Abstract
Immune checkpoint inhibitors (ICIs) have recently been used as a promising treatment for cancer, while their toxicity and immune-related side effects can be seen in any organ, including the nervous system. In contrast to other immune-related adverse events (irAEs), neurological irAEs (nAEs) are rare, with varying incidence and symptom complexity. Although nAEs are uncommon, they can sometimes be severe and even lead to death. However, little attention has been paid to nAEs, and the literature is mostly clinical reports with only a few cases. We, therefore, conducted the present review with the aim of providing a comprehensive introduction of nAEs. In this review, we summarized various nAEs, including meningitis, encephalitis, and hypophysitis in the central nervous system, and myositis, myasthenia gravis, and peripheral neuropathies in the peripheral system. We also reviewed the current diagnosis and treatment methods for nAEs commonly used in clinical practice. In addition, we discussed potential mechanisms regarding nAEs and proposed the possible approaches to prevent the risk of nAEs in patients treated with ICIs. There is still a lot to learn, such as whether and why patients with nAEs respond better to ICI-therapy. The mechanisms and significance of nAEs need to be fully clarified to address these issues and optimize the treatment strategy.
Keywords: Neurotoxicity, immune checkpoint inhibitor, immune-related adverse events, cancer, immunotherapy, neuropathy
1. INTRODUCTION
Immune checkpoint inhibitors (ICIs) can trigger an anti-tumor immune response by blocking cytotoxic inhibitory signaling. Some of them are well recognized to induce tumor eliminating effects such as cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), programmed cell death protein 1 (PD-1) on T cells, and programmed cell death ligand 1 (PD-L1) on tumor cells [1]. The use of ICIs may be beneficial for patient survival, and they have become part of the standard treatment in a number of human malignancies, resulting in a substantial increase in the number of patients receiving ICI-therapy over the last couple of years.
Immune checkpoints are receptors located on the cell surface of T cells. There are two classes of immune checkpoints. Activation receptors, including CD28, CD137, CD27, ICOS, OX40, etc., can enhance the stimulation of T cells, whereas inhibitory ones such as PD-1 and CTLA-4 usually transmit blocking signals to attenuate T-cell activation [2]. For example, PD-1 interacts with its ligands, blocking T-cell activation, cytokine production, cytotoxic function, and impairing the survival of T cells. The corresponding ligand of PD-1, known as PD-L1, is usually abnormally expressed in cancer cells, leading to the escaping of surveillance of the host immune system. ICIs can bind to these inhibitory receptors and prevent them from disturbing the activation of T-cells [2].
Ipilimumab, which targets CTLA-4, was first approved in 2011 by the Food and Drug Administration (FDA) for treating patients with advanced melanoma. Since then, more antibodies against immune checkpoints have been extensively studied. Nivolumab, pembrolizumab (anti-PD-1) and atezolizumab, durvalumab, and avelumab (anti-PD-L1) were approved by FDA subsequently for treating patients with various solid tumors. ICIs approved by FDA for treating human malignancies are listed in Table 1. Currently, the efficacy of different ICIs was evaluated in a lot of clinical trials as monotherapy or combination therapy. The objective response rate (ORR) of pembrolizumab or nivolumab among melanoma or non-small cell lung carcinoma patients was 40-45% [3-5]. Patients with bladder urothelial cancer who received PD-1/PD-L1 inhibitors treatment had an increased ORR (13-24%) [6]. Patients with triple-negative breast cancer had a relatively moderate response to PD-1 inhibitors (19%) [7]. The ORR of relapsed or refractory Hodgkin's lymphoma patients with nivolumab treatment was 87% (17% complete response) [8]. Pembrolizumab and nivolumab for treating patients with various malignancies are under phase IV clinical trials.
Table 1.
Summary of FDA-approved immune checkpoint inhibitors.
| Target |
Drug
(Trade Name) |
Immunoglobulin Class | Cancer type | Company |
First
Approved |
|---|---|---|---|---|---|
| PD-1 | Pembrolizumab (Keytruda) |
IgG4 | Melanoma, MCC, NSCLC, PMBCL, BLCA, MSI-H or dMMR cancers, HNSCC, Cervical cancer, Gastric/GEJ adenocarcinoma, HCC | Merck | 2014 |
| - | Nivolumab (Opdivo) |
IgG4 | Melanoma, NSCLC, SCLC, RCC, Hodgkin’s lymphoma, HNSC, BLCA, MSI-H or dMMR metastatic colorectal cancer | Bristol-Myers Squibb | 2014 |
| - | Cemiplimab (Libtayo) |
IgG4 | CSCC | Sanofi | 2018 |
| PD-L1 | Avelumab (Bavencio) |
IgG1 | MCC, BLCA | Merck Serono, Pfizer | 2017 |
| - | Atezolizumab (Tecentriq) |
IgG1 | BLCA, NSCLC, TNBC | Genentech | 2016 |
| - | Durvalumab (Imfinzi) |
IgG1 | BLCA, NSCLC | AstraZeneca | 2017 |
| CTLA-4 | Ipilimumab (Yervoy) |
IgG1 | Melanoma, RCC, MSI-H or dMMR metastatic colorectal cancer | Bristol-Myers Squibb | 2011 |
Abbreviations: MCC Merkel cell carcinoma, PMBCL primary mediastinal B cell lymphoma, MSI-H microsatellite instability-high, dMMR mismatch-repair deficient, GEJ gastroesophageal junction, SCLC small cell lung cancer, PD-1 programmed cell death protein 1, NSCLC non-small cell lung carcinoma, HCC hepatocellular carcinoma, RCC renal cell carcinoma, BLCA bladder urothelial carcinoma, HNSC head and neck squamous cell carcinoma, CSCC cutaneous squamous cell carcinoma, HNSCC head and neck squamous cell carcinoma, PD-L1 programmed cell death ligand 1, TNBC triple-negative breast cancer, CTLA-4 cytotoxic T-lymphocyte associated protein 4.
Although ICIs play critical roles against tumors, the activation of T cells shows the potential to make host organ systems suffer from autoimmune adverse effects. Dermatological irAEs are the first and most commonly reported in nearly half of patients treated with ipilimumab [9]. The incidence of gastrointestinal irAEs or hepatotoxicity is high, and symptoms are various. However, irAEs of the renal, respiratory, ophthalmological, and nervous systems are relatively rare [9].
With the increase in indications and widespread use of ICI-therapy, concerns have arisen about the occurrence and development of neurotoxic adverse effects. Neurological irAEs (nAEs) are not very common, accounting for no more than 3% of all irAEs. The peripheral nAEs occur more often than nAEs in the central nervous system (CNS) [10, 11]. The most common symptoms are headache, encephalitis, neuropathy, myositis, and myasthenia gravis [10-12]. Since nAEs may present with systemic symptoms, such as weakness or fatigue, which can be symptoms of cancer itself, diagnosis may be challenging. NAEs are rare but can be severe, so it is necessary for doctors to diagnose and take measures as early as possible. In this review, we intend to summarize the recent progress of nAEs, including various types of nAEs in the CNS and peripheral nervous systems (PNS), as illustrated in Fig. (1). We also introduced the possible mechanisms of nAEs based on existing clinical findings and mouse models, as well as diagnostic methods and immunomodulation treatment.
Fig. (1).
The positive and negative effects of immune checkpoint inhibitors (ICIs). (A) ICIs can help to activate T cell. Immune checkpoint receptors (CTLA-4, PD-1) expressed on T cells can interact with ligands (CD80/86, PD-L1) on antigen-presenting cells (APC) (i) or tumor cells (ii), thereby inhibiting T cell activation. These interactions can be blocked using ICIs (Usually monoclonal antibodies). (B) The simplified classification of neurological immune-related adverse events (nAEs). NAEs can be classified into two groups: central nervous system (CNS, right upper panel) or peripheral nervous system (PNS, right lower panel). (A higher resolution / colour version of this figure is available in the electronic copy of the article).
2. NEUROLOGICAL irAEs (nAEs)
Evidence on nAEs is limited, most of which are brief safety reports from clinical trials or individual case reports. The Common Terminology Criteria for Adverse Events are used as a standard to estimate toxic effects, which are also used as grading criteria to nAEs, classifying adverse events into five grades, including mild (1-2), severe (3-4), and fatal (5). As for the incidence of nAEs, 6-12% of patients experienced mild (grade 1-2) and nonspecific neurologic symptoms (headaches, dizziness, sensory impairment, etc.) [13, 14]. The incidence of grade 3-4 irAEs was lower, 0.2-0.4% with nivolumab or pembrolizumab (anti-PD-1) [13, 14], 0.3-0.8% with ipilimumab (anti-CTLA-4) [15, 16], and up to 0.7% when nivolumab and ipilimumab were used in combination [5]. Overall, grade 3-5 nAEs were regarded no more than 1% [10, 17-21]. However, the grading criteria were proposed before nAEs gained prominence, and did not show the real threat due to their dynamic and potentially rapid progress.
The onsets of nAEs are variable and extensive, ranging from a few days to several months. Some patients suffer from different types of nAEs or even other irAEs. For instance, Shigeaki et al. reported that 30% of patients with myasthenia gravis had myositis at the same time, 25% also had myocarditis [22], and Larkin et al. reported multiple cases with two nAEs and one case with three nAEs [18]. In this study, we will also look at each nAE. The percentage of each nAE, and the gender, age, and fatality, are listed in Table 2.
Table 2.
nAEs involving the central and peripheral nervous system.
| Type of nAEs | Incidence | Male/Female | Age | Dead/Alive |
|---|---|---|---|---|
| Encephalitis | 0.1-0.2% | 19/14 | 58.6 (20-83) | 9/24 |
| Aseptic meningitis | 0.1-0.2% | NA | NA | 1/7 |
| Hypophysitis | 0-18.3% | 409/224 | 63 (20-88) | 21/612 |
| Myasthenia gravis | 0.1-0.2% | 35/29 | 72.3 (45-86) | 22/42 |
| Myositis | 0.1-0.2% | 39/22 | 71.2 (25-89) | 12/68 |
| Peripheral neuropathies | < 3% | NA | NA | 1/4 |
| Guillain-Barre Syndrome | 0.2-0.3% | 7/4 | 59.1 (45-77) | 3/8 |
| Chronic Inflammatory Demyelinating Polyradiculoneuropathy | NA | 1/2 | 71.3 (44-85) | 0/3 |
| Cranial nerves palsies | NA | 4/1 | 48.2 (27-65) | 0/5 |
Abbreviations: Nivo nivolumab, Ipi ipilimumab, Pembro pembrolizumab, nAE neurological immune related adverse event, ICI Immune checkpoint inhibitors.
2.1. NAEs Involving the Central Nervous System
2.1.1. Encephalitis
The incidence of encephalitis in patients treated with ICI is 0.1-0.2% (Table 3) [18, 23-26]. Patients with encephalitis often have seizures, ataxia, episodes of increased confusion, abnormal behavior, and alterations of consciousness [23, 24]. Galmiche et al. gathered cases of encephalitis and found that the median age of onset was 60 years (ranged from 20-78), and the usual interval between the start of single ICI treatment and the onset of encephalitis was 12 weeks (13 weeks for combination therapy) [27]. Since ICI-induced encephalitis is a diagnosis of exclusion, other causes must be ruled out, such as metastases, infections, paraneoplastic conditions, and toxic metabolite [28]. Touat et al. demonstrated that patients with ICI-induced encephalitis showed a non-specific pattern, and these symptoms could be used for diagnosis, which seemed to be accurate in Galmiche’s research [10, 27].
Table 3.
Series of immune checkpoint inhibitors-associated encephalitis.
| Refs. | Year | Gender | Age | Cancer | ICIs Used | Onset Weeks | Treatments | Outcome |
|---|---|---|---|---|---|---|---|---|
| Mandel [23] | 2014 | Male | 66 | Melanoma | Nivo | 16 | No treatments | Resolved |
| Conry [35] | 2015 | Male | 41 | Melanoma | Ipi | 6 | PED + Methylpred | Improved |
| Stein [26] | 2015 | Male | 56 | Melanoma | Ipi | 15 | Methylpred | Resolved |
| Williams [36] | 2016 | Female | 50 | Melanoma | Nivo-Ipi | 2.5 | Methylpred + IVIG + Rituximab | Resolved |
| - | - | Male | 60 | SCLC | Nivo-Ipi | 0.5 | PED | Improved |
| Salam [24] | 2016 | Male | 64 | Melanoma | Pembro | 52 | Methylpred | Stable |
| Choe [37] | 2016 | Female | 45 | Melanoma | Ipi | 9 | Methylpred + IVIG | Improved |
| Brown [38] | 2017 | Male | 67 | Melanoma | Pembro | 4 | Prednisolone | Improved |
| Schneider [25] | 2017 | Male | 78 | NSCLC | Ipi | 28 | PED | Resolved |
| Bossart [39] | 2017 | Female | 60 | Melanoma | Ipi-Pembro | 12 | None | Died |
| Larkin [18] | 2017 | Female | 53 | Melanoma | Nivo-Ipi | 7 | DEX | Improved |
| - | - | Male | 61 | Melanoma | Nivo-Ipi | 15 | Methylpred + IVIG | Resolved |
| - | - | Male | 57 | Melanoma | Nivo-Ipi-nivo | 44 | Methylpred | Resolved |
| - | - | Female | 83 | Melanoma | Nivo | 8 | None | Died |
| - | - | Female | 58 | Melanoma | Ipi-ivo | 22 | Methylpred + IVIG | Resolved |
| Ito [40] | 2017 | Male | 75 | SCLC | Ipi | 13 | Methylpred + IVIG + Rituximab | Improved |
| Richard [28] | 2017 | Male | 74 | NSCLC | Nivo | 1 | Methylpred | Resolved |
| Strik [41] | 2017 | Male | 53 | Non HL | Nivo | 30 | Methylpred + IVIG + Cyclophos | Stable |
| Kyriakos [42] | 2018 | Male | 46 | EMC | Cemi | 8 | Methylpred + IVIG + Rituximab | Died |
| Chaucer [43] | 2018 | Male | 44 | RCC | Nivo | 2 | None | Died |
| Leitinger [44] | 2018 | Female | 67 | NSCLC | Nivo | 2. 5 | Methylpred + IVIG | Died |
| Zurko [45] | 2018 | Male | 20 | HL | Nivo | 2 | DEX | Improved |
| Burke [29] | 2018 | Female | 64 | OC | Nivo | 14 | Methylpred + PE | Resolved |
| Shah [46] | 2018 | Female | 66 | NSCLC | Nivo | 18 | Methylpred + PE + Rituximab | Decline |
| - | - | Female | 44 | NSCLC | Nivo | 8 | Methylpred + PE | Improved |
| Matsuoka [47] | 2018 | Male | 60 | PC | Nivo | 4 | Methylpred | Died |
| Kopecky [48] | 2018 | Male | 63 | RCC | Nivo | 12 | Methylpred + Infliximab | Died |
| De la Hoz [49] | 2018 | Female | 28 | HL | Nivo | 3 | PED | Resolved |
| Galmiche [27] | 2019 | Female | 62 | NA | Nivo, Ipi, Pembro | 6 | Methylpred + PED + IVIG | Died |
| - | - | Female | 78 | NA | Pembro | 90 | PED | Died |
| - | - | Male | 82 | NA | Pembro | 17 | Methylpred + PED + IVIG | Resolved |
| - | - | Female | 42 | NA | Nivo, Ipi | 1 | PED | Resolved |
| - | - | Male | 68 | NA | Nivo, Ipi | 2 | PED | Resolved |
Abbreviations: Nivo nivolumab, Ipi ipilimumab, Pembro pembrolizumab, Cemi cemiplimab, Cyclophos cyclophosphamide, Methylpred methylprednisolone, PED Prednisone, DEX Dexamethasone, IVIG intravenous immunoglobulins, SCLC small cell lung cancer, NSCLC non-small cell lung carcinoma, HL hodgkin lymphoma, EMC Extraskeletal Myxoid chrondrosarcoma, RCC renal cell carcinoma, OC ovarian cancer, PC pleomorphic carcinoma, ICIs Immune checkpoint inhibitors, PE plasma exchange, NA not applicable.
The patient’s brain MRI showed T2 hyperintensities in different areas, such as dentate nuclei [27], limbic structures [24], the right frontal, and left occipital lobes [23]. Dural enhancement and meningeal enhancement were also reported [23, 24]. However, some patients might have a normal brain MRI [25]. All reported cases indicated that cerebrospinal fluid (CSF) analysis was abnormal, often showing lymphocytic pleocytosis and negative cytopathology. The lumbar puncture tended to show a high white blood cell count [29].
If encephalitis is diagnosed, ICI should be interrupted immediately and followed by corticosteroid treatment, which is usually regarded as first-line therapy. Due to the rapid development of nAEs, the delay of corticosteroids treatment often leads to death [27]. Moreover, atypical cases of encephalopathies have been reported, such as brain vasculitis [30], exacerbation of multiple sclerosis [31], and posterior reversible encephalopathy syndrome-like symptoms [32-34].
2.1.2. Aseptic Meningitis
Aseptic meningitis has an incidence of approximately 0.1-0.2% in patients who received ipilimumab treatment (Table 4) [18, 23-26]. In addition, Strominger et al. reported one case with aseptic meningitis after nivolumab plus ipilimumab treatment [50], and Cordes et al. also reported one case with nivolumab monotherapy [51].
Table 4.
Series of immune checkpoint inhibitors-associated meningitis.
| Refs. | Year | Gender | Age | Cancer | ICIs Used | Onset Weeks | Treatments | Outcome |
|---|---|---|---|---|---|---|---|---|
| Yang [52] | 2007 | NA | NA | NA | Ipi | 12 | DEX | Resolved |
| Bot [54] | 2013 | Male | 51 | Melanoma | Ipi | 3 | DEX | Resolved |
| Oishi [53] | 2017 | Male | 59 | Melanoma | Ipi | 10 | Prednisolone + Methylpred | Improved |
| Spain [19] | 2016 | NA | NA | NA | Nivo-Ipi | 2 | ICI held then restarted | Resolved |
| - | - | NA | NA | NA | Ipi | 5 | ICI discontinued, no steroids | Resolved |
| - | - | NA | NA | NA | Ipi | 5 | Prednisolone | Resolved |
| Voskens [20] | 2013 | Female | 52 | Melanoma | Ipi | 4 | Steroids | Resolved |
| Cordes [51] | 2019 | Male | 58 | BLCA | Nivo | 48 | Methylpred + DEX | Died |
Abbreviations: ICI Immune checkpoint inhibitors, Nivo nivolumab, Ipi ipilimumab, Nivo-Ipi combined nivolumab and ipilimumab, BLCA bladder urothelial carcinoma, Methylpred methylprednisolone, DEX Dexamethasone, NA not applicable.
The onset of aseptic meningitis usually occurred within 1 to 7 weeks after the start of ICI treatment. Most patients got a fever, usually associated with headache [52-54]. Intermittent horizontal diplopia was observed, and MRI might show abnormalities, such as diffuse leptomeningeal and cranial nerve enhancement [50] or meningeal enhancement [53]. CSF analysis showed lymphocytic meningitis with elevated proteins. All patients’ CSF was negative for cytopathology [10]. Reported cases received discontinuation of ICI and recovered after the treatment of steroids with only one death [51].
2.1.3. Hypophysitis
For CTLA-4 drugs (ipilimumab), ICI-induced hypophysitis is presented an incidence of 0-17% [55-57], while 0.4-5% for tremelimumab [56, 58]. Other ICIs have a relatively low incidence, approximately less than 1% for nivolumab [59] or pembrolizumab [60]. It has been reported that ipilimumab treatment may have an incidence of 3.2%, compared to a two-fold (6.4%) incidence when combined ipilimumab with a PD-1 inhibitor and low (0.4%) with anti-PD-1 treatment alone [61]. The median onset time of ipilimumab-induced hypophysitis varied between studies, commonly ranging from 8.5-13 weeks, but the earliest was 6 weeks, and the latest was 24 weeks [62-65]. It has been proposed that the higher the dose, the higher the incidence of hypophysitis will be, which also varied with the application of adjuvant therapy [55, 66, 67], while some reports disagree with it [65].
The clinical manifestation often comprised a diversity of symptoms, including, but not limited to, headaches, weakness, fatigue, and weight loss [34]. Visual disturbances and polyuria/polydipsia rarely occur [68]. Other symptoms were also reported, such as confusion, hallucination, memory loss, erectile dysfunction, loss of libido, cold intolerance, dizziness, and insomnia [56, 69-71].
Patients having hypophysitis tended to show an abnormal level of the anterior pituitary hormone, usually decreased thyrotropin, corticotropin, and gonadotropin [68]. Hence, the decline of thyrotropin (usually 80% or more) might implicate the development of ipilimumab-induced hypophysitis [72]. In addition, the thyrotoxic symptom of thyroiditis could probably be another irAE, where the level of fT4 (free thyroid hormone) was raised. Therefore, it is a good way to diagnose this type of irAE by checking the fT4 level [68]. In some patients, MRI showed a uniformly enlarged pituitary gland, which in some ways resembled primary hypophysitis, possibly so-called ‘bright spot’ with thickening or disappearance of the neurohypophysis [64, 72-74].
2.2. NAE Involving the Peripheral Nervous System
2.2.1. Myasthenia Gravis
It is estimated that the incidence of myasthenia gravis is approximately 0.1-0.2% (Table 5) [22, 75-82]. It is tended to occur in men with a median age of 70, and the onset is usually within 2-6 weeks [83, 84]. The anti-PD-1 treatment seemed to be easier to cause myasthenia gravis [22]. Patients often present with fluctuating weakness of muscle, including bulbar, ocular, even respiratory muscles [85], sometimes associated with axial and proximal limb weakness and/or severe dyspnoea [22, 83].
Table 5.
Series of immune checkpoint inhibitors-associated myasthenia gravis.
| Refs. | Year | Cases | M/F | Age | PD-1, PD-L1, CTLA-4, Combinationa | Onset | AChRb | Previous MGc | CKb |
Outcome (Died/Resolved/
Improved) |
|---|---|---|---|---|---|---|---|---|---|---|
| Makarious [83] | 2017 | 23 | 13/10 | 74.5 (59-86) | 17/0/4/2 | 42 (14-84) days | 59% | 6 | 41% | 6/4/12 |
| Suzuki [22] | 2017 | 12 | 6/6 | 73.5 ± 6. 3 | 12/0/0/0 | 29 ± 13 days | 83% | NA | 83.3% | 2/2/7 |
| Gonzalez [90] | 2017 | 12 | 5/7 | 71.7 (59-81) | 8/0/3/1 | ≤ 4 cycles | 67% | 4 | 100% | 4/1/7 |
| Takamatsu [84] | 2018 | 17 | 11/6 | 69 (45-79) | 14/0/1/2 | ≤ 4 cycles | 82% | 2 | 93% | 10/0/7 |
Abbreviations: aNumber of cases treated with indicated ICIs. bPercentage of cases with abnormal indicated protein levels. cNumber of cases previously diagnosed with myasthenia gravis. Abbreviations: M/F Male/Female, CK creatine phosphokinase, AChR acetylcholine receptor, MG myasthenia gravis, PE plasma exchange, PD-1 programmed cell death protein 1, PD-L1 programmed cell death ligand 1, CTLA-4 cytotoxic T-lymphocyte associated protein 4, NA not applicable.
Noteworthy, patients with myasthenia gravis may experience a decrease in a compound muscle action potential in response to repeatedly low-frequency nerve stimulation of about 3 Hz. It is suggested that the electrodiagnosis usually results in a comparatively low positive predictive value. This was validated by the fact that only about half of assessed patients could be detected with enhanced jitter on single fiber electroneuromyography (ENMG) [86, 87]. Suzuki et al. proposed that this phenomenon could be attributed to the co-occurrence of myositis [22]. Approximately 70% of all patients with immune-related Myasthenia Gravis (irMG) tested positive for acetylcholine receptor (AChR), and some patients might have a mild-to-marked elevation of serum creatine kinase (CK) levels [22, 75-82].
The coexistence of myasthenia gravis and myositis frequently occurs, which is a risk factor for the occurrence of myasthenic crisis that requires intensive care [12, 87, 88]. Of note, most of the fatalities of irMG were associated with myositis and/or myocarditis [12]. It is therefore of great importance for physicians to diagnose this condition so that early action can be taken. It was reported that a relapsed ocular myasthenia gravis patient with positive AChR and muscle-specific kinase antibodies eventually developed a complication of irMG and immune-related myositis (irMyositis), which might provide a reference to physicians [89].
2.2.2. Myositis
For ICI therapy, one of the most common nAEs is irMyositis. IrMyositis predominantly occurs in anti-PD-1 treated patients. Selected reports of irMyositis are listed in Table 6. The clinical presentation of irMyositis is different from inflammatory paraneoplastic myopathies [87], and it can be similar to that of myasthenia gravis [10]. IrMyositis occurs more frequently in older men than older women, whose average age is about 70 [91, 92]. The onset after the initiation of ICI is often within 60 days, while it can be shorter if receiving combined treatment of ipilimumab or nivolumab [87, 93].
Table 6.
Series of immune checkpoint inhibitors-associated myositis.
| Refs. | Year | Cases | M/F | Age | PD-1, PD-L1, CTLA-4, Combinationa | Onset Days | EMG (Analyzed & Myopathy)b | Muscle Biopsyc | CK (IU/L) |
Outcome (Died/Resolved/
Improved) |
|---|---|---|---|---|---|---|---|---|---|---|
| Liewluck [92] | 2018 | 5 | 5/0 | 76 (55-86) |
5/0/0/0 | 30 (17-32) |
100% & 60% | 40% | 444 (72-7307) |
2/0/3 |
| Shah [94] | 2019 | 6 | 5/1 | 64 (50-81) |
1/1/1/3 | 37.8 (14.7-119.7) |
50% & 100% | 17% | 514-13710 | 2/2/2 |
| Touat [87] | 2018 | 10 | 7/3 | 73 (56-87) |
7/1/0/2 | 25 (5-87) |
90% &100% | 40% | 2668 (1059-16620) |
0/0/10 |
| Seki [91] | 2019 | 19 | 13/6 | 70 (25-84) |
19/0/0/0 | 29 | 58% & 78% | 21% | 5247 | 1/10/5 |
| Kadota [93] | 2019 | 15 | 7/8 | 73 (50-89) |
8/0/4/3 | 28 ± 14 | 40% & 100% | 33% | 2812 (794-20270) |
4/1/11 |
| Moreira [96] | 2019 | 19 | NA | NA | 14/0/0/5 | NA | NA | 32% | 2370 (263-7697) |
2/10/4 |
| Kimura [78] | 2016 | 1 | 1/0 | 80 | 1/0/0/0 | 14 | 100% & 100% | 100% | 7740 | 0/0/1 |
| Lecouflet [98] | 2013 | 1 | 0/1 | 67 | 0/0/1/0 | NA | NA | 100% | NA | 0/0/1 |
| Pinto [99] | 2016 | 1 | 0/1 | 86 | 1/0/0/0 | NA | 100% & 100% | 100% | 1499 | 0/0/1 |
| Bilen [95] | 2016 | 1 | 1/0 | 73 | 0/0/0/1 | 20 | NA | 100% | 13710 | 1/0/0 |
| Anna [100] | 2017 | 2 | 0/2 | 60 | 0/0/2/0 | 42, 56 | NA | NA | Normal | 0/1/0 |
Abbreviations: aNumber of cases treated with indicated ICIs. bPercentage of cases examined by EMG and percentage of those with myopathy. cPercentage of cases with abnormal muscle biopsy. Abbreviations: M/F Male/Female, EMG electromyogram, CK creatine phosphokinase, PD-1 programmed cell death protein 1, PD-L1 programmed cell death ligand 1, CTLA-4 cytotoxic T-lymphocyte associated protein 4, IVIG intravenous immunoglobulin, PE plasma exchange, NA not applicable.
Most patients may experience myalgia located on the neck, back, or proximal limbs, and then they may suffer from muscle weaknesses [85, 91]. Proximal limb with symmetrical weakness of pelvic and scapular girdle is typical, and axial muscle weakness, including cervical extensor muscles and dropping head, is common [87, 91]. Ocular involvement, including ptosis and ophthalmoparesis, facial weakness, and bulbar muscle involvement, have also been reported [87, 91-94]. Similar to other irAEs, some patients may have fevers [87], but typically without cutaneous lesions [10].
