1
|
Sadek KM, AbdEllatief HY, Mahmoud SFE, Alexiou A, Papadakis M, Al‐Hajeili M, Saad HM, Batiha GE. New insights on testicular cancer prevalence with novel diagnostic biomarkers and therapeutic approaches. Cancer Rep (Hoboken) 2024; 7:e2052. [PMID: 38507271 PMCID: PMC10953835 DOI: 10.1002/cnr2.2052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 02/23/2024] [Accepted: 03/05/2024] [Indexed: 03/22/2024] Open
Abstract
BACKGROUND Testicular cancer (TC), comprising merely 1% of male neoplasms, holds the distinction of being the most commonly encountered neoplasm among young males. RECENT FINDINGS Most cases of testicular neoplasms can be classified into two main groups, namely germ cell tumors representing approximately 95% of the cases, and sex cord-stromal tumors accounting for about 5% of the cases. Moreover, its prevalence is on the rise across the globe. TC is a neoplastic condition characterized by a favorable prognosis. The advent of cisplatin-based chemotherapeutic agents in the latter part of the 1970s has led to a significant enhancement in the 5-year survival rate, which presently surpasses 95%. Given that TC is commonly detected before reaching the age of 40, it can be anticipated that these individuals will enjoy an additional 40-50 years of life following successful treatment. The potential causes of TC are multifactorial and related to different pathologies. Accurate identification is imperative to guarantee the utmost efficacious and suitable therapy. To a certain degree, this can be accomplished through the utilization of blood examinations for neoplastic indicators; nonetheless, an unequivocal diagnosis necessitates an evaluation of the histological composition of a specimen via a pathologist. CONCLUSION TC is multifactorial and has various pathologies, therefore this review aimed to revise the prenatal and postnatal causes as well as novel diagnostic biomarkers and the therapeutic strategies of TC.
Collapse
Affiliation(s)
- Kadry M. Sadek
- Department of Biochemistry, Faculty of Veterinary MedicineDamanhour UniversityAbadiyyat DamanhurEgypt
| | - Hazem Y. AbdEllatief
- Department of Biochemistry, Faculty of Veterinary MedicineDamanhour UniversityAbadiyyat DamanhurEgypt
| | - Sahar F. E. Mahmoud
- Department of Histology, Faculty of Veterinary MedicineDamanhour UniversityAbadiyyat DamanhurEgypt
| | - Athanasios Alexiou
- University Centre for Research and DevelopmentChandigarh UniversityMohaliPunjabIndia
- Department of Research and Development, FunogenAthensGreece
- Department of Research and DevelopmentAFNP MedWienAustria
- Department of Science and EngineeringNovel Global Community Educational FoundationHebershamNew South WalesAustralia
| | - Marios Papadakis
- Department of Surgery IIUniversity Hospital Witten‐HerdeckeWuppertalGermany
| | - Marwan Al‐Hajeili
- Department of MedicineKing Abdulaziz UniversityJeddahKingdom of Saudi Arabia
| | - Hebatallah M. Saad
- Department of Pathology, Faculty of Veterinary MedicineMatrouh UniversityMarsa MatruhEgypt
| | - Gaber El‐Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary MedicineDamanhour UniversityDamanhourEgypt
| |
Collapse
|
2
|
Kim JH, Page RD, Moses GA, Columbres RC. Exceedingly Rare Bilateral Synchronous Germ Cell Testicular Tumors With Different Histopathological Features. Cureus 2023; 15:e42374. [PMID: 37492037 PMCID: PMC10364656 DOI: 10.7759/cureus.42374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/24/2023] [Indexed: 07/27/2023] Open
Abstract
Bilateral synchronous testicular tumors are a relatively uncommon occurrence, especially when they involve germ cell tumors of different histology. In this context, we present a compelling case report of a male patient who was diagnosed with bilateral synchronous germ cell testicular tumors, with one being a seminoma and the other a non-seminomatous germ cell tumor (NSGCT). The coexistence of two distinct histological types, seminoma and NSGCT, necessitates a comprehensive diagnostic approach to accurately identify and characterize each tumor. This underscores the importance of clinical history, physical examination, imaging techniques, and histopathological analysis to establish an appropriate diagnosis. Careful consideration must be given to factors such as tumor stage, histological subtype, and individual patient characteristics to determine the most suitable treatment strategy. Treatment options may encompass a combination of surgery, chemotherapy, and radiation therapy, tailored to each tumor's specific characteristics and the patient's overall health. By highlighting this unique case, we aim to underscore the significance of meticulous evaluation and accurate diagnosis when confronted with bilateral synchronous testicular tumors of different histology.
Collapse
Affiliation(s)
- Jong H Kim
- Internal Medicine, Medical City Weatherford, Weatherford, USA
| | - Ray D Page
- Oncology, Center for Cancer and Blood Disorders, Fort Worth, USA
| | - Gregory A Moses
- Pathology and Laboratory Medicine, Medical City Weatherford, Weatherford, USA
| | - Rod C Columbres
- Urology, William Carey University College of Osteopathic Medicine, Hattiesburg, USA
| |
Collapse
|
3
|
Anderson D, Razzak AN, McDonald M, Cao D, Hasoon J, Viswanath O, Kaye AD, Urits I. Mental Health in Urologic Oncology. Health Psychol Res 2022; 10:37518. [DOI: 10.52965/001c.37518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
This article is a systematic review of mental health in urologic oncology patients with prostate cancer (PCa), bladder cancer (BC), renal cell carcinoma (RCC), testicular cancer (TC), or penile cancer (PeCa). For all pathologies, a focus on increasing quality of life post-treatment demonstrated a positive impact in reducing Mental Health Illness (MHI) prevalence. Cancer specific mental health care may be given to patients to reduce suicide risk in BC patients and sexual identify and masculinity counseling may improve mental health for TC or PeCa patients. In order to better accommodate patient’s mental health needs when undergoing GU cancer treatment, we recommend incorporation of mental health metrics such as questionnaires to assess early treatment of MHI, a greater emphasis on psychosocial support with the patient’s loved ones, peers, and healthcare team, alongside advising healthy habits such as exercise which has been shown to drastically reduce MHI incidence across all pathologies. We hope that these measures conducted by urologists and oncologists, alongside possible coordination with psychiatrists and psychologists for psychotherapy, psychopharmacology, and neuro-stimulation treatment modems may be helpful in the long term to reduce MHI incidence in urology oncology patients. Given the higher incidence of MHI in oncology patients and in the patient population after the Covid-19 pandemic, MHI awareness in the sphere of urologic oncologic treatment continues to be crucial when creating a collaborative treatment platform for patients.
Collapse
Affiliation(s)
| | | | | | | | | | - Omar Viswanath
- Department of Anesthesia, Critical Care, and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School; Valley Anesthesiology and Pain Consultants, Envision Physician Services; Department of Anesthesiology, University of Arizona College of Medicine Phoenix;Department of Anesthesiology, Creighton University School of Medicine
| | | | - Ivan Urits
- Department of Anesthesia, Critical Care, and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School; Department of Anesthesiology, Louisiana State University Health Shreveport
| |
Collapse
|
4
|
Ottaviano M, Giunta EF, Rescigno P, Pereira Mestre R, Marandino L, Tortora M, Riccio V, Parola S, Casula M, Paliogiannis P, Cossu A, Vogl UM, Bosso D, Rosanova M, Mazzola B, Daniele B, Palmieri G, Palmieri G. The Enigmatic Role of TP53 in Germ Cell Tumours: Are We Missing Something? Int J Mol Sci 2021; 22:7160. [PMID: 34281219 PMCID: PMC8267694 DOI: 10.3390/ijms22137160] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/27/2021] [Accepted: 06/28/2021] [Indexed: 12/24/2022] Open
Abstract
The cure rate of germ cell tumours (GCTs) has significantly increased from the late 1970s since the introduction of cisplatin-based therapy, which to date remains the milestone for GCTs treatment. The exquisite cisplatin sensitivity has been mainly explained by the over-expression in GCTs of wild-type TP53 protein and the lack of TP53 somatic mutations; however, several other mechanisms seem to be involved, many of which remain still elusive. The findings about the role of TP53 in platinum-sensitivity and resistance, as well as the reported evidence of second cancers (SCs) in GCT patients treated only with surgery, suggesting a spectrum of cancer predisposing syndromes, highlight the need for a deepened understanding of the role of TP53 in GCTs. In the following report we explore the complex role of TP53 in GCTs cisplatin-sensitivity and resistance mechanisms, passing through several recent genomic studies, as well as its role in GCT patients with SCs, going through our experience of Center of reference for both GCTs and cancer predisposing syndromes.
Collapse
Affiliation(s)
- Margaret Ottaviano
- Oncology Unit, Ospedale del Mare, 80147 Naples, Italy; (D.B.); (M.R.); (B.D.)
- CRCTR Coordinating Rare Tumors Reference Center of Campania Region, 80131 Naples, Italy; (M.T.); (G.P.)
- IOSI (Oncology Institute of Southern Switzerland), Ente Ospedaliero Cantonale (EOC), 6500 Bellinzona, Switzerland; (R.P.M.); (L.M.); (U.M.V.)
| | - Emilio Francesco Giunta
- Oncology Unit, Department of Precision Medicine, Università Degli Studi Della Campania Luigi Vanvitelli, 80131 Naples, Italy;
| | - Pasquale Rescigno
- Interdisciplinary Group for Translational Research and Clinical Trials, Urological Cancers (GIRT-Uro), Candiolo Cancer Institute, FPO-IRCCS, Candiolo, 10160 Turin, Italy;
| | - Ricardo Pereira Mestre
- IOSI (Oncology Institute of Southern Switzerland), Ente Ospedaliero Cantonale (EOC), 6500 Bellinzona, Switzerland; (R.P.M.); (L.M.); (U.M.V.)
| | - Laura Marandino
- IOSI (Oncology Institute of Southern Switzerland), Ente Ospedaliero Cantonale (EOC), 6500 Bellinzona, Switzerland; (R.P.M.); (L.M.); (U.M.V.)
| | - Marianna Tortora
- CRCTR Coordinating Rare Tumors Reference Center of Campania Region, 80131 Naples, Italy; (M.T.); (G.P.)
| | - Vittorio Riccio
- Department of Clinical Medicine and Surgery, Università degli studi di Napoli Federico II, 80131 Naples, Italy; (V.R.); (S.P.)
| | - Sara Parola
- Department of Clinical Medicine and Surgery, Università degli studi di Napoli Federico II, 80131 Naples, Italy; (V.R.); (S.P.)
| | - Milena Casula
- Institute of Genetics and Biomedical Research (IRGB), National Research Council (CNR), 07100 Sassari, Italy; (M.C.); (G.P.)
| | - Panagiotis Paliogiannis
- Departments of Biomedical Sciences and Medical, Surgical, Experimental Sciences, University of Sassari, 07100 Sassari, Italy; (P.P.); (A.C.)
| | - Antonio Cossu
- Departments of Biomedical Sciences and Medical, Surgical, Experimental Sciences, University of Sassari, 07100 Sassari, Italy; (P.P.); (A.C.)
| | - Ursula Maria Vogl
- IOSI (Oncology Institute of Southern Switzerland), Ente Ospedaliero Cantonale (EOC), 6500 Bellinzona, Switzerland; (R.P.M.); (L.M.); (U.M.V.)
| | - Davide Bosso
- Oncology Unit, Ospedale del Mare, 80147 Naples, Italy; (D.B.); (M.R.); (B.D.)
| | - Mario Rosanova
- Oncology Unit, Ospedale del Mare, 80147 Naples, Italy; (D.B.); (M.R.); (B.D.)
| | - Brunello Mazzola
- Department of Urology, Ente Ospedaliero Cantonale (EOC), 6600 Locarno, Switzerland;
| | - Bruno Daniele
- Oncology Unit, Ospedale del Mare, 80147 Naples, Italy; (D.B.); (M.R.); (B.D.)
| | - Giuseppe Palmieri
- Institute of Genetics and Biomedical Research (IRGB), National Research Council (CNR), 07100 Sassari, Italy; (M.C.); (G.P.)
- Departments of Biomedical Sciences and Medical, Surgical, Experimental Sciences, University of Sassari, 07100 Sassari, Italy; (P.P.); (A.C.)
| | - Giovannella Palmieri
- CRCTR Coordinating Rare Tumors Reference Center of Campania Region, 80131 Naples, Italy; (M.T.); (G.P.)
| |
Collapse
|
5
|
Li Y, Lu Q, Wang Y, Ma S. Racial differences in testicular cancer in the United States: descriptive epidemiology. BMC Cancer 2020; 20:284. [PMID: 32252689 PMCID: PMC7137202 DOI: 10.1186/s12885-020-06789-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 03/26/2020] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Testicular cancer (TC) is the most common malignancy in young adult men, and in many countries the incidence rates of testicular cancer have been increasing since the middle of the twentieth century. Since disease presentation and tumor progression patterns are often heterogeneous across racial groups, there may be important racial differences in recent TC trends. METHODS In this study, Surveillance, Epidemiology, and End Results (SEER) data on TC patients diagnosed between 1973 and 2015 were analyzed, including the following racial/ethnic groups: non-Hispanic whites (NHW), Hispanic whites (HW), blacks, and Asians and Pacific Islanders (API). Patient characteristics, age-adjusted incidence rates, and survival were compared across racial groups. A multivariate Cox model was used to analyze the survival data of TC patients, in order to evaluate racial differences across several relevant factors, including marital status, age group, histologic type, treatment, stage, and tumor location. RESULTS NHWs had the highest incidence rates, followed by blacks, HWs, and APIs. There were significant survival differences among the racial groups, with NHWs having the highest survival rates and blacks having the lowest. CONCLUSION An analysis of SEER data showed that racial differences existed among TC patients in the United States with respect to patient characteristics, incidence, and survival. The results can be useful to stakeholders interested in reducing the burden of TC morbidity and mortality.
Collapse
Affiliation(s)
- Yang Li
- Center for Applied Statistics, Renmin University of China, Beijing, China
- School of Statistics, Renmin University of China, Beijing, China
| | - Qi Lu
- School of Statistics, Renmin University of China, Beijing, China
| | - Yu Wang
- Center for Applied Statistics, Renmin University of China, Beijing, China.
- School of Statistics, Renmin University of China, Beijing, China.
| | - Shuangge Ma
- School of Statistics, Renmin University of China, Beijing, China
- School of Public Health, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
6
|
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.
Collapse
|
7
|
Hellesnes R, Kvammen Ø, Myklebust TÅ, Bremnes RM, Karlsdottir Á, Negaard HFS, Tandstad T, Wilsgaard T, Fosså SD, Haugnes HS. Continuing increased risk of second cancer in long-term testicular cancer survivors after treatment in the cisplatin era. Int J Cancer 2019; 147:21-32. [PMID: 31597192 DOI: 10.1002/ijc.32704] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 08/27/2019] [Accepted: 09/10/2019] [Indexed: 12/13/2022]
Abstract
Using complete information on total treatment burden, this population-based study aimed to investigate second cancer (SC) risk in testicular cancer survivors (TCS) treated in the cisplatin era. The Cancer Registry of Norway identified 5,625 1-year TCS diagnosed 1980-2009. Standardized incidence ratios (SIRs) were calculated to evaluate the total and site-specific incidence of SC compared to the general population. Cox regression analyses evaluated the effect of treatment on the risk of SC. After a median observation time of 16.6 years, 572 TCS developed 651 nongerm cell SCs. The SC risk was increased after surgery only (SIR 1.28), with site-specific increased risks of thyroid cancer (SIR 4.95) and melanoma (SIR 1.94). After chemotherapy (CT), we observed 2.0- to 3.7-fold increased risks for cancers of the small intestine, bladder, kidney and lung. There was a 1.6- to 2.1-fold increased risk of SC after ≥2 cycles of cisplatin-based CT. Radiotherapy (RT) was associated with 1.5- to 4.4-fold increased risks for cancers of the stomach, small intestine, liver, pancreas, lung, kidney and bladder. After combined CT and RT, increased risks emerged for hematological malignancies (SIR 3.23). TCS treated in the cisplatin era have an increased risk of developing SC, in particular after treatment with cisplatin-based CT and/or RT.
Collapse
Affiliation(s)
- Ragnhild Hellesnes
- Department of Oncology, University Hospital of North Norway, Tromsø, Norway.,Department of Clinical Medicine, UiT The Arctic University, Tromsø, Norway
| | - Øivind Kvammen
- Department of Oncology, Ålesund Hospital, Ålesund, Norway.,Department of Clinical and Molecular Medicine, The Norwegian University of Science and Technology, Trondheim, Norway
| | - Tor Å Myklebust
- Department of Research and Innovation, Møre and Romsdal Hospital Trust, Ålesund, Norway.,Department of Registration, Cancer Registry of Norway, Oslo, Norway
| | - Roy M Bremnes
- Department of Oncology, University Hospital of North Norway, Tromsø, Norway.,Department of Clinical Medicine, UiT The Arctic University, Tromsø, Norway
| | - Ása Karlsdottir
- Department of Oncology, Haukeland University Hospital, Bergen, Norway
| | | | - Torgrim Tandstad
- Department of Clinical and Molecular Medicine, The Norwegian University of Science and Technology, Trondheim, Norway.,The Cancer Clinic, St. Olav's University Hospital, Trondheim, Norway
| | - Tom Wilsgaard
- Department of Community Medicine, UiT The Arctic University, Tromsø, Norway
| | - Sophie D Fosså
- Department of Registration, Cancer Registry of Norway, Oslo, Norway.,Department of Oncology, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Hege S Haugnes
- Department of Oncology, University Hospital of North Norway, Tromsø, Norway.,Department of Clinical Medicine, UiT The Arctic University, Tromsø, Norway
| |
Collapse
|
8
|
Perspectives on a Global Change in Testicular Cancer Incidence. Eur Urol 2019; 76:624-625. [DOI: 10.1016/j.eururo.2019.07.038] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 07/24/2019] [Indexed: 11/18/2022]
|
9
|
Family history of cancer and risk of paediatric and young adult's testicular cancer: A Norwegian cohort study. Br J Cancer 2019; 120:1007-1014. [PMID: 30967648 PMCID: PMC6734662 DOI: 10.1038/s41416-019-0445-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 03/13/2019] [Accepted: 03/13/2019] [Indexed: 12/17/2022] Open
Abstract
Background The aim of this study was to examine the association of a family history of cancer with the risk of testicular cancer in young adults. Methods This is a prospective cohort study including 1,974,287 males born 1951–2015, of whom 2686 were diagnosed with TC before the age of 30. Results A history of TC in male relatives was significantly associated with a diagnosis of TC among children and young adults, including brothers (6.3-fold), sons (4.7-fold), fathers (4.4-fold), paternal uncles (2.0-fold) and maternal uncles (1.9-fold). Individuals with a father diagnosed with a carcinoma or sarcoma showed an elevated risk (1.1-fold and 1.8-fold, respectively). A family history of mesothelioma was positively associated with a risk of TC [(father (2.8-fold), mother (4.6-fold) and maternal uncles and aunt (4.4-fold)]. Elevated risks were also observed when siblings were diagnosed with malignant melanoma (1.4-fold). The risk of TC was also increased when fathers (11.1-fold), paternal (4.9-fold) and maternal uncles and aunts (4.6-fold) were diagnosed with malignant neuroepithelial-tumours. Conclusion We found an increased risk of TC among children and young adults with a family history of TC, carcinoma, mesothelioma, sarcoma, malignant melanoma and malignant neuroepithelial tumours. Hereditary cancer syndromes might underlie some of the associations reported in this study.
Collapse
|
10
|
Le Fèvre C, Vigneron C, Schuster H, Walter A, Marcellin L, Massard G, Lutz P, Noël G. Metastatic mediastinal mature teratoma with malignant transformation in a young man with an adenocarcinoma in a Klinefelter's syndrome: Case report and review of the literature. Cancer Radiother 2018; 22:255-263. [DOI: 10.1016/j.canrad.2017.10.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 09/18/2017] [Accepted: 10/19/2017] [Indexed: 11/25/2022]
|
11
|
Loveday C, Litchfield K, Levy M, Holroyd A, Broderick P, Kote-Jarai Z, Dunning AM, Muir K, Peto J, Eeles R, Easton DF, Dudakia D, Orr N, Pashayan N, Reid A, Huddart RA, Houlston RS, Turnbull C. Validation of loci at 2q14.2 and 15q21.3 as risk factors for testicular cancer. Oncotarget 2018; 9:12630-12638. [PMID: 29560096 PMCID: PMC5849160 DOI: 10.18632/oncotarget.23117] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 11/15/2017] [Indexed: 01/21/2023] Open
Abstract
Testicular germ cell tumor (TGCT), the most common cancer in men aged 18 to 45 years, has a strong heritable basis. Genome-wide association studies (GWAS) have proposed single nucleotide polymorphisms (SNPs) at a number of loci influencing TGCT risk. To further evaluate the association of recently proposed risk SNPs with TGCT at 2q14.2, 3q26.2, 7q36.3, 10q26.13 and 15q21.3, we analyzed genotype data on 3,206 cases and 7,422 controls. Our analysis provides independent replication of the associations for risk SNPs at 2q14.2 (rs2713206 at P = 3.03 × 10-2; P-meta = 3.92 × 10-8; nearest gene, TFCP2L1) and rs12912292 at 15q21.3 (P = 7.96 × 10-11; P-meta = 1.55 × 10-19; nearest gene PRTG). Case-only analyses did not reveal specific associations with TGCT histology. TFCP2L1 joins the growing list of genes located within TGCT risk loci with biologically plausible roles in developmental transcriptional regulation, further highlighting the importance of this phenomenon in TGCT oncogenesis.
Collapse
Affiliation(s)
- Chey Loveday
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | - Kevin Litchfield
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
- Translational Cancer Therapeutics Laboratory, The Francis Crick Institute, London, UK
| | - Max Levy
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | - Amy Holroyd
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | - Peter Broderick
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | - Zsofia Kote-Jarai
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | - Alison M Dunning
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Kenneth Muir
- Division of Health Sciences, Warwick Medical School, Warwick University, Warwick, UK
- Institute of Population Health, University of Manchester, Manchester, UK
| | - Julian Peto
- Department of Non-communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
| | - Rosalind Eeles
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
- Royal Marsden NHS Foundation Trust, London, UK
| | - Douglas F Easton
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, UK
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Darshna Dudakia
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | - Nick Orr
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - Nora Pashayan
- Department of Applied Health Research, University College London, London, UK
| | - Alison Reid
- Academic Uro-oncology Unit, The Royal Marsden NHS Foundation Trust, Sutton, Surrey, UK
| | - Robert A Huddart
- Academic Radiotherapy Unit, Institute of Cancer Research, Sutton, Surrey, UK
| | - Richard S Houlston
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | - Clare Turnbull
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
- William Harvey Research Institute, Queen Mary University, London, UK
- Guys and St Thomas NHS Foundation Trust, London, UK
- National Cancer Registration and Analysis Service, Public Health England, London, UK
| |
Collapse
|
12
|
Abstract
Testicular germ cell tumor (TGCT) is the most common solid malignancy occurring in young men between 20 and 34 years of age, and its incidence has increased significantly over the last decades. Clinically several types of immunohistochemical markers are useful and sensitive. These new biomarkers are genes expressed in primordial germ cells/gonocytes and embryonic pluripotency-related cells but not in normal adult germ cells and they include OCT3/4, HMGA1 and 2, NANOG, SOX2, and LIN28. Gene expression in TGCT is regulated, at least in part, by DNA and histone modifications, and the epigenetic profile of these tumours is characterised by genome-wide demethylation. There are different epigenetic modifications in TGCT subtypes that reflect the normal developmental switch in primordial germ cells from an under to normally methylated genome.
Collapse
Affiliation(s)
- Paolo Chieffi
- Dipartimento di Psicologia, Università della Campania, Caserta, Italy
- Address correspondence to: Dr. Paolo Chieffi, Dipartimento di Psicologia, Viale Ellittico, 31 81100 Caserta, Italy. E-mail:
| |
Collapse
|
13
|
Abstract
PURPOSE OF REVIEW Testicular germ cell tumors (TGCTs) are a model for curable cancer because of exquisite chemosensitivity and incorporation of multimodal therapy. Nevertheless, our ability to predict metastases in early-stage disease and responders to chemotherapy in advanced disease is limited. Treatment options for cisplatin-resistant disease are sparse. A further understanding of TGCT biology may allow for more precise patient counseling and identify novel therapies in patients with cisplatin-resistant disease. RECENT FINDINGS Adult TGCTs are characterized by frequent chromosomal anomalies and low rates of somatic mutations. Large-scale integrated molecular analysis of early-stage TGCT patients is actively underway. In addition to ubiquitous gain of isochromosome 12p, current molecular studies have confirmed mutations of previously described genes (i.e., KIT and KRAS) and described novel mutations. Analysis of cisplatin-resistant cases has identified high rates of alterations within the TP53-MDM2 axis and a high proportion of patients with potentially actionable targets, including TP53-MDM2, PI3 kinase, and MAPK signaling pathway alterations. The role of epigenetics in TGCT development and prognosis is also being further characterized. SUMMARY Further molecular characterization of TGCT may allow for avoidance of unnecessary treatment in patients with early-stage disease and also provide new treatment options in patients with cisplatin-resistant disease.
Collapse
Affiliation(s)
- Solomon L Woldu
- University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| | | | | |
Collapse
|
14
|
Chieffi P. New perspective on molecular markers as promising therapeutic targets in germ cell tumors. Intractable Rare Dis Res 2016; 5:137-9. [PMID: 27195201 PMCID: PMC4869583 DOI: 10.5582/irdr.2016.01007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 02/26/2016] [Indexed: 01/28/2023] Open
Abstract
Testicular germ cell tumors (TGCTs) are the most frequent solid malignant tumors in men 20-40 years of age and the most frequent cause of death from solid tumors in this age group. TGCTs comprise two major histologic groups: seminomas and non-seminomas germ cell tumors (NSGCTs). NSGCTs can be further divided into embryonal carcinoma, Teratoma, yolk sac tumor, and choriocarcinoma. Seminomas and NSGCTs present significant differences in clinical features, therapy, and prognosis, and both show characteristics of the Primordial Germ Cells (PGCs). Many discovered biomarkers including HMGA1, GPR30, Aurora-B, estrogen receptor β, and others have given further advantages to discriminate between histological subgroups and could represent useful therapeutic targets.
Collapse
Affiliation(s)
- Paolo Chieffi
- Dipartimento di Psicologia, Seconda Università di Napoli, Caserta, Italy
| |
Collapse
|
15
|
Chieffi P, Boscia F. New discovered molecular markers as promising therapeutic targets in germ cell tumors. Expert Opin Orphan Drugs 2015. [DOI: 10.1517/21678707.2015.1074070] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
16
|
Litchfield K, Sultana R, Renwick A, Dudakia D, Seal S, Ramsay E, Powell S, Elliott A, Warren-Perry M, Eeles R, Peto J, Kote-Jarai Z, Muir K, Nsengimana J, Stratton MR, Easton DF, Bishop DT, Huddart RA, Rahman N, Turnbull C. Multi-stage genome-wide association study identifies new susceptibility locus for testicular germ cell tumour on chromosome 3q25. Hum Mol Genet 2015; 24:1169-76. [PMID: 25281660 PMCID: PMC4375409 DOI: 10.1093/hmg/ddu511] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 09/29/2014] [Indexed: 01/28/2023] Open
Abstract
Recent genome-wide association studies (GWAS) and subsequent meta-analyses have identified over 25 SNPs at 18 loci, together accounting for >15% of the genetic susceptibility to testicular germ cell tumour (TGCT). To identify further common SNPs associated with TGCT, here we report a three-stage experiment, involving 4098 cases and 18 972 controls. Stage 1 comprised previously published GWAS analysis of 307 291 SNPs in 986 cases and 4946 controls. In Stage 2, we used previously published customised Illumina iSelect genotyping array (iCOGs) data across 694 SNPs in 1064 cases and 10 082 controls. Here, we report new genotyping of eight SNPs showing some evidence of association in combined analysis of Stage 1 and Stage 2 in an additional 2048 cases of TGCT and 3944 controls (Stage 3). Through fixed-effects meta-analysis across three stages, we identified a novel locus at 3q25.31 (rs1510272) demonstrating association with TGCT [per-allele odds ratio (OR) = 1.16, 95% confidence interval (CI) = 1.06-1.27; P = 1.2 × 10(-9)].
Collapse
Affiliation(s)
- Kevin Litchfield
- Division of Genetics and Epidemiology, Institute of Cancer Research, London, UK
| | - Razvan Sultana
- Division of Genetics and Epidemiology, Institute of Cancer Research, London, UK
| | - Anthony Renwick
- Division of Genetics and Epidemiology, Institute of Cancer Research, London, UK
| | - Darshna Dudakia
- Division of Genetics and Epidemiology, Institute of Cancer Research, London, UK
| | - Sheila Seal
- Division of Genetics and Epidemiology, Institute of Cancer Research, London, UK
| | - Emma Ramsay
- Division of Genetics and Epidemiology, Institute of Cancer Research, London, UK
| | - Silvana Powell
- Division of Genetics and Epidemiology, Institute of Cancer Research, London, UK
| | - Anna Elliott
- Division of Genetics and Epidemiology, Institute of Cancer Research, London, UK
| | | | - Rosalind Eeles
- Division of Genetics and Epidemiology, Institute of Cancer Research, London, UK, Royal Marsden NHS Foundation Trust, London, UK
| | - Julian Peto
- Non-communicable Disease Epidemiology Department, London School of Hygiene and Tropical Medicine, London, UK
| | - Zsofia Kote-Jarai
- Division of Genetics and Epidemiology, Institute of Cancer Research, London, UK
| | - Kenneth Muir
- Institute of Population Health, University of Manchester, Manchester, UK
| | - Jeremie Nsengimana
- Section of Epidemiology and Biostatistics, Leeds Institute of Cancer and Pathology, Leeds, UK
| | | | - Douglas F Easton
- Genetic Epidemiology Unit, Strangeways Research Laboratory, Cancer Research UK, Cambridge, UK and
| | - D Timothy Bishop
- Section of Epidemiology and Biostatistics, Leeds Institute of Cancer and Pathology, Leeds, UK
| | - Robert A Huddart
- Academic Radiotherapy Unit, Institute of Cancer Research, Sutton, Surrey SM2 5NG, UK
| | - Nazneen Rahman
- Division of Genetics and Epidemiology, Institute of Cancer Research, London, UK, Royal Marsden NHS Foundation Trust, London, UK
| | - Clare Turnbull
- Division of Genetics and Epidemiology, Institute of Cancer Research, London, UK, Royal Marsden NHS Foundation Trust, London, UK,
| |
Collapse
|
17
|
Boublikova L, Buchler T, Stary J, Abrahamova J, Trka J. Molecular biology of testicular germ cell tumors: Unique features awaiting clinical application. Crit Rev Oncol Hematol 2014; 89:366-85. [DOI: 10.1016/j.critrevonc.2013.10.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2013] [Revised: 08/30/2013] [Accepted: 10/01/2013] [Indexed: 01/29/2023] Open
|
18
|
Mueller CM, Korde LA, McMaster ML, Peters JA, Bratslavsky G, Watkins RJ, Ling A, Kratz CP, Wulfsberg EA, Rosenberg PS, Greene MH. Familial testicular germ cell tumor: no associated syndromic pattern identified. Hered Cancer Clin Pract 2014; 12:3. [PMID: 24559313 PMCID: PMC3937045 DOI: 10.1186/1897-4287-12-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 02/12/2014] [Indexed: 11/10/2022] Open
Abstract
Background Testicular germ cell tumor (TGCT) is the most common malignancy in young men. Familial clustering, epidemiologic evidence of increased risk with family or personal history, and the association of TGCT with genitourinary (GU) tract anomalies have suggested an underlying genetic predisposition. Linkage data have not identified a rare, highly-penetrant, single gene in familial TGCT (FTGCT) cases. Based on its association with congenital GU tract anomalies and suggestions that there is an intrauterine origin to TGCT, we hypothesized the existence of unrecognized dysmorphic features in FTGCT. Methods We evaluated 38 FTGCT individuals and 41 first-degree relatives from 22 multiple-case families with detailed dysmorphology examinations, physician-based medical history and physical examination, laboratory testing, and genitourinary imaging studies. Results The prevalence of major abnormalities and minor variants did not significantly differ between either FTGCT individuals or their first-degree relatives when compared with normal population controls, except for tall stature, macrocephaly, flat midface, and retro-/micrognathia. However, these four traits were not manifest as a constellation of features in any one individual or family. We did detect an excess prevalence of the genitourinary anomalies cryptorchidism and congenital inguinal hernia in our population, as previously described in sporadic TGCT, but no congenital renal, retroperitoneal or mediastinal anomalies were detected. Conclusions Overall, our study did not identify a constellation of dysmorphic features in FTGCT individuals, which is consistent with results of genetic studies suggesting that multiple low-penetrance genes are likely responsible for FTGCT susceptibility.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Mark H Greene
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
19
|
Recent advances in molecular and cell biology of testicular germ-cell tumors. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2014; 312:79-100. [PMID: 25262239 DOI: 10.1016/b978-0-12-800178-3.00003-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Testicular germ-cell tumors (TGCTs) are the most frequent solid malignant tumors in men 20-40 years of age and the most frequent cause of death from solid tumors in this age group. TGCTs comprise two major histologic groups: seminomas and nonseminomas germ-cell tumors (NSGCTs). NSGCTs can be further divided into embryonal, carcinoma, Teratoma, yolk sac tumor, and choriocarcinoma. Seminomas and NSGCTs present significant differences in clinical features, therapy, and prognosis, and both show characteristics of the primordial germ cells. Many discovered biomarkers including OCT3/4, SOX2, SOX17, HMGA1, Nek2, GPR30, Aurora-B, estrogen receptor β, and others have given further advantages to discriminate between histological subgroups and could represent useful novel molecular targets for antineoplastic strategies. More insight into the pathogenesis of TGCTs is likely to improve disease management not only to better treatment of these tumors but also to a better understanding of stem cells and oncogenesis.
Collapse
|
20
|
Kouprina N, Lee NCO, Pavlicek A, Samoshkin A, Kim JH, Lee HS, Varma S, Reinhold WC, Otstot J, Solomon G, Davis S, Meltzer PS, Schleutker J, Larionov V. Exclusion of the 750-kb genetically unstable region at Xq27 as a candidate locus for prostate malignancy in HPCX1-linked families. Genes Chromosomes Cancer 2012; 51:933-48. [PMID: 22733720 DOI: 10.1002/gcc.21977] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Accepted: 05/08/2012] [Indexed: 12/14/2022] Open
Abstract
Several linkage studies provided evidence for the presence of the hereditary prostate cancer locus, HPCX1, at Xq27-q28. The strongest linkage peak of prostate cancer overlies a variable region of ~750 kb at Xq27 enriched by segmental duplications (SDs), suggesting that the predisposition to prostate cancer may be a genomic disorder caused by recombinational interaction between SDs. The large size of SDs and their sequence similarity make it difficult to examine this region for possible rearrangements using standard methods. To overcome this problem, direct isolation of a set of genomic segments by in vivo recombination in yeast (a TAR cloning technique) was used to perform a mutational analysis of the 750 kb region in X-linked families. We did not detect disease-specific rearrangements within this region. In addition, transcriptome and computational analyses were performed to search for nonannotated genes within the Xq27 region, which may be associated with genetic predisposition to prostate cancer. Two candidate genes were identified, one of which is a novel gene termed SPANXL that represents a highly diverged member of the SPANX gene family, and the previously described CDR1 gene that is expressed at a high level in both normal and malignant prostate cells, and mapped 210 kb of upstream the SPANX gene cluster. No disease-specific alterations were identified in these genes. Our results exclude the 750-kb genetically unstable region at Xq27 as a candidate locus for prostate malignancy. Adjacent regions appear to be the most likely candidates to identify the elusive HPCX1 locus.
Collapse
Affiliation(s)
- Natalay Kouprina
- Laboratory of Molecular Pharmacology, NCI, NIH, Bethesda, MD, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Le cancer du testicule : facteurs de risque génétiques et environnementaux. Basic Clin Androl 2012. [DOI: 10.1007/s12610-012-0164-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Abstract
Résumé
L'incidence du cancer du testicule (CT), qui est inégale d'une région à l'autre, progresse dans la plupart des pays y compris la France. L'origine la plus probable du CT est la non différenciation de cellules germinales souches pendant la vie fœtale. Ces cellules, maintenues dans un état immature, pourraient proliférer pour former une tumeur après la puberté. La fréquence de son association avec une cryptorchidie ou une infertilité a conduit à formuler l'hypothèse que le CT pouvait être un des constituants du syndrome de dysgénésie testiculaire. Le rôle des facteurs génétiques est suggéré par la fréquence de cas familiaux de CT mais aucun gène clairement responsable du cancer n'a été identifié jusqu'à présent. Parmi les nombreux gènes étudiés, ce sont ceux contrôlant la voie KITLG/KIT, qui participe à la régulation de la prolifération et de la fonction des cellules germinales primordiales, qui semblent jouer le rôle principal. Les études faites sur des populations migrantes et les jumeaux suggèrent par ailleurs que des facteurs environnementaux pourraient jouer un rôle essentiel dans la genèse du CT. Des dérégulations hormonales pendant la vie fœtale ou la puberté pourraient notamment favoriser le développement de CT. Cependant, l'exposition à des substances exogènes agissant comme des perturbateurs endocriniens est encore à démontrer.
Collapse
|
22
|
Richardson LC, Neri AJ, Tai E, Glenn JD. Testicular cancer: a narrative review of the role of socioeconomic position from risk to survivorship. Urol Oncol 2011; 30:95-101. [PMID: 22127018 DOI: 10.1016/j.urolonc.2011.09.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Revised: 09/28/2011] [Accepted: 09/30/2011] [Indexed: 01/09/2023]
Abstract
BACKGROUND Testicular cancer (TC) is one of the most curable cancers. Given survival rates of close to 100% with appropriate therapy, ensuring proper treatment is essential. We reviewed and summarized the literature on the association of socioeconomic position (SEP) along the cancer control spectrum from risk factors to survivorship. METHODS We searched PubMed from 1966 to 2011 using the following terms: testicular cancer, testicular neoplasm, poverty, and socioeconomic factors, retrieving 119 papers. After excluding papers for the non-English (10) language and non-relevance (46), we reviewed 63 papers. We abstracted information on socioeconomic position (SEP), including occupation, education, income, and combinations of the 3. Five areas were examined: risk factors, diagnosis, treatment, survival, and survivorship. RESULTS Most studies examined area-based measures, not individual measures of SEP. The majority of studies found an increased risk of developing TC with high SEP though recent papers have indicated increased risk in low-income populations. Regarding diagnosis, recent papers have indicated that lower levels of education and SEP are risk factors for later-stage TC diagnosis and hence higher TC mortality. For treatment, 1 study that examined the use of radiation therapy (RT) in stage I seminoma reported that living in a county with lower educational attainment led to lower use of RT. For survival (mortality), several studies found that men living in lower SEP geographic areas experience lower survival and higher mortality. CONCLUSION The strongest evidence for SEP impact on testicular germ cell tumor (TGCT) was found for the risk of developing cancer as well as survival. The association of SEP with TGCT risk appears to have changed over the last decade. Given the highly curable nature of TGCT, more research is needed to understand how SEP impacts diagnosis and treatment for TGCT and to design interventions to address disparities in TGCT outcomes and SEP.
Collapse
Affiliation(s)
- Lisa C Richardson
- Division of Cancer Prevention and Control, National Center for Chronic Disease Prevention and Health Promotion, Centers for Disease Control and Prevention, Atlanta, GA 30341, USA.
| | | | | | | |
Collapse
|
23
|
Turnbull C, Rahman N. Genome-wide association studies provide new insights into the genetic basis of testicular germ-cell tumour. ACTA ACUST UNITED AC 2011; 34:e86-96; discussion e96-7. [PMID: 21623831 DOI: 10.1111/j.1365-2605.2011.01162.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Testicular germ-cell tumour (TGCT) is the most common cancer in young men, and genetic epidemiological studies suggest that the disease has a strong genetic basis. Until 2009, very little of this genetic component had been explained. Genome-wide association studies have since identified eight SNPs at six loci which together account for approximately 15% of the genetic risk of TGCT and offer novel biological insights into testicular germ-cell oncogenesis. In this review, we summarize the genetic epidemiology of TGCT, detail the contribution genome-wide association studies have made to our understanding of the genetic basis of TGCT and reflect on how future technological advances may assist in revealing the remaining genetic factors underlying TGCT susceptibility.
Collapse
Affiliation(s)
- C Turnbull
- Section of Cancer Genetics, Institute of Cancer Research, Sutton, UK.
| | | |
Collapse
|
24
|
Looijenga LHJ, Gillis AJM, Stoop H, Biermann K, Oosterhuis JW. Dissecting the molecular pathways of (testicular) germ cell tumour pathogenesis; from initiation to treatment-resistance. ACTA ACUST UNITED AC 2011; 34:e234-51. [PMID: 21564133 DOI: 10.1111/j.1365-2605.2011.01157.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Human type II germ cell tumours (GCTs) originate from an embryonic germ cell, either as a primordial germ cell or gonocyte. This start determines the biological as well as clinical characteristics of this type of cancer, amongst others their totipotency as well as their overall (exceptional) sensitivity to DNA damaging agents. The histology of the precursor lesion, either carcinoma in situ or gonadoblastoma, depends on the level of testicularization (i.e. testis formation) of the gonad. The impact of either intrinsic (genetic) - and environmental factors involved in the pathogenesis is demonstrated by disorders of sex development as well as testicular dysgenesis syndrome as risk factors, including cryptorchidism, hypospadias and disturbed fertility as parameters. This knowledge allows identification of individuals at risk for development of this type of cancer, being a population of interest for screening. Factors known to regulate pluripotency during embryogenesis are proven to be of diagnostic value for type II GCTs, including OCT3/4, even applicable for non-invasive screening. In addition, presence of stem cell factor, also known as KITLG, allows distinction between delayed matured germ cells and the earliest stages of malignant transformation. This is of special interest because of the identified association between development of type II GCTs of the testis and a limited number of single nucleotide polymorphisms, including some likely related to KITL. Transition from the precursor lesion to an invasive cancer is associated with gain of the short arm of chromosome 12, in which multiple genes might be involved, including KRAS2 and possibly NANOG (pseudogenes). While most precursor lesions will progress to an invasive cancer, only a limited number of cancers will develop treatment resistance. Putative explanatory mechanisms are identified, including presence of microsatellite instability, BRAF mutations, apoptosis suppression and p21 sub-cellular localization. It remains to be investigated how these different pathways integrate to each other and how informative they are at the patient-individual level. Further understanding will allow development of more targeted treatment, which will benefit quality of life of these young cancer patients.
Collapse
Affiliation(s)
- L H J Looijenga
- Department of Pathology, Erasmus MC-University Medical Center Rotterdam, Josephine Nefkens Institute, Daniel den Hoed Cancer Center, Rotterdam, The Netherlands.
| | | | | | | | | |
Collapse
|
25
|
Sijmons RH, Vos YJ, Herkert JC, Bos KK, Lutke Holzik MF, Hoekstra-Weebers JEHM, Hofstra RMW, Hoekstra HJ. Screening for germline DND1 mutations in testicular cancer patients. Fam Cancer 2010; 9:439-42. [PMID: 20411342 PMCID: PMC2921500 DOI: 10.1007/s10689-010-9340-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Although several observations suggest that a strong genetic predisposition to developing testicular germ cell tumors (TGCT) exists, no associated, highly penetrant germline mutations have been identified so far. In the 129/Sv mouse strain, a germline mutation in the DND1 gene has been shown to strongly increase the TGCT risk. We screened 272 men with TGCT (89% sporadic cases, 11% familial) for germline mutations in the human homologue of DND1. A single nucleotide substitution c.657C > G (p.Asp219Glu) was observed in a non-familial case of testicular embryonal carcinoma. The variant was also present in the patient’s asymptomatic father and two brothers, but not observed in 210 control chromosomes. The wild type DND1 allele was not lost in the patient’s tumor. In silico analysis of the variant predicts it to be non-pathogenic. We conclude that germline DND1 mutations are unlikely to contribute significantly to human testicular germ cell tumor susceptibility. The role of human DND1 in normal physiology and disease, however, is still virtually unknown and it therefore warrants further research.
Collapse
Affiliation(s)
- Rolf H Sijmons
- Department of Genetics, University Medical Center Groningen, University of Groningen, PO Box 30001, 9700 RB, Groningen, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Baldini E, Arlot-Bonnemains Y, Mottolese M, Sentinelli S, Antoniani B, Sorrenti S, Salducci M, Comini E, Ulisse S, D'Armiento M. Deregulation of Aurora kinase gene expression in human testicular germ cell tumours. Andrologia 2010; 42:260-7. [PMID: 20629650 DOI: 10.1111/j.1439-0272.2009.00987.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The Aurora kinases regulate chromosome segregation and cytokinesis, and alterations in their expression associate with cell malignant transformation. In this study, we demonstrated by qRT-PCR analysis of 14 seminomas that Aurora-A mRNA was, with respect to control tissues, augmented in five of 14 tumour tissues by 2.17 +/- 0.30 fold (P < 0.05) and reduced in 9 to 0.38 +/- 0.10 (P < 0.01). Aurora-B mRNA was increased in 11 tumour tissues by 4.33 +/- 0.82 fold (P < 0.01) and reduced in 3 to 0.41 +/- 0.11 fold. Aurora-C mRNA was reduced to 0.20 +/- 0.32 fold (P < 0.01) in 13 seminomas and up-regulated in one case. Western blot experiments, performed on protein extracts of nine seminomas and six normal testes, showed an up-regulation of Aurora-B protein by 10.14 +/- 3.51 fold (P < 0.05), while Aurora-A protein was found increased in four seminomas by 2.16 +/- 0.43 (P < 0.05), unchanged in three and reduced in two tumour tissues. Aurora-C protein was increased by 9.2 +/- 2.90 fold (P < 0.05), suggesting that post-transcriptional mechanisms modulate its expression. In conclusion, we demonstrated that expression of Aurora kinases is deregulated in seminomas, suggesting that they may play a role in the progression of testicular cancers.
Collapse
Affiliation(s)
- E Baldini
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Alam SS, Cantwell MM, Cardwell CR, Cook MB, Murray LJ. Maternal body mass index and risk of testicular cancer in male offspring: a systematic review and meta-analysis. Cancer Epidemiol 2010; 34:509-15. [PMID: 20800565 DOI: 10.1016/j.canep.2010.07.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2010] [Revised: 07/01/2010] [Accepted: 07/02/2010] [Indexed: 01/07/2023]
Abstract
OBJECTIVES To date a number of studies have examined the association between maternal weight and testicular cancer risk although results have been largely inconsistent. This systematic review and meta-analysis investigated the nature of this association. METHODS Search strategies were conducted in Ovid Medline (1950-2009), Embase (1980-2009), Web of Science (1970-2009), and CINAHL (1937-2009) using keywords for maternal weight (BMI) and testicular cancer. RESULTS The literature search produced 1689 hits from which 63 papers were extracted. Only 7 studies met the pre-defined criteria. Random effects meta-analyses were conducted. The combined unadjusted OR (95% CI) of testicular cancer in the highest reported category of maternal BMI compared with the moderate maternal BMI was 0.82 (0.65-1.02). The Cochran's Q P value was 0.82 and the corresponding I(2) was 0%, both indicating very little variability among studies. The combined unadjusted OR (95% CI) for testicular cancer risk in the lowest reported category of maternal BMI compared to a moderate maternal BMI category was 0.88 (0.65-1.20). The Cochran's Q P value was 0.05 and the corresponding I(2) was 54%, indicating evidence of statistical heterogeneity. The combined unadjusted OR (95% CI) of testicular cancer risk per unit increase in maternal BMI was 1.01 (0.97-1.06). The Cochran's Q test had a P value of 0.05 and the corresponding I(2) was 55% indicating evidence of statistical heterogeneity. CONCLUSION This meta-analysis, which included a small number of studies, showed that a higher maternal weight does not increase the risk of testicular cancer in male offspring. Though an inverse association between high maternal BMI and testicular cancer risk was detected, it was not statistically significant. Further primary studies with adjustment for appropriate confounders are required.
Collapse
Affiliation(s)
- Shama S Alam
- Cancer Epidemiology & Health Services Research Group, Centre for Public Health, Queen's University Belfast, Northern Ireland, United Kingdom.
| | | | | | | | | |
Collapse
|
28
|
Nødtvedt A, Gamlem H, Gunnes G, Grotmol T, Indrebø A, Moe L. Breed differences in the proportional morbidity of testicular tumours and distribution of histopathologic types in a population-based canine cancer registry. Vet Comp Oncol 2010; 9:45-54. [PMID: 21303453 DOI: 10.1111/j.1476-5829.2010.00231.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Histologically verified tumours submitted to the Norwegian Canine Cancer Register from 1990 to 1998 were studied (n=14,401). The proportion of testicular tumours (n=345) was 2.4%, and the breakdown of histological tumour diagnoses is presented. The frequency of the most common histopathological types was 33% interstitial (Leydig), 26.4% Sertoli and 33.9% seminomas/germ cell tumours. The average age at diagnosis was 10 years, but was significantly lower for Sertoli cell tumours (8.6 years) than for the other tumour types. Following a histopathological re-evaluation, 22.5% of the original tumor diagnoses were modified. Proportional morbidity ratios were calculated and individuals from the breeds Shetland sheepdog and Collie were five times more likely to have testicular tumours than the overall average for the registry. Breed differences in the distribution of histopathologic types were observed. Shetland sheepdog and Collie were most commonly diagnosed with Sertoli cell tumours, while all tumours from Norwegian elkhound in this material were seminomas.
Collapse
Affiliation(s)
- A Nødtvedt
- Norwegian School of Veterinary Science, Department of Companion Animal Clinical Sciences, Oslo, Norway.
| | | | | | | | | | | |
Collapse
|
29
|
Looijenga LHJ, Hersmus R, de Leeuw BHCGM, Stoop H, Cools M, Oosterhuis JW, Drop SLS, Wolffenbuttel KP. Gonadal tumours and DSD. Best Pract Res Clin Endocrinol Metab 2010; 24:291-310. [PMID: 20541153 DOI: 10.1016/j.beem.2009.10.002] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Disorders of sex development (DSD), previously referred to as intersex, has been recognised as one of the main risk factors for development of type II germ cell tumours (GCTs), that is, seminomas/dysgerminomas and non-seminomas (e.g., embryonal carcinoma, yolk sac tumour, choriocarcinoma and teratoma). Within the testis, this type of cancer is the most frequent malignancy in adolescent and young adult Caucasian males. Although these males are not known to have dysgenetic gonads, the similarities in the resulting tumours suggest a common aetiological mechanism(s),--genetically, environmentally or a combination of both. Within the group of DSD patients, being in fact congenital conditions, the risk of malignant transformation of germ cells is highly heterogeneous, depending on a number of parameters, some of which have only recently been identified. Understanding of these recent insights will stimulate further research, with the final aim to develop an informative clinical decision tree for DSD patients, which includes optimal (early) diagnosis without overtreatment, such as prophylactic gonadectomy in the case of a low tumour risk.
Collapse
Affiliation(s)
- Leendert H J Looijenga
- Department of Pathology, Erasmus MC-University Medical Center Rotterdam, Josephine Nefkens Institute, Daniel den Hoed Cancer Center, Rotterdam, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Townsend JS, Richardson LC, German RR. Incidence of testicular cancer in the United States, 1999-2004. Am J Mens Health 2009; 4:353-60. [PMID: 20031937 DOI: 10.1177/1557988309356101] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Testicular cancer is rare but primarily affects young men. To characterize the current incidence of testicular cancer in the United States, U.S. Cancer Statistics data from 1999 through 2004 were examined. Age-adjusted (2000 U.S. standard) incidence rates were calculated for seminoma and nonseminoma testicular germ cell tumors (TGCTs). Hispanic men had the largest increase in incidence rates for nonseminomas, followed by non-Hispanic White men (annual percentage change of 3.2% and 1.9%, respectively, p < .05). Nonseminomas peaked at a younger age for Hispanic, American Indian/Alaska Native (AIAN), and Asian/Pacific Islander (API) men. Whereas 9.6% of TGCTs were diagnosed at a distant stage in non-Hispanic White men, more Hispanic (16.1%), Black (13.8%), AIAN (16.8%), and API (14.9%) men with TGCTs were diagnosed with distant stage. Monitoring incidence rates for rare cancers by race/ethnicity has improved with national population-based cancer registry coverage. Disparities in diagnosis stage have implications for effective treatment of TGCTs.
Collapse
Affiliation(s)
- Julie S Townsend
- Centers for Disease Control and Prevention, Atlanta, GA 30341, USA.
| | | | | |
Collapse
|
31
|
Abstract
Semen quality appears to have declined in recent decades in some populations, e.g. north-western Europe. At the same time, couple fertility may have increased. Hypotheses are suggested for this apparent inconsistency. Alongside the deterioration of spermatogenesis there is clear evidence of an increase in other related problems, notably testicular cancer. The sharply rising trend in this condition started a century ago--decades earlier than sometimes thought. This and other evidence clearly indicates an environmental origin, but there is also a definite genetic component. The relationship of genetics and environment is discussed in the context of the puzzle that infertility is inherited, which appears to be impossible from an evolutionary standpoint. Poor semen quality is related not only to testicular cancer but also to zygote development, in which cancer-like disruption of the genetic apparatus is observed, with serious implications for offspring health. This needs to be seen in the context that human reproduction is prone to a higher degree of impairment than that of other mammalian species, in relation to spermatogenesis, couple fertility, early pregnancy loss and embryonic aneuploidy; female- and male-mediated pathways are both implicated. It is unclear whether such human specificity originated on an evolutionary/genetic or a historico-social timescale, which is important in relation to pathogenesis. The evidence clearly indicates that the currently most popular explanation for male reproductive system impairment, the endocrine disruption hypothesis, cannot explain the main features of the descriptive epidemiology. An alternative pathogenesis is outlined, and some possible exposures considered that could be responsible.
Collapse
Affiliation(s)
- Michael Joffe
- Department of Epidemiology and Public Health, Imperial College, London W2 1PF, UK.
| |
Collapse
|
32
|
Mai PL, Chen BE, Tucker K, Friedlander M, Phillips KA, Hogg D, Jewett MAS, Bodrogi I, Geczi L, Olah E, Heimdal K, Fosså SD, Nathanson KL, Korde L, Easton DF, Dudakia D, Huddart R, Stratton MR, Bishop DT, Rapley EA, Greene MH. Younger age-at-diagnosis for familial malignant testicular germ cell tumor. Fam Cancer 2009; 8:451-6. [PMID: 19609727 DOI: 10.1007/s10689-009-9264-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2009] [Accepted: 06/30/2009] [Indexed: 11/30/2022]
Abstract
One of the clinical hallmarks of hereditary cancer susceptibility disorders is a younger-than-usual age at diagnosis. Familial aggregation of testicular germ cell tumor (TGCT) has been reported, but data on whether familial TGCT cases are diagnosed at an earlier age are inconclusive. Here we compared the age at diagnosis of familial TGCT cases with that of population cases in several countries. Familial TGCT is defined as affected individuals from families with >or=2 cases of TGCT. Age at diagnosis of familial cases from the United States, Canada, United Kingdom, Australia and New Zealand, Norway, and Hungary was compared to cases identified in population-based cancer registries from the respective country, using the generalized estimation equation method. Age at diagnosis was statistically significantly younger for familial TGCT cases from North America (P = 0.024), the United Kingdom (P < 0.0001), and Australia and New Zealand (P = 0.0033) compared with population cases. When stratified by histology, the difference in age at diagnosis distribution between familial and population cases was observed for seminoma cases from North America (P = 0.002) and the United Kingdom (P < 0.0001) and non-seminoma cases from the United Kingdom (P = 0.029) and Australia and New Zealand (P = 0.0023). In summary, we found that the age at diagnosis for familial TGCT cases is, on the average, 2-3 years younger than that for the population cases in North America, United Kingdom, and Australia and New Zealand. The younger age at diagnosis might be suggestive of a genetic basis for familial TGCT.
Collapse
Affiliation(s)
- Phuong L Mai
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MA 20852, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Rapley EA, Turnbull C, Al Olama AA, Dermitzakis ET, Linger R, Huddart RA, Renwick A, Hughes D, Hines S, Seal S, Morrison J, Nsengimana J, Deloukas P, Rahman N, Bishop DT, Easton DF, Stratton MR. A genome-wide association study of testicular germ cell tumor. Nat Genet 2009; 41:807-10. [PMID: 19483681 PMCID: PMC2871592 DOI: 10.1038/ng.394] [Citation(s) in RCA: 277] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2008] [Accepted: 05/05/2009] [Indexed: 01/20/2023]
Abstract
We conducted a genome-wide association study for testicular germ cell tumor (TGCT), genotyping 307,666 SNPs in 730 cases and 1,435 controls from the UK and replicating associations in a further 571 cases and 1,806 controls. We found strong evidence for susceptibility loci on chromosome 5 (per allele OR = 1.37 (95% CI = 1.19-1.58), P = 3 x 10(-13)), chromosome 6 (OR = 1.50 (95% CI = 1.28-1.75), P = 10(-13)) and chromosome 12 (OR = 2.55 (95% CI = 2.05-3.19), P = 10(-31)). KITLG, encoding the ligand for the receptor tyrosine kinase KIT, which has previously been implicated in the pathogenesis of TGCT and the biology of germ cells, may explain the association on chromosome 12.
Collapse
Affiliation(s)
- Elizabeth A Rapley
- Section of Cancer Genetics, Institute of Cancer Research, Sutton, Surrey, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Looijenga LHJ. Human testicular (non)seminomatous germ cell tumours: the clinical implications of recent pathobiological insights. J Pathol 2009; 218:146-62. [PMID: 19253916 DOI: 10.1002/path.2522] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Human germ cell tumours (GCTs) comprise several types of neoplasias with different pathogeneses and clinical behaviours. A classification into five subtypes has been proposed. Here, the so-called type II testicular GCTs (TGCTs), ie the seminomas and non-seminomas, will be reviewed with emphasis on pathogenesis and clinical implications. Various risk factors have been identified that define subpopulations of men who are amenable to early diagnosis. TGCTs are omnipotent, able to generate all differentiation lineages, both embryonic and extra-embryonic, as well as the germ cell lineage itself. The precursor lesion, composed of primordial germ cells/gonocytes, is referred to as carcinoma in situ of the testis (CIS) and gonadoblastoma of the dysgenetic gonad. These pre-malignant cells retain embryonic characteristics, which probably explains the unique responsiveness of the derived tumours to DNA-damaging agents. Development of CIS and gonadoblastoma is crucially dependent on the micro-environment created by Sertoli cells in the testis, and granulosa cells in the dysgenetic gonad. OCT3/4 has high sensitivity and specificity for CIS/gonadoblastoma, seminoma, and embryonal carcinoma, and is useful for the detection of CIS cells in semen, thus a promising tool for non-invasive screening. Overdiagnosis of CIS due to germ cell maturation delay can be avoided using immunohistochemical detection of stem cell factor (SCF). Immunohistochemistry is helpful in making the distinction between seminoma and embryonal carcinoma, especially SOX17 and SOX2. The different non-seminomatous histological elements can be recognized using various markers, such as AFP and hCG, while others need confirmation. The value of micro-satellite instability as well as BRAF mutations in predicting treatment resistance needs validation in prospective trials. The availability of representative cell lines, both for seminoma and for embryonal carcinoma, allows mechanistic studies into the initiation and progression of this disease.
Collapse
Affiliation(s)
- Leendert H J Looijenga
- Department of Pathology, Erasmus MC-Erasmus University Medical Center, Daniel den Hoed Cancer Center, Josephine Nefkens Institute, Rotterdam, The Netherlands.
| |
Collapse
|
35
|
Anderson PD, Lam MY, Poirier C, Bishop CE, Nadeau JH. The role of the mouse y chromosome on susceptibility to testicular germ cell tumors. Cancer Res 2009; 69:3614-8. [PMID: 19351821 DOI: 10.1158/0008-5472.can-08-4881] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Testicular germ cell tumors (TGCT) are sex limited, occurring only in males with a Y chromosome. Recently, the gr/gr deletion on the human Y chromosome was associated with increased risk of TGCTs. In addition, the presence of Y chromosome sequences is associated with TGCTs in cases of gonadal dysgenesis. TGCTs in strain 129 males recapitulate many aspects of testicular cancer in human infants and can be used to evaluate the role of the Y chromosome in TGCT risk. We used chromosome substitution strains and a sex-reversing mutant to test the role of the Y chromosome on TGCT susceptibility. Our results show that a Y-linked gene that does not differ among the tested strains is essential for tumorigenesis.
Collapse
Affiliation(s)
- Philip D Anderson
- Department of Genetics and Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | | | | | | | | |
Collapse
|
36
|
van de Geijn GJM, Hersmus R, Looijenga LHJ. Recent developments in testicular germ cell tumor research. ACTA ACUST UNITED AC 2009; 87:96-113. [DOI: 10.1002/bdrc.20140] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
37
|
Hussain SA, Ma YT, Palmer DH, Hutton P, Cullen MH. Biology of testicular germ cell tumors. Expert Rev Anticancer Ther 2009; 8:1659-73. [PMID: 18925857 DOI: 10.1586/14737140.8.10.1659] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Germ cell tumors are derived from cells of the germ cell lineage and are the most common solid malignancies to affect young Caucasian men between the ages of 15 and 40 years. All testicular germ cell tumors develop from the same precursor lesion, intratubular germ cell neoplasia unclassified, which in turn is thought to arise from malignant transformation of a primordial germ cell or gonocyte. These tumors are characterized by extreme chemosensitivity and are considered a model for curative disease. In spite of this, a small subset of patients with metastatic disease fail to achieve a complete response with cisplatin-based chemotherapy or relapse from complete remission. Understanding the molecular biology may help the design of new therapies for those patients with a poor prognosis and could also improve the treatment of cancer in general. Current understanding of the role of genetic and epigenetic factors in the etiology of germ cell tumors and the biochemical mechanisms underlying chemotherapy sensitivity and resistance is discussed in detail in this review.
Collapse
Affiliation(s)
- Syed A Hussain
- Cancer Research UK Institute for Cancer Studies, University of Birmingham, University Hospital Birmingham NHS Foundation Trust, Edgbaston, Birmingham , UK.
| | | | | | | | | |
Collapse
|
38
|
Mai PL, Friedlander M, Tucker K, Phillips KA, Hogg D, Jewett MAS, Lohynska R, Daugaard G, Richard S, Bonaïti-Pellié C, Heidenreich A, Albers P, Bodrogi I, Geczi L, Olah E, Daly PA, Guilford P, Fosså SD, Heimdal K, Liubchenko L, Tjulandin SA, Stoll H, Weber W, Easton DF, Dudakia D, Huddart R, Stratton MR, Einhorn L, Korde L, Nathanson KL, Bishop DT, Rapley EA, Greene MH. The International Testicular Cancer Linkage Consortium: a clinicopathologic descriptive analysis of 461 familial malignant testicular germ cell tumor kindred. Urol Oncol 2009; 28:492-9. [PMID: 19162511 DOI: 10.1016/j.urolonc.2008.10.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2008] [Accepted: 10/01/2008] [Indexed: 10/21/2022]
Abstract
OBJECTIVES Familial aggregation of testicular germ cell tumor (TGCT) has been reported, but it is unclear if familial TGCT represents a unique entity with distinct clinicopathologic characteristics. Here we describe a collection of familial TGCT cases from an international consortium, in an effort to elucidate any clinical characteristics that are specific to this population. MATERIALS AND METHODS Families with >or=2 cases of TGCT enrolled at 18 of the sites participating in the International Testicular Cancer Linkage Consortium were included. We analyzed clinicopathologic characteristics of 985 cases from 461 families. RESULTS A majority (88.5%) of families had only 2 cases of TGCT. Men with seminoma (50% of cases) had an older mean age at diagnosis than nonseminoma cases (P = 0.001). Among individuals with a history of cryptorchidism, TGCT was more likely to occur in the ipsilateral testis (kappa = 0.65). Cousin pairs appeared to represent a unique group, with younger age at diagnosis and a higher prevalence of cryptorchidism than other families. CONCLUSIONS Clinicopathologic characteristics in these familial TGCT cases were similar to those generally described for nonfamilial cases. However, we observed a unique presentation of familial TGCT among cousin pairs. Additional studies are needed to further explore this observation.
Collapse
Affiliation(s)
- Phuong L Mai
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD 20852, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Chieffi P, Franco R, Portella G. Molecular and cell biology of testicular germ cell tumors. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2009; 278:277-308. [PMID: 19815181 DOI: 10.1016/s1937-6448(09)78006-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Although testicular germ cell tumors (TGCTs) are relatively uncommon, they are particularly important as they tend to affect children and young men, representing the most common tumor in male aged from 20 to 40years. TGCTs are a heterogeneous group of tumors, with specific peculiarities reflecting on epidemiologic distribution and clinic-pathological features. TGCTs show a high-cure rates in both seminomas and nonseminomas and represent the model of a curable neoplasia: sensitive serum tumor markers, accurate prognostic classification, contribute to a high effectiveness of cancer therapy. However, up to 30% of patients diagnosed with metastatic nonseminomas do not achieve a durable remission, and in metastatic teratomas cisplatin-based treatment resistance has been observed. These different prognostic and therapeutic features of TGCTs highlight the need for a better understanding of the molecular biology of TGCT, that could help to improve disease management and to tailor aggressiveness of treatment to the severity of the prognosis.
Collapse
Affiliation(s)
- Paolo Chieffi
- Dipartimento di Medicina Sperimentale, II Università di Napoli, 80138 Naples, Italy
| | | | | |
Collapse
|
40
|
Peters JA, Beckjord EB, Banda Ryan DR, Carr AG, Vadaparampil ST, Loud JT, Korde L, Greene MH. Testicular cancer and genetics knowledge among familial testicular cancer family members. J Genet Couns 2008; 17:351-64. [PMID: 18481162 PMCID: PMC3111072 DOI: 10.1007/s10897-008-9153-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2007] [Accepted: 02/18/2008] [Indexed: 11/27/2022]
Abstract
PURPOSE It was our aim to determine baseline levels of testicular cancer and genetics knowledge among members of families with Familial Testicular Cancer (FTC). METHODS This is a sub-study of an ongoing National Cancer Institute (NCI) multidisciplinary, etiologically-focused, cross-sectional study of FTC. We evaluated 258 male and female participants including testicular cancer (TC) survivors, blood relatives and spouses to assess factors associated with a Genetic Knowledge Scale (GKS) and Testicular Cancer Knowledge Scale (TCKS). RESULTS Knowledge levels were generally low, with genetic knowledge lower than TC knowledge (p < 0.01). Men with a personal TC history scored highest on TC knowledge, while gender, age and education differentially influenced knowledge levels, particularly among unaffected relatives. CONCLUSIONS Prior to identifying FTC susceptibility genes, we recommend tailoring FTC genetic education to the different informational needs of TC survivors, their spouses and relatives, in preparation for the day when clinical susceptibility testing may be available.
Collapse
Affiliation(s)
- June A Peters
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, 6120 Executive Blvd., EPS 7026, Rockville, MD 20852, USA.
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Linger R, Dudakia D, Huddart R, Tucker K, Friedlander M, Phillips KA, Hogg D, Jewett MAS, Lohynska R, Daugaard G, Richard S, Chompret A, Stoppa-Lyonnet D, Bonaïti-Pellié C, Heidenreich A, Albers P, Olah E, Geczi L, Bodrogi I, Daly PA, Guilford P, Fosså SD, Heimdal K, Tjulandin SA, Liubchenko L, Stoll H, Weber W, Einhorn L, McMaster M, Korde L, Greene MH, Nathanson KL, Cortessis V, Easton DF, Bishop DT, Stratton MR, Rapley EA. Analysis of the DND1 gene in men with sporadic and familial testicular germ cell tumors. Genes Chromosomes Cancer 2008; 47:247-52. [PMID: 18069663 PMCID: PMC3109865 DOI: 10.1002/gcc.20526] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
A base substitution in the mouse Dnd1 gene resulting in a truncated Dnd protein has been shown to be responsible for germ cell loss and the development of testicular germ cell tumors (TGCT) in the 129 strain of mice. We investigated the human orthologue of this gene in 263 patients (165 with a family history of TGCT and 98 without) and found a rare heterozygous variant, p. Glu86Ala, in a single case. This variant was not present in control chromosomes (0/4,132). Analysis of the variant in an additional 842 index TGCT cases (269 with a family history of TGCT and 573 without) did not reveal any additional instances. The variant, p. Glu86Ala, is within a known functional domain of DND1 and is highly conserved through evolution. Although the variant may be a rare polymorphism, a change at such a highly conserved residue is characteristic of a disease-causing variant. Whether it is disease-causing or not, mutations in DND1 make, at most, a very small contribution to TGCT susceptibility in adults and adolescents.
Collapse
Affiliation(s)
- Rachel Linger
- Testicular Cancer Genetics Team, Section of Cancer Genetics, Institute of Cancer Research, Sutton, Surrey, UK
| | - Darshna Dudakia
- Testicular Cancer Genetics Team, Section of Cancer Genetics, Institute of Cancer Research, Sutton, Surrey, UK
| | - Robert Huddart
- Academic Radiotherapy Unit, Institute of Cancer Research, Sutton Surrey, UK
| | - Kathy Tucker
- Department of Medical Oncology, Division of Medicine, University of New South Wales and Prince of Wales Hospital Randwick, Sydney, Australia
| | - Michael Friedlander
- Department of Medical Oncology, Division of Medicine, University of New South Wales and Prince of Wales Hospital Randwick, Sydney, Australia
| | - Kelly-Anne Phillips
- Department of Haematology and Medical Oncology, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - David Hogg
- Princess Margaret Hospital and University of Toronto, Toronto, ON, Canada
| | | | - Radka Lohynska
- University Hospital, Department of Radiotherapy and Oncology, Prague, Czech Republic
| | | | - Stéphane Richard
- Génétique Oncologique EPHE-CNRS FRE 2939 Faculté de Médecine Paris-Sud, France
- Service d’Urologie, CHU, Le Kremlin-Bicêtre, France
- Service d’Urologie, Institut Gustave Roussy, Villejuif, France
| | - Agnes Chompret
- Génétique Oncologique, Institut Gustave Roussy, Villejuif, France
| | | | | | - Axel Heidenreich
- Department of Urology, Division of Oncological Urology, University of Köln, Germany
| | - Peter Albers
- Department of Urology, Klinikum Kassel GmbH, Moenchebergstr. 41-43, D-34125 Kassel, Germany
| | - Edith Olah
- Department of Molecular Genetics, National Institute of Oncology, Budapest, Hungary
- Department of Chemotherapy, National Institute of Oncology, Budapest, Hungary
| | - Lajos Geczi
- Department of Molecular Genetics, National Institute of Oncology, Budapest, Hungary
- Department of Chemotherapy, National Institute of Oncology, Budapest, Hungary
| | - Istvan Bodrogi
- Department of Molecular Genetics, National Institute of Oncology, Budapest, Hungary
- Department of Chemotherapy, National Institute of Oncology, Budapest, Hungary
| | - Peter A. Daly
- Department of Medical Oncology, St James’s Hospital, Dublin, Ireland
| | - Parry Guilford
- Cancer Genetics Laboratory, University of Otago, Dunedin, New Zealand
| | - Sophie D. Fosså
- Department of Clinical Cancer Research, Rikshospitalet-Radiumhospitalet, Oslo, Norway
- Department of Medical Genetics, Rikshospitalet-Radiumhospitalet, Oslo, Norway
| | - Ketil Heimdal
- Department of Clinical Cancer Research, Rikshospitalet-Radiumhospitalet, Oslo, Norway
- Department of Medical Genetics, Rikshospitalet-Radiumhospitalet, Oslo, Norway
| | - Sergei A. Tjulandin
- Laboratory of Clinical Genetics, Institute of Clinical Oncology, N.N. Blokhin Russian Cancer Research Center, Moscow, Russian Federation
| | - Ludmila Liubchenko
- Laboratory of Clinical Genetics, Institute of Clinical Oncology, N.N. Blokhin Russian Cancer Research Center, Moscow, Russian Federation
| | - Hans Stoll
- Medical Oncology, University Hospital, Basel, Switzerland
| | - Walter Weber
- Medical Oncology, University Hospital, Basel, Switzerland
| | - Lawrence Einhorn
- Department of Medicine, Indiana University School of Medicine, Indianapolis
| | - Mary McMaster
- Clinical Genetics Branch, Division of Cancer Epidemiology & Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland
| | - Larissa Korde
- Clinical Genetics Branch, Division of Cancer Epidemiology & Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland
| | - Mark H. Greene
- Clinical Genetics Branch, Division of Cancer Epidemiology & Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland
| | - Katherine L. Nathanson
- Departments of Medicine, Abramson Cancer Center, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Victoria Cortessis
- Department of Preventive Medicine, Keck School of Medicine, USC/Norris Comprehensive Cancer Center, Los Angeles, California
| | - Douglas F. Easton
- Cancer Research U.K. Genetic Epidemiology Unit, Strangeways Research Laboratory, Cambridge, UK
| | - D. Timothy Bishop
- Section of Epidemiology & Biostatistics, Leeds Institute of Molecular Medicine, St. James’s University Hospital, Leeds, UK
| | - Michael R. Stratton
- Testicular Cancer Genetics Team, Section of Cancer Genetics, Institute of Cancer Research, Sutton, Surrey, UK
| | - Elizabeth A. Rapley
- Testicular Cancer Genetics Team, Section of Cancer Genetics, Institute of Cancer Research, Sutton, Surrey, UK
| |
Collapse
|
42
|
Mueller CM, Korde L, Katki HA, Rosenberg PS, Peters JA, Greene MH. Constitutional cytogenetic analysis in men with hereditary testicular germ cell tumor: no evidence of disease-related abnormalities. Cancer Epidemiol Biomarkers Prev 2007; 16:2791-4. [PMID: 18086791 PMCID: PMC3125977 DOI: 10.1158/1055-9965.epi-07-0521] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Affiliation(s)
- Christine M Mueller
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Department of Health and Human Services, 6120 Executive Boulevard, EPS 7101, Rockville, MD 20852-7231, USA.
| | | | | | | | | | | |
Collapse
|
43
|
Abstract
Testis cancer is an increasing problem, especially in northern European male populations. However, survival has improved dramatically over one generation. Environmental factors may have a role in the aetiology with high oestrogen concentrations implicated. Testis cancer is subdivided between seminoma and non-seminoma. At presentation, a testicular lump is the most common finding and radical inguinal orchidectomy is recommended for most. Further multidisciplinary management is determined by histological subtype and stage and involves chemotherapy, radiotherapy and surgery, with many patients only undergoing surveillance. There is increasing emphasis on reducing toxicity of treatments in long term survivors. Treatment refractory testis cancer remains a significant challenge.
Collapse
Affiliation(s)
- Omar Khan
- Cancer Research UK, Medical Oncology Unit, Churchill Hospital, Oxford, UK.
| | | |
Collapse
|
44
|
Richiardi L, Pettersson A, Akre O. Genetic and environmental risk factors for testicular cancer. ACTA ACUST UNITED AC 2007; 30:230-40; discussion 240-1. [PMID: 17488341 DOI: 10.1111/j.1365-2605.2007.00760.x] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Germ-cell testicular cancer has a well-characterized descriptive epidemiology, whereas the aetiology remains largely unknown. It is believed that exposures acting prenatally are instrumental to germ-cell cancer development, although no specific exposure has been identified. Several epidemiological studies have investigated a number of indicators of prenatal exposures, such as birth order, gestational duration, birth weight, maternal age and nausea during pregnancy, but results are inconsistent. This paper briefly reviews the current support for genetic and environmental factors in testicular cancer aetiology. In particular, we have summarized the evidence suggesting a strong role of inherited susceptibility, which is probably carried by the effect of several unknown moderate-risk genes. We have illustrated inconsistencies in the previous studies on prenatal factors by estimating the heterogeneity and pooled odds ratios among twelve studies investigating the association between low birth weight and testicular cancer. We have discussed the possibility that puberty is another time window during which environmental factors may increase the risk of testicular cancer. Finally, we have reviewed the results from studies on cryptorchidism and impaired fertility in relation to risk for testicular cancer. In conclusion, we propose that future aetiological studies on testicular cancer should take postnatal exposures acting during puberty into account and, whenever possible, investigate both main effects and interactions among prenatal factors, genetic factors and postnatal factors.
Collapse
Affiliation(s)
- Lorenzo Richiardi
- Cancer Epidemiology Unit, CeRMS and CPO-Piemonte, University of Turin, Turin, Italy.
| | | | | |
Collapse
|
45
|
Purdue MP, Sakoda LC, Graubard BI, Welch R, Chanock SJ, Sesterhenn IA, Rubertone MV, Erickson RL, McGlynn KA. A case-control investigation of immune function gene polymorphisms and risk of testicular germ cell tumors. Cancer Epidemiol Biomarkers Prev 2007; 16:77-83. [PMID: 17220333 DOI: 10.1158/1055-9965.epi-06-0573] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
There is reason to suspect that testicular germ cell tumor (TGCT) development may be influenced by cytokines, secreted proteins that modulate tumor immune surveillance activity as well as a variety of processes in the testis. To address this hypothesis, we conducted a case-control analysis (508 cases, 608 controls) of 32 putatively functional single-nucleotide polymorphisms (SNP) in 16 immune function genes among non-Hispanic Caucasian participants in the U.S. Servicemen's Testicular Tumor Environmental and Endocrine Determinants Study. The TGFB1 Ex5-73C>T variant was positively associated with TGCT (CT/TT versus CC: odds ratio, 1.73; 95% confidence interval, 1.01-2.95; P(trend) = 0.05); additionally, haplotypes of the assessed TGFB1 SNPs (-509C>T, 327C>T, Ex1-282C>G, and Ex5-73C>T) differed in frequency between cases and controls (all TGCT, P 0.07; seminoma, P 0.04; nonseminoma, P 0.11). We also observed excess frequencies among TGCT cases versus controls of LTA 252G (P(trend) = 0.08) and of the TNF variants -1042C (P(trend) = 0.06), -1036T (P(trend) = 0.07), and -238G (P(trend) = 0.09). Analyses of haplotypes for LTA-TNF SNPs (LTA -91C>A, LTA 252A>G, TNF -863C>A, TNF -857C>T, TNF -308G>A, and -238G>A) were similarly suggestive of an association with TGCT (P = 0.06) and nonseminoma (P = 0.04), but not seminoma (P = 0.21). Polymorphisms in other genes were found to be associated only with seminoma (IL2) or nonseminoma (IFNGR2 and IL10). However, none of the associations remained noteworthy after applying the false discovery rate method to control for multiple testing. In conclusion, our findings suggest that polymorphisms in TGFB1 and LTA/TNF, and possibly other immune function genes, may influence susceptibility to TGCT.
Collapse
Affiliation(s)
- Mark P Purdue
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Department of Health and Human Services, EPS-8009, 6120 Executive Boulevard, Rockville, MD 20892-7234, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Mai PL, Korde L, Kramer J, Peters J, Mueller CM, Pfeiffer S, Stratakis CA, Pinto PA, Bratslavsky G, Merino M, Choyke P, Linehan WM, Greene MH. A possible new syndrome with growth-hormone secreting pituitary adenoma, colonic polyposis, lipomatosis, lentigines and renal carcinoma in association with familial testicular germ cell malignancy: A case report. J Med Case Rep 2007; 1:9. [PMID: 17411461 PMCID: PMC1847830 DOI: 10.1186/1752-1947-1-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2007] [Accepted: 03/28/2007] [Indexed: 12/23/2022] Open
Abstract
Background Germ-cell testicular cancer has not been definitively linked to any known hereditary cancer susceptibility disorder. Familial testicular cancer in the presence of other findings in affected and unaffected family members might indicate a previously-unidentified hereditary cancer syndrome. Case presentation The patient was diagnosed with a left testicular seminoma at age 28, and treated with left orchiectomy followed by adjuvant cobalt radiation. His family history is significant for testicular seminoma in his son, bladder cancer in his sister, and lipomatosis in his father. His evaluation as part of an etiologic study of familial testicular cancer revealed multiple colon polyps (adenomatous, hyperplastic, and hamartomatous) first found in his 50 s, multiple lipomas, multiple hyperpigmented skin lesions, left kidney cancer diagnosed at age 64, and a growth-hormone producing pituitary adenoma with associated acromegaly diagnosed at age 64. The patient underwent genetic testing for Cowden syndrome (PTEN gene), Carney complex (PRKAR1A gene), and multiple endocrine neoplasia syndrome type 1 (MEN1 gene); no deleterious mutations were identified. Discussion The constellation of benign and malignant neoplasms in the context of this patient's familial testicular cancer raised the possibility that these might be manifestations of a known hereditary susceptibility cancer syndrome; however, genetic testing for the three syndromes that were most likely to explain these findings did not show any mutation. Alternatively, this family's phenotype might represent a novel neoplasm susceptibility disorder. This possibility cannot be evaluated definitively on the basis of a single case report; additional observations and studies are necessary to investigate this hypothesis further.
Collapse
Affiliation(s)
- Phuong L Mai
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Larissa Korde
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Joan Kramer
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - June Peters
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Christine M Mueller
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Constantine A Stratakis
- Section on Endocrinology & Genetics, Developmental Endocrinology Branch, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Peter A Pinto
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Gennady Bratslavsky
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Maria Merino
- Division of Laboratory and Pathology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Peter Choyke
- Molecular Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - W Marston Linehan
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Mark H Greene
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
47
|
Peters JA, Vadaparampil ST, Kramer J, Moser RP, Court LJP, Loud J, Greene MH. Familial testicular cancer: interest in genetic testing among high-risk family members. Genet Med 2007; 8:760-70. [PMID: 17172939 DOI: 10.1097/01.gim.0000250506.15979.0c] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
PURPOSE This study is part of an ongoing National Cancer Institute multidisciplinary, etiologically-focused, cross-sectional study of Familial Testicular Cancer (FTC). The current report targets interest in clinical genetic testing for susceptibility to FTC. METHODS Demographics, knowledge, health beliefs, and psychological and social factors were evaluated as covariates related to interest in genetic testing. RESULTS The majority (66%) of 229 participants (64 affected men, 66 unaffected men, and 99 women) from 47 multiple-case FTC families expressed interest in having a genetic test within 6 months, should such a test become available. Interest was similar among the three subgroups mentioned above. Worries about insurance discrimination based on genetic test results were associated with a significantly lower interest in testing. Alternatively, participants were more likely to be interested in genetic testing if they were younger and had higher levels of family support, a physician's recommendation supporting testing, cancer distress, and a need for information to inform the health care of their children. CONCLUSIONS This study reveals social and relationship factors that FTC survivors and their relatives considered important when contemplating the use of new genetic technologies. This is the first study describing hypothetical interest in genetic testing for familial testicular cancer.
Collapse
Affiliation(s)
- June A Peters
- Clinical Genetics Branch (CGB), Division of Cancer Epidemiology and Genetics (DCEG), National Cancer Institute (NCI), National Institutes of Health (NIH), DHHS, Rockville, Maryland 20852, USA.
| | | | | | | | | | | | | |
Collapse
|
48
|
Westbrook VA, Schoppee PD, Vanage GR, Klotz KL, Diekman AB, Flickinger CJ, Coppola MA, Herr JC. Hominoid-specific SPANXA/D genes demonstrate differential expression in individuals and protein localization to a distinct nuclear envelope domain during spermatid morphogenesis. ACTA ACUST UNITED AC 2006; 12:703-16. [PMID: 17012309 DOI: 10.1093/molehr/gal079] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Human sperm protein associated with the nucleus on the X chromosome consists of a five-member gene family (SPANXA1, SPANXA2, SPANXB, SPANXC and SPANXD) clustered at Xq27.1. Evolved from an ancestral SPANX-N gene family (at Xq27 and Xp11) present in all primates as well as in rats and mice, the SPANXA/D family is present only in humans, bonobos, chimpanzees and gorillas. Among hominoid-specific genes, the SPANXA/D gene family is considered to be undergoing rapid positive selection in its coding region. In this study, RT-PCR of human testis mRNA from individuals showed that, although all SPANXA/D genes are expressed in humans, differences are evident. In particular, SPANXC is expressed only in a subset of men. The SPANXa/d protein localized to the nuclear envelope of round, condensing and elongating spermatids, specifically to regions that do not underlie the developing acrosome. During spermiogenesis, the SPANXa/d-positive domain migrated into the base of the head as the redundant nuclear envelope that protrudes into the residual cytoplasm. Post-testicular modification of the SPANXa/d proteins was noted, as were PEST (proline, glutamic acid, serine, and threonine rich regions) domains. It is concluded that the duplication of the SPANX-N gene family that occurred 6-11 MYA resulted in a new gene family, SPANXA/D, that plays a role during spermiogenesis. The SPANXa/d gene products are among the few examples of X-linked nuclear proteins expressed following meiosis. Their localization to non-acrosomal domains of the nuclear envelope adjacent to regions of euchromatin and their redistribution to the redundant nuclear envelope during spermiogenesis provide a biomarker for the redundant nuclear envelope of spermatids and spermatozoa.
Collapse
Affiliation(s)
- V A Westbrook
- Department of Cell Biology, Center for Research in Contraceptive and Reproductive Health, University of Virginia, Charlottesville, VA 22908, USA
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Tuinman MA, Hoekstra HJ, Sleijfer DT, Fleer J, Vidrine DJ, Gritz ER, Hoekstra-Weebers JEHM. Testicular cancer: a longitudinal pilot study on stress response symptoms and quality of life in couples before and after chemotherapy. Support Care Cancer 2006; 15:279-86. [PMID: 16944218 PMCID: PMC2092408 DOI: 10.1007/s00520-006-0119-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2006] [Accepted: 06/22/2006] [Indexed: 12/22/2022]
Abstract
Goals of work The current study was designed to longitudinally examine stress response symptoms (SRS) and quality of life (QoL) in couples confronted with disseminated testicular cancer. The objectives were to examine couples’ patterns of adjustment over time and possible differences in adjustment between the patient and his partner. Materials and methods Couples completed the Impact of Event Scale and the QoL subscales physical functioning, social functioning, and mental health of the RAND-36 before chemotherapy (T1), after completion of chemotherapy (T2), and 1 year later (T3). Results Before chemotherapy 26% of the patients and 50% of partners reported clinically elevated levels of SRS. Patients reported lower physical and social functioning at T2 compared to T1 and T3. Partners reported an improvement in social functioning over the year and no changes in physical functioning or mental health. No relationships between patients and partners’ functioning were found. One year after diagnosis, QoL of patients and partners was similar to that of reference groups, and patients even reported better physical functioning than the reference group. SRS of patients and partners were negatively related at T1, and patients and partners’ social functioning were positively related at T2. Conclusions According to stress response levels, the period before the start of chemotherapy was most stressful for couples. Adjustment patterns differ between testicular cancer patients and their partners with patients reporting lowered QoL after completion of chemotherapy. QoL of couples returned to normal levels 1 year after diagnosis. The effect of disseminated testicular cancer on the QoL of patients and their partners seems to be temporary. A minority may need clinical attention for severe SRS.
Collapse
Affiliation(s)
- Marrit A. Tuinman
- Department of Surgical Oncology, University Medical Center Groningen, University of Groningen, P.O. Box 30001, Groningen, 9700 RB The Netherlands
| | - Harald J. Hoekstra
- Department of Surgical Oncology, University Medical Center Groningen, University of Groningen, P.O. Box 30001, Groningen, 9700 RB The Netherlands
| | - Dirk Th. Sleijfer
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Joke Fleer
- Department of Surgical Oncology, University Medical Center Groningen, University of Groningen, P.O. Box 30001, Groningen, 9700 RB The Netherlands
| | - Damon J. Vidrine
- Department of Behavioral Science, University of Texas M. D. Anderson Cancer Center, Houston, USA
| | - Ellen R. Gritz
- Department of Behavioral Science, University of Texas M. D. Anderson Cancer Center, Houston, USA
| | - Josette E. H. M. Hoekstra-Weebers
- Department of Psychosocial Services, University Medical Center Groningen, University of Groningen, P.O. Box 30.001, Groningen, 9700 RB The Netherlands
- Comprehensive Cancer Center North-Netherlands, P.O. Box 330, Groningen, 9700 AH The Netherlands
| |
Collapse
|
50
|
Lutke Holzik MF, Hoekstra HJ, Sijmons RH, Sonneveld DJA, van der Steege G, Sleijfer DT, Nolte IM. Re-analysis of the Xq27–Xq28 region suggests a weak association of an X-linked gene with sporadic testicular germ cell tumour without cryptorchidism. Eur J Cancer 2006; 42:1869-74. [PMID: 16797968 DOI: 10.1016/j.ejca.2006.03.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2005] [Revised: 03/08/2006] [Accepted: 03/10/2006] [Indexed: 01/10/2023]
Abstract
BACKGROUND A testicular germ cell tumour (TGCT) predisposing gene has been mapped to the Xq27 region on the X chromosome. These linkage findings remain to be confirmed by other studies. METHODS In 276 patients and 169 unaffected first-degree male relatives, 12 microsatellite markers covering the candidate region were genotyped and used to study possible association of TGCT with Xq27. RESULTS In contrast to previously reported linkage of familial TGCT and cryptorchidism with Xq27, we observed an association between the subset of TGCT cases without a family history of TGCT or cryptorchism and marker DXS1193 (p=0.014). Carriers of minor alleles were at increased risk (odds ratio (OR) 4.7, confidence interval (CI) 1.1-19.6) CONCLUSION We found an association on Xq27 in a subset of TGCT cases, which suggests the presence of an X-linked gene that slightly or moderately increases risk to develop sporadic TGCT but not cryptorchidism.
Collapse
Affiliation(s)
- M F Lutke Holzik
- Department of Surgical Oncology, University Medical Centre Groningen, P.O. Box 30.001, 9700 RB Groningen, The Netherlands
| | | | | | | | | | | | | |
Collapse
|