Skip to main content

Paraneoplastic Neurological Syndromes

  • Chapter
  • First Online:
Neuroimmune Diseases

Part of the book series: Contemporary Clinical Neuroscience ((CCNE))

Abstract

Paraneoplastic neurological syndromes (PNS) are immune-mediated disorders associated with cancer. PNS usually appear in a subacute manner and may affect any level of the nervous system. They generally develop before cancer and PNS recognition leads to cancer diagnosis. Prompt tumor treatment is important to stabilize or improve PNS symptoms. Although PNS may sometimes occur without antibodies, two major groups are identified depending on the antibodies present in the serum or the CSF. Antibodies against intracellular antigens (also called onconeural antibodies) seem to play no direct role in neurological symptoms (with exceptions) but are good markers of cancer, so their detection is very useful to classify a neurological syndrome as paraneoplastic. Although these antibodies are thought to have no pathogenic effect, the immune system still plays a major role, probably mediated by cytotoxic T cells. PNS with onconeural antibodies respond poorly to immunotherapy, with the exception of anti-Ma2 PNS. A second group of antibodies against synaptic and cell surface proteins occur in well-defined neurological syndromes, which are not always PNS and may appear in younger patients without cancer. These antibodies alter the location or function of their antigens, generating the neuronal dysfunction that is the underlying cause of these PNS. Early immunotherapy improves prognosis at least in the most frequent disorders (anti-NMDA receptor encephalitis, anti-LGI1 encephalitis). In all PNS, irrespective of the group, appropriate tumor screening should be undertaken. The work-up should include the search for the most common cancers reported in PNS patients according to the age and the associated autoantibodies.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 229.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 299.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 299.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

  1. Darnell RB, Posner JB. Paraneoplastic syndromes involving the nervous system. N Engl J Med. 2003;349:1543–54.

    CAS  PubMed  Google Scholar 

  2. Rosenfeld MR, Dalmau J. Paraneoplastic neurologic syndromes. Neurol Clin. 2018;36:675–85.

    PubMed  Google Scholar 

  3. Bradshaw MJ, Linnoila JL. An overview of autoimmune and paraneoplastic encephalitides. Semin Neurol. 2018;38:330–43.

    PubMed  Google Scholar 

  4. Armangué T, Leypoldt F, Málaga I, Raspall-Chaure M, Marti I, Nichter C, et al. Herpes simplex virus encephalitis is a trigger of brain autoimmunity. Ann Neurol. 2014;75(2):317–23.

    PubMed  PubMed Central  Google Scholar 

  5. Lancaster E. Paraneoplastic disorders. Continuum. 2017;23(6):1653–79.

    PubMed  Google Scholar 

  6. Graus F, Ariño H, Dalmau J. Paraneoplastic neurological syndromes in Hodgkin and non-Hodgkin lymphomas. Blood. 2014;123(21):3230–8.

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Giometto B, Grisold W, Vitaliani R, Graus F, Honnorat J, Bertolini G. Paraneoplastic neurological syndrome in the PNS Euronetwork Database. Arch Neurol. 2010;67(3):330–5.

    PubMed  Google Scholar 

  8. Pittock SJ, Lucchinetti CF, Lennon VA. Anti-neural nuclear autoantibody type 2: paraneoplastic accompaniments. Ann Neurol. 2003;53:580–7.

    CAS  PubMed  Google Scholar 

  9. Dubey D, Pittock SJ, Kelly CR, McKeon A, Lopez-Chiriboga AS, Lennon VA, et al. Autoimmune encephalitis epidemiology and a comparison to infectious encephalitis. Ann Neurol. 2018;83:166–77.

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Gable MS, Sheriff H, Dalmau J, Tilley DH, Glaser CA. The frequency of autoimmune N-Methyl-D-Aspartate receptor encephalitis surpasses that of individual viral etiologies in young individuals enrolled in the California Encephalitis Project. Clin Infect Dis. 2012;54(7):899–904.

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Titulaer MJ, McCracken L, Gabilondo I, Armangué T, Glaser C, Iizuka T, et al. Treatment and prognostic factors for long-term outcome in patients with anti-N-Methyl-D-Aspartate (NMDA) receptor encephalitis: a cohort study. Lancet Neurol. 2013;12:157–65.

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Granerod J, Ambrose HE, Davies NW, Clewley JP, Walsh AL, Morgan D, et al. Causes of encephalitis and differences in their clinical presentations in England: a multicentre, population-based prospective study. Lancet Infect Dis. 2010;10(12):835–44.

    PubMed  Google Scholar 

  13. Irani SR, Alexander S, Waters P, Kleopa KA, Pettingill P, Zuliani L, et al. Antibodies to Kv1 potassium channel-complex proteins leucine-rich glioma inactivated protein 1 and contactin-associated protein-2 in limbic encephalitis, Morvan’s syndrome and acquired neuromyotonia. Brain. 2010;133:2734–48.

    PubMed  PubMed Central  Google Scholar 

  14. Graus F, Keime-Guibert F, Reñe R, Benhahia B, Ribalta T, Ascaso C, et al. Anti-Hu-associated paraneoplastic encephalomyelitis: analysis of 200 patients. Brain. 2001;124:1138–48.

    CAS  PubMed  Google Scholar 

  15. Honnorat J, Didelot A, Karantoni E, Ville D, Ducray F, Lambert L, et al. Autoimmune limbic encephalopathy and anti-Hu antibodies in children without cancer. Neurology. 2013;80:2226–32.

    CAS  PubMed  Google Scholar 

  16. Graus F, Delattre JY, Antoine JC, Dalmau J, Giometto B, Grisold W, et al. Recommended diagnostic criteria for paraneoplastic neurological syndromes. J Neurol Neurosurg Psychiatry. 2004;75:1135–40.

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Sharp L, Vernino S. Paraneoplastic neuromuscular disorders. Muscle Nerve. 2012;46(6):841–50.

    PubMed  Google Scholar 

  18. Aussy A, Boyer O, Cordel N. Dermatomyositis and immune-mediated necrotizing myopathies: a window on autoimmunity and cancer. Front Immunol. 2017;8:992. https://doi.org/10.3389/fimmu.2017.00992.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Yang Z, Lin F, Qin B, Liang Y, Zhong R. Polymyositis/dermatomyositis and malignancy risk: a metaanalysis study. J Rheumatol. 2015;42(2):282–91.

    PubMed  Google Scholar 

  20. Hill CL, Zhang Y, Sigurgeirsson B, Pukkala E, Mellemkjaer L, Airio A, et al. Frequency of specific cancer types in dermatomyositis and polymyositis. A population-based study. Lancet. 2011;357:96–100.

    Google Scholar 

  21. Fiorentino DF, Kuo K, Chung L, Zaba L, Li S, Casciola-Rosen L. Distinctive cutaneous and systemic features associated with antitranscriptional intermediary factor-1γ antibodies in adults with dermatomyositis. J Am Acad Dermatol. 2015;723:449–55.

    Google Scholar 

  22. Rogers A, Chung L, Li S, Casciola-Rosen L, Fiorentino DF. The cutaneous and systemic findings associated with nuclear matrix protein-2 antibodies in adult dermatomyositis patients. Arthritis Care Res. 2017; https://doi.org/10.1002/acr.23210.

  23. Allenbach Y, Keraen J, Bouvier AM, Jooste V, Champtiaux N, Hervier B, et al. High risk of cancer in autoimmune necrotizing myopathies: usefulness of myositis specific antibody. Brain. 2016;139:2131–5.

    PubMed  Google Scholar 

  24. Kadoya M, Hida A, Hashimoto Maeda M, Taira K, Ikenaga C, Uchio N, et al. Cancer association as a risk factor for antiHMGCR antibody-positive myopathy. Neurol Neuroimmunol Neuroinflamm. 2016;3(6):e290. https://doi.org/10.1212/NXI.0000000000000290.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Cetin H, Vincent A. Pathogenic mechanisms and clinical correlations in autoimmune myasthenic syndromes. Semin Neurol. 2018;38:344–54.

    PubMed  Google Scholar 

  26. Ohira M, Jeong D, Oh SJ. Seropositive myasthenia gravis associated with small-cell lung carcinoma. J Clin Neurol. 2011;7:43–6.

    PubMed  PubMed Central  Google Scholar 

  27. Simonsen M, Miyabe MM, Ouki HT, Galvao ACR, Leite D, Murayama BAR, et al. Myasthenia as a paraneoplastic manifestation of ovarian cancer. Gynecol Oncol Rep. 2018;25:35–6.

    PubMed  PubMed Central  Google Scholar 

  28. Fratalia L, Larner AJ. Late onset myasthenia gravis and carcinoid tumour: paraneoplastic syndrome? Br J Hosp Med. 2017;78(10):588–9.