Patients with severe symptoms [91] or treated with ICIs combination [93, 94] may show high CK levels. However, it was reported that patients whose muscle biopsy indicated myositis had normal CK levels [92]. Sometimes patients may have elevated levels of transaminases and troponin-T [95]. AChR and paraneoplastic antibodies are typically negative, while antibodies to signal recognition particles can be detected in individual cases [10, 87, 91, 92, 94, 96]. Myopathic motor unit potentials are recorded in most patients, while few patients also show a decrease in compound muscle action potentials [87, 94]. Noteworthy, patients with normal ENMG may also have myositis [11, 91].
Muscle biopsies often reveal varying degrees of necrotizing muscle fibers in relation to inflammatory infiltration, which may have a remarkable focal character. Hence, it is significant to select muscle biopsy sites based on clinical performance, MRI or ENMG, to avoid nonspecific or false-negative outcomes [85, 87]. The majority of ICI-related myositis cases have been reported to respond favorably to corticosteroids or other additional immune-modulating treatments [97], while clinical rehabilitation in the absence of immunomodulatory treatment is noteworthy.
2.2.3. Peripheral Neuropathies
Peripheral neuropathies occurred in less than 3% of all patients [11, 17-21]. Studies are underway to elucidate whether the incidence of peripheral neuropathies is lower than that of cytotoxic chemotherapy. For peripheral nerves, it is safer to use ICI therapy than traditional chemotherapy, based on statistics [101, 102]. The motor or sensory peripheral roots or limb may be focally or diffusely influenced by ICI-related peripheral neuropathies, presenting as demyelinating or axonal neuropathies, sometimes a mix of both. Cranial nerve palsies can affect the facial, optic, and abducens nerves [18, 20, 103-105]. Inflammations of peripheral nerves, including meningoradiculitis and meningoradiculonevrititis, are also reported [103, 106].
2.2.4. Polyradiculoneuropathy
The incidence of immune-related demyelinating polyradiculoneuropathy (irDP) is commonly considered very low, assessed only 0.1-0.3% of patients with PD-1/CTLA-4 antibodies treatment [10, 18, 107]. However, a meta-analysis revealed that peripheral neuropathy was high, 7.6% and 3.0% for PD-1 and PD-L1 inhibitors, respectively [108]. Differences in diagnostic criteria may explain this discrepancy. The onset was a median of 3 to 3.5 doses of ICIs [96]. Most patients showed symptoms that were sensor motor-related, sometimes accompanied by oligoclonal bands, including cranial nerve, manifestations of dysautonomia, or bulbar symptoms [79, 109-111]. CSF assay presented with the separation of albuminocytologic, often accompanied by lymphocytosis. Furthermore, oligoclonal bands were detected in an individual patient through CSF analysis [104, 112]. Electromyogram (EMG) showed evidence of demyelination with a sural sparing pattern, prolonged F wave latencies, and decreased motor conduction velocities and sometimes conduction block [18].
2.2.5. Guillain-Barre Syndrome (GBS)
The incidence of GBS was about 0.2-0.3% in patients treated with PD-1 inhibitors [21, 113]. As shown in Table 7, the onset is variable, which can be as early as 2 doses of anti-PD-1 agents, or much later to 18 doses (Midpoint 4.5) [88]. The clinical presentation was roughly similar to conventional GBS, such as electro-diagnostical abnormalities or CSF protein levels. Although CSF pleocytosis was mildly changed [114, 115], it could not be used as a distinguishing feature. Enhancement of the nerves in the MRI had also been reported [115]. In addition, Fukumoto et al. found a case with anti-ganglioside antibodies positive [116], although it was rarely encountered.
Table 7.
Series of immune checkpoint inhibitors-associated Guillain-Barre Syndrome.
| Refs. | Year | Gender | Age | Cancer | ICI Used | Onset | CSF (mg/dL) | Treatments | Outcome |
|---|---|---|---|---|---|---|---|---|---|
| Bot [54] | 2013 | Male | 63 | Melanoma | Ipi | 15 weeks | 89 | IVIG | Died |
| Caroline [111] | 2013 | Male | 65 | Melanoma | Ipi | 8 weeks | 160 | Tacrolimus + IVMP + PE | Died |
| Wilgenhof [112] | 2011 | Female | 57 | Melanoma | Ipi | 10 weeks | 167 | IVMP | Improved |
| Maleissye [114] | 2016 | Female | 45 | Melanoma | Pembro | 8 weeks | 56 | IVIG + Prednisolone | Not Improved |
| Supakornnumporn [117] | 2017 | Male | 77 | Melanoma | Nivo-Ipi | 10 weeks | 86 | IVIG + PED | Improved |
| Gu [110] | 2017 | Female | 49 | Melanoma | Nivo-Ipi | 5 days | 115 | IVIG + IVMP + PE + PED | Improved |
| Nukui [118] | 2018 | Male | 45 | Nasal cancer | Nivo | 10 weeks | 350 | IVIG + Steroids | Improved |
| Botha [115] | 2017 | Male | 64 | Melanoma | Ipi-Pembro | 3 months | 195 | IVIG + Steroids | Improved |
| Ong [119] | 2018 | Male | 66 | LUAD | Pembro | 7 weeks | NA | Methylpred + IVIG | Improved |
| Jacob [120] | 2016 | Female | 68 | LUSC | Nivo | 3 months | 85 | IVIG + PE | Died |
| Schneiderbauer [121] | 2016 | Male | 51 | Melanoma | Nivo | 5 months | 73 | IVIG + Corticosteroids | Improved |
Abbreviations: CB conduction block, CMAP compound muscle action potential, CSF cerebrospinal fluid, CV conduction velocity, IVMP intravenous Methylpred, PE plasma exchange, SNAP sensory nerve action potential, IVIG intravenous immunoglobulin, Nivo nivolumab, Ipi ipilimumab, Pembro pembrolizumab, LUAD lung adenocarcinoma, LUSC lung squamous cell carcinoma, Methylpred methylprednisolone, PED Prednisone, NA not applicable.
2.2.6. Chronic Inflammatory Demyelinating Polyradiculoneuropathy (CIDP)
ICI-related CIDP was so rare that only three cases were reported, as listed in Table 8. The patients were treated with PD-1/CTLA-4 antibodies. Since there were not enough cases, the onset varied enormously. EMG diagnosis presented demyelinating symptoms for all cases. However, for one patient, MRI showed enhanced dorsal roots of C7/T1. All cases presented with elevated CSF protein levels, and the last one had a mildly altered pleocytosis as described above [115]. It is noteworthy that chronic evolution was rarely reported, but the initial acute presentations eventually occurred [122].
Table 8.
Series of immune checkpoint inhibitors-associated Chronic Inflammatory Demyelinating Polyradiculoneuropathy.
| Refs. | Year | Gender | Age | Cancer | ICI used | Onset (week) | CSF (mg/dL) | Treatments | Outcome |
|---|---|---|---|---|---|---|---|---|---|
| Liao [79] | 2014 | Male | 44 | Melanoma | Ipi | 1 | 44 | PE | Improved |
| Maleissye [114] | 2016 | Female | 85 | Melanoma | Ipi, Pembro, Binimetinib | 31 | 74 | Glucocorticoids + PE | Improved |
| Tanaka [123] | 2016 | Female | 85 | Melanoma | Nivo | 2 | 358 | IVIG + Prednisolone | Improved |
Abbreviations: M male, F female, PE plasma exchange, CSF cerebrospinal fluid, IVIG intravenous immunoglobulin, Nivo nivolumab, Ipi ipilimumab, Pembro pembrolizumab, NA not applicable.
2.2.7. Motor Polyradiculopathy
Motor polyradiculopathy was rarely reported [88, 124]. Sepúlveda et al. reported a case of asymmetric limb weakness that worsened within a period of weeks to months. After the onset of symptoms, the patients underwent EMG testing several times (day 10, 36, 86, and 395, respectively) [124]. The EMG was normal at day 10, while at day 36 and later, it presented a reduced compound muscle action potential amplitudes. MRI indicated contrast-enhanced ventral lumbosacral roots. After intravenous immunoglobulins (IVIG) and plasma exchange, the patient finally improved.
2.2.8. Vasculitic Neuropathy
Vasculitic neuropathy was rare, and there were only two reported cases. Aya et al. found a case of symmetrical and progressive weakness in the lower limb [125]. Moreover, neuro-biography presented monocyte invasions of small intimal vessels, which might indicate microvascular inflammation [125]. On the contrary, the other patient had an asymmetric loss of sensation and multifocal neuropathy. In addition, CSF assay showed cell nucleation and elevated protein levels, and peripheral neuro-biopsy indicated a necrotizing vasculitis [88]. Both improved with steroid monotherapy.
2.2.9. Cranial Nerves Neuropathies
Cranial nerve palsies have been reported to affect the optic nerve, abducens, and facial nerves (as listed in Table 9). Of note, cranial nerve palsies without immune demyelinating polyradiculoneuropathy have also been reported [104, 105]. Cranial nerve palsies mainly occurred in patients treated with ipilimumab, and the onset ranged from 2 weeks to 18 weeks. The clinical presentation tended to be acute. There were reported cases of optic nerve enhancement with MRI, as well as normal MRI [126]. All cases improved with steroid therapy.
Table 9.
Series of immune checkpoint inhibitors-associated cranial nerves palsies.
| Refs. | Year | Gender | Age | Cancer | ICI used | Onset (week) | CSF | Treatments | Outcome |
|---|---|---|---|---|---|---|---|---|---|
| Altman [103] | 2015 | Male | 32 | Melanoma | Ipi | 8 | NA | PED | Improved |
| Boisseau [104] | 2017 | Female | 27 | RCC | Ipi | 20 | Elevated protein | Methylpred + steroids + PE | Improved |
| Voskens [20] | 2013 | Male | 65 | Melanoma | Ipi | 18 | NA | Methylpred + DEX | Improved |
| Walsh [127] | 2015 | Male | 53 | Melanoma | Ipi | NA | Lymphocytosis, Elevated protein | steroids | Improved |
| Fellner [126] | 2018 | Male | 64 | RCC | Nivo-Ipi | 8 | NA | PED | Improved |
Abbreviations: CSF cerebrospinal fluid, RCC renal cell carcinoma, PE plasma exchange, Nivo nivolumab, Ipi ipilimumab, Nivo-Ipi combined nivolumab and ipilimumab, Methylpred methylprednisolone, PED Prednisone, DEX Dexamethasone, NA not applicable.
3. POTENTIAL MECHANISMS OF nAEs
The exact mechanism and the pathophysiology of ICI-related nAEs are not clear, and the reason why only a small number of patients suffering from nAEs is unknown. There are two hypotheses [126]. One is the “hidden autoimmunity” theory, which suggests that the patients typically have a preexisting preclinical autoimmune disease inhibited by immunological tolerance. When treated with ICIs, immunological tolerance is broken, and clinically-evident autoimmune diseases come into existence. The other theory, “molecular mimicry,” holds that the antigen in tumor cells has similar epitopes as a neuron, which not only leads to T-cell-mediated autoimmunity against tumor cells but also affects neurons.
As to the “hidden autoimmunity” theory, the key players are autoantibodies, such as anti-ganglioside [112, 121], anti-GAD65 [46], anti-Hu [42], anti-NMDA (N-methyl-D-aspartate) receptor [36], anti-AChR, and anti-striational antibodies [84]. The suspected antibodies are listed in Table 10. Here is an explanation for how ICIs lead to the production of autoantibodies. Immune checkpoint receptors are expressed on B regulatory cells and Tfh cells, which are critical for antibody production, thereby attenuating the humoral response [128]. Hence, ICIs may block this pathway, leading to the production of autoantibodies. Moel et al. reported that some patients treated with ipilimumab developed autoantibodies, which indicated that blockade of CTLA-4 also led to the loss of B cell self-tolerance [129]. Sage et al. found that deletion of CTLA-4 on Tfr/Tfh/Treg cells contributed to the production of antibodies, which might explain the generation of autoantibodies in patients treated with ipilimumab (Anti-CTLA-4) [130]. However, some reports indicate that anti-ganglioside antibodies are undetectable in GBS patients [54, 112]. Despite this, some patients with nAEs showed no autoantibody. Hence, there may be some unknown autoantibodies that need to be identified.
Table 10.
Possible mechanisms about nAEs.
| Refs. | Associated Molecules | Disease | Cancer |
|---|---|---|---|
| Touat [10] | NMDA-R | Encephalitis | Melanoma/SCLC |
| - | CASPR2 | Encephalitis | Melanoma |
| Shah [46] | GAD65 | Encephalitis | LUAD |
| Papadopoulos [42] | Anti-Hu antibodies | Encephalitis | SCLC |
| Kimura [78] | Autoreactive CTLs | Myositis | NA |
| Bilen [95] | Anti-striated muscle antibody | Myositis | PTCC |
| Takamatsu [84] | Anti-striational antibodies | Myasthenia gravis | Melanoma/PTCC |
| Wang [139] | Anti-AChR antibodies | Myasthenia gravis | NA |
| Schneiderbauer [121] | Anti-ganglioside antibody | Guillain-Barré Syndrome | Melanoma |
Abbreviations: CTL cytotoxic T lymphocytes, NMDA-R N-Methyl-D-aspartate receptor, CASPR2 contactin-associated protein like 2, SCLC small cell lung cancer, AchR acetylcholine receptor, GAD Glutamic Acid Decarboxylase, Anti-Hu anti-neuronal nuclear antibody, PTCC papillary transitional cell carcinoma, NA not applicable.
For “molecular mimicry,” the results of mouse models and clinic findings can support this theory. Yshii et al. established a neo-antigen (HA, Haemagglutinin) in transgenic mice to study it, as shown in Table 11. HA was expressed on Purkinje cells (in situ) and transplanted breast tumor cell lines. After treatment of CTLA-4 inhibitor, they found the activation and migration of HA-specific T cells, causing manifest neural inflammation and paraneoplastic disease [131]. Their model suggested that CTLA4 blockade induced the production of antigen-specific T cells, thereby leading to neurotoxicity, possibly due to shared antigens on a neuron. A clinical study indicated that after ICI-therapy, the same infiltration T cells were identified in myocardial, skeletal muscle, and tumor samples. Whole-transcriptome sequencing indicated that there was a similar epitope between the tumor and striated muscle [132].
Table 11.
Mouse models of nAEs.
| Refs. | nAEs | Checkpoint | Models |
|---|---|---|---|
| Yshii [131] | PND | CTLA-4 | HA-transgenic mice, anti-CTLA-4 therapy |
| Kroner [135] | EAE | PD-1 | PD-1 knockout mice |
| Rui [136] | EAE | PD-1 | PD-1 knockout mice |
| Salama [137] | EAE | PD-1 | WT mice, anti-PD-1 therapy |
| EAE | PD-1 | DO11.10 TCR transgenic mice, anti-PD-1 therapy | |
| Zhu [138] | EAN | CTLA-4 | WT mice, anti-CTLA-4 therapy |
Abbreviations: nAEs neurological immune-related adverse events, PND Paraneoplastic neurological disorder, EAE Experimental autoimmune encephalomyelitis, EAN Experimental autoimmune neuritis, MOG myelin oligodendrocyte glycoprotein, B6 C57BL/6, WT wild type, HA Haemagglutinin, TCR T cell receptor.
Despite the predominance of the above two theories, other hypotheses are also proposed. For example, the polymorphism of CTLA-4 may be related to the development of nAEs. There is evidence that mouse with CTLA-4 polymorphic subtypes develops an autoimmune disease, involving the increased generation of autoantibodies [133, 134].
Although these clinical studies suggest possible mechanisms, the current effort is to develop animal models for further investigation. So far, in addition to the HA-antigen transgenic mice established by Yshii et al., as described above, a variety of models have been used to study the mechanisms of nAEs (Table 11). Using PD-1 knockout mice, Kroner et al. developed an experimental autoimmune encephalomyelitis (EAE) mouse model [135]. They found that the absence of PD-1 contributed to the expansion of T lymphocytes, the number of which was positively correlated to the severity of the EAE. A similar model was developed, and almost the same results were observed by another independent group [136]. In contrast, Salama et al. established EAE in wild-type mice by MOG35-55 peptide induction [137]. After PD-1 blockade, they observed MOG-specific T-cell infiltration in CNS and an increase in anti-MOG antibodies. They also established a splenocyte transfer mouse model, immunized with OVA peptide to induce EAE, and found that OVA-specific T cells were significantly expanded after PD-1 inhibitor treatment. To study GBS, Zhu et al. established experimental autoimmune neuritis (EAN) model, and observed T cell infiltration and demyelination after anti-CTLA-4 treatment [138]. Despite some advances, there are currently insufficient mouse models and studies. Future establishments and investigations of more nAEs mouse models will be critical to understand neurotoxicity.
4. THE DIAGNOSIS AND TREATMENTS OF nAEs
4.1. Diagnostic Approach
The diagnosis of neurological complications associated with ICIs is usually challenging due to the fact that many factors should be excluded, including opportunistic infections, metabolic, paraneoplastic, and neoplastic conditions. It is necessary to perform a systemic assessment of the nervous system. Diagnostic work, including serological tests for infections, lumbar puncture, imaging, or other diagnostic tests, is needed to be done. The diagnostic approach relies on the part of the nervous system associated. Patients who are suspected of having CNS toxicity need CT or MRI with contrast in order to search for evidence of brain metastases and further diagnostic lumbar puncture to rule out leptomeningeal disease or inflammation. Cytopathology and inspection for bacterial and viral infections in CNS ought to be covered in CSF analysis. In contrast, if patients are suspected of suffering from PNS toxicity, electromyography with nerve conductions studies and CSF analysis are suggested. In addition, muscle MRI is applicable for patients with GBS-like illness or myositis [140]. Muscle biopsy should be done for patients suspected of myositis. Notably, Touat et al. proposed a diagnostic approach for encephalitis, which has been proved to be accurate by Galmiche et al. [10, 27]. They suggested that when a patient is presented with symptoms of encephalitis, a CT or MRI should be performed to rule out cancerous meningitis or brain metastases, and CSF should be analyzed to check for bacterial or viral infections. Once these have been ruled out, the patient can be diagnosed with ICI-induced encephalitis and receive immediate immunomodulatory therapy [10].
Diagnostic algorithms or neurologic workups are suggested in some reviews [10, 140]. All in all, it is necessary to remember that the probability of co-occurring disorders needs to be promptly examined in suspicious patients, while at the same time, the application of corticosteroids or other immunomodulatory therapy should not be delayed.
4.2. Treatments for nAEs
Corticosteroids are generally used as front-line therapy when other possible causes are eliminated. At the same time, ICI treatment ought to be discontinued. Patients with mild symptoms do not need immune-modulating treatments, while regular follow-up and patient self-monitoring are necessary. For grade 1-2 nAEs, corticosteroids are recommended, together with the withdrawal of ICIs. Patients with grade 3 or higher nAEs usually need high-dose corticosteroids. Discontinuing ICIs and early intervention with high-dose steroids are significant to resolve and improve outcomes. If the patient recovers from the toxicity, corticosteroids can be discontinued by tapering the dose for at least 2 months. Some reports indicate that they also use empiric antibiotics and antivirals [10].
Despite corticosteroids, other treatments ought to be considered, such as IVIG, plasmapheresis, anti-TNF-α antibodies (e.g. infliximab), mycophenolate mofetil, and rituximab. If patients have no or only a partial response to corticosteroids, these treatments ought to be considered. Of note, corticosteroids are not preferred when treating patients with GBS. Corticosteroids have consistently shown favorable outcomes in irDP but are not typically used for traditional GBS [85], which indicates that ICI-induced GBS may differ from traditional GBS.
Patients with mild symptoms or rapid resolution can be re-challenged. Galmiche et al. reported a case of re-challenged pembrolizumab [141], and Gutzmer et al. re-challenged a case with a different class of ICI [142]. Most patients recover fully or partially after immune-modulating treatments, while fatal cases have also been reported. It is significant to note that the response can be durable [19]. Whether corticosteroids will have a negative effect on the outcomes of cancer treatment, the OS rates and time-to-treatment failure indicate that they are not influenced by nAEs or immune-modulating treatments [143].
5. CONCLUSION AND PERSPECTIVE
With the increasing use of ICIs in oncology, nAEs are thought to be more common in cancer therapy. NAEs can be serious and sometimes fatal. Clinical symptoms can be abnormal and multi-focal, sometimes disabling or life-threatening. Clinicians should be aware of these potential complications, recognizing that rapid diagnoses are necessary to minimize the risk of fatality and long-term after-effects. Discontinuation of the ICIs, early treatment with corticosteroids and immunosuppressants are necessary to improve the outcomes of patients suffering from nAEs. Whether the history of autoimmune neurological disorders can be served as a predictor for nAEs, this possibility and its mechanism need to be further studied.
There is increasing evidence that the nervous system plays an important role in the development of cancer [144]. ICI-therapy, as the most important immunotherapy currently applied, causes various types of nAEs. Here we review the common nAEs and summarize the diverse neurotoxicities reported so far. However, there is much to be learned and investigated. The interaction between the nervous system and tumor cells is the central issue, and understanding the nAEs caused by ICI-therapy is critical to address this issue.
The emerging cellular and molecular mechanisms of ICI treatment-caused nAEs are being better understood. Both “hidden autoimmunity” and “molecular mimicry” theories provide possible hypotheses [126]. However, some basic issues are still needed to be further addressed. Most recently, Parker et al. proposed a possible mechanism of neurotoxicity caused by CAR-T immunotherapy [145]. They found that CD19 (Commonly used as targets of engineered CAR-T) were expressed by brain mural cells, which could be possible CAT-T targets, facilitating T-cell infiltration and causing neurotoxicity. This report, to some extent, provides a good reference for the neurotoxicity study of ICI.
Similarly, the question is very interesting that whether any ICIs directly have some impact on the nervous system. So far, no studies have shown that ICIs have a direct effect on the nervous system. However, it has been reported that PD-L1 can be expressed on oligodendroglia and microglia [146]. Hence, it is possible that ICIs can have a direct impact on these cells once ICIs penetrate the blood-brain barrier and engage with these cells. It is also reported that, in one case of hypophysitis, CTLA-4 can be expressed by pituitary endocrine cells, indicating a potential direct effect of tremelimumab (anti-CTLA4) [147, 148]. Of note, the ICIs penetration of the blood-brain barrier or blood-nerve barrier is still a question that remains to be studied [149-151]. Although current evidence is limited, it will be valuable to investigate whether ICIs have a direct toxic effect on nerve cells. PD-L1/CTLA-4 expression is currently measured in bulk tissues, and rare low-expressing cell types may be missed in such measurements. With some new technologies, such as single-cell RNA-seq, we can detect the expression level of PD-1/PD-L1/CTLA-4 molecules at a much adequate level, which may provide some new ideas for a possible mechanism of neurotoxicity.
Two autopsy reports on nAEs provided a useful clue that T cell infiltration is one of the causes of neurotoxicity. One report of a patient with ICI-induced hypophysitis showed lymphocytic infiltration in the anterior pituitary [147]. Another nAE report showed extensive demyelination, lymphoid infiltration, and massive macrophages [152]. However, there are few studies on how T cells infiltrate into the CNS during the development of nAEs. . Under certain conditions, T cells can access the parenchyma of CNS through three separate routes [153]. However, the exact mechanism of T cell infiltration, especially in nAEs, still needs to be further investigated.
Patients with nAEs may have better responses to cancer therapy. There is one report that, compared with other patients, a NSCLC patient with encephalopathy responded better to ICI treatment [154]. Since irAEs have been extensively investigated, we speculate that the same situations of irAEs may apply to nAEs. It has been reported that the overall survival (OS) of irAEs patients is significantly higher than that of patients without irAEs [155]. Patients with multiple irAEs had better OS than patients with only one irAEs. Additionally, patients experiencing irAEs showed better ORR and progression-free survival than those without irAEs [156, 157]. Although the performance of one patient in nAEs is not enough for us to draw a conclusion, combined with the results of irAEs, we can infer that nAEs may play a potentially beneficial role in cancer immunotherapy. This is very interesting and needs to be further verified.
Cancer immunotherapy and associated neurotoxicity offer opportunities for collaboration between oncologists and neurologists. We hope that this work will contribute to the understanding of nAEs. For individual ICIs, better defining the patterns of neurotoxicity, elucidating the possible mechanism, working out potential biomarkers, and refining therapeutic options are all worthy of attention in future studies.
ACKNOWLEDGEMENTS
Declared none.
LIST OF ABBREVIATIONS
- AChR
Acetylcholine Receptor
- CK
Creatine Kinase
- CNS
Central Nervous System
- CSF
Cerebrospinal Fluid
- CTLA-4
Cytotoxic T-Lymphocyte Associated Protein 4
- EAE
Experimental Autoimmune Encephalomyelitis
- EAN
Experimental Autoimmune Neuritis
- EMG
Electromyogram
- ENMG
Electro-Neuromyography
- ICIs
Immune Checkpoint Inhibitors
- irAEs
Immune-Related Adverse Events
- irDP
Immune Related Demyelinating Polyradiculoneuropathy
- irMG
Immune-Related Myasthenia Gravis
- irMyositis
Immune Related Myositis
- IVIG
Intravenous Immunoglobu-Lins
- MOG
Myelin Oligodendrocyte Glycoprotein
- nAEs
Neurological Immune-Related Adverse Events
- NMDA
N-Methyl-D-Aspartate
- ORR
Objective Response Rate
- OS
Overall Survival
- OVA
Ovalbumin-Specific
- PD-1
Programmed Cell Death Protein 1
- PD-L1
Programmed Cell Death Ligand 1
CONSENT FOR PUBLICATION
Not applicable
FUNDING
This work was supported by the Fundamental Research Funds for the Central Universities (2019CDYGYB024), the National Natural Science Foundation of China (31300726), and Chongqing Primary and Middle School Innovation Talent Training Project (CY200118).
CONFLICT OF INTEREST
The authors declare no conflict of interest, financial or otherwise.
