Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-x24gv Total loading time: 0 Render date: 2024-06-08T02:39:05.937Z Has data issue: false hasContentIssue false

54 - Molecular targets for epithelial ovarian cancer

from Part 3.1 - Molecular pathology: carcinomas

Published online by Cambridge University Press:  05 February 2015

Grace K. Suh
Affiliation:
Departments of Experimentalherapeutics, Gynecologic Medical Oncology, Bioinformatics and Computational Biology, and Systems Biology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
Bryan T. Hennessy
Affiliation:
Departments of Experimentalherapeutics, Gynecologic Medical Oncology, Bioinformatics and Computational Biology, and Systems Biology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
Roeland Verhaak
Affiliation:
Departments of Experimentalherapeutics, Gynecologic Medical Oncology, Bioinformatics and Computational Biology, and Systems Biology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
Ji-Yeon Yang
Affiliation:
Departments of Experimentalherapeutics, Gynecologic Medical Oncology, Bioinformatics and Computational Biology, and Systems Biology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
Gordon B. Mills
Affiliation:
Departments of Experimentalherapeutics, Gynecologic Medical Oncology, Bioinformatics and Computational Biology, and Systems Biology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
Robert C. Bast
Affiliation:
Departments of Experimentalherapeutics, Gynecologic Medical Oncology, Bioinformatics and Computational Biology, and Systems Biology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
Edward P. Gelmann
Affiliation:
Columbia University, New York
Charles L. Sawyers
Affiliation:
Memorial Sloan-Kettering Cancer Center, New York
Frank J. Rauscher, III
Affiliation:
The Wistar Institute Cancer Centre, Philadelphia
Get access

Summary

Introduction

Ovarian cancer is the most lethal gynecologic malignancy in the United States. In 2010, over 21 880 new cases were diagnosed and 13 850 women died from this disease (1). Over the past two decades, advances in conventional therapy have led to an increase in five-year survival for women with ovarian cancer, but the fraction of long-term survivors remains unchanged at 30%, when all stages are considered. Traditional methods are unlikely to produce dramatic improvements in outcomes. Insights into the molecular mechanisms of ovarian cancers, the biological basis for their clinical behavior, and utilization of these targets are needed.

Cellular origin of ovarian cancers

Over 90% of ovarian cancers are of epithelial origin with mesothelial features. The remaining 10% are from germ cells or granulosa-theca cells within the ovary. Traditionally, epithelial ovarian cancers have been thought to originate from flattened cells covering the ovary or lining subserosal inclusion cysts (2). Recent evidence suggests that the majority of familial ovarian cancers, and as many as 20% of the primary peritoneal cancers, may arise from the fimbriae of the Fallopian tube (3). Epithelial ovarian cancers exhibit four histotypes (serous, endometrioid, mucinous, and clear cell) regulated by the different HOX genes implicated in normal gynecologic development (Table 54.1; 4,5). Ovarian cancers may be of low or high grade, correlating with stage at presentation, rate of growth, and response to chemotherapy. Ninety percent of epithelial ovarian cancers are clonal; thus, the genetic abnormalities found in primary cancers are also found in metastases, although the correlation is not always precise (6).

Type
Chapter
Information
Molecular Oncology
Causes of Cancer and Targets for Treatment
, pp. 606 - 618
Publisher: Cambridge University Press
Print publication year: 2013

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Jemal, A, Siegel, R, Xu, J, Ward, E. Cancer statistics, 2010. CA, A Cancer Journal for Clinicians 2010;60:277–300.CrossRef
Bast, RC Jr, Hennessy, B, Mills, GB. The biology of ovarian cancer: new opportunities for translation. Nature Reviews Cancer 2009;9:415–28.CrossRef
Mehra, K, Mehrad, M, Ning, G, et al. STICS, SCOUTs and p53 signatures: a new language for pelvic serous carcinogenesis. Frontiers in Bioscience 2011;3:625–34.
Feeley, KM, Wells, M. Precursor lesions of ovarian epithelial malignancy. Histopathology 2001;38:87–95.CrossRef
Cheng, W, Liu, J, Yoshida, H, Rosen, D, Naora, H. Lineage infidelity of epithelial ovarian cancers is controlled by HOX genes that specify regional identity in the reproductive tract. Nature Medicine 2005;11:531–7.CrossRef
Jacobs, IJ, Kohler, MF, Wiseman, RW, et al. Clonal origin of epithelial ovarian carcinoma: analysis by loss of heterozygosity, p53 mutation, and X-chromosome inactivation. Journal of the National Cancer Institute 1992;84:1793–8.CrossRefGoogle ScholarPubMed
Kohler, MF, Marks, JR, Wiseman, RW, et al. Spectrum of mutation and frequency of allelic deletion of the p53 gene in ovarian cancer. Journal of the National Cancer Institute 1993;85:1513–1519.CrossRefGoogle ScholarPubMed
Schildkraut, JM, Bastos, E, Berchuck, A. Relationship between lifetime ovulatory cycles and overexpression of mutant p53 in epithelial ovarian cancer. Journal of the National Cancer Institute 1997;89:932–8.CrossRefGoogle ScholarPubMed
Moynahan, ME, Pierce, AJ, Jasin, M. BRCA2 is required for homology-directed repair of chromosomal breaks. Molecular Cell 2001;7:263–72.CrossRef
Narod, SA, Foulkes, WD. BRCA1 and BRCA2: 1994 and beyond. Nature Reviews Cancer 2004;4:665–76.CrossRef
Lancaster, JM, Wooster, R, Mangion, J, et al. BRCA2 mutations in primary breast and ovarian cancers. Nature Genetics 1996;13:238–40.CrossRef
Rubin, SC, Blackwood, MA, Bandera, C, et al. BRCA1, BRCA2, and hereditary nonpolyposis colorectal cancer gene mutations in an unselected ovarian cancer population: relationship to family history and implications for genetic testing. American Journal of Obstetrics and Gynecology 1998;178:670–7.CrossRefGoogle Scholar
Boyd, J. Molecular genetics of hereditary ovarian cancer. Oncology (Williston Park) 1998;12:399–406.
Edwards, SL, Brough, R, Lord, CJ, et al. Resistance to therapy caused by intragenic deletion in BRCA2. Nature 2008;451:1111–15.CrossRef
Chetrit, A, Hirsh-Yechezkel, G, Ben-David, Y, et al. Effect of BRCA1/2 mutations on long-term survival of patients with invasive ovarian cancer: the national Israeli study of ovarian cancer. Journal of Clinical Oncology 2008;26:20–5.CrossRefGoogle ScholarPubMed
Sakai, W, Swisher, EM, Karlan, BY, et al. Secondary mutations as a mechanism of cisplatin resistance in BRCA2-mutated cancers. Nature 2008;451:1116–20.CrossRef
Drew, Y, Calvert, H. The potential of PARP inhibitors in genetic breast and ovarian cancers. Annals of the New York Academy of Science 2008;1138:136–45.CrossRef
Yap, TA, Carden, CP, Kaye, SB. Beyond chemotherapy: targeted therapies in ovarian cancer. Nature Reviews Cancer 2009;9:167–81.CrossRef
Hennessy, BT, Timms, KM, Carey, MS, et al. Somatic mutations in BRCA1 and BRCA2 could expand the number of patients that benefit from poly (ADP ribose) polymerase inhibitors in ovarian cancer. Journal of Clinical Oncology 2010;28:3570–6.CrossRefGoogle ScholarPubMed
Hennessy, BT, Timms, KM, Carey, MS, et al. Somatic mutations in BRCA1 and BRCA2 could expand the number of patients that benefit from poly (ADP ribose) polymerase inhibitors in ovarian cancer. Journal of Clinical Oncology 2010;28:3570–6.CrossRefGoogle ScholarPubMed
Bast, RC, Mills, GB. Personalizing therapy for ovarian cancer: BRCAness and beyond, Journal of Clinical Oncology 2010;28:3545–8.CrossRefGoogle ScholarPubMed
Jazaeri, AA, Yee, CJ, Sotiriou, C, et al. Gene expression profiles of BRCA1-linked, BRCA2-linked, and sporadic ovarian cancers. Journal of the National Cancer Institute 2002;94:990–1000.CrossRefGoogle ScholarPubMed
Ahmed, AA, Etemadmoghadam, D, Temple, J, et al. Driver mutations in TP53 are ubiquitous in high grade serous carcinoma of the ovary. Journal of Pathology 2010;221(1):49–56.CrossRefGoogle ScholarPubMed
Vousden, KH, Lane, DP. p53 in health and disease. Nature Reviews Molecular and Cellular Biology 2007;8:275–83.CrossRef
Buller, RE, Runnebaum, IB, Karlan, BY, et al. A Phase I/II trial of rAd/p53 (SCH 58500) gene replacement in recurrent ovarian cancer. Cancer Gene Therapy 2002;9:553–66.CrossRef
Vasey, PA, Shulman, LN, Campos, S, et al. Phase I trial of intraperitoneal injection of the E1B-55-kd-gene-deleted adenovirus ONYX-015 (dl1520) given on days 1 through 5 every 3 weeks in patients with recurrent/refractory epithelial ovarian cancer. Journal of Clinical Oncology 2002;20:1562–9.Google ScholarPubMed
Kojima, K, Konopleva, M, Samudio, IJ, et al. MDM2 antagonists induce p53-dependent apoptosis in AML: implications for leukemia therapy. Blood 2005;106:3150–9.CrossRef
Obata, K, Morland, SJ, Watson, RH, et al. Frequent PTEN/MMAC mutations in endometrioid but not serous or mucinous epithelial ovarian tumors. Cancer Research 1998;58:2095–7.
Cancer Genome Atlas Research Network. Integrated genomic analyses of ovarian carcinoma. Nature. 2011;474:609–15.CrossRef
Jones, S, Wang, TL, Shih, IeM, et al. Frequent mutations of chromatin remodeling gene ARID1A in ovarian clear cell carcinoma. Science 2010;330:228–31.CrossRef
Wiegand, KC, Shah, SP, Al-Agha, OM, et al. ARID1A mutations in endometriosis-associated ovarian carcinomas. New England Journal of Medicine 2010;363:1532–43.CrossRefGoogle ScholarPubMed
Yu, Y, Luo, R, Lu, Z, et al. Biochemistry and biology of ARHI (DIRAS3), an imprinted tumor suppressor gene whose expression is lost in ovarian and breast cancers. Methods in Enzymology 2006;407:455–68.CrossRef
Yu, Y, Xu, F, Peng, H, et al. NOEY2 (ARHI), an imprinted putative tumor suppressor gene in ovarian and breast carcinomas. Proceedings of the National Academy of Sciences USA 1999;96:214–19.CrossRef
Fujii, S, Luo, RZ, Yuan, J, et al. Reactivation of the silenced and imprinted alleles of ARHI is associated with increased histone H3 acetylation and decreased histone H3 lysine 9 methylation. Human Molecular Genetics 2003;12:1791–800.CrossRef
Yuan, J, Luo, RZ, Fujii, S, et al. Aberrant methylation and silencing of ARHI, an imprinted tumor suppressor gene in which the function is lost in breast cancers. Cancer Research 2003;63:4174–80.
Mackay, H, Hirte, HW, Covens, A, et al. A Phase II trial of the histone deacetylase inhibitor belinostat (PSC101) in patients with platinum resistant epithelial ovarian tumors and micropapillary/borderline (LMP) ovarian tumors. A PMH Phase II consortium trial. Journal of Clinical Oncology 2008;26(Suppl.).CrossRefGoogle Scholar
Balch, C, Huang, TH, Brown, R, Nephew, KP. The epigenetics of ovarian cancer drug resistance and resensitization. American Journal of Obstetrics and Gynecology 2004;191:1552–72.CrossRefGoogle ScholarPubMed
Bast, RC, Iyer, R, Hu, W, et al. A Phase IIa study of sequential regimen using azacitidine to reverse platinum resistance to carboplatin in patients with platinum resistant or refractory epithelial ovarian cancer. Proceedings of the American Society of Clinical Oncology 2008;26:3500.CrossRef
Eder, AM, Sui, X, Rosen, DG, et al. Atypical PKCiota contributes to poor prognosis through loss of apical-basal polarity and cyclin E overexpression in ovarian cancer. Proceedings of the National Academy of Sciences USA 2005;102:12 519–24.
Etemadmoghadam, D, DeFazio, A, Beroukhim, R, et al. Integrated genome-wide DNA copy number and expression analysis identifies distinct mechanisms of primary chemoresistance in ovarian carcinomas. Clinical Cancer Research 2009;15:1417–27CrossRef
Tuefferd, M, Couturier, J, Penault-Llorca, F, et al. HER2 status in ovarian carcinomas: a multicenter GINECO study of 320 patients. PLoS One 2007;2:e1138.
Bookman, MA, Darcy, KM, Clarke-Pearson, D, Boothby, RA, Horowitz, IR. Evaluation of monoclonal humanized anti-HER2 antibody, trastuzumab, in patients with recurrent or refractory ovarian or primary peritoneal carcinoma with overexpression of HER2: a Phase II trial of the Gynecologic Oncology Group. Journal of Clinical Oncology 2003;21:283–90.CrossRefGoogle ScholarPubMed
Cheng, KW, Lahad, JP, Gray, JW, Mills, GB. Emerging role of RAB GTPases in cancer and human disease. Cancer Research 2005;65:2516–19.CrossRef
Gautschi, O, Heighway, J, Mack, PC, et al. Aurora kinases as anticancer drug targets. Clinical Cancer Research 2008;14:1639–48.CrossRef
Li, K, Li, Y, Wu, W, et al. Modulation of Notch signaling by antibodies specific for the extracellular negative regulatory region of NOTCH3. Journal of Biological Chemistry 2008;283:8046–54.CrossRefGoogle ScholarPubMed
Cheung, HW, Cowley, GS, Weirb, BA, et al. Systematic investigation of genetic vulnerabilities across cancer cell lines reveals lineage-specific dependencies in ovarian cancer. Proceedings of the National Academy of Sciences USA 2011;108:12 372–77.
Kurman, RJ, Shih Ie, M. Pathogenesis of ovarian cancer: lessons from morphology and molecular biology and their clinical implications. International Journal of Gynecolic Pathology 2008;27:151–60.Google ScholarPubMed
Mitsuuchi, Y, Johnson, SW, Selvakumaran, M, et al. The phosphatidylinositol 3-kinase/AKT signal transduction pathway plays a critical role in the expression of p21WAF1/CIP1/SDI1 induced by cisplatin and paclitaxel. Cancer Research 2000;60:5390–4.
Shayesteh, L, Lu, Y, Kuo, WL, et al. PIK3CA is implicated as an oncogene in ovarian cancer. Nature Genetics 1999;21:99–102.CrossRef
Vivanco, I, Sawyers, CL. The phosphatidylinositol 3-Kinase AKT pathway in human cancer. Nature Reviews Cancer 2002;2:489–501.CrossRef
