Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 888
Filtrar
1.
Clin Neurol Neurosurg ; 232: 107892, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37454600

RESUMO

BACKGROUND AND PURPOSE: Aim of this study was to evaluated anxiety, depression, and possible negative implications on work activities during the Sars-CoV-2 pandemic, in a group of Multiple Sclerosis (MS) patients at risk of flu-like syndrome (FLS) compared with FLS- free treatments. METHODS: The present study included patients treated with interferon-ß (IFNß), glatiramer, and natalizumab for at least one year. Collected data included the diagnosis of COVID-19 infection, Beck Depression Inventory-II (BDI-II), Beck Anxiety Inventory (BAI), together with questions about FLS, change in work habits, use of antipyretics, anxiety, and depression. RESULTS: 100 patients were included in the study. Six patients in IFNß and 5 in the natalizumab group had a confirmed COVID-19 infection. 68% in the IFNß patients reported FLS and only one reported an increase in flu-like frequency during the pandemic; 14% reported lower compliance with treatment, and 40% reported uptake of antipyretics several times. Only one IFNß patient reported having lost more working days than the previous year. The average BAI (p = 0.039) was higher in natalizumab group. Correcting these data by age, sex and EDSS to a multivariate analysis we did not find any statistically significant difference in terms of BAI and BDI-II between the three treatment groups. CONCLUSIONS: FLS were not perceived as COVID19-like symptoms but as expected by traditional pharmacological treatments indeed. These data suggest that IFNß can be used safely.


Assuntos
Antipiréticos , COVID-19 , Esclerose Múltipla , Humanos , Esclerose Múltipla/tratamento farmacológico , Interferon beta/efeitos adversos , SARS-CoV-2 , Natalizumab/uso terapêutico , Depressão/epidemiologia , Depressão/diagnóstico , Pandemias , Antipiréticos/uso terapêutico , COVID-19/epidemiologia , Ansiedade/induzido quimicamente , Ansiedade/epidemiologia , Ansiedade/diagnóstico
2.
Br J Clin Pharmacol ; 89(5): 1682-1685, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36480744

RESUMO

AIMS: Interferon-beta (IFNß), the most widely prescribed medication for multiple sclerosis, is generally considered safe. Nevertheless, rare serious and/or life-threatening side effects have been reported such as thrombotic microangiopathy. A few mechanisms have been proposed to explain how interferon causes thrombotic microangiopathy, but immunological studies have been unable to pin this phenomenon down to a single pathophysiologic pathway. The aim of this article was to report a new mechanism explaining Interferon beta related thrombotic microangiopathy. METHODS: We report thrombotic microangiopathy in a 28-year-old male receiving interferon-beta treatment for multiple sclerosis. RESULTS: After three years of starting interferon beta therapy, the patient presented with malignant hypertension causing seizures, rapidly progressive renal failure requiring haemodialysis and haemolytic anaemia. Corticosteroid and plasma exchange sessions permitted haemolysis control. Nonetheless, the patient remained hemodialysis-dependent. Exploration of the complement system found a complement factor I deficiency whose activity normalized at the control carried out after 2 years. CONCLUSION: IFNß treatment may cause complement factor I deficit, which can lead to thrombotic microangiopathy and severe renal failure.


Assuntos
Esclerose Múltipla , Insuficiência Renal , Microangiopatias Trombóticas , Masculino , Humanos , Adulto , Interferon beta/efeitos adversos , Esclerose Múltipla/tratamento farmacológico , Microangiopatias Trombóticas/induzido quimicamente , Insuficiência Renal/complicações
3.
Eur J Neurol ; 30(2): 453-462, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36318271

RESUMO

BACKGROUND AND PURPOSE: Brain pseudoatrophy has been shown to play a pivotal role in the interpretation of brain atrophy measures during the first year of disease-modifying therapy in multiple sclerosis. Whether pseudoatrophy also affects the spinal cord remains unclear. The aim of this study was to analyze the extent of pseudoatrophy in the upper spinal cord during the first 2 years after therapy initiation and compare this to the brain. METHODS: A total of 129 patients from a prospective longitudinal multicentric national cohort study for whom magnetic resonance imaging scans at baseline, 12 months, and 24 months were available were selected for brain and spinal cord volume quantification. Annual percentage brain volume and cord area change were calculated using SIENA (Structural Image Evaluation of Normalized Atrophy) and NeuroQLab, respectively. Linear mixed model analyses were performed to compare patients on interferon-beta therapy (n = 84) and untreated patients (n = 45). RESULTS: Patients treated with interferon-beta demonstrated accelerated annual percentage brain volume and cervical cord area change in the first year after treatment initiation, whereas atrophy rates stabilized to a similar and not significantly different level compared to untreated patients during the second year. CONCLUSIONS: These results suggest that pseudoatrophy occurs not only in the brain, but also in the spinal cord during the first year of interferon-beta treatment.


Assuntos
Esclerose Múltipla , Humanos , Esclerose Múltipla/diagnóstico por imagem , Esclerose Múltipla/tratamento farmacológico , Esclerose Múltipla/patologia , Interferon beta/efeitos adversos , Estudos de Coortes , Estudos Prospectivos , Medula Espinal/diagnóstico por imagem , Medula Espinal/patologia , Encéfalo/diagnóstico por imagem , Encéfalo/patologia , Imageamento por Ressonância Magnética/métodos , Atrofia/patologia
4.
JAMA Netw Open ; 5(9): e2230439, 2022 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-36169959

RESUMO

Importance: With few approved multiple sclerosis therapies in the pediatric population, there is a need for further approved treatment options. Limited data exist for dimethyl fumarate (DMF) treatment in pediatric-onset multiple sclerosis (POMS). Objective: To compare the efficacy, safety, and tolerability of DMF vs intramuscular interferon ß-1a (IFNß-1a) in POMS. Design, Setting, and Participants: The CONNECT study was an active-controlled, open-label, rater-blinded 96-week randomized clinical trial in patients with POMS aged 10 to less than 18 years treated between August 2014 and November 2020. Data were analyzed from January through October 2021. Interventions: Patients were randomized to DMF or IFNß-1a. Main Outcomes and Measures: The primary end point was the proportion of patients free of new or newly enlarging (N or NE) T2 hyperintense lesions at week 96 among trial completers. Secondary end points included number of N or NE T2 lesions, proportion of patients free of relapse, annualized relapse rate (ARR), and safety. The estimated proportion of participants who were relapse free up to week 96 was calculated based on the Kaplan-Meier method. Adjusted ARR was obtained from a negative binomial regression adjusted for baseline relapse rate, baseline Expanded Disability Status Scale (EDSS) score, and age group. Results: Among 150 patients with POMS in the intention-to-treat (ITT) population (median [range] age, 15 [10-17] years; 101 [67.3%] female patients), 78 individuals received DMF and 72 individuals received IFNß-1a. At week 96, the proportion of patients with no N or NE T2 hyperintense lesions among 103 trial completers was 16.1% (95% CI, 8.0%-27.7%) for DMF vs 4.9% (95% CI, 0.6%-16.5%) for IFNß-1a, and in a sensitivity analysis among the ITT population, the proportions were 10 patients receiving DMF (12.8%) vs 2 patients receiving IFNß-1a (2.8%). The estimated proportion of patients who remained relapse free at week 96 was 66.2% for DMF vs 52.3% for IFNß-1a. Adjusted ARR (95% CI) at week 96 was 0.24 (95% CI, 0.15-0.39) for DMF vs 0.53 (95% CI, 0.33-0.84) for IFNß-1a; the rate ratio for DMF vs IFNß-1a was 0.46 (95% CI, 0.26-0.80; P = .006). The number of treatment-emergent adverse events (TEAEs; 74 patients [94.9%] vs 69 patients [95.8%]), serious TEAEs (18 patients [23.1%] vs 21 patients [29.2%]), and treatment discontinuations due to TEAEs (5 patients [6.4%] vs 8 patients [11.1%]) was similar for DMF vs IFNß-1a. Conclusions and Relevance: This study found that more pediatric patients with POMS treated with DMF were free of new or newly enlarging T2 lesions and that the adjusted ARR was lower among these patients compared with those treated with interferon ß-1a. DMF was well tolerated. Trial Registration: ClinicalTrials.gov Identifier: NCT02283853.


Assuntos
Esclerose Múltipla Recidivante-Remitente , Esclerose Múltipla , Adolescente , Criança , Fumarato de Dimetilo/uso terapêutico , Feminino , Humanos , Interferon beta-1a/uso terapêutico , Interferon beta/efeitos adversos , Interferon beta/uso terapêutico , Masculino , Esclerose Múltipla/tratamento farmacológico , Esclerose Múltipla Recidivante-Remitente/induzido quimicamente , Esclerose Múltipla Recidivante-Remitente/tratamento farmacológico , Recidiva Local de Neoplasia/tratamento farmacológico
7.
Curr Neuropharmacol ; 20(8): 1600-1612, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34720084

RESUMO

Multiple sclerosis (MS) is a chronic neurodegenerative, inflammatory, and autoimmune disease characterised by the demyelination of the central nervous system. One of the main approaches for treating MS is the use of disease-modifying therapies (DMTs). Among the DMTs are interferons (IFNs), which are cytokines responsible for controlling the activity of the immune system while exerting immunomodulatory, antiviral, and antiproliferative activities. IFN-beta (IFN-ß) is the first-choice drug used to treat relapsing-remitting MS. However, the administration of IFN-ß causes numerous painful adverse effects, resulting in lower adherence to the treatment. Therefore, this study aimed to investigate the headache and flu-like pain symptoms observed after IFNß injection in MS patients using a systematic review and meta-analysis of randomised controlled trials. A total of 2370 articles were identified through research databases. Nine articles were included (three involving IFNß-1b and six involving IFNß-1a). All studies included in the meta-analysis had a low risk of bias. The odds ratio of headache and flu-like pain symptoms increased in MS patients treated with IFN-ß. Thus, the adverse effects of headache and flu-like pain symptoms appear to be linked to IFN-ß treatment in MS. The protocol of the study was registered in the Prospective International Registry of Systematic Reviews (registration number CRD42021227593).


Assuntos
Cefaleia , Interferon beta , Esclerose Múltipla , Cefaleia/induzido quimicamente , Humanos , Interferon beta/efeitos adversos , Interferon beta/uso terapêutico , Esclerose Múltipla/complicações , Esclerose Múltipla/tratamento farmacológico , Dor/induzido quimicamente , Ensaios Clínicos Controlados Aleatórios como Assunto
8.
Expert Rev Anti Infect Ther ; 20(5): 741-747, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-34747295

RESUMO

OBJECTIVE: The aim of this systematic review and meta-analysis of randomized controlled trials(RCTs) was to investigate the efficacy of interferon (IFN)-ß-containing regimens in treating patients with COVID-19. METHODS: PubMed, Embase, Cochrane Central Register of Controlled Trials, and ClinicalTrials.gov were searched from inception to 17 July 2021. RCTs comparing the clinical efficacy and safety of IFN-ß-containing regimens (study group) to other antiviral treatment options or placebo (control group) in treating patients with COVID-19 were included. RESULTS: Eight RCTs were included. No significant difference in the 28-day all-cause mortality rate was observed between the study and control groups (OR, 0.74; 95% CI, 0.44-1.24; I2 = 51%). The study groups had a lower rate of intensive care unit (ICU) admissions than the control groups (OR 0.58, 95% CI 0.36-0.95; I2 = 0%). Furthermore, INF-ß was not associated with an increased risk of any adverse event (AE) or serious AE when compared with the control group. CONCLUSIONS: IFN-ß does not appear to provide an increased survival benefit in hospitalized patients with COVID-19 but may help reduce the risk of ICU admission. Moreover, IFN-ß is a safe agent for use in the treatment of COVID-19.


Assuntos
Tratamento Farmacológico da COVID-19 , Antivirais/efeitos adversos , Humanos , Interferon beta/efeitos adversos , Ensaios Clínicos Controlados Aleatórios como Assunto , SARS-CoV-2 , Resultado do Tratamento
9.
Pain ; 163(5): 923-935, 2022 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-34490849

RESUMO

ABSTRACT: Activation of toll-like receptor 4 (TLR4) in the dorsal root ganglion (DRG) and spinal cord contributes to the generation of paclitaxel-related chemotherapy-induced peripheral neuropathy (CIPN). Generalizability of TLR4 signaling in oxaliplatin-induced CIPN was tested here. Mechanical hypersensitivity developed in male SD rats by day 1 after oxaliplatin treatment, reached maximum intensity by day 14, and persisted through day 35. Western blot revealed an increase in TLR4 expression in the DRG of oxaliplatin at days 1 and 7 after oxaliplatin treatment. Cotreatment of rats with the TLR4 antagonist lipopolysaccharide derived from Rhodobacter sphaeroides ultrapure or with the nonspecific immunosuppressive minocycline with oxaliplatin resulted in significantly attenuated hyperalgesia on day 7 and 14 compared with rats that received oxaliplatin plus saline vehicle. Immunostaining of DRGs revealed an increase in the number of neurons expressing TLR4, its canonical downstream signal molecules myeloid differentiation primary response gene 88 (MyD88) and TIR-domain-containing adapter-inducing interferon-ß, at both day 7 and day 14 after oxaliplatin treatment. These increases were blocked by cotreatment with either lipopolysaccharide derived from Rhodobacter sphaeroides or minocycline. Double staining showed the localization of TLR4, MyD88, and TIR-domain-containing adapter-inducing interferon-ß in subsets of DRG neurons. Finally, there was no significant difference in oxaliplatin-induced mechanical hypersensitivity between male and female rats when observed for 2 weeks. Furthermore, upregulation of TLR4 was detected in both sexes when tested 14 days after treatment with oxaliplatin. These findings suggest that the activation of TLR4 signaling in DRG neurons is a common mechanism in CIPN induced by multiple cancer chemotherapy agents.


Assuntos
Gânglios Espinais , Doenças do Sistema Nervoso Periférico , Animais , Feminino , Gânglios Espinais/metabolismo , Hiperalgesia/induzido quimicamente , Hiperalgesia/metabolismo , Interferon beta/efeitos adversos , Interferon beta/metabolismo , Lipopolissacarídeos/toxicidade , Masculino , Minociclina/uso terapêutico , Fator 88 de Diferenciação Mieloide/metabolismo , Oxaliplatina/toxicidade , Doenças do Sistema Nervoso Periférico/tratamento farmacológico , Ratos , Ratos Sprague-Dawley , Receptor 4 Toll-Like/metabolismo
10.
BMC Ophthalmol ; 21(1): 352, 2021 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-34592956

RESUMO

BACKGROUND: Susac syndrome (SS) is characterized by the triad of encephalopathy, branch retinal artery occlusion, and sensorineural hearing loss. However, the diagnosis of SS remains difficult because the clinical triad rarely occurs at disease onset, and symptom severity varies. SS symptoms often suggest other diseases, in particular multiple sclerosis (MS), which is more common. Misdiagnosing SS as MS may cause serious complications because MS drugs, such as interferon beta-1a, can worsen the course of SS. This case report confirms previous reports that the use of interferon beta-1a in the course of misdiagnosed MS may lead to exacerbation of SS. Moreover, our case report shows that glatiramer acetate may also exacerbate the course of SS. To the best of our knowledge, this is the first reported case of exacerbation of SS by glatiramer acetate. CASE PRESENTATION: We present a case report of a patient with a primary diagnosis of MS who developed symptoms of SS during interferon beta-1a treatment for MS; these symptoms were resolved after the discontinuation of the treatment. Upon initiation of glatiramer acetate treatment, the patient developed the full clinical triad of SS. The diagnosis of MS was excluded, and glatiramer acetate therapy was discontinued. The patient's neurological state improved only after the use of a combination of corticosteroids, intravenous immunoglobulins, and azathioprine. CONCLUSIONS: The coincidence of SS signs and symptoms with treatment for MS, first with interferon beta-1a and then with glatiramer acetate, suggests that these drugs may influence the course of SS. This case report indicates that treatment with glatiramer acetate may modulate or even exacerbate the course of SS.


Assuntos
Esclerose Múltipla , Síndrome de Susac , Erros de Diagnóstico , Acetato de Glatiramer/efeitos adversos , Humanos , Interferon beta-1a/efeitos adversos , Interferon beta/efeitos adversos , Esclerose Múltipla/diagnóstico , Esclerose Múltipla/tratamento farmacológico , Síndrome de Susac/diagnóstico , Síndrome de Susac/tratamento farmacológico
12.
Sci Rep ; 11(1): 12533, 2021 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-34131191

RESUMO

Neoplasm development in Multiple Sclerosis (MS) patients treated with disease-modifying therapies (DMTs) has been widely discussed. The aim of this work is to determine neoplasm frequency, relationship with the prescription pattern of DMTs, and influence of the patients' baseline characteristics. Data from 250 MS outpatients were collected during the period 1981-2019 from the medical records of the Neurology Service of the HUPM (Hospital Universitario Puerta del Mar)-in Southern Spain-and analysed using Cox models. Neoplasm prevalence was 24%, mainly located on the skin, with cancer prevalence as expected for MS (6.8%). Latency period from MS onset to neoplasm diagnosis was 10.4 ± 6.9 years (median 9.30 [0.9-30.5]). During the observation period ß-IFN (70.4% of patients), glatiramer acetate (30.4%), natalizumab (16.8%), fingolimod (24.8%), dimethyl fumarate (24.0%), alemtuzumab (6.0%), and teriflunomide (4.8%) were administered as monotherapy. Change of pattern in step therapy was significantly different in cancer patients vs unaffected individuals (p = 0.011) (29.4% did not receive DMTs [p = 0.000]). Extended Cox model: Smoking (HR = 3.938, CI 95% 1.392-11.140, p = 0.010), being female (HR = 2.006, 1.070-3.760, p = 0.030), and age at MS diagnosis (AGE-DG) (HR = 1.036, 1.012-1.061, p = 0.004) were risk factors for neoplasm development. Secondary progressive MS (SPMS) phenotype (HR = 0.179, 0.042-0.764, p = 0.020) and treatment-time with IFN (HR = 0.923, 0.873-0.977, p = 0.006) or DMF (HR = 0.725, 0.507-1.036, p = 0.077) were protective factors. Tobacco and IFN lost their negative/positive influence as survival time increased. Cox PH model: Tobacco/AGE-DG interaction was a risk factor for cancer (HR = 1.099, 1.001-1.208, p = 0.049), followed by FLM treatment-time (HR = 1.219, 0.979-1.517). In conclusion, smoking, female sex, and AGE-DG were risk factors, and SPMS and IFN treatment-time were protective factors for neoplasm development; smoking/AGE-DG interaction was the main cancer risk factor.


Assuntos
Imunossupressores/efeitos adversos , Esclerose Múltipla Crônica Progressiva/tratamento farmacológico , Esclerose Múltipla Recidivante-Remitente/tratamento farmacológico , Esclerose Múltipla/tratamento farmacológico , Neoplasias/epidemiologia , Adulto , Idoso , Alemtuzumab/efeitos adversos , Alemtuzumab/uso terapêutico , Crotonatos/efeitos adversos , Crotonatos/uso terapêutico , Fumarato de Dimetilo/efeitos adversos , Fumarato de Dimetilo/uso terapêutico , Feminino , Cloridrato de Fingolimode/efeitos adversos , Cloridrato de Fingolimode/uso terapêutico , Acetato de Glatiramer/efeitos adversos , Acetato de Glatiramer/uso terapêutico , Humanos , Hidroxibutiratos/efeitos adversos , Hidroxibutiratos/uso terapêutico , Imunossupressores/uso terapêutico , Interferon beta/efeitos adversos , Interferon beta/uso terapêutico , Masculino , Pessoa de Meia-Idade , Esclerose Múltipla/complicações , Esclerose Múltipla/epidemiologia , Esclerose Múltipla/patologia , Esclerose Múltipla Crônica Progressiva/complicações , Esclerose Múltipla Crônica Progressiva/epidemiologia , Esclerose Múltipla Crônica Progressiva/patologia , Esclerose Múltipla Recidivante-Remitente/complicações , Esclerose Múltipla Recidivante-Remitente/epidemiologia , Esclerose Múltipla Recidivante-Remitente/patologia , Natalizumab/efeitos adversos , Natalizumab/uso terapêutico , Neoplasias/induzido quimicamente , Neoplasias/patologia , Nitrilas/efeitos adversos , Nitrilas/uso terapêutico , Pacientes Ambulatoriais , Modelos de Riscos Proporcionais , Fatores de Risco , Fumar/efeitos adversos , Espanha , Toluidinas/efeitos adversos , Toluidinas/uso terapêutico
13.
Int Immunopharmacol ; 97: 107715, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33933848

