Next Article in Journal
Association between Visceral Fat and Brain Structural Changes or Cognitive Function
Next Article in Special Issue
Intensive Care Unit-Acquired Weakness and Positioning-Related Peripheral Nerve Injuries in COVID-19: A Case Series of Three Patients and the Latest Literature Review
Previous Article in Journal
Differential Hemispheric Lateralization of Emotions and Related Display Behaviors: Emotion-Type Hypothesis
Previous Article in Special Issue
Renal Involvement in Hereditary Transthyretin Amyloidosis: An Italian Single-Centre Experience
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Presynaptic Paraneoplastic Disorders of the Neuromuscular Junction: An Update

by
Maria Pia Giannoccaro
1,2,
Patrizia Avoni
1,2 and
Rocco Liguori
1,2,*
1
Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, 40138 Bologna, Italy
2
IRCCS Istituto delle Scienze Neurologiche, UOC Clinica Neurologia, Ospedale Bellaria, Via Altura 3, 40139 Bologna, Italy
*
Author to whom correspondence should be addressed.
Submission received: 1 July 2021 / Revised: 26 July 2021 / Accepted: 2 August 2021 / Published: 3 August 2021
(This article belongs to the Special Issue Advanced Research in Neuromuscular Disorders)

Abstract

:
The neuromuscular junction (NMJ) is the target of a variety of immune-mediated disorders, usually classified as presynaptic and postsynaptic, according to the site of the antigenic target and consequently of the neuromuscular transmission alteration. Although less common than the classical autoimmune postsynaptic myasthenia gravis, presynaptic disorders are important to recognize due to the frequent association with cancer. Lambert Eaton myasthenic syndrome is due to a presynaptic failure to release acetylcholine, caused by antibodies to the presynaptic voltage-gated calcium channels. Acquired neuromyotonia is a condition characterized by nerve hyperexcitability often due to the presence of antibodies against proteins associated with voltage-gated potassium channels. This review will focus on the recent developments in the autoimmune presynaptic disorders of the NMJ.

1. Introduction

Paraneoplastic disorders are a group of diseases that develop as a consequence of an immune response against cancer. According to the classical model, the tumor cells express antigens which are shared by neuronal tissue that stimulates a T cell response directed against not only the tumor but also the nervous system. Many of these responses are accompanied also by the production of antibodies against intracellular antigens, defined as onconeural antibodies [1].
The NMJ is a highly specialized synapsis that connects the nerve with the muscle and is the target of several autoimmune disorders. On reaching the muscle, motor nerves lose their myelin sheath and branch into nerve endings forming synaptic boutons that contact the muscle surface. The gap between the presynaptic nerve terminal and the muscle, postsynaptic, membrane is called synaptic cleft and is filled with the synaptic basal lamina, a specialized extracellular matrix. The postsynaptic part of the NMJ is a specialized portion of the sarcolemma characterized by the presence of junctional folds [2].
The autoimmune disorders involving the NMJ are classified according to the site of the antigenic target and the consequent neurotransmission failure as presynaptic and postsynaptic. This review will focus on the recent developments in immune-mediated presynaptic disorders, including LEMS and acquired NMT.

2. The Neuromuscular Junction

The NMJ is a chemical synapsis, designed to transform the motor action potential into muscle fiber contraction [2]. The arrival of the motor action potential opens VGCCs located in the motor nerve terminal with consequent Ca2+ influx and exocytosis of the ACh stored in synaptic vesicles at the active zone of the presynaptic terminal. ACh binds to nicotinic AChR on the postsynaptic membrane. The AChR are ligand-gated cation channels that open when bound by two ACh, causing the influx of positively charged ions, mainly sodium, with the generation of an EPP. Besides ACh release induced by nerve activity, a motor nerve terminal spontaneously releases, at irregular intervals, single ACh quanta through exocytosis of single synaptic vesicles. The single ACh quantum leads to a postsynaptic miniature EPP [3].
In health, the EPP is usually sufficient to reach the threshold for opening voltage-gated sodium channels generating an action potential that propagates along the muscle fiber leading to contraction (Figure 1). The extent to which the EPP exceeds the threshold for the generation of an action potential is called the “safety factor” of neuromuscular transmission, and it allows the NMJ to continue functioning under different conditions. The EPP is short-lived since both AChRs and VGCCs close spontaneously while ACh disperses by diffusion or is hydrolyzed by the AChE. The opening of VGKCs at the presynaptic membrane restores the membrane potential and limits the opening of VGCCs [3].
Both LEMS and NMT affect the safety factor, although with different mechanisms. In LEMS, VGCCs antibodies decrease the safety margin by decreasing ACh release. In NMT, antibodies against VGKC compromise the safety factor by causing delayed repolarization of the axon after each action potential and prolongation of the depolarization of the muscle fiber membrane [4].

3. Lambert-Eaton Myasthenic Syndrome (LEMS)

LEMS is a neuromuscular autoimmune disorder associated with the presence of antibodies against the presynaptic VGCCs. The association with cancer was recognized since the first description of the disease in 1956 [5] and LEMS has been classically classified into paraneoplastic (SCLC-LEMS) and non-paraneoplastic (NT-LEMS) [6]. Since some clinical features overlap those of MG, many cases are misdiagnosed or diagnosed later in the course of the disease. Although rare, the clinical recognition of LEMS is relevant as more than half of patients have an associated tumor, usually, SCLC and early disease recognition and cancer investigations are fundamental to ensure a good prognosis.

3.1. Immunology and Pathophysiology

LEMS is caused by antibodies targeting presynaptic VGCCs [7], multi-subunit channels that allow the transmembrane influx of calcium in response to an action potential. In vitro these antibodies can induce cross-linking and internalization of their target [8,9], ultimately interfering with the calcium flux required for ACh release [10] (Figure 1), and the passive transfer of IgG from affected individuals to mice reproduce some of the clinical features of the disease, suggesting a pathogenic role of the antibodies [8,11]. In rats, active immunization with peptides derived from the extracellular region of the α1 subunit of the VGCC showed some LEMS-like features [12].
Studies on HEK293 cells and cerebellar granular cells showed that these antibodies reduce the Cav2.1 (P-type) currents with a compensatory increase of the expression of other VGCCs [13]. In cultured hippocampal neurons, these antibodies were shown to impair vesicular exocytosis from synaptic boutons [14].
In the majority of LEMS patients, the antibodies target the alpha1A subunit of P/Q type VGCCs [7], the most abundant at the presynaptic nerve terminal. P/Q- type VGCCs are also expressed at autonomic synapses explaining the presence of autonomic symptoms [15] as well as on the cerebellum. More rarely, patients harbor antibodies against the N-type and L-type VGCCs [13,16,17], although their significance and clinical utility are still unclear, and they are usually associated with the presence of P/Q type antibodies [17,18,19]. About 10% of patients with LEMS have concomitant PCD [20,21], associated with the reduction of the P/Q type VGCCs in the molecular layer of the cerebellum [21] at neuropathology, suggestive of a pathogenic role of the P/Q type VGCC antibodies. Recently, GRP78 antibodies were detected in 83.3% of patients with PCD-LEMS compared to only 6.6% of patients with LEMS. Functional studies on human brain microvascular endothelial cells suggested that these autoantibodies could increase the blood-brain barrier permeability and facilitate cerebellar dysfunction in patients with PCD-LEMS [22].
Autoantibodies to other proteins have been described in patients with LEMS, such as synaptotagmin and muscarinic acetylcholine receptors m1, in both seropositive and seronegative LEMS [23,24,25,26].
About 10–15% of patients with LEMS are seronegative, although their clinical phenotype is very similar to that of seropositive cases [20]. A relevant difference is an association with SCLC, seen in about 70% of VGCC positive LEMS versus 12% of seronegative [20]. Passive transfer of seronegative LEMS sera to mice generate the same phenotype and electrophysiological changes observed in mice treated with antibody-positive sera [20], suggesting that seronegative LEMS might be due to VGCC antibodies but at a relatively lower titer, or antibodies directed against VGCCs’ epitopes not recognized by current assays [6].
In more than half of LEMS patients, the etiology is paraneoplastic. SCLC cells have been shown to express functional VGCCs on their surface membranes [27] and antibodies from LEMS patients can inhibit the function of these channels in a dose-dependent manner [28].
Differently from patients with cancer, the trigger of NT-LEMS is unknown. The HLA-DR3-B8 is present in around 69% of patients with NT-LEMS compared to 23% of the control group, and 12% of paraneoplastic LEMS [29,30]. This haplotype is linked to autoimmunity, and it has been also suggested that it could improve the immunosurveillance against SCLC [30]. The underlying genetic predisposition to autoimmunity in patients with NT-LEMS is also supported by the association with other autoimmune disorders such as thyroid disorders and type 1 diabetes [31,32] and with the presence of a variety of organ-specific antibodies [33].

3.2. Clinical Features and Diagnosis

LEMS is a rare disorder with a prevalence between 2.3 and 3.8 per million population [34,35,36], mainly affecting middle-aged adults, although rare cases have been reported also in children [37]. Initial reports described a male prevalence [38,39], but this is true only for the SCLC-LEMS, which affect mainly men with a mean age of 60 years [40]. The age of onset of NT-LEMS has a bimodal distribution, with a peak age of onset of around 35 years and a second peak at age 60 years [40]. NT-LEMS has a similar frequency in men and women although a female predominance has been observed in patients below the age of 45 years, and a male predominance in those diagnosed after the age of 60 years [31].
Clinically, LEMS is characterized by proximal muscle weakness, dysautonomia, and reduced or absent deep tendon reflexes, due to a reduced release of ACh from the motor nerve terminal. Characteristically, muscle strength, as well as deep tendon reflexes, can increase transiently after a forced muscle contraction, differentiating LEMS from MG. This feature, called post-exercise facilitation, can also be observed after high-frequency nerve stimulation [41]. Indeed, LEMS is characterized by reduced evoked endplate potential amplitude with normal miniature endplate potential amplitude, in accordance with a significant reduction in ACh release and normal postsynaptic sensitivity to ACh (Figure 1). However, HFRS can overcome the calcium channel blockade [41,42]. This effect is believed to be caused by calcium accumulation at the presynaptic terminals, leading to an increased probability of ACh release (Figure 1).
Muscle weakness involves mainly the lower extremities and usually progresses slowly over time. However, a subacute or acute presentation may occur, and progression tends to be faster in patients with cancer [43,44,45]. Weakness normally spreads proximally to distally, and caudally to cranially. Cranial muscle involvement has been reported in 30–65% of LEMS patients. Ptosis and diplopia are the most common manifestations [46,47,48], but oropharyngeal symptoms have also been observed [47,48]. Mild autonomic dysfunction is very common (80–96% of patients) and includes dry mouth, impotence, constipation, and orthostatic hypotension [38,45,49].
Diagnosis is based on clinical features, neurophysiology, and antibody testing. Due to the frequent mildness and the lack of specificity of initial symptoms, diagnostic delay, as well as misdiagnosis, are frequent. The median time to diagnosis is 4 months in SCLC-LEMS and 12–19 months in NT-LEMS [40,50] and a study showed that up to 58% of patients were initially misdiagnosed [40,45].
RNS is the neurophysiological study of choice for the diagnosis, and it should be performed in at least two muscles [51]. RNS usually shows reduced CMAP amplitudes at rest, which decrements further at low frequencies of repetitive stimulation (2 to 4 Hz). A decrease of CMAP amplitude (decrement) of at least 10% is considered abnormal [51]. The decremental pattern during low-frequency RNS might help distinguish between MG and LEMS. Indeed, in MG, a train of 8–10 stimuli produces a progressive decrement in CMAP response, which is usually maximal at the fourth or fifth stimulation while it increases, although remaining below the size of the initial response, with subsequent stimuli. On the contrary, in LEMS, a low-frequency RNS produces a progressively decremental response [52,53]. Moreover, immediately after brief exercise (or with HFRS at 25 Hz to 50 Hz), CMAP amplitude increases significantly (increment) in LEMS but not in MG, helping in the differential diagnosis between MG and LEMS. Post-exercise and high-frequency stimulation have similar sensitivity of 84–96% [54,55,56] but the first is preferable as it is less painful, although high-frequency stimulation could have a slightly higher diagnostic yield [54]. An increment higher than 100% is considered 100% specific for LEMS, but a cut-off of 60% has been proposed to improve sensitivity to 97%, while maintaining a 99% specificity [54].
VGCCs antibodies can be detected by radioimmunoprecipitation. P/Q-type and N-type VGCC are extracted from human or mammalian cerebellum that is labeled with 1251-ω-Conotoxin MVIIC or GVIA derived from the Conus genus of piscivorous snails [10,16]. P/Q type VGCC antibodies have been reported to be very sensitive and specific for the diagnosis of LEMS [16,17], although low titers could be found also in controls, in patients with various types of cancers as well as in patients with amyotrophic lateral sclerosis [16,57]. However, a recent study observed the presence of these antibodies in a high proportion of patients without a neurological autoimmune or inflammatory etiology, with no difference in antibody titers between the autoimmune and the not-autoimmune groups [58], confirming that the P/Q type VGCC antibodies are not diagnostic in the absence of clinical and electrophysiologic features of LEMS. No correlation has been found between antibody titers and clinical features or outcome, although antibody levels might decrease after immunosuppressive treatment [59,60].

3.3. Cancer Association

About 60% of LEMS are paraneoplastic, usually associated with SCLC [38] expressing P/Q type VGCCs [61]. Other malignancies observed in patients with LEMS include lymphoproliferative disorders [37,60,62,63,64], prostate carcinoma [44], Merkel cell carcinoma [65], different thymic cancers [66,67], neuroblastoma [37] and atypical carcinoid [68]. Although the co-occurrence could be casual, the association with some of these tumors, including lymphoproliferative disorders, prostate cancer, and MCC, suggests causality. LEMS has been observed in patients with lymphomas and leukemias [37,60,62,63,64]. In these cases, LEMS symptoms could precede or follow the cancer diagnosis. In patients with prostate cancer, the tumor had neuroendocrine and small cell characteristics, and LEMS symptoms correlated with cancer activity [44]. MCC is an aggressive neuroendocrine tumor classified as an extrapulmonary SCLC, from which is almost histologically indistinguishable [69]. Eight cases of MCC-associated LEMS have been reported so far [65,69,70]. Most cases improved with oncological treatment and immunotherapy.
VGCC-antibody positivity does not distinguish patients with paraneoplastic LEMS however, the presence of other antibodies can help to predict the presence of cancer. In particular, SOX antibodies have been reported in 64% of patients with SCLC [71]. Moreover, N-type VGCC and GABAB receptor antibodies are associated with a higher risk of SCLC-LEMS [72]. A clinical score, the DELTA-P score, based on demographic and clinical features, can help to identify patients at high risk for SCLC who require more extensive screening. A score between 3 and 6 is associated with a probability of SCLC of 83.9–100% [40]. The presence of additional neuronal antibodies could warrant a more intensive SCLC screening in patients with DELTA-P scores of 1 or 2, which are associated with a low risk of SLCL [72]. All patients with a novel diagnosis of LEMS and no history of cancer should undergo an initial oncologic screening with a CT scan of the chest, abdomen, and pelvis. If negative, CT should be followed by 18F-PET. In case of negativity, surveillance should be continued at 3 or 6-month intervals for at least 2 years, based on the DELTA-P score and additional risk factors [40,72].
Rarely, LEMS has been reported as an adverse event of cancer treatment in patients receiving immune checkpoint inhibitors therapy [73,74,75,76,77]. These are a group of monoclonal antibodies directed against immune cell receptors expressed on T cells or their ligand, which aim at boosting the anti-tumor activity of cytotoxic T cells by interfering with inhibiting signals that reduce their activity [78]. The main targets are CTLA-4 and the PD-1 or its ligand, PD-L1. Immune-related adverse events (irAE) have been observed with every class of ICI and are related to the promotion of cellular- and antibody-mediated autoimmunity, although the exact underlying pathophysiological mechanisms remains only partially understood and several factors may play a role [79]. LEMS has been reported after treatment with different ICI (nivolumab, pembrolizumab, and ipilimumab) for different cancers, including SCLC [73,75,77], squamous cell lung carcinoma [74], and neuroendocrine tumor [76]. Response to immunotherapy, 3,4-diaminopyridine (3,4-DAP), and ICI withdrawal were variable. Diagnosis and treatments for ICI-associated LEMS may be challenging, particularly in patients with SCLC. However, in these cases, differentiating between irAE and paraneoplastic etiology is relevant as treatment can differ and affect the cancer outcome. Indeed, high-dose steroids are considered the first-line treatment for most neuromuscular irAEs [80,81]. Although contrasting results have been reported so far [82,83,84], steroids might negatively affect cancer outcomes, at least in patients with SCLC [83]. Moreover, patients with irAE should suspend ICI therapy in severe cases and consider subsequent re-challenge, whereas patients with SCLC-LEMS can continue the treatment. There are not, at present, biomarkers that help to identify patients with irAE vs paraneoplastic LEMS. The time of onset could be relevant in helping the differential diagnosis, since in SCLC-LEMS symptoms usually precede the cancer diagnosis [85], whereas the median time of irAE onset is 19 weeks from ICI treatment initiation [86]. With the expanding approved indication of ICI treatment, more cases of irAE-LEMS will likely be reported in the future, helping the identification of clues to their correct identification and management.

3.4. Treatment and Prognosis

LEMS treatment is based on symptomatic therapy and immunosuppression. In patients with cancer, oncologic treatment is of the highest priority. Cancer treatment usually leads to a clinical improvement, although, in cases with residual symptoms, additional treatment might bring further benefit. The treatment choice is based on the clinical severity of the disease. Recently, a new measure, the Triple Timed-Up-and-Go test, has been established and validated for assessing clinical function and the outcome in patients with LEMS that could be used to guide treatment [87,88].
Independently from the tumor presence, patients with moderate or severe weakness interfering with function, should be started on symptomatic treatment. The treatment of choice is amifampridine (3,4-DAP), which acts by blocking the potassium channels Kv3.3, prolonging the presynaptic nerve terminal membrane depolarization, therefore enhancing the calcium entry, and improving the ACh release (Figure 2).
More recently, a further mechanism of action, a direct agonistic effect of 3,4-DAP on Cav1 type channels, has been reported, although its relevance at therapeutic concentrations is debated [89,90,91]. Several randomized controlled trials demonstrated the superiority of 3,4-DAP compared to placebo in improving motor and autonomic symptoms [92,93,94,95,96,97,98,99], and 3,4-DAP use in LEMS has been approved in 2009 in Europe and 2018 in the US. The starting dose is 15 to 30 mg daily in adults and 7.5 to 15 mg in children, divided into 3-4 doses. The maximum approved daily dose is 80 mg in adults and 50 mg in children. The most common side effects are oral (40%) and digital (34%) paresthesia, followed by headache, nausea, and diarrhea (9.4%) [97]. High doses of 3,4-DAP have been associated with seizures and Q-T interval prolongation [92,100] and its use is contraindicated in patients with epilepsy [100]. In patients with residual or refractory weakness, immunosuppressive therapy should be introduced. Oral prednisone, alone or in combination with AZA, has been shown to be effective in a retrospective study [101]. However, IVIG is suggested as first-line treatment [102] in patients with LEMS, based on case reports [103,104] and a placebo-controlled crossover trial showing significant improvement in limb strength after treatment [59]. PLEX has been reported as LEMS treatment in case reports/series but there are not clinical trials [101,105,106]. Rituximab is suggested in patients refractory to other treatments [107,108].
Survival is shorter in patients with SCLC-LEMS and depends on the disease stage at presentation. On the other hand, LEMS is associated with longer survival in patients with SCLC [109,110,111,112] with a median of 17.3 months in these patients versus 10 in those with SCLC without neurologic symptoms [111]. Given the rarity of LEMS in association with tumors other than SCLC, there are no data on the prognosis of this group of paraneoplastic patients. In patients with NT-LEMS, life expectancy appears comparable to that of the general population [112]. Overall, functional impairment tends to be more severe in patients with paraneoplastic LEMS, although a recent study showed that the health-related quality of life does not differ significantly between patients with SCLC- and NT-LEMS [112]. A follow-up study on 47 patients with LEMS showed that 88% had an improvement of muscle strength whereas a sustained clinical remission was achieved in 43%. About a quarter of patients needed a wheelchair, at least outside the house, and more rarely, at all times [60]. In a more recent study, 62% were independent for daily living activities at diagnosis, and 85% at the 1-year follow-up. During the disease course, 73% of patients used any device to assist mobility; 52% used a wheelchair, but only 6% were fully wheelchair dependent [112].

4. Neuromyotonia (NMT)

Peripheral nerve hyperexcitability includes a spectrum of clinical manifestations ranging from cramp fasciculation syndrome to NMT, characterized by involuntary muscle activity caused by spontaneous discharges originating from the motor nerve fibers [113]. Neuromyotonia or Isaacs’ syndrome was firstly reported in 1961 by Dr Hyam Isaacs, who described a ‘syndrome of continuous muscle-fiber activity’ characterized by progressive muscle stiffness, with widespread fasciculation leading to weakness [114]. The term neuromyotonia was coined in 1965 by Mertens and Zschocke to underline the myotonia-like spasm and the origin from the peripheral nerve [115]. NMT can be hereditary or acquired. Here we will focus on the autoimmune form.

4.1. Immunology and Pathophysiology

Indirect evidence for an autoimmune etiology of NMT came from the demonstration that passive transfer of sera from affected patients into mice produced a raised threshold to d-tubocurarine in the diaphragm muscle and that PLEX significantly reduced the number of neuromyotonic discharges in patients [116,117]. In 1995, Shillito P et al. [118] developed a radioimmunoprecipitation assay using Kv1 type VGKCs extracted by the mammalian brain and labeled with iodinated alpha-dendrotoxin (125I-αDTX), a neurotoxin derived from the venom of the Dendroaspis angusticeps (Green mamba snake) which binds specifically to the Kv1.1, 1.2 and 1.6 subtypes of the Shaker channel family [119]. The authors found that three of six patients studied precipitated 125I-αDTX counts that were significantly higher than controls levels [118], indicating the presence of antibodies directed against the VGKC or related proteins. These channels functions in terminating presynaptic action potentials, therefore a reduction of their function induced by the antibodies, as shown by in vitro studies [120,121], would increase Ca2+ influx and impair action potential repolarization, leading to excessive release of AChR and NMT (Figure 3).
VGKCs antibodies were later found in a 76-year-old man with MoS [122] and shortly after in a few patients with LE [123,124,125]. Subsequently, it was discovered that the VGKCs-antibodies are not directed against the potassium channel but to associated proteins, including LGI1, CASPR2, and contactin-2 [126]. CASPR2 antibodies do not cause internalization of their target [127,128] but the passive transfer of CASPR2 immunoglobulin to mice produced nerve hyperexcitability and mechanical allodynia with loss of surface CASPR2 and VGKC Kv1 in the dorsal root ganglia [129]. LGI1 animal models did not explore the peripheral effects of the antibodies, however, both in vitro and in vivo studies showed that LGI1 antibodies inhibited the interaction between LGI1 and ADAM22/23 and reversibly reduced synaptic AMPA receptor clusters in hippocampal neurons [130,131].
The frequency of VGKC-complex antibodies in patients with NMT and related disorders ranged from 2 to 40% of cases [132,133,134,135]. In a more recent study including only NMT cases, 45% (17/38) of patients had specific antibodies (13% to CASPR2, 13% to contactin 2 (in one case associated with CASPR2), 5% to LGI1, and 16% to both CASPR2 and LGI1 [136]. Patients with LGI1-CASPR2 co-reactivity had a higher disability as detected by modified Rankin Scale, pain, dysautonomia, and sleep disorders, and thymoma [136], associations confirmed in another study [137].
About 30% of patients with CASPR2 or LGI1 have other autoimmune disorders, including MG and thyroid disorders [138], in accordance with the presence of strong HLA associations in patients developing LGI1 and/or CASPR2 antibodies [138,139]. Further observations suggested that the HLA haplotype could be relevant in determining the associated clinical phenotype and the underlying pathogenetic mechanisms in CASPR2 autoimmunity. Indeed, the HLA-DRB1*11:01, initially observed in 48% of patients with CASPR2 antibodies [138], was recently shown to be present in 93% of LE genotyped cases, but at the same rates as controls in patients with a prevalent PNH phenotype [137].
Rarely, other antibodies have been observed in patients with NMT, including collapsing response-mediator protein-5 [140], glutamic acid decarboxylase [141], muscle-specific tyrosine kinase [142], and immunoglobulin-like cell adhesion molecule 5 [143] antibodies, although their role is unclear.
Overall, about half of the patients with NMT are seronegative [133,136], suggesting the presence of other possible antibodies in these cases yet to be discovered.
Figure 3. Pathophysiology of neuromyotonia. (A) Schematic overview of the VGKC complex in the juxtaparanodal region of myelinated axons (modified from [144]). CASPR2, together with contactin-2 and other proteins, is fundamental for clustering Kv1 channels. CASPR2 antibodies could interfere with CASPR2-contactin-2 interaction leading to Kv expression down-regulation with consequent impairment of action potential repolarisation and excessive or prolonged release of acetylcholine. (B) This leads to prolonged EPP and repetitive nerve firing. The EMG shows myokymic discharges (doublets, triplets, and multiplets).
Figure 3. Pathophysiology of neuromyotonia. (A) Schematic overview of the VGKC complex in the juxtaparanodal region of myelinated axons (modified from [144]). CASPR2, together with contactin-2 and other proteins, is fundamental for clustering Kv1 channels. CASPR2 antibodies could interfere with CASPR2-contactin-2 interaction leading to Kv expression down-regulation with consequent impairment of action potential repolarisation and excessive or prolonged release of acetylcholine. (B) This leads to prolonged EPP and repetitive nerve firing. The EMG shows myokymic discharges (doublets, triplets, and multiplets).
Brainsci 11 01035 g003

4.2. Clinical Features and Diagnosis

NMT is a rare disorder characterized by spontaneous and continuous muscle activity at rest, which manifests clinically as visible muscle twitching (90%), cramps (70%), myopathy, stiffness, hypertrophy, and impaired muscle relaxation [113]. The twitches consist of myokymia, the irregular undulation of muscle fibers, resembling worms moving beneath the skin. Pseudomyotonia manifests as delayed relaxation after maximal voluntary contraction, typically elicited during a tight handgrip or eye closure [145]. Fasciculations can also be present. NMT usually starts insidiously, over months, mainly in the extremities, although the facial and truncal muscles can be involved as well [113]. The continuous muscle fiber activity is considered the cause of the hypertrophic appearance of the muscles in some patients. Muscle stiffness can be relevant, causing walking impairment, hyperlordosis, and respiratory difficulty [113,146]. Other frequently associated features include sensory symptoms such as paresthesia and pain and autonomic disturbances including hyperhidrosis, tachycardia, and diarrhea [136]. Sleep disorders, particularly insomnia, are not infrequent [136]. In its extreme form, NMT associates with signs of the central nervous system in the MoS characterized by the association of neuromyotonia with encephalopathy, sleep disorders (mostly insomnia), and dysautonomia [122].
Although epidemiological data are scarce, acquired NMT, is mainly observed in adult male patients, in the mid-40s [133,147,148]. However, few pediatric cases, associated with LGI1/CASPR2 antibodies have been reported [149,150]. Differently from adult cases, a tumor association has not been found in children.
The diagnosis of NMT is based on electrophysiological studies showing fibrillation and fasciculation potentials, myokymia, and neuromyotonic discharges, characterized by repetitive bursts of potential with short interpotential intervals (Figure 3b). Repetitive afterdischarges can be seen after the CMAP of nerve conduction studies [113]. The persistence of the abnormal nerve activity after peripheral nerve block, along with its disappearance after curare, suggests it originates from the distal motor axon or nerve terminal [114], although a proximal origin of discharges has been recorded as well [151].
Acquired neuromyotonia can occur as an idiopathic or paraneoplastic autoimmune disorder. About 25% of patients with acquired NMT have an associated tumor [133,136], supporting the need for an accurate malignancy screening in these cases. CASPR2 antibodies, in particular, are associated with thymoma in 20% of cases [152]. Rarer is the association with SLCL and lymphoma [133]. Recently, antibodies to the Netrin-1 receptors deleted in colon cancer and uncoordinated-5A, have been associated with the presence of thymoma in patients with CASPR2 antibodies, PNH, and MG [137,153,154]. The expression of Netrin-1 receptors and CASPR2 was confirmed in thymoma samples, similar to what had previously been observed for other antigens involved in paraneoplastic autoimmunity (i.e., titin) [155].

4.3. Treatment and Prognosis

In patients with an underlying tumor, oncologic treatment might induce an improvement of symptoms [117]. There are no approved symptomatic treatments for NMT. Frequently employed drugs are antiepileptics, particularly sodium-channel antagonists such as carbamazepine, sodium valproate, and phenytoin [136]. Other drugs, employed with variable success, include acetazolamide, gabapentin, lamotrigine, clonazepam, and mexiletine [145,156,157,158]. PLEX is associated with clinical and electrophysiological improvement as well as reduction of antibody titers [117]. IVIGs, steroids, AZA, methotrexate, and more recently, rituximab have been proposed as alternatives treatments [159,160].
Hypersensitivity to muscle relaxants has been suggested in patients with NMT, requiring caution during general anesthesia, and possibly lower doses of muscle relaxants, particularly when using non-depolarizing drugs [161].

5. Conclusions

LEMS and NMT are rare disorders, and the clinician’s suspicion is pivotal to pursue the proper diagnostic test and achieve a correct diagnosis. Both disorders remain under-recognized and diagnostic delays are frequent. Once the diagnosis has been established, oncological screening and appropriate treatment are fundamental for patients’ prognosis and quality of life. Although vast improvements in treatment and care have been made, further studies are needed to standardize immune therapy and improve the long-term outcome.

Author Contributions

Conceptualization, M.P.G. and R.L.; data curation, M.P.G. and P.A.; writing—original draft preparation, M.P.G.; writing—review and editing, M.P.G., P.A. and R.L.; visualization, M.P.G. and P.A.; supervision, R.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

M.P.G. and P.A. report no conflicts of interest in this work. R.L. reports personal fees from Argenx, Biogen, Sanofi-Genzyme, Argon Healthcare s.r.l., Amicus Therapeutics s.r.l. and Alfasigma for Advisory Board consultancy and Lecture fees from Dynamicom Education, SIMG Service, Adnkronos Salute Unipersonale s.r.l. and DOC Congress s.r.l., outside the submitted work.

Abbreviations

3:4-DAP, 3,4-diaminopyridine; 18F-PET, 18F-fluorodeoxyglucose-positron emission tomography; ACh, Achetilcholine; AChE, ACh esterase; AchR, ACh receptors; ADAM22/23, ADAM Metallopeptidase Domain 22/23; AMPA, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid; AZA, azathioprine; CASPR2, contactin-associated protein 2; CMAP, compound muscle action potential; CT, computed tomography; CTLA-4, cytotoxic T-lymphocyte antigen 4; DELTA-P, Dutch-English LEMS Tumor Association Prediction score; EPP, endplate potential; GABA(B), gamma-aminobutyric acid; GRP78, 78-kDa glucose-regulated protein; HEK293, Human embryonic kidney 293 cells; HFRS, high frequency repetitive stimulation; HLA, human leucocyte antigen; ICI, immune checkpoint inhibitors; irAE, immune-related adverse events; IVIG, intravenous immunoglobulin; LE, limbic encephalitis; LEMS, Lambert Eaton myasthenic syndrome; LGI1, leucine-rich glioma inactivated protein 1; MCC, Merkel cell carcinoma; MG, myasthenia gravis; MoS, Morvan’s syndrome; NMJ, neuromuscular junction; NMT, neuromyotonia; NT-LEMS, non-tumour LEMS; PCD, paraneoplastic cerebellar degeneration; PLEX, plasma exchange; PNH, peripheral nerve hyperexcitability; PD-1, programmed cell death-1; PD-1L, programmed cell death-1 ligand; RNS, repetitive nerve stimulation; SCLC, small-cell lung carcinoma; SCLC-LEMS, paraneoplastic LEMS; SOX, SRY-related high mobility group [HMG] box; VGCC, voltage gated calcium channels; VGKCs, voltage-gated potassium channels.

References

  1. Flanagan, E.P. Paraneoplastic disorders of the nervous system. J. Neurol. 2021. [Google Scholar] [CrossRef] [PubMed]
  2. Tintignac, L.A.; Brenner, H.R.; Rüegg, M.A. Mechanisms Regulating Neuromuscular Junction Development and Function and Causes of Muscle Wasting. Physiol. Rev. 2015, 95, 809–852. [Google Scholar] [CrossRef] [Green Version]
  3. Plomp, J.J. Neuromuscular Junction: Physiology and Pathophysiology. In Myasthenia Gravis and Related Disorders; Kaminski, H.J., Kusner, L.L., Eds.; Current Clinical Neurology; Humana Press: Cham, Switzerland, 2018. [Google Scholar] [CrossRef]
  4. Wood, S.J.; Slater, C.R. Safety factor at the neuromuscular junction. Prog. Neurobiol. 2001, 64, 393–429. [Google Scholar] [CrossRef]
  5. Lambert, E.H.; Eaton, L.M.; Rooke, E.D. Defect of neuromuscular conduction associated with malignant neoplasms. Am. J. Physiol. 1956, 187, 612–613. [Google Scholar]
  6. Titulaer, M.J.; Lang, B.; Verschuuren, J.J. Lambert–Eaton myasthenic syndrome: From clinical characteristics to therapeutic strategies. Lancet Neurol. 2011, 10, 1098–1107. [Google Scholar] [CrossRef]
  7. Lennon, V.A.; Lambert, E.H. Autoantibodies bind solubilized calcium channel-omega-conotoxin complexes from small cell lung carcinoma: A diagnostic aid for Lambert-Eaton myasthenic syndrome. Mayo Clin. Proc. 1989, 64, 1498–1504. [Google Scholar] [CrossRef]
  8. Lang, B.; Newsom-Davis, J.; Peers, C.; Prior, C.; Wray, D.W. The effect of myasthenic syndrome antibody on presynaptic calcium channels in the mouse. J. Physiol. 1987, 390, 257–270. [Google Scholar] [CrossRef] [PubMed]
  9. Nagel, A.; Engel, A.G.; Lang, B.; Newsom-Davis, J.; Fukuoka, T. Lambert–Eaton myasthenic syndrome IgG depletes presynaptic membrane active zone particles by antigenic modulation. Ann. Neurol. 1988, 24, 552–558. [Google Scholar] [CrossRef]
  10. Motomura, M.; Johnston, I.; Lang, B.; Vincent, A.; Newsom-Davis, J. An improved diagnostic assay for Lambert-Eaton myasthenic syndrome. J. Neurol. Neurosurg. Psychiatry 1995, 58, 85–87. [Google Scholar] [CrossRef]
  11. Lang, B.; Newsom-Davis, J.; Wray, D.; Vincent, A.; Murray, N. Autoimmune aetiology for myasthenic (Eaton–Lambert) syndrome. Lancet 1981, 2, 224–226. [Google Scholar] [CrossRef]
  12. Komai, K.; Iwasa, K.; Takamori, M. Calcium channel peptide can cause an autoimmune-mediated model of Lambert-Eaton myasthenic syndrome in rats. J. Neurol. Sci. 1999, 166, 126–130. [Google Scholar] [CrossRef]
  13. Pinto, A.; Iwasa, K.; Newland, C.; Newsom-Davis, J.; Lang, B. The action of Lambert–Eaton myasthenic syndrome immunoglobulin G on cloned human voltage-gated calcium channels. Muscle Nerve 2012, 25, 715–724. [Google Scholar] [CrossRef] [PubMed]
  14. Spillane, J.; Ermolyuk, Y.; Cano-Jaimez, M.; Lang, B.; Vincent, A.; Volynski, K.E.; Kullmann, D.M. Lambert-Eaton syndrome IgG inhibits transmitter release via P/Q Ca2+ channels. Neurology 2015, 84, 575–579. [Google Scholar] [CrossRef] [Green Version]
  15. Waterman, S.A.; Lang, B.; Newsom-Davis, J. Effect of Lambert-Eaton myasthenic syndrome antibodies on autonomic neurons in the mouse. Ann. Neurol. 1997, 42, 147–156. [Google Scholar] [CrossRef]
  16. Lennon, V.A.; Kryzer, T.J.; Griesmann, G.E.; O’Suilleabhain, P.E.; Windebank, A.J.; Woppmann, A.; Miljanich, G.P.; Lambert, E.H. Calcium-channel antibodies in the Lambert-Eaton syndrome and other paraneoplastic syndromes. N. Engl. J. Med. 1995, 332, 1467–1474. [Google Scholar] [CrossRef]
  17. Motomura, M.; Lang, B.; Johnston, I.; Palace, J.; Vincent, A.; Newsom-Davis, J. Incidence of serum anti-P/O-type and anti-N-type calcium channel autoantibodies in the Lambert-Eaton myasthenic syndrome. J. Neurol. Sci. 1997, 147, 35–42. [Google Scholar] [CrossRef]
  18. Johnston, I.; Lang, B.; Leys, K.; Newsom-Davis, J. Heterogeneity of calcium channel autoantibodies detected using a small-cell lung cancer line derived from a Lambert-Eaton myasthenic syndrome patient. Neurology 1994, 44, 334–338. [Google Scholar] [CrossRef]
  19. Martin-Moutot, N.; De, H.L.; Seagar, M. Distinct evolution of calcium channel antibody types in Lambert-Eaton myasthenic syndrome. J. Neuroimmunol. 2008, 197, 47–53. [Google Scholar] [CrossRef]
  20. Nakao, Y.K.; Motomura, M.; Fukudome, T.; Fukuda, T.; Shiraishi, H.; Yoshimura, T.; Tsujihata, M.; Eguchi, K. Seronegative Lambert-Eaton myasthenic syndrome: Study of 110 Japanese patients. Neurology 2002, 59, 1773–1775. [Google Scholar] [CrossRef] [PubMed]
  21. Fukuda, T.; Motomura, M.; Nakao, Y.; Shiraishi, H.; Yoshimura, T.; Iwanaga, K.; Tsujihata, M.; Dosaka-Akita, H.; Eguchi, K. Reduction of P/Q-type calcium channels in the postmortem cerebellum of paraneoplastic cerebellar degeneration with Lambert-Eaton myasthenic syndrome. Ann. Neurol. 2003, 53, 21–28. [Google Scholar] [CrossRef]
  22. Shimizu, F.; Takeshita, Y.; Sano, Y.; Hamamoto, Y.; Shiraishi, H.; Sato, T.; Yoshimura, S.; Maeda, T.; Fujikawa, S.; Nishihara, H.; et al. GRP78 antibodies damage the blood-brain barrier and relate to cerebellar degeneration in Lambert-Eaton myasthenic syndrome. Brain 2019, 142, 2253–2264. [Google Scholar] [CrossRef] [PubMed]
  23. Takamori, M.; Takahashi, M.; Yasukawa, Y.; Iwasa, K.; Nemoto, Y.; Suenaga, A.; Nagataki, S.; Nakamura, T. Antibodies to recombinant synaptotagmin and calcium channel subtypes in Lambert-Eaton myasthenic syndrome. J. Neurol. Sci. 1995, 133, 95–101. [Google Scholar] [CrossRef]
  24. el Far, O.; Marqueze, B.; Leveque, C.; Martin-Moutôt, N.; Lang, B.; Newsom-Davis, J.; Yoshida, A.; Takahashi, M.; Seagar, M.J. Antigens associated with N- and L-type calcium channels in Lambert-Eaton myasthenic syndrome. J. Neurochem. 1995, 64, 1696–1702. [Google Scholar] [CrossRef] [PubMed]
  25. Takamori, M.; Hamada, T.; Komai, K.; Takahashi, M.; Yoshida, A. Synaptotagmin can cause an immune-mediated model of Lambert-Eaton myasthenic syndrome in rats. Ann. Neurol. 1994, 35, 74–80. [Google Scholar] [CrossRef]
  26. Takamori, M.; Motomura, M.; Fukudome, T.; Yoshikawa, H. Autoantibodies against M1 muscarinic acetylcholine receptor in myasthenic disorders. Eur. J. Neurol. 2007, 14, 1230–1235. [Google Scholar] [CrossRef]
  27. Benatar, M.; Blaes, F.; Johnston, I.; Wilson, K.; Vincent, A.; Beeson, D.; Lang, B. Presynaptic neuronal antigens expressed by a small cell lung carcinoma cell line. J. Neuroimmunol. 2001, 113, 153–162. [Google Scholar] [CrossRef]
  28. Lang, B.; Vincent, A.; Murray, N.M.; Newsom-Davis, J. Lambert-Eaton myasthenic syndrome: Immunoglobulin G inhibition of Ca2+ flux in tumor cells correlates with disease severity. Ann. Neurol. 1989, 25, 265–271. [Google Scholar] [CrossRef]
  29. Wirtz, P.W.; Roep, B.O.; Schreuder, G.M.T.; van Doorn, P.A.; van Engelen, G.M.; Kuks, J.B.; Twijnstra, A.; De Visser, M.; Visser, L.H.; Wokke, J.H.; et al. HLA class I and II in Lambert-Eaton myasthenic syndrome without associated tumor. Hum. Immunol. 2001, 62, 809–813. [Google Scholar] [CrossRef]
  30. Wirtz, P.W.; Lang, B.; Graus, F.; van den Maagdenberg, A.M.; Saiz, A.; de Koning Gans, P.A.; Twijnstra, A.; Verschuuren, J.J. P/Q-type calcium channel antibodies, Lambert-Eaton myasthenic syndrome and survival in small cell lung cancer. J. Neuroimmunol. 2005, 164, 161–165. [Google Scholar] [CrossRef]
  31. Wirtz, P.W.; Smallegange, T.M.; Wintzen, A.R.; Verschuuren, J.J. Differences in clinical features between the Lambert-Eaton myasthenic syndrome with and without cancer: An analysis of 227 published cases. Clin. Neurol. Neurosurg. 2002, 104, 359–363. [Google Scholar] [CrossRef]
  32. Wirtz, P.W.; Bradshaw, J.; Wintzen, A.R.; Verschuuren, J.J. Associated autoimmune diseases in patients with the Lambert-Eaton myasthenic syndrome and their families. J. Neurol. 2004, 251, 1255–1259. [Google Scholar] [CrossRef] [PubMed]
  33. Lennon, V.A.; Lambert, E.H.; Whittingham, S.; Fairbanks, V. Autoimmunity in the Lambert-Eaton myasthenic syndrome. Muscle Nerve 1982, 5, S21–S25. [Google Scholar] [PubMed]
  34. Wirtz, P.W.; Nijnuis, M.G.; Sotodeh, M.; Willems, L.N.; Brahim, J.J.; Putter, H.; Wintzen, A.R.; Verschuuren, J.J. Dutch Myasthenia Study Group. The epidemiology of myasthenia gravis, Lambert-Eaton myasthenic syndrome and their associated tumours in the northern part of the province of South Holland. J. Neurol. 2003, 250, 698–701. [Google Scholar] [CrossRef]
  35. Wirtz, P.W.; van Dijk, J.G.; van Doorn, P.A.; Van Engelen, B.G.M.; Van Der Kooi, A.J.; Kuks, J.B.; Twijnstra, A.; de Visser, M.; Visser, L.H.; Wokke, J.H.; et al. The epidemiology of the Lambert–Eaton myasthenic syndrome in the Netherlands. Neurology 2004, 63, 397–398. [Google Scholar] [CrossRef] [PubMed]
