Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
3.
Eur J Pharm Sci ; 192: 106670, 2024 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-38070782

RESUMO

Aggregation has been widely described as a factor contributing to therapeutic antibody immunogenicity. Although production of high-affinity anti-drug antibodies depends on the activation of CD4 T lymphocytes, little is known about the T-cell response induced by antibody aggregates, especially for aggregates produced in mild conditions resulting from minor handling errors of vials. Large insoluble infliximab (IFX) aggregates produced in severe elevated temperature stress conditions have been previously shown to induce human monocyte-derived dendritic cell (moDC) maturation. We here showed that large IFX aggregates recruit in vitro a significantly higher number of CD4 T-cells compared to native IFX. Moreover, a larger array of T-cell epitopes encompassing the entire variable regions was evidenced compared to the native antibody. We then compared the responses of moDCs to different types of aggregates generated by submitting IFX to mild conditions of various times of incubation at an elevated temperature. Decreasing stress duration reduced aggregate size and quantity, and subsequently altered moDC activation. Of importance, IFX aggregates generated in mild conditions and not altering moDC phenotype generated an in vitro T-cell response with a higher frequency of CD4 T cells compared to native IFX. Moreover, cross-reactivity studies of aggregate-specific T cells showed that some T cells could recognize both native and aggregated IFX, while others responded only to IFX aggregates. Taken together, our results suggest that aggregation of antibodies in mild elevated temperature stress conditions is sufficient to alter moDC phenotype in a dose-dependent manner and to increase T-cell response.


Assuntos
Linfócitos T CD4-Positivos , Monócitos , Humanos , Infliximab/farmacologia , Linfócitos T CD4-Positivos/metabolismo , Temperatura
5.
Cancer Immunol Res ; 11(8): 1125-1136, 2023 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-37229623

RESUMO

Single-cell technologies have elucidated mechanisms responsible for immune checkpoint inhibitor (ICI) response, but are not amenable to a clinical diagnostic setting. In contrast, bulk RNA sequencing (RNA-seq) is now routine for research and clinical applications. Our workflow uses transcription factor (TF)-directed coexpression networks (regulons) inferred from single-cell RNA-seq data to deconvolute immune functional states from bulk RNA-seq data. Regulons preserve the phenotypic variation in CD45+ immune cells from metastatic melanoma samples (n = 19, discovery dataset) treated with ICIs, despite reducing dimensionality by >100-fold. Four cell states, termed exhausted T cells, monocyte lineage cells, memory T cells, and B cells were associated with therapy response, and were characterized by differentially active and cell state-specific regulons. Clustering of bulk RNA-seq melanoma samples from four independent studies (n = 209, validation dataset) according to regulon-inferred scores identified four groups with significantly different response outcomes (P < 0.001). An intercellular link was established between exhausted T cells and monocyte lineage cells, whereby their cell numbers were correlated, and exhausted T cells predicted prognosis as a function of monocyte lineage cell number. The ligand-receptor expression analysis suggested that monocyte lineage cells drive exhausted T cells into terminal exhaustion through programs that regulate antigen presentation, chronic inflammation, and negative costimulation. Together, our results demonstrate how regulon-based characterization of cell states provide robust and functionally informative markers that can deconvolve bulk RNA-seq data to identify ICI responders.


Assuntos
Redes Reguladoras de Genes , Melanoma , Humanos , Melanoma/tratamento farmacológico , Melanoma/genética , Imunoterapia , Leucócitos , Apresentação de Antígeno
6.
Part Fibre Toxicol ; 20(1): 12, 2023 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-37076877

RESUMO

BACKGROUND: Synthetic amorphous silica nanoparticles (SAS-NPs) are widely employed in pharmaceutics, cosmetics, food and concretes. Workers and the general population are exposed daily via diverse routes of exposure. SAS-NPs are generally recognized as safe (GRAS) by the Food and Drug Administration, but because of their nanoscale size and extensive uses, a better assessment of their immunotoxicity is required. In the presence of immune "danger signals", dendritic cells (DCs) undergo a maturation process resulting in their migration to regional lymph nodes where they activate naive T-cells. We have previously shown that fumed silica pyrogenic SAS-NPs promote the two first steps of the adaptative immune response by triggering DC maturation and T-lymphocyte response, suggesting that SAS-NPs could behave as immune "danger signals". The present work aims to identify the mechanism and the signalling pathways involved in DC phenotype modifications provoked by pyrogenic SAS-NPs. As a pivotal intracellular signalling molecule whose phosphorylation is associated with DC maturation, we hypothesized that Spleen tyrosine kinase (Syk) may play a central role in SAS-NPs-induced DC response. RESULTS: In human monocyte-derived dendritic cells (moDCs) exposed to SAS-NPs, Syk inhibition prevented the induction of CD83 and CD86 marker expression. A significant decrease in T-cell proliferation and IFN-γ, IL-17F and IL-9 production was found in an allogeneic moDC:T-cell co-culture model. These results suggested that the activation of Syk was necessary for optimal co-stimulation of T-cells. Moreover, Syk phosphorylation, observed 30 min after SAS-NP exposure, occurred upstream of the c-Jun N-terminal kinase (JNK) Mitogen-activated protein kinases (MAPK) and was elicited by the Src family of protein tyrosine kinases. Our results also showed for the first time that SAS-NPs provoked aggregation of lipid rafts in moDCs and that MßCD-mediated raft destabilisation altered Syk activation. CONCLUSIONS: We showed that SAS-NPs could act as an immune danger signal in DCs through a Syk-dependent pathway. Our findings revealed an original mechanism whereby the interaction of SAS-NPs with DC membranes promoted aggregation of lipid rafts, leading to a Src kinase-initiated activation loop triggering Syk activation and functional DC maturation.


Assuntos
Nanopartículas , Dióxido de Silício , Humanos , Dióxido de Silício/toxicidade , Dióxido de Silício/metabolismo , Proteínas Tirosina Quinases/metabolismo , Fosforilação , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Nanopartículas/toxicidade , Células Dendríticas , Quinase Syk/metabolismo
7.
Eur J Immunol ; 53(3): e2350417, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36758120
8.
J Pharm Sci ; 110(2): 952-958, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33220239

RESUMO

Sucrose is a commonly used stabilizing excipient in protein formulations. However, recent studies have indicated the presence of nanoparticulate impurities (NPIs) in the size range of 100-200 nm in pharmaceutical-grade sucrose. Furthermore, isolated NPIs have been shown to induce protein aggregation when added to monoclonal antibody formulations. Moreover, nanoparticles are popular vaccine delivery systems used to increase the immunogenicity of antigens. Therefore, we hypothesized that NPIs may have immunostimulatory properties. In this study, we evaluated the immunomodulatory effects of NPIs in presence and absence of trastuzumab in vitro with monocyte-derived dendritic cells (moDCs). Exposure of trastuzumab, the model IgG used in this study, to NPIs led to an increase in concentration of proteinaceous particles in the sub-micron range. When added to moDCs, the NPIs alone or in presence of trastuzumab did not affect cell viability or cytotoxicity. Moreover, no significant effect on the expression of surface markers, and cytokine and chemokine production was observed. Our findings showed, surprisingly, no evidence of any immunomodulatory activity of NPIs. As this study was limited to a single IgG formulation and to in vitro immunological read-outs, further work is required to fully understand the immunogenic potential of NPIs.


Assuntos
Nanopartículas , Preparações Farmacêuticas , Anticorpos Monoclonais , Excipientes , Sacarose
9.
J Immunol ; 205(9): 2351-2361, 2020 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-32989091

RESUMO

The development of anti-drug Abs in response to biological products (BP) is a major drawback in the treatment of patients. Factors related to the patient, the treatment, and the product can influence BP immunogenicity. Among these factors, BP aggregates have been suggested to promote immunogenicity by acting as danger signals recognized by dendritic cells (DC) facilitating the establishment of an anti-BP CD4 T cell-dependent adaptive immune response leading to anti-drug Abs production. To date, little is known on the mechanism supporting the effect of aggregates on DCs and consequently on the T cell response. The aim of this work was to identify key signaling pathways involved in BP aggregate DC activation and T cell response. We generated aggregates by submitting infliximab (IFX), an immunogenic anti-TNF-α chimeric Ab, to heat stress. Our results showed that IFX aggregates were able to induce human monocyte-derived DC (moDC) maturation in a concentration-dependent manner. Aggregate-treated moDCs enhanced allogeneic T cell proliferation and IL-5, IL-9, and IL-13 production compared with native Ab-treated moDCs. We then investigated the implication of FcγRIIa and spleen tyrosine kinase (Syk) in DC activation and showed that they were both strongly implicated in moDC maturation induced by IFX aggregates. Indeed, we found that neutralization of FcγRIIa inhibited DC activation, and consequently, Syk inhibition led to a decrease in T cell proliferation and cytokine production in response to IFX aggregates. Taken together, our results bring new insight, to our knowledge, on how protein aggregates could induce DC and T cell activation via the FcγRIIa-Syk signaling pathway.


Assuntos
Células Dendríticas/imunologia , Infliximab/imunologia , Ativação Linfocitária/imunologia , Receptores de IgG/imunologia , Quinase Syk/imunologia , Linfócitos T/imunologia , Proliferação de Células/fisiologia , Células Cultivadas , Humanos , Monócitos/imunologia , Transdução de Sinais/imunologia , Fator de Necrose Tumoral alfa/imunologia
10.
Front Immunol ; 11: 725, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32431697

RESUMO

Patients treated with bioproducts (BPs) frequently develop anti-drug antibodies (ADAs) with potential neutralizing capacities leading to loss of clinical response or potential hypersensitivity reactions. Many factors can influence BP immunogenicity and could be related to the patient, the treatment, as well as to the product itself. Among these latter factors, it is now well accepted that BP aggregation is associated with an increased potential for immunogenicity, as aggregates seem to be correlated with ADA development. Moreover, the presence of high-affinity ADAs suggests a CD4 T-cell dependent adaptive immune response and therefore a pivotal role for antigen-presenting cells (APCs), such as dendritic cells (DCs). In this review, we address the in vitro methods developed to evaluate how monoclonal antibodies could trigger the immunization process by focusing on the role of aggregated antibodies in the establishment of this response. In particular, we will present the different cell-based assays that have been used to assess the potential of antibodies and their aggregates to modulate cellular mechanisms leading to activation and the biological parameters (cellular activation markers, proliferation and secreted molecules) that can be measured to evaluate the different cell activation stages and their consequences in the propagation of the immune response. Indeed, the use of such strategies could help evaluate the risk of BP immunogenicity and their role in mitigating this risk.


Assuntos
Imunidade Adaptativa , Anticorpos Monoclonais/imunologia , Produtos Biológicos/imunologia , Modelos Imunológicos , Agregados Proteicos/imunologia , Agregação Patológica de Proteínas/imunologia , Anticorpos Monoclonais/uso terapêutico , Produtos Biológicos/uso terapêutico , Linfócitos T CD4-Positivos/imunologia , Células Dendríticas/imunologia , Humanos , Imunização
11.
J Pharm Sci ; 109(1): 927-932, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31520643

RESUMO

The presence of protein aggregates in biological products is suggested to promote immunogenicity, leading to the production of anti-drug antibodies with neutralizing capacities. This suggests a CD4+ T-cell dependent adaptive immune response, thus a pivotal role for antigen-presenting cells, such as dendritic cells (DCs). We previously showed that human growth hormone aggregates induced DC maturation, with notably an increase in CXCL10 production. DC phenotypic modifications were sufficient to promote allogeneic CD4+ T-cell proliferation with Th1 polarization. In this work, we identified the main intracellular signaling pathways involved in DC activation by human growth hormone aggregates, showing that aggregates induced p38 mitogen-activated protein kinase, extracellular signal-regulated kinase, and c-Jun N-terminal kinase phosphorylation, as well as nuclear factor κB subunit p65 nuclear translocation. Next, investigating the implication of Rho GTPases and phosphoinositide 3-kinase (PI3K) in activated DC showed that Rac1 and Cdc42 regulated the phosphorylation of MAP kinases, whereas PI3K was only implicated in c-Jun N-terminal kinase phosphorylation. Furthermore, we showed that Rac1 and PI3K pathways, but not Cdc42, regulated the production of CXCL10 via the MAP kinases and nuclear factor κB. Taken together, our results bring new insight on how protein aggregates could induce DC activation, leading to a better understanding of aggregates role in therapeutic proteins immunogenicity.


Assuntos
Células Dendríticas/efeitos dos fármacos , Hormônio do Crescimento Humano/farmacologia , Fosfatidilinositol 3-Quinase/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Células Cultivadas , Quimiocina CXCL10/genética , Quimiocina CXCL10/metabolismo , Células Dendríticas/enzimologia , Células Dendríticas/imunologia , Composição de Medicamentos , Hormônio do Crescimento Humano/química , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosforilação , Agregados Proteicos , Transdução de Sinais , Regulação para Cima
12.
Front Immunol ; 10: 601, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31001248

RESUMO

Therapeutic antibodies have the potential to induce immunogenicity leading to the development of anti-drug antibodies (ADA) that consequently may result in reduced serum drug concentrations, a loss of efficacy or potential hypersensitivity reactions. Among other factors, aggregated antibodies have been suggested to promote immunogenicity, thus enhancing ADA production. Dendritic cells (DC) are the most efficient antigen-presenting cell population and are crucial for the initiation of T cell responses and the subsequent generation of an adaptive immune response. This work focuses on the development of predictive in vitro assays that can monitor DC maturation, in order to determine whether drug products have direct DC stimulatory capabilities. To this end, four independent laboratories aligned a common protocol to differentiate human monocyte-derived DC (moDC) that were treated with either native or aggregated preparations of infliximab, natalizumab, adalimumab, or rituximab. These drug products were subjected to different forms of physical stress, heat and shear, resulting in aggregation and the formation of subvisible particles. Each partner developed and optimized assays to monitor diverse end-points of moDC maturation: measuring the upregulation of DC activation markers via flow cytometry, analyzing cytokine, and chemokine production via mRNA and protein quantification and identifying cell signaling pathways via quantification of protein phosphorylation. These study results indicated that infliximab, with the highest propensity to form aggregates when heat-stressed, induced a marked activation of moDC as measured by an increase in CD83 and CD86 surface expression, IL-1ß, IL-6, IL-8, IL-12, TNFα, CCL3, and CCL4 transcript upregulation and release of respective proteins, and phosphorylation of the intracellular signaling proteins Syk, ERK1/2, and Akt. In contrast, natalizumab, which does not aggregate under these stress conditions, induced no DC activation in any assay system, whereas adalimumab or rituximab aggregates induced only slight parameter variation. Importantly, the data generated in the different assay systems by each partner site correlated and supported the use of these assays to monitor drug-intrinsic propensities to drive maturation of DC. This moDC assay is also a valuable tool as an in vitro model to assess the intracellular mechanisms that drive DC activation by aggregated therapeutic proteins.


Assuntos
Anticorpos Monoclonais/farmacologia , Células Dendríticas/efeitos dos fármacos , Bioensaio , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Células Dendríticas/metabolismo , Humanos
13.
Chem Res Toxicol ; 31(12): 1323-1331, 2018 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-30421605

RESUMO

Allergic contact dermatitis (ACD) is a major cause of occupational skin disease, and nickel is among the most prevalent contact allergens. Dendritic cells (DC) play an important role in ACD and in the type of the ensuing immune response through differential phenotypes and cytokine production. The interleukin (IL)-12 cytokine family is composed of heterodimeric cytokines sharing homology at the subunit, receptors and signaling levels. We previously showed that nickel can upregulate the production of IL-12p40 and IL-23, both known to be pro-inflammatory. In this work, we aimed to extend our knowledge on nickel regulation of the IL-12 cytokine family by focusing on IL-27, a recently identified immunomodulatory cytokine from this family. We showed that nickel induced the production of IL-27 in human monocyte-derived DC (MoDC), regulating IL-22 production by human CD4+ T cells. We also showed that nickel was able to induce the expression of the two subunits of IL-27: il-27p28 and ebi3. Furthermore, we demonstrated that the production of IL-27 was dependent on the TLR4, p38 MAPK, NF-κB, and Jak-STAT signaling. However, IL-27 subunits were differentially regulated by these pathways. Indeed, both subunits were positively regulated by the TLR4 and NF-κB pathways, but only il-27p28 was also dependent on p38 MAPK and Jak-STAT pathways. Our results contribute to a better understanding of nickel-induced ACD by focusing on the IL-12 cytokine family and elucidating the mechanism of IL-27 regulation in human dendritic cells.


Assuntos
Células Dendríticas/efeitos dos fármacos , Interleucina-27/metabolismo , Níquel/toxicidade , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Células Cultivadas , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Dermatite Alérgica de Contato/metabolismo , Dermatite Alérgica de Contato/patologia , Humanos , Interleucinas/metabolismo , Monócitos/citologia , NF-kappa B/metabolismo , Níquel/química , Transdução de Sinais/efeitos dos fármacos , Receptor 4 Toll-Like/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Interleucina 22
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA