Repopulation of ovarian cancer cells after chemotherapy

Carlos M Telleria, Carlos M Telleria

Abstract

The high mortality rate caused by ovarian cancer has not changed for the past thirty years. Although most patients diagnosed with this disease respond to cytoreductive surgery and platinum-based chemotherapy and undergo remission, foci of cells almost always escape therapy, manage to survive, and acquire the capacity to repopulate the tumor. Repopulation of ovarian cancer cells that escape front-line chemotherapy, however, is a poorly understood phenomenon. Here I analyze cancer-initiating cells, transitory senescence, reverse ploidy, and cellular dormancy as putative players in ovarian cancer cell repopulation. Under standard of care, ovarian cancer patients do not receive treatment between primary cytotoxic therapy and clinical relapse; understanding the mechanisms driving cellular escape from chemotherapy should lead to the development of low toxicity, chronic treatment approaches that can be initiated right after primary therapy to interrupt cell repopulation and disease relapse by keeping it dormant and, therefore, subclinical.

References

    1. Gillies RJ, Verduzco D, Gatenby RA. Evolutionary dynamics of carcinogenesis and why targeted therapy does not work. Nat Rev Cancer. 2012;12(7):487–93.
    1. Romero I, Bast RC., Jr Minireview: human ovarian cancer: biology, current management, and paths to personalizing therapy. Endocrinology. 2012;153(4):1593–602.
    1. Vaughan S, Coward JI, Bast RC, Jr, et al. Rethinking ovarian cancer: recommendations for improving outcomes. Nat Rev Cancer. 2011;11(10):719–25.
    1. Bast RC, Jr, Hennessy B, Mills GB. The biology of ovarian cancer: new opportunities for translation. Nat Rev Cancer. 2009;9(6):415–28.
    1. Kurman RJ, Shih Ie M. Molecular pathogenesis and extraovarian origin of epithelial ovarian cancer—shifting the paradigm. Hum Pathol. 2011;42(7):918–31.
    1. Bast RC., Jr Molecular approaches to personalizing management of ovarian cancer. Ann Oncol. 2011;22(Suppl 8):viii5–15.
    1. Bast RC, Jr, Mills GB. Dissecting “PI3 Kness”: The complexity of personalized therapy for ovarian cancer. Cancer Discov. 2012;2(1):16–8.
    1. Lengyel E. Ovarian cancer development and metastasis. Am J Pathol. 2010;177(3):1053–64.
    1. Tentes AA, Courcoutsakis N, Prasopoulos P. Combined cytoreductive surgery and perioperative intraperitoneal chemotherapy for the treatment of advanced ovarian cancer. In: Farghaly SA, editor. Ovarian Cancer—Clinical and Therapeutic Perspectives. New York, NY: InTech; 2012. pp. 143–66.
    1. Ozols RF. Systemic therapy for ovarian cancer: current status and new treatments. Semin Oncol. 2006;33(2 Suppl 6):S3–11.
    1. Rabik CA, Dolan ME. Molecular mechanisms of resistance and toxicity associated with platinating agents. Cancer Treat Rev. 2007;33(1):9–23.
    1. Kelland L. The resurgence of platinum-based cancer chemotherapy. Nat Rev Cancer. 2007;7(8):573–84.
    1. Jung Y, Lippard SJ. Direct cellular responses to platinum-induced DNA damage. Chem Rev. 2007;107(5):1387–407.
    1. Cepeda V, Fuertes MA, Castilla J, Alonso C, Quevedo C, Perez JM. Biochemical mechanisms of cisplatin cytotoxicity. Anticancer Agents Med Chem. 2007;7(1):3–18.
    1. Horwitz SB. Mechanism of action of taxol. Trends Pharmacol Sci. 1992;13(4):134–6.
    1. Jekunen AP, Christen RD, Shalinsky DR, Howell SB. Synergistic interaction between cisplatin and taxol in human ovarian carcinoma cells in vitro. Br J Cancer. 1994;69(2):299–306.
    1. Ross AA, Miller GW, Moss TJ, et al. Immunocytochemical detection of tumor cells in bone marrow and peripheral blood stem cell collections from patients with ovarian cancer. Bone Marrow Transplant. 1995;15(6):929–33.
    1. Walker FE. Paclitaxel (TAXOL): side effects and patient education issues. Semin Oncol Nurs. 1993;9(4 Suppl 2):6–10.
    1. Kim JJ, Tannock IF. Repopulation of cancer cells during therapy: an important cause of treatment failure. Nat Rev Cancer. 2005;5(7):516–25.
    1. Davis AJ, Tannock JF. Repopulation of tumour cells between cycles of chemotherapy: a neglected factor. Lancet Oncol. 2000;1:86–93.
    1. Davis AJ, Tannock IF. Tumor physiology and resistance to chemotherapy: repopulation and drug penetration. Cancer Treat Res. 2002;112:1–26.
    1. Gamarra-Luques CD, Goyeneche AA, Hapon MB, Telleria CM. Mifepristone prevents repopulation of ovarian cancer cells escaping cisplatin-paclitaxel therapy. BMC Cancer. 2012;12:200.
    1. Freeburg EM, Goyeneche AA, Telleria CM. Mifepristone abrogates repopulation of ovarian cancer cells in between courses of cisplatin treatment. Int J Oncol. 2009;34(3):743–55.
    1. Coley HM. Development of drug-resistant models. Methods Mol Med. 2004;88:267–73.
    1. Davis AJ, Chapman W, Hedley DW, Oza AM, Tannock IF. Assessment of tumor cell repopulation after chemotherapy for advanced ovarian cancer: pilot study. Cytometry A. 2003;51(1):1–6.
    1. Ohtsu T, Sasaki Y, Tamura T, et al. Clinical pharmacokinetics and pharmacodynamics of paclitaxel: a 3-hour infusion versus a 24-hour infusion. Clin Cancer Res. 1995;1(6):599–606.
    1. Rowinsky EK, Jiroutek M, Bonomi P, Johnson D, Baker SD. Paclitaxel steady-state plasma concentration as a determinant of disease outcome and toxicity in lung cancer patients treated with paclitaxel and cisplatin. Clin Cancer Res. 1999;5(4):767–74.
    1. Kurata T, Tamura T, Shinkai T, et al. Phase I and pharmacological study of paclitaxel given over 3 h with cisplatin for advanced non-small cell lung cancer. Jpn J Clin Oncol. 2001;31(3):93–9.
    1. Urien S, Lokiec F. Population pharmacokinetics of total and unbound plasma cisplatin in adult patients. Br J Clin Pharmacol. 2004;57(6):756–63.
    1. Fuertes MA, Castilla J, Alonso C, Perez JM. Cisplatin biochemical mechanism of action: from cytotoxicity to induction of cell death through interconnections between apoptotic and necrotic pathways. Curr Med Chem. 2003;10(3):257–66.
    1. Taniguchi T, Tischkowitz M, Ameziane N, et al. Disruption of the Fanconi anemia-BRCA pathway in cisplatin-sensitive ovarian tumors. Nat Med. 2003;9(5):568–74.
    1. Jacquemont C, Simon JA, D’Andrea AD, Taniguchi T. Non-specific chemical inhibition of the Fanconi anemia pathway sensitizes cancer cells to cisplatin. Mol Cancer. 2012;11:26.
    1. Neuzil J, Stantic M, Zobalova R, et al. Tumour-initiating cells vs. cancer ‘stem’ cells and CD133: what’s in the name? Biochem Biophys Res Commun. 2007;355(4):855–9.
    1. Malik B, Nie D. Cancer stem cells and resistance to chemo and radio therapy. Front Biosci (Elite Ed) 2012;4:2142–9.
    1. Yu Y, Ramena G, Elble RC. The role of cancer stem cells in relapse of solid tumors. Front Biosci (Elite Ed) 2012;4:1528–41.
    1. Robertson FM, Ogasawara MA, Ye Z, et al. Imaging and analysis of 3D tumor spheroids enriched for a cancer stem cell phenotype. J Biomol Screen. 2010;15(7):820–9.
    1. Abelson S, Shamai Y, Berger L, Shouval R, Skorecki K, Tzukerman M. Intratumoral heterogeneity in the self-renewal and tumorigenic differentiation of ovarian cancer. Stem Cells. 2012;30(3):415–24.
    1. Silva IA, Bai S, McLean K, et al. Aldehyde dehydrogenase in combination with CD133 defines angiogenic ovarian cancer stem cells that portend poor patient survival. Cancer Res. 2011;71(11):3991–4001.
    1. Kryczek I, Liu S, Roh M, et al. Expression of aldehyde dehydrogenase and CD133 defines ovarian cancer stem cells. Int J Cancer. 2012;130(1):29–39.
    1. Baba T, Convery PA, Matsumura N, et al. Epigenetic regulation of CD133 and tumorigenicity of CD133+ ovarian cancer cells. Oncogene. 2009;28(2):209–18.
    1. Ferrandina G, Bonanno G, Pierelli L, et al. Expression of CD133-1 and CD133-2 in ovarian cancer. Int J Gynecol Cancer. 2007;18(3):506–14.
    1. Curley MD, Therrien VA, Cummings CL, et al. CD133 expression defines a tumor initiating cell population in primary human ovarian cancer. Stem Cells. 2009;27(12):2875–83.
    1. Long H, Xie R, Xiang T, et al. Autocrine CCL5 signaling promotes invasion and migration of CD133(+) ovarian cancer stem-like cells via NF-kappaB-mediated MMP-9 upregulation. Stem Cells. 2012;30(10):2309–19.
    1. Curley MD, Garrett LA, Schorge JO, Foster R, Rueda BR. Evidence for cancer stem cells contributing to the pathogenesis of ovarian cancer. Front Biosci. 2011;16:368–92.
    1. Heddleston JM, Li Z, McLendon RE, Hjelmeland AB, Rich JN. The hypoxic microenvironment maintains glioblastoma stem cells and promotes reprogramming towards a cancer stem cell phenotype. Cell Cycle. 2009;8(20):3274–84.
    1. Cairney CJ, Bilsland AE, Evans TR, et al. Cancer cell senescence: a new frontier in drug development. Drug Discov Today. 2012;17(5–6):269–76.
    1. Acosta JC, Gil J. Senescence: a new weapon for cancer therapy. Trends Cell Biol. 2012;22(4):211–9.
    1. Schmitt CA. Cellular senescence and cancer treatment. Biochim Biophys Acta. 2007;1775(1):5–20.
    1. Beausejour CM, Krtolica A, Galimi F, et al. Reversal of human cellular senescence: roles of the p53 and p16 pathways. EMBO J. 2003;22(16):4212–22.
    1. Roberson RS, Kussick SJ, Vallieres E, Chen SY, Wu DY. Escape from therapy-induced accelerated cellular senescence in p53-null lung cancer cells and in human lung cancers. Cancer Res. 2005;65(7):2795–803.
    1. Sliwinska MA, Mosieniak G, Wolanin K, et al. Induction of senescence with doxorubicin leads to increased genomic instability of HCT116 cells. Mech Ageing Dev. 2009;130(1–2):24–32.
    1. La Porta CA, Zapperi S, Sethna JP. Senescent cells in growing tumors: population dynamics and cancer stem cells. PLoS Comput Biol. 2012;8(1):e1002316.
    1. Achuthan S, Santhoshkumar TR, Prabhakar J, Nair SA, Pillai MR. Drug-induced senescence generates chemoresistant stemlike cells with low reactive oxygen species. J Biol Chem. 2011;286(43):37813–29.
    1. Sharma SV, Lee DY, Li B, et al. A chromatin-mediated reversible drug-tolerant state in cancer cell subpopulations. Cell. 2010;141(1):69–80.
    1. Mosieniak G, Sikora E. Polyploidy: the link between senescence and cancer. Curr Pharm Des. 2010;16(6):734–40.
    1. Erenpreisa J, Kalejs M, Cragg MS. Mitotic catastrophe and endomitosis in tumour cells: an evolutionary key to a molecular solution. Cell Biol Int. 2005;29(12):1012–8.
    1. Vakifahmetoglu H, Olsson M, Zhivotovsky B. Death through a tragedy: mitotic catastrophe. Cell Death Differ. 2008;15(7):1153–62.
    1. Vakifahmetoglu H, Olsson M, Tamm C, Heidari N, Orrenius S, Zhivotovsky B. DNA damage induces two distinct modes of cell death in ovarian carcinomas. Cell Death Differ. 2008;15(3):555–66.
    1. Vitale I, Galluzzi L, Senovilla L, et al. Illicit survival of cancer cells during polyploidization and depolyploidization. Cell Death Differ. 2011;18(9):1403–13.
    1. Puig PE, Guilly MN, Bouchot A, et al. Tumor cells can escape DNA-damaging cisplatin through DNA endoreduplication and reversible polyploidy. Cell Biol Int. 2008;32(9):1031–43.
    1. Sundaram M, Guernsey DL, Rajaraman MM, Rajaraman R. Neosis: a novel type of cell division in cancer. Cancer Biol Ther. 2004;3(2):207–18.
    1. Rajaraman R, Rajaraman MM, Rajaraman SR, Guernsey DL. Neosis–a paradigm of self-renewal in cancer. Cell Biol Int. 2005;29(12):1084–97.
    1. Ianzini F, Kosmacek EA, Nelson ES, et al. Activation of meiosis-specific genes is associated with depolyploidization of human tumor cells following radiation-induced mitotic catastrophe. Cancer Res. 2009;69(6):2296–304.
    1. Salmina K, Jankevics E, Huna A, et al. Up-regulation of the embryonic self-renewal network through reversible polyploidy in irradiated p53-mutant tumour cells. Exp Cell Res. 2010;316(13):2099–112.
    1. Erenpreisa J, Salmina K, Huna A, et al. Polyploid tumour cells elicit paradiploid progeny through depolyploidizing divisions and regulated autophagic degradation. Cell Biol Int. 2011;35(7):687–95.
    1. Amaravadi RK. Autophagy-induced tumor dormancy in ovarian cancer. J Clin Invest. 2008;118(12):3837–40.
    1. Almog N. Molecular mechanisms underlying tumor dormancy. Cancer Letters. 2010;294(2):139–46.
    1. Lu Z, Luo RZ, Lu Y, et al. The tumor suppressor gene ARHI regulates autophagy and tumor dormancy in human ovarian cancer cells. J Clin Invest. 2008;118(12):3917–29.
    1. White DE, Rayment JH, Muller WJ. Addressing the role of cell adhesion in tumor cell dormancy. Cell Cycle. 2006;5(16):1756–9.
    1. Bast RC, Jr, Spriggs DR. More than a biomarker: CA125 may contribute to ovarian cancer pathogenesis. Gynecol Oncol. 2011;121(3):429–30.
    1. Rustin GJ, van der Burg ME, Griffin CL, et al. Early versus delayed treatment of relapsed ovarian cancer (MRC OV05/EORTC 55955): a randomised trial. Lancet. 2010;376(9747):1155–63.
    1. Goonewardene TI, Hall MR, Rustin GJ. Management of asymptomatic patients on follow-up for ovarian cancer with rising CA-125 concentrations. Lancet Oncol. 2007;8(9):813–21.
    1. Licun W, Tannock IF. Selective estrogen receptor modulators as inhibitors of repopulation of human breast cancer cell lines after chemotherapy. Clin Cancer Res. 2003;9(12):4614–8.
    1. Wu L, Birle DC, Tannock IF. Effects of the mammalian target of rapamycin inhibitor CCI-779 used alone or with chemotherapy on human prostate cancer cells and xenografts. Cancer Res. 2005;65(7):2825–31.

Source: PubMed

3
订阅