Submit manuscript...
MOJ
eISSN: 2373-4442

Immunology

Review Article Volume 2 Issue 5

Improving of Antitumor Immunity and Therapeutic Efficacy of Cancer Vaccines and Adoptive Immunotherapies Using Monoclonal Antibodies

Farashi Bonab S,2 Nemat Khansari 1

1Department of Immunology, Tehran University of medical Sciences, Iran
2American Medical Diagnostic Laboratory, USA

Correspondence: Nemat Khansari, American Medical Diagnostic Laboratory, 1665 Garden Grove Blvd, Garden Grove CA 92843, USA, Tel 1(949)228-8290

Received: October 29, 2015 | Published: November 20, 2015

Citation: Farashi-Bonab S, Khansari N (2015) Improving of Antitumor Immunity and Therapeutic Efficacy of Cancer Vaccines and Adoptive Immunotherapies Using Monoclonal Antibodies. MOJ Immunol 2(5): 00062. DOI: 10.15406/moji.2015.02.00062

Download PDF

Abstract

In the past two decades, immunotherapy has become a novel therapeutic modality for cancer patients. In this therapeutic modality, the immune system of patient’s body is augmented to acquire the ability of recognition and destruction of tumor/cancer cells. Cancer immunotherapy is divided into two forms: passive immunotherapy and active immunotherapy. In passive immunotherapy monoclonal antibodies or ex vivo-proliferated/activated effector immune cells, especially T cells, are used to destroy malignant cells.

Active immunotherapy is performed using various approaches, such as tumor specific/associated antigens and antigen-loaded dendritic cell vaccines, to generate patient’s immune responses against tumor/cancer cells. Recently, some approaches of immunotherapy have been approved by U.S. FDA for use in patients with advanced cancers. However, anticancer efficacy of most immunotherapeutic strategies should be improved to obtain satisfactory results in cancer patients. In this paper, we discuss the application of monoclonal antibodies targeting the cell surface molecules related with the antitumor responses of effector immune cells and their beneficial effects on cancer vaccines and other anticancer therapies.

Keywords: Cancer; Immunotherapy; Cancer vaccine; Monoclonal antibody; Immune

Abbreviations

U.S. FDA: United States Food and Drug Administration; IL: Interleukin; TCR: T Cell Receptor; GITR: Glucocorticoid Induced TNF Receptor Family-Related Protein; TGF-β: Transforming Growth Factor-β; TNFRSF-4: Tumor Necrosis Factor Receptor Super Family Member 4; TNF: Tumor Necrosis Factor; Bcl-XL: B Cell Lymphoma-Extra Large; Bcl-2: B Cell Lymphoma-2; CTLA-4: Cytotoxic T Lymphocyte Associated Antigen-4; PD-1: Programmed Death-1; Foxp3: Fork-head Box Protein P3; NY-ESO-1: New York Esophageal Squamous-Cell Carcinoma-1; NK: Natural Killer; NKT: Natural Killer T; PD-L: PD-Ligand; MART-1: Melanoma Antigen Recognized By T Cell-1; BCG: Bacillus Calmette-Guerrin

Introduction

At present, immunotherapy is a standard therapy for some types of cancer. Immuno adjuvants, monoclonal antibodies, and anticancer vaccines are used in cancer patients. IL-2 is a major cytokine that has been used as an immuno adjuvant in some cancer patients, especially patients with metastatic melanoma [1]. However, toxicity and little therapeutic responses limit its use in cancer patients. In a few studies, vaccination with tumor specific/associated antigens and immunogenic vectors, including melanoma cancer antigen gp100 (gp209-2M) and gp209-2M-expressing fowl pox viral vector, have been successfully used to increase antitumor immune responses [2,3]. In general, cancer vaccines did not induce notable toxicity. However, vaccination of patients with cancer antigens showed limited successes [4-6] and their efficacy was lower than 5%.

In primary studies in 1980s, tumor infiltrating lymphocytes were found to have antitumor activities. In mice, tumor infiltrating lymphocytes showed potent antitumor effects after in vitro proliferation [7]. Tumor infiltrating lymphocytes from human melanoma tumors were reported to be able to recognize autologous tumor cells in a MHC-restricted manner [8]. Adoptive immunotherapy with autologous tumor infiltrating lymphocytes in combination with exogenous IL-2 induced therapeutic responses (30%) in patients with melanoma [9]. Tumor-reactive tumor infiltrating lymphocytes were also isolated and expanded from other types of cancer, including renal cell carcinoma and glioma [10,11]. However, this therapeutic approach did not have appropriate efficacy in most types of cancer. Effector T cells were also expanded from tumor draining lymph nodes [12,13]. In our study, tumor draining lymph node cells did not show appropriate antitumor activity (unpublished data).

Generation of tumor antigen-specific lymphocytes by vaccination with peptide vaccines such as gp100 did not lead to promising therapeutic antitumor responses [14]. In addition, immunotherapy with transgenic lymphocytes expressing tumor antigen-specific TCRs in mice bearing transgenic tumors expressing the cognate antigen demonstrated that high frequencies of tumor-specific cytotoxic lymphocytes is not sufficient for prevention of tumor growth [15,16]. Thus, generation of tumor-specific T cells via vaccination is not adequate for elimination of established tumors.

Tumor-specific T cells have been reported in tumor tissues, tumor draining lymph nodes, and the peripheral blood of cancer patients. But, tumor-specific T cells compose a little proportion of lymphocyte pool in cancer patients. For example, lower than 1% of peripheral blood lymphocytes and 2-15% of tumor infiltrating lymphocytes were reported to be tumor-specific T cells in human melanomas [17]. Polyclonal activation of lymphocytes by anti-CD3 and anti-CD28 antibodies as well as IL-2, which are commonly used for ex vivo proliferation of lymphocytes, leads to expansion of both tumor-specific and non-specific lymphocytes and also up-regulation of CD25 on lymphocytes. More importantly, increased levels of regulatory T cells have been observed after polyclonal activation of T cells in the presence of IL-2 [18].

Antigen-specific stimulation of T cells can enhance the number of tumor-specific T cells [19]. Vaccination with tumor antigen-pulsed dendritic cells induced antigen-specific T cells, both in vitro and in vivo. In several murine tumor models, prophylactic dendritic cell vaccines showed suitable efficacy in prevention of tumor growth. However, most therapeutic dendritic cell vaccines were unable to reject established tumors or induce long-lasting delay in tumor growth [20]. These findings indicate that improving the antitumor efficacy of ex vivo expanded lymphocytes, tumor antigen vaccines, and dendritic cell vaccines are necessary.

Augmenting of antigen-specific responses of antitumor effector immune cells
Triggering of antigen-specific immune responses of effector immune cells is a complex process and needs signaling through several co-stimulatory molecules. On the other hand, immune responses are carefully regulated through various inhibitory mechanisms to prevent autoimmunity and deleterious effects of immune responses on healthy tissues.

Several mechanisms are recruited by tumor/cancer cells to suppress antitumor activity of effector immune cells, including altered expression of MHC class I molecules, expression of Fas ligand, expression of inhibitory molecules such as B7-H1 (PD-L1), production of immunosuppressive soluble agents, as well as the immunosuppressive activity of immunoregulatory cells such as regulatory T cells and myeloid derived suppressor cells [21]. T cells that recognize tumor antigens in the absence of co-stimulatory signals become anergic or may be deleted [22]. Signaling through inhibitory molecules also results in suppression of antitumor activity of effector immune cells. Indeed, lack of signaling of co-stimulatory molecules and/or signaling through inhibitory molecules is an important reason of impaired tumor immunosurveillance in cancer patients. Agonistic/antagonistic monoclonal antibodies targeting these cell surface molecules can be beneficial for augmentation of antitumor immunity. These antibodies have also potential to improve therapeutic efficacy of adoptive immunotherapy and cancer vaccines.
Two decades ago, monoclonal antibodies have been used to enhance antitumor immune responses. In murine tumor models, targeting of several co-stimulatory/inhibitory molecules on the cell surface of effector immune cells by monoclonal antibodies led to increased antitumor immunity [23-26]. Based on satisfactory results from these preclinical studies in mice, a number of clinical trials were performed using monoclonal antibodies targeting cell surface co-stimulatory/inhibitory molecules. At present, some of these antibodies are approved by U.S. FDA for use in cancer therapy. Antagonistic anti-CTLA-4 monoclonal antibody is the earliest antibody that approved by U.S. FDA for use in patients with cancer in 2011.

Several agonistic antibodies for triggering co-stimulatory signals and antagonistic antibodies for blocking inhibitory molecules on the cell surface of immune cells as well as tumor/cancer cells and tumor stromal cells have been used in various murine tumor models and cancer patients. Some of these antibodies showed beneficial effects on antitumor immunity.

Agonistic monoclonal antibodies
Anti-GITR monoclonal antibody: GITR is expressed on regulatory T cells and other T cell
Subsets. GITR can deliver a co-stimulatory signal to naïve CD4+ T cells and CD8+ T cells, especially when TCR stimulation is weak [27]. GITR engaging with agonistic antibodies induced cytokine production and proliferation of T cells in vitro [28-29]. In addition, agonistic anti-GITR monoclonal antibodies have been reported to block suppressive activity of regulatory T cells [30,31]. In preclinical studies, Agonistic anti-GITR monoclonal antibody showed antitumor effects in various murine tumor models [32-34]. Anti-GITR monoclonal antibody increased antitumor efficacy of melanoma specific DNA vaccine such as vaccine-induced CD8+ T cell responses in a murine melanoma tumor model [35].

In another murine melanoma tumor model, vaccination with dendritic cells engineered to secrete anti-GITR antibodies had therapeutic effects [36]. Anti-GITR monoclonal antibody in combination with anti-PD-1 monoclonal antibody induced potent antitumor immunity. Importantly, combinational anti-GITR-anti-PD-1 antibody therapy together with chemotherapy (cisplatin or paclitaxel) further increased the antitumor immunity [37]. In patients with head and neck squamous cell carcinoma, regulatory T cells (CD25high) infiltrating tumor tissue expressed GITR, IL-10, and TGF-β, but, peripheral blood regulatory T cells did not expressed GITR, IL-10, and TGF-β. These GITR expressing tumor infiltrating regulatory T cells showed more suppressive activity than that peripheral blood regulatory T cells [38]. These findings suggest that administration of agonistic anti-GITR monoclonal antibodies as a monotherapy or in combination with other cancer therapy modalities may have beneficial effects.

Anti-OX40 monoclonal antibody: OX40 (CD134, TNFRSF4), another member of TNF receptor
Family is expressed on T cells and acts as a co-stimulatory molecule [39]. This molecule is transiently expressed on T cells upon TCR stimulation. The maximum expression level of OX40 is 48 hours after TCR stimulation and its expression is impeded 72-96 hours later [40]. OX40 ligand is expressed on antigen presenting cells including dendritic cells, macrophages, and B cells as well as endothelial cells after their activation [41,42]. OX40-OX40 ligand interaction leads to production of Th1 cytokines and up-regulation of anti-apoptotic proteins such as Bcl-XL and Bcl-2 [43-44].

Agonistic anti-OX40 monoclonal antibodies enhanced antitumor immune responses in several murine tumor models. Anti-OX40 monoclonal antibody increased CD8+ T cell infiltration to tumor tissue and decreased immunosuppression in the tumor [45]. OX40 engagement could deplete or block suppressive activity of regulatory T cells and augmented antitumor immunity [46,47]. Anti-OX40 monoclonal antibody together with chemotherapy (cyclophosphamide) led to apoptosis of regulatory T cells at tumor sites and produced potent antitumor immunity capable of regressing established B16 melanoma tumor [48]. Anti-OX40 monoclonal antibody together with radiotherapy resulted in therapeutic antitumor immunity against lung cancer [49]. Agonistic anti-OX40 monoclonal antibody in combination with antagonistic anti-CTLA-4 monoclonal antibody induced potent effector T cells with antitumor activity in mice [50]. Agonistic anti-OX40 monoclonal antibody in combination with antagonistic anti-PD-1 monoclonal antibody synergistically produced protective antitumor immunity against murine ovarian tumor [51].

Administration of antibodies targeting other cell surface co-stimulatory molecules such as CD28 did not show promising results. In a phase I clinical trial, agonistic anti-CD28 monoclonal antibody (TGN142) induced cytokine-release syndrome in healthy volunteers due to strong activation of T cells and subsequent release of plentiful amount of pro-inflammatory cytokines [52].

Antagonistic (Blocking/Depleting) monoclonal antibodies
Anti-CTLA-4 monoclonal antibody: CTLA-4 is a transmembrane protein expressed on the cell
Surface of T cells within 24-48 hours after T cell activation. CTLA-4 down regulates immune responses of T cells. This molecule is also expressed constitutively on regulatory T cells [53]. CTLA-4 is structurally homologous to CD28 and competes with CD28 in attachment to CD80 and CD86. In comparison with CD28, CTLA-4 has very more affinity and avidity to CD80 and CD86 molecules and confers an inhibitory signal to T cell [54]. Indeed, CTLA-4 has a crucial role in modulation of activation of naïve T cells and memory T cells. Increased frequencies of CD4+CD25+ T cells expressing high levels of Foxp3 and CTLA-4 were reported in the lymph nodes of patients with B cell non-Hodgkin's lymphoma [55].

In preclinical studies, CTLA-4 blockade was effective in some murine tumor models including glioma, sarcoma, ovarian carcinoma, and bladder carcinoma, but, was ineffective in other tumor types such as breast, prostate, and colorectal tumors [56]. In a phase I pilot study in 2002, for the first time, administration of a single dose of 3mg/kg of Ipilimumab (MDX010, BMS-734016), a human monoclonal antibody against CTLA-4, to patients with inoperable melanoma resulted in durable partial responses in two of 17 patients [57].

In other study, CTLA-4 blockade using anti-CTLA-4 monoclonal antibodies led to cancer regression as well as induction of autoimmunity in patients with metastatic melanoma [58]. Latterly, in several phase II clinical studies, Iplilimumab was used to increase antitumor immunity in patients with metastatic melanoma. Increased patient's survival was observed in some patients treated with ipilimumab [59-62]. In a phase III clinical trial in patients with advanced melanoma, patients treated with ipilimumab (10mg/kg) and Dacarbazine showed significantly more survival rate than patients treated with Dacarbazine alone [63]. Administration of ipilimumab after curative surgery also enhanced significantly overall survival in patients with recurrent able advanced melanoma [64].

In a phase II clinical trial, administration of ipilimumab resulted in some clinical effects in patients with metastatic renal cell carcinoma [65]. Ipilimumab also produced objective responses in patients with prostate cancer [66,67]. In contrast, in a phase III clinical trial, administration of ipilimumab, when compared to placebo, did not increase overall survival in patients with castration-resistant metastatic prostate cancer pretreated with doxetaxel and single dose radiotherapy [68]. Similarly, ipilimumab did not produce clinical responses in patients with metastatic pancreatic adenocarcinoma [69]. Combinational therapy with chemotherapy and ipilimumab had moderated efficacy in patients with non-small cell lung cancer [70]. In a phase II clinical study, Tremelimumab, another human monoclonal antibody against CTLA-4, did not show obvious clinical effects in 45 patients with treatment-refractory colorectal cancer [71]. Drug-related adverse effects were observed in some patients treated with ipilimumab which were usually manageable. However, a few deaths (less than 1%) were observed after treatment with anti-CTLA monoclonal antibody. In a phase III clinical trial, five patients (1%) died due to the adverse effects of ipilimumab [64].

In some studies, CTLA-4 blockade has been resulted in improved immune responses to tumor specific/associated antigens. In patients with prostate cancer, CTLA-4 blockade led to decreased levels of Prostate-specific antigen [66]. In patients with melanoma treated with ipilimumab, patients who had high serum levels of antibodies against NY-ESO-1 and NY-ESO-1-specific CD8+ T cells showed more clinical responses to ipilimumab [72]. However, this correlation between response to ipilimumab and appearance of anti-NY-ESO-1 antibodies in patient's sera was not observed in another study [73].

Ipilimumab has also been used in combination with cancer vaccines in patients with metastatic melanoma [74,75]. In a phase III clinical trial in patients with progressive metastatic melanoma pretreated with chemotherapy or IL-2, administration of ipilimumab with or without a gp100 peptide vaccine improved overall survival, when compared to peptide vaccine alone. Overall survival rates in treated groups, including ipilimumab, ipilimumab together with gp100 vaccine, and gp100 vaccine alone, were 10.1 months, 10 months, and 6.4 months, respectively [60].

Anti-PD-1 monoclonal antibody: PD-1 (CD279) is expressed on the cell surface of T cells,
B cells, NK cells, NKT cells, activated monocytes, and dendritic cells. Interaction of PD-1 with its ligands PD-L1 and PD-L2 triggers an inhibitory signaling in T cells which leads to down regulation of Bcl-XL expression and T cell differentiation. On the other hand, signaling through PD-L1 and PD-L2 alters cytokine production and maturation of dendritic cells [76]. These inhibitory signaling pathways help to avoid deleterious effects of immune responses during infection or inflammation. However, these signals can also impair antitumor immune responses [77]. Thus, blocking these pathways may improve antitumor immunity.

Nivolumab (BMS-936558), a human IgG4 anti-PD-1 monoclonal antibody, has been used in several studies to block PD-1 signaling. In a phase I clinical trial in patients with advanced solid tumors, including non-small cell lung cancer, melanoma, prostate cancer, renal cell carcinoma, and colorectal carcinoma (n=236), administration of Nivolumab led to clinical responses in patients with non-small cell lung cancer (18%), melanoma (28%), and renal cell carcinoma (27%). Drug-related adverse events were tolerable [78]. PD-1 blocking resulted in lower toxicities than CTLA-4 blockade. But, pneumonia was reported in 1.5% of patients treated with Nivolumab [79]. In 39 patients with metastatic melanoma, colorectal cancer, castration-resistant prostate cancer, non-small cell lung cancer, renal cell carcinoma, anti-PD-1 monoclonal antibody (MDX-1106) was well tolerated and induced durable complete response in one patients with colorectal cancer, two partial responses in patients with melanoma and renal cell carcinoma, and significant tumor regressions in two patients with melanoma and non-small cell lung cancer [80]. In patients with hematopoietic malignancies (n=17), PD-1 blocking using anti-PD-1 monoclonal antibody CT-011, a human IgG1 monoclonal antibody against PD-1, produced clinical responses in 33% of patients. One patient with follicular lymphoma completely treated [81]. In previously untreated patients with metastatic melanoma, administration of Nivolumab was associated with significant improved overall survival and progression-free survival when compared to Dacarbazine [79].

Administration of Pembrolizumab (MK-3475), a human IgG4 monoclonal antibody against PD-1, led to long-lasting therapeutic responses in 34% of patients with advanced melanoma. Pretreatment with ipilimumab or IL-2 did not affect the activity of Pembrolizumab in these patients [82]. Pembrolizumab was also associated with durable antitumor activity in patients with melanoma (one patient with complete response and three patients with partial response), Merkel cell carcinoma (one patient with complete response), and stable disease in patients (15 patients) with other malignancies [83]. New findings indicate that Pembrolizumab is a suitable choice for treatment of Ipilimumab-refractory melanoma [84]. In patients with reseated metastatic melanoma (n=33), Nivolumab in combination with a multi-peptide vaccine (gp100, MART-1, and NY-ESO-1 with Montanide ISA 51 VG) was well tolerated and produced immunologic activity with promising survival [85].

In 2014, U.S. FDA approved Keytruda (Pembrozomide) for use in patients with advanced or un-resectable melanoma who are no longer responding to other therapies. Recently, U.S. FDA also approved Opivido (Nivolumab) for the treatment of patients with previously treated metastatic squamous non-small cell lung cancer.

Anti-PD-L1 monoclonal antibody: PD-L1 (B7-H1) is expressed on a variety of hematopoietic
and non-hematopoietic cells and PD-L2 is expressed on dendritic cells, macrophages, B cells, and mast cells. Expression of these molecules is up regulated in inflammation [86]. More importantly, expression of PD-L1 and, to some extent, PD-L2 is reported in various types of tumor/cancer including ovarian cancer, breast cancer, cervical cancer, colon cancer, non-small cell lung cancer, glioblastoma, pancreatic cancer, gastric cancer, melanoma, and urothelial cancer, as well as hematopoietic malignancies such as Hodgkin's lymphoma, B cell lymphoma, T cell lymphoma, multiple myeloma, acute myeloid leukemia, chronic lymphocytic leukemia, and adult T cell leukemia/lymphoma. Expression of PD-L1 was correlated with disease prognosis in some cancer patients [87-90]. On the other hand, PD-1 has been reported to be expressed at high levels on tumor-specific T cells [91,92]. Thus, interaction of PD-1 on T cells with PD-L1 expressed on tumor cells and immune cells can hamper immune responses of T cells [76].

In patients with urothelial bladder carcinoma, expression of PD-L1 was high in tumor tissue of patients who did not show therapeutic responses to BCG vaccine [93]. These findings indicate that PD-1-PD-L signaling pathways can be an important immunosuppressive mechanism at tumor sites. In a multicenter phase I clinical trial, anti-PD-L1 monoclonal antibody BMS-936559, a human IgG4 monoclonal antibody against PD-L1, was administrated to 217 patients with advanced cancers, including non-small cell lung cancer (n=75), melanoma (n=55), colorectal cancer (n=18), renal cell carcinoma (n=17), ovarian cancer (n=17), pancreatic cancer (n=14), gastric cancer (n=7), and breast cancer (n=4). Anti-PD-L1 monoclonal antibody produced objective responses in patients with melanoma (17%), renal cell carcinoma (12%), and non-small cell lung carcinoma (10%). One patient with ovarian cancer also showed objective response.

