Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 Apr 1.
Published in final edited form as: Urol Oncol. 2016 Feb 28;34(4):171–181. doi: 10.1016/j.urolonc.2016.01.006

Blocking Immune Checkpoints in Prostate, Kidney and Urothelial cancer: An Overview

Angela KB Alme 1, Beerinder S Karir 3, Bishoy M Faltas 3, Charles G Drake 1,2
PMCID: PMC4834698  NIHMSID: NIHMS763968  PMID: 26923782

Abstract

Despite a long history of immunotherapeutic approaches to treatment, most genitourinary malignancies are not cured by existing immunotherapy regimens. More recently, cell-surface molecules known as immune checkpoints have become the focus of efforts to develop more effective immunotherapies. Interactions between these molecules and their ligands inhibit the proliferation and function of tumor-specific lymphocytes. A monoclonal antibody blocking one of these checkpoints was approved for the treatment of metastatic melanoma and is now being tested in other malignancies. The objective responses seen in these early trials of checkpoint blockade are driving renewed enthusiasm for cancer immunotherapy. There are several ongoing and planned trials in genitourinary malignancies of single-agent inhibitors, as well as combinations targeting multiple checkpoints or adding other types of therapies to checkpoint blockade.

Keywords: CTLA-4, PD-1, Prostate Cancer, Kidney Cancer, Bladder Cancer, PD-L1, LAG-3

I: Introduction

Genitourinary malignancies have a long history of immunotherapeutic approaches to treatment including high-dose interleukin-2 and interferon alpha for renal cell carcinoma[1], bacillus Calmette-Guerin (BCG) for bladder cancer[2]and, most recently, Sipuleucel-T for prostate cancer[3].While effective in many patients, these therapies are, in general, not curative. The development of more effective cancer immunotherapy has long been hampered by the multiple strategies that tumors use to evade destruction by the host immune system[4]. One such strategy involves the expression of cell-surface molecules, known as immune checkpoints, on tumor-specific lymphocytes[5-7]. The interactions between these molecules and their ligands inhibit the proliferation and function of cells with potentially important anti-tumor effect. The FDA approval of a monoclonal antibody that blocks the immune checkpoint CTLA-4 (Ipilimumab (BMS, Princeton, NJ)) for metastatic melanoma in 2011 marked a turning point for immunotherapy – especially for immune checkpoint blockade[8]. Here we review the various checkpoint inhibitors that are in the clinic and their particular importance in genitourinary (GU) malignancies.

II: CTLA-4 - A Prototypical Immune Checkpoint

The FDA approval in 2011 of Ipilimumab (Yervoy) for advanced melanoma opened a new chapter in the almost 20 year long story of CTLA-4 (Cytotoxic T Lymphocyte Antigen-4). Structurally homologous to the co-stimulatory molecule CD28, CTLA-4exerts its inhibitory role by binding to the same ligands (B7.1 and B7.2) CD28 does[9], though with a markedly higher affinity and avidity. This results in an effective “hijacking” of signal 2.While CD28 is found on the surface of naïve and activated T cells, CTLA-4 is only detectable after activation[10]. The therapeutic effects of CTLA-4 blockade seem to be due primarily to enhancing the effector function of T cells[11], though more recent data suggest that anti-CTLA-4 antibodies may function by depleting regulatory T cell (Treg) as well[12, 13]. The first reports of CTLA-4 blockade enhancing anti-tumor immunity in mice appeared in 1996 when the Allison group demonstrated enhanced rejection of established tumors after anti-CTLA-4 antibody treatment[14]. The broad and vital role of CTLA-4 in modulating the activation of T cells is underscored by the autoimmunity and early death seen in CTLA-4-deficient mice[15]and the significant rate of immune-related adverse events (IRAEs) seen in patients treated with CTLA-4 blockade[16].

IIA. Preclinical Studies of CTLA-4 Blockade in Prostate Cancer

The first studies showing that blockade of CTLA-4 could enhance anti-tumor immunity in a murine prostate cancer model used the TRAMP-C1 implantable tumor line. Growth of established sub-cutaneous tumors was significantly delayed, with some tumors regressing entirely[17]. Using an implanted model involving TRAMP-C2 cells, Kwon et al further demonstrated that administration of CTLA-4 blockade immediately following tumor resection (i.e. in the adjuvant setting) reduced metastatic spread to nearby lymph nodes from 97.4% to 44%[18]. Animal studies were extended to include combination treatment regimens – experiments in the TRAMP model of primary, autochronous prostate cancer tested the combination of CTLA-4 blockade along with an irradiated GM-CSF expressing whole tumor cell vaccine (similar to GVAX prostate)[19]. Assessment at 2 months post-treatment revealed a significant reduction in tumor incidence, lower tumor grade and increased accumulation of inflammatory cells when compared to the control monotherapy groups[20]. Our lab employed the antigen-bearing ProHA × TRAMP mouse to interrogate the optimal relative timing of CTLA-4 blockade and GM-CSF-secreting cell-based vaccine (GVAX) using antigen-specific adoptively transferred CD8 T cells to measure anti-tumor response. In these studies, maximum benefit was seen with anti-CTLA-4 antibody administered one day post-vaccination[21]. The addition of low-dose cyclophosphamide to the combination regimen further enhanced the anti-tumor response by abrogating immune tolerance, augmenting prostatic CD8+ T-cell infiltration and mediating depletion of regulatory T cells (Tregs)[22]. Waitz et al observed an additive effect when combining anti-CTLA-4 antibody treatment with cryoablation of primary implanted TRAMP-C2 tumors to prevent the growth of distantly-implanted secondary TRAMP-C2 tumors. Taken together, these preclinical studies provided reasonable justification for clinical trials of CTLA-4 blockade in men with prostate cancer.

Additional translational data supporting that notion came from a small microarray study showing that CTLA-4 was up-regulated in CD4+ Tregs sorted from patient prostate infiltrating lymphocytes (PIL) vs. naïve CD4 T cells from peripheral blood[23]. Perhaps more intriguing was a recent report suggesting a role for CTLA-4+ cells on patient CD8+ T cells with regulatory function[24]. In that study, patients with biochemically recurrent prostate cancer (BCR) were immunized with a PAP DNA/GM-CSF vaccine. Analysis of PBMCs from the vaccinated patients revealed a population of antigen-specific CD8+CTLA-4+ Tregs that suppressed specific T cell responses via an IL-35 dependent mechanism; i.e.anti-CTLA-4 antibody added to in vitro PAP stimulation unmasked PAP-specific effector responses that had been inhibited by this CD8 Treg population, and in vitro CTLA-4 blockade also allowed the identification of pre-existing PAP-specific CD8+ Tregs in a portion of the patients. Taken together these two studies suggest a role for CTLA-4 in inhibiting prostate cancer specific immune responses in patients.

