33 research outputs found

    Understanding API static drying with hot gas flow : design and test of a drying rig prototype and drying modelling development

    Get PDF
    Developing a continuous isolation process to produce a pure, dry, free-flowing active pharmaceutical ingredient (API) is the final barrier to the implementation of continuous end-to-end pharmaceutical manufacturing. Recent work has led to the development of continuous filtration and washing prototypes for pharmaceutical process development and small-scale manufacture. Here, we address the challenge of static drying of a solvent-wet crystalline API in a fixed bed to facilitate the design of a continuous filter dryer for pharmaceutical development, without excessive particle breakage or the formation of interparticle bridges leading to lump formation. We demonstrate the feasibility of drying small batches on a time scale suitable for continuous manufacturing, complemented by the development of a drying model that provides a design tool for process development. We also evaluate the impact of alternative washing and drying approaches on particle agglomeration. We conclude that our approach yields effective technology, with a performance that is amenable to predictive modeling

    PLoS One

    Get PDF
    Age-related macular degeneration (AMD) is a common, progressive multifactorial vision-threatening disease and many genetic and environmental risk factors have been identified. The risk of AMD is influenced by lifestyle and diet, which may be reflected by an altered metabolic profile. Therefore, measurements of metabolites could identify biomarkers for AMD, and could aid in identifying high-risk individuals. Hypothesis-free technologies such as metabolomics have a great potential to uncover biomarkers or pathways that contribute to disease pathophysiology. To date, only a limited number of metabolomic studies have been performed in AMD. Here, we aim to contribute to the discovery of novel biomarkers and metabolic pathways for AMD using a targeted metabolomics approach of 188 metabolites. This study focuses on non-advanced AMD, since there is a need for biomarkers for the early stages of disease before severe visual loss has occurred. Targeted metabolomics was performed in 72 patients with early or intermediate AMD and 72 control individuals, and metabolites predictive for AMD were identified by a sparse partial least squares discriminant analysis. In our cohort, we identified four metabolite variables that were most predictive for early and intermediate stages of AMD. Increased glutamine and phosphatidylcholine diacyl C28:1 levels were detected in non-advanced AMD cases compared to controls, while the rate of glutaminolysis and the glutamine to glutamate ratio were reduced in non-advanced AMD. The association of glutamine with non-advanced AMD corroborates a recent report demonstrating an elevated glutamine level in early AMD using a different metabolomics technique. In conclusion, this study indicates that metabolomics is a suitable method for the discovery of biomarker candidates for AMD. In the future, larger metabolomics studies could add to the discovery of novel biomarkers in yet unknown AMD pathways and expand our insights in AMD pathophysiology

    Fhit implication in the tumor invasion process

    No full text
    Dans de nombreux cancers, l’expression du gène Fhit (Fragile Histidine Triad) estfréquemment altérée. Fhit est décrit comme un important gène suppresseur de tumeur de parson rôle pro-apoptotique et anti-prolifératif. Nous avons mis en évidence que, in vivo et invitro, la diminution de l’expression de Fhit est associée au caractère infiltrant des cellulestumorales bronchiques, ce qui suggère que Fhit pourrait être impliqué dans le processusd’invasion tumorale. Nous avons en effet montré que la surexpression et l’inhibition de Fhitinduisent respectivement une diminution et une augmentation des capacités migratoires etinvasives des cellules bronchiques. Nous avons aussi mis en évidence que Fhit contrôlel’invasion des cellules tumorales bronchiques en régulant l’expression d’éléments clés de latransition épithélio-mésenchymateuse (TEM) tels que l’organisation des jonctions serrées etadhérentes, l’expression des métalloprotéinases matricielles et de la vimentine. De plus, Fhitrégule la TEM via une cascade de signalisation impliquant le récepteur au TGF-β, lesrécepteurs à tyrosine kinase (RTK), Src, Erk et Slug. Le double rôle de Fhit en tant quesuppresseur de tumeur et d’invasion renforce l’idée que Fhit pourrait représenter un nouveaubiomarqueur d’agressivité tumorale et pourrait constituer une nouvelle cible thérapeutiquedans le traitement des cancers broncho-pulmonaires.In many types of cancers, Fhit (Fragile histidine triad) expression is frequentlydecreased or lost. Fhit is described as a tumor suppressor gene by its ability to induceapoptosis and to inhibit proliferation of tumor cells. We have demonstrated that a low Fhitexpression is associated with in vivo and in vitro invasiveness of lung tumor cells. Then, wehave shown that Fhit controls the invasive phenotype of lung tumor cells by regulating keyelements of epithelial-mesenchymal transition (EMT) such as cell-cell adhesion molecules,matrix metalloproteinase and vimentin expression. Our results provide also evidence that Fhitcontrols EMT by regulating several signaling pathways implying TGF-βR, RTK, Src, ERKand Slug. The dual function of Fhit as a tumor and invasion suppressor gene strengthens theidea that Fhit could represent a new biomarker of aggressiveness of lung cancer and couldconstitute a new therapeutic target to limit tumor progression

    New targets in idiopathic pulmonary fibrosis: from inflammation and immunity to remodeling and repair

    No full text
    Introduction: Idiopathic pulmonary fibrosis (IPF) is a chronic respiratory disease whose incidence and prevalence are increasing. Recent evidence has led to a change of paradigm regarding understanding of the underlying pathophysiology. In parallel, we have witnessed the development and rise of the first anti-fibrotic drugs, namely pirfenidone and nintedanib. However, with clinical results being below expectations there is a clear need for new medications in this field. Areas covered: After covering new mechanisms involved in IPF pathogenesis, the present review discusses current clinical trials and deciphers potential new targets in light of in vitro and in vivo experimental studies. Expert Opinion: All in all, we believe that future development will require (1) a significant improvement in experimental models, (2) a proper selection and characterization of patients, allowing us to foresee which will respond to a given treatment and (3) an evaluation of combined therapies, targeting different pathways

    Alteration of human macrophage phenotypes by the anti-fibrotic drug nintedanib

    No full text
    International audienceThe tyrosine kinase inhibitor, Nintedanib (NTD), has been approved for the treatment of idiopathic pulmonary fibrosis (IPF). In cell-free systems, NTD was recently shown to inhibit kinase activity of the human recombinant colony-stimulating factor 1 (CSF1) receptor (CSF1R) which mediates major functions of pulmonary macrophages. In the present study, we have investigated the effects of NTD on the phenotype of human monocyte-derived macrophages controlled by CSF1 in order to identify its anti-inflammatory properties via CSF1R inhibition. NTD (0.01 to 1 μM) prevented the CSF1-induced phosphorylation of CSF1R and activation of the downstream signaling pathways. NTD, like the CSF1R inhibitor GW2580, significantly decreased the adhesion of macrophages and production of the chemokine ligand (CCL) 2. NTD also altered the polarization of macrophages to classical M1 and alternative M2a macrophages. It reduced the secretion of several pro-inflammatory and/or pro-fibrotic cytokines (IL-1β, IL-8, IL-10 and CXCL13) by M1 macrophages but did not prevent the expression of M1 markers. While NTD (50-200 nM) partially blocked the synthesis of M2a markers (CD11b, CD200R, CD206, and CD209), it did not reduce synthesis of the M2a pro-fibrotic cytokines CCL22 and PDGF-BB, and increased CCL18 release when used at its highest concentration (1 μM). The effects of NTD on macrophage polarization only was partially mimicked by GW2580, suggesting that the drug inhibits other molecules in addition to CSF1R. In conclusion, NTD alters the CSF1-controlled phenotype of human macrophages mainly by blocking the activation of CSF1R that thus constitutes a new molecular target of NTD, at least in vitro

    Effects of Ruxolitinib on fibrosis in preclinical models of systemic sclerosis

    No full text
    International audienceSystemic sclerosis (SSc) is an autoimmune fibrotic disorder notably characterized by the production of antinuclear autoantibodies, which have been linked to an excess of apoptotic cells, normally eliminated by a macrophagic efferocytosis. As interferon (IFN) signature and phosphorylation of JAK-STAT proteins are hallmarks of SSc tissues, we tested the hypothesis that a JAK inhibitor, ruxolitinib, targeting the IFN signaling, could improve efferocytosis of IFN-exposed human macrophages in vitro as well as skin and lung fibrosis. In vivo, BLM- and HOCl-induced skin thickness and fibrosis is associated with an increase of caspase-3 positive dermal cells and a significant increase of IFN-stimulated genes expression. In BLM-SSc model, ruxolitinib prevented dermal thickness, fibrosis and significantly decreased the number of cleaved caspase-3 cells in the dermis. Ruxolitinib also improved lung architecture and fibrosis although IFN signature was not entirely decreased by ruxolitinib. In vitro, ruxolitinib improves efferocytosis capacity of human monocyte-differentiated macrophages exposed to IFN-γ or IFN-β. In human fibroblasts derived from lung (HLF) biopsies isolated from patients with idiopathic pulmonary fibrosis, the reduced mRNA expression of typical TGF-β-activated markers by ruxolitinib was associated with a decrease of the phosphorylation of SMAD2 /3 and STAT3. Our finding supports the anti-fibrotic properties of ruxolitinib in a systemic SSc mouse model and in vitro in human lung fibroblasts

    Fhit regulates EMT targets through an EGFR/Src/ERK/Slug signaling axis in human bronchial cells.

    Full text link
    In many cancers, including lung carcinomas, Fragile histidine triad (Fhit) is frequently decreased or lost. Fhit status has recently been shown to be associated with elevated in vitro and in vivo invasiveness in lung cancer. Tumor cell invasion is facilitated by epithelial-mesenchymal transition (EMT), a process by which tumor cells lose their epithelial features to acquire a mesenchymal cell-like phenotype. In this study, the mechanism underlying Fhit-regulated EMT was deciphered. Using Slug knockdown, pharmacologic inhibitors PD98059, PP1, and gefitinib as well as an anti-EGFR antibody, it was demonstrated that Fhit silencing in bronchial cells induced overexpression of two primary EMT-associated targets, MMP-9 and vimentin, to regulate cell invasion dependent on an EGFR/Src/ERK/Slug signaling pathway. Moreover, ectopic expression of Fhit in Fhit-deficient lung cancer cells downregulated this pathway. Finally, an inverse correlation was observed between Fhit and phospho-EGFR levels in a cohort of human squamous cell lung carcinoma specimens. These results demonstrate a Fhit-dependent mechanism in the control of EMT-regulated EGFR signaling. IMPLICATIONS: This study adds new insight into the regulatory mechanism of EMT, a process known to increase resistance to conventional and targeted therapies in lung cancer

    Identification of periplakin as a major regulator of lung injury and repair in mice

    No full text
    IF 12.784 (2016)International audiencePeriplakin is a component of the desmosomes that acts as a cytolinker between intermediate filament scaffolding and the desmosomal plaque. Periplakin is strongly expressed by epithelial cells in the lung and is a target antigen for autoimmunity in idiopathic pulmonary fibrosis. The aim of this study was to determine the role of periplakin during lung injury and remodeling in a mouse model of lung fibrosis induced by bleomycin. We found that periplakin expression was downregulated in the whole lung and in alveolar epithelial cells following bleomycin-induced injury. Deletion of the Ppl gene in mice improved survival and reduced lung fibrosis development after bleomycin-induced injury. Notably, Ppl deletion promoted an antiinflammatory alveolar environment linked to profound changes in type 2 alveolar epithelial cells, including overexpression of antiinflammatory cytokines, decreased expression of profibrotic mediators, and altered cell signaling with a reduced response to TGF-β1. These results identify periplakin as a previously unidentified regulator of the response to injury in the lung
    corecore