J Med Cases
Journal of Medical Cases, ISSN 1923-4155 print, 1923-4163 online, Open Access
Article copyright, the authors; Journal compilation copyright, J Med Cases and Elmer Press Inc
Journal website https://www.journalmc.org

Case Report

Volume 12, Number 10, October 2021, pages 424-428


Nivolumab, a Double-Edged Sword: A Case Report of Nivolumab-Induced Myasthenia Gravis

Nayha Tahira, e, Anber Mahboobb, Xuanzhen Piaoc, Grace Yinga, Jishna Shresthaa, Robin Sherchana, Farha Zahrad

aChicago Medical School, Rosalind Franklin University of Medicine and Science, Internal Medicine Residency Program at Northwestern Medicine Hospital, McHenry, IL, USA
bSharif Medical and Dental College, Lahore, Pakistan
cChicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
dDepartment of Internal Medicine, Northwestern Medicine McHenry Hospital, McHenry, IL, USA
eCorresponding Author: Nayha Tahir, Chicago Medical School, Rosalind Franklin University of Medicine and Science, Internal Medicine Residency Program at Northwestern Medicine Hospital, McHenry, IL, USA

Manuscript submitted September 3, 2021, accepted September 22, 2021, published online September 29, 2021
Short title: Nivolumab: A Double-Edged Sword
doi: https://doi.org/10.14740/jmc3783

Abstract▴Top 

Nivolumab is a checkpoint inhibitor approved to treat various solid organs malignancies. Although checkpoint inhibitors are very efficacious, these medications are also associated with a variety of side effects that could be life-threatening. We present a case of nivolumab-induced myasthenia gravis in a patient with stage IV esophageal cancer, who was found to have generalized weakness, blurry vision, diplopia, and later developed acute hypoxic respiratory failure with subsequent intubation. The patient was treated with intravenous immunoglobulin and plasmapheresis, and later started on pyridostigmine and high-dose steroids with minimal improvement. Goals of care were discussed with the patient and family, and the decision was made to discharge the patient home with hospice care. Nivolumab-induced myasthenia gravis is very aggressive with a poor prognosis if not appropriately managed in time. Hence we strongly recommend a multidisciplinary approach, including neurologists, to monitor patients on nivolumab therapy to reduce morbidity and mortality associated with it.

Keywords: Nivolumab; Myasthenia gravis; Esophageal cancer; PD-L1 inhibitors

Introduction▴Top 

Nivolumab is a monoclonal antibody belonging to the checkpoint inhibitors, recently approved to treat recurrent or metastatic non-small cell lung cancer, melanoma, renal cell carcinoma, and advanced esophageal carcinoma [1]. The use of checkpoint inhibitors against various molecules, including programmed cell death protein-1 (PD-1), programmed death ligand-1 (PD-L1), and cytotoxic T-lymphocyte-associated protein-4 (CTLA-4), has become widespread in clinical practice [1]. However, although very efficacious, these medications can induce various adverse effects, the full spectrum of which is not yet thoroughly characterized.

This case highlights a case of PD-1 inhibitor-induced myasthenia gravis (MG) in a patient with stage IV esophageal cancer: a severe adverse event that has been increasingly observed with the use of PD-1/PD-L1 inhibitors.

Case Report▴Top 

Investigations

A 76-year-old man presented to the emergency department after a mechanical fall at home with head trauma. He attempted to get up from a chair using a cane and fell to the floor. His past medical history includes metastatic esophageal adenocarcinoma status post chemotherapy and radiation, radiation-induced esophageal stricture status post nasogastric tube placement, bilateral malignant pleural effusion, complete heart block status post dual-chamber pacemaker placement 1 month before presentation, recent non-ST segment elevation myocardial infarction with a normal nuclear stress test, chronic diastolic heart failure with a preserved ejection fraction of 79%, chronic normocytic anemia with baseline hemoglobin of 7.7 - 8.9 g/dL, type 2 diabetes mellitus, hypertension, hyperlipidemia, and history of prostate cancer status post radiation. The patient stated that he was also having trouble keeping his eyes open for 2 weeks before presentation, so he underwent an outpatient magnetic resonance imaging (MRI) of the brain with and without contrast, which was negative for intracranial metastasis or hemorrhage. The patient’s wife denied observing seizure activities, urinary or fecal incontinence, and slurred speech, and stated that the patient was likely unresponsive for a few seconds. Patient denied any prodromal symptoms such as chest pain, palpitation, shortness of breath, headaches, dizziness, nausea, vomiting, weakness, or numbness.

