Utilizing Superoxide Dismutase Mimetics to Enhance Radiation Therapy Response While Protecting Normal Tissues
Tài liệu tham khảo
Burdon, 1989, Cell proliferation and oxidative stress, Free Radic Res Commun, 7, 149, 10.3109/10715768909087937
Droge, 2002, Free radicals in the physiological control of cell function, Physiol Rev, 82, 47, 10.1152/physrev.00018.2001
Sarsour, 2009, Redox control of the cell cycle in health and disease, Antioxid Redox Signal, 11, 2985, 10.1089/ars.2009.2513
Handy, 2012, Redox regulation of mitochondrial function, Antioxid Redox Signal, 16, 1323, 10.1089/ars.2011.4123
Gille, 1997, Analyses of the molecular mechanism of adriamycin-induced cardiotoxicity, Free Radic Biol Med, 23, 775, 10.1016/S0891-5849(97)00025-7
Azzam, 2012, Ionizing radiation-induced metabolic oxidative stress and prolonged cell injury, Cancer Lett, 327, 48, 10.1016/j.canlet.2011.12.012
Hall, 2006
Aston, 2001, Computer-aided design (cad) of Mn(ii) complexes: Superoxide dismutase mimetics with catalytic activity exceeding the native enzyme, Inorg Chem, 40, 1779, 10.1021/ic000958v
Forman, 1973, Superoxide dismutase: A comparison of rate constants, Arch Biochem Biophys, 158, 396, 10.1016/0003-9861(73)90636-X
Mapuskar, 2017, Mitochondrial superoxide increases age-associated susceptibility of human dermal fibroblasts to radiation and chemotherapy, Cancer Res, 77, 5054, 10.1158/0008-5472.CAN-17-0106
Thompson, 2010, The manganese superoxide dismutase mimetic, M40403, protects adult mice from lethal total body irradiation, Free Radic Res, 44, 529, 10.3109/10715761003649578
Murphy, 2008, Efficacy of superoxide dismutase mimetic M40403 in attenuating radiation-induced oral mucositis in hamsters, Clin Cancer Res, 14, 4292, 10.1158/1078-0432.CCR-07-4669
Buettner, 2006, A new paradigm: Manganese superoxide dismutase influences the production of H2O2 in cells and thereby their biological state, Free Radic Biol Med, 41, 1338, 10.1016/j.freeradbiomed.2006.07.015
Anderson, 2018, Phase 1b/2a trial of the superoxide dismutase mimetic GC4419 to reduce chemoradiotherapy-induced oral mucositis in patients with oral cavity or oropharyngeal carcinoma, Int J Radiat Oncol Biol Phys, 100, 427, 10.1016/j.ijrobp.2017.10.019
Sonis, 2009, Mucositis: The impact, biology and therapeutic opportunities of oral mucositis, Oral Oncol, 45, 1015, 10.1016/j.oraloncology.2009.08.006
Henke, 2011, Palifermin decreases severe oral mucositis of patients undergoing postoperative radiochemotherapy for head and neck cancer: A randomized, placebo-controlled trial, J Clin Oncol, 29, 2815, 10.1200/JCO.2010.32.4103
Le, 2011, Palifermin reduces severe mucositis in definitive chemoradiotherapy of locally advanced head and neck cancer: A randomized, placebo-controlled study, J Clin Oncol, 29, 2808, 10.1200/JCO.2010.32.4095
Elting, 2007, Risk, outcomes, and costs of radiation-induced oral mucositis among patients with head-and-neck malignancies, Int J Radiat Oncol Biol Phys, 68, 1110, 10.1016/j.ijrobp.2007.01.053
Elting, 2008, Patient-reported measurements of oral mucositis in head and neck cancer patients treated with radiotherapy with or without chemotherapy: Demonstration of increased frequency, severity, resistance to palliation, and impact on quality of life, Cancer, 113, 2704, 10.1002/cncr.23898
Nonzee, 2008, Cancer, 113, 1446, 10.1002/cncr.23714
Traynor, 2010, Comprehensive IMRT plus weekly cisplatin for advanced head and neck cancer: The university of Wisconsin experience, Head Neck, 32, 599
Russo, 2008, Radiation treatment breaks and ulcerative mucositis in head and neck cancer, Oncologist, 13, 886, 10.1634/theoncologist.2008-0024
Vera-Llonch, 2006, Oral mucositis in patients undergoing radiation treatment for head and neck carcinoma, Cancer, 106, 329, 10.1002/cncr.21622
Allison, 2014, Multi-institutional, randomized, double-blind, placebo-controlled trial to assess the efficacy of a mucoadhesive hydrogel (MuGard) in mitigating oral mucositis symptoms in patients being treated with chemoradiation therapy for cancers of the head and neck, Cancer, 120, 1433, 10.1002/cncr.28553
Barber, 2007, Comparing pain control and ability to eat and drink with standard therapy vs gelclair: A preliminary, double centre, randomised controlled trial on patients with radiotherapy-induced oral mucositis, Support Care Cancer, 15, 427, 10.1007/s00520-006-0171-1
Lalla, 2014, MASCC/ISOO clinical practice guidelines for the management of mucositis secondary to cancer therapy, Cancer, 120, 1453, 10.1002/cncr.28592
Lambrecht, 2013, Support Care Cancer, 21, 2663, 10.1007/s00520-013-1829-0
Leenstra, 2014, Doxepin rinse versus placebo in the treatment of acute oral mucositis pain in patients receiving head and neck radiotherapy with or without chemotherapy: A phase iii, randomized, double-blind trial (NCCTG-N09C6 [alliance]), J Clin Oncol, 32, 1571, 10.1200/JCO.2013.53.2630
Rao, 2014, Phase ii multicenter trial of caphosol for the reduction of mucositis in patients receiving radiation therapy for head and neck cancer, Oral Oncol, 50, 765, 10.1016/j.oraloncology.2014.06.001
Sarvizadeh, 2015, Morphine mouthwash for the management of oral mucositis in patients with head and neck cancer, Adv Biomed Res, 4, 44, 10.4103/2277-9175.151254
Sonis, 2016, Could the biological robustness of low level laser therapy (photobiomodulation) impact its use in the management of mucositis in head and neck cancer patients, Oral Oncol, 54, 7, 10.1016/j.oraloncology.2016.01.005
Wong, 2017, A randomised controlled trial of caphosol mouthwash in management of radiation-induced mucositis in head and neck cancer, Radiother Oncol, 122, 207, 10.1016/j.radonc.2016.06.015
Spielberger, 2004, Palifermin for oral mucositis after intensive therapy for hematologic cancers, N Engl J Med, 351, 2590, 10.1056/NEJMoa040125
Sonis, 2018, Phase ii investigational oral drugs for the treatment of radio/chemotherapy induced oral mucositis, Expert Opin Investig Drugs, 27, 147, 10.1080/13543784.2018.1427732
Anderson, 2018, J Clin Oncol, 15
Hamilton, 2001, Does oxidative damage to DNA increase with age?, Proc Natl Acad Sci U S A, 98, 10469, 10.1073/pnas.171202698
Spitz, 2004, Metabolic oxidation/reduction reactions and cellular responses to ionizing radiation: A unifying concept in stress response biology, Cancer Metastasis Rev, 23, 311, 10.1023/B:CANC.0000031769.14728.bc
Suliman, 2016, Mitochondrial quality control as a therapeutic target, Pharmacol Rev, 68, 20, 10.1124/pr.115.011502
Batinic-Haberle, 1998, The ortho effect makes manganese(iii) meso-tetrakis(n-methylpyridinium-2-yl)porphyrin a powerful and potentially useful superoxide dismutase mimic, J Biol Chem, 273, 24521, 10.1074/jbc.273.38.24521
Batinic-Haberle, 2004, New class of potent catalysts of O2 -dismutation. Mn(iii) ortho-methoxyethylpyridyl- and di-ortho-methoxyethylimidazolylporphyrins, Dalton Trans, 1696, 10.1039/B400818A
Spasojevic, 2008, Pharmacokinetics of the potent redox-modulating manganese porphyrin, MnTE-2-PyP(5+), in plasma and major organs of B6C3F1 mice, Free Radic Biol Med, 45, 943, 10.1016/j.freeradbiomed.2008.05.015
Batinic-Haberle, 2018, Mn porphyrin-based redox-active drugs—differential effects as cancer therapeutics and protectors of normal tissue against oxidative injury, Antioxid Redox Signal, 10.1089/ars.2017.7453
Tovmasyan, 2013, Design, mechanism of action, bioavailability and therapeutic effects of Mn porphyrin-based redox modulators, Med Princ Pract, 22, 103, 10.1159/000341715
Weitner, 2013, Comprehensive pharmacokinetic studies and oral bioavailability of two Mn porphyrin-based sod mimics, MnTE-2-PyP5+ and MnTnHex-2-PyP5+, Free Radic Biol Med, 58, 73, 10.1016/j.freeradbiomed.2013.01.006
Leu, 2017, Cns bioavailability and radiation protection of normal hippocampal neurogenesis by a lipophilic Mn porphyrin-based superoxide dismutase mimic, MnTnBuOE-2-PyP(5), Redox Biol, 12, 864, 10.1016/j.redox.2017.04.027
Batinic-Haberle, 2015, An educational overview of the chemistry, biochemistry and therapeutic aspects of Mn porphyrins—from superoxide dismutation to ho-driven pathways, Redox Biol, 5, 43, 10.1016/j.redox.2015.01.017
Vujaskovic, 2002, A small molecular weight catalytic metalloporphyrin antioxidant with superoxide dismutase (sod) mimetic properties protects lungs from radiation-induced injury, Free Radic Biol Med, 33, 857, 10.1016/S0891-5849(02)00980-2
Gauter-Fleckenstein, 2008, Comparison of two Mn porphyrin-based mimics of superoxide dismutase in pulmonary radioprotection, Free Radic Biol Med, 44, 982, 10.1016/j.freeradbiomed.2007.10.058
Gauter-Fleckenstein, 2010, Early and late administration of MnTE-2-PyP5+ in mitigation and treatment of radiation-induced lung damage, Free Radic Biol Med, 48, 1034, 10.1016/j.freeradbiomed.2010.01.020
Gauter-Fleckenstein, 2014, Robust rat pulmonary radioprotection by a lipophilic Mn n-alkylpyridylporphyrin, MnTnHex-2-PyP(5+), Redox Biol, 2, 400, 10.1016/j.redox.2013.12.017
Cline JM, et al: Post-irradiation treatment with a superoxide dismutase mimic, MnTnHex-2-PyP(5+), mitigates radiation injury in the lungs of non-human primates after whole-thorax exposure to ionizing radiation. Antioxidants (Basel) 7:40.
Archambeau, 2013, Superoxide dismutase mimic, MnTE-2-PyP(5+) ameliorates acute and chronic proctitis following focal proton irradiation of the rat rectum, Redox Biol, 1, 599, 10.1016/j.redox.2013.10.002
Oberley-Deegan, 2012, The antioxidant, MnTE-2-PyP, prevents side-effects incurred by prostate cancer irradiation, PLoS One, 7, e44178, 10.1371/journal.pone.0044178
Shrishrimal S, et al: The sod mimic, MnTE-2-PyP, protects from chronic fibrosis and inflammation in irradiated normal pelvic tissues. Antioxidants (Basel) 6:87.
Ashcraft, 2015, Novel manganese-porphyrin superoxide dismutase-mimetic widens the therapeutic margin in a preclinical head and neck cancer model, Int J Radiat Oncol Biol Phys, 93, 892, 10.1016/j.ijrobp.2015.07.2283
Le, 2018, Ink4a/arf expression impairs neurogenesis in the brain of irradiated mice, Stem Cell Reports, 10, 1721, 10.1016/j.stemcr.2018.03.025
Weitzel, 2015, Radioprotection of the brain white matter by Mn(iii) n-butoxyethylpyridylporphyrin-based superoxide dismutase mimic MnTnBuOE-2-PyP5+, Mol Cancer Ther, 14, 70, 10.1158/1535-7163.MCT-14-0343
Weitzel, 2016, Neurobehavioral radiation mitigation to standard brain cancer therapy regimens by Mn(iii) n-butoxyethylpyridylporphyrin-based redox modifier, Environ Mol Mutagen, 57, 372, 10.1002/em.22021
Tse, 2004, Mechanistic analysis of the immunomodulatory effects of a catalytic antioxidant on antigen-presenting cells: Implication for their use in targeting oxidation-reduction reactions in innate immunity, Free Radic Biol Med, 36, 233, 10.1016/j.freeradbiomed.2003.10.029
Celic, 2014, Mn porphyrin-based sod mimic, MnTnHex-2-PyP(5+), and non-sod mimic, MnTBAP(3-), suppressed rat spinal cord ischemia/reperfusion injury via NF-kappab pathways, Free Radic Res, 48, 1426, 10.3109/10715762.2014.960865
Sheng, 2011, Neuroprotective efficacy from a lipophilic redox-modulating Mn(iii) n-hexylpyridylporphyrin, MnTnHex-2-PyP: Rodent models of ischemic stroke and subarachnoid hemorrhage, J Pharmacol Exp Ther, 338, 906, 10.1124/jpet.110.176701
Jackson, 2007, Superoxide dismutase mimetic reduces hypoxia-induced O2•−, TGF-beta, and vegf production by macrophages, Free Radic Res, 41, 8, 10.1080/10715760600913150
Chatterjee, 2017, MnTE-2-PyP treatment, or nox4 inhibition, protects against radiation-induced damage in mouse primary prostate fibroblasts by inhibiting the TGF-beta 1 signaling pathway, Radiat Res, 187, 367, 10.1667/RR14623.1
Kosmacek, 2016, MnTnBuOE-2-PyP protects normal colorectal fibroblasts from radiation damage and simultaneously enhances radio/chemotherapeutic killing of colorectal cancer cells, Oncotarget, 7, 34532, 10.18632/oncotarget.8923
Zhao, 2017, A novel redox regulator, MnTnBuOE-2-PyP (5+), enhances normal hematopoietic stem/progenitor cell function, Redox Biol, 12, 129, 10.1016/j.redox.2017.02.005
Chatterjee, 2018, The addition of manganese porphyrins during radiation inhibits prostate cancer growth and simultaneously protects normal prostate tissue from radiation damage, Antioxidants (Basel), 7
Tong, 2016, MnTE-2-PyP reduces prostate cancer growth and metastasis by suppressing p300 activity and p300/HIF-1/CREB binding to the promoter region of the PAI-1 gene, Free Radic Biol Med, 94, 185, 10.1016/j.freeradbiomed.2016.02.036
Rabbani, 2009, Antiangiogenic action of redox-modulating Mn(iii) meso-tetrakis(n-ethylpyridinium-2-yl)porphyrin, MnTE-2-PyP(5+), via suppression of oxidative stress in a mouse model of breast tumor, Free Radic Biol Med, 47, 992, 10.1016/j.freeradbiomed.2009.07.001
Zhao, 2005, A mechanism-based antioxidant approach for the reduction of skin carcinogenesis, Cancer Res, 65, 1401, 10.1158/0008-5472.CAN-04-3334
Shin, 2017, Mechanism of the antitumor and radiosensitizing effects of a manganese porphyrin, MnHex-2-PyP, Antioxid Redox Signal, 27, 1067, 10.1089/ars.2016.6889
Evans, 2014, Mn porphyrin in combination with ascorbate acts as a pro-oxidant and mediates caspase-independent cancer cell death, Free Radic Biol Med, 68, 302, 10.1016/j.freeradbiomed.2013.11.031
Tovmasyan, 2015, Anticancer therapeutic potential of Mn porphyrin/ascorbate system, Free Radic Biol Med, 89, 1231, 10.1016/j.freeradbiomed.2015.10.416
Moeller, 2004, Radiation activates HIF-1 to regulate vascular radiosensitivity in tumors: Role of reoxygenation, free radicals, and stress granules, Cancer Cell, 5, 429, 10.1016/S1535-6108(04)00115-1
Moeller, 2005, A manganese porphyrin superoxide dismutase mimetic enhances tumor radioresponsiveness, Int J Radiat Oncol Biol Phys, 63, 545, 10.1016/j.ijrobp.2005.05.026
Jaramillo, 2015, Manganese (iii) meso-tetrakis n-ethylpyridinium-2-yl porphyrin acts as a pro-oxidant to inhibit electron transport chain proteins, modulate bioenergetics, and enhance the response to chemotherapy in lymphoma cells, Free Radic Biol Med, 83, 89, 10.1016/j.freeradbiomed.2015.01.031
Jaramillo, 2012, Manganese porphyrin, MnTE-2-PyP5+, acts as a pro-oxidant to potentiate glucocorticoid-induced apoptosis in lymphoma cells, Free Radic Biol Med, 52, 1272, 10.1016/j.freeradbiomed.2012.02.001