throbber
NIH Public Access
`Author Manuscript
`Hematol Oncol Clin North Am. Author manuscript; available in PMC 2013 June 01.
`Published in final edited form as:
`Hematol Oncol Clin North Am. 2012 June ; 26(3): 447–481. doi:10.1016/j.hoc.2012.02.013.
`
`Therapeutic antibodies against cancer
`
`Mark J. Adler and Dimiter S. Dimitrov
`Protein Interactions Group, Center for Cancer Research Nanobiology Program, National Cancer
`Institute, National Institutes of Health, Frederick, Maryland 21702
`
`Abstract
`Antibody-based therapeutics against cancer are highly successful in clinic and currently enjoy
`unprecedented recognition of their potential; 13 monoclonal antibodies (mAbs) have been
`approved for clinical use in the European Union and in the United States (one, mylotarg, was
`withdrawn from market in 2010). Three of the mAbs (bevacizumab, rituximab, trastuzumab) are
`in the top six selling protein therapeutics with sales in 2010 of more than $5 bln each. Hundreds of
`mAbs including bispecific mAbs and multispecific fusion proteins, mAbs conjugated with small
`molecule drugs and mAbs with optimized pharmacokinetics are in clinical trials. However,
`challenges remain and it appears that deeper understanding of mechanisms is needed to overcome
`major problems including resistance to therapy, access to targets, complexity of biological systems
`and individual variations.
`
`Keywords
`therapeutics; antibodies; cancer; immunogenicity; safety; efficacy
`
`1. Introduction
`Antibody therapy has its roots thousands of years ago; early forms of vaccination against
`infectious diseases were developed in China as early as 200 BC. However, the history of
`true antibody therapy began much more recently with the discovery that serum from animals
`immunized with toxins, for example, diphtheria toxin or viruses, is an effective therapeutic
`against the disease caused by the same agent in humans. This discovery resulted in the
`development of the serum therapy which saved thousands of lives; von Behring who in the
`1880s developed an antitoxin that did not kill the bacteria, but neutralized the toxin that the
`bacteria release into the body was awarded the first Nobel Prize in Medicine in 1901 for his
`role in the discovery and development of a serum therapy for diphtheria. Interestingly,
`although historically successes of antibody (serum) therapy were initially mostly in the
`treatment of patients with infectious diseases currently there is only on monoclonal antibody
`(mAb) approved for treatment of any infectious disease (synagis) and it is for prevention of
`the infection not for therapy of already established infection. Initial attempts to treat cancer
`patients with serum therapy were not successful. It was not until several decades ago when a
`
`© 2012 Elsevier Inc. All rights reserved
`Address for correspondence Mark J. Adler, MD UC San Diego Cancer Center 1200 Garden View Encinitas, California 92024
`oncologic@gmail.com. Dimiter S. Dimitrov, PhD, ScD Protein Interactions Group Center for Cancer Research Nanobiology Program
`National Cancer Institute, National Institutes of Health Bldg 469, Rm 150B Frederick, Maryland 21702 Tel: (301) 846-1352 Fax:
`(301) 846-5598 dimiter.dimitrov@nih.gov.
`Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our
`customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of
`the resulting proof before it is published in its final citable form. Please note that during the production process errors may be
`discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`GENENTECH 2001
`GENZYME V. GENENTECH
`IPR2016-00383
`
`

`

`Adler and Dimitrov
`
`Page 2
`
`number of revolutionary scientific discoveries were made that allowed the development of
`recombinant therapeutic resulting in the approval of the first anti-cancer therapeutic
`antibody – mAb rituximab in 1997 (Table 1). Since than 13 mAbs have been approved for
`clinical use against cancer in the European Union and the United States and 12 are on the
`market in August 2011; one of them, Gemtuzumab ozogamicin (Mylotarg), was withdrawn
`(Table 1); in contrast we still have to wait for the first approved mAb-based therapeutic
`against an infectious disease (synagis is for prevention). In 2010 sales of the top four
`recombinant therapeutic antibodies (bevacizumab, rituximab, trastuzumab, cetuximab)
`exceeded US$ 20 bln (Table 2).).
`
`Dating back to mummies and up to the recent successes with ipilimumab it has become
`axiomatic that the human immune system has an inherent capacity for anti-tumor activity.
`This was bolstered in the 1900s by the finding of spontaneous remissions recorded—often in
`sparse anectodal findings-- in nearly ever stage and form of cancer, by the more common
`observation of spontaneous regressions of melanoma and renal carcinoma, the success of
`non-specific immune-stimulants such as BCG or Coley's toxin and the increasingly targeted
`use of antibodies against antigens more specific to certain cell types [1]. Indeed, the
`antibody specificity was perhaps the first and still the most powerful story supporting the
`ubiquitous catch-call of personalized medicine.
`
`With all of the elegance of the specificity story and more than 35 years since Kohler and
`Milstein's recipe for generating monoclonal antibodies [2], the clinical promise has been
`largely disappointing. With rare exceptions, these molecular missiles have not annihilated
`their target tumors and have fallen far short of the marvel of the antibiotic revolution. The
`rarity of cures should not dampen the substantial, if incremental, progress that has been
`made. Even in the age of single nucleotide etiologies there is a strong case that cancer, by
`the time of its clinical visibility, consists of many broken parts; hence the growing argument
`that targeted therapies may parallel the breakthrough to cure with chemotherapy in the
`1970's with the move to, not one, but a cocktail of simultaneous, combined agents. As in the
`case of combination chemotherapy, antibody therapy may come to utilize different effector
`pathways in this assault.
`
`Therapeutic mAbs and other therapeutic proteins have been reviewed previously (see recent
`reviews [3–15] and articles cited there). Therefore, here, we review the monoclonal
`antibodies used directly in treatment, shed some light on presumed primary mechanism of
`action, and survey use—from initial indication to the wider adoption based principally on
`clinical trials and trends. This line-up, with its wide spectrum of targets and mechanisms
`may give some hope yet that the long trek may yet reach the originally envisioned summit.
`If not, these agents are undoubtedly part of the solution. We focus mainly on those native,
`unconjugated antibodies that directly impact solid tumors. Bevacizumab, though its anti-
`vascular action is indirect, has gained such wide application for solid tumors (and been
`subject of much controversy) that it seemed important to include. Finally, while immune-
`conjugates have been well reviewed elsewhere [16–18] and not the present focus
`brentuximab vedontin, as the first new indication for Hodgkin's in 30 years warranted
`special inclusion. Its success represents a partial rescue of a paradigm after the first
`approved antibody-drug conjugate, gemtuzumab was withdrawn in 2010 due to lack of
`efficacy and increased deaths [19]. In the context of the present review it may also point to
`some limiting aspects in unconjugated tumor-directed antibodies, which as has been stated,
`have not delivered their quarter-century promise.
`
`Hematol Oncol Clin North Am. Author manuscript; available in PMC 2013 June 01.
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`

`

`Adler and Dimitrov
`
`Page 3
`
`2. mAbs approved for clinical use
`Currently, (as of August 2011) 13 mAbs are approved for clinical use in the European Union
`(EU) or United States (US) (Table 1). One of the approved mAbs, gemtuzumab ozogamicin
`(Mylotarg) was withdrawn from the market because of lack of clinical benefit and safety
`reasons after a clinical trial in which a greater number of deaths occurred in the group of
`patients with acute myeloid leukemia (AML) who received Mylotarg compared with those
`receiving chemotherapy alone. Mylotarg as well as removab which is not approved in the
`USA yet, and the two radiotherapeutic mAbs, Bexxar and Zevalin, will not be reviewed
`here.
`
`2.1 Rituximab
`The first candidate out of the starting box remains in many ways the poster child for both
`specificity and efficacy. Rituxamab (MabThera, Rituxan), initially developed in San Diego
`in the late 1980's, and father to that regions biotech explosion, was based upon the finding of
`CD20 antigen on normal and malignant lymphocytes; it is not appreciably expressed at
`either pole of lymphocyte ontogeny--stem cells and plasma cells--nor on other non-
`lymphoid cellular compartments. In contrast to many emerging cancer targets clearly
`connected with signal transduction circuitry there is no clear consensus on the function of
`CD20. Nonetheless, the chosen antigen-antibody duo in CD20/rituximab rendered a striking
`clinical success and ushered in a continuing wave of similarly conceived agents albeit with
`variant tactical goals and mechanisms of effect. It is interesting to note that only after many
`years afterward were clinical agents developed to target perhaps the ultimate tissue-specific
`bull's eye: the individual epitope of each B lymphocyte population—separating the
`malignant fiend from over a million brethren lymphocytes by one signature antigen
`expressed on one malignant subspecies.
`
`In 1997 rituximab was approved by the US FDA for treatment of relapsed indolent B-cell
`non-Hodgkin's lymphoma. The antibody is a mouse-human chimera utilizing murine
`variable regions to effect anti-CD20 specificity and human IgG1k constant region to
`facilitate effector function including complement mediated lysis and antibody directed
`cellular cytotoxicity [20, 21]. Additional mechanisms include caspase activation [22] a
`“vaccinal effect” based upon increased idiotype-specific T cell response to follicular
`lymphoma [23], and upregulation of proapoptotic proteins such as Bax [24, 25].
`
`Its well-known, early recognized and sometimes fatal chief toxicity has been acute infusion
`reactions. Rare fatalities, occurring mainly during first infusion, have been considered
`secondary to a cytokine reaction; generally associated with flu-like symptoms they may
`progress to life threatening hypotension, bronchospasm and hypoxia, but can usually be
`controlled by stopping or adjusting of rates of infusion and proper premedication [26].
`Blackbox events include tumor lysis syndrome, severe mucocutaneous reactions and
`progressive multifocal leukoencephalopathy (PML) resulting in death [27, 28].
`
`Rituxumab has demonstrated clinical activity across the spectrum of lymphoproliferative
`disorders but the greatest impact has been in non-Hodgkin's lymphoma, where combinations
`and optimizations, have sought to raise response rates and ultimately cure. Since its 1997
`start with relapsed indolent non-Hodgkin's lymphoma (NHL), rituximab has obtained the
`following additional indications for lymphoma per package insert: relapsed and refractory,
`follicular or low-grade, CD20-positive, B-cell NHL as single agent; previously untreated
`CD20-positive, follicular, B-cell NHL in combination with first line chemotherapy; as single
`agent maintenance therapy for patients achieving a partial or complete response to rituximab
`in combination with chemotherapy; for non-progressing (including stable), CD20 positive,
`low-grade, B-cell NHL, as a single agent after first-line combination of cyclophosphamide,
`
`Hematol Oncol Clin North Am. Author manuscript; available in PMC 2013 June 01.
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`

`

`Adler and Dimitrov
`
`Page 4
`
`vincristine, and prednisone (CVP) chemotherapy; previously untreated CD20 positive,
`diffuse large B-cell NHL in combination with anthracycline-based chemotherapy, for
`example, in the workhorse, R-CHOP [29]. It also has an oft-used indication for treatment of
`previously treated or untreated patients with CD20 chronic lymphocytic leukemia (CLL) in
`combination with fludarabine and cyclophosphamide (FC) [30].
`
`It has found off-label use in the clinic in all or nearly all malignant (and many non-
`malignant) settings where B-cells are presumed to participate in pathogenesis and been the
`subject of many scholarly reviews. Common use spans from aggressive to low grade
`lymphoproliferative disorders including: combination with chemotherapy for induction in
`second line therapy for relapsed lymphoma anticipating autologous transplant [31];
`combination with chlorambucil for indolent and with bendamustine in treatment of relapsed
`or refractory CLL [32]; induction for Burkitt's, use for gastric and non-gastric mucosa-
`associated lymphoid tissue (MALT tumors [33, 34], Mantle cell tumor [35], primary
`cutaneous B-cell [36], splenic marginal zone NHL [37] Waldenström's macroglobulinemia/
`lymphoplasmacytic lymphoma [38]. Its uses have been tailored to mutational status of
`del(17p) and del(11q) with refractory CLL (National Comprehensive Cancer Network
`(NCCN) guidelines - http://www.nccn.org/index.asp) and combined in “cocktail” with other
`antibodies such as alemtuzumab for refractory lymphoid malignancies.
`
`The evolution of treatment for CLL mirrors, in many ways, that of NHL as it leads from
`purines to chemo-immunotherapy and most recently to novel antiCD20 antibodies.
`Conventional treatment of CLL evolved from alkylators to purine analogues when it was
`demonstrated that fludarabine (F) yielded greater efficacy with better complete response
`(CR), progression-free and overall survival (PFS and OS) rates than chlorambucil as primary
`therapy [39]. Subsequently, the combination of fludarabine with cyclophosphamide (FC)
`showed better CR and PFS than F [40]. Based upon the activity of rituximab (R) alone as a
`front line agent, it was added to FC and compared to FC alone; in a phase III randomized
`trial the combination FCR demonstrated better OR, CR, and PFS, establishing both the
`regimen and the concept of chemo-immunotherapy in this setting as the upfront standard of
`care [41].
`
`2.2 Ofatumumab
`Unfortunately, the activity of rituximab as a single agent is only modest [42] and duration of
`response in relapsed disease is generally measured in months [43]. This was part of the
`impetus to develop newer anti-CD20 targeted antibodies with a goal to improve such
`characteristics as binding affinity, specificity and effector function, and efficacy [44].
`Ofatumumab (ofa), a fully human monoclonal IgG1 binds to a unique epitope [45], induces
`considerably higher complement dependent cytotoxicity (CDC) than rituximab [46] and
`shows activity in rituxan-refractory B cell lymphoma [47].
`
`On the basis of these potential biological advantages and modest early phase clinical activity
`[48] ofa was tested against CLL which was either refractory to fludarabine and alemtuzumab
`or refractory to fludarabine with disease considered too bulky for efficacy with
`alemtuzumab [49]. The drug was well tolerated, though complicated by infections in 25% of
`the patients, but the impressive clinical results including median OS of 13.7 or 15.4 months,
`within two high risk groups, respectively, contributed to the approval of ofa for disease
`refractory to fludara and for those who have failed alemtuzumab [50, 51].
`
`Given the potential advantages of ofa versus rituximab and FCR established as standard of
`care in front line, substituting ofa for rituxan in the so-call O-FC regimen was tested in a
`multinational, randomized phase II trial in treatment naïve patients [52]. Of the two tested
`doses, the higher dose arm yielded a CR rate of 50%. It remains unclear how to position this
`
`Hematol Oncol Clin North Am. Author manuscript; available in PMC 2013 June 01.
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`

`

`Adler and Dimitrov
`
`Page 5
`
`with respect to such other findings as the initial randomized phase III trial that established
`FCR as standard of care. The precedent of combining permutations of purine analogues,
`alkylators and antibodies including newer regimens like Ofa/bendamustine continues to
`inform ongoing studies [53].
`
`2.3 Ipilimumab
`The novel treatment agents for melanoma, vemurafenib (b-raf inhibitor) and ipilimumab (an
`antibody against cytotoxic T lympocyte antigen 4 (CTLA-4)), represent perhaps the most
`significant advance in oncology in several years. How they will fit in tactical treatment
`strategies, and with respect to conventional dacarbazine, IL-2, and a new gp100 based
`vaccine is a welcome and exciting challenge after decades without appreciable progress
`[54]. Blockade of the CTLA-4 has been the subject of long and intensive investigation [55,
`56].
`
`Among the most active immune inhibitory pathways is the CD28/CTLA-4:B7-1/B7-2
`receptor/ligand grouping which modulate peripheral tolerance to tumors and outgrowth of
`immune-evasive clones. Inhibition is both toward the overexpressed self targets via
`upregulation of inhibitory ligands on lymphocytes. Thus blockade of CTLA-4 has potential
`for both mono-therapy and in synergy with other therapies that enhance presentation of
`tumor epitopes to the immune system [56]. Genetic ablation of CTLA-4 leads to a massive
`and lethal lympho-proliferative disorder [57]. Antibody blockade of CTLA-4 induces potent
`anti-tumor activity through enhancing effector cells and concomitantly inhibiting T
`regulatory activity [58].
`
`Given that this inhibition is not tumor-specific it is not surprising that other tumors including
`ovarian cancer, prostate cancer, and renal cell cancer have demonstrated durable remissions
`[59].
`
`In a recent phase III trial, patients with melanoma refractory to chemotherapy or IL-2 who
`received ipilimumab had improved overall survival compared to those receiving the gp100
`peptide vaccine, and on this basis received FDA approval in 2011 [60].
`
`Ipilimumab holds an FDA indication for the treatment of unresectable or metastatic
`melanoma,with NCCN guidelines that largely elucidate specific contexts consistent with this
`approval including use as single agent for unresectable stage III in-transit metastases, local/
`satellite and/or in-transit unresectable recurrence, incompletely resected nodal recurrence,
`limited recurrence or metastatic disease, and disseminated recurrence or metastatic disease
`in patients with good performance status.
`
`Based upon its mechanism of unleashing the immune recognition and effector system there
`was rationale to test the interactive effects with tumor specific antigen. Specifically, the
`melanoma antigen, gp100, overexpressed on this tumor and among the antigens presented in
`the appropriate genetic major histocompatibility complex (MHC) context (HLA*A201)
`represented a prime vaccine candidate. In a phase III randomized trial increased response
`rates were seen when vaccine was added to IL-2 compared to IL-2 alone (16% versus 6%,
`P=0.03); progression free survival was also significantly improved with a trend toward
`improved overall survival [61]. Questions arose, nonetheless, whether gp100 vaccine was an
`appropriate control in the aforementioned phase III trial for ipilimumab. Another phase III
`randomized clinical trial treating previously untreated patients with metastatic melanoma
`compared ipilimumab (every 3 weeks for four doses followed by `maintenance' every three
`months) with and without dacarbazine as the standard control; improved OS was seen
`including a difference at 3 years of nearly 21% vs. 12% [62].
`
`Hematol Oncol Clin North Am. Author manuscript; available in PMC 2013 June 01.
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`