REFERENCES
- 1.Singh S., Hassan D., Aldawsari H.M., Molugulu N., Shukla R., Kesharwani P. Immune checkpoint inhibitors: a promising anticancer therapy. Drug Discov. Today. 2020;25(1):223–229. doi: 10.1016/j.drudis.2019.11.003. [DOI] [PubMed] [Google Scholar]
- 2.Pardoll D.M. The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer. 2012;12(4):252–264. doi: 10.1038/nrc3239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Borghaei H., Paz-Ares L., Horn L., Spigel D.R., Steins M., Ready N.E., Chow L.Q., Vokes E.E., Felip E., Holgado E., Barlesi F., Kohlhäufl M., Arrieta O., Burgio M.A., Fayette J., Lena H., Poddubskaya E., Gerber D.E., Gettinger S.N., Rudin C.M., Rizvi N., Crinò L., Blumenschein G.R., Jr, Antonia S.J., Dorange C., Harbison C.T., Graf Finckenstein F., Brahmer J.R. Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N. Engl. J. Med. 2015;373(17):1627–1639. doi: 10.1056/NEJMoa1507643. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Garon E.B., Rizvi N.A., Hui R., Leighl N., Balmanoukian A.S., Eder J.P., Patnaik A., Aggarwal C., Gubens M., Horn L., Carcereny E., Ahn M.J., Felip E., Lee J.S., Hellmann M.D., Hamid O., Goldman J.W., Soria J.C., Dolled-Filhart M., Rutledge R.Z., Zhang J., Lunceford J.K., Rangwala R., Lubiniecki G.M., Roach C., Emancipator K., Gandhi L., Investigators K. Pembrolizumab for the treatment of non-small-cell lung cancer. N. Engl. J. Med. 2015;372(21):2018–2028. doi: 10.1056/NEJMoa1501824. [DOI] [PubMed] [Google Scholar]
- 5.Larkin J., Chiarion-Sileni V., Gonzalez R., Grob J.J., Cowey C.L., Lao C.D., Schadendorf D., Dummer R., Smylie M., Rutkowski P., Ferrucci P.F., Hill A., Wagstaff J., Carlino M.S., Haanen J.B., Maio M., Marquez-Rodas I., McArthur G.A., Ascierto P.A., Long G.V., Callahan M.K., Postow M.A., Grossmann K., Sznol M., Dreno B., Bastholt L., Yang A., Rollin L.M., Horak C., Hodi F.S., Wolchok J.D. Combined nivolumab and ipilimumab or monotherapy in untreated melanoma. N. Engl. J. Med. 2015;373(1):23–34. doi: 10.1056/NEJMoa1504030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Cheng W., Fu D., Xu F., Zhang Z. Unwrapping the genomic characteristics of urothelial bladder cancer and successes with immune checkpoint blockade therapy. Oncogenesis. 2018;7(1):2. doi: 10.1038/s41389-017-0013-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Polk A., Svane I.M., Andersson M., Nielsen D. Checkpoint inhibitors in breast cancer - Current status. Cancer Treat. Rev. 2018;63:122–134. doi: 10.1016/j.ctrv.2017.12.008. [DOI] [PubMed] [Google Scholar]
- 8.Ansell S.M., Lesokhin A.M., Borrello I., Halwani A., Scott E.C., Gutierrez M., Schuster S.J., Millenson M.M., Cattry D., Freeman G.J., Rodig S.J., Chapuy B., Ligon A.H., Zhu L., Grosso J.F., Kim S.Y., Timmerman J.M., Shipp M.A., Armand P. PD-1 blockade with nivolumab in relapsed or refractory Hodgkin’s lymphoma. N. Engl. J. Med. 2015;372(4):311–319. doi: 10.1056/NEJMoa1411087. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Samaan M.A., Pavlidis P., Papa S., Powell N., Irving P.M. Gastrointestinal toxicity of immune checkpoint inhibitors: from mechanisms to management. Nat. Rev. Gastroenterol. Hepatol. 2018;15(4):222–234. doi: 10.1038/nrgastro.2018.14. [DOI] [PubMed] [Google Scholar]
- 10.Touat M., Talmasov D., Ricard D., Psimaras D. Neurological toxicities associated with immune-checkpoint inhibitors. Curr. Opin. Neurol. 2017;30(6):659–668. doi: 10.1097/WCO.0000000000000503. [DOI] [PubMed] [Google Scholar]
- 11.Hottinger A.F. Neurologic complications of immune checkpoint inhibitors. Curr. Opin. Neurol. 2016;29(6):806–812. doi: 10.1097/WCO.0000000000000391. [DOI] [PubMed] [Google Scholar]
- 12.Psimaras D., Velasco R., Birzu C., Tamburin S., Lustberg M., Bruna J., Argyriou A.A. Immune checkpoint inhibitors-induced neuromuscular toxicity: From pathogenesis to treatment. J. Peripher. Nerv. Syst. 2019;24(Suppl. 2):S74–S85. doi: 10.1111/jns.12339. [DOI] [PubMed] [Google Scholar]
- 13.Weber J.S., Hodi F.S., Wolchok J.D., Topalian S.L., Schadendorf D., Larkin J., Sznol M., Long G.V., Li H., Waxman I.M., Jiang J., Robert C. Safety profile of nivolumab monotherapy: a pooled analysis of patients with advanced melanoma. J. Clin. Oncol. 2017;35(7):785–792. doi: 10.1200/JCO.2015.66.1389. [DOI] [PubMed] [Google Scholar]
- 14.Robert C., Schachter J., Long G.V., Arance A., Grob J.J., Mortier L., Daud A., Carlino M.S., McNeil C., Lotem M., Larkin J., Lorigan P., Neyns B., Blank C.U., Hamid O., Mateus C., Shapira-Frommer R., Kosh M., Zhou H., Ibrahim N., Ebbinghaus S., Ribas A. Pembrolizumab versus ipilimumab in advanced melanoma. N. Engl. J. Med. 2015;372(26):2521–2532. doi: 10.1056/NEJMoa1503093. [DOI] [PubMed] [Google Scholar]
- 15.Eggermont A.M., Chiarion-Sileni V., Grob J.J., Dummer R., Wolchok J.D., Schmidt H., Hamid O., Robert C., Ascierto P.A., Richards J.M., Lebbé C., Ferraresi V., Smylie M., Weber J.S., Maio M., Bastholt L., Mortier L., Thomas L., Tahir S., Hauschild A., Hassel J.C., Hodi F.S., Taitt C., de Pril V., de Schaetzen G., Suciu S., Testori A. Prolonged survival in stage iii melanoma with ipilimumab adjuvant therapy. N. Engl. J. Med. 2016;375(19):1845–1855. doi: 10.1056/NEJMoa1611299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Hodi F.S., O’Day S.J., McDermott D.F., Weber R.W., Sosman J.A., Haanen J.B., Gonzalez R., Robert C., Schadendorf D., Hassel J.C., Akerley W., van den Eertwegh A.J., Lutzky J., Lorigan P., Vaubel J.M., Linette G.P., Hogg D., Ottensmeier C.H., Lebbé C., Peschel C., Quirt I., Clark J.I., Wolchok J.D., Weber J.S., Tian J., Yellin M.J., Nichol G.M., Hoos A., Urba W.J. Improved survival with ipilimumab in patients with metastatic melanoma. N. Engl. J. Med. 2010;363(8):711–723. doi: 10.1056/NEJMoa1003466. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Cuzzubbo S., Javeri F., Tissier M., Roumi A., Barlog C., Doridam J., Lebbe C., Belin C., Ursu R., Carpentier A.F. Neurological adverse events associated with immune checkpoint inhibitors: Review of the literature. Eur. J. Cancer. 2017;73:1–8. doi: 10.1016/j.ejca.2016.12.001. [DOI] [PubMed] [Google Scholar]
- 18.Larkin J., Chmielowski B., Lao C.D., Hodi F.S., Sharfman W., Weber J., Suijkerbuijk K.P.M., Azevedo S., Li H., Reshef D., Avila A., Reardon D.A. Neurologic serious adverse events associated with nivolumab plus ipilimumab or nivolumab alone in advanced melanoma, including a case series of encephalitis. Oncologist. 2017;22(6):709–718. doi: 10.1634/theoncologist.2016-0487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Spain L., Walls G., Julve M., O’Meara K., Schmid T., Kalaitzaki E., Turajlic S., Gore M., Rees J., Larkin J. Neurotoxicity from immune-checkpoint inhibition in the treatment of melanoma: a single centre experience and review of the literature. Ann. Oncol. 2017;28(2):377–385. doi: 10.1093/annonc/mdw558. [DOI] [PubMed] [Google Scholar]
- 20.Voskens C.J., Goldinger S.M., Loquai C., Robert C., Kaehler K.C., Berking C., Bergmann T., Bockmeyer C.L., Eigentler T., Fluck M., Garbe C., Gutzmer R., Grabbe S., Hauschild A., Hein R., Hundorfean G., Justich A., Keller U., Klein C., Mateus C., Mohr P., Paetzold S., Satzger I., Schadendorf D., Schlaeppi M., Schuler G., Schuler-Thurner B., Trefzer U., Ulrich J., Vaubel J., von Moos R., Weder P., Wilhelm T., Göppner D., Dummer R., Heinzerling L.M. The price of tumor control: an analysis of rare side effects of anti-CTLA-4 therapy in metastatic melanoma from the ipilimumab network. PLoS One. 2013;8(1):e53745. doi: 10.1371/journal.pone.0053745. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Zimmer L., Goldinger S.M., Hofmann L., Loquai C., Ugurel S., Thomas I., Schmidgen M.I., Gutzmer R., Utikal J.S., Göppner D., Hassel J.C., Meier F., Tietze J.K., Forschner A., Weishaupt C., Leverkus M., Wahl R., Dietrich U., Garbe C., Kirchberger M.C., Eigentler T., Berking C., Gesierich A., Krackhardt A.M., Schadendorf D., Schuler G., Dummer R., Heinzerling L.M. Neurological, respiratory, musculoskeletal, cardiac and ocular side-effects of anti-PD-1 therapy. Eur. J. Cancer. 2016;60:210–225. doi: 10.1016/j.ejca.2016.02.024. [DOI] [PubMed] [Google Scholar]
- 22.Suzuki S., Ishikawa N., Konoeda F., Seki N., Fukushima S., Takahashi K., Uhara H., Hasegawa Y., Inomata S., Otani Y., Yokota K., Hirose T., Tanaka R., Suzuki N., Matsui M. Nivolumab-related myasthenia gravis with myositis and myocarditis in Japan. Neurology. 2017;89(11):1127–1134. doi: 10.1212/WNL.0000000000004359. [DOI] [PubMed] [Google Scholar]
- 23.Mandel J.J., Olar A., Aldape K.D., Tremont-Lukats I.W. Lambrolizumab induced central nervous system (CNS) toxicity. J. Neurol. Sci. 2014;344(1-2):229–231. doi: 10.1016/j.jns.2014.06.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Salam S., Lavin T., Turan A. Limbic encephalitis following immunotherapy against metastatic malignant melanoma. BMJ Case Rep. 2016;2016 doi: 10.1136/bcr-2016-215012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Schneider S., Potthast S., Komminoth P., Schwegler G., Böhm S. PD-1 Checkpoint Inhibitor Associated Autoimmune Encephalitis. Case Rep. Oncol. 2017;10(2):473–478. doi: 10.1159/000477162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Stein M.K., Summers B.B., Wong C.A., Box H.L., Cleveland K.O. Meningoencephalitis following ipilimumab administration in metastatic melanoma. Am. J. Med. Sci. 2015;350(6):512–513. doi: 10.1097/MAJ.0000000000000584. [DOI] [PubMed] [Google Scholar]
- 27.Galmiche S., Lheure C., Kramkimel N., Franck N., Boitier F., Dupin N., Turc G., Psimaras D., Aractingi S., Guégan S. Encephalitis induced by immune checkpoint inhibitors in metastatic melanoma: a monocentric retrospective study. J. Eur. Acad. Dermatol. Venereol. 2019;33(12):e440–e443. doi: 10.1111/jdv.15756. [DOI] [PubMed] [Google Scholar]
- 28.Richard K., Weslow J., Porcella S.L., Nanjappa S. A Case report of steroid responsive nivolumab-induced encephalitis. Cancer Contr. 2017;24(5):1073274817729069. doi: 10.1177/1073274817729069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Burke M., Hardesty M., Downs W. A case of severe encephalitis while on PD-1 immunotherapy for recurrent clear cell ovarian cancer. Gynecol. Oncol. Rep. 2018;24:51–53. doi: 10.1016/j.gore.2018.03.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Khoja L., Maurice C., Chappell M., MacMillan L., Al-Habeeb A.S., Al-Faraidy N., Butler M.O., Rogalla P., Mason W., Joshua A.M., Hogg D. Eosinophilic fasciitis and acute encephalopathy toxicity from pembrolizumab treatment of a patient with metastatic melanoma. Cancer Immunol. Res. 2016;4(3):175–178. doi: 10.1158/2326-6066.CIR-15-0186. [DOI] [PubMed] [Google Scholar]
- 31.Gettings E.J., Hackett C.T., Scott T.F. Severe relapse in a multiple sclerosis patient associated with ipilimumab treatment of melanoma. Mult. Scler. 2015;21(5):670. doi: 10.1177/1352458514549403. [DOI] [PubMed] [Google Scholar]
- 32.LaPorte J., Solh M., Ouanounou S. Posterior reversible encephalopathy syndrome following pembrolizumab therapy for relapsed Hodgkin’s lymphoma. J. Oncol. Pharm. Pract. 2017;23(1):71–74. doi: 10.1177/1078155215620922. [DOI] [PubMed] [Google Scholar]
- 33.Maur M., Tomasello C., Frassoldati A., Dieci M.V., Barbieri E., Conte P. Posterior reversible encephalopathy syndrome during ipilimumab therapy for malignant melanoma. J. Clin. Oncol. 2012;30(6):e76–e78. doi: 10.1200/JCO.2011.38.7886. [DOI] [PubMed] [Google Scholar]
- 34.Astaras C., de Micheli R., Moura B., Hundsberger T., Hottinger A.F. Neurological adverse events associated with immune checkpoint inhibitors: diagnosis and management. Curr. Neurol. Neurosci. Rep. 2018;18(1):3. doi: 10.1007/s11910-018-0810-1. [DOI] [PubMed] [Google Scholar]
- 35.Conry R.M., Sullivan J.C., Nabors L.B., III Ipilimumab-induced encephalopathy with a reversible splenial lesion. Cancer Immunol. Res. 2015;3(6):598–601. doi: 10.1158/2326-6066.CIR-15-0035. [DOI] [PubMed] [Google Scholar]
- 36.Williams T.J., Benavides D.R., Patrice K.A., Dalmau J.O., de Ávila A.L., Le D.T., Lipson E.J., Probasco J.C., Mowry E.M. Association of autoimmune encephalitis with combined immune checkpoint inhibitor treatment for metastatic cancer. JAMA Neurol. 2016;73(8):928–933. doi: 10.1001/jamaneurol.2016.1399. [DOI] [PubMed] [Google Scholar]
- 37.Choe J.H., Andonian B.J., Kim G.J., Salama A.K. Autoimmune meningoencephalitis in a melanoma patient treated with ipilimumab. Immunotherapy. 2016;8(10):1163–1167. doi: 10.2217/imt-2016-0058. [DOI] [PubMed] [Google Scholar]
- 38.Brown M.P., Hissaria P., Hsieh A.H., Kneebone C., Vallat W. Autoimmune limbic encephalitis with anti-contactin-associated protein-like 2 antibody secondary to pembrolizumab therapy. . J Neuroimmunol . 2017. [DOI] [PubMed]
- 39.Bossart S., Thurneysen S., Rushing E., Frontzek K., Leske H., Mihic-Probst D., Nagel H.W., Mangana J., Goldinger S.M., Dummer R. Case report: encephalitis, with brainstem involvement, following checkpoint inhibitor therapy in metastatic melanoma. Oncologist. 2017;22(6):749–753. doi: 10.1634/theoncologist.2016-0366. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Ito M., Fujiwara S., Fujimoto D., Mori R., Yoshimura H., Hata A., Kohara N., Tomii K. Rituximab for nivolumab plus ipilimumab-induced encephalitis in a small-cell lung cancer patient. Ann. Oncol. 2017;28(9):2318–2319. doi: 10.1093/annonc/mdx252. [DOI] [PubMed] [Google Scholar]
- 41.Strik H., Keber U., Hammoud W.A., Riera-Knorrenschild J., Carl B., Dodel R., Pagenstecher A., Neubauer A. Immune checkpoint inhibitor-associated CNS autoimmune disorder (ICICAD) following nivolumab treatment: A new entity of drug-induced autoimmune encephalitis? Eur. J. Cancer. 2017;87:205–208. doi: 10.1016/j.ejca.2017.09.026. [DOI] [PubMed] [Google Scholar]
- 42.Papadopoulos K.P., Romero R.S., Gonzalez G., Dix J.E., Lowy I., Fury M. Anti-Hu-associated autoimmune limbic encephalitis in a patient with pd-1 inhibitor-responsive myxoid chondrosarcoma. Oncologist. 2018;23(1):118–120. doi: 10.1634/theoncologist.2017-0344. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Chaucer B., Stone A., Demanes A., Seibert S.M. Nivolumab-induced encephalitis in hereditary leiomyomatosis and renal cell cancer syndrome. Case Rep. Oncol. Med. 2018;2018:4273231. doi: 10.1155/2018/4273231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Leitinger M., Varosanec M.V., Pikija S., Wass R.E., Bandke D., Weis S., Studnicka M., Grinzinger S., McCoy M.R., Hauer L., Sellner J. Fatal necrotizing encephalopathy after treatment with nivolumab for squamous non-small cell lung cancer: case report and review of the literature. Front. Immunol. 2018;9:108. doi: 10.3389/fimmu.2018.00108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Zurko J., Mehta A. Association of immune-mediated cerebellitis with immune checkpoint inhibitor therapy. Mayo Clin. Proc. Innov. Qual. Outcomes. 2018;2(1):74–77. doi: 10.1016/j.mayocpiqo.2017.12.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Shah S., Dunn-Pirio A., Luedke M., Morgenlander J., Skeen M., Eckstein C. Nivolumab-induced autoimmune encephalitis in two patients with lung adenocarcinoma. Case Rep. Neurol. Med. 2018;2018:2548528. doi: 10.1155/2018/2548528. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Matsuoka H., Kimura H., Koba H., Tambo Y., Ohkura N., Hara J., Sone T., Kasahara K. Nivolumab-induced limbic encephalitis with anti-hu antibody in a patient with advanced pleomorphic carcinoma of the lung. Clin. Lung Cancer. 2018;19(5):e597–e599. doi: 10.1016/j.cllc.2018.04.009. [DOI] [PubMed] [Google Scholar]
- 48.Kopecký J., Kubeček O., Geryk T., Slováčková B., Hoffmann P., Žiaran M., Priester P. Nivolumab induced encephalopathy in a man with metastatic renal cell cancer: a case report. J. Med. Case Reports. 2018;12(1):262. doi: 10.1186/s13256-018-1786-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.De la Hoz A., Foolad F., Gallegos C., Kornblau S., Kontoyiannis D.P. Nivolumab-induced encephalitis post allogeneic stem cell transplant in a patient with Hodgkin’s disease. Bone Marrow Transplant. 2019;54(5):749–751. doi: 10.1038/s41409-018-0363-6. [DOI] [PubMed] [Google Scholar]
- 50.Strominger M. Nivolumab Plus Ipilimumab Induced Aseptic Meningitis And Bilateral Sixth Nerve Palsy In Metastatic Melanoma. 2015.
