Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-v5vhk Total loading time: 0 Render date: 2024-06-22T21:59:04.634Z Has data issue: false hasContentIssue false

7 - Biological bases of radiation injury to the brain

Published online by Cambridge University Press:  13 August 2009

Christina A. Meyers
Affiliation:
University of Texas, M. D. Anderson Cancer Center
James R. Perry
Affiliation:
University of Toronto
Get access

Summary

Introduction

Neoplasms of the central nervous system (CNS) are a pathologically diverse group of benign and malignant tumors for which a variety of management strategies, including observation, surgery, radiation therapy, and/or chemotherapy, are employed. Regardless of the type of CNS tumor treated, what usually limits the dose of radiation that can be utilized, and therefore what typically determines the local control and cure rate of that tumor, are the tolerance doses of the adjacent or underlying normal tissues in and around the CNS. This chapter will outline the biological principles of CNS radiation tolerance and radiation-induced CNS injury, with an emphasis on the brain.

Pathogenesis of radiation-induced CNS injury

Classical model of parenchymal or vascular target cells

Vascular abnormalities and demyelination are the predominant histological changes seen in radiation-induced CNS injury. Classically, late delayed injury was viewed as due solely to a reduction in the number of surviving clonogens of either parenchymal, i.e., oligodendrocyte (Van den Maazen et al., 1993), or vascular, i.e., endothelial (Calvo et al., 1988), target cell populations leading to white matter necrosis.

Vascular hypothesis

Proponents of the vascular hypothesis argue that vascular damage leads to ischemia with secondary white matter necrosis. In support of this hypothesis is the large amount of data describing radiation-induced vascular changes including blood vessel (primarily arterial) wall thickening, vessel dilation, and endothelial cell nuclear enlargement (Calvo et al., 1988; Reinhold et al., 1990; Schultheiss & Stephens, 1992).

Type
Chapter
Information
Publisher: Cambridge University Press
Print publication year: 2008

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Abayomi, OK (1996). Pathogenesis of irradiation-induced cognitive dysfunction. Acta Oncol 35: 659–663.CrossRefGoogle ScholarPubMed
Anderson, VA, Godber, T, Smibert, Eet al. (2000). Cognitive and academic outcome following cranial irradiation and chemotherapy in children: a longitudinal study. Br J Cancer 82: 255–262.CrossRefGoogle ScholarPubMed
Andratschke, NH, Nieder, C, Price, REet al. (2005). Potential role of growth factors in diminishing radiation therapy neural tissue injury. Semin Oncol 32 [2 Suppl. 3]: S67–70.CrossRefGoogle ScholarPubMed
Armstrong, CL, Gyato, K, Awadalla, AWet al. (2004). A critical review of the clinical effects of therapeutic irradiation damage to the brain: the roots of the controversy. Neuropsychol Rev 14: 65–86.CrossRefGoogle Scholar
Banisadr, G, Quéraud-Lesaux, F, Boutterin, MCet al. (2002). Distribution, cellular localization and functional role of CCR2 chemokine receptors in adult rat brain. J Neurochem 81: 257–269.CrossRefGoogle ScholarPubMed
Bassant, MH, Court, L (1978). Effect of whole-body gamma irradiation on the activity of rabbit hippocampal neurons. Radiat Res 75: 595–606.CrossRefGoogle Scholar
Bellinzona, M, Gobbel, GT, Shinohara, Cet al. (1996). Apoptosis is induced in the subependyma of young adult rats by ionizing irradiation. Neurosci Lett 208: 163–166.CrossRefGoogle ScholarPubMed
Bordet, R, Gelé, P, Duriez, Pet al. (2006). PPARs: a new target for neuroprotection. J Neurol Neurosurg Psychiatry 77: 285–287.CrossRefGoogle ScholarPubMed
Brown, WR, Thore, CR, Moody, DMet al. (2005). Vascular damage after fractionated whole-brain irradiation in rats. Radiat Res 164: 662–668.CrossRefGoogle ScholarPubMed
Calvo, W, Hopewell, JW, Reinhold, HSet al. (1988). Time- and dose-related changes in the white matter of the rat brain after single doses of X rays. Br J Radiol 61: 1043– 1052.CrossRefGoogle ScholarPubMed
Chiang, C-S, McBride, WH, Withers, HR (1993). Radiation-induced astrocytic and microglial cellular hyperplasia. Radiother Oncol 29: 60–68.CrossRefGoogle Scholar
Cooper, JS, Fu, K, Marks, Jet al. (1995). Late effects of radiation in the head and neck region. Int J Radiat Oncol Biol Phys 31: 1141–1164.CrossRefGoogle ScholarPubMed
Cox, JD, Stetz, J, Pajak, TF (1995). Toxicity criteria of the Radiation Therapy Oncology Group and the European Organization for Research and Treatment of Cancer. Int J Radiat Oncol Biol Phys 31: 1341–1346.CrossRefGoogle ScholarPubMed
Crossen, JR, Garwood, D, Glatstein, Eet al. (1994). Neurobehavioral sequelae of cranial irradiation in adults: a review of radiation-induced encephalopathy. J Clin Oncol 12: 627–642.CrossRefGoogle ScholarPubMed
El-Agamawi, AY, Hopewell, JW, Plowman, PNet al. (1996). Modulation of normal tissue responses to radiation. Br J Radiol 69: 374–375.CrossRefGoogle Scholar
Ellis, F (1969). Dose, time fractionation: a clinical hypothesis. Clin Radiol 20: 1–7.CrossRefGoogle ScholarPubMed
Emami, B, Lyman, J, Brown, Aet al. (1991). Tolerance of normal tissue to therapeutic irradiation. Int J Radiat Oncol Biol Phys 21: 109–122.CrossRefGoogle ScholarPubMed
Esik, O, Csere, T, Stefantis, Ket al. (2003). A review on radiogenic Lhermitte's sign. Pathol Oncol Res 9: 115–120.CrossRefGoogle ScholarPubMed
Feinstein, DL (2003). Therapeutic potential of peroxisomal proliferator-activated receptor agonists for neurological disease. Diabetes Technol Ther 5: 67–73.CrossRefGoogle Scholar
Fike, JR, Goebbel, GT, Martob, Jet al. (1994). Radiation brain injury is reduced by the polyamine inhibitor alpha-difluoromethylornithine. Radiat Res 138: 99–106.CrossRefGoogle ScholarPubMed
Fowler, JF (1992). Brief summary of radiobiological principles in fractionated radiotherapy. Semin Radiat Oncol 2: 16–21.CrossRefGoogle Scholar
Fukuda, H, Fukuda, A, Zhu, Cet al. (2004). Irradiation-induced progenitor cell death in the developing brain is resistant to erythropoietin treatment and caspase inhibition. Cell Death Differ 11: 1166–1178.CrossRefGoogle ScholarPubMed
Gage, FH, Kempermann, G, Palmer, TDet al. (1998). Multipotent progenitor cells in the adult dentate gyrus. J Neurobiol 36: 249–266.3.0.CO;2-9>CrossRefGoogle ScholarPubMed
Gordon, KB, Char, DH, Sagerman, RH (1995). Late effects of radiation on the eye and ocular adnexa. Int J Radiat Oncol Biol Phys 31: 1123–1140.CrossRefGoogle ScholarPubMed
Grommes, C, Landreth, GE, Heneka, MT (2004). Antineoplastic effects of peroxisome proliferator-activated receptor γ agonists. Lancet Oncol 5: 419–429.CrossRefGoogle ScholarPubMed
Hopewell, JW, Aardweg, GJMJ, Morris, GMet al. (1993). Unsaturated lipids as modulators of radiation damage in normal tissues. In Horrobin, DF (ed.). New Approaches to Cancer Treatment (pp. 88–106). London: Churchill Communications Europe.Google Scholar
Horner, PJ, Palmer, TD(2003). New roles for astrocytes: the nightlife of an “astrocyte”. La vida loca!Trends Neurosci 26: 597–603.CrossRefGoogle ScholarPubMed
Hornsey, S, Myers, R, Coultas, PGet al. (1981). Turnover of proliferative cells in the spinal cord after X irradiation and its relation to time-dependent repair of radiation damage. Br J Radiol 54: 1081–1085.CrossRefGoogle ScholarPubMed
Hornsey, S, Myers, R, Jenkinson, T (1990). The reduction of radiation damage to the spinal cord by postirradiation administration of vasoactive drugs. Int J Radiat Oncol Biol Phys 18: 1437–1442.CrossRefGoogle Scholar
Jaenke, RS, Robbins, MEC, Bywaters, Tet al. (1993). Capillary endothelium: target site of renal radiation injury. Lab Invest 57: 551–565.Google Scholar
Kim, JH, Brown, SL, Kolozsvary, Aet al. (2004). Modification of radiation injury by Ramipril, inhibitor of the angiotensin-converting enzyme, on optic neuropathy in the rat. Radiat Res 161: 137–142.CrossRefGoogle ScholarPubMed
Leibel, SA, Sheline, GE (1991). Tolerance of the brain and spinal cord to conventional irradiation. In Gutin, P, Liebel, SA, Sheline, GE (eds.) Radiation Injury to the Nervous System (1st edn.) (pp. 211–238). New York: Raven Press.Google Scholar
Limoli, CL, Rola, R, Giedzinski, Eet al. (2004). Cell-density-dependent regulation of neural precursor cells function. Proc Natl Acad Sci USA 101: 16052–16057.CrossRefGoogle Scholar
Limoli, CL, Giedzinski, E, Baure, Jet al. (2006). Altered growth and radiosensitivity in neural precursor cells subjected to oxidative stress. Int J Radiat Biol 82: 640–647.CrossRefGoogle ScholarPubMed
Lynch, CD, Sonntag, WE, Wheeler, KT (2002). Radiation-induced dementia in aged rats: effects of growth hormone and insulin-like growth factor 1 [Abstract]. Neuro-Oncology 4: 354.Google Scholar
Macbeth, FR, Wheldon, TE, Girling, DJet al. (1996). Radiation myelopathy: estimates of risk in 1048 patients in three randomized trials of palliative radiotherapy for non-small cell lung cancer. The Medical Research Council Lung Cancer Working Party. Clin Oncol (R Coll Radiol) 8: 176–181.CrossRefGoogle ScholarPubMed
McCunniff, AJ, Liang, AJ (1989). Radiation tolerance of the cervical spinal cord. Int J Radiat Oncol Biol Phys 16: 675–678.CrossRefGoogle ScholarPubMed
Mildenberger, M, Beach, TG, McGeer, EGet al. (1990). An animal model of prophylactic cranial irradiation: histologic effects at acute, early and delayed stages. Int J Radiat Oncol Biol Phys 18: 1051–1060.CrossRefGoogle ScholarPubMed
Mizumatsu, S, Monje, ML, Morhardt, et al. (2003). Extreme sensitivity of adult neurogenesis to low doses of X-irradiation. Cancer Res 63: 4021–4027.Google ScholarPubMed
Monje, ML, Mizumatsu, S, Fike, JRet al. (2002). Irradiation induced neural precursor-cell dysfunction. Nature Med 8: 955–961.CrossRefGoogle ScholarPubMed
Monje, ML, Toda, H, Palmer, TD (2003). Inflammatory blockade restores adult hippocampal neurogenesis. Science 302: 1760–1764.CrossRefGoogle ScholarPubMed
Moore, BD, Copeland, DR, Ried, Het al. (1992). Neurophysiological basis of cognitive deficits in long-term survivors of childhood cancer. Arch Neurol 49: 809–817.CrossRefGoogle ScholarPubMed
Morris, GM, Coderre, JA, Bywaters, Aet al. (1996). Boron neutron capture irradiation of the rat spinal cord: histopathological evidence of a vascular-mediated pathogenesis. Radiat Res 146: 313–320.CrossRefGoogle ScholarPubMed
Moulder, J, Robbins, MEC, Cohen, EPet al. (1998). Pharmacologic modification of radiation-induced late normal tissue injury. In Mittal, BB, Purdy, JA, Ang, KK (eds.) Radiation Therapy (pp. 129–151). Norwell, MA: Kluwer.CrossRefGoogle Scholar
Mulhern, RK, Merchant, TE, Gajjar, Aet al. (2004). Late neurocognitive sequelae in survivors of brain tumours in childhood. Lancet Oncol 5: 399–408.CrossRefGoogle ScholarPubMed
Muller, HW, Junghans, U, Kappler, J (1995). Astroglial neurotrophic and neurite-promoting factors. Pharmacol Ther 65: 1–18.CrossRefGoogle ScholarPubMed
Nakagawa, M, Bellinzona, M, Seilhan, TMet al. (1996). Microglial responses after focal radiation-induced injury are affected by alpha-difluoromethylornithine. Int J Radiat Oncol Biol Phys 36: 113–123.CrossRefGoogle ScholarPubMed
Nakaya, K, Hasegawa, T, Flickinger, JCet al. (2005). Sensitivity to radiation-induced apoptosis and neuron loss declines rapidly in the postnatal mouse neocortex. Int J Radiat Biol 81: 545–554.CrossRefGoogle ScholarPubMed
Nieder, C, Price, RE, Rivera, Bet al. (2000). Both early and delayed treatment with growth factors can modulate the development of radiation myelopathy (RM) in rats [Abstract]. Radiother Oncol 56 [Suppl. 1]: S15.Google Scholar
Noel, F, Gumin, GJ, Raju, Uet al. (1998). Increased expression of prohormone convertase-2 in the irradiated rat brain. FASEB J 12: 1725–1730.CrossRefGoogle ScholarPubMed
Palmer, TD, Takahashi, J, Gage, FH (1997). The adult rat hippocampus contains primordial neural stem cells. Mol Cell Neurosci 8: 389–404.CrossRefGoogle ScholarPubMed
Palmer, TD, Willhoite, AR, Gage, FH (2000). Vascular niche for adult hippocampal neurogenesis. J Comp Neurol 425: 479–494.3.0.CO;2-3>CrossRefGoogle ScholarPubMed
Pekny, M, Nilsson, M (2005). Astrocyte activation and reactive gliosis. Glia 50: 427–434.CrossRefGoogle ScholarPubMed
Pellmar, TC, Lepinski, DL (1993). Gamma radiation (5–10 Gy) impairs neuronal function in the guinea pig hippocampus. Radiat Res 136: 255–261.CrossRefGoogle ScholarPubMed
Raff, MC, Miller, RH, Noble, M (1983). A glial progenitor cell that develops in vitro into an astrocyte or an oligodendrocyte depending on culture medium. Nature 303: 390–396.CrossRefGoogle ScholarPubMed
Reinhold, HS, Calvo, W, Hopewell, JWet al. (1990). Development of blood vessel-related radiation damage in the fimbria of the central nervous system. Int J Radiat Oncol Biol Phys 18: 37–42.CrossRefGoogle ScholarPubMed
Rezvani, M, Birds, DA, Hodges, Het al. (2002). Modification of radiation myelopathy by the transplantation of neural stem cells in the rat. Radiat Res 156: 408–412.CrossRefGoogle Scholar
Robbins, MEC, Zhao, W (2004). Chronic oxidative stress and radiation-induced late normal tissue injury: a review. Int J Radiat Biol 80: 251–259.CrossRefGoogle ScholarPubMed
Rola, R, Raber, J, Rizk, Aet al. (2004). Radiation-induced impairment of hippocampal neurogenesis is associated with cognitive deficits in young mice. Exp Neurol 188: 316–330.CrossRefGoogle ScholarPubMed
Rola, R, Sarkissian, V, Obenaus, Aet al. (2005). High-LET radiation induces inflammation and persistent changes in markers of hippocampal neurogenesis. Radiat Res 164: 556–560.CrossRefGoogle ScholarPubMed
Roman, DD, Sperduto, PW (1995). Neuropsychological effects of cranial radiation: current knowledge and future directions. Int J Radiat Oncol Biol Phys 31: 983–998.CrossRefGoogle ScholarPubMed
Rubin, P, Casarett, GW (1968). Clinical Radiation Pathology (Vols 1 and 2). Philadelphia, PA: WB Saunders.Google ScholarPubMed
Sarkissian, V (2005). The sequelae of cranial irradiation on human cognition. Neurosci Lett 382: 118–123.CrossRefGoogle ScholarPubMed
Schultheiss, TE, Stephens, LC (1992). Permanent radiation myelopathy. Br J Radiol 65: 737–753.CrossRefGoogle ScholarPubMed
Schultheiss, TE, Kun, , Ang, KKet al. (1995). Radiation response of the central nervous system. Int J Radiat Oncol Biol Phys 31: 1093–1112.CrossRefGoogle ScholarPubMed
Shaw, E, Arusell, R, Scheithauer, Bet al. (2002). A prospective randomized trial of low-versus high-dose radiation therapy in adults with supratentorial low-grade glioma: initial report of a NCCTG-RTOG-ECOG Study. J Clin Oncol 20: 2267–2276.CrossRefGoogle ScholarPubMed
Sheline, GE, Wara, WM, Smith, V (1980). Therapeutic irradiation and brain injury. Int J Radiat Oncol Biol Phys 6: 1215–1218.CrossRefGoogle ScholarPubMed
Siegal, T, Pfeffer, MR, Meltzer, Aet al. (1996). Cellular and secretory mechanisms related to delayed radiation-induced microvessel dysfunction in the spinal cord of rats. Int J Radiat Oncol Biol Phys 36: 649–659.CrossRefGoogle ScholarPubMed
Sklar, CA, Constine, LS (1995). Chronic neuroendocrinological sequelae of radiation therapy. Int J Radiat Oncol Biol Phys 31: 1113–1122.CrossRefGoogle ScholarPubMed
Sminia, P, Kleij, AJ, Carl, UMet al. (2003). Prophylactic hyperbaric oxygen treatment and rat spinal cord irradiation. Cancer Lett 191: 59–65.CrossRefGoogle Scholar
Snyder, JS, Kee, N, Wojtowicz, JM (2003). Effects of adult neurogenesis on synaptic plasticity in the rat dentate gyrus. J Neurophysiol 85: 2423–2431.CrossRefGoogle Scholar
Song, H, Stevens, CF, Gage, FH (2002). Astroglia induce neurogenesis from adult neural stem cells. Nature 417: 39–44.CrossRefGoogle ScholarPubMed
Spence, AM, Krohn, KA, Edmonson, SWet al. (1986). Radioprotection in rat spinal cord with WR-2721 following cerebral lateral intraventricular injection. Int J Radiat Oncol Biol Phys 12: 1479–1482.CrossRefGoogle ScholarPubMed
Stoll, G, Jander, S (1999). The role of microglia and macrophages in the pathophysiology of the CNS. Prog Neurobiol 58: 233–247.CrossRefGoogle ScholarPubMed
Surma-aho, O, Niemalä, M, Vilkki, Jet al. (2001). Adverse long-term effects of brain radiotherapy in adult low-grade glioma patients. Neurology 56: 1285–1290.CrossRefGoogle ScholarPubMed
Tofilon, PJ, Fike, JR (2000). The radioresponse of the central nervous system: a dynamic process. Radiat Res 153: 357–370.CrossRefGoogle ScholarPubMed
Maazen, RWM, Kleiboer, BJ, Verhagen, Iet al. (1991a). Irradiation in vitro discriminates between different O-2A progenitor cell subpopulations in the perinatal central nervous system of rats. Radiat Res 128: 64–72.CrossRefGoogle ScholarPubMed
Maazen, RWM, Verhagen, I, Kleiboer, BJet al. (1991b). Radiosensitivity of glial progenitor cells of the prenatal and adult rat optic nerve studies by an in vitro clonogenic assay. Radiother Oncol 20: 258–264.CrossRefGoogle Scholar
Maazen, RWM, Kleiboer, BJ, Berhagen, Iet al. (1993). Repair capacity of adult rat glial progenitor cells determined by an in vitro clonogenic assay after in vitro or in vivo fractionated irradiation. Int J Radiat Biol 63: 661–666.CrossRefGoogle ScholarPubMed
Rossum, D, Hanisch, UK (2004). Microglia. Metab Brain Dis 19: 393–411.CrossRefGoogle ScholarPubMed
Vaughan, DW, Peters, A (1974). Neuroglial cells in the cerebral cortex of rats from young adulthood to old age: an electron microscopic study. J Neurocytol 3: 405–429.CrossRefGoogle Scholar
Volterra, A, Meldelosi, J (2005). Astrocytes from brain glue to communication elements: the revolution continues. Nat Rev Neurosci 6: 626–640.CrossRefGoogle ScholarPubMed
Weitzner, MA, Meyers, CA, Gelke, CKet al. (1995). The Functional Assessment of Cancer Therapy (FACT) scale: development of a brain subscale and revalidation of the general version (FACT-G) in patients with primary brain tumors. Cancer 75: 1151–1161.3.0.CO;2-Q>CrossRefGoogle ScholarPubMed
Zhao, W, Payne, V, Tommasi, Eet al. (2007). Administration of the peroxisomal proliferator-activated receptor β agonist pioglitazone during fractionated brain irradiation prevents radiation-induced cognitive impairment. Int J Radiat Oncol Biol Phys 67: 6–9.CrossRefGoogle Scholar

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×