The patient was a former smoker who quit about 35 years ago, with no history of alcohol or any recreational drugs use. Family history was pertinent for ovarian cancer in mother and lung cancer in father. Home medications include aspirin, carvedilol, furosemide, metformin, and rosuvastatin. The patient was started on radiation therapy with concurrent chemotherapy with weekly carboplatin and paclitaxel 6 months before presentation. The patient tolerated chemotherapy for 6 weeks with interruption of treatment secondary to neutropenia and was then initiated on FOLFOX chemotherapy with pegfilgrastim and nivolumab 3 and 2 months before presentation, respectively. Off note, interval reduction of chemotherapy dose took place about 5 weeks ago secondary to thrombocytopenia, and 4 weeks ago chemotherapy was held secondary to generalized weakness and functional deconditioning.

On presentation, patients’ blood pressure was 121/69 mm Hg, heart rate 83 beats per minute (bpm), temperature 36.6 °C, respiratory rate 13 breaths per minute, and oxygen saturation 96% on room air. He was alert, awake, and oriented to person, time, place, and situation. On the physical exam, the patient was noted to have mild bibasilar crackles. Additionally, bilateral ptosis was noted. Otherwise, no cranial nerve or focal neurological deficits were noted. The rest of the physical examination was unremarkable.

Diagnosis

Basic labs are included in Table 1. Electrocardiogram (EKG) showed ventricular-paced rhythm at 87 bpm with no acute ischemic changes. Computed tomography (CT) of the head showed scattered areas of subarachnoid hemorrhage involving the left frontal, right temporal and inferior right frontal lobes without significant mass or midline shift. CT angiogram of the head was negative for significant stenosis or aneurysm in the head or neck arterial vasculature. CT cervical spine without contrast was negative for acute fractures. Per neurosurgery, no surgical intervention was indicated.

Table 1.
Click to view
Table 1. Basic Labs of the Patient
 

Treatment

The patient was then started on levetiracetam for seizure prophylaxis. The repeat CT head without contrast demonstrated stable findings.

Follow-up and outcomes

A few days later, the patient started complaining of worsened blurry vision, fatigue, and diplopia in both extremes of horizontal gaze. On the physical exam, severe bilateral ptosis was noted to the point where the patient could not keep his eyes open without helping with his fingers. In terms of muscle strength, it was showed that neck flexion 3/5, extension 5/5, deltoid 5/5 bilaterally, biceps 4/5 bilaterally, triceps 4/5 bilaterally; hip flexion 2/5 on the right and 3/5 on the left, knee flexion 3/5 on the right and 4/5 on the left, and plantar flexion 4/5 bilaterally. The sensation was intact to light touch, and reflexes were 2+ throughout. The patient remained awake but somnolent, followed basic commands, and used hand signals to answer questions. Overnight, the patient was noted to have an event of unresponsiveness with oxygen saturation of 80% on room air, unable to follow commands. Lung examination revealed significant bibasilar crackles with chest radiograph showing bilateral pleural effusions; and the patient was subsequently intubated and transferred to the intensive care unit due to acute hypoxic hypercapnic respiratory failure.

A preliminary diagnosis of MG with ongoing crisis likely secondary to immune checkpoint inhibitor (ICI) therapy (nivolumab) was made, and a diagnostic workup was started. In addition to suspicion of MG, several other paraneoplastic and autoimmune conditions were also included as part of the differential diagnosis. Acetylcholine receptor modulating, binding, blocking antibodies, anti-smooth muscle antibody with titers, and autoimmune comprehensive neurology antibody panels (Table 2) were sent. Lactate dehydrogenase (LDH) was noted to be elevated up to 471 U/L (reference range: 140 - 280 U/L). Aspartate aminotransferase (AST), alanine aminotransferase (ALT), creatinine kinase (CK), thyroid stimulating hormone (TSH), adrenocorticotropic hormone (ACTH), and cortisol levels were checked and were noted to be within normal limit. CT chest without contrast was negative for any findings concerning thymoma but revealed bilateral pleural effusions. Ultrasound-guided thoracentesis was subsequently performed with 950 mL of slightly cloudy aspirate removal with fluid studies consistent with metastatic disease. Electromyography (EMG) and muscle biopsy was planned by neurology but the patient’s wife denied to pursuing any further diagnostic workup. The patient was started on pyridostigmine and high-dose prednisone by neurology service and also completed 4 days of intravenous immunoglobulin (IVIG) and 5 days of plasmapheresis. Post therapy, our patient could semi-open his eyes and be placed on pressure support for a trial of extubation. The patient expressed his wishes to pursue comfort care. Therefore, a hospice consult was placed, and the patient was discharged home with hospice care on pyridostigmine and prednisone.

Table 2.
Click to view
Table 2. Autoimmune Comprehensive Neurology Antibody Panels
 
Discussion▴Top 

Advances in cancer treatment over the years have established ICIs such as CTLA-4 and PD-1/PD-L1 blocking agents as a standard of care in several cancers [2]. PD-1 binds to specific ligands (PD-L1 (B7-H1) and PD-L2 (B7-DC)) that are expressed on tumor cells, resulting in selective immunosuppression and prevention of the immune system from rejecting the tumor. Our point of discussion is one particular drug, nivolumab, a monoclonal antibody that blocks PD-1, an immune checkpoint receptor expressed by activated T cells, from binding to its ligands [3]. Even with impressive responses to ICIs in a wide variety of malignancies, these agents cause class-related adverse effects, called immune-related adverse events (irAEs), which possibly result from an imbalanced self-activation of a T-cell immune response [4]. Immune checkpoints have an essential role in maintaining self-tolerance and warding off autoimmunity. When these physiologic “blocks” on cell-mediated immunity are removed, it can lead to unfavorable effects on a large scale beyond the tumor site. When these unchecked activated T cells circulate widely targeting self-antigens and inflammatory cytokines, it may result in inflammation and destruction in peripheral tissue, manifesting clinically as an autoimmune disease [5]. Nivolumab-induced irAE can affect all body organs, causing diabetes, hypothyroidism, adrenal insufficiency, interstitial pneumonia, colitis, renal and liver dysfunction, MG, myositis, and many others. Even though MG occurs in a smaller proportion of patients treated with nivolumab than other side effects, it does hold significant importance due to its severity and prognosis. Several cases have been recorded documenting MG development in patients on nivolumab therapy after one or more cycles. The onset and progression remain unclear, and a systematic approach is needed to establish its true incidence [6]. The exact pathogenesis of nivolumab causing MG is not entirely understood yet, but one hypothesis suggests that it promotes antigen-antibody reaction mediated by T lymphocytes or promotes the activation of B lymphocytes by T2 lymphocytes. Another hypothesis suggests that it may potentiate the autoimmune response in patients who were previously sensitive against acetylcholine receptors having positive acetylcholine receptor antibody titers but had no clinical manifestations [7]. One cohort study documented that ICI-related MG was more common in the older age group than patients with spontaneous MG. This finding indicates that elderly patients with cancer might be more susceptible to this particular adverse effect [2]. Multiple cases of ICI-associated MG have also been reported with myalgias and increased creatine phosphokinase (CPK) levels without myositis diagnosis, which suggests that the actual incidence of concurrent myositis could be higher [5]. Various studies have shown that more than one-third of such patients have overlapping myositis/myocarditis with MG, and this particular subset of patients have severe symptoms and worse clinical outcomes than patients with isolated idiopathic MG (iMG) [2]. The data available so far suggest that nivolumab-induced MG is more aggressive, has a worse prognosis, and has higher mortality than spontaneous MG; however, having older mean age at diagnosis and concurrent advanced-stage tumors may influence the prognosis [8]. Approximately 85% of patients with spontaneous MG have antibodies to the acetylcholine receptor, but these antibodies are frequently negative in ICI-mediated MG, and when they are detected, titers are mostly much lower than those seen in spontaneous MG [5]. Therefore, it is suggested to set a low threshold for prompt extensive treatment initiation in patients with iMG, particularly patients with overlap syndrome, i.e., neuropathy/myositis/MG [9]. Diagnostic workup should include acetylcholine receptor antibodies, muscle-specific antibodies, cerebrospinal fluid analysis, as many patients with irAEs show unexpectedly high cerebrospinal fluid cell counts. Other diagnostic modalities should be medical imaging or electroneurography [9]. Findings that support the diagnosis of MG are: 1) positive blood anti-AChR or anti-MuSK antibodies; 2) positive response to cholinesterase inhibitors; and 3) decremental responses to repetitive nerve stimulation or abnormal single fiber EMG [10].

ICI-associated MG is treated by first discontinuing the offending drug and then initiating low- dose corticosteroids with slow titration or methylprednisolone. Plasma exchange (PLEX) or IVIG should be added in severe cases [5]. Previous studies show that patients who received IVIG or PLEX as first-line treatment options experienced better outcomes than those treated with steroids alone. Data also suggest that IVIG or PLEX may be more effective when used as a first-line treatment option as many patients deteriorated despite second-line use of IVIG or PLEX secondary to failed initial treatment with corticosteroids. Provided the acute onset and rapid deterioration, it is recommended to use IVIG or PLEX early in the management of ICI-related MG regardless of the severity of initial symptoms. Another problem with using steroids as the only first-line treatment is their well-known side effect of acute exacerbation of iMG symptoms [2, 11]. Even though this worsening is transient, it occurs in almost 50% of patients, and it has a high potential of progressing to respiratory compromise.

