Skip to main content
Log in

S1-Leitlinie: Diagnostik bei Polyneuropathien

Leitlinie der Deutschen Gesellschaft für Neurologie

S1 guideline: diagnosis of polyneuropathies

Guideline of the German Neurological Society

  • Leitlinie
  • Published:
DGNeurologie Aims and scope

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

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Abb. 1

Notes

  1. Medikamenteninduzierte Polyneuropathien: Neben den seit langem bekannten, potenziell polyneuropathieinduzierenden Medikamenten (Chemotherapeutika, INH [Isonikotinsäurehydrazid], Thalidomid, Chloroquin usw.) wurde in den letzten Jahren die Entstehung einer Polyneuropathie bei einigen Medikamenten beschrieben, für die das bisher nicht bekannt war. Statine verursachen nach langer Anwendung selten sensible und sensomotorische Polyneuropathien, die nach ihrem Absetzen reversibel sind [19]. Bortezomib (Velcade®), ein Proteasominhibitor, der bei der Therapie des multiplen Myeloms eingesetzt wird, verursacht schmerzhafte sensomotorische Polyneuropathien, die nur teilweise reversibel sind [20]. Linezolid, ein Antibiotikum aus der Oxazolidinongruppe, verursacht nach längerer Anwendung eine schmerzhafte sensomotorische Polyneuropathie und eine toxische Optikusneuropathie [21, 22]. Auch Medikamente, die in der Therapie von Neuropathien eingesetzt werden wie Rituximab, Tumor-Nekrose-Faktor-Blocker und auch Immuncheckpointinhibitoren wie Ipilimumab, Nivolumab oder Pembrolizumab können in Einzelfällen (immunvermittelte) Neuropathien auslösen [23].

  2. Die Tabellen (Tab. 8910 und 11) erheben keinen Anspruch auf Vollständigkeit. Monatlich werden Mutationen in neuen Genen als Ursache von Neuropathien identifiziert, NGS-Panels unterscheiden sich in ihrer Zusammensetzung laborabhängig. Aktuell erschweren die Limitationen in der Abrechnung NGS-basierter Diagnostik nach EBM (evidenzbasierte Medizin) den medizinisch sinnvollen Einsatz von NGS zur molekularen Ursachenklärung

  3. Hochgestelltes a (Parametera): Jeder Nerv, der eines der Kriterien a–g erfüllt

Literatur

  1. Doppler K, Sommer C (2017) Neue Entität der Paranodopathien: eine Ziel-struktur mit therapeutischen Konsequenzen. Aktuelle Neurol 44:194–199

    Article  Google Scholar 

  2. Querol L, Devaux J, Rojas-Garcia R, Illa I (2017) Autoantibodies in chronic inflammatory neuropathies: diagnostic and therapeutic implications. Nat Rev Neurol 13(9):533–547

    Article  CAS  PubMed  Google Scholar 

  3. Eggermann K, Gess B, Hausler M, Weis J, Hahn A, Kurth I (2018) Hereditary neuropathies. Dtsch Arztebl Int 115(6):91–97

    PubMed  PubMed Central  Google Scholar 

  4. Adams D, Gonzalez-Duarte A, O’Riordan WD, Yang CC, Ueda M, Kristen AV et al (2018) Patisiran, an RNAi therapeutic, for hereditary transthyretin amyloidosis. N Engl J Med 379(1):11–21

    Article  CAS  PubMed  Google Scholar 

  5. Benson MD, Waddington-Cruz M, Berk JL, Polydefkis M, Dyck PJ, Wang AK et al (2018) Inotersen treatment for patients with hereditary transthyretin amyloidosis. N Engl J Med 379(1):22–31

    Article  CAS  PubMed  Google Scholar 

  6. Dyck P, Thomas P, Griffin J, Low P, Poduslo J (1993) Peripheral neuropathy, 3. Aufl. W.B. Saunders Company, Philadelphia

    Google Scholar 

  7. Mendell J, Kissel J, Cornblath D (2001) Diagnosis and management of peripheral nerve disorders. Oxford University Press, Oxford

    Google Scholar 

  8. Neundörfer B, Heuß D (2006) Polyneuropathien, 1. Aufl. Thieme, Stuttgart, S 129

    Google Scholar 

  9. Pestronk A Neuromuscular Disorders. http://neuromuscular.wustl.edu/

  10. Willison HJ, Jacobs BC, van Doorn PA (2016) Guillain-Barre syndrome. Lancet 388(10045):717–727

    Article  PubMed  Google Scholar 

  11. Albert SM, Wohl MA, Rechtman AM (1958) Serum neuropathy following routine use of tetanus antitoxin; report of five cases. Am Pract Dig Treat 9(8):1249–1252