RESUMO

Interferon beta (IFN-ß) has successfully been experimented with to treat multiple sclerosis (MS). However, patients sometimes do not respond effectively to treatment, and |adverse effects, including liver toxicity, accompany this therapy. |Accordingly, we decided to treat MS patients simultaneously with Silymarin (SM) as an immunomodulatory and hepatoprotective agent and IFN-ß in a clinical trial study. Complete blood count (CBC), liver enzyme levels, and the serum concentration of inflammatory and anti-inflammatory cytokines were measured. Also, the frequency of immune cells was determined by flow cytometry. Liver enzyme levels were significantly lower in the intervention group (p < 0.05). The percentage of Th17 cells in the intervention group was significantly reduced compared to the placebo group (P < 0.001). Also, the frequency of Treg cells after treatment with SM plus IFN-ß was significantly increased compared to the placebo group (p < 0.05). Furthermore, the IL-17 and IFNγ cytokine levels were significantly reduced in the intervention group (p < 0.05). Moreover, the levels of anti-inflammatory cytokines IL-10 and TGFß were significantly increased in the intervention group (P < 0.05).Overall, the results provide novel and supplementary information on SM's notable immunoregulatory effects on inflammatory response and liver function in MS patients. Clinical Trial Identifier Number: IRCTID: IRCT20171220037977N1.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Interferon beta/efeitos adversos , Esclerose Múltipla/tratamento farmacológico , Silimarina/administração & dosagem , Adulto , Doença Hepática Induzida por Substâncias e Drogas/diagnóstico , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Método Duplo-Cego , Sinergismo Farmacológico , Quimioterapia Combinada/efeitos adversos , Quimioterapia Combinada/métodos , Feminino , Humanos , Interferon beta/farmacocinética , Testes de Função Hepática , Masculino , Pessoa de Meia-Idade , Esclerose Múltipla/imunologia , Placebos/administração & dosagem , Silimarina/farmacocinética , Resultado do Tratamento
14.
Ned Tijdschr Geneeskd ; 1652021 02 18.
Artigo em Holandês | MEDLINE | ID: mdl-33651510

RESUMO

A 43-year-old male visited our dermatology department with a skin eruption diagnosed as psoriasis. It began 1 month after starting peg-interferon-α-1a therapy for multiple sclerosis. Peg-interferon is known to be able to induce psoriasis, as well as other medication, while the pathophysiology remains unclear.


Assuntos
Fatores Imunológicos/efeitos adversos , Interferon beta/efeitos adversos , Esclerose Múltipla/tratamento farmacológico , Polietilenoglicóis/efeitos adversos , Psoríase/induzido quimicamente , Adulto , Humanos , Fatores Imunológicos/uso terapêutico , Interferon beta/uso terapêutico , Masculino , Esclerose Múltipla/complicações , Polietilenoglicóis/uso terapêutico
16.
Lima; IETSI; feb. 2021.
Não convencional em Espanhol | BRISA/RedTESA | ID: biblio-1358616

RESUMO

INTRODUCCIÓN: El presente dictamen expone la evaluación de la eficacia y seguridad de fingolimod en el tratamiento de pacientes adultos con esclerosis múltiple recurrente remitente con falla al tratamiento con interferón beta. La esclerosis múltiple (EM) es una enfermedad inflamatoria y neurodegenerativa del cerebro y la médula espinal, caracterizada por episodios de disfunción neurológica que usualmente presentan recuperación; aunque también puede presentar un curso progresivo gradual. La esperanza de vida de los pacientes con EM es de 7 a 14 años menos que la población general. Más de la mitad de las muertes de pacientes con EM están directamente relacionadas a complicaciones de la enfermedad. Fingolimod, producto farmacéutico de administración oral, es un modulador del receptor de la esfingosina 1-fosfato, que, al ser metabolizado, da lugar al metabolito activo fingolimod fosfato. El metabolito activo actúa como antagonista funcional del receptor S1P, bloqueando la capacidad de los linfocitos de salir de los ganglios linfáticos; lo que produce una redistribución de los linfocitos sin disminución en su número. Este efecto reduciría la infiltración de células linfocíticas patógenas al sistema nervioso central; disminuyendo así la inflamación y lesión del tejido nervioso. A pesar que en EsSalud se dispone de interferón beta-1b para el tratamiento de la EM recurrente-remitente (EMRR) y de interferón beta-1a para pacientes con EMRR que presentan eventos adversos al tratamiento previo con interferón beta-1b, existen pacientes que no logran un control adecuado de la enfermedad con la terapia basada en interferón beta. Por ello, existe el interés de brindar una terapia que disminuya la tasa de recaída anualizada por la EMRR y mejore además la calidad de vida. Así, especialistas de EsSalud consideran que el uso de fingolimod beneficiaría a los pacientes con EMRR y falla al tratamiento con interferón beta. METODOLOGÍA: Se realizó una búsqueda de la literatura científica con el objetivo de identificar evidencia sobre la eficacia y seguridad de fingolimod, comparado con la mejor terapia de soporte, en pacientes adultos con EMRR con falla al tratamiento con interferón beta. Para identificar los documentos de interés para la presente evaluación, se buscó evidencia disponible en las siguientes bases de datos bibliográficas: PubMed, The Cochrane Library y LILACS. Adicionalmente, se realizó una búsqueda en sitios web pertenecientes a grupos que realizan evaluaciones de tecnologías sanitarias y guías de práctica clínica, incluyendo The Canadian Agency for Drugs and Technologies in Health (CADTH), Scottish Medicines Consortium (SMC), The National Institute for Health and Care Excellence (NICE), Institute for Quality and Efficiency in Health Care (IQWiG), Haute Authorité de Santé (HAS), el portal BRISA (Base Regional de Informes de Evaluación de Tecnologías en Salud de las Américas), y sitios web de organizaciones internacionales en neurología. Adicionalmente, se llevó a cabo una búsqueda manual en el portal ClinicalTrials.gov del National Institutes of Health e International Clinical Trials Registry Platform de la Organización Mundial de la Salud (http://apps.who.int/trialsearch/) para la identificación de estudios que emplearan la tecnología de interés. Finalmente, se revisaron protocolos para revisiones sistemáticas (RS) que pudieran contemplar el uso de la tecnología de interés en el portal PROSPERO del Centre for Reviews and Dissemination de la University of York (https://www.crd.york.ac.uk/PROSPERO/) y en el Systematic Review Register del Joanna Briggs Institute Centre (https://joannabriggs.org/resources/systematic_review_register). RESULTADOS: Se describe la evidencia disponible según el tipo de publicación, siguiendo lo indicado en los criterios de elegibilidad. CONCLUSIONES:  En el presente documento, se evaluó la mejor evidencia científica disponible hasta la actualidad sobre la eficacia y seguridad de fingolimod en pacientes adultos con esclerosis múltiple recurrente-remitente (EMRR) con falla al tratamiento con interferón beta.  Se incluyeron cinco documentos: dos GPC elaboradas por la AAN y la ECTRIMS/EAN, dos ETS realizadas por NICE y SMC, y el análisis post hoc del ECA FREEDOMS. Este ECA fue incluido como evidencia indirecta dado que evaluó a una población más amplia que la población de interés del presente dictamen. Las GPC de la AAN y ECTRIMS/EAN recomiendan el empleo de otra terapia modificadora de la enfermedad, incluyendo dentro las alternativas a fingolimod, en el caso de que no se presente una respuesta favorable a la terapia con una terapia modificadora de la enfermedad en pacientes con EMRR como el interferón beta. Ninguna de estas GPC establece una recomendación específica sobre el uso de fingolimod en pacientes con fallo a la terapia con interferón beta. Las ETS de NICE y SMC recomiendan fingolimod para el tratamiento de la EMRR altamente activa en adultos, solo si los pacientes tienen una tasa de recaída sin cambios o aumentada o recaídas graves en curso en comparación con el año anterior a pesar del tratamiento adecuado con interferón beta, y que la compañía fabricante proporcione fingolimod con una reducción de costo que haga costo-efectivo el empleo de fingolimod en el sistema sanitario británico y escocés. Cabe precisar que este tipo de regulaciones de costo de medicamentos no son extrapolables al sistema sanitario de EsSalud. En el análisis post hoc del ECA FREEDOMS, para el subgrupo de pacientes con EMRR previamente tratados con interferón beta, se observó menor tasa de recaída anualizada en el grupo que recibió fingolimod, comparado al que recibió placebo. No se observó diferencia en la progresión de la discapacidad para pacientes con EMRR previamente tratados con interferón beta entre los grupos de fingolimod y placebo. No se reportaron resultados sobre las pruebas para evaluar discapacidad (EDSS o MSFC), ni resultados de seguridad (eventos adversos). Entre las principales limitaciones del ECA FREEDOMS, este no fue diseñado para evaluar fingolimod en la población de interés del presente dictamen, sino para una población más amplia (pacientes con EMRR sin tratamiento o previamente tratados con interferón beta u otra terapia modificadora de la enfermedad). Dado que los resultados del ECA FREEDOMS para pacientes con EMRR previamente tratados con interferón beta provienen de un análisis post hoc, estos son de tipo exploratorio, no siendo de este modo concluyentes para determinar eficacia. Adicionalmente, el desenlace primario (tasa de recaída anualizada) presenta limitaciones, al ser un desenlace subjetivo, con la posibilidad de introducción de sesgos por parte del evaluador o el paciente. Asimismo, no se dispone de resultados específicos para pruebas de discapacidad como EDSS o MSFC, limitándose la evaluación del beneficio de esta tecnología en los pacientes de interés, siendo que se recomienda que la evaluación del beneficio de una tecnología no solo sea medida desde este desenlace, sino de la conjunción de lo observado en varios desenlaces de eficacia. En cuanto a la seguridad, la ausencia de información sobre eventos adversos en el subgrupo de pacientes con EMRR previamente tratados con interferón beta, no permite establecer conclusiones sobre la seguridad del empleo de la tecnología. Ante la limitada evidencia para la presente evaluación, se consideró la opinión del experto clínico de la institución. El clínico señala que, dado que fingolimod emplea un mecanismo de acción distinto al interferón beta, sería de utilidad para el control de la enfermedad en pacientes con EMRR y falla a interferón beta. Adicionalmente, fingolimod es fácil de administrar comparado al interferón beta, (vía oral comparada a la administración intramuscular o subcutánea del interferón beta 1a y 1b, respectivamente), lo que facilita su administración. Lo descrito por el especialista está en línea con lo descrito por las GPC, las que incluyen a fingolimod como alternativa terapéutica en pacientes con fracaso a terapia con interferón beta u otra terapia modificadora de efecto. De este modo, la evidencia científica disponible a la fecha sugiere que fingolimod tendría un beneficio en pacientes con EMRR con fallo al tratamiento con interferón beta, bajo los siguientes argumentos técnicos: 1) La evidencia indirecta procedente del ECA FREEDOMS sugiere un beneficio favorable para fingolimod en comparación con placebo en pacientes con EMRR; 2) aunque las GPC revisadas en este dictamen no generan recomendaciones para la población de interés de manera específica, se señala a fingolimod como una alternativa en pacientes con una respuesta no favorable al tratamiento previo con alguna terapia modificadora de la enfermedad (como el interferón beta) y 3) el vacío terapéutico (en el sistema sanitario de EsSalud, a la fecha, los pacientes con EMRR y fallo a la terapia con interferón beta no cuentan con una terapia específica disponible). Por lo expuesto, el IETSI aprueba el uso de fingolimod en pacientes adultos con esclerosis múltiple recurrente remitente con falla al tratamiento con interferón b. La vigencia del presente dictamen es de 1 año, según lo establecido en el Anexo N° 1. Así, la continuación de dicha aprobación estará sujeta a la evaluación de los resultados obtenidos y de nueva evidencia que pueda surgir en el tiempo.


Assuntos
Humanos , Interferon beta/efeitos adversos , Esclerose Múltipla Recidivante-Remitente/tratamento farmacológico , Cloridrato de Fingolimode/uso terapêutico , Eficácia , Análise Custo-Benefício
17.
Mult Scler Relat Disord ; 48: 102694, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33429303

RESUMO

BACKGROUND: Previous studies reported no increase in the prevalence of adverse pregnancy outcomes after exposure to interferon-beta (IFN-beta). However, no study has investigated if the prevalence of these outcomes after IFN-beta exposure is modified by maternal and newborn characteristics. Our objective was to describe the stratified prevalence of adverse pregnancy outcomes among women with multiple sclerosis (MS) exposed only to IFN-beta or unexposed to any MS disease modifying drugs (MSDMDs). METHODS: This population-based cohort study using Finnish (1996-2014) and Swedish (2005-2014) register data included pregnancies of women with MS exposed only to IFN-beta 6 months before or during pregnancy (n=718) or unexposed to MSDMDs (n=1397). The outcome prevalences were described stratified by maternal and newborn characteristics, with 95% confidence intervals (CIs). Confounder-adjusted analyses were performed if the prevalence results indicated modified effect of IFN-beta in specific strata. RESULTS: The stratified analysis indicated that the prevalence of serious (anomaly or stillbirth) and other adverse pregnancy outcomes was similar among the exposed and unexposed, with no statistically significant difference. Among women treated for MS >5 years, serious adverse pregnancy outcomes occurred in 4.3% (95%CI: 1.9-8.3%) of pregnancies exposed only to IFN-beta 6 months before or during pregnancy and in 2.7% (95%CI: 1.2-5.0%) of unexposed pregnancies. The confounder adjusted analyses did not support the hypothesis that MS treatment duration before pregnancy would modify the risk of adverse pregnancy outcomes after exposure to IFN-beta 6 months before or during pregnancy. CONCLUSION: The prevalence of adverse pregnancy outcomes was not increased after IFN-beta exposure, when pregnancies of women with MS were stratified by maternal and newborn characteristics. The stratified results were similar to the unstratified results in the same population.


Assuntos
Esclerose Múltipla , Resultado da Gravidez , Estudos de Coortes , Feminino , Finlândia/epidemiologia , Humanos , Recém-Nascido , Interferon beta/efeitos adversos , Esclerose Múltipla/tratamento farmacológico , Esclerose Múltipla/epidemiologia , Gravidez , Resultado da Gravidez/epidemiologia , Prevalência , Suécia/epidemiologia
18.
J Med Virol ; 93(2): 831-842, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32672860

RESUMO

Coronavirus disease 2019 (COVID-19) can lead to a massive cytokine release. The use of the anti-interleukin-6 receptor monoclonal antibody tocilizumab (TCZ) has been proposed in this hyperinflammatory phase, although supporting evidence is limited. We retrospectively analyzed 88 consecutive patients with COVID-19 pneumonia that received at least one dose of intravenous TCZ in our institution between 16 and 27 March 2020. Clinical status from day 0 (first TCZ dose) through day 14 was assessed by a 6-point ordinal scale. The primary outcome was clinical improvement (hospital discharge and/or a decrease of ≥2 points on the 6-point scale) by day 7. Secondary outcomes included clinical improvement by day 14 and dynamics of vital signs and laboratory values. Rates of clinical improvement by days 7 and 14 were 44.3% (39/88) and 73.9% (65/88). Previous or concomitant receipt of subcutaneous interferon-ß (adjusted odds ratio [aOR]: 0.23; 95% confidence interval [CI]: 0.06-0.94; P = .041) and serum lactate dehydrogenase more than 450 U/L at day 0 (aOR: 0.25; 95% CI: 0.06-0.99; P = .048) were negatively associated with clinical improvement by day 7. All-cause mortality was 6.8% (6/88). Body temperature and respiratory and cardiac rates significantly decreased by day 1 compared to day 0. Lymphocyte count and pulse oximetry oxygen saturation/FiO2 ratio increased by days 3 and 5, whereas C-reactive protein levels dropped by day 2. There were no TCZ-attributable adverse events. In this observational single-center study, TCZ appeared to be useful and safe as immunomodulatory therapy for severe COVID-19 pneumonia.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Antivirais/uso terapêutico , Tratamento Farmacológico da COVID-19 , Síndrome da Liberação de Citocina/prevenção & controle , Fatores Imunológicos/uso terapêutico , SARS-CoV-2/patogenicidade , Administração Intravenosa , Adulto , Temperatura Corporal/efeitos dos fármacos , Proteína C-Reativa/metabolismo , COVID-19/imunologia , COVID-19/mortalidade , COVID-19/virologia , Síndrome da Liberação de Citocina/imunologia , Síndrome da Liberação de Citocina/mortalidade , Síndrome da Liberação de Citocina/virologia , Feminino , Frequência Cardíaca/efeitos dos fármacos , Humanos , Interferon beta/efeitos adversos , L-Lactato Desidrogenase/sangue , Masculino , Pessoa de Meia-Idade , Receptores de Interleucina-6/antagonistas & inibidores , Receptores de Interleucina-6/genética , Receptores de Interleucina-6/imunologia , Taxa Respiratória/efeitos dos fármacos , Estudos Retrospectivos , SARS-CoV-2/imunologia , Índice de Gravidade de Doença , Análise de Sobrevida
19.
Brain Behav ; 11(1): e01930, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33325640

RESUMO

BACKGROUND AND AIMS: Interferon beta (IFNß) is a well-established first-line therapy for relapsing-remitting multiple sclerosis (RRMS) patients and remains the most widely prescribed agent. Atypical hemolytic uremic syndrome (aHUS) represents a rare but severe adverse effect (AE) that could occur even after many years from the beginning of IFNß therapy. Eculizumab is currently approved for treatment of aHUS and recently for neuromyelitis optica spectrum disorder (NMOSD) with aquaporin-4 antibodies (AQP4-IgG). In this article, we report the case of the latest onset of IFNß-related aHUS experienced by an MS patient and we briefly review the literature on this topic. METHODS: We performed a systematic review of the literature using PubMed, and we performed a retrospective analysis of RRMS patients that received IFNß-1a in our center and developed thrombotic microangiopathy (TMA). From this search, we identified only one patient. RESULTS: In the published literature, we identified 24 MS patients who received IFNß as disease-modifying treatment (DMT) and then developed thrombotic microangiopathy with kidney injury. The aHUS has been diagnosed in 6, all received IFNß-1a and the latest onset was after 15 years. We report a case of a 39-year-old man affected by RRMS who assumed IFNß-1a since 1999. In July 2018, he developed an IFNß-related aHUS. After the failure of plasma exchange, he underwent eculizumab, with an improvement of glomerular filtration rate and without new signs of MS activity. CONCLUSION: To our knowledge, this case represents the latest onset of IFNß-related aHUS in MS patients. Up to now, there are not literary reports about the possibility to reintroduce a DMT as add-on therapy to eculizumab.


Assuntos
Síndrome Hemolítico-Urêmica Atípica , Esclerose Múltipla Recidivante-Remitente , Esclerose Múltipla , Adulto , Humanos , Interferon beta/efeitos adversos , Masculino , Esclerose Múltipla/tratamento farmacológico , Esclerose Múltipla Recidivante-Remitente/tratamento farmacológico , Estudos Retrospectivos
20.
Eur J Neurol ; 28(5): 1609-1616, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33370478

RESUMO

BACKGROUND AND PURPOSE: There is a lack of evidence guiding discontinuation of disease-modifying therapy (DMT) in relapsing multiple sclerosis (RMS). Thus, the objective of this study was to generate and validate a risk score for disease reactivation after DMT discontinuation in RMS. METHODS: We drew a generation and validation dataset from two separate prospectively collected observational databases including RMS patients who received interferon-ß or glatiramer acetate for ≥12 months, then discontinued DMT for ≥6 months and had ≥2 years of follow-up available. In the generation sample (n = 168), regression analysis was performed to identify clinical or magnetic resonance imaging (MRI) variables independently predicting disease reactivation after DMT discontinuation. A predictive score was calculated using the variables included in the multivariable model and applied to the validation sample (n = 98). RESULTS: The variables included in the final model as independent predictors of disease reactivation were age at discontinuation, MRI activity at discontinuation, and duration of clinical stability (all p < 0.001). The resulting score was able to robustly identify patients at high (83%-85%), moderate (36%-38%), and low risk (7%) of disease reactivation within 5 years after DMT discontinuation in both cohorts. CONCLUSIONS: The composite VIAADISC score is a valuable tool to inform and support patients and neurologists in the process of decision making to discontinue injectable DMTs.


Assuntos
Esclerose Múltipla Recidivante-Remitente , Esclerose Múltipla , Acetato de Glatiramer/efeitos adversos , Humanos , Interferon beta/efeitos adversos , Interferons , Esclerose Múltipla Recidivante-Remitente/tratamento farmacológico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...