Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 104
Filtrar
3.
Clin Immunol ; 264: 110241, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38735508

RESUMEN

Primary Sjögren disease (pSD) is an autoimmune disease characterized by lymphoid infiltration of exocrine glands leading to dryness of the mucosal surfaces and by the production of autoantibodies. The pathophysiology of pSD remains elusive and no treatment with demonstrated efficacy is available yet. To better understand the biology underlying pSD heterogeneity, we aimed at identifying Consensus gene Modules (CMs) that summarize the high-dimensional transcriptomic data of whole blood samples in pSD patients. We performed unsupervised gene classification on four data sets and identified thirteen CMs. We annotated and interpreted each of these CMs as corresponding to cell type abundances or biological functions by using gene set enrichment analyses and transcriptomic profiles of sorted blood cell subsets. Correlation with independently measured cell type abundances by flow cytometry confirmed these annotations. We used these CMs to reconcile previously proposed patient stratifications of pSD. Importantly, we showed that the expression of modules representing lymphocytes and erythrocytes before treatment initiation is associated with response to hydroxychloroquine and leflunomide combination therapy in a clinical trial. These consensus modules will help the identification and translation of blood-based predictive biomarkers for the treatment of pSD.


Asunto(s)
Biomarcadores , Síndrome de Sjögren , Humanos , Síndrome de Sjögren/genética , Síndrome de Sjögren/sangre , Biomarcadores/sangre , Transcriptoma , Perfilación de la Expresión Génica/métodos , Hidroxicloroquina/uso terapéutico , Femenino , Redes Reguladoras de Genes , Linfocitos/metabolismo
4.
Med Sci (Paris) ; 40(4): 369-376, 2024 Apr.
Artículo en Francés | MEDLINE | ID: mdl-38651962

RESUMEN

Artificial intelligence and machine learning enable the construction of predictive models, which are currently used to assist in decision-making throughout the process of drug discovery and development. These computational models can be used to represent the heterogeneity of a disease, identify therapeutic targets, design and optimize drug candidates, and evaluate the efficacy of these drugs on virtual patients or digital twins. By combining detailed patient characteristics with the prediction of potential drug-candidate properties, artificial intelligence promotes the emergence of a "computational" precision medicine, allowing for more personalized treatments, better tailored to patient specificities with the aid of such predictive models. Based on such new capabilities, a mixed reality approach to the development of new drugs is being adopted by the pharmaceutical industry, which integrates the outputs of predictive virtual models with real-world empirical studies.


Title: L'intelligence artificielle, une révolution dans le développement des médicaments. Abstract: L'intelligence artificielle (IA) et l'apprentissage automatique produisent des modèles prédictifs qui aident à la prise de décisions dans le processus de découverte de nouveaux médicaments. Cette modélisation par ordinateur permet de représenter l'hétérogénéité d'une maladie, d'identifier des cibles thérapeutiques, de concevoir et optimiser des candidats-médicaments et d'évaluer ces médicaments sur des patients virtuels, ou des jumeaux numériques. En facilitant à la fois une connaissance détaillée des caractéristiques des patients et en prédisant les propriétés de multiples médicaments possibles, l'IA permet l'émergence d'une médecine de précision « computationnelle ¼ offrant des traitements parfaitement adaptés aux spécificités des patients.


Asunto(s)
Inteligencia Artificial , Desarrollo de Medicamentos , Medicina de Precisión , Inteligencia Artificial/tendencias , Humanos , Desarrollo de Medicamentos/métodos , Desarrollo de Medicamentos/tendencias , Medicina de Precisión/métodos , Medicina de Precisión/tendencias , Descubrimiento de Drogas/métodos , Descubrimiento de Drogas/tendencias , Aprendizaje Automático , Simulación por Computador
5.
J Autoimmun ; : 103147, 2023 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-38114349

RESUMEN

OBJECTIVE: While the involvement of IL-7/IL-7R axis in pSS has been described in relation to T cells, little is known about the contribution of this pathway in relationship with other immune cells, and its implication in autoimmunity. Using high-content multiomics data, we aimed at characterizing IL-7R expressing cells and the involvement of IL-7/IL-7R pathway in pSS pathophysiology. METHODS: An IL-7 signature established using RNA-sequencing of human PBMCs incubated with IL-7 was applied to 304 pSS patients, and on RNA-Seq datasets from tissue biopsies. High-content immunophenotyping using flow and imaging mass cytometry was developed to characterize peripheral and in situ IL-7R expression. RESULTS: We identified a blood 4-gene IL-7 module (IKZF4, KIAA0040, PGAP1 and SOS1) associated with anti-SSA/Ro positiveness in patients as well as disease activity, and a tissue 5-gene IL-7 module (IL7R, PCED1B, TNFSF8, ADAM19, MYBL1) associated with infiltration severity. We confirmed expression of IL-7R on T cells subsets, and further observed upregulation of IL-7R on double-negative (DN) B cells, and especially DN2 B cells. IL-7R expression was increased in pSS compared to sicca patients with variations seen according to the degree of infiltration. When expressed, IL-7R was mainly found on epithelial cells, CD4+ and CD8+ T cells, switched memory B cells, DN B cells and M1 macrophages. CONCLUSION: This exhaustive characterization of the IL-7/IL-7R pathway in pSS pathophysiology established that two IL-7 gene modules discriminate pSS patients with a high IL-7 axis involvement. Their use could guide the implementation of an anti-IL-7R targeted therapy in a precision medicine approach.

6.
Drug Discov Today ; 28(11): 103772, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37717933

RESUMEN

High-throughput computational platforms are being established to accelerate drug discovery. Servier launched the Patrimony platform to harness computational sciences and artificial intelligence (AI) to integrate massive multimodal data from internal and external sources. Patrimony has enabled researchers to prioritize therapeutic targets based on a deep understanding of the pathophysiology of immuno-inflammatory diseases. Herein, we share our experience regarding main challenges and critical success factors faced when industrializing the platform and broadening its applications to neurological diseases. We emphasize the importance of integrating such platforms in an end-to-end drug discovery process and engaging human experts early on to ensure a transforming impact.


Asunto(s)
Inteligencia Artificial , Descubrimiento de Drogas , Humanos , Investigadores
7.
Trends Pharmacol Sci ; 44(7): 411-424, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37268540

RESUMEN

Artificial intelligence (AI)-based predictive models are being used to foster a precision medicine approach to treat complex chronic diseases such as autoimmune and autoinflammatory disorders (AIIDs). In the past few years the first models of systemic lupus erythematosus (SLE), primary Sjögren syndrome (pSS), and rheumatoid arthritis (RA) have been produced by molecular profiling of patients using omic technologies and integrating the data with AI. These advances have confirmed a complex pathophysiology involving multiple proinflammatory pathways and also provide evidence for shared molecular dysregulation across different AIIDs. I discuss how models are used to stratify patients, assess causality in pathophysiology, design drug candidates in silico, and predict drug efficacy in virtual patients. By relating individual patient characteristics to the predicted properties of millions of drug candidates, these models can improve the management of AIIDs through more personalized treatments.


Asunto(s)
Artritis Reumatoide , Enfermedades Autoinmunes , Síndrome de Sjögren , Humanos , Síndrome de Sjögren/tratamiento farmacológico , Inteligencia Artificial , Enfermedades Autoinmunes/tratamiento farmacológico , Artritis Reumatoide/tratamiento farmacológico , Desarrollo de Medicamentos
8.
Drug Discov Today ; 28(7): 103605, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37146963

RESUMEN

Computational models are being explored to simulate in silico the efficacy and safety of drug candidates and medical devices. Disease models that are based on patients' profiling data are being produced to represent interactomes of genes or proteins and to infer causality in the pathophysiology, which makes it possible to mimic the impact of drugs on relevant targets. Virtual patients designed from medical records as well as digital twins are generated to simulate specific organs and to predict treatment efficacy at the individual patient level. As the acceptance of digital evidence by regulators grows, predictive artificial intelligence (AI)-based models will support the design of confirmatory trials in humans and will accelerate the development of efficient drugs and medical devices.