  36. Abenroth, D.C.; Smith, A.G.; Greenlee, J.E.; Austin, S.D.; Clardy, S.L. Lambert–Eaton myasthenic syndrome (LEMS): Epidemiology and therapeutic response in the national Veterans Affairs (VA) population. Muscle Nerve 2017, 56, 421–426. [Google Scholar] [CrossRef]
  37. Hajjar, M.; Markowitz, J.; Darras, B.T.; Kissel, J.T.; Srinivasan, J.; Jones, H.R. Lambert-Eaton syndrome, an unrecognized treatable pediatric neuromuscular disorder: Three patients and literature review. Pediatr. Neurol. 2014, 50, 11–17. [Google Scholar] [CrossRef]
  38. O’Neill, J.H.; Murray, N.M.F.; Newsom-Davis, J. The Lambert-Eaton myasthenic syndrome—A review of 50 cases. Brain 1988, 111, 577–596. [Google Scholar] [CrossRef]
  39. Sanders, D.B. Lambert-Eaton myasthenic syndrome: Clinical diagnosis, immune-mediated mechanisms, and update on therapies. Ann. Neurol. 1995, 37 (Suppl. 1), 63–73. [Google Scholar] [CrossRef]
  40. Titulaer, M.J.; Maddison, P.; Sont, J.K.; Wirtz, P.W.; Hilton-Jones, D.; Klooster, R.; Willcox, N.; Potman, M.; Smitt, P.A.S.; Kuks, J.B.; et al. Clinical Dutch-English Lambert-Eaton myasthenic syndrome (LEMS) tumor association prediction score accurately predicts small-cell lung cancer in the LEMS. J. Clin. Oncol. 2011, 29, 902–908. [Google Scholar] [CrossRef]
  41. Lambert, E.H.; Elmqvist, D. Quantal components of end-plate potentials in the myasthenic syndrome. Ann. N. Y. Acad. Sci. 1971, 183, 183–199. [Google Scholar] [CrossRef]
  42. Elmqvist, D.; Lambert, E.H. Detailed analysis of neuromuscular transmission in a patient with the myasthenic syndrome sometimes associated with bronchogenic carcinoma. Mayo Clin. Proc. 1968, 43, 689–713. [Google Scholar]
  43. Wirtz, P.W.; Wintzen, A.R.; Verschuuren, J.J. Lambert-Eaton myasthenic syndrome has a more progressive course in patients with lung cancer. Muscle Nerve 2005, 32, 226–229. [Google Scholar] [CrossRef]
  44. Titulaer, M.J.; Verschuuren, J.J. Lambert-Eaton myasthenic syndrome: Tumor versus nontumor forms. Ann. N. Y. Acad. Sci. 2008, 1132, 129–134. [Google Scholar] [CrossRef] [PubMed]
  45. Titulaer, M.J.; Wirtz, P.W.; Kuks, J.B.; Schelhaas, H.J.; van der Kooi, A.J.; Faber, C.G.; van der Pol, W.L.; de Visser, M.; Sillevis Smitt, P.A.; Verschuuren, J.J. The Lambert-Eaton myasthenic syndrome 1988–2008: A clinical picture in 97 patients. J. Neuroimmunol. 2008, 201–202, 153–158. [Google Scholar] [CrossRef] [PubMed]
  46. Young, J.D.; Leavitt, J.A. Lambert–Eaton Myasthenic Syndrome. J. Neuro-Ophthalmol. 2016, 36, 20–22. [Google Scholar] [CrossRef] [PubMed]
  47. Burns, T.M.; Russell, J.A.; LaChance, D.H.; Jones, H.R. Oculobulbar involvement is typical with Lambert-Eaton myasthenic syndrome. Ann. Neurol. 2003, 53, 270–273. [Google Scholar] [CrossRef] [PubMed]
  48. Wirtz, P.W.; Sotodeh, M.; Nijnuis, M.; Van Doorn, P.A.; Van Engelen, B.G.; Hintzen, R.Q.; De Kort, P.L.; Kuks, J.B.; Twijnstra, A.; De Visser, M.; et al. Difference in distribution of muscle weakness between myasthenia gravis and the Lambert-Eaton myasthenic syndrome. J. Neurol. Neurosurg. Psychiatry 2002, 73, 766–768. [Google Scholar] [CrossRef] [Green Version]
  49. O’Suilleabhain, P.; Low, P.A.; Lennon, V.A. Autonomic dysfunction in the Lambert-Eaton myasthenic syndrome: Serologic and clinical correlates. Neurology 1998, 50, 88–93. [Google Scholar] [CrossRef]
  50. Pellkofer, H.L.; Armbruster, L.; Linke, R.; Schumm, F.; Voltz, R. Managing non-paraneoplastic Lambert-Eaton myasthenic syndrome: Clinical characteristics in 25 German patients. J. Neuroimmunol. 2009, 217, 90–94. [Google Scholar] [CrossRef]
  51. AAEM Quality Assurance Committee. Practice parameter for repetitive nerve stimulation and single fiber EMG evaluation of adults with suspected myasthenia gravis or Lambert-Eaton myasthenic syndrome: Summary statement. Muscle Nerve 2001, 24, 1236–1238. [Google Scholar] [CrossRef]
  52. Baslo, M.B.; Deymeer, F.; Serdaroglu, P.; Parman, Y.; Ozdemir, C.; Cuttini, M. Decrement pattern in Lambert-Eaton myasthenic syndrome is different from myasthenia gravis. Neuromuscul. Disord. 2006, 16, 454–458. [Google Scholar] [CrossRef]
  53. Sanders, D.B.; Cao, L.; Massey, J.M.; Juel, V.C.; Hobson-Webb, L.; Guptill, J.T. Is the decremental pattern in Lambert-Eaton syndrome different from that in myasthenia gravis? Clin. Neurophysiol. 2014, 125, 1274–1277. [Google Scholar] [CrossRef]
  54. Oh, S.J.; Kurokawa, K.; Claussen, G.C.; Ryan, H.F., Jr. Electrophysiological diagnostic criteria of Lambert-Eaton myasthenic syndrome. Muscle Nerve 2005, 32, 515–520. [Google Scholar] [CrossRef]
  55. Tim, R.W.; Massey, J.M.; Sanders, D.B. Lambert-Eaton myasthenic syndrome (LEMS)—Clinical and electrodiagnostic features and response to therapy in 59 patients. Ann. N. Y. Acad. Sci. 1998, 841, 823–826. [Google Scholar] [CrossRef]
  56. Hatanaka, Y.; Oh, S.J. Ten-second exercise is superior to 30-second exercise for post-exercise facilitation in diagnosing Lambert-Eaton myasthenic syndrome. Muscle Nerve 2008, 37, 572–575. [Google Scholar] [CrossRef]
  57. Zalewski, N.L.; Lennon, V.A.; Lachance, D.H.; Klein, C.J.; Pittock, S.J.; Mckeon, A. P/Q- and N-type calcium-channel antibodies: Oncological, neurological, and serological accompaniments. Muscle Nerve 2016, 54, 220–227. [Google Scholar] [CrossRef] [PubMed]
  58. Di Lorenzo, R.; Mente, K.; Li, J.; Shayya, L.; Rae-Grant, A.; Li, Y.; Jammoul, A. Low specificity of voltage-gated calcium channel antibodies in Lambert-Eaton myasthenic syndrome: A call for caution. J. Neurol. 2018, 265, 2114–2119. [Google Scholar] [CrossRef]
  59. Bain, P.G.; Motomura, M.; Newsom-Davis, J.; Misbah, S.A.; Chapel, H.M.; Lee, M.L.; Vincent, A.; Lang, B. Effects of intravenous immunoglobulin on muscle weakness and calcium-channel autoantibodies in the Lambert-Eaton myasthenic syndrome. Neurology 1996, 47, 678–683. [Google Scholar] [CrossRef]
  60. Maddison, P.; Lang, B.; Mills, K.; Newsom-Davis, J. Long term outcome in Lambert-Eaton myasthenic syndrome without lung cancer. J. Neurol. Neurosurg. Psychiatry 2001, 70, 212–217. [Google Scholar] [CrossRef] [Green Version]
  61. Meriney, S.D.; Hulsizer, S.C.; Lennon, V.A.; Grinnell, A.D. Lambert–Eaton myasthenic syndrome immunoglobulins react with multiple types of calcium channels in small-cell lung carcinoma. Ann. Neurol. 1996, 40, 739–749. [Google Scholar] [CrossRef]
  62. Argov, Z.; Shapira, Y.; Averbuch-Heller, L.; Wirguin, I. Lambert-Eaton myasthenic syndrome (LEMS) in association with lymphoproliferative disorders. Muscle Nerve 1995, 18, 715–719. [Google Scholar] [CrossRef]
  63. Tyagi, A.; Connolly, S.; Hutchinson, M. Lambert-Eaton myaesthenic syndrome: A possible association with Hodgkin’s lymphoma. Ir. Med. J. 2001, 94, 18–19. [Google Scholar]
  64. Zambelis, T.; Foutsitzi, A.; Giannakopoulou, A.; Poulopoulou, K.; Karandreas, N. Lambert-Eaton myasthenic syndrome. Clinical and electrophysiological findings in seven cases. Electromyogr. Clin. Neurophysiol. 2004, 44, 289–292. [Google Scholar]
  65. Pavolucci, L.; Giannini, G.; Giannoccaro, M.P.; Foschini, M.P.; Lang, B.; Avoni, P.; Tinuper, P.; Vincent, A.; Liguori, R. Paraneoplastic cerebellar degeneration and lambert-eaton myasthenia in a patient with merkel cell carcinoma and voltage-gated calcium channel antibodies. Muscle Nerve 2017, 56, 998–1000. [Google Scholar] [CrossRef]
  66. Hashimoto, S.; Hayasaka, K.; Suzuki, K.; Endoh, M.; Yanagawa, N.; Shiono, S. Thymic Small Cell Carcinoma Associated With Lambert-Eaton Myasthenic Syndrome. Ann. Thorac. Surg. 2020, 109, e347–e348. [Google Scholar] [CrossRef]
  67. Nalbantoglu, M.; Kose, L.; Uzun, N.; Gunduz, A.; Hallac, M.; Kiziltan, M.E.; Akalin, M.A. Lambert-Eaton myasthenic syndrome associated with thymic neuroendocrine carcinoma. Muscle Nerve 2015, 51, 936–938. [Google Scholar] [CrossRef] [PubMed]
  68. Gutmann, L.; Phillips, L.H., 2nd; Gutmann, L. Trends in the association of Lambert-Eaton myasthenic syndrome with carcinoma. Neurology 1992, 42, 848–850. [Google Scholar] [CrossRef]
  69. Nguyen, N.D.; Simmons, D.B.; Bersabe, A.R.; Duginski, T.M.; Sladky, J.H.; Walton, D.; Will, M.; Renshaw, J.S. Lambert-Eaton myasthenic syndrome and merkel cell carcinoma. Cutis 2019, 103, E19–E23. [Google Scholar]
  70. Agostini, A.; Merli, M.; Avallone, G.; Burzi, L.; Mastorino, L.; Parisi, M.; Bertuzzo, D.; Ferrero, B.; Cerrato, M.; Badellino, S.; et al. Lambert-Eaton Myasthenic Syndrome and Paraneoplastic Cerebellar Degeneration Associated with Merkel Cell Carcinoma with Unknown Primary: A Case Report. Acta Derm-Venereol. 2021, 101, adv00452. [Google Scholar] [CrossRef] [PubMed]
  71. Sabater, L.; Titulaer, M.; Saiz, A.; Verschuuren, J.J.G.M.; Güre, A.O.; Graus, F. SOX1 antibodies are markers of paraneoplastic Lambert-Eaton myasthenic syndrome. Neurology 2008, 70, 924–928. [Google Scholar] [CrossRef]
  72. Maddison, P.; Gozzard, P.; Sadalage, G.; Ambrose, P.A.; Chapman, C.J.; Murray, A.; Thomsen, S.; Berretta, A.; Lang, B. Neuronal antibody detection and improved lung cancer prediction in Lambert-Eaton myasthenic syndrome. J. Neuroimmunol. 2020, 340, 577149. [Google Scholar] [CrossRef]
  73. Agrawal, K.; Agrawal, N. Lambert-Eaton Myasthenic Syndrome Secondary to Nivolumab and Ipilimumab in a Patient with Small-Cell Lung Cancer. Case Rep. Neurol. Med. 2019, 2019, 5353202. [Google Scholar] [CrossRef] [Green Version]
  74. Nakatani, Y.; Tanaka, N.; Enami, T.; Minami, S.; Okazaki, T.; Komuta, K. Lambert-Eaton Myasthenic Syndrome Caused by Nivolumab in a Patient with Squamous Cell Lung Cancer. Case Rep. Neurol. 2018, 10, 346–352. [Google Scholar] [CrossRef] [PubMed]
  75. Anderson, C.J.; Guidon, A.; Khan, F.B.; Thomas, A.A.; Riehle, C.; Hehir, M.K.; Kolb, N.A. Case Report of Lambert Eaton Myasthenic Syndrome in a Patient with Small Cell Lung Cancer on Immune Checkpoint Inhibitor Therapy. OBM Neurobiol. 2021, 5, 1. [Google Scholar] [CrossRef]
  76. Gill, A.J.; Gandhy, S.; Lancaster, E. Nivolumab-associated Lambert-Eaton myasthenic syndrome and cerebellar dysfunction in a patient with a neuroendocrine tumor. Muscle Nerve 2021, 63, E18–E21. [Google Scholar] [CrossRef] [PubMed]
  77. Krishnan, G.S.; Wilson, W.A. Immunotherapy Related Myasthenia Gravis and Lambert Eaton Syndrome Case Reports and Review of Literature. J. Immunol. Immunother. 2020, 3, 7. [Google Scholar]
  78. Darvin, P.; Toor, S.M.; Nair, V.S.; Elkord, E. Immune checkpoint inhibitors: Recent progress and potential biomarkers. Exp. Mol. Med. 2018, 50, 1–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Burke, K.P.; Grebinoski, S.; Sharpe, A.H.; Vignali, D.A.A. Understanding adverse events of immunotherapy: A mechanistic perspective. J. Exp. Med. 2021, 218, e20192179. [Google Scholar] [CrossRef] [PubMed]
  80. Brahmer, J.R.; Lacchetti, C.; Schneider, B.J.; Atkins, M.B.; Brassil, K.J.; Caterino, J.M.; Chau, I.; Ernstoff, M.S.; Gardner, J.M.; Ginex, P.; et al. Management of immune-related adverse events in patients treated with immune checkpoint inhibitor therapy: American Society of Clinical Oncology Clinical Practice Guideline. J. Clin. Oncol. 2018, 36, 1714–1768. [Google Scholar] [CrossRef]
  81. 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, 435–443. [Google Scholar] [CrossRef] [Green Version]
  82. Petrelli, F.; Signorelli, D.; Ghidini, M.; Ghidini, A.; Pizzutilo, E.G.; Ruggieri, L.; Cabiddu, M.; Borgonovo, K.; Dognini, G.; Brighenti, M.; et al. Association of Steroids use with Survival in Patients Treated with Immune Checkpoint Inhibitors: A Systematic Review and Meta-Analysis. Cancers 2020, 12, 546. [Google Scholar] [CrossRef] [Green Version]
  83. Ferrara, R.; Lai, W.V.; Hendriks, L.; Sabari, J.K.; Caramella, C.; Plodkowski, A.J.; Halpenny, D.; Chaft, J.E.; Planchard, D.; Riely, G.J. Impact of baseline steroids on efficacy of programmed cell death-1 and programmed death-ligand 1 blockade in patients with non-small-cell lung cancer. J. Clin. Oncol. 2018, 36, 2872–2878. [Google Scholar]
  84. 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.; et al. 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, 3193–3198. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Titulaer, M.J.; Wirtz, P.W.; Willems, L.N.; Van Kralingen, K.W.; Smitt, P.A.; Verschuuren, J.J. Screening for small-cell lung cancer: A follow-up study of patients with Lambert-Eaton myasthenic syndrome. J. Clin. Oncol. 2008, 26, 4276–4281. [Google Scholar] [CrossRef] [Green Version]
  86. Moreira, A.; Loquai, C.; Pföhler, C.; Kähler, K.C.; Knauss, S.; Heppt, M.V.; Gutzmer, R.; Dimitriou, F.; Meier, F.; Mitzel-Rink, H.; et al. Myositis and neuromuscular side-effects induced by immunecheckpoint inhibitors. Eur. J. Cancer 2019, 106, 12–23. [Google Scholar] [CrossRef]
  87. Sanders, D.B.; Guptill, J.T.; Aleš, K.L.; Hobson-Webb, L.D.; Jacobus, D.P.; Mahmood, R.; Massey, J.M.; Pittman, M.M.; Prather, K.; Raja, S.M.; et al. Reliability of the triple-timed up-and-go test. Muscle Nerve 2018, 57, 136–139. [Google Scholar] [CrossRef] [Green Version]
  88. Raja, S.M.; Sanders, D.B.; Juel, V.C.; Harati, Y.; Smith, A.G.; Pascuzzi, R.; Richman, D.P.; Wu, A.; Aleš, K.L.; Jacobus, L.R.; et al. Validation of the triple timed up-and-go test in Lambert-Eaton myasthenia. Muscle Nerve 2019, 60, 292–298. [Google Scholar] [CrossRef] [PubMed]
  89. Wu, Z.Z.; Li, D.P.; Chen, S.R.; Pan, H.L. Aminopyridines potentiate synaptic and neuromuscular transmission by targeting the voltage-activated calcium channel β subunit. J. Biol. Chem. 2009, 284, 36453–36461. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Li, L.; Li, D.P.; Chen, S.R.; Chen, J.; Hu, H.; Pan, H.L. Potentiation of high voltage-activated calcium channels by 4-aminopyridine depends on subunit composition. Mol. Pharmacol. 2014, 86, 760–772. [Google Scholar] [CrossRef] [Green Version]
  91. Ojala, K.S.; Ginebaugh, S.P.; Wu, M.; Miller, E.W.; Ortiz, G.; Covarrubias, M.; Meriney, S.D. A high affinity, partial antagonist effect of 3,4-diaminopyridine mediates action potential broadening and enhancement of transmitter release at NMJs. J. Biol. Chem. 2021, 296, 100302. [Google Scholar] [CrossRef]
  92. McEvoy, K.M.; Windebank, A.J.; Daube, J.R.; Low, P.A. 3,4-Diaminopyridine in the treatment of Lambert-Eaton myasthenic syndrome. N. Engl. J. Med. 1989, 321, 1567–1571. [Google Scholar] [CrossRef] [PubMed]
  93. Sanders, D.B.; Massey, J.M.; Sanders, L.L.; Edwards, L.J. A randomized trial of 3,4-diaminopyridine in Lambert-Eaton myasthenic syndrome. Neurology 2000, 54, 603–607. [Google Scholar] [CrossRef] [PubMed]
  94. Wirtz, P.W.; Verschuuren, J.J.; van Dijk, J.G.; de Kam, M.L.; Schoemaker, R.C.; van Hasselt, J.G.; Titulaer, M.J.; Tjaden, U.R.; den Hartigh, J.; van Gerven, J.M. Efficacy of 3,4-diaminopyridine and pyridostigmine in the treatment of Lambert-Eaton myasthenic syndrome: A randomized, double-blind, placebo-controlled, crossover study. Clin. Pharmacol. Ther. 2009, 86, 44–48. [Google Scholar] [CrossRef] [PubMed]
  95. Oh, S.J.; Claussen, G.G.; Hatanaka, Y.; Morgan, M.B. 3,4-Diaminopyridine is more effective than placebo in a randomized, double-blind, cross-over drug study in LEMS. Muscle Nerve 2009, 40, 795–800. [Google Scholar] [CrossRef] [PubMed]
  96. Keogh, M.; Sedehizadeh, S.; Maddison, P. Treatment for Lambert-Eaton myasthenic syndrome. Cochrane Database Syst. Rev. 2011, 2011, CD003279. [Google Scholar] [CrossRef]
  97. Oh, S.J.; Shcherbakova, N.; Kostera-Pruszczyk, A.; Alsharabati, M.; Dimachkie, M.; Blanco, J.M.; Brannagan, T.; Lavrnić, D.; Shieh, P.B.; Vial, C.; et al. Amifampridine phosphate (Firdapse(®)) is effective and safe in a phase 3 clinical trial in LEMS. Muscle Nerve 2016, 53, 717–725. [Google Scholar] [CrossRef]
  98. Sanders, D.B.; Juel, V.C.; Harati, Y.; Smith, A.G.; Peltier, A.C.; Marburger, T.; Lou, J.S.; Pascuzzi, R.M.; Richman, D.P.; Xie, T.; et al. 3,4-Diaminopyridine base effectively treats the weakness of Lambert-Eaton myasthenia. Muscle Nerve 2018, 57, 561–568. [Google Scholar] [CrossRef] [Green Version]
  99. Shieh, P.; Sharma, K.; Kohrman, B.; Oh, S.J. Amifampridine Phosphate (Firdapse) Is Effective in a Confirmatory Phase 3 Clinical Trial in LEMS. J. Clin. Neuromuscul. Dis. 2019, 20, 111–119. [Google Scholar] [CrossRef]
  100. Wirtz, P.W.; Titulaer, M.J.; Gerven, J.M.; Verschuuren, J.J. 3,4-Diaminopyridine for the treatment of Lambert-Eaton myasthenic syndrome. Expert Rev. Clin. Immunol. 2010, 6, 867–874. [Google Scholar] [CrossRef]
  101. Newsom-Davis, J.; Murray, N.M. Plasma exchange and immunosuppressive drug treatment in the Lambert-Eaton myasthenic syndrome. Neurology 1984, 34, 480–485. [Google Scholar] [CrossRef]
  102. Patwa, H.S.; Chaudhry, V.; Katzberg, H.; Rae-Grant, A.D.; So, Y.T. Evidence- based guideline: Intravenous immunoglobulin in the treatment of neuromuscular disorders: Report of the Therapeutics and Technology Assessment Subcommittee of the American Academy of Neurology. Neurology 2012, 78, 1009–1015. [Google Scholar] [CrossRef] [Green Version]
  103. Bird, S.J. Clinical and electrophysiologic improvement in Lambert-Eaton syndrome with intravenous immunoglobulin therapy. Neurology 1992, 42, 1422–1423. [Google Scholar] [CrossRef]
