Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-2xdlg Total loading time: 0 Render date: 2024-07-07T18:33:15.909Z Has data issue: false hasContentIssue false

Chapter 24 - Autoimmune Dementia: A Useful Term?

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

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

Access options

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

References

Dalmau, J, Graus, F. Antibody-mediated encephalitis. N Engl J Med 2018;378:840851.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.CrossRefGoogle ScholarPubMed
Brooks-Kayal, AR, Russek, SJ. Regulation of GABA(A) receptor gene expression and epilepsy. In: Noebels, JL, Avoli, M, Rogawski, MA, eds. Jasper’s Basic Mechanisms of the Epilepsies. Bethesda, MD: National Center for Biotechnology Information, 2012.Google Scholar
Olney, JW, Farber, NB. Glutamate receptor dysfunction and schizophrenia. Arch Gen Psychiatry 1995;52:9981007.Google Scholar
Nakanishi, S. Molecular diversity of glutamate receptors and implications for brain function. Science 1992;258:597603.Google Scholar
Irani, SR, Michell, AW, Lang, B, et al. Faciobrachial dystonic seizures precede LGI1 antibody limbic encephalitis. Ann Neurol 2011;69:892900.CrossRefGoogle ScholarPubMed
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
Dalmau, J, Armangue, T, Planaguma, J, et al. An update on anti-NMDA receptor encephalitis for neurologists and psychiatrists: mechanisms and models. Lancet Neurol 2019;18:10451057.Google Scholar
Do, LD, Chanson, E, Desestret, V, et al. Characteristics in limbic encephalitis with anti-adenylate kinase 5 autoantibodies. Neurology 2017;88:514524.CrossRefGoogle ScholarPubMed
Flanagan, EP, McKeon, A, Lennon, VA, et al. Autoimmune dementia: clinical course and predictors of immunotherapy response. Mayo Clin Proc 2010;85:881897.CrossRefGoogle ScholarPubMed
Dubey, D, Singh, J, Britton, JW, et al. Predictive models in the diagnosis and treatment of autoimmune epilepsy. Epilepsia 2017;58:11811189.Google Scholar
Pollak, TA, Lennox, BR, Müller, 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.CrossRefGoogle Scholar
Toledano, M, Pittock, SJ. Autoimmune epilepsy. Semin Neurol 2015;35:245258.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.CrossRefGoogle ScholarPubMed
Graus, F, Saiz, A, Dalmau, J. GAD antibodies in neurological disorders: insights and challenges. Nat Rev Neurol 2020;16:353365.CrossRefGoogle ScholarPubMed
Bozzetti, S, Rossini, F, Ferrari, S, et al. Epileptic seizures of suspected autoimmune origin: a multicentre retrospective study. J Neurol Neurosurg Psychiatry 2020;91:11451153.CrossRefGoogle ScholarPubMed
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
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.CrossRefGoogle ScholarPubMed
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
Dahm, L, Ott, C, Steiner, J, et al. Seroprevalence of autoantibodies against brain antigens in health and disease. Ann Neurol 2014;76:8294.Google Scholar
Hammer, C, Stepniak, B, Schneider, A, et al. Neuropsychiatric disease relevance of circulating anti-NMDA receptor autoantibodies depends on blood–brain barrier integrity. Mol Psychiatry 2014;19:11431149.CrossRefGoogle ScholarPubMed
Castillo-Gomez, E, Oliveira, B, Tapken, D, et al. All naturally occurring autoantibodies against the NMDA receptor subunit NR1 have pathogenic potential irrespective of epitope and immunoglobulin class. Mol Psychiatry 2016;22:17761784.Google Scholar
Lennox, BR, Palmer-Cooper, EC, Pollak, T, et al. Prevalence and clinical characteristics of serum neuronal cell surface antibodies in first-episode psychosis: a case–control study. Lancet Psychiatry 2017;4:4248.Google Scholar
Schou, MB, Saether, SG, Drange, OK, et al. The significance of anti-neuronal antibodies for acute psychiatric disorders: a retrospective case-controlled study. BMC Neurosci 2018;19:68.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
Michaelson, DM, Alroy, G, Goldstein, D, Chapman, J, Feldon, J. Characterization of an experimental autoimmune dementia model in the rat. Ann N Y Acad Sci 1991;640:290294.Google Scholar
Vernino, S, Geschwind, M, Boeve, B. Autoimmune encephalopathies. Neurologist 2007;13:140147.Google Scholar
Hébert, J, Gros, P, Lapointe, S, et al. Searching for autoimmune encephalitis: beware of normal CSF. J Neuroimmunol 2020;345:577285.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
McKeon, A. Autoimmune encephalopathies and dementias. Continuum (Minneapolis, Minn) 2016;22:538558.Google Scholar
Sechi, E, Flanagan, EP. Diagnosis and management of autoimmune dementia. Curr Treat Options Neurol 2019;21:11.CrossRefGoogle ScholarPubMed
Long, JM, Day, GS. Autoimmune dementia. Semin Neurol 2018;38:303315.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
Prüss, H, Holtje, M, Maier, N, et al. IgA NMDA receptor antibodies are markers of synaptic immunity in slow cognitive impairment. Neurology 2012;78:17431753.CrossRefGoogle ScholarPubMed
Doss, S, Wandinger, KP, Hyman, BT, et al. High prevalence of NMDA receptor IgA/IgM antibodies in different dementia types. Ann Clin Transl Neurol 2014;1:822832.CrossRefGoogle ScholarPubMed
Hara, M, Martinez-Hernandez, E, Ariño, H, et al. Clinical and pathogenic significance of IgG, IgA, and IgM antibodies against the NMDA receptor. Neurology 2018;90:e1386e1394.Google Scholar
Steiner, J, Teegen, B, Schiltz, K, et al. Prevalence of N-methyl-D-aspartate receptor autoantibodies in the peripheral blood: healthy control samples revisited. JAMA Psychiatry 2014;71:838839.Google Scholar
Sperber, PS, Siegerink, B, Huo, S, et al. Serum anti-NMDA (N-methyl-D-aspartate)-receptor antibodies and long-term clinical outcome after stroke (PROSCIS-B). Stroke 2019;50:32133219.Google Scholar
Geschwind, MD, Shu, H, Haman, A, Sejvar, JJ, Miller, BL. Rapidly progressive dementia. Ann Neurol 2008;64:97108.Google Scholar
Geschwind, MD. Rapidly progressive dementia. Continuum (Minneapolis, Minn) 2016;22:510537.Google ScholarPubMed
Grau-Rivera, O, Gelpi, E, Nos, C, et al. Clinicopathological correlations and concomitant pathologies in rapidly progressive dementia: a brain bank series. Neuro-degener Dis 2015;15:350360.Google Scholar
Chitravas, N, Jung, RS, Kofskey, DM, et al. Treatable neurological disorders misdiagnosed as Creutzfeldt–Jakob disease. Ann Neurol 2011;70:437444.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
Anuja, P, Venugopalan, V, Darakhshan, N, et al. Rapidly progressive dementia: an eight year (2008–2016) retrospective study. PLoS One 2018;13:e0189832.Google Scholar
Studart Neto, A, Soares Neto, HR, Simabukuro, MM, et al. Rapidly progressive dementia: prevalence and causes in a neurologic unit of a tertiary hospital in Brazil. Alzheimer Dis Assoc Disord 2017;31:239243.CrossRefGoogle Scholar
Sala, I, Marquié, M, Sánchez-Saudinós, MB, et al. Rapidly progressive dementia: experience in a tertiary care medical center. Alzheimer Dis Assoc Disord 2012;26:267271.Google Scholar
Papageorgiou, SG, Kontaxis, T, Bonakis, A, et al. Rapidly progressive dementia: causes found in a Greek tertiary referral center in Athens. Alzheimer Dis Assoc Disord 2009;23:337346.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
Irani, SR, Michell, AW, Lang, B, et al. Faciobrachial dystonic seizures precede Lgi1 antibody limbic encephalitis. Ann Neurol 2011;69:892900.Google Scholar
Gaig, C, Graus, F, Compta, Y, et al. Clinical manifestations of the anti-IgLON5 disease. Neurology 2017;88:17361743.Google Scholar
Saito, Y, Ruberu, NN, Sawabe, M, et al. Staging of argyrophilic grains: an age-associated tauopathy. J Neuropathol Exp Neurol 2004;63:911918.Google Scholar
Zarow, C, Sitzer, TE, Chui, HC. Understanding hippocampal sclerosis in the elderly: epidemiology, characterization, and diagnostic issues. Curr Neurol Neurosci Rep 2008;8:363370.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
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
Knopman, DS, Parisi, JE, Salviati, A, et al. Neuropathology of cognitively normal elderly. J Neuropathol Exp Neurol 2003;62:10871095.Google Scholar
Beach, TG, Sue, L, Scott, S, et al. Hippocampal sclerosis dementia with tauopathy. Brain Pathol 2003;13:263278.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
Geschwind, MD, Tan, KM, Lennon, VA, et al. Voltage-gated potassium channel autoimmunity mimicking Creutzfeldt-Jakob disease. Arch Neurol 2008;65:13411346.CrossRefGoogle ScholarPubMed
Geschwind, MD, Martindale, J, Miller, D, et al. Challenging the clinical utility of the 14-3-3 protein for the diagnosis of sporadic Creutzfeldt–Jakob disease. Arch Neurol 2003;60:813816.Google Scholar
Tartaglia, MC, Johnson, DY, Thai, JN, et al. Clinical overlap between Jakob–Creutzfeldt disease and Lewy body disease. Can J Neurol Sci 2012;39:304310.Google Scholar
Candelise, N, Baiardi, S, Franceschini, A, Rossi, M, Parchi, P. Towards an improved early diagnosis of neurodegenerative diseases: the emerging role of in vitro conversion assays for protein amyloids. Acta Neuropathologica Commun 2020;8:117.CrossRefGoogle ScholarPubMed
Tan, KM, Lennon, VA, Klein, CJ, Boeve, BF, Pittock, SJ. Clinical spectrum of voltage-gated potassium channel autoimmunity. Neurology 2008;70:18831890.Google Scholar
Rossi, M, Mead, S, Collinge, J, Rudge, P, Vincent, A. Neuronal antibodies in patients with suspected or confirmed sporadic Creutzfeldt–Jakob disease. J Neurol Neurosurg Psychiatry 2015;86:692694.Google Scholar
Fujita, K, Yuasa, T, Watanabe, O, et al. Voltage-gated potassium channel complex antibodies in Creutzfeldt–Jakob disease. J Neurol 2012;259:22492250.CrossRefGoogle ScholarPubMed
Newey, CR, Appleby, BS, Shook, S, Sarwal, A. Patient with voltage-gated potassium-channel (VGKC) limbic encephalitis found to have Creutzfeldt–Jakob disease (CJD) at autopsy. J Neuropsychiatry Clin Neurosci 2013;25:E05E07.CrossRefGoogle ScholarPubMed
Jammoul, A, Lederman, RJ, Tavee, J, Li, Y. Presence of voltage-gated potassium channel complex antibody in a case of genetic prion disease. BMJ Case Rep 2014;2014:bcr2013201622.Google Scholar
Jones, M, Odunsi, S, du Plessis, D, et al. Gerstmann–Straüssler–Scheinker disease: novel PRNP mutation and VGKC-complex antibodies. Neurology 2014;82:21072111.Google Scholar
Angus-Leppan, H, Rudge, P, Mead, S, Collinge, J, Vincent, A. Autoantibodies in sporadic Creutzfeldt–Jakob disease. JAMA Neurol 2013;70:919922.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.CrossRefGoogle ScholarPubMed
Mackay, G, Ahmad, K, Stone, J, et al. NMDA receptor autoantibodies in sporadic Creutzfeldt–Jakob disease. J Neurol 2012;259:19791981.Google Scholar
Fujita, K, Yuasa, T, Takahashi, Y, et al. Antibodies to N-methyl-D-aspartate glutamate receptors in Creutzfeldt–Jakob disease patients. J Neuroimmunol 2012;251:9093.Google Scholar
Zuhorn, F, Hübenthal, A, Rogalewski, A, et al. Creutzfeldt–Jakob disease mimicking autoimmune encephalitis with CASPR2 antibodies. BMC Neurol 2014;14:227.Google Scholar
Grau-Rivera, O, Sanchez-Valle, R, Saiz, A, et al. Determination of neuronal antibodies in suspected and definite Creutzfeldt–Jakob disease. JAMA Neurol 2014;71:7478.Google Scholar
Cavallieri, F, Mandrioli, J, Tondelli, M, et al. Pearls & Oy-sters: rapidly progressive dementia: prions or immunomediated? Neurology 2014;82:e149152.Google Scholar
McKeon, A, Marnane, M, O’Connell, M, et al. Potassium channel antibody associated encephalopathy presenting with a frontotemporal dementia like syndrome. Arch Neurol 2007;64:15281530.CrossRefGoogle ScholarPubMed
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
Uttley, L, Carroll, C, Wong, R, Hilton, DA, Stevenson, M. Creutzfeldt–Jakob disease: a systematic review of global incidence, prevalence, infectivity, and incubation. Lancet Infect Dis 2020;20:e2e10.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
Marquetand, J, van Lessen, M, Bender, B, et al. Slowly progressive LGI1 encephalitis with isolated late-onset cognitive dysfunction: a treatable mimic of Alzheimer’s disease. Eur J Neurol 2016;23:e28e29.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
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
Yonelinas, AP. The hippocampus supports high-resolution binding in the service of perception, working memory and long-term memory. Behav Brain Res 2013;254:3444.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
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
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
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.CrossRefGoogle ScholarPubMed
Loane, C, Argyropoulos, GPD, Roca-Fernandez, A, et al. Hippocampal network abnormalities explain amnesia after VGKCC-Ab related autoimmune limbic encephalitis. J Neurol Neurosurg Psychiatry 2019;90:965974.Google Scholar
Griffith, SP, Malpas, CB, Alpitsis, R, O’Brien, TJ, Monif, M. The neuropsychological spectrum of anti-LGI1 antibody mediated autoimmune encephalitis. J Neuroimmunol 2020;345:577271.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
Knopman, DS, DeKosky, ST, Cummings, JL, et al. Practice parameter: diagnosis of dementia (an evidence-based review). Report of the Quality Standards Subcommittee of the American Academy of Neurology. Neurology 2001;56:11431153.CrossRefGoogle Scholar
Miller, TD, Chong, TT, Aimola Davies, AM, et al. Human hippocampal CA3 damage disrupts both recent and remote episodic memories. Elife 2020;9.Google Scholar
Stern, Y. Cognitive reserve in ageing and Alzheimer’s disease. Lancet Neurol 2012;11:10061012.Google Scholar
Stern, Y, Arenaza-Urquijo, EM, Bartres-Faz, D, et al. Whitepaper: defining and investigating cognitive reserve, brain reserve, and brain maintenance. Alzheimers Dement 2020;16:13051311.CrossRefGoogle ScholarPubMed
Matyas, N, Keser Aschenberger, F, Wagner, G, et al. Continuing education for the prevention of mild cognitive impairment and Alzheimer’s-type dementia: a systematic review and overview of systematic reviews. BMJ Open 2019;9:e027719.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
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
Gelpi, E, Hoftberger, R, Graus, F, et al. Neuropathological criteria of anti-IgLON5-related tauopathy. Acta Neuropathol 2016;132:531543.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
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
Gaig, C, Compta, Y. Neurological profiles beyond the sleep disorder in patients with anti-IgLON5 disease. Curr Opin Neurol 2019;32:493499.CrossRefGoogle ScholarPubMed
Hansen, N, Hirschel, S, Stöcker, W, et al. Figural memory impairment in conjunction with neuropsychiatric symptoms in IgLON5 antibody-associated autoimmune encephalitis. Front Psychiatry 2020;11:576.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.CrossRefGoogle ScholarPubMed
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
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
Gaig, C, Iranzo, A, Cajochen, C, et al. Characterization of the sleep disorder of anti-IgLON5 disease. Sleep 2019;42:zsz133.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.CrossRefGoogle ScholarPubMed
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
Zhou, Q, Zhu, X, Meng, H, Zhang, M, Chen, S. Anti-dipeptidyl-peptidase-like protein 6 encephalitis, a rare cause of reversible rapid progressive dementia and insomnia. J Neuroimmunol 2020;339:577114.CrossRefGoogle ScholarPubMed
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
Titulaer, MJ, McCracken, L, Gabilondo, I, et al. Late-onset anti-NMDA receptor encephalitis. Neurology 2013;81:10581063.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.Google Scholar
Ricken, G, Zrzavy, T, Macher, S, et al. Autoimmune global amnesia as manifestation of AMPAR encephalitis and neuropathologic findings. Neurol Neuroimmunol Neuroinflamm 2021;8:e1019Google Scholar
Danve, A, Grafe, M, Deodhar, A. Amyloid beta-related angiitis: a case report and comprehensive review of literature of 94 cases. Semin Arthrit Rheumat 2014;44:8692.Google Scholar
Schielke, E, Nolte, C, Müller, W, Brück, W. Sarcoidosis presenting as rapidly progressive dementia: clinical and neuropathological evaluation. J Neurol 2001;248:522524.Google Scholar
De Maindreville, A, Bedos, L, Bakchine, S. Systemic sarcoidosis mimicking a behavioural variant of frontotemporal dementia. Cae Rep Neurol Med 2015;2015:409126.Google Scholar
Gómez-Puerta, JA, Cervera, R, Calvo, LM, et al. Dementia associated with the antiphospholipid syndrome: clinical and radiological characteristics of 30 patients. Rheumatology (Oxford, England) 2005;44:9599.CrossRefGoogle ScholarPubMed
Abraham, P, Neel, I, Bishay, S, Sewell, DD. Central nervous system systemic lupus erythematosus (CNS-SLE) vasculitis mimicking Lewy body dementia: a case report emphasizing the role of imaging with an analysis of 33 comparable cases from the scientific literature. J Geriatr Psychiatry Neurol 2021;34:128141.Google Scholar
Seipelt, M, Zerr, I, Nau, R, et al. Hashimoto’s encephalitis as a differential diagnosis of Creutzfeldt–Jakob disease. J Neurol Neurosurg Psychiatry 1999;66:172176.Google Scholar
Mattozzi, S, Sabater, L, Escudero, D, et al. Hashimoto encephalopathy in the 21st century. Neurology 2020;94:e217e224.CrossRefGoogle Scholar
Chang, T, Riffsy, MT, Gunaratne, PS. Hashimoto encephalopathy: clinical and MRI improvement following high-dose corticosteroid therapy. Neurologist 2010;16:394396.Google Scholar
Castillo, P, Woodruff, B, Caselli, R, et al. Steroid-responsive encephalopathy associated with autoimmune thyroiditis. Arch Neurol 2006;63:197202.Google Scholar
Slooter, AJC, Otte, WM, Devlin, JW, et al. Updated nomenclature of delirium and acute encephalopathy: statement of ten Societies. Intensive Care Med 2020;46:10201022.Google Scholar
Oldham, MA, Holloway, RG. Delirium disorder: integrating delirium and acute encephalopathy. Neurology 2020;95:173178.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
×