Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
BMC Neurol ; 16: 98, 2016 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-27405225

RESUMO

BACKGROUND: Natalizumab provides rapid and high-efficacy control of multiple sclerosis disease activity with long-term stabilization. However, the benefits of the drug are countered by a risk of developing progressive multifocal leukoencephalopathy in patients infected with the John Cunningham Virus. Close monitoring is required in patients with increased progressive multifocal leukoencephalopathy risk receiving natalizumab in the long-term for an optimal benefit-risk evaluation. Standardized high-quality monitoring procedures may provide a superior basis for individual benefit and risk evaluation and thus improve treatment decisions. The non-interventional study TRUST was designed to capture natalizumab effectiveness under real-life conditions and to examine alternate approaches for clinical assessments, magnetic resonance imaging monitoring and use of biomarkers for progressive multifocal leukoencephalopathy risk stratification. METHODS/DESIGN: TRUST is a non-interventional, multicenter, prospective cohort study conducted at approximately 200 German neurological centers. The study is intended to enroll 1260 relapsing-remitting multiple sclerosis patients with ongoing natalizumab therapy for at least 12 months. Patients will be followed for a period of 3 years, irrespective of treatment changes after study start. Data on clinical, subclinical and patient-centric outcomes will be documented in order to compare the effectiveness of continuous versus discontinued natalizumab treatment. Furthermore, the type and frequency of clinical, magnetic resonance imaging and biomarker assessments, reasons for continuation or discontinuation of therapy and the safety profile of natalizumab will be collected to explore the impact of a systematic patient management approach and its potential impact on patient outcome. Specifically, the role of biomarkers, the use of expert opinions, the impact of high-frequency magnetic resonance imaging assessment for early progressive multifocal leukoencephalopathy detection and the role of additional radiological and clinical expert advice will be explored. DISCUSSION: TRUST was initiated in spring 2014 and enrollment is anticipated to be completed by mid 2016. Annual interim analyses will deliver continuous information and transparency with regard to the patient cohorts and the completeness and quality of data as well as closely monitor any safety signals in the natalizumab-treated cohort. The study's results may provide insights into opportunities to improve the benefit-risk assessment in clinical practice and support treatment decisions.


Assuntos
Protocolos Clínicos , Gerenciamento Clínico , Leucoencefalopatia Multifocal Progressiva/diagnóstico , Esclerose Múltipla Recidivante-Remitente/tratamento farmacológico , Natalizumab/efeitos adversos , Natalizumab/uso terapêutico , Anticorpos Monoclonais Humanizados/efeitos adversos , Anticorpos Monoclonais Humanizados/uso terapêutico , Feminino , Humanos , Leucoencefalopatia Multifocal Progressiva/induzido quimicamente , Leucoencefalopatia Multifocal Progressiva/complicações , Imageamento por Ressonância Magnética , Masculino , Esclerose Múltipla Recidivante-Remitente/complicações , Estudos Prospectivos , Medição de Risco
2.
Viruses ; 11(12)2019 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-31795203

RESUMO

Marek's disease virus (MDV) is an alphaherpesvirus that causes Marek's disease, a malignant lymphoproliferative disease of domestic chickens. While MDV vaccines protect animals from clinical disease, they do not provide sterilizing immunity and allow field strains to circulate and evolve in vaccinated flocks. Therefore, there is a need for improved vaccines and for a better understanding of innate and adaptive immune responses against MDV infections. Interferons (IFNs) play important roles in the innate immune defenses against viruses and induce upregulation of a cellular antiviral state. In this report, we quantified the potent antiviral effect of IFNα and IFNγ against MDV infections in vitro. Moreover, we demonstrate that both cytokines can delay Marek's disease onset and progression in vivo. Additionally, blocking of endogenous IFNα using a specific monoclonal antibody, in turn, accelerated disease. In summary, our data reveal the effects of IFNα and IFNγ on MDV infection and improve our understanding of innate immune responses against this oncogenic virus.


Assuntos
Galinhas/virologia , Herpesvirus Galináceo 2/imunologia , Interferon-alfa/imunologia , Interferon gama/imunologia , Doença de Marek/virologia , Doenças das Aves Domésticas/virologia , Animais , Anticorpos Monoclonais/imunologia , Progressão da Doença , Imunidade Inata , Doença de Marek/patologia , Doença de Marek/prevenção & controle , Vacinas contra Doença de Marek/imunologia , Doenças das Aves Domésticas/patologia , Doenças das Aves Domésticas/prevenção & controle
3.
Vet J ; 173(3): 697-700, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-16624600

RESUMO

Aural haematomas occur commonly in dogs in which predisposing factors include trauma to the pinna, a history of violent head shaking, and acute or chronic otitis externa. Treatment usually involves invasive surgery performed under general anaesthesia but these techniques can create wounds requiring intensive aftercare. Furthermore, certain breeds of dog and/or older animals are often at greater risk due to complications arising from the use of anaesthetics. Therefore, a need exists for a less invasive procedure which can be performed easily in general practice and with minimal reliance on anaesthetics. Fibrin sealants fulfil these criteria and have been used successfully to treat aural haematomas in humans. This is the first known report on the use of fibrin sealant to treat a canine aural haematoma. In the present case, the haematoma recurred shortly after removal of the protective collar, and was subsequently treated by conventional surgery. Possible reasons for initial success of the fibrin sealant and then later recurrence of the haematoma (e.g. premature removal of the collar) are discussed.


Assuntos
Doenças do Cão/cirurgia , Adesivo Tecidual de Fibrina/uso terapêutico , Hematoma/veterinária , Infecção da Ferida Cirúrgica/veterinária , Cicatrização/efeitos dos fármacos , Animais , Cães , Hematoma/cirurgia , Infecção da Ferida Cirúrgica/prevenção & controle , Resultado do Tratamento
4.
Lancet Haematol ; 4(8): e350-e361, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28712941

RESUMO

BACKGROUND: GP2013 is a rituximab biosimilar developed to stringent development guidelines, including non-clinical and preclinical investigations and clinical trials in rheumatoid arthritis and follicular lymphoma. We aimed to compare the efficacy, safety, tolerability, pharmacokinetics, and pharmacodynamics of GP2013 plus cyclophosphamide, vincristine, and prednisone (GP2013-CVP) with rituximab-CVP (R-CVP) in patients with follicular lymphoma. METHODS: In this phase 3, multinational, double-blind, randomised, controlled trial, adults (aged 18 years or older) with previously untreated, advanced stage (Ann Arbor stage III or IV) follicular lymphoma of WHO histological grades 1, 2, or 3a were randomly assigned (1:1) using interactive response technology to eight cycles of GP2013-CVP or R-CVP (combination phase), followed by monotherapy maintenance in responders for a 2-year period. Randomisation was stratified by Follicular Lymphoma International Prognostic Index risk group and geographic region. The primary endpoint was comparability in overall response, with equivalence concluded if the entire 95% CI was within a margin of -12% to 12%. The primary endpoint was analysed using the per-protocol set, which included all patients who received at least one (partial or complete) dose of investigational treatment and who did not have any major protocol deviations. The trial is registered with ClinicalTrials.gov, number NCT01419665, and is ongoing. FINDINGS: Between Dec 1, 2011, and Jan 15, 2015, 858 patients were screened for eligibility. 314 patients were randomly assigned to GP2013, of whom 312 were given GP2013, and 315 were assigned to reference rituximab. Median follow-up was 11·6 months (IQR 5·8-18·2) for the primary analysis. The primary endpoint, equivalence of overall response, was met (271 [87%] of 311 patients with GP2013 and 274 [88%] of 313 patients with reference rituximab achieved an overall response; difference -0·40% [95% CI -5·94 to 5·14]). Occurrence of adverse events and serious adverse events was similar between the treatment groups (289 [93%] of 312 patients in the GP2013-CVP group had an adverse event and 71 [23%] of 312 patients had a serious adverse event; 288 [91%] of 315 patients in the R-CVP group had an adverse event and 63 [20%] had a serious adverse event). The most common adverse event was neutropenia (80 [26%] of 312 patients in the GP2013-CVP group and 93 [30%] of 315 patients in the R-CVP group in the combination phase and 23 [10%] of 231 patients in the GP2013-CVP group and 13 [6%] of 231 patients in the R-CVP group in the maintenance phase). The most common grade 3 or 4 adverse event during the combination and maintenance phase was neutropenia (55 [18%] of 312 patients in the GP2013-CVP group and 65 [21%] of 315 patients in the R-CVP group in the combination phase and 17 [7%] of 231 patients in the GP2013-CVP group and nine [4%] of 231 patients in the R-CVP group in the maintenance phase). The occurrence of anti-drug antibodies was similar in the treatment groups (five [2%] of 268 patients in the GP2013-CVP; three [1%] in the R-CVP group). INTERPRETATION: Our results show that GP2013 represents a viable rituximab biosimilar candidate for patients with previously untreated advanced follicular lymphoma. The introduction of biosimilars provides additional therapeutic options with potential to increase access to effective and life-saving biological therapies such as rituximab. FUNDING: Hexal.


Assuntos
Medicamentos Biossimilares/uso terapêutico , Linfoma Folicular/tratamento farmacológico , Linfoma Folicular/patologia , Rituximab/uso terapêutico , Adolescente , Adulto , Idoso , Medicamentos Biossimilares/efeitos adversos , Intervalo Livre de Doença , Método Duplo-Cego , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Padrões de Referência , Rituximab/efeitos adversos , Segurança , Adulto Jovem
5.
J Interferon Cytokine Res ; 24(3): 179-84, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15035851

RESUMO

Transcripts of interferon-alpha (IFN-alpha) and IFN-beta genes are present in virus-infected chicken cells, but because of a lack of appropriate assays and reagents, it was unclear if biologically active IFN-beta is secreted. We have established a nonviral bioassay for the sensitive detection of chicken IFN (ChIFN). This assay is based on a quail cell line that carries a luciferase gene that is controlled by the IFN-responsive chicken Mx promoter. Luciferase activity was strongly stimulated when the indicator cells were incubated with ChIFN-alpha, ChIFN-beta, or ChIFN-gamma but not with chicken interleukin-1beta (ChIL-1beta). Unlike the classic antiviral assay that preferentially detects ChIFN-alpha, the Mx-luciferase assay detected ChIFN-alpha and ChIFN-beta with similar sensitivity. With the help of this novel assay and with rabbit antisera specific for either IFN-alpha or IFN-beta, we analyzed the composition of IFN in supernatants of virus-infected chicken embryo cells. Virtually all IFN produced in response to Newcastle disease virus (NDV) was IFN-alpha. However, IFN produced in response to influenza A or vaccinia virus (VV) was a mixture of usually more than 80% IFN-alpha and up to 20% IFN-beta. Thus, IFN-alpha and IFN-beta both contribute to the cytokine activity in supernatants of virus-infected chicken cells. Furthermore, the infecting virus appears to determine the IFN subtype composition.


Assuntos
Bioensaio/métodos , Embrião de Galinha/imunologia , Embrião de Galinha/virologia , Soros Imunes , Interleucina-1/análise , Interleucina-1/biossíntese , Animais , Especificidade de Anticorpos , Linhagem Celular , Proteínas de Ligação ao GTP/genética , Regulação da Expressão Gênica , Genes Reporter , Humanos , Soros Imunes/imunologia , Vírus da Influenza A/patogenicidade , Interleucina-1/imunologia , Luciferases/análise , Luciferases/biossíntese , Luciferases/genética , Proteínas de Resistência a Myxovirus , Vírus da Doença de Newcastle/patogenicidade , Infecções por Orthomyxoviridae/imunologia , Regiões Promotoras Genéticas , Codorniz , Vaccinia virus/patogenicidade
6.
J Virol ; 77(15): 8394-407, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12857909

RESUMO

Highly attenuated modified vaccinia virus Ankara (MVA) serves as a candidate vaccine to immunize against infectious diseases and cancer. MVA was randomly obtained by serial growth in cultures of chicken embryo fibroblasts (CEF), resulting in the loss of substantial genomic information including many genes regulating virus-host interactions. The vaccinia virus interferon (IFN) resistance gene E3L is among the few conserved open reading frames encoding viral immune defense proteins. To investigate the relevance of E3L in the MVA life cycle, we generated the deletion mutant MVA-DeltaE3L. Surprisingly, we found that MVA-DeltaE3L had lost the ability to grow in CEF, which is the first finding of a vaccinia virus host range phenotype in this otherwise highly permissive cell culture. Reinsertion of E3L led to the generation of revertant virus MVA-E3rev and rescued productive replication in CEF. Nonproductive infection of CEF with MVA-DeltaE3L allowed viral DNA replication to occur but resulted in an abrupt inhibition of viral protein synthesis at late times. Under these nonpermissive conditions, CEF underwent apoptosis starting as early as 6 h after infection, as shown by DNA fragmentation, Hoechst staining, and caspase activation. Moreover, we detected high levels of active chicken alpha/beta IFN (IFN-alpha/beta) in supernatants of MVA-DeltaE3L-infected CEF, while moderate IFN quantities were found after MVA or MVA-E3rev infection and no IFN activity was present upon infection with wild-type vaccinia viruses. Interestingly, pretreatment of CEF with similar amounts of recombinant chicken IFN-alpha inhibited growth of vaccinia viruses, including MVA. We conclude that efficient propagation of MVA in CEF, the tissue culture system used for production of MVA-based vaccines, essentially requires conserved E3L gene function as an inhibitor of apoptosis and/or IFN induction.


Assuntos
Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Vaccinia virus/fisiologia , Proteínas Virais/genética , Proteínas Virais/metabolismo , Replicação Viral , Animais , Apoptose , Linhagem Celular , Células Cultivadas , Embrião de Galinha , Galinhas , Cricetinae , DNA Viral/biossíntese , Fibroblastos , Deleção de Genes , Interferon Tipo I/farmacologia , Interferon-alfa/biossíntese , Interferon beta/biossíntese , Coelhos , Proteínas Recombinantes , Vaccinia virus/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA