Antibody Therapy Brandon G Smaglo, Dalal Aldeghaither, and Louis M Weiner, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA Ó 2016 Elsevier Ltd. All rights reserved.
Abstract Antibody therapy has emerged as an important weapon in the anticancer armament. These therapies take advantage of their ability to target proteins uniquely expressed on the surface of cancer cells. A number of monoclonal antibodies, which target a specific tumor cell surface protein and in doing so result in cellular destruction, have been approved for clinical use, and many more continue to be developed. Expanding upon monoclonal antibody therapy are immunoconjugates, which deliver a therapeutic entity to the cancer cell with high precision by linking it to the antibody. Cancer antibody therapy may also target the immune system, preventing its natural downregulation and thus augmenting the body’s natural immune response against the cancer. An important component of all of these therapies is the design of antibodies that specifically target specific cancer epitopes, and much of the ongoing research into cancer antibody therapy is focused on the engineering antibodies that bind with high specificity.
Antibodies Targeting Cancer Antibodies Already Approved Antibodies are proteins secreted by B cells in response to antigens. The basic building blocks of the antibody structure are the heavy and light chains that together shape two functional units: the constant region (Fc) and the fragment of antigen-binding region (Fab) (Weiner et al., 2010). The Fab domain is composed of three hypervariable complementarity-determining regions (CDRs) which serve as determinants of antibody specificity and antigen recognition (Weiner et al., 2010). The Fc domain acts as a link between the Fab’s capacity to bind antigens and immune system activation and recruit effector cells by engaging their Fcg. Antibodies have five structural categories: IgM, IgG, IgA, IgE, and IgD, based on their heavy chain compositions and Fc regions. IgG is the most commonly used antibody structure for monoclonal antibody therapy.
Monoclonal Antibodies Monoclonal antibodies were first produced in 1975 using the then newly developed hybridoma technology (Köhler and Milstein, 1975). Their murine origin resulted in immunogenicity in humans and accompanying inability to induce robust human immune effector responses against the targeted tumor antigens and cancer cells, limiting their clinical application (Shawler et al., 1985; Khazaeli et al., 1994). Scientific and technical advancements have enabled the design and development of chimeric antibodies, composed of murine variable regions transferred onto human Fc regions, humanized antibodies, with murine CDRs grafted onto a human antibody scaffold, and fully human antibodies derived from xenogeneic mice or by recombinant human antibody display technologies (Robinson et al., 2004). The modified structures constitute improvements in target binding, effector potency, and reduced immunogenicity. These modified antibody structures now dominate the human antibody therapy landscape. To date, 14 conjugated and unconjugated mAbs have been FDA approved for the treatment of cancer (Table 1).
550
The chimeric monoclonal antibody rituximab targets and destroys B cells by binding the surface protein CD20. One of the first monoclonal antibodies to enter into clinical practice, rituximab is generally well-tolerated and routinely used as a part of systemic treatment for a number of B cell leukemias and lymphomas, in both first and refractory lines of therapy (Marcus et al., 2005; Coiffier et al., 1998; Coiffier et al., 2002; Tam et al., 2008; Pfreundschuh et al., 2011). Ofatumumab, a humanized monoclonal antibody, also targets and binds CD20 and, in doing so, appears to inhibit early-stage activation of the B cell lymphocytes. In the treatment of chemotherapyrefractory CLL, ofatumumab was found to be active and welltolerated (Wierda et al., 2010). Ofatumumab is being explored in the first line treatment of CLL and also in other B cell malignancies, although questions of toxicity may limit its development in all clinical settings, and it is generally reserved for the treatment of rituximab-refractory B cell malignancies (Shanafelt et al., 2013; Czuczman et al., 2012; Ujjani et al., 2015). Alemtuzumab is a humanized monoclonal antibody that targets CD52; this surface protein is highly expressed on lymphocytes. A 42% reduction in risk of disease progression or death was reported when alemtuzumab was used in firstline therapy for patients with CLL, compared to standard therapy with chlorambucil (Hillmen et al., 2007). Presently, alemtuzumab is not available for clinical use, though it may be developed for future use in other diseases. Cetuximab, a chimeric monoclonal antibody, and panitumumab, a fully humanized monoclonal antibody, both target the epidermal growth factor receptor (EGFR). EGFR is overexpressed on the surface of some cancer cells. When ligands bind to EGFR, several intracellular protein pathways are stimulated that ultimately result in cellular growth and proliferation. In metastatic colorectal cancer, cetuximab and panitumumab have demonstrated benefit when added to standard chemotherapy regimens (Van Cutsem et al., 2009; Bokemeyer et al., 2009). Importantly, in colorectal cancer, these antibodies are only effective in patients with tumors that do not possess mutations in the Ras protein; Ras mutations result in
Encyclopedia of Immunobiology, Volume 4
http://dx.doi.org/10.1016/B978-0-12-374279-7.17021-3
Tumor Immunology j Antibody Therapy
551
Table 1 Monoclonal antibodies approved for use, as identified by target and cancer type, with the studies that support their use in these diseases Monoclonal antibody
Target
Disease type
References
Rituximab
CD20
B cell lymphoma, CLL
Ofatumumab
CD20
B cell lymphoma, CLL
Alemtuzumab Cetuximab
CD52 EGFR
Panitumumab Trastuzumab
EGFR HER2
Ziv-aflibercept Bevacizumab
VEGF VEGF
Ramucirumab
VEGF-R2
B cell malignancies Colorectal cancer Head/neck squamous cell cancer Nonsmall cell lung cancer Colorectal cancer Breast cancer Gastric cancer Colorectal cancer Colorectal cancer Nonsmall cell lung cancer Glioblastoma Gastric cancer Colon cancer, lung cancer
Marcus et al. (2005), Coiffier et al. (1998), Coiffier et al. (2002), Tam et al. (2008), and Pfreundschuh et al. (2011) Wierda et al. (2010), Shanafelt et al. (2013), Czuczman et al. (2012), and Ujjani et al. (2015) Hillmen et al. (2007) Van Cutsem et al. (2009), Bonner et al. (2010), Vermorken et al. (2008), and Pirker et al. (2009)
constitutively activated proteins, regardless of upstream influence from EGFR. Cetuximab also has benefit in the treatment of head and neck squamous cell cancers, both as primary therapy in conjunction with definitive radiation (Bonner et al., 2010) and in conjunction with chemotherapy for unresectable disease (Vermorken et al., 2008). Finally, when added to chemotherapy, cetuximab leads to a modest increase in survival over chemotherapy alone in metastatic nonsmall cell lung cancer (Pirker et al., 2009). In the same family as EGFR, HER2 receptors are responsible for stimulation of cellular proliferation; overexpression of this protein by certain cancers results in uncontrollable cancer cell growth and reproduction (Hudis, 2007). This overexpression has been taken advantage of clinically, with the design of monoclonal antibodies targeting this protein. Trastuzumab, a recombinant monoclonal antibody-targeting HER2, was first demonstrated to have clinical benefit for the treatment of patients with metastatic breast cancer whose tumors overexpressed HER2 in 2001 (Slamon et al., 2001). For the management of women with a HER2 overexpressing breast cancer, this benefit has since been demonstrated to extend into the adjuvant setting as well (Leyland-Jones et al., 2005; Romond et al., 2005). Overexpression of HER2 can also be seen in patients with gastric cancer. The results of the ToGA trial (Bang et al., 2010) demonstrated an improvement in overall survival when trastuzumab was added to standard chemotherapy for the management of patients with metastatic gastric cancer whose tumors were found to overexpress HER2. Use of this monoclonal antibody is now standard for the treatment of these patients. An important therapeutic strategy in cancer treatment is targeting angiogenesis, the process of new blood vessel formation necessary for tumor growth. This process is primarily driven by ligand-binding vascular endothelial growth factor, or VEGF, which consists of a family of six different proteins delineated as VEGF A through E, and PIGF (Smaglo and Hwang, 2013). These proteins have proven to be important targets for
Bokemeyer et al. (2009) Slamon et al. (2001), Leyland-Jones et al. (2005), Romond et al. (2005), and Bang et al. (2010) Van Cutsem et al. (2012) Fuchs et al. (2007), Hochster et al. (2008), Giantonio et al. (2007), Bennouna et al. (2013), Sandler et al. (2006), and Vredenburgh et al. (2007) Fuchs et al. (2014), Wilke et al. (2014), Garon et al. (2014), and Tabernero et al. (2015)
monoclonal antibodies. The most thoroughly explored antiVEGF antibody is bevacizumab, which binds to VEGF-A and prevents its subsequent receptor binding. In the management of metastatic colorectal cancer, bevacizumab has demonstrated survival benefit when added to both conventional oxaliplatin and irinotecan chemotherapy regimens (Fuchs et al., 2007; Hochster et al., 2008). This benefit holds true regardless of line of therapy or previous bevacizumab exposure (Giantonio et al., 2007; Bennouna et al., 2013). Bevacizumab is also employed in the treatment of certain lung cancers and in glioblastoma multiforme (Sandler et al., 2006; Vredenburgh et al., 2007). A second monoclonal antibody-like structure targeting the VEGF system, ziv-aflibercept, works by binding several of the VEGF ligand proteins together and thus preventing their receptor binding and activation. The Velour study demonstrated the benefit of this newer antibody, which presently has only the single indication for treatment in second line metastatic colon cancer in combination with irinotecan-based chemotherapy (Van Cutsem et al., 2012).
