3,006
Views
56
CrossRef citations to date
0
Altmetric
Review

Nonhuman primates as models for the discovery and development of radiation countermeasures

&
Pages 695-709 | Received 09 Mar 2017, Accepted 24 Apr 2017, Published online: 05 May 2017

References

  • Singh VK, Newman VL, Berg AN, et al. Animal models for acute radiation syndrome drug discovery. Expert Opin Drug Discov. 2015;10:497–517. .
  • Kim JW, Lee DW, Choi WH, et al. Development of a porcine skin injury model and characterization of the dose-dependent response to high-dose radiation. J Radiat Res. 2013;54:823–831.
  • King GL, Rabin BM, Weatherspoon JK. 5-HT3 receptor antagonists ameliorate emesis in the ferret evoked by neutron or proton radiation. Aviat Space Environ Med. 1999;70:485–492.
  • Lankau EW, Turner PV, Mullan RJ, et al. Use of nonhuman primates in research in North America. J Am Assoc Lab Anim Science. 2014;53:278–282.
  • Williams JP, Brown SL, Georges GE, et al. Animal models for medical countermeasures to radiation exposure. Radiat Res. 2010;173:557–578.
  • Williams JP, Jackson IL, Shah JR, et al. Animal models and medical countermeasures development for radiation-induced lung damage: report from an NIAID Workshop. Radiat Res. 2012;177:e0025–39. .
  • Augustine AD, Gondre-Lewis T, McBride W, et al. Animal models for radiation injury, protection and therapy. Radiat Res. 2005;164:100–109.
  • Flynn JL, Capuano SV, Croix D, et al. Non-human primates: a model for tuberculosis research. Tuberculosis (Edinb). 2003;83:116–118.
  • Mahmud N, Pang W, Cobbs C, et al. Studies of the route of administration and role of conditioning with radiation on unrelated allogeneic mismatched mesenchymal stem cell engraftment in a nonhuman primate model. Exp Hematol. 2004;32:494–501.
  • Norol F, Drouet M, Mathieu J, et al. Ex vivo expanded mobilized peripheral blood CD34+ cells accelerate haematological recovery in a baboon model of autologous transplantation. Br J Haematol. 2000;109:162–172.
  • Herodin F, Richard S, Grenier N, et al. Assessment of total- and partial-body irradiation in a baboon model: preliminary results of a kinetic study including clinical, physical, and biological parameters. Health Phys. 2012;103:143–149.
  • Herodin F, Mestries JC, Janodet D, et al. Recombinant glycosylated human interleukin-6 accelerates peripheral blood platelet count recovery in radiation-induced bone marrow depression in baboons. Blood. 1992;80:688–695.
  • Port M, Herodin F, Valente M, et al. MicroRNA expression for early prediction of late occurring hematologic acute radiation syndrome in baboons. PLoS One. 2016;11:e0165307.
  • Port M, Herodin F, Valente M, et al. First generation gene expression signature for early prediction of late occurring hematological acute radiation syndrome in baboons. Radiat Res. 2016;186:39–54.
  • Valente M, Denis J, Grenier N, et al. Revisiting biomarkers of total-body and partial-body exposure in a baboon model of irradiation. PLoS One. 2015;10:e0132194.
  • Port M, Herodin F, Valente M, et al. Gene expression signature for early prediction of late occurring pancytopenia in irradiated baboons. Ann Hematol. Forthcoming 2017.
  • Port M, Herodin F, Valente M, et al. Pre-exposure gene expression in baboons with and without pancytopenia after radiation exposure. Int J Mol Sci. 2017;18:E541.
  • Farese AM, MacVittie TJ. Filgrastim for the treatment of hematopoietic acute radiation syndrome. Drugs Today (Barc). 2015;51:537–548.
  • Hankey KG, Farese AM, Blaauw EC, et al. Pegfilgrastim improves survival of lethally irradiated nonhuman primates. Radiat Res. 2015;183:643–655. .
  • Clayton NP, Charpentier EJJ, LaCasse ER, et al. Sargramostim significantly improved the mortality rate at Day 60 in a non-human primate model of hematopoietic acute radiation syndrome with minimal supportive care when administered 48 h after total body irradiation. 42nd Conference of the European Radiation Reasearch Society; 2016; Amsterdam, Netherlands; p. 190
  • Farese AM, Williams DE, Seiler FR, et al. Combination protocols of cytokine therapy with interleukin-3 and granulocyte-macrophage colony-stimulating factor in a primate model of radiation-induced marrow aplasia. Blood. 1993;82:3012–3018.
  • 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. .
  • Krivokrysenko VI, Toshkov IA, Gleiberman AS, et al. The Toll-like receptor 5 agonist Entolimod mitigates lethal acute radiation syndrome in non-human primates. Plos One. 2015;10:e0135388.
  • Gluzman-Poltorak Z, Mendonca SR, Vainstein V, et al. Randomized comparison of single dose of recombinant human IL-12 versus placebo for restoration of hematopoiesis and improved survival in rhesus monkeys exposed to lethal radiation. J Hematol Oncol. 2014;7:31.
  • Basile LA, Ellefson D, Gluzman-Poltorak Z, et al. HemaMax, a recombinant human interleukin-12, is a potent mitigator of acute radiation injury in mice and non-human primates. Plos One. 2012;7:e30434. .
  • Gluzman-Poltorak Z, Vainstein V, Basile LA. Recombinant interleukin-12, but not granulocyte-colony stimulating factor, improves survival in lethally irradiated nonhuman primates in the absence of supportive care: evidence for the development of a frontline radiation medical countermeasure. Am J Hematol. 2014;89:868–873.
  • Stickney DR, Dowding C, Authier S, et al. 5-androstenediol improves survival in clinically unsupported rhesus monkeys with radiation-induced myelosuppression. Int Immunopharmacol. 2007;7:500–505. .
  • Singh VK, Kulkarni S, Fatanmi OO, et al. Radioprotective efficacy of gamma-tocotrienol in nonhuman primates. Radiat Res. 2016;185:285–298.
  • MacVittie TJ, Gibbs A, Farese AM, et al. AEOL 10150 mitigates radiation-induced lung injury in the nonhuman primate: morbidity and mortality are administration schedule-dependent. Radiat Res. 2017;187:298–318.
  • Carter CL, Jones JW, Barrow K, et al. A MALDI-MSI approach to the characterization of radiation-induced lung injury and medical countermeasure development. Health Phys. 2015;109:466–478.
  • Faller DV, Castaneda SA, Zhou D, et al. An oral HemokineTM, alpha-methylhydrocinnamate, enhances myeloid and neutrophil recovery following irradiation in vivo. Blood Cells Mol Dis. 2017;63:1–8.
  • Herodin F, Roy L, Grenier N, et al. Antiapoptotic cytokines in combination with pegfilgrastim soon after irradiation mitigates myelosuppression in nonhuman primates exposed to high irradiation dose. Exp Hematol. 2007;35:1172–1181.
  • Drouet M, Mourcin F, Grenier N, et al. Single administration of stem cell factor, FLT-3 ligand, megakaryocyte growth and development factor, and interleukin-3 in combination soon after irradiation prevents nonhuman primates from myelosuppression: long-term follow-up of hematopoiesis. Blood. 2004;103:878–885.
  • Zeidler C, Kanz L, Hurkuck F, et al. In vivo effects of interleukin-6 on thrombopoiesis in healthy and irradiated primates. Blood. 1992;80:2740–2745.
  • MacVittie TJ, Farese AM, Patchen ML, et al. Therapeutic efficacy of recombinant interleukin-6 (IL-6) alone and combined with recombinant human IL-3 in a nonhuman primate model of high-dose, sublethal radiation-induced marrow aplasia. Blood. 1994;84:2515–2522.
  • Chen BJ, Deoliveira D, Spasojevic I, et al. Growth hormone mitigates against lethal irradiation and enhances hematologic and immune recovery in mice and nonhuman primates. PLoS One. 2010;5:e11056.
  • Farese AM, Smith WG, Giri JG, et al. Promegapoietin-1a, an engineered chimeric IL-3 and Mpl-L receptor agonist, stimulates hematopoietic recovery in conventional and abbreviated schedules following radiation-induced myelosuppression in nonhuman primates. Stem Cells. 2001;19:329–338.
  • Vasin MV, Semenov LF, Suvorov NN, et al. Protective effect and the therapeutic index of indralin in juvenile rhesus monkeys. J Radiat Res. 2014;55:1048–1055.
  • Laver J, Abboud M, Gasparetto C, et al. Effects of IL-1 on hematopoietic progenitors after myelosuppressive chemoradiotherapy. Biotherapy. 1989;1:293–300.
  • Berenson RJ, Andrews RG, Bensinger WI, et al. Antigen CD34+ marrow cells engraft lethally irradiated baboons. J Clin Invest. 1988;81:951–955.
  • Farese AM, Casey DB, Smith WG, et al. Leridistim, a chimeric dual G-CSF and IL-3 receptor agonist, enhances multilineage hematopoietic recovery in a nonhuman primate model of radiation-induced myelosuppression: effect of schedule, dose, and route of administration. Stem Cells. 2001;19:522–533.
  • Farese AM, Casey DB, Vigneulle RM, et al. A single dose of pegylated leridistim significantly improves neutrophil recovery in sublethally irradiated rhesus macaques. Stem Cells. 2001;19:514–521.
  • MacVittie TJ, Farese AM, Smith WG, et al. Myelopoietin, an engineered chimeric IL-3 and G-CSF receptor agonist, stimulates multilineage hematopoietic recovery in a nonhuman primate model of radiation-induced myelosuppression. Blood. 2000;95:837–845.
  • Farese AM, Hunt P, Grab LB, et al. Combined administration of recombinant human megakaryocyte growth and development factor and granulocyte colony-stimulating factor enhances multilineage hematopoietic reconstitution in nonhuman primates after radiation-induced marrow aplasia. J Clin Invest. 1996;97:2145–2151.
  • Farese AM, Herodin F, McKearn JP, et al. Acceleration of hematopoietic reconstitution with a synthetic cytokine (SC-55494) after radiation-induced bone marrow aplasia. Blood. 1996;87:581–591.
  • MacVittie TJ, Farese AM, Herodin F, et al. Combination therapy for radiation-induced bone marrow aplasia in nonhuman primates using synthokine SC-55494 and recombinant human granulocyte colony-stimulating factor. Blood. 1996;87:4129–4135.
  • Ma YC, Li M, Xing S, et al. [Therapeutic effect of combined cytokines on nonhuman primate model of severe haemopoietic acute radiation sickness]. Zhongguo Shi Yan Xue Ye Xue Za Zhi. 2016;24:573–579.
  • Wagemaker G, Neelis KJ, Hartong SCC, et al. The efficacy of recombinant TPO in murine And nonhuman primate models for myelosuppression and stem cell transplantation. Stem Cells. 1998;16(Suppl 2):127–141.
  • Farese AM, MacVittie TJ, Roskos L, et al. Hematopoietic recovery following autologous bone marrow transplantation in a nonhuman primate: effect of variation in treatment schedule with PEG-rHuMGDF. Stem Cells. 2003;21:79–89.
  • U.S. Food and Drug Administration. Guidance for industry: product developoment under the animal rule. 2015. [cited 2016 Jul 5]. Available from: http://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/UCM399217.pdf
  • Hall EJ, Giaccia AJ. Radiobiology for the radiobiologist. 7th ed. Philadelphia (PA): Lippincott Williams and Wilkins; 2012.
  • Bushberg JT. Radiation exposure and contamination - Merck manual for health care professionals. Rahway (NJ): Merck Publishing Group; 2016.
  • McCann DGC. Radiation poisoning: current concepts in the acute radiation syndrome. Am J Clin Med. 2006;3:13–21.
  • 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. Br J Radiol Suppl. 2005;27:1–8.
  • Hill RP. Radiation effects on the respiratory system. Br J Radiol Suppl. 2005;27:75–81.
  • Moulder JE, Cohen EP. Radiation-induced multi-organ involvement and failure: the contribution of radiation effects on the renal system. Br J Radiol Suppl. 2005;27:82–88.
  • 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;162:711–728. .
  • Dumont F, Le Roux A, Bischoff P. Radiation countermeasure agents: an update. Expert Opin Ther Pat. 2010;20:73–101. .
  • Farese AM, Cohen MV, Katz BP, et al. Filgrastim improves survival in lethally irradiated nonhuman primates. Radiat Res. 2013;179:89–100. .
  • Farese AM, Cohen MV, Katz BP, et al. A nonhuman primate model of the hematopoietic acute radiation syndrome plus medical management. Health Phys. 2012;103:367–382.
  • MacVittie TJ, Farese AM, Bennett A, et al. The acute gastrointestinal subsyndrome of the acute radiation syndrome: a rhesus macaque model. Health Phys. 2012;103:411–426. .
  • Geraci JP, Jackson KL, Mariano MS. The intestinal radiation syndrome: sepsis and endotoxin. Radiat Res. 1985;101:442–450.
  • Carr KE. Effects of radiation damage on intestinal morphology. Int Rev Cytol. 2001;208:1–119.
  • Mason KA, Withers HR, McBride WH, et al. Comparison of the gastrointestinal syndrome after total-body or total-abdominal irradiation. Radiat Res. 1989;117:480–488.
  • Krimsky M, Dagan A, Aptekar L, et al. Assessment of intestinal permeability in rats by permeation of inulin-fluorescein. J Basic Clin Physiol Pharmacol. 2000;11:143–153.
  • Kobayashi T, Ohmori T, Yanai M, et al. The analysis of the defense mechanism against indigenous bacterial translocation in X-irradiated mice. Microbiol Immunol. 1991;35:315–324.
  • Lutgens LC, Blijlevens NM, Deutz NE, et al. Monitoring myeloablative therapy-induced small bowel toxicity by serum citrulline concentration: a comparison with sugar permeability tests. Cancer. 2005;103:191–199.
  • Lang IM, Sarna SK, Condon RE. Gastrointestinal motor correlates of vomiting in the dog: quantification and characterization as an independent phenomenon. Gastroenterology. 1986;90:40–47.
  • Danquechin Dorval E, Mueller GP, Eng RR, et al. Effect of ionizing radiation on gastric secretion and gastric motility in monkeys. Gastroenterology. 1985;89:374–380.
  • Lopez M, Martin M. Medical management of the acute radiation syndrome. Rep Pract Oncol Radiother. 2011;16:138–146.
  • Robbins ME, Bourland JD, Cline JM, et al. A model for assessing cognitive impairment after fractionated whole-brain irradiation in nonhuman primates. Radiat Res. 2011;175:519–525.
  • Hanbury DB, Peiffer AM, Dugan G, et al. Long-term cognitive functioning in single-dose total-body gamma-irradiated rhesus monkeys (Macaca mulatta). Radiat Res. 2016;186:447–454.
  • 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;109:335–341.
  • Travis EL. Organizational response of normal tissues to irradiation. Semin Radiat Oncol. 2001;11:184–196.
  • Marks LB, Yu X, Vujaskovic Z, et al. Radiation-induced lung injury. Semin Radiat Oncol. 2003;13:333–345.
  • Sharplin J, Franko AJ. A quantitative histological study of strain-dependent differences in the effects of irradiation on mouse lung during the intermediate and late phases. Radiat Res. 1989;119:15–31.
  • Sharplin J, Franko AJ. A quantitative histological study of strain-dependent differences in the effects of irradiation on mouse lung during the early phase. Radiat Res. 1989;119:1–14.
  • McLaughlin RF Jr., Tyler WS, Canada RO. Subgross pulmonary anatomy of the rabbit, rat, and guinea pig, with additional notes on the human lung. Am Rev Respir Dis. 1966;94:380–387.
  • Vujaskovic Z, Marks LB, Anscher MS. The physical parameters and molecular events associated with radiation-induced lung toxicity. Semin Radiat Oncol. 2000;10:296–307.
  • Haston CK, Zhou X, Gumbiner-Russo L, et al. Universal and radiation-specific loci influence murine susceptibility to radiation-induced pulmonary fibrosis. Cancer Res. 2002;62:3782–3788.
  • Garofalo M, Bennett A, Farese AM, et al. The delayed pulmonary syndrome following acute high-dose irradiation: a rhesus macaque model. Health Phys. 2014;106:56–72.
  • DiCarlo AL, Jackson IL, Shah JR, et al. Development and licensure of medical countermeasures to treat lung damage resulting from a radiological or nuclear incident. Radiat Res. 2012;177:717–721.
  • Peter RU. Cutaneous radiation syndrome in multi-organ failure. BJR Suppl. 2005;27:180–184.
  • DeBo RJ, Lees CJ, Dugan GO, et al. Late effects of total-body gamma irradiation on cardiac structure and function in male rhesus macaques. Radiat Res. 2016;186:55–64.
  • van Kleef EM, Zurcher C, Oussoren YG, et al. Long-term effects of total-body irradiation on the kidney of Rhesus monkeys. Int J Radiat Biol. 2000;76:641–648.
  • Singh VK, Romaine PL, Newman VL, et al. Medical countermeasures for unwanted CBRN exposures: part II radiological and nuclear threats with review of recent countermeasure patents. Expert Opin Ther Pat. 2016;26:1399–1408.
  • Sanofi-aventis U.S. LLC. Leukine. 2013. [cited 2016 Oct 20]. Available from: http://products.sanofi.us/Leukine/Leukine.html
  • Gupta S. FDA advisory committee meeting: study drug Leukine. 2013. [cited 2016 Oct 2]. Available from: http://www.fda.gov/downloads/advisorycommittees/committeesmeetingmaterials/drugs/medicalimagingdrugsadvisorycommittee/ucm350156.pdf
  • Neumedicines. HemaMax™ (rHuIL-12) for acute radiation syndrome. 2014. [cited 2014 Oct 27]. Available from: http://www.neumedicines.com/
  • Gridley DS, Makinde AY, Luo X, et al. Radiation and a metalloporphyrin radioprotectant in a mouse prostate tumor model. Anticancer Res. 2007;27:3101–3109.
  • Pearlstein RD, Higuchi Y, Moldovan M, et al. Metalloporphyrin antioxidants ameliorate normal tissue radiation damage in rat brain. Int J Radiat Biol. 2010;86:145–163.
  • Zhang Y, Zhang X, Rabbani ZN, et al. Oxidative stress mediates radiation lung injury by inducing apoptosis. Int J Radiat Oncol Biol Phys. 2012;83:740–748.
  • Batinic-Haberle I, Tovmasyan A, Roberts E, et al. SOD therapeutics: latest insights into their structure-activity relationships and impact upon the cellular redox-based pathways. Antioxid Redox Signal. 2013;20:2372–2415.
  • Orrell RW. AEOL-10150 (Aeolus). Curr Opin Investig Drugs. 2006;7:70–80.
  • Garofalo MC, Ward AA, Farese AM, et al. A pilot study in rhesus macaques to assess the treatment efficacy of a small molecular weight catalytic metalloporphyrin antioxidant (AEOL 10150) in mitigating radiation-induced lung damage. Health Phys. 2014;106:73–83.
  • Stickney DR, Dowding C, Garsd A, et al. 5-androstenediol stimulates multilineage hematopoiesis in rhesus monkeys with radiation-induced myelosuppression. Int Immunopharmacol. 2006;6:1706–1713.
  • Singh VK, Newman VL, Romaine PL, et al. Radiation countermeasure agents: an update (2011-2014). Expert Opin Ther Pat. 2014;24:1229–1255. .
  • Vasin MV, Antipov VV, Chernov GA, et al. Studies of the radiation-protective effects of indralin on the hematopoietic system of different species of animals. Radiats Biol Radioecol. 1996;36:168–189.
  • 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. .
  • U.S. Food and Drug Administration. Table of pharmacogenomic biomarkers in drug labeling. 2015. [cited 2015 Oct 25]. Available from: http://www.fda.gov/drugs/scienceresearch/researchareas/pharmacogenetics/ucm083378.htm
  • European Medicines Agency. Qualification of novel methodologies for medicine development. 2015. [cited 2015 Oct 25]. Available from: http://www.ema.europa.eu/ema/index.jsp?curl=pages/regulation/document_listing/document_listing_000319.jsp&mid=WC0b01ac0580022bb0
  • Pharmaceutical and Medical Devices Agency. Record of consultations on pharmacogenomics/biomarkers. 2010. [cited 2015 Oct 25]. Available from: https://www.pmda.go.jp/english/review-services/consultations/0001.html
  • Hu S, Blakely WF, Cucinotta FA. HEMODOSE: a biodosimetry tool based on multi-type blood cell counts. Health Phys. 2015;109:54–68.
  • 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;71:22–37.
  • Gluzman-Poltorak Z, Vainstein V, Basile LA. Association of hematological nadirs and survival in a nonhuman primate model of hematopoietic syndrome of acute radiation syndrome. Radiat Res. 2015;184:226–230.
  • Blakely WF, Ossetrova NI, Whitnall MH, et al. Multiple parameter radiation injury assessment using a nonhuman primate radiation model-biodosimetry applications. Health Phys. 2010;98:153–159.
  • Blakely WF, Madrid JP, Sandgren DJ. Biodosimetry medical recording-use of the biodosimetry assessment tool. Health Phys. 2010;99(Suppl 5):S184–191.
  • Singh VK, Fatanmi OO, Singh PK, et al. Role of radiation-induced granulocyte colony-stimulating factor in recovery from whole body gamma-irradiation. Cytokine. 2012;58:406–414.
  • 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.
  • Redon CE, Nakamura AJ, Martin OA, et al. Recent developments in the use of gamma-H2AX as a quantitative DNA double-strand break biomarker. Aging (Albany NY). 2011;3:168–174.
  • Ha CT, Li X, Fu D, et al. Circulating IL-18 binding protein (IL-18BP) and IL-18 as dual biomarkers of total-body irradiation in mice. Radiat Res. 2016;185:375–383.
  • Xiao M. The role of proinflammatory cytokine interleukin-18 in radiation injury. Health Phys. 2016;111:212–217.
  • Ha CT, Li X-H, Fu D, et al. Circulating interleukin-18 as a biomarker of total-body radiation exposure in mice, minipigs, and nonhuman primates (NHP). PLoS One. 2014;9:e109249.
  • Moroni M, Lombardini E, Salber R, et al. Hematological changes as prognostic indicators of survival: similarities between Gottingen minipigs, humans, and other large animal models. PLoS One. 2011;6:e25210.
  • 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;186:71–78.
  • Jones JW, Bennett A, Carter CL, et al. Citrulline as a biomarker in the non-human primate total- and partial-body irradiation models: correlation of circulating citrulline to acute and prolonged gastrointestinal injury. Health Phys. 2015;109:440–451.
  • 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;184:545–553.
  • Jones JW, Tudor G, Bennett A, et al. Development and validation of a LC-MS/MS assay for quantitation of plasma citrulline for application to animal models of the acute radiation syndrome across multiple species. Anal Bioanal Chem. 2014;406:4663–4675.
  • Jones JW, Scott AJ, Tudor G, et al. Identification and quantitation of biomarkers for radiation-induced injury via mass spectrometry. Health Phys. 2014;106:106–119.
  • Cortez MA, Bueso-Ramos C, Ferdin J, et al. MicroRNAs in body fluids–the mix of hormones and biomarkers. Nat Rev Clin Oncol. 2011;8:467–477.
  • Chim SS, Shing TK, Hung EC, et al. Detection and characterization of placental microRNAs in maternal plasma. Clin Chem. 2008;54:482–490.
  • Menon N, Rogers CJ, Lukaszewicz AI, et al. Detection of acute radiation sickness: a feasibility study in non-human primates circulating miRNAs for triage in radiological events. PLoS One. 2016;11:e0167333.
  • 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.
  • Zhang C, Peng G. Non-coding RNAs: an emerging player in DNA damage response. Mutation Res Rev Mutat Res. 2015;763:202–211.
  • Guo L, Zhao Y, Yang S, et al. An integrated evolutionary analysis of miRNA-lncRNA in mammals. Mol Biol Rep. 2014;41:201–207.
  • Rana S, Kumar R, Sultana S, et al. Radiation-induced biomarkers for the detection and assessment of absorbed radiation doses. J Pharm Bioallied Sci. 2010;2:189–196.
  • Swartz HM, Williams BB, Zaki BI, et al. Clinical EPR: unique opportunities and some challenges. Acad Radiol. 2014;21:197–206.
  • Swartz HM, Williams BB, Flood AB. Overview of the principles and practice of biodosimetry. Radiat Environ Biophys. 2014;53:221–232.
  • Byrum SD, Burdine MS, Orr L, et al. Time- and radiation-dose dependent changes in the plasma proteome after total body irradiation of non-human primates: implications for biomarker selection. PLoS One. 2017;12:e0174771.
  • Byrum SD, Burdine MS, Orr L, et al. A quantitative proteomic analysis of urine from gamma-irradiated nonhuman primates. J Proteomics Bioinform. 2016;9.
  • 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. Forthcoming 2017.
  • Pannkuk EL, Laiakis EC, Mak TD, et al. A lipidomic and metabolomic serum signature from nonhuman primates exposed to ionizing radiation. Metabolomics. 2016;12:pii: 80.
  • 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 Advances. 2016;6:51192–51202.
  • Laiakis EC, Pannkuk EL, Diaz-Rubio ME, et al. Implications of genotypic differences in the generation of a urinary metabolomics radiation signature. Mutat Res. 2016;788:41–49.
  • Pannkuk EL, Laiakis EC, Authier S, et al. Global metabolomic identification of long-term dose-dependent urinary biomarkers in nonhuman primates exposed to ionizing radiation. Radiat Res. 2015;184:121–133.
  • Chen Z, Coy SL, Pannkuk EL, et al. Rapid and high-throughput detection and quantitation of radiation biomarkers in human and nonhuman primates by differential mobility spectrometry-mass spectrometry. J Am Soc Mass Spectrom. 2016;27:1626–1636.
  • Pannkuk EL, Fornace AJ Jr., Laiakis EC. Metabolomic applications in radiation biodosimetry: exploring radiation effects through small molecules. Int J Radiat Biol. 2017;1–26.
  • Johnson CH, Gonzalez FJ. Challenges and opportunities of metabolomics. J Cell Physiol. 2012;227:2975–2981.
  • Johnson CH, Patterson AD, Krausz KW, et al. Radiation metabolomics. 5. Identification of urinary biomarkers of ionizing radiation exposure in nonhuman primates by mass spectrometry-based metabolomics. Radiat Res. 2012;178:328–340.
  • Goudarzi M, Mak TD, Jacobs JP, et al. An Integrated multi-omic approach to assess radiation injury on the host-microbiome axis. Radiat Res. 2016;186:219–234.
  • Amundson SA, Do KT, Shahab S, et al. Identification of potential mRNA biomarkers in peripheral blood lymphocytes for human exposure to ionizing radiation. Radiat Res. 2000;154:342–346.
  • Ahlem CN, White SK, Page TM, et al. Differential metabolism of androst-5-ene-3beta,17beta-diol between rats, canines, monkeys and humans. Steroids. 2011;76:669–674.
  • Amgen Inc. Neupogen (filgrastim) injection for subcutaneous or intravenous use. 2015. [cited 2015 Apr 2]. Available from: http://pi.amgen.com/united_states/neupogen/neupogen_pi_hcp_english.pdf
  • Amgen Inc. Neulasta (pegfilgrastim) injection for subcutaneous use. 2015. [cited 2015 Nov 19]. Available from: http://pi.amgen.com/united_states/neulasta/neulasta_pi_hcp_english.pdf
  • Johnke RM, Sattler JA, Allison RR. Radioprotective agents for radiation therapy: future trends. Future Oncol. 2014;10:2345–2357.