Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.970
Filter
1.
Zh Nevrol Psikhiatr Im S S Korsakova ; 124(7. Vyp. 2): 72-78, 2024.
Article in Russian | MEDLINE | ID: mdl-39175243

ABSTRACT

OBJECTIVE: To evaluate the effect of discontinuation or prolongation of DMT on the activity of the disease during pregnancy and in the postpartum period in patients with aggressive MS from the Moscow region. MATERIAL AND METHODS: The study included female patients with an aggressive course of MS receiving DMT at the time of pregnancy. The patients were followed-up for the period 2016 to February 2024. RESULTS: There were 17 cases of pregnancy during natalizumab (NZ) therapy; discontinuation of therapy in the first trimester of pregnancy provoked a resumption of disease activity in half of the patients. There were no exacerbations in patients whose therapy was prolonged until the 34th week of pregnancy. In 5 patients receiving fingolimod (FGL), therapy was discontinued upon the establishment of pregnancy, which caused the resumption of disease activity in three out of 5 cases. In 3 patients receiving anti-B-cell therapy, pregnancy occurred within a few months after the next infusion, there were no exacerbations during pregnancy. CONCLUSION: The cancellation of NS therapy in the early stages of pregnancy in most cases leads to the resumption of disease activity during pregnancy. Exacerbations in the postpartum period also correlated with early discontinuation of therapy and with a long period before the restart of infusions. Prolongation of infusions to 30-34 weeks of pregnancy contributed to stabilization of the condition throughout the perinatal period. Discontinuation of FGL therapy at the onset of pregnancy increased the risk of repeated relapses of the disease, up to the development of inflammatory immune restoration syndrome during pregnancy and contributed to the increase in disability in the postpartum period.


Subject(s)
Fingolimod Hydrochloride , Multiple Sclerosis , Natalizumab , Pregnancy Complications , Humans , Female , Pregnancy , Moscow/epidemiology , Adult , Pregnancy Complications/drug therapy , Natalizumab/therapeutic use , Multiple Sclerosis/drug therapy , Fingolimod Hydrochloride/therapeutic use , Postpartum Period , Immunosuppressive Agents/therapeutic use , Young Adult
2.
Mult Scler ; 30(9): 1163-1175, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39087208

ABSTRACT

BACKGROUND: Comparisons between cladribine and other potent immunotherapies for multiple sclerosis (MS) are lacking. OBJECTIVES: To compare the effectiveness of cladribine against fingolimod, natalizumab, ocrelizumab and alemtuzumab in relapsing-remitting MS. METHODS: Patients with relapsing-remitting MS treated with cladribine, fingolimod, natalizumab, ocrelizumab or alemtuzumab were identified in the global MSBase cohort and two additional UK centres. Patients were followed for ⩾6/12 and had ⩾3 in-person disability assessments. Patients were matched using propensity score. Four pairwise analyses compared annualised relapse rates (ARRs) and disability outcomes. RESULTS: The eligible cohorts consisted of 853 (fingolimod), 464 (natalizumab), 1131 (ocrelizumab), 123 (alemtuzumab) or 493 (cladribine) patients. Cladribine was associated with a lower ARR than fingolimod (0.07 vs. 0.12, p = 0.006) and a higher ARR than natalizumab (0.10 vs. 0.06, p = 0.03), ocrelizumab (0.09 vs. 0.05, p = 0.008) and alemtuzumab (0.17 vs. 0.04, p < 0.001). Compared to cladribine, the risk of disability worsening did not differ in patients treated with fingolimod (hazard ratio (HR) 1.08, 95% confidence interval (CI) 0.47-2.47) or alemtuzumab (HR 0.73, 95% CI 0.26-2.07), but was lower for patients treated with natalizumab (HR 0.35, 95% CI 0.13-0.94) and ocrelizumab (HR 0.45, 95% CI 0.26-0.78). There was no evidence for a difference in disability improvement. CONCLUSION: Cladribine is an effective therapy that can be viewed as a step up in effectiveness from fingolimod, but is less effective than the most potent intravenous MS therapies.


Subject(s)
Alemtuzumab , Antibodies, Monoclonal, Humanized , Cladribine , Fingolimod Hydrochloride , Immunosuppressive Agents , Multiple Sclerosis, Relapsing-Remitting , Natalizumab , Humans , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Female , Male , Cladribine/therapeutic use , Cladribine/adverse effects , Alemtuzumab/adverse effects , Alemtuzumab/therapeutic use , Fingolimod Hydrochloride/therapeutic use , Fingolimod Hydrochloride/adverse effects , Adult , Natalizumab/therapeutic use , Natalizumab/adverse effects , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Humanized/adverse effects , Immunosuppressive Agents/therapeutic use , Immunosuppressive Agents/adverse effects , Middle Aged , Immunologic Factors/adverse effects , Treatment Outcome
3.
FASEB J ; 38(15): e23872, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39126272

ABSTRACT

Paclitaxel is among the most active chemotherapy drugs for the aggressive triple negative breast cancer (TNBC). Unfortunately, it often induces painful peripheral neuropathy (CIPN), a major debilitating side effect. Here we demonstrate that in naive and breast tumor-bearing immunocompetent mice, a clinically relevant dose of FTY720/Fingolimod that targets sphingosine-1-phosphate receptor 1 (S1PR1), alleviated paclitaxel-induced neuropathic pain. FTY720 also significantly attenuated paclitaxel-stimulated glial fibrillary acidic protein (GFAP), a marker for activated astrocytes, and expression of the astrocyte-secreted synaptogenic protein Sparcl1/Hevin, a key regulator of synapse formation. Notably, the formation of excitatory synapses containing VGluT2 in the spinal cord dorsal horn induced by paclitaxel was also inhibited by FTY720 treatment, supporting the involvement of astrocytes and Sparcl1 in CIPN. Furthermore, in this TNBC mouse model that mimics human breast cancer, FTY720 administration also enhanced the anti-tumor effects of paclitaxel, leading to reduced tumor progression and lung metastasis. Taken together, our findings suggest that targeting the S1P/S1PR1 axis with FTY720 is a multipronged approach that holds promise as a therapeutic strategy for alleviating both CIPN and enhancing the efficacy of chemotherapy in TNBC treatment.


Subject(s)
Fingolimod Hydrochloride , Neuralgia , Paclitaxel , Animals , Fingolimod Hydrochloride/pharmacology , Paclitaxel/pharmacology , Neuralgia/chemically induced , Neuralgia/drug therapy , Neuralgia/metabolism , Neuralgia/pathology , Mice , Female , Calcium-Binding Proteins/metabolism , Calcium-Binding Proteins/genetics , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Astrocytes/metabolism , Astrocytes/drug effects , Extracellular Matrix Proteins/metabolism , Extracellular Matrix Proteins/genetics , Cell Line, Tumor , Sphingosine-1-Phosphate Receptors/metabolism , Humans , Disease Progression , Antineoplastic Agents, Phytogenic/pharmacology , Glial Fibrillary Acidic Protein/metabolism , Glial Fibrillary Acidic Protein/genetics
4.
J Integr Neurosci ; 23(8): 142, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39207074

ABSTRACT

BACKGROUND: Clinically, ischemic reperfusion injury is the main cause of stroke injury. This study aimed to assess the effectiveness of fingolimod in suppressing inflammation caused by ischemic brain injury and explore its pharmacological mechanisms. METHODS: In total, 75 male Sprague-Dawley rats were randomly and equally assigned to five distinct groups: sham, middle cerebral artery occlusion/reperfusion (MCAO/R) surgery, fingolimod low-dose (F-L), fingolimod medium-dose (F-M), and fingolimod high-dose (F-H). Neurobehavioral tests, 2,3,5-triphenyltetrazolium chloride staining, and the brain tissue drying-wet method were conducted to evaluate neurological impairment, cerebral infarction size, and brain water content. Enzyme-linked immunosorbent assay was employed to quantify pro-inflammatory cytokines interleukin (IL)-1ß, IL-6, and tumor necrosis factor-alpha (TNF-α) protein levels. Western blotting and immunohistochemical staining were performed to assess high mobility group box 1 (HMGB1), toll-like receptor 4 (TLR4), and nuclear factor kappa-B p65 (NF-κBp65) levels. RESULTS: Rats in the F-L, F-M, and F-H groups exhibited lower Longa scores, reduced infarction volumes, and decreased brain edema than those in the MCAO/R group. Additionally, the F-L, F-M, and F-H groups exhibited lower serum levels of IL-1ß, IL-6, and TNF-α than those of the MCAO/R group. Additionally, F-L, F-M, and F-H treatments resulted in decreased HMGB1, TLR4, and NF-κBp65 protein expression levels in the hippocampus of MCAO/R rats. CONCLUSIONS: Fingolimod was found to reduce ischemic brain injury in a dose-dependent manner. Moreover, it was also found to alleviate inflammation following ischemic brain injury via the HMGB1/TLR4/NF­κB signaling pathway.


Subject(s)
Brain Ischemia , Fingolimod Hydrochloride , HMGB1 Protein , NF-kappa B , Rats, Sprague-Dawley , Signal Transduction , Toll-Like Receptor 4 , Animals , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 4/drug effects , HMGB1 Protein/metabolism , HMGB1 Protein/drug effects , Male , Fingolimod Hydrochloride/pharmacology , Fingolimod Hydrochloride/administration & dosage , Rats , Signal Transduction/drug effects , Signal Transduction/physiology , Brain Ischemia/drug therapy , Brain Ischemia/metabolism , NF-kappa B/metabolism , NF-kappa B/drug effects , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/complications , Inflammation/drug therapy , Inflammation/metabolism , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/etiology , Disease Models, Animal , Reperfusion Injury/drug therapy , Reperfusion Injury/metabolism
5.
Front Immunol ; 15: 1416464, 2024.
Article in English | MEDLINE | ID: mdl-39076966

ABSTRACT

Introduction: Disease-modifying therapies (DMTs) have been shown to improve disease outcomes in multiple sclerosis (MS) patients. They may also impair the immune response to vaccines, including the SARS-CoV-2 vaccine. However, available data on both the intrinsic immune effects of DMTs and their influence on cellular response to the SARS-CoV-2 vaccine are still incomplete. Methods: Here, we evaluated the immune cell effects of 3 DMTs on the response to mRNA SARS-CoV-2 vaccination by comparing MS patients treated with one specific therapy (fingolimod, dimethyl fumarate, or natalizumab) with both healthy controls and untreated patients. We profiled 23 B-cell traits, 57 T-cell traits, and 10 cytokines, both at basal level and after stimulation with a pool of SARS-CoV-2 spike peptides, in 79 MS patients, treated with DMTs or untreated, and 32 healthy controls. Measurements were made before vaccination and at three time points after immunization. Results and Discussion: MS patients treated with fingolimod showed the strongest immune cell dysregulation characterized by a reduction in all measured lymphocyte cell classes; the patients also had increased immune cell activation at baseline, accompanied by reduced specific immune cell response to the SARS-CoV-2 vaccine. Also, anti-spike specific B cells progressively increased over the three time points after vaccination, even when antibodies measured from the same samples instead showed a decline. Our findings demonstrate that repeated booster vaccinations in MS patients are crucial to overcoming the immune cell impairment caused by DMTs and achieving an immune response to the SARS-CoV-2 vaccine comparable to that of healthy controls.


Subject(s)
COVID-19 Vaccines , COVID-19 , Dimethyl Fumarate , Fingolimod Hydrochloride , Multiple Sclerosis , SARS-CoV-2 , Humans , COVID-19/immunology , COVID-19/prevention & control , Male , Multiple Sclerosis/immunology , Multiple Sclerosis/drug therapy , Female , SARS-CoV-2/immunology , COVID-19 Vaccines/immunology , Middle Aged , Adult , Fingolimod Hydrochloride/therapeutic use , Fingolimod Hydrochloride/pharmacology , Dimethyl Fumarate/therapeutic use , Dimethyl Fumarate/pharmacology , Immunosuppressive Agents/therapeutic use , Natalizumab/therapeutic use , B-Lymphocytes/immunology , Vaccination , T-Lymphocytes/immunology , Cytokines/metabolism
6.
Mult Scler ; 30(8): 1026-1035, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39054846

ABSTRACT

INTRODUCTION: One strategy to mitigate progressive multifocal leukoencephalopathy (PML) risk is to switch to other highly effective disease-modifying therapies (DMTs). However, the optimal switch DMT following natalizumab (NTZ) discontinuation is yet to be determined. OBJECTIVE: The objective of the study is to determine the most effective and tolerable DMTs to switch to following NTZ discontinuation due to John Cunningham virus (JCV) antibody positivity. METHODS: This is a multicenter observational cohort study that included all stable relapsing-remitting multiple sclerosis (MS) patients who were treated with NTZ for at least 6 months before switching therapy due to JCV antibody positivity. RESULTS: Of 321 patients, 255 switched from NTZ to rituximab/ocrelizumab, 52 to fingolimod, and 14 to alemtuzumab, with higher annualized relapse rate (ARR) in fingolimod switchers (0.193) compared with rituximab/ocrelizumab or alemtuzumab (0.028 and 0.032, respectively). Fingolimod switchers also had increased disability progression (p = 0.014) and a higher proportion developed magnetic resonance imaging (MRI) lesions compared with rituximab/ocrelizumab (62.9% vs. 13.0%, p < 0.001, and 66.6% vs. 24.0%, p < 0.001, respectively). Mean drug survival favored rituximab/ocrelizumab or alemtuzumab over fingolimod (p < 0.001). CONCLUSION: Our study shows superior effectiveness of rituximab/ocrelizumab and alemtuzumab compared with fingolimod in stable patients switching from NTZ due to JC virus antibody positivity.


Subject(s)
Immunologic Factors , JC Virus , Leukoencephalopathy, Progressive Multifocal , Multiple Sclerosis, Relapsing-Remitting , Natalizumab , Humans , Natalizumab/therapeutic use , Natalizumab/adverse effects , Female , Adult , Male , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Immunologic Factors/adverse effects , Leukoencephalopathy, Progressive Multifocal/chemically induced , JC Virus/immunology , Middle Aged , Drug Substitution , Rituximab/adverse effects , Rituximab/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Humanized/adverse effects , Fingolimod Hydrochloride/therapeutic use , Alemtuzumab/adverse effects , Alemtuzumab/therapeutic use
7.
BMC Neurol ; 24(1): 255, 2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39048953

ABSTRACT

BACKGROUND: Multiple Sclerosis (MS) a central nervous system autoimmune disorder, mainly affecting young adults and more prevalent among women, can lead to sexual dysfunction (SD) among both males and females with MS. Female sexual dysfunction can be defined as dyspareunia, a lack of sexual desire, disorders in the arousal and orgasm phases, and sexual pain disorders. The purpose of this study is to investigate the changes in sexual function among females with MS whose treatment was switched from first-line injectable medications to other agents after a six-month duration. And assess the changes in all three domains of SD. METHODS: In this longitudinal study females diagnosed with MS, aged between 18 and 50 years old, and were candidates for switching their treatment from interferon beta-1a (intra-muscular and subcutaneous), and Glatiramer Acetate (GA), to Fingolimod, Dimethyl Fumarate (DMF), or Natalizumab (NTZ) due to patients' convenience and tolerability and adverse events were included. "Multiple Sclerosis Intimacy and Sexuality Questionnaire-19" was used to evaluate the SD changes before and six months after the new treatment initiation. Statistical analysis was conducted using SPSS V.24 software. Histograms and the Shapiro-Wilk test were used to assess the normality of the variables; due to the non-normal distribution of quantitative variables (except for age), the Wilcoxon signed-rank test was used to compare the scores, before and six months after the medication change. The level of significance was considered less than 0.05. RESULTS: Out of 107 female participants (average age: 35.09 ± 5.61), The mean of overall MSISQ-19 scores, before and six months after the medication change were not significant (p-value = 0.091). However, considering the subdomains, the medication changes only affected the tertiary subdomain of MSISQ-19 (p-value = 0.017). Still, the scores of other subdomains did not change significantly (p-value = 0.761 for primary SD and 0.479 for secondary SD). Also, there wasn't any significant difference between EDSS before and after the medication change (p-value = 0.461). CONCLUSIONS: To our knowledge, this was the first study, assessing the effect of MS medication change on the improvement of SD among patients. According to the results of the presented cross-sectional study, we found that during a six-month period, the tertiary subdomain of MSISQ-19 symptoms improved significantly, while the changes in other SD domains were not significant.


Subject(s)
Glatiramer Acetate , Multiple Sclerosis , Sexual Dysfunction, Physiological , Humans , Female , Adult , Multiple Sclerosis/drug therapy , Multiple Sclerosis/complications , Middle Aged , Sexual Dysfunction, Physiological/etiology , Sexual Dysfunction, Physiological/drug therapy , Longitudinal Studies , Glatiramer Acetate/administration & dosage , Glatiramer Acetate/therapeutic use , Young Adult , Immunologic Factors/administration & dosage , Immunologic Factors/therapeutic use , Dimethyl Fumarate/administration & dosage , Dimethyl Fumarate/therapeutic use , Adolescent , Immunosuppressive Agents/therapeutic use , Immunosuppressive Agents/administration & dosage , Interferon beta-1a/administration & dosage , Interferon beta-1a/therapeutic use , Drug Substitution/methods , Fingolimod Hydrochloride/therapeutic use , Fingolimod Hydrochloride/administration & dosage , Natalizumab/administration & dosage , Natalizumab/therapeutic use
8.
Article in English | MEDLINE | ID: mdl-39052026

ABSTRACT

Fingolimod is a sphingosine-1-phosphate receptor modulator used to treat multiple sclerosis. While fingolimod has been associated with an increased risk of cryptococcal meningitis, its correlation with other deep mycoses remains unclear. In this study, we conducted a scoping review of fingolimod associated with histoplasmosis, based on a case report, a literature review, and data from the FDA Adverse Events Reporting System (FAERS) as of January 24th, 2023. A 30-year-old Brazilian woman diagnosed with relapsing-remitting multiple sclerosis, receiving a daily dose of 0.5 mg of fingolimod, presented with a two-month history of fever and unintended weight loss, accompanied by lymphadenopathy, splenomegaly, and lung involvement was investigated. Biopsy of a lung nodule revealed fungal structures suggestive of Histoplasma sp. Additionally, serological testing yielded positive for Histoplasma capsulatum. Disseminated histoplasmosis should be considered in the differential diagnosis of febrile syndromes in patients undergoing fingolimod therapy for multiple sclerosis, particularly in the Americas, where this mycosis is endemic. Treatment with itraconazole and modification of immunotherapy can achieve excellent clinical outcomes.


Subject(s)
Fingolimod Hydrochloride , Histoplasmosis , Multiple Sclerosis, Relapsing-Remitting , Humans , Histoplasmosis/drug therapy , Histoplasmosis/diagnosis , Fingolimod Hydrochloride/adverse effects , Female , Adult , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Antifungal Agents/adverse effects , Antifungal Agents/therapeutic use , Immunosuppressive Agents/adverse effects , Histoplasma
9.
BMJ Case Rep ; 17(7)2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39059796

ABSTRACT

This case report details a female patient with multiple sclerosis in her 30s, who experienced a significant fingolimod rebound syndrome post partum, characterised by worsening neurological symptoms and severe demyelinating lesions. Traditional treatments, including steroids and plasmapheresis, were ineffective. However, the introduction of intravenous immunoglobulin (IVIG) led to remarkable improvement in her symptoms and disability status. This case highlights the complex immunological changes associated with fingolimod cessation and underscores IVIG's potential as a valuable treatment in managing such rebounds.


Subject(s)
Fingolimod Hydrochloride , Immunoglobulins, Intravenous , Immunosuppressive Agents , Postpartum Period , Humans , Female , Fingolimod Hydrochloride/therapeutic use , Immunoglobulins, Intravenous/therapeutic use , Adult , Immunosuppressive Agents/therapeutic use , Multiple Sclerosis/drug therapy , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Treatment Outcome
10.
Mult Scler Relat Disord ; 88: 105757, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38972107

ABSTRACT

BACKGROUND: This study investigates the gap in understanding the dynamics of recurring disease activity (RDA) in RRMS patients after fingolimod (FGL) treatment discontinuation. The aim is to investigate RDA in RRMS patients after stopping FGL, aiming to improve management and comprehension of disease progression post-treatment. METHODS: In this multicenter, retrospective study, data from 172 of 944 RRMS patients aged 18-55, across nine centers in Turkey, who discontinued FGL treatment, were analyzed. The collected data included EDSS scores, annualized relapse rates (ARR), lymphocyte counts, and MRI findings, with follow-up assessments conducted at 6 months, 1 year, and up to 2 years. RESULTS: RDA was observed in 31.9 % of patients, with incidences of rebound and reactivation at 20.3 % and 11.6 %, respectively. Factors like younger age, longer treatment duration, lower lymphocyte counts, and higher lesion burden increased RDA risk. Notably, 52.9 % of pregnant patients experienced RDA (16.4 % of the overall RDA group), with rebound occurring in six and reactivation in three. Patients with RDA had longer medication-free intervals and increased ARR. Discontinuation reasons varied, with disease progression linked to a lower RDA risk. CONCLUSION: Findings highlight the necessity for personalized management and vigilant monitoring after FGL discontinuation in RRMS patients, offering critical insights into RDA risk factors, and the complex interplay between treatment cessation, pregnancy, and disease progression.


Subject(s)
Disease Progression , Fingolimod Hydrochloride , Immunosuppressive Agents , Multiple Sclerosis, Relapsing-Remitting , Recurrence , Humans , Adult , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Multiple Sclerosis, Relapsing-Remitting/diagnostic imaging , Multiple Sclerosis, Relapsing-Remitting/physiopathology , Female , Male , Retrospective Studies , Young Adult , Middle Aged , Adolescent , Fingolimod Hydrochloride/administration & dosage , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/therapeutic use , Turkey , Pregnancy , Follow-Up Studies , Magnetic Resonance Imaging
11.
Neurosciences (Riyadh) ; 29(3): 184-189, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38981628

