throbber
Targeted Therapy of Cancer
`
`Targeted Therapy of Cancer: New
`Prospects for Antibodies and
`Immunoconjugates1
`
`Robert M. Sharkey, PhD; David M. Goldenberg, ScD, MD
`
`Dr. Sharkey is Member and Direc-
`tor of Clinical Research, Garden
`State Cancer Center at the Center for
`Molecular Medicine and Immunol-
`ogy, Belleville, NJ.
`
`Dr. Goldenberg is President, Gar-
`den State Cancer Center at the Cen-
`ter
`for Molecular Medicine and
`Immunology, Belleville, NJ.
`
`This article is available online at
`http://CAonline.AmCancerSoc.org
`
`ABSTRACT Immunotherapy of cancer has been explored for over a century, but it is only in the
`last decade that various antibody-based products have been introduced into the management of
`patients with diverse cancers. At present, this is one of the most active areas of clinical research, with
`eight therapeutic products already approved in oncology. Antibodies against tumor-associated
`markers have been a part of medical practice in immunohistology and in vitro immunoassays for
`several decades, have even been used as radioconjugates in diagnostic imaging, and are now
`becoming increasingly recognized as important biological agents for the detection and treatment of
`cancer. Molecular engineering has improved the prospects for such antibody-based therapeutics,
`resulting in different constructs and humanized/human antibodies that can be administered fre-
`quently. Consequently, a renewed interest in the development of antibodies conjugated with radio-
`nuclides, drugs, and toxins has emerged. We review how antibodies and immunoconjugates have influenced cancer detection and therapy,
`and also describe promising new developments and challenges for broader applications. (CA Cancer J Clin 2006;56:226–243.) © American
`Cancer Society, Inc., 2006.
`
`INTRODUCTION
`
`The search for a mechanism to target diseases selectively was first realized when resistance to infectious disease
`could be transferred from one animal to another through their serum, a process known as passive serotherapy.1 Five
`years later, in 1895, Hericourt and Richet immunized dogs with a human sarcoma and then transferred the serum
`to patients.2 This anticipated the “magic bullet” concept of Paul Ehrlich in 1908, that “toxins” could be targeted to
`cancer and other diseases.3 Another half-century passed before antibodies were identified as the substance in serum
`responsible for these effects.
`Despite being potent immune system instigators for killing infectious agents, clinical research initially focused on
`immunoconjugates prepared with radionuclides, drugs, or toxins, since unconjugated or “naked” antibodies had little
`therapeutic benefit in oncology compared with the immunoconjugates. Early immunotherapy trials failed to show
`substantial responses,4 – 6 but antibodies against carcinoembryonic antigen (CEA) could selectively target and disclose
`sites of CEA-expressing cancers in patients, and also deliver cytotoxic radioactivity in human colonic cancer
`xenografts having CEA.7,8 Thereafter, DeNardo, et al.9 reported responses in lymphoma patients to radiolabeled
`antibodies, and soon others confirmed that radiolabeled antibodies had antitumor activity in non-Hodgkin lym-
`phoma (NHL), but there was also early evidence that the naked antibodies themselves might be effective.10 –12 It was
`during this same period that rituximab (Rituxan, Genentech, and biogen idec), an anti-CD20 IgG, became of interest
`as a therapeutic for NHL without being radiolabeled.13 The experience and subsequent introduction of rituximab
`into the treatment of NHL can be credited for the expanded interest in unconjugated antibodies for cancer therapy.
`
`1This work was supported in part by USPHS grant P01-CA103985 from the National Cancer Institute, NIH, and grant 06-1853-FS-N0 from the New Jersey
`Department of Health and Senior Services.
`
`226
`
`CA A Cancer Journal for Clinicians
`
`IMMUNOGEN 2136, pg. 1
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`CA Cancer J Clin 2006;56:226–243
`
`Antibodies (eg, IgG, which is the most com-
`monly used immunoglobulin form, Figure 1)
`are unique proteins with dual
`functionality.
`All naturally occurring antibodies are multi-
`valent, with IgG having two binding ‘arms.’
