Skip to main content Accessibility help
×
Hostname: page-component-76fb5796d-zzh7m Total loading time: 0 Render date: 2024-04-27T15:34:35.677Z Has data issue: false hasContentIssue false

Chapter 21 - Abnormal Movements in Neurological Autoimmune Disorders

from Section 4 - Autoimmunity in Neurological and Psychiatric Diseases

Published online by Cambridge University Press:  27 January 2022

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

Summary

In this chapter we describe different types of movement disorders that associate with autoimmune encephalitis, and the antibodies more frequently involved. In children the most common disorders are Sydenham chorea and anti-NMDAR encephalitis. Abnormal movements occur in ~80% of patients with anti-NMDAR encephalitis and include multiple different types such as chorea, oromandibular dystonia, stereotypies, opistotonus, catatonia, or myorhythmia. Children who develop anti-NMDAR encephalitis as a complication of previous herpes simplex viral encephalitis present prominent generalized chorea or choreoathetosis. In adults the most frequent autoimmune neurological disease that associates with movement disorders is anti-IgLON5 disease. More than 80% of patients this disease develop at least one type of movement disorder; gait instability or ataxia associated with craniofacial dyskinesias or generalized chorea are the most common combination of movement disorders. Hyperekplexia is a major manifestation of progressive encephalomyelitis with rigidity and myoclonus (PERM), which is usually associated with glycine receptor antibodies; some patients with similar symptoms have DPPX antibodies. Autoimmune chorea in adults may also be a paraneoplastic manifestation of small-cell lung cancer and CRMP5 antibodies. The most common paroxysmal abnormal movement of autoimmune origin is faciobrachial dystonic seizures associated with LGI1 antibodies. Patients with anti-CASPR2 encephalitis may have paroxysmal episodes of cerebellar ataxia that precede the encephalitis. Anti-CASPR2 encephalitis can also cause orthostatic myoclonus.

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

Jankovic, J. Movement disorders in 2016: progress in Parkinson disease and other movement disorders. Nat Rev Neurol 2017;13:7678.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
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
Baizabal-Carvallo, JF, Jankovic, J. Autoimmune and paraneoplastic movement disorders: an update. J Neurol Sci 2018;385:175184.Google Scholar
Caviness, JN, Forsyth, PA, Layton, DD, McPhee, TJ. The movement disorder of adult opsoclonus. Mov Disord 1995;10:2227.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
Hara, M, Arino, H, Petit-Pedrol, M, et al. DPPX antibody-associated encephalitis: main syndrome and antibody effects. Neurology 2017;88:13401348.CrossRefGoogle ScholarPubMed
Gaig, C, Iranzo, A, Cajochen, C, et al. Characterization of the sleep disorder of anti-IgLON5 disease. Sleep 2019;42:zsz133.CrossRefGoogle ScholarPubMed
Gaig, C, Compta, Y. Neurological profiles beyond the sleep disorder in patients with anti-IgLON5 disease. Curr Opin Neurol 2019;32:493499.Google Scholar
Dalmau, J, Graus, F, Villarejo, A, et al. Clinical analysis of anti-Ma2-associated encephalitis. Brain 2004;127:18311844.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
Ali, F, Wijdicks, EF. Treatment of movement disorder emergencies in autoimmune encephalitis in the neurosciences ICU. Neurocrit Care 2020;32:286294.Google Scholar
Cardoso, F, Seppi, K, Mair, KJ, Wenning, GK, Poewe, W. Seminar on choreas. Lancet Neurol 2006;5:589602.Google Scholar
Sanger, TD, Chen, D, Fehlings, DL, et al. Definition and classification of hyperkinetic movements in childhood. Mov Disord 2010;25:15381549.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. 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
Duan, BC, Weng, WC, Lin, KL, et al. Variations of movement disorders in anti-N-methyl-D-aspartate receptor encephalitis: a nationwide study in Taiwan. Medicine (Baltimore) 2016;95:e4365.CrossRefGoogle ScholarPubMed
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
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
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
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
Erer Ozbek, S, Yapici, Z, Tuzun, E, et al. A case of hyperkinetic movement disorder associated with LGI1 antibodies. Turkish J Pediatr 2015;57:514517.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
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
Dale, RC, Merheb, V, Pillai, S, et al. Antibodies to surface dopamine-2 receptor in autoimmune movement and psychiatric disorders. Brain 2012;135:34533468.Google Scholar
Dale, RC, Church, AJ, Surtees, RA, et al. Encephalitis lethargica syndrome: 20 new cases and evidence of basal ganglia autoimmunity. Brain 2004;127:2133.Google Scholar
Dale, RC, Irani, SR, Brilot, F, et al. N-methyl-D-aspartate receptor antibodies in pediatric dyskinetic encephalitis lethargica. Ann Neurol 2009;66:704709.Google Scholar
Sinmaz, N, Tea, F, Pilli, D, et al. Dopamine-2 receptor extracellular N-terminus regulates receptor surface availability and is the target of human pathogenic antibodies from children with movement and psychiatric disorders. Acta neuropathologica communications 2016;4:126.Google Scholar
Kirvan, CA, Swedo, SE, Kurahara, D, Cunningham, MW. Streptococcal mimicry and antibody-mediated cell signaling in the pathogenesis of Sydenham’s chorea. Autoimmunity 2006;39:2129.Google Scholar
Cunningham, MW, Cox, CJ. Autoimmunity against dopamine receptors in neuropsychiatric and movement disorders: a review of Sydenham chorea and beyond. Acta physiologica (Oxford, England) 2016;216:90100.Google Scholar
Cardoso, F. Autoimmune choreas. J Neurol Neurosurg Psychiatry 2017;88:412417.Google Scholar
Cardoso, F, Eduardo, C, Silva, AP, Mota, CC. Chorea in fifty consecutive patients with rheumatic fever. Mov Disord 1997;12:701703.CrossRefGoogle ScholarPubMed
Maia, DP, Teixeira, AL Jr, Quintao Cunningham, MC, Cardoso, F. Obsessive compulsive behavior, hyperactivity, and attention deficit disorder in Sydenham chorea. Neurology 2005;64:17991801.Google Scholar
Singer, HS. Autoantibody-associated movement disorders in children: proven and proposed. Semin Pediatr Neurol 2017;24:168179.Google Scholar
Brilot, F, Merheb, V, Ding, A, Murphy, T, Dale, RC. Antibody binding to neuronal surface in Sydenham chorea, but not in PANDAS or Tourette syndrome. Neurology 2011;76:15081513.Google Scholar
Wilbur, C, Bitnun, A, Kronenberg, S, et al. PANDAS/PANS in childhood: Controversies and evidence. Paediatr Child Health 2019;24:8591.Google Scholar
Weiner, SG, Normandin, PA. Sydenham chorea: a case report and review of the literature. Pediatr Emerg Care 2007;23:2024.Google Scholar
Elevli, M, Celebi, A, Tombul, T, Gokalp, AS. Cardiac involvement in Sydenham’s chorea: clinical and Doppler echocardiographic findings. Acta Paediatr 1999;88:10741077.Google Scholar
Gurkas, E, Karalok, ZS, Taskin, BD, et al. Predictors of recurrence in Sydenham’s chorea: clinical observation from a single center. Brain Dev 2016;38:827834.Google Scholar
Cardoso, F, Vargas, AP, Oliveira, LD, Guerra, AA, Amaral, SV. Persistent Sydenham’s chorea. Mov Disord 1999;14:805807.Google Scholar
Korn-Lubetzki, I, Brand, A. Sydenham’s chorea in Jerusalem: still present. Israel Med Assoc J 2004;6:460462.Google Scholar
Dean, SL, Singer, HS. Treatment of Sydenham’s chorea: a review of the current evidence. Tremor Other Hyperkinet Mov 2017;7:456.Google Scholar
Pena, J, Mora, E, Cardozo, J, Molina, O, Montiel, C. Comparison of the efficacy of carbamazepine, haloperidol and valproic acid in the treatment of children with Sydenham’s chorea: clinical follow-up of 18 patients. Arq Neuropsiquiatr 2002;60:374377.Google Scholar
Genel, F, Arslanoglu, S, Uran, N, Saylan, B. Sydenham’s chorea: clinical findings and comparison of the efficacies of sodium valproate and carbamazepine regimens. Brain Dev 2002;24:7376.Google Scholar
Teixeira, AL, Cardoso, F, Maia, DP, Cunningham, MC. Sydenham’s chorea may be a risk factor for drug induced parkinsonism. J Neurol Neurosurg Psychiatry 2003;74:13501351.Google Scholar
Paz, JA, Silva, CA, Marques-Dias, MJ. Randomized double-blind study with prednisone in Sydenham’s chorea. Pediatr Neurol 2006;34:264269.Google Scholar
Garvey, MA, Snider, LA, Leitman, SF, Werden, R, Swedo, SE. Treatment of Sydenham’s chorea with intravenous immunoglobulin, plasma exchange, or prednisone. J Child Neurol 2005;20:424429.Google Scholar
Husby, G, van de Rijn, I, Zabriskie, JB, Abdin, ZH, Williams, RC Jr. Antibodies reacting with cytoplasm of subthalamic and caudate nuclei neurons in chorea and acute rheumatic fever. J Exp Med 1976;144:10941110.Google Scholar
Singer, HS, Loiselle, CR, Lee, O, Garvey, MA, Grus, FH. Anti-basal ganglia antibody abnormalities in Sydenham chorea. J Neuroimmunol 2003;136:154161.Google Scholar
Kirvan, CA, Swedo, SE, Heuser, JS, Cunningham, MW. Mimicry and autoantibody-mediated neuronal cell signaling in Sydenham chorea. Nat Med 2003;9:914920.Google Scholar
Kirvan, CA, Cox, CJ, Swedo, SE, Cunningham, MW. Tubulin is a neuronal target of autoantibodies in Sydenham’s chorea. J Immunol 2007;178:74127421.Google Scholar
Shimasaki, C, Frye, RE, Trifiletti, R, et al. Evaluation of the Cunningham Panel in pediatric autoimmune neuropsychiatric disorder associated with streptococcal infection (PANDAS) and pediatric acute-onset neuropsychiatric syndrome (PANS): changes in antineuronal antibody titers parallel changes in patient symptoms. J Neuroimmunol 2019;339:577138.Google Scholar
Hesselmark, E, Bejerot, S. Biomarkers for diagnosis of Pediatric Acute Neuropsychiatric Syndrome (PANS): sensitivity and specificity of the Cunningham Panel. J Neuroimmunol 2017;312:3137.CrossRefGoogle ScholarPubMed
O’Toole, O, Lennon, VA, Ahlskog, JE, et al. Autoimmune chorea in adults. Neurology 2013;80:11331144.Google Scholar
Lamby, N, Leypoldt, F, Schulz, JB, Tauber, SC. Atypical presentation of anti-Ma2-associated encephalitis with choreiform movement. Neurol Neuroimmunol Neuroinflamm 2019;6:e557.Google Scholar
Asherson, RA, Derksen, RH, Harris, EN, et al. Chorea in systemic lupus erythematosus and ‘lupus-like’ disease: association with antiphospholipid antibodies. Semin Arthritis Rheumat 1987;16:253259.CrossRefGoogle ScholarPubMed
Cervera, R, Piette, JC, Font, J, et al. Antiphospholipid syndrome: clinical and immunologic manifestations and patterns of disease expression in a cohort of 1,000 patients. Arthritis Rheum 2002;46:10191027.Google Scholar
Cervera, R, Asherson, RA, Font, J, et al. Chorea in the antiphospholipid syndrome. Clinical, radiologic, and immunologic characteristics of 50 patients from our clinics and the recent literature. Medicine (Baltimore) 1997;76:203212.Google Scholar
Reiner, P, Galanaud, D, Leroux, G, et al. Long-term outcome of 32 patients with chorea and systemic lupus erythematosus or antiphospholipid antibodies. Mov Disord 2011;26:24222427.Google Scholar
Dale, RC, Yin, K, Ding, A, et al. Antibody binding to neuronal surface in movement disorders associated with lupus and antiphospholipid antibodies. Dev Med Child Neurol 2011;53:522528.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
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
Kujawa, KA, Niemi, VR, Tomasi, MA, et al. Ballistic-choreic movements as the presenting feature of renal cancer. Arch Neurol 2001;58:11331135.Google Scholar
Sheen, VL, Asimakopoulos, F, Heyman, E, Henderson, G, Feske, SK. Hemichorea as a presentation of recurrent non-Hodgkin’s lymphoma. J Neurol 2002;249:17461748.CrossRefGoogle ScholarPubMed
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;12:262.Google Scholar
Gupta, HV, Gervais, C, Ross, MA, Mehta, SH. Purkinje cell cytoplasmic antibody (PCA-2)-related chorea-dystonia syndrome. Tremor Other Hyperkinet Mov 2016;6:420.Google Scholar
Goldstein, L, Djaldetti, R, Benninger, F. Anti-Yo, chorea and hemiballismus: a case report. J Clin Neurosci 2017;42:113114.CrossRefGoogle ScholarPubMed
Feinstein, E, Walker, R. An update on the treatment of chorea. Curr Treat Options Neurol 2018;20:44.CrossRefGoogle ScholarPubMed
Gaig, C, Graus, F, Compta, Y, et al. Clinical manifestations of the anti-IgLON5 disease. Neurology 2017;88:17361743.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
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
Dalmau, J, Graus, F. Antibody-mediated encephalitis. N Engl J Med 2018;378:840851.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 2014;4:tre-04-213-4821-1.Google Scholar
Colletta, K, Kartha, N, Chawla, J. Paraneoplastic puzzle: an unusual case of hemichorea, renal cell carcinoma, and LGI1 antibody. Mov Disord Clin Pract 2018;5:337338.Google Scholar
Edwards, MJ, Lang, AE, Bhatia, KP. Stereotypies: a critical appraisal and suggestion of a clinically useful definition. Mov Disord 2012;27:179185.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
Mohammad, SS, Fung, VS, Grattan-Smith, P, et al. Movement disorders in children with anti-NMDAR encephalitis and other autoimmune encephalopathies. Mov Disord 2014;29:15391542.Google Scholar
Spatola, M, Petit-Pedrol, M, Simabukuro, MM, et al. Investigations in GABAA receptor antibody-associated encephalitis. Neurology 2017;88:10121020.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
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
Baizabal-Carvallo, JF, Cardoso, F, Jankovic, J. Myorhythmia: phenomenology, etiology, and treatment. Mov Disord 2015;30:171179.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
Simpson, DA, Wishnow, R, Gargulinski, RB, Pawlak, AM. Oculofacial-skeletal myorhythmia in central nervous system Whipple’s disease: additional case and review of the literature. Mov Disord 1995;10:195200.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
Une, H, Matsuse, D, Uehara, T, et al. Branchial myorhythmia in a case of systemic lupus erythematosus. J Neurol Sci 2020;408:116501.Google Scholar
Pittock, SJ, Lucchinetti, CF, Lennon, VA. Anti-neuronal nuclear autoantibody type 2: paraneoplastic accompaniments. Ann Neurol 2003;53:580587.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
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
Clerinx, K, Breban, T, Schrooten, M, et al. Progressive encephalomyelitis with rigidity and myoclonus: resolution after thymectomy. Neurology 2011;76:303304.Google Scholar
Sarkis, RA, Coffey, MJ, Cooper, JJ, Hassan, I, Lennox, B. Anti-N-methyl-D-aspartate receptor encephalitis: a review of psychiatric phenotypes and management considerations: a report of the American Neuropsychiatric Association Committee on Research. J Neuropsychiatr Clin Neurosci 2019;31:137142.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
Kurtis, MM, Toledano, R, Garcia-Morales, I, Gil-Nagel, A. Immunomodulated parkinsonism as a presenting symptom of LGI1 antibody encephalitis. Parkinsonism Relat Disord 2015;21:12861287.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
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
Pittock, SJ, Lucchinetti, CF, Lennon, VA. Anti-neuronal nuclear autoantibody type 2: paraneoplastic accompaniments. Ann Neurol 2003;53:580587.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
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
Hoglinger, GU, Respondek, G, Stamelou, M, et al. Clinical diagnosis of progressive supranuclear palsy: the Movement Disorder Society criteria. Mov Disord 2017;32:853864.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
Gaig, C, Compta, Y, Heidbreder, A, et al. Frequency and characterization of movement disorders in anti-IgLON5 disease. Neurology 2021;97:e1367–1381.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
Bonello, M, Jacob, A, Ellul, MA, et al. IgLON5 disease responsive to immunotherapy. Neurol Neuroimmunol Neuroinflamm 2017;4:e383.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
Chang, VC, Frucht, SJ. Myoclonus. Curr Treat Options Neurol 2008;10:222229.Google Scholar
Fahn, S, Marsden, CD, Van Woert, MH. Definition and classification of myoclonus. Adv Neurol 1986;43:15.Google Scholar
Shibasaki, H, Hallett, M. Electrophysiological studies of myoclonus. Muscle Nerve 2005;31:157174.Google Scholar
Shibasaki, H. Neurophysiological classification of myoclonus. Neurophysiologie clinique 2006;36:267269.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
Tan, KM, Lennon, VA, Klein, CJ, Boeve, BF, Pittock, SJ. Clinical spectrum of voltage-gated potassium channel autoimmunity. Neurology 2008;70:18831890.Google Scholar
Roobol, TH, Kazzaz, BA, Vecht, CJ. Segmental rigidity and spinal myoclonus as a paraneoplastic syndrome. J Neurol Neurosurg Psychiatry 1987;50:628631.Google Scholar
Pittock, SJ, Lucchinetti, CF, Parisi, JE, et al. Amphiphysin autoimmunity: paraneoplastic accompaniments. Ann Neurol 2005;58:96107.Google Scholar
Hines, H, Murray, NM, Ahmad, S, Jaradeh, S, Gold, CA. Video NeuroImages: paraneoplastic spinal myoclonus associated with Caspr2 antibodies. Neurology 2018;90:660661.Google Scholar
Govert, F, Witt, K, Erro, R, et al. Orthostatic myoclonus associated with Caspr2 antibodies. Neurology 2016;86:13531355.Google Scholar
Nanaura, H, Kataoka, H, Kiriyama, T, et al. Spinal segmental myoclonus in both legs associated with antibodies to glycine receptors. Neurol Clin Pract 2019;9:176177.Google Scholar
Bien, CG, Elger, CE. Epilepsia partialis continua: semiology and differential diagnoses. Epileptic Disord 2008;10:37.Google Scholar
Obeso, JA, Rothwell, JC, Marsden, CD. The spectrum of cortical myoclonus: from focal reflex jerks to spontaneous motor epilepsy. Brain 1985;108:193–224.Google Scholar
Cockerell, OC, Rothwell, J, Thompson, PD, Marsden, CD, Shorvon, SD. Clinical and physiological features of epilepsia partialis continua. Cases ascertained in the UK. Brain 1996;119:393–407.Google Scholar
Olson, JA, Olson, DM, Sandborg, C, Alexander, S, Buckingham, B. Type 1 diabetes mellitus and epilepsia partialis continua in a 6-year-old boy with elevated anti-GAD65 antibodies. Pediatrics 2002;109:E50.Google Scholar
Mukherjee, V, Mukherjee, A, Mukherjee, A, Halder, A. Type I diabetes mellitus in a child presenting with epilepsy partialis continua. J Indian Med Assoc 2007;105:340342.Google Scholar
Baglietto, MG, Mancardi, MM, Giannattasio, A, et al. Epilepsia partialis continua in type 1 diabetes: evolution into epileptic encephalopathy with continuous spike-waves during slow sleep. Neurol Sci 2009;30:509512.Google Scholar
Triplett, J, Vijayan, S, MacDonald, A, et al. Fulminant anti-GAD antibody encephalitis presenting with status epilepticus requiring aggressive immunosuppression. J Neuroimmunol 2018;323:119124.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
Kim, EH, Kim, YJ, Ko, TS, Yum, MS, Lee, JH. A young child of anti-NMDA receptor encephalitis presenting with epilepsia partialis continua: the first pediatric case in Korea. Korean J Pediatr 2016;59:S133S138.Google Scholar
Katsuse, K, Shimizu, G, Saito Sato, N, et al. Epilepsia partialis continua as an early sign of anti-myelin oligodendrocyte glycoprotein antibody-positive encephalitis. Intern Med (Tokyo, Japan) 2020;59:14451449.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
Rudzinski, LA, Pittock, SJ, McKeon, A, et al. Extratemporal EEG and MRI findings in ANNA-1 (anti-Hu) encephalitis. Epilepsy Res 2011;95:255262.Google Scholar
Khasani, S, Becker, K, Meinck, HM. Hyperekplexia and stiff-man syndrome: abnormal brainstem reflexes suggest a physiological relationship. J Neurol Neurosurg Psychiatry 2004;75:12651269.Google Scholar
Bakker, MJ, van Dijk, JG, van den Maagdenberg, AM, Tijssen, MA. Startle syndromes. Lancet Neurol 2006;5:513524.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
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
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
Hutchinson, M, Waters, P, McHugh, J, et al. Progressive encephalomyelitis, rigidity, and myoclonus: a novel glycine receptor antibody. Neurology 2008;71:12911292.Google Scholar
Zutt, R, Elting, JW, van Zijl, JC, et al. Electrophysiologic testing aids diagnosis and subtyping of myoclonus. Neurology 2018;90:e647e657.Google Scholar
Murinson, BB, Guarnaccia, JB. Stiff-person syndrome with amphiphysin antibodies: distinctive features of a rare disease. Neurology 2008;71:19551958.Google Scholar
Wijntjes, J, Bechakra, M, Schreurs, MWJ, et al. Pruritus in anti-DPPX encephalitis. Neurol Neuroimmunol Neuroinflamm 2018;5:e455.Google Scholar
Mas, N, Saiz, A, Leite, MI, et al. Antiglycine-receptor encephalomyelitis with rigidity. J Neurol Neurosurg Psychiatry 2011;82:13991401.Google Scholar
Netravathi, M, Saini, J, Mahadevan, A, et al. Is pruritus an indicator of aquaporin-positive neuromyelitis optica? Mult Scler 2017;23:810817.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
Hart, IK, Maddison, P, Newsom-Davis, J, Vincent, A, Mills, KR. Phenotypic variants of autoimmune peripheral nerve hyperexcitability. Brain 2002;125:18871895.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
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
Maddison, P. Neuromyotonia. Clin Neurophysiol 2006;117:21182127.Google Scholar
Elangovan, C, Morawo, A, Ahmed, A. Current treatment options for peripheral nerve hyperexcitability syndromes. Curr Treat Options Neurol 2018;20:23.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. J Neurol Neurosurg Psychiatry 1994;57:840842.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
Rubio-Agusti, I, Perez-Miralles, F, Sevilla, T, et al. Peripheral nerve hyperexcitability: a clinical and immunologic study of 38 patients. Neurology 2011;76:172178.Google Scholar
Evoli, A, Minicuci, GM, Vitaliani, R, et al. Paraneoplastic diseases associated with thymoma. J Neurol 2007;254:756762.Google Scholar
Gastaldi, M, De Rosa, A, Maestri, M, et al. Acquired neuromyotonia in thymoma-associated myasthenia gravis: a clinical and serological study. Eur J Neurol 2019;26:992999.Google Scholar
van Sonderen, A, Wirtz, PW, Verschuuren, JJ, Titulaer, MJ. Paraneoplastic syndromes of the neuromuscular junction: therapeutic options in myasthenia gravis, Lambert–Eaton myasthenic syndrome, and neuromyotonia. Curr Treat Options Neurol 2013;15:224239.Google Scholar
Rana, SS, Ramanathan, RS, Small, G, Adamovich, B. Paraneoplastic Isaacs’ syndrome: a case series and review of the literature. J Clin Neuromusc Dis 2012;13:228233.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
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
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
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
Rubio, I, Bataller, L, Perez-Miralles, F, et al. Peripheral nerve hyperexcitability (PNH): a clinical study of 26 patients. Neurology 2009;72:A56.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
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
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
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
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
Lopez Chiriboga, AS, Pittock, S. Episodic ataxia in CASPR2 autoimmunity. Neurol Neuroimmunol Neuroinflamm 2019;6:e536.Google Scholar
Xia, C, Dubeau, F. Teaching video NeuroImages: dystonic posturing in anti-NMDA receptor encephalitis. Neurology 2011;76:e80.Google Scholar
Rubio-Agusti, I, Dalmau, J, Sevilla, T, et al. Isolated hemidystonia associated with NMDA receptor antibodies. Mov Disord 2011;26:351352.Google Scholar
Liu, J, Zhang, Q, Lian, Z, et al. Painful tonic spasm in neuromyelitis optica spectrum disorders: prevalence, clinical implications and treatment options. Mult Scler Relat Disord 2017;17:99102.Google Scholar
Li, QY, Wang, B, Yang, J, et al. Painful tonic spasm in Chinese patients with neuromyelitis optica spectrum disorder: prevalence, subtype, and features. Mult Scler Relat Disord 2020;45:102408.Google Scholar
Kim, SM, Go, MJ, Sung, JJ, Park, KS, Lee, KW. Painful tonic spasm in neuromyelitis optica: incidence, diagnostic utility, and clinical characteristics. Arch Neurol 2012;69:10261031.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
Meinck, HM, Thompson, PD. Stiff man syndrome and related conditions. Mov Disord 2002;17:853866.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
×