51
|
Zhang Q, Tian X, Hu G, Shi P, Wu J, Li S, Zhou H, Jin BK, Yang J, Zhang S, Tian Y. Dual-Functional Analogous cis-Platinum Complex with High Antitumor Activities and Two-Photon Bioimaging. Biochemistry 2015; 54:2177-80. [DOI: 10.1021/bi5014062] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Qiong Zhang
- Key
Laboratory of Functional Inorganic Material Chemistry of Anhui Province,
Department of Chemistry, Anhui University, Hefei 230039, P. R. China
| | - Xiaohe Tian
- Department
of Chemistry, The MRC/UCL Centre for Medical Molecular Virology, University College London, London WC1H 0AJ, U.K
| | - Guiju Hu
- Key
Laboratory of Functional Inorganic Material Chemistry of Anhui Province,
Department of Chemistry, Anhui University, Hefei 230039, P. R. China
| | - Pengfei Shi
- Key
Laboratory of Functional Inorganic Material Chemistry of Anhui Province,
Department of Chemistry, Anhui University, Hefei 230039, P. R. China
| | - Jieying Wu
- Key
Laboratory of Functional Inorganic Material Chemistry of Anhui Province,
Department of Chemistry, Anhui University, Hefei 230039, P. R. China
| | - Shengli Li
- Key
Laboratory of Functional Inorganic Material Chemistry of Anhui Province,
Department of Chemistry, Anhui University, Hefei 230039, P. R. China
| | - Hongping Zhou
- Key
Laboratory of Functional Inorganic Material Chemistry of Anhui Province,
Department of Chemistry, Anhui University, Hefei 230039, P. R. China
| | - Bao-Kang Jin
- Key
Laboratory of Functional Inorganic Material Chemistry of Anhui Province,
Department of Chemistry, Anhui University, Hefei 230039, P. R. China
| | - Jiaxiang Yang
- Key
Laboratory of Functional Inorganic Material Chemistry of Anhui Province,
Department of Chemistry, Anhui University, Hefei 230039, P. R. China
| | - Shengyi Zhang
- Key
Laboratory of Functional Inorganic Material Chemistry of Anhui Province,
Department of Chemistry, Anhui University, Hefei 230039, P. R. China
| | - Yupeng Tian
- Key
Laboratory of Functional Inorganic Material Chemistry of Anhui Province,
Department of Chemistry, Anhui University, Hefei 230039, P. R. China
| |
Collapse
|
52
|
Malvezzi S, Sturla SJ, Tanasova M. Quantification of pyrophosphate as a universal approach to determine polymerase activity and assay polymerase inhibitors. Anal Biochem 2015; 478:1-7. [PMID: 25772306 DOI: 10.1016/j.ab.2015.03.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 02/24/2015] [Accepted: 03/03/2015] [Indexed: 11/17/2022]
Abstract
The importance of DNA polymerases in biology and biotechnology, and their recognition as potential therapeutic targets, drives development of methods for deriving kinetic characteristics of polymerases and their propensity to perform polynucleotide synthesis over modified DNA templates. Among various polymerases, translesion synthesis (TLS) polymerases enable cells to avoid the cytotoxic stalling of replicative DNA polymerases at chemotherapy-induced DNA lesions, thereby leading to drug resistance. Identification of TLS inhibitors to overcome drug-resistance necessitates the development of appropriate high-throughput assays. Since polymerase-mediated DNA synthesis involves the release of inorganic pyrophosphate (PPi), we established a universal and fast method for monitoring the progress of DNA polymerases based on the quantification of PPi with a fluorescence-based assay that we coupled to in vitro primer extension reactions. The established assay has a nanomolar detection limit in PPi and enables the evaluation of single nucleotide incorporation and DNA synthesis progression kinetics. The results demonstrated that the developed assay is a reliable method for monitoring TLS and identifying nucleoside and nucleotide-based TLS inhibitors.
Collapse
Affiliation(s)
- S Malvezzi
- Department of Health Sciences and Technology, ETH Zurich Schmelzbergstrasse 9, 8092 Zurich, Switzerland
| | - S J Sturla
- Department of Health Sciences and Technology, ETH Zurich Schmelzbergstrasse 9, 8092 Zurich, Switzerland
| | - M Tanasova
- Department of Chemistry, Michigan Technological University, 1400 Townsend Drive, Houghton, MI 49931, USA.
| |
Collapse
|
53
|
Platinum-induced kidney damage: Unraveling the DNA damage response (DDR) of renal tubular epithelial and glomerular endothelial cells following platinum injury. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:685-98. [DOI: 10.1016/j.bbamcr.2014.12.033] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 12/17/2014] [Accepted: 12/29/2014] [Indexed: 11/19/2022]
|
54
|
Selbo PK, Bostad M, Olsen CE, Edwards VT, Høgset A, Weyergang A, Berg K. Photochemical internalisation, a minimally invasive strategy for light-controlled endosomal escape of cancer stem cell-targeting therapeutics. Photochem Photobiol Sci 2015; 14:1433-50. [DOI: 10.1039/c5pp00027k] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Despite progress in radio-, chemo- and photodynamic-therapy (PDT) of cancer, treatment resistance still remains a major problem for patients with aggressive tumours.
Collapse
Affiliation(s)
- Pål Kristian Selbo
- Department of Radiation Biology
- Institute for Cancer Research
- The Norwegian Radium Hospital
- Oslo University Hospital
- Montebello
| | - Monica Bostad
- Department of Radiation Biology
- Institute for Cancer Research
- The Norwegian Radium Hospital
- Oslo University Hospital
- Montebello
| | - Cathrine Elisabeth Olsen
- Department of Radiation Biology
- Institute for Cancer Research
- The Norwegian Radium Hospital
- Oslo University Hospital
- Montebello
| | - Victoria Tudor Edwards
- Department of Radiation Biology
- Institute for Cancer Research
- The Norwegian Radium Hospital
- Oslo University Hospital
- Montebello
| | - Anders Høgset
- Cancer Stem Cell Innovation Center (SFI-CAST)
- Institute for Cancer Research
- Norwegian Radium Hospital
- Oslo University Hospital
- Oslo
| | - Anette Weyergang
- Department of Radiation Biology
- Institute for Cancer Research
- The Norwegian Radium Hospital
- Oslo University Hospital
- Montebello
| | - Kristian Berg
- Department of Radiation Biology
- Institute for Cancer Research
- The Norwegian Radium Hospital
- Oslo University Hospital
- Montebello
| |
Collapse
|
55
|
Targeted therapies in germ cell tumors. Urol Oncol 2014; 33:363-9. [PMID: 25544153 DOI: 10.1016/j.urolonc.2014.09.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 09/05/2014] [Accepted: 09/08/2014] [Indexed: 11/20/2022]
|
56
|
Guth AM, Deogracias M, Dow SW. Comparison of cancer stem cell antigen expression by tumor cell lines and by tumor biopsies from dogs with melanoma and osteosarcoma. Vet Immunol Immunopathol 2014; 161:132-40. [PMID: 25146881 DOI: 10.1016/j.vetimm.2014.07.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Revised: 04/25/2014] [Accepted: 07/17/2014] [Indexed: 12/18/2022]
Abstract
Cancer stem cells (CSCs) represent a small subpopulation of tumor cells that play a critical role in initiating and sustaining tumor growth. However, we currently have an incomplete understanding of the expression patterns of CSC antigens in tumors of dogs, nor do we understand how expression of these antigens vary between tumor cell lines and tumor biopsy specimens. Therefore, we used flow cytometry and commonly reported CSC surface and intracellular markers to evaluate the phenotype and overall frequency of CSC subpopulations in tumor cell lines and primary tumor biopsy samples from dogs with melanoma and osteosarcoma. We found that cells expressing common CSC antigens were rare in tumor cell lines, with the exception of tumor cells expressing CD44 and CD90. In contrast, tumor cells expressing conventional CSC antigens such as CD133, CD34, CD44, CD24 and Oct3/4 were much more common in tumor biopsy samples. Notably, the frequency and types of putative CSC subpopulations were very similar in biopsy samples from dogs with either melanoma or osteosarcoma. Our results suggest that the tumor microenvironment significantly influences CSC subpopulations within tumors and that tumor cell lines may not accurately reflect the actual frequency or types of CSC subpopulations present in tumor tissues in vivo.
Collapse
Affiliation(s)
- Amanda M Guth
- Animal Cancer Center, Department of Clinical Sciences, Colorado State University, Ft. Collins, CO 80523, United States
| | - Mike Deogracias
- Animal Cancer Center, Department of Clinical Sciences, Colorado State University, Ft. Collins, CO 80523, United States
| | - Steven W Dow
- Animal Cancer Center, Department of Clinical Sciences, Colorado State University, Ft. Collins, CO 80523, United States.
| |
Collapse
|
57
|
Levine PM, Garabedian MJ, Kirshenbaum K. Targeting the androgen receptor with steroid conjugates. J Med Chem 2014; 57:8224-37. [PMID: 24936953 PMCID: PMC4207530 DOI: 10.1021/jm500101h] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The androgen receptor (AR) is a major therapeutic target in prostate cancer pharmacology. Progression of prostate cancer has been linked to elevated expression of AR in malignant tissue, suggesting that AR plays a central role in prostate cancer cell biology. Potent therapeutic agents can be precisely crafted to specifically target AR, potentially averting systemic toxicities associated with nonspecific chemotherapies. In this review, we describe various strategies to generate steroid conjugates that can selectively engage AR with high potency. Analogies to recent developments in nonsteroidal conjugates targeting AR are also evaluated. Particular focus is placed on potential applications in AR pharmacology. The review culminates with a description of future prospects for targeting AR.
Collapse
Affiliation(s)
- Paul M Levine
- Department of Chemistry, New York University , New York, New York 10003, United States
| | | | | |
Collapse
|
58
|
Mazo JD, García-López J, Weber M. Epigenetic traits of testicular cancer: from primordial germ cells to germ cell tumors. Epigenomics 2014; 6:253-5. [DOI: 10.2217/epi.14.24] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Affiliation(s)
- Jesús del Mazo
- Centro de Investigaciones Biológicas, C.I.B. (CSIC), Ramiro de Maeztu 9, 28040-Madrid, Spain
| | - Jesús García-López
- Centro de Investigaciones Biológicas, C.I.B. (CSIC), Ramiro de Maeztu 9, 28040-Madrid, Spain
| | - Michael Weber
- Biotechnology & Cell Signaling, CNRS UMR7242, University of Strasbourg, 300 Bd., Sébastien Brant, BP 10413, 67412 Illkirch Cedex, France
| |
Collapse
|
59
|
Epithelial-mesenchymal status renders differential responses to cisplatin in ovarian cancer. Oncogene 2014; 34:1899-907. [PMID: 24858042 DOI: 10.1038/onc.2014.136] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Revised: 03/20/2014] [Accepted: 03/28/2014] [Indexed: 12/22/2022]
Abstract
Chemoresistance to platinums, such as cisplatin, is of critical concern in the treatment of ovarian cancer. Recent evidence has linked epithelial-mesenchymal transition (EMT) as a contributing mechanism. The current study explored the connection between cellular responses to cisplatin and EMT in ovarian cancer. Expression microarrays were utilized to estimate the EMT status as a binary phenotype, and the transcriptional responses of 46 ovarian cancer cell lines to cisplatin were measured at dosages equivalent to 50% growth inhibition. Phenotypic responses to cisplatin were quantified with respect to cell number, proliferation rate and apoptosis, and then compared with the epithelial or mesenchymal status. Ovarian cancer cell lines with an epithelial status exhibited higher resistance to cisplatin treatment in the MTS assay than those with a mesenchymal status. Pathway analyses revealed the induction of G1/S- and S-phase genes (P=0.001) and the activation of multiple NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells) downstream genes (P=0.0016) by cisplatin selectively in epithelial-like cell lines. BrdU incorporation and Caspase-3/7 release assays confirmed impaired apoptosis in epithelial-like ovarian cancer cells. In clinical samples, we observed resistance to single platinum treatment and the selective activation of the NF-κB pathway by platinum in ovarian cancers with an epithelial status. Overall, our results suggest that, in epithelial-like ovarian cancer cells, NF-κB activation by cisplatin may lead to defective apoptosis, preferential proliferation arrest and a consequential decreased sensitivity to cisplatin.
Collapse
|
60
|
Machado CR, Vieira-da-Rocha JP, Mendes IC, Rajão MA, Marcello L, Bitar M, Drummond MG, Grynberg P, Oliveira DAA, Marques C, Van Houten B, McCulloch R. Nucleotide excision repair in Trypanosoma brucei: specialization of transcription-coupled repair due to multigenic transcription. Mol Microbiol 2014; 92:756-76. [PMID: 24661334 PMCID: PMC4138998 DOI: 10.1111/mmi.12589] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/22/2014] [Indexed: 02/02/2023]
Abstract
Nucleotide excision repair (NER) is a highly conserved genome repair pathway acting on helix distorting DNA lesions. NER is divided into two subpathways: global genome NER (GG-NER), which is responsible for repair throughout genomes, and transcription-coupled NER (TC-NER), which acts on lesions that impede transcription. The extent of the Trypanosoma brucei genome that is transcribed is highly unusual, since most genes are organized in multigene transcription units, each transcribed from a single promoter. Given this transcription organization, we have addressed the importance of NER to T. brucei genome maintenance by performing RNAi against all predicted contributing factors. Our results indicate that TC-NER is the main pathway of NER repair, but only CSB, XPBz and XPG contribute. Moreover, we show that UV lesions are inefficiently repaired in T. brucei, perhaps due to preferential use of RNA polymerase translesion synthesis. RNAi of XPC and DDB was found to be lethal, and we show that these factors act in inter-strand cross-link repair. XPD and XPB appear only to act in transcription, not repair. This work indicates that the predominance of multigenic transcription in T. brucei has resulted in pronounced adaptation of NER relative to the host and may be an attractive drug target.
Collapse
Affiliation(s)
- Carlos R Machado
- Departamento de Bioquímica e Imunologia, ICB, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Caixa Postal 486, Belo Horizonte, 30161-970, MG, Brazil
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
61
|
Abada PB, Howell SB. Cisplatin induces resistance by triggering differentiation of testicular embryonal carcinoma cells. PLoS One 2014; 9:e87444. [PMID: 24475288 PMCID: PMC3903721 DOI: 10.1371/journal.pone.0087444] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Accepted: 12/27/2013] [Indexed: 12/13/2022] Open
Abstract
Although testicular germ cell tumors are generally quite responsive to treatment with cisplatin, a small fraction of them acquire resistance during therapy. Even when cisplatin treatment is successful the patient is often left with a residual teratoma at the site of the primary tumor suggesting that cisplatin may trigger differentiation in some tumors. Using the human embryonal carcinoma cell line NTera2/D1, we confirmed that exposure to the differentiating agent retinoic acid produced a reduction in pluripotency markers NANOG and POU5F1 (Oct3/4) and an acute concentration-dependent increase in resistance to both cisplatin and paclitaxel that reached as high as 18-fold for cisplatin and 61-fold for paclitaxel within four days. A two day exposure to cisplatin also produced a concentration-dependent decrease in the expression of the NANOG and POU5F1 and increased expression of three markers whose levels increase with differentiation including Nestin, SCG10 and Fibronectin. In parallel, exposure to cisplatin induced up to 6.2-fold resistance to itself and 104-fold resistance to paclitaxel. Paclitaxel did not induce differentiation or resistance to either itself or cisplatin. Neither retinoic acid nor cisplatin induced resistance in cervical or prostate cancer cell lines or other germ cell tumor lines in which they failed to alter the expression of NANOG and POU5F1. Forced expression of NANOG prevented the induction of resistance to cisplatin by retinoic acid. We conclude that cisplatin can acutely induce resistance to itself and paclitaxel by triggering a differentiation response in pluripotent germ cell tumor cells.
Collapse
Affiliation(s)
- Paolo B. Abada
- Department of Medicine and the Moores UCSD Cancer Center, University of California San Diego, La Jolla, California, United States of America
| | - Stephen B. Howell
- Department of Medicine and the Moores UCSD Cancer Center, University of California San Diego, La Jolla, California, United States of America
- * E-mail:
| |
Collapse
|
62
|
Oberoi HS, Nukolova NV, Kabanov AV, Bronich TK. Nanocarriers for delivery of platinum anticancer drugs. Adv Drug Deliv Rev 2013; 65:1667-85. [PMID: 24113520 PMCID: PMC4197009 DOI: 10.1016/j.addr.2013.09.014] [Citation(s) in RCA: 298] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Revised: 09/19/2013] [Accepted: 09/25/2013] [Indexed: 12/18/2022]
Abstract
Platinum based anticancer drugs have revolutionized cancer chemotherapy, and continue to be in widespread clinical use especially for management of tumors of the ovary, testes, and the head and neck. However, several dose limiting toxicities associated with platinum drug use, partial anti-tumor response in most patients, development of drug resistance, tumor relapse, and many other challenges have severely limited the patient quality of life. These limitations have motivated an extensive research effort towards development of new strategies for improving platinum therapy. Nanocarrier-based delivery of platinum compounds is one such area of intense research effort beginning to provide encouraging preclinical and clinical results and may allow the development of the next generation of platinum chemotherapy. This review highlights current understanding on the pharmacology and limitations of platinum compounds in clinical use, and provides a comprehensive analysis of various platinum-polymer complexes, micelles, dendrimers, liposomes and other nanoparticles currently under investigation for delivery of platinum drugs.
Collapse
Affiliation(s)
- Hardeep S. Oberoi
- Department of Pharmaceutical Sciences and Center for Drug Delivery and Nanomedicine, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Natalia V. Nukolova
- Department of Chemistry, M.V. Lomonosov Moscow State University, Leninskie Gory, Moscow 119992, Russia
- Russian State Medical University, Department of Medical Nanobiotechnology, Ostrovityanova 1, Moscow 117997, Russia
| | - Alexander V. Kabanov
- Department of Chemistry, M.V. Lomonosov Moscow State University, Leninskie Gory, Moscow 119992, Russia
- Center for Nanotechnology in Drug Delivery and Division of Molecular Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Tatiana K. Bronich
- Department of Pharmaceutical Sciences and Center for Drug Delivery and Nanomedicine, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
63
|
Uema N, Ooshio T, Harada K, Naito M, Naka K, Hoshii T, Tadokoro Y, Ohta K, Ali MAE, Katano M, Soga T, Nakanuma Y, Okuda A, Hirao A. Abundant nucleostemin expression supports the undifferentiated properties of germ cell tumors. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 183:592-603. [PMID: 23885716 DOI: 10.1016/j.ajpath.2013.04.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Revised: 03/21/2013] [Accepted: 04/30/2013] [Indexed: 12/23/2022]
Abstract
Nucleostemin (NS) is a nucleolar GTP-binding protein that is involved in ribosomal biogenesis and protection of telomeres. We investigated the expression of NS in human germ cell tumors and its function in a mouse germ cell tumor model. NS was abundantly expressed in undifferentiated, but not differentiated, types of human testicular germ cell tumors. NS was expressed concomitantly with OCT3/4, a critical regulator of the undifferentiated status of pluripotent stem cells in primordial germ cells and embryonal carcinomas. To investigate the roles of NS in tumor growth in vivo, we used a mouse teratoma model. Analysis of teratomas derived from embryonic stem cells in which the NS promoter drives GFP expression showed that cells highly expressing NS were actively proliferating and exhibited the characteristics of tumor-initiating cells, including the ability to initiate and propagate tumor cells in vivo. NS-expressing cells exhibited higher levels of GTP than non-NS-expressing cells. Because NS protein is stabilized by intracellular GTP, metabolic changes may contribute to abundant NS expression in the undifferentiated cells. OCT3/4 deficiency in teratomas led to loss of NS expression, resulting in growth retardation. Finally, we found that teratomas deficient in NS lost their undifferentiated characteristics, resulting in defective tumor proliferation. These data indicate that abundant expression of NS supports the undifferentiated properties of germ cell tumors.
Collapse
Affiliation(s)
- Noriyuki Uema
- Division of Molecular Genetics, Cancer and Stem Cell Program, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
64
|
Zeron-Medina J, Wang X, Repapi E, Campbell MR, Su D, Castro-Giner F, Davies B, Peterse EF, Sacilotto N, Walker GJ, Terzian T, Tomlinson IP, Box NF, Meinshausen N, De Val S, Bell DA, Bond GL. A polymorphic p53 response element in KIT ligand influences cancer risk and has undergone natural selection. Cell 2013; 155:410-22. [PMID: 24120139 PMCID: PMC4171736 DOI: 10.1016/j.cell.2013.09.017] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Revised: 07/09/2013] [Accepted: 09/10/2013] [Indexed: 12/13/2022]
Abstract
The ability of p53 to regulate transcription is crucial for tumor suppression and implies that inherited polymorphisms in functional p53-binding sites could influence cancer. Here, we identify a polymorphic p53 responsive element and demonstrate its influence on cancer risk using genome-wide data sets of cancer susceptibility loci, genetic variation, p53 occupancy, and p53-binding sites. We uncover a single-nucleotide polymorphism (SNP) in a functional p53-binding site and establish its influence on the ability of p53 to bind to and regulate transcription of the KITLG gene. The SNP resides in KITLG and associates with one of the largest risks identified among cancer genome-wide association studies. We establish that the SNP has undergone positive selection throughout evolution, signifying a selective benefit, but go on to show that similar SNPs are rare in the genome due to negative selection, indicating that polymorphisms in p53-binding sites are primarily detrimental to humans.
Collapse
Affiliation(s)
- Jorge Zeron-Medina
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Xuting Wang
- Environmental Genomics Group, Laboratory of Molecular Genetics, National Institute of Environmental Health Sciences-National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Emmanouela Repapi
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Michelle R. Campbell
- Environmental Genomics Group, Laboratory of Molecular Genetics, National Institute of Environmental Health Sciences-National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Dan Su
- Environmental Genomics Group, Laboratory of Molecular Genetics, National Institute of Environmental Health Sciences-National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Francesc Castro-Giner
- Molecular and Population Genetics Laboratory, The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Benjamin Davies
- Transgenic Technology Research Group, The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Elisabeth F.P. Peterse
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Natalia Sacilotto
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Graeme J. Walker
- Skin Carcinogenesis Laboratory, Queensland Institute of Medical Research, Herston, QLD 4006, Australia
| | - Tamara Terzian
- Department of Dermatology, University of Colorado Denver, Aurora, CO 80045, USA
- Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado Denver, Aurora, CO 80045, USA
| | - Ian P. Tomlinson
- Molecular and Population Genetics Laboratory, The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Neil F. Box
- Department of Dermatology, University of Colorado Denver, Aurora, CO 80045, USA
- Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado Denver, Aurora, CO 80045, USA
| | - Nicolai Meinshausen
- Department of Statistics, University of Oxford, 1 South Parks Road, Oxford OX1 3TG, UK
| | - Sarah De Val
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Douglas A. Bell
- Environmental Genomics Group, Laboratory of Molecular Genetics, National Institute of Environmental Health Sciences-National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Gareth L. Bond
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| |
Collapse
|
65
|
Unravelling mechanisms of cisplatin sensitivity and resistance in testicular cancer. Expert Rev Mol Med 2013; 15:e12. [PMID: 24074238 DOI: 10.1017/erm.2013.13] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Testicular cancer is the most frequent solid malignant tumour type in men 20-40 years of age. At the time of diagnosis up to 50% of the patients suffer from metastatic disease. In contrast to most other metastatic solid tumours, the majority of metastatic testicular cancer patients can be cured with highly effective cisplatin-based chemotherapy. This review aims to summarise the current knowledge on response to chemotherapy and the biological basis of cisplatin-induced apoptosis in testicular cancer. The frequent presence of wild-type TP53 and the low levels of p53 in complex with the p53 negative feed-back regulator MDM2 contribute to cisplatin sensitivity. Moreover, the high levels of the pluripotency regulator Oct4 and as a consequence of Oct4 expression high levels of miR-17/106b seed family and pro-apoptotic Noxa and the low levels of cytoplasmic p21 (WAF1/Cip1) appear to be causative for the exquisite sensitivity to cisplatin-based therapy of testicular cancer. However, resistance of testicular cancer to cisplatin-based therapy does occur and can be mediated through aberrant levels of the above mentioned key players. Drugs targeting these key players showed, at least pre-clinically, a sensitising effect to cisplatin treatment. Further clinical development of such treatment strategies will lead to new treatment options for platinum-resistant testicular cancers.
Collapse
|
66
|
Harrington JA, Wheeler GM, Sweeting MJ, Mander AP, Jodrell DI. Adaptive designs for dual-agent phase I dose-escalation studies. Nat Rev Clin Oncol 2013; 10:277-88. [PMID: 23507740 DOI: 10.1038/nrclinonc.2013.35] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Anticancer agents used in combination are fundamental to successful cancer treatment, particularly in a curative setting. For dual-agent phase I trials, the goal is to identify drug doses and schedules for further clinical testing. However, current methods for establishing the recommended phase II dose for agents in combination can fail to fully explore drug interactions. With increasing numbers of anticancer drugs requiring testing, new adaptive model-based trial designs that improve on current practice have been proposed, although uptake has been minimal. We describe the methods available and discuss some of the opportunities and challenges faced in dual-agent phase I trials, as well as giving examples of trials in which adaptive designs have been implemented successfully. Improving the design and execution of phase I trials of drug combinations critically relies on collaboration between the statistical and clinical communities to facilitate the implementation of adaptive, model-based designs.
Collapse
Affiliation(s)
- Jennifer A Harrington
- Department of Oncology, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK.
| | | | | | | | | |
Collapse
|
67
|
Gutekunst M, Mueller T, Weilbacher A, Dengler MA, Bedke J, Kruck S, Oren M, Aulitzky WE, van der Kuip H. Cisplatin Hypersensitivity of Testicular Germ Cell Tumors Is Determined by High Constitutive Noxa Levels Mediated by Oct-4. Cancer Res 2013; 73:1460-9. [DOI: 10.1158/0008-5472.can-12-2876] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
68
|
Sears RB, Joyce LE, Ojaimi M, Gallucci JC, Thummel RP, Turro C. Photoinduced ligand exchange and DNA binding of cis-[Ru(phpy)(phen)(CH3CN)2]+ with long wavelength visible light. J Inorg Biochem 2012; 121:77-87. [PMID: 23353083 DOI: 10.1016/j.jinorgbio.2012.12.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Revised: 12/02/2012] [Accepted: 12/07/2012] [Indexed: 01/28/2023]
Abstract
The complex cis-[Ru(phpy)(phen)(CH3CN)2](+) (phpy=2-phenylpyridine, phen=1,10-phenanthroline) was investigated as a potential photodynamic therapy (PDT) agent. This complex presents desirable photochemical characteristics including a low energy absorption tail extending into the PDT window (600-850nm) and photoinduced exchange of the CH3CN ligands, generating a species analogous to the chemotherapy drug cisplatin. Furthermore, photochemical reactivity can be controlled through selective irradiation into the Ru-phen singlet metal-to-ligand charge transfer ((1)MLCT) band (λirr=500 nm) of [Ru(phpy)(phen)(CH3CN)2](+) in the presence of excess t-butylammonium chloride (TBACl) resulting in efficient photoinduced production of [Ru(phpy)(phen)(CH3CN)Cl] (Φ=0.25). This lower energy irradiation resulted in greater quantum yield of photosubstitution when compared to direct irradiation into the Ru-phpy (1)MLCT peak (λirr=450 nm; Φ=0.08) in CH2Cl2. It was found that the lower quantum yield observed for irradiation into the Ru→phpy(-)(1)MLCT band results from significant orbital mixing of the phpy(-) ligand with the t2g-type filled set in the metal, giving this state significant ligand-centered character. Lastly, this complex produced a decrease in the mobility of linearized ds-DNA when irradiated with λirr≥420nm, indicative of covalent binding by the transition metal complex similar to that observed for cisplatin. No change in mobility was found for the same samples kept in the dark indicating, unlike cisplatin, DNA binding of cis-[Ru(phpy)(phen)(CH3CN)2](+) only occurs with the activation of light. These observations support the use of cis-[Ru(phpy)(phen)(CH3CN)2](+) as a potential PDT agent by the photoinduced generation of a cisplatin analog.
Collapse
Affiliation(s)
- R Bryan Sears
- Department of Chemistry, The Ohio State University, Columbus, OH 43210, USA
| | | | | | | | | | | |
Collapse
|
69
|
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 2012; 7:e51563. [PMID: 23251575 PMCID: PMC3520950 DOI: 10.1371/journal.pone.0051563] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2012] [Accepted: 11/01/2012] [Indexed: 02/02/2023] Open
Abstract
Testicular Germ Cell Tumors (TGCT) and patient-derived cell lines are extremely sensitive to cisplatin and other interstrand cross-link (ICL) inducing agents. Nevertheless, a subset of TGCTs are either innately resistant or acquire resistance to cisplatin during treatment. Understanding the mechanisms underlying TGCT sensitivity/resistance to cisplatin as well as the identification of novel strategies to target cisplatin-resistant TGCTs have major clinical implications. Herein, we have examined the proficiency of five embryonal carcinoma (EC) cell lines to repair cisplatin-induced ICLs. Using γH2AX staining as a marker of double strand break formation, we found that EC cell lines were either incapable of or had a reduced ability to repair ICL-induced damage. The defect correlated with reduced Homologous Recombination (HR) repair, as demonstrated by the reduction of RAD51 foci formation and by direct evaluation of HR efficiency using a GFP-reporter substrate. HR-defective tumors cells are known to be sensitive to the treatment with poly(ADP-ribose) polymerase (PARP) inhibitor. In line with this observation, we found that EC cell lines were also sensitive to PARP inhibitor monotherapy. The magnitude of sensitivity correlated with HR-repair reduced proficiency and with the expression levels and activity of PARP1 protein. In addition, we found that PARP inhibition strongly enhanced the response of the most resistant EC cells to cisplatin, by reducing their ability to overcome the damage. These results point to a reduced proficiency of HR repair as a source of sensitivity of ECs to ICL-inducing agents and PARP inhibitor monotherapy, and suggest that pharmacological inhibition of PARP can be exploited to target the stem cell component of the TGCTs (namely ECs) and to enhance the sensitivity of cisplatin-resistant TGCTs to standard treatments.
Collapse
|
70
|
Fenske AE, Glaesener S, Bokemeyer C, Thomale J, Dahm-Daphi J, Honecker F, Dartsch DC. Cisplatin resistance induced in germ cell tumour cells is due to reduced susceptibility towards cell death but not to altered DNA damage induction or repair. Cancer Lett 2012; 324:171-8. [DOI: 10.1016/j.canlet.2012.05.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Revised: 05/09/2012] [Accepted: 05/11/2012] [Indexed: 11/26/2022]
|
71
|
Abstract
AbstractTesticular cancer is rare but is the most common cancer in males between 15 and 34 years of age. Two principal types of testicular cancer are distinguished: seminomas and non-seminomas. If detected early, the overall cure rate for testicular cancer exceeds 90%. In this study, artificial neural network (ANN) analysis as a prognostic tool was demonstrated regard to five year recurrence after the non-seminoma treatment. Data from 202 patients treated for non-seminoma were available for evaluation and comparison. A total of 32 variables were analysed using the ANN. The ANN approach, as an advanced multivariate data processing method, was demon-strated to provide objective prognostic data. Some of these prognostic factors are consistent or even imperceptible with previously evaluated by other statistical methods.
Collapse
|
72
|
Chui MH. Insights into cancer metastasis from a clinicopathologic perspective: Epithelial-Mesenchymal Transition is not a necessary step. Int J Cancer 2012; 132:1487-95. [PMID: 22833228 DOI: 10.1002/ijc.27745] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2012] [Revised: 06/25/2012] [Accepted: 07/10/2012] [Indexed: 12/12/2022]
Abstract
Epithelial-mesenchymal transition (EMT) has been implicated as the critical event initiating cancer invasion and metastasis. After disseminating through the circulation, the malignant cells have been proposed to undergo subsequent mesenchymal-epithelial transition (MET) to form secondary tumors. However, strong evidence from human tumor specimens for this paradigm is lacking. In carcinomas, cancers derived from epithelial tissues, epithelial morphology and gene expression are always retained to some degree. While mesenchymal transdifferentiation may be involved in the pathogenesis of carcinosarcomas, even in these neoplasms, as well as in germ cell tumors capable of multilineage differentiation, the mesenchymal phenotype does not facilitate metastatic progression. Indeed, most cancers invade and travel through lymphatic and blood vessels via cohesive epithelial migration, rather than going through the EMT-MET sequence. EMT gene expression is also consistently associated with high histologic grade and while the transcription factors, Snail, Slug and Twist have traditionally been thought of as inducers of EMT, under certain conditions, they also mediate dedifferentiation and maintenance of the stem cell state. In various malignancies, including basal-like breast cancer and colorectal cancer, the genetically unstable, undifferentiated phenotype predicts early metastatic spread and poor prognosis. This article discusses some of the controversies surrounding differentiation and metastasis from a clinicopathologic perspective and presents evidence that the epithelial phenotype is maintained throughout the process of cancer metastasis.
Collapse
Affiliation(s)
- Michael Herman Chui
- Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, Canada.
| |
Collapse
|
73
|
Abstract
Proteins in the HMG family are important transcription factors. They recognize cisplatin-damaged DNA lesions with a structure-specific preference and account for more than 70% of all proteins that interact with the cisplatin 1,2-intrastrand d(GpG) cross-link. HMGB4, a new member of the mammalian HMGB protein family expressed preferentially in the testis, was generated recombinantly, and its interactions with cisplatin-modified DNA were investigated in vitro. The binding affinities of the two individual DNA-binding domains of HMGB4 to DNA carrying a cisplatin 1,2-intrastrand d(GpG) cross-link are weaker than those of the DNA-binding domains of HMGB1. Full-length HMGB4, however, has a 28-fold stronger binding affinity (K(d) = 4.35 nM) for the platinated adduct compared to that of HMGB1 (K(d) = 120 nM), presumably because the former lacks a C-terminal acidic tail. The residue Phe37 plays a critical role in stabilizing the binding complex of HMGB4 with the cisplatin-modified DNA, as it does for HMGB1. Hydroxyl radical footprinting analysis of the HMGB4/platinated DNA complex reveals a footprinting pattern very different from that of HMGB1, however, revealing very little binding asymmetry with respect to the platinated lesion. An in vitro repair assay revealed that HMGB4, at 1 μM, interferes with repair of cisplatin 1,2-intrastrand cross-link damage by >90% compared to control, whereas HMGB1 at the same concentration inhibits repair by 45%. This repair inhibition capability is highly dependent on both the binding affinity and the size of the proteins. The putative role of HMGB4 in the mechanism of action of cisplatin, and especially its potential relevance to the hypersensitivity of testicular germ cell tumors to cisplatin, are discussed.
Collapse
Affiliation(s)
- Semi Park
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139-4307, USA
| | | |
Collapse
|
74
|
Ushida H, Chano T, Minami K, Kita H, Kawakami T, Okabe H, Okada Y, Okamoto K. Therapeutic Potential of SOX2 Inhibition for Embryonal Carcinoma. J Urol 2012; 187:1876-81. [DOI: 10.1016/j.juro.2011.12.058] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2011] [Indexed: 01/26/2023]
Affiliation(s)
- Hiroshi Ushida
- Department of Urology, Shiga University of Medical Science, Shiga, Japan
| | - Tokuhiro Chano
- Department of Clinical Laboratory Medicine, Shiga University of Medical Science, Shiga, Japan
| | - Kahori Minami
- Department of Clinical Laboratory Medicine, Shiga University of Medical Science, Shiga, Japan
| | - Hiroko Kita
- Department of Clinical Laboratory Medicine, Shiga University of Medical Science, Shiga, Japan
| | - Takahiro Kawakami
- Department of Clinical Laboratory Medicine, Shiga University of Medical Science, Shiga, Japan
| | - Hidetoshi Okabe
- Department of Clinical Laboratory Medicine, Shiga University of Medical Science, Shiga, Japan
| | - Yusaku Okada
- Department of Urology, Shiga University of Medical Science, Shiga, Japan
| | - Keisei Okamoto
- Department of Urology, Shiga University of Medical Science, Shiga, Japan
| |
Collapse
|
75
|
Vermeulen L, de Sousa e Melo F, Richel DJ, Medema JP. The developing cancer stem-cell model: clinical challenges and opportunities. Lancet Oncol 2012; 13:e83-9. [PMID: 22300863 DOI: 10.1016/s1470-2045(11)70257-1] [Citation(s) in RCA: 267] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
During the past decade, a stem-cell-like subset of cancer cells has been identified in many malignancies. These cells, referred to as cancer stem cells (CSCs), are of particular interest because they are believed to be the clonogenic core of the tumour and therefore represent the cell population that drives growth and progression. Many efforts have been made to design therapies that specifically target the CSC population, since this was predicted to be the crucial population to eliminate. However, recent insights have complicated the initial elegant model, by showing a dominant role for the tumour microenvironment in determining CSC characteristics within a malignancy. This is particularly important since dedifferentiation of non-tumorigenic tumour cells towards CSCs can occur, and therefore the CSC population in a neoplasm is expected to vary over time. Moreover, evidence suggests that not all tumours are driven by rare CSCs, but might instead contain a large population of tumorigenic cells. Even though these results suggest that specific targeting of the CSC population might not be a useful therapeutic strategy, research into the hierarchical cellular organisation of malignancies has provided many important new insights in the biology of tumours. In this Personal View, we highlight how the CSC concept is developing and influences our thinking on future treatment for solid tumours, and recommend ways to design clinical trials to assess drugs that target malignant disease in a rational fashion.
Collapse
Affiliation(s)
- Louis Vermeulen
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental Molecular Medicine, Academic Medical Center, Amsterdam, Netherlands.
| | | | | | | |
Collapse
|
76
|
Okamoto K. Epigenetics: A way to understand the origin and biology of testicular germ cell tumors. Int J Urol 2012; 19:504-11. [DOI: 10.1111/j.1442-2042.2012.02986.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
77
|
Abstract
PURPOSE OF REVIEW Male infertility impacts a substantial proportion of men and has serious implication for a man's quality of life. Advances in reproductive technology may allow men to bypass urologic care in order to achieve their family planning goals. Recent data suggests that male reproductive failure may be a harbinger of future urologic diseases, including prostate cancer (CaP), thus emphasizing the importance of dedicated urologic evaluation and care for all male infertility patients. RECENT FINDINGS We will review the epidemiologic data that explores an association between male reproductive health and CaP. We will review the potential biologic mechanisms that may underlie this association, and explore possible reasons for inconsistencies in study findings. SUMMARY Studies of the association between male infertility and CaP are inconsistent. Despite this, the association between reproductive health in a man's fourth decade (30s) and his development of aggressive CaP in his sixth decade (50s) should not be ignored. These findings, combined with the robustness of the potential common underlying mechanisms, provide a foundation for future studies of male reproductive health that are more specific in their approach to answering questions about the association between male reproductive failure and future systemic disease.
Collapse
Affiliation(s)
- Thomas J Walsh
- Department of Urology, University of Washington School of Medicine, Seattle, Washington, USA.
| |
Collapse
|
78
|
Rahman M, Deleyrolle L, Vedam-Mai V, Azari H, Abd-El-Barr M, Reynolds BA. The cancer stem cell hypothesis: failures and pitfalls. Neurosurgery 2011; 68:531-45; discussion 545. [PMID: 21135745 DOI: 10.1227/neu.0b013e3181ff9eb5] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Based on the clonal evolution model and the assumption that the vast majority of tumor cells are able to propagate and drive tumor growth, the goal of cancer treatment has traditionally been to kill all cancerous cells. This theory has been challenged recently by the cancer stem cell (CSC) hypothesis, that a rare population of tumor cells, with stem cell characteristics, is responsible for tumor growth, resistance, and recurrence. Evidence for putative CSCs has been described in blood, breast, lung, prostate, colon, liver, pancreas, and brain. This new hypothesis would propose that indiscriminate killing of cancer cells would not be as effective as selective targeting of the cells that are driving long-term growth (ie, the CSCs) and that treatment failure is often the result of CSCs escaping traditional therapies.The CSC hypothesis has gained a great deal of attention because of the identification of a new target that may be responsible for poor outcomes of many aggressive cancers, including malignant glioma. As attractive as this hypothesis sounds, especially when applied to tumors that respond poorly to current treatments, we will argue in this article that the proposal of a stemlike cell that initiates and drives solid tissue cancer growth and is responsible for therapeutic failure is far from proven. We will present the point of view that for most advanced solid tissue cancers such as glioblastoma multiforme, targeting a putative rare CSC population will have little effect on patient outcomes. This review will cover problems with the CSC hypothesis, including applicability of the hierarchical model, inconsistencies with xenotransplantation data, and nonspecificity of CSC markers.
Collapse
Affiliation(s)
- Maryam Rahman
- Department of Neurosurgery, University of Florida, Gainesville, Florida, USA.
| | | | | | | | | | | |
Collapse
|
79
|
Abstract
Testicular cancer, though generally uncommon, is the most common cancer NPs will see in young men. Advances in surgery, radiation, and cisplatin-based chemotherapy, have made testicular cancer a "model" for a curable cancer. Survivors will likely live many years, but will require lifelong follow-up for possible recurrence and long-term treatment sequelae.
Collapse
Affiliation(s)
- Barbara H Zoltick
- Division of Hematology/Oncology, University of Pennsylvania Health System, Philadelphia, PA, USA
| |
Collapse
|
80
|
Third-Line Chemotherapy and Novel Agents for Metastatic Germ Cell Tumors. Hematol Oncol Clin North Am 2011; 25:577-91, ix. [DOI: 10.1016/j.hoc.2011.03.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
81
|
LeBron C, Pal P, Brait M, Dasgupta S, Guerrero-Preston R, Looijenga LH, Kowalski J, Netto G, Hoque MO. Genome-wide analysis of genetic alterations in testicular primary seminoma using high resolution single nucleotide polymorphism arrays. Genomics 2011; 97:341-9. [DOI: 10.1016/j.ygeno.2011.02.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Revised: 01/11/2011] [Accepted: 02/25/2011] [Indexed: 11/26/2022]
|
82
|
|
83
|
Abstract
Over the last decade, the notion that tumors are maintained by their own stem cells, the so-called cancer stem cells, has created great excitement in the research community. This review attempts to summarize the underlying concepts of this notion, to distinguish hard facts from beliefs and to define the future challenges of the field.
Collapse
|
84
|
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: 3.9] [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
|
85
|
Gutekunst M, Oren M, Weilbacher A, Dengler MA, Markwardt C, Thomale J, Aulitzky WE, van der Kuip H. p53 hypersensitivity is the predominant mechanism of the unique responsiveness of testicular germ cell tumor (TGCT) cells to cisplatin. PLoS One 2011; 6:e19198. [PMID: 21532991 PMCID: PMC3080918 DOI: 10.1371/journal.pone.0019198] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2010] [Accepted: 03/29/2011] [Indexed: 12/20/2022] Open
Abstract
Consistent with the excellent clinical results in testicular germ cell tumors
(TGCT), most cell lines derived from this cancer show an exquisite sensitivity
to Cisplatin. It is well accepted that the high susceptibility of TGCT cells to
apoptosis plays a central role in this hypersensitive phenotype. The role of the
tumor suppressor p53 in this response, however, remains controversial. Here we
show that siRNA-mediated silencing of p53 is sufficient to completely abrogate
hypersensitivity not only to Cisplatin but also to non-genotoxic inducers of p53
such as the Mdm2 antagonist Nutlin-3 and the proteasome inhibitor Bortezomib.
The close relationship between p53 protein levels and induction of apoptosis is
lost upon short-term differentiation, indicating that this predominant
pro-apoptotic function of p53 is unique in pluripotent embryonal carcinoma (EC)
cells. RNA interference experiments as well as microarray analysis demonstrated
a central role of the pro-apoptotic p53 target gene NOXA in the p53-dependent
apoptotic response of these cells. In conclusion, our data indicate that the
hypersensitivity of TGCT cells is a result of their unique sensitivity to p53
activation. Furthermore, in the very specific cellular context of germ
cell-derived pluripotent EC cells, p53 function appears to be limited to
induction of apoptosis.
Collapse
Affiliation(s)
- Matthias Gutekunst
- Dr Margarete-Fischer-Bosch Institute of Clinical Pharmacology and
University of Tuebingen, Stuttgart, Germany
| | - Moshe Oren
- Department of Molecular Cell Biology, Weizmann Institute of Science,
Rehovot, Israel
| | - Andrea Weilbacher
- Dr Margarete-Fischer-Bosch Institute of Clinical Pharmacology and
University of Tuebingen, Stuttgart, Germany
| | - Michael A. Dengler
- Dr Margarete-Fischer-Bosch Institute of Clinical Pharmacology and
University of Tuebingen, Stuttgart, Germany
| | - Christiane Markwardt
- Dr Margarete-Fischer-Bosch Institute of Clinical Pharmacology and
University of Tuebingen, Stuttgart, Germany
| | - Jürgen Thomale
- Institute for Cell Biology, University of Duisburg-Essen Medical School,
Essen, Germany
| | - Walter E. Aulitzky
- 2 Department of Internal Medicine, Robert-Bosch-Hospital, Stuttgart,
Germany
| | - Heiko van der Kuip
- Dr Margarete-Fischer-Bosch Institute of Clinical Pharmacology and
University of Tuebingen, Stuttgart, Germany
- * E-mail:
| |
Collapse
|
86
|
Mao P, Hever MP, Niemaszyk LM, Haghkerdar JM, Yanco EG, Desai D, Beyrouthy MJ, Kerley-Hamilton JS, Freemantle SJ, Spinella MJ. Serine/threonine kinase 17A is a novel p53 target gene and modulator of cisplatin toxicity and reactive oxygen species in testicular cancer cells. J Biol Chem 2011; 286:19381-91. [PMID: 21489989 DOI: 10.1074/jbc.m111.218040] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Testicular cancer is highly curable with cisplatin-based therapy, and testicular cancer-derived human embryonal carcinoma (EC) cells undergo a p53-dominant transcriptional response to cisplatin. In this study, we have discovered that a poorly characterized member of the death-associated protein family of serine/threonine kinases, STK17A (also called DRAK1), is a novel p53 target gene. Cisplatin-mediated induction of STK17A in the EC cell line NT2/D1 was prevented with p53 siRNA. Furthermore, STK17A was induced with cisplatin in HCT116 and MCF10A cells but to a much lesser extent in isogenic p53-suppressed cells. A functional p53 response element that binds endogenous p53 in a cisplatin-dependent manner was identified 5 kb upstream of the first coding exon of STK17A. STK17A is not present in the mouse genome, but the closely related gene STK17B is induced with cisplatin in mouse NIH3T3 cells, although this induction is p53-independent. Interestingly, in human cells containing both STK17A and STK17B, only STK17A is induced with cisplatin. Knockdown of STK17A conferred resistance to cisplatin-induced growth suppression and apoptotic cell death in EC cells. This was associated with the up-regulation of detoxifying and antioxidant genes, including metallothioneins MT1H, MT1M, and MT1X that have previously been implicated in cisplatin resistance. In addition, knockdown of STK17A resulted in decreased cellular reactive oxygen species, whereas STK17A overexpression increased reactive oxygen species. In summary, we have identified STK17A as a novel direct target of p53 and a modulator of cisplatin toxicity and reactive oxygen species in testicular cancer cells.
Collapse
Affiliation(s)
- Pingping Mao
- Department of Pharmacology and Toxicology, Dartmouth Medical School, Hanover, New Hampshire 03755, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
87
|
Gilbert D, Rapley E, Shipley J. Testicular germ cell tumours: predisposition genes and the male germ cell niche. Nat Rev Cancer 2011; 11:278-88. [PMID: 21412254 DOI: 10.1038/nrc3021] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Testicular germ cell tumours (TGCTs) of adults and adolescents are putatively derived from primordial germ cells or gonocytes. Recently reported genome-wide association studies implicate six gene loci that predispose to TGCT development. Remarkably, the functions of proteins encoded by genes within these regions bridge our understanding between the pathways involved in primordial germ cell physiology, male germ cell development and the molecular pathology of TGCTs. Furthermore, this improved understanding of the mechanisms underlying TGCT development and dissemination has clinical relevance for the management of patients with these tumours.
Collapse
Affiliation(s)
- Duncan Gilbert
- Sussex Cancer Centre, Royal Sussex County Hospital, Eastern Road, Brighton BN2 5BE, East Sussex, UK
| | | | | |
Collapse
|
88
|
Lu X, Kang Y. Cell fusion hypothesis of the cancer stem cell. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 714:129-40. [PMID: 21506011 DOI: 10.1007/978-94-007-0782-5_6] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
A major advance in recent cancer research is the identification of tumor cells with stem cell-like properties. Cancer stem cells (CSCs) often represent a rare population in the tumor mass and possess the exclusive ability to initiate the growth of a heterogeneous tumor. The origin of CSCs remains elusive and is likely to be cancer type specific. One possible but under-appreciated potential mechanism for the generation of CSCs is through fusion between stem cells and differentiated cells. The cell fusion hypothesis of CSCs adds an important functional underpinning to the potential multifaceted roles of cell fusion in the initiation and progression of cancer.
Collapse
Affiliation(s)
- Xin Lu
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA.
| | | |
Collapse
|
89
|
Usanova S, Piée-Staffa A, Sied U, Thomale J, Schneider A, Kaina B, Köberle B. Cisplatin sensitivity of testis tumour cells is due to deficiency in interstrand-crosslink repair and low ERCC1-XPF expression. Mol Cancer 2010; 9:248. [PMID: 20846399 PMCID: PMC3098011 DOI: 10.1186/1476-4598-9-248] [Citation(s) in RCA: 120] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2010] [Accepted: 09/16/2010] [Indexed: 12/11/2022] Open
Abstract
Background Cisplatin based chemotherapy cures over 80% of metastatic testicular germ cell tumours (TGCT). In contrast, almost all other solid cancers in adults are incurable once they have spread beyond the primary site. Cell lines derived from TGCTs are hypersensitive to cisplatin reflecting the clinical response. Earlier findings suggested that a reduced repair capacity might contribute to the cisplatin hypersensitivity of testis tumour cells (TTC), but the critical DNA damage has not been defined. This study was aimed at investigating the formation and repair of intrastrand and interstrand crosslinks (ICLs) induced by cisplatin in TTC and their contribution to TTC hypersensitivity. Results We observed that repair of intrastrand crosslinks is similar in cisplatin sensitive TTC and resistant bladder cancer cells, whereas repair of ICLs was significantly reduced in TTC. γH2AX formation, which serves as a marker of DNA breaks formed in response to ICLs, persisted in cisplatin-treated TTC and correlated with sustained phosphorylation of Chk2 and enhanced PARP-1 cleavage. Expression of the nucleotide excision repair factor ERCC1-XPF, which is implicated in the processing of ICLs, is reduced in TTC. To analyse the causal role of ERCC1-XPF for ICL repair and cisplatin sensitivity, we over-expressed ERCC1-XPF in TTC by transient transfection. Over-expression increased ICL repair and rendered TTC more resistant to cisplatin, which suggests that ERCC1-XPF is rate-limiting for repair of ICLs resulting in the observed cisplatin hypersensitivity of TTC. Conclusion Our data indicate for the first time that the exceptional sensitivity of TTC and, therefore, very likely the curability of TGCT rests on their limited ICL repair due to low level of expression of ERCC1-XPF.
Collapse
Affiliation(s)
- Svetlana Usanova
- Institute of Toxicology, Clinical Centre of University of Mainz, Obere Zahlbacher Strasse 67, 55131 Mainz, Germany
| | | | | | | | | | | | | |
Collapse
|
90
|
Turnbull C, Rapley EA, Seal S, Pernet D, Renwick A, Hughes D, Ricketts M, Linger R, Nsengimana J, Deloukas P, Huddart RA, Bishop DT, Easton DF, Stratton MR, Rahman N. Variants near DMRT1, TERT and ATF7IP are associated with testicular germ cell cancer. Nat Genet 2010; 42:604-7. [PMID: 20543847 PMCID: PMC3773909 DOI: 10.1038/ng.607] [Citation(s) in RCA: 270] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2010] [Accepted: 05/17/2010] [Indexed: 12/13/2022]
Abstract
We conducted a genome-wide association study for testicular germ cell tumor, genotyping 298,782 SNPs in 979 affected individuals and 4,947 controls from the UK and replicating associations in a further 664 cases and 3,456 controls. We identified three new susceptibility loci, two of which include genes that are involved in telomere regulation. We identified two independent signals within the TERT-CLPTM1L locus on chromosome 5, which has previously been associated with multiple other cancers (rs4635969, OR=1.54, P=1.14x10(-23); rs2736100, OR=1.33, P=7.55x10(-15)). We also identified a locus on chromosome 12 (rs2900333, OR=1.27, P=6.16x10(-10)) that contains ATF7IP, a regulator of TERT expression. Finally, we identified a locus on chromosome 9 (rs755383, OR=1.37, P=1.12x10(-23)), containing the sex determination gene DMRT1, which has been linked to teratoma susceptibility in mice.
Collapse
Affiliation(s)
- Clare Turnbull
- Section of Cancer Genetics, Institute of Cancer Research, Sutton, Surrey, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
91
|
Mueller T, Mueller LP, Holzhausen HJ, Witthuhn R, Albers P, Schmoll HJ. Histological evidence for the existence of germ cell tumor cells showing embryonal carcinoma morphology but lacking OCT4 expression and cisplatin sensitivity. Histochem Cell Biol 2010; 134:197-204. [PMID: 20532795 DOI: 10.1007/s00418-010-0710-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/12/2010] [Indexed: 10/19/2022]
Abstract
The biological basis for manifestation of chemotherapy resistance in metastatic testicular germ cell tumors (GCT) remains obscure and is of particular clinical interest. In nonseminomatous GCT (NSGCT) the pluripotent embryonal carcinoma (EC) cells are the precursors of the manifold differentiated structures but also drive the malignant growth. They are known to be hypersensitive towards DNA-damaging agents and to express the embryonal transcription factor OCT4. We recently characterized EC cells that lack OCT4 expression and show cisplatin resistance. In the present, immunohistochemical study we analyzed the composition of NSGCT with the focus on such OCT4-negative EC cells using a NSGCT xenograft model as well as patient-derived NSGCT samples. In the xenograft model, the cisplatin-sensitive cell line H12.1 gives rise to xenografts where EC structures are mainly composed of OCT4-positive cells, whereas xenografts from the resistant cell line 1411HP exclusively comprise OCT4-negative EC areas. We found that post-chemotherapy residual metastatic tumors of patients can be comprised of exclusively OCT4-negative EC, whereas the matched testicular primary tumor harbors OCT4-positive EC. Thorough histological analyses revealed a few examples of such OCT4-negative EC cells also in the testicular primary tumor as well as in xenografts from the cisplatin-sensitive NSGCT-cell line. For these cells we propose an identity as early extraembryonal progenitor cells directly derived from OCT4-expressing EC cells. This challenges the use of the term EC cell. The data also support our hypothesis that malignant growth of resistant NSGCT may be driven by this cell type.
Collapse
Affiliation(s)
- Thomas Mueller
- Department of Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, 06120 Halle, Germany.
| | | | | | | | | | | |
Collapse
|
92
|
Abstract
OCT4 is a transcription factor critical for the pluripotency of human embryonal stem (ES) and induced pluipotency stem (IPS) cells. OCT4 is commonly expressed in germ-cell tumors as well as putative cancer stem cells in several tumors, and is a key determinant of oncogenic fate in germ-cell tumors. The capacity of the human immune system to recognize this critical stem-cell gene is not known, but has implications for preventing tumors with ES/IPS-based therapies and targeting stem-cell pathways in cancer. Here we show that OCT4-specific T cells can be readily detected in freshly isolated T cells from most (>80%) healthy donors. The reactivity to OCT4-derived peptides resides primarily in the CD45RO(+) memory T-cell compartment and consists predominantly of CD4(+) T cells. T cells reactive against OCT4-derived peptides can be readily expanded in culture using peptide-loaded dendritic cells. In contrast to healthy donors, immunity to OCT4 was detected in only 35% of patients with newly diagnosed germ-cell tumors. However, chemotherapy of germ-cell tumors led to the induction of anti-OCT4 immunity in vivo in patients lacking such responses at baseline. These data demonstrate the surprising lack of immune tolerance to this critical pluripotency antigen in humans. Harnessing natural immunity to this antigen may allow immune-based targeting of pluripotency-related pathways for prevention of cancers, including those in the setting of ES/IPS-based therapies.
Collapse
|
93
|
Noel EE, Yeste-Velasco M, Mao X, Perry J, Kudahetti SC, Li NF, Sharp S, Chaplin T, Xue L, McIntyre A, Shan L, Powles T, Oliver RTD, Young BD, Shipley J, Berney DM, Joel SP, Lu YJ. The association of CCND1 overexpression and cisplatin resistance in testicular germ cell tumors and other cancers. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 176:2607-15. [PMID: 20395447 DOI: 10.2353/ajpath.2010.090780] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Development of chemoresistance limits the clinical efficiency of platinum-based therapy. Although many resistance mechanisms have been demonstrated, genetic/molecular alterations responsible for drug resistance in the majority of clinical cases have not been identified. We analyzed three pairs of testicular germ cell tumor cell lines using Affymetrix expression microarrays and revealed a limited number of differentially expressed genes across the cell lines when comparing the parental and resistant cells. Among them, CCND1 was the most significantly differentially expressed gene. Analysis of testicular germ cell tumor clinical samples by quantitative reverse transcription PCR analysis revealed that overall expression of CCND1 was significantly higher in resistant cases compared with sensitive samples (P < 0.0001). We also found that CCND1 was dramatically overexpressed both in induced and intrinsically resistant samples of ovarian and prostate cancer. Finally combined CCND1 knockdown using small-interfering RNA and cisplatin treatment inhibited cell growth in vitro significantly more effectively than any of these single treatments. Therefore, deregulation of CCND1 may be a major cause of cisplatin resistance in testicular germ cell tumors and may also be implicated in ovarian and prostate cancers. CCND1 could be potentially used as a marker for treatment stratification and as a molecular target to improve the treatment of platinum-resistant tumors.
Collapse
Affiliation(s)
- Elodie E Noel
- Medical Oncology Centre, Cancer Institute, Barts and London School of Medicine and Dentistry, Queen Mary, University of London, Charterhouse Square, London EC1M 6BQ, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
94
|
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: 5.6] [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
|
95
|
Li B, Cheng Q, Li Z, Chen J. p53 inactivation by MDM2 and MDMX negative feedback loops in testicular germ cell tumors. Cell Cycle 2010; 9:1411-20. [PMID: 20372076 DOI: 10.4161/cc.9.7.11255] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Testicular germ cell tumors (TGCT) are unique in their excellent response to DNA-damaging chemotherapy. Mutation of p53 is rare in both untreated and relapsed TGCTs, suggesting that p53 fails to respond effectively against malignant transformation in germ cells. Previous studies implicated the presence of a poorly defined TGCT-specific mechanism of p53 inactivation. Here we show that disruption of p53-MDM2 binding using the MDM2-specific inhibitor Nutlin activates p53 in TGCT cells and is sufficient to induce strong apoptosis. Knockdown of MDMX cooperates with Nutlin to activate p53. Surprisingly, we found that p53 activation induced a two-fold increase in MDMX mRNA and protein expression in TGCT cells. A p53-responsive promoter is identified in MDMX intron 1 that contains a functional p53-binding site, suggesting that MDMX also functions as a negative feedback regulator of p53 in a cell line-dependent fashion. These findings suggest that MDM2 and MDMX are responsible for the functional inactivation of p53 in TGCT. Furthermore, TGCT cells are unique in having a strong apoptosis response to p53. Direct activation of p53 by targeting MDM2 and MDMX may provide a backup approach for the treatment of TGCTs resistant to DNA-damaging drugs.
Collapse
Affiliation(s)
- Baozong Li
- Molecular Oncology Department, Moffitt Cancer Center, Tampa, FL, USA
| | | | | | | |
Collapse
|
96
|
Piulats JM, Jiménez L, García del Muro X, Villanueva A, Viñals F, Germà-Lluch JR. Molecular mechanisms behind the resistance of cisplatin in germ cell tumours. Clin Transl Oncol 2010; 11:780-6. [PMID: 20045784 DOI: 10.1007/s12094-009-0446-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Cisplatin has been one of the principal chemotherapy agents for the last 30 years and is still used widely in the treatment of testicular, ovarian, lung, head and neck, bladder and several other tumours. Resistance to chemotherapeutic agents is a major obstacle for successful treatment. Treatment effect on germ cell tumours (GCTs) is more successful than in adults suffering from almost any other solid tumour, but resistance still appears in 20% of patients with metastatic disease. However, because of the young age of patients and few data regarding the process of becoming resistant, this situation is still a challenge. In this review we are going to analyse the published literature on cisplatin resistance in GCTs and explain the initiatives that the Spanish Germ Cell Cancer Group (GG) is taking to try to elucidate the molecular mechanisms behind this process.
Collapse
Affiliation(s)
- Josep Ma Piulats
- Medical Oncology Department, IDIBELL Institut Català d'Oncologia, Hospital Duran i Reynals, L'Hospitalet de Llobregat, Barcelona, Spain
| | | | | | | | | | | |
Collapse
|
97
|
Korkola JE, Houldsworth J, Bosl GJ, Chaganti RSK. Molecular events in germ cell tumours: linking chromosome-12 gain, acquisition of pluripotency and response to cisplatin. BJU Int 2009; 104:1334-8. [PMID: 19840009 DOI: 10.1111/j.1464-410x.2009.08855.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Germ cell tumours (GCTs) represent the leading cause of cancer-related morbidity and mortality in young men aged 18-35 years. Transformation of the cell of origin results in tumours with several unique properties. GCTs are characterized by gain of the short arm of chromosome 12 in almost all cases, a frequency of genomic alteration not seen in any other solid tumours. GCTs are truly pluripotent, giving rise to cells of somatic and extra-embryonic lineages, which results in tumours with a spectrum of differentiation that rivals that seen in normal embryogenesis and development. Despite the presence of genomic instability and many oncogenic changes, GCTs are highly curable, even in the metastatic setting, due to their extreme sensitivity to cisplatin-based chemotherapy. In this review we highlight some of the molecular events associated with the genesis, differentiation and chemotherapeutic response of these tumours, and discuss how these alterations are linked with biological features unique to germ cells.
Collapse
Affiliation(s)
- James E Korkola
- Cell Biology Division, Memorial Sloan-Kettering Cancer Center, New York, NY, USA.
| | | | | | | |
Collapse
|
98
|
Perry J, Powles T, Shamash J, Veerupillai A, McGrowder E, Noel E, Lu YJ, Oliver T, Joel S. The relative activity of cisplatin, oxaliplatin and satraplatin in testicular germ cell tumour sensitive and resistant cell lines. Cancer Chemother Pharmacol 2009; 64:925-33. [PMID: 19263053 DOI: 10.1007/s00280-009-0944-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2008] [Accepted: 01/20/2009] [Indexed: 10/21/2022]
Abstract
BACKGROUND Germ cell tumours (GCT) can become resistant to cisplatin, which is associated with a relatively poor prognosis. Oxaliplatin and satraplatin have been developed to overcome cisplatin resistance in other cancers, but their effect in cisplatin resistant (cisR) GCTs is unclear. In this work we address this issue by comparing their efficacy in three paired sensitive and cisR GCT cell lines. METHODS Three established cisplatin sensitive (cisS) and resistant cell line pairs were used (GCT27, GCT27r: SUSA, SUSAr: 833k, 833kr). Viability was assessed using a luciferase based ATP assay and EC(50) and EC(80) concentrations were calculated. Western blot analysis and flow cytometry was used for further assessment. RESULTS Sensitivity to the three platinum compounds was broadly similar in the three cisS lines GCT cell lines (EC(50) = 0.27-0.51 microM for cisplatin, 0.52-0.79 microM for oxaliplatin, 0.31-1.26 microM for satraplatin). EC(50) values for cisplatin in the three cisR sub lines were 1.8- to 3.8-fold higher than in the sensitive parental lines. Cross resistance to satraplatin and oxaliplatin occurred in all three cisR cell lines (resistance factor 1.9-4.4), with the exception of oxaliplatin in the 833Kr (resistance factor 0.9). Differences in the effect of specific drugs on cell cycle distribution, p53, p21 and MDM2 were observed. CONCLUSIONS These data suggest that satraplatin and oxaliplatin could theoretically be used in chemo-naive GCTs and support the further clinical evaluation of these agents in this setting. The mechanism of cross resistance to these drugs appears multifactorial.
Collapse
Affiliation(s)
- Jackie Perry
- Orchid Cancer Pharmacology Group, Centre for Experimental Cancer Medicine, Institute of Cancer, St Bartholomew's Hospital, West Smithfield, London, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
99
|
Shackleton M, Quintana E, Fearon ER, Morrison SJ. Heterogeneity in cancer: cancer stem cells versus clonal evolution. Cell 2009; 138:822-9. [PMID: 19737509 DOI: 10.1016/j.cell.2009.08.017] [Citation(s) in RCA: 802] [Impact Index Per Article: 53.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The identification and characterization of cancer stem cells might lead to more effective treatments for some cancers by focusing therapy on the most malignant cells. To achieve this goal it will be necessary to determine which cancers follow a cancer stem cell model and which do not, to address technical issues related to tumorigenesis assays, and to test the extent to which cancer cell heterogeneity arises from genetic versus epigenetic differences.
Collapse
Affiliation(s)
- Mark Shackleton
- Howard Hughes Medical Institute, Life Sciences Institute, Center for Stem Cell Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | | | | | | |
Collapse
|
100
|
Korkola JE, Houldsworth J, Feldman DR, Olshen AB, Qin LX, Patil S, Reuter VE, Bosl GJ, Chaganti RSK. Identification and validation of a gene expression signature that predicts outcome in adult men with germ cell tumors. J Clin Oncol 2009; 27:5240-7. [PMID: 19770384 DOI: 10.1200/jco.2008.20.0386] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Germ cell tumor (GCT) is the most common malignancy in young adult men. Currently, patients are risk-stratified on the basis of clinical presentation and serum tumor markers. The introduction of molecular markers could improve outcome prediction. PATIENTS AND METHODS Expression profiling was performed on 74 nonseminomatous GCTs (NSGCTs) from cisplatin-treated patients (ie, training set) and on 34 similarly treated patients with NSGCTs (ie, validation set). A gene classifier was developed by using prediction analysis for microarrays (PAM) for the binary end point of 5-year overall survival (OS). A predictive score was developed for OS by using the univariate Cox model. RESULTS In the training set, PAM identified 140 genes that predicted 5-year OS (cross-validated classification rate, 60%). The PAM model correctly classified 90% of patients in the validation set. Patients predicted to have good outcome had significantly longer survival than those with poor predicted outcome (P < .001). For the OS end point, a 10-gene model had a predictive accuracy (ie, concordance index) of 0.66 in the training set and a concordance index of 0.83 in the validation set. Dichotomization of the samples on the basis of the median score resulted in significant differences in survival (P = .002). For both end points, the gene-based predictor was an independent prognostic factor in a multivariate model that included clinical risk stratification (P < .01 for both). CONCLUSION We have identified gene expression signatures that accurately predict outcome in patients with GCTs. These predictive genes should be useful for the prediction of patient outcome and could provide novel targets for therapeutic intervention.
Collapse
Affiliation(s)
- James E Korkola
- Cell Biology Program, Sloan-Kettering Institute for Cancer Research, New York, USA
| | | | | | | | | | | | | | | | | |
Collapse
|