  104. Okada, A.; Koike, H.; Nakamura, T.; Motomura, M.; Sobue, G. Efficacy of intravenous immunoglobulin for treatment of Lambert-Eaton myasthenic syndrome without anti-presynaptic P/Q-type voltage-gated calcium channel antibodies: A case report. Neuromuscul. Disord. 2015, 25, 70–72. [Google Scholar] [CrossRef] [PubMed]
  105. Seneviratne, U.; De Silva, R. Lambert-Eaton myasthenic syndrome. Postgrad. Med. J. 1999, 75, 516–520. [Google Scholar] [CrossRef] [PubMed]
  106. Motomura, M.; Hamasaki, S.; Nakane, S.; Fukuda, T.; Nakao, Y.K. Apheresis treatment in Lambert-Eaton myasthenic syndrome. Ther. Apher. 2000, 4, 287–290. [Google Scholar] [CrossRef] [PubMed]
  107. Maddison, P.; McConville, J.; Farrugia, M.E.; Davies, N.; Rose, M.; Norwood, F.; Jungbluth, H.; Robb, S.; Hilton-Jones, D. The use of rituximab in myasthenia gravis and Lambert-Eaton myasthenic syndrome. J. Neurol. Neurosurg. Psychiatry 2011, 82, 671–673. [Google Scholar] [CrossRef]
  108. Boutin, E.; Rey, C.; Romeu, M.; Pouget, J.; Franques, J. Favourable outcome after treatment with rituximab in a case of seronegative non-paraneoplastic Lambert-Eaton myasthenic syndrome. Rev. Med. Interne 2013, 34, 493–496. [Google Scholar] [CrossRef]
  109. Maddison, P.; Newsom-Davis, J.; Mills, K.R.; Souhami, R.L. Favourable prognosis in Lambert-Eaton myasthenic syndrome and small-cell lung carcinoma. Lancet 1999, 353, 117–118. [Google Scholar] [CrossRef]
  110. Wirtz, P.W.; Willcox, N.; van der Slik, A.R.; Lang, B.; Maddison, P.; Koeleman, B.P.; Giphart, M.J.; Wintzen, A.R.; Roep, B.O.; Verschuuren, J.J. HLA and smoking in prediction and prognosis of small cell lung cancer in autoimmune Lambert-Eaton myasthenic syndrome. J. Neuroimmunol. 2005, 159, 230–237. [Google Scholar] [CrossRef] [PubMed]
  111. Maddison, P.; Gozzard, P.; Grainge, M.J.; Lang, B. Long-term survival in paraneoplastic Lambert-Eaton myasthenic syndrome. Neurology 2017, 88, 1334–1339. [Google Scholar] [CrossRef]
  112. Lipka, A.F.; Boldingh, M.I.; van Zwet, E.W.; Schreurs, M.W.J.; Kuks, J.B.M.; Tallaksen, C.M.; Titulaer, M.J.; Verschuuren, J.J.G.M. Long-term follow-up, quality of life, and survival of patients with Lambert-Eaton myasthenic syndrome. Neurology 2020, 94, e511–e520. [Google Scholar] [CrossRef] [Green Version]
  113. Maddison, P. Neuromyotonia. Clin. Neurophysiol. 2006, 117, 2118–2127. [Google Scholar] [CrossRef]
  114. Isaacs, H. A syndrome of continuous muscle-fibre activity. J. Neurol. Neurosurg. Psychiatry 1961, 24, 319–325. [Google Scholar] [CrossRef] [Green Version]
  115. Mertens, H.G.; Zschocke, S. Neuromyotonie [Neuromyotonia]. Klin. Wochenschr. 1965, 43, 917–925. [Google Scholar] [CrossRef] [PubMed]
  116. Sinha, S.; Newsom-Davis, J.; Mills, K.; Byrne, N.; Lang, B.; Vincent, A. Autoimmune aetiology for acquired neuromyotonia (Isaacs’ syndrome). Lancet 1991, 338, 75–77. [Google Scholar] [CrossRef]
  117. Newsom-Davis, J.; Mills, K.R. Immunological associations of acquired neuromyotonia (Isaacs’ syndrome). Report of five cases and literature review. Brain 1993, 116 Pt 2, 453–469. [Google Scholar] [CrossRef]
  118. Shillito, P.; Molenaar, P.C.; Vincent, A.; Leys, K.; Zheng, W.; van den Berg, R.J.; Plomp, J.J.; van Kempen, G.T.; Chauplannaz, G.; Wintzen, A.R.; et al. Acquired neuromyotonia: Evidence for autoantibodies directed against K+ channels of peripheral nerves. Ann. Neurol. 1995, 38, 714–722. [Google Scholar] [CrossRef] [PubMed]
  119. Scott, V.E.; Muniz, Z.M.; Sewing, S.; Lichtinghagen, R.; Parcej, D.N.; Pongs, O.; Dolly, J.O. Antibodies specific for distinct Kv subunits unveil a heterooligomeric basis for subtypes of alpha-dendrotoxin-sensitive potassium channels in bovine brain. Biochemistry 2002, 33, 1617–1623. [Google Scholar] [CrossRef]
  120. Sonoda, Y.; Arimura, K.; Kurono, A.; Suehara, M.; Kameyama, M.; Minato, S.; Hayashi, A.; Osame, M. Serum of Isaacs’ syndrome suppresses potassium channels in PC-12 cell lines. Muscle Nerve 1996, 19, 1439–1446. [Google Scholar] [CrossRef]
  121. Nagado, T.; Arimura, K.; Sonoda, Y.; Kurono, A.; Horikiri, Y.; Kameyama, A.; Kameyama, M.; Pongs, O.; Osame, M. Potassium current suppression in patients with peripheral nerve hyperexcitability. Brain 1999, 122 Pt 11, 2057–2066. [Google Scholar] [CrossRef] [Green Version]
  122. Liguori, R.; Vincent, A.; Clover, L.; Avoni, P.; Plazzi, G.; Cortelli, P.; Baruzzi, A.; Carey, T.; Gambetti, P.; Lugaresi, E.; et al. Morvan’s syndrome: Peripheral and central nervous system and cardiac involvement with antibodies to voltage-gated potassium channels. Brain 2001, 124 Pt 12, 2417–2426. [Google Scholar] [CrossRef] [Green Version]
  123. Buckley, C.; Oger, J.; Clover, L.; Tüzün, E.; Carpenter, K.; Jackson, M.; Vincent, A. Potassium channel antibodies in two patients with reversible limbic encephalitis. Ann. Neurol. 2001, 50, 73–78. [Google Scholar] [CrossRef] [PubMed]
  124. Vincent, A.; Buckley, C.; Schott, J.M.; Baker, I.; Dewar, B.K.; Detert, N.; Clover, L.; Parkinson, A.; Bien, C.G.; Omer, S.; et al. Potassium channel antibody-associated encephalopathy: A potentially immunotherapy-responsive form of limbic encephalitis. Brain 2004, 127, 701–712. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Thieben, M.J.; Lennon, V.A.; Boeve, B.F.; Aksamit, A.J.; Keegan, M.; Vernino, S. Potentially reversible autoimmune limbic encephalitis with neuronal potassium channel antibody. Neurology 2004, 62, 1177–1182. [Google Scholar] [CrossRef]
  126. Irani, S.R.; Alexander, S.; Waters, P.; Kleopa, K.A.; Pettingill, P.; Zuliani, L.; Peles, E.; Buckley, C.; Lang, B.; Vincent, A. Antibodies to Kv1 potassium channel-complex proteins leucine-rich, glioma inactivated 1 protein and contactin-associated protein-2 in limbic encephalitis, Morvan’s syndrome and acquired neuromyotonia. Brain 2010, 133, 2734–2748. [Google Scholar] [CrossRef]
  127. Patterson, K.R.; Dalmau, J.; Lancaster, E. Mechanisms of Caspr2 antibodies in autoimmune encephalitis and neuromyotonia. Ann. Neurol. 2018, 83, 40–51. [Google Scholar] [CrossRef] [PubMed]
  128. Giannoccaro, M.P.; Menassa, D.A.; Jacobson, L.; Coutinho, E.; Prota, G.; Lang, B.; Leite, M.I.; Cerundolo, V.; Liguori, R.; Vincent, A. Behaviour and neuropathology in mice injected with human contactin-associated protein 2 antibodies. Brain 2019, 142, 2000–2012. [Google Scholar] [CrossRef]
  129. Dawes, J.M.; Weir, G.A.; Middleton, S.J.; Patel, R.; Chisholm, K.I.; Pettingill, P.; Peck, L.J.; Sheridan, J.; Shakir, A.; Jacobson, L.; et al. Immune or Genetic-Mediated Disruption of CASPR2 Causes Pain Hypersensitivity Due to Enhanced Primary Afferent Excitability. Neuron 2018, 97, 806–822.e10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Ohkawa, T.; Fukata, Y.; Yamasaki, M.; Miyazaki, T.; Yokoi, N.; Takashima, H.; Watanabe, M.; Watanabe, O.; Fukata, M. Autoantibodies to epilepsy-related LGI1 in limbic encephalitis neutralize LGI1-ADAM22 interaction and reduce synaptic AMPA receptors. J. Neurosci. 2013, 33, 18161–18174. [Google Scholar] [CrossRef] [PubMed]
  131. Petit-Pedrol, M.; Sell, J.; Planagumà, J.; Mannara, F.; Radosevic, M.; Haselmann, H.; Ceanga, M.; Sabater, L.; Spatola, M.; Soto, D.; et al. LGI1 antibodies alter Kv1.1 and AMPA receptors changing synaptic excitability, plasticity and memory. Brain 2018, 141, 3144–3159. [Google Scholar] [CrossRef]
  132. Vernino, S.; Lennon, V.A. Ion channel and striational antibodies define a continuum of autoimmune neuromuscular hyperexcitability. Muscle Nerve 2002, 26, 702–707. [Google Scholar] [CrossRef]
  133. Hart, I.K.; Maddison, P.; Newsom-Davis, J.; Vincent, A.; Mills, K.R. Phenotypic variants of autoimmune peripheral nerve hyperexcitability. Brain 2002, 125 Pt 8, 1887–1895. [Google Scholar] [CrossRef] [Green Version]
  134. Rubio-Agusti, I.; Perez-Miralles, F.; Sevilla, T.; Muelas, N.; Chumillas, M.J.; Mayordomo, F.; Azorin, I.; Carmona, E.; Moscardo, F.; Palau, J.; et al. Peripheral nerve hyperexcitability: A clinical and immunologic study of 38 patients. Neurology 2011, 76, 172–178. [Google Scholar] [CrossRef]
  135. Liewluck, T.; Klein, C.J.; Jones, L.K., Jr. Cramp-fasciculation syndrome in patients with and without neural autoantibodies. Muscle Nerve 2014, 49, 351–356. [Google Scholar] [CrossRef] [PubMed]
  136. Vincent, A.; Pettingill, P.; Pettingill, R.; Lang, B.; Birch, R.; Waters, P.; Irani, S.R.; Buckley, C.; Watanabe, O.; Arimura, K.; et al. Association of Leucine-Rich Glioma Inactivated Protein 1, Contactin-Associated Protein 2, and Contactin 2 Antibodies With Clinical Features and Patient-Reported Pain in Acquired Neuromyotonia. JAMA Neurol. 2018, 75, 1519–1527. [Google Scholar] [CrossRef] [Green Version]
  137. Muñiz-Castrillo, S.; Joubert, B.; Elsensohn, M.H.; Pinto, A.L.; Saint-Martin, M.; Vogrig, A.; Picard, G.; Rogemond, V.; Dubois, V.; Tamouza, R.; et al. Anti-CASPR2 clinical phenotypes correlate with HLA and immunological features. J. Neurol. Neurosurg. Psychiatry 2020, 91, 1076–1084. [Google Scholar] [CrossRef]
  138. Binks, S.; Varley, J.; Lee, W.; Makuch, M.; Elliott, K.; Gelfand, J.M.; Jacob, S.; Leite, M.I.; Maddison, P.; Chen, M.; et al. Distinct HLA associations of LGI1 and CASPR2-antibody diseases. Brain 2018, 141, 2263–2271. [Google Scholar] [CrossRef] [PubMed]
  139. van Sonderen, A.; Roelen, D.L.; Stoop, J.A.; Verdijk, R.M.; Haasnoot, G.W.; Thijs, R.D.; Wirtz, P.W.; Schreurs, M.W.; Claas, F.H.; Sillevis Smitt, P.A.; et al. Anti-LGI1 encephalitis is strongly associated with HLA-DR7 and HLA-DRB4. Ann. Neurol. 2017, 81, 193–198. [Google Scholar] [CrossRef] [PubMed]
  140. Nakazato, T.; Tsuji, Y.; Kanai, K.; Noto, Y.I.; Hoshino, Y.; Yamashiro, K.; Yokoyama, K.; Nishioka, K.; Shimo, Y.; Watanabe, O.; et al. Isaacs syndrome: A slow potassium channelopathy caused by autoantibodies? Clin. Neurophysiol. 2018, 129, 956–958. [Google Scholar] [CrossRef]
  141. Antozzi, C.; Frassoni, C.; Vincent, A.; Regondi, M.C.; Andreetta, F.; Bernasconi, P.; Ciano, C.; Chang, T.; Cornelio, F.; Spreafico, R.; et al. Sequential antibodies to potassium channels and glutamic acid decarboxylase in neuromyotonia. Neurology 2005, 64, 1290–1293. [Google Scholar] [CrossRef]
  142. Simon, N.G.; Reddel, S.W.; Kiernan, M.C.; Layzer, R. Muscle-specific kinase antibodies: A novel cause of peripheral nerve hyperexcitability? Muscle Nerve 2013, 48, 819–823. [Google Scholar] [CrossRef]
  143. Wenninger, S. Expanding the Clinical Spectrum of IgLON5-Syndrome. J. Neuromuscul. Dis. 2017, 4, 337–339. [Google Scholar] [CrossRef]
  144. Giannoccaro, M.P.; Crisp, S.J.; Vincent, A. Antibody-mediated central nervous system diseases. Brain Neurosci. Adv. 2018, 2, 2398212818817497. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Ahmed, A.; Simmons, Z. Isaacs syndrome: A review. Muscle Nerve 2015, 52, 5–12. [Google Scholar] [CrossRef] [PubMed]
  146. Gutmann, L.; Gutmann, L. Myokymia and neuromyotonia. J. Neurol. 2004, 251, 138–142. [Google Scholar] [CrossRef]
  147. Joubert, B.; Honnorat, J. Autoimmune channelopathies in paraneoplastic neurological syndromes. Biochim. Biophys. Acta 2015, 1848, 2665–2676. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Gonzalez Primomo, S.N.; Blas, L.; Bertotti, A.C.; Ameri, C. Urinary manifestations in Isaacs’s syndrome. Our experience in 8 cases. Neurourol. Urodyn. 2018, 37, 496–500. [Google Scholar] [CrossRef] [Green Version]
  149. Surana, S.; Kumar, R.; Pitt, M.; Hafner, P.; Mclellan, A.; Davidson, J.; Prabakhar, P.; Vincent, A.; Hacohen, Y.; Wright, S. Acquired neuromyotonia in children with CASPR2 and LGI1 antibodies. Dev. Med. Child Neurol. 2019, 61, 1344–1347. [Google Scholar] [CrossRef] [Green Version]
  150. López-Chiriboga, A.S.; Klein, C.; Zekeridou, A.; McKeon, A.; Dubey, D.; Flanagan, E.P.; Lennon, V.A.; Tillema, J.M.; Wirrell, E.C.; Patterson, M.C.; et al. LGI1 and CASPR2 neurological autoimmunity in children. Ann. Neurol. 2018, 84, 473–480. [Google Scholar] [CrossRef] [PubMed]
  151. Santos, M.O.; Swash, M.; de Carvalho, M. The generator site in acquired autoimmune neuromyotonia. Clin. Neurophysiol. 2017, 128, 643–646. [Google Scholar] [CrossRef]
  152. Boyko, M.; Au, K.L.K.; Casault, C.; de Robles, P.; Pfeffer, G. Systematic review of the clinical spectrum of CASPR2 antibody syndrome. J. Neurol. 2020, 267, 1137–1146. [Google Scholar] [CrossRef] [PubMed]
  153. Torres-Vega, E.; Mancheño, N.; Cebrián-Silla, A.; Herranz-Pérez, V.; Chumillas, M.J.; Moris, G.; Joubert, B.; Honnorat, J.; Sevilla, T.; Vílchez, J.J.; et al. Netrin-1 receptor antibodies in thymoma-associated neuromyotonia with myasthenia gravis. Neurology 2017, 88, 1235–1242. [Google Scholar] [CrossRef] [Green Version]
  154. Gastaldi, M.; De Rosa, A.; Maestri, M.; Zardini, E.; Scaranzin, S.; Guida, M.; Borrelli, P.; Ferraro, O.E.; Lampasona, V.; Furlan, R.; et al. Acquired neuromyotonia in thymoma-associated myasthenia gravis: A clinical and serological study. Eur. J. Neurol. 2019, 26, 992–999. [Google Scholar] [CrossRef]
  155. Marx, A.; Porubsky, S.; Belharazem, D.; Saruhan-Direskeneli, G.; Schalke, B.; Ströbel, P.; Weis, C.A. Thymoma related myasthenia gravis in humans and potential animal models. Exp. Neurol. 2015, 270, 55–65. [Google Scholar] [CrossRef] [PubMed]
  156. Dhand, U.K. Isaacs syndrome: Clinical and electrophysiological response to gabapentin. Muscle Nerve 2006, 34, 646–650. [Google Scholar] [CrossRef] [PubMed]
  157. Wasserstein, P.H.; Shadlen, M.N.; Dolfman, L.J. Successful treatment of neuromyotonia with mexiletine. Neurology 1992, 42 (Suppl. 3), 270. [Google Scholar]
  158. Celebisoy, N.; Colakoglu, Z.; Akbaba, Y.; Yuceyar, N. Continuous muscle fibre activity: A case treated with acetazolamide. J. Neurol. Neurosurg. Psychiatry 1998, 64, 256–258. [Google Scholar] [CrossRef] [PubMed]
  159. Skeie, G.O.; Apostolski, S.; Evoli, A.; Gilhus, N.E.; Illa, I.; Harms, L.; Hilton-Jones, D.; Melms, A.; Verschuuren, J.; Horge, H.W. Guidelines for treatment of autoimmune neuromuscular transmission disorders. Eur. J. Neurol. 2010, 17, 893–902. [Google Scholar] [CrossRef]
  160. van Sonderen, A.; Wirtz, P.W.; Verschuuren, J.J.; Titulaer, M.J. Paraneoplastic syndromes of the neuromuscular junction: Therapeutic options in myasthenia gravis, lambert-eaton myasthenic syndrome, and neuromyotonia. Curr. Treat. Options Neurol. 2013, 15, 224–239. [Google Scholar] [CrossRef]
  161. Ginsburg, G.; Forde, R.; Martyn, J.A.; Eikermann, M. Increased sensitivity to a nondepolarizing muscle relaxant in a patient with acquired neuromyotonia. Muscle Nerve 2009, 40, 139–142. [Google Scholar] [CrossRef]
Figure 1. Pathophysiology of LEMS. In normal conditions, the depolarization of the presynaptic nerve terminal leads to calcium ions influx, acetylcholine (ACh) release, and binding to the ACh receptors (AChR) with a consequent influx of positively charged ions, mainly sodium, generation of an endplate potential (EPP) and muscle contraction. In LEMS, voltage gated calcium channel (VGCC) antibodies block calcium influx causing a reduction of the ACh released at the presynaptic terminal with consequent reduction of the EPP amplitude. High-frequency repetitive nerve stimulation however can increase the EPP amplitude through calcium accumulation in the presynaptic terminal and increased ACh release.
Figure 1. Pathophysiology of LEMS. In normal conditions, the depolarization of the presynaptic nerve terminal leads to calcium ions influx, acetylcholine (ACh) release, and binding to the ACh receptors (AChR) with a consequent influx of positively charged ions, mainly sodium, generation of an endplate potential (EPP) and muscle contraction. In LEMS, voltage gated calcium channel (VGCC) antibodies block calcium influx causing a reduction of the ACh released at the presynaptic terminal with consequent reduction of the EPP amplitude. High-frequency repetitive nerve stimulation however can increase the EPP amplitude through calcium accumulation in the presynaptic terminal and increased ACh release.
Brainsci 11 01035 g001
Figure 2. Effect of 3,4-diamniopyride treatment in LEMS. 3,4-diaminopyridine (3,4-DAP) blocks the efflux of potassium (K+) prolonging the presynaptic depolarization, which in turn prolongs the VGCCs opening time, allowing the influx of an increased amount of calcium ions with consequently increased ACh release.
Figure 2. Effect of 3,4-diamniopyride treatment in LEMS. 3,4-diaminopyridine (3,4-DAP) blocks the efflux of potassium (K+) prolonging the presynaptic depolarization, which in turn prolongs the VGCCs opening time, allowing the influx of an increased amount of calcium ions with consequently increased ACh release.
Brainsci 11 01035 g002
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Giannoccaro, M.P.; Avoni, P.; Liguori, R. Presynaptic Paraneoplastic Disorders of the Neuromuscular Junction: An Update. Brain Sci. 2021, 11, 1035. https://0-doi-org.brum.beds.ac.uk/10.3390/brainsci11081035

AMA Style

Giannoccaro MP, Avoni P, Liguori R. Presynaptic Paraneoplastic Disorders of the Neuromuscular Junction: An Update. Brain Sciences. 2021; 11(8):1035. https://0-doi-org.brum.beds.ac.uk/10.3390/brainsci11081035

Chicago/Turabian Style

Giannoccaro, Maria Pia, Patrizia Avoni, and Rocco Liguori. 2021. "Presynaptic Paraneoplastic Disorders of the Neuromuscular Junction: An Update" Brain Sciences 11, no. 8: 1035. https://0-doi-org.brum.beds.ac.uk/10.3390/brainsci11081035

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop