Skip to main content
Top
Published in: Molecular and Cellular Pediatrics 1/2018

Open Access 01-12-2018 | Mini review

Chemotherapy and the pediatric brain

Author: Chrysanthy Ikonomidou

Published in: Molecular and Cellular Pediatrics | Issue 1/2018

Login to get access

Abstract

Survival rates of children with cancer are steadily increasing. This urges our attention to neurocognitive and psychiatric outcomes, as these can markedly influence the quality of life of these children. Neurobehavioral morbidity in childhood cancer survivors affects diverse aspects of cognitive function, which can include attention, memory, processing speed, intellect, academic achievement, and emotional health. Reasons for neurobehavioral morbidity are multiple with one major contributor being chemotherapy-induced central nervous system (CNS) toxicity. Clinical studies investigating the effects of chemotherapy on the CNS in children with cancer have reported causative associations with the development of leukoencephalopathies as well as smaller regional grey and white matter volumes, which have been found to correlate with neurocognitive deficits.
Preclinical work has provided compelling evidence that chemotherapy drugs are potent neuro- and gliotoxins in vitro and in vivo and can cause brain injury via excitotoxic and apoptotic mechanisms. Furthermore, chemotherapy triggers DNA (deoxyribonucleic acid) damage directly or through increased oxidative stress. It can shorten telomeres and accelerate cell aging, cause cytokine deregulation, inhibit hippocampal neurogenesis, and reduce brain vascularization and blood flow. These mechanisms, when allowed to operate on the developing brain of a child, have high potential to not only cause brain injury, but also alter crucial developmental events, such as myelination, synaptogenesis, neurogenesis, cortical thinning, and formation of neuronal networks.
This short review summarizes key publications describing neurotoxicity of chemotherapy in pediatric cancers and potential underlying pathomechanisms.
Literature
1.
go back to reference Henderson TO, Friedman DL, Meadows AT (2010) Childhood cancer survivors: transition to adult-focused risk-based care. Pediatrics 126:129–136PubMedCrossRef Henderson TO, Friedman DL, Meadows AT (2010) Childhood cancer survivors: transition to adult-focused risk-based care. Pediatrics 126:129–136PubMedCrossRef
2.
go back to reference Kadan-Lottick NS, Zeltzer LK, Liu Q et al (2010) Neurocognitive functioning in adult survivors of childhood noncentral nervous system cancers. J Natl Cancer Inst 102:881–893PubMedPubMedCentralCrossRef Kadan-Lottick NS, Zeltzer LK, Liu Q et al (2010) Neurocognitive functioning in adult survivors of childhood noncentral nervous system cancers. J Natl Cancer Inst 102:881–893PubMedPubMedCentralCrossRef
3.
go back to reference Oeffinger KC, Nathan PC, Kremer LC (2010) Challenges after curative treatment for childhood cancer and long-term follow up of survivors. Hematol Oncol Clin North Am 24:129–149PubMedCrossRef Oeffinger KC, Nathan PC, Kremer LC (2010) Challenges after curative treatment for childhood cancer and long-term follow up of survivors. Hematol Oncol Clin North Am 24:129–149PubMedCrossRef
4.
go back to reference Zeltzer LK, Recklitis C, Buchbinder D et al (2009) Psychological status in childhood cancer survivors: a report from the Childhood Cancer Survivor Study. J Clin Oncol 27:2396–2404PubMedPubMedCentralCrossRef Zeltzer LK, Recklitis C, Buchbinder D et al (2009) Psychological status in childhood cancer survivors: a report from the Childhood Cancer Survivor Study. J Clin Oncol 27:2396–2404PubMedPubMedCentralCrossRef
5.
go back to reference Sleurs C, Lemiere J, Vercruysse T et al (2017) Intellectual development of childhood ALL patients: a multicenter longitudinal study. Psycho-Oncology 26:508–514PubMedCrossRef Sleurs C, Lemiere J, Vercruysse T et al (2017) Intellectual development of childhood ALL patients: a multicenter longitudinal study. Psycho-Oncology 26:508–514PubMedCrossRef
6.
go back to reference Duffner PK, Armstrong FD, Chen L et al (2014) Neurocognitive and neuroradiologic central nervous system late effects in children treated on Pediatric Oncology Group (POG) P9605 (standard risk) and P9201 (lesser risk) acute lymphoblastic leukemia protocols (ACCL0131): a methotrexate consequence? A report from the Children’s Oncology Group. J Pediatr Hematol Oncol 36:8–15PubMedPubMedCentralCrossRef Duffner PK, Armstrong FD, Chen L et al (2014) Neurocognitive and neuroradiologic central nervous system late effects in children treated on Pediatric Oncology Group (POG) P9605 (standard risk) and P9201 (lesser risk) acute lymphoblastic leukemia protocols (ACCL0131): a methotrexate consequence? A report from the Children’s Oncology Group. J Pediatr Hematol Oncol 36:8–15PubMedPubMedCentralCrossRef
7.
go back to reference Anderson FS, Kunin-Batson AS (2009) Neurocognitive late effects of chemotherapy in children: the past 10 years of research on brain structure and function. Pediatr Blood Cancer 52:159–164PubMedCrossRef Anderson FS, Kunin-Batson AS (2009) Neurocognitive late effects of chemotherapy in children: the past 10 years of research on brain structure and function. Pediatr Blood Cancer 52:159–164PubMedCrossRef
8.
go back to reference Ashford J, Schoffstall C, Reddick WE et al (2010) Attention and working memory abilities in children treated for acute lymphoblastic leukemia. Cancer 116:1173–1186CrossRef Ashford J, Schoffstall C, Reddick WE et al (2010) Attention and working memory abilities in children treated for acute lymphoblastic leukemia. Cancer 116:1173–1186CrossRef
9.
go back to reference Buizer AI, de Sonneville LM, Veerman AJ (2009) Effects of chemotherapy on neurocognitive function in children with acute lymphoblastic leukemia: a critical review of the literature. Pediatr Blood Cancer 52:447–454PubMedCrossRef Buizer AI, de Sonneville LM, Veerman AJ (2009) Effects of chemotherapy on neurocognitive function in children with acute lymphoblastic leukemia: a critical review of the literature. Pediatr Blood Cancer 52:447–454PubMedCrossRef
10.
go back to reference Lofstad GE, Reinfjell T, Hestad K et al (2009) Cognitive outcome in children and adolescents treated for acute lymphoblastic leukaemia with chemotherapy only. Acta Paediatr 98:180–186PubMedPubMedCentralCrossRef Lofstad GE, Reinfjell T, Hestad K et al (2009) Cognitive outcome in children and adolescents treated for acute lymphoblastic leukaemia with chemotherapy only. Acta Paediatr 98:180–186PubMedPubMedCentralCrossRef
11.
go back to reference Moleski M (2000) Neuropsychological, neuroanatomical, and neurophysiological consequences of CNS chemotherapy for acute lymphoblastic leukemia. Arch Clin Neuropsychol 15:603–630PubMedCrossRef Moleski M (2000) Neuropsychological, neuroanatomical, and neurophysiological consequences of CNS chemotherapy for acute lymphoblastic leukemia. Arch Clin Neuropsychol 15:603–630PubMedCrossRef
12.
go back to reference von der Weid N, Mosimann I, Hirt A et al (2003) Intellectual outcome in children and adolescents with acute lymphoblastic leukaemia treated with chemotherapy alone: age- and sex-related differences. Eur J Cancer 39:359–365PubMedCrossRef von der Weid N, Mosimann I, Hirt A et al (2003) Intellectual outcome in children and adolescents with acute lymphoblastic leukaemia treated with chemotherapy alone: age- and sex-related differences. Eur J Cancer 39:359–365PubMedCrossRef
13.
go back to reference Jansen NC, Kingma A, Schuitema A et al (2008) Neuropsychological outcome in chemotherapy-only-treated children with acute lymphoblastic leukemia. J Clin Oncol 26:3025–3030PubMedCrossRef Jansen NC, Kingma A, Schuitema A et al (2008) Neuropsychological outcome in chemotherapy-only-treated children with acute lymphoblastic leukemia. J Clin Oncol 26:3025–3030PubMedCrossRef
14.
go back to reference Bhojwani D, Sabin ND, Pei D et al (2014) Methotrexate-induced neurotoxicity and leukoencephalopathy in childhood acute lymphoblastic leukemia. J Clin Oncol 32:949–959PubMedPubMedCentralCrossRef Bhojwani D, Sabin ND, Pei D et al (2014) Methotrexate-induced neurotoxicity and leukoencephalopathy in childhood acute lymphoblastic leukemia. J Clin Oncol 32:949–959PubMedPubMedCentralCrossRef
15.
go back to reference Carey ME, Haut MW, Reminger SL et al (2008) Reduced frontal white matter volume in long-term childhood leukemia survivors: a voxel-based morphometry study. AJNR Am J Neuroradiol 29:792–797PubMedCrossRefPubMedCentral Carey ME, Haut MW, Reminger SL et al (2008) Reduced frontal white matter volume in long-term childhood leukemia survivors: a voxel-based morphometry study. AJNR Am J Neuroradiol 29:792–797PubMedCrossRefPubMedCentral
17.
go back to reference Asato R, Akiyama Y, Ito M et al (1992) Nuclear magnetic resonance abnormalities of the cerebral white matter in children with acute lymphoblastic leukemia and malignant lymphoma during and after central nervous system prophylactic treatment with intrathecal methotrexate. Cancer 70:1997–2004PubMedCrossRef Asato R, Akiyama Y, Ito M et al (1992) Nuclear magnetic resonance abnormalities of the cerebral white matter in children with acute lymphoblastic leukemia and malignant lymphoma during and after central nervous system prophylactic treatment with intrathecal methotrexate. Cancer 70:1997–2004PubMedCrossRef
18.
go back to reference Dellani PR, Eder S, Gawehn J et al (2008) Late structural alterations of cerebral white matter in long-term survivors of childhood leukemia. J Magn Reson Imaging 27:1250–1255PubMedCrossRef Dellani PR, Eder S, Gawehn J et al (2008) Late structural alterations of cerebral white matter in long-term survivors of childhood leukemia. J Magn Reson Imaging 27:1250–1255PubMedCrossRef
19.
go back to reference Reddick WE, Glass JO, Johnson DP, Laningham FH, Pui C-H (2009) Voxel-based analysis of T2 hyperintensities in white matter during treatment of childhood leukemia. Am J Neuroradiol 30:1947–1954PubMedCrossRefPubMedCentral Reddick WE, Glass JO, Johnson DP, Laningham FH, Pui C-H (2009) Voxel-based analysis of T2 hyperintensities in white matter during treatment of childhood leukemia. Am J Neuroradiol 30:1947–1954PubMedCrossRefPubMedCentral
20.
go back to reference Deprez S, Amant F, Smeets A et al (2012) Longitudinal assessment of chemotherapy-induced changes in cerebral white matter and its correlation with impaired cognitive functioning. J Clin Oncol 30:274–281PubMedCrossRef Deprez S, Amant F, Smeets A et al (2012) Longitudinal assessment of chemotherapy-induced changes in cerebral white matter and its correlation with impaired cognitive functioning. J Clin Oncol 30:274–281PubMedCrossRef
21.
go back to reference Khong P-L, Leung LHT, Fung ASM et al (2006) White matter anisotropy in post-treatment childhood cancer survivors: preliminary evidence of association with neurocognitive function. J Clin Oncol 24:884–890PubMedCrossRef Khong P-L, Leung LHT, Fung ASM et al (2006) White matter anisotropy in post-treatment childhood cancer survivors: preliminary evidence of association with neurocognitive function. J Clin Oncol 24:884–890PubMedCrossRef
22.
go back to reference Edelmann MN, Krull KR, Liu W et al (2014) Diffusion tenson imaging and neurocognition in survivors of childhood acute lymphoblastic leukaemia. Brain 137:2973–2983PubMedPubMedCentralCrossRef Edelmann MN, Krull KR, Liu W et al (2014) Diffusion tenson imaging and neurocognition in survivors of childhood acute lymphoblastic leukaemia. Brain 137:2973–2983PubMedPubMedCentralCrossRef
23.
go back to reference ElAlfy M, Ragab I, Azab I, Amin S, Abdel-Maguid M (2014) Neurocognitive outcome and white matter anisotropy in childhood acute lymphoblastic leukemia survivors treated with different protocols. Pediatr Hematol Oncol 31:194–204PubMedCrossRef ElAlfy M, Ragab I, Azab I, Amin S, Abdel-Maguid M (2014) Neurocognitive outcome and white matter anisotropy in childhood acute lymphoblastic leukemia survivors treated with different protocols. Pediatr Hematol Oncol 31:194–204PubMedCrossRef
24.
go back to reference Kesler SR, Gugel M, Huston-Warren E, Watson C (2016) Atypical structural connectome organization and cognitive impairment in young survivors of acute lymphoblastic leukemia. Brain Connect 6:273–282PubMedPubMedCentralCrossRef Kesler SR, Gugel M, Huston-Warren E, Watson C (2016) Atypical structural connectome organization and cognitive impairment in young survivors of acute lymphoblastic leukemia. Brain Connect 6:273–282PubMedPubMedCentralCrossRef
25.
go back to reference Sleurs C, Lemiere J, Christiaens D et al (2018) Advanced MR diffusion imaging and chemotherapy-related changes in cerebral white matter microstructure of survivors of childhood bone and soft tissue sarcoma. Hum Brain Mapp:1–13 Sleurs C, Lemiere J, Christiaens D et al (2018) Advanced MR diffusion imaging and chemotherapy-related changes in cerebral white matter microstructure of survivors of childhood bone and soft tissue sarcoma. Hum Brain Mapp:1–13
26.
go back to reference McDonald BC, Conroy SK, Ahles TA, West JD, Saykin AJ (2010) Gray matter reduction associated with systemic chemotherapy for breast cancer: a prospective MRI study. Breast Canc Res Treat 123:819–828CrossRef McDonald BC, Conroy SK, Ahles TA, West JD, Saykin AJ (2010) Gray matter reduction associated with systemic chemotherapy for breast cancer: a prospective MRI study. Breast Canc Res Treat 123:819–828CrossRef
27.
go back to reference McDonald BC, Conroy SK, Smith DJ, West JD, Saykin AJ (2013) Frontal gray matter reduction after breast cancer chemotherapy and association with executive symptoms: a replication and extension study. Brain Behav Immun 30:S117–S125PubMedCrossRef McDonald BC, Conroy SK, Smith DJ, West JD, Saykin AJ (2013) Frontal gray matter reduction after breast cancer chemotherapy and association with executive symptoms: a replication and extension study. Brain Behav Immun 30:S117–S125PubMedCrossRef
28.
go back to reference Tamnes CT, Zeller B, Amlien IK et al (2015) Cortical surface area and thickness in adult survivors of pediatric acute lymphoblastic leukemia. Pediatr Blood Cancer 62:1027–1034PubMedCrossRef Tamnes CT, Zeller B, Amlien IK et al (2015) Cortical surface area and thickness in adult survivors of pediatric acute lymphoblastic leukemia. Pediatr Blood Cancer 62:1027–1034PubMedCrossRef
29.
go back to reference Oesterlundh G, Kjellmer I, Lannering B et al (2008) Neurochemical markers of brain damage in cerebrospinal fluid during induction treatment of acute lymphoblastic leukemia in children. Pediatr Blood Cancer 50:793–798CrossRef Oesterlundh G, Kjellmer I, Lannering B et al (2008) Neurochemical markers of brain damage in cerebrospinal fluid during induction treatment of acute lymphoblastic leukemia in children. Pediatr Blood Cancer 50:793–798CrossRef
30.
go back to reference Quinn CT, Griener JC, Bottiglieri T et al (1997) Elevation of homocysteine and excitatory amino acid neurotransmitters in the CSF of children who receive methotrexate for the treatment of cancer. J Clin Oncol 15:2800–2806PubMedCrossRef Quinn CT, Griener JC, Bottiglieri T et al (1997) Elevation of homocysteine and excitatory amino acid neurotransmitters in the CSF of children who receive methotrexate for the treatment of cancer. J Clin Oncol 15:2800–2806PubMedCrossRef
31.