- 51.Cordes L.M., Davarpanah N.N., Reoma L.B., Gasmi B., Quezado M., Khan O.I., Nath A., Apolo A.B. Neurotoxicities associated with checkpoint inhibitors: Two case reports and a review of the literature. Clin. Case Rep. 2019;8(1):24–32. doi: 10.1002/ccr3.2534. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Yang J.C., Hughes M., Kammula U., Royal R., Sherry R.M., Topalian S.L., Suri K.B., Levy C., Allen T., Mavroukakis S., Lowy I., White D.E., Rosenberg S.A. Ipilimumab (anti-CTLA4 antibody) causes regression of metastatic renal cell cancer associated with enteritis and hypophysitis. J. Immunother. 2007;30(8):825–830. doi: 10.1097/CJI.0b013e318156e47e. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Oishi K., Nakao M., Maeda S., Matsushita T., Ikeda T., Yamada T., Takehara K., Hamaguchi Y. A case of aseptic meningitis without neck rigidity occurring in a metastatic melanoma patient treated with ipilimumab. Eur. J. Dermatol. 2017;27(2):193–194. doi: 10.1684/ejd.2016.2943. [DOI] [PubMed] [Google Scholar]
- 54.Bot I., Blank C.U., Boogerd W., Brandsma D. Neurological immune-related adverse events of ipilimumab. Pract. Neurol. 2013;13(4):278–280. doi: 10.1136/practneurol-2012-000447. [DOI] [PubMed] [Google Scholar]
- 55.Corsello S.M., Barnabei A., Marchetti P., De Vecchis L., Salvatori R., Torino F. Endocrine side effects induced by immune checkpoint inhibitors. J. Clin. Endocrinol. Metab. 2013;98(4):1361–1375. doi: 10.1210/jc.2012-4075. [DOI] [PubMed] [Google Scholar]
- 56.Torino F., Barnabei A., Paragliola R.M., Marchetti P., Salvatori R., Corsello S.M. Endocrine side-effects of anti-cancer drugs: mAbs and pituitary dysfunction: clinical evidence and pathogenic hypotheses. Eur. J. Endocrinol. 2013;169(6):R153–R164. doi: 10.1530/EJE-13-0434. [DOI] [PubMed] [Google Scholar]
- 57.Guerrero E., Johnson D.B., Bachelot A., Lebrun-Vignes B., Moslehi J.J., Salem J-E. Immune checkpoint inhibitor-associated hypophysitis-world health organisation vigibase report analysis. Eur. J. Cancer. 2019;113:10–13. doi: 10.1016/j.ejca.2019.03.002. [DOI] [PubMed] [Google Scholar]
- 58.Ribas A., Camacho L.H., Lopez-Berestein G., Pavlov D., Bulanhagui C.A., Millham R., Comin-Anduix B., Reuben J.M., Seja E., Parker C.A., Sharma A., Glaspy J.A., Gomez-Navarro J. Antitumor activity in melanoma and anti-self responses in a phase I trial with the anti-cytotoxic T lymphocyte-associated antigen 4 monoclonal antibody CP-675,206. J. Clin. Oncol. 2005;23(35):8968–8977. doi: 10.1200/JCO.2005.01.109. [DOI] [PubMed] [Google Scholar]
- 59.Topalian S.L., Sznol M., McDermott D.F., Kluger H.M., Carvajal R.D., Sharfman W.H., Brahmer J.R., Lawrence D.P., Atkins M.B., Powderly J.D., Leming P.D., Lipson E.J., Puzanov I., Smith D.C., Taube J.M., Wigginton J.M., Kollia G.D., Gupta A., Pardoll D.M., Sosman J.A., Hodi F.S. Survival, durable tumor remission, and long-term safety in patients with advanced melanoma receiving nivolumab. J. Clin. Oncol. 2014;32(10):1020–1030. doi: 10.1200/JCO.2013.53.0105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Robert C., Ribas A., Wolchok J.D., Hodi F.S., Hamid O., Kefford R., Weber J.S., Joshua A.M., Hwu W.J., Gangadhar T.C., Patnaik A., Dronca R., Zarour H., Joseph R.W., Boasberg P., Chmielowski B., Mateus C., Postow M.A., Gergich K., Elassaiss-Schaap J., Li X.N., Iannone R., Ebbinghaus S.W., Kang S.P., Daud A. Anti-programmed-death-receptor-1 treatment with pembrolizumab in ipilimumab-refractory advanced melanoma: a randomised dose-comparison cohort of a phase 1 trial. Lancet. 2014;384(9948):1109–1117. doi: 10.1016/S0140-6736(14)60958-2. [DOI] [PubMed] [Google Scholar]
- 61.Barroso-Sousa R., Barry W.T., Garrido-Castro A.C., Hodi F.S., Min L., Krop I.E., Tolaney S.M. Incidence of endocrine dysfunction following the use of different immune checkpoint inhibitor regimens: a systematic review and meta-analysis. JAMA Oncol. 2018;4(2):173–182. doi: 10.1001/jamaoncol.2017.3064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Scott E.S., Long G.V., Guminski A., Clifton-Bligh R.J., Menzies A.M., Tsang V.H. The spectrum, incidence, kinetics and management of endocrinopathies with immune checkpoint inhibitors for metastatic melanoma. Eur. J. Endocrinol. 2018;178(2):173–180. doi: 10.1530/EJE-17-0810. [DOI] [PubMed] [Google Scholar]
- 63.Min L., Hodi F.S., Giobbie-Hurder A., Ott P.A., Luke J.J., Donahue H., Davis M., Carroll R.S., Kaiser U.B. Systemic high-dose corticosteroid treatment does not improve the outcome of ipilimumab-related hypophysitis: a retrospective cohort study. Clin. Cancer Res. 2015;21(4):749–755. doi: 10.1158/1078-0432.CCR-14-2353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Lam T., Chan M.M., Sweeting A.N., De Sousa S.M., Clements A., Carlino M.S., Long G.V., Tonks K., Chua E., Kefford R.F., Chipps D.R. Ipilimumab-induced hypophysitis in melanoma patients: an Australian case series. Intern. Med. J. 2015;45(10):1066–1073. doi: 10.1111/imj.12819. [DOI] [PubMed] [Google Scholar]
- 65.Faje A.T., Sullivan R., Lawrence D., Tritos N.A., Fadden R., Klibanski A., Nachtigall L. Ipilimumab-induced hypophysitis: a detailed longitudinal analysis in a large cohort of patients with metastatic melanoma. J. Clin. Endocrinol. Metab. 2014;99(11):4078–4085. doi: 10.1210/jc.2014-2306. [DOI] [PubMed] [Google Scholar]
- 66.Ascierto P.A., Del Vecchio M., Robert C., Mackiewicz A., Chiarion-Sileni V., Arance A., Lebbé C., Bastholt L., Hamid O., Rutkowski P., McNeil C., Garbe C., Loquai C., Dreno B., Thomas L., Grob J.J., Liszkay G., Nyakas M., Gutzmer R., Pikiel J., Grange F., Hoeller C., Ferraresi V., Smylie M., Schadendorf D., Mortier L., Svane I.M., Hennicken D., Qureshi A., Maio M. Ipilimumab 10 mg/kg versus ipilimumab 3 mg/kg in patients with unresectable or metastatic melanoma: a randomised, double-blind, multicentre, phase 3 trial. Lancet Oncol. 2017;18(5):611–622. doi: 10.1016/S1470-2045(17)30231-0. [DOI] [PubMed] [Google Scholar]
- 67.Maker A.V., Yang J.C., Sherry R.M., Topalian S.L., Kammula U.S., Royal R.E., Hughes M., Yellin M.J., Haworth L.R., Levy C., Allen T., Mavroukakis S.A., Attia P., Rosenberg S.A. Intrapatient dose escalation of anti-CTLA-4 antibody in patients with metastatic melanoma. J. Immunother. 2006;29(4):455–463. doi: 10.1097/01.cji.0000208259.73167.58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Wehbeh L., Alreddawi S., Salvatori R. Hypophysitis in the era of immune checkpoint inhibitors and immunoglobulin G4-related disease. Expert Rev. Endocrinol. Metab. 2019;14(3):167–178. doi: 10.1080/17446651.2019.1598260. [DOI] [PubMed] [Google Scholar]
- 69.Dillard T., Yedinak C.G., Alumkal J., Fleseriu M. Anti-CTLA-4 antibody therapy associated autoimmune hypophysitis: serious immune related adverse events across a spectrum of cancer subtypes. Pituitary. 2010;13(1):29–38. doi: 10.1007/s11102-009-0193-z. [DOI] [PubMed] [Google Scholar]
- 70.Blansfield J.A., Beck K.E., Tran K., Yang J.C., Hughes M.S., Kammula U.S., Royal R.E., Topalian S.L., Haworth L.R., Levy C., Rosenberg S.A., Sherry R.M. Cytotoxic T-lymphocyte-associated antigen-4 blockage can induce autoimmune hypophysitis in patients with metastatic melanoma and renal cancer. J. Immunother. 2005;28(6):593–598. doi: 10.1097/01.cji.0000178913.41256.06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Bertrand A., Kostine M., Barnetche T., Truchetet M.E., Schaeverbeke T. Immune related adverse events associated with anti-CTLA-4 antibodies: systematic review and meta-analysis. BMC Med. 2015;13(1):211. doi: 10.1186/s12916-015-0455-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.De Sousa S.M.C., Sheriff N., Tran C.H., Menzies A.M., Tsang V.H.M., Long G.V., Tonks K.T.T. Fall in thyroid stimulating hormone (TSH) may be an early marker of ipilimumab-induced hypophysitis. Pituitary. 2018;21(3):274–282. doi: 10.1007/s11102-018-0866-6. [DOI] [PubMed] [Google Scholar]
- 73.Albarel F., Gaudy C., Castinetti F., Carré T., Morange I., Conte-Devolx B., Grob J.J., Brue T. Long-term follow-up of ipilimumab-induced hypophysitis, a common adverse event of the anti-CTLA-4 antibody in melanoma. Eur. J. Endocrinol. 2015;172(2):195–204. doi: 10.1530/EJE-14-0845. [DOI] [PubMed] [Google Scholar]
- 74.Min L., Vaidya A., Becker C. Association of ipilimumab therapy for advanced melanoma with secondary adrenal insufficiency: a case series. Endocr. Pract. 2012;18(3):351–355. doi: 10.4158/EP11273.OR. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.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(6):E25–E26. doi: 10.1002/mus.25496. [DOI] [PubMed] [Google Scholar]
- 76.Chang E., Sabichi A.L., Sada Y.H. Myasthenia gravis after nivolumab therapy for squamous cell carcinoma of the bladder. J. Immunother. 2017;40(3):114–116. doi: 10.1097/CJI.0000000000000161. [DOI] [PubMed] [Google Scholar]
- 77.Johnson D.B., Saranga-Perry V., Lavin P.J., Burnette W.B., Clark S.W., Uskavitch D.R., Wallace D.E., Dickson M.A., Kudchadkar R.R., Sosman J.A. Myasthenia gravis induced by ipilimumab in patients with metastatic melanoma. J. Clin. Oncol. 2015;33(33):e122–e124. doi: 10.1200/JCO.2013.51.1683. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Kimura T., Fukushima S., Miyashita A., Aoi J., Jinnin M., Kosaka T., Ando Y., Matsukawa M., Inoue H., Kiyotani K., Park J.H., Nakamura Y., Ihn H. Myasthenic crisis and polymyositis induced by one dose of nivolumab. Cancer Sci. 2016;107(7):1055–1058. doi: 10.1111/cas.12961. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Liao B., Shroff S., Kamiya-Matsuoka C., Tummala S. Atypical neurological complications of ipilimumab therapy in patients with metastatic melanoma. Neuro-oncol. 2014;16(4):589–593. doi: 10.1093/neuonc/nou001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Loochtan A.I., Nickolich M.S., Hobson-Webb L.D. Myasthenia gravis associated with ipilimumab and nivolumab in the treatment of small cell lung cancer. Muscle Nerve. 2015;52(2):307–308. doi: 10.1002/mus.24648. [DOI] [PubMed] [Google Scholar]
- 81.March K.L., Samarin M.J., Sodhi A., Owens R.E. Pembrolizumab-induced myasthenia gravis: A fatal case report. J. Oncol. Pharm. Pract. 2018;24(2):146–149. doi: 10.1177/1078155216687389. [DOI] [PubMed] [Google Scholar]
- 82.Shirai T., Sano T., Kamijo F., Saito N., Miyake T., Kodaira M., Katoh N., Nishie K., Okuyama R., Uhara H. Acetylcholine receptor binding antibody-associated myasthenia gravis and rhabdomyolysis induced by nivolumab in a patient with melanoma. Jpn. J. Clin. Oncol. 2016;46(1):86–88. doi: 10.1093/jjco/hyv158. [DOI] [PubMed] [Google Scholar]
- 83.Makarious D., Horwood K., Coward J.I.G. Myasthenia gravis: An emerging toxicity of immune checkpoint inhibitors. Eur. J. Cancer. 2017;82:128–136. doi: 10.1016/j.ejca.2017.05.041. [DOI] [PubMed] [Google Scholar]
- 84.Takamatsu K., Nakane S., Suzuki S., Kosaka T., Fukushima S., Kimura T., Miyashita A., Mukaino A., Yamakawa S., Watanabe K., Jinnin M., Komohara Y., Ihn H., Ando Y. Immune checkpoint inhibitors in the onset of myasthenia gravis with hyperCKemia. Ann. Clin. Transl. Neurol. 2018;5(11):1421–1427. doi: 10.1002/acn3.654. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Psimaras D. Neuromuscular complications of immune checkpoint inhibitors. Presse Med. 2018;47(11-12 Pt 2):e253–e259. doi: 10.1016/j.lpm.2018.10.009. [DOI] [PubMed] [Google Scholar]
- 86.Chen J.H., Lee K.Y., Hu C.J., Chung C.C. 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(50):e9262. doi: 10.1097/MD.0000000000009262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Touat M., Maisonobe T., Knauss S., Ben Hadj Salem O., Hervier B., Auré K., Szwebel T.A., Kramkimel N., Lethrosne C., Bruch J.F., Laly P., Cadranel J., Weiss N., Béhin A., Allenbach Y., Benveniste O., Lenglet T., Psimaras D., Stenzel W., Léonard-Louis S. Immune checkpoint inhibitor-related myositis and myocarditis in patients with cancer. Neurology. 2018;91(10):e985–e994. doi: 10.1212/WNL.0000000000006124. [DOI] [PubMed] [Google Scholar]
- 88.Kao J.C., Brickshawana A., Liewluck T. Neuromuscular Complications of Programmed Cell Death-1 (PD-1) Inhibitors. Curr. Neurol. Neurosci. Rep. 2018;18(10):63. doi: 10.1007/s11910-018-0878-7. [DOI] [PubMed] [Google Scholar]
- 89.Mitsune A., Yanagisawa S., Fukuhara T., Miyauchi E., Morita M., Ono M., Tojo Y., Ichinose M. Relapsed myasthenia gravis after nivolumab treatment. Intern. Med. 2018;57(13):1893–1897. doi: 10.2169/internalmedicine.9153-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Gonzalez N.L., Puwanant A., Lu A., Marks S.M., Živković S.A. Myasthenia triggered by immune checkpoint inhibitors: New case and literature review. Neuromuscul. Disord. 2017;27(3):266–268. doi: 10.1016/j.nmd.2017.01.002. [DOI] [PubMed] [Google Scholar]
- 91.Seki M., Uruha A., Ohnuki Y., Kamada S., Noda T., Onda A., Ohira M., Isami A., Hiramatsu S., Hibino M., Nakane S., Noda S., Yutani S., Hanazono A., Yaguchi H., Takao M., Shiina T., Katsuno M., Nakahara J., Matsubara S., Nishino I., Suzuki S. Inflammatory myopathy associated with PD-1 inhibitors. J. Autoimmun. 2019;100:105–113. doi: 10.1016/j.jaut.2019.03.005. [DOI] [PubMed] [Google Scholar]
- 92.Liewluck T., Kao J.C., Mauermann M.L. PD-1 Inhibitor-associated myopathies: emerging immune-mediated myopathies. J. Immunother. 2018;41(4):208–211. doi: 10.1097/CJI.0000000000000196. [DOI] [PubMed] [Google Scholar]
- 93.Kadota H., Gono T., Shirai Y., Okazaki Y., Takeno M., Kuwana M. Immune checkpoint inhibitor-induced myositis: a case report and literature review. Curr. Rheumatol. Rep. 2019;21(4):10. doi: 10.1007/s11926-019-0811-3. [DOI] [PubMed] [Google Scholar]
- 94.Shah M., Tayar J.H., Abdel-Wahab N., Suarez-Almazor M.E. Myositis as an adverse event of immune checkpoint blockade for cancer therapy. Semin. Arthritis Rheum. 2019;48(4):736–740. doi: 10.1016/j.semarthrit.2018.05.006. [DOI] [PubMed] [Google Scholar]
- 95.Bilen M.A., Subudhi S.K., Gao J., Tannir N.M., Tu S.M., 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(1):36. doi: 10.1186/s40425-016-0139-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Moreira A., Loquai C., Pföhler C., Kähler K.C., Knauss S., Heppt M.V., Gutzmer R., Dimitriou F., Meier F., Mitzel-Rink H., Schuler G., Terheyden P., Thoms K.M., Türk M., Dummer R., Zimmer L., Schröder R., Heinzerling L. Myositis and neuromuscular side-effects induced by immune checkpoint inhibitors. Eur. J. Cancer. 2019;106:12–23. doi: 10.1016/j.ejca.2018.09.033. [DOI] [PubMed] [Google Scholar]
- 97.Haddox C.L., Shenoy N., Shah K.K., Kao J.C., Jain S., Halfdanarson T.R., Wijdicks E.F., Goetz M.P. Pembrolizumab induced bulbar myopathy and respiratory failure with necrotizing myositis of the diaphragm. Ann. Oncol. 2017;28(3):673–675. doi: 10.1093/annonc/mdw655. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Lecouflet M., Verschoore M., Giard C., Gohier P., Le Corre Y., Milea D., Martin L. Orbital myositis associated with ipilimumab. Ann. Dermatol. Venereol. 2013;140(6-7):448–451. doi: 10.1016/j.annder.2013.02.029. [DOI] [PubMed] [Google Scholar]
- 99.Vallet H., Gaillet A., Weiss N., Vanhaecke C., Saheb S., Touitou V., Franck N., Kramkimel N., Borden A., Touat M., Ricard D., Verny M., Maisonobe T., Psimaras D. Pembrolizumab-induced necrotic myositis in a patient with metastatic melanoma. Ann. Oncol. 2016;27(7):1352–1353. doi: 10.1093/annonc/mdw126. [DOI] [PubMed] [Google Scholar]
- 100.Pushkarevskaya A., Neuberger U., Dimitrakopoulou-Strauss A., Enk A., Hassel J.C. Severe ocular myositis after ipilimumab treatment for melanoma: a report of 2 cases. J. Immunother. 2017;40(7):282–285. doi: 10.1097/CJI.0000000000000178. [DOI] [PubMed] [Google Scholar]
- 101.Luo W., Wang Z., Tian P., Li W. Safety and tolerability of PD-1/PD-L1 inhibitors in the treatment of non-small cell lung cancer: a meta-analysis of randomized controlled trials. J. Cancer Res. Clin. Oncol. 2018;144(10):1851–1859. doi: 10.1007/s00432-018-2707-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Nishijima T.F., Shachar S.S., Nyrop K.A., Muss H.B. Safety and Tolerability of PD-1/PD-L1 Inhibitors compared with chemotherapy in patients with advanced cancer: A meta-analysis. Oncologist. 2017;22(4):470–479. doi: 10.1634/theoncologist.2016-0419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Altman A.L., Golub J.S., Pensak M.L., Samy R.N. Bilateral Facial palsy following ipilimumab infusion for melanoma. Otolaryngol. Head Neck Surg. 2015;153(5):894–895. doi: 10.1177/0194599815606701. [DOI] [PubMed] [Google Scholar]
- 104.Boisseau W., Touat M., Berzero G., Savatovsky J., Marabelle A., Touitou V., Ricard D., Malouf G., Psimaras D. Safety of treatment with nivolumab after ipilimumab-related meningoradiculitis and bilateral optic neuropathy. Eur. J. Cancer. 2017;83:28–31. doi: 10.1016/j.ejca.2017.05.036. [DOI] [PubMed] [Google Scholar]
- 105.Yeh O.L., Francis C.E. Ipilimumab-associated bilateral optic neuropathy. J. Neuroophthalmol. 2015;35(2):144–147. doi: 10.1097/WNO.0000000000000217. [DOI] [PubMed] [Google Scholar]
- 106.Bompaire F., Mateus C., Taillia H., De Greslan T., Lahutte M., Sallansonnet-Froment M., Ouologuem M., Renard J.L., Gorochov G., Robert C., Ricard D. Severe meningo-radiculo-neuritis associated with ipilimumab. Invest. New Drugs. 2012;30(6):2407–2410. doi: 10.1007/s10637-011-9787-1. [DOI] [PubMed] [Google Scholar]
- 107.Xu M., Nie Y., Yang Y., Lu Y.T., Su Q. Risk of neurological toxicities following the use of different immune checkpoint inhibitor regimens in solid tumors: a systematic review and meta-analysis. Neurologist. 2019;24(3):75–83. doi: 10.1097/NRL.0000000000000230. [DOI] [PubMed] [Google Scholar]
- 108.Man J., Ritchie G., Links M., Lord S., Lee C.K. Treatment-related toxicities of immune checkpoint inhibitors in advanced cancers: A meta-analysis. Asia Pac. J. Clin. Oncol. 2018;14(3):141–152. doi: 10.1111/ajco.12838. [DOI] [PubMed] [Google Scholar]
- 109.Johansen A., Christensen S.J., Scheie D., Højgaard J.L.S., Kondziella D. Neuromuscular adverse events associated with anti-PD-1 monoclonal antibodies: Systematic review. Neurology. 2019;92(14):663–674. doi: 10.1212/WNL.0000000000007235. [DOI] [PubMed] [Google Scholar]
- 110.Gu Y., Menzies A.M., Long G.V., Fernando S.L., Herkes G. Immune mediated neuropathy following checkpoint immunotherapy. J. Clin. Neurosci. 2017;45:14–17. doi: 10.1016/j.jocn.2017.07.014. [DOI] [PubMed] [Google Scholar]
- 111.Gaudy-Marqueste C., Monestier S., Franques J., Cantais E., Richard M.A., Grob J.J. A severe case of ipilimumab-induced guillain-barré syndrome revealed by an occlusive enteric neuropathy: a differential diagnosis for ipilimumab-induced colitis. J. Immunother. 2013;36(1):77–78. doi: 10.1097/CJI.0b013e31827807dd. [DOI] [PubMed] [Google Scholar]
- 112.Wilgenhof S., Neyns B. Anti-CTLA-4 antibody-induced Guillain-Barré syndrome in a melanoma patient. Ann. Oncol. 2011;22(4):991–993. doi: 10.1093/annonc/mdr028. [DOI] [PubMed] [Google Scholar]
- 113.Kao J.C., Liao B., Markovic S.N., Klein C.J., Naddaf E., Staff N.P., Liewluck T., Hammack J.E., Sandroni P., Finnes H., Mauermann M.L. Neurological complications associated with anti-programmed death 1 (PD-1) Antibodies. JAMA Neurol. 2017;74(10):1216–1222. doi: 10.1001/jamaneurol.2017.1912. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.de Maleissye M.F., Nicolas G., Saiag P. Pembrolizumab-induced demyelinating polyradiculoneuropathy. N. Engl. J. Med. 2016;375(3):296–297. doi: 10.1056/NEJMc1515584. [DOI] [PubMed] [Google Scholar]
- 115.Yost M.D., Chou C.Z., Botha H., Block M.S., Liewluck T. Facial diplegia after pembrolizumab treatment. Muscle Nerve. 2017;56(3):E20–E21. doi: 10.1002/mus.25663. [DOI] [PubMed] [Google Scholar]
- 116.Fukumoto Y., Kuwahara M., Kawai S., Nakahama K., Kusunoki S. Acute demyelinating polyneuropathy induced by nivolumab. J. Neurol. Neurosurg. Psychiatry. 2018;89(4):435–437. doi: 10.1136/jnnp-2017-316510. [DOI] [PubMed] [Google Scholar]
- 117.Supakornnumporn S., Katirji B. Guillain-Barré syndrome triggered by immune checkpoint inhibitors: a case report and literature review. J. Clin. Neuromuscul. Dis. 2017;19(2):80–83. doi: 10.1097/CND.0000000000000193. [DOI] [PubMed] [Google Scholar]
- 118.Nukui T., Nakayama Y., Yamamoto M., Taguchi Y., Dougu N., Konishi H., Hayashi T., Nakatsuji Y. Nivolumab-induced acute demyelinating polyradiculoneuropathy mimicking Guillain-Barré syndrome. J. Neurol. Sci. 2018;390:115–116. doi: 10.1016/j.jns.2018.04.028. [DOI] [PubMed] [Google Scholar]
- 119.Ong S., Chapman J., Young G., Mansy T. Guillain-Barré-like syndrome during pembrolizumab treatment. Muscle Nerve. 2018 doi: 10.1002/mus.26101. [DOI] [PubMed] [Google Scholar]
- 120.Jacob A., Unnikrishnan D.C., 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(8):1869–1870. doi: 10.1007/s00432-016-2191-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Schneiderbauer R., Schneiderbauer M., Wick W., Enk A.H., Haenssle H.A., Hassel J.C. PD-1 Antibody-induced Guillain-Barré Syndrome in a patient with metastatic melanoma. Acta Derm. Venereol. 2017;97(3):395–396. doi: 10.2340/00015555-2548. [DOI] [PubMed] [Google Scholar]
- 122.Ryota T., Hiroshi M., Yasushi T., Kumi Y., Takumi H., Akiko I., Mari O., Sae I., Ikuo S., Akira T.J.J.J.o.C.O. Nivolumab-induced chronic inflammatory demyelinating polyradiculoneuropathy mimicking rapid-onset Guillain–Barré syndrome: A case report. 2016. [DOI] [PubMed]
- 123.Tanaka R., Maruyama H., Tomidokoro Y., Yanagiha K., Hirabayashi T., Ishii A., Okune M., Inoue S., Sekine I., Tamaoka A., Fujimoto M. Nivolumab-induced chronic inflammatory demyelinating polyradiculoneuropathy mimicking rapid-onset Guillain-Barré syndrome: a case report. Jpn. J. Clin. Oncol. 2016;46(9):875–878. doi: 10.1093/jjco/hyw090. [DOI] [PubMed] [Google Scholar]
- 124.Sepúlveda M., Martinez-Hernandez E., Gaba L., Victoria I., Sola-Valls N., Falgàs N., Casanova-Molla J., Graus F. Motor polyradiculopathy during pembrolizumab treatment of metastatic melanoma. Muscle Nerve. 2017;56(6):E162–E167. doi: 10.1002/mus.25672. [DOI] [PubMed] [Google Scholar]
- 125.Aya F., Ruiz-Esquide V., Viladot M., Font C., Prieto-González S., Prat A., Arance A. Vasculitic neuropathy induced by pembrolizumab. Ann. Oncol. 2017;28(2):433–434. doi: 10.1093/annonc/mdw613. [DOI] [PubMed] [Google Scholar]
- 126.Fellner A., Makranz C., Lotem M., Bokstein F., Taliansky A., Rosenberg S., Blumenthal D.T., Mandel J., Fichman S., Kogan E., Steiner I., Siegal T., Lossos A., Yust-Katz S. Neurologic complications of immune checkpoint inhibitors. J. Neurooncol. 2018;137(3):601–609. doi: 10.1007/s11060-018-2752-5. [DOI] [PubMed] [Google Scholar]
- 127. Walsh; Ryan; Galetta; Steven; Tamhankar. Meeting, M.J.N.A. Ipilimumab-associated optic neuropathy . 2012.
- 128.Khan A.R., Hams E., Floudas A., Sparwasser T., Weaver C.T., Fallon P.G. PD-L1hi B cells are critical regulators of humoral immunity. Nat. Commun. 2015;6:5997. doi: 10.1038/ncomms6997. [DOI] [PubMed] [Google Scholar]
- 129.de Moel E.C., Rozeman E.A., Kapiteijn E.H., Verdegaal E.M.E., Grummels A., Bakker J.A., Huizinga T.W.J., Haanen J.B., Toes R.E.M., van der Woude D. Autoantibody development under treatment with immune-checkpoint inhibitors. Cancer Immunol. Res. 2019;7(1):6–11. doi: 10.1158/2326-6066.CIR-18-0245. [DOI] [PubMed] [Google Scholar]
- 130.Sage P.T., Paterson A.M., Lovitch S.B., Sharpe A.H. The coinhibitory receptor CTLA-4 controls B cell responses by modulating T follicular helper, T follicular regulatory, and T regulatory cells. Immunity. 2014;41(6):1026–1039. doi: 10.1016/j.immuni.2014.12.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Yshii L.M., Gebauer C.M., Pignolet B., Mauré E., Quériault C., Pierau M., Saito H., Suzuki N., Brunner-Weinzierl M., Bauer J., Liblau R. CTLA4 blockade elicits paraneoplastic neurological disease in a mouse model. Brain. 2016;139(11):2923–2934. doi: 10.1093/brain/aww225. [DOI] [PubMed] [Google Scholar]
- 132.Johnson D.B., Balko J.M., Compton M.L., Chalkias S., Gorham J., Xu Y., Hicks M., Puzanov I., Alexander M.R., Bloomer T.L., Becker J.R., Slosky D.A., Phillips E.J., Pilkinton M.A., Craig-Owens L., Kola N., Plautz G., Reshef D.S., Deutsch J.S., Deering R.P., Olenchock B.A., Lichtman A.H., Roden D.M., Seidman C.E., Koralnik I.J., Seidman J.G., Hoffman R.D., Taube J.M., Diaz L.A., Jr, Anders R.A., Sosman J.A., Moslehi J.J. Fulminant Myocarditis with Combination Immune Checkpoint Blockade. N. Engl. J. Med. 2016;375(18):1749–1755. doi: 10.1056/NEJMoa1609214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Sanderson K., Scotland R., Lee P., Liu D., Groshen S., Snively J., Sian S., Nichol G., Davis T., Keler T., Yellin M., Weber J. Autoimmunity in a phase I trial of a fully human anti-cytotoxic T-lymphocyte antigen-4 monoclonal antibody with multiple melanoma peptides and Montanide ISA 51 for patients with resected stages III and IV melanoma. J. Clin. Oncol. 2005;23(4):741–750. doi: 10.1200/JCO.2005.01.128. [DOI] [PubMed] [Google Scholar]
- 134.Liu S.M., Sutherland A.P., Zhang Z., Rainbow D.B., Quintana F.J., Paterson A.M., Sharpe A.H., Oukka M., Wicker L.S., Kuchroo V.K. Overexpression of the Ctla-4 isoform lacking exons 2 and 3 causes autoimmunity. J. Immunol. 2012;188(1):155–162. doi: 10.4049/jimmunol.1102042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Kroner A., Schwab N., Ip C.W., Ortler S., Göbel K., Nave K-A., Mäurer M., Martini R., Wiendl H. Accelerated course of experimental autoimmune encephalomyelitis in PD-1-deficient central nervous system myelin mutants. Am. J. Pathol. 2009;174(6):2290–2299. doi: 10.2353/ajpath.2009.081012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Rui Y., Honjo T., Chikuma S. Programmed cell death 1 inhibits inflammatory helper T-cell development through controlling the innate immune response. Proc. Natl. Acad. Sci. USA. 2013;110(40):16073–16078. doi: 10.1073/pnas.1315828110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Salama A.D., Chitnis T., Imitola J., Ansari M.J.I., Akiba H., Tushima F., Azuma M., Yagita H., Sayegh M.H., Khoury S.J. Critical role of the programmed death-1 (PD-1) pathway in regulation of experimental autoimmune encephalomyelitis. J. Exp. Med. 2003;198(1):71–78. doi: 10.1084/jem.20022119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Zhu J., Zou L., Zhu S., Mix E., Shi F., Wang H., Volkmann I., Winblad B., Schalling M., Ljunggren H. Cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) blockade enhances incidence and severity of experimental autoimmune neuritis in resistant mice. J. Neuroimmunol. 2001;115(1-2):111–117. doi: 10.1016/S0165-5728(01)00255-7. [DOI] [PubMed] [Google Scholar]
- 139.Wang H.B., Shi F.D., Li H., Chambers B.J., Link H., Ljunggren H.G. Anti-CTLA-4 antibody treatment triggers determinant spreading and enhances murine myasthenia gravis. J. Immunol. 2001;166(10):6430–6436. doi: 10.4049/jimmunol.166.10.6430. [DOI] [PubMed] [Google Scholar]
- 140.Reynolds K.L., Guidon A.C. Diagnosis and management of immune checkpoint inhibitor-associated neurologic toxicity: illustrative case and review of the literature. Oncologist. 2019;24(4):435–443. doi: 10.1634/theoncologist.2018-0359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Morrone A., Scarabello A., Sperduti I., Cota C., Donà M.G., Orsini D., Cristaudo A. Donovanosis in migrants: a clinical case series in an Italian dermatological hospital. J. Eur. Acad. Dermatol. Venereol. 2019;33(12):e438–e440. doi: 10.1111/jdv.15809. [DOI] [PubMed] [Google Scholar]
- 142.Gutzmer R., Koop A., Meier F., Hassel J.C., Terheyden P., Zimmer L., Heinzerling L., Ugurel S., Pföhler C., Gesierich A., Livingstone E., Satzger I., Kähler K.C. Programmed cell death protein-1 (PD-1) inhibitor therapy in patients with advanced melanoma and preexisting autoimmunity or ipilimumab-triggered autoimmunity. Eur. J. Cancer. 2017;75:24–32. doi: 10.1016/j.ejca.2016.12.038. [DOI] [PubMed] [Google Scholar]
- 143.Horvat T.Z., Adel N.G., Dang T.O., Momtaz P., Postow M.A., Callahan M.K., Carvajal R.D., Dickson M.A., D’Angelo S.P., Woo K.M., Panageas K.S., Wolchok J.D., Chapman P.B. Immune-Related Adverse Events, Need for Systemic immunosuppression, and effects on survival and time to treatment failure in patients with melanoma treated with ipilimumab at memorial sloan kettering cancer center. J. Clin. Oncol. 2015;33(28):3193–3198. doi: 10.1200/JCO.2015.60.8448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Monje M., Borniger J.C., D’Silva N.J., Deneen B., Dirks P.B., Fattahi F., Frenette P.S., Garzia L., Gutmann D.H., Hanahan D., Hervey-Jumper S.L., Hondermarck H., Hurov J.B., Kepecs A., Knox S.M., Lloyd A.C., Magnon C., Saloman J.L., Segal R.A., Sloan E.K., Sun X., Taylor M.D., Tracey K.J., Trotman L.C., Tuveson D.A., Wang T.C., White R.A., Winkler F. Roadmap for the emerging field of cancer neuroscience. Cell. 2020;181(2):219–222. doi: 10.1016/j.cell.2020.03.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Parker K.R., Migliorini D., Perkey E., Yost K.E., Bhaduri A., Bagga P., Haris M., Wilson N.E., Liu F., Gabunia K., Scholler J., Montine T.J., Bhoj V.G., Reddy R., Mohan S., Maillard I., Kriegstein A.R., June C.H., Chang H.Y., Posey A.D., Jr, Satpathy A.T. Single-cell analyses identify brain mural cells expressing cd19 as potential off-tumor targets for CAR-T Immunotherapies. Cell. 2020;183(1):126–142.e17. doi: 10.1016/j.cell.2020.08.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Phares T., Ramakrishna C., Parra G., Epstein A., Chen L., Atkinson R., Stohlman S., Bergmann C. Target-dependent b7-h1 regulation contributes to clearance of central nervous sysyem infection and dampens morbidity. . J. Immunol (Baltimore, Md. 2009. [DOI] [PMC free article] [PubMed]
- 147.Caturegli P., Di Dalmazi G., Lombardi M., Grosso F., Larman H.B., Larman T., Taverna G., Cosottini M., Lupi I. Hypophysitis Secondary to Cytotoxic T-Lymphocyte-associated protein 4 blockade: insights into pathogenesis from an autopsy series. Am. J. Pathol. 2016;186(12):3225–3235. doi: 10.1016/j.ajpath.2016.08.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Iwama S., De Remigis A., Callahan M.K., Slovin S.F., Wolchok J.D., Caturegli P. Pituitary expression of CTLA-4 mediates hypophysitis secondary to administration of CTLA-4 blocking antibody. Sci. Transl. Med. 2014;6(230):230ra45. doi: 10.1126/scitranslmed.3008002. [DOI] [PubMed] [Google Scholar]
- 149.Iwasaki A. Immune regulation of antibody access to neuronal tissues. Trends Mol. Med. 2017;23(3):227–245. doi: 10.1016/j.molmed.2017.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Pluim D., Ros W., van Bussel M.T.J., Brandsma D., Beijnen J.H., Schellens J.H.M. Enzyme linked immunosorbent assay for the quantification of nivolumab and pembrolizumab in human serum and cerebrospinal fluid. J. Pharm. Biomed. Anal. 2019;164:128–134. doi: 10.1016/j.jpba.2018.10.025. [DOI] [PubMed] [Google Scholar]
- 151.Guo H., Wang R., Wang D., Wang S., Zhou J., Chai Z., Yao S., Li J., Lu L., Liu Y., Xie C., Lu W. Deliver anti-PD-L1 into brain by p-hydroxybenzoic acid to enhance immunotherapeutic effect for glioblastoma. J. Control. Release. 2020;320:63–72. doi: 10.1016/j.jconrel.2020.01.005. [DOI] [PubMed] [Google Scholar]
- 152.Maurice C., Schneider R., Kiehl T-R., Bavi P., Roehrl M.H.A., Mason W.P., Hogg D. Subacute CNS demyelination after treatment with nivolumab for melanoma. Cancer Immunol. Res. 2015;3(12):1299–1302. doi: 10.1158/2326-6066.CIR-15-0141. [DOI] [PubMed] [Google Scholar]
- 153.Ransohoff R.M., Kivisäkk P., Kidd G. Three or more routes for leukocyte migration into the central nervous system. Nat. Rev. Immunol. 2003;3(7):569–581. doi: 10.1038/nri1130. [DOI] [PubMed] [Google Scholar]
- 154.Feng S., Coward J., McCaffrey E., Coucher J., Kalokerinos P., O’Byrne K. Pembrolizumab-induced encephalopathy: a review of neurological toxicities with immune checkpoint inhibitors. J. Thorac. Oncol. 2017;12(11):1626–1635. doi: 10.1016/j.jtho.2017.08.007. [DOI] [PubMed] [Google Scholar]
- 155.Freeman-Keller M., Kim Y., Cronin H., Richards A., Gibney G., Weber J.S. Nivolumab in resected and unresectable metastatic melanoma: characteristics of immune-related adverse events and association with outcomes. Clin. Cancer Res. 2016;22(4):886–894. doi: 10.1158/1078-0432.CCR-15-1136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Sato K., Akamatsu H., Murakami E., Sasaki S., Kanai K., Hayata A., Tokudome N., Akamatsu K., Koh Y., Ueda H., Nakanishi M., Yamamoto N. Correlation between immune-related adverse events and efficacy in non-small cell lung cancer treated with nivolumab. Lung Cancer. 2018;115:71–74. doi: 10.1016/j.lungcan.2017.11.019. [DOI] [PubMed] [Google Scholar]
- 157.Teraoka S., Fujimoto D., Morimoto T., Kawachi H., Ito M., Sato Y., Nagata K., Nakagawa A., Otsuka K., Uehara K., Imai Y., Ishida K., Fukuoka J., Tomii K. Early immune-related adverse events and association with outcome in advanced non-small cell lung cancer patients treated with nivolumab: a prospective cohort study. J. Thorac. Oncol. 2017;12(12):1798–1805. doi: 10.1016/j.jtho.2017.08.022. [DOI] [PubMed] [Google Scholar]

