+ All Categories
Home > Documents > Immuno-oncology Combinations: A Review of Clinical Experience and Future...

Immuno-oncology Combinations: A Review of Clinical Experience and Future...

Date post: 10-Mar-2018
Category:
Upload: trannhi
View: 217 times
Download: 2 times
Share this document with a friend
12
Review Immuno-oncology Combinations: A Review of Clinical Experience and Future Prospects Scott J. Antonia 1 , James Larkin 2 , and Paolo A. Ascierto 3 Abstract Immuno-oncology is an evolving treatment modality that includes immunotherapies designed to harness the patient’s own immune system. This approach is being studied for its potential to improve long-term survival across multiple tumor types. It is now important to determine how immunotherapies may be most effectively used to achieve the best possible patient outcomes. Combining or sequencing immunotherapies that target distinct immune pathways is a logical approach, with the potential to further enhance the magnitude of the antitumor immune response over single agents. Early clinical data in patients with melanoma treated with two immune checkpoint inhibitors, ipilimumab and nivolu- mab, suggest support for this combination approach. Numerous other combination approaches are being evaluated in early-phase clinical trials; however, their clinical activity remains unknown. Clinical experience to date has shown that when combining an immuno-oncology agent with an existing therapeutic modality, it is important to determine the optimal dose, schedule, and sequence. Clin Cancer Res; 20(24); 6258–68. Ó2014 AACR. Introduction Tumors avoid immune destruction by a range of complex and often overlapping mechanisms that disrupt key compo- nents of the immune system involved in mounting an effective antitumor response (1–4). Tumors can avoid rec- ognition and elimination by the immune system by disrupt- ing antigen presentation mechanisms, either through down- regulation of MHC class I molecules or by disabling antigen- processing machinery. Alternatively, or additionally, tumors may suppress the immune system by disrupting pathways involved in controlling T-cell inhibition (checkpoint) and activation (3, 5), or by recruiting immunosuppressive cell types, such as regulatory T cells (Treg) and myeloid-derived suppressor cells (MDSC). The release of factors, including adenosine and prostaglandin E2, and the enzyme indolea- mine 2,3-dioxygenase (IDO) is another mechanism that tumors may use to suppress immune activity (3). The idea of targeting the immune system as a therapeutic approach in cancer is not new. Cytokines [interleukin-2 (IL2) and interferon-a (IFNa)] have been used for decades, pre- dominantly in patients with renal cell carcinoma (RCC) and melanoma. However, these cytokines are not target specific, and have been associated with significant toxicity and limited efficacy; these factors restrict use to healthy patients and only a select group of these patients will derive benefit (6, 7). Immuno-oncology is an evolving treatment modality that includes immunotherapies designed to target and harness the patient’s immune system directly to kill tumor cells (8, 9). Numerous strategies for overcoming tumor immune evasion are under evaluation (Table 1). Because these approaches directly target the patient’s immune sys- tem, they have the potential for activity across multiple types of cancer. Examples of immunotherapeutic appro- aches under clinical investigation include T-cell checkpoint inhibitors or agonists for T-cell–activating pathways, novel cytokines such as IL12 and IL15, therapeutic vaccines, elimination of immunosuppressive cells, and other agents and approaches designed to enhance immune cell function (Table 1; refs. 10–12). Since the approval of IL2, sipuleucel-T (a therapeutic vaccine composed of recombinant antigen protein designed to stimulate T-cell responses) and ipilimumab [a cytotoxic T-lymphocyte antigen 4 (CTLA-4) immune checkpoint inhibitor] were the first immunotherapies to be approved for patients with cancer. Sipuleucel-T was approved in 2010 for asymptomatic or minimally symptomatic metastatic castrate–resistant prostate cancer (CRPC) and ipilimumab in 2011 for unresectable or metastatic melanoma. Both agents were shown to significantly improve overall survival (OS) in phase III clinical trials (Fig. 1; refs. 13–16). Monoclonal antibodies targeting programmed death-1 (PD-1) ligand (PD-L1) interaction, another immune check- point pathway, are the most advanced in clinical develop- ment after ipilimumab and sipuleucel-T, and various agents are being tested in clinical trials across a range of tumor types. 1 Moftt Cancer Center and Research Institute, Tampa, Florida. 2 The Royal Marsden, London, United Kingdom. 3 Istituto Nazionale Tumori Fondazione G. Pascale, Naples, Italy. Note: S.J. Antonia and P.A. Ascierto share senior authorship. Corresponding Author: Scott J. Antonia, Moftt Cancer Center and Research Institute, Tampa, FL 33612. Phone: 813-745-8470; E-mail: Scott.Antonia@moftt.org doi: 10.1158/1078-0432.CCR-14-1457 Ó2014 American Association for Cancer Research. Clinical Cancer Research Clin Cancer Res; 20(24) December 15, 2014 6258 on May 13, 2018. © 2014 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from Published OnlineFirst October 23, 2014; DOI: 10.1158/1078-0432.CCR-14-1457
Transcript

Review

Immuno-oncology Combinations: A Review of ClinicalExperience and Future Prospects

Scott J. Antonia1, James Larkin2, and Paolo A. Ascierto3

AbstractImmuno-oncology is an evolving treatment modality that includes immunotherapies designed to

harness the patient’s own immune system. This approach is being studied for its potential to improve

long-term survival across multiple tumor types. It is now important to determine how immunotherapies

may be most effectively used to achieve the best possible patient outcomes. Combining or sequencing

immunotherapies that target distinct immune pathways is a logical approach, with the potential to

further enhance the magnitude of the antitumor immune response over single agents. Early clinical data

in patients with melanoma treated with two immune checkpoint inhibitors, ipilimumab and nivolu-

mab, suggest support for this combination approach. Numerous other combination approaches are

being evaluated in early-phase clinical trials; however, their clinical activity remains unknown. Clinical

experience to date has shown that when combining an immuno-oncology agent with an existing

therapeutic modality, it is important to determine the optimal dose, schedule, and sequence. Clin Cancer

Res; 20(24); 6258–68. �2014 AACR.

IntroductionTumors avoid immune destruction by a range of complex

and often overlappingmechanisms that disrupt key compo-nents of the immune system involved in mounting aneffective antitumor response (1–4). Tumors can avoid rec-ognition and elimination by the immune system by disrupt-ing antigen presentationmechanisms, either through down-regulation ofMHC class Imolecules or by disabling antigen-processingmachinery. Alternatively, or additionally, tumorsmay suppress the immune system by disrupting pathwaysinvolved in controlling T-cell inhibition (checkpoint) andactivation (3, 5), or by recruiting immunosuppressive celltypes, such as regulatory T cells (Treg) and myeloid-derivedsuppressor cells (MDSC). The release of factors, includingadenosine and prostaglandin E2, and the enzyme indolea-mine 2,3-dioxygenase (IDO) is another mechanism thattumors may use to suppress immune activity (3).

The idea of targeting the immune system as a therapeuticapproach in cancer is not new.Cytokines [interleukin-2 (IL2)and interferon-a (IFNa)] have been used for decades, pre-dominantly in patients with renal cell carcinoma (RCC) andmelanoma. However, these cytokines are not target specific,

andhavebeen associatedwith significant toxicity and limitedefficacy; these factors restrict use to healthy patients and onlya select group of these patients will derive benefit (6, 7).

Immuno-oncology is an evolving treatment modalitythat includes immunotherapies designed to target andharness the patient’s immune system directly to kill tumorcells (8, 9). Numerous strategies for overcoming tumorimmune evasion are under evaluation (Table 1). Becausethese approaches directly target the patient’s immune sys-tem, they have the potential for activity across multipletypes of cancer. Examples of immunotherapeutic appro-aches under clinical investigation include T-cell checkpointinhibitors or agonists for T-cell–activating pathways, novelcytokines such as IL12 and IL15, therapeutic vaccines,elimination of immunosuppressive cells, and other agentsand approaches designed to enhance immune cell function(Table 1; refs. 10–12).

Since the approval of IL2, sipuleucel-T (a therapeuticvaccine composedof recombinant antigenproteindesignedto stimulate T-cell responses) and ipilimumab [a cytotoxicT-lymphocyte antigen 4 (CTLA-4) immune checkpointinhibitor] were the first immunotherapies to be approvedfor patients with cancer. Sipuleucel-T was approved in 2010for asymptomatic or minimally symptomatic metastaticcastrate–resistant prostate cancer (CRPC) and ipilimumabin 2011 for unresectable or metastatic melanoma. Bothagents were shown to significantly improve overall survival(OS) in phase III clinical trials (Fig. 1; refs. 13–16).

Monoclonal antibodies targeting programmed death-1(PD-1) ligand (PD-L1) interaction, another immune check-point pathway, are the most advanced in clinical develop-ment after ipilimumab and sipuleucel-T, and various agentsarebeing tested in clinical trials across a rangeof tumor types.

1Moffitt Cancer Center and Research Institute, Tampa, Florida. 2The RoyalMarsden, London, United Kingdom. 3IstitutoNazionale Tumori FondazioneG. Pascale, Naples, Italy.

Note: S.J. Antonia and P.A. Ascierto share senior authorship.

Corresponding Author: Scott J. Antonia, Moffitt Cancer Center andResearch Institute, Tampa, FL 33612. Phone: 813-745-8470; E-mail:[email protected]

doi: 10.1158/1078-0432.CCR-14-1457

�2014 American Association for Cancer Research.

ClinicalCancer

Research

Clin Cancer Res; 20(24) December 15, 20146258

on May 13, 2018. © 2014 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst October 23, 2014; DOI: 10.1158/1078-0432.CCR-14-1457

Table 1. Potential strategies for overcoming tumor immune evasion mechanisms and examples of agentsin clinical development (12)

Treatment strategy Examples of agents in clinical development

Reversing the inhibition of adaptive immunity (blocking T-cell checkpoint pathways)

* Inhibiting the CTLA-4 checkpointmoleculea

Ipilimumab: approved for melanomaTremelimumab: phase II for malignant mesothelioma, HCC, melanoma

* Inhibiting the interaction between PD-1checkpoint and its ligandsa

Nivolumab (anti–PD-1): phase III for melanoma, NSCLC, RCC

Pembrolizumab (MK-3475; anti–PD-1): phase III for NSCLC, melanomaMPDL3280A (RG7446; anti–PD-L1): phase III for NSCLCPidilizumab (CT-011; anti–PD-1): phase II for FL, prostate, pancreatic, melanomaAMP-514 (MEDI0680; anti–PD-1): phase I for solid tumorsMEDI4736 (anti–PD-L1): phase I for solid tumorsAMP-224 (recombinant PD-L-Fc fusion protein): phase I for solid tumorsrHIgM12B7 (anti–PD-L2): phase I for melanoma

* Inhibiting the LAG-3 checkpointmolecule

IMP321: phase I for breast, RCC; phase II for melanomaBMS-986016: phase I for solid tumors

* Inhibiting the TIM-3 checkpoint No agent undergoing clinical evaluation

* Inhibiting the adenosine A2A receptor No agent undergoing clinical evaluation

Switching on adaptive immunity (promoting T-cell costimulatory receptor signaling using agonist antibodies)* Promoting CD137 signaling Urelumab: phase I for B-cell NHL, CLL, solid tumors

PF-05082566: phase I for NHL

* Enhancing OX-40 signaling MEDI6469: phase II for breast, prostate, solid tumors

* Promoting GITR signaling TRX518: phase I for melanoma, solid tumors

* Enhancing CD27 signaling CDX-1127: phase I for CD27-expressing hematologic malignancies and solid tumors* CD40 Activation CP-870,893: phase I for pancreatic, melanoma

Chi Lob 7/4: phase I for advanced malignancies* Systemic recombinant IL21administration (range of effects thatenhance immune cell function)

Denenicokin: phase II for melanoma, ovarian and phase I for RCC, NHL

* Systemic recombinant IL15administration (range of effects thatenhance immune cell function)

rhIL 15: phase I for melanoma, kidney, NSCLC, SCHN

* Systemic recombinant IL7administration (range of effects,including on T-cell development)

rhIL 7: phase II for various solid tumors

Improving the function of innate immune cells* Manipulating the activation of NK-cellinhibitory receptors (KIR)

Lirilumab: phase II for AML (maintenance); phase I for solid tumors

* Stimulating macrophages and DCs Toll-like receptor agonists:Bacillus Calmette-Gu�erin (TLR 2/4 agonist): approved for bladder carcinoma

Hiltonol (TLR7 agonist): phase II various solid and hematologic malignanciesImiquimod (TLR7 agonist): approved basal cell carcinomaResiquimod (TRL7/8 agonist): phase I/II various solid and hematologic malignanciesCpG 7909 (TLR 9 agonist): phase II various solid tumors

Activating the immune system (potentiating immune-cell effector function)* IDO inhibition INCB024360: phase II for melanoma, EOC, PPC, FTC

Indoximod: phase II for breast, prostate

(Continued on the following page)

Immuno-oncology Combinations

www.aacrjournals.org Clin Cancer Res; 20(24) December 15, 2014 6259

on May 13, 2018. © 2014 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst October 23, 2014; DOI: 10.1158/1078-0432.CCR-14-1457

One of the more exciting aspects of immunotherapies isdemonstrated with data from clinical trials for ipilimu-mab, nivolumab, and pembrolizumab that show thepotential for long-term survival. In a phase III study ofipilimumab in previously treated patients with metastaticmelanoma (study MDX010-020), the survival rate at 2and 3 years was 25% for each (17). In addition, in apooled analysis of data from 12 ipilimumab clinicalstudies with follow-up of up to 10 years in some patients,an OS plateau started at approximately 3 years and the 3-year survival rate was 22% (18). The PD-1 immunecheckpoint inhibitors nivolumab and pembrolizumabhave also shown durable responses in phase I studies(19–21).

Although targeting the immune systemhas emerged as aneffective treatment approach for patients with CRPC andmetastatic melanoma (17, 18), for the development of thistreatmentmodality to progress, it is important to determinehow agents should be used to achieve the best possiblepatient outcomes.Combining immunotherapieswithotherestablished and investigational cancer therapies is a field ofactive investigation, with a multitude of approaches underconsideration. This review focuses on (i) combining orsequencing immunotherapies that target distinct immunepathways, particularly T-cell checkpoints, and (ii) combin-ing immunotherapies with existing therapeutic modalities,specifically BRAF-targeted therapies, chemotherapies, andradiotherapy.

Table 1. Potential strategies for overcoming tumor immune evasion mechanisms and examples of agentsin clinical development (12) (Cont'd )

Treatment strategy Examples of agents in clinical development

* Inhibition of TGF-b signaling GC1008; phase I for melanoma, RCCLY2157299: phase I/II various solid tumors

TEW 7197: phase I solid tumorsIMC-TR1 (LY3022859): phase I solid tumors

* Systemic IL2 or IFNa administrationa Agents approved

* Various vaccine-based strategiesa Various approaches under clinical evaluation

Abbreviations: ADCC, antibody-dependent cell-mediated cytotoxicity; AML, acute myeloid leukemia; CLL, chronic lymphocyticleukemia; EOC, epithelial ovarian cancer; FL, follicular lymphoma; FTC, fallopian tube cancer; GITR, glucocorticoid-induced tumornecrosis factor related gene; HCC, hepatocellular carcinoma; NHL, non-Hodgkin lymphoma; PPC, primary peritoneal cancer; SCHN,squamous cell head and neck cancer; TIM, T-cell immunoglobulin mucin; TLR, Toll-like receptor.aApproaches in which approved compounds or investigational compounds are being studied in phase III trials.

CCR Reviews

0 3 6 9 12

1-year survival

of 82%

95% CI

(69.0%–94.4%)

1 mg/kg nivolumab + 3 mg/kg ipilimumab

All concurrent regimen

2/17

n = 53

n = 17

9/53

15 18 21 24 27 30 33 36

0.0

0.1

0.2

0.3

0.4

0.5

0.6

0.7

0.8

0.9

1.0

Months

17 16 16 14 10 5 3 2 2 1 0 0 0

53 47 36 29 19 10 7 4 4 3 1 1 0

1 mg + 3 mg

All concurrent

Patients at risk

Censored Died/treatedPro

po

rtio

n o

f su

rviv

al

Figure 1. Targeting two distinctimmune checkpoint pathways:interim data from a phase I study ofconcurrent ipilimumab andnivolumab. Patients with advancedmelanoma treated with ipilimumabin combination with nivolumab hada preliminary 1-year OS rate of82%. These data provided therationale for initiation of a phase IIItrial of the ipilimumab/nivolumabcombination in previouslyuntreated patients with metastaticmelanoma. Reprinted withpermission fromWolchok et al. (16).

Antonia et al.

Clin Cancer Res; 20(24) December 15, 2014 Clinical Cancer Research6260

on May 13, 2018. © 2014 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst October 23, 2014; DOI: 10.1158/1078-0432.CCR-14-1457

Combining immunotherapies that target distinctimmune pathwaysCombining or sequencing immunotherapies that

target distinct immune pathways is a rational strategy todetermine whether the magnitude of the antitumorimmune response may be improved over that generatedwith a single agent. Potential combination approachesundergoing clinical evaluation include dual T-cell check-point inhibition, T-cell checkpoint inhibition combinedwith immunomodulatory antibodies designed to enhanceT-cell activity through agonistic interaction with costimu-latory receptors (aiming to switch on adaptive immunity),T-cell checkpoint inhibition combined with approaches toimprove the function of innate immune cells, and T-cellcheckpoint inhibition combined with other approaches toenhance the immune response (Table 2; refs. 3, 5, 12).

Dual T-cell checkpoint inhibitionGiven that T-cell checkpoint inhibitors (e.g., ipilimu-

mab, nivolumab, pembrolizumab) have shown single-agent clinical activity in several tumor types (5, 13),and preclinical data suggest checkpoint molecules mayact synergistically to regulate T-cell function and pro-mote tumor immune escape, it is rational to evaluatewhether combining checkpoint inhibitors improvesactivity, achieving an OS benefit in a greater proportionof patients compared with either agent alone (Fig. 2;refs. (16, 22–24). Initial support for dual T-cell check-point inhibition has come from a phase I study inwhich patients with advanced stage III or IV melanomawere treated with both ipilimumab (1 or 3 mg/kg) andnivolumab (0.3 mg/kg, 1 mg/kg, or 3 mg/kg) in aconcurrent or sequenced regimen (16, 25). An objectiveresponse rate (ORR) rate of 40% was achieved inpatients treated with the concurrent regimen (ORR was53% at the maximum tolerated dose, nivolumab 1 mg/kg, ipilimumab 3 mg/kg). The preliminary 1-year OSrate with the concurrent regimen was 82% [95% con-fidence interval (CI) 69.0–94.4; Fig. 1; ref. 16]. Thesepromising results prompted the initiation of a phase IIIstudy (CheckMate 067) to further evaluate concurrenttreatment with ipilimumab and nivolumab (12).Phase I studies are in progress to evaluate ipilimumab

plus nivolumab in patients with a range of solid tumors[including RCC, non–small cell lung cancer (NSCLC),colon cancer, triple-negative breast cancer, gastric cancer,pancreatic cancer, and small-cell lung cancer (SCLC); ipi-limumab plus pembrolizumab (anti–PD-1) in patientswith melanoma, RCC, and NSCLC; tremelimumab (ananti–CTLA-4 agent) plus MEDI 4736 (anti–PD-L1 agent)in NSCLC; and an anti-lymphocyte activation gene 3 (LAG-3) monoclonal antibody BMS-986016 plusnivolumab(anti–PD-1) in patients with solid tumors (Table 2;ref. 12)]. The latter combination is supported by preclinicaldata that showed strong synergistic antitumor activity whenboth the PD-1 and LAG-3 immune checkpoint pathwayswere blocked (23). Dual anti–LAG-3/anti–PD-1 antibodytreatment curedmost mice of established fibrosarcoma and

colon adenocarcinoma tumors that were largely resistant tosingle antibody treatment (22).

T-cell checkpoint inhibition combined with agonisticantibodies against T-cell costimulatory receptors

In theory, if agents designed to release the checkpoint-mediated inhibition of T cells were combined with agonistantibodies designed to enhance costimulatory T-cell signal-ing, a more effective immune response may be generated(5). To date, no data are available from clinical trialsevaluating these combinations, but studies are in progress.CD40 plays a key role in the development of T-cell–depen-dent antitumor immunity, and is essential in enablingantigen-presenting cells to process and present antigeneffectively to T cells (26–28). Combining T-cell checkpointblockade (using anti–CTLA-4 agent tremelimumab) withan agent that targets the costimulatory molecule CD40(CP-870,893) is being investigated in a phase I trial inpatients with melanoma (12).

Other agonist antibodies designed to target receptors,including OX-40, CD27, GITR, and CD137, are in devel-opment. The clinical evaluation of these agents as mono-therapy is at an early stage, although the limited dataavailable suggest they canbe safety administered topatients.Data from a large phase I trial with urelumab (anti-CD137)in more than 100 patients did show liver toxicity, with2 deaths reported at higher doses. Clinical evaluation ofurelumab is continuing at lower doses in advanced solidtumors and hematologic malignancies (29). Evaluatingcombinations of these antibodies with checkpoint inhibi-tors and other immunotherapies is an exciting possibility,but one that should be evaluated with caution.

Another agonistic therapeutic approach that was evalu-ated with catastrophic effects was the CD28 agonistTGN1412. In a first-in-human phase I trial, TGN1412administration resulted in a cytokine storm that causedsevere adverse events in the six volunteers (30). As explainedbyCurran and colleagues (31), CD28 iswidely expressed onall mature T-cell populations; therefore, an agonistic CD28antibody may be expected to have a polyclonal "superagonist" effect—this is in contrast to other costimulatorymodules, such as CD137 or OX-40, which are onlyexpressed on a proportion of T cells, so agonist antibodiesare likely to have a more selective effect.

T-cell checkpoint inhibition combined withapproaches to improve the function of innate immunecells

Adaptive immune responses to cancer involve variouscomponents of innate immunity. In viewof this, combiningtherapies designed to enhance T-cell function with agentsdesigned to improve innate immune cell function areworthy of evaluation. Natural killer (NK) cells are innateeffector cells that maintain tolerance to self-tissue via theexpression of killer cell immunoglobulin-like receptors(KIR), which negatively regulate NK-cell activity by bindingto the MHC class I molecules expressed on most "normal"cells (32–35). Tumor cells may appear like normal cells by

Immuno-oncology Combinations

www.aacrjournals.org Clin Cancer Res; 20(24) December 15, 2014 6261

on May 13, 2018. © 2014 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst October 23, 2014; DOI: 10.1158/1078-0432.CCR-14-1457

Table 2. Combination approaches in clinical development (12)

Selected clinical trials of immunotherapies

Drugs Target Development phase/tumor type

Dual T-cell checkpoint blockadeIpilimumab þ nivolumab CTLA-4 þ PD-1 Phase III: melanoma; phase I/II: RCC, colon, NSCLC, triple-negative

breast cancer, gastric cancer pancreatic cancer, and SCLCIpilimumab þ pembrolizumab CTLA-4 þ PD-1 Phase I: melanoma, RCC, and NSCLCTremelimumab þ MEDI4736 CTLA-4 þ PD-L1 Phase I: NSCLC, solid tumorsNivolumab þ BMS-986016 PD-1 þ LAG-3 Phase I: solid tumors

T-cell blockade þ costimulatory receptor agonistsCP-870,893 þ tremelimumab CTLA -4 þ CD40 Phase I: metastatic melanoma

T-cell blockade þ improving the function of innate immune cellsLirilumab þ ipilimumab CTLA-4 þ KIR Phase I: solid tumorsLirilumab þ nivolumab PD-1 þ KIR Phase I: solid tumors

T-cell blockade þ other immune system activatorsDenenicokin þ ipilimumab CTLA-4 þ IL21 Phase I: melanomaDenenicokin þ nivolumab PD-1 þ IL21 Phase I: solid tumorsINCB024360 þ ipilimumab CTLA-4 þ IDO Phase I: melanomaIndoximod þ sipuleucel-T IDO þ vaccine Phase II: prostateNivolumab þ gp100, NY-ESO-1 PD-1 þ vaccine Phase I: melanomaIpilimumab þ sipuleucel-T CTLA-4 þ vaccine Phase II: prostate cancerIpilimumab þ TriMix-DC CTLA-4 þ vaccine Phase II: melanomaIpilimumab þ NY-ESO-1 vaccine CTLA-4 þ vaccine Phase I: melanomaIpilimumab þ adoptive cell transfer CTLA-4 þ passive

immunotherapyPhase I/II: melanoma

Selected clinical trials of immunotherapies (excluding ipilimumab) in combination with other treatment modalities

Drugs Target Combination treatmentmodality

Development phase/tumortype

Tremelimumab CTLA-4 Gefitinib Phase I: NSCLCNivolumab PD-1 Chemotherapya Phase I: NSCLC

Dasatinib Phase I: CMLBevacizumab Phase I: NSCLCErlotinib Phase I: NSCLCSunitinib or pazopanib Phase II: RCC

Pembrolizumab PD-1 Pazopanib Phase I: RCCLenalidomide þdexamethasone

Phase I: multiple myeloma

MPDL3280A PD-L1 Bevcizumab Phase II: RCCErlotinib Phase I: NSCLCVemurafenib Phase I: melanoma

MEDI14736 PD-L1 Trametinib � dabrafenib Phase I/II: melanomaPidilizumab PD-1 Rituximab Phase II: FL

Gemcitabine Phase II: pancreaticFOLFOX Phase II: CRC (completed)Sipuleucil-T þcyclophosphamide

Phase II: prostate cancer

IMP321 LAG-3 Paclitaxel Phase I: breast (completed)Urelumab CD137 Rituximab Phase I: NHL

Chemotherapy Phase I: solid tumorsPF-05082566 CD137 Rituximab Phase I: NHLCP-870,893 CD40 Paclitaxel/carboplatin Phase I: solid tumors

(completed)Denenicokin IL21 Sunitinib Phase I/II: RCC

Sorafenib Phase I/II: RCCRituximab Phase I: NHL

Indoximod IDO Docetaxel Phase II: breast

Abbreviations: CML, chronic myeloid leukemia; CRC, colorectal cancer; FL, follicular lymphoma; NHL, non-Hodgkin lymphoma.aChemotherapy regimens include gemcitabine/cisplatin, pemetrexed/cisplatin, and carboplatin/paclitaxel.

Antonia et al.

Clin Cancer Res; 20(24) December 15, 2014 Clinical Cancer Research6262

on May 13, 2018. © 2014 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst October 23, 2014; DOI: 10.1158/1078-0432.CCR-14-1457

retaining or upregulating MHC class I to escape immuno-surveillance by NK cells (33). Lirilumab is an anti-KIRantibody that blocks the inhibitory KIR signal, therebypotentiating NK-cell killing of tumor cells, despite expres-sion of MHC I. A regimen designed to enhance innate andadaptive immunity, respectively, could theoreticallyachieve more favorable biologic and clinical activity com-pared with either agent alone (36, 37). This could beachieved in a variety ways, such as by using an anti-KIRagent (lirilumab) in combination with PD-1 or CTLA-4immune checkpoint inhibitors. Clinical trials are underway evaluating such combinations (Table 2; ref. 12).

Other immunotherapy combination partnersCytokine therapy. Cytokines have the capacity to stim-

ulate an immune response, although arguably less specif-ically comparedwithother immunotherapeutic approaches(3). IL21 has a role inNK and T-cell activation, and systemicadministration of a recombinant IL21 (rIL21) has demon-strated antitumor activity in tumors, including metastaticmelanoma (38). On the basis of preclinical studies inmouse tumor models which showed enhanced antitumoractivity when rIL21 was combined with either anti–CTLA-4or anti–PD-1 agents (39), phase I dose-escalation studiesare evaluating these combinations in patients withadvanced or metastatic melanoma (ipilimumab; ref. 40)or solid tumors (nivolumab; ref. 41).Other cytokines are under evaluation asmonotherapy for

cancer therapy, but a phase II trial is ongoing with IL7,which has a wide range of biologic activities, including arole in T-cell development, after standard therapy withsipuleucel-T for patients with asymptomatic or minimallysymptomatic metastatic CRPC (12).IDO inhibition. IDO is an immunosuppressive enzyme

that is involved in maintaining peripheral immune toler-ance by suppressing the function of both innate and adap-

tive immune cells. Data frompreclinical studies suggest thatinhibiting IDO can promote the proliferation, survival, andfunction of various immune cells [e.g., T cells, NK cells, anddendritic cells (DC)], reduce the generation of Tregs, andsignificantly inhibit tumor growth (42, 43). Furthermore,studies in murine models showed that host-derived IDOcan suppress the antitumor activity of an anti–CTLA-4antibody. However, inhibition or absence of IDO com-bined with therapies targeting immune checkpoints, suchas CTLA-4, PD-1/PD-L1, and GITR, acts synergistically tocontrol tumor growth and improve OS (44). Thus, com-bining an agent that inhibits IDO with another immuno-therapywould appear to be a rational approach and is beingevaluated in several clinical trials (Table 2). A phase II trial isevaluating the IDO inhibitor indoximod in combinationwith the therapeutic vaccine sipuleucel-T in patients withprostate cancer (Table 2; ref. 12).

Adoptive cell transfer and T-cell engineering. Adoptivecell transfer (ACT) involves the collection of tumor-infil-trating lymphocytes (TIL) from patients, the in vitro expan-sion of autologous lymphocytes with reactivity to tumorantigens, and the subsequent transfer back to the patient,with the expectation that the tumor-specific lymphocyteswill attack the tumor (11, 45). ACT has demonstrateddurable complete responses in patients with melanoma(45). In a phase II study, 20 of 93 patients with metastaticmelanoma (22%) had durable, complete remissions(>3–7 years) after treatment with IL2 and ACT of TILs.

In addition to the expansion and transfer of TILs,approaches to modify the patient’s T cells are underevaluation. These include engineering T cells using chi-meric antigen receptors (CAR) to redirect them to specifictumor-antigen targets before reinfusion. T-cell receptor(TCR) gene therapy is another strategy in development;the objective is to induce immune reactivity againsttumors by introducing genes encoding a tumor-reactiveTCR into patients’ T cells, improving immune reactivity.Combining these types of approaches with other immu-notherapies may further improve clinical efficacy. Trialsof ACT, CARs, and TCR gene therapy in combination withimmune checkpoint inhibitors or other approaches areongoing or under consideration.

Therapeutic vaccines. Although the variousmechanismsof actionof therapeutic vaccines are beyond the scopeof thisreview (46, 47), most vaccines are designed to (i) presenttumor antigens to the immune system and (ii) provideimmune modulation. Because of their differing mechan-isms of action, vaccines and other immunotherapies arepotential combination partners. Clinical data have shownpromising results with some combinations, for example,gp100 peptide vaccine and IL2 in melanoma, and ipilimu-mab combined with granulocyte macrophage-colony stim-ulating factor cell-based vaccine in pancreatic cancer (48,49). However, no survival advantage was seen in patientswithmelanoma treated with gp100 plus ipilimumab versusthose given ipilimumab alone in a phase III trial (13, 17).

Various phase I and II clinical trials combining a vaccinewith a checkpoint inhibitor are ongoing in patients with

CCR Reviews

Combination

PD-1 pathway blockade

CTLA-4 pathway blockade

0 1 2 3 4

0

20

40

60

80

100

Years

Per

cent

age

aliv

e

Figure 2. Hypothetical effect on OS of blocking two T-cell checkpointpathways. Adapted with permission from Urba (24).

Immuno-oncology Combinations

www.aacrjournals.org Clin Cancer Res; 20(24) December 15, 2014 6263

on May 13, 2018. © 2014 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst October 23, 2014; DOI: 10.1158/1078-0432.CCR-14-1457

melanoma or prostate cancer (Table 2; ref. 12). However, aclear demonstration of the vaccine’s ability to induce clin-ically relevant antitumor responses in patients is required,as historically, the clinical translation of cancer vaccinesinto efficacious therapies has been challenging (with theexception of sipuleucel-T, the only approved therapeuticcancer vaccine; ref. 47). Data suggest that T cells activated atthe vaccine site are "shut down" when they enter the tumormicroenvironment, most likely due to tumor-mediated T-cell–suppressive mechanisms (50, 51). With tools such asPD-1 immune checkpoint inhibitors that are designed toblock tumor-mediated T-cell suppression in the tumormicroenvironment, it is worth evaluating whether vaccinesmay improve clinical efficacy when combined with a check-point inhibitor. However, data from the only publishedvaccine/PD-1 checkpoint inhibitor study showed the addi-tion of a vaccine did not improve the efficacy of PD-1inhibition (52).

Integrating immunotherapies with existingtherapeutic modalities

Existing treatment modalities, (e.g., chemotherapy, radio-therapy, and molecularly targeted therapies) cause tumorreduction, not only through cytotoxic/cytostatic effects, butalso through mechanisms that may potentiate immuneactivity, including modification of the tumor microenviron-ment and release of tumor antigens. This activity may becomplementary, even synergistic, to the immunotherapiesdesigned to support an antitumor immune response.

The immune effects of chemotherapy and radiotherapyare widely recognized and reviewed elsewhere (53–62).Immune potentiatingmechanisms include release of tumorantigens for immune presentation, depletion of immuno-suppressive cells (e.g., MDSCs, Tregs), activation ofimmune effectors (NK cells, DCs, B cells, conventionaleffector T cells), and sensitization of tumor cells to lysis.

Targeted therapies may also sensitize tumor cells toimmune-mediated killing by a variety of mechanisms.These have been reviewed by Vanneman and colleagues(58), and includepromoting effectiveDCmaturation, T-cellpriming, activation, and differentiation into long-livedmemory T cells, increasing expression of death receptorsor "distress" ligands, reducing expression of prosurvivalsignals, abrogating the production of tumorigenic inflam-mation, and inhibiting immunosuppressive cell types (63).BRAF inhibitorsmay also increase TILs and enhance antigenpresentation (64, 65). Interestingly, while the BRAF inhi-bitors have a potentiating effect on the immune system,MEK inhibitors have a possible reverse effect, reducing thesecretion of cytokines (66) and reducing the activity ofT lymphocytes (65) and DCs (67).

Clinical experience and considerations in combiningnovel immunotherapies with existing treatmentmodalities

Ipilimumab is the most widely studied combinationpartner for existing treatmentmodalities, anddata highlightthe need for careful consideration in the choice of combi-

nation partner and approach to treatment. Preliminary datafor ipilimumab in combination with chemotherapy, radio-therapy, and targeted therapy with BRAF inhibitors, arediscussed below, alongside data with other immuno-therapies. Table 2 provides a summary of ongoing clinicaltrials with immunotherapies (excluding ipilimumab) incombination studies with chemotherapy, radiotherapy,and targeted therapies (12).

Chemotherapy combinations. Ipilimumab has shownpromising results when combined with chemotherapy inpatients with melanoma and lung cancer; however, dataindicate that careful consideration of the combinationapproach is going to be important in regard to tolerabilityand optimizing patient outcomes.

Patients with previously untreated melanoma whoreceived ipilimumab (10mg/kg) plus chemotherapy (dacar-bazine) had significantly improvedOS comparedwith thosewho received chemotherapy alone (11.2 months vs. 9.1months; ref. 15). However, the benefit of the combinationrelative to ipilimumab alone remains unclear, as there wasnot an ipilimumab-alone arm in the trial. The combinationwas also less well tolerated compared with dacarbazinealone. Grade 3 or 4 adverse events occurred in 56.3% ofpatients treated with ipilimumab/dacarbazine comparedwith 27.5% treated with dacarbazine/placebo (P < 0.001;ref. 15). Similarly, data from a three-arm, phase I studyshowed that ipilimumab could be safety combined witheither dacarbazine or carboplatin/paclitaxel in patients withmelanoma (68).

Combining ipilimumab with paclitaxel and carbopla-tin significantly improved immune-related progression-free survival (irPFS) compared with chemotherapy alonein a phase II study in patients with NSCLC and extensive-disease SCLC (69, 70). However, the improvement inirPFS was only evident when the drugs were given on aphased schedule (e.g., two doses of placebo plus pacli-taxel/carboplatin followed by four doses of ipilimumabplus paclitaxel/carboplatin), not when they were givenconcurrently. Phased ipilimumab, concurrent ipilimu-mab, and control, respectively, were associated withmedian irPFS of 5.7, 5.5, and 4.6 months in patientswith NSCLC, and 6.4, 5.7, and 5.3 months in patientswith SCLC. The overall incidence of treatment-relatedgrade 3/4 adverse events was similar across the arms, andipilimumab did not appear to exacerbate the adverseevents associated with chemotherapy (69, 70). Ongoingtrials are further evaluating ipilimumab/chemotherapycombinations in melanoma and lung cancer, as well asin various other solid tumors, and will hopefully provideinformation about how best to combine these treatmentmodalities.

Nivolumab is being investigated in combination with avariety of agents in a large phase I trial (CheckMate 012,NCT01454102) in chemotherapy-na€�ve patients withNSCLC. Treatment arms include nivolumab monotherapyand nivolumab in combination with three, platinum-baseddoublet chemotherapy regimens, bevacizumab given afterat least four cycles of platinum doublet chemotherapy, and

Antonia et al.

Clin Cancer Res; 20(24) December 15, 2014 Clinical Cancer Research6264

on May 13, 2018. © 2014 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst October 23, 2014; DOI: 10.1158/1078-0432.CCR-14-1457

erlotinib (EGFR-mutation positive nonsquamous NSCLCpatients). Preliminary data indicate that nivolumab plusplatinum-based chemotherapy has a manageable safetyprofile with no drug-related deaths reported so far. Objec-tive responses have been observed in each arm, and 1-yearOS rates ranged from 50% to 87% (71).Radiotherapy combinations. Ipilimumab has been eval-

uated in combination with radiotherapy in patients withmetastatic CRPC and melanoma. Promising activity withmanageable tolerability was observed in a phase I/II trialin patients with CRPC who had progressed after antian-drogen therapy (72); however, results from a phase IIItrial showed no significant improvement in OS with theaddition of ipilimumab to radiotherapy in post-docetaxelCRPC. A subgroup analysis did suggest benefit forpatients with less advanced disease (73). An analysis ofclinical data from 21 patients with advanced melanomawho had received radiotherapy after ipilimumab progres-sion on the Italian Expanded Access Program indicatedthat radiotherapy after ipilimumab treatment may furtherpotentiate its effect (74). A local response to radiotherapywas detected in 13 patients (62%), while 8 patients (38%)did not show any local regression. The median OS for all21 patients was 13 months (range 6–26). Eleven (85%) of13 patients with local response showed an abscopaleffect, suggesting that local response to radiotherapy maybe predictive for the abscopal response and outcome. Themedian OS for patients with and without abscopalresponses was, respectively, of 22.4 months (range 2.5–50.3) and 8.3 months (range 7.6–9.0). There are nowover 15 clinical trials alone in progress to evaluate ipili-mumab plus radiotherapy.Initial data from a phase I trial of MPDL3280A, an anti–

PD-L1 monoclonal antibody, in combination with localradiotherapy showed evidence of activity in the five patientstreated (75). Overall, case reports and data from severalsmall clinical studies showing successful, sometimes dra-matic, outcomes with radiotherapy/immunotherapy com-binations in patients with melanoma provide additionalsupport for further evaluation; these are comprehensivelydiscussed by Barker and Postow (76).Targeted therapy combinations. Clinical data are lim-

ited on the efficacy of combining ipilimumab with tar-geted agents, although numerous trials are ongoing,particularly in melanoma, where three targeted therapiesare now approved in the United States for patients withmelanoma and mutated BRAF (dabrafenib, vemurafenib,and trametinib).Immunotherapy and BRAF inhibitor combinations are

extensively reviewed by Hu-Lieskovan and colleagues (77).Some data indicate that the sequencing of BRAF inhibitorsand ipilimumab has a marked effect on the efficacy andtolerability of the combination in patients with BRAF-mutant melanoma, and indicate that the drugs should besequenced (78–80). Data from a recent retrospective anal-ysis of a cohort of patients treatedwith immunotherapy andthen a BRAF inhibitor (with or without a MEK inhibitor)showed prior immunotherapy did not appear to have an

adverse effect on response to a BRAF inhibitor. However,outcomes were poor when ipilimumab was given afterBRAF inhibitor discontinuation (81).More data are needed,but there is some rationale to use either agent first in asequencing approach, depending on the disease kinetics. Inmore rapid progressors, a BRAF inhibitor may be used firstto reduce tumor load followed by ipilimumab tomaintain aresponse; in patients with more indolent disease, ipilimu-mab may be given first followed by vemurafenib to reducetumor burden (78).

In a phase I trial, concurrent administration of vemur-afenib and ipilimumab at the approved monotherapydoses or with a lower dose of vemurafenib resulted inhepatotoxicity that was greater than expected for eitheragent alone (80). These safety analyses demonstrate therisk of using vemurafenib and ipilimumab concurrently,and these drugs should not be used in combinationoutside of a clinical trial. Ongoing studies are evaluatingthe optimal sequence of these agents in patients withBRAF-mutant metastatic melanoma. Severe cutaneousand neurologic toxicity has also been reported in twopatients with melanoma during therapy with vemurafe-nib after receiving treatment with a PD-1 immune check-point inhibitor (nivolumab or pembrolizumab; ref. 82).It is also noteworthy that dose-limiting toxicities havebeen observed in patients with RCC treated with thetargeted agent sunitinib and either rhIL21 (hematologictoxicity) or the anti–CTLA-4 agent tremelimumab (renalfailure), further emphasizing the need for caution whenevaluating combinations (83, 84).

RCC is a tumor for which combining immunotherapyand targeted therapy is of substantial interest. Preliminarydata from aphase II trial of nivolumab in combinationwithpazopanib or sunitinib in patients with metastatic RCCshowed evidence of activity with ORRs of 45% and 52%,respectively, and amanageable safety profile (85). This trialand others evaluating various combinations in RCCcontinue.

The anti-CD137 agents urelumab and PF-05082566 areboth in phase I trials in combination with rituximab inpatients with non-Hodgkin lymphoma (Table 2). Clinicalstudy of these agents with rituximab is based on preclin-ical data that have shown enhanced tumor regressionwhen an anti-CD137 agent was used after a therapeuticmonoclonal antibody (86, 87). The anti-CD137 antibodyis proposed to enhance rituximab-dependent cytotoxicitythrough antigen-dependent cell-mediated cytotoxicity(86). Recent preclinical data showing enhanced antilym-phoma activity with rituximab combined with KIR block-ade (lirilumab) also support clinical investigation of thiscombination (34).

ConclusionsImmuno-oncology is an evolving treatment modality,

with agents being studied for their potential to providelong-term survival across a broad range of tumor types,and for their synergistic activity when combined with othertreatmentmodalities. It is important now to determine how

www.aacrjournals.org Clin Cancer Res; 20(24) December 15, 2014 6265

Immuno-oncology Combinations

on May 13, 2018. © 2014 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst October 23, 2014; DOI: 10.1158/1078-0432.CCR-14-1457

to advance this field and how to use these new immu-notherapies most effectively to achieve the best patientoutcomes. Areas of investigation are broad, and includecombining or sequencing immunotherapies that targetdistinct immune pathways, combining or sequencing animmunotherapeutic agent with existing treatment modal-ities, and determining the optimal schedule of therapies incombination regimens. At present, it is difficult to identifythe best combination approaches to pursue given the lim-ited data and the somewhat unexpected occurrence oftoxicity with some combinations (e.g., ipilimumab andvemurafenib). Future data from preliminary clinical studieswill help to direct research.

Combining immunotherapies has the potential to over-comemore than one of the barriers that tumor cells developto evade the immune system, and may provide an OSbenefit in a greater portion of patients comparedwith eitheragent alone (Fig. 1). However, the ideal sequence, schedule,and combination of immunotherapies need to be deter-mined. Likewise, it is important to determine optimal dose,schedule, and sequence when combining an immunother-apywith radiotherapy, chemotherapy, or targeted agents, asthese therapies all have different mechanisms of action. Afinal consideration for combining immunotherapieswill beto identify the regimens with the best risk–benefit profile.We can expect improvements in overall clinical efficacy asnew agents targeting alternative or overlapping tumor-asso-ciated immunosuppressive mechanisms are developed andused in combination or sequentially.

Disclosure of Potential Conflicts of InterestS.J. Antonia is a consultant/advisory board member for Bristol-Myers

Squibb and MedImmune/AstraZeneca. J. Larkin reports receiving com-mercial research grants from Bristol-Myers Squibb, Novartis, and Pfizer,and is a consultant/advisory board member for Bristol-Myers Squibb,GlaxoSmithKline, Merck, Novartis, Pfizer, and Roche/Genentech. (Note:all consultancy/advisory board membership was uncompensated after2012.) P.A. Ascierto reports receiving commercial research grants fromBristol-Myers Squibb, Merck, Roche/Genentech, and Ventana; speakersbureau honoraria from Bristol-Myers Squibb, GlaxoSmithKline, andRoche/Genentech; and is a consultant/advisory board member for Bris-tol-Myers Squibb, GlaxoSmithKline, Merck, Novartis, Roche/Genentech,and Ventana. No other potential conflicts of interest were disclosed.

Authors' ContributionsConception and design: J. Larkin, P.A. AsciertoDevelopment of methodology: J. Larkin, P.A. AsciertoAcquisitionofdata (provided animals, acquired andmanagedpatients,provided facilities, etc.): P.A. AsciertoAnalysis and interpretation of data (e.g., statistical analysis, biosta-tistics, computational analysis): J. Larkin, P.A. AsciertoWriting, review, and/or revision of the manuscript: S.J. Antonia,J. Larkin, P.A. AsciertoAdministrative, technical, or material support (i.e., reporting or orga-nizing data, constructing databases): P.A. AsciertoStudy supervision: P.A. Ascierto

AcknowledgmentsThe authors thank Rebecca Turner of StemScientific, a health care com-

munications firm funded by Bristol–Myers Squibb, for providingwriting andeditorial support.

Grant SupportJ. Larkin is supported by the NIH Research Royal Marsden Hospital/

Institute of Cancer Research Biomedical Research Centre for Cancer.

Received July 2, 2014; revised September 17, 2014; accepted October 4,2014; published OnlineFirst October 23, 2014.

References1. Drake CG, Jaffee E, Pardoll DM. Mechanisms of immune evasion by

tumors. Adv Immunol 2006;90:51–81.2. Vesely MD, Kershaw MH, Schreiber RD, Smyth MJ. Natural innate

and adaptive immunity to cancer. Annu Rev Immunol 2011;29:235–71.

3. Mellman I, Coukos G, Dranoff G. Cancer Immunotherapy comes ofage. Nature 2011;480:21–9.

4. Kerkar SP, Restifo NP. Cellular constituents of immune escape withinthe tumor microenvironment. Cancer Res 2012;72:3125–30.

5. Pardoll DM. The blockade of immune checkpoints in cancer immu-notherapy. Nat Rev Cancer 2012;2:252–64.

6. Amin A, White RJ Jr. High-dose interleukin-2: is it still indicated formelanoma and RCC in an era of targeted therapies? Oncology2013;27:680–91.

7. Antony GK, Dudek AZ. Interleukin 2 in cancer therapy. Curr MedChem2010;17:3297–302.

8. Finn OJ. Immuno-oncology: understanding the function and dysfunc-tion of the immune system in cancer. Ann Oncol 2012;23:viii6–9.

9. Eggermont A. Can immuno-oncology offer a truly pan-tumourapproach to therapy? Ann Oncol 2012;23:viii53–7.

10. Brody J, Kohrt H, Marabelle A, Levy R. Active and passive immuno-therapy for lymphoma: proving principles and improving results. J ClinOncol 2011;29:1864–75.

11. Smits EL, Berneman ZN, Van Tendeloo VF. Immunotherapy ofacute myeloid leukemia: current approaches. Oncologist 2009;14:240–52.

12. ClinicalTrials.gov [database on the internet]. Bethesda (MD): The NIH.c2000 - [cited 2014 Jul 2]. Available from: www.clinicaltrials.gov.

13. Hodi FS, O'Day SJ, McDermott DF, Weber RW, Sosman JA, HaanenJB, et al. Improved survival with ipilimumab in patients with metastaticmelanoma. N Engl J Med 2010;363:711–23.

14. Kantoff PW, Higano CS, Shore ND, Berger ER, Small EJ, Penson DF,et al. Sipuleucel-T immunotherapy for castration-resistant prostatecancer. N Engl J Med 2010;363:411–22.

15. Robert C, Thomas L, Bondarenko I, O'DayS,Weber JW,GarbeC, et al.Ipilimumab plus dacarbazine for previously untreated metastatic mel-anoma. N Engl J Med 2011;364:2517–26.

16. Wolchok J, Kluger HM, Callahan MK, Postow MA, Gordon RA, SegalNH, et al. Safety and clinical activity of nivolumab (anti-PD-1, BMS-936558, ONO-4538) in combination with ipilimumab in patients (pts)with advanced melanoma (MEL). J Clin Oncol 31, 2013 (suppl; abstr9012).

17. McDermott D, Haanen J, Chen TT, Lorigan P, O'Day S ;MDX010)–(20 Investigators. Efficacy and safety of ipilimumab in meta-static melanoma patients surviving more than 2 years followingtreatment in a phase III trial (MDX010)–(20). Ann Oncol 2013;24:2694–8.

18. Schadendorf D, Hodi FS, Robert C, Weber JS, Margolin K,Hamid O, et al. Pooled analysis of long-term survival datafrom phase II and phase III trials of ipilimumab in metastatic orlocally advanced, unresectable melanoma [abstract].In: Proceedings of the European Cancer Congress 2013(ECCO-ESMO-ESTRO); 2013 Sept 27–Oct 1; Amsterdam, theNetherlands. Brussels, Belgium: ECCO; 2013. Abstract nr24LBA.

19. Topalian SL, Sznol M, McDermott DF, Kluger HM, Carvajal RD, Sharf-manWH, et al. Survival, durable tumor remission, and long-term safetyin patientswith advancedmelanoma receiving nivolumab. JClinOncol2014;32:1020–30.

20. Hamid O, Robert C, Daud A, Hodi FS, HwuWJ, Kefford R, et al. Safetyand tumor responses with lambrolizumab (anti-PD-1) in melanoma.N Engl J Med 2013;369:134–44.

Clin Cancer Res; 20(24) December 15, 2014 Clinical Cancer Research6266

Antonia et al.

on May 13, 2018. © 2014 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst October 23, 2014; DOI: 10.1158/1078-0432.CCR-14-1457

21. Ribas A, Hodi FS, Kefford R, Hamid O, Daud A, Wolchok JD, et al.Efficacy and safety of the anti-PD-1 monoclonal antibody MK-3475 in411 patients (pts) withmelanoma (MEL). J Clin Oncol 2014;32:5s, 2014(suppl; abstr LBA9000).

22. Drake CG. Combination immunotherapy approaches. Ann Oncol2012;23:viii41–6.

23. WooSR, TumisME,GoldbergMV, Bankoti J, SelbyM,Nirschl CJ, et al.Immune inhibitory molecules LAG-3 and PD-1 synergistically regulateT-cell function to promote tumoral immune escape. Cancer Res2012;72:917–27.

24. Urba WJ. With the checkpoint inhibitors, can we call checkmatefor cancer? [video on the Internet]. Alexandria (VA): AmericanSociety of Clinical Oncology; 2013 [cited 2014 Jul 2]. Available from:http://meetinglibrary.asco.org/content/83771.

25. Wolchok JD, Kluger H, Callahan MK, Postow MA, Rizvi NA, LesokhinAM, et al. Nivolumab plus ipilimumab in advancedmelanoma. N Engl JMed 2013;369:122–33.

26. Fonsatti E, Maio M, Altomonte M, Hersey P. Biology and clinicalapplications of CD40 in cancer treatment. Semin Oncol 2010;37:517–23.

27. French RR, Chan HT, Tutt AL, Glennie MJ. CD40 antibody evokes acytotoxic T-cell response that eradicates lymphoma and bypassesT-cell help. Nat Med 1999;5:548–53.

28. Schoenberger SP, Toes RE, van der Voort EI, Offringa R, Melief CJ.T-cell help for cytotoxic T lymphocytes is mediated by CD40-CD40Linteractions. Nature 1998;393:480–3.

29. Melero I, Hirschhorn-Cymerman D, Morales-Kastresana A, San-mamed MF, Wolchok JD. Agonist antibodies to TNFR molecules thatcostimulate T and NK cells. Clin Cancer Res 2013;19:1044–53.

30. Suntharalingam G, Perry MR, Ward S, Brett SJ, Castello-Cortes J,Brunner MD, et al. Cytokine storm in a phase 1 trial of the anti-CD28monoclonal antibody TGN1412. N Engl J Med 2006;355:1018–28.

31. Curran MA, Callahan MK, Subudhi SK, Allison JP. Response to"Ipilimumab (Yervoy) and the TGN1412 catastrophe". Immunobiology2012;217:590–2.

32. Purdy AK, Campbell KS. Natural killer cells and cancer: regulation bythe killer cell Ig-like receptors (KIR). Cancer Biol Ther 2009;8:2211–20.

33. Romagn�e F, Andr�e P, Spee P, Zahn S, Anfossi N, Gauthier L, et al.Preclinical characterization of 1-7F9, a novel human anti-KIR receptortherapeutic antibody that augments natural killer-mediated killing oftumor cells. Blood 2009;114:2667–77.

34. Kohrt HE, Thielens A, Marabelle A, Sagiv-Barfi I, Sola C, Chanuc F,et al. Anti-KIR antibody enhancement of anti-lymphoma activity ofnatural killer cells as monotherapy and in combination with anti-CD20antibodies. Blood 2014;123:678–86.

35. Benson DM Jr, Bakan CE, Zhang S, Collins SM, Liang J, Srivastava S,et al. IPH2101, a novel anti-inhibitory KIR antibody, and lenalidomidecombine to enhance the natural killer cell versus multiple myelomaeffect. Blood 2011;118:6387–91.

36. SanbornRE, SharfmanWH,Segal NH,Hodi FS,Wolchok JD,UrbaWJ,et al. Aphase I dose-escalation andcohort expansion studyof lirilumab(anti-KIR; BMS-986015) administered in combination with nivolumab(anti-PD-1; BMS-936558; ONO-4538) in patients (Pts) with advancedrefractory solid tumors. J Clin Oncol 31, 2013 (suppl; abstr TPS3110).

37. Rizvi NA, Infante JR, Gibney GT, Bertino EM, Cooley SA, Lekatiset K,et al. A phase I study of lirilumab (BMS-986015), an anti-KIR mono-clonal antibody, administered in combination with ipilimumab, an anti-CTLA4 monoclonal antibody, in patients (Pts) with select advancedsolid tumors. J Clin Oncol 31, 2013 (suppl; abstr TPS3106).

38. Hashmi MH, Van Veldhuizen PJ. Interleukin-21: updated review ofphase I and II clinical trials in metastatic renal cell carcinoma, meta-static melanoma and relapsed/refractory indolent non-Hodgkin's lym-phoma. Expert Opin Biol Ther 2010;10:807–17.

39. Jure-Kunkel M, Selby M, Lewis K, Masters G, Valle J, Grosso J, et al.Nonclinical evaluation of the combination of mouse IL-21 and anti-mouse CTLA-4 or PD-1 blocking antibodies in mouse tumor models.J Clin Oncol 31, 2013 (suppl; abstr 3019).

40. Bhatia S, Curti BD, Gordon MS, Chesney J, Logan T, Thompson JA,et al. Phase 1 dose-escalation study is evaluating recombinant humanIL-21 (BMS-982470) in combination with ipilimumab (ipi) in patients

with advanced or metastatic melanoma (MM). J Clin Oncol 31, 2013(suppl; abstr TPS3109).

41. Chow LQM, Gordon MS, Logan TF, Antonia SJ, Bhatia S, ThompsonJA, et al. Phase 1 dose-escalation study of recombinant humaninterleukin-21 (rIL-21, BMS-982470) in combination with nivolumab(anti-PD-1; BMS-936558; ONO-4538) in patients (pts) with advancedor metastatic solid tumors. J Clin Oncol 31, 2013 (suppl; abstrTPS3112).

42. FribergM, Jennings R, Alsarraj M, Dessureault S, Cantor A, ExtermannM, et al. Indoleamine 2,3-dioxygenase contributes to tumor cell eva-sion of T cell-mediated rejection. Int J Cancer 2002;101:151–5.

43. Liu X, Shin N, Koblish HK, Yang G, Wang Q, Wang K, et al. Selectiveinhibition of IDO1 effectively regulates mediators of antitumor immu-nity. Blood 2010;115:3520–30.

44. Holmgaard RB, Zamarin D, Munn DH, Wolchok JD, Allison JP. Indo-leamine 2,3-dioxygenase is a critical resistance mechanism in antitu-mor T cell immunotherapy targeting CTLA-4. J Exp Med 2013;210:1389–402.

45. Rosenberg SA, Yang JC, Sherry RM, Kammula US, Hughes MS, PhanGQ, et al. Durable complete responses in heavily pretreated patientswith metastatic melanoma using T-cell transfer immunotherapy. ClinCancer Res 2011;17:4550–7.

46. Palena C, Schlom J. Vaccines against human carcinomas: strategiesto improve antitumor immune responses. J Biomed Biotechnol2010;2010:380697.

47. Guo C, Manjili MH, Subjeck JR, Sarkar D, Fisher PB, Wang XY, et al.Therapeutic cancer vaccines: past, present and future. Adv CancerRes 2013;119:421–75.

48. LeDT, Lutz E, Uram JN, Sugar EA, Onners B, Solt S, et al. Evaluation ofipilimumab in combination with allogeneic pancreatic tumor cellstransfected with a GM-CSF gene in previously treated pancreaticcancer. J Immunother 2013;36:382–9.

49. Schwartzentruber DJ, LawsonDH, Richards JM, Conry RM,Miller DM,Treisman J, et al. gp100 peptide vaccine and interleukin-2 in patientswith advanced melanoma. N Engl J Med 2013;364:2110–27.

50. Nielsen MB, Marincola FM. Melanoma vaccines: the paradox of T cellactivation without clinical response. Cancer Chemother Pharmacol2000;46:S62–6.

51. Zippelius A, Batard P, Rubio-Godoy V, Bioley G, Li�enard D, Lejeune F,et al. Effector function of human tumor-specific CD8 T cells in mela-noma lesions: a state of local functional tolerance. Cancer Res2004;64:2865–73.

52. Weber JS,KudchadkarRR,YuB,GallensteinD,HorakCE, InzunzaHD,Zhao X, et al. Safety, efficacy, and biomarkers of nivolumab withvaccine in ipilimumab-refractory or -naive melanoma. J Clin Oncol2013;31:4311–8.

53. KershawMH,DevaudC, JohnLB,WestwoodJA,DarcyPK. Enhancingimmunotherapyusing chemotherapy and radiation tomodify the tumormicroenvironment. Oncoimmunology 2013;2:e25962.

54. Formenti SC, Demaria S. Combining radiotherapy and cancerimmunotherapy: a paradigm shift. J Natl Cancer Inst 2013;105:256–65.

55. Ferrara TA, Hodge JW, Gulley JL. Combining radiation and immuno-therapy for synergistic antitumor therapy. Curr Opin Mol Ther2009;11:37–42.

56. Hannani D, Sistigu A, Kepp O, et al. Prerequisites for the antitumorvaccine-like effect of chemotherapy and radiotherapy. Cancer J2011;17:351–8.

57. M�enard C, Martin F, Apetoh L, Bouyer F, Ghiringhelli F. Cancerchemotherapy: not only a direct cytotoxic effect, but also an adjuvantfor antitumor immunity. Cancer Immunol Immunother 2008;57:1579–87.

58. Vanneman M, Dranoff G. Combining immunotherapy and targetedtherapies in cancer treatment. Nat Rev Cancer 2012;12:237–51.

59. Hodge JW, Ardiani A, Farsaci B, Kwilas AR, Gameiro SR. The tippingpoint for combination therapy: cancer vaccines with radiation, che-motherapy, or targeted small molecule inhibitors. Semin Oncol 2012;39:323–39.

60. Kroemer G, Galluzzi L, Kepp O, Zitvogel L. Immunogenic cell death incancer therapy. Annu Rev Immunol 2013;31:51–72.

www.aacrjournals.org Clin Cancer Res; 20(24) December 15, 2014 6267

Immuno-oncology Combinations

on May 13, 2018. © 2014 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst October 23, 2014; DOI: 10.1158/1078-0432.CCR-14-1457

61. Ramakrishnan R, Assudani D, Nagaraj S, Hunter T, Cho HI, Antonia S,et al. Chemotherapy enhances tumor cell susceptibility to CTL-medi-ated killing during cancer immunotherapy in mice. J Clin Invest2010;120:1111–24.

62. Hunter TB, Manimala NJ, Luddy KA, Catlin T, Antonia SJ. Paclitaxeland TRAIL synergize to kill paclitaxel-resistant small cell lung cancercells through a caspase-independent mechanism mediated throughAIF. Anticancer Res 2011;31:3193–204.

63. Schilling B, Sucker A, Griewank K, Zhao F, Weide B, G€orgens A, et al.Vemurafenib reverses immunosuppression by myeloid derived sup-pressor cells. Int J Cancer 2013;133:1653–63.

64. Frederick DT, Piris A, Cogdill AP, Cooper ZA, Lezcano C, Ferrone CR,et al. BRAF inhibition is associated with enhanced melanoma antigenexpression and a more favorable tumor microenvironment in patientswith metastatic melanoma. Clin Cancer Res 2013;19:1225–31.

65. Boni A, Cogdill AP, DangP, Udayakumar D,NjauwCN, SlossCM, et al.Selective BRAFV600E inhibition enhances T-cell recognition of mel-anoma without affecting lymphocyte function. Cancer Res 2010;70:5213–9.

66. Shindo T, Kim TK, Benjamin CL, Wieder ED, Levy RB, Komanduri KV.MEK inhibitors selectively suppress alloreactivity and graft-versus-host disease in a memory stage-dependent manner. Blood 2013;121:4617–26.

67. Ott PA, Henry T, Baranda SJ, Frleta D, Manches O, Bogunovic D, et al.Inhibition of both BRAF and MEK in BRAF(V600E) mutant melanomarestores compromised dendritic cell (DC) function while having differ-ential direct effects on DC properties. Cancer Immunol Immunother2013;62:811–22.

68. Weber J, Hamid O, Amin A, O'Day S, Masson E, Goldberg SM, et al.Randomized phase I pharmacokinetic study of ipilimumab with orwithout one of two different chemotherapy regimens in patients withuntreated advanced melanoma. Cancer Immun 2013;13:7.

69. Lynch TJ, Bondarenko I, Luft A, Serwatowski P, Barlesi F, Chacko R,et al. Ipilimumab in combinationwith paclitaxel and carboplatin as first-line treatment in stage IIIB/IV non-small-cell lung cancer: results from arandomized, double-blind, multicenter phase II study. J Clin Oncol2012;30:2046–54.

70. Reck M, Bondarenko I, Luft A, Serwatowski P, Barlesi F, Chacko R,et al. Ipilimumab in combinationwith paclitaxel and carboplatin as first-line therapy in extensive-disease-small-cell lung cancer: results from arandomized, double-blind, multicenter phase 2 trial. Ann Oncol2013;24:75–83.

71. Antonia SJ, Brahmer JR, Gettinger SN, Chow LQM, Juergens RA,Shepherd FA, et al. Nivolumab (anti-PD-1; BMS-936558, ONO-4538)in combination with platinum-based doublet chemotherapy (PT-DC) inadvanced non-small cell lung cancer (NSCLC). J Clin Oncol 32:5s,2014 (suppl; abstr 8113).

72. Slovin SF, Higano CS, Hamid O, Tejwani S, Harzstark A, Alumkal JJ,et al. Ipilimumab alone or in combination with radiotherapy in meta-static castration-resistant prostate cancer: results from an open-label,multicenter phase I/II study. Ann Oncol 2013;24:1813–21.

73. Kwon ED, Drake CG, Scher HI, Fizazi K, Bossi A, van den EertweghAJ,et al. Ipilimumab versus placebo after radiotherapy in patients withmetastatic castration-resistant prostate cancer that had progressedafter docetaxel chemotherapy (CA184-043): a multicentre, rando-mised, double-blind, phase 3 trial. Lancet Oncol 2014;15:700–12.

74. Grimaldi AM, Simeone E, Giannarelli D, Muto P, Falivene S, Borzillo V,et al. Abscopal effects of radiotherapy on advanced melanomapatients who progressed after ipilimumab immunotherapy. Oncoim-munology 2014;3:e28780.

75. Sagiv-Barfi I, Rajapaksa A, Czerwinski D, Chang S, Hebb J, Chester C,et al. Local tumor irradiation combined with a-PDL-1 immune check-point inhibition results in local and systemic anti-tumor responses:successful translation of a mouse model to a human case series[abstract]. In: Proceedings of the 105th Annual Meeting of the Amer-ican Association for Cancer Research; 2014 Apr 5–9; San Diego, CA.Philadelphia (PA): AACR; 2014. Abstract nr 2941.

76. Barker CA, PostowMA. Combinations of radiation therapy and immu-notherapy for melanoma: a review of clinical outcomes. Int J RadiatOncol Biol Phys 2014;88:986–97.

77. Hu-Lieskovan S, Robert L, Homet Moreno B, Ribas A. Combiningtargeted therapy with immunotherapy in BRAF-mutant melanoma:promise and challenges. J Clin Oncol 2014;32:2248–54.

78. Ascierto PA, Simeone E, Giannarelli D, Grimaldi AM, Romano A,Mozzillo N, et al. Sequencing of BRAF inhibitors and ipilimumab inpatients with metastatic melanoma: a possible algorithm for clinicaluse. J Trans Med 2012;10:107.

79. Ascierto PA, Simeone E, Sileni VC, Del Vecchio M, Marchetti P,Cappellini GC, et al. Sequential treatment with Ipilimumab and BRAFinhibitors in patients with metastatic melanoma: data from the ItalianCohort of the ipilimumab expanded access program. Cancer Invest2014;32:144–9.

80. Ribas A, Hodi FS, Callahan M, Konto C, Wolchok J. Hepatotoxicitywith combination of vemurafenib and ipilimumab. N Engl J Med 2013;368:1365–6.

81. Ackerman A, Klein O, McDermott DF, Wang W, Ibrahim N, LawrenceDP, et al. Outcomes of patients withmetastaticmelanoma treatedwithimmunotherapy prior to or after BRAF inhibitors. Cancer 2014;120:1695–701.

82. Johnson DB, Wallender EK, Cohen DN, Likhari SS, Zwerner JP,Powers JG, et al. Severe cutaneous and neurologic toxicity in mela-noma patients during vemurafenib administration following anti-PD-1therapy. Cancer Immunol Res 2013;1:373–7.

83. Gr€unwald V, Desar IM, Haanen J, Fiedler W, Mouritzen U, Olsen MW,et al. A phase I study of recombinant human interleukin-21 (rIL-21) incombination with sunitinib in patients with metastatic renal cell car-cinoma (RCC). Acta Oncol 2011;50:121–6.

84. Rini BI, Stein M, Shannon P, Eddy S, Tyler A, Stephenson JJ Jr,et al. Phase 1 dose-escalation trial of tremelimumab plus sunitinibin patients with metastatic renal cell cancer. Cancer 2011;117:758–67.

85. Amin A, Plimack ER, Infante JR, Ernstoff MS, Rini BI, McDermot DF,et al. Nivolumab (anti-PD-1; BMS-936558, ONO-4538) in combi-nation with sunitinib or pazopanib in patients (pts) with metastaticrenal cell carcinoma (mRCC). J Clin Oncol 32:5s, 2014 (suppl; abstr5010).

86. Houot R, Kohrt H, Levy R. Boosting antibody-dependent cellularcytotoxicity against tumor cells with a CD137 stimulatory antibody.Oncoimmunology 2012;1:957–8.

87. Kohrt HE, Houot R, Goldstein MJ, Weiskopf K, Alizadeh AA, Brody J,et al. CD137 stimulation enhances the antilymphoma activity of anti-CD20 antibodies. Blood 2011;117:2423–32.

Clin Cancer Res; 20(24) December 15, 2014 Clinical Cancer Research6268

Antonia et al.

on May 13, 2018. © 2014 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst October 23, 2014; DOI: 10.1158/1078-0432.CCR-14-1457

2014;20:6258-6268. Published OnlineFirst October 23, 2014.Clin Cancer Res   Scott J. Antonia, James Larkin and Paolo A. Ascierto  and Future ProspectsImmuno-oncology Combinations: A Review of Clinical Experience

  Updated version

  10.1158/1078-0432.CCR-14-1457doi:

Access the most recent version of this article at:

   

   

  Cited articles

  http://clincancerres.aacrjournals.org/content/20/24/6258.full#ref-list-1

This article cites 77 articles, 22 of which you can access for free at:

  Citing articles

  http://clincancerres.aacrjournals.org/content/20/24/6258.full#related-urls

This article has been cited by 10 HighWire-hosted articles. Access the articles at:

   

  E-mail alerts related to this article or journal.Sign up to receive free email-alerts

  Subscriptions

Reprints and

  [email protected]

To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at

  Permissions

  Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

.http://clincancerres.aacrjournals.org/content/20/24/6258To request permission to re-use all or part of this article, use this link

on May 13, 2018. © 2014 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst October 23, 2014; DOI: 10.1158/1078-0432.CCR-14-1457


Recommended