Anti–Programed 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 anti–PD-1 nor the incidence of an immunoresponsive tumor microenvironment has been described for GCTs. A patient initially diagnosed with melanoma via fine needle aspiration was treated with one dose of antibody to PD-1. A core needle biopsy was subsequently performed to acquire sufficient 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 benefit from immunotherapy. Primary tumors (N = 134) were categorized as high (T-cell–inflamed), medium, or low (non–T-cell-inflamed) by their T-cell signature derived from RNAseq data. Anti–PD-1 induced decreases in serum markers 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%) tumor subtypes. Expression of alpha-fetoprotein (AFP) RNA correlated with lack of the T-cell signature, with increasing AFP RNA inversely correlating with the inflamed signature and expression of IFNγ-associated genes. These data suggest that GCTs can respond to anti–PD-1 and that gene expression profiling supports investigation of immunotherapy for treatment of GCTs. Cancer Immunol Res; 4(11); 903–9. ©2016 AACR.

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 nonseminomatous germ cell tumor (NSGCT) with treatment dictated both by American Joint Committee on Cancer and risk modeling–based assessment (2, 3). Early-stage germ cell tumors respond well to orchiectomy and adjuvant radiotherapy or chemotherapy. After using a risk stratification system for more advanced disease, germ cell tumors are commonly treated with 3 to 4 cycles of chemotherapy. Though disease refractory to initial chemotherapy can be treated with effective second- and sometimes third-line chemotherapy (4, 5), progressive chemotherapy is associated with significant toxicity. Long-term sequelae of treatment include second malignancies, hypogonadism, infertility, and cardiovascular disease (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 anti–cytotoxic T lymphocyte antigen 4 (CTLA-4) and anti–programmed death 1 (PD-1)/programmed death-ligand 1 (PD-L1). These approaches have changed clinical paradigms in melanoma, non–small cell lung cancer, and clear cell renal cell carcinoma, in which phase III trials have documented improvements in overall survival and an improvement in toxicity profile 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 efficacy of immune-checkpoint immunotherapy, the majority of patients currently do not respond. Multiple predictive biomarkers for immunotherapy have been put forward, particularly immunohistochemistry for PD-L1 or tumor-infiltrating lymphocytes (TIL). However, each of these is only an aspect of an effective antitumor immune response, in which infiltrating T cells elaborate cytokines such as IFNγ that leads to upregulation of immune effector molecules (9). A more robust approach to tumor microenvironmental evaluation may thus be facilitated by gene expression profiling where an active antitumor immune environment has been coined the “T cell–inflamed tumor microenvironment” (10). This phenotype has prognostic significance and is associated with the presence of microenvironmental factors—such 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).

Here, we describe the effective treatment of a patient with NSGCT with anti–PD-1 immunotherapy and then describe an analysis of the TGCT cohort of The Cancer Genome Atlas (TCGA). Based on gene expression profiling using a previously described T-cell gene signature (13), we found that a substantial proportion of GCTs are likely to be susceptible to checkpoint immunotherapy. Finally, we identified a potential biochemical mediator of immunosuppression whose potential warrants further investigation in prospective clinical research.

A 32-year-old Caucasian male developed a palpable mass on the left of his neck without other symptoms. CT showed a large confluent nodal mass in the left neck (pretreatment, Fig. 1A and B; posttreatment, Fig. 1C and D) measuring 7.0 × 7.3 cm as well as retroperitoneal lymphadenopathy that included an aortocaval node of 2.1 × 1.5 cm and paracaval node of 1.2 × 0.9 cm. A fluorodeoxyglucose-positron-emission tomography scan showed hypermetabolism in the left cervical and retroperitoneal lymph nodes. A fine needle aspiration (FNA) demonstrated a malignant neoplasm of large epithelioid cells with high nuclear to cytoplasmic ratio, large and irregular nuclei, and coarse chromatin pattern. Immunohistochemical staining revealed likely positivity of 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 metastatic malignant melanoma of unknown primary.

Figure 1.

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

Figure 1.

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

Close modal
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.

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.

Close modal

The patient was then referred to medical oncology. No obvious skin lesions were observed by physical exam, while baseline laboratory values were within normal limits with the exception of a lactate dehydrogenase (LDH) concentration of 461 U/L (normal range < 200 U/L). The patient began anti–PD-1 therapy with nivolumab, receiving a single 3 mg/kg dose. For consideration of all potential melanoma treatment options, a core needle biopsy was performed to obtain adequate tissue for molecular analysis. In contrast with the FNA, histologic review of the core biopsy specimen was consistent with NSGCT (embryonal cell carcinoma) with immunohistochemistry positive for cytokeratin (OSCAR antibody), CD30, and OCT 3/4 (Fig. 2D–F) and negative for S-100, MART-1 (Fig. 2G), SOX10, PLAP, and TTF1/Napsin. Ultrasonography of the scrotum revealed no definitive radiographic 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 initial serum tumor markers, measured after nivolumab treatment, included concentrations of α-fetoprotein (AFP) of 1.3 ng/mL (normal range < 8.4), β-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 commercial vendor (Pathline Progressive Pathology). TIL were described as high-positive staining for PD-1 with >25% distribution and 2+ staining intensity, as well as low-positive staining for PD-L1 on TIL with 1% to 24% distribution and 1+ staining intensity. Tumor cells were described as low-positive staining for PD-L1 with 1% to 24% staining in cytoplasmic and membranous location and 1+ intensity.

A follow-up CT scan after treatment with a single dose of nivolumab noted the left supraclavicular mass measuring 5.3 × 5.1 cm, aortocaval node 1 × 0.7 cm, and paracaval lymph node 0.5 × 0.5 cm, consistent with 33% disease reduction by RECIST version 1.1 and 49% regression by immune-related Response Criteria (14). On clinical exam, the lymphadenopathy in the neck was no longer palpable.

Despite evidence for a rapid response to immunotherapy, tumor board consensus was to transition the patient to standard chemotherapy with bleomycin (30 units/dose, days 1, 8, and 15), etoposide (100 mg/m2, days 1 to 5), and cisplatin (20 mg/m2, days 1 to 5) administered on a 21-day cycle for 3 cycles, given the curative intent of that approach. After chemotherapy, a radiographic complete response was documented and further decline in tumor markers was observed (LDH 263 IU/L, AFP 3.1 ng/mL, HCG < 0.6 IU/L). The patient subsequently underwent right orchiectomy and left neck dissection with evidence of complete pathologic response. As the retroperitoneal lymphadenopathy had also resolved, resection of these sites was avoided consistent with professional society guidelines (15).

Analysis of TCGA data set

RNA-seq gene expression data, whole-exome sequencing somatic mutation data, and clinical information of TCGA TGCT were obtained from Broad Genome Data Analysis Center (GDAC; release date November 1, 2015, level 4). A total of 134 primary tumor samples were included in this study. RNA-seq raw read counts were summarized at gene level using the expectation maximization (RSEM) method, followed by upper quartile-normalization and log2 transformation.

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

The tumor groups were identified by our previously published method (13). In brief, unsupervised hierarchical clustering was used to cluster 18,475 genes that are expressed in at least 50% of the samples into 12 groups based on similar expression patterns. A cluster of 1,015 genes including 12 genes of a previously identified 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 a consensus sample clustering method. Gene ontology (GO) enrichment analysis of the genes of interest was performed using DAVID v6.7 (16).

AFP expression and correlation with IFNγ-associated immune genes

Tumors from TCGA were divided into five categories based on the expression of AFP. Samples with 0 raw read counts (AFP negative) were defined as no expression (category 1). For those that express AFP gene (AFP positive), the first, second, third, and fourth quartiles of normalized AFP expression values (Q1 to Q4, equivalent to 25%, 50%, 75%, and 100%) were calculated and used to bin the samples to categories 2 to 5. The distribution of T-cell–inflamed and non–T-cell-inflamed groups was calculated in each AFP-positive or -negative category and vice versa. Seminoma and nonseminoma tumor types were retrieved from the patient histologic type in the TCGA clinical table. The Pearson's product–moment correlation coefficient was calculated between AFP and genes of interest. Significance of the association (P values) was computed using an R function correlation test, with the alternative hypothesis being expecting “less” negative association between AFP and select genes.

To better understand the spectrum of IFNγ-associated T-cell inflammation in GCTs, and by extension the relative likelihood that T-cell–based checkpoint immunotherapies will have clinical utility, we analyzed of the gene expression data from the TGCT cohort of TCGA, separating patient tumors into T-cell–inflamed, intermediate, and non–T-cell-inflamed cohorts. The list of TCGA samples and related information is provided in Supplementary Table S1.

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

By clustering the 134 TGCT samples of TCGA based on the expression of select 1,015 genes (Supplementary Table S2), we identified three distinct tumor groups associated with low (non–T-cell-inflamed), high (T-cell–inflamed), and mixed (intermediate) expression of the T-cell gene signature (Fig. 3). Overall, the presence of the T-cell–inflamed tumor microenvironment was identified in 47% of TGCT and the non–T-cell-inflamed tumor microenvironment in 32% (with 21% intermediate). GO analysis suggested that expression of these genes is significantly enriched in biological processes that include immune responses, defense responses, positive regulation of immune system processes, and leukocyte activation (Supplementary Table S3).

Figure 3.

Gene expression heatmap of non–T-cell-inflamed, intermediate, and T-cell–inflamed testicular germ cell tumors from TCGA. Genes are on the row, and samples on the column. Two heatmap panels are presented. Top smaller panel shows the expression pattern of HCG genes (CGA and CGB 1/2/3/5/7/8). Bottom larger panel shows that of 1,015 select genes used to categorize samples into T-cell–inflamed and non–T-cell-inflamed tumor groups, with the gene clustering dendrogram shown to the left side. Blue, red, and green horizontal bars indicate tumor groups associated with low (non–T-cell-inflamed), high (T-cell–inflamed), and mixed (intermediate) expression of the T-cell gene signature. Three horizontal annotation bars are added above the heatmap (from top to the bottom): consensus cluster assignment of 134 primary tumors (clusters 1 to 12); samples colored by AFP expression levels; seminoma (red) and nonseminoma samples (black).

Figure 3.

Gene expression heatmap of non–T-cell-inflamed, intermediate, and T-cell–inflamed testicular germ cell tumors from TCGA. Genes are on the row, and samples on the column. Two heatmap panels are presented. Top smaller panel shows the expression pattern of HCG genes (CGA and CGB 1/2/3/5/7/8). Bottom larger panel shows that of 1,015 select genes used to categorize samples into T-cell–inflamed and non–T-cell-inflamed tumor groups, with the gene clustering dendrogram shown to the left side. Blue, red, and green horizontal bars indicate tumor groups associated with low (non–T-cell-inflamed), high (T-cell–inflamed), and mixed (intermediate) expression of the T-cell gene signature. Three horizontal annotation bars are added above the heatmap (from top to the bottom): consensus cluster assignment of 134 primary tumors (clusters 1 to 12); samples colored by AFP expression levels; seminoma (red) and nonseminoma samples (black).

Close modal

AFP expression inversely correlates with a T-cell–inflamed tumor microenvironment

To explore the role of the T-cell–inflamed tumor microenvironment in GCT, we evaluated clinical factors, including nonseminoma and seminomatous histology. In the seminoma population, 75% of samples were T-cell–inflamed, 19% intermediate, and 6% non–T-cell-inflamed. In contrast, within nonseminoma, a majority 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–inflamed phenotype in seminoma compared with that of nonseminoma, the relative impact of AFP expression in the T-cell–inflamed tumor microenvironment was investigated. We categorized AFP expression into five levels, from no expression through four quartiles, and calculated the relative abundance of each category within the overall T-cell–inflamed, intermediate, and non–T-cell-inflamed groups. T-cell–inflamed tumors with no AFP expression made up 51% of the samples, with another 26% having 1st quartile AFP expression. Conversely, the non–T-cell-inflamed population was made up of 35% 3rd quartile and 25% 4th quartile AFP expression. The second quartile AFP expression was similar across inflamed, intermediate, and noninflamed tumors at 14%, 14%, and 19%, respectively. Distribution of AFP expression by quartile is shown relative to tumor groups in Fig. 4A. Principal component analysis confirmed associations of no/low AFP cohorts with T-cell inflammation and high AFP cohorts with a lack of T-cell inflammation, with 77% of T-cell–inflamed tumors having no/low AFP expression (no AFP: 51%; 1st quartile AFP: 26%), whereas 60% of non–T-cell-inflamed tumors (3rd quartile AFP: 35%; 4th quartile AFP: 25%) had high AFP expression. Additional analysis revealed statistically significant inverse correlations between AFP and IFNγ-associated genes including CD8A, GZMB, PD-L1, FOXP3, BATF3, and IRF8 (Fig. 4B). A similar analysis was performed on HCG subunit genes (CGA, CGB1, CGB2, CGB3, CGB5, CGB7, and CGB8) with no significant associations observed with T-cell inflammation.

Figure 4.

Distribution of AFP expression quartiles within non–T-cell-inflamed, intermediate, and T-cell–inflamed tumor groups. A, samples were categorized into 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, AFP+). 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 above each panel. Blue and red colors indicate the noninflamed and inflamed tumor group, respectively.

Figure 4.

Distribution of AFP expression quartiles within non–T-cell-inflamed, intermediate, and T-cell–inflamed tumor groups. A, samples were categorized into 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, AFP+). 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 above each panel. Blue and red colors indicate the noninflamed and inflamed tumor group, respectively.

Close modal

Here, we report a RECIST quality partial response to anti–PD-1 immunotherapy in a patient with advanced NSGCT. This case suggests that further investigation of immunotherapy in this disease is warranted. Our data analysis and assessment were hampered by the inadequate availability of pretreatment tissue for histologic diagnosis. Therefore, it is clearly a necessity for robust and high-quality tissue biopsy specimens to be collected and preserved for later analysis, before initiation of anticancer treatments.

Building upon our clinical case report describing effective treatment of a patient with NSGCT with anti–PD-1, we analyzed the presence or absence of a T-cell–inflamed tumor microenvironment of TGCT utilizing an IFNγ-associated gene set. Overall, 47% of TGCT had the T-cell–inflamed tumor microenvironment and 32% had the non–T-cell-inflamed tumor microenvironment. The T-cell–inflamed tumor microenvironment was more likely in seminomatous TGCT and became less frequent in NSGCT as AFP expression increased. Regarding AFP specifically, its expression inversely correlated with several known molecular mediators of immunotherapy.

Immunotherapy with checkpoint-blocking approaches, especially antibodies to PD-1 and PD-L1, has changed the standard of care for several tumor histologies and is being investigated in many cancers. Multiple biomarkers are being pursued to find those that can predict benefit from checkpoint-blockade immunotherapy, including expression of PD-L1 (17) and the presence of infiltrating TIL (18), although both of these are components of an effective IFNγ-associated T-cell response. Thus, gene expression profiling has the potential to be a more encompassing modality, because it reflects the tumor–immune system interaction more broadly.

Despite high cure rates for TGCT, investigation into the immune response associated with these diseases is of increasing urgency given the unmet needs of refractory patients and the long-term toxicities associated with chemotherapy (19). Immunohistochemical analysis for PD-L1 expression has been reported to be present in 73% of seminomas, 64% of nonseminoma, and no staining in intratubular germ cell neoplasia or normal testicular specimens (20). Substantial stromal expression of PD-L1 was also reported, though not in direct comparison with corresponding malignant cells. The presence of PD-L1 expression by immunohistochemistry has been suggested as a prognostic marker in TGCT with a correlation between low PD-L1 expression and improved progression-free and overall survival to nonimmunotherapy interventions (21). A previous bioinformatic investigation of TCGA suggested high PD-L1 and CD8A expression in 48% of TGCT samples (22). This is in line with our observations using a T-cell gene signature. Previously, we presented preliminary data 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 signature among solid tumors (as in other PD-1 blockade–responsive cancers, including clear cell kidney cancer and lung adenocarcinoma; ref. 23). The dual PD-L1 and CD8A approach above also ranks TGCT highly relative to other tumors, adding further support to the concept of high T-cell inflammation in TGCT. An additional advantage of our approach, however, lies in the definition of the non–T-cell-inflamed tumor microenvironment, in which we can identify factors driving the immune-exclusion phenotype.

The presence of the T-cell–inflamed tumor microenvironment was inversely associated with AFP expression. While this observation requires prospective validation, it is of interest to consider in the context of the known clinical management of GCTs in which concentration of AFP can partially guide the selection of chemotherapy. The intersection of chemotherapy and immunotherapy is of relevance in TGCT, given recent reports suggesting the potential for platinum in sensitizing tumors to checkpoint blockade in preclinical models (24) and clinical reports from multiple tumors of preliminary clinical success combining these modalities (25).

We acknowledge limitations to the data analyses we have advanced here. Although presentation of a case report that demonstrates a clinical response to anti–PD-1 in TGCT suggests the potential for clinical benefit, patients are heterogeneous, making extrapolation of the significance of this case to clinical practice difficult. This patient may have had good-risk, platinum-sensitive GCT, which is not the population of GCT patients most in need of novel treatments. We also recognize that the patients comprising the TCGA data were not treated with immunotherapy and do not represent a homogenous population of patients. Therefore, reference to any individual patient clinical outcomes is not possible and will necessitate prospective studies. In addition, NSGCT are a heterogeneous group of disease histologies and that the use of AFP as a broad marker will require further refinement, particularly the presence of T-cell inflammation in primary testis tumors versus metastatic disease. Given the inferred immune privilege of the testes under physiologic conditions, tumors arising in a different setting could have different properties. We are not aware of research investigating differences in the immune response between anatomic sites in germ cell tumors, and sample sizes within TCGA were inadequate to address this in a robust manner.

In summary, we have described a RECIST partial response in a patient with NSGCT, via a single dose of anti–PD-1 and have further described the immune landscape of TGCTs. In particular, a T-cell gene signature in seminomatous GCT was enriched relative to NSGCT, and the presence of the T-cell–inflamed tumor microenvironment and the expression of AFP were inversely correlated. These findings could have immediate impact for clinical translation, given that they suggest that a substantial fraction of GCT may be 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.

No potential conflicts of interest were disclosed.

Conception and design: S. Shah, J.E. Ward, B.E. Brockstein, J.J. Luke

Development of methodology: R. Bao, B.E. Brockstein, J.J. Luke

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): S. Shah, J.E. Ward, B.E. Brockstein, J.J. Luke

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): S. Shah, R. Bao, J.J. Luke

Writing, review, and/or revision of the manuscript: S. Shah, J.E. Ward, R. Bao, C.R. Hall, B.E. Brockstein, J.J. Luke

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): J.E. Ward, B.E. Brockstein, J.J. Luke

Study supervision: B.E. Brockstein, J.J. Luke

Other (reviewed and interpreted the tissue from the referenced patient to arrive at a pathologic diagnosis): C.R. Hall

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

J.J. Luke was supported by Paul Calabresi Career Development in Clinical Oncology Award (NIH 1K12CA139160-05), Young Investigator Award from the Cancer Research Foundation, and the Arthur J Schreiner Family Melanoma Research Fund, with support from The Center for Research Informatics of The University of Chicago Biological Science Division and The Institute for Translational Medicine/CTSA (NIH UL1 RR024999).

1.
Hanna
NH
,
Einhorn
LH
. 
Testicular cancer–discoveries and updates
.
N Engl J Med
2014
;
371
:
2005
16
.
2.
International
Germ Cell Consensus Classification: A prognostic factor-based staging system for metastatic germ cell cancers. International Germ Cell Cancer Collaborative Group
.
J Clin Oncol
1997
;
15
:
594
603
.
3.
Edge
SB
,
American Joint Committee on Cancer
.
AJCC cancer staging manual
. 7th ed.
New York, NY
:
Springer
; 
2010
.
4.
Loehrer
PJ
 Sr
,
Lauer
R
,
Roth
BJ
,
Williams
SD
,
Kalasinski
LA
,
Einhorn
LH
. 
Salvage therapy in recurrent germ cell cancer: Ifosfamide and cisplatin 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 germ cell 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 testis cancer
.
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 untreated melanoma
.
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 microenvironment is driven by CD8(+) 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 CD8+ T-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) blockade with pembrolizumab (MK-3475) with an interferon-inflammatory immune 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
An immune-active tumor microenvironment favors clinical response to ipilimumab
.
Cancer Immunol Immunother
2012
;
61
:
1019
31
.
13.
Spranger
S
,
Bao
R
,
Gajewski
TF
. 
Melanoma-intrinsic beta-catenin signalling prevents anti-tumour immunity
.
Nature
2015
;
523
:
231
5
.
14.
Wolchok
JD
,
Hoos
A
,
O'Day
S
,
Weber
JS
,
Hamid
O
,
Lebbé
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.
Huang
da W
,
Sherman
BT
,
Lempicki
RA
. 
Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources
.
Nat Protoc
2009
;
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 Med
2015
;
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 resistance
.
Nature
2014
;
515
:
568
71
.
19.
Willemse
PM
,
Burggraaf
J
,
Hamdy
NA
,
Weijl
NI
,
Vossen
CY
,
van Wulften
L
, et al
Prevalence of the metabolic syndrome and cardiovascular disease risk in chemotherapy-treated testicular germ cell tumour survivors
.
Br J Cancer
2013
;
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
,
Svetlovska
D
, et al
Prognostic value of programmed-death-1 receptor (PD-1) and its ligand 1 (PD-L1) in testicular germ cell tumors
.
Ann Oncol
2016
;
27
:
300
5
.
22.
Ock
CY
,
Keam
B
,
Kim
S
,
Lee
JS
,
Kim
M
,
Kim
TM
, et al
Pan-cancer immunogenomic perspective on the tumor microenvironment based on PD-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 of the T-cell non-inflamed tumor microenvironment 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 blockade 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 chemotherapy in non-small cell lung cancer (NSCLC): A phase Ib safety and efficacy update [abstract]
.
In: Proceedings of the the 16th World Conference on Lung Cancer; September 6–9; Denver, CO. Abstract nr 0207. Available from:
http://library.iaslc.org/virtual-library-search?product_id=1&author=&category=&date=&session_type=&session=&presentation=&keyword=&page=4
accessed September 23rd, 2016
.