DOI QR코드

DOI QR Code

Mechanisms of radiation-induced normal tissue toxicity and implications for future clinical trials

  • Kim, Jae Ho (Department of Radiation Oncology, Henry Ford Health System) ;
  • Jenrow, Kenneth A. (Department of Radiation Oncology, Henry Ford Health System) ;
  • Brown, Stephen L. (Department of Radiation Oncology, Henry Ford Health System)
  • Received : 2014.08.01
  • Accepted : 2014.08.18
  • Published : 2014.09.30

Abstract

To summarize current knowledge regarding mechanisms of radiation-induced normal tissue injury and medical countermeasures available to reduce its severity. Advances in radiation delivery using megavoltage and intensity-modulated radiation therapy have permitted delivery of higher doses of radiation to well-defined tumor target tissues. Injury to critical normal tissues and organs, however, poses substantial risks in the curative treatment of cancers, especially when radiation is administered in combination with chemotherapy. The principal pathogenesis is initiated by depletion of tissue stem cells and progenitor cells and damage to vascular endothelial microvessels. Emerging concepts of radiation-induced normal tissue toxicity suggest that the recovery and repopulation of stromal stem cells remain chronically impaired by long-lived free radicals, reactive oxygen species, and pro-inflammatory cytokines/chemokines resulting in progressive damage after radiation exposure. Better understanding the mechanisms mediating interactions among excessive generation of reactive oxygen species, production of pro-inflammatory cytokines and activated macrophages, and role of bone marrow-derived progenitor and stem cells may provide novel insight on the pathogenesis of radiation-induced injury of tissues. Further understanding the molecular signaling pathways of cytokines and chemokines would reveal novel targets for protecting or mitigating radiation injury of tissues and organs.

Keywords

References

  1. Otsuka S, Coderre JA, Micca PL, et al. Depletion of neural precursor cells after local brain irradiation is due to radiation dose to the parenchyma, not the vasculature. Radiat Res 2006;165:582-91. https://doi.org/10.1667/RR3539.1
  2. Schuller BW, Binns PJ, Riley KJ, Ma L, Hawthorne MF, Coderre JA. Selective irradiation of the vascular endothelium has no effect on the survival of murine intestinal crypt stem cells. Proc Natl Acad Sci U S A 2006;103:3787-92. https://doi.org/10.1073/pnas.0600133103
  3. Schuller BW, Rogers AB, Cormier KS, et al. No significant endothelial apoptosis in the radiation-induced gastrointestinal syndrome. Int J Radiat Oncol Biol Phys 2007;68:205-10. https://doi.org/10.1016/j.ijrobp.2006.12.069
  4. Rhee SG, Chae HZ, Kim K. Peroxiredoxins: a historical overview and speculative preview of novel mechanisms and emerging concepts in cell signaling. Free Radic Biol Med 2005;38:1543-52. https://doi.org/10.1016/j.freeradbiomed.2005.02.026
  5. Ehrhart EJ, Segarini P, Tsang ML, Carroll AG, Barcellos-Hoff MH. Latent transforming growth factor beta1 activation in situ: quantitative and functional evidence after low-dose gamma-irradiation. FASEB J 1997;11:991-1002.
  6. Martin M, Lefaix J, Delanian S. TGF-beta1 and radiation fibrosis: a master switch and a specific therapeutic target? Int J Radiat Oncol Biol Phys 2000;47:277-90. https://doi.org/10.1016/S0360-3016(00)00435-1
  7. Anscher MS, Marks LB, Shafman TD, et al. Risk of long-term complications after TFG-beta1-guided very-high-dose thoracic radiotherapy. Int J Radiat Oncol Biol Phys 2003;56:988-95. https://doi.org/10.1016/S0360-3016(03)00184-6
  8. Flanders KC, Sullivan CD, Fujii M, et al. Mice lacking Smad3 are protected against cutaneous injury induced by ionizing radiation. Am J Pathol 2002;160:1057-68. https://doi.org/10.1016/S0002-9440(10)64926-7
  9. Janko M, Ontiveros F, Fitzgerald TJ, Deng A, DeCicco M, Rock KL. IL-1 generated subsequent to radiation-induced tissue injury contributes to the pathogenesis of radiodermatitis. Radiat Res 2012;178:166-72. https://doi.org/10.1667/RR3097.1
  10. Nawroth I, Alsner J, Behlke MA, et al. Intraperitoneal administration of chitosan/DsiRNA nanoparticles targeting TNF${\alpha}$ prevents radiation-induced fibrosis. Radiother Oncol 2010;97: 143-8. https://doi.org/10.1016/j.radonc.2010.09.010
  11. Lyubimova N, Hopewell JW. Experimental evidence to support the hypothesis that damage to vascular endothelium plays the primary role in the development of late radiation-induced CNS injury. Br J Radiol 2004;77:488-92. https://doi.org/10.1259/bjr/15169876
  12. Li YQ, Ballinger JR, Nordal RA, Su ZF, Wong CS. Hypoxia in radiation-induced blood-spinal cord barrier breakdown. Cancer Res 2001;61:3348-54.