Hennessy, BT, Smith, DL, Ram, PT, Lu, Y, Mills, GB. Exploiting the PI3K/AKT pathway for cancer drug discovery. Nature Reviews Drug Discovery 2005;4:988–1004.CrossRef
Jiang, BH, Liu, LZ. AKT signaling in regulating angiogenesis. Current Cancer Drug Targets 2008;8:19–26.CrossRef
Levine, DA, Bogomolniy, F, Yee, CJ, et al. Frequent mutation of the PIK3CA gene in ovarian and breast cancers. Clinical Cancer Research 2005;11:2875–8.CrossRef
Trotman, LC, Pandolfi, PP. PTEN and p53: who will get the upper hand? Cancer Cell 2003;3:97–9.
Cheng, JQ, Lindsley, CW, Cheng, GZ, Yang, H, Nicosia, SV. The Akt/PKB pathway: molecular target for cancer drug discovery. Oncogene 2005;24:7482–92.CrossRef
Yang, X, Fraser, M, Moll, UM, Basak, A, Tsang, BK. Akt-mediated cisplatin resistance in ovarian cancer: modulation of p53 action on caspase-dependent mitochondrial death pathway. Cancer Research 2006;66:3126–36.CrossRef
Hu, L, Hofmann, J, Lu, Y, Mills, GB, Jaffe, RB. Inhibition of phosphatidylinositol 3ʹ-kinase increases efficacy of paclitaxel in in vitro and in vivo ovarian cancer models. Cancer Research 2002;62:1087–92.
Nimeiri, HS, Oza, AM, Morgan, RJ, et al. Efficacy and safety of bevacizumab plus erlotinib for patients with recurrent ovarian, primary peritoneal, and fallopian tube cancer: a trial of the Chicago, PMH, and California Phase II Consortia. Gynecologic Oncology 2008;110:49–55.CrossRef
Schilder, RJ, Sill, MW, Chen, X, et al. Phase II study of gefitinib in patients with relapsed or persistent ovarian or primary peritoneal carcinoma and evaluation of epidermal growth factor receptor mutations and immunohistochemical expression: a Gynecologic Oncology Group Study. Clinical Cancer Research 2005;11:5539–48.CrossRef
Lacroix, L, Pautier, P, Duvillard, P, et al. Response of ovarian carcinomas to gefitinib-carboplatin-paclitaxel combination is not associated with EGFR kinase domain somatic mutations. International Journal of Cancer 2006;118:1068–9.CrossRefGoogle Scholar
Gordon, AN, Finkler, N, Edwards, RP, et al. Efficacy and safety of erlotinib HCl, an epidermal growth factor receptor (HER1/EGFR) tyrosine kinase inhibitor, in patients with advanced ovarian carcinoma: results from a Phase II multicenter study. International Journal of Gynecological Cancer 2005;15:785–92.CrossRefGoogle ScholarPubMed
Wagner, U, du Bois, A, Pfisterer, J, et al. Gefitinib in combination with tamoxifen in patients with ovarian cancer refractory or resistant to platinum-taxane based therapy–a Phase II trial of the AGO Ovarian Cancer Study Group (AGO-OVAR 2.6). Gynecologic Oncology 2007;105:132–7.
Rosen, DG, Mercado-Uribe, I, Yang, G, et al. The role of constitutively active signal transducer and activator of transcription 3 in ovarian tumorigenesis and prognosis. Cancer 2006;107:2730–40.CrossRef
Chan, KS, Carbajal, S, Kiguchi, K, et al. Epidermal growth factor receptor-mediated activation of Stat3 during multistage skin carcinogenesis. Cancer Research 2004;64:2382–9.CrossRef
Carbia-Nagashima, A, Arzt, E. Intracellular proteins and mechanisms involved in the control of gp130/JAK/STAT cytokine signaling. IUBMB Life 2004;56:83–8.CrossRef
Yamashita, S, Miyagi, C, Carmany-Rampey, A, et al. Stat3 controls cell movements during zebrafish gastrulation. Developmental Cell 2002;2:363–75.CrossRef
McMurray, JS. A new small-molecule Stat3 inhibitor. Chemical Biology 2006;13:1123–4.CrossRef
Estrella, VC, Eder, AM, Liu, S, et al. Lysophosphatidic acid induction of urokinase plasminogen activator secretion requires activation of the p38MAPK pathway. International Journal of Oncology 2007;31:441–9.Google ScholarPubMed
Mills, GB, Eder, A, Fang, X, et al. Critical role of lysophospholipids in the pathophysiology, diagnosis, and management of ovarian cancer. Cancer Treatment and Research 2002;107:259–83.
Beck, HP, Kohn, T, Rubenstein, S, et al. Discovery of potent LPA2 (EDG4) antagonists as potential anticancer agents. Bioorganic and Medicinal Chemistry Letters 2008;18:1037–41.CrossRef
Samanta, AK, Huang, HJ, Bast, RC, Jr., Liao, WS. Overexpression of MEKK3 confers resistance to apoptosis through activation of NFkappaB. Journal of Biological Chemistry 2004;279:7576–83.CrossRefGoogle ScholarPubMed
Hacker, H, Karin, M. Regulation and function of IKK and IKK-related kinases. Science STKE 2006;re13.
Marquez, RT, Baggerly, KA, Patterson, AP, et al. Patterns of gene expression in different histotypes of epithelial ovarian cancer correlate with those in normal fallopian tube, endometrium and colon. Clinical Cancer Research 2005;11:6116–26.CrossRef
Tothill, RW, Tinker, AV, George, J, et al. Novel molecular subtypes of serous and endometrioid ovarian cancer linked to clinical outcome. Clinical Cancer Research 2008;14:5198–208.CrossRef
Liang, XH, Jackson, S, Seaman, M, et al. Induction of autophagy and inhibition of tumorigenesis by beclin 1. Nature 1999;402:672–6.CrossRef
Lu, Z, Luo, RZ, Lu, Y, et al. The tumor suppressor gene ARHI regulates autophagy and tumor dormancy in human ovarian cancer cells. Journal of Clinical Investigation 2008;118:3917–29.Google ScholarPubMed
Folkman, J, Watson, K, Ingber, D, Hanahan, D. Induction of angiogenesis during the transition from hyperplasia to neoplasia. Nature 1989;339:58–61.CrossRef
Hanahan, D, Folkman, J. Patterns and emerging mechanisms of the angiogenic switch during tumorigenesis. Cell 1996;86:353–64.CrossRef
Zebrowski, BK, Liu, W, Ramirez, K, et al. Markedly elevated levels of vascular endothelial growth factor in malignant ascites. Annals of Surgical Oncology 1999;6:373–8.CrossRef
Mok, SC, Bonome, T, Vathipadiekal, V, et al. A gene signature predictive for outcome in advanced ovarian cancer identifies a survival factor: microfibril-associated glycoprotein 2. Cancer Cell 2009;16:521–32.CrossRef
Burger, RA, Sill, MW, Monk, BJ, Greer, BE, Sorosky, JI. Phase II trial of bevacizumab in persistent or recurrent epithelial ovarian cancer or primary peritoneal cancer: a Gynecologic Oncology Group Study. Journal of Clinical Oncology 2007;25:5165–71.CrossRefGoogle ScholarPubMed
Cannistra, SA, Matulonis, UA, Penson, RT, et al. Phase II study of bevacizumab in patients with platinum-resistant ovarian cancer or peritoneal serous cancer. Journal of Clinical Oncology 2007;25:5180–6.CrossRefGoogle ScholarPubMed
Kamat, AA, Kim, TJ, Landen, CN, Jr., et al. Metronomic chemotherapy enhances the efficacy of antivascular therapy in ovarian cancer. Cancer Research 2007;67:281–8.CrossRef
Biagi, JJ. A Phase II study of sunitinib (SU11248) in patients with recurrent epithelial ovarian, fallopian tube, or primary peritoneal carcinoma – NCIC GTG IND 185. Journal of Clinical Oncology 2007;25(Suppl.).Google Scholar
Friedlander, M, Hancock, KC, Benigno, B, et al. Pazopanib (GW786034) is active in women with advanced epithelial ovarian, fallopian tube, and peritoneal cancers: initial results of a Phase II study. Journal of Clinical Oncology 2007;25(Suppl.).Google Scholar
Hirte, JW, Vidal, L, Fleming, GF, et al. A Phase II study of cediranib (AZD2171) in recurrent or persistent ovarian, peritoneal, or fallopian tube cancer: final results of a PMH, Chicago and California consortia trial. Journal of Clinical Oncology 2008;26(Suppl).CrossRefGoogle Scholar
Matulonis, UA, Berlin, ST, Krasner, CN, et al. Cediranib (AZD2171) is an active agent in recurrent epithelial ovarian cancer. Journal of Clinical Oncology 2008;26(Suppl).CrossRefGoogle Scholar
Muggia, F. Platinum compounds 30 years after the introduction of cisplatin: implications for the treatment of ovarian cancer. Gynecologic Oncology 2009;112:275–81.CrossRef
Yan, X, Yin, J, Yao, H, et al. Increased expression of annexin A3 is a mechanism of platinum resistance in ovarian cancer. Cancer Research 2010;70:1616–24.CrossRef
Schuijer, M, Berns, EM. TP53 and ovarian cancer. Human Mutation 2003;21:285–91.CrossRef
Mansouri, A, Zhang, Q, Ridgway, LD, Tian, L, Claret, FX. Cisplatin resistance in an ovarian carcinoma is associated with a defect in programmed cell death control through XIAP regulation. Oncology Research 2003;13:399–404.CrossRef
Selvakumaran, M, Pisarcik, DA, Bao, R, Yeung, AT, Hamilton, TC. Enhanced cisplatin cytotoxicity by disturbing the nucleotide excision repair pathway in ovarian cancer cell lines. Cancer Research 2003;63:1311–16.
Mangala, LS, Zuzel, V, Schmandt, R. Therapeutic targeting of ATP7B. Clinical Cancer Research 2009;15:3770–80.CrossRef
Livingston, DM, Silver, DP. Cancer: crossing over to drug resistance. Nature 2008;451:1066–7.CrossRef
Perez, EA. Impact, mechanisms and novel chemotherapeutic strategies for overcoming resistance to anthracyclines and taxanes in metastatic breast cancer. Breast Cancer Research and Treatment 2009;114:195–201.CrossRef
Muggia, FM, Braly, PS, Brady, MF et al. Phase III randomized study of cisplatin versus paclitaxel versus cisplatin and paclitaxel in patients with suboptimal stage III or IV ovarian cancer: a Gynecologic Oncology Group study. Journal of Clinical Oncology 2000;18:106–115.CrossRefGoogle ScholarPubMed
Ahmed, AA, Wang, X, Lu, Z et al. Modulating microtubule stability enhances the cytotoxic response of cancer cells to paclitaxel. Cancer Research 2011;71:5806–17.CrossRef
Ahmed, AA, Lu, Z, Jennings, NB et al. SIK2 is a centrosome kinase required for bipolar spindle formation that provides a potential target for therapy in ovarian cancer. Cancer Cell 2010;18:109–121.CrossRef
Whitehurst, AW, Xie, Y, Purinton, SC et al. Tumor antigen acrosin binding protein normalizes mitotic spindle function to promote cancer cell proliferation. Cancer Research 2010;70:7652–61.CrossRef
Iglehart, JD, Silver, DP. Synthetic lethality: a new direction in cancer-drug development. New England Journal of Medicine 2009;361:189–91.CrossRefGoogle ScholarPubMed

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

  • Molecular targets for epithelial ovarian cancer
    • By Grace K. Suh, Departments of Experimentalherapeutics, Gynecologic Medical Oncology, Bioinformatics and Computational Biology, and Systems Biology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA, Bryan T. Hennessy, Departments of Experimentalherapeutics, Gynecologic Medical Oncology, Bioinformatics and Computational Biology, and Systems Biology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA, Roeland Verhaak, Departments of Experimentalherapeutics, Gynecologic Medical Oncology, Bioinformatics and Computational Biology, and Systems Biology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA, Ji-Yeon Yang, Departments of Experimentalherapeutics, Gynecologic Medical Oncology, Bioinformatics and Computational Biology, and Systems Biology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA, Gordon B. Mills, Departments of Experimentalherapeutics, Gynecologic Medical Oncology, Bioinformatics and Computational Biology, and Systems Biology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA, Robert C. Bast, Departments of Experimentalherapeutics, Gynecologic Medical Oncology, Bioinformatics and Computational Biology, and Systems Biology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
  • Edited by Edward P. Gelmann, Columbia University, New York, Charles L. Sawyers, Memorial Sloan-Kettering Cancer Center, New York, Frank J. Rauscher, III
  • Book: Molecular Oncology
  • Online publication: 05 February 2015
  • Chapter DOI: https://doi.org/10.1017/CBO9781139046947.055
Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

  • Molecular targets for epithelial ovarian cancer
    • By Grace K. Suh, Departments of Experimentalherapeutics, Gynecologic Medical Oncology, Bioinformatics and Computational Biology, and Systems Biology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA, Bryan T. Hennessy, Departments of Experimentalherapeutics, Gynecologic Medical Oncology, Bioinformatics and Computational Biology, and Systems Biology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA, Roeland Verhaak, Departments of Experimentalherapeutics, Gynecologic Medical Oncology, Bioinformatics and Computational Biology, and Systems Biology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA, Ji-Yeon Yang, Departments of Experimentalherapeutics, Gynecologic Medical Oncology, Bioinformatics and Computational Biology, and Systems Biology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA, Gordon B. Mills, Departments of Experimentalherapeutics, Gynecologic Medical Oncology, Bioinformatics and Computational Biology, and Systems Biology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA, Robert C. Bast, Departments of Experimentalherapeutics, Gynecologic Medical Oncology, Bioinformatics and Computational Biology, and Systems Biology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
  • Edited by Edward P. Gelmann, Columbia University, New York, Charles L. Sawyers, Memorial Sloan-Kettering Cancer Center, New York, Frank J. Rauscher, III
  • Book: Molecular Oncology
  • Online publication: 05 February 2015
  • Chapter DOI: https://doi.org/10.1017/CBO9781139046947.055
Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

  • Molecular targets for epithelial ovarian cancer
    • By Grace K. Suh, Departments of Experimentalherapeutics, Gynecologic Medical Oncology, Bioinformatics and Computational Biology, and Systems Biology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA, Bryan T. Hennessy, Departments of Experimentalherapeutics, Gynecologic Medical Oncology, Bioinformatics and Computational Biology, and Systems Biology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA, Roeland Verhaak, Departments of Experimentalherapeutics, Gynecologic Medical Oncology, Bioinformatics and Computational Biology, and Systems Biology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA, Ji-Yeon Yang, Departments of Experimentalherapeutics, Gynecologic Medical Oncology, Bioinformatics and Computational Biology, and Systems Biology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA, Gordon B. Mills, Departments of Experimentalherapeutics, Gynecologic Medical Oncology, Bioinformatics and Computational Biology, and Systems Biology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA, Robert C. Bast, Departments of Experimentalherapeutics, Gynecologic Medical Oncology, Bioinformatics and Computational Biology, and Systems Biology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
  • Edited by Edward P. Gelmann, Columbia University, New York, Charles L. Sawyers, Memorial Sloan-Kettering Cancer Center, New York, Frank J. Rauscher, III
  • Book: Molecular Oncology
  • Online publication: 05 February 2015
  • Chapter DOI: https://doi.org/10.1017/CBO9781139046947.055
Available formats
×