Skip to main content Accessibility help
×
Hostname: page-component-5c6d5d7d68-wpx84 Total loading time: 0 Render date: 2024-08-17T14:12:36.149Z Has data issue: false hasContentIssue false

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
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

References

Graus, F, Titulaer, MJ, Balu, R, et al. A clinical approach to diagnosis of autoimmune encephalitis. Lancet Neurol 2016;15:391404.Google Scholar
Rolls, ET. Limbic systems for emotion and for memory, but no single limbic system. Cortex 2015;62:119157.Google Scholar
Dalmau, J, Graus, F. Antibody-mediated encephalitis. N Engl J Med 2018;378:840851.CrossRefGoogle ScholarPubMed
Ehling, P, Melzer, N, Budde, T, Meuth, SG. CD8(+) T cell-mediated neuronal dysfunction and degeneration in limbic encephalitis. Front Neurol 2015;6:163.Google Scholar
Bien, CG, Vincent, A, Barnett, MH, et al. Immunopathology of autoantibody-associated encephalitides: clues for pathogenesis. Brain 2012;135:16221638.Google Scholar
Dubey, D, Pittock, SJ, Kelly, CR, et al. Autoimmune encephalitis epidemiology and a comparison to infectious encephalitis. Ann Neurol 2018;83:166177.Google Scholar
Gultekin, SH, Rosenfeld, MR, Voltz, R, et al. Paraneoplastic limbic encephalitis: neurological symptoms, immunological findings and tumour association in 50 patients. Brain 2000;123(Pt 7):14811494.CrossRefGoogle ScholarPubMed
Graus, F, Escudero, D, Oleaga, L, et al. Syndrome and outcome of antibody-negative limbic encephalitis. Eur J Neurol 2018;25:10111016.CrossRefGoogle ScholarPubMed
Kasper, BS, Taylor, DC, Janz, D, et al. Neuropathology of epilepsy and psychosis: the contributions of J.A.N. Corsellis. Brain 2010;133:37953805.CrossRefGoogle ScholarPubMed
Brierley, JB, Corsellis, JAN, Hierons, R, et al. Subacute encephalitis of later adult life: mainly affecting the limbic areas. Brain 1960;83:357368.Google Scholar
Henson, RA, Hoffman, HL, Urich, H. Encephalomyelitis with carcinoma. Brain 1965;88:449464.CrossRefGoogle ScholarPubMed
Brain, WR, Norris, FH. The Remote Effects of Cancer on the Nervous System. New York: Grune and Stratton, 1965.Google Scholar
Corsellis, JA, Goldberg, GJ, Norton, AR. ‘Limbic encephalitis’ and its association with carcinoma. Brain 1968;91:481496.CrossRefGoogle ScholarPubMed
Case records of the Massachusetts General Hospital. Weekly clinicopathological exercises. Case 39–1988: a 76-year-old man with confusion, agitation, and a gait disorder. N Engl J Med 1988;319:849860.CrossRefGoogle Scholar
Dalmau, J, Graus, F, Rosenblum, MK, Posner, JB. Anti-Hu–associated paraneoplastic encephalomyelitis/sensory neuronopathy: a clinical study of 71 patients. Medicine (Baltimore) 1992;71:5972.CrossRefGoogle ScholarPubMed
Graus, F, Elkon, KB, Cordon-Cardo, C, Posner, JB. Sensory neuronopathy and small cell lung cancer. Antineuronal antibody that also reacts with the tumor. Am J Med 1986;80:4552.Google Scholar
Alamowitch, S, Graus, F, Uchuya, M, et al. Limbic encephalitis and small cell lung cancer: clinical and immunological features. Brain 1997;120:923928.Google Scholar
Buckley, C, Oger, J, Clover, L, et al. Potassium channel antibodies in two patients with reversible limbic encephalitis. Ann Neurol 2001;50:7378.Google Scholar
Lai, M, Huijbers, MG, Lancaster, E, et al. Investigation of LGI1 as the antigen in limbic encephalitis previously attributed to potassium channels: a case series. Lancet Neurol 2010;9:776785.CrossRefGoogle ScholarPubMed
Irani, SR, Alexander, S, Waters, P, et al. Antibodies to Kv1 potassium channel-complex proteins leucine-rich, glioma inactivated 1 protein and contactin-associated protein-2 in limbic encephalitis, Morvan’s syndrome and acquired neuromyotonia. Brain 2010;133:27342748.CrossRefGoogle ScholarPubMed
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.Google Scholar
Lai, M, Hughes, EG, Peng, X, et al. AMPA receptor antibodies in limbic encephalitis alter synaptic receptor location. Ann Neurol 2009;65:424434.Google Scholar
Lancaster, E, Lai, M, Peng, X, et al. Antibodies to the GABA(B) receptor in limbic encephalitis with seizures: case series and characterisation of the antigen. Lancet Neurol 2010;9:6776.CrossRefGoogle Scholar
Nokura, K, Yamamoto, H, Okawara, Y, et al. Reversible limbic encephalitis caused by ovarian teratoma. Acta Neurol Scand 1997;95:367373.Google Scholar
Rajappa, S, Digumarti, R, Immaneni, SR, Parage, M. Primary renal lymphoma presenting with paraneoplastic limbic encephalitis. J Clin Oncol 2007;25:37833785.Google Scholar
Bien, CG, Urbach, H, Schramm, J, et al. Limbic encephalitis as a precipitating event in adult-onset temporal lobe epilepsy. Neurology 2007;69:12361244.CrossRefGoogle ScholarPubMed
Gultekin, HS, Rosenfeld, MR, Voltz, RD, et al. Paraneoplastic limbic encephalitis: neurological symptoms, immunological findings and tumor association in 50 patients. Brain 2000;123:14811494.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
Newman, NJ, Bell, IR, McKee, AC. Paraneoplastic limbic encephalitis: neuropsychiatric presentation. Biol Psychiatry 1990;27:529542.Google Scholar
Iranzo, A, Graus, F, Clover, L, et al. Rapid eye movement sleep behavior disorder and potassium channel antibody-associated limbic encephalitis. Ann Neurol 2006;59:178181.CrossRefGoogle ScholarPubMed
Irani, SR, Michell, AW, Lang, B, et al. Faciobrachial dystonic seizures precede Lgi1 antibody limbic encephalitis. Ann Neurol 2011;69:892900.Google Scholar
Maureille, A, Fenouil, T, Joubert, B, et al. Isolated seizures are a common early feature of paraneoplastic anti-GABAB receptor encephalitis. J Neurol 2019;266:195206.Google Scholar
Harrison, NA, Johnston, K, Corno, F, et al. Psychogenic amnesia: syndromes, outcome, and patterns of retrograde amnesia. Brain 2017;140:24982510.Google Scholar
Pertzov, Y, Miller, TD, Gorgoraptis, N, et al. Binding deficits in memory following medial temporal lobe damage in patients with voltage-gated potassium channel complex antibody-associated limbic encephalitis. Brain 2013;136:24742485.CrossRefGoogle ScholarPubMed
Butler, CR, Miller, TD, Kaur, MS, et al. Persistent anterograde amnesia following limbic encephalitis associated with antibodies to the voltage-gated potassium channel complex. J Neurol Neurosurg Psychiatry 2014;85:387391.Google Scholar
Baddeley, A. Working memory. Science 1992;255:556559.Google Scholar
Baddeley, A. The concept of episodic memory. Philos Trans Roy Soc Lond B Biol Sci 2001;356:13451350.Google Scholar
Finke, C, Pruss, H, Heine, J, et al. Evaluation of cognitive deficits and structural hippocampal damage in encephalitis with leucine-rich, glioma-inactivated 1 antibodies. JAMA Neurol 2017;74:5059.Google Scholar
Kayser, MS, Kohler, CG, Dalmau, J. Psychiatric manifestations of paraneoplastic disorders. Am J Psychiatry 2010;167:10391050.CrossRefGoogle ScholarPubMed
Jang, Y, Lee, ST, Lim, JA, et al. Psychiatric symptoms delay the diagnosis of anti-LGI1 encephalitis. J Neuroimmunol 2018;317:814.Google Scholar
Kayser, MS, Titulaer, MJ, Gresa-Arribas, N, Dalmau, J. Frequency and characteristics of isolated psychiatric episodes in anti-N-methyl-d-aspartate receptor encephalitis. JAMA Neurol 2013;70:11331139.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.CrossRefGoogle Scholar
Gadoth, A, Pittock, SJ, Dubey, D, et al. Expanded phenotypes and outcomes among 256 LGI1/CASPR2-IgG-positive patients. Ann Neurol 2017;82:7992.CrossRefGoogle ScholarPubMed
Rocamora, R, Becerra, JL, Fossas, P, et al. Pilomotor seizures: an autonomic semiology of limbic encephalitis? Seizure 2014;23:670673.Google Scholar
Tenyi, D, Bone, B, Horvath, R, et al. Ictal piloerection is associated with high-grade glioma and autoimmune encephalitis: results from a systematic review. Seizure 2019;64:15.CrossRefGoogle ScholarPubMed
Kohler, J, Hufschmidt, A, Hermle, L, Volk, B, Lucking, CH. Limbic encephalitis: two cases. J Neuroimmunol 1988;20:177178.CrossRefGoogle ScholarPubMed
Lacomis, D, Khoshbin, S, Schick, RM. MR imaging of paraneoplastic limbic encephalitis. J Comput Assist Tomogr 1990;14:115117.CrossRefGoogle ScholarPubMed
Urbach, H, Soeder, BM, Jeub, M, et al. Serial MRI of limbic encephalitis. Neuroradiology 2006;48:380386.Google Scholar
Dalmau, J, Graus, F, Villarejo, A, et al. Clinical analysis of anti-Ma2-associated encephalitis. Brain 2004;127:18311844.Google Scholar
Dirr, LY, Elster, AD, Donofrio, PD, Smith, M. Evolution of brain MRI abnormalities in limbic encephalitis. Neurology 1990;40:13041306.Google Scholar
Miller, TD, Chong, TT, Aimola Davies, AM, et al. Focal CA3 hippocampal subfield atrophy following LGI1 VGKC-complex antibody limbic encephalitis. Brain 2017;140:12121219.Google Scholar
Arino, H, Armangue, T, Petit-Pedrol, M, et al. Anti-LGI1-associated cognitive impairment: presentation and long-term outcome. Neurology 2016;87:759765.Google Scholar
Morbelli, S, Arbizu, J, Booij, J, et al. The need of standardization and of large clinical studies in an emerging indication of [(18)F]FDG PET: the autoimmune encephalitis. Eur J Nucl Med Molec Imag 2017;44:353357.Google Scholar
Ances, BM, Vitaliani, R, Taylor, RA, et al. Treatment-responsive limbic encephalitis identified by neuropil antibodies: MRI and PET correlates. Brain 2005;128:17641777.Google Scholar
Baumgartner, A, Rauer, S, Mader, I, Meyer, PT. Cerebral FDG-PET and MRI findings in autoimmune limbic encephalitis: correlation with autoantibody types. J Neurol 2013;260:27442753.CrossRefGoogle ScholarPubMed
Fakhoury, T, Abou-Khalil, B, Kessler, RM. Limbic encephalitis and hyperactive foci on PET scan. Seizure 1999;8:427431.Google Scholar
Salmon, E, Sadzot, B, Maquet, P, Franck, G. Results on coregistration of mediotemporal 18F-fluoro-2-deoxy-D-glucose-positron emission tomography (FDG-PET) hyperactivity and 3D magnetic resonance imaging hyperintense lesions in limbic encephalitis. J Neuroimag 2002;12:282.Google Scholar
Scheid, R, Lincke, T, Voltz, R, von Cramon, DY, Sabri, O. Serial 18F-fluoro-2-deoxy-D-glucose positron emission tomography and magnetic resonance imaging of paraneoplastic limbic encephalitis. Arch Neurol 2004;61:17851789.CrossRefGoogle ScholarPubMed
Henry, TR, Babb, TL, Engel, J Jr., et al. Hippocampal neuronal loss and regional hypometabolism in temporal lobe epilepsy. Ann Neurol 1994;36:925927.Google Scholar
Spatola, M, Stojanova, V, Prior, JO, Dalmau, J, Rossetti, AO. Serial brain (1)(8)FDG-PET in anti-AMPA receptor limbic encephalitis. J Neuroimmunol 2014;271:5355.CrossRefGoogle Scholar
Provenzale, JM, Barboriak, DP, Coleman, RE. Limbic encephalitis: comparison of FDG PET and MR imaging findings. AJR Am J Roentgenol 1998;170:16591660.Google Scholar
Kassubek, J, Juengling, FD, Nitzsche, EU, Lucking, CH. Limbic encephalitis investigated by 18FDG-PET and 3D MRI. J Neuroimaging 2001;11:5559.CrossRefGoogle Scholar
Na, DL, Hahm, DS, Park, JM, Kim, SE. Hypermetabolism of the medial temporal lobe in limbic encephalitis on (18)FDG-PET scan: a case report. Eur Neurol 2001;45:187189.Google Scholar
Troester, F, Weske, G, Schlaudraff, E, Passlick, B, Kraemer, K. Image of the month. FDG-PET in paraneoplastic limbic encephalitis. Eur J Nucl Med Molec Imag 2009;36:539.Google Scholar
Maffione, AM, Chondrogiannis, S, Ferretti, A, Al-Nahhas, A, Rubello, D. Correlative imaging with (18)F-FDG PET/CT and MRI in paraneoplastic limbic encephalitis. Clin Nucl Med 2013;38:463464.Google Scholar
Su, M, Xu, D, Tian, R. (18)F-FDG PET/CT and MRI findings in a patient with anti-GABA(B) receptor encephalitis. Clin Nucl Med 2015;40:515517.CrossRefGoogle Scholar
Kim, TJ, Lee, ST, Shin, JW, et al. Clinical manifestations and outcomes of the treatment of patients with GABAB encephalitis. J Neuroimmunol 2014;270:4550.CrossRefGoogle ScholarPubMed
Cozar Santiago Mdel, P, Sanchez Jurado, R, Sanz Llorens, R, Aguilar Barrios, JE, Ferrer Rebolleda, J. Limbic encephalitis diagnosed with 18F-FDG PET/CT. Clin Nucl Med 2016;41:e101103.Google Scholar
Castagnoli, H, Manni, C, Marchesani, F, et al. The role of 18F-FDG PET/CT in management of paraneoplastic limbic encephalitis combined with small cell lung cancer: a case report. Medicine (Baltimore) 2019;98:e16593.Google Scholar
Taneja, S, Suri, V, Ahuja, A, Jena, A. Simultaneous 18F-FDG PET/MRI in autoimmune limbic encephalitis. Ind J Nucl Med 2018;33:174176.Google Scholar
Deuschl, C, Ruber, T, Ernst, L, et al. 18F-FDG-PET/MRI in the diagnostic work-up of limbic encephalitis. PLoS One 2020;15:e0227906.Google Scholar
Longo, R, Wagner, M, Savenkoff, B, et al. A paraneoplastic limbic encephalitis from an anorectal small cell neuroendocrine carcinoma: a case report. BMC Neurol 2019;19:304.Google Scholar
Rey, C, Koric, L, Guedj, E, et al. Striatal hypermetabolism in limbic encephalitis. J Neurol 2012;259:11061110.Google Scholar
Moloney, P, Boylan, R, Elamin, M, et al. Semi-quantitative analysis of cerebral FDG-PET reveals striatal hypermetabolism and normal cortical metabolism in a case of VGKCC limbic encephalitis. Neuroradiol J 2017;30:160163.Google Scholar
Maeder-Ingvar, M, Prior, JO, Irani, SR, et al. FDG-PET hyperactivity in basal ganglia correlating with clinical course in anti-NDMA-R antibodies encephalitis. J Neurol Neurosurg Psychiatry 2011;82:235236.Google Scholar
Krakauer, M, Law, I FDG PET brain imaging in neuropsychiatric systemic lupus erythematosis with choreic symptoms. Clin Nucl Med 2009;34:122123.Google Scholar
Goldman, S, Amrom, D, Szliwowski, HB Reversible striatalhypermetabolismin a case of Sydenham’s chorea. Mov Disord 1993;8:355358.Google Scholar
Psimaras, D, Carpentier, AF, Rossi, C. Cerebrospinal fluid study in paraneoplastic syndromes. J Neurol Neurosurg Psychiatry 2010;81:4245.CrossRefGoogle ScholarPubMed
Venkatesan, A, Michael, BD, Probasco, JC, Geocadin, RG, Solomon, T. Acute encephalitis in immunocompetent adults. Lancet 2019;393:702716.Google Scholar
Tyler, KL. Acute viral encephalitis. N Engl J Med 2018;379:557566.Google Scholar
Oyanguren, B, Sanchez, V, Gonzalez, FJ, et al. Limbic encephalitis: a clinical-radiological comparison between herpetic and autoimmune etiologies. Eur J Neurol 2013;20:15661570.Google Scholar
Chow, FC, Glaser, CA, Sheriff, H, et al. Use of clinical and neuroimaging characteristics to distinguish temporal lobe herpes simplex encephalitis from its mimics. Clin Infect Dis 2015;60:13771383.Google Scholar
Ward, KN. Child and adult forms of human herpesvirus 6 encephalitis: looking back, looking forward. Curr Opin Neurol 2014;27:349355.Google Scholar
Ongradi, J, Ablashi, DV, Yoshikawa, T, Stercz, B, Ogata, M. Roseolovirus-associated encephalitis in immunocompetent and immunocompromised individuals. J Neurovirol 2017;23:119.Google Scholar
Isaacson, E, Glaser, CA, Forghani, B, et al. Evidence of human herpesvirus 6 infection in 4 immunocompetent patients with encephalitis. Clin Infect Dis 2005;40:890893.Google Scholar
Seeley, WW, Marty, FM, Holmes, TM, et al. Post-transplant acute limbic encephalitis: clinical features and relationship to HHV6. Neurology 2007;69:156165.Google Scholar
Vinnard, C, Barton, T, Jerud, E, Blumberg, E. A report of human herpesvirus 6-associated encephalitis in a solid organ transplant recipient and a review of previously published cases. Liver Transpl 2009;15:12421246.Google Scholar
Bhanushali, MJ, Kranick, SM, Freeman, AF, et al. Human herpes 6 virus encephalitis complicating allogeneic hematopoietic stem cell transplantation. Neurology 2013;80:14941500.CrossRefGoogle ScholarPubMed
Yilmaz, M, Yasar, C, Aydin, S, et al. Human herpesvirus 6 encephalitis in an immunocompetent pregnant patient and review of the literature. Clin Neurol Neurosurg 2018;171:106108.Google Scholar
Filippova, A, Charles, J, Epaulard, O, et al. Exogenous human herpesvirus 6 reinfection after tumor-infiltrating T-lymphocyte therapy. Cytotherapy 2018;20:521523.Google Scholar
Athauda, D, Delamont, RS, Pablo-Fernandez, ED. High grade glioma mimicking voltage gated potassium channel complex associated antibody limbic encephalitis. Case Rep Neurologic Med 2014;2014:458790.Google Scholar
Vogrig, A, Joubert, B, Ducray, F, et al. Glioblastoma as differential diagnosis of autoimmune encephalitis. J Neurol 2018;265:669677.Google Scholar
Hainsworth, JB, Shishido, A, Theeler, BJ, Carroll, CG, Fasano, RE. Treatment responsive GABA(B)-receptor limbic encephalitis presenting as new-onset super-refractory status epilepticus (NORSE) in a deployed U.S. soldier. Epileptic Disord 2014;16:486493.Google Scholar
Chevret, L, Husson, B, Nguefack, S, Nehlig, A, Bouilleret, V. Prolonged refractory status epilepticus with early and persistent restricted hippocampal signal MRI abnormality. J Neurol 2008;255:112116.Google Scholar
Tien, RD, Felsberg, GJ. The hippocampus in status epilepticus: demonstration of signal intensity and morphologic changes with sequential fast spin-echo MR imaging. Radiology 1995;194:249256.Google Scholar
Chan, S, Chin, SS, Kartha, K, et al. Reversible signal abnormalities in the hippocampus and neocortex after prolonged seizures. Am J Neuroradiol 1996;17:17251731.Google Scholar
Kim, JA, Chung, JI, Yoon, PH, et al. Transient MR signal changes in patients with generalized tonicoclonic seizure or status epilepticus: periictal diffusion-weighted imaging. Am J Neuroradiol 2001;22:11491160.Google Scholar
Budhram, A, Britton, JW, Liebo, GB, et al. Use of diffusion-weighted imaging to distinguish seizure-related change from limbic encephalitis. J Neurol 2020;267:33373342.Google Scholar
Scriven, J, Davies, S, Banerjee, AK, Jenkins, N, Watson, J. Limbic encephalitis secondary to HIV seroconversion. Int J STD AIDS 2011;22:236237.Google Scholar
Blanc, F, Ben Abdelghani, K, Schramm, F, et al. Whipple limbic encephalitis. Arch Neurol 2011;68:14711473.Google Scholar
Scheid, R, Voltz, R, Vetter, T, Sabri, O, von Cramon, DY. Neurosyphilis and paraneoplastic limbic encephalitis: important differential diagnoses. J Neurol 2005;252:11291132.Google Scholar
Derouich, I, Messouak, O, Belahsen, MF. Syphilitic limbic encephalitis revealed by status epilepticus. BMJ Case Rep 2013;2013.Google Scholar
Kano, O, Arasaki, K, Ikeda, K, et al. Limbic encephalitis associated with systemic lupus erythematosus. Lupus 2009;18:13161319.Google Scholar
Finelli, PF, Inoa, V. Limbic encephalitis as the presenting feature of Sjogren syndrome. Neurol Clin Pract 2013;3:165167.Google Scholar
Kumar, N, Leep Hunderfund, AN, Kutzbach, BR, Pulido, JS, Miller, GM. A limbic encephalitis MR imaging in a patient with Behcet disease and relapsing polychondritis. AM J Neuroradiol 2009;30:E96.Google Scholar
Yoneda, M, Fujii, A, Ito, A, et al. High prevalence of serum autoantibodies against the amino terminal of alpha-enolase in Hashimoto’s encephalopathy. J Neuroimmunol 2007;185:195200.Google Scholar
Fujii, A, Yoneda, M, Ito, T, et al. Autoantibodies against the amino terminal of alpha-enolase are a useful diagnostic marker of Hashimoto’s encephalopathy. J Neuroimmunol 2005;162:130136.Google Scholar
Kishitani, T, Matsunaga, A, Ikawa, M, et al. Limbic encephalitis associated with anti-NH2-terminal of alpha-enolase antibodies: a clinical subtype of Hashimoto encephalopathy. Medicine (Baltimore) 2017;96:e6181.Google Scholar
Hayashi, Y, Yamada, M, Kimura, A, et al. Clinical findings of a probable case of MM2-cortical-type sporadic Creutzfeldt-Jakob disease with antibodies to anti-N-terminus of alpha-enolase. Prion 2017;11:454464.Google Scholar
Macchi, ZA, Kleinschmidt-DeMasters, BK, Orjuela, KD, et al. Glioblastoma as an autoimmune limbic encephalitis mimic: a case and review of the literature. J Neuroimmunol 2020;342:577214.Google Scholar
Verhelst, H, Van Coster, R, Bockaert, N, et al. Limbic encephalitis as presentation of a SAP deficiency. Neurology 2007;69:218219.CrossRefGoogle ScholarPubMed
Langheinrich, TC, Romanowski, CA, Wharton, S, Hadjivassiliou, M. Presenilin-1 mutation associated with amnesia, ataxia, and medial temporal lobe T2 signal changes. Neurology 2011;76:14351436.Google Scholar
Goncalves, LF, Debelenko, LV, Bhambhani, KJ, Scheid, A, Altinok, D. Histiocytic necrotizing lymphadenitis (Kikuchi-Fujimoto disease) with CNS involvement in a child. Pediatr Radiol 2014;44:234238.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
Dorresteijn, LD, Kappelle, AC, Renier, WO, Gijtenbeek, JM. Anti-amphiphysin associated limbic encephalitis: a paraneoplastic presentation of small-cell lung carcinoma. J Neurol 2002;249:13071308.CrossRefGoogle ScholarPubMed
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
Zuliani, L, Saiz, A, Tavolato, B, et al. Paraneoplastic limbic encephalitis associated with potassium channel antibodies: value of anti-glial nuclear antibodies in identifying the tumour. J Neurol Neurosurg Psychiatry 2007;78:204205.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.Google Scholar
Kellinghaus, C, Kraus, J, Blaes, F, Nabavi, DG, Schabitz, WR. CRMP-5-autoantibodies in testicular cancer associated with limbic encephalitis and choreiform dyskinesias. Eur Neurol 2007;57:241243.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
Arino, H, Hoftberger, R, Gresa-Arribas, N, et al. Paraneoplastic neurological syndromes and glutamic acid decarboxylase antibodies. JAMA Neurol 2015;72:874881.Google Scholar
van Sonderen, A, Arino, H, Petit-Pedrol, M, et al. The clinical spectrum of Caspr2 antibody-associated disease. Neurology 2016;87:521528.CrossRefGoogle ScholarPubMed
Mitchell, AN, Bakhos, CT, Zimmerman, EA. Anti-Ri-associated paraneoplastic brainstem cerebellar syndrome with coexisting limbic encephalitis in a patient with mixed large cell neuroendocrine lung carcinoma. J Clin Neurosci 2015;22:421423.Google Scholar
Sutton, I, Winer, J, Rowlands, D, Dalmau, J. Limbic encephalitis and antibodies to Ma2: a paraneoplastic presentation of breast cancer. J Neurol Neurosurg Psychiatry 2000;69:266268.Google Scholar
Krishna, VR, Knievel, K, Ladha, S, Sivakumar, K. Lower extremity predominant stiff-person syndrome and limbic encephalitis with amphiphysin antibodies in breast cancer. J Clin Neuromusc Dis 2012;14:7274.Google Scholar
Spatola, M, Sabater, L, Planaguma, J, et al. Encephalitis with mGluR5 antibodies: symptoms and antibody effects. Neurology 2018;90:e1964e1972.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
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, Dalmou, 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
Yu, Z, Kryzer, TJ, Griesmann, GE, Kim, K, Benarroch, EE, Lennon, VA. CRMP-5 neuronal autoantibody: marker of lung cancer and thymoma-related autoimmunity. Ann Neurol 2001;49:146154.Google Scholar
Rosenfeld, MR, Eichen, JG, Wade, DF, Posner, JB, Dalmau, J. Molecular and clinical diversity in paraneoplastic immunity to Ma proteins. Ann Neurol 2001;50:339348.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
Carreno, M, Bien, CG, Asadi-Pooya, AA, et al. Epilepsy surgery in drug resistant temporal lobe epilepsy associated with neuronal antibodies. Epilepsy Res 2017;129:101105.CrossRefGoogle ScholarPubMed
Engel, J Jr, Van Ness, P, Rasmussen, TB, Ojemann, LM. Outcome with respect to epileptic seizures. In: Engel, J Jr, ed. Surgical Treatment of the Epilepsies, 2nd ed. New York: Raven Press, 1993:609621.Google Scholar
Spatola, M, Dalmau, J. Seizures and risk of epilepsy in autoimmune and other inflammatory encephalitis. Curr Opin Neurol 2017;30:345353.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
McKay, JH, Dimberg, EL, Lopez Chiriboga, AS. A systematic review of gamma-aminobutyric acid receptor type B autoimmunity. Neurologia i neurochirurgia polska 2019;53:17.Google Scholar
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
Joubert, B, Kerschen, P, Zekeridou, A, et al. Clinical spectrum of encephalitis associated with antibodies against the alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor: case series and review of the literature. JAMA Neurol 2015;72:11631169.CrossRefGoogle ScholarPubMed
Graus, F, Boronat, A, Xifro, X, et al. The expanding clinical profile of anti-AMPA receptor encephalitis. Neurology 2010;74:857859.Google Scholar
Titulaer, MJ, McCracken, L, Gabilondo, I, et al. Treatment and prognostic factors for long-term outcome in patients with anti-NMDA receptor encephalitis: an observational cohort study. Lancet Neurol 2013;12:157165.Google Scholar
Jia, Y, Wang, J, Xue, L, Hou, Y. Limbic encephalitis associated with AMPA receptor and CRMP5 antibodies: a case report and literature review. Brain Behav 2020;10:e01528.Google Scholar
Laurido-Soto, O, Brier, MR, Simon, LE, et al. Patient characteristics and outcome associations in AMPA receptor encephalitis. J Neurol 2019;266:450460.Google Scholar
Carr, I. The Ophelia syndrome: memory loss in Hodgkin’s disease. Lancet 1982;1:844845.Google Scholar
Graus, F, Arino, H, Dalmau, J. Paraneoplastic neurological syndromes in Hodgkin and non-Hodgkin lymphomas. Blood 2014;123:32303238.Google Scholar
Epaulard, O, Courby, S, Pavese, P, et al. Paraneoplastic acute diffuse encephalitis revealing Hodgkin’s disease. Leuk Lymphoma 2004;45:25092512.Google Scholar
Lancaster, E, Martinez-Hernandez, E, Titulaer, MJ, et al. Antibodies to metabotropic glutamate receptor 5 in the Ophelia syndrome. Neurology 2011;77:16981701.Google Scholar
van Sonderen, A, Thijs, RD, Coenders, EC, et al. Anti-LGI1 encephalitis: clinical syndrome and long-term follow-up. Neurology 2016;87:14491456.Google Scholar
Tuzun, E, Rossi, JE, Karner, SF, Centurion, AF, Dalmau, J. Adenylate kinase 5 autoimmunity in treatment refractory limbic encephalitis. J Neuroimmunol 2007;186:177180.Google Scholar
Do, LD, Chanson, E, Desestret, V, et al. Characteristics in limbic encephalitis with anti-adenylate kinase 5 autoantibodies. Neurology 2017;88:514524.Google Scholar
Bien, CI, Nehls, F, Kollmar, R, et al. Identification of adenylate kinase 5 antibodies during routine diagnostics in a tissue-based assay: three new cases and a review of the literature. J Neuroimmunol 2019;334:576975.CrossRefGoogle Scholar
Giometto, B, Nicolao, P, Macucci, M, et al. Temporal-lobe epilepsy associated with glutamic-acid-decarboxylase autoantibodies. Lancet 1998;352:457.Google Scholar
Peltola, J, Kulmala, P, Isojarvi, J, et al. Autoantibodies to glutamic acid decarboxylase in patients with therapy-resistant epilepsy. Neurology 2000;55:4650.Google Scholar
Mata, S, Muscas, GC, Naldi, I, et al. Non-paraneoplastic limbic encephalitis associated with anti-glutamic acid decarboxylase antibodies. J Neuroimmunol 2008;199:155159.Google Scholar
Malter, MP, Helmstaedter, C, Urbach, H, Vincent, A, Bien, CG. Antibodies to glutamic acid decarboxylase define a form of limbic encephalitis. Ann Neurol 2010;67:470478.Google Scholar
Finelli, PF. Autoimmune limbic encephalitis with GAD antibodies. Neurohospitalist 2011;1:178181.Google Scholar
Markakis, I, Alexopoulos, H, Poulopoulou, C, et al. Immunotherapy-responsive limbic encephalitis with antibodies to glutamic acid decarboxylase. J Neurol Sci 2014;343:192194.Google Scholar
Lopez-Sublet, M, Bihan, H, Reach, G, et al. Limbic encephalitis and type 1 diabetes with glutamic acid decarboxylase 65 (GAD65) autoimmunity: improvement with high-dose intravenous immunoglobulin therapy. Diabetes Metab 2012;38:273275.Google Scholar
Blanc, F, Ruppert, E, Kleitz, C, et al. Acute limbic encephalitis and glutamic acid decarboxylase antibodies: a reality? J Neurol Sci 2009;287:6971.Google Scholar
Van Ael, Y, Amir, R, Cras, P. Anti-GAD antibodies, a rare cause of limbic encephalitis: a case report. Acta Neurol Belg 2016;116:105107.Google Scholar
Mishra, N, Rodan, LH, Nita, DA, et al. Anti-glutamic acid decarboxylase antibody associated limbic encephalitis in a child: expanding the spectrum of pediatric inflammatory brain diseases. J Child Neurol 2014;29:677683.Google Scholar
Arcani, R, Jean, E, Pozzo Di Borgo, J, et al. Anti-glutamic acid decarboxylase antibody paraneoplastic limbic encephalitis associated with acute myeloid leukemia. Clin Neurol Neurosurg 2020;189:105618.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 2019;267:10231025.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
Ng, AS, Kramer, J, Centurion, A, et al. Clinico-pathological correlation in adenylate kinase 5 autoimmune limbic encephalitis. J Neuroimmunol 2015;287:3135.Google Scholar
Golombeck, KS, Bonte, K, Monig, C, et al. Evidence of a pathogenic role for CD8(+) T cells in anti-GABAB receptor limbic encephalitis. Neurol Neuroimmunol Neuroinflamm 2016;3:e232.Google Scholar
Kortvelyessy, P, Bauer, J, Stoppel, CM, et al. Complement-associated neuronal loss in a patient with CASPR2 antibody-associated encephalitis. Neurol Neuroimmunol Neuroinflamm 2015;2:e75.Google Scholar
Schultze-Amberger, J, Pehl, D, Stenzel, W. LGI-1-positive limbic encephalitis: a clinicopathological study. J Neurol 2012;259:24782480.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
Lee, WJ, Lee, ST, Byun, JI, et al. Rituximab treatment for autoimmune limbic encephalitis in an institutional cohort. Neurology 2016;86:16831691.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
Lee, WJ, Lee, ST, Moon, J, et al. Tocilizumab in autoimmune encephalitis refractory to rituximab: an institutional cohort study. Neurotherapeutics 2016;13:824832.Google Scholar

References

Vincent, A, Roberts, M, Willison, H, Lang, B, Newsom-Davis, J. Autoantibodies, neurotoxins and the nervous system. J Physiol Paris 1995;89:129136.Google Scholar
Motomura, M, Johnston, I, Lang, B, Vincent, A, Newsom-Davis, J. An improved diagnostic assay for Lambert–Eaton myasthenic syndrome. J Neurol Neurosurg Psychiatry 1995;58:8587.Google Scholar
Vincent, A, Newsom Davis, J. Anti-acetylcholine receptor antibodies. J Neurol Neurosurg Psychiatry 1980;43:590600.Google Scholar
Shillito, P, Molenaar, PC, Vincent, A, et al. Acquired neuromyotonia: evidence for autoantibodies directed against K+ channels of peripheral nerves. Ann Neurol 1995;38:714722.Google Scholar
Graus, F, Saiz, A, Dalmau, J. Antibodies and neuronal autoimmune disorders of the CNS. J Neurol 2009;257:509517Google 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
Ances, BM, Vitaliani, R, Taylor, RA, et al. Treatment-responsive limbic encephalitis identified by neuropil antibodies: MRI and PET correlates. Brain 2005;128:17641777.CrossRefGoogle ScholarPubMed
Dalmau, JG, Christian, G, Graus, F. Autoantibodies to synaptic receptors and neuronal cell surface proteins in autoimmune diseases of the central nervous system. Physiol Rev 2017;97:839887.Google Scholar
Jarius, S, Wildemann, B. ‘Medusa head ataxia’: the expanding spectrum of Purkinje cell antibodies in autoimmune cerebellar ataxia. Part 2: anti-PKC-gamma, anti-GluR-delta2, anti-Ca/ARHGAP26 and anti-VGCC. J Neuroinflammation 2015;12:167.Google Scholar
Jarius, S, Wildemann, B. ‘Medusa head ataxia’: the expanding spectrum of Purkinje cell antibodies in autoimmune cerebellar ataxia. Part 3: anti-Yo/CDR2, anti-Nb/AP3B2, PCA-2, anti-Tr/DNER, other antibodies, diagnostic pitfalls, summary and outlook. J Neuroinflammation 2015;12:168.Google Scholar
Jarius, S, Wildemann, B. ‘Medusa-head ataxia’: the expanding spectrum of Purkinje cell antibodies in autoimmune cerebellar ataxia. Part 1: anti-mGluR1, anti-Homer-3, anti-Sj/ITPR1 and anti-CARP VIII. J Neuroinflammation 2015;12:166.Google Scholar
Newsom-Davis, J, Mills, KR. Immunological associations of acquired neuromyotonia (Isaac’s syndrome): report of five cases and literature review. Brain 1993;116:453469.Google Scholar
Sinha, S, Newsom-Davis, J, Mills, K, et al. Autoimmune aetiology for acquired neuromyotonia (Isaacs’ syndrome). Lancet 1991;338:7577.Google Scholar
Newsom-Davis, J, Buckley, C, Clover, L, et al. Autoimmune disorders of neuronal potassium channels. Ann N Y Acad Sci 2003;998:202210.Google Scholar
Hart, IK, Maddison, P, Newsom-Davis, J, Vincent, A, Mills, KR. Phenotypic variants of autoimmune peripheral nerve hyperexcitability. Brain 2002;125:18871895.Google Scholar
Liguori, R, Vincent, A, Clover, L, et al. Morvan’s syndrome: peripheral and central nervous system and cardiac involvement with antibodies to voltage-gated potassium channels. Brain 2001;124:24172426.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
Buckley, C, Oger, J, Clover, L, et al. Potassium channel antibodies in two patients with reversible limbic encephalitis. Ann Neurol 2001;50:7378.Google Scholar
Vincent, A, Buckley, C, Schott, JM, et al. Potassium channel antibody-associated encephalopathy: a potentially immunotherapy-responsive form of limbic encephalitis. Brain 2004;127:701712.Google Scholar
Thieben, MJ, Lennon, VA, Boeve, BF, et al. Potentially reversible autoimmune limbic encephalitis with neuronal potassium channel antibody. Neurology 2004;62:11771182.Google Scholar
McKnight, K, Jiang, Y, Hart, Y, et al. Serum antibodies in epilepsy and seizure-associated disorders. Neurology 2005;65:17301736.Google Scholar
Majoie, HJ, de Baets, M, Renier, W, Lang, B, Vincent, A. Antibodies to voltage-gated potassium and calcium channels in epilepsy. Epilepsy Res 2006;71:135141.Google Scholar
Tan, KM, Lennon, VA, Klein, CJ, Boeve, BF, Pittock, SJ. Clinical spectrum of voltage-gated potassium channel autoimmunity. Neurology 2008;70:18831890.Google Scholar
Dhamija, R, Renaud, DL, Pittock, SJ, et al. Neuronal voltage-gated potassium channel complex autoimmunity in children. Pediatr Neurol 2011;44:275281.Google Scholar
Vincent, A, Buckley, C, Lang, B, Irani, S. Clinical spectrum of voltage-gated potassium channel autoimmunity. Neurology 2009;72:99.Google Scholar
Klein, CJ, Lennon, VA, Aston, PA, McKeon, A, Pittock, SJ. Chronic pain as a manifestation of potassium channel-complex autoimmunity. Neurology 2012;79:11361144.Google Scholar
Lai, M, Huijbers, MG, Lancaster, E, et al. Investigation of LGI1 as the antigen in limbic encephalitis previously attributed to potassium channels: a case series. Lancet Neurol 2010;9:776785.Google Scholar
Irani, SR, Alexander, S, Waters, P, et al. Antibodies to Kv1 potassium channel-complex proteins leucine-rich, glioma inactivated 1 protein and contactin-associated protein-2 in limbic encephalitis, Morvan’s syndrome and acquired neuromyotonia. Brain 2010;133:27342748.Google Scholar
Rosenfeld, MR, Titulaer, MJ, Dalmau, J. Paraneoplastic syndromes and autoimmune encephalitis, five new things. Neurol Clin Pract 2012;1:215222.Google Scholar
Lang, B, Makuch, M, Moloney, T, et al. Intracellular and non-neuronal targets of voltage-gated potassium channel complex antibodies. J Neurol Neurosurg Psychiatry 2017;88:353361.Google Scholar
Paterson, RW, Zandi, MS, Armstrong, R, Vincent, A, Schott, JM. Clinical relevance of positive voltage-gated potassium channel (VGKC)-complex antibodies: experience from a tertiary referral centre. J Neurol Neurosurg Psychiatry 2014;85:625630.Google Scholar
Hacohen, Y, Singh, R, Rossi, M, et al. Clinical relevance of voltage-gated potassium channel-complex antibodies in children. Neurology 2015;85:967975.Google Scholar
van Sonderen, A, Schreurs, MW, de Bruijn, MA, et al. The relevance of VGKC positivity in the absence of LGI1 and Caspr2 antibodies. Neurology 2016;86:16921699.Google Scholar
Graus, F, Gorman, MP. Voltage-gated potassium channel antibodies: game over. Neurology 2016;86:16571658.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
Gadoth, A, Pittock, SJ, Dubey, D, et al. Expanded phenotypes and outcomes among 256 LGI1/CASPR2-IgG-positive patients. Ann Neurol 2017;82:7992.Google Scholar
Dubey, D, Alqallaf, A, Hays, R, et al. Neurological autoantibody prevalence in epilepsy of unknown etiology. JAMA Neurol 2017;74:397402.Google Scholar
Elisak, M, Krysl, D, Hanzalova, J, et al. The prevalence of neural antibodies in temporal lobe epilepsy and the clinical characteristics of seropositive patients. Seizure 2018;63:16.Google Scholar
Brenner, T, Sills, GJ, Hart, Y, et al. Prevalence of neurologic autoantibodies in cohorts of patients with new and established epilepsy. Epilepsia 2013;54:10281035.Google Scholar
Vincent, A, Pettingill, P, Pettingill, R, et al. Association of leucine-rich glioma inactivated protein 1, contactin-associated protein 2, and contactin 2 antibodies with clinical features and patient-reported pain in acquired neuromyotonia. JAMA Neurol 2018;75:15191527.Google Scholar
Klein, CJ, Lennon, VA, Aston, PA, et al. Insights from LGI1 and CASPR2 potassium channel complex autoantibody subtyping. JAMA Neurol 2013;70:229234.Google Scholar
Binks, SNM, Klein, CJ, Waters, P, Pittock, SJ, Irani, SR. LGI1, CASPR2 and related antibodies: a molecular evolution of the phenotypes. J Neurol Neurosurg Psychiatry 2018;89:526534.Google Scholar
Ohkawa, T, Fukata, Y, Yamasaki, M, et al. Autoantibodies to epilepsy-related LGI1 in limbic encephalitis neutralize LGI1–ADAM22 interaction and reduce synaptic AMPA receptors. J Neurosci 2013;33:1816118174.Google Scholar
Petit-Pedrol, M, Sell, J, Planaguma, J, et al. LGI1 antibodies alter Kv1.1 and AMPA receptors changing synaptic excitability, plasticity and memory. Brain 2018;141:31443159.Google Scholar
van Sonderen, A, Thijs, RD, Coenders, EC, et al. Anti-LGI1 encephalitis: clinical syndrome and long-term follow-up. Neurology 2016;87:14491456.Google Scholar
Celicanin, M, Blaabjerg, M, Maersk-Moller, C, et al. Autoimmune encephalitis associated with voltage-gated potassium channels-complex and leucine-rich glioma-inactivated 1 antibodies: a national cohort study. Eur J Neurol 2017;24:9991005.Google Scholar
Binks, S, Varley, J, Lee, W, et al. Distinct HLA associations of LGI1 and CASPR2-antibody diseases. Brain 2018;141:22632271.Google Scholar
van Sonderen, A, Roelen, DL, Stoop, JA, et al. Anti-LGI1 encephalitis is strongly associated with HLA-DR7 and HLA-DRB4. Ann Neurol 2017;81:193198.Google Scholar
Kim, TJ, Lee, ST, Moon, J, et al. Anti-LGI1 encephalitis is associated with unique HLA subtypes. Ann Neurol 2017;81:183192.Google Scholar
Mueller, SH, Farber, A, Pruss, H, et al. Genetic predisposition in anti-LGI1 and anti-NMDA receptor encephalitis. Ann Neurol 2018;83:863869.Google Scholar
Bien, CG, Vincent, A, Barnett, MH, et al. Immunopathology of autoantibody-associated encephalitides: clues for pathogenesis. Brain 2012;135:16221638.Google Scholar
Arino, H, Armangue, T, Petit-Pedrol, M, et al. Anti-LGI1-associated cognitive impairment: presentation and long-term outcome. Neurology 2016;87:759765.Google Scholar
Park, DC, Murman, DL, Perry, KD, Bruch, LA. An autopsy case of limbic encephalitis with voltage-gated potassium channel antibodies. Eur J Neurol 2007;14:e56.Google Scholar
Dunstan, EJ, Winer, JB. Autoimmune limbic encephalitis causing fits, rapidly progressive confusion and hyponatraemia. Age Ageing 2006;35:536537.Google Scholar
Khan, NL, Jeffree, MA, Good, C, Macleod, W, Al-Sarraj, S. Histopathology of VGKC antibody-associated limbic encephalitis. Neurology 2009;72:17031705.Google Scholar
Schultze-Amberger, J, Pehl, D, Stenzel, W. LGI-1-positive limbic encephalitis: a clinicopathological study. J Neurol 2012;259:24782480.Google Scholar
Körtvelyessy, P, Bauer, J, Stoppel, CM, et al. Complement-associated neuronal loss in a patient with CASPR2 antibody-associated encephalitis. Neurol Neuroimmunol Neuroinflamm 2015;2:e75.Google Scholar
Sundal, C, Vedeler, C, Miletic, H, Andersen, O. Morvan syndrome with Caspr2 antibodies: clinical and autopsy report. J Neurol Sci 2017;372:453455.Google Scholar
Maat, P, de Beukelaar, JW, Jansen, C, et al. Pathologically confirmed autoimmune encephalitis in suspected Creutzfeldt-Jakob disease. Neurol Neuroimmunol Neuroinflamm 2015;2:e178.Google Scholar
Thompson, J, Bi, M, Murchison, AG, et al. The importance of early immunotherapy in patients with faciobrachial dystonic seizures. Brain 2018;141:348356.Google Scholar
Irani, SR, Michell, AW, Lang, B, et al. Faciobrachial dystonic seizures precede LGI1 antibody limbic encephalitis. Ann Neurol 2011;69:892900.Google Scholar
Navarro, V, Kas, A, Apartis, E, et al. Motor cortex and hippocampus are the two main cortical targets in LGI1-antibody encephalitis. Brain 2016;139:10791093.Google Scholar
Irani, SR, Stagg, CJ, Schott, JM, et al. Faciobrachial dystonic seizures: the influence of immunotherapy on seizure control and prevention of cognitive impairment in a broadening phenotype. Brain 2013;136:31513162.Google Scholar
Rachdi, A, Dupouy, J, Benaiteau, M, et al. Leucine-rich glioma-inactivated 1 encephalitis: broadening the sphere. Tremor Other Hyperkinet Mov (N Y) 2019;9.Google Scholar
Loddenkemper, T, Kellinghaus, C, Gandjour, J, et al. Localising and lateralising value of ictal piloerection. J Neurol Neurosurg Psychiatry 2004;75:879883.Google Scholar
Rocamora, R, Becerra, JL, Fossas, P, et al. Pilomotor seizures: an autonomic semiology of limbic encephalitis? Seizure 2014;23:670673.Google Scholar
Wieser, S, Kelemen, A, Barsi, P, et al. Pilomotor seizures and status in non-paraneoplastic limbic encephalitis. Epileptic Disord 2005;7:205211.Google ScholarPubMed
Naasan, G, Irani, SR, Bettcher, BM, Geschwind, MD, Gelfand, JM. Episodic bradycardia as neurocardiac prodrome to voltage-gated potassium channel complex/leucine-rich, glioma inactivated 1 antibody encephalitis. JAMA Neurol 2014;71:13001304.Google Scholar
Nilsson, AC, Blaabjerg, M. More evidence of a neurocardiac prodrome in anti-LGI1 encephalitis. J Neurol Sci 2015;357:310311.Google Scholar
Britton, JW, Ghearing, GR, Benarroch, EE, Cascino, GD. The ictal bradycardia syndrome: localization and lateralization. Epilepsia 2006;47:737744.Google Scholar
Tofaris, GK, Irani, SR, Cheeran, BJ, et al. Immunotherapy-responsive chorea as the presenting feature of LGI1-antibody encephalitis. Neurology 2012;79:195196.Google Scholar
Ramdhani, RA, Frucht, SJ. Isolated chorea associated with LGI1 antibody. Tremor Other Hyperkinet Mov (N Y) 2014;4.Google Scholar
Iranzo, A, Graus, F, Clover, L, et al. Rapid eye movement sleep behavior disorder and potassium channel antibody-associated limbic encephalitis. Ann Neurol 2006;59:178181.Google Scholar
Cornelius, JR, Pittock, SJ, McKeon, A, et al. Sleep manifestations of voltage-gated potassium channel complex autoimmunity. Arch Neurol 2011;68:733738.Google Scholar
Butler, CR, Miller, TD, Kaur, MS, et al. Persistent anterograde amnesia following limbic encephalitis associated with antibodies to the voltage-gated potassium channel complex. J Neurol Neurosurg Psychiatry 2014;85:387391.Google Scholar
Finke, C, Pruss, H, Heine, J, et al. Evaluation of cognitive deficits and structural hippocampal damage in encephalitis with leucine-rich, glioma-inactivated 1 antibodies. JAMA Neurol 2017;74:5059.Google Scholar
Bettcher, BM, Gelfand, JM, Irani, SR, et al. More than memory impairment in voltage-gated potassium channel complex encephalopathy. Eur J Neurol 2014;21:13011310.Google Scholar
Jang, Y, Lee, ST, Lim, JA, et al. Psychiatric symptoms delay the diagnosis of anti-LGI1 encephalitis. J Neuroimmunol 2018;317:814.Google Scholar
Balint, B, Vincent, A, Meinck, HM, Irani, SR, Bhatia, KP. Movement disorders with neuronal antibodies: syndromic approach, genetic parallels and pathophysiology. Brain 2018;141:1336.Google Scholar
Geschwind, MD, Tan, KM, Lennon, VA, et al. Voltage-gated potassium channel autoimmunity mimicking Creutzfeldt-Jakob disease. Arch Neurol 2008;65:13411346.Google Scholar
McQuillan, RF, Bargman, JM. Hyponatraemia caused by LGI1-associated limbic encephalitis. NDT plus 2011;4:424426.Google Scholar
Head, K, Gong, S, Joseph, S, et al. Defining the expression pattern of the LGI1 gene in BAC transgenic mice. Mammalian Genome 2007;18:328337.Google Scholar
Gadoth, A, Zekeridou, A, Klein, CJ, et al. Elevated LGI1-IgG CSF index predicts worse neurological outcome. Ann Clin Transl Neurol 2018;5:646650.Google Scholar
Escudero, D, Guasp, M, Arino, H, et al. Antibody-associated CNS syndromes without signs of inflammation in the elderly. Neurology 2017;89:14711475.Google Scholar
Flanagan, EP, Kotsenas, AL, Britton, JW, et al. Basal ganglia T1 hyperintensity in LGI1-autoantibody faciobrachial dystonic seizures. Neurol Neuroimmunol Neuroinflamm 2015;2:e161.Google Scholar
Grativvol, RS, Simabukuro, MM, Spera, RR, et al. Imaging findings in faciobrachial dystonic seizures associated with LGI-1 antibodies. Arq Neuropsiquiatr 2016;74:947.CrossRefGoogle ScholarPubMed
Lopez Chiriboga, AS, Siegel, JL, Tatum, WO, Shih, JJ, Flanagan, EP. Striking basal ganglia imaging abnormalities in LGI1 ab faciobrachial dystonic seizures. Neurol Neuroimmunol Neuroinflamm 2017;4:e336.Google Scholar
Heine, J, Pruss, H, Bartsch, T, et al. Imaging of autoimmune encephalitis: relevance for clinical practice and hippocampal function. Neuroscience 2015;309:6883.Google Scholar
Moloney, P, Boylan, R, Elamin, M, et al. Semi-quantitative analysis of cerebral FDG-PET reveals striatal hypermetabolism and normal cortical metabolism in a case of VGKCC limbic encephalitis. Neuroradiol J 2017;30:160163.Google Scholar
Rey, C, Koric, L, Guedj, E, et al. Striatal hypermetabolism in limbic encephalitis. J Neurol 2012;259:11061110.Google Scholar
Maeder-Ingvar, M, Prior, JO, Irani, SR, et al. FDG-PET hyperactivity in basal ganglia correlating with clinical course in anti-NDMA-R antibodies encephalitis. J Neurol Neurosurg Psychiatry 2011;82:235236.Google Scholar
Morbelli, S, Arbizu, J, Booij, J, et al. The need of standardization and of large clinical studies in an emerging indication of [(18)F]FDG PET: the autoimmune encephalitis. Eur J Nucl Med Molec Imag 2017;44:353357.Google Scholar
Fauser, S, Talazko, J, Wagner, K, et al. FDG-PET and MRI in potassium channel antibody-associated non-paraneoplastic limbic encephalitis: correlation with clinical course and neuropsychology. Acta Neurol Scand 2005;111:338343.Google Scholar
Ances, BM, Vitaliani, R, Taylor, RA, et al. Treatment-responsive limbic encephalitis identified by neuropil antibodies: MRI and PET correlates. Brain 2005;128:17641777.Google Scholar
Kamaleshwaran, KK, Iyer, RS, Antony, J, Radhakrishnan, EK, Shinto, A. 18F-FDG PET/CT findings in voltage-gated potassium channel limbic encephalitis. Clin Nucl Med 2013;38:392394.Google Scholar
Chatzikonstantinou, A, Szabo, K, Ottomeyer, C, Kern, R, Hennerici, MG. Successive affection of bilateral temporomesial structures in a case of non-paraneoplastic limbic encephalitis demonstrated by serial MRI and FDG-PET. J Neurol 2009;256:17531755.Google Scholar
Baumgartner, A, Rauer, S, Mader, I, Meyer, PT. Cerebral FDG-PET and MRI findings in autoimmune limbic encephalitis: correlation with autoantibody types. J Neurol 2013;260:27442753.Google Scholar
Kunze, A, Drescher, R, Kaiser, K, et al. Serial FDG PET/CT in autoimmune encephalitis with faciobrachial dystonic seizures. Clin Nucl Med 2014;39:e436438.Google Scholar
Park, S, Choi, H, Cheon, GJ, Wook Kang, K, Lee, DS. 18F-FDG PET/CT in anti-LGI1 encephalitis: initial and follow-up findings. CLin Nucl Med 2015;40:156158.Google Scholar
Serrano Vicente, J, Garcia Bernardo, L, Duran Barquero, C, et al. Brain metabolic changes in limbic encephalitis evidenced by 18FDG PET: correlation with symptomatology. Revista espanola de medicina nuclear e imagen molecular 2013;32:201202.Google Scholar
Newey, CR, Sarwal, A, Hantus, S. [(18)F]-fluoro-deoxy-glucose positron emission tomography scan should be obtained early in cases of autoimmune encephalitis. Autoimmune Dis 2016;2016:9450452.Google Scholar
Chen, C, Wang, X, Zhang, C, et al. Seizure semiology in leucine-rich glioma-inactivated protein 1 antibody-associated limbic encephalitis. Epilepsy Behav 2017;77:9095.Google Scholar
Dodich, A, Cerami, C, Iannaccone, S, et al. Neuropsychological and FDG-PET profiles in VGKC autoimmune limbic encephalitis. Brain Cogn 2016;108:8187.Google Scholar
Shin, YW, Lee, ST, Shin, JW, et al. VGKC-complex/LGI1-antibody encephalitis: clinical manifestations and response to immunotherapy. J Neuroimmunol 2013;265:7581.Google Scholar
Tripathi, M, Tripathi, M, Roy, SG, et al. Metabolic topography of autoimmune non-paraneoplastic encephalitis. Neuroradiology 2018;60:189198.Google Scholar
Jang, Y, Lee, ST, Bae, JY, et al. LGI1 expression and human brain asymmetry: insights from patients with LGI1-antibody encephalitis. J Neuroinflammation 2018;15:279.Google Scholar
d’Orsi, G, Martino, T, Lalla, A, et al. Faciobrachial dystonic seizures expressed as epileptic spasms, followed by focal seizures in anti-LGI1 encephalitis: a video-polygraphic study. Epileptic Disord 2018;20:525529.Google Scholar
Shan, W, Liu, X, Wang, Q. Teaching neuroimages: (18)F-FDG-PET/SPM analysis in 3 different stages from a patient with LGI-1 autoimmune encephalitis. Neurology 2019;93:e1917e1918.Google Scholar
Zuliani, L, Nosadini, M, Gastaldi, M, et al. Management of antibody-mediated autoimmune encephalitis in adults and children: literature review and consensus-based practical recommendations. Neurol Sci 2019;40:20172030.Google Scholar
Irani, SR, Gelfand, JM, Bettcher, BM, Singhal, NS, Geschwind, MD. Effect of rituximab in patients with leucine-rich, glioma-inactivated 1 antibody-associated encephalopathy. JAMA Neurol 2014;71:896900.Google Scholar
Brown, JW, Martin, PJ, Thorpe, JW, et al. Long-term remission with rituximab in refractory leucine-rich glioma inactivated 1 antibody encephalitis. J Neuroimmunol 2014;271:6668.Google Scholar
Abboud, H, Probasco, JC, Irani, S, et al. Autoimmune encephalitis: proposed best practice recommendations for diagnosis and acute management. J Neurol Neurosurg Psychiatry 2021;92:757768.Google Scholar
Sola-Valls, N, Arino, H, Escudero, D, et al. Telemedicine assessment of long-term cognitive and functional status in anti-leucine-rich, glioma-inactivated 1 encephalitis. Neurol Neuroimmunol Neuroinflamm 2020;7:e652.Google Scholar
Miller, TD, Chong, TT, Aimola Davies, AM, et al. Human hippocampal CA3 damage disrupts both recent and remote episodic memories. Elife 2020;9:e41836.Google Scholar
Miller, TD, Chong, TT, Aimola Davies, AM, et al. Focal CA3 hippocampal subfield atrophy following LGI1 VGKC-complex antibody limbic encephalitis. Brain 2017;140:12121219.Google Scholar
Xu, M, Bennett, DLH, Querol, LA, et al. Pain and the immune system: emerging concepts of IgG-mediated autoimmune pain and immunotherapies. J Neurol Neurosurg Psychiatry 2020;91:177188.Google Scholar
Laurencin, C, Andre-Obadia, N, Camdessanche, JP, et al. Peripheral small fiber dysfunction and neuropathic pain in patients with Morvan syndrome. Neurology 2015;85:20762078.Google Scholar
Lancaster, E, Huijbers, MG, Bar, V, et al. Investigations of caspr2, an autoantigen of encephalitis and neuromyotonia. Ann Neurol 2011;69:303311.Google Scholar
Giannoccaro, MP, Menassa, DA, Jacobson, L, et al. Behaviour and neuropathology in mice injected with human contactin-associated protein 2 antibodies. Brain 2019;142:20002012.Google Scholar
Pinatel, D, Hivert, B, Saint-Martin, M, et al. The Kv1-associated molecules TAG-1 and Caspr2 are selectively targeted to the axon initial segment in hippocampal neurons. J Cell Sci 2017;130:22092220.Google Scholar
Boyko, M, Au, KLK, Casault, C, de Robles, P, Pfeffer, G. Systematic review of the clinical spectrum of CASPR2 antibody syndrome. J Neurol 2020;267:11371146.Google Scholar
Joubert, B, Gobert, F, Thomas, L, et al. Autoimmune episodic ataxia in patients with anti-CASPR2 antibody-associated encephalitis. Neurol Neuroimmunol Neuroinflamm 2017;4:e371.Google Scholar
Govert, F, Witt, K, Erro, R, et al. Orthostatic myoclonus associated with Caspr2 antibodies. Neurology 2016;86:13531355.Google Scholar
van Sonderen, A, Arino, H, Petit-Pedrol, M, et al. The clinical spectrum of Caspr2 antibody-associated disease. Neurology 2016;87:521528.Google Scholar
Joubert, B, Saint-Martin, M, Noraz, N, et al. Characterization of a subtype of autoimmune encephalitis with anti-contactin-associated protein-like 2 antibodies in the cerebrospinal fluid, prominent limbic symptoms, and seizures. JAMA Neurol 2016;73:11151124.Google Scholar
Becker, EB, Zuliani, L, Pettingill, R, et al. Contactin-associated protein-2 antibodies in non-paraneoplastic cerebellar ataxia. J Neurol Neurosurg Psychiatry 2012;83:437440.Google Scholar
Klockgether, T. Sporadic ataxia with adult onset: classification and diagnostic criteria. Lancet Neurol 2010;9:94104.Google Scholar
Fabbri, M, Giannoccaro, MP, Leta, C, et al. Non-paraneoplastic ataxia in a patient with contactin-associated protein-2 antibodies and benign course. Eur J Neurol 2015;22:e6263.Google Scholar
Melzer, N, Golombeck, KS, Gross, CC, Meuth, SG, Wiendl, H. Cytotoxic CD8+ T cells and CD138+ plasma cells prevail in cerebrospinal fluid in non-paraneoplastic cerebellar ataxia with contactin-associated protein-2 antibodies. J Neuroinflammation 2012;9:160.Google Scholar
Muñiz-Castrillo, S, Joubert, B, Elsensohn, MH, et al. Anti-CASPR2 clinical phenotypes correlate with HLA and immunological features. J Neurol Neurosurg Psychiatry 2020;91:10761084.Google Scholar
Malter, MP, Frisch, C, Schoene-Bake, JC, et al. Outcome of limbic encephalitis with VGKC-complex antibodies: relation to antigenic specificity. J Neurol 2014;261:16951705.Google Scholar
Titulaer, MJ, McCracken, L, Gabilondo, I, et al. Treatment and prognostic factors for long-term outcome in patients with anti-NMDA receptor encephalitis: an observational cohort study. Lancet Neurol 2013;12:157165.Google Scholar
Scheibe, F, Ostendorf, L, Reincke, SM, et al. Daratumumab treatment for therapy-refractory anti-CASPR2 encephalitis. J Neurol 2020;267:317323.Google Scholar
Walusinski, O, Honnorat, J. Augustin Morvan (1819–1897), a little-known rural physician and neurologist. Rev Neurol (Paris) 2013;169:28.Google Scholar
Serratrice, G, Azulay, JP. [What is left of Morvan’s fibrillary chorea?]. Rev Neurol (Paris) 1994;150:257265.Google ScholarPubMed
Barber, PA, Anderson, NE, Vincent, A. Morvan’s syndrome associated with voltage-gated K+ channel antibodies. Neurology 2000;54:771772.Google Scholar
Cottrell, DA, Blackmore, KJ, Fawcett, PR, et al. Sub-acute presentation of Morvan’s syndrome after thymectomy. J Neurol Neurosurg Psychiatry 2004;75:15041505.Google Scholar
Lee, EK, Maselli, RA, Ellis, WG, Agius, MA. Morvan’s fibrillary chorea: a paraneoplastic manifestation of thymoma. J Neurol Neurosurg Psychiatry 1998;65:857862.Google Scholar
Josephs, KA, Silber, MH, Fealey, RD, et al. Neurophysiologic studies in Morvan syndrome. J Clin Neurophysiol 2004;21:440445.Google Scholar
Abou-Zeid, E, Boursoulian, LJ, Metzer, WS, Gundogdu, B. Morvan syndrome: a case report and review of the literature. J Clin Neuromuscul Dis 2012;13:214227.Google Scholar
Lugaresi, E, Provini, F. Agrypnia excitata: clinical features and pathophysiological implications. Sleep Med Rev 2001;5:313322.Google Scholar
Provini, F, Marconi, S, Amadori, M, et al. Morvan chorea and agrypnia excitata: when video-polysomnographic recording guides the diagnosis. Sleep Med 2011;12:10411043.Google Scholar
Lugaresi, E, Provini, F, Cortelli, P. Agrypnia excitata. Sleep Med 2011;12(Suppl. 2):S310.Google Scholar
Madrid, A, Gil-Peralta, A, Gil-Neciga, E, Gonzalez, JR, Jarrin, S. Morvan’s fibrillary chorea: remission after plasmapheresis. J Neurol 1996;243:350353.Google Scholar
van den Berg, JS, Van Engelen, BG, Boerman, RH, De Baets, MH. Acquired neuromyotonia: superiority of plasma exchange over high-dose intravenous human immunoglobulin. J Neurol 1999;246:623625.Google Scholar
Ishii, A, Hayashi, A, Ohkoshi, N, et al. Clinical evaluation of plasma exchange and high dose intravenous immunoglobulin in a patient with Isaacs’ syndrome [see comments]. J Neurol Neurosurg Psychiatry 1994;57:840842.Google Scholar
Fischer-Perroudon, C, Trillet, M, Mouret, J, et al. [Polygraphic and metabolic studies of persistent insomnia with hallucinations. Apropos of an antomo-clinical study of a case of Morvan’s fibrillar chorea]. Rev Neurol (Paris) 1974;130:111125.Google Scholar
Patterson, KR, Dalmau, J, Lancaster, E. Mechanisms of Caspr2 antibodies in autoimmune encephalitis and neuromyotonia. Ann Neurol 2018;83:4051.Google Scholar
Ong, E, Viaccoz, A, Ducray, F, et al. Dramatic improvement after rituximab in a patient with paraneoplastic treatment-refractory Morvan syndrome associated with anti-CASPR2 antibodies. Eur J Neurol 2013;20:e9697.Google Scholar
Lopez-Chiriboga, AS, Klein, C, Zekeridou, A, et al. LGI1 and CASPR2 neurological autoimmunity in children. Ann Neurol 2018;84:473480.Google Scholar
Zhang, J, Ji, T, Chen, Q, et al. Pediatric autoimmune encephalitis: case series from two Chinese tertiary pediatric neurology centers. Front Neurol 2019;10:906.Google Scholar
Nosadini, M, Toldo, I, Tascini, B, et al. LGI1 and CASPR2 autoimmunity in children: systematic literature review and report of a young girl with Morvan syndrome. J Neuroimmunol 2019;335:577008.Google Scholar
Wright, S, Geerts, AT, Jol-van der Zijde, CM, et al. Neuronal antibodies in pediatric epilepsy: clinical features and long-term outcomes of a historical cohort not treated with immunotherapy. Epilepsia 2016;57:823831.Google Scholar
Suleiman, J, Wright, S, Gill, D, et al. Autoantibodies to neuronal antigens in children with new-onset seizures classified according to the revised ILAE organization of seizures and epilepsies. Epilepsia 2013;54:20912100.Google Scholar
Garcia-Tarodo, S, Datta, AN, Ramelli, GP, et al. Circulating neural antibodies in unselected children with new-onset seizures. Eur J Paediatr Neurol 2018;22:396403.Google Scholar
Borusiak, P, Bettendorf, U, Wiegand, G, et al. Autoantibodies to neuronal antigens in children with focal epilepsy and no prima facie signs of encephalitis. Eur J Paediatr Neurol 2016;20:573579.Google Scholar
Tekturk, P, Baykan, B, Erdag, E, et al. Investigation of neuronal auto-antibodies in children diagnosed with epileptic encephalopathy of unknown cause. Brain Dev 2018;40:909917.Google Scholar
Syrbe, S, Stettner, GM, Bally, J, et al. CASPR2 autoimmunity in children expanding to mild encephalopathy with hypertension. Neurology 2020;94:e2290e2301.Google Scholar

References

Buckley, C, Oger, J, Clover, L, et al. Potassium channel antibodies in two patients with reversible limbic encephalitis. Ann Neurol 2001;50:7378.Google Scholar
Lai, M, Huijbers, MG, Lancaster, E, et al. Investigation of LGI1 as the antigen in limbic encephalitis previously attributed to potassium channels: a case series. Lancet Neurol 2010;9:776785.Google Scholar
Irani, SR, Alexander, S, Waters, P, et al. Antibodies to Kv1 potassium channel-complex proteins leucine-rich, glioma inactivated 1 protein and contactin-associated protein-2 in limbic encephalitis, Morvan’s syndrome and acquired neuromyotonia. Brain 2010;133:27342748.Google Scholar
Darnell, RB, Posner, JB. Paraneoplastic syndromes involving the nervous system. N Engl J Med 2003;349:15431554.Google Scholar
Bataller, L, Kleopa, KA, Wu, GF, et al. Autoimmune limbic encephalitis in 39 patients: immunophenotypes and outcomes. J Neurol Neurosurg Psychiatry 2007;78:381385.Google Scholar
Ances, BM, Vitaliani, R, Taylor, RA, et al. Treatment-responsive limbic encephalitis identified by neuropil antibodies: MRI and PET correlates. Brain 2005;128:17641777.Google Scholar
Vitaliani, R, Mason, W, Ances, B, et al. Paraneoplastic encephalitis, psychiatric symptoms, and hypoventilation in ovarian teratoma. Ann Neurol 2005;58:594604.Google Scholar
Dalmau, J, Tuzun, E, Wu, HY, et al. Paraneoplastic anti-N-methyl-D-aspartate receptor encephalitis associated with ovarian teratoma. Ann Neurol 2007;61:2536.Google Scholar
Dalmau, J, Gleichman, AJ, Hughes, EG, et al. Anti-NMDA-receptor encephalitis: case series and analysis of the effects of antibodies. Lancet Neurol 2008;7:10911098.Google Scholar
Dalmau, J. NMDA receptor encephalitis and other antibody-mediated disorders of the synapse: the 2016 Cotzias Lecture. Neurology 2016;87:24712482.Google Scholar
DeLong, GR, Bean, SC, Brown, FR, III. Acquired reversible autistic syndrome in acute encephalopathic illness in children. Arch Neurol 1981;38:191194.Google Scholar
Sebire, G, Devictor, D, Huault, G, et al. Coma associated with intense bursts of abnormal movements and long-lasting cognitive disturbances: an acute encephalopathy of obscure origin. J Pediatr 1992;121:845851.Google Scholar
Haimi-Cohen, Y, Soen, G, Amir, J, Kimia, A, Varsano, I. Coma with abnormal movements and prolonged cognitive disturbances: a new subset of acute encephalopathy. Neuropediatrics 1996;27:270272.Google Scholar
Hartley, LM, Ng, SY, Dale, RC, et al. Immune mediated chorea encephalopathy syndrome in childhood. Dev Med Child Neurol 2002;44:273277.Google Scholar
Poloni, C, Korff, CM, Ricotti, V, et al. Severe childhood encephalopathy with dyskinesia and prolonged cognitive disturbances: evidence for anti-N-methyl-d-aspartate receptor encephalitis. Dev Med Child Neurol 2010;52:e7882.CrossRefGoogle ScholarPubMed
Sansing, LH, Tuzun, E, Ko, MW, et al. A patient with encephalitis associated with NMDA receptor antibodies. Nat Clin Pract Neurol 2007;3:291296.Google Scholar
Lejuste, F, Thomas, L, Picard, G, et al. Neuroleptic intolerance in patients with anti-NMDAR encephalitis. Neurol Neuroimmunol Neuroinflamm 2016;3:e280.Google Scholar
Lim, SY, Panikkath, R, Mankongpaisarnrung, C, et al. Anti-N-methyl-D-aspartate receptor encephalitis. Am J Med Sci 2013;345:491493.Google Scholar
Salehi, N, Yuan, AK, Stevens, G, Koshy, R, Klein, WF. A case of severe anti-n-methyl d-aspartate (anti-nmda) receptor encephalitis with refractory autonomic instability and elevated intracranial pressure. Am J Case Rep 2018;19:12161221.Google Scholar
Mammele, S, Thompson, KS, Abe, KK. A rapidly fatal case of anti-NMDA receptor encephalitis due to acute brain edema and herniation. Neurology 2019;92:10141016.Google Scholar
Iizuka, T, Sakai, F, Ide, T, et al. Anti-NMDA receptor encephalitis in Japan: long-term outcome without tumor removal. Neurology 2008;70:504511.Google Scholar
Sebire, G. In search of lost time from ‘Demonic Possession’ to anti-N-methyl-D-aspartate receptor encephalitis. Ann Neurol 2010;67:141142.Google Scholar
Bickerstaff, ER, Cloake, PC. Mesencephalitis and rhombencephalitis. Br Med J 1951;2:7781.Google Scholar
Merwick, A, Dalmau, J, Delanty, N. Insights into antibody-associated encephalitis: Bickerstaff’s 1950’s papers revisted. J Neurol Sci 2013;334:167168.Google Scholar
Odaka, M, Yuki, N, Yamada, M, et al. Bickerstaff’s brainstem encephalitis: clinical features of 62 cases and a subgroup associated with Guillain-Barre syndrome. Brain 2003;126:22792290.Google Scholar
Titulaer, MJ, Hoftberger, R, Iizuka, T, et al. Overlapping demyelinating syndromes and anti-N-methyl-D-aspartate receptor encephalitis. Ann Neurol 2014;75:411428.Google Scholar
Hacohen, Y, Absoud, M, Hemingway, C, et al. NMDA receptor antibodies associated with distinct white matter syndromes. Neurol Neuroimmunol Neuroinflamm 2014;1:e2.Google Scholar
Tenyi, D, Szucs, A, Kovacs, N, Illes, Z, Janszky, J. Paraneoplastic anti-NMDA receptor encephalitis in 1830? Neurol Neuroimmunol Neuroinflamm 2020;7:e887.Google Scholar
van Sonderen, A, Petit-Pedrol, M, Dalmau, J, Titulaer, MJ. The value of LGI1, Caspr2 and voltage-gated potassium channel antibodies in encephalitis. Nat Rev Neurol 2017;13:290301.Google Scholar
Granerod, J, Ambrose, HE, Davies, NW, et al. Causes of encephalitis and differences in their clinical presentations in England: a multicentre, population-based prospective study. Lancet Infect Dis 2010;10:835844.Google Scholar
Gable, MS, Sheriff, H, Dalmau, J, Tilley, DH, Glaser, CA. The frequency of autoimmune N-methyl-D-aspartate receptor encephalitis surpasses that of individual viral etiologies in young individuals enrolled in the California Encephalitis Project. Clin Infect Dis 2012;54:899904.Google Scholar
Pruss, H, Dalmau, J, Harms, L, et al. Retrospective analysis of NMDA receptor antibodies in encephalitis of unknown origin. Neurology 2010;75:17351739.Google Scholar
Armangue, T, Spatola, M, Vlagea, A, et al. Frequency, symptoms, risk factors, and outcomes of autoimmune encephalitis after herpes simplex encephalitis: a prospective observational study and retrospective analysis. Lancet Neurol 2018;17:760772.Google Scholar
Erickson, TA, Muscal, E, Munoz, FM, et al. Infectious and autoimmune causes of encephalitis in children. Pediatrics 2020;145:e20192543.Google Scholar
Titulaer, MJ, McCracken, L, Gabilondo, I, et al. Treatment and prognostic factors for long-term outcome in patients with anti-NMDA receptor encephalitis: an observational cohort study. Lancet Neurol 2013;12:157165.Google Scholar
Viaccoz, A, Desestret, V, Ducray, F, et al. Clinical specificities of adult male patients with NMDA receptor antibodies encephalitis. Neurology 2014;82:556563.Google Scholar
Irani, SR, Bera, K, Waters, P, et al. N-methyl-D-aspartate antibody encephalitis: temporal progression of clinical and paraclinical observations in a predominantly non-paraneoplastic disorder of both sexes. Brain 2010;133:16551667.Google Scholar
Florance, NR, Davis, RL, Lam, C, et al. Anti-N-methyl-D-aspartate receptor (NMDAR) encephalitis in children and adolescents. Ann Neurol 2009;66:1118.Google Scholar
Titulaer, MJ, McCracken, L, Gabilondo, I, et al. Late-onset anti-NMDA receptor encephalitis. Neurology 2013;81:10581063.Google Scholar
Bost, C, Chanson, E, Picard, G, et al. Malignant tumors in autoimmune encephalitis with anti-NMDA receptor antibodies. J Neurol 2018;265:21902200.Google Scholar
Chefdeville, A, Treilleux, I, Mayeur, ME, et al. Immunopathological characterization of ovarian teratomas associated with anti-N-methyl-D-aspartate receptor encephalitis. Acta Neuropathol Commun 2019;7:38.Google Scholar
Dai, Y, Zhang, J, Ren, H, et al. Surgical outcomes in patients with anti-N-methyl D-aspartate receptor encephalitis with ovarian teratoma. Am J Obstet Gynecol 2019;221:e481-485.Google Scholar
Afanasiev, V, Brechemier, ML, Boisseau, W, et al. Anti-NMDA receptor antibody encephalitis and neuroendocrine pancreatic tumor: causal link? Neurology 2016;87:112113.Google Scholar
Hara, M, Morita, A, Kamei, S, et al. Anti-N-methyl-D-aspartate receptor encephalitis associated with carcinosarcoma with neuroendocrine differentiation of the uterus. J Neurol 2011;258:13511353.Google Scholar
Kobayashi, M, Nishioka, K, Takanashi, M, et al. Anti-NMDA receptor encephalitis due to large-cell neuroendocrine carcinoma of the uterus. J Neurol Sci 2017;383:7274.Google Scholar
Jeraiby, M, Depince-Berger, A, Bossy, V, Antoine, JC, Paul, S. A case of anti-NMDA receptor encephalitis in a woman with a NMDA-R(+) small cell lung carcinoma (SCLC). Clin Immunol 2016;166–167:9699.Google Scholar
Coban, A, Gundogdu, G, Poyraz, M, et al. NMDA receptor encephalitis with cancer of unknown primary origin. Tumori 2016;102(Suppl. 2). doi: 10.5301/tj.5000447.Google Scholar
Kobayashi, Y, Sato, M, Kinoshita, M, et al. An elderly case of paraneoplastic anti-NMDA receptor encephalitis associated with small-cell lung cancer expressing NR1 subunits. Intern Med 2020;59:23072309.Google Scholar
Boangher, S, Mespouille, P, Filip, CM, Goffette, S. Small-cell lung cancer with positive anti-NMDAR and anti-AMPAR antibodies paraneoplastic limbic encephalitis. Case Rep Neurol Med 2016;2016:3263718.Google Scholar
Wu, YY, He, XJ, Zhang, ML, Shi, YY, Zhang, JW. Anti-N-methyl-D-aspartate receptor encephalitis with lung adenocarcinoma. Neurol Sci 2016;37:15731575.Google Scholar
Lim, JA, Lee, ST, Jung, KH, et al. Anti-N-methyl-d-aspartate receptor encephalitis in Korea: clinical features, treatment, and outcome. J Clin Neurol 2014;10:157161.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
Shalhout, SZ, Emerick, KS, Sadow, PM, Linnoila, JJ, Miller, DM. Regionally metastatic Merkel cell carcinoma associated with paraneoplastic anti-N-methyl-D-aspartate receptor encephalitis. Case Rep Oncol Med 2020;2020:1257587.Google Scholar
Lim, EW, Yip, CW. Anti-N-methyl-D-aspartate receptor encephalitis associated with hepatic neuroendocrine carcinoma: a case report. J Clin Neurosci 2017;41:7072.Google Scholar
Zandi, MS, Irani, SR, Follows, G, et al. Limbic encephalitis associated with antibodies to the NMDA receptor in Hodgkin lymphoma. Neurology 2009;73:20392040.Google Scholar
Park, BS, Son, GM, Kim, HS, Medina, J, Cho, JW. Anti-N-methyl-D-aspartate receptor encephalitis in a patient with colon cancer. Clin Neurol Neurosurg 2019;177:114116.Google Scholar
Ding, L, Tan, H, Li, Z, Ji, J, Song, X. Case report: anaesthetic management of radical gastrectomy for gastric cancer associated with anti-N-methyl-D-aspartate receptor encephalitis. BMC Anesthesiol 2017;17:90.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
Cho, EH, Byun, JM, Park, HY, et al. The first case of anti-N-methyl-D-aspartate receptor encephalitis (anti-NMDAR encephalitis) associated with ovarian mucinous cystadenoma: a case report. Taiwan J Obstet Gynecol 2019;58:557559.Google Scholar
Yang, J, Li, B, Li, X, Lai, Z. Anti-N-Methyl-D-aspartate receptor encephalitis associated with clear cell renal carcinoma: a case report. Front Oncol 2020;10:350.Google Scholar
Kaddu-Mulindwa, D, Roth, S, Klees-Rollmann, A, Fassbender, K, Fousse, M. Primary HIV infection presenting with Kaposi sarcoma and limbic encephalitis. J Neurovirol 2020;26:292296.Google Scholar
Mahadeen, A, Mullaguri, N, George, P, Rabinowitz, L, Newey, CR. Anti-N-methyl-D-aspartate encephalitis concomitantly with tall-cell variant papillary thyroid carcinoma. Cureus 2019;11:e5415.Google Scholar
Hattori, Y, Yamashita, Y, Mizuno, M, et al. Anti-N-methyl-D-aspartate receptor limbic encephalitis associated with mature cystic teratoma of the fallopian tube. J Obstet Gynaecol Res 2017;43:412415.Google Scholar
Nosadini, M, Mohammad, SS, Corazza, F, et al. Herpes simplex virus-induced anti-N-methyl-D-aspartate receptor encephalitis: a systematic literature review with analysis of 43 cases. Dev Med Child Neurol 2017;59:796805.Google Scholar
Ma, J, Han, W, Jiang, L. Japanese encephalitis-induced anti-N-methyl-D-aspartate receptor encephalitis: a hospital-based prospective study. Brain Dev 2020;42:179184.Google Scholar
Mueller, SH, Farber, A, Pruss, H, et al. Genetic predisposition in anti-LGI1 and anti-NMDA receptor encephalitis. Ann Neurol 2018;83:863869.Google Scholar
Shu, Y, Qiu, W, Zheng, J, et al. HLA class II allele DRB1*16:02 is associated with anti-NMDAR encephalitis. J Neurol Neurosurg Psychiatry 2019;90:652658.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
Garre, 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
Derksen, SJ, Goraj, B, Molenaar, JP, Hoeven, J. Severe anti NMDA encephalitis and EBV infection. Neth J Crit Care 2013;17:1921.Google Scholar
Cohen, DA, Lopez-Chiriboga, AS, Pittock, SJ, et al. Posttransplant autoimmune encephalitis. Neurol Neuroimmunol Neuroinflamm 2018;5:e497.Google Scholar
Toda, J, Maeda, T, Akuta, K, et al. Limbic encephalitis with antibodies to N-methyl-D-aspartate (NMDA)-type glutamate receptor after allogeneic transplantation. Int J Hematol 2020;112:254257.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
Tominaga, N, Kanazawa, N, Kaneko, A, et al. Prodromal headache in anti-NMDAR encephalitis: an epiphenomenon of NMDAR autoimmunity. Brain Behav 2018;8:e01012.Google Scholar
Kayser, MS, Dalmau, J. Anti-NMDA receptor encephalitis in psychiatry. Curr Psychiatry Rev 2011;7:189193.Google Scholar
Warren, N, Siskind, D, O’Gorman, C. Refining the psychiatric syndrome of anti-N-methyl-d-aspartate receptor encephalitis. Acta Psychiatr Scand 2018;138:401408.Google Scholar
Xu, X, Lu, Q, Huang, Y, et al. Anti-NMDAR encephalitis: A single-center, longitudinal study in China. Neurol Neuroimmunol Neuroinflamm 2020;7:e633.Google Scholar
Sabbula, BR, Yemmanur, S, Sanivarapu, R, Kagolanu, D, Shadab, A. Finding the cause of psychosis: a challenging case of anti-NMDAR encephalitis. Case Rep Med 2020;2020:2074704.Google Scholar
Guasp, M, Gine-Serven, E, Maudes, E, et al. Clinical, neuro-immunological, and CSF investigations in first episode psychosis. Neurology 2021;97:e61e75.Google Scholar
Pollak, TA, Lennox, BR, Muller, S, et al. Autoimmune psychosis: an international consensus on an approach to the diagnosis and management of psychosis of suspected autoimmune origin. Lancet Psychiatry 2020;7:93108.Google Scholar
Arino, H, Munoz-Lopetegi, A, Martinez-Hernandez, E, et al. Sleep disorders in anti-NMDAR encephalitis. Neurology 2020;95:e671e684.Google Scholar
Munoz-Lopetegi, A, Graus, F, Dalmau, J, Santamaria, J. Sleep disorders in autoimmune encephalitis. Lancet Neurol 2020;19:10101022.Google Scholar
Liu, X, Yan, B, Wang, R, et al. Seizure outcomes in patients with anti-NMDAR encephalitis: a follow-up study. Epilepsia 2017;58:21042111.Google Scholar
Haberlandt, E, Ensslen, M, Gruber-Sedlmayr, U, et al. Epileptic phenotypes, electroclinical features and clinical characteristics in 17 children with anti-NMDAR encephalitis. Eur J Paediatr Neurol 2017;21:457464.Google Scholar
de Bruijn, M, van Sonderen, A, van Coevorden-Hameete, MH, et al. Evaluation of seizure treatment in anti-LGI1, anti-NMDAR, and anti-GABABR encephalitis. Neurology 2019;92:e2185e2196.Google Scholar
Schmitt, SE, Pargeon, K, Frechette, ES, et al. Extreme delta brush: a unique EEG pattern in adults with anti-NMDA receptor encephalitis. Neurology 2012;79:10941100.Google Scholar
Qu, XP, Vidaurre, J, Peng, XL, et al. Seizure characteristics, outcome, and risk of epilepsy in pediatric anti-N-methyl-D-aspartate receptor encephalitis. Pediatr Neurol 2020;105:3540.Google Scholar
Chavez-Castillo, M, Ruiz-Garcia, M, Herrera-Mora, P. Characterization and outcomes of epileptic seizures in Mexican pediatric patients with anti-N-methyl-D-aspartate receptor encephalitis. Cureus 2020;12:e8211.Google Scholar
Geis, C, Planaguma, J, Carreno, M, Graus, F, Dalmau, J. Autoimmune seizures and epilepsy. J Clin Invest 2019;129:926940.Google Scholar
Steriade, C, Britton, J, Dale, RC, et al. Acute symptomatic seizures secondary to autoimmune encephalitis and autoimmune-associated epilepsy: conceptual definitions. Epilepsia 2020;61:13411351.Google Scholar
Shen, CH, Fang, GL, Yang, F, et al. Seizures and risk of epilepsy in anti-NMDAR, anti-LGI1, and anti-GABAB R encephalitis. Ann Clin Transl Neurol 2020;7:13921399.Google Scholar
Baizabal-Carvallo, JF, Stocco, A, Muscal, E, Jankovic, J. The spectrum of movement disorders in children with anti-NMDA receptor encephalitis. Mov Disord 2013;28:543547.Google Scholar
Varley, JA, Webb, AJS, Balint, B, et al. The movement disorder associated with NMDAR antibody-encephalitis is complex and characteristic: an expert video-rating study. J Neurol Neurosurg Psychiatry 2019;90:724726.Google Scholar
Frechette, ES, Zhou, L, Galetta, SL, Chen, L, Dalmau, J. Prolonged follow-up and CSF antibody titers in a patient with anti-NMDA receptor encephalitis. Neurology 2011;76:S64S66.Google Scholar
Uchino, A, Iizuka, T, Urano, Y, et al. Pseudo-piano playing motions and nocturnal hypoventilation in anti-NMDA receptor encephalitis: response to prompt tumor removal and immunotherapy. Intern Med 2011;50:627630.Google Scholar
Ali, F, Wijdicks, EF. Treatment of movement disorder emergencies in autoimmune encephalitis in the neurosciences ICU. Neurocrit Care 2020;32:286294.Google Scholar
Zheng, F, Ye, X, Shi, X, Poonit, ND, Lin, Z. Management of refractory orofacial dyskinesia caused by anti-N-methyl-D-aspartate receptor encephalitis using botulinum toxin. Front Neurol 2018;9:81.Google Scholar
Gumbinger, C, Hametner, C, Wildemann, B, Veltkamp, R, Bosel, J. Administration of isoflurane-controlled dyskinetic movements caused by anti-NMDAR encephalitis. Neurology 2013;80:19971998.Google Scholar
de Montmollin, E, Demeret, S, Brule, N, et al. Anti-N-methyl-D-aspartate receptor encephalitis in adult patients requiring intensive care. Am J Respir Crit Care Med 2017;195:491499.Google Scholar
Lim, JA, Lee, ST, Kim, TJ, et al. Frequent rhabdomyolysis in anti-NMDA receptor encephalitis. J Neuroimmunol 2016;298:178180.Google Scholar
Howard, CM, Kass, JS, Bandi, VDP, Guntupalli, KK. Challenges in providing critical care for patients with anti-N-methyl-D-aspartate receptor encephalitis. Chest 2014;145:11431147.Google Scholar
Mehr, SR, Neeley, RC, Wiley, M, Kumar, AB. Profound autonomic instability complicated by multiple episodes of cardiac asystole and refractory bradycardia in a patient with anti-NMDA encephalitis. Case Rep Neurol Med 2016;2016:7967526.Google Scholar
Kumar, MA, Jain, A, Dechant, VE, et al. Anti-N-methyl-D-aspartate receptor encephalitis during pregnancy. Arch Neurol 2010;67:884887.Google Scholar
Chia, PL, Tan, K, Foo, D. Profound sinus node dysfunction in anti-N-methyl-D-aspartate receptor limbic encephalitis. Pacing Clin Electrophysiol 2013;36:e9092.Google Scholar
Yan, L, Zhang, S, Huang, X, Tang, Y, Wu, J. Clinical study of autonomic dysfunction in patients with anti-NMDA receptor encephalitis. Front Neurol 2021;12:609750.Google Scholar
Lee, M, Lawn, N, Prentice, D, Chan, J. Anti-NMDA receptor encephalitis associated with ictal asystole. J Clin Neurosci 2011;18:17161718.Google Scholar
Millichap, JJ, Goldstein, JL, Laux, LC, et al. Ictal asystole and anti-N-methyl-D-aspartate receptor antibody encephalitis. Pediatrics 2011;127:e781e786.Google Scholar
Schuele, SU, Bermeo, AC, Alexopoulos, AV, et al. Video-electrographic and clinical features in patients with ictal asystole. Neurology 2007;69:434441.Google Scholar
Britton, JW, Ghearing, GR, Benarroch, EE, Cascino, GD. The ictal bradycardia syndrome: localization and lateralization. Epilepsia 2006;47:737744.Google Scholar
Inayat, F, Hung Pinto, WA, Ahmad, S, Hussain, A, Ullah, W. Anti-N-methyl-D-aspartate receptor encephalitis associated with ictal torsades de pointes and cardiac arrest. Cureus 2019;11:e4837.Google Scholar
Bell, M, Friedman, M, Matar, M, et al. Anesthesia for pediatric patients with anti-NMDA receptor encephalitis: a retrospective case series. Paediatr Anaesth 2021;31:316322.Google Scholar
Lapebie, FX, Kennel, C, Magy, L, et al. Potential side effect of propofol and sevoflurane for anesthesia of anti-NMDA-R encephalitis. BMC Anesthesiol 2014;14:5.Google Scholar
Sato, M, Yasumoto, H, Arai, T. General anesthesia with propofol for ovarian teratoma excision associated with anti-N-methyl-D-aspartate receptor encephalitis. JA Clin Rep 2018;4:14.Google Scholar
Broderick, DK, Raines, DE, Nanji, KC. Total intravenous anesthesia using N-methyl-D-aspartate (NMDA) receptor-sparing drugs in a patient with anti-NMDA receptor encephalitis. A A Case Rep 2014;2:8385.Google Scholar
Yamanaka, D, Kawano, T, Tateiwa, H, et al. Successful management of dexmedetomidine for postoperative intensive care sedation in a patient with anti-NMDA receptor encephalitis: a case report and animal experiment. Springerplus 2016;5:1380.Google Scholar
Wada, N, Tashima, K, Motoyasu, A, et al. Anesthesia for patient with anti-N-methyl-D-aspartate receptor encephalitis: a case report with a brief review of the literature. Medicine (Baltimore) 2018;97:e13651.Google Scholar
Al Ghamdi, F, Uffman, JC, Kim, SS, Nafiu, OO, Tobias, JD. Anesthetic care for patients with anti-NMDA receptor encephalitis. Saudi J Anaesth 2020;14:164168.Google Scholar
Liu, H, Jian, M, Liang, F, Yue, H, Han, R. Anti-N-methyl-D-aspartate receptor encephalitis associated with an ovarian teratoma: two cases report and anesthesia considerations. BMC Anesthesiol 2015;15:150.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
Sakamoto, T, Endo, A, Yoshitomi, H, Tanabe, K. Takotsubo cardiomyopathy caused by intense emotional stress induced by voluntary quarantine during the coronavirus disease crisis. Circ Rep 2020;2:382383.Google Scholar
Gelow, J, Kruer, M, Yadav, V, Kaul, S. Apical ballooning resulting from limbic encephalitis. Am J Med 2009;122:583586.Google Scholar
Finsterer, J, Wahbi, K. CNS disease triggering Takotsubo stress cardiomyopathy. Int J Cardiol 2014;177:322329.Google Scholar
Dubey, D, Singh, J, Britton, JW, et al. Predictive models in the diagnosis and treatment of autoimmune epilepsy. Epilepsia 2017;58:11811189.Google Scholar
Dubey, D, Kothapalli, N, McKeon, A, et al. Predictors of neural-specific autoantibodies and immunotherapy response in patients with cognitive dysfunction. J Neuroimmunol 2018;323:6272.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
Ho, ACC, Mohammad, SS, Pillai, SC, et al. High sensitivity and specificity in proposed clinical diagnostic criteria for anti-N-methyl-D-aspartate receptor encephalitis. Dev Med Child Neurol 2017;59:12561260.Google Scholar
Kaneko, A, Kaneko, J, Tominaga, N, et al. Pitfalls in clinical diagnosis of anti-NMDA receptor encephalitis. J Neurol 2018;265:586596.Google Scholar
Nishida, H, Kohyama, K, Kumada, S, et al. Evaluation of the diagnostic criteria for anti-NMDA receptor encephalitis in Japanese children. Neurology 2021;96:e2070e2077.Google Scholar
Nicolle, DCM, Moses, JL. A systematic review of the neuropsychological sequelae of people diagnosed with anti-N-methyl-D-aspartate receptor encephalitis in the acute and chronic phases. Arch Clin Neuropsychol 2018;33:964983.Google Scholar
Iizuka, T, Kaneko, J, Tominaga, N, et al. Association of progressive cerebellar atrophy with long-term outcome in patients with anti-N-methyl-d-aspartate receptor encephalitis. JAMA Neurol 2016;73:706713.Google Scholar
Peer, M, Pruss, H, Ben-Dayan, I, et al. Functional connectivity of large-scale brain networks in patients with anti-NMDA receptor encephalitis: an observational study. Lancet Psychiatry 2017;4:768774.Google Scholar
Finke, C, Kopp, UA, Pajkert, A, et al. Structural hippocampal damage following anti-N-methyl-D-aspartate receptor encephalitis. Biol Psychiatry 2016;79:727734.Google Scholar
Phillips, OR, Joshi, SH, Narr, KL, et al. Superficial white matter damage in anti-NMDA receptor encephalitis. J Neurol Neurosurg Psychiatry 2018;89:518525.Google Scholar
Sonderen, AV, Arends, S, Tavy, DLJ, et al. Predictive value of electroencephalography in anti-NMDA receptor encephalitis. J Neurol Neurosurg Psychiatry 2018;89:11011106.Google Scholar
Jeannin-Mayer, S, Andre-Obadia, N, Rosenberg, S, et al. EEG analysis in anti-NMDA receptor encephalitis: description of typical patterns. Clin Neurophysiol 2019;130:289296.Google Scholar
Yuan, J, Guan, H, Zhou, X, et al. Changing brain metabolism patterns in patients with ANMDARE: serial 18F-FDG PET/CT findings. Clin Nucl Med 2016;41:366370.Google Scholar
Probasco, JC, Solnes, L, Nalluri, A, et al. Abnormal brain metabolism on FDG-PET/CT is a common early finding in autoimmune encephalitis. Neurol Neuroimmunol Neuroinflamm 2017;4:e352.Google Scholar
Lagarde, S, Lepine, A, Caietta, E, et al. Cerebral (18)fluorodeoxy-glucose positron emission tomography in paediatric anti N-methyl-D-aspartate receptor encephalitis: acase series. Brain Dev 2016;38:461470.Google Scholar
Leypoldt, F, Hoftberger, R, Titulaer, MJ, et al. Investigations on CXCL13 in anti-N-methyl-D-aspartate receptor encephalitis: a potential biomarker of treatment response. JAMA Neurol 2015;72:180186.Google Scholar
Chen, J, Ding, Y, Zheng, D, et al. Elevation of YKL-40 in the CSF of anti-NMDAR encephalitis patients is associated with poor prognosis. Front Neurol 2018;9:727.Google Scholar
Ding, YW, Pan, SY, Xie, W, Shen, HY, Wang, HH. Elevated soluble Fas and FasL in cerebrospinal fluid and serum of patients with anti-N-methyl-D-aspartate receptor encephalitis. Front Neurol 2018;9:904.Google Scholar
Iizuka, T, Yoshii, S, Kan, S, et al. Reversible brain atrophy in anti-NMDA receptor encephalitis: a long-term observational study. J Neurol 2010;257:16861691.Google Scholar
Matute, C, Palma, A, Serrano-Regal, MP, et al. N-methyl-D-aspartate receptor antibodies in autoimmune encephalopathy alter oligodendrocyte function. Ann Neurol 2020;87:670676.Google Scholar
Veciana, M, Becerra, JL, Fossas, P, et al. EEG extreme delta brush: an ictal pattern in patients with anti-NMDA receptor encephalitis. Epilepsy Behav 2015;49:280285.Google Scholar
Gillinder, L, Warren, N, Hartel, G, Dionisio, S, O’Gorman, C. EEG findings in NMDA encephalitis: a systematic review. Seizure 2019;65:2024.Google Scholar
Sands, TT, Nash, K, Tong, S, Sullivan, J. Focal seizures in children with anti-NMDA receptor antibody encephalitis. Epilepsy Res 2015;112:3136.Google Scholar
Leypoldt, F, Buchert, R, Kleiter, I, et al. Fluorodeoxyglucose positron emission tomography in anti-N-methyl-D-aspartate receptor encephalitis: distinct pattern of disease. J Neurol Neurosurg Psychiatry 2012;83:681686.Google Scholar
Peng, Y, Liu, B, Pei, S, et al. Higher CSF levels of NLRP3 inflammasome is associated with poor prognosis of anti-N-methyl-D-aspartate receptor encephalitis. Front Immunol 2019;10:905.Google Scholar
Li, J, Gu, Y, An, H, et al. Cerebrospinal fluid light and heavy neurofilament level increased in anti-N-methyl-d-aspartate receptor encephalitis. Brain Behav 2019;9:e01354.Google Scholar
Zou, C, Pei, S, Yan, W, et al. Cerebrospinal fluid osteopontin and inflammation-associated cytokines in patients with anti-N-methyl-D-aspartate receptor encephalitis. Front Neurol 2020;11:519692.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
Camdessanche, JP, Streichenberger, N, Cavillon, G, et al. Brain immunohistopathological study in a patient with anti-NMDAR encephalitis. Eur J Neurol 2011;18:929931.Google Scholar
Day, GS, Laiq, S, Tang-Wai, DF, Munoz, DG. Abnormal neurons in teratomas in NMDAR encephalitis. JAMA Neurol 2014;71:717724.Google Scholar
Nolan, A, Buza, N, Margeta, M, Rabban, JT. Ovarian teratomas in women with anti-N-methyl-D-aspartate receptor encephalitis: topography and composition of immune cell and neuroglial populations is compatible with an autoimmune mechanism of disease. Am J Surg Pathol 2019;43:949964.Google Scholar
Hong, SB, Shin, YW, Shin, YW, Lee, SK, Chu, K. Occult growing teratoma as the cause of protracted symptoms in a patient with anti-NMDA-receptor encephalitis and prior ovarian teratoma removal: implications for continued monitoring and treatment. J Clin Neurol 2021;17:131133.Google Scholar
Dabner, M, McCluggage, WG, Bundell, C, et al. Ovarian teratoma associated with anti-n-methyl D-aspartate receptor encephalitis: a report of 5 cases documenting prominent intratumoral lymphoid infiltrates. Int J Gynecol Pathol 2012;31:429437.Google Scholar
Johnson, N, Henry, C, Fessler, AJ, Dalmau, J. Anti-NMDA receptor encephalitis causing prolonged nonconvulsive status epilepticus. Neurology 2010;75:14801482.Google Scholar
Sahin, H, Abdullazade, S, Sanci, M. Mature cystic teratoma of the ovary: a cutting edge overview on imaging features. Insights Imaging 2017;8:227241.Google Scholar
Lwanga, A, Kamson, DO, Wilkins, TE, et al. Occult teratoma in a case of N-methyl-D-aspartate receptor encephalitis. Neuroradiol J 2018;31:415419.Google Scholar
Alexopoulos, H, Kosmidis, ML, Dalmau, J, Dalakas, MC. Paraneoplastic anti-NMDAR encephalitis: long term follow-up reveals persistent serum antibodies. J Neurol 2011;258:15681570.Google Scholar
Tuzun, E, Zhou, L, Baehring, JM, et al. Evidence for antibody-mediated pathogenesis in anti-NMDAR encephalitis associated with ovarian teratoma. Acta Neuropathol 2009;118:737743.Google Scholar
Jiang, XY, Lei, S, Zhang, L, et al. Co-expression of NMDA-receptor subunits NR1, NR2A, and NR2B in dysplastic neurons of teratomas in patients with paraneoplastic NMDA-receptor-encephalitis: a retrospective clinico-pathology study of 159 patients. Acta Neuropathol Commun 2020;8:130.Google Scholar
Makuch, M, Wilson, R, Al-Diwani, A, et al. N-methyl-D-aspartate receptor antibody production from germinal center reactions: therapeutic implications. Ann Neurol 2018;83:553561.Google Scholar
Abdul-Rahman, ZM, Panegyres, PK, Roeck, M, et al. Anti-N-methyl-D-aspartate receptor encephalitis with an imaging-invisible ovarian teratoma: a case report. J Med Case Rep 2016;10:296.Google Scholar
Delangle, R, Demeret, S, Canlorbe, G, et al. Anti-NMDA receptor encephalitis associated with ovarian tumor: the gynecologist point of view. Arch Gynecol Obstet 2020;302:315320.Google Scholar
Boeck, AL, Logemann, F, Krauss, T, et al. Ovarectomy despite negative imaging in anti-NMDA receptor encephalitis: effective even late. Case Rep Neurol Med 2013;2013:843192.Google Scholar
Willis, RA. A further study of the structure of teratomata. J Pathol Bacteriol 1937;45:4965.Google Scholar
Mangler, M, Trebesch de Perez, I, Teegen, B, et al. Seroprevalence of anti-N-methyl-D-aspartate receptor antibodies in women with ovarian teratoma. J Neurol 2013;260:28312835.Google Scholar
Gong, S, Zhou, M, Shi, G, et al. Absence of NMDA receptor antibodies in patients with ovarian teratoma without encephalitis. Neurol Neuroimmunol Neuroinflamm 2017;4:e344.Google Scholar
Trillsch, F, Eichhorn, P, Oliveira-Ferrer, L, et al. No need for NMDA receptor antibody screening in neurologically asymptomatic patients with ovarian teratomas. J Neurol 2018;265:431432.Google Scholar
Nosadini, M, Thomas, T, Eyre, M, et al. International consensus recommendations for the treatment of paediatric NMDAR-antibody encephalitis. Neurol Neuroimmunol Neuroinflamm 2021;8:e1052.Google Scholar
Nepal, G, Shing, YK, Yadav, JK, et al. Efficacy and safety of rituximab in autoimmune encephalitis: a meta-analysis. Acta Neurol Scand 2020;142:449459.Google Scholar
Schumacher, LT, Mann, AP, MacKenzie, JG. Agitation management in pediatric males with anti-N-methyl-D-aspartate receptor encephalitis. J Child Adolesc Psychopharmacol 2016;26:939943.Google Scholar
Mohammad, SS, Jones, H, Hong, M, et al. Symptomatic treatment of children with anti-NMDAR encephalitis. Dev Med Child Neurol 2016;58:376384.Google Scholar
Jun, JS, Seo, HG, Lee, ST, Chu, K, Lee, SK. Botulinum toxin treatment for hypersalivation in anti-NMDA receptor encephalitis. Ann Clin Transl Neurol 2017;4:830834.Google Scholar
Scott, RA, Rabinstein, AA. Paroxysmal sympathetic hyperactivity. Semin Neurol 2020;40:485491.Google Scholar
Dale, RC, Brilot, F, Duffy, LV, et al. Utility and safety of rituximab in pediatric autoimmune and inflammatory CNS disease. Neurology 2014;83:142150.Google Scholar
Lee, WJ, Lee, ST, Moon, J, et al. Tocilizumab in autoimmune encephalitis refractory to rituximab: an institutional cohort study. Neurotherapeutics 2016;13:824832.Google Scholar
Scheibe, F, Pruss, H, Mengel, AM, et al. Bortezomib for treatment of therapy-refractory anti-NMDA receptor encephalitis. Neurology 2017;88:366370.Google Scholar
Behrendt, V, Krogias, C, Reinacher-Schick, A, Gold, R, Kleiter, I. Bortezomib treatment for patients with anti-N-methyl-d-aspartate receptor encephalitis. JAMA Neurol 2016;73:12511253.Google Scholar
Cordani, R, Micalizzi, C, Giacomini, T, et al. Bortezomib-responsive refractory anti-N-methyl-D-aspartate receptor encephalitis. Pediatr Neurol 2020;103:6164.Google Scholar
Keddie, S, Crisp, SJ, Blackaby, J, et al. Plasma cell depletion with bortezomib in the treatment of refractory N-methyl-D-aspartate (NMDA) receptor antibody encephalitis: rational developments in neuroimmunological treatment. Eur J Neurol 2018;25:13841388.Google Scholar
Kim, LN, Edwards, L, Goonetilleke, N, et al. Bortezomib for the treatment of refractory anti-N-methyl-D-aspartate receptor encephalitis. Intern Med J 2020;50:15911592.Google Scholar
Lazzarin, SM, Vabanesi, M, Cecchetti, G, et al. Refractory anti-NMDAR encephalitis successfully treated with bortezomib and associated movements disorders controlled with tramadol: a case report with literature review. J Neurol 2020;267:24622468.Google Scholar
Schroeder, C, Back, C, Koc, U, et al. Breakthrough treatment with bortezomib for a patient with anti-NMDAR encephalitis. Clin Neurol Neurosurg 2018;172:2426.Google Scholar
Sveinsson, O, Granqvist, M, Forslin, Y, et al. Successful combined targeting of B- and plasma cells in treatment refractory anti-NMDAR encephalitis. J Neuroimmunol 2017;312:1518.Google Scholar
Turnbull, MT, Siegel, JL, Becker, TL, et al. Early bortezomib therapy for refractory anti-NMDA receptor encephalitis. Front Neurol 2020;11:188.Google Scholar
Zhang, XT, Wang, CJ, Wang, BJ, Guo, SG. The short-term efficacy of combined treatments targeting B cell and plasma cell in severe and refractory anti-N-methyl-D-aspartate receptor encephalitis: two case reports. CNS Neurosci Ther 2019;25:151153.Google Scholar
Wang, T, Wang, B, Zeng, Z, et al. Efficacy and safety of bortezomib in rituximab-resistant anti-N-methyl-D-aspartate receptor (anti-NMDAR) encephalitis as well as the clinical characteristics: an observational study. J Neuroimmunol 2021;354:577527.Google Scholar
Shin, YW, Lee, ST, Kim, TJ, Jun, JS, Chu, K. Bortezomib treatment for severe refractory anti-NMDA receptor encephalitis. Ann Clin Transl Neurol 2018;5:598605.Google Scholar
Ratuszny, D, Skripuletz, T, Wegner, F, et al. Case report: daratumumab in a patient with severe refractory anti-NMDA receptor encephalitis. Front Neurol 2020;11:602102.Google Scholar
Parratt, KL, Allan, M, Lewis, SJ, et al. Acute psychiatric illness in a young woman: an unusual form of encephalitis. Med J Aust 2009;191:284286.Google Scholar
Maccaferri, GE, Rossetti, AO, Dalmau, J, Berney, A. Anti-N-methyl-D-aspartate receptor encephalitis: a new challenging entity for consultation-liaison psychiatrist. Brain Disord Ther 2016;5:215.Google Scholar
Kashihara, T, Nozaki, I, Sakai, K, et al. Recovery from multidisciplinary therapy-refractory anti-NMDA receptor encephalitis after over three years of mechanical ventilation. Clin Neurol Neurosurg 2021;202:106477.Google Scholar
Lee, WJ, Lee, ST, Shin, YW, et al. Teratoma removal, steroid, IVIG, rituximab and tocilizumab (T-SIRT) in anti-NMDAR encephalitis. Neurotherapeutics 2020;18:474487.Google Scholar
Tatencloux, S, Chretien, P, Rogemond, V, et al. Intrathecal treatment of anti-N-methyl-D-aspartate receptor encephalitis in children. Dev Med Child Neurol 2015;57:9599.Google Scholar
Bravo-Oro, A, Abud-Mendoza, C, Quezada-Corona, A, Dalmau, J, Campos-Guevara, V. [Anti-N-methyl-D-aspartate (NMDA) receptor encephalitis: experience with six pediatric patients: potential efficacy of methotrexate]. Rev Neurol 2013;57:405410.Google Scholar
Dogan Onugoren, M, Golombeck, KS, Bien, C, et al. Immunoadsorption therapy in autoimmune encephalitides. Neurol Neuroimmunol Neuroinflamm 2016;3:e207.Google Scholar
Nosadini, M, Mohammad, SS, Toldo, I, Sartori, S, Dale, RC. Mycophenolate mofetil, azathioprine and methotrexate usage in paediatric anti-NMDAR encephalitis: a systematic literature review. Eur J Paediatr Neurol 2019;23:718.Google Scholar
Cooper, JJ, Afzal, KI. Safety of electroconvulsive therapy in 2 very young pediatric patients with catatonia related to anti-N-methyl-D-aspartate receptor encephalitis. J ECT 2019;35:216217.Google Scholar
Moussa, T, Afzal, K, Cooper, J, et al. Pediatric anti-NMDA receptor encephalitis with catatonia: treatment with electroconvulsive therapy. Pediatr Rheumatol Online J 2019;17:8.Google Scholar
Medina, M, Cooper, JJ. Refractory catatonia due to N-methyl-D-aspartate receptor encephalitis responsive to electroconvulsive therapy: the clinical use of the clock drawing test. J ECT 2017;33:223224.Google Scholar
Sunwoo, JS, Jung, DC, Choi, JY, et al. Successful treatment of refractory dyskinesia secondary to anti-N-methyl-D-aspartate receptor encephalitis with electroconvulsive therapy. J ECT 2016;32:e1314.Google Scholar
Matsumoto, T, Matsumoto, K, Kobayashi, T, Kato, S. Electroconvulsive therapy can improve psychotic symptoms in anti-NMDA-receptor encephalitis. Psychiatry Clin Neurosci 2012;66:242243.Google Scholar
Braakman, HM, Moers-Hornikx, VM, Arts, BM, et al. Pearls & oy-sters: electroconvulsive therapy in anti-NMDA receptor encephalitis. Neurology 2010;75:e44e46.Google Scholar
Coffey, MJ, Cooper, JJ. Electroconvulsive therapy in anti-N-methyl-D-aspartate receptor encephalitis: a case report and review of the literature. J ECT 2016;32:225229.Google Scholar
Warren, N, Grote, V, O’Gorman, C, Siskind, D. Electroconvulsive therapy for anti-N-methyl-d-aspartate (NMDA) receptor encephalitis: a systematic review of cases. Brain Stimul 2019;12:329334.Google Scholar
Creten, C, van der Zwaan, S, Blankespoor, RJ, et al. Late onset autism and anti-NMDA-receptor encephalitis. Lancet 2011;378:98.Google Scholar
Gonzalez-Valcarcel, J, Rosenfeld, MR, Dalmau, J. [Differential diagnosis of encephalitis due to anti-NMDA receptor antibodies]. Neurologia 2010;25:409413.Google Scholar
Sinha, A, Ewies, AA. Ovarian mature cystic teratoma: challenges of surgical management. Obstet Gynecol Int 2016;2016:2390178.Google Scholar
Jorge, S, Jones, NL, Chen, L, et al. Characteristics, treatment and outcomes of women with immature ovarian teratoma, 1998–2012. Gynecol Oncol 2016;142:261266.Google Scholar
Tanyi, JL, Marsh, EB, Dalmau, J, Chu, CS. Reversible paraneoplastic encephalitis in three patients with ovarian neoplasms. Acta Obstet Gynecol Scand 2012;91:630634.Google Scholar
Anderson, D, Nathoo, N, Henry, M, et al. Oophorectomy in NMDA receptor encephalitis and negative pelvic imaging. Pract Neurol 2020. doi: 10.1136/practneurol-2020-002676.Google Scholar
Masghati, S, Nosratian, M, Dorigo, O. Anti-N-methyl-aspartate receptor encephalitis in identical twin sisters: role for oophorectomy. Obstet Gynecol 2014;123:433435.Google Scholar
Hansen, HC, Klingbeil, C, Dalmau, J, et al. Persistent intrathecal antibody synthesis 15 years after recovering from anti-N-methyl-D-aspartate receptor encephalitis. JAMA Neurol 2013;70:117119.Google Scholar
Gresa-Arribas, N, Titulaer, MJ, Torrents, A, et al. Antibody titres at diagnosis and during follow-up of anti-NMDA receptor encephalitis: a retrospective study. Lancet Neurol 2014;13:167177.Google Scholar
Wang, W, Li, JM, Hu, FY, et al. Anti-NMDA receptor encephalitis: clinical characteristics, predictors of outcome and the knowledge gap in southwest China. Eur J Neurol 2016;23:621629.Google Scholar
Shim, Y, Kim, SY, Kim, H, et al. Clinical outcomes of pediatric Anti-NMDA receptor encephalitis. Eur J Paediatr Neurol 2020;29:8791.Google Scholar
Zhang, Y, Liu, G, Jiang, M, et al. Clinical characteristics and prognosis of severe anti-N-methyl-D-aspartate receptor encephalitis patients. Neurocrit Care 2018;29:264272.Google Scholar
Armangue, T, Titulaer, MJ, Malaga, I, et al. Pediatric anti-N-methyl-D-aspartate receptor encephalitis-clinical analysis and novel findings in a series of 20 patients. J Pediatr 2013;162:850856.Google Scholar
Byrne, S, Walsh, C, Hacohen, Y, et al. Earlier treatment of NMDAR antibody encephalitis in children results in a better outcome. Neurol Neuroimmunol Neuroinflamm 2015;2:e130.Google Scholar
Gordon-Lipkin, E, Yeshokumar, AK, Saylor, D, Arenivas, A, Probasco, JC. Comparative outcomes in children and adults with Anti-N-methyl-D-aspartate (anti-NMDA) receptor encephalitis. J Child Neurol 2017;32:930935.Google Scholar
Balu, R, McCracken, L, Lancaster, E, et al. A score that predicts 1-year functional status in patients with anti-NMDA receptor encephalitis. Neurology 2019;92:e244e252.Google Scholar
Peng, Y, Dai, F, Liu, L, et al. Validation of the NEOS score in Chinese patients with anti-NMDAR encephalitis. Neurol Neuroimmunol Neuroinflamm 2020;7:e860.Google Scholar
Finke, C, Kopp, UA, Pruss, H, et al. Cognitive deficits following anti-NMDA receptor encephalitis. J Neurol Neurosurg Psychiatry 2012;83:195198.Google Scholar
Blum, RA, Tomlinson, AR, Jette, N, et al. Assessment of long-term psychosocial outcomes in anti-NMDA receptor encephalitis. Epilepsy Behav 2020;108:107088.Google Scholar
de Bruijn, M, Aarsen, FK, van Oosterhout, MP, et al. Long-term neuropsychological outcome following pediatric anti-NMDAR encephalitis. Neurology 2018;90:e1997e2005.Google Scholar
Tomlinson, AR, Blum, RA, Jette, N, et al. Assessment of care transitions and caregiver burden in anti-NMDA receptor encephalitis. Epilepsy Behav 2020;108:107066.Google Scholar
Cainelli, E, Nosadini, M, Sartori, S, Suppiej, A. Neuropsychological and psychopathological profile of anti-NNMDA encephalitis: a possible pathophysiological model for pediatric neuropsychiatric disorders. Arch Clin Neuropsychol 2019;34:13091319.Google Scholar
Arnulf, I. Kleine-Levin syndrome. Sleep Med Clin 2015;10:151161.Google Scholar
Lim, JA, Lee, ST, Moon, J, et al. Development of the clinical assessment scale in autoimmune encephalitis. Ann Neurol 2019;85:352358.Google Scholar
Gabilondo, I, Saiz, A, Galan, L, et al. Analysis of relapses in anti-NMDAR encephalitis. Neurology 2011;77:996999.Google Scholar
Joubert, B, Garcia-Serra, A, Planaguma, J, et al. Pregnancy outcomes in anti-NMDA receptor encephalitis: case series. Neurol Neuroimmunol Neuroinflamm 2020;7:e668.Google Scholar
Chourasia, N, Watkins, MW, Lankford, JE, Kass, JS, Kamdar, A. An infant born to a mother with anti-N-methyl-D-aspartate receptor encephalitis. Pediatr Neurol 2018;79:6568.Google Scholar
Lamale-Smith, LM, Moore, GS, Guntupalli, SR, Scott, JB. Maternal–fetal transfer of anti-N-methyl-D-aspartate receptor antibodies. Obstet Gynecol 2015;125:10561058.Google Scholar
Hilderink, M, Titulaer, MJ, Schreurs, MW, Keizer, K, Bunt, JE. Transient anti-NMDAR encephalitis in a newborn infant due to transplacental transmission. Neurol Neuroimmunol Neuroinflamm 2015;2:e126.Google Scholar
Jagota, P, Vincent, A, Bhidayasiri, R. Transplacental transfer of NMDA receptor antibodies in an infant with cortical dysplasia. Neurology 2014;82:16621663.Google Scholar
Das, G, Damotte, V, Gelfand, JM, et al. Rituximab before and during pregnancy: a systematic review, and a case series in MS and NMOSD. Neurol Neuroimmunol Neuroinflamm 2018;5:e453.Google Scholar
Kumpfel, T, Thiel, S, Meinl, I, et al. Anti-CD20 therapies and pregnancy in neuroimmunologic disorders: a cohort study from Germany. Neurol Neuroimmunol Neuroinflamm 2021;8:e913.Google Scholar
Fleischmann, R, Pruss, H, Rosche, B, et al. Severe cognitive impairment associated with intrathecal antibodies to the NR1 subunit of the N-methyl-D-aspartate receptor in a patient with multiple sclerosis. JAMA Neurol 2015;72:9699.Google Scholar
Ramberger, M, Bsteh, G, Schanda, K, et al. NMDA receptor antibodies: a rare association in inflammatory demyelinating diseases. Neurol Neuroimmunol Neuroinflamm 2015;2:e141.Google Scholar
Du, L, Wang, H, Zhou, H, et al. Anti-NMDA receptor encephalitis concomitant with myelin oligodendrocyte glycoprotein antibody diseases: a retrospective observational study. Medicine (Baltimore) 2020;99:e21238.Google Scholar
Tao, S, Zhang, Y, Ye, H, Guo, D. AQP4-IgG-seropositive neuromyelitis optica spectrum disorder (NMOSD) coexisting with anti-N-methyl-D-aspartate receptor (NMDAR) encephalitis: a case report and literature review. Mult Scler Relat Disord 2019;35:185192.Google Scholar
Martinez-Hernandez, E, Guasp, M, Garcia-Serra, A, et al. Clinical significance of anti-NMDAR concurrent with glial or neuronal surface antibodies. Neurology 2020;94:e2302e2310.Google Scholar
Dubey, D, Hinson, SR, Jolliffe, EA, et al. Autoimmune GFAP astrocytopathy: prospective evaluation of 90 patients in 1 year. J Neuroimmunol 2018;321:157163.Google Scholar
Kunchok, A, Zekeridou, A, McKeon, A. Autoimmune glial fibrillary acidic protein astrocytopathy. Curr Opin Neurol 2019;32:452458.Google Scholar
Hjalmarsson, A, Blomqvist, P, Skoldenberg, B. Herpes simplex encephalitis in Sweden, 1990–2001: incidence, morbidity, and mortality. Clin Infect Dis 2007;45:875880.Google Scholar
Gnann, JW Jr., Whitley, RJ. Herpes simplex encephalitis: an update. Curr Infect Dis Rep 2017;19:13.Google Scholar
Skoldenberg, B, Aurelius, E, Hjalmarsson, A, et al. Incidence and pathogenesis of clinical relapse after herpes simplex encephalitis in adults. J Neurol 2006;253:163170.Google Scholar
Schleede, L, Bueter, W, Baumgartner-Sigl, S, et al. Pediatric herpes simplex virus encephalitis: a retrospective multicenter experience. J Child Neurol 2013;28:321331.Google Scholar
Kimura, H, Aso, K, Kuzushima, K, et al. Relapse of herpes simplex encephalitis in children. Pediatrics 1992;89:891894.Google Scholar
Barthez-Carpentier, MA, Rozenberg, F, Dussaix, E, et al. Relapse of herpes simplex encephalitis. J Child Neurol 1995;10:363368.Google Scholar
Ito, Y, Kimura, H, Yabuta, Y, et al. Exacerbation of herpes simplex encephalitis after successful treatment with acyclovir. Clin Infect Dis 2000;30:185187.Google Scholar
De Tiege, X, Rozenberg, F, Des Portes, V, et al. Herpes simplex encephalitis relapses in children: differentiation of two neurologic entities. Neurology 2003;61:241243.Google Scholar
Hacohen, Y, Deiva, K, Pettingill, P, et al. N-methyl-D-aspartate receptor antibodies in post-herpes simplex virus encephalitis neurological relapse. Mov Disord 2014;29:9096.Google Scholar
Mohammad, SS, Sinclair, K, Pillai, S, et al. Herpes simplex encephalitis relapse with chorea is associated with autoantibodies to N-methyl-D-aspartate receptor or dopamine-2 receptor. Mov Disord 2014;29:117122.Google Scholar
Petit-Pedrol, M, Armangue, T, Peng, X, et al. Encephalitis with refractory seizures, status epilepticus, and antibodies to the GABAA receptor: a case series, characterisation of the antigen, and analysis of the effects of antibodies. Lancet Neurol 2014;13:276286.Google Scholar
Armangue, T, Moris, G, Cantarin-Extremera, V, et al. Autoimmune post-herpes simplex encephalitis of adults and teenagers. Neurology 2015;85:17361743.Google Scholar
Armangue, T, Leypoldt, F, Malaga, I, et al. Herpes simplex virus encephalitis is a trigger of brain autoimmunity. Ann Neurol 2014;75:317323.Google Scholar
Lim, HK, Seppanen, M, Hautala, T, et al. TLR3 deficiency in herpes simplex encephalitis: high allelic heterogeneity and recurrence risk. Neurology 2014;83:18881897.Google Scholar
Lafaille, FG, Pessach, IM, Zhang, SY, et al. Impaired intrinsic immunity to HSV-1 in human iPSC-derived TLR3-deficient CNS cells. Nature 2012;491:769773.Google Scholar
Armangue, T, Baucells, BJ, Vlagea, A, et al. Toll-like receptor 3 deficiency in autoimmune encephalitis post-herpes simplex encephalitis. Neurol Neuroimmunol Neuroinflamm 2019;6:e611.Google Scholar

References

Marchi, N, Granata, T, Janigro, D. Inflammatory pathways of seizure disorders. Trends Neurosci 2014;37:5565.Google Scholar
Korff, CM, Dale, RC. The immune system in pediatric seizures and epilepsies. Pediatrics 2017;140:e20163534.Google Scholar
van Vliet, EA, Aronica, E, Vezzani, A, Ravizza, T. Review: neuroinflammatory pathways as treatment targets and biomarker candidates in epilepsy – emerging evidence from preclinical and clinical studies. Neuropathol Appl Neurobiol 2018;44:91111.Google Scholar
Vezzani, A, Dingledine, R, Rossetti, AO. Immunity and inflammation in status epilepticus and its sequelae: possibilities for therapeutic application. Expert Rev Neurother 2015;15:10811092.Google Scholar
Geis, C, Planaguma, J, Carreno, M, Graus, F, Dalmau, J. Autoimmune seizures and epilepsy. J Clin Invest 2019;129:926940.Google Scholar
Kim, SY, Senatorov, VV Jr., Morrissey, CS, et al. TGFbeta signaling is associated with changes in inflammatory gene expression and perineuronal net degradation around inhibitory neurons following various neurological insults. Sci Rep 2017;7:7711.Google Scholar
Vezzani, A, Balosso, S, Ravizza, T. Neuroinflammatory pathways as treatment targets and biomarkers in epilepsy. Nat Rev Neurol 2019;15:459472.Google Scholar
Yeshokumar, AK, Coughlin, A, Fastman, J, et al. Seizures in autoimmune encephalitis-A systematic review and quantitative synthesis. Epilepsia 2021;62:397407.Google Scholar
Sillevis, SP, Kinoshita, A, De, LB, et al. Paraneoplastic cerebellar ataxia due to autoantibodies against a glutamate receptor. N Engl J Med 2000;342:2127.Google Scholar
de Graaff, E, Maat, P, Hulsenboom, E, et al. Identification of Delta/Notch-like epidermal growth factor-related receptor as the Tr antigen in paraneoplastic cerebellar degeneration. Ann Neurol 2012;71:815824.Google Scholar
Sabater, L, Gaig, C, Gelpi, E, et al. A novel non-rapid-eye movement and rapid-eye-movement parasomnia with sleep breathing disorder associated with antibodies to IgLON5: a case series, characterisation of the antigen, and post-mortem study. Lancet Neurol 2014;13:575586.Google Scholar
Gaig, C, Graus, F, Compta, Y, et al. Clinical manifestations of the anti-IgLON5 disease. Neurology 2017;88:17361743.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.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
Darnell, RB, Posner, JB. Paraneoplastic syndromes involving the nervous system. N Engl J Med 2003;349:15431554.Google Scholar
Peltola, J, Kulmala, P, Isojarvi, J, et al. Autoantibodies to glutamic acid decarboxylase in patients with therapy-resistant epilepsy. Neurology 2000;55:4650.Google Scholar
Giometto, B, Nicolao, P, Macucci, M, et al. Temporal-lobe epilepsy associated with glutamic-acid-decarboxylase autoantibodies. Lancet 1998;352:457.Google Scholar
Bien, CG, Granata, T, Antozzi, C, et al. Pathogenesis, diagnosis and treatment of Rasmussen encephalitis: a European consensus statement. Brain 2005;128:454471.Google Scholar
Iizuka, T, Kanazawa, N, Kaneko, J, et al. Cryptogenic NORSE: its distinctive clinical features and response to immunotherapy. Neurol Neuroimmunol Neuroinflamm 2017;4:e396.Google Scholar
Gaspard, N, Foreman, BP, Alvarez, V, et al. New-onset refractory status epilepticus: etiology, clinical features, and outcome. Neurology 2015;85:16041613.Google Scholar
Zhou, JY, Xu, B, Lopes, J, Blamoun, J, Li, L. Hashimoto encephalopathy: literature review. Acta Neurol Scand 2017;135:285290.Google Scholar
Alink, J, de Vries, TW. Unexplained seizures, confusion or hallucinations: think Hashimoto encephalopathy. Acta Paediatr 2008;97:451453.Google Scholar
Jarius, S, Paul, F, Aktas, O, et al. MOG encephalomyelitis: international recommendations on diagnosis and antibody testing. J Neuroinflammation 2018;15:134.Google Scholar
Hamid, SHM, Whittam, D, Saviour, M, et al. Seizures and encephalitis in myelin oligodendrocyte glycoprotein IgG disease vs aquaporin 4 IgG disease. JAMA Neurol 2018;75:6571.Google Scholar
Dubey, D, Hinson, SR, Jolliffe, EA, et al. Autoimmune GFAP astrocytopathy: prospective evaluation of 90 patients in 1 year. J Neuroimmunol 2018;321:157163.Google Scholar
de Bruijn, M, van Sonderen, A, van Coevorden-Hameete, MH, et al. Evaluation of seizure treatment in anti-LGI1, anti-NMDAR, and anti-GABABR encephalitis. Neurology 2019;92:e2185e2196.Google Scholar
Irani, SR, Stagg, CJ, Schott, JM, et al. Faciobrachial dystonic seizures: the influence of immunotherapy on seizure control and prevention of cognitive impairment in a broadening phenotype. Brain 2013;136:31513162.Google Scholar
Jeffery, OJ, Lennon, VA, Pittock, SJ, et al. GABAB receptor autoantibody frequency in service serologic evaluation. Neurology 2013;81:882887.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
Spatola, M, Petit-Pedrol, M, Simabukuro, MM, et al. Investigations in GABAA receptor antibody-associated encephalitis. Neurology 2017;88:10121020.Google Scholar
Petit-Pedrol, M, Armangue, T, Peng, X, et al. Encephalitis with refractory seizures, status epilepticus, and antibodies to the GABAA receptor: a case series, characterisation of the antigen, and analysis of the effects of antibodies. Lancet Neurol 2014;13:276286.Google Scholar
Titulaer, MJ, McCracken, L, Gabilondo, I, et al. Treatment and prognostic factors for long-term outcome in patients with anti-NMDA receptor encephalitis: an observational cohort study. Lancet Neurol 2013;12:157165.Google Scholar
Schmitt, SE, Pargeon, K, Frechette, ES, et al. Extreme delta brush: a unique EEG pattern in adults with anti-NMDA receptor encephalitis. Neurology 2012;79:10941100.Google Scholar
Sonderen, AV, Arends, S, Tavy, DLJ, et al. Predictive value of electroencephalography in anti-NMDA receptor encephalitis. J Neurol Neurosurg Psychiatry 2018;89:11011106.Google Scholar
Aurangzeb, S, Symmonds, M, Knight, RK, et al. LGI1-antibody encephalitis is characterised by frequent, multifocal clinical and subclinical seizures. Seizure 2017;50:1417.Google Scholar
Steriade, C, Mirsattari, SM, Murray, BJ, Wennberg, R. Subclinical temporal EEG seizure pattern in LGI1-antibody-mediated encephalitis. Epilepsia 2016;57:e155160.Google Scholar
van Sonderen, A, Thijs, RD, Coenders, EC, et al. Anti-LGI1 encephalitis: clinical syndrome and long-term follow-up. Neurology 2016;87:14491456.Google Scholar
Arino, H, Armangue, T, Petit-Pedrol, M, et al. Anti-LGI1-associated cognitive impairment: presentation and long-term outcome. Neurology 2016;87:759765.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
Lai, M, Hughes, EG, Peng, X, et al. AMPA receptor antibodies in limbic encephalitis alter synaptic receptor location. Ann Neurol 2009;65:424434.CrossRefGoogle ScholarPubMed
van Sonderen, A, Arino, H, Petit-Pedrol, M, et al. The clinical spectrum of Caspr2 antibody-associated disease. Neurology 2016;87:521528.CrossRefGoogle ScholarPubMed
Joubert, B, Saint-Martin, M, Noraz, N, et al. Characterization of a subtype of autoimmune encephalitis with anti-contactin-associated protein-like 2 antibodies in the cerebrospinal fluid, prominent limbic symptoms, and seizures. JAMA Neurol 2016;73:11151124.CrossRefGoogle ScholarPubMed
van Sonderen, A, Schreurs, MW, de Bruijn, MA, et al. The relevance of VGKC positivity in the absence of LGI1 and Caspr2 antibodies. Neurology 2016;86:16921699.Google Scholar
Spatola, M, Sabater, L, Planaguma, J, et al. Encephalitis with mGluR5 antibodies: symptoms and antibody effects. Neurology 2018;90:e1964e1972.Google Scholar
Dale, RC, Merheb, V, Pillai, S, et al. Antibodies to surface dopamine-2 receptor in autoimmune movement and psychiatric disorders. Brain 2012;135:34533468.CrossRefGoogle ScholarPubMed
Hara, M, Arino, H, Petit-Pedrol, M, et al. DPPX antibody-associated encephalitis: main syndrome and antibody effects. Neurology 2017;88:13401348.Google Scholar
Tobin, WO, Lennon, VA, Komorowski, L, et al. DPPX potassium channel antibody: frequency, clinical accompaniments, and outcomes in 20 patients. Neurology 2014;83:17971803.Google Scholar
Carvajal-Gonzalez, A, Leite, MI, Waters, P, et al. Glycine receptor antibodies in PERM and related syndromes: characteristics, clinical features and outcomes. Brain 2014;137:21782192.Google Scholar
Schuler, V, Luscher, C, Blanchet, C, et al. Epilepsy, hyperalgesia, impaired memory, and loss of pre- and postsynaptic GABA(B) responses in mice lacking GABA(B(1)). Neuron 2001;31:4758.Google Scholar
Prosser, HM, Gill, CH, Hirst, WD, et al. Epileptogenesis and enhanced prepulse inhibition in GABA(B1)-deficient mice. Mol Cell Neurosci 2001;17:10591070.Google Scholar
Waldmeier, PC, Kaupmann, K, Urwyler, S. Roles of GABAB receptor subtypes in presynaptic auto- and heteroreceptor function regulating GABA and glutamate release. J Neural Transm (Vienna) 2008;115:14011411.Google Scholar
Ohkawa, T, Satake, S, Yokoi, N, et al. Identification and characterization of GABA(A) receptor autoantibodies in autoimmune encephalitis. J Neurosci 2014;34:81518163.Google Scholar
Hirose, S. Mutant GABA(A) receptor subunits in genetic (idiopathic) epilepsy. Prog Brain Res 2014;213:5585.CrossRefGoogle Scholar
Planaguma, J, Leypoldt, F, Mannara, F, et al. Human N-methyl D-aspartate receptor antibodies alter memory and behaviour in mice. Brain 2015;138:94109.Google Scholar
Planaguma, J, Haselmann, H, Mannara, F, et al. Ephrin-B2 prevents N-methyl-D-aspartate receptor antibody effects on memory and neuroplasticity. Ann Neurol 2016;80:388400.Google Scholar
Ladepeche, L, Planaguma, J, Thakur, S, et al. NMDA receptor autoantibodies in autoimmune encephalitis cause a subunit-specific nanoscale redistribution of NMDA receptors. Cell Rep 2018;23:37593768.Google Scholar
Mikasova, L, De Rossi, P, Bouchet, D, et al. Disrupted surface cross-talk between NMDA and Ephrin-B2 receptors in anti-NMDA encephalitis. Brain 2012;135:16061621.Google Scholar
Wright, S, Hashemi, K, Stasiak, L, et al. Epileptogenic effects of NMDAR antibodies in a passive transfer mouse model. Brain 2015;138:31593167.CrossRefGoogle Scholar
Forrest, D, Yuzaki, M, Soares, HD, et al. Targeted disruption of NMDA receptor 1 gene abolishes NMDA response and results in neonatal death. Neuron 1994;13:325338.CrossRefGoogle ScholarPubMed
Nakazawa, K, Quirk, MC, Chitwood, RA, et al. Requirement for hippocampal CA3 NMDA receptors in associative memory recall. Science 2002;297:211218.Google Scholar
Shimizu, E, Tang, YP, Rampon, C, Tsien, JZ. NMDA receptor-dependent synaptic reinforcement as a crucial process for memory consolidation. Science 2000;290:11701174.CrossRefGoogle ScholarPubMed
Korotkova, T, Fuchs, EC, Ponomarenko, A, von Engelhardt, J, Monyer, H. NMDA receptor ablation on parvalbumin-positive interneurons impairs hippocampal synchrony, spatial representations, and working memory. Neuron 2010;68:557569.Google Scholar
Ohkawa, T, Fukata, Y, Yamasaki, M, et al. Autoantibodies to epilepsy-related LGI1 in limbic encephalitis neutralize LGI1–ADAM22 interaction and reduce synaptic AMPA receptors. J Neurosci 2013;33:1816118174.Google Scholar
Petit-Pedrol, M, Sell, J, Planaguma, J, et al. LGI1 antibodies alter Kv1.1 and AMPA receptors changing synaptic excitability, plasticity and memory. Brain 2018;141:31443159.Google Scholar
Fukata, Y, Lovero, KL, Iwanaga, T, et al. Disruption of LGI1-linked synaptic complex causes abnormal synaptic transmission and epilepsy. Proc Natl Acad Sci USA 2010;107:37993804.Google Scholar
Zhou, YD, Lee, S, Jin, Z, et al. Arrested maturation of excitatory synapses in autosomal dominant lateral temporal lobe epilepsy. Nat Med 2009;15:12081214.Google Scholar
Fukata, Y, Adesnik, H, Iwanaga, T, et al. Epilepsy-related ligand/receptor complex LGI1 and ADAM22 regulate synaptic transmission. Science 2006;313:17921795.Google Scholar
Seagar, M, Russier, M, Caillard, O, et al. LGI1 tunes intrinsic excitability by regulating the density of axonal Kv1 channels. Proc Natl Acad Sci US 2017;114:77197724.Google Scholar
Boillot, M, Lee, CY, Allene, C, et al. LGI1 acts presynaptically to regulate excitatory synaptic transmission during early postnatal development. Sci Rep 2016;6:21769.Google Scholar
Gleichman, AJ, Panzer, JA, Baumann, BH, Dalmau, J, Lynch, DR. Antigenic and mechanistic characterization of anti-AMPA receptor encephalitis. Ann Clin Transl Neurol 2014;1:180189.Google Scholar
Peng, X, Hughes, EG, Moscato, EH, et al. Cellular plasticity induced by anti-alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor encephalitis antibodies. Ann Neurol 2015;77:381398.Google Scholar
Haselmann, H, Mannara, F, Werner, C, et al. Human autoantibodies against the AMPA receptor subunit GluA2 induce receptor reorganization and memory dysfunction. Neuron 2018;100:91105.Google Scholar
Shimshek, DR, Jensen, V, Celikel, T, et al. Forebrain-specific glutamate receptor B deletion impairs spatial memory but not hippocampal field long-term potentiation. J Neurosci 2006;26:84288440.Google Scholar
Shimshek, DR, Bus, T, Kim, J, et al. Enhanced odor discrimination and impaired olfactory memory by spatially controlled switch of AMPA receptors. PLoS Biol 2005;3:e354.Google Scholar
Lu, W, Shi, Y, Jackson, AC, et al. Subunit composition of synaptic AMPA receptors revealed by a single-cell genetic approach. Neuron 2009;62:254268.Google Scholar
Pinatel, D, Hivert, B, Boucraut, J, et al. Inhibitory axons are targeted in hippocampal cell culture by anti-Caspr2 autoantibodies associated with limbic encephalitis. Front Cell Neurosci 2015;9:265.Google Scholar
Poliak, S, Salomon, D, Elhanany, H, et al. Juxtaparanodal clustering of Shaker-like K+ channels in myelinated axons depends on Caspr2 and TAG-1. J Cell Biol 2003;162:11491160.Google Scholar
Horresh, I, Poliak, S, Grant, S, et al. Multiple molecular interactions determine the clustering of Caspr2 and Kv1 channels in myelinated axons. J Neurosci 2008;28:1421314222.Google Scholar
Jia, Z, Lu, Y, Henderson, J, et al. Selective abolition of the NMDA component of long-term potentiation in mice lacking mGluR5. Learn Mem 1998;5:331343.Google Scholar
Lu, YM, Jia, Z, Janus, C, et al. Mice lacking metabotropic glutamate receptor 5 show impaired learning and reduced CA1 long-term potentiation (LTP) but normal CA3 LTP. J Neurosci 1997;17:51965205.Google Scholar
Witkin, JM, Baez, M, Yu, J, Eiler, WJ, 2nd. mGlu5 receptor deletion does not confer seizure protection to mice. Life Sci 2008;83:377380.Google Scholar
Guo, W, Molinaro, G, Collins, KA, et al. Selective disruption of metabotropic glutamate receptor 5-homer interactions mimics phenotypes of fragile X syndrome in mice. J Neurosci 2016;36:21312147.Google Scholar
Sun, W, Maffie, JK, Lin, L, et al. DPP6 establishes the A-type K(+) current gradient critical for the regulation of dendritic excitability in CA1 hippocampal neurons. Neuron 2011;71:11021115.Google Scholar
Lin, L, Murphy, JG, Karlsson, RM, et al. DPP6 loss impacts hippocampal synaptic development and induces behavioral impairments in recognition, learning and memory. Front Cell Neurosci 2018;12:84.Google Scholar
Crisp, SJ, Dixon, CL, Jacobson, L, et al. Glycine receptor autoantibodies disrupt inhibitory neurotransmission. Brain 2019;142:33983410.Google Scholar
Shiang, R, Ryan, SG, Zhu, YZ, et al. Mutations in the alpha 1 subunit of the inhibitory glycine receptor cause the dominant neurologic disorder, hyperekplexia. Nat Genet 1993;5:351358.Google Scholar
Buckwalter, MS, Cook, SA, Davisson, MT, White, WF, Camper, SA. A frameshift mutation in the mouse alpha 1 glycine receptor gene (Glra1) results in progressive neurological symptoms and juvenile death. Hum Mol Genet 1994;3:20252030.Google Scholar
Findlay, GS, Phelan, R, Roberts, MT, et al. Glycine receptor knock-in mice and hyperekplexia-like phenotypes: comparisons with the null mutant. J Neurosci 2003;23:80518059.Google Scholar
Gresa-Arribas, N, Planaguma, J, Petit-Pedrol, M, et al. Human neurexin-3alpha antibodies associate with encephalitis and alter synapse development. Neurology 2016;86:22352242.Google Scholar
Missler, M, Zhang, W, Rohlmann, A, et al. Alpha-neurexins couple Ca2+ channels to synaptic vesicle exocytosis. Nature 2003;423:939948.Google Scholar
Aoto, J, Foldy, C, Ilcus, SM, Tabuchi, K, Sudhof, TC. Distinct circuit-dependent functions of presynaptic neurexin-3 at GABAergic and glutamatergic synapses. Nat Neurosci 2015;18:9971007.Google Scholar
Yao, L, Yue, W, Xunyi, W, et al. Clinical features and long-term outcomes of seizures associated with autoimmune encephalitis: a follow-up study in East China. J Clin Neurosci 2019;68:7379.Google Scholar
Lancaster, E, Lai, M, Peng, X, et al. Antibodies to the GABA(B) receptor in limbic encephalitis with seizures: case series and characterisation of the antigen. Lancet Neurol 2010;9:6776.Google Scholar
Zhao, XH, Yang, X, Liu, XW, Wang, SJ. Clinical features and outcomes of Chinese patients with anti-gamma-aminobutyric acid B receptor encephalitis. Exp Ther Med 2020;20:617622.Google Scholar
Dogan Onugoren, M, Deuretzbacher, D, Haensch, CA, et al. Limbic encephalitis due to GABAB and AMPA receptor antibodies: a case series. J Neurol Neurosurg Psychiatry 2015;86:965972.Google Scholar
Maureille, A, Fenouil, T, Joubert, B, et al. Isolated seizures are a common early feature of paraneoplastic anti-GABAB receptor encephalitis. J Neurol 2019;266:195206.Google Scholar
Chen, X, Liu, F, Li, JM, et al. Encephalitis with antibodies against the GABAB receptor: seizures as the most common presentation at admission. Neurol Res 2017;39:973980.Google Scholar
Cui, J, Bu, H, He, J, et al. The gamma-aminobutyric acid-B receptor (GABAB) encephalitis: clinical manifestations and response to immunotherapy. Int J Neurosci 2018;128:627633.Google Scholar
McKay, JH, Dimberg, EL, Lopez Chiriboga, AS. A systematic review of gamma-aminobutyric acid receptor type B autoimmunity. Neurol Neurochir Pol 2019;53:17.Google Scholar
Boronat, A, Sabater, L, Saiz, A, Dalmau, J, Graus, F. GABAB receptor antibodies in limbic encephalitis and anti-GAD-associated neurologic disorders. Neurology 2011;76:795800.Google Scholar
Kruer, MC, Hoeftberger, R, Lim, KY, et al. Aggressive course in encephalitis with opsoclonus, ataxia, chorea, and seizures: the first pediatric case of gamma-aminobutyric acid type B receptor autoimmunity. JAMA Neurol 2014;71:620623.Google Scholar
Guasp, M, Landa, J, Martinez-Hernandez, E, et al. Thymoma and autoimmune encephalitis: clinical manifestations and antibodies. Neurol Neuroimmunol Neuroinflamm 2021;8:e1053.Google Scholar
Pettingill, P, Kramer, HB, Coebergh, JA, et al. Antibodies to GABAA receptor alpha1 and gamma2 subunits: clinical and serologic characterization. Neurology 2015;84:12331241.Google Scholar
Armangue, T, Olive-Cirera, G, Martinez-Hernandez, E, et al. Associations of paediatric demyelinating and encephalitic syndromes with myelin oligodendrocyte glycoprotein antibodies: a multicentre observational study. Lancet Neurol 2020;19:234246.Google Scholar
O’Connor, K, Waters, P, Komorowski, L, et al. GABAA receptor autoimmunity: a multicenter experience. Neurol Neuroimmunol Neuroinflamm 2019;6:e552.Google Scholar
Simabukuro, MM, Petit-Pedrol, M, Castro, LH, et al. GABAA receptor and LGI1 antibody encephalitis in a patient with thymoma. Neurol Neuroimmunol Neuroinflamm 2015;2:e73.Google Scholar
Figlerowicz, M, Kemnitz, P, Mania, A, et al. Autoimmune encephalitis with GABAA receptor antibodies in a 10-year-old girl. Clin Neurol Neurosurg 2018;164:160163.Google Scholar
Florance, NR, Davis, RL, Lam, C, et al. Anti-N-methyl-D-aspartate receptor (NMDAR) encephalitis in children and adolescents. Ann Neurol 2009;66:1118.Google Scholar
Armangue, T, Titulaer, MJ, Malaga, I, et al. Pediatric anti-N-methyl-D-aspartate receptor encephalitis: clinical analysis and novel findings in a series of 20 patients. J Pediatr 2013;162:850856.Google Scholar
Qu, XP, Vidaurre, J, Peng, XL, et al. Seizure characteristics, outcome, and risk of epilepsy in pediatric anti-N-methyl-D-aspartate receptor encephalitis. Pediatr Neurol 2020;105:3540.Google Scholar
Chavez-Castillo, M, Ruiz-Garcia, M, Herrera-Mora, P. Characterization and outcomes of epileptic seizures in Mexican pediatric patients with anti-N-methyl-D-aspartate receptor encephalitis. Cureus 2020;12:e8211.Google Scholar
Irani, SR, Bera, K, Waters, P, et al. N-methyl-D-aspartate antibody encephalitis: temporal progression of clinical and paraclinical observations in a predominantly non-paraneoplastic disorder of both sexes. Brain 2010;133:16551667.Google Scholar
Dalmau, J, Gleichman, AJ, Hughes, EG, et al. Anti-NMDA-receptor encephalitis: case series and analysis of the effects of antibodies. Lancet Neurol 2008;7:10911098.Google Scholar
Viaccoz, A, Desestret, V, Ducray, F, et al. Clinical specificities of adult male patients with NMDA receptor antibodies encephalitis. Neurology 2014;82:556563.Google Scholar
Gofshteyn, JS, Yeshokumar, AK, Jette, N, et al. Clinical and electrographic features of persistent seizures and status epilepticus associated with anti-NMDA receptor encephalitis (anti-NMDARE). Epileptic Disord 2020;22:739751.Google Scholar
Liu, X, Yan, B, Wang, R, et al. Seizure outcomes in patients with anti-NMDAR encephalitis: a follow-up study. Epilepsia 2017;58:21042111.Google Scholar
Haberlandt, E, Ensslen, M, Gruber-Sedlmayr, U, et al. Epileptic phenotypes, electroclinical features and clinical characteristics in 17 children with anti-NMDAR encephalitis. Eur J Paediatr Neurol 2017;21:457464.Google Scholar
Favier, M, Joubert, B, Picard, G, et al. Initial clinical presentation of young children with N-methyl-D-aspartate receptor encephalitis. Eur J Paediatr Neurol 2017;22:404411.Google Scholar
Ariño, H, Muñoz-Lopetegi, A, Martinez-Hernandez, E, et al. Sleep disorders in anti-NMDAR encephalitis. Neurology 2020;95:e671e684.Google Scholar
Munoz-Lopetegi, A, Graus, F, Dalmau, J, Santamaria, J. Sleep disorders in autoimmune encephalitis. Lancet Neurol 2020;19:10101022.Google Scholar
Gillinder, L, Warren, N, Hartel, G, Dionisio, S, O’Gorman, C. EEG findings in NMDA encephalitis: a systematic review. Seizure 2019;65:2024.Google Scholar
Veciana, M, Becerra, JL, Fossas, P, et al. EEG extreme delta brush: an ictal pattern in patients with anti-NMDA receptor encephalitis. Epilepsy Behav 2015;49:280285.Google Scholar
da Silva-Junior, FP, Castro, LH, Andrade, JQ, et al. Serial and prolonged EEG monitoring in anti-N-methyl-D-aspartate receptor encephalitis. Clin Neurophysiol 2014;125:15411544.Google Scholar
Tan, YL, Tan, K, Tan, NC. Antiepileptic treatment for anti-NMDA receptor encephalitis: the need for video-EEG monitoring. Epileptic Disord 2013;15:468.Google Scholar
Howard, CM, Kass, JS, Bandi, VDP, Guntupalli, KK. Challenges in providing critical care for patients with anti-N-methyl-D-aspartate receptor encephalitis. Chest 2014;145:11431147.Google Scholar
Chanson, E, Bicilli, E, Lauxerois, M, et al. Anti-NMDA-R encephalitis: should we consider extreme delta brush as electrical status epilepticus? Neurophysiol Clin 2016;46:1725.Google Scholar
Jeannin-Mayer, S, Andre-Obadia, N, Rosenberg, S, et al. EEG analysis in anti-NMDA receptor encephalitis: description of typical patterns. Clin Neurophysiol 2019;130:289296.Google Scholar
Sands, TT, Nash, K, Tong, S, Sullivan, J. Focal seizures in children with anti-NMDA receptor antibody encephalitis. Epilepsy Res 2015;112:3136.Google Scholar
Shen, CH, Fang, GL, Yang, F, et al. Seizures and risk of epilepsy in anti-NMDAR, anti-LGI1, and anti-GABAB R encephalitis. Ann Clin Transl Neurol 2020;7:13921399.Google Scholar
Santoro, JD, Filippakis, A, Chitnis, T. Ketamine use in refractory status epilepticus associated with anti-NMDA receptor antibody encephalitis. Epilepsy Behav Rep 2019;12:100326.Google Scholar
Millichap, JJ, Goldstein, JL, Laux, LC, et al. Ictal asystole and anti-N-methyl-D-aspartate receptor antibody encephalitis. Pediatrics 2011;127:e781e786.Google Scholar
Lee, M, Lawn, N, Prentice, D, Chan, J. Anti-NMDA receptor encephalitis associated with ictal asystole. J Clin Neurosci 2011;18:17161718.Google Scholar
Sansing, LH, Tuzun, E, Ko, MW, et al. A patient with encephalitis associated with NMDA receptor antibodies. Nat Clin Pract Neurol 2007;3:291296.Google Scholar
Kumar, MA, Jain, A, Dechant, VE, et al. Anti-N-methyl-D-aspartate receptor encephalitis during pregnancy. Arch Neurol 2010;67:884887.Google Scholar
de Montmollin, E, Demeret, S, Brule, N, et al. Anti-N-methyl-D-aspartate receptor encephalitis in adult patients requiring intensive care. Am J Respir Crit Care Med 2017;195:491499.Google Scholar
Mehr, SR, Neeley, RC, Wiley, M, Kumar, AB. Profound autonomic instability complicated by multiple episodes of cardiac asystole and refractory bradycardia in a patient with anti-NMDA encephalitis. Case Rep Neurol Med 2016;2016:7967526.Google Scholar
Fisher, RS, Acevedo, C, Arzimanoglou, A, et al. ILAE official report: a practical clinical definition of epilepsy. Epilepsia 2014;55:475482.Google Scholar
Zhang, W, Wang, X, Shao, N, Ma, R, Meng, H. Seizure characteristics, treatment, and outcome in autoimmune synaptic encephalitis: a long-term study. Epilepsy Behav 2019;94:198203.Google Scholar
Gabilondo, I, Saiz, A, Galan, L, et al. Analysis of relapses in anti-NMDAR encephalitis. Neurology 2011;77:996999.Google Scholar
Armangue, T, Spatola, M, Vlagea, A, et al. Frequency, symptoms, risk factors, and outcomes of autoimmune encephalitis after herpes simplex encephalitis: a prospective observational study and retrospective analysis. Lancet Neurol 2018;17:760772.Google Scholar
Paolicchi, JM. The timing of pediatric epilepsy syndromes: what are the developmental triggers? Ann N Y Acad Sci 2013;1304:4551.Google Scholar
Aznar Lain, G, Dellatolas, G, Eisermann, M, et al. Children often present with infantile spasms after herpetic encephalitis. Epilepsia 2013;54:15711576.Google Scholar
Yildirim, M, Konuskan, B, Yalnizoglu, D, et al. Electroencephalographic findings in anti-N-methyl-D-aspartate receptor encephalitis in children: a series of 12 patients. Epilepsy Behav 2018;78:118123.Google Scholar
Zhang, Y, Liu, G, Jiang, MD, Li, LP, Su, YY. Analysis of electroencephalogram characteristics of anti-NMDA receptor encephalitis patients in China. Clin Neurophysiol 2017;128:12271233.Google Scholar
Mohammad, SS, Soe, SM, Pillai, SC, et al. Etiological associations and outcome predictors of acute electroencephalography in childhood encephalitis. Clin Neurophysiol 2016;127:32173224.Google Scholar
Gibbs, EL, Gibbs, FA. Extreme spindles: correlation of electroencephalographic sleep pattern with mental retardation. Science 1962;138:11061107.Google Scholar
Kirkpatrick, MP, Clarke, CD, Sonmezturk, HH, Abou-Khalil, B. Rhythmic delta activity represents a form of nonconvulsive status epilepticus in anti-NMDA receptor antibody encephalitis. Epilepsy Behav 2011;20:392394.Google Scholar
Probasco, JC, Benavides, DR, Ciarallo, A, et al. Electroencephalographic and fluorodeoxyglucose-positron emission tomography correlates in anti-N-methyl-D-aspartate receptor autoimmune encephalitis. Epilepsy Behav Case Rep 2014;2:174178.Google Scholar
Zhang, Y, Llinas, RR, Lisman, JE. Inhibition of NMDARs in the nucleus reticularis of the thalamus produces delta frequency bursting. Front Neural Circuits 2009;3:20.Google Scholar
Gataullina, S, Plouin, P, Vincent, A, et al. Paroxysmal EEG pattern in a child with N-methyl-D-aspartate receptor antibody encephalitis. Dev Med Child Neurol 2011;53:764767.Google Scholar
Ikeda, A, Matsui, M, Hase, Y, et al. ‘Burst and slow complexes’ in nonconvulsive epileptic status. Epileptic Disord 2006;8:6164.Google Scholar
Clancy, RR, Bergqvist, AGC, Dlugos, DJ. Neonatal electroencephalography. In: Ebersole, JS, Pedley, TA, eds. Current practice of clinical electroencephalography, 3rd ed. Philadelphia, PA: Lippincott Williams & Wilkins, 2003: 160234.Google Scholar
Foff, EP, Taplinger, D, Suski, J, Lopes, MB, Quigg, M. EEG findings may serve as a potential biomarker for anti-NMDA receptor encephalitis. Clin EEG Neurosci 2017;48:4853.Google Scholar
Sankaranarayanan, M, Shah, S, Thomas, P, Kannoth, S, Radhakrishnan, K. Persistent extreme delta brush in anti-NMDA-receptor encephalitis: does it portend a poor prognosis? Epilepsy Behav Rep 2019;12:100324.Google Scholar
Baykan, B, Gungor Tuncer, O, Vanli-Yavuz, EN, et al. Delta brush pattern is not unique to NMDAR encephalitis: evaluation of two independent long-term EEG cohorts. Clin EEG Neurosci 2018;49:278284.Google Scholar
Heresco-Levy, U, Durrant, AR, Ermilov, M, et al. Clinical and electrophysiological effects of D-serine in a schizophrenia patient positive for anti-N-methyl-D-aspartate receptor antibodies. Biol Psychiatry 2015;77:e2729.Google Scholar
Gitiaux, C, Simonnet, H, Eisermann, M, et al. Early electro-clinical features may contribute to diagnosis of the anti-NMDA receptor encephalitis in children. Clin Neurophysiol 2013;124:23542361.Google Scholar
Brenton, JN, Kim, J, Schwartz, RH. Approach to the management of pediatric-onset anti-N-methyl-D-aspartate (anti-NMDA) receptor encephalitis: a case series. J Child Neurol 2016;31:11501155.Google Scholar
Lai, M, Huijbers, MG, Lancaster, E, et al. Investigation of LGI1 as the antigen in limbic encephalitis previously attributed to potassium channels: a case series. Lancet Neurol 2010;9:776785.Google Scholar
van Sonderen, A, Petit-Pedrol, M, Dalmau, J, Titulaer, MJ. The value of LGI1, Caspr2 and voltage-gated potassium channel antibodies in encephalitis. Nat Rev Neurol 2017;13:290301.Google Scholar
Irani, SR, Alexander, S, Waters, P, et al. Antibodies to Kv1 potassium channel-complex proteins leucine-rich, glioma inactivated 1 protein and contactin-associated protein-2 in limbic encephalitis, Morvan’s syndrome and acquired neuromyotonia. Brain 2010;133:27342748.Google Scholar
Gadoth, A, Pittock, SJ, Dubey, D, et al. Expanded phenotypes and outcomes among 256 LGI1/CASPR2-IgG-positive patients. Ann Neurol 2017;82:7992.Google Scholar
Andrade, DM, Tai, P, Dalmau, J, Wennberg, R. Tonic seizures: a diagnostic clue of anti-LGI1 encephalitis? Neurology 2011;76:13551357.Google Scholar
Wennberg, R, Steriade, C, Chen, R, Andrade, D. Frontal infraslow activity marks the motor spasms of anti-LGI1 encephalitis. Clin Neurophysiol 2018;129:5968.Google Scholar
Navarro, V, Kas, A, Apartis, E, et al. Motor cortex and hippocampus are the two main cortical targets in LGI1-antibody encephalitis. Brain 2016;139:10791093.Google Scholar
Irani, SR, Michell, AW, Lang, B, et al. Faciobrachial dystonic seizures precede Lgi1 antibody limbic encephalitis. Ann Neurol 2011;69:892900.Google Scholar
Vincent, A, Buckley, C, Schott, JM, et al. Potassium channel antibody-associated encephalopathy: a potentially immunotherapy-responsive form of limbic encephalitis. Brain 2004;127:701712.Google Scholar
Borusiak, P, Bettendorf, U, Wiegand, G, et al. Autoantibodies to neuronal antigens in children with focal epilepsy and no prima facie signs of encephalitis. Eur J Paediatr Neurol 2016;20:573579.Google Scholar
Suleiman, J, Brenner, T, Gill, D, et al. VGKC antibodies in pediatric encephalitis presenting with status epilepticus. Neurology 2011;76:12521255.Google Scholar
Suleiman, J, Brilot, F, Lang, B, Vincent, A, Dale, RC. Autoimmune epilepsy in children: case series and proposed guidelines for identification. Epilepsia 2013;54:10361045.Google Scholar
Wright, S, Geerts, AT, Jol-van der Zijde, CM, et al. Neuronal antibodies in pediatric epilepsy: clinical features and long-term outcomes of a historical cohort not treated with immunotherapy. Epilepsia 2016;57:823831.Google Scholar
Hacohen, Y, Singh, R, Rossi, M, et al. Clinical relevance of voltage-gated potassium channel-complex antibodies in children. Neurology 2015;85:967975.Google Scholar
Lopez-Chiriboga, AS, Klein, C, Zekeridou, A, et al. LGI1 and CASPR2 neurological autoimmunity in children. Ann Neurol 2018;84:473480.Google Scholar
Beimer, NJ, Selwa, LM. Seizure semiology of anti-LGI1 antibody encephalitis. Epileptic Disord 2017;19:461464.Google Scholar
Thompson, J, Bi, M, Murchison, AG, et al. The importance of early immunotherapy in patients with faciobrachial dystonic seizures. Brain 2018;141:348356.Google Scholar
Lopez Chiriboga, AS, Siegel, JL, Tatum, WO, Shih, JJ, Flanagan, EP. Striking basal ganglia imaging abnormalities in LGI1 ab faciobrachial dystonic seizures. Neurol Neuroimmunol Neuroinflamm 2017;4:e336.Google Scholar
Flanagan, EP, Kotsenas, AL, Britton, JW, et al. Basal ganglia T1 hyperintensity in LGI1-autoantibody faciobrachial dystonic seizures. Neurol Neuroimmunol Neuroinflamm 2015;2:e161.Google Scholar
Naasan, G, Irani, SR, Bettcher, BM, Geschwind, MD, Gelfand, JM. Episodic bradycardia as neurocardiac prodrome to voltage-gated potassium channel complex/leucine-rich, glioma inactivated 1 antibody encephalitis. JAMA Neurol 2014;71:13001304.Google Scholar
Rachdi, A, Dupouy, J, Benaiteau, M, et al. Leucine-rich glioma-inactivated 1 encephalitis: broadening the sphere. Tremor Other Hyperkinet Mov (N Y) 2019;9.Google Scholar
Chen, C, Wang, X, Zhang, C, et al. Seizure semiology in leucine-rich glioma-inactivated protein 1 antibody-associated limbic encephalitis. Epilepsy Behav 2017;77:9095.Google Scholar
Finke, C, Pruss, H, Heine, J, et al. Evaluation of cognitive deficits and structural hippocampal damage in encephalitis with leucine-rich, glioma-inactivated 1 antibodies. JAMA Neurol 2017;74:5059.Google Scholar
Shin, YW, Lee, ST, Shin, JW, et al. VGKC-complex/LGI1-antibody encephalitis: clinical manifestations and response to immunotherapy. J Neuroimmunol 2013;265:7581.Google Scholar
Liu, X, Han, Y, Yang, L, et al. The exploration of the spectrum of motor manifestations of anti-LGI1 encephalitis beyond FBDS. Seizure 2020;76:2227.Google Scholar
Iranzo, A, Graus, F, Clover, L, et al. Rapid eye movement sleep behavior disorder and potassium channel antibody-associated limbic encephalitis. Ann Neurol 2006;59:178181.CrossRefGoogle ScholarPubMed
Irani, SR, Gelfand, JM, Bettcher, BM, Singhal, NS, Geschwind, MD. Effect of rituximab in patients with leucine-rich, glioma-inactivated 1 antibody-associated encephalopathy. JAMA Neurol 2014;71:896900.Google Scholar
Smith, KM, Dubey, D, Liebo, GB, Flanagan, EP, Britton, JW. Clinical course and features of seizures associated with LGI1-antibody encephalitis. Neurology 2021;97:e1141–e1149.Google Scholar
Striano, P, Belcastro, V, Striano, S. Tonic seizures: a diagnostic clue of anti-LGI1 encephalitis? Neurology 2011;77:21412143.Google Scholar
Chatrian, GE, Shaw, CM, Plum, F. Focal periodic slow transients in epilepsia partialis continua: clinical and pathological correlations in two cases. Electroencephalogr Clin Neurophysiol 1964;16:387393.Google Scholar
Oga, T, Ikeda, A, Nagamine, T, et al. Implication of sensorimotor integration in the generation of periodic dystonic myoclonus in subacute sclerosing panencephalitis (SSPE). Mov Disord 2000;15:11731183.Google Scholar
Hirsch, LJ, Gaspard, N, van Baalen, A, et al. Proposed consensus definitions for new-onset refractory status epilepticus (NORSE), febrile infection-related epilepsy syndrome (FIRES), and related conditions. Epilepsia 2018;59:739744.Google Scholar
Nabbout, R. FIRES and IHHE: delineation of the syndromes. Epilepsia 2013;54(Suppl. 6):5456.Google Scholar
van Baalen, A, Hausler, M, Plecko-Startinig, B, et al. Febrile infection-related epilepsy syndrome without detectable autoantibodies and response to immunotherapy: a case series and discussion of epileptogenesis in FIRES. Neuropediatrics 2012;43:209216.Google Scholar
Lang, B, Makuch, M, Moloney, T, et al. Intracellular and non-neuronal targets of voltage-gated potassium channel complex antibodies. J Neurol Neurosurg Psychiatry 2017;88:353361.Google Scholar
Graus, F, Gorman, MP. Voltage-gated potassium channel antibodies: game over. Neurology 2016;86:16571658.Google Scholar
Barros, P, Brito, H, Ferreira, PC, et al. Resective surgery in the treatment of super-refractory partial status epilepticus secondary to NMDAR antibody encephalitis. Eur J Paediatr Neurol 2014;18:449452.Google Scholar
Kaplan, PW, Probasco, J. Limbic and new onset refractory tonic status epilepticus (NORSE) in anti-NMDAR encephalitis. Clin Neurophysiol Pract 2017;2:140143.Google Scholar
Monti, G, Giovannini, G, Marudi, A, et al. Anti-NMDA receptor encephalitis presenting as new onset refractory status epilepticus in COVID-19. Seizure 2020;81:1820.Google Scholar
Sculier, C, Gaspard, N. New onset refractory status epilepticus (NORSE). Seizure 2019;68:7278.Google Scholar
Hainsworth, JB, Shishido, A, Theeler, BJ, Carroll, CG, Fasano, RE. Treatment responsive GABA(B)-receptor limbic encephalitis presenting as new-onset super-refractory status epilepticus (NORSE) in a deployed U.S. soldier. Epileptic Disord 2014;16:486493.Google Scholar
Budhram, A, Mirian, A, Le, C, et al. Unilateral cortical FLAIR-hyperintense Lesions in Anti-MOG-associated Encephalitis with Seizures (FLAMES): characterization of a distinct clinico-radiographic syndrome. J Neurol 2019;266:24812487.Google Scholar
Specchio, N, Pietrafusa, N. New-onset refractory status epilepticus and febrile infection-related epilepsy syndrome. Dev Med Child Neurol 2020;62:897905.Google Scholar
Kothur, K, Bandodkar, S, Wienholt, L, et al. Etiology is the key determinant of neuroinflammation in epilepsy: elevation of cerebrospinal fluid cytokines and chemokines in febrile infection-related epilepsy syndrome and febrile status epilepticus. Epilepsia 2019;60:16781688.Google Scholar
Howell, KB, Katanyuwong, K, Mackay, MT, et al. Long-term follow-up of febrile infection-related epilepsy syndrome. Epilepsia 2012;53:101110.Google Scholar

References

Reindl, M, Waters, P. Myelin oligodendrocyte glycoprotein antibodies in neurological disease. Nat Rev Neurol 2019;15:89102.Google Scholar
Jarius, S, Wandinger, KP, Borowski, K, Stoecker, W, Wildemann, B. Antibodies to CV2/CRMP5 in neuromyelitis optica-like disease: case report and review of the literature. Clin Neurol Neurosurg 2012;114:331335.Google Scholar
Matute, C, Palma, A, Serrano-Regal, MP, et al. N-methyl-D-aspartate receptor antibodies in autoimmune encephalopathy alter oligodendrocyte function. Ann Neurol 2020;87:670676.Google Scholar
Spadaro, M, Winklmeier, S, Beltran, E, et al. Pathogenicity of human antibodies against myelin oligodendrocyte glycoprotein. Ann Neurol 2018;84:315328.Google Scholar
Hillebrand, S, Schanda, K, Nigritinou, M, et al. Circulating AQP4-specific auto-antibodies alone can induce neuromyelitis optica spectrum disorder in the rat. Acta Neuropathol 2019;137:467485.Google Scholar
Wingerchuk, DM, Banwell, B, Bennett, JL, et al. International consensus diagnostic criteria for neuromyelitis optica spectrum disorders. Neurology 2015;85:177189.Google Scholar
Dubey, D, Pittock, SJ, Kelly, CR, et al. Autoimmune encephalitis epidemiology and a comparison to infectious encephalitis. Ann Neurol 2018;83:166177.Google Scholar
Britton, PN, Dale, RC, Blyth, CC, et al. Causes and clinical features of childhood encephalitis: a multicenter, prospective cohort study. Clin Infect Dis 2020;70:25172526.Google Scholar
Krupp, LB, Banwell, B, Tenembaum, S. Consensus definitions proposed for pediatric multiple sclerosis and related disorders. Neurology 2007;68:S712.Google Scholar
Cole, J, Evans, E, Mwangi, M, Mar, S. Acute disseminated encephalomyelitis in children: an updated review based on current diagnostic criteria. Pediatr Neurol 2019;100:2634.Google Scholar
Pohl, D, Alper, G, Van Haren, K, et al. Acute disseminated encephalomyelitis: updates on an inflammatory CNS syndrome. Neurology 2016;87:S38S45.Google Scholar
Koelman, DL, Mateen, FJ. Acute disseminated encephalomyelitis: current controversies in diagnosis and outcome. J Neurol 2015;262:20132024.Google Scholar
Van Bogaert, L. Post-infectious encephalomyelitis and multiple sclerosis: the significance of perivenous encephalomyelitis. J Neuropathol Exp Neurol 1950;9:219249.Google Scholar
Torisu, H, Kira, R, Ishizaki, Y, et al. Clinical study of childhood acute disseminated encephalomyelitis, multiple sclerosis, and acute transverse myelitis in Fukuoka Prefecture, Japan. Brain Dev 2010;32:454462.Google Scholar
Banwell, B, Kennedy, J, Sadovnick, D, et al. Incidence of acquired demyelination of the CNS in Canadian children. Neurology 2009;72:232239.Google Scholar
Leake, JA, Albani, S, Kao, AS, et al. Acute disseminated encephalomyelitis in childhood: epidemiologic, clinical and laboratory features. Pediatr Infect Dis J 2004;23:756764.Google Scholar
Pohl, D, Hennemuth, I, von Kries, R, Hanefeld, F. Paediatric multiple sclerosis and acute disseminated encephalomyelitis in Germany: results of a nationwide survey. Eur J Pediatr 2007;166:405412.Google Scholar
Yamaguchi, Y, Torisu, H, Kira, R, et al. A nationwide survey of pediatric acquired demyelinating syndromes in Japan. Neurology 2016;87:20062015.Google Scholar
Otallah, S. Acute disseminated encephalomyelitis in children and adults: a focused review emphasizing new developments. Mult Scler 2021:27:11531160.Google Scholar
Chen, Y, Ma, F, Xu, Y, Chu, X, Zhang, J. Incidence of acute disseminated encephalomyelitis in the Jiangsu province of China, 2008–2011. Mult Scler J Exp Transl Clin 2015;1:2055217315594831.Google Scholar
Koelman, DL, Chahin, S, Mar, SS, et al. Acute disseminated encephalomyelitis in 228 patients: a retrospective, multicenter US study. Neurology 2016;86:20852093.Google Scholar
Dale, RC, de Sousa, C, Chong, WK, et al. Acute disseminated encephalomyelitis, multiphasic disseminated encephalomyelitis and multiple sclerosis in children. Brain 2000;123:24072422.Google Scholar
Sejvar, JJ, Kohl, KS, Bilynsky, R, et al. Encephalitis, myelitis, and acute disseminated encephalomyelitis (ADEM): case definitions and guidelines for collection, analysis, and presentation of immunization safety data. Vaccine 2007;25:57715792.Google Scholar
Novi, G, Rossi, T, Pedemonte, E, et al. Acute disseminated encephalomyelitis after SARS-CoV-2 infection. Neurol Neuroimmunol Neuroinflamm 2020;7:e797.Google Scholar
Parsons, T, Banks, S, Bae, C, et al. COVID-19-associated acute disseminated encephalomyelitis (ADEM). J Neurol 2020:267:27992802.Google Scholar
Karussis, D, Petrou, P. The spectrum of post-vaccination inflammatory CNS demyelinating syndromes. Autoimmun Rev 2014;13:215224.Google Scholar
Chen, Y, Ma, F, Xu, Y, Chu, X, Zhang, J. Vaccines and the risk of acute disseminated encephalomyelitis. Vaccine 2018;36:37333739.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
Erol, I, Ozkale, Y, Alkan, O, Alehan, F. Acute disseminated encephalomyelitis in children and adolescents: a single center experience. Pediatr Neurol 2013;49:266273.Google Scholar
Ketelslegers, IA, Visser, IE, Neuteboom, RF, et al. Disease course and outcome of acute disseminated encephalomyelitis is more severe in adults than in children. Mult Scler 2011;17:441448.Google Scholar
Koelman, DL, Benkeser, DC, Xu, Y, et al. Acute disseminated encephalomyelitis in China, Singapore and Japan: a comparison with the USA. Eur J Neurol 2017;24:391396.Google Scholar
Boesen, MS, Blinkenberg, M, Koch-Henriksen, N, et al. Implications of the International Paediatric Multiple Sclerosis Study Group consensus criteria for paediatric acute disseminated encephalomyelitis: a nationwide validation study. Dev Med Child Neurol 2018;60:11231131.Google Scholar
Panicker, JN, Nagaraja, D, Kovoor, JM, Subbakrishna, DK. Descriptive study of acute disseminated encephalomyelitis and evaluation of functional outcome predictors. J Postgrad Med 2010;56:1216.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
Alper, G, Heyman, R, Wang, L. Multiple sclerosis and acute disseminated encephalomyelitis diagnosed in children after long-term follow-up: comparison of presenting features. Dev Med Child Neurol 2009;51:480486.Google Scholar
Fridinger, SE, Alper, G. Defining encephalopathy in acute disseminated encephalomyelitis. J Child Neurol 2014;29:751755.Google Scholar
Tenembaum, S, Chamoles, N, Fejerman, N. Acute disseminated encephalomyelitis: a long-term follow-up study of 84 pediatric patients. Neurology 2002;59:12241231.Google Scholar
Lim, KE, Hsu, YY, Hsu, WC, Chan, CY. Multiple complete ring-shaped enhanced MRI lesions in acute disseminated encephalomyelitis. Clinical imaging 2003;27:281284.Google Scholar
Tenembaum, S, Chitnis, T, Ness, J, Hahn, JS. Acute disseminated encephalomyelitis. Neurology 2007;68:S23S36.Google Scholar
Zhang, L, Wu, A, Zhang, B, et al. Comparison of deep gray matter lesions on magnetic resonance imaging among adults with acute disseminated encephalomyelitis, multiple sclerosis, and neuromyelitis optica. Mult Scler 2014;20:418423.Google Scholar
Lu, Z, Zhang, B, Qiu, W, et al. Comparative brain stem lesions on MRI of acute disseminated encephalomyelitis, neuromyelitis optica, and multiple sclerosis. PLoS One 2011;6:e22766.Google Scholar
Wong, YYM, van Pelt, ED, Ketelslegers, IA, et al. Evolution of MRI abnormalities in paediatric acute disseminated encephalomyelitis. Eur J Paediatr Neurol 2017;21:300304.Google Scholar
Brilot, F, Dale, RC, Selter, RC, et al. Antibodies to native myelin oligodendrocyte glycoprotein in children with inflammatory demyelinating central nervous system disease. Ann Neurol 2009;66:833842.Google Scholar
Baumann, M, Sahin, K, Lechner, C, et al. Clinical and neuroradiological differences of paediatric acute disseminating encephalomyelitis with and without antibodies to the myelin oligodendrocyte glycoprotein. J Neurol Neurosurg Psychiatry 2015;86:265272.Google Scholar
Greenlee, JE. Encephalitis and postinfectious encephalitis. Continuum (Minneapolis, Minn) 2012;18:12711289.Google Scholar
Hart, MN, Earle, KM. Haemorrhagic and perivenous encephalitis: a clinical-pathological review of 38 cases. J Neurol Neurosurg Psychiatry 1975;38:585591.Google Scholar
Young, NP, Weinshenker, BG, Parisi, JE, et al. Perivenous demyelination: association with clinically defined acute disseminated encephalomyelitis and comparison with pathologically confirmed multiple sclerosis. Brain 2010;133:333348.Google Scholar
Waters, P, Fadda, G, Woodhall, M, et al. Serial anti-myelin oligodendrocyte glycoprotein antibody analyses and outcomes in children with demyelinating syndromes. JAMA Neurol 2020;77:8293.Google Scholar
Kennedy, PG. Viral encephalitis: causes, differential diagnosis, and management. J Neurol Neurosurg Psychiatry 2004;75(Suppl. 1):i10i15.Google Scholar
Titulaer, MJ, Hoftberger, R, Iizuka, T, et al. Overlapping demyelinating syndromes and anti-N-methyl-D-aspartate receptor encephalitis. Ann Neurol 2014;75:411428.Google Scholar
Martinez-Hernandez, E, Guasp, M, Garcia-Serra, A, et al. Clinical significance of anti-NMDAR concurrent with glial or neuronal surface antibodies. Neurology 2020;94:e23022310.Google Scholar
Spatola, M, Petit-Pedrol, M, Simabukuro, MM, et al. Investigations in GABAa receptor antibody-associated encephalitis. Neurology 2017;88:10121020.Google Scholar
Dorr, J, Krautwald, S, Wildemann, B, et al. Characteristics of Susac syndrome: a review of all reported cases. Nat Rev Neurol 2013;9:307316.Google Scholar
Kleffner, I, Dorr, J, Ringelstein, M, et al. Diagnostic criteria for Susac syndrome. J Neurol Neurosurg Psychiatry 2016;87:12871295.Google Scholar
Siva, A, Saip, S. The spectrum of nervous system involvement in Behcet’s syndrome and its differential diagnosis. J Neurol 2009;256:513529.Google Scholar
Kocer, N, Islak, C, Siva, A, et al. CNS involvement in neuro-Behcet syndrome: an MR study. Am J Neuroradiol 1999;20:10151024.Google Scholar
Uygunoglu, U, Zeydan, B, Ozguler, Y, et al. Myelopathy in Behcet’s disease: the bagel sign. Ann Neurol 2017;82:288298.Google Scholar
Twilt, M, Benseler, SM. CNS vasculitis in children. Mult Scler Relat Disord 2013;2:162171.Google Scholar
Neuteboom, RF, Boon, M, Catsman Berrevoets, CE, et al. Prognostic factors after a first attack of inflammatory CNS demyelination in children. Neurology 2008;71:967973.Google Scholar
Banwell, B, Bar-Or, A, Arnold, DL, et al. Clinical, environmental, and genetic determinants of multiple sclerosis in children with acute demyelination: a prospective national cohort study. Lancet Neurol 2011;10:436445.Google Scholar
Banwell, B, Krupp, L, Kennedy, J, et al. Clinical features and viral serologies in children with multiple sclerosis: a multinational observational study. Lancet Neurol 2007;6:773781.Google Scholar
Peche, SS, Alshekhlee, A, Kelly, J, Lenox, J, Mar, S. A long-term follow-up study using IPMSSG criteria in children with CNS demyelination. Pediatr Neurol 2013;49:329334.Google Scholar
Mar, S, Lenox, J, Benzinger, T, Brown, S, Noetzel, M. Long-term prognosis of pediatric patients with relapsing acute disseminated encephalomyelitis. J Child Neurol 2010;25:681688.Google Scholar
Barkhof, F, Filippi, M, Miller, DH, et al. Comparison of MRI criteria at first presentation to predict conversion to clinically definite multiple sclerosis. Brain 1997;120:20592069.Google Scholar
Callen, DJ, Shroff, MM, Branson, HM, et al. Role of MRI in the differentiation of ADEM from MS in children. Neurology 2009;72:968973.Google Scholar
Verhey, LH, Branson, HM, Shroff, MM, et al. MRI parameters for prediction of multiple sclerosis diagnosis in children with acute CNS demyelination: a prospective national cohort study. Lancet Neurol 2011;10:10651073.Google Scholar
Fadda, G, Brown, RA, Longoni, G, et al. MRI and laboratory features and the performance of international criteria in the diagnosis of multiple sclerosis in children and adolescents: a prospective cohort study. Lancet Child Adolesc Health 2018;2:191204.Google Scholar
Pohl, D, Tenembaum, S. Treatment of acute disseminated encephalomyelitis. Curr Treat Opt Neurol 2012;14:264275.Google Scholar
Anlar, B, Basaran, C, Kose, G, et al. Acute disseminated encephalomyelitis in children: outcome and prognosis. Neuropediatrics 2003;34:194199.Google Scholar
Gadian, J, Kirk, E, Holliday, K, Lim, M, Absoud, M. Systematic review of immunoglobulin use in paediatric neurological and neurodevelopmental disorders. Dev Med Child Neurol 2017;59:136144.Google Scholar
Weiner, HL, Dau, PC, Khatri, BO, et al. Double-blind study of true vs. sham plasma exchange in patients treated with immunosuppression for acute attacks of multiple sclerosis. Neurology 1989;39:11431149.Google Scholar
Weinshenker, BG, O’Brien, PC, Petterson, TM, et al. A randomized trial of plasma exchange in acute central nervous system inflammatory demyelinating disease. Ann Neurol 1999;46:878886.Google Scholar
Absoud, M, Parslow, RC, Wassmer, E, et al. Severe acute disseminated encephalomyelitis: a paediatric intensive care population-based study. Mult Scler 2011;17:12581261.Google Scholar
Ahmed, AI, Eynon, CA, Kinton, L, Nicoll, JA, Belli, A. Decompressive craniectomy for acute disseminated encephalomyelitis. Neurocrit Care 2010;13:393395.Google Scholar
Dombrowski, KE, Mehta, AI, Turner, DA, McDonagh, DL. Life-saving hemicraniectomy for fulminant acute disseminated encephalomyelitis. Br J Neurosurg 2011;25:249252.Google Scholar
Granget, E, Milh, M, Pech-Gourg, G, et al. Life-saving decompressive craniectomy for acute disseminated encephalomyelitis in a child: a case report. Childs Nerv Syst 2012;28:11211124.Google Scholar
Nishiyama, M, Nagase, H, Tomioka, K, et al. Clinical time course of pediatric acute disseminated encephalomyelitis. Brain Dev 2019;41:531537.Google Scholar
Suppiej, A, Cainelli, E, Casara, G, et al. Long-term neurocognitive outcome and quality of life in pediatric acute disseminated encephalomyelitis. Pediatr Neurol 2014;50:363367.Google Scholar
Shilo, S, Michaeli, O, Shahar, E, Ravid, S. Long-term motor, cognitive and behavioral outcome of acute disseminated encephalomyelitis. Eur J Paediatr Neurol 2016;20:361367.Google Scholar
Burton, KLO, Williams, TA, Catchpoole, SE, Brunsdon, RK. Long-term neuropsychological outcomes of childhood onset acute disseminated encephalomyelitis (ADEM): a meta-analysis. Neuropsychol Rev 2017;27:124133.Google Scholar
Iype, M, Ts, A, Kunju, PM, et al. Factors related to long term motor, behavioral, and scholastic outcome in children with acute disseminated encephalomyelitis. Pediatr Neurol 2018;89:4957.Google Scholar
Rossor, T, Benetou, C, Wright, S, et al. Early predictors of epilepsy and subsequent relapse in children with acute disseminated encephalomyelitis. Mult Scler 2020;26:333342.Google Scholar
Kariyawasam, S, Singh, RR, Gadian, J, et al. Clinical and radiological features of recurrent demyelination following acute disseminated encephalomyelitis (ADEM). Mult Scler Relat Disord 2015;4:451456.Google Scholar
Wong, YYM, Hacohen, Y, Armangue, T, et al. Paediatric acute disseminated encephalomyelitis followed by optic neuritis: disease course, treatment response and outcome. Eur J Neurol 2018;25:782786.Google Scholar
Hacohen, Y, Absoud, M, Deiva, K, et al. Myelin oligodendrocyte glycoprotein antibodies are associated with a non-MS course in children. Neurol Neuroimmunol Neuroinflamm 2015;2:e81.Google Scholar
Reindl, M, Di Pauli, F, Rostasy, K, Berger, T. The spectrum of MOG autoantibody-associated demyelinating diseases. Nat Rev Neurol 2013;9:455461.Google Scholar
Ketelslegers, IA, Van Pelt, DE, Bryde, S, et al. Anti-MOG antibodies plead against MS diagnosis in an acquired demyelinating syndromes cohort. Mult Scler 2015;21:15131520.Google Scholar
Jurynczyk, M, Messina, S, Woodhall, MR, et al. Clinical presentation and prognosis in MOG-antibody disease: a UK study. Brain 2017;140:31283138.CrossRefGoogle ScholarPubMed
Hegen, H, Reindl, M. Recent developments in MOG-IgG associated neurological disorders. Therapeut Adv Neurologic Disord 2020;13:1756286420945135.Google Scholar
Lebar, R, Baudrimont, M, Vincent, C. Chronic experimental autoimmune encephalomyelitis in the guinea pig: presence of anti-M2 antibodies in central nervous system tissue and the possible role of M2 autoantigen in the induction of the disease. J Autoimmun 1989;2:115132.Google Scholar
Lebar, R, Lubetzki, C, Vincent, C, Lombrail, P, Boutry, JM. The M2 autoantigen of central nervous system myelin, a glycoprotein present in oligodendrocyte membrane. Clin Exp Immunol 1986;66:423434.Google Scholar
Linington, C, Bradl, M, Lassmann, H, Brunner, C, Vass, K. Augmentation of demyelination in rat acute allergic encephalomyelitis by circulating mouse monoclonal antibodies directed against a myelin/oligodendrocyte glycoprotein. Am J Pathol 1988;130:443454.Google Scholar
Kerlero de Rosbo, N, Mendel, I, Ben-Nun, A. Chronic relapsing experimental autoimmune encephalomyelitis with a delayed onset and an atypical clinical course, induced in PL/J mice by myelin oligodendrocyte glycoprotein (MOG)-derived peptide: preliminary analysis of MOG T cell epitopes. Eur J Immunol 1995;25:985993.Google Scholar
Lalive, PH, Molnarfi, N, Benkhoucha, M, Weber, MS, Santiago-Raber, ML. Antibody response in MOG (35–55) induced EAE. J Neuroimmunol 2011;240–241:2833.Google Scholar
Iglesias, A, Bauer, J, Litzenburger, T, Schubart, A, Linington, C. T- and B-cell responses to myelin oligodendrocyte glycoprotein in experimental autoimmune encephalomyelitis and multiple sclerosis. Glia 2001;36:220234.Google Scholar
Bourquin, C, Iglesias, A, Berger, T, Wekerle, H, Linington, C. Myelin oligodendrocyte glycoprotein-DNA vaccination induces antibody-mediated autoaggression in experimental autoimmune encephalomyelitis. Eur J Immunol 2000;30:36633671.Google Scholar
Brunner, C, Lassmann, H, Waehneldt, TV, Matthieu, JM, Linington, C. Differential ultrastructural localization of myelin basic protein, myelin/oligodendroglial glycoprotein, and 2′,3′-cyclic nucleotide 3′-phosphodiesterase in the CNS of adult rats. J Neurochem 1989;52:296304.Google Scholar
Reindl, M, Linington, C, Brehm, U, et al. Antibodies against the myelin oligodendrocyte glycoprotein and the myelin basic protein in multiple sclerosis and other neurological diseases: a comparative study. Brain 1999;122:20472056.Google Scholar
Berger, T, Rubner, P, Schautzer, F, et al. Antimyelin antibodies as a predictor of clinically definite multiple sclerosis after a first demyelinating event. N Engl J Med 2003;349:139145.Google Scholar
Karni, A, Bakimer-Kleiner, R, Abramsky, O, Ben-Nun, A. Elevated levels of antibody to myelin oligodendrocyte glycoprotein is not specific for patients with multiple sclerosis. Arch Neurol 1999;56:311315.Google Scholar
Mantegazza, R, Cristaldini, P, Bernasconi, P, et al. Anti-MOG autoantibodies in Italian multiple sclerosis patients: specificity, sensitivity and clinical association. Int Immunol 2004;16:559565.Google Scholar
von Budingen, HC, Hauser, SL, Fuhrmann, A, et al. Molecular characterization of antibody specificities against myelin/oligodendrocyte glycoprotein in autoimmune demyelination. Proc Natl Acad Sci USA 2002;99:82078212.Google Scholar
O’Connor, KC, McLaughlin, KA, De Jager, PL, et al. Self-antigen tetramers discriminate between myelin autoantibodies to native or denatured protein. Nat Med 2007;13:211217.Google Scholar
Jarius, S, Paul, F, Aktas, O, et al. MOG encephalomyelitis: international recommendations on diagnosis and antibody testing. J Neuroinflammation 2018;15:134.Google Scholar
Reindl, M, Schanda, K, Woodhall, M, et al. International multicenter examination of MOG antibody assays. Neurol Neuroimmunol Neuroinflamm 2020;7:e674.Google Scholar
Sepulveda, M, Armangue, T, Martinez-Hernandez, E, et al. Clinical spectrum associated with MOG autoimmunity in adults: significance of sharing rodent MOG epitopes. J Neurol 2016;263:13491360.Google Scholar
Jarius, S, Ruprecht, K, Kleiter, I, et al. MOG-IgG in NMO and related disorders: a multicenter study of 50 patients. Part 1: Frequency, syndrome specificity, influence of disease activity, long-term course, association with AQP4-IgG, and origin. J Neuroinflammation 2016;13:279.Google Scholar
Waters, P, Woodhall, M, O’Connor, KC, et al. MOG cell-based assay detects non-MS patients with inflammatory neurologic disease. Neurol Neuroimmunol Neuroinflamm 2015;2:e89.Google Scholar
Pedreno, M, Sepulveda, M, Armangue, T, et al. Frequency and relevance of IgM, and IgA antibodies against MOG in MOG-IgG-associated disease. Mult Scler Relat Disord 2019;28:230234.Google Scholar
Waters, PJ, Komorowski, L, Woodhall, M, et al. A multicenter comparison of MOG-IgG cell-based assays. Neurology 2019;92:e1250e1255.Google Scholar
Duignan, S, Wright, S, Rossor, T, et al. Myelin oligodendrocyte glycoprotein and aquaporin-4 antibodies are highly specific in children with acquired demyelinating syndromes. Dev Med Child Neurol 2018;60:958962.Google Scholar
Armangue, T, Olive-Cirera, G, Martinez-Hernandez, E, et al. Associations of paediatric demyelinating and encephalitic syndromes with myelin oligodendrocyte glycoprotein antibodies: a multicentre observational study. Lancet Neurol 2020;19:234246.Google Scholar
de Mol, CL, Wong, Y, van Pelt, ED, et al. The clinical spectrum and incidence of anti-MOG-associated acquired demyelinating syndromes in children and adults. Mult Scler 2020;26:806814.Google Scholar
O’Connell, K, Hamilton-Shield, A, Woodhall, M, et al. Prevalence and incidence of neuromyelitis optica spectrum disorder, aquaporin-4 antibody-positive NMOSD and MOG antibody-positive disease in Oxfordshire, UK. J Neurol Neurosurg Psychiatry 2020;91:11261128.Google Scholar
Hennes, EM, Baumann, M, Schanda, K, et al. Prognostic relevance of MOG antibodies in children with an acquired demyelinating syndrome. Neurology 2017;89:900908.Google Scholar
Cobo-Calvo, A, Ruiz, A, Maillart, E, et al. Clinical spectrum and prognostic value of CNS MOG autoimmunity in adults: the MOGADOR study. Neurology 2018;90:e1858e1869.Google Scholar
Kim, SM, Woodhall, MR, Kim, JS, et al. Antibodies to MOG in adults with inflammatory demyelinating disease of the CNS. Neurol Neuroimmunol Neuroinflamm 2015;2:e163.Google Scholar
Ogawa, R, Nakashima, I, Takahashi, T, et al. MOG antibody-positive, benign, unilateral, cerebral cortical encephalitis with epilepsy. Neurol Neuroimmunol Neuroinflamm 2017;4:e322.Google Scholar
Cobo-Calvo, A, Ayrignac, X, Kerschen, P, et al. Cranial nerve involvement in patients with MOG antibody-associated disease. Neurol Neuroimmunol Neuroinflamm 2019;6:e543.Google Scholar
Jarius, S, Kleiter, I, Ruprecht, K, et al. MOG-IgG in NMO and related disorders: a multicenter study of 50 patients. Part 3: Brainstem involvement – frequency, presentation and outcome. J Neuroinflammation 2016;13:281.Google Scholar
Katsuse, K, Kurihara, M, Sugiyama, Y, et al. Aphasic status epilepticus preceding tumefactive left hemisphere lesion in anti-MOG antibody associated disease. Mult Scler Relat Disord 2019;27:9194.Google Scholar
Miyaue, N, Yamanishi, Y, Tada, S, et al. A case of ADEM-like presentation with anti-MOG antibody following tumefactive demyelinating lesion. Mult Scler Relat Disord 2019;31:6264.Google Scholar
Berzero, G, Taieb, G, Marignier, R, et al. CLIPPERS mimickers: relapsing brainstem encephalitis associated with anti-MOG antibodies. Eur J Neurol 2018;25:e16e17.Google Scholar
Bruijstens, AL, Wong, YYM, van Pelt, DE, et al. HLA association in MOG-IgG- and AQP4-IgG-related disorders of the CNS in the Dutch population. Neurol Neuroimmunol Neuroinflamm 2020;7:e702.Google Scholar
Jarius, S, Ruprecht, K, Kleiter, I, et al. MOG-IgG in NMO and related disorders: a multicenter study of 50 patients. Part 2: Epidemiology, clinical presentation, radiological and laboratory features, treatment responses, and long-term outcome. J Neuroinflammation 2016;13:280.Google Scholar
Jurynczyk, M, Geraldes, R, Probert, F, et al. Distinct brain imaging characteristics of autoantibody-mediated CNS conditions and multiple sclerosis. Brain 2017;140:617627.Google Scholar
Jurynczyk, M, Tackley, G, Kong, Y, et al. Brain lesion distribution criteria distinguish MS from AQP4-antibody NMOSD and MOG-antibody disease. J Neurol Neurosurg Psychiatry 2017;88:132136.Google Scholar
Huppke, P, Rostasy, K, Karenfort, M, et al. Acute disseminated encephalomyelitis followed by recurrent or monophasic optic neuritis in pediatric patients. Mult Scler 2013;19:941946.Google Scholar
Baumann, M, Hennes, EM, Schanda, K, et al. Children with multiphasic disseminated encephalomyelitis and antibodies to the myelin oligodendrocyte glycoprotein (MOG): extending the spectrum of MOG antibody positive diseases. Mult Scler 2016;22:18211829.Google Scholar
Serra, M, Presicci, A, Fucci, M, et al. Acute disseminated encephalomyelitis followed by optic neuritis: a rare syndrome of uncertain treatment and prognosis. Neuropediatrics 2020;51:286291.Google Scholar
Biotti, D, Bonneville, F, Tournaire, E, et al. Optic neuritis in patients with anti-MOG antibodies spectrum disorder: MRI and clinical features from a large multicentric cohort in France. J Neurol 2017;264:21732175.Google Scholar
Chen, JJ, Flanagan, EP, Jitprapaikulsan, J, et al. Myelin oligodendrocyte glycoprotein antibody-positive optic neuritis: clinical characteristics, radiologic clues, and outcome. Am J Ophthalmol 2018;195:815.Google Scholar
Lee, HJ, Kim, B, Waters, P, et al. Chronic relapsing inflammatory optic neuropathy (CRION): a manifestation of myelin oligodendrocyte glycoprotein antibodies. J Neuroinflammation 2018;15:302.Google Scholar
Chalmoukou, K, Alexopoulos, H, Akrivou, S, et al. Anti-MOG antibodies are frequently associated with steroid-sensitive recurrent optic neuritis. Neurol Neuroimmunol Neuroinflamm 2015;2:e131.Google Scholar
Petzold, A, Plant, GT. Chronic relapsing inflammatory optic neuropathy: a systematic review of 122 cases reported. J Neurol 2014;261:1726.Google Scholar
Narayan, RN, McCreary, M, Conger, D, Wang, C, Greenberg, BM. Unique characteristics of optical coherence tomography (OCT) results and visual acuity testing in myelin oligodendrocyte glycoprotein (MOG) antibody positive pediatric patients. Mult Scler Relat Disord 2019;28:8690.Google Scholar
Shor, N, Aboab, J, Maillart, E, et al. Clinical, imaging and follow-up study of optic neuritis associated with myelin oligodendrocyte glycoprotein antibody: a multicentre study of 62 adult patients. Eur J Neurol 2020;27:384391.Google Scholar
Ramanathan, S, Prelog, K, Barnes, EH, et al. Radiological differentiation of optic neuritis with myelin oligodendrocyte glycoprotein antibodies, aquaporin-4 antibodies, and multiple sclerosis. Mult Scler 2016;22:470482.Google Scholar
Purvin, V, Kawasaki, A, Jacobson, DM. Optic perineuritis: clinical and radiographic features. Arch Ophthalmol 2001;119:12991306.Google Scholar
Jang, Y, Kim, SM, Yun, YI, et al. Comparison between optic neuritis associated with antibody against myelin oligodendrocyte glycoprotein and presumed idiopathic optic perineuritis. Neurol Sci 2020;41:27552760.Google Scholar
Ramanathan, S, Fraser, C, Curnow, SR, et al. Uveitis and optic perineuritis in the context of myelin oligodendrocyte glycoprotein antibody seropositivity. Eur J Neurol 2019;26:1137-e75.Google Scholar
Sepulveda, M, Aldea, M, Escudero, D, et al. Epidemiology of NMOSD in Catalonia: influence of the new 2015 criteria in incidence and prevalence estimates. Mult Scler 2018:24:18431851.Google Scholar
Sepulveda, M, Armangue, T, Sola-Valls, N, et al. Neuromyelitis optica spectrum disorders: comparison according to the phenotype and serostatus. Neurol Neuroimmunol Neuroinflamm 2016;3:e225.Google Scholar
Kitley, J, Waters, P, Woodhall, M, et al. Neuromyelitis optica spectrum disorders with aquaporin-4 and myelin-oligodendrocyte glycoprotein antibodies: a comparative study. JAMA Neurol 2014;71:276283.Google Scholar
Dubey, D, Pittock, SJ, Krecke, KN, et al. Clinical, radiologic, and prognostic features of myelitis associated with myelin oligodendrocyte glycoprotein autoantibody. JAMA Neurol 2019;76:301309.Google Scholar
Hacohen, Y, Nishimoto, Y, Fukami, Y, et al. Paediatric brainstem encephalitis associated with glial and neuronal autoantibodies. Dev Med Child Neurol 2016;58:836841.Google Scholar
Banks, SA, Morris, PP, Chen, JJ, et al. Brainstem and cerebellar involvement in MOG-IgG-associated disorder versus aquaporin-4-IgG and MS. J Neurol Neurosurg Psychiatry 2020. doi: 10.1136/jnnp-2020-325121.Google Scholar
Iorio, R, Lucchinetti, CF, Lennon, VA, et al. Intractable nausea and vomiting from autoantibodies against a brain water channel. Clin Gastroenterol Hepatol 2013;11:240245.Google Scholar
Kunchok, A, Krecke, KN, Flanagan, EP, et al. Does area postrema syndrome occur in myelin oligodendrocyte glycoprotein-IgG-associated disorders (MOGAD)? Neurology 2020;94:8588.Google Scholar
Durozard, P, Rico, A, Boutiere, C, et al. Comparison of the response to rituximab between myelin oligodendrocyte glycoprotein and aquaporin-4 antibody diseases. Ann Neurol 2020;87:256266.Google Scholar
Budhram, A, Mirian, A, Le, C, et al. Unilateral cortical FLAIR-hyperintense Lesions in Anti-MOG-associated Encephalitis with Seizures (FLAMES): characterization of a distinct clinico-radiographic syndrome. J Neurol 2019;266:24812487.Google Scholar
Hamid, SHM, Whittam, D, Saviour, M, et al. Seizures and encephalitis in myelin oligodendrocyte glycoprotein IgG disease vs aquaporin 4 IgG disease. JAMA Neurol 2018;75:6571.Google Scholar
Wang, L, ZhangBao, J, Zhou, L, et al. Encephalitis is an important clinical component of myelin oligodendrocyte glycoprotein antibody associated demyelination: a single-center cohort study in Shanghai, China. Eur J Neurol 2019;26:168174.Google Scholar
Takai, Y, Misu, T, Kaneko, K, et al. Myelin oligodendrocyte glycoprotein antibody-associated disease: an immunopathological study. Brain 2020;143:14311446.Google Scholar
Ikeda, T, Yamada, K, Ogawa, R, et al. The pathological features of MOG antibody-positive cerebral cortical encephalitis as a new spectrum associated with MOG antibodies: a case report. J Neurol Sci 2018;392:113115.Google Scholar
Salama, S, Khan, M, Pardo, S, Izbudak, I, Levy, M. MOG antibody-associated encephalomyelitis/encephalitis. Mult Scler 2019;25:14271433.Google Scholar
Wegener-Panzer, A, Cleaveland, R, Wendel, EM, et al. Clinical and imaging features of children with autoimmune encephalitis and MOG antibodies. Neurol Neuroimmunol Neuroinflamm 2020;7:e731.Google Scholar
Robinson, CP, Busl, KM. Neurologic manifestations of severe respiratory viral contagions. Crit Care Explor 2020;2:e0107.Google Scholar
Johns, TG, Bernard, CC. The structure and function of myelin oligodendrocyte glycoprotein. J Neurochem 1999;72:19.Google Scholar
Delarasse, C, Daubas, P, Mars, LT, et al. Myelin/oligodendrocyte glycoprotein-deficient (MOG-deficient) mice reveal lack of immune tolerance to MOG in wild-type mice. J Clin Invest 2003;112:544553.Google Scholar
Dale, RC, Tantsis, EM, Merheb, V, et al. Antibodies to MOG have a demyelination phenotype and affect oligodendrocyte cytoskeleton. Neurol Neuroimmunol Neuroinflamm 2014;1:e12.Google Scholar
Peschl, P, Schanda, K, Zeka, B, et al. Human antibodies against the myelin oligodendrocyte glycoprotein can cause complement-dependent demyelination. J Neuroinflammation 2017;14:208.Google Scholar
Wang, JJ, Jaunmuktane, Z, Mummery, C, et al. Inflammatory demyelination without astrocyte loss in MOG antibody-positive NMOSD. Neurology 2016;87:229231.Google Scholar
Kortvelyessy, P, Breu, M, Pawlitzki, M, et al. ADEM-like presentation, anti-MOG antibodies, and MS pathology: TWO case reports. Neurol Neuroimmunol Neuroinflamm 2017;4:e335.Google Scholar
Spadaro, M, Gerdes, LA, Mayer, MC, et al. Histopathology and clinical course of MOG-antibody-associated encephalomyelitis. Ann Clin Transl Neurol 2015;2:295301.Google Scholar
Lucchinetti, C, Bruck, W, Parisi, J, et al. Heterogeneity of multiple sclerosis lesions: implications for the pathogenesis of demyelination. Ann Neurol 2000;47:707717.Google Scholar
Höftberger, R, Guo, Y, Flanagan, EP, et al. The pathology of central nervous system inflammatory demyelinating disease accompanying myelin oligodendrocyte glycoprotein autoantibody. Acta Neuropathol 2020;139:875892.Google Scholar
Saadoun, S, Waters, P, Owens, GP, et al. Neuromyelitis optica MOG-IgG causes reversible lesions in mouse brain. Acta Neuropathol Commun 2014;2:35.Google Scholar
Kinzel, S, Weber, MS. The role of peripheral CNS-directed antibodies in promoting inflammatory CNS demyelination. Brain Sci 2017;7:70.Google Scholar
Kinzel, S, Lehmann-Horn, K, Torke, S, et al. Myelin-reactive antibodies initiate T cell-mediated CNS autoimmune disease by opsonization of endogenous antigen. Acta Neuropathol 2016;132:4358.Google Scholar
Bonnan, M, Valentino, R, Debeugny, S, et al. Short delay to initiate plasma exchange is the strongest predictor of outcome in severe attacks of NMO spectrum disorders. J Neurol Neurosurg Psychiatry 2018;89:346351.Google Scholar
Hacohen, Y, Banwell, B. Treatment approaches for MOG-Ab-associated demyelination in children. Curr Treat Opt Neurol 2019;21:2.Google Scholar
Ramanathan, S, Mohammad, S, Tantsis, E, et al. Clinical course, therapeutic responses and outcomes in relapsing MOG antibody-associated demyelination. J Neurol Neurosurg Psychiatry 2018;89:127137.Google Scholar
Hacohen, Y, Wong, YY, Lechner, C, et al. Disease course and treatment responses in children with relapsing myelin oligodendrocyte glycoprotein antibody-associated disease. JAMA Neurol 2018;75:478487.Google Scholar
Hyun, JW, Woodhall, MR, Kim, SH, et al. Longitudinal analysis of myelin oligodendrocyte glycoprotein antibodies in CNS inflammatory diseases. J Neurol Neurosurg Psychiatry 2017;88:811817.Google Scholar
Probstel, AK, Dornmair, K, Bittner, R, et al. Antibodies to MOG are transient in childhood acute disseminated encephalomyelitis. Neurology 2011;77:580588.Google Scholar
Oliveira, LM, Apóstolos-Pereira, SL, Pitombeira, MS et al. Persistent MOG-IgG positivity is a predictor of recurrence in MOG-IgG-associated optic neuritis, encephalitis and myelitis. Mult Scler 2019;25:19071914.Google Scholar
Lopez-Chiriboga, AS, Majed, M, Fryer, J, et al. Association of MOG-IgG serostatus with relapse after acute disseminated encephalomyelitis and proposed diagnostic criteria for MOG-IgG-associated disorders. JAMA Neurol 2018;75:13551363.Google Scholar
Li, S, Ren, H, Xu, Y, et al. Long-term efficacy of mycophenolate mofetil in myelin oligodendrocyte glycoprotein antibody-associated disorders: a prospective study. Neurol Neuroimmunol Neuroinflamm 2020;7:e705.Google Scholar
Chen, JJ, Flanagan, EP, Bhatti, MT, et al. Steroid-sparing maintenance immunotherapy for MOG-IgG associated disorder. Neurology 2020;95:e111e120.Google Scholar
Whittam, DH, Cobo-Calvo, A, Lopez-Chiriboga, AS, et al. Treatment of MOG-IgG-associated disorder with rituximab: an international study of 121 patients. Mult Scler Relat Disord 2020;44:102251.Google Scholar
Levy, M, Fujihara, K, Palace, J. New therapies for neuromyelitis optica spectrum disorder. Lancet Neurol 2021;20:6067.Google Scholar
Rigal, J, Pugnet, G, Ciron, J, Lépine, Z, Biotti, D. Off-label use of tocilizumab in neuromyelitis optica spectrum disorders and MOG-antibody-associated diseases: a case-series. Mult Scler Relat Disord 2020;46:102483.Google Scholar
Elsbernd, PM, Hoffman, WR, Carter, JL, Wingerchuk, DM. Interleukin-6 inhibition with tocilizumab for relapsing MOG-IgG associated disorder (MOGAD): a case-series and review. Mult Scler Relat Disord 2020;48:102696.Google Scholar
Phillips, OR, Joshi, SH, Narr, KL, et al. Superficial white matter damage in anti-NMDA receptor encephalitis. J Neurol Neurosurg Psychiatry 2018;89:518525.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
Bernal, F, Graus, F, Pifarre, A, et al. Immunohistochemical analysis of anti-Hu-associated paraneoplastic encephalomyelitis. Acta Neuropathol (Berl) 2002;103:509515.Google Scholar
Henson, RA, Hoffman, HL, Urich, H. Encephalomyelitis with carcinoma. Brain 1965;88:449464.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
Ricard, D, Rogemond, V, Charrier, E, et al. Isolation and expression pattern of human Unc-33-like phosphoprotein 6/collapsin response mediator protein 5 (Ulip6/CRMP5): coexistence with Ulip2/CRMP2 in Sema3a- sensitive oligodendrocytes. J Neurosci 2001;21:72037214.Google Scholar
Ricard, D, Stankoff, B, Bagnard, D, et al. Differential expression of collapsin response mediator proteins (CRMP/ULIP) in subsets of oligodendrocytes in the postnatal rodent brain. Molec Cell Neurosci 2000;16:324337.Google Scholar
Cohen, DA, Bhatti, MT, Pulido, JS, et al. Collapsin response-mediator protein 5-associated retinitis, vitritis, and optic disc edema. Ophthalmology 2020;127:221229.Google Scholar
Keegan, BM, Pittock, SJ, Lennon, VA. Autoimmune myelopathy associated with collapsin response-mediator protein-5 immunoglobulin G. Ann Neurol 2008;63:531534.Google Scholar
Ducray, F, Roos-Weil, R, Garcia, PY, et al. Devic’s syndrome-like phenotype associated with thymoma and anti-CV2/CRMP5 antibodies. J Neurol Neurosurg Psychiatry 2007;78:325327.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
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
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
Antoine, JC, Honnorat, J, Vocanson, C, et al. Posterior uveitis, paraneoplastic encephalomyelitis and auto- antibodies reacting with developmental protein of brain and retina. J Neurol Sci 1993;117:215223.Google Scholar
Margolin, E, Flint, A, Trobe, JD. High-titer collapsin response-mediating protein-associated (CRMP-5) paraneoplastic optic neuropathy and vitritis as the only clinical manifestations in a patient with small cell lung carcinoma. J Neuroophthalmol 2008;28:1722.Google Scholar
Morita, M, Fukuhara, T, Takahashi, H, Maemondo, M. Small cell lung cancer and progressive retinopathy. BMJ Case Rep 2014;2014:bcr2014205888.Google Scholar
Murphy, MA, Thirkill, CE, Hart, WM Jr. Paraneoplastic retinopathy: a novel autoantibody reaction associated with small-cell lung carcinoma. J Neuroophthalmol 1997;17:7783.Google Scholar
Luiz, JE, Lee, AG, Keltner, JL, Thirkill, CE, Lai, EC. Paraneoplastic optic neuropathy and autoantibody production in small-cell carcinoma of the lung. J Neuroophthalmol 1998;18:178181.Google Scholar
Dubey, D, Lennon, VA, Gadoth, A, et al. Autoimmune CRMP5 neuropathy phenotype and outcome defined from 105 cases. Neurology 2018;90:e103e110.Google Scholar
Camdessanche, JP, Antoine, JC, Honnorat, J, et al. Paraneoplastic peripheral neuropathy associated with anti-Hu antibodies: a clinical and electrophysiological study of 20 patients. Brain 2002;125:166175.Google Scholar
Moss, HE, Liu, GT, Dalmau, J. Glazed (vision) and confused. Survey Ophthalmol 2010;55:169173.Google Scholar

References

Parpura, V, Basarsky, TA, Liu, F, et al. Glutamate-mediated astrocyte-neuron signalling. Nature 1994;369:744747.Google Scholar
Bowman, CL, Kimelberg, HK. Excitatory amino acids directly depolarize rat brain astrocytes in primary culture. Nature 1984;311:656659.Google Scholar
Haydon, PG. Astrocytes and the modulation of sleep. Curr Opin Neurobiol 2017;44:2833.Google Scholar
Araque, A, Parpura, V, Sanzgiri, RP, Haydon, PG. Tripartite synapses: glia, the unacknowledged partner. Trends Neurosci 1999;22:208215.Google Scholar
Hasan, U, Singh, SK. The astrocyte–neuron interface: an overview on molecular and cellular dynamics controlling formation and maintenance of the tripartite synapse. Method Molec Biol (Clifton, NJ) 2019;1938:318.Google Scholar
Perea, G, Navarrete, M, Araque, A. Tripartite synapses: astrocytes process and control synaptic information. Trends Neurosci 2009;32:421431.Google Scholar
Simard, M, Arcuino, G, Takano, T, Liu, QS, Nedergaard, M. Signaling at the gliovascular interface. J Neurosci 2003;23:92549262.Google Scholar
Pfrieger, FW. Role of glial cells in the formation and maintenance of synapses. Brain Res Revs 2010;63:3946.Google Scholar
Iliff, JJ, Wang, M, Liao, Y, et al. A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid beta. Sci Transl Med 2012;4:147ra111.Google Scholar
Papouin, T, Dunphy, J, Tolman, M, Foley, JC, Haydon, PG. Astrocytic control of synaptic function. Philos Trans Roy Soc Lond B Biol Sci 2017;372:20160154.Google Scholar
Yao, X, Hrabetova, S, Nicholson, C, Manley, GT. Aquaporin-4-deficient mice have increased extracellular space without tortuosity change. J Neurosci 2008;28:54605464.Google Scholar
Ransohoff, RM, Engelhardt, B. The anatomical and cellular basis of immune surveillance in the central nervous system. Nat Rev Immunol 2012;12:623635.Google Scholar
Farina, C, Aloisi, F, Meinl, E. Astrocytes are active players in cerebral innate immunity. Trends Immunol 2007;28:138145.Google Scholar
Colombo, E, Farina, C. Astrocytes: key regulators of neuroinflammation. Trends Immunol 2016;37:608620.Google Scholar
Lennon, VA, Kryzer, TJ, Pittock, SJ, Verkman, AS, Hinson, SR. IgG marker of optic-spinal multiple sclerosis binds to the aquaporin-4 water channel. J Exp Med 2005;202:473477.Google Scholar
Wingerchuk, DM, Banwell, B, Bennett, JL, et al. International consensus diagnostic criteria for neuromyelitis optica spectrum disorders. Neurology 2015;85:177189.Google Scholar
Jarius, S, Wildemann, B. The history of neuromyelitis optica. J Neuroinflammation 2013;10:8.Google Scholar
Flanagan, EP, Hinson, SR, Lennon, VA, et al. Glial fibrillary acidic protein immunoglobulin G as biomarker of autoimmune astrocytopathy: analysis of 102 patients. Ann Neurol 2017;81:298309.Google Scholar
Saida, T. [Treatment of NMO]. Rinsho Shinkeigaku 2009;49:902905.Google Scholar
Lucchinetti, CF, Guo, Y, Popescu, BF, et al. The pathology of an autoimmune astrocytopathy: lessons learned from neuromyelitis optica. Brain Pathol 2014;24:8397.Google Scholar
Fang, B, McKeon, A, Hinson, SR, et al. Autoimmune glial fibrillary acidic protein astrocytopathy: a novel meningoencephalomyelitis. JAMA Neurol 2016;73:12971307.Google Scholar
Bradl, M, Lassmann, H. Experimental models of neuromyelitis optica. Brain Pathol 2014;24:7482.Google Scholar
Shu, Y, Long, Y, Chang, Y, et al. Brain immunohistopathology in a patient with autoimmune glial fibrillary acidic protein astrocytopathy. Neuroimmunomodulation 2018;25:16.Google Scholar
Long, Y, Liang, J, Xu, H, et al. Autoimmune glial fibrillary acidic protein astrocytopathy in Chinese patients: a retrospective study. Eur J Neurol 2018;25:477483.Google Scholar
Kuroiwa, Y, Shibasaki, H. Epidemiologic and clinical studies of multiple sclerosis in Japan. Neurology 1976;26:810.Google Scholar
O’Riordan, JI, Gallagher, HL, Thompson, AJ, et al. Clinical, CSF, and MRI findings in Devic’s neuromyelitis optica. J Neurol Neurosurg Psychiatry 1996;60:382387.Google Scholar
Wingerchuk, DM, Hogancamp, WF, O’Brien, PC, Weinshenker, BG. The clinical course of neuromyelitis optica (Devic’s syndrome). Neurology 1999;53:11071114.Google Scholar
Lennon, VA, Wingerchuk, DM, Kryzer, TJ, et al. A serum autoantibody marker of neuromyelitis optica: distinction from multiple sclerosis. Lancet 2004;364:21062112.Google Scholar
Wingerchuk, DM, Lennon, VA, Pittock, SJ, Lucchinetti, CF, Weinshenker, BG. Revised diagnostic criteria for neuromyelitis optica. Neurology 2006;66:14851489.Google Scholar
Sepulveda, M, Aldea, M, Escudero, D, et al. Epidemiology of NMOSD in Catalonia: influence of the new 2015 criteria in incidence and prevalence estimates. Mult Scler 2018;24:18431851.Google Scholar
Bukhari, W, Prain, KM, Waters, P, et al. Incidence and prevalence of NMOSD in Australia and New Zealand. J Neurol Neurosurg Psychiatry 2017;88:632638.Google Scholar
Papp, V, Illes, Z, Magyari, M, et al. Nationwide prevalence and incidence study of neuromyelitis optica spectrum disorder in Denmark. Neurology 2018;91:e2265e2275.Google Scholar
Papp, V, Iljicsov, A, Rajda, C, et al. A population-based epidemiological study of neuromyelitis optica spectrum disorder in Hungary. Eur J Neurol 2020;27:308317.Google Scholar
Kim, SH, Kim, W, Li, XF, Jung, IJ, Kim, HJ. Clinical spectrum of CNS aquaporin-4 autoimmunity. Neurology 2012;78:11791185.Google Scholar
Sepulveda, M, Armangue, T, Sola-Valls, N, et al. Neuromyelitis optica spectrum disorders: comparison according to the phenotype and serostatus. Neurol Neuroimmunol Neuroinflamm 2016;3:e225.Google Scholar
Wingerchuk, DM, Lennon, VA, Lucchinetti, CF, Pittock, SJ, Weinshenker, BG. The spectrum of neuromyelitis optica. Lancet Neurol 2007;6:805815.Google Scholar
Kim, SH, Mealy, MA, Levy, M, et al. Racial differences in neuromyelitis optica spectrum disorder. Neurology 2018;91:e2089e2099.Google Scholar
Flanagan, EP, Cabre, P, Weinshenker, BG, et al. Epidemiology of aquaporin-4 autoimmunity and neuromyelitis optica spectrum. Ann Neurol 2016;79:775783.Google Scholar
Hor, JY, Asgari, N, Nakashima, I, et al. Epidemiology of neuromyelitis optica spectrum disorder and its prevalence and incidence worldwide. Front Neurol 2020;11:501.Google Scholar
O’Connell, K, Hamilton-Shield, A, Woodhall, M, et al. Prevalence and incidence of neuromyelitis optica spectrum disorder, aquaporin-4 antibody-positive NMOSD and MOG antibody-positive disease in Oxfordshire, UK. J Neurol Neurosurg Psychiatry 2020;91:11261128.Google Scholar
Banwell, B, Tenembaum, S, Lennon, VA, et al. Neuromyelitis optica-IgG in childhood inflammatory demyelinating CNS disorders. Neurology 2008;70:344352.Google Scholar
McKeon, A, Lennon, VA, Lotze, T, et al. CNS aquaporin-4 autoimmunity in children. Neurology 2008;71:93100.Google Scholar
Collongues, N, Marignier, R, Zephir, H, et al. Long-term follow-up of neuromyelitis optica with a pediatric onset. Neurology 2010;75:10841088.Google Scholar
Shahmohammadi, S, Doosti, R, Shahmohammadi, A, et al. Autoimmune diseases associated with neuromyelitis optica spectrum disorders: a literature review. Mult Scler Relat Disord 2019;27:350363.Google Scholar
Iyer, A, Elsone, L, Appleton, R, Jacob, A. A review of the current literature and a guide to the early diagnosis of autoimmune disorders associated with neuromyelitis optica. Autoimmunity 2014;47:154161.Google 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
Titulaer, MJ, Hoftberger, R, Iizuka, T, et al. Overlapping demyelinating syndromes and anti-N-methyl-D-aspartate receptor encephalitis. Ann Neurol 2014;75:411428.Google Scholar
Martinez-Hernandez, E, Guasp, M, Garcia-Serra, A, et al. Clinical significance of anti-NMDAR concurrent with glial or neuronal surface antibodies. Neurology 2020;94:e2302e2310.Google Scholar
Asgari, N, Jarius, S, Laustrup, H, et al. Aquaporin-4-autoimmunity in patients with systemic lupus erythematosus: a predominantly population-based study. Mult Scler 2018;24:331339.Google Scholar
Katsumata, Y, Kawachi, I, Kawaguchi, Y, et al. Semiquantitative measurement of aquaporin-4 antibodies as a possible surrogate marker of neuromyelitis optica spectrum disorders with systemic autoimmune diseases. Mod Rheumatol 2012;22:676684.Google Scholar
Qiao, L, Wang, Q, Fei, Y, et al. The clinical characteristics of primary Sjogren’s syndrome with neuromyelitis optica spectrum disorder in China: a STROBE-compliant article. Medicine (Baltimore) 2015;94:e1145.Google Scholar
Piga, M, Chessa, E, Peltz, MT, et al. Demyelinating syndrome in SLE encompasses different subtypes: do we need new classification criteria? Pooled results from systematic literature review and monocentric cohort analysis. Autoimmunity Rev 2017;16:244252.Google Scholar
Chourkani, N, El Moutawakil, B, Sibai, M, et al. [Primary Sjogren’s syndrome and neuromyelitis optica]. La Revue de medecine interne 2010;31:e13e15.Google Scholar
Spillane, J, Christofi, G, Sidle, KC, Kullmann, DM, Howard, RS. Myasthenia gravis and neuromyelitis opica: a causal link. Mult Scler Relat Disord 2013;2:233237.Google Scholar
Jarius, S, Paul, F, Franciotta, D, et al. Neuromyelitis optica spectrum disorders in patients with myasthenia gravis: ten new aquaporin-4 antibody positive cases and a review of the literature. Mult Scler 2012;18:11351143.Google Scholar
McKeon, A, Lennon, VA, Jacob, A, et al. Coexistence of myasthenia gravis and serological markers of neurological autoimmunity in neuromyelitis optica. Muscle Nerve 2009;39:8790.Google Scholar
Leite, MI, Coutinho, E, Lana-Peixoto, M, et al. Myasthenia gravis and neuromyelitis optica spectrum disorder: a multicenter study of 16 patients. Neurology 2012;78:16011607.Google Scholar
Vaknin-Dembinsky, A, Abramsky, O, Petrou, P, et al. Myasthenia gravis-associated neuromyelitis optica-like disease: an immunological link between the central nervous system and muscle? Arch Neurol 2011;68:15571561.Google Scholar
Pittock, SJ, Lennon, VA. Aquaporin-4 autoantibodies in a paraneoplastic context. Arch Neurol 2008;65:629632.Google Scholar
Verschuur, CV, Kooi, AJ, Troost, D. Anti-aquaporin 4 related paraneoplastic neuromyelitis optica in the presence of adenocarcinoma of the lung. Clin Neuropathol 2015;34:232236.Google Scholar
Figueroa, M, Guo, Y, Tselis, A, et al. Paraneoplastic neuromyelitis optica spectrum disorder associated with metastatic carcinoid expressing aquaporin-4. JAMA Neurol 2014;71:495498.Google Scholar
Iorio, R, Rindi, G, Erra, C, et al. Neuromyelitis optica spectrum disorder as a paraneoplastic manifestation of lung adenocarcinoma expressing aquaporin-4. Mult Scler 2015;21:791794.Google Scholar
Ontaneda, D, Fox, RJ. Is neuromyelitis optica with advanced age of onset a paraneoplastic disorder? Int J Neurosci 2014;124:509511.Google Scholar
Flanagan, EP, McKeon, A, Lennon, VA, et al. Paraneoplastic isolated myelopathy: clinical course and neuroimaging clues. Neurology 2011;76:20892095.Google Scholar
Jarius, S, Wandinger, KP, Borowski, K, Stoecker, W, Wildemann, B. Antibodies to CV2/CRMP5 in neuromyelitis optica-like disease: case report and review of the literature. Clin Neurol Neurosurg 2012;114:331335.Google Scholar
Cai, G, He, D, Chu, L, et al. Paraneoplastic neuromyelitis optica spectrum disorders: three new cases and a review of the literature. Int J Neurosci 2016;126:660668.Google Scholar
Al-Harbi, T, Al-Sarawi, A, Binfalah, M, Dermime, S. Paraneoplastic neuromyelitis optica spectrum disorder associated with stomach carcinoid tumor. Hematol Oncol Stem Cell Ther 2014;7:116119.Google Scholar
Nakayama-Ichiyama, S, Yokote, T, Hiraoka, N, et al. A paraneoplastic neuromyelitis optica spectrum disorder associated with a mature B-cell neoplasm. Leukemia Res 2011;35:e111113.Google Scholar
Armagan, H, Tuzun, E, Icoz, S, et al. Long extensive transverse myelitis associated with aquaporin-4 antibody and breast cancer: favorable response to cancer treatment. J Spinal Cord Med 2012;35:267269.Google Scholar
Moussawi, K, Lin, DJ, Matiello, M, et al. Brainstem and limbic encephalitis with paraneoplastic neuromyelitis optica. J Clin Neurosci 2016;23:159161.Google Scholar
Mueller, S, Dubal, DB, Josephson, SA. A case of paraneoplastic myelopathy associated with the neuromyelitis optica antibody. Nat Clin Pract Neurol 2008;4:284288.Google Scholar
Kitazawa, Y, Warabi, Y, Bandoh, M, Takahashi, T, Matsubara, S. Elderly-onset neuromyelitis optica which developed after the diagnosis of prostate adenocarcinoma and relapsed after a 23-valent pneumococcal polysaccharide vaccination. Intern Med (Tokyo, Japan) 2012;51:103107.Google Scholar
Kon, T, Ueno, T, Suzuki, C, et al. Aquaporin-4 antibody positive neuromyelitis optica spectrum disorder associated with esophageal cancer. J Neuroimmunol 2017;309:3840.Google Scholar
Annus, A, Bencsik, K, Obal, I, et al. Paraneoplastic neuromyelitis optica spectrum disorder: a case report and review of the literature. J Clin Neurosci 2018;48:710.Google Scholar
Beauchemin, P, Iorio, R, Traboulsee, AL, et al. Paraneoplastic neuromyelitis optica spectrum disorder: a single center cohort description with two cases of histological validation. Mult Scler Relat Disord 2018;20:3742.Google Scholar
Deuel, LM, Bunch, ME. A case of paraneoplastic neuromyelitis optica associated with small cell lung carcinoma. J Neuroimmunol 2018;316:130132.Google Scholar
Wiener, DC, Kaplan, TB, Bravo-Iniguez, CE, et al. Paraneoplastic neuromyelitis optica spectrum disorder as presentation of esophageal adenocarcinoma. Ann Thoracic Surg 2018;105:e133e135.Google Scholar
Baik, KW, Kim, SH, Shin, HY. Paraneoplastic neuromyelitis optica associated with lung adenocarcinoma in a young woman. J Clin Neurol (Seoul, Korea) 2018;14:246247.Google Scholar
Liao, W, Li, C, Tang, Y, et al. Aquaporin-4 antibody positive short transverse myelitis associated with breast cancer. Mult Scler Relat Disord 2019;30:119122.Google Scholar
Bernard-Valnet, R, Cobo-Calvo, A, Siegfried, A, et al. Paraneoplastic neuromyelitis optica and ovarian teratoma: a case series. Mult Scler Relat Disord 2019;31:97100.Google Scholar
Sachdeva, J, Bansal, R, Takkar, A, Singh, R. Neuromyelitis optica spectrum disorder and adenocarcinoma of ovary: a novel association. BMJ Case Rep 2019;12:e227435.Google Scholar
Sudo, A, Chihara, N, Takenaka, Y, et al. Paraneoplastic NMOSD associated with EG junction adenocarcinoma expressing unprotected AQP4. Neurol Neuroimmunol Neuroinflamm 2018;5:e482.Google Scholar
Fang, W, Zheng, Y, Cai, MT, Zhang, YX. Neuromyelitis optica spectrum disorder with lung adenocarcinoma and intraductal papillary mucinous neoplasm. Mult Scler Relat Disord 2019;32:7780.Google Scholar
Blackburn, K, Wang, C, Greenberg, B. Two cases of aquaporin-4 positive neuromyelitis optica associated with T-cell lymphoma. J Neuroimmunol 2020;338:577092.Google Scholar
Dinoto, A, Bosco, A, Sartori, A, et al. Hiccups, severe vomiting and longitudinally extensive transverse myelitis in a patient with prostatic adenocarcinoma and aquaporin-4 antibodies. J Neuroimmunol 2021;352:577488.Google Scholar
Reindl, M, Waters, P. Myelin oligodendrocyte glycoprotein antibodies in neurological disease. Nat Rev Neurol 2019;15:89102.Google Scholar
Jarius, S, Ruprecht, K, Wildemann, B, et al. Contrasting disease patterns in seropositive and seronegative neuromyelitis optica: a multicentre study of 175 patients. J Neuroinflammation 2012;9:14.Google Scholar
Collongues, N, Marignier, R, Zephir, H, et al. Neuromyelitis optica in France: a multicenter study of 125 patients. Neurology 2010;74:736742.Google Scholar
Petzold, A, Pittock, S, Lennon, V, et al. Neuromyelitis optica-IgG (aquaporin-4) autoantibodies in immune mediated optic neuritis. J Neurol Neurosurg Psychiatry 2010;81:109111.Google Scholar
Nakajima, H, Motomura, M, Tanaka, K, et al. Antibodies to myelin oligodendrocyte glycoprotein in idiopathic optic neuritis. BMJ Open 2015;5:e007766.Google Scholar
Rostasy, K, Mader, S, Schanda, K, et al. Anti-myelin oligodendrocyte glycoprotein antibodies in pediatric patients with optic neuritis. Arch Neurol 2012;69:752756.Google Scholar
Hoftberger, R, Sepulveda, M, Armangue, T, et al. Antibodies to MOG and AQP4 in adults with neuromyelitis optica and suspected limited forms of the disease. Mult Scler 2015;21:866874.Google Scholar
Martinez-Hernandez, E, Sepulveda, M, Rostasy, K, et al. Antibodies to aquaporin 4, myelin-oligodendrocyte glycoprotein, and the glycine receptor alpha1 subunit in patients with isolated optic neuritis. JAMA Neurol 2015;72:187193.Google Scholar
Petzold, A, Woodhall, M, Khaleeli, Z, et al. Aquaporin-4 and myelin oligodendrocyte glycoprotein antibodies in immune-mediated optic neuritis at long-term follow-up. J Neurol Neurosurg Psychiatry 2019;90:10211026.Google Scholar
Lotan, I, Hellmann, MA, Benninger, F, Stiebel-Kalish, H, Steiner, I. Recurrent optic neuritis: different patterns in multiple sclerosis, neuromyelitis optica spectrum disorders and MOG-antibody disease. J Neuroimmunol 2018;324:115118.Google Scholar
Chen, JJ, Flanagan, EP, Jitprapaikulsan, J, et al. Myelin oligodendrocyte glycoprotein antibody-positive optic neuritis: clinical characteristics, radiologic clues, and outcome. Am J Ophthalmol 2018;195:815.Google Scholar
Kim, SM, Woodhall, MR, Kim, JS, et al. Antibodies to MOG in adults with inflammatory demyelinating disease of the CNS. Neurol Neuroimmunol Neuroinflamm 2015;2:e163.Google Scholar
Ramanathan, S, Prelog, K, Barnes, EH, et al. Radiological differentiation of optic neuritis with myelin oligodendrocyte glycoprotein antibodies, aquaporin-4 antibodies, and multiple sclerosis. Mult Scler 2016;22:470482.Google Scholar
Storoni, M, Davagnanam, I, Radon, M, Siddiqui, A, Plant, GT. Distinguishing optic neuritis in neuromyelitis optica spectrum disease from multiple sclerosis: a novel magnetic resonance imaging scoring system. J Neuroophthalmol 2013;33:123127.Google Scholar
Costello, F, Coupland, S, Hodge, W, et al. Quantifying axonal loss after optic neuritis with optical coherence tomography. Ann Neurol 2006;59:963969.Google Scholar
Gabilondo, I, Martínez-Lapiscina, EH, Fraga-Pumar, E, et al. Dynamics of retinal injury after acute optic neuritis. Ann Neurol 2015;77:517528.Google Scholar
Martinez-Lapiscina, EH, Arnow, S, Wilson, JA, et al. Retinal thickness measured with optical coherence tomography and risk of disability worsening in multiple sclerosis: a cohort study. Lancet Neurol 2016;15:574584.Google Scholar
Xu, SC, Kardon, RH, Leavitt, JA, et al. Optical coherence tomography is highly sensitive in detecting prior optic neuritis. Neurology 2019;92:e527e535.Google Scholar
Specovius, S, Zimmermann, HG, Oertel, FC, et al. Cohort profile: a collaborative multicentre study of retinal optical coherence tomography in 539 patients with neuromyelitis optica spectrum disorders (CROCTINO). BMJ Open 2020;10:e035397.Google Scholar
Martinez-Lapiscina, EH, Sepulveda, M, Torres-Torres, R, et al. Usefulness of optical coherence tomography to distinguish optic neuritis associated with AQP4 or MOG in neuromyelitis optica spectrum disorders. Therapeut Adv Neurol Disord 2016;9:436440.Google Scholar
Filippatou, AG, Mukharesh, L, Saidha, S, Calabresi, PA, Sotirchos, ES. AQP4-IgG and MOG-IgG related optic neuritis-prevalence, optical coherence tomography findings, and visual outcomes: a systematic review and meta-analysis. Front Neurol 2020;11:540156.Google Scholar
Huang, Y, Zhou, L, ZhangBao, J, et al. Peripapillary and parafoveal vascular network assessment by optical coherence tomography angiography in aquaporin-4 antibody-positive neuromyelitis optica spectrum disorders. Br J Ophthalmol 2018;103:789796.Google Scholar
Weinshenker, BG, Wingerchuk, DM, Vukusic, S, et al. Neuromyelitis optica IgG predicts relapse after longitudinally extensive transverse myelitis. Ann Neurol 2006;59:566569.Google Scholar
Transverse Myelitis Consortium Working Group.Proposed diagnostic criteria and nosology of acute transverse myelitis. Neurology 2002;59:499505.Google Scholar
Iorio, R, Damato, V, Mirabella, M, et al. Distinctive clinical and neuroimaging characteristics of longitudinally extensive transverse myelitis associated with aquaporin-4 autoantibodies. J Neurol 2013;260:23962402.Google Scholar
Narayan, R, Simpson, A, Fritsche, K, et al. MOG antibody disease: a review of MOG antibody seropositive neuromyelitis optica spectrum disorder. Mult Scler Relat Disord 2018;25:6672.Google Scholar
Flanagan, EP, Weinshenker, BG, Krecke, KN, et al. Short myelitis lesions in aquaporin-4-IgG-positive neuromyelitis optica spectrum disorders. JAMA Neurol 2015;72:8187.Google Scholar
Cacciaguerra, L, Meani, A, Mesaros, S, et al. Brain and cord imaging features in neuromyelitis optica spectrum disorders. Ann Neurol 2019;85:371384.Google Scholar
Cheng, C, Jiang, Y, Lu, X, et al. The role of anti-aquaporin 4 antibody in the conversion of acute brainstem syndrome to neuromyelitis optica. BMC Neurology 2016;16:203.Google Scholar
Kremer, L, Mealy, M, Jacob, A, et al. Brainstem manifestations in neuromyelitis optica: a multicenter study of 258 patients. Mult Scler 2014;20:843847.Google Scholar
Shosha, E, Dubey, D, Palace, J, et al. Area postrema syndrome: frequency, criteria, and severity in AQP4-IgG-positive NMOSD. Neurology 2018;91:e1642e1651.Google Scholar
Takahashi, T, Miyazawa, I, Misu, T, et al. Intractable hiccup and nausea in neuromyelitis optica with anti-aquaporin-4 antibody: a herald of acute exacerbations. J Neurol Neurosurg Psychiatry 2008;79:10751078.Google Scholar
Apiwattanakul, M, Popescu, BF, Matiello, M, et al. Intractable vomiting as the initial presentation of neuromyelitis optica. Ann Neurol 2010;68:757761.Google Scholar
Misu, T, Fujihara, K, Nakashima, I, Sato, S, Itoyama, Y. Intractable hiccup and nausea with periaqueductal lesions in neuromyelitis optica. Neurology 2005;65:14791482.Google Scholar
Jarius, S, Kleiter, I, Ruprecht, K, et al. MOG-IgG in NMO and related disorders: a multicenter study of 50 patients. Part 3: brainstem involvement – frequency, presentation and outcome. J Neuroinflammation 2016;13:281.Google Scholar
Suzuki, K, Nakamura, T, Hashimoto, K, et al. Hypothermia, hypotension, hypersomnia, and obesity associated with hypothalamic lesions in a patient positive for the anti-aquaporin 4 antibody: a case report and literature review. Arch Neurol 2012;69:13551359.Google Scholar
Inoue, K, Nakayama, T, Kamisawa, A, Saito, J. Syndrome of inappropriate antidiuretic hormone accompanied by bilateral hypothalamic and anterior thalamic lesions with serum antiaquaporin 4 antibody. BMJ Case Rep 2017;2017:bcr2017219721.Google Scholar
Poppe, AY, Lapierre, Y, Melancon, D, et al. Neuromyelitis optica with hypothalamic involvement. Mult Scler 2005;11:617621.Google Scholar
Viegas, S, Weir, A, Esiri, M, et al. Symptomatic, radiological and pathological involvement of the hypothalamus in neuromyelitis optica. J Neurol Neurosurg Psychiatry 2009;80:679682.Google Scholar
Baba, T, Nakashima, I, Kanbayashi, T, et al. Narcolepsy as an initial manifestation of neuromyelitis optica with anti-aquaporin-4 antibody. J Neurol 2009;256:287288.Google Scholar
Zhang, L, Wu, A, Zhang, B, et al. Comparison of deep gray matter lesions on magnetic resonance imaging among adults with acute disseminated encephalomyelitis, multiple sclerosis, and neuromyelitis optica. Mult Scler 2014;20:418423.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
Cheng, C, Jiang, Y, Chen, X, et al. Clinical, radiographic characteristics and immunomodulating changes in neuromyelitis optica with extensive brain lesions. BMC Neurology 2013;13:72.Google Scholar
Kim, W, Park, MS, Lee, SH, et al. Characteristic brain magnetic resonance imaging abnormalities in central nervous system aquaporin-4 autoimmunity. Mult Scler 2010;16:12291236.Google Scholar
Magana, SM, Matiello, M, Pittock, SJ, et al. Posterior reversible encephalopathy syndrome in neuromyelitis optica spectrum disorders. Neurology 2009;72:712717.Google Scholar
Kim, W, Kim, SH, Huh, SY, Kim, HJ. Brain abnormalities in neuromyelitis optica spectrum disorder. Mult Scler Int 2012;2012:735486.Google Scholar
Armangue, T, Olive-Cirera, G, Martinez-Hernandez, E, et al. Associations of paediatric demyelinating and encephalitic syndromes with myelin oligodendrocyte glycoprotein antibodies: a multicentre observational study. Lancet Neurol 2020;19:234246.Google Scholar
Kim, SM, Kim, SJ, Lee, HJ, et al. Differential diagnosis of neuromyelitis optica spectrum disorders. Therapeut Adv Neurol Disord 2017;10:265289.Google Scholar
Polman, CH, Reingold, SC, Banwell, B, et al. Diagnostic criteria for multiple sclerosis: 2010 revisions to the McDonald criteria. Ann Neurol 2011;69:292302.Google Scholar
Bergamaschi, R, Tonietti, S, Franciotta, D, et al. Oligoclonal bands in Devic’s neuromyelitis optica and multiple sclerosis: differences in repeated cerebrospinal fluid examinations. Mult Scler 2004;10:24.Google Scholar
Thompson, AJ, Baranzini, SE, Geurts, J, Hemmer, B, Ciccarelli, O. Multiple sclerosis. Lancet 2018;391:16221636.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
Berry, S, Lin, WV, Sadaka, A, Lee, AG. Nonarteritic anterior ischemic optic neuropathy: cause, effect, and management. Eye Brain 2017;9:2328.Google Scholar
Yu-Wai-Man, P, Turnbull, DM, Chinnery, PF. Leber hereditary optic neuropathy. J Med Genet 2002;39:162169.Google Scholar
Scott, AM, Yinh, J, McAlindon, T, Kalish, R. Two cases of sarcoidosis presenting as longitudinally extensive transverse myelitis. Clin Rheumatol 2018;37:28992905.Google Scholar
Lee, HS, Kim do, Y, Shin, HY, et al. Spinal cord involvement in Behcet’s disease. Mult Scler 2016;22:960963.Google Scholar
Uygunoglu, U, Zeydan, B, Ozguler, Y, et al. Myelopathy in Behçet’s disease: the bagel sign. Ann Neurol 2017;82:288298.Google Scholar
Isobe, N, Kanamori, Y, Yonekawa, T, et al. First diagnostic criteria for atopic myelitis with special reference to discrimination from myelitis-onset multiple sclerosis. J Neurol Sci 2012;316:3035.Google Scholar
Flanagan, EP, Krecke, KN, Marsh, RW, et al. Specific pattern of gadolinium enhancement in spondylotic myelopathy. Ann Neurol 2014;76:5465.Google Scholar
Atkinson, JL, Miller, GM, Krauss, WE, et al. Clinical and radiographic features of dural arteriovenous fistula, a treatable cause of myelopathy. Mayo Clin Proc 2001;76:11201130.Google Scholar
Cabrera, M, Paradas, C, Marquez, C, Gonzalez, A. Acute paraparesis following intravenous steroid therapy in a case of dural spinal arteriovenous fistula. J Neurol 2008;255:14321433.Google Scholar
Zalewski, NL, Rabinstein, AA, Krecke, KN, et al. Characteristics of spontaneous spinal cord infarction and proposed diagnostic criteria. JAMA Neurol 2019;76:5663.Google Scholar
Fatemi, Y, Chakraborty, R. Acute flaccid myelitis: a clinical overview for 2019. Mayo Clin Proc 2019;94:875881.Google Scholar
Mihai, C, Jubelt, B. Infectious myelitis. Curr Neurol Neurosci Rep 2012;12:633641.Google Scholar
Tobin, WO, Guo, Y, Krecke, KN, et al. Diagnostic criteria for chronic lymphocytic inflammation with pontine perivascular enhancement responsive to steroids (CLIPPERS). Brain 2017;140:24152425.Google Scholar
Dalmau, J, Graus, F, Villarejo, A, et al. Clinical analysis of anti-Ma2-associated encephalitis. Brain 2004;127:18311844.Google Scholar
Schwartz, MA, Selhorst, JB, Ochs, AL, et al. Oculomasticatory myorhythmia: a unique movement disorder occurring in Whipple’s disease. Ann Neurol 1986;20:677683.Google Scholar
Spatola, M, Petit-Pedrol, M, Simabukuro, MM, et al. Investigations in GABAa receptor antibody-associated encephalitis. Neurology 2017;88:1012–1020.Google Scholar
Izquierdo, C, Velasco, R, Vidal, N, et al. Lymphomatosis cerebri: a rare form of primary central nervous system lymphoma: analysis of 7 cases and systematic review of the literature. Neuro-oncology 2016;18:707715.Google Scholar
Koelman, DL, Chahin, S, Mar, SS, et al. Acute disseminated encephalomyelitis in 228 patients: a retrospective, multicenter US study. Neurology 2016;86:20852093.Google Scholar
Tobin, WO, Weinshenker, BG, Lucchinetti, CF. Longitudinally extensive transverse myelitis. Curr Opin Neurol 2014;27:279289.Google Scholar
Nardone, R, Versace, V, Brigo, F, et al. Herpes simplex virus type 2 myelitis: case report and review of the literature. Front Neurol 2017;8:199.Google Scholar
Steiner, I, Kennedy, PG, Pachner, AR. The neurotropic herpes viruses: herpes simplex and varicella-zoster. Lancet Neurol 2007;6:10151028.Google Scholar
Greninger, AL, Naccache, SN, Messacar, K, et al. A novel outbreak enterovirus D68 strain associated with acute flaccid myelitis cases in the USA (2012–14): a retrospective cohort study. Lancet Infect Dis 2015;15:671682.Google Scholar
Heerlein, K, Jarius, S, Jacobi, C, et al. Aquaporin-4 antibody positive longitudinally extensive transverse myelitis following varicella zoster infection. J Neurol Sci 2009;276:184186.Google Scholar
Sejvar, JJ, Lopez, AS, Cortese, MM, et al. Acute flaccid myelitis in the United States, August–December 2014: results of nationwide surveillance. Clin Infect Dis 2016;63:737745.Google Scholar
McMinn, P, Stratov, I, Nagarajan, L, Davis, S. Neurological manifestations of enterovirus 71 infection in children during an outbreak of hand, foot, and mouth disease in Western Australia. Clin Infect Dis 2001;32:236242.Google Scholar
Chow, CCN, Magnussen, J, Ip, J, Su, Y. Acute transverse myelitis in COVID-19 infection. BMJ Case Rep 2020;13:236720.Google Scholar
Kaur, H, Mason, JA, Bajracharya, M, et al. Transverse myelitis in a child with COVID-19. Pediatr Neurol 2020;112:56.Google Scholar
Sotoca, J, Rodríguez-Álvarez, Y. COVID-19-associated acute necrotizing myelitis. Neurol Neuroimmunol Neuroinflamm 2020;7:e803.Google Scholar
Valiuddin, H, Skwirsk, B, Paz-Arabo, P. Acute transverse myelitis associated with SARS-CoV-2: a case-report. Brain Behav Immun Health 2020;5:100091.Google Scholar
Liu, Z, Jiao, L, Qiu, Z, et al. Clinical characteristics of patients with paraneoplastic myelopathy. J Neuroimmunol 2019;330:136142.Google Scholar
Keegan, BM, Pittock, SJ, Lennon, VA. Autoimmune myelopathy associated with collapsin response-mediator protein-5 immunoglobulin G. Ann Neurol 2008;63:531534.Google Scholar
Zalewski, NL, Flanagan, EP. Autoimmune and paraneoplastic myelopathies. Semin Neurol 2018;38:278289.Google Scholar
Mancall, EL, Rosales, RK. Necrotizing myelopathy associated with visceral carcinoma. Brain 1964;87:639656.Google Scholar
Flanagan, EP, Kaufmann, TJ, Krecke, KN, et al. Discriminating long myelitis of neuromyelitis optica from sarcoidosis. Ann Neurol 2016;79:437447.Google Scholar
Zalewski, NL, Rabinstein, AA, Brinjikji, W, et al. Unique gadolinium enhancement pattern in spinal dural arteriovenous fistulas. JAMA Neurol 2018;75:15421545.Google Scholar
O’Keeffe, DT, Mikhail, MA, Lanzino, G, Kallmes, DF, Weinshenker, BG. Corticosteroid-induced paraplegia: a diagnostic clue for spinal dural arterial venous fistula. JAMA Neurol 2015;72:833834.Google Scholar
Zhang, S, Liu, H, Li, J. Cervical myelopathy caused by intracranial dural arteriovenous fistula with acute worsening after steroid administration. World Neurosurg 2018;120:328330.Google Scholar
Akman-Demir, G, Serdaroglu, P, Tasci, B. Clinical patterns of neurological involvement in Behcet’s disease: evaluation of 200 patients. The Neuro-Behcet Study Group. Brain 1999;122:21712182.Google Scholar
Serdaroglu, P, Yazici, H, Ozdemir, C, et al. Neurologic involvement in Behcet’s syndrome. A prospective study. Arch Neurol 1989;46:265269.Google Scholar
Tan, IL, Mowry, EM, Steele, SU, et al. Brainstem encephalitis: etiologies, treatment, and predictors of outcome. J Neurol 2013;260:23122319.Google Scholar
Moragas, M, Martinez-Yelamos, S, Majos, C, et al. Rhombencephalitis: a series of 97 patients. Medicine (Baltimore) 2011;90:256261.Google Scholar
Al Sawaf, A, Berger, JR. Longitudinally extensive transverse myelitis suspected for isolated Neuro-Behcet: a diagnostic conundrum. Mult Scler Relat Disord 2015;4:395399.Google Scholar
Compain, C, Sacre, K, Puechal, X, et al. Central nervous system involvement in Whipple disease: clinical study of 18 patients and long-term follow-up. Medicine (Baltimore) 2013;92:324330.Google Scholar
Black, DF, Aksamit, AJ, Morris, JM. MR imaging of central nervous system Whipple disease: a 15-year review. AM J Neuroradiol 2010;31:14931497.Google Scholar
Compta, Y, Iranzo, A, Santamaria, J, Casamitjana, R, Graus, F. REM sleep behavior disorder and narcoleptic features in anti-Ma2-associated encephalitis. Sleep 2007;30:767769.Google Scholar
Rojas-Marcos, I, Graus, F, Sanz, G, Robledo, A, Diaz-Espejo, C. Hypersomnia as presenting symptom of anti-Ma2-associated encephalitis: case study. Neuro-oncol 2007;9:7577.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
Nielsen, S, Nagelhus, EA, Amiry-Moghaddam, M, et al. Specialized membrane domains for water transport in glial cells: high-resolution immunogold cytochemistry of aquaporin-4 in rat brain. J Neurosci 1997;17:171180.Google Scholar
Papadopoulos, MC, Saadoun, S. Key roles of aquaporins in tumor biology. Biochim Biophys Acta 2015;1848:25762583.Google Scholar
Roemer, SF, Parisi, JE, Lennon, VA, et al. Pattern-specific loss of aquaporin-4 immunoreactivity distinguishes neuromyelitis optica from multiple sclerosis. Brain 2007;130:11941205.Google Scholar
Lucchinetti, CF, Mandler, RN, McGavern, D, et al. A role for humoral mechanisms in the pathogenesis of Devic’s neuromyelitis optica. Brain 2002;125:14501461.Google Scholar
Misu, T, Hoftberger, R, Fujihara, K, et al. Presence of six different lesion types suggests diverse mechanisms of tissue injury in neuromyelitis optica. Acta Neuropathol 2013;125:815827.Google Scholar
Hinson, SR, Roemer, SF, Lucchinetti, CF, et al. Aquaporin-4-binding autoantibodies in patients with neuromyelitis optica impair glutamate transport by down-regulating EAAT2. J Exp Med 2008;205:24732481.Google Scholar
Richard, C, Ruiz, A, Cavagna, S, et al. Connexins in neuromyelitis optica: a link between astrocytopathy and demyelination. Brain 2020;143:27212732.Google Scholar
Jacob, A, Saadoun, S, Kitley, J, et al. Detrimental role of granulocyte-colony stimulating factor in neuromyelitis optica: clinical case and histological evidence. Mult Scler 2012;18:18011803.Google Scholar
Palace, J, Leite, MI, Nairne, A, Vincent, A. Interferon beta treatment in neuromyelitis optica: increase in relapses and aquaporin 4 antibody titers. Arch Neurol 2010;67:10161017.Google Scholar
Pohl, M, Fischer, MT, Mader, S, et al. Pathogenic T cell responses against aquaporin 4. Acta Neuropathol 2011;122:2134.Google Scholar
Hillebrand, S, Schanda, K, Nigritinou, M, et al. Circulating AQP4-specific auto-antibodies alone can induce neuromyelitis optica spectrum disorder in the rat. Acta Neuropathol 2019;137:467485.Google Scholar
Ketelslegers, IA, Modderman, PW, Vennegoor, A, et al. Antibodies against aquaporin-4 in neuromyelitis optica: distinction between recurrent and monophasic patients. Mult Scler 2011;17:15271530.Google Scholar
Palace, J, Lin, DY, Zeng, D, et al. Outcome prediction models in AQP4-IgG positive neuromyelitis optica spectrum disorders. Brain 2019;142:13101323.Google Scholar
Marignier, R, Cobo Calvo, A, Vukusic, S. Neuromyelitis optica and neuromyelitis optica spectrum disorders. Curr Opin Neurol 2017;30:208215.Google Scholar
Abboud, H, Petrak, A, Mealy, M, et al. Treatment of acute relapses in neuromyelitis optica: steroids alone versus steroids plus plasma exchange. Mult Scler 2016;22:185192.Google Scholar
Kleiter, I, Gahlen, A, Borisow, N, et al. Neuromyelitis optica: evaluation of 871 attacks and 1,153 treatment courses. Ann Neurol 2016;79:206216.Google Scholar
Kleiter, I, Gahlen, A, Borisow, N, et al. Apheresis therapies for NMOSD attacks: a retrospective study of 207 therapeutic interventions. Neurol Neuroimmunol Neuroinflamm 2018;5:e504.Google Scholar
Elsone, L, Panicker, J, Mutch, K, et al. Role of intravenous immunoglobulin in the treatment of acute relapses of neuromyelitis optica: experience in 10 patients. Mult Scler 2014;20:501504.Google Scholar
Magraner, MJ, Coret, F, Casanova, B. The effect of intravenous immunoglobulin on neuromyelitis optica. Neurologia 2013;28:6572.Google Scholar
Absoud, M, Brex, P, Ciccarelli, O, et al. A multicentre randomiSed controlled TRial of IntraVEnous immunoglobulin compared with standard therapy for the treatment of transverse myelitis in adults and children (STRIVE). Health Technol Assess (Winchester, England) 2017;21:150.Google Scholar
Stellmann, JP, Krumbholz, M, Friede, T, et al. Immunotherapies in neuromyelitis optica spectrum disorder: efficacy and predictors of response. J Neurol Neurosurg Psychiatry 2017;88:639647.Google Scholar
Nikoo, Z, Badihian, S, Shaygannejad, V, Asgari, N, Ashtari, F. Comparison of the efficacy of azathioprine and rituximab in neuromyelitis optica spectrum disorder: a randomized clinical trial. J Neurol 2017;264:20032009.Google Scholar
Mealy, MA, Wingerchuk, DM, Palace, J, Greenberg, BM, Levy, M. Comparison of relapse and treatment failure rates among patients with neuromyelitis optica: multicenter study of treatment efficacy. JAMA Neurol 2014;71:324330.Google Scholar
Jeong, IH, Park, B, Kim, SH, et al. Comparative analysis of treatment outcomes in patients with neuromyelitis optica spectrum disorder using multifaceted endpoints. Mult Scler 2016;22:329339.Google Scholar
Damato, V, Evoli, A, Iorio, R. Efficacy and safety of rituximab therapy in neuromyelitis optica spectrum disorders: a systematic review and meta-analysis. JAMA Neurol 2016;73:13421348.Google Scholar
Montcuquet, A, Collongues, N, Papeix, C, et al. Effectiveness of mycophenolate mofetil as first-line therapy in AQP4-IgG, MOG-IgG, and seronegative neuromyelitis optica spectrum disorders. Mult Scler 2017;23:13771384.Google Scholar
Kojima, M, Oji, S, Tanaka, S, et al. Tacrolimus is effective for neuromyelitis optica spectrum disorders with or without anti-AQP4 antibody. Mult Scler Relat Disord 2019;39:101907.Google Scholar
Zhang, C, Tian, DC, Yang, CS, et al. Safety and efficacy of bortezomib in patients with highly relapsing neuromyelitis optica spectrum disorder. JAMA Neurol 2017;74:10101012.Google Scholar
Ringelstein, M, Ayzenberg, I, Harmel, J, et al. Long-term therapy with interleukin 6 receptor blockade in highly active neuromyelitis optica spectrum disorder. JAMA Neurol 2015;72:756763.Google Scholar
Carreon Guarnizo, E, Hernandez Clares, R, Castillo Trivino, T, et al. Experience with tocilizumab in patients with neuromyelitis optica spectrum disorders. Neurologia 2019;S0213-4853(19)30033-7.Google Scholar
Zhang, C, Zhang, M, Qiu, W, et al. Safety and efficacy of tocilizumab versus azathioprine in highly relapsing neuromyelitis optica spectrum disorder (TANGO): an open-label, multicentre, randomised, phase 2 trial. Lancet Neurol 2020;19:391401.Google Scholar
Snowden, JA, Pearce, RM, Lee, J, et al. Haematopoietic stem cell transplantation (HSCT) in severe autoimmune diseases: analysis of UK outcomes from the British Society of Blood and Marrow Transplantation (BSBMT) data registry 1997–2009. Br J Haematol 2012;157:742746.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 2020;55:283306.Google Scholar
Greco, R, Bondanza, A, Oliveira, MC, et al. Autologous hematopoietic stem cell transplantation in neuromyelitis optica: a registry study of the EBMT Autoimmune Diseases Working Party. Mult Scler 2015;21:189197.Google Scholar
Burt, RK, Balabanov, R, Han, X, et al. Autologous nonmyeloablative hematopoietic stem cell transplantation for neuromyelitis optica. Neurology 2019;93:e1732e1741.Google Scholar
Pittock, SJ, Berthele, A, Fujihara, K, et al. Eculizumab in aquaporin-4-positive neuromyelitis optica spectrum disorder. N Engl J Med 2019;381:614625.Google Scholar
Traboulsee, A, Greenberg, BM, Bennett, JL, et al. Safety and efficacy of satralizumab monotherapy in neuromyelitis optica spectrum disorder: a randomised, double-blind, multicentre, placebo-controlled phase 3 trial. Lancet Neurol 2020;19:402412.Google Scholar
Yamamura, T, Kleiter, I, Fujihara, K, et al. Trial of satralizumab in neuromyelitis optica spectrum disorder. N Engl J Med 2019;381:21142124.Google Scholar
Cree, BAC, Bennett, JL, Kim, HJ, et al. Inebilizumab for the treatment of neuromyelitis optica spectrum disorder (N-MOmentum): a double-blind, randomised placebo-controlled phase 2/3 trial. Lancet 2019;394:13521363.Google Scholar
Tahara, M, Oeda, T, Okada, K, et al. Safety and efficacy of rituximab in neuromyelitis optica spectrum disorders (RIN-1 study): a multicentre, randomised, double-blind, placebo-controlled trial. Lancet Neurol 2020;19:298306.Google Scholar
Xue, T, Yang, Y, Lu, Q, et al. Efficacy and safety of monoclonal antibody therapy in neuromyelitis optica spectrum disorders: evidence from randomized controlled trials. Mult Scler Relat Disord 2020;43:102166.Google Scholar
Maillart, E, Durand-Dubief, F, Louapre, C, et al. Outcome and risk of recurrence in a large cohort of idiopathic longitudinally extensive transverse myelitis without AQP4/MOG antibodies. J Neuroinflammation 2020;17:128.Google Scholar
Levy, M, Fujihara, K, Palace, J. New therapies for neuromyelitis optica spectrum disorder. Lancet Neurol 2021;20:6067.Google Scholar
Singh, VK, Warren, R, Averett, R, Ghaziuddin, M. Circulating autoantibodies to neuronal and glial filament proteins in autism. Pediatr Neurol 1997;17:8890.Google Scholar
Wang, KK, Yang, Z, Yue, JK, et al. Plasma anti-glial fibrillary acidic protein autoantibody levels during the acute and chronic phases of traumatic brain injury: a transforming research and clinical knowledge in traumatic brain injury pilot study. J Neurotrauma 2016;33:12701277.Google Scholar
Poletaev, AB, Morozov, SG, Gnedenko, BB, Zlunikin, VM, Korzhenevskey, DA. Serum anti-S100b, anti-GFAP and anti-NGF autoantibodies of IgG class in healthy persons and patients with mental and neurological disorders. Autoimmunity 2000;32:3338.Google Scholar
El Rahman, HAA, Salama, M, Gad El-Hak, SA, et al. A panel of autoantibodies against neural proteins as peripheral biomarker for pesticide-induced neurotoxicity. Neurotox Res 2018;33:316336.Google Scholar
Dubey, D, Hinson, SR, Jolliffe, EA, et al. Autoimmune GFAP astrocytopathy: prospective evaluation of 90 patients in 1 year. J Neuroimmunol 2018;321:157163.Google Scholar
Kunchok, A, Zekeridou, A, McKeon, A. Autoimmune glial fibrillary acidic protein astrocytopathy. Curr Opin Neurol 2019;32:452458.Google Scholar
Issa, N, Martin, C, Dulau, C, Camou, F. Severe anti-GFAP meningo-encephalomyelitis following viral infection. Mult Scler Relat Disord 2020;45:102448.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
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
Iorio, R, Damato, V, Evoli, A, et al. Clinical and immunological characteristics of the spectrum of GFAP autoimmunity: a case series of 22 patients. J Neurol Neurosurg Psychiatry 2018;89:138146.Google Scholar
Kimura, A, Takekoshi, A, Yoshikura, N, Hayashi, Y, Shimohata, T. Clinical characteristics of autoimmune GFAP astrocytopathy. J Neuroimmunol 2019;332:9198.Google Scholar
Wickel, J, Chung, HY, Kirchhof, K, et al. Encephalitis with radial perivascular emphasis: not necessarily associated with GFAP antibodies. Neurol Neuroimmunol Neuroinflamm 2020;7:e670.Google Scholar
Sechi, E, Morris, PP, McKeon, A, et al. Glial fibrillary acidic protein IgG related myelitis: characterisation and comparison with aquaporin-4-IgG myelitis. J Neurol Neurosurg Psychiatry 2019;90:488490.Google Scholar

References

Hadjivassiliou, M, Boscolo, S, Tongiorgi, E, et al. Cerebellar ataxia as a possible organ-specific autoimmune disease. Mov Disord 2008;23:13701377.Google Scholar
Sivera, R, Martin, N, Bosca, I, et al. Autoimmunity as a prognostic factor in sporadic adult onset cerebellar ataxia. J Neurol 2012;259:851854.Google Scholar
Abele, M, Burk, K, Schols, L, et al. The aetiology of sporadic adult-onset ataxia. Brain 2002;125:961968.Google Scholar
Lin, CY, Wang, MJ, Tse, W, et al. Serum antigliadin antibodies in cerebellar ataxias: a systematic review and meta-analysis. J Neurol Neurosurg Psychiatry 2018;89:11741180.Google Scholar
Mitoma, H, Manto, M, Hampe, CS. Time is cerebellum. Cerebellum 2018;17:387391.Google Scholar
Jones, AL, Flanagan, EP, Pittock, SJ, et al. Responses to and outcomes of treatment of autoimmune cerebellar ataxia in adults. JAMA Neurol 2015;72:13041312.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
Mitoma, H, Adhikari, K, Aeschlimann, D, et al. Consensus paper: neuroimmune mechanisms of cerebellar ataxias. Cerebellum 2016;15:213232.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
Honnorat, J, Saiz, A, Giometto, B, et al. Cerebellar ataxia with anti-glutamic acid decarboxylase antibodies: study of 14 patients. Arch Neurol 2001;58:225230.Google Scholar
Lopez-Chiriboga, AS, Komorowski, L, Kumpfel, T, et al. Metabotropic glutamate receptor type 1 autoimmunity: clinical features and treatment outcomes. Neurology 2016;86:10091013.Google Scholar
Klockgether, T. Sporadic ataxia with adult onset: classification and diagnostic criteria. Lancet Neurol 2010;9:94104.Google Scholar
Graus, F, Saiz, A, Dalmau, J. GAD antibodies in neurological disorders: insights and challenges. Nat Rev Neurol 2020;16:353365.Google Scholar
Manto, M. Toxic agents causing cerebellar ataxias. Handb Clin Neurol 2012;103:201213.Google Scholar
Hadjivassiliou, M, Martindale, J, Shanmugarajah, P, et al. Causes of progressive cerebellar ataxia: prospective evaluation of 1500 patients. J Neurol Neurosurg Psychiatry 2017;88:301309.Google Scholar
Synofzik, M, Nemeth, AH. Recessive ataxias. Handb Clin Neurol 2018;155:7389.Google Scholar
Hagerman, RJ, Hagerman, P. Fragile X-associated tremor/ataxia syndrome: features, mechanisms and management. Nat Rev Neurol 2016;12:403412.Google Scholar
Fanciulli, A, Wenning, GK. Multiple-system atrophy. N Engl J Med 2015;372:249263.Google Scholar
Baiardi, S, Magherini, A, Capellari, S, et al. Towards an early clinical diagnosis of sporadic CJD VV2 (ataxic type). J Neurol Neurosurg Psychiatry 2017;88:764772.Google Scholar
Pichler, M, Vemuri, P, Rabinstein, AA, et al. Prevalence and natural history of superficial siderosis: a population-based study. Stroke 2017;48:32103214.Google Scholar
Matthews, BR, Jones, LK, Saad, DA, Aksamit, AJ, Josephs, KA. Cerebellar ataxia and central nervous system Whipple disease. Arch Neurol 2005;62:618620.Google Scholar
Wijburg, MT, van Oosten, BW, Murk, JL, et al. Heterogeneous imaging characteristics of JC virus granule cell neuronopathy (GCN): a case series and review of the literature. J Neurol 2015;262:6573.Google Scholar
Pedroso, JL, Vale, TC, Gama, MTD, et al. Cerebellar degeneration and progressive ataxia associated with HIV-virus infection. Parkinsonism Relat Disord 2018;54:9598.Google Scholar
Hadjivassiliou, M, Sanders, DD, Aeschlimann, DP. Gluten-related disorders: gluten ataxia. Digestive Dis (Basel, Switzerland) 2015;33:264268.Google Scholar
McKeon, A, Lennon, VA, Pittock, SJ, Kryzer, TJ, Murray, J. The neurologic significance of celiac disease biomarkers. Neurology 2014;83:17891796.Google Scholar
Matsunaga, A, Ikawa, M, Fujii, A, et al. Hashimoto’s encephalopathy as a treatable adult-onset cerebellar ataxia mimicking spinocerebellar degeneration. Eur Neurol 2013;69:1420.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
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
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
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, Shams’ili, S, Rojas, I, et al. Anti-Tr antibodies as markers of paraneoplastic cerebellar degeneration and Hodgkin’s disease. Neurology 2003;60:230234.Google Scholar
Pittock, SJ, Lucchinetti, CF, Lennon, VA. Anti-neuronal nuclear autoantibody type 2: paraneoplastic accompaniments. Ann Neurol 2003;53:580587.Google Scholar
Dalmau, J, Graus, F, Villarejo, A, et al. Clinical analysis of anti-Ma2-associated encephalitis. Brain 2004;127:18311844.Google Scholar
Gadoth, A, Kryzer, TJ, Fryer, J, et al. Microtubule-associated protein 1B: novel paraneoplastic biomarker. Ann Neurol 2017;81:266277.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
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
Bataller, L, Wade, DF, Rosenfeld, MR, Dalmau, J. Immunity to Zic proteins frequently associates with paraneoplastic neurologic disorders (PND) and predicts small-cell lung cancer. Ann Neurol 2002;59:19851988.Google Scholar
Pittock, SJ, Lucchinetti, CF, Parisi, JE, et al. Amphiphysin autoimmunity: paraneoplastic accompaniments. Ann Neurol 2005;58:96107.Google Scholar
Tetsuka, S, Tominaga, K, Ohta, E, et al. Paraneoplastic cerebellar degeneration associated with an onconeural antibody against creatine kinase, brain-type. J Neurol Sci 2013;335:4857.Google Scholar
Do, LD, Gupton, SL, Tanji, K, et al. TRIM9 and TRIM67 are new targets in paraneoplastic cerebellar degeneration. Cerebellum 2019;18:245254.Google Scholar
van Coevorden-Hameete, MH, van Beuningen, SFB, Perrenoud, M, et al. Antibodies to TRIM46 are associated with paraneoplastic neurological syndromes. Ann Clin Transl Neurol 2017;4:680686.Google Scholar
Bataller, L, Sabater, L, Saiz, A, et al. Carbonic anhydrase-related protein VIII: autoantigen in paraneoplastic cerebellar degeneration. Ann Neurol 2004;56:575579.Google Scholar
Prevezianou, A, Tzartos, JS, Dagklis, IE, et al. Paraneoplastic cerebellar degeneration in a patient with breast cancer associated with carbonic anhydrase-related protein VIII autoantibodies. J Neuroimmunol 2020;344:577242.Google Scholar
Hoftberger, R, Kovacs, GG, Sabater, L, et al. Protein kinase Cγ antibodies and paraneoplastic cerebellar degeneration. J Neuroimmunol 2013;256:9193.Google Scholar
Ren, H, Zhao, D, Xu, X, et al. Paraneoplastic cerebellar degeneration associated with anti-protein kinase Cgamma antibodies in a Chinese patient. J Neuroimmunol 2020;350:577408.Google Scholar
Ruiz-Garcia, R, Martinez-Hernandez, E, Joubert, B, et al. Paraneoplastic cerebellar ataxia and antibodies to metabotropic glutamate receptor 2. Neurol Neuroimmunol Neuroinflamm 2020;7:e658.Google Scholar
van Coevorden-Hameete, MH, de Graaff, E, Titulaer, MJ, et al. Plasticity-related gene 5: a novel surface autoantigen in paraneoplastic cerebellar degeneration. Neurol Neuroimmunol Neuroinflamm 2015;2:e156.Google Scholar
Arino, H, Gresa-Arribas, N, Blanco, Y, et al. Cerebellar ataxia and glutamic acid decarboxylase antibodies: immunologic profile and long-term effect of immunotherapy. JAMA Neurol 2014;71:10091016.Google Scholar
Spatola, M, Pedrol, MP, Maudes, E, et al. Clinical features, prognostic factors, and antibody effects in anti-mGluR1 encephalitis. Neurology 2020;95:e3012e3025.Google Scholar
Jarius, S, Wandinger, KP, Horn, S, Heuer, H, Wildemann, B. A new Purkinje cell antibody (anti-Ca) associated with subacute cerebellar ataxia: immunological characterization. J Neuroinflammation 2010;7:21.Google Scholar
Pittock, SJ, Alfugham, N, O’Connor, K, et al. GTPase regulator associated with focal adhesion kinase 1 (GRAF1) immunoglobulin-associated ataxia and neuropathy. Mov Disord Clin Pract 2020;7:904909.Google Scholar
Alfugham, N, Gadoth, A, Lennon, VA, et al. ITPR1 autoimmunity: frequency, neurologic phenotype, and cancer association. Neurol Neuroimmunol Neuroinflamm 2018;5:e418.Google Scholar
Jarius, S, Scharf, M, Begemann, N, et al. Antibodies to the inositol 1,4,5-trisphosphate receptor type 1 (ITPR1) in cerebellar ataxia. J Neuroinflammation 2014;11:206.Google Scholar
Miske, R, Gross, CC, Scharf, M, et al. Neurochondrin is a neuronal target antigen in autoimmune cerebellar degeneration. Neurol Neuroimmunol Neuroinflamm 2017;4:e307.Google Scholar
Shelly, S, Kryzer, TJ, Komorowski, L, et al. Neurochondrin neurological autoimmunity. Neurol Neuroimmunol Neuroinflamm 2019;6:e612.Google Scholar
Joubert, B, Gobert, F, Thomas, L, et al. Autoimmune episodic ataxia in patients with anti-CASPR2 antibody-associated encephalitis. Neurol Neuroimmunol Neuroinflamm 2017;4:e371.Google Scholar
Yaguchi, H, Yabe, I, Takahashi, H, et al. Identification of anti-Sez6l2 antibody in a patient with cerebellar ataxia and retinopathy. J Neurol 2014;261:224226.Google Scholar
Landa, J, Guasp, M, Petit-Pedrol, M, et al. Seizure-related 6 homolog like 2 autoimmunity: neurologic syndrome and antibody effects. Neurol Neuroimmunol Neuroinflamm 2021;8:e916.Google Scholar
Honorat, JA, Lopez-Chiriboga, AS, Kryzer, TJ, et al. Autoimmune gait disturbance accompanying adaptor protein-3B2-IgG. Neurology 2019;93:e954e963.Google Scholar
Jarius, S, Wildemann, B. ‘Medusa head ataxia’: the expanding spectrum of Purkinje cell antibodies in autoimmune cerebellar ataxia. Part 2: Anti-PKC-gamma, anti-GluR-delta2, anti-Ca/ARHGAP26 and anti-VGCC. J Neuroinflammation 2015;12:167.Google Scholar
Landa, J, Guasp, M, Míguez-Cabello, F, et al. Encephalitis with autoantibodies against the Glutamate Kainate Receptors GluK2. Ann Neurol 2021;90:101117.Google Scholar
Honorat, JA, Lopez-Chiriboga, AS, Kryzer, TJ, et al. Autoimmune septin-5 cerebellar ataxia. Neurol Neuroimmunol Neuroinflamm 2018;5:e474.Google Scholar
Hoftberger, R, Sabater, L, Ortega, A, Dalmau, J, Graus, F. Patient with homer-3 antibodies and cerebellitis. JAMA Neurol 2013;70:506509.Google Scholar
Darnell, RB, Furneaux, HM, Posner, JB. Antiserum from a patient with cerebellar degeneration identifies a novel protein in Purkinje cells, cortical neurons and neuroectodermal tumors. J Neurosci 1991;11:12241230.Google Scholar
Hadjivassiliou, M, Graus, F, Honnorat, J, et al. Diagnostic criteria for primary autoimmune cerebellar ataxia: guidelines from an international task force on immune-mediated cerebellar ataxias. Cerebellum 2020;19:605610.Google Scholar
Brouwer, B. Beitrag zur Kenntnis der chronischen diffusen Kleinhirnerkrankungen. Neurol Zentralbl 1919;38:674682.Google Scholar
Brain, WR, Daniel, PM, Greenfield, JG. Subacute cortical cerebellar degeneration and its relation to carcinoma. J Neurol Neurosurg Psychiat 1951;14:5975.Google Scholar
Brain, WR, Wilkinson, M. Subacute cerebellar degeneration associated with neoplasms. Brain 1965;88:465478.Google Scholar
Henson, RA, Urich, HE. Cancer and the Nervous System: The Neurological Manifestations of Systemic Malignant Disease. London: Blackwell Scientific, 1982.Google Scholar
Porta-Etessam, J, Berbel, A, Martinez-Salio, A, et al. [Paraneoplastic cerebellar degeneration with asymmetrical pan-cerebellar syndrome]. Rev Neurol 1998;26:10151017.Google Scholar
Krolak-Salmon, P, Androdias, G, Meyronet, D, et al. Slow evolution of cerebellar degeneration and chorea in a man with anti-Yo antibodies. Eur J Neurol 2006;13:307308.Google Scholar
Bodranghien, F, Bastian, A, Casali, C, et al. Consensus paper: revisiting the symptoms and signs of cerebellar syndrome. Cerebellum 2016;15:369391.Google Scholar
Schmahmann, JD. The cerebellum and cognition. Neurosci Lett 2019;688:6275.Google Scholar
de Andres C, Esquivel, A, de Villoria, JG, Graus, F, Sanchez-Ramon, S. Unusual magnetic resonance imaging and cerebrospinal fluid findings in paraneoplastic cerebellar degeneration: a sequential study. J Neurol Neurosurg Psychiatry 2006;77:562563.Google Scholar
Suri, V, Khan, NI, Jadhao, N, Gupta, R. Paraneoplastic cerebellar degeneration in Hodgkin’s lymphoma. Ann Ind Acad Neurol 2012;15:205207.Google Scholar
Choi, KD, Kim, JS, Park, SH, et al. Cerebellar hypermetabolism in paraneoplastic cerebellar degeneration. J Neurol Neurosurg Psychiatry 2006;77:525528.Google Scholar
Abdulaziz, ATA, Yu, XQ, Zhang, L, et al. Paraneoplastic cerebellar degeneration associated with cerebellar hypermetabolism: case report. Medicine (Baltimore) 2018;97:e10717.Google Scholar
Gheysens, O, Deroose, CM, Tousseyn, T, et al. Hodgkin lymphoma-associated paraneoplastic cerebellar degeneration on FDG-PET/CT. Br J Haematol 2014;164:468.Google Scholar
Saiz, A, Graus, F, Dalmau, J, et al. Detection of 14-3-3 brain protein in the cerebrospinal fluid of patients with paraneoplastic neurological disorders. Ann Neurol 1999;46:774777.Google Scholar
Psimaras, D, Carpentier, AF, Rossi, C. Cerebrospinal fluid study in paraneoplastic syndromes. J Neurol Neurosurg Psychiatry 2010;81:4245.Google Scholar
Schmid, AH, Riede, UN. A morphometric study of the cerebellar cortex from patients with carcinoma: a contribution on quantitative aspects in carcinotoxic cerebellar atrophy. Acta Neuropathologica 1974;28:343352.Google Scholar
Storstein, A, Krossnes, B, Vedeler, CA. Autopsy findings in the nervous system and ovarian tumour of two patients with paraneoplastic cerebellar degeneration. Acta Neurol Scand Suppl 2006;183:6970.Google Scholar
Verschuuren, J, Chuang, L, Rosenblum, MK, et al. Inflammatory infiltrates and complete absence of Purkinje cells in anti-Yo-associated paraneoplastic cerebellar degeneration. Acta Neuropathol (Berl) 1996;91:519525.Google Scholar
Giometto, B, Marchiori, GC, Nicolao, P, et al. Sub-acute cerebellar degeneration with anti-Yo autoantibodies: immunohistochemical analysis of the immune reaction in the central nervous system [published erratum appears in Neuropathol Appl Neurobiol 1998 Feb;24(1):2]. Neuropathol Appl Neurobiol 1997;23:468474.Google Scholar
Hormigo, A, Dalmau, J, Rosenblum, MK, River, ME, Posner, JB. Immunological and pathological study of anti-Ri-associated encephalopathy. Ann Neurol 1994;36:896902.Google Scholar
Brieva-Ruiz, L, Diaz-Hurtado, M, Matias-Guiu, X, et al. Anti-Ri-associated paraneoplastic cerebellar degeneration and breast cancer: an autopsy case study. Clin Neurol Neurosurg 2008;110:10441046.Google Scholar
McKeon, A, Tracy, JA, Pittock, SJ, et al. Purkinje cell cytoplasmic autoantibody type 1 accompaniments: the cerebellum and beyond. Arch Neurol 2011;68:12821289.Google Scholar
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
Rojas-Marcos, I, Picard, G, Chinchon, D, et al. Human epidermal growth factor receptor 2 overexpression in breast cancer of patients with anti-Yo–associated paraneoplastic cerebellar degeneration. Neuro-oncology 2012;14:506510.Google Scholar
Openshaw, H, Stuve, O, Antel, JP, et al. Multiple sclerosis flares associated with recombinant granulocyte colony-stimulating factor. Neurology 2000;54:21472150.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
Luque, FA, Furneaux, HM, Ferziger, R, et al. Anti-Ri: an antibody associated with paraneoplastic opsoclonus and breast cancer. Ann Neurol 1991;29:241251.Google Scholar
Shams’ili, S, Grefkens, J, De Leeuw, B, et al. Paraneoplastic cerebellar degeneration associated with antineuronal antibodies: analysis of 50 patients. Brain 2003;126:14091418.Google Scholar
Pittock, SJ, Parisi, JE, McKeon, A, et al. Paraneoplastic jaw dystonia and laryngospasm with antineuronal nuclear autoantibody type 2 (anti-Ri). Arch Neurol 2010;67:11091115.Google Scholar
Simard, C, Vogrig, A, Joubert, B, et al. Clinical spectrum and diagnostic pitfalls of neurologic syndromes with Ri antibodies. Neurol Neuroimmunol Neuroinflamm 2020;7:e699.Google Scholar
Bechich, S, Graus, F, Arboix, A, et al. Anti-Hu-associated paraneoplastic sensory neuropathy and breast cancer. J Neurol 2000;247:552553.Google Scholar
Hoftberger, R, Sabater, L, Velasco, F, et al. Carbonic anhydrase-related protein VIII antibodies and paraneoplastic cerebellar degeneration. Neuropathol Appl Neurobiol 2014;40:650653.Google Scholar
Kerasnoudis, A, Rockhoff, M, Federlein, J, Gold, R, Krogias, C. Isolated ZIC4 antibodies in paraneoplastic cerebellar syndrome with an underlying ovarian tumor. Arch Neurol 2011;68:1073.Google Scholar
David, YB, Warner, E, Levitan, M, et al. Autoimmune paraneoplastic cerebellar degeneration in ovarian carcinoma patients treated with plasmapheresis and immunoglobulin: a case report. Cancer 1996;78:21532156.Google Scholar
Stark, E, Wurster, U, Patzold, U, Sailer, M, Haas, J. Immunological and clinical response to immunosuppressive treatment in paraneoplastic cerebellar degeneration. Arch Neurol 1995;52:814818.Google Scholar
Widdess-Walsh, P, Tavee, JO, Schuele, S, Stevens, GH. Response to intravenous immunoglobulin in anti-Yo associated paraneoplastic cerebellar degeneration: case report and review of the literature. J Neurooncol 2003;63:187190.Google Scholar
Thone, J, Hohaus, A, Lamprecht, S, Bickel, A, Erbguth, F. Effective immunosuppressant therapy with cyclophosphamide and corticosteroids in paraneoplastic cerebellar degeneration. J Neurol Sci 2008;272:171173.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;89:798792.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
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
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
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
Trotter, JL, Hendin, BA, Osterland, K. Cerebellar degeneration with Hodgkin’s disease: an immunological study. Arch Neurol 1976;33:660661.Google Scholar
Graus, F, Dalmau, J, Valldeoriola, F, et al. Immunological characterization of a neuronal antibody (anti-Tr) associated with paraneoplastic cerebellar degeneration and Hodgkin’s disease. J Neuroimmunol 1997;74:5561.Google Scholar
de Graaff, E, Maat, P, Hulsenboom, E, et al. Identification of delta/notch-like epidermal growth factor-related receptor as the Tr antigen in paraneoplastic cerebellar degeneration. Ann Neurol 2012;71:815824.Google Scholar
Khan, N. Paraneoplastic cerebellar degeneration in a patient with anaplastic non-Hodgkin’s lymphoma. BMJ Case Rep 2018;2018:bcr2017224100.Google Scholar
Inui, R, Saito, K, Shimomura, Y, et al. Anti-Ma-associated paraneoplastic cerebellar degeneration in a patient with nodular lymphocyte-predominant Hodgkin lymphoma: a case report. BMC Neurol 2020;20:355.Google Scholar
Hammack, J, Kotanides, H, Rosenblum, MK, Posner, JB. Paraneoplastic cerebellar degeneration. II. Clinical and immunologic findings in 21 patients with Hodgkin’s disease. Neurology 1992;42:19381943.Google Scholar
Greene, M, Lai, Y, Baella, N, Dalmau, J, Lancaster, E. Antibodies to Delta/notch-like epidermal growth factor-related receptor in patients with anti-Tr, paraneoplastic cerebellar degeneration, and Hodgkin lymphoma. JAMA Neurol 2014;71:10031008.Google Scholar
Taniguchi, Y, Tanji, C, Kawai, T, et al. A case report of plasmapheresis in paraneoplastic cerebellar ataxia associated with anti-Tr antibody. Ther Apher Dial 2006;10:9093.Google Scholar
Yeo, KK, Walter, AW, Miller, RE, Dalmau, J. Rituximab as potential therapy for paraneoplastic cerebellar degeneration in pediatric Hodgkin disease. Pediatr Blood Cancer 2012;58:986987.Google Scholar
Peltola, J, Hietaharju, A, Rantala, I, Lehtinen, T, Haapasalo, H. A reversible neuronal antibody (anti-Tr) associated paraneoplastic cerebellar degeneration in Hodgkin’s disease. Acta Neurol Scand 1998;98:360363.Google Scholar
Gungor, S, Kilic, B, Arslan, M, Ozgen, U. Hodgkin’s lymphoma associated with paraneoplastic cerebellar degeneration in children: a case report and review of the literature. Childs Nerv Syst 2017;33:509512.Google Scholar
Avramova, BE, Hristova, T, Yordanova, M, et al. Cerebellar degeneration as a rare paraneoplastic syndrome in a child with Hodgkin lymphoma. J Pediatr Hematol Oncol 2016;38:470472.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
Bataller, L, Wade, DF, Graus, F, et al. Antibodies to Zic4 in paraneoplastic neurologic disorders and small-cell lung cancer. Neurology 2004;62:778782.Google Scholar
Rogemond, V, Honnorat, J. Anti-CV2 autoantibodies and paraneoplastic neurological syndromes. CLin Rev Allerg Immunol 2000;19:5159.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
Pavolucci, L, Giannini, G, Giannoccaro, MP, et al. Paraneoplastic cerebellar degeneration and Lambert-Eaton myasthenia in a patient with merkel cell carcinoma and voltage-gated calcium channel antibodies. Muscle Nerve 2017;56:9981000.Google Scholar
Takasugi, J, Shimamura, M, Koda, T, et al. Paraneoplastic cerebellar degeneration and Lambert–Eaton myasthenic syndrome associated with neuroendocrine carcinoma of the oropharynx. Intern Med (Tokyo, Japan) 2018;57:587590.Google Scholar
Goldstein, JM, Waxman, SG, Vollmer, TL, et al. Subacute cerebellar degeneration and Lambert–Eaton myasthenic syndrome associated with antibodies to voltage-gated calcium channels: differential effect of immunosuppressive therapy on central and peripheral defects. J Neurol Neurosurg Psychiatry 1994;57:11381139.Google Scholar
Clouston, PD, Saper, CB, Arbizu, T, et al. Paraneoplastic cerebellar degeneration. III. Cerebellar degeneration, cancer, and the Lambert–Eaton myasthenic syndrome. Neurology 1992;42:19441950.Google Scholar
Zalewski, N, Lennon, VA, Pittock, SJ, McKeon, A. Calcium channel autoimmunity: Cerebellar ataxia and lambert-eaton syndrome coexisting. Muscle Nerve 2017. doi: 10.1002/mus.26053.Google Scholar
Fukuda, T, Motomura, M, Nakao, Y, et al. Reduction of P/Q-type calcium channels in the postmortem cerebellum of paraneoplastic cerebellar degeneration with Lambert–Eaton myasthenic syndrome. Ann Neurol 2003;53:2128.Google Scholar
Counsell, CE, McLeod, M, Grant, R. Reversal of subacute paraneoplastic cerebellar syndrome with intravenous immunoglobulin. Neurology 1994;44:11841185.Google Scholar
Pellkofer, HL, Voltz, R, Kuempfel, T. Favorable response to rituximab in a patient with anti-VGCC-positive Lambert–Eaton myasthenic syndrome and cerebellar dysfunction. Muscle Nerve 2009;40:305308.Google Scholar
Shimizu, F, Takeshita, Y, Sano, Y, et al. GRP78 antibodies damage the blood–brain barrier and relate to cerebellar degeneration in Lambert–Eaton myasthenic syndrome. Brain 2019;142:22532264.Google Scholar
Graus, F, Lang, B, Pozo-Rosich, P, et al. P/Q type calcium-channel antibodies in paraneoplastic cerebellar degeneration with lung cancer. Neurology 2002;59:764766.Google Scholar
Dalmau, J, Gultekin, SH, Voltz, R, et al. Ma1, a novel neuron- and testis-specific protein, is recognized by the serum of patients with paraneoplastic neurological disorders. Brain 1999;122 (Pt 1):2739.Google Scholar
Ducray, F, Demarquay, G, Graus, F, et al. Seronegative paraneoplastic cerebellar degeneration: the PNS Euronetwork experience. Eur J Neurol 2014;21:731735.Google Scholar
Iyer, JG, Parvathaneni, K, Bhatia, S, et al. Paraneoplastic syndromes (PNS) associated with Merkel cell carcinoma (MCC): a case series of 8 patients highlighting different clinical manifestations. J Am Acad Dermatol 2016;75:541547.Google Scholar
Storstein, A, Raspotnig, M, Vitaliani, R, et al. Prostate cancer, Hu antibodies and paraneoplastic neurological syndromes. J Neurol 2016;263:10011007.Google Scholar
Linnoila, J, Guo, Y, Gadoth, A, et al. Purkinje cell cytoplasmic antibody type I (anti-Yo): predictive of gastrointestinal adenocarcinomas in men. J Neurol Neurosurg Psychiatry 2018;89:11161117.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
Bataller, L, Sabater, L, Saiz, A, et al. Carbonic anhydrase-related protein VIII: autoantigen in paraneoplastic cerebellar degeneration. Ann Neurol 2004;56:575579.Google Scholar
Ammar, H, Brown, SH, Malani, A, et al. A case of paraneoplastic cerebellar ataxia secondary to renal cell carcinoma. Southern Med J 2008;101:556557.Google Scholar
Zhu, Y, Chen, S, Chen, S, et al. An uncommon manifestation of paraneoplastic cerebellar degeneration in a patient with high grade urothelial, carcinoma with squamous differentiation: a case report and literature review. BMC Cancer 2016;16:324.Google Scholar
Henke, C, Rieger, J, Hartmann, S, et al. Paraneoplastic cerebellar degeneration associated with lymphoepithelial carcinoma of the tonsil. BMC Neurol 2013;13:147.Google Scholar
Lakshmaiah, KC, Viveka, BK, Anil Kumar, N, et al. Gastric diffuse large B cell lymphoma presenting as paraneoplastic cerebellar degeneration: case report and review of literature. J Egyptian National Cancer Institute 2013;25:231235.Google Scholar
Sola-Valls, N, Gaba, L, Munoz, E, et al. Paraneoplastic cerebellar degeneration associated with thymic germinoma. J Neurol Sci 2012;320:153155.Google Scholar
Solimena, M, Folli, F, Aparisi, R, Pozza, G, De Camilli, P. Autoantibodies to GABA-ergic neurons and pancreatic beta cells in stiff-man syndrome. N Engl J Med 1990;322:15551560.Google Scholar
Solimena, M, De Camilli, P. Autoimmunity to glutamic acid decarboxylase (GAD) in stiff-man syndrome and insulin-dependent diabetes mellitus. Trends Neurosci 1991;14:452457.Google Scholar
Saiz, A, Blanco, Y, Sabater, L, et al. Spectrum of neurological syndromes associated with glutamic acid decarboxylase antibodies: diagnostic clues for this association. Brain 2008;131:25532563.Google Scholar
Saiz, A, Arpa, J, Sagasta, A, et al. Autoantibodies to glutamic acid decarboxylase in three patients with cerebellar ataxia, late-onset insulin-dependent diabetes mellitus, and polyendocrine autoimmunity. Neurology 1997;49:10261030.Google Scholar
Bataller, L, Valero, C, Diaz, R, et al. Cerebellar ataxia associated with neuroendocrine thymic carcinoma and GAD antibodies. J Neurol Neurosurg Psychiatry 2009;80:696697.Google Scholar
Piccolo, G, Tavazzi, E, Cavallaro, T, et al. Clinico-pathological findings in a patient with progressive cerebellar ataxia, autoimmune polyendocrine syndrome, hepatocellular carcinoma and anti-GAD autoantibodies. J Neurol Sci 2010;290:148149.Google Scholar
Muñiz-Castrillo, S, Vogrig, A, Joubert, B, et al. Transient neurological symptoms preceding cerebellar ataxia with glutamic acid decarboxylase antibodies. Cerebellum 2020;19:715721.Google Scholar
Guasp, M, Sola-Valls, N, Martinez-Hernandez, E, et al. Cerebellar ataxia and autoantibodies restricted to glutamic acid decarboxylase 67 (GAD67). J Neuroimmunol 2016;300:1517.Google Scholar
Gresa-Arribas, N, Arino, H, Martinez-Hernandez, E, et al. Antibodies to inhibitory synaptic proteins in neurological syndromes associated with glutamic acid decarboxylase autoimmunity. PLoS One 2015;10:e0121364.Google Scholar
Planche, V, Marques, A, Ulla, M, Ruivard, M, Durif, F. Intravenous immunoglobulin and rituximab for cerebellar ataxia with glutamic acid decarboxylase autoantibodies. Cerebellum 2014;13:318322.Google Scholar
Mitoma, H, Hadjivassiliou, M, Honnorat, J. Guidelines for treatment of immune-mediated cerebellar ataxias. Cerebellum Ataxias 2015;2:14.Google Scholar
Virgilio, R, Corti, S, Agazzi, P, et al. Effect of steroid treatment in cerebellar ataxia associated with anti-glutamic acid decarboxylase antibodies. J Neurol Neurosurg Psychiatry 2009;80:9596.Google Scholar
Pedroso, JL, Braga-Neto, P, Dutra, LA, Barsottini, OG. Cerebellar ataxia associated to anti-glutamic acid decarboxylase autoantibody (anti-GAD): partial improvement with intravenous immunoglobulin therapy. Arq Neuropsiquiatr 2011;69:993.Google Scholar
Nanri, K, Niwa, H, Mitoma, H, et al. Low-titer anti-GAD-antibody-positive cerebellar ataxia. Cerebellum 2013;12:171175.Google Scholar
Petrijan, T, Menih, M. Low-titre gad antibody-associated late-onset cerebellar ataxia with a significant clinical response to intravenous immunoglobulin treatment. Cerebellum 2017;16:868871.Google Scholar
Meinck, HM, Faber, L, Morgenthaler, N, et al. Antibodies against glutamic acid decarboxylase: prevalence in neurological diseases. J Neurol Neurosurg Psychiatry 2001;71:100103.Google Scholar
Walikonis, JE, Lennon, VA. Radioimmunoassay for glutamic acid decarboxylase (GAD65) autoantibodies as a diagnostic aid for stiff-man syndrome and a correlate of susceptibility to type 1 diabetes mellitus. Mayo Clin Proc 1998;73:11611166.Google Scholar
Sillevis, SP, Kinoshita, A, De, LB, et al. Paraneoplastic cerebellar ataxia due to autoantibodies against a glutamate receptor. N Engl J Med 2000;342:2127.Google Scholar
Iorio, R, Damato, V, Mirabella, M, et al. Cerebellar degeneration associated with mGluR1 autoantibodies as a paraneoplastic manifestation of prostate adenocarcinoma. J Neuroimmunol 2013;263:155158.Google Scholar
Marignier, R, Chenevier, F, Rogemond, V, et al. Metabotropic glutamate receptor type 1 autoantibody-associated cerebellitis: a primary autoimmune disease? Arch Neurol 2010;67:627630.Google Scholar
Lancaster, E, Martinez-Hernandez, E, Titulaer, MJ, et al. Antibodies to metabotropic glutamate receptor 5 in the Ophelia syndrome. Neurology 2011;77:16981701.Google Scholar
Pedroso, JL, Dutra, LA, Espay, AJ, Hoftberger, R, Barsottini, OGP. Video NeuroImages: head titubation in anti-mGluR1 autoantibody-associated cerebellitis. Neurology 2018;90:746747.Google Scholar
Christ, M, Muller, T, Bien, C, et al. Autoimmune encephalitis associated with antibodies against the metabotropic glutamate receptor type 1: case report and review of the literature. Therapeut Adv Neurol Disord 2019;12:1756286419847418.Google Scholar
Kammermeier, PJ, Xiao, B, Tu, JC, Worley, PF, Ikeda, SR. Homer proteins regulate coupling of group I metabotropic glutamate receptors to N-type calcium and M-type potassium channels. J Neurosci 2000;20:72387245.Google Scholar
Zuliani, L, Sabater, L, Saiz, A, et al. Homer 3 autoimmunity in subacute idiopathic cerebellar ataxia. Neurology 2007;68:239240.Google Scholar
Fouka, P, Alexopoulos, H, Chatzi, I, et al. Antibodies to inositol 1,4,5-triphosphate receptor 1 in patients with cerebellar disease. Neurol Neuroimmunol Neuroinflamm 2017;4:e306.Google Scholar
Berzero, G, Hacohen, Y, Komorowski, L, et al. Paraneoplastic cerebellar degeneration associated with anti-ITPR1 antibodies. Neurol Neuroimmunol Neuroinflamm 2017;4:e326.Google Scholar
Xu, X, Ren, H, Li, L, et al. Anti-Homer-3 antibody associated cerebellar ataxia: a rare case report and literature review. J Neuroimmunol 2019;330:155158.Google Scholar
Jarius, S, Ringelstein, M, Haas, J, et al. Inositol 1,4,5-trisphosphate receptor type 1 autoantibodies in paraneoplastic and non-paraneoplastic peripheral neuropathy. J Neuroinflammation 2016;13:278.Google Scholar
Weihua, Z, Haitao, R, Fang, F, et al. Neurochondrin antibody serum positivity in three cases of autoimmune cerebellar ataxia. Cerebellum 2019;18:11371142.Google Scholar
Jarius, S, Martinez-Garcia, P, Hernandez, AL, et al. Two new cases of anti-Ca (anti-ARHGAP26/GRAF) autoantibody-associated cerebellar ataxia. J Neuroinflammation 2013;10:7.Google Scholar
Doss, S, Numann, A, Ziegler, A, et al. Anti-Ca/anti-ARHGAP26 antibodies associated with cerebellar atrophy and cognitive decline. J Neuroimmunol 2014;267:102104.Google Scholar
Wallwitz, U, Brock, S, Schunck, A, et al. From dizziness to severe ataxia and dysarthria: new cases of anti-Ca/ARHGAP26 autoantibody-associated cerebellar ataxia suggest a broad clinical spectrum. J Neuroimmunol 2017;309:7781.Google Scholar
Newman, LS, McKeever, MO, Okano, HJ, Darnell, RB. Beta-NAP, a cerebellar degeneration antigen, is a neuron-specific vesicle coat protein. Cell 1995;82:773783.Google Scholar
Geis, C, Weishaupt, A, Hallermann, S, et al. Stiff person syndrome-associated autoantibodies to amphiphysin mediate reduced GABAergic inhibition. Brain 2010;133:31663180.Google Scholar
Zis, P, Rao, DG, Hoggard, N, Sarrigiannis, PG, Hadjivassiliou, M. Anti-MAG associated cerebellar ataxia and response to rituximab. J Neurol 2018;265:115118.Google Scholar
Dalakas, MC, Teravainen, H, Engel, WK. Tremor as a feature of chronic relapsing and dysgammaglobulinemic polyneuropathies: incidence and management. Arch Neurol 1984;41:711714.Google Scholar
Govert, F, Witt, K, Erro, R, et al. Orthostatic myoclonus associated with Caspr2 antibodies. Neurology 2016;86:13531355.Google Scholar
Becker, EB, Zuliani, L, Pettingill, R, et al. Contactin-associated protein-2 antibodies in non-paraneoplastic cerebellar ataxia. J Neurol Neurosurg Psychiatry 2012;83:437440.Google Scholar
Borsche, M, Hahn, S, Hanssen, H, et al. Sez6l2-antibody-associated progressive cerebellar ataxia: a differential diagnosis of atypical parkinsonism. J Neurol 2019;266:522524.Google Scholar
Bartels, F, Pruss, H, Finke, C. Anti-ARHGAP26 autoantibodies are associated with isolated cognitive impairment. Front Neurol 2018;9:656.Google Scholar
Jarius, S, Wildemann, B, Stocker, W, Moser, A, Wandinger, KP. Psychotic syndrome associated with anti-Ca/ARHGAP26 and voltage-gated potassium channel antibodies. J Neuroimmunol 2015;286:7982.Google Scholar
Rommel, FR, Miske, R, Stocker, W, et al. Chorea minor associated with anti-neurochondrin autoantibodies. Neuropediatrics 2017;48:482483.Google Scholar
Hadjivassiliou, M, Grunewald, RA, Chattopadhyay, AK, et al. Clinical, radiological, neurophysiological, and neuropathological characteristics of gluten ataxia. Lancet 1998;352:15821585.Google Scholar
Sapone, A, Bai, JC, Ciacci, C, et al. Spectrum of gluten-related disorders: consensus on new nomenclature and classification. BMC Med 2012;10:13.Google Scholar
Bushara, KO, Goebel, SU, Shill, H, Goldfarb, LG, Hallett, M. Gluten sensitivity in sporadic and hereditary cerebellar ataxia. Ann Neurol 2001;49:540543.Google Scholar
Hadjivassiliou, M, Sanders, DS, Woodroofe, N, Williamson, C, Grunewald, RA. Gluten ataxia. Cerebellum 2008;7:494498.Google Scholar
Tarlac, V, Kelly, L, Anderson, RP, Bye, N, Storey, E. Coeliac disease patients do not produce antibodies to a common cerebellar epitope. Cerebellum Ataxias 2014;1:18.Google Scholar
Hadjivassiliou, M, Boscolo, S, Davies-Jones, GA, et al. The humoral response in the pathogenesis of gluten ataxia. Neurology 2002;58:12211226.Google Scholar
Boscolo, S, Sarich, A, Lorenzon, A, et al. Gluten ataxia: passive transfer in a mouse model. Ann N Y Acad Sci 2007;1107:319328.Google Scholar
Boscolo, S, Lorenzon, A, Sblattero, D, et al. Anti transglutaminase antibodies cause ataxia in mice. PLoS One 2010;5:e9698.Google Scholar
Bürk, K, Bösch, S, Müller, CA, et al. Sporadic cerebellar ataxia associated with gluten sensitivity. Brain 2001;124:10131019.Google Scholar
Hadjivassiliou, M, Sanders, DS, Grunewald, RA, et al. Gluten sensitivity: from gut to brain. Lancet Neurol 2010;9:318330.Google Scholar
Burk, K, Bosch, S, Muller, CA, et al. Sporadic cerebellar ataxia associated with gluten sensitivity. Brain 2001;124:10131019.Google Scholar
Wilkinson, ID, Hadjivassiliou, M, Dickson, JM, et al. Cerebellar abnormalities on proton MR spectroscopy in gluten ataxia. J Neurol Neurosurg Psychiatry 2005;76:10111013.Google Scholar
Hadjivassiliou, M, Wallis, LI, Hoggard, N, et al. MR spectroscopy and atrophy in Gluten, Friedreich’s and SCA6 ataxias. Acta Neurol Scand 2012;126:138143.Google Scholar
Hadjivassiliou, M, Aeschlimann, P, Sanders, DS, et al. Transglutaminase 6 antibodies in the diagnosis of gluten ataxia. Neurology 2013;80:17401745.Google Scholar
Cascella, NG, Santora, D, Gregory, P, et al. Increased prevalence of transglutaminase 6 antibodies in sera from schizophrenia patients. Schizophr Bull 2013;39:867871.Google Scholar
Hadjivassiliou, M, Davies-Jones, GA, Sanders, DS, Grunewald, RA. Dietary treatment of gluten ataxia. J Neurol Neurosurg Psychiatry 2003;74:12211224.Google Scholar
Souayah, N, Chin, RL, Brannagan, TH, et al. Effect of intravenous immunoglobulin on cerebellar ataxia and neuropathic pain associated with celiac disease. Eur J Neurol 2008;15:13001303.Google Scholar
Thakkar, K, Maricich, SM, Alper, G. Acute ataxia in childhood: 11-year experience at a major pediatric neurology referral center. J Child Neurol 2016;31:11561160.Google Scholar
Van Samkar, A, Poulsen, MNF, Bienfait, HP, Van Leeuwen, RB. Acute cerebellitis in adults: a case report and review of the literature. BMC Res Notes 2017;10:610.Google Scholar
Emelifeonwu, JA, Shetty, J, Kaliaperumal, C, et al. Acute cerebellitis in children: a variable clinical entity. J Child Neurol 2018;33:675684.Google Scholar
Bozzola, E, Bozzola, M, Tozzi, AE, et al. Acute cerebellitis in varicella: a ten year case series and systematic review of the literature. Ital J Pediatr 2014;40:57.Google Scholar
Connolly, AM, Dodson, WE, Prensky, AL, Rust, RS. Course and outcome of acute cerebellar ataxia. Ann Neurol 1994;35:673679.Google Scholar
Lancella, L, Esposito, S, Galli, ML, et al. Acute cerebellitis in children: an eleven year retrospective multicentric study in Italy. Ital J Pediatr 2017;43:54.Google Scholar
Shimokaze, T, Kato, M, Yoshimura, Y, Takahashi, Y, Hayasaka, K. A case of acute cerebellitis accompanied by autoantibodies against glutamate receptor delta2. Brain Dev 2007;29:224226.Google Scholar
Shiihara, T, Kato, M, Konno, A, Takahashi, Y, Hayasaka, K. Acute cerebellar ataxia and consecutive cerebellitis produced by glutamate receptor delta2 autoantibody. Brain Dev 2007;29:254256.Google Scholar
Kubota, M, Takahashi, Y. Steroid-responsive chronic cerebellitis with positive glutamate receptor delta 2 antibody. J Child Neurol 2008;23:228230.Google Scholar
Berridge, G, Menassa, DA, Moloney, T, et al. Glutamate receptor delta2 serum antibodies in pediatric opsoclonus myoclonus ataxia syndrome. Neurology 2018;91:e714e723.Google Scholar
Matsumoto, H, Okabe, S, Hirakawa-Yamada, M, et al. Steroid-responsive focal epilepsy with focal dystonia accompanied by glutamate receptor delta2 antibody. J Neuroimmunol 2012;249:101104.Google Scholar
Fukuoka, T, Takeda, H, Ohe, Y, et al. Anti-glutamate receptor delta2 antibody-positive migrating focal encephalitis. Clin Neurol Neurosurg 2012;114:13511354.Google Scholar
De Bruecker, Y, Claus, F, Demaerel, P, et al. MRI findings in acute cerebellitis. Eur Radiol 2004;14:14781483.Google Scholar
Jabbour, P, Samaha, E, Abi Lahoud, G, et al. Hemicerebellitis mimicking a tumour on MRI. Childs Nerv Syst 2003;19:122125.Google Scholar
Levy, EI, Harris, AE, Omalu, BI, et al. Sudden death from fulminant acute cerebellitis. Pediatr Neurosurg 2001;35:2428.Google Scholar
Ishida, K, Mitoma, H, Wada, Y, et al. Selective loss of Purkinje cells in a patient with anti-glutamic acid decarboxylase antibody-associated cerebellar ataxia. J Neurol Neurosurg Psychiatry 2007;78:190192.Google Scholar
Coesmans, M, Smitt, PA, Linden, DJ, et al. Mechanisms underlying cerebellar motor deficits due to mGluR1-autoantibodies. Ann Neurol 2003;53:325336.Google Scholar
Fabian, RH, Petroff, G. Intraneuronal IgG in the central nervous system: uptake by retrograde axonal transport. Neurology 1987;37:17801784.Google Scholar
Greenlee, JE, Clawson, SA, Hill, KE, et al. Anti-Yo antibody uptake and interaction with its intracellular target antigen causes Purkinje cell death in rat cerebellar slice cultures: a possible mechanism for paraneoplastic cerebellar degeneration in humans with gynecological or breast cancers. PLoS One 2015;10:e0123446.Google Scholar
Graus, F, Illa, I, Agusti, M, et al. Effect of intraventricular injection of an anti-Purkinje cell antibody (anti-Yo) in a guinea pig model. J Neurol Sci 1991;106:8287.Google Scholar
Greenlee, JE, Burns, JB, Rose, JW, Jaeckle, KA, Clawson, S. Uptake of systemically administered human anticerebellar antibody by rat Purkinje cells following blood–brain barrier disruption. Acta Neuropathologica 1995;89:341345.Google Scholar
Sakai, K, Gofuku, M, Kitagawa, Y, Ogasawara, T, Hirose, G. Induction of anti-Purkinje cell antibodies in vivo by immunizing with a recombinant 52-kDa paraneoplastic cerebellar degeneration-associated protein. J Neuroimmunol 1995;60:135141.Google Scholar
McKasson, M, Clardy, SL, Clawson, SA, et al. Voltage-gated calcium channel autoimmune cerebellar degeneration: case and study of cytotoxicity. Neurol Neuroimmunol Neuroinflamm 2016;3:e222.Google Scholar
Martin-Garcia, E, Mannara, F, Gutierrez-Cuesta, J, et al. Intrathecal injection of P/Q type voltage-gated calcium channel antibodies from paraneoplastic cerebellar degeneration cause ataxia in mice. J Neuroimmunol 2013;261:5359.Google Scholar
Mitoma, H, Song, SY, Ishida, K, et al. Presynaptic impairment of cerebellar inhibitory synapses by an autoantibody to glutamate decarboxylase. J Neurol Sci 2000;175:4044.Google Scholar
Manto, M, Mitoma, H, Hampe, CS. Anti-GAD antibodies and the cerebellum: where do we stand? Cerebellum 2018;18:153156.Google Scholar
Chang, T, Alexopoulos, H, Pettingill, P, et al. Immunization against GAD induces antibody binding to GAD-independent antigens and brainstem GABAergic neuronal loss. PLoS One 2013;8:e72921.Google Scholar
Ishida, K, Mitoma, H, Song, SY, et al. Selective suppression of cerebellar GABAergic transmission by an autoantibody to glutamic acid decarboxylase. Ann Neurol 1999;46:263267.Google Scholar
Manto, MU, Laute, MA, Aguera, M, et al. Effects of anti-glutamic acid decarboxylase antibodies associated with neurological diseases. Ann Neurol 2007;61:544551.Google Scholar
Manto, MU, Hampe, CS, Rogemond, V, Honnorat, J. Respective implications of glutamate decarboxylase antibodies in stiff person syndrome and cerebellar ataxia. Orphanet J Rare Dis 2011;6:3.Google Scholar
Manto, M, Honnorat, J, Hampe, CS, et al. Disease-specific monoclonal antibodies targeting glutamate decarboxylase impair GABAergic neurotransmission and affect motor learning and behavioral functions. Front Behav Neurosci 2015;9:78.Google Scholar
Chang, T, Alexopoulos, H, McMenamin, M, et al. Neuronal surface and glutamic acid decarboxylase autoantibodies in nonparaneoplastic stiff person syndrome. JAMA Neurol 2013;70:11401149.Google Scholar
Geis, C, Weishaupt, A, Grunewald, B, et al. Human stiff-person syndrome IgG induces anxious behavior in rats. PLoS One 2011;6:e16775.Google Scholar
Hansen, N, Grunewald, B, Weishaupt, A, et al. Human stiff person syndrome IgG-containing high-titer anti-GAD65 autoantibodies induce motor dysfunction in rats. Exp Neurol 2013;239:202209.Google Scholar
Albert, ML, Austin, LM, Darnell, RB. Detection and treatment of activated T cells in the cerebrospinal fluid of patients with paraneoplastic cerebellar degeneration [see comments]. Ann Neurol 2000;47:917.Google Scholar
Tanaka, M, Tanaka, K, Tsuji, S, et al. Cytotoxic T cell activity against the peptide, AYRARALEL, from Yo protein of patients with the HLA A24 or B27 supertype and paraneoplastic cerebellar degeneration. J Neurol Sci 2001;188:6165.Google Scholar
Skorstad, G, Hestvik, AL, Vartdal, F, Holmoy, T. Cerebrospinal fluid T cell responses against glutamic acid decarboxylase 65 in patients with stiff person syndrome. J Autoimmun 2009;32:2432.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

References

Tan, IL, Mowry, EM, Steele, SU, et al. Brainstem encephalitis: etiologies, treatment, and predictors of outcome. J Neurol 2013;260:23122319.Google Scholar
Moragas, M, Martinez-Yelamos, S, Majos, C, et al. Rhombencephalitis: a series of 97 patients. Medicine (Baltimore) 2011;90:256261.Google Scholar
Carvajal-Gonzalez, A, Leite, MI, Waters, P, et al. Glycine receptor antibodies in PERM and related syndromes: characteristics, clinical features and outcomes. Brain 2014;137: 21782192.Google Scholar
Caviness, JN, Forsyth, PA, Layton, DD, McPhee, TJ. The movement disorder of adult opsoclonus. Mov Disord 1995;10:2227.Google Scholar
Shahrizaila, N, Yuki, N. Bickerstaff brainstem encephalitis and Fisher syndrome: anti-GQ1b antibody syndrome. J Neurol Neurosurg Psychiatry 2013;84:576583.Google Scholar
Tobin, WO, Guo, Y, Krecke, KN, et al. Diagnostic criteria for chronic lymphocytic inflammation with pontine perivascular enhancement responsive to steroids (CLIPPERS). Brain 2017;140:24152425.Google Scholar
Desai, J, Mitchell, WG. Acute cerebellar ataxia, acute cerebellitis, and opsoclonus-myoclonus syndrome. J Child Neurol 2012;27:14821488.Google Scholar
Joubert, B, Rostasy, K, Honnorat, J. Immune-mediated ataxias. Handb Clin Neurol 2018;155:313332.Google Scholar
Luque, FA, Furneaux, HM, Ferziger, R, et al. Anti-Ri: an antibody associated with paraneoplastic opsoclonus and breast cancer. Ann Neurol 1991;29:241251.Google Scholar
Chen, CL, Chou, HH, Chen, CM, Shen, CY, Tsai, SJ. Post-infectious opsoclonus and reversible magnetic resonance imaging changes: a case report and review of the literatures. Acta neurologica Taiwanica 2012;21:7983.Google Scholar
Taieb, G, Mulero, P, Psimaras, D, et al. CLIPPERS and its mimics: evaluation of new criteria for the diagnosis of CLIPPERS. J Neurol Neurosurg Psychiatry 2019;90:10271038.Google Scholar
Saiz, A, Bruna, J, Stourac, P, et al. Anti-Hu-associated brainstem encephalitis. J Neurol Neurosurg Psychiatry 2009;80:404407.Google Scholar
Sabater, L, Gaig, C, Gelpi, E, et al. A novel non-rapid-eye movement and rapid-eye-movement parasomnia with sleep breathing disorder associated with antibodies to IgLON5: a case series, characterisation of the antigen, and post-mortem study. Lancet Neurol 2014;13:575586.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.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
Klaas, JP, Ahlskog, JE, Pittock, SJ, et al. Adult-onset opsoclonus-myoclonus syndrome. Arch Neurol 2012;69:15981607.Google Scholar
Dalmau, J, Graus, F, Villarejo, A, et al. Clinical analysis of anti-Ma2-associated encephalitis. Brain 2004;127:18311844.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
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
Pittock, SJ, Lucchinetti, CF, Lennon, VA. Anti-neuronal nuclear autoantibody type 2: paraneoplastic accompaniments. Ann Neurol 2003;53:580587.Google Scholar
Tintore, M, Rovira, A, Arrambide, G, et al. Brainstem lesions in clinically isolated syndromes. Neurology 2010;75:19331938.Google Scholar
Jarius, S, Kleiter, I, Ruprecht, K, et al. MOG-IgG in NMO and related disorders: a multicenter study of 50 patients. Part 3: Brainstem involvement – frequency, presentation and outcome. J Neuroinflammation 2016;13:281.Google Scholar
Shosha, E, Dubey, D, Palace, J, et al. Area postrema syndrome: frequency, criteria, and severity in AQP4-IgG-positive NMOSD. Neurology 2018;91:e1642e1651.Google Scholar
Mundiyanapurath, S, Jarius, S, Probst, C, et al. GABA-B-receptor antibodies in paraneoplastic brainstem encephalitis. J Neuroimmunol 2013;259:8891.Google Scholar
Jubelt, B, Mihai, C, Li, TM, Veerapaneni, P. Rhombencephalitis/brainstem encephalitis. Curr Neurol Neurosci Rep 2011;11:543552.Google Scholar
Bossart, S, Thurneysen, S, Rushing, E, et al. Case report: encephalitis, with brainstem involvement, following checkpoint inhibitor therapy in metastatic melanoma. Oncologist 2017;22:749753.Google Scholar
Miller, DH, Chard, DT, Ciccarelli, O. Clinically isolated syndromes. Lancet Neurol 2012;11:157169.Google Scholar
Digre, KB. Opsoclonus in adults: report of three cases and review of the literature. Arch Neurol 1986;43:11651175.Google Scholar
Grubbs, J Jr., Trobe, JD, Fisher-Hubbard, A. Opsoclonus-myoclonus syndrome in primary central nervous system lymphoma. J Neuroophthalmol 2016;36:408411.Google Scholar
Matsumura, K, Sonoh, M, Tamaoka, A, Sakuta, M. Syndrome of opsoclonus-myoclonus in hyperosmolar nonketotic coma. Ann Neurol 1985;18:623624.Google Scholar
Pullicino, P, Aquilina, J. Opsoclonus in organophosphate poisoning. Arch Neurol 1989;46:704705.Google Scholar
Wong, A. An update on opsoclonus. Curr Opin Neurol 2007;20:2531.Google Scholar
Kinsbourne, M. Myoclonic encephalopathy of infants. J Neurol Neurosurg Psychiatry 1962;25:271276.Google Scholar
Solomon, GE, Chutorian, AM. Opsoclonus and occult neuroblastoma. N Engl J Med 1968;279:475477.Google Scholar
Klein, A, Schmitt, B, Boltshauser, E. Long-term outcome of ten children with opsoclonus-myoclonus syndrome. Eur J Pediatr 2007;166:359363.Google Scholar
Mitchell, WG, Brumm, VL, Azen, CG, et al. Longitudinal neurodevelopmental evaluation of children with opsoclonus-ataxia. Pediatrics 2005;116:901907.Google Scholar
Catsman-Berrevoets, CE, Aarsen, FK, van Hemsbergen, ML, et al.. Improvement of neurological status and quality of life in children with opsoclonus myoclonus syndrome at long-term follow-up. Pediatr Blood Cancer 2009;53:10481053.Google Scholar
Pang, KK, de Sousa, C, Lang, B, Pike, MG. A prospective study of the presentation and management of dancing eye syndrome/opsoclonus-myoclonus syndrome in the United Kingdom. Eur J Paediatr Neurol 2010;14:156161.Google Scholar
Hasegawa, S, Matsushige, T, Kajimoto, M, et al. A nationwide survey of opsoclonus-myoclonus syndrome in Japanese children. Brain Dev 2015;37:656660.Google Scholar
De, GE, Parodi, S, Conte, M, et al. Long-term follow-up of neuroblastoma-associated opsoclonus-myoclonus-ataxia syndrome. Neuropediatrics 2009;40:103111.Google Scholar
Pranzatelli, MR, Tate, ED, McGee, NR. Multifactorial analysis of opsoclonus-myoclonus syndrome etiology (‘Tumor’ vs. ‘No tumor’) in a cohort of 356 US children. Pediatr Blood Cancer 2018;65:e27097.Google Scholar
Krug, P, Schleiermacher, G, Michon, J, et al. Opsoclonus-myoclonus in children associated or not with neuroblastoma. Eur J Paediatr Neurol 2010;14:400409.Google Scholar
Connolly, AM, Dodson, WE, Prensky, AL, Rust, RS. Course and outcome of acute cerebellar ataxia. Ann Neurol 1994;35:673679.Google Scholar
Matthay, KK, Blaes, F, Hero, B, et al. Opsoclonus myoclonus syndrome in neuroblastoma: a report from a workshop on the dancing eyes syndrome at the advances in neuroblastoma meeting in Genoa, Italy, 2004. Cancer Lett 2005;228:275282.Google Scholar
Pranzatelli, MR, Tate, ED, Alber, M, et al. Rituximab, IVIg, and tetracosactide (acth1-24) combination immunotherapy (‘rite-ci’) for pediatric opsoclonus-myoclonus syndrome: immunomarkers and clinical observations. Neuropediatrics 2018;49:123134.Google Scholar
Pranzatelli, MR, Tate, ED. Dexamethasone, intravenous immunoglobulin, and rituximab combination immunotherapy for pediatric opsoclonus-myoclonus syndrome. Pediatr Neurol 2017;73:4856.Google Scholar
Anderson, NE, Budde-Steffen, C, Rosenblum, MK, et al. Opsoclonus, myoclonus, ataxia, and encephalopathy in adults with cancer: a distinct paraneoplastic syndrome.Medicine 1988;67:100109.Google Scholar
Baringer, JR, Sweeney, VP, Winkler, GF. An acute syndrome of ocular oscillations and truncal myoclonus. Brain 1968;91:473480.Google Scholar
Bataller, L, Graus, F, Saiz, A, Vilchez, J. Clinical course and response to immunotherapy in 24 patients with idiopathic or paraneoplastic opsoclonus-myoclonus syndrome. Brain 2000;124:437443.Google Scholar
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
Belcastro, V, Piola, M, Binda, S, et al. Opsoclonus-myoclonus syndrome associated with human herpes virus-6 rhomboencephalitis. J Neurol Sci 2014;341:165166.Google Scholar
Zaganas, I, Prinianakis, G, Xirouchaki, N, Mavridis, M. Opsoclonus-myoclonus syndrome associated with cytomegalovirus encephalitis. Neurology 2007;68:1636.Google Scholar
Guedes, BF, Vieira Filho, MAA, Listik, C, et al. HIV-associated opsoclonus-myoclonus-ataxia syndrome: early infection, immune reconstitution syndrome or secondary to other diseases? Case report and literature review. J Neurovirol 2018;24:123127.Google Scholar
Ayarza, A, Parisi, V, Altclas, J, et al. Opsoclonus-myoclonus-ataxia syndrome and HIV seroconversion. J Neurol 2009;256:10241025.Google Scholar
van Toorn, R, Rabie, H, Warwick, JM. Opsoclonus-myoclonus in an HIV-infected child on antiretroviral therapy: possible immune reconstitution inflammatory syndrome. Eur J Paediatr Neurol 2005;9:423426.Google Scholar
Kanjanasut, N, Phanthumchinda, K, Bhidayasiri, R. HIV-related opsoclonus-myoclonus-ataxia syndrome: report on two cases. Clin Neurol Neurosurg 2010;112:572574.Google Scholar
Syrbe, S, Merkenschlager, A, Bernhard, MK, et al. Opsoclonus-myoclonus syndrome after adenovirus infection. SpringerPlus 2015;4:636.Google Scholar
Skeie, GO, Eldoen, G, Skeie, BS, et al. Opsoclonus myoclonus syndrome in two cases with neuroborreliosis. Eur J Neurol 2007;14:e12.Google Scholar
Sanguinetti, S, Ramdhani, RA. Opsoclonus myoclonus ataxia syndrome related to the novel coronavirus (COVID-19). J Neuroophthalmol 2021;41:e288e289.Google Scholar
Shah, PB, Desai, SD. Opsoclonus myoclonus ataxia syndrome (OMAS) in the setting of COVID-19 infection. Neurology 2021;96:33.Google Scholar
Tan, AH, Linn, K, Ramli, NM, et al. Opsoclonus-myoclonus-ataxia syndrome associated with dengue virus infection. Parkinsonism Relat Disord 2014;20:13091310.Google Scholar
Akiyama, K, Imazeki, R, Yoshii, F, Koide, T, Muto, J. An adult case of hand, foot, and mouth disease caused by enterovirus 71 accompanied by opsoclonus myoclonica. Tokai J Exp Clin Med 2008;33:143145.Google Scholar
Sahly, A, Gauquelin, L, Sebire, G. Rapid resolution of enterovirus 71-associated opsoclonus myoclonus syndrome on intravenous immunoglobulin. Child Neurol Open 2017;4:2329048x17733215.Google Scholar
Verma, A, Brozman, B. Opsoclonus-myoclonus syndrome following Epstein–Barr virus infection. Neurology 2002;58:11311132.Google Scholar
Ertekin, V, Tan, H. Opsoclonus-myoclonus syndrome attributable to hepatitis C infection. Pediatr Neurol 2010;42:441442.Google Scholar
Singh, D, Sinha, M, Kumar, R, Shukla, R, Ahuja, RC. Opsoclonus-myoclonus syndrome caused by varicella-zoster virus. Ann Ind Acad Neurol 2010;13:211212.Google Scholar
Morita, A, Ishihara, M, Kamei, S, Ishikawa, H. Opsoclonus-myoclonus syndrome following influenza a infection. Intern Med (Tokyo, Japan) 2012;51:24292431.Google Scholar
Sountharalingam, S, Herath, H, Wijegunasinghe, D, Senanayke, S. Opsoclonus myoclonus syndrome in a patient with Japanese encephalitis: a case report. J Med Case Rep 2017;11:294.Google Scholar
Nunes, JC, Bruscato, AM, Walz, R, Lin, K. Opsoclonus-myoclonus syndrome associated with Mycoplasma pneumoniae infection in an elderly patient. J Neurol Sci 2011;305:147148.Google Scholar
Huber, BM, Strozzi, S, Steinlin, M, Aebi, C, Fluri, S. Mycoplasma pneumoniae associated opsoclonus-myoclonus syndrome in three cases. Eur J Pediatr 2010;169:441445.Google Scholar
Kang, BH, Kim, JI. Opsoclonus-myoclonus syndrome associated with mumps virus infection. J Clin Neurol (Seoul, Korea) 2014;10:272275.Google Scholar
D’sa, S, Singh, S, Sowmya, S. Opsoclonus in scrub typhus. J Postgrad Med 2012;58:296297.Google Scholar
Gurkas, E, Gucuyener, K, Yilmaz, U, Havali, C, Demir, E. Opsoclonus-myoclonus syndrome following rotavirus gastroenteritis. Pediatr Int 2014;56:e86e87.Google Scholar
Evans, RW, Welch, K. Opsoclonus in a confirmed case of St. Louis encephalitis. J Neurol Neurosurg Psychiatry 1982;45:660661.Google Scholar
Ahn, AK, Bradley, K, Pina-Garza, JE. Opsoclonus associated with salmonellosis in a 6-week-old infant. J Child Neurol 2014;29:952954.Google Scholar
Flabeau, O, Meissner, W, Foubert-Samier, A, et al. Opsoclonus myoclonus syndrome in the context of Salmonellosis. Mov Disord 2009;24:23062308.Google Scholar
Radic, B, Cajic, I, Petelin Gadze, Z, Sulentic, V, Nankovic, S. A case of adult-onset poststreptococcal opsoclonus-myoclonus syndrome. Acta Neurol Belg 2018;118:541542.Google Scholar
Jones, CE, Smyth, DP, Faust, SN. Opsoclonus-myoclonus syndrome associated with group a streptococcal infection. Pediatr Infect Dis J 2007;26:358359.Google Scholar
Radu, RA, Terecoasa, EO, Ene, A, Bajenaru, OA, Tiu, C. Opsoclonus-myoclonus syndrome associated with West-Nile virus infection: case report and review of the literature. Front Neurol 2018;9:864.Google Scholar
Armangue, T, Titulaer, MJ, Sabater, L, et al. A novel treatment-responsive encephalitis with frequent opsoclonus and teratoma. Ann Neurol 2014;75:435441.Google Scholar
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
Musunuru, K, Kesari, S. Paraneoplastic opsoclonus-myoclonus ataxia associated with non-small-cell lung carcinoma. J Neurooncol 2008;90:213216.Google Scholar
Fumal, A, Jobe, J, Pepin, JL, et al. Intravenous immunoglobulins in paraneoplastic brainstem encephalitis with anti-Ri antibodies. J Neurol 2006;253:13601361.Google Scholar
Jongen, JL, Moll, WJ, Sillevis Smitt, PA, Vecht, CJ, Tijssen, CC. Anti-Ri positive opsoclonus-myoclonus-ataxia in ovarian duct cancer. J Neurol 1998;245:691692.Google Scholar
Scholz, J, Vieregge, P, Ruff, C. Paraneoplastic opsoclonus-myoclonus syndrome in metastatic ovarian carcinoma. J Neurol Neurosurg Psychiatry 1994;57:763764.Google Scholar
Biotti, D, Viaccoz, A, Olivier, N, et al. Opsoclonus, limbic encephalitis, anti-Ma2 antibodies and gastric adenocarcinoma. Eur J Neurol 2012;19:e144e145.Google Scholar
Honnorat, J, Trillet, M, Antoine, JC, et al. Paraneoplastic opsomyoclonus, cerebellar ataxia and encephalopathy associated with anti-Purkinje cell antibodies [letter]. J Neurol 1997;244:333335.Google Scholar
Aggarwal, A, Williams, D. Opsoclonus as a paraneoplastic manifestation of pancreatic carcinoma. J Neurol Neurosurg Psychiatry 1997;63:687688.Google Scholar
Corcia, P, De, TB, Hommet, C, Saudeau, D, Autret, A. Paraneoplastic opsoclonus associated with cancer of the gall bladder. J Neurol Neurosurg Psychiatry 1997;62:293.Google Scholar
Rossor, AM, Perry, F, Botha, A, Norwood, F. Opsoclonus myoclonus syndrome due to squamous cell carcinoma of the oesophagus. BMJ Case Rep 2014;2014:bcr2013202849.Google Scholar
Graus, F, Arino, H, Dalmau, J. Paraneoplastic neurological syndromes in Hodgkin and non-Hodgkin lymphomas. Blood 2014;123:32303238.Google Scholar
Jung, KY, Youn, J, Chung, CS. Opsoclonus-myoclonus syndrome in an adult with malignant melanoma. J Neurol 2006;253:942943.Google Scholar
Dresco, F, Aubin, F, Deveza, E, et al. Paraneoplastic opsoclonus-myoclonus syndrome preceding a mucosal malignant melanoma. Acta dermato-venereologica 2019;99:337338.Google Scholar
Zamecnik, J, Cerny, R, Bartos, A, Jerabek, J, Bojar, M. Paraneoplastic opsoclonus-myoclonus syndrome associated with malignant fibrous histiocytoma: neuropathological findings. Ceskoslovenska patologie 2004;40:6367.Google Scholar
Kearsley, JH, Johnson, P, Halmagyi, GM. Paraneoplastic cerebellar disease: remission with excision of the primary tumor. Arch Neurol 1985;42:12081210.Google Scholar
Mitoma, H, Orimo, S, Sodeyama, N, Tamaki, M. Paraneoplastic opsoclonus-myoclonus syndrome and neurofibrosarcoma. Eur Neurol 1996;36:322.Google Scholar
Prestigiacomo, CJ, Balmaceda, C, Dalmau, J. Anti-Ri-associated paraneoplastic opsoclonus-ataxia syndrome in a man with transitional cell carcinoma. Cancer 2001;91:14231428.Google Scholar
Helmchen, C, Rambold, H, Sprenger, A, Erdmann, C, Binkofski, F. Cerebellar activation in opsoclonus: an fMRI study. Neurology 2003;61:412415.Google Scholar
Taib, BG, Kinshuck, AJ, Milburn-McNulty, P, et al. Opsoclonus-myoclonus syndrome associated with a nasopharyngeal tumor in an adult: a case report. J Med Case Rep 2015;9:128.Google Scholar
Van Diest, D, De Raeve, H, Claes, J, et al. Paraneoplastic opsoclonus-myoclonus-ataxia (OMA) syndrome in an adult patient with esthesioneuroblastoma. J Neurol 2008;255:594596.Google Scholar
De, LS, Terrone, C, Crivellaro, S, et al. Opsoclonus-myoclonus syndrome as a paraneoplastic manifestation of renal cell carcinoma. a case report and review of the literature. Urol Int 2002;68:206208.Google Scholar
Yamaguchi, Y, Wada, M, Tanji, H, et al. Marked improvement in opsoclonus and cerebellar ataxia after the surgical removal of a squamous cell carcinoma of the thymus: a case report. J Neurol Sci 2013;325:156159.Google Scholar
Schwartz, M, Sharf, B, Zidan, J. Opsoclonus as a presenting symptom in thymic carcinoma. J Neurol Neurosurg Psychiatry 1990;53:534.Google Scholar
Dropcho, E, Payne, R. Paraneoplastic opsoclonus-myoclonus: association with medullary thyroid carcinoma and review of the literature. Arch Neurol 1986;43:410415.Google Scholar
Hattori, T, Hirayama, K, Imai, T, et al. Pontine lesion in opsoclonus-myoclonus syndrome shown by MRI. J Neurol Neurosurg Psychiat 1988;51:15721575.Google Scholar
Bartos, A. Effective high-dose clonazepam treatment in two patients with opsoclonus and myoclonus: GABAergic hypothesis. Eur Neurol 2006;56:240242.Google Scholar
Fernandes, TD, Bazan, R, Betting, LE, da Rocha, FC. Topiramate effect in opsoclonus-myoclonus-ataxia syndrome. Arch Neurol 2012;69:133.Google Scholar
Oh, SY, Kim, JS, Dieterich, M. Update on opsoclonus-myoclonus syndrome in adults. J Neurol 2019;266:15411548.Google Scholar
Ross, AT, Zeman, W. Opsoclonus, occult carcinoma, and chemical pathology in dentate nuclei. Arch Neurol 1967;17:546551.Google Scholar
Ridley, A, Kennard, C, Scholtz, CL, et al. Omnipause neurons in two cases of opsoclonus associated with oat cell carcinoma of the lung. Brain 1987;110:16991709.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
Oh, SY, Boegle, R, Eulenburg, PZ, et al. Longitudinal multi-modal neuroimaging in opsoclonus-myoclonus syndrome. J Neurol 2017;264:512519.Google Scholar
Hormigo, A, Dalmau, J, Rosenblum, MK, River, ME, Posner, JB. Immunological and pathological study of anti-Ri-associated encephalopathy. Ann Neurol 1994;36:896902.Google Scholar
Pranzatelli, MR, Travelstead, AL, Tate, ED, et al. B- and T-cell markers in opsoclonus-myoclonus syndrome: immunophenotyping of CSF lymphocytes. Neurology 2004;62:15261532.Google Scholar
Fuhlhuber, V, Bick, S, Kirsten, A, et al. Elevated B-cell activating factor BAFF, but not APRIL, correlates with CSF cerebellar autoantibodies in pediatric opsoclonus-myoclonus syndrome. J Neuroimmunol 2009;210:8791.Google Scholar
Bickerstaff, ER, Cloake, PC. Mesencephalitis and rhombencephalitis. Br Med J 1951;2:7781.Google Scholar
Bickerstaff, ER. Brain-stem encephalitis: further observations on a grave syndrome with benign prognosis. Br Med J 1957;1:13841387.Google Scholar
Fisher, M. An unusual variant of acute idiopathic polyneuritis (syndrome of ophthalmoplegia, ataxia and areflexia). N Engl J Med 1956;255:5765.Google Scholar
Odaka, M, Yuki, N, Hirata, K. Anti-GQ1b IgG antibody syndrome: clinical and immunological range. J Neurol Neurosurg Psychiatry 2001;70:5055.Google Scholar
Ito, M, Kuwabara, S, Odaka, M, et al. Bickerstaff’s brainstem encephalitis and Fisher syndrome form a continuous spectrum: clinical analysis of 581 cases. J Neurol 2008;255:674682.Google Scholar
Odaka, M, Yuki, N, Hirata, K. Patients with chronic inflammatory demyelinating polyneuropathy initially diagnosed as Guillain–Barre syndrome. J Neurol 2003;250:913916.Google Scholar
Liu, JX, Willison, HJ, Pedrosa-Domellof, F. Immunolocalization of GQ1b and related gangliosides in human extraocular neuromuscular junctions and muscle spindles. Invest Ophthalmol Vis Sci 2009;50:32263232.Google Scholar
Chiba, A, Kusunoki, S, Obata, H, Machinami, R, Kanazawa, I. Ganglioside composition of the human cranial nerves, with special reference to pathophysiology of Miller Fisher syndrome. Brain Res 1997;745:3236.Google Scholar
Zeiner, PS, Brandhofe, A, Muller-Eschner, M, Steinmetz, H, Pfeilschifter, W. Area postrema syndrome as frequent feature of Bickerstaff brainstem encephalitis. Ann Clin Transl Neurol 2018;5:15341542.Google Scholar
Santoro, JD, Lazzareschi, DV, Campen, CJ, Van Haren, KP. Pediatric Bickerstaff brainstem encephalitis: a systematic review of literature and case series. J Neurol 2018;265:141150.Google Scholar
Koga, M, Kusunoki, S, Kaida, K, et al. Nationwide survey of patients in Japan with Bickerstaff brainstem encephalitis: epidemiological and clinical characteristics. J Neurol Neurosurg Psychiatry 2012;83:12101215.Google Scholar
Odaka, M, Yuki, N, Yamada, M, et al. Bickerstaff’s brainstem encephalitis: clinical features of 62 cases and a subgroup associated with Guillain–Barre syndrome. Brain 2003;126:22792290.Google Scholar
Wakerley, BR, Uncini, A, Yuki, N. Guillain–Barre and Miller Fisher syndromes: new diagnostic classification. Nat Rev Neurol 2014;10:537544.Google Scholar
Yoshikawa, K, Kuwahara, M, Morikawa, M, Kusunoki, S. Bickerstaff brainstem encephalitis with or without anti-GQ1b antibody. Neurol Neuroimmunol Neuroinflamm 2020;7:e889.Google Scholar
Yuki, N, Shahrizaila, N. How do we identify infectious agents that trigger Guillain–Barre syndrome, Fisher syndrome and Bickerstaff brainstem encephalitis? J Neurol Sci 2011;302:15.Google Scholar
Pittock, SJ, Kryzer, TJ, Lennon, VA. Paraneoplastic antibodies coexist and predict cancer, not neurological syndrome. Ann Neurol 2004;56:715719.Google Scholar
Dauvilliers, Y, Bauer, J, Rigau, V, et al. Hypothalamic immunopathology in anti-Ma-associated diencephalitis with narcolepsy-cataplexy. JAMA Neurol 2013;70:13051310.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
Blaes, F. Paraneopla`stic brain stem encephalitis. Curr Treat Options Neurol 2013;15:201209.Google Scholar
Pruss, H, Voltz, R, Gelderblom, H, et al. Spontaneous remission of anti-Ma associated paraneoplastic mesodiencephalic and brainstem encephalitis. J Neurol 2008;255:292294.Google Scholar
Ortega Suero, G, Sola-Valls, N, Escudero, D, Saiz, A, Graus, F. Anti-Ma and anti-Ma2-associated paraneoplastic neurological syndromes. Neurologia 2018;33:1827.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
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
Simard, C, Vogrig, A, Joubert, B, et al. Clinical spectrum and diagnostic pitfalls of neurologic syndromes with Ri antibodies. Neurol Neuroimmunol Neuroinflamm 2020;7:e699.Google Scholar
Adams, C, McKeon, A, Silber, MH, Kumar, R. Narcolepsy, REM sleep behavior disorder, and supranuclear gaze palsy associated with Ma1 and Ma2 antibodies and tonsillar carcinoma. Arch Neurol 2011;68:521524.Google Scholar
Garcia-Reitboeck, P, Thompson, G, Johns, P, et al. Upbeat nystagmus in anti-Ma2 encephalitis. Practical Neurol 2014;14:3638.Google Scholar
Wagner, J, Schankin, C, Birnbaum, T, Popperl, G, Straube, A. Ocular motor and lid apraxia as initial symptom of anti-Ma1/Ma2-associated encephalitis. Neurology 2009;72:466467.Google Scholar
Barnett, M, Prosser, J, Sutton, I, et al. Paraneoplastic brain stem encephalitis in a woman with anti-Ma2 antibody. J Neurol Neurosurg Psychiatry 2001;70:222225.Google Scholar
Compta, Y, Iranzo, A, Santamaria, J, Casamitjana, R, Graus, F. REM sleep behavior disorder and narcoleptic features in anti-Ma2-associated encephalitis. Sleep 2007;30:767769.Google Scholar
Edvardsson, B. Anti-Ma2-positive paraneoplastic brainstem encephalitis associated with prostatic adenocarcinoma. Neurol India 2011;59:912913.Google Scholar
Rosenfeld, MR, Eichen, JG, Wade, DF, Posner, JB, Dalmau, J. Molecular and clinical diversity in paraneoplastic immunity to Ma proteins. Ann Neurol 2001;50:339348.Google Scholar
Kraemer, M, Berlit, P. Anti-Ma2 antibodies in B-cell primary CNS lymphoma. J Neurol 2007;254:12861287.Google Scholar
Desestret, V, Didelot, A, Meyronet, D, et al. Neurosarcoidosis with diencephalitis and anti-Ma2 antibodies. Neurology 2010;74:772774.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
Yshii, LM, Gebauer, CM, Pignolet, B, et al. CTLA4 blockade elicits paraneoplastic neurological disease in a mouse model. Brain 2016;139:29232934.Google Scholar
Greene, JJ, Keefe, MW, Harris, JP, Matsuoka, AJ. Paraneoplastic syndrome: a masquerade of autoimmune inner ear disease. Otol Neurotol 2015;36:e310.Google Scholar
Narayan, RN, McKeon, A, Fife, TD. Autoimmune vestibulocerebellar syndromes. Semin Neurol 2020;40:97115.Google Scholar
Budde-Steffen, C, Anderson, NE, Rosenblum, MK, et al. An antineuronal autoantibody in paraneoplastic opsoclonus. Ann Neurol 1988;23:528531.Google Scholar
Sutton, IJ, Barnett, MH, Watson, JD, Ell, JJ, Dalmau, J. Paraneoplastic brainstem encephalitis and anti-Ri antibodies. J Neurol 2002;249:15971598.Google Scholar
Kastrup, O, Meyring, S, Diener, HC. Atypical paraneoplastic brainstem encephalitis associated with anti-ri-antibodies due to thymic carcinoma with possible clinical response to immunoglobulins. Eur Neurol 2001;45:285287.Google Scholar
Boch, M, Rinke, A, Rexin, P, et al. Paraneoplastic brainstem encephalitis in a patient with exceptionally long course of a metastasized neuroendocrine rectum neoplasm. BMC Cancer 2014;14:691.Google Scholar
Kim, KJ, Yun, JY, Lee, JY, Kim, YE, Jeon, BS. Ondine’s curse in anti-Ri antibody associated paraneoplastic brainstem syndrome. Sleep Med 2013;14:382.Google Scholar
Casado, JL, Gil-Peralta, A, Graus, F, et al. Anti-Ri antibodies associated with opsoclonus and progressive encephalomyelitis with rigidity. Neurology 1994;44:15211522.Google Scholar
Thumen, A, Moser, A. An uncommon paraneoplastic Ri-positive opsoclonus-myoclonus-like syndrome and stiff-person syndrome with elevated glutamate/GABA ratio in the cerebrospinal fluid after breast cancer. J Neurol 2010;257:12151217.Google Scholar
Pittock, SJ, Parisi, JE, McKeon, A, et al. Paraneoplastic jaw dystonia and laryngospasm with antineuronal nuclear autoantibody type 2 (anti-Ri). Arch Neurol 2010;67:11091115.Google Scholar
Rojas-Marcos, I, Picard, G, Chinchon, D, et al. Human epidermal growth factor receptor 2 overexpression in breast cancer of patients with anti-Yo–associated paraneoplastic cerebellar degeneration. Neuro-oncology 2012;14:506510.Google Scholar
Kim, H, Lim, Y, Kim, KK. Anti-ri-antibody-associated paraneoplastic syndrome in a man with breast cancer showing a reversible pontine lesion on MRI. J Clin Neurol (Seoul, Korea) 2009;5:151152.Google Scholar
Tesseki, K, Kataoka, H, Terashima, M, et al. Biphasic paraneoplastic brainstem encephalitis associated with anti-Ri antibody. J Neurooncol 2010;100:141143.Google Scholar
Angstwurm, K, Schielke, E, Zimmer, C, Kivelitz, D, Weber, JR. Superficial siderosis of the central nervous system: response to steroid therapy. J Neurol 2002;249:12231225.Google Scholar
Wirtz, PW, Sillevis Smitt, PA, Hoff, JI, et al. Anti-Ri antibody positive opsoclonus-myoclonus in a male patient with breast carcinoma. J Neurol 2002;249:17101712.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
Saiz, A, Bruna, J, Stourac, P, et al. Anti-Hu-associated brainstem encephalitis. J Neurol Neurosurg Psychiatry 2009;80:404407.Google Scholar
Montaut, S, Mallaret, M, Laguna, AE, et al. Anti-Hu-associated brainstem encephalitis with ganglioneuroblastoma in a young adult. J Neurol 2014;261:18221824.Google Scholar
Malek, N, Damian, M. Trismus caused by paraneoplastic brainstem encephalitis. Practical Neurol 2018;18:146150.Google Scholar
Mendoza, M, Latorre, JG. Pearls and oy-sters: reversible Ondine’s curse in a case of lateral medullary infarction. Neurology 2013;80:e13e16.Google Scholar
Kay, L, Bauer, S, Koczulla, AR, et al. Ondine’s curse and temporal lobe seizures: anti-Hu- and Zic4-associated paraneoplastic brainstem and limbic encephalitis. Eur J Neurol 2018;25:e59e60.Google Scholar
Najjar, M, Taylor, A, Agrawal, S, et al. Anti-Hu paraneoplastic brainstem encephalitis caused by a pancreatic neuroendocrine tumor presenting with central hypoventilation. J Clin Neurosci 2017;40:7273.Google Scholar
Pittock, SJ, Debruyne, J, Krecke, KN, et al. Chronic lymphocytic inflammation with pontine perivascular enhancement responsive to steroids (CLIPPERS). Brain 2010;133:26262634.Google Scholar
Dudesek, A, Rimmele, F, Tesar, S, et al. CLIPPERS: chronic lymphocytic inflammation with pontine perivascular enhancement responsive to steroids. Review of an increasingly recognized entity within the spectrum of inflammatory central nervous system disorders. Clin Exp Immunol 2014;175:385396.Google Scholar
Taieb, G, Labauge, P. CLIPPERS, a possible symptomatic lymphohistiocytic immune reaction. Brain 2018;141:e5.Google Scholar
Taieb, G, Kaphan, E, Duflos, C, et al. Hemophagocytic lymphohistiocytosis gene mutations in adult patients presenting with CLIPPERS-like syndrome. Neurol Neuroimmunol Neuroinflamm 2021;8:e970.Google Scholar
Griffin, G, Shenoi, S, Hughes, GC. Hemophagocytic lymphohistiocytosis: an update on pathogenesis, diagnosis, and therapy. Best Pract Res Clin Rheumatol 2020;34:101515.Google Scholar
Benson, LA, Li, H, Henderson, LA, et al. Pediatric CNS-isolated hemophagocytic lymphohistiocytosis. Neurol Neuroimmunol Neuroinflamm 2019;6:e560.Google Scholar
Veerapandiyan, A, Chaudhari, A, Deo, P, Ming, X. Chronic lymphocytic inflammation with pontine perivascular enhancement responsive to steroids (CLIPPERS): a pediatric case report with six year follow-up. Mult Scler Relat Disord 2017;17:9598.Google Scholar
Sa, M, Green, L, Abdel-Mannan, O, et al. Is chronic lymphocytic inflammation with pontine perivascular enhancement responsive to steroids (CLIPPERS) in children the same condition as in adults? Dev Med Child Neurol 2019;61:490496.Google Scholar
Ortega, MR, Usmani, N, Parra-Herran, C, et al. CLIPPERS complicating multiple sclerosis causing concerns of CNS lymphoma. Neurology 2012;79:715716.Google Scholar
Ferreira, RM, Machado, G, Souza, AS, Lin, K, Correa-Neto, Y. CLIPPERS-like MRI findings in a patient with multiple sclerosis. J Neurol Sci 2013;327:6162.Google Scholar
Blaabjerg, M, Ruprecht, K, Sinnecker, T, et al. Widespread inflammation in CLIPPERS syndrome indicated by autopsy and ultra-high-field 7T MRI. Neurol Neuroimmunol Neuroinflamm 2016;3:e226.Google Scholar
De Graaff, HJ, Wattjes, MP, Rozemuller-Kwakkel, AJ, Petzold, A, Killestein, J. Fatal B-cell lymphoma following chronic lymphocytic inflammation with pontine perivascular enhancement responsive to steroids. JAMA Neurol 2013;70:915918.Google Scholar
Buttmann, M, Metz, I, Brecht, I, Bruck, W, Warmuth-Metz, M. Atypical chronic lymphocytic inflammation with pontocerebellar perivascular enhancement responsive to steroids (CLIPPERS), primary angiitis of the CNS mimicking CLIPPERS or overlap syndrome? A case report. J Neurol Sci 2013;324:183186.Google Scholar
Taieb, G, Uro-Coste, E, Clanet, M, et al. A central nervous system B-cell lymphoma arising two years after initial diagnosis of CLIPPERS. J Neurol Sci 2014;344:224226.Google Scholar
Taieb, G, Duflos, C, Renard, D, et al. Long-term outcomes of CLIPPERS (chronic lymphocytic inflammation with pontine perivascular enhancement responsive to steroids) in a consecutive series of 12 patients. Arch Neurol 2012;69:847855.Google Scholar
Taieb, G, Allou, T, Labauge, P. Therapeutic approaches in CLIPPERS. Curr Treat Options Neurol 2017;19:17.Google Scholar

References

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.Google Scholar
Pelkey, KA, Chittajallu, R, Craig, MT, et al. Hippocampal GABAergic inhibitory interneurons. Physiol Rev 2017;97:16191747.Google Scholar
Moore, LA, Trussell, LO. Corelease of inhibitory neurotransmitters in the mouse auditory midbrain. J Neurosci 2017;37:94539464.Google Scholar
Wojcik, SM, Katsurabayashi, S, Guillemin, I, et al. A shared vesicular carrier allows synaptic corelease of GABA and glycine. Neuron 2006;50:575587.Google Scholar
Hirose, S. Mutant GABA(A) receptor subunits in genetic (idiopathic) epilepsy. Progr Brain Res 2014;213:5585.Google Scholar
Dreissen, YE, Tijssen, MA. The startle syndromes: physiology and treatment. Epilepsia 2012;53(Suppl 7):311.Google Scholar
Warich-Kirches, M, Von Bossanyi, P, Treuheit, T, et al. Stiff-man syndrome: possible autoimmune etiology targeted against GABA-ergic cells. Clin Neuropathol 1997;16:214219.Google Scholar
De Felipe, J, Marco, P, Fairen, A, Jones, EG. Inhibitory synaptogenesis in mouse somatosensory cortex. Cereb Cortex 1997;7:619634.Google Scholar
Betz, H, Langosch, D, Hoch, W, et al. Structure and expression of inhibitory glycine receptors. Adv Exp Med Biol 1991;287:421429.Google Scholar
Jacob, TC, Moss, SJ, Jurd, R. GABA(A) receptor trafficking and its role in the dynamic modulation of neuronal inhibition. Nat Rev Neurosci 2008;9:331343.Google Scholar
Tamas, G, Lorincz, A, Simon, A, Szabadics, J. Identified sources and targets of slow inhibition in the neocortex. Science 2003;299:19021905.Google Scholar
Butler, MH, Hayashi, A, Ohkoshi, N, et al. Autoimmunity to gephyrin in stiff-man syndrome. Neuron 2000;26:307312.Google Scholar
Kasaragod, VB, Schindelin, H. Structure–function relationships of glycine and GABAA receptors and their interplay with the scaffolding protein gephyrin. Front Molec Neurosci 2018;11:317.Google Scholar
Spatola, M, Petit-Pedrol, M, Simabukuro, MM, et al. Investigations in GABAA receptor antibody-associated encephalitis. Neurology 2017;88:10121020.Google Scholar
Bode, A, Lynch, JW. The impact of human hyperekplexia mutations on glycine receptor structure and function. Molec Brain 2014;7:2.Google Scholar
Schaefer, N, Roemer, V, Janzen, D, Villmann, C. Impaired glycine receptor trafficking in neurological diseases. Front Molec Neurosci 2018;11:291.Google Scholar
Macdonald, RL, Kang, JQ, Gallagher, MJ. Mutations in GABAA receptor subunits associated with genetic epilepsies. J Physiol 2010;588:18611869.Google Scholar
Pruss, H, Kirmse, K. Pathogenic role of autoantibodies against inhibitory synapses. Brain Res 2018;1701:146152.Google Scholar
Erlander, MG, Tobin, AJ. The structural and functional heterogeneity of glutamic acid decarboxylase: a review. Neurochem Research 1991;16:215226.Google Scholar
Werner, C, Pauli, M, Doose, S, et al. Human autoantibodies to amphiphysin induce defective presynaptic vesicle dynamics and composition. Brain 2016;139:365379.Google Scholar
McKeon, A, Robinson, MT, McEvoy, KM, et al. Stiff-man syndrome and variants: clinical course, treatments, and outcomes. Arch Neurol 2012;69:230238.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
Carvajal-Gonzalez, A, Leite, MI, Waters, P, et al. Glycine receptor antibodies in PERM and related syndromes: characteristics, clinical features and outcomes. Brain 2014;137:21782192.Google Scholar
Piepgras, J, Holtje, M, Michel, K, et al. Anti-DPPX encephalitis: pathogenic effects of antibodies on gut and brain neurons. Neurology 2015;85:890897.Google Scholar
Balint, B, Jarius, S, Nagel, S, et al. Progressive encephalomyelitis with rigidity and myoclonus: a new variant with DPPX antibodies. Neurology 2014;82:15211528.Google Scholar
Gresa-Arribas, N, Arino, H, Martinez-Hernandez, E, et al. Antibodies to inhibitory synaptic proteins in neurological syndromes associated with glutamic acid decarboxylase autoimmunity. PLoS One 2015;10:e0121364.Google Scholar
Ariño, H, Hoftberger, R, Gresa-Arribas, N, et al. Paraneoplastic neurological syndromes and glutamic acid decarboxylase antibodies. JAMA Neurol 2015;72:874881.Google Scholar
Pittock, SJ, Lucchinetti, CF, Parisi, JE, et al. Amphiphysin autoimmunity: paraneoplastic accompaniments. Ann Neurol 2005;58:96107.Google Scholar
Moersch, F, Woltman, H. Progressive fluctuating muscular rigidity and spasm (‘stiffman syndrome’): report of a case and some observations in 13 other cases. Mayo Clin Proc 1956;31:421427.Google Scholar
Solimena, M, Folli, F, Denis-Donini, S, et al. Autoantibodies to glutamic acid decarboxylase in a patient with stiff-man syndrome, epilepsy, and type I diabetes mellitus. N Engl J Med 1988;318:10121020.Google Scholar
Solimena, M, Folli, F, Aparisi, R, Pozza, G, De Camilli, P. Autoantibodies to GABA-ergic neurons and pancreatic beta cells in stiff-man syndrome. N Engl J Med 1990;322:15551560.Google Scholar
Levy, LM, Dalakas, MC, Floeter, MK. The stiff-person syndrome: an autoimmune disorder affecting neurotransmission of gamma-aminobutyric acid. Ann Intern Med 1999;131:522530.Google Scholar
Martinez-Hernandez, E, Arino, H, McKeon, A, et al. Clinical and immunological investigations in 121 patients with stiff-person spectrum disorder. JAMA Neurol 2016;73:714720.Google Scholar
Saiz, A, Graus, F, Valldeoriola, F, Valls-Sole, J, Tolosa, E. Stiff-leg syndrome: a focal form of stiff-man syndrome. Ann Neurol 1998;43:400403.Google Scholar
Brown, P, Rothwell, JC, Marsden, CD. The stiff leg syndrome. J Neurol Neurosurg Psychiatry 1997;62:3137.Google Scholar
Leigh, PN, Rothwell, JC, Traub, M, Marsden, CD. A patient with reflex myoclonus and muscle rigidity: ‘jerking stiff-man syndrome’. J Neurol Neurosurg Psychiatry 1980;43:11251131.Google Scholar
Meinck, HM, Thompson, PD. Stiff man syndrome and related conditions. Mov Disord 2002;17:853866.Google Scholar
Rakocevic, G, Alexopoulos, H, Dalakas, MC. Quantitative clinical and autoimmune assessments in stiff person syndrome: evidence for a progressive disorder. BMC Neurol 2019;19:1.Google Scholar
Henningsen, P, Meinck, HM. Specific phobia is a frequent non-motor feature in stiff man syndrome. J Neurol Neurosurg Psychiatry 2003;74:462465.Google Scholar
Ameli, R, Snow, J, Rakocevic, G, Dalakas, MC. A neuropsychological assessment of phobias in patients with stiff person syndrome. Neurology 2005;64:19611963.Google Scholar
Barker, RA, Revesz, T, Thom, M, Marsden, CD, Brown, P. Review of 23 patients affected by the stiff man syndrome: clinical subdivision into stiff trunk (man) syndrome, stiff limb syndrome, and progressive encephalomyelitis with rigidity. J Neurol Neurosurg Psychiatry 1998;65:633640.Google Scholar
Saiz, A, Blanco, Y, Sabater, L, et al. Spectrum of neurological syndromes associated with glutamic acid decarboxylase antibodies: diagnostic clues for this association. Brain 2008;131:25532563.Google Scholar
Maddison, P, Mills, KR, Newsom-Davis, J. Clinical electrophysiological characterization of the acquired neuromyotonia phenotype of autoimmune peripheral nerve hyperexcitability. Muscle Nerve 2006;33:801808.Google Scholar
Mamoli, B, Heiss, WD, Maida, E, Podreka, I. Electrophysiological studies on the ‘stiff-man’ syndrome. J Neurol 1977;217:111121.Google Scholar
Espay, AJ, Chen, R. Rigidity and spasms from autoimmune encephalomyelopathies: stiff-person syndrome. Muscle Nerve 2006;34:677690.Google Scholar
Floeter, MK, Valls-Sole, J, Toro, C, Jacobowitz, D, Hallett, M. Physiologic studies of spinal inhibitory circuits in patients with stiff-person syndrome. Neurology 1998;51:8593.Google Scholar
Meinck, HM, Kuster, S, Benecke, R, Conrad, B. The flexor reflex–influence of stimulus parameters on the reflex response. Electroencephalogr Clin Neurophysiol 1985;61:287298.Google Scholar
Meinck, HM, Ricker, K, Conrad, B. The stiff-man syndrome: new pathophysiological aspects from abnormal exteroceptive reflexes and the response to clomipramine, clonidine, and tizanidine. J Neurol Neurosurg Psychiatry 1984;47:280287.Google Scholar
Sandbrink, F, Syed, NA, Fujii, MD, Dalakas, MC, Floeter, MK. Motor cortex excitability in stiff-person syndrome. Brain 2000;123:22312239.Google Scholar
Koerner, C, Wieland, B, Richter, W, Meinck, HM. Stiff-person syndromes: motor cortex hyperexcitability correlates with anti-GAD autoimmunity. Neurology 2004;62:13571362.Google Scholar
Dalakas, MC, Li, M, Fujii, M, Jacobowitz, DM. Stiff person syndrome: quantification, specificity, and intrathecal synthesis of GAD65 antibodies. Neurology 2001;57:780784.Google Scholar
Ellis, TM, Atkinson, MA. The clinical significance of an autoimmune response against glutamic acid decarboxylase. Nat Med 1996;2:148153.Google Scholar
Saiz, A, Arpa, J, Sagasta, A, et al. Autoantibodies to glutamic acid decarboxylase in three patients with cerebellar ataxia, late-onset insulin-dependent diabetes mellitus, and polyendocrine autoimmunity. Neurology 1997;49:10261030.Google Scholar
Meinck, HM, Faber, L, Morgenthaler, N, et al. Antibodies against glutamic acid decarboxylase: prevalence in neurological diseases. J Neurol Neurosurg Psychiatry 2001;71:100103.Google Scholar
Walikonis, JE, Lennon, VA. Radioimmunoassay for glutamic acid decarboxylase (GAD65) autoantibodies as a diagnostic aid for stiff-man syndrome and a correlate of susceptibility to type 1 diabetes mellitus. Mayo Clin Proc 1998;73:11611166.Google Scholar
Schmidli, RS, Colman, PG, Bonifacio, E. Disease sensitivity and specificity of 52 assays for glutamic acid decarboxylase antibodies: the Second International GADAB Workshop. Diabetes 1995;44:636640.Google Scholar
Graus, F, Saiz, A, Dalmau, J. GAD antibodies in neurological disorders: insights and challenges. Nat Rev Neurol 2020;16:353365.Google Scholar
McKeon, A, Martinez-Hernandez, E, Lancaster, E, et al. Glycine receptor autoimmune spectrum with stiff-man syndrome phenotype. JAMA Neurol 2013;70:4450.Google Scholar
Brown, P, Marsden, CD. The stiff man and stiff man plus syndromes. J Neurol 1999;246:648652.Google Scholar
Lorish, TR, Thorsteinsson, G, Howard, FM Jr. Stiff-man syndrome updated. Mayo Clin Proc 1989;64:629636.Google Scholar
Wu, Y, Matsui, H, Tomizawa, K. Amphiphysin I and regulation of synaptic vesicle endocytosis. Acta medica Okayama 2009;63:305323.Google Scholar
Folli, F, Solimena, M, Cofiell, R, et al. Autoantibodies to a 128-kd synaptic protein in three women with the stiff-man syndrome and breast cancer. N Engl J Med 1993;328:546551.Google Scholar
De Camilli, P, Thomas, A, Cofiell, R, et al. The synaptic vesicle-associated protein amphiphysin is the 128-kD autoantigen of stiff-man syndrome with breast cancer. J Exp Med 1993;178:22192223.Google Scholar
Murinson, BB, Guarnaccia, JB. Stiff-person syndrome with amphiphysin antibodies: distinctive features of a rare disease. Neurology 2008;71:19551958.Google Scholar
Saiz, A, Dalmau, J, Butler, MH, et al. Anti-amphiphysin I antibodies in patients with paraneoplastic neurological disorders associated with small cell lung carcinoma. J Neurol Neurosurg Psychiatry 1999;66:214217.Google Scholar
Nguyen-Huu, BK, Urban, PP, Schreckenberger, M, Dieterich, M, Werhahn, KJ. Antiamphiphysin-positive stiff-person syndrome associated with small cell lung cancer. Mov Disord 2006;21:12851287.Google Scholar
Dubey, D, Jitprapaikulsan, J, Bi, H, et al. Amphiphysin-IgG autoimmune neuropathy: a recognizable clinicopathologic syndrome. Neurology 2019;93:e1873e1880.Google Scholar
Hagiwara, H, Enomoto-Nakatani, S, Sakai, K, et al. Stiff-person syndrome associated with invasive thymoma: a case report. J Neurol Sci 2001;193:5962.Google Scholar
Thomas, S, Critchley, P, Lawden, M, et al. Stiff person syndrome with eye movement abnormality, myasthenia gravis, and thymoma. J Neurol Neurosurg Psychiatry 2005;76:141142.Google Scholar
Tanaka, H, Matsumura, A, Okumura, M, et al. Stiff man syndrome with thymoma. Ann Thorac Surg 2005;80:739741.Google Scholar
Iwata, T, Inoue, K, Mizuguchi, S, et al. Thymectomy for paraneoplastic stiff-person syndrome associated with invasive thymoma. J Thorac Cardiovasc Surg 2006;132:196197.Google Scholar
Kobayashi, R, Kaji, M, Horiuchi, S, et al. Recurrent thymoma with stiff-person syndrome and pure red blood cell aplasia. Ann Thorac Surg 2014;97:18021804.Google Scholar
Aghajanzadeh, M, Alavi, A, Aghajanzadeh, G, Massahania, S. Stiff man syndrome with invasive thymic carcinoma. Arch Iran Med 2013;16:195196.Google Scholar
Agarwal, PA, Ichaporia, NR. Glutamic acid decarboxylase antibody-positive paraneoplastic stiff limb syndrome associated with carcinoma of the breast. Neurol India 2010;58:449451.Google Scholar
Silverman, IE. Paraneoplastic stiff limb syndrome. J Neurol Neurosurg Psychiatry 1999;67:126127.Google Scholar
Sinnreich, M, Assal, F, Hefft, S, et al. Anti-GAD antibodies and breast cancer in a patient with stiff-person syndrome: a puzzling association. Eur Neurol 2001;46:5152.Google Scholar
Schiff, D, Dalmau, J, Myers, DJ. Anti-GAD antibody positive stiff-limb syndrome in multiple myeloma. J Neurooncol 2003;65:173175.Google Scholar
McHugh, JC, Murray, B, Renganathan, R, Connolly, S, Lynch, T. GAD antibody positive paraneoplastic stiff person syndrome in a patient with renal cell carcinoma. Mov Disord 2007;22:13431346.Google Scholar
Ferrari, P, Federico, M, Grimaldi, LM, Silingardi, V. Stiff-man syndrome in a patient with Hodgkin’s disease: an unusual paraneoplastic syndrome. Haematologica 1990;75:570572.Google Scholar
Rakocevic, G, Hussain, A. Stiff person syndrome improvement with chemotherapy in a patient with cutaneous T cell lymphoma. Muscle Nerve 2013;47:938939.Google Scholar
Koca, I, Ucar, M, Kalender, ME, Alkan, S. The horses are the first thought but one must not forget the zebras even if they are rare: stiff person syndrome associated with malignant mesothelioma. BMJ Case Rep 2014;2014:bcr2013203455.Google Scholar
Sarwari, N, Galili, Y, Perez, A, Avgeropoulos, N, Tseng, J. Unusual presentation of lung adenocarcinoma with paraneoplastic stiff person syndrome: role of EGFR tyrosine kinase inhibitors. J Thorac Oncol 2019;14:e179e180.Google Scholar
Yong, SYS, Teo, JY, Yong, KP, Goh, BKP. Paraneoplastic stiff person syndrome secondary to pancreatic adenocarcinoma. J Gastrointest Surg 2018;22:172174.Google Scholar
Levy, LM, Levy-Reis, I, Fujii, M, Dalakas, MC. Brain gamma-aminobutyric acid changes in stiff-person syndrome. Arch Neurol 2005;62:970974.Google Scholar
Alexopoulos, H, Dalakas, MC. Immunology of stiff person syndrome and other GAD-associated neurological disorders. Exp Rev Clin Immunol 2013;9:10431053.Google Scholar
Bu, DF, Erlander, MG, Hitz, BC, et al. Two human glutamate decarboxylases, 65-kDa GAD and 67-kDa GAD, are each encoded by a single gene. Proc Natl Acad Sci USA 1992;89:21152119.Google Scholar
Dinkel, K, Meinck, HM, Jury, KM, Karges, W, Richter, W. Inhibition of gamma-aminobutyric acid synthesis by glutamic acid decarboxylase autoantibodies in stiff-man syndrome. Ann Neurol 1998;44:194201.Google Scholar
Chang, T, Alexopoulos, H, McMenamin, M, et al. Neuronal surface and glutamic acid decarboxylase autoantibodies in nonparaneoplastic stiff person syndrome. JAMA Neurol 2013;70:11401149.Google Scholar
Chang, T, Alexopoulos, H, Pettingill, P, et al. Immunization against GAD induces antibody binding to GAD-independent antigens and brainstem GABAergic neuronal loss. PLoS One 2013;8:e72921.Google Scholar
Nemni, R, Caniatti, LM, Gironi, M, Bazzigaluppi, E, De Grandis, D. Stiff person syndrome does not always occur with maternal passive transfer of GAD65 antibodies. Neurology 2004;62:21012102.Google Scholar
Bjork, E, Velloso, LA, Kampe, O, Karlsson, FA. GAD autoantibodies in IDDM, stiff-man syndrome, and autoimmune polyendocrine syndrome type I recognize different epitopes. Diabetes 1994;43:161165.Google Scholar
Raju, R, Foote, J, Banga, JP, et al. Analysis of GAD65 autoantibodies in stiff-person syndrome patients. J Immunol 2005;175:77557762.Google Scholar
Fouka, P, Alexopoulos, H, Akrivou, S, et al. GAD65 epitope mapping and search for novel autoantibodies in GAD-associated neurological disorders. J Neuroimmunol 2015;281:7377.Google Scholar
Manto, MU, Laute, MA, Aguera, M, et al. Effects of anti-glutamic acid decarboxylase antibodies associated with neurological diseases. Ann Neurol 2007;61:544551.Google Scholar
Geis, C, Weishaupt, A, Grunewald, B, et al. Human stiff-person syndrome IgG induces anxious behavior in rats. PLoS One 2011;6:e16775.Google Scholar
Hansen, N, Grunewald, B, Weishaupt, A, et al. Human stiff person syndrome IgG-containing high-titer anti-GAD65 autoantibodies induce motor dysfunction in rats. Exp Neurol 2013;239:202209.Google Scholar
Manto, M, Honnorat, J, Hampe, CS, et al. Disease-specific monoclonal antibodies targeting glutamate decarboxylase impair GABAergic neurotransmission and affect motor learning and behavioral functions. Front Behav Neurosci 2015;9:78.Google Scholar
Sommer, C, Weishaupt, A, Brinkhoff, J, et al. Paraneoplastic stiff-person syndrome: passive transfer to rats by means of IgG antibodies to amphiphysin. Lancet 2005;365:14061411.Google Scholar
Geis, C, Weishaupt, A, Hallermann, S, et al. Stiff person syndrome-associated autoantibodies to amphiphysin mediate reduced GABAergic inhibition. Brain 2010;133:31663180.Google Scholar
Balint, B, Meinck, HM. Pragmatic treatment of stiff person spectrum disorders. Mov Disord Clin Pract 2018;5:394401.Google Scholar
Miller, F, Korsvik, H. Baclofen in the treatment of stiff-man syndrome. Ann Neurol 1981;9:511512.Google Scholar
Sechi, G, Barrocu, M, Piluzza, MG, et al. Levetiracetam in stiff-person syndrome. J Neurol 2008;255:17211725.Google Scholar
Squintani, G, Bovi, T, Ferigo, L, et al. Efficacy of pregabalin in a case of stiff-person syndrome: clinical and neurophysiological evidence. J Neurol Sci 2012;314:166168.Google Scholar
Meinck, HM, Conrad, B. Neuropharmacological investigations in the stiff-man syndrome. J Neurol 1986;233:340347.Google Scholar
Murinson, BB, Rizzo, M. Improvement of stiff-person syndrome with tiagabine. Neurology 2001;57:366.Google Scholar
Spehlmann, R, Norcross, K, Rasmus, SC, Schlageter, NL. Improvement of stiff-man syndrome with sodium valproate. Neurology 1981;31:11621163.Google Scholar
Seitz, RJ, Blank, B, Kiwit, JC, Benecke, R. Stiff-person syndrome with anti-glutamic acid decarboxylase autoantibodies: complete remission of symptoms after intrathecal baclofen administration. J Neurol 1995;242:618622.Google Scholar
Newton, JC, Harned, ME, Sloan, PA, Salles, SS. Trialing of intrathecal baclofen therapy for refractory stiff-person syndrome. Reg Anesth Pain Med 2013;38:248250.Google Scholar
Maramattom, BV. Intrathecal baclofen pump implantation in a case of stiff person syndrome. Neurol India 2010;58:115117.Google Scholar
Ho, BL, Shih, PY. Successful intrathecal baclofen therapy for seronegative stiff-person syndrome: a case report. Acta Neurologica Taiwanica 2008;17:172176.Google Scholar
Silbert, PL, Matsumoto, JY, McManis, PG, et al. Intrathecal baclofen therapy in stiff-man syndrome: a double-blind, placebo-controlled trial. Neurology 1995;45:18931897.Google Scholar
Stayer, C, Tronnier, V, Dressnandt, J, et al. Intrathecal baclofen therapy for stiff-man syndrome and progressive encephalomyelopathy with rigidity and myoclonus. Neurology 1997;49:15911597.Google Scholar
Abbatemarco, JR, Willis, MA, Wilson, RG, et al. Case series: intrathecal baclofen therapy in stiff-person syndrome. Neuromodulation 2018;21:655659.Google Scholar
Liguori, R, Cordivari, C, Lugaresi, E, Montagna, P. Botulinum toxin A improves muscle spasms and rigidity in stiff-person syndrome. Mov Disord 1997;12:10601063.Google Scholar
Davis, D, Jabbari, B. Significant improvement of stiff-person syndrome after paraspinal injection of botulinum toxin A. Mov Disord 1993;8:371373.Google Scholar
Anagnostou, E, Zambelis, T. Botulinum toxin A in anti-GAD-positive stiff-limb syndrome. Muscle Nerve 2012;46:457458.Google Scholar
Dalakas, MC, Fujii, M, Li, M, et al. High-dose intravenous immune globulin for stiff-person syndrome. N Engl J Med 2001;345:18701876.Google Scholar
Pagano, MB, Murinson, BB, Tobian, AA, King, KE. Efficacy of therapeutic plasma exchange for treatment of stiff-person syndrome. Transfusion 2014;54:18511856.Google Scholar
McKeon, A, Tracy, JA. GAD65 neurological autoimmunity. Muscle Nerve 2017;56:1527.Google Scholar
Baizabal-Carvallo, JF, Jankovic, J. Stiff-person syndrome: insights into a complex autoimmune disorder. J Neurol Neurosurg Psychiatry 2015;86:840848.Google Scholar
Fekete, R, Jankovic, J. Childhood stiff-person syndrome improved with rituximab. Case Rep Neurol 2012;4:9296.Google Scholar
Bacorro, EA, Tehrani, R. Stiff-person syndrome: persistent elevation of glutamic acid decarboxylase antibodies despite successful treatment with rituximab. J Clin Rheumatol 2010;16:237239.Google Scholar
Qureshi, A, Hennessy, M. Stiff person syndrome (SPS) complicated by respiratory failure: successful treatment with rituximab. J Neurol 2012;259:180181.Google Scholar
Lobo, ME, Araujo, ML, Tomaz, CA, Allam, N. Stiff-person syndrome treated with rituximab. BMJ Case Rep 2010;2010:bcr0520103021.Google Scholar
Baker, MR, Das, M, Isaacs, J, Fawcett, PR, Bates, D. Treatment of stiff person syndrome with rituximab. J Neurol Neurosurg Psychiatry 2005;76:9991001.Google Scholar
Dalakas, MC, Rakocevic, G, Dambrosia, JM, Alexopoulos, H, McElroy, B. A double-blind, placebo-controlled study of rituximab in patients with stiff person syndrome. Ann Neurol 2017;82:271277.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 2020;55:283306.Google Scholar
Sanders, S, Bredeson, C, Pringle, CE, et al. Autologous stem cell transplantation for stiff person syndrome: two cases from the Ottawa blood and marrow transplant program. JAMA Neurol 2014;71:12961299.Google Scholar
Kass-Iliyya, L, Snowden, JA, Thorpe, A, et al. Autologous haematopoietic stem cell transplantation for refractory stiff-person syndrome: the UK experience. J Neurol 2021;268:265275.Google Scholar
Burt, RK, Balabanov, R, Han, X, et al. Autologous hematopoietic stem cell transplantation for Stiff Person Spectrum Disorder: a clinical trial. Neurology 2020;96:e817e830.Google Scholar
Whiteley, AM, Swash, M, Urich, H. Progressive encephalomyelitis with rigidity. Brain 1976;99:2742.Google Scholar
Campbell, AM, Garland, H. Subacute myoclonic spinal neuronitis. J Neurol Neurosurg Psychiatry 1956;19:268274.Google Scholar
Kasperek, S, Zebrowski, S. Stiff-man syndrome and encephalomyelitis: report of a case. Arch Neurol 1971;24:2230.Google Scholar
Lhermitte, F, Chain, F, Escourolle, R, et al. [A further case of tetanus-like contracture distinct from the stiff man syndrome. Pharmacological and neuropathological study of a case of predominantly spinal encephalomyelitis]. Revue neurologique 1973;128:321.Google Scholar
Howell, DA, Lees, AJ, Toghill, PJ. Spinal internuncial neurones in progressive encephalomyelitis with rigidity. J Neurol Neurosurg Psychiatry 1979;42:773785.Google Scholar
Hutchinson, M, Waters, P, McHugh, J, et al. Progressive encephalomyelitis, rigidity, and myoclonus: a novel glycine receptor antibody. Neurology 2008;71:12911292.Google Scholar
Shugaiv, E, Leite, MI, Sehitoglu, E, et al. Progressive encephalomyelitis with rigidity and myoclonus: a syndrome with diverse clinical features and antibody responses. Eur Neurol 2013;69:257262.Google Scholar
Kyskan, R, Chapman, K, Mattman, A, Sin, D. Antiglycine receptor antibody and encephalomyelitis with rigidity and myoclonus (PERM) related to small cell lung cancer. BMJ Case Rep 2013;2013:bcr2013010027.Google Scholar
Spitz, M, Ferraz, HB, Barsottini, OG, Gabbai, AA. Progressive encephalomyelitis with rigidity: a paraneoplastic presentation of oat cell carcinoma of the lung: case report. Arq Neuropsiquiatr 2004;62:547549.Google Scholar
Mas, N, Saiz, A, Leite, MI, et al. Antiglycine-receptor encephalomyelitis with rigidity. J Neurol Neurosurg Psychiatry 2011;82:13991401.Google Scholar
Peeters, E, Vanacker, P, Woodhall, M, et al. Supranuclear gaze palsy in glycine receptor antibody-positive progressive encephalomyelitis with rigidity and myoclonus. Mov Disord 2012;27:18301832.Google Scholar
Bourke, D, Roxburgh, R, Vincent, A, et al. Hypoventilation in glycine-receptor antibody related progressive encephalomyelitis, rigidity and myoclonus. J Clin Neurosci 2014;21:876878.Google Scholar
Stern, WM, Howard, R, Chalmers, RM, et al. Glycine receptor antibody mediated Progressive Encephalomyelitis with Rigidity and Myoclonus (PERM): a rare but treatable neurological syndrome. Pract Neurol 2014;14:123127.Google Scholar
Piotrowicz, A, Thumen, A, Leite, MI, Vincent, A, Moser, A. A case of glycine-receptor antibody-associated encephalomyelitis with rigidity and myoclonus (PERM): clinical course, treatment and CSF findings. J Neurol 2011;258:22682270.Google Scholar
Jungehulsing, GJ, Behse, F, Grosse, P. Can progressive encephalomyelitis with rigidity mimic motor neuron disease? Case report and review of the literature. J Neurol 2005;252:863865.Google Scholar
Kenda, J, Svigelj, V, Rodi, Z, et al. Glycine receptor antibodies and progressive encephalomyelitis with rigidity and myoclonus with predominant motor neuron degeneration: expanding the clinical spectrum. J Neurol Sci 2015;353:177178.Google Scholar
Hinson, SR, Lopez-Chiriboga, AS, Bower, JH, et al. Glycine receptor modulating antibody predicting treatable stiff-person spectrum disorders. Neurol Neuroimmunol Neuroinflamm 2018;5:e438.Google Scholar
Crisp, SJ, Balint, B, Vincent, A. Redefining progressive encephalomyelitis with rigidity and myoclonus after the discovery of antibodies to glycine receptors. Curr Opin Neurol 2017;30:310316.Google Scholar
Piquet, AL, Khan, M, Warner, JEA, et al. Novel clinical features of glycine receptor antibody syndrome: a series of 17 cases. Neurol Neuroimmunol Neuroinflamm 2019;6:e592.Google Scholar
Casado, JL, Gil-Peralta, A, Graus, F, et al. Anti-Ri antibodies associated with opsoclonus and progressive encephalomyelitis with rigidity. Neurology 1994;44:15211522.Google Scholar
Ishii, A, Hayashi, A, Ohkoshi, N, Matsuno, S, Shoji, S. Progressive encephalomyelitis with rigidity associated with anti-amphiphysin antibodies. J Neurol Neurosurg Psychiatry 2004;75:661662.Google Scholar
Hara, M, Ariño, H, Petit-Pedrol, M, et al. DPPX-antibody associated encephalitis: main syndrome and antibody effects. Neurology 2017;88:13401348.Google Scholar
Rauschenberger, V, von Wardenburg, N, Schaefer, N, et al. Glycine receptor autoantibodies impair receptor function and induce motor dysfunction. Ann Neurol 2020;88:544561.Google Scholar
Crisp, SJ, Dixon, CL, Jacobson, L, et al. Glycine receptor autoantibodies disrupt inhibitory neurotransmission. Brain 2019;142:33983410.Google Scholar
Ozaki, K, Ohkubo, T, Yamada, T, et al. Progressive encephalomyelitis with rigidity and myoclonus resolving after thymectomy with subsequent anasarca: an autopsy case. Intern Med (Tokyo, Japan) 2018;57:34513458.Google Scholar
Turner, MR, Irani, SR, Leite, MI, et al. Progressive encephalomyelitis with rigidity and myoclonus: glycine and NMDA receptor antibodies. Neurology 2011;77:439443.Google Scholar
Swayne, A, Tjoa, L, Broadley, S, et al. Antiglycine receptor antibody related disease: a case series and literature review. Eur J Neurol 2018;25:12901298.Google Scholar
Boronat, A, Gelfand, JM, Gresa-Arribas, N, et al. Encephalitis and antibodies to dipeptidyl-peptidase-like protein-6, a subunit of Kv4.2 potassium channels. Ann Neurol 2013;73:120128.Google Scholar
Hara, M, Arino, H, Petit-Pedrol, M, et al. DPPX antibody-associated encephalitis: main syndrome and antibody effects. Neurology 2017;88:13401348.Google Scholar
Tobin, WO, Lennon, VA, Komorowski, L, et al. DPPX potassium channel antibody: frequency, clinical accompaniments, and outcomes in 20 patients. Neurology 2014;83:17971803.Google Scholar
Doherty, L, Gold, D, Solnes, L, Probasco, J, Venkatesan, A. Anti-DPPX encephalitis: prominent nystagmus reflected by extraocular muscle FDG-PET avidity. Neurol Neuroimmunol Neuroinflamm 2017;4:e361.Google Scholar
Stokin, GB, Popovic, M, Gelpi, E, et al. Neuropathologic features of anti-dipeptidyl-peptidase-like protein-6 antibody encephalitis. Neurology 2015;84:430432.Google Scholar
Dougherty, K, Covarrubias, M. A dipeptidyl aminopeptidase-like protein remodels gating charge dynamics in Kv4.2 channels. J Gen Physiol 2006;128:745753.Google Scholar
Ohkawa, T, Fukata, Y, Yamasaki, M, et al. Autoantibodies to epilepsy-related LGI1 in limbic encephalitis neutralize LGI1-ADAM22 interaction and reduce synaptic AMPA receptors. J Neurosci 2013;33:1816118174.Google Scholar
Petit-Pedrol, M, Sell, J, Planaguma, J, et al. LGI1 antibodies alter Kv1.1 and AMPA receptors changing synaptic excitability, plasticity and memory. Brain 2018;141:31443159.Google Scholar
Huijbers, MG, Querol, LA, Niks, EH, et al. The expanding field of IgG4-mediated neurological autoimmune disorders. Eur J Neurol 2015;22:11511161.Google Scholar

References

Sabater, L, Gaig, C, Gelpi, E, et al. A novel non-rapid-eye movement and rapid-eye-movement parasomnia with sleep breathing disorder associated with antibodies to IgLON5: a case series, characterisation of the antigen, and post-mortem study. Lancet Neurol 2014;13:575586.Google Scholar
Lugaresi, E, Provini, F. Agrypnia excitata: clinical features and pathophysiological implications. Sleep Med Rev 2001;5:313322.Google Scholar
Lugaresi, E, Provini, F, Cortelli, P. Agrypnia excitata. Sleep Med 2011;12(Suppl.2):S3S10.Google Scholar
Provini, F, Marconi, S, Amadori, M, et al. Morvan chorea and agrypnia excitata: when video-polysomnographic recording guides the diagnosis. Sleep Med 2011;12:10411043.Google Scholar
Gaig, C, Iranzo, A, Cajochen, C, et al. Characterization of the sleep disorder of anti-IgLON5 disease. Sleep 2019;42:zsz133.Google Scholar
Gaig, C, Graus, F, Compta, Y, et al. Clinical manifestations of the anti-IgLON5 disease. Neurology 2017;88:17361743.Google Scholar
Honorat, JA, Komorowski, L, Josephs, KA, et al. IgLON5 antibody: neurological accompaniments and outcomes in 20 patients. Neurol Neuroimmunol Neuroinflamm 2017;4:e385.Google Scholar
Escudero, D, Guasp, M, Arino, H, et al. Antibody-associated CNS syndromes without signs of inflammation in the elderly. Neurology 2017;89:14711475.Google Scholar
Cagnin, A, Mariotto, S, Fiorini, M, et al. Microglial and neuronal TDP-43 pathology in anti-IgLON5-related tauopathy. J Alzheimer Dis 2017;59:1320.Google Scholar
Erro, ME, Sabater, L, Martinez, L, et al. Anti-IGLON5 disease: a new case without neuropathologic evidence of brainstem tauopathy. Neurol Neuroimmunol Neuroinflamm 2020;7:e651.Google Scholar
Landa, J, Gaig, C, Planagumà, J, et al. Effects of IgLON5 antibodies on neuronal cytoskeleton: a link between autoimmunity and neurodegeneration. Ann Neurol 2020;88:10231027.Google Scholar
Haitao, R, Huiqin, L, Tao, Q, et al. Autoimmune encephalitis associated with vitiligo? J Neuroimmunol 2017;310:1416.Google Scholar
Ramanan, VK, Crum, BA, McKeon, A. Subacute encephalitis with recovery in IgLON5 autoimmunity. Neurol Neuroimmunol Neuroinflamm 2018;5:e485.Google Scholar
Werner, J, Jelcic, I, Schwarz, EI, et al. Anti-IgLON5 disease: A new bulbar-onset motor neuron mimic syndrome. Neurol Neuroimmunol Neuroinflamm 2021;8:e962.Google Scholar
Gaig, C, Iranzo, A, Santamaria, J, Graus, F. The sleep disorder in Anti-lgLON5 disease. Curr Neurol Neurosci Rep 2018;18:41.Google Scholar
Schoberl, F, Levin, J, Remi, J, et al. IgLON5: a case with predominant cerebellar tau deposits and leptomeningeal inflammation. Neurology 2018;91:180182.Google Scholar
Gaig, C, Compta, Y. Neurological profiles beyond the sleep disorder in patients with anti-IgLON5 disease. Curr Opin Neurol 2019;32:493499.Google Scholar
Schroder, JB, Melzer, N, Ruck, T, et al. Isolated dysphagia as initial sign of anti-IgLON5 syndrome. Neurol Neuroimmunol Neuroinflamm 2017;4:e302.Google Scholar
Bonello, M, Jacob, A, Ellul, MA, et al. IgLON5 disease responsive to immunotherapy. Neurol Neuroimmunol Neuroinflamm 2017;4:e383.Google Scholar
Nutt, JG, Marsden, CD, Thompson, PD. Human walking and higher-level gait disorders, particularly in the elderly. Neurology 1993;43:268279.Google Scholar
Demain, A, Westby, GW, Fernandez-Vidal, S, et al. High-level gait and balance disorders in the elderly: a midbrain disease? J Neurol 2014;261:196206.Google Scholar
Bruggemann, N, Wandinger, KP, Gaig, C, et al. Dystonia, lower limb stiffness, and upward gaze palsy in a patient with IgLON5 antibodies. Mov Disord 2016;31:762764.Google Scholar
Haitao, R, Yingmai, Y, Yan, H, et al. Chorea and parkinsonism associated with autoantibodies to IgLON5 and responsive to immunotherapy. J Neuroimmunol 2016;300:910.Google Scholar
Litvan, I, Agid, Y, Jankovic, J, et al. Accuracy of clinical criteria for the diagnosis of progressive supranuclear palsy (Steele–Richardson–Olszewski syndrome). Neurology 1996;46:922930.Google Scholar
Mangesius, S, Sprenger, F, Hoftberger, R, et al. IgLON5 autoimmunity tested negative in patients with progressive supranuclear palsy and corticobasal syndrome. Parkinsonism Relat Disord 2017;38:102103.Google Scholar
Gaig, C, Compta, Y, Heidbreder, A, et al. Frequency and characterization of movement disorders in anti-IgLON5 disease. Neurology 2021;97:e1367–e1381.Google Scholar
Simabukuro, MM, Sabater, L, Adoni, T, et al. Sleep disorder, chorea, and dementia associated with IgLON5 antibodies. Neurol Neuroimmunol Neuroinflamm 2015;2:e136.Google Scholar
Morales-Briceno, H, Cruse, B, Fois, AF, et al. IgLON5-mediated neurodegeneration is a differential diagnosis of CNS Whipple disease. Neurology 2018;90:11131115.Google Scholar
Vetter, E, Olmes, DG, Linker, R, Seifert, F. Teaching video NeuroImages: facial myokymia and myorhythmia in anti-IgLON5 disease – the bitten lip. Neurology 2018;91:e1659.Google Scholar
Peeters, I, Wiels, W, De Raedt, S, Flamez, A. Unusual head movements in Anti-IgLON5 disease. Mov Disord Clin Pract 2020;7:708709.Google Scholar
Baizabal-Carvallo, JF, Cardoso, F, Jankovic, J. Myorhythmia: phenomenology, etiology, and treatment. Mov Disord 2015;30:171179.Google Scholar
Montagna, M, Amir, R, De Volder, I, et al. IgLON5-associated encephalitis with atypical brain magnetic resonance imaging and cerebrospinal fluid changes. Front Neurol 2018;9:329.Google Scholar
Tao, QQ, Wei, Q, Song, SJ, Yin, XZ. Motor neuron disease-like phenotype associated with anti-IgLON5 disease. CNS Neurosci Therapeut 2018;24:13051308.Google Scholar
Muñoz-Lopetegi, A, Graus, F, Dalmau, J, Santamaria, J. Sleep disorders in autoimmune encephalitis. Lancet Neurol 2020;19:10101022.Google Scholar
Blinder, T, Lewerenz, J. Cerebrospinal fluid findings in patients with autoimmune encephalitis: a systematic analysis. Front Neurol 2019;10:804.Google Scholar
Moreno-Estebanez, A, Garcia-Ormaechea, M, Tijero, B, et al. Anti-IgLON5 disease responsive to immunotherapy: a case report with an abnormal MRI. Mov Disord Clin Pract 2018;5:653656.Google Scholar
Rossling, R, Pehl, D, Lingnau, M, Pruss, H. A case of CLIPPERS challenging the new diagnostic criteria. Brain 2018;141:e12.Google Scholar
Hasselbacher, K, Steffen, A, Wandinger, KP, Bruggemann, N. IgLON5 antibodies are infrequent in patients with isolated sleep apnea. Eur J Neurol 2018;25:e46e47.Google Scholar
Gaig, C, Ercilla, G, Daura, X, et al. HLA and microtubule-associated protein tau H1 haplotype associations in anti-IgLON5 disease. Neurol Neuroimmunol Neuroinflamm 2019;6:e605.Google Scholar
Iranzo, A, Santamaria, J. Severe obstructive sleep apnea/hypopnea mimicking REM sleep behavior disorder. Sleep 2005;28:203206.Google Scholar
Montagna, P, Lugaresi, E. Agrypnia excitata: a generalized overactivity syndrome and a useful concept in the neurophysiopathology of sleep. Clin Neurophysiol 2002;113:552560.Google Scholar
Mahowald, MW, Schenck, CH. Status dissociatus: a perspective on states of being. Sleep 1991;14:6979.Google Scholar
Fernandez-Arcos, A, Iranzo, A, Serradell, M, Gaig, C, Santamaria, J. The clinical phenotype of idiopathic rapid eye movement sleep behavior disorder at presentation: a study in 203 consecutive patients. Sleep 2016;39:121132.Google Scholar
Boxer, AL, Yu, JT, Golbe, LI, et al. Advances in progressive supranuclear palsy: new diagnostic criteria, biomarkers, and therapeutic approaches. Lancet Neurol 2017;16:552563.Google Scholar
Fanciulli, A, Wenning, GK. Multiple-system atrophy. N Engl J Med 2015;372:13751376.Google Scholar
Montojo, T, Piren, V, Benkhadra, F, et al. Gaze palsy, sleep and gait disorder, as well as Tako-Tsubo syndrome in a patient with IgLON5 antibodies. Mov Disord Clin Pract 2017;4:441443.Google Scholar
Brunetti, V, Della Marca, G, Spagni, G, Iorio, R. Immunotherapy improves sleep and cognitive impairment in anti-IgLON5 encephalopathy. Neurol Neuroimmunol Neuroinflamm 2019;6:e577.Google Scholar
Schwartz, MA, Selhorst, JB, Ochs, AL, et al. Oculomasticatory myorhythmia: a unique movement disorder occurring in Whipple’s disease. Ann Neurol 1986;20:677683.Google Scholar
Chung, HY, Wickel, J, Voss, A, et al. Autoimmune encephalitis with anti-IgLON5 and anti-GABAB-receptor antibodies: a case report. Medicine (Baltimore) 2019;98:e15706.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
Hongo, Y, Iizuka, T, Kaneko, A, et al. An autopsy case of MM2-thalamic subtype of sporadic Creutzfeldt–Jakob disease with Lewy bodies presenting as a sleep disorder mimicking anti-IgLON5 disease. J Neurol Sci 2019;404:3639.Google Scholar
Gelpi, E, Hoftberger, R, Graus, F, et al. Neuropathological criteria of anti-IgLON5-related tauopathy. Acta Neuropathol 2016;132:531543.Google Scholar
Grimwood, J, Gordon, LA, Olsen, A, et al. The DNA sequence and biology of human chromosome 19. Nature 2004;428:529535.Google Scholar
Karagogeos, D. Neural GPI-anchored cell adhesion molecules. Front Biosci 2003;8:s13041320.Google Scholar
Tan, RPA, Leshchyns’ka, I, Sytnyk, V. Glycosylphosphatidylinositol-anchored immunoglobulin superfamily cell adhesion molecules and their role in neuronal development and synapse regulation. Front Molec Neurosci 2017;10:378.Google Scholar
Hashimoto, T, Yamada, M, Maekawa, S, Nakashima, T, Miyata, S. IgLON cell adhesion molecule Kilon is a crucial modulator for synapse number in hippocampal neurons. Brain Res 2008;1224:111.Google Scholar
Ranaivoson, FM, Turk, LS, Ozgul, S, et al. A proteomic screen of neuronal cell-surface molecules reveals IgLONs as Structurally conserved interaction modules at the synapse. Structure 2019;27:893906.Google Scholar
Venkannagari, H, Kasper, JM, Misra, A, et al. Highly conserved molecular features in IgLONs contrast their distinct structural and biological outcomes. J Molec Biol 2020;432:52875303.Google Scholar
Vanaveski, T, Singh, K, Narvik, J, et al. Promoter-specific expression and genomic structure of IgLON family genes in mouse. Front Neurosci 2017;11:38.Google Scholar
Miyata, S, Matsumoto, N, Taguchi, K, et al. Biochemical and ultrastructural analyses of IgLON cell adhesion molecules, Kilon and OBCAM in the rat brain. Neuroscience 2003;117:645658.Google Scholar
Struyk, AF, Canoll, PD, Wolfgang, MJ, et al. Cloning of neurotrimin defines a new subfamily of differentially expressed neural cell adhesion molecules. J Neurosci 1995;15:21412156.Google Scholar
Zacco, A, Cooper, V, Chantler, PD, et al. Isolation, biochemical characterization and ultrastructural analysis of the limbic system-associated membrane protein (LAMP), a protein expressed by neurons comprising functional neural circuits. J Neurosci 1990;10:7390.Google Scholar
Sabater, L, Planaguma, J, Dalmau, J, Graus, F. Cellular investigations with human antibodies associated with the anti-IgLON5 syndrome. J Neuroinflammation 2016;13:226.Google Scholar
Huijbers, MG, Querol, LA, Niks, EH, et al. The expanding field of IgG4-mediated neurological autoimmune disorders. Eur J Neurol 2015;22:11511161.Google Scholar
Sollid, LM, Pos, W, Wucherpfennig, KW. Molecular mechanisms for contribution of MHC molecules to autoimmune diseases. Curr Opin Immunol 2014;31:2430.Google Scholar
Leshchyns’ka, I, Sytnyk, V. Reciprocal interactions between cell adhesion molecules of the immunoglobulin superfamily and the cytoskeleton in neurons. Front Cell Dev Biol 2016;4:9.Google Scholar
Dale, RC, Tantsis, EM, Merheb, V, et al. Antibodies to MOG have a demyelination phenotype and affect oligodendrocyte cytoskeleton. Neurol Neuroimmunol Neuroinflamm 2014;1:e12.Google Scholar
Ryding, M, Gamre, M, Nissen, MS, et al. Neurodegeneration induced by anti-IgLON5 antibodies studied in induced pluripotent stem cell-derived human neurons. Cells 2021;10:837.Google Scholar
Grüter, T, Behrendt, V, Bien, CI, Gold, R, Ayzenberg, I. Early immunotherapy is highly effective in IgG1/IgG4 positive IgLON5 disease. J Neurol 2020;267:21512153.Google Scholar
Logmin, K, Moldovan, AS, Elben, S, Schnitzler, A, Groiss, SJ. Intravenous immunoglobulins as first-line therapy for IgLON5 encephalopathy. J Neurol 2019;266:10311033.Google Scholar
Nissen, MS, Blaabjerg, M. Anti-IgLON5 disease: a case with 11-year clinical course and review of the literature. Front Neurol 2019;10:1056.Google Scholar
Cabezudo-García, P, Mena-Vázquez, N, Estivill Torrús, G, Serrano-Castro, P. Response to immunotherapy in anti-IgLON5 disease: a systematic review. Acta Neurol Scand 2020;141:263270.Google Scholar
Fuseya, K, Kimura, A, Yoshikura, N, et al. Corticobasal syndrome in a patient with anti-IgLON5 antibodies. Mov Disord Clin Pract 2020;7:557559.Google Scholar

References

Darnell, RB, Posner, JB. Paraneoplastic syndromes involving the nervous system. N Engl J Med 2003;349:15431554.Google Scholar
Rosenfeld, MR, Dalmau, J. Update on paraneoplastic and autoimmune disorders of the central nervous system. Semin Neurol 2010;30:320331.Google Scholar
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.Google Scholar
Soussain, C, Ricard, D, Fike, JR, et al. CNS complications of radiotherapy and chemotherapy. Lancet 2009;374:16391651.Google Scholar
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.Google Scholar
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.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
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.Google Scholar
Alamowitch, S, Graus, F, Uchuya, M, et al. Limbic encephalitis and small cell lung cancer: clinical and immunological features. Brain 1997;120:923928.Google Scholar
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.Google Scholar
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.Google Scholar
Armangue, T, Titulaer, MJ, Sabater, L, et al. A novel treatment-responsive encephalitis with frequent opsoclonus and teratoma. Ann Neurol 2014;75:435441.Google Scholar
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.Google Scholar
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.Google Scholar
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.Google 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.Google Scholar
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 Scholar
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.Google Scholar
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

References

Hauser, W, Fitzcharles, MA. Facts and myths pertaining to fibromyalgia. Dialogues Clin Neurosci 2018;20:5362.Google Scholar
Chong, JY, Rowland, LP, Utiger, RD. Hashimoto encephalopathy: syndrome or myth? Arch Neurol 2003;60:164171.Google Scholar
Chen, KA, Brilot, F, Dale, RC, Lafferty, AR, Andrews, PI. Hashimoto’s encephalopathy and anti-MOG antibody encephalitis: 50 years after Lord Brain’s description. Eur J Paediatr Neurol 2017;21:898901.Google Scholar
Mirabelli-Badenier, M, Biancheri, R, Morana, G, et al. Anti-NMDAR encephalitis misdiagnosed as Hashimoto’s encephalopathy. Eur J Paediatr Neurol 2014;18:7274.Google Scholar
Montagna, G, Imperiali, M, Agazzi, P, et al. Hashimoto’s encephalopathy: a rare proteiform disorder. Autoimmunity Rev 2016;15:466476.Google Scholar
Laurent, C, Capron, J, Quillerou, B, et al. Steroid-responsive encephalopathy associated with autoimmune thyroiditis (SREAT): characteristics, treatment and outcome in 251 cases from the literature. Autoimmunity Rev 2016;15:11291133.Google Scholar
Brain, L, Jellinek, EH, Ball, K. Hashimoto’s disease and encephalopathy. Lancet 1966;2:512514.Google Scholar
Thrush, DC, Boddie, HG. Episodic encephalopathy associated with thyroid disorders. J Neurol Neurosurg Psychiatry 1974;37:696700.Google Scholar
Becker, KL, Ferguson, RH, Mc, CW. The connective-tissue diseases and symptoms associated with Hashimoto’s thyroiditis. N Engl J Med 1963;268:277280.Google Scholar
Shein, M, Apter, A, Dickerman, Z, Tyano, S, Gadoth, N. Encephalopathy in compensated Hashimoto thyroiditis: a clinical expression of autoimmune cerebral vasculitis. Brain Dev 1986;8:6064.Google Scholar
Latinville, D, Bernardi, O, Cougoule, JP, et al. [Hashimoto’s thyroiditis and myoclonic encephalopathy. Pathogenic hypothesis]. Rev Neurol (Paris) 1985;141:5558.Google Scholar
Shaw, PJ, Walls, TJ, Newman, PK, Cleland, PG, Cartlidge, NE. Hashimoto’s encephalopathy: a steroid-responsive disorder associated with high anti-thyroid antibody titers – report of 5 cases. Neurology 1991;41:228233.Google Scholar
Kothbauer-Margreiter, I, Sturzenegger, M, Komor, J, Baumgartner, R, Hess, CW. Encephalopathy associated with Hashimoto thyroiditis: diagnosis and treatment. J Neurol 1996;243:585593.Google Scholar
v Maydell, B, Kopp, M, v Komorowski, G, et al. Hashimoto encephalopathy: is it underdiagnosed in pediatric patients? Neuropediatrics 2002;33:8689.Google Scholar
Ghika-Schmid, F, Ghika, J, Regli, F, et al. Hashimoto’s myoclonic encephalopathy: an underdiagnosed treatable condition? Mov Disord 1996;11:555562.Google Scholar
Sunil, GS, Mariash, CN. Hashimoto’s encephalitis. J Clin Endocrinol Metab 2001;86:947.Google Scholar
Peschen-Rosin, R, Schabet, M, Dichgans, J. Manifestation of Hashimoto’s encephalopathy years before onset of thyroid disease. Eur Neurol 1999;41:7984.Google Scholar
Selim, M, Drachman, DA. Ataxia associated with Hashimoto’s disease: progressive non-familial adult onset cerebellar degeneration with autoimmune thyroiditis. J Neurol Neurosurg Psychiatry 2001;71:8187.Google Scholar
Caselli, RJ, Boeve, BF, Scheithauer, BW, O’Duffy, JD, Hunder, GG. Nonvasculitic autoimmune inflammatory meningoencephalitis (NAIM): a reversible form of encephalopathy. Neurology 1999;53:15791581.Google Scholar
Chong, JY, Rowland, LP. What’s in a NAIM? Hashimoto encephalopathy, steroid-responsive encephalopathy associated with autoimmune thyroiditis, or nonvasculitic autoimmune meningoencephalitis? Arch Neurol 2006;63:175176.Google Scholar
Josephs, KA, Rubino, FA, Dickson, DW. Nonvasculitic autoimmune inflammatory meningoencephalitis. Neuropathology 2004;24:149152.Google Scholar
Schiess, N, Pardo, CA. Hashimoto’s encephalopathy. Ann N Y Acad Sci 2008;1142:254265.Google Scholar
Striano, P, Pagliuca, M, Andreone, V, et al. Unfavourable outcome of Hashimoto encephalopathy due to status epilepticus: one autopsy case. J Neurol 2006;253:248249.Google Scholar
Oide, T, Tokuda, T, Yazaki, M, et al. Anti-neuronal autoantibody in Hashimoto’s encephalopathy: neuropathological, immunohistochemical, and biochemical analysis of two patients. J Neurol Sci 2004;217:712.Google Scholar
Duffey, P, Yee, S, Reid, IN, Bridges, LR. Hashimoto’s encephalopathy: postmortem findings after fatal status epilepticus. Neurology 2003;61:11241126.Google Scholar
Nolte, KW, Unbehaun, A, Sieker, H, Kloss, TM, Paulus, W. Hashimoto encephalopathy: a brainstem vasculitis? Neurology 2000;54:769770.Google Scholar
Castillo, P, Woodruff, B, Caselli, R, et al. Steroid-responsive encephalopathy associated with autoimmune thyroiditis. Arch Neurol 2006;63:197202.Google Scholar
Ergul, AB, Altuner Torun, Y, Altug, U, et al. Congenital hemophilia A presenting with hashimoto’s encephalopathy and myocarditis: the first reported case. J Pediatr Hematol Oncol 2018;40:e435e438.Google Scholar
Cooper, BL, Appel, SE, Ammar, HM. A young female with urinary retention: Hashimoto’s encephalopathy. Am J Emerg Med 2017;35:943.e941943.e942.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
Litmeier, S, Pruss, H, Witsch, E, Witsch, J. Initial serum thyroid peroxidase antibodies and long-term outcomes in SREAT. Acta Neurol Scand 2016;134:452457.Google Scholar
Mattozzi, S, Sabater, L, Escudero, D, et al. Hashimoto encephalopathy in the 21st century. Neurology 2020;94:e217e224.Google Scholar
Hollowell, JG, Staehling, NW, Flanders, WD, et al. Serum TSH, T(4), and thyroid antibodies in the United States population (1988 to 1994): National Health and Nutrition Examination Survey (NHANES III). J Clin Endocrinol Metab 2002;87:489499.Google Scholar
Sinclair, D. Analytical aspects of thyroid antibodies estimation. Autoimmunity 2008;41:4654.Google Scholar
Ochi, H, Horiuchi, I, Araki, N, et al. Proteomic analysis of human brain identifies alpha-enolase as a novel autoantigen in Hashimoto’s encephalopathy. FEBS Lett 2002;528:197202.Google Scholar
Gitlits, VM, Toh, BH, Sentry, JW. Disease association, origin, and clinical relevance of autoantibodies to the glycolytic enzyme enolase. J Investig Med 2001;49:138145.Google Scholar
Vermeulen, N, Arijs, I, Joossens, S, et al. Anti-alpha-enolase antibodies in patients with inflammatory bowel disease. Clin Chem 2008;54:534541.Google Scholar
Pratesi, F, Moscato, S, Sabbatini, A, et al. Autoantibodies specific for alpha-enolase in systemic autoimmune disorders. J Rheumatol 2000;27:109115.Google Scholar
Adamus, G, Amundson, D, Seigel, GM, Machnicki, M. Anti-enolase-alpha autoantibodies in cancer-associated retinopathy: epitope mapping and cytotoxicity on retinal cells. J Autoimmun 1998;11:671677.Google Scholar
Yoneda, M, Fujii, A, Ito, A, et al. High prevalence of serum autoantibodies against the amino terminal of alpha-enolase in Hashimoto’s encephalopathy. J Neuroimmunol 2007;185:195200.Google Scholar
Fujii, A, Yoneda, M, Ito, T, et al. Autoantibodies against the amino terminal of alpha-enolase are a useful diagnostic marker of Hashimoto’s encephalopathy. J Neuroimmunol 2005;162:130136.Google Scholar
Kishitani, T, Matsunaga, A, Ikawa, M, et al. Limbic encephalitis associated with anti-NH2-terminal of alpha-enolase antibodies: a clinical subtype of Hashimoto encephalopathy. Medicine (Baltimore) 2017;96:e6181.Google Scholar
Hayashi, Y, Yamada, M, Kimura, A, et al. Clinical findings of a probable case of MM2-cortical-type sporadic Creutzfeldt-Jakob disease with antibodies to anti-N-terminus of alpha-enolase. Prion 2017;11:454464.Google Scholar
Ferracci, F, Bertiato, G, Moretto, G. Hashimoto’s encephalopathy: epidemiologic data and pathogenetic considerations. J Neurol Sci 2004;217:165168.Google Scholar
Dubey, D, Pittock, SJ, Kelly, CR, et al. Autoimmune encephalitis epidemiology and a comparison to infectious encephalitis. Ann Neurol 2018;83:166177.Google Scholar
Grande, ML, Constantino, A, Rayo, JI, et al. Brain hypoperfusion on Tc-99 m-ethylene dicysteine diethyl ester single-photon emission computed tomography in Hashimoto’s encephalopathy. Indian J Nucl Med 2013;28:102104.Google Scholar
Yeh, SJ, Tsai, LK, Tang, SC, Jeng, JS. Pin-point pupils with drowsiness as a presentation of Hashimoto’s encephalopathy mimicking pontine infarction. Acta neurologica Taiwanica 2014;23:1923.Google Scholar
Zimmermann, P, Stranzinger, E. Steroid-responsive encephalopathy associated with Hashimoto thyroiditis. Pediatr Radiol 2012;42:891893.Google Scholar
Hirose, D, Hirao, K, Kaneko, Y, et al. Case of Hashimoto’s encephalopathy showing atypical clinical course with magnetic resonance imaging abnormalities. Geriatr Gerontol Intl 2017;17:12351237.Google Scholar
Bonmann, E, Prumbaum, M, Stippich, C, Ringleb, P, Sliwka, U. Hashimoto encephalopathy: rare cause of stroke in young patient. Neurologist 2009;15:342344.Google Scholar
Yildiz, OK, Segmen, H, Oztoprak, I, Bolayir, E, Topaktas, S. Posterior reversible encephalopathy and alexia without agraphia in a patient with Hashimoto’s encephalopathy. Neurol Sci 2010;31:523525.Google Scholar
Lalanne, L, Meriot, ME, Ruppert, E, et al. Attempted infanticide and suicide inaugurating catatonia associated with Hashimoto’s encephalopathy: a case report. BMC Psychiatry 2016;16:13.Google Scholar
Liu, MY, Zhang, SQ, Hao, Y, Zheng, HM. Paroxysmal kinesigenic dyskinesia as the initial symptom of Hashimoto encephalopathy. CNS Neurosci Therapeut 2012;18:271273.Google Scholar
Georgiev, D, Kojovic, M, Klanjscek, G, Dolenc-Groselj, L. Hashimoto encephalopathy associated rapid onset narcolepsy type 1. Sleep Med 2017;29:9495.Google Scholar
Olmez, I, Moses, H, Sriram, S, et al. Diagnostic and therapeutic aspects of Hashimoto’s encephalopathy. J Neurol Sci 2013;331:6771.Google Scholar
Ramcharan, K, Hosein, N, Teelucksingh, JD, Rampersad, F, Teelucksingh, S. Prominent bilateral hand tremor in hashimoto’s encephalopathy: a video demonstration. Tremor Other Hyperkinet Mov 2016;6:419.Google Scholar
Schnedl, WJ, Mirzaei, S, Wallner-Liebmann, SJ, et al. Improvement of cerebral hypoperfusion with levothyroxine therapy in Hashimoto’s encephalopathy demonstrated by (99 m)Tc-HMPAO-SPECT. Eur Thyroid J 2013;2:116119.Google Scholar
Zhao, W, Li, J, Wang, J, et al. A case of Hashimoto encephalopathy: clinical manifestation, imaging, pathology, treatment, and prognosis. Neurologist 2011;17:141143.Google Scholar
Zhu, Y, Yang, H, Xiao, F. Hashimoto’s encephalopathy: a report of three cases and relevant literature reviews. Intl J Clin Exp Med 2015;8:1681716826.Google Scholar
He, L, Li, M, Long, XH, Li, XP, Peng, Y. A case of Hashimoto’s encephalopathy misdiagnosed as viral encephalitis. Am J Case Rep 2013;14:366369.Google Scholar
Uwatoko, H, Yabe, I, Sato, S, et al. Hashimoto’s encephalopathy mimicking a brain tumor and its pathological findings: a case report. J Neurol Sci 2018;394:141143.Google Scholar
Chang, T, Riffsy, MT, Gunaratne, PS. Hashimoto encephalopathy: clinical and MRI improvement following high-dose corticosteroid therapy. Neurologist 2010;16:394396.Google Scholar
Graham, BR, Shiff, N, Nour, M, et al. Hashimoto encephalopathy presenting with stroke-like episodes in an adolescent female: a case report and literature review. Pediatr Neurol 2016;59:6270.Google Scholar
Gresa-Arribas, N, Titulaer, MJ, Torrents, A, et al. Antibody titres at diagnosis and during follow-up of anti-NMDA receptor encephalitis: a retrospective study. Lancet Neurol 2014;13:167177.Google Scholar
Guasp, M, Modena, Y, Armangue, T, Dalmau, J, Graus, F. Clinical features of seronegative, but CSF antibody-positive, anti-NMDA receptor encephalitis. Neurol Neuroimmunol Neuroinflamm 2020;7:e659.Google Scholar
Gabilondo, I, Saiz, A, Galan, L, et al. Analysis of relapses in anti-NMDAR encephalitis. Neurology 2011;77:996999.Google Scholar
Kayser, MS, Titulaer, MJ, Gresa-Arribas, N, Dalmau, J. Frequency and characteristics of isolated psychiatric episodes in anti-N-methyl-d-aspartate receptor encephalitis. JAMA Neurol 2013;70:11331139.Google Scholar
Koros, C, Economou, A, Mastorakos, G, et al. A selective memory deficit caused by autoimmune encephalopathy associated with Hashimoto thyroiditis. Cogn Behav Neurol 2012;25:144148.Google Scholar
Laycock, K, Chaudhuri, A, Fuller, C, et al. A novel assessment and treatment approach to patients with Hashimoto’s encephalopathy. Endocrinol Diabetes Metab Case Rep 2018;2018:17-0117.Google Scholar
Sheetal, SK, Mathew, R, Peethambaran, B. Hashimoto’s encephalopathy as a treatable cause of corticobasal disease. Ann Indian Acad Neurol 2016;19:285286.Google Scholar
Gliebus, G, Lippa, CF. Cerebrospinal immunoglobulin level changes and clinical response to treatment of Hashimoto’s encephalopathy. Am J Alzheimer Dis Other Dementia 2009;24:373376.Google Scholar
de Holanda, NC, de Lima, DD, Cavalcanti, TB, Lucena, CS, Bandeira, F. Hashimoto’s encephalopathy: systematic review of the literature and an additional case. J Neuropsychiatr Clin Neurosci 2011;23:384390.Google Scholar
Aquino, RT, Mutarelli, EG. Hashimoto’s encephalopathy. Arq Neuropsiquiatr 2009;67:724725.Google Scholar
Mijajlovic, M, Mirkovic, M, Dackovic, J, Zidverc-Trajkovic, J, Sternic, N. Clinical manifestations, diagnostic criteria and therapy of Hashimoto’s encephalopathy: report of two cases. J Neurol Sci 2010;288:194196.Google Scholar
Chong, CS, Leung, JL, Wong, IH, Ng, PW, Miao, MY. Presenile dementia: a case of Hashimoto’s encephalopathy. E Asian Arch Psychiatr 2011;21:3236.Google Scholar
Guirgis, H, Amar, C. A Case of Hashimoto’s encephalopathy presenting with acute psychosis. J Neuropsychiatr Clin Neurosci 2014;26:E1E2.Google Scholar
Lee, Y, House, EM. Treatment of steroid-resistant Hashimoto encephalopathy with misidentification delusions and catatonia. Psychosomatics 2017;58:322327.Google Scholar
Segers, K, Braconnier, P, Corazza, F, et al. Subacute cognitive deterioration with high serum anti-thyroid peroxidase antibodies: two cases and a plea for pragmatism. Psychogeriatrics 2013;13:175179.Google Scholar
Vivek, AK, Arun, AB, Menon, V, Kandasamy, P. Acute polymorphic psychosis as a presenting feature of Hashimoto’s encephalopathy. Asian J Psychiatr 2016;19:1920.Google Scholar
Horikoshi, S, Miura, I, Kunii, Y, et al. Hashimoto encephalopathy with high plasma monoamine metabolite levels: a case report. Neuropsychiatr Dis Treat 2017;13:10431045.Google Scholar
Liu, CY, Tseng, MC, Lin, PH. Encephalopathy associated with autoimmune thyroid disease (Hashimoto’s thyroiditis) presenting as depression: a case report. Gen Hosp Psychiatry 2011;33:641.e647649.Google Scholar
Chang, JS, Chang, TC. Hashimoto’s encephalopathy: report of three cases. J Formosan Med Assoc 2014;113:862866.Google Scholar
Normann, C, Frase, L, Berger, M, Nissen, C. Steroid-responsive depression. BMJ Case Rep 2013;2013:bcr2013009101.Google Scholar
Endres, D, Perlov, E, Riering, AN, et al. Steroid-responsive chronic schizophreniform syndrome in the context of mildly increased antithyroid peroxidase antibodies. Front Psychiatry 2017;8:64.Google Scholar
Endres, D, Perlov, E, Stich, O, et al. Steroid responsive encephalopathy associated with autoimmune thyroiditis (SREAT) presenting as major depression. BMC Psychiatry 2016;16:184.Google Scholar
Prat, S, Jouan, Y, Magnant, J, Graux, J, El-Hage, W. Hashimoto encephalopathy diagnosis after 40 years of a schizophrenia-like disorder. Schizophr Res 2012;139:269270.Google Scholar
Lin, YT, Liao, SC. Hashimoto encephalopathy presenting as schizophrenia-like disorder. Cogn Behav Neurol 2009;22:197201.Google Scholar
Boban, M, Malojcic, B. Rumination syndrome in a patient with Hashimoto’s encephalopathy. J Neuropsychiatr Clin Neurosci 2011;23:E11E12.Google Scholar
Haider, AS, Alam, M, Adetutu, E, et al. Autoimmune schizophrenia? Psychiatric manifestations of Hashimoto’s encephalitis. Cureus 2016;8:e672.Google Scholar
Al-Busaidi, M, Burad, J, Al-Belushi, A, Gujjar, A. Super refractory status epilepticus in Hashimoto’s encephalopathy. Oman Med J 2017;32:247250.Google Scholar
Yu, HJ, Lee, J, Seo, DW, Lee, M. Clinical manifestations and treatment response of steroid in pediatric Hashimoto encephalopathy. J Child Neurol 2014;29:938942.Google Scholar
Visée, H, Mabiglia, C, Vanderaspoilden, V, Gazagnes, MD, Glibert, G. Recurrent status epilepticus associated with Hashimoto’s encephalopathy. Epilepsy Behav Case Rep 2013;1:113117.Google Scholar
Bektas, O, Yilmaz, A, Kendirli, T, Siklar, Z, Deda, G. Hashimoto encephalopathy causing drug-resistant status epilepticus treated with plasmapheresis. Pediatr Neurol 2012;46:132135.Google Scholar
Hilberath, JM, Schmidt, H, Wolf, GK. Steroid-responsive encephalopathy associated with autoimmune thyroiditis (SREAT): case report of reversible coma and status epilepticus in an adolescent patient and review of the literature. Eur J Pediatr 2014;173:12631273.Google Scholar
Monti, G, Pugnaghi, M, Ariatti, A, et al. Non-convulsive status epilepticus of frontal origin as the first manifestation of Hashimoto’s encephalopathy. Epileptic Disord 2011;13:253258.Google Scholar
Spatola, M, Petit-Pedrol, M, Simabukuro, MM, et al. Investigations in GABAA receptor antibody-associated encephalitis. Neurology 2017;88:10121020.Google Scholar
Maureille, A, Fenouil, T, Joubert, B, et al. Isolated seizures are a common early feature of paraneoplastic anti-GABAB receptor encephalitis. J Neurol 2019;266:195206.Google Scholar
Hacohen, Y, Joseph, S, Kneen, R, et al. Limbic encephalitis associated with elevated antithyroid antibodies. J Child Neurol 2014;29:769773.Google Scholar
Graus, F, Escudero, D, Oleaga, L, et al. Syndrome and outcome of antibody-negative limbic encephalitis. Eur J Neurol 2018;25:10111016.Google Scholar
Termsarasab, P, Pitakpatapee, Y, Frucht, SJ, Srivanitchapoom, P. Steroid-responsive encephalopathy associated with autoimmune thyroiditis (SREAT) presenting with pure cerebellar ataxia. Tremor Other Hyperkinet Mov 2018;8:585.Google Scholar
Matsunaga, A, Ikawa, M, Fujii, A, et al. Hashimoto’s encephalopathy as a treatable adult-onset cerebellar ataxia mimicking spinocerebellar degeneration. Eur Neurol 2013;69:1420.Google Scholar
Marignier, R, Chenevier, F, Rogemond, V, et al. Metabotropic glutamate receptor type 1 autoantibody-associated cerebellitis: a primary autoimmune disease? Arch Neurol 2010;67:627630.Google Scholar
Yaguchi, H, Yabe, I, Takahashi, H, et al. Identification of anti-Sez6l2 antibody in a patient with cerebellar ataxia and retinopathy. J Neurol 2014;261:224226.Google Scholar
Honorat, JA, Lopez-Chiriboga, AS, Kryzer, TJ, et al. Autoimmune septin-5 cerebellar ataxia. Neurol Neuroimmunol Neuroinflamm 2018;5:e474.Google Scholar
Rukmangadachar, LA, Dandapat, S, Bit-Ivan, EN, Peng, YY. Hashimoto’s encephalopathy: presenting with epilepsia partialis continua and a frontal lobe lesion. Clin Case Rep 2018;6:136142.Google Scholar
Spatola, M, Sabater, L, Planaguma, J, et al. Encephalitis with mGluR5 antibodies: symptoms and antibody effects. Neurology 2018;90:e1964e1972.Google Scholar
Gini, B, Lovato, L, Cianti, R, et al. Novel autoantigens recognized by CSF IgG from Hashimoto’s encephalitis revealed by a proteomic approach. J Neuroimmunol 2008;196:153158.Google Scholar

References

Moore, E, Huang, MW, Putterman, C. Advances in the diagnosis, pathogenesis and treatment of neuropsychiatric systemic lupus erythematosus. Curr Opin Rheumatol 2020;32:152158.Google Scholar
ACR. The American College of Rheumatology nomenclature and case definitions for neuropsychiatric lupus syndromes. Arthritis Rheum 1999;42:599608.Google Scholar
Bertsias, GK, Boumpas, DT. Pathogenesis, diagnosis and management of neuropsychiatric SLE manifestations. Nat Rev Rheumatol 2010;6:358367.Google Scholar
Ainiala, H, Hietaharju, A, Loukkola, J, et al. Validity of the new American College of Rheumatology criteria for neuropsychiatric lupus syndromes: a population-based evaluation. Arthritis Rheum 2001;45:419423.Google Scholar
Govoni, M, Bombardieri, S, Bortoluzzi, A, et al. Factors and comorbidities associated with first neuropsychiatric event in systemic lupus erythematosus: does a risk profile exist? A large multicentre retrospective cross-sectional study on 959 Italian patients. Rheumatology (Oxford, England) 2012;51:157168.Google Scholar
Mikdashi, J, Handwerger, B, Langenberg, P, Miller, M, Kittner, S. Baseline disease activity, hyperlipidemia, and hypertension are predictive factors for ischemic stroke and stroke severity in systemic lupus erythematosus. Stroke 2007;38:281285.Google Scholar
Hanly, JG, Walsh, NM, Sangalang, V. Brain pathology in systemic lupus erythematosus. J Rheumatol 1992;19:732741.Google Scholar
Mikdashi, JA, Esdaile, JM, Alarcon, GS, et al. Proposed response criteria for neurocognitive impairment in systemic lupus erythematosus clinical trials. Lupus 2007;16:418425.Google Scholar
Tomietto, P, Annese, V, D’Agostini, S, et al. General and specific factors associated with severity of cognitive impairment in systemic lupus erythematosus. Arthritis Rheum 2007;57:14611472.Google Scholar
Tay, SH, Mak, A. Diagnosing and attributing neuropsychiatric events to systemic lupus erythematosus: time to untie the Gordian knot? Rheumatology (Oxford, England) 2017;56:i14i23.Google Scholar
Kampylafka, EI, Alexopoulos, H, Kosmidis, ML, et al. Incidence and prevalence of major central nervous system involvement in systemic lupus erythematosus: a 3-year prospective study of 370 patients. PLoS One 2013;8:e55843.Google Scholar
Baizabal-Carvallo, JF, Delgadillo-Marquez, G, Estanol, B, Garcia-Ramos, G. Clinical characteristics and outcomes of the meningitides in systemic lupus erythematosus. Eur Neurol 2009;61:143148.Google Scholar
Kim, JM, Kim, KJ, Yoon, HS, et al. Meningitis in Korean patients with systemic lupus erythematosus: analysis of demographics, clinical features and outcomes; experience from affiliated hospitals of the Catholic University of Korea. Lupus 2011;20:531536.Google Scholar
Pego-Reigosa, JM, Isenberg, DA. Psychosis due to systemic lupus erythematosus: characteristics and long-term outcome of this rare manifestation of the disease. Rheumatology (Oxford, England) 2008;47:14981502.Google Scholar
Sanna, G, Bertolaccini, ML, Cuadrado, MJ, et al. Neuropsychiatric manifestations in systemic lupus erythematosus: prevalence and association with antiphospholipid antibodies. J Rheumatol 2003;30:985992.Google Scholar
Hanly, JG, Urowitz, MB, Siannis, F, et al. Autoantibodies and neuropsychiatric events at the time of systemic lupus erythematosus diagnosis: results from an international inception cohort study. Arthritis Rheum 2008;58:843853.Google Scholar
Gonzalez, A, Massardo, L. Antibodies and the brain: antiribosomal P protein antibody and the clinical effects in patients with systemic lupus erythematosus. Curr Opin Neurol 2018;31:300305.Google Scholar
Govoni, M, Bortoluzzi, A, Padovan, M, et al. The diagnosis and clinical management of the neuropsychiatric manifestations of lupus. J Autoimmun 2016;74:4172.Google Scholar
Eber, T, Chapman, J, Shoenfeld, Y. Anti-ribosomal P-protein and its role in psychiatric manifestations of systemic lupus erythematosus: myth or reality? Lupus 2005;14:571575.Google Scholar
Sciascia, S, Bertolaccini, ML, Roccatello, D, Khamashta, MA, Sanna, G. Autoantibodies involved in neuropsychiatric manifestations associated with systemic lupus erythematosus: a systematic review. J Neurol 2014;261:17061714.Google Scholar
Bortoluzzi, A, Scire, CA, Bombardieri, S, et al. Development and validation of a new algorithm for attribution of neuropsychiatric events in systemic lupus erythematosus. Rheumatology (Oxford, England) 2015;54:891898.Google Scholar
Karassa, FB, Afeltra, A, Ambrozic, A, et al. Accuracy of anti-ribosomal P protein antibody testing for the diagnosis of neuropsychiatric systemic lupus erythematosus: an international meta-analysis. Arthritis Rheum 2006;54:312324.Google Scholar
Choi, MY, FitzPatrick, RD, Buhler, K, Mahler, M, Fritzler, MJ. A review and meta-analysis of anti-ribosomal P autoantibodies in systemic lupus erythematosus. Autoimmunity Rev 2020;19:102463.Google Scholar
DeGiorgio, LA, Konstantinov, KN, Lee, SC, et al. A subset of lupus anti-DNA antibodies cross-reacts with the NR2 glutamate receptor in systemic lupus erythematosus. Nat Med 2001;7:11891193.Google Scholar
Hughes, EG, Peng, X, Gleichman, AJ, et al. Cellular and synaptic mechanisms of anti-NMDA receptor encephalitis. J Neurosci 2010;30:58665875.Google Scholar
Hirohata, S, Tanaka, K. Differential expression of antibodies to NMDA receptor in anti-NMDA receptor encephalitis and in neuropsychiatric systemic lupus erythematosus. Lupus Sci Med 2019;6:e000359.Google Scholar
Tay, SH, Fairhurst, AM, Mak, A. Clinical utility of circulating anti-N-methyl-d-aspartate receptor subunits NR2A/B antibody for the diagnosis of neuropsychiatric syndromes in systemic lupus erythematosus and Sjogren’s syndrome: an updated meta-analysis. Autoimmunity Rev 2017;16:114122.Google Scholar
Arinuma, Y, Yanagida, T, Hirohata, S. Association of cerebrospinal fluid anti-NR2 glutamate receptor antibodies with diffuse neuropsychiatric systemic lupus erythematosus. Arthritis Rheum 2008;58:11301135.Google Scholar
Matus, S, Burgos, PV, Bravo-Zehnder, M, et al. Antiribosomal-P autoantibodies from psychiatric lupus target a novel neuronal surface protein causing calcium influx and apoptosis. J Exp Med 2007;204:32213234.Google Scholar
Bravo-Zehnder, M, Toledo, EM, Segovia-Miranda, F, et al. Anti-ribosomal P protein autoantibodies from patients with neuropsychiatric lupus impair memory in mice. Arthritis Rheumatol (Hoboken, NJ) 2015;67:204214.Google Scholar
Katzav, A, Solodeev, I, Brodsky, O, et al. Induction of autoimmune depression in mice by anti-ribosomal P antibodies via the limbic system. Arthritis Rheum 2007;56:938948.Google Scholar
Katzav, A, Ben-Ziv, T, Chapman, J, et al. Anti-P ribosomal antibodies induce defect in smell capability in a model of CNS-SLE (depression). J Autoimmun 2008;31:393398.Google Scholar
Gaburo, N Jr, de Carvalho, JF, Timo-Iaria, CIM, et al. Electrophysiological dysfunction induced by anti-ribosomal P protein antibodies injection into the lateral ventricle of the rat brain. Lupus 2017;26:463469.Google Scholar
Kowal, C, Degiorgio, LA, Lee, JY, et al. Human lupus autoantibodies against NMDA receptors mediate cognitive impairment. Proc Natl Acad Sci USA 2006;103:1985419859.Google Scholar
Planaguma, J, Leypoldt, F, Mannara, F, et al. Human N-methyl D-aspartate receptor antibodies alter memory and behaviour in mice. Brain 2015;138:94109.Google Scholar
Chang, EH, Volpe, BT, Mackay, M, et al. Selective impairment of spatial cognition caused by autoantibodies to the N-methyl-D-aspartate receptor. EBioMedicine 2015;2:755764.Google Scholar
Huerta, PT, Kowal, C, DeGiorgio, LA, Volpe, BT, Diamond, B. Immunity and behavior: antibodies alter emotion. Proc Natl Acad Sci USA 2006;103:678683.Google Scholar
Lee, JY, Huerta, PT, Zhang, J, et al. Neurotoxic autoantibodies mediate congenital cortical impairment of offspring in maternal lupus. Nat Med 2009;15:9196.Google Scholar
Faust, TW, Chang, EH, Kowal, C, et al. Neurotoxic lupus autoantibodies alter brain function through two distinct mechanisms. Proc Natl Acad Sci USA 2010;107:1856918574.Google Scholar
Kowal, C, DeGiorgio, LA, Nakaoka, T, et al. Cognition and immunity; antibody impairs memory. Immunity 2004;21:179188.Google Scholar
Chan, K, Nestor, J, Huerta, TS, et al. Lupus autoantibodies act as positive allosteric modulators at GluN2A-containing NMDA receptors and impair spatial memory. Nat Commun 2020;11:1403.Google Scholar
Diamond, B, Huerta, PT, Mina-Osorio, P, Kowal, C, Volpe, BT. Losing your nerves? Maybe it’s the antibodies. Nat Rev Immunol 2009;9:449456.Google Scholar
Varley, JA, Andersson, M, Grant, E, et al. Absence of neuronal autoantibodies in neuropsychiatric systemic lupus erythematosus. Ann Neurol 2020;88:12441250.Google Scholar
Borhani-Haghighi, A, Kardeh, B, Banerjee, S, et al. Neuro-Behcet’s disease: an update on diagnosis, differential diagnoses, and treatment. Mult Scler Relat Disord 2019;39:101906.Google Scholar
International Study Group for Behcet’s Disease. Criteria for diagnosis of Behcet’s disease. Lancet 1990;335:10781080.Google Scholar
Al-Araji, A, Kidd, DP. Neuro-Behcet’s disease: epidemiology, clinical characteristics, and management. Lancet Neurol 2009;8:192204.Google Scholar
Akman-Demir, G, Serdaroglu, P, Tasci, B. Clinical patterns of neurological involvement in Behcet’s disease: evaluation of 200 patients. The Neuro-Behcet Study Group. Brain 1999;122:21712182.Google Scholar
Bolek, EC, Sari, A, Kilic, L, et al. Clinical features and disease course of neurological involvement in Behcet’s disease: HUVAC experience. Mult Scler Relat Disord 2019;38:101512.Google Scholar
Kidd, D, Steuer, A, Denman, AM, Rudge, P. Neurological complications in Behcet’s syndrome. Brain 1999;122:21832194.Google Scholar
Albayram, S, Saip, S, Hasiloglu, ZI, et al. Evaluation of parenchymal neuro-Behcet disease by using susceptibility-weighted imaging. Am J Neuroradiol 2011;32:10501055.Google Scholar
Akman-Demir, G, Bahar, S, Coban, O, Tasci, B, Serdaroglu, P. Cranial MRI in Behcet’s disease: 134 examinations of 98 patients. Neuroradiology 2003;45:851859.Google Scholar
Law, LY, Riminton, DS, Nguyen, M, et al. The spectrum of immune-mediated and inflammatory lesions of the brainstem: clues to diagnosis. Neurology 2019;93:390405.Google Scholar
Coban, O, Bahar, S, Akman-Demir, G, et al. Masked assessment of MRI findings: is it possible to differentiate neuro-Behcet’s disease from other central nervous system diseases? [corrected]. Neuroradiology 1999;41:255260.Google Scholar
Kalra, S, Silman, A, Akman-Demir, G, et al. Diagnosis and management of neuro-Behcet’s disease: international consensus recommendations. J Neurol 2014;261:16621676.Google Scholar
International Team for the Revision of the International Criteria for Behçet’s Disease (ITR-ICBD). The International Criteria for Behcet’s Disease (ICBD): a collaborative study of 27 countries on the sensitivity and specificity of the new criteria. J Eur Acad Dermatol Venereol 2014;28:338347.Google Scholar
Tobin, WO, Guo, Y, Krecke, KN, et al. Diagnostic criteria for chronic lymphocytic inflammation with pontine perivascular enhancement responsive to steroids (CLIPPERS). Brain 2017;140:24152425.Google Scholar
Taieb, G, Mulero, P, Psimaras, D, et al. CLIPPERS and its mimics: evaluation of new criteria for the diagnosis of CLIPPERS. J Neurol Neurosurg Psychiatry 2019;90:10271038.Google Scholar
Lee, HS, Kim do, Y, Shin, HY, Choi, YC, Kim, SM. Spinal cord involvement in Behcet’s disease. Mult Scler 2016;22:960963.Google Scholar
Uygunoglu, U, Zeydan, B, Ozguler, Y, et al. Myelopathy in Behcet’s disease: the bagel sign. Ann Neurol 2017;82:288298.Google Scholar
Bennett, DL, McCabe, DJ, Stevens, JM, et al. Tumefactive neuro-Behcet disease. Neurology 2004;63:709.Google Scholar
Tramontini, PL, Finkelsztejn, A, Duarte, JA, et al. Neuro-Behcet disease mimicking brain tumor: a case report. Surg Neurol Intl 2017;8:97.Google Scholar
Jade, J, Chung, K, Arendse, M, Hussain, Z, White, D. Neuro-Behcet’s disease presenting with tumour-like lesions and responding to rituximab. J Clin Neurosci 2016;32:139141.Google Scholar
Siva, A, Kantarci, OH, Saip, S, et al. Behcet’s disease: diagnostic and prognostic aspects of neurological involvement. J Neurol 2001;248:95103.Google Scholar
Zeydan, B, Uygunoglu, U, Saip, S, et al. Infliximab is a plausible alternative for neurologic complications of Behcet disease. Neurol Neuroimmunol Neuroinflamm 2016;3:e258.Google Scholar
Hatemi, G, Christensen, R, Bang, D, et al. 2018 update of the EULAR recommendations for the management of Behcet’s syndrome. Ann Rheumat Dis 2018;77:808818.Google Scholar
Akman-Demir, G, Tüzün, E, Içöz, S, et al. Interleukin-6 in neuro-Behçet’s disease: association with disease subsets and long-term outcome. Cytokine 2008;44:373376.Google Scholar
Akiyama, M, Kaneko, Y, Takeuchi, T. Effectiveness of tocilizumab in Behcet’s disease: a systematic literature review. Semin Arthritis Rheumat 2020;50:797804.Google Scholar
Iannuzzi, MC, Rybicki, BA, Teirstein, AS. Sarcoidosis. N Engl J Med 2007;357:21532165.Google Scholar
Fritz, D, van de Beek, D, Brouwer, MC. Clinical features, treatment and outcome in neurosarcoidosis: systematic review and meta-analysis. BMC Neurol 2016;16:220.Google Scholar
Arkema, EV, Cozier, YC. Epidemiology of sarcoidosis: current findings and future directions. Therapeut Adv Chronic Dis 2018;9:227240.Google Scholar
Dumas, O, Abramovitz, L, Wiley, AS, Cozier, YC, Camargo, CA Jr. Epidemiology of sarcoidosis in a prospective cohort study of U.S. women. Ann Am Thorac Soc 2016;13:6771.Google Scholar
Chen, ES, Moller, DR. Sarcoidosis–scientific progress and clinical challenges. Nat Rev Rheumatol 2011;7:457467.Google Scholar
Muller-Quernheim, J, Prasse, A, Zissel, G. Pathogenesis of sarcoidosis. Presse Med 2012;41:e275287.Google Scholar
Rosen, Y. Pathology of sarcoidosis. Semin Respirat Crit Care Med 2007;28:3652.Google Scholar
Paparel, P, Devonec, M, Perrin, P, et al. Association between sarcoidosis and testicular carcinoma: a diagnostic pitfall. Sarcoidosis Vasc Diffuse Lung Dis 2007;24:95101.Google Scholar
Massey, J, Walker, S, Galloway, M, et al. Granulomatous CNS inflammation associated with seminoma. J Neurol 2019;266:13891393.Google Scholar
Kim, SM, Kim, SJ, Lee, HJ, et al. Differential diagnosis of neuromyelitis optica spectrum disorders. Therapeut Adv Neurol Disord 2017;10:265289.Google Scholar
Murphy, OC, Salazar-Camelo, A, Jimenez, JA, et al. Clinical and MRI phenotypes of sarcoidosis-associated myelopathy. Neurol Neuroimmunol Neuroinflamm 2020;7:e722.Google Scholar
Zalewski, NL, Krecke, KN, Weinshenker, BG, et al. Central canal enhancement and the trident sign in spinal cord sarcoidosis. Neurology 2016;87:743744.Google Scholar
Flanagan, EP, Kaufmann, TJ, Krecke, KN, et al. Discriminating long myelitis of neuromyelitis optica from sarcoidosis. Ann Neurol 2016;79:437447.Google Scholar
Kidd, DP. Sarcoidosis of the central nervous system: clinical features, imaging, and CSF results. J Neurol 2018;265:19061915.Google Scholar
Flanagan, EP, Hinson, SR, Lennon, VA, et al. Glial fibrillary acidic protein immunoglobulin G as biomarker of autoimmune astrocytopathy: analysis of 102 patients. Ann Neurol 2017;81:298309.Google Scholar
Shah, R, Roberson, GH, Cure, JK. Correlation of MR imaging findings and clinical manifestations in neurosarcoidosis. Am J Neuroradiol 2009;30:953961.Google Scholar
Smith, JK, Matheus, MG, Castillo, M. Imaging manifestations of neurosarcoidosis. Am J Roentgenol 2004;182:289295.Google Scholar
Fritz, D, van de Beek, D, Brouwer, MC, Booij, J. Whole-body 18F-FDG PET-CT in the diagnosis of neurosarcoidosis. Mayo Clin Proc 2020;95:10821084.Google Scholar
Stern, BJ, Royal, W III, Gelfand, JM, et al. Definition and consensus diagnostic criteria for neurosarcoidosis: From the Neurosarcoidosis Consortium Consensus Group. JAMA Neurol 2018;75:15461553.Google Scholar
Zajicek, JP, Scolding, NJ, Foster, O, et al. Central nervous system sarcoidosis: diagnosis and management. Month J Assoc Physician 1999;92:103117.Google Scholar
Joubert, B, Chapelon-Abric, C, Biard, L, et al. Association of prognostic factors and immunosuppressive treatment with long-term outcomes in neurosarcoidosis. JAMA Neurol 2017;74:13361344.Google Scholar
Cohen Aubart, F, Bouvry, D, Galanaud, D, et al. Long-term outcomes of refractory neurosarcoidosis treated with infliximab. J Neurol 2017;264:891897.Google Scholar
Gelfand, JM, Bradshaw, MJ, Stern, BJ, et al. Infliximab for the treatment of CNS sarcoidosis: a multi-institutional series. Neurology 2017;89:20922100.Google Scholar
Fritz, D, Timmermans, WMC, van Laar, JAM, et al. Infliximab treatment in pathology-confirmed neurosarcoidosis. Neurol Neuroimmunol Neuroinflamm 2020;7:e847.Google Scholar
Jamilloux, Y, Cohen-Aubart, F, Chapelon-Abric, C, et al. Efficacy and safety of tumor necrosis factor antagonists in refractory sarcoidosis: a multicenter study of 132 patients. Semin Arthritis Rheumat 2017;47:288294.Google Scholar
Stone, JH, Zen, Y, Deshpande, V. IgG4-related disease. N Engl J Med 2012;366:539551.Google Scholar
Stone, JH, Khosroshahi, A, Deshpande, V, et al. Recommendations for the nomenclature of IgG4-related disease and its individual organ system manifestations. Arthritis Rheum 2012;64:30613067.Google Scholar
Lu, LX, Della-Torre, E, Stone, JH, Clark, SW. IgG4-related hypertrophic pachymeningitis: clinical features, diagnostic criteria, and treatment. JAMA Neurol 2014;71:785793.Google Scholar
Regev, K, Nussbaum, T, Cagnano, E, Giladi, N, Karni, A. Central nervous system manifestation of IgG4-related disease. JAMA Neurol 2014;71:767770.Google Scholar
Melenotte, C, Seguier, J, Ebbo, M, et al. Clinical presentation, treatment and outcome of IgG4-related pachymeningitis: from a national case registry and literature review. Semin Arthritis Rheumat 2019;49:430437.Google Scholar
AbdelRazek, MA, Venna, N, Stone, JH. IgG4-related disease of the central and peripheral nervous systems. Lancet Neurol 2018;17:183192.Google Scholar
Kupersmith, MJ, Martin, V, Heller, G, Shah, A, Mitnick, HJ. Idiopathic hypertrophic pachymeningitis. Neurology 2004;62:686694.Google Scholar
Amrhein, TJ, Kranz, PG. Spontaneous intracranial hypotension: imaging in diagnosis and treatment. Radiol Clin North Am 2019;57:439451.Google Scholar
Della-Torre, E, Galli, L, Franciotta, D, et al. Diagnostic value of IgG4 Indices in IgG4-related hypertrophic pachymeningitis. J Neuroimmunol 2014;266:8286.Google Scholar
Wallace, ZS, Carruthers, MN, Khosroshahi, A, et al. IgG4-related disease and hypertrophic pachymeningitis. Medicine (Baltimore) 2013;92:206216.Google Scholar
Deshpande, V, Zen, Y, Chan, JK, et al. Consensus statement on the pathology of IgG4-related disease. Mod Pathol 2012;25:11811192.Google Scholar
Khosroshahi, A, Wallace, ZS, Crowe, JL, et al. International consensus guidance statement on the management and treatment of IgG4-related disease. Arthritis Rheumatol (Hoboken, NJ) 2015;67:16881699.Google Scholar
Campochiaro, C, Della-Torre, E, Lanzillotta, M, et al. Long-term efficacy of maintenance therapy with rituximab for IgG4-related disease. Eur J Intern Med 2020;74:9298.Google Scholar
Ebbo, M, Grados, A, Samson, M, et al. Long-term efficacy and safety of rituximab in IgG4-related disease: data from a French nationwide study of thirty-three patients. PLoS One 2017;12:e0183844.Google Scholar
Levraut, M, Cohen, M, Bresch, S, et al. Immunoglobulin G4-related hypertrophic pachymeningitis: a case-oriented review. Neurol Neuroimmunol Neuroinflamm 2019;6:e568.Google Scholar
Rice, CM, Scolding, NJ. The diagnosis of primary central nervous system vasculitis. Pract Neurol 2020;20:109114.Google Scholar
Cravioto, H, Feigin, I. Noninfectious granulomatous angiitis with a predilection for the nervous system. Neurology 1959;9:599609.Google Scholar
Calabrese, LH, Mallek, JA. Primary angiitis of the central nervous system: report of 8 new cases, review of the literature, and proposal for diagnostic criteria. Medicine (Baltimore) 1988;67:2039.Google Scholar
Benseler, SM. Central nervous system vasculitis in children. Curr Rheumatol Rep 2006;8:442449.Google Scholar
Salvarani, C, Brown, RD Jr, Calamia, KT, et al. Primary central nervous system vasculitis: analysis of 101 patients. Ann Neurol 2007;62:442451.Google Scholar
Salvarani, C, Brown, RD Jr, Christianson, T, et al. An update of the Mayo Clinic cohort of patients with adult primary central nervous system vasculitis: description of 163 patients. Medicine (Baltimore) 2015;94:e738.Google Scholar
Gallagher, KT, Shaham, B, Reiff, A, et al. Primary angiitis of the central nervous system in children: 5 cases. J Rheumatol 2001;28:616623.Google Scholar
Benseler, SM, Silverman, E, Aviv, RI, et al. Primary central nervous system vasculitis in children. Arthritis Rheum 2006;54:12911297.Google Scholar
Salvarani, C, Brown, RD Jr., Calamia, KT, et al. Primary central nervous system vasculitis presenting with intracranial hemorrhage. Arthritis Rheum 2011;63:35983606.Google Scholar
Salvarani, C, Brown, RD Jr., Hunder, GG. Adult primary central nervous system vasculitis. Lancet 2012;380:767777.Google Scholar
Salvarani, C, Brown, RD Jr., Christianson, TJH, et al. Primary central nervous system vasculitis mimicking brain tumor: comprehensive analysis of 13 cases from a single institutional cohort of 191 cases. J Autoimmun 2019;97:2228.Google Scholar
Zhu, DS, Yang, XL, Lv, HH, et al. Seizure syndrome as a first manifestation of solitary tumor-like mass lesion of PACNS: two case reports. Medicine (Baltimore) 2017;96:e6018.Google Scholar
de Boysson, H, Boulouis, G, Dequatre, N, et al. Tumor-like presentation of primary angiitis of the central nervous system. Stroke 2016;47:24012404.Google Scholar
Killeen, T, Jucker, D, Went, P, et al. Solitary tumour-like mass lesions of the central nervous system: primary angiitis of the CNS and inflammatory pseudotumour. Clin Neurol Neurosurg 2015;135:3437.Google Scholar
Salvarani, C, Brown, RD Jr, Calamia, KT, et al. Primary CNS vasculitis with spinal cord involvement. Neurology 2008;70:23942400.Google Scholar
Twilt, M, Benseler, SM. CNS vasculitis in children. Mult Scler Relat Disord 2013;2:162171.Google Scholar
Ganta, K, Malik, AM, Wood, JB, Levin, MC. Radial contrast enhancement on brain magnetic resonance imaging diagnostic of primary angiitis of the central nervous system: a case report and review of the literature. J Med Care Rep 2014;8:26.Google Scholar
Hajj-Ali, RA, Singhal, AB, Benseler, S, Molloy, E, Calabrese, LH. Primary angiitis of the CNS. Lancet Neurol 2011;10:561572.Google Scholar
Boulouis, G, de Boysson, H, Zuber, M, et al. Primary angiitis of the central nervous system: magnetic resonance imaging spectrum of parenchymal, meningeal, and vascular lesions at baseline. Stroke 2017;48:12481255.Google Scholar
Singhal, AB, Topcuoglu, MA, Fok, JW, et al. Reversible cerebral vasoconstriction syndromes and primary angiitis of the central nervous system: clinical, imaging, and angiographic comparison. Ann Neurol 2016;79:882894.Google Scholar
Caputi, L, Erbetta, A, Marucci, G, et al. Biopsy-proven primary angiitis of the central nervous system mimicking leukodystrophy: a case report and review of the literature. J Clin Neurosci 2019;64:4244.Google Scholar
de Boysson, H, Boulouis, G, Aouba, A, et al. Adult primary angiitis of the central nervous system: isolated small-vessel vasculitis represents distinct disease pattern. Rheumatology (Oxford, England) 2017;56:439444.Google Scholar
Schuster, S, Bachmann, H, Thom, V, et al. Subtypes of primary angiitis of the CNS identified by MRI patterns reflect the size of affected vessels. J Neurol Neurosurg Psychiatry 2017;88:749755.Google Scholar
Mossa-Basha, M, Hwang, WD, De Havenon, A, et al. Multicontrast high-resolution vessel wall magnetic resonance imaging and its value in differentiating intracranial vasculopathic processes. Stroke 2015;46:15671573.Google Scholar
Kuker, W, Gaertner, S, Nagele, T, et al. Vessel wall contrast enhancement: a diagnostic sign of cerebral vasculitis. Cerebrovasc Dis (Basel, Switzerland) 2008;26:2329.Google Scholar
Twilt, M, Benseler, SM. The spectrum of CNS vasculitis in children and adults. Nat Rev Rheumatol 2011;8:97107.Google Scholar
Aviv, RI, Benseler, SM, DeVeber, G, et al. Angiography of primary central nervous system angiitis of childhood: conventional angiography versus magnetic resonance angiography at presentation. Am J Neuroradiol 2007;28:915.Google Scholar
Duna, GF, Calabrese, LH. Limitations of invasive modalities in the diagnosis of primary angiitis of the central nervous system. J Rheumatol 1995;22:662667.Google Scholar
Salvarani, C, Brown, RD Jr, Calamia, KT, et al. Primary central nervous system vasculitis: analysis of 101 patients. Ann Neurol 2007;62:442451.Google Scholar
Harris, KG, Tran, DD, Sickels, WJ, Cornell, SH, Yuh, WT. Diagnosing intracranial vasculitis: the roles of MR and angiography. Am J Neuroradiol 1994;15:317330.Google Scholar
Raghavan, A, Wright, JM, Huang Wright, C, et al. Concordance of angiography and cerebral biopsy results for suspected primary central nervous system vasculitis: a multi-center retrospective review. Clin Neurol Neurosurg 2019;185:105482.Google Scholar
de Boysson, H, Parienti, JJ, Mawet, J, et al. Primary angiitis of the CNS and reversible cerebral vasoconstriction syndrome: a comparative study. Neurology 2018;91:e1468e1478.Google Scholar
Miller, DV, Salvarani, C, Hunder, GG, et al. Biopsy findings in primary angiitis of the central nervous system. Am J Surg Pathol 2009;33:3543.Google Scholar
Hajj-Ali, RA, Saygin, D, Ray, E, et al. Long-term outcomes of patients with primary angiitis of the central nervous system. Clin Exp Rheumatol 2019;37(Suppl. 117):4551.Google Scholar
de Boysson, H, Zuber, M, Naggara, O, et al. Primary angiitis of the central nervous system: description of the first fifty-two adults enrolled in the French cohort of patients with primary vasculitis of the central nervous system. Arthritis Rheumatol (Hoboken, NJ) 2014;66:13151326.Google Scholar
Beelen, J, Benseler, SM, Dropol, A, Ghali, B, Twilt, M. Strategies for treatment of childhood primary angiitis of the central nervous system. Neurol Neuroimmunol Neuroinflamm 2019;6:e567.Google Scholar
Beuker, C, Schmidt, A, Strunk, D, et al. Primary angiitis of the central nervous system: diagnosis and treatment. Therapeut Adv Neurol Disord 2018;11:1756286418785071.Google Scholar
Hutchinson, C, Elbers, J, Halliday, W, et al. Treatment of small vessel primary CNS vasculitis in children: an open-label cohort study. Lancet Neurol 2010;9:10781084.Google Scholar
Salvarani, C, Brown, RD Jr, Christianson, TJ, et al. Adult primary central nervous system vasculitis treatment and course: analysis of one hundred sixty-three patients. Arthritis Rheumatol (Hoboken, NJ) 2015;67:16371645.Google Scholar
Schuster, S, Ozga, AK, Stellmann, JP, et al. Relapse rates and long-term outcome in primary angiitis of the central nervous system. J Neurol 2019;266:14811489.Google Scholar
Salvarani, C, Brown, RD Jr, Muratore, F, et al. Rituximab therapy for primary central nervous system vasculitis: a 6 patient experience and review of the literature. Autoimmunity Rev 2019;18:399405.Google Scholar
de Boysson, H, Arquizan, C, Touze, E, et al. Treatment and long-term outcomes of primary central nervous system vasculitis. Stroke 2018;49:19461952.Google Scholar
Rottino, A, Hoffman, G. A sarcoid form of encephalitis in a patient with Hodgkin’s disease: case report with review of the literature. J Neuropathol Exp Neurol 1950;9:103108.Google Scholar
Rewcastle, NB, Tom, MI. Non-infectious granulomatous angiitis of the nervous system associated with Hodgkin’s disease. J Neurol Neurosurg Psychiatry 1962;25:5158.Google Scholar
Salvarani, C, Brown, RD Jr, Christianson, TJH, et al. Primary central nervous system vasculitis associated with lymphoma. Neurology 2018;90:e847e855.Google Scholar
Rosen, CL, DePalma, L, Morita, A. Primary angiitis of the central nervous system as a first presentation in Hodgkin’s disease: a case report and review of the literature. Neurosurgery 2000;46:15041508.Google Scholar
Delobel, P, Brassat, D, Danjoux, M, et al. Granulomatous angiitis of the central nervous system revealing Hodgkin’s disease. J Neurol 2004;251:611612.Google Scholar
Le Guennec, L, Roos-Weil, D, Mokhtari, K, et al. Granulomatous angiitis of the CNS revealing a Hodgkin lymphoma. Neurology 2013;80:323324.Google Scholar
Lopez-Chiriboga, AS, Yoon, JW, Siegel, JL, et al. Granulomatous angiitis of the central nervous system associated with hodgkin’s lymphoma: case report and literature review. J Stroke Cerebrovasc Dis 2018;27:e5e8.Google Scholar
Rosenblum, WI, Hadfield, MG. Granulomatous angiitis of the nervous system in cases of herpes zoster and lymphosarcoma. Neurology 1972;22:348354.Google Scholar
Graus, F, Arino, H, Dalmau, J. Paraneoplastic neurological syndromes in Hodgkin and non-Hodgkin lymphomas. Blood 2014;123:32303238.Google Scholar
Sheehy, N, Sheehan, K, Brett, F, et al. Hodgkins disease presenting with granulomatous angiitis of the central nervous system. J Neurol 2003;250:112113.Google Scholar
Power, DG, Mullholland, PJ, Sheehy, N, Farrell, MA, Daly, PA. Relapsing granulomatous angiitis of the central nervous system in a patient while in remission from Hodgkin lymphoma. Irish Med J 2006;99:282.Google Scholar
Fuehrer, NE, Hammack, JE, Morris, JM, Kaufmann, TJ, Giannini, C. Teaching NeuroImages: granulomatous angiitis of the CNS associated with Hodgkin lymphoma. Neurology 2011;77:e110e111.Google Scholar
Smith, EE, Charidimou, A, Ayata, C, Werring, DJ, Greenberg, SM. Cerebral amyloid angiopathy-related transient focal neurologic episodes. Neurology 2021;97:231238.Google Scholar
Probst, A, Ulrich, J. Amyloid angiopathy combined with granulomatous angiitis of the central nervous system: report on two patients. Clin Neuropathol 1985;4:250259.Google Scholar
Murphy, MN, Sima, AA. Cerebral amyloid angiopathy associated with giant cell arteritis: a case report. Stroke 1985;16:514517.Google Scholar
Schwab, P, Lidov, HG, Schwartz, RB, Anderson, RJ. Cerebral amyloid angiopathy associated with primary angiitis of the central nervous system: report of 2 cases and review of the literature. Arthritis Rheum 2003;49:421427.Google Scholar
Kinnecom, C, Lev, MH, Wendell, L, et al. Course of cerebral amyloid angiopathy-related inflammation. Neurology 2007;68:14111416.Google Scholar
Scolding, NJ, Joseph, F, Kirby, PA, et al. Abeta-related angiitis: primary angiitis of the central nervous system associated with cerebral amyloid angiopathy. Brain 2005;128:500515.Google Scholar
Corovic, A, Kelly, S, Markus, HS. Cerebral amyloid angiopathy associated with inflammation: a systematic review of clinical and imaging features and outcome. Intl J Stroke 2018;13:257267.Google Scholar
Danve, A, Grafe, M, Deodhar, A. Amyloid beta-related angiitis: a case report and comprehensive review of literature of 94 cases. Semin Arthritis Rheumat 2014;44:8692.Google Scholar
Sakai, K, Ueda, M, Fukushima, W, et al. Nationwide survey on cerebral amyloid angiopathy in Japan. Eur J Neurol 2019;26:14871493.Google Scholar
Salvarani, C, Hunder, GG, Morris, JM, et al. Abeta-related angiitis: comparison with CAA without inflammation and primary CNS vasculitis. Neurology 2013;81:15961603.Google Scholar
Salvarani, C, Morris, JM, Giannini, C, et al. Imaging findings of cerebral amyloid angiopathy, abeta-related angiitis (ABRA), and cerebral amyloid angiopathy-related inflammation: a single-institution 25-year experience. Medicine (Baltimore) 2016;95:e3613.Google Scholar
Kusakabe, K, Inoue, A, Matsumoto, S, et al. Cerebral amyloid angiopathy-related inflammation with epilepsy mimicking a presentation of brain tumor: a case report and review of the literature. Intl J Surg Case Rep 2018;48:95100.Google Scholar
Ronsin, S, Deiana, G, Geraldo, AF, et al. Pseudotumoral presentation of cerebral amyloid angiopathy-related inflammation. Neurology 2016;86:912919.Google Scholar
Chung, KK, Anderson, NE, Hutchinson, D, Synek, B, Barber, PA. Cerebral amyloid angiopathy related inflammation: three case reports and a review. J Neurol Neurosurg Psychiatry 2011;82:2026.Google Scholar
Renard, D, Tatu, L, Collombier, L, et al. Cerebral amyloid angiopathy and cerebral amyloid angiopathy-related inflammation: comparison of hemorrhagic and DWI MRI Features. J Alzheimer Dis 2018;64:11131121.Google Scholar
Auriel, E, Charidimou, A, Gurol, ME, et al. Validation of clinicoradiological criteria for the diagnosis of cerebral amyloid angiopathy-related inflammation. JAMA Neurol 2016;73:197202.Google Scholar
Regenhardt, RW, Thon, JM, Das, AS, et al. Association between immunosuppressive treatment and outcomes of cerebral amyloid angiopathy-related inflammation. JAMA Neurol 2020;77:110.Google Scholar
Kloppenborg, RP, Richard, E, Sprengers, ME, et al. Steroid responsive encephalopathy in cerebral amyloid angiopathy: a case report and review of evidence for immunosuppressive treatment. J Neuroinflammation 2010;7:18.Google Scholar
Traschutz, A, Tzaridis, T, Penner, AH, et al. Reduction of microbleeds by immunosuppression in a patient with Abeta-related vascular inflammation. Neurol Neuroimmunol Neuroinflamm 2015;2:e165.Google Scholar
Kleffner, I, Dorr, J, Ringelstein, M, et al. Diagnostic criteria for Susac syndrome. J Neurol Neurosurg Psychiatry 2016;87:12871295.Google Scholar
Susac, JO, Hardman, JM, Selhorst, JB. Microangiopathy of the brain and retina. Neurology 1979;29:313316.Google Scholar
Dorr, J, Krautwald, S, Wildemann, B, et al. Characteristics of Susac syndrome: a review of all reported cases. Nat Rev Neurol 2013;9:307316.Google Scholar
Mateen, FJ, Zubkov, AY, Muralidharan, R, et al. Susac syndrome: clinical characteristics and treatment in 29 new cases. Eur J Neurol 2012;19:800811.Google Scholar
Susac, JO, Murtagh, FR, Egan, RA, et al. MRI findings in Susac’s syndrome. Neurology 2003;61:17831787.Google Scholar
Ringelstein, M, Albrecht, P, Kleffner, I, et al. Retinal pathology in Susac syndrome detected by spectral-domain optical coherence tomography. Neurology 2015;85:610618.Google Scholar
Agamanolis, DP, Klonk, C, Bigley, K, Rennebohm, RM. Neuropathological findings in Susac syndrome: an autopsy report. J Neuropathol Exp Neurol 2019;78:515519.Google Scholar
Heiskala, H, Somer, H, Kovanen, J, et al. Microangiopathy with encephalopathy, hearing loss and retinal arteriolar occlusions: two new cases. J Neurol Sci 1988;86:239250.Google Scholar
Hardy, TA, O’Brien, B, Gerbis, N, et al. Brain histopathology in three cases of Susac’s syndrome: implications for lesion pathogenesis and treatment. J Neurol Neurosurg Psychiatry 2015;86:582584.Google Scholar
Magro, CM, Poe, JC, Lubow, M, Susac, JO. Susac syndrome: an organ-specific autoimmune endotheliopathy syndrome associated with anti-endothelial cell antibodies. Am J Clin Pathol 2011;136:903912.Google Scholar
Jarius, S, Kleffner, I, Dorr, JM, et al. Clinical, paraclinical and serological findings in Susac syndrome: an international multicenter study. J Neuroinflammation 2014;11:46.Google Scholar
Servettaz, A, Guilpain, P, Tamas, N, et al. Natural anti-endothelial cell antibodies. Autoimmunity Rev 2008;7:426430.Google Scholar
Gross, CC, Meyer, C, Bhatia, U, et al. CD8(+) T cell-mediated endotheliopathy is a targetable mechanism of neuro-inflammation in Susac syndrome. Nat Commun 2019;10:5779.Google Scholar
Rennebohm, RM, Susac, JO. Treatment of Susac’s syndrome. J Neurol Sci 2007;257:215220.Google Scholar
Rennebohm, RM, Asdaghi, N, Srivastava, S, Gertner, E. Guidelines for treatment of Susac syndrome: an update. Intl J Stroke 2018;15:484494.Google Scholar
Vodopivec, I, Prasad, S. Treatment of Susac syndrome. Curr Treat Options Neurol 2016;18:3.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
×