Abstract
The clinical success of monoclonal antibody immune checkpoint modulators such as ipilimumab, which targets cytotoxic T lymphocyte-associated antigen 4 (CTLA-4), and the recently approved agents nivolumab and pembrolizumab, which target programmed cell death receptor 1 (PD-1), has stimulated renewed enthusiasm for anticancer immunotherapy, which was heralded by Science as ‘Breakthrough of the Year’ in 2013. As the potential of cancer immunotherapy has been recognized since the 1890s when William Coley showed that bacterial products could be beneficial in cancer patients, leveraging the immune system in the treatment of cancer is certainly not a new concept; however, earlier attempts to develop effective therapeutic vaccines and antibodies against solid tumors, for example, melanoma, frequently met with failure due in part to self-tolerance and the development of an immunosuppressive tumor microenvironment. Increased knowledge of the mechanisms through which cancer evades the immune system and the identification of tumor-associated antigens (TAAs) and negative immune checkpoint regulators have led to the development of vaccines and monoclonal antibodies targeting specific tumor antigens and immune checkpoints such as CTLA-4 and PD-1. This review first discusses the established targets of currently approved cancer immunotherapies and then focuses on investigational cancer antigens and their clinical potential. Because of the highly heterogeneous nature of tumors, effective anticancer immunotherapy-based treatment regimens will likely require a personalized combination of therapeutic vaccines, antibodies and chemotherapy that fit the specific biology of a patient’s disease.
Keywords: antitumor antibody, cancer-testis antigen, cancer vaccine, immunotherapy, oncofetal antigen, tumor-associated antigen
Introduction
The possibility that immunotherapy could be useful for the treatment of cancer was first realized in the 1890s when William B. Coley showed that injecting killed bacterial cultures, known as Coley toxins, had beneficial effects in cancer patients [Nauts et al. 1953]. Decades later, studies performed in chemically induced sarcoma mouse models showed that syngeneic mice injected with irradiated sarcoma cells displayed immunity when later challenged with live sarcoma cells [Foley, 1953; Prehn and Main, 1957; Klein et al. 1960]. The fact that the immunity conferred in this manner was tumor-specific suggested that tumors express unique antigens that are specifically recognized by the adaptive immune system. Indeed, the first such tumor-associated antigen (TAA), known as melanoma antigen 1 (MAGE-1, also known as MAGE-A1), was identified in human melanoma cells by Boon and colleagues in 1991 [van der Bruggen et al. 1991]. Since that seminal discovery, the number of new TAAs has grown steadily to the point where there are now over 400 T-cell-defined human tumor antigenic peptides that have been identified [Vigneron et al. 2013]. Until the late 20th century, the role of immunosurveillance in cancer control had been the subject of much debate [Schreiber et al. 2011]; however, two studies by Schreiber and colleagues involving interferon gamma and tumor immunogenicity in immunocompetent hosts are considered to have played a major role in renewing interest in tumor immunology [Kaplan et al. 1998; Shankaran et al. 2001]. Over the years, a number of different approaches to cancer immunotherapy, including antibodies, cytokines, adoptive cell therapy (ACT) and therapeutic vaccines, have been attempted, but the overall response rates have been largely disappointing [Kirkwood et al. 2012; Savage et al. 2014]. Only recently, as knowledge of tumor biology and immunology has improved, has the complex nature of the interactions between the immune system and cancer come into focus, which has allowed the development of more specifically targeted agents [Kirkwood et al. 2012; Galluzzi et al. 2014].
Nonmutated, shared self-antigens constitute the majority of currently identified TAAs and can be classified into three major categories: (a) tumor-specific or cancer-testis antigens (CTAs), for example, MAGE-1, normally found only in the testes but are aberrantly expressed by a number of different cancers; (b) differentiation antigens expressed by both tumors and the normal differentiated cells from which the tumors arise, for example, melanoma antigen recognized by T cells (MART-1, also known as Melan-A); and (c) self-antigens that are overexpressed by tumors, for example, mucin 1 (MUC1) [Savage et al. 2014]. CTAs were the first type of TAA identified, and there are currently over 200 genes that have been classified as CTAs [Almeida et al. 2009]. It is believed that the observed antigenicity of CTAs is ascribed to the privileged immune status of the testis, where the blood–testis barrier prevents the entry of immune cells. Developing spermatozoa also do not express major histocompatibility complex (MHC) class I molecules, allowing them to evade immunosurveillance by infiltrating T cells [Whitehurst, 2014]. Thus, CTAs make attractive targets for immunotherapy; however, medullary thymic epithelial cells (mTECs) have been reported to express CTAs such as MAGE and New York esophageal squamous cell carcinoma 1 (NY-ESO-1) [Gotter et al. 2004], which suggests that central tolerance to CTAs can develop. A transcriptional regulator known as Aire (autoimmune regulator), which is expressed by mTECs, promotes the promiscuous expression of tissue-restricted antigens such as CTAs [Anderson et al. 2002; Derbinski et al. 2005], resulting in promotion of tolerance to TAAs through thymic deletion of self-reactive T cells [Savage et al. 2014].
In general, active immunotherapy of solid tumors requires the induction of cellular [T helper type 1 (TH1) and cytotoxic T lymphocyte (CTL)-mediated] as opposed to humoral [T helper type 2 (TH2), antibody-mediated] immune responses in order to be effective [Rosenberg, 2001; Kirkwood et al. 2012; Melero et al. 2014]. A TH1-polarized immune response involving CTLs and natural killer (NK) cells mediates the elimination of tumor cells, while a TH2-polarized immune response can have deleterious effects by promoting tumor development and progression [Kirkwood et al. 2012; Curigliano et al. 2013]. This is not to say that antibody-based anticancer immunotherapies are ineffective against solid tumors. Indeed, the past decade has seen the clinical development of numerous monoclonal antibodies directed at growth factors such as vascular endothelial growth factor A (VEGFA; e.g. bevacizumab), growth factor receptors such as human epidermal growth factor receptor (EGFR or HER1; e.g. cetuximab, panitumumab) and EGFR 2 (HER2; e.g. trastuzumab, pertuzumab), and negative immune checkpoint regulators such as cytotoxic T lymphocyte-associated antigen 4 (CTLA-4; e.g. ipilimumab) and programmed cell death receptor 1 (PD-1; e.g. nivolumab, pembrolizumab) [Sliwkowski and Mellman, 2013]. In addition to the direct, modulatory effects on signal transduction, antibody-dependent cellular cytotoxicity (ADCC), mediated through engagement of NK cells and macrophages, also likely contributes to the activity of these antibodies [Kohrt et al. 2012; Sliwkowski and Mellman, 2013]. The tumor-targeting monoclonal antibodies, perhaps the most commonly utilized form of anticancer immunotherapy [Weiner et al. 2010; Vacchelli et al. 2014], are examples of passive immunotherapy, while immune checkpoint inhibitors are considered to be active immunotherapies [Galluzzi et al. 2014].
The recent clinical development and approval of the immune checkpoint inhibitors ipilimumab, which targets CTLA-4, and nivolumab and pembrolizumab, both of which target PD-1, has generated a great deal of excitement in the field of cancer immunotherapy [Swaika et al. 2015]. In a phase III clinical trial, ipilimumab was the first immunotherapeutic to significantly improve overall survival (OS) in patients with advanced, metastatic melanoma [Hodi et al. 2010], and since then both nivolumab [Robert et al. 2015a] and pembrolizumab [Robert et al. 2015b] have demonstrated significant improvement in OS in this patient population. This review will first discuss CTLA-4, PD-1, and other established targets of cancer immunotherapy and then focus on novel tumor antigens and their clinical potential.
Established targets of anticancer immunotherapies
Currently approved anticancer immunotherapies, including monoclonal antibodies and cancer vaccines, target a number of different antigens including CTLA-4, PD-1, EGFR, HER2, VEGF, VEGF-R2, and PAP (prostatic acid phosphatase). Another well-known TAA is BCR-ABL (breakpoint cluster region Abelson tyrosine kinase), for which several small molecule tyrosine kinase inhibitors (TKIs) have been approved and for which immunotherapies are currently under development. Monoclonal antibodies are by far the most commonly approved form of antigen-specific anticancer immunotherapy, with only a single therapeutic cancer vaccine, sipuleucel-T (trade name Provenge®), which targets the prostate cancer antigen PAP, having received Food and Drug Administration (FDA) approval to date. These immunotherapeutic targets and the clinical efficacy of approved treatments are discussed below. For a comprehensive list of the antigen targets of established anticancer immunotherapies, refer to Table 1.
Table 1.
Abbreviations: ALL, acute lymphoblastic leukemia; AML, acute myelogenous leukemia; BCR-ABL, breakpoint cluster region Abelson tyrosine kinase; CLL, chronic lymphocytic leukemia; CTLA-4, cytotoxic T-lymphocyte-associated antigen 4; CRC, colorectal cancer; EGFR, epidermal growth factor receptor; EpCAM, epithelial cell adhesion molecule; HER2, human epidermal growth factor receptor 2; NHL, non-Hodgkin’s lymphoma; NSCLC, non-small cell lung cancer; PAP, prostatic acid phosphatase; PD-1, programmed cell death receptor 1; RCC, renal cell carcinoma; VEGF, vascular endothelial growth factor; VEGF-R2, vascular endothelial growth factor receptor 2.
Immune checkpoint blockade: CTLA-4 and PD-1
A great deal of research in cancer immunotherapy is currently being concentrated on the development of immune checkpoint modulators, particularly those targeting CTLA-4 and PD-1 and its ligands PD-L1/L2. The first checkpoint modulator to be approved, ipilimumab is a monoclonal antibody directed against CTLA-4, a molecule that downregulates the activation of T cells. A phase III clinical trial in patients with metastatic melanoma comparing ipilimumab and gp100 peptide vaccine monotherapies to the combination showed that median OS was significantly increased from 6.4 months (gp100 vaccine alone) to 10.0 months (ipilimumab plus gp100 vaccine). Ipilimumab monotherapy was equally effective [Hodi et al. 2010]. The results of this trial, which was the first demonstration of an immunotherapeutic agent significantly increasing OS in patients with metastatic melanoma, led to the approval of ipilimumab in 2011. A subsequent phase III trial in patients with metastatic melanoma showed that a combination of ipilimumab and dacarbazine significantly increased OS (11.2 versus 9.1 months) compared with dacarbazine alone [Robert et al. 2011].
Still in clinical development, tremelimumab is another anti-CTLA-4 monoclonal antibody. Unlike ipilimumab, tremelimumab failed to demonstrate any significant clinical benefit in patients with advanced melanoma [Ribas et al. 2013], although a survival benefit was revealed in patients achieving higher drug exposures following a retrospective analysis of phase II and III pharmacokinetic data [Calabro et al. 2015]. Tremelimumab is currently being developed for the treatment of malignant mesothelioma [Calabro et al. 2015], where it has demonstrated long-term clinical benefit in phase II studies [Calabro et al. 2013, 2015]. The FDA recently granted tremelimumab Orphan Drug Designation for this indication.
Two new monoclonal antibody-based therapies targeting PD-1, pembrolizumab (formerly lambrolizumab) and nivolumab, were approved by the FDA within the last year. The interaction of PD-1, expressed on the surface of activated T cells, and PD-L1 on the surface of tumor cells results in immunosuppression. An expansion cohort of a phase I trial [Hamid et al. 2013] compared pembrolizumab treatment at doses of 2 or 10 mg in metastatic melanoma patients that had progressed on ipilimumab. At both dose levels, the overall response rate, which was the primary endpoint, was 26% and the treatment was well tolerated [Robert et al. 2014]. The results of this study led to accelerated FDA approval of pembrolizumab for metastatic melanoma in September 2014, representing the first such approval of an anti-PD-1 immunotherapy. Just 3 months later, nivolumab received accelerated FDA approval for advanced melanoma on the basis of phase I/II clinical trial data showing a 31% overall response rate with a median duration of response of two years, 43% two-year survival, and a median OS of approximately 17 months [Topalian et al. 2012, 2014]. A recently published phase III trial comparing a combination of nivolumab and dacarbazine to dacarbazine alone in patients with metastatic melanoma showed significant increases in progression-free survival (PFS) and OS with the combination [Robert et al. 2015a]. Clinical trials evaluating the use of nivolumab and pembrolizumab alone and in combination with other agents such as ipilimumab, TKIs, and anti-PD-L1 agents for the treatment of a wide variety of other cancers are currently ongoing or completed, with promising results [Scott, 2015; Swaika et al. 2015].
The results of clinical trials involving pembrolizumab and nivolumab in the treatment of non-small cell lung cancer (NSCLC) and advanced melanoma were recently reported. Pembrolizumab was found to be superior to ipilimumab in the treatment of patients with advanced melanoma in a phase III clinical trial [Robert et al. 2015b]. The 6-month PFS rate was 47.3% in patients treated with pembrolizumab compared with 26.5% in patients treated with ipilimumab, and the 12-month survival rate for pembrolizumab was 74.1% compared with 58.2% for ipilimumab, differences that were highly significant. A response rate of 33.7% was seen with pembrolizumab treatment compared with 11.9% for ipilimumab, with a lower incidence of adverse events in patients treated with pembrolizumab [Robert et al. 2015b]. In a phase I study in advanced NSCLC, pembrolizumab demonstrated antitumor activity, with a response rate of 45% among patients with a PD-L1 tumor cell expression rate of at least 50%, and a response rate of 19% among all patients. Median PFS was 3.7 months among all patients and 6.3 months in patients with high PD-L1 expression [Garon et al. 2015]. The results of this trial led to the recent FDA approval of pembrolizumab for NSCLC.
Nivolumab was also recently FDA approved for the treatment of NSCLC based on the results of a phase III study showing a significant increase in OS in patients treated with nivolumab compared with docetaxel [Brahmer et al. 2015]. In previously untreated patients with unresectable stage III/IV melanoma, a randomized, double-blind phase III trial showed that nivolumab treatment, either alone or in combination with ipilimumab, was superior to ipilimumab alone, with median PFS increasing significantly from 2.9 months with ipilimumab to 6.9 and 11.5 months with nivolumab and the combination, respectively. Among patients with PD-L1-positive tumors, median PFS was 14.0 months for nivolumab alone and the combination compared to 3.9 months for ipilimumab alone [Larkin et al. 2015]. The combination of nivolumab and ipilimumab recently received accelerated FDA approval for the treatment of BRAF V600 wild-type unresectable or metastatic melanoma based on the results of a phase II trial [Postow et al. 2015]. This was the first approval of an immunotherapy combination for cancer patients.
Investigational, PD-1-targeted agents still in clinical development include pidilizumab, which has been evaluated in phase II clinical trials in patients with B-cell lymphoma [Armand et al. 2013] and metastatic melanoma [Atkins et al. 2014], AMP-514 (MEDI0680), being evaluated in ongoing phase I trials in patients with advanced malignancies [ClinicalTrials.gov identifiers: NCT02118337; NCT02013804], and AMP-224, which is currently being evaluated in a phase I trial [ClinicalTrials.gov identifier: NCT02298946] in patients with metastatic colorectal cancer (CRC) [Homet Moreno et al. 2015].
EGFR receptor tyrosine kinase family
The human epidermal growth factor receptor (EGFR or HER1) is a receptor tyrosine kinase (RTK) that is overexpressed in a variety of different cancers including lung, breast and colon cancer. Specific mutations in the EGFR proto-oncogene result in the RTK becoming constitutively active [Chung, 2015]. Upon activation, EGFR stimulates a number of different signaling pathways, for example, MAPK (mitogen-activated protein kinase), PI3K (phosphoinositide 3-kinase) and STAT (signal transducer and activator of transcription), which lead to cancer cell proliferation and survival [Hynes and Lane, 2005]. This has made EGFR an attractive target for immunotherapy, and indeed several different monoclonal antibodies as well as small molecule TKIs have been developed and approved [Sliwkowski and Mellman, 2013]. Among these, gefitinib, erlotinib and afatinib are TKIs approved for the treatment of NSCLC. While TKIs are quite effective in treating EGFR mutation-positive NSCLC, the development of resistance is virtually universal, commonly through the T790M mutation [Pao et al. 2005]. Interestingly, a peptide antigen derived from the T790M EGFR mutation has been reported to be immunogenic and is a potential target for antigen-specific immunotherapy [Yamada et al. 2013; Ofuji et al. 2015].
Cetuximab and panitumumab
Approved monoclonal antibodies targeting EGFR include cetuximab for colon and head and neck cancers, and panitumumab for colon cancer. Cetuximab received accelerated FDA approval in 2004 for the treatment of metastatic CRC refractory to irinotecan based on the results of clinical trials showing acceptable safety [Saltz et al. 2004] and significant increases in overall response rate and time to progression in patients treated with a combination of cetuximab and irinotecan [Cunningham et al. 2004]. Phase III clinical trial data reaffirming prior results and demonstrating significant increases in PFS and OS in patients with refractory metastatic CRC [Jonker et al. 2007; Sobrero et al. 2008] led to regular FDA approval of single-agent cetuximab in 2007 for the treatment of chemotherapy-refractory metastatic CRC. More recently, cetuximab received FDA approval in 2012 as first-line treatment for metastatic CRC based on the results of the phase III CRYSTAL trial [Van Cutsem et al. 2009] and supporting trials [Bokemeyer et al. 2009, 2012]. A pooled analysis of these trials showed that cetuximab combined with chemotherapy significantly increased overall response rate, PFS and OS in patients with KRAS wild type, EGFR-expressing metastatic CRC [Bokemeyer et al. 2012].
Cetuximab is also used in the treatment of squamous cell carcinoma of the head and neck (SCCHN), having received FDA approval in 2006 for single-agent use in recurrent/metastatic SCCHN refractory to platinum-based chemotherapy, and in combination with radiotherapy for locally or regionally advanced SCCHN. This approval was based on the results of a phase III clinical trial showing significant increases in PFS, median duration of locoregional control and OS with cetuximab plus radiotherapy compared to radiotherapy alone in patients with locoregionally advanced SCCHN [Bonner et al. 2006], and a phase II study in patients with refractory recurrent/metastatic SCCHN showing a 13% response rate and median OS of approximately 6 months with cetuximab monotherapy [Vermorken et al. 2007]. Cetuximab was subsequently approved as first-line therapy in combination with chemotherapy for recurrent/metastatic SCCHN on the strength of the results of a phase III clinical trial showing significant increases in median OS, PFS and response rate with the combination of cetuximab and chemotherapy compared with chemotherapy alone in patients with recurrent/metastatic SCCHN [Vermorken et al. 2008]. The activity of cetuximab has also been evaluated in NSCLC, where a meta-analysis of four phase II/III clinical trials showed that the combination of cetuximab and first-line chemotherapy slightly, yet significantly, increased OS compared with platinum-based chemotherapy alone [Pujol et al. 2014].
Panitumumab is also used in the treatment of metastatic CRC. The results of a phase III clinical trial showing significant increases in PFS and response rate in patients with chemotherapy-refractory metastatic CRC led to the accelerated FDA approval of panitumumab in 2006 [Van Cutsem et al. 2007]. Panitumumab was most recently approved in 2014 as first-line treatment in combination with chemotherapy in KRAS wild-type, metastatic CRC following the results of phase III clinical trials showing a significant increase in PFS, an increase in OS [Douillard et al. 2010], and noninferiority with cetuximab [Price et al. 2014].
Trastuzumab
Trastuzumab is a monoclonal antibody used for the treatment of breast cancer overexpressing the human EGFR 2 (HER2) oncogene, which includes approximately 15–30% of breast cancers and is associated with a poor prognosis [Slamon et al. 1987, 1989]. The FDA first approved trastuzumab in combination with chemotherapy in 1998 for the first-line treatment of patients with metastatic breast cancer. This original approval was based on the results of a phase III clinical trial showing that treatment with trastuzumab plus standard chemotherapy significantly increased time to progression, response rate, duration of response and OS compared with chemotherapy alone [Slamon et al. 2001]. Based on clinical trials data showing significant increases in disease-free survival (DFS) and OS [Romond et al. 2005; Perez et al. 2011, 2014], trastuzumab later received FDA approval in 2006 for the treatment of breast cancer in the adjuvant setting in combination with chemotherapy. Trastuzumab was most recently approved in 2010 for the first-line treatment of metastatic gastric or gastro-esophageal junction adenocarcinoma in combination with chemotherapy based on a phase III clinical trial showing significantly increased OS following treatment with trastuzumab combined with chemotherapy [Bang et al. 2010]. Currently, trastuzumab in combination with pembrolizumab is being evaluated in a phase I/II clinical trial in patients with trastuzumab-resistant, HER2-positive metastatic breast cancer [ClinicalTrials.gov identifier: NCT02129556]. It has been previously shown that trastuzumab and anti-PD-1 treatment are synergistic in mice [Stagg et al. 2011].
Necitumumab
Still in clinical development, necitumumab is a second-generation anti-EGFR monoclonal antibody currently being evaluated for the treatment of NSCLC. Recently published data from a phase III clinical trial in 1093 patients with stage IV squamous NSCLC showed that necitumumab in combination with chemotherapy as first-line treatment resulted in a modest survival benefit, with a median OS of 11.5 months in patients treated with necitumumab plus chemotherapy compared with 9.9 months in patients treated with chemotherapy alone, a difference that was significant [Thatcher et al. 2015]. In patients with stage IV nonsquamous NSCLC, however, combining necitumumab with standard chemotherapy as first-line treatment provided no additional clinical benefit compared with chemotherapy alone [Paz-Ares et al. 2015].
VEGFA and VEGF-R2
Vascular endothelial growth factor A (VEGFA) plays a critical role in angiogenesis, and it is the target of the monoclonal antibody bevacizumab. In addition to its anti-angiogenic effects [Ferrara et al. 2004], bevacizumab has been found to augment tumor infiltration by T and B lymphocytes [Manzoni et al. 2010; Hodi et al. 2014] and to inhibit regulatory T cells (Tregs) [Terme et al. 2013]. Bevacizumab has been FDA approved for the treatment of CRC (2004), glioblastoma (2009), cervical cancer (2014), lung cancer (2006), renal cell cancer (2009), breast cancer (2008), and ovarian cancer (2014).
Bevacizumab was initially approved for the first-line treatment of metastatic CRC in combination with chemotherapy based on a phase III study showing significant increases in PFS and median OS compared with chemotherapy alone [Hurwitz et al. 2004]. Shortly thereafter, bevacizumab received approval as second-line treatment in combination with chemotherapy for patients with previously treated metastatic CRC following the results of a phase III clinical trial showing significant improvement in PFS and median OS [Giantonio et al. 2007] compared with chemotherapy alone. Two years later, another Phase III trial in patients with recurrent or advanced NSCLC showed significant increases in response rate, PFS and OS following treatment with bevacizumab plus chemotherapy [Sandler et al. 2006], which led to approval of bevacizumab combined with chemotherapy as first-line treatment of recurrent/advanced NSCLC. The approval of bevacizumab as first-line treatment of metastatic breast cancer was supported by the results of a phase III clinical trial in patients with HER2-negative metastatic breast cancer showing that treatment with bevacizumab and paclitaxel led to significant increases in PFS and objective response rate (ORR) compared with paclitaxel alone [Miller et al. 2007]. A phase II study used to support the 2009 FDA approval of bevacizumab combined with chemotherapy for patients with recurrent glioblastoma showed that treatment with bevacizumab combined with chemotherapy resulted in improved PFS and ORR, differences that were significant compared to expected effects of salvage chemotherapy [Friedman et al. 2009]. The same year, bevacizumab combined with interferon α was approved as first-line treatment of metastatic renal cell carcinoma (RCC) based on phase III data demonstrating significant improvement in PFS [Escudier et al. 2007]. No significant improvement in OS was observed in this trial [Escudier et al. 2010]. Most recently, bevacizumab was approved in 2014 for the treatment of recurrent or metastatic cervical cancer and recurrent, platinum-refractory ovarian carcinoma based on phase III clinical trials showing significant increases in OS and response rate in cervical cancer [Tewari et al. 2014], and significant improvement in PFS and ORR in ovarian cancer [Pujade-Lauraine et al. 2014], compared with chemotherapy alone. Like trastuzumab, bevacizumab and anti-PD-1 combination treatment is currently being clinically evaluated [ClinicalTrials.gov identifiers: NCT02210117; NCT02039674; NCT02348008].
VEGF-R2, also known as KDR (kinase insert domain receptor) and Flk-1 (fetal liver kinase 1), is an RTK and the primary receptor through which VEGF mediates its angiogenic, mitogenic, and permeability-enhancing effects [Ferrara et al. 2004]. A monoclonal antibody that targets VEGF-R2, ramucirumab has received FDA approval for the second-line treatment of gastric cancer and NSCLC within the past year. Ramucirumab was approved as monotherapy and in combination with paclitaxel for the treatment of gastric or gastroesophageal junction adenocarcinoma after progression on first-line chemotherapy based on the results of two phase III trials showing significant increases in median OS compared with placebo [Fuchs et al. 2014] and paclitaxel plus placebo [Wilke et al. 2014]. Efficacy as second-line treatment for metastatic NSCLC after progression on platinum-based chemotherapy was also demonstrated in a phase III trial showing significant improvement in PFS and OS with the combination of docetaxel and ramucirumab compared with docetaxel plus placebo [Garon et al. 2014].
PAP
PAP is the target of the only currently FDA-approved therapeutic anticancer vaccine, sipuleucel-T (Provenge®). Sipuleucel-T was approved for the treatment of asymptomatic and minimally symptomatic metastatic castration-resistant prostate cancer in 2010 based on the results of the phase III IMPACT trial showing a significant increase in median OS in patients treated with sipuleucel-T compared with those treated with placebo [Kantoff et al. 2010a]. To prepare sipuleucel-T, peripheral blood mononuclear cells from the patient are cultured with a fusion protein that incorporates PAP and GM-CSF (granulocyte-macrophage colony-stimulating factor). The patient is then infused with these cells, and the process is repeated for three cycles. Thus, sipuleucel-T is actually a combination of a cell-based vaccine and autologous ACT [Melero et al. 2014]. Interestingly, in addition to immune responses against the primary antigenic target PAP, humoral immune responses against several secondary antigens such as PSA (prostate specific antigen), K-ras and KLK2/hK2 were also observed to be elevated in patients treated with sipuleucel-T but not control in the phase III IMPACT trial, and these responses were associated with improved OS [GuhaThakurta et al. 2015].
Investigational tumor antigens and their clinical potential
In addition to the established tumor antigens for which targeted immunotherapies have already been approved (Table 1), there are numerous novel tumor antigens currently being investigated as potential targets for new immunotherapies, including MUC1, PD-L1, lymphocyte activation gene 3 (LAG-3), T-cell immunoglobulin domain and mucin domain 3 (TIM-3), V-domain immunoglobulin-containing suppressor of T-cell activation (VISTA), NY-ESO-1, carcinoembryonic antigen (CEA), PSA, MAGE-A3, 5T4, survivin and indoleamine-2,3-dioxygenase (IDO1). These selected antigens and their clinical potential as targets for investigational immunotherapies are discussed below. Table 2 includes a more complete listing of tumor antigen targets for which immunotherapies are currently being explored.
Table 2.
Antigen | Cancer types | Targeted agents | References |
---|---|---|---|
A2aR | NSCLC | PBF-509 | [Pinna, 2014] |
AKAP4 | NSCLC Ovarian |
Preclinical | [Agarwal et al. 2013; Mirandola et al. 2015] |
BAGE | Glioblastoma Ovarian |
Preclinical | [Zhang et al. 2010; Akiyama et al. 2014] |
BORIS | Prostate, Lung Esophageal |
Preclinical | [Okabayashi et al. 2012; Cheema et al. 2014; Lee et al. 2014] |
CD22 | ALL | Epratuzumab Moxetumomab Inotuzumab ozogamicin |
[Raetz et al. 2008; Advani et al. 2014; Annesley and Brown, 2015; Raetz et al. 2015] |
CD73 | Advanced solid tumors | MEDI9447 | [Hay et al. 2015] |
CD137 | Advanced solid tumors | Urelumab PF-05082566 |
[Yonezawa et al. 2015] |
CEA | CRC | PANVAC™ Ad5-[E1-, E2b-]-CEA(6D) |
[Turriziani et al. 2012; Morse et al. 2013a, 2013b] |
CS1 | Multiple myeloma | Elotuzumab | [Veillette and Guo, 2013] |
CTLA-4 | Malignant mesothelioma | Tremelimumab | [Calabro et al. 2015] |
EBAG9 | Bladder | Preclinical | [Miyazaki et al. 2014] |
EGF | NSCLC | CIMAvax | [Gonzalez et al. 2007; Neninger Vinageras et al. 2008; Cheng and Kananathan, 2012; Ruiz et al. 2014] |
EGFR | NSCLC | Necitumumab | [Thatcher et al. 2015] |
GAGE | Cervical | Preclinical | [Kular et al. 2010] |
GD2 | Neuroblastoma Retinoblastoma Melanoma other solid tumors |
Dinutuximab, hu3F8 hu14.18-IL-2, 3F8/OKT3BsAb anti-GD2 CAR GD2-KLH |
[Suzuki and Cheung, 2015] |
gp100 | Melanoma | gp100:209-217(210M) | [Schwartzentruber et al. 2011] |
HPV-16 | Cervical SCCHN |
HPV-16 (E6, E7) TG4001, Lm-LLO-E7 pNGVL4a-CRT/E7, INO-3112 |
[Kenter et al. 2009; Brun et al. 2011; Bagarazzi et al. 2012; Zaravinos, 2014] |
HSP105 | CRC Bladder |
Preclinical | [Zappasodi et al. 2011; Kawai et al. 2014; Sawada et al. 2014] |
IDH1 | Glioma | IDH1(R132H) p123-142 | [Schumacher et al. 2014] |
Idiotype (NeuGcGM3) | NSCLC, Breast Melanoma |
Racotumomab | [Alfonso et al. 2002, 2014; Diaz et al. 2003] |
IDO1 | Breast, Melanoma NSCLC |
Indoximod INCB024360 IDO1 peptide vaccine |
[Iversen et al. 2014, 2015; Soliman et al. 2014] |
KIR | Lymphoma | Lirilumab | [Kohrt et al. 2014] |
LAG-3 | Breast, Hemato- logical, Advanced solid tumors |
BMS-986016 IMP321 |
[Brignone et al. 2010; Creelan, 2014; Nguyen and Ohashi, 2015] |
LY6K | Gastric SCCHN |
LY6K-177 peptide LY6K, CDCA1, IMP3 |
[Ishikawa et al. 2014; Yoshitake et al. 2015] |
MAGE-A3 | Melanoma NSCLC |
recMAGE-A3 Zastumotide |
[Kruit et al. 2013; Ulloa-Montoya et al. 2013; Vansteenkiste et al. 2013; Melero et al. 2014] |
MAGE-C2 | Gastric, Melanoma Multiple myeloma |
Preclinical | [Zhang et al. 2013; Ghadban et al. 2014; Reinhard et al. 2014] |
MAGE-D4 | CRC | Preclinical | [Zhang et al. 2014] |
Melan-A | Melanoma | MART-1 (26-35, 27L) | [Tarhini et al. 2012; Romano et al. 2014] |
MET | NSCLC | Onartuzumab Tivantinib |
[Scagliotti et al. 2012; Spigel et al. 2013; Stinchcombe, 2014] |
MUC1 | NSCLC, Breast Prostate |
Tecemotide, TG4010 PANVAC™ |
[Kimura and Finn, 2013; Butts et al. 2014; Wurz et al. 2014] |
MUC4 | Pancreatic | Preclinical | [Wu et al. 2009; Torres et al. 2012; Zhu et al. 2014] |
MUC16 | Ovarian | Abagovomab Oregovomab |
[Buzzonetti et al. 2014; Felder et al. 2014] |
NY-ESO-1 | Ovarian Melanoma |
NY-ESO-1/ISCOMATRIX™ rV-NY-ESO-1; rF-NY-ESO-1 |
[Robbins et al. 2011; Odunsi et al. 2012; Klein et al. 2015] |
PD-1 | B-cell lymphoma Melanoma, CRC |
Pidilizumab AMP-224, AMP-514 |
[Armand et al. 2013; Homet Moreno et al. 2015] |
PD-L1 | NSCLC, RCC Bladder, Breast Melanoma, SCCHN |
BMS-936559, Atezolizumab Durvalumab, Avelumab |
[Cha et al. 2015; Ibrahim et al. 2015; Sunshine and Taube, 2015; Swaika et al. 2015] |
PRAME | NSCLC | Preclinical | [De Pas et al. 2012] |
PSA | Prostate | PROSTVAC®-VF | [Gulley et al. 2010; Kantoff et al. 2010b; Shore, 2014] |
ROR1 | CLL, Pancreatic Lung, Breast |
Preclinical | [Hojjat-Farsangi et al. 2014; Berger et al. 2015; Shabani et al. 2015] |
Sialyl-Tn | Breast | Theratope | [Miles et al. 2011; Ibrahim et al. 2013] |
SPAG-9 | Prostate, CRC NSCLC, Ovarian |
Preclinical | [Garg et al. 2007; Kanojia et al. 2011; Wang et al. 2013; Chen et al. 2014] |
SSX1 | Prostate Multiple myeloma |
Preclinical | [Smith et al. 2011; He et al. 2014] |
Survivin | Melanoma Glioma, Solid tumors |
EMD640744 Trivalent peptide vaccine Tripeptide vaccine |
[Becker et al. 2012; Lennerz et al. 2014; Pollack et al. 2014] |
Telomerase | Pancreatic | Tertomotide | [Melero et al. 2014; Staff et al. 2014] |
TIM-3 | Melanoma, NHL NSCLC | Preclinical | [Sakuishi et al. 2010; Ngiow et al. 2011; Gao et al. 2012; Fourcade et al. 2014; Jiang et al. 2015] |
VISTA | Melanoma, Bladder | Preclinical | [Le Mercier et al. 2014; Lines et al. 2014b] |
WT1 | Ovarian, Uterine, AML Multiple myeloma |
WT1 peptide vaccine | [Coosemans et al. 2014; Di Stasi et al. 2015] |
XAGE-1b | Prostate | DC-based tumor vaccine | [Zhou et al. 2008; Xie and Wang, 2015] |
5T4 | RCC, CRC Prostate |
TroVax®
Naptumomab estafenatox |
[Amato et al. 2010; Zhang et al. 2012; Harrop et al. 2013; Eisen et al. 2014; Rowe and Cen, 2014; Stern et al. 2014] |
Abbreviations: A2aR, adenosine A2a receptor; AKAP4, A kinase anchor protein 4; AML, acute myelogenous leukemia; ALL, acute lymphoblastic leukemia; BAGE, B melanoma antigen; BORIS, brother of the regulator of imprinted sites; CEA, carcinoembryonic antigen; CLL, chronic lymphocytic leukemia; CRC, colorectal cancer; CS1, CD2 subset 1; CTLA-4, cytotoxic T-lymphocyte-associated antigen 4; EBAG9, estrogen receptor binding site associated antigen 9; EGF, epidermal growth factor; EGFR, epidermal growth factor receptor; NSCLC, non-small cell lung cancer; GAGE, G antigen; GD2, disialoganglioside GD2; gp100, glycoprotein 100; HPV-16, human papillomavirus 16; HSP105, heat-shock protein 105; IDH1, isocitrate dehydrogenase type 1; IDO1, indoleamine-2,3-dioxygenase 1; KIR, killer cell immunoglobulin-like receptor; LAG-3, lymphocyte activation gene 3; LY6K, lymphocyte antigen 6 complex K; MAGE-A3, melanoma antigen 3; MAGE-C2, melanoma antigen C2; MAGE-D4, melanoma antigen D4; Melan-A/MART-1, melanoma antigen recognized by T-cells 1; MET, N-methyl-N’-nitroso-guanidine human osteosarcoma transforming gene; MUC1, mucin 1; MUC4, mucin 4; MUC16, mucin 16; NHL, non-Hodgkin lymphoma; NY-ESO-1, New York esophageal squamous cell carcinoma 1; PD-1, programmed cell death receptor 1; PD-L1, programmed cell death receptor ligand 1; PRAME, preferentially expressed antigen of melanoma; PSA, prostate specific antigen; RCC, renal cell carcinoma; ROR1, receptor tyrosine kinase orphan receptor 1; SCCHN, squamous cell carcinoma of the head and neck; SPAG-9, sperm-associated antigen 9; SSX1, synovial sarcoma X-chromosome breakpoint 1; TIM-3, T-cell immunoglobulin domain and mucin domain-3; VISTA, V-domain immunoglobulin-containing suppressor of T-cell activation; WT1, Wilms’ Tumor-1; XAGE-1b, X chromosome antigen 1b.
