Skip to main content
Journal of Leukocyte Biology logoLink to Journal of Leukocyte Biology
. 2013 Jul;94(1):41–53. doi: 10.1189/jlb.1212631

At the Bedside: CTLA-4- and PD-1-blocking antibodies in cancer immunotherapy

Margaret K Callahan *,, Jedd D Wolchok *,†,‡,1
PMCID: PMC4051187  PMID: 23667165

Clinical Review for Basic Researchers: Treating patients with CTLA-4 and PD-1 pathway blocking antibodies, plus clinical progress and application of agents in earlier stages of development.

Keywords: tumor, overall survival, endpoints

Abstract

It is increasingly appreciated that cancers are recognized by the immune system, and under some circumstances, the immune system may control or even eliminate tumors. The modulation of signaling via coinhibitory or costimulatory receptors expressed on T cells has proven to be a potent way to amplify antitumor immune responses. This approach has been exploited successfully for the generation of a new class of anticancer therapies, “checkpoint-blocking” antibodies, exemplified by the recently FDA-approved agent, ipilimumab, an antibody that blocks the coinhibitory receptor CTLA-4. Capitalizing on the success of ipilimumab, agents that target a second coinhibitory receptor, PD-1, or its ligand, PD-L1, are in clinical development. Lessons learned from treating patients with CTLA-4 and PD-1 pathway-blocking antibodies will be reviewed, with a focus on concepts likely to inform the clinical development and application of agents in earlier stages of development. See related review At the bench: Preclinical rationale for CTLA-4 and PD-1 blockade as cancer immunotherapy.

Introduction

The development of an antigen-specific T cell response is a complex, highly regulated process. Early studies investigating T cell activation led to the “two-signal” model, wherein activation requires antigen-specific stimulation via the TCR (signal 1) and a costimulatory signal (signal 2)[14]. Subsequent studies have largely validated the two-signal model and added layers of complexity to this framework. It is now clear that a variety of immunomodulatory signals, both costimulatory and coinhibitory, are needed to orchestrate an antigen-specific immune response.

It is increasingly appreciated that cancers are recognized by the immune system, and under certain circumstances, the immune system may control or even eliminate tumors [5]. Studies in mouse models of transplantable tumors have demonstrated that manipulation of costimulatory or coinhibitory signals can amplify T cell responses against tumors [6]. This may be accomplished by blockade of coinhibitory molecules, such as CTLA-4, PD-1, and LAG-3, or by enhanced signaling of costimulatory molecules, such as GITR, OX40, and 4-1BB [719]. Based on robust preclinical activity in mouse models, antibodies targeting a variety of coinhibitory and costimulatory molecules are in clinical develop as anticancer agents.

Building on the basic science and preclinical studies reviewed in the companion article by Intlekofer and Thompson [20], the present review focuses on the clinical development of antibodies that block signaling via the inhibitory molecules CTLA-4 or PD-1, including agents that are already approved by the FDA (anti-CTLA-4) or in the earlier stages of clinical development (anti-PD-1, anti-PD-L1). These checkpoint-blocking antibodies have demonstrated clinical activity in a variety of tumor types, including melanoma, RCC, and NSCLC. As novel anticancer agents, they have a distinct profile of antitumor activity and toxicity, underscoring their unique mechanism of activity. Whereas CTLA-4 and PD-1 function as negative regulators, each plays a nonredundant role in modulating immune responses. CLTA-4, through engagement with its ligands CD80 and CD86, plays a pivotal role in attenuating the early activation of naïve and memory T cells. In contrast, PD-1 is primarily involved in modulating T cell activity in peripheral tissues via its interaction with PD-L1 and PD-L2 [21]. Differences in the biological role of each molecule are likely to explain unique, clinical features of agents that target each pathway. The understanding of tumor-specific and patient-specific characteristics that shape tumor-immune interaction may allow selection of checkpoint-blocking strategies tailored to individual patients. Moreover, as CTLA-4 and PD-1 regulate immune responses in a nonredundant fashion, combined blockade of both pathways may achieve superior antitumor activity. Lessons learned from treating patients with CTLA-4 and PD-1 pathway-blocking antibodies are likely to inform the clinical development of the next generation of antibodies targeting T cells via a diversity of coinhibitory and costimulatory molecules.

CTLA-4-BLOCKING ANTIBODIES

Based on the promising preclinical data generated in mouse models, two antibodies that block CTLA-4 in humans have been developed: ipilimumab (previously MDX-010; Medarex, Princeton, NY, USA, and Bristol-Myers Squibb, Princeton, NJ, USA) and tremelimumab (previously CP-675,206 or ticilimumab; Pfizer, New York, NY, USA, and now Medimmune, Gaithersburg, MD, USA; Table 1). Ipilimumab is a fully human IgG1 κ mAb, generated using mice transgenic for the human Ig heavy and light chains. Ipilimumab has high affinity for human and cynomolgus monkey CTLA-4 and not rodent CTLA-4 in vitro. Preclinical safety testing, performed in the cynomolgus monkey, showed a favorable profile. Ipilimumab has a half-life of 12–14 days. Tremelimumab, a fully human mAb belonging to the IgG2 antibody class, has a longer half-life of 22 days. It also has high affinity for human and cynomolgus monkey CTLA-4 in vitro.

Table 1. Checkpoint-Blocking Antibodies in Clinical Development.

Antibody Target Isotype Clinical development
Ipilimumab (Bristol-Myers Squibb) CTLA-4 IgG1k FDA-approved
Tremelimumab (Pfizer, now Medimmune) CTLA-4 IgG2 Completed Phase III
Nivolumab/BMS-936558/MDX1106 (Bristol-Myers Squibb) PD-1 IgG4 Phase I interim results, Phase III open
CT-011 (CureTech, Yavne, Israel) PD-1 IgG1 Completed Phase I
MK-3475 (Merck, Whitehouse Station, NJ, USA) PD-1 IgG4 Phase I ongoing, interim results
AMP-224a (Amplimmune, Gaithersburg, MD, USA) PD-1 NA Phase I ongoing
BMS-936559/MDX-1105 (Bristol-Myers Squibb) PD-L1 IgG4 Completed Phase I
MEDI4736 (MedImmune/AstraZeneca, London, UK) PD-L1 IgG1k Phase I ongoing
MPDL3280A/RG7446 (Genentech, South San Francisco, CA, USA/Roche, Basel, Switzerland) PD-L1 Phase I ongoing
a

Fusion protein.

CTLA-4-blocking antibodies in the clinic

  • Ipilimumab

  • —Fully human IgG1 κ mAb

  • —Half-life 12–14 days

  • Tremelimumab

  • —Fully human IgG2 mAb

  • —Half-life ∼22 days

Research Question: How does the isotype impact antitumor activity of checkpoint-blocking antibodies?

IPILIMUMAB: PATHWAY TO FDA APPROVAL

Ipilimumab (commercial name Yervoy) was approved on March 25, 2011, by the FDA for the treatment of unresectable or metastatic melanoma. The clinical testing of ipilimumab in patients began one decade earlier. Pilot studies of ipilimumab were first reported in 2002, with a Phase I study showing two PR in a cohort of 17 patients with unresectable melanoma treated with a single dose of ipilimumab, dosed at 3 mg/kg [22]. Treatment was well-tolerated, with only a mild rash noted. Subsequent studies focused on establishing appropriate dosing and schedule of the drug. A schedule of dosing every 3 weeks was adopted in several early studies, and the first evidence of a unique toxicity profile emerged from these trials. Collectively, these toxicities have been described as irAE, with the most common events including dermatitis, colitis, and hepatitis. These toxicities appeared to reflect a pattern of tissue-specific inflammation. A dose-response relationship was clearly defined in a double-blind Phase II study comparing ipilimumab at doses of 0.3, 3, and 10 mg/kg every 3 weeks, followed by maintenance doses administered every 12 weeks [23]. The highest dose cohort, 10 mg/kg, had the greatest response rate (11%), followed by 3 mg/kg (4.2%) and 0.3 mg/kg (0%). The rate of irAE was also higher with increased ipilimumab dose.

Ultimately, FDA approval was based on a benefit in OS seen in a randomized Phase III trial for patients with previously treated, unresectable Stage III or Stage IV melanoma [24]. This study randomized patients in a 3:1:1 ratio to receive ipilimumab at a dose of 3 mg/kg with a peptide vaccine (two HLA-A*0201-restricted peptides derived from the melanosomal antigen gp100 emulsified in Montanide), ipilimumab alone, or the peptide vaccine alone as the control arm. Median OS in the combination ipilimumab and peptide vaccine arm (10.0 months) was similar to the ipilimumab alone arm (10.1 months) but significantly higher than the peptide vaccine alone arm (6.4 months). Moreover, survival at 1 and 2 years was clearly superior in the ipilimumab-treated group when compared with peptide vaccine alone (45.6% vs. 25.3% at 1 year; 23.8% vs. 16.3% at 2 years), delivering on the promise of immunotherapy to establish durable disease control in a subset of patients. A second randomized, placebo-controlled Phase III trial comparing ipilimumab at a dose of 10 mg/kg plus dacarbazine chemotherapy versus dacarbazine alone confirmed the survival advantage of ipilimumab treatment in patients with treatment of naïve, unresectable Stage III or Stage IV melanoma [25]. A significant survival advantage for ipilimumab-treated patients was reported at 1 year (47.3% vs. 36.3%), 2 years (28.5% vs. 17.9%), and 3 years (20.8% vs. 12.2%), highlighting the long-term survival benefit to receiving ipilimumab.

Clinical development of tremelimumab suggests that this antibody may have similar activity to ipilimumab, although the two drugs have not been compared directly. In Phase I and II studies of tremelimumab, durable responses were seen in a significant minority of patients, along with a similar profile of irAE. The dosing and schedule chosen for tremelimumab, 15 mg/kg every 3 months, in part, reflect the difference in the half-life of tremelimumab versus ipilimumab. In one Phase II trial that enrolled 251 patients with metastatic melanoma, tremelimumab was associated with a durable overall response rate of 6.6%, lasting from 8.9 months to 29.8 months [26]. A number of additional patients had SD, and a total of 21% of patients achieved an objective response or prolonged SD. A randomized, open-label Phase III trial for patients with advanced melanoma, comparing tremelimumab with chemotherapy (dacarbazine or temozolomide) was conducted. After an interim analysis failed to demonstrate a benefit (OS 10.7 months vs. 11.7 months), the study was halted [27]. These results may have been complicated, however, by an unintentional crossover in the control arm that may have had access to ipilimumab. Additionally, the optimal dosing and schedule for tremelimumab have yet to be defined. Recently updated interim results demonstrate a nonsignificant trend, favoring an OS benefit for tremelimumab (P=0.14) in patients treated at locations where ipilimumab was not available [28].

Ipilimumab, the first checkpoint-blocking antibody approved by the FDA

  • —OS benefit demonstrated in two Phase III studies of patients with advanced melanoma

  • —Approved by the FDA in March 2011

  • —Approved dose of 3 mg/kg administered every 3 weeks for four doses

TOXICITIES ASSOCIATED WITH CTLA-4-BLOCKING ANTIBODIES

The clinical testing of the human CTLA-4-blocking antibodies ipilimumab and tremelimumab uncovered novel toxicities consisting of tissue-specific inflammation seen in a unique distribution of sites. These toxicities have been characterized as irAE based on their presumed link to the activation of the immune system by CTLA-4 blockade. Tissues most commonly affected include the skin (rash, pruritus, vitiligo), bowel (diarrhea, colitis), liver (hepatitis, elevated liver enzymes), the pituitary, and other endocrine glands (hypophysitis, hypothyroidism, thyroiditis, adrenal insufficiency; Table 2) [24]. Much more rarely reported irAE include uveitis, conjunctivitis, neuropathy, myopathy, pancreatitis, cytopenias, and nephritis [3236]. irAE are typically responsive to interruption or discontinuation of CTLA-4 blockade in combination with immunosuppressive drugs, such as corticosteroids or occasionally TNF-blocking antibodies (for colitis) or mycophenolate mofetil (for hepatitis) [37, 38]. Irreversible damage to endocrine organs during this period of inflammation may necessitate hormone supplementation [3941].

Table 2. Selected Clinical Trials of Checkpoint-Blocking Antibodies in Patients with Advanced Melanoma.

Agent tested Patients Treatment arms Response rates Median OS
CTLA-4 blockade
    Ipilimumab [24] Six-hundred seventy-six patients with previously treated, unresectable Stage III or IV melanoma Ipilimumab versus gp100 peptide vaccine versus combination Ipilimumab alone
BORR 10.9% 10.1 months
DCR 28.5% 45.6% at 1 year
Ipilimumab 3 mg/kg q3 weeks × 4 doses two CR, 13 PR, 24 SD 23.5% at 2 years
Ipilimumab + gp100 peptide vaccine
BORR 5.7% 10.0 months
DCR 20.1% 43.6% at 1 year
one CR, 22 PR, 58 SD 21.6% at 2 years
gp100 Peptide vaccine alone
BORR 1.5% 6.4 months
DCR 11.0% 25.3% at 1 year
zero CR, two PR, 13 SD 13.7% at 2 years
    Ipilimumab [29] Two-hundred seventeen patients with previously treated metastatic melanoma Ipilimumab 10 mg/kg
0.3 mg/kg BORR 11.1% 11.4 months
versus 3 mg/kg DCR 29.2% 48.6% at 1 year
versus 10 mg/kg two CR, six PR, 13 SD
q3 weeks × 4 doses, then q 3 months
3 mg/kg
BORR 4.2% 8.7 months
DCR 26.4% 39.3% at 1 year
zero CR, three PR, 16 SD
0.3 mg/kg
BORR 9% 8.6 months
DCR 13.7% 39.6% at 1 year
zero CR, zero PR, 10 SD 39.6% at 1 year
zero CR, zero PR, 10 SD
PD-1 or PD-L1 blockade
    Nivolumab [30] One-hundred four patients with advanced melanoma (94 evaluable for response) Nivolumab (BMS-936558) 0.1, 0.3, 1, 3, or 10 mg/kg q 2 weeks for up to 96 weeks All dose levels
ORR 28% NR
    BMS-936559 [31] Fifty-five patients with advanced melanoma (52 evaluable for response) BMS-936559 (MDX-1105) 0.3, 1, 3, or 10 mg/kg q 2 weeks for up to 96 weeks All dose levels
ORR 17% NR
three CR, six PR, 14 SD

BORR, Best overall response rate; DCR, disease control rate; ORR, overall response rate; NR, not reported.

Research Questions: What are the patient-related risk factors for the development of an irAE? What antibody-related factors determine the type(s) of irAEs that develop?

Curiously, this spectrum of irAE was not generally observed in the preclinical testing of ipilimumab or tremelimumab in animal models [42]. In some specific preclinical models, evidence that CTLA-4 blockade might lead to immune-related damage of nontumor tissue was reported. For example, successful treatment of B16 tumor-bearing mice with a combination of CTLA-4 blockade plus the GVAX melanoma vaccine could lead to depigmentation, presumably through the targeting of normal melanocytic antigens [43]. However, more generalized evidence of tissue inflammation, such as colitis or dermatitis, was not seen in mice treated with CTLA-4 blockade as a monotherapy, perhaps reflecting differences in the half-life of the murine antibody, lack of genetic diversity, or the pathogen-free housing conditions of the mice. In a variety of mouse models of autoimmunity, CTLA-4 blockade can exacerbate the underlying disease [44]. Accordingly, patients with autoimmune disease have been excluded from clinical trials of ipilimumab or tremelimumab.

KINETICS OF TUMOR RESPONSE AND RE-EVALUATION OF TRADITIONAL RADIOGRAPHIC RESPONSE ENDPOINTS

Standard criteria for evaluating responses to chemotherapy have been developed to promote objectivity and to facilitate comparisons across trials. One commonly used response criteria is the RECIST, which is based on patterns of responses to chemotherapeutic agents that correlate with clinical outcomes. According to RECIST and the closely related, modified WHO criteria, an increase in tumor size and/or development of new lesions are defined as progressive disease, typically implicating that a change in therapy is warranted (Table 3). Observations in Phase I and Phase II clinical trials with CTLA-4 blockade suggested that patterns of response to immunotherapy may differ significantly from standard responses to chemotherapy. Assessing efficacy of immunotherapy by RECIST or WHO criteria, therefore, may not be appropriate.

Table 3. Comparison between irRC and WHO Criteria.

Parameter-evaluated irRC WHO
CR Disappearance of all lesions in two consecutive observations not less than 4 weeks apart Disappearance of all lesions in two consecutive observations not less than 4 weeks apart
PR ⩾50% Decrease in tumor burden compared with baseline in two observations at least 4 weeks apart ⩾50% Decrease in SPD of all index lesions compared with baseline in two observations at least 4 weeks apart, in the absence of new lesions or unequivocal progression of nonindex lesions
SD 50% Decrease in tumor burden compared with baseline cannot be established, nor 25% increase compared with nadir 50% Decrease in SPD compared with baseline cannot be established, nor 25% increase compared with nadir, in the absence of new lesions or unequivocal progression of nonindex lesions
Progressive disease At least 25% increase in tumor burden compared with nadir (at any single time-point) in two consecutive observations at least 4 weeks apart At least 25% increase in SPD compared with nadir and/or unequivocal progression of nonindex lesions and/or appearance of new lesions (at any single time-point)

SPD, Sum of the products of the two largest perpendicular diameters. Tumor burden = SPDindex lesions + SPDnew, measurable lesions. Adapted from ref. [29].

To address this question, the radiographic responses of 487 patients treated on three multicenter Phase II clinical trials of ipilimumab were analyzed, and four patterns were identified: decreased baseline lesions without new lesions; durable SD; initial increase in total tumor burden but eventual response; and response of initial lesions with development of new lesions [29]. Interestingly, each of these patterns was associated with favorable survival. Based on these observations, irRC were proposed to evaluate the benefits of immunotherapy, previously, potentially misrepresented by traditional RECIST criteria. The main difference of irRC compared with traditional WHO or RECIST criteria is that irRC requires new lesions to be considered as part of the “total tumor burden” and not considered immediately as progressive disease. A detailed comparison of irRC and WHO criteria is presented in Table 3. The irRC is already being used in parallel with traditional criteria in current clinical protocols, and prospective validation of its correlation with additional clinical endpoints is ongoing.

Clinical and Research Questions: What factor(s) determine the kinetics of an antitumor response in patients treated with checkpoint-blocking antibodies?

What features of the tumor or host immune system explain the cases of durable responses?

In cases of tumor relapse, what features of the tumor or host immune system allow the tumor to escape?

Although radiographic response rate, whether by irRC or traditional criteria, is important, OS is perhaps the most important and least subjective study endpoint in therapeutic oncology trials. Results involving other forms of tumor immunotherapy, such as sipuleucel-T, an autologous cellular vaccine product, have demonstrated that immunotherapeutic strategies can confer a survival benefit in the absence of meaningful traditional disease responses [4547]. Although many questions remain, the presence of a survival benefit without a convincing tumor response suggests that traditional methods of measuring disease response may need to be revised when considering immunotherapeutic approaches. Lastly, another unique aspect of the responses to CTLA-4 blockade has been the very slow kinetics of responses and the remarkable durability of responses. Prieto et al. [48] reported on the long-term follow-up for 177 patients treated in some of the earliest clinical trials of ipilimumab [49]. For the 15 patients who achieved a CR, a median of 30 months was required to achieve this endpoint. Encouragingly, all of these responses, except for one, are ongoing, with the longest lasting 99+ months.

DOSING AND SCHEDULE

The FDA-approved schedule for ipilimumab treatment is one dose every 3 weeks for a total of four doses. Some clinical trials have permitted additional, so-called “maintenance” doses of ipilimumab, administered every 3 months after initial induction therapy. Alternatively, some trials have permitted repeat dosing or “reinduction” therapy using the original four-dose induction schedule. The Hodi study [24]provides some limited evidence that reinduction with ipilimumab may help patients who have benefitted previously but then ultimately developed disease progression. In this study, 31 patients with progressive disease after a confirmed PR, CR, or at least 3 months of SD were offered reinduction with subsequent evaluation showing one CR, five PRs, and 15 with SD. The use of the maintenance-dosing schedule has not been tested in a randomized fashion.

Clinical Questions: What is the optimal dose of ipilimumab?

Is there a role for maintenance dosing of ipilimumab?

Is there a role for reinduction dosing of ipilimumab?

The FDA has approved ipilimumab at a dose of 3 mg/kg. However, it is not clear that this reflects the best clinical activity for ipilimumab, and several important questions related to dosing and schedule have not been answered. What is the most effective dose of ipilimumab? Phase I studies did not identify a MTD. Furthermore, the randomized, double-blinded Phase II study, comparing ipilimumab at three dose levels—0.3, 3, and 10 mg/kg—demonstrated dose-dependent antitumor activity. The improved response rate (0% vs. 4.2% vs. 11.1%) must be balanced against an increased rate of Grade 3/4 irAE (0% vs. 7% vs. 25%). A double-blind Phase III study comparing the activity of ipilimumab at the 10 mg/kg versus the 3 mg/kg dose is ongoing (NCT01515189).

PD-1-BLOCKING ANTIBODIES

PD-1 is a second coinhibitory receptor expressed on T cells and a promising therapeutic target. Although CTLA-4 and PD-1 function as negative regulators, they play unique roles in the regulatory pathway of activated T cells. PD-1 appears to play a more prominent role in modulating T cell activity in peripheral tissues via interaction with PD-L1 and PD-L2. The expression pattern of PD-L1, expressed broadly on hematopoietic and nonhematopoietic tissues, and PD-L2, expressed on DCs, macrophages, mast cells, and B cells, supports this notion. Furthermore, PD-L1 and PD-L2 are up-regulated under inflammatory conditions, mediated, in part, by type I and type II IFNs [5054]. PD-L1 and to a lesser extent, PD-L2 are expressed on many human tumors, including urothelial, ovarian, breast, cervical, colon, pancreatic, gastric cancers, as well as melanoma glioblastoma and NSCLC [5566]. In addition, these molecules have also been detected on hematologic malignancies, including Hodgkin lymphoma, primary mediastinal B cell lymphoma, angioimmunoblastic T cell lymphoma, multiple myeloma, acute myeloid leukemia, chronic lymphocytic leukemia, and adult T cell leukemia/lymphoma [54, 6770]. Expression of PD-L1 has been correlated with prognosis in many of these malignancies, fueling the hypothesis that PD-L1 expression is a mechanism for tumor immune evasion [17, 58, 62, 71]. However, these findings must be interpreted with caution, given the technical difficulties with detecting PD-L1 in formalin-fixed tissues and the lack of a uniform assay. Additionally, PD-1 is highly expressed on lymphocytes infiltrating human tumors and circulating tumor-specific T cells, a phenotype that may be correlated with impaired T cell function in some settings [7275]. Together, these findings suggest that interrupting the PD-1:PD-L1/PD-L2 interaction could be an effective anticancer therapy by blunting inhibition of immune responses in the tumor microenvironment [76, 77].

Research Question: How do the mechanisms of activity for CTLA-4 and PD-1 or PD-L1 blockade translate into clinical activity for each agent? What features are tumor-type-specific or specific to the individual tumor or host immune system?

Several antibodies that inhibit the PD-1 pathway, by blockade of the PD-1 molecule or PD-L1, have been developed, and their characteristics are described in Table 1 [78]. This area is evolving rapidly; this review will focus on agents for which clinical data have been published. Nivolumab (BMS-936558) is a fully human IgG4 antibody. The largest experience with nivolumab reported to date comes from a Phase Ib dose-escalation study, including 269 patients with advanced solid tumors, including melanoma, NSCLC, prostate cancer, RCC, and CRC [79]. Patients received nivolumab at doses ranging from 0.1 to 10 mg/kg on an every-other-week schedule. In the initial reports, notable response rates were observed in patients with NSCLC (18%), melanoma (28%), and RCC (27%). These data have since been updated and presented recently at the European Society for Medical Oncology, now including a total of 304 patients. Updated results support the original findings; for example, in 106 evaluable patients with advanced melanoma, the response rate was 31% [80]. Responses in NSCLC were especially remarkable, as this cancer has not been described previously as amenable to immunologic approaches to disease control. In comparison with CTLA-4 blockade, rates of previously defined irAE (diarrhea, rash) appeared to be less frequent. However, a new and potentially serious irAE—pneumonitis—was observed in 3% of patients (Table 2).

MK-3475 (Merck) is a humanized IgG4 antibody directed against PD-1. A Phase I open-label, dose-escalation study tested doses of 1, 3, or 10 mg/kg in 27 patients with advanced solid tumors [81]. No dose-limiting toxicity was identified within this range, and clinical activity was noted. Notably, approximately one-half of the patients treated with MK-3475 in this study had been treated previously with ipilimumab. Thus, these data suggest that the PD-1-blocking antibodies may have activity in patients with advanced melanoma who are refractory to treatment with ipilimumab. More recently, the interim results of a Phase Ib study testing doses of 3 or 10 mg/kg were reported for 85 patients with advanced melanoma [31]. An objective response rate of 51% (43/85) and a CR rate of 9% (8/85) were reported. Adverse events appear consistent with irAE noted previously for nivolumab and included rash, diarrhea, itching, and arthralgias. CT-011 (CureTech) is a humanized IgG1 antibody that was tested initially in a Phase I open-label, dose-escalation, single-dose (0.2–6 mg/kg) study of 17 patients with hematological malignancies [30]. The MTD was not achieved, and the doses tested were well-tolerated. The most frequent adverse event was diarrhea (11.8%). Patients who achieved a response or sustained SD totaled 33% of the study population, and one patient with follicular lymphoma showed no evidence of malignancy following therapy. Based on these promising results, several Phase II protocols of CT-011 are being conducted.

Clinical Question: How does the response (or lack of response) to one checkpoint-blocking antibody predict response (or lack of response) to another checkpoint-blocking antibody?

Blockade of PD-L1 represents an alternative approach to disrupting PD-1 signaling, and three antibodies have been developed for clinical use (Table 1). BMS-936559 (previously MDX-1105; Bristol-Myers Squibb) is a fully human, PD-L1-specific IgG4 mAb. Results of a Phase I study evaluating the clinical activity of BMS-936559 in 207 patients with advanced solid tumors, including NSCLC, melanoma, CRC, RCC, ovarian cancer, pancreatic cancer, gastric cancer, and breast cancer, were reported recently [82]. As with PD-1 blockade, objective responses were seen in patients with melanoma (17%), RCC (12%), and NSCLC (10%). Additionally, one patient with ovarian cancer had an objective response. As with nivolumab, irAE appeared less common with PD-L1 blockade than with CTLA-4 blockade. In contrast to nivolumab, no cases of pneumonitis were reported with BMS-936559. The preservation of regulatory signaling via PD-L2 may be one explanation for the relative infrequency of irAE for PD-L1 blockade [83].

CANDIDATE BIOMARKERS OF RESPONSE

Monitoring of immunological parameters has been an integral component of completed and ongoing clinical trials of CTLA-4 and PD-1 blockade. Recent studies investigating the tumor microenvironment highlight the important differences in immune activity in the tumor compared with peripheral blood. The majority of putative biomarkers has been identified in small, retrospective analyses, and prospective validation is an area of active research.

Early studies of ipilimumab-treated patients identified the ALC as a marker of interest. In a retrospective review of 379 patients treated with ipilimumab, followed by prospective validation involving 64 additional patients, the rate of rise in ALC correlated with patients meeting a CR, PR, or SD, lasting >24 weeks [84]. A second, independent study of 51 patients showed that patients with ALC, >1000 after the second dose of ipilimumab, had a higher rate of CR, PR, or SD at Week 24 of therapy [85]. In this study, the high ALC group had higher OS at 6 and 12 months. Furthermore, it appears that CD8+ T cells may be the pertinent lymphocyte subset responsible for the beneficial response to ipilimumab, as suggested by analysis of lymphocyte subgroups [86].

Clinical and Research Question: What features of individual patients (i.e., their immune system) determine the likelihood of response to checkpoint blockade?

Antigen-specific immune responses during CTLA-4 blockade have been evaluated for a number of cancer-related antigens, such as NY-ESO-1, MAGE, Melan-A, melanoma antigen recognized by T cell 1, gp-100, tyrosinase, prostate-specific antigen, prostate acid phosphatase, and prostate-specific membrane antigen. Immune responses to the cancer-testis antigen NY-ESO-1 have been characterized the most extensively to correlate with clinical activity following CTLA-4 inhibition. In one study of 15 patients with metastatic melanoma treated with ipilimumab, five of eight (62.5%) with tumor shrinkage or stabilization had developed NY-ESO-1 antibodies [87]. All five of these seropositive patients had clearly detectable CD4+ and CD8+ T cells against NY-ESO-1. In contrast, none of the seven patients with progressive disease was seropositive for NY-ESO-1. There was, however, one patient with progressive disease who had a CD4+ T cell response. These findings imply that the development of polyfunctional NY-ESO-1-specific T cells, as a surrogate of a more developed antitumor immune response or as direct mediators of antitumor immunity, may identify patients likely to benefit from ipilimumab. This connection was also supported by the findings from a larger, retrospective study of 144 ipilimumab-treated patients with melanoma [88]. In this study, NY-ESO-1-seropositive patients were more likely to have a CR, PR, or prolonged SD after treatment with ipilimumab, with an even stronger positive correlation in patients that were seropositive and positive for NY-ESO-1 CD8+ T cells. This correlation between ipilimumab and NY-ESO-1 serology was not found, however, in another study [89].

Clinical and Research Question: What features of individual tumors determine the likelihood of response to checkpoint blockade?

ICOS is a costimulatory molecule thought to play an especially important role in T cell survival, proliferation, and generation of memory [90]. ICOS is up-regulated on activated T cells, and ICOS expression on T cells has been proposed as a potential biomarker for clinical activity in patients treated with checkpoint-blocking antibodies. The first studies correlating ICOS expression with clinical activity came from patients with bladder cancer, treated with neoadjuvant ipilimumab. In this setting, ICOS was identified as a biomarker up-regulated on tumor-infiltrating T cells [91]. Subsequent studies extended these findings to patients with prostate or breast cancer, treated with CTLA-4 blockade [92, 93]. Finally, in a retrospective analysis of melanoma patients treated with ipilimumab, increased frequency of CD4+ICOShigh T cells, sustained over a period of 12 weeks, correlated positively with increased OS [94].

Recent studies have highlighted the important role that the tumor microenvironment plays in modulating antitumor immune responses and the diversity of mechanisms that may interfere with effective elimination of tumor cells [95]. In a prospective Phase II study of patients with melanoma treated with ipilimumab, several immune-related biomarkers were identified in pretreatment tumor biopsies that correlated with favorable clinical outcomes [96]. High baseline expression levels of forkhead box P3 and IDO in the tumor, as well as increased levels of tumor-infiltrating lymphocytes during treatment, were all associated with clinical activity. In a further analysis of this study using gene-expression profiling, pretreated expression of immune-related genes, especially IFN-γ-responsive genes, was correlated positively with clinical activity [97].

Consistent with this emphasis on the clinical relevance of the tumor microenvironment, Topalian et al. [30] have suggested that tumor expression of PD-L1 may correlate favorably with clinical responses to nivoumab. In the Phase I study of nivoumab, PD-L1 expression, detected by immunohistochemistry, was positive in tumor biopsy samples from 25 patients and negative in 17 cases. For patients with PD-L1-positive tumors, the objective response rate was 36% (nine of 25), whereas the response rate in the PD-L1-negative group was 0%. Larger, prospective studies will be necessary to explore this further. It is worthwhile to remark that PD-L1 expression in the tumor may be dynamic and heterogeneous, and this may pose a challenge in the use of PD-L1 as a biomarker. In a retrospective review of 150 melanoma tumor samples, tumor PD-L1 expression was heterogeneous and highly concordant in colocalizing with immune infiltrate and the presence of IFN-γ, supporting the concept that tumor PD-L1 expression is a dynamic, adaptive response to immune cells in the microenvironment [98].

AREAS OF PROMISE FOR CLINICAL DEVELOPMENT OF CHECKPOINT BLOCKADE

Activity for checkpoint blockade outside of melanoma

Checkpoint-blocking antibodies have been tested most extensively in patients with advanced melanoma, but evolving data support testing the clinical activity of these agents more widely. In mouse models of transplantable tumors, CTLA-4 blockade has demonstrated activity in diverse tumor types, including glioma, sarcoma, ovarian carcinoma, and bladder carcinoma (reviewed by Grosso and Jure-Kunkel [98]). In some tumor types, such as breast, prostate, and colorectal tumors, CTLA-4 blockade was not active in the monotherapy setting, but activity was seen in combination with other agents. Recent Phase I studies of the PD-1-blocking antibody (BMS-936558) and the PD-L1-blocking antibody (BMS-936559) have underscored the wider potential of checkpoint blockade. In a Phase I study of BMS-936558, clinical activity was seen in patients with NSCLC (18% objective response rate;14/76 patients) and RCC (27% objective response rate; nine of 33 patients), in addition to patients with melanoma (28% objective response rate; 26/94 patients) [30]. No objective responses were seen in patients with prostate or CRC. In a Phase I study evaluating BMS-936559, evidence of clinical activity was seen in patients with NSCLC, RCC, and melanoma, and one patient with ovarian cancer also had a response [31]. No objective responses were seen in patients with pancreatic or CRC. Phase III studies of BMS-936558 in NSCLC, RCC, and melanoma are ongoing. Ipilimumab has also been evaluated in nonmelanoma tumors with modest evidence of clinical activity in some tumor types. Objective radiographic and biochemical responses have been reported in patients with prostate cancer [99102]. At present, two Phase III trials of ipilimumab in prostate cancer are ongoing. A study combining chemotherapy with ipilimumab for NSCLC suggested a modest benefit [103]. A Phase II study in patients with metastatic RCC also demonstrated some clinical activity [104]. In a study of metastatic pancreatic cancer, no responses were seen in patients treated with ipilimumab [105]. Additional studies will be necessary to understand the diversity in responses to checkpoint blockade across the spectrum of tumor types and evaluate whether novel combinations of therapies may demonstrate activity in tumors where robust activity has not been seen thus far.

Clinical and Research Question: What tumor types are most (or least) amenable to treatment with checkpoint-blocking antibodies and why?

Novel combinations

As the clinical activity of checkpoint-blocking antibodies in the monotherapy setting is established, the next step will be to assess the safety, feasibility, and activity of a variety of combinations (Table 4). CTLA-4-blocking antibodies and PD-1-blocking antibodies have demonstrated clinical antitumor activity against melanoma and other solid tumors. Preclinical studies suggest that CTLA-4 and PD-1- or PD-L1-blocking antibodies may be combined to achieve superior antitumor activity [106, 107]. The nonredundant functions of CTLA-4 and PD-1 support the concept that combined blockade could have favorable clinical activity. However, similarities in the toxicity profile for CTLA-4-blocking and PD-1/PD-L1-blocking agents (Table 5) highlight the importance of evaluating toxicity of combined blockade. A first clinical trial in humans, combining ipilimumab with nivolumab in a concomitant schedule, is presently ongoing (NCT01024231). Anecdotal experience with individual patients who have responded to PD-1 blockade after failing to respond to ipilimumab, or vice-versa, supports the notion that these agents have distinct mechanisms of action. However, additional studies will be needed to better delineate the optimal sequencing of these two agents.

Table 4. Planned or Ongoing Studies of Combination Therapies with Checkpoint-Blocking Antibodies.

Combinations Tumor type(s) NCT number
Combinations with targeted inhibitors
Ipilimumab + vemurafenib Melanoma NCT01673854
NCT01400451
Ipilimumab + dabrafenib ± trematenib Melanoma NCT01767454
Ipilimumab + BMS-908662 Melanoma NCT01245556
Ipilimumab + imatinib Ckit mutant tumors NCT01738139
Ipilimumab + radiation Melanoma NCT01565837
NCT01689974
NCT01703507
Ipilimumab + androgen-deprivation therapy Prostate cancer NCT01377389
NCT01688492
NCT01498978
Ipilimumab + dasatinib Gastrointestinal stromal tumor NCT01643278
Combinations with immunotherapies
Ipilimumab + nivolumab Melanoma NCT01783938
NCT01024231
Ipilimumab + anti-OX40 Melanoma NCT01689870
Ipilimumab + anti-KIR antibody Advanced cancers NCT01750580
Ipilimumab + rIL-21 Melanoma NCT01489059
Ipilimumab + GM-CSF Melanoma, prostate cancer NCT01134614
NCT01530984
NCT00064129
NCT01363206
Ipilimumab + adoptive cell transfer Melanoma NCT01701674
Ipilimumab after allogeneic stem cell transplant Advanced cancers NCT00060372
Ipilimumab + IFN-α-2b Melanoma NCT01496807
NCT01708941
Ipilimumab + autologous DC vaccine Melanoma NCT01302496
Ipilimumab + IDO inhibitor Melanoma NCT01604889
Ipilimumab + intratumoral IL-2 Melanoma NCT01672450
NCT01480323
Ipilimumab + OncoVex Melanoma NCT01740297
Other combinations
Ipilimumab + bevacizumab Melanoma NCT00790010
Ipilimumab + radiation Melanoma and other tumor types NCT01565837
NCT01689974
NCT01703507
NCT01769222
NCT01449279
NCT01557114
NCT01497808
Ipilimumab + lenolidamide Advanced cancers NCT01750983
Ipilimumab + isolated limb infusion Melanoma NCT01323517
Ipilimumab + doxycycline + temozolamide Melanoma NCT01590082
Ipilimumab + paclitaxel + carboplatin NSCLC NCT01165216
NCT01285609
Ipilimumab + cryoablation Breast cancer NCT01502592
Ipilimumab + gemcitabine Pancreatic cancer NCT01473940
Ipilimumab + rituximab Lymphoma NCT01729806
Ipilimumab + carboplatin + etoposide Small cell lung cancer NCT01331525
Ipilimumab + radioembolization Melanoma NCT01730157
Nivolumab + combination chemotherapy NSCLC NCT01454102
Ipilimumab + gemcitabin + cisplatin Urothelial carcinoma NCT01524991
Ipilimumab + low-dose cyclophosphamide Melanoma NCT01740401
Ipilimumab + fomustine Melanoma NCT01654692

NCT, National Clinical Trial.

Table 5. irAE Associated with Checkpoint Blockade.

Toxicity Any grade Grade 3/4
Ipilimumaba (data from Phase III study; ref. [23])
Rash 18% (93/511) 1% (six/511)
Pruritus 19% (99/511) <1% (one/511)
Diarrhea 30% (151/511) 4% (20/511)
Hepatitis 1% (five/511) <1% (two/511)
Hypophysitis <1% (four/511) <1% (four/511)
Hypothyroidism 2% (eight/511) <1% (one/511)
Pneumonitis NR NR
Nivolumabb (data from Phase I study; ref. [30])
Rash 12% (31/259) NR
Pruritus 10% (26/259) <1% (one/259)
Diarrhea 12% (30/259) 1% (three/259)
Hepatitis (increased alanine aminotransferase) 4% (11/296) 1% (two/296)
Hypophysitis <1% <1%
Hypothyroidism 2% (six/259) <1% (one/259)
Pneumonitis 3% (nine/259) 1% (three/259)
a

Ipilimumab monotherapy or ipilimumab plus peptide vaccine at a dose of 3 mg/kg; patients with advanced melanoma.

b

Nivolumab monotherapy at doses of 0.1–10 mg/kg; patients with advanced solids tumors.

Clinical Question: Which combinations are feasible and safe in the clinic?

CTLA-4 blockade and PD-1 blockade are also being tested in combination with traditional anticancer modalities. In the context of these combination studies, traditional therapies are being looked at through the lens of immunology. The potential for cytotoxic therapies, including chemotherapy, radiotherapy, and cryotherapy, to modulate immune activity is increasingly being appreciated, adding a layer of complexity to the design of combination regimens [108110]. With the use of radiation therapy as an example, systemic antitumor effects from local radiation (described as “abscopal” effects) and are thought to be mediated via changes in the immune system [111]. Preclinical mouse model studies provide evidence that radiation-induced changes impact the antitumor immune response and that radiation may partner favorably with immunotherapies [112114]. In case reports, patients treated with ipilimumab have been described to benefit from the abscopal effect of local radiation [115, 116]. These observations have provided the rationale for several clinical trials, now ongoing, combining radiation therapy with ipilimumab. Another area of active exploration is the combination of checkpoint blockade with targeted inhibitors, a topic of particular interest in melanoma, where the BRAF inhibitor vemurafenib and ipilimumab are the two therapies that have demonstrated a survival benefit.

Clinical Question: Can combinations of checkpoint-blocking antibodies be tailored to an individual's tumor and/or immune system to improve clinical activity?

SUMMARY AND FUTURE DIRECTIONS

The immune system has established an elaborate system of self-regulation to prevent against excessively damaging immune responses. Although essential for immunologic homeostasis, in the presence of active malignancy, inhibitory signaling may predominate and prevent effective antitumor immune responses. Research over the past two decades has highlighted the important components of several immune-regulatory circuits, and insight into these mechanisms has led to novel approaches to tumor immunotherapy. Antibodies that block inhibitory pathways, such as those directed against CTLA-4 and PD-1, have been developed for clinical use. These checkpoint-blocking antibodies are revitalizing interest in solid tumor immunotherapy and have resulted in promising clinical outcomes, exemplified by the recent FDA approval of ipilimumab. Lessons learned from studies of CTLA-4 and PD-1 blockade provide a foundation for the further development of checkpoint-blocking antibodies but highlight a number of outstanding questions that remain (Table 6). These studies have opened the door for clinical development of agents targeting a variety of additional coinhibitory and costimulatory molecules, including LAG-3, T cell Ig and mucin domain-3, GITR, OX-40, and 4-1BB, among others.

Table 6. Summary and Future Questions for the Clinical Development of Checkpoint-Blocking Antibodies.

Lessons learned Outstanding questions
CBA can generate potent antitumor immune responses with evidence of clinical activity demonstrated in patients with melanoma, RCC, and NSCLC and anecdotal reports of activity in other solid tumors. What tumor types are most (or least) amenable to treatment with CBA and why?
Many patients do not have objective responses to CBA monotherapy based on the dosing and schedule of antibodies tested thus far. What features of individual tumors determine the likelihood of response to checkpoint blockade?
What features of individual patients (i.e., their immune system) determine the likelihood of response to checkpoint blockade?
Can the type of CBA be customized based on tumor or patient characteristics to enhance the likelihood of response?
Compared with CTLA-4 blockade, early studies suggest that PD-1-blocking antibodies have a favorable response rate; data on long-term survival for larger numbers of patients treated with PD-1-blocking antibodies are awaited. How do the mechanisms of activity for CTLA-4 and PD-1 or PD-L1 blockade translate into clinical activity for CBA? What features are tumor-type-specific or specific to the individual tumor or patient?
Does response (or lack of response) to one CBA predict response (or lack of response) to another CBA?
CBA can generate immune activation leading to tissue-specific inflammation and consequent toxicities in a variety of organs. What determines which patients develop toxicity and which type(s) of toxicity develop?
In early studies, PD-1- and PD-L1-blocking antibodies appear to have an acceptable toxicity profile. How can toxicity be uncoupled from antitumor activity (some patients have a tumor response without toxicity and others, vice versa)?
CTLA-4- and PD-1-blocking antibodies generate overlapping and unique toxicities. How do the mechanisms of activity for CTLA-4 blockade and PD-1 or PD-L1 blockade translate into the toxicity profile for each agent (what explains unique toxicities such as pneumonitis)?
Compared with traditional cytotoxic anticancer agents, CBA may have unique and delayed kinetics of response and remarkable durability of responses. What factors determine the kinetics of an antitumor response in patients treated with CBA, and what explains the cases of delayed responses?
In cases where durable responses are seen, what features of the tumor or the immune system determine durable disease control?
In cases of tumor relapse, what features of the tumor or the immune system allow the tumor to escape?
Preclinical studies suggest that CBA may be combined successfully with a diversity of anticancer agents, generating superior antitumor activity. Which combinations are feasible and safe in the clinic?
Which combinations generate the most robust antitumor activity and in what setting?
Can combinations of CBA be tailored to an individual's tumor and/or immune system to improve clinical activity?

CBA, Checkpoint-blocking antibodies.

ACKNOWLEDGMENTS

The authors thank Andrew M. Intlekofer and Craig B. Thompson for comments and critique during the preparation of this review.

SEE CORRESPONDING ARTICLE ON PAGE 25

ALC
absolute lymphocyte count
CR
complete response
CRC
colorectal cancer
FDA
U.S. Food and Drug Administration
GITR
glucocorticoid-induced TNFR family-related gene
irAE
immune-related adverse event(s)
irRC
immune-related response criteria
LAG-3
lymphocyte-activation gene-3
MTD
maximum tolerated dose
NSCLC
nonsmall cell lung cancer
OS
overall survival
PD-1
programmed death-1
PD-L1
programmed death-1 ligand
PR
partial response(s)
RCC
renal cell cancer
RECIST
Response Evaluation Criteria in Solid Tumors
SD
stable disease
WHO
World Health Organization

AUTHORSHIP

M.K.C. and J.D.W. wrote the review.

REFERENCES

  • 1. Baxter A. G., Hodgkin P. D. (2002) Activation rules: the two-signal theories of immune activation. Nat. Rev. Immunol. 2, 439–446 [DOI] [PubMed] [Google Scholar]
  • 2. Jenkins M. K., Schwartz R. H. (1987) Antigen presentation by chemically modified splenocytes induces antigen-specific T cell unresponsiveness in vitro and in vivo. J. Exp. Med. 165, 302–319 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Lafferty K. J., Cunningham A. J. (1975) A new analysis of allogeneic interactions. Aust. J. Exp. Biol. Med. Sci. 53, 27–42 [DOI] [PubMed] [Google Scholar]
  • 4. Bretscher P., Cohn M. (1970) A theory of self-nonself discrimination. Science 169, 1042–1049 [DOI] [PubMed] [Google Scholar]
  • 5. Dunn G. P., Old L. J., Schreiber R. D. (2004) The three Es of cancer immunoediting. Annu. Rev. Immunol. 22, 329–360 [DOI] [PubMed] [Google Scholar]
  • 6. Peggs K. S., Quezada S. A., Allison J. P. (2009) Cancer immunotherapy: co-stimulatory agonists and co-inhibitory antagonists. Clin. Exp. Immunol. 157, 9–19 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Leach D. R., Krummel M. F., Allison J. P. (1996) Enhancement of antitumor immunity by CTLA-4 blockade. Science 271, 1734–1736 [DOI] [PubMed] [Google Scholar]
  • 8. Kwon E. D., Hurwitz A. A., Foster B. A., Madias C., Feldhaus A. L., Greenberg N. M., Burg M. B., Allison J. P. (1997) Manipulation of T cell costimulatory and inhibitory signals for immunotherapy of prostate cancer. Proc. Natl. Acad. Sci. USA 94, 8099–8103 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Yang Y. F., Zou J. P., Mu J., Wijesuriya R., Ono S., Walunas T., Bluestone J., Fujiwara H., Hamaoka T. (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, 4036–4041 [PubMed] [Google Scholar]
  • 10. Shrikant P., Khoruts A., Mescher M. F. (1999) CTLA-4 blockade reverses CD8+ T cell tolerance to tumor by a CD4+ T cell- and IL-2-dependent mechanism. Immunity 11, 483–493 [DOI] [PubMed] [Google Scholar]
  • 11. Sotomayor E. M., Borrello I., Tubb E., Allison J. P., Levitsky H. I. (1999) In vivo blockade of CTLA-4 enhances the priming of responsive T cells but fails to prevent the induction of tumor antigen-specific tolerance. Proc. Natl. Acad. Sci. USA 96, 11476–11481 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Allison J. P., Hurwitz A. A., Leach D. R. (1995) Manipulation of costimulatory signals to enhance antitumor T-cell responses. Curr. Opin. Immunol. 7, 682–686 [DOI] [PubMed] [Google Scholar]
  • 13. Grosso J. F., Kelleher C. C., Harris T. J., Maris C. H., Hipkiss E. L., De Marzo A., Anders R., Netto G., Getnet D., Bruno T. C., Goldberg M. V., Pardoll D. M., Drake C. G. (2007) LAG-3 regulates CD8+ T cell accumulation and effector function in murine self- and tumor-tolerance systems. J. Clin. Invest. 117, 3383–3392 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Hirano F., Kaneko K., Tamura H., Dong H., Wang S., Ichikawa M., Rietz C., Flies D. B., Lau J. S., Zhu G., Tamada K., Chen L. (2005) Blockade of B7-H1 and PD-1 by monoclonal antibodies potentiates cancer therapeutic immunity. Cancer Res. 65, 1089–1096 [PubMed] [Google Scholar]
  • 15. Curiel T. J., Wei S., Dong H., Alvarez X., Cheng P., Mottram P., Krzysiek R., Knutson K. L., Daniel B., Zimmermann M. C., David O., Burow M., Gordon A., Dhurandhar N., Myers L., Berggren R., Hemminki A., Alvarez R. D., Emilie D., Curiel D. T., Chen L., Zou W. (2003) Blockade of B7-H1 improves myeloid dendritic cell-mediated antitumor immunity. Nat. Med. 9, 562–567 [DOI] [PubMed] [Google Scholar]
  • 16. Strome S. E., Dong H., Tamura H., Voss S. G., Flies D. B., Tamada K., Salomao D., Cheville J., Hirano F., Lin W., Kasperbauer J. L., Ballman K. V., Chen L. (2003) B7-H1 blockade augments adoptive T-cell immunotherapy for squamous cell carcinoma. Cancer Res. 63, 6501–6505 [PubMed] [Google Scholar]
  • 17. Iwai Y., Ishida M., Tanaka Y., Okazaki T., Honjo T., Minato N. (2002) Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade. Proc. Natl. Acad. Sci. USA 99, 12293–12297 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Iwai Y., Terawaki S., Honjo T. (2005) PD-1 blockade inhibits hematogenous spread of poorly immunogenic tumor cells by enhanced recruitment of effector T cells. Int. Immunol. 17, 133–144 [DOI] [PubMed] [Google Scholar]
  • 19. Radhakrishnan S., Nguyen L. T., Ciric B., Flies D., Van Keulen V. P., Tamada K., Chen L., Rodriguez M., Pease L. R. (2004) Immunotherapeutic potential of B7-DC (PD-L2) cross-linking antibody in conferring antitumor immunity. Cancer Res. 64, 4965–4972 [DOI] [PubMed] [Google Scholar]
  • 20. Intlekofer A. M., Thompson C. B. At the bench: Preclinical rationale for CTLA-4 and PD-1 blockade as cancer immunotherapy. J. Leukoc. Biol. 94, 25–39 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Ishida Y., Agata Y., Shibahara K., Honjo T. (1992) Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. EMBO J. 11, 3887–3895 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Tchekmedyian S., et al. (2002) MDX-010 (human anti-CTLA4): a Phase I trial in malignant melanoma. Proc. Am. Soc. Clin. Oncol. 21 [Google Scholar]
  • 23. Wolchok J.D., Neyns B., Linette G., Negrier S., Lutzky J., Thomas L., Waterfield W., Schadendorf D., Smylie M., Guthrie T., Jr., Grob J.J., Chesney J., Chin K., Chen K., Hoos A., O'Day S.J., Lebbe C. (2010) Ipilimumab monotherapy in patients with pretreated advanced melanoma: a randomised, double-blind, multicentre, Phase II, dose-ranging study. Lancet Oncol. 11, 155–164 [DOI] [PubMed] [Google Scholar]
  • 24. Hodi F. S., O'Day S. J., McDermott D. F., Weber R. W., Sosman J. A., Haanen J. B., Gonzalez R., Robert C., Schadendorf D., Hassel J. C., Akerley W., van den Eertwegh A. J., Lutzky J., Lorigan P., Vaubel J. M., Linette G. P., Hogg D., Ottensmeier C. H., Lebbe C., Peschel C., Quirt I., Clark J. I., Wolchok J. D., Weber J. S., Tian J., Yellin M. J., Nichol G. M., Hoos A., Urba W. J. (2010) Improved survival with ipilimumab in patients with metastatic melanoma. N. Engl. J. Med. 363, 711–723 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Robert C., Thomas L., Bondarenko I., O'Day S., M D, W. J., Garbe C., Lebbe C., Baurain J. F., Testori A., Grob J. J., Davidson N., Richards J., Maio M., Hauschild A., Miller W. H., Jr., Gascon P., Lotem M., Harmankaya K., Ibrahim R., Francis S., Chen T. T., Humphrey R., Hoos A., Wolchok J. D. (2011) Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. N. Engl. J. Med. 364, 2517–2526 [DOI] [PubMed] [Google Scholar]
  • 26. Kirkwood J. M., Lorigan P., Hersey P., Hauschild A., Robert C., McDermott D., Marshall M. A., Gomez-Navarro J., Liang J. Q., Bulanhagui C. A. (2010) Phase II trial of tremelimumab (CP-675,206) in patients with advanced refractory or relapsed melanoma. Clin. Care Res. 16, 1042–1048 [DOI] [PubMed] [Google Scholar]
  • 27. Ribas A., et al. (2008) Phase III, open-label, randomized, comparative study of tremelimumab (CP-675,206) and chemotherapy (temozolomide or dacarbazine) in patients with advanced melanoma. J. Clin. Oncol. 26 (Suppl.; abs. LBA9011). [Google Scholar]
  • 28. Marshall M. A., Ribas A., Huang B. (2010) Evaluation of baseline serum C-reactive protein (CRP) and benefit from tremelimumab compared to chemotherapy in first-line melanoma. J. Clin. Oncol. 28 (Suppl; abs. 2609). [Google Scholar]
  • 29. Wolchok J. D., Hoos A., O'Day S., Weber J. S., Hamid O., Lebbe C., Maio M., Binder M., Bohnsack O., Nichol G., Humphrey R., Hodi F. S. (2009) Guidelines for the evaluation of immune therapy activity in solid tumors: immune-related response criteria. Clin. Care Res. 15, 7412–7420 [DOI] [PubMed] [Google Scholar]
  • 30. Topalian S. L., Hodi F. S., Brahmer J. R., Gettinger S. N., Smith D. C., McDermott D. F., Powderly J. D., Carvajal R. D., Sosman J. A., Atkins M. B., Leming P. D., Spigel D. R., Antonia S. J., Horn L., Drake C. G., Pardoll D. M., Chen L., Sharfman W. H., Anders R. A., Taube J. M., McMiller T. L., Xu H., Korman A. J., Jure-Kunkel M., Agrawal S., McDonald D., Kollia G. D., Gupta A., Wigginton J. M., Sznol M. (2012) Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N. Engl. J. Med. 366, 2443–2454 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Brahmer J. R., Tykodi S. S., Chow L. Q., Hwu W. J., Topalian S. L., Hwu P., Drake C. G., Camacho L. H., Kauh J., Odunsi K., Pitot H. C., Hamid O., Bhatia S., Martins R., Eaton K., Chen S., Salay T. M., Alaparthy S., Grosso J. F., Korman A. J., Parker S. M., Agrawal S., Goldberg S. M., Pardoll D. M., Gupta A., Wigginton J. M. (2012) Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N. Engl. J. Med. 366, 2455–2465 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Weber J. (2009) Ipilimumab: controversies in its development, utility and autoimmune adverse events. Cancer Immunol. Immunother. 58, 823–830 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Di Giacomo A. M., Biagioli M., Maio M. (2010) The emerging toxicity profiles of anti-CTLA-4 antibodies across clinical indications. Semin. Oncol. 37, 499–507 [DOI] [PubMed] [Google Scholar]
  • 34. Forde P.M., Rock K., Wilson G., O'Byrne K. J. (2012) Ipilimumab-induced immune-related renal failure—a case report. Anticancer Res. 32, 4607–4608 [PubMed] [Google Scholar]
  • 35. Maur M., Tomasello C., Frassoldati A., Dieci M. V., Barbieri E., Conte P. (2012) Posterior reversible encephalopathy syndrome during ipilimumab therapy for malignant melanoma. J. Clin. Oncol. 30, e76–e78 [DOI] [PubMed] [Google Scholar]
  • 36. Andrews S., Holden R. (2012) Characteristics and management of immunerelated adverse effects associated with ipilimumab, a new immunotherapy for metastatic melanoma. Cancer Manag. Res. 4, 299–307 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Beck K. E., Blansfield J. A., Tran K. Q., Feldman A. L., Hughes M. S., Royal R. E., Kammula U. S., Topalian S. L., Sherry R. M., Kleiner D., Quezado M., Lowy I., Yellin M., Rosenberg S. A., Yang J. C. (2006) Enterocolitis in patients with cancer after antibody blockade of cytotoxic T-lymphocyte-associated antigen 4. J. Clin. Oncol. 24, 2283–2289 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Wolchok J. (2012) How recent advances in immunotherapy are changing the standard of care for patients with metastatic melanoma. Ann. Oncol. 23 (Suppl. 8), viii15–viii21 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Juszczak A., Gupta A., Karavitaki N., Middleton M. R., Grossman A. B. (2012) Ipilimumab: a novel immunomodulating therapy causing autoimmune hypophysitis: a case report and review. Eur. J. Endocrinol. 167, 1–5 [DOI] [PubMed] [Google Scholar]
  • 40. Dillard T., Yedinak C. G., Alumkal J., Fleseriu M. (2010) Anti-CTLA-4 antibody therapy associated autoimmune hypophysitis: serious immune related adverse events across a spectrum of cancer subtypes. Pituitary 13, 29–38 [DOI] [PubMed] [Google Scholar]
  • 41. Kaehler K. C., Egberts F., Lorigan P., Hauschild A. (2009) Anti-CTLA-4 therapy-related autoimmune hypophysitis in a melanoma patient. Melanoma Res. 19, 333–334 [DOI] [PubMed] [Google Scholar]
  • 42. Peggs K. S., Quezada S. A., Allison J. P. (2008) Cell intrinsic mechanisms of T-cell inhibition and application to cancer therapy. Immunol. Rev. 224, 141–165 [DOI] [PubMed] [Google Scholar]
  • 43. van Elsas A., Hurwitz A. A., Allison J. P. (1999) Combination immunotherapy of B16 melanoma using anti-cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) and granulocyte/macrophage colony-stimulating factor (GM-CSF)-producing vaccines induces rejection of subcutaneous and metastatic tumors accompanied by autoimmune depigmentation. J. Exp. Med. 190, 355–366 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Hurwitz A. A., Sullivan T. J., Krummel M. F., Sobel R. A., Allison J. P. (1997) Specific blockade of CTLA-4/B7 interactions results in exacerbated clinical and histologic disease in an actively-induced model of experimental allergic encephalomyelitis. J. Neuroimmunol. 73, 57–62 [DOI] [PubMed] [Google Scholar]
  • 45. Kantoff P. W., Higano C. S., Shore N. D., Berger E. R., Small E. J., Penson D. F., Redfern C. H., Ferrari A. C., Dreicer R., Sims R. B., Xu Y., Frohlich M. W., Schellhammer P. F. (2010) Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N. Engl. J. Med. 363, 411–422 [DOI] [PubMed] [Google Scholar]
  • 46. Small E. J., Schellhammer P. F., Higano C. S., Redfern C. H., Nemunaitis J. J., Valone F. H., Verjee S. S., Jones L. A., Hershberg R. M. (2006) Placebo-controlled Phase III trial of immunologic therapy with sipuleucel-T (APC8015) in patients with metastatic, asymptomatic hormone refractory prostate cancer. J. Clin. Oncol. 24, 3089–3094 [DOI] [PubMed] [Google Scholar]
  • 47. Kantoff P. W., Schuetz T. J., Blumenstein B. A., Glode L. M., Bilhartz D. L., Wyand M., Manson K., Panicali D. L., Laus R., Schlom J., Dahut W. L., Arlen P. M., Gulley J. L., Godfrey W. R. (2010) Overall survival analysis of a Phase II randomized controlled trial of a poxviral-based PSA-targeted immunotherapy in metastatic castration-resistant prostate cancer. J. Clin. Oncol. 28, 1099–1105 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Prieto P. A., Yang J. C., Sherry R. M., Hughes M. S., Kammula U. S., White D. E., Levy C. L., Rosenberg S. A., Phan G. Q. (2012) CTLA-4 blockade with ipilimumab: long-term follow-up of 177 patients with metastatic melanoma. Clin. Cancer Res. 18, 2039–2047 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Postow M. A., Callahan M. K., Wolchok J. D. (2012) The antitumor immunity of ipilimumab: (T-cell) memories to last a lifetime? Clin. Cancer Res. 18, 1821–1823 [DOI] [PubMed] [Google Scholar]
  • 50. Yamazaki T., Akiba H., Iwai H., Matsuda H., Aoki M., Tanno Y., Shin T., Tsuchiya H., Pardoll D. M., Okumura K., Azuma M., Yagita H. (2002) Expression of programmed death 1 ligands by murine T cells and APC. J. Immunol. 169, 5538–5545 [DOI] [PubMed] [Google Scholar]
  • 51. Eppihimer M. J., Gunn J., Freeman G. J., Greenfield E. A., Chenova T., Erickson J., Leonard J. P. (2002) Expression and regulation of the PD-L1 immunoinhibitory molecule on microvascular endothelial cells. Microcirculation 9, 133–145 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Schreiner B., Mitsdoerffer M., Kieseier B. C., Chen L., Hartung H. P., Weller M., Wiendl H. (2004) Interferon-β enhances monocyte and dendritic cell expression of B7-H1 (PD-L1), a strong inhibitor of autologous T-cell activation: relevance for the immune modulatory effect in multiple sclerosis. J. Neuroimmunol. 155, 172–182 [DOI] [PubMed] [Google Scholar]
  • 53. Lee S. J., Jang B. C., Lee S. W., Yang Y. I., Suh S. I., Park Y. M., Oh S., Shin J. G., Yao S., Chen L., Choi I. H. (2006) Interferon regulatory factor-1 is prerequisite to the constitutive expression and IFN-γ-induced upregulation of B7-H1 (CD274). FEBS Lett. 580, 755–762 [DOI] [PubMed] [Google Scholar]
  • 54. Liu J., Hamrouni A., Wolowiec D., Coiteux V., Kuliczkowski K., Hetuin D., Saudemont A., Quesnel B. (2007) Plasma cells from multiple myeloma patients express B7-H1 (PD-L1) and increase expression after stimulation with IFN-{γ} and TLR ligands via a MyD88-, TRAF6-, and MEK-dependent pathway. Blood 110, 296–304 [DOI] [PubMed] [Google Scholar]
  • 55. Thompson R. H., Gillett M. D., Cheville J. C., Lohse C. M., Dong H., Webster W. S., Chen L., Zincke H., Blute M. L., Leibovich B. C., Kwon E. D. (2005) Costimulatory molecule B7-H1 in primary and metastatic clear cell renal cell carcinoma. Cancer 104, 2084–2091 [DOI] [PubMed] [Google Scholar]
  • 56. Thompson R. H., Gillett M. D., Cheville J. C., Lohse C. M., Dong H., Webster W. S., Krejci K. G., Lobo J. R., Sengupta S., Chen L., Zincke H., Blute M. L., Strome S. E., Leibovich B. C., Kwon E. D. (2004) Costimulatory B7-H1 in renal cell carcinoma patients: indicator of tumor aggressiveness and potential therapeutic target. Proc. Natl. Acad. Sci. USA 101, 17174–17179 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Inman B. A., Sebo T. J., Frigola X., Dong H., Bergstralh E. J., Frank I., Fradet Y., Lacombe L., Kwon E. D. (2007) PD-L1 (B7-H1) expression by urothelial carcinoma of the bladder and BCG-induced granulomata: associations with localized stage progression. Cancer 109, 1499–1505 [DOI] [PubMed] [Google Scholar]
  • 58. Nakanishi J., Wada Y., Matsumoto K., Azuma M., Kikuchi K., Ueda S. (2007) Overexpression of B7-H1 (PD-L1) significantly associates with tumor grade and postoperative prognosis in human urothelial cancers. Cancer Immunol. Immunother. 56, 1173–1182 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Hamanishi J., Mandai M., Iwasaki M., Okazaki T., Tanaka Y., Yamaguchi K., Higuchi T., Yagi H., Takakura K., Minato N., Honjo T., Fujii S. (2007) Programmed cell death 1 ligand 1 and tumor-infiltrating CD8+ T lymphocytes are prognostic factors of human ovarian cancer. Proc. Natl. Acad. Sci. USA 104, 3360–3365 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Nomi T., Sho M., Akahori T., Hamada K., Kubo A., Kanehiro H., Nakamura S., Enomoto K., Yagita H., Azuma M., Nakajima Y. (2007) Clinical significance and therapeutic potential of the programmed death-1 ligand/programmed death-1 pathway in human pancreatic cancer. Clin. Cancer Res. 13, 2151–2157 [DOI] [PubMed] [Google Scholar]
  • 61. Wu C., Zhu Y., Jiang J., Zhao J., Zhang X. G., Xu N. (2006) Immunohistochemical localization of programmed death-1 ligand-1 (PD-L1) in gastric carcinoma and its clinical significance. Acta Histochem. 108, 19–24 [DOI] [PubMed] [Google Scholar]
  • 62. Dong H., Strome S. E., Salomao D. R., Tamura H., Hirano F., Flies D. B., Roche P. C., Lu J., Zhu G., Tamada K., Lennon V. A., Celis E., Chen L. (2002) Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat. Med. 8, 793–800 [DOI] [PubMed] [Google Scholar]
  • 63. Wintterle S., Schreiner B., Mitsdoerffer M., Schneider D., Chen L., Meyermann R., Weller M., Wiendl H. (2003) Expression of the B7-related molecule B7-H1 by glioma cells: a potential mechanism of immune paralysis. Cancer Res. 63, 7462–7467 [PubMed] [Google Scholar]
  • 64. Brown J. A., Dorfman D. M., Ma F. R., Sullivan E. L., Munoz O., Wood C. R., Greenfield E. A., Freeman G. J. (2003) Blockade of programmed death-1 ligands on dendritic cells enhances T cell activation and cytokine production. J. Immunol. 170, 1257–1266 [DOI] [PubMed] [Google Scholar]
  • 65. Thompson R. H., Kuntz S. M., Leibovich B. C., Dong H., Lohse C. M., Webster W. S., Sengupta S., Frank I., Parker A. S., Zincke H., Blute M. L., Sebo T. J., Cheville J. C., Kwon E. D. (2006) Tumor B7-H1 is associated with poor prognosis in renal cell carcinoma patients with long-term follow-up. Cancer Res. 66, 3381–3385 [DOI] [PubMed] [Google Scholar]
  • 66. Konishi J., Yamazaki K., Azuma M., Kinoshita I., Dosaka-Akita H., Nishimura M. (2004) B7-H1 expression on non-small cell lung cancer cells and its relationship with tumor-infiltrating lymphocytes and their PD-1 expression. Clin. Cancer Res. 10, 5094–5100 [DOI] [PubMed] [Google Scholar]
  • 67. Xerri L., Chetaille B., Serriari N., Attias C., Guillaume Y., Arnoulet C., Olive D. (2008) Programmed death 1 is a marker of angioimmunoblastic T-cell lymphoma and B-cell small lymphocytic lymphoma/chronic lymphocytic leukemia. Hum. Pathol. 39, 1050–1058 [DOI] [PubMed] [Google Scholar]
  • 68. Dorfman D. M., Brown J. A., Shahsafaei A., Freeman G. J. (2006) Programmed death-1 (PD-1) is a marker of germinal center-associated T cells and angioimmunoblastic T-cell lymphoma. Am. J. Surg. Pathol. 30, 802–810 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Rosenwald A., Wright G., Leroy K., Yu X., Gaulard P., Gascoyne R. D., Chan W. C., Zhao T., Haioun C., Greiner T. C., et al. (2003) Molecular diagnosis of primary mediastinal B cell lymphoma identifies a clinically favorable subgroup of diffuse large B cell lymphoma related to Hodgkin lymphoma. J. Exp. Med. 198, 851–862 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Tamura H., Dan K., Tamada K., Nakamura K., Shioi Y., Hyodo H., Wang S. D., Dong H., Chen L., Ogata K. (2005) Expression of functional B7-H2 and B7.2 costimulatory molecules and their prognostic implications in de novo acute myeloid leukemia. Clin. Cancer Res. 11, 5708–5717 [DOI] [PubMed] [Google Scholar]
  • 71. Thompson R. H., Webster W. S., Cheville J. C., Lohse C. M., Dong H., Leibovich B. C., Kuntz S. M., Sengupta S., Kwon E. D., Blute M. L. (2005) B7-H1 glycoprotein blockade: a novel strategy to enhance immunotherapy in patients with renal cell carcinoma. Urology 66, 10–14 [DOI] [PubMed] [Google Scholar]
  • 72. Wong R. M., Scotland R. R., Lau R. L., Wang C., Korman A. J., Kast W. M., Weber J. S. (2007) Programmed death-1 blockade enhances expansion and functional capacity of human melanoma antigen-specific CTLs. Int. Immunol. 19, 1223–1234 [DOI] [PubMed] [Google Scholar]
  • 73. Fourcade J., Kudela P., Sun Z., Shen H., Land S. R., Lenzner D., Guillaume P., Luescher I. F., Sander C., Ferrone S., Kirkwood J. M., Zarour H. M. (2009) PD-1 is a regulator of NY-ESO-1-specific CD8+ T cell expansion in melanoma patients. J. Immunol. 182, 5240–5249 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Matsuzaki J., Gnjatic S., Mhawech-Fauceglia P., Beck A., Miller A., Tsuji T., Eppolito C., Qian F., Lele S., Shrikant P., Old L. J., Odunsi K. (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, 7875–7880 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Ahmadzadeh M., Johnson L. A., Heemskerk B., Wunderlich J. R., Dudley M. E., White D. E., Rosenberg S. A. (2009) Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired. Blood 114, 1537–1544 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Dong H., Zhu G., Tamada K., Chen L. (1999) B7-H1, a third member of the B7 family, co-stimulates T-cell proliferation and interleukin-10 secretion. Nat. Med. 5, 1365–1369 [DOI] [PubMed] [Google Scholar]
  • 77. Freeman G. J., Long A. J., Iwai Y., Bourque K., Chernova T., Nishimura H., Fitz L. J., Malenkovich N., Okazaki T., Byrne M. C., Horton H. F., Fouser L., Carter L., Ling V., Bowman M. R., Carreno B. M., Collins M., Wood C. R., Honjo T. (2000) Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J. Exp. Med. 192, 1027–1034 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Berger R., Rotem-Yehudar R., Slama G., Landes S., Kneller A., Leiba M., Koren-Michowitz M., Shimoni A., Nagler A. (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, 3044–3051 [DOI] [PubMed] [Google Scholar]
  • 79. Sosman J., Sznol M., McDermott D., Carvsjal R., Lawrence D., Topalian S. L., Wigginton J. M., Kollia G., Gupta A., Hodi F. (2012) Clinical activity and safety of anti-programmed death-1 (PD-1) (BMS-936558/MDX-1106/ONO-4538) in patients (PTS) with advanced melanoma (MEL) in ESMO. [Google Scholar]
  • 80. Patnaik A., Kang S. P., Tolcher A. W., Rasco D. W., Papadopoulos K. P., Beeram M., Drengler R., Chen C., Smith L., Perez C., Gergich K. (2012) Phase I study of MK-3475 (anti-PD-1 monoclonal antibody) in patients with advanced solid tumors. J. Clin. Oncol. 30 (Suppl.; abs. 2512). [DOI] [PubMed] [Google Scholar]
  • 81. Hamid O. (2012) Efficacy and Safety of MK-3475 in Patients with Advanced Melanoma in Society for Melanoma Research. 2012 Congress, Hollywood, CA, USA [Google Scholar]
  • 82. Chen D. S., Irving B. A., Hodi F. S. (2012) Molecular pathways: next-generation immunotherapy—inhibiting programmed death-ligand 1 and programmed death-1. Clin. Cancer Res. 18, 6580–6587 [DOI] [PubMed] [Google Scholar]
  • 83. Berman D., et al. (2009) Association of peripheral blood absolute lymphocyte count (ALC) and clinical activity in patients (pts) with advanced melanoma treated with ipilimumab. J. Clin. Oncol. 27, 3020 (abs.)19470921 [Google Scholar]
  • 84. Ku G. Y., Yuan J., Page D. B., Schroeder S. E., Panageas K. S., Carvajal R. D., Chapman P. B., Schwartz G. K., Allison J. P., Wolchok J. D. (2010) Single-institution experience with ipilimumab in advanced melanoma patients in the compassionate use setting: lymphocyte count after 2 doses correlates with survival. Cancer 116, 1767–1775 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Yang R. K. (2010) CTLA-4 blockade with ipilimumab increases peripheral CD8+ T cells: correlation with clinical outcomes. J. Clin. Oncol. 28, 2555 (abs.) [Google Scholar]
  • 86. Yuan J., Gnjatic S., Li H., Powel S., Gallardo H. F., Ritter E., Ku G. Y., Jungbluth A. A., Segal N. H., Rasalan T. S., Manukian G., Xu Y., Roman R. A., Terzulli S. L., Heywood M., Pogoriler E., Ritter G., Old L. J., Allison J. P., Wolchok J. D. (2008) CTLA-4 blockade enhances polyfunctional NY-ESO-1 specific T cell responses in metastatic melanoma patients with clinical benefit. Proc. Natl. Acad. Sci. USA 105, 20410–20415 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Yuan J., Adamow M., Ginsberg B. A., Rasalan T. S., Ritter E., Gallardo H. F., Xu Y., Pogoriler E., Terzulli S. L., Kuk D., Panageas K. S., Ritter G., Sznol M., Halaban R., Jungbluth A. A., Allison J. P., Old L. J., Wolchok J. D., Gnjatic S. (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, 16723–16728 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Goff S. L., Robbins P. F., El-Gamil M., Rosenberg S. A. (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, 884–885 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Burmeister Y., Lischke T., Dahler A. C., Mages H. W., Lam K. P., Coyle A. J., Kroczek R. A., Hutloff A. (2008) ICOS controls the pool size of effector-memory and regulatory T cells. J. Immunol. 180, 774–782 [DOI] [PubMed] [Google Scholar]
  • 90. Liakou C. I., Kamat A., Tang D. N., Chen H., Sun J., Troncoso P., Logothetis C., Sharma P. (2008) CTLA-4 blockade increases IFNγ-producing CD4+ICOShi cells to shift the ratio of effector to regulatory T cells in cancer patients. Proc. Natl. Acad. Sci. USA 105, 14987–14992 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Chen H., Liakou C. I., Kamat A., Pettaway C., Ward J. F., Tang D. N., Sun J., Jungbluth A. A., Troncoso P., Logothetis C., Sharma P. (2009) Anti-CTLA-4 therapy results in higher CD4+ICOShi T cell frequency and IFN-γ levels in both nonmalignant and malignant prostate tissues. Proc. Natl. Acad. Sci. USA 106, 2729–2734 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Vonderheide R. H., LoRusso P. M., Khalil M., Gartner E. M., Khaira D., Soulieres D., Dorazio P., Trosko J. A., Ruter J., Mariani G. L., Usari T., Domchek S. M. (2010) Tremelimumab in combination with exemestane in patients with advanced breast cancer and treatment-associated modulation of inducible costimulator expression on patient T cells. Clin. Cancer Res. 16, 3485–3494 [DOI] [PubMed] [Google Scholar]
  • 93. Carthon B. C., Wolchok J. D., Yuan J., Kamat A., Ng Tang D. S., Sun J., Ku G., Troncoso P., Logothetis C. J., Allison J. P., Sharma P. (2010) Preoperative CTLA-4 blockade: tolerability and immune monitoring in the setting of a presurgical clinical trial. Clin. Cancer Res. 16, 2861–2871 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Gajewski T. F., Fuertes M., Spaapen R., Zheng Y., Kline J. (2011) Molecular profiling to identify relevant immune resistance mechanisms in the tumor microenvironment. Curr. Opin. Immunol. 23, 286–292 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Hamid O., Schmidt H., Nissan A., Ridolfi L., Aamdal S., Hansson J., Guida M., Hyams D. M., Gomez H., Bastholt L., Chasalow S. D., Berman D. (2011) A prospective Phase II trial exploring the association between tumor microenvironment biomarkers and clinical activity of ipilimumab in advanced melanoma. J. Transl. Med. 9, 204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Ji R. R., Chasalow S. D., Wang L., Hamid O., Schmidt H., Cogswell J., Alaparthy S., Berman D., Jure-Kunkel M., Siemers N. O., Jackson J. R., Shahabi V. (2012) An immune-active tumor microenvironment favors clinical response to ipilimumab. Cancer Immunol. Immunother. 61, 1019–1031 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Taube J. M., Anders R. A., Young G. D., Xu H., Sharma R., McMiller T. L., Chen S., Klein A. P., Pardoll D. M., Topalian S. L., Chen L. (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, 127ra37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Grosso J. F., Jure-Kunkel M. N. (2013) CTLA-4 blockade in tumor models: an overview of preclinical and translational research. Cancer Immun. 13, 5. [PMC free article] [PubMed] [Google Scholar]
  • 99. Fong L., Kwek S. S., O'Brien S., Kavanagh B., McNeel D. G., Weinberg V., Lin A. M., Rosenberg J., Ryan C. J., Rini B. I., Small E. J. (2009) Potentiating endogenous antitumor immunity to prostate cancer through combination immunotherapy with CTLA4 blockade and GM-CSF. Cancer Res. 69, 609–615 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Slovin S. F., Beer T. M., Higano C. S., Tejwani S., Hamid O., Picus J., Harzstark A., Scher H. I., Lan Z., Lowy I. (2009) Initial Phase II experience of ipilimumab (IPI) alone and in combination with radiotherapy (XRT) in patients with metastatic castration-resistant prostate cancer (mCRPC). J. Clin. Oncol. 27 (Suppl.), 15s (abs. 5138) [Google Scholar]
  • 101. Mohebtash M., Madan R. A., Arlen P. M., Rauckhorst M., Tsang K. Y., Cereda V., Vergati M., Poole D. J., Dahut W. L., Schlom J., Gulley J. L. (2009) Phase I trial of targeted therapy with PSA-TRICOM vaccine (V) and ipilimumab (ipi) in patients (pts) with metastatic castration-resistant prostate cancer (mCRPC). J. Clin. Oncol. 27 (Suppl.), 15s (abs. 5138) [Google Scholar]
  • 102. Small E. J., Tchekmedyian N. S., Rini B. I., Fong L., Lowy I., Allison J. P. (2007) A pilot trial of CTLA-4 blockade with human anti-CTLA-4 in patients with hormone-refractory prostate cancer. Clin. Cancer Res. 13, 1810–1815 [DOI] [PubMed] [Google Scholar]
  • 103. Lynch T. J., Bondarenko I. N., Luft A., Serwatowski P., Barlesi F., Chacko R. T., Sebastian M., Siegel J., Cuillerot J., Reck M. (2010) Phase II trial of ipilimumab (IPI) and paclitaxel/carboplatin (P/C) in first-line Stage IIIb/IV non-small cell lung cancer (NSCLC). J. Clin. Oncol. 28 (Suppl.), 15s (abs. 7531) [Google Scholar]
  • 104. Yang J. C., Hughes M., Kammula U., Royal R., Sherry R. M., Topalian S. L., Suri K. B., Levy C., Allen T., Mavroukakis S., Lowy I., White D. E., Rosenberg S. A. (2007) Ipilimumab (anti-CTLA4 antibody) causes regression of metastatic renal cell cancer associated with enteritis and hypophysitis. J. Immunother. 30, 825–830 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Royal R. E., Levy C., Turner K., Mathur A., Hughes M., Kammula U. S., Sherry R. M., Topalian S. L., Yang J. C., Lowy I., Rosenberg S. A. (2010) Phase 2 trial of single agent ipilimumab (anti-CTLA-4) for locally advanced or metastatic pancreatic adenocarcinoma. J. Immunother. 33, 828–833 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Curran M. A., Montalvo W., Yagita H., Allison J. P. (2010) PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors. Proc. Natl. Acad. Sci. USA 107, 4275–4280 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Mangsbo S. M., Sandin L. C., Anger K., Korman A. J., Loskog A., Totterman T. H. (2010) Enhanced tumor eradication by combining CTLA-4 or PD-1 blockade with CpG therapy. J. Immunother. 33, 225–235 [DOI] [PubMed] [Google Scholar]
  • 108. Kroemer G., Galluzzi L., Kepp O., Zitvogel L. (2013) Immunogenic cell death in cancer therapy. Annu. Rev. Immunol. 31, 51–72 [DOI] [PubMed] [Google Scholar]
  • 109. Ma Y., Conforti R., Aymeric L., Locher C., Kepp O., Kroemer G., Zitvogel L. (2011) How to improve the immunogenicity of chemotherapy and radiotherapy. Cancer Metastasis Rev. 30, 71–82 [DOI] [PubMed] [Google Scholar]
  • 110. Ma Y., Kepp O., Ghiringhelli F., Apetoh L., Aymeric L., Locher C., Tesniere A., Martins I., Ly A., Haynes N. M., Smyth M. J., Kroemer G., Zitvogel L. (2010) Chemotherapy and radiotherapy: cryptic anticancer vaccines. Semin. Immunol. 22, 113–124 [DOI] [PubMed] [Google Scholar]
  • 111. Kroemer G., Zitvogel L. (2012) Abscopal but desirable: the contribution of immune responses to the efficacy of radiotherapy. Oncoimmunology 1, 407–408 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Dewan M. Z., Galloway A. E., Kawashima N., Dewyngaert J. K., Babb J. S., Formenti S. C., Demaria S. (2009) Fractionated but not single-dose radiotherapy induces an immune-mediated abscopal effect when combined with anti-CTLA-4 antibody. Clin. Cancer Res. 15, 5379–5388 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113. Demaria S., Kawashima N., Yang A. M., Devitt M. L., Babb J. S., Allison J. P., Formenti S. C. (2005) Immune-mediated inhibition of metastases after treatment with local radiation and CTLA-4 blockade in a mouse model of breast cancer. Clin. Cancer Res. 11, 728–734 [PubMed] [Google Scholar]
  • 114. Chakravarty P. K., Alfieri A., Thomas E. K., Beri V., Tanaka K. E., Vikram B., Guha C. (1999) Flt3-ligand administration after radiation therapy prolongs survival in a murine model of metastatic lung cancer. Cancer Res. 59, 6028–6032 [PubMed] [Google Scholar]
  • 115. Postow M. A., Callahan M. K., Barker C. A., Yamada Y., Yuan J., Kitano S., Mu Z., Rasalan T., Adamow M., Ritter E., Sedrak C., Jungbluth A. A., Chua R., Yang A. S., Roman R. A., Rosner S., Benson B., Allison J. P., Lesokhin A. M., Gnjatic S., Wolchok J. D. (2012) Immunologic correlates of the abscopal effect in a patient with melanoma. N. Engl. J. Med. 366, 925–931 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Hiniker S. M., Chen D. S., Knox S. J. (2012) Abscopal effect in a patient with melanoma. N. Engl. J. Med. 366, 2035. [DOI] [PubMed] [Google Scholar]

Articles from Journal of Leukocyte Biology are provided here courtesy of The Society for Leukocyte Biology

RESOURCES