Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Pharm Res ; 37(7): 120, 2020 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-32514880

RESUMO

PURPOSE: Manufacturing processes for polypeptide/protein drugs are designed to ensure robust quality, efficacy and safety. Process differences introduced by follow-on manufacturers may result in changes in quality and clinical outcomes. This study investigated the impact of production methods on the stability and impurities of liraglutide and semaglutide drug substances/products, and the potential impact on drug quality, efficacy and safety. METHODS: State-of-the-art analytical methods were used to compare physical and chemical stability, and impurity profiles of drug substances/products from different suppliers. Identified polypeptide-related impurities were evaluated for immunogenicity potential by in silico T cell epitope prediction. Semaglutide immunogenicity in clinical trials (SUSTAIN) was evaluated using a tiered antibody analysis. RESULTS: Manufacturing scale and process strongly impacted the physical stability of the products. Trace metals increased high-molecular-weight protein formation for liraglutide and semaglutide. Synthetic and recombinant liraglutide produced by five suppliers had distinct impurity profiles compared with the originator. In silico evaluation suggested that new impurities could be immunogenic. Immunogenicity of semaglutide in clinical trials was lower than for liraglutide. CONCLUSIONS: Differences in manufacturing processes affect chemical/physical stability and impurity profile, and may impact immunogenicity. Follow-on versions of liraglutide and semaglutide, and possibly other polypeptides, should be clinically evaluated for efficacy and safety.


Assuntos
Peptídeos Semelhantes ao Glucagon/farmacologia , Liraglutida/farmacologia , Peptídeos/farmacologia , Sequência de Aminoácidos , Animais , Linhagem Celular , Química Farmacêutica , Simulação por Computador , Cricetinae , Contaminação de Medicamentos , Estabilidade de Medicamentos , Peptídeos Semelhantes ao Glucagon/síntese química , Humanos , Rim/citologia , Liraglutida/síntese química , Metais/análise , Peso Molecular , Peptídeos/síntese química
2.
J Virol ; 87(11): 6283-95, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23536658

RESUMO

It has been reported that adenovirus (Ad)-primed CD8 T cells may display a distinct and partially exhausted phenotype. Given the practical implications of this claim, we decided to analyze in detail the quality of Ad-primed CD8 T cells by directly comparing these cells to CD8 T cells induced through infection with lymphocytic choriomeningitis virus (LCMV). We found that localized immunization with intermediate doses of Ad vector induces a moderate number of functional CD8 T cells which qualitatively match those found in LCMV-infected mice. The numbers of these cells may be efficiently increased by additional adenoviral boosting, and, importantly, the generated secondary memory cells cannot be qualitatively differentiated from those induced by primary infection with replicating virus. Quantitatively, DNA priming prior to Ad vaccination led to even higher numbers of memory cells. In this case, the vaccination led to the generation of a population of memory cells characterized by relatively low CD27 expression and high CD127 and killer cell lectin-like receptor subfamily G member 1 (KLRG1) expression. These memory CD8 T cells were capable of proliferating in response to viral challenge and protecting against infection with live virus. Furthermore, viral challenge was followed by sustained expansion of the memory CD8 T-cell population, and the generated memory cells did not appear to have been driven toward exhaustive differentiation. Based on these findings, we suggest that adenovirus-based prime-boost regimens (including Ad serotype 5 [Ad5] and Ad5-like vectors) represent an effective means to induce a substantially expanded, long-lived population of high-quality transgene-specific memory CD8 T cells.


Assuntos
Adenovírus Humanos/imunologia , Antígenos Virais/imunologia , Linfócitos T CD8-Positivos/imunologia , Vetores Genéticos/imunologia , Glicoproteínas/imunologia , Memória Imunológica , Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/imunologia , Proteínas Virais/imunologia , Adenovírus Humanos/genética , Animais , Antígenos Virais/administração & dosagem , Antígenos Virais/genética , Feminino , Vetores Genéticos/genética , Glicoproteínas/administração & dosagem , Glicoproteínas/genética , Humanos , Coriomeningite Linfocítica/prevenção & controle , Coriomeningite Linfocítica/virologia , Vírus da Coriomeningite Linfocítica/genética , Camundongos , Camundongos Endogâmicos C57BL , Vacinação , Proteínas Virais/administração & dosagem , Proteínas Virais/genética , Vacinas Virais/administração & dosagem , Vacinas Virais/genética , Vacinas Virais/imunologia
3.
J Immunol ; 180(4): 2474-85, 2008 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-18250457

RESUMO

Type III IFNs (IFN-lambda/IL-28/29) are cytokines with type I IFN-like antiviral activities, which remain poorly characterized. We herein show that most cell types expressed both types I and III IFNs after TLR stimulation or virus infection, whereas the ability of cells to respond to IFN-lambda was restricted to a narrow subset of cells, including plasmacytoid dendritic cells and epithelial cells. To examine the role of type III IFN in antiviral defense, we generated IL-28Ralpha-deficient mice. These mice were indistinguishable from wild-type mice with respect to clearance of a panel of different viruses, whereas mice lacking the type I IFN receptor (IFNAR(-/-)) were significantly impaired. However, the strong antiviral activity evoked by treatment of mice with TLR3 or TLR9 agonists was significantly reduced in both IL-28RA(-/-) and IFNAR(-/-) mice. The type I IFN receptor system has been shown to mediate positive feedback on IFN-alphabeta expression, and we found that the type I IFN receptor system also mediates positive feedback on IFN-lambda expression, whereas IL-28Ralpha signaling does not provide feedback on either type I or type III IFN expression in vivo. Finally, using bone-marrow chimeric mice we showed that TLR-activated antiviral defense requires expression of IL-28Ralpha only on nonhemopoietic cells. In this compartment, epithelial cells responded to IFN-lambda and directly restricted virus replication. Our data suggest type III IFN to target a specific subset of cells and to contribute to the antiviral response evoked by TLRs.


Assuntos
Antivirais/metabolismo , Citocinas/fisiologia , Herpes Genital/imunologia , Herpes Genital/prevenção & controle , Receptores Toll-Like/fisiologia , Animais , Antivirais/farmacologia , Cruzamentos Genéticos , Citocinas/biossíntese , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Feminino , Células-Tronco Hematopoéticas/imunologia , Células-Tronco Hematopoéticas/metabolismo , Herpes Genital/metabolismo , Herpesvirus Humano 2/imunologia , Ligantes , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Quimera por Radiação , Receptores de Citocinas/deficiência , Receptores de Citocinas/genética , Receptores Toll-Like/metabolismo
4.
J Gen Virol ; 88(Pt 6): 1708-1716, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17485530

RESUMO

Many novel vaccine strategies rely on recombinant viral vectors for antigen delivery, and adenovirus vectors have emerged among the most potent of these. In this report, we have compared the immune response induced through priming with adenovirus vector-encoded full-length viral protein to that elicited with an adenovirus-encoded minimal epitope covalently linked to beta(2)-microglobulin. We demonstrate that the beta(2)-microglobulin-linked epitope induced an accelerated and augmented CD8(+) T-cell response. Furthermore, the immunity conferred by vaccination with beta(2)-microglobulin-linked lymphocytic choriomeningitis virus (LCMV)-derived epitopes was long-lived and protective. Notably, in contrast to full-length protein, the response elicited with the beta(2)-microglobulin-linked LCMV-derived epitope was CD4(+) T-cell independent. Furthermore, virus-specific CD8(+) T cells primed in the absence of CD4(+) T-cell help were sustained in the long term and able to expand and control a secondary challenge with LCMV. Our results demonstrate that modifications to the antigen used in adenovirus vaccines may be used to improve the induced T-cell response. Such a strategy for CD4(+) T-cell-independent immunity from adenovirus vectors offers prospects for vaccination against opportunistic pathogens in AIDS patients and possibly immunotherapy in chronic virus infections.


Assuntos
Adenoviridae/imunologia , Antígenos Virais/imunologia , Linfócitos T CD8-Positivos/imunologia , Vetores Genéticos , Vacinas Virais/imunologia , Animais , Antígenos Virais/genética , Linfócitos T CD4-Positivos/imunologia , Modelos Animais de Doenças , Epitopos/genética , Epitopos/imunologia , Citometria de Fluxo , Fígado/virologia , Pulmão/virologia , Subpopulações de Linfócitos/imunologia , Coriomeningite Linfocítica/imunologia , Coriomeningite Linfocítica/patologia , Coriomeningite Linfocítica/prevenção & controle , Vírus da Coriomeningite Linfocítica/imunologia , Camundongos , Baço/virologia , Análise de Sobrevida , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Vacinas Virais/genética , Microglobulina beta-2/genética , Microglobulina beta-2/imunologia
5.
J Immunol ; 178(3): 1662-70, 2007 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-17237416

RESUMO

Previous work has shown that agonistic Abs to CD40 (anti-CD40) can boost weak CD8 T cell responses as well as substitute for CD4 T cell function during chronic gammaherpes virus infection. Agonistic anti-CD40 treatment has, therefore, been suggested as a potential therapeutic strategy in immunocompromised patients. In this study, we investigated whether agonistic anti-CD40 could substitute for CD4 T cell help in generating a sustained CD8 T cell response and prevent viral recrudescence following infection with lymphocytic choriomeningitis virus (LCMV). Contrary to expectations, we found that anti-CD40 treatment of MHC class II-deficient mice infected with a moderate dose of LCMV resulted in severe suppression of the antiviral CD8 T cell response and uncontrolled virus spread, rather than improved CD8 T cell immune surveillance. In Ab-treated wild-type mice, the antiviral CD8 T cell response also collapsed prematurely, and virus clearance was delayed. Additional analysis revealed that, following anti-CD40 treatment, the virus-specific CD8 T cells initially proliferated normally, but an increased cell loss compared with that in untreated mice was observed. The anti-CD40-induced abortion of virus-specific CD8 T cells during LCMV infection was IL-12 independent, but depended partly on Fas expression. Notably, similar anti-CD40 treatment of vesicular stomatitis virus-infected mice resulted in an improved antiviral CD8 T cell response, demonstrating that the effect of anti-CD40 treatment varies with the virus infection studied. For this reason, we recommend further evaluation of the safety of this regimen before being applied to human patients.


Assuntos
Anticorpos/efeitos adversos , Antígenos CD40/imunologia , Imunidade/efeitos dos fármacos , Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/imunologia , Animais , Anticorpos/administração & dosagem , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/virologia , Antígenos de Histocompatibilidade Classe II/genética , Terapia de Imunossupressão , Camundongos , Camundongos Knockout
6.
J Virol ; 80(9): 4501-9, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16611910

RESUMO

Type III interferons (IFNs) (interleukin-28/29 or lambda interferon [IFN-lambda]) are cytokines with IFN-like activities. Here we show that several classes of viruses induce expression of IFN-lambda1 and -lambda2/3 in similar patterns. The IFN-lambdas were-unlike alpha/beta interferon (IFN-alpha/beta)-induced directly by stimulation with IFN-alpha or -lambda, thus identifying type III IFNs as IFN-stimulated genes. In vitro assays revealed that IFN-lambdas have appreciable antiviral activity against encephalomyocarditis virus (EMCV) but limited activity against herpes simplex virus type 2 (HSV-2), whereas IFN-alpha potently restricted both viruses. Using three murine models for generalized virus infections, we found that while recombinant IFN-alpha reduced the viral load after infection with EMCV, lymphocytic choriomeningitis virus (LCMV), and HSV-2, treatment with recombinant IFN-lambda in vivo did not affect viral load after infection with EMCV or LCMV but did reduce the hepatic viral titer of HSV-2. In a model for a localized HSV-2 infection, we further found that IFN-lambda completely blocked virus replication in the vaginal mucosa and totally prevented development of disease, in contrast to IFN-alpha, which had a more modest antiviral activity. Finally, pretreatment with IFN-lambda enhanced the levels of IFN-gamma in serum after HSV-2 infection. Thus, type III IFNs are expressed in response to most viruses and display potent antiviral activity in vivo against select viruses. The discrepancy between the observed antiviral activity in vitro and in vivo may suggest that IFN-lambda exerts a significant portion of its antiviral activity in vivo via stimulation of the immune system rather than through induction of the antiviral state.


