Antioxidants and Therapeutic Targets in Ovarian Clear Cell Carcinoma
Abstract
:1. Introduction
2. Linking Oxidative Stress and Carcinogenesis from Endometriosis to OCCCs
3. Attempts to Prevent OCCCs Developing from Endometriosis
4. Molecular Characteristics in OCCCs Related to Anti-Oxidative Pathway
5. Oxidative Stress and Cancer Stemness of OCCC
6. Therapeutic Targets for OCCCs in the Present and Future
Category | Target Molecules | Clinical Research | Result | Reference |
---|---|---|---|---|
RTKs and related molecules | MET | already completed | minimal activity | [84] |
PI3K/AKT/mTOR | already completed | clinically ineffective | [85] | |
VEGFR/PDGFR | already completed | minimal activity | [86] | |
VEGFR/PDGFR/FGFR | Currently conducting | ― | NCT02866370 | |
ARID1A chromatin remodeling factor | EZH/glutathione | ― | ― | [87,88,89] |
Immune checkpoint proteins | PD-L1 | Currently conducting | ― | NCT03405454 |
PD-1/CTLA-4 | Currently conducting | ― | NCT03355976 | |
TIM-1 | Currently conducting | ― | NCT02837991 |
7. Conclusions
Supplementary Materials
Author Contributions
Funding
Conflicts of Interest
References
- Available online: http://gco.iarc.fr/today/home (accessed on 15 December 2020).
- Available online: https://www.cancer.org/research/cancer-facts-statistics/all-cancer-facts-figures/cancer-facts-figures-2018.html (accessed on 15 December 2020).
- Available online: https://ganjoho.jp/reg_stat/statistics/stat/short_pred.html (accessed on 15 December 2020).
- Yamagami, W.; Nagase, S.; Takahashi, F.; Ino, K.; Hachisuga, T.; Aoki, D.; Katabuchi, H. Clinical statistics of gynecologic cancers in Japan. J. Gynecol. Oncol. 2017, 28. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Itamochi, H.; Kigawa, J.; Terakawa, N. Mechanisms of chemoresistance and poor prognosis in ovarian clear cell carcinoma. Cancer Sci. 2008, 99, 653–658. [Google Scholar] [CrossRef] [PubMed]
- Shih, I.-M.; Kurman, R.J. Ovarian Tumorigenesis. Am. J. Pathol. 2004, 164, 1511–1518. [Google Scholar] [CrossRef]
- Köbel, M.; Reuss, A.; Du Bois, A.; Kommoss, S.; Kommoss, F.; Gao, D.; Kalloger, S.E.; Huntsman, D.G.; Gilks, C.B. The biological and clinical value of p53 expression in pelvic high-grade serous carcinomas. J. Pathol. 2010, 222, 191–198. [Google Scholar] [CrossRef] [PubMed]
- Kaldawy, A.; Segev, Y.; Lavie, O.; Auslender, R.; Sopik, V.; Narod, S.A. Low-grade serous ovarian cancer: A review. Gynecol. Oncol. 2016, 143, 433–438. [Google Scholar] [CrossRef] [PubMed]
- Seidman, J.D.; Kurman, R.J.; Ronnett, B.M. Primary and Metastatic Mucinous Adenocarcinomas in the Ovaries. Am. J. Surg. Pathol. 2003, 27, 985–993. [Google Scholar] [CrossRef] [PubMed]
- Köbel, M.; Kalloger, S.E.; Boyd, N.; McKinney, S.; Mehl, E.; Palmer, C.; Leung, S.; Bowen, N.J.; Ionescu, D.N.; Rajput, A.; et al. Ovarian Carcinoma Subtypes Are Different Diseases: Implications for Biomarker Studies. PLoS Med. 2008, 5, e232. [Google Scholar] [CrossRef]
- Bouchard-Fortier, G.; Panzarella, T.; Rosen, B.; Chapman, W.; Gien, L.T. Endometrioid Carcinoma of the Ovary: Outcomes Compared to Serous Carcinoma After 10 Years of Follow-Up. J. Obstet. Gynaecol. Can. 2017, 39, 34–41. [Google Scholar] [CrossRef]
- Kuo, K.-T.; Mao, T.-L.; Jones, S.; Veras, E.; Ayhan, A.; Wang, T.-L.; Glas, R.; Slamon, D.; Velculescu, V.E.; Kuman, R.J.; et al. Frequent Activating Mutations of PIK3CA in Ovarian Clear Cell Carcinoma. Am. J. Pathol. 2009, 174, 1597–1601. [Google Scholar] [CrossRef] [Green Version]
- Jones, S.; Wang, T.-L.; Shih, I.-M.; Mao, T.-L.; Nakayama, K.; Roden, R.; Glas, R.; Slamon, D.; Diaz, L.A.; Vogelstein, B.; et al. Frequent Mutations of Chromatin Remodeling Gene ARID1A in Ovarian Clear Cell Carcinoma. Science 2010, 330, 228–231. [Google Scholar] [CrossRef] [Green Version]
- Tsuchiya, A.; Sakamoto, M.; Yasuda, J.; Chuma, M.; Ohta, T.; Ohki, M.; Yasugi, T.; Taketani, Y.; Hirohashi, S. Expression Profiling in Ovarian Clear Cell Carcinoma. Am. J. Pathol. 2003, 163, 2503–2512. [Google Scholar] [CrossRef]
- Zondervan, K.; Becker, C.M.; Missmer, S.A. Endometriosis. N. Engl. J. Med. 2020, 382, 1244–1256. [Google Scholar] [CrossRef] [PubMed]
- Pearce, C.L.; Templeman, C.; Rossing, M.A.; Lee, A.; Near, A.M.; Webb, P.M.; Nagle, C.M.; Doherty, J.A.; Cushing-Haugen, K.L.; Wicklund, K.G.; et al. Association between endometriosis and risk of histological subtypes of ovarian cancer: Apooled analysis of case–control studies. Lancet Oncol. 2012, 13, 385–394. [Google Scholar] [CrossRef] [Green Version]
- Nezhat, F.; Datta, M.S.; Hanson, V.; Pejovic, T.; Nezhat, C.; Nezhat, C. The relationship of endometriosis and ovarian malignancy: A review. Fertil. Steril. 2008, 90, 1559–1570. [Google Scholar] [CrossRef] [PubMed]
- Landskron, G.; De La Fuente, M.; Thuwajit, P.; Thuwajit, C.; Hermoso, M.A. Chronic Inflammation and Cytokines in the Tumor Microenvironment. J. Immunol. Res. 2014, 2014, 149185. [Google Scholar] [CrossRef] [Green Version]
- Balkwill, F.R.; Mantovani, A. Inflammation and cancer: Back to Virchow? Lancet 2001, 357, 539–545. [Google Scholar] [CrossRef]
- Worley, J.M.J.; Welch, W.R.; Berkowitz, R.S.; Ng, S.-W. Endometriosis-Associated Ovarian Cancer: A Review of Pathogenesis. Int. J. Mol. Sci. 2013, 14, 5367–5379. [Google Scholar] [CrossRef] [Green Version]
- Bulun, S.E. Endometriosis. N. Engl. J. Med. 2009, 360, 268–279. [Google Scholar] [CrossRef]
- Yamaguchi, K.; Mandai, M.; Oura, T.; Matsumura, N.; Hamanishi, J.; Baba, T.; Matsui, S.; Murphy, S.K.; Konishi, I. Identification of an ovarian clear cell carcinoma gene signature that reflects inherent disease biology and the carcinogenic processes. Oncogene 2010, 29, 1741–1752. [Google Scholar] [CrossRef] [Green Version]
- Toyokuni, S. Role of iron in carcinogenesis: Cancer as a ferrotoxic disease. Cancer Sci. 2009, 100, 9–16. [Google Scholar] [CrossRef]
- Yamaguchi, K.; Mandai, M.; Toyokuni, S.; Hamanishi, J.; Higuchi, T.; Takakura, K.; Fujii, S. Contents of Endometriotic Cysts, Especially the High Concentration of Free Iron, Are a Possible Cause of Carcinogenesis in the Cysts through the Iron-Induced Persistent Oxidative Stress. Clin. Cancer Res. 2008, 14, 32–40. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, J.L.; Okada, S.; Hamazaki, S.; Ebina, Y.; Midorikawa, O. Subacute nephrotoxicity and in-duction of renal cell carcinoma in mice treated with ferric nitrilotriacetate. Cancer Res. 1987, 47, 1867–1869. [Google Scholar] [PubMed]
- Liu, M.; Okada, S. Induction of free radicals and tumors in the kidneys of Wistar rats by ferric ethylenediamine-N,N′-diacetate. Carcinogenesis 1994, 15, 2817–2821. [Google Scholar] [CrossRef] [PubMed]
- Munksgaard, P.S.; Blaakaer, J. The association between endometriosis and ovarian cancer: A review of histological, genetic and molecular alterations. Gynecol. Oncol. 2012, 124, 164–169. [Google Scholar] [CrossRef] [PubMed]
- Melin, A.-S.; Lundholm, C.; Malki, N.; Swahn, M.-L.; Sparén, P.; Bergqvist, A. Hormonal and surgical treatments for endometriosis and risk of epithelial ovarian cancer. Acta Obstet. Gynecol. Scand. 2013, 92, 546–554. [Google Scholar] [CrossRef] [PubMed]
- Yoshino, O.; Minamisaka, T.; Ono, Y.; Tsuda, S.; Samejima, A.; Shima, T.; Nakashima, A.; Koga, K.; Osuga, Y.; Saito, S. Three cases of clear-cell adenocarcinoma arising from endometrioma during hormonal treatments. J. Obstet. Gynaecol. Res. 2018, 44, 1850–1858. [Google Scholar] [CrossRef]
- Modugno, F.; Ness, R.B.; Allen, G.O.; Schildkraut, J.M.; Davis, F.G.; Goodman, M.T. Oral contraceptive use, reproductive history, and risk of epithelial ovarian cancer in women with and without endometriosis. Am. J. Obstet. Gynecol. 2004, 191, 733–740. [Google Scholar] [CrossRef]
- Collaborative Group on Epidemiological Studies of Ovarian Cancer Ovarian cancer and oral contraceptives: Collaborative reanalysis of data from 45 epidemiological studies including 23,257 women with ovarian cancer and 87,303 controls. Lancet 2008, 371, 303–314. [CrossRef] [Green Version]
- Nishida, Y.; Yamashita, E.; Miki, W. Quenching activities of common hydrophilic and lip-ophilic antioxidants against singlet oxygen using chemiluminescence detection System. Carotenoid Sci. 2007, 11, 16–20. [Google Scholar]
- Martin, H.D.; Ruck, C.; Schmidt, M.; Sell, S.; Beutner, S.; Mayer, B.; Walsh, R. Chemistry of carotenoid oxidation and free radical reactions. Pure Appl. Chem. 1999, 71, 2253–2262. [Google Scholar] [CrossRef]
- Kuroki, T.; Ikeda, S.; Okada, T.; Maoka, T.; Kitamura, A.; Sugimoto, M.; Kume, S. Astaxanthin ameliorates heat stress-induced impairment of blastocyst development In Vitro: Astaxanthin colocalization with and action on mitochondria. J. Assist. Reprod. Genet. 2013, 30, 623–631. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Park, J.S.; Mathison, B.D.; Hayek, M.G.; Zhang, J.; Reinhart, G.A.; Chew, B.P. Astaxanthin modulates age-associated mitochondrial dysfunction in healthy dogs1. J. Anim. Sci. 2013, 91, 268–275. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ames, B.N. Dietary carcinogens and anticarcinogens. Oxygen radicals and degenerative diseases. Science 1983, 221, 1256–1264. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dušinská, M.; Kažimírová, A.; Barančoková, M.; Beňo, M.; Smolkova, B.; Horská, A.; Rašlová, K.; Wsólová, L.; Collins, A. Nutritional supplementation with antioxidants decreases chromosomal damage in humans. Mutagenesis 2003, 18, 371–376. [Google Scholar] [CrossRef] [Green Version]
- Federico, A.; Morgillo, F.; Tuccillo, C.; Ciardiello, F.; Loguercio, C. Chronic inflammation and oxidative stress in human carcinogenesis. Int. J. Cancer 2007, 121, 2381–2386. [Google Scholar] [CrossRef]
- Choi, H.D.; Youn, Y.K.; Shin, W.G. Positive Effects of Astaxanthin on Lipid Profiles and Oxidative Stress in Overweight Subjects. Plant Foods Hum. Nutr. 2011, 66, 363–369. [Google Scholar] [CrossRef]
- Wolf, A.M.; Asoh, S.; Hiranuma, H.; Ohsawa, I.; Iio, K.; Satou, A.; Ishikura, M.; Ohta, S. Astaxanthin protects mitochondrial redox state and functional integrity against oxidative stress. J. Nutr. Biochem. 2010, 21, 381–389. [Google Scholar] [CrossRef]
- Aoi, W.; Naito, Y.; Takanami, Y.; Ishii, T.; Kawai, Y.; Akagiri, S.; Kato, Y.; Osawa, T.; Yoshikawa, T. Astaxanthin improves muscle lipid metabolism in exercise via inhibitory effect of oxidative CPT I modification. Biochem. Biophys. Res. Commun. 2008, 366, 892–897. [Google Scholar] [CrossRef]
- Park, J.S.; Chyun, J.H.; Kim, Y.K.; Line, L.L.; Chew, B.P. Astaxanthin decreased oxidative stress and inflammation and enhanced immune response in humans. Nutr. Metab. 2010, 7, 18. [Google Scholar] [CrossRef] [Green Version]
- Kavitha, K.; Kowshik, J.; Kishore, T.K.K.; Baba, A.B.; Nagini, S. Astaxanthin inhibits NF-κB and Wnt/β-catenin signaling pathways via inactivation of Erk/MAPK and PI3K/Akt to induce intrinsic apoptosis in a hamster model of oral cancer. Biochim. Biophys. Acta (BBA) Gen. Subj. 2013, 1830, 4433–4444. [Google Scholar] [CrossRef]
- Kowshik, J.; Baba, A.B.; Giri, H.; Reddy, G.D.; Dixit, M.; Nagini, S. Astaxanthin Inhibits JAK/STAT-3 Signaling to Abrogate Cell Proliferation, Invasion and Angiogenesis in a Hamster Model of Oral Cancer. PLoS ONE 2014, 9, e109114. [Google Scholar] [CrossRef] [PubMed]
- Palozza, P.; Torelli, C.; Boninsegna, A.; Simone, R.; Catalano, A.; Mele, M.C.; Picci, N. Growth-inhibitory effects of the astaxanthin-rich alga Haematococcus pluvialis in human colon cancer cells. Cancer Lett. 2009, 283, 108–117. [Google Scholar] [CrossRef] [PubMed]
- Tanaka, T.; Makita, H.; Ohnishi, M.; Mori, H.; Satoh, K.; Hara, A. Chemoprevention of rat oral carcinogenesis by naturally occurring xanthophylls, astaxanthin and canthaxanthin. Cancer Res. 1995, 55, 4059–4064. [Google Scholar] [PubMed]
- Brito, A.; Ribeiro, M.; Abrantes, A.M.; Pires, A.; Teixo, R.; Tralhão, J.; Botelho, M.F. Quercetin in Cancer Treatment, Alone or in Combination with Conventional Therapeutics? Curr. Med. Chem. 2015, 22, 3025–3039. [Google Scholar] [CrossRef] [Green Version]
- Srivastava, S.; Somasagara, R.R.; Hegde, M.; Nishana, M.; Tadi, S.K.; Srivastava, M.; Choudhary, B.; Raghavan, S.C. Quercetin, a Natural Flavonoid Interacts with DNA, Arrests Cell Cycle and Causes Tumor Regression by Activating Mitochondrial Pathway of Apoptosis. Sci. Rep. 2016, 6, 24049. [Google Scholar] [CrossRef] [Green Version]
- Satyan, K.; Swamy, N.; Dizon, D.S.; Singh, R.; Granai, C.O.; Brard, L. Phenethyl isothiocyanate (PEITC) inhibits growth of ovarian cancer cells by inducing apoptosis: Role of caspase and MAPK activation. Gynecol. Oncol. 2006, 103, 261–270. [Google Scholar] [CrossRef]
- Wattenberg, L.W. Inhibitory effects of benzyl isothiocyanate administered shortly before diethylnitrosamine or benzo[a]pyrene on pulmonary and forestomach neoplasia in A/J mice. Carcinogenesis 1987, 8, 1971–1973. [Google Scholar] [CrossRef]
- Long, Y.; Fei, H.; Xu, S.; Wen, J.; Ye, L.; Su, Z. Association about dietary vitamin C intake on the risk of ovarian cancer: A meta-analysis. Biosci. Rep. 2020, 40. [Google Scholar] [CrossRef] [Green Version]
- L’Espérance, K.; Datta, G.D.; Qureshi, S.; Koushik, A. Vitamin D Exposure and Ovarian Cancer Risk and Prognosis. Int. J. Environ. Res. Public Health 2020, 17, 1168. [Google Scholar] [CrossRef] [Green Version]
- Leng, Y.; Zhou, H.; Meng, F.; Tian, T.; Xu, J.; Yan, F. Association of vitamin E on the risk of ovarian cancer: A meta-analysis. Biosci. Rep. 2019, 39. [Google Scholar] [CrossRef]
- Alpha-Tocopherol, Beta Carotene Cancer Prevention Study Group. The Effect of Vitamin E and Beta Carotene on the Incidence of Lung Cancer and Other Cancers in Male Smokers. N. Engl. J. Med. 1994, 330, 1029–1035. [Google Scholar] [CrossRef]
- Wang, Q.; He, C. Dietary vitamin A intake and the risk of ovarian cancer: A meta-analysis. Biosci. Rep. 2020, 40, 40. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hua, X.; Yu, L.; You, R.; Yang, Y.; Liao, J.; Chen, D.; Yu, L. Association among Dietary Flavonoids, Flavonoid Subclasses and Ovarian Cancer Risk: A Meta-Analysis. PLoS ONE 2016, 11, e0151134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chang, E.T.; Lee, V.S.; Canchola, A.J.; Clarke, C.A.; Purdie, D.M.; Reynolds, P.; Anton-Culver, H.; Bernstein, L.; Deapen, D.; Peel, D.; et al. Diet and Risk of Ovarian Cancer in the California Teachers Study Cohort. Am. J. Epidemiol. 2007, 165, 802–813. [Google Scholar] [CrossRef]
- Warburg, O. On the Origin of Cancer Cells. Science 1956, 123, 309–314. [Google Scholar] [CrossRef] [PubMed]
- Kato, N.; Tamura, G.; Motoyama, T. Hypomethylation of hepatocyte nuclear factor-1beta (HNF-1beta) CpG island in clear cell carcinoma of the ovary. Virchows Archiv. 2008, 452, 175–180. [Google Scholar] [CrossRef]
- Zhu, J.-N.; Jiang, L.; Jiang, J.-H.; Yang, X.; Li, X.-Y.; Zeng, J.-X.; Shi, R.-Y.; Shi, Y.; Pan, X.-R.; Han, Z.-P.; et al. Hepatocyte nuclear factor-1beta enhances the stemness of hepatocellular carcinoma cells through activation of the Notch pathway. Sci. Rep. 2017, 7, 4793. [Google Scholar] [CrossRef] [Green Version]
- Sugiyama, T.; Kamura, T.; Kigawa, J.; Terakawa, N.; Kikuchi, Y.; Kita, T.; Suzuki, M.; Sato, I.; Taguchi, K. Clinical characteristics of clear cell carcinoma of the ovary: A distinct histologic type with poor prognosis and resistance to platinum-based chemotherapy. Cancer 2000, 88, 2584–2589. [Google Scholar] [CrossRef]
- Goff, B.A.; De La Cuesta, R.S.; Muntz, H.G.; Fleischhacker, D.; Ek, M.; Rice, L.W.; Nikrui, N.; Tamimi, H.K.; Cain, J.M.; Greer, B.E.; et al. Clear Cell Carcinoma of the Ovary: A Distinct Histologic Type with Poor Prognosis and Resistance to Platinum-Based Chemotherapy in Stage III Disease. Gynecol. Oncol. 1996, 60, 412–417. [Google Scholar] [CrossRef]
- Senkel, S.; Lucas, B.; Klein-Hitpass, L.; Ryffel, G.U. Identification of target genes of the transcription factor HNF1β and HNF1α in a human embryonic kidney cell line. Biochim. Biophys. Acta (BBA) Gene Struct. Expr. 2005, 1731, 179–190. [Google Scholar] [CrossRef]
- Okamoto, T.; Mandai, M.; Matsumura, N.; Yamaguchi, K.; Kondoh, H.; Amano, Y.; Baba, T.; Hamanishi, J.; Abiko, K.; Kosaka, K.; et al. Hepatocyte nuclear factor-1β (HNF-1β) promotes glucose uptake and glycolytic activity in ovarian clear cell carcinoma. Mol. Carcinog. 2015, 54, 35–49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, P.; Khurana, A.; Rattan, R.; He, X.; Kalloger, S.; Dowdy, S.; Gilks, B.; Shridhar, V. Regulation of HSulf-1 Expression by Variant Hepatic Nuclear Factor 1 in Ovarian Cancer. Cancer Res. 2009, 69, 4843–4850. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, C.; Zhou, Y.; Hu, C.; Wang, Y.; Yan, Z.; Li, Z.; Wu, R. Mitochondria and oxidative stress in ovarian endometriosis. Free Radic. Biol. Med. 2019, 136, 22–34. [Google Scholar] [CrossRef] [PubMed]
- Hemachandra, L.P.M.P.; Shin, D.-H.; Dier, U.; Iuliano, J.N.; Engelberth, S.A.; Uusitalo, L.M.; Murphy, S.K.; Hempel, N. Mitochondrial Superoxide Dismutase Has a Protumorigenic Role in Ovarian Clear Cell Carcinoma. Cancer Res. 2015, 75, 4973–4984. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Amano, T.; Chano, T.; Isono, T.; Kimura, F.; Kushima, R.; Murakami, T. Abundance of mitochondrial superoxide dismutase is a negative predictive biomarker for endometriosis-associated ovarian cancers. World J. Surg. Oncol. 2019, 17, 1–7. [Google Scholar] [CrossRef]
- Mizuno, T.; Suzuki, N.; Makino, H.; Furui, T.; Morii, E.; Aoki, H.; Kunisada, T.; Yano, M.; Kuji, S.; Hirashima, Y.; et al. Cancer stem-like cells of ovarian clear cell carcinoma are enriched in the ALDH-high population associated with an accelerated scavenging system in reactive oxygen species. Gynecol. Oncol. 2015, 137, 299–305. [Google Scholar] [CrossRef]
- Visvader, J.E.; Lindeman, G.J. Cancer stem cells in solid tumours: Accumulating evidence and unresolved questions. Nat. Rev. Cancer 2008, 8, 755–768. [Google Scholar] [CrossRef]
- Chano, T.; Kita, H.; Avnet, S.; Lemma, S.; Baldini, N. Prominent role of RAB39A-RXRB axis in cancer development and stemness. Oncotarget 2018, 9, 9852–9866. [Google Scholar] [CrossRef] [Green Version]
- Metzler, M.A.; Sandell, L.L. Enzymatic Metabolism of Vitamin A in Developing Vertebrate Embryos. Nutrients 2016, 8, 812. [Google Scholar] [CrossRef]
- Shannon, S.R.; Moise, A.R.; Trainor, P. New insights and changing paradigms in the regulation of vitamin A metabolism in development. Wiley Interdiscip. Rev. Dev. Biol. 2017, 6, e264. [Google Scholar] [CrossRef] [Green Version]
- Obrochta, K.M.; Krois, C.R.; Campos, B.; Napoli, J.L.; Duan, X.-L.; Liu, N.-N.; Yang, Y.-T.; Li, H.-H.; Li, M.; Dou, S.-X.; et al. Insulin Regulates Retinol Dehydrogenase Expression and All-trans-retinoic Acid Biosynthesis through FoxO1. J. Biol. Chem. 2015, 290, 7259–7268. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Felix, E.L.; Loyd, B.; Cohen, M.H. Inhibition of the growth and development of a transplantable murine melanoma by vitamin A. Science 1975, 189, 886–888. [Google Scholar] [CrossRef] [PubMed]
- Dillehay, D.L.; Shealy, Y.F.; Lamon, E.W. Inhibition of Moloney murine lymphoma and sarcoma growth in vivo by dietary retinoids. Cancer Res. 1989, 49, 44–50. [Google Scholar] [PubMed]
- Aebi, S.; Kröning, R.; Cenni, B.; Sharma, A.; Fink, D.; Los, G.; Weisman, R.; Howell, S.B.; Christen, R.D. all-trans retinoic acid enhances cisplatin-induced apoptosis in human ovarian adenocarcinoma and in squamous head and neck cancer cells. Clin. Cancer Res. 1997, 3, 2033–2038. [Google Scholar]
- Schug, T.T.; Berry, D.C.; Shaw, N.S.; Travis, S.N.; Noy, N. Opposing Effects of Retinoic Acid on Cell Growth Result from Alternate Activation of Two Different Nuclear Receptors. Cell 2007, 129, 723–733. [Google Scholar] [CrossRef] [Green Version]
- Available online: https://www.proteinatlas.org/ENSG00000164687-FABP5/pathology (accessed on 15 December 2020).
- Guan, F.; Wang, L.; Hao, S.; Wu, Z.; Bai, J.; Kang, Z.; Zhou, Q.; Chang, H.; Yin, H.; Li, D.; et al. Retinol dehydrogenase-10 promotes development and progression of human glioma via the TWEAK-NF-κB axis. Oncotarget 2017, 8, 105262–105275. [Google Scholar] [CrossRef] [Green Version]
- Fujimura, M.; Katsumata, N.; Tsuda, H.; Uchi, N.; Miyazaki, S.; Hidaka, T.; Sakai, M.; Saito, S. HER2 Is Frequently Over-expressed in Ovarian Clear Cell Adenocarcinoma: Possible Novel Treatment Modality Using Recombinant Monoclonal Antibody against HER2, Trastuzumab. Jpn. J. Cancer Res. 2002, 93, 1250–1257. [Google Scholar] [CrossRef]
- Kim, H.-J.; Yoon, A.; Ryu, J.-Y.; Cho, Y.-J.; Choi, J.-J.; Song, S.Y.; Bang, H.; Lee, J.S.; Cho, W.C.; Choi, C.H.; et al. c-MET as a Potential Therapeutic Target in Ovarian Clear Cell Carcinoma. Sci. Rep. 2016, 6, 38502. [Google Scholar] [CrossRef] [Green Version]
- Wakinoue, S.; Chano, T.; Amano, T.; Isono, T.; Kimura, F.; Kushima, R.; Murakami, T. ADP-ribosylation factor-like 4C predicts worse prognosis in endometriosis-associated ovarian cancers. Cancer Biomark. 2019, 24, 223–229. [Google Scholar] [CrossRef]
- Konstantinopoulos, P.A.; Brady, W.E.; Farley, J.H.; Armstrong, A.; Uyar, D.; Gershenson, D. Phase II study of single-agent cabozantinib in patients with recurrent clear cell ovarian, primary peritoneal or fallopian tube cancer (NRG-GY001). Gynecol. Oncol. 2018, 150, 9–13. [Google Scholar] [CrossRef]
- Farley, J.H.; Brady, W.E.; Fujiwara, K.; Nomura, H.; Yunokawa, M.; Tokunaga, H.; Saitou, M.; Gershenson, D.M. A phase II evaluation of temsirolimus in combination with carboplatin and paclitaxel followed by temsirolimus consolidation as first-line therapy in the treatment of stage III-IV clear cell carcinoma of the ovary. J. Clin. Oncol. 2016, 34, 5531. [Google Scholar] [CrossRef]
- DeVorkin, L.; Hattersley, M.; Kim, P.; Ries, J.; Spowart, J.; Anglesio, M.S.; Levi, S.M.; Huntsman, D.G.; Amaravadi, R.K.; Winkler, J.D.; et al. Autophagy Inhibition Enhances Sunitinib Efficacy in Clear Cell Ovarian Carcinoma. Mol. Cancer Res. 2017, 15, 250–258. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bitler, B.G.; Aird, K.M.; Garipov, A.; Li, H.; Amatangelo, M.; Kossenkov, A.V.; Schultz, D.C.; Liu, Q.; Shih, I.-M.; Conejo-Garcia, J.R.; et al. Synthetic lethality by targeting EZH2 methyltransferase activity in ARID1A-mutated cancers. Nat. Med. 2015, 21, 231–238. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Berns, K.; Caumanns, J.J.; Hijmans, E.M.; Gennissen, A.M.C.; Severson, T.M.; Evers, B.; Wisman, G.B.A.; Meersma, G.J.; Lieftink, C.; Beijersbergen, R.L.; et al. ARID1A mutation sensitizes most ovarian clear cell carcinomas to BET inhibitors. Oncogene 2018, 37, 4611–4625. [Google Scholar] [CrossRef] [PubMed]
- Ogiwara, H.; Takahashi, K.; Sasaki, M.; Kuroda, T.; Yoshida, H.; Watanabe, R.; Maruyama, A.; Makinoshima, H.; Chiwaki, F.; Sasaki, H.; et al. Targeting the Vulnerability of Glutathione Metabolism in ARID1A-Deficient Cancers. Cancer Cell 2019, 35, 177–190.e8. [Google Scholar] [CrossRef] [Green Version]
- Wiegand, K.C.; Hennessy, B.T.; Leung, S.; Wang, Y.; Ju, Z.; Murray, M.; Kalloger, S.; Finlayson, S.; Stemke-Hale, K.; Lu, Y.; et al. A functional proteogenomic analysis of endometrioid and clear cell carcinomas using reverse phase protein array and mutation analysis: Protein expression is histotype-specific and loss of ARID1A/BAF250a is associated with AKT phosphorylation. BMC Cancer 2014, 14, 120. [Google Scholar] [CrossRef] [Green Version]
- Le, D.T.; Durham, J.N.; Smith, K.N.; Wang, H.; Bartlett, B.R.; Aulakh, L.K.; Lu, S.; Kemberling, H.; Wilt, C.; Luber, B.S.; et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science 2017, 357, 409–413. [Google Scholar] [CrossRef] [Green Version]
- Le, D.T.; Uram, J.N.; Wang, H.; Bartlett, B.R.; Kemberling, H.; Eyring, A.D.; Skora, A.D.; Luber, B.S.; Azad, N.S.; Laheru, D.; et al. PD-1 Blockade in Tumors with Mismatch-Repair Deficiency. N. Engl. J. Med. 2015, 372, 2509–2520. [Google Scholar] [CrossRef]
- Samstein, R.M.; Lee, C.-H.; Shoushtari, A.N.; Hellmann, M.D.; Shen, R.; Janjigian, Y.Y.; Barron, D.A.; Zehir, A.; Jordan, E.J.; Omuro, A.; et al. Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nat. Genet. 2019, 51, 202–206. [Google Scholar] [CrossRef]
- Yarchoan, M.; Hopkins, A.; Jaffee, E.M. Tumor Mutational Burden and Response Rate to PD-1 Inhibition. N. Engl. J. Med. 2017, 377, 2500–2501. [Google Scholar] [CrossRef]
- Akbari, M.R.; Zhang, S.; Cragun, D.; Lee, J.-H.; Coppola, D.; McLaughlin, J.R.; Risch, H.A.; Rosen, B.; Shaw, P.; Sellers, T.A.; et al. Correlation between germline mutations in MMR genes and microsatellite instability in ovarian cancer specimens. Fam. Cancer 2017, 16, 351–355. [Google Scholar] [CrossRef] [PubMed]
- Feinberg, J.; Elvin, J.A.; Bellone, S.; Santin, A.D. Identification of ovarian cancer patients for immunotherapy by concurrent assessment of tumor mutation burden (TMB), microsatellite instability (MSI) status, and targetable genomic alterations (GA). Gynecol. Oncol. 2018, 149 (Suppl. 1), 36. [Google Scholar] [CrossRef]
- Jiang, T.; Chen, X.; Su, C.; Ren, S.; Zhou, C. Pan-cancer analysis of ARID1A Alterations as Biomarkers for Immunotherapy Outcomes. J. Cancer 2020, 11, 776–780. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Thomas, L.J.; Vitale, L.; O’Neill, T.; Dolnick, R.Y.; Wallace, P.K.; Minderman, H.; Gergel, L.E.; Forsberg, E.M.; Boyer, J.M.; Storey, J.R.; et al. Development of a Novel Antibody-Drug Conjugate for the Potential Treatment of Ovarian, Lung, and Renal Cell Carcinoma Expressing TIM-1. Mol. Cancer Ther. 2016, 15, 2946–2954. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Goto, M.; Miwa, H.; Shikami, M.; Tsunekawa-Imai, N.; Suganuma, K.; Mizuno, S.; Takahashi, M.; Mizutani, M.; Hanamura, I.; Nitta, M. Importance of Glutamine Metabolism in Leukemia Cells by Energy Production Through TCA Cycle and by Redox Homeostasis. Cancer Investig. 2014, 32, 241–247. [Google Scholar] [CrossRef] [PubMed]
- Roesch, A.; Vultur, A.; Bogeski, I.; Wang, H.; Zimmermann, K.M.; Speicher, D.W.; Körbel, C.; Laschke, M.W.; Gimotty, P.A.; Philipp, S.E.; et al. Overcoming Intrinsic Multidrug Resistance in Melanoma by Blocking the Mitochondrial Respiratory Chain of Slow-Cycling JARID1Bhigh Cells. Cancer Cell 2013, 23, 811–825. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vazquez, F.; Lim, J.-H.; Chim, H.; Bhalla, K.; Girnun, G.; Pierce, K.; Clish, C.B.; Granter, S.R.; Widlund, H.R.; Spiegelman, B.M.; et al. PGC1α Expression Defines a Subset of Human Melanoma Tumors with Increased Mitochondrial Capacity and Resistance to Oxidative Stress. Cancer Cell 2013, 23, 287–301. [Google Scholar] [CrossRef] [Green Version]
- Viale, A.; Pettazzoni, P.; Lyssiotis, C.A.; Ying, H.; Sanchez, N.; Marchesini, M.; Carugo, A.; Green, T.; Seth, S.; Giuliani, V.; et al. Oncogene ablation-resistant pancreatic cancer cells depend on mitochondrial function. Nature 2014, 514, 628–632. [Google Scholar] [CrossRef] [Green Version]
- Molina, J.R.; Sun, Y.; Protopopova, M.; Gera, S.; Bandi, M.; Bristow, C.; McAfoos, T.; Morlacchi, P.; Ackroyd, J.J.; Agip, A.-N.A.; et al. An inhibitor of oxidative phosphorylation exploits cancer vulnerability. Nat. Med. 2018, 24, 1036–1046. [Google Scholar] [CrossRef] [Green Version]
- El-Mir, M.-Y.; Nogueira, V.; Fontaine, E.; Avéret, N.; Rigoulet, M.; Leverve, X. Dimethylbiguanide Inhibits Cell Respiration via an Indirect Effect Targeted on the Respiratory Chain Complex I. J. Biol. Chem. 2000, 275, 223–228. [Google Scholar] [CrossRef] [Green Version]
- Thakur, S.; Daley, B.; Gaskins, K.; Vasko, V.V.; Boufraqech, M.; Patel, D.; Sourbier, C.; Reece, J.M.; Cheng, S.-Y.; Kebebew, E.; et al. Metformin Targets Mitochondrial Glycerophosphate Dehydrogenase to Control Rate of Oxidative Phosphorylation and Growth of Thyroid Cancer In Vitro and In Vivo. Clin. Cancer Res. 2018, 24, 4030–4043. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wheaton, W.W.; Weinberg, S.E.; Hamanaka, R.B.; Soberanes, S.; Sullivan, L.B.; Anso, E.; Glasauer, A.; Dufour, E.; Mutlu, G.M.; Budigner, G.S.; et al. Metformin inhibits mitochondrial complex I of cancer cells to reduce tumorigenesis. eLife 2014, 3, e02242. [Google Scholar] [CrossRef] [PubMed]
- Bridges, H.R.; Jones, A.J.Y.; Pollak, M.N.; Hirst, J. Effects of metformin and other biguanides on oxidative phosphorylation in mitochondria. Biochem. J. 2014, 462, 475–487. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wilcock, C.; Bailey, C.J. Accumulation of metformin by tissues of the normal and diabetic mouse. Xenobiotica 1994, 24, 49–57. [Google Scholar] [CrossRef]
- Owen, M.R.; Doran, E.; Halestrap, A.P. Evidence that metformin exerts its anti-diabetic effects through inhibition of complex 1 of the mitochondrial respiratory chain. Biochem. J. 2000, 348, 607–614. [Google Scholar] [CrossRef]
- Eikawa, S.; Nishida, M.; Mizukami, S.; Yamazaki, C.; Nakayama, E.; Udono, H. Immune-mediated antitumor effect by type 2 diabetes drug, metformin. Proc. Natl. Acad. Sci. USA 2015, 112, 1809–1814. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sanada, S.; Futami, K.; Terada, A.; Yonemoto, K.; Ogasawara, S.; Akiba, J.; Yasumoto, M.; Sumi, A.; Ushijima, K.; Kamura, T.; et al. RECQL1 DNA Repair Helicase: A Potential Therapeutic Target and a Proliferative Marker against Ovarian Cancer. PLoS ONE 2013, 8, e72820. [Google Scholar] [CrossRef]
- Arai, A.; Chano, T.; Futami, K.; Furuichi, Y.; Ikebuchi, K.; Inui, T.; Tameno, H.; Ochi, Y.; Shimada, T.; Hisa, Y.; et al. RECQL1 and WRN Proteins Are Potential Therapeutic Targets in Head and Neck Squamous Cell Carcinoma. Cancer Res. 2011, 71, 4598–4607. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chan, E.M.; Shibue, T.; McFarland, J.M.; Gaeta, B.; Ghandi, M.; Dumont, N.; Gonzalez, A.; McPartlan, J.S.; Li, T.; Zhang, Y.; et al. WRN helicase is a synthetic lethal target in microsatellite unstable cancers. Nat. Cell Biol. 2019, 568, 551–556. [Google Scholar] [CrossRef] [Green Version]
- Sakharkar, M.K.; Sakharkar, K.R.; Pervaiz, S. Druggability of human disease genes. Int. J. Biochem. Cell Biol. 2007, 39, 1156–1164. [Google Scholar] [CrossRef]
Clinical Research | Antioxidant | Result | Reference |
---|---|---|---|
Meta-analysis | Vitamin A | Reduced the risk of ovarian cancer. | [52] |
Meta-analysis | Vitamin C | No significant effect on the risk of ovarian cancer. | [53] |
Systematic review | Vitamin D | No significant effect on the risk of ovarian cancer. | [55] |
Meta-analysis | Vitamin E | No significant effect on the risk of ovarian cancer. | [51] |
Meta-analysis | Flavonoid | Reduced the risk of ovarian cancer. | [56] |
Cohort study | Flavonoid Isothiocyanate | Reduced the risk of ovarian cancer. No significant effect on the risk of ovarian cancer. | [57] |
Pathway | p-Value (RNAseq from Vividly Growing Cancer Spheres) |
---|---|
Hs_Integrated_Cancer_Pathway_WP1971_82939 | 1.02 × 104 |
Hs_Intrinsic_Pathway_for_Apoptosis_WP1841_83332 | 0.002799811 |
Hs_Signaling_by_Retinoic_Acid_WP3323_83286 | 0.003082763 |
Hs_Vitamin_A_and_Carotenoid_Metabolism_WP716_83589 | 0.003229062 |
Hs_BMAL1-CLOCK, NPAS2_activates_circadian_gene_expression_WP3355_83343 | 0.003687094 |
Hs_Integrated_Breast_Cancer_Pathway_WP1984_82941 | 0.004416806 |
Hs_Pre-NOTCH_Expression_and_Processing_WP2786_83418 | 0.004513508 |
Hs_Lipid_storage_and_perilipins_in_skeletal_muscle_WP2887_85092 | 0.011662396 |
Hs_Uptake_and_function_of_anthrax_toxins_WP3390_83389 | 0.013593029 |
Hs_miRNA_Regulation_of_DNA_Damage_Response_WP1530_84694 | 0.013785134 |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 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 (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Amano, T.; Murakami, A.; Murakami, T.; Chano, T. Antioxidants and Therapeutic Targets in Ovarian Clear Cell Carcinoma. Antioxidants 2021, 10, 187. https://doi.org/10.3390/antiox10020187
Amano T, Murakami A, Murakami T, Chano T. Antioxidants and Therapeutic Targets in Ovarian Clear Cell Carcinoma. Antioxidants. 2021; 10(2):187. https://doi.org/10.3390/antiox10020187
Chicago/Turabian StyleAmano, Tsukuru, Atsushi Murakami, Takashi Murakami, and Tokuhiro Chano. 2021. "Antioxidants and Therapeutic Targets in Ovarian Clear Cell Carcinoma" Antioxidants 10, no. 2: 187. https://doi.org/10.3390/antiox10020187
APA StyleAmano, T., Murakami, A., Murakami, T., & Chano, T. (2021). Antioxidants and Therapeutic Targets in Ovarian Clear Cell Carcinoma. Antioxidants, 10(2), 187. https://doi.org/10.3390/antiox10020187