Asunto(s)
Inteligencia Artificial , Simulación por Computador , Humanos
9.
Expert Rev Clin Immunol ; 19(3): 305-314, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36680799

RESUMEN

INTRODUCTION: Auto-immune diseases are complex and heterogeneous. Various types of biomarkers can be used to support precision medicine approaches to autoimmune diseases, ensuring that the right patient receives the most appropriate therapy to improve treatment outcomes. AREAS COVERED: We review the recent progress made in modeling several autoimmune diseases such as Systemic Lupus Erythematosus, primary Sjogren Syndrome, and Rheumatoid Arthritis following extensive molecular profiling of large cohorts of patients. From this knowledge, BMKs are being identified which support diagnostic as well as patient stratification and prediction of response to treatment. The identification of biomarkers should be initiated early in drug development and properly validated during subsequent clinical trials. To ensure the robustness and reproducibility of biomarkers, the PERMIT Consortium recently established recommendations highlighting the importance of relevant study design, sample size, and appropriate validation of analytical methods. EXPERT OPINION: The integration by AI-powered analytics of massive data provided by multi-omics technologies, high-resolution medical imaging and sensors borne by patients will eventually allow the identification of clinically relevant BMKs, likely in the form of combinatorial predictive algorithms, to support future drug development for autoimmune diseases.


Asunto(s)
Artritis Reumatoide , Enfermedades Autoinmunes , Lupus Eritematoso Sistémico , Humanos , Reproducibilidad de los Resultados , Enfermedades Autoinmunes/terapia , Enfermedades Autoinmunes/tratamiento farmacológico , Lupus Eritematoso Sistémico/tratamiento farmacológico , Artritis Reumatoide/diagnóstico , Artritis Reumatoide/tratamiento farmacológico , Biomarcadores
10.
Expert Opin Drug Discov ; 17(8): 815-824, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35786124

RESUMEN

INTRODUCTION: As a mid-size international pharmaceutical company, we initiated 4 years ago the launch of a dedicated high-throughput computing platform supporting drug discovery. The platform named 'Patrimony' was built up on the initial predicate to capitalize on our proprietary data while leveraging public data sources in order to foster a Computational Precision Medicine approach with the power of artificial intelligence. AREAS COVERED: Specifically, Patrimony is designed to identify novel therapeutic target candidates. With several successful use cases in immuno-inflammatory diseases, and current ongoing extension to applications to oncology and neurology, we document how this industrial computational platform has had a transformational impact on our R&D, making it more competitive, as well time and cost effective through a model-based educated selection of therapeutic targets and drug candidates. EXPERT OPINION: We report our achievements, but also our challenges in implementing data access and governance processes, building up hardware and user interfaces, and acculturing scientists to use predictive models to inform decisions.


Asunto(s)
Inteligencia Artificial , Descubrimiento de Drogas , Humanos , Medicina de Precisión
13.
Expert Rev Clin Immunol ; 18(1): 47-56, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34842494

RESUMEN

INTRODUCTION: The complex pathophysiology of autoimmune diseases (AIDs) is being progressively deciphered, providing evidence for a multiplicity of pro-inflammatory pathways underlying heterogeneous clinical phenotypes and disease evolution. AREAS COVERED: Treatment strategies involving drug combinations are emerging as a preferred option to achieve remission in a vast majority of patients affected by systemic AIDs. The design of appropriate drug combinations can benefit from AID modeling following a comprehensive multi-omics molecular profiling of patients combined with Artificial Intelligence (AI)-powered computational analyses. Such disease models support patient stratification in homogeneous subgroups, shed light on dysregulated pro-inflammatory pathways and yield hypotheses regarding potential therapeutic targets and candidate biomarkers to stratify and monitor patients during treatment. AID models inform the rational design of combination therapies interfering with independent pro-inflammatory pathways related to either one of five prominent immune compartments contributing to the pathophysiology of AIDs, i.e. pro-inflammatory signals originating from tissues, innate immune mechanisms, T lymphocyte activation, autoantibodies and B cell activation, as well as soluble mediators involved in immune cross-talk. EXPERT OPINION: The optimal management of AIDs in the future will rely upon rationally designed combination therapies, as a modality of a model-based Computational Precision Medicine taking into account the patients' biological and clinical specificities.


