Skip to main content Accessibility help
×
Hostname: page-component-77c89778f8-sh8wx Total loading time: 0 Render date: 2024-07-23T07:08:52.807Z Has data issue: false hasContentIssue false

Chapter 16 - Autoimmune and Inflammatory Encephalopathies as Complications of Cancer

from Section 3 - Specific Syndromes and Diseases

Published online by Cambridge University Press:  27 January 2022

Josep Dalmau
Affiliation:
Universitat de Barcelona
Francesc Graus
Affiliation:
Universitat de Barcelona
Get access

Summary

In this chapter we review the CNS syndromes mediated by autoimmune or inflammatory mechanisms in patients with cancer. Paraneoplastic neurological syndromes (PNS) are considered to be immune-mediated disorders against proteins expressed by the tumour and nervous system. The autoimmune hypothesis is supported by the presence in serum and CSF of antibodies against neural proteins that are also expressed in the tumour. Less frequently the tumour does not express neuronal proteins but predisposes to immune dysregulation and autoimmune mechanisms. Novel cancer therapies that enhance anti-tumour immune responses frequently cause inflammatory CNS disorders. Immune checkpoint inhibitors have been associated with a wide range of immune-related adverse effects, including an increased incidence of PNS. Another type of cancer therapy is based on the use of T cells genetically engineered to express chimeric antigen receptors (CARs) that recognize molecules present on the surface of tumour cells. CAR T cell therapy can cause severe, potentially lethal, encephalopathy syndromes mediated by massive release of cytokines instead of autoimmune mechanisms. Post-transplant autoimmune encephalitis are rare disorders that mostly occur after allogeneic haematopoietic stem cell transplantation. They are related to graft versus host disease and, sometimes, they associate with antibodies against neuronal surface antigens.

Type
Chapter
Information
Publisher: Cambridge University Press
Print publication year: 2022

Access options

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

References

Darnell, RB, Posner, JB. Paraneoplastic syndromes involving the nervous system. N Engl J Med 2003;349:15431554.CrossRefGoogle ScholarPubMed
Rosenfeld, MR, Dalmau, J. Update on paraneoplastic and autoimmune disorders of the central nervous system. Semin Neurol 2010;30:320331.CrossRefGoogle ScholarPubMed
Yshii, LM, Hohlfeld, R, Liblau, RS. Inflammatory CNS disease caused by immune checkpoint inhibitors: status and perspectives. Nat Rev Neurol 2017;13:755763.Google Scholar
Graus, F, Saiz, A, Dalmau, J. Antibodies and neuronal autoimmune disorders of the CNS. J Neurol 2009;257:509517.Google Scholar
Bernal, F, Graus, F, Pifarre, A, et al. Immunohistochemical analysis of anti-Hu-associated paraneoplastic encephalomyelitis. Acta Neuropathol (Berl) 2002;103:509515.Google Scholar
Bien, CG, Vincent, A, Barnett, MH, et al. Immunopathology of autoantibody-associated encephalitides: clues for pathogenesis. Brain 2012;135:16221638.Google Scholar
Graus, F, Delattre, JY, Antoine, JC, et al. Recommended diagnostic criteria for paraneoplastic neurological syndromes. J Neurol Neurosurg Psychiatry 2004;75:11351140.Google Scholar
Dalmau, J, Geis, C, Graus, F. Autoantibodies to synaptic receptors and neuronal cell surface proteins in autoimmune diseases of the central nervous system. Physiol Rev 2017;97:839887.CrossRefGoogle ScholarPubMed
Soussain, C, Ricard, D, Fike, JR, et al. CNS complications of radiotherapy and chemotherapy. Lancet 2009;374:16391651.CrossRefGoogle ScholarPubMed
Staff, NP, Grisold, A, Grisold, W, Windebank, AJ. Chemotherapy-induced peripheral neuropathy: a current review. Ann Neurol 2017;81:772781.Google Scholar
Wu, VC, Huang, JW, Lien, HC, et al. Levamisole-induced multifocal inflammatory leukoencephalopathy: clinical characteristics, outcome, and impact of treatment in 31 patients. Medicine (Baltimore) 2006;85:203213.Google Scholar
Vosoughi, R, Schmidt, BJ. Multifocal leukoencephalopathy in cocaine users: a report of two cases and review of the literature. BMC Neurol 2015;15:208.Google Scholar
Graus, F, Dalmau, J. Paraneoplastic neurological syndromes in the era of immune-checkpoint inhibitors. Nat Rev Clin Oncol 2019;16:535548.Google Scholar
Vogrig, A, Fouret, M, Joubert, B, et al. Increased frequency of anti-Ma2 encephalitis associated with immune checkpoint inhibitors. Neurol Neuroimmunol Neuroinflamm 2019;6:e604.Google Scholar
June, CH, Sadelain, M. Chimeric antigen receptor therapy. N Engl J Med 2018;379:6473.Google Scholar
Neelapu, SS, Tummala, S, Kebriaei, P, et al. Chimeric antigen receptor T-cell therapy: assessment and management of toxicities. Nat Rev Clin Oncol 2018;15:4762.CrossRefGoogle ScholarPubMed
Cohen, JA, Baldassari, LE, Atkins, HL, et al. Autologous hematopoietic cell transplantation for treatment-refractory relapsing multiple sclerosis: position statement from the American Society for Blood and Marrow Transplantation. Biol Blood Marrow Transplant 2019;25:845854.Google Scholar
Sharrack, B, Saccardi, R, Alexander, T, et al. Autologous haematopoietic stem cell transplantation and other cellular therapy in multiple sclerosis and immune-mediated neurological diseases: updated guidelines and recommendations from the EBMT Autoimmune Diseases Working Party (ADWP) and the Joint Accreditation Committee of EBMT and ISCT (JACIE). Bone Marrow Transplant 2019;55:283306.Google Scholar
Maffini, E, Festuccia, M, Brunello, L, et al. Neurologic complications after allogeneic hematopoietic stem cell transplantation. Biol Blood Marrow Transplantat 2017;23:388397.Google Scholar
Saiz, A, Graus, F. Neurologic complications of hematopoietic cell transplantation. Semin Neurol 2010;30:287295.Google Scholar
Graus, F, Titulaer, MJ, Balu, R, et al. A clinical approach to diagnosis of autoimmune encephalitis. Lancet Neurol 2016;15:391404.Google Scholar
Graus, F, Escudero, D, Oleaga, L, et al. Syndrome and outcome of antibody-negative limbic encephalitis. Eur J Neurol 2018;25:10111016.CrossRefGoogle ScholarPubMed
Graus, F, Delattre, JY, Antoine, JC, et al. Recommended diagnostic criteria for paraneoplastic neurological syndromes. J Neurol Neurosurg Psychiatry 2004;75:11351140.Google Scholar
Graus, F, Keime-Guibert, F, Rene, R, et al. Anti-Hu-associated paraneoplastic encephalomyelitis: analysis of 200 patients. Brain 2001;124:11381148.Google Scholar
Honnorat, J, Cartalat-Carel, S, Ricard, D, et al. Onco-neural antibodies and tumour type determine survival and neurological symptoms in paraneoplastic neurological syndromes with Hu or CV2/CRMP5 antibodies. J Neurol Neurosurg Psychiatry 2009;80:412416.Google Scholar
Dalmau, J, Graus, F, Villarejo, A, et al. Clinical analysis of anti-Ma2-associated encephalitis. Brain 2004;127:18311844.CrossRefGoogle ScholarPubMed
Alamowitch, S, Graus, F, Uchuya, M, et al. Limbic encephalitis and small cell lung cancer: clinical and immunological features. Brain 1997;120:923928.CrossRefGoogle ScholarPubMed
van Coevorden-Hameete, MH, de Bruijn, M, de Graaff, E, et al. The expanded clinical spectrum of anti-GABABR encephalitis and added value of KCTD16 autoantibodies. Brain 2019;142:16311643.Google Scholar
Voltz, R, Gultekin, SH, Rosenfeld, MR, et al. A serologic marker of paraneoplastic limbic and brain-stem encephalitis in patients with testicular cancer [see comments]. N Engl J Med 1999;340:17881795.CrossRefGoogle ScholarPubMed
Spatola, M, Sabater, L, Planaguma, J, et al. Encephalitis with mGluR5 antibodies: symptoms and antibody effects. Neurology 2018;90:e1964e1972.Google Scholar
Shavit, YB, Graus, F, Probst, A, Rene, R, Steck, AJ. Epilepsia partialis continua: a new manifestation of anti-Hu-associated paraneoplastic encephalomyelitis. Ann Neurol 1999;45:255258.Google Scholar
Graus, F, Boronat, A, Xifro, X, et al. The expanding clinical profile of anti-AMPA receptor encephalitis. Neurology 2010;74:857859.Google Scholar
Hoftberger, R, Titulaer, MJ, Sabater, L, et al. Encephalitis and GABAB receptor antibodies: novel findings in a new case series of 20 patients. Neurology 2013;81:15001506.Google Scholar
Hoftberger, R, van Sonderen, A, Leypoldt, F, et al. Encephalitis and AMPA receptor antibodies: Novel findings in a case series of 22 patients. Neurology 2015;84:24032412.Google Scholar
Dalmau, J, Lancaster, E, Martinez-Hernandez, E, Rosenfeld, MR, Balice-Gordon, R. Clinical experience and laboratory investigations in patients with anti-NMDAR encephalitis. Lancet Neurol 2011;10:6374.Google Scholar
Spatola, M, Petit-Pedrol, M, Simabukuro, MM, et al. Investigations in GABAA receptor antibody-associated encephalitis. Neurology 2017;88:10121020.Google Scholar
Arino, H, Hoftberger, R, Gresa-Arribas, N, et al. Paraneoplastic neurological syndromes and glutamic acid decarboxylase antibodies. JAMA Neurol 2015;72:874881.Google Scholar
Irani, SR, Pettingill, P, Kleopa, KA, et al. Morvan syndrome: clinical and serological observations in 29 cases. Ann Neurol 2012;72:241255.Google Scholar
Vernino, S, Tuite, P, Adler, CH, et al. Paraneoplastic chorea associated with CRMP-5 neuronal antibody and lung carcinoma. Ann Neurol 2002;51:625630.Google Scholar
Peterson, K, Rosenblum, MK, Kotanides, H, Posner, JB. Paraneoplastic cerebellar degeneration: I. A clinical analysis of 55 anti-Yo antibody-positive patients. Neurology 1992;42:19311937.Google Scholar
Mason, WP, Graus, F, Lang, B, et al. Small-cell lung cancer, paraneoplastic cerebellar degeneration and the Lambert–Eaton myasthenic syndrome. Brain 1997;120:12791300.Google Scholar
Bernal, F, Shamsili, S, Rojas, I, et al. Clinical and immunological features of patients with anti-Tr antibodies. Neurology 2003;60:230234.Google Scholar
Sabater, L, Hoftberger, R, Boronat, A, et al. Antibody repertoire in paraneoplastic cerebellar degeneration and small cell lung cancer. PLoS One 2013;8:e60438.Google Scholar
Pranzatelli, MR, Tate, ED, McGee, NR. Demographic, clinical, and immunologic features of 389 children with opsoclonus-myoclonus syndrome: a cross-sectional study. Front Neurol 2017;8:113.CrossRefGoogle ScholarPubMed
Armangue, T, Titulaer, MJ, Sabater, L, et al. A novel treatment-responsive encephalitis with frequent opsoclonus and teratoma. Ann Neurol 2014;75:435441.CrossRefGoogle ScholarPubMed
Armangue, T, Sabater, L, Torres-Vega, E, et al. Clinical and immunological features of opsoclonus-myoclonus syndrome in the era of neuronal cell surface antibodies. JAMA Neurol 2016;73:417424.Google Scholar
Klaas, JP, Ahlskog, JE, Pittock, SJ, et al. Adult-onset opsoclonus-myoclonus syndrome. Arch Neurol 2012;69:15981607.Google Scholar
Mandel-Brehm, C, Dubey, D, Kryzer, TJ, et al. Kelch-like protein 11 antibodies in seminoma-associated paraneoplastic encephalitis. N Engl J Med 2019;381:4754.Google Scholar
Saiz, A, Bruna, J, Stourac, P, et al. Anti-Hu-associated brainstem encephalitis. J Neurol Neurosurg Psychiatry 2009;80:404407.Google Scholar
Pittock, SJ, Lucchinetti, CF, Lennon, VA. Anti-neuronal nuclear autoantibody type 2: paraneoplastic accompaniments. Ann Neurol 2003;53:580587.Google Scholar
Murinson, BB, Guarnaccia, JB. Stiff-person syndrome with amphiphysin antibodies: distinctive features of a rare disease. Neurology 2008;71:19551958.Google Scholar
Flanagan, EP, McKeon, A, Lennon, VA, et al. Paraneoplastic isolated myelopathy: clinical course and neuroimaging clues. Neurology 2011;76:20892095.Google Scholar
Hoffmann, LA, Jarius, S, Pellkofer, HL, et al. Anti-Ma and anti-Ta associated paraneoplastic neurological syndromes: twenty-two newly diagnosed patients and review of previous cases. J Neurol Neurosurg Psychiatry 2008;79:767773.Google Scholar
Henson, RA, Urich, HE. Cancer and the Nervous System: The Neurological Manifestations of Systemic Malignant Disease. London: Blackwell Scientific, 1982.Google Scholar
Titulaer, MJ, Wirtz, PW, Willems, LN, et al. Screening for small-cell lung cancer: a follow-up study of patients with Lambert–Eaton myasthenic syndrome. J Clin Oncol 2008;26:42764281.CrossRefGoogle ScholarPubMed
Rojas, I, Graus, F, Keime-Guibert, F, et al. Long-term clinical outcome of paraneoplastic cerebellar degeneration and anti-Yo antibodies. Neurology 2000;55:713715.Google Scholar
Pakkala, S, Owonikoko, TK. Immune checkpoint inhibitors in small cell lung cancer. J Thorac Dis 2018;10:S460S467.Google Scholar
Rudin, CM, Awad, MM, Navarro, A, et al. Pembrolizumab or placebo plus etoposide and platinum as first-line therapy for extensive-stage small-cell lung cancer: randomized, double-blind, phase III KEYNOTE-604 study. J Clin Oncol 2020;38:23692379.Google Scholar
Elrington, GM, Murray, NM, Spiro, SG, Newsom-Davis, J. Neurological paraneoplastic syndromes in patients with small cell lung cancer: a prospective survey of 150 patients. J Neurol Neurosurg Psychiat 1991;54:764767.Google Scholar
Gozzard, P, Woodhall, M, Chapman, C, et al. Paraneoplastic neurologic disorders in small cell lung carcinoma: a prospective study. Neurology 2015;85:235239.CrossRefGoogle ScholarPubMed
Vogrig, A, Gigli, GL, Segatti, S, et al. Epidemiology of paraneoplastic neurological syndromes: a population-based study. J Neurol 2019;267:2635.Google Scholar
Hébert, J, Riche, B, Vogrig, A, et al. Epidemiology of paraneoplastic neurologic syndromes and autoimmune encephalitides in France. Neurol Neuroimmunol Neuroinflamm 2020;7:e883.Google Scholar
Henson, RA, Hoffman, HL, Urich, H. Encephalomyelitis with carcinoma. Brain 1965;88:449464.Google Scholar
Pittock, SJ, Lucchinetti, CF, Parisi, JE, et al. Amphiphysin autoimmunity: paraneoplastic accompaniments. Ann Neurol 2005;58:96107.Google Scholar
Yu, Z, Kryzer, TJ, Griesmann, GE, et al.CRMP-5 neuronal autoantibody: marker of lung cancer and thymoma-related autoimmunity. Ann Neurol 2001;49:146154.Google Scholar
Gadoth, A, Kryzer, TJ, Fryer, J, et al. Microtubule-associated protein 1B: novel paraneoplastic biomarker. Ann Neurol 2017;81:266277.Google Scholar
Cross, SA, Salomao, DR, Parisi, JE, et al. Paraneoplastic autoimmune optic neuritis with retinitis defined by CRMP-5-IgG. Ann Neurol 2003;54:3850.Google Scholar
O’Toole, O, Lennon, VA, Ahlskog, JE, et al. Autoimmune chorea in adults. Neurology 2013;80:11331144.Google Scholar
Vigliani, MC, Honnorat, J, Antoine, JC, et al. Chorea and related movement disorders of paraneoplastic origin: the PNS EuroNetwork experience. J Neurol 2011;258:20582068.Google Scholar
Maudes, E, Landa, J, Munoz-Lopetegi, A, et al. Clinical significance of Kelch-like protein 11 antibodies. Neurol Neuroimmunol Neuroinflamm 2020;7:e666.Google Scholar
Dubey, D, Wilson, MR, Clarkson, B, et al. Expanded clinical phenotype, oncological associations, and immunopathologic insights of paraneoplastic Kelch-like protein-11 encephalitis. JAMA Neurol 2020;77:14201429.Google Scholar
Llado, A, Carpentier, AF, Honnorat, J, et al. Hu-antibody-positive patients with or without cancer have similar clinical profiles. J Neurol Neurosurg Psychiatry 2006;77:996997.Google Scholar
Graus, F, Dalmau, J, Rene, R, et al. Anti-Hu antibodies in patients with small-cell lung cancer: association with complete response to therapy and improved survival. J Clin Oncol 1997;15:28662872.Google Scholar
Graus, F, Vogrig, A, Muñiz-Castrillo, S, et al. Updated diagnostic criteria for paraneoplastic neurological syndromes. Neurol Neuroimmunol Neuroinflamm 2021;8:e1014.CrossRefGoogle Scholar
Sepulveda, M, Sola-Valls, N, Escudero, D, et al. Clinical profile of patients with paraneoplastic neuromyelitis optica spectrum disorder and aquaporin-4 antibodies. Mult Scler 2017;24:17531759.Google Scholar
Abboud, H, Rossman, I, Mealy, MA, et al. Neuronal autoantibodies: differentiating clinically relevant and clinically irrelevant results. J Neurol 2017;264:22842292.Google Scholar
Budhram, A, Nicolle, MW, Yang, L. The positive predictive value of onconeural antibody testing: a retrospective review. Can J Neurol Sci 2018;45:577579.Google Scholar
Ebright, MJ, Li, SH, Reynolds, E, et al. Unintended consequences of Mayo paraneoplastic evaluations. Neurology 2018;91:e2057e2066.Google Scholar
Zidan, A, Fein, A, Zuchowski, K. The use, misuse and abuse of paraneoplastic panels in neurological disorders. A retrospective study. Clin Neurol Neurosurg 2019;186:105545.Google Scholar
Seluk, L, Taliansky, A, Yonath, H, et al. A large screen for paraneoplastic neurological autoantibodies; diagnosis and predictive values. Clin Immunol (Orlando, Fla) 2019;199:2936.Google Scholar
Brier, MR, Bucelli, RC, Day, GS. Reader response: unintended consequences of Mayo paraneoplastic evaluations. Neurology 2019;93:603.Google Scholar
Ruiz-García, R, Martínez-Hernández, E, Saiz, A, Dalmau, J, Graus, F. The diagnostic value of onconeural antibodies depends on how they are tested. Front Immunol 2020;11:1482.Google Scholar
Dechelotte, B, Muniz-Castrillo, S, Joubert, B, et al. Diagnostic yield of commercial immunodots to diagnose paraneoplastic neurologic syndromes. Neurol Neuroimmunol Neuroinflamm 2020;7:e701.CrossRefGoogle ScholarPubMed
Dalmau, J, Furneaux, HM, Cordon-Cardo, C, Posner, JB. The expression of the Hu (paraneoplastic encephalomyelitis/sensory neuronopathy) antigen in human normal and tumor tissues. Am J Pathol 1992;141:881886.Google ScholarPubMed
D’Alessandro, V, Muscarella, LA, la Torre, A, et al. Molecular analysis of the HuD gene in neuroendocrine lung cancers. Lung Cancer 2010;67:6975.Google Scholar
Joseph, CG, Darrah, E, Shah, AA, et al. Association of the autoimmune disease scleroderma with an immunologic response to cancer. Science 2014;343:152157.Google Scholar
Small, M, Treilleux, I, Couillault, C, et al. Genetic alterations and tumor immune attack in Yo paraneoplastic cerebellar degeneration. Acta Neuropathol 2018;135:569579.Google Scholar
Pignolet, BS, Gebauer, CM, Liblau, RS. Immunopathogenesis of paraneoplastic neurological syndromes associated with anti-Hu antibodies: a beneficial antitumor immune response going awry. Oncoimmunology 2013;2:e27384.Google Scholar
Hillary, RP, Ollila, HM, Lin, L, et al. Complex HLA association in paraneoplastic cerebellar ataxia with anti-Yo antibodies. J Neuroimmunol 2018;315:2832.Google Scholar
de Graaf, MT, de Beukelaar, JW, Haasnoot, GW, et al. HLA-DQ2+ individuals are susceptible to Hu-Ab associated paraneoplastic neurological syndromes. J Neuroimmunol 2010;226:147149.CrossRefGoogle ScholarPubMed
Carpentier, AF, Rosenfeld, MR, Delattre, JY, et al. DNA vaccination with HuD inhibits growth of a neuroblastoma in mice. Clin Cancer Res 1998;4:28192824.Google Scholar
Yshii, LM, Gebauer, CM, Pignolet, B, et al. CTLA4 blockade elicits paraneoplastic neurological disease in a mouse model. Brain 2016;139:29232934.Google Scholar
Blachere, NE, Orange, DE, Santomasso, BD, et al. T cells targeting a neuronal paraneoplastic antigen mediate tumor rejection and trigger CNS autoimmunity with humoral activation. Eur J Immunol 2014;44:32403251.Google Scholar
Monstad, SE, Drivsholm, L, Storstein, A, et al. Hu and voltage-gated calcium channel (VGCC) antibodies related to the prognosis of small-cell lung cancer. J Clin Oncol 2004;22:795800.Google Scholar
Titulaer, MJ, Klooster, R, Potman, M, et al. SOX antibodies in small-cell lung cancer and Lambert–Eaton myasthenic syndrome: frequency and relation with survival. J Clin Oncol 2009;27:42604267.Google Scholar
Gozzard, P, Chapman, C, Vincent, A, Lang, B, Maddison, P. Novel humoral prognostic markers in small-cell lung carcinoma: a prospective study. PLoS One 2015;10:e0143558.Google Scholar
de Jongste, AH, de Graaf, MT, Martinuzzi, E, et al. Three sensitive assays do not provide evidence for circulating HuD-specific T cells in the blood of patients with paraneoplastic neurological syndromes with anti-Hu antibodies. Neuro-oncol 2012;14:841848.Google Scholar
Ohara, S, Iijima, N, Hayashida, K, Oide, T, Katai, S. Autopsy case of opsoclonus-myoclonus-ataxia and cerebellar cognitive affective syndrome associated with small cell carcinoma of the lung. Mov Disord 2007;22:13201324.Google Scholar
Martinez-Hernandez, E, Horvath, J, Shiloh-Malawsky, Y, et al. Analysis of complement and plasma cells in the brain of patients with anti-NMDAR encephalitis. Neurology 2011;77:589593.Google Scholar
Titulaer, MJ, Soffietti, R, Dalmau, J, et al. Screening for tumours in paraneoplastic syndromes: report of an EFNS Task Force. Eur J Neurol 2011;18:19–e13.Google Scholar
McKeon, A, Apiwattanakul, M, Lachance, DH, et al. Positron emission tomography-computed tomography in paraneoplastic neurologic disorders: systematic analysis and review. Arch Neurol 2010;67:322329.Google Scholar
Darnell, RB, DeAngelis, LM. Regression of small-cell lung carcinoma in patients with paraneoplastic neuronal antibodies. Lancet 1993;341:2122.Google Scholar
Bataller, L, Graus, F, Saiz, A, Vilchez, JJ. Clinical outcome in adult onset idiopathic or paraneoplastic opsoclonus-myoclonus. Brain 2001;124:437443.Google Scholar
Chalk, CH, Murray, NM, Newsom-Davis, J, O’Neill, JH, Spiro, SG. Response of the Lambert–Eaton myasthenic syndrome to treatment of associated small-cell lung carcinoma. Neurology 1990;40:15521556.Google Scholar
Keime-Guibert, F, Graus, F, Broet, P, et al. Clinical outcome of patients with anti-Hu-associated encephalomyelitis after treatment of the tumor. Neurology 1999;53:17191723.Google Scholar
Lipka, AF, Boldingh, MI, van Zwet, EW, et al. Long-term follow-up, quality of life, and survival of patients with Lambert–Eaton myasthenic syndrome. Neurology 2020;94:e511e520.Google Scholar
Mathew, RM, Vandenberghe, R, Garcia-Merino, A, et al. Orchiectomy for suspected microscopic tumor in patients with anti-Ma2-associated encephalitis. Neurology 2007;68:900905.Google Scholar
Keime-Guibert, F, Graus, F, Fleury, A, et al. Treatment of paraneoplastic neurological syndromes with antineuronal antibodies (Anti-Hu, anti-Yo) with a combination of immunoglobulins, cyclophosphamide, and methylprednisolone. J Neurol Neurosurg Psychiatry 2000;68:479482.Google Scholar
Uchuya, M, Graus, F, Vega, F, Reñé, R, Delattre, JY. Intravenous immunoglobulin treatment in paraneoplastic neurological syndromes with antineuronal autoantibodies. J Neurol Neurosurg Psychiat 1996;60:388392.Google Scholar
Vernino, S, O’Neill, BP, Marks, RS, O’Fallon, JR, Kimmel, DW. Immunomodulatory treatment trial for paraneoplastic neurological disorders. Neuro-oncol 2004;6:5562.Google Scholar
Berzero, G, Karantoni, E, Dehais, C, et al. Early intravenous immunoglobulin treatment in paraneoplastic neurological syndromes with onconeural antibodies. J Neurol Neurosurg Psychiatry 2018;89:798–792.Google Scholar
Shams’ili, S, de Beukelaar, J, Gratama, JW, et al. An uncontrolled trial of rituximab for antibody associated paraneoplastic neurological syndromes. J Neurol 2006;253:1620.Google Scholar
Orange, D, Frank, M, Tian, S, et al. Cellular immune suppression in paraneoplastic neurologic syndromes targeting intracellular antigens. Arch Neurol 2012;69:11321140.Google Scholar
de Jongste, AH, van Gelder, T, Bromberg, JE, et al. A prospective open-label study of sirolimus for the treatment of anti-Hu associated paraneoplastic neurological syndromes. Neuro-oncol 2015;17:145150.Google Scholar
van Broekhoven, F, de Graaf, MT, Bromberg, JE, et al. Human chorionic gonadotropin treatment of anti-Hu-associated paraneoplastic neurological syndromes. J Neurol Neurosurg Psychiatry 2010;81:13411344.Google Scholar
Wilbur, C, Yea, C, Licht, C, Irwin, MS, Yeh, EA. An upfront immunomodulatory therapy protocol for pediatric opsoclonus-myoclonus syndrome. Pediatr Blood Cancer 2019;66:e27776.Google Scholar
Korman, AJ, Peggs, KS, Allison, JP. Checkpoint blockade in cancer immunotherapy. Adv Immunol 2006;90:297339.Google Scholar
Ribas, A, Wolchok, JD. Cancer immunotherapy using checkpoint blockade. Science 2018;359:13501355.Google Scholar
Leach, DR, Krummel, MF, Allison, JP. Enhancement of antitumor immunity by CTLA-4 blockade. Science 1996;271:17341736.Google Scholar
Boussiotis, VA. Molecular and biochemical aspects of the PD-1 checkpoint pathway. N Engl J Med 2016;375:17671778.Google Scholar
Sharma, P, Allison, JP. Immune checkpoint targeting in cancer therapy: toward combination strategies with curative potential. Cell 2015;161:205214.Google Scholar
Sharma, P, Allison, JP. Dissecting the mechanisms of immune checkpoint therapy. Nat Rev Immunol 2020;20:7576.Google Scholar
Mikami, T, Liaw, B, Asada, M, et al. Neuroimmunological adverse events associated with immune checkpoint inhibitor: a retrospective, pharmacovigilance study using FAERS database. J Neurooncol 2021;152:135144.Google Scholar
Astaras, C, de Micheli, R, Moura, B, Hundsberger, T, Hottinger, AF. Neurological adverse events associated with immune checkpoint inhibitors: diagnosis and management. Curr Neurol Neurosci Rep 2018;18:19.Google Scholar
Johnson, DB, Manouchehri, A, Haugh, AM, et al. Neurologic toxicity associated with immune checkpoint inhibitors: a pharmacovigilance study. J Immunother Cancer 2019;7:134.Google Scholar
Johnson, DB, McDonnell, WJ, Gonzalez-Ericsson, PI, et al. A case report of clonal EBV-like memory CD4(+) T cell activation in fatal checkpoint inhibitor-induced encephalitis. Nat Med 2019;25:12431250.Google Scholar
Maurice C, Schneider R, Kiehl TR, et al. Subacute CNS Demyelination after Treatment with Nivolumab for Melanoma. Cancer immunology research 2015;3:1299–1302.Google Scholar
Mandel JJ, Olar A, Aldape KD, Tremont-Lukats IW. Lambrolizumab induced central nervous system (CNS) toxicity. J Neurol Sci 2014;344:229–231.Google Scholar
Velasco R, Villagrán M, Jové M, et al. Encephalitis induced by immune checkpoint inhibitors: A systematic review. JAMA Neurol. 2021;78:864–873.Google Scholar
Galmiche, S, Lheure, C, Kramkimel, N, et al. Encephalitis induced by immune checkpoint inhibitors in metastatic melanoma: a monocentric retrospective study. J Eur Acad Dermatol Venereol 2019;33:e440e443.Google Scholar
Larkin, J, Chmielowski, B, Lao, CD, et al. Neurologic serious adverse events associated with nivolumab plus ipilimumab or nivolumab alone in advanced melanoma, including a case series of encephalitis. Oncologist 2017;22:709718.Google Scholar
Stein, MK, Summers, BB, Wong, CA, Box, HL, Cleveland, KO. Meningoencephalitis following ipilimumab administration in metastatic melanoma. Am J Med Sci 2015;350:512513.Google Scholar
Dubey, D, David, WS, Reynolds, KL, et al. Severe neurological toxicity of immune checkpoint inhibitors: growing spectrum. Ann Neurol 2020;87:659669.Google Scholar
Sechi, E, Markovic, SN, McKeon, A, et al. Neurological autoimmunity and immune checkpoint inhibitors: autoantibody profiles and outcomes. Neurology 2020;95:e2442e2452.Google Scholar
Sato, K, Mano, T, Iwata, A, Toda, T. Neurological and related adverse events in immune checkpoint inhibitors: a pharmacovigilance study from the Japanese Adverse Drug Event Report database. J Neurooncol 2019;145:19.Google Scholar
Burke, M, Hardesty, M, Downs, W. A case of severe encephalitis while on PD-1 immunotherapy for recurrent clear cell ovarian cancer. Gynecol Oncol Rep 2018;24:5153.Google Scholar
Cabral, G, Ladeira, F, Gil, N. Nivolumab-induced seronegative encephalitis. J Neuroimmunol 2020;347:577350.Google Scholar
Conry, RM, Sullivan, JC, Nabors, LB, III. Ipilimumab-induced encephalopathy with a reversible splenial lesion. Cancer Immunol Res 2015;3:598601.Google Scholar
Garcia-Monco, JC, Cortina, IE, Ferreira, E, et al. Reversible splenial lesion syndrome (RESLES): what’s in a name? J Neuroimag 2011;21:e114.Google Scholar
Gkoufa, A, Gogas, H, Diamantopoulos, PT, Ziogas, DC, Psichogiou, M. Encephalitis in a patient with melanoma treated with immune checkpoint inhibitors: case presentation and review of the literature. J Immunother (Hagerstown, Md : 1997) 2020;43:224229.Google Scholar
Kapadia, RK, Ney, DE, Hannan, M, et al. Glial fibrillary acidic protein (GFAP) associated autoimmune meningoencephalitis in a patient receiving nivolumab. J Neuroimmunol 2020;344:577259.Google Scholar
Leitinger, M, Varosanec, MV, Pikija, S, et al. Fatal necrotizing encephalopathy after treatment with nivolumab for squamous non-small cell lung cancer: case report and review of the literature. Front Immunol 2018;9:108.Google Scholar
Robert, L, Langner-Lemercier, S, Angibaud, A, et al. Immune-related encephalitis in two patients treated with immune checkpoint inhibitor. Clin Lung Cancer 2020;21:e474e477.Google Scholar
Schneider, S, Potthast, S, Komminoth, P, Schwegler, G, Bohm, S. PD-1 checkpoint inhibitor associated autoimmune encephalitis. Case Rep Oncol 2017;10:473478.Google Scholar
Sanchis-Borja, M, Ricordel, C, Chiappa, AM, et al. Encephalitis related to immunotherapy for lung cancer: analysis of a multicenter cohort. Lung Cancer 2020;143:3639.Google Scholar
Tatsumi, S, Uryu, K, Iwasaki, S, Harada, H. A case of anti-crmp5 paraneoplastic neurological syndrome induced by atezolizumab for small cell lung cancer. Intern Med (Tokyo, Japan) 2020. doi: 10.2169/internalmedicine.4889-20.Google Scholar
Yamaguchi, Y, Nagasawa, H, Katagiri, Y, Wada, M. Atezolizumab-associated encephalitis in metastatic lung adenocarcinoma: a case report. J Med Case Rep 2020;14:88.Google Scholar
Zafar, Z, Vogler, C, Hudali, T, Bhattarai, M. Nivolumab-associated acute demyelinating encephalitis: a case report and literature review. Clin Med Res 2019;17:2933.Google Scholar
Quach, HT, Robbins, CJ, Balko, JM, et al. Severe epididymo-orchitis and encephalitis complicating anti-PD-1 therapy. Oncologist 2019;24:872876.Google Scholar
Sechi, E, Markovic, SN, McKeon, A, et al. Neurologic autoimmunity and immune checkpoint inhibitors: autoantibody profiles and outcomes. Neurology 2020;95:e2442e2452.Google Scholar
Vogrig, A, Muñiz-Castrillo, S, Joubert, B, et al. Central nervous system complications associated with immune checkpoint inhibitors. J Neurol Neurosurg Psychiatry 2020;91:772778.Google Scholar
Chung, M, Jaffer, M, Verma, N, et al. Immune checkpoint inhibitor induced anti-glutamic acid decarboxylase 65 (Anti-GAD 65) limbic encephalitis responsive to intravenous immunoglobulin and plasma exchange. J Neurol 2020;267:10231025.Google Scholar
Williams, TJ, Benavides, DR, Patrice, KA, et al. Association of autoimmune encephalitis with combined immune checkpoint inhibitor treatment for metastatic cancer. JAMA Neurol 2016;73:928933.Google Scholar
Hottinger, AF, de Micheli, R, Guido, V, et al. Natalizumab may control immune checkpoint inhibitor-induced limbic encephalitis. Neurol Neuroimmunol Neuroinflamm 2018;5:e439.Google Scholar
Fellner, A, Makranz, C, Lotem, M, et al. Neurologic complications of immune checkpoint inhibitors. J Neurooncol 2018;137:601609.Google Scholar
Salam, S, Lavin, T, Turan, A. Limbic encephalitis following immunotherapy against metastatic malignant melanoma. BMJ Case Rep 2016;2016:bcr2016215012.Google Scholar
Brown, MP, Hissaria, P, Hsieh, AH, Kneebone, C, Vallat, W. Autoimmune limbic encephalitis with anti-contactin-associated protein-like 2 antibody secondary to pembrolizumab therapy. J Neuroimmunol 2017;305:1618.Google Scholar
Shah, S, Dunn-Pirio, A, Luedke, M, et al. Nivolumab-induced autoimmune encephalitis in two patients with lung adenocarcinoma. Case Rep Neurol Med 2018;2018:2548528.Google Scholar
Papadopoulos, KP, Romero, RS, Gonzalez, G, et al. Anti-Hu-associated autoimmune limbic encephalitis in a patient with PD-1 inhibitor-responsive myxoid chondrosarcoma. Oncologist 2018;23:118120.Google Scholar
Matsuoka, H, Kimura, H, Koba, H, et al. Nivolumab-induced limbic encephalitis with Anti-Hu antibody in a patient with advanced pleomorphic carcinoma of the lung. Clin Lung Cancer 2018;19:e597e599.Google Scholar
Cordes, LM, Davarpanah, NN, Reoma, LB, et al. Neurotoxicities associated with checkpoint inhibitors: two case reports and a review of the literature. Clin Case Rep 2020;8:2432.Google Scholar
Taillefer, VT, Pigeon, M, Chen, M, et al. Very high-dose methylprednisolone for treatment of nivolumab-induced limbic encephalitis: acase report. J Oncol Pharmacy Pract 2020;26:15381543.Google Scholar
Lyons, S, Joyce, R, Moynagh, P, et al. Autoimmune encephalitis associated with Ma2 antibodies and immune checkpoint inhibitor therapy. Pract Neurol 2020;20:256259.Google Scholar
Erol-Yıldız, R, Kızılay, T, Tüzün, E, Mısırlı, H, Türkoğlu, R. Nivolumab-induced autoantibody negative limbic encephalitis in a patient with Hodgkin lymphoma. Leuk Lymphoma 2020;61:15191521.Google Scholar
Hoftberger, R, Titulaer, MJ, Sabater, L, et al. Encephalitis and GABAB receptor antibodies: novel findings in a new case series of 20 patients. Neurology 2013;81:15001506.Google Scholar
Kawamura, R, Nagata, E, Mukai, M, et al. Acute cerebellar ataxia induced by nivolumab. Intern Med (Tokyo, Japan) 2017;56:33573359.Google Scholar
Kopecky, J, Kubecek, O, Geryk, T, et al. Nivolumab induced encephalopathy in a man with metastatic renal cell cancer: a case report. J Med Case Rep 2018;12:262269.Google Scholar
Schröter, N, Weiller, C, Rauer, S, Waller, CF. Anti-glycin-receptor antibody related stiff-person syndrome under treatment with an immune checkpoint inhibitor. J Neurol 2021;268:709711.Google Scholar
Zurko, J, Mehta, A. Association of immune-mediated cerebellitis with immune checkpoint inhibitor therapy. Mayo Clin Proc Innov Qual Outcomes 2018;2:7477.Google Scholar
Vitt, JR, Kreple, C, Mahmood, N, et al. Autoimmune pancerebellitis associated with pembrolizumab therapy. Neurology 2018;91:9193.Google Scholar
Shibaki, R, Murakami, S, Oki, K, Ohe, Y. Nivolumab-induced autoimmune encephalitis in an anti-neuronal autoantibody-positive patient. Japan J Clin Oncol 2019;49:793794.Google Scholar
Gill, A, Perez, MA, Perrone, CM, et al. A case series of PD-1 inhibitor-associated paraneoplastic neurologic syndromes. J Neuroimmunol 2019;334:576980.Google Scholar
Mongay-Ochoa, N, Vogrig, A, Muñiz-Castrillo, S, Honnorat, J. Anti-Hu-associated paraneoplastic syndromes triggered by immune-checkpoint inhibitor treatment. J Neurol 2020;267:21542156.Google Scholar
Brahmer, JR, Lacchetti, C, Schneider, BJ, et al. Management of immune-related adverse events in patients treated with immune checkpoint inhibitor therapy: American Society of Clinical Oncology Clinical Practice Guideline. J Clin Oncol 2018;36:17141768.Google Scholar
Leonardi, GC, Gainor, JF, Altan, M, et al. Safety of Programmed Death-1 pathway inhibitors among patients with non-small-cell lung cancer and preexisting autoimmune disorders. J Clin Oncol 2018;36:19051912.Google Scholar
Manson, G, Maria, ATJ, Poizeau, F, et al. Worsening and newly diagnosed paraneoplastic syndromes following anti-PD-1 or anti-PD-L1 immunotherapies, a descriptive study. J Immunother Cancer 2019;7:337.Google Scholar
Raibagkar, P, Ho, D, Gunturu, KS, Srinivasan, J. Worsening of anti-Hu paraneoplastic neurological syndrome related to anti-PD-1 treatment: case report and review of literature. J Neuroimmunol 2020;341:577184.Google Scholar
Sermer, D, Brentjens, R. CAR T-cell therapy: full speed ahead. Hematol Oncol 2019;37(Suppl. 1):95100.Google Scholar
Wang, Z, Chen, W, Zhang, X, Cai, Z, Huang, W. A long way to the battlefront: CAR T cell therapy against solid cancers. J Cancer 2019;10:31123123.Google Scholar
Roddie, C, O’Reilly, M, Dias Alves Pinto, J, et al. Manufacturing chimeric antigen receptor T cells: issues and challenges. Cytotherapy 2019;21:327340.Google Scholar
Feins, S, Kong, W, Williams, EF, Milone, MC, Fraietta, JA. An introduction to chimeric antigen receptor (CAR) T-cell immunotherapy for human cancer. Am J Hematol 2019;94:S3S9.Google Scholar
Neill, L, Rees, J, Roddie, C. Neurotoxicity-CAR T-cell therapy: what the neurologist needs to know. Pract Neurol 2020;20:285293.Google Scholar
Lee, DW, Santomasso, BD, Locke, FL, et al. ASTCT consensus grading for cytokine release syndrome and neurologic toxicity associated with immune effector cells. Biol Blood Marrow Transplant 2019;25:625638.Google Scholar
Ramos-Casals, M, Brito-Zeron, P, Lopez-Guillermo, A, Khamashta, MA, Bosch, X. Adult haemophagocytic syndrome. Lancet 2014;383:15031516.Google Scholar
Gratton, SM, Powell, TR, Theeler, BJ, et al. Neurological involvement and characterization in acquired hemophagocytic lymphohistiocytosis in adulthood. J Neurol Sci 2015;357:136142.Google Scholar
Rubin, DB, Danish, HH, Ali, AB, et al. Neurological toxicities associated with chimeric antigen receptor T-cell therapy. Brain 2019;142:13341348.Google Scholar
Santomasso, BD, Park, JH, Salloum, D, et al. Clinical and biological correlates of neurotoxicity associated with CAR T-cell therapy in patients with B-cell acute lymphoblastic leukemia. Cancer Discov 2018;8:958971.Google Scholar
Rubin, DB, Al Jarrah, A, Li, K, et al. Clinical predictors of neurotoxicity after chimeric antigen receptor T-cell therapy. JAMA Neurol 2020;77:15361542.Google Scholar
Karschnia, P, Jordan, JT, Forst, DA, et al. Clinical presentation, management, and biomarkers of neurotoxicity after adoptive immunotherapy with CAR T cells. Blood 2019;133:22122221.Google Scholar
Hu, Y, Sun, J, Wu, Z, et al. Predominant cerebral cytokine release syndrome in CD19-directed chimeric antigen receptor-modified T cell therapy. J Hematol Oncol 2016;9:70.Google Scholar
Giavridis, T, van der Stegen, SJC, Eyquem, J, et al. CAR T cell-induced cytokine release syndrome is mediated by macrophages and abated by IL-1 blockade. Nat Med 2018;24:731738.Google Scholar
Sterner, RM, Sakemura, R, Cox, MJ, et al. GM-CSF inhibition reduces cytokine release syndrome and neuroinflammation but enhances CAR-T cell function in xenografts. Blood 2019;133:697709.Google Scholar
Norelli, M, Camisa, B, Barbiera, G, et al. Monocyte-derived IL-1 and IL-6 are differentially required for cytokine-release syndrome and neurotoxicity due to CAR T cells. Nat Med 2018;24:739748.Google Scholar
Gust, J, Hay, KA, Hanafi, LA, et al. Endothelial activation and blood–brain barrier disruption in neurotoxicity after adoptive immunotherapy with CD19 CAR-T Cells. Cancer Discov 2017;7:14041419.Google Scholar
Torre, M, Solomon, IH, Sutherland, CL, et al. Neuropathology of a case with fatal CAR T-Cell-associated cerebral edema. J Neuropathol Exp Neurol 2018;77:877882.Google Scholar
Saiz, A, Graus, F. Neurological complications of hematopoietic cell transplantation. Semin Neurol 2004;24:427434.Google Scholar
Denier, C, Bourhis, JH, Lacroix, C, et al. Spectrum and prognosis of neurologic complications after hematopoietic transplantation. Neurology 2006;67:19901997.Google Scholar
Pruitt, AA, Graus, F, Rosenfeld, MR. Neurological complications of transplantation: part I: hematopoietic cell transplantation. Neurohospitalist 2013;3:2438.Google Scholar
Graus, F, Saiz, A, Sierra, J, et al. Neurologic complications of autologous and allogeneic bone marrow transplantation in patients with leukemia: a comparative study. Neurology 1996;46:10041009.Google Scholar
Siegal, D, Keller, A, Xu, W, et al. Central nervous system complications after allogeneic hematopoietic stem cell transplantation: incidence, manifestations, and clinical significance. Biol Blood Marrow Transplant 2007;13:13691379.Google Scholar
Holbro, A, Abinun, M, Daikeler, T. Management of autoimmune diseases after haematopoietic stem cell transplantation. Br J Haematol 2012;157:281290.Google Scholar
Balaguer-Rosello, A, Bataller, L, Piñana, JL, et al. Noninfectious neurologic complications after allogeneic hematopoietic stem cell transplantation. Biol Blood Marrow Transplant 2019;25:18181824.Google Scholar
Delios, AM, Rosenblum, M, Jakubowski, AA, DeAngelis, LM. Central and peripheral nervous system immune mediated demyelinating disease after allogeneic hemopoietic stem cell transplantation for hematologic disease. J Neurooncol 2012;110:251256.Google Scholar
Krupp, LB, Tardieu, M, Amato, MP, et al. International Pediatric Multiple Sclerosis Study Group criteria for pediatric multiple sclerosis and immune-mediated central nervous system demyelinating disorders: revisions to the 2007 definitions. Mult Scler 2013;19:12611267.Google Scholar
Smith, CI, Aarli, JA, Biberfeld, P, et al. Myasthenia gravis after bone-marrow transplantation. Evidence for a donor origin. N Engl J Med 1983;309:15651568.Google Scholar
Sherer, Y, Shoenfeld, Y. Autoimmune diseases and autoimmunity post-bone marrow transplantation. Bone Marrow Transplant 1998;22:873881.Google Scholar
King, C, Ilic, A, Koelsch, K, Sarvetnick, N. Homeostatic expansion of T cells during immune insufficiency generates autoimmunity. Cell 2004;117:265277.Google Scholar
Das, J, Gill, A, Lo, C, et al. A case of multiple sclerosis-like relapsing remitting encephalomyelitis following allogeneic hematopoietic stem cell transplantation and a review of the published literature. Front Immunol 2020;11:668.Google Scholar
Grauer, O, Wolff, D, Bertz, H, et al. Neurological manifestations of chronic graft-versus-host disease after allogeneic haematopoietic stem cell transplantation: report from the Consensus Conference on Clinical Practice in chronic graft-versus-host disease. Brain 2010;133:28522865.Google Scholar
Thummalapalli, R, Sena, LA, Probasco, JC, Gladstone, DE. Checkpoint inhibitor-induced autoimmune encephalitis reversed by rituximab after allogeneic bone marrow transplant in a patient with Hodgkin lymphoma. Leuk Lymphoma 2019;61:228–230.Google Scholar
Lowas, SR, Lettieri, CK. A case of anti-NMDA receptor encephalitis during dinutuximab therapy for neuroblastoma. J Pediatr Hematol Oncol 2021;43:e127e129.Google Scholar
Nagai, K, Maekawa, T, Terashima, H, Kubota, M, Ishiguro, A. Severe anti-GAD antibody-associated encephalitis after stem cell transplantation. Brain Dev 2019;41:301304.Google Scholar
Rathore, GS, Leung, KS, Muscal, E. Autoimmune encephalitis following bone marrow transplantation. Pediatr Neurol 2015;53:253256.Google Scholar
Pirotte, M, Forte, F, Lutteri, L, et al. Neuronal surface antibody-mediated encephalopathy as manifestation of chronic graft-versus-host disease after allogeneic hematopoietic stem cell transplantation. J Neuroimmunol 2018;323:115118.Google Scholar
Cohen, DA, Lopez-Chiriboga, AS, Pittock, SJ, et al. Posttransplant autoimmune encephalitis. Neurol Neuroimmunol Neuroinflamm 2018;5:e497.Google Scholar
Garré, J, Sprengers, M, Van Melkebeke, D, Laureys, G. EBV-NMDA double positive encephalitis in an immunocompromised patient. J Neurol Sci 2019;396:7677.Google Scholar
Konen, FF, Schwenkenbecher, P, Jendretzky, KF, et al. Severe anti-N-Methyl-D-aspartate receptor encephalitis under immunosuppression after liver transplantation. Front Neurol 2019;10:987.Google Scholar
Randall, A, Huda, S, Jacob, A, Larner, AJ. Autoimmune encephalitis (NMDAR antibody) in a patient receiving chronic post-transplant immunosuppression. Pract Neurol 2018;18:320322.Google Scholar
Zhao, CZ, Erickson, J, Dalmau, J. Clinical reasoning: agitation and psychosis in a patient after renal transplantation. Neurology 2012;79:e41e44.Google Scholar
Mohrmann, RL, Mah, V, Vinters, HV. Neuropathologic findings after bone marrow transplantation: an autopsy study. Hum Pathol 1990;21:630639.Google Scholar
Patchell, RA, White, CL, III, Clark, AW, Beschorner, WE, Santos, GW. Neurologic complications of bone marrow transplantation. Neurology 1985;35:300306.Google Scholar
Ruggiu, M, Cuccuini, W, Mokhtari, K, et al. Case report: central nervous system involvement of human graft versus host disease – report of 7 cases and a review of literature. Medicine (Baltimore) 2017;96:e8303.Google Scholar
Padovan, CS, Yousry, TA, Schleuning, M, et al. Neurological and neuroradiological findings in long-term survivors of allogeneic bone marrow transplantation. Ann Neurol 1998;43:627633.Google Scholar
Min, GJ, Park, S, Park, SS, et al. A case of central nervous system graft-versus-host disease following allogeneic stem cell transplantation. Int J Hematol 2019;110:635639.Google Scholar
Kamble, RT, Chang, CC, Sanchez, S, Carrum, G. Central nervous system graft-versus-host disease: report of two cases and literature review. Bone Marrow Transplant 2007;39:4952.Google Scholar
Sostak, P, Padovan, CS, Eigenbrod, S, et al. Cerebral angiitis in four patients with chronic GVHD. Bone Marrow Transplant 2010;45:11811188.Google Scholar
Nakayama, Y, Kamio, Y, Kato, N, Murayama, Y. Extracranial-intracranial bypass for cerebral vasculitis after graft-versus-host disease: case report and review of the literature. World Neurosurg 2019;123:193196.Google Scholar
Ma, M, Barnes, G, Pulliam, J, et al. CNS angiitis in graft vs host disease. Neurology 2002;59:19941997.Google Scholar
Campbell, JN, Morris, PP. Cerebral vasculitis in graft-versus-host disease: a case report. Am J Neuroradiol 2005;26:654656.Google Scholar
Harvey, CM, Gottipati, R, Schwarz, S, et al. Acute disseminated encephalomyelitis following allo-SCT: central nervous system manifestation of GVHD. Bone Marrow Transplant 2014;49:854856.Google Scholar
Matsuo, Y, Kamezaki, K, Takeishi, S, et al. Encephalomyelitis mimicking multiple sclerosis associated with chronic graft-versus-host disease after allogeneic bone marrow transplantation. Intern Med (Tokyo, Japan) 2009;48:14531456.Google Scholar
Iwasaki, Y, Sako, K, Ohara, Y, et al. Subacute panencephalitis associated with chronic graft-versus- host disease. Acta Neuropathol 1993;85:566572.Google Scholar
Saad, AG, Alyea, EP, III, Wen, PY, Degirolami, U, Kesari, S. Graft-versus-host disease of the CNS after allogeneic bone marrow transplantation. J Clin Oncol 2009;27:e147149.Google Scholar

Save book to Kindle

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

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

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

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

Available formats
×

Save book to Google Drive

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

Available formats
×