Immunoconjugates Antibodies have also been utilized against tumors by delivering cytotoxic payloads using immunoconjugates. Immunoconjugates are complex therapeutic molecules that make use of antibodies’ cancer-targeting specificities, linking therapeutic agents to the antibody in order to deliver toxic payloads to the tumor target. Three components are essential to the construction of an effective immunoconjugate: the antibody, the therapeutic payload, and the linker that joins the two components. Immunoconjugates are divided based upon the mechanism of action of the therapeutic agent conjugated to the antibody, which may be a pharmacologic agent, a radionucleotide, or a catalytic toxin. Of these therapeutic entities, immunoconjugates employing pharmacologic and radionucleotide agents have obtained FDA approval. Pharmacologic agents that block tubulin polymerization (such as auristatin and maytansinoids) or induce DNA strand
552
Tumor Immunology j Antibody Therapy
scission (such as calicheamicin) have been successfully employed as therapeutic conjugates in cancer therapy (Dosio et al., 2011). Perhaps the two most successful immunoconjugates to date are brentuximab vedotin and trastuzumab emtansine, which hold FDA approvals for the treatment of lymphomas and breast cancers, respectively. Brentuximab vedotin is an immunoconjugate that links an anti-CD30 antibody to the antimicrotubule monomethylauristatin E (MMAE) (Pro et al., 2012). This immunoconjugate is internalized when the antibody binds the CD30 antigen and enters into the lysosome, where MMAE is cleaved off and released; MMAE can then interrupt the microtubule network, inducing cellular apoptosis of the CD30-expressing tumor cell. In patients with anaplastic large cell lymphoma refractory to at least one prior therapy, treatment with brentuximab vedotin demonstrated an 86% objective response rate, with 57% of patients achieving a complete remission (Sutherland et al., 2006). In patients who had relapsed Hodgkin’s disease despite autologous stem cell transplant, treatment with brentuximab vedotin resulted in an overall response rate of 75%, with 34% of patients achieving a complete remission (Younes et al., 2012). Trastuzumab emtansine is an immunoconjugate of the anti-HER2 monoclonal antibody trastuzumab and DM1, a maytansinoid toxic to tumor cells through inhibition of tubulin polymerization (LoRusso et al., 2011). In the 2012 EMILIA study, efficacy of trastuzumab emtansine was compared to the combination of lapatinib and capecitabine in patients with HER2-positive breast cancer whose disease was refractory to trastuzumab therapy (Verma et al., 2012). This study demonstrated a superior progression free survival with the use of trastuzumab emtansine when compared with lapatinib/capecitabine. The activity of trastuzumab emtansine, even in patients who had been previously treated with trastuzumab illustrates the therapeutic potential of immunoconjugates beyond unconjugated antibody therapy. The immunoconjugate gemtuzumab ozogamicin, which links calicheamicin to a CD33-targeting antibody, received accelerated FDA approval in 2000 for the treatment of relapsed acute myeloid leukemias (Bross et al., 2001). This accelerated approval was granted because the agent, in early phase studies, appeared to be well-tolerated and effective, whereas conventional chemotherapy in this setting is often poorly tolerated and ineffective. However, in a regulatory agency–required postapproval study, gemtuzumab ozogamicin did not improve clinical benefit and demonstrated higher rates of toxicity compared to conventional chemotherapy (Petersdorf et al., 2009). Gemtuzumab ozogamicin was subsequently withdrawn in 2010. The immunoconjugate ibritumomab tiuxetan is the only radioimmunoconjugate currently holding FDA approval. This agent has antitumor activity in rituximab-refractory follicular lymphomas. Ibritumomab is a monoclonal antibody that binds the CD20 antigen; tiuxetan is a linker molecule, which allows the antibody to deliver the radioactive isotope 90yttrium to the tumor cell. In non-Hodgkin lymphoma patients who were refractory to therapy with the monoclonal antibody rituximab and were heavily pretreated with systemic therapies, the administration of ibritumomab tiuxetan produced an overall response rate of 74%, with 15% of patients achieving a complete response (Witzig et al., 2002).
Of note, another FDA-approved radionucleotide immunoconjugate, iodine tositumomab, encountered challenges that prompted its manufacturer to remove it from the market in February 2014. Kaminski and colleagues reported in 2001 that iodine tositumomab delivers radioactive 131-iodine to CD20 expressing tumor cells via an anti-CD20 monoclonal antibody and is effective treatment of follicular lymphoma (Kaminski et al., 2001). Reported median survival with this agent is 22.8 months. However, following regulatory approval and marketing, practitioner familiarity with the monoclonal antibody rituximab also may have limited the widespread use of this radionucleotide immunoconjugate, and treatment using the new agent was considered to be complicated to deliver.
Antibodies in Clinical Trials Monoclonal Antibodies Presently, nearly 700 studies are listed in clinicaltrials.gov, recruiting patients with various cancers for treatment with monoclonal antibodies, including 96 studies that have reached Phase III level of investigation. Specific information about clinical trials can be found at the url: http://clinicaltrials.gov/. While many of these trials are exploring the activity of the agents already approved in either other cancer types, lines of therapy, or combinations, a number of new monoclonal antibodies are being developed as well. Rilotumumab is a fully humanized monoclonal antibody that targets hepatocyte growth factor, preventing its interaction with cMET, and thus prevents cMET activation and tumor proliferation (Giordano, 2009). This agent is being investigated in the treatment of gastric, lung, and brain cancers. Bavituximab is a chimeric antibody, which targets the membrane phospholipid phosphatidylserine, resulting in occlusion of tumor blood vessels (DeRose et al., 2011), is being evaluated for the treatment of rectal, lung, liver, and skin cancers. Some of these new monoclonal antibodies are making their way into clinical practice. Ramucirumab, a fully humanized monoclonal antibody directed against the VEGF receptor 2, prevents ligand binding and thus downstreams signaling that promotes tumor angiogenesis. Based on the 2014 results of the REGARD trial (Fuchs et al., 2014) and the RAINBOW trial (Wilke et al., 2014), ramucirumab is approved for the secondline treatment of patients with metastatic gastric cancer. Ramucirumab has approvals for the treatment of certain lung and colon cancers as well (Garon et al., 2014; Tabernero et al., 2015). Siltuximab, a chimeric monoclonal antibodytargeting IL-6, is being investigated for the treatment of both solid tumors (renal cell, prostate, and ovarian cancers) and liquid tumors (non-Hodgkin’s lymphomas, multiple myeloma). In 2014, siltuximab became the first-approved treatment for multicentric Castleman’s disease, through the FDA’s priority review program (Markman and Patel, 2014).
Immunoconjugates A number of pharmacologic immunoconjugates are being developed; these molecules exploit known specific surface markers as targets for therapeutic delivery. Table 2 summarizes the pharmacologic immunoconjugates under active investigation. Such immunoconjugates include other calicheamicin conjugates: the agent inotuzumab ozogamicin, for example, consists of calicheamicin linked to a humanized antibody
Tumor Immunology j Antibody Therapy
553
Table 2 Selected pharmacologic immunoconjugates in various stages of development are summarized by disease type, pharmacologic entity, and antibody, with the studies that support their use in these diseases Agent
Diseases treated
Cytotoxic agent
Antibody type
Studies
Inotuzumab ozogamicin
B cell non-Hodgkin’s lymphoma or acute lymphoid leukemias Metastatic breast cancer or unresectable melanoma Ovarian, Merkel cell, and small cell lung cancers NaPi2b expressing ovarian and NSCLC Glioblastoma B cell non-Hodgkin’s lymphoma or Acute lymphoid leukemia
Calicheamicin
Humanized IgG4 targeting CD22
Ricart (2011)
MMAE DM1
Humanized IgG2-targeting transmembrane glycoprotein NMB Humanized IgG1-targeting CD56
Maric et al. (2013), Hamid et al. (2010) Woll (2009)
MMAE MMAF DM4
Humanized IgG-targeting NaPi2b Humanized IgG-targeting EGFR Humanized IgG1-targeting CD19
Burris et al. (2014) Gan et al. (2014) Ribrag et al. (2014)
Glembatumumab vedotin Lorvotuzumab mertansine DNIB0600A ABT-414 SAR3419
directed against CD22 (Ricart, 2011). This agent is being studied for treatment of relapsed or refractory acute lymphoblastic leukemia, in B cell non-Hodgkin’s lymphomas, and as a part of a preparative regimen for stem cell transplant. SAR3419, an immunoconjugate of a CD19 antibody and a maytansinoid (DM4), is under investigation for the treatment of relapsed or refractory B cell non-Hodgkin’s lymphoma and in relapsed or refractory acute lymphocytic leukemia (Ribrag et al., 2014). A number of additional antibody targets in solid tumors are actively being investigated in the development of pharmacologic immunoconjugates as well. One tumor target, glycoprotein nonmetastatic B (GPNMB), is overexpressed in certain solid tumors, including triple-negative breast cancer and melanoma. It is being investigated as the antibody target for the immunoconjugate glembatumumab vedotin in these cancer types (Maric et al., 2013; Hamid et al., 2010). Lorvotuzumab mertansine, which combines the maytansinoid DM1 with an anti-CD56–targeting antibody, is under phase I investigation for the treatment of patients with advanced ovarian, Merkel cell, and small cell lung cancers (Woll, 2009). Gan et al. (2014) have described ABT-414, an immunoconjugate with an Auristatin moiety linked to an antibody-targeting–activated EGFR, which demonstrates preliminary promise in patients with glioblastoma (Gan et al., 2014). Studies of DNIB0600A, which links MMAE to an antibody-targeting NaPi2b, are underway to evaluate for efficacy, following encouraging phase I data presented by Burris and colleagues at the 2014 ASCO meeting for the treatment of ovarian and nonsmall cell lung cancers (Burris et al., 2014). Compared to pharmacologic immunoconjugate therapies, in the wake of the discontinuation of iodine tositumomab
production, further radioimmunoconjugate development has slowed, and it remains to be seen if such agents will find an expanded role in the treatment of human cancers. However, a third class of immunoconjugates, which employs a catalytic toxin as the therapeutic entity, remains interesting, relatively underexplored, and potentially useful. As a systemic treatment, an intact bacterial or other pathologic toxin cannot be readily used as a cancer treatment, due to the significant collateral toxicity to healthy patient tissue. However, if such molecules can be reliably delivered to the target, i.e., the cancer, with high selectivity, then they have the potential to be extremely effective. Pastan and colleagues at the Laboratory of Molecular Biology of the National Cancer Institute’s Center for Cancer Research have described their progress in developing this class of immunoconjugate. For example, Kreitman reported that treatment with moxetumomab pasudotox, an immunoconjugate that delivers pseudomonas exotoxin A to the CD22expressing leukemic cells was well-tolerated without the development of dose-limiting toxicities to treat relapsed/ refractory hairy cell leukemia (Kreitman et al., 2012). Treatment with moxetumomab pasudotox is also being explored in treatment of acute lymphocytic leukemia and non-Hodgkin’s lymphoma. In a separate trial also led by the Laboratory of Molecular Biology, the SS1P anti-mesothelin immunotoxin is being administered to patients along with T cell and B cell– depleting chemotherapies to minimize antitoxin antibody responses. Major tumor regressions were reported in 3 of 10 patients with mesothelioma who were treated with this regimen, supporting the further development of this therapy in solid tumors (Hassan et al., 2013). Immunotoxin immunoconjugates under development are summarized in Table 3.
Table 3 Toxin immunoconjugates in development are summarized by disease type, toxin employed, and antibody, with the studies that support their use in these diseases Agent
Diseases treated
Toxin
Antibody type
References
Moxetumomab pasudotox
Relapsed/refractory hairy cell leukemia Relapsed/refractory acute lymphoid leukemia Non-Hodgkin lymphoma Mesothelioma
Pseudomonas exotoxin A
Fv antibody fragment targeting CD22
Kreitman et al. (2012)
Pseudomonas exotoxin A
Fv antibody fragment targeting mesothelin
Hassan et al. (2013)
SS1P
554
Tumor Immunology j Antibody Therapy
New Candidates – Preclinical Evidence In addition to the development of new agents and novel combinations, a number of approaches to optimize radionucleotide immunoconjugate therapy have been explored. One particular approach employed a pretargeting strategy in the treatment of tumor cells with radioimmunotherapies (Press et al., 2001). Pretargeting initially uses an unlabeled antibody to localize the tumor; a radionucleotide–hapten complex is subsequently administered, which reacts with the pretargeted antibody to concentrate the radionucleotide to the tumor. While preclinical evaluation of this method targeting CD20 in this manner demonstrated promise, results were disappointing in a phase II study of the treatment of patients with metastatic colorectal cancer with pretargeted radioimmunotherapy, both in terms of high rates of toxicity and low rate of response (Knox et al., 2000). Preclinical efforts to optimize pretargeting radionucleotide immunoconjugate therapy are ongoing in order to further the development of radioimmunoconjugate therapy as well. The work of Breichbiel and colleagues has focused on the use of targeted-a therapy, in which a particles are delivered by appropriate vectors to tumor cells (Baidoo et al., 2013). Exquisitely cytotoxic and exhibiting a short path length, the a particles are particularly suited to the situations of minimal residual disease or micrometastases.
Identification and Molecular Engineering of Therapeutic Antibodies
Improvements in antibody engineering techniques have allowed for the customization of therapeutic antibodies to overcome traditional mAb limitations. Modification in the antibody structure has resulted in the manipulation of immunogenicity, size, valence, affinity for target antigen, and Fc domain–directed interactions. Specific examples include:
Fab Domain Modification The Fab domain is the antigen-binding domain of antibodies. Antibody engineering has utilized hybridoma technology and phage- or yeast-display libraries to create a 25-kDa, monovalent single chain Fv (scFv) composed of the variable domains (VH and VL) of an antibody fused together with short peptide linker. The scFv became the basic building block of antibodybased fragments such as the enzymatically cleaved fragments, Fab and F(ab’)2, and genetically engineered formats such as scFv, (scFv)2, diabody, and minibody (Figure 1; reviewed by Robinson et al., 2004). Moreover, techniques such as chain-shuffling and site-directed mutagenesis (Swers et al., 2004; Crameri et al., 1996; Marks et al., 1992) are employed to manipulate antigen-binding properties and define the roles of binding affinity and avidity on in vitro and in vivo tumor targeting, immune effector functions such as antibodydependent cell-mediated cytotoxicity (ADCC) and therapeutic efficacy.
Fc Domain Modification
The size of antibodies (typically about 150 kDa) is regarded as a major limitation of their therapeutic application and effectiveness due to its role in hindering tissue penetration and delivery (Chames et al., 2009). Size also attributes to nonspecific uptake of antibodies or immunoconjugates by the reticuloendothelial system, limited efficacy, and the induction of dose-limiting toxicity to the liver and bone marrow (Weiner et al., 2012).
The significance of the Fc domain is due to its role in immune effector cell activation. Fcg on the surface of immune effector cells (NK cells, dendritic cells, neutrophils, and mononuclear phagocytes) recognizes the Fc domain and transduces activating signals through immunoreceptor tyrosine-based activation motifs (ITAMs) or delivers inhibitory signals though immunoreceptor tyrosine-based inhibitor motifs (ITIMs). Most Fc stimulatory signals are transduced by FcgR1 (CD64) and FcgRIIIA (CD16a). Cross-linking of FcgRs on effector cells
Enzymacally derived fragments
F(ab’)2
Fab’
Fc
scFv
(scFv)2
Minibody
Diabody
Triabody
Genecally engineered fragments Figure 1
Structures of enzymatically derived and genetically engineered fragments of Ig molecule.
Tetrabody
Tumor Immunology j Antibody Therapy promotes ADCC and tumor cell destruction (Nimmerjahn and Ravetch, 2012). ADCC has been shown to be an important mechanism for monoclonal antibodies used in cancer therapy. Manipulation of the Fc domain effector functions has been via two general approaches. The first approach entails altering the amino acid structure of the domain to influence binding affinity to Fc receptors (Shields et al., 2001; Lazar et al., 2006; Nimmerjahn and Ravetch, 2012; Stavenhagen et al., 2008). Altered amino acid sequences have led to the creation of Fc domains with higher affinity for FcgRIIIA and enhanced ADCC properties. The second approach is by modifying the oligosaccharide content of the Fc domain. The defucosylation of antibody oligosaccharides has shown enhancement of ADCC in vitro and enhanced in vivo antitumor activity (Kubota et al., 2009).
Ig-Like Scaffolds Our knowledge of the structure and function of the immunoglobulin molecule has allowed us to recognize the limitations of current forms of antibody therapy. In an effort to address such limitations, scientists have utilized current engineering technologies and naturally occurring structures to developed recombinant single chain Ig scaffolds.
Nanobodies Nanobodies are heavy chain antibodies (HCAB) based on distinct Ig structures found naturally in sharks and camelid. The structure of HCAB is unique, composed of a single heavy chain with one variable domain (VHH) (Figure 2(a)).The VHH is equivalent to the Fab fragment of IgG antibodies (Muyldermans et al., 1994). Nanobodies are smaller and have longer VHH domains facilitating their reach to targets inaccessible to conventional antibodies (Muyldermans et al., 1994). They are also easily manipulated yielding
(a)
555
bispecific/bifunctional structures and posses advantageous properties such as specificity, stability, thermotolerance, and low immunogenicity. The structural and functional appeal of nanobodies has increased their attractiveness as potential drugs leading to their commercial development and selection by phage display libraries. There are currently six nanobodies undergoing clinical trials, none of which are for anticancer purposes (Muyldermans, 2013).
Domain Antibodies (dAbs) dAbs or engineered antibody domains (eAds) are small engineered monomers (11–15 kDa) based on the variable domain of an antibody heavy chain (VH domain) (Ward et al., 1989) or light chain (VL domain) (Pereira et al., 1998). Their small size facilitates and ease of modification makes them attractive candidates for pharmaceutical development. The goal is to produce stable novel structures with favorable biophysical properties unique to dAb (Holt et al., 2003). Currently, the only dAb undergoing clinical trials is CEP-37247; an antiTNF bivalent dAb fused to a human IgG1 Fc region (Gay et al., 2010), for the treatment of sciatica. In cancer therapy, dAbs are yet to establish therapeutic efficacy and application.
Chemically Programmed Antibodies (cpAbs) CpAbs are novel scaffolds composed of an antibody fragment joined to a synthetic component (peptide or small molecule) (Figure 2(b)). The synthetic component functions as an antigen recognizing and binding region, while the antibody fragment carries out the effector functions, in addition to contributing to the half-life and bivalency. The minimum antibody requirement in cpAbs is the Fc fragment, while the Fab portion is usually replaced by a synthetic component (reviewed by Rader, 2014). The only cpAbs undergoing clinical trials have peptides as their synthetic component and are based on the CovX-Body platform. There has been a halt in the development
(b)
VHH
Pepde or small molecule Fc
VHH (nanobody) Fc
Heavy chain anbody (HCAB)
(c)
BiTes
DaRTs
TandAB
Figure 2 Schematic representation of Ig-like scaffolds (a) Heavy chain antibody (HCAB) and VHH derivative (nanobody) (b) Chemically programmed antibodies (CpAbs), composed of an antibody component and a synthetic component (peptide or small molecule) and (c) Bispecific scaffolds BiTes, DaRTs, and TandABs.
556
Tumor Immunology j Antibody Therapy
of cpAbs, yet they still demonstrate potential due to a range of their possible synthetic components.
specific cancers, including melanoma and non-small cell lung cancer (Larkin et al., 2015; Weber et al., 2015; Robert et al., 2015a; Gettinger et al., 2015; Brahmer et al., 2015; Robert et al., 2015b).
Antibodies Targeting the Immune System Targets of antibody therapy are not limited to the tumor cell; they can also be used to modulate a desired immune response or signaling pathway. The immune system is very attractive therapeutically. Manipulating entities such as immunoregulatory coreceptors and other cell surface signaling molecules, could lead to enhanced antitumor response and influence the outcome of lymphocyte-mediated immune responses by modulating antigen-specific T cell receptor and B cell receptor signals (Weiner et al., 2010). Ipilimumab is the first FDA-approved immune-targeting monoclonal antibody against cytotoxic T lymphocyte–associated antigen 4 (CTLA-4), an inhibitory receptor of T cell activation.
Antibodies in Clinical Trials Manipulating the immune system to fight the tumor has proven to be a difficult task. Yet, understanding the mechanisms in which the effector cells operate, and the success of ipilimumab has opened the door for several other targets that are currently in clinical trials. Other targets under study that serve as costimulatory molecules include PD-1/PDL-1, CD40, and CD137 (Pardoll, 2012). There are currently 22 antibodies in clinical trials that target the immune system (Table 4), 19 of which are for the treatment of cancer (Yao et al., 2013). Several of these therapies have garnered approval for treatment of
Table 4
New Candidates The manipulation and stimulation of the immune system can be achieved not only by mAb but also by structurally modified antibodies such as bispecific antibodies (bsAbs) and trispecific antibodies (triomAbs). These modified structures are able to simultaneously engage tumor antigen and an immune activator, redirecting the immune response toward the tumor antigen. A common target of bsAbs and triomAb is CD3, the activating receptor for T cells, whose engagement activates and retargets T cells to the tumor. An example of a developed triomAb is catumaxomab, which binds the tumor antigen EpCAM, CD3, and innate effector cells through its intact Fc portion (Ruf and Lindhofer, 2001). Other tumor antigens targeted by triomAbs include HER2/neu (ertumaxomab), CD20 (Bi20/FBTA05), GD2, and GD3 (Ektomun) (Weiner et al., 2012).
Identification and Genetic Engineering of Therapeutic Antibodies Genetic engineering has also been utilized to create multifunctional protein scaffolds derived from the scFv antibody fragment that are able to modulate the immune system (Figure 2(c)). Bispecific formats have demonstrated notable clinical success (Shahied et al., 2004; Müller et al., 2010;
Antibodies targeting the immune system in clinical trials (Yao et al., 2013)
Name
Antibody type
Target
Activity
Diseases treated
Trial phase
Tremelimumab Galiximab
Human IgG2 Chimeric IgG1 Human IgG4 Humanized IgG1 IgG4 Fusion protein (B7DC þ IgG1) Human IgG4 Engineered human IgG1 Engineered human IgG1 Humanized IgG1 Fusion protein (LAG3 þ IgG1)
CTLA4 CD80 PD1–B7H1, PD1–B7DC PD1–B7H1, PD1–B7DC PD1–B7H1, PD1–B7DC PD1–B7H1, PD1–B7DC PD1–B7H1 PD1–B7H1 PD1–B7H1 B7H3 LAG3–MHCII
Solid tumors Lymphoma Multiple cancers Solid tumors Solid tumors Multiple cancers Solid tumors
II II
Anti-OX40 TRX518 CP-870,893 Lucatumumab
Human IgG4 Human IgG Human IgG1 OX40-specific mouse IgG GITR-specific humanized IgG1 Human IgG1 Human IgG1
CD137 CD137 CD27 OX40 GITR–GITRL CD40 CD40
T cell priming and activation B cell proliferation T cell activation and tolerance T cell activation and tolerance T cell activation and tolerance T cell activation and tolerance T cell activation and tolerance T cell activation and tolerance T cell activation and tolerance T Cell activation and tolerance DC maturation and T cell activation T cell activation T cell activation T cell activation CD4 T cell activation T cell activation APC activation and B cell maturation APC activation and B cell maturation
Dacetuzumab
Humanized IgG1
CD40
AMP224 BMS-936559 MPDL3280A MGA271 IMP321
PF-05082566
Solid tumors Solid tumors Multiple cancers
Solid tumors Lymphoma Multiple cancers Prostate cancer Solid tumors Multiple cancers Lymphoma and leukemia APC activation and B cell maturation Lymphoma and multiple myeloma
I I
I I/II I/II I I II I I I/II II
B7H1, B7 homolog 1 (programmed death-1 ligand-1); B7DC, B7 dendritic cell (programmed death-1 ligand-2); CTLA4, cytotoxic T lymphocyte antigen 4; GITR, glucocorticoidinduced TNFR-related protein; GITRL, GITR ligand; IgG1, immunoglobulin G1; LAG3, lymphocyte activation gene 3; MHCII, major histocompatibility complex class II; OX40L, OX40 ligand; PD1, programmed cell death protein 1.
Tumor Immunology j Antibody Therapy Rossi et al., 2003; Baeuerle et al., 2009; Kellner et al., 2008), the most successful of which is the BiTE (bispecific T cell engager). BiTEs simultaneously engage CD3 and a tumor antigen such as CD19, EpCAM or EGFR. Blinatumomab (MT-103) is the most clinically advanced BiTE targeting CD19 and CD3 (Baeuerle et al., 2009), it is undergoing Phase III clinical trials for treatment of acute lymphoblastic leukemia and has completed Phase I evaluations in non-Hodgkin’s lymphoma. Other bispecific scaffolds being developed are DARTs (dual affinity retargeting) (Rader, 2011) and TandAbs (tetravalent tandem antibodies) (Mølhøj et al., 2007). Their structures and functions resemble BiTes but differ in the linkage of heavy and light chain variable domains. DARTs have yet to reach clinical trials while TandAb have been in clinical development, with the completion of Phase I clinical trial of AFM13 against CD30 and CD16a (FcgRIIIA) (Mølhøj et al., 2007; Baeuerle et al., 2008; Rothe et al., 2011). Clinical testing of several additional anticancer therapies are underway that expand upon and apply the immunoconjugate concept in novel ways. In 1999, Marks et al. developed a method to select antibodies optimal for internalization by cancer cells though the recovery of infectious phage from the cancer cells to be targeted (Becerril et al., 1999). In 2009, this method was used to create highly specific antibodies coupled to liposomes containing doxorubicin, which demonstrated significant antitumor effects in mice (ElBayoumi and Torchilin, 2009). Exploration of this method in human subjects has potential merit. This concept of using antibody-loaded liposomes to deliver a therapeutic entity has also been explored as a novel method of immunoconjugate development. Immunoliposomes also have been employed by Xu and colleagues as delivery vehicles for the p53 tumor suppressor (Xu, 2001). The therapeutic potential of immunoliposomes has been explored in some phase I studies. In addition to this repurposing of antibodies already in clinical use, future advancement of immunoconjugate therapy will draw on new techniques being developed in other areas of cancer medicine that can be adapted to further antibody selection and design. These techniques involve the selection of antigens to serve as cancer-specific antibody targets as well as optimization of the internalization of the antibodies by the tumor cells. Some tumor-specific antigens that could be targeted by immunoconjugates may emerge from intense genomic analysis of tumors, some of which may be identifying driver mutations. The tumor-specific protein products of these mutations would be attractive targets for an immunoconjugate, as toxicity to normal cells based upon on-target toxicity would be minimized. New approaches may further widen the use of immunoconjugates. For example, Adams, Rudnick, and colleagues have reported in 2011, the utility of a novel radiotracer linked to a HER2 receptor antibody for positron emission tomography imaging, and have identified antibody structural features that influence cellular internalization and subsequent tumor catabolism that is associated with variability in antibody affinity for the antigen (Rudnick et al., 2011). Finally, antibody therapy can be incorporated into vaccination strategies. Schlom and colleagues demonstrated in 2012 how vaccine-induced immune responses can be enhanced by trastuzumab therapy in breast cancer patients (Schlom, 2012; Disis et al., 2009; Norton et al., 2014). This
557
approach may have additional utility in augmenting immunoconjugate therapy that uses trastuzumab or other monoclonal antibodies as the delivery vehicle of the therapeutic entity to the cancer. Through these and other techniques, the engineering of therapeutic antibodies capable of targeting cancerspecific epitopes with ever-higher degrees of specificity will allow for even greater anticancer effects.
See also: Myeloid and B Cell Development: Regulation of IgL Chain Recombination; Regulation of Igh Recombination and Allelic Exclusion. Structure and Function of Diversifying Receptors: IgG Structure and Function; Structure and Function of Camelid VHH. Tumor Immunology: Cancer Immunosurveillance: Immunoediting; Inflammation and Cancer; Therapeutic and Prophylactic Cancer Vaccines; Tumor Antigens, Viral Origin; Vaccines and Their Role in CD8 T CellMediated Antitumor Immunity.
References Adams, G.P., et al., 2001. High affinity restricts the localization and tumor penetration of single-chain fv antibody molecules. Cancer Res. 61, 4750–4755. Adams, G.P., et al., 1998. Increased affinity leads to improved selective tumor delivery of single-chain Fv antibodies. Cancer Res. 58, 485–490. Baeuerle, P.A., Kufer, P., Bargou, R., 2009. BiTE: teaching antibodies to engage T-cells for cancer therapy. Curr. Opin. Mol. Ther. 11, 22–30. Baeuerle, P.A., Reinhardt, C., Kufer, P., 2008. BiTE: a new class of antibodies that recruit T-cells. Drugs Fut. 33, 2. Baidoo, K.E., Yong, K., Brechbiel, M.W., 2013. Molecular pathways: targeted a-particle radiation therapy. Clin. Cancer Res. 19, 530–537. Bang, Y.-J., et al., 2010. Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastrooesophageal junction cancer (ToGA): a phase 3, open-label, randomised controlled trial. Lancet 376, 687–697. Becerril, B., Poul, M.A., Marks, J.D., 1999. Toward selection of internalizing antibodies from phage libraries. Biochem. Biophys. Res. Commun. 255, 386–393. Bennouna, J., et al., 2013. Continuation of bevacizumab after first progression in metastatic colorectal cancer (ML18147): a randomised phase 3 trial. Lancet Oncol. 14, 29–37. Bokemeyer, C., et al., 2009. Fluorouracil, leucovorin, and oxaliplatin with and without cetuximab in the first-line treatment of metastatic colorectal cancer. J. Clin. Oncol. 27, 663–671. Bonner, J.A., et al., 2010. Radiotherapy plus cetuximab for locoregionally advanced head and neck cancer: 5-year survival data from a phase 3 randomised trial, and relation between cetuximab-induced rash and survival. Lancet Oncol. 11, 21–28. Brahmer, J., et al., 2015. Nivolumab versus docetaxel in advanced squamous-cell non-small-cell lung cancer. N. Engl. J. Med. 373, 123–135. Bross, P.F., et al., 2001. Approval summary: gemtuzumab ozogamicin in relapsed acute myeloid leukemia. Clin. Cancer Res. 7, 1490–1496. Burris, H.A., et al., 2014. A phase I study of DNIB0600A, an antibody-drug conjugate targeting NaPi2b, in patients with non-small cell lung cancer or platinum-resistant ovarian cancer. J. Clin. Oncol. 32, (5s). Chames, P., et al., 2009. Therapeutic antibodies: successes, limitations and hopes for the future. Br. J. Pharmacol. 157, 220–233. Clinicaltrials.gov, 2015. Coiffier, B., et al., 2002. CHOP plus rituximab with CHOP chemotherapy in elderly patients with diffuse large B-cell lymphoma. N. Engl. J. Med. 346, 235–242. Coiffier, B., et al., 1998. Rituximab (anti-CD20 monoclonal antibody) for the treatment of patients with relapsing or refractory aggressive lymphoma: a multicenter phase II study. Blood 92, 1927–1932. Crameri, A., Cwirla, S., Stemmer, W.P., 1996. Construction and evolution of antibodyphage libraries by DNA shuffling. Nat. Med. 2, 100–102. Czuczman, M.S., et al., 2012. Ofatumumab monotherapy in rituximab-refractory follicular lymphoma: results from a multicenter study. Blood 119, 3698–3704. DeRose, P., Thorpe, P.E., Gerber, D.E., 2011. Development of bavituximab, a vascular targeting agent with immune-modulating properties, for lung cancer treatment. Immunotherapy 3, 933–944.
558
Tumor Immunology j Antibody Therapy
Disis, M.L., et al., 2009. Concurrent trastuzumab and HER2/neu-specific vaccination in patients with metastatic breast cancer. J. Clin. Oncol. 27, 4685–4692. Dosio, F., Brusa, P., Cattel, L., 2011. Immunotoxins and anticancer drug conjugate assemblies: the role of the linkage between components. Toxins 3, 848–883. ElBayoumi, T.A., Torchilin, V.P., 2009. Tumor-targeted nanomedicines: enhanced antitumor efficacy in vivo of doxorubicin-loaded, long-circulating liposomes modified with cancer-specific monoclonal antibody. Clin. Cancer Res. 15, 1973–1980. Fuchs, C.S., et al., 2014. Ramucirumab monotherapy for previously treated advanced gastric or gastro-oesophageal junction adenocarcinoma (REGARD): an international, randomised, multicentre, placebo-controlled, phase 3 trial. Lancet 383, 31–39. Fuchs, C.S., et al., 2007. Randomized, controlled trial of irinotecan plus infusional, bolus, or oral fluoropyrimidines in first-line treatment of metastatic colorectal cancer: results from the BICC-c study. J. Clin. Oncol. 25, 4779–4786. Gan, H.K., et al., 2014. A phase I study evaluating ABT-414 in combination with temozolamide for subjects with recurrent or unresectable glioblastoma. J. Clin. Oncol. 32, 5s. Garon, E.B., et al., 2014. Ramucirumab plus docetaxel versus placebo plus docetaxel for second-line treatment of stage IV non-small-cell lung cancer after disease progression on platinum-based therapy (REVEL): a multicentre, double-blind, randomised phase 3 trial. Lancet 384, 665–673. Gay, R.D., et al., 2010. Anti-TNFa domain antibody construct CEP-37247: full antibody functionality at half the size. mAbs 2, 625–638. Gettinger, S.N., et al., 2015. Overall survival and long-term safety of nivolumab in patients with previously treated advanced non-small-cell lung cancer. J. Clin. Oncol. 33, 2004–2012. Giantonio, B.J., et al., 2007. Bevacizumab in combination with oxaliplatin, fluorouracil, and leucovorin (FOLFOX4) for previously treated metastatic colorectal cancer: results from the Eastern Cooperative Oncology Group Study E3200. J. Clin. Oncol. 25, 1539–1544. Giordano, S., 2009. Rilotumumab, a mAb against human hepatocyte growth factor for the treatment of cancer. Curr. Opin. Mol. Ther. 11, 448–455. Hamid, O., Sznol, M., Pavlick, A.C., 2010. Frequent dosing and GPNMB expression with CDX011 (CR011vcMMAE), an antibody–drug conjugate (ADC), in patients with advanced melanoma. J. Clin. Oncol. 28, 15s. Hassan, R., et al., 2013. Major cancer regressions in mesothelioma after treatment with an anti-mesothelin immunotoxin and immune suppression. Sci. Transl. Med. 5 (208), ra147. Hillmen, P., et al., 2007. Alemtuzumab compared with chlorambucil as first-line therapy for chronic lymphocytic leukemia. J. Clin. Oncol. 25, 5616–5623. Hochster, H.S., et al., 2008. Safety and efficacy of oxaliplatin-fluoropyrimidine regimens with or without bevacizumab as first-line treatment of metastatic colorectal cancer (mCRC): final analysis of the TREE-Study. J. Clin. Oncol. 24, 18s. Holt, L.J., et al., 2003. Domain antibodies: proteins for therapy. Trends Biotechnol. 21, 484–490. Hudis, C.A., 2007. Trastuzumabdmechanism of action and use in clinical practice. N. Engl. J. Med. 357, 39–51. Kaminski, M.S., Zelenetz, A.D., Press, O.W., 2001. Pivotal study of iodine I 131 tositumomab for chemotherapy-refractory low-grade or transformed low-grade B-cell non-Hodgkin’s lymphomas. J. Clin. Oncol. 19, 3918–3928. Kellner, C., et al., 2008. A novel CD19-directed recombinant bispecific antibody derivative with enhanced immune effector functions for human leukemic cells. J. Immunother. 31, 871–884. Khazaeli, M.B., Conry, R.M., LoBuglio, A.F., 1994. Human immune response to monoclonal antibodies. J. Immunother. 15, 42–52. Köhler, G., Milstein, C., 1975. Continuous cultures of fused cells secreting antibody of predefined specificity. Nature 256, 495–497. Knox, S.J., et al., 2000. Phase II trial of yttrium-90-DOTA-biotin pretargeted by NR-LU-10 antibody/streptavidin in patients with metastatic colon cancer. Clin. Cancer Res. 6, 406–414. Kubota, T., et al., 2009. Engineered therapeutic antibodies with improved effector functions. Cancer Sci. 100, 1566–1572. Kreitman, R.J., et al., 2012. Phase I trial of anti-CD22 recombinant immunotoxin moxetumomab pasudotox (CAT-8015 or HA22) in patients with hairy cell leukemia. J. Clin. Oncol. 30, 1822–1828. Larkin, J., et al., 2015. Combined nivolumab and ipilimumab or monotherapy in untreated melanoma. N. Engl. J. Med. 373, 23–34. Lazar, G.A., et al., 2006. Engineered antibody Fc variants with enhanced effector function. Proc. Natl. Acad. Sci. U.S.A. 103, 4005–4010. Leyland-Jones, B., et al., 2005. Trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer. N. Engl. J. Med. 353, 1659–1672.
LoRusso, P.M., et al., 2011. Trastuzumab emtansine: a unique antibody-drug conjugate in development for human epidermal growth factor receptor 2–positive cancer. Clin. Cancer Res. 17, 6437–6447. Maric, G., Rose, A.A., Annis, M.G., Siegel, P.M., 2013. Glycoprotein non-metastatic b (GPNMB): a metastatic mediator and emerging therapeutic target in cancer. Onco Targets Ther. 6, 839–852. Marcus, R., et al., 2005. CVP chemotherapy plus rituximab compared with CVP as first-line treatment for advanced follicular lymphoma. Blood 105, 1417–1423. Markman, A., Patel, T., 2014. Siltuximab: first global approval. Drugs. 74, 1147–1152. Marks, J.D., et al., 1992. By-passing immunization: building high affinity human antibodies by chain shuffling. Biotechnology 10, 779–783. Muyldermans, S., 2013. Nanobodies: natural single-domain antibodies. Annu. Rev. Biochem. 82, 775–797. Muyldermans, S., et al., 1994. Sequence and structure of VH domain from naturally occurring camel heavy chain immunoglobulins lacking light chains. Protein Eng. 7, 1129–1135. Müller, D., et al., 2010. Bispecific antibodies for cancer immunotherapy. BioDrugs 24, 89–98. Mølhøj, M., et al., 2007. CD19-/CD3-bispecific antibody of the BiTE class is far superior to tandem diabody with respect to redirected tumor cell lysis. Mol. Immunol. 44, 1935–1943. Nimmerjahn, F., Ravetch, J.V., 2012. Translating basic mechanisms of IgG effector activity into next generation cancer therapies. Cancer Immun. 12, 13. Norton, N., et al., 2014. Association studies of Fcg receptor polymorphisms with outcome in HER2þ breast Cancer patients treated with trastuzumab in NCCTG (Alliance) trial N9831. Cancer Immunol. Res. 2, 962–969. Pardoll, D.M., 2012. The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer 12, 252–264. Pereira, B., et al., 1998. Cardiolipin binding a light chain from lupus-prone mice. Biochemistry 37, 1430–1437. Petersdorf, S., et al., 2009. Preliminary results of Southwest Oncology Group Study S0106: an international intergroup phase 3 randomized trial comparing the addition of gemtuzumab ozogamicin to standard induction therapy versus standard induction therapy followed by a second randomization to post-consolidation gemtuzumab ozogamicin versus no additional therapy for previously untreated acute myeloid leukemia. ASH Annu. Meet. Abstr. 114, 790. Pfreundschuh, M., et al., 2011. CHOP-like chemotherapy with or without rituximab in young patients with good-prognosis diffuse large-B-cell lymphoma: 6-year results of an open-label randomized study of the MabThera International Trial (MInT) Group. Lancet Oncol. 12, 1013–1022. Pirker, R., et al., 2009. Cetuximab plus chemotherapy in patients with advanced nonsmall-cell lung cancer (FLEX): an open-label randomised phase III trial. Lancet 373, 1525–1531. Press, O.W., et al., 2001. A comparative evaluation of conventional and pretargeted radioimmunotherapy of CD20-expressing lymphoma xenografts. Blood 98, 2535–2543. Pro, B., et al., 2012. Brentuximab vedotin (SGN-35) in patients with relapsed or refractory systemic anaplastic large-cell lymphoma: results of a phase II study. J. Clin. Oncol. 30, 2190–2196. Rader, C., 2014. Chemically programmed antibodies. Trends Biotechnol. 32 (4), 186–197. http://dx.doi.org/10.1016/j.tibtech.2014.02.003. Rader, C., 2011. DARTs take aim at BiTEs. Blood 117, 4403–4404. Ribrag, V., et al., 2014. A dose-escalation study of SAR3419, an anti-CD19 antibody maytansinoid conjugate, administered by intravenous infusion once weekly in patients with relapsed/refractory b-cell non-hodgkin lymphoma. Clin. Cancer Res. 20, 213–220. Ricart, A.D., 2011. Antibody-drug conjugates of calicheamicin derivative: gemtuzumab ozogamicin and inotuzumab ozogamicin. Clin. Cancer Res. 17, 6417–6427. Robert, C., et al., 2015a. Nivolumab in previously untreated melanoma without BRAF mutation. N. Engl. J. Med. 372, 320–330. Robert, C., et al., 2015b. Pembrolizumab versus ipilimumab in advanced melanoma. N. Engl. J. Med. 372, 2521–2532. Robinson, M.K., Weiner, L.M., Adams, G.P., 2004. Improving monoclonal antibodies for cancer therapy. Drug Devel. Res. 61, 172–187. Romond, E.H., et al., 2005. Trastuzumab plus adjuvant chemotherapy for operable HER2-positive breast cancer. N. Engl. J. Med. 353, 1673–1684. Rossi, E.A., et al., 2003. Development of new multivalent-bispecific agents for pretargeting tumor localization and therapy. Clin. Cancer Res. 9, 3886S–3896S. Rothe, A., et al., 2011. A phase I study with the bispecific anti-CD30 x anti-CD16A antibody construct AFM13 in patients with relapsed or refractory hodgkin lymphoma. ASH Annu. Meet. Abstr. 118, 3709.
Tumor Immunology j Antibody Therapy Rudnick, S.I., et al., 2011. Influence of affinity and antigen internalization on the uptake and penetration of anti-HER2 antibodies in solid tumors. Cancer Res. 71, 2250–2259. Ruf, P., Lindhofer, H., 2001. Induction of a long-lasting antitumor immunity by a trifunctional bispecific antibody. Blood 98, 2526–2534. Sandler, A., et al., 2006. Paclitaxel–carboplatin alone or with bevacizumab for non– small-cell lung cancer. N. Engl. J. Med. 355, 2542–2550. Schlom, J., 2012. Therapeutic cancer vaccines: current status and moving forward. J. Natl. Cancer Inst. 104, 599–613. Shahied, L.S., et al., 2004. Bispecific minibodies targeting HER2/neu and CD16 exhibit improved tumor lysis when placed in a divalent tumor antigen binding format. J. Biol. Chem. 279, 53907–53914. Shanafelt, T., et al., 2013. Ofatumumab-based chemoimmunotherapy is effective and well tolerated in patients with previously untreated chronic lymphocytic leukemia (CLL). Cancer 119, 3788–3796. Shawler, D.L., et al., 1985. Human immune responce to multiple injections of murine monoclonal IgG. J. Immunol. 135, 1530–1535. Shields, R.L., et al., 2001. High resolution mapping of the binding site on human IgG1 for Fc gamma RI, Fc gamma RII, Fc gamma RIII, and FcRn and design of IgG1 variants with improved binding to the Fc gamma R. J. Biol. Chem. 276, 6591–6604. Slamon, D.J., et al., 2001. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N. Engl. J. Med. 344, 783–792. Smaglo, B.G., Hwang, J., 2013. Continuum of care with anti-angiogenic therapies in metastatic colorectal cancer. J. Gastrointest. Onco. 4, 299–307. Stavenhagen, J.B., et al., 2008. Enhancing the potency of therapeutic monoclonal antibodies via Fc optimization. Adv. Enzym. Regul. 48, 152–164. Sutherland, M., et al., 2006. Lysosomal trafficking and cysteine protease metabolism confer target-specific cytotoxicity by peptide-linked anti-CD30-auristatin conjugates. J. Biol. Chem. 281, 10540–10547. Swers, J.S., Kellogg, B.A., Wittrup, K.D., 2004. Shuffled antibody libraries created by in vivo homologous recombination and yeast surface display. Nucleic Acids Res. 32, e36–36. Tabernero, J., et al., 2015. Ramucirumab versus placebo in combination with secondline FOLFIRI in patients with metastatic colorectal carcinoma that progressed during or after first-line therapy with bevacizumab, oxaliplatin, and a fluoropyrimidine (RAISE): a randomised, double-blind, multicentre, phase 3 study. Lancet Oncol. 16, 499–508. Tam, C.S., et al., 2008. Long-term results of the fludarabine, cyclophosphamide, and rituximab regimen as initial therapy of chronic lymphocytic leukemia. Blood 112, 975–980. Ujjani, C., et al., 2015. Ofatumumab and bendamustine in previously treated chronic lymphocytic leukemia and small lymphocytic lymphoma. Leuk. Lymphoma. 56, 915–920.
559
Van Cutsem, E., et al., 2012. Addition of aflibercept to fluorouracil, leucovorin, and irinotecan improves survival in a phase III randomized trial in patients with metastatic colorectal cancer previously treated with an oxaliplatin-based regimen. J. Clin. Oncol. 30, 3499–3506. Van Cutsem, E., et al., 2009. Cetuximab and chemotherapy as initial treatment for metastatic colorectal cancer. N. Engl. J. Med. 360, 1408–1417. Verma, S., et al., 2012. Trastuzumab emtansine for HER2-positive advanced breast cancer. N. Engl. J. Med. 367, 1783–1791. Vermorken, J.B., et al., 2008. Platinum-based chemotherapy plus cetuximab in head and neck cancer. N. Engl. J. Med. 359, 1116–1127. Vredenburgh, J.J., et al., 2007. Phase II trial of bevacizumab and irinotecan in recurrent malignant glioma. Clin. Cancer Res. 13, 1253–1259. Ward, E.S., et al., 1989. Binding activities of a repertoire of single immunoglobulin variable domains secreted from Escherichia coli. Nature 341, 544–546. Weber, J.S., et al., 2015. Nivolumab versus chemotherapy in patients with advanced melanoma who progressed after anti-CTLA4 treatment (CheckMate 037): a randomised, controlled, open-label, phase 3 trial. Lancet Oncol. 16, 375–384. Weiner, L.M., Murray, J.C., Shuptrine, C.W., 2012. Antibody-based immunotherapy of cancer. Cell 148, 1081–1084. Weiner, L.M., Surana, R., Wang, S., 2010. Monoclonal antibodies: versatile platforms for cancer immunotherapy. Nat. Rev. Immunol. 10, 317–327. Wierda, W.G., et al., 2010. Ofatumumab as single-agent CD20 immunotherapy in fludarabine-refractory chronic lymphocytic leukemia. J. Clin. Oncol. 28, 1749–1755. Wilke, H., et al., 2014. RAINBOW: a global, phase III, randomized, double-blind study of ramucirumab plus paclitaxel versus placebo plus paclitaxel in the treatment of metastatic gastroesophageal junction (GEJ) and gastric adenocarcinoma following disease progression on first-line platinum- and fluoropyrimidine-containing combination therapy rainbow IMCL CP12-0922 (I4T-IE-JVBE). J. Clin. Oncol. 32, s3. Witzig, T.E., et al., 2002. Treatment with ibritumomab tiuxetan radioimmunotherapy in patients with rituximab-refractory follicular non-Hodgkin’s lymphoma. J. Clin. Oncol. 20, 3262–3269. Woll, P., 2009. Clinical experience of IMGN901 (BB-10901, huN901-DM1) in patients with Merkel cell carcinoma (MCC). Mol. Cancer Ther. 8, B237. Xu, L., et al., 2001. Systemic p53 gene therapy of cancer with immunolipoplexes targeted by anti-transferrin receptor scFv. Mol. Med. 7, 723–734. Yao, S., Zhu, Y., Chen, L., 2013. Advances in targeting cell surface signaling molecules for immune modulation. Nat. Rev. Drug Discov. 12, 130–146. Younes, A., et al., 2012. Results of a pivotal phase II study of brentuximab vedotin for patients with relapsed or refractory Hodgkin’s lymphoma. J. Clin. Oncol. 30, 2183–2189.