Elsevier

European Journal of Cancer

Volume 82, September 2017, Pages 128-136
European Journal of Cancer

Review
Myasthenia gravis: An emerging toxicity of immune checkpoint inhibitors

https://doi.org/10.1016/j.ejca.2017.05.041Get rights and content

Highlights

  • Twenty-three reported cases of immune checkpoint inhibitor-associated myasthenia gravis (MG).

  • Approximately, 70% of cases were de novo presentations.

  • There is a 30.4% MG-specific mortality associated with immune checkpoint inhibition.

  • Average onset of MG symptoms is within 6 weeks of treatment initiation.

  • Early recognition of symptoms and robust treatment is essential.

Abstract

The advent of immunotherapy has heralded a number of significant advances in the treatment of particular malignancies associated with poor prognosis (melanoma, non-small-cell lung, renal and head/neck cancers). The success witnessed with therapeutic agents targeting cytotoxic T-lymphocyte-associated protein 4, programmed cell death protein 1 and programmed cell death ligand 1 immune checkpoints has inevitably led to an explosion in their clinical application and the subsequent recognition of specific toxicity profiles distinct from those long recognised with chemotherapy. Consequently, as the utility of such therapies broaden, understanding the nature, timing and management of these immune-related adverse events (irAEs) becomes increasingly significant. Although neurological irAEs are considered relatively rare in comparison with hepatitis, colitis, pneumonitis and endocrinopathies, one emerging side-effect is myasthenia gravis (MG). Among the 23 reported cases of immune checkpoint inhibitor-associated MG, 72.7% were de novo presentations, 18.2% were exacerbations of pre-existing MG and 9.1% were exacerbations of subclinical MG. The average onset of symptoms was within 6 weeks (range 2–12 weeks) of treatment initiation. In addition, there was no consistent association with elevated acetylcholine antibody titres and the development of immune checkpoint inhibitor-related MG. Significantly, there was a 30.4% MG-specific-related mortality, which further emphasises the importance of early recognition and robust treatment of this toxicity. In addition to a review of the existing literature, we present a new case of pembrolizumab-induced MG and provide insights into the underlying mechanisms of action of this phenomenon.

Introduction

Over the past 5 years, targeted immunotherapy has created a major paradigm shift within the therapeutic landscape of numerous solid tumours. Manipulation of pathways which mediate blunting of anti-tumour immunity has predominantly focussed on the development of inhibitors directed against the immune checkpoint modulators such as cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), programmed cell death protein 1 (PD-1) and its ligand, PD-L1. The introduction of these novel agents has not only witnessed unprecedented extensions in survival outcomes for patients with malignant melanoma, lung, renal and head/neck cancers but reveals a plethora of immune-related toxicities for which early recognition and appropriate clinical management are paramount. Autoimmune events including hepatitis, colitis, pneumonitis, dermatitis, nephritis, endocrinopathies and hypophysitis are well-recognised phenomena which represent a manifestation of dysregulated inflammation induced by immune checkpoint inhibition. As the use of these targeted agents expand, further toxicity issues have emerged. Recently, this has been exemplified by both de novo presentations and exacerbations of pre-existing myasthenia gravis (MG) which, to date, has been reported in 22 cases in the literature. Herein, we present a new presentation of ocular MG (oMG) followed by a succinct review of cases highlighting this phenomenon.

Section snippets

Case report

An 85-year-old woman with metastatic melanoma with left axillary lymphadenopathy commenced single-agent pembrolizumab (2 mg/kg; every 3 weeks) and tolerated the first cycle without any significant toxicity. Shortly after the second cycle, she presented with diplopia that was subsequently followed by asymmetrical bilateral ptosis (L > R). The remaining neurological examination was unremarkable, and there was no evidence of oesophageal dysmotility or respiratory compromise. Brain magnetic

Anti-PD-1 inhibitor-induced myasthenia gravis

In addition to this aforementioned report, since the introduction of PD-1 and PD-L1 immune checkpoint inhibitors, there have been several publications highlighting a causal relationship with the manifestation of de novo MG (Table 1).

Indeed, March et al. [1] recently documented a fatal case in a 63-year-old male with malignant melanoma of the right scalp, which had metastasised to the liver and brain, who was also treated with pembrolizumab. Two weeks after the first cycle, he presented with

Anti-CTLA-4 inhibitor-induced myasthenia gravis

To date, there have been four cases of MG with the anti-CTLA-4 antibody, ipilimumab; all of which have been de novo presentations [12], [13], [14]. Intriguingly, all cases occurred within 6 weeks of initiation of ipilimumab and were not associated with any mortality. Liao et al. [12] reported a case of a 70-year-old female with uveal melanoma and liver metastases who was commenced on ipilimumab. After the second infusion, she developed generalised myalgia, dysphagia, odynophagia, bilateral

Combination immune checkpoint inhibitor therapy-induced myasthenia gravis

Currently, there are two reports of combination immune checkpoint inhibitor therapy inducing MG. The first case by Antonia et al. [15] notes a patient who received durvalumab (anti-PD-L1 inhibitor; 10 mg/kg) and tremelimumab (anti-CTLA-4 inhibitor; 1 mg/kg) delivered on a 4-week schedule [15]. The patient was diagnosed with advanced NSCLC and developed MG within 10 d of initiating treatment. Unfortunately, there is a paucity of further information on the patient profile and treatment history

PD-1 inhibitor exacerbation of myasthenia gravis

In addition to de novo MG, there have been four such reports [18], [19], [20], [21] of patients with exacerbations of pre-existing MG as a direct consequence of immune checkpoint inhibition.

One such case reported by Lau et al. [18] documents a 75-year-old man, with a known history of non-thymomatous AChR-Ab positive generalised MG maintained on azathioprine, who was diagnosed with metastatic melanoma. Two weeks after the second cycle of pembrolizumab, he presented with ataxia, neck weakness and

Discussion

Historically, the excitement generated by the introduction of novel therapies within the treatment armament for oncology patients is also moderated by the emergence of an often unique constellation of toxicities which require prompt recognition and robust clinical management. This is particularly emphasised by the immune-related adverse events (irAEs) associated with immune checkpoint inhibitors. Zimmer et al. [6] published a study of 496 patients being treated with anti-PD-1 therapy in which

Conclusion

With the advent of immune checkpoint inhibitors, there have been significant advances in the treatment of a number of malignancies. However, the recognition of timing and treatment of irAEs with these therapies is integral to the clinical management of patients undertaking immunotherapy. Although neurological sequelae of immune checkpoint inhibitors are relatively rare, MG, in particular, is becoming an increasingly recognised phenomenon, with potentially fatal outcomes witnessed in just under

Conflict of interest statement

None declared.

References (34)

  • T. Kimura et al.

    Myasthenic crisis and polymyositis induced by one dose of nivolumab

    Cancer Sci

    (2016)
  • I. Alnahhas et al.

    A case of new onset antibody-positive myasthenia gravis in a patient treated with pembrolizumab for melanoma

    Muscle Nerve

    (2017 Jun)
  • P. Polat et al.

    Myasthenia gravis induced by nivolumab therapy in a patient with non-small-cell lung cancer

    Muscle Nerve

    (2016)
  • G. Sciacca et al.

    Benign form of myasthenia gravis after nivolumab treatment

    Muscle Nerve

    (2016)
  • E. Chang et al.

    Myasthenia gravis after nivolumab therapy for squamous cell carcinoma of the bladder

    J Immunother

    (2017)
  • D. Lopez et al.

    Myasthenia gravis and rhabdomyolysis in a patient with advanced renal cell cancer treated with nivolumab: a case report and review of the literature

    Br J Med Health Res

    (2015)
  • B. Liao et al.

    Atypical neurological complications of ipilimumab therapy in patients with metastatic melanoma

    Neuro Oncol

    (2014)
  • Cited by (197)

    • Treatment of rheumatic adverse events of cancer immunotherapy

      2022, Best Practice and Research: Clinical Rheumatology
      Citation Excerpt :

      For ICI-myositis with muscle weakness severely limiting mobility and/or cardiac or respiratory compromise, the recommendation is for high-dose IV steroids at 1–2 mg/kg per day but with strong consideration for pulse dose steroids with 500–1000 mg IV solumedrol per day for 3–5 days. For severe and/or rapidly progressing presentations, IVIg should be considered and is particularly effective for cases with myasthenia gravis [22,92]. In general, treatment beyond corticosteroids will be impacted by the severity and extent of neuromuscular involvement and/or cardiac involvement, as well as the acuity of the patient's clinical status.

    View all citing articles on Scopus
    View full text