules. Superantigens bind to the T cell receptor outside the antigen recogni­
`tion site and to MHC class II molecules outside the peptide binding groove normally
`involved in antigenic recognition, bypassing normal immunological specificity. Super­
`antigens are thus the most potent known activators of T cells. Fab'-superantigen fusion
`proteins have been produced which stimulate T cells and target to tumour cells. The
`fusion proteins are intended to replace MHC class II binding with tumour cell binding
`through the Fab'. Such fusion proteins have been shown to be capable of inhibiting
`tumour growth in animal models through causing activated T cells to infiltrate and attack
`the tumour (Dohlstein et al., 1995). One problem with the use of such agents, however,
`is residual MHC class II binding which leads to relatively high toxicity through systemic
`T cell activation and accumulation of inflammatory cytokines in serum. Attempts to
`improve Fab'-superantigen fusion proteins for tumour therapy are thus being made by
`mutations in the superantigen binding site for MHC class II which prevent binding and
`reduce systemic toxicity (Hansson et al., 1 997).
`Other approaches in attempts to improve the recruitment of the immune system by
`antibodies have included the use of antibody-cytokine fusion proteins. Certain cytokines,
`such as inter!eukin-2 (IL-2) are able to activate multiple- immune mechanisms, although
`attempts at tumour therapy with cytokine alone have led to severe systemic toxicity. Fusion
`proteins have thus been designed to target a high local concentration of cytokine to the
`tumour site, minimising systemic toxicity. Several different cytokines have been investig­
`ated. IL-2 stimulates T cell proliferation and T cell mediated killing and has thus been
`
`widely investigated, but tumour necrosis factor a (TNFa), TNF�, GM-CSF, IL-5, IL-8
`and interferon a have also been tested. Fusion proteins have been produced with several
`
`antibodies and encouraging pre-clinical results obtained. Chimeric 14. 1 8, an antibody that
`recognises the GD2 ganglioside expressed on neuroblastoma, melanoma and certain other
`tumours, has been used to produce fusions with several cytokines, the most promising of
`which appears to be IL-2 (Hank et al., 1 996). The fusion protein maintained antigen bind­
`ing and IL-2 activity and was able to exert anti-tumour effects in mice bearing human
`tumour xenografts of neuroblastoma and melanoma. Activation of human effector cells
`has also been demonstrated (Hank et al., 1 996). Cytokine fusion proteins have also been
`produced with small antibody fragments such as scFv, which may allow more effective
`tumour penetration (Dorai et al., 1 994 ). However, whether such fusion proteins will be
`sufficiently less toxic than free IL-2 when administered to patients remains to be seen.
`
`HOSPIRA EX. 1 029
`Page 1 3 5
`
`
`
`135 of 254
`
`Celltrion, Inc. 1195
`Celltrion v. Genentech
`IPR2017-01374
`
`

`

`1 28
`
`Applications and Engineering of Monoclonal Antibodies
`
`4.2.5 Radioimmunotherapy
`As with diagnostic tumour imaging (Section 3.9. 1), the design of antibodies for
`radioimmunotherapy (RIT) requires careful consideration of the individual components
`of the immunoconjugate, including the fom1 of the antibody, the radioisotope and the
`means of attachment of the radioisotope to the antibody. In addition, the choice of a
`suitable tumour-associated antigen becomes more critical with therapeutic doses of iso­
`tope, as the presence of antigen on normal tissues will result in the deposition of activity,
`which may lead to unacceptable toxicity. Many factors influence the degree of tumour
`localisation in patients. These include heterogeneous expression of antigen, presence of
`circulating antigen, tumour vascularisation and penetration of antibody into tumour tissue
`(Boxer et al., 1992). However, there may not be a need to achieve localisation to each
`individual tumour cell, as a radioisotope can be chosen with a pathlength of many cell
`diameters, allowing for 'bystander cell' killing at the tumour site. Nevertheless, a high
`proportion of tumour cells expressing the antigen is desirable. This is because the major
`limitation to RIT is the dose of isotope which can be delivered at the tumour site without
`resulting in unacceptable toxicity. In general, only a small proportion of the dose admin­
`istered localises to the tumour site in patients. Typically tumour-localisation levels of
`1 0% injected dose per kg of tumour (0.0 1 % injected dose per gram of tumour) are
`observed in human studies where good targeting is achieved (van Hof et al., 1 996). The
`total administered dose is usually dependent on the toxicity to normal body tissues. Bone
`marrow is the most sensitive normal tissue to radiation and therefore doses are usually
`limited by the dose to bone marrow from radiolabelled antibody circulating in the blood
`(Badger, 1990). Several strategies have been investigated in attempts to reduce bone
`marrow toxicity, including the use of short-range radioisotopes which require cell inter­
`nalisation for cytotoxicity (Auger emitters), the use of specific clearing mechanisms to
`remove circulating activity, two-step targeting strategies and the use of rapidly clearing
`antibody fragments.
`Careful optimisation of an immunoconjugate for RIT, through antibody engineering
`and development of suitable chemistry for radioisotope attachment, can lead to improved
`properties both in vitro and in vivo (King et al., 1 994), and it is likely that such optimised
`immunoconjugates will provide the next generation of molecules of clinical utility for
`RIT. Some of the parameters which need to

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