Skip to main content
Top
Published in: Immunologic Research 6/2022

20-07-2022 | Encephalopathy | Review

Role of inflammation and oxidative stress in chemotherapy-induced neurotoxicity

Authors: Pooja Gupta, Tavneet Kaur Makkar, Lavisha Goel, Monika Pahuja

Published in: Immunologic Research | Issue 6/2022

Login to get access

Abstract

Chemotherapeutic agents may adversely affect the nervous system, including the neural precursor cells as well as the white matter. Although the mechanisms are not completely understood, several hypotheses connecting inflammation and oxidative stress with neurotoxicity are now emerging. The proposed mechanisms differ depending on the class of drug. For example, toxicity due to cisplatin occurs due to activation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), which alters hippocampal long-term potentiation. Free radical injury is also involved in the cisplatin-mediated neurotoxicity as dysregulation of nuclear factor erythroid 2-related factor 2 (Nrf2) has been seen which protects against the free radical injury by regulating glutathione S-transferases and hemeoxygenase-1 (HO-1). Thus, correcting the imbalance between NF-κB and Nrf2/HO-1 pathways may alleviate cisplatin-induced neurotoxicity. With newer agents like bortezomib, peripheral neuropathy occurs due to up-regulation of TNF-α and IL-6 in the sensory neurons. Superoxide dismutase dysregulation is also involved in bortezomib-induced neuropathy. This article reviews the available literature on inflammation and oxidative stress in neurotoxicity caused by various classes of chemotherapeutic agents. It covers the conventional medicines like platinum compounds, vinca alkaloids, and methotrexate, as well as the newer therapeutic agents like immunomodulators and immune checkpoint inhibitors. A better understanding of the pathophysiology will lead to further advancement in strategies for management of chemotherapy-induced neurotoxicity.
Literature
1.
go back to reference de Moor JS, Mariotto AB, Parry C, Alfano CM, Padgett L, Kent EE, Forsythe L, Scoppa S, Hachey M, Rowland JH. Cancer survivors in the United States: prevalence across the survivorship trajectory and implications for care. Cancer Epidemiol Biomarkers Prev. 2013;22:561–70.PubMedPubMedCentralCrossRef de Moor JS, Mariotto AB, Parry C, Alfano CM, Padgett L, Kent EE, Forsythe L, Scoppa S, Hachey M, Rowland JH. Cancer survivors in the United States: prevalence across the survivorship trajectory and implications for care. Cancer Epidemiol Biomarkers Prev. 2013;22:561–70.PubMedPubMedCentralCrossRef
3.
go back to reference Magge RS, DeAngelis LM. The double-edged sword: neurotoxicity of chemotherapy. Blood Rev. 2015;29:93–100.PubMedCrossRef Magge RS, DeAngelis LM. The double-edged sword: neurotoxicity of chemotherapy. Blood Rev. 2015;29:93–100.PubMedCrossRef
4.
go back to reference Wigmore P. The effect of systemic chemotherapy on neurogenesis, plasticity and memory. Curr Top Behav Neurosci. 2013;15:211–40.PubMedCrossRef Wigmore P. The effect of systemic chemotherapy on neurogenesis, plasticity and memory. Curr Top Behav Neurosci. 2013;15:211–40.PubMedCrossRef
5.
go back to reference Sul JK, DeAngelis LM. Neurologic complications of cancer chemotherapy. Semin Oncol. 2006;33:324–32.PubMedCrossRef Sul JK, DeAngelis LM. Neurologic complications of cancer chemotherapy. Semin Oncol. 2006;33:324–32.PubMedCrossRef
9.
go back to reference Das A, Ranadive N, Kinra M, Nampoothiri M, Arora D, Mudgal J. An overview on chemotherapy-induced cognitive impairment and potential role of antidepressants. Curr Neuropharmacol. 2020;18:838–51.PubMedPubMedCentralCrossRef Das A, Ranadive N, Kinra M, Nampoothiri M, Arora D, Mudgal J. An overview on chemotherapy-induced cognitive impairment and potential role of antidepressants. Curr Neuropharmacol. 2020;18:838–51.PubMedPubMedCentralCrossRef
10.
go back to reference Kiguchi N, Maeda T, Kobayashi Y, Kishioka S. Up-regulation of tumor necrosis factor-alpha in spinal cord contributes to vincristine-induced mechanical allodynia in mice. Neurosci Lett. 2008;445:140–3.PubMedCrossRef Kiguchi N, Maeda T, Kobayashi Y, Kishioka S. Up-regulation of tumor necrosis factor-alpha in spinal cord contributes to vincristine-induced mechanical allodynia in mice. Neurosci Lett. 2008;445:140–3.PubMedCrossRef
11.
go back to reference Muthuraman A, Singh N, Jaggi AS. Protective effect of Acorus calamus L. in rat model of vincristine induced painful neuropathy: evidence of anti-inflammatory and anti-oxidative activity. Food Chem Toxicol. 2011;49:2557–63.PubMedCrossRef Muthuraman A, Singh N, Jaggi AS. Protective effect of Acorus calamus L. in rat model of vincristine induced painful neuropathy: evidence of anti-inflammatory and anti-oxidative activity. Food Chem Toxicol. 2011;49:2557–63.PubMedCrossRef
12.
go back to reference Briones TL, Woods J. Dysregulation in myelination mediated by persistent neuroinflammation: possible mechanisms in chemotherapy-related cognitive impairment. Brain Behav Immun. 2014;35:23–32.PubMedCrossRef Briones TL, Woods J. Dysregulation in myelination mediated by persistent neuroinflammation: possible mechanisms in chemotherapy-related cognitive impairment. Brain Behav Immun. 2014;35:23–32.PubMedCrossRef
13.
go back to reference Jangra A, Kwatra M, Singh T, Pant R, Kushwah P, Ahmed S, et al. Edaravone alleviates cisplatin-induced neurobehavioral deficits via modulation of oxidative stress and inflammatory mediators in the rat hippocampus. Eur J Pharmacol. 2016;791:51–61.PubMedCrossRef Jangra A, Kwatra M, Singh T, Pant R, Kushwah P, Ahmed S, et al. Edaravone alleviates cisplatin-induced neurobehavioral deficits via modulation of oxidative stress and inflammatory mediators in the rat hippocampus. Eur J Pharmacol. 2016;791:51–61.PubMedCrossRef
17.
go back to reference Weiss RB, Issell BF. The nitrosoureas: carmustine (BCNU) and lomustine (CCNU). Cancer Treat Rev. 1982;9:313–30.PubMedCrossRef Weiss RB, Issell BF. The nitrosoureas: carmustine (BCNU) and lomustine (CCNU). Cancer Treat Rev. 1982;9:313–30.PubMedCrossRef
18.
go back to reference More GS, Thomas AB, Chitlange SS, Nanda RK, Gajbhiye RL. Nitrogen mustards as alkylating agents: a review on chemistry, mechanism of action and current USFDA status of drugs. Anticancer Agents Med Chem. 2019;19:1080–102.PubMedCrossRef More GS, Thomas AB, Chitlange SS, Nanda RK, Gajbhiye RL. Nitrogen mustards as alkylating agents: a review on chemistry, mechanism of action and current USFDA status of drugs. Anticancer Agents Med Chem. 2019;19:1080–102.PubMedCrossRef
19.
go back to reference Singh RK, Kumar S, Prasad DN, Bhardwaj TR. Therapeutic journery of nitrogen mustard as alkylating anticancer agents: Historic to future perspectives. Eur J Med Chem. 2018;151:401–33.PubMedCrossRef Singh RK, Kumar S, Prasad DN, Bhardwaj TR. Therapeutic journery of nitrogen mustard as alkylating anticancer agents: Historic to future perspectives. Eur J Med Chem. 2018;151:401–33.PubMedCrossRef
20.
go back to reference Warwick GP. The mechanism of action of alkylating agents. Cancer Res. 1963;23:1315–33.PubMed Warwick GP. The mechanism of action of alkylating agents. Cancer Res. 1963;23:1315–33.PubMed
21.
go back to reference Meanwell CA, Blake AE, Kelly KA, Honigsberger L, Blackledge G. Prediction of ifosfamide/mesna associated encephalopathy. Eur J Cancer Clin Oncol. 1986;22:815–9.PubMedCrossRef Meanwell CA, Blake AE, Kelly KA, Honigsberger L, Blackledge G. Prediction of ifosfamide/mesna associated encephalopathy. Eur J Cancer Clin Oncol. 1986;22:815–9.PubMedCrossRef
22.
go back to reference Rieger C, Fiegl M, Tischer J, Ostermann H, Schiel X. Incidence and severity of ifosfamide-induced encephalopathy. Anticancer Drugs. 2004;15:347–50.PubMedCrossRef Rieger C, Fiegl M, Tischer J, Ostermann H, Schiel X. Incidence and severity of ifosfamide-induced encephalopathy. Anticancer Drugs. 2004;15:347–50.PubMedCrossRef
23.
go back to reference Pratt CB, Goren MP, Meyer WH, Singh B, Dodge RK. Ifosfamide neurotoxicity is related to previous cisplatin treatment for pediatric solid tumors. J Clin Oncol. 1990;8:1399–401.PubMedCrossRef Pratt CB, Goren MP, Meyer WH, Singh B, Dodge RK. Ifosfamide neurotoxicity is related to previous cisplatin treatment for pediatric solid tumors. J Clin Oncol. 1990;8:1399–401.PubMedCrossRef
24.
go back to reference David KA, Picus J. Evaluating risk factors for the development of ifosfamide encephalopathy. Am J Clin Oncol. 2005;28:277–80.PubMedCrossRef David KA, Picus J. Evaluating risk factors for the development of ifosfamide encephalopathy. Am J Clin Oncol. 2005;28:277–80.PubMedCrossRef
26.
go back to reference Savica R, Rabinstein AA, Josephs KA. Ifosfamide associated myoclonus-encephalopathy syndrome. J Neurol. 2011;258:1729–31.PubMedCrossRef Savica R, Rabinstein AA, Josephs KA. Ifosfamide associated myoclonus-encephalopathy syndrome. J Neurol. 2011;258:1729–31.PubMedCrossRef
27.
go back to reference Ames B, Lewis LD, Chaffee S, Kim J, Morse R. Ifosfamide-induced encephalopathy and movement disorder. Pediatr Blood Cancer. 2010;54:624–6.PubMedCrossRef Ames B, Lewis LD, Chaffee S, Kim J, Morse R. Ifosfamide-induced encephalopathy and movement disorder. Pediatr Blood Cancer. 2010;54:624–6.PubMedCrossRef
28.
go back to reference Taupin D, Racela R, Friedman D. Ifosfamide chemotherapy and nonconvulsive status epilepticus: case report and review of the literature. Clin EEG Neurosci. 2014;45:222–5.PubMedCrossRef Taupin D, Racela R, Friedman D. Ifosfamide chemotherapy and nonconvulsive status epilepticus: case report and review of the literature. Clin EEG Neurosci. 2014;45:222–5.PubMedCrossRef
29.
go back to reference Shapiro WR, Green SB. Reevaluating the efficacy of intra-arterial BCNU. J Neurosurg. 1987;66:313–5.PubMed Shapiro WR, Green SB. Reevaluating the efficacy of intra-arterial BCNU. J Neurosurg. 1987;66:313–5.PubMed
30.
go back to reference Shapiro WR, Green SB, Burger PC, Selker RG, VanGilder JC, Robertson JT, et al. A randomized comparison of intra-arterial versus intravenous BCNU, with or without intravenous 5-fluorouracil, for newly diagnosed patients with malignant glioma. J Neurosurg. 1992;76:772–81.PubMedCrossRef Shapiro WR, Green SB, Burger PC, Selker RG, VanGilder JC, Robertson JT, et al. A randomized comparison of intra-arterial versus intravenous BCNU, with or without intravenous 5-fluorouracil, for newly diagnosed patients with malignant glioma. J Neurosurg. 1992;76:772–81.PubMedCrossRef
31.
go back to reference Hiesiger EM, Green SB, Shapiro WR, Burger PC, Selker RG, Mahaley MS, et al. Results of a randomized trial comparing intra-arterial cisplatin and intravenous PCNU for the treatment of primary brain tumors in adults: Brain Tumor Cooperative Group trial 8420A. J Neurooncol. 1995;25:143–54.PubMedCrossRef Hiesiger EM, Green SB, Shapiro WR, Burger PC, Selker RG, Mahaley MS, et al. Results of a randomized trial comparing intra-arterial cisplatin and intravenous PCNU for the treatment of primary brain tumors in adults: Brain Tumor Cooperative Group trial 8420A. J Neurooncol. 1995;25:143–54.PubMedCrossRef
32.
go back to reference Burger PC, Kamenar E, Schold SC, Fay JW, Phillips GL, Herzig GP. Encephalomyelopathy following high-dose BCNU therapy. Cancer. 1981;48:1318–27.CrossRef Burger PC, Kamenar E, Schold SC, Fay JW, Phillips GL, Herzig GP. Encephalomyelopathy following high-dose BCNU therapy. Cancer. 1981;48:1318–27.CrossRef
34.
go back to reference Stone JB, DeAngelis LM. Cancer-treatment-induced neurotoxicity–focus on newer treatments. Nat Rev Clin Oncol. 2016;13:92–105.PubMedCrossRef Stone JB, DeAngelis LM. Cancer-treatment-induced neurotoxicity–focus on newer treatments. Nat Rev Clin Oncol. 2016;13:92–105.PubMedCrossRef
35.
go back to reference Eberly AL, Anderson GD, Bubalo JS, McCune JS. Optimal prevention of seizures induced by high-dose busulfan. Pharmacotherapy. 2008;28:1502–10.PubMedCrossRef Eberly AL, Anderson GD, Bubalo JS, McCune JS. Optimal prevention of seizures induced by high-dose busulfan. Pharmacotherapy. 2008;28:1502–10.PubMedCrossRef
36.
go back to reference DeAngelis LM, Posner JB. Neurologic complications of cancer. 2nd ed. New York, USA: Contemporary Neurology Series. Oxford University Press Inc; 2009. p. 447–510. DeAngelis LM, Posner JB. Neurologic complications of cancer. 2nd ed. New York, USA: Contemporary Neurology Series. Oxford University Press Inc; 2009. p. 447–510.
37.
go back to reference Makranz C, Khutsurauli S, Kalish Y, Eliahou R, Kadouri L, Gomori J, Lossos A. Neurological variability in chemotherapy-induced posterior reversible encephalopathy syndrome associated with thrombotic microangiopathy: Case reports and literature review. Mol Clin Oncol. 2018;8:178–82.PubMed Makranz C, Khutsurauli S, Kalish Y, Eliahou R, Kadouri L, Gomori J, Lossos A. Neurological variability in chemotherapy-induced posterior reversible encephalopathy syndrome associated with thrombotic microangiopathy: Case reports and literature review. Mol Clin Oncol. 2018;8:178–82.PubMed
38.
go back to reference Bae SH, Park MJ, Lee MM, Kim TM, Lee SH, Cho SY, et al. Toxicity profile of temozolomide in the treatment of 300 malignant glioma patients in Korea. J Korean Med Sci. 2014;29:980–4.PubMedPubMedCentralCrossRef Bae SH, Park MJ, Lee MM, Kim TM, Lee SH, Cho SY, et al. Toxicity profile of temozolomide in the treatment of 300 malignant glioma patients in Korea. J Korean Med Sci. 2014;29:980–4.PubMedPubMedCentralCrossRef
39.
go back to reference Boland MP, Foster SJ, O’Neill LAJ. Daunorubicin activates NFkappaB and induces kappaB-dependent gene expression in HL-60 promyelocytic and Jurkat T lymphoma cells. J Biol Chem. 1997;272:12952–60.PubMedCrossRef Boland MP, Foster SJ, O’Neill LAJ. Daunorubicin activates NFkappaB and induces kappaB-dependent gene expression in HL-60 promyelocytic and Jurkat T lymphoma cells. J Biol Chem. 1997;272:12952–60.PubMedCrossRef
40.
go back to reference Kasibhatla S, Brunner T, Genestier L, Echeverri F, Mahboubi A, Green DR. DNA damaging agents induce expression of Fas ligand and subsequent apoptosis in T lymphocytes via the activation of NF-kappa B and AP-1. Mol Cell. 1998;1:543–51.PubMedCrossRef Kasibhatla S, Brunner T, Genestier L, Echeverri F, Mahboubi A, Green DR. DNA damaging agents induce expression of Fas ligand and subsequent apoptosis in T lymphocytes via the activation of NF-kappa B and AP-1. Mol Cell. 1998;1:543–51.PubMedCrossRef
41.
go back to reference Bohuslav J, Chen LF, Kwon H, Mu Y, Greene WC. p53 induces NF-kappaB activation by an IkappaB kinase-independent mechanism involving phosphorylation of p65 by ribosomal S6 kinase 1. J Biol Chem. 2004;279:26115–25.PubMedCrossRef Bohuslav J, Chen LF, Kwon H, Mu Y, Greene WC. p53 induces NF-kappaB activation by an IkappaB kinase-independent mechanism involving phosphorylation of p65 by ribosomal S6 kinase 1. J Biol Chem. 2004;279:26115–25.PubMedCrossRef
42.
go back to reference Minsavage GD, Dillman JF. Bifunctional alkylating agent-induced p53 and nonclassical nuclear factor kappaB responses and cell death are altered by caffeic acid phenethyl ester: a potential role for antioxidant/electrophilic response-element signaling. J Pharmacol Exp Ther. 2007;321:202–12.PubMedCrossRef Minsavage GD, Dillman JF. Bifunctional alkylating agent-induced p53 and nonclassical nuclear factor kappaB responses and cell death are altered by caffeic acid phenethyl ester: a potential role for antioxidant/electrophilic response-element signaling. J Pharmacol Exp Ther. 2007;321:202–12.PubMedCrossRef
43.
go back to reference Zhang Y, Broser M, Rom WN. Activation of the interleukin 6 gene by Mycobacterium tuberculosis or lipopolysaccharide is mediated by nuclear factors NF-IL6 and NF-kappa B. Proc Natl Acad Sci U S A. 1994;91:2225–9.PubMedPubMedCentralCrossRef Zhang Y, Broser M, Rom WN. Activation of the interleukin 6 gene by Mycobacterium tuberculosis or lipopolysaccharide is mediated by nuclear factors NF-IL6 and NF-kappa B. Proc Natl Acad Sci U S A. 1994;91:2225–9.PubMedPubMedCentralCrossRef
44.
go back to reference Ritchie MH, Fillmore RA, Lausch RN, Oakes JE. A role for NF-kappa B binding motifs in the differential induction of chemokine gene expression in human corneal epithelial cells. Invest Ophthalmol Vis Sci. 2004;45:2299–305.PubMedCrossRef Ritchie MH, Fillmore RA, Lausch RN, Oakes JE. A role for NF-kappa B binding motifs in the differential induction of chemokine gene expression in human corneal epithelial cells. Invest Ophthalmol Vis Sci. 2004;45:2299–305.PubMedCrossRef
45.
go back to reference Dillman JF, McGary KL, Schlager JJ. An inhibitor of p38 MAP kinase downregulates cytokine release induced by sulfur mustard exposure in human epidermal keratinocytes. Toxicol In Vitro. 2004;18:593–9.PubMedCrossRef Dillman JF, McGary KL, Schlager JJ. An inhibitor of p38 MAP kinase downregulates cytokine release induced by sulfur mustard exposure in human epidermal keratinocytes. Toxicol In Vitro. 2004;18:593–9.PubMedCrossRef
46.
go back to reference Ryan KM, O’Prey J, Vousden KH. Loss of nuclear factor-kappaB is tumor promoting but does not substitute for loss of p53. Cancer Res. 2004;64:4415–8.PubMedCrossRef Ryan KM, O’Prey J, Vousden KH. Loss of nuclear factor-kappaB is tumor promoting but does not substitute for loss of p53. Cancer Res. 2004;64:4415–8.PubMedCrossRef
47.
go back to reference Rosenthal DS, Simbulan-Rosenthal CMG, Iyer S, Spoonde A, Smith W, Ray R, et al. Sulfur mustard induces markers of terminal differentiation and apoptosis in keratinocytes via a Ca2+-calmodulin and caspase-dependent pathway. J Invest Dermatol. 1998;111:64–71.PubMedCrossRef Rosenthal DS, Simbulan-Rosenthal CMG, Iyer S, Spoonde A, Smith W, Ray R, et al. Sulfur mustard induces markers of terminal differentiation and apoptosis in keratinocytes via a Ca2+-calmodulin and caspase-dependent pathway. J Invest Dermatol. 1998;111:64–71.PubMedCrossRef
49.
go back to reference Martindale JL, Holbrook NJ. Cellular response to oxidative stress: signaling for suicide and survival. J Cell Physiol. 2002;192:1–15.PubMedCrossRef Martindale JL, Holbrook NJ. Cellular response to oxidative stress: signaling for suicide and survival. J Cell Physiol. 2002;192:1–15.PubMedCrossRef
50.
go back to reference Fujioka S, Schmidt C, Sclabas GM, Li Z, Pelicano H, Peng B, et al. Stabilization of p53 is a novel mechanism for proapoptotic function of NF-kappaB. J Biol Chem. 2004;279:27549–59.PubMedCrossRef Fujioka S, Schmidt C, Sclabas GM, Li Z, Pelicano H, Peng B, et al. Stabilization of p53 is a novel mechanism for proapoptotic function of NF-kappaB. J Biol Chem. 2004;279:27549–59.PubMedCrossRef
51.
go back to reference Kohn KW, Pommier Y. Molecular interaction map of the p53 and Mdm2 logic elements, which control the Off-On switch of p53 in response to DNA damage. Biochem Biophys Res Commun. 2005;331:816–27.PubMedCrossRef Kohn KW, Pommier Y. Molecular interaction map of the p53 and Mdm2 logic elements, which control the Off-On switch of p53 in response to DNA damage. Biochem Biophys Res Commun. 2005;331:816–27.PubMedCrossRef
52.
go back to reference Shieh SY, Ikeda M, Taya Y, Prives C. DNA damage-induced phosphorylation of p53 alleviates inhibition by MDM2. Cell. 1997;91:325–34.PubMedCrossRef Shieh SY, Ikeda M, Taya Y, Prives C. DNA damage-induced phosphorylation of p53 alleviates inhibition by MDM2. Cell. 1997;91:325–34.PubMedCrossRef
54.
go back to reference Podratz JL, Knight AM, Ta LE, Staff NP, Gass JM, Genelin K, et al. Cisplatin induced mitochondrial DNA damage in dorsal root ganglion neurons. Neurobiol Dis. 2011;41:661–8.PubMedCrossRef Podratz JL, Knight AM, Ta LE, Staff NP, Gass JM, Genelin K, et al. Cisplatin induced mitochondrial DNA damage in dorsal root ganglion neurons. Neurobiol Dis. 2011;41:661–8.PubMedCrossRef
56.
go back to reference Hinduja S, Kraus KS, Manohar S, Salvi RJ. D-methionine protects against cisplatin-induced neurotoxicity in the hippocampus of the adult rat. Neurotox Res. 2015;27:199–204.PubMedCrossRef Hinduja S, Kraus KS, Manohar S, Salvi RJ. D-methionine protects against cisplatin-induced neurotoxicity in the hippocampus of the adult rat. Neurotox Res. 2015;27:199–204.PubMedCrossRef
57.
go back to reference Kaasa S, Olsnes BT, Mastekaasa A. Neuropsychological evaluation of patients with inoperable non-small cell lung cancer treated with combination chemotherapy or radiotherapy. Acta Oncol. 1988;27:241–6.PubMedCrossRef Kaasa S, Olsnes BT, Mastekaasa A. Neuropsychological evaluation of patients with inoperable non-small cell lung cancer treated with combination chemotherapy or radiotherapy. Acta Oncol. 1988;27:241–6.PubMedCrossRef
58.
go back to reference Troy L, McFarland K, Littman-Power S, Kelly BJ, Walpole ET, Wyld D, et al. Cisplatin-based therapy: a neurological and neuropsychological review. Psychooncology. 2000;9:29–39.PubMedCrossRef Troy L, McFarland K, Littman-Power S, Kelly BJ, Walpole ET, Wyld D, et al. Cisplatin-based therapy: a neurological and neuropsychological review. Psychooncology. 2000;9:29–39.PubMedCrossRef
59.
go back to reference Roelofs RI, Hrushesky W, Rogin J, Rosenberg L. Peripheral sensory neuropathy and cisplatin chemotherapy. Neurology. 1984;34:934–8.PubMedCrossRef Roelofs RI, Hrushesky W, Rogin J, Rosenberg L. Peripheral sensory neuropathy and cisplatin chemotherapy. Neurology. 1984;34:934–8.PubMedCrossRef
60.
go back to reference Thompson SW, Davis LE, Kornfeld M, Hilgers RD, Standefer JC. Cisplatin neuropathy. Clinical, electrophysiologic, morphologic, and toxicologic studies. Cancer. 1984;54:1269–75.PubMedCrossRef Thompson SW, Davis LE, Kornfeld M, Hilgers RD, Standefer JC. Cisplatin neuropathy. Clinical, electrophysiologic, morphologic, and toxicologic studies. Cancer. 1984;54:1269–75.PubMedCrossRef
61.
go back to reference Berman IJ, Mann MP. Seizures and transient cortical blindness associated with cis-platinum (II) diamminedichloride (PDD) therapy in a thirty-year-old man. Cancer. 1980;45:764–6.PubMedCrossRef Berman IJ, Mann MP. Seizures and transient cortical blindness associated with cis-platinum (II) diamminedichloride (PDD) therapy in a thirty-year-old man. Cancer. 1980;45:764–6.PubMedCrossRef
62.
go back to reference Lyass O, Lossos A, Hubert A, Gips M, Peretz T. Cisplatin-induced non-convulsive encephalopathy. Anticancer Drugs. 1998;9:100–4.PubMedCrossRef Lyass O, Lossos A, Hubert A, Gips M, Peretz T. Cisplatin-induced non-convulsive encephalopathy. Anticancer Drugs. 1998;9:100–4.PubMedCrossRef
63.
go back to reference Içli F, Karaoğuz H, Dinçol D, Demirkazik A, Günel N, Karaoğuz R, et al. Severe vascular toxicity associated with cisplatin-based chemotherapy. Cancer. 1993;72:587–93.PubMedCrossRef Içli F, Karaoğuz H, Dinçol D, Demirkazik A, Günel N, Karaoğuz R, et al. Severe vascular toxicity associated with cisplatin-based chemotherapy. Cancer. 1993;72:587–93.PubMedCrossRef
64.
go back to reference Dietrich J, Marienhagen J, Schalke B, Bogdahn U, Schlachetzki F. Vascular neurotoxicity following chemotherapy with cisplatin, ifosfamide, and etoposide. Ann Pharmacother. 2004;38:242–6.PubMedCrossRef Dietrich J, Marienhagen J, Schalke B, Bogdahn U, Schlachetzki F. Vascular neurotoxicity following chemotherapy with cisplatin, ifosfamide, and etoposide. Ann Pharmacother. 2004;38:242–6.PubMedCrossRef
65.
go back to reference Pasetto LM, D’Andrea MR, Rossi E, Monfardini S. Oxaliplatin-related neurotoxicity: how and why? Crit Rev Oncol Hematol. 2006;59:159–68.PubMedCrossRef Pasetto LM, D’Andrea MR, Rossi E, Monfardini S. Oxaliplatin-related neurotoxicity: how and why? Crit Rev Oncol Hematol. 2006;59:159–68.PubMedCrossRef
66.
go back to reference Heinzlef O, Lotz JP, Roullet E. Severe neuropathy after high dose carboplatin in three patients receiving multidrug chemotherapy. J Neurol Neurosurg Psychiatry. 1998;64:667–9.PubMedPubMedCentralCrossRef Heinzlef O, Lotz JP, Roullet E. Severe neuropathy after high dose carboplatin in three patients receiving multidrug chemotherapy. J Neurol Neurosurg Psychiatry. 1998;64:667–9.PubMedPubMedCentralCrossRef
67.
go back to reference So H, Kim HJ, Lee JH, Park C, Kim Y, Kim E, et al. Cisplatin cytotoxicity of auditory cells requires secretions of proinflammatory cytokines via activation of ERK and NF-kappaB. J Assoc Res Otolaryngol. 2007;8:338–55.PubMedPubMedCentralCrossRef So H, Kim HJ, Lee JH, Park C, Kim Y, Kim E, et al. Cisplatin cytotoxicity of auditory cells requires secretions of proinflammatory cytokines via activation of ERK and NF-kappaB. J Assoc Res Otolaryngol. 2007;8:338–55.PubMedPubMedCentralCrossRef
68.
go back to reference Kim SJ, Lim JY, Lee JN, Choe SK, Kim Y-i, Song SR, et al. Activation of β-catenin by inhibitors of glycogen synthase kinase-3 ameliorates cisplatin-induced cytotoxicity and pro-inflammatory cytokine expression in HEI-OC1 cells. Toxicology. 2014;320:74–82.PubMedCrossRef Kim SJ, Lim JY, Lee JN, Choe SK, Kim Y-i, Song SR, et al. Activation of β-catenin by inhibitors of glycogen synthase kinase-3 ameliorates cisplatin-induced cytotoxicity and pro-inflammatory cytokine expression in HEI-OC1 cells. Toxicology. 2014;320:74–82.PubMedCrossRef
69.
go back to reference Shabani M, Larizadeh MH, Parsania S, Hajali V, Shojaei A. Evaluation of destructive effects of exposure to cisplatin during developmental stage: no profound evidence for sex differences in impaired motor and memory performance. Int J Neurosci. 2012;122:439–48.PubMedCrossRef Shabani M, Larizadeh MH, Parsania S, Hajali V, Shojaei A. Evaluation of destructive effects of exposure to cisplatin during developmental stage: no profound evidence for sex differences in impaired motor and memory performance. Int J Neurosci. 2012;122:439–48.PubMedCrossRef
71.
go back to reference Sahin K, Tuzcu M, Sahin N, Ali S, Kucuk O. Nrf2/HO-1 signaling pathway may be the prime target for chemoprevention of cisplatin-induced nephrotoxicity by lycopene. Food Chem Toxicol. 2010;48:2670–4.PubMedCrossRef Sahin K, Tuzcu M, Sahin N, Ali S, Kucuk O. Nrf2/HO-1 signaling pathway may be the prime target for chemoprevention of cisplatin-induced nephrotoxicity by lycopene. Food Chem Toxicol. 2010;48:2670–4.PubMedCrossRef
74.
go back to reference da Silva Machado C, Mendonça LM, Venancio V de P, Bianchi MLP, Antunes LMG. Coenzyme Q10 protects Pc12 cells from cisplatin-induced DNA damage and neurotoxicity. Neurotoxicology. 2013;36:10–6. da Silva Machado C, Mendonça LM, Venancio V de P, Bianchi MLP, Antunes LMG. Coenzyme Q10 protects Pc12 cells from cisplatin-induced DNA damage and neurotoxicity. Neurotoxicology. 2013;36:10–6.
75.
go back to reference Shabani M, Nazeri M, Parsania S, Razavinasab M, Zangiabadi N, Esmaeilpour K, et al. Walnut consumption protects rats against cisplatin-induced neurotoxicity. Neurotoxicology. 2012;33:1314–21.PubMedCrossRef Shabani M, Nazeri M, Parsania S, Razavinasab M, Zangiabadi N, Esmaeilpour K, et al. Walnut consumption protects rats against cisplatin-induced neurotoxicity. Neurotoxicology. 2012;33:1314–21.PubMedCrossRef
76.
go back to reference Gopal KV, Wu C, Shrestha B, Campbell KCM, Moore EJ, Gross GW. d-Methionine protects against cisplatin-induced neurotoxicity in cortical networks. Neurotoxicol Teratol. 2012;34:495–504.PubMedCrossRef Gopal KV, Wu C, Shrestha B, Campbell KCM, Moore EJ, Gross GW. d-Methionine protects against cisplatin-induced neurotoxicity in cortical networks. Neurotoxicol Teratol. 2012;34:495–504.PubMedCrossRef
77.
go back to reference Tuncer S, Dalkilic N, Akif Dunbar M, Keles B. Comparative effects of α lipoic acid and melatonin on cisplatin-induced neurotoxicity. Int J Neurosci. 2010;120:655–63.PubMedCrossRef Tuncer S, Dalkilic N, Akif Dunbar M, Keles B. Comparative effects of α lipoic acid and melatonin on cisplatin-induced neurotoxicity. Int J Neurosci. 2010;120:655–63.PubMedCrossRef
78.
go back to reference Oz M, Nurullahoglu Atalik KE, Yerlikaya FH, Demir EA. Curcumin alleviates cisplatin-induced learning and memory impairments. Neurobiol Learn Mem. 2015;123:43–9.PubMedCrossRef Oz M, Nurullahoglu Atalik KE, Yerlikaya FH, Demir EA. Curcumin alleviates cisplatin-induced learning and memory impairments. Neurobiol Learn Mem. 2015;123:43–9.PubMedCrossRef
79.
go back to reference Weijl NI, Hopman GD, Wipkink-Bakker A, Lentjes EG, Berger HM, Cleton FJ, Osanto S. Cisplatin combination chemotherapy induces a fall in plasma antioxidants of cancer patients. Ann Oncol. 1998;9:1331–7.PubMedCrossRef Weijl NI, Hopman GD, Wipkink-Bakker A, Lentjes EG, Berger HM, Cleton FJ, Osanto S. Cisplatin combination chemotherapy induces a fall in plasma antioxidants of cancer patients. Ann Oncol. 1998;9:1331–7.PubMedCrossRef
80.
go back to reference Pace A, Savarese A, Picardo M, Maresca V, Pacetti U, del Monte G, et al. Neuroprotective effect of vitamin E supplementation in patients treated with cisplatin chemotherapy. J Clin Oncol. 2003;21:927–31.PubMedCrossRef Pace A, Savarese A, Picardo M, Maresca V, Pacetti U, del Monte G, et al. Neuroprotective effect of vitamin E supplementation in patients treated with cisplatin chemotherapy. J Clin Oncol. 2003;21:927–31.PubMedCrossRef
82.
go back to reference King KL, Boder GB. Correlation of the clinical neurotoxicity of the vinca alkaloids vincristine, vinblastine, and vindesine with their effects on cultured rat midbrain cells. Cancer Chemother Pharmacol. 1979;2:239–42.PubMedCrossRef King KL, Boder GB. Correlation of the clinical neurotoxicity of the vinca alkaloids vincristine, vinblastine, and vindesine with their effects on cultured rat midbrain cells. Cancer Chemother Pharmacol. 1979;2:239–42.PubMedCrossRef
83.
go back to reference Casey EB, Jellife AM, le Quesne PM, Millett YL. Vincristine neuropathy. Clin Electrophysiol Obs Brain. 1973;96:69–86. Casey EB, Jellife AM, le Quesne PM, Millett YL. Vincristine neuropathy. Clin Electrophysiol Obs Brain. 1973;96:69–86.
84.
85.
go back to reference LaPointe NE, Morfini G, Brady ST, Feinstein SC, Wilson L, Jordan MA. Effects of eribulin, vincristine, paclitaxel and ixabepilone on fast axonal transport and kinesin-1 driven microtubule gliding: implications for chemotherapy-induced peripheral neuropathy. Neurotoxicology. 2013;37:231–9.PubMedPubMedCentralCrossRef LaPointe NE, Morfini G, Brady ST, Feinstein SC, Wilson L, Jordan MA. Effects of eribulin, vincristine, paclitaxel and ixabepilone on fast axonal transport and kinesin-1 driven microtubule gliding: implications for chemotherapy-induced peripheral neuropathy. Neurotoxicology. 2013;37:231–9.PubMedPubMedCentralCrossRef
86.
go back to reference Pal P. Clinical and electrophysiological studies in vincristine induced neuropathy. Electromyogr Clin Neurophysiol. 1999;39:323–30.PubMed Pal P. Clinical and electrophysiological studies in vincristine induced neuropathy. Electromyogr Clin Neurophysiol. 1999;39:323–30.PubMed
87.
go back to reference Robertson G, Bhoopalam N, Zelkowitz L. Vincristine neurotoxicity and abnormal secretion of antidiuretic hormone. Arch Intern Med. 1973;132:717–20.PubMedCrossRef Robertson G, Bhoopalam N, Zelkowitz L. Vincristine neurotoxicity and abnormal secretion of antidiuretic hormone. Arch Intern Med. 1973;132:717–20.PubMedCrossRef
88.
go back to reference Byrd R, Rohrbaugh T, Raney R Jr, Norris D. Transient cortical blindness secondary to vincristine therapy in childhood malignancies. Cancer. 1981;47:37–40.PubMedCrossRef Byrd R, Rohrbaugh T, Raney R Jr, Norris D. Transient cortical blindness secondary to vincristine therapy in childhood malignancies. Cancer. 1981;47:37–40.PubMedCrossRef
89.
go back to reference Gomber S, Dewan P, Chhonker D. Vincristine induced neurotoxicity in cancer patients. Indian J Pediatr. 2010;77:97–100.PubMedCrossRef Gomber S, Dewan P, Chhonker D. Vincristine induced neurotoxicity in cancer patients. Indian J Pediatr. 2010;77:97–100.PubMedCrossRef
90.
go back to reference Kiguchi N, Maeda T, Kobayashi Y, Saika F, Kishioka S. Involvement of inflammatory mediators in neuropathic pain caused by vincristine. Int Rev Neurobiol. 2009;85:179–90.PubMedCrossRef Kiguchi N, Maeda T, Kobayashi Y, Saika F, Kishioka S. Involvement of inflammatory mediators in neuropathic pain caused by vincristine. Int Rev Neurobiol. 2009;85:179–90.PubMedCrossRef
91.
go back to reference Sweitzer SM, Pahl JL, DeLeo JA. Propentofylline attenuates vincristine-induced peripheral neuropathy in the rat. Neurosci Lett. 2006;400:258–61.PubMedCrossRef Sweitzer SM, Pahl JL, DeLeo JA. Propentofylline attenuates vincristine-induced peripheral neuropathy in the rat. Neurosci Lett. 2006;400:258–61.PubMedCrossRef
92.
go back to reference Bujalska M, Tatarkiewicz J, de Cordé A, Gumułka SW. Effect of cyclooxygenase and nitric oxide synthase inhibitors on streptozotocin-induced hyperalgesia in rats. Pharmacology. 2008;81:151–7.PubMedCrossRef Bujalska M, Tatarkiewicz J, de Cordé A, Gumułka SW. Effect of cyclooxygenase and nitric oxide synthase inhibitors on streptozotocin-induced hyperalgesia in rats. Pharmacology. 2008;81:151–7.PubMedCrossRef
93.
go back to reference Kamei J, Tamura N, Saitoh A. Possible involvement of the spinal nitric oxide/cGMP pathway in vincristine-induced painful neuropathy in mice. Pain. 2005;117:112–20.PubMedCrossRef Kamei J, Tamura N, Saitoh A. Possible involvement of the spinal nitric oxide/cGMP pathway in vincristine-induced painful neuropathy in mice. Pain. 2005;117:112–20.PubMedCrossRef
94.
go back to reference Wei Y, Pu X, Zhao L. Preclinical studies for the combination of paclitaxel and curcumin in cancer therapy (Review). Oncol Rep. 2017;37:3159–66.PubMedCrossRef Wei Y, Pu X, Zhao L. Preclinical studies for the combination of paclitaxel and curcumin in cancer therapy (Review). Oncol Rep. 2017;37:3159–66.PubMedCrossRef
95.
go back to reference Nogales E, Grayer Wolf S, Khan IA, Ludueña RF, Downing KH. Structure of tubulin at 6.5 A and location of the taxol-binding site. Nature. 1995;375:424–7.PubMedCrossRef Nogales E, Grayer Wolf S, Khan IA, Ludueña RF, Downing KH. Structure of tubulin at 6.5 A and location of the taxol-binding site. Nature. 1995;375:424–7.PubMedCrossRef
96.
go back to reference Lee JJ, Swain SM. Peripheral neuropathy induced by microtubule-stabilizing agents. J Clin Oncol. 2006;24:1633–42.PubMedCrossRef Lee JJ, Swain SM. Peripheral neuropathy induced by microtubule-stabilizing agents. J Clin Oncol. 2006;24:1633–42.PubMedCrossRef
98.
go back to reference Perry JR, Warner E. Transient encephalopathy after paclitaxel (Taxol) infusion. Neurology. 1996;46:1596–9.PubMedCrossRef Perry JR, Warner E. Transient encephalopathy after paclitaxel (Taxol) infusion. Neurology. 1996;46:1596–9.PubMedCrossRef
99.
go back to reference Rook J, Rosser T, Fangusaro J, Finlay J. Acute transient encephalopathy following paclitaxel treatment in an adolescent with a recurrent suprasellar germinoma. Pediatr Blood Cancer. 2008;50:699–700.PubMedCrossRef Rook J, Rosser T, Fangusaro J, Finlay J. Acute transient encephalopathy following paclitaxel treatment in an adolescent with a recurrent suprasellar germinoma. Pediatr Blood Cancer. 2008;50:699–700.PubMedCrossRef
100.
go back to reference Pusztai L, Mendoza TR, Reuben JM, Martinez MM, Willey JS, Lara J, et al. Changes in plasma levels of inflammatory cytokines in response to paclitaxel chemotherapy. Cytokine. 2004;25:94–102.PubMedCrossRef Pusztai L, Mendoza TR, Reuben JM, Martinez MM, Willey JS, Lara J, et al. Changes in plasma levels of inflammatory cytokines in response to paclitaxel chemotherapy. Cytokine. 2004;25:94–102.PubMedCrossRef
102.
go back to reference Peters CM, Jimenez-Andrade JM, Jonas BM, Sevcik MA, Koewler NJ, Ghilardi JR, et al. Intravenous paclitaxel administration in the rat induces a peripheral sensory neuropathy characterized by macrophage infiltration and injury to sensory neurons and their supporting cells. Exp Neurol. 2007;203:42–54.PubMedCrossRef Peters CM, Jimenez-Andrade JM, Jonas BM, Sevcik MA, Koewler NJ, Ghilardi JR, et al. Intravenous paclitaxel administration in the rat induces a peripheral sensory neuropathy characterized by macrophage infiltration and injury to sensory neurons and their supporting cells. Exp Neurol. 2007;203:42–54.PubMedCrossRef
103.
go back to reference Argyriou AA, Koltzenburg M, Polychronopoulos P, Papapetropoulos S, Kalofonos HP. Peripheral nerve damage associated with administration of taxanes in patients with cancer. Crit Rev Oncol Hematol. 2008;66:218–28.PubMedCrossRef Argyriou AA, Koltzenburg M, Polychronopoulos P, Papapetropoulos S, Kalofonos HP. Peripheral nerve damage associated with administration of taxanes in patients with cancer. Crit Rev Oncol Hematol. 2008;66:218–28.PubMedCrossRef
106.
go back to reference Duggett NA, Griffiths LA, McKenna OE, de Santis V, Yongsanguanchai N, Mokori EB, et al. Oxidative stress in the development, maintenance and resolution of paclitaxel-induced painful neuropathy. Neuroscience. 2016;333:13–26.PubMedCrossRef Duggett NA, Griffiths LA, McKenna OE, de Santis V, Yongsanguanchai N, Mokori EB, et al. Oxidative stress in the development, maintenance and resolution of paclitaxel-induced painful neuropathy. Neuroscience. 2016;333:13–26.PubMedCrossRef
109.
110.
go back to reference Bhojwani D, Sabin ND, Pei D, Yang JJ, Khan RB, Panetta JC, et al. Methotrexate-induced neurotoxicity and leukoencephalopathy in childhood acute lymphoblastic leukemia. J Clin Oncol. 2014;32:949–59.PubMedPubMedCentralCrossRef Bhojwani D, Sabin ND, Pei D, Yang JJ, Khan RB, Panetta JC, et al. Methotrexate-induced neurotoxicity and leukoencephalopathy in childhood acute lymphoblastic leukemia. J Clin Oncol. 2014;32:949–59.PubMedPubMedCentralCrossRef
112.
go back to reference Gibson EM, Nagaraja S, Ocampo A, Tam LT, Wood LS, Pallegar PN, et al. Methotrexate chemotherapy induces persistent tri-glial dysregulation that underlies chemotherapy-related cognitive impairment. Cell. 2019;176:43-55.e13.PubMedCrossRef Gibson EM, Nagaraja S, Ocampo A, Tam LT, Wood LS, Pallegar PN, et al. Methotrexate chemotherapy induces persistent tri-glial dysregulation that underlies chemotherapy-related cognitive impairment. Cell. 2019;176:43-55.e13.PubMedCrossRef
113.
go back to reference Seigers R, Timmermans J, van der Horn HJ, de Vries EFJ, Dierckx RA, Visser L, et al. Methotrexate reduces hippocampal blood vessel density and activates microglia in rats but does not elevate central cytokine release. Behav Brain Res. 2010;207:265–72.PubMedCrossRef Seigers R, Timmermans J, van der Horn HJ, de Vries EFJ, Dierckx RA, Visser L, et al. Methotrexate reduces hippocampal blood vessel density and activates microglia in rats but does not elevate central cytokine release. Behav Brain Res. 2010;207:265–72.PubMedCrossRef
115.
116.
go back to reference Riehl JL, Brown WJ. Acute cerebellar syndrome secondary to 5-fluorouracil therapy. Neurology. 1964;14:961–7.PubMedCrossRef Riehl JL, Brown WJ. Acute cerebellar syndrome secondary to 5-fluorouracil therapy. Neurology. 1964;14:961–7.PubMedCrossRef
117.
go back to reference Gottlieb JA, Luce JK. Cerebellar ataxia with weekly 5-fluorouracil administration. Lancet. 1971;1:138–9.PubMedCrossRef Gottlieb JA, Luce JK. Cerebellar ataxia with weekly 5-fluorouracil administration. Lancet. 1971;1:138–9.PubMedCrossRef
118.
go back to reference Liaw CC, Liaw SJ, Wang CH, Chiu MC, Huang JS. Transient hyperammonemia related to chemotherapy with continuous infusion of high-dose 5-fluorouracil. Anticancer Drugs. 1993;4:311–5.PubMedCrossRef Liaw CC, Liaw SJ, Wang CH, Chiu MC, Huang JS. Transient hyperammonemia related to chemotherapy with continuous infusion of high-dose 5-fluorouracil. Anticancer Drugs. 1993;4:311–5.PubMedCrossRef
119.
go back to reference Liaw CC, Wang HM, Wang CH, Yang TS, Chen JS, Chang HK, et al. Risk of transient hyperammonemic encephalopathy in cancer patients who received continuous infusion of 5-fluorouracil with the complication of dehydration and infection. Anticancer Drugs. 1999;10:275–81.PubMedCrossRef Liaw CC, Wang HM, Wang CH, Yang TS, Chen JS, Chang HK, et al. Risk of transient hyperammonemic encephalopathy in cancer patients who received continuous infusion of 5-fluorouracil with the complication of dehydration and infection. Anticancer Drugs. 1999;10:275–81.PubMedCrossRef
120.
go back to reference Nott L, Price TJ, Pittman K, Patterson K, Fletcher J. Hyperammonemia encephalopathy: an important cause of neurological deterioration following chemotherapy. Leuk Lymphoma. 2007;48:1702–11.PubMedCrossRef Nott L, Price TJ, Pittman K, Patterson K, Fletcher J. Hyperammonemia encephalopathy: an important cause of neurological deterioration following chemotherapy. Leuk Lymphoma. 2007;48:1702–11.PubMedCrossRef
121.
go back to reference Kikuta S, Asakage T, Nakao K, Sugasawa M, Kubota A. The aggravating factors of hyperammonemia related to 5-fluorouracil infusion–a report of two cases. Auris Nasus Larynx. 2008;35:295–9.PubMedCrossRef Kikuta S, Asakage T, Nakao K, Sugasawa M, Kubota A. The aggravating factors of hyperammonemia related to 5-fluorouracil infusion–a report of two cases. Auris Nasus Larynx. 2008;35:295–9.PubMedCrossRef
122.
go back to reference Brashear A, Siemers E. Focal dystonia after chemotherapy: a case series. J Neurooncol. 1997;34:163–7.PubMedCrossRef Brashear A, Siemers E. Focal dystonia after chemotherapy: a case series. J Neurooncol. 1997;34:163–7.PubMedCrossRef
123.
go back to reference Bergevin PR, Patwardhan VC, Weissman J, Sui Moi L. Letter: Neurotoxicity of 5-fluorouracil. Lancet. 1975;1:410. Bergevin PR, Patwardhan VC, Weissman J, Sui Moi L. Letter: Neurotoxicity of 5-fluorouracil. Lancet. 1975;1:410.
124.
go back to reference Pirzada NA, Ali II, Dafer RM. Fluorouracil-induced neurotoxicity. Ann Pharmacother. 2000;34:35–8.PubMedCrossRef Pirzada NA, Ali II, Dafer RM. Fluorouracil-induced neurotoxicity. Ann Pharmacother. 2000;34:35–8.PubMedCrossRef
125.
go back to reference el Amrani M, Heinzlef O, Debroucker T, Roullet É, Bousser MG, Amarenco P. Brain infarction following 5-fluorouracil and cisplatin therapy. Neurology. 1998;51:899–901.PubMedCrossRef el Amrani M, Heinzlef O, Debroucker T, Roullet É, Bousser MG, Amarenco P. Brain infarction following 5-fluorouracil and cisplatin therapy. Neurology. 1998;51:899–901.PubMedCrossRef
126.
go back to reference Delval L, Klastersky J. Optic neuropathy in cancer patients. Report of a case possibly related to 5 fluorouracil toxicity and review of the literature. J Neurooncol. 2002;60:165–9.PubMedCrossRef Delval L, Klastersky J. Optic neuropathy in cancer patients. Report of a case possibly related to 5 fluorouracil toxicity and review of the literature. J Neurooncol. 2002;60:165–9.PubMedCrossRef
127.
go back to reference Bixenman WW, Nicholls JVV, Warwick OH. Oculomotor disturbances associated with 5-fluorouracil chemotherapy. Am J Ophthalmol. 1977;83:789–93.PubMedCrossRef Bixenman WW, Nicholls JVV, Warwick OH. Oculomotor disturbances associated with 5-fluorouracil chemotherapy. Am J Ophthalmol. 1977;83:789–93.PubMedCrossRef
128.
go back to reference Stein ME, Drumea K, Yarnitsky D, Benny A, Tzuk-Shina T. A rare event of 5-fluorouracil-associated peripheral neuropathy: a report of two patients. Am J Clin Oncol. 1998;21:248–9.PubMedCrossRef Stein ME, Drumea K, Yarnitsky D, Benny A, Tzuk-Shina T. A rare event of 5-fluorouracil-associated peripheral neuropathy: a report of two patients. Am J Clin Oncol. 1998;21:248–9.PubMedCrossRef
129.
go back to reference Takimoto CH, Lu ZH, Zhang R, Liang MD, Larson LV, Cantilena LR Jr, et al. Severe neurotoxicity following 5-fluorouracil-based chemotherapy in a patient with dihydropyrimidine dehydrogenase deficiency. Clin Cancer Res. 1996;2:477–81.PubMed Takimoto CH, Lu ZH, Zhang R, Liang MD, Larson LV, Cantilena LR Jr, et al. Severe neurotoxicity following 5-fluorouracil-based chemotherapy in a patient with dihydropyrimidine dehydrogenase deficiency. Clin Cancer Res. 1996;2:477–81.PubMed
130.
go back to reference Heier MS, Fosså SD. Neurological manifestations in a phase 2 study of 13 patients treated with doxyfluridine. Acta Neurol Scand. 1985;72:171–5.PubMedCrossRef Heier MS, Fosså SD. Neurological manifestations in a phase 2 study of 13 patients treated with doxyfluridine. Acta Neurol Scand. 1985;72:171–5.PubMedCrossRef
131.
go back to reference Kuzuhara S, Ohkoshi N, Kanemaru K, Hashimoto H, Nakanishi T, Toyokura Y. Subacute leucoencephalopathy induced by carmofur, a 5-fluorouracil derivative. J Neurol. 1987;234:365–70.PubMedCrossRef Kuzuhara S, Ohkoshi N, Kanemaru K, Hashimoto H, Nakanishi T, Toyokura Y. Subacute leucoencephalopathy induced by carmofur, a 5-fluorouracil derivative. J Neurol. 1987;234:365–70.PubMedCrossRef
132.
go back to reference Ohara S, Hayashi R, Hata S, Itoh N, Hanyu N, Yamamoto K. Leukoencephalopathy induced by chemotherapy with tegafur, a 5-fluorouracil derivative. Acta Neuropathol. 1998;96:527–31.PubMedCrossRef Ohara S, Hayashi R, Hata S, Itoh N, Hanyu N, Yamamoto K. Leukoencephalopathy induced by chemotherapy with tegafur, a 5-fluorouracil derivative. Acta Neuropathol. 1998;96:527–31.PubMedCrossRef
133.
go back to reference Renouf D, Gill S. Capecitabine-induced cerebellar toxicity. Clin Colorectal Cancer. 2006;6:70–1. Renouf D, Gill S. Capecitabine-induced cerebellar toxicity. Clin Colorectal Cancer. 2006;6:70–1.
134.
go back to reference Formica V, Leary A, Cunningham D, Chua YJ. 5-Fluorouracil can cross brain-blood barrier and cause encephalopathy: should we expect the same from capecitabine? A case report on capecitabine-induced central neurotoxicity progressing to coma. Cancer Chemother Pharmacol. 2006;58:276–8.PubMedCrossRef Formica V, Leary A, Cunningham D, Chua YJ. 5-Fluorouracil can cross brain-blood barrier and cause encephalopathy: should we expect the same from capecitabine? A case report on capecitabine-induced central neurotoxicity progressing to coma. Cancer Chemother Pharmacol. 2006;58:276–8.PubMedCrossRef
135.
go back to reference Videnovic A, Semenov I, Chua-Adajar R, Baddi L, Blumenthal DT, Beck AC, et al. Capecitabine-induced multifocal leukoencephalopathy: a report of five cases. Neurology. 2005;65:1792–4.PubMedCrossRef Videnovic A, Semenov I, Chua-Adajar R, Baddi L, Blumenthal DT, Beck AC, et al. Capecitabine-induced multifocal leukoencephalopathy: a report of five cases. Neurology. 2005;65:1792–4.PubMedCrossRef
136.
go back to reference Benger A, Browman G, Walker I, Preisler H. Clinical evidence of a cumulative effect of high-dose cytarabine on the cerebellum in patients with acute leukemia: a leukemia intergroup report. Cancer Treat Rep. 1985;69:240–1.PubMed Benger A, Browman G, Walker I, Preisler H. Clinical evidence of a cumulative effect of high-dose cytarabine on the cerebellum in patients with acute leukemia: a leukemia intergroup report. Cancer Treat Rep. 1985;69:240–1.PubMed
137.
go back to reference Dworkin LA, Goldman RD, Zivin LS, Fuchs PC. Cerebellar toxicity following high-dose cytosine arabinoside. J Clin Oncol. 1985;3:613–6.PubMedCrossRef Dworkin LA, Goldman RD, Zivin LS, Fuchs PC. Cerebellar toxicity following high-dose cytosine arabinoside. J Clin Oncol. 1985;3:613–6.PubMedCrossRef
138.
go back to reference Herzig RH, Hines JD, Herzig GP, Wolff SN, Cassileth PA, Lazarus HM, et al. Cerebellar toxicity with high-dose cytosine arabinoside. J Clin Oncol. 1987;5:927–32.PubMedCrossRef Herzig RH, Hines JD, Herzig GP, Wolff SN, Cassileth PA, Lazarus HM, et al. Cerebellar toxicity with high-dose cytosine arabinoside. J Clin Oncol. 1987;5:927–32.PubMedCrossRef
139.
go back to reference Dietrich J, Wen PY. Neurologic complications of chemotherapy. In: Schiff D, Kesari S, Wen PY (Eds). Cancer neurology in Clinical Practice Humana Press; New Jersey. 2008:287–326. Dietrich J, Wen PY. Neurologic complications of chemotherapy. In: Schiff D, Kesari S, Wen PY (Eds). Cancer neurology in Clinical Practice Humana Press; New Jersey. 2008:287–326.
140.
go back to reference Warrell RP, Berman E. Phase I and II study of fludarabine phosphate in leukemia: therapeutic efficacy with delayed central nervous system toxicity. J Clin Oncol. 1986;4:74–9.PubMedCrossRef Warrell RP, Berman E. Phase I and II study of fludarabine phosphate in leukemia: therapeutic efficacy with delayed central nervous system toxicity. J Clin Oncol. 1986;4:74–9.PubMedCrossRef
141.
go back to reference Cheson BD, Vena DA, Foss FM, Sorensen JM. Neurotoxicity of purine analogs: a review. J Clin Oncol. 1994;12:2216–28.PubMedCrossRef Cheson BD, Vena DA, Foss FM, Sorensen JM. Neurotoxicity of purine analogs: a review. J Clin Oncol. 1994;12:2216–28.PubMedCrossRef
142.
go back to reference Berg SL, Blaney SM, Devidas M, Lampkin TA, Murgo A, Bernstein M, et al. Phase II study of nelarabine (compound 506U78) in children and young adults with refractory T-cell malignancies: a report from the Children’s Oncology Group. J Clin Oncol. 2005;23:3376–82.PubMedCrossRef Berg SL, Blaney SM, Devidas M, Lampkin TA, Murgo A, Bernstein M, et al. Phase II study of nelarabine (compound 506U78) in children and young adults with refractory T-cell malignancies: a report from the Children’s Oncology Group. J Clin Oncol. 2005;23:3376–82.PubMedCrossRef
143.
go back to reference Schiff D, Wen PY, van den Bent MJ. Neurological adverse effects caused by cytotoxic and targeted therapies. Nat Rev Clin Oncol. 2009;6:596–603.PubMedCrossRef Schiff D, Wen PY, van den Bent MJ. Neurological adverse effects caused by cytotoxic and targeted therapies. Nat Rev Clin Oncol. 2009;6:596–603.PubMedCrossRef
144.
go back to reference Cohen MH, Johnson JR, Massie T, Sridhara R, McGuinn WD, Abraham S, et al. Approval summary: nelarabine for the treatment of T-cell lymphoblastic leukemia/lymphoma. Clin Cancer Res. 2006;12:5329–35.PubMedCrossRef Cohen MH, Johnson JR, Massie T, Sridhara R, McGuinn WD, Abraham S, et al. Approval summary: nelarabine for the treatment of T-cell lymphoblastic leukemia/lymphoma. Clin Cancer Res. 2006;12:5329–35.PubMedCrossRef
145.
146.
go back to reference DeAngelo DJ, Yu D, Johnson JL, Coutre SE, Stone RM, Stopeck AT, et al. Nelarabine induces complete remissions in adults with relapsed or refractory T-lineage acute lymphoblastic leukemia or lymphoblastic lymphoma: Cancer and Leukemia Group B study 19801. Blood. 2007;109:5136–42.PubMedPubMedCentralCrossRef DeAngelo DJ, Yu D, Johnson JL, Coutre SE, Stone RM, Stopeck AT, et al. Nelarabine induces complete remissions in adults with relapsed or refractory T-lineage acute lymphoblastic leukemia or lymphoblastic lymphoma: Cancer and Leukemia Group B study 19801. Blood. 2007;109:5136–42.PubMedPubMedCentralCrossRef
149.
150.
go back to reference Nunes AT, Annunziata CM. Proteasome inhibitors: structure and function. Semin Oncol. 2017;44:377–80.PubMedCrossRef Nunes AT, Annunziata CM. Proteasome inhibitors: structure and function. Semin Oncol. 2017;44:377–80.PubMedCrossRef
152.
go back to reference Mushtaq A, Kapoor V, Latif A, Iftikhar A, Zahid U, McBride A, et al. Efficacy and toxicity profile of carfilzomib-based regimens for treatment of multiple myeloma. Crit Rev Oncol Hematol. 2018;125:1–11.PubMedPubMedCentralCrossRef Mushtaq A, Kapoor V, Latif A, Iftikhar A, Zahid U, McBride A, et al. Efficacy and toxicity profile of carfilzomib-based regimens for treatment of multiple myeloma. Crit Rev Oncol Hematol. 2018;125:1–11.PubMedPubMedCentralCrossRef
153.
go back to reference Schlafer D, Shah KS, Panjic EH, Lonial S. Safety of proteasome inhibitors for treatment of multiple myeloma. Expert Opin Drug Saf. 2017;16:167–83.PubMed Schlafer D, Shah KS, Panjic EH, Lonial S. Safety of proteasome inhibitors for treatment of multiple myeloma. Expert Opin Drug Saf. 2017;16:167–83.PubMed
156.
go back to reference Yamamoto S, Egashira N. Pathological mechanisms of bortezomib-induced peripheral neuropathy. Int J Mol Sci. 2021;22:1–14.CrossRef Yamamoto S, Egashira N. Pathological mechanisms of bortezomib-induced peripheral neuropathy. Int J Mol Sci. 2021;22:1–14.CrossRef
157.
go back to reference Liu H, Xu R, Huang H. Peripheral neuropathy outcomes and efficacy of subcutaneous bortezomib when combined with thalidomide and dexamethasone in the treatment of multiple myeloma. Exp Ther Med. 2016;12:3041–6.PubMedPubMedCentralCrossRef Liu H, Xu R, Huang H. Peripheral neuropathy outcomes and efficacy of subcutaneous bortezomib when combined with thalidomide and dexamethasone in the treatment of multiple myeloma. Exp Ther Med. 2016;12:3041–6.PubMedPubMedCentralCrossRef
160.
go back to reference Xie JD, Chen SR, Chen H, Pan HL. Bortezomib induces neuropathic pain through protein kinase C-mediated activation of presynaptic NMDA receptors in the spinal cord. Neuropharmacology. 2017;123:477–87.PubMedPubMedCentralCrossRef Xie JD, Chen SR, Chen H, Pan HL. Bortezomib induces neuropathic pain through protein kinase C-mediated activation of presynaptic NMDA receptors in the spinal cord. Neuropharmacology. 2017;123:477–87.PubMedPubMedCentralCrossRef
161.
go back to reference Stockstill K, Doyle TM, Yan X, Chen Z, Janes K, Little JW, et al. Dysregulation of sphingolipid metabolism contributes to bortezomib-induced neuropathic pain. J Exp Med. 2018;215:1301–13.PubMedPubMedCentralCrossRef Stockstill K, Doyle TM, Yan X, Chen Z, Janes K, Little JW, et al. Dysregulation of sphingolipid metabolism contributes to bortezomib-induced neuropathic pain. J Exp Med. 2018;215:1301–13.PubMedPubMedCentralCrossRef
163.
go back to reference Zámečníkova A. Novel approaches to the development of tyrosine kinase inhibitors and their role in the fight against cancer. Expert Opin Drug Discov. 2014;9:77–92.PubMedCrossRef Zámečníkova A. Novel approaches to the development of tyrosine kinase inhibitors and their role in the fight against cancer. Expert Opin Drug Discov. 2014;9:77–92.PubMedCrossRef
165.
go back to reference Broekman F, Giovannetti E, Peters GJ. Tyrosine kinase inhibitors: multi-targeted or single-targeted? World J Clin Oncol. 2011;2:89–93.CrossRef Broekman F, Giovannetti E, Peters GJ. Tyrosine kinase inhibitors: multi-targeted or single-targeted? World J Clin Oncol. 2011;2:89–93.CrossRef
166.
go back to reference Kavanagh S, Nee A, Lipton JH. Emerging alternatives to tyrosine kinase inhibitors for treating chronic myeloid leukemia. Expert Opin Emerg Drugs. 2018;23:51–62.PubMedCrossRef Kavanagh S, Nee A, Lipton JH. Emerging alternatives to tyrosine kinase inhibitors for treating chronic myeloid leukemia. Expert Opin Emerg Drugs. 2018;23:51–62.PubMedCrossRef
167.
go back to reference Rafei H, Jabbour EJ, Kantarjian H, Sinicrope KD, Kamiya-Matsuoka C, Mehta RS, et al. Neurotoxic events associated with BCR-ABL1 tyrosine kinase inhibitors: a case series. Leuk Lymphoma. 2019;60:3292–5.PubMedCrossRef Rafei H, Jabbour EJ, Kantarjian H, Sinicrope KD, Kamiya-Matsuoka C, Mehta RS, et al. Neurotoxic events associated with BCR-ABL1 tyrosine kinase inhibitors: a case series. Leuk Lymphoma. 2019;60:3292–5.PubMedCrossRef
168.
go back to reference Alhawiti N, Burbury KL, Kwa FA, O’Malley CJ, Shuttleworth P, Grigg AP, et al. The tyrosine kinase inhibitor, nilotinib potentiates a prothrombotic state. Thromb Res. 2016;145:54–64.PubMedCrossRef Alhawiti N, Burbury KL, Kwa FA, O’Malley CJ, Shuttleworth P, Grigg AP, et al. The tyrosine kinase inhibitor, nilotinib potentiates a prothrombotic state. Thromb Res. 2016;145:54–64.PubMedCrossRef
169.
go back to reference Coon EA, Zalewski NL, Hoffman EM, Tefferi A, Flemming KD. Nilotinib treatment-associated cerebrovascular disease and stroke. Am J Hematol. 2013;88:534–5.PubMedCrossRef Coon EA, Zalewski NL, Hoffman EM, Tefferi A, Flemming KD. Nilotinib treatment-associated cerebrovascular disease and stroke. Am J Hematol. 2013;88:534–5.PubMedCrossRef
171.
go back to reference Mitsudomi T, Morita S, Yatabe Y, Negoro S, Okamoto I, Tsurutani J, et al. Gefitinib versus cisplatin plus docetaxel in patients with non-small-cell lung cancer harbouring mutations of the epidermal growth factor receptor (WJTOG3405): an open label, randomised phase 3 trial. Lancet Oncol. 2010;11:121–8.PubMedCrossRef Mitsudomi T, Morita S, Yatabe Y, Negoro S, Okamoto I, Tsurutani J, et al. Gefitinib versus cisplatin plus docetaxel in patients with non-small-cell lung cancer harbouring mutations of the epidermal growth factor receptor (WJTOG3405): an open label, randomised phase 3 trial. Lancet Oncol. 2010;11:121–8.PubMedCrossRef
172.
go back to reference Rosell R, Carcereny E, Gervais R, Vergnenegre A, Massuti B, Felip E, et al. Erlotinib versus standard chemotherapy as first-line treatment for European patients with advanced EGFR mutation-positive non-small-cell lung cancer (EURTAC): a multicentre, open-label, randomised phase 3 trial. Lancet Oncol. 2012;13:239–46.PubMedCrossRef Rosell R, Carcereny E, Gervais R, Vergnenegre A, Massuti B, Felip E, et al. Erlotinib versus standard chemotherapy as first-line treatment for European patients with advanced EGFR mutation-positive non-small-cell lung cancer (EURTAC): a multicentre, open-label, randomised phase 3 trial. Lancet Oncol. 2012;13:239–46.PubMedCrossRef
173.
go back to reference Moore MJ, Goldstein D, Hamm J, Figer A, Hecht JR, Gallinger S, et al. Erlotinib plus gemcitabine compared with gemcitabine alone in patients with advanced pancreatic cancer: a phase III trial of the National Cancer Institute of Canada Clinical Trials Group. J Clin Oncol. 2007;25:1960–6.PubMedCrossRef Moore MJ, Goldstein D, Hamm J, Figer A, Hecht JR, Gallinger S, et al. Erlotinib plus gemcitabine compared with gemcitabine alone in patients with advanced pancreatic cancer: a phase III trial of the National Cancer Institute of Canada Clinical Trials Group. J Clin Oncol. 2007;25:1960–6.PubMedCrossRef
174.
go back to reference Diéras V, Miles D, Verma S, Pegram M, Welslau M, Baselga J, et al. Trastuzumab emtansine versus capecitabine plus lapatinib in patients with previously treated HER2-positive advanced breast cancer (EMILIA): a descriptive analysis of final overall survival results from a randomised, open-label, phase 3 trial. Lancet Oncol. 2017;18:732–42.PubMedPubMedCentralCrossRef Diéras V, Miles D, Verma S, Pegram M, Welslau M, Baselga J, et al. Trastuzumab emtansine versus capecitabine plus lapatinib in patients with previously treated HER2-positive advanced breast cancer (EMILIA): a descriptive analysis of final overall survival results from a randomised, open-label, phase 3 trial. Lancet Oncol. 2017;18:732–42.PubMedPubMedCentralCrossRef
175.
go back to reference Bence AK, Anderson EB, Halepota MA, Doukas MA, DeSimone PA, Davis GA, et al. Phase I pharmacokinetic studies evaluating single and multiple doses of oral GW572016, a dual EGFR-ErbB2 inhibitor, in healthy subjects. Invest New Drugs. 2005;23:39–49.PubMedCrossRef Bence AK, Anderson EB, Halepota MA, Doukas MA, DeSimone PA, Davis GA, et al. Phase I pharmacokinetic studies evaluating single and multiple doses of oral GW572016, a dual EGFR-ErbB2 inhibitor, in healthy subjects. Invest New Drugs. 2005;23:39–49.PubMedCrossRef
176.
go back to reference Paz-Ares L, Mezger J, Ciuleanu TE, Fischer JR, von Pawel J, Provencio M, Kazarnowicz A, Losonczy G, de Castro G Jr, Szczesna A, Crino L, Reck M, Ramlau R, Ulsperger E, Schumann C, Miziara JE, Lessa ÁE, Dediu M, Bálint B, Depenbrock H, Soldatenkova V, Kurek R, Hirsch FR, Thatcher N, Socinski MA, INSPIRE investigators. Necitumumab plus pemetrexed and cisplatin as first-line therapy in patients with stage IV non-squamous non-small-cell lung cancer (INSPIRE): an open-label, randomised, controlled phase 3 study. Lancet Oncol. 2015;16:328–37.PubMedCrossRef Paz-Ares L, Mezger J, Ciuleanu TE, Fischer JR, von Pawel J, Provencio M, Kazarnowicz A, Losonczy G, de Castro G Jr, Szczesna A, Crino L, Reck M, Ramlau R, Ulsperger E, Schumann C, Miziara JE, Lessa ÁE, Dediu M, Bálint B, Depenbrock H, Soldatenkova V, Kurek R, Hirsch FR, Thatcher N, Socinski MA, INSPIRE investigators. Necitumumab plus pemetrexed and cisplatin as first-line therapy in patients with stage IV non-squamous non-small-cell lung cancer (INSPIRE): an open-label, randomised, controlled phase 3 study. Lancet Oncol. 2015;16:328–37.PubMedCrossRef
178.
go back to reference Kanaan Z, Kulairi Z, Titianu M, Saha S, Kumar S. Guillain-Barré syndrome following treatment with sunitinib malate. Case Rep Oncol Med. 2014:1–2. Kanaan Z, Kulairi Z, Titianu M, Saha S, Kumar S. Guillain-Barré syndrome following treatment with sunitinib malate. Case Rep Oncol Med. 2014:1–2.
182.
go back to reference Cooper MR, Chim H, Chan H, Durand C. Ceritinib: a new tyrosine kinase inhibitor for non-small-cell lung cancer. Ann Pharmacother. 2015;49:107–12.PubMedCrossRef Cooper MR, Chim H, Chan H, Durand C. Ceritinib: a new tyrosine kinase inhibitor for non-small-cell lung cancer. Ann Pharmacother. 2015;49:107–12.PubMedCrossRef
183.
go back to reference Kroeze SGC, Fritz C, Hoyer M, Lo SS, Ricardi U, Sahgal A, et al. Toxicity of concurrent stereotactic radiotherapy and targeted therapy or immunotherapy: a systematic review. Cancer Treat Rev. 2017;53:25–37.PubMedCrossRef Kroeze SGC, Fritz C, Hoyer M, Lo SS, Ricardi U, Sahgal A, et al. Toxicity of concurrent stereotactic radiotherapy and targeted therapy or immunotherapy: a systematic review. Cancer Treat Rev. 2017;53:25–37.PubMedCrossRef
184.
go back to reference DeMichele A, Clark AS, Tan KS, Heitjan DF, Gramlich K, Gallagher M, et al. CDK 4/6 inhibitor palbociclib (PD0332991) in Rb+ advanced breast cancer: phase II activity, safety, and predictive biomarker assessment. Clin Cancer Res. 2015;21:995–1001.PubMedCrossRef DeMichele A, Clark AS, Tan KS, Heitjan DF, Gramlich K, Gallagher M, et al. CDK 4/6 inhibitor palbociclib (PD0332991) in Rb+ advanced breast cancer: phase II activity, safety, and predictive biomarker assessment. Clin Cancer Res. 2015;21:995–1001.PubMedCrossRef
185.
go back to reference Gurule NJ, Heasley LE. Linking tyrosine kinase inhibitor-mediated inflammation with normal epithelial cell homeostasis and tumor therapeutic responses. Cancer Drug Resist. 2018;1:118–25.PubMed Gurule NJ, Heasley LE. Linking tyrosine kinase inhibitor-mediated inflammation with normal epithelial cell homeostasis and tumor therapeutic responses. Cancer Drug Resist. 2018;1:118–25.PubMed
188.
go back to reference Calabrese V, Scapagnini G, Ravagna A, Colombrita C, Spadaro F, Butterfield DA, et al. Increased expression of heat shock proteins in rat brain during aging: relationship with mitochondrial function and glutathione redox state. Mech Ageing Dev. 2004;125:325–35.PubMedCrossRef Calabrese V, Scapagnini G, Ravagna A, Colombrita C, Spadaro F, Butterfield DA, et al. Increased expression of heat shock proteins in rat brain during aging: relationship with mitochondrial function and glutathione redox state. Mech Ageing Dev. 2004;125:325–35.PubMedCrossRef
190.
go back to reference Spain L, Diem S, Larkin J. Management of toxicities of immune checkpoint inhibitors. Cancer Treat Rev. 2016;44:51–60.PubMedCrossRef Spain L, Diem S, Larkin J. Management of toxicities of immune checkpoint inhibitors. Cancer Treat Rev. 2016;44:51–60.PubMedCrossRef
191.
go back to reference Darvin P, Toor SM, Sasidharan Nair V, Elkord E. Immune checkpoint inhibitors: recent progress and potential biomarkers. Exp Mol Med. 2018;50:1–11.PubMedCrossRef Darvin P, Toor SM, Sasidharan Nair V, Elkord E. Immune checkpoint inhibitors: recent progress and potential biomarkers. Exp Mol Med. 2018;50:1–11.PubMedCrossRef
192.
go back to reference Robert C, Thomas L, Bondarenko I, O’Day S, Weber J, Garbe C, et al. Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. N Engl J Med. 2011;364:2517–26.PubMedCrossRef Robert C, Thomas L, Bondarenko I, O’Day S, Weber J, Garbe C, et al. Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. N Engl J Med. 2011;364:2517–26.PubMedCrossRef
194.
go back to reference Eggermont AMM, Chiarion-Sileni V, Grob JJ, Dummer R, Wolchok JD, Schmidt H, et al. Adjuvant ipilimumab versus placebo after complete resection of high-risk stage III melanoma (EORTC 18071): a randomised, double-blind, phase 3 trial. Lancet Oncol. 2015;16:522–30.PubMedCrossRef Eggermont AMM, Chiarion-Sileni V, Grob JJ, Dummer R, Wolchok JD, Schmidt H, et al. Adjuvant ipilimumab versus placebo after complete resection of high-risk stage III melanoma (EORTC 18071): a randomised, double-blind, phase 3 trial. Lancet Oncol. 2015;16:522–30.PubMedCrossRef
195.
go back to reference Bruna J, Argyriou AA, Anastopoulou GG, Alemany M, Nadal E, Kalofonou F, et al. Incidence and characteristics of neurotoxicity in immune checkpoint inhibitors with focus on neuromuscular events: Experience beyond the clinical trials. J Peripher Nerv Syst. 2020;25:171–7.PubMedCrossRef Bruna J, Argyriou AA, Anastopoulou GG, Alemany M, Nadal E, Kalofonou F, et al. Incidence and characteristics of neurotoxicity in immune checkpoint inhibitors with focus on neuromuscular events: Experience beyond the clinical trials. J Peripher Nerv Syst. 2020;25:171–7.PubMedCrossRef
196.
go back to reference Hui E. Understanding T cell signaling using membrane reconstitution. Immunol Rev. 2019;291:44–56.PubMedCrossRef Hui E. Understanding T cell signaling using membrane reconstitution. Immunol Rev. 2019;291:44–56.PubMedCrossRef
197.
go back to reference Rota G, Niogret C, Dang AT, Barros CR, Fonta NP, Alfei F, et al. Shp-2 is dispensable for establishing T cell exhaustion and for PD-1 signaling in vivo. Cell Rep. 2018;23:39–49.PubMedCrossRef Rota G, Niogret C, Dang AT, Barros CR, Fonta NP, Alfei F, et al. Shp-2 is dispensable for establishing T cell exhaustion and for PD-1 signaling in vivo. Cell Rep. 2018;23:39–49.PubMedCrossRef
200.
go back to reference Bronte V, Zanovello P. Regulation of immune responses by L-arginine metabolism. Nat Rev Immunol. 2005;5:641–54.PubMedCrossRef Bronte V, Zanovello P. Regulation of immune responses by L-arginine metabolism. Nat Rev Immunol. 2005;5:641–54.PubMedCrossRef
201.
go back to reference Aboelella NS, Brandle C, Kim T, Ding ZC, Zhou G. Oxidative stress in the tumor microenvironment and its relevance to cancer immunotherapy. Cancers. 2021;13:1–25.CrossRef Aboelella NS, Brandle C, Kim T, Ding ZC, Zhou G. Oxidative stress in the tumor microenvironment and its relevance to cancer immunotherapy. Cancers. 2021;13:1–25.CrossRef
202.
go back to reference Schachter S, Freeman R. Transient ischemic attack and adriamycin cardiomyopathy. Neurology. 1982;32:1380–1.PubMedCrossRef Schachter S, Freeman R. Transient ischemic attack and adriamycin cardiomyopathy. Neurology. 1982;32:1380–1.PubMedCrossRef
203.
go back to reference Grimm S, Chamberlain M. Hodgkin’s lymphoma: a review of neurologic complications. Adv Hematol. 2011. Grimm S, Chamberlain M. Hodgkin’s lymphoma: a review of neurologic complications. Adv Hematol. 2011.
205.
206.
go back to reference Allan LA, Morrice N, Brady S, Magee G, Pathak S, Clarke PR. Inhibition of caspase-9 through phosphorylation at Thr 125 by ERK MAPK. Nat Cell Biol. 2003;5:647–54.PubMedCrossRef Allan LA, Morrice N, Brady S, Magee G, Pathak S, Clarke PR. Inhibition of caspase-9 through phosphorylation at Thr 125 by ERK MAPK. Nat Cell Biol. 2003;5:647–54.PubMedCrossRef
207.
go back to reference Tangpong J, Cole MP, Sultana R, Joshi G, Estus S, Vore M, et al. Adriamycin-induced, TNF-alpha-mediated central nervous system toxicity. Neurobiol Dis. 2006;23:127–39.PubMedCrossRef Tangpong J, Cole MP, Sultana R, Joshi G, Estus S, Vore M, et al. Adriamycin-induced, TNF-alpha-mediated central nervous system toxicity. Neurobiol Dis. 2006;23:127–39.PubMedCrossRef
208.
go back to reference Rangamani P, Sirovich L. Survival and apoptotic pathways initiated by TNF-alpha: modeling and predictions. Biotechnol Bioeng. 2007;97:1216–29.PubMedCrossRef Rangamani P, Sirovich L. Survival and apoptotic pathways initiated by TNF-alpha: modeling and predictions. Biotechnol Bioeng. 2007;97:1216–29.PubMedCrossRef
209.
go back to reference Alhowail AH, Bloemer J, Majrashi M, Pinky PD, Bhattacharya S, Yongli Z, et al. Doxorubicin-induced neurotoxicity is associated with acute alterations in synaptic plasticity, apoptosis, and lipid peroxidation. Toxicol Mech Methods. 2019;29:457–66.PubMedCrossRef Alhowail AH, Bloemer J, Majrashi M, Pinky PD, Bhattacharya S, Yongli Z, et al. Doxorubicin-induced neurotoxicity is associated with acute alterations in synaptic plasticity, apoptosis, and lipid peroxidation. Toxicol Mech Methods. 2019;29:457–66.PubMedCrossRef
210.
go back to reference Kuznetsov AV, Margreiter R, Amberger A, Saks V, Grimm M. Changes in mitochondrial redox state, membrane potential and calcium precede mitochondrial dysfunction in doxorubicin-induced cell death. Biochim Biophys Acta. 2011;1813:1144–52.PubMedCrossRef Kuznetsov AV, Margreiter R, Amberger A, Saks V, Grimm M. Changes in mitochondrial redox state, membrane potential and calcium precede mitochondrial dysfunction in doxorubicin-induced cell death. Biochim Biophys Acta. 2011;1813:1144–52.PubMedCrossRef
211.
go back to reference Salas-Ramirez KY, Bagnall C, Frias L, Abdali SA, Ahles TA, Hubbard K. Doxorubicin and cyclophosphamide induce cognitive dysfunction and activate the ERK and AKT signaling pathways. Behav Brain Res. 2015;292:133–41.PubMedPubMedCentralCrossRef Salas-Ramirez KY, Bagnall C, Frias L, Abdali SA, Ahles TA, Hubbard K. Doxorubicin and cyclophosphamide induce cognitive dysfunction and activate the ERK and AKT signaling pathways. Behav Brain Res. 2015;292:133–41.PubMedPubMedCentralCrossRef
212.
go back to reference Green PS, Leeuwenburgh C. Mitochondrial dysfunction is an early indicator of doxorubicin-induced apoptosis. Biochim Biophys Acta. 2002;1588:94–101.PubMedCrossRef Green PS, Leeuwenburgh C. Mitochondrial dysfunction is an early indicator of doxorubicin-induced apoptosis. Biochim Biophys Acta. 2002;1588:94–101.PubMedCrossRef
213.
go back to reference Sardão VA, Oliveira PJ, Holy J, Oliveira CR, Wallace KB. Doxorubicin-induced mitochondrial dysfunction is secondary to nuclear p53 activation in H9c2 cardiomyoblasts. Cancer Chemother Pharmacol. 2009;64:811–27.PubMedCrossRef Sardão VA, Oliveira PJ, Holy J, Oliveira CR, Wallace KB. Doxorubicin-induced mitochondrial dysfunction is secondary to nuclear p53 activation in H9c2 cardiomyoblasts. Cancer Chemother Pharmacol. 2009;64:811–27.PubMedCrossRef
215.
go back to reference Mai Y, Yu JJ, Bartholdy B, Xu-Monette ZY, Knapp EE, Yuan F, et al. An oxidative stress-based mechanism of doxorubicin cytotoxicity suggests new therapeutic strategies in ABC-DLBCL. Blood. 2016;128:2797–807.PubMedPubMedCentralCrossRef Mai Y, Yu JJ, Bartholdy B, Xu-Monette ZY, Knapp EE, Yuan F, et al. An oxidative stress-based mechanism of doxorubicin cytotoxicity suggests new therapeutic strategies in ABC-DLBCL. Blood. 2016;128:2797–807.PubMedPubMedCentralCrossRef
218.
go back to reference Feinberg WM, Swenson MR. Cerebrovascular complications of L-asparaginase therapy. Neurology. 1988;38:127–33.PubMedCrossRef Feinberg WM, Swenson MR. Cerebrovascular complications of L-asparaginase therapy. Neurology. 1988;38:127–33.PubMedCrossRef
220.
go back to reference Selleri C, Pane F, Notaro R, Catalano L, Santoro LEF, Luciano L, et al. All-trans-retinoic acid (ATRA) responsive skin relapses of acute promyelocytic leukaemia followed by ATRA-induced pseudotumour cerebri. Br J Haematol. 1996;92:937–40.PubMedCrossRef Selleri C, Pane F, Notaro R, Catalano L, Santoro LEF, Luciano L, et al. All-trans-retinoic acid (ATRA) responsive skin relapses of acute promyelocytic leukaemia followed by ATRA-induced pseudotumour cerebri. Br J Haematol. 1996;92:937–40.PubMedCrossRef
222.
go back to reference Chaudhary UB, Haldas JR. Long-term complications of chemotherapy for germ cell tumours. Drugs. 2003;63:1565–77.PubMedCrossRef Chaudhary UB, Haldas JR. Long-term complications of chemotherapy for germ cell tumours. Drugs. 2003;63:1565–77.PubMedCrossRef
223.
go back to reference Shahab N, Haider S, Doll DC. Vascular toxicity of antineoplastic agents. Semin Oncol. 2006;33:121–38.PubMedCrossRef Shahab N, Haider S, Doll DC. Vascular toxicity of antineoplastic agents. Semin Oncol. 2006;33:121–38.PubMedCrossRef
225.
go back to reference Younes A, Gopal AK, Smith SE, Ansell SM, Rosenblatt JD, Savage KJ, Ramchandren R, Bartlett NL, Cheson BD, de Vos S, Forero-Torres A, Moskowitz CH, Connors JM, Engert A, Larsen EK, Kennedy DA, Sievers EL, Chen R. Results of a pivotal phase II study of brentuximab vedotin for patients with relapsed or refractory Hodgkin’s lymphoma. J Clin Oncol. 2012;30:2183–9.PubMedPubMedCentralCrossRef Younes A, Gopal AK, Smith SE, Ansell SM, Rosenblatt JD, Savage KJ, Ramchandren R, Bartlett NL, Cheson BD, de Vos S, Forero-Torres A, Moskowitz CH, Connors JM, Engert A, Larsen EK, Kennedy DA, Sievers EL, Chen R. Results of a pivotal phase II study of brentuximab vedotin for patients with relapsed or refractory Hodgkin’s lymphoma. J Clin Oncol. 2012;30:2183–9.PubMedPubMedCentralCrossRef
226.
go back to reference Moskowitz CH, Nademanee A, Masszi T, Agura E, Holowiecki J, Abidi MH, Chen AI, Stiff P, Gianni AM, Carella A, Osmanov D, Bachanova V, Sweetenham J, Sureda A, Huebner D, Sievers EL, Chi A, Larsen EK, Hunder NN, Walewski J, AETHERA Study Group. Brentuximab vedotin as consolidation therapy after autologous stem-cell transplantation in patients with Hodgkin’s lymphoma at risk of relapse or progression (AETHERA): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. 2015;385:1853–62.PubMedCrossRef Moskowitz CH, Nademanee A, Masszi T, Agura E, Holowiecki J, Abidi MH, Chen AI, Stiff P, Gianni AM, Carella A, Osmanov D, Bachanova V, Sweetenham J, Sureda A, Huebner D, Sievers EL, Chi A, Larsen EK, Hunder NN, Walewski J, AETHERA Study Group. Brentuximab vedotin as consolidation therapy after autologous stem-cell transplantation in patients with Hodgkin’s lymphoma at risk of relapse or progression (AETHERA): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. 2015;385:1853–62.PubMedCrossRef
229.
230.
go back to reference Dalgleish A, Galustian C. The potential of immunomodulatory drugs in the treatment of solid tumors. Future Oncol. 2010;6:1479–84.PubMedCrossRef Dalgleish A, Galustian C. The potential of immunomodulatory drugs in the treatment of solid tumors. Future Oncol. 2010;6:1479–84.PubMedCrossRef
231.
go back to reference Piccolomo A, Schifone CP, Strafella V, Specchia G, Musto P, Albano F. Immunomodulatory drugs in acute myeloid leukemia treatment. Cancers. 2020;12:1–14.CrossRef Piccolomo A, Schifone CP, Strafella V, Specchia G, Musto P, Albano F. Immunomodulatory drugs in acute myeloid leukemia treatment. Cancers. 2020;12:1–14.CrossRef
233.
go back to reference Mountjoy L, Sebastian S, Jain T, Hilal T, Gonzalez-Velez M, Girardo M, et al. Prediction of immunomodulatory drugs (IMiDs) sensitivity in myeloma via determination of baseline anti-oxidative stress capacity. Leukemia. 2020;34:3060–3.PubMedCrossRef Mountjoy L, Sebastian S, Jain T, Hilal T, Gonzalez-Velez M, Girardo M, et al. Prediction of immunomodulatory drugs (IMiDs) sensitivity in myeloma via determination of baseline anti-oxidative stress capacity. Leukemia. 2020;34:3060–3.PubMedCrossRef
234.
go back to reference Chaudhry V, Cornblath DR, Polydefkis M, Ferguson A, Borrello I. Characteristics of bortezomib- and thalidomide-induced peripheral neuropathy. J Peripher Nerv Syst. 2008;13:275–82.PubMedPubMedCentralCrossRef Chaudhry V, Cornblath DR, Polydefkis M, Ferguson A, Borrello I. Characteristics of bortezomib- and thalidomide-induced peripheral neuropathy. J Peripher Nerv Syst. 2008;13:275–82.PubMedPubMedCentralCrossRef
235.
go back to reference Patel UH, Mir MA, Sivik JK, Raheja D, Pandey MK, Talamo G. Central neurotoxicity of immunomodulatory drugs in multiple myeloma. Hematol Rep. 2015;7:12–4.CrossRef Patel UH, Mir MA, Sivik JK, Raheja D, Pandey MK, Talamo G. Central neurotoxicity of immunomodulatory drugs in multiple myeloma. Hematol Rep. 2015;7:12–4.CrossRef
237.
go back to reference Decourt B, Wilson J, Ritter A, Dardis C, Difilippo FP, Zhuang X, et al. MCLENA-1: a phase II clinical trial for the assessment of safety, tolerability, and efficacy of lenalidomide in patients with mild cognitive impairment due to Alzheimer’s disease. Open Access J Clin Trials. 2020;12:1–13.PubMedPubMedCentralCrossRef Decourt B, Wilson J, Ritter A, Dardis C, Difilippo FP, Zhuang X, et al. MCLENA-1: a phase II clinical trial for the assessment of safety, tolerability, and efficacy of lenalidomide in patients with mild cognitive impairment due to Alzheimer’s disease. Open Access J Clin Trials. 2020;12:1–13.PubMedPubMedCentralCrossRef
239.
go back to reference Sriram K, O’Callaghan JP. Divergent roles for tumor necrosis factor-alpha in the brain. J Neuroimmune Pharmacol. 2007;2:140–53.PubMedCrossRef Sriram K, O’Callaghan JP. Divergent roles for tumor necrosis factor-alpha in the brain. J Neuroimmune Pharmacol. 2007;2:140–53.PubMedCrossRef
241.
go back to reference Xue HX, Fu WY, Cui HD, Yang LL, Zhang N, Zhao LJ. High-dose thalidomide increases the risk of peripheral neuropathy in the treatment of ankylosing spondylitis. Neural Regen Res. 2015;10:814–8.PubMedPubMedCentralCrossRef Xue HX, Fu WY, Cui HD, Yang LL, Zhang N, Zhao LJ. High-dose thalidomide increases the risk of peripheral neuropathy in the treatment of ankylosing spondylitis. Neural Regen Res. 2015;10:814–8.PubMedPubMedCentralCrossRef
242.
go back to reference Kompoliti K, Horn SS. Drug-Induced and iatrogenic neurological disorders. In Textbook of Clinical Neurology: Third Edition. Elsevier Inc.; 2007. p. 1285–318. Kompoliti K, Horn SS. Drug-Induced and iatrogenic neurological disorders. In Textbook of Clinical Neurology: Third Edition. Elsevier Inc.; 2007. p. 1285–318.
Metadata
Title
Role of inflammation and oxidative stress in chemotherapy-induced neurotoxicity
Authors
Pooja Gupta
Tavneet Kaur Makkar
Lavisha Goel
Monika Pahuja
Publication date
20-07-2022
Publisher
Springer US
Published in
Immunologic Research / Issue 6/2022
Print ISSN: 0257-277X
Electronic ISSN: 1559-0755
DOI
https://doi.org/10.1007/s12026-022-09307-7

Other articles of this Issue 6/2022

Immunologic Research 6/2022 Go to the issue