Skip to main content Accessibility help
×
Hostname: page-component-77c89778f8-fv566 Total loading time: 0 Render date: 2024-07-20T21:25:07.776Z Has data issue: false hasContentIssue false

Chapter 5 - Antibodies to Neural Cell Surface Antigens

from Section 2 - Antibodies and Antigens

Published online by Cambridge University Press:  27 January 2022

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

Summary

Antibodies against cell surface antigens (neuronal or glial) usually recognize conformational epitopes. Therefore, these antibodies can only be detected by techniques that preserve the native conformation of the target antigen, for instance, indirect immunofluorescence with tissue adapted to cell surface or synaptic proteins, cultured neurons, cells transfected with the antigen, or radioimmunoassay. Studies using in-vitro models or passive transfer of antibodies to experimental animals strongly suggest that these antibodies are pathogenic. Some antibodies frequently occur in encephalitis associated with cancer (for example, anti-GABAbR antibodies and small-cell lung cancer), but the incidence of cancer is usually lower than that observed in patients with onconeural antibodies. Two antibodies against surface antigens, DNER (Tr) and amphiphysin, are exceptions to the characteristics mentioned above as they recognize linear epitopes and are considered biomarkers of paraneoplastic neurological syndromes associated with Hodgkin lymphoma (DNER) or small-cell lung cancer (amphiphysin). Antibodies against AMPAR, GABAbR, and LGI1 usually associate with limbic encephalitis. Other antibodies characteristically associate with distinct syndromes, for example, anti-NMDAR encephalitis that manifests with a highly characteristic neuropsychiatric syndrome; CASPR2 antibodies that can manifest as Morvan syndrome; aquaporin 4 antibodies that typically associate with neuromyelitis optica spectrum disorders, and glycine receptor antibodies that predominantly associate with progressive encephalomyelitis with rigidity and myoclonus (a disorder in the spectrum of stiff-person syndrome). Antibodies against GABAaR, MOG, and GluK2 subunit of the kainate receptor occur with different types of encephalitis characterized by multiple FLAIR/T2 hyperintense lesion in the brain MRI.

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

Gultekin, HS, Rosenfeld, MR, Voltz, RD, et al. Paraneoplastic limbic encephalitis: neurological symptoms, immunological findings and tumor association in 50 patients. Brain 2000;123:14811494Google Scholar
Bien, CG, Schulze-Bonhage, A, Deckert, M, et al. Limbic encephalitis not associated with neoplasm as a cause of temporal lobe epilepsy. Neurology 2000;55:18231828.CrossRefGoogle Scholar
Watanabe, Y, Shimizu, Y, Ooi, S, et al. Steroid-responsive limbic encephalitis. Intern Med 2003;42:428432.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
Dalmau, J, Geis, C, Graus, F. Autoantibodies to synaptic receptors and neuronal cell surface proteins in autoimmune diseases of the central nervous system. Physiol Rev 2017;97:839887.CrossRefGoogle ScholarPubMed
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
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.CrossRefGoogle ScholarPubMed
Dalmau, J, Graus, F. Antibody-mediated encephalitis. N Engl J Med 2018;378:840851.Google Scholar
Cutillo, G, Saariaho, AH, Meri, S. Physiology of gangliosides and the role of antiganglioside antibodies in human diseases. Cell Mol Immunol 2020;17:313322.Google Scholar
Reiner, A, Levitz, J. Glutamatergic signaling in the central nervous system: ionotropic and metabotropic receptors in concert. Neuron 2018;98:10801098.Google Scholar
Vitaliani, R, Mason, W, Ances, B, et al. Paraneoplastic encephalitis, psychiatric symptoms, and hypoventilation in ovarian teratoma. Ann Neurol 2005;58:594604.CrossRefGoogle ScholarPubMed
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
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.CrossRefGoogle ScholarPubMed
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
Titulaer, MJ, McCracken, L, Gabilondo, I, et al. Late-onset anti-NMDA receptor encephalitis. Neurology 2013;81:10581063.Google Scholar
Gleichman, AJ, Spruce, LA, Dalmau, J, Seeholzer, SH, Lynch, DR. Anti-NMDA receptor encephalitis antibody binding is dependent on amino acid identity of a small region within the GluN1 amino terminal domain. J Neurosci 2012;32:1108211094.CrossRefGoogle ScholarPubMed
Hughes, EG, Peng, X, Gleichman, AJ, et al. Cellular and synaptic mechanisms of anti-NMDA receptor encephalitis. J Neurosci 2010;30:58665875.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.CrossRefGoogle ScholarPubMed
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
Moscato, EH, Peng, X, Jain, A, et al. Acute mechanisms underlying antibody effects in anti-N-methyl-D-aspartate receptor encephalitis. Ann Neurol 2014;76:108119.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
Carceles-Cordon, M, Mannara, F, Aguilar, E, et al. NMDAR antibodies alter dopamine receptors and cause psychotic behavior in mice. Ann Neurol 2020;88:603613.Google Scholar
Grea, H, Bouchet, D, Rogemond, V, et al. Human autoantibodies against N-methyl-D-aspartate receptor modestly alter dopamine D1 receptor surface dynamics. Front Psychiatry 2019;10:670.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
Graus, F, Boronat, A, Xifro, X, et al. The expanding clinical profile of anti-AMPA receptor encephalitis. Neurology 2010;74:857859.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
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
Bataller, L, Galiano, R, Garcia-Escrig, M, et al. Reversible paraneoplastic limbic encephalitis associated with antibodies to the AMPA receptor. Neurology 2010;74:265267.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.CrossRefGoogle ScholarPubMed
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
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
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
Landa, J, Guasp, M, Míguez-Cabello, F, et al. Encephalitis with autoantibodies against the glutamate kainate receptors GluK2. Ann Neurol 2021;90:101117.CrossRefGoogle ScholarPubMed
Contractor, A, Mulle, C, Swanson, GT. Kainate receptors coming of age: milestones of two decades of research. Trends Neurosci 2011;34:154163.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
Spatola, M, Petit-Pedrol, M, Simabukuro, MM, et al. Investigations in GABAA receptor antibody-associated encephalitis. Neurology 2017;88:10121020.Google Scholar
Caputo, D, Iorio, R, Vigevano, F, Fusco, L. Febrile infection-related epilepsy syndrome (FIRES) with super-refractory status epilepticus revealing autoimmune encephalitis due to GABAAR antibodies. Eur J Paediatr Neurol 2018;22:182185.Google Scholar
Nikolaus, M, Knierim, E, Meisel, C, et al. Severe GABAA receptor encephalitis without seizures: a paediatric case successfully treated with early immunomodulation. Eur J Paediatr Neurol 2018;22:558562.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
Armangue, T, Moris, G, Cantarin-Extremera, V, et al. Autoimmune post-herpes simplex encephalitis of adults and teenagers. Neurology 2015;85:17361743.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
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
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
Samra, K, Rogers, J, Mahdi-Rogers, M, Stanton, B. Catatonia with GABAA receptor antibodies. Pract Neurol 2020;20:139143.CrossRefGoogle 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
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
Jarius, S, Steinmeyer, F, Knobel, A, et al. GABAB receptor antibodies in paraneoplastic cerebellar ataxia. J Neuroimmunol 2013;256:9496.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
Jeffery, OJ, Lennon, VA, Pittock, SJ, et al. GABAB receptor autoantibody frequency in service serologic evaluation. Neurology 2013;81:882887.Google Scholar
Ohta, K, Seki, M, Dalmau, J, Shinohara, Y. Perfusion IMP-SPECT shows reversible abnormalities in GABA(B) receptor antibody associated encephalitis with normal MRI. Brain Behav 2011;1:7072.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
Jain, A, Lancaster, E, Dalmau, J, Balice-Gordon, RJ. Autoantibodies in the CSF of anti-GABA receptor encephalitis patients block activation of GABA receptors in vitro. Ann Neurol 2015;78:S77.Google Scholar
Nibber, A, Mann, EO, Pettingill, P, et al. Pathogenic potential of antibodies to the GABAB receptor. Epilepsia Open 2017;2:355359.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
Ferraguti, F, Shigemoto, R. Metabotropic glutamate receptors. Cell Tissue Res 2006;326:483504.CrossRefGoogle ScholarPubMed
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
Spatola, M, Petit-Pedrol, M, Maudes, E, et al. Clinical features, prognostic factors, and antibody effects in anti-mGluR1 encephalitis. Neurology, 2020;95:e3012e3025.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
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