`

`Adler and Dimitrov
`
`Page 6
`
`The cluster of well-identified side effects induced by CTLA-4 inhibition have been referred
`to as “immune-related adverse events” (IRAEs). These unique adverse effects are likely a
`direct effect of impairing immune tolerance. They include colitis/diarrhea, dermatitis,
`hepatitis, uveitis, nephritis, inflammatory myopathy and endocrinopathies. While these
`reactions have gained a blackbox designation for occasional severe and even fatal instances
`they are generally manageable and reversible with treatment guidelines which include
`systemic corticosteroids [63]. These toxicities may be prolonged, suggestive of sustained
`release from immune tolerance, and perhaps a different response profile including long
`periodsof stable disease, and correlation of toxicity with efficacy. In one report with high-
`risk melanoma ipilimumab-treated patients who experienced high grade IRAEs had a
`significantly higher rate of tumor regression than those without IRAEs (36% versus 5% of
`patients) [64].
`
`Based upon a mechanism of action clearly different from IL-2, which increases
`responsiveness to immune targets, and is non-overlapping with chemotherapy, earlier phase
`trials and future efforts will focus upon combinations of vaccines, chemo, and other
`immune-modulators [59]. Furthermore, given the prolonged time course of side effects and
`the resulting requirement for prolonged steroids, timing of its use with respect to IL-2 and
`vaccines will be the subject of much attention [65].
`
`2.4 Trastuzumab
`HER2 is overexpressed in 20–30 % of invasive breast cancers and associated with a worse
`prognosis [66]. Trastuzumab is a humanized monoclonal antibody targeting the human
`epidermal growth factor receptor 2 (HER2) which was approved by the FDA in 1998 as the
`first monoclonal for a solid tumor indicated for patients with invasive breast cancer that
`overexpresses HER2. It is now a standard part of treatment for HER2 positive tumors in
`both metastatic and adjuvant settings. Since, across the range of studies, tumors that
`overexpress HER2 receptor respond better, considerable effort has been expended to
`accurately assess receptor status [67–69].
`
`HER2 is part of a family of transmembrane tyrosine kinase receptors which normally
`regulate cell growth and survival, differentiation and migration [70]. It consists of an
`extracellular binding domain, a transmembrane segment and an intracellular tyrosine kinase
`domain. The receptor is activated by homo- or hetero- dimerization generally, but not
`always activated through ligand binding; it can dimerize and thus activate, independent of
`ligand [71] through either overexpression or mutation [72]. Thus activated by
`overexpression, signal-transduction cascades act to promote a host of pro-growth activities
`including proliferation, survival, and invasion. Such signal transduction is mediated through
`the RAS-MAKP pathway, inhibiting cell death through the m-TOR pathway [73].
`Additionally, it inhibits P13K pathway, reducing PTEN phosphorylation and AKT
`dephosphorylation and thus increasing cell death [74] [75].
`
`The human IgG1 is capable of inducing antibody dependent cell-mediated cytotoxicity
`(ADCC) in vitro [76] and of recruitment of effector cells in animal studies [77]. An immune
`mechanism is suggested by the increased lymphoid infiltration into tumor after preoperative
`administration of trastuzumab [78]. There is also evidence that it causes regression of
`vasculature by modulating angiogenic factors [79].
`
`As a single agent in metastatic breast cancer, and receptor status using earlier
`immunohistochemistry (IHC) expression criteria, trastuzumab produced response rates of 11
`to 26% [80]. From the earliest studies, though time has sharpened the assessment, it has
`been clear that the best results occur in tumors that overexpress Her2. The breakthrough trial
`for trastuzumab in metatstatic disease came in a randomized phase III trial when it was used
`
`Hematol Oncol Clin North Am. Author manuscript; available in PMC 2013 June 01.
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`

`

`Adler and Dimitrov
`
`Page 7
`
`in combination with chemotherapy for Her-2 positive patients [81]. As first line therapy for
`metastatic disease patients were given either chemo alone or in combination. Patients were
`given an anthracycline and cyclophosphamide or, paclitaxel (if they had previous
`anthracycline in adjuvant settting). Results showed not only improvement in response rate
`(RR) and progression free interval but in overall survival. Trastuzumab was subsequently
`showed to have efficacy and safety with a variety of other chemotherapeutics including
`docetaxel [82], vinorelbine [83], and doxil [84] in nonrandomized trials.
`
`As for the adjuvant setting, large randomized trials established significant benefits from the
`addition of trastuzumab to both anthracycline and non-anthracycline early breast cancer
`[85]. Four major adjuvant trials including >13,000 women with HER2-positive early breast
`cancer utilized different adjuvant regimens with trastuzumab; in these studies overall,
`trastuzumab reduced the 3-year risk of recurrence by about half in this population [86]. On
`this basis, trastuzumab has become part of standard adjuvant therapy. Both the international
`Consensus Group and NCCN recommend its use for women with HER2 positive, node
`positive tumors as well as for node negative disease when the primary is >1cm.
`
`Trastuzumab, combined with chemotherapy has also shown improvement in pathological
`responses and event free survival when used in the neoadjuvant setting prior to surgery [87].
`In a randomized phase III trial patients with advanced gastroesophageal and gastric
`adenocarcinoma tumors which overexpressed HER2 showed significant increase in overall
`survival when trastuzumab was added to their chemotherapy [88]. Trastuzumab now has an
`FDA indication for use in combination with cisplatinum and fluorouracil (5FU) or
`capecitabine for first line treatment of gastric and gastroesophageal tumor which
`overexpress HER 2.
`
`The most significant toxicity associated with trastuzumab is cardiomyopathy ranging from
`subclinical decreases in left ventricular ejection fraction (LVEF) to cardiac failure
`manifesting as congestive heart failure (CHF). The risk is greatest when administered
`concurrently with anthracyclines [81]. Use following anthracyclines was associated
`commonly with asymptomatic cardiac dysfunction but most severe decreases recovered with
`time [89]. Close monitoring of clinical status and cardiac function, sequential rather than
`concomitant use, and development of non-anthracycline regimens [90] [91] have all been
`objectives.
`
`2.5 Bevacizumab
`The discussion of bevacizumab (bev) here will be asymmetric in bulk and breadth compared
`to the other antibodies owing to its conceptual and actual application in many tumor types,
`it's unique mechanism and toxicity profile. Bevacizumab is a humanized IgG1 mAb that
`binds to and neutralizes the ligand vascular endothelial growth factor (VEGF) rather than
`binding the cell surface receptor. In fact many tissues and most malignancies produce VEGF
`whose native function, whether acting from a distance or in an autocrine loop, operates
`through binding and activation of the VEGF receptor [91]. The latter includes an
`extracellular binding domain and a cytoplasmic kinase domain. Following VEGF binding,
`the otherwise inactive monomer receptor undergoes dimerization, autophosphorylation of
`the tyrosisne kinase domain and downstream activation of many of the usual signal
`transduction suspects including MAPK and protein kinase C pathways which mediate
`proliferative events - in this setting, endothelial proliferation and angiogenesis [92]; such
`neoangiogensis is required by tumors once they grow greater than 2 mm [93].
`
`Many of this antibody's common toxicities are related to its impact on microvasculature
`including hypertension, proteinuria, rare bowel perforation, impaired wound healing and
`bleeding [94]. Other than the rare bowel perforation these can generally be managed and
`
`Hematol Oncol Clin North Am. Author manuscript; available in PMC 2013 June 01.
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`

`

`Adler and Dimitrov
`
`Page 8
`
`without necessitating cessation of therapy. While there will naturally be some specificity of
`side effects and adverse reactions dependent upon the drugs with which bevacizumab is
`paired, with some notable exceptions, toxicities are generally neither drug combination nor
`tumor specific.
`
`More severe and fatal consequences of bevacizumab have been the subject of a number of
`meta-analyses and reports of large institution experience. In perhaps the largest of these,
`fatal adverse events (FAEs) were considered in a meta-analysis of over 10,000 patients with
`various solid tumor types, comparing regimens with and without the addition of
`bevacizumab. The overall incidences of fatal adverse events were 2.5% and among these the
`highest percentages were nearly a quarter attributable to hemorrhage, about half of that
`related to neutropenia, and a smaller amount to perforation. There was increased relative risk
`attributable to combining bevacizumab with taxanes or platinum but not with other agents,
`nor were there significant tumor-specific increases. In another large meta-analysis
`bevacizumab was associated with high-grade congestive heart failure in breast cancer with
`an overall incidence of 1.6% [95]. Yet a third large meta-analysis identified a 12% risk of
`thromboembolic events [96]. Of note, a pooled analysis of phase II and phase III trials did
`not show an increase in venous thromboembolic events--important to recognize with a
`baseline of tumor--associated venous thromboembolic events (VTEs) of around 10% with or
`without this agent [97]. Massive hemoptysis has been linked to large central lesions at risk
`for cavitation [98], and avoided in these circumstances, and more generally in squamous
`cancer where this risk is increased. Bowel perforation occurred with an incidence under 2%
`in a large institution with a treated population of over 1400 patients; it was generally
`managed without the need for surgical intervention [99].
`
`Bevacizumab demonstrated nil [100] to small [101] response rates as mono-therapy and,
`with such exceptions as maintenance regimens and single agent use with recurrent
`glioblastoma, its predominant clinical role lies in combination with chemotherapy. In 2004,
`based upon improvement of response rates progression-free survival, and overall survival,
`bevacizumab, when combined with chemotherapy in metastatic colorectal cancer [102],
`became the first anti-angiogenic agent approved for clinical use. Since then it has gained
`indications for metastatic breast, metastatic renal cancer, metastatic (as well as advanced or
`recurrent) non-small cell lung cancer, and glioblastoma. Increasing use of bevacizumab is
`also being seen with hepatocellular and ovarian cancer.
`
`2.5.1 Colorectal cancer—At this time bevacizumab has an indication in metastatic
`colorectal cancer in both first and second line settings. The initial approval followed its use
`with bolus irinotecan, fluorouracil, and leucovorin (IFL) where addition of bevacizumab
`significantly improved response rate and median survival (20 versus 16 months) compared
`to chemo only [102]. While bolus IFL has fallen out of general use due to its toxicity profile,
`studies have supported the value of bevacizumab in combination with more widely used
`treatments including FOLFIRI (FOL – leucovorin plus F – fluorouracil (5-FU) and IRI –
`irinotecan (Camptosar)) [103, 104], and three oxaliplatin-containing regimens [105]. In
`addition, when bevacizumab was added to 5FU/leucovorin in the absence of irinotecan or
`oxaliplatin, response rates were approximately doubled and median survival improved
`compared to chemo alone [106, 107].
`
`Efforts to apply bevacizumab in the adjuvant setting for colorectal cancer moved from initial
`enthusiasm to disappointment. As noted above, bevacizumab had shown favorable impact in
`metastatic disease in a number of settings including in combination with IFL (irinotecan,
`5FU and leucovorin) for metastatic colorectal cancer. Borrowing the prevailing paradigm for
`chemotherapy--which attempts to apply results in metastatic disease to adjuvant use on the
`presumption of potential elimination of micro-metastases - bevacizumab was studied in the
`
`Hematol Oncol Clin North Am. Author manuscript; available in PMC 2013 June 01.
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`

`

`Adler and Dimitrov
`
`Page 9
`
`adjuvant setting for colorectal cancer. Two recently publish

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