Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 3 de 3
Filtrer
1.
Transplantation ; 107(7): 1502-1512, 2023 07 01.
Article de Anglais | MEDLINE | ID: mdl-36584373

RÉSUMÉ

BACKGROUND: During solid organ transplantation, donor leukocytes, including myeloid cells, are transferred within the organ to the recipient. Both tolerogenic and alloreactive roles have been attributed to donor myeloid cells; however, their subset-specific retention posttransplantation has not been investigated in detail. METHODS: Major histocompatibility complex (MHC)-matched and mismatched liver transplants were performed in mice, and the fate of donor and recipient myeloid cells was assessed. RESULTS: Following MHC-matched transplantation, a proportion of donor myeloid cells was retained in the graft, whereas others egressed and persisted in the blood, spleen, and bone marrow but not the lymph nodes. In contrast, after MHC-mismatched transplantation, all donor myeloid cells, except Kupffer cells, were depleted. This depletion was caused by recipient T and B cells because all donor myeloid subsets were retained in MHC-mismatched grafts when recipients lacked T and B cells. Recipient myeloid cells rapidly infiltrated MHC-matched and, to a greater extent, MHC-mismatched liver grafts. MHC-mismatched grafts underwent a transient rejection episode on day 7, coinciding with a transition in macrophages to a regulatory phenotype, after which rejection resolved. CONCLUSIONS: Phenotypic and kinetic differences in the myeloid cell responses between MHC-matched and mismatched grafts were identified. A detailed understanding of the dynamics of immune responses to transplantation is critical to improving graft outcomes.


Sujet(s)
Transplantation hépatique , Souris , Animaux , Transplantation hépatique/effets indésirables , Transplantation de moelle osseuse , Transplantation homologue , Complexe majeur d'histocompatibilité , Antigènes d'histocompatibilité , Cellules myéloïdes
2.
Sci Rep ; 11(1): 15312, 2021 07 28.
Article de Anglais | MEDLINE | ID: mdl-34321489

RÉSUMÉ

Advances in cancer immunology have increased the use of immune checkpoint inhibitors in clinical practice, however not all patients respond, and treatment can have severe side-effects. Blood-based immunological biomarkers are an attractive method for predicting which patients will respond to therapy, however, reliable biomarkers for immune checkpoint blockade are lacking. This study aimed to identify patients before or early in treatment who would best respond to PD-1 inhibitors. We hypothesised that higher baseline PD-L1 and/or PD-1 on peripheral blood T cells could predict radiological response to PD-1 inhibitors. This pilot prospective cohort study assessed 26 patients with melanoma or non-small cell lung cancer, treated with pembrolizumab, nivolumab, or nivolumab/ipilimumab combined. Response was assessed by RECIST 1.1. Peripheral blood lymphocytes collected at baseline, after one cycle, 10 weeks and at discontinuation of therapy were analysed by flow cytometry. Patients with a higher proportion of PD-L1+ T cells at baseline had improved objective response to PD-1 inhibitor therapy, and patients with a lower proportion of regulatory T cells at baseline experienced more immune-related adverse events. These findings may prove useful to assist in clinical decision making. Further studies with larger cohorts are required to validate these findings.


Sujet(s)
Antigène CD274/biosynthèse , Carcinome pulmonaire non à petites cellules/métabolisme , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Tumeurs du poumon/métabolisme , Mélanome/métabolisme , Thérapie moléculaire ciblée , Protéines tumorales/biosynthèse , Lymphocytes T/effets des médicaments et des substances chimiques , Sujet âgé , Sujet âgé de 80 ans ou plus , Anticorps monoclonaux humanisés/effets indésirables , Anticorps monoclonaux humanisés/pharmacologie , Anticorps monoclonaux humanisés/usage thérapeutique , Antigène CD274/antagonistes et inhibiteurs , Antigène CD274/génétique , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Femelle , Humains , Inhibiteurs de points de contrôle immunitaires/effets indésirables , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Ipilimumab/administration et posologie , Ipilimumab/effets indésirables , Ipilimumab/pharmacologie , Estimation de Kaplan-Meier , Tumeurs du poumon/traitement médicamenteux , Mâle , Mélanome/traitement médicamenteux , Protéines tumorales/antagonistes et inhibiteurs , Protéines tumorales/génétique , Nivolumab/administration et posologie , Nivolumab/effets indésirables , Nivolumab/pharmacologie , Nivolumab/usage thérapeutique , Projets pilotes , Survie sans progression , Études prospectives , Lymphocytes T régulateurs/effets des médicaments et des substances chimiques
3.
Sci Transl Med ; 11(501)2019 07 17.
Article de Anglais | MEDLINE | ID: mdl-31316010

RÉSUMÉ

Cancer immunotherapy using antibodies that target immune checkpoints has delivered outstanding results. However, responses only occur in a subset of patients, and it is not fully understood what biological processes determine an effective outcome. This lack of understanding hinders the development of rational combination treatments. We set out to define the pretreatment microenvironment associated with an effective outcome by using the fact that inbred mouse strains bearing monoclonal cancer cell line-derived tumors respond in a dichotomous manner to immune checkpoint blockade (ICB). We compared the cellular composition and gene expression profiles of responsive and nonresponsive tumors from mice before ICB and validated the findings in cohorts of patients with cancer treated with ICB antibodies. We found that responsive tumors were characterized by an inflammatory gene expression signature consistent with up-regulation of signal transducer and activator of transcription 1 (STAT1) and Toll-like receptor 3 (TLR3) signaling and down-regulation of interleukin-10 (IL-10) signaling. In addition, responsive tumors had more infiltrating-activated natural killer (NK) cells, which were necessary for response. Pretreatment of mice with large established tumors using the STAT1-activating cytokine interferon-γ (IFNγ), the TLR3 ligand poly(I:C), and an anti-IL-10 antibody sensitized tumors to ICB by attracting IFNγ-producing NK cells into the tumor, resulting in increased cure rates. Our results identify a pretreatment tumor microenvironment that predicts response to ICB, which can be therapeutically attained. These data suggest a biomarker-driven approach to patient management to establish whether a patient would benefit from treatment with sensitizing therapeutics before ICB.


Sujet(s)
Immunothérapie , Cellules tueuses naturelles/immunologie , Facteur de transcription STAT-1/métabolisme , Transduction du signal , Microenvironnement tumoral/immunologie , Animaux , Lignée cellulaire tumorale , Prolifération cellulaire , Clones cellulaires , Association thérapeutique , Modèles animaux de maladie humaine , Régulation de l'expression des gènes tumoraux , Humains , Inflammation/anatomopathologie , Souris de lignée BALB C , Souris de lignée C57BL , Tumeurs/génétique , Tumeurs/immunologie , Tumeurs/anatomopathologie , Phénotype
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE