Molecular biology of testicular germ cell tumors: Unique features awaiting clinical application

https://doi.org/10.1016/j.critrevonc.2013.10.001Get rights and content

Abstract

Testicular germ cell tumors (TGCTs) are the most common solid tumors in young adult men characterized by distinct biologic features and clinical behavior. Both genetic predispositions and environmental factors probably play a substantial role in their etiology. TGTCs arise from a malignant transformation of primordial germ cells in a process that starts prenatally, is often associated with a certain degree of gonadal dysgenesis, and involves the acquirement of several specific aberrations, including activation of SCF–CKIT, amplification of 12p with up-regulation of stem cell genes, and subsequent genetic and epigenetic alterations. Their embryonic and germ origin determines the unique sensitivity of TGCTs to platinum-based chemotherapy. Contrary to the vast majority of other malignancies, no molecular prognostic/predictive factors nor targeted therapy is available for patients with these tumors. This review summarizes the principal molecular characteristics of TGCTs that could represent a potential basis for development of novel diagnostic and treatment approaches.

Introduction

Though relatively rare, testicular (and extragonadal) germ cell tumors (TGCTs) are the most common solid tumors in young men aged 18–35 years, and represent the leading cause of cancer-related morbidity and mortality in this group. Their incidence is steadily increasing worldwide, ranging around 5–10/100,000 in developed countries and displaying profound ethnic and geographic differences (the highest incidence in Europe estimated for 2012 was in Denmark and Norway – over 12/100,000, in contrast to 5/100,000 in neighboring Finland or 3/100,000 in Spain). Their mortality has been slightly decreasing to 0.5/100,000 in developed countries, largely attributable to the platinum-based chemotherapy and multidisciplinary approach in their management [1], [2], [3], [4], [5], [6]. The principal risk factors for TGCTs include the family history of testicular tumor (relative risk (RR)  4–6 for sons and 8–10 for brothers), cryptorchism (RR  4–8), and contralateral testicular tumor diagnosed previously (RR  25). The increasing incidence of TGCTs seems to correspond with decreasing fertility among men, male sub/infertility being also related to TCGTs (RR up to 20 for infertile men) [2], [4], [7], [8].

Both genetic predispositions and environmental factors are thought to play a significant role in the etiopathogenesis of these tumors. Several gene loci with low penetrance probably contribute to the TGCT susceptibility (e.g. proposed gene TGCT1 on Xq27, also linked to cryptorchism; gr/gr deletion in AZFc region on Y chromosome; etc.) [6], [8], [9]. In recent genome-wide association studies, six gene loci were related to TGCT predisposition: KITLG, SPRY4, BAK1, DMRT1, TERT and ATF7IP [10], [11], [12], [13], [14], [15]; and the International Testicular Cancer Linkage Consortium (ITCLC) continues to recruit participants for studies on genetics of familial TGCTs. Regarding environmental factors, increased exposure to estrogens (including xenoestrogens, endocrine disrupting chemicals – EDC) in prenatal and early postnatal period is hypothesized to contribute to the rising occurrence of TGCTs and sub/infertility. Among other environmental toxicants, phtalates and nanoparticles have been shown to affect both somatic and developing germ cells but their contribution to testicular problems such as infertility or tumors is unclear [16], [17].

The traditional histologic classification of TGCTs is based on the type of their differentiation dividing them into two major groups: seminomas (typical, undifferentiated, spermatocytic) and non-seminomas (embryonal carcinoma, teratoma and teratocarcinoma, yolk sac tumor, choriocarcioma, mixed tumors). Their precursor lesion of carcinoma in situ (CIS) character is the intratubular germ cell neoplasm, unclassified (ITGCNU). TGCT cells express several characteristic antigens (SOX2 and 17, NANOG, OCT3/4, HMGA1 and 2, PATZ1, CKIT, Aurora-B, PLAP, etc.) that may help to identify the TGCT subtypes and distinguish TGCTs from other tumors [2], [18]; they may also release proteins including human choriogonadotropin (HCG) and alpha-fetoprotein (AFP) useful as serum tumor markers for the disease monitoring [19], [20], [21].

Although in the clinical course TGCTs, particularly non-seminomas, tend to metastasize relatively early to lymph nodes (retroperitoneal, mediastinal, left supraclavicular) and other organs (lungs, liver, brain), due to their exceptional chemosensitivity to platinum derivates they are curable even in the stage of advanced metastatic disease – a characteristic which is unparalleled among any other solid tumors in adults. Seminomas differ from non-seminomas by their later onset (mostly during the fourth decade of life, about 10 years later than non-seminomas), usually slower growth and dissemination, and also very high radiosensitivity. Patients with TGCTs are stratified into risk groups according to the histology, TNM stage and serum markers (IGCCCG classification). Therapy of TGCTs is multimodal including surgery, platinum-based chemotherapy, and/or radiotherapy. The 5-year overall survival is estimated around 95% for good-prognosis, 80% for intermediate-prognosis, and 70% for poor-prognosis tumors. The overall cure rates of TGCTs approach 85%; over 95% for localized disease and approximately 80% for metastatic disease – the best response by any solid tumor [22], [23], [24], [25]. Despite that, TGCTs still remain a significant cause of death in adult young men and the management and outcomes of TGCT patients have not changed much since the introduction of cisplatin into the clinical practice in the late 1970s, newer approaches (high-dose chemotherapy) or cytostatics (taxanes, gemcitabine) bringing rather modest improvement to patients’ survival.

Section snippets

TGCT development

According to the cell of origin, genetic and other characteristics, human germ cell tumors (GCTs) have been recently divided by an alternative WHO classification system into five entities (Table 1) [26], [27]. TGCTs of young adults that are in the focus of this review belong to WHO type II GCTs. Pediatric TGCTs (type I) and spermatocytic seminomas (type III) are rare, have different etiopathogenesis, biologic and clinical features, and are not further discussed here. On the contrary,

Typical genetic aberrations

The genetic constitution of TGCTs reflects the embryonic character of the tumor cells, with generally low incidence of mutations, frequent uniparental disomies, loss of parental pattern of genomic imprinting (erased) and distinct DNA methylation profile (hypomethylation), in which they differ from other somatic tissue-derived tumors.

Genomic imprinting and uniparental disomy

Genomic imprinting is an inherited uniparental, monoallelic expression of certain genes due to the epigenetic silencing

DNA damage response (DDR), TP53 signaling and cell cycle control

Embryonic and germ cells, which the tumor cells originate from, employ specific mechanisms to ensure their genetic integrity remains pristine, as the accumulation of mutations would have detrimental effect for the whole organism or even the next generations. In the context of stem cell gene overexpression, modified activity of DNA repair systems and apoptotic programs has been observed in these cells. This results in the suppression of spontaneous mutation frequency to ≈10−6 compared to ≈10−4

Potential molecular prognostic/predictive factors and therapeutic targets

In an increasing number of various malignancies, molecular aberrations have become an integral part of tumor classification, diagnostic process and treatment strategy. They may characterize specific tumor subtypes, act as key prognostic factors, represent targets for development of new therapeutic approaches and predict the overall response to the treatment. Despite the presence of unique molecular features and recurrent aberrations, none of these has been implemented into TGCT clinical

Conclusion and take-home message

TGCTs are malignant tumors with distinct molecular characteristics and clinical behavior. In their etiology, both genetic predispositions and environmental factors play a substantial role – probably more than in other tumors, though no single cause has been identified so far. The initial steps of the process of malignant transformation occur prenatally and are associated with disturbed gonadal development, which can range from discrete changes to severe gonadal dysgenesis syndromes. The

Conflict of interest

All authors declare no conflict of interests.

Funding sources

Supported by grant IGA NT/12414-5.

Reviewers

Dr Friedemann Honecker, Medizinische Klinik II, Universitätsklinikum Hamburg-Eppendorf, Labor für Experimentelle Onkologie, Zentrum für Innere Medizin, D-20246 Hamburg, Germany.

Dr John Fitzpatrick, Head of Research, Irish Cancer Society, Ireland.

Dr. Ludmila Boublikova is a consultant Medical Oncologist at the Department of Oncology, 1st Faculty of Medicine, Charles University and Thomayer Hospital, and a Principal Investigator at research laboratories of the Department of Pediatric Hematology and Oncology, 2nd Faculty of Medicine, Charles University and University Hospital in Motol, Prague, Czech Republic. She obtained her MD degree in Medicine from Charles University and completed the postgraduate clinical training in Medical Oncology

References (176)

  • N.B. Atkin et al.

    Specific chromosome change, i(12p), in testicular tumours?

    Lancet

    (1982)
  • A.M. Ottesen et al.

    Cytogenetic investigation of testicular carcinoma in situ and early seminoma by high-resolution comparative genomic hybridization analysis of subpopulations flow sorted according to DNA content

    Cancer Genet Cytogenet

    (2004)
  • B. Koberle et al.

    Cisplatin resistance: preclinical findings and clinical implications

    Biochim Biophys Acta

    (2010)
  • F. Honecker et al.

    Xeroderma pigmentosum group a protein and chemotherapy resistance in human germ cell tumors

    Lab Invest

    (2003)
  • R.H. Jones et al.

    New directions in testicular cancer; molecular determinants of oncogenesis and treatment success

    Eur J Cancer

    (2003)
  • A.E. Fenske et al.

    Cisplatin resistance induced in germ cell tumour cells is due to reduced susceptibility towards cell death but not to altered DNA damage induction or repair

    Cancer Lett

    (2012)
  • P.M. Voorhoeve et al.

    A genetic screen implicates miRNA-372 and miRNA-373 as oncogenes in testicular germ cell tumors

    Cell

    (2006)
  • M.I. Aladjem et al.

    ES cells do not activate p53-dependent stress responses and undergo p53-independent apoptosis in response to DNA damage

    Curr Biol

    (1998)
  • IARC....
  • R.P. Manecksha et al.

    Epidemiology of testicular cancer

    BJU Int

    (2009)
  • NCI....
  • E. Rajpert-De Meyts

    Developmental model for the pathogenesis of testicular carcinoma in situ: genetic and environmental aspects

    Hum Reprod Update

    (2006)
  • N. Jorgensen et al.

    Recent adverse trends in semen quality and testis cancer incidence among Finnish men

    Int J Androl

    (2011)
  • M.F. Lutke Holzik et al.

    Genetic predisposition to testicular germ-cell tumours

    Lancet Oncol

    (2004)
  • E. Rapley

    Susceptibility alleles for testicular germ cell tumour: a review

    Int J Androl

    (2007)
  • G.P. Crockford et al.

    Genome-wide linkage screen for testicular germ cell tumour susceptibility loci

    Hum Mol Genet

    (2006)
  • D. Gilbert et al.

    Testicular germ cell tumours: predisposition genes and the male germ cell niche

    Nat Rev Cancer

    (2011)
  • P.A. Kanetsky et al.

    Common variation in KITLG and at 5q31 3 predisposes to testicular germ cell cancer

    Nat Genet

    (2009)
  • E.A. Rapley et al.

    A genome-wide association study of testicular germ cell tumor

    Nat Genet

    (2009)
  • C. Turnbull et al.

    Genome-wide association studies provide new insights into the genetic basis of testicular germ-cell tumour

    Int J Androl

    (2011)
  • C. Turnbull et al.

    Variants near DMRT1, TERT and ATF7IP are associated with testicular germ cell cancer

    Nat Genet

    (2010)
  • M. Garner et al.

    Testicular cancer and hormonally active agents

    J Toxicol Environ Health B Crit Rev

    (2008)
  • B. Lucas et al.

    Signaling pathways in spermatogonial stem cells and their disruption by toxicants

    Birth Defects Res C Embryo Today

    (2009)
  • G.J. van de Geijn et al.

    Recent developments in testicular germ cell tumor research

    Birth Defects Res C Embryo Today

    (2009)
  • E. Rajpert-de Meyts et al.

    From gonocytes to testicular cancer: the role of impaired gonadal development

    Ann N Y Acad Sci

    (2007)
  • Y. Sheikine et al.

    Molecular genetics of testicular germ cell tumors

    Am J Cancer Res

    (2012)
  • D.R. Feldman et al.

    Progression-free and overall survival in patients with relapsed/refractory germ cell tumors treated with single-agent chemotherapy: endpoints for clinical trial design

    Cancer

    (2011)
  • H.D. Mannuel et al.

    Update on testicular germ cell tumors

    Curr Opin Oncol

    (2011)
  • L.H. Looijenga et al.

    A molecular model for the development of germ cell cancer

    Int J Cancer

    (1999)
  • J.W. Oosterhuis et al.

    Testicular germ-cell tumours in a broader perspective

    Nat Rev Cancer

    (2005)
  • L.H. Looijenga et al.

    Dissecting the molecular pathways of (testicular) germ cell tumour pathogenesis; from initiation to treatment-resistance

    Int J Androl

    (2011)
  • L.H. Looijenga et al.

    Chromosomes and expression in human testicular germ-cell tumors: insight into their cell of origin and pathogenesis

    Ann N Y Acad Sci

    (2007)
  • E. Rajpert-De Meyts et al.

    Pathogenesis of testicular carcinoma in situ and germ cell cancer: still more questions than answers

    Int J Androl

    (2011)
  • K. Almstrup et al.

    Improved gene expression signature of testicular carcinoma in situ

    Int J Androl

    (2007)
  • J. Pleskacova et al.

    Tumor risk in disorders of sex development

    Sex Dev

    (2011)
  • K.M. Grigor et al.

    Pathogenesis and cell biology of germ cell neoplasia: general discussion

    Eur Urol

    (1993)
  • A. Frigyesi et al.

    A model for karyotypic evolution in testicular germ cell tumors

    Genes Chromosomes Cancer

    (2004)
  • R.S. Chaganti et al.

    The cytogenetic theory of the pathogenesis of human adult male germ cell tumors

    Apmis

    (1998)
  • R.S. Chaganti et al.

    Genetics and biology of adult human male germ cell tumors

    Cancer Res

    (2000)
  • N.E. Skakkebaek et al.

    Testicular dysgenesis syndrome: an increasingly common developmental disorder with environmental aspects

    Hum Reprod

    (2001)
  • Cited by (36)

    • The clinical value of circulating free tumor DNA in testicular germ cell tumor patients

      2022, Urologic Oncology: Seminars and Original Investigations
    • Perspectives on testicular germ cell neoplasms

      2017, Human Pathology
      Citation Excerpt :

      GCNIS has been recognized as a common precursor lesion for most malignant testicular GCTs since 1972 (Fig. 1). It is found in the adjacent seminiferous tubules of approximately 88% of adult testicular GCTs and in approximately 5% of cases in the contralateral testis [10-13]. However, GCNIS is absent in yolk sac tumor and teratoma of the prepubertal testis and in spermatocytic tumor of the adult testis.

    • Sperm mitochondria dysfunction in response to testicular cancer

      2024, European Journal of Clinical Investigation
    • Epigenetic Regulation of Driver Genes in Testicular Tumorigenesis

      2023, International Journal of Molecular Sciences
    View all citing articles on Scopus

    Dr. Ludmila Boublikova is a consultant Medical Oncologist at the Department of Oncology, 1st Faculty of Medicine, Charles University and Thomayer Hospital, and a Principal Investigator at research laboratories of the Department of Pediatric Hematology and Oncology, 2nd Faculty of Medicine, Charles University and University Hospital in Motol, Prague, Czech Republic. She obtained her MD degree in Medicine from Charles University and completed the postgraduate clinical training in Medical Oncology at Thomayer Hospital in Prague, Czech Republic. Then she finished her PhD study in Molecular biology at Trinity College Dublin, Ireland. She regularly publishes in peer-reviewed journals and acts as a reviewer for international journals. Her main areas of professional interest involve the research on malignant cell development and molecular prognostic factors, and clinical management of patients with testicular tumors.

    Dr. Tomas Buchler is an Associated Professor of Oncology and the Head of the Department of Oncology of 1st Faculty of Medicine, Charles University and Thomayer Hospital in Prague, Czech Republic. He trained in Internal Medicine and Medical Oncology at the University Hospital in Brno, Czech Republic, where he obtained his MD and PhD degrees. He spent several years as a researcher in cancer immunology at Oxford University, UK, and as a senior registrar in oncology at University College Hospital and St. Bartholomew's Hospital in London, UK. He is a full member of the European Society of Medical Oncology (ESMO), the author of more than 40 peer-reviewed articles and reviewer for medical journals and grant agencies. His professional interests include the epidemiology and treatment of testicular tumors, renal and colorectal cancer.

    Dr. Jan Trka is currently a Professor of Medical Genetics and the Head of the Laboratory Center of Department of Pediatric Hematology and Oncology of 2nd Faculty of Medicine, Charles University and University Hospital in Prague – Motol, Czech Republic. He obtained his MD degree in Medicine and PhD in Molecular biology from Charles University in Prague, and spent several years on PhD and postdoc positions in research institutes in Austria and the UK. He has founded and leads the Childhood Leukemia Investigation Prague (CLIP) group. He is an active member of the International Members Committee and the Committee on Communication of the American Society of Hematology (ASH) and the head of the Biology and Diagnosis Committee of the International BFM Study Group (I-BFM SG). He has published over 80 peer-reviewed articles, has received a number of grants and scientific awards and is a reviewer for international journals and grant agencies. He is devoted to the childhood acute leukemia research with special interests in leukemic clone origin and development and minimal residual disease detection.

    1

    On behalf of TTIP – Testicular Tumor Investigation Group Prague.

    View full text