Asunto(s)
Enfermedades Autoinmunes , Medicina de Precisión , Inteligencia Artificial , Enfermedades Autoinmunes/tratamiento farmacológico , Biomarcadores , Terapia Combinada , Humanos
14.
Expert Rev Proteomics ; 19(1): 33-42, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34937491

RESUMEN

INTRODUCTION: Proteomics encompasses a wide and expanding range of methods to identify, characterize, and quantify thousands of proteins from a variety of biological samples, including blood samples, tumors, and tissues. Such methods are supportive of various forms of immunotherapy applied to chronic conditions such as allergies, autoimmune diseases, cancers, and infectious diseases. AREAS COVERED: In support of immunotherapy, proteomics based on mass spectrometry has multiple specific applications related to (i) disease modeling and patient stratification, (ii) antigen/ autoantigen/neoantigen/ allergen identification, (iii) characterization of proteins and monoclonal antibodies used for immunotherapeutic or diagnostic purposes, (iv) identification of biomarkers and companion diagnostics and (v) monitoring by immunoproteomics of immune responses elicited in the course of the disease or following immunotherapy. EXPERT OPINION: Proteomics contributes as an enabling technology to an evolution of immunotherapy toward a precision medicine approach aiming to better tailor treatments to patients' specificities in multiple disease areas. This trend is favored by a better understanding through multi-omics profiling of both the patient's characteristics, his/her immune status as well as of the features of the immunotherapeutic drug.


Asunto(s)
Medicina de Precisión , Proteómica , Biomarcadores , Femenino , Humanos , Inmunoterapia/métodos , Masculino , Espectrometría de Masas , Proteómica/métodos
15.
Drug Discov Today ; 27(1): 215-222, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34555509

RESUMEN

Artificial Intelligence (AI) relies upon a convergence of technologies with further synergies with life science technologies to capture the value of massive multi-modal data in the form of predictive models supporting decision-making. AI and machine learning (ML) enhance drug design and development by improving our understanding of disease heterogeneity, identifying dysregulated molecular pathways and therapeutic targets, designing and optimizing drug candidates, as well as evaluating in silico clinical efficacy. By providing an unprecedented level of knowledge on both patient specificities and drug candidate properties, AI is fostering the emergence of a computational precision medicine allowing the design of therapies or preventive measures tailored to the singularities of individual patients in terms of their physiology, disease features, and exposure to environmental risks.


Asunto(s)
Inteligencia Artificial , Diseño de Fármacos/tendencias , Desarrollo de Medicamentos/tendencias , Evaluación de Medicamentos , Medicina de Precisión , Tecnología Biomédica/métodos , Tecnología Biomédica/tendencias , Técnicas de Apoyo para la Decisión , Evaluación de Medicamentos/métodos , Evaluación de Medicamentos/tendencias , Humanos , Informática Médica , Medicina de Precisión/métodos , Medicina de Precisión/tendencias
16.
Arthritis Res Ther ; 23(1): 262, 2021 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-34663440

RESUMEN

BACKGROUND: The identification of patients with knee osteoarthritis (OA) likely to progress rapidly in terms of structure is critical to facilitate the development of disease-modifying drugs. METHODS: Using 9280 knee magnetic resonance (MR) images (3268 patients) from the Osteoarthritis Initiative (OAI) database , we implemented a deep learning method to predict, from MR images and clinical variables including body mass index (BMI), further cartilage degradation measured by joint space narrowing at 12 months. RESULTS: Using COR IW TSE images, our classification model achieved a ROC AUC score of 65%. On a similar task, trained radiologists obtained a ROC AUC score of 58.7% highlighting the difficulty of the classification task. Additional analyses conducted in parallel to predict pain grade evaluated by the WOMAC pain index achieved a ROC AUC score of 72%. Attention maps provided evidence for distinct specific areas as being relevant in those two predictive models, including the medial joint space for JSN progression and the intra-articular space for pain prediction. CONCLUSIONS: This feasibility study demonstrates the interest of deep learning applied to OA, with a potential to support even trained radiologists in the challenging task of identifying patients with a high-risk of disease progression.


Asunto(s)
Cartílago Articular , Aprendizaje Profundo , Osteoartritis de la Rodilla , Progresión de la Enfermedad , Humanos , Articulación de la Rodilla , Imagen por Resonancia Magnética , Osteoartritis de la Rodilla/diagnóstico por imagen
17.
Drug Discov Today ; 26(10): 2465-2473, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34224903

RESUMEN

Interferon (IFN)-α has emerged as a major therapeutic target for several autoimmune rheumatic diseases. In this review, we focus on clinical and preclinical advances in anti-IFN-α treatments in systemic lupus erythematosus (SLE), primary Sjögren syndrome (pSS), systemic sclerosis (SSc), and dermatomyositis (DM), for which a high medical need persists. Promising achievements were obtained following direct IFN-α neutralization, targeting its production through the cytosolic nucleic acid sensor pathways or by blocking its downstream effects through the type I IFN receptor. We further focus on molecular profiling and data integration approaches as crucial steps to select patients most likely to benefit from anti-IFN-α therapies within a precision medicine approach.


Asunto(s)
Enfermedades Autoinmunes/terapia , Interferón-alfa/antagonistas & inhibidores , Enfermedades Reumáticas/terapia , Animales , Enfermedades Autoinmunes/inmunología , Humanos , Interferón-alfa/inmunología , Terapia Molecular Dirigida , Selección de Paciente , Medicina de Precisión/métodos , Receptor de Interferón alfa y beta/inmunología , Enfermedades Reumáticas/inmunología
18.
PLoS One ; 16(7): e0254374, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34293006

RESUMEN

While establishing worldwide collective immunity with anti SARS-CoV-2 vaccines, COVID-19 remains a major health issue with dramatic ensuing economic consequences. In the transition, repurposing existing drugs remains the fastest cost-effective approach to alleviate the burden on health services, most particularly by reducing the incidence of the acute respiratory distress syndrome associated with severe COVID-19. We undertook a computational repurposing approach to identify candidate therapeutic drugs to control progression towards severe airways inflammation during COVID-19. Molecular profiling data were obtained from public sources regarding SARS-CoV-2 infected epithelial or endothelial cells, immune dysregulations associated with severe COVID-19 and lung inflammation induced by other respiratory viruses. From these data, we generated a protein-protein interactome modeling the evolution of lung inflammation during COVID-19 from inception to an established cytokine release syndrome. This predictive model assembling severe COVID-19-related proteins supports a role for known contributors to the cytokine storm such as IL1ß, IL6, TNFα, JAK2, but also less prominent actors such as IL17, IL23 and C5a. Importantly our analysis points out to alarmins such as TSLP, IL33, members of the S100 family and their receptors (ST2, RAGE) as targets of major therapeutic interest. By evaluating the network-based distances between severe COVID-19-related proteins and known drug targets, network computing identified drugs which could be repurposed to prevent or slow down progression towards severe airways inflammation. This analysis confirmed the interest of dexamethasone, JAK2 inhibitors, estrogens and further identified various drugs either available or in development interacting with the aforementioned targets. We most particularly recommend considering various inhibitors of alarmins or their receptors, currently receiving little attention in this indication, as candidate treatments for severe COVID-19.


