3,466
Views
9
CrossRef citations to date
0
Altmetric
Drug Evaluation

BIO 300: a promising radiation countermeasure under advanced development for acute radiation syndrome and the delayed effects of acute radiation exposure

&
Pages 429-441 | Received 10 Mar 2020, Accepted 15 Apr 2020, Published online: 25 May 2020

References

  • Koenig KL, Goans RE, Hatchett RJ, et al. Medical treatment of radiological casualties: current concepts. Ann Emerg Med. 2005;45:643–652.
  • Ohnishi T. The disaster at Japan’s Fukushima-Daiichi nuclear power plant after the March 11, 2011 earthquake and tsunami, and the resulting spread of radioisotope contamination. Radiat Res. 2012;177:1–14.
  • Williams JP, McBride WH. After the bomb drops: a new look at radiation-induced multiple organ dysfunction syndrome (MODS). Int J Radiat Biol. 2011;87:851–868.
  • Need JT, Mothershead JL. Strategic national stockpile program: implications for military medicine. Mil Med. 2006;171:698–702.
  • Fushiki S. Radiation hazards in children - lessons from Chernobyl, Three Mile Island and Fukushima. Brain Dev. 2013;35:220–227.
  • Hu TW, Slaysman KS. Health-related economic costs of the Three-Mile Island accident. Socioecon Plann Sci. 1984;18: 183–193.
  • Andersson KG, Mikkelsen T, Astrup P, et al. Estimation of health hazards resulting from a radiological terrorist attack in a city. Radiat Prot Dosimetry. 2008;131:297–307.
  • Bar-Dayan Y, Hagby M, Goldberg A, et al. Health implications of radiological terrorism: perspectives from Israel. J Emerg Trauma Shcock. 2009;2(2):117–123.
  • Biological Effects of Ionizing Radiation VII. Health risks from exposure to low levels of ionizing radiation. Washington, DC: National Academy Press; 2005
  • Gale RP, Armitage JO. Are we prepared for nuclear terrorism? N Engl J Med. 2018;378:1246–1254.
  • Seed TM. Radiation protectants: current status and future prospects. Health Phys. 2005;89:531–545.
  • Singh VK, Seed TM. A review of radiation countermeasures focusing on injury-specific medicinals and regulatory approval status: part I. Radiation sub-syndromes, animal models and FDA-approved countermeasures. Int J Radiat Biol. 2017;93:851–869.
  • Singh VK, Hanlon BK, Santiago PT, et al. A review of radiation countermeasures focusing on injury-specific medicinals and regulatory approval status: part III. Countermeasures under early stages of development along with ‘standard of care’ medicinal and procedures not requiring regulatory approval for use. Int J Radiat Biol. 2017;93:885–906.
  • Singh VK, Garcia M, Seed TM. A review of radiation countermeasures focusing on injury-specific medicinals and regulatory approval status: part II. Countermeasures for limited indications, internalized radionuclides, emesis, late effects, and agents demonstrating efficacy in large animals with or without FDA IND status. Int J Radiat Biol. 2017;93(9):870–884.
  • Stone HB, Moulder JE, Coleman CN, et al. Models for evaluating agents intended for the prophylaxis, mitigation and treatment of radiation injuries. Report of an NCI Workshop, December 3–4, 2003. Radiat Res. 2004;162:711–728.
  • McCann DGC. Radiation poisoning: current concepts in the acute radiation syndrome. Am J Clin Med. 2006;3:13–21.
  • Armed Forces Radiobiology Research Institute. Medical management of radiological casualities. 4th ed. Bethesda, MD, USA: Armed Forces Radiobiology Research Institute; 2013.
  • Singh VK, Newman VL, Romaine PL, et al. Radiation countermeasure agents: an update (2011–2014). Expert Opin Ther Pat. 2014;24:1229–1255.
  • Manas ES, Xu ZB, Unwalla RJ, et al. Understanding the selectivity of genistein for human estrogen receptor-beta using X-ray crystallography and computational methods. Structure. 2004;12:2197–2207.
  • Weiss JF, Landauer MR. History and development of radiation-protective agents. Int J Radiat Biol. 2009;85:539–573.
  • Humanetics Pharmaceuticals. BIO 300 – A unique, highly selective radiation modulator. 2019 [cited 2020 Jan 3]. Available from: https://www.humaneticscorp.com/
  • U.S. Food and Drug Administration. Guidance document: product development under the animal rule. 2015 [cited 2018 Oct 20]. Available from: http://www.fda.gov/downloads/drugs/guidancecomplianceregulatoryinformation/guidances/ucm399217.pdf
  • Singh VK, Newman VL, Romaine PL, et al. Use of biomarkers for assessing radiation injury and efficacy of countermeasures. Expert Rev Mol Diagn. 2016;16:65–81.
  • Pannkuk EL, Fornace AJ Jr., Laiakis EC. Metabolomic applications in radiation biodosimetry: exploring radiation effects through small molecules. Int J Radiat Biol. 2017;93:1151–1176.
  • Nair AB, Jacob S. A simple practice guide for dose conversion between animals and human. J Basic Clin Pharm. 2016;7:27–31.
  • Ye H, Shaw IC. Food flavonoid ligand structure/estrogen receptor-alpha affinity relationships - toxicity or food functionality? Food Chem Toxicol. 2019;129:328–336.
  • Nilsson S, Gustafsson JA. Estrogen receptors: therapies targeted to receptor subtypes. Clin Pharmacol Ther. 2011;89:44–55.
  • Heldring N, Pike A, Andersson S, et al. Estrogen receptors: how do they signal and what are their targets. Physiol Rev. 2007;87:905–931.
  • Warner M, Huang B, Gustafsson JA. Estrogen receptor beta as a pharmaceutical target. Trends Pharmacol Sci. 2017;38:92–99.
  • Weihua Z, Saji S, Makinen S, et al. Estrogen receptor (ER) beta, a modulator of ERalpha in the uterus. Proc Natl Acad Sci USA. 2000;97:5936–5941.
  • Shim GJ, Wang L, Andersson S, et al. Disruption of the estrogen receptor beta gene in mice causes myeloproliferative disease resembling chronic myeloid leukemia with lymphoid blast crisis. Proc Natl Acad Sci USA. 2003;100:6694–6699.
  • Nakajima Y, Akaogi K, Suzuki T, et al. Estrogen regulates tumor growth through a nonclassical pathway that includes the transcription factors ERbeta and KLF5. Sci Signal. 2011;4:ra22.
  • Rizza P, Barone I, Zito D, et al. Estrogen receptor beta as a novel target of androgen receptor action in breast cancer cell lines. Breast Cancer Res. 2014;16:R21.
  • Mizukami Y. In vivo functions of GPR30/GPER-1, a membrane receptor for estrogen: from discovery to functions in vivo. Endocr J. 2010;57:101–107.
  • Prossnitz ER, Hathaway HJ. What have we learned about GPER function in physiology and disease from knockout mice? J Steroid Biochem Mol Biol. 2015;153:114–126.
  • Ignatov T, Modl S, Thulig M, et al. GPER-1 acts as a tumor suppressor in ovarian cancer. J Ovarian Res. 2013;6:51.
  • Martin SG, Lebot MN, Sukkarn B, et al. Low expression of G protein-coupled oestrogen receptor 1 (GPER) is associated with adverse survival of breast cancer patients. Oncotarget. 2018;9:25946–25956.
  • Weissenborn C, Ignatov T, Ochel HJ, et al. GPER functions as a tumor suppressor in triple-negative breast cancer cells. J Cancer Res Clin Oncol. 2014;140:713–723.
  • Du ZR, Feng XQ, Li N, et al. G protein-coupled estrogen receptor is involved in the anti-inflammatory effects of genistein in microglia. Phytomedicine. 2018;43:11–20.
  • Liang S, Chen Z, Jiang G, et al. Activation of GPER suppresses migration and angiogenesis of triple negative breast cancer via inhibition of NF-kappaB/IL-6 signals. Cancer Lett. 2017;386:12–23.
  • Luo LJ, Liu F, Lin ZK, et al. Genistein regulates the IL-1 beta induced activation of MAPKs in human periodontal ligament cells through G protein-coupled receptor 30. Arch Biochem Biophys. 2012;522:9–16.
  • Vitale DC, Piazza C, Melilli B, et al. Isoflavones: estrogenic activity, biological effect and bioavailability. Eur J Drug Metab Pharmacokinet. 2013;38:15–25.
  • Kuiper GG, Lemmen JG, Carlsson B, et al. Interaction of estrogenic chemicals and phytoestrogens with estrogen receptor beta. Endocrinology. 1998;139:4252–4263.
  • Thomas P, Dong J. Binding and activation of the seven-transmembrane estrogen receptor GPR30 by environmental estrogens: A potential novel mechanism of endocrine disruption. J Steroid Biochem Mol Biol. 2006;102:175–179.