MUC1
Mucin 1 (MUC1) is one of the best-characterized TAAs, and it is overexpressed and aberrantly glycosylated in over 90% of adenocarcinomas such as lung and breast cancer [Szabo, 2003; Brayman et al. 2004]. MUC1 is a transmembrane glycoprotein expressed in several epithelial tissues, but the normal glycosylation pattern shields the peptide core of the extracellular domain from immunosurveillance. In cancer, the aberrant underglycosylation of MUC1 leads to exposure of the immunogenic tandem repeat regions of the core peptide, making MUC1 an attractive target for immunotherapies [Apostolopoulos and McKenzie, 1994; Vlad et al. 2004].
While several different MUC1-targeted immunotherapies have been assessed in clinical trials [Kimura and Finn, 2013], the most advanced agent is tecemotide, an antigen-specific, liposome-based, peptide cancer vaccine [Wurz et al. 2014] that has been evaluated in a phase III clinical trial as maintenance therapy for patients with locally advanced, unresectable stage IIIA/IIIB NSCLC who had not progressed following primary chemoradiotherapy [Butts et al. 2014]. The results failed to demonstrate significant improvement in OS with tecemotide compared with placebo; however, a predefined subgroup analysis of the patients who received concurrent chemoradiotherapy followed by tecemotide showed a significant survival benefit, while no survival benefit was seen with sequential chemoradiotherapy [Butts et al. 2014]. After approximately 20 months of additional follow up, the survival benefit of tecemotide following concurrent chemoradiotherapy remained significant [Mitchell et al. 2015]. These results suggest that the timing of radiotherapy, chemotherapy and immunotherapy is important and that monitoring immune status may be necessary when designing treatment regimens combining immunotherapy with chemoradiotherapy [Kao et al. 2014, 2015]. Another phase III trial, recently discontinued, was initiated to study tecemotide maintenance therapy following concurrent chemoradiotherapy in NSCLC [DeGregorio et al. 2014].
Immune checkpoints: PD-L1; LAG-3; TIM-3; VISTA
PD-L1
As already mentioned, the development of immune checkpoint inhibitors is currently the focus of intense research efforts. The interaction of PD-L1, which is expressed on the surface of tumor cells, with PD-1 on activated T cells results in immunosuppression and tumor immune escape [Iwai et al. 2002; Swaika et al. 2015]. PD-L1 expression has been found to be inducible by interferon γ [Muhlbauer et al. 2006]. Several investigational immunotherapies targeting PD-L1 are presently in clinical development for metastatic melanoma, NSCLC, RCC and bladder cancer, among others.
BMS-936559 (MDX-1105) is a monoclonal antibody that inhibits the binding of PD-L1 to both PD-1 and CD80. The safety and antitumor activity of BMS-936559 in patients with a variety of advanced cancers have been evaluated in a phase I clinical trial involving 207 patients [Brahmer et al. 2012]. In NSCLC, an ORR of 10% (all partial responses) and PFS rate of 31% at 24 weeks were observed. Patients with RCC showed a 12% ORR and 53% progression-free response rate at 24 weeks. In ovarian cancer, an ORR of only 6% was observed [Brahmer et al. 2012]. No more recent clinical data regarding BMS-936559 in cancer therapy have emerged, and it appears that clinical development of this agent for oncology applications has been discontinued [Mullard, 2013; Sunshine and Taube, 2015].
Atezolizumab (MPDL3280A) is an anti-PD-L1 monoclonal antibody that has demonstrated activity against a variety of advanced cancers in phase I clinical trials. The Fc region of the antibody is modified to prevent both ADCC and complement-dependent cytotoxicity (CDC) [Lu et al. 2014], which prevents the killing of immune cells that also express PD-L1, such as activated T cells. Atezolizumab is currently being evaluated in patients with locally advanced or metastatic solid tumors in an ongoing phase I study. In a cohort of patients with metastatic urothelial bladder cancer, an ORR of 43% was observed at 6 weeks, increasing to 52% at 12 weeks of treatment [Powles et al. 2014]. As there had been no significant advances in the treatment of metastatic urothelial cancer in decades, these results led to breakthrough designation status by the FDA in June 2014. Updated response and survival data from this study after an additional 8 months of follow up continued to show promising PFS and OS results [Petrylak et al. 2015]. For patients with NSCLC, an ORR of 23% and PFS rate of 45% at 24 weeks was seen with atezolizumab treatment [Herbst et al. 2014], and the most recent data continued to show clinical benefit in NSCLC [Horn et al. 2015]. Atezolizumab has also been studied in combination with first-line chemotherapy in metastatic NSCLC patients, and was found to improve ORR in all combinations tested in a phase Ib trial [Liu et al. 2015b]. In patients with metastatic melanoma or RCC, ORRs of 30% and 14%, and PFS rates of 41% and 48% at 24 weeks, respectively, were observed [Herbst et al. 2014]. Patients in the triple-negative breast cancer cohort had an ORR of 19% and a PFS rate of 27% at 24 weeks [Emens et al. 2015].
Preliminary results from two phase II clinical trials of atezolizumab in patients with advanced NSCLC were recently reported. In both chemotherapy-naïve and previously treated patients, an increasing ORR to atezolizumab treatment was associated with increasing PD-L1 expression [Spigel et al. 2015]. Compared with docetaxel in previously treated NSCLC, treatment with atezolizumab was found to increase OS, PFS and ORR in association with PD-L1 expression [Spira et al. 2015]. There are currently several other phase I/II and phase III clinical trials of atezolizumab currently underway [Swaika et al. 2015].
Durvalumab (MEDI4736) is an anti-PD-L1 monoclonal antibody similar to atezolizumab, with an Fc region modification that prevents ADCC and CDC. In a phase I clinical trial, response rates of 13% and 14% were observed in patients with metastatic NSCLC and SCCHN, respectively [Lee and Chow, 2014]. A phase II study [ClinicalTrials.gov identifier: NCT02336165] in patients with glioblastoma multiforme is currently underway [Reardon et al. 2015]. Durvalumab has also entered a phase III clinical trial in combination with concurrent chemoradiotherapy in patients with stage III unresectable NSCLC [ClinicalTrials.gov identifier: NCT02125461] and a phase II/III clinical trial [ClinicalTrials.gov identifier: NCT02154490] in patients with recurrent stage IIIB/IV squamous cell NSCLC [Lee and Chow, 2014; Ibrahim et al. 2015]. Other ongoing phase II and phase III clinical trials of durvalumab include third-line treatment of stage IIIB/IV NSCLC [ClinicalTrials.gov identifier: NCT02087423], second-line treatment of SCCHN [ClinicalTrials.gov identifier: NCT02207530; Ibrahim et al. 2015] and treatment of advanced NSCLC alone or in combination with tremelimumab [ClinicalTrials.gov identifier: NCT02352948; Planchard et al. 2015].
A fourth anti-PD-L1 monoclonal antibody, avelumab (MSB0010718C), is currently in phase I development in subjects with metastatic or locally advanced solid tumors [ClinicalTrials.gov identifier: NCT01772004], with a separate phase I trial in Japanese subjects [Lu et al. 2014]. Results of this phase I trial in extensively pretreated patients with a variety of different tumors have so far demonstrated that avelumab has an acceptable safety profile [Kelly et al. 2015]. A phase Ib expansion study in advanced NSCLC patients who had progressed after platinum-based chemotherapy demonstrated increased activity with avelumab treatment in patients with PD-L1-positive tumors [Gulley et al. 2015]. A phase II study of avelumab in patients with metastatic Merkel cell carcinoma is currently underway [Kaufman et al. 2015]. Interestingly, unlike the other anti-PD-L1 antibodies being developed, avelumab appears to possess ADCC activity [Boyerinas et al. 2015].
LAG-3
LAG-3 is a negative regulator of effector T-cell function [Sierro et al. 2011], making it an attractive target for immunotherapy. LAG-3 is expressed on the membranes of activated T cells and is upregulated on exhausted T cells [Nguyen and Ohashi, 2015]. Because LAG-3 is also expressed on activated Tregs, LAG-3 blockade may inhibit the suppressive effects of Tregs in addition to the direct action on effector cells [Huang et al. 2004]. The major ligand of LAG-3 is MHC class II, and their interaction may play a role in modulating dendritic cell function [Goldberg and Drake, 2011; Camisaschi et al. 2014]. A monoclonal antibody targeting LAG-3, known as BMS-986016, is currently in early phase I clinical development. Ongoing clinical trials are investigating single agent BMS-986016 for hematological malignancies and a combination of nivolumab and BMS-986016 in patients with advanced solid tumors [Creelan, 2014; Nguyen and Ohashi, 2015]. In addition to the membrane-bound form of LAG-3, the LAG3 gene also codes for a soluble variant, sLAG-3, that functions as an immune adjuvant [Nguyen and Ohashi, 2015]. A sLAG-3 immunoglobulin fusion protein (IMP321) is in early clinical development as an immune adjuvant. In a Phase I clinical trial evaluating a combination of taxane-based chemotherapy and IMP321 in women with metastatic breast cancer, a 50% ORR was observed [Brignone et al. 2010].
TIM-3
TIM-3 is an inhibitory immune checkpoint receptor that limits the duration and magnitude of TH1 CD4+ and CD8+ CTL cellular responses [Anderson, 2014]. Galectin-9 has been identified as the ligand for TIM-3, and it is through this pathway that TH1 immunity may be suppressed [Zhu et al. 2005]. The expression of TIM-3 on dysfunctional CD8+ T cells and Tregs makes it an attractive target for anticancer immunotherapy. Preclinical animal models have shown that antibody inhibition of TIM-3 is very effective in suppressing tumor growth [Ngiow et al. 2011]. Synergistic antitumor effects in multiple cancer types have been observed in preclinical models with the combination of anti-PD-1/PD-L1 and TIM-3 blockade [Sakuishi et al. 2010; Ngiow et al. 2011; Jing et al. 2015]. Clinically, increased expression of TIM-3 and PD-1 in melanoma patients has been associated with tumor NY-ESO-1-specific CD8+ T-cell dysfunction [Fourcade et al. 2014]. In NSCLC, TIM-3 is expressed on tumor-infiltrating CD4+ and CD8+ T cells [Gao et al. 2012], and the presence of CD4+ T cells that express TIM-3 is strongly correlated with advanced tumor grade in NSCLC [Gao et al. 2012] and poor survival in follicular B-cell non-Hodgkin lymphoma [Yang et al. 2012]. Thus, TIM-3 has great clinical potential as a target of novel immunotherapies [Cheng and Ruan, 2015].
VISTA
VISTA is a newly identified negative immune checkpoint regulator [Lines et al. 2014b]. Unlike PD-L1, VISTA is expressed primarily in hematopoietic cells in both mice [Wang et al. 2011] and humans [Lines et al. 2014a]. In murine models of melanoma and bladder cancer, leukocytes within the tumor microenvironment and in tumor-draining lymph nodes were found to have elevated VISTA expression. Treatment with an anti-VISTA monoclonal antibody resulted in tumor growth inhibition, especially in combination with a tumor vaccine [Le Mercier et al. 2014]. In these studies, VISTA blockade was found to be effective with no detectable expression of VISTA and high expression levels of PD-L1 on tumor cells. This observation could have important clinical implications for combination treatments targeting the VISTA and PD-1/PD-L1 checkpoint pathways, allowing for potential synergy due to the fact that these pathways operate independently [Lines et al. 2014b; Liu et al. 2015a].
CTAs: NY-ESO-1 and MAGE-A3
CTAs constitute the majority of the currently identified TAAs, and numerous CTAs are being investigated as potential targets of new anticancer immunotherapies for a variety of malignancies. Among these, immunotherapies targeting NY-ESO-1 and MAGE-A3 have reached the clinical stage of development. These antigens are normally expressed only in adult testis germ cells and become aberrantly re-expressed in various cancers such as lung, bladder and melanoma [Jungbluth et al. 2001].
NY-ESO-1
NY-ESO-1 is encoded by the CTAG1B gene and was first identified in 1997 [Chen et al. 1997]. Expressed in a variety of carcinomas and sarcomas, NY-ESO-1 is highly immunogenic and is thus a logical target for anticancer immunotherapy [Nicholaou et al. 2006; Endo et al. 2015]. In a phase I clinical trial in patients with advanced synovial cell sarcoma and melanoma, subjects were administered autologous ACT using cells transduced with a T-cell receptor (TCR) targeting NY-ESO-1. Objective response rates of 67% (4/6) and 45% (5/11) were observed for synovial cell sarcoma and melanoma patients, respectively [Robbins et al. 2011]. Autologous ACT has also been performed using NY-ESO-1-specific CD4+ cells [Hunder et al. 2008]. A prime-boost vaccine strategy was assessed in patients with stage III/IV melanoma and stage II-IV ovarian carcinoma in a phase II clinical trial. Patients were primed with a recombinant NY-ESO-1-expressing vaccinia virus and then boosted with an NY-ESO-1-expressing fowlpox virus. In the melanoma patients, CD8+ T-cell responses increased from 40% pretreatment to 88% following vaccination. Median OS was significantly increased in the melanoma and ovarian cancer patients who had immune responses following treatment [Odunsi et al. 2012]. An NY-ESO-1-specific vaccine (NY-ESO-1/ISCOMATRIX™) in combination with cyclophosphamide has also been evaluated in advanced melanoma in phase II. Treatment led to a significant increase in antigen-specific CD4+ T-cell immune responses [Klein et al. 2015] compared to a cohort of patients treated with the vaccine alone [Nicholaou et al. 2009]. A prime-boost treatment strategy employing NY-ESO-1/ISCOMATRIX™ in high-risk, resected melanoma patients was found to augment CD8+ T-cell immune responses [Chen et al. 2015].
MAGE-A3
MAGE-A3 is a CTA that has been extensively clinically investigated as a target for immunotherapy in melanoma and lung cancer. In a clinical trial involving nine patients with advanced cancers, five responses were observed following autologous ACT with T cells transduced with anti-MAGE-A3 TCRs. However, neurological toxicities were seen in three patients, resulting in two deaths due to previously unknown expression of MAGE family antigens in the brain [Morgan et al. 2013]. A Phase II study examined vaccination with recombinant MAGE-A3 protein combined with either AS15 [a combination of QS21 saponin, monophosphoryl lipid A (MPL-A), and CpG7909, a Toll-like receptor (TLR)-9 agonist, in a liposomal formulation] or AS02B (a combination of QS21 saponin and MPL-A, a TLR-4 agonist) immunostimulants in patients with stage III/IV melanoma. Trends towards greater ORR, PFS and median OS were observed in the patients who received the AS15 immunostimulant compared with AS02B [Kruit et al. 2013]. The peptide vaccine zastumotide (GSK2132231A) is being evaluated in a phase III clinical trial in patients with resected stage IIIB/IIIC melanoma. This trial failed to demonstrate a significant prolongation of DFS in patients with positive expression of MAGE-A3 [Melero et al. 2014], but it is ongoing in a subset of patients who are positive for a predictive marker of response [Ulloa-Montoya et al. 2013]. A phase II clinical trial of zastumotide (GSK1572932A) in patients with resected stage IB/II NSCLC exhibited a trend toward positive treatment outcomes compared with placebo [Vansteenkiste et al. 2013]; however, a large phase III study in patients with resected stage IB-IIIA NSCLC failed to meet its primary endpoint of significant prolongation of DFS, resulting in termination of the study [Melero et al. 2014]. Clinical development of zastumotide appears to have been discontinued.
Oncofetal antigens: CEA and 5T4
Oncofetal antigens are so-called because though widely expressed during fetal development, normal expression is generally limited in adults. In cancer, the expression of these antigens becomes upregulated, making them potential targets for immunotherapeutic intervention.
CEA
CEA is a well-known oncofetal antigen normally expressed in the stomach, tongue, esophagus, cervix and prostate in adults, but becomes highly overexpressed in colorectal and gastric cancers [Beauchemin and Arabzadeh, 2013]. Clinically, CEA has been investigated extensively as an immunotherapeutic target for colorectal and various other cancers [Turriziani et al. 2012]. In a phase I/II clinical trial, treatment with an adenoviral gene delivery platform encoding the CEA antigen produced cell-mediated immunity in 61% of patients with advanced CRC and a 48% OS rate at 12 months [Morse et al. 2013a]. A phase II study in CRC patients who were disease-free following metastasectomy and perioperative chemotherapy compared the effectiveness of autologous dendritic cells modified with PANVAC™, a poxvector encoding both CEA and MUC1, to the PANVAC™ poxvector administered in combination with GM-CSF. Survival and recurrence-free survival were similar between the two treatment groups, and vaccinated patients overall experienced superior survival compared with a contemporary group of patients not treated with immunotherapy [Morse et al. 2013b]. In a recently reported phase II trial comparing treatment with PANVAC™ in combination with docetaxel to docetaxel alone in 48 patients with metastatic breast cancer, patients treated with the combination experienced improved PFS compared with docetaxel alone, a result which, while not significant, was suggestive of a clinical benefit [Heery et al. 2015]. Increased CEA expression has been associated with shorter relapse-free survival in breast cancer patients [Saadatmand et al. 2013].
5T4
The oncofetal antigen 5T4 was identified by screening for shared surface molecules in human trophoblasts and human cancer cells [Stern et al. 2014]. A heavily glycosylated, membrane-bound protein, 5T4 is highly expressed in cervical, colorectal, gastric, ovarian, prostate, lung and renal cancers [Southall et al. 1990]. The expression of 5T4 has been associated with epithelial mesenchymal transition (EMT), which is involved in the metastasis of epithelial cancers [Nieto and Cano, 2012]. Three different 5T4-based immunotherapeutic strategies are being clinically evaluated: a vaccine known as TroVax® (modified vaccinia virus Ankara- MVA), an antibody–superantigen [Staphylococcal Enterotoxin A (SEA)] fusion protein, and an antibody–drug conjugate (ADC) combining a 5T4-specific monoclonal antibody with a tubulin inhibitor.
5T4 vaccine (TroVax®)
Favorable trends toward clinical benefit have been observed in early phase I and phase II clinical trials of the TroVax® 5T4-MVA vaccine in advanced RCC [Zhang et al. 2012], CRC [Rowe and Cen, 2014], and castration-resistant prostate cancer [Harrop et al. 2013]. To determine whether treatment with TroVax® can improve survival, a phase III clinical trial in patients with metastatic RCC compared treatment with TroVax® to standard of care treatment [Amato et al. 2010]. While this trial failed to demonstrate any significant effects of TroVax® on OS, a subgroup of patients with good prognosis and receiving concurrent IL-2 experienced a significant survival benefit compared with placebo. The patients with the greatest increases in 5T4-specific antibodies also experienced a survival benefit with TroVax® compared with placebo, which is consistent with a pooled analysis of prior phase I/II TroVax® studies [Harrop et al. 2010].
5T4 superantigen–antibody fusion protein (naptumomab estafenatox)
The tumor-targeted super-antigen concept employs bacterial superantigens, which are the most potent known activators of T cells, to attract and activate large numbers of T cells to the target [Eisen et al. 2014]. A phase II study of the superantigen-antibody fusion protein targeting 5T4 in RCC showed a significant survival benefit [Shaw et al. 2007], which led to the development of ANYARA (naptumomab estafenatox) [Eisen et al. 2014]. Naptumomab estafenatox has been evaluated in combination with interferon α in a phase II/III clinical trial involving patients with advanced RCC. Although the study failed to meet its primary endpoint, survival benefits were seen in a subgroup of patients with low levels of IL-6 and normal levels of SEA antibodies [Elkord et al. 2015]. The results of this study are still undergoing analysis, and a phase II/III clinical trial of naptumomab estafenatox in combination with a TKI for metastatic RCC is being designed [Stern et al. 2014]. Lastly, promising preclinical activity has been seen with an ADC targeting 5T4 [Sapra et al. 2013], and clinical evaluation is now underway [Stern et al. 2014].
PSA
PSA is a well-known prostate cancer biomarker that is now a target for antigen-specific immunotherapy. An anticancer vaccine known as PROSTVAC®-VF (PSA-TRICOM; rilimogene galvacirepvec) has been developed that incorporates three co-stimulatory molecules (CD80, CD54 and CD58, collectively referred to as TRICOM) and two recombinant viral vectors encoding PSA transgenes [Singh et al. 2015]. Patients are primed with the vaccinia-based vector and then boosted with a fowlpox vector, both of which are administered with GM-CSF. Early phase I and phase II clinical trials in patients with metastatic castration-resistant prostate cancer (mCRPC) demonstrated that PROSTVAC®-VF is safe, induces a high rate of immune responses and has survival benefits [DiPaola et al. 2006; Arlen et al. 2007; Gulley et al. 2010]. A double-blind, randomized, controlled phase II clinical trial in patients with mCRPC showed that treatment with PROSTVAC®-VF significantly improved OS by 8.5 months compared to treatment with control vectors [Kantoff et al. 2010b]. A phase III clinical trial [ClinicalTrials.gov identifier: NCT01322490] in 1200 patients with asymptomatic or minimally symptomatic mCRPC is currently ongoing [Singh et al. 2015].
Survivin
Also known as BIRC5 (baculoviral inhibitor of apoptosis protein repeat-containing 5), survivin is an inhibitor of the intrinsic apoptosis pathway that is widely overexpressed in cancers, making it an attractive target for antigen-specific immunotherapy [Mobahat et al. 2014]. Peptide cancer vaccines targeting survivin have been evaluated in phase I and phase II clinical trials. A phase I study of a multi-epitope anti-survivin peptide vaccine (EMD640744) in patients with advanced solid tumors found an antigen-specific T-cell response in 63% of subjects, and 28% of vaccinated subjects achieved stable disease [Lennerz et al. 2014]. Another phase I study in pediatric brainstem glioma patients using a tripeptide vaccine containing survivin and given in combination with the immunoadjuvant poly-ICLC (polyinosinic-polycytidylic acid stabilized by lysine and carboxymethylcellulose) showed evidence of survivin-specific immune responses and clinical benefit [Pollack et al. 2014]. In grade 2 low-grade glioma patients, vaccination with survivin and three other glioma-associated peptides combined with poly-ICLC produced robust TH1 immune responses [Okada et al. 2015]. The efficacy of another survivin-targeted peptide vaccine in metastatic melanoma patients was evaluated in a phase II clinical trial. Treatment with the vaccine, which contained three survivin peptide epitopes, significantly prolonged OS in patients with antigen-specific T-cell responses [Becker et al. 2012].
IDO1
Upregulation of indolamine-2,3-dioxygenase (IDO1), which catabolizes the essential amino acid tryptophan, is a recently discovered mechanism of tumor cell immune escape. Tryptophan is required for normal T-cell function, and its depletion can lead to immunosuppression [Jiang et al. 2015]. The IDO1 enzyme is overexpressed in various malignancies including breast, NSCLC, prostate and gastric cancers [Uyttenhove et al. 2003]. Two small molecule IDO1 enzyme inhibitors and a peptide-based vaccine are currently being evaluated in clinical trials. Indoximod (1-methly-D-tryptophan) combined with docetaxel was evaluated in a phase I clinical trial in patients with metastatic solid tumors. The treatment was well tolerated with evidence of clinical activity [Soliman et al. 2014]. A phase II trial of this agent in combination with docetaxel as first-line treatment for metastatic breast cancer is ongoing [ClinicalTrials.gov identifier: NCT01792050], in addition to a number of other phase II studies in metastatic melanoma, prostate and pancreatic cancers.
The other small molecule IDO1 inhibitor, INCB024360, is currently being evaluated in combination with ipilimumab in a phase I/II study [ClinicalTrials.gov identifier: NCT02077114 (completed)] in metastatic melanoma [Iversen et al. 2015], a phase I/II study [ClinicalTrials.gov identifier: NCT02318277] of INCB024360 in combination with durvalumab in patients with advanced solid tumors [Ibrahim et al. 2015] and a phase Ib study [ClinicalTrials.gov identifier: NCT02298153] in stage IIIB/IV NSCLC in combination with atezolizumab [Cha et al. 2015]. A peptide vaccine targeting IDO1 was evaluated in a phase I clinical trial in patients with metastatic NSCLC. Median OS was approximately 26 months, and long-lasting disease stabilization was observed in 47% of patients. A currently ongoing phase II study in patients with metastatic melanoma is evaluating the effects of a peptide vaccine containing epitopes to survivin and IDO1 in combination with temozolomide [Iversen et al. 2014].
Discussion
With the recent clinical development and therapeutic successes of immune checkpoint inhibitors targeting the CTLA-4 and PD-1/PD-L1 pathways, such as ipilimumab, pembrolizumab and nivolumab, enthusiasm surrounding anticancer immunotherapy has become intense. However, despite the impressive improvements in clinical outcome seen with these agents, the response rates remain modest, but encouraging. Clearly, we must expand our knowledge about how these agents work and the safest way to administer them in order to improve their efficacy. With respect to antigen-specific immunotherapies, the abundance of tumor antigens that have been identified has allowed for the development of multiple monoclonal antibodies and peptide cancer vaccines targeting these antigens, but the responses to these agents as well remain modest, due in part to mechanisms of tumor evasion and immunosuppression, and the fact that TAA expression profiles are patient-specific. With the availability of checkpoint inhibitors, it is now possible to alter the immunosuppressive tumor microenvironment, which could potentially increase the efficacy of antigen-specific immunotherapies. It is thus becoming quite clear that in order to derive the maximum benefit from these novel immunotherapies, we must learn more about how to best use these agents in combination with chemotherapy and with each other. Numerous clinical trials investigating various combinations of immunotherapies and chemotherapy are already underway.
Another point to consider with the use of cancer vaccines is that while they are not often associated with clinically significant objective responses or improvements in PFS, they have been shown to significantly prolong OS, and thus OS may be the most valid clinical endpoint for assessment of the efficacy of these agents [DeGregorio et al. 2012; Dillman, 2015]. Ultimately, personalized immunotherapy, whereby patients are screened for the expression of tumor antigens and PD-1/PD-L1 so that they can be matched with the appropriate antigen-specific agent, checkpoint inhibitor and chemotherapeutic agent, may be the best means of deriving the maximum therapeutic benefits from these treatments [Kao et al. 2015]. The future of personalized cancer immunotherapy has never been more promising, and the results of ongoing clinical trials are eagerly awaited.
Footnotes
Funding: This research received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors.
Conflict of interest statement: The author(s) declare(s) that there is no conflict of interest.
Contributor Information
Gregory T. Wurz, Department of Internal Medicine, Division of Hematology and Oncology, University of California, Davis, Sacramento, CA, USA
Chiao-Jung Kao, Department of Obstetrics and Gynecology, University of California, Davis Sacramento, CA, USA.
Michael W. DeGregorio, Department of Internal Medicine, Division of Hematology and Oncology, University of California, Davis, 4501 X Street Suite 3016, Sacramento, CA 95817, USA.
References
- Advani A., McDonough S., Coutre S., Wood B., Radich J., Mims M., et al. (2014) SWOG S0910: a phase 2 trial of clofarabine/cytarabine/epratuzumab for relapsed/refractory acute lymphocytic leukaemia. Br J Haematol 165: 504–509. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Agarwal S., Saini S., Parashar D., Verma A., Sinha A., Jagadish N., et al. (2013) The novel cancer-testis antigen A-kinase anchor protein 4 (AKAP4) is a potential target for immunotherapy of ovarian serous carcinoma. Oncoimmunology 2: e24270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ai J., Advani A. (2015) Current status of antibody therapy in ALL. Br J Haematol 168: 471–480. [DOI] [PubMed] [Google Scholar]
- Akiyama Y., Komiyama M., Miyata H., Yagoto M., Ashizawa T., Iizuka A., et al. (2014) Novel cancer-testis antigen expression on glioma cell lines derived from high-grade glioma patients. Oncol Rep 31: 1683–1690. [DOI] [PubMed] [Google Scholar]
- Alfonso M., Diaz A., Hernandez A., Perez A., Rodriguez E., Bitton R., et al. (2002) An anti-idiotype vaccine elicits a specific response to N-glycolyl sialic acid residues of glycoconjugates in melanoma patients. J Immunol 168: 2523–2529. [DOI] [PubMed] [Google Scholar]
- Alfonso S., Valdes-Zayas A., Santiesteban E., Flores Y., Areces F., Hernandez M., et al. (2014) A randomized, multicenter, placebo-controlled clinical trial of racotumomab-alum vaccine as switch maintenance therapy in advanced non-small cell lung cancer patients. Clin Cancer Res 20: 3660–3671. [DOI] [PubMed] [Google Scholar]
- Almeida L., Sakabe N., deOliveira A., Silva M., Mundstein A., Cohen T., et al. (2009) CTdatabase: a knowledge-base of high-throughput and curated data on cancer-testis antigens. Nucleic Acids Res 37: D816–D819. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Amato R., Hawkins R., Kaufman H., Thompson J., Tomczak P., Szczylik C., et al. (2010) Vaccination of metastatic renal cancer patients with MVA-5T4: a randomized, double-blind, placebo-controlled phase III study. Clin Cancer Res 16: 5539–5547. [DOI] [PubMed] [Google Scholar]
- Anderson A. (2014) TIM-3: an emerging target in the cancer immunotherapy landscape. Cancer Immunol Res 2: 393–398. [DOI] [PubMed] [Google Scholar]
- Anderson M., Venanzi E., Klein L., Chen Z., Berzins S., Turley S., et al. (2002) Projection of an immunological self shadow within the thymus by the Aire protein. Science 298: 1395–1401. [DOI] [PubMed] [Google Scholar]
- Annesley C., Brown P. (2015) Novel agents for the treatment of childhood acute leukemia. Ther Adv Hematol 6: 61–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Apostolopoulos V., McKenzie I. (1994) Cellular mucins: targets for immunotherapy. Crit Rev Immunol 14: 293–309. [DOI] [PubMed] [Google Scholar]
- Arlen P., Skarupa L., Pazdur M., Seetharam M., Tsang K., Grosenbach D., et al. (2007) Clinical safety of a viral vector based prostate cancer vaccine strategy. J Urol 178: 1515–1520. [DOI] [PubMed] [Google Scholar]
- Armand P., Nagler A., Weller E., Devine S., Avigan D., Chen Y., et al. (2013) Disabling immune tolerance by programmed death-1 blockade with pidilizumab after autologous hematopoietic stem-cell transplantation for diffuse large B-cell lymphoma: results of an international phase II trial. J Clin Oncol 31: 4199–4206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Atkins M., Kudchadkar R., Sznol M., McDermott D., Lotem M., Schachter J., et al. (2014) Phase 2, multicenter, safety and efficacy study of pidilizumab in patients with metastatic melanoma. J Clin Oncol 32(Suppl. 5): abstract 9001. [Google Scholar]
- Bagarazzi M., Yan J., Morrow M., Shen X., Parker R., Lee J., et al. (2012) Immunotherapy against HPV16/18 generates potent TH1 and cytotoxic cellular immune responses. Sci Transl Med 4: 155ra138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bang Y., Van Cutsem E., Feyereislova A., Chung H., Shen L., Sawaki A., et al. (2010) Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): a phase 3, open-label, randomised controlled trial. Lancet 376: 687–697. [DOI] [PubMed] [Google Scholar]
- Beauchemin N., Arabzadeh A. (2013) Carcinoembryonic antigen-related cell adhesion molecules (CEACAMs) in cancer progression and metastasis. Cancer Metastasis Rev 32: 643–671. [DOI] [PubMed] [Google Scholar]
- Becker J., Andersen M., Hofmeister-Muller V., Wobser M., Frey L., Sandig C., et al. (2012) Survivin-specific T-cell reactivity correlates with tumor response and patient survival: a phase-II peptide vaccination trial in metastatic melanoma. Cancer Immunol Immunother 61: 2091–2103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Berger C., Sommermeyer D., Hudecek M., Berger M., Balakrishnan A., Paszkiewicz P., et al. (2015) Safety of targeting ROR1 in primates with chimeric antigen receptor-modified T cells. Cancer Immunol Res 3: 206–216. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bokemeyer C., Bondarenko I., Makhson A., Hartmann J., Aparicio J., de Braud F., 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. [DOI] [PubMed] [Google Scholar]
- Bokemeyer C., Van Cutsem E., Rougier P., Ciardiello F., Heeger S., Schlichting M., et al. (2012) Addition of cetuximab to chemotherapy as first-line treatment for KRAS wild-type metastatic colorectal cancer: pooled analysis of the CRYSTAL and OPUS randomised clinical trials. Eur J Cancer 48: 1466–1475. [DOI] [PubMed] [Google Scholar]
- Bonner J., Harari P., Giralt J., Azarnia N., Shin D., Cohen R., et al. (2006) Radiotherapy plus cetuximab for squamous-cell carcinoma of the head and neck. N Engl J Med 354: 567–578. [DOI] [PubMed] [Google Scholar]
- Boyerinas B., Jochems C., Fantini M., Heery C., Gulley J., Tsang K., et al. (2015) Antibody-dependent cellular cytotoxicity activity of a novel anti-PD-L1 antibody avelumab (MSB0010718C) on human tumor cells. Cancer Immunol Res 3: 1148–1157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brahmer J., Reckamp K., Baas P., Crino L., Eberhardt W., Poddubskaya E., et al. (2015) Nivolumab versus docetaxel in advanced squamous-cell non-small-cell lung cancer. N Engl J Med 373: 123–135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brahmer J., Tykodi S., Chow L., Hwu W., Topalian S., Hwu P., et al. (2012) Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med 366: 2455–2465. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brayman M., Thathiah A., Carson D. (2004) MUC1: a multifunctional cell surface component of reproductive tissue epithelia. Reprod Biol Endocrinol 2: 4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brignone C., Gutierrez M., Mefti F., Brain E., Jarcau R., Cvitkovic F., et al. (2010) First-line chemoimmunotherapy in metastatic breast carcinoma: combination of paclitaxel and IMP321 (LAG-3Ig) enhances immune responses and antitumor activity. J Transl Med 8: 71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brun J., Dalstein V., Leveque J., Mathevet P., Raulic P., Baldauf J., et al. (2011) Regression of high-grade cervical intraepithelial neoplasia with TG4001 targeted immunotherapy. Am J Obstet Gynecol 204: 169.e1–169.e8. [DOI] [PubMed] [Google Scholar]
- Butts C., Socinski M., Mitchell P., Thatcher N., Havel L., Krzakowski M., et al. (2014) Tecemotide (L-BLP25) versus placebo after chemoradiotherapy for stage III non-small-cell lung cancer (START): a randomised, double-blind, phase 3 trial. Lancet Oncol 15: 59–68. [DOI] [PubMed] [Google Scholar]
- Buzzonetti A., Fossati M., Catzola V., Scambia G., Fattorossi A., Battaglia A. (2014) Immunological response induced by abagovomab as a maintenance therapy in patients with epithelial ovarian cancer: relationship with survival - a substudy of the MIMOSA trial. Cancer Immunol Immunother 63: 1037–1045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Calabro L., Ceresoli G., di Pietro A., Cutaia O., Morra A., Ibrahim R., et al. (2015) CTLA4 blockade in mesothelioma: finally a competing strategy over cytotoxic/target therapy? Cancer Immunol Immunother 64: 105–112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Calabro L., Morra A., Fonsatti E., Cutaia O., Amato G., Giannarelli D., et al. (2013) Tremelimumab for patients with chemotherapy-resistant advanced malignant mesothelioma: an open-label, single-arm, phase 2 trial. Lancet Oncol 14: 1104–1111. [DOI] [PubMed] [Google Scholar]
- Camisaschi C., De Filippo A., Beretta V., Vergani B., Villa A., Vergani E., et al. (2014) Alternative activation of human plasmacytoid DCs in vitro and in melanoma lesions: involvement of LAG-3. J Invest Dermatol 134: 1893–1902. [DOI] [PubMed] [Google Scholar]
- Cha E., Wallin J., Kowanetz M. (2015) PD-L1 inhibition with MPDL3280A for solid tumors. Semin Oncol 42: 484–487. [DOI] [PubMed] [Google Scholar]
- Cheema Z., Hari-Gupta Y., Kita G., Farrar D., Seddon I., Corr J., et al. (2014) Expression of the cancer-testis antigen BORIS correlates with prostate cancer. Prostate 74: 164–176. [DOI] [PubMed] [Google Scholar]
- Chen F., Lu Z., Deng J., Han X., Bai J., Liu Q., et al. (2014) SPAG9 expression is increased in human prostate cancer and promotes cell motility, invasion and angiogenesis in vitro. Oncol Rep 32: 2533–2540. [DOI] [PubMed] [Google Scholar]
- Chen J., Dawoodji A., Tarlton A., Gnjatic S., Tajar A., Karydis I., et al. (2015) NY-ESO-1 specific antibody and cellular responses in melanoma patients primed with NY-ESO-1 protein in ISCOMATRIX and boosted with recombinant NY-ESO-1 fowlpox virus. Int J Cancer 136: E590–E601. [DOI] [PubMed] [Google Scholar]
- Chen R., Chen B. (2015) Brentuximab vedotin for relapsed or refractory Hodgkin’s lymphoma. Drug Des Devel Ther 9: 1729–1733. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen Y., Scanlan M., Sahin U., Tureci O., Gure A., Tsang S., et al. (1997) A testicular antigen aberrantly expressed in human cancers detected by autologous antibody screening. Proc Natl Acad Sci U S A 94: 1914–1918. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cheng J., Kananathan R. (2012) CIMAvax EGF vaccine for stage IIIb/IV non-small cell lung carcinoma. Hum Vaccin Immunother 8: 1799–1801. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cheng L., Ruan Z. (2015) Tim-3 and Tim-4 as the potential targets for antitumor therapy. Hum Vaccin Immunother 11: 2458–2462. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chung C. (2015) Tyrosine kinase inhibitors for epidermal growth factor receptor gene mutation-positive non-small cell lung cancers: an update for recent advances in therapeutics. J Oncol Pharm Pract. DOI: 10.1177/1078155215577810. [DOI] [PubMed] [Google Scholar]
- Coosemans A., Vergote I., Van Gool S. (2014) Wilms’ tumor gene 1 immunotherapy in pelvic gynecological malignancies. Expert Rev Clin Immunol 10: 705–711. [DOI] [PubMed] [Google Scholar]
- Creelan B. (2014) Update on immune checkpoint inhibitors in lung cancer. Cancer Control 21: 80–89. [DOI] [PubMed] [Google Scholar]
- Cunningham D., Humblet Y., Siena S., Khayat D., Bleiberg H., Santoro A., et al. (2004) Cetuximab monotherapy and cetuximab plus irinotecan in irinotecan-refractory metastatic colorectal cancer. N Engl J Med 351: 337–345. [DOI] [PubMed] [Google Scholar]
- Curigliano G., Criscitiello C., Esposito A., Fumagalli L., Gelao L., Locatelli M., et al. (2013) Developing an effective breast cancer vaccine: challenges to achieving sterile immunity versus resetting equilibrium. Breast 22(Suppl. 2): S96–S99. [DOI] [PubMed] [Google Scholar]
- De Pas T., Giovannini M., Rescigno M., Catania C., Toffalorio F., Spitaleri G., et al. (2012) Vaccines in non-small cell lung cancer: rationale, combination strategies and update on clinical trials. Crit Rev Oncol Hematol 83: 432–443. [DOI] [PubMed] [Google Scholar]
- DeGregorio M., Soe L., Wolf M. (2014) Tecemotide (L-BLP25) versus placebo after chemoradiotherapy for stage III non-small cell lung cancer (START): a randomized, double-blind, phase III trial. J Thorac Dis 6: 571–573. [DOI] [PMC free article] [PubMed] [Google Scholar]
- DeGregorio M., Wurz G., Gutierrez A., Wolf M. (2012) L-BLP25 vaccine plus letrozole for breast cancer: Is translation possible? OncoImmunology 1: 1422–1424. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Derbinski J., Gabler J., Brors B., Tierling S., Jonnakuty S., Hergenhahn M., et al. (2005) Promiscuous gene expression in thymic epithelial cells is regulated at multiple levels. J Exp Med 202: 33–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Di Stasi A., Jimenez A., Minagawa K., Al-Obaidi M., Rezvani K. (2015) Review of the results of WT1 peptide vaccination strategies for myelodysplastic syndromes and acute myeloid leukemia from nine different studies. Front Immunol 6: 36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Diaz A., Alfonso M., Alonso R., Saurez G., Troche M., Catala M., et al. (2003) Immune responses in breast cancer patients immunized with an anti-idiotype antibody mimicking NeuGc-containing gangliosides. Clin Immunol 107: 80–89. [DOI] [PubMed] [Google Scholar]
- Dillman R. (2015) Cancer vaccines: can they improve survival? Cancer Biother Radiopharm 30: 147–151. [DOI] [PubMed] [Google Scholar]
- DiPaola R., Plante M., Kaufman H., Petrylak D., Israeli R., Lattime E., et al. (2006) A phase I trial of pox PSA vaccines (PROSTVAC-VF) with B7-1, ICAM-1, and LFA-3 co-stimulatory molecules (TRICOM) in patients with prostate cancer. J Transl Med 4: 1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Douillard J., Siena S., Cassidy J., Tabernero J., Burkes R., Barugel M., et al. (2010) Randomized, phase III trial of panitumumab with infusional fluorouracil, leucovorin, and oxaliplatin (FOLFOX4) versus FOLFOX4 alone as first-line treatment in patients with previously untreated metastatic colorectal cancer: the PRIME study. J Clin Oncol 28: 4697–4705. [DOI] [PubMed] [Google Scholar]
- Eisen T., Hedlund G., Forsberg G., Hawkins R. (2014) Naptumomab estafenatox: targeted immunotherapy with a novel immunotoxin. Curr Oncol Rep 16: 370. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Elkord E., Burt D., Sundstedt A., Nordle O., Hedlund G., Hawkins R. (2015) Immunological response and overall survival in a subset of advanced renal cell carcinoma patients from a randomized phase 2/3 study of naptumomab estafenatox plus IFN-alpha versus IFN-alpha. Oncotarget 6: 4428–4439. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Emens L., Braiteh F., Cassier P., Delord J., Eder J., Fasso M., et al. (2015) Inhibition of PD-L1 by MPDL3280A leads to clinical activity in patients with metastatic triple-negative breast cancer (TNBC) (Abstract 2859). In: Annual Meeting of the American Association for Cancer Research, Philadelphia, PA, 18–22 April. [Google Scholar]
- Endo M., de Graaff M., Ingram D., Lim S., Lev D., Briaire-de Bruijn I., et al. (2015) NY-ESO-1 (CTAG1B) expression in mesenchymal tumors. Mod Pathol 28: 587–595. [DOI] [PubMed] [Google Scholar]
- Escudier B., Bellmunt J., Negrier S., Bajetta E., Melichar B., Bracarda S., et al. (2010) Phase III trial of bevacizumab plus interferon alfa-2a in patients with metastatic renal cell carcinoma (AVOREN): final analysis of overall survival. J Clin Oncol 28: 2144–2150. [DOI] [PubMed] [Google Scholar]
- Escudier B., Pluzanska A., Koralewski P., Ravaud A., Bracarda S., Szczylik C., et al. (2007) Bevacizumab plus interferon alfa-2a for treatment of metastatic renal cell carcinoma: a randomised, double-blind phase III trial. Lancet 370: 2103–2111. [DOI] [PubMed] [Google Scholar]
- Felder M., Kapur A., Gonzalez-Bosquet J., Horibata S., Heintz J., Albrecht R., et al. (2014) MUC16 (CA125): tumor biomarker to cancer therapy, a work in progress. Mol Cancer 13: 129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ferrara N., Hillan K., Gerber H., Novotny W. (2004) Discovery and development of bevacizumab, an anti-VEGF antibody for treating cancer. Nat Rev Drug Discov 3: 391–400. [DOI] [PubMed] [Google Scholar]
- Foley E. (1953) Antigenic properties of methylcholanthrene-induced tumors in mice of the strain of origin. Cancer Res 13: 835–837. [PubMed] [Google Scholar]
- Fourcade J., Sun Z., Pagliano O., Chauvin J., Sander C., Janjic B., et al. (2014) PD-1 and TIM-3 regulate the expansion of tumor antigen-specific CD8+ T cells induced by melanoma vaccines. Cancer Res 74: 1045–1055. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Friedman H., Prados M., Wen P., Mikkelsen T., Schiff D., Abrey L., et al. (2009) Bevacizumab alone and in combination with irinotecan in recurrent glioblastoma. J Clin Oncol 27: 4733–4740. [DOI] [PubMed] [Google Scholar]
- Fuchs C., Tomasek J., Yong C., Dumitru F., Passalacqua R., Goswami C., 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. [DOI] [PubMed] [Google Scholar]
- Galluzzi L., Vacchelli E., Bravo-San Pedro J., Buque A., Senovilla L., Baracco E., et al. (2014) Classification of current anticancer immunotherapies. Oncotarget 5: 12472–12508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gao X., Zhu Y., Li G., Huang H., Zhang G., Wang F., et al. (2012) TIM-3 expression characterizes regulatory T cells in tumor tissues and is associated with lung cancer progression. PLoS One 7: e30676. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Garg M., Chaurasiya D., Rana R., Jagadish N., Kanojia D., Dudha N., et al. (2007) Sperm-associated antigen 9, a novel cancer testis antigen, is a potential target for immunotherapy in epithelial ovarian cancer. Clin Cancer Res 13: 1421–1428. [DOI] [PubMed] [Google Scholar]
- Garon E., Ciuleanu T., Arrieta O., Prabhash K., Syrigos K., Goksel T., 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. [DOI] [PubMed] [Google Scholar]
- Garon E., Rizvi N., Hui R., Leighl N., Balmanoukian A., Eder J., et al. (2015) Pembrolizumab for the treatment of non-small-cell lung cancer. N Engl J Med 372: 2018–2028. [DOI] [PubMed] [Google Scholar]
- Ghadban T., Perez D., Vashist Y., Bockhorn M., Koenig A., El Gammal A., et al. (2014) Expression of cancer testis antigens CT10 (MAGE-C2) and GAGE in gastrointestinal stromal tumors. Eur J Surg Oncol 40: 1307–1312. [DOI] [PubMed] [Google Scholar]
- Giantonio B., Catalano P., Meropol N., O’Dwyer P., Mitchell E., Alberts S., 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. [DOI] [PubMed] [Google Scholar]
- Goldberg M., Drake C. (2011) LAG-3 in Cancer Immunotherapy. Curr Top Microbiol Immunol 344: 269–278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gonzalez G., Crombet T., Neninger E., Viada C., Lage A. (2007) Therapeutic vaccination with epidermal growth factor (EGF) in advanced lung cancer: analysis of pooled data from three clinical trials. Hum Vaccin 3: 8–13. [DOI] [PubMed] [Google Scholar]
- Gotter J., Brors B., Hergenhahn M., Kyewski B. (2004) Medullary epithelial cells of the human thymus express a highly diverse selection of tissue-specific genes colocalized in chromosomal clusters. J Exp Med 199: 155–166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- GuhaThakurta D., Sheikh N., Fan L., Kandadi H., Meagher T., Hall S., et al. (2015) Humoral immune response against nontargeted tumor antigens after treatment with sipuleucel-T and its association with improved clinical outcome. Clin Cancer Res 21: 3619–3630. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gulley J., Arlen P., Madan R., Tsang K., Pazdur M., Skarupa L., et al. (2010) Immunologic and prognostic factors associated with overall survival employing a poxviral-based PSA vaccine in metastatic castrate-resistant prostate cancer. Cancer Immunol Immunother 59: 663–674. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gulley J., Spigel D., Kelly K., Chandler J., Rajan A., Hassan R., et al. (2015) Avelumab (MSB0010718C), an anti-PD-L1 antibody, in advanced NSCLC patients: A phase Ib, open-label expansion trial in patients progressing after platinum-based chemotherapy. J Clin Oncol 33(Suppl.): abstract 8034. [Google Scholar]
- Hamid O., Robert C., Daud A., Hodi F., Hwu W., Kefford R., et al. (2013) Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. N Engl J Med 369: 134–144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Harrop R., Chu F., Gabrail N., Srinivas S., Blount D., Ferrari A. (2013) Vaccination of castration-resistant prostate cancer patients with TroVax (MVA-5T4) in combination with docetaxel: a randomized phase II trial. Cancer Immunol Immunother 62: 1511–1520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Harrop R., Shingler W., Kelleher M., de Belin J., Treasure P. (2010) Cross-trial analysis of immunologic and clinical data resulting from phase I and II trials of MVA-5T4 (TroVax) in colorectal, renal, and prostate cancer patients. J Immunother 33: 999–1005. [DOI] [PubMed] [Google Scholar]
- Hay C., Sult E., Huang Q., Hammond S., Mulgrew K., McGlinchey K., et al. (2015) MEDI9447: Enhancing anti-tumor immunity by targeting CD73 in the tumor microenvironment. Cancer Res 75(Suppl. 15): abstract 285. [Google Scholar]
- He L., Ji J., Liu S., Xue E., Liang Q., Ma Z. (2014) Expression of cancer-testis antigen in multiple myeloma. J Huazhong Univ Sci Technolog Med Sci 34: 181–185. [DOI] [PubMed] [Google Scholar]
- Heery C., Ibrahim N., Arlen P., Mohebtash M., Murray J., Koenig K., et al. (2015) Docetaxel alone or in combination with a therapeutic cancer vaccine (PANVAC) in patients with metastatic breast cancer: A randomized clinical trial. JAMA Oncol. DOI: 10.1001/jamaoncol.2015.2736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Herbst R., Soria J., Kowanetz M., Fine G., Hamid O., Gordon M., et al. (2014) Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature 515: 563–567. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hodi F., Lawrence D., Lezcano C., Wu X., Zhou J., Sasada T., et al. (2014) Bevacizumab plus ipilimumab in patients with metastatic melanoma. Cancer Immunol Res 2: 632–642. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hodi F., O’Day S., McDermott D., Weber R., Sosman J., Haanen J., et al. (2010) Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med 363: 711–723. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hoffman L., Gore L. (2014) Blinatumomab, a bi-specific anti-CD19/CD3 BiTE((R)) antibody for the treatment of acute lymphoblastic leukemia: perspectives and current pediatric applications. Front Oncol 4: 63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hojjat-Farsangi M., Moshfegh A., Daneshmanesh A., Khan A., Mikaelsson E., Osterborg A., et al. (2014) The receptor tyrosine kinase ROR1–an oncofetal antigen for targeted cancer therapy. Semin Cancer Biol 29: 21–31. [DOI] [PubMed] [Google Scholar]
- Homet Moreno B., Parisi G., Robert L., Ribas A. (2015) Anti-PD-1 therapy in melanoma. Semin Oncol 42: 466–473. [DOI] [PubMed] [Google Scholar]
- Horn L., Spigel D., Gettinger S., Antonia S., Gordon M., Herbst R., et al. (2015) Clinical activity, safety and predictive biomarkers of the engineered antibody MPDL3280A (anti-PD-L1) in non-small cell lung cancer (NSCLC): update from a phase Ia study. J Clin Oncol 33(suppl.): abstract 8029. [Google Scholar]
- Huang C., Workman C., Flies D., Pan X., Marson A., Zhou G., et al. (2004) Role of LAG-3 in regulatory T cells. Immunity 21: 503–513. [DOI] [PubMed] [Google Scholar]
- Hunder N., Wallen H., Cao J., Hendricks D., Reilly J., Rodmyre R., et al. (2008) Treatment of metastatic melanoma with autologous CD4+ T cells against NY-ESO-1. N Engl J Med 358: 2698–2703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hurwitz H., Fehrenbacher L., Novotny W., Cartwright T., Hainsworth J., Heim W., et al. (2004) Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N Engl J Med 350: 2335–2342. [DOI] [PubMed] [Google Scholar]
- Hynes N., Lane H. (2005) ERBB receptors and cancer: the complexity of targeted inhibitors. Nat Rev Cancer 5: 341–354. [DOI] [PubMed] [Google Scholar]
- Ibrahim N., Murray J., Zhou D., Mittendorf E., Sample D., Tautchin M., et al. (2013) Survival advantage in patients with metastatic breast cancer receiving endocrine therapy plus sialyl Tn-KLH vaccine: post hoc analysis of a large randomized trial. J Cancer 4: 577–584. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ibrahim R., Stewart R., Shalabi A. (2015) PD-L1 blockade for cancer treatment: MEDI4736. Semin Oncol 42: 474–483. [DOI] [PubMed] [Google Scholar]
- Isfort S., Keller-v Amsberg G., Schafhausen P., Koschmieder S., Brummendorf T. (2014) Bosutinib: a novel second-generation tyrosine kinase inhibitor. Recent Results Cancer Res 201: 81–97. [DOI] [PubMed] [Google Scholar]
- Ishikawa H., Imano M., Shiraishi O., Yasuda A., Peng Y., Shinkai M., et al. (2014) Phase I clinical trial of vaccination with LY6K-derived peptide in patients with advanced gastric cancer. Gastric Cancer 17: 173–180. [DOI] [PubMed] [Google Scholar]
- Iversen T., Andersen M., Svane I. (2015) The targeting of indoleamine 2,3 dioxygenase -mediated immune escape in cancer. Basic Clin Pharmacol Toxicol 116: 19–24. [DOI] [PubMed] [Google Scholar]
- Iversen T., Engell-Noerregaard L., Ellebaek E., Andersen R., Larsen S., Bjoern J., et al. (2014) Long-lasting disease stabilization in the absence of toxicity in metastatic lung cancer patients vaccinated with an epitope derived from indoleamine 2,3 dioxygenase. Clin Cancer Res 20: 221–232. [DOI] [PubMed] [Google Scholar]
- Iwai Y., Ishida M., Tanaka Y., Okazaki T., Honjo T., Minato N. (2002) Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade. Proc Natl Acad Sci U S A 99: 12293–12297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jiang T., Sun Y., Yin Z., Feng S., Sun L., Li Z. (2015) Research progress of indoleamine 2,3-dioxygenase inhibitors. Future Med Chem 7: 185–201. [DOI] [PubMed] [Google Scholar]
- Jing W., Gershan J., Weber J., Tlomak D., McOlash L., Sabatos-Peyton C., et al. (2015) Combined immune checkpoint protein blockade and low dose whole body irradiation as immunotherapy for myeloma. J Immunother Cancer 3: 2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jonker D., O’Callaghan C., Karapetis C., Zalcberg J., Tu D., Au H., et al. (2007) Cetuximab for the treatment of colorectal cancer. N Engl J Med 357: 2040–2048. [DOI] [PubMed] [Google Scholar]
- Jungbluth A., Chen Y., Stockert E., Busam K., Kolb D., Iversen K., et al. (2001) Immunohistochemical analysis of NY-ESO-1 antigen expression in normal and malignant human tissues. Int J Cancer 92: 856–860. [DOI] [PubMed] [Google Scholar]
- Kanojia D., Garg M., Gupta S., Gupta A., Suri A. (2011) Sperm-associated antigen 9 is a novel biomarker for colorectal cancer and is involved in tumor growth and tumorigenicity. Am J Pathol 178: 1009–1020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kantoff P., Higano C., Shore N., Berger E., Small E., Penson D., et al. (2010a) Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med 363: 411–422. [DOI] [PubMed] [Google Scholar]
- Kantoff P., Schuetz T., Blumenstein B., Glode L., Bilhartz D., Wyand M., et al. (2010b) Overall survival analysis of a phase II randomized controlled trial of a Poxviral-based PSA-targeted immunotherapy in metastatic castration-resistant prostate cancer. J Clin Oncol 28: 1099–1105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kao C., Wurz G., Lin Y., Vang D., Griffey S., Wolf M., et al. (2015) Assessing the effects of concurrent versus sequential cisplatin/radiotherapy on immune status in lung tumor-bearing C57BL/6 mice. Cancer Immunol Res 3: 741–750. [DOI] [PubMed] [Google Scholar]
- Kao C., Wurz G., Monjazeb A., Vang D., Cadman T., Griffey S., et al. (2014) Antitumor effects of cisplatin combined with tecemotide immunotherapy in a human MUC1 transgenic lung cancer mouse model. Cancer Immunol Res 2: 581–589. [DOI] [PubMed] [Google Scholar]
- Kaplan D., Shankaran V., Dighe A., Stockert E., Aguet M., Old L., et al. (1998) Demonstration of an interferon gamma-dependent tumor surveillance system in immunocompetent mice. Proc Natl Acad Sci U S A 95: 7556–7561. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kaufman H., Hamid O., D’Angelo S., Yuan G., Chin K., Bhatia S., et al. (2015) A phase II, open-label, multicenter trial to investigate the clinical activity and safety of avelumab (MSB0010718C) in patients with metastatic Merkel cell carcinoma. J Clin Oncol 33(Suppl.): abstract TPS9086. [Google Scholar]
- Kawai T., Enomoto Y., Morikawa T., Matsushita H., Kume H., Fukayama M., et al. (2014) High expression of heat shock protein 105 predicts a favorable prognosis for patients with urinary bladder cancer treated with radical cystectomy. Mol Clin Oncol 2: 38–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kelly K., Patel M., Infante J., Iannotti N., Nikolinakos P., Leach J., et al. (2015) Avelumab (MSB0010718C), an anti-PD-L1 antibody, in patients with metastatic or locally advanced solid tumors: assessment of safety and tolerability in a phase I, open-label expansion study. J Clin Oncol 33(Suppl.): abstract 3044. [Google Scholar]
- Kenter G., Welters M., Valentijn A., Lowik M., Berends-van der Meer D., Vloon A., et al. (2009) Vaccination against HPV-16 oncoproteins for vulvar intraepithelial neoplasia. N Engl J Med 361: 1838–1847. [DOI] [PubMed] [Google Scholar]
- Kimura T., Finn O. (2013) MUC1 immunotherapy is here to stay. Expert Opin Biol Ther 13: 35–49. [DOI] [PubMed] [Google Scholar]
- Kirkwood J., Butterfield L., Tarhini A., Zarour H., Kalinski P., Ferrone S. (2012) Immunotherapy of cancer in 2012. CA Cancer J Clin 62: 309–335. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Klein G., Sjogren H., Klein E., Hellstrom K. (1960) Demonstration of resistance against methylcholanthrene-induced sarcomas in the primary autochthonous host. Cancer Res 20: 1561–1572. [PubMed] [Google Scholar]
- Klein O., Davis I., McArthur G., Chen L., Haydon A., Parente P., et al. (2015) Low-dose cyclophosphamide enhances antigen-specific CD4(+) T cell responses to NY-ESO-1/ISCOMATRIX vaccine in patients with advanced melanoma. Cancer Immunol Immunother 64: 507–518. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kohrt H., Houot R., Marabelle A., Cho H., Osman K., Goldstein M., et al. (2012) Combination strategies to enhance antitumor ADCC. Immunotherapy 4: 511–527. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kohrt H., Thielens A., Marabelle A., Sagiv-Barfi I., Sola C., Chanuc F., et al. (2014) Anti-KIR antibody enhancement of anti-lymphoma activity of natural killer cells as monotherapy and in combination with anti-CD20 antibodies. Blood 123: 678–686. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kruit W., Suciu S., Dreno B., Mortier L., Robert C., Chiarion-Sileni V., et al. (2013) Selection of immunostimulant AS15 for active immunization with MAGE-A3 protein: results of a randomized phase II study of the European Organisation for Research and Treatment of Cancer Melanoma Group in Metastatic Melanoma. J Clin Oncol 31: 2413–2420. [DOI] [PubMed] [Google Scholar]
- Kular R., Yehiely F., Deiss L. (2010) Rational drug design: a GAGE derived peptide kills tumor cells. Cancer Biol Ther 9: 825–831. [DOI] [PubMed] [Google Scholar]
- Larkin J., Chiarion-Sileni V., Gonzalez R., Grob J., Cowey C., Lao C., et al. (2015) Combined nivolumab and ipilimumab or monotherapy in untreated melanoma. N Engl J Med 373: 23–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Le Mercier I., Chen W., Lines J., Day M., Li J., Sergent P., et al. (2014) VISTA Regulates the development of protective antitumor immunity. Cancer Res 74: 1933–1944. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee H., Kim J., Cho S., Ko T., Kim H., Park S., et al. (2014) Expression of the brother of the regulator of imprinted sites gene in the sputum of patients with lung cancer. Korean J Thorac Cardiovasc Surg 47: 378–383. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee S., Chow L. (2014) A new addition to the PD-1 checkpoint inhibitors for non-small cell lung cancer-the anti-PDL1 antibody-MEDI4736. Transl Lung Cancer Res 3: 408–410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lennerz V., Gross S., Gallerani E., Sessa C., Mach N., Boehm S., et al. (2014) Immunologic response to the survivin-derived multi-epitope vaccine EMD640744 in patients with advanced solid tumors. Cancer Immunol Immunother 63: 381–394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lindauer M., Hochhaus A. (2014) Dasatinib. Recent Results Cancer Res 201: 27–65. [DOI] [PubMed] [Google Scholar]
- Lines J., Pantazi E., Mak J., Sempere L., Wang L., O’Connell S., et al. (2014a) VISTA is an immune checkpoint molecule for human T cells. Cancer Res 74: 1924–1932. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lines J., Sempere L., Broughton T., Wang L., Noelle R. (2014b) VISTA is a novel broad-spectrum negative checkpoint regulator for cancer immunotherapy. Cancer Immunol Res 2: 510–517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu J., Yuan Y., Chen W., Putra J., Suriawinata A., Schenk A., et al. (2015a) Immune-checkpoint proteins VISTA and PD-1 nonredundantly regulate murine T-cell responses. Proc Natl Acad Sci U S A 112: 6682–6687. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu S., Powderly J., Camidge D., Ready N., Heist R., Hodi S., et al. (2015b) Safety and efficacy of MPDL3280A (anti-PD-L1) in combination with platinum-based doublet chemotherapy in patients with advanced non-small cell lung cancer (NSCLC). J Clin Oncol 33(Suppl.): abstract 8030. [Google Scholar]
- Loke J., Khan J., Wilson J., Craddock C., Wheatley K. (2015) Mylotarg has potent anti-leukaemic effect: a systematic review and meta-analysis of anti-CD33 antibody treatment in acute myeloid leukaemia. Ann Hematol 94: 361–373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lu J., Lee-Gabel L., Nadeau M., Ferencz T., Soefje S. (2014) Clinical evaluation of compounds targeting PD-1/PD-L1 pathway for cancer immunotherapy. J Oncol Pharm Pract. DOI: 10.1177/1078155214538087. [DOI] [PubMed] [Google Scholar]
- Manzoni M., Rovati B., Ronzoni M., Loupakis F., Mariucci S., Ricci V., et al. (2010) Immunological effects of bevacizumab-based treatment in metastatic colorectal cancer. Oncology 79: 187–196. [DOI] [PubMed] [Google Scholar]
- Melero I., Gaudernack G., Gerritsen W., Huber C., Parmiani G., Scholl S., et al. (2014) Therapeutic vaccines for cancer: an overview of clinical trials. Nat Rev Clin Oncol 11: 509–524. [DOI] [PubMed] [Google Scholar]
- Miles D., Roche H., Martin M., Perren T., Cameron D., Glaspy J., et al. (2011) Phase III multicenter clinical trial of the sialyl-TN (STn)-keyhole limpet hemocyanin (KLH) vaccine for metastatic breast cancer. Oncologist 16: 1092–1100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Miller K., Wang M., Gralow J., Dickler M., Cobleigh M., Perez E., et al. (2007) Paclitaxel plus bevacizumab versus paclitaxel alone for metastatic breast cancer. N Engl J Med 357: 2666–2676. [DOI] [PubMed] [Google Scholar]
- Mirandola L., Figueroa J., Phan T., Grizzi F., Kim M., Rahman R., et al. (2015) Novel antigens in non-small cell lung cancer: SP17, AKAP4, and PTTG1 are potential immunotherapeutic targets. Oncotarget 6: 2812–2826. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mitchell P., Thatcher N., Socinski M., Wasilewska-Tesluk E., Horwood K., Szczesna A., et al. (2015) Tecemotide in unresectable stage III non-small-cell lung cancer in the phase III START study: updated overall survival and biomarker analyses. Ann Oncol 26: 1134–1142. [DOI] [PubMed] [Google Scholar]
- Miyazaki T., Ikeda K., Horie-Inoue K., Kondo T., Takahashi S., Inoue S. (2014) EBAG9 modulates host immune defense against tumor formation and metastasis by regulating cytotoxic activity of T lymphocytes. Oncogenesis 3: e126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mobahat M., Narendran A., Riabowol K. (2014) Survivin as a preferential target for cancer therapy. Int J Mol Sci 15: 2494–2516. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morgan R., Chinnasamy N., Abate-Daga D., Gros A., Robbins P., Zheng Z., et al. (2013) Cancer regression and neurological toxicity following anti-MAGE-A3 TCR gene therapy. J Immunother 36: 133–151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morse M., Chaudhry A., Gabitzsch E., Hobeika A., Osada T., Clay T., et al. (2013a) Novel adenoviral vector induces T-cell responses despite anti-adenoviral neutralizing antibodies in colorectal cancer patients. Cancer Immunol Immunother 62: 1293–1301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morse M., Niedzwiecki D., Marshall J., Garrett C., Chang D., Aklilu M., et al. (2013b) A randomized phase II study of immunization with dendritic cells modified with poxvectors encoding CEA and MUC1 compared with the same poxvectors plus GM-CSF for resected metastatic colorectal cancer. Ann Surg 258: 879–886. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Muhlbauer M., Fleck M., Schutz C., Weiss T., Froh M., Blank C., et al. (2006) PD-L1 is induced in hepatocytes by viral infection and by interferon-alpha and -gamma and mediates T cell apoptosis. J Hepatol 45: 520–528. [DOI] [PubMed] [Google Scholar]
- Mullard A. (2013) New checkpoint inhibitors ride the immunotherapy tsunami. Nat Rev Drug Discov 12: 489–492. [DOI] [PubMed] [Google Scholar]
- Nauts H., Fowler G., Bogatko F. (1953) A review of the influence of bacterial infection and of bacterial products (Coley’s toxins) on malignant tumors in man; a critical analysis of 30 inoperable cases treated by Coley’s mixed toxins, in which diagnosis was confirmed by microscopic examination selected for special study. Acta Med Scand Suppl 276: 1–103. [PubMed] [Google Scholar]
- Neninger Vinageras E., de la Torre A., Osorio Rodriguez M., Catala Ferrer M., Bravo I., Mendoza del Pino M., et al. (2008) Phase II randomized controlled trial of an epidermal growth factor vaccine in advanced non-small-cell lung cancer. J Clin Oncol 26: 1452–1458. [DOI] [PubMed] [Google Scholar]
- Ngiow S., von Scheidt B., Akiba H., Yagita H., Teng M., Smyth M. (2011) Anti-TIM3 antibody promotes T cell IFN-gamma-mediated antitumor immunity and suppresses established tumors. Cancer Res 71: 3540–3551. [DOI] [PubMed] [Google Scholar]
- Nguyen L., Ohashi P. (2015) Clinical blockade of PD1 and LAG3–potential mechanisms of action. Nat Rev Immunol 15: 45–56. [DOI] [PubMed] [Google Scholar]
- Nicholaou T., Ebert L., Davis I., Robson N., Klein O., Maraskovsky E., et al. (2006) Directions in the immune targeting of cancer: lessons learned from the cancer-testis Ag NY-ESO-1. Immunol Cell Biol 84: 303–317. [DOI] [PubMed] [Google Scholar]
- Nicholaou T., Ebert L., Davis I., McArthur G., Jackson H., Dimopoulos N., et al. (2009) Regulatory T-cell-mediated attenuation of T-cell responses to the NY-ESO-1 ISCOMATRIX vaccine in patients with advanced malignant melanoma. Clin Cancer Res 15: 2166–2173. [DOI] [PubMed] [Google Scholar]
- Nieto M., Cano A. (2012) The epithelial-mesenchymal transition under control: global programs to regulate epithelial plasticity. Semin Cancer Biol 22: 361–368. [DOI] [PubMed] [Google Scholar]
- O’Sullivan C., Connolly R. (2014) Pertuzumab and its accelerated approval: evolving treatment paradigms and new challenges in the management of HER2-positive breast cancer. Oncology (Williston Park) 28: 186–194, 196. [PubMed] [Google Scholar]
- Odunsi K., Matsuzaki J., Karbach J., Neumann A., Mhawech-Fauceglia P., Miller A., et al. (2012) Efficacy of vaccination with recombinant vaccinia and fowlpox vectors expressing NY-ESO-1 antigen in ovarian cancer and melanoma patients. Proc Natl Acad Sci U S A 109: 5797–5802. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ofuji K., Tada Y., Yoshikawa T., Shimomura M., Yoshimura M., Saito K., et al. (2015) A peptide antigen derived from EGFR T790M is immunogenic in nonsmall cell lung cancer. Int J Oncol 46: 497–504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Okabayashi K., Fujita T., Miyazaki J., Okada T., Iwata T., Hirao N., et al. (2012) Cancer-testis antigen BORIS is a novel prognostic marker for patients with esophageal cancer. Cancer Sci 103: 1617–1624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Okada H., Butterfield L., Hamilton R., Hoji A., Sakaki M., Ahn B., et al. (2015) Induction of robust type-I CD8+ T-cell responses in WHO grade 2 low-grade glioma patients receiving peptide-based vaccines in combination with poly-ICLC. Clin Cancer Res 21: 286–294. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ostendorf B., le Coutre P., Kim T., Quintas-Cardama A. (2014) Nilotinib. Recent Results Cancer Res 201: 67–80. [DOI] [PubMed] [Google Scholar]
- Pao W., Miller V., Politi K., Riely G., Somwar R., Zakowski M., et al. (2005) Acquired resistance of lung adenocarcinomas to gefitinib or erlotinib is associated with a second mutation in the EGFR kinase domain. PLoS Med 2: e73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Paz-Ares L., Mezger J., Ciuleanu T., Fischer J., von Pawel J., Provencio M., et al. (2015) Necitumumab plus pemetrexed and cisplatin as first-line therapy in patients with stage IV non-squamous non-small-cell lung cancer (INSPIRE): an open-label, randomised, controlled phase 3 study. Lancet Oncol 16: 328–337. [DOI] [PubMed] [Google Scholar]
- Perez E., Romond E., Suman V., Jeong J., Davidson N., Geyer C., Jr., et al. (2011) Four-year follow-up of trastuzumab plus adjuvant chemotherapy for operable human epidermal growth factor receptor 2-positive breast cancer: joint analysis of data from NCCTG N9831 and NSABP B-31. J Clin Oncol 29: 3366–3373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Perez E., Romond E., Suman V., Jeong J., Sledge G., Geyer C., Jr., et al. (2014) Trastuzumab plus adjuvant chemotherapy for human epidermal growth factor receptor 2-positive breast cancer: planned joint analysis of overall survival from NSABP B-31 and NCCTG N9831. J Clin Oncol 32: 3744–3752. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Petrylak D., Powles T., Bellmunt J., Braiteh F., Loriot Y., Zambrano C., et al. (2015) A phase Ia study of MPDL3280A (anti-PD-L1): updated response and survival data in urothelial bladder cancer (UBC). J Clin Oncol 33(Suppl.): abstract 4501. [Google Scholar]
- Pinna A. (2014) Adenosine A2A receptor antagonists in Parkinson’s disease: progress in clinical trials from the newly approved istradefylline to drugs in early development and those already discontinued. CNS Drugs 28: 455–474. [DOI] [PubMed] [Google Scholar]
- Planchard D., Shtivelband M., Shi K., Ibrahim R., Ballas M., Sorio J., et al. (2015) A phase III study of MEDI4736 (M), an anti-PD-L1 antibody, in monotherapy or in combination with tremelimumab (T), versus standard of care (SOC) in patients (pts) with advanced non-small cell lung cancer (NSCLC) who have received at least two prior systemic treatment regimens (ARCTIC). J Clin Oncol 33(Suppl.): abstract TPS8104. [Google Scholar]
- Pollack I., Jakacki R., Butterfield L., Hamilton R., Panigrahy A., Potter D., et al. (2014) Antigen-specific immune responses and clinical outcome after vaccination with glioma-associated antigen peptides and polyinosinic-polycytidylic acid stabilized by lysine and carboxymethylcellulose in children with newly diagnosed malignant brainstem and nonbrainstem gliomas. J Clin Oncol 32: 2050–2058. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Postow M., Chesney J., Pavlick A., Robert C., Grossmann K., McDermott D., et al. (2015) Nivolumab and ipilimumab versus ipilimumab in untreated melanoma. N Engl J Med 372: 2006–2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Powles T., Eder J., Fine G., Braiteh F., Loriot Y., Cruz C., et al. (2014) MPDL3280A (anti-PD-L1) treatment leads to clinical activity in metastatic bladder cancer. Nature 515: 558–562. [DOI] [PubMed] [Google Scholar]
- Prehn R., Main J. (1957) Immunity to methylcholanthrene-induced sarcomas. J Natl Cancer Inst 18: 769–778. [PubMed] [Google Scholar]
- Price T., Peeters M., Kim T., Li J., Cascinu S., Ruff P., et al. (2014) Panitumumab versus cetuximab in patients with chemotherapy-refractory wild-type KRAS exon 2 metastatic colorectal cancer (ASPECCT): a randomised, multicentre, open-label, non-inferiority phase 3 study. Lancet Oncol 15: 569–579. [DOI] [PubMed] [Google Scholar]
- Pujade-Lauraine E., Hilpert F., Weber B., Reuss A., Poveda A., Kristensen G., et al. (2014) Bevacizumab combined with chemotherapy for platinum-resistant recurrent ovarian cancer: The AURELIA open-label randomized phase III trial. J Clin Oncol 32: 1302–1308. [DOI] [PubMed] [Google Scholar]
- Pujol J., Pirker R., Lynch T., Butts C., Rosell R., Shepherd F., et al. (2014) Meta-analysis of individual patient data from randomized trials of chemotherapy plus cetuximab as first-line treatment for advanced non-small cell lung cancer. Lung Cancer 83: 211–218. [DOI] [PubMed] [Google Scholar]
- Raetz E., Cairo M., Borowitz M., Blaney S., Krailo M., Leil T., et al. (2008) Chemoimmunotherapy reinduction with epratuzumab in children with acute lymphoblastic leukemia in marrow relapse: a Children’s Oncology Group Pilot Study. J Clin Oncol 26: 3756–3762. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Raetz E., Cairo M., Borowitz M., Lu X., Devidas M., Reid J., et al. (2015) Re-induction chemoimmunotherapy with epratuzumab in relapsed acute lymphoblastic leukemia (ALL): phase II results from Children’s Oncology Group (COG) study ADVL04P2. Pediatr Blood Cancer 62: 1171–1175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reardon D., Dietrich J., Kaley T., Gan H., Dunn G., Cloughesy T., et al. (2015) Phase II study to evaluate the clinical efficacy and safety of MEDI4736 in patients with glioblastoma (GBM). J Clin Oncol 33(Suppl.): abstract TPS2077. [Google Scholar]
- Reinhard H., Yousef S., Luetkens T., Fehse B., Berdien B., Kroger N., et al. (2014) Cancer-testis antigen MAGE-C2/CT10 induces spontaneous CD4+ and CD8+ T-cell responses in multiple myeloma patients. Blood Cancer J 4: e212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ribas A., Kefford R., Marshall M., Punt C., Haanen J., Marmol M., et al. (2013) Phase III randomized clinical trial comparing tremelimumab with standard-of-care chemotherapy in patients with advanced melanoma. J Clin Oncol 31: 616–622. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Robbins P., Morgan R., Feldman S., Yang J., Sherry R., Dudley M., et al. (2011) Tumor regression in patients with metastatic synovial cell sarcoma and melanoma using genetically engineered lymphocytes reactive with NY-ESO-1. J Clin Oncol 29: 917–924. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Robert C., Long G., Brady B., Dutriaux C., Maio M., Mortier L., et al. (2015a) Nivolumab in previously untreated melanoma without BRAF mutation. N Engl J Med 372: 320–330. [DOI] [PubMed] [Google Scholar]
- Robert C., Ribas A., Wolchok J., Hodi F., Hamid O., Kefford R., et al. (2014) Anti-programmed-death-receptor-1 treatment with pembrolizumab in ipilimumab-refractory advanced melanoma: a randomised dose-comparison cohort of a phase 1 trial. Lancet 384: 1109–1117. [DOI] [PubMed] [Google Scholar]
- Robert C., Schachter J., Long G., Arance A., Grob J., Mortier L., et al. (2015b) Pembrolizumab versus ipilimumab in advanced melanoma. N Engl J Med 372: 2521–2532. [DOI] [PubMed] [Google Scholar]
- Robert C., Thomas L., Bondarenko I., O’Day S., Weber J., Garbe C., et al. (2011) Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. N Engl J Med 364: 2517–2526. [DOI] [PubMed] [Google Scholar]
- Romano E., Michielin O., Voelter V., Laurent J., Bichat H., Stravodimou A., et al. (2014) MART-1 peptide vaccination plus IMP321 (LAG-3Ig fusion protein) in patients receiving autologous PBMCs after lymphodepletion: results of a phase I trial. J Transl Med 12: 97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Romond E., Perez E., Bryant J., Suman V., Geyer C., Jr., Davidson N., et al. (2005) Trastuzumab plus adjuvant chemotherapy for operable HER2-positive breast cancer. N Engl J Med 353: 1673–1684. [DOI] [PubMed] [Google Scholar]
- Rosenberg S. (2001) Progress in human tumour immunology and immunotherapy. Nature 411: 380–384. [DOI] [PubMed] [Google Scholar]
- Rowe J., Cen P. (2014) TroVax in colorectal cancer. Hum Vaccin Immunother 10: 3196–3200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ruiz R., Hunis B., Raez L. (2014) Immunotherapeutic agents in non-small-cell lung cancer finally coming to the front lines. Curr Oncol Rep 16: 400. [DOI] [PubMed] [Google Scholar]
- Saadatmand S., de Kruijf E., Sajet A., Dekker-Ensink N., van Nes J., Putter H., et al. (2013) Expression of cell adhesion molecules and prognosis in breast cancer. Br J Surg 100: 252–260. [DOI] [PubMed] [Google Scholar]
- Sakuishi K., Apetoh L., Sullivan J., Blazar B., Kuchroo V., Anderson A. (2010) Targeting tim-3 and PD-1 pathways to reverse T cell exhaustion and restore anti-tumor immunity. J Exp Med 207: 2187–2194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Saltz L., Meropol N., Loehrer P., Needle M., Kopit J., Mayer R. (2004) Phase II trial of cetuximab in patients with refractory colorectal cancer that expresses the epidermal growth factor receptor. J Clin Oncol 22: 1201–1208. [DOI] [PubMed] [Google Scholar]
- Sandler A., Gray R., Perry M., Brahmer J., Schiller J., Dowlati A., et al. (2006) Paclitaxel-carboplatin alone or with bevacizumab for non-small-cell lung cancer. N Engl J Med 355: 2542–2550. [DOI] [PubMed] [Google Scholar]
- Sapra P., Damelin M., Dijoseph J., Marquette K., Geles K., Golas J., et al. (2013) Long-term tumor regression induced by an antibody–drug conjugate that targets 5T4, an oncofetal antigen expressed on tumor-initiating cells. Mol Cancer Ther 12: 38–47. [DOI] [PubMed] [Google Scholar]
- Savage P., Leventhal D., Malchow S. (2014) Shaping the repertoire of tumor-infiltrating effector and regulatory T cells. Immunol Rev 259: 245–258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sawada Y., Komori H., Tsunoda Y., Shimomura M., Takahashi M., Baba H., et al. (2014) Identification of HLA-A2 or HLA-A24-restricted CTL epitopes for potential HSP105-targeted immunotherapy in colorectal cancer. Oncol Rep 31: 1051–1058. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Scagliotti G., Novello S., Schiller J., Hirsh V., Sequist L., Soria J., et al. (2012) Rationale and design of MARQUEE: a phase III, randomized, double-blind study of tivantinib plus erlotinib versus placebo plus erlotinib in previously treated patients with locally advanced or metastatic, nonsquamous, non-small-cell lung cancer. Clin Lung Cancer 13: 391–395. [DOI] [PubMed] [Google Scholar]
- Schreiber R., Old L., Smyth M. (2011) Cancer immunoediting: integrating immunity’s roles in cancer suppression and promotion. Science 331: 1565–1570. [DOI] [PubMed] [Google Scholar]
- Schumacher T., Bunse L., Pusch S., Sahm F., Wiestler B., Quandt J., et al. (2014) A vaccine targeting mutant IDH1 induces antitumour immunity. Nature 512: 324–327. [DOI] [PubMed] [Google Scholar]
- Schwartzentruber D., Lawson D., Richards J., Conry R., Miller D., Treisman J., et al. (2011) gp100 peptide vaccine and interleukin-2 in patients with advanced melanoma. N Engl J Med 364: 2119–2127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Scott L. (2015) Nivolumab: a review in advanced melanoma. Drugs 75: 1413–1424. [DOI] [PubMed] [Google Scholar]
- Seimetz D. (2011) Novel monoclonal antibodies for cancer treatment: the trifunctional antibody catumaxomab (removab). J Cancer 2: 309–316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shabani M., Naseri J., Shokri F. (2015) Receptor tyrosine kinase-like orphan receptor 1: a novel target for cancer immunotherapy. Expert Opin Ther Targets 19: 941–955. [DOI] [PubMed] [Google Scholar]
- Shankaran V., Ikeda H., Bruce A., White J., Swanson P., Old L., et al. (2001) IFNgamma and lymphocytes prevent primary tumour development and shape tumour immunogenicity. Nature 410: 1107–1111. [DOI] [PubMed] [Google Scholar]
- Shaw D., Connolly N., Patel P., Kilany S., Hedlund G., Nordle O., et al. (2007) A phase II study of a 5T4 oncofoetal antigen tumour-targeted superantigen (ABR-214936) therapy in patients with advanced renal cell carcinoma. Br J Cancer 96: 567–574. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shore N. (2014) PROSTVAC(R) targeted immunotherapy candidate for prostate cancer. Immunotherapy 6: 235–247. [DOI] [PubMed] [Google Scholar]
- Sierro S., Romero P., Speiser D. (2011) The CD4-like molecule LAG-3, biology and therapeutic applications. Expert Opin Ther Targets 15: 91–101. [DOI] [PubMed] [Google Scholar]
- Singh P., Pal S., Alex A., Agarwal N. (2015) Development of PROSTVAC immunotherapy in prostate cancer. Future Oncol 11: 2137–2148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Skoetz N., Bauer K., Elter T., Monsef I., Roloff V., Hallek M., et al. (2012) Alemtuzumab for patients with chronic lymphocytic leukaemia. Cochrane Database Syst Rev 2: CD008078. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Slamon D., Clark G., Wong S., Levin W., Ullrich A., McGuire W. (1987) Human breast cancer: correlation of relapse and survival with amplification of the HER-2/neu oncogene. Science 235: 177–182. [DOI] [PubMed] [Google Scholar]
- Slamon D., Godolphin W., Jones L., Holt J., Wong S., Keith D., et al. (1989) Studies of the HER-2/neu proto-oncogene in human breast and ovarian cancer. Science 244: 707–712. [DOI] [PubMed] [Google Scholar]
- Slamon D., Leyland-Jones B., Shak S., Fuchs H., Paton V., Bajamonde A., 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. [DOI] [PubMed] [Google Scholar]
- Sliwkowski M., Mellman I. (2013) Antibody therapeutics in cancer. Science 341: 1192–1198. [DOI] [PubMed] [Google Scholar]
- Smith H., Cronk R., Lang J., McNeel D. (2011) Expression and immunotherapeutic targeting of the SSX family of cancer-testis antigens in prostate cancer. Cancer Res 71: 6785–6795. [DOI] [PubMed] [Google Scholar]
- Sobrero A., Maurel J., Fehrenbacher L., Scheithauer W., Abubakr Y., Lutz M., et al. (2008) EPIC: phase III trial of cetuximab plus irinotecan after fluoropyrimidine and oxaliplatin failure in patients with metastatic colorectal cancer. J Clin Oncol 26: 2311–2319. [DOI] [PubMed] [Google Scholar]
- Soliman H., Jackson E., Neuger T., Dees E., Harvey R., Han H., et al. (2014) A first in man phase I trial of the oral immunomodulator, indoximod, combined with docetaxel in patients with metastatic solid tumors. Oncotarget 5: 8136–8146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Southall P., Boxer G., Bagshawe K., Hole N., Bromley M., Stern P. (1990) Immunohistological distribution of 5T4 antigen in normal and malignant tissues. Br J Cancer 61: 89–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Spigel D., Chaft J., Gettinger S., Chao B., Dirix L., Schmid P., et al. (2015) Clinical activity and safety from a phase II study (FIR) of MPDL3280A (anti-PD-L1) in PD-L1-selected patients with non-small cell lung cancer (NSCLC). J Clin Oncol 33(Suppl.): abstract 8028. [Google Scholar]
- Spigel D., Ervin T., Ramlau R., Daniel D., Goldschmidt J., Jr., Blumenschein G., Jr., et al. (2013) Randomized phase II trial of Onartuzumab in combination with erlotinib in patients with advanced non-small-cell lung cancer. J Clin Oncol 31: 4105–4114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Spira A., Park K., Mazieres J., Vansteenkiste J., Rittmeyer A., Ballinger M., et al. (2015) Efficacy, safety and predictive biomarker results from a randomized phase II study comparing MPDL3280A versus docetaxel in 2L/3L NSCLC (POPLAR). J Clin Oncol 33(Suppl.): abstract 8010. [Google Scholar]
- Staff C., Mozaffari F., Frodin J., Mellstedt H., Liljefors M. (2014) Telomerase (GV1001) vaccination together with gemcitabine in advanced pancreatic cancer patients. Int J Oncol 45: 1293–1303. [DOI] [PubMed] [Google Scholar]
- Stagg J., Loi S., Divisekera U., Ngiow S., Duret H., Yagita H., et al. (2011) Anti-ErbB-2 mAb therapy requires type I and II interferons and synergizes with anti-PD-1 or anti-CD137 mAb therapy. Proc Natl Acad Sci U S A 108: 7142–7147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stern P., Brazzatti J., Sawan S., McGinn O. (2014) Understanding and exploiting 5T4 oncofoetal glycoprotein expression. Semin Cancer Biol 29: 13–20. [DOI] [PubMed] [Google Scholar]
- Stinchcombe T. (2014) Novel agents in development for advanced non-small cell lung cancer. Ther Adv Med Oncol 6: 240–253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sunshine J., Taube J. (2015) PD-1/PD-L1 inhibitors. Curr Opin Pharmacol 23: 32–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Suzuki M., Cheung N. (2015) Disialoganglioside GD2 as a therapeutic target for human diseases. Expert Opin Ther Targets 19: 349–362. [DOI] [PubMed] [Google Scholar]
- Swaika A., Hammond W., Joseph R. (2015) Current state of anti-PD-L1 and anti-PD-1 agents in cancer therapy. Mol Immunol 67: 4–17. [DOI] [PubMed] [Google Scholar]
- Szabo E. (2003) MUC1 expression in lung cancer. Methods Mol Med 74: 251–258. [DOI] [PubMed] [Google Scholar]
- Tarhini A., Leng S., Moschos S., Yin Y., Sander C., Lin Y., et al. (2012) Safety and immunogenicity of vaccination with MART-1 (26–35, 27L), gp100 (209–217, 210M), and tyrosinase (368–376, 370D) in adjuvant with PF-3512676 and GM-CSF in metastatic melanoma. J Immunother 35: 359–366. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Terme M., Pernot S., Marcheteau E., Sandoval F., Benhamouda N., Colussi O., et al. (2013) VEGFA-VEGFR pathway blockade inhibits tumor-induced regulatory T-cell proliferation in colorectal cancer. Cancer Res 73: 539–549. [DOI] [PubMed] [Google Scholar]
- Tewari K., Sill M., Long H., 3rd, Penson R., Huang H., Ramondetta L., et al. (2014) Improved survival with bevacizumab in advanced cervical cancer. N Engl J Med 370: 734–743. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thatcher N., Hirsch F., Luft A., Szczesna A., Ciuleanu T., Dediu M., et al. (2015) Necitumumab plus gemcitabine and cisplatin versus gemcitabine and cisplatin alone as first-line therapy in patients with stage IV squamous non-small-cell lung cancer (SQUIRE): an open-label, randomised, controlled phase 3 trial. Lancet Oncol 16: 763–774. [DOI] [PubMed] [Google Scholar]
- Topalian S., Hodi F., Brahmer J., Gettinger S., Smith D., McDermott D., et al. (2012) Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med 366: 2443–2454. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Topalian S., Sznol M., McDermott D., Kluger H., Carvajal R., Sharfman W., et al. (2014) Survival, durable tumor remission, and long-term safety in patients with advanced melanoma receiving nivolumab. J Clin Oncol 32: 1020–1030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Torres M., Chakraborty S., Souchek J., Batra S. (2012) Mucin-based targeted pancreatic cancer therapy. Curr Pharm Des 18: 2472–2481. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Turriziani M., Fantini M., Benvenuto M., Izzi V., Masuelli L., Sacchetti P., et al. (2012) Carcinoembryonic antigen (CEA)-based cancer vaccines: recent patents and antitumor effects from experimental models to clinical trials. Recent Pat Anticancer Drug Discov 7: 265–296. [DOI] [PubMed] [Google Scholar]
- Ulloa-Montoya F., Louahed J., Dizier B., Gruselle O., Spiessens B., Lehmann F., et al. (2013) Predictive gene signature in MAGE-A3 antigen-specific cancer immunotherapy. J Clin Oncol 31: 2388–2395. [DOI] [PubMed] [Google Scholar]
- Uyttenhove C., Pilotte L., Theate I., Stroobant V., Colau D., Parmentier N., et al. (2003) Evidence for a tumoral immune resistance mechanism based on tryptophan degradation by indoleamine 2,3-dioxygenase. Nat Med 9: 1269–1274. [DOI] [PubMed] [Google Scholar]
- Vacchelli E., Aranda F., Eggermont A., Galon J., Sautes-Fridman C., Zitvogel L., et al. (2014) Trial watch: tumor-targeting monoclonal antibodies in cancer therapy. Oncoimmunology 3: e27048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Van Cutsem E., Kohne C., Hitre E., Zaluski J., Chang Chien C., Makhson A., et al. (2009) Cetuximab and chemotherapy as initial treatment for metastatic colorectal cancer. N Engl J Med 360: 1408–1417. [DOI] [PubMed] [Google Scholar]
- Van Cutsem E., Peeters M., Siena S., Humblet Y., Hendlisz A., Neyns B., et al. (2007) Open-label phase III trial of panitumumab plus best supportive care compared with best supportive care alone in patients with chemotherapy-refractory metastatic colorectal cancer. J Clin Oncol 25: 1658–1664. [DOI] [PubMed] [Google Scholar]
- van der Bruggen P., Traversari C., Chomez P., Lurquin C., De Plaen E., Van den Eynde B., et al. (1991) A gene encoding an antigen recognized by cytolytic T lymphocytes on a human melanoma. Science 254: 1643–1647. [DOI] [PubMed] [Google Scholar]
- Vansteenkiste J., Zielinski M., Linder A., Dahabreh J., Gonzalez E., Malinowski W., et al. (2013) Adjuvant MAGE-A3 immunotherapy in resected non-small-cell lung cancer: phase II randomized study results. J Clin Oncol 31: 2396–2403. [DOI] [PubMed] [Google Scholar]
- Veillette A., Guo H. (2013) CS1, a SLAM family receptor involved in immune regulation, is a therapeutic target in multiple myeloma. Crit Rev Oncol Hematol 88: 168–177. [DOI] [PubMed] [Google Scholar]
- Vermorken J., Mesia R., Rivera F., Remenar E., Kawecki A., Rottey S., et al. (2008) Platinum-based chemotherapy plus cetuximab in head and neck cancer. N Engl J Med 359: 1116–1127. [DOI] [PubMed] [Google Scholar]
- Vermorken J., Trigo J., Hitt R., Koralewski P., Diaz-Rubio E., Rolland F., et al. (2007) Open-label, uncontrolled, multicenter phase II study to evaluate the efficacy and toxicity of cetuximab as a single agent in patients with recurrent and/or metastatic squamous cell carcinoma of the head and neck who failed to respond to platinum-based therapy. J Clin Oncol 25: 2171–2177. [DOI] [PubMed] [Google Scholar]
- Vigneron N., Stroobant V., Van den Eynde B., van der Bruggen P. (2013) Database of T cell-defined human tumor antigens: the 2013 update. Cancer Immun 13: 15. [PMC free article] [PubMed] [Google Scholar]
- Vlad A., Kettel J., Alajez N., Carlos C., Finn O. (2004) MUC1 immunobiology: from discovery to clinical applications. Adv Immunol 82: 249–293. [DOI] [PubMed] [Google Scholar]
- Waller C. (2014) Imatinib mesylate. Recent Results Cancer Res 201: 1–25. [DOI] [PubMed] [Google Scholar]
- Wang L., Rubinstein R., Lines J., Wasiuk A., Ahonen C., Guo Y., et al. (2011) VISTA, a novel mouse Ig superfamily ligand that negatively regulates T cell responses. J Exp Med 208: 577–592. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang Y., Dong Q., Miao Y., Fu L., Lin X., Wang E. (2013) Clinical significance and biological roles of SPAG9 overexpression in non-small cell lung cancer. Lung Cancer 81: 266–272. [DOI] [PubMed] [Google Scholar]
- Wehrle J., Pahl H., von Bubnoff N. (2014) Ponatinib: a third-generation inhibitor for the treatment of CML. Recent Results Cancer Res 201: 99–107. [DOI] [PubMed] [Google Scholar]
- Weiner L., Surana R., Wang S. (2010) Monoclonal antibodies: versatile platforms for cancer immunotherapy. Nat Rev Immunol 10: 317–327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Whitehurst A. (2014) Cause and consequence of cancer/testis antigen activation in cancer. Annu Rev Pharmacol Toxicol 54: 251–272. [DOI] [PubMed] [Google Scholar]
- Wilke H., Muro K., Van Cutsem E., Oh S., Bodoky G., Shimada Y., et al. (2014) Ramucirumab plus paclitaxel versus placebo plus paclitaxel in patients with previously treated advanced gastric or gastro-oesophageal junction adenocarcinoma (RAINBOW): a double-blind, randomised phase 3 trial. Lancet Oncol 15: 1224–1235. [DOI] [PubMed] [Google Scholar]
- Wu J., Wei J., Meng K., Chen J., Gao W., Zhang J., et al. (2009) Identification of an HLA-A*0201-restrictive CTL epitope from MUC4 for applicable vaccine therapy. Immunopharmacol Immunotoxicol 31: 468–476. [DOI] [PubMed] [Google Scholar]
- Wurz G., Kao C., Wolf M., DeGregorio M. (2014) Tecemotide: an antigen-specific cancer immunotherapy. Hum Vaccin Immunother 10: 3383–3393. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xie C., Wang G. (2015) XAGE-1b Cancer/Testis Antigen Is a Potential Target for Immunotherapy in Prostate Cancer. Urol Int 94: 354–362. [DOI] [PubMed] [Google Scholar]
- Yamada T., Azuma K., Muta E., Kim J., Sugawara S., Zhang G., et al. (2013) EGFR T790M mutation as a possible target for immunotherapy; identification of HLA-A*0201-restricted T cell epitopes derived from the EGFR T790M mutation. PLoS One 8: e78389. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang Z., Grote D., Ziesmer S., Niki T., Hirashima M., Novak A., et al. (2012) IL-12 upregulates TIM-3 expression and induces T cell exhaustion in patients with follicular B cell non-Hodgkin lymphoma. J Clin Invest 122: 1271–1282. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yonezawa A., Dutt S., Chester C., Kim J., Kohrt H. (2015) Boosting Cancer Immunotherapy with Anti-CD137 Antibody Therapy. Clin Cancer Res 21: 3113–3120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yoshitake Y., Fukuma D., Yuno A., Hirayama M., Nakayama H., Tanaka T., et al. (2015) Phase II clinical trial of multiple peptide vaccination for advanced head and neck cancer patients revealed induction of immune responses and improved OS. Clin Cancer Res 21: 312–321. [DOI] [PubMed] [Google Scholar]
- Zappasodi R., Bongarzone I., Ghedini G., Castagnoli L., Cabras A., Messina A., et al. (2011) Serological identification of HSP105 as a novel non-Hodgkin lymphoma therapeutic target. Blood 118: 4421–4430. [DOI] [PubMed] [Google Scholar]
- Zaravinos A. (2014) An updated overview of HPV-associated head and neck carcinomas. Oncotarget 5: 3956–3969. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang L., Chen J., Song X., Wen W., Li Y., Zhang Y., et al. (2013) Cancer/testis antigen HCA587-derived long peptide vaccine generates potent immunologic responses and antitumor effects in mouse model. Oncol Res 21: 193–200. [DOI] [PubMed] [Google Scholar]
- Zhang Q., He S., Shen N., Luo B., Fan R., Fu J., et al. (2014) Overexpression of MAGE-D4 in colorectal cancer is a potentially prognostic biomarker and immunotherapy target. Int J Clin Exp Pathol 7: 3918–3927. [PMC free article] [PubMed] [Google Scholar]
- Zhang R., Bines S., Ruby C., Kaufman H. (2012) TroVax((R)) vaccine therapy for renal cell carcinoma. Immunotherapy 4: 27–42. [DOI] [PubMed] [Google Scholar]
- Zhang S., Zhou X., Yu H., Yu Y. (2010) Expression of tumor-specific antigen MAGE, GAGE and BAGE in ovarian cancer tissues and cell lines. BMC Cancer 10: 163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhou Q., Guo A., Xu C., An S., Wang Z., Yang S., et al. (2008) A dendritic cell-based tumour vaccine for lung cancer: full-length XAGE-1b protein-pulsed dendritic cells induce specific cytotoxic T lymphocytes in vitro. Clin Exp Immunol 153: 392–400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhu C., Anderson A., Schubart A., Xiong H., Imitola J., Khoury S., et al. (2005) The Tim-3 ligand galectin-9 negatively regulates T helper type 1 immunity. Nat Immunol 6: 1245–1252. [DOI] [PubMed] [Google Scholar]
- Zhu Y., Zhang J., Liang W., Zhu R., Wang B., Miao Y., et al. (2014) Pancreatic cancer counterattack: MUC4 mediates Fas-independent apoptosis of antigen-specific cytotoxic T lymphocyte. Oncol Rep 31: 1768–1776. [DOI] [PubMed] [Google Scholar]