`Antigen-binding specificity is encoded by
`three complementarity-determining regions (CDRs),
`while the Fc-region is responsible for binding to
`serum proteins (eg, complement) or cells. An
`antibody itself usually is not responsible for
`killing target cells, but instead marks the cells
`that other components or effector cells of the
`body’s immune system should attack, or it can
`initiate signaling mechanisms in the targeted
`cell that leads to the cell’s self-destruction (Fig-
`ure 2). The former two attack mechanisms are
`referred to as antibody-dependent complement-
`mediated
`cytotoxicity
`and antibody-
`(CMC)
`dependent cellular cytotoxicity (ADCC). ADCC
`involves the recognition of the antibody by
`immune cells that engage the antibody-marked
`cells and either through their direct action, or
`through the recruitment of other cell types,
`lead to the tagged-cell’s death. CMC is a pro-
`cess where a cascade of different complement
`proteins become activated, usually when sev-
`eral IgGs are in close proximity to each other,
`either with one direct outcome being cell lysis,
`or one indirect outcome being attracting other
`immune cells to this location for effector cell
`function.
`Antibodies, when bound to key substances
`found on the cell surface, also can induce cells
`to undergo programmed cell death, orapoptosis
`(Figure 2). For example, if rituximab binds to
`two CD20 molecules, this triggers signals in-
`side the cell that can induce apoptosis.14 If
`rituximab is cross-linked by other antiantibod-
`ies, the apoptotic signal is intensified.15 This
`cross-linking could also occur when the anti-
`body is bound by another immune cell through
`its Fc-gamma receptors (Fc␥R). Other anti-
`bodies, such as trastuzumab (anti-HER2/neu;
`Herceptin, Genentech) and cetuximab (anti-
`epidermal growth factor receptor, EGFR; Er-
`bitux,
`ImClone Systems and Bristol-Myers
`Squibb) also have the ability to inhibit cell
`proliferation.16 –18 Because cells
`frequently
`have alternative pathways for critical functions,
`interrupting a single signaling pathway alone
`
`might not be sufficient to ensure cell death.
`From this perspective, it is not surprising that
`antibodies are often best used in combination
`with chemotherapy and radiation therapy to
`augment their antitumor effects.19 –21
`Bevacizumab (Avastin, Genentech) is yet
`another example of how antibodies can be
`used therapeutically. This antibody binds to
`vascular endothelial growth factor (VEGF)
`that is made by tumor cells to promote vessel
`formation, thereby preventing it from inter-
`acting with endothelial cells to form new
`blood vessels (Figure 2).22 Antibodies can
`also be used to modulate immune response.
`Antibodies to the cytotoxic T-lymphocyte
`associated antigen-4 (CTLA-4)
`stimulate
`T-cell
`immune responses by blocking the
`inhibitory effects of CTLA-4, which can en-
`hance tumor rejection.23 However, release of
`this innate inhibitory mechanism can also
`increase the risk of autoimmunity.24 Two
`human anti-CTLA-4 antibodies are currently
`in early clinical trials (MDX-010, Medarex,
`and CP-675,206, Pfizer), with evidence that
`they may have activity in melanoma.24 There are
`already a number of antibodies used or be-
`ing studied as therapeutic agents in cancer as
`well as autoimmune diseases (eg, alemtuzumab,
`daclizumab,
`infliximab,
`rituximab,
`epratu-
`zumab).25–31 Antibodies also can block molecules
`associated with cell adhesion, thereby inhibiting
`tumor metastasis.32,33 With such diverse mecha-
`nisms of action, there are a number of opportu-
`nities for building antibody-based therapeutics.
`Antibodies naturally have long serum half-
`lives. For immunotherapy,
`this property is
`helpful because the antibody is maintained in
`the body fluids, where it can continually inter-
`act with its target. For other targeting strategies,
`most notably with radioconjugates, it can be
`harmful because the highly radiosensitive bone
`marrow is continually exposed to radiation,
`resulting in dose-limiting myelosuppression.
`The large size of an antibody impacts its ability
`to move through a tumor mass. A high inter-
`stitial pressure inhibits the diffusion of larger
`molecules within the tumor.34 Migration
`within the tumor is also inhibited by a binding-
`site barrier, a process where the antibody as it is
`leaving the tumor’s blood vessels binds to the
`
`Volume 56 Y Number 4 Y July/August 2006
`
`227
`
`IMMUNOGEN 2136, pg. 2
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`Targeted Therapy of Cancer
`
`FIGURE 1 Schematic representation of an IgG molecule, its chemically produced fragments, and several recombinant
`antibody fragments with their nominal molecular weights. At the bottom, a schematic representation of the process in-
`volved in engineering murine MAbs to reduce their immunogenicity is provided. A chimeric antibody splices the VL and
`VH portions of the murine IgG to a human IgG. A humanized antibody splices only the CDR portions from the murine
`MAb, along with some of the adjacent “framework” regions to help maintain the conformational structure of the CDRs. A
`fully human IgG can be isolated from specialized transgenic mice bred to produce human IgG after immunizing with tu-
`mor antigen or by a specialized phage display method.
`
`228
`
`CA A Cancer Journal for Clinicians
`
`IMMUNOGEN 2136, pg. 3
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`CA Cancer J Clin 2006;56:226–243
`
`FIGURE 2 Mechanisms of action associated with unconjugated antibodies. In this example, the antigen is shown to be
`floating in lipid rafts within the tumor cell membrane. (A) Antibodies can activate apoptotic signals by cross-linking anti-
`gen, particularly across different lipid rafts. Additional cross-linking of antibody by immune cells can also enhance cellu-
`lar signaling. (B) Immune cells themselves can attack the antibody-coated cell (eg, phagocytosis), and/or they can
`liberate additional factors, such as cytokines that attract other cytotoxic cells. (C) If antibodies are positioned closely to-
`gether, they can initiate the complement cascade that can disrupt the membrane, but some of the complement compo-
`nents also are chemo-attractants for immune effector cells and stimulate blood flow. (D) Tumors also can produce
`angiogenic factors that initiate neovascularization. Antibodies can neutralize these substances by binding to them, or
`they can bind directly to unique antigens presented in the new blood vessels, where they could exert similar activities.
`
`first available antigen, concentrating the anti-
`body in the perivascular space.35 High-affinity
`antibodies are less likely to migrate into the
`tumor bed.36 Administering higher doses of the
`antibody can reduce the effect of the binding
`site barrier and allow the antibody to diffuse
`more deeply into the tumor bed.37 For cyto-
`toxic agents that must be internalized to kill the
`cell (eg, toxins, cytotoxic drugs), the ability to
`distribute throughout the tumor is important.
`Radioconjugates are less affected by this be-
`cause some radioactive particles can traverse as
`much as 1.0 cm from where they are deposited
`(bystander or crossfire effect).
`
`THERAPY WITH UNCONJUGATED ANTIBODIES
`
`A renewed interest in the effects of un-
`conjugated antibodies in cancer began in the
`early 1980s, after murine monoclonal anti-
`(MAbs) became available.38 These
`bodies
`initial trials were performed in hematological
`malignancies, as well as in colorectal cancer
`and melanoma.4 – 6,39 – 41 As with many inno-
`
`vative treatment approaches that are some-
`times introduced before the technology has
`matured sufficiently to extract maximum
`benefit, only occasional clinical
`responses
`were observed. With insufficient efficacy and
`the immunogenicity of the foreign murine
`MAb, most of these studies were terminated.
`Fortunately, some investigators persevered.
`An excellent lesson on the tribulations of the
`development of an antibody product be-
`tween an academic group and industry is that
`of alemtuzumab (Campath, Berlex, and Gen-
`zyme).42 Alemtuzumab (anti-CD52) had one
`of the earliest and protracted developments
`of an antibody ultimately commercialized. It
`took over 20 years from the development of
`the first rat immunoglobulin against CD52,
`changing the immunoglobulin type, and fi-
`nally developing a humanized, recombinant
`form, and involved several commercial firms
`during this
`time. Chemotherapy-refractive
`chronic lymphocytic leukemia was the indi-
`cation finally approved in 2001.
