Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 756
Filter
1.
Br J Surg ; 111(5)2024 May 03.
Article in English | MEDLINE | ID: mdl-38743864

ABSTRACT

BACKGROUND: Postoperative ileus, driven by the cholinergic anti-inflammatory pathway, is the most common complication in patients undergoing colorectal surgery. By inhibiting acetylcholinesterase, pyridostigmine can potentially modulate the cholinergic anti-inflammatory pathway and accelerate gastrointestinal recovery. This study aimed to assess the efficacy of pyridostigmine in improving gastrointestinal recovery after colorectal surgery. METHODS: This double-blinded RCT enrolled adult patients undergoing elective colorectal surgery at two hospitals in South Australia. Patients were randomized to 60 mg oral pyridostigmine or placebo twice daily starting 6 h after surgery until the first passage of stool. The primary outcome was GI-2, a validated composite measure of time to first stool and tolerance of oral diet. Secondary outcomes included incidence of postoperative ileus (defined as GI-2 greater than 4 days), duration of hospital stay, and 30-day complications, evaluated by intention-to-treat univariate analysis. RESULTS: Of 130 patients recruited (mean(s.d.) age 58.4(16.4) years; 73 men, 56%), 65 were allocated to each arm. The median GI-2 was 1 day shorter with pyridostigmine compared with placebo (2 (i.q.r. 1-3) versus 3 (2-4) days; P = 0.015). However, there were no significant differences in postoperative ileus (17.2 versus 21.5%; P = 0.532) or duration of hospital stay (median 5 (i.q.r. 4-8.75) versus 5 (4-7.5) days; P = 0.921). Similarly, there were no significant differences in overall complications, anastomotic leak, cardiac complications, or patient-reported side effects. CONCLUSION: Pyridostigmine resulted in a quicker return of GI-2 and was well tolerated. Larger multicentre studies are required to determine the optimal dosing and evaluate the impact of pyridostigmine in different surgical settings. Registration number: ACTRN12621000530820 (https://anzctr.org.au).


Subject(s)
Cholinesterase Inhibitors , Ileus , Postoperative Complications , Pyridostigmine Bromide , Humans , Male , Ileus/prevention & control , Ileus/etiology , Female , Double-Blind Method , Middle Aged , Postoperative Complications/prevention & control , Cholinesterase Inhibitors/administration & dosage , Cholinesterase Inhibitors/adverse effects , Cholinesterase Inhibitors/therapeutic use , Pyridostigmine Bromide/administration & dosage , Pyridostigmine Bromide/therapeutic use , Aged , Length of Stay , Adult , Treatment Outcome
2.
Digit J Ophthalmol ; 30(1): 15-18, 2024.
Article in English | MEDLINE | ID: mdl-38601901

ABSTRACT

We report 2 cases of pediatric ocular myasthenia gravis. The first case was a 7-year-old girl who presented with bilateral ophthalmoplegia and ptosis that correlated with the onset of upper respiratory symptoms. Neuroimaging and acetylcholine receptor antibody testing were unremarkable. The ice pack test was positive. Symptoms greatly improved with pyridostigmine, with full resolution of ophthalmoplegia achieved by 8-month follow-up. The second case was a 4-year-old girl who presented emergently with ptosis and bilateral ophthalmoplegia. Acetylcholine receptor antibodies testing was positive. The patient was started on pyridostigmine and intravenous immunoglobulin and is scheduled to follow-up with pediatric ophthalmology in the outpatient setting.


Subject(s)
Blepharoptosis , Myasthenia Gravis , Ophthalmoplegia , Female , Child , Humans , Child, Preschool , Pyridostigmine Bromide/therapeutic use , Myasthenia Gravis/complications , Myasthenia Gravis/diagnosis , Blepharoptosis/diagnosis , Blepharoptosis/etiology , Ophthalmoplegia/diagnosis , Ophthalmoplegia/etiology , Receptors, Cholinergic , Autoantibodies
3.
Eur J Neurol ; 31(5): e16229, 2024 May.
Article in English | MEDLINE | ID: mdl-38321574

ABSTRACT

BACKGROUND: Generalized myasthenia gravis (MG) with antibodies against the acetylcholine receptor is a chronic disease causing muscle weakness. Access to novel treatments warrants authoritative treatment recommendations. The Nordic countries have similar, comprehensive health systems, mandatory health registers, and extensive MG research. METHODS: MG experts and patient representatives from the five Nordic countries formed a working group to prepare treatment guidance for MG based on a systematic literature search and consensus meetings. RESULTS: Pyridostigmine represents the first-line symptomatic treatment, while ambenonium and beta adrenergic agonists are second-line options. Early thymectomy should be undertaken if a thymoma, and in non-thymoma patients up to the age of 50-65 years if not obtaining remission on symptomatic treatment. Most patients need immunosuppressive drug treatment. Combining corticosteroids at the lowest possible dose with azathioprine is recommended, rituximab being an alternative first-line option. Mycophenolate, methotrexate, and tacrolimus represent second-line immunosuppression. Plasma exchange and intravenous immunoglobulin are used for myasthenic crises and acute exacerbations. Novel complement inhibitors and FcRn blockers are effective and fast-acting treatments with promising safety profiles. Their use depends on local availability, refunding policies, and cost-benefit analyses. Adapted physical training is recommended. Planning of pregnancies with optimal treatment, information, and awareness of neonatal MG is necessary. Social support and adaptation of work and daily life activities are recommended. CONCLUSIONS: Successful treatment of MG rests on timely combination of different interventions. Due to spontaneous disease fluctuations, comorbidities, and changes in life conditions, regular long-term specialized follow-up is needed. Most patients do reasonably well but there is room for further improvement. Novel treatments are promising, though subject to restricted access due to costs.


Subject(s)
Myasthenia Gravis , Neuromuscular Diseases , Thymus Neoplasms , Pregnancy , Female , Infant, Newborn , Humans , Middle Aged , Aged , Myasthenia Gravis/drug therapy , Receptors, Cholinergic , Pyridostigmine Bromide/therapeutic use , Immunosuppressive Agents/therapeutic use , Autoantibodies , Thymectomy
4.
Pediatr Neurol ; 153: 137-143, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38382246

ABSTRACT

BACKGROUND: Currently, there is no universally accepted standard treatment for ocular myasthenia gravis (OMG) in children. We aimed to investigate the possible proper regimens and timing of treatment for pediatric OMG cases based on the clinical manifestations: OMG with ptosis only and OMG with other features. METHODS: One hundred and forty two OMG cases attended at the Department of Pediatrics, Xiangya Hospital, Central South University, from 2010 to 2019 were included, and information from medical records was reviewed and recorded. Comparisons of clinical characteristics between patients with OMG with ptosis only and patients with OMG with other features as well as between patients treated with glucocorticoid (GC) within or after six months from disease onset were performed. RESULTS: OMG with other features constituted about 54.9% of the cases, and 66.2% of the patients achieved optimal outcome. Patients with OMG with ptosis only responded to pyridostigmine alone more than patients with OMG with other features who required several therapies (P < 0.001). Patients with OMG with ptosis only had a larger proportion of optimal outcome than the patients with OMG with other features (P = 0.002), and the difference remained significant even when the individual outcome groups were compared (P < 0.001). Patients who received GC within six months had a greater proportion of optimal outcome than those who received it after six months (P < 0.001). CONCLUSIONS: Although OMG with other features is a more common subtype of OMG, it is also more severe than OMG with ptosis only. An earlier addition of GC leads to optimal outcome.


Subject(s)
Blepharoptosis , Myasthenia Gravis , Humans , Child , Myasthenia Gravis/diagnosis , Myasthenia Gravis/drug therapy , Blepharoptosis/drug therapy , Blepharoptosis/etiology , Pyridostigmine Bromide/therapeutic use , Glucocorticoids/therapeutic use , Retrospective Studies
5.
Medicine (Baltimore) ; 103(4): e36989, 2024 Jan 26.
Article in English | MEDLINE | ID: mdl-38277576

ABSTRACT

RATIONALE: Patients with elderly-onset myasthenia gravis can have a good prognosis with appropriate diagnosis and response, although it is difficult to differentiate between exacerbations of myasthenia gravis in elderly patients and age-related changes. Therefore, it is important for physicians to understand the clinical characteristics and safe assessment methods for patients with elderly-onset myasthenia gravis. PATIENT CONCERNS: An 82-year-old male diagnosed with myasthenia gravis 6 months prior had no difficulty in daily living. After falling on a golf course, he was diagnosed with a right femoral neck fracture on the 1st day and underwent right total hip replacement surgery on the 12th day, being transferred to our hospital for rehabilitation therapy on the 32nd day. However, immediately after transfer, the patient showed fatigability during training and difficulty swallowing food. DIAGNOSES: This case was diagnosed as an exacerbation of myasthenia gravis. INTERVENTIONS: Pyridostigmine was initiated with the expectation of immediate effect on the 54th day. OUTCOMES: His symptoms and physical functions improved immediately, and walking distance and food intake increased. From this clinical course, it was judged that immunosuppressive therapy was indicated as a transition to generalized myasthenia gravis. For this reason, he was discharged after arranging postdischarge visits to the department of neurology, accordingly. LESSONS: A better understanding of the characteristics of elderly-onset myasthenia gravis may allow for relatively safe assessment of the condition and improve its diagnosis and treatment.


Subject(s)
Deglutition Disorders , Myasthenia Gravis , Aged, 80 and over , Humans , Male , Aftercare , Deglutition Disorders/drug therapy , Myasthenia Gravis/diagnosis , Myasthenia Gravis/drug therapy , Patient Discharge , Pyridostigmine Bromide/therapeutic use
6.
Am J Case Rep ; 24: e941792, 2023 Nov 25.
Article in English | MEDLINE | ID: mdl-38006204

ABSTRACT

BACKGROUND Myasthenia gravis is a neuromuscular disorder that is strongly associated with thymoma. Although the presence of myasthenia gravis with other tumors is uncommon, approximately 50% of patients with thymoma have myasthenia gravis. Thymic Hodgkin lymphoma should be considered due to the multiple reported cases of patients with myasthenia gravis and Hodgkin lymphoma. In this report, we present the case of 24-year-old woman with myasthenia gravis who was incidentally found to have coexisting thymoma with thymic Hodgkin lymphoma. CASE REPORT A 24-year-old woman with a known case of vitiligo presented with a 2-year history of diplopia and incidental anterior mediastinal mass. Following investigations, myasthenia gravis was diagnosed and managed by pyridostigmine, prednisolone, and azathioprine. Regarding the anterior mediastinal mass, thymoma was suspected based on the presence of myasthenia gravis and radiological findings. She underwent extended transsternal thymectomy. The final histopathological report of the dissected thymus disclosed Hodgkin lymphoma pathology coexisting with thymoma. After the diagnosis of Hodgkin lymphoma nodular sclerosis type IIA was confirmed, 6 cycles of chemotherapy were administered. Four years of follow-up revealed no evidence of Hodgkin lymphoma. However, her symptoms of myasthenia gravis persisted despite Hodgkin lymphoma remission. CONCLUSIONS There is an unclear association between myasthenia gravies and Hodgkin lymphoma. Prior reports revealed regression of myasthenia gravies following Hodgkin lymphoma management, which suggests that myasthenia could be a complication of Hodgkin lymphoma. However, in our case, myasthenia gravis persisted after Hodgkin lymphoma management; therefore, further studies are needed to explore this association.


Subject(s)
Hodgkin Disease , Myasthenia Gravis , Thymoma , Thymus Neoplasms , Female , Humans , Young Adult , Hodgkin Disease/complications , Hodgkin Disease/diagnosis , Myasthenia Gravis/complications , Myasthenia Gravis/diagnosis , Pyridostigmine Bromide/therapeutic use , Thymoma/complications , Thymoma/diagnosis , Thymoma/pathology , Thymus Neoplasms/complications , Thymus Neoplasms/diagnosis , Thymus Neoplasms/pathology
7.
J Am Anim Hosp Assoc ; 59(5): 249-254, 2023 Sep 01.
Article in English | MEDLINE | ID: mdl-37708471

ABSTRACT

Acquired myasthenia gravis (MG) in dogs can present with focal or generalized weakness and is diagnosed by the presence of circulating antibodies to the acetylcholine receptor. Megaesophagus is the most common focal form of MG. Although exacerbation of MG has been associated with the use of fluoroquinolones in humans, it has not been previously described in dogs. The medical records of 46 dogs diagnosed with MG based on acetylcholine receptor antibody testing from 1997 to 2021 were retrospectively evaluated to identify any dogs who demonstrated exacerbation of MG after the administration of a fluoroquinolone. Exacerbation of MG, from focal to generalized, occurred in a median of 4.5 days after initiation of fluoroquinolone therapy in six dogs. In addition, one dog with generalized MG and megaesophagus developed pyridostigmine resistance subsequent to fluoroquinolone therapy. Marked improvement in generalized weakness was reported 36 hr after discontinuation of fluoroquinolone therapy alone in one dog and in combination with pyridostigmine in two dogs. Fluoroquinolone therapy was never stopped in three dogs who were euthanized because of severe weakness and one dog who died of respiratory arrest.


Subject(s)
Dog Diseases , Esophageal Achalasia , Myasthenia Gravis , Humans , Dogs , Animals , Pyridostigmine Bromide/therapeutic use , Esophageal Achalasia/veterinary , Retrospective Studies , Dog Diseases/chemically induced , Dog Diseases/drug therapy , Myasthenia Gravis/chemically induced , Myasthenia Gravis/drug therapy , Myasthenia Gravis/veterinary , Fluoroquinolones/adverse effects , Receptors, Cholinergic
8.
J Med Case Rep ; 17(1): 402, 2023 Sep 08.
Article in English | MEDLINE | ID: mdl-37679826

ABSTRACT

BACKGROUND: Myasthenia gravis is an autoimmune condition affecting the neuromuscular junction and causing muscle weakness along with fatigue (myasthenia). When the clinical manifestations of myasthenia gravis are isolated to the eye muscles, only causing weak eye movements, it is referred to as ocular myasthenia gravis, which can mimic a 1 and ½ syndrome. CASE PRESENTATION: An African-American female in her fifties with past medical history of hypertension presented to our outpatient clinic with complaints of blurred vision for two weeks. Her symptoms were associated with facial discomfort and a generalized headache. On physical examination upon her initial presentation, there was demonstratable swelling of the left upper eyelid with drooping. Her extraocular movements revealed defects with the abduction and adduction of the right eye, and the left eye would not adduct, although the outward movement was normal. The left eye failed to lift/elevate completely when looking upwards, a pseudo 1 and ½ syndrome. A positive Cogan lid twitch was also noticed. Imaging of the brain and orbit ruled out central causes. Diagnosis of ocular myasthenia gravis was made in accordance with positive anti-acetylcholine receptor antibodies. With 120 mg pyridostigmine oral dose, the patient experienced improvement subjectively and objectively, and the patient was discharged on oral pyridostigmine and prednisone. Six months later, with prednisone having been tapered off, the patient developed a myasthenic crisis and was treated with plasmapheresis and intravenous immunoglobulins. After recovering from the myasthenic crisis, efgartigimod infusions were instituted, which helped our patient restore normal life. CONCLUSION: Our patient who presented with "blurred vision" was discovered to have binocular diplopia due to significant dysconjugate eye movements. After diligently ruling out central etiologies, we concluded that her presentation was due to a peripheral etiology. Her serologies and her presentation helped confirm a diagnosis of ocular myasthenia gravis. Also, as in most cases, our patient also progressed to develop generalized myasthenia gravis while on pyridostigmine. Efgartigimod infusions instituted after our patient recovered from a myasthenic crisis have helped her restore a normal life.


Subject(s)
Diplopia , Myasthenia Gravis , Female , Humans , Diplopia/etiology , Pyridostigmine Bromide/therapeutic use , Prednisone/therapeutic use , Vision Disorders , Myasthenia Gravis/complications , Myasthenia Gravis/diagnosis , Myasthenia Gravis/therapy , Muscle Weakness
9.
Clin Neurophysiol ; 154: 100-106, 2023 10.
Article in English | MEDLINE | ID: mdl-37595479

ABSTRACT

OBJECTIVE: To investigate the electrophysiological basis of pyridostigmine enhancement of endurance performance documented earlier in patients with spinal muscular atrophy (SMA). METHODS: We recorded surface electromyography (sEMG) in four upper extremity muscles of 31 patients with SMA types 2 and 3 performing endurance shuttle tests (EST) and maximal voluntary contraction (MVC) measurements during a randomized, double blind, cross-over, phase II trial. Linear mixed effect models (LMM) were used to assess the effect of pyridostigmine on (i) time courses of median frequencies and of root mean square (RMS) amplitudes of sEMG signals and (ii) maximal RMS amplitudes during MVC measurements. These sEMG changes over time indicate levels of peripheral muscle fatigue and recruitment of new motor units, respectively. RESULTS: In comparison to a placebo, patients with SMA using pyridostigmine had fourfold smaller decreases in frequency and twofold smaller increases in amplitudes of sEMG signals in some muscles, recorded during ESTs (p < 0.05). We found no effect of pyridostigmine on MVC RMS amplitudes. CONCLUSIONS: sEMG parameters indicate enhanced low-threshold (LT) motor unit (MU) function in upper-extremity muscles of patients with SMA treated with pyridostigmine. This may underlie their improved endurance. SIGNIFICANCE: Our results suggest that enhancing LT MU function may constitute a therapeutic strategy to reduce fatigability in patients with SMA.


Subject(s)
Muscular Atrophy, Spinal , Pyridostigmine Bromide , Humans , Pyridostigmine Bromide/pharmacology , Pyridostigmine Bromide/therapeutic use , Electromyography/methods , Muscles/physiology , Muscle Fatigue/physiology , Muscle, Skeletal/physiology
10.
J Am Assoc Nurse Pract ; 35(8): 519-523, 2023 Aug 01.
Article in English | MEDLINE | ID: mdl-37141567

ABSTRACT

ABSTRACT: Congenital myasthenic syndrome (CMS) is a group of rare genetic disorders that mimics the symptoms of myasthenia gravis, but it is due to a genetic defect. We present a case of a male CMS patient, and the course of the disease through the years. The patient initially presented with generalized muscle weakness and difficulty swallowing. During the follow-up, he developed difficulty in chewing, bilateral external ophthalmoparesis with an almost full block of eye movements and bulbar syndrome. The case illustrates both the clinical heterogeneity and the progressive worsening of the symptoms of the disease over the years. The optimal treatment for CMS is based on the molecular defect and its localization in the neuromuscular junction. In our case, treatment with pyridostigmine resulted in good long-term control of symptoms. As a result of the patient's good compliance with treatment, he was not admitted to hospital because of respiratory distress. The lack of a unified protocol for the treatment of CMS highlights the need for a more personalized approach when dealing with patients with rare diseases.


Subject(s)
Myasthenia Gravis , Myasthenic Syndromes, Congenital , Humans , Male , Mutation/genetics , Myasthenia Gravis/diagnosis , Myasthenic Syndromes, Congenital/drug therapy , Myasthenic Syndromes, Congenital/genetics , Myasthenic Syndromes, Congenital/diagnosis , Pyridostigmine Bromide/therapeutic use , Adult , Treatment Outcome
11.
Neurotoxicology ; 96: 197-206, 2023 05.
Article in English | MEDLINE | ID: mdl-37160207

ABSTRACT

Gulf War Illness (GWI) is an unrelenting multi-symptom illness with chronic central nervous system and peripheral pathology affecting veterans from the 1991 Gulf War and for which effective treatment is lacking. An increasing number of studies indicate that persistent neuroinflammation is likely the underlying cause of cognitive and mood dysfunction that affects veterans with GWI. We have previously reported that fingolimod, a drug approved for the treatment of relapsing-remitting multiple sclerosis, decreases neuroinflammation and improves cognition in a mouse model of Alzheimer's disease. In this study, we investigated the effect of fingolimod treatment on cognition and neuroinflammation in a mouse model of GWI. We exposed C57BL/6 J male mice to GWI-related chemicals pyridostigmine bromide, DEET, and permethrin, and to mild restraint stress for 28 days (GWI mice). Control mice were exposed to the chemicals' vehicle only. Starting 3 months post-exposure, half of the GWI mice and control mice were orally treated with fingolimod (1 mg/kg/day) for 1 month, and the other half were left untreated. Decreased memory on the Morris water maze test was detected in GWI mice compared to control mice and was reversed by fingolimod treatment. Immunohistochemical analysis of brain sections with antibodies to Iba1 and GFAP revealed that GWI mice had increased microglia activation in the hippocampal dentate gyrus, but no difference in reactive astrocytes was detected. The increased activation of microglia in GWI mice was decreased to the level in control mice by treatment with fingolimod. No effect of fingolimod treatment on gliosis in control mice was detected. To explore the signaling pathways by which decreased memory and increased neuroinflammation in GWI may be protected by fingolimod, we investigated the involvement of the inflammatory signaling pathways of protein kinase R (PKR) in the cerebral cortex of these mice. We found increased phosphorylation of PKR in the brain of GWI mice compared to controls, as well as increased phosphorylation of its most recognized downstream effectors: the α subunit of eukaryotic initiation factor 2 (eIF2α), IκB kinase (IKK), and the p65 subunit of nuclear factor-κB (NFκB-p65). Furthermore, we found that the increased phosphorylation level of these three proteins were suppressed in GWI mice treated with fingolimod. These results suggest that activation of PKR and NFκB signaling may be important for the regulation of cognition and neuroinflammation in the GWI condition and that fingolimod, a drug already approved for human use, may be a potential candidate for the treatment of GWI.


Subject(s)
Fingolimod Hydrochloride , Persian Gulf Syndrome , Animals , Male , Mice , Amnesia/metabolism , Disease Models, Animal , Fingolimod Hydrochloride/therapeutic use , Fingolimod Hydrochloride/metabolism , Fingolimod Hydrochloride/pharmacology , Gulf War , Memory Disorders/chemically induced , Memory Disorders/drug therapy , Memory Disorders/metabolism , Mice, Inbred C57BL , Microglia , Neuroinflammatory Diseases , NF-kappa B/metabolism , Persian Gulf Syndrome/chemically induced , Persian Gulf Syndrome/drug therapy , Persian Gulf Syndrome/metabolism , Protein Kinases/metabolism , Protein Kinases/pharmacology , Protein Kinases/therapeutic use , Pyridostigmine Bromide/therapeutic use , Pyridostigmine Bromide/pharmacology
12.
ANZ J Surg ; 93(9): 2086-2091, 2023 09.
Article in English | MEDLINE | ID: mdl-37132128

ABSTRACT

BACKGROUND: Chronic intestinal pseudo-obstruction (CIPO) may be a primary or secondary phenomenon and is often multifactorial. Treatment is largely directed at improving colonic motility. The use of cholinesterase inhibitors such as pyridostigmine has been hypothesized to increase acetylcholine in the bowel, improving symptoms and transit times. METHODS: A systematic review of the use of pyridostigmine in CIPO was conducted using scientific and commercial search engines identifying scientific studies enrolling adult human subjects, published from 2000 to 2022 in the English language. RESULTS: Four studies were identified including two randomized controlled trials (RCT) and two observational studies. The studies had heterogenous inclusion criteria, dosing regimens and reported outcomes. Two studies were identified as being at high risk of bias. All studies reported improved patient outcomes with use of pyridostigmine, and low rates (4.3%) of mild cholinergic side effects. No major side effects were reported. CONCLUSION: The use of pyridostigmine in management of CIPO is biologically plausible due to its ability to increase colonic motility, and early studies on its role are uniformly suggestive of benefit with low side-effect profile. Four clinical studies have been conducted to date, with small sample sizes, heterogeneity and high risk of bias. Further high-quality studies are required to enable assessment of pyridostigmine's utility as an effective management strategy in CIPO.


Subject(s)
Intestinal Pseudo-Obstruction , Pyridostigmine Bromide , Adult , Humans , Pyridostigmine Bromide/therapeutic use , Pyridostigmine Bromide/pharmacology , Gastrointestinal Motility , Intestinal Pseudo-Obstruction/drug therapy , Intestinal Pseudo-Obstruction/diagnosis , Cholinesterase Inhibitors/therapeutic use , Cholinesterase Inhibitors/pharmacology , Chronic Disease
13.
Int Ophthalmol ; 43(8): 2777-2785, 2023 Aug.
Article in English | MEDLINE | ID: mdl-36879110

ABSTRACT

BACKGROUND: Ocular myasthenia gravis (OMG) is an autoimmune disease which causes ptosis, diplopia, or both. It can be categorized as early or late onset, with differing presenting characteristics and prognoses. Currently, there is limited information available to compare characteristics and outcomes in onset groups in Thailand. OBJECTIVE: To describe and compare baseline characteristics and outcomes in OMG patients classified by onset groups and to investigate the factors associated with the disease, especially in terms of treatment responses classified according to the MGFA Post-Intervention Status (MGFA-PIS). METHODS: OMG patients diagnosed between January 2014 and March 2021 at Rajavithi Hospital, Thailand, were categorized into 2 groups based on age of onset, and baseline characteristics were analyzed and compared. The treatment responses of each group in terms of time to achievement of minimal manifestations (MM) were analyzed. RESULTS: Eighty-one patients (38 with early and 43 with late onset) were included, and the mean (SD) follow-up time was 35.85 months (17.25). There was no significant difference between the baseline characteristics of the two groups. A low dose of pyridostigmine was more commonly used in the early-onset group (p = 0.01), while the mean dose of corticosteroids was significantly lower in the late-onset patients (p < 0.001). We found that seropositivity of acetylcholine receptor antibody decreased the odds ratio of achievement of MM (OR 0.185, 95% CI 0.043-0.789, p = 0.023) and receiving a high dose of pyridostigmine (≥ 120 mg/day) increased the odds ratio of achieving it (OR 8.296, 95% CI 2.136-32.226, p = 0.002). CONCLUSIONS: A higher dose of pyridostigmine may be necessary for achievement of favorable treatment response. AChRAb seropositivity is a predictor for unfavorable treatment response in Thai populations.


Subject(s)
Myasthenia Gravis , Pyridostigmine Bromide , Humans , Pyridostigmine Bromide/therapeutic use , Age of Onset , Retrospective Studies , Myasthenia Gravis/diagnosis , Myasthenia Gravis/drug therapy , Myasthenia Gravis/epidemiology , Receptors, Cholinergic/therapeutic use
14.
J Oncol Pharm Pract ; 29(5): 1271-1274, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36734133

ABSTRACT

INTRODUCTION: Blepharoptosis, commonly referred to as ptosis or eyelid sagging, is a condition where the upper eyelid droops over the eye. It can be congenital or acquired and is caused by the weakening of the eyelid muscles. CASE REPORT: We present a case of a 3-year-old boy with T-cell acute lymphoblastic leukemia who developed bilateral ptosis while on treatment with Berlin-Frankfurt Munster-98 protocol. MANAGEMENT & OUTCOME: The patient was diagnosed with bilateral ptosis due to vincristine, the primary agent in the induction phase of the protocol. The addition of the neuroregenerative agents and supportive measures led to marked improvement, followed by complete resolution within 3 weeks. DISCUSSION: Vincristine is an anticancer agent with known neurotoxicity, which has a significant role in treating hematological malignancies and sarcoma. In many studies, the addition of neuroregenerative agents such as pyridoxine and pyridostigmine has been noted to hasten recovery without any documented side effects. Similar findings were also drawn from our research due to India's higher incidence of vincristine-induced neurotoxicity. It is essential to promptly diagnose and manage symptoms at the earliest to prevent the risk of permanent nerve damage and inferior quality of life for the patient.


Subject(s)
Blepharoptosis , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Male , Humans , Child, Preschool , Vincristine/adverse effects , Blepharoptosis/chemically induced , Blepharoptosis/diagnosis , Blepharoptosis/drug therapy , Pyridostigmine Bromide/therapeutic use , Pyridoxine/therapeutic use , Quality of Life , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy
15.
Acta Neurol Belg ; 123(2): 375-384, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36658451

ABSTRACT

INTRODUCTION: As new treatments are becoming available for patients with myasthenia gravis (MG), it is worth reflecting on the actual status of MG treatment to determine which patients would most likely benefit from the new treatments. METHODS: We reviewed the clinical files of all MG patients seen at the Department of Neurology of the Antwerp University Hospital during the years 2019, 2020 and 2021. RESULTS: 163 patients were included. Age at diagnosis varied from the first to the eighth decades, with a peak of incidence from 60 to 70 years for both genders, and an additional peak from 20 to 30 years in women. Diplopia and ptosis were by far the most common onset symptom. At maximum disease severity, 24% of the patients still had purely ocular symptoms and 4% needed mechanical ventilation. 97% of the patients received a treatment with pyridostigmine and 68% with corticosteroids, often in combination with immunosuppressants. More than half reported side effects. At the latest visit, 50% of the patients were symptom-free. Also, half of the symptomatic patients were fulltime at work or retired with no or mild limitations in daily living. The remaining patients were working part-time, on sick leave, or retired with severe limitations. DISCUSSION AND CONCLUSION: The majority of MG patients are doing well with currently available treatments, but often at the cost of side effects in the short and in the long term. A significant group is in need of better treatments.


Subject(s)
Blepharoptosis , Myasthenia Gravis , Humans , Female , Male , Belgium , Myasthenia Gravis/diagnosis , Myasthenia Gravis/drug therapy , Myasthenia Gravis/epidemiology , Pyridostigmine Bromide/therapeutic use , Blepharoptosis/drug therapy , Diplopia/drug therapy
16.
Thorac Cancer ; 14(5): 517-523, 2023 02.
Article in English | MEDLINE | ID: mdl-36594520

ABSTRACT

OBJECTIVE: To study the influencing factors of myasthenic crisis in patients with myasthenia gravis during perioperative period. METHODS: A total of 564 myasthenia gravis (MG) patients who underwent standard expanded resection of thymoma/thymoma in the Department of Thoracic Surgery of Beijing Hospital from January 2011 to March 2022 were retrospectively included in the study. Clinical indicators such as gender, age, thymoma, American Society of Anesthesiologists (ASA) score, operation time, intraoperative blood loss, and some others were recorded. RESULTS: Osserman-stages IIB + III + IV (odds ratio [OR] 16.091, 95% confidence interval [CI] 5.170-50.076, p value < 0.001), the dosage of pyridostigmine bromide more than 240 mg (OR 6.462, 95% CI 3.110-13.427, p value < 0.001), ASA score 2 and 3 (OR 3.203, 95% CI 1.461-7.020, p value = 0.004), low diffusion lung capacity for carbon monoxide (DLCO%) (OR 0.981, 95% CI 0.963-1.000 p value = 0.049), and blood loss greater than 1000 ml (OR 16.590, 95% CI 1.911-144.011, p value = 0.011) were independent risk factors for myasthenic crisis. CONCLUSIONS: Patients with poor Osserman stages, higher preoperative dosage of pyridostigmine bromide, higher ASA score, poor pulmonary function (low DLCO%), and more intraoperative bleeding should be highly vigilant for the occurrence of postoperative myasthenic crisis.


Subject(s)
Myasthenia Gravis , Thymoma , Thymus Neoplasms , Humans , Thymoma/surgery , Pyridostigmine Bromide/therapeutic use , Retrospective Studies , Thymectomy/adverse effects , Postoperative Complications/epidemiology , Postoperative Complications/etiology , Myasthenia Gravis/surgery , Thymus Neoplasms/surgery
17.
Hum Mol Genet ; 32(9): 1552-1564, 2023 04 20.
Article in English | MEDLINE | ID: mdl-36611016

ABSTRACT

Congenital myasthenic syndrome (CMS) is a heterogeneous condition associated with 34 different genes, including SLC5A7, which encodes the high-affinity choline transporter 1 (CHT1). CHT1 is expressed in presynaptic neurons of the neuromuscular junction where it uses the inward sodium gradient to reuptake choline. Biallelic CHT1 mutations often lead to neonatal lethality, and less commonly to non-lethal motor weakness and developmental delays. Here, we report detailed biochemical characterization of two novel mutations in CHT1, p.I294T and p.D349N, which we identified in an 11-year-old patient with a history of neonatal respiratory distress, and subsequent hypotonia and global developmental delay. Heterologous expression of each CHT1 mutant in human embryonic kidney cells showed two different mechanisms of reduced protein function. The p.I294T CHT1 mutant transporter function was detectable, but its abundance and half-life were significantly reduced. In contrast, the p.D349N CHT1 mutant was abundantly expressed at the cell membrane, but transporter function was absent. The residual function of the p.I294T CHT1 mutant may explain the non-lethal form of CMS in this patient, and the divergent mechanisms of reduced CHT1 function that we identified may guide future functional studies of the CHT1 myasthenic syndrome. Based on these in vitro studies that provided a diagnosis, treatment with cholinesterase inhibitor together with physical and occupational therapy significantly improved the patient's strength and quality of life.


Subject(s)
Mutant Proteins , Mutation , Myasthenic Syndromes, Congenital , Symporters , Myasthenic Syndromes, Congenital/drug therapy , Myasthenic Syndromes, Congenital/genetics , Myasthenic Syndromes, Congenital/metabolism , Myasthenic Syndromes, Congenital/rehabilitation , Humans , Male , Child , HEK293 Cells , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Half-Life , Cell Membrane/metabolism , Protein Transport , Staurosporine/pharmacology , Pyridostigmine Bromide/therapeutic use , Quality of Life , Symporters/chemistry , Symporters/genetics , Symporters/metabolism
18.
J Cardiothorac Surg ; 18(1): 20, 2023 Jan 12.
Article in English | MEDLINE | ID: mdl-36635776

ABSTRACT

OBJECTIVE: To study the influencing factors of myasthenic crisis in non-thymoma myasthenia gravis (MG) patients during perioperative period. METHODS: We retrospectively analyzed a total of 387 non-thymoma MG patients who underwent extended thymoma resection in the Department of Thoracic Surgery of Beijing Hospital from February 2011 to December 2021, recorded ASA score, Osserman classification, preoperative course, pyridostigmine dosage, operation method, operation time, and intraoperative blood loss, then analyzed the factors associated with postoperative myasthenic crisis by univariate and multivariate logistic regression. RESULTS: Osserman classification IIB + III + IV (P < 0.001), history of myasthenic crisis (P = 0.013), pyridostigmine dosage greater than 240 (P < 0.001), ASA score 2 and 3 (P = 0.001) are independent risk factors for myasthenic crisis. CONCLUSION: Patients with poor Osserman classification, history of myasthenic crisis before surgery, larger preoperative dosage of pyridostigmine, and higher ASA scores should be highly alert to the occurrence of postoperative myasthenic crisis.


Subject(s)
Myasthenia Gravis , Thymoma , Thymus Neoplasms , Humans , Pyridostigmine Bromide/therapeutic use , Retrospective Studies , Thymectomy/adverse effects , Thymectomy/methods , Postoperative Complications/etiology , Myasthenia Gravis/complications , Myasthenia Gravis/surgery , Thymoma/complications , Thymoma/surgery , Thymus Neoplasms/complications , Thymus Neoplasms/surgery
19.
Photodiagnosis Photodyn Ther ; 41: 103240, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36592783

ABSTRACT

BACKGROUND: Although the mechanism is not clear, the inability of the orbicularis oculi muscle, especially the deeper segment (Horner muscle), is thought to be responsible in epiphora. This study evaluates the effect of the anticholinergic drug pyridostigmine (Mestinon) in patients with patent but dysfunctional lacrimal drainage system. MATERIAL AND METHODS: Twenty patients with bilateral epiphora (mean age:60.78 ± 6.49 yrs) were included in this study. Patients with a patent lacrimal irrigation test based on persistent and symptomatic epiphora, wiping >10 times daily or continuous tearing and grade 4-5 epiphora according to Munk scale, showing neuropathic involvement in the orbicularis oculi muscle by the quantitative motor unit potential (MUP) analysis method were evaluated prospectively. Fluorescein dye disappearance test (a semi-quantitative assessment of delayed tear outflow) together with a Schirmer test reading were performed in order to detect dry eye. The patients were evaluated for tear meniscus measurements by anterior segment optical coherence topography (OCT) and non-invasive tear break-up time (NI-BUT) was measured by Oculus Keratograph 5 M. Those with a NI-BUT value above 10 s, without eyelid laxity, previous ocular surgery or ocular surface disease, or nasolacrimal duct obstruction, and who agreed to use the drug were included in the study. Each subject underwent OCT measurements of the lower tear meniscus of both eyes before and 15 mins after taking Mestinon (1 × 60 mg tablet). Upon measurement of the positive effect of the drug on tear meniscus height (TMH), the patients were asked to continue this regime daily for 1 month and then evaluated for relief in their epiphora complaints and any systemic drug side effects. RESULTS: A total of 20 patients (40 eyes) with bilateral epiphora were included in the study. All eyes had grade 4 Munk-score epiphora, Schirmer's test was within the normal range in all eyes (mean, 14 ± 4 mm), and patent lacrimal irrigation test. The lower mean TMH reductions 15 min after Mestinon in the right and left eyes were 135.41 ± 85.47 and 55.44 ± 61.56 mm, respectively, a statistically significant decrease in both eyes (p = 0.001, p < 0.01). The mean tear meniscus area (TMA) in the right and left eyes was 131.83 ± 68.27 mm2 and 62.72 ± 50.57 mm2, respectively; 15 mins after administration of Mestinon, the mean TMA in the right and left eyes was 77.27 ± 48.34 and 59.18 ± 44.74 mm2, respectively (p = 0.001, p < 0.01). The mean decreases of 54.56 ± 39.34 mm2 in the right eye area and 3.53 ± 42.32 mm2 in the left eye area were statistically significant (p = 0.041, p < 0.05). CONCLUSION: Symptomatic relief for epiphora cannot be achieved with known treatment options due to lacrimal pump dysfunction. We found that pyridostigmine (Mestinon) provided relief in patients' complaints of epiphora consistent with a significant reduction in TMH levels.


Subject(s)
Lacrimal Duct Obstruction , Nasolacrimal Duct , Photochemotherapy , Humans , Middle Aged , Aged , Pyridostigmine Bromide/therapeutic use , Tomography, Optical Coherence/methods , Photochemotherapy/methods , Photosensitizing Agents
20.
Eur J Med Genet ; 66(3): 104706, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36669590

ABSTRACT

Disease causing variants in the Ryanodine receptor 1 (RYR1) gene are a common cause for congenital myopathy and for malignant hyperthermia susceptibility. We report a 17 year old boy with congenital muscle weakness progressing to a myasthenia like myopathy with muscle weakness, fatigability, ptosis, and ophthalmoplegia. Muscle biopsy showed predominance and atrophy of type 1 fibers. Whole-exome trio sequencing revealed three variants in the RYR1-gene in the patient: c.6721C > T,p.(Arg2241*) and c.2122G > A,p.(Asp708Asn) in cis position, and the c.325C > T,p.(Arg109Trp) variant in trans. Treatment with pyridostigmine improved symptoms. This case supports that a myasthenia like phenotype is part of the phenotypic spectrum of RYR1 related disorders, and that treatment with pyridostigmine can be beneficial for patients with this phenotype.


Subject(s)
Muscular Diseases , Pyridostigmine Bromide , Adolescent , Humans , Male , Muscle Weakness/genetics , Muscle, Skeletal/pathology , Muscular Diseases/genetics , Mutation , Phenotype , Pyridostigmine Bromide/therapeutic use , Ryanodine Receptor Calcium Release Channel/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...