New Insights into Ferroptosis Initiating Therapies (FIT) by Targeting the Rewired Lipid Metabolism in Ovarian Cancer Peritoneal Metastases
Abstract
:1. Introduction—An Overview of the Prevalence and Burden of Ovarian Cancer
2. Difficulties in Current Ovarian Cancer Treatment
3. Alternative Therapeutic Approaches to Ovarian Cancer
3.1. Targeted Therapy in Ovarian Cancer
3.2. Immunotherapy in Ovarian Cancer
4. The Tumor Microenvironment (TME)-Stimulated Chemoresistance Is a Major Hurdle in the Successive Treatment of Ovarian Cancer
5. Augmented Fatty Acid Metabolism (FAM) Is Conducive to the Metastatic Aggression of Ovarian Cancer
6. Targeting Aberrant FAM with Ferroptosis-Inducing Compounds (FINs) as the Swiss Army Knife against Metastatic Ovarian Cancer
7. Conclusions and Perspectives
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
References
- Reid, B.M.; Permuth, J.B.; Sellers, T.A. Epidemiology of ovarian cancer: A review. Cancer Biol. Med. 2017, 14, 9–32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Key Statistics for Ovarian Cancer, American Cancer Society’s Cancer Statistics Center. Available online: https://www.cancer.org/cancer/ovarian-cancer/about/key-statistics.html#written_by (accessed on 12 January 2022).
- Pokhriyal, R.; Hariprasad, R.; Kumar, L.; Hariprasad, G. Chemotherapy Resistance in Advanced Ovarian Cancer Patients. Biomark. Cancer 2019, 11, 1179299X19860815. [Google Scholar] [CrossRef] [PubMed]
- Holschneider, C.H.; Berek, J.S. Ovarian cancer: Epidemiology, biology, and prognostic factors. Semin. Surg. Oncol. 2000, 19, 3–10. [Google Scholar] [CrossRef] [PubMed]
- Ovarian and Peritoneal Cancer, Centre for Health Protection, Department of Health, The Government of the Hong Kong Special Administrative Region. Available online: https://www.chp.gov.hk/en/healthtopics/content/25/6625.html (accessed on 4 January 2022).
- Preston, C.C.; Goode, E.L.; Hartmann, L.C.; Kalli, K.R.; Knutson, K.L. Immunity and immune suppression in human ovarian cancer. Immunotherapy 2011, 3, 539–556. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lengyel, E. Ovarian cancer development and metastasis. Am. J. Pathol. 2010, 177, 1053–1064. [Google Scholar] [CrossRef]
- Ahmed, N.; Stenvers, K.L. Getting to know ovarian cancer ascites: Opportunities for targeted therapy-based translational research. Front. Oncol. 2013, 3, 256. [Google Scholar] [CrossRef] [Green Version]
- Yung, M.M.H.; Siu, M.K.Y.; Ngan, H.Y.S.; Chan, D.W.; Chan, K.K.L. Orchestrated Action of AMPK Activation and Combined VEGF/PD-1 Blockade with Lipid Metabolic Tunning as Multi-Target Therapeutics against Ovarian Cancers. Int. J. Mol. Sci. 2022, 23, 6857. [Google Scholar] [CrossRef]
- Ivanova, V.; Dikov, T.; Dimitrova, N. Histologic subtypes of ovarian carcinoma: Selected diagnostic and classification problems in Bulgaria: Is low hospital volume an issue? Tumori 2017, 103, 148–154. [Google Scholar] [CrossRef]
- Matulonis, U.A.; Sood, A.K.; Fallowfield, L.; Howitt, B.E.; Sehouli, J.; Karlan, B.Y. Ovarian cancer. Nat. Rev. Dis. Prim. 2016, 2, 16061. [Google Scholar] [CrossRef]
- Hossain, K.R.; Escobar Bermeo, J.D.; Warton, K.; Valenzuela, S.M. New Approaches and Biomarker Candidates for the Early Detection of Ovarian Cancer. Front. Bioeng. Biotechnol. 2022, 10, 819183. [Google Scholar] [CrossRef]
- Rakina, M.; Kazakova, A.; Villert, A.; Kolomiets, L.; Larionova, I. Spheroid Formation and Peritoneal Metastasis in Ovarian Cancer: The Role of Stromal and Immune Components. Int. J. Mol. Sci. 2022, 23, 6215. [Google Scholar] [CrossRef]
- Bast, R.C., Jr.; Matulonis, U.A.; Sood, A.K.; Ahmed, A.A.; Amobi, A.E.; Balkwill, F.R.; Wielgos-Bonvallet, M.; Bowtell, D.D.L.; Brenton, J.D.; Brugge, J.S.; et al. Critical questions in ovarian cancer research and treatment: Report of an American Association for Cancer Research Special Conference. Cancer 2019, 125, 1963–1972. [Google Scholar] [CrossRef]
- Wright, A.A.; Bohlke, K.; Armstrong, D.K.; Bookman, M.A.; Cliby, W.A.; Coleman, R.L.; Dizon, D.S.; Kash, J.J.; Meyer, L.A.; Moore, K.N.; et al. Neoadjuvant Chemotherapy for Newly Diagnosed, Advanced Ovarian Cancer: Society of Gynecologic Oncology and American Society of Clinical Oncology Clinical Practice Guideline. J. Clin. Oncol. 2016, 34, 3460–3473. [Google Scholar] [CrossRef]
- du Bois, A.; Reuss, A.; Pujade-Lauraine, E.; Harter, P.; Ray-Coquard, I.; Pfisterer, J. Role of surgical outcome as prognostic factor in advanced epithelial ovarian cancer: A combined exploratory analysis of 3 prospectively randomized phase 3 multicenter trials: By the Arbeitsgemeinschaft Gynaekologische Onkologie Studiengruppe Ovarialkarzinom (AGO-OVAR) and the Groupe d’Investigateurs Nationaux Pour les Etudes des Cancers de l’Ovaire (GINECO). Cancer 2009, 115, 1234–1244. [Google Scholar] [CrossRef]
- Barbolina, M.V. Molecular Mechanisms Regulating Organ-Specific Metastases in Epithelial Ovarian Carcinoma. Cancers 2018, 10, 444. [Google Scholar] [CrossRef] [Green Version]
- Kuroki, L.; Guntupalli, S.R. Treatment of epithelial ovarian cancer. BMJ 2020, 371, m3773. [Google Scholar] [CrossRef]
- Curley, M.D.; Garrett, L.A.; Schorge, J.O.; Foster, R.; Rueda, B.R. Evidence for cancer stem cells contributing to the pathogenesis of ovarian cancer. Front. Biosci. 2011, 16, 368–392. [Google Scholar] [CrossRef] [Green Version]
- 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] [Green Version]
- Tan, D.S.; Agarwal, R.; Kaye, S.B. Mechanisms of transcoelomic metastasis in ovarian cancer. Lancet Oncol. 2006, 7, 925–934. [Google Scholar] [CrossRef]
- Yim, G.W.; Eoh, K.J.; Kim, S.W.; Nam, E.J.; Kim, Y.T. Malnutrition Identified by the Nutritional Risk Index and Poor Prognosis in Advanced Epithelial Ovarian Carcinoma. Nutr. Cancer 2016, 68, 772–779. [Google Scholar] [CrossRef]
- Balogun, N.; Forbes, A.; Widschwendter, M.; Lanceley, A. Noninvasive nutritional management of ovarian cancer patients: Beyond intestinal obstruction. Int. J. Gynecol. Cancer Off. J. Int. Gynecol. Cancer Soc. 2012, 22, 1089–1095. [Google Scholar] [CrossRef] [PubMed]
- Sato, M.; Kawana, K.; Adachi, K.; Fujimoto, A.; Yoshida, M.; Nakamura, H.; Nishida, H.; Inoue, T.; Taguchi, A.; Ogishima, J.; et al. Detachment from the primary site and suspension in ascites as the initial step in metabolic reprogramming and metastasis to the omentum in ovarian cancer. Oncol. Lett. 2018, 15, 1357–1361. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Padma, V.V. An overview of targeted cancer therapy. Biomedicine 2015, 5, 19. [Google Scholar] [CrossRef] [PubMed]
- Zhong, L.; Li, Y.; Xiong, L.; Wang, W.; Wu, M.; Yuan, T.; Yang, W.; Tian, C.; Miao, Z.; Wang, T.; et al. Small molecules in targeted cancer therapy: Advances, challenges, and future perspectives. Signal Transduct. Target. Ther. 2021, 6, 201. [Google Scholar] [CrossRef] [PubMed]
- Zahavi, D.; Weiner, L. Monoclonal Antibodies in Cancer Therapy. Antibodies 2020, 9, 34. [Google Scholar] [CrossRef]
- Wiggans, A.J.; Cass, G.K.; Bryant, A.; Lawrie, T.A.; Morrison, J. Poly(ADP-ribose) polymerase (PARP) inhibitors for the treatment of ovarian cancer. Cochrane Database Syst. Rev. 2015, CD007929. [Google Scholar] [CrossRef]
- Curtin, N.J.; Szabo, C. Therapeutic applications of PARP inhibitors: Anticancer therapy and beyond. Mol. Aspects Med. 2013, 34, 1217–1256. [Google Scholar] [CrossRef] [Green Version]
- Mukherjee, S.; Pal, M.; Mukhopadhyay, S.; Das, I.; Hazra, R.; Ghosh, S.; Mondal, R.K.; Bal, R. VEGF Expression to Support Targeted Therapy in Ovarian Surface Epithelial Neoplasms. J. Clin. Diagn. Res. 2017, 11, EC43–EC46. [Google Scholar] [CrossRef]
- Choi, H.J.; Armaiz Pena, G.N.; Pradeep, S.; Cho, M.S.; Coleman, R.L.; Sood, A.K. Anti-vascular therapies in ovarian cancer: Moving beyond anti-VEGF approaches. Cancer Metastasis Rev. 2015, 34, 19–40. [Google Scholar] [CrossRef] [Green Version]
- Fraser, M.; Leung, B.; Jahani-Asl, A.; Yan, X.; Thompson, W.E.; Tsang, B.K. Chemoresistance in human ovarian cancer: The role of apoptotic regulators. Reprod. Biol. Endocrinol. 2003, 1, 66. [Google Scholar] [CrossRef]
- Kim, S.; Han, Y.; Kim, S.I.; Kim, H.S.; Kim, S.J.; Song, Y.S. Tumor evolution and chemoresistance in ovarian cancer. NPJ Precis. Oncol. 2018, 2, 20. [Google Scholar] [CrossRef] [Green Version]
- Limagne, E.; Nuttin, L.; Thibaudin, M.; Jacquin, E.; Aucagne, R.; Bon, M.; Revy, S.; Barnestein, R.; Ballot, E.; Truntzer, C.; et al. MEK inhibition overcomes chemoimmunotherapy resistance by inducing CXCL10 in cancer cells. Cancer Cell 2022, 40, 136–152.e112. [Google Scholar] [CrossRef]
- Yung, M.M.; Ross, F.A.; Hardie, D.G.; Leung, T.H.; Zhan, J.; Ngan, H.Y.; Chan, D.W. Bitter Melon (Momordica charantia) Extract Inhibits Tumorigenicity and Overcomes Cisplatin-Resistance in Ovarian Cancer Cells Through Targeting AMPK Signaling Cascade. Integr. Cancer Ther. 2015, 15, 376–389. [Google Scholar] [CrossRef] [Green Version]
- Chan, D.W.; Yung, M.M.; Chan, Y.S.; Xuan, Y.; Yang, H.; Xu, D.; Zhan, J.B.; Chan, K.K.; Ng, T.B.; Ngan, H.Y. MAP30 protein from Momordica charantia is therapeutic and has synergic activity with cisplatin against ovarian cancer in vivo by altering metabolism and inducing ferroptosis. Pharmacol. Res. 2020, 161, 105157. [Google Scholar] [CrossRef]
- Yung, M.M.; Ngan, H.Y.; Chan, D.W. Targeting AMPK signaling in combating ovarian cancers: Opportunities and challenges. Acta Biochim. Biophys. Sin. 2016, 48, 301–317. [Google Scholar] [CrossRef] [Green Version]
- Agarwal, R.; Kaye, S.B. Ovarian cancer: Strategies for overcoming resistance to chemotherapy. Nat. Rev. Cancer 2003, 3, 502–516. [Google Scholar] [CrossRef]
- Cramer-van der Welle, C.M.; Verschueren, M.V.; Tonn, M.; Peters, B.J.M.; Schramel, F.; Klungel, O.H.; Groen, H.J.M.; van de Garde, E.M.W.; Santeon, N.S.G. Real-world outcomes versus clinical trial results of immunotherapy in stage IV non-small cell lung cancer (NSCLC) in the Netherlands. Sci. Rep. 2021, 11, 6306. [Google Scholar] [CrossRef]
- Mantia-Smaldone, G.M.; Corr, B.; Chu, C.S. Immunotherapy in ovarian cancer. Hum. Vaccin. Immunother. 2012, 8, 1179–1191. [Google Scholar] [CrossRef] [Green Version]
- Zhang, L.; Conejo-Garcia, J.R.; Katsaros, D.; Gimotty, P.A.; Massobrio, M.; Regnani, G.; Makrigiannakis, A.; Gray, H.; Schlienger, K.; Liebman, M.N.; et al. Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer. N. Engl. J. Med. 2003, 348, 203–213. [Google Scholar] [CrossRef] [Green Version]
- Santoiemma, P.P.; Powell, D.J., Jr. Tumor infiltrating lymphocytes in ovarian cancer. Cancer Biol. Ther. 2015, 16, 807–820. [Google Scholar] [CrossRef]
- Gandhi, L.; Rodriguez-Abreu, D.; Gadgeel, S.; Esteban, E.; Felip, E.; De Angelis, F.; Domine, M.; Clingan, P.; Hochmair, M.J.; Powell, S.F.; et al. Pembrolizumab plus Chemotherapy in Metastatic Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2018, 378, 2078–2092. [Google Scholar] [CrossRef] [PubMed]
- Yeldag, G.; Rice, A.; Del Rio Hernandez, A. Chemoresistance and the Self-Maintaining Tumor Microenvironment. Cancers 2018, 10, 471. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Perez-Ruiz, E.; Melero, I.; Kopecka, J.; Sarmento-Ribeiro, A.B.; Garcia-Aranda, M.; De Las Rivas, J. Cancer immunotherapy resistance based on immune checkpoints inhibitors: Targets, biomarkers, and remedies. Drug Resist. Updat. 2020, 53, 100718. [Google Scholar] [CrossRef] [PubMed]
- Wang, S.; Xie, K.; Liu, T. Cancer Immunotherapies: From Efficacy to Resistance Mechanisms—Not Only Checkpoint Matters. Front. Immunol. 2021, 12, 690112. [Google Scholar] [CrossRef] [PubMed]
- Kim, S.; Kim, B.; Song, Y.S. Ascites modulates cancer cell behavior, contributing to tumor heterogeneity in ovarian cancer. Cancer Sci. 2016, 107, 1173–1178. [Google Scholar] [CrossRef] [Green Version]
- Paraiso, K.H.; Smalley, K.S. Fibroblast-mediated drug resistance in cancer. Biochem. Pharmacol. 2013, 85, 1033–1041. [Google Scholar] [CrossRef]
- Gao, Q.; Yang, Z.; Xu, S.; Li, X.; Yang, X.; Jin, P.; Liu, Y.; Zhou, X.; Zhang, T.; Gong, C.; et al. Heterotypic CAF-tumor spheroids promote early peritoneal metastatis of ovarian cancer. J. Exp. Med. 2019, 216, 688–703. [Google Scholar] [CrossRef] [Green Version]
- Lane, D.; Matte, I.; Garde-Granger, P.; Laplante, C.; Carignan, A.; Rancourt, C.; Piche, A. Inflammation-regulating factors in ascites as predictive biomarkers of drug resistance and progression-free survival in serous epithelial ovarian cancers. BMC Cancer 2015, 15, 492. [Google Scholar] [CrossRef] [Green Version]
- Finley, L.W.; Carracedo, A.; Lee, J.; Souza, A.; Egia, A.; Zhang, J.; Teruya-Feldstein, J.; Moreira, P.I.; Cardoso, S.M.; Clish, C.B.; et al. SIRT3 opposes reprogramming of cancer cell metabolism through HIF1alpha destabilization. Cancer Cell 2011, 19, 416–428. [Google Scholar] [CrossRef] [Green Version]
- Chen, R.R.; Yung, M.M.H.; Xuan, Y.; Zhan, S.; Leung, L.L.; Liang, R.R.; Leung, T.H.Y.; Yang, H.; Xu, D.; Sharma, R.; et al. Targeting of lipid metabolism with a metabolic inhibitor cocktail eradicates peritoneal metastases in ovarian cancer cells. Commun. Biol. 2019, 2, 281. [Google Scholar] [CrossRef]
- Xuan, Y.; Wang, H.; Yung, M.M.; Chen, F.; Chan, W.S.; Chan, Y.S.; Tsui, S.W.; Ngan, Y.S.; Chan, K.L.; Chan, D.W. SCD1/FADS2 fatty acid desaturases equipoise lipid metabolic activity and redox-driven ferroptosis in ascites-derived ovarian cancer cells. Theranostics 2022, 12, 3534–3552. [Google Scholar] [CrossRef]
- Reymond, M.A. Definition and semantics: “Peritoneal Carcinomatosis” should be abandoned and replaced by “Peritoneal Metastasis”. Pleura Peritoneum 2017, 2, 119–120. [Google Scholar] [CrossRef]
- Harter, P.; Hahmann, M.; Lueck, H.J.; Poelcher, M.; Wimberger, P.; Ortmann, O.; Canzler, U.; Richter, B.; Wagner, U.; Hasenburg, A.; et al. Surgery for recurrent ovarian cancer: Role of peritoneal carcinomatosis: Exploratory analysis of the DESKTOP I Trial about risk factors, surgical implications, and prognostic value of peritoneal carcinomatosis. Ann. Surg. Oncol. 2009, 16, 1324–1330. [Google Scholar] [CrossRef]
- Gerber, S.A.; Rybalko, V.Y.; Bigelow, C.E.; Lugade, A.A.; Foster, T.H.; Frelinger, J.G.; Lord, E.M. Preferential attachment of peritoneal tumor metastases to omental immune aggregates and possible role of a unique vascular microenvironment in metastatic survival and growth. Am. J. Pathol. 2006, 169, 1739–1752. [Google Scholar] [CrossRef] [Green Version]
- Nieman, K.M.; Kenny, H.A.; Penicka, C.V.; Ladanyi, A.; Buell-Gutbrod, R.; Zillhardt, M.R.; Romero, I.L.; Carey, M.S.; Mills, G.B.; Hotamisligil, G.S.; et al. Adipocytes promote ovarian cancer metastasis and provide energy for rapid tumor growth. Nat. Med. 2011, 17, 1498–1503. [Google Scholar] [CrossRef] [Green Version]
- Ma, X. The omentum, a niche for premetastatic ovarian cancer. J. Exp. Med. 2020, 217, e20192312. [Google Scholar] [CrossRef] [Green Version]
- Evans, K.J. Solid Omental Tumors, Medscape. Available online: https://emedicine.medscape.com/article/193622-overview (accessed on 3 September 2021).
- Sangisetty, S.L.; Miner, T.J. Malignant ascites: A review of prognostic factors, pathophysiology and therapeutic measures. World J. Gastrointest. Surg. 2012, 4, 87–95. [Google Scholar] [CrossRef]
- Yung, M.M.; Tang, H.W.; Cai, P.C.; Leung, T.H.; Ngu, S.F.; Chan, K.K.; Xu, D.; Yang, H.; Ngan, H.Y.; Chan, D.W. GRO-alpha and IL-8 enhance ovarian cancer metastatic potential via the CXCR2-mediated TAK1/NFkappaB signaling cascade. Theranostics 2018, 8, 1270–1285. [Google Scholar] [CrossRef]
- Clark, R.; Krishnan, V.; Schoof, M.; Rodriguez, I.; Theriault, B.; Chekmareva, M.; Rinker-Schaeffer, C. Milky spots promote ovarian cancer metastatic colonization of peritoneal adipose in experimental models. Am. J. Pathol. 2013, 183, 576–591. [Google Scholar] [CrossRef] [Green Version]
- Liberti, M.V.; Locasale, J.W. The Warburg Effect: How Does it Benefit Cancer Cells? Trends Biochem. Sci. 2016, 41, 211–218. [Google Scholar] [CrossRef]
- Motohara, T.; Masuda, K.; Morotti, M.; Zheng, Y.; El-Sahhar, S.; Chong, K.Y.; Wietek, N.; Alsaadi, A.; Carrami, E.M.; Hu, Z.; et al. An evolving story of the metastatic voyage of ovarian cancer cells: Cellular and molecular orchestration of the adipose-rich metastatic microenvironment. Oncogene 2019, 38, 2885–2898. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Leinster, D.A.; Kulbe, H.; Everitt, G.; Thompson, R.; Perretti, M.; Gavins, F.N.; Cooper, D.; Gould, D.; Ennis, D.P.; Lockley, M.; et al. The peritoneal tumour microenvironment of high-grade serous ovarian cancer. J. Pathol. 2012, 227, 136–145. [Google Scholar] [CrossRef] [Green Version]
- Wang, X.; Yung, M.M.H.; Sharma, R.; Chen, F.; Poon, Y.T.; Lam, W.Y.; Li, B.; Ngan, H.Y.S.; Chan, K.K.L.; Chan, D.W. Epigenetic Silencing of miR-33b Promotes Peritoneal Metastases of Ovarian Cancer by Modulating the TAK1/FASN/CPT1A/NF-kappaB Axis. Cancers 2021, 13, 4795. [Google Scholar] [CrossRef]
- Lu, J. The Warburg metabolism fuels tumor metastasis. Cancer Metastasis Rev. 2019, 38, 157–164. [Google Scholar] [CrossRef] [PubMed]
- Yeung, T.L.; Leung, C.S.; Yip, K.P.; Au Yeung, C.L.; Wong, S.T.; Mok, S.C. Cellular and molecular processes in ovarian cancer metastasis. A Review in the Theme: Cell and Molecular Processes in Cancer Metastasis. Am. J. Physiol. Cell Physiol. 2015, 309, C444–C456. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ma, Y.; Temkin, S.M.; Hawkridge, A.M.; Guo, C.; Wang, W.; Wang, X.Y.; Fang, X. Fatty acid oxidation: An emerging facet of metabolic transformation in cancer. Cancer Lett. 2018, 435, 92–100. [Google Scholar] [CrossRef]
- Ferreira, C.A.; Ni, D.; Rosenkrans, Z.T.; Cai, W. Scavenging of reactive oxygen and nitrogen species with nanomaterials. Nano Res. 2018, 11, 4955–4984. [Google Scholar] [CrossRef]
- Barrera, G. Oxidative stress and lipid peroxidation products in cancer progression and therapy. ISRN Oncol. 2012, 2012, 137289. [Google Scholar] [CrossRef] [Green Version]
- Bhattacharyya, A.; Chattopadhyay, R.; Mitra, S.; Crowe, S.E. Oxidative stress: An essential factor in the pathogenesis of gastrointestinal mucosal diseases. Physiol. Rev. 2014, 94, 329–354. [Google Scholar] [CrossRef] [Green Version]
- Singh, A.; Kukreti, R.; Saso, L.; Kukreti, S. Oxidative Stress: A Key Modulator in Neurodegenerative Diseases. Molecules 2019, 24, 1583. [Google Scholar] [CrossRef]
- Dixon, S.J.; Lemberg, K.M.; Lamprecht, M.R.; Skouta, R.; Zaitsev, E.M.; Gleason, C.E.; Patel, D.N.; Bauer, A.J.; Cantley, A.M.; Yang, W.S.; et al. Ferroptosis: An iron-dependent form of nonapoptotic cell death. Cell 2012, 149, 1060–1072. [Google Scholar] [CrossRef] [Green Version]
- Li, D.; Li, Y. The interaction between ferroptosis and lipid metabolism in cancer. Signal Transduct. Target. Ther. 2020, 5, 108. [Google Scholar] [CrossRef]
- Li, J.; Cao, F.; Yin, H.L.; Huang, Z.J.; Lin, Z.T.; Mao, N.; Sun, B.; Wang, G. Ferroptosis: Past, present and future. Cell Death Dis. 2020, 11, 88. [Google Scholar] [CrossRef] [Green Version]
- Yagoda, N.; von Rechenberg, M.; Zaganjor, E.; Bauer, A.J.; Yang, W.S.; Fridman, D.J.; Wolpaw, A.J.; Smukste, I.; Peltier, J.M.; Boniface, J.J.; et al. RAS-RAF-MEK-dependent oxidative cell death involving voltage-dependent anion channels. Nature 2007, 447, 864–868. [Google Scholar] [CrossRef] [Green Version]
- Yu, H.; Guo, P.; Xie, X.; Wang, Y.; Chen, G. Ferroptosis, a new form of cell death, and its relationships with tumourous diseases. J. Cell Mol. Med. 2017, 21, 648–657. [Google Scholar] [CrossRef] [Green Version]
- Latunde-Dada, G.O. Ferroptosis: Role of lipid peroxidation, iron and ferritinophagy. Biochim. Biophys. Acta Gen. Subj. 2017, 1861, 1893–1900. [Google Scholar] [CrossRef] [Green Version]
- Cao, J.Y.; Dixon, S.J. Mechanisms of ferroptosis. Cell. Mol. Life Sci. 2016, 73, 2195–2209. [Google Scholar] [CrossRef] [Green Version]
- Xie, Y.; Hou, W.; Song, X.; Yu, Y.; Huang, J.; Sun, X.; Kang, R.; Tang, D. Ferroptosis: Process and function. Cell Death Differ. 2016, 23, 369–379. [Google Scholar] [CrossRef] [Green Version]
- Yang, W.S.; Stockwell, B.R. Synthetic lethal screening identifies compounds activating iron-dependent, nonapoptotic cell death in oncogenic-RAS-harboring cancer cells. Chem. Biol. 2008, 15, 234–245. [Google Scholar] [CrossRef] [Green Version]
- Mou, Y.; Wang, J.; Wu, J.; He, D.; Zhang, C.; Duan, C.; Li, B. Ferroptosis, a new form of cell death: Opportunities and challenges in cancer. J. Hematol. Oncol. 2019, 12, 34. [Google Scholar] [CrossRef]
- Zhang, C.; Liu, X.; Jin, S.; Chen, Y.; Guo, R. Ferroptosis in cancer therapy: A novel approach to reversing drug resistance. Mol. Cancer 2022, 21, 47. [Google Scholar] [CrossRef] [PubMed]
- Li, Y.; Yan, H.; Xu, X.; Liu, H.; Wu, C.; Zhao, L. Erastin/sorafenib induces cisplatin-resistant non-small cell lung cancer cell ferroptosis through inhibition of the Nrf2/xCT pathway. Oncol. Lett. 2020, 19, 323–333. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hong, T.; Lei, G.; Chen, X.; Li, H.; Zhang, X.; Wu, N.; Zhao, Y.; Zhang, Y.; Wang, J. PARP inhibition promotes ferroptosis via repressing SLC7A11 and synergizes with ferroptosis inducers in BRCA-proficient ovarian cancer. Redox Biol. 2021, 42, 101928. [Google Scholar] [CrossRef] [PubMed]
- Chen, T.C.; Chuang, J.Y.; Ko, C.Y.; Kao, T.J.; Yang, P.Y.; Yu, C.H.; Liu, M.S.; Hu, S.L.; Tsai, Y.T.; Chan, H.; et al. AR ubiquitination induced by the curcumin analog suppresses growth of temozolomide-resistant glioblastoma through disrupting GPX4-Mediated redox homeostasis. Redox Biol. 2020, 30, 101413. [Google Scholar] [CrossRef] [PubMed]
- Doll, S.; Proneth, B.; Tyurina, Y.Y.; Panzilius, E.; Kobayashi, S.; Ingold, I.; Irmler, M.; Beckers, J.; Aichler, M.; Walch, A.; et al. ACSL4 dictates ferroptosis sensitivity by shaping cellular lipid composition. Nat. Chem. Biol. 2017, 13, 91–98. [Google Scholar] [CrossRef]
- Kagan, V.E.; Mao, G.; Qu, F.; Angeli, J.P.; Doll, S.; Croix, C.S.; Dar, H.H.; Liu, B.; Tyurin, V.A.; Ritov, V.B.; et al. Oxidized arachidonic and adrenic PEs navigate cells to ferroptosis. Nat. Chem. Biol. 2017, 13, 81–90. [Google Scholar] [CrossRef] [Green Version]
- Weibel, G.L.; Joshi, M.R.; Wei, C.; Bates, S.R.; Blair, I.A.; Rothblat, G.H. 15(S)-Lipoxygenase-1 associates with neutral lipid droplets in macrophage foam cells: Evidence of lipid droplet metabolism. J. Lipid Res. 2009, 50, 2371–2376. [Google Scholar] [CrossRef] [Green Version]
- Shah, R.; Shchepinov, M.S.; Pratt, D.A. Resolving the Role of Lipoxygenases in the Initiation and Execution of Ferroptosis. ACS Cent. Sci. 2018, 4, 387–396. [Google Scholar] [CrossRef]
- Tang, Z.; Huang, Z.; Huang, Y.; Chen, Y.; Huang, M.; Liu, H.; Ye, Q.A.; Zhao, J.; Jia, B. Ferroptosis: The Silver Lining of Cancer Therapy. Front Cell. Dev. Biol. 2021, 9, 765859. [Google Scholar] [CrossRef]
- Lu, B.; Chen, X.B.; Ying, M.D.; He, Q.J.; Cao, J.; Yang, B. The Role of Ferroptosis in Cancer Development and Treatment Response. Front. Pharmacol. 2017, 8, 992. [Google Scholar] [CrossRef]
- Sato, M.; Kusumi, R.; Hamashima, S.; Kobayashi, S.; Sasaki, S.; Komiyama, Y.; Izumikawa, T.; Conrad, M.; Bannai, S.; Sato, H. The ferroptosis inducer erastin irreversibly inhibits system xc- and synergizes with cisplatin to increase cisplatin’s cytotoxicity in cancer cells. Sci. Rep. 2018, 8, 968. [Google Scholar] [CrossRef] [Green Version]
- Zhou, H.H.; Chen, X.; Cai, L.Y.; Nan, X.W.; Chen, J.H.; Chen, X.X.; Yang, Y.; Xing, Z.H.; Wei, M.N.; Li, Y.; et al. Erastin Reverses ABCB1-Mediated Docetaxel Resistance in Ovarian Cancer. Front. Oncol. 2019, 9, 1398. [Google Scholar] [CrossRef] [Green Version]
- Lu, X.; Meng, T. Depletion of tumor-associated macrophages enhances the anti-tumor effect of docetaxel in a murine epithelial ovarian cancer. Immunobiology 2019, 224, 355–361. [Google Scholar] [CrossRef]
- Tesfay, L.; Paul, B.T.; Konstorum, A.; Deng, Z.; Cox, A.O.; Lee, J.; Furdui, C.M.; Hegde, P.; Torti, F.M.; Torti, S.V. Stearoyl-CoA Desaturase 1 Protects Ovarian Cancer Cells from Ferroptotic Cell Death. Cancer Res. 2019, 79, 5355–5366. [Google Scholar] [CrossRef]
- Wu, Y.; Yu, C.; Luo, M.; Cen, C.; Qiu, J.; Zhang, S.; Hu, K. Ferroptosis in Cancer Treatment: Another Way to Rome. Front. Oncol. 2020, 10, 571127. [Google Scholar] [CrossRef]
- Sacco, A.; Battaglia, A.M.; Botta, C.; Aversa, I.; Mancuso, S.; Costanzo, F.; Biamonte, F. Iron Metabolism in the Tumor Microenvironment-Implications for Anti-Cancer Immune Response. Cells 2021, 10, 303. [Google Scholar] [CrossRef]
- Ludwig, H.; Evstatiev, R.; Kornek, G.; Aapro, M.; Bauernhofer, T.; Buxhofer-Ausch, V.; Fridrik, M.; Geissler, D.; Geissler, K.; Gisslinger, H.; et al. Iron metabolism and iron supplementation in cancer patients. Wien Klin. Wochenschr. 2015, 127, 907–919. [Google Scholar] [CrossRef] [Green Version]
- Pfeifhofer-Obermair, C.; Tymoszuk, P.; Petzer, V.; Weiss, G.; Nairz, M. Iron in the Tumor Microenvironment-Connecting the Dots. Front. Oncol. 2018, 8, 549. [Google Scholar] [CrossRef] [Green Version]
- Basuli, D.; Tesfay, L.; Deng, Z.; Paul, B.; Yamamoto, Y.; Ning, G.; Xian, W.; McKeon, F.; Lynch, M.; Crum, C.P.; et al. Iron addiction: A novel therapeutic target in ovarian cancer. Oncogene 2017, 36, 4089–4099. [Google Scholar] [CrossRef] [Green Version]
- Wang, F.; Lv, H.; Zhao, B.; Zhou, L.; Wang, S.; Luo, J.; Liu, J.; Shang, P. Iron and leukemia: New insights for future treatments. J. Exp. Clin. Cancer Res. 2019, 38, 406. [Google Scholar] [CrossRef]
- Wu, J.; Lu, Y.; Lee, A.; Pan, X.; Yang, X.; Zhao, X.; Lee, R.J. Reversal of multidrug resistance by transferrin-conjugated liposomes co-encapsulating doxorubicin and verapamil. J. Pharm. Pharm. Sci. 2007, 10, 350–357. [Google Scholar] [PubMed]
- Zhou, Y.; Shen, Y.; Chen, C.; Sui, X.; Yang, J.; Wang, L.; Zhou, J. The crosstalk between autophagy and ferroptosis: What can we learn to target drug resistance in cancer? Cancer Biol. Med. 2019, 16, 630–646. [Google Scholar] [CrossRef] [PubMed]
- Liang, W.; Ferrara, N. Iron Metabolism in the Tumor Microenvironment: Contributions of Innate Immune Cells. Front. Immunol. 2020, 11, 626812. [Google Scholar] [CrossRef] [PubMed]
- Zou, Y.; Li, H.; Graham, E.T.; Deik, A.A.; Eaton, J.K.; Wang, W.; Sandoval-Gomez, G.; Clish, C.B.; Doench, J.G.; Schreiber, S.L. Cytochrome P450 oxidoreductase contributes to phospholipid peroxidation in ferroptosis. Nat. Chem. Biol. 2020, 16, 302–309. [Google Scholar] [CrossRef]
- Dufrusine, B.; Di Francesco, A.; Oddi, S.; Scipioni, L.; Angelucci, C.B.; D’Addario, C.; Serafini, M.; Hafner, A.K.; Steinhilber, D.; Maccarrone, M.; et al. Iron-Dependent Trafficking of 5-Lipoxygenase and Impact on Human Macrophage Activation. Front. Immunol. 2019, 10, 1347. [Google Scholar] [CrossRef] [Green Version]
- Stamatikos, A.D.; Paton, C.M. Role of stearoyl-CoA desaturase-1 in skeletal muscle function and metabolism. Am. J. Physiol. Endocrinol. Metab. 2013, 305, E767–E775. [Google Scholar] [CrossRef] [Green Version]
- Rockfield, S.; Chhabra, R.; Robertson, M.; Rehman, N.; Bisht, R.; Nanjundan, M. Links Between Iron and Lipids: Implications in Some Major Human Diseases. Pharmaceuticals 2018, 11, 113. [Google Scholar] [CrossRef] [Green Version]
- Pan, Q.; Law, C.O.K.; Yung, M.M.H.; Han, K.C.; Pon, Y.L.; Lau, T.C.K. Novel RNA aptamers targeting gastrointestinal cancer biomarkers CEA, CA50 and CA72-4 with superior affinity and specificity. PLoS ONE 2018, 13, e0198980. [Google Scholar] [CrossRef] [Green Version]
- Wang, H.; Yung, M.M.H.; Ngan, H.Y.S.; Chan, K.K.L.; Chan, D.W. The Impact of the Tumor Microenvironment on Macrophage Polarization in Cancer Metastatic Progression. Int. J. Mol. Sci. 2021, 22, 6560. [Google Scholar] [CrossRef]
- Zuo, S.; Yu, J.; Pan, H.; Lu, L. Novel insights on targeting ferroptosis in cancer therapy. Biomark. Res. 2020, 8, 50. [Google Scholar] [CrossRef]
- Wu, J.; Minikes, A.M.; Gao, M.; Bian, H.; Li, Y.; Stockwell, B.R.; Chen, Z.N.; Jiang, X. Intercellular interaction dictates cancer cell ferroptosis via NF2-YAP signalling. Nature 2019, 572, 402–406. [Google Scholar] [CrossRef]
- Wang, H.; Lin, D.; Yu, Q.; Li, Z.; Lenahan, C.; Dong, Y.; Wei, Q.; Shao, A. A Promising Future of Ferroptosis in Tumor Therapy. Front. Cell. Dev. Biol. 2021, 9, 629150. [Google Scholar] [CrossRef]
- Sun, Y.; Xue, Z.; Huang, T.; Che, X.; Wu, G. Lipid metabolism in ferroptosis and ferroptosis-based cancer therapy. Front. Oncol. 2022, 12, 941618. [Google Scholar] [CrossRef]
- Song, X.; Zhu, S.; Chen, P.; Hou, W.; Wen, Q.; Liu, J.; Xie, Y.; Liu, J.; Klionsky, D.J.; Kroemer, G.; et al. AMPK-Mediated BECN1 Phosphorylation Promotes Ferroptosis by Directly Blocking System Xc(-) Activity. Curr. Biol. 2018, 28, 2388–2399.e5. [Google Scholar] [CrossRef] [Green Version]
- Liang, C.; Feng, P.; Ku, B.; Dotan, I.; Canaani, D.; Oh, B.H.; Jung, J.U. Autophagic and tumour suppressor activity of a novel Beclin1-binding protein UVRAG. Nat. Cell. Biol. 2006, 8, 688–699. [Google Scholar] [CrossRef]
- Kusnick, J.; Bruneau, A.; Tacke, F.; Hammerich, L. Ferroptosis in Cancer Immunotherapy—Implications for Hepatocellular Carcinoma. Immuno 2022, 2, 185–217. [Google Scholar] [CrossRef]
- Lei, G.; Zhang, Y.; Koppula, P.; Liu, X.; Zhang, J.; Lin, S.H.; Ajani, J.A.; Xiao, Q.; Liao, Z.; Wang, H.; et al. The role of ferroptosis in ionizing radiation-induced cell death and tumor suppression. Cell Res. 2020, 30, 146–162. [Google Scholar] [CrossRef]
- Chekhun, V.F.; Lukyanova, N.Y.; Burlaka, C.A.; Bezdenezhnykh, N.A.; Shpyleva, S.I.; Tryndyak, V.P.; Beland, F.A.; Pogribny, I.P. Iron metabolism disturbances in the MCF-7 human breast cancer cells with acquired resistance to doxorubicin and cisplatin. Int. J. Oncol. 2013, 43, 1481–1486. [Google Scholar] [CrossRef] [Green Version]
- Wang, W.; Green, M.; Choi, J.E.; Gijon, M.; Kennedy, P.D.; Johnson, J.K.; Liao, P.; Lang, X.; Kryczek, I.; Sell, A.; et al. CD8(+) T cells regulate tumor ferroptosis during cancer immunotherapy. Nature 2019, 569, 270–274. [Google Scholar] [CrossRef]
- Chatterjee, A.; Rodger, E.J.; Eccles, M.R. Epigenetic drivers of tumourigenesis and cancer metastasis. Semin. Cancer Biol. 2018, 51, 149–159. [Google Scholar] [CrossRef]
- Reyes, H.D.; Devor, E.J.; Warrier, A.; Newtson, A.M.; Mattson, J.; Wagner, V.; Duncan, G.N.; Leslie, K.K.; Gonzalez-Bosquet, J. Differential DNA methylation in high-grade serous ovarian cancer (HGSOC) is associated with tumor behavior. Sci. Rep. 2019, 9, 17996. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Drayton, R.M.; Dudziec, E.; Peter, S.; Bertz, S.; Hartmann, A.; Bryant, H.E.; Catto, J.W. Reduced expression of miRNA-27a modulates cisplatin resistance in bladder cancer by targeting the cystine/glutamate exchanger SLC7A11. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2014, 20, 1990–2000. [Google Scholar] [CrossRef] [Green Version]
- Liu, X.X.; Li, X.J.; Zhang, B.; Liang, Y.J.; Zhou, C.X.; Cao, D.X.; He, M.; Chen, G.Q.; He, J.R.; Zhao, Q. MicroRNA-26b is underexpressed in human breast cancer and induces cell apoptosis by targeting SLC7A11. FEBS Lett. 2011, 585, 1363–1367. [Google Scholar] [CrossRef] [Green Version]
- Mao, C.; Wang, X.; Liu, Y.; Wang, M.; Yan, B.; Jiang, Y.; Shi, Y.; Shen, Y.; Liu, X.; Lai, W.; et al. A G3BP1-Interacting lncRNA Promotes Ferroptosis and Apoptosis in Cancer via Nuclear Sequestration of p53. Cancer Res. 2018, 78, 3484–3496. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yoshioka, Y.; Kosaka, N.; Ochiya, T.; Kato, T. Micromanaging Iron Homeostasis: Hypoxia-inducible micro-RNA-210 suppresses iron homeostasis-related proteins. J. Biol. Chem. 2012, 287, 34110–34119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Babu, K.R.; Muckenthaler, M.U. miR-20a regulates expression of the iron exporter ferroportin in lung cancer. J. Mol. Med. 2016, 94, 347–359. [Google Scholar] [CrossRef] [Green Version]
- Park, S.; Oh, J.; Kim, M.; Jin, E.J. Bromelain effectively suppresses Kras-mutant colorectal cancer by stimulating ferroptosis. Anim. Cells Syst. 2018, 22, 334–340. [Google Scholar] [CrossRef] [Green Version]
- Peng, Y.; Xiang, H.; Chen, C.; Zheng, R.; Chai, J.; Peng, J.; Jiang, S. MiR-224 impairs adipocyte early differentiation and regulates fatty acid metabolism. Int. J. Biochem. Cell Biol. 2013, 45, 1585–1593. [Google Scholar] [CrossRef]
- Cui, M.; Xiao, Z.; Sun, B.; Wang, Y.; Zheng, M.; Ye, L.; Zhang, X. Involvement of cholesterol in hepatitis B virus X protein-induced abnormal lipid metabolism of hepatoma cells via up-regulating miR-205-targeted ACSL4. Biochem. Biophys. Res. Commun. 2014, 445, 651–655. [Google Scholar] [CrossRef]
Combination Drugs | Target | Mechanism | References |
---|---|---|---|
Erastin & Cisplatin | System Xc- | Erastin inhibited system Xc- and enhanced the cytotoxic effects of Cisplatin to eradicate tumor cells. | [94] |
Erastin & Docetaxel | SLC7A11 | Combination of Erastin and Docetaxel significantly promoted cell apoptosis, and induced cell cycle arrest at G2/M in ovarian cancer cells with ABCB1 overexpression. | [95] |
Sotuletinib (BLZ945) & Docetaxel | CD8+ T cells | BLZ945 combined with Docetaxel increased the infiltration of CD8+ T cells in tumor tissues. | [96] |
SCD1 inhibitors & RSL3/Erastin | Lipid peroxidation ROS | Stearoyl-CoA desaturase 1 (SCD1) inhibitors reduced an endogenous membrane antioxidant, CoQ10, which significantly potentiated the antitumor effect of Erastin and RSL3 in ovarian cancer cells. | [97,98] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 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
Zhan, S.; Yung, M.M.H.; Siu, M.K.Y.; Jiao, P.; Ngan, H.Y.S.; Chan, D.W.; Chan, K.K.L. New Insights into Ferroptosis Initiating Therapies (FIT) by Targeting the Rewired Lipid Metabolism in Ovarian Cancer Peritoneal Metastases. Int. J. Mol. Sci. 2022, 23, 15263. https://doi.org/10.3390/ijms232315263
Zhan S, Yung MMH, Siu MKY, Jiao P, Ngan HYS, Chan DW, Chan KKL. New Insights into Ferroptosis Initiating Therapies (FIT) by Targeting the Rewired Lipid Metabolism in Ovarian Cancer Peritoneal Metastases. International Journal of Molecular Sciences. 2022; 23(23):15263. https://doi.org/10.3390/ijms232315263
Chicago/Turabian StyleZhan, Shijie, Mingo M. H. Yung, Michelle K. Y. Siu, Peili Jiao, Hextan Y. S. Ngan, David W. Chan, and Karen K. L. Chan. 2022. "New Insights into Ferroptosis Initiating Therapies (FIT) by Targeting the Rewired Lipid Metabolism in Ovarian Cancer Peritoneal Metastases" International Journal of Molecular Sciences 23, no. 23: 15263. https://doi.org/10.3390/ijms232315263
APA StyleZhan, S., Yung, M. M. H., Siu, M. K. Y., Jiao, P., Ngan, H. Y. S., Chan, D. W., & Chan, K. K. L. (2022). New Insights into Ferroptosis Initiating Therapies (FIT) by Targeting the Rewired Lipid Metabolism in Ovarian Cancer Peritoneal Metastases. International Journal of Molecular Sciences, 23(23), 15263. https://doi.org/10.3390/ijms232315263