    CAS  Google Scholar 

  29. Vernino S, Lennon VA. Autoantiboy profiles and neurological correlations of thymoma. Clin Cancer Res. 2004;10:7270–5.

    CAS  PubMed  Google Scholar 

  30. Romi F. Thymoma in myasthenia gravis: from diagnosis to treatment. Autoimmune Dis. 2011;2011:474512. https://doi.org/10.4061/2011/474512.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Romi F, Skeie GO, Gilhus NE, Aarli JA. Striational antibodies in myasthenia gravis: reactivity and possible clinical significance. Arch Neurol. 2005;62(3):442–6.

    PubMed  Google Scholar 

  32. Wirtz PW, Smallegange TM, Wintzen AR, Verschuuren JJ. Differences in clinical features between the Lamber-Eaton myasthenic syndrome with and without cancer: an analysis of 227 published cases. Clin Neurol Neurosurg. 2002;104:359–63.

    PubMed  Google Scholar 

  33. Titulaer MJ, Verschuuren JJ. Lambert-Eaton myasthenic syndrome: tumor versus nontumor forms. Ann N Y Acad Sci. 2008;1132:129–34.

    PubMed  Google Scholar 

  34. Titulaer MJ, Maddison P, Sont JK, Wirtz PW, Hilton-Jones D, Klooster R, et al. Clinical Dutch-English Lambert-Eaton myasthenic syndrome (LEMS) tumor association prediction score accurately predicts small-lung cancer in the LEMS. J Clin Oncol. 2011;29(07):902–8.

    PubMed  Google Scholar 

  35. Nakao YK, Motomura M, Fukudome T, Fukuda T, Shirashi H, Yoshimura T, et al. Seronegative Lambert-Eaton myasthenic syndrome: study of 110 Japanese patients. Neurology. 2002;59(11):1773–5.

    CAS  PubMed  Google Scholar 

  36. Graus F, Vincent A, Pozo-Rosich P, Sabater L, Saiz A, Lang B, et al. Anti-glial nuclear antibody: marker of lung cancer-related paraneoplastic neurological syndromes. J Neuroimmunol. 2005;165:166–71.

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Sabater L, Titulaer M, Saiz A, Verschuuren J, Gure AO, Graus F. SOX1 antibodies are markers of paraneoplastic Lambert-Eaton myasthenic syndrome. Neurology. 2008;70:924–8.

    CAS  PubMed  Google Scholar 

  38. Titulaer MJ, Klooster R, Potman M, Sabater L, Graus F, Hegeman IM, et al. SOX antibodies in small-cell lung cancer and Lambert-Eaton myasthenic syndrome: frequency and relation with survival. J Clin Oncol. 2009;27:4260–7.

    CAS  PubMed  Google Scholar 

  39. Sawlani K, Katirji B. Peripheral nerve hyperexcitability syndromes. Continuum. 2017;23(5):1437–50.

    PubMed  Google Scholar 

  40. Liewluck T, Klein CJ, Jones LK. Cramp-fasciculation syndrome in patients with and without neural autoantibodies. Muscle Nerve. 2014;49:351–6.

    CAS  PubMed  Google Scholar 

  41. Vernino S, Lennon VA. Ion channel and striational antibodies define a continuum of autoimmune neuromuscular hyperexcitability. Muscle Nerve. 2002;26:702–7.

    CAS  PubMed  Google Scholar 

  42. Hart IK, Maddison P, Newsom-Davis J, Vincent A, Mills KR. Phenotypic variants of autoimmune peripheral nerve hyperexcitability. Brain. 2002;125(8):1887–95.

    PubMed  Google Scholar 

  43. Vincent A, Pettingill P, Pettingill R, Lang B, Birch R, Waters P, et al. Association of leucine-rich glioma inactivated protein 1, contactin-associated protein 2 and contactin 2 antibodies with clinical features and patient-reported pain in acquired neuromyotonia. JAMA Neurol. 2018; https://doi.org/10.1001/jamaneurol.2018.2681.

  44. Klein CJ, Lennon VA, Aston PA, McKeon A, O’Toole O, Quek A, et al. Insights from LGI1 and CASPR2 potassium channel complex autoantibody subtyping. JAMA Neurol. 2013;70(2):229–34.

    PubMed  PubMed Central  Google Scholar 

  45. Gadoth A, Pittock SJ, Dubey D, McKeon A, Britton JW, Schmeling JE, et al. Expanded phenotypes and outcomes among 256 LGI1/CASPR2-IgG positive patients. Ann Neurol. 2017;82(1):79–92.

    CAS  PubMed  Google Scholar 

  46. Rubio-Agusti I, Perez-Miralles F, Sevilla T, Muelas N, Chumillas MJ, Mayordomo F, et al. Peripheral nerve hyperexcitability: a clinical and immunological study of 38 patients. Neurology. 2011;76:172–8.

    CAS  PubMed  Google Scholar 

  47. Irani SR, Pettingil P, Kleopa KA, Schiza N, Waters P, Mazia C, et al. Morvan syndrome: clinical and serological observations in 29 cases. Ann Neurol. 2012;72(2):241–55.

    PubMed  Google Scholar 

  48. Laurencin C, André-Obadia N, Camdessanché JP, Maugière F, Ong E, Vukusic S, et al. Peripheral small fiber dysfunction and neuropathic pain in patients with Morvan syndrome. Neurology. 2015;85:2076–8.

    PubMed  Google Scholar 

  49. Joubert B, Saint-Martin M, Noraz N, Picard G, Rogemond V, Ducray F, 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;73:1115–24.

    PubMed  Google Scholar 

  50. Torres-Vega E, Mancheño N, Cebrián-Silla A, Herranz-Pérez V, Chumillas MJ, Morís G, et al. Netrin-1 receptor antibodies in thymoma-associated neuromyotonia with myasthenia gravis. Neurology. 2017;88:1–8.

    Google Scholar 

  51. Camdessanché JP, Antoine JC, Honnorat J, Vial C, Petiot P, Convers P, et al. Paraneoplastic peripheral neuropathy associated with anti-Hu antibodies. A clinical and electrophysiological study. Brain. 2002;125:166–75.

    PubMed  Google Scholar 

  52. Yu Z, Kryzer TJ, Griesmann GE, Kim KK, Benarroch EE, Lennon VA. CRMP-5 neuronal autoantibody: marker of lung cancer and thymoma-related autoimmunity. Ann Neurol. 2001;49:146–54.

    CAS  PubMed  Google Scholar 

  53. Dubey D, Lennon VA, Gadoth A, Pittock SJ, Flanagan EP, Schmeling JE, et al. Autoimmune CRMP5 neuropathy phenotype and outcome defined from 105 cases. Neurology. 2018;90:e103–10. https://doi.org/10.1212/WNL.0000000000004803.

    Article  CAS  PubMed  Google Scholar 

  54. Lee HR, Lennon VA, Camilleri M, Prather CM. Paraneoplastic gastrointestinal motor dysfunction: clinical and laboratory characteristics. Am J Gastroenterol. 2001;96:373–9.

    CAS  PubMed  Google Scholar 

  55. Badari A, Farolino D, Nasser E, Mehboob S, Crossland D. A novel approach to paraneoplastic intestinal pseudo-obstruction. Support Care Cancer. 2012;20:425–8.

    PubMed  Google Scholar 

  56. Taverna JA, Babiker HM, Yun S, Bishop MC, Lau-Braunhut S, Meyer PN, et al. The great masquerader of malignancy: chronic intestinal pseudo-obstruction. Biomarker Res. 2014;2:23. https://doi.org/10.1186/s40364-014-0023-y.

    Article  Google Scholar 

  57. Vernino S, Lox PA, Fealey RD, Stewart JD, Farrugia G, Lennon VA. Autoantibodies to ganglionic acetylcholine receptors in autoimmune autonomic neuropathies. N Engl J Med. 2000;343:847–55.

    CAS  PubMed  Google Scholar 

  58. Li Y, Jammoul A, Mente K, Li J, Shields RW, Vernino S, et al. Clinical experience of seropositive ganglionic acetylcholine receptor antibody in a tertiary neurology referral center. Muscle Nerve. 2015;52:386–91.

    CAS  PubMed  Google Scholar 

  59. McKeon A, Lennon VA, Lachance DH, Fealey RD, Pittock SJ. Ganglionic acetylcholine receptor autoantibody: oncological, neurological and serological accompaniments. Arch Neurol. 2009;66(6):735–41.