Drug-related toxicities were lower than that induce by CTLA-4 blockade. Furthermore, pneumonia was not observed in patients treated with this antibody [94]. In a phase I clinical study, administration of anti-PD-L1 monoclonal antibody (MPDL3280A) led to clinical activity in patients with metastatic urothelial bladder carcinoma, and this clinical activity was correlated with PD-L1 expression on tumor infiltrating immune cells [95]. Expression of PD-L1 at tumor sites was correlated with patient's responses to anti-PD-1 monoclonal antibody therapy [96-100]. Other anti-PD-L1 monoclonal antibodies (MPDL3280A and MED14736) also produced objective responses in patients with non-small cell lung cancer, metastatic renal cell carcinoma, metastatic bladder carcinoma, and head and neck squamous cell carcinoma [100-105]. Table 1 shows recently reported clinical trials of anti-PD-1 and anti-PD-L1 monoclonal antibodies in cancer patients.

Anti-CD25 monoclonal antibody: Regulatory T cells constitutively express high levels of CD25
on their cell surface. Increased frequencies of regulatory T cells have been reported in most types of cancer and elevated levels of this T cell subset were associated with poor prognosis in some cancer patients. Accordingly, administration of anti-CD25 monoclonal antibody resulted in enhanced antitumor immunity [21]. Co-administration of anti-CD25 and anti-CTLA-4 monoclonal antibodies synergistically decreased suppression of cytotoxic T cells and NK cells [106]. Anti-CD25 monoclonal antibody in combination with IL-12 gene transduction led to rejection of tumors in mice. However, anti-CD25 monoclonal antibody did not affect tumor growth when used as a monotherapy [107].

Anti-CD25 monoclonal antibody can also be used in combination with adoptive immunotherapy or cancer vaccines. But, expression of CD25 is induced on T cells after activation [108] and anti-CD25 monoclonal antibody may deplete these recently activated effector T cells. Therefore, the appropriate time of anti-CD25 monoclonal antibody injection should be considered in combinational therapy of anti-CD25 antibody with adoptive immunotherapy and cancer vaccine. In several studies, depletion of CD25+ regulatory T cells before vaccination has increased vaccine-mediated antitumor immune responses. Anti-CD25 monoclonal antibody as a monotherapy and also in combination with whole tumor cell vaccine (irradiated pancreas adenocarcinoma cells) induced tumor specific immune responses [109]. Furthermore, anti-CD25 monoclonal antibody in combination with dendritic cell vaccine led to long-term immunity against experimental glioma in mice [110]. In a murine melanoma model, combinational therapy with anti-CD25 monoclonal antibody and dendritic cell-tumor fusion vaccine significantly reduced pulmonary metastasis compared to monoclonal antibody or fusion vaccine alone [111].

Target

Antibody

Cancer Type

Patient’s Number

Therapeutic Responses

PD-1

Pembrolizumab

Progressive metastatic colorectal carcinoma with or without mismatch-repair deficiency

41 patients

Objective response rate and progression-free survival rate were 40% (4 of 10 patients) and 78% (7 of 9 patients), respectively, for mismatch repair–deficient colorectal cancers and 0% (0 of 18 patients) and 11% (2 of 18 patients) for mismatch repair–proficient colorectal cancers.

PD-1

Nivolumab

Previously heavily treated relapsed or refractory Hodgkin’s lymphoma

23 patients

Objective response was observed in 20 patients (87%), including 17% with a complete response and 70% with a partial response. Three patients (13%) had stable disease. The rate of progression-free survival at 24 weeks was 86%.

PD-1

Nivolumab

Advanced treatment-refractory melanoma

107 patients

Objective responses were observed in 31% of patients (33 of 107) with melanoma. Seven of 107 patients (7%) experienced stable disease lasting for 24 weeks or more.  Overall survival rates of 62% at 1 year and 43% at 2 years were achieved, with a median overall survival of 16.8 months. 

PD-1

Lambrolizumab

Advanced melanoma

135 patients

The confirmed response rate, evaluated by central radiologic review according to the Response Evaluation Criteria in Solid Tumors (RECIST), version 1.1, was 38%.

PD-1

Nivolumab

Advanced melanoma, non-small cell lung cancer, castration-resistant prostate cancer, renal-cell cancer, and colorectal cancer

296 patients

Among 236 patients in whom response could be evaluated, objective responses (complete or partial responses) were observed in 18% of patients with non-small cell lung cancer (14 of 76 patients), 28% of patients with melanoma (26 of 94 patients), and 27% of patients with renal-cell cancer (9 of 33 patients). 

PD-1

Nivolumab

Advanced metastatic melanoma, colorectal cancer, castrate-resistant prostate cancer, non-small cell lung cancer, and renal cell carcinoma

39 patients

Durable complete response was observed in one patient with colorectal cancer and partial responses were observed in two patients with melanoma and renal cell carcinoma. Two patients with melanoma and non-small cell lung cancer experienced significant lesional tumor regressions not meeting partial response criteria.

PD-L1

BMS-936559

Non-small cell lung cancer,

207 patients

Durable objective

 

 

melanoma, colorectal cancer, renal cell cancer, ovarian cancer, pancreatic cancer, gastric cancer, and breast cancer

 

Response rate of 6 to 17% and prolonged stabilization of disease (rates of 12 to 41%

 

 

 

 

At 24 weeks) were observed in patients with non-small cell lung cancer, melanoma, and renal cell cancer.

PD-L1

MPDL3280A

Different tumor types (including locally advanced or metastatic solid tumors, and hematological malignancies)

175 patients

Complete and partial responses (evaluated by Response Evaluation Criteria in Solid Tumors, version 1.1) were observed in 32 of 175 (18%) patients with all tumor types.  Complete and partial responses were observed in 11 of 53 (21%), 11 of 43 (26%), 7 of 56 (13%), and 3 of 23 (13%) of patients with non-small cell lung cancer, melanoma, renal cell carcinoma, and other tumors (including colorectal cancer, gastric cancer, and head and neck squamous cell carcinoma), respectively.

PD-L1

MPDL3280A

Metastatic bladder cancer

68 patients

For patients with a minimum of 6 weeks of follow-up, objective response rates were 43% (13 of 30) for patients with high level of PD-L1 expression in tumor tissues and 11% (4 of 35) for patients with low/without PD-L1 expression in tumors.

Table 1: Clinical trials in which agonistic and antagonistic antibodies were employed.

Conclusion

In cancer patients effector immune cells do not respond to cancer cells. Indeed, cancer cells recruit several mechanisms to evade from cancer immunosurveillance. These mechanisms also impede the therapeutic efficacy of cancer vaccines and adoptive immunotherapies. Augmenting of co-stimulatory signaling and modulation of inhibitory signaling in effector immune cells may induce antitumor immunity. Monoclonal antibodies that target co-stimulatory/inhibitory molecules on the cell surface of effector immune cells, tumor cells, and tumor stromal cells can be a promising approach for enhancing of antitumor immune responses. At present, some of these antibodies are approved by U.S. FDA for use in patients with some advanced cancers which are refractory to other cancer therapy modalities. Agonistic/antagonistic monoclonal antibodies can be used in combination with cancer vaccines and adoptive immunotherapies, and even with conventional cancer therapies, to improve their therapeutic efficacy.

References

  1. Rosenberg SA, Yang JC, White DE, Steinberg SM (1998) Durability of complete responses in patients with metastatic cancer treated with high-dose interleukin-2: identification of the antigens mediating response. Ann Surg 228(3): 307-319.
  2. Salgaller ML, Marincola FM, Cormier JN, Rosenberg SA (1996) Immunization against epitopes in the human melanoma antigen gp100 following patient immunization with synthetic peptides. Cancer Res 56(20): 4749-4757.
  3. Rosenberg SA, Yang JC, Schwartzentruber DJ, Hwu P, Topalian SL, et al. (2003) Recombinant fowl pox viruses encoding the anchor-modified gp100 melanoma antigen can generate antitumor immune responses in patients with metastatic melanoma. Clin Cancer Res 9(8): 2973-2980.
  4. Rosenberg SA, Yang JC, Restifo NP (2004) Cancer immunotherapy: moving beyond current vaccines. Nat Med 10(9): 909-915.
  5. Choudhury A, Mosolits S, Kokhaei P, Hansson L, Palma M, et al. (2006) Clinical results of vaccine therapy for cancer: learning from history for improving the future. Adv Cancer Res 95: 147-202.
  6. Stan R, Wolchok JD, Cohen AD (2006) DNA vaccines against cancer. Hematol Oncol Clin North Am 20(3): 613-636.
  7. Spiess PJ, Yang JC, Rosenberg SA (1987) In vivo antitumor activity of tumor-infiltrating lymphocytes expanded in recombinant interleukin-2. J Natl Cancer Inst 79(5): 1067-1075.
  8. Muul LM, Spiess PJ, Director EP, Rosenberg SA (1987) Identification of specific cytolytic immune responses against autologous tumor in humans bearing malignant melanoma. J Immunol 138(3): 989-995.
  9. Rosenberg SA, Yannelli JR, Yang JC, Topalian SL, Schwartzentruber DJ, et al. (1994) Treatment of patients with metastatic melanoma using autologous tumor-infiltrating lymphocytes and interleukin-2. J Natl Cancer Inst 86(15): 1159-1166.
  10. Figlin RA, Pierce WC, Kaboo R, Tso CL, Moldawer N, et al. (1997) Treatment of metastatic renal cell carcinoma with nephrectomy, interleukin-2 and cytokine-primed or CD8(+) selected tumor infiltrating lymphocytes from primary tumor. J Urol 158(3 Pt 1): 740-745.
  11. Quattrocchi KB, Miller CH, Cush S, Bernard SA, Dull ST, et al. (1999) Pilot study of local autologous tumor infiltrating lymphocytes for the treatment of recurrent malignant gliomas. J Neurooncol 45(2): 141-157.
  12. Yoshizawa H, Sakai K, Chang AE, Shu SY (1991) Activation by anti-CD3 of tumor-draining lymph node cells for specific adoptive immunotherapy. Cell Immunol 134(2): 473-479.
  13. Li Q, Yu B, Grover AC, Zeng X, Chang AE (2002) Therapeutic effects of tumor reactive CD4+ cells generated from tumor-primed lymph nodes using anti-CD3/anti-CD28 monoclonal antibodies. J Immunother 25(4): 304-313.
  14. Rosenberg SA, Yang JC, Restifo NP (2004) Cancer immunotherapy: moving beyond current vaccines. Nat Med 10(9): 909-915.
  15. Wick M, Dubey P, Koeppen H, Siegel CT, Fields PE, et al. (1997) Antigenic cancer cells grow progressively in human hosts without evidence for T cell exhaustion or systemic anergy. J Exp Med 186(2): 229-238.
  16. Prevost-Blondel A, Zimmermann C, Stemmer C, Kulmburg P, Rosenthal FM, et al. (1998) Tumor-infiltrating lymphocytes exhibiting high ex vivo cytolytic activity fail to prevent murine melanoma tumor growth in vivo. J Immunol 161(5): 2187-2194.
  17. Topalian SL, Solomon D, Rosenberg SA (1989) Tumor-specific cytolysis by lymphocytes infiltrating human melanomas. J Immunol 142(10): 3714-3725.
  18. Rouse M, Nagarkatti M, Nagarkatti PS (2013) The role of IL-2 in the activation and expansion of regulatory T-cells and the development of experimental autoimmune encephalomyelitis. Immunobiology 218(4): 674-682.
  19. Dang Y, Knutson KL, Goodell V, dela Rosa C, Salazar LG, et al. (2007) Tumor antigen-specific T-cell expansion is greatly facilitated by in vivo priming. Clin Cancer Res 13(6): 1883-1891.
  20. Farashi-Bonab S, Khansari N (2015) Dendritic cell vaccine and its application in cancer therapy. Int J Vaccines Vaccin 1(1): 00002.
  21. Farashi-Bonab S, Khansari N (2014) Regulatory T cells in cancer patients and their roles in cancer development/progression. MOJ Immunol 1(4): 0024.
  22. Chen L, Linsley PS, Hellstrom, KE (1993) Costimulation of T cells for tumor immunity. Immunol Today 14(10): 483-486.
  23. Allison JP, Hurwitz AA, Leach DR (1995) Manipulation of costimulatory signals to enhance antitumor T-cell responses. Curr Opin Immunol 7(5): 682-686.
  24. Leach DR, Krummel MF, Allison JP (1996) Enhancement of antitumor immunity by CTLA-4 blockade. Science 271(5256): 1734-1736.
  25. Kwon ED, Hurwitz AA, Foster BA, Madias C, Feldhaus AL, et al. (1997) Manipulation of T cell costimulatory and inhibitory signals for immunotherapy of prostate cancer. Proc Natl Acad Sci USA 94(15): 8099-8103.
  26. Yang YF, Zou JP, Mu J, Wijesuriya R, Ono S, et al. (1997) Enhanced induction of antitumor T-cell responses by cytotoxic T lymphocyte-associated molecule-4 blockade: the effect is manifested only at the restricted tumor-bearing stages. Cancer Res 57(18): 4036-4041.
  27. Nocentini G, Ronchetti S, Petrillo MG, Riccardi C (2012) Pharmacological modulation of GITRL/GITR system: therapeutic perspectives. Br J Pharmacol 165(7): 2089-2099.
  28. Kanamaru, F, Youngnak P, Hashiguchi M, Nishioka T, Takahashi T, et al. (2004) Costimulation via glucocorticoid-induced TNF receptor in both conventional and CD25+ regulatory CD4+ T cells. J Immunol 172(12): 7306-7314.
  29. Ronchetti S, Nocentini G, Bianchini R, Krausz LT, Migliorati G, et al. (2007) Glucocorticoid-induced TNFR-related protein lowers the threshold of CD28 costimulation in CD8+ T cells. J Immunol 179(9): 5916-5926.
  30. McHugh RS, Whitters MJ, Piccirillo CA, Young DA, Shevach EM, et al. (2002) CD4(+)CD25(+) immunoregulatory T cells: gene expression analysis reveals a functional role for the glucocorticoid-induced TNF receptor. Immunity 16(2): 311-323.
  31. Shimizu J, Yamazaki S, Takahashi T, Ishida Y, Sakaguchi S (2002) Stimulation of CD25(+)CD4(+) regulatory T cells through GITR breaks immunological self-tolerance. Nat Immunol 3(2): 135-142.
  32. Turk MJ, Guevara-Patino JA, Rizzuto GA, Engelhorn ME, Sakaguchi S, et al. (2004) Concomitant tumor immunity to a poorly immunogenic melanoma is prevented by regulatory T cells. J Exp Med 200(6): 771-782.
  33. Ko K, Yamazaki S, Nakamura K, Nishioka T, Hirota K, et al. (2005) Treatment of advanced tumors with agonistic anti-GITR mAb and its effects on tumor-infiltrating Foxp3+CD25+CD4+ regulatory T cells. J Exp Med 202(7): 885-891.
  34. Cohen AD, Schaer DA, Liu C, Li Y, Hirschhorn-Cymmerman D, et al. (2010) Agonist anti-GITR monoclonal antibody induces melanoma tumor immunity in mice by altering regulatory T cell stability and intra-tumor accumulation. PloS One 5(5): e10436.
  35. Cohen AD, Diab A, Perales MA, Wolchok JD, Rizzuto G, et al. (2006) Agonist anti-GITR antibody enhances vaccine-induced CD8+ T-cell responses and tumor immunity. Cancer Res 66(9): 4904-4912.
  36. Boczkowski D, Lee J, Pruitt S, Nair S (2009) Dendritic cells engineered to secrete anti-GITR antibodies are effective adjuvants to dendritic cell-based immunotherapy. Cancer Gene Ther 16(12): 900-911.
  37. Lu L, Xu X, Zhang B, Zhang R, Ji H, et al. (2014) Combined PD-1 blockade and GITR triggering induce a potent antitumor immunity in murine cancer models and synergizes with chemotherapeutic drugs. J Transl Med 12: 36.
  38. Strauss L, Bergmann C, Szczepanski M, Gooding W, Johnson JT, et al. (2007) A unique subset of CD4+CD25highFoxp3+ T cells secreting IL-10 and TGF-β1 mediates suppression in the tumor microenvironment. Clin Cancer Res 13(15 Pt 1): 4345-4354.
  39. Takeda I, Ine S, Killeen N, Ndhlovu LC, Murata K, et al. (2004) Distinct roles for the OX40-OX40 ligand interaction in regulatory and nonregulatory T cells. J Immunol 172(6): 3580-3589.
  40. Gramaglia I, Weinberg AD, Lemon M, Croft M (1998) OX-40 ligand: a potent costimulatory molecule for sustaining primary CD4 T cell responses. J Immunol 161(12): 6510-6517.
  41. Brocker, T., Gulbranson-Jadge A, Flynn S, Riedinger M, Raykundalia C, et al. (1999) CD4 T cell traffic control: in vivo evidence that ligation of OX40 on CD4 T cells by OX40-ligand expressed on dendritic cells leads to the accumulation of CD4 T cells in B follicles. Eur J Immunol 29(5): 1610-1616.
  42. Sato T, Ishii N, Murata K, Kikuchi K, Nakagawa S, et al. (2002) Consequences of OX40-OX40 ligand interactions in Langerhans cell function: enhanced contact hypersensitivity responses in OX40L-transgenic mice. Eur J Immunol 32(11): 3326-3335.
  43. Evans, DE, Prell RA, Thalhofer CJ, Hurwitz AA, Weinberg AD (2001) Engagement of OX40 enhances antigen-specific CD4(+) T cell mobilization/memory development and humoral immunity: comparison of alphaOX-40 with alphaCTLA-4. J Immunol 167(12): 6804-6811.
  44. Rogers PR, Song J, Gramaglia I, Kileen N, Croft M (2001) OX40 promotes Bcl-xL and Bcl-2 expression and is essential for long-term survival of CD4 T cells. Immunity 15(3): 445-455.
  45. Gough MJ, Ruby CE, Redmond WL, Dhungel B, Brown A, et al. (2008) OX40 agonist therapy enhances CD8 infiltration and decreases immune suppression in the tumor. Cancer Res 68(13): 5206-5215.
  46. Piconese S, Valzasina B, Colombo MP (2008) OX40 triggering blocks suppression by regulatory T cells and facilitates tumor rejection. J Exp Med 205 (4): 825-839.
  47. Bulliard Y, Jolicoeur R, Zhang J, Dranoff G, Wilson NS, et al. (2014) OX40 engagement depletes intratumoral Tregs via activating Fcgamma Rs, leading to antitumor efficacy. Immunol Cell Biol 92(6): 475-480.
  48. Hirschhorn-Cymerman D, Rizzuto GA, Merghoub T, Cohen AD, Avogadri F, et al. (2009) OX40 engagement and chemotherapy combination provides potent antitumor immunity with concomitant regulatory T cell apoptosis. J Exp Med 206(5): 1103-1116.
  49. Yokouchi H, Yamazaki K, Chamoto K, Kikuchi E, Shinagawa N, et al. (2008) Anti-OX40 monoclonal antibody therapy in combination with radiotherapy results in therapeutic antitumor immunity to murine lung cancer. Cancer Sci 99(2): 361-367.
  50. Redmond WL, Linch SN, Kasiewicz MJ (2014) Combined targeting of co-stimulatory (OX40) and co-inhibitory (CTLA-4) pathways elicits potent effector T cells capable of driving robust antitumor immunity. Cancer Immunol Res 2(2): 142-153.
  51. Guo Z, Wang X, Cheng D, Xia Z, Luan M, et al. (2014) PD-1 blockade and OX40 triggering synergistically protects against tumor growth in a murine model of ovarian cancer. PLoS One 9(2): e89350.
  52. Suntharalingam G, Perry MR, Ward S, Brett SJ, Castello-Cortes A, et al. (2006) Cytokine storm in a phase 1 trial of the anti-CD28 monoclonal antibody TGN1412. N Engl J Med 355(10): 1018-1028.
  53. Walunas TL, Lenschow DJ, Bakker CY, Linsley PS, Freeman GJ, et al. (1994) CTLA-4 can function as a negative regulator of T cell activation. Immunity 1(5): 405-413.
  54. Salama, AK, Hodi FS (2011) Cytotoxic T-lymphocyte-associated antigen-4. Clin Cancer Res 17(14): 4622-4628.
  55. Yang ZZ, Novak AJ, Stenson MJ, Witzig TE, Ansell SM (2006) Intratumoral CD4+CD25+ regulatory T-cell-mediated suppression of infiltrating CD4+ T cells in B-cell non-Hodgkin lymphoma. Blood 107(9): 3639-3646.
  56. Grosso JF, Jure-Kunkel MN (2013) CTLA-4 blockade in tumor models: an overview of preclinical and translational research. Cancer Immun 13: 5.
  57. Tchekmedyian S, Glasby JA, Korman A, Keler T, Deo Y, et al. (2002) MDX-010 (human anti-CTLA4): a Phase I trial in malignant melanoma. Proc Am Soc Clin Oncol 21: 223 (abstr 56).
  58. Phan GQ, Yang JC, Sherry RM, Hwu P, Topalian SL, et al. (2003) Cancer regression and autoimmunity induced by cytotoxic T lymphocyte-associated antigen 4 blockade in patients with metastatic melanoma. Proc Natl Acad Sci USA 100(14): 8372-8377.
  59. Eisen T, Trefzer U, Hamilton A, Hersey P, Millward M, et al. (2010) Results of a multicenter, randomized, double-blind phase 2/3 study of lenalidomide in the treatment of pretreated relapsed or refractory metastatic malignant melanoma. Cancer 116(1): 146-154.
  60. Hodi FS, O'Day SJ, McDermott DF, Weber RW, Sosman JA, et al. (2010) Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med 363(8): 711-723.
  61. O’Day SJ, Maio M, Chiarion-Sileni V, Gajewski TF, Pehamberger IN, et al. (2010) Efficacy and safety of ipilimumab monotherapy in patients with previously treated, advanced melanoma: a multicenter, single-arm phase II study. Ann Oncol 21(8): 1712-1717.
  62. Wolchok JD, Neyns B, Linette G, Negrier S, Lutzky J, et al. (2010) Ipilimumab monotherapy in patients with pretreated advanced melanoma: a randomized, double-blind, multicentre, phase 2, dose-ranging study. Lancet Oncol 11(2): 155-164.
  63. Robert C, Thomas L, Bondarenko I, O'Day S, Weber J, et al. (2011) Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. N Engl J Med 364(26): 2517-2526.
  64. Eggermont AM, Chiarion-Sileni V, Grob JJ, Dummer R, Wolchok JD, et al. (2014) Ipilimumab versus placebo after complete resection of stage III melanoma: initial efficacy and safety results from the EORTC 18071 phase III trial. J Clin Oncol 32(18 suppl): abstr LBA9008.
  65. Yang JC, Hughes M, Kammula U, Royal R, Sherry RM, et al. (2007) Ipilimumab (anti-CTLA4 antibody) causes regression of metastatic renal cell cancer associated with enteritis and hypophysitis. J Immunother 30(8): 825-830.
  66. Small EJ, Tchekmedyian NS, Rini Bl, Fong L, Lowy I, et al. (2007) A pilot trial of CTLA-4 blockade with human anti-CTLA-4 in patients with hormone-refractory prostate cancer. Clin Cancer Res 13(6): 1810-1815.
  67. Slovin SF, Higano CS, Hamid O, Tejwani A, Harzstark, A, et al. (2013) Ipilimumab alone and in combination with radiotherapy in metastatic castration-resistant prostate cancer: results from an open-label, multicenter phase I/II study. Ann Oncol 24(7): 1813-1821.
  68. Kwon ED, Drake CG, Scher HI, Fizazi K, Bossi A, et al. (2014) Ipilimumab versus placebo after radiotherapy in patients with metastatic castration-resistant prostate cancer that had progressed after docetaxel chemotherapy (CA184-043): a multicentre, randomised, double-blind, phase 3 trial. Lancet Oncol 15(7): 700-712.
  69. Royal RE, Levy C, Turner K, Mathur A, Hughes M, et al. (2010) Phase 2 trial of single agent ipilimumab (anti-CTLA-4) for locally advanced or metastatic pancreatic adenocarcinoma. J Immunother 33(8): 828-833.
  70. Lynch TJ, Bondarenko I, Luft A, Serwatowski P, Barlesi F, et al. (2012) Phase II trial of ipilimumab (IPI) and paclitaxel/carboplatin (P/C) in first-line Stage IIIb/IV non-small cell lung cancer (NSCLC): results from randomized, double bind, multicenter phase II study. J Clin Oncol 30(17): 2046-2054.
  71. Chung KY, Gore I, Fong L, Venook A, Beck SB, et al. (2010) Phase II study of the anti-cytotoxic T-lymohocyte associated antigen 4 monoclonal antibody, tremelimumab, in patients with refractory metastatic colorectal cancer. J Clin Oncol 28(21): 3485-3490.
  72. Yuan J, Adamow M, Ginsberg BA, Rasalan TS, Ritter E, et al. (2011) Integrated NY-ESO-1 antibody and CD8+ T-cell responses correlate with clinical benefit in advanced melanoma patients treated with ipilimumab. Proc Natl Acad Sci USA 108(40): 16723-16728.
  73. Goff SL, Robbins PF, El-Gamil M, Rosenberg SA (2009) No correlation between clinical response to CTLA-4 blockade and presence of NY-ESO-1 antibody in patients with metastatic melanoma. J Immunother 32(8): 884-885.
  74. Attia P, Phan GQ, Maker AV, Robinson MR, Quezado MM (2005) Autoimmunity correlates with tumor regression in patients with metastatic melanoma treated with anti-cytotoxic T-lymphocyte antigen-4. J Clin Oncol 23(5): 6043-6053.
  75. Downey SG, Klapper JA, Smith FO, Yang JC, Sherry RM, et al. (2007) Prognostic factors related to clinical response in patients with metastatic melanoma treated by CTL-associated antigen-4 blockade. Clin Cancer Res 13(22 Pt 1): 6681-6688.
  76. Keir ME, Butte MJ, Freeman GJ, Sharpe AH (2008) PD-1 and its ligands in tolerance and immunity. Annu Rev Immunol 26: 677-704.
  77. Taube JM, Anders RA, Young GD, Xu H, Sharma R, et al. (2012) Colocalization of inflammatory response with B7-h1 expression in human melanocytic lesions supports an adaptive resistance mechanism of immune escape. Sci Transl Med 4(127): 127ra37.
  78. Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, et al. (2012) Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med 366(26): 2443-2454.
  79. Robert C, Long GV, Brady B, Dutriaux C, Maio M, et al. (2015) Nivolumab in previously untreated melanoma without BRAF mutation. N Eng J Med 372(4): 320-330.
  80. Brahmer JR, Drake CG, Wollner I, Powderly JD, Picus J, et al. (2010) Phase I study of single-agent anti-programmed death-1 (MDX-1106) in refractory solid tumors: safety, clinical activity, pharmacodynamics, and immunologic correlates. J Clin Oncol 28(19): 3167-3175.
  81. Berger R, Rotem-Yehudar R, Slama G, Landes S, Kneller A, et al. (2008) Phase I Safety and Pharmacokinetic Study of CT-011, a Humanized Antibody Interacting with PD-1, in Patients with Advanced Hematologic Malignancies. Clin Cancer Res 14(10): 3044-3051.
  82. Ribas A, Hodi FS, Kefford R, Hamid O, Daud A, et al. (2014) Efficacy and safety of the anti-PD-1 monoclonal antibody MK-3475 in 411 patients (pts) with melanoma (MEL). J Clin Oncol 32(18 suppl): abstr LBA9000.
  83. Patnaik A, Kang SP, Rasco D, Papadopoulos KP, Elassaiss-Schaap J, et al. (2012) Phase I study of MK-3475 (anti-PD-1 monoclonal antibody) in patients with advanced solid tumors. Clin Cancer Res 21(19): 4286-4293.
  84. Ribas A, Puzanov I, Dummer R, Schadendrof D, Hamid O, et al. (2015) Pembrolizumab versus investigator-choice chemotherapy for ipilimumab-refractory melanoma (KEYNOTE-002): a randomised, controlled, phase 2 trial. Lancet Oncol 16(8): 908-918.
  85. Gibney GT, Kudchadkar RR, DeConti RC, Thebeau MS, Czupryn MP, et al. (2015) Safety, correlative markers, and clinical results of adjuvant nivolumab in combination with vaccine in resected high-risk metastatic melanoma. Clin Cancer Res 21(4): 712-720.
  86. Yamazaki T, Akiba H, Iwai H, Matsuda H, Aoki M, et al. (2002) Expression of programmed death 1 ligands by murine T cells and APC. J Immunol 169(10): 5538-5545.
  87. Nakanishi J, Wada Y, Matsumoto K, Azuma M, Kikuchi K, et al. (2007) Overexpression of B7-H1 (PD-L1) significantly associates with tumor grade and postoperative prognosis in human urothelial cancers. Cancer Immunol 56(8): 1173-1182.
  88. Hino R, Kabashima K, Kato Y, Yagi H, Nakamura M, et al. (2010) Tumor cell expression of programmed cell death-1 ligand 1 is a prognostic factor for malignant melanoma. Cancer 116(7): 1757-1766.
  89. Zhang Y, Huang S, Gong D, Qin Y, Shen Q (2010) Programmed death-1 upregulation is correlated with dysfunction of tumor-infiltrating CD8+ T lymphocytes in human non-small cell lung cancer. Cell Mol Immunol 7(5): 389-395.
  90. Xylinas E, Robinson BD, Kluth LA, Volkmer BG, Hautmann R, et al. (2014) Association of T-cell co-regulatory protein expression with clinical outcomes following radical cystectomy for urothelial carcinoma of the bladder. Eur J Surg Oncol 40(1): 121-127.
  91. Fourcade J, Kudela P, Sun Z, Shen H, Land SR, et al. (2009) PD-1 is a regulator of NY-ESO-1-specific CD8+ T cell expansion in melanoma patients. J Immunol 182(9): 5240-5249.
  92. Matsuzaki J, Gnjatic S, Mhawech-Fauceglia P, Beck A, Miller A, et al. (2010) Tumor-infiltrating NY-ESO-1-specific CD8+ T cells are negatively regulated by LAG-3 and PD-1 in human ovarian cancer. Proc Natl Acad Sci USA 107(17): 7875-7880.
  93. Inman BA, Sebo TJ, Frigola X, Dong H, Bergstralh EJ, et al. (2007) PD-L1 (B7-H1) expression by urothelial carcinoma of the bladder and BCG-induced granulomata: associations with localized stage progression. Cancer 109(8): 1499-1505.
  94. Brahmer JR, Tykodi SS, Chow LQ, Hwu WJ, Topalian SL, et al. (2012) Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med 366(26): 2455-2465.
  95. Bellmunt J, Petrylak DP, Powles T, Braiteh F, Vogelzang N, et al. (2014) Inhibition of PD-L1 by MPDL3280A leads to clinical activity in pts with metastatic urothelial bladder cancer (UBC). Ann Oncol 25(Suppl 4): iv280-iv304.
  96. Taube JM, Klein A, Brahmer JR, Xu H, Pan X, et al. (2014) Association of PD-1, PD-1 ligands, and other features of the tumor immune microenvironment with response to anti-PD-1 therapy. Clin Cancer Res 20(19): 5064-5074.
  97. Cho DC, Sosman JA, Sznol M, Gordon MS, Hollebecque A, et al. (2013) Clinical activity, safety and biomarkers of MPDL3280A, an engineered PD-L1 antibody in patients with metastatic renal cell carcinoma (RCC). J Clin Oncol 31(15 suppl): abstr 4505.
  98. Powles T, Eder JP, Fine GD, Braiteh FS, Loriot Y, et al. (2014) MPDL3280A (anti-PD-L1) treatment leads to clinical activity in metastatic bladder cancer. Nature 515(7528): 558-562.
  99. Segal NH, Hamid O, Hwu W, Massard M, Butler M, et al. (2014) A phase I multi-arm dose-expansion study of the anti-programmed cell death-ligand-1 (PD-L1) antibody MEDI473: Preliminary data. Ann Oncol 25(Suppl 4): iv361-iv372.
  100. Spigel DR, Chaft JE, Gettinger SN, Chao BH, Dirix LY, et al. (2015) Clinical activity and safety from a phase II study (FIR) of MPDL3280A (anti-PDL1) in PD-L1-selected patients with non-small cell lung cancer (NSCLC). J Clin Oncol 33: 8028.
  101. Le DT, Uram JN, Wang H, Bartlett BR, Kemberling H, et al. (2015) PD-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med 372(26): 2509-2520.
  102. Ansell SM, Lesokhin AM, Borrello I, Halwani A, Scott EC, et al. (2015) PD-1 blockade with nivolumab in relapsed or refractory Hodgkin’s lymphoma. N Engl J Med 372(4): 311-319.
  103. Topalian SL, Sznol M, McDermott DF, Kluger HM, Carvajal RD, et al. (2014) Survival, durable tumor remission, and long-term safety in patients with advanced melanoma receiving nivolumab. J Clin Oncol 32(10): 1020-1030.
  104. Hamid O, Robert C, Daud A, Hodi FS, Hwu WJ, et al. (2013) Safety and tumor responses with lambrolizumab (anti–PD-1) in melanoma. N Engl J Med 369(2): 134-144.
  105. Herbst RS, Soria JC, Kowanetz M, Fine GD, Hamid O, et al. (2014) Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature 515(7528): 563-567.
  106. Sutmuller RP, van Duivenvoorde LM, van Elsas A, Schumacher TN, Wildenberg ME, et al. (2001) Synergism of cytotoxic T lymphocyte-associated antigen 4 blockade and depletion of CD25(+) regulatory T cells in antitumor therapy reveals alternative pathways for suppression of auto reactive cytotoxic T lymphocyte responses. J Exp Med 194(6): 823-832.
  107. Nagai H, Horikawa T, Hara I, Fukunaga A, Oniki S, et al. (2004) In vivo elimination of CD25+ regulatory T cells leads to tumor rejection of B16F10 melanoma, when combined with interleukin-12 gene transfer. Exp Dermatol 13(10): 613-620  .
  108. Yi H, Zhen Y, Jiang L, Zheng J, Zhao Y (2006) The phenotypic characterization of naturally occurring regulatory CD4+CD25+ T cells. Cell Mol Immunol 3(3): 189-195.
  109. Viehl CT, Moore TT, Liyanage UK, Frey DM, Ehlers JP, et al. (2006) Depletion of CD4+CD25+ regulatory T cells promotes a tumor-specific immune response in pancreas cancer-bearing mice. Ann Surg Oncol 13(9): 1252-1258.
  110. Maes W, Rosas GG, Verbinnen B, Boon L, De Vleeschouwer S, et al. (2009) DC vaccination with anti-CD25 treatment leads to long-term immunity against experimental glioma. Neuro Oncol 11(5): 529-542.
  111. Tan C, Reddy V, Dannull J, Ding E, Nair SK, et al. (2013) Impact of anti-CD25 monoclonal antibody on dendritic cell-tumor fusion vaccine efficacy in a murine melanoma model. J Transl Med 11: 148.
Creative Commons Attribution License

©2015 Farashi-Bonab, et al. This is an open access article distributed under the terms of the, which permits unrestricted use, distribution, and build upon your work non-commercially.