Additionally, steroids need several weeks to show clinical response [12]. Based on documented improvement in outcomes with IVIG and PLEX in the majority of the patients with severe iMG [13-15], their early use is recommended before or simultaneously with steroids to combat the risk of a transient worsening, especially in severe cases [11, 16, 17]. Based on our experience and previously documented literature, we suggest that a multidisciplinary approach [2] should be implemented while treating patients with ICI so that life-threatening adverse effects could be managed in a time-sensitive manner. The team should specifically include neurologists and neuro-oncologists [18], as neurotoxicity can be challenging to diagnose mainly when these agents are administered and monitored by non-neurological professionals hence the risk of underreporting stays high [19]. A standardized stepwise checklist [19] should also be developed to detect the neurological irAEs early, particularly in those hospitals where neurologists are either not readily available or are not directly involved in the care and monitoring of cancer patients [19].

Conclusions

Although ICIs’ exceptional efficacy has transformed the treatment of many cancers, they come with many potentially lethal immune-related adverse effects affecting multiple organs. Nivolumab-induced MG, developed through an unknown mechanism, is very aggressive with poor prognosis if not appropriately managed in time; hence we strongly recommend a multidisciplinary approach, including neurologists, to monitor patients on nivolumab therapy in order to reduce morbidity and mortality associated with it. An aggressive phase IV surveillance can help understand the pathogenesis and probability of MG occurrence and develop an effective management strategy to enhance the safe use of an otherwise excellent treatment option.

Acknowledgments

None to declare.

Financial Disclosure

No financial disclosure or funding to declare.

Conflict of Interest

No known conflict of interest to disclose.

Informed Consent

Not applicable.

Author Contributions

NT wrote the majority of the manuscript. AM, XP, GY, JS, and RS all contributed to writing and revising. FZ assisted in revisions of final draft.

Data Availability

The authors declare that data supporting the findings of this study are available within the article.


References▴Top 
  1. Rajan A, Kim C, Heery CR, Guha U, Gulley JL. Nivolumab, anti-programmed death-1 (PD-1) monoclonal antibody immunotherapy: Role in advanced cancers. Hum Vaccin Immunother. 2016;12(9):2219-2231.
    doi pubmed
  2. Safa H, Johnson DH, Trinh VA, Rodgers TE, Lin H, Suarez-Almazor ME, Fa'ak F, et al. Immune checkpoint inhibitor related myasthenia gravis: single center experience and systematic review of the literature. J Immunother Cancer. 2019;7(1):319.
    doi pubmed
  3. Sosman JA. Immunotherapy of advanced melanoma with immune checkpoint inhibition. Literature review current through: Jun 2021. This topic last updated: Mar 04, 2021. In UpToDate: Accessed on July 19th, 2021.
  4. Cortellini A, Napoleoni L, Cimini N, Parisi A, Pavese F, D'Orazio C, Verna L, et al. Immune checkpoint inhibitors and myasthenic syndromes: a case report of a metastatic renal cell carcinoma patient treated with nivolumab. J Clin Neuromuscul Dis. 2018;20(2):99-100.
    doi pubmed
  5. Haugh AM, Probasco JC, Johnson DB. Neurologic complications of immune checkpoint inhibitors. Expert Opin Drug Saf. 2020;19(4):479-488.
    doi pubmed
  6. Nakanishi S, Nishida S, Miyazato M, Goya M, Saito S. A case report of nivolumab-induced myasthenia gravis and myositis in a metastatic renal cell carcinoma patient. Urol Case Rep. 2020;29:101105.
    doi pubmed
  7. Minon-Fernandez B, Losada-Domingo JM, Sanchez-Horvath MT, Barcena-Llona J. [Myasthenia gravis associated with nivolumab]. Rev Neurol. 2020;70(2):72-73.
    doi pubmed
  8. Makarious D, Horwood K, Coward JIG. Myasthenia gravis: An emerging toxicity of immune checkpoint inhibitors. Eur J Cancer. 2017;82:128-136.
    doi pubmed
  9. Mohn N, Beutel G, Gutzmer R, Ivanyi P, Satzger I, Skripuletz T. Neurological immune related adverse events associated with nivolumab, ipilimumab, and pembrolizumab therapy-review of the literature and future outlook. J Clin Med. 2019;8(11):1777.
    doi pubmed
  10. Valenti-Azcarate R, Esparragosa Vazquez I, Toledano Illan C, Idoate Gastearena MA, Gallego Perez-Larraya J. Nivolumab and Ipilimumab-induced myositis and myocarditis mimicking a myasthenia gravis presentation. Neuromuscul Disord. 2020;30(1):67-69.
    doi pubmed
  11. Miller RG, Milner-Brown HS, Mirka A. Prednisone-induced worsening of neuromuscular function in myasthenia gravis. Neurology. 1986;36(5):729-732.
    doi pubmed
  12. Guptill JT, Soni M, Meriggioli MN. Current treatment, emerging translational therapies, and new therapeutic targets for autoimmune myasthenia gravis. Neurotherapeutics. 2016;13(1):118-131.
    doi pubmed
  13. Gajdos P, Chevret S, Clair B, Tranchant C, Chastang C. Clinical trial of plasma exchange and high-dose intravenous immunoglobulin in myasthenia gravis. Myasthenia Gravis Clinical Study Group. Ann Neurol. 1997;41(6):789-796.
    doi pubmed
  14. Barth D, Nabavi Nouri M, Ng E, Nwe P, Bril V. Comparison of IVIg and PLEX in patients with myasthenia gravis. Neurology. 2011;76(23):2017-2023.
    doi pubmed
  15. Zinman L, Ng E, Bril V. IV immunoglobulin in patients with myasthenia gravis: a randomized controlled trial. Neurology. 2007;68(11):837-841.
    doi pubmed
  16. Hoffmann S, Kohler S, Ziegler A, Meisel A. Glucocorticoids in myasthenia gravis - if, when, how, and how much? Acta Neurol Scand. 2014;130(4):211-221.
    doi pubmed
  17. Melzer N, Ruck T, Fuhr P, Gold R, Hohlfeld R, Marx A, Melms A, et al. Clinical features, pathogenesis, and treatment of myasthenia gravis: a supplement to the Guidelines of the German Neurological Society. J Neurol. 2016;263(8):1473-1494.
    doi pubmed
  18. Werner JM, Schweinsberg V, Schroeter M, von Reutern B, Malter MP, Schlaak M, Fink GR, et al. Successful treatment of myasthenia gravis following PD-1/CTLA-4 combination checkpoint blockade in a patient with metastatic melanoma. Front Oncol. 2019;9:84.
    doi pubmed
  19. Bolz S, Ramakrishnan T, Fleischer M, Livingstone E, Stolte B, Thimm A, Kizina K, et al. Detect it so you can treat it: A case series and proposed checklist to detect neurotoxicity in checkpoint therapy. eNeurologicalSci. 2021;22:100324.
    doi pubmed


This article is distributed under the terms of the Creative Commons Attribution Non-Commercial 4.0 International License, which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.


Journal of Medical Cases is published by Elmer Press Inc.

 

Browse  Journals  

 

Journal of Clinical Medicine Research

Journal of Endocrinology and Metabolism

Journal of Clinical Gynecology and Obstetrics

 

World Journal of Oncology

Gastroenterology Research

Journal of Hematology

 

Journal of Medical Cases

Journal of Current Surgery

Clinical Infection and Immunity

 

Cardiology Research

World Journal of Nephrology and Urology

Cellular and Molecular Medicine Research

 

Journal of Neurology Research

International Journal of Clinical Pediatrics

 

 
       
 

Journal of Medical Cases, monthly, ISSN 1923-4155 (print), 1923-4163 (online), published by Elmer Press Inc.                     
The content of this site is intended for health care professionals.
This is an open-access journal distributed under the terms of the Creative Commons Attribution-NonCommercial 4.0 International License, which permits unrestricted
non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.
Creative Commons Attribution license (Attribution-NonCommercial 4.0 International CC-BY-NC 4.0)


This journal follows the International Committee of Medical Journal Editors (ICMJE) recommendations for manuscripts submitted to biomedical journals,
the Committee on Publication Ethics (COPE) guidelines, and the Principles of Transparency and Best Practice in Scholarly Publishing.

website: www.journalmc.org   editorial contact: editor@journalmc.org
Address: 9225 Leslie Street, Suite 201, Richmond Hill, Ontario, L4B 3H6, Canada

© Elmer Press Inc. All Rights Reserved.


Disclaimer: The views and opinions expressed in the published articles are those of the authors and do not necessarily reflect the views or opinions of the editors and Elmer Press Inc. This website is provided for medical research and informational purposes only and does not constitute any medical advice or professional services. The information provided in this journal should not be used for diagnosis and treatment, those seeking medical advice should always consult with a licensed physician.