go back to reference Viacha V, Eliopoulou M, Haidas S, Beratis NG (2004) Correlation of cerebrospinal fluid betal-glucuronidase activity with plasma methotrexate concentrations in leukemic children receiving high-dose methotrexate. Pediatr Blood Cancer 42:350–356CrossRef Viacha V, Eliopoulou M, Haidas S, Beratis NG (2004) Correlation of cerebrospinal fluid betal-glucuronidase activity with plasma methotrexate concentrations in leukemic children receiving high-dose methotrexate. Pediatr Blood Cancer 42:350–356CrossRef
32.
go back to reference Chiaretti A, Ruggiero A, Coccia P et al (2011) Expression of liquoral neuroprotection markers in children with acute lymphoblastic leukemia. Leukemia Res 35:1467–1471CrossRef Chiaretti A, Ruggiero A, Coccia P et al (2011) Expression of liquoral neuroprotection markers in children with acute lymphoblastic leukemia. Leukemia Res 35:1467–1471CrossRef
33.
go back to reference Van Gool SW, De Meyer G, van de Voorde A, Vanmechelen E, Vanderstichele H (2004) Neurotoxicity marker profiles in the CSF are not age-dependent but show variation in children treated for acute lymphoblastic leukemia. Neurotoxicology 25:471–480PubMedCrossRef Van Gool SW, De Meyer G, van de Voorde A, Vanmechelen E, Vanderstichele H (2004) Neurotoxicity marker profiles in the CSF are not age-dependent but show variation in children treated for acute lymphoblastic leukemia. Neurotoxicology 25:471–480PubMedCrossRef
34.
go back to reference Krawczuk-Rybak M, Grabowska A, Protal PT, Muszynska-Roslan K, Braszko J (2012) Intellectual functioning of childhood leukemia survivors – relation to Tau protein – a marker of white matter injury. Adv Med Sci 57:266–272PubMedCrossRef Krawczuk-Rybak M, Grabowska A, Protal PT, Muszynska-Roslan K, Braszko J (2012) Intellectual functioning of childhood leukemia survivors – relation to Tau protein – a marker of white matter injury. Adv Med Sci 57:266–272PubMedCrossRef
35.
go back to reference Caron JE, Krull KR, Hockenberry M et al (2009) Oxidative stress and executive function in children receiving chemotherapy for acute lymphoblastic leukemia. Pediatr Blood Cancer 53:551–556PubMedPubMedCentralCrossRef Caron JE, Krull KR, Hockenberry M et al (2009) Oxidative stress and executive function in children receiving chemotherapy for acute lymphoblastic leukemia. Pediatr Blood Cancer 53:551–556PubMedPubMedCentralCrossRef
36.
go back to reference Taylor OA, Hockenberry MJ, McCarthy K et al (2015) Evaluation of biomarkers of oxidative stress and apoptosis in patients with severe methotrexate neurotoxicity: a case series. J Pediatr Oncol Nurs 32:320–325PubMedCrossRef Taylor OA, Hockenberry MJ, McCarthy K et al (2015) Evaluation of biomarkers of oxidative stress and apoptosis in patients with severe methotrexate neurotoxicity: a case series. J Pediatr Oncol Nurs 32:320–325PubMedCrossRef
37.
go back to reference Cole PD, Beckwith KA, Vijayanathan V et al (2009) Folate homeostasis in cerebrospinal fluid during therapy for acute lymphoblastic leukemia. Pediatr Neurol 40:34–41PubMedCrossRef Cole PD, Beckwith KA, Vijayanathan V et al (2009) Folate homeostasis in cerebrospinal fluid during therapy for acute lymphoblastic leukemia. Pediatr Neurol 40:34–41PubMedCrossRef
38.
go back to reference DeVita V, Hellman S, Rosenberg S (2005) Cancer: principles&practice of oncology, 7th edn, pp 332–422 DeVita V, Hellman S, Rosenberg S (2005) Cancer: principles&practice of oncology, 7th edn, pp 332–422
39.
go back to reference Seigers R, Fardell JE (2011) Neurobiological basis of chemotherapy-induced cognitive impairment: a review of rodent research. Neurosci Biobehav Rev 35:729–741PubMedCrossRef Seigers R, Fardell JE (2011) Neurobiological basis of chemotherapy-induced cognitive impairment: a review of rodent research. Neurosci Biobehav Rev 35:729–741PubMedCrossRef
40.
go back to reference Seigers R, Schagen SB, Beerling W et al (2008) Long-lasting suppression of hippocampal cell proliferation and impaired cognitive performance by methotrexate in the rat. Behav Brain Res 186:168–175PubMedCrossRef Seigers R, Schagen SB, Beerling W et al (2008) Long-lasting suppression of hippocampal cell proliferation and impaired cognitive performance by methotrexate in the rat. Behav Brain Res 186:168–175PubMedCrossRef
41.
go back to reference Seigers R, Schagen SB, Coppens CM et al (2009) Methotrexate decreases hippocampal cell proliferation and induces memory deficits in rats. Behav Brain Res 2:279–284CrossRef Seigers R, Schagen SB, Coppens CM et al (2009) Methotrexate decreases hippocampal cell proliferation and induces memory deficits in rats. Behav Brain Res 2:279–284CrossRef
42.
go back to reference Geller HM, Cheng KY, Goldsmith NK et al (2001) Oxidative stress mediates neuronal DNA damage and apoptosis in response to cytosine arabinoside. J Neurochem 78:265–275PubMedCrossRef Geller HM, Cheng KY, Goldsmith NK et al (2001) Oxidative stress mediates neuronal DNA damage and apoptosis in response to cytosine arabinoside. J Neurochem 78:265–275PubMedCrossRef
43.
go back to reference Husain K, Whitworth C, Hazelrigg S, Rybak L (2003) Carboplatin-induced oxidative injury in rat inferior colliculus. Int J Toxicol 22:335–342PubMedCrossRef Husain K, Whitworth C, Hazelrigg S, Rybak L (2003) Carboplatin-induced oxidative injury in rat inferior colliculus. Int J Toxicol 22:335–342PubMedCrossRef
44.
go back to reference Husain K, Whitworth C, Somani SM, Rybak LP (2001) Carboplatin-induced oxidative stress in rat cochlea. Hear Res 159:14–22PubMedCrossRef Husain K, Whitworth C, Somani SM, Rybak LP (2001) Carboplatin-induced oxidative stress in rat cochlea. Hear Res 159:14–22PubMedCrossRef
45.
go back to reference Oboh G, Ogunruku OO (2010) Cyclophosphamide-induced oxidative stress in brain: protective effect of hot short pepper (Capsicum frutescens L. var. abbreviatum). Exp Toxicol Pathol 63:227–233CrossRef Oboh G, Ogunruku OO (2010) Cyclophosphamide-induced oxidative stress in brain: protective effect of hot short pepper (Capsicum frutescens L. var. abbreviatum). Exp Toxicol Pathol 63:227–233CrossRef
46.
go back to reference Koros C, Kitraki E (2009) Neurofilament isoform alterations in the rat cerebellum following cytosine arabinoside administration. Toxicol Lett 189:215–218PubMedCrossRef Koros C, Kitraki E (2009) Neurofilament isoform alterations in the rat cerebellum following cytosine arabinoside administration. Toxicol Lett 189:215–218PubMedCrossRef
47.
go back to reference Joshi G, Sultana R, Tangpong J et al (2005) Free radical mediated oxidative stress and toxic side effects in brain induced by the anti cancer drug adriamycin: insight into chemobrain. Free Radic Res 39:1147–1154PubMedCrossRef Joshi G, Sultana R, Tangpong J et al (2005) Free radical mediated oxidative stress and toxic side effects in brain induced by the anti cancer drug adriamycin: insight into chemobrain. Free Radic Res 39:1147–1154PubMedCrossRef
48.
go back to reference Montilla P, Tunez I, Munoz MC, Soria JV, Lopez A (1997) Antioxidative effect of melatonin in rat brain oxidative stress induced by Adriamycin. Rev Esp Fisiol 53:301–305PubMed Montilla P, Tunez I, Munoz MC, Soria JV, Lopez A (1997) Antioxidative effect of melatonin in rat brain oxidative stress induced by Adriamycin. Rev Esp Fisiol 53:301–305PubMed
49.
go back to reference Öz E, Ilhan MN (2006) Effects of melatonin in reducing the toxic effects of doxorubicin. Mol Cell Biochem 286:11–15PubMedCrossRef Öz E, Ilhan MN (2006) Effects of melatonin in reducing the toxic effects of doxorubicin. Mol Cell Biochem 286:11–15PubMedCrossRef
50.
go back to reference Rajamani R, Muthuvel A, Senthilvelan M, Sheeladevi R (2006) Oxidative stress induced by methotrexate alone and in the presence of methanol in discrete regions of the rodent brain, retina and optic nerve. Toxicol Lett 165:265–273PubMedCrossRef Rajamani R, Muthuvel A, Senthilvelan M, Sheeladevi R (2006) Oxidative stress induced by methotrexate alone and in the presence of methanol in discrete regions of the rodent brain, retina and optic nerve. Toxicol Lett 165:265–273PubMedCrossRef
51.
go back to reference Uzar E, Koyuncuoglu HR, Uz E et al (2006) The activities of antioxidant enzymes and the level of malondialdehyde in cerebellum of rats subjected to methotrexate: protective effect of caffeic acid phenethyl ester. Mol Cell Biochem 291:63–68PubMedCrossRef Uzar E, Koyuncuoglu HR, Uz E et al (2006) The activities of antioxidant enzymes and the level of malondialdehyde in cerebellum of rats subjected to methotrexate: protective effect of caffeic acid phenethyl ester. Mol Cell Biochem 291:63–68PubMedCrossRef
52.
go back to reference Dietrich J, Han R, Yang Y, Mayer-Proschel M, Noble M (2006) CNS progenitor cells and oligodendrocytes are targets of chemotherapeutic agents in vitro and in vivo. J Biol 5:22.1–22.23CrossRef Dietrich J, Han R, Yang Y, Mayer-Proschel M, Noble M (2006) CNS progenitor cells and oligodendrocytes are targets of chemotherapeutic agents in vitro and in vivo. J Biol 5:22.1–22.23CrossRef
53.
go back to reference Dietrich J, Prust M, Kaiser J (2015) Chemotherapy, cognitive impairment and hippocampal toxicity. Neuroscience 308:224–232CrossRef Dietrich J, Prust M, Kaiser J (2015) Chemotherapy, cognitive impairment and hippocampal toxicity. Neuroscience 308:224–232CrossRef
54.
go back to reference Han R, Yang YM, Dietrich J et al (2008) Systemic 5-fluorouracil treatment causes a syndrome of delayed myelin destruction in the central nervous system. J Biol 7:12.1–12.22CrossRef Han R, Yang YM, Dietrich J et al (2008) Systemic 5-fluorouracil treatment causes a syndrome of delayed myelin destruction in the central nervous system. J Biol 7:12.1–12.22CrossRef
55.
go back to reference Mignone RG, Weber ET (2006) Potent inhibition of cell proliferation in the hippocampal dentate gyrus of mice by the chemotherapeutic drug thioTEPA. Brain Res 1111:26–29PubMedCrossRef Mignone RG, Weber ET (2006) Potent inhibition of cell proliferation in the hippocampal dentate gyrus of mice by the chemotherapeutic drug thioTEPA. Brain Res 1111:26–29PubMedCrossRef
56.
go back to reference Mondie CM, Vandergrift KA, Wilson CL, Gulinello ME, Weber ET (2010) The chemotherapy agent, thioTEPA, yields long-term impairment of hippocampal cell proliferation and memory deficits but not depression-related behaviors in mice. Behav Brain Res 209:66–72PubMedPubMedCentralCrossRef Mondie CM, Vandergrift KA, Wilson CL, Gulinello ME, Weber ET (2010) The chemotherapy agent, thioTEPA, yields long-term impairment of hippocampal cell proliferation and memory deficits but not depression-related behaviors in mice. Behav Brain Res 209:66–72PubMedPubMedCentralCrossRef
57.
go back to reference Mustafa S, Walker A, Bennett G, Wigmore PM (2008) 5-Fluorouracil chemotherapy affects spatial working memory and newborn neurons in the adult rat hippocampus. Eur J Neurosci 28:323–330PubMedCrossRef Mustafa S, Walker A, Bennett G, Wigmore PM (2008) 5-Fluorouracil chemotherapy affects spatial working memory and newborn neurons in the adult rat hippocampus. Eur J Neurosci 28:323–330PubMedCrossRef
58.
go back to reference Yang M, Kim JS, Song MS et al (2010) Cyclophosphamide impairs hippocampus-dependent learning and memory in adult mice: possible involvement of hippocampal neurogenesis in chemotherapy-induced memory deficits. Neurobiol Learn Mem 93:487–494PubMedCrossRef Yang M, Kim JS, Song MS et al (2010) Cyclophosphamide impairs hippocampus-dependent learning and memory in adult mice: possible involvement of hippocampal neurogenesis in chemotherapy-induced memory deficits. Neurobiol Learn Mem 93:487–494PubMedCrossRef
59.
go back to reference Bittigau P, Sifringer M, Pohl D (1999) Apoptotic neurodegeneration following trauma is markedly enhanced in the immature brain. Ann Neurol 45:724–735PubMedCrossRef Bittigau P, Sifringer M, Pohl D (1999) Apoptotic neurodegeneration following trauma is markedly enhanced in the immature brain. Ann Neurol 45:724–735PubMedCrossRef
60.
go back to reference Bossy-Wetzel E, Barsoum MJ, Godzik A et al (2003) Mitochondrial function in apoptosis, neurodegeneration and aging. Curr Opin Cell Biol 15:706–716PubMedCrossRef Bossy-Wetzel E, Barsoum MJ, Godzik A et al (2003) Mitochondrial function in apoptosis, neurodegeneration and aging. Curr Opin Cell Biol 15:706–716PubMedCrossRef
61.
go back to reference Lipton SA, Rosenberg PA (1994) Excitatory amino acids as a final common pathway for neurologic disorders. N Engl J Med 330:613–622PubMedCrossRef Lipton SA, Rosenberg PA (1994) Excitatory amino acids as a final common pathway for neurologic disorders. N Engl J Med 330:613–622PubMedCrossRef
62.
go back to reference Murphy AN, Fiskum G, Beal MF (1999) Mitochondria in neurodegeneration: bioenergetic function in cell life and death. J Cereb Blood Flow Metab 19:231–245PubMedCrossRef Murphy AN, Fiskum G, Beal MF (1999) Mitochondria in neurodegeneration: bioenergetic function in cell life and death. J Cereb Blood Flow Metab 19:231–245PubMedCrossRef
63.
go back to reference Rothman SM, Olney JW (1995) Excitotoxicity and the NMDA receptor—still lethal after eight years. Trends Neurosci 18:57–58PubMed Rothman SM, Olney JW (1995) Excitotoxicity and the NMDA receptor—still lethal after eight years. Trends Neurosci 18:57–58PubMed
64.
go back to reference Lee JM, Zipfel GJ, Choi DW (1999) The changing landscape of ischaemic brain injury mechanisms. Nature 399(suppl):A7–A14PubMedCrossRef Lee JM, Zipfel GJ, Choi DW (1999) The changing landscape of ischaemic brain injury mechanisms. Nature 399(suppl):A7–A14PubMedCrossRef
65.
go back to reference Bonfoco E, Krainc D, Ankarcrona M et al (1995) Apoptosis and necrosis: two distinct events induced, respectively, by mild and intense insults with N-methyl-D-aspartate or nitric oxide/superoxide in cortical cell cultures. Proc Natl Acad Sci U S A 92:7162–7166PubMedPubMedCentralCrossRef Bonfoco E, Krainc D, Ankarcrona M et al (1995) Apoptosis and necrosis: two distinct events induced, respectively, by mild and intense insults with N-methyl-D-aspartate or nitric oxide/superoxide in cortical cell cultures. Proc Natl Acad Sci U S A 92:7162–7166PubMedPubMedCentralCrossRef
66.
go back to reference Northington FJ, Ferriero DM, Graham EM et al (2001) Early neurodegeneration after hypoxia-ischemia in neonatal rat is necrosis while delayed neuronal death is apoptosis. Neurobiol Dis 8:207–219PubMedCrossRef Northington FJ, Ferriero DM, Graham EM et al (2001) Early neurodegeneration after hypoxia-ischemia in neonatal rat is necrosis while delayed neuronal death is apoptosis. Neurobiol Dis 8:207–219PubMedCrossRef
67.
go back to reference Pohl D, Bittigau P, Ishimaru MJ et al (1999) NMDA antagonists and apoptotic cell death triggered by head trauma in developing rat brain. Proc Natl Acad Sci U S A 96:2508–2513PubMedPubMedCentralCrossRef Pohl D, Bittigau P, Ishimaru MJ et al (1999) NMDA antagonists and apoptotic cell death triggered by head trauma in developing rat brain. Proc Natl Acad Sci U S A 96:2508–2513PubMedPubMedCentralCrossRef
68.
go back to reference Rzeski W, Pruskil S, Macke A et al (2004) Anticancer agents are potent neurotoxins in vitro and in vivo. Ann Neurol 56:351–360PubMedCrossRef Rzeski W, Pruskil S, Macke A et al (2004) Anticancer agents are potent neurotoxins in vitro and in vivo. Ann Neurol 56:351–360PubMedCrossRef
69.
go back to reference Courtney MJ, Coffey ET (1999) The mechanism of Ara-C-induced apoptosis of differentiating cerebellar granule neurons. Eur J Neurosci 11:1073–1084PubMedCrossRef Courtney MJ, Coffey ET (1999) The mechanism of Ara-C-induced apoptosis of differentiating cerebellar granule neurons. Eur J Neurosci 11:1073–1084PubMedCrossRef
70.
go back to reference Wick A, Wick W, Hirrlinger J et al (2004) Chemotherapy-induced cell death in primary cerebellar granule neurons but not in astrocytes: in vitro paradigm of differential neurotoxicity. J Neurochem 91:1067–1074PubMedCrossRef Wick A, Wick W, Hirrlinger J et al (2004) Chemotherapy-induced cell death in primary cerebellar granule neurons but not in astrocytes: in vitro paradigm of differential neurotoxicity. J Neurochem 91:1067–1074PubMedCrossRef
71.
go back to reference Gregorios JB, Gregorios AB, Mora J et al (1989) Morphologic alterations in rat brain following systemic and intraventricular methotrexate injection: light and electron microscopic studies. J Neuropathol Exp Neurol 48:33–47PubMedCrossRef Gregorios JB, Gregorios AB, Mora J et al (1989) Morphologic alterations in rat brain following systemic and intraventricular methotrexate injection: light and electron microscopic studies. J Neuropathol Exp Neurol 48:33–47PubMedCrossRef
72.
go back to reference Das S, Basu A (2008) Inflammation: a new candidate in modulating adult neurogenesis. J Neurosci Res 86:1199–1208PubMedCrossRef Das S, Basu A (2008) Inflammation: a new candidate in modulating adult neurogenesis. J Neurosci Res 86:1199–1208PubMedCrossRef
73.
go back to reference de Koning BA, van Dieren JM, Lindenbergh-Kortleve DJ et al (2006) Contributions of mucosal immune cells to methotrexate-induced mucositis. Int Immunol 18:941–949PubMedCrossRef de Koning BA, van Dieren JM, Lindenbergh-Kortleve DJ et al (2006) Contributions of mucosal immune cells to methotrexate-induced mucositis. Int Immunol 18:941–949PubMedCrossRef
74.
go back to reference De Visser KE, Eichten A, Coussens LM (2006) Paradoxical roles of the immune system during cancer development. Nat Rev Cancer 6:24–37PubMedCrossRef De Visser KE, Eichten A, Coussens LM (2006) Paradoxical roles of the immune system during cancer development. Nat Rev Cancer 6:24–37PubMedCrossRef
75.
go back to reference Ekdahl CT, Claasen JH, Bonde S, Kokaia Z, Lindvall O (2003) Inflammation is detrimental for neurogenesis in adult brain. Proc Natl Acad Sci U S A 100:13632–13637PubMedPubMedCentralCrossRef Ekdahl CT, Claasen JH, Bonde S, Kokaia Z, Lindvall O (2003) Inflammation is detrimental for neurogenesis in adult brain. Proc Natl Acad Sci U S A 100:13632–13637PubMedPubMedCentralCrossRef
76.
go back to reference Seruga B, Zhang H, Bernstein LJ, Tannock IF (2008) Cytokines and their relationship to the symptoms and outcome of cancer. Nat Rev Cancer 8:887–899PubMedCrossRef Seruga B, Zhang H, Bernstein LJ, Tannock IF (2008) Cytokines and their relationship to the symptoms and outcome of cancer. Nat Rev Cancer 8:887–899PubMedCrossRef
77.
go back to reference Wilson CJ, Finch CE, Cohen HJ (2002) Cytokines and cognition—the case for a head-to-toe inflammatory paradigm. J Am Geriatr Soc 50:2041–2056PubMedCrossRef Wilson CJ, Finch CE, Cohen HJ (2002) Cytokines and cognition—the case for a head-to-toe inflammatory paradigm. J Am Geriatr Soc 50:2041–2056PubMedCrossRef
78.
go back to reference de Vos FY, Willemse PH, De Vries EG, Gietema JA (2004) Endothelial cell effects of cytotoxics: balance between desired and unwanted effects. Cancer Treat Rev 30:495–513PubMedCrossRef de Vos FY, Willemse PH, De Vries EG, Gietema JA (2004) Endothelial cell effects of cytotoxics: balance between desired and unwanted effects. Cancer Treat Rev 30:495–513PubMedCrossRef
79.
go back to reference Mizusawa S, Kondoh Y, Murakami M et al (1988) Effect of methotrexate on local cerebral blood flow in conscious rats. Jpn J Pharmacol 48:499–501PubMedCrossRef Mizusawa S, Kondoh Y, Murakami M et al (1988) Effect of methotrexate on local cerebral blood flow in conscious rats. Jpn J Pharmacol 48:499–501PubMedCrossRef
80.
go back to reference Seigers R, Timmermans J, van der Horn HJ et al (2010) Methotrexate reduces hippocampal blood vessel density and activates microglia in rats but does not elevate central cytokine release. Behav Brain Res 207:265–272PubMedCrossRef Seigers R, Timmermans J, van der Horn HJ et al (2010) Methotrexate reduces hippocampal blood vessel density and activates microglia in rats but does not elevate central cytokine release. Behav Brain Res 207:265–272PubMedCrossRef
81.
go back to reference Theruvath AJ, Ilivitzki A, Muehe A et al (2017) A PET/MRI imaging approach for the integrated assessment of chemotherapy-induced brain, heart, and bone injuries in pediatric cancer survivors: a pilot study. Radiology 285:971–979PubMedCrossRef Theruvath AJ, Ilivitzki A, Muehe A et al (2017) A PET/MRI imaging approach for the integrated assessment of chemotherapy-induced brain, heart, and bone injuries in pediatric cancer survivors: a pilot study. Radiology 285:971–979PubMedCrossRef
82.
83.
go back to reference Saunders NR, Knott GW, Dziegielewska KM (2000) Barriers in the immature brain. Cell Mol Neurobiol 20:29–40PubMedCrossRef Saunders NR, Knott GW, Dziegielewska KM (2000) Barriers in the immature brain. Cell Mol Neurobiol 20:29–40PubMedCrossRef
84.
go back to reference Virgintino D, Errede M, Girolamo F et al (2008) Fetal blood-brain barrier P-glycoprotein contributes to brain protection during human development. J Neuropathol Exp Neurol 67:50–61PubMedCrossRef Virgintino D, Errede M, Girolamo F et al (2008) Fetal blood-brain barrier P-glycoprotein contributes to brain protection during human development. J Neuropathol Exp Neurol 67:50–61PubMedCrossRef
85.
go back to reference de Vries NA, Beijnen JH, Boogerd W, van Tellingen O (2006) Blood–brain barrier and chemotherapeutic treatment of brain tumors. Expert Rev Neurother 6:1199–1209PubMedCrossRef de Vries NA, Beijnen JH, Boogerd W, van Tellingen O (2006) Blood–brain barrier and chemotherapeutic treatment of brain tumors. Expert Rev Neurother 6:1199–1209PubMedCrossRef
86.
go back to reference Perry A, Schmidt RE (2006) Cancer therapy-associated CNS neuropathology: an update and review of the literature. Acta Neuropathol 111:197–212PubMedCrossRef Perry A, Schmidt RE (2006) Cancer therapy-associated CNS neuropathology: an update and review of the literature. Acta Neuropathol 111:197–212PubMedCrossRef
87.
go back to reference Briones TL, Woods J (2011) Chemotherapy-induced cognitive impairment is associated with decreases in cell proliferation and histone modifications. BMC Neurosci 12:124PubMedPubMedCentralCrossRef Briones TL, Woods J (2011) Chemotherapy-induced cognitive impairment is associated with decreases in cell proliferation and histone modifications. BMC Neurosci 12:124PubMedPubMedCentralCrossRef
88.
go back to reference Briones TL, Woods J (2013) Dysregulation in myelination mediated by persistent neuroinflammation: possible mechanisms in chemotherapy-related cognitive impairment. Brain Behav Immun 35:23–32PubMedCrossRef Briones TL, Woods J (2013) Dysregulation in myelination mediated by persistent neuroinflammation: possible mechanisms in chemotherapy-related cognitive impairment. Brain Behav Immun 35:23–32PubMedCrossRef
89.
go back to reference Lyons L, Elbeltagy M, Bennett G, Wigmore P (2011) The effects of cyclophosphamide on hippocampal cell proliferation and spatial working memory in rat. PLoS One 6:e21445PubMedPubMedCentralCrossRef Lyons L, Elbeltagy M, Bennett G, Wigmore P (2011) The effects of cyclophosphamide on hippocampal cell proliferation and spatial working memory in rat. PLoS One 6:e21445PubMedPubMedCentralCrossRef
90.
go back to reference Lee GD, Longo DI, Wang Y et al (2006) Transient improvement in cognitive function and synaptic plasticity in rats following cancer chemotherapy. Clin Cancer Res 12:198–205PubMedCrossRef Lee GD, Longo DI, Wang Y et al (2006) Transient improvement in cognitive function and synaptic plasticity in rats following cancer chemotherapy. Clin Cancer Res 12:198–205PubMedCrossRef
91.
go back to reference Xiao R, Yu HL, Zhao HF et al (2007) Developmental neurotoxicity role of cyclophosphamide onpost-neural tube closure of rodents in vitro and in vivo. Int J Dev Neurosci 25:531–537PubMedCrossRef Xiao R, Yu HL, Zhao HF et al (2007) Developmental neurotoxicity role of cyclophosphamide onpost-neural tube closure of rodents in vitro and in vivo. Int J Dev Neurosci 25:531–537PubMedCrossRef
92.
go back to reference Avella D, Pisu MB, Roda E, Gravati M, Bernocchi G (2006) Reorganization of the rat cerebellar cortex during postnatal development following cisplatin treatment. Exp Neurol 201:131–143PubMedCrossRef Avella D, Pisu MB, Roda E, Gravati M, Bernocchi G (2006) Reorganization of the rat cerebellar cortex during postnatal development following cisplatin treatment. Exp Neurol 201:131–143PubMedCrossRef
93.
go back to reference Andres AL, Gong C, Di K, Bota DA (2014) Low-doses of cisplatin injure hippocampal synapses: a mechanism for ‘chemo’ brain? Exp Neurol 255:137–144PubMedPubMedCentralCrossRef Andres AL, Gong C, Di K, Bota DA (2014) Low-doses of cisplatin injure hippocampal synapses: a mechanism for ‘chemo’ brain? Exp Neurol 255:137–144PubMedPubMedCentralCrossRef
94.
go back to reference Cerri S, Piccolini VM, Santin G et al (2011) The developmental neurotoxicity study of platinum compounds: effects of cisplatin versus a novel Pt(II) complex on rat cerebellum. Neurotoxicol Teratol 33:273–281PubMedCrossRef Cerri S, Piccolini VM, Santin G et al (2011) The developmental neurotoxicity study of platinum compounds: effects of cisplatin versus a novel Pt(II) complex on rat cerebellum. Neurotoxicol Teratol 33:273–281PubMedCrossRef
95.
go back to reference Gopal KV, Wu C, Shrestha B et al (2012) D-Methionine protects against cisplatin-induced neurotoxicity in cortical networks. Neurotoxicol Teratol 34:495–504PubMedCrossRef Gopal KV, Wu C, Shrestha B et al (2012) D-Methionine protects against cisplatin-induced neurotoxicity in cortical networks. Neurotoxicol Teratol 34:495–504PubMedCrossRef
96.
go back to reference Sugimoto S, Yamamoto YL, Nagahiro S, Diksic M (1995) Permeability change and brain tissue damage after intracarotid administration of cisplatin studied by double-tracer autoradiography in rats. J Neuro-Oncol 24:229–240CrossRef Sugimoto S, Yamamoto YL, Nagahiro S, Diksic M (1995) Permeability change and brain tissue damage after intracarotid administration of cisplatin studied by double-tracer autoradiography in rats. J Neuro-Oncol 24:229–240CrossRef
97.
go back to reference Piccolini VM, Cerri S, Romanelli E, Bernocchi G (2012) Interactions of neurotransmitter systems duringpostnatal development of the rat hippocampal formation: effects of cisplatin. Exp Neurol 234:239–252PubMedCrossRef Piccolini VM, Cerri S, Romanelli E, Bernocchi G (2012) Interactions of neurotransmitter systems duringpostnatal development of the rat hippocampal formation: effects of cisplatin. Exp Neurol 234:239–252PubMedCrossRef
98.
go back to reference Turan MI, Cayir A, Cetin N et al (2014) An investigation of the effect of thiamine pyrophosphateon cisplatin-induced oxidative stress and DNA damage in rat brain tissue compared with thiamine: thiamine and thiamine pyrophosphate effects on cisplatin neurotoxicity. Hum Exp Toxicol 33:14–21PubMedCrossRef Turan MI, Cayir A, Cetin N et al (2014) An investigation of the effect of thiamine pyrophosphateon cisplatin-induced oxidative stress and DNA damage in rat brain tissue compared with thiamine: thiamine and thiamine pyrophosphate effects on cisplatin neurotoxicity. Hum Exp Toxicol 33:14–21PubMedCrossRef
99.
go back to reference van der Plas E, Schachar RJ, Hitzler J et al (2016) Brain structure, working memory and response inhibition in childhood leukemia survivors. Brain Behav 7:e00621PubMedPubMedCentralCrossRef van der Plas E, Schachar RJ, Hitzler J et al (2016) Brain structure, working memory and response inhibition in childhood leukemia survivors. Brain Behav 7:e00621PubMedPubMedCentralCrossRef
100.
go back to reference Cossaart N, SantaCruz KS, Preston D, Johnson P, Skikne BS (2003) Fatal chemotherapy-induced encephalopathy following high-dose therapy for metastatic breast cancer: a case report and review of the literature. Bone Marrow Transplant 31:57–60PubMedCrossRef Cossaart N, SantaCruz KS, Preston D, Johnson P, Skikne BS (2003) Fatal chemotherapy-induced encephalopathy following high-dose therapy for metastatic breast cancer: a case report and review of the literature. Bone Marrow Transplant 31:57–60PubMedCrossRef
101.
go back to reference Cruz-Sanchez FF, Artigas J, Cervos-Navarro J, Rossi ML, Ferszt R (1991) Brain lesions following combined treatment with methotrexate and craniospinal irradiation. J Neuro-Oncol 10:165–171CrossRef Cruz-Sanchez FF, Artigas J, Cervos-Navarro J, Rossi ML, Ferszt R (1991) Brain lesions following combined treatment with methotrexate and craniospinal irradiation. J Neuro-Oncol 10:165–171CrossRef
102.
go back to reference Fassas AB, Gattani AM, Morgello S (1994) Cerebral demyelination with 5-fluorouracil and levamisole. Cancer Investig 12:379–383CrossRef Fassas AB, Gattani AM, Morgello S (1994) Cerebral demyelination with 5-fluorouracil and levamisole. Cancer Investig 12:379–383CrossRef
103.
go back to reference Liu HM, Maurer HS, Vongsvivut S, Conway JJ (1978) Methotrexate encephalopathy. A neuropathologic study. Hum Pathol 9:635–648PubMedCrossRef Liu HM, Maurer HS, Vongsvivut S, Conway JJ (1978) Methotrexate encephalopathy. A neuropathologic study. Hum Pathol 9:635–648PubMedCrossRef
104.
go back to reference Moore-Maxwell CA, Datto MB, Hulette CM (2004) Chemotherapy-induced toxic leukoencephalopathy causes a wide range of symptoms: a series of four autopsies. Mod Pathol 17:241–247PubMedCrossRef Moore-Maxwell CA, Datto MB, Hulette CM (2004) Chemotherapy-induced toxic leukoencephalopathy causes a wide range of symptoms: a series of four autopsies. Mod Pathol 17:241–247PubMedCrossRef
105.
go back to reference Antunes NL, Souweidane MM, Lis E, Rosenblum MK, Steinherz PG (2002) Methotrexate leukoencephalopathy presenting as Kluver–Bucy syndrome and uncinate seizures. Pediatr Neurol 26:305–308PubMedCrossRef Antunes NL, Souweidane MM, Lis E, Rosenblum MK, Steinherz PG (2002) Methotrexate leukoencephalopathy presenting as Kluver–Bucy syndrome and uncinate seizures. Pediatr Neurol 26:305–308PubMedCrossRef
106.
go back to reference DeAngelis LM, Seiferheld W, Schold SC, Fisher B, Schultz CJ (2002) Combination chemotherapy and radiotherapy for primary central nervous system lymphoma: radiation therapy oncology group study 93–10. J Clin Oncol 20:4643–4648PubMedCrossRef DeAngelis LM, Seiferheld W, Schold SC, Fisher B, Schultz CJ (2002) Combination chemotherapy and radiotherapy for primary central nervous system lymphoma: radiation therapy oncology group study 93–10. J Clin Oncol 20:4643–4648PubMedCrossRef
107.
go back to reference Lai R, Abrey LE, Rosenblum MK, DeAngelis LM (2004) Treatment-induced leukoencephalopathy in primary CNS lymphoma: a clinical and autopsy study. Neurology 62:451–456PubMedCrossRef Lai R, Abrey LE, Rosenblum MK, DeAngelis LM (2004) Treatment-induced leukoencephalopathy in primary CNS lymphoma: a clinical and autopsy study. Neurology 62:451–456PubMedCrossRef
108.
go back to reference Rubinstein LJ, Herman MM, Long TF, Wilbur JR (1975) Disseminated necrotizing leukoencephalopathy: a complication of treated central nervous system leukemia and lymphoma. Cancer 35:291–305PubMedCrossRef Rubinstein LJ, Herman MM, Long TF, Wilbur JR (1975) Disseminated necrotizing leukoencephalopathy: a complication of treated central nervous system leukemia and lymphoma. Cancer 35:291–305PubMedCrossRef
109.
go back to reference Stone JA, Castillo M, Mukherji SK (1999) Leukoencephalopathy complicating an Ommaya reservoir and chemotherapy. Neuroradiology 41:134–136PubMedCrossRef Stone JA, Castillo M, Mukherji SK (1999) Leukoencephalopathy complicating an Ommaya reservoir and chemotherapy. Neuroradiology 41:134–136PubMedCrossRef
110.
go back to reference Abelson HT (1978) Methotrexate and central nervous system toxicity. Cancer Treat Rep 2:1999–2001 Abelson HT (1978) Methotrexate and central nervous system toxicity. Cancer Treat Rep 2:1999–2001
111.
go back to reference Allen JC, Rosen G, Mehta BM, Horten B (1980) Leukoencephalopathy following high-dose iv methotrexate chemotherapy with leucovorin rescue. Cancer Treat Rep 64:1261–1273PubMed Allen JC, Rosen G, Mehta BM, Horten B (1980) Leukoencephalopathy following high-dose iv methotrexate chemotherapy with leucovorin rescue. Cancer Treat Rep 64:1261–1273PubMed
112.
go back to reference Omuro AM, DeAngelis LM, Yahalom J, Abrey LE (2005) Chemotherapy for primary CNS lymphoma: an intent-to-treat analysis with complete follow-up. Neurology 64:69-74PubMedCrossRef Omuro AM, DeAngelis LM, Yahalom J, Abrey LE (2005) Chemotherapy for primary CNS lymphoma: an intent-to-treat analysis with complete follow-up. Neurology 64:69-74PubMedCrossRef
113.
go back to reference Lovblad K, Kelkar P, Ozdoba C et al (1998) Pure methotrexate encephalopathy presenting with seizures: CT and MRI features. Pediatr Radiol 28:86–91PubMedCrossRef Lovblad K, Kelkar P, Ozdoba C et al (1998) Pure methotrexate encephalopathy presenting with seizures: CT and MRI features. Pediatr Radiol 28:86–91PubMedCrossRef
114.
go back to reference de Waal R, Algra PR, Heimans JJ, Wolbers JG, Scheltens P (1993) Methotrexate induced brain necrosis and severe leukoencephalopathy due to disconnection of an Ommaya device. J Neuro-Oncol 15:269–273CrossRef de Waal R, Algra PR, Heimans JJ, Wolbers JG, Scheltens P (1993) Methotrexate induced brain necrosis and severe leukoencephalopathy due to disconnection of an Ommaya device. J Neuro-Oncol 15:269–273CrossRef
115.
go back to reference Packer RJ, Zimmerman RA, Rosenstock J et al (1981) Focal encephalopathy following methotrexate therapy. Administration via a misplaced intraventricular catheter. Arch Neurol 38:450–452PubMedCrossRef Packer RJ, Zimmerman RA, Rosenstock J et al (1981) Focal encephalopathy following methotrexate therapy. Administration via a misplaced intraventricular catheter. Arch Neurol 38:450–452PubMedCrossRef
116.
go back to reference Colamaria V, Caraballo R, Borgna-Pignatti C et al (1990) Transient focal leukoencephalopathy following intraventricular methotrexate and cytarabine. A complication of the Ommaya reservoir: case report and review of the literature. Childs Nerv Syst 6:231–235PubMedCrossRef Colamaria V, Caraballo R, Borgna-Pignatti C et al (1990) Transient focal leukoencephalopathy following intraventricular methotrexate and cytarabine. A complication of the Ommaya reservoir: case report and review of the literature. Childs Nerv Syst 6:231–235PubMedCrossRef
117.
go back to reference Fouladi M, Chintagumpala M, Laningham FH et al (2004) White matter lesions detected by magnetic resonance imaging after radiotherapy and high-dose chemotherapy in children with medulloblastoma or primitive neuroectodermal tumor. J Clin Oncol 22:4551–4560PubMedCrossRef Fouladi M, Chintagumpala M, Laningham FH et al (2004) White matter lesions detected by magnetic resonance imaging after radiotherapy and high-dose chemotherapy in children with medulloblastoma or primitive neuroectodermal tumor. J Clin Oncol 22:4551–4560PubMedCrossRef
118.
go back to reference Rutkowski S, Bode U, Deinlein F et al (2005) Treatment of early childhood medulloblastoma by postoperative chemotherapy alone. N Engl J Med 352:978–986PubMedCrossRef Rutkowski S, Bode U, Deinlein F et al (2005) Treatment of early childhood medulloblastoma by postoperative chemotherapy alone. N Engl J Med 352:978–986PubMedCrossRef
119.
go back to reference Fouladi M, Langston J, Mulhern R et al (2000) Silent lacunar lesions detected by magnetic resonance imaging in children with brain tumors: a late sequela of therapy. J Clin Oncol 18:824-831PubMedCrossRef Fouladi M, Langston J, Mulhern R et al (2000) Silent lacunar lesions detected by magnetic resonance imaging in children with brain tumors: a late sequela of therapy. J Clin Oncol 18:824-831PubMedCrossRef
120.
go back to reference Rubnitz JE, Relling MV, Harrison PL et al (1998) Transient encephalopathy following high-dose methotrexate treatment in childhood acute lymphoblastic leukemia. Leukemia 12:1176–1181PubMedCrossRef Rubnitz JE, Relling MV, Harrison PL et al (1998) Transient encephalopathy following high-dose methotrexate treatment in childhood acute lymphoblastic leukemia. Leukemia 12:1176–1181PubMedCrossRef
121.
go back to reference Price RA, Jamieson PA (1975) The central nervous system in childhood leukemia. II. Subacute leukoencephalopathy. Cancer 35:306–318PubMedCrossRef Price RA, Jamieson PA (1975) The central nervous system in childhood leukemia. II. Subacute leukoencephalopathy. Cancer 35:306–318PubMedCrossRef
123.
go back to reference Rubinstein JL, Herman MM, Long TF, Wilbur JR (1975) Leukoencephalopathy following combined therapy of central nervous system leukemia and lymphoma. Acta Neuropathol Suppl (Berl) Suppl 6:251–255 Rubinstein JL, Herman MM, Long TF, Wilbur JR (1975) Leukoencephalopathy following combined therapy of central nervous system leukemia and lymphoma. Acta Neuropathol Suppl (Berl) Suppl 6:251–255
124.
go back to reference Atlas SW, Grossman RI, Packer RJ et al (1987) Magnetic resonance imaging diagnosis of disseminated necrotizing leukoencephalopathy. J Comput Tomogr 11:39–43PubMedCrossRef Atlas SW, Grossman RI, Packer RJ et al (1987) Magnetic resonance imaging diagnosis of disseminated necrotizing leukoencephalopathy. J Comput Tomogr 11:39–43PubMedCrossRef
125.
go back to reference Batara JF, Grossman SA (2003) Primary central nervous system lymphomas. Curr Opin Neurol 16:671–675PubMedCrossRef Batara JF, Grossman SA (2003) Primary central nervous system lymphomas. Curr Opin Neurol 16:671–675PubMedCrossRef
127.
go back to reference Omuro AM, Ben-Porat LS, Panageas KS et al (2005) Delayed neurotoxicity in primary central nervous system lymphoma. Arch Neurol 62:1595–1600PubMedCrossRef Omuro AM, Ben-Porat LS, Panageas KS et al (2005) Delayed neurotoxicity in primary central nervous system lymphoma. Arch Neurol 62:1595–1600PubMedCrossRef
128.
go back to reference Bashir R, Hochberg FH, Linggood RM, Hottleman K (1988) Pre-irradiation internal carotid artery BCNU in treatment of glioblastoma multiforme. J Neurosurg 68:917–919PubMedCrossRef Bashir R, Hochberg FH, Linggood RM, Hottleman K (1988) Pre-irradiation internal carotid artery BCNU in treatment of glioblastoma multiforme. J Neurosurg 68:917–919PubMedCrossRef
129.
130.
go back to reference Kleinschmidt-DeMasters BK, Geier JM (1989) Pathology of high-dose intra-arterial BCNU. Surg Neurol 31:435–443PubMedCrossRef Kleinschmidt-DeMasters BK, Geier JM (1989) Pathology of high-dose intra-arterial BCNU. Surg Neurol 31:435–443PubMedCrossRef
131.
go back to reference Rosenblum MK, Delattre JY, Walker RW, Shapiro WR (1989) Fatal necrotizing encephalopathy complicating treatment of malignant gliomas with intra-arterial BCNU and irradiation: a pathological study. J Neuro-Oncol 7:269–281CrossRef Rosenblum MK, Delattre JY, Walker RW, Shapiro WR (1989) Fatal necrotizing encephalopathy complicating treatment of malignant gliomas with intra-arterial BCNU and irradiation: a pathological study. J Neuro-Oncol 7:269–281CrossRef
132.
go back to reference Hinchey J, Chaves C, Appignani B et al (1996) A reversible posterior leukoencephalopathy syndrome. N Engl J Med 334:494–500PubMedCrossRef Hinchey J, Chaves C, Appignani B et al (1996) A reversible posterior leukoencephalopathy syndrome. N Engl J Med 334:494–500PubMedCrossRef
133.
go back to reference Kahana A, Rowley HA, Weinstein JM (2005) Cortical blindness: clinical and radiologic findings in reversible posterior leukoencephalopathy syndrome: case report and review of the literature. Ophthalmology 112:e7–e11PubMedCrossRef Kahana A, Rowley HA, Weinstein JM (2005) Cortical blindness: clinical and radiologic findings in reversible posterior leukoencephalopathy syndrome: case report and review of the literature. Ophthalmology 112:e7–e11PubMedCrossRef
134.
go back to reference Pavlakis SG, Frank Y, Chusid R (1999) Hypertensive encephalopathy, reversible occipitoparietal encephalopathy, or reversible posterior leukoencephalopathy: three names for an old syndrome. J Child Neurol 14:277–281PubMedCrossRef Pavlakis SG, Frank Y, Chusid R (1999) Hypertensive encephalopathy, reversible occipitoparietal encephalopathy, or reversible posterior leukoencephalopathy: three names for an old syndrome. J Child Neurol 14:277–281PubMedCrossRef
135.
go back to reference Rangi PS, Partridge WJ, Newlands ES, Waldman AD (2005) Posterior reversible encephalopathy syndrome: a possible late interaction between cytotoxic agents and general anaesthesia. Neuroradiology 47:586–590PubMedCrossRef Rangi PS, Partridge WJ, Newlands ES, Waldman AD (2005) Posterior reversible encephalopathy syndrome: a possible late interaction between cytotoxic agents and general anaesthesia. Neuroradiology 47:586–590PubMedCrossRef
136.
go back to reference Sanchez-Carpintero R, Narbona J, Lopez de Mesa R, Arbizu J, Sierrasesumaga L (2001) Transient posterior encephalopathy induced by chemotherapy in children. Pediatr Neurol 24:145–148PubMedCrossRef Sanchez-Carpintero R, Narbona J, Lopez de Mesa R, Arbizu J, Sierrasesumaga L (2001) Transient posterior encephalopathy induced by chemotherapy in children. Pediatr Neurol 24:145–148PubMedCrossRef
137.
go back to reference Schiff D, Lopes MB (2005) Neuropathological correlates of reversible posterior leukoencephalopathy. Neurocrit Care 2:303–305PubMedCrossRef Schiff D, Lopes MB (2005) Neuropathological correlates of reversible posterior leukoencephalopathy. Neurocrit Care 2:303–305PubMedCrossRef
138.
go back to reference Shin RK, Stern JW, Janss AJ, Hunter JV, Liu GT (2001) Reversible posterior leukoencephalopathy during the treatment of acute lymphoblastic leukemia. Neurology 56:388–391PubMedCrossRef Shin RK, Stern JW, Janss AJ, Hunter JV, Liu GT (2001) Reversible posterior leukoencephalopathy during the treatment of acute lymphoblastic leukemia. Neurology 56:388–391PubMedCrossRef
139.
go back to reference Stott VL, Hurrell MA, Anderson TJ (2005) Reversible posterior leukoencephalopathy syndrome: a misnomer reviewed. Intern Med J 35:83–90PubMedCrossRef Stott VL, Hurrell MA, Anderson TJ (2005) Reversible posterior leukoencephalopathy syndrome: a misnomer reviewed. Intern Med J 35:83–90PubMedCrossRef
140.
go back to reference Tam CS, Galanos J, Seymour JF et al (2004) Reversible posterior leukoencephalopathy syndrome complicating cytotoxic chemotherapy for hematologic malignancies. Am J Hematol 77:72–76PubMedCrossRef Tam CS, Galanos J, Seymour JF et al (2004) Reversible posterior leukoencephalopathy syndrome complicating cytotoxic chemotherapy for hematologic malignancies. Am J Hematol 77:72–76PubMedCrossRef
Metadata
Title
Chemotherapy and the pediatric brain
Author
Chrysanthy Ikonomidou
Publication date
01-12-2018
Publisher
Springer Berlin Heidelberg
Published in
Molecular and Cellular Pediatrics / Issue 1/2018
Electronic ISSN: 2194-7791
DOI
https://doi.org/10.1186/s40348-018-0087-0

Other articles of this Issue 1/2018

Molecular and Cellular Pediatrics 1/2018 Go to the issue