The Role of Cancer Stem Cell Markers in Ovarian Cancer
Abstract
:Simple Summary
Abstract
1. Introduction
2. Ovarian Cancer
3. Cancer Stem Cells
4. Interactions between CSC and Non-Tumour Cells
5. Targeting CSCs in Ovarian Cancer
6. Ovarian Cancer Stem Cells Markers
6.1. CD133 (Prominin-1)
6.2. CD44
6.3. ALDH1
6.4. CD24
6.5. CD117
6.6. Other Biomarkers Related to CSCs
7. Conclusions and Future Directions
Author Contributions
Funding
Conflicts of Interest
References
- Ottevanger, P.B. Ovarian cancer stem cells more questions than answers. Semin. Cancer Biol. 2017, 44, 67–71. [Google Scholar] [CrossRef] [PubMed]
- WCRF. Ovarian Cancer Statistics. 2020. Available online: https://www.wcrf.org/cancer-trends/ovarian-cancer-statistics/ (accessed on 9 October 2023).
- Saha, S.; Parte, S.; Roy, P.; Kakar, S.S. Ovarian Cancer Stem Cells: Characterization and Role in Tumorigenesis. Adv. Exp. Med. Biol. 2021, 1330, 151–169. [Google Scholar] [CrossRef] [PubMed]
- Doubeni, C.A.; Doubeni, A.R.; Myers, A.E. Diagnosis and Management of Ovarian Cancer. Am. Fam. Physician 2016, 93, 937–944. [Google Scholar] [PubMed]
- Lheureux, S.; Gourley, C.; Vergote, I.; Oza, A.M. Epithelial ovarian cancer. Lancet 2019, 393, 1240–1253. [Google Scholar] [CrossRef] [PubMed]
- Keyvani, V.; Farshchian, M.; Esmaeili, S.A.; Yari, H.; Moghbeli, M.; Nezhad, S.K.; Abbaszadegan, M.R. Ovarian cancer stem cells and targeted therapy. J. Ovarian Res. 2019, 12, 120. [Google Scholar] [CrossRef] [PubMed]
- Izycka, N.; Rucinski, M.; Andrzejewska, M.; Szubert, S.; Nowak-Markwitz, E.; Sterzynska, K. The Prognostic Value of Cancer Stem Cell Markers (CSCs) Expression—ALDH1A1, CD133, CD44—For Survival and Long-Term Follow-Up of Ovarian Cancer Patients. Int. J. Mol. Sci. 2023, 24, 2400. [Google Scholar] [CrossRef] [PubMed]
- Miree, O.; Srivastava, S.K.; Dasgupta, S.; Singh, S.; Rocconi, R.; Singh, A.P. Current and Futuristic Roadmap of Ovarian Cancer Management: An Overview. In Ovarian Cancer: Molecular & Diagnostic Imaging and Treatment Strategies; Advances in Experimental Medicine and Biology; Springer: Cham, Switzerland, 2021; Volume 1330, pp. 1–19. [Google Scholar] [CrossRef]
- Meinhold-Heerlein, I.; Hauptmann, S. The heterogeneity of ovarian cancer. Arch. Gynecol. Obstet. 2014, 289, 237–239. [Google Scholar] [CrossRef] [PubMed]
- Edson, M.A.; Nagaraja, A.K.; Matzuk, M.M. The mammalian ovary from genesis to revelation. Endocr. Rev. 2009, 30, 624–712. [Google Scholar] [CrossRef]
- Kurman, R.J.; Visvanathan, K.; Roden, R.; Wu, T.C.; Shih Ie, M. Early detection and treatment of ovarian cancer: Shifting from early stage to minimal volume of disease based on a new model of carcinogenesis. Am. J. Obstet. Gynecol. 2008, 198, 351–356. [Google Scholar] [CrossRef]
- Prat, J.; D’Angelo, E.; Espinosa, I. Ovarian carcinomas: At least five different diseases with distinct histological features and molecular genetics. Hum. Pathol. 2018, 80, 11–27. [Google Scholar] [CrossRef]
- Kurman, R.J.; Shih Ie, M. The Dualistic Model of Ovarian Carcinogenesis: Revisited, Revised, and Expanded. Am. J. Pathol. 2016, 186, 733–747. [Google Scholar] [CrossRef] [PubMed]
- Muñoz-Galván, S.; Carnero, A. Targeting Cancer Stem Cells to Overcome Therapy Resistance in Ovarian Cancer. Cells 2020, 9, 1402. [Google Scholar] [CrossRef] [PubMed]
- Fuller, P.J.; Leung, D.; Chu, S. Genetics and genomics of ovarian sex cord-stromal tumors. Clin. Genet. 2017, 91, 285–291. [Google Scholar] [CrossRef] [PubMed]
- Horta, M.; Cunha, T.M. Sex cord-stromal tumors of the ovary: A comprehensive review and update for radiologists. Diagn. Interv. Radiol. 2015, 21, 277–286. [Google Scholar] [CrossRef] [PubMed]
- Ramalingam, P. Germ Cell Tumors of the Ovary: A Review. Semin. Diagn. Pathol. 2023, 40, 22–36. [Google Scholar] [CrossRef] [PubMed]
- American Cancer Society. Ovarian Cancer Risk Factors; American Cancer Society: Tulsa, OK, USA, 2021. [Google Scholar]
- Cass, I.; Baldwin, R.L.; Varkey, T.; Moslehi, R.; Narod, S.A.; Karlan, B.Y. Improved survival in women with BRCA-associated ovarian carcinoma. Cancer 2003, 97, 2187–2195. [Google Scholar] [CrossRef] [PubMed]
- Kurman, R.J.; Shih, I.-M. Pathogenesis of ovarian cancer. Lessons from morphology and molecular biology and their clinical implications. Int. J. Gynecol. Pathol. Off. J. Int. Soc. Gynecol. Pathol. 2008, 27, 151–160. [Google Scholar]
- Mahajan, A.; Liu, Z.; Gellert, L.; Zou, X.; Yang, G.; Lee, P.; Yang, X.; Wei, J.J. HMGA2: A biomarker significantly overexpressed in high-grade ovarian serous carcinoma. Mod. Pathol. 2010, 23, 673–681. [Google Scholar] [CrossRef]
- Hunn, J.; Rodriguez, G.C. Ovarian cancer: Etiology, risk factors, and epidemiology. Clin. Obstet. Gynecol. 2012, 55, 3–23. [Google Scholar] [CrossRef]
- Vergote, I.; Tropé, C.G.; Amant, F.; Kristensen, G.B.; Ehlen, T.; Johnson, N.; Verheijen, R.H.; Van Der Burg, M.E.; Lacave, A.J.; Panici, P.B. Neoadjuvant chemotherapy or primary surgery in stage IIIC or IV ovarian cancer. N. Engl. J. Med. 2010, 363, 943–953. [Google Scholar] [CrossRef]
- Kehoe, S.; Hook, J.; Nankivell, M.; Jayson, G.C.; Kitchener, H.; Lopes, T.; Luesley, D.; Perren, T.; Bannoo, S.; Mascarenhas, M. Primary chemotherapy versus primary surgery for newly diagnosed advanced ovarian cancer (CHORUS): An open-label, randomised, controlled, non-inferiority trial. Lancet 2015, 386, 249–257. [Google Scholar] [CrossRef] [PubMed]
- Pignata, S.; Scambia, G.; Ferrandina, G.; Savarese, A.; Sorio, R.; Breda, E.; Gebbia, V.; Musso, P.; Frigerio, L.; Del Medico, P. Carboplatin plus paclitaxel versus carboplatin plus pegylated liposomal doxorubicin as first-line treatment for patients with ovarian cancer: The MITO-2 randomized phase III trial. J. Clin. Oncol. 2011, 29, 3628–3635. [Google Scholar] [CrossRef] [PubMed]
- Karam, A.; Ledermann, J.; Kim, J.-W.; Sehouli, J.; Lu, K.; Gourley, C.; Katsumata, N.; Burger, R.; Nam, B.-H.; Bacon, M. Fifth ovarian cancer consensus conference of the gynecologic cancer intergroup: First-line interventions. Ann. Oncol. 2017, 28, 711–717. [Google Scholar] [CrossRef] [PubMed]
- Katsumata, N.; Yasuda, M.; Isonishi, S.; Takahashi, F.; Michimae, H.; Kimura, E.; Aoki, D.; Jobo, T.; Kodama, S.; Terauchi, F. Long-term results of dose-dense paclitaxel and carboplatin versus conventional paclitaxel and carboplatin for treatment of advanced epithelial ovarian, fallopian tube, or primary peritoneal cancer (JGOG 3016): A randomised, controlled, open-label trial. Lancet Oncol. 2013, 14, 1020–1026. [Google Scholar] [CrossRef] [PubMed]
- Armstrong, D.K.; Bundy, B.; Wenzel, L.; Huang, H.Q.; Baergen, R.; Lele, S.; Copeland, L.J.; Walker, J.L.; Burger, R.A. Intraperitoneal cisplatin and paclitaxel in ovarian cancer. N. Engl. J. Med. 2006, 354, 34–43. [Google Scholar] [CrossRef] [PubMed]
- Ray-Coquard, I.; Mirza, M.R.; Pignata, S.; Walther, A.; Romero, I.; du Bois, A. Therapeutic options following second-line platinum-based chemotherapy in patients with recurrent ovarian cancer: Comparison of active surveillance and maintenance treatment. Cancer Treat. Rev. 2020, 90, 102107. [Google Scholar] [CrossRef]
- Soliman, A.A.; Elzarkaa, A.A.; Malik, E. Epithelial Ovarian Cancer and Cancer Stem Cells. Adv. Exp. Med. Biol. 2021, 1330, 21–32. [Google Scholar] [CrossRef]
- Kenda Suster, N.; Virant-Klun, I. Presence and role of stem cells in ovarian cancer. World J. Stem Cells 2019, 11, 383–397. [Google Scholar] [CrossRef]
- Fotopoulou, C. Limitations to the use of carboplatin-based therapy in advanced ovarian cancer. EJC Suppl. 2014, 12, 13–16. [Google Scholar] [CrossRef]
- Pieterse, Z.; Amaya-Padilla, M.A.; Singomat, T.; Binju, M.; Madjid, B.D.; Yu, Y.; Kaur, P. Ovarian cancer stem cells and their role in drug resistance. Int. J. Biochem. Cell Biol. 2019, 106, 117–126. [Google Scholar] [CrossRef]
- Zuber, E.; Schweitzer, D.; Allen, D.; Parte, S.; Kakar, S.S. Stem Cells in Ovarian Cancer and Potential Therapies. Proc. Stem Cell Res. Oncog. 2020, 8, e1001. [Google Scholar] [PubMed]
- Markowska, J.; Kojs, Z.; Twardawa, D. Cancer stem cells in targeted therapy. Curr. Gynecol. Oncol. 2018, 16, 96–100. [Google Scholar] [CrossRef]
- Lapidot, T.; Sirard, C.; Vormoor, J.; Murdoch, B.; Hoang, T.; Caceres-Cortes, J.; Minden, M.; Paterson, B.; Caligiuri, M.A.; Dick, J.E. A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature 1994, 367, 645–648. [Google Scholar] [CrossRef] [PubMed]
- Bapat, S.A.; Mali, A.M.; Koppikar, C.B.; Kurrey, N.K. Stem and progenitor-like cells contribute to the aggressive behavior of human epithelial ovarian cancer. Cancer Res. 2005, 65, 3025–3029. [Google Scholar] [CrossRef] [PubMed]
- Jubelin, C.; Muñoz-Garcia, J.; Cochonneau, D.; Moranton, E.; Heymann, M.F.; Heymann, D. Biological evidence of cancer stem-like cells and recurrent disease in osteosarcoma. Cancer Drug Resist. 2022, 5, 184–198. [Google Scholar] [CrossRef] [PubMed]
- Keyvani-Ghamsari, S.; Khorsandi, K.; Rasul, A.; Zaman, M.K. Current understanding of epigenetics mechanism as a novel target in reducing cancer stem cells resistance. Clin. Epigenetics 2021, 13, 120. [Google Scholar] [CrossRef] [PubMed]
- Atashzar, M.R.; Baharlou, R.; Karami, J.; Abdollahi, H.; Rezaei, R.; Pourramezan, F.; Zoljalali Moghaddam, S.H. Cancer stem cells: A review from origin to therapeutic implications. J. Cell. Physiol. 2020, 235, 790–803. [Google Scholar] [CrossRef] [PubMed]
- Nimmakayala, R.K.; Batra, S.K.; Ponnusamy, M.P. Unraveling the journey of cancer stem cells from origin to metastasis. Biochim. Biophys. Acta (BBA)-Rev. Cancer 2019, 1871, 50–63. [Google Scholar] [CrossRef]
- Wan Kamarul Zaman, W.S.; Nurul, A.A.; Nordin, F. Stem cells and cancer stem cells: The Jekyll and Hyde Scenario and their implications in stem cell therapy. Biomedicines 2021, 9, 1245. [Google Scholar] [CrossRef]
- Kitagawa, K.; Murata, A.; Matsuura, N.; Tohya, K.; Takaichi, S.; Monden, M.; Inoue, M. Epithelial-mesenchymal transformation of a newly established cell line from ovarian adenosarcoma by transforming growth factor-beta1. Int. J. Cancer 1996, 66, 91–97. [Google Scholar] [CrossRef]
- Rich, J.N. Cancer stem cells: Understanding tumor hierarchy and heterogeneity. Medicine 2016, 95 (Suppl. S1), S2–S7. [Google Scholar] [CrossRef] [PubMed]
- Al-Alem, L.F.; Pandya, U.M.; Baker, A.T.; Bellio, C.; Zarrella, B.D.; Clark, J.; DiGloria, C.M.; Rueda, B.R. Ovarian cancer stem cells: What progress have we made? Int. J. Biochem. Cell Biol. 2019, 107, 92–103. [Google Scholar] [CrossRef] [PubMed]
- Palorini, R.; Votta, G.; Balestrieri, C.; Monestiroli, A.; Olivieri, S.; Vento, R.; Chiaradonna, F. Energy Metabolism Characterization of a Novel Cancer Stem Cell-L ike Line 3 AB-OS. J. Cell. Biochem. 2014, 115, 368–379. [Google Scholar] [CrossRef] [PubMed]
- Pastò, A.; Bellio, C.; Pilotto, G.; Ciminale, V.; Silic-Benussi, M.; Guzzo, G.; Rasola, A.; Frasson, C.; Nardo, G.; Zulato, E. Cancer stem cells from epithelial ovarian cancer patients privilege oxidative phosphorylation, and resist glucose deprivation. Oncotarget 2014, 5, 4305–4319. [Google Scholar] [CrossRef] [PubMed]
- Szaryńska, M.; Olejniczak, A.; Kmieć, Z. The role of cancer stem cells in pathogenesis of colorectal cancer. Adv. Hyg. Exp. Med. 2016, 70, 1469–1482. [Google Scholar] [CrossRef] [PubMed]
- Królewska-Daszczyńska, P.; Wendlocha, D.; Smycz-Kubańska, M.; Stępień, S.; Mielczarek-Palacz, A. Cancer stem cells markers in ovarian cancer: Clinical and therapeutic significance (Review). Oncol. Lett. 2022, 24, 465. [Google Scholar] [CrossRef]
- Derks, L.L.M.; van Boxtel, R. Stem cell mutations, associated cancer risk, and consequences for regenerative medicine. Cell Stem Cell 2023, 30, 1421–1433. [Google Scholar] [CrossRef]
- Plaks, V.; Kong, N.; Werb, Z. The cancer stem cell niche: How essential is the niche in regulating stemness of tumor cells? Cell Stem Cell 2015, 16, 225–238. [Google Scholar] [CrossRef]
- Klemba, A.; Purzycka-Olewiecka, J.K.; Wcisło, G.; Czarnecka, A.M.; Lewicki, S.; Lesyng, B.; Szczylik, C.; Kieda, C. Surface markers of cancer stem-like cells of ovarian cancer and their clinical relevance. Contemp. Oncol. 2018, 22, 48–55. [Google Scholar] [CrossRef]
- Bar, J.K.; Grelewski, P.; Lis-Nawara, A.; Drobnikowska, K. The role of cancer stem cells in progressive growth and resistance of ovarian cancer: True or fiction? Adv. Hyg. Exp. Med. 2015, 69, 1077–1086. [Google Scholar]
- Bapat, S.A. Human ovarian cancer stem cells. Reproduction 2010, 140, 33–41. [Google Scholar] [CrossRef] [PubMed]
- Bighetti-Trevisan, R.L.; Sousa, L.O.; Castilho, R.M.; Almeida, L.O. Cancer Stem Cells: Powerful Targets to Improve Current Anticancer Therapeutics. Stem Cells Int. 2019, 2019, 9618065. [Google Scholar] [CrossRef] [PubMed]
- Prieto-Vila, M.; Takahashi, R.-u.; Usuba, W.; Kohama, I.; Ochiya, T. Drug resistance driven by cancer stem cells and their niche. Int. J. Mol. Sci. 2017, 18, 2574. [Google Scholar] [CrossRef] [PubMed]
- Shibata, M.; Hoque, M.O. Targeting Cancer Stem Cells: A Strategy for Effective Eradication of Cancer. Cancers 2019, 11, 732. [Google Scholar] [CrossRef] [PubMed]
- Konrad, C.V.; Murali, R.; Varghese, B.A.; Nair, R. The role of cancer stem cells in tumor heterogeneity and resistance to therapy. Can. J. Physiol. Pharmacol. 2017, 95, 1–15. [Google Scholar] [CrossRef] [PubMed]
- Ebben, J.D.; Treisman, D.M.; Zorniak, M.; Kutty, R.G.; Clark, P.A.; Kuo, J.S. The cancer stem cell paradigm: A new understanding of tumor development and treatment. Expert. Opin. Ther. Targets 2010, 14, 621–632. [Google Scholar] [CrossRef] [PubMed]
- Brabletz, T.; Jung, A.; Spaderna, S.; Hlubek, F.; Kirchner, T. Opinion: Migrating cancer stem cells—An integrated concept of malignant tumour progression. Nat. Rev. Cancer 2005, 5, 744–749. [Google Scholar] [CrossRef] [PubMed]
- Diaz-Cano, S.J. Tumor heterogeneity: Mechanisms and bases for a reliable application of molecular marker design. Int. J. Mol. Sci. 2012, 13, 1951–2011. [Google Scholar] [CrossRef]
- Tirino, V.; Desiderio, V.; Paino, F.; De Rosa, A.; Papaccio, F.; Fazioli, F.; Pirozzi, G.; Papaccio, G. Human primary bone sarcomas contain CD133+ cancer stem cells displaying high tumorigenicity in vivo. FASEB J. 2011, 25, 2022–2030. [Google Scholar] [CrossRef]
- Alwosaibai, K.; Aalmri, S.; Mashhour, M.; Ghandorah, S.; Alshangiti, A.; Azam, F.; Selwi, W.; Gharaibeh, L.; Alatawi, Y.; Alruwaii, Z.; et al. PD-L1 is highly expressed in ovarian cancer and associated with cancer stem cells populations expressing CD44 and other stem cell markers. BMC Cancer 2023, 23, 13. [Google Scholar] [CrossRef]
- Kise, K.; Kinugasa-Katayama, Y.; Takakura, N. Tumor microenvironment for cancer stem cells. Adv. Drug Deliv. Rev. 2016, 99 Pt B, 197–205. [Google Scholar] [CrossRef]
- Abdullah, L.N.; Chow, E.K. Mechanisms of chemoresistance in cancer stem cells. Clin. Transl. Med. 2013, 2, 3. [Google Scholar] [CrossRef] [PubMed]
- Eyre, R.; Harvey, I.; Stemke-Hale, K.; Lennard, T.W.J.; Tyson-Capper, A.; Meeson, A.P. Reversing paclitaxel resistance in ovarian cancer cells via inhibition of the ABCB1 expressing side population. Tumor Biol. 2014, 35, 9879–9892. [Google Scholar] [CrossRef] [PubMed]
- Hu, L.; McArthur, C.; Jaffe, R.B. Ovarian cancer stem-like side-population cells are tumourigenic and chemoresistant. Br. J. Cancer 2010, 102, 1276–1283. [Google Scholar] [CrossRef] [PubMed]
- Kim, D.K.; Seo, E.J.; Choi, E.J.; Lee, S.I.; Kwon, Y.W.; Jang, I.H.; Kim, S.-C.; Kim, K.-H.; Suh, D.-S.; Seong-Jang, K.; et al. Crucial role of HMGA1 in the self-renewal and drug resistance of ovarian cancer stem cells. Exp. Mol. Med. 2016, 48, e255. [Google Scholar] [CrossRef]
- Opferman, J.T.; Kothari, A. Anti-apoptotic BCL-2 family members in development. Cell Death Differ. 2018, 25, 37–45. [Google Scholar] [CrossRef] [PubMed]
- He, K.; Gan, W.J. Wnt/β-Catenin Signaling Pathway in the Development and Progression of Colorectal Cancer. Cancer Manag. Res. 2023, 15, 435–448. [Google Scholar] [CrossRef] [PubMed]
- Ranganathan, P.; Weaver, K.L.; Capobianco, A.J. Notch signalling in solid tumours: A little bit of everything but not all the time. Nat. Rev. Cancer 2011, 11, 338–351. [Google Scholar] [CrossRef]
- Dylla, S.J.; Beviglia, L.; Park, I.K.; Chartier, C.; Raval, J.; Ngan, L.; Pickell, K.; Aguilar, J.; Lazetic, S.; Smith-Berdan, S.; et al. Colorectal cancer stem cells are enriched in xenogeneic tumors following chemotherapy. PLoS ONE 2008, 3, e2428. [Google Scholar] [CrossRef]
- Davis, A.; Tinker, A.V.; Friedlander, M. “Platinum resistant” ovarian cancer: What is it, who to treat and how to measure benefit? Gynecol. Oncol. 2014, 133, 624–631. [Google Scholar] [CrossRef]
- Deng, J.; Bai, X.; Feng, X.; Ni, J.; Beretov, J.; Graham, P.; Li, Y. Inhibition of PI3K/Akt/mTOR signaling pathway alleviates ovarian cancer chemoresistance through reversing epithelial-mesenchymal transition and decreasing cancer stem cell marker expression. BMC Cancer 2019, 19, 618. [Google Scholar] [CrossRef] [PubMed]
- Altomare, D.A.; Wang, H.Q.; Skele, K.L.; De Rienzo, A.; Klein-Szanto, A.J.; Godwin, A.K.; Testa, J.R. AKT and mTOR phosphorylation is frequently detected in ovarian cancer and can be targeted to disrupt ovarian tumor cell growth. Oncogene 2004, 23, 5853–5857. [Google Scholar] [CrossRef] [PubMed]
- Abubaker, K.; Luwor, R.B.; Zhu, H.; McNally, O.; Quinn, M.A.; Burns, C.J.; Thompson, E.W.; Findlay, J.K.; Ahmed, N. Inhibition of the JAK2/STAT3 pathway in ovarian cancer results in the loss of cancer stem cell-like characteristics and a reduced tumor burden. BMC Cancer 2014, 14, 317. [Google Scholar] [CrossRef] [PubMed]
- Vathipadiekal, V.; Saxena, D.; Mok, S.C.; Hauschka, P.V.; Ozbun, L.; Birrer, M.J. Identification of a potential ovarian cancer stem cell gene expression profile from advanced stage papillary serous ovarian cancer. PLoS ONE 2012, 7, e29079. [Google Scholar] [CrossRef] [PubMed]
- Chau, W.K.; Ip, C.K.; Mak, A.S.; Lai, H.C.; Wong, A.S. c-Kit mediates chemoresistance and tumor-initiating capacity of ovarian cancer cells through activation of Wnt/β-catenin-ATP-binding cassette G2 signaling. Oncogene 2013, 32, 2767–2781. [Google Scholar] [CrossRef] [PubMed]
- Ray, A.; Meng, E.; Reed, E.; Shevde, L.A.; Rocconi, R.P. Hedgehog signaling pathway regulates the growth of ovarian cancer spheroid forming cells. Int. J. Oncol. 2011, 39, 797–804. [Google Scholar] [CrossRef] [PubMed]
- Chung, K.P.S.; Leung, R.W.H.; Lee, T.K.W. Hampering Stromal Cells in the Tumor Microenvironment as a Therapeutic Strategy to Destem Cancer Stem Cells. Cancers 2021, 13, 3191. [Google Scholar] [CrossRef] [PubMed]
- Korkaya, H.; Liu, S.; Wicha, M.S. Breast cancer stem cells, cytokine networks, and the tumor microenvironment. J. Clin. Investig. 2011, 121, 3804–3809. [Google Scholar] [CrossRef]
- Lee, T.K.W.; Castilho, A.; Cheung, V.C.H.; Tang, K.H.; Ma, S.; Ng, I.O.L. CD24+ liver tumor-initiating cells drive self-renewal and tumor initiation through STAT3-mediated NANOG regulation. Cell Stem Cell 2011, 9, 50–63. [Google Scholar] [CrossRef]
- Izar, B.; Tirosh, I.; Stover, E.H.; Wakiro, I.; Cuoco, M.S.; Alter, I.; Rodman, C.; Leeson, R.; Su, M.-J.; Shah, P. A single-cell landscape of high-grade serous ovarian cancer. Nat. Med. 2020, 26, 1271–1279. [Google Scholar] [CrossRef]
- Kommoss, S.; Winterhoff, B.; Oberg, A.L.; Konecny, G.E.; Wang, C.; Riska, S.M.; Fan, J.-B.; Maurer, M.J.; April, C.; Shridhar, V. Bevacizumab may differentially improve ovarian cancer outcome in patients with proliferative and mesenchymal molecular subtypes. Clin. Cancer Res. 2017, 23, 3794–3801. [Google Scholar] [CrossRef] [PubMed]
- Raghavan, S.; Snyder, C.S.; Wang, A.; McLean, K.; Zamarin, D.; Buckanovich, R.J.; Mehta, G. Carcinoma-Associated Mesenchymal Stem Cells Promote Chemoresistance in Ovarian Cancer Stem Cells via PDGF Signaling. Cancers 2020, 12, 2063. [Google Scholar] [CrossRef] [PubMed]
- Miranda, A.; Hamilton, P.T.; Zhang, A.W.; Pattnaik, S.; Becht, E.; Mezheyeuski, A.; Bruun, J.; Micke, P.; de Reynies, A.; Nelson, B.H. Cancer stemness, intratumoral heterogeneity, and immune response across cancers. Proc. Natl. Acad. Sci. USA 2019, 116, 9020–9029. [Google Scholar] [CrossRef] [PubMed]
- Quatromoni, J.G.; Eruslanov, E. Tumor-associated macrophages: Function, phenotype, and link to prognosis in human lung cancer. Am. J. Transl. Res. 2012, 4, 376–389. [Google Scholar] [PubMed]
- Raghavan, S.; Mehta, P.; Xie, Y.; Lei, Y.L.; Mehta, G. Ovarian cancer stem cells and macrophages reciprocally interact through the WNT pathway to promote pro-tumoral and malignant phenotypes in 3D engineered microenvironments. J. Immunother. Cancer 2019, 7, 190. [Google Scholar] [CrossRef]
- Wang, C.; Lin, Y.; Zhu, H.; Zhou, Y.; Mao, F.; Huang, X.; Sun, Q.; Li, C. The Prognostic and Clinical Value of Tumor-Associated Macrophages in Patients With Breast Cancer: A Systematic Review and Meta-Analysis. Front. Oncol. 2022, 12, 905846. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Q.-W.; Liu, L.; Gong, C.-Y.; Shi, H.-S.; Zeng, Y.-H.; Wang, X.-Z.; Zhao, Y.-W.; Wei, Y.-Q. Prognostic Significance of Tumor-Associated Macrophages in Solid Tumor: A Meta-Analysis of the Literature. PLoS ONE 2012, 7, e50946. [Google Scholar] [CrossRef]
- Zhu, S.; Zhang, T.; Zheng, L.; Liu, H.; Song, W.; Liu, D.; Li, Z.; Pan, C.-X. Combination strategies to maximize the benefits of cancer immunotherapy. J. Hematol. Oncol. 2021, 14, 156. [Google Scholar] [CrossRef]
- Wu, Y.; Yi, M.; Niu, M.; Mei, Q.; Wu, K. Myeloid-derived suppressor cells: An emerging target for anticancer immunotherapy. Mol. Cancer 2022, 21, 184. [Google Scholar] [CrossRef]
- Gabrilovich, D.I.; Nagaraj, S. Myeloid-derived suppressor cells as regulators of the immune system. Nat. Rev. Immunol. 2009, 9, 162–174. [Google Scholar] [CrossRef]
- Komura, N.; Mabuchi, S.; Shimura, K.; Yokoi, E.; Kozasa, K.; Kuroda, H.; Takahashi, R.; Sasano, T.; Kawano, M.; Matsumoto, Y. The role of myeloid-derived suppressor cells in increasing cancer stem-like cells and promoting PD-L1 expression in epithelial ovarian cancer. Cancer Immunol. Immunother. 2020, 69, 2477–2499. [Google Scholar] [CrossRef] [PubMed]
- Lu, C.; Redd, P.S.; Lee, J.R.; Savage, N.; Liu, K. The expression profiles and regulation of PD-L1 in tumor-induced myeloid-derived suppressor cells. Oncoimmunology 2016, 5, e1247135. [Google Scholar] [CrossRef] [PubMed]
- Ohms, M.; Möller, S.; Laskay, T. An Attempt to Polarize Human Neutrophils Toward N1 and N2 Phenotypes in vitro. Front. Immunol. 2020, 11, 532. [Google Scholar] [CrossRef] [PubMed]
- Que, H.; Fu, Q.; Lan, T.; Tian, X.; Wei, X. Tumor-associated neutrophils and neutrophil-targeted cancer therapies. Biochim. Biophys. Acta (BBA)-Rev. Cancer 2022, 1877, 188762. [Google Scholar] [CrossRef] [PubMed]
- Sato, S.; Matsushita, H.; Shintani, D.; Kobayashi, Y.; Fujieda, N.; Yabuno, A.; Nishikawa, T.; Fujiwara, K.; Kakimi, K.; Hasegawa, K. Association between effector-type regulatory T cells and immune checkpoint expression on CD8+ T cells in malignant ascites from epithelial ovarian cancer. BMC Cancer 2022, 22, 437. [Google Scholar] [CrossRef] [PubMed]
- Napoletano, C.; Bellati, F.; Ruscito, I.; Pernice, M.; Zizzari, I.G.; Caponnetto, S.; Tomao, F.; Frigerio, L.; Liberati, M.; Rughetti, A. Immunological and clinical impact of cancer stem cells in vulvar cancer: Role of CD133/CD24/ABCG2-expressing cells. Anticancer Res. 2016, 36, 5109–5116. [Google Scholar] [CrossRef] [PubMed]
- Ganguly, D.; Chandra, R.; Karalis, J.; Teke, M.; Aguilera, T.; Maddipati, R.; Wachsmann, M.B.; Ghersi, D.; Siravegna, G.; Zeh, H.J., III. Cancer-associated fibroblasts: Versatile players in the tumor microenvironment. Cancers 2020, 12, 2652. [Google Scholar] [CrossRef]
- Tokuda, K.; Morine, Y.; Miyazaki, K.; Yamada, S.; Saito, Y.; Nishi, M.; Tokunaga, T.; Ikemoto, T.; Imura, S.; Shimada, M. The interaction between cancer associated fibroblasts and tumor associated macrophages via the osteopontin pathway in the tumor microenvironment of hepatocellular carcinoma. Oncotarget 2021, 12, 333–343. [Google Scholar] [CrossRef]
- Lewis, S.; Jones, D. The Role of Cancer Associated Fibroblasts in Maintaining Cancer Stem Cells. FASEB J. 2020, 34, 1. [Google Scholar] [CrossRef]
- You, J.; Li, M.; Cao, L.; Gu, Q.; Deng, P.; Tan, Y.; Hu, C. Snail1-dependent cancer-associated fibroblasts induce epithelial-mesenchymal transition in lung cancer cells via exosomes. QJM Int. J. Med. 2019, 112, 581–590. [Google Scholar] [CrossRef]
- Khaledian, B.; Thibes, L.; Shimono, Y. Adipocyte regulation of cancer stem cells. Cancer Sci. 2023, 114, 4134–4144. [Google Scholar] [CrossRef] [PubMed]
- Pagnotta, P.; Gantov, M.; Fletcher, S.; Lombardi, A.; Crosbie, M.L.; Santiso, N.; Ursino, A.; Frascarolli, C.; Amato, A.; Dreszman, R.; et al. Peritumoral adipose tissue promotes lipolysis and white adipocytes browning by paracrine action. Front. Endocrinol. 2023, 14, 1144016. [Google Scholar] [CrossRef] [PubMed]
- Lamb, R.; Ablett, M.P.; Spence, K.; Landberg, G.; Sims, A.H.; Clarke, R.B. Wnt pathway activity in breast cancer sub-types and stem-like cells. PLoS ONE 2013, 8, e67811. [Google Scholar] [CrossRef] [PubMed]
- Cao, Y. Adipocyte and lipid metabolism in cancer drug resistance. J. Clin. Investig. 2019, 129, 3006–3017. [Google Scholar] [CrossRef] [PubMed]
- Iyoshi, S.; Yoshihara, M.; Nakamura, K.; Sugiyama, M.; Koya, Y.; Kitami, K.; Uno, K.; Mogi, K.; Tano, S.; Tomita, H. Pro-tumoral behavior of omental adipocyte-derived fibroblasts in tumor microenvironment at the metastatic site of ovarian cancer. Int. J. Cancer 2021, 149, 1961–1972. [Google Scholar] [CrossRef] [PubMed]
- Cardenas, C.; Montagna, M.K.; Pitruzzello, M.; Lima, E.; Mor, G.; Alvero, A.B. Adipocyte microenvironment promotes Bcl xl expression and confers chemoresistance in ovarian cancer cells. Apoptosis 2017, 22, 558–569. [Google Scholar] [CrossRef] [PubMed]
- Di Franco, S.; Bianca, P.; Sardina, D.S.; Turdo, A.; Gaggianesi, M.; Veschi, V.; Nicotra, A.; Mangiapane, L.R.; Lo Iacono, M.; Pillitteri, I. Adipose stem cell niche reprograms the colorectal cancer stem cell metastatic machinery. Nat. Commun. 2021, 12, 5006. [Google Scholar] [CrossRef] [PubMed]
- Ji, Y.; Ge, Q.; Zhang, W.; Qu, P. Molecular Features, Prognostic Value, and Cancer Immune Interactions of Angiogenesis-Related Genes in Ovarian Cancer. Reprod. Sci. 2023, 30, 1637–1650. [Google Scholar] [CrossRef]
- Iliopoulos, D.; Hirsch, H.A.; Struhl, K. Metformin decreases the dose of chemotherapy for prolonging tumor remission in mouse xenografts involving multiple cancer cell types. Cancer Res. 2011, 71, 3196–3201. [Google Scholar] [CrossRef]
- Shank, J.J.; Yang, K.; Ghannam, J.; Cabrera, L.; Johnston, C.J.; Reynolds, R.K.; Buckanovich, R.J. Metformin targets ovarian cancer stem cells in vitro and in vivo. Gynecol. Oncol. 2012, 127, 390–397. [Google Scholar] [CrossRef]
- Brown, J.R.; Chan, D.K.; Shank, J.J.; Griffith, K.A.; Fan, H.; Szulawski, R.; Yang, K.; Reynolds, R.K.; Johnston, C.; McLean, K.; et al. Phase II clinical trial of metformin as a cancer stem cell-targeting agent in ovarian cancer. JCI Insight 2020, 5, e133247. [Google Scholar] [CrossRef] [PubMed]
- Landen, C.N.; Buckanovich, R.J.; Sill, M.; Mannel, R.S.; Walker, J.L.; Disilvestro, P.; Mathews, C.A.; Mutch, D.G.; Hernandez, M.; Martin, L.P.; et al. A phase I/II study of ruxolitinib with frontline neoadjuvant and post-surgical therapy in patients with advanced epithelial ovarian, Fallopian tube, or primary peritoneal cancer. J. Clin. Oncol. 2022, 40 (Suppl. S16), 5501. [Google Scholar] [CrossRef]
- Ning, N.; Pan, Q.; Zheng, F.; Teitz-Tennenbaum, S.; Egenti, M.; Yet, J.; Li, M.; Ginestier, C.; Wicha, M.S.; Moyer, J.S.; et al. Cancer stem cell vaccination confers significant antitumor immunity. Cancer Res. 2012, 72, 1853–1864. [Google Scholar] [CrossRef] [PubMed]
- Markowska, A.; Sajdak, S.; Huczyński, A.; Rehlis, S.; Markowska, J. Ovarian cancer stem cells: A target for oncological therapy. Adv. Clin. Exp. Med. 2018, 27, 1017–1020. [Google Scholar] [CrossRef] [PubMed]
- Chung, H.; Kim, Y.H.; Kwon, M.; Shin, S.J.; Kwon, S.H.; Cha, S.D.; Cho, C.H. The effect of salinomycin on ovarian cancer stem-like cells. Obstet. Gynecol. Sci. 2016, 59, 261–268. [Google Scholar] [CrossRef] [PubMed]
- Lee, H.G.; Shin, S.J.; Chung, H.W.; Kwon, S.H.; Cha, S.D.; Lee, J.E.; Cho, C.H. Salinomycin reduces stemness and induces apoptosis on human ovarian cancer stem cell. J. Gynecol. Oncol. 2017, 28, e14. [Google Scholar] [CrossRef] [PubMed]
- Wen, Y.; Hou, Y.; Huang, Z.; Cai, J.; Wang, Z. SOX2 is required to maintain cancer stem cells in ovarian cancer. Cancer Sci. 2017, 108, 719–731. [Google Scholar] [CrossRef]
- Mi, Y.; Huang, Y.; Deng, J. The enhanced delivery of salinomycin to CD133(+) ovarian cancer stem cells through CD133 antibody conjugation with poly(lactic-co-glycolic acid)-poly(ethylene glycol) nanoparticles. Oncol. Lett. 2018, 15, 6611–6621. [Google Scholar] [CrossRef]
- Berridge, M.J.; Bootman, M.D.; Roderick, H.L. Calcium signalling: Dynamics, homeostasis and remodelling. Nat. Rev. Mol. Cell Biol. 2003, 4, 517–529. [Google Scholar] [CrossRef]
- Lee, H.; Kim, J.W.; Kim, D.K.; Choi, D.K.; Lee, S.; Yu, J.H.; Kwon, O.B.; Lee, J.; Lee, D.S.; Kim, J.H.; et al. Calcium Channels as Novel Therapeutic Targets for Ovarian Cancer Stem Cells. Int. J. Mol. Sci. 2020, 21, 2327. [Google Scholar] [CrossRef]
- Lee, H.; Kim, J.W.; Lee, D.S.; Min, S.H. Combined Poziotinib with Manidipine Treatment Suppresses Ovarian Cancer Stem-Cell Proliferation and Stemness. Int. J. Mol. Sci. 2020, 21, 7379. [Google Scholar] [CrossRef] [PubMed]
- Jiang, L.-y.; Zhang, X.-l.; Du, P.; Zheng, J.-h. γ-Secretase inhibitor, DAPT inhibits self-renewal and stemness maintenance of ovarian cancer stem-like cells in vitro. Chin. J. Cancer Res. 2011, 23, 140–146. [Google Scholar] [CrossRef] [PubMed]
- Kakar, S.S.; Ratajczak, M.Z.; Powell, K.S.; Moghadamfalahi, M.; Miller, D.M.; Batra, S.K.; Singh, S.K. Withaferin a alone and in combination with cisplatin suppresses growth and metastasis of ovarian cancer by targeting putative cancer stem cells. PLoS ONE 2014, 9, e107596. [Google Scholar] [CrossRef] [PubMed]
- Ediriweera, M.K.; Tennekoon, K.H.; Samarakoon, S.R. Role of the PI3K/AKT/mTOR signaling pathway in ovarian cancer: Biological and therapeutic significance. Semin. Cancer Biol. 2019, 59, 147–160. [Google Scholar] [CrossRef] [PubMed]
- Green, J.M.; Alvero, A.B.; Kohen, F.; Mor, G. 7-(O)-Carboxymethyl daidzein conjugated to Nt-Boc-hexylenediamine: A novel compound capable of inducing cell death in epithelial ovarian cancer stem cells. Cancer Biol. Ther. 2009, 8, 1747–1753. [Google Scholar] [CrossRef] [PubMed]
- Casagrande, F.; Cocco, E.; Bellone, S.; Richter, C.E.; Bellone, M.; Todeschini, P.; Siegel, E.; Varughese, J.; Arin-Silasi, D.; Azodi, M. Eradication of chemotherapy-resistant CD44+ human ovarian cancer stem cells in mice by intraperitoneal administration of clostridium perfringens enterotoxin. Cancer 2011, 117, 5519–5528. [Google Scholar] [CrossRef] [PubMed]
- Burgos-Ojeda, D.; Rueda, B.R.; Buckanovich, R.J. Ovarian cancer stem cell markers: Prognostic and therapeutic implications. Cancer Lett. 2012, 322, 1–7. [Google Scholar] [CrossRef]
- Liu, M.; Mor, G.; Cheng, H.; Xiang, X.; Hui, P.; Rutherford, T.; Yin, G.; Rimm, D.L.; Holmberg, J.; Alvero, A.; et al. High frequency of putative ovarian cancer stem cells with CD44/CK19 coexpression is associated with decreased progression-free intervals in patients with recurrent epithelial ovarian cancer. Reprod. Sci. 2013, 20, 605–615. [Google Scholar] [CrossRef]
- Ran, X.; Zhou, P.; Zhang, K. Autophagy plays an important role in stemness mediation and the novel dual function of EIG121 in both autophagy and stemness regulation of endometrial carcinoma JEC cells. Int. J. Oncol. 2017, 51, 644–656. [Google Scholar] [CrossRef]
- Roy, L.; Bobbs, A.; Sattler, R.; Kurkewich, J.L.; Dausinas, P.B.; Nallathamby, P.; Cowden Dahl, K.D. CD133 Promotes Adhesion to the Ovarian Cancer Metastatic Niche. Cancer Growth Metastasis 2018, 11, 1179064418767882. [Google Scholar] [CrossRef]
- Min, K.-J.; So, K.A.; Ouh, Y.-T.; Hong, J.-H.; Lee, J.-K. The effects of DNA methylation and epigenetic factors on the expression of CD133 in ovarian cancers. J. Ovarian Res. 2012, 5, 28. [Google Scholar] [CrossRef] [PubMed]
- Ferrandina, G.; Bonanno, G.; Pierelli, L.; Perillo, A.; Procoli, A.; Mariotti, A.; Corallo, M.; Martinelli, E.; Rutella, S.; Paglia, A. Expression of CD133-1 and CD133-2 in ovarian cancer. Int. J. Gynecol. Cancer 2008, 18, 506–514. [Google Scholar] [CrossRef] [PubMed]
- Zhou, Q.; Chen, A.; Song, H.; Tao, J.; Yang, H.; Zuo, M. Prognostic value of cancer stem cell marker CD133 in ovarian cancer: A meta-analysis. Int. J. Clin. Exp. Med. 2015, 8, 3080–3088. [Google Scholar] [PubMed]
- Ruscito, I.; Castillo-Tong, D.C.; Vergote, I.; Ignat, I.; Stanske, M.; Vanderstichele, A.; Ganapathi, R.N.; Glajzer, J.; Kulbe, H.; Trillsch, F. Exploring the clonal evolution of CD133/aldehyde-dehydrogenase-1 (ALDH1)-positive cancer stem-like cells from primary to recurrent high-grade serous ovarian cancer (HGSOC). A study of the Ovarian Cancer Therapy–Innovative Models Prolong Survival (OCTIPS) Consortium. Eur. J. Cancer 2017, 79, 214–225. [Google Scholar] [PubMed]
- Tao, Y.; Li, H.; Huang, R.; Mo, D.; Zeng, T.; Fang, M.; Li, M. Clinicopathological and Prognostic Significance of Cancer Stem Cell Markers in Ovarian Cancer Patients: Evidence from 52 Studies. Cell Physiol. Biochem. 2018, 46, 1716–1726. [Google Scholar] [CrossRef] [PubMed]
- Steg, A.D.; Bevis, K.S.; Katre, A.A.; Ziebarth, A.; Dobbin, Z.C.; Alvarez, R.D.; Zhang, K.; Conner, M.; Landen, C.N. Stem cell pathways contribute to clinical chemoresistance in ovarian cancer. Clin. Cancer Res. 2012, 18, 869–881. [Google Scholar] [CrossRef] [PubMed]
- Liu, B.L.; Liu, S.J.; Baskys, A.; Cheng, H.; Han, Y.; Xie, C.; Song, H.; Li, J.; Xin, X.Y. Platinum sensitivity and CD133 expression as risk and prognostic predictors of central nervous system metastases in patients with epithelial ovarian cancer. BMC Cancer 2014, 14, 829. [Google Scholar] [CrossRef]
- Zhang, J.; Guo, X.; Chang, D.Y.; Rosen, D.G.; Mercado-Uribe, I.; Liu, J. CD133 expression associated with poor prognosis in ovarian cancer. Mod. Pathol. 2012, 25, 456–464. [Google Scholar] [CrossRef]
- Kryczek, I.; Liu, S.; Roh, M.; Vatan, L.; Szeliga, W.; Wei, S.; Banerjee, M.; Mao, Y.; Kotarski, J.; Wicha, M.S. Expression of aldehyde dehydrogenase and CD133 defines ovarian cancer stem cells. Int. J. Cancer 2012, 130, 29–39. [Google Scholar] [CrossRef]
- Silva, I.A.; Bai, S.; McLean, K.; Yang, K.; Griffith, K.; Thomas, D.; Ginestier, C.; Johnston, C.; Kueck, A.; Reynolds, R.K. Aldehyde Dehydrogenase in Combination with CD133 Defines Angiogenic Ovarian Cancer Stem Cells That Portend Poor Patient SurvivalALDH and CD133 Define Ovarian Cancer Stem Cells. Cancer Res. 2011, 71, 3991–4001. [Google Scholar] [CrossRef]
- Coffman, L.; Mooney, C.; Lim, J.; Bai, S.; Silva, I.; Gong, Y.; Yang, K.; Buckanovich, R.J. Endothelin receptor-A is required for the recruitment of antitumor T cells and modulates chemotherapy induction of cancer stem cells. Cancer Biol. Ther. 2013, 14, 184–192. [Google Scholar] [CrossRef] [PubMed]
- Long, H.; Xie, R.; Xiang, T.; Zhao, Z.; Lin, S.; Liang, Z.; Chen, Z.; Zhu, B. Autocrine CCL5 signaling promotes invasion and migration of CD133+ ovarian cancer stem-like cells via NF-κB-mediated MMP-9 upregulation. Stem Cells 2012, 30, 2309–2319. [Google Scholar] [CrossRef] [PubMed]
- Xiang, T.; Long, H.; He, L.; Han, X.; Lin, K.; Liang, Z.; Zhuo, W.; Xie, R.; Zhu, B. Interleukin-17 produced by tumor microenvironment promotes self-renewal of CD133+ cancer stem-like cells in ovarian cancer. Oncogene 2015, 34, 165–176. [Google Scholar] [CrossRef] [PubMed]
- Wu, Q.; Guo, R.; Lin, M.; Zhou, B.; Wang, Y. MicroRNA-200a inhibits CD133/1+ ovarian cancer stem cells migration and invasion by targeting E-cadherin repressor ZEB2. Gynecol. Oncol. 2011, 122, 149–154. [Google Scholar] [CrossRef] [PubMed]
- Ponnusamy, M.P.; Seshacharyulu, P.; Vaz, A.; Dey, P.; Batra, S.K. MUC4 stabilizes HER2 expression and maintains the cancer stem cell population in ovarian cancer cells. J. Ovarian Res. 2011, 4, 7. [Google Scholar] [CrossRef] [PubMed]
- Sherman, L.; Sleeman, J.; Herrlich, P.; Ponta, H. Hyaluronate receptors: Key players in growth, differentiation, migration and tumor progression. Curr. Opin. Cell Biol. 1994, 6, 726–733. [Google Scholar] [CrossRef] [PubMed]
- Louderbough, J.M.V.; Schroeder, J.A. Understanding the Dual Nature of CD44 in Breast Cancer Progression. Mol. Cancer Res. 2011, 9, 1573–1586. [Google Scholar] [CrossRef] [PubMed]
- Siegelman, M.H.; DeGrendele, H.C.; Estess, P. Activation and interaction of CD44 and hyaluronan in immunological systems. J. Leukoc. Biol. 1999, 66, 315–321. [Google Scholar] [CrossRef]
- Li, Y.; Jiang, D.; Liang, J.; Meltzer, E.B.; Gray, A.; Miura, R.; Wogensen, L.; Yamaguchi, Y.; Noble, P.W. Severe lung fibrosis requires an invasive fibroblast phenotype regulated by hyaluronan and CD44. J. Exp. Med. 2011, 208, 1459–1471. [Google Scholar] [CrossRef]
- Orian-Rousseau, V.; Morrison, H.; Matzke, A.; Kastilan, T.; Pace, G.; Herrlich, P.; Ponta, H. Hepatocyte growth factor-induced Ras activation requires ERM proteins linked to both CD44v6 and F-actin. Mol. Biol. Cell 2007, 18, 76–83. [Google Scholar] [CrossRef]
- Jain, M.; He, Q.; Lee, W.S.; Kashiki, S.; Foster, L.C.; Tsai, J.C.; Lee, M.E.; Haber, E. Role of CD44 in the reaction of vascular smooth muscle cells to arterial wall injury. J. Clin. Investig. 1996, 97, 596–603. [Google Scholar] [CrossRef]
- Slevin, M.; Krupinski, J.; Gaffney, J.; Matou, S.; West, D.; Delisser, H.; Savani, R.C.; Kumar, S. Hyaluronan-mediated angiogenesis in vascular disease: Uncovering RHAMM and CD44 receptor signaling pathways. Matrix Biol. 2007, 26, 58–68. [Google Scholar] [CrossRef]
- Elzarkaa, A.A.; Sabaa, B.E.; Abdelkhalik, D.; Mansour, H.; Melis, M.; Shaalan, W.; Farouk, M.; Malik, E.; Soliman, A.A. Clinical relevance of CD44 surface expression in advanced stage serous epithelial ovarian cancer: A prospective study. J. Cancer Res. Clin. Oncol. 2016, 142, 949–958. [Google Scholar] [CrossRef]
- Sihombing, U.H.M.; Andrijono, A.; Purwoto, G.; Gandamihardja, S.; Harahap, A.R.; Rustamadji, P.; Kekalih, A.; Widyawati, R.; Fuady, D.R. CD44+/CD24- Expression as predictors of ovarian cancer chemoresistance: Immunohistochemistry and flow cytometry study. J. Egypt. Natl. Cancer Inst. 2022, 34, 44. [Google Scholar] [CrossRef]
- Kraushaar, D.C.; Yamaguchi, Y.; Wang, L. Heparan sulfate is required for embryonic stem cells to exit from self-renewal. J. Biol. Chem. 2010, 285, 5907–5916. [Google Scholar] [CrossRef]
- Gomez, K.E.; Wu, F.; Keysar, S.B.; Morton, J.J.; Miller, B.; Chimed, T.S.; Le, P.N.; Nieto, C.; Chowdhury, F.N.; Tyagi, A.; et al. Cancer Cell CD44 Mediates Macrophage/Monocyte-Driven Regulation of Head and Neck Cancer Stem Cells. Cancer Res. 2020, 80, 4185–4198. [Google Scholar] [CrossRef]
- Bourguignon, L.Y.; Peyrollier, K.; Xia, W.; Gilad, E. Hyaluronan-CD44 interaction activates stem cell marker Nanog, Stat-3-mediated MDR1 gene expression, and ankyrin-regulated multidrug efflux in breast and ovarian tumor cells. J. Biol. Chem. 2008, 283, 17635–17651. [Google Scholar] [CrossRef]
- Chanmee, T.; Ontong, P.; Mochizuki, N.; Kongtawelert, P.; Konno, K.; Itano, N. Excessive hyaluronan production promotes acquisition of cancer stem cell signatures through the coordinated regulation of Twist and the transforming growth factor β (TGF-β)-Snail signaling axis. J. Biol. Chem. 2014, 289, 26038–26056. [Google Scholar] [CrossRef]
- Anttila, M.A.; Tammi, R.H.; Tammi, M.I.; Syrjänen, K.J.; Saarikoski, S.V.; Kosma, V.-M. High levels of stromal hyaluronan predict poor disease outcome in epithelial ovarian cancer. Cancer Res. 2000, 60, 150–155. [Google Scholar]
- Salmi, M.; Karikoski, M.; Elima, K.; Rantakari, P.; Jalkanen, S. CD44 Binds to Macrophage Mannose Receptor on Lymphatic Endothelium and Supports Lymphocyte Migration via Afferent Lymphatics. Circ. Res. 2013, 112, 1577–1582. [Google Scholar] [CrossRef]
- Zhou, J.; Du, Y.; Lu, Y.; Luan, B.; Xu, C.; Yu, Y.; Zhao, H. CD44 Expression Predicts Prognosis of Ovarian Cancer Patients through Promoting Epithelial-Mesenchymal Transition (EMT) by Regulating Snail, ZEB1, and Caveolin-1. Front. Oncol. 2019, 9, 802. [Google Scholar] [CrossRef]
- Zhu, Y.; Zhang, H.; Zhang, G.; Shi, Y.; Huang, J. Co-expression of CD44/MyD88 is a poor prognostic factor in advanced epithelial ovarian cancer. Ann. Transl. Med. 2019, 7, 91. [Google Scholar] [CrossRef]
- Sihombing, U.; Andrijono, A.; Purwoto, G.; Gandamihardja, S.; Harahap, A.; Rustamadji, P.; Kekalih, A.; Widyawati, R.; Fuady, D. Expression of CD44+/CD24-, RAD6 and DDB2 on chemotherapy response in ovarian Cancer: A prospective flow cytometry study. Gynecol. Oncol. Rep. 2022, 42, 101005. [Google Scholar] [CrossRef]
- Lin, J.; Ding, D. The prognostic role of the cancer stem cell marker CD44 in ovarian cancer: A meta-analysis. Cancer Cell Int. 2017, 17, 8. [Google Scholar] [CrossRef]
- Zhang, J.; Yuan, B.; Zhang, H.; Li, H. Human epithelial ovarian cancer cells expressing CD105, CD44 and CD106 surface markers exhibit increased invasive capacity and drug resistance. Oncol. Lett. 2019, 17, 5351–5360. [Google Scholar] [CrossRef]
- Alvero, A.B.; Chen, R.; Fu, H.H.; Montagna, M.; Schwartz, P.E.; Rutherford, T.; Silasi, D.A.; Steffensen, K.D.; Waldstrom, M.; Visintin, I.; et al. Molecular phenotyping of human ovarian cancer stem cells unravels the mechanisms for repair and chemoresistance. Cell Cycle 2009, 8, 158–166. [Google Scholar] [CrossRef]
- Zhang, H.-F.; Hu, P.; Fang, S.-Q. Understanding the role of CD44V6 in ovarian cancer. Oncol. Lett. 2017, 14, 1989–1992. [Google Scholar] [CrossRef]
- Motohara, T.; Fujimoto, K.; Tayama, S.; Narantuya, D.; Sakaguchi, I.; Tashiro, H.; Katabuchi, H. CD44 Variant 6 as a Predictive Biomarker for Distant Metastasis in Patients With Epithelial Ovarian Cancer. Obstet. Gynecol. 2016, 127, 1003–1011. [Google Scholar] [CrossRef]
- Shah, V.; Taratula, O.; Garbuzenko, O.B.; Taratula, O.R.; Rodriguez-Rodriguez, L.; Minko, T. Targeted nanomedicine for suppression of CD44 and simultaneous cell death induction in ovarian cancer: An optimal delivery of siRNA and anticancer drug. Clin. Cancer Res. 2013, 19, 6193–6204. [Google Scholar] [CrossRef]
- Sosulski, A.; Horn, H.; Zhang, L.; Coletti, C.; Vathipadiekal, V.; Castro, C.M.; Birrer, M.J.; Nagano, O.; Saya, H.; Lage, K.; et al. CD44 Splice Variant v8-10 as a Marker of Serous Ovarian Cancer Prognosis. PLoS ONE 2016, 11, e0156595. [Google Scholar] [CrossRef]
- Zhang, S.; Balch, C.; Chan, M.W.; Lai, H.-C.; Matei, D.; Schilder, J.M.; Yan, P.S.; Huang, T.H.; Nephew, K.P. Identification and characterization of ovarian cancer-initiating cells from primary human tumors. Cancer Res. 2008, 68, 4311–4320. [Google Scholar] [CrossRef]
- Cao, L.; Shao, M.; Schilder, J.; Guise, T.; Mohammad, K.; Matei, D. Tissue transglutaminase links TGF-β, epithelial to mesenchymal transition and a stem cell phenotype in ovarian cancer. Oncogene 2012, 31, 2521–2534. [Google Scholar] [CrossRef]
- Wintzell, M.; Löfstedt, L.; Johansson, J.; Pedersen, A.B.; Fuxe, J.; Shoshan, M. Repeated cisplatin treatment can lead to a multiresistant tumor cell population with stem cell features and sensitivity to 3-bromopyruvate. Cancer Biol. Ther. 2012, 13, 1454–1462. [Google Scholar] [CrossRef]
- Chen, D.; Zhang, Y.; Wang, J.; Chen, J.; Yang, C.; Cai, K.; Wang, X.; Shi, F.; Dou, J. MicroRNA-200c overexpression inhibits tumorigenicity and metastasis of CD117+CD44+ ovarian cancer stem cells by regulating epithelial-mesenchymal transition. J. Ovarian Res. 2013, 6, 50. [Google Scholar] [CrossRef]
- Meng, E.; Long, B.; Sullivan, P.; McClellan, S.; Finan, M.A.; Reed, E.; Shevde, L.; Rocconi, R.P. CD44+/CD24− ovarian cancer cells demonstrate cancer stem cell properties and correlate to survival. Clin. Exp. Metastasis 2012, 29, 939–948. [Google Scholar] [CrossRef]
- Clark, D.W.; Palle, K. Aldehyde dehydrogenases in cancer stem cells: Potential as therapeutic targets. Ann. Transl. Med. 2016, 4, 518. [Google Scholar] [CrossRef]
- House, C.D.; Jordan, E.; Hernandez, L.; Ozaki, M.; James, J.M.; Kim, M.; Kruhlak, M.J.; Batchelor, E.; Elloumi, F.; Cam, M.C. NFκB Promotes Ovarian Tumorigenesis via Classical Pathways That Support Proliferative Cancer Cells and Alternative Pathways That Support ALDH+ Cancer Stem–like CellsAlternative NFκB Regulates ALDH+ Ovarian Cancer Cells. Cancer Res. 2017, 77, 6927–6940. [Google Scholar] [CrossRef]
- Grimley, E.; Cole, A.J.; Luong, T.T.; McGonigal, S.C.; Sinno, S.; Yang, D.; Bernstein, K.A.; Buckanovich, R.J. Aldehyde dehydrogenase inhibitors promote DNA damage in ovarian cancer and synergize with ATM/ATR inhibitors. Theranostics 2021, 11, 3540–3551. [Google Scholar] [CrossRef]
- Nwani, N.G.; Condello, S.; Wang, Y.; Swetzig, W.M.; Barber, E.; Hurley, T.; Matei, D. A novel ALDH1A1 inhibitor targets cells with stem cell characteristics in ovarian cancer. Cancers 2019, 11, 502. [Google Scholar] [CrossRef]
- Xiong, S.; Feng, Y.; Cheng, L. Cellular reprogramming as a therapeutic target in cancer. Trends Cell Biol. 2019, 29, 623–634. [Google Scholar] [CrossRef]
- Landen, C.N., Jr.; Goodman, B.; Katre, A.A.; Steg, A.D.; Nick, A.M.; Stone, R.L.; Miller, L.D.; Mejia, P.V.; Jennings, N.B.; Gershenson, D.M. Targeting aldehyde dehydrogenase cancer stem cells in ovarian cancer. Mol. Cancer Ther. 2010, 9, 3186–3199. [Google Scholar] [CrossRef]
- Deng, S.; Yang, X.; Lassus, H.; Liang, S.; Kaur, S.; Ye, Q.; Li, C.; Wang, L.-P.; Roby, K.F.; Orsulic, S. Distinct expression levels and patterns of stem cell marker, aldehyde dehydrogenase isoform 1 (ALDH1), in human epithelial cancers. PLoS ONE 2010, 5, e10277. [Google Scholar] [CrossRef]
- Kuroda, T.; Hirohashi, Y.; Torigoe, T.; Yasuda, K.; Takahashi, A.; Asanuma, H.; Morita, R.; Mariya, T.; Asano, T.; Mizuuchi, M. ALDH1-high ovarian cancer stem-like cells can be isolated from serous and clear cell adenocarcinoma cells, and ALDH1 high expression is associated with poor prognosis. PLoS ONE 2013, 8, e65158. [Google Scholar] [CrossRef]
- Wang, Y.-C.; Yo, Y.-T.; Lee, H.-Y.; Liao, Y.-P.; Chao, T.-K.; Su, P.-H.; Lai, H.-C. ALDH1-bright epithelial ovarian cancer cells are associated with CD44 expression, drug resistance, and poor clinical outcome. Am. J. Pathol. 2012, 180, 1159–1169. [Google Scholar] [CrossRef]
- Zhao, W.; Zang, C.; Zhang, T.; Li, J.; Liu, R.; Feng, F.; Lv, Q.; Zheng, L.; Tian, J.; Sun, C. Clinicopathological characteristics and prognostic value of the cancer stem cell marker ALDH1 in ovarian cancer: A meta-analysis. OncoTargets Ther. 2018, 11, 1821–1831. [Google Scholar] [CrossRef]
- Uddin, M.H.; Kim, B.; Cho, U.; Azmi, A.S.; Song, Y.S. Association of ALDH1A1-NEK-2 axis in cisplatin resistance in ovarian cancer cells. Heliyon 2020, 6, e05442. [Google Scholar] [CrossRef]
- Roy, M.; Connor, J.; Al-Niaimi, A.; Rose, S.L.; Mahajan, A. Aldehyde dehydrogenase 1A1 (ALDH1A1) expression by immunohistochemistry is associated with chemo-refractoriness in patients with high-grade ovarian serous carcinoma. Hum. Pathol. 2018, 73, 1–6. [Google Scholar] [CrossRef]
- Chang, B.; Liu, G.; Xue, F.; Rosen, D.G.; Xiao, L.; Wang, X.; Liu, J. ALDH1 expression correlates with favorable prognosis in ovarian cancers. Mod. Pathol. 2009, 22, 817–823. [Google Scholar] [CrossRef]
- Huang, R.; Li, X.; Holm, R.; Trope, C.G.; Nesland, J.M.; Suo, Z. The expression of aldehyde dehydrogenase 1 (ALDH1) in ovarian carcinomas and its clinicopathological associations: A retrospective study. BMC Cancer 2015, 15, 502. [Google Scholar] [CrossRef]
- Hough, M.R.; Rosten, P.M.; Sexton, T.L.; Kay, R.; Humphries, R.K. Mapping of CD24 and homologous sequences to multiple chromosomal loci. Genomics 1994, 22, 154–161. [Google Scholar] [CrossRef] [PubMed]
- Rougon, G.; Alterman, L.A.; Dennis, K.; Guo, X.J.; Kinnon, C. The murine heat-stable antigen: A differentiation antigen expressed in both the hematolymphoid and neural cell lineages. Eur. J. Immunol. 1991, 21, 1397–1402. [Google Scholar] [CrossRef] [PubMed]
- Bai, X.F.; Li, O.; Zhou, Q.; Zhang, H.; Joshi, P.S.; Zheng, X.; Liu, Y.; Wang, Y.; Zheng, P.; Liu, Y. CD24 controls expansion and persistence of autoreactive T cells in the central nervous system during experimental autoimmune encephalomyelitis. J. Exp. Med. 2004, 200, 447–458. [Google Scholar] [CrossRef] [PubMed]
- Tarhriz, V.; Bandehpour, M.; Dastmalchi, S.; Ouladsahebmadarek, E.; Zarredar, H.; Eyvazi, S. Overview of CD24 as a new molecular marker in ovarian cancer. J. Cell. Physiol. 2019, 234, 2134–2142. [Google Scholar] [CrossRef] [PubMed]
- Nagare, R.P.; Sneha, S.; Sidhanth, C.; Roopa, S.; Murhekar, K.; Shirley, S.; Swaminathan, R.; Sridevi, V.; Ganesan, T.S. Expression of cancer stem cell markers CD24, EPHA1 and CD9 and their correlation with clinical outcome in epithelial ovarian tumours. Cancer Biomark. 2020, 28, 397–408. [Google Scholar] [CrossRef] [PubMed]
- Nakamura, K.; Terai, Y.; Tanabe, A.; Ono, Y.J.; Hayashi, M.; Maeda, K.; Fujiwara, S.; Ashihara, K.; Nakamura, M.; Tanaka, Y.; et al. CD24 expression is a marker for predicting clinical outcome and regulates the epithelial-mesenchymal transition in ovarian cancer via both the Akt and ERK pathways. Oncol. Rep. 2017, 37, 3189–3200. [Google Scholar] [CrossRef] [PubMed]
- Kristiansen, G.; Denkert, C.; Schlüns, K.; Dahl, E.; Pilarsky, C.; Hauptmann, S. CD24 is expressed in ovarian cancer and is a new independent prognostic marker of patient survival. Am. J. Pathol. 2002, 161, 1215–1221. [Google Scholar] [CrossRef] [PubMed]
- Soltész, B.; Lukács, J.; Szilágyi, E.; Márton, É.; Szilágyi Bónizs, M.; Penyige, A.; Póka, R.; Nagy, B. Expression of CD24 in plasma, exosome and ovarian tissue samples of serous ovarian cancer patients. J. Biotechnol. 2019, 298, 16–20. [Google Scholar] [CrossRef] [PubMed]
- Gao, M.Q.; Choi, Y.P.; Kang, S.; Youn, J.H.; Cho, N.H. CD24+ cells from hierarchically organized ovarian cancer are enriched in cancer stem cells. Oncogene 2010, 29, 2672–2680. [Google Scholar] [CrossRef]
- Burgos-Ojeda, D.; Wu, R.; McLean, K.; Chen, Y.C.; Talpaz, M.; Yoon, E.; Cho, K.R.; Buckanovich, R.J. CD24+ Ovarian Cancer Cells Are Enriched for Cancer-Initiating Cells and Dependent on JAK2 Signaling for Growth and Metastasis. Mol. Cancer Ther. 2015, 14, 1717–1727. [Google Scholar] [CrossRef]
- Foster, B.M.; Zaidi, D.; Young, T.R.; Mobley, M.E.; Kerr, B.A. CD117/c-kit in Cancer Stem Cell-Mediated Progression and Therapeutic Resistance. Biomedicines 2018, 6, 31. [Google Scholar] [CrossRef]
- Longley, B.J.; Reguera, M.J.; Ma, Y. Classes of c-KIT activating mutations: Proposed mechanisms of action and implications for disease classification and therapy. Leuk. Res. 2001, 25, 571–576. [Google Scholar] [CrossRef] [PubMed]
- Luo, L.; Zeng, J.; Liang, B.; Zhao, Z.; Sun, L.; Cao, D.; Yang, J.; Shen, K. Ovarian cancer cells with the CD117 phenotype are highly tumorigenic and are related to chemotherapy outcome. Exp. Mol. Pathol. 2011, 91, 596–602. [Google Scholar] [CrossRef] [PubMed]
- Yang, B.; Yan, X.; Liu, L.; Jiang, C.; Hou, S. Overexpression of the cancer stem cell marker CD117 predicts poor prognosis in epithelial ovarian cancer patients: Evidence from meta-analysis. Onco Targets Ther. 2017, 10, 2951–2961. [Google Scholar] [CrossRef] [PubMed]
- Liu, W.; Zhang, J.; Gan, X.; Shen, F.; Yang, X.; Du, N.; Xia, D.; Liu, L.; Qiao, L.; Pan, J.; et al. LGR5 promotes epithelial ovarian cancer proliferation, metastasis, and epithelial-mesenchymal transition through the Notch1 signaling pathway. Cancer Med. 2018, 7, 3132–3142. [Google Scholar] [CrossRef] [PubMed]
- McClanahan, T.; Koseoglu, S.; Smith, K.; Grein, J.; Gustafson, E.; Black, S.; Kirschmeier, P.; Samatar, A.A. Identification of overexpression of orphan G protein-coupled receptor GPR49 in human colon and ovarian primary tumors. Cancer Biol. Ther. 2006, 5, 419–426. [Google Scholar] [CrossRef]
- Wilczyński, J.R.; Wilczyński, M.; Paradowska, E. Cancer Stem Cells in Ovarian Cancer—A Source of Tumor Success and a Challenging Target for Novel Therapies. Int. J. Mol. Sci. 2022, 23, 2496. [Google Scholar] [CrossRef] [PubMed]
- Motohara, T.; Masuko, S.; Ishimoto, T.; Yae, T.; Onishi, N.; Muraguchi, T.; Hirao, A.; Matsuzaki, Y.; Tashiro, H.; Katabuchi, H. Transient depletion of p53 followed by transduction of c-Myc and K-Ras converts ovarian stem-like cells into tumor-initiating cells. Carcinogenesis 2011, 32, 1597–1606. [Google Scholar] [CrossRef]
- Robinson, M.; Gilbert, S.F.; Waters, J.A.; Lujano-Olazaba, O.; Lara, J.; Alexander, L.J.; Green, S.E.; Burkeen, G.A.; Patrus, O.; Sarwar, Z.; et al. Characterization of SOX2, OCT4 and NANOG in Ovarian Cancer Tumor-Initiating Cells. Cancers 2021, 13, 262. [Google Scholar] [CrossRef]
- Parte, S.C.; Batra, S.K.; Kakar, S.S. Characterization of stem cell and cancer stem cell populations in ovary and ovarian tumors. J. Ovarian Res. 2018, 11, 69. [Google Scholar] [CrossRef]
- Saunders, A.; Li, D.; Faiola, F.; Huang, X.; Fidalgo, M.; Guallar, D.; Ding, J.; Yang, F.; Xu, Y.; Zhou, H.; et al. Context-Dependent Functions of NANOG Phosphorylation in Pluripotency and Reprogramming. Stem Cell Rep. 2017, 8, 1115–1123. [Google Scholar] [CrossRef]
- Yun, H.; Han, G.H.; Kim, J.; Chung, J.Y.; Kim, J.H.; Cho, H. NANOG regulates epithelial-mesenchymal transition via AMPK/mTOR signalling pathway in ovarian cancer SKOV-3 and A2780 cells. J. Cell Mol. Med. 2022, 26, 5277–5291. [Google Scholar] [CrossRef] [PubMed]
- Kim, D.K.; Lee, J.-S.; Lee, E.Y.; Jang, H.; Han, S.; Kim, H.Y.; Hwang, I.-Y.; Choi, J.-W.; Shin, H.M.; You, H.J.; et al. O-GlcNAcylation of Sox2 at threonine 258 regulates the self-renewal and early cell fate of embryonic stem cells. Exp. Mol. Med. 2021, 53, 1759–1768. [Google Scholar] [CrossRef] [PubMed]
- Gong, R.; Li, Z.-Q.; Fu, K.; Ma, C.; Wang, W.; Chen, J.-C. Long noncoding RNA PVT1 promotes stemness and temozolomide resistance through miR-365/ELF4/SOX2 axis in glioma. Exp. Neurobiol. 2021, 30, 244–255. [Google Scholar] [CrossRef]
- Rao, R.S.; Raju, K.L.; Augustine, D.; Patil, S. Prognostic Significance of ALDH1, Bmi1, and OCT4 Expression in Oral Epithelial Dysplasia and Oral Squamous Cell Carcinoma. Cancer Control 2020, 27, 1073274820904959. [Google Scholar] [CrossRef]
- Barger, C.J.; Branick, C.; Chee, L.; Karpf, A.R. Pan-Cancer Analyses Reveal Genomic Features of FOXM1 Overexpression in Cancer. Cancers 2019, 11, 251. [Google Scholar] [CrossRef] [PubMed]
- Zhou, J.; Wang, Y.; Wang, Y.; Yin, X.; He, Y.; Chen, L.; Wang, W.; Liu, T.; Di, W. FOXM1 modulates cisplatin sensitivity by regulating EXO1 in ovarian cancer. PLoS ONE 2014, 9, e96989. [Google Scholar] [CrossRef]
- Kim, D.K.; Ham, M.H.; Lee, S.Y.; Shin, M.J.; Kim, Y.E.; Song, P.; Suh, D.-S.; Kim, J.H. CD166 promotes the cancer stem-like properties of primary epithelial ovarian cancer cells. BMB Rep. 2020, 53, 622–627. [Google Scholar] [CrossRef]
- Umezu-Goto, M.; Kishi, Y.; Taira, A.; Hama, K.; Dohmae, N.; Takio, K.; Yamori, T.; Mills, G.B.; Inoue, K.; Aoki, J. Autotaxin has lysophospholipase D activity leading to tumor cell growth and motility by lysophosphatidic acid production. J. Cell Biol. 2002, 158, 227–233. [Google Scholar] [CrossRef]
- Brindley, D.N.; Lin, F.-T.; Tigyi, G.J. Role of the autotaxin–lysophosphatidate axis in cancer resistance to chemotherapy and radiotherapy. Biochim. Biophys. Acta (BBA)-Mol. Cell Biol. Lipids 2013, 1831, 74–85. [Google Scholar] [CrossRef]
- Seo, E.J.; Kwon, Y.W.; Jang, I.H.; Kim, D.K.; Lee, S.I.; Choi, E.J.; Kim, K.-H.; Suh, D.-S.; Lee, J.H.; Choi, K.U. Autotaxin regulates maintenance of ovarian cancer stem cells through lysophosphatidic acid-mediated autocrine mechanism. Stem Cells 2016, 34, 551–564. [Google Scholar] [CrossRef]
- Enriquez, V.A.; Cleys, E.R.; Da Silveira, J.C.; Spillman, M.A.; Winger, Q.A.; Bouma, G.J. High LIN28A Expressing Ovarian Cancer Cells Secrete Exosomes That Induce Invasion and Migration in HEK293 Cells. BioMed. Res. Int. 2015, 2015, 701390. [Google Scholar] [CrossRef] [PubMed]
- Rupp, A.-K.; Rupp, C.; Keller, S.; Brase, J.C.; Ehehalt, R.; Fogel, M.; Moldenhauer, G.; Marmé, F.; Sültmann, H.; Altevogt, P. Loss of EpCAM expression in breast cancer derived serum exosomes: Role of proteolytic cleavage. Gynecol. Oncol. 2011, 122, 437–446. [Google Scholar] [CrossRef] [PubMed]
- Hsu, Y.-T.; Osmulski, P.; Wang, Y.; Huang, Y.-W.; Liu, L.; Ruan, J.; Jin, V.X.; Kirma, N.B.; Gaczynska, M.E.; Huang, T.H.-M. EpCAM-regulated transcription exerts influences on nanomechanical properties of endometrial cancer cells that promote epithelial-to-mesenchymal transition. Cancer Res. 2016, 76, 6171–6182. [Google Scholar] [CrossRef] [PubMed]
- Breed, C.; Hicks, D.A.; Webb, P.G.; Galimanis, C.E.; Bitler, B.G.; Behbakht, K.; Baumgartner, H.K. Ovarian Tumor Cell Expression of Claudin-4 Reduces Apoptotic Response to Paclitaxel. Mol. Cancer Res. 2019, 17, 741–750. [Google Scholar] [CrossRef] [PubMed]
- Huang, D.; Rao, D.; Xi, X.; Zhang, Z.; Zhong, T. Application of extracellular vesicles proteins in cancer diagnosis. Front. Cell Dev. Biol. 2022, 10, 1007360. [Google Scholar] [CrossRef] [PubMed]
- López de Andrés, J.; Griñán-Lisón, C.; Jiménez, G.; Marchal, J.A. Cancer stem cell secretome in the tumor microenvironment: A key point for an effective personalized cancer treatment. J. Hematol. Oncol. 2020, 13, 136. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Q.; Cai, D.J.; Li, B. Ovarian cancer stem-like cells elicit the polarization of M2 macrophages. Mol. Med. Rep. 2015, 11, 4685–4693. [Google Scholar] [CrossRef] [PubMed]
- Bonello, M.; Sims, A.H.; Langdon, S.P. Human epidermal growth factor receptor targeted inhibitors for the treatment of ovarian cancer. Cancer Biol. Med. 2018, 15, 375–388. [Google Scholar]
- Ren, L.; Yu, Y.; Wang, L.; Zhu, Z.; Lu, R.; Yao, Z. Hypoxia-induced CCL28 promotes recruitment of regulatory T cells and tumor growth in liver cancer. Oncotarget 2016, 7, 75763–75773. [Google Scholar] [CrossRef]
- Kim, W.T.; Ryu, C.J. Cancer stem cell surface markers on normal stem cells. BMB Rep. 2017, 50, 285–298. [Google Scholar] [CrossRef]
- Badrinath, N.; Yoo, S.Y. Recent Advances in Cancer Stem Cell-Targeted Immunotherapy. Cancers 2019, 11, 310. [Google Scholar] [CrossRef] [PubMed]
- Wang, L.; Xu, T.; Cui, M. Are ovarian cancer stem cells the target for innovative immunotherapy? Onco Targets Ther. 2018, 11, 2615–2626. [Google Scholar] [CrossRef] [PubMed]
Identifier/Official Title | Phase | Population/Intervention/Treatment | Primary Endpoints | Results |
---|---|---|---|---|
Completed Trials | ||||
NCT02178670 Study of Cancer Stem Cell Vaccine That as a Specific Antigen in Metastatic Adenocarcinoma of the Ovarian | 1, 2 | 40 participants with stage III epithelial ovarian cancer in remission after surgery (hysterectomy and ovariectomy) and the first primary chemotherapy Treatment: biological: CSC-DC | Primary outcome: Safety of immunisation with the cancer stem cell vaccine (number of participants with an adverse event) Secondary outcome:
| No results have been published |
NCT01579812 A Phase II Evaluation of Metformin, Targeting Cancer Stem Cells for the Prevention of Relapse in Patients with Stage IIC/III/IV Ovarian, Fallopian Tube, and Primary Peritoneal Cancer | 2 | 90 participants with advanced ovarian, fallopian tube, or primary peritoneal cancer | Primary outcome: Recurrence-free survival (time frame: 18 months) Primary endpoints:
Overall survival (time frame: up to 3 years) |
|
Ongoing Trials | ||||
Identifier/official title | Phase | Population/intervention/treatment | Primary endpoints | |
NCT03949283 Standard Chemotherapy Versus Cancer Stem Cell Assay Directed Chemotherapy in Recurrent Platinum Resistant Ovarian Cancer | 3 | 150 participants with recurrent platinum-resistant ovarian cancer Diagnostic test: ChemoID assay Drug: standard chemotherapy | Primary outcome: ORR in patients with recurrent epithelial ovarian cancer who had ChemoID-guided treatment versus physician choice control treatment Secondary outcomes:
| |
NCT03632798 Avastin Plus Chemotherapy vs. Avastin Plus Chemotherapy Chosen by Cancer Stem Cell Chemosensitivity Testing in the Management of Patients with Recurrent Platinum-Resistant or -Sensitive Epithelial Ovarian, Fallopian Tube, or Primary Peritoneal Cancer | 3 | 300 participants experiencing first, second, or third recurrence of any stage of epithelial ovarian cancer Diagnostic test: ChemoID assay Drug: chemotherapy | Primary outcome: PFS in patients with recurrent epithelial cancer who received standard treatment with bevacizumab plus chemotherapy chosen by the physician versus bevacizumab plus ChemoID drug response assay-directed chemotherapy Secondary outcome:
| |
NCT05576519 Immunohistochemical Expression of Epithelial Cell Adhesion Molecule (EpCAM) in Epithelial Ovarian Carcinoma | * | 50 paraffin blocks of epithelial ovarian cancer collected from patients who underwent surgery | Primary outcome: Evaluation of EPCAM expression in ovarian carcinoma | |
NCT02713386 A Phase I/II Study of Ruxolitinib with Front-Line Neoadjuvant and Post-Surgical Therapy in Patients with Advanced Epithelial Ovarian, Fallopian Tube or Primary Peritoneal Cancer | 1,2 | 147 participants with advanced epithelial ovarian, fallopian tube, or primary peritoneal cancer Drug: carboplatin Drug: paclitaxel Drug: ruxolitinib phosphate Procedure: therapeutic conventional surgery | Primary outcome:
|
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Frąszczak, K.; Barczyński, B. The Role of Cancer Stem Cell Markers in Ovarian Cancer. Cancers 2024, 16, 40. https://doi.org/10.3390/cancers16010040
Frąszczak K, Barczyński B. The Role of Cancer Stem Cell Markers in Ovarian Cancer. Cancers. 2024; 16(1):40. https://doi.org/10.3390/cancers16010040
Chicago/Turabian StyleFrąszczak, Karolina, and Bartłomiej Barczyński. 2024. "The Role of Cancer Stem Cell Markers in Ovarian Cancer" Cancers 16, no. 1: 40. https://doi.org/10.3390/cancers16010040
APA StyleFrąszczak, K., & Barczyński, B. (2024). The Role of Cancer Stem Cell Markers in Ovarian Cancer. Cancers, 16(1), 40. https://doi.org/10.3390/cancers16010040