827
Views
0
CrossRef citations to date
0
Altmetric
REVIEW

Myasthenia Gravis and Lambert-Eaton Myasthenic Syndrome: New Developments in Diagnosis and Treatment

ORCID Icon &
Pages 3001-3022 | Received 19 Sep 2022, Accepted 24 Nov 2022, Published online: 22 Dec 2022

References

  • Grob D, Brunner NG, Namba T. The natural course of myasthenia gravis and effect of therapeutic measures. NYAS. 1981;377:652–669.
  • Oosterhuis HJ. The natural course of myasthenia gravis: a long term follow up study. J Neurol Neurosurg Psychiatry. 1989;52(10):1121–1127. PMID: 2795037; PMCID: PMC1031695. doi:10.1136/jnnp.52.10.1121
  • Lazaridis K, Tzartos SJ. Autoantibody specificities in myasthenia gravis; implications for improved diagnostics and therapeutics. Front Immunol. 2020;11:212. doi:10.3389/fimmu.2020.00212
  • Hughes BW, Kusner LL, Kaminski HJ. Molecular architecture of the neuromuscular junction. Muscle Nerve. 2006;33(4):445–461. PMID: 16228970. doi:10.1002/mus.20440
  • Bacchi S, Kramer P, Chalk C. Autoantibodies to low-density lipoprotein receptor-related protein 4 in double seronegative myasthenia gravis: a systematic review. Can J Neurol Sci. 2018;45:62–67. doi:10.1017/cjn.2017.253
  • Zhang B, Shen C, Bealmear B, et al. Autoantibodies to agrin in myasthenia gravis patients. PLoS One. 2014;9:e91816. doi:10.1371/journal.pone.0091816
  • Gasperi C, Melms A, Schoser B, et al. Anti-agrin autoantibodies in myasthenia gravis. Neurology. 2014;82(22):1976–1983. doi:10.1212/WNL.0000000000000478
  • Yoganathan K, Stevenson A, Tahir A, Sadler R, Radunovic A, Malek N. Bedside and laboratory diagnostic testing in myasthenia. J Neurol. 2022;269(6):3372–3384. PMID: 35142871; PMCID: PMC9119875. doi:10.1007/s00415-022-10986-3
  • Skolka M, Lamb CJ, Rubin DI, Klein CJ, Laughlin RS. Electrodiagnostic characteristics suggestive of muscle-specific kinase myasthenia gravis. Neurol Clin Pract. 2022;12(3):211–217. doi:10.1212/CPJ.0000000000001166
  • Pascuzzi RM. The edrophonium test. Semin Neurol. 2003;23(1):83–88. PMID: 12870109. doi:10.1055/s-2003-40755
  • Giannoccaro MP, Paolucci M, Zenesini C, et al. Comparison of ice pack test and single-fiber EMG diagnostic accuracy in patients referred for myasthenic ptosis. Neurology. 2020;95(13):e1800–e1806. PMID: 32788239. doi:10.1212/WNL.0000000000010619
  • Pascuzzi RM, Coslett HB, Johns TR. Long-term corticosteroid treatment of myasthenia gravis: report of 116 patients. Ann Neurol. 1984;15(3):291–298. PMID: 6721451. doi:10.1002/ana.410150316
  • Menon D, Bril V. Pharmacotherapy of generalized myasthenia gravis with special emphasis on newer biologicals. Drugs. 2022;82:865–887. doi:10.1007/s40265-022-01726-y
  • Tindall RS, Phillips JT, Rollins JA, Wells L, Hall K. A clinical therapeutic trial of cyclosporine in myasthenia gravis. Ann N Y Acad Sci. 1993;681:539–551. PMID: 8357194. doi:10.1111/j.1749-6632.1993.tb22937.x
  • Sanders DB, Hart IK, Mantegazza R, et al. An international, Phase III, randomized trial of mycophenolate mofetil in myasthenia gravis. Neurology. 2008;71(6):400–406. doi:10.1212/01.wnl.0000312374.95186.cc
  • Cahoon WD Jr, Kockler DR. Mycophenolate mofetil treatment of myasthenia gravis. Ann Pharmacother. 2006;40(2):295–298. doi:10.1345/aph.1G501
  • Wang L, Zhang S, Xi J, et al. Efficacy and safety of tacrolimus for myasthenia gravis: a systematic review and meta-analysis. J Neurol. 2017;264(11):2191–2200. doi:10.1007/s00415-017-8616-7
  • Yoshikawa H, Kiuchi T, Saida T, Takamori M. Randomised, double-blind, placebo-controlled study of tacrolimus in myasthenia gravis. J Neurol Neurosurg Psychiatry. 2011;82(9):970–977. doi:10.1136/jnnp-2011-300148
  • Pasnoor M, He J, Herbelin L, et al. A randomized controlled trial of methotrexate for patients with generalized myasthenia gravis. Neurology. 2016;87(1):57–64. doi:10.1212/WNL.0000000000002795
  • Howard JF Jr, Utsugisawa K, Benatar M, et al. Safety and efficacy of eculizumab in anti-acetylcholine receptor antibody-positive refractory generalised myasthenia gravis (REGAIN): a phase 3, randomised, double-blind, placebo-controlled, multicentre study. Lancet Neurol. 2017;16:976–986. doi:10.1016/S1474-4422(17)30369-1
  • Muppidi S, Utsugisawa K, Benatar M, et al. Long-term safety and efficacy of eculizumab in generalized myasthenia gravis. Muscle Nerve. 2019;60:14–24. doi:10.1002/mus.26447
  • Vissing J, Jacob S, Fujita KP, O’Brien F, Howard JF; REGAIN study group. ‘Minimal symptom expression’ in patients with acetylcholine receptor antibody-positive refractory generalized myasthenia gravis treated with eculizumab. J Neurol. 2020;267(7):1991–2001. PMID: 32189108; PMCID: PMC7320935. doi:10.1007/s00415-020-09770-y
  • Howard JF, Nowak RJ, Wolfe GI, et al. Clinical effects of the self-administered subcutaneous complement inhibitor zilucoplan in patients with moderate to severe generalized myasthenia gravis: results of a phase 2 randomized, double-blind, placebo-controlled, multicenter clinical trial. JAMA Neurol. 2020;77(5):582–592. doi:10.1001/jamaneurol.2019.5125
  • Howard JF, Genge A, Hussain Y, et al. Outcomes from raise: a randomized, placebo-controlled, double-blind, phase 3 trial of zilucoplan in generalized myasthenia gravis. mus mgfa Abstracts; 2022.
  • Vu T, Meisel A, Mantegazza R, et al. Efficacy and safety of ravulizumab, a long-acting terminal complement inhibitor, in adults with anti-acetylcholine receptor antibody-positive generalized myasthenia gravis: results from the phase 3 CHAMPION MG study. Neurology. 2022;98(18 Supplement):791.
  • Hehir MK, Hobson-Webb LD, Benatar M, et al. Rituximab as treatment for anti-MuSK myasthenia gravis: multicenter blinded prospective review. Neurology. 2017;89:1069–1077. doi:10.1212/WNL.0000000000004341
  • Tandan R, Hehir MK 2nd, Waheed W, Howard DB. Rituximab treatment of myasthenia gravis: a systematic review. Muscle Nerve. 2017;56:185–196. doi:10.1002/mus.25597
  • Topakian R, Zimprich F, Iglseder S, et al. High efficacy of rituximab for myasthenia gravis: a comprehensive nationwide study in Austria. J Neurol. 2019;266:699–706. doi:10.1007/s00415-019-09191-6
  • Di Stefano V, Lupica A, Rispoli MG, Di Muzio A, Brighina F, Rodolico C. Rituximab in AChR subtype of myasthenia gravis: systematic review. J Neurol Neurosurg Psychiatry. 2020;91:392–395. doi:10.1136/jnnp-2019-322606
  • Young C, McGill SC. Rituximab for the Treatment of Myasthenia Gravis: A 2021 Update [Internet]. Ottawa (ON): Canadian Agency for Drugs and Technologies in Health; 2021. Available from https://www.ncbi.nlm.nih.gov/books/NBK571915/. Accessed November 25, 2022.
  • Howard JF Jr, Bril V, Vu T, et al; ADAPT Investigator Study Group. Safety, efficacy, and tolerability of efgartigimod in patients with generalised myasthenia gravis (ADAPT): a multicentre, randomised, placebo-controlled, phase 3 trial. Lancet Neurol. 2021;20(7):526–536. Erratum in: Lancet Neurol. 2021 Aug;20(8):e5.PMID: 34146511. doi:10.1016/S1474-4422(21)00159-9
  • Bril V, Benatar M, Andersen H, et al; MG0002 Investigators. Efficacy and safety of rozanolixizumab in moderate to severe generalized myasthenia gravis: a phase 2 randomized control trial. Neurology. 2021;96(6):e853–e865. PMID: 33219142; PMCID: PMC8105899. doi:10.1212/WNL.0000000000011108
  • Bril V, Druzdz A, Grosskreutz J, et al. Efficacy and safety of rozanolixizumab in patients with generalized myasthenia gravis: a randomized, multicenter, double-blind, placebo-controlled phase 3 study MGFA abstracts. Neurology. 2022;65:S35. doi:10.1002/mus.27540
  • Usmani A, Kwan L, Wahib-Khalil D, Trivedi J, Nations S, Sarode R. Excellent response to therapeutic plasma exchange in myasthenia gravis patients irrespective of antibody status. J Clin Apher. 2019;34:416–422. doi:10.1002/jca.21694
  • Yamada C, Teener JW, Davenport RD, Cooling L. Maintenance plasma exchange treatment for muscle specific kinase antibody positive myasthenia gravis patients. J Clin Apher. 2015;30:314–319. doi:10.1002/jca.21377
  • Ortiz-Salas P, Velez-Van-Meerbeke A, Galvis-Gomez CA, Rodriguez QJ. Human immunoglobulin versus plasmapheresis in guillain-barre syndrome and myasthenia gravis: a meta-analysis. J Clin Neuromuscul Dis. 2016;18:1–11. doi:10.1097/CND.0000000000000119
  • Sanders DB, Wolfe GI, Benatar M, et al. International consensus guidance for management of myasthenia gravis: executive summary. Neurology. 2016;87:419–425. doi:10.1212/WNL.0000000000002790
  • Adiao KJB, Espiritu AI, Roque VLA, Reyes J. Efficacy and tolerability of subcutaneously administered immunoglobulin in myasthenia gravis: a systematic review. J Clin Neurosci. 2020;72:316–321.
  • Hellmann MA, Mosberg-Galili R, Lotan I, Steiner I. Maintenance IVIg therapy in myasthenia gravis does not affect disease activity. J Neurol Sci. 2014;338:39–42. doi:10.1016/j.jns.2013.10.043
  • Alipour-Faz A, Shojaei M, Peyvandi H, et al. A comparison between IVIG and plasma exchange as preparations before thymectomy in myasthenia gravis patients. Acta Neurol Belg. 2017;117:245–249. doi:10.1007/s13760-016-0689-z
  • Rahbek MA, Mikkelsen EE, Overgaard K, Vinge L, Andersen H, Dalgas U. Exercise in myasthenia gravis: a feasibility study of aerobic and resistance training. Muscle Nerve. 2017;56:700–709. doi:10.1002/mus.25552
  • McQuillen MP, Leone MG. A treatment carol: thymectomy revisited. Neurology. 1977;27:1103–1106. doi:10.1212/WNL.27.12.1103
  • Wolfe GI, Kaminski HJ, Aban IB, et al. Randomized trial of thymectomy in myasthenia gravis. N Engl J Med. 2016;375:511–522. doi:10.1056/NEJMoa1602489
  • Wolfe GI, Kaminski HJ, Aban IB, et al. Long-term effect of thymectomy plus prednisone versus prednisone alone in patients with non-thymomatous myasthenia gravis: 2-year extension of the MGTX randomised trial. Lancet Neurol. 2019;18:259–268. doi:10.1016/S1474-4422(18)30392-2
  • Gronseth GS, Barohn R, Narayanaswami P. Practice advisory: thymectomy for myasthenia gravis (practice parameter update): report of the guideline development, dissemination, and implementation subcommittee of the American Academy of Neurology. Neurology. 2020;94:705–709. doi:10.1212/WNL.0000000000009294
  • Zhu K, Li J, Huang X, et al. Thymectomy is a beneficial therapy for patients with non-thymomatous ocular myasthenia gravis: a systematic review and meta-analysis. Neurol Sci. 2017;38:1753–1760. doi:10.1007/s10072-017-3058-7
  • Mehrizi M, Fontem RF, Gearhart TR, Pascuzzi RM. Medications and myasthenia gravis (a reference for health care professionals) prepared for the myasthenia gravis foundation of America; 2012. Available from: www.myasthenia.org. Accessed November 25, 2022.
  • Mehrizi M, Pascuzzi RM. Complications of radiologic contrast in patients with myasthenia gravis. Muscle Nerve. 2014;50:443–444. doi:10.1002/mus.24254
  • Abdel-Wahab N, Shah M, Lopez-Olivo MA, Suarez-Almazor ME. Use of immune checkpoint inhibitors in the treatment of patients with cancer and preexisting autoimmune disease: a systematic review. Ann Intern Med. 2018;168:121–130. doi:10.7326/M17-2073
  • Safa H, Johnson DH, Trinh VA, et al. Immune checkpoint inhibitor related myasthenia gravis: single center experience and systematic review of the literature. J Immunother Cancer. 2019;7:319. doi:10.1186/s40425-019-0774-y
  • Kochhar PK, Schumacher RE, Sarkar S. Transient neonatal myasthenia gravis: refining risk estimate for infants born to women with myasthenia gravis. J Perinatol. 2021;41:2279–2283. doi:10.1038/s41372-021-00970
  • Vanhaesebrouck AE, Beeson D. The congenital myasthenic syndromes: expanding genetic and phenotypic spectrums and refining treatment strategies. Curr Opin Neurol. 2019;32:696–703. doi:10.1097/WCO.0000000000000736
  • Burns TM, Russell JA, LaChance DH, Jones HR. Oculobulbar involvement is typical with Lambert-Eaton myasthenic syndrome. Ann Neurol. 2003;53:270–273. doi:10.1002/ana.10477
  • Young JD, Leavitt JA. Lambert-Eaton myasthenic syndrome: ocular signs and symptoms. J Neuroophthalmol. 2016;36:20–22. doi:10.1097/WNO.0000000000000258
  • Kennelly KD, Dodick DW, Pascuzzi RM, et al. Neuronal autoantibodies and paraneoplastic neurological syndromes associated with extrapulmonary small cell carsinoma. Neurology. 1997;48:A31–A32.
  • Hong BY, An HJ, Lim SH. Long-Standing Lambert–Eaton myasthenic syndrome caused by undetectable small-cell lung cancer: why we should follow-up LEMS. Diagnostics. 2022;12:1542. doi:10.3390/diagnostics12071542
  • Sabater L, Titulaer M, Saiz A, Verschuuren J, Gure AO, Graus F. SOX1 antibodies are markers of paraneoplastic Lambert-Eaton myasthenic syndrome. Neurology. 2008;70:924–928. doi:10.1212/01.wnl.0000281663.81079.24
  • Kesner VG, Oh SJ, Dimachkie MM, Barohn RJ. Lambert-Eaton myasthenic syndrome. Neurol Clin. 2018;36:379–394. doi:10.1016/j.ncl.2018.01.008
  • Maddison P, Gozzard P, Grainge MJ, Lang B. Long-term survival in paraneoplastic Lambert-Eaton myasthenic syndrome. Neurology. 2017;88:1334–1339. doi:10.1212/WNL.0000000000003794
  • McEvoy KM, Windebank AJ, Daube JR, Low PA. 3,4-Diaminopyridine in the treatment of Lambert-Eaton myasthenic syndrome. N Engl J Med. 1989;321:1567–1571. doi:10.1056/NEJM198912073212303
  • Oh SJ, Shcherbakova N, Kostera-Pruszczyk A, et al. Amifampridine phosphate (Firdapse((R))) is effective and safe in a phase 3 clinical trial in LEMS. Muscle Nerve. 2016;53:717–725. doi:10.1002/mus.25070
  • Sanders DB, Juel VC, Harati Y, et al. 3,4-diaminopyridine base effectively treats the weakness of Lambert-Eaton myasthenia. Muscle Nerve. 2018;57:561–568. doi:10.1002/mus.26052
  • Anwar A, Saleem S, Ahmed MF, Ashraf S, Ashraf S. Recent advances and therapeutic options in Lambert-Eaton myasthenic syndrome. Cureus. 2019;11(8):e5450. PMID: 31637147; PMCID: PMC6799875. doi:10.7759/cureus.5450
  • Meisel A, Sieb JP, Le Masson G, et al. The European Lambert–Eaton myasthenic syndrome registry: long-term outcomes following symptomatic treatment. Neurol Ther. 2022;11(3):1071–1083. doi:10.1007/s40120-022-00354-8
  • Johns TR, editor. Myasthenia gravis. In: Seminars in Neurology. Vol. 2. StatPearls [Internet]; 1982:3. doi:10.1055/s-002-11126
  • Nowak RJ, Coffey CS, Goldstein JM, et al. (29a) rituximab in acetylcholine receptor antibody-positive generalized myasthenia gravis: the BeatMG study. Neurology. 2022;98(4):e376–e389. doi:10.1212/WNL.0000000000013121
  • Jaretzki AI, Barohn RJ, Ernstoff RM, et al. Myasthenia gravis recommendations for clinical research standards task force of the medical scientific advisory board of the myasthenia gravis foundation of America. Neurology. 2000;55(1):16–23. doi:10.1212/WNL.55.1.16
  • Barohn RJ, McIntire D, Herbelin L, Wolfe GI, Nations S, Bryan WW. Reliability testing of the quantitative myasthenia gravis score. Ann N Y Acad Sci. 1998;841:769–772. doi:10.1111/j.1749-6632.1998.tb11015.x
  • Wolfe GI, Herbelin L, Nations SP, Foster B, Bryan WW, Barohn RJ. Myasthenia gravis activities of daily living profile. Neurology. 1999;52:1487–1489. doi:10.1212/WNL.52.7.1487
  • Burns TM, Conaway MR, Cutter GR, Sanders DB. Less is more, or almost as much: a 15-item quality-of-life instrument for myasthenia gravis. Muscle Nerve. 2008;38(2):957–963. doi:10.1002/mus.21053
  • Burns TM, Sadjadi R, Utsugisawa K, et al. International clinimetric evaluation of the MG-QOL15, resulting in slight revision and subsequent validation of the MG-QOL15r. Muscle Nerve. 2016;54(6):1015–1022. doi:10.1002/mus.25198
  • Burns TM, Conaway MR, Cutter GR, Sanders DB. Construction of an efficient evaluative instrument for myasthenia gravis: the MG composite. Muscle Nerve. 2008;38(6):1553–1562. doi:10.1002/mus.21185
  • Burns TM, Conaway M, Sanders DB; MG Composite and MG-QOL15 Study Group. The MG composite: a valid and reliable outcome measure for myasthenia gravis. Neurology. 2010;74(18):1434–1440. doi:10.1212/WNL.0b013e3181dc1b1e
  • Thomsen JLS, Andersen H. Outcome measures in clinical trials of patients with myasthenia gravis. Front Neurol. 2020;11:596382. PMID: 33424747; PMCID: PMC7793650. doi:10.3389/fneur.2020.596382