IIB. CTLA-4 Blockade in Prostate Cancer - Monotherapy

The first reported pilot trial of anti-CTLA-4 antibody in prostate cancer patients tested a single 3 mg/kg dose in fourteen patients with advanced mCRPC. While treatment at this dose was well-tolerated, only two patients demonstrated PSA declines of ≤50% before eventually progressing [25]. In a larger study,Slovin et al tested Ipilimumab alone or in combination with radiotherapy in 71 patients with mCRPC[26]. Of the fifty patients who received the highest dose of Ipilimumab (10 mg/kg) alone or in concert with radiotherapy, eight experienced PSA reduction of ≤50%, six had stable disease and one patient had an ongoing complete response. Across all groups, 80% of patients experienced IRAEs with grade 3/4 IRAEs reported in 32%. Fourteen patients (28%) in the 10 mg/kg cohorts discontinued treatment due to AEs. Until recently, this was the largest experience of Ipilimumab monotherapy in prostate cancer, and set the stage for two randomized Phase III trials, launched in 2009-2010. The first of these, CA184-043 (NCT00861614) randomized approximately 800 men with mCPRC who had progressed on chemotherapy to either placebo or to Ipilimumab at a dose of 10 mg/kg q 3 weeks × 4 doses, followed by q 3 month maintenance for non-progressing patients[27]. Based on preclinical data showing that treating animals with implanted tumors with radiation therapy plus anti-CTLA-4 was more effective than either treatment alone[28], this trial also included a low dose (8 Gy) of radiation therapy to at least one lesion in both groups. It should be noted that these men, in general, had multiple sites of disease, so this radiation treatment would not be expected to significantly reduce a tolerogenic tumor burden. Instead, the notion here was that antigen “liberation” might serve to prime an immune response which would then be boosted by anti-CTLA-4 treatment. As reported, the trial missed its primary endpoint of overall survival (O.S.) with treatment arm showing median OS of 11.2 months vs. 10 months in control arm (hazard ratio [HR] = 0.85, 95% confidence interval [CI] = 0.72–1.00, P = .0530). The secondary endpoint of progression free survival (PFS) was met, with a PFS of 4.0 months in the Ipilimumab arm as compared to 3.1 months in the placebo group. Pre-planned and exploratory subgroup analyses showed that patients with an alkaline phosphatase of < 1.5 times the upper limit of normal and a hemoglobin > 11 mg/dL might derive benefit. Perhaps most interestingly, analyses for interaction showed that the presence of visceral metastases strongly interacted with a treatment effect in that Ipilimumab appeared to have no effect on O.S. in patients with visceral metastases[29]. This surprising finding suggests that visceral metastases in prostate cancer might be immunologically different than bone lesions, and has profound implications for future immunotherapy trials in prostate cancer. It is worth noting that the pivotal trial of the prostate cancer vaccine Sipuleucel-T excluded patients with visceral metastases[30]; in retrospect this was likely a wise decision. Recently updated O.S. data of this trial was found to be consistent with the initial analysis, demonstrating larger benefits in patients with lower disease burden and especially when patients did not have visceral metastasis. Median OS was reported to be 11.2 months (9.6–12.6) in the ipilimumab arm vs. 10.0 months (8.4–11.2) in control arm (HR 0.84, p=0.03) [31]. A second large randomized Phase III trial of Ipilimumab in prostate cancer has completed accrual. This trial (CA184-095, NCT01057810) randomized 600 men who had not yet received chemotherapy to either Ipilimumab or placebo. This trial did not include “priming” radiotherapy, but did exclude men with visceral disease. Disappointingly, initial reports suggest that this trial is negative; results are expected to be released in 2016.

IIC. CTLA-4 Blockade in Prostate Cancer – Combination Regimens

Several combination trials involving anti-CTLA-4 have been completed thus far (Table 1). In one of these, Fong et al [32] treated 24 mCRPC patients with increasing doses of Ipilimumab plus a fixed dose of GM-CSF. Immunologically, the role of GM-CSF in this strategy is likely to be the activation and potentially the expansion of antigen presenting cells (APC) although an opposing inhibitory role cannot be excluded. Of the six patients receiving the highest Ipilimumab dose (3 mg/kg), 3 had confirmed PSA declines of >50%. One of these three responding patients also exhibited a partial response in visceral metastases. Correlative studies showed that patients treated with the two highest doses of Ipilimumab had increased levels of activated CD8+ T cells - above those previously seen with GM-CSF alone. These data are interesting; patients in previous trials who received CTLA-4 blockade alone demonstrated no reported increases in activated CD8 T cells. Additional combination trials have paired prostate cancer vaccines with Ipilimumab. In the first of these, patients were treated with an allogeneic GM-CSF secreting cell based prostate cancer vaccine (GVAX Prostate), along with escalating doses of Ipilimumab[33]. Of the seven patients (25%) who demonstrated PSA declines of ≤50%, all had received the two highest doses of Ipilimumab (3 or 5 mg/kg). Overall the combination was well tolerated, and patients appeared to have a longer than predicted overall survival (O.S.), although the trial was not powered to draw firm conclusions. In a second relevant trial, Ipilimumab was co-administered with the pox-virus based anti-PSA vaccine known as ProstVac VF[34]. In this Phase I dose escalation trial, 30 patients were treated with a fixed dose of a pox-viral PSA vaccine (PSA-Tricom) and one of four different Ipilimumab doses[35]. Among the 24 chemotherapy naïve subjects, 14 had PSA declines from baseline but only 6 were >50%. Of the nine HLA-A2 patients, six demonstrated antigen-specific T cell responses via ELISpot. Taken together these trials show that combinations of Ipilimumab and other immunologically active agents can be well-tolerated, and suggest the potential for possible additive efficacy. Based on data from several groups showing that androgen-ablation increases T cell infiltration into the prostate gland[36, 37], and may mitigate tolerance[38], there are at least four ongoing trials combining Ipilimumab with androgen ablation (Table 1). In one of these trials (NCT01194271), the M.D. Anderson group is treating men with high-risk disease for 4 months with the combination of hormonal therapy and Ipilimumab, followed by radical prostatectomy. Tissue gathered at surgery will be critical in determining the tissue-level effects of combined treatment in the clinical setting.

Table 1.

Kidney cancer
Agent Phase Trial Status Design/Description Number Of Subjects Results/Comments Reference or NCCT Identifier
Nivolumab Monotherapy III Completed Randomized, open- label study of anti-PD-1 versus the mTOR inhibitor everolimus in patients with advanced / metastatic RCC. Patients have received prior anti-angiogenic therapy. 821 Primary endpoint = OS 25 Vs.19.6 months Motzer RJ et al NEJM 2015
MPDL3280A (anti-PD-L1) +/- Avastinvs. Sunitinib III Ongoing Randomized, open- label study comparing MPDL3280A + avastin to sunitinib alone in patients with untreated locally advanced or metastatic RCC 550 Primary endpoint = PFS, Secondary Endpoints = Overall RR/duration, OS, DOR, Adverse Event Incidence & pharmacokinetics NCT02420821
Nivolumab + Ipilimumab III Ongoing Randomized open label study of nivolumab + ipilimumab compared with sunitinib monotherapy in previously untreated advanced or mRCC 1070 Primary Outcome Measures: Progression-free survival (PFS), Overall survival (OS)
Secondary Outcome Measures: Objective response rate (ORR) & Adverse Event Incidence
NCT02231749
Prostate cancer
Agent Phase Trial Status/PC Design/Description Number of Subjects Results/Comments Reference or NCCT Identifier
Ipilimumab Monotherapy III Completed Randomized, double blind trial of ipilimumabvs. placebo in pts with chemotherapy-naïve mCRPC 600 Primary endpoint- OS
Secondary endpoints = PFS, pain progression, time to subsequent therapy and safety.
Results- not yet reported
NCT01057810
Ipilimumab Following radiotherapy III Completed Randomized, double blind trial of ipilimumabvs placebo following radiotherapy in patients previously treated with docetaxel 799 Primary endpoint = OS
Secondary endpoints = PFS, pain response and safety
OS- 11.2 vs 10 months (p=0.05)
Kwon ED et al Lancet Onc. 2014
Bladder cancer
Agent Phase Trial Status Design/Description Number Of Subjects Results/Comments Reference or NCCT Identifier
Pembrolizumab (anti-PD-1) monotherapy III Ongoing Randomized open label stud of Pembrolizumab (MK-3475) Versus Paclitaxel, Docetaxel or Vinflunine in Recurrent or Progressive Metastatic UC patients 470 Primary Outcome Measures: Overall survival (OS), Progression-free survival (PFS) per RECIST 1.1 NCT02256436
Atezolizumab (MPDL3280A) III Ongoing Open-Label study in Patients With PD-L1-Selected, High-Risk Muscle Invasive UC patients after Cystectomy Randomized to Atezolizumab Vs. Observation as Adjuvant Therapy 440 Primary Outcome: Disease-Free Survival (DFS)
Secondary Outcome: Overall Survival (OS), Disease-Specific Survival (DSS), Distant Metastasis-Free Survival (DMFS), Adverse Events (AEs), Percentage of Anti-Therapeutic Antibody (ATA) Response
NCT02450331
Atezolizumab (MPDL3280A) III Ongoing Open label randomized study of Atezolizumab vs. chemotherapy in metastatic UC patients who have failed platinum based chemotherapy 767 Primary Outcome: Overall survival Secondary Outcome: Objective response rate (ORR), Progression-free survival (PFS), Duration of response (DOR), Incidence of adverse events (AEs), Incidence of anti-therapeutic antibodies to Atezolizumab NCT02302807

IID. Blocking CTLA-4 in Kidney Cancer

A 2007 study found an association between CTLA-4 polymorphisms and the risk of developing RCC, as well as an association between a particular SNP and tumor grade in RCC patients[39]. Unfortunately, this finding did not translate to broad success in treating RCC with anti-CTLA-4[40]. A phase II trial conducted primarily at the National Cancer Institute (NCI) treated 61 patients with 3 mg/kg doses of Ipilimumab every 3 weeks, or with a single 3 mg/kg “loading dose” followed by 1 mg/kg doses every 3 weeks. In this trial, sequential cohorts were assessed [41]. Partial responses were observed in 5/40 (13%) patients receiving the higher dose. Grade 3 or 4 IRAEs were observed in 33% of patients, potentially a higher rate than that observed in melanoma patients, and likely reflecting the q 3 week dosing regimen used in this trial. At the current time, single-agent CTLA-4 blockade is not in clinical trials in RCC, most likely due to competition from the relative plethora of targeted agents, and other immunotherapy agents such as anti-PD-1. A phase I trial investigating CTLA-4 blockade in combination with anti-PD1 is ongoing and preliminary results are discussed below (section IIIB)

IIE. Blocking CTLA-4 in Bladder Cancer

To date, the only reported trial of Ipilimumab in bladder cancer is a relatively small pre-operative trial with the primary endpoints of safety/tolerability and immune monitoring[42]. In this study, 12 patients received 2 doses of Ipilimumab (either 10 mg/kg or 3 mg/kg) at 7 weeks and 4 before radical cystectomy. While the majority of IRAE's were Grade 1/2 and did not delay surgery, in the higher dose cohort, one patient did not receive the second Ipilimumab dose due to Grade 3 diarrhea and 2 patients had their surgery delayed due to IRAEs. This small trial led to the somewhat unexpected finding of a significantly higher number of ICOShigh CD4 T-cells, both circulating and in the tumor tissue, and an increased ratio of ICOS+ to FoxP3+ CD4 T-cells in treated patients. ICOS (Inducible COStimulator) is related to CD28 and CTLA-4 and plays a role in immune cell responses and proliferation. These ICOShigh CD4 T-cells produced IFN-γ upon stimulation, suggesting that they have the potential to be involved in an anti-tumor immune response. This important clinical observation has also led to interesting preclinical studies suggesting the importance of ICOS as a checkpoint, as well as a potential biomarker for the efficacy of anti-CTLA-4. In further trials, an ongoing multi-center Phase II trial(NCT01524991) is currently recruiting patients with advanced/metastatic urothelial carcinoma for treatment with a regimen combining Gemcitabine/Cisplatin with Ipilimumab (10 mg/kg every 3 weeks). The primary endpoint is one-year overall survival with progression-free survival, overall response rate and safety/AEs as secondary outcome measures. Taken together these data suggest that bladder cancer is an intriguing target for immune checkpoint blockade. Additional trials blocking PD-L1 and PD-1 in bladder cancer are ongoing as well(Table I).

III: The Immune Checkpoint PD-1 (Programmed Death–1)

In contrast to the early, widespread and lethal autoimmunity seen in mice lacking CTLA-4, PD-1 knockout mice exhibit late-onset, milder, strain-specific autoimmunity that is generally limited in scope[43, 44]. Together with its expression on the “exhausted” CD8 cells seen in chronic infections[45], these data speak to a role for PD-1 in modulating T cell responses to prevent autoimmunity and restrain inflammatory responses in the face of persistent antigen. First described by the Honjo group in 1992[46], PD-1 can be found on T cells, B cells, natural killer T cells, dendritic cells and activated monocytes[6], and is well-described on tumor-infiltrating lymphocytes in numerous human cancers[47-51]. T cell expression of PD-1 occurs upon activation via the TCR[52]. Given the role of PD-1 in limiting inflammatory responses, it is no surprise that its ligands, PD-L1/B7-H1 and PD-L2/B7-DC, are up-regulated on multiple cell types by pro-inflammatory cytokines[5]. PD-L1 is widely expressed in numerous human carcinomas, including lung, ovary, colon, melanoma, kidney and bladder and has been shown to correlate with progression and poor prognosis for some malignancies[53, 54]. The IFNγ-induced expression of PD-L1 on tumors is a mechanism of “adaptive resistance” in response to the immune infiltrate, as opposed to an oncogene-driven, constitutive means of escape. In melanoma, immunohistochemical(IHC) examination of 150 benign and cancerous lesions revealed a highly significant association of B7-H1 expression with inflammatory infiltrates (P <0.0001)[55], suggesting that here the expression of PD-L1 is primarily adaptive. In terms of genitourinary cancers, expression of PD-L1 is common in kidney cancer and bladder cancer, but exceedingly rare in prostate cancer. In terms of translation, multiple preclinical studies using murine tumor models demonstrated success with antibody-mediated blockade of the PD-1/PD-L1 interaction[56, 57]. In 2010 the results of the first pilot study of anti-PD-1[58]were reported, followed by Phase Ib multi-dose results for PD-1[59]and anti-PD-L1 in 2012[60].In comparison with anti-CTLA-4, grade 3 or 4 IRAEs were less frequent, and with the exception of a small number of serious pneumonitis cases, proved manageable. In general, 15-30% of patients with kidney cancer, melanoma and lung cancer showed objective responses to these agents, providing important clinical proof of concept for monotherapy.

IIIA. Blocking PD-1 in Prostate Cancer

Despite evidence from at least two groups showing that the CD8 T cells that infiltrate prostate tumors express PD-1[47, 61], no objective responses to single-agent PD-1 blockade were reported in 17 patients with prostate cancer treated on the Phase Ib multi-dose trial of anti-PD-1[59]. The precise mechanisms underlying this lack of response are not immediately obvious, but may involve the phenotype of prostate infiltrating lymphocytes, which are generally refractory to stimulation [62]. Another possible explanation involves the expression of PD-L1, which is generally associated with an increased response to anti-PD-1 monotherapy[63], and which is generally absent in prostate cancer. Indeed, the relative paucity of PD-L1 expression in human prostate tumors is somewhat puzzling, given pre-clinical data suggesting that loss of PTEN appears to be associated with PD-L1 up-regulation in both prostate cancer[64] and glioblastoma[65], and the notion that between 10 and 70% of prostate tumors lose PTEN[66]. Regardless of the precise mechanism, clinical data thus far suggest that PD-1 blockade is not as likely to be as effective as monotherapy for prostate cancer as it is for kidney cancer, lung cancer or melanoma.

IIIB. Blocking PD-1 in Kidney Cancer

Given the presence of PD-1+ mononuclear cells and PD-L1+ tumor cells in RCC patients, it was not especially surprising that phase I dose-escalation trials of anti-PD-1 monotherapy yielded objective responses in RCC[58, 67]. Perhaps more impressive, however, is the case of an advanced RCC patient who demonstrated a stable partial response that, over time, evolved into a documented complete response. In 2013, Lipson et al reported that the patient has remained off-treatment for over 5 years[68]. Longer-term follow-up data from a phase Ib study of nivolumab showed that the objective response rate to anti-PD-1 monotherapy in RCC patients was in the 30-35% range with prolonged stable disease in another 10% of patients [69]. Similar long term follow-up study of MPDL3280A in RCC patients showed durable median response of 54 weeks (2.7+ to 68.1+ weeks) and positive association between PD-L1 intensity and response to MPDL3280A [70]. Based on these long term efficacy activities and safe tolerability profile, further studies of anti PD-1 agents especially Nivolumab in RCC were undertaken and have been successfully completed (Table 1).

Phase II study of nivolumab in previously treated RCC patients involved three randomized cohorts treated every three weeks at doses of 0.3, 2 or 10 mg/kg respectively. Median PFS was reported as 2.7, 4.0, and 4.2 months respectively in three cohorts (P = 0.9) with median OS of 18.2 months (80% CI, 16.2 to 24.0 months), 25.5 months (80% CI, 19.8 to 28.8 months), and 24.7 months (80% CI, 15.3 to 26.0 months) across 3 cohorts. 19/168 (11%) of patients experienced grade3/4 study drug related toxicities [71]. In a Phase I “biomarker” trial of nivolumab, immunomodulatory activity of this drug was assessed using pre- and post-treatment biopsies as well as peripheral blood samples. Study included 3 cohorts that received nivolumab every three weeks at doses of 0.3, 2 or 10 mg/kg respectively. An additional cohort in this study enrolled treatment-naïve RCC patients. PD-L1+ patients demonstrated better ORR of 22% (4/18) as compared to 8% (3/38) seen in PD-L1– patients. During the course of treatment from baseline to cycle 2 day 8, T cell infiltrates increased by a median of 70% (CD3+; range 53– 220%) and 88% (CD8+; 61–257%). Such transition in biomarkers along the study drug treatment course proved immunomodulatory effects of nivolumab [72].

Nivolumab and MPDL3280A have also been investigated in combination with FDA approved drugs. A Phase I dose escalation study(NCT01472081) of Nivolumab (anti-PD-1 agent) in combination with tyrosine kinase inhibitors sunitinib or pazopanib in mRCC patients has been reported. No dose limiting toxicity was observed in sunitinib arm at starting dose of nivolumab (2 mg/kg IV Q3W) leading to expansion of higher dose arm (5mg/kg IV Q3W). But DLT's were observed in pazopinib arm at starting dose of nivolumab leading to closure of the arm. Overall both combinations demonstrated safe toxicity profiles along with anti-tumor activity [73]. Another phase I study of nivolumab in combination with ipilimumab in mRCC patients has demonstrated acceptable safety with ongoing anti-tumor responses. 16% patients in the study experienced dose limiting AEs that most commonly included increased lipase or ALT. Objective response rate (ORR) was reported in 29% pts. (nivolumab 3 mg/kg + ipilimumab 1 mg/kg cohort) and 39% patients (nivolumab 1 mg/kg + ipilimumab 3 mg/kg cohort) [74]. MPDL3280Ahas also been investigated along with bevacizumab in mRCC patients with good tolerability of the treatment combination [75].

Based on promising phase II study results, phase III randomized trial comparing Nivolumab to the mTOR inhibitor everolimus (1:1 randomization) in RCC patients was undertaken. While overall survival was the study's primary endpoint, numerous secondary outcomes such as safety, progression-free survival, objective responses and disease-related symptom progression were also measured. A total of 821 RCC patients (clear cell histology) who had previously received one or two lines of anti-angiogenic regimens were accrued. After a minimum follow-up period of 14 months, median OS was reported at 25.0 months (95% confidence interval [CI], 21.8 to not estimable) in the nivolumab arm vs. 19.6 months (95% CI, 17.6 to 23.1) in the everolimus arm. The objective response rate was 25% with nivolumab and 5% with everolimus (odds ratio 5.98; 95% CI, 3.68 to 9.72; P<0.001). The median progression-free survival was found to be 4.6 months in the nivolumab group and 4.4 months in the everolimus group (hazard ratio, 0.88; 95% CI, 0.75 to 1.03; P=0.11) [76]. These results eventually led to FDA approval of nivolumab for advanced RCC patients.

IV: Other Checkpoint Molecules – LAG-3, TIM-3,B7-H3 and B7-H4/B7-Hx

IVA. Blocking LAG-3

LAG-3 is a cell surface molecule found on several different types of immune cells that is structurally quite similar to CD4[77]. Both molecules bind to Class II MHC but LAG-3's affinity for Class II is likely higher than that of CD4[78]. Though LAG-3 is up-regulated upon activation of either CD4 or CD8 T-cells[79], it serves as a negative regulator of homeostatic proliferation[80]and can be found on CD8+ TILs as well as CD4+ induced Treg[50, 51, 81]. Analysis of tumor-infiltrating lymphocytes (TILs) from both primary and metastatic RCC tumors showed significant expression of LAG-3 on CD8 T cells. Expression, particularly in concert with other checkpoint molecules, has been observed in melanoma as well[51]. An interesting synergy between PD-1 and LAG-3 blockade has been shown in murine tumor models[82]and ovarian cancer patients[50]. Both studies demonstrated that combined antibody blockade of PD-1 and LAG-3 was more effective than either alone and may be a promising future checkpoint blockade strategy for some malignancies[82, 73].One LAG-3 related molecule currently in clinical trials is a recombinant, soluble, dimeric LAG-3-Ig fusion protein known as IMP321 or ImmuFact (Immutep, Orsay, France) that is intended to condition dendritic cells without inducing inflammation. There have been several Phase I trials of IMP321 in various cancers, including a non-randomized, fixed dose-escalation Phase I study of 21 patients with advanced or metastatic RCC (NCT00351949)[84]. The only adverse events attributed to IMP321 in this trial were grade 1 local reactions at the injection sites. Increases in activated CD8+ T cells were seen at the two highest dosages but there were no objective responses. Progression-free survival was significantly better in the higher doses vs. the lower doses with 7/8 (87.5%) high dose patients experiencing stable disease at 3 months vs. 3/11 (27%) in lower dose cohort (P=0.015). At this time, there is only one IMP321 study recruiting patients – a multi-peptide vaccine for melanoma with IMP321 as adjuvant. A LAG-3 blocking monoclonal antibody, BMS-986016, (BMS, Princeton NJ) has very recently entered a Phase I dose escalation trial, both as a monotherapy and in combination with PD-1 blockade (NCT01968109).

IVB. TIM-3 as an Immune Checkpoint

TIM-3 (T-cell immunoglobulin mucin-3) was identified in 2002 by the Kuchroo lab as part of an effort to find a reliable marker for TH1 CD4 T-cells[85]. Originally thought to be expressed only on differentiated TH1 and Tc1 cells as a means of restraining TH1 responses, later studies demonstrated expression on innate immune cells including dendritic cells[86]. Murine studies found that the majority of CD8 TILs expressing TIM-3 also expressed PD-1, and that these double-expressors were characterized by the most severe inhibition of effector cytokine secretion (IL-2, IFN-γ and TNF-α). Few studies have addressed the expression of TIM-3 on human TIL; in one relevant study it was shown that advanced melanoma patients exhibited co-expression of TIM-3 and PD-1 on tumor infiltrating, antigen-specific CD8 T cells[87]. Combined in vitro blockade with both TIM-3 and PD-1 was able to restore effector cytokine secretion. A recent study of benign and cancerous tissue from 137 treatment-naïve prostate cancer patients found weak TIM-3 expression in benign tissue compared to up-regulation in both PIN and invasive carcinomas[88]. Univariate analysis showed a significant association with TNM stage, nuclear grade and recurrence-free or progression-free survival. In RCC patients, percentages of TIM-3+ and TIM-3+PD-1+ CD8 T cells and CD4 T cells were significantly higher in tumor-infiltrate than in peripheral blood[89]. Despite ample preclinical interest, at this time, there are no ongoing clinical trials of TIM-3 blockade in cancer patients.

IVC. B7-H3

First described by the Chen lab in 2001[90], B7-H3 is a member of the B7 superfamily with inducible cell surface expression on T cells, monocytes and dendritic cells as well as low-level constitutive expression on many non-immune cells and tissues. It was originally characterized as a co-stimulatory molecule that, when combined with anti-CD3, could induce IFNγ production in T cells. Later work suggestedthat ligation of B7-H3 could mediate suppression of TH1 responses[91]. Interestingly, B7-H3 may have a dual role in the immune response to cancer, in some conditions up-regulating a response and in others down-modulating immune responsiveness[92]. Multiple studies of prostate cancer patient samples showed strong immunohistochemical staining of B7-H3 on adenocarcinomas and high-grade PIN as well as some cell lines[93, 94]. Of 823 prostatectomy samples, 93% showed high expression which was correlated with metastases at time of surgery as well as a significantly higher risk of recurrence and death attributable to prostate cancer. More intense staining of B7-H3 in prostate cancer was also associated with greater risk of biochemical recurrence after salvage radiation therapy[95],staining of the proliferation marker Ki-67[96] and lower numbers of intratumoralCD4 T cells, CD8 T cells and DCs[97]. Interestingly, B7-H3 expression was not affected by androgen deprivation before radical prostatectomy[98]. In 743 RCC patients, only 17% had tumoral B7-H3 expression while 95% were positive for expression in the tumor vasculature[99]. Both tumor expression and diffuse vascular expression were associated with greater risk of disease progression and death due to RCC. In UCC of the bladder, B7-H3 was found to be widely expressed (>70% of samples) across all tumor stages, though BCG recipients tended to have increased expression[100]. Clinically, Loo and colleagues developed an Fc-enhanced monoclonal antibody that targets B7-H3 expressing tumors via ADCC[101]. This antibody exhibited potent anti-tumor activity in both in vitro and xenograft studies with no adverse events seen in primate studies. At the current time, a B7-H3 targeted antibody (MGA271, MacroGenics, Frederick MD), is being clinically evaluated in a Phase I trial in patients with melanoma or prostate cancer (NCT01391143).

IVD. B7-H4/B7x/B7S1

B7-H4, also known as B7x/B7S1, is a member of the B7 super family first described in 2003 by three independent research groups and is considered to be an inhibitor of proliferation and cytokine production in CD4 and CD8 T-cells[102-105]. Primarily expressed on activated T cells, B cells, dendritic cells and monocytes, surface expression is low in most non-lymphoid tissues, though somewhat higher in prostate, testis and a small number of other sites. B7-H4 expression has been described in numerous malignancies including prostate and renal cancers. In both prostate cancer and RCC, more robust expression is associated with a higher risk of death, metastatic disease and recurrence[94, 106]. B7-H4 expression is not confined to the tumor cells themselves – in one study 211 of 259 RCC patient specimens (81.5%) were positive for tumor vasculature endothelium expression via IHC[105]. In preclinical studies, a recombinant human antibody delayed tumor growth in an ovarian cancer model involving humanized mice with established sub-cutaneous tumors[107].vAt this time, there are no ongoing clinical trials targeting B7-H4.

V: Conclusions and Future Directions

The objective responses seen with anti-PD-1 and anti-CTLA-4 antibodies are driving renewed enthusiasm for cancer immunotherapy. While single-agent CTLA-4 blockade shows efficacy in multiple tumor types, the high rates of serious IRAEs cannot be overlooked. The results of ongoing Phase III trials will clarify how Ipilimumab can best be used going forward, especially in prostate cancer where a randomized Phase III trial in the pre-chemotherapy space has accrued and is maturing. PD-1 blockade monotherapy, in contrast, results in durable responses in multiple tumor types. With a potentially lower incidence of serious IRAEs, Nivolimumab and other PD-1 / PD-L1 targeting agents like MK-3475 (Merck) and MPDL3280A (Roche / Genentech) have great potential, though simultaneous inhibition of more than one checkpoint may be likely to be more effective than targeting a single pathway[108]. Checkpoint blockade in combination with other therapies, such as vaccines, androgen ablation, targeted therapies and radiation, has proved effective in murine models. With several ongoing or planned trials to explore these approaches, the field is eager to see the same efficacy in patients.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Figlin RA, Abi-Aad AS, Belldegrun A, deKernion JB. The role of interferon and interleukin-2 in the immunotherapeutic approach to renal cell carcinoma. Semin Oncol. 1991;18:102–7. [PubMed] [Google Scholar]
  • 2.Herr HW, Morales A. History of bacillus Calmette-Guerin and bladder cancer: an immunotherapy success story. J Urol. 2008;179:53–6. doi: 10.1016/j.juro.2007.08.122. [DOI] [PubMed] [Google Scholar]
  • 3.Frohlich MW. Sipuleucel-T for the treatment of advanced prostate cancer. Semin Oncol. 2012;39:245–52. doi: 10.1053/j.seminoncol.2012.02.004. [DOI] [PubMed] [Google Scholar]
  • 4.Drake CG, Jaffee E, Pardoll DM. Mechanisms of immune evasion by tumors. Adv Immunol. 2006;90:51–81. doi: 10.1016/S0065-2776(06)90002-9. [DOI] [PubMed] [Google Scholar]
  • 5.Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012;12:252–64. doi: 10.1038/nrc3239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Keir ME, Butte MJ, Freeman GJ, Sharpe AH. PD-1 and its ligands in tolerance and immunity. Annu Rev Immunol. 2008;26:677–704. doi: 10.1146/annurev.immunol.26.021607.090331. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Chen L. Co-inhibitory molecules of the B7-CD28 family in the control of T-cell immunity. Nat Rev Immunol. 2004;4:336–47. doi: 10.1038/nri1349. [DOI] [PubMed] [Google Scholar]
  • 8.Lipson EJ, Drake CG. Ipilimumab: an anti-CTLA-4 antibody for metastatic melanoma. Clin Cancer Res. 2011;17:6958–62. doi: 10.1158/1078-0432.CCR-11-1595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Linsley PS, Ledbetter JA. The role of the CD28 receptor during T cell responses to antigen. Annu Rev Immunol. 1993;11:191–212. doi: 10.1146/annurev.iy.11.040193.001203. [DOI] [PubMed] [Google Scholar]
  • 10.Walunas TL, Lenschow DJ, Bakker CY, et al. CTLA-4 can function as a negative regulator of T cell activation. Immunity. 1994;1:405–13. doi: 10.1016/1074-7613(94)90071-x. [DOI] [PubMed] [Google Scholar]
  • 11.Peggs KS, Quezada SA, Chambers CA, Korman AJ, Allison JP. Blockade of CTLA-4 on both effector and regulatory T cell compartments contributes to the antitumor activity of anti-CTLA-4 antibodies. J Exp Med. 2009;206:1717–25. doi: 10.1084/jem.20082492. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Selby MJ, Engelhardt JJ, Quigley M, et al. Anti-CTLA-4 Antibodies of IgG2a Isotype Enhance Antitumor Activity through Reduction of Intratumoral Regulatory T Cells. Cancer Immunology Research. 2013;1:32–42. doi: 10.1158/2326-6066.CIR-13-0013. [DOI] [PubMed] [Google Scholar]
  • 13.Simpson TR, Li F, Montalvo-Ortiz W, et al. Fc-dependent depletion of tumor-infiltrating regulatory T cells co-defines the efficacy of anti-CTLA-4 therapy against melanoma. J Exp Med. 2013;210:1695–710. doi: 10.1084/jem.20130579. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Leach DR, Krummel MF, Allison JP. Enhancement of antitumor immunity by CTLA-4 blockade. Science. 1996;271:1734–6. doi: 10.1126/science.271.5256.1734. [DOI] [PubMed] [Google Scholar]
  • 15.Tivol EA, Borriello F, Schweitzer AN, Lynch WP, Bluestone JA, Sharpe AH. Loss of CTLA-4 leads to massive lymphoproliferation and fatal multiorgan tissue destruction, revealing a critical negative regulatory role of CTLA-4. Immunity. 1995;3:541–7. doi: 10.1016/1074-7613(95)90125-6. [DOI] [PubMed] [Google Scholar]
  • 16.Weber J. Ipilimumab: controversies in its development, utility and autoimmune adverse events. Cancer Immunol Immunother. 2009;58:823–30. doi: 10.1007/s00262-008-0653-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Kwon ED, Hurwitz AA, Foster BA, et al. Manipulation of T cell costimulatory and inhibitory signals for immunotherapy of prostate cancer. Proc Natl Acad Sci U S A. 1997;94:8099–103. doi: 10.1073/pnas.94.15.8099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Kwon ED, Foster BA, Hurwitz AA, et al. Elimination of residual metastatic prostate cancer after surgery and adjunctive cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) blockade immunotherapy. Proc Natl Acad Sci U S A. 1999;96:15074–9. doi: 10.1073/pnas.96.26.15074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Simons JW, Sacks N. Granulocyte-macrophage colony-stimulating factor-transduced allogeneic cancer cellular immunotherapy: the GVAX vaccine for prostate cancer. Urol Oncol. 2006;24:419–24. doi: 10.1016/j.urolonc.2005.08.021. [DOI] [PubMed] [Google Scholar]
  • 20.Hurwitz AA, Foster BA, Kwon ED, et al. Combination immunotherapy of primary prostate cancer in a transgenic mouse model using CTLA-4 blockade. Cancer Res. 2000;60:2444–8. [PubMed] [Google Scholar]
  • 21.Wada S, Jackson CM, Yoshimura K, et al. Sequencing CTLA-4 blockade with cell-based immunotherapy for prostate cancer. J Transl Med. 2013;11:89. doi: 10.1186/1479-5876-11-89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Wada S, Yoshimura K, Hipkiss EL, et al. Cyclophosphamide augments antitumor immunity: studies in an autochthonous prostate cancer model. Cancer Res. 2009;69:4309–18. doi: 10.1158/0008-5472.CAN-08-4102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Sfanos KS, Bruno TC, Maris CH, et al. Phenotypic Analysis of Prostate-Infiltrating Lymphocytes Reveals TH17 and Treg Skewing. Clin Cancer Res. 2008;14:3254–61. doi: 10.1158/1078-0432.CCR-07-5164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Olson BM, Jankowska-Gan E, Becker JT, Vignali DA, Burlingham WJ, McNeel DG. Human prostate tumor antigen-specific CD8+ regulatory T cells are inhibited by CTLA-4 or IL-35 blockade. J Immunol. 2012;189:5590–601. doi: 10.4049/jimmunol.1201744. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Small EJ, Tchekmedyian NS, Rini BI, Fong L, Lowy I, Allison JP. A pilot trial of CTLA-4 blockade with human anti-CTLA-4 in patients with hormone-refractory prostate cancer. Clin Cancer Res. 2007;13:1810–5. doi: 10.1158/1078-0432.CCR-06-2318. [DOI] [PubMed] [Google Scholar]
  • 26.Slovin SF, Higano CS, Hamid O, et al. Ipilimumab alone or in combination with radiotherapy in metastatic castration-resistant prostate cancer: results from an open-label, multicenter phase I/II study. Ann Oncol. 2013 doi: 10.1093/annonc/mdt107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Gerritsen WR, Kwon ED, Fizazi K, et al. CA184-043: A randomized, multicenter, double-blind phase 3 trial comparing overall survival (OS) in patients (pts) with post-docetaxel castration-resistant prostate cancer (CRPC) and bone metastases treated with ipilimumab (ipi) vs placebo (pbo), each following single-dose radiotherapy (RT) European Journal of Cancer. 2013;49:S678–S679. [Google Scholar]
  • 28.Demaria S, Kawashima N, Yang AM, et al. Immune-mediated inhibition of metastases after treatment with local radiation and CTLA-4 blockade in a mouse model of breast cancer. Clin Cancer Res. 2005;11:728–34. [PubMed] [Google Scholar]
  • 29.Drake CG, Kwon ED, Fizazi K, et al. Results of subset analyses on overall survival (OS) from study CA184-043: Ipilimumab (Ipi) versus placebo (Pbo) in post-docetaxel metastatic castration-resistant prostate cancer (mCRPC) ASCO Meeting Abstracts. 2014;32:2. [Google Scholar]
  • 30.Kantoff PW, Higano CS, Shore ND, et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med. 2010;363:411–22. doi: 10.1056/NEJMoa1001294. [DOI] [PubMed] [Google Scholar]
  • 31.Fizazi K, Drake C, Kwon E, et al. 763PD - Updated overall survival (OS) from the phase 3 trial, CA184-043: Ipilimumab (Ipi) vs placebo (Pbo) in patients with post-docetaxel metastatic castration resistant prostate cancer mCRPC. Annals of Oncology. 2014;25(suppl_4):iv255–iv279. 10.1093/annonc/mdu336. [Google Scholar]
  • 32.Fong L, Kwek SS, O'Brien S, et al. Potentiating endogenous antitumor immunity to prostate cancer through combination immunotherapy with CTLA4 blockade and GM-CSF. Cancer Res. 2009;69:609–15. doi: 10.1158/0008-5472.CAN-08-3529. [DOI] [PubMed] [Google Scholar]
  • 33.van den Eertwegh AJ, Versluis J, van den Berg HP, et al. Combined immunotherapy with granulocyte-macrophage colony-stimulating factor-transduced allogeneic prostate cancer cells and ipilimumab in patients with metastatic castration-resistant prostate cancer: a phase 1 dose-escalation trial. Lancet Oncol. 2012;13:509–17. doi: 10.1016/S1470-2045(12)70007-4. [DOI] [PubMed] [Google Scholar]
  • 34.Arlen PM, Kaufman HL, DiPaola RS. Pox viral vaccine approaches. Semin Oncol. 2005;32:549–55. doi: 10.1053/j.seminoncol.2005.09.006. [DOI] [PubMed] [Google Scholar]
  • 35.Madan RA, Mohebtash M, Arlen PM, et al. Ipilimumab and a poxviral vaccine targeting prostate-specific antigen in metastatic castration-resistant prostate cancer: a phase 1 dose-escalation trial. Lancet Oncol. 2012;13:501–8. doi: 10.1016/S1470-2045(12)70006-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Mercader M, Bodner BK, Moser MT, et al. T cell infiltration of the prostate induced by androgen withdrawal in patients with prostate cancer. Proc Natl Acad Sci U S A. 2001;98:14565–70. doi: 10.1073/pnas.251140998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Gannon PO, Poisson AO, Delvoye N, Lapointe R, Mes-Masson AM, Saad F. Characterization of the intra-prostatic immune cell infiltration in androgen-deprived prostate cancer patients. J Immunol Methods. 2009;348:9–17. doi: 10.1016/j.jim.2009.06.004. [DOI] [PubMed] [Google Scholar]
  • 38.Drake CG, Doody AD, Mihalyo MA, et al. Androgen ablation mitigates tolerance to a prostate/prostate cancer-restricted antigen. Cancer Cell. 2005;7:239–49. doi: 10.1016/j.ccr.2005.01.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Cozar JM, Romero JM, Aptsiauri N, et al. High incidence of CTLA-4 AA (CT60) polymorphism in renal cell cancer. Hum Immunol. 2007;68:698–704. doi: 10.1016/j.humimm.2007.05.002. [DOI] [PubMed] [Google Scholar]
  • 40.Drake CG, Lipson EJ, Brahmer JR. Breathing new life into immunotherapy: review of melanoma, lung and kidney cancer. Nat Rev Clin Oncol. 2014;11:24–37. doi: 10.1038/nrclinonc.2013.208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Yang JC, Hughes M, Kammula U, et al. Ipilimumab (anti-CTLA4 antibody) causes regression of metastatic renal cell cancer associated with enteritis and hypophysitis. J Immunother. 2007;30:825–30. doi: 10.1097/CJI.0b013e318156e47e. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Carthon BC, Wolchok JD, Yuan JD, et al. Preoperative CTLA-4 Blockade: Tolerability and Immune Monitoring in the Setting of a Presurgical Clinical Trial. Clinical Cancer Research. 2010;16:2861–71. doi: 10.1158/1078-0432.CCR-10-0569. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Nishimura H, Nose M, Hiai H, Minato N, Honjo T. Development of lupus-like autoimmune diseases by disruption of the PD-1 gene encoding an ITIM motif-carrying immunoreceptor. Immunity. 1999;11:141–51. doi: 10.1016/s1074-7613(00)80089-8. [DOI] [PubMed] [Google Scholar]
  • 44.Okazaki T, Tanaka Y, Nishio R, et al. Autoantibodies against cardiac troponin I are responsible for dilated cardiomyopathy in PD-1-deficient mice. Nat Med. 2003;9:1477–83. doi: 10.1038/nm955. [DOI] [PubMed] [Google Scholar]
  • 45.Barber DL, Wherry EJ, Masopust D, et al. Restoring function in exhausted CD8 T cells during chronic viral infection. Nature. 2006;439:682–7. doi: 10.1038/nature04444. [DOI] [PubMed] [Google Scholar]
  • 46.Ishida Y, Agata Y, Shibahara K, Honjo T. Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. EMBO J. 1992;11:3887–95. doi: 10.1002/j.1460-2075.1992.tb05481.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Sfanos KS, Bruno TC, Meeker AK, De Marzo AM, Isaacs WB, Drake CG. Human prostate-infiltrating CD8+ T lymphocytes are oligoclonal and PD-1+ Prostate. 2009;69:1694–703. doi: 10.1002/pros.21020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Ahmadzadeh M, Johnson LA, Heemskerk B, et al. Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired. Blood. 2009;114:1537–44. doi: 10.1182/blood-2008-12-195792. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Thompson RH, Dong H, Lohse CM, et al. PD-1 is expressed by tumor-infiltrating immune cells and is associated with poor outcome for patients with renal cell carcinoma. Clin Cancer Res. 2007;13:1757–61. doi: 10.1158/1078-0432.CCR-06-2599. [DOI] [PubMed] [Google Scholar]
  • 50.Matsuzaki J, Gnjatic S, Mhawech-Fauceglia P, et al. 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 U S A. 2010;107:7875–80. doi: 10.1073/pnas.1003345107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Baitsch L, Legat A, Barba L, et al. Extended co-expression of inhibitory receptors by human CD8 T-cells depending on differentiation, antigen-specificity and anatomical localization. PLoS ONE. 2012;7:e30852. doi: 10.1371/journal.pone.0030852. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Agata Y, Kawasaki A, Nishimura H, et al. Expression of the PD-1 antigen on the surface of stimulated mouse T and B lymphocytes. Int Immunol. 1996;8:765–72. doi: 10.1093/intimm/8.5.765. [DOI] [PubMed] [Google Scholar]
  • 53.Dong H, Strome SE, Salomao DR, et al. Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat Med. 2002;8:793–800. doi: 10.1038/nm730. [DOI] [PubMed] [Google Scholar]
  • 54.Thompson RH, Gillett MD, Cheville JC, et al. Costimulatory B7-H1 in renal cell carcinoma patients: Indicator of tumor aggressiveness and potential therapeutic target. Proc Natl Acad Sci U S A. 2004;101:17174–9. doi: 10.1073/pnas.0406351101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Taube JM, Anders RA, Young GD, et al. Colocalization of inflammatory response with b7-h1 expression in human melanocytic lesions supports an adaptive resistance mechanism of immune escape. Sci Transl Med. 2012;4:127ra37. doi: 10.1126/scitranslmed.3003689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Dong H, Strome SE, Salomao DR, et al. Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat Med. 2002;8:793–800. doi: 10.1038/nm730. [DOI] [PubMed] [Google Scholar]
  • 57.Iwai Y, Ishida M, Tanaka Y, Okazaki T, Honjo T, Minato N. 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. 2002;99:12293–7. doi: 10.1073/pnas.192461099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Brahmer JR, Drake CG, Wollner I, et al. 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. 2010;28:3167–75. doi: 10.1200/JCO.2009.26.7609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Topalian SL, Hodi FS, Brahmer JR, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 2012;366:2443–54. doi: 10.1056/NEJMoa1200690. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Brahmer JR, Tykodi SS, Chow LQ, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med. 2012;366:2455–65. doi: 10.1056/NEJMoa1200694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Ebelt K, Babaryka G, Frankenberger B, et al. Prostate cancer lesions are surrounded by FOXP3+, PD-1+ and B7-H1+ lymphocyte clusters. Eur J Cancer. 2009;45:1664–72. doi: 10.1016/j.ejca.2009.02.015. [DOI] [PubMed] [Google Scholar]
  • 62.Bronte V, Kasic T, Gri G, et al. Boosting antitumor responses of T lymphocytes infiltrating human prostate cancers. J Exp Med. 2005;201:1257–68. doi: 10.1084/jem.20042028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Grosso J, Horak CE, Inzunza D, et al. Association of tumor PD-L1 expression and immune biomarkers with clinical activity in patients (pts) with advanced solid tumors treated with nivolumab (anti-PD-1; BMS-936558; ONO-4538) ASCO Meeting Abstracts. 2013;31:3016. [Google Scholar]
  • 64.Crane CA, Panner A, Murray JC, et al. PI(3) kinase is associated with a mechanism of immunoresistance in breast and prostate cancer. Oncogene. 2009;28:306–12. doi: 10.1038/onc.2008.384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Parsa AT, Waldron JS, Panner A, et al. Loss of tumor suppressor PTEN function increases B7-H1 expression and immunoresistance in glioma. Nat Med. 2007;13:84–8. doi: 10.1038/nm1517. [DOI] [PubMed] [Google Scholar]
  • 66.McMenamin ME, Soung P, Perera S, Kaplan I, Loda M, Sellers WR. Loss of PTEN expression in paraffin-embedded primary prostate cancer correlates with high Gleason score and advanced stage. Cancer Res. 1999;59:4291–6. [PubMed] [Google Scholar]
  • 67.Cho DC, Sosman JA, Sznol M, et al. Clinical activity, safety, and biomarkers of MPDL3280A, an engineered PD-L1 antibody in patients with metastatic renal cell carcinoma (mRCC) ASCO Meeting Abstracts. 2013;31:4505. [Google Scholar]
  • 68.Lipson EJ, Sharfman WH, Drake CG, et al. Durable Cancer Regression Off-Treatment and Effective Reinduction Therapy with an Anti-PD-1 Antibody. Clin Cancer Res. 2013;19:462–8. doi: 10.1158/1078-0432.CCR-12-2625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Drake CG, McDermott DF, Sznol M, et al. Survival, safety, and response duration results of nivolumab (Anti-PD-1; BMS-936558; ONO-4538) in a phase I trial in patients with previously treated metastatic renal cell carcinoma (mRCC): Long-term patient follow-up. ASCO Meeting Abstracts. 2013;31:4514. [Google Scholar]
  • 70.McDermott DF, Sznol M, Sosman JA, et al. Immune correlates and long term follow up of a phase Ia study of MPDL3280A, an engineered PD-L1 antibody, in patients with metastatic renal cell carcinoma mRCC. Annals of Oncology. 2014;25(suppl_4):iv280–iv304. 10.1093/annonc/mdu337. [Google Scholar]
  • 71.Motzer RJ, Rini BI, McDermott DF, et al. Nivolumab for Metastatic Renal Cell Carcinoma: Results of a Randomized Phase II Trial. J Clin Oncol. 2015;33(13):1430–7. doi: 10.1200/JCO.2014.59.0703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Choueiri TK, Fishman MN, Escudier BJ, et al. Immunomodulatory activity of nivolumab in previously treated and untreated metastatic renal cell carcinoma (mRCC): Biomarker-based results from a randomized clinical trial. ASCO 2014. J Clin Oncol. 2014;32(5s) suppl; abstr 5012. [Google Scholar]
  • 73.Amin A, Plimack ER, Infante JR, et al. Nivolumab (anti-PD-1; BMS-936558, ONO-4538) in combination with sunitinib or pazopanib in patients (pts) with metastatic renal cell carcinoma (mRCC). ASCO 2014. J Clin Oncol. 2014;32(5s) suppl; abstr 5010. [Google Scholar]
  • 74.Hammers HJ, Plimack ER, Infante JR, et al. Phase I study of nivolumab in combination with ipilimumab in metastatic renal cell carcinoma (mRCC). ASCO 2014. J Clin Oncol. 2014;32(5s) doi: 10.1200/JCO.2016.72.1985. suppl; abstr 4504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Lieu C, Bendell J, Powderly JD, et al. Safety and efficacy of MPDL3280A (anti-PDL1) in combination with bevacizumab (bev) and/or chemotherapy (chemo) in patients (pts) with locally advanced or metastatic solid tumors. ESMO 2014. Annals of Oncology. 2014;25(suppl_41):iv361–iv372. 10.1093/annonc/mdu342. [Google Scholar]
  • 76.Motzer RJ, Escudier B, McDermott DF, et al. Nivolumab versus Everolimus in Advanced Renal-Cell Carcinoma. N Engl J Med. 2015;373:1803–1813. doi: 10.1056/NEJMoa1510665. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Goldberg MV, Drake CG. LAG-3 in Cancer Immunotherapy. Curr Top Microbiol Immunol. 2011;344:269–78. doi: 10.1007/82_2010_114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Huard B, Prigent P, Tournier M, Bruniquel D, Triebel F. CD4/major histocompatibility complex class II interaction analyzed with CD4- and lymphocyte activation gene-3 (LAG-3)-Ig fusion proteins. Eur J Immunol. 1995;25:2718–21. doi: 10.1002/eji.1830250949. [DOI] [PubMed] [Google Scholar]
  • 79.Huard B, Tournier M, Hercend T, Triebel F, Faure F. Lymphocyte-activation gene 3/major histocompatibility complex class II interaction modulates the antigenic response of CD4+ T lymphocytes. Eur J Immunol. 1994;24:3216–21. doi: 10.1002/eji.1830241246. [DOI] [PubMed] [Google Scholar]
  • 80.Workman CJ, Vignali DA. Negative regulation of T cell homeostasis by lymphocyte activation gene-3 (CD223) J Immunol. 2005;174:688–95. doi: 10.4049/jimmunol.174.2.688. [DOI] [PubMed] [Google Scholar]
  • 81.Huang CT, Workman CJ, Flies D, et al. Role of LAG-3 in regulatory T cells. Immunity. 2004;21:503–13. doi: 10.1016/j.immuni.2004.08.010. [DOI] [PubMed] [Google Scholar]
  • 82.Woo SR, Turnis ME, Goldberg MV, et al. Immune inhibitory molecules LAG-3 and PD-1 synergistically regulate T-cell function to promote tumoral immune escape. Cancer Res. 2012;72:917–27. doi: 10.1158/0008-5472.CAN-11-1620. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Nirschl CJ, Drake CG. Molecular Pathways: Co-Expression of Immune Checkpoint Molecules: Signaling Pathways and Implications for Cancer Immunotherapy. Clin Cancer Res. 2013 doi: 10.1158/1078-0432.CCR-12-1972. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Brignone C, Escudier B, Grygar C, Marcu M, Triebel F. A phase I pharmacokinetic and biological correlative study of IMP321, a novel MHC class II agonist, in patients with advanced renal cell carcinoma. Clin Cancer Res. 2009;15:6225–31. doi: 10.1158/1078-0432.CCR-09-0068. [DOI] [PubMed] [Google Scholar]
  • 85.Monney L, Sabatos CA, Gaglia JL, et al. Th1-specific cell surface protein Tim-3 regulates macrophage activation and severity of an autoimmune disease. Nature. 2002;415:536–41. doi: 10.1038/415536a. [DOI] [PubMed] [Google Scholar]
  • 86.Anderson AC, Anderson DE, Bregoli L, et al. Promotion of tissue inflammation by the immune receptor Tim-3 expressed on innate immune cells. Science. 2007;318:1141–3. doi: 10.1126/science.1148536. [DOI] [PubMed] [Google Scholar]
  • 87.Fourcade J, Sun Z, Benallaoua M, et al. Upregulation of Tim-3 and PD-1 expression is associated with tumor antigen-specific CD8+ T cell dysfunction in melanoma patients. J Exp Med. 2010;207:2175–86. doi: 10.1084/jem.20100637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Piao YR, Piao LZ, Zhu LH, Jin ZH, Dong XZ. Prognostic value of T cell immunoglobulin mucin-3 in prostate cancer. Asian Pac J Cancer Prev. 2013;14:3897–901. doi: 10.7314/apjcp.2013.14.6.3897. [DOI] [PubMed] [Google Scholar]
  • 89.Dannenmann SR, Thielicke J, Stockli M, et al. Tumor-associated macrophages subvert T-cell function and correlate with reduced survival in clear cell renal cell carcinoma. Oncoimmunology. 2013;2:e23562. doi: 10.4161/onci.23562. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Chapoval AI, Ni J, Lau JS, et al. B7-H3: a costimulatory molecule for T cell activation and IFN-gamma production. Nat Immunol. 2001;2:269–74. doi: 10.1038/85339. [DOI] [PubMed] [Google Scholar]
  • 91.Suh WK, Gajewska BU, Okada H, et al. The B7 family member B7-H3 preferentially down-regulates T helper type 1-mediated immune responses. Nat Immunol. 2003;4:899–906. doi: 10.1038/ni967. [DOI] [PubMed] [Google Scholar]
  • 92.Loos M, Hedderich DM, Friess H, Kleeff J. B7-h3 and its role in antitumor immunity. Clin Dev Immunol. 2010;2010:683875. doi: 10.1155/2010/683875. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Roth TJ, Sheinin Y, Lohse CM, et al. B7-H3 ligand expression by prostate cancer: a novel marker of prognosis and potential target for therapy. Cancer Res. 2007;67:7893–900. doi: 10.1158/0008-5472.CAN-07-1068. [DOI] [PubMed] [Google Scholar]
  • 94.Zang X, Thompson RH, Al-Ahmadie HA, et al. B7-H3 and B7× are highly expressed in human prostate cancer and associated with disease spread and poor outcome. Proc Natl Acad Sci U S A. 2007;104:19458–63. doi: 10.1073/pnas.0709802104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Parker AS, Heckman MG, Sheinin Y, et al. Evaluation of B7-H3 expression as a biomarker of biochemical recurrence after salvage radiation therapy for recurrent prostate cancer. Int J Radiat Oncol Biol Phys. 2011;79:1343–9. doi: 10.1016/j.ijrobp.2010.01.061. [DOI] [PubMed] [Google Scholar]
  • 96.Liu Y, Vlatkovic L, Saeter T, et al. Is the clinical malignant phenotype of prostate cancer a result of a highly proliferative immune-evasive B7-H3-expressing cell population? Int J Urol. 2012;19:749–56. doi: 10.1111/j.1442-2042.2012.03017.x. [DOI] [PubMed] [Google Scholar]
  • 97.Liu Y, Saeter T, Vlatkovic L, et al. Dendritic and lymphocytic cell infiltration in prostate carcinoma. Histol Histopathol. 2013;28:1621–8. doi: 10.14670/HH-28.1621. [DOI] [PubMed] [Google Scholar]
  • 98.Chavin G, Sheinin Y, Crispen PL, et al. Expression of immunosuppresive B7-H3 ligand by hormone-treated prostate cancer tumors and metastases. Clin Cancer Res. 2009;15:2174–80. doi: 10.1158/1078-0432.CCR-08-2262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Crispen PL, Sheinin Y, Roth TJ, et al. Tumor cell and tumor vasculature expression of B7-H3 predict survival in clear cell renal cell carcinoma. Clin Cancer Res. 2008;14:5150–7. doi: 10.1158/1078-0432.CCR-08-0536. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Boorjian SA, Sheinin Y, Crispen PL, et al. T-cell coregulatory molecule expression in urothelial cell carcinoma: clinicopathologic correlations and association with survival. Clin Cancer Res. 2008;14:4800–8. doi: 10.1158/1078-0432.CCR-08-0731. [DOI] [PubMed] [Google Scholar]
  • 101.Loo D, Alderson RF, Chen FZ, et al. Development of an Fc-enhanced anti-B7-H3 monoclonal antibody with potent antitumor activity. Clin Cancer Res. 2012;18:3834–45. doi: 10.1158/1078-0432.CCR-12-0715. [DOI] [PubMed] [Google Scholar]
  • 102.Zang X, Loke P, Kim J, Murphy K, Waitz R, Allison JP. B7×: a widely expressed B7 family member that inhibits T cell activation. Proc Natl Acad Sci U S A. 2003;100:10388–92. doi: 10.1073/pnas.1434299100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Prasad DV, Richards S, Mai XM, Dong C. B7S1, a novel B7 family member that negatively regulates T cell activation. Immunity. 2003;18:863–73. doi: 10.1016/s1074-7613(03)00147-x. [DOI] [PubMed] [Google Scholar]
  • 104.Sica GL, Choi IH, Zhu G, et al. B7-H4, a molecule of the B7 family, negatively regulates T cell immunity. Immunity. 2003;18:849–61. doi: 10.1016/s1074-7613(03)00152-3. [DOI] [PubMed] [Google Scholar]
  • 105.Krambeck AE, Thompson RH, Dong H, et al. B7-H4 expression in renal cell carcinoma and tumor vasculature: associations with cancer progression and survival. Proc Natl Acad Sci U S A. 2006;103:10391–6. doi: 10.1073/pnas.0600937103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Thompson RH, Zang X, Lohse CM, et al. Serum-soluble B7× is elevated in renal cell carcinoma patients and is associated with advanced stage. Cancer Res. 2008;68:6054–8. doi: 10.1158/0008-5472.CAN-08-0869. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Dangaj D, Lanitis E, Zhao A, et al. Novel recombinant human b7-h4 antibodies overcome tumoral immune escape to potentiate T-cell antitumor responses. Cancer Res. 2013;73:4820–9. doi: 10.1158/0008-5472.CAN-12-3457. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Wolchok JD, Kluger H, Callahan MK, et al. Nivolumab plus Ipilimumab in Advanced Melanoma. N Engl J Med. 2013 doi: 10.1056/NEJMoa1302369. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES