Abstract
Immune checkpoint inhibitors can cause immune side effects, with myasthenia gravis (MG) being relatively rare. With this review, we present 66-year-old man with melanoma treated with pembrolizumab who developed MG. With immuno-oncology (IO) single agent usage, 42 cases reported new-onset MG and 9 cases reported exacerbation of pre-existing MG. Among the patients who had new-onset MG after administration of programmed cell death protein 1 (PD-1) inhibitors, 14 patients (38.8%) developed severe respiratory failure and required intubation and 10 patients (27.02%) died. Among the patients with exacerbation of pre-existing MG after receiving PD-1 inhibitors, 1 patient (11.1%) required intubation, and no death was reported. Combination IO therapy-induced MG was reported in seven cases, with at least two cases complicated by respiratory failure and one death. Our observations suggest a possible difference in the severity of the disease and outcome among different IO therapy options.
Keywords: oncology, neuromuscular disease
Background
For the last decade, advancements in immuno-oncology revolutionised cancer patient’s management.1 Currently, inhibiting cytotoxic lymphocyte-associated protein 4 (CTLA-4) and programmed cell death protein 1 (PD-1)/programmed death-ligand 1 (PD-L1) axis is implicated for treatment of many malignancies.2 This leads to disinhibition of cytotoxic T cells, resulting in antitumour activity.2 As a result, unique immune-related side effects affecting any organ or tissue may arise.3 Interestingly, these complications may manifest as known autoimmune syndrome. Little is known about how immune check point inhibitors induced side effects are different from sporadic alloimmunisation.4 Neurological immune side effects, though are rare, may lead to rapid and severe deterioration of the patient condition.5 Rarely, some patient may experience myasthenia gravis (MG) type syndrome, which can lead to significant morbidity and mortality if not recognised promptly.6 In such cases, to provide best diagnostic and therapeutic interventions, multidisciplinary approach, involving neurologist, medical oncologist and other medical specialties, is required.7 Here, we report a case of a 66-year-old man with metastatic melanoma who developed MG after receiving the second dose of pembrolizumab. We also performed an extensive review of already existing reports in relation to this topic.
Case presentation
A 66-year-old man was diagnosed with stage IB cutaneous malignant melanoma in 2017. Primary tumour was 1.5-millimetre thick without ulceration, located over left periaricular site. His oncological history is also significant for locally advanced left tonsillar squamous cell carcinoma, which was treated with radiation therapy and cetuximab. Unfortunately, patient was diagnosed with metastatic recurrent melanoma in July 2018. He was found to have bilateral axillary lymphadenopathy and treatment with pembrolizumab was initiated. The patient presented to the clinic on day 10 of cycle 2 of pembrolizumab therapy complaining of diplopia, hoarseness, dysphagia and dysphonia. Laboratory analysis revealed an elevation in both aspartate transaminase and alanine transaminase, consistent with grade two hepatic toxicity. The patient was found to have progressive ptosis during the day, bilateral vocal paralysis and nasal regurgitation. Constellation of these symptoms raised concern for MG-type syndrome. Anti-titin, anti-acetylcholine receptor and Purkinje cell/neuronal nuclear antibodies were not present in the serum. MRI of the brain showed no evidence of metastatic disease.
Differential diagnosis
Although the clinical picture is similar to MG, underlying disease pathology may not be the same and likely not completely understood for immune toxicity. The case we reported could be a case of seronegative MG. Around 10%–20% of patients with MG do not have acetylcholine receptor (AChR) antibodies (seronegative). Among this group of patients, some might have antibodies to a membrane-linked muscle-specific kinase (MuSK).
Treatment
Pembrolizumab was discontinued and the patient was started on pulse steroid therapy with intravenous methylprednisolone 60 mg three times per day for both grade two hepatic toxicity and MG. The patient’s severe dysphagia and muscular weakness necessitated percutaneous endoscopic gastrostomy tube insertion and tracheostomy placement due to the high risk of aspiration and vocal cord paralysis.
Outcome and follow-up
Later, hoarseness and double vision did resolve with these interventions. Unfortunately, vocal cord paralysis was persistent. A positron emission tomography scan in 3 months showed a decrease in the size and the intensity of activity in both axillae. The patient had no progression of metastatic melanoma thereafter. Unique findings in our case is with just two doses of pembrolizumab and since then even without any further line of therapy, the relative stability of metastatic adenopathy and survival reaching >30 months.
Discussion
We performed a detailed and extensive search of medical databases, including Google Scholar and PubMed, looking for all published cases of MG (new cases or exacerbation of pre-existing disease) initiation of immune checkpoint inhibitors (ICI) treatment. We used the following medical subject heading terms: MG, ICI, anti-PD-1 antibody, anti-PD-L1 antibody and anti-CTLA-4 antibody. We searched the reference lists found in relevant articles, including previously published literature reviews and textbooks, manually. As a result of this extensive search and screening of articles, besides our own reported case, we were able to identify 58 reported cases of MG (new cases or exacerbation of pre-existing disease) during or after ICI treatment. Of these, 36 cases reported new-onset MG (table 1) and 9 cases reported exacerbation of pre-existing MG after initiation (table 2) of PD-1 inhibitors. Administration of CTLA-4 inhibitors and PD-L1 inhibitors caused the new onset of MG in four cases (table 3) and two cases (table 4), respectively. In addition, seven cases reported that they developed MG after administration of a combination of ICI medications (table 5).
Table 1.
New onset MG after utilisation of PD-1 inhibitors
| Study | Publish year | Age | Gender | PD-1 inhibitor | Cancer type and stage | Anti-AChR Ab | Anti-MuSK/anti-striational Abs | Previous history of MG | Time between initiation of treatment and onset of symptoms (days) | Initial symptoms | Severe respiratory failure requiring intubation | Initial treatment | MG-related outcome | 
| Our case | 2020 | 66 | M | Pembrolizumab | Metastatic melanoma | Negative | Negative | No | 24 | Diplopia, hoarseness and dysphagia | No | Pulse steroid therapy, percutaneous endoscopic gastrostomy (PEG) and tracheostomy placement | Improved | 
| Zimmer et al8 | 2016 | 69 | F | Pembrolizumab | Metastatic melanoma | Negative | Negative | No | 28 | Eye movement disorder, bilateral diplopia and shortness of breath  | 
No | Pyridostigmine, prednisone, methylprednisolone, intravenous immune globulin (IVIG) and plasmapheresis | Died | 
| Alnahhas and Wong9 | 2017 | 84 | M | Pembrolizumab | Metastatic melanoma | Positive | Negative | No | 30 | Progressive weakness and dysphagia | Yes (refused) | Pyridostigmine, prednosine and IVIG | Died (unclear if it was related to MG) | 
| Makarious et al (ocular MG)10  | 
2017 | 85 | F | Pembrolizumab | Metastatic melanoma | Negative | Negative | No | 31 | Diplopia and asymmetrical bilateral ptosis (L>R)  | 
No | IVIG, prednisone and pyridostigmine | Improved (died later from unrelated cardiac issues) | 
| Nguyen et al11 | 2017 | 81 | M | Pembrolizumab | Metastatic melanoma | Negative | Negative | No | 58 | Bilateral ptosis | No | Prednisone | Improved | 
| Nguyen et al11 | 2017 | 86 | F | Pembrolizumab | Metastatic melanoma | Negative | Negative | No | 41 | Bilateral ptosis and dysphagia  | 
No | Methylprednisolone and oral prednisolone | Improved | 
| Gonzalez et al12 | 2017 | 71 | F | Pembrolizumab | Metastatic uterine carcinoma (CA) | Negative | Negative | No | 63 | Dysphagia, diplopia and dysartheria | No | Pyridostigmine and prednisone | Improved | 
| March et al13 | 2018 | 63 | M | Pembrolizumab | Metastatic melanoma | Positive | Negative | No | 14 | Right ptosis, puffiness, blurred vision and shortness of breath | Yes | Pyridostigmine, prednosine, IVIG and plasma exchange | Died | 
| Algaeed et al14 | 2018 | 73 | M | Pembrolizumab | Recurrent melanoma | Positive | Not reported | No | 21 | Left eyelid droop and shortness of breath | Yes | Pyridostigmine | Improved | 
| Hibino et al15  | 
2018 | 83 | M | Pembrolizumab | Lung squamous cell carcinoma (SCC) stage IVb | Negative | Negative | No | 38 | Narrowing visual field and easy fatigability eye leads | No | Pyridostigmine and prednisone | Improved | 
| Huh et al16 | 2018 | 34 | F | Pembrolizumab | Thymic SCC | Positive | Unknown | No | unknown | Diplopia, dysphagia and dyspnoea | No | IVIG, methylprednisolone, prednisone and plasmapheresis | Improved | 
| Erkilinc et al17 | 2018 | 57 | M | Pembrolizumab | Thymoma | Negative | Negative | No | 30 | Double vision, drooping eyelids and difficulty with talking and swallowing | IVIG, methylprednisolone and pyridostigmine | Improved | |
| Onda et al (ocular MG)18  | 
2019 | 73 | M | Pembrolizumab | Lung adenocarcinoma | Negative | Positive (anti-titin Ab positive)  | 
No | 23 | Diplopia, ptosis and external ophthalmoplegia without general muscle weakness | No | Ascending-dose regimen of prednisolone and steroid pulse therapy | Improved | 
| Noda et al19 | 2019 | 77 | F | Pembrolizumab | Lung adenocarcinoma | Positive | Positive (anti-titin Ab positive)  | 
No | 49 | Limb and neck weakness, dyspnoea and dysphasia | Yes (non-invasive positive-pressure ventilation and tracheotomy positive pressure ventilation) | IVIG, immune absorption therapy and plasma exchange therapy | Improved | 
| Todo et al20 | 2020 | 63 | M | Pembrolizumab | Bladder carcinoma | Negative | Negative | No | 12–34 | Left ptosis and diplopia | No | Prednisolone | Improved | 
| Szuchan et al21 | 2020 | 70 | F | Pembrolizumab | Metastatic thymic cancer | Yes | Unknown | No | 21 | Shortness of breath | Yes | High-dose intravenous methylprednisolone | Improved | 
| Lopez et al22 | 2015 | 65 | M | Nivolumab | Renal cell carcinoma (RCC) | Positive | Negative | No | 15 | Dyspnoea, diplopia and bilateral ptosis | Yes | High dose glucocorticoids (GC)+IVIG | Died | 
| Polat and Donofrio23 | 2016 | 65 | M | Nivolomab | Non-small cell lung cancer (NSCLC) | Negative | Negative | No | 31 | Blurred vision and bilateral ptosis | No | Pyridostigmine | Improved | 
| Sciacca et al24 | 2016 | 81 | M | Nivolumab | NSCLC stage IV  | 
Negative | Unknown | No | 28 | Bilateral ptosis, nasal speech and proximal limb weakness | No | Prednisone | Improved | 
| Shirai et al25 | 2016 | 81 | F | Nivolumab | Melanoma | Positive | Negative | No | 21 | Dyspnoea | Yes (refused) | Treatment refused by the patient | Died | 
| Kimura et al26 | 2016 | 80 | M | Nivolumab | Melanoma | Positive | Negative | No | 14 | Fatigue, anorexia, stable dyspnoea | Yes | High dose GC, IVIG and plasma exchange | Stable disease | 
| Chang et al27 | 2017 | 75 | M | Nivolumab | Bladder SCC | Positive | Negative | No | 39 | Double vision, fatigue and severe shortness of breath (SOB) | No Bilevel positive airway pressure (BiPAP)  | 
Pyridostigmine and IVIG | Died(Hospice) | 
| Chen et al28 | 2017 | 65 | M | Nivolumab | Lung SCC | Negative | Unknown | No | 33 | Weakness of four extremities | Yes (family refused) | Pyridostigmine+high dose GC | Died | 
| Mehta et al29 | 2017 | 73 | M | Nivolumab | Metastatic RCC | Positive | Unknown | No | 14 | Fatigue, haematuria and progressive weakness | Yes | Pyridostigmine+high dose GC | Long-term ventilator support | 
| Tanaka et al30 | 2017 | 70 s | F | Nivolumab | Malignant melanoma | Positive | Unknown | No | 31 | Ptosis | No | Prednisone and pyridostigmine | Improved | 
| Fukasawa et al31 | 2017 | 69 | F | Nivolumab | Advanced lung adenocarcinoma | Positive | Unknown | No | 1 week after the third cycle | Generalised weakness and double vision | No | Methylprednisolone and prednisone | Improved | 
| Konoeda et al32 | 2017 | 74 | F | Nivolumab | Advanced colon cancer | Positive | Unknown | No | 5 days after the second course | Bilateral ptosis | No (required BiPAP) | Oral prednisolone, IVIG and plasma exchange | Improved | 
| Tan et al33 | 2017 | 45 | M | Nivolumab | NSCLC | Positive | Unknown | No | 14 | Dyspnoea, ptosis and ophthalmoplegia | Yes | Methylprednisolone, pyridostigmine and IVIG | Improved | 
| Hasegawa et al34 | 2017 | 76 | F | Nivolumab | NSCLC | Positive | Unknown | No | After the second cycle of nivolumab | Left eyelid ptosis, dyspnoea and muscle weakness | No | Plasma exchange, IVIG and low-dose prednisolone | Improved | 
| Kang et al35 | 2018 | 75 | N | Nivolumab | H&N SCC | Positive | Positive | No | 21 | Severe fatigue, generalised weakness and bilateral ptosis | Yes | Methylprednisolone, oral prednisolone and plasma exchange | Died | 
| Yousef et al36 | 67 | M | Nivolumab | SCC left parotid gland stage IV | Negative | Positive | No | 35 | Difficulty rising from chair and climbing stairs | No (BiPAP) | IVIG+prednisone | Improved | |
| Sawai et al37 | 2019 | 84 | F | Nivolumab | RCC | Negative | Unknown | No | 15 | Blepharoptosis, diplopia, weakness of extremities and dysphagia | Yes (refused by patient and family) | IVIG and steroid pulse therapy | Died | 
| Isami et al38 | 2019 | 53 | M | Nivolumab | NSCLC | Negative | Positive | No | 30 | Bilateral ptosis and bulbar palsy | No | Prednisolone | Improved | 
| Sun et al39 | 2019 | 83 | M | Nivolumab | Recurrent urothelial CA | Positive | Positive | No | 21 | Hoarseness, difficulty speaking and swallowing, and generalised weakness | Yes | Methylprednisolone+plasma exchange | Home hospice with tracheostomy | 
| Kim et al40  | 
2019 | 76 | M | Nivolumab | NSCLC | Positive | Unknown | No | 45 | Gait disturbance | No | Methylprednisolone, prednisone and pyridostigmine | Improved | 
| Nakanishi et al41 | 2020 | 78 | M | Nivolumab | RCC | Positive | Unknown | No | After the second course of nivolumab | Neck pain and altered mental status | Yes (refused by family) | Prednisolone | Died | 
| Pouchelon et al42 | 2020 | 78 | M | Nivolumab | Bronchial squamous cell carcinoma | Negative | Negative | No | Unknown | Difficulty holding his head and ptosis of the right eye | Yes (intubation was not done) | Prednisone, IVIG and plasma exchange | Died | 
Ab, antibody; AChR, acetylcholine receptor; MG, myasthenia gravis; MuSK, muscle-specific kinase; PD-1, programmed cell death protein 1; PEG, percutaneous endoscopic gastrostomy.
Table 2.
Exacerbation of pre-existing MG after initiation of PD-1 inhibitors
| Study | Publish year | Age | Gender | PD-1 inhibitor | Cancer type and stage | Anti-AChR Ab | Anti-MuSK/anti-striated muscle Ab | Previous history of MG | Time between initiation of treatment and onset of symptoms (days) | Initial symptoms | Severe respiratory failure requiring intubation | Initial treatment | MG-related outcome | 
| Zhu and Li43 | 2016 | 59 | F | Pembrolizumab | Metastatic melanoma | Negative | Negative | Yes (generalised MG) | 42 | Rapidly progressive hoarseness and dysphagia | No | IVIG, plasmapheresis and prednisone | Improved | 
| Phadke et al44 | 2016 | 75 | M | Pembrolizumab | Metastatic melanoma | Positive | Unknown | Yes | 21 | Diplopia, bilateral ptosis and respiratory distress | No (BiPAP) | Pyridostigmine, plasma exchange, IVIG and rituximab | Improved | 
| Lau et al45 | 2016 | 75 | M | Pembrolizumab | Metastatic melanoma | Positive | Unknown | Yes | 25 | Difficulty walking and neck weakness | No bilevel positive airway pressure (BiPAP) | Methylprednisolone, prednisone, IVIG and azathioprine | Improved | 
| Earl et al46 | 2018 | 74 | M | Pembrolizumab | Metastatic melanoma | Positive | Negative | Yes | 12 days after his second infusion | Ptosis, diplopia, dysphagia and proximal limb weakness | No | Pyridostigmine, prednisone and plasma exchange mycophenolate | Discharged to inpatient hospice and ultimately died | 
| Maeda et al47 | 2016 | 79 | M | Nivolumab | Metastatic melanoma | Positive | Unknown | Yes | 106 | Diplopia and facial weakness | No | Pyridostigmine+prednisone | Improved | 
| Cooper et al48 | 2017 | 68 | F | Nivolumab | Non-small cell lung cancer (NSCLC) stage IV | Positive | Negative | Yes | 84 days | Difficulty walking, dysartheria and diplopia | No (refused by patient) | Pyridostigmine, prednisone and plasma exchange | Admitted to hospice | 
| Mitsune et al49 | 2018 | 62 | F | Nivolumab | Primary neuroendocrine carcinoma (CA) of trachea | Positive | Positive | Yes (ocular MG)  | 
25 days  | 
Generalised fatigue and muscle weakness | No | Methylprednisolone | Improved | 
| Agrawal Yash et al50 | 2019 | 51 | M | Nivolumab | Melanoma stage IIIB | Negative | Negative | Yes and history of Interferon alfa (IFNa) induced MG | 6 days | Constipation, worsening right arm and hand numbness and paraesthesia | No | Pyridostigmine+high-dose glucocorticoid | Improved | 
| Kamien et al51 | 2019 | 76 | M | Nivolumab | Oesophageal CA | Positive (orignal GM positive but significantly uptrended during exacerbation | Unknown | Yes (generalised MG)  | 
7 days after the second dose. 79 days after the first dose (72 days gap between the first and the second doses | Diplopia, weakness of upper extremities and drolling | No | Pyridostigmine+high-dose GC | Improved | 
Ab, antibody; AChR, acetylcholine receptor; MG, myasthenia gravis; MuSK, muscle-specific kinase; PD-1, programmed cell death protein 1.
Table 3.
Cases of CTLA-4-induced MG
| Study | Publish year | Age | Gender | Cancer type and stage | Anti-AChR Ab | Anti-MuSK/anti-striated muscle Ab | Previous history of MG | Time between initiation of treatment (Rx) and onset of symptoms | Initial symptoms | Severe respiratory failure requiring intubation | Initial treatment | MG-related outcome | 
| Liao et al63 | 2014 | 70 | F | Metastatic melanoma | Positive | Positive | No | 70 days  | 
Generalised myalgia and dysphagia | No | Solu-medrol, plasmapheresis and intravenous immune globulin (IVIG) | Improved | 
| Johnson et al56 | 2015 | 69 | F | Melanoma stage IIIA | Positive | Negative | No | 42 days | Diplopia, dysphagia and ptosis | No | Pyridostigmine | Improved | 
| Johnson et al56 | 2015 | 73 | M | Melanoma stage IIIB | Positive | Unknown | No | After 2 doses (probably 21 days) | Shortness of breath (SOB) and proximal limb weakness | No | Pyridostigmine and high-dose glucocorticoid | Improved | 
| Montes et al64 | 2018 | 74 | M | Metastatic melanoma | Negative | Unknown | No | 43 days | SOB and proximal limb weakness | No | Pyridostigmine and high-dose glucocorticoid | Improved | 
Ab, antibody; AChR, acetylcholine receptor; CTLA-4, cytotoxic lymphocyte-associated protein 4; MG, myasthenia gravis; MuSK, muscle-specific kinase.
Table 4.
Cases of PD-L1-induced MG
| Study | Publish year | Age | Gender | PD-L1 inhibitor | Cancer type and stage | Anti-AChR Ab | Anti-MuSK/anti-striated muscle Ab | Previous history of MG | Time between initiation of treatment (Rx) and onset of symptoms | Initial symptoms | Severe respiratory failure requiring intubation | Initial treatment | MG-related outcome | 
| Brahmer et al53 | 2012 | Unknown | Unknown | Anti-PD-L1 antibody (not specified) | Unknown | Unknown | Unknown | Unknown | Unknown | Unknown | Unknown | Unknown | Unknown | 
| Thakolwiboon et al65 | 2019 | 87 | M | Atezolizumab | Urothelial carcinoma | Positive | Positive | No | 21 days | Double vision and ptosis | No | Intravenous immune globulin (IVIG) and low-dose oral pyridostigmine | Died due to cardiac arrest | 
| Shah et al66 | 2019 | 81 | F | Atezolizumab | Urachal cancer | Negative | Negative | No | 60 days | Head droop, left facial droop and voice change | Yes | Plasma exchange and rituximab | Comfort care | 
Ab, antibody; AChR, acetylcholine recpetor; MG, myasthenia gravis; MuSK, muscle-specific kinase; PD-L1, programmed death-ligand 1.
Table 5.
Cases of MG induced by combination of ICI therapy
| Study | Publish year | Age | Gender | Immune checkpoint inhibitors (ICI) | Cancer type and stage | Anti-AChR Ab | Anti-MuSK/anti-striated muscle Ab | Previous history of MG | Time between initiation of treatment (Rx) and onset of symptoms | Initial symptoms | Severe respiratory failure requiring intubation | Initial treatment | MG-related outcome | 
| Loochtan et al67 | 2015 | 70 | M | Nivolumab+ipilimumab | Small cell lung cancer (SCLC) stage | Positive | Positive | No | 16 days | Ptosis and diplopia | Yes | Prednisone and plasmapheresis | Comfort care | 
| Chen et al52 | 2017 | 57 | M | Nivolumab+ipilimumab | Non-small cell lung cancer (NSCLC) | Positive | Unknown | No | 14 days | Drooping of eyelids, dropped head, limb weakness, unsteadiness in walking and mild dyspnoea | No | Prednisolone and pyridostigmine | Improved from MG but died from sepsis in the same hospitalisation | 
| Werner et al68 | 2019 | 62 | M | Nivolumab+ipilimumab | Melanoma (clark-level III) | Negative | Negative | No | 28 days | Fatigue and ptosis of right eye | No | Pyridostigmine+high-dose glucocorticoids | Improved | 
| Fazel et al54 | 2019 | 78 | F | Nivolumab+ipilimumab | Metastatic melanoma | Unknown | Positive | No | 5 days | Diplopia, proximal muscle weakness and myalgia | Yes (refused by the patient)  | 
Methylprednisolone+ Intravenous immune globulin (IVIG) | Inpatient hospice | 
| Leaver et al69 | 2020 | 55 | M | Nivolumab+ipilimumab | Metastatic melanoma | Negative | Negative | No | 28 days | Blurred vision, mild bilateral ptosis and fatigable left arm abduction | No | Prednisolone | Improved | 
| Antonia et al55 | 2016 | Durvaluma+tremelimumab | Advanced NSCLC | Unknown | Unknown | Unknown | 14 days | Unknown | Unknown | Unknown | Died | ||
| Yousef et al36 | 65 | F | Durvalumab+tremelimumab | Liomyosarcoma stage IV with lung metastasis | Positive | Positive | No | After the first dose | Back pain and stooped posture with difficulty lifting her head | No | Plasma exchange+ Intravenous immune globulin (IVIG) + prednisone | Improved | 
Ab, antibody; AChR, acetylcholine receptor; MG, myasthenia gravis; MuSK, muscle-specific kinase.
Among 36 reported cases of new onset of the MG after administration of PD-1 inhibitors, 15 cases were related to pembrolizumab8–21 and 21 cases happened after using nivolumab.22–42 In the patients with exacerbation of the pre-existing MG, four were related to pembrolizumab43–46 and five were related to nivolumab.47–51 AChR antibody was positive in 20 patients (51.2%) with new onset of MG and 7 patients (77.7%) with exacerbation of the pre-existing MG. We considered developing severe respiratory failure requiring bilevel positive airway pressure (BiPAP) or endotracheal intubation and death as unfavourable outcomes of disease. Among the patients who had new-onset MG after administration of PD-1 inhibitors, 14 patients (38.8%) developed severe respiratory failure due to MG and required intubation from which 11 patients received nivolumab,22 25 26 28 29 33 35 37 39 41 42 and 3 received pembrolizumab.9 13 14 Three patients (8%) who all received nivolumab required BiPAP.27 32 36 Ten patients (27.02%) passed away as a result of MG, among which 8 patients received nivolumab22 25 27 28 35 37 41 42 and 2 patients were on pembrolizumab.8 13 One patient required long-term ventilation support29 and one patient was placed on hospice care.39
Among the patients with exacerbation of pre-existing MG after receiving PD-1 inhibitors, patient (11.1%) who was receiving nivolumab required intubation,48 and 2 patients (22.1%) required BiPAP.44 45 Both these patients that required BiPAP were receiving pembrolizumab. No death was reported in patients with exacerbation of pre-existing MG, but two of them were placed on hospice care.46 48 Among reported cases of PD-1 inhibitor-associated new-onset MG, the mean duration for the onset of symptoms was 29.06 days post initiation of the PD-1 inhibitor. Among those with exacerbation of the pre-existing MG after receiving PD-1 inhibitor, the mean duration for the onset of symptoms was 43.2 days post initiation of the medication. Among four reported cases of ipilimumab-associated MG, all were new-onset cases. The mean duration for the onset of symptoms was 44 days post initiation of ipilimumab. No death was reported (table 4).
We found seven reported cases of immune checkpoint inhibitor combination therapy-associated MG (table 5). Five patients received nivolumab+ipilimumab and the other two received durvalumab+tremelimumab. Among these patients, the mean duration for the onset of symptoms was 17.5 days post initiation of combination therapy. Among this group of patients, at least 2 patients (28.5%) developed severe respiratory failure requiring endotracheal intubation (28.5%) and 1 (14.2%) died as a result of MG. In one case, the symptoms of MG were improving, but he died from sepsis in the same hospitalisation.52 Two patients were placed on comfort/hospice care.53 54 Antonia et al developed a multicentre study to evaluate for safety and antitumor activity of durvalumab plus tremelimumab in non-small cell lung cancer. In this study, they reported a patient with NSCLC who received durvalumab 20 mg/kg every 4 weeks plus tremelimumab 1 mg/kg. This patient developed MG within 10 days of initiating treatment and eventually passed away as a result of MG. Although considering the outcome of death, we can guess this patient developed severe respiratory failure requiring endotracheal intubation but unfortunately despite detailed evaluation of the manuscript, we could not find more information about this patient, including the course of treatment or the need for intubation.55
Over the last couple of years, immune checkpoint inhibitors utilisation has emerged as an effective treatment for different types of advanced cancers.56 57 Despite impressive benefits were observed from using ICI agents in patients with different types of cancer, utilisation of these medications was associated with the occurrence of serious adverse events related to excessive immune system activation, known as immune-related adverse events (irAEs).58 MG is an autoimmune disorder that targets neuromuscular junction where acetylcholine is the main neurotransmitter.59 It is believed that autoantibodies against the AChR or muscle-specific tyrosine kinase are responsible for the clinical features.60 61 The main characteristic of the disease is fatigable weakness variably involving ocular, bulbar, respiratory and limb muscles.62 MG is estimated to occur in 0.1%–0.2% of patients receiving immunotherapy.56 63
In June 2012, Brahmer et al reported the first case of new onset of MG in a patient treated with an ICI medication. They conducted the study of ‘Safety and activity of anti-PD-L1 antibody in patients with advanced cancer’. In which they have enrolled patients with different types of advanced cancer in a dose-escalation phase of the trial of anti-PD-L1 antibody administration. Based on the reported results, among 125 patients who received 10 mg/kg of anti-PD-L1 antibody (the highest dose), 1 patient developed MG.53 Since then, multiple cases reported indicating the development of MG after using ICI agents. The number of reported cases who developed new-onset MG or exacerbation of pre-existing disease after initiation of ICI treatment over the last couple of years has been continuously increasing. We found 7 cases reported in 2018. This number increased to 13 cases in 2019. Regarding the reported cases in 2020, as of 31 May, we found 6 reported cases, including our case. This increasing pattern indicates the importance of gathering more information about this side effect of ICI therapy for different cancers. Our observations show that the prevalence of developing severe respiratory failure due to MG, which required intubation of patients, was higher in cases with new onset of MG after using PD-1 inhibitors compared to those with pre-existing MG who had exacerbation of the disease after using PD-1 inhibitors (38.8% vs 11.1%). We had a similar observation for the reported death outcome (27.02% vs no death). In addition, the mean duration for the onset of symptoms after initiation of PD-1 inhibitor was much shorter in patients with new-onset MG compared to exacerbation of the pre-existing disease (29.06 days vs 43.2 days). These data could probably suggest a more severe presentation of the disease and a worse outcome in the new onset of MG after using PD-1 inhibitors. Although more studies are needed to confirm this observation, we can suggest more careful monitoring of the patients without previous history of MG for the symptoms of this disease while being treated with PD-1 inhibitors.
None of the four patients who received ipilimumab developed the severe disease and they all improved without any unfavourable outcome. Also, the mean duration for the onset of symptoms after initiation of ipilimumab was 44 days, which is the longest duration among all ICI agents. This might be suggestive of milder presentation of ipilimumab-induced MG.
In contrast, 42.8% of patients who received the combination of ICI medications developed severe respiratory failure requiring endotracheal intubation, which is the highest rate among the patients who developed MG after administration of ICI agents. In addition, the mean duration for the onset of symptoms of MG was 17.5 days post initiation of combination therapy, which is the shortest duration among all ICI agents. We can suggest that the patient who develops MG after combination therapy with ICI medications probably is at a higher risk to develop a more severe presentation of the disease.
All of these suggestions need to be evaluated more by performing further studies. This will lead to a better understanding of these irAEs of ICI therapy and can help in better management and eventually better outcome of this disease.
Although MG is considered asone of the rare irAEs of ICI therapy, considering an increasing number of reported cases with MG related to ICI therapy, the clinicians need to enhance their understanding of this phenomenon. Our observations suggest possible differences in the severity of the disease and outcome among different ICI therapy options. More studies are needed to evaluate these suggestions.
Learning points.
Considering an increasing number of reported cases with myasthenia gravis (MG) related to immune checkpoint inhibitors (ICI) therapy, clinicians need to enhance their understanding of this phenomenon.
Our observations suggest a possible difference in the severity of MG related to ICI therapy among different ICI therapy regimens.
Footnotes
Contributors: BH and AV worked on the review of literature and prepared initial draft. SM has edited the entire manuscript to meet BMJ requirements, and worked on submission. KC supervised the entire process, edited and approved the final draft.
Funding: The authors have not declared a specific grant for this research from any funding agency in the public, commercial or not-for-profit sectors.
Competing interests: None declared.
Provenance and peer review: Not commissioned; externally peer reviewed.
Ethics statements
Patient consent for publication
Obtained.
References
- 1.Dubois M, Ardin C, André F, et al. L’immunothérapie, une révolution en oncologie - Revue de l’efficacité des inhibiteurs de points de contrôle immunitaire [The revolution of immuno-oncology therapy: review of immune checkpoint inhibitors efficacy]. Med Sci 2019;35:937–45. 10.1051/medsci/2019225 [DOI] [PubMed] [Google Scholar]
 - 2.La-Beck NM, Jean GW, Huynh C. Immune Checkpoint Inhibitors: New Insights and Current Place in Cancer Therapy [published correction appears in Pharmacotherapy. 2015 Dec;35(12):1205]. Pharmacotherapy 2015;35:963–76. 10.1002/phar.1643 [DOI] [PubMed] [Google Scholar]
 - 3.Spiers L, Coupe N, Payne M. Toxicities associated with checkpoint inhibitors-an overview. Rheumatology 2019;58:vii7–16. 10.1093/rheumatology/kez418 [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 4.Tocut M, Brenner R, Zandman-Goddard G. Autoimmune phenomena and disease in cancer patients treated with immune checkpoint inhibitors. Autoimmun Rev 2018;17:610–6. 10.1016/j.autrev.2018.01.010 [DOI] [PubMed] [Google Scholar]
 - 5.Dalakas MC. Neurological complications of immune checkpoint inhibitors: what happens when you 'take the brakes off' the immune system. Ther Adv Neurol Disord 2018;11:1756286418799864. 10.1177/1756286418799864 [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 6.Becquart O, Lacotte J, Malissart P, et al. Myasthenia gravis induced by immune checkpoint inhibitors. J Immunother 2019;42:309–12. 10.1097/CJI.0000000000000278 [DOI] [PubMed] [Google Scholar]
 - 7.Kottschade L, Brys A, Peikert T, et al. A multidisciplinary approach to toxicity management of modern immune checkpoint inhibitors in cancer therapy. Melanoma Res 2016;26:469–80. 10.1097/CMR.0000000000000273 [DOI] [PubMed] [Google Scholar]
 - 8.Zimmer L, Goldinger SM, Hofmann L, et al. Neurological, respiratory, musculoskeletal, cardiac and ocular side-effects of anti-PD-1 therapy. Eur J Cancer 2016;60:210–25. 10.1016/j.ejca.2016.02.024 [DOI] [PubMed] [Google Scholar]
 - 9.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:E25–6. 10.1002/mus.25496 [DOI] [PubMed] [Google Scholar]
 - 10.Makarious D, Horwood K, Coward JIG. Myasthenia gravis: an emerging toxicity of immune checkpoint inhibitors. Eur J Cancer 2017;82:128–36. 10.1016/j.ejca.2017.05.041 [DOI] [PubMed] [Google Scholar]
 - 11.Nguyen BHV, Kuo J, Budiman A, et al. Two cases of clinical myasthenia gravis associated with pembrolizumab use in responding melanoma patients. Melanoma Res 2017;27:152–4. 10.1097/CMR.0000000000000310 [DOI] [PubMed] [Google Scholar]
 - 12.Gonzalez NL, Puwanant A, Lu A, et al. Myasthenia triggered by immune checkpoint inhibitors: new case and literature review. Neuromuscul Disord 2017;27:266–8. 10.1016/j.nmd.2017.01.002 [DOI] [PubMed] [Google Scholar]
 - 13.March KL, Samarin MJ, Sodhi A, et al. Pembrolizumab-induced myasthenia gravis: a fatal case report. J Oncol Pharm Pract 2018;24:146–9. 10.1177/1078155216687389 [DOI] [PubMed] [Google Scholar]
 - 14.Algaeed M, Mukharesh L, Heinzelmann M, et al. Pearls & Oy-sters: Pembrolizumab-induced myasthenia gravis. Neurology 2018;91:e1365–7. 10.1212/WNL.0000000000006278 [DOI] [PubMed] [Google Scholar]
 - 15.Hibino M, Maeda K, Horiuchi S, et al. Pembrolizumab-induced myasthenia gravis with myositis in a patient with lung cancer. Respirol Case Rep 2018;6:e00355. 10.1002/rcr2.355 [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 16.Huh SY, Shin SH, Kim MK, et al. Emergence of myasthenia gravis with myositis in a patient treated with pembrolizumab for thymic cancer. J Clin Neurol 2018;14:115–7. 10.3988/jcn.2018.14.1.115 [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 17.Erkilinc B, Karagozlu E, Uludag B. S52. A case of myasthenia gravis related to use of pembrolizumab. Clinical Neurophysiology. Volume 129, Supplement 1, May 2018, Page e161 (Abstracts of the 31st International Congress of Clinical Neurophysiology (ICCN) of the IFCN, May 1–6, 2018, Washington, DC, USA).
 - 18.Onda A, Miyagawa S, Takahashi N, et al. Pembrolizumab-induced ocular myasthenia gravis with anti-titin antibody and necrotizing myopathy. Intern Med 2019;58:1635–8. 10.2169/internalmedicine.1956-18 [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 19.Noda T, Kageyama H, Miura M, et al. [A case of myasthenia gravis and myositis induced by pembrolizumab]. Rinsho Shinkeigaku 2019;59:502–8. 10.5692/clinicalneurol.cn-001251 [DOI] [PubMed] [Google Scholar]
 - 20.Todo M, Kaneko G, Shirotake S, et al. Pembrolizumab-induced myasthenia gravis with myositis and Presumable myocarditis in a patient with bladder cancer. IJU Case Rep 2020;3:17–20. 10.1002/iju5.12128 [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 21.Szuchan C, Elson L, Alley E, et al. Checkpoint inhibitor-induced myocarditis and myasthenia gravis in a recurrent/metastatic thymic carcinoma patient: a case report. Eur Heart J Case Rep 2020;4:1–8. 10.1093/ehjcr/ytaa051 [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 22.Lopez D, Calvao A, Fershko A. 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;2:11–16. [Google Scholar]
 - 23.Polat P, Donofrio PD. Myasthenia gravis induced by nivolumab therapy in a patient with non-small-cell lung cancer. Muscle Nerve 2016;54:507. 10.1002/mus.25163 [DOI] [PubMed] [Google Scholar]
 - 24.Sciacca G, Nicoletti A, Rampello L, et al. Benign form of myasthenia gravis after nivolumab treatment. Muscle Nerve 2016;54:507–9. 10.1002/mus.25212 [DOI] [PubMed] [Google Scholar]
 - 25.Shirai T, Sano T, Kamijo F, 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. 10.1093/jjco/hyv158 [DOI] [PubMed] [Google Scholar]
 - 26.Kimura T, Fukushima S, Miyashita A, et al. Myasthenic crisis and polymyositis induced by one dose of nivolumab. Cancer Sci 2016;107:1055–8. 10.1111/cas.12961 [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 27.Chang E, Sabichi AL, Sada YH. Myasthenia gravis after nivolumab therapy for squamous cell carcinoma of the bladder. J Immunother 2017;40:114–6. 10.1097/CJI.0000000000000161 [DOI] [PubMed] [Google Scholar]
 - 28.Chen Y-H, Liu F-C, Hsu C-H, et al. Nivolumab-induced myasthenia gravis in a patient with squamous cell lung carcinoma: case report. Medicine 2017;96:e7350. 10.1097/MD.0000000000007350 [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 29.Mehta JJ, Maloney E, Srinivasan S, et al. Myasthenia gravis induced by nivolumab: a case report. Cureus 2017;9:e1702. 10.7759/cureus.1702 [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 30.Tanaka R, Sunada Y, Wataru F. Successful reinstitution of nivolumab in combination with corticosteroids for metastatic malignant melanoma with myasthenia gravis as an immune-related adverse event. Kawasaki Medical Journal 2017;43:59–61. [Google Scholar]
 - 31.Fukasawa Y, Sasaki K, Natsume M, et al. Nivolumab-Induced myocarditis concomitant with myasthenia gravis. Case Rep Oncol 2017;10:809–12. 10.1159/000479958 [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 32.Konoeda F, Suzuki S, Nishimoto Y, et al. A case of myasthenia gravis and myositis induced by nivolumab. Rinsho Shinkeigaku 2017;57:373–7. 10.5692/clinicalneurol.cn-000991 [DOI] [PubMed] [Google Scholar]
 - 33.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. 10.1016/j.jtho.2017.02.024 [DOI] [PubMed] [Google Scholar]
 - 34.Hasegawa Y, Kawai S, Ota T, et al. Myasthenia gravis induced by nivolumab in patients with non-small-cell lung cancer: a case report and literature review. Immunotherapy 2017;9:701–7. 10.2217/imt-2017-0043 [DOI] [PubMed] [Google Scholar]
 - 35.Kang KH, Grubb W, Sawlani K, et al. Immune checkpoint-mediated myositis and myasthenia gravis: a case report and review of evaluation and management. Am J Otolaryngol 2018;39:642–5. 10.1016/j.amjoto.2018.06.003 [DOI] [PubMed] [Google Scholar]
 - 36.Yousef ML, Harrison RA, Woodman K, et al. Neurologic complications in cancer patients exposed to checkpoint inhibitors. poster presentation of case series from MD Anderson cancer center.
 - 37.Sawai T, Hosokawa T, Shigekiyo T, et al. [An autopsy case of nivolumab-induced myasthenia gravis and myositis]. Rinsho Shinkeigaku 2019;59:360–4. 10.5692/clinicalneurol.cn-001282 [DOI] [PubMed] [Google Scholar]
 - 38.Isami A, Uchiyama A, Shimaoka Y, et al. [A case of anti-titin antibody positive nivolumab-related necrotizing myopathy with myasthenia gravis]. Rinsho Shinkeigaku 2019;59:431–5. 10.5692/clinicalneurol.cn-001270 [DOI] [PubMed] [Google Scholar]
 - 39.Sun R, Shah V, Tummala S, et al. Nivolumab-induced myasthenia gravis with myositis in patients with genitourinary cancer (P4.2-005). Neurology 2019;92. [Google Scholar]
 - 40.Kim J-S, Nam T-S, Kim J, et al. Myasthenia gravis and myopathy after nivolumab treatment for non-small cell lung carcinoma: a case report. Thorac Cancer 2019;10:2045–9. 10.1111/1759-7714.13177 [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 41.Nakanishi S, Nishida S, Miyazato M, et al. A case report of nivolumab-induced myasthenia gravis and myositis in a metastatic renal cell carcinoma patient. Urol Case Rep 2020;29:101105. 10.1016/j.eucr.2019.101105 [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 42.Pouchelon CA, Afanasiev V, Evrard S. Myasthénie auto-immune induite PAR Le nivolumab, un inhibiteur de checkpoint immunitaireMyasthenia gravis induced by nivolumab, an immune checkpoint inhibitor. Pratique Neurologique – FMC 2020;11:39–43. [Google Scholar]
 - 43.Zhu J, Li Y. Myasthenia gravis exacerbation associated with pembrolizumab. Muscle Nerve 2016;54:506–7. 10.1002/mus.25055 [DOI] [PubMed] [Google Scholar]
 - 44.Phadke SD, Ghabour R, Swick BL, et al. 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. 10.1177/2324709616674316 [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 45.Lau KHV, Kumar A, Yang IH, et al. Exacerbation of myasthenia gravis in a patient with melanoma treated with pembrolizumab. Muscle Nerve 2016;54:157–61. 10.1002/mus.25141 [DOI] [PubMed] [Google Scholar]
 - 46.Earl DE, Loochtan AI, Bedlack RS. Refractory myasthenia gravis exacerbation triggered by pembrolizumab. Muscle Nerve 2018;57:E120–1. 10.1002/mus.26021 [DOI] [PubMed] [Google Scholar]
 - 47.Maeda O, Yokota K, Atsuta N, et al. Nivolumab for the treatment of malignant melanoma in a patient with pre-existing myasthenia gravis. Nagoya J Med Sci 2016;78:119–22. [PMC free article] [PubMed] [Google Scholar]
 - 48.Cooper DS, Meriggioli MN, Bonomi PD, et al. Severe exacerbation of myasthenia gravis associated with checkpoint inhibitor immunotherapy. J Neuromuscul Dis 2017;4:169–73. 10.3233/JND-170219 [DOI] [PubMed] [Google Scholar]
 - 49.Mitsune A, Yanagisawa S, Fukuhara T, et al. Relapsed myasthenia gravis after nivolumab treatment. Intern Med 2018;57:1893–7. 10.2169/internalmedicine.9153-17 [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 50.Agrawal Yash N, James H, Frances C. Case report and review of literature management of myasthenia gravis without significant exacerbation during nivolumab therapy for metastatic melanoma: a case report and review of literature. Annals of Clinical Oncology 2019. [Google Scholar]
 - 51.Kamien A, Knuth A, Santhosh-Kumar C. Reactivation of myasthenia gravis secondary to nivolumab: case report and literature review. J Hematol Oncol Pharm 2019;9:24–9. [Google Scholar]
 - 52.Chen J-H, Lee K-Y, Hu C-J, et al. 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 2017;96:e9262. 10.1097/MD.0000000000009262 [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 53.Brahmer JR, Tykodi SS, Chow LQM, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med 2012;366:2455–65. 10.1056/NEJMoa1200694 [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 54.Fazel M, Jedlowski PM, Myositis S. Severe myositis, myocarditis, and myasthenia gravis with elevated anti-striated muscle antibody following single dose of Ipilimumab-Nivolumab therapy in a patient with metastatic melanoma. Case Reports Immunol 2019;2019:1–3. 10.1155/2019/2539493 [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 55.Antonia SJ, López-Martin JA, Bendell J, et al. Nivolumab alone and nivolumab plus ipilimumab in recurrent small-cell lung cancer (CheckMate 032): a multicentre, open-label, phase 1/2 trial. Lancet Oncol 2016;17:883–95. 10.1016/S1470-2045(16)30098-5 [DOI] [PubMed] [Google Scholar]
 - 56.Johnson DB, Saranga-Perry V, Lavin PJM, et al. Myasthenia gravis induced by ipilimumab in patients with metastatic melanoma. J Clin Oncol 2015;33:e122–4. 10.1200/JCO.2013.51.1683 [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 57.Abdel-Wahab N, Shah M, Suarez-Almazor ME. Adverse events associated with immune checkpoint blockade in patients with cancer: a systematic review of case reports. PLoS One 2016;11:e0160221. 10.1371/journal.pone.0160221 [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 58.Villadolid J, Amin A. Immune checkpoint inhibitors in clinical practice: update on management of immune-related toxicities. Transl Lung Cancer Res 2015;4:560–75. 10.3978/j.issn.2218-6751.2015.06.06 [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 59.Drachman DB. Myasthenia gravis. N Engl J Med 1994;330:1797–810. 10.1056/NEJM199406233302507 [DOI] [PubMed] [Google Scholar]
 - 60.Patrick J, Lindstrom J. Autoimmune response to acetylcholine receptor. Science 1973;180:871–2. 10.1126/science.180.4088.871 [DOI] [PubMed] [Google Scholar]
 - 61.Hoch W, McConville J, Helms S, et al. Auto-Antibodies to the receptor tyrosine kinase MuSK in patients with myasthenia gravis without acetylcholine receptor antibodies. Nat Med 2001;7:365–8. 10.1038/85520 [DOI] [PubMed] [Google Scholar]
 - 62.Grob D. Course and management of myasthenia gravis. J Am Med Assoc 1953;153:529–32. 10.1001/jama.1953.02940230001001 [DOI] [PubMed] [Google Scholar]
 - 63.Liao B, Shroff S, Kamiya-Matsuoka C, et al. Atypical neurological complications of ipilimumab therapy in patients with metastatic melanoma. Neuro Oncol 2014;16:589–93. 10.1093/neuonc/nou001 [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 64.Montes V, Sousa S, Pita F, et al. Myasthenia gravis induced by ipilimumab in a patient with metastatic melanoma. Front Neurol 2018;9:150. 10.3389/fneur.2018.00150 [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 65.Thakolwiboon S, Karukote A, Wilms H. De novo myasthenia gravis induced by Atezolizumab in a patient with urothelial carcinoma. Cureus 2019;11:e5002. 10.7759/cureus.5002 [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 66.Shah M, Tayar JH, Abdel-Wahab N, et al. Myositis as an adverse event of immune checkpoint blockade for cancer therapy. Semin Arthritis Rheum 2019;48:4736–40. 10.1016/j.semarthrit.2018.05.006 [DOI] [PubMed] [Google Scholar]
 - 67.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. 10.1002/mus.24648 [DOI] [PubMed] [Google Scholar]
 - 68.Werner J-M, Schweinsberg V, Schroeter M, 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. 10.3389/fonc.2019.00084 [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 69.Leaver PJ, Jang HS-I, Vernon ST, et al. Immune checkpoint inhibitor-mediated myasthenia gravis with focal subclinical myocarditis progressing to symptomatic cardiac disease. BMJ Case Rep 2020;13. 10.1136/bcr-2019-232920. [Epub ahead of print: 13 May 2020]. [DOI] [PMC free article] [PubMed] [Google Scholar]
 