Gollion, C, Dupouy, J, Roberts, M, et al. Reversible myoclonus-ataxia encephalitis related to anti-mGLUR1 autoantibodies. Mov Disord 2019;34:438439.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
Ichise, T, Kano, M, Hashimoto, K, et al. mGluR1 in cerebellar Purkinje cells essential for long-term depression, synapse elimination, and motor coordination. Science 2000;288:18321835.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
Ruiz-Garcia, R, Martinez-Hernandez, E, Joubert, B, et al. Paraneoplastic cerebellar ataxia and antibodies to metabotropic glutamate receptor 2. Neurol Neuroimmunol Neuroinflamm 2019;7:e658.CrossRefGoogle ScholarPubMed
Carr, I. The Ophelia syndrome: memory loss in Hodgkin’s disease. Lancet 1982;1:844845.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
Mat, A, Adler, H, Merwick, A, et al. Ophelia syndrome with metabotropic glutamate receptor 5 antibodies in CSF. Neurology 2013;80:13491350.Google Scholar
Pruss, H, Rothkirch, M, Kopp, U, et al. Limbic encephalitis with mGluR5 antibodies and immunotherapy-responsive prosopagnosia. Neurology 2014;83:13841386.Google Scholar
Spatola, M, Sabater, L, Planaguma, J, et al. Encephalitis with mGluR5 antibodies: Symptoms and antibody effects. Neurology 2018;90:e1964e1972.Google Scholar
Guevara, C, Farias, G, Silva-Rosas, C, et al. Encephalitis associated to metabotropic glutamate receptor 5 (mGluR5) antibodies in cerebrospinal fluid. Front Immunol 2018;9:2568.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
Brimberg, L, Benhar, I, Mascaro-Blanco, A, et al. Behavioral, pharmacological, and immunological abnormalities after Streptococcal exposure: a novel rat model of Sydenham chorea and related neuropsychiatric disorders. Neuropsychopharmacology 2012;37:20762087.Google Scholar
Marques-Matos, C, Melo, C, Sampaio, M, et al. Child neurology: treatable bilateral striatal lesions related to anti-dopamine 2 receptor autoimmunity. Neurology 2018;91:98101.Google Scholar
Pathmanandavel, K, Starling, J, Merheb, V, et al. Antibodies to surface dopamine-2 receptor and N-methyl-D-aspartate receptor in the first episode of acute psychosis in children. Biol Psychiatry 2015;77:537547.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
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
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 Neuropathol Commun 2016;4:126.Google Scholar
Hutchinson, M, Waters, P, McHugh, J, et al. Progressive encephalomyelitis, rigidity, and myoclonus: a novel glycine receptor antibody. Neurology 2008;71:12911292.CrossRefGoogle ScholarPubMed
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
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
Martinez-Hernandez, E, Arino, H, McKeon, A, et al. Clinical and immunologic investigations in patients with stiff-person spectrum disorder. JAMA Neurol 2016;73:714720.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
Ekizoglu, E, Baykan, B, Sezgin, M, et al. Follow-up of patients with epilepsy harboring antiglycine receptor antibodies. Epilepsy Behav 2019;92:103107.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
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
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
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
Crisp, SJ, Dixon, CL, Jacobson, L, et al. Glycine receptor autoantibodies disrupt inhibitory neurotransmission. Brain 2019;142:33983410.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
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
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
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
Irani, SR, Michell, AW, Lang, B, et al. Faciobrachial dystonic seizures precede Lgi1 antibody limbic encephalitis. Ann Neurol 2011;69:892900.CrossRefGoogle ScholarPubMed
Andrade, DM, Tai, P, Dalmau, J, Wennberg, R. Tonic seizures: a diagnostic clue of anti-LGI1 encephalitis? Neurology 2011;76:13551357.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
Kim, TJ, Lee, ST, Moon, J, et al. Anti-LGI1 encephalitis is associated with unique HLA subtypes. Ann Neurol 2017;81:183192.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
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
Irani, SR, Pettingill, P, Kleopa, KA, et al. Morvan syndrome: clinical and serological observations in 29 cases. Ann Neurol 2012;72:241255.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
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
Ramberger, M, Berretta, A, Tan, JMM, et al. Distinctive binding properties of human monoclonal LGI1 autoantibodies determine pathogenic mechanisms. Brain 2020;143:17311745.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
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
Binks, S, Varley, J, Lee, W, et al. Distinct HLA associations of LGI1 and CASPR2-antibody diseases. Brain 2018;141:22632271.Google Scholar
Muniz-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
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
Syrbe, S, Stettner, GM, Bally, J, et al. CASPR2 autoimmunity in children expanding to mild encephalopathy with hypertension. Neurology 2020;94:e2290e2301.Google Scholar
Olsen, AL, Lai, Y, Dalmau, J, Scherer, SS, Lancaster, E. Caspr2 autoantibodies target multiple epitopes. Neurol Neuroimmunol Neuroinflamm 2015;2:e127.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
Liang, W, Zhang, J, Saint-Martin, M, et al. Structural mapping of hot spots within human CASPR2 discoidin domain for autoantibody recognition. J Autoimmun 2019;96:168177.Google Scholar
Patterson, KR, Dalmau, J, Lancaster, E. Mechanisms of Caspr2 antibodies in autoimmune encephalitis and neuromyotonia. Ann Neurol 2018;83:4051.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
Balint, B, Jarius, S, Nagel, S, et al. Progressive encephalomyelitis with rigidity and myoclonus: a new variant with DPPX antibodies. Neurology 2014;82:15211528.CrossRefGoogle ScholarPubMed
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
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.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
Stoeck, K, Carstens, PO, Jarius, S, et al. Prednisolone and azathioprine are effective in DPPX antibody-positive autoimmune encephalitis. Neurol Neuroimmunol Neuroinflamm 2015;2:e86.Google Scholar
Zagha, E, Ozaita, A, Chang, SY, et al. DPP10 modulates Kv4-mediated A-type potassium channels. J Biol Chem 2005;280:1885318861.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
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
Koh, SJ, Ang, CH, Tham, HLC, Chua, HC. Refractory status epilepticus secondary to neurexin-3α encephalitis: a case report. Neurology Asia 2018;23:273277.Google Scholar
Costa, A, Silva-Pinto, A, Alves, J, et al. Postmalaria neurologic syndrome associated with neurexin-3alpha antibodies. Neurol Neuroimmunol Neuroinflamm 2017;4:e392.Google Scholar
Loehrer, PA, Bien, CI, Dusoi, AE, Timmermann, L, Simon, OJ. Neurexin-3alpha associated autoimmune encephalitis: full recovery after rituximab therapy – a case report. Eur J Neurol 2020;27:e91e93.Google Scholar
Sudhof, TC. Neuroligins and neurexins link synaptic function to cognitive disease. Nature 2008;455:903911.Google Scholar
Siddiqui, TJ, Pancaroglu, R, Kang, Y, Rooyakkers, A, Craig, AM. LRRTMs and neuroligins bind neurexins with a differential code to cooperate in glutamate synapse development. J Neurosci 2010;30:74957506.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
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.CrossRefGoogle ScholarPubMed
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
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
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
Gelpi, E, Hoftberger, R, Graus, F, et al. Neuropathological criteria of anti-IgLON5-related tauopathy. Acta Neuropathol 2016;132:531543.Google Scholar
Landa, J, Gaig, C, Planaguma, J, et al. Effects of IgLON5 antibodies on neuronal cytoskeleton: a link between autoimmunity and neurodegeneration. Ann Neurol 2020;88:10231027.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
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
Tan, RPA, Leshchyns’ka, I, Sytnyk, V. Glycosylphosphatidylinositol-anchored immunoglobulin superfamily cell adhesion molecules and their role in neuronal development and synapse regulation. Front Mol Neurosci 2017;10:378.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
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
Miyazaki, T, Hashimoto, K, Uda, A, et al. Disturbance of cerebellar synaptic maturation in mutant mice lacking BSRPs, a novel brain-specific receptor-like protein family. FEBS Lett 2006;580:40574064.Google Scholar
Nash, A, Aumann, TD, Pigoni, M, et al. Lack of Sez6 family proteins impairs motor functions, short-term memory, and cognitive flexibility and alters dendritic spine properties. Cereb Cortex 2020;30:21672184.Google Scholar
Landa, J, Guasp, M, M P-P, et al. Seizure-related 6 homolog like 2 (SEZ6L2) autoimmunity: neurologic syndrome and antibody effects. Neurol Neuroimmunol Neuroinflamm 2021;8:e916.Google Scholar
