Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Jul 1.
Published in final edited form as: Curr Opin Urol. 2019 Jul;29(4):344–349. doi: 10.1097/MOU.0000000000000642

Genetics of Testicular Germ Cell Tumors

Nirmish Singla 1, John T Lafin 1, Rashed A Ghandour 1, Samuel Kaffenberger 2, James F Amatruda 3, Aditya Bagrodia 1
PMCID: PMC6659740  NIHMSID: NIHMS1533891  PMID: 31045925

Abstract

Purpose of review:

Understanding the molecular basis underlying testicular germ cell tumors (TGCTs) may help improve patient outcomes, particularly for patients with poorer risk or chemoresistant disease. Here, we review the major contemporary advances in elucidating TGCT genetics by discussing patterns of TGCT inheritance, recent genomic and transcriptomic discoveries in TGCT, and the role of genetics in predicting therapeutic resistance and in guiding treatment.

Recent findings:

In the absence of a major high-penetrance TGCT susceptibility gene, inheritance is likely driven by a complex polygenic model with considerable variation. The most common genomic alterations found in TGCTs include gains in chromosome 12p and mutations in KIT, KRAS, and NRAS, particularly in seminomas. Sensitivity to cisplatin-based chemotherapy likely relies on intact TP53, reciprocal loss of heterozygosity, and high mitochondrial priming. Targetable mutations are uncommon in TGCTs, however, posing a challenge for the development of effective personalized therapies. Consistent with the characteristically low tumor mutational burden, immune checkpoint inhibitors do not appear to be effective for most TGCTs.

Summary:

Refinements in next-generation sequencing techniques over the last few years have enabled considerable advances in elucidating the genomic, transcriptomic, and epigenetic landscape of TGCTs. Future efforts focused on developing novel treatment modalities are needed.

Keywords: testis cancer, germ cell tumor, genetics, inheritance, chemoresistance

INTRODUCTION

Testicular germ cell tumors (TGCTs) represent the most common malignancy in young men, with a projected incidence of 9,560 new diagnoses in the U.S. in 2019.(1) TGCTs exhibit a heterogeneous clinical and pathologic spectrum, broadly classified into seminomatous and non-seminomatous (NSGCT) groups. NSGCT is further characterized by various histologic subtypes, which may be pure or mixed. Understanding pathologic differences between TGCT subtypes is an important distinction given the implications both for prognosis and in guiding treatment strategies. Depending on stage and histology, patients may be effectively managed with surveillance, surgery, chemotherapy, radiation, or some combination thereof. As a whole, TGCT patients tend to do well clinically, even in the advanced setting, as evidenced by >80% 5-year overall survival and fewer than 500 deaths from TGCT projected for 2019 in the U.S.(1, 2)

A better understanding of the molecular and genetic basis underlying these heterogeneous tumors may help further improve upon patient outcomes, particularly for those with poorer risk or chemoresistant disease. It is this very heterogeneity, however, that makes it particularly difficult to study genetic correlates in TGCTs. Nevertheless, with the refinement of next-generation sequencing (NGS) techniques, the last few years have witnessed considerable advances in elucidating the genomic, transcriptomic, and epigenetic landscape of TGCTs. Herein, we review the major contemporary advances and future directions in understanding TGCT genetics by discussing patterns of TGCT inheritance, recent efforts to elucidate the genomic landscape of TGCT, and the role of genetics in predicting therapeutic resistance and in guiding treatment. Rather than provide a comprehensive overview of the genetics of TGCT, the present review will focus primarily on novel developments that emerged over the last couple years.

COMPLEX GENETIC PATTERNS UNDERLYING TGCT INHERITANCE

A thorough understanding of the genomic and environmental factors contributing to the rising incidence of TGCTs may shed light on the reasons for this trend and potentially identify approaches to counteract it.(3) Identified risk factors for developing TGCT include cryptorchidism, prior GCT, subfertility, disorders of sexual differentiation, and family history.(3-8) Among these, family history represents the strongest risk factor and may increase risk by up to 10-fold.(8-11)

Causative germline mutations for TGCTs remain largely undetected, though several single nucleotide polymorphisms (SNPs) have been associated with familial TGCT risk. Proposed candidate genes have included those linked to germ cell differentiation pathways (including those affecting the androgen receptor(12, 13)), cilia-microtubule function (e.g., DNAAF1 inactivation(14)), and the receptor tyrosine kinase KIT/KITLG signaling pathway (e.g., rare deleterious variants in the phosphodiesterase PDE11A(15)). In an earlier genome-wide association study (GWAS) for TGCT, Rapley et al. found strong evidence for susceptibility loci on chromosomes 5, 6, and 12 accounting for 7% of the risk to siblings and 10% of the risk to offspring of TGCT patients.(16) They also noted that KIT/KITLG involvement, largely found on chromosome 12, may account for the strongest association. In a subsequent multistage GWAS of over 25,000 individuals, Litchfield et al. identified 4 additional susceptibility loci for TGCT including 3q23, 11q14.1, 16p13.13, and 16q24.2, supporting a polygenic model of TGCT.(17) It is important to note, however, that the genetic defects identified are rare and together account for only a minority of TGCT cases.

The role and interaction of genetic and non-genetic factors in driving familial TGCT were explored more extensively in two recent studies.(**18, **19) In one study, Litchfield et al. analyzed germline whole-exome (WES) data for 919 TGCT patients (306 familial cases) and 1,609 healthy controls, encompassing nearly one million rare gene variants that included 114 cancer susceptibility genes and 49 TCGT susceptibility loci from prior GWAS.(**19) They were unable to find a major high-penetrance TGCT susceptibility gene, again supporting a polygenic model of inherited susceptibility with considerable variation.

In another larger-scale study Loveday et al. performed polygenic risk score analysis of 37 TGCT susceptibility SNPs in 3,931 sporadic and 236 familial TGCT cases and 12,368 controls.(**18) In contrast to their prior work,(**19) they found a clear enrichment for TGCT susceptibility alleles in familial compared to sporadic cases, and they noted that many SNPs would not have been previously detected as they mapped to noncoding regions of DNA. The majority of familial TGCT cases were attributable to polygenic enrichment, suggesting that in the absence of a major high-penetrance TGCT susceptibility gene, familial clustering is likely driven by aggregate effects of polygenic variation.

Together, these studies highlight the complex nature of the genetic basis underlying inheritance patterns in TGCT. Although there is presently no defined role for genetic testing of family members of affected individuals, clinicians must maintain a high degree of vigilance, as these members are at an increased baseline risk of developing TGCT.

RECENT EFFORTS TO ELUCIDATE THE GENETIC LANDSCAPE OF TGCT

Chromosome 12p gains constitute the most common copy number alteration seen in TGCTs.(20-22) Single-gene point mutations are generally uncommon, though mutations in KIT, TP53 (for mediastinal primary tumors), KRAS/BRAF, and NRAS have been found to be recurrently mutated.(23) The KIT proto-oncogene in particular is the most commonly mutated gene in TGCTs, though such mutations are seen in only 19% of seminomas and 2% of NSGCTs.(24-27) Notably, different frequencies of chromosome 12q gains and KITLG variants between Caucasians and African-Americans may account for some of the ethnic variation seen in TGCTs.(16, 28-30)

In multiple NGS studies, although the common genomic alterations identified in TGCTs were recurrently identified, including 12p gains and KRAS and KIT mutations, targetable alterations were not as well-defined.(20, 22, **31-34) For example, in a recent comprehensive study of the mutational profile of 42 TGCT patients, Litchfield et al. noted a relatively low total mutational burden (TMB) compared to other malignancies and found common copy number alterations to include 12p gains and amplifications of the spermatocyte development gene FSIP2 and Xq28, along with recurrent mutations in KIT and the tumor suppressor gene CDC27.(22) Likewise, in another clinically integrated molecular analysis of tumors from 47 patients with TGCTs and 2 additional patients with primary mediastinal GCTs, Taylor-Weiner et al. used WES and RNA sequencing to identify highly recurrent chromosome arm-level amplifications and reciprocal deletions (reciprocal loss of heterozygosity (LOH)) as the primary somatic feature of GCTs.(**31) They also noted that KRAS and RPL5 were significantly mutated, with KRAS mutations likely occurring phylogenetically after chromosome 12p gains.

In 2018, The Cancer Genome Atlas (TCGA) Research Network published their comprehensive analysis of 137 primary TGCTs using genomic, epigenomic, transcriptomic, and proteomic analysis.(**35) As previously identified, Shen et al. similarly noted that recurrent somatic mutations were rare with low overall TMB, and activating mutations were found in just 3 genes: KIT (18%), KRAS (14%), and NRAS (4%). These gene mutations were found primarily in seminoma patients, and enhanced KIT-PI3K-RAS signaling appeared to play a significant role in most seminomas. The authors also found that the seminoma, embryonal carcinoma, yolk sac tumor, and teratoma histologic subtypes harbored distinct DNA methylation, miRNA expression, immune infiltration, and copy number aberration profiles. Furthermore, they reported potential biomarkers for risk stratification, including microRNA (miRNA) expressed in teratoma, and methylation at non-canonical cytosine sites (CpH sites), observed in the setting of embryonal carcinoma. Together, their findings may provide biomarkers for studying TGCTs, risk-stratifying patients, and identifying potential therapeutic targets.

MICRORNAS: EMERGING BIOMARKERS FOR TGCT

The role of miRNA as an emerging serum biomarker for TGCT has been explored in several contemporary studies. miRNAs are small noncoding regions of RNA that are involved with epigenetic regulation of gene expression.(36) Several groups have identified a group of miRNAs that are secreted into circulation by testicular cancer tissue, specifically the miRNA-371–3 and miRNA-302/367 clusters.(37) These miRNA clusters, particularly miRNA-371a-3p, have been shown to exhibit higher sensitivity and specificity for GCT compared to traditional serum tumor markers in GCT (alpha-fetoprotein, beta-human chorionic gonadotropin, lactate dehydrogenase), and over 85% of patients with seminoma notably express miRNA-371a-3p.(38-41) The levels of these markers seem to be associated with both clinical stage and tumor bulk,(42, 43) supporting their role as potentially useful biomarkers in monitoring therapeutic response.

Recently, Dieckmann et al. conducted the largest prospective study to examine the performance characteristics of serum miRNA-371a-3p in the diagnosis and monitoring of patients with testicular cancer.(**44) Across an entire cohort of 616 patients with testis cancer, they noted that the miRNA-371a-3p test had 90% sensitivity, 94% specificity, 97% positive predictive value, 83% negative predictive value, and an area under the curve of 0.966 on receiver operating characteristic analysis for the primary diagnosis of GCT. The test outperformed the combination of classic serum tumor markers and was additionally associated with clinical stage, primary tumor size, and response to treatment. Notably, the marker was expressed in all histologic subtypes except teratoma. Their findings provide encouraging data to further support the clinical implementation of an miRNA-based test for patients with TGCT pending further validation.

THERAPEUTIC IMPLICATIONS OF TGCT GENETICS AND FUTURE DIRECTIONS

Platinum-based chemotherapy constitutes the first-line systemic regimen for advanced TGCT, and while most patients are cured with this approach, 10–20% of patients with metastatic TGCT will ultimately succumb to their disease.(3) Unfortunately, attempts to develop noncytotoxic targeted therapies for TCGT have been largely unsuccessful.(45) Indeed, there is a strong need to better understand molecular determinants of chemoresistance and identify actionable targets that may lead to the development of effective therapies in this setting.

In their integrated molecular analysis of TGCT, Taylor-Weiner et al. used BH3 profiling to functionally measure apoptotic signaling in tumors and suggested that chemosensitivity in TGCTs may rely on intact TP53, reciprocal LOH, and high mitochondrial priming.(**31) In contrast, chemoresistant tumors appeared to accumulate copy number events, including additional reciprocal LOH, and lose expression of pluripotency markers and apoptosis regulators (NANOG and POU5F1).(46, 47)

To explore genetic determinants of cisplatin resistance in GCTs further, Bagrodia et al. performed WES or targeted exon-capture-based sequencing on 180 tumors and, in line with the findings from Taylor-Weiner et al.,(**31) reported TP53 alterations, along with MDM2 amplifications, to be present exclusively in cisplatin-resistant tumors and prevalent in patients with primary mediastinal NSGCTs.(*48) Furthermore, actionable alterations, including RAC1 mutations, were found in over half of cisplatin-resistant GCTs, which may hold therapeutic implications for patients in the cisplatin-resistant space.

More recently, Necchi et al. performed NGS on 107 TGCTs (23 seminoma, 84 NSGCT) in patients who relapsed despite receipt of cisplatin-based chemotherapy, with most samples derived from chemorefractory metastatic lesions.(**49) RAS-RAF pathway (mostly KRAS alterations at the single-gene level) and cell-cycle pathway (largely non-targetable) alterations were the most common genomic alterations found in both seminomas and NSGCTs, while KIT and PI3K pathway gene alterations were more frequently seen in seminomas (22% and 26% of seminomas, respectively). Aside from KIT and PI3K pathway genes, only very few potentially targetable mutations were found, including BRAF (<5% of NSGCTs), ERBB2 (<4% of NSGCTs), and DNA repair genes (13% of TGCTs).

Immune checkpoint inhibitors (ICI) targeting programmed death-1 (PD-1), its ligand (PD-L1), or cytotoxic T-lymphocyte associated protein 4 (CTLA-4) have emerged as a promising class of systemic therapy in the frontline or chemorefractory setting for multiple malignancies, including lung cancer, bladder cancer, melanoma, and several others. The role of ICIs has been studied to a limited extent in TGCTs as well, but unfortunately results have not been encouraging.(50-52) This is not surprising in light of the low TMB and low degree of microsatellite instability found in TGCTs.(**49) Hence, without additional preclinical rationale and improved patient selection, the use of ICIs for TGCT will unlikely gain the same prominence as in other cancers.

Alternative targets in TGCT are under active investigation. In a small study of 7 patients, 5 with TGCTs, treatment with the antibody-drug conjugate brentuximab vedotin targeting CD30 resulted in a durable complete response (>6 months) in one patient and a brief partial response in another patient.(*53) The uncommon nature of targetable mutations in TGCTs remain a challenge in developing therapies for chemoresistant patients, and future efforts focusing on preclinical models and novel treatment modalities are much needed.

CONCLUSION

Considerable advances have been made over the last couple years to elucidate the complex genetic basis underlying TGCTs. Without an identifiable major high-penetrance TGCT susceptibility gene, familial clustering is likely driven by polygenic variation. Most commonly, gains in chromosome 12p and mutations in KIT and KRAS can be found in TGCTs, particularly among seminoma patients, though the uncommon nature of targetable mutations in TGCTs remains a challenge in developing therapies for patients resistant to cisplatin-based systemic chemotherapy. Nonetheless, chemosensitivity appears to rely on intact TP53, reciprocal LOH, and high mitochondrial priming. Consistent with the low TMB seen in TGCTs compared to other malignancies, ICI has not proven to be an effective management strategy for these patients. Serum miRNA is emerging as a promising biomarker for both diagnosis and therapeutic monitoring in patients with TGCT, and additional validation concerning its clinical utility is warranted. Indeed, future efforts focusing on developing novel, personalized treatment modalities are much needed.

KEY POINTS:

  • Refinements in next-generation sequencing techniques over the last few years have enabled considerable advances in elucidating the genomic, transcriptomic, and epigenetic landscape of TGCTs.

  • A major high-penetrance TGCT susceptibility gene has not been identified. Multiple studies support a complex polygenic model of inherited susceptibility to TGCTs with considerable variation.

  • The most common alterations in TGCTs, particularly seminomas, include gains in chromosome 12p and single-gene point mutations in KIT, KRAS, and NRAS. It appears that enhanced KIT-PI3K-RAS signaling may play a significant role in most seminomas.

  • Sensitivity to cisplatin-based chemotherapy appears to rely on intact TP53, reciprocal loss of heterozygosity, and high mitochondrial priming.

  • Serum microRNAs (miRNA), particularly miRNA371–3, are an emerging candidate biomarker for both the diagnosis and therapeutic monitoring of patients with TGCT, and contemporary studies have demonstrated early promise.

  • Consistent with the relatively low tumor mutational burden and microsatellite instability, immune checkpoint inhibitors do not appear to be effective for most TGCTs. The uncommon nature of targetable mutations in TGCTs remain a challenge in developing therapies for chemoresistant patients, and future efforts focused on developing novel treatment modalities are needed.

ACKNOWLEDGEMENTS

Financial support and sponsorship: This work was supported by the Ruth L. Kirschstein National Research Service Award T32 CA136515–09 (N.S.), the University of Texas Southwestern Medical Center Physician Scientist Training Program (N.S.), and Dedman Family Scholarship in Clinical Care (A.B).

Footnotes

Conflicts of interest: none

REFERENCES

  • 1.Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA Cancer J Clin 2019. January;69(1):7–34. [DOI] [PubMed] [Google Scholar]
  • 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. February;15(2):594–603. [DOI] [PubMed] [Google Scholar]
  • 3.Rajpert-De Meyts E, McGlynn KA, Okamoto K, et al. Testicular germ cell tumours. Lancet 2016. April 23;387(10029):1762–74. [DOI] [PubMed] [Google Scholar]
  • 4.Cook MB, Akre O, Forman D, et al. A systematic review and meta-analysis of perinatal variables in relation to the risk of testicular cancer--experiences of the mother. Int J Epidemiol 2009. December;38(6):1532–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Fossa SD, Chen J, Schonfeld SJ, et al. Risk of contralateral testicular cancer: a population-based study of 29,515 U.S. men. J Natl Cancer Inst 2005. July 20;97(14):1056–66. [DOI] [PubMed] [Google Scholar]
  • 6.Moller H, Skakkebaek NE. Risk of testicular cancer in subfertile men: case-control study. BMJ 1999. February 27;318(7183):559–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Hemminki K, Li X. Familial risk in testicular cancer as a clue to a heritable and environmental aetiology. Br J Cancer 2004. May 4;90(9):1765–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Swerdlow AJ, De Stavola BL, Swanwick MA, et al. Risks of breast and testicular cancers in young adult twins in England and Wales: evidence on prenatal and genetic aetiology. Lancet 1997. December 13;350(9093):1723–8. [DOI] [PubMed] [Google Scholar]
  • 9.Forman D, Oliver RT, Brett AR, et al. Familial testicular cancer: a report of the UK family register, estimation of risk and an HLA class 1 sib-pair analysis. Br J Cancer 1992. February;65(2):255–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Kharazmi E, Hemminki K, Pukkala E, et al. Cancer Risk in Relatives of Testicular Cancer Patients by Histology Type and Age at Diagnosis: A Joint Study from Five Nordic Countries. Eur Urol 2015. August;68(2):283–9. [DOI] [PubMed] [Google Scholar]
  • 11.Lutke Holzik MF, Rapley EA, Hoekstra HJ, et al. Genetic predisposition to testicular germ-cell tumours. Lancet Oncol 2004. June;5(6):363–71. [DOI] [PubMed] [Google Scholar]
  • 12.Pleskacova J, Hersmus R, Oosterhuis JW, et al. Tumor risk in disorders of sex development. Sex Dev 2010. September;4(4–5):259–69. [DOI] [PubMed] [Google Scholar]
  • 13.Lottrup G, Jorgensen A, Nielsen JE, et al. Identification of a novel androgen receptor mutation in a family with multiple components compatible with the testicular dysgenesis syndrome. J Clin Endocrinol Metab 2013. June;98(6):2223–9. [DOI] [PubMed] [Google Scholar]
  • 14.Litchfield K, Levy M, Dudakia D, et al. Rare disruptive mutations in ciliary function genes contribute to testicular cancer susceptibility. Nat Commun 2016. December 20;7:13840. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Pathak A, Stewart DR, Faucz FR, et al. Rare inactivating PDE11A variants associated with testicular germ cell tumors. Endocr Relat Cancer 2015. December;22(6):909–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Rapley EA, Turnbull C, Al Olama AA, et al. A genome-wide association study of testicular germ cell tumor. Nat Genet 2009. July;41(7):807–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Litchfield K, Holroyd A, Lloyd A, et al. Identification of four new susceptibility loci for testicular germ cell tumour. Nat Commun 2015. October 27;6:8690. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • **18.Loveday C, Law P, Litchfield K, et al. Large-scale Analysis Demonstrates Familial Testicular Cancer to have Polygenic Aetiology. Eur Urol 2018. September;74(3):248–52.**In this study of of 919 TGCT patients, including 306 familial cases, and 1,609 healthy controls, the authors performed germline whole-exome sequencing to help define the role and interaction of genetic and non-genetic factors in driving familial TGCT. The authors were not able to find a major high-penetrance susceptibility gene for TGCT, supporting a polygenic model fo inherited susceptibility with considerable variation.
  • **19.Litchfield K, Loveday C, Levy M, et al. Large-scale Sequencing of Testicular Germ Cell Tumour (TGCT) Cases Excludes Major TGCT Predisposition Gene. Eur Urol 2018. June;73(6):828–31.**In this large-scale study of 3,931 sporadic and 236 familial TCGT cases and 12,368 controls using polygenic risk score analysis, the authors note that familial clustering in TCGT is likely driven by aggregate effects of polygenic variation.
  • 20.Atkin NB, Baker MC. Specific chromosome change, i(12p), in testicular tumours? Lancet 1982. December 11;2(8311):1349. [DOI] [PubMed] [Google Scholar]
  • 21.Oosterhuis JW, Castedo SM, de Jong B, et al. Ploidy of primary germ cell tumors of the testis. Pathogenetic and clinical relevance. Lab Invest 1989. January;60(1):14–21. [PubMed] [Google Scholar]
  • 22.Litchfield K, Summersgill B, Yost S, et al. Whole-exome sequencing reveals the mutational spectrum of testicular germ cell tumours. Nat Commun 2015. January 22;6:5973. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Gonzalez-Exposito R, Merino M, Aguayo C. Molecular biology of testicular germ cell tumors. Clin Transl Oncol 2016. June;18(6):550–6. [DOI] [PubMed] [Google Scholar]
  • 24.Sheikine Y, Genega E, Melamed J, et al. Molecular genetics of testicular germ cell tumors. Am J Cancer Res 2012;2(2):153–67. [PMC free article] [PubMed] [Google Scholar]
  • 25.Ezeh UI, Turek PJ, Reijo RA, et al. Human embryonic stem cell genes OCT4, NANOG, STELLAR, and GDF3 are expressed in both seminoma and breast carcinoma. Cancer 2005. November 15;104(10):2255–65. [DOI] [PubMed] [Google Scholar]
  • 26.Bamford S, Dawson E, Forbes S, et al. The COSMIC (Catalogue of Somatic Mutations in Cancer) database and website. Br J Cancer 2004. July 19;91(2):355–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Forbes SA, Beare D, Gunasekaran P, et al. COSMIC: exploring the world’s knowledge of somatic mutations in human cancer. Nucleic Acids Res 2015. January;43(Database issue):D805–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Kanetsky PA, Mitra N, Vardhanabhuti S, et al. Common variation in KITLG and at 5q31.3 predisposes to testicular germ cell cancer. Nat Genet 2009. July;41(7):811–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.McGlynn KA, Devesa SS, Graubard BI, et al. Increasing incidence of testicular germ cell tumors among black men in the United States. J Clin Oncol 2005. August 20;23(24):5757–61. [DOI] [PubMed] [Google Scholar]
  • 30.Gajendran VK, Nguyen M, Ellison LM. Testicular cancer patterns in African-American men. Urology 2005. September;66(3):602–5. [DOI] [PubMed] [Google Scholar]
  • **31.Taylor-Weiner A, Zack T, O’Donnell E, et al. Genomic evolution and chemoresistance in germ-cell tumours. Nature 2016. November 30;540(7631):114–18.**This study is one of the initial well-executed clinically integrated molecular analyses of GCT patients. Using whole-exome and transcriptomic sequencing, the authors were able to identify recurrent chromosome-arm level amplifications and reciprocal deletions as the primary somatic feature of GCTs. They also purported mechanisms of chemosensitivity using BH3 profiling.
  • 32.Goddard NC, McIntyre A, Summersgill B, et al. KIT and RAS signalling pathways in testicular germ cell tumours: new data and a review of the literature. Int J Androl 2007. August;30(4):337–48; discussion 49. [DOI] [PubMed] [Google Scholar]
  • 33.Tian Q, Frierson HF Jr., Krystal GW, et al. Activating c-kit gene mutations in human germ cell tumors. Am J Pathol 1999. June;154(6):1643–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Biermann K, Goke F, Nettersheim D, et al. c-KIT is frequently mutated in bilateral germ cell tumours and down-regulated during progression from intratubular germ cell neoplasia to seminoma. J Pathol 2007. November;213(3):311–8. [DOI] [PubMed] [Google Scholar]
  • **35.Shen H, Shih J, Hollern DP, et al. Integrated Molecular Characterization of Testicular Germ Cell Tumors. Cell Rep 2018. June 12;23(11):3392–406.**This study recently emerged from The Cancer Genome Atlas (TCGA) Research Network. The investigators performed a comprehensive molecular analysis of 137 primary TGCT samples using genomic, epigenomic, transcriptomic, and proteomic approaches. They noted that total mutational burden was relatively low in TGCT, and enhanced KIT-PI3K-RAS signaling seems to play an important role in seminomas. Different histologic subtypes also vary in their molecular profiles, which may have implications for risk stratification and treatment strategies.
  • 36.Li Z, Xu R, Li N. MicroRNAs from plants to animals, do they define a new messenger for communication? Nutr Metab (Lond) 2018;15:68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Murray MJ, Halsall DJ, Hook CE, et al. Identification of microRNAs From the miR-371~373 and miR-302 clusters as potential serum biomarkers of malignant germ cell tumors. Am J Clin Pathol 2011. January;135(1):119–25. [DOI] [PubMed] [Google Scholar]
  • 38.Belge G, Dieckmann KP, Spiekermann M, et al. Serum levels of microRNAs miR-371–3: a novel class of serum biomarkers for testicular germ cell tumors? Eur Urol 2012. May;61(5):1068–9. [DOI] [PubMed] [Google Scholar]
  • 39.Syring I, Bartels J, Holdenrieder S, et al. Circulating serum miRNA (miR-367–3p, miR-371a-3p, miR-372–3p and miR-373–3p) as biomarkers in patients with testicular germ cell cancer. J Urol 2015. January;193(1):331–7. [DOI] [PubMed] [Google Scholar]
  • 40.van Agthoven T, Looijenga LHJ. Accurate primary germ cell cancer diagnosis using serum based microRNA detection (ampTSmiR test). Oncotarget 2017. August 29;8(35):58037–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Bezan A, Gerger A, Pichler M. MicroRNAs in testicular cancer: implications for pathogenesis, diagnosis, prognosis and therapy. Anticancer Res 2014. June;34(6):2709–13. [PubMed] [Google Scholar]
  • 42.van Agthoven T, Eijkenboom WMH, Looijenga LHJ. microRNA-371a-3p as informative biomarker for the follow-up of testicular germ cell cancer patients. Cell Oncol (Dordr) 2017. August;40(4):379–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Radtke A, Hennig F, Ikogho R, et al. The Novel Biomarker of Germ Cell Tumours, Micro-RNA-371a-3p, Has a Very Rapid Decay in Patients with Clinical Stage 1. Urol Int 2018;100(4):470–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • **44.Dieckmann KP, Radtke A, Geczi L, et al. Serum Levels of MicroRNA-371a-3p (M371 Test) as a New Biomarker of Testicular Germ Cell Tumors: Results of a Prospective Multicentric Study. J Clin Oncol 2019. March 15:JCO1801480.**This is the largest prospective study to examine the performance characteristics of miRNA-371a-3p in the diagnosis and therapeutic monitoring of patients with TGCT. The authors found the test to outperform traditional serum tumor markers. They noted that miRNA levels associated with clinical stage, tumor size, and response to treatment, supporting the clinical role of an miRNA-based test for TGCT patients.
  • 45.Allen JC, Kirschner A, Scarpato KR, et al. Current Management of Refractory Germ Cell Tumors and Future Directions. Curr Oncol Rep 2017. February;19(2):8. [DOI] [PubMed] [Google Scholar]
  • 46.Looijenga LH, Stoop H, de Leeuw HP, et al. POU5F1 (OCT¾) identifies cells with pluripotent potential in human germ cell tumors. Cancer Res 2003. May 1;63(9):2244–50. [PubMed] [Google Scholar]
  • 47.Hart AH, Hartley L, Parker K, et al. The pluripotency homeobox gene NANOG is expressed in human germ cell tumors. Cancer 2005. November 15;104(10):2092–8. [DOI] [PubMed] [Google Scholar]
  • *48.Bagrodia A, Lee BH, Lee W, et al. Genetic Determinants of Cisplatin Resistance in Patients With Advanced Germ Cell Tumors. J Clin Oncol 2016. November 20;34(33):4000–07.*This novel study explored genetic determinants of cisplatin resistance in GCTs, finding that TP53 alterations, along with MDM2 amplifications, were present exclusively in cisplatin-resistant tumors and prevalent in patients with primary mediastinal NSGCTs. Actionable alterations were found in several chemoresistant paitents.
  • **49.Necchi A, Bratslavsky G, Corona RJ, et al. Genomic Characterization of Testicular Germ Cell Tumors Relapsing After Chemotherapy. Eur Urol Focus 2018. July 16.**In this more recent study of chemorefractory patients, the authors noted that ras-RAF pathway and cell cycle pathway alterations were the most common genomic alterations found in TGCTs, while KIT and PI3K pathway gene alterations were more frequently seen in seminomas. Only very few potentially targetable mutations were found.
  • 50.Adra N, Einhorn LH, Althouse SK, et al. Phase II trial of pembrolizumab in patients with platinum refractory germ-cell tumors: a Hoosier Cancer Research Network Study GU14–206. Ann Oncol 2018. January 1;29(1):209–14. [DOI] [PubMed] [Google Scholar]
  • 51.Necchi A, Giannatempo P, Raggi D, et al. An Open-label Randomized Phase 2 study of Durvalumab Alone or in Combination with Tremelimumab in Patients with Advanced Germ Cell Tumors (APACHE): Results from the First Planned Interim Analysis. Eur Urol 2019. January;75(1):201–03. [DOI] [PubMed] [Google Scholar]
  • 52.Fankhauser CD, Curioni-Fontecedro A, Allmann V, et al. Frequent PD-L1 expression in testicular germ cell tumors. Br J Cancer 2015. July 28;113(3):411–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • *53.Albany C, Einhorn L, Garbo L, et al. Treatment of CD30-Expressing Germ Cell Tumors and Sex Cord Stromal Tumors with Brentuximab Vedotin: Identification and Report of Seven Cases. Oncologist 2018. March;23(3):316–23.*This is a novel, small study evaluating the effectiveness of an antibody-drug conjugate (brentuximab vedotin) targeting CD30, showing a durable complete resopnse in one patient and a brief partial response in another patient with TGCT

RESOURCES