  13. Nordal RA, Nagy A, Pintilie M, Wong CS. Hypoxia and hypoxiainducible factor-1 target genes in central nervous system radiation injury: a role for vascular endothelial growth factor. Clin Cancer Res 2004;10:3342-53. https://doi.org/10.1158/1078-0432.CCR-03-0426
  14. Gonzalez J, Kumar AJ, Conrad CA, Levin VA. Effect of bevacizumab on radiation necrosis of the brain. Int J Radiat Oncol Biol Phys 2007;67:323-6. https://doi.org/10.1016/j.ijrobp.2006.10.010
  15. Patan S. Vasculogenesis and angiogenesis. Cancer Treat Res 2004;117:3-32. https://doi.org/10.1007/978-1-4419-8871-3_1
  16. Bastianutto C, Mian A, Symes J, et al. Local radiotherapy induces homing of hematopoietic stem cells to the irradiated bone marrow. Cancer Res 2007;67:10112-6. https://doi.org/10.1158/0008-5472.CAN-07-2192
  17. Burrell K, Hill RP, Zadeh G. High-resolution in-vivo analysis of normal brain response to cranial irradiation. PLoS One 2012;7:e38366. https://doi.org/10.1371/journal.pone.0038366
  18. Kim JH, Kolozsvary AJ, Jenrow KA, Brown SL. Mechanisms of radiation-induced skin injury and implications for future clinical trials. Int J Radiat Biol 2013;89:311-8. https://doi.org/10.3109/09553002.2013.765055
  19. Thanasegaran S, Ito S, Nishio N, Uddin MN, Sun Y, Isobe KI. Recruitment of Gr1+CD11b+F4/80+ Population in the Bone Marrow and Spleen by Irradiation-Induced Pulmonary Damage. Inflammation. 2014 Jul 10 [Epub]. http://dx.doi.org/10.1007/s10753-014-9952-8.
  20. Mantovani A. Macrophage diversity and polarization: in vivo veritas. Blood 2006;108:408-9. https://doi.org/10.1182/blood-2006-05-019430
  21. Jaal J, Richter C, Dorr W. Effect of recombinant human keratinocyte growth factor ($\Delta$23rHuKGF, Palifermin) on inflammatory and immune changes in mouse tongue during fractionated irradiation. Int J Radiat Biol 2010;86:860-6. https://doi.org/10.3109/09553002.2010.487025
  22. Jenrow KA, Brown SL, Kolozsvary AJ, Lapanowski K, Kim JH. Time-dependent inhibition of pan-inflammatory cytokines mitigates radiation-induced skin injury in mice. Radiat Res 2014;182:316-21. https://doi.org/10.1667/RR13711.1
  23. Yarnold J, Brotons MC. Pathogenetic mechanisms in radiation fibrosis. Radiother Oncol 2010;97:149-61. https://doi.org/10.1016/j.radonc.2010.09.002
  24. Ahmed EA, Agay D, Schrock G, Drouet M, Meineke V, Scherthan H. Persistent DNA damage after high dose in vivo gamma exposure of minipig skin. PLoS One 2012;7:e39521. https://doi.org/10.1371/journal.pone.0039521
  25. Wang J, Zheng H, Ou X, et al. Hirudin ameliorates intestinal radiation toxicity in the rat: support for thrombin inhibition as strategy to minimize side-effects after radiation therapy and as countermeasure against radiation exposure. J Thromb Haemost 2004;2:2027-35. https://doi.org/10.1111/j.1538-7836.2004.00960.x
  26. Paris F, Fuks Z, Kang A, et al. Endothelial apoptosis as the primary lesion initiating intestinal radiation damage in mice. Science 2001;293:293-7. https://doi.org/10.1126/science.1060191
  27. Sekine I, Sumi M, Ito Y, et al. Retrospective analysis of steroid therapy for radiation-induced lung injury in lung cancer patients. Radiother Oncol 2006;80:93-7. https://doi.org/10.1016/j.radonc.2006.06.007
  28. Wynn TA. Cellular and molecular mechanisms of fibrosis. J Pathol 2008;214:199-210. https://doi.org/10.1002/path.2277
  29. Yarnold J, Brotons MC. Pathogenetic mechanisms in radiation fibrosis. Radiother Oncol 2010;97:149-61. https://doi.org/10.1016/j.radonc.2010.09.002
  30. Beinert T, Binder D, Oehm C, et al. Further evidence for oxidant-induced vascular endothelial growth factor upregulation in the bronchoalveolar lavage fluid of lung cancer patients undergoing radio-chemotherapy. J Cancer Res Clin Oncol 2000;126:352-6. https://doi.org/10.1007/s004320050355
  31. Fleckenstein K, Gauter-Fleckenstein B, Jackson IL, Rabbani Z, Anscher M, Vujaskovic Z. Using biological markers to predict risk of radiation injury. Semin Radiat Oncol 2007;17:89-98. https://doi.org/10.1016/j.semradonc.2006.11.004
  32. Waghray M, Cui Z, Horowitz JC, et al. Hydrogen peroxide is a diffusible paracrine signal for the induction of epithelial cell death by activated myofibroblasts. FASEB J 2005;19:854-6.
  33. Macartney G, Harrison MB, VanDenKerkhof E, Stacey D, McCarthy P. Quality of life and symptoms in pediatric brain tumor survivors: a systematic review. J Pediatr Oncol Nurs 2014;31:65-77. https://doi.org/10.1177/1043454213520191
  34. Mizumatsu S, Monje ML, Morhardt DR, Rola R, Palmer TD, Fike JR. Extreme sensitivity of adult neurogenesis to low doses of X-irradiation. Cancer Res 2003;63:4021-7.