Yaguchi, H, Yabe, I, Takahashi, H, et al. Anti-Sez6l2 antibody detected in a patient with immune-mediated cerebellar ataxia inhibits complex formation of GluR1 and Sez6l2. J Neurol 2018;265:962965.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
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
Trotter, JL, Hendin, BA, Osterland, CK. Cerebellar degeneration with Hodgkin disease: an immunological study. Arch Neurol 1976;33:660661.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.CrossRefGoogle ScholarPubMed
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
Tohgo, A, Eiraku, M, Miyazaki, T, et al. Impaired cerebellar functions in mutant mice lacking DNER. Mol Cell Neurosci 2006;31:326333.Google Scholar
Saito, SY, Takeshima, H. DNER as key molecule for cerebellar maturation. Cerebellum 2006;5:227231.Google Scholar
Fukuoka, T, Engel, AG, Lang, B, Newsom-Davis, J, Vincent, A. Lambert–Eaton myasthenic syndrome: II. Immunoelectron microscopy localization of IgG at the mouse motor end-plate. Ann Neurol 1987;22:200211.Google Scholar
Fukuoka, T, Engel, AG, Lang, B, et al. Lambert–Eaton myasthenic syndrome: I. Early morphological effects of IgG on the presynaptic membrane active zones. Ann Neurol 1987;22:193199.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.CrossRefGoogle ScholarPubMed
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
Graus, F, Lang, B, Pozo-Rosich, P, Saiz, A, Casamitjana, R, Vincent, A. P/Q type calcium-channel antibodies in paraneoplastic cerebellar degeneration with lung cancer. Neurology 2002;59:764766.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
Peter, BJ, Kent, HM, Mills, IG, et al. BAR domains as sensors of membrane curvature: the amphiphysin BAR structure. Science 2004;303:495499.Google Scholar
David, C, McPherson, PS, Mundigl, O, De Camilli, P. A role of amphiphysin in synaptic vesicle endocytosis suggested by its binding to dynamin in nerve terminals. Proc Natl Acad Sci USA 1996;93:331335.Google Scholar
Ramjaun, AR, Micheva, KD, Bouchelet, I, McPherson, PS. Identification and characterization of a nerve terminal-enriched amphiphysin isoform. J Biol Chem 1997;272:1670016706.Google Scholar
David, C, Solimena, M, De Camilli, P. Autoimmunity in Stiff-Man syndrome with breast cancer is targeted to the C-terminal region of human amphiphysin, a protein similar to the yeast proteins, Rvs167 and Rvs161. FEBS Lett 1994;351:7379.Google Scholar
Yang, H, Wan, Z, Huang, C, Yin, H, Song, D. AMPH-1 is a tumor suppressor of lung cancer by inhibiting Ras-Raf-MEK-ERK signal pathway. Lasers Med Sci 2019;34:473478.Google Scholar
Chen, Y, Liu, J, Li, L, et al. AMPH-1 is critical for breast cancer progression. J Cancer 2018;9:21752182.Google Scholar
Prokic, I, Cowling, BS, Laporte, J. Amphiphysin 2 (BIN1) in physiology and diseases. J Mol Med (Berl) 2014;92:453463.CrossRefGoogle ScholarPubMed
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
Weinshenker, BG. Neuromyelitis optica: what it is and what it might be. Lancet 2003;361:889890.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
de Seze, J, Lebrun, C, Stojkovic, T, et al. Is Devic’s neuromyelitis optica a separate disease? A comparative study with multiple sclerosis. Mult Scler 2003;9:521525.Google Scholar
Mandler, RN, Ahmed, W, Dencoff, JE. Devic’s neuromyelitis optica: a prospective study of seven patients treated with prednisone and azathioprine. Neurology 1998;51:12191220.Google Scholar
Keegan, M, Pineda, AA, McClelland, RL, et al. Plasma exchange for severe attacks of CNS demyelination: predictors of response. Neurology 2002;58:143146.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
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
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
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
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
Nagelhus, EA, Ottersen, OP. Physiological roles of aquaporin-4 in brain. Physiol Rev 2013;93:15431562.Google Scholar
Wingerchuk, DM, Lennon, VA, Pittock, SJ, Lucchinetti, CF, Weinshenker, BG. Revised diagnostic criteria for neuromyelitis optica. Neurology 2006;66:14851489.Google Scholar
Saiz, A, Zuliani, L, Blanco, Y, et al. Revised diagnostic criteria for neuromyelitis optica (NMO). Application in a series of suspected patients. J Neurol 2007;254:12331237.Google Scholar
Dinkin, MJ, Cestari, DM, Stein, MC, Brass, SD, Lessell, S. NMO antibody-positive recurrent optic neuritis without clear evidence of transverse myelitis. Arch Ophthalmol 2008;126:566570.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
Wingerchuk, DM, Lennon, VA, Lucchinetti, CF, Pittock, SJ, Weinshenker, BG. The spectrum of neuromyelitis optica. Lancet Neurol 2007;6:805815.Google Scholar
Waters, PJ, McKeon, A, Leite, MI, et al. Serologic diagnosis of NMO: a multicenter comparison of aquaporin-4-IgG assays. Neurology 2012;78:665671.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, 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
Mader, S, Gredler, V, Schanda, K, et al. Complement activating antibodies to myelin oligodendrocyte glycoprotein in neuromyelitis optica and related disorders. J Neuroinflammation 2011;8:184.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
Tenembaum, S, Chitnis, T, Nakashima, I, et al. Neuromyelitis optica spectrum disorders in children and adolescents. Neurology 2016;87:S5966.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
Tenembaum, S, Yeh, EA, Guthy-Jackson Foundation International Clinical Consortium. Pediatric NMOSD: a review and position statement on approach to work-up and diagnosis. Front Pediatr 2020;8:339.Google Scholar
Saadoun, S, Waters, P, Bell, BA, et al. Intra-cerebral injection of neuromyelitis optica immunoglobulin G and human complement produces neuromyelitis optica lesions in mice. Brain 2010;133:349361.Google Scholar
Hinson, SR, Pittock, SJ, Lucchinetti, CF, et al. Pathogenic potential of IgG binding to water channel extracellular domain in neuromyelitis optica. Neurology 2007;69:22212231.CrossRefGoogle ScholarPubMed
Vincent, T, Saikali, P, Cayrol, R, et al. Functional consequences of neuromyelitis optica-IgG astrocyte interactions on blood–brain barrier permeability and granulocyte recruitment. J Immunol 2008;181:57305737.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
Asavapanumas, N, Ratelade, J, Papadopoulos, MC, et al. Experimental mouse model of optic neuritis with inflammatory demyelination produced by passive transfer of neuromyelitis optica-immunoglobulin G. J Neuroinflammation 2014;11:16.Google Scholar
Peschl, P, Bradl, M, Hoftberger, R, Berger, T, Reindl, M. Myelin oligodendrocyte glycoprotein: deciphering a target in inflammatory demyelinating diseases. Front Immunol 2017;8:529.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
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
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
Kitley, J, Woodhall, M, Waters, P, et al. Myelin-oligodendrocyte glycoprotein antibodies in adults with a neuromyelitis optica phenotype. Neurology 2012;79:12731277.Google Scholar
Lechner, C, Baumann, M, Hennes, EM, et al. Antibodies to MOG and AQP4 in children with neuromyelitis optica and limited forms of the disease. J Neurol Neurosurg Psychiatry 2016;87:897905.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
Jarius, S, Metz, I, Konig, FB, et al. Screening for MOG-IgG and 27 other anti-glial and anti-neuronal autoantibodies in ‘pattern II multiple sclerosis’ and brain biopsy findings in a MOG-IgG-positive case. Mult Scler 2016;22:15411549.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
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
Pham-Dinh, D, Mattei, MG, Nussbaum, JL, et al. Myelin/oligodendrocyte glycoprotein is a member of a subset of the immunoglobulin superfamily encoded within the major histocompatibility complex. Proc Natl Acad Sci USA 1993;90:79907994.Google Scholar
von Budingen, HC, Mei, F, Greenfield, A, et al. The myelin oligodendrocyte glycoprotein directly binds nerve growth factor to modulate central axon circuitry. J Cell Biol 2015;210:891898.Google Scholar
Tea, F, Lopez, JA, Ramanathan, S, et al. Characterization of the human myelin oligodendrocyte glycoprotein antibody response in demyelination. Acta Neuropathol Commun 2019;7:145.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
Jarius, S, Paul, F, Aktas, O, et al. MOG encephalomyelitis: international recommendations on diagnosis and antibody testing. J Neuroinflammation 2018;15:134.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
Storch, MK, Stefferl, A, Brehm, U, et al. Autoimmunity to myelin oligodendrocyte glycoprotein in rats mimics the spectrum of multiple sclerosis pathology. Brain Pathol 1998;8:681694.Google Scholar
Mayer, MC, Meinl, E. Glycoproteins as targets of autoantibodies in CNS inflammation: MOG and more. Ther Adv Neurol Disord 2012;5:147159.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

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
×