    PubMed  PubMed Central  Google Scholar 

  60. Zivkovic SA, Ascherman D, Lacomis D. Vasculitis neuropathy-electrodiagnostic findings and association with malignancies. Acta Neurol Scand. 2007;115:432–6.

    CAS  PubMed  Google Scholar 

  61. Oh SJ. Paraneoplastic vasculitis of the peripheral nervous system. Neurol Clin. 1997;15:849–63.

    CAS  PubMed  Google Scholar 

  62. Choi HS, Kim DH, Yang SN, Sung HJ, Choi SJ. A case or paraneoplastic vasculitic neuropathy associated with gastric cancer. Clin Neurol Neurosurg. 2013;115:218–21.

    PubMed  Google Scholar 

  63. Kannan ME, Challa S, Kandadai RM, Uppin MS, Jabeen SA, Borgohain R. Series of paraneoplastic vasculitic neuropathy: a rare, potentially treatable neuropathy. Neurol India. 2015;63:30–4.

    PubMed  Google Scholar 

  64. Vigliani MC, Magistrello M, Polo P, Mutani R, Chiò A. Risk of cancer in patients with Guillain-Barré syndrome (GBS). A population-based study. J Neurol. 2004;251:321–6.

    PubMed  Google Scholar 

  65. Hiew FL, Rajabally YA. Malignancy in Guillain-Barré syndrome: a twelve-year single-center study. J Neurol Sci. 2017;375:275–8.

    PubMed  Google Scholar 

  66. Graus F, Dalmau J. Paraneoplastic neuropathies. Curr Opin Neurol. 2013;26:489–95.

    CAS  PubMed  Google Scholar 

  67. Rajabally YA, Attarian S. Chronic inflammatory demyelinatin polyneuropathy and malignancy: a systematic review. Muscle Nerve. 2018;57(6):875–83.

    PubMed  Google Scholar 

  68. Briani C, Vitaliani R, Grisold W, Honnorat J, Graus F, Antoine JC, et al. Spectrum of paraneoplastic disease associated with lymphoma. Neurology. 2011;76:705–10.

    CAS  PubMed  Google Scholar 

  69. Stern BV, Baehring JM, Kleopa KA, Hochberg FH. Multifocal motor neuropathy with conduction block associated with metastatic lymphoma of the nervous system. J Neurooncol. 2006;78:81–4.

    PubMed  Google Scholar 

  70. Rigamonti A, Lauria G, Stanzani L, Piamarta F, Agostoni E. A case of multifocal motor neuropathy with conduction block associated with gastric and lung adenocarcinoma. J Peripher Nerv Syst. 2012;17:226–8.

    PubMed  Google Scholar 

  71. Antoine JC, Camdessanché JP. Paraneoplastic neuropathies. Curr Opin Neurol. 2017;30(5):513–20.

    CAS  PubMed  Google Scholar 

  72. Antoine JC, Mosnier JF, Absi L, Convers P, Honnorat J, Michel D. Carcinoma associated paraneoplastic peripheral neuropathies in patients with and without anti-onconeural antibodies. J Neurol Neurosurg Psychiatry. 1999;67:7–14.

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Camdessanché JP, Jousserand G, Ferraud K, Vial C, Petiot P, Honnorat J, et al. The pattern and diagnostic criteria of sensory neuronopathy: a case-control study. Brain. 2009;132:1723–33.

    PubMed  PubMed Central  Google Scholar 

  74. Antoine JC, Robert-Varvat F, Maisonobe T, Créange A, Franques J, Mathis S, et al. Testing the validity of a set of diagnostic criteria for sensory neuronopathies: a francophone collaborative study. J Neurol. 2014;261:2093–100.

    PubMed  Google Scholar 

  75. Oki Y, Koike H, Iijima M, Mori K, Hattori N, Katsuno M, et al. Ataxic vs painful form of paraneoplastic neuropathy. Neurology. 2007;69:564–72.

    CAS  PubMed  Google Scholar 

  76. Camdessanché JP, Jousserand G, Franques J, Pouget J, Delmont E, Créange A, et al. A clinical pattern-based etiological diagnostic strategy for sensory neuronopathies: a French collaborative study. J Peripher Nerv Syst. 2012;17:331–40.

    PubMed  Google Scholar 

  77. Oh SJ, Gürtekin Y, Dropcho E, King P, Claussen GC. Anti-Hu antibody neuropathy: a clinical, electrophysiological and pathological study. Clin Neurophysiol. 2005;116:28–34.

    PubMed  Google Scholar 

  78. Storstein A, Raspotnig M, Vitaliani R, Giametto B, Graus F, Grisold W, et al. Prostate cancer, Hu antibodies and paraneoplastic neurological syndromes. J Neurol. 2016;263(5):1001–7.

    CAS  PubMed  Google Scholar 

  79. Honnorat J, Cartalat-Carel S, Ricard D, Camdessanché JP, Carpentier AF, Rogemond V, et al. Onconeural antibodies and tumour type determine survival and neurological symptoms in paraneoplastic neurological syndromes with Hu or CV2/CRMP5 antibodies. J Neurol Neurosurg Psychiatry. 2009;80(4):412–6.

    CAS  PubMed  Google Scholar 

  80. Antoine JC, Honnorat J, Camdessanché JP, Magistris M, Absi L, Mosnier JF, et al. Paraneoplastic anti-CV2 antibodies react with peripheral nerve and are associated with a mixed axonal and demyelinating peripheral neuropathy. Ann Neurol. 2001;49:214–21.

    CAS  PubMed  Google Scholar 

  81. Pittock SJ, Luchinetti CF, Parisi JE, Benarroch EE, Mokri B, Stephan CL, et al. Amphiphysin autoimmunity: paraneoplastic accompaniments. Ann Neurol. 2005;58:96–107.

    PubMed  Google Scholar 

  82. Mélé N, Berzero G, Maisonobe T, Salachas F, Nicolas G, Weiss N, et al. Motor neuron disease of paraneoplastic origin: a rare but treatable condition. J Neurol. 2018;265(7):1590–9.

    PubMed  Google Scholar 

  83. Verschueren A, Gallard J, Boucraut J, Honnorat J, Pouget J, Attarian S. Paraneoplastic subacute lower motor neuron syndrome associated with solid cancer. J Neurol Sci. 2015;358:413–6.

    PubMed  Google Scholar 

  84. Younger DS, Graber J, Hayakawa-Yano Y, Parveen S, Frank M, Darnell RB. Ri/NOVA gene-associated paraneoplastic subacute motor neuronopathy. Muscle Nerve. 2013;47(4):617–8.

    CAS  PubMed  Google Scholar 

  85. Flanagan EP, Sandroni P, Pittock SJ, Inwards DJ, Jones LK. Paraneoplastic lower motor neuronopathy associated with Hodgkin lymphoma. Muscle Nerve. 2012;46:823–7.

    PubMed  Google Scholar 

  86. Vogrig A, Joubert B, Maureille A, Thomas L, Bernard E, Streichenberger N, et al. Motor neuron involvement in anti-Ma2-associated paraneoplastic neurological syndrome. J Neurol. 2018; https://doi.org/10.1007/s00415-018-9143-x.

  87. Flanagan EP, McKeon A, Lennon VA, Kearns J, Weinshenker BG, Krecke KN, et al. Paraneoplastic isolated myelopathy: clinical course and neuroimaging clues. Neurology. 2011;76:2089–95.

    CAS  PubMed  Google Scholar 

  88. Flanagan EP, Keegan BM. Paraneoplastic myelopathy. Neurol Clin. 2013;31:307–18.

    PubMed  Google Scholar 

  89. Cross SA, Salomao DR, Parisi JE, Kryzer TJ, Bradley EA, Mines JA, et al. Paraneoplastic autoimmune optic neuritis with retinitis defined by CRMP-5-IgG. Ann Neurol. 2003;54:38–50.

    PubMed  Google Scholar 

  90. Ducray F, Roos-Weil R, Garcia PY, Slesar J, Heinzlef O, Chatelain D. Devic’s syndrome-like phenotype associated with thymoma and anti-CV2/CRMP5 antibodies. J Neurol Neurosurg Psychiatry. 2007;78(3):325–7.

    PubMed  PubMed Central  Google Scholar 

  91. Pittock SJ, Lennon VA. Aquaporin-4 antibodies in a paraneoplastic context. Arch Neurol. 2008;65(5):629–32.

    PubMed  Google Scholar 

  92. Beauchemin P, Iorio R, Traboulsee AL, Field T, Tinker AV, Carruthers L. Paraneoplastic neuromyelitis optica spectrum disorder: a single center cohort description with two cases of histological validation. Mult Scler Relat Disord. 2018;20:37–42.