`Due in part to the contributions made by the
`groups led by Morrison (Columbia and Stan-
`
`Volume 56 Y Number 4 Y July/August 2006
`
`229
`
`IMMUNOGEN 2136, pg. 4
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`Targeted Therapy of Cancer
`
`ford Universities) and Winter (Cambridge),
`MAbs now are engineered to remove a signif-
`icant portion of the murine component of the
`IgG, substituting human IgG components be-
`fore entering clinical studies.43– 45 Chimeric
`antibodies essentially splice VL and VH regions
`on the murine antibody to the human IgG,
`making a molecule that is 75% human and 25%
`murine IgG, whereas a humanized antibody
`grafts the CDR regions from a murine MAb,
`along with some of the surrounding “frame-
`work” regions to maintain CDR conforma-
`tion, onto a human IgG, essentially making a
`molecule with 5% of its sequence from the
`parental MAb (Figure 1).45 More recent ad-
`vances have made available, either by genetic
`or phage-display methods, the development of
`fully human MAbs that have now entered clin-
`ical trials.46 Such engineered MAbs are postu-
`lated to greatly reduce the immunogenicity of
`antibodies, allowing multiple injections to be
`given, and the human Fc enhances the inter-
`action with other immune system elements.
`Rituximab is perhaps the most prominent
`example of a highly successful paradigm of an-
`tibody therapy. As a chimeric antibody, not
`only did it have reduced immunogenicity, but
`its effector function (associated with the Fc-
`portion) was improved. For example, when
`testing ADCC activity against follicular lym-
`phoma isolated from 43 patients, Weng, et al.
`reported that only rituximab, not its parent
`murine anti-CD20 IgG (2B8), had activity in
`vitro.47 Rituximab was initially approved as a
`single agent therapy for relapsed or refractory
`low-grade, follicular B-cell NHL, having an
`overall response rate of 48% (10% were com-
`plete responses, CR) with a median duration of
`11.8 months.48,49 Since CD20 is not expressed
`on precursors B-cells, rituximab induces a de-
`pletion of only mature B-cells. Rituximab’s
`major side effects, which are thought to be
`associated with the activation of complement
`pathways, occur during or shortly after its in-
`fusion. Other less common side effects include
`symptoms associated with tumor lysis
`syn-
`drome, severe mucotaneous reactions, renal
`toxicity, cardiac arrhythmias, hypersensitivity
`reactions, and reactivation of hepatitis B (pri-
`
`marily when used in combination with chemo-
`therapy).49
`Rituximab’s activity is unique among cancer
`treatments because 40% of
`the patients re-
`treated with rituximab could again respond
`with a similar duration.50 Extending the dura-
`tion of rituximab therapy can improve the re-
`sponse
`rate, particularly
`the number of
`complete responses, and its duration. However,
`whether given as a maintenance regimen or
`retreating at the first sign of progression, the
`time to chemotherapy was the same.51 With
`both approaches having equal benefit, retreat-
`ment is generally favored because of the higher
`expense of a maintenance regimen. Despite the
`success of rituximab as a monotherapy, there
`are still a number of patients who do not re-
`spond to the initial treatment, and over time,
`many of those who do will relapse. In an at-
`tempt to improve outcome, rituximab has been
`combined with chemotherapy regimens,
`in-
`cluding CHOP, CVP, and MCP, as front-line
`treatments, with very promising results in not
`only follicular B-cell lymphomas, but also in
`lymphomas.52,53 Indeed,
`diffuse large B-cell
`trials examining front-line combinations of rit-
`uximab and chemotherapy have already dem-
`onstrated improvements in response rates, time
`to progression, and event-free survival, and
`while the overall response rates are promising
`based on current 2- to 3-year follow-up data,
`more time will be required to fully appre-
`ciate its impact.52 Even in chronic lymphocytic
`leukemia (CLL), where initial testing of ritux-
`imab was disappointing, dose intensifica-
`tion and combinations with chemotherapy
`have provided significant
`improvements
`in
`response.54,55 Early clinical studies combining
`rituximab with a humanized anti-CD22,
`epratuzumab (Immunomedics, Inc.) suggested
`the potential for additional benefit, particularly
`in patients with diffuse large B-cell lympho-
`mas.56,57 Studies have also assessed the possible
`role of an anti-CD80 MAb (galiximab, biogen
`idec) as a monotherapy in NHL,58,59 and clin-
`ical trials are in progress testing its combination
`with rituximab.
`Considerable attention has been devoted to
`understanding the mechanism of action of rit-
`uximab, particularly why some B-cell lympho-
`
`230
`
`CA A Cancer Journal for Clinicians
`
`IMMUNOGEN 2136, pg. 5
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`CA Cancer J Clin 2006;56:226–243
`
`mas are affected and why not all patients with
`follicular lymphomas respond. As mentioned
`earlier, rituximab has been shown to have
`CMC, ADCC, and apoptotic activity, with the
`former two mechanisms believed to have the
`greatest impact, although there are conflicting
`views of which of these two pathways contrib-
`utes the most to the response.14,60 – 66 Studies in
`transgenic and other mouse models have sup-
`ported the importance of
`the Fc-receptor-
`mediated mechanism of
`action
`for
`rituximab.67,68 These efforts have contributed
`in part to a better understanding of the role of
`various Fc receptors found on a variety of im-
`mune effector cells (eg, B-cells, neutrophils,
`natural killer cells, and monocytes) on (in the
`case of rituximab) the clearance of B-cells, as
`well as the plasma half-life of antibodies.69 Not
`only do the various Fc-receptors
`influence
`binding, but the absence of certain carbohy-
`drates on the Fc portion of the IgG can affect
`both ADCC and CMC activities.70,71 Cartron,
`et al. found that the expression of the homozy-
`gous Fc-gamma RIIIa receptor (CD16) 158V
`genotype correlated with a higher response rate
`to rituximab, but it did not have an impact on
`the progression-free survival.72 Weng, et al.
`found a similar correlation and also noted that
`the homozygous expression of the Fc-gamma
`RIIa histidine/histidine genotype correlated
`independently with a higher response rate, par-
`ticularly when assessing the response status ⱖ6
`months from treatment.47 By unraveling the
`molecular basis for antibody cytotoxicity, not
`only can more effective antibodies be designed,
`but it could lead to a more rational approach
`for combinations to enhance activity, such as
`the finding that G-CSF up-regulates CD64
`(Fc-gamma receptor I), which can enhance the
`binding of neutrophils and monocytes
`to
`B-cells coated with rituximab.73 IL-12 has a
`similar stimulatory effect in mouse models and
`more recently has been applied clinically with
`promising results.74,75 These discoveries are
`also having an impact on the development of
`antibodies for treating other cancers.76 – 80
`The approved antibodies listed in Table 1 in-
`dicate that immunotherapy is not restricted to
`hematological malignancies, but includes diverse
`target antigens and receptors having different
`
`functions. Trastuzumab is an anti-
`biological
`HER2/neu antibody that has had a major impact
`on the therapy of breast cancer and is used alone
`and in combination with drugs.81– 83 HER2/neu
`is overexpressed on a proportion of breast and
`other cancers, and trastuzumab binds with an
`extracellular epitope of
`this
`target molecule.
`About 15% of women whose tumors overexpress
`HER2/neu respond to trastuzumab, but its effi-
`cacy is clearly best when used in combination
`with chemotherapy, where a 25% increase in the
`median survival (to 29 months) has been report-
`ed.81 Further, the addition of this antibody to
`adjuvant chemotherapy for breast cancer has im-
`proved survival markedly.83 Since only a portion
`of breast cancer patients overexpress HER2/neu
`and respond to trastuzumab, selection of suitable
`patients is important. New data are emerging that
`suggest trastuzumab treatment after adjuvant che-
`motherapy can have a significant benefit com-
`pared with observation, particularly in reducing
`the rate of distant recurrence.82
`As a member of a family of receptor tyrosine
`kinases, the binding of HER2 by trastuzumab
`can interrupt intracellular signaling and affect
`tumor cell growth. Izumi, et al. showed that
`trastuzumab also has antiangiogenic proper-
`ties.84 While this may be an important under-
`lying mechanism of action, other evidence
`suggests that trastuzumab’s activity is princi-
`pally governed by ADCC.85 However, trastu-
`zumab combined with chemotherapy improves
`response rates, despite the immunosuppressive
`activity of the chemotherapy, and trastuzum-
`ab’s activity is enhanced when combined with
`other, nonantibody, Erb inhibitors, such as ge-
`fitinib and erlotinib, all of which suggest that its
`ability to interfere with signaling is impor-
`tant.86 Since HER2 is a member of a family of
`growth factors known as
`the neuregulins/
`heregulin and is expressed in multiple neuronal
`and non-neuronal tissues in embryos and adult
`animals, including the heart, it is not surprising
`that cardiomyopathy has been associated with
`trastuzumab, particularly when combined with
`paclitaxel and anthracyclines.87–90
`EGFR is also overexpressed in many solid
`cancers, and when bound by its ligand, cell
`growth is stimulated. However, when engaged
`by an EGFR-specific antibody, receptor phos-
`
`Volume 56 Y Number 4 Y July/August 2006
`
`231
`
`IMMUNOGEN 2136, pg. 6
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`Targeted Therapy of Cancer
`
`TABLE 1 FDA-approved Antibodies for the Parenteral Use in Detection and Treatment of Cancer
`
`Generic Name
`
`Trade name
`
`Agent/Target
`
`Cancer Indication
`
`Approval
`
`Unconjugated
`Rituximab
`Trastuzumab
`Alemtuzumab
`Cetuximab
`
`Bevacizumab
`Radioconjugates
`Satumomab pendetide
`Nofetumomab merpentan
`Arcitumomab
`Capromab pendetide
`Ibritumomab tiuxetan
`Tositumomab
`Drug conjugates
`Gemtuzumab ozogamicin
`
`Rituxan
`Herceptin
`Campath-1H
`Erbitux
`
`Avastin
`
`OncoScint*
`Verluma*
`CEA-Scan*
`ProstaScint
`Zevalin
`Bexxar
`
`Mylotarg
`
`*No longer commercially available.
`†CLL ⫽ chronic lymphocytic leukemia.
`‡SCLC ⫽ small cell lung cancer.
`§AML ⫽ acute myelogenous leukemia.
`
`Chimeric anti-CD20 IgG1
`Humanized anti-HER2 IgG1
`Humanized anti-CD52
`Chimeric anti-EGFR
`
`Chimeric anti-VEGF
`
`111In-murine anti-TAG-72 IgG
`99mTc-murine anti-EGP-1 Fab’
`99mTc-murine anti-CEA Fab’
`111In-murine anti-PSMA
`90Y-murine anti-CD20 IgG ⫹ rituximab
`131I-murine anti-CD20 IgG ⫹ unlabeled tositumomab
`
`B-cell lymphoma
`Breast
`CLL†
`Colorectal
`Head/neck
`Colorectal
`
`Colorectal, ovarian
`SCLC‡
`Colorectal
`Prostate
`B-cell lymphoma
`B-cell lymphoma
`
`Humanized anti-CD33 IgG4 conjugated to colicheamicin
`
`AML§
`
`1997
`1998
`2001
`2004
`2006
`2004
`
`1992
`1996
`1996
`1996
`2002
`2003
`
`2000
`
`phorylation is decreased and cell growth is inhib-
`ited. The chimeric antibody against EGFR,
`cetuximab, also has an effect on neovasculariza-
`tion.91,92 Cetuximab works best in combination
`with chemotherapy in colorectal cancer,
`for
`which it was initially approved, and with external
`irradiation in head and neck cancers, which was
`recently FDA-approved.17,93 Beside the usual
`risks associated with antibody infusions, cetux-
`imab causes an acneform rash and other skin
`reactions in most patients, with 10% of these
`being severe. There is evidence suggesting that
`the intensity of the skin rash is associated with its
`antitumor response and even survival.94 Other
`EGFR antibodies, particularly humanized and
`fully human forms, also are in development, as
`indicated in Table 2, and may in fact be better
`tolerated and show evidence of activity without
`being combined with cytotoxic chemotherapy,
`which is currently being evaluated in Phase III
`trials. It is too early to speculate whether they
`will, in fact, provide any therapeutic advantages
`over cetuximab.
`Bevacizumab targets and blocks vascular en-
`dothelial growth factor (VEGF) and VEGF’s
`binding to its receptor on the vascular endo-
`thelium. Since VEGF is released by many
`cancers to stimulate proliferation of new blood
`vessels, the combination of bevacizumab and
`chemotherapy was found to increase objective
`
`responses, median time to progression, and sur-
`vival
`in patients with metastatic colorectal
`cancer, compared with chemotherapy alone,
`but earlier preclinical
`studies
`indicated that
`anti-VEGF antibodies were active alone, as
`well as in combination with radiation.22,95,96 It
`is currently being studied clinically in renal cell,
`breast, and lung cancers, as well as in a number
`of other nonhematological and hematological
`malignancies.97–99 As might be expected, bev-
`acizumab may cause gastrointestinal perfora-
`tions and delayed wound healing, as well as
`hemorrhagic events (primarily seen in small cell
`lung cancer trials, where bevacizumab is not
`approved). Arterial
`thromboembolic events
`(eg, cerebral infarction, transient ischemic at-
`tacks, myocardial infarction, angina) and pro-
`teinurea also have been reported.100
`
`IMMUNOCONJUGATES
`
`Antibodies also function as carriers of cyto-
`toxic substances, such as radioisotopes, drugs, and
`toxins (Figure 3). In NHL, anti-CD20 radiocon-
`jugates have superior antitumor activity com-
`pared with their unconjugated antibody coun-
`terparts, but there is increased, albeit manageable,
`hematological toxicity.101,102 These findings are
`
`232
`
`CA A Cancer Journal for Clinicians
`
`IMMUNOGEN 2136, pg. 7
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`TABLE 2 Selected Unconjugated Antibody Therapeutics in Advanced Clinical Testing
`
`Generic Name
`
`Agent/Target
`
`Apolizumab
`Chimeric 14.18
`Epratuzumab
`Galiximab
`HuMax-CD4
`Lumiliximab
`MDX-010
`Matuzumab
`Orgegovomab
`Panitumumab
`Pertuzumab
`Rencarex
`Vitaxin
`
`Human anti-HLA-DR
`Chimeric anti-ganglioside (GD2)
`Humanized anti-CD22
`Humanized anti-CD80
`Fully human anti-CD4
`Humanized anti-CD23 (Fc-epsilon RII)
`Anti-CTLA-4
`Humanized anti-EGFR
`Murine anti-CA-125
`Human anti-EGFR
`Humanized anti-HER-2
`Chimeric anti-G250
`Humanized anti-␣v␤3 integrin
`
`*CLL ⫽ chronic lymphocytic leukemia.
`†CRC ⫽ colorectal cancer.
`‡CTCL ⫽ cutaneous T-cell lymphoma.
`§NHL ⫽ non-Hodgkin lymphoma.
`¶NSCLC ⫽ non-small cell lung cancer.
`**SLL ⫽ small lymphocytic lymphomas.
`
`CA Cancer J Clin 2006;56:226–243
`
`Cancer
`
`CLL*, SLL**
`Neuroblastoma
`NHL§
`NHL§
`CTCL‡
`CLL*
`Melanoma
`CRC†
`Ovarian
`NSCLC¶, CRC†, renal
`Breast, prostate, ovarian
`Kidney
`Melanoma, prostate
`
`FIGURE 3 Immunoconjugates are formed primarily by chemical reactions. Radioconjugates can be formed by coupling
`radioiodine to tyrosine residues, or by binding chelates to lysine residues, which are then used to bind a variety of radio-
`metals, such as 90Y. Cysteine residues are also useful for coupling radionuclides, particularly 99mTc and rhenium, but
`cysteine is also used for conjugation of drugs and toxins, which can also be coupled to lysine residues. In addition, the
`carbohydrates found on IgG can be modified to allow the coupling of chelates or drugs. Drugs have also been coupled
`to an intermediate carrier that allows for a higher number of drugs to be bound to the antibody. Toxin conjugates usually
`need to be modified to remove their innate cell binding properties, with the biologically active portion then coupled to the
`antibody or used as a portion of a recombinant fusion protein.
`
`Volume 56 Y Number 4 Y July/August 2006
`
`233
`
`IMMUNOGEN 2136, pg. 8
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`Targeted Therapy of Cancer
`
`strong incentives to continue the pursuit of im-
`munoconjugates for cancer therapy.
