Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-m9kch Total loading time: 0 Render date: 2024-05-12T05:55:08.727Z Has data issue: false hasContentIssue false

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

Access options

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

References

References

Patrick, J, Lindstrom, J. Autoimmune response to acetylcholine receptor. Science 1973;180:871872.Google Scholar
Lindstrom, JM, Lambert, EH. Content of acetylcholine receptor and antibodies bound to receptor in myasthenia gravis, experimental autoimmune myasthenia gravis, and Eaton–Lambert syndrome. Neurology 1978;28:130138.Google Scholar
Motomura, M, Johnston, I, Lang, B, Newsom-Davis, J. Anticalcium channel antibodies detected using Conus magus toxin in Lambert–Eaton myasthenic syndrome sera. Ann Neurol 1994;36:324.Google Scholar
Shillito, P, Molenaar, PC, Vincent, A, et al. Acquired neuromyotonia: evidence for autoantibodies directed against K+ channels of peripheral nerves. Ann Neurol 1995;38:714722.Google Scholar
Vernino, S, Adamski, J, Kryzer, TJ, Fealey, RD, Lennon, VA. Neuronal nicotinic ACh receptor antibody in subacute autonomic neuropathy and cancer-related syndromes. Neurology 1998;50:18061813.Google Scholar
Greenlee, JE, Brashear, HR. Antibodies to cerebellar Purkinje cells in patients with paraneoplastic cerebellar degeneration and ovarian carcinoma. Ann Neurol 1983;14:609613.Google Scholar
Graus, F, Cordon-Cardo, C, Posner, JB. Neuronal antinuclear antibody in sensory neuronopathy from lung cancer. Neurology 1985;35:538543.Google Scholar
Budde-Steffen, C, Anderson, NE, Rosenblum, MK, et al. An antineuronal autoantibody in paraneoplastic opsoclonus. Ann Neurol 1988;23:528531.Google Scholar
Darnell, RB. Onconeural antigens and the paraneoplastic neurologic disorders: at the intersection of cancer, immunity, and the brain. Proc Natl Acad Sci USA 1996;93:45294536.Google Scholar
Graus, F, Cordon-Cardo, C, Posner, JB. Neuronal antinuclear antibody in sensory neuronopathy from lung cancer. Neurology 1985;35:538543.Google Scholar
Budde-Steffen, C, Anderson, NE, Rosenblum, MK, et al. An antineuronal autoantibody in paraneoplastic opsoclonus. Ann Neurol 1988;23:528531.Google Scholar
Solimena, M, Folli, F, Denis-Donini, S, et al. Autoantibodies to glutamic acid decarboxylase in a patient with stiff-man syndrome, epilepsy, and type I diabetes mellitus. N Engl J Med 1988;318:10121020.Google Scholar
Thirkill, CE, Fitzgerald, P, Sergott, RC, et al. Cancer-associated retinopathy (CAR syndrome) with antibodies reacting with retinal, optic-nerve, and cancer cells [see comments]. N Eng J Med 1989;321:15891594.CrossRefGoogle ScholarPubMed
Sakai, K, Mitchell, DJ, Tsukamoto, T, Steinman, L. Isolation of a complementary DNA clone encoding an autoantigen recognized by an anti-neuronal cell antibody from a patient with paraneoplastic cerebellar degeneration [published erratum appears in Ann Neurol 1991 Nov;30(5):738]. Ann Neurol 1990;28:692698.Google Scholar
Solimena, M, Folli, F, Aparisi, R, Pozza, G, De Camilli, P. Autoantibodies to GABA-ergic neurons and pancreatic beta cells in stiff-man syndrome. N Eng J Med 1990;322:15551560.Google Scholar
Polans, AS, Buczylko, J, Crabb, J, Palczewski, K. A photoreceptor calcium binding protein is recognized by autoantibodies obtained from patients with cancer-associated retinopathy. J Cell Biol 1991;112:981989.CrossRefGoogle ScholarPubMed
Luque, FA, Furneaux, HM, Ferziger, R, et al. Anti-Ri: an antibody associated with paraneoplastic opsoclonus and breast cancer. Ann Neurol 1991;29:241251.CrossRefGoogle ScholarPubMed
Szabo, A, Dalmau, J, Manley, G, et al. HuD, a paraneoplastic encephalomyelitis antigen, contains RNA-binding domains and is homologous to Elav and Sex-lethal. Cell 1991;67:325333.Google Scholar
Peterson, K, Rosenblum, MK, Kotanides, H, Posner, JB. Paraneoplastic cerebellar degeneration. I. A clinical analysis of 55 anti-Yo antibody-positive patients. Neurology 1992;42:19311937.Google Scholar
Dalmau, J, Graus, F, Rosenblum, MK, Posner, JB. Anti-Hu–associated paraneoplastic encephalomyelitis/sensory neuronopathy: a clinical study of 71 patients. Medicine (Baltimore) 1992;71:5972.Google Scholar
Buckanovich, RJ, Posner, JB, Darnell, RB. Nova, the paraneoplastic Ri antigen, is homologous to an RNA-binding protein and is specifically expressed in the developing motor system. Neuron 1993;11:657672.Google Scholar
Antoine, JC, Honnorat, J, Vocanson, C, et al. Posterior uveitis, paraneoplastic encephalomyelitis and auto-antibodies reacting with developmental protein of brain and retina. J Neurol Sci 1993;117:215223.Google Scholar
Folli, F, Solimena, M, Cofiell, R, et al. Autoantibodies to a 128-kd synaptic protein in three women with the stiff-man syndrome and breast cancer. N Eng J Med 1993;328:546551.Google Scholar
De Camilli, P, Thomas, A, Cofiell, R, et al. The synaptic vesicle-associated protein amphiphysin is the 128-kD autoantigen of stiff-man syndrome with breast cancer. J Exp Med 1993;178:22192223.Google Scholar
Dropcho, EJ. Antiamphiphysin antibodies with small-cell lung carcinoma and paraneoplastic encephalomyelitis. Ann Neural 1996;39:659667.Google Scholar
Graus, F, Dalmau, J, Valldeoriola, F, et al. Immunological characterization of a neuronal antibody (anti-Tr) associated with paraneoplastic cerebellar degeneration and Hodgkin’s disease. J Neuroimmunol 1997;74:5561.Google Scholar
Saiz, A, Arpa, J, Sagasta, A, et al. Autoantibodies to glutamic acid decarboxylase in three patients with cerebellar ataxia, late-onset insulin-dependent diabetes mellitus, and polyendocrine autoimmunity. Neurology 1997;49:10261030.CrossRefGoogle ScholarPubMed
Giometto, B, Nicolao, P, Macucci, M, et al. Temporal-lobe epilepsy associated with glutamic-acid-decarboxylase autoantibodies. Lancet 1998;352:457.Google Scholar
Dalmau, J, Gultekin, SH, Voltz, R, et al. Ma1, a novel neuron- and testis-specific protein, is recognized by the serum of patients with paraneoplastic neurological disorders. Brain 1999; 122:2739.Google Scholar
Voltz, R, Gultekin, SH, Rosenfeld, MR, et al. A serologic marker of paraneoplastic limbic and brain-stem encephalitis in patients with testicular cancer [see comments]. N Engl J Med 1999;340:17881795.Google Scholar
Gure, AO, Stockert, E, Scanlan, MJ, et al. Serological identification of embryonic neural proteins as highly immunogenic tumor antigens in small cell lung cancer. Proc Natl Acad Sci USA 2000;97:41984203.Google Scholar
Vernino, S, Lennon, VA. New Purkinje cell antibody (PCA-2): marker of lung cancer-related neurological autoimmunity. Ann Neurol 2000;47:297305.Google Scholar
Yu, Z, Kryzer, TJ, Griesmann, GE, et al. CRMP-5 neuronal autoantibody: marker of lung cancer and thymoma-related autoimmunity. Ann Neurol 2001;49:146154.Google Scholar
Dalmau, J, Graus, F, Villarejo, A, et al. Clinical analysis of anti-Ma2-associated encephalitis. Brain 2004;127:18311844.Google Scholar
Sommer, C, Weishaupt, A, Brinkhoff, J, et al. Paraneoplastic stiff-person syndrome: passive transfer to rats by means of IgG antibodies to amphiphysin. Lancet 2005;365:14061411.Google Scholar
Sabater, L, Titulaer, M, Saiz, A, et al. SOX1 antibodies are markers of paraneoplastic Lambert Eaton myasthenic syndrome. Neurology 2008;70:924928.Google Scholar
de Graaff, E, Maat, P, Hulsenboom, E, et al. Identification of delta/notch-like epidermal growth factor-related receptor as the Tr antigen in paraneoplastic cerebellar degeneration. Ann Neurol 2012;71:815824.Google Scholar
Fang, B, McKeon, A, Hinson, SR, et al. Autoimmune glial fibrillary acidic protein astrocytopathy: a novel meningoencephalomyelitis. JAMA Neurol 2016;73:12971307.Google Scholar
Gadoth, A, Kryzer, TJ, Fryer, J, et al. Microtubule-associated protein 1B: novel paraneoplastic biomarker. Ann Neurol 2017;81:266277.Google Scholar
Krakenes, T, Herdlevaer, I, Raspotnig, M, et al. CDR2L is the major Yo antibody target in paraneoplastic cerebellar degeneration. Ann Neurol 2019;86:316321.Google Scholar
Mandel-Brehm, C, Dubey, D, Kryzer, TJ, et al. Kelch-like protein 11 antibodies in seminoma-associated paraneoplastic encephalitis. N Engl J Med 2019;381:4754.Google Scholar
Seeger, RC, Zeltzer, PM, Rayner, SA. Onco-neural antigen: a new neural differentiation antigen expressed by neuroblastoma, oat cell carcinoma, Wilms’ tumor, and sarcoma cells. J Immunol 1979;122:15481555.Google Scholar
Graus, F, Delattre, JY, Antoine, JC, et al. Recommended diagnostic criteria for paraneoplastic neurological syndromes. J Neurol Neurosurg Psychiatry 2004;75:11351140.Google Scholar
Bernal, F, Graus, F, Pifarre, A, et al. Immunohistochemical analysis of anti-Hu-associated paraneoplastic encephalomyelitis. Acta Neuropathol (Berl) 2002;103:509515.Google Scholar
Graus, F, Dalmau, J. Paraneoplastic neurological syndromes. Curr Opin Neurol 2012;25:795801.Google Scholar
Greenlee, JE, Boyden, JW, Pingree, M, et al. Antibody types and IgG subclasses in paraneoplastic neurological syndromes. J Neurol Sci 2001;184:131137.Google Scholar
Graus, F, Illa, I, Agusti, M, Ribalta, T, Cruz-Sanchez, F. Effect of intraventricular injection of an anti-Purkinje cell antibody (anti-Yo) in a guinea pig model. J Neurol Sci 1991;106:8287.Google Scholar
Carpentier, AF, Rosenfeld, MR, Delattre, JY, et al. DNA vaccination with HuD inhibits growth of a neuroblastoma in mice. Clin Cancer Res 1998;4:28192824.Google Scholar
Pellkofer, H, Schubart, AS, Hoftberger, R, et al. Modelling paraneoplastic CNS disease: T-cells specific for the onconeuronal antigen PNMA1 mediate autoimmune encephalomyelitis in the rat. Brain 2004;127:18221830.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
Graus, F, Vogrig, A, Muñiz-Castrillo, S, et al. Updated diagnostic criteria for paraneoplastic neurologic syndromes. Neurol Neuroimmunol Neuroinflamm 2021;8:e1014.Google Scholar
Graus, F, Keime-Guibert, F, Rene, R, et al. Anti-Hu-associated paraneoplastic encephalomyelitis: analysis of 200 patients. Brain 2001;124:11381148.CrossRefGoogle Scholar
Pittock, SJ, Lucchinetti, CF, Lennon, VA. Anti-neuronal nuclear autoantibody type 2: paraneoplastic accompaniments. Ann Neurol 2003;53:580587.Google Scholar
Pittock, SJ, Lucchinetti, CF, Parisi, JE, et al. Amphiphysin autoimmunity: paraneoplastic accompaniments. Ann Neurol 2005;58:96107.Google Scholar
Maudes, E, Landa, J, Munoz-Lopetegi, A, et al. Clinical significance of Kelch-like protein 11 antibodies. Neurol Neuroimmunol Neuroinflamm 2020;7;e666.Google Scholar
Dubey, D, Wilson, MR, Clarkson, B, et al. Expanded clinical phenotype, oncological associations, and immunopathologic insights of paraneoplastic Kelch-like protein-11 encephalitis. JAMA Neurol 2020;77:14201429.Google Scholar
Bronicki, LM, Jasmin, BJ. Emerging complexity of the HuD/ELAVl4 gene: implications for neuronal development, function, and dysfunction. RNA (New York, NY) 2013;19:10191037.Google Scholar
Graus, F, Dalmau, J, Rene, R, et al. Anti-Hu antibodies in patients with small-cell lung cancer: association with complete response to therapy and improved survival. Chin Ger J Clin Oncol 1997;15:28662872.Google Scholar
Brot, S, Smaoune, H, Youssef-Issa, M, et al. Collapsin response-mediator protein 5 (CRMP5) phosphorylation at threonine 516 regulates neurite outgrowth inhibition. Eur J Neurosci 2014;40:30103020.Google Scholar
Sabater, L, Saiz, A, Dalmau, J, Graus, F. Pitfalls in the detection of CV2 (CRMP5) antibodies. J Neuroimmunol 2016;290:8083.Google Scholar
Julian, LM, McDonald, AC, Stanford, WL. Direct reprogramming with SOX factors: masters of cell fate. Curr Opin Genet Dev 2017;46:2436.Google Scholar
Ruiz-Garcia, R, Martinez-Hernandez, E, Garcia-Ormaechea, M, et al. Caveats and pitfalls of SOX1 autoantibody testing with a commercial line blot assay in paraneoplastic neurological investigations. Front Immunol 2019;10:769.Google Scholar
Herdlevær, I, Haugen, M, Mazengia, K, Totland, C, Vedeler, C. Paraneoplastic cerebellar degeneration: the importance of including cdr2l as a diagnostic marker. Neurol Neuroimmunol Neuroinflamm 2021;8:e963.Google Scholar
Werner, C, Pauli, M, Doose, S, et al. Human autoantibodies to amphiphysin induce defective presynaptic vesicle dynamics and composition. Brain 2016;139:365379.Google Scholar
Dalmau, J, Voltz, R, Eichen, JG, et al. Antibodies against Ma1–Ma5 define distinct paraneoplastic neurologic syndromes associated with limbic, brainstem, or cerebellar dysfunction. Neurology 1999;52:197198.Google Scholar
Villarroel-Campos, D, Gonzalez-Billault, C. The MAP1B case: an old MAP that is new again. Dev Neurobiol 2014;74:953971.Google Scholar
McKeon, A, Tracy, JA, Pittock, SJ, et al. Purkinje cell cytoplasmic autoantibody type 1 accompaniments: the cerebellum and beyond. Arch Neurol 2011;68:12821289.Google Scholar
Petit, T, Janser, JC, Achour, NR, Borel, C, Haegele, P. Paraneoplastic temporal lobe epilepsy and anti-Yo autoantibody. Ann Oncol 1997;8:919.Google Scholar
Goldstein, L, Djaldetti, R, Benninger, F. Anti-Yo, chorea and hemiballismus: a case report. J Clin Neurosci 2017;42:113114.Google Scholar
Khwaja, S, Sripathi, N, Ahmad, BK, Lennon, VA. Paraneoplastic motor neuron disease with type 1 Purkinje cell antibodies. Muscle Nerve 1998;21:943945.Google Scholar
McNamara, P, Costelloe, L, Langan, Y, Redmond, J. Anti-Yo positive dorsal root ganglionopathy. J Neurol 2011;258:519520.Google Scholar
Altermatt, HJ, Rodriguez, M, Scheithauer, BW, Lennon, VA. Paraneoplastic anti-Purkinje and type I anti-neuronal nuclear autoantibodies bind selectively to central, peripheral, and autonomic nervous system cells. Lab Investig 1991;65:412420.Google ScholarPubMed
Monstad, SE, Storstein, A, Dorum, A, et al. Yo antibodies in ovarian and breast cancer patients detected by a sensitive immunoprecipitation technique. Clin Exp Immunol 2006;144:5358.Google Scholar
Linnoila, J, Guo, Y, Gadoth, A, et al. Purkinje cell cytoplasmic antibody type I (anti-Yo): predictive of gastrointestinal adenocarcinomas in men. J Neurol Neurosurg Psychiatry 2018;89:11161117.Google Scholar
Rojas, I, Graus, F, Keime-Guibert, F, et al. Long-term clinical outcome of paraneoplastic cerebellar degeneration and anti-Yo antibodies. Neurology 2000;55:713715.Google Scholar
Cunningham, J, Graus, F, Anderson, N, Posner, JB. Partial characterization of the Purkinje cell antigens in paraneoplastic cerebellar degeneration. Neurology 1986;36:11631168.Google Scholar
Dropcho, EJ, Chen, YT, Posner, JB, Old, LJ. Cloning of a brain protein identified by autoantibodies from a patient with paraneoplastic cerebellar degeneration. Proc Natl Acad Sci USA 1987;84:45524556.Google Scholar
Furneaux, HM, Dropcho, EJ, Barbut, D, et al. Characterization of a cDNA encoding a 34-kDa Purkinje neuron protein recognized by sera from patients with paraneoplastic cerebellar degeneration. Proc Natl Acad Sci USA 1989;86:28732877.Google Scholar
Totland, C, Krakenes, T, Mazengia, K, Haugen, M, Vedeler, C. Expression of the onconeural protein CDR1 in cerebellum and ovarian cancer. Oncotarget 2018;9:2397523986.Google Scholar
Fathallah-Shaykh, H, Wolf, S, Wong, E, Posner, JB, Furneaux, HM. Cloning of a leucine-zipper protein recognized by the sera of patients with antibody-associated paraneoplastic cerebellar degeneration. Proc Natl Acad Sci USA 1991;88:34513454.Google Scholar
Darnell, JC, Albert, ML, Darnell, RB. Cdr2, a target antigen of naturally occuring human tumor immunity, is widely expressed in gynecological tumors. Cancer Res 2000;60:21362139.Google Scholar
Corradi, JP, Yang, CW, Darnell, JC, Dalmau, J, Darnell, RB. A post-transcriptional regulatory mechanism restricts expression of the paraneoplastic cerebellar degeneration antigen cdr2 to immune privileged tissues. J Neurosci 1997;17:14061415.Google Scholar
Sakai, K, Ogasawara, T, Hirose, G, Jaeckle, KA, Greenlee, JE. Analysis of autoantibody binding to 52-kd paraneoplastic cerebellar degeneration-associated antigen expressed in recombinant proteins. Ann Neurol 1993;33:373380.Google Scholar
Herdlevaer, I, Kråkenes, T, Schubert, M, Vedeler, CA. Localization of CDR2L and CDR2 in paraneoplastic cerebellar degeneration. Ann Clin Transl Neurol 2020;7;22312242.Google Scholar
Okano, HJ, Park, WY, Corradi, JP, Darnell, RB. The cytoplasmic Purkinje onconeural antigen cdr2 down-regulates c-Myc function: implications for neuronal and tumor cell survival. Genes Dev 1999;13:20872097.Google Scholar
O’Donovan, KJ, Diedler, J, Couture, GC, Fak, JJ, Darnell, RB. The onconeural antigen cdr2 is a novel APC/C target that acts in mitosis to regulate c-myc target genes in mammalian tumor cells. PLoS One 2010;5:e10045.Google Scholar
Eichler, TW, Totland, C, Haugen, M, et al. CDR2L antibodies: a new player in paraneoplastic cerebellar degeneration. PLoS One 2013;8:e66002.Google Scholar
Raspotnig, M, Haugen, M, Thorsteinsdottir, M, et al. Cerebellar degeneration-related proteins 2 and 2-like are present in ovarian cancer in patients with and without Yo antibodies. Cancer Immunol Immunother 2017;66:14631471.Google Scholar
Ruiz-García, R, Martínez-Hernández, E, Saiz, A, Dalmau, J, Graus, F. The diagnostic value of onconeural antibodies depends on how they are tested. Front Immunol 2020;11:1482.Google Scholar
Small, M, Treilleux, I, Couillault, C, et al. Genetic alterations and tumor immune attack in Yo paraneoplastic cerebellar degeneration. Acta Neuropathol 2018;135:569579.Google Scholar
Graus, F, Elkon, KB, Cordon-Cardo, C, Posner, JB. Sensory neuronopathy and small cell lung cancer: antineuronal antibody that also reacts with the tumor. Am J Med 1986;80:4552.Google Scholar
Henson, RA, Hoffman, HL, Urich, H. Encephalomyelitis with carcinoma. Brain 1965;88:449464.Google Scholar
Honnorat, J, Didelot, A, Karantoni, E, et al. Autoimmune limbic encephalopathy and anti-Hu antibodies in children without cancer. Neurology 2013;80:22262232.Google Scholar
Antunes, NL, Khakoo, Y, Matthay, KK, et al. Antineuronal antibodies in patients with neuroblastoma and paraneoplastic opsoclonus-myoclonus. J Pediatr Hematol Oncol 2000;22:315320.Google Scholar
Liu, J, Dalmau, J, Szabo, A, et al. Paraneoplastic encephalomyelitis antigens bind to the AU-rich elements of mRNA. Neurology 1995;45:544550.Google Scholar
Ince-Dunn, G, Okano, HJ, Jensen, KB, et al. Neuronal Elav-like (Hu) proteins regulate RNA splicing and abundance to control glutamate levels and neuronal excitability. Neuron 2012;75:10671080.CrossRefGoogle ScholarPubMed
Manley, GT, Smitt, PS, Dalmau, J, Posner, JB. Hu antigens: reactivity with Hu antibodies, tumor expression, and major immunogenic sites. Ann Neurol 1995;38:102110.Google Scholar
Kumagai, T, Kitagawa, Y, Hirose, G, Sakai, K. Antibody recognition and RNA binding of a neuronal nuclear autoantigen associated with paraneoplastic neurological syndromes and small cell lung carcinoma. J Neuroimmunol 1999;93:3744.Google Scholar
Dalmau, J, Furneaux, HM, Cordon-Cardo, C, Posner, JB. The expression of the Hu (paraneoplastic encephalomyelitis/sensory neuronopathy) antigen in human normal and tumor tissues. Am J Pathol 1992;141:881886.Google Scholar
Carpentier, AF, Voltz, R, DesChamps, T, et al. Absence of HuD gene mutations in paraneoplastic small cell lung cancer tissue. Neurology 1998;50:1919.Google Scholar
D’Alessandro, V, Muscarella, LA, la Torre, A, et al. Molecular analysis of the HuD gene in neuroendocrine lung cancers. Lung Cancer 2010;67:6975.Google Scholar
Pulido, MA, DerHartunian, MK, Qin, Z, et al. Isoaspartylation appears to trigger small cell lung cancer-associated autoimmunity against neuronal protein ELAVL4. J Neuroimmunol 2016;299:7078.Google Scholar
Fueyo, J, Ferrer, I, Valldeoriola, F, Graus, F. The expression of a neuronal nuclear antigen (Ri) recognized by the human anti-Ri autoantibody in the developing rat nervous system. Neurosci Lett 1993;162:141144.Google Scholar
McCabe, DJ, Turner, NC, Chao, D, et al. Paraneoplastic ‘stiff person syndrome’ with metastatic adenocarcinoma and anti-Ri antibodies. Neurology 2004;62:14021404.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
Brieva-Ruiz, L, Diaz-Hurtado, M, Matias-Guiu, X, et al. Anti-Ri-associated paraneoplastic cerebellar degeneration and breast cancer: an autopsy case study. Clin Neurol Neurosurg 2008;110:10441046.Google Scholar
Younger, DS, Graber, J, Hayakawa-Yano, Y, et al. Ri/Nova gene-associated paraneoplastic subacute motor neuronopathy. Muscle Nerve 2013;47:617618.Google Scholar
Sutton, IJ, Barnett, MH, Watson, JD, Ell, JJ, Dalmau, J. Paraneoplastic brainstem encephalitis and anti-Ri antibodies. J Neurol 2002;249:15971598.Google Scholar
Drlicek, M, Bianchi, G, Bogliun, G, et al. Antibodies of the anti-Yo and anti-Ri type in the absence of paraneoplastic neurological syndromes: a long-term survey of ovarian cancer patients. J Neurol 1997;244:8589.Google Scholar
Monstad, SE, Knudsen, A, Salvesen, HB, Aarseth, JH, Vedeler, CA. Onconeural antibodies in sera from patients with various types of tumours. Cancer Immunol Immunother 2009;58:17951800.Google Scholar
Graus, F, Rowe, G, Fueyo, J, Darnell, RB, Dalmau, J. The neuronal nuclear antigen recognized by the human anti-Ri autoantibody is expressed in central but not peripheral nervous system neurons. Neurosci Lett 1993;150:212214.Google Scholar
Yang, YY, Yin, GL, Darnell, RB. The neuronal RNA-binding protein Nova-2 is implicated as the autoantigen targeted in POMA patients with dementia. Proc Natl Acad Sci USA 1998;95:1325413259.CrossRefGoogle ScholarPubMed
Jensen, KB, Dredge, BK, Stefani, G, et al. Nova-1 regulates neuron-specific alternative splicing and is essential for neuronal viability [see comments]. Neuron 2000;25:359371.Google Scholar
Buckanovich, RJ, Yang, YY, Darnell, RB. The onconeural antigen Nova-1 is a neuron-specific RNA-binding protein, the activity of which is inhibited by paraneoplastic antibodies. J Neurosci 1996;16:11141122.Google Scholar
Zhang, YA, Liu, HN, Zhu, JM, et al. RNA binding protein Nova1 promotes tumor growth in vivo and its potential mechanism as an oncogene may due to its interaction with GABAA Receptor-gamma2. J Biomed Sci 2016;23:71.Google Scholar
Li, C, He, Y, Ma, H, Han, S. NOVA1 acts as an oncogene in osteosarcoma. Am J Transl Res 2017;9:44504457.Google Scholar
Antoine, JC, Honnorat, J, Anterion, CT, et al. Limbic encephalitis and immunological perturbations in two patients with thymoma. J Neurol Neurosurg Psychiatry 1995;58:706710.Google Scholar
Honnorat, J, Antoine, JC, Derrington, E, Aguera, M, Belin, MF. Antibodies to a subpopulation of glial cells and a 66 kDa developmental protein in patients with paraneoplastic neurological syndromes. J Neurol Neurosurg Psychiatry 1996;61:270278.Google Scholar
Cross, SA, Salomao, DR, Parisi, JE, et al. Paraneoplastic autoimmune optic neuritis with retinitis defined by CRMP-5-IgG. Ann Neurol 2003;54:3850.Google Scholar
Cohen, DA, Bhatti, MT, Pulido, JS, et al. Collapsin response-mediator protein 5-associated retinitis, vitritis, and optic disc edema. Ophthalmology 2020;127:221229.Google Scholar
Jarius, S, Wandinger, KP, Borowski, K, Stoecker, W, Wildemann, B. Antibodies to CV2/CRMP5 in neuromyelitis optica-like disease: case report and review of the literature. Clin Neurol Neurosurg 2012;114:331335.Google Scholar
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
Dubey, D, Lennon, VA, Gadoth, A, et al. Autoimmune CRMP5 neuropathy phenotype and outcome defined from 105 cases. Neurology 2018;90:e103e110.Google Scholar
Antoine, JC, Honnorat, J, Camdessanche, JP, et al. Paraneoplastic anti-CV2 antibodies react with peripheral nerve and are associated with a mixed axonal and demyelinating peripheral neuropathy. Ann Neurol 2001;49:214221.Google Scholar
Bataller, L, Wade, DF, Graus, F, et al. Antibodies to Zic4 in paraneoplastic neurologic disorders and small-cell lung cancer. Neurology 2004;62:778782.Google Scholar
Monstad, SE, Drivsholm, L, Skeie, GO, Aarseth, JH, Vedeler, CA. CRMP5 antibodies in patients with small-cell lung cancer or thymoma. Cancer Immunol Immunother 2008;57:227232.Google Scholar
Schmidt, EF, Strittmatter, SM. The CRMP family of proteins and their role in Sema3A signaling. Adv Exp Med Biol 2007;600:111.Google Scholar
Naudet, N, Moutal, A, Vu, HN, et al. Transcriptional regulation of CRMP5 controls neurite outgrowth through Sox5. Cell Molec Life Sci 2018;75:6779.Google Scholar
Ricard, D, Rogemond, V, Charrier, E, et al. Isolation and expression pattern of human Unc-33-like phosphoprotein 6/collapsin response mediator protein 5 (Ulip6/CRMP5): coexistence with Ulip2/CRMP2 in Sema3a-sensitive oligodendrocytes. J Neurosci 2001;21:72037214.Google Scholar
Brot, S, Malleval, C, Benetollo, C, et al. Identification of a new CRMP5 isoform present in the nucleus of cancer cells and enhancing their proliferation. Exp Cell Res 2013;319:588599.Google Scholar
Murinson, BB, Guarnaccia, JB. Stiff-person syndrome with amphiphysin antibodies: distinctive features of a rare disease. Neurology 2008;71:19551958.Google Scholar
Saiz, A, Dalmau, J, Butler, MH, et al. Anti-amphiphysin I antibodies in patients with paraneoplastic neurological disorders associated with small cell lung carcinoma. J Neurol Neurosurg Psychiatry 1999;66:214217.Google Scholar
Irani, SR. ‘Moonlighting’ surface antigens: a paradigm for autoantibody pathogenicity in neurology? Brain 2016;139:304306.Google Scholar
Trotter, JL, Hendin, BA, Osterland, K. Cerebellar degeneration with Hodgkin’s disease: an immunological study. Arch Neurol 1976;33:660661.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
Probst, C, Komorowski, L, de Graaff, E, et al. Standardized test for anti-Tr/DNER in patients with paraneoplastic cerebellar degeneration. Neurol Neuroimmunol Neuroinflamm 2015;2:e68.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
Bergner, CG, Lang, C, Spreer, A, et al. Teaching NeuroImages: Ma2 encephalitis presenting as acute panhypopituitarism in a young man. Neurology 2013;81:e146147.Google Scholar
Murphy, SM, Khan, U, Alifrangis, C, et al. Anti Ma2-associated myeloradiculopathy: expanding the phenotype of anti-Ma2 associated paraneoplastic syndromes. J Neurol Neurosurg Psychiatry 2012;83:232233.Google Scholar
Ayrignac, X, Castelnovo, G, Landrault, E, et al. Ma2 antibody and multiple mononeuropathies. Rev Neurol (Paris) 2008;164:608611.Google Scholar
Pang, SW, Lahiri, C, Poh, CL, Tan, KO. PNMA family: protein interaction network and cell signalling pathways implicated in cancer and apoptosis. Cell Signall 2018;45:5462.Google Scholar
Lee, YH, Pang, SW, Tan, KO. PNMA2 mediates heterodimeric interactions and antagonizes chemo-sensitizing activities mediated by members of PNMA family. Biochem Biophys Res Commun 2016;473:224229.Google Scholar
Chen, HL, D’Mello, SR. Induction of neuronal cell death by paraneoplastic Ma1 antigen. J Neurosci Res 2010;88:35083519.Google Scholar
Rosenfeld, MR, Eichen, JG, Wade, DF, Posner, JB, Dalmau, J. Molecular and clinical diversity in paraneoplastic immunity to Ma proteins. Ann Neurol 2001;50:339348.Google Scholar
Johannis, W, Renno, JH, Wielckens, K, Voltz, R. Ma2 antibodies: an evaluation of commercially available detection methods. Clin Lab 2011;57:321326.Google Scholar
Titulaer, MJ, Klooster, R, Potman, M, et al. SOX antibodies in small-cell lung cancer and Lambert–Eaton myasthenic syndrome: frequency and relation with survival. J Clin Oncol 2009;27:42604267.Google Scholar
Schepers, GE, Teasdale, RD, Koopman, P. Twenty pairs of sox: extent, homology, and nomenclature of the mouse and human sox transcription factor gene families. Dev Cell 2002;3:167170.Google Scholar
Vural, B, Chen, LC, Saip, P, et al. Frequency of SOX Group B (SOX1, 2, 3) and ZIC2 antibodies in Turkish patients with small cell lung carcinoma and their correlation with clinical parameters. Cancer 2005;103:25752583.Google Scholar
Maddison, P, Titulaer, MJ, Verschuuren, JJ, et al. The utility of anti-SOX2 antibodies for cancer prediction in patients with paraneoplastic neurological disorders. J Neuroimmunol 2019;326:1418.Google Scholar
Tschernatsch, M, Gross, O, Kneifel, N, Kaps, M, Blaes, F. SOX-1 autoantibodies in patients with paraneoplastic neurological syndromes. Autoimmunity Reviews 2009;8:549551.Google Scholar
Berger, B, Dersch, R, Ruthardt, E, et al. Prevalence of anti-SOX1 reactivity in various neurological disorders. J Neurol Sci 2016;369:342346.Google Scholar
Sottile, V, Li, M, Scotting, PJ. Stem cell marker expression in the Bergmann glia population of the adult mouse brain. Brain Res 2006;1099:817.Google Scholar
Xu, YR, Yang, WX. SOX-mediated molecular crosstalk during the progression of tumorigenesis. Seminar Cell Dev Biol 2017;63:2334.Google Scholar
Graus, F, Vincent, A, Pozo-Rosich, P, et al. Anti-glial nuclear antibody: marker of lung cancer-related paraneoplastic neurological syndromes. J Neuroimmunol 2005;165:166171.Google Scholar
Dhanoa, BS, Cogliati, T, Satish, AG, Bruford, EA, Friedman, JS. Update on the Kelch-like (KLHL) gene family. Hum Genomics 2013;7:13.Google Scholar
Jitprapaikulsan, J, Klein, CJ, Pittock, SJ, et al. Phenotypic presentations of paraneoplastic neuropathies associated with MAP1B-IgG. J Neurol Neurosurg Psychiatry 2020;91:328330.Google Scholar
Tortosa, E, Montenegro-Venegas, C, Benoist, M, et al. Microtubule-associated protein 1B (MAP1B) is required for dendritic spine development and synaptic maturation. J Biol Chem 2011;286:4063840648.Google Scholar
Chien, TM, Chan, TC, Huang, SK, et al. Role of microtubule-associated protein 1b in urothelial carcinoma: overexpression predicts poor prognosis. Cancers 2020;12;630.Google Scholar
Chan, KH, Vernino, S, Lennon, VA. ANNA-3 anti-neuronal nuclear antibody: marker of lung cancer-related autoimmunity. Ann Neurol 2001;50:301311.Google Scholar
Bataller, L, Wade, DF, Graus, F, et al. Antibodies to Zic4 in paraneoplastic neurologic disorders and small-cell lung cancer. Neurology 2004;62:778782.Google Scholar
Basal, E, Zalewski, N, Kryzer, TJ, et al. Paraneoplastic neuronal intermediate filament autoimmunity. Neurology 2018;91:e1677e1689.Google Scholar
Zekeridou, A, Kryzer, T, Guo, Y, et al. Phosphodiesterase 10 A IgG: a novel biomarker of paraneoplastic neurologic autoimmunity. Neurology 2019;93:e815e822.Google Scholar
Sabater, L, Gomez-Choco, M, Saiz, A, Graus, F. BR serine/threonine kinase 2: a new autoantigen in paraneoplastic limbic encephalitis. J Neuroimmunol 2005;170:186190.Google Scholar
Popkirov, S, Ayzenberg, I, Hahn, S, et al. Rho-associated protein kinase 2 (ROCK2): a new target of autoimmunity in paraneoplastic encephalitis. Acta Neuropathologica Commun 2017;5:40.Google Scholar
Sabater, L, Bataller, L, Carpentier, AF, et al. Protein kinase Cgamma autoimmunity in paraneoplastic cerebellar degeneration and non-small-cell lung cancer. J Neurol Neurosurg Psychiatry 2006;77:13591362.Google Scholar
Tetsuka, S, Tominaga, K, Ohta, E, et al. Paraneoplastic cerebellar degeneration associated with an onconeural antibody against creatine kinase, brain-type. J Neurol Sci 2013;335:4857.Google Scholar
Do, LD, Gupton, SL, Tanji, K, et al. TRIM9 and TRIM67 are new targets in paraneoplastic cerebellar degeneration. Cerebellum 2018;18:245254.Google Scholar
van Coevorden-Hameete, MH, van Beuningen, SFB, Perrenoud, M, et al. Antibodies to TRIM46 are associated with paraneoplastic neurological syndromes. Ann Clin Transl Neurol 2017;4:680686.Google Scholar
Bataller, L, Sabater, L, Saiz, A, et al. Carbonic anhydrase-related protein VIII: autoantigen in paraneoplastic cerebellar degeneration. Ann Neurol 2004;56:575579.Google Scholar
Hoftberger, R, Kovacs, GG, Sabater, L, et al. Protein kinase Cgamma antibodies and paraneoplastic cerebellar degeneration. J Neuroimmunol 2012;256:9193.Google Scholar
Tschernatsch, M, Klotz, M, Probst, C, et al. Synaptophysin is an autoantigen in paraneoplastic neuropathy. J Neuroimmunol 2008;197:8186.Google Scholar
Butler, MH, Hayashi, A, Ohkoshi, N, et al. Autoimmunity to gephyrin in Stiff-Man syndrome. Neuron 2000;26:307312.Google Scholar
van Coevorden-Hameete, MH, de Graaff, E, Titulaer, MJ, et al. Plasticity-related gene 5: a novel surface autoantigen in paraneoplastic cerebellar degeneration. Neurol Neuroimmunol Neuroinflamm 2015;2:e156.Google Scholar
Bataller, L, Wade, DF, Fuller, GN, Rosenfeld, MR, Dalmau, J. Cerebellar degeneration and autoimmunity to zinc-finger proteins of the cerebellum. Neurology 2002;59:19851987.CrossRefGoogle ScholarPubMed
Sabater, L, Hoftberger, R, Boronat, A, et al. Antibody repertoire in paraneoplastic cerebellar degeneration and small cell lung cancer. PLoS One 2013;8:e60438.Google Scholar
Aydin, C, Celik, SY, Icoz, S, et al. Prognostic factors in anti-neuronal antibody positive patients. Noro psikiyatri arsivi 2018;55:189194.Google Scholar
Eye, PG, Wang, B, Keung, ES, Tagg, NT. Anti-ZIC4 associated paraneoplastic cerebellar degeneration in a patient with both diffuse large B-cell lymphoma and incidental smoldering multiple myeloma. J Neurol Sci 2018;384:3637.Google Scholar
Kerasnoudis, A, Rockhoff, M, Federlein, J, Gold, R, Krogias, C. Isolated ZIC4 antibodies in paraneoplastic cerebellar syndrome with an underlying ovarian tumor. Arch Neurol 2011;68:1073.Google Scholar
Dechelotte, B, Muniz-Castrillo, S, Joubert, B, et al. Diagnostic yield of commercial immunodots to diagnose paraneoplastic neurologic syndromes. Neurol Neuroimmunol Neuroinflamm 2020;7;e701.Google Scholar
Keltner, JL, Thirkill, CE, Yip, PT. Clinical and immunologic characteristics of melanoma-associated retinopathy syndrome: eleven new cases and a review of 51 previously published cases. J Neuroophthalmol 2001;21:173187.Google Scholar
Grewal, DS, Fishman, GA, Jampol, LM. Autoimmune retinopathy and antiretinal antibodies: a review. Retina (Philadelphia, Pa) 2014;34:827845.Google Scholar
Adamus, G. Are anti-retinal autoantibodies a cause or a consequence of retinal degeneration in autoimmune retinopathies? Front Immunol 2018;9:765.Google Scholar
Braithwaite, T, Holder, GE, Lee, RW, Plant, GT, Tufail, A. Diagnostic features of the autoimmune retinopathies. Autoimmun Rev 2014;13:534538.Google Scholar
Fox, AR, Gordon, LK, Heckenlively, JR, et al. Consensus on the diagnosis and management of nonparaneoplastic autoimmune retinopathy using a modified Delphi approach. Am J Ophthalmol 2016;168:183190.Google Scholar
Faez, S, Loewenstein, J, Sobrin, L. Concordance of antiretinal antibody testing results between laboratories in autoimmune retinopathy. JAMA Ophthalmol 2013;131:113115.Google Scholar
Yang, S, Dizhoor, A, Wilson, DJ, Adamus, G. GCAP1, Rab6, and HSP27: novel autoantibody targets in cancer-associated retinopathy and autoimmune retinopathy. Transl Vision Sci Technol 2016;5:1.Google Scholar
Forooghian, F. The uncertainty regarding antiretinal antibodies. JAMA Ophthalmol 2015;133:744745.Google Scholar
Adamus, G, Champaigne, R, Yang, S. Occurrence of major anti-retinal autoantibodies associated with paraneoplastic autoimmune retinopathy. Clin Immunol (Orlando, Fla) 2020;210:108317.Google Scholar
Adamus, G, Ren, G, Weleber, RG. Autoantibodies against retinal proteins in paraneoplastic and autoimmune retinopathy. BMC Ophthalmol 2004;4:5.Google Scholar
Bazhin, AV, Schadendorf, D, Philippov, PP, Eichmuller, SB. Recoverin as a cancer-retina antigen. Cancer Immunol Immunother 2007;56:110116.Google Scholar
Gibbs, E, Matsubara, J, Cao, S, Cui, J, Forooghian, F. Antigen-specificity of antiretinal antibodies in patients with noninfectious uveitis. Can J Ophthalmol 2017;52:463467.Google Scholar
Ten Berge, JC, van Rosmalen, J, Vermeer, J, et al. Serum autoantibody profiling of patients with paraneoplastic and non-paraneoplastic autoimmune retinopathy. PLoS One 2016;11:e0167909.Google Scholar
Bazhin, AV, Savchenko, MS, Shifrina, ON, et al. Recoverin as a paraneoplastic antigen in lung cancer: the occurrence of anti-recoverin autoantibodies in sera and recoverin in tumors. Lung Cancer 2004;44:193198.Google Scholar
Gorodovikova, EN, Gimelbrant, AA, Senin, II, Philippov, PP. Recoverin mediates the calcium effect upon rhodopsin phosphorylation and cGMP hydrolysis in bovine retina rod cells. FEBS Lett 1994;349:187190.Google Scholar
Adamus, G, Machnicki, M, Seigel, GM. Apoptotic retinal cell death induced by antirecoverin autoantibodies of cancer-associated retinopathy. Investig Ophthalmol Visual Sci 1997;38:283291.Google Scholar
Shiraga, S, Adamus, G. Mechanism of CAR syndrome: anti-recoverin antibodies are the inducers of retinal cell apoptotic death via the caspase 9- and caspase 3-dependent pathway. J Neuroimmunol 2002;132:7282.Google Scholar
Adamus, G, Machnicki, M, Elerding, H, et al. Antibodies to recoverin induce apoptosis of photoreceptor and bipolar cells in vivo. J Autoimmun 1998;11:523533.Google Scholar
Doss, S, Numann, A, Ziegler, A, et al. Anti-Ca/anti-ARHGAP26 antibodies associated with cerebellar atrophy and cognitive decline. J Neuroimmunol 2014;267:102104.Google Scholar
Pittock, SJ, Alfugham, N, O’Connor, K, et al. GTPase regulator associated with focal adhesion kinase 1 (GRAF1) immunoglobulin-associated ataxia and neuropathy. Mov Disord Clin Pract 2020;7:904909.Google Scholar
Jarius, S, Scharf, M, Begemann, N, et al. Antibodies to the inositol 1,4,5-trisphosphate receptor type 1 (ITPR1) in cerebellar ataxia. J Neuroinflammation 2014;11:206.Google Scholar
Alfugham, N, Gadoth, A, Lennon, VA, et al. ITPR1 autoimmunity: frequency, neurologic phenotype, and cancer association. Neurol Neuroimmunol Neuroinflamm 2018;5:e418.Google Scholar
Do, LD, Chanson, E, Desestret, V, et al. Characteristics in limbic encephalitis with anti-adenylate kinase 5 autoantibodies. Neurology 2017;88:514524.Google Scholar
Miske, R, Gross, CC, Scharf, M, et al. Neurochondrin is a neuronal target antigen in autoimmune cerebellar degeneration. Neurol Neuroimmunol Neuroinflamm 2017;4:e307.Google Scholar
Shelly, S, Kryzer, TJ, Komorowski, L, et al. Neurochondrin neurological autoimmunity. Neurol Neuroimmunol Neuroinflamm 2019;6;e612.Google Scholar
Lancaster, E. CNS syndromes associated with antibodies against metabotropic receptors. Curr Opin Neurol 2017;30:354360.Google Scholar
Honorat, JA, Lopez-Chiriboga, AS, Kryzer, TJ, et al. Autoimmune gait disturbance accompanying adaptor protein-3B2-IgG. Neurology 2019;93:e954e963.Google Scholar
Honorat, JA, Lopez-Chiriboga, AS, Kryzer, TJ, et al. Autoimmune septin-5 cerebellar ataxia. Neurol Neuroimmunol Neuroinflamm 2018;5:e474.Google Scholar
Zuliani, L, Sabater, L, Saiz, A, et al. Homer 3 autoimmunity in subacute idiopathic cerebellar ataxia. Neurology 2007;68:239240.Google Scholar
Piepgras, J, Holtje, M, Otto, C, et al. Intrathecal immunoglobulin A and G antibodies to synapsin in a patient with limbic encephalitis. Neurol Neuroimmunol Neuroinflamm 2015;2:e169.Google Scholar
Darnell, RB, Furneaux, HM, Posner, JB. Antiserum from a patient with cerebellar degeneration identifies a novel protein in Purkinje cells, cortical neurons and neuroectodermal tumors. J Neurosci 1991;11:12241230.Google Scholar
McKeon, A, Tracy, JA. GAD65 neurological autoimmunity. Muscle Nerve 2017;56:1527.Google Scholar
Graus, F, Saiz, A, Dalmau, J. GAD antibodies in neurological disorders: insights and challenges. Nat Rev Neurol 2020;16:353365.Google Scholar
Bhandari, HS. Presentation of opsoclonus myoclonus ataxia syndrome with glutamic acid decarboxylase antibodies. BMJ Case Rep 2012;2012:bcr2012006339.Google Scholar
Martins, AI, Carvalho, JN, Amorim, AM, et al. Disabling central paroxysmal positioning upbeat nystagmus and vertigo associated with the presence of anti-glutamic acid decarboxylase antibodies. J Neuroophthalmol 2018;38:3235.Google Scholar
Vianello, M, Morello, F, Scaravilli, T, Tavolato, B, Giometto, B. Tremor of the mouth floor and anti-glutamic acid decarboxylase autoantibodies. Eur J Neurol 2003;10:513514.Google Scholar
Macaron, G, Willis, MA, Ontaneda, D, et al. Palatal myoclonus, abnormal eye movements, and olivary hypertrophy in GAD65-related disorder. Neurology 2020;94:273275.Google Scholar
Ellis, TM, Atkinson, MA. The clinical significance of an autoimmune response against glutamic acid decarboxylase. Nat Med 1996;2:148153.Google Scholar
Erlander, MG, Tobin, AJ. The structural and functional heterogeneity of glutamic acid decarboxylase: a review. Neurochem Res 1991;16:215226.Google Scholar
Kaufman, DL, Houser, CR, Tobin, AJ. Two forms of the gamma-aminobutyric acid synthetic enzyme glutamate decarboxylase have distinct intraneuronal distributions and cofactor interactions. J Neurochem 1991;56:720723.Google Scholar
Gresa-Arribas, N, Arino, H, Martinez-Hernandez, E, et al. Antibodies to inhibitory synaptic proteins in neurological syndromes associated with glutamic acid decarboxylase autoimmunity. PLoS One 2015;10:e0121364.Google Scholar
Burbelo, PD, Groot, S, Dalakas, MC, Iadarola, MJ. High definition profiling of autoantibodies to glutamic acid decarboxylases GAD65/GAD67 in stiff-person syndrome. Biochem Biophys Res Commun 2008;366:17.Google Scholar
Dinkel, K, Meinck, HM, Jury, KM, Karges, W, Richter, W. Inhibition of gamma-aminobutyric acid synthesis by glutamic acid decarboxylase autoantibodies in stiff-man syndrome. Ann Neurol 1998;44:194201.Google Scholar
Dalakas, MC, Li, M, Fujii, M, Jacobowitz, DM. Stiff person syndrome: quantification, specificity, and intrathecal synthesis of GAD65 antibodies. Neurology 2001;57:780784.Google Scholar
Meinck, HM, Faber, L, Morgenthaler, N, et al. Antibodies against glutamic acid decarboxylase: prevalence in neurological diseases. J Neurol Neurosurg Psychiatry 2001;71:100103.Google Scholar
Saiz, A, Blanco, Y, Sabater, L, et al. Spectrum of neurological syndromes associated with glutamic acid decarboxylase antibodies: diagnostic clues for this association. Brain 2008;131:25532563.Google Scholar
Flanagan, EP, Hinson, SR, Lennon, VA, et al. Glial fibrillary acidic protein immunoglobulin G as biomarker of autoimmune astrocytopathy: analysis of 102 patients. Ann Neurol 2017;81:298309.Google Scholar
Kunchok, A, Zekeridou, A, McKeon, A. Autoimmune glial fibrillary acidic protein astrocytopathy. Curr Opin Neurol 2019;32:452458.Google Scholar

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.Google 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.Google Scholar
Lai, M, Hughes, EG, Peng, X, et al. AMPA receptor antibodies in limbic encephalitis alter synaptic receptor location. Ann Neurol 2009;65:424434.Google Scholar
Dalmau, J, Tuzun, E, Wu, HY, et al. Paraneoplastic anti-N-methyl-D-aspartate receptor encephalitis associated with ovarian teratoma. Ann Neurol 2007;61:2536.Google Scholar
Dalmau, J, 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.Google Scholar
Dalmau, J, Gleichman, AJ, Hughes, EG, et al. Anti-NMDA-receptor encephalitis: case series and analysis of the effects of antibodies. Lancet Neurol 2008;7:10911098.Google Scholar
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
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.Google Scholar
Hughes, EG, Peng, X, Gleichman, AJ, et al. Cellular and synaptic mechanisms of anti-NMDA receptor encephalitis. J Neurosci 2010;30:58665875.Google Scholar
Mikasova, L, De Rossi, P, Bouchet, D, et al. Disrupted surface cross-talk between NMDA and Ephrin-B2 receptors in anti-NMDA encephalitis. Brain 2012;135:16061621.Google Scholar
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.Google Scholar
Martinez-Hernandez, E, Guasp, M, Garcia-Serra, A, et al. Clinical significance of anti-NMDAR concurrent with glial or neuronal surface antibodies. Neurology 2020;94:e2302e2310.Google Scholar
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.Google Scholar
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.Google Scholar
Lancaster, E, Lai, M, Peng, X, et al. Antibodies to the GABA(B) receptor in limbic encephalitis with seizures: case series and characterisation of the antigen. Lancet Neurol 2010;9:6776.Google Scholar
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.Google Scholar
Sillevis, SP, Kinoshita, A, De, LB, et al. Paraneoplastic cerebellar ataxia due to autoantibodies against a glutamate receptor. N Engl J Med 2000;342:2127.Google Scholar
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.Google Scholar
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.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
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.Google Scholar
Andrade, DM, Tai, P, Dalmau, J, Wennberg, R. Tonic seizures: a diagnostic clue of anti-LGI1 encephalitis? Neurology 2011;76:13551357.Google Scholar
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.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
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.Google Scholar
Gaig, C, Iranzo, A, Cajochen, C, et al. Characterization of the sleep disorder of anti-IgLON5 disease. Sleep 2019;42:zsz133.Google Scholar
Gaig, C, Graus, F, Compta, Y, et al. Clinical manifestations of the anti-IgLON5 disease. Neurology 2017;88:17361743.Google Scholar
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.Google Scholar
Greene, M, Lai, Y, Baella, N, Dalmau, J, Lancaster, E. Antibodies to Delta/notch-like epidermal growth factor-related receptor in patients with anti-Tr, paraneoplastic cerebellar degeneration, and Hodgkin lymphoma. JAMA Neurol 2014;71:10031008.Google Scholar
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.Google Scholar
Mason, WP, Graus, F, Lang, B, et al. Small-cell lung cancer, paraneoplastic cerebellar degeneration and the Lambert–Eaton myasthenic syndrome. Brain 1997;120:12791300.Google Scholar
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.Google Scholar
Werner, C, Pauli, M, Doose, S, et al. Human autoantibodies to amphiphysin induce defective presynaptic vesicle dynamics and composition. Brain 2016;139:365379.Google Scholar
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.Google Scholar
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
×