Skip to main content
Top
Published in: Current Neurology and Neuroscience Reports 10/2018

01-10-2018 | Nerve and Muscle (L H Weimer, Section Editor)

Neuromuscular Complications of Programmed Cell Death-1 (PD-1) Inhibitors

Authors: Justin C. Kao, Adipong Brickshawana, Teerin Liewluck

Published in: Current Neurology and Neuroscience Reports | Issue 10/2018

Login to get access

Abstract

Purpose of Review

In recent years, immune checkpoint inhibitors have been increasingly used in patients with metastatic cancers with favorable oncological outcomes; however, there have also been increasing number of cancer survivors who have developed immune-related adverse events. Little is known about PD-1 inhibitor-associated neuromuscular complications.

Recent Findings

Neuromuscular disorders are the most common neurological complication reported in PD-1 inhibitor-treated patients. Myasthenia gravis, immune-mediated myopathies, and Guillain-Barre syndrome are among commonly reported immune-related neuromuscular complications. HyperCKemia occurs frequently in patients with PD-1 inhibitor-associated myasthenia gravis, indicating coexisting myopathies or myocarditis. Oculobulbar weakness is a unique and common presentation of PD-1 inhibitor-associated immune-mediated myopathies with or without concomitant myasthenia gravis. High-dose steroid monotherapy may be associated with clinical deterioration in some patients with PD-1 inhibitor-associated myasthenia gravis, immune-mediated myopathies, or Guillain-Barre syndrome.

Summary

PD-1 inhibitor-associated neuromuscular complications have some characteristic features compared to their idiopathic counterparts. Although steroid monotherapy is commonly used in non-neuromuscular autoimmune disorders triggered by anti-PD-1 therapy, this may lead to unfavorable outcomes in some patients with PD-1 inhibitor-associated neuromuscular complications.
Literature
4.
go back to reference Sharpe AH, Pauken KE. The diverse functions of the PD1 inhibitory pathway. Nat Rev Immunol. 2018;18:153–67.CrossRefPubMed Sharpe AH, Pauken KE. The diverse functions of the PD1 inhibitory pathway. Nat Rev Immunol. 2018;18:153–67.CrossRefPubMed
5.
go back to reference Jiang TT, Martinov T, Xin L, Kinder JM, Spanier JA, Fife BT, et al. Programmed death-1 culls peripheral accumulation of high-affinity autoreactive CD4 T cells to protect against autoimmunity. Cell Rep. 2016;17:1783–94.CrossRefPubMedPubMedCentral Jiang TT, Martinov T, Xin L, Kinder JM, Spanier JA, Fife BT, et al. Programmed death-1 culls peripheral accumulation of high-affinity autoreactive CD4 T cells to protect against autoimmunity. Cell Rep. 2016;17:1783–94.CrossRefPubMedPubMedCentral
6.
go back to reference Garcia-Diaz A, Shin DS, Moreno BH, Saco J, Escuin-Ordinas H, Rodriguez GA, et al. Interferon receptor signaling pathways regulating PD-L1 and PD-L2 expression. Cell Rep. 2017;19:1189–201.CrossRefPubMedPubMedCentral Garcia-Diaz A, Shin DS, Moreno BH, Saco J, Escuin-Ordinas H, Rodriguez GA, et al. Interferon receptor signaling pathways regulating PD-L1 and PD-L2 expression. Cell Rep. 2017;19:1189–201.CrossRefPubMedPubMedCentral
8.
go back to reference • Ribas A, Wolchok JD. Cancer immunotherapy using checkpoint blockade. Science. 2018;359:1350–5. A comprehensive review of cancer immunotherapy using immune checkpoint imhibitors. CrossRefPubMedPubMedCentral • Ribas A, Wolchok JD. Cancer immunotherapy using checkpoint blockade. Science. 2018;359:1350–5. A comprehensive review of cancer immunotherapy using immune checkpoint imhibitors. CrossRefPubMedPubMedCentral
9.
go back to reference Zamani MR, Aslani S, Salmaninejad A, Javan MR, Rezaei N. PD-1/PD-L and autoimmunity: a growing relationship. Cell Immunol. 2016;310:27–41.CrossRefPubMed Zamani MR, Aslani S, Salmaninejad A, Javan MR, Rezaei N. PD-1/PD-L and autoimmunity: a growing relationship. Cell Immunol. 2016;310:27–41.CrossRefPubMed
10.
go back to reference Weber JS, D’Angelo SP, Minor D, Hodi FS, Gutzmer R, Neyns B, et al. Nivolumab versus chemotherapy in patients with advanced melanoma who progressed after anti-CTLA-4 treatment (CheckMate 037): a randomised, controlled, open-label, phase 3 trial. Lancet Oncol. 2015;16:375–84.CrossRefPubMed Weber JS, D’Angelo SP, Minor D, Hodi FS, Gutzmer R, Neyns B, et al. Nivolumab versus chemotherapy in patients with advanced melanoma who progressed after anti-CTLA-4 treatment (CheckMate 037): a randomised, controlled, open-label, phase 3 trial. Lancet Oncol. 2015;16:375–84.CrossRefPubMed
11.
go back to reference Robert C, Schachter J, Long GV, Arance A, Grob JJ, Mortier L, et al. Pembrolizumab versus Ipilimumab in advanced melanoma. N Engl J Med. 2015;372:2521–32.CrossRefPubMed Robert C, Schachter J, Long GV, Arance A, Grob JJ, Mortier L, et al. Pembrolizumab versus Ipilimumab in advanced melanoma. N Engl J Med. 2015;372:2521–32.CrossRefPubMed
12.
go back to reference •• Kao JC, Liao B, Markovic SN, Klein CJ, Naddaf E, Staff NP, et al. Neurological complications associated with anti-programmed death 1 (PD-1) antibodies. JAMA Neurol. 2017;74:1216–22. A series of patients with PD-1 inhibitor-associated neurologic complications highlighting the breadth, diversity, and frequency of neuromuscular complications. CrossRefPubMedPubMedCentral •• Kao JC, Liao B, Markovic SN, Klein CJ, Naddaf E, Staff NP, et al. Neurological complications associated with anti-programmed death 1 (PD-1) antibodies. JAMA Neurol. 2017;74:1216–22. A series of patients with PD-1 inhibitor-associated neurologic complications highlighting the breadth, diversity, and frequency of neuromuscular complications. CrossRefPubMedPubMedCentral
13.
go back to reference •• Zimmer L, Goldinger SM, Hofmann L, Loquai C, Ugurel S, Thomas I, et al. Neurological, respiratory, musculoskeletal, cardiac and ocular side-effects of anti-PD-1 therapy. Eur J Cancer. 2016;60:210–25. A series of patients with PD-1 inhibitor-associated neurologic complications highlighting the breadth, diversity, and frequency of neuromuscular complications. CrossRefPubMed •• Zimmer L, Goldinger SM, Hofmann L, Loquai C, Ugurel S, Thomas I, et al. Neurological, respiratory, musculoskeletal, cardiac and ocular side-effects of anti-PD-1 therapy. Eur J Cancer. 2016;60:210–25. A series of patients with PD-1 inhibitor-associated neurologic complications highlighting the breadth, diversity, and frequency of neuromuscular complications. CrossRefPubMed
14.
go back to reference Spain L, Diem S, Larkin J. Management of toxicities of immune checkpoint inhibitors. Cancer Treat Rev. 2016;44:51–60.CrossRefPubMed Spain L, Diem S, Larkin J. Management of toxicities of immune checkpoint inhibitors. Cancer Treat Rev. 2016;44:51–60.CrossRefPubMed
15.
go back to reference •• Liewluck T, Kao JC, Mauermann ML. PD-1 inhibitor-associated myopathies: emerging immune-mediated myopathies. J Immunother. 2018;41:208–11. A series of patients with PD-1 inhibitor-associated immune-mediated myopathies highlighting a unique oculobulbar involvement in these patients, mimicking myasthenia gravis. CrossRefPubMed •• Liewluck T, Kao JC, Mauermann ML. PD-1 inhibitor-associated myopathies: emerging immune-mediated myopathies. J Immunother. 2018;41:208–11. A series of patients with PD-1 inhibitor-associated immune-mediated myopathies highlighting a unique oculobulbar involvement in these patients, mimicking myasthenia gravis. CrossRefPubMed
16.
go back to reference Lau KH, Kumar A, Yang IH, Nowak RJ. Exacerbation of myasthenia gravis in a patient with melanoma treated with pembrolizumab. Muscle Nerve. 2016;54:157–61.CrossRefPubMed Lau KH, Kumar A, Yang IH, Nowak RJ. Exacerbation of myasthenia gravis in a patient with melanoma treated with pembrolizumab. Muscle Nerve. 2016;54:157–61.CrossRefPubMed
17.
go back to reference Nguyen BH, Kuo J, Budiman A, Christie H, Ali S. Two cases of clinical myasthenia gravis associated with pembrolizumab use in responding melanoma patients. Melanoma Res. 2017;27:152–4.CrossRefPubMed Nguyen BH, Kuo J, Budiman A, Christie H, Ali S. Two cases of clinical myasthenia gravis associated with pembrolizumab use in responding melanoma patients. Melanoma Res. 2017;27:152–4.CrossRefPubMed
18.
go back to reference Phadke SD, Ghabour R, Swick BL, Swenson A, Milhem M, Zakharia Y. Pembrolizumab therapy triggering an exacerbation of preexisting autoimmune disease: a report of 2 patient cases. J Investig Med High Impact Case Rep. 2016;4:2324709616674316.PubMedPubMedCentral Phadke SD, Ghabour R, Swick BL, Swenson A, Milhem M, Zakharia Y. Pembrolizumab therapy triggering an exacerbation of preexisting autoimmune disease: a report of 2 patient cases. J Investig Med High Impact Case Rep. 2016;4:2324709616674316.PubMedPubMedCentral
19.
go back to reference Alnahhas I, Wong J. A case of new-onset antibody-positive myasthenia gravis in a patient treated with pembrolizumab for melanoma. Muscle Nerve. 2017;55:E25-E26.CrossRef Alnahhas I, Wong J. A case of new-onset antibody-positive myasthenia gravis in a patient treated with pembrolizumab for melanoma. Muscle Nerve. 2017;55:E25-E26.CrossRef
20.
go back to reference Maeda O, Yokota K, Atsuta N, Katsuno M, Akiyama M, Ando Y. Nivolumab for the treatment of malignant melanoma in a patient with pre-existing myasthenia gravis. Nagoya J Med Sci. 2016;78:119–22.PubMedPubMedCentral Maeda O, Yokota K, Atsuta N, Katsuno M, Akiyama M, Ando Y. Nivolumab for the treatment of malignant melanoma in a patient with pre-existing myasthenia gravis. Nagoya J Med Sci. 2016;78:119–22.PubMedPubMedCentral
21.
go back to reference Zhu J, Li Y. Myasthenia gravis exacerbation associated with pembrolizumab. Muscle Nerve. 2016;54:506–7.CrossRefPubMed Zhu J, Li Y. Myasthenia gravis exacerbation associated with pembrolizumab. Muscle Nerve. 2016;54:506–7.CrossRefPubMed
22.
go back to reference Polat P, Donofrio PD. Myasthenia gravis induced by nivolumab therapy in a patient with non-small-cell lung cancer. Muscle Nerve. 2016;54:507.CrossRefPubMed Polat P, Donofrio PD. Myasthenia gravis induced by nivolumab therapy in a patient with non-small-cell lung cancer. Muscle Nerve. 2016;54:507.CrossRefPubMed
23.
go back to reference Sciacca G, Nicoletti A, Rampello L, Noto L, Parra HJ, Zappia M. Benign form of myasthenia gravis after nivolumab treatment. Muscle Nerve. 2016;54:507–9.CrossRefPubMed Sciacca G, Nicoletti A, Rampello L, Noto L, Parra HJ, Zappia M. Benign form of myasthenia gravis after nivolumab treatment. Muscle Nerve. 2016;54:507–9.CrossRefPubMed
24.
go back to reference Gonzalez NL, Puwanant A, Lu A, Marks SM, Zivkovic SA. Myasthenia triggered by immune checkpoint inhibitors: new case and literature review. Neuromuscul Disord. 2017;27:266–8.CrossRefPubMed Gonzalez NL, Puwanant A, Lu A, Marks SM, Zivkovic SA. Myasthenia triggered by immune checkpoint inhibitors: new case and literature review. Neuromuscul Disord. 2017;27:266–8.CrossRefPubMed
25.
go back to reference Kimura T, Fukushima S, Miyashita A, Aoi J, Jinnin M, Kosaka T, et al. Myasthenic crisis and polymyositis induced by one dose of nivolumab. Cancer Sci. 2016;107:1055–8.CrossRefPubMedPubMedCentral Kimura T, Fukushima S, Miyashita A, Aoi J, Jinnin M, Kosaka T, et al. Myasthenic crisis and polymyositis induced by one dose of nivolumab. Cancer Sci. 2016;107:1055–8.CrossRefPubMedPubMedCentral
26.
go back to reference Chang E, Sabichi AL, Sada YH. Myasthenia gravis after nivolumab therapy for squamous cell carcinoma of the bladder. J Immunother. 2017;40:114–6.CrossRefPubMed Chang E, Sabichi AL, Sada YH. Myasthenia gravis after nivolumab therapy for squamous cell carcinoma of the bladder. J Immunother. 2017;40:114–6.CrossRefPubMed
27.
go back to reference March KL, Samarin MJ, Sodhi A, Owens RE. Pembrolizumab-induced myasthenia gravis: a fatal case report. J Oncol Pharm Pract. 2017;1078155216687389. March KL, Samarin MJ, Sodhi A, Owens RE. Pembrolizumab-induced myasthenia gravis: a fatal case report. J Oncol Pharm Pract. 2017;1078155216687389.
28.
go back to reference Shirai T, Sano T, Kamijo F, Saito N, Miyake T, Kodaira M, et al. Acetylcholine receptor binding antibody-associated myasthenia gravis and rhabdomyolysis induced by nivolumab in a patient with melanoma. Jpn J Clin Oncol. 2016;46:86–8.CrossRefPubMed Shirai T, Sano T, Kamijo F, Saito N, Miyake T, Kodaira M, et al. Acetylcholine receptor binding antibody-associated myasthenia gravis and rhabdomyolysis induced by nivolumab in a patient with melanoma. Jpn J Clin Oncol. 2016;46:86–8.CrossRefPubMed
29.
go back to reference Mehta JJ, Maloney E, Srinivasan S, Seitz P, Cannon M. Myasthenia gravis induced by nivolumab: a case report. Cureus. 2017;9:e1702.PubMedPubMedCentral Mehta JJ, Maloney E, Srinivasan S, Seitz P, Cannon M. Myasthenia gravis induced by nivolumab: a case report. Cureus. 2017;9:e1702.PubMedPubMedCentral
30.
go back to reference Loochtan AI, Nickolich MS, Hobson-Webb LD. Myasthenia gravis associated with ipilimumab and nivolumab in the treatment of small cell lung cancer. Muscle Nerve. 2015;52:307–8.CrossRefPubMed Loochtan AI, Nickolich MS, Hobson-Webb LD. Myasthenia gravis associated with ipilimumab and nivolumab in the treatment of small cell lung cancer. Muscle Nerve. 2015;52:307–8.CrossRefPubMed
31.
go back to reference Makarious D, Horwood K, Coward JIG. Myasthenia gravis: an emerging toxicity of immune checkpoint inhibitors. Eur J Cancer. 2017;82:128–36.CrossRefPubMed Makarious D, Horwood K, Coward JIG. Myasthenia gravis: an emerging toxicity of immune checkpoint inhibitors. Eur J Cancer. 2017;82:128–36.CrossRefPubMed
32.
go back to reference Huh SY, Shin SH, Kim MK, Lee SY, Son KH, Shin HY. Emergence of myasthenia gravis with myositis in a patient treated with pembrolizumab for thymic cancer. J Clin Neurol. 2018;14:115–7.CrossRefPubMed Huh SY, Shin SH, Kim MK, Lee SY, Son KH, Shin HY. Emergence of myasthenia gravis with myositis in a patient treated with pembrolizumab for thymic cancer. J Clin Neurol. 2018;14:115–7.CrossRefPubMed
33.
go back to reference Chen YH, Liu FC, Hsu CH, Chian CF. Nivolumab-induced myasthenia gravis in a patient with squamous cell lung carcinoma: case report. Medicine (Baltimore). 2017;96:e7350.CrossRef Chen YH, Liu FC, Hsu CH, Chian CF. Nivolumab-induced myasthenia gravis in a patient with squamous cell lung carcinoma: case report. Medicine (Baltimore). 2017;96:e7350.CrossRef
34.
go back to reference Chen JH, Lee KY, Hu CJ, Chung CC. Coexisting myasthenia gravis, myositis, and polyneuropathy induced by ipilimumab and nivolumab in a patient with non-small-cell lung cancer: a case report and literature review. Medicine (Baltimore). 2017;96:e9262.CrossRef Chen JH, Lee KY, Hu CJ, Chung CC. Coexisting myasthenia gravis, myositis, and polyneuropathy induced by ipilimumab and nivolumab in a patient with non-small-cell lung cancer: a case report and literature review. Medicine (Baltimore). 2017;96:e9262.CrossRef
35.
go back to reference Tan RYC, Toh CK, Takano A. Continued response to one dose of nivolumab complicated by myasthenic crisis and myositis. J Thorac Oncol. 2017;12:e90–1.CrossRefPubMed Tan RYC, Toh CK, Takano A. Continued response to one dose of nivolumab complicated by myasthenic crisis and myositis. J Thorac Oncol. 2017;12:e90–1.CrossRefPubMed
36.
go back to reference Fellner A, Makranz C, Lotem M, Bokstein F, Taliansky A, Rosenberg S, et al. Neurologic complications of immune checkpoint inhibitors. J Neuro-Oncol. 2018;137:601–9.CrossRef Fellner A, Makranz C, Lotem M, Bokstein F, Taliansky A, Rosenberg S, et al. Neurologic complications of immune checkpoint inhibitors. J Neuro-Oncol. 2018;137:601–9.CrossRef
37.
go back to reference •• Suzuki S, Ishikawa N, Konoeda F, et al. Nivolumab-related myasthenia gravis with myositis and myocarditis in Japan. Neurology. 2017;89:1127-34. A well-designed study of myasthenia gravis in nivolumab-treated patients highlighting the differences between myasthenia gravis patients with and without nivolumab exposure and the myasthenia gravis-myositis-myocarditis overlap syndrome. CrossRefPubMed •• Suzuki S, Ishikawa N, Konoeda F, et al. Nivolumab-related myasthenia gravis with myositis and myocarditis in Japan. Neurology. 2017;89:1127-34. A well-designed study of myasthenia gravis in nivolumab-treated patients highlighting the differences between myasthenia gravis patients with and without nivolumab exposure and the myasthenia gravis-myositis-myocarditis overlap syndrome. CrossRefPubMed
38.
go back to reference Agata Y, Kawasaki A, Nishimura H, Ishida Y, Tsubat T, Yagita H, et al. Expression of the PD-1 antigen on the surface of stimulated mouse T and B lymphocytes. Int Immunol. 1996;8:765–72.CrossRefPubMed Agata Y, Kawasaki A, Nishimura H, Ishida Y, Tsubat T, Yagita H, et al. Expression of the PD-1 antigen on the surface of stimulated mouse T and B lymphocytes. Int Immunol. 1996;8:765–72.CrossRefPubMed
39.
go back to reference Thibult ML, Mamessier E, Gertner-Dardenne J, Pastor S, Just-Landi S, Xerri L, et al. PD-1 is a novel regulator of human B-cell activation. Int Immunol. 2013;25:129–37.CrossRefPubMed Thibult ML, Mamessier E, Gertner-Dardenne J, Pastor S, Just-Landi S, Xerri L, et al. PD-1 is a novel regulator of human B-cell activation. Int Immunol. 2013;25:129–37.CrossRefPubMed
40.
go back to reference Vallet H, Gaillet A, Weiss N, Vanhaecke C, Saheb S, Touitou V, et al. Pembrolizumab-induced necrotic myositis in a patient with metastatic melanoma. Ann Oncol. 2016;27:1352–3.CrossRefPubMed Vallet H, Gaillet A, Weiss N, Vanhaecke C, Saheb S, Touitou V, et al. Pembrolizumab-induced necrotic myositis in a patient with metastatic melanoma. Ann Oncol. 2016;27:1352–3.CrossRefPubMed
41.
go back to reference Suzuki S, Utsugisawa K, Yoshikawa H, Motomura M, Matsubara S, Yokoyama K, et al. Autoimmune targets of heart and skeletal muscles in myasthenia gravis. Arch Neurol. 2009;66:1334–8.CrossRefPubMed Suzuki S, Utsugisawa K, Yoshikawa H, Motomura M, Matsubara S, Yokoyama K, et al. Autoimmune targets of heart and skeletal muscles in myasthenia gravis. Arch Neurol. 2009;66:1334–8.CrossRefPubMed
42.
go back to reference Santos E, Coutinho E, Martins da Silva A, Marinho A, Vasconcelos C, Taipa R, et al. Inflammatory myopathy associated with myasthenia gravis with and without thymic pathology: report of four cases and literature review. Autoimmun Rev. 2017;16:644–9.CrossRefPubMed Santos E, Coutinho E, Martins da Silva A, Marinho A, Vasconcelos C, Taipa R, et al. Inflammatory myopathy associated with myasthenia gravis with and without thymic pathology: report of four cases and literature review. Autoimmun Rev. 2017;16:644–9.CrossRefPubMed
43.
go back to reference Bilen MA, Subudhi SK, Gao J, Tannir NM, Tu SM, Sharma P. Acute rhabdomyolysis with severe polymyositis following ipilimumab-nivolumab treatment in a cancer patient with elevated anti-striated muscle antibody. J Immunother Cancer. 2016;4:36.CrossRefPubMedPubMedCentral Bilen MA, Subudhi SK, Gao J, Tannir NM, Tu SM, Sharma P. Acute rhabdomyolysis with severe polymyositis following ipilimumab-nivolumab treatment in a cancer patient with elevated anti-striated muscle antibody. J Immunother Cancer. 2016;4:36.CrossRefPubMedPubMedCentral
44.
go back to reference Juel VC. Myasthenia gravis: management of myasthenic crisis and perioperative care. Semin Neurol. 2004;24:75–81.CrossRefPubMed Juel VC. Myasthenia gravis: management of myasthenic crisis and perioperative care. Semin Neurol. 2004;24:75–81.CrossRefPubMed
45.
go back to reference Grob D, Brunner N, Namba T, Pagala M. Lifetime course of myasthenia gravis. Muscle Nerve. 2008;37:141–9.CrossRefPubMed Grob D, Brunner N, Namba T, Pagala M. Lifetime course of myasthenia gravis. Muscle Nerve. 2008;37:141–9.CrossRefPubMed
46.
go back to reference Johnson DB, Balko JM, Compton ML, Chalkias S, Gorham J, Xu Y, et al. Fulminant myocarditis with combination immune checkpoint blockade. N Engl J Med. 2016;375:1749–55.CrossRefPubMedPubMedCentral Johnson DB, Balko JM, Compton ML, Chalkias S, Gorham J, Xu Y, et al. Fulminant myocarditis with combination immune checkpoint blockade. N Engl J Med. 2016;375:1749–55.CrossRefPubMedPubMedCentral
47.
go back to reference Gandiga PC, Wang AR, Gonzalez-Rivera T, Sreih AG. Pembrolizumab-associated inflammatory myopathy. Rheumatology (Oxford). 2018;57:397–8.CrossRef Gandiga PC, Wang AR, Gonzalez-Rivera T, Sreih AG. Pembrolizumab-associated inflammatory myopathy. Rheumatology (Oxford). 2018;57:397–8.CrossRef
48.
go back to reference Bourgeois-Vionnet J, Joubert B, Bernard E, Sia MA, Pante V, Fabien N, et al. Nivolumab-induced myositis: a case report and a literature review. J Neurol Sci. 2018;387:51–3.CrossRefPubMed Bourgeois-Vionnet J, Joubert B, Bernard E, Sia MA, Pante V, Fabien N, et al. Nivolumab-induced myositis: a case report and a literature review. J Neurol Sci. 2018;387:51–3.CrossRefPubMed
49.
go back to reference Uchio N, Taira K, Ikenaga C, Unuma A, Kadoya M, Kubota A, et al. Granulomatous myositis induced by anti-PD-1 monoclonal antibodies. Neurol Neuroimmunol Neuroinflamm. 2018;5:e464.CrossRefPubMedCentral Uchio N, Taira K, Ikenaga C, Unuma A, Kadoya M, Kubota A, et al. Granulomatous myositis induced by anti-PD-1 monoclonal antibodies. Neurol Neuroimmunol Neuroinflamm. 2018;5:e464.CrossRefPubMedCentral
50.
go back to reference Yoshioka M, Kambe N, Yamamoto Y, Suehiro K, Matsue H. Case of respiratory discomfort due to myositis after administration of nivolumab. J Dermatol. 2015;42:1008–9.CrossRefPubMed Yoshioka M, Kambe N, Yamamoto Y, Suehiro K, Matsue H. Case of respiratory discomfort due to myositis after administration of nivolumab. J Dermatol. 2015;42:1008–9.CrossRefPubMed
52.
go back to reference Diamantopoulos PT, Tsatsou K, Benopoulou O, Anastasopoulou A, Gogas H. Inflammatory myopathy and axonal neuropathy in a patient with melanoma following pembrolizumab treatment. J Immunother. 2017;40:221–3.CrossRefPubMed Diamantopoulos PT, Tsatsou K, Benopoulou O, Anastasopoulou A, Gogas H. Inflammatory myopathy and axonal neuropathy in a patient with melanoma following pembrolizumab treatment. J Immunother. 2017;40:221–3.CrossRefPubMed
53.
go back to reference Saini L, Chua N. Severe inflammatory myositis in a patient receiving concurrent nivolumab and azacitidine. Leuk Lymphoma. 2017;58:2011–3.CrossRefPubMed Saini L, Chua N. Severe inflammatory myositis in a patient receiving concurrent nivolumab and azacitidine. Leuk Lymphoma. 2017;58:2011–3.CrossRefPubMed
54.
go back to reference Behling J, Kaes J, Munzel T, Grabbe S, Loquai C. New-onset third-degree atrioventricular block because of autoimmune-induced myositis under treatment with anti-programmed cell death-1 (nivolumab) for metastatic melanoma. Melanoma Res. 2017;27:155–8.CrossRefPubMed Behling J, Kaes J, Munzel T, Grabbe S, Loquai C. New-onset third-degree atrioventricular block because of autoimmune-induced myositis under treatment with anti-programmed cell death-1 (nivolumab) for metastatic melanoma. Melanoma Res. 2017;27:155–8.CrossRefPubMed
55.
go back to reference Min L, Hodi FS. Anti-PD1 following ipilimumab for mucosal melanoma: durable tumor response associated with severe hypothyroidism and rhabdomyolysis. Cancer Immunol Res. 2014;2:15–8.CrossRefPubMed Min L, Hodi FS. Anti-PD1 following ipilimumab for mucosal melanoma: durable tumor response associated with severe hypothyroidism and rhabdomyolysis. Cancer Immunol Res. 2014;2:15–8.CrossRefPubMed
56.
go back to reference Milone M. Diagnosis and management of immune-mediated myopathies. Mayo Clin Proc. 2017;92:826–37.CrossRefPubMed Milone M. Diagnosis and management of immune-mediated myopathies. Mayo Clin Proc. 2017;92:826–37.CrossRefPubMed
57.
go back to reference Moslehi JJ, Salem JE, Sosman JA, Lebrun-Vignes B, Johnson DB. Increased reporting of fatal immune checkpoint inhibitor-associated myocarditis. Lancet. 2018;391:933.CrossRefPubMedPubMedCentral Moslehi JJ, Salem JE, Sosman JA, Lebrun-Vignes B, Johnson DB. Increased reporting of fatal immune checkpoint inhibitor-associated myocarditis. Lancet. 2018;391:933.CrossRefPubMedPubMedCentral
58.
go back to reference Gordon SR, Maute RL, Dulken BW, Hutter G, George BM, McCracken MN, et al. PD-1 expression by tumour-associated macrophages inhibits phagocytosis and tumour immunity. Nature. 2017;545:495–9.CrossRefPubMedPubMedCentral Gordon SR, Maute RL, Dulken BW, Hutter G, George BM, McCracken MN, et al. PD-1 expression by tumour-associated macrophages inhibits phagocytosis and tumour immunity. Nature. 2017;545:495–9.CrossRefPubMedPubMedCentral
59.
go back to reference Kahler KC, Hassel JC, Heinzerling L, et al. Management of side effects of immune checkpoint blockade by anti-CTLA-4 and anti-PD-1 antibodies in metastatic melanoma. J Dtsch Dermatol Ges. 2016;14:662–81.PubMed Kahler KC, Hassel JC, Heinzerling L, et al. Management of side effects of immune checkpoint blockade by anti-CTLA-4 and anti-PD-1 antibodies in metastatic melanoma. J Dtsch Dermatol Ges. 2016;14:662–81.PubMed
60.
go back to reference Yost MD, Chou CZ, Botha H, Block MS, Liewluck T. Facial diplegia after pembrolizumab treatment. Muscle Nerve. 2017;56:E20–1.CrossRefPubMed Yost MD, Chou CZ, Botha H, Block MS, Liewluck T. Facial diplegia after pembrolizumab treatment. Muscle Nerve. 2017;56:E20–1.CrossRefPubMed
61.
go back to reference Ong S, Chapman J, Young G, Mansy T. Guillain-Barre-like syndrome during pembrolizumab treatment. Muscle Nerve. 2018;58:E8-E10.CrossRef Ong S, Chapman J, Young G, Mansy T. Guillain-Barre-like syndrome during pembrolizumab treatment. Muscle Nerve. 2018;58:E8-E10.CrossRef
62.
go back to reference Jacob A, Unnikrishnan DC, Mathew A, Thyagarajan B, Patel S. A case of fatal Guillain-Barre syndrome from anti-PD1 monoclonal antibody use. J Cancer Res Clin Oncol. 2016;142:1869–70.CrossRefPubMed Jacob A, Unnikrishnan DC, Mathew A, Thyagarajan B, Patel S. A case of fatal Guillain-Barre syndrome from anti-PD1 monoclonal antibody use. J Cancer Res Clin Oncol. 2016;142:1869–70.CrossRefPubMed
63.
go back to reference Schneiderbauer R, Schneiderbauer M, Wick W, Enk AH, Haenssle HA, Hassel JC. PD-1 antibody-induced Guillain-Barre syndrome in a patient with metastatic melanoma. Acta Derm Venereol. 2017;97:395–6.CrossRefPubMed Schneiderbauer R, Schneiderbauer M, Wick W, Enk AH, Haenssle HA, Hassel JC. PD-1 antibody-induced Guillain-Barre syndrome in a patient with metastatic melanoma. Acta Derm Venereol. 2017;97:395–6.CrossRefPubMed
64.
go back to reference de Maleissye MF, Nicolas G, Saiag P. Pembrolizumab-induced demyelinating polyradiculoneuropathy. N Engl J Med. 2016;375:296–7.CrossRefPubMed de Maleissye MF, Nicolas G, Saiag P. Pembrolizumab-induced demyelinating polyradiculoneuropathy. N Engl J Med. 2016;375:296–7.CrossRefPubMed
65.
go back to reference Fukumoto Y, Kuwahara M, Kawai S, Nakahama K, Kusunoki S. Acute demyelinating polyneuropathy induced by nivolumab. J Neurol Neurosurg Psychiatry. 2018;89:435–7.CrossRefPubMed Fukumoto Y, Kuwahara M, Kawai S, Nakahama K, Kusunoki S. Acute demyelinating polyneuropathy induced by nivolumab. J Neurol Neurosurg Psychiatry. 2018;89:435–7.CrossRefPubMed
66.
go back to reference Supakornnumporn S, Katirji B. Guillain-Barre syndrome triggered by immune checkpoint inhibitors: a case report and literature review. J Clin Neuromuscul Dis. 2017;19:80–3.CrossRefPubMed Supakornnumporn S, Katirji B. Guillain-Barre syndrome triggered by immune checkpoint inhibitors: a case report and literature review. J Clin Neuromuscul Dis. 2017;19:80–3.CrossRefPubMed
67.
go back to reference Dimachkie MM, Saperstein DS. Acquired immune demyelinating neuropathies. Continuum (Minneap Minn). 2014;20:1241–60. Dimachkie MM, Saperstein DS. Acquired immune demyelinating neuropathies. Continuum (Minneap Minn). 2014;20:1241–60.
68.
go back to reference Tanaka R, Maruyama H, Tomidokoro Y, Yanagiha K, Hirabayashi T, Ishii A, et al. Nivolumab-induced chronic inflammatory demyelinating polyradiculoneuropathy mimicking rapid-onset Guillain-Barre syndrome: a case report. Jpn J Clin Oncol. 2016;46:875–8.CrossRefPubMed Tanaka R, Maruyama H, Tomidokoro Y, Yanagiha K, Hirabayashi T, Ishii A, et al. Nivolumab-induced chronic inflammatory demyelinating polyradiculoneuropathy mimicking rapid-onset Guillain-Barre syndrome: a case report. Jpn J Clin Oncol. 2016;46:875–8.CrossRefPubMed
69.
go back to reference Sepulveda M, Martinez-Hernandez E, Gaba L, et al. Motor polyradiculopathy during pembrolizumab treatment of metastatic melanoma. Muscle Nerve. 2017;56:E162–7.CrossRefPubMed Sepulveda M, Martinez-Hernandez E, Gaba L, et al. Motor polyradiculopathy during pembrolizumab treatment of metastatic melanoma. Muscle Nerve. 2017;56:E162–7.CrossRefPubMed
70.
go back to reference Aya F, Ruiz-Esquide V, Viladot M, Font C, Prieto-González S, Prat A, et al. Vasculitic neuropathy induced by pembrolizumab. Ann Oncol. 2017;28:433–4.PubMed Aya F, Ruiz-Esquide V, Viladot M, Font C, Prieto-González S, Prat A, et al. Vasculitic neuropathy induced by pembrolizumab. Ann Oncol. 2017;28:433–4.PubMed
Metadata
Title
Neuromuscular Complications of Programmed Cell Death-1 (PD-1) Inhibitors
Authors
Justin C. Kao
Adipong Brickshawana
Teerin Liewluck
Publication date
01-10-2018
Publisher
Springer US
Published in
Current Neurology and Neuroscience Reports / Issue 10/2018
Print ISSN: 1528-4042
Electronic ISSN: 1534-6293
DOI
https://doi.org/10.1007/s11910-018-0878-7

Other articles of this Issue 10/2018

Current Neurology and Neuroscience Reports 10/2018 Go to the issue

Headache (R B Halker, Section Editor)

The Neuralgias

Sleep (M Thorpy and M Billiard, Section Editors)

Sleep in Parkinson’s Disease with Impulse Control Disorder

Sleep (M Thorpy and M Billiard, Section Editors)

Circadian Rhythm Disturbances in the Blind