  35. Panagiotakos G, Alshamy G, Chan B, et al. Long-term impact of radiation on the stem cell and oligodendrocyte precursors in the brain. PLoS One 2007;2:e588. https://doi.org/10.1371/journal.pone.0000588
  36. Snyder JS, Hong NS, McDonald RJ, Wojtowicz JM. A role for adult neurogenesis in spatial long-term memory. Neuroscience 2005;130:843-52. https://doi.org/10.1016/j.neuroscience.2004.10.009
  37. Kermen F, Sultan S, Sacquet J, Mandairon N, Didier A. Consolidation of an olfactory memory trace in the olfactory bulb is required for learning-induced survival of adult-born neurons and long-term memory. PLoS One 2010;5:e12118. https://doi.org/10.1371/journal.pone.0012118
  38. Jessberger S, Toni N, Clemenson GD Jr, Ray J, Gage FH. Directed differentiation of hippocampal stem/progenitor cells in the adult brain. Nat Neurosci 2008;11:888-93. https://doi.org/10.1038/nn.2148
  39. Toni N, Laplagne DA, Zhao C, et al. Neurons born in the adult dentate gyrus form functional synapses with target cells. Nat Neurosci 2008;11:901-7. https://doi.org/10.1038/nn.2156
  40. Toni N, Teng EM, Bushong EA, et al. Synapse formation on neurons born in the adult hippocampus. Nat Neurosci 2007;10:727-34. https://doi.org/10.1038/nn1908
  41. Snyder JS, Ferrante SC, Cameron HA. Late maturation of adult-born neurons in the temporal dentate gyrus. PLoS One 2012;7:e48757. https://doi.org/10.1371/journal.pone.0048757
  42. Guzowski JF, Lyford GL, Stevenson GD, et al. Inhibition of activity-dependent arc protein expression in the rat hippocampus impairs the maintenance of long-term potentiation and the consolidation of long-term memory. J Neurosci 2000;20:3993-4001.
  43. Rola R, Raber J, Rizk A, et al. Radiation-induced impairment of hippocampal neurogenesis is associated with cognitive deficits in young mice. Exp Neurol 2004;188:316-30. https://doi.org/10.1016/j.expneurol.2004.05.005
  44. Rosi S, Andres-Mach M, Fishman KM, Levy W, Ferguson RA, Fike JR. Cranial irradiation alters the behaviorally induced immediate-early gene arc (activity-regulated cytoskeletonassociated protein). Cancer Res 2008;68:9763-70. https://doi.org/10.1158/0008-5472.CAN-08-1861
  45. Rosi S, Ramirez-Amaya V, Vazdarjanova A, Worley PF, Barnes CA, Wenk GL. Neuroinflammation alters the hippocampal pattern of behaviorally induced Arc expression. J Neurosci 2005;25:723-31. https://doi.org/10.1523/JNEUROSCI.4469-04.2005
  46. Rosi S, Ramirez-Amaya V, Vazdarjanova A, et al. Accuracy of hippocampal network activity is disrupted by neuroinflammation: rescue by memantine. Brain 2009;132(Pt 9):2464-77. https://doi.org/10.1093/brain/awp148
  47. Jenrow KA, Brown SL, Lapanowski K, Naei H, Kolozsvary A, Kim JH. Selective inhibition of microglia-mediated neuroinflammation mitigates radiation-induced cognitive impairment. Radiat Res 2013;179:549-56. https://doi.org/10.1667/RR3026.1
  48. Zhao W, Payne V, Tommasi E, Diz DI, Hsu FC, Robbins ME. Administration of the peroxisomal proliferator-activated receptor gamma agonist pioglitazone during fractionated brain irradiation prevents radiation-induced cognitive impairment. Int J Radiat Oncol Biol Phys 2007;67:6-9. https://doi.org/10.1016/j.ijrobp.2006.09.036
  49. Allen AR, Eilertson K, Sharma S, et al. Effects of radiation combined injury on hippocampal function are modulated in mice deficient in chemokine receptor 2 (CCR2). Radiat Res 2013;180:78-88. https://doi.org/10.1667/RR3344.1
  50. Acharya MM, Martirosian V, Christie LA, Limoli CL. Long-term cognitive effects of human stem cell transplantation in the irradiated brain. Int J Radiat Biol 2014;90:816-20. https://doi.org/10.3109/09553002.2014.927934
  51. Abayomi OK. Pathogenesis of cognitive decline following therapeutic irradiation for head and neck tumors. Acta Oncol 2002;41:346-51. https://doi.org/10.1080/028418602760169389
  52. Butler JM, Rapp SR, Shaw EG. Managing the cognitive effects of brain tumor radiation therapy. Curr Treat Options Oncol 2006;7:517-23. https://doi.org/10.1007/s11864-006-0026-5
  53. Singer BH, Gamelli AE, Fuller CL, Temme SJ, Parent JM, Murphy GG. Compensatory network changes in the dentate gyrus restore long-term potentiation following ablation of neurogenesis in young-adult mice. Proc Natl Acad Sci U S A 2011;108:5437-42. https://doi.org/10.1073/pnas.1015425108
  54. Liang L, Hu D, Liu W, Williams JP, Okunieff P, Ding I. Celecoxib reduces skin damage after radiation: selective reduction of chemokine and receptor mRNA expression in irradiated skin but not in irradiated mammary tumor. Am J Clin Oncol 2003;26:S114-21.
  55. Delanian S, Lefaix JL. Current management for late normal tissue injury: radiation-induced fibrosis and necrosis. Semin Radiat Oncol 2007;17:99-107. https://doi.org/10.1016/j.semradonc.2006.11.006
  56. Lefaix JL, Delanian S, Vozenin MC, Leplat JJ, Tricaud Y, Martin M. Striking regression of subcutaneous fibrosis induced by high doses of gamma rays using a combination of pentoxifylline and alpha-tocopherol: an experimental study. Int J Radiat Oncol Biol Phys 1999;43:839-47. https://doi.org/10.1016/S0360-3016(98)00419-2
  57. Delanian S, Porcher R, Balla-Mekias S, Lefaix JL. Randomized, placebo-controlled trial of combined pentoxifylline and tocopherol for regression of superficial radiation-induced fibrosis. J Clin Oncol 2003;21:2545-50. https://doi.org/10.1200/JCO.2003.06.064
  58. ClinicalTrials.gov. Trial of combined pentoxifylline-tocopherolclodronate vs placebo in radiation-induced brachial plexopathy (PENTOCLO) [Internet]. Washington, DC: US National Institute of Health; c2014 [cited 2014 Aug 15]. Available from: http://clinicaltrials.gov/ct2/show/NCT01291433.
  59. Singh VK, Brown DS, Kao TC. Alpha-tocopherol succinate protects mice from gamma-radiation by induction of granulocyte-colony stimulating factor. Int J Radiat Biol 2010;86:12-21. https://doi.org/10.3109/09553000903264515
  60. Kulkarni S, Singh PK, Ghosh SP, Posarac A, Singh VK. Granulocyte colony-stimulating factor antibody abrogates radioprotective efficacy of gamma-tocotrienol, a promising radiation countermeasure. Cytokine 2013;62:278-85. https://doi.org/10.1016/j.cyto.2013.03.009
  61. Singh VK, Beattie LA, Seed TM. Vitamin E: tocopherols and tocotrienols as potential radiation countermeasures. J Radiat Res 2013;54:973-88. https://doi.org/10.1093/jrr/rrt048
  62. Xavier S, Piek E, Fujii M, et al. Amelioration of radiationinduced fibrosis: inhibition of transforming growth factorbeta signaling by halofuginone. J Biol Chem 2004;279:15167-76. https://doi.org/10.1074/jbc.M309798200
  63. Avraham T, Yan A, Zampell JC, et al. Radiation therapy causes loss of dermal lymphatic vessels and interferes with lymphatic function by TGF-beta1-mediated tissue fibrosis. Am J Physiol Cell Physiol 2010;299:C589-605. https://doi.org/10.1152/ajpcell.00535.2009
  64. Lee JW, Tutela JP, Zoumalan RA, et al. Inhibition of Smad3 expression in radiation-induced fibrosis using a novel method for topical transcutaneous gene therapy. Arch Otolaryngol Head Neck Surg 2010;136:714-9. https://doi.org/10.1001/archoto.2010.107
  65. Brown SL, Kolozsvary A, Jenrow KA, Lapanowski K, Kim JH. TNF alpha receptor antagonist mitigates radiation induced acute and sub-acute skin injury in mice (poster PS4-47). In: Presented at the 58th Annual Meeting of the Radiation Research Society; 2012 Sep 29-Oct 4; Puerto Rico.
  66. Delanian S, Baillet F, Huart J, Lefaix JL, Maulard C, Housset M. Successful treatment of radiation-induced fibrosis using liposomal Cu/Zn superoxide dismutase: clinical trial. Radiother Oncol 1994;32:12-20. https://doi.org/10.1016/0167-8140(94)90444-8
  67. Campana F, Zervoudis S, Perdereau B, et al. Topical superoxide dismutase reduces post-irradiation breast cancer fibrosis. J Cell Mol Med 2004;8:109-16. https://doi.org/10.1111/j.1582-4934.2004.tb00265.x
  68. Rabbani ZN, Anscher MS, Folz RJ, et al. Overexpression of extracellular superoxide dismutase reduces acute radiation induced lung toxicity. BMC Cancer 2005;5:59. https://doi.org/10.1186/1471-2407-5-59
  69. Rosenthal RA, Fish B, Hill RP, et al. Salen Mn complexes mitigate radiation injury in normal tissues. Anticancer Agents Med Chem 2011;11:359-72. https://doi.org/10.2174/187152011795677490
  70. Yan S, Brown SL, Kolozsvary A, Freytag SO, Lu M, Kim JH. Mitigation of radiation-induced skin injury by AAV2-mediated MnSOD gene therapy. J Gene Med 2008;10:1012-8. https://doi.org/10.1002/jgm.1226
  71. Chan AW, Bhatt DL, Chew DP, et al. Relation of inflammation and benefit of statins after percutaneous coronary interventions. Circulation 2003;107:1750-6. https://doi.org/10.1161/01.CIR.0000060541.18923.E9
  72. Shishehbor MH, Brennan ML, Aviles RJ, et al. Statins promote potent systemic antioxidant effects through specific inflammatory pathways. Circulation 2003;108:426-31. https://doi.org/10.1161/01.CIR.0000080895.05158.8B
  73. Chen J, Zhang ZG, Li Y, et al. Statins induce angiogenesis, neurogenesis, and synaptogenesis after stroke. Ann Neurol 2003;53:743-51. https://doi.org/10.1002/ana.10555
  74. Lu D, Goussev A, Chen J, et al. Atorvastatin reduces neurological deficit and increases synaptogenesis, angiogenesis, and neuronal survival in rats subjected to traumatic brain injury. J Neurotrauma 2004;21:21-32. https://doi.org/10.1089/089771504772695913
  75. Williams JP, Hernady E, Johnston CJ, et al. Effect of administration of lovastatin on the development of late pulmonary effects after whole-lung irradiation in a murine model. Radiat Res 2004;161:560-7. https://doi.org/10.1667/RR3168
  76. Haydont V, Gilliot O, Rivera S, et al. Successful mitigation of delayed intestinal radiation injury using pravastatin is not associated with acute injury improvement or tumor protection. Int J Radiat Oncol Biol Phys 2007;68:1471-82. https://doi.org/10.1016/j.ijrobp.2007.03.044
  77. Wang J, Boerma M, Fu Q, Kulkarni A, Fink LM, Hauer- Jensen M. Simvastatin ameliorates radiation enteropathy development after localized, fractionated irradiation by a protein C-independent mechanism. Int J Radiat Oncol Biol Phys 2007;68:1483-90. https://doi.org/10.1016/j.ijrobp.2007.03.036
  78. Jenrow KA, Liu J, Brown SL, Kolozsvary A, Lapanowski K, Kim JH. Combined atorvastatin and ramipril mitigate radiationinduced impairment of dentate gyrus neurogenesis. J Neurooncol 2011;101:449-56. https://doi.org/10.1007/s11060-010-0282-x
  79. Liu YH, Yang XP, Sharov VG, et al. Effects of angiotensinconverting enzyme inhibitors and angiotensin II type 1 receptor antagonists in rats with heart failure: role of kinins and angiotensin II type 2 receptors. J Clin Invest 1997;99:1926-35. https://doi.org/10.1172/JCI119360
  80. Robbins ME, Diz DI. Pathogenic role of the renin-angiotensin system in modulating radiation-induced late effects. Int J Radiat Oncol Biol Phys 2006;64:6-12. https://doi.org/10.1016/j.ijrobp.2005.08.033
  81. Ward WF, Kim YT, Molteni A, Solliday NH. Radiation-induced pulmonary endothelial dysfunction in rats: modification by an inhibitor of angiotensin converting enzyme. Int J Radiat Oncol Biol Phys 1988;15:135-40. https://doi.org/10.1016/0360-3016(88)90357-4
  82. Moulder JE, Fish BL, Cohen EP. Treatment of radiation nephropathy with ACE inhibitors. Int J Radiat Oncol Biol Phys 1993;27:93-9. https://doi.org/10.1016/0360-3016(93)90425-U
  83. Moulder JE, Fish BL, Cohen EP. Radiation nephropathy is treatable with an angiotensin converting enzyme inhibitor or an angiotensin II type-1 (AT1) receptor antagonist. Radiother Oncol 1998;46:307-15. https://doi.org/10.1016/S0167-8140(97)00175-8
  84. Moulder JE, Fish BL, Regner KR, Cohen EP. Angiotensin II blockade reduces radiation-induced proliferation in experimental radiation nephropathy. Radiat Res 2002;157:393-401. https://doi.org/10.1667/0033-7587(2002)157[0393:AIBRRI]2.0.CO;2
  85. Kim JH, Brown SL, Kolozsvary A, et al. Modification of radiation injury by ramipril, inhibitor of angiotensin-converting enzyme, on optic neuropathy in the rat. Radiat Res 2004;161:137-42. https://doi.org/10.1667/RR3124
  86. Ryu S, Kolozsvary A, Jenrow KA, Brown SL, Kim JH. Mitigation of radiation-induced optic neuropathy in rats by ACE inhibitor ramipril: importance of ramipril dose and treatment time. J Neurooncol 2007;82:119-24. https://doi.org/10.1007/s11060-006-9256-4
  87. Jenrow KA, Brown SL, Liu J, Kolozsvary A, Lapanowski K, Kim JH. Ramipril mitigates radiation-induced impairment of neurogenesis in the rat dentate gyrus. Radiat Oncol 2010;5:6. https://doi.org/10.1186/1748-717X-5-6
  88. Hill RP, Rodemann HP, Hendry JH, Roberts SA, Anscher MS. Normal tissue radiobiology: from the laboratory to the clinic. Int J Radiat Oncol Biol Phys 2001;49:353-65. https://doi.org/10.1016/S0360-3016(00)01484-X
  89. Francois S, Mouiseddine M, Mathieu N, et al. Human mesenchymal stem cells favour healing of the cutaneous radiation syndrome in a xenogenic transplant model. Ann Hematol 2007;86:1-8.
  90. Lombaert IM, Wierenga PK, Kok T, Kampinga HH, deHaan G, Coppes RP. Mobilization of bone marrow stem cells by granulocyte colony-stimulating factor ameliorates radiationinduced damage to salivary glands. Clin Cancer Res 2006;12:1804-12. https://doi.org/10.1158/1078-0432.CCR-05-2381
  91. Kim JH, Kolozsvary A, Jenrow KA, Brown SL. Plerixafor, a CXCR4 antagonist, mitigates skin radiation-induced injury in mice. Radiat Res 2012;178:202-6. https://doi.org/10.1667/RR2886.1
  92. Donahue RE, Jin P, Bonifacino AC, et al. Plerixafor (AMD3100) and granulocyte colony-stimulating factor (G-CSF) mobilize different CD34+ cell populations based on global gene and microRNA expression signatures. Blood 2009;114:2530-41. https://doi.org/10.1182/blood-2009-04-214403
  93. Kioi M, Vogel H, Schultz G, Hoffman RM, Harsh GR, Brown JM. Inhibition of vasculogenesis, but not angiogenesis, prevents the recurrence of glioblastoma after irradiation in mice. J Clin Invest 2010;120:694-705. https://doi.org/10.1172/JCI40283
  94. Kozin SV, Kamoun WS, Huang Y, Dawson MR, Jain RK, Duda DG. Recruitment of myeloid but not endothelial precursor cells facilitates tumor regrowth after local irradiation. Cancer Res 2010;70:5679-85. https://doi.org/10.1158/0008-5472.CAN-09-4446
  95. Wong D, Korz W. Translating an antagonist of chemokine receptor CXCR4: from bench to bedside. Clin Cancer Res 2008;14:7975-80. https://doi.org/10.1158/1078-0432.CCR-07-4846
  96. Uchida D, Onoue T, Kuribayashi N, et al. Blockade of CXCR4 in oral squamous cell carcinoma inhibits lymph node metastases. Eur J Cancer 2011;47:452-9. https://doi.org/10.1016/j.ejca.2010.09.028

Cited by

  1. Innate Immune Activation by Tissue Injury and Cell Death in the Setting of Hematopoietic Stem Cell Transplantation vol.6, pp.None, 2014, https://doi.org/10.3389/fimmu.2015.00101
  2. Contemporary treatment with radiosurgery for spine metastasis and spinal cord compression in 2015 vol.33, pp.1, 2014, https://doi.org/10.3857/roj.2015.33.1.1
  3. Management of toxicities following pelvic irradiation for gynaecological cancers vol.27, pp.5, 2014, https://doi.org/10.1097/cco.0000000000000215
  4. Variation in human cancer cell external phosphatidylserine is regulated by flippase activity and intracellular calcium vol.6, pp.33, 2014, https://doi.org/10.18632/oncotarget.6045
  5. Selective cytoprotective effect of histamine on doxorubicin-induced hepatic and cardiac toxicity in animal models vol.1, pp.None, 2014, https://doi.org/10.1038/cddiscovery.2015.59
  6. Clinical outcomes of tissue expanders on adjuvant radiotherapy of resected retroperitoneal sarcoma vol.95, pp.28, 2014, https://doi.org/10.1097/md.0000000000004123
  7. Reverse geroscience: how does exposure to early diseases accelerate the age‐related decline in health? vol.1386, pp.1, 2016, https://doi.org/10.1111/nyas.13297
  8. Polymorphic Variants in Oxidative Stress Genes and Acute Toxicity in Breast Cancer Patients Receiving Radiotherapy vol.48, pp.3, 2014, https://doi.org/10.4143/crt.2015.360
  9. Soft Tissue Necrosis in Head and Neck Cancer Patients After Transoral Robotic Surgery or Wide Excision With Primary Closure Followed by Radiation Therapy vol.95, pp.9, 2016, https://doi.org/10.1097/md.0000000000002852
  10. Volumetric modulated arc therapy for carotid sparing in the management of early glottic cancer vol.34, pp.1, 2016, https://doi.org/10.3857/roj.2016.34.1.18
  11. MDP: A Deinococcus Mn 2+ -Decapeptide Complex Protects Mice from Ionizing Radiation vol.11, pp.8, 2014, https://doi.org/10.1371/journal.pone.0160575
  12. The Influence of C-Ions and X-rays on Human Umbilical Vein Endothelial Cells vol.6, pp.None, 2014, https://doi.org/10.3389/fonc.2016.00005
  13. Protection against Radiotherapy-Induced Toxicity vol.5, pp.3, 2014, https://doi.org/10.3390/antiox5030022
  14. Hybrid Manganese Dioxide Nanoparticles Potentiate Radiation Therapy by Modulating Tumor Hypoxia vol.76, pp.22, 2014, https://doi.org/10.1158/0008-5472.can-15-3475
  15. The evaluation of protective effect of lycopene against genotoxic influence of X-irradiation in human blood lymphocytes vol.56, pp.4, 2014, https://doi.org/10.1007/s00411-017-0713-6
  16. Management of Urethral Stricture and Bladder Neck Contracture Following Primary and Salvage Treatment of Prostate Cancer vol.18, pp.10, 2014, https://doi.org/10.1007/s11934-017-0729-0
  17. Radioprotective effect of vitamin E on salivary glands after radioiodine therapy for differentiated thyroid cancer: a randomized-controlled trial vol.38, pp.11, 2014, https://doi.org/10.1097/mnm.0000000000000727
  18. Ionizing Radiation Induces Innate Immune Responses in Macrophages by Generation of Mitochondrial Reactive Oxygen Species vol.187, pp.1, 2014, https://doi.org/10.1667/rr14346.1
  19. Microglial Activation in Traumatic Brain Injury vol.9, pp.None, 2017, https://doi.org/10.3389/fnagi.2017.00208
  20. Mechanisms of radiotherapy-associated cognitive disability in patients with brain tumours vol.13, pp.1, 2014, https://doi.org/10.1038/nrneurol.2016.185
  21. Genetic Variants in MTHFR Gene Predict ≥ 2 Radiation Pneumonitis in Esophageal Squamous Cell Carcinoma Patients Treated with Thoracic Radiotherapy vol.12, pp.1, 2014, https://doi.org/10.1371/journal.pone.0169147
  22. Assessing Oxidative Stress in Tumors by Measuring the Rate of Hyperpolarized [1- 13 C]Dehydroascorbic Acid Reduction Using 13 C Magnetic Resonance Spectroscopy vol.292, pp.5, 2014, https://doi.org/10.1074/jbc.m116.761536
  23. NLRP3 inflammasome activation mediates radiation-induced pyroptosis in bone marrow-derived macrophages vol.8, pp.2, 2017, https://doi.org/10.1038/cddis.2016.460
  24. Adverse effect of excess body weight on survival in cervical cancer patients after surgery and radiotherapy vol.35, pp.1, 2014, https://doi.org/10.3857/roj.2016.01977
  25. Panax ginseng Meyer prevents radiation-induced liver injury via modulation of oxidative stress and apoptosis vol.41, pp.2, 2014, https://doi.org/10.1016/j.jgr.2016.02.006
  26. The role of adjuvant external beam radiation therapy for papillary thyroid carcinoma invading the trachea vol.35, pp.2, 2017, https://doi.org/10.3857/roj.2017.00192
  27. 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 vol.93, pp.9, 2014, https://doi.org/10.1080/09553002.2017.1332438
  28. Synthesis and Radioprotective Activity of Mitochondria Targeted Dihydropyridines In Vitro vol.18, pp.11, 2014, https://doi.org/10.3390/ijms18112233
  29. Longitudinal brain structural alterations in patients with nasopharyngeal carcinoma early after radiotherapy vol.19, pp.None, 2014, https://doi.org/10.1016/j.nicl.2018.04.019
  30. Cerebral blood flow changes after radiation therapy identifies pseudoprogression in diffuse intrinsic pontine gliomas vol.20, pp.7, 2014, https://doi.org/10.1093/neuonc/nox227
  31. Radiation Induces Apoptosis and Osteogenic Impairment through miR-22-Mediated Intracellular Oxidative Stress in Bone Marrow Mesenchymal Stem Cells vol.2018, pp.None, 2018, https://doi.org/10.1155/2018/5845402
  32. Treatment-Related Adverse Effects in Lung Cancer Patients after Stereotactic Ablative Radiation Therapy vol.2018, pp.None, 2018, https://doi.org/10.1155/2018/6483626
  33. Impact of Unilateral Orbital Radiotherapy on the Structure and Function of Bilateral Human Meibomian Gland vol.2018, pp.None, 2014, https://doi.org/10.1155/2018/9308649
  34. Comparative study of radioprotective effects of selenium nanoparticles and sodium selenite in irradiation-induced nephropathy of mice model vol.94, pp.1, 2014, https://doi.org/10.1080/09553002.2018.1400709
  35. Prognostic Significance of Sarcopenia With Inflammation in Patients With Head and Neck Cancer Who Underwent Definitive Chemoradiotherapy vol.8, pp.None, 2014, https://doi.org/10.3389/fonc.2018.00457
  36. Heart in space: effect of the extraterrestrial environment on the cardiovascular system vol.15, pp.3, 2018, https://doi.org/10.1038/nrcardio.2017.157
  37. Perfusion magnetic resonance imaging changes in normal appearing brain tissue after radiotherapy in glioblastoma patients may confound longitudinal evaluation of treatment response vol.52, pp.2, 2014, https://doi.org/10.2478/raon-2018-0022
  38. Receptor tyrosine kinase signaling in cancer radiotherapy and its targeting for tumor radiosensitization vol.94, pp.7, 2014, https://doi.org/10.1080/09553002.2018.1478160
  39. Novel biological strategies to enhance the radiation therapeutic ratio vol.36, pp.3, 2014, https://doi.org/10.3857/roj.2018.00332
  40. Plasminogen activation is required for the development of radiation-induced dermatitis vol.9, pp.11, 2014, https://doi.org/10.1038/s41419-018-1106-8
  41. Inhibition of Radiation-Induced Ccl2 Signaling Protects Lungs from Vascular Dysfunction and Endothelial Cell Loss vol.30, pp.2, 2019, https://doi.org/10.1089/ars.2017.7458
  42. Radiation-induced Osteomyelitis/Osteonecrosis of the Rib: SPECT/CT Imaging for Successful Surgical Management vol.7, pp.12, 2014, https://doi.org/10.1097/gox.0000000000002536
  43. Free vastus lateralis musculocutaneous flap transfer for radiation-induced chest wall fistula combined with osteomyelitis : Two case report vol.98, pp.22, 2014, https://doi.org/10.1097/md.0000000000015859
  44. Combining Radiotherapy and Immunotherapy in Lung Cancer: Can We Expect Limitations Due to Altered Normal Tissue Toxicity? vol.20, pp.1, 2014, https://doi.org/10.3390/ijms20010024
  45. The healing effect of hydrogen-rich water on acute radiation-induced skin injury in rats vol.60, pp.1, 2014, https://doi.org/10.1093/jrr/rry074
  46. Targeting the Immunomodulatory CD73/Adenosine System to Improve the Therapeutic Gain of Radiotherapy vol.10, pp.None, 2014, https://doi.org/10.3389/fimmu.2019.00698
  47. Gut microbiota: implications for radiotherapy response and radiotherapy-induced mucositis vol.13, pp.5, 2014, https://doi.org/10.1080/17474124.2019.1595586
  48. Outcomes of reconstructive urinary tract surgery after pelvic radiotherapy vol.53, pp.2, 2014, https://doi.org/10.1080/21681805.2019.1611631
  49. Plasma Fibrinogen-Like 1 as a Potential Biomarker for Radiation-Induced Liver Injury vol.8, pp.9, 2014, https://doi.org/10.3390/cells8091042
  50. Melatonin Modulates Regulation of NOX2 and NOX4 Following Irradiation in the Lung vol.14, pp.3, 2014, https://doi.org/10.2174/1574884714666190502151733
  51. The CD73/Ado System-A New Player in RT Induced Adverse Late Effects vol.11, pp.10, 2014, https://doi.org/10.3390/cancers11101578
  52. The Use of Radioprotective Agents to Prevent Effects Associated with Aging vol.46, pp.12, 2014, https://doi.org/10.1134/s1062359019120021
  53. Radio-detoxified LPS alters bone marrow-derived extracellular vesicles and endothelial progenitor cells vol.10, pp.1, 2019, https://doi.org/10.1186/s13287-019-1417-4
  54. Therapeutic Reversal of Radiotherapy Injury to Pro-fibrotic Dysfunctional Fibroblasts In Vitro Using Adipose-derived Stem Cells vol.8, pp.3, 2014, https://doi.org/10.1097/gox.0000000000002706
  55. Enteric-Coated Strategies in Colorectal Cancer Nanoparticle Drug Delivery System vol.14, pp.None, 2020, https://doi.org/10.2147/dddt.s273612
  56. Assessment of monoamine neurotransmitters in the cortex and cerebellum of gamma-irradiated mice: A neuromodulatory role of Cynodon dactylon vol.19, pp.1, 2014, https://doi.org/10.4103/jcar.jcar_13_19
  57. Synergetic role of senna and fennel extracts as antioxidant, anti-inflammatory and anti-mutagenic agents in irradiated human blood lymphocyte cultures vol.13, pp.1, 2014, https://doi.org/10.1080/16878507.2020.1723948
  58. Assessment of antioxidant, immune enhancement, and antimutagenic efficacy of fennel seed extracts in irradiated human blood cultures vol.13, pp.1, 2014, https://doi.org/10.1080/16878507.2020.1728963
  59. Plasminogen is a master regulator and a potential drug candidate for the healing of radiation wounds vol.11, pp.3, 2020, https://doi.org/10.1038/s41419-020-2397-0
  60. Modulation of radiation‐induced damage of human glomerular endothelial cells by SMPDL3B vol.34, pp.6, 2020, https://doi.org/10.1096/fj.201902179r
  61. Radiotherapy and Its Impact on the Nervous System of Cancer Survivors vol.19, pp.5, 2014, https://doi.org/10.2174/1871527319666200708125741
  62. Comparing the Role of the p53 Gene and Telomerase Enzyme in ‘Accelerated Aging Due to Cancer’: A Literature Review vol.12, pp.10, 2020, https://doi.org/10.7759/cureus.10794
  63. Can a comparison of clinical and deep space irradiation scenarios shed light on the radiation response of the brain? vol.93, pp.1115, 2014, https://doi.org/10.1259/bjr.20200245
  64. Prostate Cancer Radiotherapy: Increased Biochemical Control and Late Toxicity in Men With Medication Allergies vol.4, pp.6, 2014, https://doi.org/10.1093/jncics/pkaa081
  65. Gold nanoparticle mediated radiation response among key cell components of the tumour microenvironment for the advancement of cancer nanotechnology vol.10, pp.None, 2014, https://doi.org/10.1038/s41598-020-68994-0
  66. Provisional Clinical Opinions for Radiation Therapy in Prefrail Elderly Colorectal Cancer Patients: Part One vol.74, pp.7, 2014, https://doi.org/10.3862/jcoloproctology.74.413
  67. Extracellular Vesicles for the Treatment of Radiation-Induced Normal Tissue Toxicity in the Lung vol.10, pp.None, 2014, https://doi.org/10.3389/fonc.2020.602763
  68. Toxicity Profile of Combining PD-1/PD-L1 Inhibitors and Thoracic Radiotherapy in Non-Small Cell Lung Cancer: A Systematic Review vol.12, pp.None, 2014, https://doi.org/10.3389/fimmu.2021.627197
  69. Radiation-Induced Vascular Disease-A State-of-the-Art Review vol.8, pp.None, 2014, https://doi.org/10.3389/fcvm.2021.652761
  70. Pharmacological Ascorbate Promotes the Tumor Radiosensitization of Au@Pd Nanoparticles with Simultaneous Protection of Normal Tissues vol.4, pp.2, 2014, https://doi.org/10.1021/acsabm.0c01537
  71. Three-Dimensional Tumor Spheroids as a Tool for Reliable Investigation of Combined Gold Nanoparticle and Docetaxel Treatment vol.13, pp.6, 2014, https://doi.org/10.3390/cancers13061465
  72. Psychotherapy with Music Intervention Improves Anxiety, Depression and the Redox Status in Breast Cancer Patients Undergoing Radiotherapy: A Randomized Controlled Clinical Trial vol.13, pp.8, 2014, https://doi.org/10.3390/cancers13081752
  73. Radiation-Induced Metabolic Shifts in the Hepatic Parenchyma: Findings from 18F-FDG PET Imaging and Tissue NMR Metabolomics in a Mouse Model for Hepatocellular Carcinoma vol.26, pp.9, 2014, https://doi.org/10.3390/molecules26092573
  74. NLRP3 protects mice from radiation-induced colon and skin damage via attenuating cGAS-STING signaling vol.418, pp.None, 2014, https://doi.org/10.1016/j.taap.2021.115495
  75. Radiation-Induced Bilateral Oculomotor Nerve Palsy 20 Years After Radiation Treatment vol.96, pp.20, 2021, https://doi.org/10.1212/wnl.0000000000011936
  76. Adverse Vascular Functional and Structural Changes Secondary to Breast Cancer and its Treatments with Adjuvant Therapy: a Systematic Review vol.3, pp.7, 2021, https://doi.org/10.1007/s42399-021-00859-7
  77. The Ability of the Nitric Oxide Synthases Inhibitor T1023 to Selectively Protect the Non-Malignant Tissues vol.22, pp.17, 2014, https://doi.org/10.3390/ijms22179340
  78. Devastated Bladder Outlet in Pelvic Cancer Survivors: Issues on Surgical Reconstruction and Quality of Life vol.10, pp.21, 2021, https://doi.org/10.3390/jcm10214920
  79. Clinical factors affecting the determination of radiotherapy-induced skin toxicity in breast cancer vol.39, pp.4, 2021, https://doi.org/10.3857/roj.2020.00395
  80. Medical countermeasures for radiation induced health effects: report of an Interagency Panel Session held at the NASA Human Research Program Investigator’s Workshop, 26 January 2017 vol.97, pp.suppl1, 2014, https://doi.org/10.1080/09553002.2019.1665214
  81. Phytochemicals: Potential Therapeutic Modulators of Radiation Induced Signaling Pathways vol.11, pp.1, 2014, https://doi.org/10.3390/antiox11010049