`
`Radionuclides
`
`Radiolabeled antibodies were the first group
`of immunoconjugates to be examined.103 Ta-
`ble 3 lists some of the more commonly used
`radionuclides conjugated to antibodies
`for
`cancer treatment. Because the radioactivity can
`be detected easily by external scintigraphy, it is
`also noteworthy to mention the additional ap-
`plication of radiolabeled antibodies for imag-
`ing. The demonstration of cancer targeting
`with a radiolabeled antibody fragment to CEA
`resulted in the development of radiolabeled
`antibodies for cancer imaging.7 Since then,
`99mTc- and 111In-radioconjugates have been
`commonly used for this application, but with
`the advent of positron-emission tomography
`(PET), investigators are now beginning to take
`advantage of this technologically superior im-
`aging system by radiolabeling tumor-associated
`antibodies with positron-emitters.104 –107
`Whereas external beam radiation delivers a
`focused beam of high dose rate radiation for
`short bursts that are divided over several weeks
`and is designed to treat local disease, radioim-
`munotherapy (RAIT) is typically given as an
`intravenous injection, thereby allowing radio-
`activity to be delivered to tumors throughout
`the body. Tumor uptake of a radiolabeled IgG
`occurs gradually, taking 1 to 2 days before peak
`uptake occurs. Peak uptake is typically ⬍0.01%
`of the total injected dose per gram tumor, but
`the radioactivity deposited in the tumor can be
`detected several weeks later.108 Because of its
`kinetics, the radiation-absorbed dose delivered
`by RAIT occurs at a much lower dose rate than
`external beam irradiation, but is continually
`present for a period of time defined by the
`physical half-life of the radionuclide and the
`biological half-life of the antibody residing in
`the tumor. This continuous, low dose rate ra-
`diation exposure can be as effective as intermit-
`tent, high dose rate radiation.109,110
`When it comes to choices of radionuclides for
`therapy, tumor size is the primary consideration.
`Medium-energy beta-emitters, such as 131I (0.5
`MeV) and 177Lu (0.8 MeV), can traverse 1.0 mm,
`
`while high-energy beta-emitters, such as 90Y or
`188Re (2.1 MeV), can penetrate up to 11 mm,
`making it possible for beta-emitters to kill across
`several hundred cells, referred to earlier as a by-
`stander or crossfire effect.111 This is considered a
`significant attribute for radioconjugates compared
`with other immunoconjugates, since they can be
`therapeutically active even if heterogeneous anti-
`gen expression, tumor architecture, or other fac-
`tors impede targeting of every cell. Although
`higher energy beta-emitters have the potential of
`killing cells across a longer path-length, the ab-
`sorbed fraction is higher for the lower energy
`beta-emitters (ie, probability of hitting the nu-
`clear DNA), making them efficient killers. Alpha-
`emitters, such as 213Bi and 211At, traverse only a
`few cell diameters, but an alpha particle is also a
`far more efficient (energetic) killer than even a
`low-energy beta particle, requiring fewer “hits”
`to damage cellular processes.111 Low-energy
`electrons, such as are produced by Auger emitters
`(125I, 67Ga, or 111In, for example) have to be in
`close contact, preferably inside a cell or in the
`nucleus, to exert a cytotoxic effect. As one might
`expect, beta-emitters are most likely best applied
`in situations where the tumors are ⱖ0.5 cm in
`diameter; otherwise a substantial portion of the
`energy from the radioactive decay will be ab-
`sorbed in the surrounding normal tissue. The
`alpha and low-energy electron emitters are best
`applied when the disease burden is smaller, more
`localized, or where there may be single or small
`clusters of cells (eg,
`leukemia, malignant as-
`cites).112,113
`The primary concern for using radionuclide-
`labeled IgG is that it remains in the blood for an
`extended period of time, which continually exposes
`the highly sensitive red marrow to radiation, result-
`ing in dose-limiting myelosuppression. Smaller
`forms of the antibodies, such as a F(ab=)2 or Fab’, and
`more recently, molecularly engineered antibody
`subfragments (Figure 1) with more favorable phar-
`macokinetic properties, are removed more rapidly
`from the blood, thereby improving tumor/blood
`ratios.114,115 There have been reports of improved
`therapeutic responses using smaller-sized antibodies,
`but these smaller entities frequently are cleared from
`the blood by renal filtration, and as a result, many
`radionuclides (eg, radiometals) become trapped in a
`higher concentration in the kidneys than in the
`
`234
`
`CA A Cancer Journal for Clinicians
`
`IMMUNOGEN 2136, pg. 9
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`CA Cancer J Clin 2006;56:226–243
`
`TABLE 3 Physical Properties of Several Examples of Radionuclides Used for Radioconjugate Therapy
`
`Radionuclide
`
`Emission
`
`Half

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