Assuntos
Antivirais/imunologia , Vírus da Encefalomiocardite/imunologia , Herpesvirus Humano 2/imunologia , Interferons/biossíntese , Interferons/imunologia , Vírus da Coriomeningite Linfocítica/imunologia , Animais , Infecções por Arenaviridae/prevenção & controle , Infecções por Arenaviridae/virologia , Infecções por Cardiovirus/prevenção & controle , Infecções por Cardiovirus/virologia , Linhagem Celular Tumoral , Progressão da Doença , Vírus da Encefalomiocardite/fisiologia , Feminino , Regulação Viral da Expressão Gênica , Herpes Genital/patologia , Herpes Genital/prevenção & controle , Herpes Genital/virologia , Humanos , Interferons/deficiência , Interferons/genética , Vírus da Coriomeningite Linfocítica/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Vagina/patologia , Vagina/virologia
7.
J Virol ; 79(15): 10073-6, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16014969

RESUMO

Bone marrow chimeras were used to determine the cellular target(s) for the antiviral activity of gamma interferon (IFN-gamma). By transfusing such mice with high numbers of naive virus-specific CD8(+) T cells, a system was created in which the majority of virus-specific CD8(+) T cells would be capable of responding to IFN-gamma, but expression of the relevant receptor on non-T cells could be experimentally controlled. Only when the IFN-gamma receptor is absent on both radioresistant parenchymal and bone marrow-derived cells will chimeric mice challenged with a highly invasive, noncytolytic virus completely lack the ability to control the infection and develop severe wasting disease. Further, the study shows that IFN-gamma receptor expression on parenchymal cells in the viscera is more important for virus control than IFN-gamma receptor expression on bone marrow-derived cells.


Assuntos
Infecções por Arenaviridae/imunologia , Infecções por Arenaviridae/virologia , Linfócitos T CD8-Positivos/imunologia , Vírus da Coriomeningite Linfocítica/imunologia , Animais , Infecções por Arenaviridae/patologia , Células da Medula Óssea/imunologia , Quimera , Fígado/imunologia , Fígado/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Interferon/deficiência , Receptores de Interferon/genética , Baço/imunologia , Baço/virologia , Síndrome de Emaciação/patologia , Receptor de Interferon gama
8.
J Immunol ; 173(10): 6284-93, 2004 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-15528367

RESUMO

Induction of a monospecific antiviral CD8+ T cell response may pose a risk to the host due to the narrow T cell response induced. At the individual level, this may result in selection of CD8+ T cell escape variants, particularly during chronic viral infection. Second, prior immunization toward a single dominant epitope may suppress the response to other viral epitopes, and this may lead to increased susceptibility to reinfection with escape variants circulating in the host population. To address these issues, we induced a memory response consisting solely of monospecific, CD8+ T cells by use of DNA vaccines encoding immunodominant epitopes of lymphocytic choriomeningitis virus (LCMV). We analyzed the spectrum of the CD8+ T cell response and the susceptibility to infection in H-2(b) and H-2(d) mice. Priming for a monospecific, CD8+ T cell response did not render mice susceptible to viral variants. Thus, vaccinated mice were protected against chronic infection with LCMV, and no evidence indicating biologically relevant viral escape was obtained. In parallel, a broad and sustained CD8+ T cell response was generated upon infection, and in H-2(d) mice epitope spreading was observed. Even after acute LCMV infection, DNA vaccination did not significantly impair naturally induced immunity. Thus, the response to the other immunogenic epitopes was not dramatically suppressed in DNA-immunized mice undergoing normal immunizing infection, and the majority of mice were protected against rechallenge with escape variants. These findings underscore that a monospecific vaccine may induce efficient protective immunity given the right set of circumstances.


Assuntos
Antivirais/imunologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/virologia , Epitopos de Linfócito T/imunologia , Coriomeningite Linfocítica/prevenção & controle , Vírus da Coriomeningite Linfocítica/imunologia , Vacinas de DNA/imunologia , Vacinas Virais/imunologia , Animais , Antivirais/administração & dosagem , Células Cultivadas , Doença Crônica , Citotoxicidade Imunológica , Suscetibilidade a Doenças/imunologia , Relação Dose-Resposta Imunológica , Epitopos de Linfócito T/administração & dosagem , Epitopos de Linfócito T/genética , Feminino , Variação Genética/imunologia , Antígenos H-2/imunologia , Antígeno de Histocompatibilidade H-2D , Memória Imunológica , Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Prevenção Secundária , Vacinas de DNA/administração & dosagem , Vacinas Virais/administração & dosagem , Vacinas Virais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA