The Double-Faced Role of Nitric Oxide and Reactive Oxygen Species in Solid Tumors
Abstract
:1. Introduction
2. The Double-Faced Role of ROS/RNS in Cancer
3. ROS-Induced Genetic and Epigenetic Instability in Cancer Cells
4. Pleiotropic Role of Nitric Oxide in Cancer
5. High Steady-State ROS Levels in Tumor Cells
6. Multiple Roles of ROS/RNS in Cancer Proliferation
7. NO-Related Intracellular Hypoxia Provokes Tumor Progression
8. ROS and RNS Effects in the Tumor Microenvironment
9. NO in Initial and Acquired Resistance to Therapy
10. ROS/RNS-Mediated Mechanisms of Targeted Cancer Therapy
10.1. Free-Radical-Mediated Effects of Therapy Targeting VEGFR
10.2. Free-Radical-Mediated Effects of EGFR-Targeted Therapy
10.3. Free-Radical-Mediated Effects of HER-Targeted Therapy
10.4. Free-Radical-Mediated Effects of PDGFRα, KIT, ABL, and CSF-1 Receptor-Targeted Therapy
10.5. Free-Radical-Mediated Effects of BRAF-Targeted Therapy
11. Conclusions
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
Abbreviations
ABL = nonreceptor tyrosine kinase | AMPK = AMP-activated protein kinase |
AP-1 = activating protein-1 | CAF = cancer-associated fibroblast |
CAT = catalase | cGMP = cyclin guanosine monophosphate |
CREB = cAMP response element-binding protein | CSF-1 = colony-stimulating factor-1 |
DC = dendritic cell | DR5 = Death receptor 5 |
EGF = epidermal growth factor | EGFR = epidermal growth factor receptor |
EMT = epithelial-mesenchymal transition | FGF = fibroblast growth factor |
GCL = glutamate cysteine ligase | GPX = glutathione peroxidase |
GSH = glutathione | GST = glutathione S-transferase |
HER = human epidermal growth factor receptor | HDAC = Histone deacetylase |
HIF-1 = hypoxia-inducible factor 1 | JNK = c-Jun N-terminal kinase |
Keap1 = kelch-like protein 19 | KIT (CD117) = proto-oncogene, receptor tyrosine kinase |
MAPK = mitogen activated protein kinase | MDSC = myeloid-derived suppressor cell |
MMP = matrix metalloproteinases | NFE2L2 = nuclear factor erythroid-derived 2-like 2 |
NFkB = Nuclear Factor-κB | NOX = NADPH oxidase |
Nrf2 = nuclear factor erythroid 2-related factor 2 | PDGF = platelet-derived growth factor |
PDGFR = platelet-derived growth factor receptor | PGC-1 = peroxisome proliferator-activated receptor-gamma coactivator-1 |
PI3K/Akt = phosphoinositide-3-kinase–protein kinase B | PTEN = phosphatidylinositol-3,4,5-trisphosphate 3-phosphatase |
PTX = paclitaxel | PX-12-1 = methylpropyl 2-imidazolyl disulphide |
RKIP = Raf kinase inhibitor protein | Snail = transcription factor |
SOD = superoxide dismutase | TAF = tumor associated fibroblast |
TAM = tumor-associated macrophage | TIL = Tumor infiltrated lymphocyte |
TKI = tyrosine kinase inhibitor | Trx = thioredoxin |
VEGF = vascular endothelial growth factor | YY1 = Yin Yang 1 transcription factor |
References
- Jones, D.P.; Sies, H. The Redox Code. Antioxidants Redox Signal. 2015, 23, 734–746. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Laurent, A.; Nicco, C.; Chéreau, C.; Goulvestre, C.; Alexandre, J.; Alves, A.; Lévy, E.; Goldwasser, F.; Panis, Y.; Soubrane, O.; et al. Controlling tumor growth by modulating endogenous production of reactive oxygen species. Cancer Res. 2005, 65, 948–956. [Google Scholar] [PubMed]
- Li, P.; Wu, M.; Wang, J.; Sui, Y.; Liu, S.; Shi, D. NAC selectively inhibit cancer telomerase activity: A higher redox homeostasis threshold exists in cancer cells. Redox Biol. 2016, 8, 91–97. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Davies, M.J. Protein oxidation and peroxidation. Biochem. J. 2016, 473, 805–825. [Google Scholar] [CrossRef] [Green Version]
- Halliwell, B. Oxidative stress and cancer: Have we moved forward? Biochem. J. 2007, 401, 1–11. [Google Scholar] [CrossRef]
- Sosa, V.; Moliné, T.; Somoza, R.; Paciucci, R.; Kondoh, H.; LLeonart, M.E. Oxidative stress and cancer: An overview. Ageing Res. Rev. 2013, 12, 376–390. [Google Scholar] [CrossRef]
- Assi, M. The differential role of reactive oxygen species in early and late stages of cancer. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2017, 313, R646–R653. [Google Scholar] [CrossRef] [Green Version]
- Nguyen, K.S.H.; Neal, J.W.; Wakelee, H. Review of the current targeted therapies for non-small-cell lung cancer. World J. Clin. Oncol. 2014, 5, 576–587. [Google Scholar] [CrossRef]
- Halliwell, B.; Gutteridge, J.M.C. (Eds.) Free Radicals in Biology and Medicine; Oxford University Press: Oxford, UK, 2015; ISBN 9780198717478. [Google Scholar]
- Marinho, H.S.; Real, C.; Cyrne, L.; Soares, H.; Antunes, F. Hydrogen peroxide sensing, signaling and regulation of transcription factors. Redox Biol. 2014, 2, 535–562. [Google Scholar] [CrossRef] [Green Version]
- Rhee, S.G. H2O2, a necessary evil for cell signaling. Science 2006, 312, 1882–1883. [Google Scholar] [CrossRef]
- Chaiswing, L.; Clair, W.H.S.; Clair, D.K.S. Redox Paradox: A Novel Approach to Therapeutics-Resistant Cancer. Antioxidants Redox Signal. 2018, 29, 1237–1272. [Google Scholar] [CrossRef] [PubMed]
- Pervaiz, S.; Clement, M.V. Superoxide anion: Oncogenic reactive oxygen species? Int. J. Biochem. Cell Biol. 2007, 39, 1297–1304. [Google Scholar] [CrossRef] [PubMed]
- Sies, H. Hydrogen peroxide as a central redox signaling molecule in physiological oxidative stress: Oxidative eustress. Redox Biol. 2017, 11, 613–619. [Google Scholar] [CrossRef] [PubMed]
- Kim, Y.M.; Kim, T.H.; Chung, H.T.; Talanian, R.V.; Yin, X.M.; Billiar, T.R. Nitric oxide prevents tumor necrosis factor α-induced rat hepatocyte apoptosis by the interruption of mitochondrial apoptotic signaling through S-nitrosylation of caspase-8. Hepatology 2000, 32, 770–778. [Google Scholar] [CrossRef] [PubMed]
- Veskoukis, A.S.; Tsatsakis, A.M.; Kouretas, D. Dietary oxidative stress and antioxidant defense with an emphasis on plant extract administration. Cell Stress Chaperones 2012, 17, 11–21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Moloney, J.N.; Cotter, T.G. ROS signalling in the biology of cancer. Semin. Cell Dev. Biol. 2018, 80, 50–64. [Google Scholar] [CrossRef]
- Chia, N.; Wang, L.; Lu, X.; Senut, M.C.; Brenner, C.; Ruden, D.M. Hypothesis: Environmental regulation of 5-hydroxymethyl-cytosine by oxidative stress. Epigenetics 2011, 6, 853–856. [Google Scholar] [CrossRef] [Green Version]
- Lewandowska, J.; Bartoszek, A. DNA Methylation in Cancer Development, Diagnosis and Therapy—Multiple Opportunities for Genotoxic Agents to Act as Methylome Disruptors or Remediators. Mutagenesis 2011, 26, 475–487. [Google Scholar] [CrossRef]
- Simpson, N.E.; Tryndyak, V.P.; Pogribna, M.; Beland, F.A.; Pogribny, I.P. Modifying metabolically sensitive histone marks by inhibiting glutamine metabolism affects gene expression and alters cancer cell phenotype. Epigenetics 2012, 7, 1413–1420. [Google Scholar] [CrossRef] [Green Version]
- Rose, N.R.; Klose, R.J. Understanding the relationship between DNA methylation and histone lysine methylation. Biochim. Biophys. Acta Gene Regul. Mech. 2014, 1839, 1362–1372. [Google Scholar] [CrossRef] [Green Version]
- Santana, M.M.; Gonzalez, J.M.; Cruz, C. Nitric oxide accumulation: The evolutionary trigger for phytopathogenesis. Front. Microbiol. 2017, 8, 8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vanini, F.; Kashfi, K.; Nath, N. The dual role of iNOS in cancer. Redox Biol. 2015, 6, 334–343. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lipton, S.A.; Choi, Y.B.; Pan, Z.H.; Lei, S.Z.; Chen, H.S.V.; Sucher, N.J.; Loscalzo, J.; Singel, D.J.; Stamler, J.S. A redox-based mechanism for the neuroprotective and neurodestructive effects of nitric oxide and related nitroso-compounds. Nature 1993, 364, 626–632. [Google Scholar] [CrossRef] [PubMed]
- Keshet, R.; Erez, A. Arginine and the metabolic regulation of nitric oxide synthesis in cancer. DMM Dis. Model. Mech. 2018, 11. [Google Scholar] [CrossRef] [Green Version]
- Adams, L.; Franco, M.C.; Estevez, A.G. Reactive nitrogen species in cellular signaling. Exp. Biol. Med. 2015, 240, 711–717. [Google Scholar] [CrossRef] [Green Version]
- Bogdan, C. Nitric oxide and the regulation of gene expression. Trends Cell Biol. 2001, 11, 66–75. [Google Scholar] [CrossRef]
- Salimian Rizi, B.; Achreja, A.; Nagrath, D. Nitric Oxide: The Forgotten Child of Tumor Metabolism. Trends Cancer 2017, 3, 659–672. [Google Scholar] [CrossRef]
- Rigas, B. The use of nitric oxide-donating nonsteroidal anti-inflammatory drugs in the chemoprevention of colorectal neoplasia. Curr. Opin. Gastroenterol. 2007, 23, 55–59. [Google Scholar] [CrossRef]
- Pendyala, S.; Natarajan, V. Redox regulation of Nox proteins. Respir. Physiol. Neurobiol. 2010, 174, 265–271. [Google Scholar] [CrossRef] [Green Version]
- Vaquero, E.C.; Edderkaoui, M.; Pandol, S.J.; Gukovsky, I.; Gukovskaya, A.S. Reactive oxygen species produced by NAD(P)H oxidase inhibit apoptosis in pancreatic cancer cells. J. Biol. Chem. 2004, 279, 34643–34654. [Google Scholar] [CrossRef] [Green Version]
- Murphy, M.P. How mitochondria produce reactive oxygen species. Biochem. J. 2009, 417, 1–13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nazarewicz, R.R.; Dikalova, A.; Bikineyeva, A.; Ivanov, S.; Kirilyuk, I.A.; Grigor’Ev, I.A.; Dikalov, S.I. Does scavenging of mitochondrial superoxide attenuate cancer prosurvival signaling pathways? Antioxidants Redox Signal. 2013, 19, 344–349. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pelicano, H.; Lu, W.; Zhou, Y.; Zhang, W.; Chen, Z.; Hu, Y.; Huang, P. Mitochondrial dysfunction and reactive oxygen species imbalance promote breast cancer cell motility through a CXCL14-mediated mechanism. Cancer Res. 2009, 69, 2375–2383. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Porporato, P.E.; Payen, V.L.; Pérez-Escuredo, J.; De Saedeleer, C.J.; Danhier, P.; Copetti, T.; Dhup, S.; Tardy, M.; Vazeille, T.; Bouzin, C.; et al. A mitochondrial switch promotes tumor metastasis. Cell Rep. 2014, 8, 754–766. [Google Scholar] [CrossRef] [Green Version]
- Yoshida, S.; Tsutsumi, S.; Muhlebach, G.; Sourbier, C.; Lee, M.J.; Lee, S.; Vartholomaiou, E.; Tatokoro, M.; Beebe, K.; Miyajima, N.; et al. Molecular chaperone TRAP1 regulates a metabolic switch between mitochondrial respiration and aerobic glycolysis. Proc. Natl. Acad. Sci. USA 2013, 110, E1604–E1612. [Google Scholar] [CrossRef] [Green Version]
- Yuan, Y.; Wang, W.; Li, H.; Yu, Y.; Tao, J.; Huang, S.; Zeng, Z. Nonsense and missense mutation of mitochondrial ND6 gene promotes cell migration and invasion in human lung adenocarcinoma. BMC Cancer 2015, 15, 346. [Google Scholar] [CrossRef] [Green Version]
- Ishikawa, K.; Takenaga, K.; Akimoto, M.; Koshikawa, N.; Yamaguchi, A.; Imanishi, H.; Nakada, K.; Honma, Y.; Hayashi, J.I. ROS-generating mitochondrial DNA mutations can regulate tumor cell metastasis. Science 2008, 320, 661–664. [Google Scholar] [CrossRef] [Green Version]
- Pljesa-Ercegovac, M.; Savic-Radojevic, A.; Matic, M.; Coric, V.; Djukic, T.; Radic, T.; Simic, T. Glutathione transferases: Potential targets to overcome chemoresistance in solid tumors. Int. J. Mol. Sci. 2018, 19, 3785. [Google Scholar] [CrossRef] [Green Version]
- Savic-Radojevic, A.; Mimic-Oka, J.; Pljesa-Ercegovac, M.; Opacic, M.; Dragicevic, D.; Kravic, T.; Djokic, M.; Micic, S.; Simic, T. Glutathione S-Transferase-P1 Expression Correlates with Increased Antioxidant Capacity in Transitional Cell Carcinoma of the Urinary Bladder. Eur. Urol. 2007, 52, 470–477. [Google Scholar] [CrossRef]
- Simic, T.; Savic-Radojevic, A.; Pljesa-Ercegovac, M.; Matic, M.; Mimic-Oka, J. Glutathione S-transferases in kidney and urinary bladder tumors. Nat. Rev. Urol. 2009, 6, 281–289. [Google Scholar] [CrossRef]
- Morelli, M.B.; Amantini, C.; Santoni, M.; Soriani, A.; Nabissi, M.; Cardinali, C.; Santoni, A.; Santoni, G. Axitinib induces DNA damage response leading to senescence, mitotic catastrophe, and increased NK cell recognition in human renal carcinoma cells. Oncotarget 2015, 6, 36245–36259. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Djukic, T.; Simic, T.; Pljesa-Ercegovac, M.; Matic, M.; Suvakov, S.; Coric, V.; Dragicevic, D.; Savic-Radojevic, A. Upregulated glutathione transferase omega-1 correlates with progression of urinary bladder carcinoma. Redox Rep. 2017, 22, 486–492. [Google Scholar] [CrossRef] [PubMed]
- Wu, C.L.; Huang, A.C.; Yang, J.S.; Liao, C.L.; Lu, H.F.; Chou, S.T.; Ma, C.Y.; Hsia, T.C.; Ko, Y.C.; Chung, J.G. Benzyl isothiocyanate (BITC) and phenethyl isothiocyanate (PEITC)-mediated generation of reactive oxygen species causes cell cycle arrest and induces apoptosis via activation of caspase-3, mitochondria dysfunction and nitric oxide (NO) in human osteogenic sarcoma U-2 OS cells. J. Orthop. Res. 2011, 29, 1199–1209. [Google Scholar] [PubMed]
- Harris, I.S.; Treloar, A.E.; Inoue, S.; Sasaki, M.; Gorrini, C.; Lee, K.C.; Yung, K.Y.; Brenner, D.; Knobbe-Thomsen, C.B.; Cox, M.A.; et al. Glutathione and Thioredoxin Antioxidant Pathways Synergize to Drive Cancer Initiation and Progression. Cancer Cell 2015, 27, 211–222. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tobe, R.; Carlson, B.A.; Tsuji, P.A.; Lee, B.J.; Gladyshev, V.N.; Hatfield, D.L. Differences in redox regulatory systems in human lung and liver tumors suggest different avenues for therapy. Cancers (Basel) 2015, 7, 2262–2276. [Google Scholar] [CrossRef]
- Harvie, M. Nutritional Supplements and Cancer: Potential Benefits and Proven Harms. Am. Soc. Clin. Oncol. Educ. Book 2014, 34, e478–e486. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gupta, S.C.; Hevia, D.; Patchva, S.; Park, B.; Koh, W.; Aggarwal, B.B. Upsides and downsides of reactive oxygen species for Cancer: The roles of reactive oxygen species in tumorigenesis, prevention, and therapy. Antioxidants Redox Signal. 2012, 16, 1295–1322. [Google Scholar] [CrossRef] [Green Version]
- Rodic, S.; Vincent, M.D. Reactive oxygen species (ROS) are a key determinant of cancer’s metabolic phenotype. Int. J. Cancer 2018, 142, 440–448. [Google Scholar] [CrossRef]
- Michiels, C.; Tellier, C.; Feron, O. Cycling hypoxia: A key feature of the tumor microenvironment. Biochim. Biophys. Acta Rev. Cancer 2016, 1866, 76–86. [Google Scholar] [CrossRef]
- Almeida, M.; Han, L.; Martin-Millan, M.; O’Brien, C.A.; Manolagas, S.C. Oxidative stress antagonizes Wnt signaling in osteoblast precursors by diverting β-catenin from T cell factor- to forkhead box O-mediated transcription. J. Biol. Chem. 2007, 282, 27298–27305. [Google Scholar] [CrossRef] [Green Version]
- Mani, S.A.; Guo, W.; Liao, M.J.; Eaton, E.N.; Ayyanan, A.; Zhou, A.Y.; Brooks, M.; Reinhard, F.; Zhang, C.C.; Shipitsin, M.; et al. The Epithelial-Mesenchymal Transition Generates Cells with Properties of Stem Cells. Cell 2008, 133, 704–715. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Omori, E.; Inagaki, M.; Mishina, Y.; Matsumoto, K.; Ninomiya-Tsuji, J. Epithelial transforming growth factor β-activated kinase 1 (TAK1) is activated through two independent mechanisms and regulates reactive oxygen species. Proc. Natl. Acad. Sci. USA 2012, 109, 3365–3370. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Díaz, B.; Courtneidge, S.A. Redox signaling at invasive microdomains in cancer cells. Free Radic. Biol. Med. 2012, 52, 247–256. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tonelli, C.; Chio, I.I.C.; Tuveson, D.A. Transcriptional Regulation by Nrf2. Antioxidants Redox Signal. 2018, 29, 1727–1745. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wiemer, E.A.C. Stressed tumor cell, chemosensitized cancer. Nat. Med. 2011, 17, 1552–1554. [Google Scholar] [CrossRef] [PubMed]
- Fei, J.; Hong, A.; Dobbins, T.A.; Jones, D.; Lee, C.S.; Loo, C.; Al-Ghamdi, M.; Harnett, G.B.; Clark, J.; O’Brien, C.J.; et al. Prognostic significance of vascular endothelial growth factor in squamous cell carcinomas of the tonsil in relation to human papillomavirus status and epidermal growth factor receptor. Ann. Surg. Oncol. 2009, 16, 2908–2917. [Google Scholar] [CrossRef]
- Ushio-Fukai, M.; Nakamura, Y. Reactive oxygen species and angiogenesis: NADPH oxidase as target for cancer therapy. Cancer Lett. 2008, 266, 37–52. [Google Scholar] [CrossRef] [Green Version]
- Masson, N.; Ratcliffe, P.J. Hypoxia signaling pathways in cancer metabolism: The importance of co-selecting interconnected physiological pathways. Cancer Metab. 2014, 2, 3. [Google Scholar] [CrossRef] [Green Version]
- Semenza, G.L. HIF-1 mediates metabolic responses to intratumoral hypoxia and oncogenic mutations. J. Clin. Investig. 2013, 123, 3664–3671. [Google Scholar] [CrossRef] [Green Version]
- Mehdi, A.; Riazalhosseini, Y. Epigenome aberrations: Emerging driving factors of the clear cell renal cell carcinoma. Int. J. Mol. Sci. 2017, 18, 1774. [Google Scholar] [CrossRef] [Green Version]
- Poyton, R.; Hendrickson, M. Crosstalk between nitric oxide and hypoxia-inducible factor signaling pathways: An update. Res. Rep. Biochem. 2015, 5, 147. [Google Scholar] [CrossRef] [Green Version]
- Xu, W.; Liu, L.Z.; Loizidou, M.; Ahmed, M.; Charles, I.G. The role of nitric oxide in cancer. Cell Res. 2002, 12, 311–320. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Alderton, W.K.; Cooper, C.E.; Knowles, R.G. Nitric oxide synthases: Structure, function and inhibition. Biochem. J. 2001, 357, 593–615. [Google Scholar] [CrossRef] [PubMed]
- Van Faassen, E.E.; Bahrami, S.; Feelisch, M.; Hogg, N.; Kelm, M.; Kim-Shapiro, D.B.; Kozlov, A.V.; Li, H.; Lundberg, J.O.; Mason, R.; et al. Nitrite as regulator of hypoxic signaling in mammalian physiology. Med. Res. Rev. 2009, 29, 683–741. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, F.; Sonveaux, P.; Rabbani, Z.N.; Liu, S.; Yan, B.; Huang, Q.; Vujaskovic, Z.; Dewhirst, M.W.W.; Li, C.Y. Regulation of HIF-1α Stability through S-Nitrosylation. Mol. Cell 2007, 26, 63–74. [Google Scholar] [CrossRef] [Green Version]
- Hudson, C.C.; Liu, M.; Chiang, G.G.; Otterness, D.M.; Loomis, D.C.; Kaper, F.; Giaccia, A.J.; Abraham, R.T. Regulation of Hypoxia-Inducible Factor 1 Expression and Function by the Mammalian Target of Rapamycin. Mol. Cell. Biol. 2002, 22, 7004–7014. [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]
- Rizi, B.S.; Caneba, C.; Nowicka, A.; Nabiyar, A.W.; Liu, X.; Chen, K.; Klopp, A.; Nagrath, D. Nitric oxide mediates metabolic coupling of omentum-derived adipose stroma to ovarian and endometrial cancer cells. Cancer Res. 2015, 75, 456–471. [Google Scholar] [CrossRef] [Green Version]
- Marozkina, N.V.; Wei, C.; Yemen, S.; Wallrabe, H.; Nagji, A.S.; Liu, L.; Morozkina, T.; Jones, D.R.; Gaston, B. S-Nitrosoglutathione reductase in human lung cancer. Am. J. Respir. Cell Mol. Biol. 2012, 46, 63–70. [Google Scholar] [CrossRef] [Green Version]
- Basile, M.S.; Mazzon, E.; Krajnovic, T.; Draca, D.; Cavalli, E.; Al-Abed, Y.; Bramanti, P.; Nicoletti, F.; Mijatovic, S.; Maksimovic-Ivanic, D. Anticancer and Differentiation Properties of the Nitric Oxide Derivative of Lopinavir in Human Glioblastoma Cells. Molecules 2018, 23, 2463. [Google Scholar] [CrossRef] [Green Version]
- Maksimovic-Ivanic, D.; Mijatovic, S.; Miljkovic, D.; Harhaji-Trajkovic, L.; Timotijevic, G.; Mojic, M.; Dabideen, D.; Cheng, K.F.; McCubrey, J.A.; Mangano, K.; et al. The antitumor properties of a nontoxic, nitric oxide-modified version of saquinavir are independent of Akt. Mol. Cancer Ther. 2009, 8, 1169–1178. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mijatovic, S.; Maksimovic-Ivanic, D.; Mojic, M.; Malaponte, G.; Libra, M.; Cardile, V.; Miljkovic, D.; Harhaji, L.; Dabideen, D.; Cheng, K.F.; et al. Novel nitric oxide-donating compound (S,R)-3-phenyl-4,5-dihydro-5-isoxazole acetic acid–nitric oxide (GIT-27NO) induces p53 mediated apoptosis in human A375 melanoma cells. Nitric Oxide 2008, 19, 177–183. [Google Scholar] [CrossRef] [PubMed]
- Mijatovic, S.; Maksimovic-Ivanic, D.; Mojic, M.; Timotijevic, G.; Miljkovic, D.; Mangano, K.; Donia, M.; Di Cataldo, A.; Al-Abed, Y.; Cheng, K.F.; et al. Cytotoxic and immune-sensitizing properties of nitric oxide-modified saquinavir in iNOS-positive human melanoma cells. J. Cell. Physiol. 2011, 226, 1803–1812. [Google Scholar] [CrossRef] [PubMed]
- Mojic, M.; Mijatovic, S.; Maksimovic-Ivanic, D.; Dinic, S.; Grdovic, N.; Miljkovic, D.; Stosic-Grujicic, S.; Tumino, S.; Fagone, P.; Mangano, K.; et al. Saquinavir-NO-targeted S6 protein mediates sensitivity of androgen-dependent prostate cancer cells to TRAIL. Cell Cycle 2012, 11, 1174–1182. [Google Scholar] [CrossRef] [Green Version]
- Mojic, M.; Mijatovic, S.; Maksimovic-Ivanic, D.; Miljkovic, D.; Stosic-Grujicic, S.; Stankovic, M.; Mangano, K.; Travali, S.; Donia, M.; Fagone, P.; et al. Therapeutic Potential of Nitric Oxide-Modified Drugs in Colon Cancer Cells. Mol. Pharmacol. 2012, 82, 700–710. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Paskaš, S.; Krajnović, T.; Basile, M.S.; Dunđerović, D.; Cavalli, E.; Mangano, K.; Mammana, S.; Al-Abed, Y.; Nicoletti, F.; Mijatović, S.; et al. Senescence as a main mechanism of Ritonavir and Ritonavir-NO action against melanoma. Mol. Carcinog. 2019, 58, 1362–1375. [Google Scholar] [CrossRef]
- Paskas, S.; Mazzon, E.; Basile, M.S.; Cavalli, E.; Al-Abed, Y.; He, M.; Rakocevic, S.; Nicoletti, F.; Mijatovic, S.; Maksimovic-Ivanic, D. Lopinavir-NO, a nitric oxide-releasing HIV protease inhibitor, suppresses the growth of melanoma cells in vitro and in vivo. Investig. New Drugs 2019, 37, 1014–1028. [Google Scholar] [CrossRef]
- Duan, W.; Hou, J.; Chu, X.; Li, X.; Zhang, J.; Li, J.; Xu, W.; Zhang, Y. Synthesis and biological evaluation of novel histone deacetylases inhibitors with nitric oxide releasing activity. Bioorg. Med. Chem. 2015, 23, 4481–4488. [Google Scholar] [CrossRef]
- Bigagli, E.; Luceri, C.; De Angioletti, M.; Chegaev, K.; D’Ambrosio, M.; Riganti, C.; Gazzano, E.; Saponara, S.; Longini, M.; Luceri, F.; et al. New NO- and H2S-releasing doxorubicins as targeted therapy against chemoresistance in castration-resistant prostate cancer: In vitro and in vivo evaluations. Investig. New Drugs 2018, 36, 985–998. [Google Scholar] [CrossRef]
- Donia, M.; Maksimovic-Ivanic, D.; Mijatovic, S.; Mojic, M.; Miljkovic, D.; Timotijevic, G.; Fagone, P.; Caponnetto, S.; Al-Abed, Y.; McCubrey, J.A.; et al. In vitro and in vivo anticancer action of Saquinavir-NO, a novel nitric oxide-derivative of the protease inhibitor saquinavir, on hormone resistant prostate cancer cells. Cell Cycle 2011, 10, 492–499. [Google Scholar] [CrossRef] [Green Version]
- Donia, M.; Mangano, K.; Fagone, P.; De Pasquale, R.; Dinotta, F.; Coco, M.; Padron, J.; Al-Abed, Y.; Lombardo, G.A.G.; Maksimovic-Ivanic, D.; et al. Unique antineoplastic profile of Saquinavir-NO, a novel NO-derivative of the protease inhibitor Saquinavir, on the in vitro and in vivo tumor formation of A375 human melanoma cells. Oncol. Rep. 2012, 28, 682–688. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Maksimovic-Ivanic, D.; Fagone, P.; McCubrey, J.; Bendtzen, K.; Mijatovic, S.; Nicoletti, F. HIV-protease inhibitors for the treatment of cancer: Repositioning HIV protease inhibitors while developing more potent NO-hybridized derivatives? Int. J. Cancer 2017, 140, 1713–1726. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Maksimovic-Ivanic, D.; Mijatovic, S.; Harhaji, L.; Miljkovic, D.; Dabideen, D.; Kai, F.C.; Mangano, K.; Malaponte, G.; Al-Abed, Y.; Libra, M.; et al. Anticancer properties of the novel nitric oxide-donating compound (S,R)-3-phenyl-4,5-dihydro-5-isoxazole acetic acid-nitric oxide in vitro and in vivo. Mol. Cancer Ther. 2008, 7, 510–520. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mijatović, S.; Bramanti, A.; Nicoletti, F.; Fagone, P.; Kaluđerović, G.N.; Maksimović-Ivanić, D. Naturally occurring compounds in differentiation based therapy of cancer. Biotechnol. Adv. 2018, 36, 1622–1632. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mijatovic, S.; Maksimovic-Ivanic, D.; Timotijevic, G.; Miljkovic, D.; Donia, M.; Libra, M.; Coco, M.; McCubrey, J.; Al-Abed, Y.; Korac, A.; et al. Induction of caspase-independent apoptotic-like cell death of mouse mammary tumor TA3Ha cells in vitro and reduction of their lethality in vivo by the novel chemotherapeutic agent GIT-27NO. Free Radic. Biol. Med. 2010, 48, 1090–1099. [Google Scholar] [CrossRef]
- Nisoli, E.; Carruba, M.O. Nitric oxide and mitochondrial biogenesis. J. Cell Sci. 2006, 119, 2855–2862. [Google Scholar] [CrossRef] [Green Version]
- Hamanaka, R.B.; Chandel, N.S. Mitochondrial reactive oxygen species regulate cellular signaling and dictate biological outcomes. Trends Biochem. Sci. 2010, 35, 505–513. [Google Scholar] [CrossRef] [Green Version]
- Fischer, K.; Hoffmann, P.; Voelkl, S.; Meidenbauer, N.; Ammer, J.; Edinger, M.; Gottfried, E.; Schwarz, S.; Rothe, G.; Hoves, S.; et al. Inhibitory effect of tumor cell-derived lactic acid on human T cells. Blood 2007, 109, 3812–3819. [Google Scholar] [CrossRef]
- Colegio, O.R.; Chu, N.Q.; Szabo, A.L.; Chu, T.; Rhebergen, A.M.; Jairam, V.; Cyrus, N.; Brokowski, C.E.; Eisenbarth, S.C.; Phillips, G.M.; et al. Functional polarization of tumour-associated macrophages by tumour-derived lactic acid. Nature 2014, 513, 559–563. [Google Scholar] [CrossRef]
- De Palma, M.; Lewis, C.E. Macrophage regulation of tumor responses to anticancer therapies. Cancer Cell 2013, 23, 277–286. [Google Scholar] [CrossRef] [Green Version]
- Branco-Price, C.; Evans, C.E.; Johnson, R.S. Endothelial hypoxic metabolism in carcinogenesis and dissemination: HIF-A isoforms are a NO metastatic phenomenon. Oncotarget 2013, 4, 2567–2576. [Google Scholar] [CrossRef] [Green Version]
- Ehrenfeld, P.; Cordova, F.; Duran, W.N.; Sanchez, F.A. S-nitrosylation and its role in breast cancer angiogenesis and metastasis. Nitric Oxide Biol. Chem. 2019, 87, 52–59. [Google Scholar] [CrossRef]
- Weinberg, F.; Ramnath, N.; Nagrath, D. Reactive oxygen species in the tumor microenvironment: An overview. Cancers (Basel) 2019, 11, 1191. [Google Scholar] [CrossRef] [Green Version]
- Capparelli, C.; Whitaker-Menezes, D.; Guido, C.; Balliet, R.; Pestell, T.G.; Howell, A.; Sneddon, S.; Pestell, R.G.; Martinez-Outschoorn, U.; Lisanti, M.P.; et al. CTGF drives autophagy, glycolysis and senescence in cancer-associated fibroblasts via HIF1 activation, metabolically promoting tumor growth. Cell Cycle 2012, 11, 2272–2284. [Google Scholar] [CrossRef] [Green Version]
- Martinez-Outschoorn, U.E.; Pavlides, S.; Howell, A.; Pestell, R.G.; Tanowitz, H.B.; Sotgia, F.; Lisanti, M.P. Stromal-epithelial metabolic coupling in cancer: Integrating autophagy and metabolism in the tumor microenvironment. Int. J. Biochem. Cell Biol. 2011, 43, 1045–1051. [Google Scholar] [CrossRef] [Green Version]
- Pavlides, S.; Vera, I.; Gandara, R.; Sneddon, S.; Pestell, R.G.; Mercier, I.; Martinez-Outschoorn, U.E.; Whitaker-Menezes, D.; Howell, A.; Sotgia, F.; et al. Warburg meets autophagy: Cancer-Associated fibroblasts accelerate tumor growth and metastasis via oxidative stress, mitophagy, and aerobic glycolysis. Antioxidants Redox Signal. 2012, 16, 1264–1284. [Google Scholar] [CrossRef] [Green Version]
- Corbet, C.; Feron, O. Tumour acidosis: From the passenger to the driver’s seat. Nat. Rev. Cancer 2017, 17, 577–593. [Google Scholar] [CrossRef]
- Dietl, K.; Renner, K.; Dettmer, K.; Timischl, B.; Eberhart, K.; Dorn, C.; Hellerbrand, C.; Kastenberger, M.; Kunz-Schughart, L.A.; Oefner, P.J.; et al. Lactic Acid and Acidification Inhibit TNF Secretion and Glycolysis of Human Monocytes. J. Immunol. 2010, 184, 1200–1209. [Google Scholar] [CrossRef]
- 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]
- Mojia, M.; Pristov, J.B.; Maksimović-Ivanić, D.; Jones, D.R.; Stanić, M.; Mijatović, S.; Spasojević, I. Extracellular iron diminishes anticancer effects of vitamin C: An in vitro study. Sci. Rep. 2014, 4, 4. [Google Scholar] [CrossRef] [Green Version]
- Chang, C.H.; Qiu, J.; O’Sullivan, D.; Buck, M.D.; Noguchi, T.; Curtis, J.D.; Chen, Q.; Gindin, M.; Gubin, M.M.; Van Der Windt, G.J.W.; et al. Metabolic Competition in the Tumor Microenvironment Is a Driver of Cancer Progression. Cell 2015, 162, 1229–1241. [Google Scholar] [CrossRef] [Green Version]
- Le Bourgeois, T.; Strauss, L.; Aksoylar, H.I.; Daneshmandi, S.; Seth, P.; Patsoukis, N.; Boussiotis, V.A. Targeting T cell metabolism for improvement of cancer immunotherapy. Front. Oncol. 2018, 8, 8. [Google Scholar] [CrossRef]
- Hossain, F.; Al-Khami, A.A.; Wyczechowska, D.; Hernandez, C.; Zheng, L.; Reiss, K.; Del Valle, L.; Trillo-Tinoco, J.; Maj, T.; Zou, W.; et al. Inhibition of Fatty Acid Oxidation Modulates Immunosuppressive Functions of Myeloid-Derived Suppressor Cells and Enhances Cancer Therapies. Cancer Immunol. Res. 2015, 3, 1236–1247. [Google Scholar] [CrossRef] [Green Version]
- Van den Bossche, J.; Baardman, J.; Otto, N.A.; van der Velden, S.; Neele, A.E.; van den Berg, S.M.; Luque-Martin, R.; Chen, H.J.; Boshuizen, M.C.S.; Ahmed, M.; et al. Mitochondrial Dysfunction Prevents Repolarization of Inflammatory Macrophages. Cell Rep. 2016, 17, 684–696. [Google Scholar] [CrossRef] [Green Version]
- Everts, B.; Amiel, E.; Van Der Windt, G.J.W.; Freitas, T.C.; Chott, R.; Yarasheski, K.E.; Pearce, E.L.; Pearce, E.J. Commitment to glycolysis sustains survival of NO-producing inflammatory dendritic cells. Blood 2012, 120, 1422–1431. [Google Scholar] [CrossRef] [Green Version]
- Barsoum, I.B.; Koti, M.; Siemens, D.R.; Graham, C.H. Mechanisms of hypoxia-mediated immune escape in cancer. Cancer Res. 2014, 74, 7185–7190. [Google Scholar] [CrossRef] [Green Version]
- Chandel, N.S.; McClintock, D.S.; Feliciano, C.E.; Wood, T.M.; Melendez, J.A.; Rodriguez, A.M.; Schumacker, P.T. Reactive oxygen species generated at mitochondrial Complex III stabilize hypoxia-inducible factor-1α during hypoxia: A mechanism of O2 sensing. J. Biol. Chem. 2000, 275, 25130–25138. [Google Scholar] [CrossRef] [Green Version]
- LaGory, E.L.; Wu, C.; Taniguchi, C.M.; Ding, C.K.C.; Chi, J.T.; von Eyben, R.; Scott, D.A.; Richardson, A.D.; Giaccia, A.J. Suppression of PGC-1α Is Critical for Reprogramming Oxidative Metabolism in Renal Cell Carcinoma. Cell Rep. 2015, 12, 116–127. [Google Scholar] [CrossRef] [Green Version]
- Doménech, E.; Maestre, C.; Esteban-Martínez, L.; Partida, D.; Pascual, R.; Fernández-Miranda, G.; Seco, E.; Campos-Olivas, R.; Pérez, M.; Megias, D.; et al. AMPK and PFKFB3 mediate glycolysis and survival in response to mitophagy during mitotic arrest. Nat. Cell Biol. 2015, 17, 1304–1316. [Google Scholar] [CrossRef]
- Faubert, B.; Boily, G.; Izreig, S.; Griss, T.; Samborska, B.; Dong, Z.; Dupuy, F.; Chambers, C.; Fuerth, B.J.; Viollet, B.; et al. AMPK is a negative regulator of the warburg effect and suppresses tumor growth in vivo. Cell Metab. 2013, 17, 113–124. [Google Scholar] [CrossRef] [Green Version]
- Shao, D.; Oka, S.I.; Liu, T.; Zhai, P.; Ago, T.; Sciarretta, S.; Li, H.; Sadoshima, J. A redox-dependent mechanism for regulation of AMPK activation by thioredoxin1 during energy starvation. Cell Metab. 2014, 19, 232–245. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, L.; Ishdorj, G.; Gibson, S.B. Reactive oxygen species regulation of autophagy in cancer: Implications for cancer treatment. Free Radic. Biol. Med. 2012, 53, 1399–1410. [Google Scholar] [CrossRef] [PubMed]
- Lebleu, V.S.; O’Connell, J.T.; Gonzalez Herrera, K.N.; Wikman, H.; Pantel, K.; Haigis, M.C.; De Carvalho, F.M.; Damascena, A.; Domingos Chinen, L.T.; Rocha, R.M.; et al. PGC-1α mediates mitochondrial biogenesis and oxidative phosphorylation in cancer cells to promote metastasis. Nat. Cell Biol. 2014, 16, 992–1003. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shi, Y.H.; Wang, Y.X.; Bingle, L.; Gong, L.H.; Heng, W.J.; Li, Y.; Fang, W.G. In vitro study of HIF-1 activation and VEGF release by bFGF in the T47D breast cancer cell line under normoxic conditions: Involvement of PI-3K/Akt and MEK1/ERK pathways. J. Pathol. 2005, 205, 530–536. [Google Scholar] [CrossRef] [PubMed]
- Addeo, A.; Banna, G.L.; Metro, G.; DI Maio, M. Chemotherapy in combination with immune checkpoint inhibitors for the first-line treatment of patients with advanced non-small cell lung cancer: A systematic review and literature-based meta-analysis. Front. Oncol. 2019, 9, 264. [Google Scholar] [CrossRef] [Green Version]
- Kim, P.K.M.; Kwon, Y.G.; Chung, H.T.; Kim, Y.M. Regulation of caspases by nitric oxide. Ann. N. Y. Acad. Sci. 2002, 962, 42–52. [Google Scholar] [CrossRef]
- Kim, Y.M.; Talanian, R.V.; Billiar, T.R. Nitric oxide inhibits apoptosis by preventing increases in caspase-3- like activity via two distinct mechanisms. J. Biol. Chem. 1997, 272, 31138–31148. [Google Scholar] [CrossRef] [Green Version]
- Liao, W.; Ye, T.; Liu, H. Prognostic Value of Inducible Nitric Oxide Synthase (iNOS) in Human Cancer: A Systematic Review and Meta-Analysis. Biomed Res. Int. 2019, 2019. [Google Scholar] [CrossRef] [Green Version]
- Mijatovic, S.; Maksimovic-Ivanic, D.; Radovic, J.; Popadic, D.; Momcilovic, M.; Harhaji, L.; Miljkovic, D.; Trajkovic, V. Aloe-emodin prevents cytokine-induced tumor cell death: The inhibition of auto-toxic nitric oxide release as a potential mechanism. Cell. Mol. Life Sci. 2004, 61, 1805–1815. [Google Scholar] [CrossRef]
- Xu, Y.; Ren, H.; Liu, J.; Wang, Y.; Meng, Z.; He, Z.; Miao, W.; Chen, G.; Li, X. A switchable NO-releasing nanomedicine for enhanced cancer therapy and inhibition of metastasis. Nanoscale 2019, 11, 5474–5488. [Google Scholar] [CrossRef]
- Hu, Q.; Peng, J.; Liu, W.; He, X.; Cui, L.; Chen, X.; Yang, M.; Liu, H.; Liu, S.; Wang, H. Elevated cleaved caspase-3 is associated with shortened overall survival in several cancer types. Int. J. Clin. Exp. Pathol. 2014, 7, 5057–5070. [Google Scholar] [PubMed]
- Ichim, G.; Tait, S.W.G. A fate worse than death: Apoptosis as an oncogenic process. Nat. Rev. Cancer 2016, 16, 539–548. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bonavida, B.; Garban, H. Nitric oxide-mediated sensitization of resistant tumor cells to apoptosis by chemo-immunotherapeutics. Redox Biol. 2015, 6, 486–494. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Teppo, H.-R.; Soini, Y.; Karihtala, P. Reactive Oxygen Species-Mediated Mechanisms of Action of Targeted Cancer Therapy. Oxid. Med. Cell. Longev. 2017, 2017. [Google Scholar] [CrossRef]
- Chang, S.P.; Shen, S.C.; Lee, W.R.; Yang, L.L.; Chen, Y.C. Imatinib mesylate induction of ROS-dependent apoptosis in melanoma B16F0 cells. J. Dermatol. Sci. 2011, 62, 183–191. [Google Scholar] [CrossRef]
- Cui, W.; Zhang, Z.J.; Hu, S.Q.; Mak, S.H.; Xu, D.P.; Choi, C.L.; Wang, Y.Q.; Tsim, W.K.; Lee, M.Y.; Rong, J.H.; et al. Sunitinib produces neuroprotective effect via inhibiting nitric oxide overproduction. CNS Neurosci. Ther. 2014, 20, 244–252. [Google Scholar] [CrossRef]
- Suddek, G.M. Sunitinib improves chemotherapeutic efficacy and ameliorates cisplatin-induced nephrotoxicity in experimental animals. Cancer Chemother. Pharmacol. 2011, 67, 1035–1044. [Google Scholar] [CrossRef]
- Tesori, V.; Piscaglia, A.C.; Samengo, D.; Barba, M.; Bernardini, C.; Scatena, R.; Pontoglio, A.; Castellini, L.; Spelbrink, J.N.; Maulucci, G.; et al. The multikinase inhibitor Sorafenib enhances glycolysis and synergizes with glycolysis blockade for cancer cell killing. Sci. Rep. 2015, 5, 9149. [Google Scholar] [CrossRef] [Green Version]
- Doherty, K.R.; Wappel, R.L.; Talbert, D.R.; Trusk, P.B.; Moran, D.M.; Kramer, J.W.; Brown, A.M.; Shell, S.A.; Bacus, S. Multi-parameter in vitro toxicity testing of crizotinib, sunitinib, erlotinib, and nilotinib in human cardiomyocytes. Toxicol. Appl. Pharmacol. 2013, 272, 245–255. [Google Scholar] [CrossRef]
- Truong, T.H.; Ung, P.M.U.; Palde, P.B.; Paulsen, C.E.; Schlessinger, A.; Carroll, K.S. Molecular Basis for Redox Activation of Epidermal Growth Factor Receptor Kinase. Cell Chem. Biol. 2016, 23, 837–848. [Google Scholar] [CrossRef] [Green Version]
- Shan, F.; Shao, Z.; Jiang, S.; Cheng, Z. Erlotinib induces the human non–small-cell lung cancer cells apoptosis via activating ROS-dependent JNK pathways. Cancer Med. 2016, 5, 3166–3175. [Google Scholar] [CrossRef] [PubMed]
- Kwon, T.; Rho, J.K.; Lee, J.C.; Park, Y.H.; Shin, H.J.; Cho, S.; Kang, Y.K.; Kim, B.Y.; Yoon, D.Y.; Yu, D.Y. An important role for peroxiredoxin II in survival of A549 lung cancer cells resistant to gefitinib. Exp. Mol. Med. 2015, 47, e165. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Aird, K.M.; Allensworth, J.L.; Batinic-Haberle, I.; Lyerly, H.K.; Dewhirst, M.W.; Devi, G.R. ErbB1/2 tyrosine kinase inhibitor mediates oxidative stress-induced apoptosis in inflammatory breast cancer cells. Breast Cancer Res. Treat. 2012, 132, 109–119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sadeghirizi, A.; Yazdanparast, R.; Aghazadeh, S. Combating trastuzumab resistance by targeting thioredoxin-1/PTEN interaction. Tumor Biol. 2016, 37, 6737–6747. [Google Scholar] [CrossRef] [PubMed]
- Bauer, D.; Werth, F.; Nguyen, H.A.; Kiecker, F.; Eberle, J. Critical role of reactive oxygen species (ROS) for synergistic enhancement of apoptosis by vemurafenib and the potassium channel inhibitor TRAM-34 in melanoma cells. Cell Death Dis. 2017, 8, e2594. [Google Scholar] [CrossRef]
- Yu, L.; Gao, L.X.; Ma, X.Q.; Hu, F.X.; Li, C.M.; Lu, Z. Involvement of superoxide and nitric oxide in BRAFV600E inhibitor PLX4032-induced growth inhibition of melanoma cells. Integr. Biol. (United Kingdom) 2014, 6, 1211–1217. [Google Scholar] [CrossRef]
- Hartmann, J.; Haap, M.; Kopp, H.-G.; Lipp, H.-P. Tyrosine Kinase Inhibitors—A Review on Pharmacology, Metabolism and Side Effects. Curr. Drug Metab. 2009, 10, 470–481. [Google Scholar] [CrossRef]
- Chiou, J.F.; Tai, C.J.; Wang, Y.H.; Liu, T.Z.; Jen, Y.M.; Shiau, C.Y. Sorafenib induces preferential apoptotic killing of a drug- and radio-resistant Hep G2 cells through a mitochondria-dependent oxidative stress mechanism. Cancer Biol. Ther. 2009, 8, 1904–1913. [Google Scholar] [CrossRef] [Green Version]
- Thijs, A.M.J.; van Herpen, C.M.L.; Verweij, V.; Pertijs, J.; van den Broek, P.H.H.; van der Graaf, W.T.A.; Rongen, G.A. Impaired endothelium-dependent vasodilation does not initiate the development of sunitinib-associated hypertension. J. Hypertens. 2015, 33, 2075–2082. [Google Scholar] [CrossRef]
- Abdel-Aziz, A.K.; Shouman, S.; El-Demerdash, E.; Elgendy, M.; Abdel-Naim, A.B. Chloroquine synergizes sunitinib cytotoxicity via modulating autophagic, apoptotic and angiogenic machineries. Chem. Biol. Interact. 2014, 217, 28–40. [Google Scholar] [CrossRef]
- Ji, S.; Xiong, Y.; Zhao, X.; Liu, Y.; Yu, L.Q. Effect of the nrf2-are signaling pathway on biological characteristics and sensitivity to sunitinib in renal cell carcinoma. Oncol. Lett. 2019, 17, 5175–5186. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Coriat, R.; Nicco, C.; Cheŕeau, C.; Mir, O.; Alexandre, J.; Ropert, S.; Weill, B.; Chaussade, S.; Goldwasser, F.; Batteux, F. Sorafenib-induced hepatocellular carcinoma cell death depends on reactive oxygen species production in vitro and in vivo. Mol. Cancer Ther. 2012, 11, 2284–2293. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gao, L.; Wang, X.; Tang, Y.; Huang, S.; Hu, C.A.A.; Teng, Y. FGF19/FGFR4 signaling contributes to the resistance of hepatocellular carcinoma to sorafenib. J. Exp. Clin. Cancer Res. 2017, 36, 8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Itsumi, M.; Tatsugami, K. Immunotherapy for Renal Cell Carcinoma. Clin. Dev. Immunol. 2011, 2010, 1–8. [Google Scholar] [CrossRef]
- Qian, X.; Li, J.; Ding, J.; Wang, Z.; Zhang, W.; Hu, G. ERLOTINIB ACTIVATES MITOCHONDRIAL DEATH PATHWAYS RELATED TO THE PRODUCTION OF REACTIVE OXYGEN SPECIES IN THE HUMAN NON-SMALL CELL LUNG CANCER CELL LINE A549. Clin. Exp. Pharmacol. Physiol. 2009, 36, 487–494. [Google Scholar] [CrossRef]
- Korashy, H.M.; Attafi, I.M.; Ansari, M.A.; Assiri, M.A.; Belali, O.M.; Ahmad, S.F.; AL-Alallah, I.A.; Al Anazi, F.E.; Alhaider, A.A. Molecular mechanisms of cardiotoxicity of gefitinib in vivo and in vitro rat cardiomyocyte: Role of apoptosis and oxidative stress. Toxicol. Lett. 2016, 252, 50–61. [Google Scholar] [CrossRef]
- Okon, I.S.; Coughlan, K.A.; Zhang, M.; Wang, Q.; Zou, M.H. Gefitinib-mediated reactive oxygen specie (ROS) instigates mitochondrial dysfunction and drug resistance in lung cancer cells. J. Biol. Chem. 2015, 290, 9101–9110. [Google Scholar] [CrossRef] [Green Version]
- Krall, E.B.; Wang, B.; Munoz, D.M.; Ilic, N.; Raghavan, S.; Niederst, M.J.; Yu, K.; Ruddy, D.A.; Aguirre, A.J.; Kim, J.W.; et al. KEAP1 loss modulates sensitivity to kinase targeted therapy in lung cancer. Elife 2017, 6, e18970. [Google Scholar] [CrossRef] [Green Version]
- Yamadori, T.; Ishii, Y.; Homma, S.; Morishima, Y.; Kurishima, K.; Itoh, K.; Yamamoto, M.; Minami, Y.; Noguchi, M.; Hizawa, N. Molecular mechanisms for the regulation of Nrf2-mediated cell proliferation in non-small-cell lung cancers. Oncogene 2012, 31, 4768–4777. [Google Scholar] [CrossRef] [Green Version]
- Orcutt, K.P.; Parsons, A.D.; Sibenaller, Z.A.; Scarbrough, P.M.; Zhu, Y.; Sobhakumari, A.; Wilke, W.W.; Kalen, A.L.; Goswami, P.; Miller, F.J.; et al. Erlotinib-mediated inhibition of EGFR signaling induces metabolic oxidative stress through NOX4. Cancer Res. 2011, 71, 3932–3940. [Google Scholar] [CrossRef] [Green Version]
- Ebelt, N.D.; Kaoud, T.S.; Edupuganti, R.; Van Ravenstein, S.; Dalby, K.N.; Van Den Berg, C.L. A c-Jun N-terminal kinase inhibitor, JNK-IN-8, sensitizes triple negative breast cancer cells to lapatinib. Oncotarget 2017, 8, 104894–104912. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vu, T.; Claret, F.X. Trastuzumab: Updated mechanisms of action and resistance in breast cancer. Front. Oncol. 2012, 2. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Swain, S.M.; Baselga, J.; Kim, S.B.; Ro, J.; Semiglazov, V.; Campone, M.; Ciruelos, E.; Ferrero, J.M.; Schneeweiss, A.; Heeson, S.; et al. Pertuzumab, trastuzumab, and docetaxel in HER2-positive metastatic breast cancer. N. Engl. J. Med. 2014, 372, 724–734. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Luo, C.; Lim, J.H.; Lee, Y.; Granter, S.R.; Thomas, A.; Vazquez, F.; Widlund, H.R.; Puigserver, P. A PGC1α-mediated transcriptional axis suppresses melanoma metastasis. Nature 2016, 537, 422–426. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Target | Drug Name | Implicated ROS | Suggested Mechanism | Putative Redox Biomarkers | References |
---|---|---|---|---|---|
VEGFR | Axitinib | Not specified | Not specified | Oxidative DNA damage byproducts | [126] |
Sunitinib | ↓NO | Increased GSH down regulated NOS | - | [127,128] | |
Sorafenib | ↑H2O2, O2−, NO | Mitochondrial dysfunction and GSH depletion | Advanced oxidation protein products | [129] | |
EGFR | Crizotinib | ↑O2− | Prx upregulation associated with drug resistance | - | [130] |
Afatinib | Not specified | Oxidative stress associated with drug resistance | - | [131] | |
Erlotinib | Not specified | Induced ROS-mediated apoptosis | - | [132] | |
Gefitinib | Not specified | Prx II upregulation associated with drug resistance | - | [133] | |
HER1/HER2 | Lapatinib | Not specified | Upregulated SOD1/ SOD2 and GSH associated with drug resistance | - | [134] |
HER2 dimerization | Trastuzumab | Not specified | Increased Trx-1 associated with drug resistance | Restoration of plasma antioxidant activity | [135] |
PDGFRα, KIT, ABL, CSF-1 receptor | Imatinib | Not specified | ROS-dependent apoptosis | - | [126] |
BRAF V600E | Vemurafenib | ↑ NO and O2− production | Depolarization of mitochondrial membrane, induced PGC1α | - | [136,137] |
© 2020 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
Mijatović, S.; Savić-Radojević, A.; Plješa-Ercegovac, M.; Simić, T.; Nicoletti, F.; Maksimović-Ivanić, D. The Double-Faced Role of Nitric Oxide and Reactive Oxygen Species in Solid Tumors. Antioxidants 2020, 9, 374. https://doi.org/10.3390/antiox9050374
Mijatović S, Savić-Radojević A, Plješa-Ercegovac M, Simić T, Nicoletti F, Maksimović-Ivanić D. The Double-Faced Role of Nitric Oxide and Reactive Oxygen Species in Solid Tumors. Antioxidants. 2020; 9(5):374. https://doi.org/10.3390/antiox9050374
Chicago/Turabian StyleMijatović, Sanja, Ana Savić-Radojević, Marija Plješa-Ercegovac, Tatjana Simić, Ferdinando Nicoletti, and Danijela Maksimović-Ivanić. 2020. "The Double-Faced Role of Nitric Oxide and Reactive Oxygen Species in Solid Tumors" Antioxidants 9, no. 5: 374. https://doi.org/10.3390/antiox9050374
APA StyleMijatović, S., Savić-Radojević, A., Plješa-Ercegovac, M., Simić, T., Nicoletti, F., & Maksimović-Ivanić, D. (2020). The Double-Faced Role of Nitric Oxide and Reactive Oxygen Species in Solid Tumors. Antioxidants, 9(5), 374. https://doi.org/10.3390/antiox9050374