    CAS  PubMed  Google Scholar 

  12. Baust W, Meyer D, Wachsmuth W (1979) Peripheral neuropathy after administration of tetanus toxoid. J Neurol 222(2):131–133

    Article  CAS  PubMed  Google Scholar 

  13. Blumstein GI, Kreithen H (1966) Peripheral neuropathy following tetanus toxoid administration. JAMA 198(9):1030–1031

    Article  CAS  PubMed  Google Scholar 

  14. Pollard JD, Selby G (1978) Relapsing neuropathy due to tetanus toxoid. Report of a case. J Neurol Sci 37(1–2):113–125

    Article  CAS  PubMed  Google Scholar 

  15. Reinstein L, Pargament JM, Goodman JS (1982) Peripheral neuropathy after multiple tetanus toxoid injections. Arch Phys Med Rehabil 63(7):332–334

    CAS  PubMed  Google Scholar 

  16. Prasad PL, Rai PL (2018) Prospective study of diphtheria for neurological complications. J Pediatr Neurosci 13(3):313–316

    Article  PubMed  PubMed Central  Google Scholar 

  17. Rattananan W, Thaisetthawatkul P, Dyck PJ (2014) Postsurgical inflammatory neuropathy: a report of five cases. J Neurol Sci 337(1–2):137–140

    Article  PubMed  Google Scholar 

  18. Unfallversicherung DDG (2018) BK 1317 – Polyneuropathie oder Enzephalopathie durch organische Lösungsmittel oder deren Gemische (BK-Report 1/2018). https://www.dguv.de/ifa/publikationen/reports-download/reports-2018/bk-report-1-2018/index.jsp

  19. de Langen JJ, van Puijenbroek EP (2006) HMG-CoA-reductase inhibitors and neuropathy: reports to the Netherlands Pharmacovigilance Centre. Neth J Med 64(9):334–338

    PubMed  Google Scholar 

  20. Richardson PG, Briemberg H, Jagannath S, Wen PY, Barlogie B, Berenson J et al (2006) Frequency, characteristics, and reversibility of peripheral neuropathy during treatment of advanced multiple myeloma with bortezomib. J Clin Oncol 24(19):3113–3120

    Article  CAS  PubMed  Google Scholar 

  21. Bressler AM, Zimmer SM, Gilmore JL, Somani J (2004) Peripheral neuropathy associated with prolonged use of linezolid. Lancet Infect Dis 4(8):528–531

    Article  PubMed  Google Scholar 

  22. Rucker JC, Hamilton SR, Bardenstein D, Isada CM, Lee MS (2006) Linezolid-associated toxic optic neuropathy. Neurology 66(4):595–598

    Article  CAS  PubMed  Google Scholar 

  23. Gu Y, Menzies AM, Long GV, Fernando SL, Herkes G (2017) Immune mediated neuropathy following checkpoint immunotherapy. J Clin Neurosci 45:14–17

    Article  PubMed  Google Scholar 

  24. Reinhardt F, Wetzel T, Vetten S, Radespiel-Troger M, Hilz MJ, Heuss D et al (2000) Peripheral neuropathy in chronic venous insufficiency. Muscle Nerve 23(6):883–887

    Article  CAS  PubMed  Google Scholar 

  25. Tankisi H, Pugdahl K, Johnsen B, Fuglsang-Frederiksen A (2007) Correlations of nerve conduction measures in axonal and demyelinating polyneuropathies. Clin Neurophysiol 118(11):2383–2392

    Article  PubMed  Google Scholar 

  26. Buchner H, Schönlau L, Ferbert A (2018) Neurographie des N. Suralis. Klin Neurophysiol 49:188–207

    Article  Google Scholar 

  27. Stöhr M (1998) Atlas der klinischen Elektromyographie und Neurographie, 4. Aufl. Kohlhammer, Stuttgart

    Google Scholar 

  28. Bromberg MB, Franssen H (2015) Practical rules for electrodiagnosis in suspected multifocal motor neuropathy. J Clin Neuromuscul Dis 16(3):141–152

    Article  PubMed  Google Scholar 

  29. Heuß D, Müller-Felber W, Schulte-Mattler W, Dengler R, Toyka K, Sommer C (2002) Diagnostik und Therapie der multifokalen motorischen Neuropathie (MMN). Bundeseinheitliche Konsensuspapiere der Muskelzentren im Auftrag der Deutschen Gesellschaft für Muskelkranke e. V. (DGM). Nervenheilkunde (21):100–107

  30. Joint Task Force of the E, the PNS (2010) European Federation of Neurological Societies/Peripheral Nerve Society guideline on management of multifocal motor neuropathy. Report of a joint task force of the European Federation of Neurological Societies and the Peripheral Nerve Society—first revision. J Peripher Nerv Syst 15(4):295–301

    Article  Google Scholar 

  31. Akaza M, Kanouchi T, Inaba A, Numasawa Y, Irioka T, Mizusawa H et al (2011) Motor nerve conduction study in cauda equina with high-voltage electrical stimulation in multifocal motor neuropathy and amyotrophic lateral sclerosis. Muscle Nerve 43(2):274–282

    Article  PubMed  Google Scholar 

  32. Jaspert A, Claus D, Grehl H, Kerling F, Neundorfer B (1995) Value of proximal conduction block study in diagnosis of inflammatory neuropathies. Nervenarzt 66(6):445–454

    CAS  PubMed  Google Scholar 

  33. Atherton DD, Facer P, Roberts KM, Misra VP, Chizh BA, Bountra C et al (2007) Use of the novel Contact Heat Evoked Potential Stimulator (CHEPS) for the assessment of small fibre neuropathy: correlations with skin flare responses and intra-epidermal nerve fibre counts. BMC Neurol 7:21

    Article  PubMed  PubMed Central  Google Scholar 

  34. Obermann M, Katsarava Z, Esser S, Sommer C, He L, Selter L et al (2008) Correlation of epidermal nerve fiber density with pain-related evoked potentials in HIV neuropathy. Pain 138(1):79–86

    Article  PubMed  Google Scholar 

  35. Casanova-Molla J, Grau-Junyent JM, Morales M, Valls-Sole J (2011) On the relationship between nociceptive evoked potentials and intraepidermal nerve fiber density in painful sensory polyneuropathies. Pain 152(2):410–418

    Article  PubMed  Google Scholar 

  36. Rage M, Van Acker N, Knaapen MW, Timmers M, Streffer J, Hermans MP et al (2011) Asymptomatic small fiber neuropathy in diabetes mellitus: investigations with intraepidermal nerve fiber density, quantitative sensory testing and laser-evoked potentials. J Neurol 258:1852

    Article  PubMed  Google Scholar 

  37. Tavakoli M, Marshall A, Pitceathly R, Fadavi H, Gow D, Roberts ME et al (2010) Corneal confocal microscopy: a novel means to detect nerve fibre damage in idiopathic small fibre neuropathy. Exp Neurol 223(1):245–250

    Article  PubMed  Google Scholar 

  38. Wilbourn A (2000) Multiple mononeuropathies and polyneuropathies. In: Levin K, Luders HO (Hrsg) Comprehensive clinical neurophysiology. W.B. Saunders Company, Philadelphia, London, New York, St. Louis, Sydney, Toronto, S 215–233

    Google Scholar 

  39. Hamel J, Logigian EL (2018) Acute nutritional axonal neuropathy. Muscle Nerve 57(1):33–39

    Article  PubMed  Google Scholar 

  40. Pestronk A Neuromuscular. Neuromuscular Disease Center Washington University, St. Louis ( https://neuromuscular.wustl.edu/autonomic.html .)

  41. van den Berg B, van der Eijk AA, Pas SD, Hunter JG, Madden RG, Tio-Gillen AP et al (2014) Guillain-Barre syndrome associated with preceding hepatitis E virus infection. Neurology 82(6):491–497

    Article  CAS  PubMed  Google Scholar 

  42. Dirlikov E, Major CG, Medina NA, Lugo-Robles R, Matos D, Munoz-Jordan JL et al (2018) Clinical features of Guillain-barre syndrome with vs without Zika virus infection, Puerto Rico, 2016. JAMA Neurol 75(9):1089–1097

    Article  PubMed  PubMed Central  Google Scholar 

  43. Goodfellow JA, Willison HJ (2018) Gangliosides and autoimmune peripheral nerve diseases. Prog Mol Biol Transl Sci 156:355–382

    Article  PubMed  Google Scholar 

  44. Klehmet J, Marschenz S, Ruprecht K, Wunderlich B, Buttner T, Hiemann R et al (2018) Analysis of anti-ganglioside antibodies by a line immunoassay in patients with chronic-inflammatory demyelinating polyneuropathies (CIDP). Clin Chem Lab Med 56(6):919–926

    Article  CAS  PubMed  Google Scholar 

  45. Fehmi J, Scherer SS, Willison HJ, Rinaldi S (2018) Nodes, paranodes and neuropathies. J Neurol Neurosurg Psychiatry 89(1):61–71

    Article  PubMed  Google Scholar 

  46. Uncini A, Vallat JM (2018) Autoimmune nodo-paranodopathies of peripheral nerve: the concept is gaining ground. J Neurol Neurosurg Psychiatry 89(6):627–635

    Article  PubMed  Google Scholar 

  47. Binks S, Varley J, Lee W, Makuch M, Elliott K, Gelfand JM et al (2018) Distinct HLA associations of LGI1 and CASPR2-antibody diseases. Brain 141(8):2263–2271

    Article  PubMed  PubMed Central  Google Scholar 

  48. Sghirlanzoni A, Pareyson D, Lauria G (2005) Sensory neuron diseases. Lancet Neurol 4(6):349–361

    Article  CAS  PubMed  Google Scholar 

  49. Antoine JC, Boutahar N, Lassabliere F, Reynaud E, Ferraud K, Rogemond V et al (2015) Antifibroblast growth factor receptor 3 antibodies identify a subgroup of patients with sensory neuropathy. J Neurol Neurosurg Psychiatry 86(12):1347–1355

    Article  PubMed  Google Scholar 

  50. Gibbons CH, Freeman R (2015) Treatment-induced neuropathy of diabetes: an acute, iatrogenic complication of diabetes. Brain 138(Pt 1):43–52

    Article  PubMed  Google Scholar 

  51. Mohseni S (2001) Hypoglycemic neuropathy. Acta Neuropathol 102(5):413–421

    CAS  PubMed  Google Scholar 

  52. Ozaki K, Sano T, Tsuji N, Matsuura T, Narama I (2010) Insulin-induced hypoglycemic peripheral motor neuropathy in spontaneously diabetic WBN/Kob rats. Comp Med 60(4):282–287

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Kumar N (2014) Neurologic aspects of cobalamin (B12) deficiency. Handb Clin Neurol 120:915–926

    Article  PubMed  Google Scholar 

  54. Santos-Garcia D, de la Fuente-Fernandez R, Valldeoriola F, Palasi A, Carrillo F, Grande M et al (2012) Polyneuropathy while on duodenal levodopa infusion in Parkinson’s disease patients: we must be alert. J Neurol 259(8):1668–1672

    Article  PubMed  Google Scholar 

  55. Prasse A, Katic C, Germann M, Buchwald A, Zissel G, Muller-Quernheim J (2008) Phenotyping sarcoidosis from a pulmonary perspective. Am J Respir Crit Care Med 177(3):330–336

    Article  CAS  PubMed  Google Scholar 

  56. Ufer F, Friese M (2015) Neurosarkoidose. Aktuelle Neurol 42:218–227

    Article  Google Scholar 

  57. Nobile-Orazio E, Bianco M, Nozza A (2017) Advances in the treatment of paraproteinemic neuropathy. Curr Treat Options Neurol 19(12):43

    Article  PubMed  Google Scholar 

  58. Lozeron P, Mariani LL, Dodet P, Beaudonnet G, Theaudin M, Adam C et al (2018) Transthyretin amyloid polyneuropathies mimicking a demyelinating polyneuropathy. Neurology 91(2):e143–e152

    Article  CAS  PubMed  Google Scholar 

  59. Vlam L, van der Pol WL, Cats EA, Straver DC, Piepers S, Franssen H et al (2011) Multifocal motor neuropathy: diagnosis, pathogenesis and treatment strategies. Nat Rev Neurol 8(1):48–58

    Article  CAS  PubMed  Google Scholar 

  60. van Sonderen A, Arino H, Petit-Pedrol M, Leypoldt F, Kortvelyessy P, Wandinger KP et al (2016) The clinical spectrum of Caspr2 antibody-associated disease. Neurology 87(5):521–528

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Devaux JJ, Miura Y, Fukami Y, Inoue T, Manso C, Belghazi M et al (2016) Neurofascin-155 IgG4 in chronic inflammatory demyelinating polyneuropathy. Neurology 86(9):800–807

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Doppler K, Appeltshauser L, Wilhelmi K, Villmann C, Dib-Hajj SD, Waxman SG et al (2015) Destruction of paranodal architecture in inflammatory neuropathy with anti-contactin-1 autoantibodies. J Neurol Neurosurg Psychiatry 86(7):720–728

    Article  PubMed  Google Scholar 

  63. Mathey EK, Garg N, Park SB, Nguyen T, Baker S, Yuki N et al (2017) Autoantibody responses to nodal and paranodal antigens in chronic inflammatory neuropathies. J Neuroimmunol 309:41–46

    Article  CAS  PubMed  Google Scholar 

  64. Miura Y, Devaux JJ, Fukami Y, Manso C, Belghazi M, Wong AH et al (2015) Contactin 1 IgG4 associates to chronic inflammatory demyelinating polyneuropathy with sensory ataxia. Brain 138(Pt 6):1484–1491

    Article  PubMed  PubMed Central  Google Scholar 

  65. Querol L, Nogales-Gadea G, Rojas-Garcia R, Diaz-Manera J, Pardo J, Ortega-Moreno A et al (2014) Neurofascin IgG4 antibodies in CIDP associate with disabling tremor and poor response to IVIg. Neurology 82(10):879–886

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Heuß D (2006) Untersuchungen des Liquor cerebrospinalis. In: Neundörfer B, Heuß D (Hrsg) Polyneuropathien. Referenzreihe Neurologie RRN. Thieme, Stuttgart, S 32–35 (Tafel IX)

    Google Scholar 

  67. Bourque PR, Breiner A, Moher D, Brooks J, Hegen H, Deisenhammer F et al (2019) Adult CSF total protein: Higher upper reference limits should be considered worldwide. A web-based survey. J Neurol Sci 396:48–51

    Article  CAS  PubMed  Google Scholar 

  68. Allen JA, Lewis RA (2015) CIDP diagnostic pitfalls and perception of treatment benefit. Neurology 85(6):498–504

    Article  CAS  PubMed  Google Scholar 

  69. Kindstrand E, Nilsson BY, Hovmark A, Pirskanen R, Asbrink E (1997) Peripheral neuropathy in acrodermatitis chronica atrophicans—a late Borrelia manifestation. Acta Neurol Scand 95(6):338–345

    Article  CAS  PubMed  Google Scholar 

  70. Rauer S (2018) Neuroborreliose. https://www.dgn.org/leitlinien/3567-ll-030-071-2018-neuroborreliose

  71. Antoniadi T, Buxton C, Dennis G, Forrester N, Smith D, Lunt P et al (2015) Application of targeted multi-gene panel testing for the diagnosis of inherited peripheral neuropathy provides a high diagnostic yield with unexpected phenotype-genotype variability. BMC Med Genet 16:84

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Baets J, Deconinck T, De Vriendt E, Zimon M, Yperzeele L, Van Hoorenbeeck K et al (2011) Genetic spectrum of hereditary neuropathies with onset in the first year of life. Brain 134(Pt 9):2664–2676

    Article  PubMed  PubMed Central  Google Scholar 

  73. Bansagi B, Griffin H, Whittaker RG, Antoniadi T, Evangelista T, Miller J et al (2017) Genetic heterogeneity of motor neuropathies. Neurology 88(13):1226–1234

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Hoeijmakers JG, Faber CG, Lauria G, Merkies IS, Waxman SG (2012) Small-fibre neuropathies—advances in diagnosis, pathophysiology and management. Nat Rev Neurol 8(7):369–379

    Article  CAS  PubMed  Google Scholar 

  75. Kurth I (2015) Sensory and autonomic neuropathies and pain-related channelopathies. Schmerz 29(4):445–457

    Article  CAS  PubMed  Google Scholar 

  76. Rossor AM, Polke JM, Houlden H, Reilly MM (2013) Clinical implications of genetic advances in Charcot-Marie-Tooth disease. Nat Rev Neurol 9(10):562–571

    Article  CAS  PubMed  Google Scholar 

  77. Tazir M, Bellatache M, Nouioua S, Vallat JM (2013) Autosomal recessive Charcot-Marie-Tooth disease: from genes to phenotypes. J Peripher Nerv Syst 18(2):113–129

    Article  CAS  PubMed  Google Scholar 

  78. England JD, Gronseth GS, Franklin G, Carter GT, Kinsella LJ, Cohen JA et al (2009) Practice parameter: the evaluation of distal symmetric polyneuropathy: the role of laboratory and genetic testing (an evidence-based review). Report of the American Academy of Neurology, the American Association of Neuromuscular and Electrodiagnostic Medicine, and the American Academy of Physical Medicine and Rehabilitation. Pm R 1(1):5–13

    Article  CAS  PubMed  Google Scholar 

  79. Neundörfer B, Rautenstrauss B (2006) Hereditäre Polyneuropathien. In: Neundörfer B, Heuß D (Hrsg) Polyneuropathien. Thieme, Stuttgart, S 100–109

    Google Scholar 

  80. Pestronk A Neuromuscular disorders—hereditary neuropathies. http://neuromuscular.wustl.edu/time/hmsn.html

  81. Saporta AS, Sottile SL, Miller LJ, Feely SM, Siskind CE, Shy ME (2011) Charcot-Marie-Tooth disease subtypes and genetic testing strategies. Ann Neurol 69(1):22–33

    Article  PubMed  PubMed Central  Google Scholar 

  82. Hund E (2014) Hereditary transthyretin amyloidosis. Nervenarzt 85(10):1291–1297

    Article  CAS  PubMed  Google Scholar 

  83. Conceicao I, Gonzalez-Duarte A, Obici L, Schmidt HH, Simoneau D, Ong ML et al (2016) „Red flag“ symptom clusters in transthyretin familial amyloid polyneuropathy. J Peripher Nerv Syst 21(1):5–9

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Goedee HS, Brekelmans GJ, van Asseldonk JT, Beekman R, Mess WH, Visser LH (2013) High resolution sonography in the evaluation of the peripheral nervous system in polyneuropathy—a review of the literature. Eur J Neurol 20(10):1342–1351

    Article  CAS  PubMed  Google Scholar 

  85. Üçeyler N (2016) Ultraschall in der Diagnostik von Polyneuropathien. Aktuelle Neurol 43:548–556

    Article  Google Scholar 

  86. Goedee HS, van der Pol WL, van Asseldonk JH, Franssen H, Notermans NC, Vrancken AJ et al (2017) Diagnostic value of sonography in treatment-naive chronic inflammatory neuropathies. Neurology 88(2):143–151

    Article  PubMed  Google Scholar 

  87. Bernabeu A, Lopez-Celada S, Alfaro A, Mas JJ, Sanchez-Gonzalez J (2016) Is diffusion tensor imaging useful in the assessment of the sciatic nerve and its pathologies? Our clinical experience. Br J Radiol 89(1066):20150728

    Article  PubMed  PubMed Central  Google Scholar 

  88. Lichtenstein T, Sprenger A, Weiss K, Slebocki K, Cervantes B, Karampinos D et al (2018) MRI biomarkers of proximal nerve injury in CIDP. Ann Clin Transl Neurol 5(1):19–28

    Article  CAS  PubMed  Google Scholar 

  89. Sinclair CD, Miranda MA, Cowley P, Morrow JM, Davagnanam I, Mehta H et al (2011) MRI shows increased sciatic nerve cross sectional area in inherited and inflammatory neuropathies. J Neurol Neurosurg Psychiatry 82(11):1283–1286

    Article  CAS  PubMed  Google Scholar 

  90. Kronlage M, Pitarokoili K, Schwarz D, Godel T, Heiland S, Yoon MS et al (2017) Diffusion tensor imaging in chronic inflammatory demyelinating polyneuropathy: diagnostic accuracy and correlation with electrophysiology. Invest Radiol 52(11):701–707

    Article  PubMed  Google Scholar 

  91. Goedee HS, Jongbloed BA, van Asseldonk JH, Hendrikse J, Vrancken A, Franssen H et al (2017) A comparative study of brachial plexus sonography and magnetic resonance imaging in chronic inflammatory demyelinating neuropathy and multifocal motor neuropathy. Eur J Neurol 24(10):1307–1313

    Article  CAS  PubMed  Google Scholar 

  92. Antoine JC, Camdessanche JP (2017) Paraneoplastic neuropathies. Curr Opin Neurol 30(5):513–520

    Article  CAS  PubMed  Google Scholar 

  93. Camdessanche JP, Jousserand G, Franques J, Pouget J, Delmont E, Creange A et al (2012) A clinical pattern-based etiological diagnostic strategy for sensory neuronopathies: a French collaborative study. J Peripher Nerv Syst 17(3):331–340

    Article  PubMed  Google Scholar 

  94. Titulaer MJ, Soffietti R, Dalmau J, Gilhus NE, Giometto B, Graus F et al (2011) Screening for tumours in paraneoplastic syndromes: report of an EFNS task force. Eur J Neurol 18(1):19–19e3

    Article  CAS  PubMed  Google Scholar 

  95. Dispenzieri A (2017) POEMS syndrome: 2017 Update on diagnosis, risk stratification, and management. Am J Hematol 92(8):814–829

    Article  PubMed  Google Scholar 

  96. Adams D, Cauquil C, Labeyrie C (2017) Familial amyloid polyneuropathy. Curr Opin Neurol 30(5):481–489

    Article  CAS  PubMed  Google Scholar 

  97. Adams D, Lozeron P, Lacroix C (2012) Amyloid neuropathies. Curr Opin Neurol 25(5):564–572

    Article  CAS  PubMed  Google Scholar 

  98. Collins MP, Dyck PJ, Gronseth GS, Guillevin L, Hadden RD, Heuss D et al (2010) Peripheral Nerve Society Guideline on the classification, diagnosis, investigation, and immunosuppressive therapy of non-systemic vasculitic neuropathy: executive summary. J Peripher Nerv Syst 15(3):176–184

    Article  PubMed  Google Scholar 

  99. Collins MP, Hadden RD (2017) The nonsystemic vasculitic neuropathies. Nat Rev Neurol 13(5):302–316

    Article  PubMed  Google Scholar 

  100. Heuß D (2006) Morphologische Untersuchungen bei Polyneuropathien – Nerven- und Muskelbiopsie. In: Neundörfer B, Heuß D (Hrsg) Polyneuropathien. Referenzreihe Neurologie RRN. Thieme, Stuttgart, S 19–22 (Tafel I–VII.)

    Google Scholar 

  101. Sommer CL, Brandner S, Dyck PJ, Harati Y, LaCroix C, Lammens M et al (2010) Peripheral Nerve Society Guideline on processing and evaluation of nerve biopsies. J Peripher Nerv Syst 15(3):164–175

    Article  PubMed  Google Scholar 

  102. Collins MP, Mendell JR, Periquet MI, Sahenk Z, Amato AA, Gronseth GS et al (2000) Superficial peroneal nerve/peroneus brevis muscle biopsy in vasculitic neuropathy. Neurology 55(5):636–643

    Article  CAS  PubMed  Google Scholar 

  103. Leuschner T, Probst-Cousin S, Kayser C, Neundörfer B, Heuß D (2001) Reliabilität morphologischer Methoden in der Diagnostik einer vaskulitischen Neuropathie. Nervenheilkunde 40:20

    Google Scholar 

  104. Vital C, Vital A, Canron MH, Jaffre A, Viallard JF, Ragnaud JM et al (2006) Combined nerve and muscle biopsy in the diagnosis of vasculitic neuropathy. A 16-year retrospective study of 202 cases. J Peripher Nerv Syst 11(1):20–29

    Article  PubMed  Google Scholar 

  105. Vrancken AF, Gathier CS, Cats EA, Notermans NC, Collins MP (2011) The additional yield of combined nerve/muscle biopsy in vasculitic neuropathy. Eur J Neurol 18(1):49–58

    Article  CAS  PubMed  Google Scholar 

  106. Üçeyler N, Devigili G, Toyka KV, Sommer C (2010) Skin biopsy as an additional diagnostic tool in non-systemic vasculitic neuropathy. Acta Neuropathol 120(1):109–116

    Article  PubMed  Google Scholar 

  107. Verschueren A (2007) Immune-mediated neuropathies: indications and value of nerve biopsy. Rev Neurol 163(Spec No 1):3S58–3S60

    PubMed  Google Scholar 

  108. Allen JA, Gorson KC, Gelinas D (2018) Challenges in the diagnosis of chronic inflammatory demyelinating polyneuropathy. Brain Behav 8(3):e932

    Article  PubMed  PubMed Central  Google Scholar 

  109. Sommer C (2018) Fortschr Neurol Psychiatr 86:509–518

    Article  PubMed  Google Scholar 

  110. Terkelsen AJ, Karlsson P, Lauria G, Freeman R, Finnerup NB, Jensen TS (2017) The diagnostic challenge of small fibre neuropathy: clinical presentations, evaluations, and causes. Lancet Neurol 16(11):934–944

    Article  PubMed  Google Scholar 

  111. Levine TD (2018) Small fiber neuropathy: disease classification beyond pain and burning. J Cent Nerv Syst Dis 10:1179573518771703

    Article  PubMed  PubMed Central  Google Scholar 

  112. Blackmore D, Siddiqi ZA (2017) Diagnostic criteria for small fiber neuropathy. J Clin Neuromuscul Dis 18(3):125–131

    Article  PubMed  Google Scholar 

  113. Joint Task Force of the E, the PNS (2010) European Federation of Neurological Societies/Peripheral Nerve Society Guideline on management of chronic inflammatory demyelinating polyradiculoneuropathy: report of a joint task force of the European Federation of Neurological Societies and the Peripheral Nerve Society—First Revision. J Peripher Nerv Syst 15(1):1–9

    Article  Google Scholar 

  114. Hottinger AF (2016) Neurologic complications of immune checkpoint inhibitors. Curr Opin Neurol 29(6):806–812

    Article  CAS  PubMed  Google Scholar 

  115. Echaniz-Laguna A, de Seze J, Chanson JB (2012) Chronic inflammatory demyelinating polyradiculoneuropathy in solid organ transplant recipients: a prospective study. J Neurol Neurosurg Psychiatry 83(7):699–705

    Article  PubMed  Google Scholar 

  116. Karam C, Mauermann ML, Johnston PB, Lahoria R, Engelstad JK, Dyck PJ (2014) Immune-mediated neuropathies following stem cell transplantation. J Neurol Neurosurg Psychiatry 85(6):638–642

    Article  PubMed  Google Scholar 

  117. Romero S, Montoro J, Guinot M, Almenar L, Andreu R, Balaguer A et al (2018) Post-transplant lymphoproliferative disorders after solid organ and hematopoietic stem cell transplantation. Leuk Lymphoma 60:1–9

    Google Scholar 

  118. Julian T, Glascow N, Syeed R, Zis P (2018) Alcohol-related peripheral neuropathy: a systematic review and meta-analysis. J Neurol. https://doi.org/10.3201/eid2202.141762

    Article  PubMed  PubMed Central  Google Scholar 

  119. Mellion M, Gilchrist JM, de la Monte S (2011) Alcohol-related peripheral neuropathy: nutritional, toxic, or both? Muscle Nerve 43(3):309–316

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Mellion ML, Silbermann E, Gilchrist JM, Machan JT, Leggio L, de la Monte S (2014) Small-fiber degeneration in alcohol-related peripheral neuropathy. Alcohol Clin Exp Res 38(7):1965–1972

    Article  PubMed  PubMed Central  Google Scholar 

  121. Callaghan BC, Hur J, Feldman EL (2012) Diabetic neuropathy: one disease or two? Curr Opin Neurol 25(5):536–541

    Article  PubMed  PubMed Central  Google Scholar 

  122. Polydefkis M, Griffin JW, McArthur J (2003) New insights into diabetic polyneuropathy. JAMA 290(10):1371–1376

    Article  CAS  PubMed  Google Scholar 

  123. Visser NA, Notermans NC, Linssen RS, van den Berg LH, Vrancken AF (2015) Incidence of polyneuropathy in Utrecht, the Netherlands. Neurology 84(3):259–264

    Article  CAS  PubMed  Google Scholar 

  124. Zis P, Sarrigiannis PG, Rao DG, Hewamadduma C, Hadjivassiliou M (2016) Chronic idiopathic axonal polyneuropathy: a systematic review. J Neurol 263(10):1903–1910

    Article  PubMed  Google Scholar 

  125. Finsterer J, Wanschitz J, Quasthoff S, Iglseder S, Loscher W, Grisold W (2017) Causally treatable, hereditary neuropathies in Fabry’s disease, transthyretin-related familial amyloidosis, and Pompe’s disease. Acta Neurol Scand 136(6):558–569

    Article  CAS  PubMed  Google Scholar 

  126. Heuss D (2019) Therapie der Polyneuropathien. In: Therapie-Handbuch. Elsevier – Urban & Fischer ( http://www1.us.elsevierhealth.com/THB/chapter_P011.php )

  127. Sommer C, Gold R, Heuss D et al.Therapie akuter und chronischer immunvermittelter Neuropathien und Neuritiden, S2e-Leitlinie. In: Neurologie DGf, editor. Leitlinien für Diagnostik und Therapie in der Neurologie: AWMF online. Das Portal der wissenschaftlichen Medizin; 2018.

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to D. Heuß.

Ethics declarations

Interessenkonflikt

Siehe Interessenkonflikterklärung auf www.dgn.org/leitlinien.

Für diesen Beitrag wurden von den Autoren keine Studien an Menschen oder Tieren durchgeführt. Für die aufgeführten Studien gelten die jeweils dort angegebenen ethischen Richtlinien.

Additional information

Redaktion

P. Berlit, Berlin

Fachgesellschaft

Deutsche Gesellschaft für Neurologie (DGN)

Federführend

Prof. Dr. D. Heuß, Neurologische Klinik, Universitätsklinikum Erlangen, Schwabachanlage 6, 91054 Erlangen, E‑Mail: dieter.heuss@uk-erlangen.de

Redaktionskomitee

D. Heuß, Neurologische Klinik, Universitätsklinikum Erlangen

E. Hund, Neurologische Klinik, Amyloidosezentrum, Universitätsklinikum Heidelberg

J. Klehmet, Klinik für Neurologie, Charité Universitätsmedizin Berlin

I. Kurth, Institut für Humangenetik, Universitätsklinik RWTH (Rheinisch-Westfälische Technische Hochschule) Aachen

H. Lehmann, Klinik und Poliklinik für Neurologie, Universitätsklinikum Köln

C. Sommer, Neurologische Klinik, Universitätsklinikum Würzburg

Redaktionskomitee Österreich

W. Löscher, Neurologische Klinik, Medizinische Universität Innsbruck, Österreich

Redaktionskomitee Schweiz

S. Renaud, Département médecine, Hôpital neuchâtelois, Neuchâtel, Schweiz

Die vollständige Leitlinie wurde unter https://www.dgn.org/leitlinien/3754-ll-030-067-diagnostik-bei-polyneuropathien-2019 am 10.04.2019 publiziert.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Heuß, D. S1-Leitlinie: Diagnostik bei Polyneuropathien. DGNeurologie 2, 359–382 (2019). https://doi.org/10.1007/s42451-019-0099-6

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s42451-019-0099-6

Navigation