  • Landauer MR, Harvey AJ, Kaytor MD, et al. Mechanism and therapeutic window of a genistein nanosuspension to protect against hematopoietic-acute radiation syndrome. J Radiat Res. 2019;60:308–317.
  • Si H, Liu D. Phytochemical genistein in the regulation of vascular function: new insights. Curr Med Chem. 2007;14:2581–2589.
  • Majid S, Kikuno N, Nelles J, et al. Genistein induces the p21WAF1/CIP1 and p16INK4a tumor suppressor genes in prostate cancer cells by epigenetic mechanisms involving active chromatin modification. Cancer Res. 2008;68:2736–2744.
  • Frey RS, Li J, Singletary KW. Effects of genistein on cell proliferation and cell cycle arrest in nonneoplastic human mammary epithelial cells: involvement of Cdc2, p21(waf/cip1), p27(kip1), and Cdc25C expression. Biochem Pharmacol. 2001;61:979–989.
  • Ding H, Duan W, Zhu W-G, et al. p21 response to DNA damage induced by genistein and etoposide in human lung cancer cells. Biochem Biophys Res Commun. 2003;305:950–956.
  • Zhang Z, Wang CZ, Du GJ, et al. Genistein induces G2/M cell cycle arrest and apoptosis via ATM/p53-dependent pathway in human colon cancer cells. Int J Oncol. 2013;43:289–296.
  • Oki T, Sowa Y, Hirose T, et al. Genistein induces Gadd45 gene and G2/M cell cycle arrest in the DU145 human prostate cancer cell line. FEBS Lett. 2004;577:55–59.
  • La Rosa FA, Pierce JW, Sonenshein GE. Differential regulation of the c-myc oncogene promoter by the NF-kappa B rel family of transcription factors. Mol Cell Biol. 1994;14:1039–1044.
  • Gartel AL, Shchors K. Mechanisms of c-myc-mediated transcriptional repression of growth arrest genes. Exp Cell Res. 2003;283:17–21.
  • Su Y, Simmen RC. Soy isoflavone genistein upregulates epithelial adhesion molecule E-cadherin expression and attenuates beta-catenin signaling in mammary epithelial cells. Carcinogenesis. 2009;30:331–339.
  • Jagadeesh S, Kyo S, Banerjee PP. Genistein represses telomerase activity via both transcriptional and posttranslational mechanisms in human prostate cancer cells. Cancer Res. 2006;66:2107–2115.
  • Zhu J, Ren J, Tang L. Genistein inhibits invasion and migration of colon cancer cells by recovering WIF1 expression. Mol Med Rep. 2018;17:7265–7273.
  • Zenk JL. New therapy for the prevention and prophylactic treatment of acute radiation syndrome. Expert Opin Investig Drugs. 2007;16:767–770.
  • Landauer, M.R. et al. Radioprotection by oral administration of genistein in mice receiving total body irradiation. 48th Annual Meeting and ToxExpo; March 15–19; Baltimore, MD. 2009.
  • Landauer M. Radioprotection by the Soy Isoflavone Genistein. In: Arora R, editor. Herbal radiomodulators: applications in medicine, Homeland defense and space. Wallingford, England: CABI Publishing; 2008. p. 163–173.
  • Singh VK, Seed TM. Pharmacological management of ionizing radiation injuries: current and prospective agents and targeted organ systems. Expert Opin Pharmacother. 2020;21:317–337.
  • Landauer MR, Srinivasan V, Seed TM. Genistein treatment protects mice from ionizing radiation injury. J Appl Toxicol. 2003;23:379–385.
  • Davis TA, Clarke TK, Mog SR, et al. Subcutaneous administration of genistein prior to lethal irradiation supports multilineage, hematopoietic progenitor cell recovery and survival. Int J Radiat Biol. 2007;83(3):141–151.
  • Ha CT, Li XH, Fu D, et al. Genistein nanoparticles protect mouse hematopoietic system and prevent proinflammatory factors after gamma irradiation. Radiat Res. 2013;180:316–325.
  • Davis TA, Mungunsukh O, Zins S, et al. Genistein induces radioprotection by hematopoietic stem cell quiescence. Int J Radiat Biol. 2008;84:713–726.
  • Day RM, Davis TA, Barshishat-Kupper M, et al. Enhanced hematopoietic protection from radiation by the combination of genistein and captopril. Int Immunopharmacol. 2013;15:348–356.
  • Day RM, Barshishat-Kupper M, Mog SR, et al. Genistein protects against biomarkers of delayed lung sequelae in mice surviving high-dose total body irradiation. J Radiat Res. 2008;49:361–372.
  • Davis TA, Landauer MR, Mog SR, et al. Timing of captopril administration determines radiation protection or radiation sensitization in a murine model of total body irradiation. Exp Hematol. 2010;38:270–281.
  • Dykstra JC, Harvey AJ, Ingram MAC, et al. Development of BIO 300 as a Medical Countermeasure for H-ARS and DEARE-lung. In Policies and Regulatory Pathways to FDA Licensure: Radiation Countermeasures and Biodosimetry Devices. Rockville, MD, USA: RNCP, NIAID; 2018
  • Jackson DP, Sorensen DK, Cronkite EP, et al. Effectiveness of transfusions of fresh and lyophilized platelets in controlling bleeding due to thrombocytopenia. J Clin Invest. 1959;38:1689–1697.
  • Jackson IL, Vujaskovic Z, Down JD. Revisiting strain-related differences in radiation sensitivity of the mouse lung: recognizing and avoiding the confounding effects of pleural effusions. Radiat Res. 2010;173:10–20.
  • Jackson IL, Vujaskovic Z, Down JD. A further comparison of pathologies after thoracic irradiation among different mouse strains: finding the best preclinical model for evaluating therapies directed against radiation-induced lung damage. Radiat Res. 2011;175:510–518.
  • Jackson IL, Zodda A, Gurung G, et al. BIO 300, a nanosuspension of genistein, mitigates pneumonitis/fibrosis following high-dose radiation exposure in the C57L/J murine model. Br J Pharmacol. 2017;174:4738–4750.
  • Para AE, Bezjak A, Yeung IW, et al. Effects of genistein following fractionated lung irradiation in mice. Radiother Oncol. 2009;92:500–510.
  • Mahmood J, Jelveh S, Calveley V, et al. Mitigation of radiation-induced lung injury by genistein and EUK-207. Int J Radiat Biol. 2011;87:889–901.
  • Jackson IL, Pavlovic R, Alexander AA, et al. BIO 300, a nanosuspension of genistein, mitigates radiation-induced erectile dysfunction and sensitizes human prostate cancer xenografts to radiation therapy. Int J Radiat Oncol Biol Phys. 2019;105:400–409.
  • Brown SL, Kaytor MD, Lapanowski K, et al. BIO 300 (genistein oral nanosuspension) protects normal lung tissue and radiosensitizes tumor tissue in a mouse xenograft model of non-small cell lung cancer (NSCLC). In The 60th Annual Meeting of the Radiation Research Society; Las Vegas, NV; 2014.
  • Ossetrova NI, Sandgren DJ, Blakely WF. Protein biomarkers for enhancement of radiation dose and injury assessment in nonhuman primate total-body irradiation model. Radiat Prot Dosimetry. 2014;159:61–76.
  • Ossetrova NI, Condliffe DP, Ney PH, et al. Early-response biomarkers for assessment of radiation exposure in a mouse total-body irradiation model. Health Phys. 2014;106(6):772–786.
  • Krivokrysenko VI, Shakhov AN, Singh VK, et al. Identification of granulocyte colony-stimulating factor and interleukin-6 as candidate biomarkers of CBLB502 efficacy as a medical radiation countermeasure. J Pharmacol Exp Ther. 2012;343:497–508.
  • Singh VK, Simas M, Pollard H. Biomarkers for acute radiation syndrome: challenges for developing radiation countermeasures following animal rule. Expert Rev Mol Diagn. 2018;18:921–924.
  • Singh VK, Santiago PT, Simas M, et al. Acute radiation syndrome: an update on biomarkers for radiation injury. J Radiat Cancer Res. 2019;9:132–146.
  • Singh VK, Grace MB, Parekh VI, et al. Effects of genistein administration on cytokine induction in whole-body gamma irradiated mice. Int Immunopharmacol. 2009;9:1401–1410.
  • Kong FM, Ao X, Wang L, et al. The use of blood biomarkers to predict radiation lung toxicity: a potential strategy to individualize thoracic radiation therapy. Cancer Control. 2008;15:140–150.
  • Allen TC, Kurdowska A. Interleukin 8 and acute lung injury. Arch Pathol Lab Med. 2014;138:266–269.
  • Jones JW, Jackson IL, Vujaskovic Z, et al. Targeted metabolomics identifies pharmacodynamic biomarkers for BIO 300 mitigation of radiation-induced lung injury. Pharm Res. 2017;34:2698–2709.
  • Cheema AK, Mehta KY, Santiago PT, et al. Pharmacokinetic and metabolomic studies with BIO 300, a nanosuspension of genistein, in a nonhuman primate model. Int J Mol Sci. 2019;20:1231.
  • Chang KL, Kung ML, Chow NH, et al. Genistein arrests hepatoma cells at G2/M phase: involvement of ATM activation and upregulation of p21waf1/cip1 and Wee1. Biochem Pharmacol. 2004;67:717–726.
  • Ye R, Bodero A, Zhou BB, et al. The plant isoflavenoid genistein activates p53 and Chk2 in an ATM-dependent manner. J Biol Chem. 2001;276:4828–4833.
  • Zhu J, Zhang C, Qing Y, et al. Genistein induces apoptosis by stabilizing intracellular p53 protein through an APE1-mediated pathway. Free Radic Biol Med. 2015;86:209–218.
  • Landauer, M.R. et al. Radioprotection by oral administration of genistein in mice receiving total body irradiation. 48th Annual Meeting and ToxExpo; March 15–19; Baltimore, MD. 2009.
  • McClain RM, Wolz E, Davidovich A, et al. Reproductive safety studies with genistein in rats. Food Chem Toxicol. 2007;45:1319–1332.
  • McClain RM, Wolz E, Davidovich A, et al. Subchronic and chronic safety studies with genistein in dogs. Food Chem Toxicol. 2005;43:1461–1482.
  • McClain RM, Wolz E, Davidovich A, et al. Acute, subchronic and chronic safety studies with genistein in rats. Food Chem Toxicol. 2006;44:56–80.
  • Michael McClain R, Wolz E, Davidovich A, et al. Genetic toxicity studies with genistein. Food Chem Toxicol. 2006;44:42–55.
  • Ullmann U, Metzner J, Frank T, et al. Safety, tolerability, and pharmacokinetics of single ascending doses of synthetic genistein (Bonistein) in healthy volunteers. Adv Ther. 2005;22:65–78.
  • Ullmann U, Oberwittle H, Grossmann M, et al. Repeated oral once daily intake of increasing doses of the novel synthetic genistein product Bonistein in healthy volunteers. Planta Med. 2005;71:891–896.
  • Metzner JE, Frank T, Kunz I, et al. Study on the pharmacokinetics of synthetic genistein after multiple oral intake in post-menopausal women. Arzneimittelforschung. 2009;59:513–520.
  • Kim KH, Dodsworth C, Paras A, et al. High dose genistein aglycone therapy is safe in patients with mucopolysaccharidoses involving the central nervous system. Mol Genet Metab. 2013;109:382–385.
  • Busby MG, Jeffcoat AR, Bloedon LT, et al. Clinical characteristics and pharmacokinetics of purified soy isoflavones: single-dose administration to healthy men. Am J Clin Nutr. 2002;75:126–136.
  • Fischer L, Mahoney C, Jeffcoat AR, et al. Clinical characteristics and pharmacokinetics of purified soy isoflavones: multiple-dose administration to men with prostate neoplasia. Nutr Cancer. 2004;48:160–170.
  • Bloedon LT, Jeffcoat AR, Lopaczynski W, et al. Safety and pharmacokinetics of purified soy isoflavones: single-dose administration to postmenopausal women. Am J Clin Nutr. 2002;76:1126–1137.
  • Tacyildiz N, Ozyoruk D, Yavuz G, et al. Soy isoflavones ameliorate the adverse effects of chemotherapy in children. Nutr Cancer. 2010;62:1001–1005.
  • Ahmad IU, Forman JD, Sarkar FH, et al. Soy isoflavones in conjunction with radiation therapy in patients with prostate cancer. Nutr Cancer. 2010;62:996–1000.
  • Kaytor MD, Brown SL, Arapi I, et al. A phase 1b/2a study evaluating the pharmacokinetics, safety, and efficacy of nano-genistein and chemoradiotherapy for non-small cell lung cancer. In Annual Meeting of the Amercian Society for Radiation Oncology; 2018 Oct 21–24; San Antonio, TX; 2018. p. E688.
  • U. S. Food and Drug Administration. Search orphan drug designations and approvals. 2019 [cited 2020 Feb 5]. Available from: https://www.accessdata.fda.gov/scripts/opdlisting/oopd/
  • Singh VK, Pollard HB. Ionizing radiation-induced altered microRNA expression as biomarkers for assessing acute radiation injury. Expert Rev Mol Diagn. 2017;17:871–874.