    PubMed  Google Scholar 

  93. Sepúlveda M, Sola-Valls N, Escudero D, Rojc B, Barón M, Hernandez-Echebarría L, et al. Clinical profile of patients with paraneoplastic neuromyelitis optica spectrum disorder and aquaporin-4 antibodies. Mult Scler. 2017; https://doi.org/10.1177/1352458517731914.

  94. Fang B, McKeon A, Hinson SR, Kryzer TJ, Pittock SJ, Aksamit AJ, et al. Autoimmune glial fibrillary acidic protein astrocytopathy: a novel meningoencephalomyelitis. JAMA Neurol. 2006;73(11):1297–307.

    Google Scholar 

  95. Flanagan EP, Hinson SR, Lennon VA, Fang B, Aksamit AJ, Morris PP, et al. Glial fibrillary acidic protein immunoglobin G as a biomarker of autoimmune astrocytopathy: analysis of 102 patients. Ann Neurol. 2017;81(2):298–309.

    CAS  PubMed  Google Scholar 

  96. Alexopulos H, Dalakas MC. A critical update on the immunopathogenesis of stiff person syndrome. Eur J Clin Invest. 2010;40(11):1018–25.

    Google Scholar 

  97. Meinck HM, Thompson PD. Stiff man syndrome and related conditions. Mov Disord. 2002;17(5):853–66.

    PubMed  Google Scholar 

  98. Martínez-Hernandez E, Ariño H, McKeon A, Iizuka T, Titulaer MJ, Simabukuro MM, et al. Clinical and immunologic investigations in patients with stiff-person spectrum disorder. JAMA Neurol. 2016;73(6):714–20.

    PubMed  PubMed Central  Google Scholar 

  99. Ariño H, Hötberger R, Gresa-Arribas N, Martínez-Hernandez E, Armangué T, Kruer MC, et al. Paraneoplastic neurological syndromes and glutamic acid decarboxylase antibodies. JAMA Neurol. 2015;72(8):874–81.

    PubMed  PubMed Central  Google Scholar 

  100. Murinson BB, Guarnaccia JB. Stiff-person syndrome with amphiphysin antibodies: distinctive features of a rare disease. Neurology. 2008;71:1955–8.

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Faissner S, Lukas C, Reinacher-Schick A, Tannapfel A, Gold R, Kleiter I. Amphiphysin-positive paraneoplastic myelitis and stiff-person syndrome. Neurol Neuroimmunol Neuroinflamm. 2016;3(6):e285. https://doi.org/10.1212/NXI.0000000000000285.

    Article  PubMed  PubMed Central  Google Scholar 

  102. Vinjam MR, Shanmugarajah P, Ford HL. Ophthalmoplegia heralding the onset of anti-amphiphysin related paraneoplastic stiff person syndrome. J Neurol. 2016;263(5):1017–8.

    PubMed  Google Scholar 

  103. Nguyen-Huu BK, Urban PP, Schreckenberger M, Dieterich M, Werhahn KJ. Antiamphiphysin-positive stiff-person syndrome associated with small cell lung cancer. Mov Disord. 2006;21(8):1285–7.

    CAS  PubMed  Google Scholar 

  104. Butler MH, Hayashi A, Ohkoshi N, Villmann C, Becker CM, Feng G, et al. Autoimmunity to gephyrin in stiff-man syndrome. Neuron. 2000;26:307–12.

    CAS  PubMed  Google Scholar 

  105. McCabe DJH, Turner NC, Chao D, Leff A, Gregson NA, Womersley HJ, et al. Paraneoplastic “stiff person syndrome” with metastasic adenocarcinoma and anti-Ri antibodies. Neurology. 2004;62:1402–4.

    CAS  PubMed  Google Scholar 

  106. Albóndiga-Chindurza A, Riva E, Jiménez-Huete A, Graus F, Franch O. Paraneoplastic stiff person syndrome with small cell carcinoma of the bladder and anti-Ri antibodies. Clin Neurol Neurosurg. 2018;173:194–5.

    Google Scholar 

  107. Hutchinson M, Waters P, McHugh J, Gorman G, O’Riordan S, Connolly S. Progressive encephalomyelitis, rigidity and myoclonus: a novel glycine receptor antibody. Neurology. 2008;71:1291–2.

    CAS  PubMed  Google Scholar 

  108. Carvajal-González A, Leite MI, Waters P, Woodhall M, Coutinho E, Balint B, et al. Glycine receptor antibodies in PERM and related syndromes: characteristics, clinical features and outcomes. Brain. 2014;137:2178–92.

    PubMed  PubMed Central  Google Scholar 

  109. Uehara T, Murai H, Yamasaki R, Kikuchi H, Shigeto H, Ohyagi Y, et al. Thymoma-associated progressive encephalomyelitis, rigidity and myoclonus successfully treated with thymectomy and intravenous immunoglobulin. Eur Neurol. 2011;66:328–30.

    PubMed  Google Scholar 

  110. Clerinx K, Breban T, Schrooten M, Leite MI, Vincent A, Verschakelen J, et al. Progressive encephalomyelitis, rigidity and myoclonus: resolution after thymectomy. Neurology. 2011;76:303–4.

    CAS  PubMed  Google Scholar 

  111. Borellini L, Lanfranconi S, Bonato S, Trezzi I, Franco G, Torretta L, et al. Progressive encephalomyelitis, rigidity and myoclonus associated with anti-GlyR antibodies and Hodgkin’s lymphoma: a case report. Front Neurol. 2017;8:401. https://doi.org/10.3389/fneur.2017.00401.

    Article  PubMed  PubMed Central  Google Scholar 

  112. Balint B, Jarius S, Nagel S, Haberkorn U, Probst C, Blöcker IM, et al. Progressive encephalomyelitis, rigidity and myoclonus: a new variant with DPPX antibodies. Neurology. 2014;82:1521–8.

    CAS  PubMed  Google Scholar 

  113. Tobin WO, Lennon VA, Komorowski L, Probst C, Clardy SL, Aksamit AJ, et al. DPPX potassium channel antibody: frequency, clinical accompaniments, and outcomes in 20 patients. Neurology. 2014;83:1797–803.

    CAS  PubMed  PubMed Central  Google Scholar 

  114. Pittock SJ, Lucchinetti CF, Lennon VA. Anti-neuronal nuclear autoantibody type 2: paraneoplastic accompaniments. Ann Neurol. 2003;53:580–7.

    CAS  PubMed  Google Scholar 

  115. Chan KH, Vernino S, Lennon VA. ANNA-3 anti-neuronal nuclear antibody: marker of lung cancer-related autoimmunity. Ann Neurol. 2001;50:301–11.

    CAS  PubMed  Google Scholar 

  116. Gadoth A, Kryzer TJ, Fryer J, McKeon A, Lennon VA, Pittock SJ. Microtubule associated protein 1B: novel paraneoplastic biomarker. Ann Neurol. 2017;81:266–77.

    CAS  PubMed  Google Scholar 

  117. Bataller L, Wade DF, Graus F, Stacey HD, Rosenfeld MR, Dalmau J. Antibodies to Zic4 in paraneoplastic neurologic disorders and small-cell lung cancer. Neurology. 2004;62(5):778–82.

    CAS  PubMed  Google Scholar 

  118. Pike M. Opsoclonus-myoclonus syndrome. Handb Clin Neurol. 2013;112:1209–11.

    PubMed  Google Scholar 

  119. Klaas JP, Ahlskog E, Pittock SJ, Matsumoto JY, Aksamit AJ, Bartleson JD, et al. Adult-onset opsoclonus-myoclonus syndrome. Arch Neurol. 2012;69(12):1598–607.

    PubMed  Google Scholar 

  120. Pranzatelli MR, Tate ED, McGee NR. Children with opsoclonus-myoclonus syndrome: a cross-sectional study. Front Neurol. 2017;8:468. https://doi.org/10.3389/fneur.2017.00468.

    Article  PubMed  PubMed Central  Google Scholar 

  121. Armangué T, Sabater L, Torres-Vega E, Martínez-Hernández E, Ariño H, Petit-Pedrol M, et al. Clinical and immunological features of opsoclonus-myoclonus syndrome in the era of neuronal cell surface antibodies. JAMA Neurol. 2016;73(4):417–24.

    PubMed  PubMed Central  Google Scholar 

  122. Bataller L, Graus F, Saiz A, Vilchez JJ. Clinical outcome in adult onset idiopathic or paraneoplastic opsoclonus-myoclonus. Brain. 2001;124:437–43.

    CAS  PubMed  Google Scholar 

  123. Shams’ili S, Grefkens J, de Leeuw B, van den Bent M, Hooijkaas H, van der Holt B, et al. Paraneoplastic cerebellar degeneration associated with antineuronal antibodies: analysis of 50 patients. Brain. 2003;126:1409–18.

    PubMed  Google Scholar 

  124. Ducray F, Demarquay G, Graus F, Decullier E, Antoine JC, Giometto B, et al. Seronegative paraneoplastic cerebellar degeneration: the PNS euronetwork experience. Eur J Neurol. 2014;21:731–5.

    CAS  PubMed  Google Scholar 

  125. Peterson K, Rosenblum MK, Kotanides H, Posner JB. Paraneoplastic cerebellar degeneration: a clinical analysis of 55 anti-Yo antibody-positive patients. Neurology. 1992;42:1931–7.

    CAS  PubMed  Google Scholar 

  126. McKeon A, Tracy JA, Pittock SJ, Parisi JE, Klein CJ, Lennon VA. Purkinje-cell cytoplasmatic autoantibody type 1 accompaniments. Arch Neurol. 2011;68(10):1282–9.

    PubMed  Google Scholar 

  127. Sabater L, Höftberger R, Boronat A, Saiz A, Dalmau J, Graus F. Antibody repertoire in paraneoplastic cerebellar degeneration and small cell lung cancer. PLoS One. 2013;8(3):e60438. https://doi.org/10.1371/journal.pone.0060438.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Graus F, Dalmau J, Valldeoriola F, Ferrer I, Reñé R, Marin C, et al. Immunological characterization of a neuronal antibody (anti-Tr) associated with paraneoplastic cerebellar degeneration and Hodgkin’s disease. J Neuroimmunol. 1997;74:55–61.

    CAS  PubMed  Google Scholar 

  129. Bernal F, Shams’ili S, Rojas I, Sanchez-Valle R, Saiz A, Dalmau J, et al. Anti-Tr antibodies as markers of paraneoplastic cerebellar degeneration and Hodgkin’s disease. Neurology. 2003;60:230–4.

    CAS  PubMed  Google Scholar 

  130. Sillevis Smitt P, Kinoshita A, de Leeuw B, Moll W, Coesmans M, Jaarsma D, et al. Paraneoplastic cerebellar ataxia due to autoantibodies against glutamate receptor. N Engl J Med. 2000;342(1):21–7.

    CAS  PubMed  Google Scholar 

  131. Marignier R, Chenevier F, Rogemond V, Sillevis Smitt P, Renoux C, Cavillon G, et al. Metabotropic glutamate receptor type 1 autoantibody-associated cerebellitis: a primary autoimmune disease? Arch Neurol. 2010;67(5):627–30.

    PubMed  Google Scholar 

  132. Dalmau J, Graus F, Villarejo A, Posner JB, Blumenthal D, Thiessen B, et al. Clinical analysis of anti-ma-2 associated encephalitis. Brain. 2004;127:1831–44.

    PubMed  Google Scholar 

  133. Rosenfeld MR, Eichen JG, Wade DF, Posner JB, Dalmau J. Molecular and clinical diversity in paraneoplastic immunity to Ma proteins. Ann Neurol. 2001;50:339–48.

    CAS  PubMed  Google Scholar 

  134. Hoffmann LA, Jarius S, Pellkofer HL, Schueller M, Krumbholz M, Koenig F, et al. Anti-Ma and anti-Ta associated paraneoplastic neurological syndrome: 22 newly diagnosed patients and review of previous cases. J Neurol Neurosurg Psychiatry. 2008;79:767–73.

    CAS  PubMed  Google Scholar 

  135. Ortega-Suero G, Sola-Valls N, Escudero D, Saiz A, Graus F. Síndromes neurológicos paraneoplásicos asociados a anticuerpos anti-Ma y anti-Ma2. Neurologia. 2018;33(1):18–27.

    CAS  PubMed  Google Scholar 

  136. Saiz A, Bruna J, Stourac P, Vigliani MC, Giometto B, Grisold W, et al. Anti-Hu-associated brainstem encephalitis. J Neurol Neurosurg Psychiatry. 2009;80:404–7.

    CAS  PubMed  Google Scholar 

  137. Graus F, Titulaer MJ, Balu R, Benseler S, Bien CG, Cellucci T, et al. A clinical approach to diagnosis of autoimmune encephalitis. Lancet Neurol. 2016;15(4):391–404.

    PubMed  PubMed Central  Google Scholar 

  138. Oyanguren B, Sánchez V, González FJ, de Felipe A, Esteban L, López-Sendón JL, et al. Limbic encephalitis: a clinical-radiological comparison between herpetic and autoimmune etiologies. Eur J Neurol. 2013;20(12):1566–70.

    CAS  PubMed  Google Scholar 

  139. Graus F, Saiz A, Lai M, Bruna J, López F, Sabater L, et al. Neuronal surface antigen antibodies in limbic encephalitis: clinical-immunologic associations. Neurology. 2008;71:930–6.

    CAS  PubMed  PubMed Central  Google Scholar 

  140. Graus F, Escudero D, Oleaga L, Bruna J, Villarejo-Galende A, Ballabriga J, et al. Syndrome and outcome of antibody-negative limbic encephalitis. Eur J Neurol. 2018;25(8):1011–6.

    CAS  PubMed  PubMed Central  Google Scholar 

  141. Gultekin SH, Rosenfeld MR, Voltz R, Eichen J, Posner JB, Dalmau J. Paraneoplastic limbic encephalitis: neurological symptoms, immunological findings and tumour association in 50 patients. Brain. 2000;123:1481–94.

    PubMed  Google Scholar 

  142. Bataller L, Kleopa KA, Wu GF, Rosenfeld MR, Dalmau J. Autoimmune limbic encephalitis in 39 patients: immunophenotypes and outcomes. J Neurol Neurosurg Psychiatry. 2007;78:381–5.

    CAS  PubMed  Google Scholar 

  143. Lai M, Huijbers MGM, Lancaster E, Graus F, Bataller L, Balice-Gordon R, et al. Investigation of LGI1 as the antigen in limbic encephalitis previously attributed to potassium channels: a case series. Lancet Neurol. 2010;9(8):776–85.

    CAS  PubMed  PubMed Central  Google Scholar 

  144. Irani SR, Stagg CJ, Schott JM, Rosenthal CR, Schneider SA, Pettingill P, et al. Faciobrachial dystonic seizures: the influence of immunotherapy on seizure control and prevention of cognitive impairment in a broadening phenotype. Brain. 2013;136:3151–62.

    PubMed  Google Scholar 

  145. Navarro V, Kas A, Apartis E, Chami L, Rogemond V, Levy P, et al. Motor cortex and hippocampus are the two main cortical targets in LGI1-antibody encephalitis. Brain. 2016;139:1079–93.

    PubMed  Google Scholar 

  146. van Sonderen A, Thijs RD, Coenders EC, Jiskoot LC, Sanchez E, de Bruijn MAAM, et al. Anti-LGI1 encephalitis: clinical syndrome and long-term follow-up. Neurology. 2016;87:1–8.

    Google Scholar 

  147. Joubert B, Gobert F, Thomas L, Saint-Martin M, Detestret V, Convers P, et al. Autoimmune episodic ataxia in patients with anti-CASPR2 antibody-associated encephalitis. Neurol Neuroimmunol Neuroinflam. 2017;4:e371. https://doi.org/10.1212/NXI.0000000000000371.

    Article  Google Scholar 

  148. Lai M, Hughes EG, Peng X, Zhou L, Gleichman AJ, Shu H, et al. AMPA receptor antibodies in limbic encephalitis alter synaptic receptor location. Ann Neurol. 2009;65(9):424–34.

    CAS  PubMed  PubMed Central  Google Scholar 

  149. Höftberger R, van Sonderen A, Leypoldt F, Houghton D, Geschwind M, Gelfand J, et al. Encephalitis and AMPA receptor antibodies: novel findings in a case series of 22 patients. Neurology. 2015;84:2403–12.

    PubMed  PubMed Central  Google Scholar 

  150. Joubert B, Kerschen P, Zekeridou A, Detestret V, Rogemond V, Chaffois MO, et al. Antibodies against the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor: case series and review of the literature. JAMA Neurol. 2015;72(10):1163–9.

    PubMed  Google Scholar 

  151. Graus F, Boronat A, Xifró X, Boix M, Svigelj V, García A, et al. The expanding clinical profile of anti-AMPA receptor encephalitis. Neurology. 2010;74:857–9.

    CAS  PubMed  Google Scholar 

  152. Wei YC, Liu CH, Lin JJ, Lin KJ, Huang KL, Lee TH, et al. Rapid progression and brain atrophy in anti-AMPA receptor encephalitis. J Neuroimmunol. 2013;261:129–33.

    CAS  PubMed  Google Scholar 

  153. Bataller L, Galiano R, García-Escrig M, Martínez B, Sevilla T, Blasco R, et al. Reversible paraneoplastic limbic encephalitis associated with antibodies to the AMPA receptor. Neurology. 2010;74:857–9.

    Google Scholar 

  154. Lancaster E, Lai M, Peng X, Hughes E, Constantinescu R, Raizer J, et al. Antibodies to the GABA(B) receptor in limbic encephalitis with seizures: case series and characterization of the antigen. Lancet Neurol. 2010;9(1):67–76.

    CAS  PubMed  Google Scholar 

  155. Jeffery OJ, Lennon VA, Pittock SJ, Gregory JK, Britton JW, McKeon A. GABA(B) receptor autoantibody frequency in service serologic evaluation. Neurology. 2013;81:882–7.

    CAS  PubMed  PubMed Central  Google Scholar 

  156. Höftberger R, Titulaer MJ, Sabater L, Dome B, Rózsás A, Hegedus B, et al. Encephalitis and GABA(B) receptor antibodies: novel findings in a new case series of 20 patients. Neurology. 2013;81:1500–6.

    PubMed  PubMed Central  Google Scholar 

  157. Dogan Onugoren M, Deuretzbacher D, Haensch CA, Hagedorn HJ, Halve S, Isenmann S, et al. Limbic encephalitis due to GABA(B) and AMPA receptor antibodies: a case series. J Neurol Neurosurg Psychiatry. 2015;86:965–72.

    CAS  PubMed  Google Scholar 

  158. Boronat A, Sabater L, Saiz A, Dalmau J, Graus F. GABA(B) receptor antibodies in limbic encephalitis and anti-GAD-associated neurologic disorders. Neurology. 2011;76:795–800.

    CAS  PubMed  PubMed Central  Google Scholar 

  159. Malter MP, Helmstaedter C, Urbach H, Vincent A, Bien CG. Antibodies to glutamic acid decarboxylase define a form of limbic encephalitis. Ann Neurol. 2010;67:470–8.

    PubMed  Google Scholar 

  160. Carr I. The Ophelia syndrome: memory loss in Hodgkin’s disease. Neurology. 1982;1:844–5.

    CAS  Google Scholar 

  161. Lancaster E, Martinez-Hernandez E, Titulaer MJ, Boulos M, Weaver S, Antoine JC, et al. Antibodies to metabotropic glutamate receptor 5 in the Ophelia syndrome. Neurology. 2011;77:1698–701.

    CAS  PubMed  PubMed Central  Google Scholar 

  162. Mat A, Adler H, Merwick A, Chadwick G, Gullo G, Dalmau JO, et al. Ophelia syndrome with metabotropic glutamate receptor 5 antibodies in CSF. Neurology. 2013;42(suppl 5):7–8.

    Google Scholar 

  163. Prüss H, Rothkirch M, Kopp U, Hamer HM, Hagge M, Sterzer P, et al. Limbic encephalitis with mGluR5 antibodies and immunotherapy-responsive prosopagnosia. Neurology. 2014;83:1384–6.

    PubMed  Google Scholar 

  164. Spatola M, Sabater L, Planaguma J, Martinez-Hernandez E, Armangué T, Prüss H et al. Encephalitis with mGluR5 antibodies: symptoms and antibody effects. Neurology 2018;0:1–9. doi:https://doi.org/10.1212/WNL.0000000000005614.

  165. Petit-Pedrol M, Armangue T, Peng X, Bataller L, Cellucci T, Davis R, et al. Encephalitis with refractory seizures, status epilepticus and antibodies to the GABA(A) receptor: a case series, characterisation of the antigen, and analysis of the effects of antibodies. Lancet Neurol. 2014;13:276–86.

    CAS  PubMed  PubMed Central  Google Scholar 

  166. Pettingill P, Kramer HB, Coebergh JA, Pettingill R, Maxwell S, Nibber A, et al. Antibodies to GABA(A) receptor α1 and 2 subunits. Neurology. 2015;84:1233–41.

    CAS  PubMed  PubMed Central  Google Scholar 

  167. Spatola M, Petit-Pedrol M, Simabukuro MM, Armangue T, Castro FJ, Barcelo-Artigues MI, et al. Investigations in GABA(A) receptor antibody-associated encephalitis. Neurology. 2017;88:1012–20.

    CAS  PubMed  PubMed Central  Google Scholar 

  168. Ohkawa T, Satake S, Yokoi N. Identification and characterization of GABA(A) receptor autoantibodies in autoimmune encephalitis. J Neurosci. 2014;34:8151–63.

    PubMed  PubMed Central  Google Scholar 

  169. Simabukuro MM, Petit-Pedrol M, Castro LH, Nitrini R, Lucato L, Zambon AA, et al. GABA(A) receptor and LGI1 antibody encephalitis in a patient with thymoma. Neurol Neuroimmunol Neuroinflamm. 2015;2:e73. https://doi.org/10.1212/NXI.0000000000000073.

    Article  PubMed  PubMed Central  Google Scholar 

  170. Boronat A, Gelfand JM, Gresa-Arribas N, Jeong HY, Walsh M, Roberts K, et al. Encephalitis and antibodies to DPPX, a subunit of Kv4.2 potassium channels. Ann Neurol. 2013;73(1):120–8.

    CAS  PubMed  Google Scholar 

  171. Hara M, Ariño H, Petit-Pedrol M, Sabater L, Titulaer MJ, Martinez-Hernandez E, et al. DPPX antibody-associated encephalitis: main syndrome and antibody effects. Neurology. 2017;88:1340–8.

    CAS  PubMed  PubMed Central  Google Scholar 

  172. Tüzün E, Rossi JE, Karner SF, Centurion AF, Dalmau J. Adenylate kinase 5 autoimmunity in treatment refractory limbic encephalitis. J Neuroimmunol. 2007;186(1–2):177–80.

    PubMed  PubMed Central  Google Scholar 

  173. Do LD, Chanson E, Detestret V, Joubert B, Ducray F, Brugière S, et al. Characteristics in limbic encephalitis with anti-adenylate kinase 5 autoantibodies. Neurology. 2017;88:514–24.

    CAS  PubMed  Google Scholar 

  174. Dalmau J, Tüzün E, Wu H, Masjuan J, Rossi JE, Voloschin A, et al. Paraneoplastic anti-N-methyl-D-aspartate receptor encephalitis associated with ovarian teratoma. Ann Neurol. 2007;61(1):25–36.

    CAS  PubMed  PubMed Central  Google Scholar 

  175. Dalmau J, Gleichman AJ, Hughes EG, Rossi JE, Peng XP, Meizan L, Dessain SK, et al. Anti-NMDA-receptor encephalitis: case series and analysis of the effects of antibodies. Lancet Neurol. 2008;7(12):1091–8.

    CAS  PubMed  PubMed Central  Google Scholar 

  176. Irani SR, Bera K, Waters P, Zuliani L, Maxwell S, Zandi MS, et al. N-methyl-D-aspartate receptor encephalitis: temporal progression of clinical and paraclinical observations in a predominantly non-paraneoplastic disorder of both sexes. Brain. 2010;133:1655–67.

    PubMed  PubMed Central  Google Scholar 

  177. Titulaer MJ, McCracken L, Gabilondo I, Iizuka T, Kawachi I, Bataller L, et al. Late-onset anti-NMDA receptor encephalitis. Neurology. 2013;81:1058–63.

    CAS  PubMed  PubMed Central  Google Scholar 

  178. Viaccoz A, Detestret V, Ducray F, Picard G, Cavillon G, Rogemond V, et al. Clinical specificities of adult male patients with NMDA receptor antibodies encephalitis. Neurology. 2014;82:556–63.

    CAS  PubMed  Google Scholar 

  179. Titulaer MJ, Dalmau J. Seizures as first symptom of anti-NMDA receptor encephalitis are more common in men. Neurology. 2014;82:550–1.

    PubMed  Google Scholar 

  180. Schmitt SE, Pargeon K, Frechette ES, Hirsch L, Dalmau J, Friedman D. Extreme delta brush: a unique EEG pattern in adults with anti-NMDA receptor encephalitis. Neurology. 2012;79:1094–100.

    PubMed  PubMed Central  Google Scholar 

  181. Armangué A, Morís G, Cantarín-Extremera V, Conde CE, Rostasy K, Erro ME, et al. Autoimmune post-herpes simplex encephalitis of adults and teenagers. Neurology. 2015;85:1736–43.

    PubMed  PubMed Central  Google Scholar 

  182. Armangue T, Spatola M, Vlagea A, Mattozzi S, Cárceles-Cordon M, Martinez-Heras E, 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(9):760–72.

    PubMed  PubMed Central  Google Scholar 

  183. Vigliani MC, Honnorat J, Antoine JC, Vitaliani R, Giometto B, Psimaras D, et al. Chorea and related movement disorders of paraneoplastic origin: the PNS EuroNetwork experience. J Neurol. 2011;258:2058–68.

    PubMed  Google Scholar 

  184. Dale RC, Merheb V, Pillai S, Wang D, Cantrill L, Murphy TK, et al. Antibodies to surface dopamine-2 receptor in autoimmune movement and psychiatric disorders. Brain. 2012;135:3453–68.

    PubMed  Google Scholar 

  185. Sabater L, Gaig C, Gelpi E, Bataller L, Lewerenz J, Torres-Vega E, et al. A novel NREM and REM parasomnia with sleep breathing disorder associated with antibodies against IgLON5: a case series, pathological features, and characterization of the antigen. Lancet Neurol. 2014;13(6):575–86.

    CAS  PubMed  PubMed Central  Google Scholar 

  186. Gaig C, Graus F, Compta Y, Högl B, Bataller L, Brüggermann N, et al. Clinical manifestations of the anti-IgLON5 disease. Neurology. 2017;88:1736–43.

    CAS  PubMed  PubMed Central  Google Scholar 

  187. Graus F, Saiz A, Dalmau J. Antibodies and neuronal autoimmune disorders of the CNS. J Neurol. 2010;257:509–17.

    CAS  PubMed  Google Scholar 

  188. McKeon A, Pittock SJ. Paraneoplastic encephalomyelopathies: pathology and mechanisms. Acta Neuropathol. 2011;122:381–400.

    CAS  PubMed  Google Scholar 

  189. Lancaster E, Dalmau J. Neuronal autoantigens-pathogenesis, associated disorders and antibody testing. Nat Rev Neurol. 2012;8(7):380–90.

    CAS  PubMed  PubMed Central  Google Scholar 

  190. Sillevis Smitt PA, Manley GT, Posner JB. Immunization with the paraneoplastic encephalomyelitis antigen HuD does not cause neurological disease in mice. Neurology. 1995;45:1873–8.

    CAS  PubMed  Google Scholar 

  191. Carpentier AF, Rosenfeld MR, Delattre JY, Whalen RG, Posner JB, Dalmau J. DNA vaccination with HuD inhibits growth of a neuroblastoma in mice. Clin Cancer Res. 1998;4(11):2819–24.

    CAS  PubMed  Google Scholar 

  192. Dalmau J, Furneaux HM, Gralla RJ, Kris MG, Posner JB. Detection of the anti-Hu antibody in the serum of patients with small cell lung cancer: a quantitative western blot analysis. Ann Neurol. 1990;27(5):544–52.

    CAS  PubMed  Google Scholar 

  193. Stich O, Jarius S, Kleer B, Rasiah C, Voltz R, Rauer S. Specific antibody index in cerebrospinal fluid from patients with central and peripheral paraneoplastic neurological syndromes. J Neuroimmunol. 2007;183:220–4.

    CAS  PubMed  Google Scholar 

  194. Roberts WK, Deluca IJ, Thomas A, Fak J, Williams T, Buckley N, et al. Patients with lung cancer and paraneoplastic Hu syndrome harbor HuD-specific type 2 CD8+ T cells. J Clin Invest. 2009;119:2042–51.

    CAS  PubMed  PubMed Central  Google Scholar 

  195. Albert ML, Darnell JC, Bender A, Francisco LM, Bhardwaj N, Darnell RB. Tumor-specific killer cells in paraneoplastic cerebellar degeneration. Nat Med. 1998;4:1321–4.

    CAS  PubMed  Google Scholar 

  196. Albert ML, Austin LM, Darnell RB. Detection and treatment of activated T cells in the cerebrospinal fluid of patients with paraneoplastic cerebellar degeneration. Ann Neurol. 2000;47:9–17.

    CAS  PubMed  Google Scholar 

  197. DeLuca I, Blachère NE, Santomasso B, Darnell RB. Tolerance to the neuron-specific paraneoplastic HuD antigen. PLoS One. 2009;4:e5739.

    PubMed  PubMed Central  Google Scholar 

  198. Blachère NE, Orange DE, Santomasso BD, Doerner J, Foo PK, Herre M, et al. T cells targeting a neuronal paraneoplastic antigen mediate tumor rejection and trigger autoimmunity with humoral activation. Eur J Immunol. 2014;44(11):3240–51.

    PubMed  PubMed Central  Google Scholar 

  199. Gebauer C, Pignolet B, Yshii L, Mauré E, Bauer J, Liblau R. CD4+ and CD8+ T cells are both needed to induce paraneoplastic neurological disease in a mouse model. Oncoimmunology. 2016;6(2):e1260212. https://doi.org/10.1080/2162402X.2016.1260212.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  200. Sommer C, Weishaupt A, Brinkhoff J, Biko L, Wessig C, Gold R, et al. Paraneoplastic stiff-person syndrome: passive transfer to rats by means of IgG antibodies to amphiphysin. Lancet. 2005;365:1406–11.

    CAS  PubMed  Google Scholar 

  201. Geis C, Weishaupt A, Hallermann S, Grünewald B, Wessig C, Wultsch T, et al. Stiff person syndrome-associated autoantibodies to amphiphysin mediate reduced GABAergic inhibition. Brain. 2010;133:3166–80.

    PubMed  Google Scholar 

  202. Manto MU, Laute M-A, Aguera M, Rogemond V, Pandolfo M, Honnorat J. Effects of anti-glutamic acid decarboxylase antibodies associated with neurological diseases. Ann Neurol. 2007;61:544–51.

    CAS  PubMed  Google Scholar 

  203. Manto MU, Hampe CS, Rogemond V, Honnorat J. Respective implications of glutamate decarboxylase antibodies in stiff person syndrome and cerebellar ataxia. Orphanet J Rare Dis. 2011;6:3. https://doi.org/10.1186/1750-1172-6-3.

    Article  PubMed  PubMed Central  Google Scholar 

  204. 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;97:839–87.

    CAS  PubMed  PubMed Central  Google Scholar 

  205. Weiner AL, Vieira L, McKay CA, Bayer MJ. Ketamine abuser presenting to the emergency department: a case series. J Emerg Med. 2000;18(4):447–51.

    CAS  PubMed  Google Scholar 

  206. Belforte JE, Zsiros V, Sklar ER, Jiang Z, Yu G, Li Y, et al. Postnatal NMDA receptor ablation in corticolimbic interneuron confers schizophrenia-like phenotypes. Nat Neurosci. 2010;13:76–83.

    CAS  PubMed  Google Scholar 

  207. Morante-Redolat JM, Gorostidi-Pagola A, Piquer-Sirerol S, Sáenz A, Poza JJ, Galán J, et al. Mutations in the LGI1/Epitempin gene on 19q24 cause autosomal dominant lateral epilepsy. Hum Mol Genet. 2002;11:1119–28.

    CAS  PubMed  Google Scholar 

  208. Strauss KA, Puffenberger EG, Huentelman MJ, Gottlieb S, Dobrin SE, Parod JM, et al. Recessive symptomatic focal epilepsy and mutant contactin-associated protein-like 2. N Engl J Med. 2006;354:1370–7.

    CAS  PubMed  Google Scholar 

  209. Bettler B, Kaupmann K, Mosbacher J, Gassmann M. Molecular structure and physiological function of GABA(B) receptors. Physiol Rev. 2004;84:835–67.

    CAS  PubMed  Google Scholar 

  210. Shiang R, Ryan SG, Zhu YZ, Hahn AF, O’Connel P, Wasmuth JJ. Mutations in the 𝛼1 subunit of the inhibitory glycine receptor cause the dominant neurological disorder, hyperekplexia. Nat Genet. 1993;5:351–8.

    Google Scholar 

  211. Hughes EG, Peng X, Gleichman AJ, Lai M, Zhou L, Tsou R, et al. Cellular and synaptic mechanisms of anti-NMDA receptor encephalitis. J Neurosci. 2010;30:5866–75.

    CAS  PubMed  PubMed Central  Google Scholar 

  212. Peng X, Hughes EG, Moscato EH, Parsons TD, Dalmau J, Balice-Gordon RJ. Cellular plasticity induced by anti-alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor encephalitis antibodies. Ann Neurol. 2015;77:381–98.

    CAS  PubMed  PubMed Central  Google Scholar 

  213. 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 

  214. Ohkawa T, Fukata Y, Yamasaki M, Miyazaki T, Yokoi N, Takashima H, et al. Autoantibodies to epilepsy-related LGI1 in limbic encephalitis neutralize LGI1-ADAM22 interaction and reduce synaptic AMPA receptors. J Neurosci. 2013;33:18161–74.

    CAS  PubMed  PubMed Central  Google Scholar 

  215. Pinatel D, Hivert B, Boucraut J, Saint-Martin M, Rogemond V, Zoupi L, et al. Inhibitory axons are targeted in hippocampal cell culture by anti-Caspr2 autoantibodies associated with limbic encephalitis. Front Cell Neurosci. 2015;9:265. https://doi.org/10.3389/fncel.2015.00265.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  216. Piepgras J, Holtje M, Michel K, Li Q, Otto C, Drenckhahn C, et al. Anti-DPPX encephalitis: pathogenic effects of antibodies on gut and brain neurons. Neurology. 2015;85:890–7.

    CAS  PubMed  PubMed Central  Google Scholar 

  217. de Graaff E, Maat P, Hulsenboom E, van den Berg R, van den Bent M, Demmers J, et al. Identification of delta/notch-like epidermal growth factor-related receptor as the Tr antigen in paraneoplastic cerebellar degeneration. Ann Neurol. 2012;71:815–24.

    PubMed  Google Scholar 

  218. Planaguma J, Leypoldt F, Mannara F, Gutierrez-Cuesta J, Martin-Garcia E, Aguilar E, et al. Human N-methyl D-aspartate receptor antibodies alter memory and behaviour in mice. Brain. 2015;138:94–109.

    PubMed  Google Scholar 

  219. de Graaf MT, Beukelaar JW, Haasnoot GW, Levering WH, Rogemond V, Didelot A, et al. HLDA-DQ2+ individuals are susceptible to Hu-Ab associated paraneoplastic neurological syndromes. J Neuroimmunol. 2010;226:147–9.

    PubMed  Google Scholar 

  220. Hillary RP, Ollila HM, Lin L, Detestret V, Rogemond V, Picard G, et al. Complex HLA association in paraneoplastic cerebellar ataxia with anti-Yo antibodies. J Neuroimmunol. 2018;315:28–32.

    CAS  PubMed  Google Scholar 

  221. van Sonderen A, Roelen DL, Stoop JA, Verdijk RM, Haasnoot GW, Thijs RD, et al. Anti-LGI1 encephalitis is strongly associated with HLA-DR7 and HLA-DRB4. Ann Neurol. 2017;81(2):193–8.

    PubMed  Google Scholar 

  222. Kim T-J, Lee S-T, Moon J, Sunwoo J-S, Byun J-L, Lim J-A, et al. Anti-LGI1 encephalitis is associated with unique HLA subtypes. Ann Neurol. 2017;81(2):183–92.

    CAS  PubMed  Google Scholar 

  223. Mueller SH, Färber A, Prüss H, Melzer N, Golombeck KS, Kümpfel T, et al. Genetic predisposition in anti-LGI1 and anti-NMDA receptor encephalitis. Ann Neurol. 2018;83(4):863–9.

    CAS  PubMed  Google Scholar 

  224. Zoccarato M, Gastaldi M, Zuliani L, Biagioli T, Brogi M, Bernardi G, et al. Diagnostic of paraneoplastic neurological syndromes. Neurol Sci. 2017;38(2):237–42.

    PubMed  Google Scholar 

  225. Baumgartner A, Rauer S, Mader I, Meyer PT. Cerebral FDG-PET and MRI findings in autoimmune limbic encephalitis: correlation with autoantibody types. J Neurol. 2013;260:2744–53.

    PubMed  Google Scholar 

  226. Heine J, Prüss H, Bartsch T, Ploner CJ, Paul F, Finke C. Imaging of autoimmune encephalitis-relevance for clinical practice and hippocampal function. Neuroscience. 2015;309:68–83.

    CAS  PubMed  Google Scholar 

  227. Leypoldt F, Buchert R, Kleiter I, Marienhagen J, Gelderblom M, Magnus T, et al. Fluorodeoxyglucose positron emission tomography in anti-N-methyl-D-aspartate recetr encephalitis: a distinct pattern of disease. J Neurol Neurosurg Psychiatry. 2012;83(7):681–6.

    PubMed  Google Scholar 

  228. Psimaras D, Carpentier AF, Rossi C. Cerebrospinal fluid study in paraneoplastic syndromes. J Neurol Neurosurg Psychiatry. 2010;81:42–5.

    CAS  PubMed  Google Scholar 

  229. Höftberger R, Dalmau J, Graus F. Clinical neuropathology practice guide 5-2012: updated guideline for the diagnosis of anti-neuronal antibodies. Clin Neuropathol. 2012;31(5):337–41.

    PubMed  PubMed Central  Google Scholar 

  230. Gresa-Arribas N, Titulaer MJ, Torrents A, Aguilar E, McCraken L, Leypoldt F, et al. Diagnosis and significance of antibody titers in anti-NMDA receptor encephalitis, a retrospective study. Lancet Neurol. 2014;13(2):167–77.

    CAS  PubMed  Google Scholar 

  231. Alexopoulos H, Kosmidis ML, Dalmau J, Dalakas MC. Paraneoplastic anti-NMDAR encephalitis: long term follow-up reveals persistent serum antibodies. J Neurol. 2011;258:1568–70.

    PubMed  Google Scholar 

  232. Hansen H-C, Klingbeil C, Dalmau J, Li W, Weissbrich B, Wandinger K-P. Persistent intrathecal antibody synthesis 15 years after recovering from anti-N-methyl-D-aspartate receptor encephalitis. JAMA Neurol. 2013;70(1):117–9.

    PubMed  PubMed Central  Google Scholar 

  233. Titulaer MJ, Soffietti R, Dalmau J, Gilhus NE, Giometto B, Graus F, et al. Screening for tumours in paraneoplastic syndromes: report of an EFNS Task Force. Eur J Neurol. 2011;18(1):19–e3. https://doi.org/10.1111/j.1468-1331.2010.03220.x.

    Article  CAS  PubMed  Google Scholar 

  234. Antoine JC, Camdessanché JP. Treatment options in paraneoplastic disorders of the peripheral nervous system. Curr Treat Options Neurol. 2013;15(2):210–23.

    PubMed  Google Scholar 

  235. Dalmau J, Lancaster E, Martinez-Hernandez E, Rosenfeld MR, Balice-Gordon R. Clinical experience and laboratory investigations in patients with anti-NMDA-R encephalitis. Lancet Neurol. 2011;10(1):63–74.

    CAS  PubMed  PubMed Central  Google Scholar 

  236. Keime-Guibert F, Graus F, Fleury A, René R, Honnorat J, Broet P, et al. Treatment of paraneoplastic neurological syndromes with antineural antibodies (anti-Hu, anti-Yo) with combination of immunoglobulins, cyclophosphamide and methylprednisolone. J Neurol Neurosurg Psychiatry. 2000;68:479–82.

    CAS  PubMed  PubMed Central  Google Scholar 

  237. Vernino S, O’Neill BP, Marks RS, O’Fallon JR, Kimmel DW. Immunomodulatory treatment trial for paraneoplastic neurological disorders. Neuro Oncol. 2004;6(1):55–62.

    PubMed  PubMed Central  Google Scholar 

  238. Shams’ili S, de Beukelaar J, Gratama JW, Hooijkaas H, van den Bent M, van’t Veer M, et al. An uncontrolled trial of rituximab for antibody associated paraneoplastic syndromes. J Neurol. 2006;253:16–20.

    PubMed  Google Scholar 

  239. Lee W-J, Lee S-T, Byun J-I, Sunwoo J-S, Kim T-J, Lim J-A, et al. Rituximab treatment for autoimmune limbic encephalitis in an institutional cohort. Neurology. 2016;86:1–9.

    Google Scholar 

  240. Berzero G, Karantoni E, Dehais C, Ducray F, Thomas L, Picard G, et al. Early intravenous immunoglobulin treatment in paraneoplastic neurological syndromes with onconeural antibodies. J Neurol Neurosurg Psychiatry. 2018;89:789–92.

    PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Jérôme Honnorat .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2019 Springer Nature Switzerland AG

About this chapter

Check for updates. Verify currency and authenticity via CrossMark

Cite this chapter

Muñiz-Castrillo, S., Honnorat, J. (2019). Paraneoplastic Neurological Syndromes. In: Mitoma, H., Manto, M. (eds) Neuroimmune Diseases. Contemporary Clinical Neuroscience. Springer, Cham. https://doi.org/10.1007/978-3-030-19515-1_14

Download citation

Publish with us

Policies and ethics