Asunto(s)
Alarminas/inmunología , Antivirales/farmacología , COVID-19/complicaciones , Reposicionamiento de Medicamentos , Neumonía/complicaciones , Neumonía/tratamiento farmacológico , Antivirales/inmunología , Antivirales/uso terapéutico , Humanos , Neumonía/inmunología
19.
Nat Commun ; 12(1): 3523, 2021 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-34112769

RESUMEN

There is currently no approved treatment for primary Sjögren's syndrome, a disease that primarily affects adult women. The difficulty in developing effective therapies is -in part- because of the heterogeneity in the clinical manifestation and pathophysiology of the disease. Finding common molecular signatures among patient subgroups could improve our understanding of disease etiology, and facilitate the development of targeted therapeutics. Here, we report, in a cross-sectional cohort, a molecular classification scheme for Sjögren's syndrome patients based on the multi-omic profiling of whole blood samples from a European cohort of over 300 patients, and a similar number of age and gender-matched healthy volunteers. Using transcriptomic, genomic, epigenetic, cytokine expression and flow cytometry data, combined with clinical parameters, we identify four groups of patients with distinct patterns of immune dysregulation. The biomarkers we identify can be used by machine learning classifiers to sort future patients into subgroups, allowing the re-evaluation of response to treatments in clinical trials.


Asunto(s)
Citocinas/sangre , Metilación de ADN , Interferones/sangre , Proteoma/metabolismo , Síndrome de Sjögren/inmunología , Transcriptoma/genética , Adulto , Autoanticuerpos/sangre , Biomarcadores/sangre , Quimiocinas/análisis , Quimiocinas/genética , Quimiocinas/metabolismo , Estudios de Cohortes , Biología Computacional , Simulación por Computador , Estudios Transversales , Citocinas/análisis , Citocinas/genética , Metilación de ADN/genética , Bases de Datos Genéticas , Bases de Datos de Proteínas , Femenino , Citometría de Flujo , Estudio de Asociación del Genoma Completo , Humanos , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Interferones/genética , Masculino , Persona de Mediana Edad , Familia de Multigenes , Polimorfismo de Nucleótido Simple , Proteoma/genética , RNA-Seq , Síndrome de Sjögren/sangre , Síndrome de Sjögren/genética , Síndrome de Sjögren/fisiopatología
20.
J Transl Autoimmun ; 4: 100093, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33748735

RESUMEN

Increased interferon-α (IFN-α) production is a critical component in the pathophysiology of systemic lupus erythematosus (SLE) and other rheumatic autoimmune diseases. Herein, we report the characterization of S95021, a fully human IgG1 anti-IFN-α monoclonal antibody (mAb) as a novel therapeutic candidate for targeted patient populations. S95021 was expressed in CHOZN GS-/- cells, purified by chromatography and characterized by using electrophoresis, size exclusion chromatography and liquid chromatography-mass spectrometry. High purity S95021 was obtained as a monomeric entity comprising different charge variants mainly due to N-glycosylation. Surface plasmon resonance kinetics experiments showed strong association rates with all IFN-α subtypes and estimated KDs below picomolar values. Pan-IFN-α-binding properties were confirmed by immunoprecipitation assays and neutralization capacity with reporter HEK-Blue IFN-α/ß cells. S95021 was IFN-α-selective and exhibited superior potency and broader neutralization profile when compared with the benchmark anti-IFN-α mAbs rontalizumab and sifalimumab. STAT-1 phosphorylation and the type I IFN gene signature induced in human peripheral blood mononuclear cells by recombinant IFN-α subtypes or plasmas from selected autoimmune patients were efficiently reduced by S95021 in a dose-dependent manner. Together, our results show that S95021 is a new potent, selective and pan IFN-α-neutralizing mAb. It is currently further evaluated as a valid therapeutic candidate in selected autoimmune diseases in which the IFN-α pro-inflammatory pathway is dysregulated.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA