8
Cancer Immunology Miniatures Clinical Response of a Patient to AntiPD-1 Immunotherapy and the Immune Landscape of Testicular Germ Cell Tumors Shalin Shah 1 , James E. Ward 1,2 , Riyue Bao 2 , Curtis R. Hall 1,2 , Bruce E. Brockstein 1,2 , and Jason J. Luke 2 Abstract AntiProgramed Death 1 (PD-1) is standard immunotherapy for multiple cancers, and the expression of one of its ligands, PD-L1, has been described in germ cell tumors (GCT). Neither the clinical activity of antiPD-1 nor the incidence of an immunoresponsive tumor microenvironment has been described for GCTs. A patient initially diagnosed with mela- noma via ne needle aspiration was treated with one dose of antibody to PD-1. A core needle biopsy was subsequently performed to acquire sufcient tissue for molecular analysis, which led to a change in diagnosis to metastatic embryonal carcinoma. The testicular GCT cohort of The Cancer Genome Atlas was analyzed using a T-cell gene signature associated with benet from immunotherapy. Primary tumors (N ¼ 134) were categorized as high (T-cellinamed), medium, or low (nonT-cell-inamed) by their T-cell signature derived from RNAseq data. AntiPD-1 induced decreases in serum markers and a 33% reduction in tumor volume. Gene expression revealed a T-cellinamed tumor microenvironment in 47% of testicular GCTs, including seminoma (83%) and nonseminoma (17%) tumor subtypes. Expression of alpha-fetoprotein (AFP) RNA correlat- ed with lack of the T-cell signature, with increasing AFP RNA inversely correlating with the inamed signature and expres- sion of IFNg -associated genes. These data suggest that GCTs can respond to antiPD-1 and that gene expression proling sup- ports investigation of immunotherapy for treatment of GCTs. Cancer Immunol Res; 4(11); 9039. Ó2016 AACR. Introduction Testicular germ cell tumors (TGCT), although one of the most common malignancies of young men, are highly treatable with modern chemotherapy and have cure rates exceeding 95% (1). Germ cell tumors are categorized by histologic and biochemical parameters as either being a pure seminoma or a nonseminoma- tous germ cell tumor (NSGCT) with treatment dictated both by American Joint Committee on Cancer and risk modelingbased assessment (2, 3). Early-stage germ cell tumors respond well to orchiectomy and adjuvant radiotherapy or chemotherapy. After using a risk stratication system for more advanced disease, germ cell tumors are commonly treated with 3 to 4 cycles of chemo- therapy. Though disease refractory to initial chemotherapy can be treated with effective second- and sometimes third-line chemo- therapy (4, 5), progressive chemotherapy is associated with sig- nicant toxicity. Long-term sequelae of treatment include second malignancies, hypogonadism, infertility, and cardiovascular dis- ease (6). As a result, new treatment strategies and novel drug approaches are needed. Cancer immunotherapy has undergone a resurgence in recent years with the development of immune-checkpoint blocking antibodies such as anticytotoxic T lymphocyte antigen 4 (CTLA-4) and antiprogrammed death 1 (PD-1)/programmed death-ligand 1 (PD-L1). These approaches have changed clinical paradigms in melanoma, nonsmall cell lung cancer, and clear cell renal cell carcinoma, in which phase III trials have documen- ted improvements in overall survival and an improvement in toxicity prole for most patients (7, 8). In many other tumor types, it is expected that checkpoint immunotherapy will become a part of the treatment armamentarium over a short period of time. Despite tremendous optimism regarding the clinical efcacy of immune-checkpoint immunotherapy, the majority of patients currently do not respond. Multiple predictive biomarkers for immunotherapy have been put forward, particularly immuno- histochemistry for PD-L1 or tumor-inltrating lymphocytes (TIL). However, each of these is only an aspect of an effective antitumor immune response, in which inltrating T cells elaborate cytokines such as IFNg that leads to upregulation of immune effector molecules (9). A more robust approach to tumor microenviron- mental evaluation may thus be facilitated by gene expression proling where an active antitumor immune environment has been coined the "T cellinamed tumor microenvironment" (10). This phenotype has prognostic signicance and is associated with the presence of microenvironmental factorssuch as PD-L1, indolamine-2,3-dioxygenase (IDO), and regulatory T cells (9)as well as with clinical responses to immunotherapeutics, such as antibodies to CTLA-4 and PD-1 (11, 12). 1 Northshore University HealthSystem, Evanston, Illinois. 2 University of Chicago, Chicago, Illinois. Note: Supplementary data for this article are available at Cancer Immunology Research Online (http://cancerimmunolres.aacrjournals.org/). B.E. Brockstein and J.J. Luke as cosenior authors contributed equally to this article. Corresponding Author: Jason J. Luke, University of Chicago, 5841 S. Maryland Ave. MC2115, Chicago, IL 60637. Phone: 773-834-3096; Fax: 773-702-0963; E-mail: [email protected] doi: 10.1158/2326-6066.CIR-16-0087 Ó2016 American Association for Cancer Research. Cancer Immunology Research www.aacrjournals.org 903 on July 25, 2020. © 2016 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from Published OnlineFirst September 16, 2016; DOI: 10.1158/2326-6066.CIR-16-0087

Clinical Response of a Patient to Anti PD-1 Immunotherapy ...Immunotherapy and the Immune Landscape of Testicular Germ Cell Tumors Shalin Shah1, James E.Ward1,2, Riyue Bao2, Curtis

  • Upload
    others

  • View
    7

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Clinical Response of a Patient to Anti PD-1 Immunotherapy ...Immunotherapy and the Immune Landscape of Testicular Germ Cell Tumors Shalin Shah1, James E.Ward1,2, Riyue Bao2, Curtis

Cancer Immunology Miniatures

Clinical Response of a Patient to Anti–PD-1Immunotherapy and the Immune Landscape ofTesticular Germ Cell TumorsShalin Shah1, James E.Ward1,2, Riyue Bao2, Curtis R. Hall1,2, Bruce E. Brockstein1,2, andJason J. Luke2

Abstract

Anti–Programed Death 1 (PD-1) is standard immunotherapyfor multiple cancers, and the expression of one of its ligands,PD-L1, has been described in germ cell tumors (GCT). Neitherthe clinical activity of anti–PD-1 nor the incidence of animmunoresponsive tumor microenvironment has beendescribed for GCTs. A patient initially diagnosed with mela-noma via fine needle aspiration was treated with one dose ofantibody to PD-1. A core needle biopsy was subsequentlyperformed to acquire sufficient tissue for molecular analysis,which led to a change in diagnosis to metastatic embryonalcarcinoma. The testicular GCT cohort of The Cancer GenomeAtlas was analyzed using a T-cell gene signature associated withbenefit from immunotherapy. Primary tumors (N ¼ 134) were

categorized as high (T-cell–inflamed), medium, or low (non–T-cell-inflamed) by their T-cell signature derived from RNAseqdata. Anti–PD-1 induced decreases in serummarkers and a 33%reduction in tumor volume. Gene expression revealed a T-cell–inflamed tumor microenvironment in 47% of testicular GCTs,including seminoma (83%) and nonseminoma (17%) tumorsubtypes. Expression of alpha-fetoprotein (AFP) RNA correlat-ed with lack of the T-cell signature, with increasing AFP RNAinversely correlating with the inflamed signature and expres-sion of IFNg-associated genes. These data suggest that GCTs canrespond to anti–PD-1 and that gene expression profiling sup-ports investigation of immunotherapy for treatment of GCTs.Cancer Immunol Res; 4(11); 903–9. �2016 AACR.

IntroductionTesticular germ cell tumors (TGCT), although one of the most

common malignancies of young men, are highly treatable withmodern chemotherapy and have cure rates exceeding 95% (1).Germ cell tumors are categorized by histologic and biochemicalparameters as either being a pure seminoma or a nonseminoma-tous germ cell tumor (NSGCT) with treatment dictated both byAmerican Joint Committee on Cancer and risk modeling–basedassessment (2, 3). Early-stage germ cell tumors respond well toorchiectomy and adjuvant radiotherapy or chemotherapy. Afterusing a risk stratification system for more advanced disease, germcell tumors are commonly treated with 3 to 4 cycles of chemo-therapy. Though disease refractory to initial chemotherapy can betreated with effective second- and sometimes third-line chemo-therapy (4, 5), progressive chemotherapy is associated with sig-nificant toxicity. Long-term sequelae of treatment include secondmalignancies, hypogonadism, infertility, and cardiovascular dis-

ease (6). As a result, new treatment strategies and novel drugapproaches are needed.

Cancer immunotherapy has undergone a resurgence in recentyears with the development of immune-checkpoint blockingantibodies such as anti–cytotoxic T lymphocyte antigen 4(CTLA-4) and anti–programmed death 1 (PD-1)/programmeddeath-ligand 1 (PD-L1). These approaches have changed clinicalparadigms in melanoma, non–small cell lung cancer, and clearcell renal cell carcinoma, in which phase III trials have documen-ted improvements in overall survival and an improvement intoxicity profile for most patients (7, 8). In many other tumortypes, it is expected that checkpoint immunotherapy will becomea part of the treatment armamentarium over a short periodof time.

Despite tremendous optimism regarding the clinical efficacy ofimmune-checkpoint immunotherapy, the majority of patientscurrently do not respond. Multiple predictive biomarkers forimmunotherapy have been put forward, particularly immuno-histochemistry for PD-L1or tumor-infiltrating lymphocytes (TIL).However, each of these is only an aspect of an effective antitumorimmune response, inwhich infiltrating T cells elaborate cytokinessuch as IFNg that leads to upregulation of immune effectormolecules (9). A more robust approach to tumor microenviron-mental evaluation may thus be facilitated by gene expressionprofiling where an active antitumor immune environment hasbeen coined the "T cell–inflamed tumor microenvironment"(10). This phenotype has prognostic significance and is associatedwith the presence ofmicroenvironmental factors—such as PD-L1,indolamine-2,3-dioxygenase (IDO), and regulatory T cells (9)—as well as with clinical responses to immunotherapeutics, such asantibodies to CTLA-4 and PD-1 (11, 12).

1Northshore University HealthSystem, Evanston, Illinois. 2University ofChicago, Chicago, Illinois.

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

B.E. Brockstein and J.J. Luke as cosenior authors contributed equally to thisarticle.

Corresponding Author: Jason J. Luke, University of Chicago, 5841 S. MarylandAve. MC2115, Chicago, IL 60637. Phone: 773-834-3096; Fax: 773-702-0963;E-mail: [email protected]

doi: 10.1158/2326-6066.CIR-16-0087

�2016 American Association for Cancer Research.

CancerImmunologyResearch

www.aacrjournals.org 903

on July 25, 2020. © 2016 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst September 16, 2016; DOI: 10.1158/2326-6066.CIR-16-0087

Page 2: Clinical Response of a Patient to Anti PD-1 Immunotherapy ...Immunotherapy and the Immune Landscape of Testicular Germ Cell Tumors Shalin Shah1, James E.Ward1,2, Riyue Bao2, Curtis

Here, we describe the effective treatment of a patient withNSGCT with anti–PD-1 immunotherapy and then describe ananalysis of the TGCT cohort of The Cancer GenomeAtlas (TCGA).Based on gene expression profiling using a previously described T-cell gene signature (13), we found that a substantial proportion ofGCTs are likely to be susceptible to checkpoint immunotherapy.Finally, we identified a potential biochemical mediator of immu-nosuppression whose potential warrants further investigation inprospective clinical research.

Case ReportA 32-year-old Caucasian male developed a palpable mass on

the left of his neck without other symptoms. CT showed a largeconfluent nodalmass in the left neck (pretreatment, Fig. 1A and B;posttreatment, Fig. 1C and D) measuring 7.0 � 7.3 cm as well asretroperitoneal lymphadenopathy that included an aortocavalnode of 2.1 � 1.5 cm and paracaval node of 1.2 � 0.9 cm. Afluorodeoxyglucose-positron-emission tomography scan showedhypermetabolism in the left cervical and retroperitoneal lymphnodes. A fine needle aspiration (FNA) demonstrated a malignantneoplasm of large epithelioid cells with high nuclear to cyto-plasmic ratio, large and irregular nuclei, and coarse chromatinpattern. Immunohistochemical staining revealed likely positivityof tumor cells for PanMelanoma Cocktail (Mart-1, HMB 45,and tyrosinase) and focal weak staining for pancytokeratin (Fig.2A–C). These findings were deemed consistent with metastaticmalignant melanoma of unknown primary.

The patient was then referred tomedical oncology. No obviousskin lesions were observed by physical exam, while baselinelaboratory values were within normal limits with the exceptionof a lactate dehydrogenase (LDH) concentration of 461 U/L(normal range < 200 U/L). The patient began anti–PD-1 therapywith nivolumab, receiving a single 3 mg/kg dose. For consider-

ation of all potential melanoma treatment options, a core needlebiopsy was performed to obtain adequate tissue for molecularanalysis. In contrast with the FNA, histologic review of the corebiopsy specimen was consistent with NSGCT (embryonal cellcarcinoma) with immunohistochemistry positive for cytoker-atin (OSCAR antibody), CD30, and OCT 3/4 (Fig. 2D–F) andnegative for S-100, MART-1 (Fig. 2G), SOX10, PLAP, and TTF1/Napsin. Ultrasonography of the scrotum revealed no definitiveradiographic evidence of a primary testicular tumor; however,the right testicle was markedly atrophic compared with the left,a possible indication of an occult primary tumor. The initialserum tumor markers, measured after nivolumab treatment,included concentrations of a-fetoprotein (AFP) of 1.3 ng/mL(normal range < 8.4), b-human chorionic gonadotropin (HCG)of 3.1 IU/L (normal range < 1.4), and LDH of 388 U/L.Expressions of PD-1 and PD-L1 were determined by a com-mercial vendor (Pathline Progressive Pathology). TIL weredescribed as high-positive staining for PD-1 with >25% distri-bution and 2þ staining intensity, as well as low-positive stain-ing for PD-L1 on TIL with 1% to 24% distribution and 1þstaining intensity. Tumor cells were described as low-positivestaining for PD-L1 with 1% to 24% staining in cytoplasmic andmembranous location and 1þ intensity.

A follow-up CT scan after treatment with a single dose ofnivolumab noted the left supraclavicular mass measuring 5.3 �5.1 cm, aortocaval node 1 � 0.7 cm, and paracaval lymph node0.5 � 0.5 cm, consistent with 33% disease reduction by RECISTversion 1.1 and 49% regression by immune-related ResponseCriteria (14). On clinical exam, the lymphadenopathy in the neckwas no longer palpable.

Despite evidence for a rapid response to immunotherapy,tumor board consensus was to transition the patient to standardchemotherapy with bleomycin (30 units/dose, days 1, 8, and 15),etoposide (100 mg/m2, days 1 to 5), and cisplatin (20 mg/m2,

Figure 1.

CT images of tumor lesion in neck. A,before treatment axial view. B, beforetreatment coronal view. C, after onedose of nivolumab in axial view.D, after one dose of nivolumab incoronal view.

Shah et al.

Cancer Immunol Res; 4(11) November 2016 Cancer Immunology Research904

on July 25, 2020. © 2016 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst September 16, 2016; DOI: 10.1158/2326-6066.CIR-16-0087

Page 3: Clinical Response of a Patient to Anti PD-1 Immunotherapy ...Immunotherapy and the Immune Landscape of Testicular Germ Cell Tumors Shalin Shah1, James E.Ward1,2, Riyue Bao2, Curtis

days 1 to 5) administered on a 21-day cycle for 3 cycles, given thecurative intent of that approach. After chemotherapy, a radio-graphic complete response was documented and further declinein tumor markers was observed (LDH 263 IU/L, AFP 3.1 ng/mL,HCG < 0.6 IU/L). The patient subsequently underwent rightorchiectomy and left neck dissection with evidence of completepathologic response. As the retroperitoneal lymphadenopathyhad also resolved, resection of these sites was avoided consistentwith professional society guidelines (15).

Materials and MethodsAnalysis of TCGA data set

RNA-seq gene expression data, whole-exome sequencingsomatic mutation data, and clinical information of TCGA TGCTwere obtained fromBroadGenomeData Analysis Center (GDAC;release date November 1, 2015, level 4). A total of 134 primarytumor samples were included in this study. RNA-seq raw readcounts were summarized at gene level using the expectationmaximization (RSEM) method, followed by upper quartile-nor-malization and log2 transformation.

Identification of T-cell–inflamed and non–T-cell-inflamedgroups

The tumor groups were identified by our previously publishedmethod (13). In brief, unsupervised hierarchical clustering wasused to cluster 18,475 genes that are expressed in at least 50% ofthe samples into 12 groups based on similar expression patterns.A cluster of 1,015 genes including 12 genes of a previouslyidentified T-cell gene signature (CD8A, CCL3, CCL4, CXCL9,CXCL10, ICOS, IRF1, GZMK, HLA-DMA, HLA-DMB, HLA-DOA,HLA-DOB) was selected for identification of T-cell–inflamed,intermediate, and non–T-cell-inflamed tumor groups using aconsensus sample clustering method. Gene ontology (GO)enrichment analysis of the genes of interest was performed usingDAVID v6.7 (16).

AFP expression and correlation with IFNg-associated immunegenes

Tumors from TCGA were divided into five categories based onthe expression of AFP. Samples with 0 raw read counts (AFPnegative) were defined as no expression (category 1). For thosethat express AFP gene (AFP positive), the first, second, third, and

Figure 2.

Immunohistochemistry from pretreatment and on-treatment biopsy. All images, 40x magnification. A–C, pan-melanoma cocktail pre-treatment biopsy;D, H&E on-treatment biopsy; E, Oct3-4 on-treatment biopsy; F, CD30 on-treatment biopsy; G, Mart1 on-treatment biopsy.

Anti–PD-1 Response and Immune Landscape in Testicular Cancer

www.aacrjournals.org Cancer Immunol Res; 4(11) November 2016 905

on July 25, 2020. © 2016 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst September 16, 2016; DOI: 10.1158/2326-6066.CIR-16-0087

Page 4: Clinical Response of a Patient to Anti PD-1 Immunotherapy ...Immunotherapy and the Immune Landscape of Testicular Germ Cell Tumors Shalin Shah1, James E.Ward1,2, Riyue Bao2, Curtis

fourth quartiles of normalized AFP expression values (Q1 to Q4,equivalent to 25%, 50%, 75%, and 100%) were calculatedand used to bin the samples to categories 2 to 5. The distributionof T-cell–inflamed and non–T-cell-inflamed groups was calculat-ed in each AFP-positive or -negative category and vice versa.Seminoma and nonseminoma tumor types were retrieved fromthe patient histologic type in the TCGA clinical table. The Pear-son's product–moment correlation coefficient was calculatedbetween AFP and genes of interest. Significance of the association(P values)was computed using anR function correlation test, withthe alternative hypothesis being expecting "less" negative associ-ation between AFP and select genes.

ResultsTo better understand the spectrum of IFNg-associated T-cell

inflammation in GCTs, and by extension the relative likelihoodthat T-cell–based checkpoint immunotherapies will have clinicalutility, we analyzed of the gene expression data from the TGCTcohort of TCGA, separating patient tumors into T-cell–inflamed,intermediate, and non–T-cell-inflamed cohorts. The list of TCGAsamples and related information is provided in SupplementaryTable S1.

Spectrum of T-cell–inflamed and non–T-cell-inflamedgerm cell tumors

By clustering the 134 TGCT samples of TCGA based on theexpression of select 1,015 genes (Supplementary Table S2), weidentified three distinct tumor groups associated with low (non–T-cell-inflamed), high (T-cell–inflamed), and mixed (intermedi-ate) expression of the T-cell gene signature (Fig. 3). Overall, thepresence of the T-cell–inflamed tumor microenvironment was

identified in 47% of TGCT and the non–T-cell-inflamed tumormicroenvironment in 32% (with 21% intermediate). GO analysissuggested that expression of these genes is significantly enrichedin biological processes that include immune responses, defenseresponses, positive regulation of immune system processes, andleukocyte activation (Supplementary Table S3).

AFP expression inversely correlates with a T-cell–inflamedtumor microenvironment

To explore the role of the T-cell–inflamed tumor microenvi-ronment in GCT, we evaluated clinical factors, including non-seminoma and seminomatous histology. In the seminoma pop-ulation, 75%of samples were T-cell–inflamed, 19% intermediate,and6%non–T-cell-inflamed. In contrast, within nonseminoma, amajority of samples were non–T-cell-inflamed at 60%, with 23%intermediate, and 17% inflamed.

Based on the finding of a higher percentage of T-cell–inflamedphenotype in seminoma compared with that of nonseminoma,the relative impact ofAFP expression in the T-cell–inflamed tumormicroenvironment was investigated. We categorized AFP expres-sion into five levels, from no expression through four quartiles,and calculated the relative abundance of each category within theoverall T-cell–inflamed, intermediate, and non–T-cell-inflamedgroups. T-cell–inflamed tumors with no AFP expression made up51% of the samples, with another 26% having 1st quartile AFPexpression. Conversely, the non–T-cell-inflamed population wasmade up of 35% 3rd quartile and 25% 4th quartile AFP expres-sion. The second quartile AFP expression was similar acrossinflamed, intermediate, and noninflamed tumors at 14%, 14%,and 19%, respectively. Distribution of AFP expression by quartileis shown relative to tumor groups in Fig. 4A. Principal componentanalysis confirmed associations of no/lowAFP cohorts with T-cell

Figure 3.

Gene expression heatmap of non–T-cell-inflamed, intermediate, and T-cell–inflamed testicular germ cell tumorsfrom TCGA. Genes are on the row, andsamples on the column. Two heatmappanels are presented. Top smaller panelshows the expression pattern of HCGgenes (CGA and CGB 1/2/3/5/7/8).Bottom larger panel shows that of 1,015select genes used to categorize samplesinto T-cell–inflamed and non–T-cell-inflamed tumor groups, with the geneclustering dendrogram shown to the leftside. Blue, red, and green horizontal barsindicate tumor groups associated withlow (non–T-cell-inflamed), high (T-cell–inflamed), and mixed (intermediate)expression of the T-cell gene signature.Three horizontal annotation bars areadded above the heatmap (from top tothe bottom): consensus clusterassignment of 134 primary tumors(clusters 1 to 12); samples colored by AFPexpression levels; seminoma (red) andnonseminoma samples (black).

Shah et al.

Cancer Immunol Res; 4(11) November 2016 Cancer Immunology Research906

on July 25, 2020. © 2016 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst September 16, 2016; DOI: 10.1158/2326-6066.CIR-16-0087

Page 5: Clinical Response of a Patient to Anti PD-1 Immunotherapy ...Immunotherapy and the Immune Landscape of Testicular Germ Cell Tumors Shalin Shah1, James E.Ward1,2, Riyue Bao2, Curtis

inflammation and high AFP cohorts with a lack of T-cell inflam-mation, with 77% of T-cell–inflamed tumors having no/low AFPexpression (noAFP: 51%; 1st quartileAFP: 26%), whereas 60%ofnon–T-cell-inflamed tumors (3rd quartile AFP: 35%; 4th quartileAFP: 25%) had high AFP expression. Additional analysis revealedstatistically significant inverse correlations between AFP andIFNg-associated genes including CD8A, GZMB, PD-L1, FOXP3,BATF3, and IRF8 (Fig. 4B). A similar analysis was performed onHCG subunit genes (CGA, CGB1, CGB2, CGB3, CGB5, CGB7, andCGB8) with no significant associations observed with T-cellinflammation.

DiscussionHere, we report a RECIST quality partial response to anti–PD-1

immunotherapy in a patient with advanced NSGCT. This casesuggests that further investigation of immunotherapy in thisdisease is warranted. Our data analysis and assessment werehampered by the inadequate availability of pretreatment tissuefor histologic diagnosis. Therefore, it is clearly a necessity forrobust and high-quality tissue biopsy specimens to be collected

and preserved for later analysis, before initiation of anticancertreatments.

Building upon our clinical case report describing effectivetreatment of a patient with NSGCT with anti–PD-1, we analyzedthe presence or absence of a T-cell–inflamed tumor microenvi-ronment of TGCT utilizing an IFNg-associated gene set. Overall,47% of TGCT had the T-cell–inflamed tumor microenvironmentand 32%had the non–T-cell-inflamed tumormicroenvironment.The T-cell–inflamed tumor microenvironment was more likely inseminomatous TGCT and became less frequent in NSGCT as AFPexpression increased. Regarding AFP specifically, its expressioninversely correlated with several known molecular mediators ofimmunotherapy.

Immunotherapy with checkpoint-blocking approaches, espe-cially antibodies to PD-1 and PD-L1, has changed the standard ofcare for several tumor histologies and is being investigated inmany cancers. Multiple biomarkers are being pursued to findthose that can predict benefit from checkpoint-blockade immu-notherapy, including expression of PD-L1 (17) and the presenceof infiltrating TIL (18), although both of these are components ofan effective IFNg-associated T-cell response. Thus, gene expressionprofiling has the potential to be a more encompassing modality,because it reflects the tumor–immune system interaction morebroadly.

Despite high cure rates for TGCT, investigation into theimmune response associated with these diseases is of increasingurgency given the unmet needs of refractory patients and the long-term toxicities associated with chemotherapy (19). Immunohis-tochemical analysis for PD-L1 expression has been reported to bepresent in 73% of seminomas, 64% of nonseminoma, and nostaining in intratubular germ cell neoplasia or normal testicularspecimens (20). Substantial stromal expression of PD-L1was alsoreported, though not in direct comparison with correspondingmalignant cells. The presence of PD-L1 expression by immuno-histochemistry has been suggested as a prognostic marker inTGCT with a correlation between low PD-L1 expression andimproved progression-free and overall survival to nonimmu-notherapy interventions (21). A previous bioinformatic investi-gation of TCGA suggested high PD-L1 and CD8A expression in48% of TGCT samples (22). This is in line with our observationsusing a T-cell gene signature. Previously,wepresentedpreliminarydata on an extended 160 gene T-cell signature across TCGA,describing the TGCT cohort as one of the most highly T-cell–inflamed, based on percentage of samples with that signatureamong solid tumors (as in other PD-1 blockade–responsivecancers, including clear cell kidney cancer and lung adenocarci-noma; ref. 23). The dual PD-L1 and CD8A approach above alsoranks TGCT highly relative to other tumors, adding further sup-port to the concept of high T-cell inflammation in TGCT. Anadditional advantage of our approach, however, lies in the def-inition of the non–T-cell-inflamed tumor microenvironment, inwhich we can identify factors driving the immune-exclusionphenotype.

The presence of the T-cell–inflamed tumor microenvironmentwas inversely associated with AFP expression. While this obser-vation requires prospective validation, it is of interest to considerin the context of the known clinical management of GCTs inwhich concentration of AFP can partially guide the selection ofchemotherapy. The intersection of chemotherapy and immuno-therapy is of relevance inTGCT, given recent reports suggesting thepotential for platinum in sensitizing tumors to checkpoint

Figure 4.

Distribution of AFP expression quartiles within non–T-cell-inflamed,intermediate, and T-cell–inflamed tumor groups. A, samples were categorizedinto five bins based on the expression of AFP: 1, no expression (AFP-);2–5, 1st, 2nd, 3rd, and 4th quartile of AFP expression level (Q1–Q4, AFPþ).B, correlation plots of AFP vs. CD8A, GZMB, PD-L1, FOXP3, BATF3, and IRF8.P values of the Pearson's product–moment correlation are indicated aboveeach panel. Blue and red colors indicate the noninflamed and inflamed tumorgroup, respectively.

Anti–PD-1 Response and Immune Landscape in Testicular Cancer

www.aacrjournals.org Cancer Immunol Res; 4(11) November 2016 907

on July 25, 2020. © 2016 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst September 16, 2016; DOI: 10.1158/2326-6066.CIR-16-0087

Page 6: Clinical Response of a Patient to Anti PD-1 Immunotherapy ...Immunotherapy and the Immune Landscape of Testicular Germ Cell Tumors Shalin Shah1, James E.Ward1,2, Riyue Bao2, Curtis

blockade in preclinical models (24) and clinical reports frommultiple tumors of preliminary clinical success combining thesemodalities (25).

We acknowledge limitations to the data analyses we haveadvanced here. Although presentation of a case report thatdemonstrates a clinical response to anti–PD-1 in TGCT suggeststhe potential for clinical benefit, patients are heterogeneous,making extrapolation of the significance of this case to clinicalpractice difficult. This patient may have had good-risk, platinum-sensitive GCT, which is not the population of GCT patients mostin need of novel treatments. We also recognize that the patientscomprising the TCGA data were not treated with immunotherapyand do not represent a homogenous population of patients.Therefore, reference to any individual patient clinical outcomesis not possible and will necessitate prospective studies. In addi-tion, NSGCT are a heterogeneous group of disease histologies andthat the use of AFP as a broad marker will require furtherrefinement, particularly the presence of T-cell inflammation inprimary testis tumors versusmetastatic disease. Given the inferredimmune privilege of the testes under physiologic conditions,tumors arising in a different setting could have different proper-ties. We are not aware of research investigating differences in theimmune response between anatomic sites in germ cell tumors,and sample sizeswithin TCGAwere inadequate to address this in arobust manner.

In summary, we have described a RECIST partial response in apatient with NSGCT, via a single dose of anti–PD-1 and havefurther described the immune landscape of TGCTs. In particular, aT-cell gene signature in seminomatous GCT was enriched relativeto NSGCT, and the presence of the T-cell–inflamed tumor micro-environment and the expression of AFPwere inversely correlated.These findings could have immediate impact for clinical transla-tion, given that they suggest that a substantial fraction ofGCTmaybe amendable to targeting of T-cell inflammation through

approaches such as blockade of PD-1. A phase II study of anti–PD-1 in GCT has been launched (Clinicaltrials.gov identifier:NCT02499952), and results are eagerly awaited.

Disclosure of Potential Conflicts of InterestNo potential conflicts of interest were disclosed.

Authors' ContributionsConception and design: S. Shah, J.E. Ward, B.E. Brockstein, J.J. LukeDevelopment of methodology: R. Bao, B.E. Brockstein, J.J. LukeAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): S. Shah, J.E. Ward, B.E. Brockstein, J.J. LukeAnalysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis): S. Shah, R. Bao, J.J. LukeWriting, review, and/or revision of the manuscript: S. Shah, J.E. Ward, R. Bao,C.R. Hall, B.E. Brockstein, J.J. LukeAdministrative, technical, or material support (i.e., reporting or organizingdata, constructing databases): J.E. Ward, B.E. Brockstein, J.J. LukeStudy supervision: B.E. Brockstein, J.J. LukeOther (reviewed and interpreted the tissue from the referenced patient toarrive at a pathologic diagnosis): C.R. Hall

AcknowledgmentsWe recognize Thomas A. Victor, MD, PhD, for helpful contributions to this

research.

Grant SupportJ.J. Luke was supported by Paul Calabresi Career Development in Clinical

Oncology Award (NIH 1K12CA139160-05), Young Investigator Award fromthe Cancer Research Foundation, and the Arthur J Schreiner Family MelanomaResearch Fund, with support from The Center for Research Informatics of TheUniversity of Chicago Biological Science Division and The Institute for Trans-lational Medicine/CTSA (NIH UL1 RR024999).

Received April 24, 2016; revised August 17, 2016; accepted August 17, 2016;published OnlineFirst September 16, 2016.

References1. HannaNH, Einhorn LH. Testicular cancer–discoveries and updates. N Engl

J Med 2014;371:2005–16.2. InternationalGerm Cell Consensus Classification: A prognostic factor-

based staging system for metastatic germ cell cancers. International GermCell Cancer Collaborative Group. J Clin Oncol 1997;15:594–603.

3. Edge SB, American Joint Committee on Cancer. AJCC cancer stagingmanual. 7th ed. New York, NY: Springer; 2010.

4. Loehrer PJSr, Lauer R, Roth BJ, Williams SD, Kalasinski LA, EinhornLH. Salvage therapy in recurrent germ cell cancer: Ifosfamide andcisplatin plus either vinblastine or etoposide. Ann Intern Med 1988;109:540–6.

5. Feldman DR, Sheinfeld J, Bajorin DF, Fischer P, Turkula S, Ishill N, et al.TI-CE high-dose chemotherapy for patients with previously treated germcell tumors: Results and prognostic factor analysis. J Clin Oncol 2010;28:1706–13.

6. Abouassaly R, Fossa SD, Giwercman A, Kollmannsberger C, Motzer RJ,Schmoll HJ, et al. Sequelae of treatment in long-term survivors of testiscancer. Eur Urol 2011;60:516–26.

7. Larkin J, Chiarion-Sileni V, Gonzalez R, Grob JJ, Cowey CL, Lao CD, et al.Combined nivolumab and ipilimumab or monotherapy in untreatedmelanoma. N Engl J Med 2015;373:23–34.

8. Motzer RJ, Escudier B, McDermott DF, George S, Hammers HJ, Srinivas S,et al. Nivolumab versus everolimus in advanced renal-cell carcinoma.N Engl J Med 2015;373:1803–13.

9. Spranger S, Spaapen RM, Zha Y, Williams J, Meng Y, Ha TT, et al. Up-regulation of PD-L1, IDO, and T(regs) in the melanoma tumor microen-vironment is driven by CD8(þ) T cells. Sci Transl Med 2013;5:200ra116.

10. Harlin H, Meng Y, Peterson AC, Zha Y, Tretiakova M, Slingluff C, et al.Chemokine expression in melanoma metastases associated with CD8þT-cell recruitment. Cancer Res 2009;69:3077–85.

11. Ribas A, Robert C, Hodi FS, Wolchok JD, Joshua AM, Hwu WJ, et al.Association of response to programmed death receptor 1 (PD-1) blockadewith pembrolizumab (MK-3475) with an interferon-inflammatoryimmune gene signature. J Clin Oncol 33, 2015(suppl; abstr 3001).

12. Ji RR, Chasalow SD, Wang L, Hamid O, Schmidt H, Cogswell J, et al. Animmune-active tumor microenvironment favors clinical response to ipi-limumab. Cancer Immunol Immunother 2012;61:1019–31.

13. Spranger S, BaoR,Gajewski TF.Melanoma-intrinsic beta-catenin signallingprevents anti-tumour immunity. Nature 2015;523:231–5.

14. Wolchok JD, Hoos A, O'Day S, Weber JS, Hamid O, Lebb�e C, et al.Guidelines for the evaluation of immune therapy activity in solid tumors:Immune-related response criteria. Clin Cancer Res 2009;15:7412–20.

15. Motzer RJ, Jonasch E, Agarwal N, Beard C, Bhayani S, Bolger GB, et al.Testicular Cancer, Version 2.2015. J Natl Compr Canc Netw 2015;13:772–99.

16. HuangdaW, ShermanBT, Lempicki RA. Systematic and integrative analysisof large gene lists using DAVID bioinformatics resources. Nat Protoc2009;4:44–57.

17. Robert C, Schachter J, Long GV, Arance A, Grob JJ, Mortier L, et al.Pembrolizumab versus ipilimumab in advanced melanoma. N Engl J Med2015;372:2521–32.

18. Tumeh PC, Harview CL, Yearley JH, Shintaku IP, Taylor EJ, Robert L, et al.PD-1 blockade induces responses by inhibiting adaptive immune resis-tance. Nature 2014;515:568–71.

Shah et al.

Cancer Immunol Res; 4(11) November 2016 Cancer Immunology Research908

on July 25, 2020. © 2016 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst September 16, 2016; DOI: 10.1158/2326-6066.CIR-16-0087

Page 7: Clinical Response of a Patient to Anti PD-1 Immunotherapy ...Immunotherapy and the Immune Landscape of Testicular Germ Cell Tumors Shalin Shah1, James E.Ward1,2, Riyue Bao2, Curtis

19. Willemse PM, Burggraaf J, Hamdy NA,Weijl NI, Vossen CY, vanWulften L,et al. Prevalence of themetabolic syndrome and cardiovascular disease riskin chemotherapy-treated testicular germ cell tumour survivors. Br J Cancer2013;109:60–7.

20. Fankhauser CD, Curioni-Fontecedro A, Allmann V, Beyer J, Tischler V,Sulser T, et al. Frequent PD-L1 expression in testicular germ cell tumors.Br J Cancer 2015;113:411–3.

21. Cierna Z, Mego M, Miskovska V, Machalekova K, Chovanec M, Sve-tlovska D, et al. Prognostic value of programmed-death-1 receptor(PD-1) and its ligand 1 (PD-L1) in testicular germ cell tumors. AnnOncol 2016;27:300–5.

22. Ock CY, Keam B, Kim S, Lee JS, Kim M, Kim TM, et al. Pan-cancerimmunogenomic perspective on the tumor microenvironment based onPD-L1 and CD8 T-cell infiltration. Clin Cancer Res 2016;22:2261–70.

23. Luke JJ, Spranger S, Bao R, Andrade J, Gajewski TF. The genetic landscape ofthe T-cell non-inflamed tumormicroenvironment in human solid tumors.J Immunother Cancer 2015;3:P97–P.

24. Pfirschke C, Engblom C, Rickelt S, Cortez-Retamozo V, Garris C, Pucci F,et al. Immunogenic chemotherapy sensitizes tumors to checkpoint block-ade therapy. Immunity 2016;44:343–54.

25. Camidge R, Liu SV, Powderly J, Ready N, Hodi S, Gettinger SN, et al.Atezolizumab (MPDL3280A) combined with platinum-based chemo-therapy in non-small cell lung cancer (NSCLC): A phase Ib safety andefficacy update [abstract]. In: Proceedings of the the 16th World Con-ference on Lung Cancer; September 6–9; Denver, CO. Abstract nr 0207.Available from: http://library.iaslc.org/virtual-library-search?produc-t_id=1&author=&category=&date=&session_type=&session=&presenta-tion=&keyword=&page=4 accessed September 23rd, 2016.

www.aacrjournals.org Cancer Immunol Res; 4(11) November 2016 909

Anti–PD-1 Response and Immune Landscape in Testicular Cancer

on July 25, 2020. © 2016 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst September 16, 2016; DOI: 10.1158/2326-6066.CIR-16-0087

Page 8: Clinical Response of a Patient to Anti PD-1 Immunotherapy ...Immunotherapy and the Immune Landscape of Testicular Germ Cell Tumors Shalin Shah1, James E.Ward1,2, Riyue Bao2, Curtis

2016;4:903-909. Published OnlineFirst September 16, 2016.Cancer Immunol Res   Shalin Shah, James E. Ward, Riyue Bao, et al.   the Immune Landscape of Testicular Germ Cell Tumors

PD-1 Immunotherapy and−Clinical Response of a Patient to Anti

  Updated version

  10.1158/2326-6066.CIR-16-0087doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://cancerimmunolres.aacrjournals.org/content/suppl/2016/09/16/2326-6066.CIR-16-0087.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://cancerimmunolres.aacrjournals.org/content/4/11/903.full#ref-list-1

This article cites 22 articles, 7 of which you can access for free 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/4/11/903To request permission to re-use all or part of this article, use this link

on July 25, 2020. © 2016 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst September 16, 2016; DOI: 10.1158/2326-6066.CIR-16-0087