Targeted Anti-Mitochondrial Therapy: The Future of Oncology
Abstract
:1. Introduction
2. Mitochondria’s Benefits for Cancer Cells
2.1. Surviving in the Harsh Tumor Microenvironment
2.1.1. Metabolic Switch to Glycolysis
2.1.2. Redox Homeostasis
2.1.3. Protective Cell-Cycle Arrest (Dormancy or Quiescence)
2.1.4. pH Homeostasis
2.1.5. Autophagy
2.1.6. Intercellular Mitochondria Trafficking
2.1.7. Angiogenesis
2.2. Immune Evasion
2.2.1. TME Acidification
2.2.2. Glucose Influx
2.2.3. Mitochondrial Hijacking
2.2.4. Recruitment of Myeloid-Derived Suppressor Cells (MDSCs) toward TME
2.2.5. Expression of Immune Checkpoints
2.2.6. Defective Antigen Presentation
2.2.7. Immunosuppressive Mediators
2.3. Cancer Progression
2.3.1. Genomic Instability
2.3.2. Quiescence Evasion
2.3.3. Metastasis
2.4. Resistance to Treatment
2.4.1. Chemotherapy
2.4.2. Radiotherapy
2.4.3. Immunotherapy
3. Discussion
3.1. An Energy Battle between Immune and Cancer Cells
3.2. Mitochondria Improve Treatment Resistance
3.3. Reactive Oxygen Species: The Main Weapon of Mitochondria
3.4. Cancer Stem Cells Can Be Defeated by Targeting Mitochondria
3.5. Future Directions
4. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Halvorson, H.M.; Wyatt, K.H.; Kuehn, K.A. Ecological significance of autotroph–heterotroph microbial interactions in freshwaters. Freshw. Biol. 2020, 65, 1183–1188. [Google Scholar] [CrossRef]
- Saha, T.; Dash, C.; Jayabalan, R.; Khiste, S.; Kulkarni, A.; Kurmi, K.; Mondal, J.; Majumder, P.K.; Bardia, A.; Jang, H.L. Intercellular nanotubes mediate mitochondrial trafficking between cancer and immune cells. Nat. Nanotechnol. 2021, 17, 98–106. [Google Scholar] [CrossRef] [PubMed]
- García-Heredia, J.M.; Carnero, A. Role of mitochondria in cancer stem cell resistance. Cells 2020, 9, 1693. [Google Scholar] [CrossRef]
- Wu, Y.T.; Wu, S.B.; Wei, Y.H. Metabolic reprogramming of human cells in response to oxidative stress: Implications in the pathophysiology and therapy of mitochondrial diseases. Curr. Pharm. Des. 2014, 20, 5510–5526. [Google Scholar] [CrossRef]
- Akbari, H.; Taghizadeh-Hesary, F.; Heike, Y.; Bahadori, M. Cell Energy: A New Hypothesis in Decoding Cancer Evolution. Arch. Iran. Med. 2019, 22, 733–735. [Google Scholar]
- Al Tameemi, W.; Dale, T.P.; Al-Jumaily, R.M.K.; Forsyth, N.R. Hypoxia-Modified Cancer Cell Metabolism. Front. Cell Dev. Biol. 2019, 7, 4. [Google Scholar] [CrossRef]
- Paredes, F.; Williams, H.C.; San Martin, A. Metabolic adaptation in hypoxia and cancer. Cancer Lett. 2021, 502, 133–142. [Google Scholar] [CrossRef]
- Lee, S.; Hallis, S.P.; Jung, K.A.; Ryu, D.; Kwak, M.K. Impairment of HIF-1α-mediated metabolic adaption by NRF2-silencing in breast cancer cells. Redox Biol. 2019, 24, 101210. [Google Scholar] [CrossRef]
- Ishida, T.; Nakao, S.; Ueyama, T.; Harada, Y.; Kawamura, T. Metabolic remodeling during somatic cell reprogramming to induced pluripotent stem cells: Involvement of hypoxia-inducible factor 1. Inflamm. Regen. 2020, 40, 8. [Google Scholar] [CrossRef]
- Baumeister, J.; Chatain, N.; Hubrich, A.; Maié, T.; Costa, I.G.; Denecke, B.; Han, L.; Küstermann, C.; Sontag, S.; Seré, K.; et al. Hypoxia-inducible factor 1 (HIF-1) is a new therapeutic target in JAK2V617F-positive myeloproliferative neoplasms. Leukemia 2020, 34, 1062–1074. [Google Scholar] [CrossRef]
- van Gisbergen, M.W.; Offermans, K.; Voets, A.M.; Lieuwes, N.G.; Biemans, R.; Hoffmann, R.F.; Dubois, L.J.; Lambin, P. Mitochondrial Dysfunction Inhibits Hypoxia-Induced HIF-1α Stabilization and Expression of Its Downstream Targets. Front. Oncol. 2020, 10, 770. [Google Scholar] [CrossRef]
- Tormos, K.V.; Chandel, N.S. Inter-connection between mitochondria and HIFs. J. Cell. Mol. Med. 2010, 14, 795–804. [Google Scholar] [CrossRef]
- Vaupel, P.; Multhoff, G. Revisiting the Warburg effect: Historical dogma versus current understanding. J. Physiol. 2021, 599, 1745–1757. [Google Scholar] [CrossRef] [PubMed]
- Ramos-Ibeas, P.; Barandalla, M.; Colleoni, S.; Lazzari, G. Pyruvate antioxidant roles in human fibroblasts and embryonic stem cells. Mol. Cell. Biochem. 2017, 429, 137–150. [Google Scholar] [CrossRef]
- Chang, C.-W.; Lo, J.-F.; Wang, X.W. Roles of mitochondria in liver cancer stem cells. Differentiation 2019, 107, 35–41. [Google Scholar] [CrossRef] [PubMed]
- Hur, W.; Ryu, J.Y.; Kim, H.U.; Hong, S.W.; Lee, E.B.; Lee, S.Y.; Yoon, S.K. Systems approach to characterize the metabolism of liver cancer stem cells expressing CD133. Sci. Rep. 2017, 7, 45557. [Google Scholar] [CrossRef]
- Song, K.; Kwon, H.; Han, C.; Zhang, J.; Dash, S.; Lim, K.; Wu, T. Active glycolytic metabolism in CD133(+) hepatocellular cancer stem cells: Regulation by MIR-122. Oncotarget 2015, 6, 40822–40835. [Google Scholar] [CrossRef]
- Li, P.; Zhang, D.; Shen, L.; Dong, K.; Wu, M.; Ou, Z.; Shi, D. Redox homeostasis protects mitochondria through accelerating ROS conversion to enhance hypoxia resistance in cancer cells. Sci. Rep. 2016, 6, 22831. [Google Scholar] [CrossRef]
- Mitsuishi, Y.; Taguchi, K.; Kawatani, Y.; Shibata, T.; Nukiwa, T.; Aburatani, H.; Yamamoto, M.; Motohashi, H. Nrf2 redirects glucose and glutamine into anabolic pathways in metabolic reprogramming. Cancer Cell 2012, 22, 66–79. [Google Scholar] [CrossRef]
- Wang, X.; Perez, E.; Liu, R.; Yan, L.J.; Mallet, R.T.; Yang, S.H. Pyruvate protects mitochondria from oxidative stress in human neuroblastoma SK-N-SH cells. Brain Res. 2007, 1132, 1–9. [Google Scholar] [CrossRef]
- Tauffenberger, A.; Fiumelli, H.; Almustafa, S.; Magistretti, P.J. Lactate and pyruvate promote oxidative stress resistance through hormetic ROS signaling. Cell Death Dis. 2019, 10, 653. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jitschin, R.; Hofmann, A.D.; Bruns, H.; Gießl, A.; Bricks, J.; Berger, J.; Saul, D.; Eckart, M.J.; Mackensen, A.; Mougiakakos, D. Mitochondrial metabolism contributes to oxidative stress and reveals therapeutic targets in chronic lymphocytic leukemia. Blood 2014, 123, 2663–2672. [Google Scholar] [CrossRef]
- Haraguchi, N.; Ishii, H.; Mimori, K.; Tanaka, F.; Ohkuma, M.; Kim, H.M.; Akita, H.; Takiuchi, D.; Hatano, H.; Nagano, H. CD13 is a therapeutic target in human liver cancer stem cells. J. Clin. Investig. 2010, 120, 3326–3339. [Google Scholar] [CrossRef] [PubMed]
- Kim, H.M.; Haraguchi, N.; Ishii, H.; Ohkuma, M.; Okano, M.; Mimori, K.; Eguchi, H.; Yamamoto, H.; Nagano, H.; Sekimoto, M. Increased CD13 expression reduces reactive oxygen species, promoting survival of liver cancer stem cells via an epithelial–mesenchymal transition-like phenomenon. Ann. Surg. Oncol. 2012, 19, 539–548. [Google Scholar] [CrossRef] [PubMed]
- Thanee, M.; Loilome, W.; Techasen, A.; Sugihara, E.; Okazaki, S.; Abe, S.; Ueda, S.; Masuko, T.; Namwat, N.; Khuntikeo, N. CD44 variant-dependent redox status regulation in liver fluke-associated cholangiocarcinoma: A target for cholangiocarcinoma treatment. Cancer Sci. 2016, 107, 991–1000. [Google Scholar] [CrossRef] [PubMed]
- Wicks, E.E.; Semenza, G.L. Hypoxia-inducible factors: Cancer progression and clinical translation. J. Clin. Investig. 2022, 132, e159839. [Google Scholar] [CrossRef]
- Druker, J.; Wilson, J.W.; Child, F.; Shakir, D.; Fasanya, T.; Rocha, S. Role of Hypoxia in the Control of the Cell Cycle. Int. J. Mol. Sci. 2021, 22, 4874. [Google Scholar] [CrossRef]
- Marescal, O.; Cheeseman, I.M. Cellular Mechanisms and Regulation of Quiescence. Dev. Cell 2020, 55, 259–271. [Google Scholar] [CrossRef]
- Koshiji, M.; Kageyama, Y.; Pete, E.A.; Horikawa, I.; Barrett, J.C.; Huang, L.E. HIF-1α induces cell cycle arrest by functionally counteracting Myc. EMBO J. 2004, 23, 1949–1956. [Google Scholar] [CrossRef]
- Yu-Lee, L.-Y.; Lee, Y.-C.; Pan, J.; Lin, S.-C.; Pan, T.; Yu, G.; Hawke, D.H.; Pan, B.-F.; Lin, S.-H. Bone secreted factors induce cellular quiescence in prostate cancer cells. Sci. Rep. 2019, 9, 18635. [Google Scholar] [CrossRef]
- Mandal, J.P.; Shiue, C.-N.; Chen, Y.-C.; Lee, M.-C.; Yang, H.-H.; Chang, H.-H.; Hu, C.-T.; Liao, P.-C.; Hui, L.-C.; You, R.-I.; et al. PKCδ mediates mitochondrial ROS generation and oxidation of HSP60 to relieve RKIP inhibition on MAPK pathway for HCC progression. Free Radic. Biol. Med. 2021, 163, 69–87. [Google Scholar] [CrossRef] [PubMed]
- O’Reilly, E.; Zeinabad, H.A.; Szegezdi, E. Hematopoietic versus leukemic stem cell quiescence: Challenges and therapeutic opportunities. Blood Rev. 2021, 50, 100850. [Google Scholar] [CrossRef] [PubMed]
- Shiau, J.P.; Chuang, Y.T.; Cheng, Y.B.; Tang, J.Y.; Hou, M.F.; Yen, C.Y.; Chang, H.W. Impacts of Oxidative Stress and PI3K/AKT/mTOR on Metabolism and the Future Direction of Investigating Fucoidan-Modulated Metabolism. Antioxidants 2022, 11, 911. [Google Scholar] [CrossRef] [PubMed]
- Dunn, J.D.; Alvarez, L.A.; Zhang, X.; Soldati, T. Reactive oxygen species and mitochondria: A nexus of cellular homeostasis. Redox Biol. 2015, 6, 472–485. [Google Scholar] [CrossRef] [PubMed]
- Carcereri de Prati, A.; Butturini, E.; Rigo, A.; Oppici, E.; Rossin, M.; Boriero, D.; Mariotto, S. Metastatic breast cancer cells enter into dormant state and express cancer stem cells phenotype under chronic hypoxia. J. Cell. Biochem. 2017, 118, 3237–3248. [Google Scholar] [CrossRef]
- Czowski, B.J.; Romero-Moreno, R.; Trull, K.J.; White, K.A. Cancer and pH Dynamics: Transcriptional Regulation, Proteostasis, and the Need for New Molecular Tools. Cancers 2020, 12, 2760. [Google Scholar] [CrossRef]
- White, K.A.; Grillo-Hill, B.K.; Barber, D.L. Cancer cell behaviors mediated by dysregulated pH dynamics at a glance. J. Cell Sci. 2017, 130, 663–669. [Google Scholar] [CrossRef]
- Becker, H.M. Carbonic anhydrase IX and acid transport in cancer. Br. J. Cancer 2020, 122, 157–167. [Google Scholar] [CrossRef]
- Supuran, C.T.; Scozzafava, A. Carbonic anhydrase inhibitors: Aromatic sulfonamides and disulfonamides act as efficient tumor growth inhibitors. J. Enzym. Inhib. 2000, 15, 597–610. [Google Scholar] [CrossRef]
- Ding, W.X.; Ni, H.M.; Li, M.; Liao, Y.; Chen, X.; Stolz, D.B.; Dorn, G.W., 2nd; Yin, X.M. Nix is critical to two distinct phases of mitophagy, reactive oxygen species-mediated autophagy induction and Parkin-ubiquitin-p62-mediated mitochondrial priming. J. Biol. Chem. 2010, 285, 27879–27890. [Google Scholar] [CrossRef]
- Towers, C.G.; Fitzwalter, B.E.; Regan, D.; Goodspeed, A.; Morgan, M.J.; Liu, C.W.; Gustafson, D.L.; Thorburn, A. Cancer Cells Upregulate NRF2 Signaling to Adapt to Autophagy Inhibition. Dev. Cell 2019, 50, 690–703.e696. [Google Scholar] [CrossRef] [PubMed]
- Kasai, S.; Shimizu, S.; Tatara, Y.; Mimura, J.; Itoh, K. Regulation of Nrf2 by Mitochondrial Reactive Oxygen Species in Physiology and Pathology. Biomolecules 2020, 10, 320. [Google Scholar] [CrossRef] [PubMed]
- Rothe, K.; Porter, V.; Jiang, X. Current Outlook on Autophagy in Human Leukemia: Foe in Cancer Stem Cells and Drug Resistance, Friend in New Therapeutic Interventions. Int. J. Mol. Sci. 2019, 20, 461. [Google Scholar] [CrossRef] [PubMed]
- Nazio, F.; Bordi, M.; Cianfanelli, V.; Locatelli, F.; Cecconi, F. Autophagy and cancer stem cells: Molecular mechanisms and therapeutic applications. Cell Death Differ. 2019, 26, 690–702. [Google Scholar] [CrossRef]
- Lambeth, D.O. What is the function of GTP produced in the Krebs citric acid cycle? IUBMB Life 2002, 54, 143–144. [Google Scholar] [CrossRef]
- Akbari, H.; Taghizadeh-Hesary, F.; Bahadori, M. Mitochondria determine response to anti-programmed cell death protein-1 (anti-PD-1) immunotherapy: An evidence-based hypothesis. Mitochondrion 2022, 62, 151–158. [Google Scholar] [CrossRef]
- Lu, J.; Zheng, X.; Li, F.; Yu, Y.; Chen, Z.; Liu, Z.; Wang, Z.; Xu, H.; Yang, W. Tunneling nanotubes promote intercellular mitochondria transfer followed by increased invasiveness in bladder cancer cells. Oncotarget 2017, 8, 15539–15552. [Google Scholar] [CrossRef]
- Zhu, H.; Zhang, S. Hypoxia inducible factor-1α/vascular endothelial growth factor signaling activation correlates with response to radiotherapy and its inhibition reduces hypoxia-induced angiogenesis in lung cancer. J. Cell. Biochem. 2018, 119, 7707–7718. [Google Scholar] [CrossRef]
- Sun, X.; Vale, M.; Jiang, X.; Gupta, R.; Krissansen, G.W. Antisense HIF-1α prevents acquired tumor resistance to angiostatin gene therapy. Cancer Gene Ther. 2010, 17, 532–540. [Google Scholar] [CrossRef]
- Zhang, Q.; Han, Z.; Zhu, Y.; Chen, J.; Li, W. Role of hypoxia inducible factor-1 in cancer stem cells (Review). Mol. Med. Rep. 2021, 23, 17. [Google Scholar] [CrossRef]
- Rohani, N.; Hao, L.; Alexis, M.S.; Joughin, B.A.; Krismer, K.; Moufarrej, M.N.; Soltis, A.R.; Lauffenburger, D.A.; Yaffe, M.B.; Burge, C.B.; et al. Acidification of Tumor at Stromal Boundaries Drives Transcriptome Alterations Associated with Aggressive Phenotypes. Cancer Res. 2019, 79, 1952–1966. [Google Scholar] [CrossRef] [Green Version]
- Huber, V.; Camisaschi, C.; Berzi, A.; Ferro, S.; Lugini, L.; Triulzi, T.; Tuccitto, A.; Tagliabue, E.; Castelli, C.; Rivoltini, L. Cancer acidity: An ultimate frontier of tumor immune escape and a novel target of immunomodulation. Semin. Cancer Biol. 2017, 43, 74–89. [Google Scholar] [CrossRef]
- Bohloli, M.; Atashi, A.; Soleimani, M.; Kaviani, S.; Anbarlou, A. Investigating Effects of Acidic pH on Proliferation, Invasion and Drug-Induced Apoptosis in Lymphoblastic Leukemia. Cancer Microenviron. 2016, 9, 119–126. [Google Scholar] [CrossRef] [PubMed]
- Klein, K.; He, K.; Younes, A.I.; Barsoumian, H.B.; Chen, D.; Ozgen, T.; Mosaffa, S.; Patel, R.R.; Gu, M.; Novaes, J. Role of mitochondria in cancer immune evasion and potential therapeutic approaches. Front. Immunol. 2020, 11, 573326. [Google Scholar] [CrossRef] [PubMed]
- Yu, X.-J.; Song, J.-C.; Du, J.; Shi, Y.-Q.; Liu, Y.-X.; Shen, Y. GLUT-1 and its regulating factor HIF-1α expression in epithelial ovarian tumors: GLUT-1 is associated with molecular typing and grade of epithelial ovarian cancer. Int. J. Clin. Exp. Pathol. 2017, 10, 4479–4487. [Google Scholar]
- Heydarzadeh, S.; Moshtaghie, A.A.; Daneshpoor, M.; Hedayati, M. Regulators of glucose uptake in thyroid cancer cell lines. Cell Commun. Signal. 2020, 18, 83. [Google Scholar] [CrossRef]
- Herbel, C.; Patsoukis, N.; Bardhan, K.; Seth, P.; Weaver, J.D.; Boussiotis, V.A. Clinical significance of T cell metabolic reprogramming in cancer. Clin. Transl. Med. 2016, 5, 29. [Google Scholar] [CrossRef]
- Yang, Y.; Li, C.; Liu, T.; Dai, X.; Bazhin, A.V. Myeloid-Derived Suppressor Cells in Tumors: From Mechanisms to Antigen Specificity and Microenvironmental Regulation. Front. Immunol. 2020, 11, 1371. [Google Scholar] [CrossRef]
- Chiu, D.K.C.; Xu, I.M.J.; Lai, R.K.H.; Tse, A.P.W.; Wei, L.L.; Koh, H.Y.; Li, L.L.; Lee, D.; Lo, R.C.L.; Wong, C.M. Hypoxia induces myeloid-derived suppressor cell recruitment to hepatocellular carcinoma through chemokine (C-C motif) ligand 26. Hepatology 2016, 64, 797–813. [Google Scholar] [CrossRef]
- Watson, M.J.; Vignali, P.D.A.; Mullett, S.J.; Overacre-Delgoffe, A.E.; Peralta, R.M.; Grebinoski, S.; Menk, A.V.; Rittenhouse, N.L.; DePeaux, K.; Whetstone, R.D.; et al. Metabolic support of tumour-infiltrating regulatory T cells by lactic acid. Nature 2021, 591, 645–651. [Google Scholar] [CrossRef]
- Cheng, A.N.; Cheng, L.-C.; Kuo, C.-L.; Lo, Y.K.; Chou, H.-Y.; Chen, C.-H.; Wang, Y.-H.; Chuang, T.-H.; Cheng, S.-J.; Lee, A.Y.-L. Mitochondrial Lon-induced mtDNA leakage contributes to PD-L1–mediated immunoescape via STING-IFN signaling and extracellular vesicles. J. Immunother. Cancer 2020, 8, e001372. [Google Scholar] [CrossRef] [PubMed]
- Noman, M.Z.; Desantis, G.; Janji, B.; Hasmim, M.; Karray, S.; Dessen, P.; Bronte, V.; Chouaib, S. PD-L1 is a novel direct target of HIF-1α, and its blockade under hypoxia enhanced MDSC-mediated T cell activation. J. Exp. Med. 2014, 211, 781–790. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ohl, K.; Tenbrock, K. Reactive oxygen species as regulators of MDSC-mediated immune suppression. Front. Immunol. 2018, 9, 2499. [Google Scholar] [CrossRef] [PubMed]
- Voron, T.; Colussi, O.; Marcheteau, E.; Pernot, S.; Nizard, M.; Pointet, A.-L.; Latreche, S.; Bergaya, S.; Benhamouda, N.; Tanchot, C. VEGF-A modulates expression of inhibitory checkpoints on CD8+ T cells in tumors. J. Exp. Med. 2015, 212, 139–148. [Google Scholar] [CrossRef] [PubMed]
- You, L.; Wu, W.; Wang, X.; Fang, L.; Adam, V.; Nepovimova, E.; Wu, Q.; Kuca, K. The role of hypoxia-inducible factor 1 in tumor immune evasion. Med. Res. Rev. 2021, 41, 1622–1643. [Google Scholar] [CrossRef]
- Cornel, A.M.; Mimpen, I.L.; Nierkens, S. MHC Class I Downregulation in Cancer: Underlying Mechanisms and Potential Targets for Cancer Immunotherapy. Cancers 2020, 12, 1760. [Google Scholar] [CrossRef]
- Taylor, B.C.; Balko, J.M. Mechanisms of MHC-I Downregulation and Role in Immunotherapy Response. Front. Immunol. 2022, 13, 844866. [Google Scholar] [CrossRef]
- Sethumadhavan, S.; Silva, M.; Philbrook, P.; Nguyen, T.; Hatfield, S.M.; Ohta, A.; Sitkovsky, M.V. Hypoxia and hypoxia-inducible factor (HIF) downregulate antigen-presenting MHC class I molecules limiting tumor cell recognition by T cells. PLoS ONE 2017, 12, e0187314. [Google Scholar] [CrossRef]
- Brea, E.J.; Oh, C.Y.; Manchado, E.; Budhu, S.; Gejman, R.S.; Mo, G.; Mondello, P.; Han, J.E.; Jarvis, C.A.; Ulmert, D.; et al. Kinase Regulation of Human MHC Class I Molecule Expression on Cancer Cells. Cancer Immunol. Res. 2016, 4, 936–947. [Google Scholar] [CrossRef]
- Franklin, D.A.; James, J.L.; Axelrod, M.L.; Balko, J.M. MEK inhibition activates STAT signaling to increase breast cancer immunogenicity via MHC-I expression. Cancer Drug Resist. 2020, 3, 603–612. [Google Scholar] [CrossRef]
- Sun, P.; Meng, L.H. Emerging roles of class I PI3K inhibitors in modulating tumor microenvironment and immunity. Acta Pharm. Sin. 2020, 41, 1395–1402. [Google Scholar] [CrossRef] [PubMed]
- Osipov, A.; Blair, A.B.; Liberto, J.; Wang, J.; Li, K.; Herbst, B.; Xu, Y.; Li, S.; Niu, N.; Rashid, R.; et al. Inhibition of focal adhesion kinase enhances antitumor response of radiation therapy in pancreatic cancer through CD8+ T cells. Cancer Biol. Med. 2021, 18, 206–214. [Google Scholar] [CrossRef]
- Liu, Y.T.; Sun, Z.J. Turning cold tumors into hot tumors by improving T-cell infiltration. Theranostics 2021, 11, 5365–5386. [Google Scholar] [CrossRef] [PubMed]
- Wang, H.; Jiang, H.; Van De Gucht, M.; De Ridder, M. Hypoxic radioresistance: Can ROS be the key to overcome it? Cancers 2019, 11, 112. [Google Scholar] [CrossRef] [PubMed]
- Aggarwal, V.; Tuli, H.S.; Varol, A.; Thakral, F.; Yerer, M.B.; Sak, K.; Varol, M.; Jain, A.; Khan, M.; Sethi, G. Role of reactive oxygen species in cancer progression: Molecular mechanisms and recent advancements. Biomolecules 2019, 9, 735. [Google Scholar] [CrossRef]
- Bonora, M.; Missiroli, S.; Perrone, M.; Fiorica, F.; Pinton, P.; Giorgi, C. Mitochondrial control of genomic instability in cancer. Cancers 2021, 13, 1914. [Google Scholar] [CrossRef]
- Kalkavan, H.; Green, D.R. MOMP, cell suicide as a BCL-2 family business. Cell Death Differ. 2018, 25, 46–55. [Google Scholar] [CrossRef] [PubMed]
- Xu, Y.; Surman, D.R.; Diggs, L.; Xi, S.; Gao, S.; Gurusamy, D.; McLoughlin, K.; Drake, J.; Feingold, P.; Brown, K.; et al. Bile acid-induced “Minority MOMP” promotes esophageal carcinogenesis while maintaining apoptotic resistance via Mcl-1. Oncogene 2020, 39, 877–890. [Google Scholar] [CrossRef]
- Wu, F.; Tong, D.D.; Ni, L.; Wang, L.M.; Wang, M.C. HIF-1α suppresses myeloma progression by targeting Mcl-1. Int. J. Clin. Exp. Pathol. 2020, 13, 1483–1491. [Google Scholar]
- Pećina-Šlaus, N.; Kafka, A.; Salamon, I.; Bukovac, A. Mismatch Repair Pathway, Genome Stability and Cancer. Front. Mol. Biosci. 2020, 7, 122. [Google Scholar] [CrossRef]
- Koshiji, M.; To, K.K.-W.; Hammer, S.; Kumamoto, K.; Harris, A.L.; Modrich, P.; Huang, L.E. HIF-1α induces genetic instability by transcriptionally downregulating MutSα expression. Mol. Cell 2005, 17, 793–803. [Google Scholar] [CrossRef] [PubMed]
- Huang, J.X.; Yan, W.; Song, Z.X.; Qian, R.Y.; Chen, P.; Salminen, E.; Toppari, J. Relationship between proliferative activity of cancer cells and clinicopathological factors in patients with esophageal squamous cell carcinoma. World J. Gastroenterol. 2005, 11, 2956–2959. [Google Scholar] [CrossRef] [PubMed]
- Ganguly, K.K.; Pal, S.; Moulik, S.; Chatterjee, A. Integrins and metastasis. Cell Adhes. Migr. 2013, 7, 251–261. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fiore, A.P.Z.P.; Ribeiro, P.d.F.; Bruni-Cardoso, A. Sleeping Beauty and the Microenvironment Enchantment: Microenvironmental Regulation of the Proliferation-Quiescence Decision in Normal Tissues and in Cancer Development. Front. Cell Dev. Biol. 2018, 6, 59. [Google Scholar] [CrossRef]
- Nunes, J.B.; Peixoto, J.; Soares, P.; Maximo, V.; Carvalho, S.; Pinho, S.S.; Vieira, A.F.; Paredes, J.; Rego, A.C.; Ferreira, I.L.; et al. OXPHOS dysfunction regulates integrin-β1 modifications and enhances cell motility and migration. Hum. Mol. Genet. 2014, 24, 1977–1990. [Google Scholar] [CrossRef]
- Kadonosono, T.; Miyamoto, K.; Sakai, S.; Matsuo, Y.; Kitajima, S.; Wang, Q.; Endo, M.; Niibori, M.; Kuchimaru, T.; Soga, T.; et al. AGE/RAGE axis regulates reversible transition to quiescent states of ALK-rearranged NSCLC and pancreatic cancer cells in monolayer cultures. Sci. Rep. 2022, 12, 9886. [Google Scholar] [CrossRef]
- Serasinghe, M.N.; Wieder, S.Y.; Renault, T.T.; Elkholi, R.; Asciolla, J.J.; Yao, J.L.; Jabado, O.; Hoehn, K.; Kageyama, Y.; Sesaki, H.; et al. Mitochondrial division is requisite to RAS-induced transformation and targeted by oncogenic MAPK pathway inhibitors. Mol. Cell 2015, 57, 521–536. [Google Scholar] [CrossRef]
- Zhou, D.; Shao, L.; Spitz, D.R. Reactive oxygen species in normal and tumor stem cells. Adv. Cancer Res. 2014, 122, 1–67. [Google Scholar] [CrossRef]
- Mittal, V. Epithelial mesenchymal transition in tumor metastasis. Annu. Rev. Pathol. Mech. Dis. 2018, 13, 395–412. [Google Scholar] [CrossRef]
- Fazilaty, H.; Gardaneh, M.; Akbari, P.; Zekri, A.; Behnam, B. SLUG and SOX9 cooperatively regulate tumor initiating niche factors in breast cancer. Cancer Microenviron. 2016, 9, 71–74. [Google Scholar] [CrossRef]
- Fazilaty, H.; Gardaneh, M.; Bahrami, T.; Salmaninejad, A.; Behnam, B. Crosstalk between breast cancer stem cells and metastatic niche: Emerging molecular metastasis pathway? Tumor Biol. 2013, 34, 2019–2030. [Google Scholar] [CrossRef] [PubMed]
- Fazilaty, H.; Behnam, B. The perivascular niche governs an autoregulatory network to support breast cancer metastasis. Cell Biol. Int. 2014, 38, 691–694. [Google Scholar] [CrossRef] [PubMed]
- Kim, M.; Jang, K.; Miller, P.; Picon-Ruiz, M.; Yeasky, T.M.; El-Ashry, D.; Slingerland, J.M. VEGFA links self-renewal and metastasis by inducing Sox2 to repress miR-452, driving Slug. Oncogene 2017, 36, 5199–5211. [Google Scholar] [CrossRef]
- Erler, J.T.; Bennewith, K.L.; Nicolau, M.; Dornhöfer, N.; Kong, C.; Le, Q.-T.; Chi, J.-T.A.; Jeffrey, S.S.; Giaccia, A.J. Lysyl oxidase is essential for hypoxia-induced metastasis. Nature 2006, 440, 1222–1226. [Google Scholar] [CrossRef] [PubMed]
- Amendola, P.G.; Reuten, R.; Erler, J.T. Interplay Between LOX Enzymes and Integrins in the Tumor Microenvironment. Cancers 2019, 11, 729. [Google Scholar] [CrossRef] [PubMed]
- Yang, H.; Villani, R.M.; Wang, H.; Simpson, M.J.; Roberts, M.S.; Tang, M.; Liang, X. The role of cellular reactive oxygen species in cancer chemotherapy. J. Exp. Clin. Cancer Res. 2018, 37, 266. [Google Scholar] [CrossRef] [PubMed]
- Ji, L.; Li, H.; Gao, P.; Shang, G.; Zhang, D.D.; Zhang, N.; Jiang, T. Nrf2 pathway regulates multidrug-resistance-associated protein 1 in small cell lung cancer. PLoS ONE 2013, 8, e63404. [Google Scholar] [CrossRef]
- Perillo, B.; Di Donato, M.; Pezone, A.; Di Zazzo, E.; Giovannelli, P.; Galasso, G.; Castoria, G.; Migliaccio, A. ROS in cancer therapy: The bright side of the moon. Exp. Mol. Med. 2020, 52, 192–203. [Google Scholar] [CrossRef]
- Dornfeld, K.; Bjork, J.; Folkert, G.; Skildum, A.; Wallace, K.B. Mitochondrial activities play a pivotal role in regulating cell cycle in response to doxorubicin. Cell Cycle 2021, 20, 1067–1079. [Google Scholar] [CrossRef]
- Desouky, O.; Ding, N.; Zhou, G. Targeted and non-targeted effects of ionizing radiation. J. Radiat. Res. Appl. Sci. 2015, 8, 247–254. [Google Scholar] [CrossRef]
- Lee, J.J.; van de Ven, R.A.H.; Zaganjor, E.; Ng, M.R.; Barakat, A.; Demmers, J.; Finley, L.W.S.; Gonzalez Herrera, K.N.; Hung, Y.P.; Harris, I.S.; et al. Inhibition of epithelial cell migration and Src/FAK signaling by SIRT3. Proc. Natl. Acad. Sci. USA 2018, 115, 7057–7062. [Google Scholar] [CrossRef] [PubMed]
- Desdín-Micó, G.; Soto-Heredero, G.; Mittelbrunn, M. Mitochondrial activity in T cells. Mitochondrion 2018, 41, 51–57. [Google Scholar] [CrossRef] [PubMed]
- Surace, L.; Doisne, J.-M.; Escoll, P.; Marie, S.; Dardalhon, V.; Croft, C.; Thaller, A.; Topazio, D.; Sparaneo, A.; Cama, A. Polarized mitochondria as guardians of NK cell fitness. Blood Adv. 2021, 5, 26–38. [Google Scholar] [CrossRef] [PubMed]
- Memme, J.M.; Erlich, A.T.; Phukan, G.; Hood, D.A. Exercise and mitochondrial health. J. Physiol. 2021, 599, 803–817. [Google Scholar] [CrossRef]
- Luoma, R.L.; Butler, M.W.; Stahlschmidt, Z.R. Plasticity of immunity in response to eating. J. Exp. Biol. 2016, 219, 1965–1968. [Google Scholar] [CrossRef]
- Rodrigues, N.R.; Macedo, G.E.; Martins, I.K.; Gomes, K.K.; de Carvalho, N.R.; Posser, T.; Franco, J.L. Short-term sleep deprivation with exposure to nocturnal light alters mitochondrial bioenergetics in Drosophila. Free Radic. Biol. Med. 2018, 120, 395–406. [Google Scholar] [CrossRef]
- de Mello, A.H.; Costa, A.B.; Engel, J.D.G.; Rezin, G.T. Mitochondrial dysfunction in obesity. Life Sci. 2018, 192, 26–32. [Google Scholar] [CrossRef]
- Malińska, D.; Więckowski, M.R.; Michalska, B.; Drabik, K.; Prill, M.; Patalas-Krawczyk, P.; Walczak, J.; Szymański, J.; Mathis, C.; Van der Toorn, M. Mitochondria as a possible target for nicotine action. J. Bioenerg. Biomembr. 2019, 51, 259–276. [Google Scholar] [CrossRef]
- Chamoto, K.; Chowdhury, P.S.; Kumar, A.; Sonomura, K.; Matsuda, F.; Fagarasan, S.; Honjo, T. Mitochondrial activation chemicals synergize with surface receptor PD-1 blockade for T cell-dependent antitumor activity. Proc. Natl. Acad. Sci. USA 2017, 114, E761–E770. [Google Scholar] [CrossRef]
- van der Merwe, M.; van Niekerk, G.; Fourie, C.; du Plessis, M.; Engelbrecht, A.-M. The impact of mitochondria on cancer treatment resistance. Cell. Oncol. 2021, 44, 983–995. [Google Scholar] [CrossRef]
- Sabnis, A.J.; Bivona, T.G. Principles of Resistance to Targeted Cancer Therapy: Lessons from Basic and Translational Cancer Biology. Trends Mol. Med. 2019, 25, 185–197. [Google Scholar] [CrossRef] [PubMed]
- Shah, N.P.; Tran, C.; Lee, F.Y.; Chen, P.; Norris, D.; Sawyers, C.L. Overriding imatinib resistance with a novel ABL kinase inhibitor. Science 2004, 305, 399–401. [Google Scholar] [CrossRef] [PubMed]
- Nazarian, R.; Shi, H.; Wang, Q.; Kong, X.; Koya, R.C.; Lee, H.; Chen, Z.; Lee, M.-K.; Attar, N.; Sazegar, H. Melanomas acquire resistance to B-RAF (V600E) inhibition by RTK or N-RAS upregulation. Nature 2010, 468, 973–977. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sequist, L.V.; Waltman, B.A.; Dias-Santagata, D.; Digumarthy, S.; Turke, A.B.; Fidias, P.; Bergethon, K.; Shaw, A.T.; Gettinger, S.; Cosper, A.K.; et al. Genotypic and histological evolution of lung cancers acquiring resistance to EGFR inhibitors. Sci. Transl. Med. 2011, 3, 75ra26. [Google Scholar] [CrossRef]
- Mutch, D.G.; Orlando, M.; Goss, T.; Teneriello, M.G.; Gordon, A.N.; McMeekin, S.D.; Wang, Y.; Scribner, D.R., Jr.; Marciniack, M.; Naumann, R.W.; et al. Randomized phase III trial of gemcitabine compared with pegylated liposomal doxorubicin in patients with platinum-resistant ovarian cancer. J. Clin. Oncol. 2007, 25, 2811–2818. [Google Scholar] [CrossRef]
- Kudoh, K.; Takano, M.; Kouta, H.; Kikuchi, R.; Kita, T.; Miyamoto, M.; Watanabe, A.; Kato, M.; Goto, T.; Kikuchi, Y. Effects of bevacizumab and pegylated liposomal doxorubicin for the patients with recurrent or refractory ovarian cancers. Gynecol. Oncol. 2011, 122, 233–237. [Google Scholar] [CrossRef]
- Michels, J.; Ghiringhelli, F.; Frenel, J.-S.; Brard, C.; You, B.; Floquet, A.; Eberst, L.; Bahleda, R.; Genestie, C.; Balleyguier, C.; et al. Pembrolizumab in combination with bevacizumab and pegylated liposomal doxorubicin in patients with platinum-resistant epithelial ovarian cancer. J. Clin. Oncol. 2021, 39, 5522. [Google Scholar] [CrossRef]
- Liao, Z.; Chua, D.; Tan, N.S. Reactive oxygen species: A volatile driver of field cancerization and metastasis. Mol. Cancer 2019, 18, 65. [Google Scholar] [CrossRef]
- Reczek, C.R.; Chandel, N.S. The Two Faces of Reactive Oxygen Species in Cancer. Annu. Rev. Cancer Biol. 2017, 1, 79–98. [Google Scholar] [CrossRef]
- Ježek, J.; Cooper, K.F.; Strich, R. Reactive Oxygen Species and Mitochondrial Dynamics: The Yin and Yang of Mitochondrial Dysfunction and Cancer Progression. Antioxidants 2018, 7, 13. [Google Scholar] [CrossRef]
- Iranmanesh, Y.; Jiang, B.; Favour, O.C.; Dou, Z.; Wu, J.; Li, J.; Sun, C. Mitochondria’s role in the maintenance of cancer stem cells in glioblastoma. Front. Oncol. 2021, 11, 582694. [Google Scholar] [CrossRef] [PubMed]
- Sighel, D.; Notarangelo, M.; Aibara, S.; Re, A.; Ricci, G.; Guida, M.; Soldano, A.; Adami, V.; Ambrosini, C.; Broso, F. Inhibition of mitochondrial translation suppresses glioblastoma stem cell growth. Cell Rep. 2021, 35, 109024. [Google Scholar] [CrossRef] [PubMed]
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
Taghizadeh-Hesary, F.; Akbari, H.; Bahadori, M.; Behnam, B. Targeted Anti-Mitochondrial Therapy: The Future of Oncology. Genes 2022, 13, 1728. https://doi.org/10.3390/genes13101728
Taghizadeh-Hesary F, Akbari H, Bahadori M, Behnam B. Targeted Anti-Mitochondrial Therapy: The Future of Oncology. Genes. 2022; 13(10):1728. https://doi.org/10.3390/genes13101728
Chicago/Turabian StyleTaghizadeh-Hesary, Farzad, Hassan Akbari, Moslem Bahadori, and Babak Behnam. 2022. "Targeted Anti-Mitochondrial Therapy: The Future of Oncology" Genes 13, no. 10: 1728. https://doi.org/10.3390/genes13101728
APA StyleTaghizadeh-Hesary, F., Akbari, H., Bahadori, M., & Behnam, B. (2022). Targeted Anti-Mitochondrial Therapy: The Future of Oncology. Genes, 13(10), 1728. https://doi.org/10.3390/genes13101728