combination of 4-1bb agonist and pd-1 ...€¦ · 2oncology research unit, pfizer inc., san diego,...

13
Research Article Combination of 4-1BB Agonist and PD-1 Antagonist Promotes Antitumor Effector/Memory CD8 T Cells in a Poorly Immunogenic Tumor Model Shihao Chen 1 , Li-Fen Lee 1 ,Timothy S. Fisher 2 , Bart Jessen 3 , Mark Elliott 2 ,Winston Evering 3 , Kathryn Logronio 1 , Guang Huan Tu 1 , Konstantinos Tsaparikos 2 , Xiaoai Li 1 , Hui Wang 2 , Chi Ying 1 , Mengli Xiong 1 , Todd VanArsdale 2 , and John C. Lin 1 Abstract Immunotherapies targeting the programmed death 1 (PD-1) coinhibitory receptor have shown great promise for a subset of patients with cancer. However, robust and safe combination therapies are still needed to bring the benet of cancer immuno- therapy to broader patient populations. To search for an optimal strategy of combinatorial immunotherapy, we have compared the antitumor activity of the anti4-1BB/antiPD-1 combination with that of the antiPD-1/antiLAG-3 combination in the poorly immunogenic B16F10 melanoma model. Pronounced tumor inhibition occurred only in animals receiving antiPD-1 and anti4-1BB concomitantly, while combining antiPD-1 with antiLAG-3 led to a modest degree of tumor suppression. The activity of the anti4-1BB/antiPD-1 combination was depen- dent on IFNg and CD8 þ T cells. Both 4-1BB and PD-1 proteins were elevated on the surface of CD8 þ T cells by anti4-1BB/antiPD-1 cotreatment. In the tumor microenvironment, an effective antitumor immune response was induced as indicated by the increased CD8 þ /Treg ratio and the enrichment of genes such as Cd3e, Cd8a, Ifng, and Eomes. In the spleen, the combination treatment shaped the immune system to an effector/memory phenotype and increased the overall activity of tumor-specic CD8 þ CTLs, reecting a long-lasting systemic antitumor response. Furthermore, combination treatment in C57BL/6 mice showed no additional safety signals, and only minimally increased sever- ity of the known toxicity relative to 4-1BB agonist alone. There- fore, in the absence of any cancer vaccine, anti4-1BB/antiPD-1 combination therapy is sufcient to elicit a robust antitumor effector/memory T-cell response in an aggressive tumor model and is therefore a candidate for combination trials in patients. Cancer Immunol Res; 3(2); 14960. Ó2014 AACR. Introduction Immunotherapy has emerged as a promising treatment for cancer in recent years, showing durable responses in subsets of patients, and thus was selected as the Breakthrough of Year 2013 by the journal Science (1). For example, ipilimumab, an anti-CTL antigen 4 (CTLA-4) antagonistic antibody, showed an objective response rate (10%) in patients with unresectable or metastatic melanoma in a phase III trial, whereas severe or life-threatening toxicity was seen in 10% to 15% of the patients (2). Targeting the programmed death 1/programmed cell death ligand 1 (PD-1/PD- L1) axis resulted in a higher response rate of up to 37%, whereas treatment-related grade 3 toxicity rates were 9% to 33% in the melanoma cohorts of multiple phase I trials (3). The combination of two checkpoint inhibitors, antiCTLA-4 and antiPD-1, increased the response rate up to 53% in a phase I trial with advanced melanoma; however, severe treatment-related adverse events (grade 3) were also reported in 53% of cases and treatment was discontinued in 21% of the patients (3, 4). Although these results are promising, it is clear that increasing the frequency of response while limiting toxicities will be crucial for the success of combinatorial cancer immunotherapy in a wider population of patients with cancer. 4-1BB (CD137 and TNFRSF9) belongs to the TNF receptor superfamily (TNFRSF), and its expression is generally activation dependent on a subset of immune cells, including T cells and natural killer (NK) cells (5). The ligand that stimulates 4-1BB (4- 1BBL) is expressed on antigen-presenting cells, including dendrit- ic cells (DC), B cells, and macrophages (6). Ligation of 4-1BB receptors elicits a variety of immune cellspecic biologic responses. For instance, 4-1BB activation promotes cell prolifer- ation, survival, and cytokine production in T cells. In several tumor models, 4-1BB agonists have demonstrated antitumor efcacy (6, 7). Interestingly, in some autoimmunity models, 4- 1BB agonist also inhibited autoimmune reactions partly by antagonizing Th17 cell polarization (8, 9). This dual immuno- regulatory activity of 4-1BB offers the possibility to enhance antitumor activity while simultaneously dampening autoim- mune side effects associated with immunotherapy approaches 1 Rinat Laboratories, Pzer Inc., South San Francisco, California. 2 Oncology Research Unit, Pzer Inc., San Diego, California. 3 Drug Safety R&D, Pzer Inc., San Diego, California. Note: Supplementary data for this article are available at Cancer Immunology Research Online (http://cancerimmunolres.aacrjournals.org/). S. Chen and L.-F. Lee contributed equally to this article. Corresponding Authors: Shihao Chen, Rinat Laboratories, Pzer Inc., 230 East Grand Avenue, South San Francisco, CA 94080. Phone: 650-615-7329; E-mail: shihao.chen@pzer.com; and John C. Lin, john.lin@pzer.com doi: 10.1158/2326-6066.CIR-14-0118 Ó2014 American Association for Cancer Research. Cancer Immunology Research www.aacrjournals.org 149 on September 13, 2020. © 2015 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from Published OnlineFirst November 11, 2014; DOI: 10.1158/2326-6066.CIR-14-0118

Upload: others

Post on 20-Jul-2020

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Combination of 4-1BB Agonist and PD-1 ...€¦ · 2Oncology Research Unit, Pfizer Inc., San Diego, California. 3Drug Safety R&D, Pfizer Inc., San Diego, California. Note: Supplementary

Research Article

Combination of 4-1BB Agonist and PD-1Antagonist PromotesAntitumorEffector/MemoryCD8 T Cells in a Poorly Immunogenic TumorModelShihaoChen1, Li-FenLee1,TimothyS. Fisher2, Bart Jessen3,MarkElliott2,WinstonEvering3,Kathryn Logronio1, Guang Huan Tu1, Konstantinos Tsaparikos2, Xiaoai Li1, Hui Wang2,Chi Ying1, Mengli Xiong1, Todd VanArsdale2, and John C. Lin1

Abstract

Immunotherapies targeting the programmed death 1 (PD-1)coinhibitory receptor have shown great promise for a subset ofpatients with cancer. However, robust and safe combinationtherapies are still needed to bring the benefit of cancer immuno-therapy to broader patient populations. To search for an optimalstrategy of combinatorial immunotherapy, we have compared theantitumor activity of the anti–4-1BB/anti–PD-1 combinationwith that of the anti–PD-1/anti–LAG-3 combination in the poorlyimmunogenic B16F10 melanoma model. Pronounced tumorinhibition occurred only in animals receiving anti–PD-1 andanti–4-1BB concomitantly, while combining anti–PD-1 withanti–LAG-3 led to a modest degree of tumor suppression. Theactivity of the anti–4-1BB/anti–PD-1 combination was depen-dent on IFNg and CD8þ T cells. Both 4-1BB and PD-1 proteinswere elevated on the surface of CD8þ T cells by anti–4-1BB/anti–

PD-1 cotreatment. In the tumor microenvironment, an effectiveantitumor immune response was induced as indicated by theincreased CD8þ/Treg ratio and the enrichment of genes such asCd3e, Cd8a, Ifng, and Eomes. In the spleen, the combinationtreatment shaped the immune system to an effector/memoryphenotype and increased the overall activity of tumor-specificCD8þCTLs, reflecting a long-lasting systemic antitumor response.Furthermore, combination treatment in C57BL/6 mice showedno additional safety signals, and only minimally increased sever-ity of the known toxicity relative to 4-1BB agonist alone. There-fore, in the absence of any cancer vaccine, anti–4-1BB/anti–PD-1combination therapy is sufficient to elicit a robust antitumoreffector/memory T-cell response in an aggressive tumor modeland is therefore a candidate for combination trials in patients.Cancer Immunol Res; 3(2); 149–60. �2014 AACR.

IntroductionImmunotherapy has emerged as a promising treatment for

cancer in recent years, showing durable responses in subsets ofpatients, and thus was selected as the Breakthrough of Year 2013by the journal Science (1). For example, ipilimumab, an anti-CTLantigen 4 (CTLA-4) antagonistic antibody, showed an objectiveresponse rate (�10%) in patients with unresectable or metastaticmelanoma in a phase III trial, whereas severe or life-threateningtoxicity was seen in 10% to 15% of the patients (2). Targeting theprogrammed death 1/programmed cell death ligand 1 (PD-1/PD-L1) axis resulted in a higher response rate of up to 37%, whereastreatment-related grade � 3 toxicity rates were 9% to 33% in the

melanoma cohorts ofmultiple phase I trials (3). The combinationof two checkpoint inhibitors, anti–CTLA-4 and anti–PD-1,increased the response rate up to 53% in a phase I trial withadvanced melanoma; however, severe treatment-related adverseevents (grade � 3) were also reported in 53% of cases andtreatment was discontinued in 21% of the patients (3, 4).Although these results are promising, it is clear that increasingthe frequency of response while limiting toxicities will be crucialfor the success of combinatorial cancer immunotherapy in awiderpopulation of patients with cancer.

4-1BB (CD137 and TNFRSF9) belongs to the TNF receptorsuperfamily (TNFRSF), and its expression is generally activationdependent on a subset of immune cells, including T cells andnatural killer (NK) cells (5). The ligand that stimulates 4-1BB (4-1BBL) is expressed on antigen-presenting cells, including dendrit-ic cells (DC), B cells, and macrophages (6). Ligation of 4-1BBreceptors elicits a variety of immune cell–specific biologicresponses. For instance, 4-1BB activation promotes cell prolifer-ation, survival, and cytokine production in T cells. In severaltumor models, 4-1BB agonists have demonstrated antitumorefficacy (6, 7). Interestingly, in some autoimmunity models, 4-1BB agonist also inhibited autoimmune reactions partly byantagonizing Th17 cell polarization (8, 9). This dual immuno-regulatory activity of 4-1BB offers the possibility to enhanceantitumor activity while simultaneously dampening autoim-mune side effects associated with immunotherapy approaches

1Rinat Laboratories, Pfizer Inc., South San Francisco, California.2Oncology Research Unit, Pfizer Inc., San Diego, California. 3DrugSafety R&D, Pfizer Inc., San Diego, California.

Note: Supplementary data for this article are available at Cancer ImmunologyResearch Online (http://cancerimmunolres.aacrjournals.org/).

S. Chen and L.-F. Lee contributed equally to this article.

Corresponding Authors: Shihao Chen, Rinat Laboratories, Pfizer Inc., 230 EastGrand Avenue, South San Francisco, CA 94080. Phone: 650-615-7329; E-mail:[email protected]; and John C. Lin, [email protected]

doi: 10.1158/2326-6066.CIR-14-0118

�2014 American Association for Cancer Research.

CancerImmunologyResearch

www.aacrjournals.org 149

on September 13, 2020. © 2015 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst November 11, 2014; DOI: 10.1158/2326-6066.CIR-14-0118

Page 2: Combination of 4-1BB Agonist and PD-1 ...€¦ · 2Oncology Research Unit, Pfizer Inc., San Diego, California. 3Drug Safety R&D, Pfizer Inc., San Diego, California. Note: Supplementary

that break immune tolerance. Agonistic antibodies to 4-1BB(urelumab, BMS, and PF-05082566; Pfizer) are currently beingtested in humans with advanced cancer, and PF-05082566 wasfound well tolerated with evidence of disease stabilization inmultiple patients (10, 11).

PD-1 is an inhibitory receptor expressed on activated T cells aswell as other immune cells, and its expression can be associatedwith T-cell exhaustion (12). PD-1 binds to two ligands, PD-L1(B7-H1) and PD-L2 (B7-DC) that are often overexpressed ontumor cells. Such interactions deliver a negative signal to T cells todampen the immune response. Antibody blockade of PD-1 wasshown to enhance effector T-cell responses and induce T-cell–mediated tumor rejection in some mouse models (12). Additiveor synergistic antitumor effects were also reported when PD-1blockadewas combinedwith other immune checkpoint blockadesuch as T-cell immunoglobulin mucin 3 (TIM-3) or lymphocyteactivation gene 3 (LAG-3) in different tumor models (13, 14).

Anti–4-1BB agonist has been investigated extensively in pre-clinical studies to treat cancer when combined with other immu-nomodulatory antibodies. Synergistic antitumor effects have beenobserved for combinations of anti–4-1BBwith anti–TIM-3 (15) orwith anti–PD-1 (16, 17) in the ID8murine ovarian cancer model.Studies on ID8 ovarian carcinoma also indicated that the anti–4-1BB/anti–PD-L1 combination had moderate impact on survival,which could be further improved with additional vaccination(18). The anti–4-1BB/anti–CTLA-4 combination significantlydelayed MC38 colon carcinoma growth over anti–4-1BB oranti–CTLA-4 alone; however, the same regimen failed to controlB16F1 melanoma growth (19). The anti–4-1BB/anti–CTLA-4combination also failed to prolong the survival of B16F10tumor–bearing mice (20). Further studies showed that activevaccination was required for the anti–4-1BB/anti–CTLA-4 com-bination to effectively control the growth of B16-BL6 melanoma(21). These studies suggested that not all combinations aretherapeutically equivalent and different models might respondto treatment differently.Here, we report that the anti–4-1BB/anti–PD-1 combination is more efficacious than that of anti–PD-1/anti–LAG-3 in suppressing B16F10 melanoma growth withoutadjuvant or vaccination. We investigated the cellular mechanismunderlying this synergy. Finally, we showed that the combinationwas well tolerated in na€�ve or tumor-bearing mice, indicative of apotential therapeutic index for the combination.

Materials and MethodsMice

Six- to 8-week-old female C57BL/6 mice and IFNg-deficientmice were purchased from The Jackson Laboratory. Mice weremaintained and all animal experiments were conducted accord-ing to the protocols approved by the Institutional Animal Careand Use Committee of Rinat (South San Francisco) and World-wide Research and Development (La Jolla), Pfizer Inc.

Cell linesThe B16F10 melanoma cell line was purchased from the

ATCC in 2010. The MC38 colon carcinoma cell line was kindlyprovided by Dr. Antoni Ribas at UCLA (Los Angeles, CA) in2011. Cells were cultured in DMEM medium supplementedwith 10% FBS and 2 mmol/L L-glutamine, and IMPACT testedfor pathogens at the Research Animal Diagnostic Laboratory.No other authentication assay was performed. Pathogen-free

cells growing in an exponential growth phase were harvestedand used for tumor inoculation.

Antibodies for immunotherapy and flow cytometryTherapeutic rat anti-mouse 4-1BB mAb (clone MAB9371) was

purchased from R&D systems. Rat anti-mouse PD-1 mAb (cloneRMP1-14) and rat IgG2a isotype control were purchased fromBioXcell. Rat anti-mouse LAG-3 mAb (clone eBioC9B7W)was purchased from eBioscience. mAbs used for cell-surface orintracellular stains were purchased from BD Biosciences oreBioscience. They were hamster anti-mouse CD3e (clone 145-2C11), rat anti-mouse CD4 (clone RM4-5), rat anti-mouse CD8a(clone 53-6.7), rat anti-mouse CD25 (clone PC61), hamster anti-mouse CD137 (4-1BB; clone 17B5), hamster anti-mouse CD279(PD-1; clone J43), rat anti-mouse FoxP3 (clone FJK-16s), rat anti-mouse Eomes (clone, Dan11mag), hamster anti-mouse KLRG1(clone 2F1), and rat anti-mouse NK-1.1 (clone PK136). Live cellswere separated from dead cells using the LIVE/DEAD Fixable BlueDead Cell Stain Kit (Invitrogen).

Immune-cell phenotyping using flow cytometrySpleens from tumor-bearing mice were harvested and dissoci-

ated mechanically into single-cell suspension in ice-cold PBS.Splenocytes were treatedwith Red BloodCell Lysing Buffer Hybri-Max (Sigma-Aldrich), washed twice with PBS, and resuspended inPBS supplemented with 2% FBS and 0.9% NaN3. An aliquotof approximately 1 � 106 splenocytes was preincubated with 10mg/mL of mouse BD Fc Block (BD Biosciences) for 10 minutesbefore phenotyping mAb cocktail was added to specifically stainimmune cells.

Tumor-infiltrating lymphocytes (TIL) were prepared using theMouse Tumor Dissociation Kit and the GentleMACS Dissociatoraccording to the manufacturer's instructions (Miltenyi Biotec).

Cell-surface antigens were labeled by incubating cells at 4�C for30 minutes. Intracellular staining was carried out using a FoxP3/Transcription Factor Staining Buffer set according to the manu-facturer's protocol (eBioscience). Flow-cytometry data wereacquired using LSR Fortessa (BD Biosciences) and analyzed usingFlowJo (TreeStar Inc.).

In vivo tumor efficacy studiesC57BL/6 mice were inoculated subcutaneously at the right

flank with 1 � 106 B16F10 or MC38 cells in 0.1 mL of serum-free DMEMmedium or PBS. Treatment was started when tumorsreached 50 to 154 mm3 in size. Antibodies to 4-1BB (1 mg/kg),PD-1 (10 mg/kg), and LAG-3 (10 mg/kg), alone or in combina-tion, were administrated twice i.p. 5 days apart. In the study oftreating larger tumors (range of 126–350 mm3), antibodies weregiven four times i.p. 3 days apart. To treat IFNg-deficient mice,antibodies were given three times 3 days apart. To deplete CD8þ Tcells, anti-CD8 (53.5.8, BioXcell) was given as described previ-ously (22). Briefly, depleting antibody (100 mg) was administrat-ed 1 day before anti–4-1BB/anti–PD-1 administration and every 5days thereafter for a total of three doses. Tumor sizewasmeasuredin two dimensions using calipers, and the volume was expressedinmm3 using the following formula:V¼ 0.5 L�W2, where L andW are the long and short diameters of the tumor, respectively.

Antigen-specific immune response assayFrozen splenocytes from MC38 tumor–bearing mice after

antibody treatment were thawed and rested for 12 hours. Viable

Chen et al.

Cancer Immunol Res; 3(2) February 2015 Cancer Immunology Research150

on September 13, 2020. © 2015 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst November 11, 2014; DOI: 10.1158/2326-6066.CIR-14-0118

Page 3: Combination of 4-1BB Agonist and PD-1 ...€¦ · 2Oncology Research Unit, Pfizer Inc., San Diego, California. 3Drug Safety R&D, Pfizer Inc., San Diego, California. Note: Supplementary

cells were plated at 1 � 106 per well in duplicate in the presenceof 0.5 mg/mL of CD8-restricted MC38 immunodominant peptideKSPWFTTL (Proimmune) or the positive activation controlConA at 5 mg/mL (Sigma-Aldrich) for 48 hours. The assay wasdeveloped according to the manufacturer's instructions and readon a CTL ELISpot reader followed by spot enumeration usingImmunospot 5.0.3. The upper sensitivity limit of the assay was500 spots. Samples with no detectable ConA response wereremoved from the analysis.

Quantitative RT-PCRB16F10 orMC38 tumor samples (�2mm3)were collected into

RNAlater stabilization reagent at the endof efficacy studies. Tumorpieces were homogenized into RLT buffer containing 1% 2-mercaptoethanol using a Qiagen Tissuelyser and 5-mm steelbeads. RNA was isolated from the homogenate using RNeasymini spin columns, and quantified via UV absorbance using aNanodrop 8000 spectrophotometer. Total RNAper sample (1 mg)was used as template to prepare cDNA used as template forTaqMan primer probe sets targeting mouse Cd3e, Cd8a, Ifng,Eomes, Pdcd1, Cd274, Tnfrsf9, Cd4, Foxp3, Gapdh, and Actb. Sam-ples were amplified in duplicate in 384-well plates using anApplied Biosystems ViiA7 Real Time PCR system. Relative quan-tity (RQ) was calculated via the DDCt method relative to theaverage of the PBS control group.

Mouse toxicology studyNa€�ve C57BL/6 (n¼ 5/group) were given a single subcutaneous

administration of test articles or control (PBS) and clinical andanatomic pathology was analyzed 10 days later. At necropsy, thefollowing tissues were collected for histology: cecum, colon, duo-denum, epididymides, heart, ileum, jejunum, kidneys, liver, lung,spleen, pancreas, stomach, and testes. The testes and epididymideswere preserved in modified Davidson's solution, and all othertissues were fixed in 10% neutral-buffered formalin. All tissueswere routinely processed, embedded in paraffin and sectioned,stained with hematoxylin and eosin (H&E), and analyzed by aboard certified veterinary pathologist. In addition, blood andserumwere collected for hematology using the Advia 2120 (Bayer)and clinical chemistry analysis using the Advair 1200 (Siemens).

Potential toxicity was also assessed in B16F10 tumor–bearingmice. Mice with average tumors of approximately 100 mm3 (n ¼5/group) received a single s.c. dose of antibodies and clinical andanatomic pathology was analyzed 10 days later. Spleen and liverwere processed and evaluated as described above for the tumor-free mice. Whole blood was analyzed for complete blood countand liver enzymes by the Diagnostic laboratory, Department ofComparative Medicine, Stanford University (Stanford, CA).

Statistical analysisResults were expressed asmean� SEM. Statistical analyseswere

performed using GraphPad Prism 6.0 one-way or two-wayANOVA to compare the differences among multiple groups. AP value of <0.05 was considered as significant difference.

Results4-1BB activation and PD-1 blockade but not LAG-3 and PD-1dual blockade resulted in synergistic antitumor effects inmurine B16F10 melanoma

To evaluate the antitumor efficacy of combination immuno-therapy against established B16F10 tumors, an anti–4-1BBdosing

regimen was selected on the basis of prior studies that showedefficacy in the CT26 model (23) and anti–PD-1 and anti–LAG-3dosing regimens as previously reported (14, 21). Consistent withpublished results, anti–4-1BB, anti–PD-1, or anti–LAG-3 alonedid not consistently inhibit B16F10 tumor growth when treat-ments were started on tumors of 50 to 154 mm3 in size (Fig. 1Aand B). Combining anti–PD-1 with anti–LAG-3 resulted in 54%of tumor growth inhibition (TGI) relative to the isotype control(P < 0.001), but no mice were tumor free. In contrast, whenanimals were concurrently administered anti–4-1BB and anti–PD-1 antibodies, a dramatic efficacy of 85% TGI relative to theisotype control (P < 0.0001) was observed and 7 of 10 treatedanimals were tumor free (Fig. 1A and B). Combinatory efficacywas reproducible in an independent study by different experi-menters (Supplementary Fig. S1). Furthermore, significant TGI(51% relative to the control group; P < 0.0001) was observed forthe anti–4-1BB/anti–PD-1 combination when treatment wasapplied to very large tumors (size between 126 and 350 mm3),whereas the anti–PD-1/anti–LAG-3 combination was ineffective(Fig. 1C).

Likewise, a robust antitumor effect of the anti–4-1BB/anti–PD-1combination was observed in the MC38 colon cancer model(Fig. 1D). At the end of the study (day 21 after tumor implant),the TGI for the combination treatmentwas 63% relative to the PBScontrol (P < 0.0001). The suppression was also significant whencompared with the single agent alone (P < 0.05 vs. anti–4-1BBalone and P < 0.001 vs. anti–PD-1 alone).

The efficacy of anti–4-1BB/anti–PD-1 combination wasdependent on IFNg and CD8þ T cells

Given the antitumor efficacy of anti–4-1BB/anti–PD-1 combi-nation, it is important to understand how the synergy wasachieved. First, we compared tumor growth in response toanti–4-1BB/anti–PD-1 cotreatment in wild-type and IFNg-defi-cient mice. The combination suppressed tumor growth in wild-type animals, whereas such an antitumor effect was completelyabolished in the IFNg-deficient mice (Fig. 2A). To determine theimmune-cell subset that is required for the anti–4-1BB/anti–PD-1combination,wedepletedCD8þT cells in B16F10 tumor–bearingmice and compared tumor growth after treatment. In the absenceof CD8þ T cells, combination treatment–related tumor suppres-sion was completely abrogated (Fig. 2B).

Anti–4-1BB/anti–PD-1 combination increased the availabilityof target molecules for therapeutic antibodies on CD8þ T cells

To further understand the mechanisms underlying the syner-gistic effects achieved by the anti–4-1BB/anti–PD combination,we examined the expression of 4-1BB and PD-1 on CD8þ T cellsin response to antibody treatment. Splenocytes or TILs fromB16F10 tumor–bearing mice after treatment were isolated,immunostained, and analyzed by flow cytometry. A representa-tive flow cytometric gating strategy is shown in SupplementaryFig. S2. The anti–PD-1 (J43) used for detection does not competewith the therapeutic anti–PD-1 (RMP1-14) for the binding to PD-1, as determined by BIAcore analysis (data not shown). In PBS-and isotype-treated animals, PD-1 expression was barely detected(<1%)on splenic CD8þ T cells (Fig. 2C). Anti–PD-1 treatment didnot affect its own expression or that of 4-1BB. However, anti–4-1BB treatment induced the expression of PD-1 on 34% of splenicCD8þ T cells when administrated alone (P < 0.0001 vs. isotypecontrol) and on 29% of CD8þ T cells when coadministered with

4-1BB and PD-1 Potentiate CD8-Mediated Antitumor Immunity

www.aacrjournals.org Cancer Immunol Res; 3(2) February 2015 151

on September 13, 2020. © 2015 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst November 11, 2014; DOI: 10.1158/2326-6066.CIR-14-0118

Page 4: Combination of 4-1BB Agonist and PD-1 ...€¦ · 2Oncology Research Unit, Pfizer Inc., San Diego, California. 3Drug Safety R&D, Pfizer Inc., San Diego, California. Note: Supplementary

anti–PD-1 (P < 0.001 vs. isotype control). Combination therapyalso induced PD-1 expression on 74% of TIL CD8þ T cells (P <0.001 vs. isotype control; Fig. 2D).

Therapeutic 4-1BB agonist relies on 4-1BB expressed on primedT cells for efficacy (24, 25). Anti–4-1BB treatment induced its ownexpression, with the cell-surface expression detectable on 15% ofsplenic CD8þ T cells in the anti–4-1BB–treated group and 10% of

the combination treatment group, comparedwith 0.2% inPBS- orisotype-treated groups (P < 0.0001 anti–4-1BB treatment vs.isotype control and P < 0.01 combination treatment vs. isotypecontrol; Fig. 2E). There was a trend that the combination treat-ment upregulated 4-1BB expression on TILCD8þ T cells, althoughthe increase was not statistically significant (Fig. 2F). These resultsindicated that more abundant targets were available for

Figure 1.4-1BB activation and PD-1 blockade synergistically amplify the antitumor effects in B16F10 melanoma and MC38 colon carcinoma models. C57BL/6 micewere inoculated s.c. (day 0) with 1 � 106 B16F10 (A–C) or 1 � 106 MC38 (D) cells. A and B, after tumors were established, mice were randomized into groupsof 10 animals per group with an average tumor volume of approximately 85 mm3 (range, 50–154 mm3) on day 9. Animals were administered i.p. rat IgG2a(isotype control, 10mg/kg), anti–4-1BB (1mg/kg), anti–PD-1 (10mg/kg), or anti–LAG-3 (10mg/kg)mAbs in avolumeof 200mL, alone or in combinations as indicatedin the figure, on days 9 and 14 (indicated by arrows). C, when B16F10 tumor volume reached 126 to 350 mm3 (day 10), mice were randomized into groupsof 10 animals per group and treatedwith rat IgG2a, anti–PD-1/anti–LAG-3 or anti–4-1BB/anti–PD-1 mAbs every 3 days for a total of four doses (indicated by arrows).D,MC38 tumor–bearing animals (8mice/group)with an average tumor volumeof approximately 140mm3 (day 10)were treatedwith anti–4-1BB and anti–PD-1mAbsi.p. on days 10 and 15 (indicated by arrows). Tumor size was measured two to three times a week. Mean � SEM of each treatment group is shown in A, C, and D.B, the individual tumor growth over time for plot (A). Statistics were generated using two-way ANOVA, � , P < 0.05; �� , P < 0.01; ��� , P < 0.001; and ���� , P < 0.0001,when comparing groups as indicated by the vertical lines.

Chen et al.

Cancer Immunol Res; 3(2) February 2015 Cancer Immunology Research152

on September 13, 2020. © 2015 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst November 11, 2014; DOI: 10.1158/2326-6066.CIR-14-0118

Page 5: Combination of 4-1BB Agonist and PD-1 ...€¦ · 2Oncology Research Unit, Pfizer Inc., San Diego, California. 3Drug Safety R&D, Pfizer Inc., San Diego, California. Note: Supplementary

therapeutic antibodies both in the tumor and in the spleen aftercombination treatment.

Anti–4-1BB/anti–PD-1 treatment promoted antitumorimmune response in the tumor microenvironment

To provide further evidence of a localized antitumor immuneresponse, TILs were isolated from B16F10 tumors 3 days after thesecond dose of antibodies and an immune-cell subset analysis wasperformed by flow cytometry. The combination treatmentenriched CD8þ T cells with an 8-fold increase in the CD8þ/CD4þ

ratio over the isotype group (P < 0.0001; Fig. 3A). Regulatory T-cell(Treg) abundance was not altered (Fig. 3B). However, the ratio ofCD8þ/Tregwas increased by7-fold in response to the combinationover the isotype treatment (P < 0.0001; Fig. 3C). In agreementwithpublished results (9), we observed the induction of KLRG1 onapproximately 30% of the CD8þ T cells in the anti–4-1BB aloneand combination groups (P < 0.001 vs. isotype; Fig. 3D).

In addition, we evaluated the genes associated with antitu-mor immune responses in B16F10 tumor samples by quanti-tative PCR. The RQ of each target gene was related to theaverage of the PBS control samples and calculated by the DDCt

method using GAPDH and actin housekeeping genes. Com-pared with the PBS group, combination treatment led to anenrichment of signals for Cd3e (P < 0.001), Cd8a (P < 0.001),

Ifng (P < 0.001), Eomes (P < 0.001), Tnfrsf9 (P < 0.05), Pdcd1 (P¼ 0.0001), Cd274 (P < 0.001), and Foxp3 (P < 0.05; Fig. 4). Nosignificant changes were noted for CD4 expression. The profileof mRNA enrichment observed here and in MC38 tumors(Supplementary Fig. S3) is largely congruent with the flowcytometric findings in the B16F10 tumors, supporting a strongenhancement of CD8 effector/memory cell phenotype induc-tion by the anti–4-1BB/anti–PD-1 combination.

Anti–4-1BB/anti–PD-1 combination induced T-cell effector/memory differentiation

To explore modulation of the immune system systemicallyfollowing anti–4-1BB/anti–PD-1 treatment, splenocytes were iso-lated from tumor-bearing mice and detailed phenotypic analysisof the immune cells was performed by flow cytometry. One of thecharacteristics of 4-1BB activation is to induce the expression ofeomesodermin (Eomes), a transcription factor associated with T-cell effector/memory differentiation (9, 26). We detected Eomesexpression on 64% of CD8þ T cells in the anti–4-1BB–treatedanimals and on 69%of CD8þ T cells in the anti–4-1BB/anti–PD-1combination-treated animals, but only on 19%of CD8þ T cells inthe isotype-treated animals (P < 0.0001 vs. isotype control; Fig.5A). Anti–PD-1 treatment alone did not alter Eomes expression(18% EomesþCD8þ T cells). CD4þ T cells from control groups

Figure 2.CD8þ T cells were immunomodulated and required for the efficacy of anti–4-1BB/anti–PD-1 combination treatment.Wild-type C57BL/6 or IFNg-deficient mice wereinoculated s.c. (day 0) with 1 � 106 B16F10. Mice were randomized into groups of 10 animals per group with an average tumor volume of approximately80 mm3. A, mice were randomized on day 8, and anti–4-1BB (1 mg/kg)/anti–PD-1 (10 mg/kg) mAbs were dosed on days 8, 11, and 14 (indicated by arrows). B, micewere randomized on day 10. Isotype control IgG (100 mg) or anti-CD8 mAb was administered i.p. on days 11, 16, and 21 (indicated by blue arrows). Anti–4-1BB(1 mg/kg)/anti–PD-1 (10 mg/kg) mAbs were dosed on days 12, 17, and 22 (indicated by black arrows). Tumor size was measured two to three times a week. Mean� SEMof each treatment group is shown. C to F, spleens and tumor fragments fromB16F10 tumor–bearingmice after antibody treatment (Fig. 1A and SupplementaryFig. S1) were harvested and disaggregated into a single-cell suspension. After immunostaining, cells were analyzed by flow cytometry for the expression ofCD3, CD4, CD8, PD-1, and 4-1BB. The expression of PD-1 on splenic and TIL CD8þ T cells is shown in C and D, respectively. The expression of 4-1BB on splenic and TILCD8þ T cells is shown in E and F, respectively. Each symbol represents an individual animal within the same treatment group. �� , P < 0.01; ��� , P < 0.001; and���� , P < 0.0001, when comparing groups as indicated by the horizontal lines.

4-1BB and PD-1 Potentiate CD8-Mediated Antitumor Immunity

www.aacrjournals.org Cancer Immunol Res; 3(2) February 2015 153

on September 13, 2020. © 2015 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst November 11, 2014; DOI: 10.1158/2326-6066.CIR-14-0118

Page 6: Combination of 4-1BB Agonist and PD-1 ...€¦ · 2Oncology Research Unit, Pfizer Inc., San Diego, California. 3Drug Safety R&D, Pfizer Inc., San Diego, California. Note: Supplementary

(PBS or isotype) did not express Eomes (<1%), whereas expres-sion was increased to 2.8% of CD4þ T cells in the anti–4-1BB–treated group, and further increased to 8.3%of CD4þ T cells in theanti–4-1BB/anti–PD-1 combination group (P<0.0001 vs. isotypegroup; Fig. 5B).

Memory is the hallmark of the acquired immune system. Thus,we characterized the memory phenotype of T cells at the end of aB16F10 efficacy study. The combination treatment significantlyincreased both the central memory (Tcm, CD44þ CD62Lþ; P <0.0001 vs. PBS control) and effector memory (Tem, CD44þ

CD62L�; P < 0.0001 vs. PBS control) CD8þ T cells in the spleen(Fig. 5C and D). CD8þ Tem subset increase was also observed inthe MC38 model (Supplementary Fig. S4).

Anti–4-1BB/anti–PD-1 treatment generated antigen-specificCTL response

To determine whether anti–4-1BB/anti–PD-1 treatmentinduced an antigen-specific immune response, we isolated sple-nocytes from MC38 tumor–bearing mice after treatment andstimulated with the CD8-restricted MC38 immunodominantpeptide KSPWFTTL ex vivo. IFNg production assayed by ELISpotwas evaluated as a measurement of response. Anti–4-1BB treat-ment increased the number of IFNg-producing cells (250 � 36)compared with that of the PBS group (117� 28; P < 0.05; Fig. 6).Anti–PD-1 treatment also increased IFNg production (242�46, P< 0.05 vs. PBS group). The combination of anti–4-1BB and anti–

PD-1 further increased the numbers of peptide/tumor-specific Tcells to 377 � 24 and the increase was statistically significant ascompared with those treated with PBS (P < 0.0001), anti–4-1BBalone (P<0.05), or anti–PD-1 alone (P<0.05). Thedata indicatedthat the combination elicited a greater antigen-specific CTLresponse than either single-agent alone.

Anti–4-1BB/anti–PD-1 combination immunotherapy resultedin moderate liver enzyme elevation in mice compared withanti–4-1BB alone

To understand the safety profile of the anti–4-1BB/anti–PD-1combination therapy, we tested these antibodies either within orabove their respective therapeutic dose range in non–tumor-bearing mice. C57BL/6 mice were administered a single s.c.injection of PBS, anti–4-1BB alone (at 0.1, 1, or 5 mg/kg),anti–PD-1 alone (at 20mg/kg), and three combination treatmentgroups consisting of 20 mg/kg anti–PD-1 with each of the threedose levels of anti–4-1BB listed above. Ten days after dosing, thestudy was terminated and tissue, blood, and serum collected forclinical and anatomic pathology assessment.

Of the clinical chemistry pathology measured, only thoseassociated primarily with liver toxicity and certain peripheralblood subsets were significantly affected. The groups treated with1 or 5mg/kg anti–4-1BB alone demonstrated increases in alanineaminotransferase (ALT) of 6- (P < 0.01) and 5-fold (P < 0.05),respectively, compared with the PBS group (Fig. 7A). The

Figure 3.Anti–4-1BB/PD-1 combinationenriched CD8þ T cells in TILs. Tumorfragments were harvested fromB16F10 tumor–bearing mice afterantibody treatment anddisaggregated into a single-cellsuspension. After surface staining,cells were fixed and permeabilized forintracellular marker staining, andfinally analyzed by flow cytometry forthe expression of CD3, CD4, CD8,CD25, KLRG1, and Foxp3. A,the ratio of CD8þ to CD4þ

T cells; B, the frequency of Treg(CD4þCD25þFoxp3þ); C, the ratio ofCD8þ T cells to Treg; and D, theexpression of KLRG1 on CD8þ T cells.��� , P < 0.001 and ���� , P < 0.0001,when comparing groups as indicatedby the horizontal lines.

Chen et al.

Cancer Immunol Res; 3(2) February 2015 Cancer Immunology Research154

on September 13, 2020. © 2015 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst November 11, 2014; DOI: 10.1158/2326-6066.CIR-14-0118

Page 7: Combination of 4-1BB Agonist and PD-1 ...€¦ · 2Oncology Research Unit, Pfizer Inc., San Diego, California. 3Drug Safety R&D, Pfizer Inc., San Diego, California. Note: Supplementary

combination groups with anti–PD-1 had slightly greater increasesin ALT of 10- (P < 0.0001) and 8-fold (P < 0.0001), respectively,compared with the PBS group. Glutamate dehydrogenase (GLDH)potentially offers differential diagnostic potential in the investiga-tion of liver disease, particularly when interpreted in conjunctionwithother hepatic enzyme test results. It ismore concentrated in thecentral areas of the liver lobules than in the periportal zones and isusually only released from necrotic cells due to its exclusivemitochondrial localization (27). The results of theGLDHmeasure-ments were similar to those of ALT, with increases in the 1 mg/kg

and 5 mg/kg anti–4-1BB single-agent and combination groups of4-fold (P<0.001), 3-fold (P<0.01), 6-fold (P<0.0001), and5-fold(P < 0.0001), respectively, relative to the PBS group (Fig. 7B).Although the difference between the combination and the corre-sponding anti–4-1BB–alone groups was statistically significant(Fig. 7A and B), the magnitude of difference was small (�50%increase in combination vs. single agent for either ALT or GLDH).No effects on ALT or GLDH were observed in the other groups.Aspartate aminotransferase (AST) followed a similar pattern as theother liver enzyme markers (Fig. 7C).

PBS

Anti–4-1

BB

Anti–PD-1

Anti–4-1

BB/Anti–

PD1–1

0

1

2

3

4

Cd3

e - L

og (R

Q)

Cd8

a - L

og (R

Q)

Ifng

- Log

(RQ

)

*****

PBS

Anti–4-1

BB

Anti–PD-1

Anti–4-1

BB/Anti–

PD1–1

0

1

2

3

4*****

PBS

Anti–4-1

BB

Anti–PD-1

Anti–4-1

BB/Anti–

PD1–2

–1

0

1

2

3

4*****

PBS

Anti–4-1

BB

Anti–PD-1

Anti–4-1

BB/Anti–

PD1–1

5 *

4

3

2

1

0

0

1

2

3******

PBS

Anti–4-1

BB

Anti–PD-1

Anti–4-1

BB/Anti–

PD1–1

0

1

2

3

4Pd

cd1

- Log

(RQ

)

Eom

es -

Log

(RQ

)

*****

PBS

Anti–4-1

BB

Anti–PD-1

Anti–4-1

BB/Anti–

PD1–0.5

0.0

0.5

1.0

1.5

2.0

Cd2

74 -

Log

(RQ

)

******

PBS

Anti–4-1

BB

Anti–PD-1

Anti–4-1

BB/Anti–

PD1

Tnfr

sf9

- RQ

PBS

Anti–4-1

BB

Anti–PD-1

Anti–4-1

BB/Anti–

PD1–1

0

1

2

3

Cd4

- Lo

g (R

Q)

PBS

Anti–4-1

BB

Anti–PD-1

Anti–4-1

BB/Anti–

PD1–1.0

–0.5

0.0

0.5

1.0

1.5

2.0

Foxp

3 - L

og (R

Q) *

Figure 4.Anti–4-1BB/PD-1 combination enriched genes associated with antitumor immune response. B16F10 tumor samples were collected after antibody treatmentand total RNA was isolated. Gene expression from each treatment group was calculated using real-time PCR analysis with Gapdh and Actb as the endogenouscontrols. Column dot plots are shown for the genes of Cd3e, Cd8a, Ifng, Eomes, Pdcd1, Cd274, Tnfrsf9, Cd4, and Foxp3. Except for 4-1BB expression, therelative expression for all other genes is shown in log scale due to the great variance within the group. Each symbol represents an individual animal within the sametreatment group. � , P < 0.05; �� , P < 0.01; and ��� , P < 0.001, when comparing groups as indicated by the horizontal lines.

4-1BB and PD-1 Potentiate CD8-Mediated Antitumor Immunity

www.aacrjournals.org Cancer Immunol Res; 3(2) February 2015 155

on September 13, 2020. © 2015 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst November 11, 2014; DOI: 10.1158/2326-6066.CIR-14-0118

Page 8: Combination of 4-1BB Agonist and PD-1 ...€¦ · 2Oncology Research Unit, Pfizer Inc., San Diego, California. 3Drug Safety R&D, Pfizer Inc., San Diego, California. Note: Supplementary

Several hematology parameters were affected by the studytreatments. There were decreases in platelet counts in the 1 and5mg/kg anti–4-1BB alone (P < 0.001 and P < 0.0001, respectively,vs. PBS) and combination with anti–PD-1 groups (P < 0.0001 vs.PBS; Fig. 7D). Although there was a statistically significantdecrease in platelets in the combination groups relative to thecorresponding anti–4-1BB single-agent groups (P < 0.05 for the1mg/kg anti–4-1BB/anti–PD-1 vs. 1mg/kg anti–4-1BB alone, andP < 0.01 for 5 mg/kg anti–4-1BB/anti–PD-1 vs. 5 mg/kg anti–4-1BB alone), the magnitude of the additional increase was quitemodest (roughly 15%). No effects on platelets were observed inthe other treatment groups. The 5-mg/kg anti–4-1BB alone andanti–PD-1 combination at 1 and 5 mg/kg anti–4-1BB showeddramatic lymphocyte decreases (�75%; P < 0.0001 vs. PBS),whereas the decrease in other groups ranged from 18% to 35%relative to the PBS group (Fig. 7E). Not only is the differencebetween the combination at 1mg/kg anti–4-1BB and the 1mg/kganti–4-1BB alone statistically significant (P < 0.0001), but furtherdecrease of lymphocyte count by 55% (combination vs. 4-1BBalone) may be biologically significant. Only the 5-mg/kg anti–4-

1BB–alone group showed a statistically significant decrease inneutrophils (P < 0.001 vs. PBS), and addition of anti–PD-1appeared to block the effects of anti–4-1BB on neutrophildecrease (Fig. 7F). Total white blood cells were also affected bythe treatments, but this changewas dominated by the lymphocyteeffects (data not shown).

Among the tissues examined histopathologically, treatment-related findings were only observed in the liver and spleen. In theliver (Supplementary Table S1A), treatment with anti–4-1BB at 1and5mg/kg led to an increased incidence and severityofmixed cellinfiltrate and caused single-cell necrosis that ranged fromminimalto moderate in severity. Addition of anti–PD-1 did not causeadditional effects. Mixed cell infiltration was present withminimalseverity, and single-cell necrosis was absent in all other treatmentgroups. In the spleen (Supplementary Table S1B), the incidence oflymphoidhyperplasiawas increased inall antibody-treatedgroups.The severity increased fromminimal in the PBS group to minimaltomild inall other groups except the5-mg/kg anti–4-1BB/anti–PD-1 combination group, in which it was mild to moderate. Theincidence and severity of extramedullary hematopoiesis were

Figure 5.Anti–4-1BB/PD-1 combination induced T-cell effector/memory differentiation. At the end of efficacy studies, spleens from B16F10 tumor–bearing mice wereharvested and disaggregated into a single-cell suspension. After surface staining, cells were fixed and permeabilized for intracellular marker staining, andfinally analyzed by flow cytometry for the expression of CD3, CD4, CD8, CD44, CD62L, and Eomes. The expression of Eomes in CD8þ and CD4þ T cells is shown in Aand B, respectively. The percentages of central memory (CD44þCD62Lþ) and effector memory (CD44þCD62L�) CD8þ T cells are shown in C and D, respectively.Each symbol represents an individual animal within the same treatment group. �� , P < 0.01; ��� , P < 0.001; and ���� , P < 0.0001, versus PBS or rat IgG2a.

Chen et al.

Cancer Immunol Res; 3(2) February 2015 Cancer Immunology Research156

on September 13, 2020. © 2015 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst November 11, 2014; DOI: 10.1158/2326-6066.CIR-14-0118

Page 9: Combination of 4-1BB Agonist and PD-1 ...€¦ · 2Oncology Research Unit, Pfizer Inc., San Diego, California. 3Drug Safety R&D, Pfizer Inc., San Diego, California. Note: Supplementary

increased in the 1- and 5-mg/kg anti–4-1BB combination groups,but not significantly altered in all other groups.

To assess potential toxicity in tumor-bearing animals, micewith established B16F10 tumors were treated with isotype (20mg/kg), anti–4-1BB (1 mg/kg), anti–PD-1 (20 mg/kg), or theanti–4-1BB (1 mg/kg)/anti–PD-1 (20 mg/kg) combination. Anti-body treatment, tissue collection, and data analysis were per-formed in a similar manner to those for tumor-free mice. Therewere no statistically significant changes in the absolute number ofperipheral platelets, lymphocytes, and neutrophils among allgroups. ALT was elevated in the combination group (P < 0.05vs. isotype; Supplementary Fig. S5). However, the AST levels werehighly variable and greatest in the control group, which preventedus from evaluating its changes in response to treatments (data notshown). The histopathology of the spleen and liver after treat-ments displayed similar patterns as described in the tumor-freemice (data not shown).

DiscussionT-cell activation is regulated through effective coordination of

costimulatory signals provided by cell-surface coregulatory recep-tors in addition to the antigen-specific TCR signaling (28). Ago-nistic antibodies directed against activating receptors and block-ing antibodies to inhibitory receptors may enhance T-cell stim-ulation and are under active investigation for cancer therapy

(7, 29). Blockade of the suppressive signals via anti–CTLA-4,anti–PD-1, and anti–PD-L1 agents has shown clinical benefit(3). Dual blockade of immunosuppressive pathways via anti–CTLA-4 and anti–PD-1 or via anti–PD-1 and anti–LAG-3 hasentered clinical trials (3, 4). Modulation of stimulatory signalsby 4-1BB, OX40, or GITR pathway is still in the early stages ofclinical evaluation (30). In preclinical studies, combinations ofimmunomodulators as well as in combination with chemother-apy, targeted agents, vaccination or irradiation, have been eval-uated in various models (15–18, 31–33). Here, we show that thecombination of anti–4-1BB with anti–PD-1 synergistically inhib-ited the growth of B16F10melanoma andMC38 colon carcinomain syngeneic C57BL/6 mice, and that the combination was rea-sonably well tolerated, supporting the clinical development of ananti–4-1BB/anti–PD-1 combination immunotherapy.

Anti–4-1BB therapydemonstratedpromisingbut limited clinicalbenefits in early clinical trials (34). Potential mechanisms of 4-1BBagonistic antibody–mediated tumor regressions include breakingof immunologic ignorance and prevention of T-cell deletion andanergy (5, 35, 36). Given that 4-1BB is specifically expressed onantigen-experiencedT cells (7, 37), the observed increase in 4-1BBþ

T cells may implicate an enhanced immune response to tumorchallenge in anti–4-1BB–treated animals. We and others haveshown that 4-1BB activation upregulated PD-1 expression oneffector T cells, and the PD-1/PD-L1 axis has been reported toconfer tumor resistance to 4-1BB costimulatory therapy (38). Inaddition, PD-L1 was induced in anti–4-1BB/anti–PD-1–treatedtumors in vivo (Fig. 4 and Supplementary Fig. S3) as well asIFNg-treated cells in vitro (data not shown and ref. 38). Therefore,cotreatment of anti–4-1BB with anti–PD-1 represents a mechanis-tically rational combination to maximize the antitumor potential.Thishypothesis is further supportedbydirectly comparingdifferentcombinations in the samemodel. Here, we show that anti–4-1BB/anti–PD-1 combination was more potent than anti–PD-1/anti–LAG-3 combination in suppression of both small and largeB16F10 tumors (Fig. 1). The combination of anti–4-1BB withanti–PD-1 was also more efficacious than dual combinations ofanti–CLTA-4/anti–PD-1 and anti–4-1BB/anti–CTLA-4 in the ID8model (16, 17). Therefore, in at least twoof themost aggressive andpoorly immunogenic tumor models (ID8 and B16), anti–4-1BB/anti–PD-1 combination appears to exhibit the strongest antitumoreffect among thedual-agent combinationsof severalwidely studiedpathways (CTLA-4, PD-1, LAG-3, and 4-1BB).

Previousworkhas implicated that the T-box transcription factorsEomes and T-bet act as master regulators in CD8þ T-cell effectorand memory differentiation and function (26, 39–41). Recently,Eomes has been shown to be required for the antitumor immunitymediatedby 4-1BBagonist immunotherapy (42). 4-1BB activationwas able to induce Eomes expression in tumor-infiltrating T cellswithout affecting T-bet expression, and the high expression ofEomes promoted the development of novel KLRG1þ Eomesþ

T-cell subsets characterized by enhanced, multipotent cytotoxicity(9).We also showhere that 4-1BB stimulation increased Eomes onsplenicCD8þT cells and to a lesser degree onCD4þT cells,whereasanti–PD-1 alonehadnoeffect. Consistentwith the roleof Eomes inT-cell polarization, we found the accumulation of CD8þ effectorand central memory T cells in the spleen of tumor-bearing miceafter anti–4-1BB/anti–PD-1 treatment. Furthermore, these CD8þ Tcells were functionally active, and produced large amounts of IFNgupon tumor antigen-specific restimulation ex vivo. The loss ofantitumor protection when CD8þ T cells were depleted further

Figure 6.Anti–4-1BB/PD-1 combination enhanced antigen-specific T-cell response. Atthe end of the efficacy study as described in Fig. 1D, splenocyteswere isolatedand fresh frozen. Viable cells after thaw were rested for 12 hours andchallenged with CD8-restricted MC38 immunodominant peptide KSPWFTTLex vivo. IFNg production was assessed by ELISpot. The number ofspots, an indication of the number of IFNg-producing cells, was reported.Each symbol represents an individual animal within the same treatmentgroup. � , P < 0.05 and ���� , P < 0.0001, when comparing groups as indicatedby the horizontal lines.

4-1BB and PD-1 Potentiate CD8-Mediated Antitumor Immunity

www.aacrjournals.org Cancer Immunol Res; 3(2) February 2015 157

on September 13, 2020. © 2015 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst November 11, 2014; DOI: 10.1158/2326-6066.CIR-14-0118

Page 10: Combination of 4-1BB Agonist and PD-1 ...€¦ · 2Oncology Research Unit, Pfizer Inc., San Diego, California. 3Drug Safety R&D, Pfizer Inc., San Diego, California. Note: Supplementary

supported their pivotal role in antitumor immunity. In agreementwith the essential role of IFNg for 4-1BB agonistic activity (43), thecombination also required IFNg as the antitumor activity wasabolished in IFNg-deficient mice (Fig. 2A). Durable efficacy islikely to occur given that the combination treatment inducedtumor antigen-specific and memory phenotype T cells. In thetumors, we observed signals associated with antitumor activity,such as the increased CD8þ/Treg ratio and induced KLRG1 onCD8þ T cells. By gene-expression analysis, we were able to dem-onstrate that an active antitumor immune response was inducedwith increases in the expression ofmRNA encodingCd8a, Ifng, andEomes. Unlike published data (15, 17), our findings did not showchanges in myeloid-derived suppressor cells in response to treat-ment (data not shown).

Finally, the combination of anti–4-1BB and anti–PD-1 anti-bodies was well tolerated in mice. There were no clinical signs oftoxicity or mortalities from either of the single agents or thecombination, and anti–PD-1 alone caused only minimaldecreases in peripheral lymphocytes and splenic lymphoid hyper-plasia. The effects in the combination arms, which included livermixed cell infiltration and single-cell necrosis with associatedincreases in plasma liver enzymes, splenic lymphoid hyperplasia

and extramedulary hematopoiesis, and decreases in peripheralwhite blood cell subsets and platelets, were driven predominantlyby the anti–4-1BB antibody. These findings were observed withsimilar severity in the anti–4-1BB–alone treatment arms and havebeen described previously with another mouse-reactive anti–4-1BB (44). The human relevance of themouse findings is apparentin a previous report listing neutropenia and transaminitis as twoof the most common adverse events in a phase I trial of a 4-1BBagonist antibody in patients with cancer (45). The most notablecombination effect in the mouse study was the synergisticdecrease in peripheral lymphocytes observed at the 1-mg/kg doseof anti–4-1BB in combination with anti–PD-1. This observationlikely represents the desired combined pharmacologic effect oflymphocyte activation and tissue sequestration.

Taken together, the pharmacologic effects of the anti–4-1BB/anti–PD-1 combination demonstrate amuch greater synergywithrespect to the antitumor efficacy than toxicity in syngeneicmodelswith an intact immune system. This preclinical study provides astrong rationale for developing anti–4-1BB/anti–PD-1 combina-tion immunotherapy in human patients. A study of 4-1BB agonistPF-05082566 plus the PD-1 inhibitor MK-3475 in patients withsolid tumors (B1641003/KEYNOTE-0036) has been proposed

Figure 7.Serum liver enzyme levels and hematology alterations in response to 4-1BB activation therapy in normal mice. Serum samples collected 10 days following a singleadministration of treatments to C57BL/6 mice (n ¼ 5/group) were analyzed for the amount of ALT (A), GLDH (B), or AST (C). Whole-blood samples weresubjected to platelet counts (D), lymphocyte counts (E), or neutrophil counts (F). The anti–4-1BB-0.1, anti–4-1BB-1, and anti–4-1BB-5 represent the 0.1-, 1-, and5-mg/kg doses of anti–4-1BB alone groups, respectively. Anti–PD-1-20 represents the group of anti–PD-1 treatment at 20 mg/kg. The combo-0.1, combo-1, andcombo-5 represent the groups of 0.1, 1, and 5mg/kg anti–4-1BBmAb in combinationwith 20mg/kg anti–PD-1mAb. � ,P<0.05; �� ,P <0.01; ��� ,P <0.001; and ���� ,P <0.0001, when comparing groups as indicated by the horizontal lines.

Chen et al.

Cancer Immunol Res; 3(2) February 2015 Cancer Immunology Research158

on September 13, 2020. © 2015 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst November 11, 2014; DOI: 10.1158/2326-6066.CIR-14-0118

Page 11: Combination of 4-1BB Agonist and PD-1 ...€¦ · 2Oncology Research Unit, Pfizer Inc., San Diego, California. 3Drug Safety R&D, Pfizer Inc., San Diego, California. Note: Supplementary

(clinicaltrials.gov). The cellular phenotypes and immunemechanisms underlying the synergism as we delineated here alsoprovide a framework for patient biomarker investigation in com-bination clinical trials.

Disclosure of Potential Conflicts of InterestB. Jessen reports receiving commercial research support from Pfizer Inc.

J.C. Lin has ownership interest (including patents) in Pfizer Inc. No potentialconflicts of interest were disclosed by the other authors.

Authors' ContributionsConception and design: S. Chen, L.-F. Lee, T.S. Fisher, B. Jessen, M. Elliott,H. Wang, T. VanArsdale, J.C. LinDevelopment of methodology: S. Chen, T.S. Fisher, M. Elliott, K. Logronio, X.Li, T. VanArsdaleAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): S. Chen, L.-F. Lee, T.S. Fisher, M. Elliott, W. Evering,K. Logronio, G.H. Tu, K. Tsaparikos, X. Li, H. Wang, C. Ying, M. Xiong

Analysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis): S. Chen, L.-F. Lee, T.S. Fisher, B. Jessen,W. Evering, K.Logronio, K. Tsaparikos, X. Li, T. VanArsdale, J.C. LinWriting, review, and/or revision of the manuscript: S. Chen, L.-F. Lee, T.S.Fisher, B. Jessen, M. Elliott, W. Evering, J.C. LinAdministrative, technical, or material support (i.e., reporting or organizingdata, constructing databases): S. Chen, K. Logronio, H. WangStudy supervision: S. Chen, L.-F. Lee, K. Logronio, K. Tsaparikos, H. Wang, T.VanArsdale, J.C. Lin

AcknowledgmentsThe authors thank Dr. Barbra Sasu at Rinat Laboratories (South San Fran-

cisco, CA) for critical reading and comments on the article.The costs of publication of this articlewere defrayed inpart by the payment of

page charges. This article must therefore be hereby marked advertisement inaccordance with 18 U.S.C. Section 1734 solely to indicate this fact.

Received June18, 2014; revisedOctober 13, 2014; acceptedOctober 27, 2014;published OnlineFirst November 11, 2014.

References1. Couzin-Frankel J. Breakthrough of the year 2013. Cancer immunotherapy.

Science 2013;342:1432–3.2. Hodi FS,O'Day SJ,McDermott DF,Weber RW, Sosman JA,Haanen JB, et al.

Improved survivalwith ipilimumab in patients withmetastaticmelanoma.N Engl J Med 2010;363:711–23.

3. Page DB, Postow MA, Callahan MK, Allison JP, Wolchok JD. Immunemodulation in cancer with antibodies. Ann Rev Med 2014;65:185–202.

4. Wolchok JD, Kluger H, Callahan MK, Postow MA, Rizvi NA, Lesokhin AM,et al. Nivolumab plus ipilimumab in advanced melanoma. N Engl J Med2013;369:122–33.

5. Wang C, Lin GH, McPherson AJ, Watts TH. Immune regulation by 4-1BBand 4-1BBL: complexities and challenges. Immunol Rev 2009;229:192–215.

6. Vinay DS, Kwon BS. Immunotherapy of cancer with 4-1BB. Mol CancerTher 2012;11:1062–70.

7. Wang S, Chen L. Immunobiology of cancer therapies targeting CD137 andB7-H1/PD-1 cosignal pathways. Curr Topics Microbiol Immunol 2011;344:245–67.

8. Vinay DS, Cha K, Kwon BS. Dual immunoregulatory pathways of 4-1BBsignaling. J Mol Med 2006;84:726–36.

9. Curran MA, Geiger TL, Montalvo W, Kim M, Reiner SL, Al-Shamkhani A,et al. Systemic 4-1BB activation induces a novel T cell phenotype driven byhigh expression of Eomesodermin. J Exp Med 2013;210:743–55.

10. Kohrt HEK, Godwin JE, Lossos IS, Williams ME, Timmerman J, Link BK,et al. A phase Ib, open-label, multicenter study of urelumab (BMS-663513)in combination with rituximab in subjects with relapsed/refractory B-cellmalignancies. J Clin Oncol 2013;31:abstr TPS3108.

11. SegalNH,Gopal AK, Bhatia S, KohrtHE, LevyR, PishvaianMJ, et al. A phase1 study of PF-05082566 (anti-4-1BB) in patients with advanced cancer. JClin Oncol 2014;32:abstr 3007.

12. McDermott DF, Atkins MB. PD-1 as a potential target in cancer therapy.Cancer Med 2013;2:662–73.

13. Ngiow SF, Teng MW, Smyth MJ. Prospects for TIM3-targeted antitumorimmunotherapy. Cancer Res 2011;71:6567–71.

14. Woo SR, Turnis ME, Goldberg MV, Bankoti J, Selby M, Nirschl CJ, et al.Immune inhibitory molecules LAG-3 and PD-1 synergistically regulate T-cell function to promote tumoral immune escape. Cancer Res 2012;72:917–27.

15. Guo Z, Cheng D, Xia Z, Luan M, Wu L, Wang G, et al. Combined TIM-3blockade and CD137 activation affords the long-term protection in amurine model of ovarian cancer. J Transl Med 2013;11:215.

16. Dai M, Wei H, Yip YY, Feng Q, He K, Popov V, et al. Long-lasting completeregression of established mouse tumors by counteracting Th2 inflamma-tion. J Immunother 2013;36:248–57.

17. Wei H, Zhao L, Li W, Fan K, Qian W, Hou S, et al. Combinatorial PD-1blockade and CD137 activation has therapeutic efficacy in murine cancermodels and synergizes with cisplatin. PLoS ONE 2013;8:e84927.

18. Duraiswamy J, Freeman GJ, Coukos G. Therapeutic PD-1 pathwayblockade augments with other modalities of immunotherapy T-cellfunction to prevent immune decline in ovarian cancer. Cancer Res2013;73:6900–12.

19. Kocak E, Lute K, ChangX,MayKF Jr, ExtenKR, ZhangH, et al. Combinationtherapy with anti-CTL antigen-4 and anti-4-1BB antibodies enhancescancer immunity and reduces autoimmunity. Cancer Res 2006;66:7276–84.

20. Jensen BA, Pedersen SR, Christensen JP, Thomsen AR. The availability of afunctional tumor targeting T-cell repertoire determines the anti-tumorefficiency of combination therapy with anti-CTLA-4 and anti-4-1BB anti-bodies. PLoS ONE 2013;8:e66081.

21. Curran MA, Kim M, Montalvo W, Al-Shamkhani A, Allison JP. Combina-tion CTLA-4 blockade and 4-1BB activation enhances tumor rejection byincreasing T-cell infiltration, proliferation, and cytokine production. PLoSONE 2011;6:e19499.

22. Butler NS, Moebius J, Pewe LL, Traore B, Doumbo OK, Tygrett LT, et al.Therapeutic blockade of PD-L1 and LAG-3 rapidly clears establishedblood-stage Plasmodium infection. Nat Immunol 2012;13:188–95.

23. Escuin-Ordinas H, Elliott M, Atefi M, Lee M, Ng C, Wei L, et al. PETimaging to non-invasively study immune activation leading to antitu-mor responses with a 4-1BB agonistic antibody. J Immuno Ther Cancer2013;1:14.

24. Melero I, Shuford WW, Newby SA, Aruffo A, Ledbetter JA, Hellstrom KE,et al. Monoclonal antibodies against the 4-1BB T-cell activation moleculeeradicate established tumors. Nat Med 1997;3:682–5.

25. Quetglas JI, Dubrot J, Bezunartea J, Sanmamed MF, Hervas-Stubbs S,Smerdou C, et al. Immunotherapeutic synergy between anti-CD137 mAband intratumoral administration of a cytopathic Semliki Forest virusencoding IL-12. Mol Ther 2012;20:1664–75.

26. Iborra S, Ramos M, Arana DM, Lazaro S, Aguilar F, Santos E, et al. N-rascouples antigen receptor signaling to Eomesodermin and to functionalCD8þ T-cell memory but not to effector differentiation. J Exp Med 2013;210:1463–79.

27. Panteghini M, Bais R, vanSolinge WW. Enzymes. In:Burtis CA, AshwoodER, Bruns DE, editors. Tietz textbook of clinical chemistry and moleculardiagnostics, 4th edn. St. Louis: Elsevier Saunders; 2006. p. 597–643.

28. Sharpe AH, Abbas AK. T-cell costimulation–biology, therapeutic potential,and challenges. N Engl J Med 2006;355:973–5.

29. Mellman I, Coukos G, Dranoff G. Cancer immunotherapy comes of age.Nature 2011;480:480–9.

30. Melero I,Hirschhorn-CymermanD,Morales-KastresanaA, SanmamedMF,Wolchok JD. Agonist antibodies to TNFRmolecules that costimulate T andNK cells. Clin Cancer Res 2013;19:1044–53.

31. Deng L, Liang H, Burnette B, Beckett M, Darga T, Weichselbaum RR, et al.Irradiation and anti-PD-L1 treatment synergistically promote antitumorimmunity in mice. J Clin Invest 2014;124:687–95.

www.aacrjournals.org Cancer Immunol Res; 3(2) February 2015 159

4-1BB and PD-1 Potentiate CD8-Mediated Antitumor Immunity

on September 13, 2020. © 2015 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst November 11, 2014; DOI: 10.1158/2326-6066.CIR-14-0118

Page 12: Combination of 4-1BB Agonist and PD-1 ...€¦ · 2Oncology Research Unit, Pfizer Inc., San Diego, California. 3Drug Safety R&D, Pfizer Inc., San Diego, California. Note: Supplementary

32. Duraiswamy J, Kaluza KM, Freeman GJ, Coukos G. Dual blockade ofPD-1 and CTLA-4 combined with tumor vaccine effectively restoresT-cell rejection function in tumors. Cancer Res 2013;73:3591–603.

33. Morales-Kastresana A, Sanmamed MF, Rodriguez I, Palazon A, Martinez-Forero I, Labiano S, et al. Combined immunostimulatory monoclonalantibodies extend survival in an aggressive transgenic hepatocellular car-cinoma mouse model. Clin Cancer Res 2013;19:6151–62.

34. Ascierto PA, Simeone E, SznolM, Fu YX,Melero I. Clinical experiences withanti-CD137 and anti-PD1 therapeutic antibodies. Seminars Oncol2010;37:508–16.

35. Melero I, Murillo O, Dubrot J, Hervas-Stubbs S, Perez-Gracia JL. Multi-layered actionmechanisms of CD137 (4-1BB)-targeted immunotherapies.Trends Pharmacol Sci 2008;29:383–90.

36. Wilcox RA, Flies DB, Zhu G, Johnson AJ, Tamada K, Chapoval AI, et al.Provision of antigen and CD137 signaling breaks immunological igno-rance, promoting regression of poorly immunogenic tumors. J Clin Invest2002;109:651–9.

37. Ye Q, Song DG, Poussin M, Yamamoto T, Best A, Li C, et al. CD137accurately identifies and enriches for naturally occurring tumor-reactive Tcells in tumor. Clin Cancer Res 2014;20:44–55.

38. Hirano F, Kaneko K, Tamura H, Dong H, Wang S, Ichikawa M, et al.Blockade of B7-H1 and PD-1 bymonoclonal antibodies potentiates cancertherapeutic immunity. Cancer Res 2005;65:1089–96.

39. Intlekofer AM, Takemoto N, Wherry EJ, Longworth SA, Northrup JT,Palanivel VR, et al. Effector and memory CD8þ T cell fate coupled by T-bet and eomesodermin. Nat Immunol 2005;6:1236–44.

40. Li G, Yang Q, Zhu Y, Wang HR, Chen X, Zhang X, et al. T-Bet and eomesregulate the balance between the effector/central memory T cells versusmemory stem like T cells. PLoS ONE 2013;8:e67401.

41. Pearce EL, Mullen AC, Martins GA, Krawczyk CM, Hutchins AS, Zediak VP,et al. Control of effector CD8þ T cell function by the transcription factorEomesodermin. Science 2003;302:1041–3.

42. Song C, Sadashivaiah K, Furusawa A, Davila E, Tamada K, Banerjee A.Eomesodermin is required for antitumor immunity mediated by 4-1BB-agonist immunotherapy. Oncoimmunology 2014;3:e27680.

43. Wilcox RA, Flies DB, Wang H, Tamada K, Johnson AJ, Pease LR, et al.Impaired infiltration of tumor-specific cytolytic T cells in the absence ofinterferon-gamma despite their normal maturation in lymphoid organsduringCD137monoclonal antibody therapy.CancerRes 2002;62:4413–8.

44. Niu L, Strahotin S, Hewes B, Zhang B, Zhang Y, Archer D, et al. Cytokine-mediated disruption of lymphocyte trafficking, hemopoiesis, and induc-tion of lymphopenia, anemia, and thrombocytopenia in anti-CD137-treated mice. J Immunol 2007;178:4194–213.

45. Sznol M, Hodi FS, Margolin K, McDermott DF, Ernstoff MS, Kirkwood JM,et al. Phase I study of BMS-663513, a fully human anti-CD137 agonistmonoclonal antibody, in patients (pts) with advanced cancer (CA). J ClinOncol 2008;26:3007.

Cancer Immunol Res; 3(2) February 2015 Cancer Immunology Research160

Chen et al.

on September 13, 2020. © 2015 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst November 11, 2014; DOI: 10.1158/2326-6066.CIR-14-0118

Page 13: Combination of 4-1BB Agonist and PD-1 ...€¦ · 2Oncology Research Unit, Pfizer Inc., San Diego, California. 3Drug Safety R&D, Pfizer Inc., San Diego, California. Note: Supplementary

2015;3:149-160. Published OnlineFirst November 11, 2014.Cancer Immunol Res   Shihao Chen, Li-Fen Lee, Timothy S. Fisher, et al.   Tumor ModelAntitumor Effector/Memory CD8 T Cells in a Poorly Immunogenic Combination of 4-1BB Agonist and PD-1 Antagonist Promotes

  Updated version

  10.1158/2326-6066.CIR-14-0118doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://cancerimmunolres.aacrjournals.org/content/suppl/2014/11/11/2326-6066.CIR-14-0118.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://cancerimmunolres.aacrjournals.org/content/3/2/149.full#ref-list-1

This article cites 42 articles, 16 of which you can access for free at:

  Citing articles

  http://cancerimmunolres.aacrjournals.org/content/3/2/149.full#related-urls

This article has been cited by 35 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

  Permissions

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

.http://cancerimmunolres.aacrjournals.org/content/3/2/149To request permission to re-use all or part of this article, use this link

on September 13, 2020. © 2015 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst November 11, 2014; DOI: 10.1158/2326-6066.CIR-14-0118