ABSTRACT

OBJECTIVES: To assess clinicians' adherence to fingolimod's effective use according to the prescribed recommendations to reduce safety risk, identify the consequences, and highlight areas for improvement to policy makers for the benefit of both patient and care-giver. METHODS: A retrospective observational study conducted at a tertiary hospital targeting multiple sclerosis patients on fingolimod from January 2017 to December 2021. The physicians' adherence to the manufacturer's instructions was assessed and categorized into good, moderate, and poor based on adherence to fingolimod instructions and monitoring measures. Four monitoring measures were assessed: bradycardia observation, ophthalmic examination, liver enzymes, and infections. In addition, the impact of adherence on patient safety was also assessed. RESULTS: A total of 140 patients were included. Seventy-twopatients (51.4%) had physician with poor adherence (followed only one instruction or none). Sixty-five patients (46.4%) had 2-3 manufacture recommendations where physician's adherence was moderate. Three patients (2.10%) had all manufacturer's recommendations. In terms of fingolimod complications, 18 patients found to have bradycardia after the first does, macular oedema and infections was reported in 4 patients, and the elevation in hepatic enzymes was reported in 6 patients. Poor physician's adherence has resulted in treatment incompleteness and highest fingolimod discontinuation or switching to other treatment options. CONCLUSION: Adherence to fingolimod instructions was poor among physicians which resulted in highest drug switching or discontinuing rate.


Subject(s)
Fingolimod Hydrochloride , Immunosuppressive Agents , Multiple Sclerosis , Patient Safety , Humans , Fingolimod Hydrochloride/therapeutic use , Fingolimod Hydrochloride/adverse effects , Female , Male , Retrospective Studies , Adult , Immunosuppressive Agents/therapeutic use , Immunosuppressive Agents/adverse effects , Multiple Sclerosis/drug therapy , Middle Aged , Guideline Adherence/statistics & numerical data
12.
J Neurol Sci ; 463: 123132, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39002188

ABSTRACT

The most prevalent disease course of Multiple Sclerosis (MS) is relapsing remitting multiple sclerosis (RRMS). Fingolimod (Gilenya®) was the first oral disease-modifying therapy to RRMS. Patients affected by MS require long-term treatment, making the ongoing evaluation of the safety profile of fingolimod imperative. The aim of this study was to analyze the post-marketing pharmacovigilance data of fingolimod in Europe. Data of 12-year period (1 January 2011-19 June 2023) were obtained from EudraVigilance, and a descriptive analysis using drug-reaction pairs was performed. A total of 22,957 reports were collected. The most reported adverse events (AEs) were related to nervous system disorders SOC (multiple sclerosis relapse n = 2271; 3.51%, headache n = 921; 1.42%, central nervous system lesion n = 893; 1,38%, dizziness 769; 1,19%, hypoaesthesia 487; 0.75% and multiple sclerosis 449; 0.69%), followed by investigations (lymphocyte count decreased n = 1648; 2.55%, white blood cell count decreased n = 833; 1.29%), blood and lymphatic system disorder (lymphopenia n = 1146; 1.77%), and general disorders and administration site condition (fatigue n = 1106; 1.71%, gait disturbance 564; 0.87%). A percentage of 23.00% of serious adverse events (SAEs), among the most reported were multiple sclerosis relapse (n = 2271; 15.27%), macular oedema (n = 793; 5.33%), bradycardia (n = 678; 4.56%), leukopenia (n = 533; 3.58%), and multiple sclerosis (n = 449; 3.02%). Most of AEs were non-serious, some SAEs related to cardiac, ophthalmic and infectious disorders emerged: their prevalence, along with the alignment of reported AEs with existing literature, supports the overall safety of fingolimod. Considering the rare and long-term ADRs that may arise in patients chronically treated for MS, continuous pharmacovigilance remains essential.


Subject(s)
Databases, Factual , Fingolimod Hydrochloride , Immunosuppressive Agents , Multiple Sclerosis, Relapsing-Remitting , Pharmacovigilance , Humans , Fingolimod Hydrochloride/adverse effects , Fingolimod Hydrochloride/therapeutic use , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Multiple Sclerosis, Relapsing-Remitting/epidemiology , Male , Female , Adult , Immunosuppressive Agents/adverse effects , Immunosuppressive Agents/therapeutic use , Middle Aged , Europe/epidemiology , Young Adult
13.
ACS Appl Mater Interfaces ; 16(30): 38880-38892, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39016239

ABSTRACT

Post-translational modification, mitochondrial abruptions, neuroinflammation, and α-synuclein (α-Syn) aggregation are considered as major causes of Parkinson's disease (PD) pathogenesis. The recent literature highlights neuroimmune cross talk and the negative role of immune effector T (Teff) and positive regulation by regulatory T (Treg) cells in PD treatment. Herein, a strategy to endow Treg action paves the path for development of PD treatment. Thus, we explored the neuroprotective efficiency of the immunomodulator and PP2A (protein phosphatase 2) activator, FTY720 nanoparticles in in vivo experimental PD models. Repurposing of FTY720 for PD is known due to its protective effect by reducing PD and its camouflaged role in endowing EZH2-mediated epigenetic regulation of PD. EZH2-FOXP3 interaction is necessary for the neuroprotective Treg cell activity. Therefore, we synthesized FTY720 nanoparticles to improve FTY720 protective efficacy in an in vivo PD model to explore the PP2A mediated signaling. We confirmed the formation of FTY720NPs, and the results of the behavioral and protein expression study showed the significant neuroprotective efficiency of our nanoformulations. In the exploration of neuroprotective mechanism, several lines of evidence confirmed FTY720NPs mediated induction of PP2A/EZH2/FOXP3 signaling in the induction of Treg cells effect in in vivo PD treatment. In summary, our nanoformulations have novel potential to alleviate PD by inducing PP2A-induced epigenetic regulation-mediated neuroimmunomodulation at the clinical setup.


Subject(s)
Fingolimod Hydrochloride , Nanoparticles , Neuroprotective Agents , T-Lymphocytes, Regulatory , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Animals , Nanoparticles/chemistry , Mice , Neuroprotective Agents/pharmacology , Neuroprotective Agents/chemistry , Fingolimod Hydrochloride/pharmacology , Fingolimod Hydrochloride/chemistry , Fingolimod Hydrochloride/therapeutic use , Mice, Inbred C57BL , Enhancer of Zeste Homolog 2 Protein/metabolism , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Protein Phosphatase 2/metabolism , Immunomodulating Agents/pharmacology , Immunomodulating Agents/chemistry , Male , Immunologic Factors/pharmacology , Immunologic Factors/chemistry , Forkhead Transcription Factors/metabolism , Humans , Parkinsonian Disorders/drug therapy
14.
J Neurol ; 271(9): 6181-6196, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39073436

ABSTRACT

BACKGROUND AND OBJECTIVES: In multiple sclerosis (MS), MRI markers can measure the potential neuroprotective effects of fingolimod beyond its anti-inflammatory activity. In this study we aimed to comprehensively explore, in the real-word setting, whether fingolimod not only reduces clinical/MRI inflammatory activity, but also influences the progression of irreversible focal and whole brain damage in relapsing-remitting [RR] MS patients. METHODS: The "EVOLUTION" study, a 24-month observational, prospective, single-arm, multicenter study, enrolled 261 RRMS patients who started fingolimod at 32 Italian MS centers and underwent biannual neurological assessments and annual MRI evaluations. Study outcomes included the proportions of evaluable RRMS patients achieving at 24 months: (1) no new/enlarging T2-hyperintense white matter (WM) lesions and/or clinical relapses; (2) a modified classification of "No Evidence of Disease Activity 4" ("modified NEDA-4") defined as no new/enlarging T2-hyperintense WM lesions, clinical relapses, and 6-month confirmed disability progression, and a yearly percentage lateral ventricular volume change on T2-FLAIR images < 2%; (3) less than 40% of active lesions at baseline and month 12 evolving to permanent black holes (PBHs). RESULTS: At month 24, 76/160 (47.5%; 95% confidence interval [CI] = 39.8%;55.2%) RRMS patients had no clinical/MRI activity. Thirty-nine of 170 RRMS patients (22.9%; 95% CI = 16.6%;29.3%) achieved "modified NEDA-4" status. Forty-four of 72 RRMS patients (61.1%; 95% CI = 49.8%;72.4%) had less than 40% of active WM lesions evolving to PBHs. The study confirmed the established safety and tolerability profile of fingolimod. DISCUSSION: By comparing our results with those from the literature, the EVOLUTION study seems to indicate a neuroprotective effect of fingolimod, limiting inflammatory activity, brain atrophy and PBH development.


Subject(s)
Fingolimod Hydrochloride , Magnetic Resonance Imaging , Multiple Sclerosis, Relapsing-Remitting , Humans , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Multiple Sclerosis, Relapsing-Remitting/diagnostic imaging , Multiple Sclerosis, Relapsing-Remitting/pathology , Fingolimod Hydrochloride/therapeutic use , Female , Male , Adult , Middle Aged , Prospective Studies , Immunosuppressive Agents/therapeutic use , Disease Progression , Brain/diagnostic imaging , Brain/pathology , Brain/drug effects , White Matter/diagnostic imaging , White Matter/pathology , White Matter/drug effects
15.
Actas Dermosifiliogr ; 115(8): T781-T790, 2024 Sep.
Article in English, Spanish | MEDLINE | ID: mdl-38972584

ABSTRACT

The use of disease-modifying therapies (DMT) has led to a paradigm shift in the management of multiple sclerosis. A comprehensive narrative review was conducted through an extensive literature search including Medline and Google Scholar to elucidate the link between DMT and the propensity of cutaneous malignancies. Sphingosine-1-phosphate receptor modulators, such as fingolimod and siponimod are associated with a higher risk of basal cell carcinoma (BCC), but not squamous cell carcinoma, or melanoma. The associated physiopathological mechanisms are not fully understood. Alemtuzumab and cladribine show isolated associations with skin cancer. Regarding other DMT, no increased risk has ever been found. Given the evidence currently available, it is of paramount importance to advocate for necessary dermatological assessments that should be individualized to the risk profile of each patient. Nonetheless, additional prospective studies are still needed to establish efficient dermatological follow-up protocols.


Subject(s)
Carcinoma, Basal Cell , Multiple Sclerosis , Skin Neoplasms , Humans , Multiple Sclerosis/drug therapy , Multiple Sclerosis/complications , Carcinoma, Basal Cell/drug therapy , Fingolimod Hydrochloride/therapeutic use , Fingolimod Hydrochloride/adverse effects , Immunosuppressive Agents/adverse effects , Immunosuppressive Agents/therapeutic use , Alemtuzumab/adverse effects , Alemtuzumab/therapeutic use , Sphingosine 1 Phosphate Receptor Modulators/therapeutic use , Sphingosine 1 Phosphate Receptor Modulators/adverse effects , Melanoma/drug therapy , Cladribine/therapeutic use , Cladribine/adverse effects , Carcinoma, Squamous Cell/etiology , Carcinoma, Squamous Cell/chemically induced
16.
J Neuroimmunol ; 394: 578410, 2024 09 15.
Article in English | MEDLINE | ID: mdl-39067241

ABSTRACT

Anti-N-methyl-d-aspartate receptor (NMDAR) encephalitis is an autoimmune disorder. With the method of indirect immunofluorescence assay (IIF), more anti-NMDAR encephalitis patients have been discovered when its first onset. But it was rare that anti-NMDAR encephalitis overlapped with multiple sclerosis (MS) documented in literatures. Here, we present a case who initially developed anti-NMDAR encephalitis and MS. Furthermore, we concluded the characteristics of patients who were diagnosed as anti-NMDAR encephalitis overlapping with MS. Additionally, due to the relapsing process, mycophenolate mofetil and sequentially fingolimod for the treatment were taken, which subsequently led to the development of a lymphoproliferative disease in his brain and other organs. This case illustrates the complex role of immunosuppressive agents.


Subject(s)
Fingolimod Hydrochloride , Immunosuppressive Agents , Lymphoproliferative Disorders , Mycophenolic Acid , Humans , Fingolimod Hydrochloride/therapeutic use , Fingolimod Hydrochloride/adverse effects , Mycophenolic Acid/therapeutic use , Mycophenolic Acid/adverse effects , Male , Immunosuppressive Agents/therapeutic use , Immunosuppressive Agents/adverse effects , Lymphoproliferative Disorders/drug therapy , Adult , Anti-N-Methyl-D-Aspartate Receptor Encephalitis/drug therapy , Brain/diagnostic imaging , Brain/pathology , Brain/drug effects , Multiple Sclerosis/drug therapy
17.
Front Immunol ; 15: 1404316, 2024.
Article in English | MEDLINE | ID: mdl-38938576

ABSTRACT

Background: The primary treatment for acute relapses in multiple sclerosis (MS) is the intravenous administration of high-dose methylprednisolone (IVMP). However, the mechanisms through which corticosteroid treatment impacts acute neuroinflammation in people with MS (pwMS) remain not fully understood. In particular, the changes induced by glucocorticoids (GCs) on cells of the innate immune system and the differences between patients with distinct immunotherapies have received little attention to date. Methods: We conducted immunophenotyping using flow cytometry on peripheral blood mononuclear cells of pwMS who received IVMP treatment during a relapse. We compared the impact of an IVMP treatment on a broad variety of immune cell subsets within three groups: twelve patients who were treatment-naïve to disease modifying therapies (wDMT) to ten patients on platform therapies (PT) and eighteen patients on fingolimod therapy (FTY). Results: We observed pronounced interindividual short- and intermediate-term effects of IVMP on distinct immune cells subsets. In addition to the well-documented decrease in T-helper cells (Th cells), we detected significant alterations after the first IVMP infusion within the innate immune response among neutrophil, eosinophil and basophil granulocytes, monocytes and plasmacytoid dendritic cells (pDCs). When comparing patients wDMT to the PT and FTY cohorts, we found that IVMP had a similar impact on innate immune cells across all treatment groups. However, we did not observe a significant further decline in T lymphocyte counts during IVMP in patients with pre-existing lymphopenia under FTY treatment. Although T cell apoptosis is considered the main mechanism of action of GCs, patients with FTY still reported symptom improvement following IVMP treatment. Conclusion: In addition to T cell suppression, our data suggests that further immunoregulatory mechanisms of GC, particularly on cells of the innate immune response, are of greater significance than previously understood. Due to the regulation of the adaptive immune cells by DMTs, the impact of GC on these cells varies depending on the underlying DMT. Additional studies involving larger cohorts and cerebrospinal fluid samples are necessary to gain a deeper understanding of the immune response to GC in pwMS with different DMTs during relapse to define and explain differences in clinical response profiles.


Subject(s)
Multiple Sclerosis , Humans , Female , Male , Adult , Middle Aged , Multiple Sclerosis/drug therapy , Multiple Sclerosis/immunology , Fingolimod Hydrochloride/therapeutic use , Fingolimod Hydrochloride/administration & dosage , Immunity, Innate/drug effects , Methylprednisolone/therapeutic use , Methylprednisolone/administration & dosage , Immunophenotyping , Leukocytes, Mononuclear/immunology , Adrenal Cortex Hormones/therapeutic use , Adrenal Cortex Hormones/administration & dosage , Immunosuppressive Agents/therapeutic use , Glucocorticoids/therapeutic use , Glucocorticoids/administration & dosage
18.
Front Immunol ; 15: 1384411, 2024.
Article in English | MEDLINE | ID: mdl-38911861

ABSTRACT

Background: Although fingolimod, a sphingosine 1-phosphate receptor agonist, has shown to be an effective treatment reducing relapse rate and also slowing down the disability progression in relapsing-remitting multiple sclerosis (RRMS) patients, it is important to quickly identify those suboptimal responders. Objective: The main objective was to assess different clinical, radiological, genetic and environmental factors as possible early predictors of response in MS patients treated with fingolimod for 24 months. The secondary objective was to analyze the possible contribution of the environmental factors analyzed to the progression and activity of the disease along the 2-years of follow-up. Methods: A retrospective study with 151 patients diagnosed with MS, under fingolimod treatment for 24 months, with serum samples at initiation and six months later, and with clinical and radiological data at initiation and 24 months later, were included in the study. Clinical and radiological variables were collected to establish NEDA-3 (no evidence of disease activity: patients without relapses, disability progression and new T2 lesions or Gd+ lesions) and EDA (evidence of disease activity: patients with relapses and/or progression and/or new T2 lesions or gadolinium-positive [Gd+] lesions) conditions. Human leukocyte antigen II (HLA-II), EBNA-1 IgG and VCA IgG from Epstein-Barr virus (EBV) and antibody titers against Human herpesvirus 6A/B (HHV-6A/B) were also analyzed. Results: A total of 151 MS patients fulfilled the inclusion criteria: 27.8% was NEDA-3 (37.5% among those previously treated with high efficacy therapies >24 months). The following early predictors were statistically significantly associated with NEDA-3 condition: sex (male; p=0.002), age at baseline (older; p=0.009), relapses 2-years before fingolimod initiation ≤1 (p=0.010), and absence of Gd+ lesions at baseline (p=0.006). Regarding the possible contribution of the environmental factors included in the study to the activity or the progression of the disease, we only found that EBNA-1 IgG titers decreased in 20.0% of PIRA (progression independent from relapse activity) patients vs. 73.3% of RAW (relapse-associated worsening) patients (p=0.006; O.R. = 11.0). Conclusion: MS patients that are male, older, and with a low clinical and radiological activity at fingolimod initiation have a greater probability to reach NEDA-3 condition after two years with this therapy. An intriguing association of EBV with the progression of the disease has also been described, but it should be further study in a larger cohort to confirm these results.


Subject(s)
Disease Progression , Epstein-Barr Virus Nuclear Antigens , Fingolimod Hydrochloride , Immunoglobulin G , Humans , Fingolimod Hydrochloride/therapeutic use , Female , Male , Adult , Epstein-Barr Virus Nuclear Antigens/immunology , Retrospective Studies , Immunoglobulin G/blood , Middle Aged , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Multiple Sclerosis, Relapsing-Remitting/immunology , Multiple Sclerosis, Relapsing-Remitting/blood , Treatment Outcome , Immunosuppressive Agents/therapeutic use , Multiple Sclerosis/drug therapy , Multiple Sclerosis/immunology , Multiple Sclerosis/blood
19.
Eur J Med Res ; 29(1): 344, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38918831

ABSTRACT

Multiple Sclerosis (MS) is a complex autoimmune disorder that significantly impacts the central nervous system, leading to a range of complications. While intracranial haemorrhage (ICH) is a rare but highly morbid complication, more common CNS complications include progressive multifocal leukoencephalopathy (PML) and other CNS infections. This severe form of stroke, known for its high morbidity and mortality rates, presents a critical challenge in the management of MS. The use of disease-modifying drugs (DMDs) in treating MS introduces a nuanced aspect to patient care, with certain medications like Dimethyl Fumarate and Fingolimod showing potential in reducing the risk of ICH, while others such as Alemtuzumab and Mitoxantrone are associated with an increased risk. Understanding the intricate relationship between these DMDs, the pathophysiological mechanisms of ICH, and the individualised aspects of each patient's condition is paramount. Factors such as genetic predispositions, existing comorbidities, and lifestyle choices play a crucial role in tailoring treatment approaches, emphasising the importance of a personalised, vigilant therapeutic strategy. The necessity for ongoing and detailed research cannot be overstated. It is crucial to explore the long-term effects of DMDs on ICH occurrence and prognosis in MS patients, aiming to refine clinical practices and promote patient-centric, informed therapeutic decisions. This approach ensures that the management of MS is not only comprehensive but also adaptable to the evolving understanding of the disease and its treatments.


Subject(s)
Cerebral Hemorrhage , Multiple Sclerosis , Humans , Multiple Sclerosis/drug therapy , Multiple Sclerosis/complications , Cerebral Hemorrhage/etiology , Cerebral Hemorrhage/chemically induced , Immunosuppressive Agents/adverse effects , Immunosuppressive Agents/therapeutic use , Mitoxantrone/therapeutic use , Mitoxantrone/adverse effects , Fingolimod Hydrochloride/therapeutic use , Fingolimod Hydrochloride/adverse effects , Dimethyl Fumarate/therapeutic use , Dimethyl Fumarate/adverse effects
20.
Cancer Res Commun ; 4(7): 1802-1814, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38881188

ABSTRACT

Recent progress in single-cell profiling technologies has revealed significant phenotypic and transcriptional heterogeneity in tumor-infiltrating CD8+ T cells. However, the transition between the different states of intratumoral antigen-specific CD8+ T cells remains elusive. Here, we sought to examine the generation, transcriptomic states, and the clinical relevance of melanoma-infiltrating CD8+ T cells expressing a chemokine receptor and T-cell differentiation marker, CX3C chemokine receptor 1 (CX3CR1). Analysis of single-cell datasets revealed distinct human melanoma-infiltrating CD8+ T-cell clusters expressing genes associated with effector T-cell function but with distinguishing expression of CX3CR1 or PDCD1. No obvious impact of CX3CR1 expression in melanoma on the response to immune checkpoint inhibitor therapy was observed while increased pretreatment and on-treatment frequency of a CD8+ T-cell cluster expressing high levels of exhaustion markers was associated with poor response to the treatment. Adoptively transferred antigen-specific CX3CR1- CD8+ T cells differentiated into the CX3CR1+ subset in mice treated with FTY720, which inhibits lymphocyte egress from secondary lymphoid tissues, suggesting the intratumoral generation of CX3CR1+ CD8+ T cells rather than their trafficking from secondary lymphoid organs. Furthermore, analysis of adoptively transferred antigen-specific CD8+ T cells, in which the Cx3cr1 gene was replaced with a marker gene confirmed that CX3CR1+ CD8+ T cells could directly differentiate from the intratumoral CX3CR1- subset. These findings highlight that tumor antigen-specific CX3CR1- CD8+ T cells can fully differentiate outside the secondary lymphoid organs and generate CX3CR1+ CD8+ T cells in the tumor microenvironment, which are distinct from CD8+ T cells that express markers of exhaustion. SIGNIFICANCE: Intratumoral T cells are composed of heterogeneous subpopulations with various phenotypic and transcriptional states. This study illustrates the intratumoral generation of antigen-specific CX3CR1+ CD8+ T cells that exhibit distinct transcriptomic signatures and clinical relevance from CD8+ T cells expressing markers of exhaustion.


Subject(s)
CD8-Positive T-Lymphocytes , CX3C Chemokine Receptor 1 , Lymphocytes, Tumor-Infiltrating , Melanoma , Transcriptome , CX3C Chemokine Receptor 1/genetics , CX3C Chemokine Receptor 1/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Melanoma/immunology , Melanoma/genetics , Melanoma/pathology , Animals , Humans , Mice , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Fingolimod Hydrochloride/pharmacology , Fingolimod Hydrochloride/therapeutic use , Mice, Inbred C57BL , Single-Cell Analysis , Clinical Relevance
SELECTION OF CITATIONS
SEARCH DETAIL