Skip to main content
Top
Published in: Cellular Oncology 2/2014

01-04-2014 | Review

The chemosensitivity of testicular germ cell tumors

Author: Ioannis A. Voutsadakis

Published in: Cellular Oncology | Issue 2/2014

Login to get access

Abstract

Although rare cancers overall, testicular germ cell tumors (TGCTs) are the most common type of cancer in young males below 40 years of age. Both subtypes of TGCTs, i.e., seminomas and non-seminomas, are highly curable and the majority of even metastatic patients may expect to be cured. These high cure rates are not due to the indolent nature of these cancers, but rather to their sensitivity to chemotherapy (and for seminomas to radiotherapy). The delineation of the cause of chemosensitivity at the molecular level is of paramount importance, because it may provide insights into the minority of TGCTs that are chemo-resistant and, thereby, provide opportunities for specific therapeutic interventions aimed at reverting them to chemosensitivity. In addition, delineation of the molecular basis of TGCT chemo-sensitivity may be informative for the cause of chemo-resistance of other more common types of cancer and, thus, may create new therapeutic leads. p53, a frequently mutated tumor suppressor in cancers in general, is not mutated in TGCTs, a fact that has implications for their chemo-sensitivity. Oct4, an embryonic transcription factor, is uniformly expressed in the seminoma and embryonic carcinoma components of non-seminomas, and its interplay with p53 may be important in the chemotherapy response of these tumors. This interplay, together with other features of TGCTs such as the gain of genetic material from the short arm of chromosome 12 and the association with disorders of testicular development, will be discussed in this paper and integrated in a unifying hypothesis that may explain their chemo-sensitivity.
Literature
1.
go back to reference A. Di Pietro, E.G.E. de Vries, J.A. Gietema, D.C.J. Spierings, S. de Jong, Testicular germ cell tumours: the paradigm of chemo-sensitive solid tumours. Int. J. Biochem. Cell Biol. 37, 2437–2456 (2005)PubMed A. Di Pietro, E.G.E. de Vries, J.A. Gietema, D.C.J. Spierings, S. de Jong, Testicular germ cell tumours: the paradigm of chemo-sensitive solid tumours. Int. J. Biochem. Cell Biol. 37, 2437–2456 (2005)PubMed
2.
go back to reference L.H.J. Looijenga, A.J.M. Gillis, H. Stoop, K. Biermann, J.W. Oosterhuis, Dissecting the molecular pathways of (testicular) germ cell tumour pathogenesis; from initiation to treatment-resistance. Int. J. Androl. 34, e234–e251 (2011)PubMed L.H.J. Looijenga, A.J.M. Gillis, H. Stoop, K. Biermann, J.W. Oosterhuis, Dissecting the molecular pathways of (testicular) germ cell tumour pathogenesis; from initiation to treatment-resistance. Int. J. Androl. 34, e234–e251 (2011)PubMed
3.
go back to reference International Germ Cell Cancer Collaborative Group, International Germ Cell Consensus Classification: a prognostic factor-based staging system for metastatic germ cell cancers. J. Clin. Oncol. 15, 594–603 (1997) International Germ Cell Cancer Collaborative Group, International Germ Cell Consensus Classification: a prognostic factor-based staging system for metastatic germ cell cancers. J. Clin. Oncol. 15, 594–603 (1997)
4.
go back to reference F. Mayer, F. Honecker, L.H.J. Looijenga, C. Bokemeyer, Towards an understanding of the biological basis of response to cisplatin-based chemotherapy in germ-cell tumors. Ann. Oncol. 14, 825–832 (2003)PubMed F. Mayer, F. Honecker, L.H.J. Looijenga, C. Bokemeyer, Towards an understanding of the biological basis of response to cisplatin-based chemotherapy in germ-cell tumors. Ann. Oncol. 14, 825–832 (2003)PubMed
5.
go back to reference H.Q. Peng, D. Hogg, D. Malkin, D. Bailey, B.L. Gallie, M. Balbul, M. Jewett, J. Buchanan, P.E. Goss, Mutations of the p53 gene do not occur in testis cancer. Cancer Res. 53, 3574–3578 (1993)PubMed H.Q. Peng, D. Hogg, D. Malkin, D. Bailey, B.L. Gallie, M. Balbul, M. Jewett, J. Buchanan, P.E. Goss, Mutations of the p53 gene do not occur in testis cancer. Cancer Res. 53, 3574–3578 (1993)PubMed
6.
go back to reference L. Guillou, A. Estreicher, P. Chaubert, J. Hurlimann, A.M. Kurt, G. Metthez, R. Iggo, A.C. Gray, P. Jichlinski, H.-J. Leisinger, J. Benhattar, Germ cell tumors of the testis overexpress wild-type p53. Am. J. Pathol. 149, 1221–1228 (1996)PubMedCentralPubMed L. Guillou, A. Estreicher, P. Chaubert, J. Hurlimann, A.M. Kurt, G. Metthez, R. Iggo, A.C. Gray, P. Jichlinski, H.-J. Leisinger, J. Benhattar, Germ cell tumors of the testis overexpress wild-type p53. Am. J. Pathol. 149, 1221–1228 (1996)PubMedCentralPubMed
7.
go back to reference J.S. Kerley-Hamilton, A.M. Pike, N. Li, J. DiRenzo, M.J. Spinella, A p53-dominant transcriptional response to cisplatin in testicular germ cell tumor-derived human embryonal carcinoma. Oncogene 24, 6090–6100 (2005)PubMed J.S. Kerley-Hamilton, A.M. Pike, N. Li, J. DiRenzo, M.J. Spinella, A p53-dominant transcriptional response to cisplatin in testicular germ cell tumor-derived human embryonal carcinoma. Oncogene 24, 6090–6100 (2005)PubMed
9.
go back to reference C.R. Leemans, B.J.M. Braakhuis, R.H. Brakenhoff, The molecular biology of head and neck cancer. Nat. Rev. Cancer 11, 9–22 (2011)PubMed C.R. Leemans, B.J.M. Braakhuis, R.H. Brakenhoff, The molecular biology of head and neck cancer. Nat. Rev. Cancer 11, 9–22 (2011)PubMed
10.
go back to reference I.A. Voutsadakis, Ubiquitination and the ubiquitin-proteasome system in the pathogenesis and treatment of squamous head and neck carcinoma. Anticancer Res. 33, 3527–3541 (2013)PubMed I.A. Voutsadakis, Ubiquitination and the ubiquitin-proteasome system in the pathogenesis and treatment of squamous head and neck carcinoma. Anticancer Res. 33, 3527–3541 (2013)PubMed
11.
go back to reference A.V. Pinho, I. Rooman, F.X. Real, p53-dependent regulation of growth, epithelial-mesenchymal transition and stemness in normal pancreatic epithelial cells. Cell Cycle 10, 1312–1321 (2011)PubMed A.V. Pinho, I. Rooman, F.X. Real, p53-dependent regulation of growth, epithelial-mesenchymal transition and stemness in normal pancreatic epithelial cells. Cell Cycle 10, 1312–1321 (2011)PubMed
13.
go back to reference I.N. Colaluca, D. Tosoni, P. Nuciforo, F. Senic-Matuglia, V. Galimberti, G. Viale, S. Pece, P.P. Di Fiore, NUMB controls p53 tumour suppressor activity. Nature 451, 76–80 (2008)PubMed I.N. Colaluca, D. Tosoni, P. Nuciforo, F. Senic-Matuglia, V. Galimberti, G. Viale, S. Pece, P.P. Di Fiore, NUMB controls p53 tumour suppressor activity. Nature 451, 76–80 (2008)PubMed
14.
15.
go back to reference L.A. Carvajal, J.J. Manfredi, Another fork in the road- life or death decisions by the tumour suppressor p53. EMBO Rep. 14, 414–421 (2013)PubMed L.A. Carvajal, J.J. Manfredi, Another fork in the road- life or death decisions by the tumour suppressor p53. EMBO Rep. 14, 414–421 (2013)PubMed
16.
go back to reference H. Solomon, S. Madar, V. Rotter, Mutant p53 gain of function is interwoven into the hallmarks of cancer. J. Pathol. 225, 475–478 (2011)PubMed H. Solomon, S. Madar, V. Rotter, Mutant p53 gain of function is interwoven into the hallmarks of cancer. J. Pathol. 225, 475–478 (2011)PubMed
17.
go back to reference D. Hanahan, R.A. Weinberg, Hallmarks of cancer: the next generation. Cell 144, 646–674 (2011)PubMed D. Hanahan, R.A. Weinberg, Hallmarks of cancer: the next generation. Cell 144, 646–674 (2011)PubMed
18.
go back to reference P. Campomenosi, P. Monti, A. Aprile, A. Abbondandolo, T. Frebourg, B. Gold, T. Crook, A. Inga, M.A. Resnick, R. Iggo, G. Fronza, p53 mutants can often transactivate promoters containing a p21 but not Bax or PIG3 responsive elements. Oncogene 20, 3573–3579 (2001)PubMed P. Campomenosi, P. Monti, A. Aprile, A. Abbondandolo, T. Frebourg, B. Gold, T. Crook, A. Inga, M.A. Resnick, R. Iggo, G. Fronza, p53 mutants can often transactivate promoters containing a p21 but not Bax or PIG3 responsive elements. Oncogene 20, 3573–3579 (2001)PubMed
19.
go back to reference S. Rowan, R.L. Ludwig, Y. Haupt, S. Bates, X. Lu, M. Oren, K.H. Vousden, Specific loss of apoptotic but not cell-cycle arrest function in a human tumor derived p53 mutant. EMBO J. 15, 827–838 (1996)PubMedCentralPubMed S. Rowan, R.L. Ludwig, Y. Haupt, S. Bates, X. Lu, M. Oren, K.H. Vousden, Specific loss of apoptotic but not cell-cycle arrest function in a human tumor derived p53 mutant. EMBO J. 15, 827–838 (1996)PubMedCentralPubMed
20.
go back to reference G. Blandino, W. Deppert, P. Hainaut, A. Levine, G. Lozano, M. Olivier, V. Rotter, K. Wiman, M. Oren, Mutant p53 protein, master regulator of human malignancies: a report of the fifth Mutant p53 Workshop. Cell Death Differ. 19, 180–183 (2012)PubMedCentralPubMed G. Blandino, W. Deppert, P. Hainaut, A. Levine, G. Lozano, M. Olivier, V. Rotter, K. Wiman, M. Oren, Mutant p53 protein, master regulator of human malignancies: a report of the fifth Mutant p53 Workshop. Cell Death Differ. 19, 180–183 (2012)PubMedCentralPubMed
21.
go back to reference J.G. Jackson, S.M. Post, G. Lozano, Regulation of tissue- and stimulus-specific cell fate decisions by p53 in vivo. J. Pathol. 223, 127–136 (2011)PubMed J.G. Jackson, S.M. Post, G. Lozano, Regulation of tissue- and stimulus-specific cell fate decisions by p53 in vivo. J. Pathol. 223, 127–136 (2011)PubMed
22.
go back to reference K. Bensaad, A. Tsuruta, M.A. Selak, M.N. Vidal, K. Nakano, R. Bartrons, E. Gottlieb, K.H. Vousden, TIGAR, a p53-inducible regulator of glycolysis and apoptosis. Cell 126, 107–120 (2006)PubMed K. Bensaad, A. Tsuruta, M.A. Selak, M.N. Vidal, K. Nakano, R. Bartrons, E. Gottlieb, K.H. Vousden, TIGAR, a p53-inducible regulator of glycolysis and apoptosis. Cell 126, 107–120 (2006)PubMed
23.
go back to reference F. Mayer, H. Stoop, G.L. Scheffer, R. Scheper, J.W. Oosterhuis, L.H.J. Looijenga, C. Bokemeyer, Molecular determinants of treatment response in human germ cell tumors. Clin. Cancer Res. 9, 767–773 (2003)PubMed F. Mayer, H. Stoop, G.L. Scheffer, R. Scheper, J.W. Oosterhuis, L.H.J. Looijenga, C. Bokemeyer, Molecular determinants of treatment response in human germ cell tumors. Clin. Cancer Res. 9, 767–773 (2003)PubMed
24.
go back to reference D.C.J. Spierings, E.G.E. de Vries, A.J. Stel, N. te Rietstap, E. Vellenga, S. de Jong, Low p21Waf1/Cip1 protein level sensitizes testicular germ cell tumor cells to Fas-mediated apoptosis. Oncogene 23, 4862–4872 (2004)PubMed D.C.J. Spierings, E.G.E. de Vries, A.J. Stel, N. te Rietstap, E. Vellenga, S. de Jong, Low p21Waf1/Cip1 protein level sensitizes testicular germ cell tumor cells to Fas-mediated apoptosis. Oncogene 23, 4862–4872 (2004)PubMed
25.
go back to reference A. Di Pietro, R. Koster, W. Boersma-van Eck, W.A. Dam, N.H. Mulder, J.A. Gietema, E.G.E. de Vries, S. de Jong, Pro- and anti-apoptotic effects of p53 in cisplatin-treated human testicular cancer are cell context-dependent. Cell Cycle 11, 4552–4562 (2012)PubMedCentralPubMed A. Di Pietro, R. Koster, W. Boersma-van Eck, W.A. Dam, N.H. Mulder, J.A. Gietema, E.G.E. de Vries, S. de Jong, Pro- and anti-apoptotic effects of p53 in cisplatin-treated human testicular cancer are cell context-dependent. Cell Cycle 11, 4552–4562 (2012)PubMedCentralPubMed
26.
go back to reference M. Gutekunst, M. Oren, A. Weilbacher, M.A. Dengler, C. Markwardt, J. Thomale, W.E. Aulitzky, H. van der Kuip, p53 hypersensitivity is the predominant mechanism of the unique responsiveness of testicular germ cell tumor (TGCT) cells to cisplatin. PLoS ONE 6, e19198 (2011)PubMedCentralPubMed M. Gutekunst, M. Oren, A. Weilbacher, M.A. Dengler, C. Markwardt, J. Thomale, W.E. Aulitzky, H. van der Kuip, p53 hypersensitivity is the predominant mechanism of the unique responsiveness of testicular germ cell tumor (TGCT) cells to cisplatin. PLoS ONE 6, e19198 (2011)PubMedCentralPubMed
27.
go back to reference C. Dai, W. Gu, p53 post-translational modification: deregulated in tumorigenesis. Trends Mol. Med. 16, 528–535 (2010)PubMedCentralPubMed C. Dai, W. Gu, p53 post-translational modification: deregulated in tumorigenesis. Trends Mol. Med. 16, 528–535 (2010)PubMedCentralPubMed
28.
29.
go back to reference J.M. Stommel, G.M. Wahl, A new twist in the feedback loop. Stress-activated MDM2 destabilization is required for p53 activation. Cell Cycle 4, 413–417 (2005) J.M. Stommel, G.M. Wahl, A new twist in the feedback loop. Stress-activated MDM2 destabilization is required for p53 activation. Cell Cycle 4, 413–417 (2005)
30.
go back to reference B. Li, Q. Cheng, Z. Li, J. Chen, p53 inactivation by MDM2 and MDMX negative feedback loops in testicular germ cell tumors. Cell Cycle 9, 1411–1420 (2010)PubMedCentralPubMed B. Li, Q. Cheng, Z. Li, J. Chen, p53 inactivation by MDM2 and MDMX negative feedback loops in testicular germ cell tumors. Cell Cycle 9, 1411–1420 (2010)PubMedCentralPubMed
31.
32.
go back to reference J. Yuan, K. Luo, L. Zhang, J.C. Cheville, Z. Lou, USP10 regulates p53 localization and stability by deubiquitinating p53. Cell 140, 384–396 (2010)PubMedCentralPubMed J. Yuan, K. Luo, L. Zhang, J.C. Cheville, Z. Lou, USP10 regulates p53 localization and stability by deubiquitinating p53. Cell 140, 384–396 (2010)PubMedCentralPubMed
33.
go back to reference M.B. Kastan, G.P. Zambetti, Parc-ing p53 in the cytoplasm. Cell 112, 1–5 (2003)PubMed M.B. Kastan, G.P. Zambetti, Parc-ing p53 in the cytoplasm. Cell 112, 1–5 (2003)PubMed
34.
go back to reference A.Y. Nikolaev, M. Li, N. Puskas, J. Qin, W. Gu, Parc: a cytoplasmic anchor for p53. Cell 112, 29–40 (2003)PubMed A.Y. Nikolaev, M. Li, N. Puskas, J. Qin, W. Gu, Parc: a cytoplasmic anchor for p53. Cell 112, 29–40 (2003)PubMed
35.
go back to reference R.P. Leng, Y. Lin, W. Ma, H. Wu, B. Lemmers, S. Chung, J.M. Parant, G. Lozano, R. Hakem, S. Benchimol, Pirh2, a p53-induced ubiquitin-protein ligase, promotes p53 degradation. Cell 112, 779–791 (2003)PubMed R.P. Leng, Y. Lin, W. Ma, H. Wu, B. Lemmers, S. Chung, J.M. Parant, G. Lozano, R. Hakem, S. Benchimol, Pirh2, a p53-induced ubiquitin-protein ligase, promotes p53 degradation. Cell 112, 779–791 (2003)PubMed
36.
go back to reference D. Chen, N. Kon, M. Li, W. Zhang, J. Qin, W. Gu, ARF-BP1/Mule is a critical mediator of the ARF tumor suppressor. Cell 121, 1071–1083 (2005)PubMed D. Chen, N. Kon, M. Li, W. Zhang, J. Qin, W. Gu, ARF-BP1/Mule is a critical mediator of the ARF tumor suppressor. Cell 121, 1071–1083 (2005)PubMed
37.
go back to reference D. Dornan, I. Wertz, H. Shimizu, D. Arnott, G.D. Frantz, P. Dowd, K. O’Rourke, H. Koeppen, V.M. Dixit, The ubiquitin ligase COP is a critical negative regulator of p53. Nature 429, 86–92 (2004)PubMed D. Dornan, I. Wertz, H. Shimizu, D. Arnott, G.D. Frantz, P. Dowd, K. O’Rourke, H. Koeppen, V.M. Dixit, The ubiquitin ligase COP is a critical negative regulator of p53. Nature 429, 86–92 (2004)PubMed
38.
go back to reference S.V. Khoronenkova, G.L. Dianov, USP7S-dependent inactivation of Mule regulates DNA damage signalling and repair. Nucleic Acids Res. 41, 1750–1756 (2013)PubMedCentralPubMed S.V. Khoronenkova, G.L. Dianov, USP7S-dependent inactivation of Mule regulates DNA damage signalling and repair. Nucleic Acids Res. 41, 1750–1756 (2013)PubMedCentralPubMed
39.
go back to reference Q. Zhong, W. Gao, F. Du, X. Wang, Mule/ARF-BP1, a BH3-only E3 ubiquitin ligase, catalyzes the polyubiquitination of Mcl-1 and regulates apoptosis. Cell 121, 1085–1095 (2005)PubMed Q. Zhong, W. Gao, F. Du, X. Wang, Mule/ARF-BP1, a BH3-only E3 ubiquitin ligase, catalyzes the polyubiquitination of Mcl-1 and regulates apoptosis. Cell 121, 1085–1095 (2005)PubMed
40.
go back to reference A. Shmueli, M. Oren, Life, death, and ubiquitin: taming the Mule. Cell 121, 963–965 (2005)PubMed A. Shmueli, M. Oren, Life, death, and ubiquitin: taming the Mule. Cell 121, 963–965 (2005)PubMed
41.
go back to reference N. Taira, K. Nihira, T. Yamaguchi, Y. Miki, K. Yoshida, DYRK2 is targeted to the nucleus and controls p53 via Ser46 phosphorylation in the apoptotic response to DNA damage. Mol. Cell 25, 725–738 (2007)PubMed N. Taira, K. Nihira, T. Yamaguchi, Y. Miki, K. Yoshida, DYRK2 is targeted to the nucleus and controls p53 via Ser46 phosphorylation in the apoptotic response to DNA damage. Mol. Cell 25, 725–738 (2007)PubMed
42.
go back to reference C. Rinaldo, A. Prodosmo, F. Mancini, S. Iacovelli, A. Sacchi, F. Moretti, S. Soddu, MDM2-regulated degradation of HIPK2 prevents p53Ser46 phosphorylation and DNA damage-induced apoptosis. Mol. Cell 25, 739–750 (2007)PubMed C. Rinaldo, A. Prodosmo, F. Mancini, S. Iacovelli, A. Sacchi, F. Moretti, S. Soddu, MDM2-regulated degradation of HIPK2 prevents p53Ser46 phosphorylation and DNA damage-induced apoptosis. Mol. Cell 25, 739–750 (2007)PubMed
43.
go back to reference S.M. Sykes, H.S. Mellert, M.A. Holbert, K. Li, R. Marmorstein, W.S. Lane, S.B. McMahon, Acetylation of the p53 DNA-binding domain regulates apoptosis induction. Mol. Cell 24, 841–851 (2006)PubMedCentralPubMed S.M. Sykes, H.S. Mellert, M.A. Holbert, K. Li, R. Marmorstein, W.S. Lane, S.B. McMahon, Acetylation of the p53 DNA-binding domain regulates apoptosis induction. Mol. Cell 24, 841–851 (2006)PubMedCentralPubMed
44.
go back to reference Y. Tang, J. Luo, W. Zhang, W. Gu, Tip60-dependent acetylation of p53 modulates the decision between cell-cycle arrest and apoptosis. Mol. Cell 24, 827–839 (2006)PubMed Y. Tang, J. Luo, W. Zhang, W. Gu, Tip60-dependent acetylation of p53 modulates the decision between cell-cycle arrest and apoptosis. Mol. Cell 24, 827–839 (2006)PubMed
45.
go back to reference S.M. Sykes, T.J. Stanek, A. Frank, M.E. Murphy, S.B. McMahon, Acetylation of the DNA binding domain regulates transcription-independent apoptosis by p53. J. Biol. Chem. 284, 20197–20205 (2009)PubMedCentralPubMed S.M. Sykes, T.J. Stanek, A. Frank, M.E. Murphy, S.B. McMahon, Acetylation of the DNA binding domain regulates transcription-independent apoptosis by p53. J. Biol. Chem. 284, 20197–20205 (2009)PubMedCentralPubMed
46.
go back to reference C.D. Knights, J. Di Catania, S. Giovanni, S. Muratoglu, R. Perez, A. Swartzbeck, A.A. Quong, X. Zhang, T. Beerman, R.G. Pestell, M.L. Avantaggiati, Distinct p53 acetylation cassettes differentially influence gene-expression patterns and cell fate. J. Cell Biol. 173, 533–544 (2006)PubMedCentralPubMed C.D. Knights, J. Di Catania, S. Giovanni, S. Muratoglu, R. Perez, A. Swartzbeck, A.A. Quong, X. Zhang, T. Beerman, R.G. Pestell, M.L. Avantaggiati, Distinct p53 acetylation cassettes differentially influence gene-expression patterns and cell fate. J. Cell Biol. 173, 533–544 (2006)PubMedCentralPubMed
47.
go back to reference J. Luo, A.Y. Nikolaev, S. Imai, D. Chen, F. Su, A. Shiloh, L. Guarente, W. Gu, Negative control of p53 by Sir2alpha promotes cell survival under stress. Cell 107, 137–148 (2001)PubMed J. Luo, A.Y. Nikolaev, S. Imai, D. Chen, F. Su, A. Shiloh, L. Guarente, W. Gu, Negative control of p53 by Sir2alpha promotes cell survival under stress. Cell 107, 137–148 (2001)PubMed
48.
go back to reference J. Yi, J. Luo, SIRT1 and p53, effect on cancer, senescence and beyond. Biochim. Biophys. Acta 2010, 1684–1689 (1804) J. Yi, J. Luo, SIRT1 and p53, effect on cancer, senescence and beyond. Biochim. Biophys. Acta 2010, 1684–1689 (1804)
49.
go back to reference M. Han, E. Song, Y. Guo, X. Ou, C. Mantel, H.E. Broxmeyer, SIRT1 regulates apoptosis and Nanog expression in mouse embryonic stem cells by controlling p53 subcellular localization. Cell Stem Cell 2, 241–251 (2008)PubMedCentralPubMed M. Han, E. Song, Y. Guo, X. Ou, C. Mantel, H.E. Broxmeyer, SIRT1 regulates apoptosis and Nanog expression in mouse embryonic stem cells by controlling p53 subcellular localization. Cell Stem Cell 2, 241–251 (2008)PubMedCentralPubMed
50.
go back to reference M. Miettinen, I. Virtanen, A. Talerman, Intermediate filament proteins in human testis and testicular germ-cell tumors. Am. J. Pathol. 120, 402–410 (1985)PubMedCentralPubMed M. Miettinen, I. Virtanen, A. Talerman, Intermediate filament proteins in human testis and testicular germ-cell tumors. Am. J. Pathol. 120, 402–410 (1985)PubMedCentralPubMed
51.
go back to reference M.A. Calzado, L. De La Vega, E. Munoz, M.L. Schmitz, From top to bottom. The two faces of HIPK2 for regulation of the hypoxic response. Cell Cycle 8, 1659–1664 (2009)PubMed M.A. Calzado, L. De La Vega, E. Munoz, M.L. Schmitz, From top to bottom. The two faces of HIPK2 for regulation of the hypoxic response. Cell Cycle 8, 1659–1664 (2009)PubMed
52.
go back to reference A.S. Coutts, L. Weston, N.B. La Thangue, Actin nucleation by a transcription co-factor that links cytoskeletal events with the p53 response. Cell Cycle 9, 1511–1515 (2010)PubMed A.S. Coutts, L. Weston, N.B. La Thangue, Actin nucleation by a transcription co-factor that links cytoskeletal events with the p53 response. Cell Cycle 9, 1511–1515 (2010)PubMed
53.
go back to reference N. Shikama, C.-W. Lee, S. France, L. Delavaine, J. Lyon, M. Krstic-Demonacos, N.B. La Thangue, A novel cofactor for p300 that regulates the p53 response. Mol. Cell 4, 365–376 (1999)PubMed N. Shikama, C.-W. Lee, S. France, L. Delavaine, J. Lyon, M. Krstic-Demonacos, N.B. La Thangue, A novel cofactor for p300 that regulates the p53 response. Mol. Cell 4, 365–376 (1999)PubMed
54.
55.
go back to reference Y. Samuels-Lev, D.J. O’Connor, D. Bergamaschi, G. Trigiante, J.-K. Hsieh, S. Zhong, I. Campargue, L. Naumovski, T. Crook, X. Lu, ASPP proteins specifically stimulate the apoptotic function of p53. Mol. Cell 8, 781–794 (2001)PubMed Y. Samuels-Lev, D.J. O’Connor, D. Bergamaschi, G. Trigiante, J.-K. Hsieh, S. Zhong, I. Campargue, L. Naumovski, T. Crook, X. Lu, ASPP proteins specifically stimulate the apoptotic function of p53. Mol. Cell 8, 781–794 (2001)PubMed
56.
go back to reference S. Gillotin, X. Lu, The ASPP proteins complex and cooperate with p300 to modulate the transcriptional activity of p53. FEBS Lett. 585, 1778–1782 (2011)PubMed S. Gillotin, X. Lu, The ASPP proteins complex and cooperate with p300 to modulate the transcriptional activity of p53. FEBS Lett. 585, 1778–1782 (2011)PubMed
57.
go back to reference L. Yuan, C. Tian, H. Wang, S. Song, D. Li, G. Xing, Y. Yin, F. He, L. Zhang, Apak competes with p53 for direct binding to intron 1 of p53AIP1 to regulate apoptosis. EMBO Rep. 13, 363–370 (2012)PubMedCentralPubMed L. Yuan, C. Tian, H. Wang, S. Song, D. Li, G. Xing, Y. Yin, F. He, L. Zhang, Apak competes with p53 for direct binding to intron 1 of p53AIP1 to regulate apoptosis. EMBO Rep. 13, 363–370 (2012)PubMedCentralPubMed
58.
go back to reference C. Tian, G. Xing, P. Xie, K. Lu, J. Nie, J. Wang, L. Li, M. Gao, L. Zhang, F. He, KRAB-type zinc-finger protein Apak specifically regulates p53-dependent apoptosis. Nat. Cell Biol. 11, 580–591 (2009)PubMed C. Tian, G. Xing, P. Xie, K. Lu, J. Nie, J. Wang, L. Li, M. Gao, L. Zhang, F. He, KRAB-type zinc-finger protein Apak specifically regulates p53-dependent apoptosis. Nat. Cell Biol. 11, 580–591 (2009)PubMed
59.
go back to reference L. Cheng, M.-T. Sung, P. Cossu-Rocca, T.D. Jones, G.T. MacLennan, J. De Jong, A. Lopez-Beltran, R. Montironi, L.H.J. Looijenga, OCT4: biological functions and clinical applications as a marker of germ cell neoplasia. J. Pathol. 211, 1–9 (2007)PubMed L. Cheng, M.-T. Sung, P. Cossu-Rocca, T.D. Jones, G.T. MacLennan, J. De Jong, A. Lopez-Beltran, R. Montironi, L.H.J. Looijenga, OCT4: biological functions and clinical applications as a marker of germ cell neoplasia. J. Pathol. 211, 1–9 (2007)PubMed
60.
go back to reference S. Stefanovic, M. Pucéat, L’octamanie continue. Le double jeu de OCT4. Med. Sci 26, 411–416 (2010) S. Stefanovic, M. Pucéat, L’octamanie continue. Le double jeu de OCT4. Med. Sci 26, 411–416 (2010)
61.
go back to reference J. Kehler, E. Tolkunova, B. Koschorz, M. Pesce, L. Gentile, M. Boiani, Oct4 is required for primordial germ cell survival. EMBO Rep. 5, 1078–1083 (2004)PubMedCentralPubMed J. Kehler, E. Tolkunova, B. Koschorz, M. Pesce, L. Gentile, M. Boiani, Oct4 is required for primordial germ cell survival. EMBO Rep. 5, 1078–1083 (2004)PubMedCentralPubMed
62.
go back to reference S. Stefanovic, N. Abboud, S. Désilets, D. Nury, C. Cowan, M. Pucéat, Interplay of Oct4 with Sox2 and Sox17: a molecular switch from stem cell pluripotency to specifying a cardiac fate. J. Cell Biol. 186, 665–673 (2009)PubMedCentralPubMed S. Stefanovic, N. Abboud, S. Désilets, D. Nury, C. Cowan, M. Pucéat, Interplay of Oct4 with Sox2 and Sox17: a molecular switch from stem cell pluripotency to specifying a cardiac fate. J. Cell Biol. 186, 665–673 (2009)PubMedCentralPubMed
63.
go back to reference Y.H. Huang, C.C. Chin, H.N. Ho, C.K. Chou, C.N. Shen, H.C. Kuo, Y.C. Wu, Y.C. Hung, C.C. Chang, T.Y. Ling, Pluripotency of mouse spermatogonial stem cells maintened by IGF-1-dependent pathway. FASEB J. 23, 2076–2087 (2009)PubMed Y.H. Huang, C.C. Chin, H.N. Ho, C.K. Chou, C.N. Shen, H.C. Kuo, Y.C. Wu, Y.C. Hung, C.C. Chang, T.Y. Ling, Pluripotency of mouse spermatogonial stem cells maintened by IGF-1-dependent pathway. FASEB J. 23, 2076–2087 (2009)PubMed
64.
go back to reference Y. Lin, Y. Yang, W. Li, Q. Chen, J. Li, X. Pan, L. Zhou, C. Liu, C. Chen, J. He, H. Cao, H. Yao, L. Zheng, X. Xu, Z. Xia, J. Ren, L. Xiao, L. Li, B. Shen, H. Zhou, Y.-J. Wang, Reciprocal regulation of Akt and Oct4 promotes the self-renewal and survival of embryonal carcinoma cells. Mol. Cell 48, 627–640 (2012)PubMedCentralPubMed Y. Lin, Y. Yang, W. Li, Q. Chen, J. Li, X. Pan, L. Zhou, C. Liu, C. Chen, J. He, H. Cao, H. Yao, L. Zheng, X. Xu, Z. Xia, J. Ren, L. Xiao, L. Li, B. Shen, H. Zhou, Y.-J. Wang, Reciprocal regulation of Akt and Oct4 promotes the self-renewal and survival of embryonal carcinoma cells. Mol. Cell 48, 627–640 (2012)PubMedCentralPubMed
65.
go back to reference P. Deb-Rinker, D. Ly, A. Jezierski, M. Sikorska, P.R. Walker, Sequential DNA methylation of the Nanog and Oct-4 upstream regions in human NT2 cells during neuronal differentiation. J. Biol. Chem. 280, 6257–6260 (2005)PubMed P. Deb-Rinker, D. Ly, A. Jezierski, M. Sikorska, P.R. Walker, Sequential DNA methylation of the Nanog and Oct-4 upstream regions in human NT2 cells during neuronal differentiation. J. Biol. Chem. 280, 6257–6260 (2005)PubMed
66.
go back to reference Y.C. Wu, T.Y. Ling, S.H. Lu, H.C. Kuo, H.N. Ho, S.D. Yeh, C.N. Shen, Y.H. Huang, Chemotherapeutic sensitivity of testicular germ cell tumors under hypoxic conditions is negatively regulated by SENP1-controlled sumoylation of OCT4. Cancer Res. 72, 4963–4973 (2012)PubMed Y.C. Wu, T.Y. Ling, S.H. Lu, H.C. Kuo, H.N. Ho, S.D. Yeh, C.N. Shen, Y.H. Huang, Chemotherapeutic sensitivity of testicular germ cell tumors under hypoxic conditions is negatively regulated by SENP1-controlled sumoylation of OCT4. Cancer Res. 72, 4963–4973 (2012)PubMed
67.
go back to reference T.D. Jones, T.M. Ulbright, J.N. Eble, L.A. Baldridge, L. Cheng, OCT4 staining in testicular tumors: a sensitive and specific marker for seminoma and embryonal carcinoma. Am. J. Surg. Pathol. 28, 935–940 (2004)PubMed T.D. Jones, T.M. Ulbright, J.N. Eble, L.A. Baldridge, L. Cheng, OCT4 staining in testicular tumors: a sensitive and specific marker for seminoma and embryonal carcinoma. Am. J. Surg. Pathol. 28, 935–940 (2004)PubMed
68.
go back to reference G. Pan, J.A. Thomson, Nanog and transcriptional networks in embryonic stem cell pluripotency. Cell Res. 17, 42–49 (2007)PubMed G. Pan, J.A. Thomson, Nanog and transcriptional networks in embryonic stem cell pluripotency. Cell Res. 17, 42–49 (2007)PubMed
69.
go back to reference L. Mándoky, B. Szende, L. Géczi, I. Bodrogi, M. Kásler, M. Bak, Apoptosis regulation and spontaneous apoptosis index of testicular germ cell tumors are associated with differentiation and resistance to systemic treatment. Anticancer Res. 28, 1641–1650 (2008)PubMed L. Mándoky, B. Szende, L. Géczi, I. Bodrogi, M. Kásler, M. Bak, Apoptosis regulation and spontaneous apoptosis index of testicular germ cell tumors are associated with differentiation and resistance to systemic treatment. Anticancer Res. 28, 1641–1650 (2008)PubMed
70.
go back to reference D. Juric, S. Sale, R.A. Hromas, R. Yu, Y. Wang, G.E. Duran, R. Tibshirani, L.H. Einhorn, B.I. Sikic, Gene expression profiling differentiates germ cell tumors from other cancers and defines subtype-specific signatures. Proc. Natl. Acad. Sci. U. S. A. 102, 17763–17768 (2005)PubMedCentralPubMed D. Juric, S. Sale, R.A. Hromas, R. Yu, Y. Wang, G.E. Duran, R. Tibshirani, L.H. Einhorn, B.I. Sikic, Gene expression profiling differentiates germ cell tumors from other cancers and defines subtype-specific signatures. Proc. Natl. Acad. Sci. U. S. A. 102, 17763–17768 (2005)PubMedCentralPubMed
71.
go back to reference M.W. Datta, E. Macri, S. Signoretti, A.A. Renshaw, M. Loda, Transition from in situ to invasive testicular germ cell neoplasia is associated with the loss of p21 and gain of mdm-2 expression. Mod. Pathol. 14, 437–442 (2001)PubMed M.W. Datta, E. Macri, S. Signoretti, A.A. Renshaw, M. Loda, Transition from in situ to invasive testicular germ cell neoplasia is associated with the loss of p21 and gain of mdm-2 expression. Mod. Pathol. 14, 437–442 (2001)PubMed
72.
go back to reference R. Koster, A. di Pietro, H. Timmer-Bosscha, J.H. Gibcus, A. van den Berg, A.J. Suurmeijer, R. Bischoff, J.A. Gietema, S. de Jong, Cytoplasmic p21 expression levels determine cisplatin resistance in human testicular cancer. J. Clin. Invest. 120, 3594–3605 (2010)PubMedCentralPubMed R. Koster, A. di Pietro, H. Timmer-Bosscha, J.H. Gibcus, A. van den Berg, A.J. Suurmeijer, R. Bischoff, J.A. Gietema, S. de Jong, Cytoplasmic p21 expression levels determine cisplatin resistance in human testicular cancer. J. Clin. Invest. 120, 3594–3605 (2010)PubMedCentralPubMed
73.
go back to reference T. Mueller, L.P. Mueller, J. Luetzkendorf, W. Voigt, H. Simon, H.J. Schmoll, Loss of Oct-3/4 expression in embryonal carcinoma cells is associated with induction of cisplatin resistance. Tumour Biol. 27, 71–83 (2006)PubMed T. Mueller, L.P. Mueller, J. Luetzkendorf, W. Voigt, H. Simon, H.J. Schmoll, Loss of Oct-3/4 expression in embryonal carcinoma cells is associated with induction of cisplatin resistance. Tumour Biol. 27, 71–83 (2006)PubMed
74.
go back to reference A. Singh, J. Settleman, EMT, cancer stem cells and drug resistance: an emerging axis of evil in the war on cancer. Oncogene 29, 4741–4751 (2010)PubMedCentralPubMed A. Singh, J. Settleman, EMT, cancer stem cells and drug resistance: an emerging axis of evil in the war on cancer. Oncogene 29, 4741–4751 (2010)PubMedCentralPubMed
75.
go back to reference I.A. Voutsadakis, Molecular predictors of gemcitabine response in pancreatic cancer. World J. Gastrointest. Oncol. 3, 153–164 (2011)PubMedCentralPubMed I.A. Voutsadakis, Molecular predictors of gemcitabine response in pancreatic cancer. World J. Gastrointest. Oncol. 3, 153–164 (2011)PubMedCentralPubMed
76.
go back to reference L.-L. Tsai, C.-C. Yu, Y.-C. Chang, C.-H. Yu, M.-Y. Chou, Markedly increased Oct4 and Nanog expression correlates with cisplatin resistance in oral squamous cell carcinoma. J. Oral Pathol. Med. 40, 621–628 (2011)PubMed L.-L. Tsai, C.-C. Yu, Y.-C. Chang, C.-H. Yu, M.-Y. Chou, Markedly increased Oct4 and Nanog expression correlates with cisplatin resistance in oral squamous cell carcinoma. J. Oral Pathol. Med. 40, 621–628 (2011)PubMed
77.
go back to reference N.S. Hillbertz, J.M. Hirsch, J. Jalouli, M.M. Jalouli, L. Sand, Viral and molecular aspects of oral cancer. Anticancer Res. 32, 4201–4212 (2012)PubMed N.S. Hillbertz, J.M. Hirsch, J. Jalouli, M.M. Jalouli, L. Sand, Viral and molecular aspects of oral cancer. Anticancer Res. 32, 4201–4212 (2012)PubMed
78.
go back to reference J. Houldsworth, H. Xiao, V.V.V.S. Murty, W. Chen, B. Ray, V.E. Reuter, G.J. Bosl, R.S.K. Chaganti, Human male germ cell tumor resistance to cisplatin is linked to TP53 gene mutation. Oncogene 16, 2345–2349 (1998)PubMed J. Houldsworth, H. Xiao, V.V.V.S. Murty, W. Chen, B. Ray, V.E. Reuter, G.J. Bosl, R.S.K. Chaganti, Human male germ cell tumor resistance to cisplatin is linked to TP53 gene mutation. Oncogene 16, 2345–2349 (1998)PubMed
79.
go back to reference T. Mueller, L.P. Mueller, H.-J. Holzhausen, R. Witthuhn, P. Albers, H.J. Schmoll, Histological evidence for the existence of germ cell tumor cells showing embryonal carcinoma morphology but lacking OCT4 expression and cisplatin sensitivity. Histochem. Cell Biol. 134, 197–204 (2010)PubMed T. Mueller, L.P. Mueller, H.-J. Holzhausen, R. Witthuhn, P. Albers, H.J. Schmoll, Histological evidence for the existence of germ cell tumor cells showing embryonal carcinoma morphology but lacking OCT4 expression and cisplatin sensitivity. Histochem. Cell Biol. 134, 197–204 (2010)PubMed
80.
go back to reference K. Takahashi, S. Yamanaka, Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126, 663–676 (2006)PubMed K. Takahashi, S. Yamanaka, Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126, 663–676 (2006)PubMed
81.
go back to reference N. Tapia, H.R. Scholer, p53 connects tumorigenesis and reprogramming to pluripotency. J. Exp. Med. 207, 2045–2048 (2010)PubMedCentralPubMed N. Tapia, H.R. Scholer, p53 connects tumorigenesis and reprogramming to pluripotency. J. Exp. Med. 207, 2045–2048 (2010)PubMedCentralPubMed
82.
go back to reference R.R. Chivukula, J.T. Mendell, Abate and switch: miR-145 in stem cell differentiation. Cell 137, 606–608 (2009)PubMed R.R. Chivukula, J.T. Mendell, Abate and switch: miR-145 in stem cell differentiation. Cell 137, 606–608 (2009)PubMed
83.
go back to reference P. Svoboda, M. Flemr, The role of miRNAs and endogenous siRNAs in maternal-to-zygotic reprogramming and the establishment of pluripotency. EMBO Rep. 11, 590–597 (2010)PubMedCentralPubMed P. Svoboda, M. Flemr, The role of miRNAs and endogenous siRNAs in maternal-to-zygotic reprogramming and the establishment of pluripotency. EMBO Rep. 11, 590–597 (2010)PubMedCentralPubMed
84.
go back to reference L. Liu, J. Lian, H. Zhang, H. Tian, M. Liang, M. Yin, F. Sun, Micro-RNA-302a sensitizes testicular embryonal carcinoma cells to cisplatin-induced cell death. J. Cell. Physiol. 228, 2294–2304 (2013)PubMed L. Liu, J. Lian, H. Zhang, H. Tian, M. Liang, M. Yin, F. Sun, Micro-RNA-302a sensitizes testicular embryonal carcinoma cells to cisplatin-induced cell death. J. Cell. Physiol. 228, 2294–2304 (2013)PubMed
85.
go back to reference N. Xu, T. Papagiannakopoulos, G. Pan, J.A. Thomson, K.S. Kosik, MicroRNA-145 regulates OCT4, SOX2 and KLF4 and represses pluripotency in human embryonic stem cells. Cell 137, 647–658 (2009)PubMed N. Xu, T. Papagiannakopoulos, G. Pan, J.A. Thomson, K.S. Kosik, MicroRNA-145 regulates OCT4, SOX2 and KLF4 and represses pluripotency in human embryonic stem cells. Cell 137, 647–658 (2009)PubMed
86.
go back to reference S.O. Suh, Y. Chen, M.S. Zaman, H. Hirata, S. Yamamura, V. Shahryari, J. Liu, Z.L. Tabatabai, S. Kakar, G. Deng, Y. Tanaka, R. Dahiya, MicroRNA-145 is regulated by methylation and p53 gene mutation in prostate cancer. Carcinogenesis 32, 772–778 (2011)PubMedCentralPubMed S.O. Suh, Y. Chen, M.S. Zaman, H. Hirata, S. Yamamura, V. Shahryari, J. Liu, Z.L. Tabatabai, S. Kakar, G. Deng, Y. Tanaka, R. Dahiya, MicroRNA-145 is regulated by methylation and p53 gene mutation in prostate cancer. Carcinogenesis 32, 772–778 (2011)PubMedCentralPubMed
87.
go back to reference N. Riggi, M.-L. Suvà, C. De Vito, P. Provero, J.-C. Stehle, K. Baumer, L. Cironi, M. Janiszewska, T. Petricevic, D. Suvà, S. Tercier, J.-M. Joseph, L. Guillou, I. Stamenkovic, EWS-FLI-1 modulates miRNA145 and SOX2 expression to initiate mesenchymal stem cell reprogramming toward Ewing sarcoma cancer stem cells. Genes Dev. 24, 916–932 (2010)PubMedCentralPubMed N. Riggi, M.-L. Suvà, C. De Vito, P. Provero, J.-C. Stehle, K. Baumer, L. Cironi, M. Janiszewska, T. Petricevic, D. Suvà, S. Tercier, J.-M. Joseph, L. Guillou, I. Stamenkovic, EWS-FLI-1 modulates miRNA145 and SOX2 expression to initiate mesenchymal stem cell reprogramming toward Ewing sarcoma cancer stem cells. Genes Dev. 24, 916–932 (2010)PubMedCentralPubMed
88.
go back to reference C. Krausz, L.H.J. Looijenga, Genetic aspects of testicular germ cell tumors. Cell Cycle 7, 3519–3524 (2008)PubMed C. Krausz, L.H.J. Looijenga, Genetic aspects of testicular germ cell tumors. Cell Cycle 7, 3519–3524 (2008)PubMed
89.
go back to reference F. Anokye-Danso, C.M. Trivedi, D. Juhr, M. Gupta, Z. Cui, Y. Tian, Y. Zhang, W. Yang, P.J. Gruber, J.A. Epstein, E.E. Morrisey, Highly efficient miRNA-mediated reprogramming of mouse and human somatic cells to pluripotency. Cell Stem Cell 8, 376–388 (2011)PubMedCentralPubMed F. Anokye-Danso, C.M. Trivedi, D. Juhr, M. Gupta, Z. Cui, Y. Tian, Y. Zhang, W. Yang, P.J. Gruber, J.A. Epstein, E.E. Morrisey, Highly efficient miRNA-mediated reprogramming of mouse and human somatic cells to pluripotency. Cell Stem Cell 8, 376–388 (2011)PubMedCentralPubMed
90.
go back to reference S.-L. Lin, Concise review: deciphering the mechanism behind induced pluripotent stem cell generation. Stem Cells 29, 1645–1649 (2011)PubMedCentralPubMed S.-L. Lin, Concise review: deciphering the mechanism behind induced pluripotent stem cell generation. Stem Cells 29, 1645–1649 (2011)PubMedCentralPubMed
91.
go back to reference D.K. Ma, J.U. Guo, G. Ming, H. Song, DNA excision repair proteins and Gadd45 as molecular players for active DNA demethylation. Cell Cycle 8, 1526–1531 (2009)PubMedCentralPubMed D.K. Ma, J.U. Guo, G. Ming, H. Song, DNA excision repair proteins and Gadd45 as molecular players for active DNA demethylation. Cell Cycle 8, 1526–1531 (2009)PubMedCentralPubMed
92.
go back to reference P.M. Voorhoeve, C. le Sage, M. Schrier, A.J.M. Gillis, H. Stoop, R. Nagel, Y.-P. Liu, J. van Duijse, J. Drost, A. Griekspoor, E. Zlotorynski, N. Yabuta, G. De Vita, H. Nojima, L.H.J. Looijenga, R. Agami, A genetic screen implicates miRNA-372 and miRNA-373 as oncogenes in testicular germ cell tumors. Cell 124, 1169–1181 (2006)PubMed P.M. Voorhoeve, C. le Sage, M. Schrier, A.J.M. Gillis, H. Stoop, R. Nagel, Y.-P. Liu, J. van Duijse, J. Drost, A. Griekspoor, E. Zlotorynski, N. Yabuta, G. De Vita, H. Nojima, L.H.J. Looijenga, R. Agami, A genetic screen implicates miRNA-372 and miRNA-373 as oncogenes in testicular germ cell tumors. Cell 124, 1169–1181 (2006)PubMed
93.
go back to reference M.L. Wang, S.H. Chiou, C.W. Wu, Targeting cancer stem cells: emerging role of Nanog transcription factor. Oncol. Targets Ther. 6, 1207–1220 (2013) M.L. Wang, S.H. Chiou, C.W. Wu, Targeting cancer stem cells: emerging role of Nanog transcription factor. Oncol. Targets Ther. 6, 1207–1220 (2013)
94.
go back to reference P. Navarro, P. Avner, When X-inactivation meets pluripotency: an intimate rendezvous. FEBS Lett. 583, 1721–1727 (2009)PubMed P. Navarro, P. Avner, When X-inactivation meets pluripotency: an intimate rendezvous. FEBS Lett. 583, 1721–1727 (2009)PubMed
95.
go back to reference A.H. Hart, L. Hartley, K. Parker, M. Ibrahim, L.H.J. Looijenga, M. Pauchnik, C.W. Chow, L. Robb, The pluripotency homeobox gene NANOG is expressed in human germ cell tumors. Cancer 104, 2092–2098 (2005)PubMed A.H. Hart, L. Hartley, K. Parker, M. Ibrahim, L.H.J. Looijenga, M. Pauchnik, C.W. Chow, L. Robb, The pluripotency homeobox gene NANOG is expressed in human germ cell tumors. Cancer 104, 2092–2098 (2005)PubMed
96.
go back to reference P.E. Szabo, K. Hubner, H. Scholer, J.R. Mann, Allele-specific expression of imprinted genes in mouse migratory primordial germ cells. Mech. Dev. 115, 157–160 (2002)PubMed P.E. Szabo, K. Hubner, H. Scholer, J.R. Mann, Allele-specific expression of imprinted genes in mouse migratory primordial germ cells. Mech. Dev. 115, 157–160 (2002)PubMed
97.
go back to reference D. Gilbert, E. Rapley, J. Shipley, Testicular germ cell tumours: predisposition genes and the male germ cell niche. Nat. Rev. Cancer 11, 278–288 (2011)PubMed D. Gilbert, E. Rapley, J. Shipley, Testicular germ cell tumours: predisposition genes and the male germ cell niche. Nat. Rev. Cancer 11, 278–288 (2011)PubMed
98.
go back to reference M. Kucia, R. Reca, K. Miekus, J. Wanzeck, W. Wojakowski, A. Janowska-Wieczorek, J. Ratajczak, M.Z. Ratajczak, Trafficking of normal stem cells and metastasis of cancer stem cells involve similar mechanisms: pivotal role of the SDF-1-CXCR4 axis. Stem Cells 23, 879–894 (2005)PubMed M. Kucia, R. Reca, K. Miekus, J. Wanzeck, W. Wojakowski, A. Janowska-Wieczorek, J. Ratajczak, M.Z. Ratajczak, Trafficking of normal stem cells and metastasis of cancer stem cells involve similar mechanisms: pivotal role of the SDF-1-CXCR4 axis. Stem Cells 23, 879–894 (2005)PubMed
99.
go back to reference M.C. Mostert, A.J.M.H. Verkerk, M. van de Pol, J. Heighway, P. Marynen, C. Rosenberg, A. Geurts van Kessel, J. van Echten, B. de Jong, J.W. Oosterhuis, L.H.J. Looijenga, Identification of the critical region of 12p over-representation in testicular germ cell tumors of adolescents and adults. Oncogene 16, 2617–2627 (1998)PubMed M.C. Mostert, A.J.M.H. Verkerk, M. van de Pol, J. Heighway, P. Marynen, C. Rosenberg, A. Geurts van Kessel, J. van Echten, B. de Jong, J.W. Oosterhuis, L.H.J. Looijenga, Identification of the critical region of 12p over-representation in testicular germ cell tumors of adolescents and adults. Oncogene 16, 2617–2627 (1998)PubMed
100.
go back to reference S. Rodriguez, O. Jafer, H. Goker, B.M. Summersgill, G. Zafarana, A.J.M. Gillis, R.J.H.L.M. van Gurp, J.W. Oosterhuis, Y.-J. Lu, R. Huddart, C.S. Cooper, J. Clark, L.H.J. Looijenga, J.M. Shipley, Expression profile of genes from 12p in testicular germ cell tumors of adolescents and adults associated with i(12p) and amplification at 12p11.2-p12.1. Oncogene 22, 1880–1891 (2003)PubMed S. Rodriguez, O. Jafer, H. Goker, B.M. Summersgill, G. Zafarana, A.J.M. Gillis, R.J.H.L.M. van Gurp, J.W. Oosterhuis, Y.-J. Lu, R. Huddart, C.S. Cooper, J. Clark, L.H.J. Looijenga, J.M. Shipley, Expression profile of genes from 12p in testicular germ cell tumors of adolescents and adults associated with i(12p) and amplification at 12p11.2-p12.1. Oncogene 22, 1880–1891 (2003)PubMed
101.
go back to reference J.E. Korkola, J. Houldsworth, R.S.V. Chadalavada, A.B.. Olshen, D. Dobrzynski, V.E. Reuter, G.J. Bosl, R.S.K. Chaganti, Down-regulation of stem cell genes, including those in a 200-kb gene cluster at 12p13.31, is associated with in vivo differentiation of human male germ cell tumors. Cancer Res. 66, 820–827 (2006) J.E. Korkola, J. Houldsworth, R.S.V. Chadalavada, A.B.. Olshen, D. Dobrzynski, V.E. Reuter, G.J. Bosl, R.S.K. Chaganti, Down-regulation of stem cell genes, including those in a 200-kb gene cluster at 12p13.31, is associated with in vivo differentiation of human male germ cell tumors. Cancer Res. 66, 820–827 (2006)
102.
go back to reference K. Tanaka, S. Okamoto, Y. Ishikawa, H. Tamura, T. Hara, DDX1 is required for testicular tumorigenesis, partially through the transcriptional activation of 12p stem cell genes. Oncogene 28, 2142–2151 (2009)PubMed K. Tanaka, S. Okamoto, Y. Ishikawa, H. Tamura, T. Hara, DDX1 is required for testicular tumorigenesis, partially through the transcriptional activation of 12p stem cell genes. Oncogene 28, 2142–2151 (2009)PubMed
103.
go back to reference C.J. Giuliano, J.S. Kerley-Hamilton, T. Bee, S.J. Freemantle, R. Manickaratnam, E. Dmitrovsky, M.J. Spinella, Retinoic acid represses a cassette of candidate pluripotency chromosome 12p genes during induced loss of human embryonal carcinoma tumorigenicity. Biochim. Biophys. Acta 1731, 48–56 (2005)PubMed C.J. Giuliano, J.S. Kerley-Hamilton, T. Bee, S.J. Freemantle, R. Manickaratnam, E. Dmitrovsky, M.J. Spinella, Retinoic acid represses a cassette of candidate pluripotency chromosome 12p genes during induced loss of human embryonal carcinoma tumorigenicity. Biochim. Biophys. Acta 1731, 48–56 (2005)PubMed
104.
go back to reference E. Rajpert-De Meyts, R. Hanstein, N. Jorgensen, N. Graem, P.H. Vogt, N.E. Skakkebaek, Developmental expression of POU5F1 (OCT-3/4) in normal and dysgenetic human gonads. Hum. Reprod. 19, 1338–1344 (2004)PubMed E. Rajpert-De Meyts, R. Hanstein, N. Jorgensen, N. Graem, P.H. Vogt, N.E. Skakkebaek, Developmental expression of POU5F1 (OCT-3/4) in normal and dysgenetic human gonads. Hum. Reprod. 19, 1338–1344 (2004)PubMed
105.
go back to reference M. Cools, S.L.S. Drop, K.P. Wolfenbuttel, J.W. Oosterhuis, L.H.J. Looijenga, Germ cell tumors in the intersex gonad: old paths, new directions, moving frontiers. Endocr. Rev. 27, 468–484 (2006)PubMed M. Cools, S.L.S. Drop, K.P. Wolfenbuttel, J.W. Oosterhuis, L.H.J. Looijenga, Germ cell tumors in the intersex gonad: old paths, new directions, moving frontiers. Endocr. Rev. 27, 468–484 (2006)PubMed
106.
go back to reference K. McClelland, J. Bowles, P. Koopman, Male sex determination: insights into molecular mechanisms. Asian J. Androl. 14, 164–171 (2012)PubMedCentralPubMed K. McClelland, J. Bowles, P. Koopman, Male sex determination: insights into molecular mechanisms. Asian J. Androl. 14, 164–171 (2012)PubMedCentralPubMed
107.
go back to reference J.S. Fisher, Environmental anti-androgens and male reproductive health: focus on phthalates and testicular dysgenesis syndrome. Reproduction 127, 305–315 (2004)PubMed J.S. Fisher, Environmental anti-androgens and male reproductive health: focus on phthalates and testicular dysgenesis syndrome. Reproduction 127, 305–315 (2004)PubMed
108.
go back to reference F. Orso, E. Cottone, M.D. Hasleton, Activator protein-2gamma (AP-2gamma) expression is specifically induced by oestrogens through binding of the oestrogen receptor to a canonical element within the5′-untranslated region. Biochem. J. 377, 429–438 (2004)PubMedCentralPubMed F. Orso, E. Cottone, M.D. Hasleton, Activator protein-2gamma (AP-2gamma) expression is specifically induced by oestrogens through binding of the oestrogen receptor to a canonical element within the5′-untranslated region. Biochem. J. 377, 429–438 (2004)PubMedCentralPubMed
109.
go back to reference D. Eckert, S. Buhl, S. Weber, R. Jӓger, H. Schorle, The AP-2 family of transcription factors. Genome Biol. 6, 246 (2005)PubMedCentralPubMed D. Eckert, S. Buhl, S. Weber, R. Jӓger, H. Schorle, The AP-2 family of transcription factors. Genome Biol. 6, 246 (2005)PubMedCentralPubMed
110.
go back to reference S. Schӓfer, J. Anschlag, D. Nettersheim, N. Haas, L. Pawig, H. Schorle, The role of BLIMP1 and its putative downstream target TFAP2C in germ cell development and germ cell tumours. Int. J. Androl. 34, e152–e159 (2011) S. Schӓfer, J. Anschlag, D. Nettersheim, N. Haas, L. Pawig, H. Schorle, The role of BLIMP1 and its putative downstream target TFAP2C in germ cell development and germ cell tumours. Int. J. Androl. 34, e152–e159 (2011)
111.
go back to reference C.E. Hoei-Hansen, J.E. Nielsen, K. Almstrup, S.B. Sonne, N. Graem, N.E. Skakkebaek, H. Leffers, E. Rajpert-De Meyts, Transcription Factor AP-2γ is a developmentally regulated marker of testicular carcinoma in situ and germ cell tumors. Clin. Cancer Res. 10, 8521–8530 (2004)PubMed C.E. Hoei-Hansen, J.E. Nielsen, K. Almstrup, S.B. Sonne, N. Graem, N.E. Skakkebaek, H. Leffers, E. Rajpert-De Meyts, Transcription Factor AP-2γ is a developmentally regulated marker of testicular carcinoma in situ and germ cell tumors. Clin. Cancer Res. 10, 8521–8530 (2004)PubMed
112.
go back to reference A. Jørgensen, J.E. Nielsen, M. Blomberg Jensen, N. Graem, E. Rajpert-De Meyts, Analysis of meiosis regulators in human gonads: a sexually dimorphic spatio-temporal expression pattern suggests involvement of DMRT1 in meiotic entry. Mol. Hum. Reprod. 18, 523–534 (2012)PubMed A. Jørgensen, J.E. Nielsen, M. Blomberg Jensen, N. Graem, E. Rajpert-De Meyts, Analysis of meiosis regulators in human gonads: a sexually dimorphic spatio-temporal expression pattern suggests involvement of DMRT1 in meiotic entry. Mol. Hum. Reprod. 18, 523–534 (2012)PubMed
113.
go back to reference A. Jørgensen, J.E. Nielsen, K. Almstrup, B.G. Toft, B.L. Petersen, E. Rajpert-De Meyts, Dysregulation of the mitosis-meiosis switch in testicular carcinoma in situ. J. Pathol. 229, 588–598 (2013)PubMed A. Jørgensen, J.E. Nielsen, K. Almstrup, B.G. Toft, B.L. Petersen, E. Rajpert-De Meyts, Dysregulation of the mitosis-meiosis switch in testicular carcinoma in situ. J. Pathol. 229, 588–598 (2013)PubMed
114.
go back to reference S.M. Kraggerud, C.E. Hoei-Hansen, S. Alagaratnam, R.I. Skotheim, V.M. Abeler, E. Rajpert-De Meyts, R.A. Lothe, Molecular characteristics of malignant ovarian germ cell tumors and comparison with testicular counterparts: implications for pathogenesis. Endocr. Rev. 34, 339–376 (2013)PubMedCentralPubMed S.M. Kraggerud, C.E. Hoei-Hansen, S. Alagaratnam, R.I. Skotheim, V.M. Abeler, E. Rajpert-De Meyts, R.A. Lothe, Molecular characteristics of malignant ovarian germ cell tumors and comparison with testicular counterparts: implications for pathogenesis. Endocr. Rev. 34, 339–376 (2013)PubMedCentralPubMed
115.
go back to reference P. Arnaud, Genomic imprinting in germ cells: imprints are under control. Reproduction 140, 411–423 (2010)PubMed P. Arnaud, Genomic imprinting in germ cells: imprints are under control. Reproduction 140, 411–423 (2010)PubMed
117.
go back to reference P. Savage, J. Stebbing, M. Bower, T. Crook, Why does cytotoxic chemotherapy cure only some cancers? Nat. Clin. Pract. Oncol. 6, 43–52 (2009)PubMed P. Savage, J. Stebbing, M. Bower, T. Crook, Why does cytotoxic chemotherapy cure only some cancers? Nat. Clin. Pract. Oncol. 6, 43–52 (2009)PubMed
118.
go back to reference J. Bartkova, C.J. Bakkenist, E. Rajpert-De Meyts, N.E. Skakkebaek, M. Sehested, J. Lukas, M.B. Kastan, J. Bartek, ATM activation in normal human tissues and testicular cancer. Cell Cycle 4, 838–845 (2005)PubMed J. Bartkova, C.J. Bakkenist, E. Rajpert-De Meyts, N.E. Skakkebaek, M. Sehested, J. Lukas, M.B. Kastan, J. Bartek, ATM activation in normal human tissues and testicular cancer. Cell Cycle 4, 838–845 (2005)PubMed
119.
go back to reference F. Cavallo, D.R. Feldman, M. Barchi, Revisiting DNA damage repair, p53-mediated apoptosis and cisplatin sensitivity in germ cell tumors. Int. J. Dev. Biol. 57, 273–280 (2013)PubMed F. Cavallo, D.R. Feldman, M. Barchi, Revisiting DNA damage repair, p53-mediated apoptosis and cisplatin sensitivity in germ cell tumors. Int. J. Dev. Biol. 57, 273–280 (2013)PubMed
120.
go back to reference B. Köberle, J.R. Masters, J.A. Hartley, R.D. Wood, Defective repair of cisplatin-induced DNA damage caused by reduced XPA protein in testicular germ cell tumours. Curr. Biol. 9, 273–276 (1999)PubMed B. Köberle, J.R. Masters, J.A. Hartley, R.D. Wood, Defective repair of cisplatin-induced DNA damage caused by reduced XPA protein in testicular germ cell tumours. Curr. Biol. 9, 273–276 (1999)PubMed
121.
go back to reference C. Welsh, R. Day, C. McGurk, J.R.W. Masters, R.D. Wood, B. Köberle, Reduced levels of XPA, ERCC1 and XPF DNA repair proteins in testis tumor cell lines. Int. J. Cancer 110, 352–361 (2004)PubMed C. Welsh, R. Day, C. McGurk, J.R.W. Masters, R.D. Wood, B. Köberle, Reduced levels of XPA, ERCC1 and XPF DNA repair proteins in testis tumor cell lines. Int. J. Cancer 110, 352–361 (2004)PubMed
122.
go back to reference B. Köberle, W. Brenner, A. Albers, S. Usanova, J.W. Thüroff, B. Kaina, ERCC1 and XPF expression in human testicular germ cell tumors. Oncol. Rep. 23, 223–227 (2010)PubMed B. Köberle, W. Brenner, A. Albers, S. Usanova, J.W. Thüroff, B. Kaina, ERCC1 and XPF expression in human testicular germ cell tumors. Oncol. Rep. 23, 223–227 (2010)PubMed
123.
go back to reference J. Mendoza, J. Martínez, C. Hernández, D. Pérez-Montiel, C. Castro, E. Fabián-Morales, M. Santibáñez, R. González-Barrios, J. Díaz-Chávez, M.A. Andonegui, N. Reynoso, L.F. Oñate, M.A. Jiménez, M. Núñez, R. Dyer, L.A. Herrera, Association between ERCC1 and XPA expression and polymorphisms and the response to cisplatin in testicular germ cell tumours. Br. J. Cancer 109, 68–75 (2013)PubMedCentralPubMed J. Mendoza, J. Martínez, C. Hernández, D. Pérez-Montiel, C. Castro, E. Fabián-Morales, M. Santibáñez, R. González-Barrios, J. Díaz-Chávez, M.A. Andonegui, N. Reynoso, L.F. Oñate, M.A. Jiménez, M. Núñez, R. Dyer, L.A. Herrera, Association between ERCC1 and XPA expression and polymorphisms and the response to cisplatin in testicular germ cell tumours. Br. J. Cancer 109, 68–75 (2013)PubMedCentralPubMed
124.
go back to reference S. Bhana, A. Hewer, D.H. Phillips, D.R. Lloyd, p53-dependent global nucleoside excision repair of cisplatin-induced intrastand cross links in human cells. Mutagenesis 23, 131–136 (2008)PubMed S. Bhana, A. Hewer, D.H. Phillips, D.R. Lloyd, p53-dependent global nucleoside excision repair of cisplatin-induced intrastand cross links in human cells. Mutagenesis 23, 131–136 (2008)PubMed
125.
go back to reference F. Cavallo, G. Graziani, C. Antinozzi, D.R. Feldman, J. Houldsworth, G.J. Bosl, R.S. Chaganti, M.E. Moynahan, M. Jasin, M. Barchi, Reduced proficiency in homologous recombination underlies the high sensitivity of embryonal carcinoma testicular germ cell tumors to Cisplatin and poly (adp-ribose) polymerase inhibition. PLoS One 7, e51563 (2012)PubMedCentralPubMed F. Cavallo, G. Graziani, C. Antinozzi, D.R. Feldman, J. Houldsworth, G.J. Bosl, R.S. Chaganti, M.E. Moynahan, M. Jasin, M. Barchi, Reduced proficiency in homologous recombination underlies the high sensitivity of embryonal carcinoma testicular germ cell tumors to Cisplatin and poly (adp-ribose) polymerase inhibition. PLoS One 7, e51563 (2012)PubMedCentralPubMed
126.
go back to reference K.A. Olaussen, A. Dunant, P. Fouret, E. Brambilla, F. André, V. Haddad, E. Taranchon, M. Filipits, J.-P. Pignon, T. Tursz, T. Le Chevalier, J.-C. Soria, DNA repair by ERCC1 in Non-Small-Cell Lung Cancer and cisplatin-based adjuvant chemotherapy. New Engl. J. Med. 355, 983–991 (2006)PubMed K.A. Olaussen, A. Dunant, P. Fouret, E. Brambilla, F. André, V. Haddad, E. Taranchon, M. Filipits, J.-P. Pignon, T. Tursz, T. Le Chevalier, J.-C. Soria, DNA repair by ERCC1 in Non-Small-Cell Lung Cancer and cisplatin-based adjuvant chemotherapy. New Engl. J. Med. 355, 983–991 (2006)PubMed
127.
go back to reference E. Ribeiro, M. Ganzinelli, D. Andreis, R. Bertoni, R. Giardini, S.B. Fox, M. Broggini, A. Bottini, V. Zanoni, L. Bazzola, C. Foroni, D. Generali, G. Damia, Triple negative breast cancers have a reduced expression of DNA repair genes. PLoS ONE 8, e66243 (2013)PubMedCentralPubMed E. Ribeiro, M. Ganzinelli, D. Andreis, R. Bertoni, R. Giardini, S.B. Fox, M. Broggini, A. Bottini, V. Zanoni, L. Bazzola, C. Foroni, D. Generali, G. Damia, Triple negative breast cancers have a reduced expression of DNA repair genes. PLoS ONE 8, e66243 (2013)PubMedCentralPubMed
128.
go back to reference Z. Zheng, T. Chen, X. Li, E. Haura, A. Sharma, G. Bepler, DNA synthesis and repair genes RRM1 and ERCC1 in lung cancer. New Engl. J. Med. 356, 800–808 (2007)PubMed Z. Zheng, T. Chen, X. Li, E. Haura, A. Sharma, G. Bepler, DNA synthesis and repair genes RRM1 and ERCC1 in lung cancer. New Engl. J. Med. 356, 800–808 (2007)PubMed
129.
go back to reference R. Vélez-Cruz, D.G. Johnson, E2F1 and p53 transcription factors as accessory factors for nucleotide excision repair. Int. J. Mol. Sci. 13, 13554–13568 (2012)PubMedCentralPubMed R. Vélez-Cruz, D.G. Johnson, E2F1 and p53 transcription factors as accessory factors for nucleotide excision repair. Int. J. Mol. Sci. 13, 13554–13568 (2012)PubMedCentralPubMed
130.
go back to reference S.R. Martens-de Kemp, S.U. Dalm, F.M.J. Wijnolts, A. Brink, R.J. Honeywell, G.J. Peters, B.J.M. Braakhuis, R.H. Brakenhoff, DNA-bound platinum is the major determinant of cisplatin sensitivity in head and necksquamous carcinoma cells. PLoS ONE 8, e61555 (2013)PubMedCentralPubMed S.R. Martens-de Kemp, S.U. Dalm, F.M.J. Wijnolts, A. Brink, R.J. Honeywell, G.J. Peters, B.J.M. Braakhuis, R.H. Brakenhoff, DNA-bound platinum is the major determinant of cisplatin sensitivity in head and necksquamous carcinoma cells. PLoS ONE 8, e61555 (2013)PubMedCentralPubMed
131.
go back to reference L. Friboulet, K.A. Olaussen, J.P. Pignon, F.A. Shepherd, M.S. Tsao, S. Graziano, R. Kratzke, J.Y. Douillard, L. Seymour, R. Pirker, M. Filipits, F. André, E. Solary, F. Ponsonnailles, A. Robin, A. Stoclin, N. Dorvault, F. Commo, J. Adam, E. Vanhecke, P. Saulnier, J. Thomale, T. Le Chevalier, A. Dunant, V. Rousseau, G. Le Teuff, E. Brambilla, J.C. Soria, ERCC1 isoform expression and DNA repair in non-small-cell lung cancer. New Engl. J. Med. 368, 1101–1110 (2013)PubMed L. Friboulet, K.A. Olaussen, J.P. Pignon, F.A. Shepherd, M.S. Tsao, S. Graziano, R. Kratzke, J.Y. Douillard, L. Seymour, R. Pirker, M. Filipits, F. André, E. Solary, F. Ponsonnailles, A. Robin, A. Stoclin, N. Dorvault, F. Commo, J. Adam, E. Vanhecke, P. Saulnier, J. Thomale, T. Le Chevalier, A. Dunant, V. Rousseau, G. Le Teuff, E. Brambilla, J.C. Soria, ERCC1 isoform expression and DNA repair in non-small-cell lung cancer. New Engl. J. Med. 368, 1101–1110 (2013)PubMed
132.
go back to reference M.J.P. Welters, B.J.M. Braakhuis, A.J. Jacobs-Bergmans, A. Kegel, R.A. Baan, W.J.F. van der Vijgh, A.M.J. Fichtinger-Schepman, The potential of platinum-DNA adduct determination in ex vivo treated tumor fragments for the prediction of sensitivity to cisplatin chemotherapy. Ann. Oncol. 10, 97–103 (1999)PubMed M.J.P. Welters, B.J.M. Braakhuis, A.J. Jacobs-Bergmans, A. Kegel, R.A. Baan, W.J.F. van der Vijgh, A.M.J. Fichtinger-Schepman, The potential of platinum-DNA adduct determination in ex vivo treated tumor fragments for the prediction of sensitivity to cisplatin chemotherapy. Ann. Oncol. 10, 97–103 (1999)PubMed
133.
go back to reference X. Chen, L.J. Ko, L. Jayaraman, C. Prives, p53 levels, functional domains, and DNA damage determine the extent of the apoptotic response of tumor cells. Genes Dev. 10, 2438–2451 (1996)PubMed X. Chen, L.J. Ko, L. Jayaraman, C. Prives, p53 levels, functional domains, and DNA damage determine the extent of the apoptotic response of tumor cells. Genes Dev. 10, 2438–2451 (1996)PubMed
134.
go back to reference J. Fan, J.R. Bertino, Modulation of cisplatinum cytotoxicity byp53: effect of p53-mediated apoptosis and DNA repair. Mol. Pharmacol. 56, 966–972 (1999)PubMed J. Fan, J.R. Bertino, Modulation of cisplatinum cytotoxicity byp53: effect of p53-mediated apoptosis and DNA repair. Mol. Pharmacol. 56, 966–972 (1999)PubMed
135.
go back to reference J.C. Huang, D.B. Zamble, J.T. Reardon, S.J. Lippard, A. Sancar, HMG-domain proteins specifically inhibit the repair of the major DNA adduct of the anticancer drug cisplatin by human excision nuclease. Proc. Natl. Acad. Sci. U. S. A. 91, 10394–10398 (1994)PubMedCentralPubMed J.C. Huang, D.B. Zamble, J.T. Reardon, S.J. Lippard, A. Sancar, HMG-domain proteins specifically inhibit the repair of the major DNA adduct of the anticancer drug cisplatin by human excision nuclease. Proc. Natl. Acad. Sci. U. S. A. 91, 10394–10398 (1994)PubMedCentralPubMed
136.
go back to reference L. Jayaraman, N. Chandra Moorthy, K.G.K. Murthy, J.L. Manley, M. Bustin, C. Prives, High mobility group protein-1 (HMG-1) is a unique activator of p53. Genes Dev. 12, 462–472 (1998)PubMedCentralPubMed L. Jayaraman, N. Chandra Moorthy, K.G.K. Murthy, J.L. Manley, M. Bustin, C. Prives, High mobility group protein-1 (HMG-1) is a unique activator of p53. Genes Dev. 12, 462–472 (1998)PubMedCentralPubMed
137.
go back to reference Q. He, C.H. Liang, S.J. Lippard, Steroid hormones induce HMG1 overexpression and sensitize breast cancer cells to cisplatin and carboplatin. Proc. Natl. Acad. Sci. U. S. A. 97, 5768–5772 (2000)PubMedCentralPubMed Q. He, C.H. Liang, S.J. Lippard, Steroid hormones induce HMG1 overexpression and sensitize breast cancer cells to cisplatin and carboplatin. Proc. Natl. Acad. Sci. U. S. A. 97, 5768–5772 (2000)PubMedCentralPubMed
138.
go back to reference G. Nagatani, M. Nomoto, H. Takano, T. Ise, K. Kato, T. Imamura, H. Izumi, K. Makishima, K. Kohno, Transcriptional activation of the human HMG1 gene in cisplatin-resistant human cancer cells. Cancer Res. 61, 1592–1597 (2001)PubMed G. Nagatani, M. Nomoto, H. Takano, T. Ise, K. Kato, T. Imamura, H. Izumi, K. Makishima, K. Kohno, Transcriptional activation of the human HMG1 gene in cisplatin-resistant human cancer cells. Cancer Res. 61, 1592–1597 (2001)PubMed
139.
go back to reference S.S. Lange, K.M. Vasquez, HMGB1: The jack-of-all-trades protein is a master DNA repair mechanic. Mol. Carcinog. 48, 571–580 (2009)PubMedCentralPubMed S.S. Lange, K.M. Vasquez, HMGB1: The jack-of-all-trades protein is a master DNA repair mechanic. Mol. Carcinog. 48, 571–580 (2009)PubMedCentralPubMed
140.
go back to reference K.M. Livesey, R. Kang, P. Vernon, W. Buchser, P. Loughran, S.C. Watkins, L. Zhang, J.J. Manfredi, H.J. Zeh 3rd, L. Li, M.T. Lotze, D. Tang, p53/HMGB1 complexes regulate autophagy and apoptosis. Cancer Res. 72, 1996–2005 (2012)PubMedCentralPubMed K.M. Livesey, R. Kang, P. Vernon, W. Buchser, P. Loughran, S.C. Watkins, L. Zhang, J.J. Manfredi, H.J. Zeh 3rd, L. Li, M.T. Lotze, D. Tang, p53/HMGB1 complexes regulate autophagy and apoptosis. Cancer Res. 72, 1996–2005 (2012)PubMedCentralPubMed
141.
go back to reference K.M. Livesey, R. Kang, H.J. Zeh III, M.T. Lotze, D. Tang, Direct molecular interactions between HMGB1 and TP53 in colorectal cancer. Autophagy 8, 846–848 (2012)PubMedCentralPubMed K.M. Livesey, R. Kang, H.J. Zeh III, M.T. Lotze, D. Tang, Direct molecular interactions between HMGB1 and TP53 in colorectal cancer. Autophagy 8, 846–848 (2012)PubMedCentralPubMed
142.
go back to reference D. Tang, R. Kang, H.J. Zeh III, M.T. Lotze, High-Mobility Group Box 1, oxidative stress, and disease. Antioxid. Redox Signal 14, 1315–1335 (2011)PubMedCentralPubMed D. Tang, R. Kang, H.J. Zeh III, M.T. Lotze, High-Mobility Group Box 1, oxidative stress, and disease. Antioxid. Redox Signal 14, 1315–1335 (2011)PubMedCentralPubMed
143.
144.
go back to reference R. Franco, F. Esposito, M. Fedele, G. Liguori, G.M. Pierantoni, G. Botti, D. Tramontano, A. Fusco, P. Chieffi, Detection of high-mobility group proteins A1 and A2 represents a valid diagnostic marker in post-pubertal testicular germ cell tumours. J. Pathol. 214, 58–64 (2008)PubMed R. Franco, F. Esposito, M. Fedele, G. Liguori, G.M. Pierantoni, G. Botti, D. Tramontano, A. Fusco, P. Chieffi, Detection of high-mobility group proteins A1 and A2 represents a valid diagnostic marker in post-pubertal testicular germ cell tumours. J. Pathol. 214, 58–64 (2008)PubMed
145.
go back to reference G. D’Orazi, C. Rinaldo, S. Soddu, Updates on HIPK2: a resourceful oncosuppressor for clearing cancer. J. Exp. Clin. Cancer Res. 31, 63 (2012)PubMedCentralPubMed G. D’Orazi, C. Rinaldo, S. Soddu, Updates on HIPK2: a resourceful oncosuppressor for clearing cancer. J. Exp. Clin. Cancer Res. 31, 63 (2012)PubMedCentralPubMed
146.
go back to reference L.M.S. Resar, The High Mobility Group A1 gene: transforming inflammatory signals into cancer. Cancer Res. 70, 436–439 (2010)PubMedCentralPubMed L.M.S. Resar, The High Mobility Group A1 gene: transforming inflammatory signals into cancer. Cancer Res. 70, 436–439 (2010)PubMedCentralPubMed
147.
go back to reference I. Ben-Porath, M.W. Thomson, V.J. Carey, R. Ge, G.W. Bell, A. Regev, R.A. Weinberg, An embryonicstem cell-like gene expression signature in poorly differentiated aggressive human tumors. Nat. Genet. 40, 499–507 (2008)PubMedCentralPubMed I. Ben-Porath, M.W. Thomson, V.J. Carey, R. Ge, G.W. Bell, A. Regev, R.A. Weinberg, An embryonicstem cell-like gene expression signature in poorly differentiated aggressive human tumors. Nat. Genet. 40, 499–507 (2008)PubMedCentralPubMed
148.
go back to reference F. Honecker, H. Wermann, F. Mayer, A.J.M. Gillis, H. Stoop, R.J.L.M. van Gurp, K. Oechsle, E. Steyerberg, J.T. Hartmann, W.N.M. Dinjens, J.W. Oosterhuis, C. Bokemeyer, L.H.J. Looijenga, Microsatellite instability, Mismatch Repair deficiency, and BRAF mutation in treatment-resistant germ cell tumors. J. Clin. Oncol. 27, 2129–2136 (2009)PubMed F. Honecker, H. Wermann, F. Mayer, A.J.M. Gillis, H. Stoop, R.J.L.M. van Gurp, K. Oechsle, E. Steyerberg, J.T. Hartmann, W.N.M. Dinjens, J.W. Oosterhuis, C. Bokemeyer, L.H.J. Looijenga, Microsatellite instability, Mismatch Repair deficiency, and BRAF mutation in treatment-resistant germ cell tumors. J. Clin. Oncol. 27, 2129–2136 (2009)PubMed
149.
go back to reference C.M. Ribic, D.J. Sergent, M.J. Moore, S.N. Thibodeau, A.J. French, R.M. Goldberg, S.R. Hamilton, P. Laurent-Puig, R. Gryfe, L.E. Shepherd, D. Tu, M. Redston, S. Gallinger, Tumor microsatellite-instability status as a predictor of benefit from fluorouracil-based adjuvant chemotherapy for colon cancer. New Engl. J. Med. 349, 247–257 (2003)PubMedCentralPubMed C.M. Ribic, D.J. Sergent, M.J. Moore, S.N. Thibodeau, A.J. French, R.M. Goldberg, S.R. Hamilton, P. Laurent-Puig, R. Gryfe, L.E. Shepherd, D. Tu, M. Redston, S. Gallinger, Tumor microsatellite-instability status as a predictor of benefit from fluorouracil-based adjuvant chemotherapy for colon cancer. New Engl. J. Med. 349, 247–257 (2003)PubMedCentralPubMed
150.
go back to reference F.A. Sinicrope, M.R. Mahoney, T.C. Smyrk, S.N. Thibodeau, R.S. Warren, M.M. Bertagnolli, G.D. Nelson, R.M. Goldberg, D.J. Sargent, S.R. Alberts, Prognostic impact of deficient DNA Mismatch Repair in patients with stage III colon cancer from a randomized trial of FOLFOX-based adjuvant chemotherapy. J. Clin. Oncol. 31, 3664–3672 (2013)PubMed F.A. Sinicrope, M.R. Mahoney, T.C. Smyrk, S.N. Thibodeau, R.S. Warren, M.M. Bertagnolli, G.D. Nelson, R.M. Goldberg, D.J. Sargent, S.R. Alberts, Prognostic impact of deficient DNA Mismatch Repair in patients with stage III colon cancer from a randomized trial of FOLFOX-based adjuvant chemotherapy. J. Clin. Oncol. 31, 3664–3672 (2013)PubMed
151.
go back to reference D. Fink, S. Nebel, S. Aebi, H. Zheng, B. Cenni, A. Nehmé, R.D. Christen, S.B. Howell, The role of DNA mismatch repair in platinum drug resistance. Cancer Res. 56, 4881–4886 (1996)PubMed D. Fink, S. Nebel, S. Aebi, H. Zheng, B. Cenni, A. Nehmé, R.D. Christen, S.B. Howell, The role of DNA mismatch repair in platinum drug resistance. Cancer Res. 56, 4881–4886 (1996)PubMed
152.
go back to reference P.B. Chapman, A. Hauschild, C. Robert, J.B. Haanen, P. Ascierto, J. Larkin, R. Dummer, C. Garbe, A. Testori, M. Maio, D. Hogg, P. Lorigan, C. Lebbe, T. Jouary, D. Schadendorf, A. Ribas, S.J. O’Day, J.A. Sosman, J.M. Kirkwood, A.M. Eggermont, B. Dreno, K. Nolop, J. Li, B. Nelson, J. Hou, R.J. Lee, K.T. Flaherty, G.A. McArthur, BRIM-3 Study Group: improved survival with vemurafenib in melanoma with BRAF V600E mutation. N. Engl. J. Med. 364, 2507–2516 (2011)PubMedCentralPubMed P.B. Chapman, A. Hauschild, C. Robert, J.B. Haanen, P. Ascierto, J. Larkin, R. Dummer, C. Garbe, A. Testori, M. Maio, D. Hogg, P. Lorigan, C. Lebbe, T. Jouary, D. Schadendorf, A. Ribas, S.J. O’Day, J.A. Sosman, J.M. Kirkwood, A.M. Eggermont, B. Dreno, K. Nolop, J. Li, B. Nelson, J. Hou, R.J. Lee, K.T. Flaherty, G.A. McArthur, BRIM-3 Study Group: improved survival with vemurafenib in melanoma with BRAF V600E mutation. N. Engl. J. Med. 364, 2507–2516 (2011)PubMedCentralPubMed
153.
go back to reference S.P. Wang, W.L. Wang, Y.L. Chang, C.T. Wu, Y.C. Chao, S.H. Kao, A. Yuan, C.W. Lin, S.C. Yang, W.K. Chan, K.C. Li, T.M. Hong, P.C. Yang, p53 controls cancer cell invasion by inducing the MDM2-mediated degradation of Slug. Nat. Cell Biol. 11, 694–704 (2009)PubMed S.P. Wang, W.L. Wang, Y.L. Chang, C.T. Wu, Y.C. Chao, S.H. Kao, A. Yuan, C.W. Lin, S.C. Yang, W.K. Chan, K.C. Li, T.M. Hong, P.C. Yang, p53 controls cancer cell invasion by inducing the MDM2-mediated degradation of Slug. Nat. Cell Biol. 11, 694–704 (2009)PubMed
154.
go back to reference R. Li, J. Liang, S. Ni, T. Zhou, X. Qing, H. Li, W. He, J. Chen, F. Li, Q. Zhuang, B. Qin, J. Xu, W. Li, J. Yang, Y. Gan, D. Qin, S. Feng, H. Song, D. Yang, B. Zhang, L. Zeng, L. Lai, M.A. Esteban, D. Pei, A mesenchymal-to-epithelial transition initiates and is required for the nuclear reprogramming of mouse fibroblasts. Cell Stem Cell 7, 51–63 (2010)PubMed R. Li, J. Liang, S. Ni, T. Zhou, X. Qing, H. Li, W. He, J. Chen, F. Li, Q. Zhuang, B. Qin, J. Xu, W. Li, J. Yang, Y. Gan, D. Qin, S. Feng, H. Song, D. Yang, B. Zhang, L. Zeng, L. Lai, M.A. Esteban, D. Pei, A mesenchymal-to-epithelial transition initiates and is required for the nuclear reprogramming of mouse fibroblasts. Cell Stem Cell 7, 51–63 (2010)PubMed
155.
go back to reference S.H. Chiou, M.L. Wang, Y.T. Chou, C.J. Chen, C.F. Hong, W.J. Hsieh, H.T. Chang, Y.S. Chen, T.W. Lin, H.S. Hsu, C.W. Wu, Coexpression of oct4 and nanog enhances malignancy in lung adenocarcinoma by inducing cancer stem cell-like properties and epithelial-mesenchymal transdifferentiation. Cancer Res. 70, 10433–10444 (2010)PubMed S.H. Chiou, M.L. Wang, Y.T. Chou, C.J. Chen, C.F. Hong, W.J. Hsieh, H.T. Chang, Y.S. Chen, T.W. Lin, H.S. Hsu, C.W. Wu, Coexpression of oct4 and nanog enhances malignancy in lung adenocarcinoma by inducing cancer stem cell-like properties and epithelial-mesenchymal transdifferentiation. Cancer Res. 70, 10433–10444 (2010)PubMed
156.
go back to reference M.K. Siu, E.S. Wong, D.S. Kong, H.Y. Chan, L. Jiang, O.G. Wong, E.W. Lam, K.K. Chan, H.Y. Ngan, X.F. Le, A.N. Cheung, Stem cell transcription factor NANOG controls cell migration and invasion via dysregulation of E-cadherin and FoxJ1 and contributes to adverse clinical outcome in ovarian cancers. Oncogene 32, 3500–3509 (2013)PubMed M.K. Siu, E.S. Wong, D.S. Kong, H.Y. Chan, L. Jiang, O.G. Wong, E.W. Lam, K.K. Chan, H.Y. Ngan, X.F. Le, A.N. Cheung, Stem cell transcription factor NANOG controls cell migration and invasion via dysregulation of E-cadherin and FoxJ1 and contributes to adverse clinical outcome in ovarian cancers. Oncogene 32, 3500–3509 (2013)PubMed
157.
go back to reference U.I. Ezeh, P.J. Turek, R.A. Reijo, A.T. Clark, Human embryonic stem cell genes OCT4, NANOG, STELLAR, and GDF3 are expressed in both seminoma and breast carcinoma. Cancer 104, 2255–2265 (2005)PubMed U.I. Ezeh, P.J. Turek, R.A. Reijo, A.T. Clark, Human embryonic stem cell genes OCT4, NANOG, STELLAR, and GDF3 are expressed in both seminoma and breast carcinoma. Cancer 104, 2255–2265 (2005)PubMed
Metadata
Title
The chemosensitivity of testicular germ cell tumors
Author
Ioannis A. Voutsadakis
Publication date
01-04-2014
Publisher
Springer Netherlands
Published in
Cellular Oncology / Issue 2/2014
Print ISSN: 2211-3428
Electronic ISSN: 2211-3436
DOI
https://doi.org/10.1007/s13402-014-0168-6

Other articles of this Issue 2/2014

Cellular Oncology 2/2014 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine