2,541
Views
8
CrossRef citations to date
0
Altmetric
Review

Metabolomics-based predictive biomarkers of radiation injury and countermeasure efficacy: current status and future perspectives

ORCID Icon, &
Pages 641-654 | Received 20 Jan 2021, Accepted 19 May 2021, Published online: 07 Jun 2021

References

  • Singh VK, Newman VL, Romaine PL, et al., Use of biomarkers for assessing radiation injury and efficacy of countermeasures. Expert Rev Mol Diagn. 16(1): 65–81. 2016. .
  • Singh VK, Pollard HB. Ionizing radiation-induced altered microRNA expression as biomarkers for assessing acute radiation injury. Expert Rev Mol Diagn. 2017;17(10):871–874.
  • Hall EJ, Giaccia AJ. Radiobiology for the Radiobiologist. 7th ed. Philadelphia, PA: Lippincott Williams and Wilkins; 2012.
  • Bushberg JT, Kroger LA, Hartman MB, et al. Nuclear/radiological terrorism: emergency department management of radiation casualties. J Emerg Med. 2007;32(1):71–85.
  • Farese AM, MacVittie TJ. Filgrastim for the treatment of hematopoietic acute radiation syndrome. Drugs Today (Barc). 2015;51:537–548.
  • Zhong Y, Pouliot M, Downey AM, et al. Efficacy of delayed administration of sargramostim up to 120 hours post exposure in a nonhuman primate total body radiation model. Int J Radiat Biol. 2020:1–17. 10.1080/09553002.2019.1673499. in press.
  • Hankey KG, Farese AM, Blaauw EC, et al. Pegfilgrastim improves survival of lethally irradiated nonhuman primates. Radiat Res. 2015;183(6):643–655.
  • 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(11):921–924.
  • Pannkuk EL, Fornace AJ Jr., Laiakis EC. Metabolomic applications in radiation biodosimetry: exploring radiation effects through small molecules. Int J Radiat Biol. 2017;93(10):1151–1176.
  • Jones JW, Scott AJ, Tudor G, et al. Identification and quantitation of biomarkers for radiation-induced injury via mass spectrometry. Health Phys. 2014;106(1):106–119.
  • Gatto L, Hansen KD, Hoopmann MR, et al. Testing and validation of computational methods for mass spectrometry. J Proteome Res. 2016;15(3):809–814.
  • Rustam YH, Reid GE. Analytical challenges and recent advances in mass spectrometry based lipidomics. Anal Chem. 2018;90(1):374–397.
  • Coy SL, Cheema AK, Tyburski JB, et al., Radiation metabolomics and its potential in biodosimetry. Int J Radiat Biol. 87(8): 802–823. 2011. .
  • Vicente E, Vujaskovic Z, Jackson IL. A systematic review of metabolomic and lipidomic candidates for biomarkers in radiation injury. Metabolites. 2020;10(6):259.
  • Menon SS, Uppal M, Randhawa S, et al. Radiation metabolomics: current status and future directions. Front Oncol. 2016;6:20.
  • Laiakis EC, Canadell MP, Grilj V, et al. Serum lipidomic analysis from mixed neutron/X-ray radiation fields reveals a hyperlipidemic and pro-inflammatory phenotype. Sci Rep. 2019;9(1):4539.
  • Laiakis EC, Wang YW, Young EF, et al. Metabolic Dysregulation after Neutron Exposures Expected from an Improvised Nuclear Device. Radiat Res. 2017;188(1):21–34.
  • Quehenberger O, Armando AM, Brown AH, et al. Lipidomics reveals a remarkable diversity of lipids in human plasma. J Lipid Res. 2010;51(11):3299–3305.
  • Jelonek K, Pietrowska M, Ros M, et al. Radiation-induced changes in serum lipidome of head and neck cancer patients. Int J Mol Sci. 2014;15(4):6609–6624.
  • Lin X, Fuks Z, Kolesnick R. Ceramide mediates radiation-induced death of endothelium. Crit Care Med. 2000;28(Supplement):N87–93.
  • Kolesnick R, Fuks Z. Radiation and ceramide-induced apoptosis. Oncogene. 2003;22(37):5897–5906.
  • Jones JW, Carter CL, Li F, et al. Ultraperformance convergence chromatography-high resolution tandem mass spectrometry for lipid biomarker profiling and identification. Biomed Chromatogr. 2017;39(3):e3822.
  • Pannkuk EL, Laiakis EC, Authier S, et al. Targeted metabolomics of nonhuman primate serum after exposure to ionizing radiation: potential tools for high-throughput biodosimetry. RSC Adv. 2016;6(56):51192–51202.
  • Goudarzi M, Weber WM, Chung J, et al. Serum dyslipidemia is induced by internal exposure to Strontium-90 in Mice, Lipidomic profiling using a data-independent liquid Chromatography–Mass Spectrometry Approach. J Proteome Res. 2015;14(9):4039–4049.
  • Carter CL, Jones JW, Farese AM, et al. Lipidomic dysregulation within the lung parenchyma following whole-thorax lung irradiation: markers of injury, inflammation and fibrosis detected by MALDI-MSI. Sci Rep. 2017;7(1):10343.
  • Pannkuk EL, Laiakis EC, Mak TD, et al. A lipidomic and metabolomic serum signature from nonhuman primates exposed to ionizing radiation. Metabolomics. 2016;12(5):80.
  • Pannkuk EL, Laiakis EC, Singh VK, et al., Lipidomic signatures of nonhuman primates with radiation-induced hematopoietic syndrome. Sci Rep. 7(1): 9777. 2017. .
  • Cheema AK, Hinzman CP, Mehta KY, et al., Plasma derived exosomal biomarkers of exposure to ionizing radiation in nonhuman primates. Int J Mol Sci. 19(11): 3427. 2018.
  • Khan AR, Rana P, Devi MM, et al. Nuclear magnetic resonance spectroscopy-based metabonomic investigation of biochemical effects in serum of γ-irradiated mice. Int J Radiat Biol. 2011;87(1):91–97.
  • Kurland IJ, Broin P, Golden A, et al. Integrative Metabolic Signatures for Hepatic Radiation Injury. PloS One. 2015;89(6):e0124795.
  • Pannkuk EL, Laiakis EC, Fornace AJ Jr., et al., A metabolomic serum signature from nonhuman primates treated with a radiation countermeasure, gamma-tocotrienol, and exposed to ionizing radiation. Health Phys. 115(1): 3–11. 2018. .
  • Pannkuk EL, Laiakis EC, Authier S, et al. Gas chromatography/mass spectrometry metabolomics of urine and serum from nonhuman primates exposed to ionizing radiation: impacts on the tricarboxylic acid cycle and protein metabolism. J Proteome Res. 2017;16(5):2091–2100.
  • Bujold K, Hauer-Jensen M, Donini O, et al. Citrulline as a biomarker for gastrointestinal-acute radiation syndrome: species differences and experimental condition effects. Radiat Res. 2016;186(1):71–78.
  • Wang J, Shao L, Hendrickson HP, et al. Total body irradiation in the “hematopoietic” dose range induces substantial intestinal injury in non-human primates. Radiat Res. 2015;184(5):545–553.
  • Jang WG, Park JY, Lee J, et al. Investigation of relative metabolic changes in the organs and plasma of rats exposed to X-ray radiation using HR-MAS1 HNMR and solution 1HNMR. NMR Biomed. 2016;29(4):507–518.
  • Cheema AK, Li Y, Girgis M, et al., Metabolomic studies in tissues of mice treated with amifostine and exposed to gamma-radiation. Sci Rep. 9(1): 15701. 2019. .
  • Cheema AK, Suman S, Kaur P, et al. Long-term differential changes in mouse intestinal metabolomics after γ and heavy ion radiation exposure. PloS One. 2014;9(1):87079.
  • Rotolo JA, Fong CS, Bodo S, et al. Anti-ceramide single-chain variable fragment mitigates radiation GI syndrome mortality independent of DNA repair. JCI Insight. 2021;6(8). 10.1172/jci.insight.145380.
  • Wang Z, Zhao Y. Gut microbiota derived metabolites in cardiovascular health and disease. Protein Cell. 2018;9(5):416–431.
  • Comai S, Bertazzo A, Brughera M, et al. Tryptophan in health and disease. Adv Clin Chem. 2020;95:165–218.
  • Satyamitra MM, Cassatt DR, Hollingsworth BA, et al., Metabolomics in Radiation Biodosimetry: current Approaches and Advances. Metabolites. 10(8): 328. 2020. .
  • Dorr H, Meineke V. Acute radiation syndrome caused by accidental radiation exposure - therapeutic principles. BMC Med. 2011;9(1):126.
  • Gourmelon P, Benderitter M, Bertho JM, et al. European consensus on the medical management of acute radiation syndrome and analysis of the radiation accidents in Belgium and Senegal. Health Phys. 2010;98(6):825–832.
  • McCann DGC. Radiation poisoning: current concepts in the acute radiation syndrome. Am J Clin Med. 2006;3:13–21.
  • Peter RU, Gottlober P, Nadeshina N, et al. Radiation lentigo. A distinct cutaneous lesion after accidental radiation exposure. Arch Dermatol. 1997;133(2):209–211.
  • Peter RU, Gottlober P. Management of cutaneous radiation injuries: diagnostic and therapeutic principles of the cutaneous radiation syndrome. Mil Med. 2002;167(suppl_1):110–112.
  • Peter RU, Braun-Falco O, Birioukov A, et al. Chronic cutaneous damage after accidental exposure to ionizing radiation: the Chernobyl experience. J Am Acad Dermatol. 1994;30(5):719–723.
  • Dainiak N, Gent RN, Carr Z, et al. Literature review and global consensus on management of acute radiation syndrome affecting nonhematopoietic organ systems. Disaster Med Public Health Prep. 2011;184(3):183–201.
  • Fliedner TM, Dorr DH, Meineke V. Multi-organ involvement as a pathogenetic principle of the radiation syndromes: a study involving 110 case histories documented in SEARCH and classified as the bases of haematopoietic indicators of effect. British Journal of Radiology Suppl. 2005;Supplement_27(1):1–8.
  • Hill RP. Radiation effects on the respiratory system. British Journal of Radiology Suppl. 2005;27(1):75–81.
  • Moulder JE, Cohen EP. Radiation-induced multi-organ involvement and failure: the contribution of radiation effects on the renal system. British Journal of Radiology Suppl. 2005;27(1):82–88.
  • 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(6):711–728.
  • Singh VK, Seed TM. Pharmacological management of ionizing radiation injuries: current and prospective agents and targeted organ systems. Expert Opin Pharmacother. 2020;21(3):317–337.
  • MacVittie TJ. The MCART consortium animal model series: MCART animal model refinement and MCM development: defining organ dose, organ-specific tissue imaging, model validation and the natural history between the acute radiation syndrome (ARS) and the delayed effects of acute radiation exposure (DEARE). Health Phys. 2015;109(5):335–341.
  • Molls M, Budach V, Bamberg M. Total body irradiation: the lung as critical organ. Strahlenther Onkol. 1986;162(4):226–232.
  • MacVittie TJ, Farese AM, Kane MAARS. DEARE, and multiple-organ injury: a strategic and tactical approach to link radiation effects, animal models, medical countermeasures, and biomarker development to predict clinical outcome. Health Phys. 2019;116(4):453.
  • MacVittie TJ, Farese AM, Parker GA, et al. The time course of radiation-induced lung injury in a nonhuman primate model of partial-body irradiation with minimal bone marrow sparing: clinical and radiographic evidence and the effect of neupogen administration. Health Phys. 2019;10(3):328–382.
  • Seed TM, Blakely WF, Knudson GB, et al. International conference on low-level radiation injury and medical countermeasures. Mil Med. 2002;167:1–143.
  • Seed TM, Fry SA, Neta R, et al. Prevention and treatments: summary statement. Mil Med. 2002;98(suppl_1):87–93.
  • Ralston-Hooper K, Jannasch A, Adamec J, et al. The use of two-dimensional gas chromatography-time-of-flight mass spectrometry (GCxGC-TOF-MS) for metabolomic analysis of polar metabolites. Methods Mol Biol. 2011;708:205–211.
  • Forster J, Famili I, Fu P, et al. Genome-scale reconstruction of the Saccharomyces cerevisiae metabolic network. Genome Res. 2003;13(2):244–253.
  • Fiehn O. Metabolomics–the link between genotypes and phenotypes. Plant Mol Biol. 2002;48(1/2):155–171.
  • Pannkuk EL, Laiakis EC, Girgis M, et al. Temporal effects on radiation responses in nonhuman primates: identification of biofluid small molecule signatures by gas chromatography–mass spectrometry metabolomics. Metabolites. 2019;9(5):98.
  • Horning EC, Horning MG. Metabolic profiles: chromatographic methods for isolation and characterization of a variety of metabolites in man. Methods in Medical Research. 1970;12:369–371.
  • Markley JL, Bruschweiler R, Edison AS, et al. The future of NMR-based metabolomics. Curr Opin Biotechnol. 2017;43:34–40.
  • Cheng J, Lan W, Zheng G, et al. Metabolomics: a high-throughput platform for metabolite profile exploration. Methods Mol Biol. 2018;1754:265–292.
  • Clendinen CS, Monge ME, Fernandez FM. Ambient mass spectrometry in metabolomics. Analyst. 2017;142(17):3101–3117.
  • Fuhrer T, Zamboni N. High-throughput discovery metabolomics. Curr Opin Biotechnol. 2015;31:73–78.
  • Taraboletti A, Goudarzi M, Kabir A, et al. Fabric Phase Sorptive Extraction—A metabolomic preprocessing approach for ionizing radiation exposure assessment. J Proteome Res. 2019;18(8):3020–3031.
  • Laiakis EC. Metabolomic applications in radiation biodosimetry. Methods Mol Biol. 2019;1978:391–402.
  • Singh VK, Seed TM. An update on sargramostim for treatment of acute radiation syndrome. Drugs Today (Barc). 2018;54(11):679–693.
  • Gale RP, Armitage JO. Use of molecularly-cloned haematopoietic growth factors in persons exposed to acute high-dose, high-dose rate whole-body ionizing radiations. Blood Rev. 2021;45:100690.
  • Singh VK, Newman VL, Seed TM. Colony-stimulating factors for the treatment of the hematopoietic component of the acute radiation syndrome (H-ARS): a review. Cytokine. 2015;71(1):22–37.
  • Amgen Inc. Neupogen (filgrastim) injection for subcutaneous or intravenous use. 2015. Available at: http://pi.amgen.com/united_states/neupogen/neupogen_pi_hcp_english.pdf [Last accessed 2015 Apr 02]
  • Amgen Inc. Neulasta (pegfilgrastim) injection for subcutaneous use. 2015. Available at: http://pi.amgen.com/united_states/neulasta/neulasta_pi_hcp_english.pdf [Last accessed 2015 Nov 19]
  • Sanofi-Aventis U.S. LLC. LEUKINE® (sargramostim) for injection, for subcutaneous or intravenous use. 2018. Available at: https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/103362s5240lbl.pdf?utm_campaign=20180329%20MCMi&utm_medium=email&utm_source=Eloqua [Last accessed 2018 Apr 01]
  • Farese AM, Cohen MV, Katz BP, et al. Filgrastim improves survival in lethally irradiated nonhuman primates. Radiat Res. 2013;179(1):89–100.
  • Singh VK, Seed TM, Olabisi AO. Drug discovery strategies for acute radiation syndrome. Expert Opin Drug Discov. 2019;14(7):701–715.
  • U.S. Food and Drug Administration. Guidance for industry: product developoment under the Animal Rule. 2015. Available at: http://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/UCM399217.pdf [Last accessed 2016 Jul 5].
  • Nair AB, Jacob S. A simple practice guide for dose conversion between animals and human. J Basic Clin Pharm. 2016;54(2):27–31.
  • U.S. Food and Drug Administration. More about biomarkers & qualification. 2018. Available at: https://www.fda.gov/Drugs/DevelopmentApprovalProcess/DrugDevelopmentToolsQualificationProgram/BiomarkerQualificationProgram/ucm535408.htm [Last accessed 2018 May 18]
  • Aebersold P. FDA experience with medical countermeasures under the animal rule. Adv Prev Med. 2012;2012:507571.
  • Administration USFaD. CDER Drug and biologics animal rule approvals. 2020. Available at: https://www.fda.gov/media/107839/download [Last accessed 2010 Oct 11]
  • Patterson AD, Lanz C, Gonzalez FJ, et al. The role of mass spectrometry-based metabolomics in medical countermeasures against radiation. Mass Spectrom Rev. 2010;29(3):503–521.
  • Fendler W, Malachowska B, Meghani K, et al. Evolutionarily conserved serum microRNAs predict radiation-induced fatality in nonhuman primates. Sci Transl Med. 2017;9:eaal2408.
  • Ghosh SP, Pathak R, Kumar P, et al. Gamma-tocotrienol modulates radiation-induced microRNA expression in mouse spleen. Radiat Res. 2016;185(5):485–495.
  • Singh VK, Seed TM. BIO 300: a promising radiation countermeasure under advanced development for acute radiation syndrome and the delayed effects of acute radiation exposure. Expert Opin Investig Drugs. 2020;29(5):429–441.
  • 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(24):4738–4750.
  • Jones JW, Jackson IL, Vujaskovic Z, et al., Targeted metabolomics identifies pharmacodynamic biomarkers for BIO 300 mitigation of radiation-induced lung injury. Pharm Res. 12(12): 369–371. 2017. .
  • Niaudet C, Bonnaud S, Guillonneau M, et al. Plasma membrane reorganization links acid sphingomyelinase/ceramide to p38 MAPK pathways in endothelial cells apoptosis. Cell Signal. 2017;33:10–21.
  • Corre I, Guillonneau M, Paris F. Membrane signaling induced by high doses of ionizing radiation in the endothelial compartment. Relevance in radiation toxicity. Int J Mol Sci. 2013;14(11):22678–22696.
  • 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. 20(5): 1231. 2019. .
  • Sen CK, Khanna S, Roy S. Tocotrienols: vitamin E beyond tocopherols. Life Sci. 2006;78(18):2088–2098.
  • Singh VK, Beattie LA, Seed TM. Vitamin E: tocopherols and tocotrienols as potential radiation countermeasures. J Radiat Res. 2013;54(6):973–988.
  • Singh VK, Hauer-Jensen M. γ-Tocotrienol as a Promising Countermeasure for Acute Radiation Syndrome: current Status. Int J Mol Sci. 2016;17(5):e663.
  • Singh VK, Kulkarni S, Fatanmi OO, et al. Radioprotective efficacy of gamma-tocotrienol in nonhuman primates. Radiat Res. 2016;185(3):285–298.
  • Cheema AK, Mehta KY, Fatanmi OO, et al., A Metabolomic and lipidomic serum signature from nonhuman primates administered with a promising radiation countermeasure, gamma-tocotrienol. Int J Mol Sci. 185(1): 79. 2017. .
  • Singh VK, Seed TM. The efficacy and safety of amifostine for the acute radiation syndrome. Expert Opin Drug Saf. 2019;18(11):1077–1090.
  • Carnes BA, Grdina DJ. In Vivo Protection by the Aminothiol WR-2721 against Neutron-induced Carcinogenesis. Int J Radiat Biol. 1992;61(5):567–576.
  • Rasey JS, Spence AM, Badger CC, et al. Specific protection of different normal tissues. Pharmacol Ther. 1988;39(1–3):33–43.
  • Weiss JF. Pharmacologic approaches to protection against radiation-induced lethality and other damage. Environ Health Perspect. 1997;105(Suppl 6):1473–1478.
  • Seed TM, Inal CE, Singh VK. Radioprotection of hematopoietic progenitors by low dose amifostine prophylaxis. Int J Radiat Biol. 2014;90(7):594–604.
  • Cumberland Pharmaceuticals Inc. ETHYOL- amifostine injection, powder, lyophilized, for solution. 2017. Available at: https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/020221s033lbl.pdf [Last accessed 2020 Feb 12]
  • Brizel DM, Wasserman TH, Henke M, et al. Phase III randomized trial of amifostine as a radioprotector in head and neck cancer. J Clin Oncol. 2000;18(19):3339–3345.
  • Schuchter LM. Exploration of platinum-based dose-intensive chemotherapy strategies with amifostine (Ethyol). Eur J Cancer. 1996;32(Suppl 4):S40–2.
  • Ali BH, Al Moundhri MS. Agents ameliorating or augmenting the nephrotoxicity of cisplatin and other platinum compounds: a review of some recent research. Food Chem Toxicol. 2006;44(8):1173–1183.
  • Chirino YI, Pedraza-Chaverri J. Role of oxidative and nitrosative stress in cisplatin-induced nephrotoxicity. Experimental and toxicologic pathology: official journal of the Gesellschaft fur Toxikologische Pathologie. 2009;61(3):223–242.
  • Seed TM. Radiation protectants: current status and future prospects. Health Phys. 2005;89(5):531–545.
  • Singh VK, Fatanmi OO, Wise SY, et al. The potentiation of the radioprotective efficacy of two medical countermeasures, gamma-tocotrienol and amifostine, by a combination prophylactic modality. Radiat Prot Dosimetry. 2016;172(1–3):302–310.
  • Cheema AK, Li Y, Girgis M, et al., Alterations in tissue metabolite profiles with amifostine-prophylaxed mice exposed to gamma radiation. Metabolites. 10(5): 211. 2020. .
  • Singh VK, Santiago PT, Simas M, et al. Acute radiation syndrome: an update on biomarkers for radiation Injury. Journal of Radiation and Cancer Research. 2018;9(4):132–146.
  • Rotolo J, Stancevic B, Zhang J, et al. Anti-ceramide antibody prevents the radiation gastrointestinal syndrome in mice. J Clin Invest. 2012;122(5):1786–1790.
  • Singh VK, Fuller J, Rotolo J, et al. Anti-ceramide antibody as treatment for the acute radiation gastrointestinal syndrome. 3rd International Conference on the Molecular Medicine of Sphingolipids. French Lick Resort, French Lick, Indiana, USA, 2016
  • Singh VK, Fuller J, Rotolo J, et al. Anti-ceramide antibody as treatment for the acute radiation gastrointestinal syndrome. Conference on normal tissue radiation effects and countermeasures. Winthrop Rockfeller Institute, University of Arkansas Medical Sciences, Little Rock, AR, USA, 2018
  • Kiang JG, Jiao W, Cary LH, et al. Wound trauma increases radiation-induced mortality by activation of iNOS pathway and elevation of cytokine concentrations and bacterial infection. Radiat Res. 2010;173(3):319–332.
  • DiCarlo AL, Maher C, Hick JL, et al. Radiation injury after a nuclear detonation: medical consequences and the need for scarce resources allocation. Disaster Med Public Health Prep. 2011;5(Suppl S1):S32–44.
  • DiCarlo AL, Ramakrishnan N, Hatchett RJ. Radiation combined injury: overview of NIAID research. Health Phys. 2010;98(6):863–867.