Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 9 de 9
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Science ; 380(6649): eabo2296, 2023 06 09.
Article de Anglais | MEDLINE | ID: mdl-37289890

RÉSUMÉ

Antibiotics (ABX) compromise the efficacy of programmed cell death protein 1 (PD-1) blockade in cancer patients, but the mechanisms underlying their immunosuppressive effects remain unknown. By inducing the down-regulation of mucosal addressin cell adhesion molecule 1 (MAdCAM-1) in the ileum, post-ABX gut recolonization by Enterocloster species drove the emigration of enterotropic α4ß7+CD4+ regulatory T 17 cells into the tumor. These deleterious ABX effects were mimicked by oral gavage of Enterocloster species, by genetic deficiency, or by antibody-mediated neutralization of MAdCAM-1 and its receptor, α4ß7 integrin. By contrast, fecal microbiota transplantation or interleukin-17A neutralization prevented ABX-induced immunosuppression. In independent lung, kidney, and bladder cancer patient cohorts, low serum levels of soluble MAdCAM-1 had a negative prognostic impact. Thus, the MAdCAM-1-α4ß7 axis constitutes an actionable gut immune checkpoint in cancer immunosurveillance.


Sujet(s)
Antibactériens , Molécules d'adhérence cellulaire , Résistance aux médicaments antinéoplasiques , Microbiome gastro-intestinal , Inhibiteurs de points de contrôle immunitaires , Tolérance immunitaire , Surveillance immunologique , Intégrines , Mucoprotéines , Tumeurs , Animaux , Humains , Souris , Antibactériens/effets indésirables , Bactéries/immunologie , Molécules d'adhérence cellulaire/métabolisme , Mouvement cellulaire , Transplantation de microbiote fécal , Microbiome gastro-intestinal/immunologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Tolérance immunitaire/effets des médicaments et des substances chimiques , Intégrines/métabolisme , Interleukine-17/métabolisme , Mucoprotéines/métabolisme , Tumeurs/immunologie , Tumeurs/thérapie , Cellules Th17/immunologie , Tube digestif/immunologie , Tube digestif/microbiologie
2.
J Immunother Cancer ; 10(3)2022 03.
Article de Anglais | MEDLINE | ID: mdl-35296557

RÉSUMÉ

BACKGROUND: Prostate cancer (PC) responds to androgen deprivation therapy (ADT) usually in a transient fashion, progressing from hormone-sensitive PC (HSPC) to castration-resistant PC (CRPC). We investigated a mouse model of PC as well as specimens from PC patients to unravel an unsuspected contribution of thymus-derived T lymphocytes and the intestinal microbiota in the efficacy of ADT. METHODS: Preclinical experiments were performed in PC-bearing mice, immunocompetent or immunodeficient. In parallel, we prospectively included 65 HSPC and CRPC patients (Oncobiotic trial) to analyze their feces and blood specimens. RESULTS: In PC-bearing mice, ADT increased thymic cellularity and output. PC implanted in T lymphocyte-depleted or athymic mice responded less efficiently to ADT than in immunocompetent mice. Moreover, depletion of the intestinal microbiota by oral antibiotics reduced the efficacy of ADT. PC reduced the relative abundance of Akkermansia muciniphila in the gut, and this effect was reversed by ADT. Moreover, cohousing of PC-bearing mice with tumor-free mice or oral gavage with Akkermansia improved the efficacy of ADT. This appears to be applicable to PC patients because long-term ADT resulted in an increase of thymic output, as demonstrated by an increase in circulating recent thymic emigrant cells (sjTRECs). Moreover, as compared with HSPC controls, CRPC patients demonstrated a shift in their intestinal microbiota that significantly correlated with sjTRECs. While feces from healthy volunteers restored ADT efficacy, feces from PC patients failed to do so. CONCLUSIONS: These findings suggest the potential clinical utility of reversing intestinal dysbiosis and repairing acquired immune defects in PC patients.


Sujet(s)
Microbiome gastro-intestinal , Tumeurs prostatiques résistantes à la castration , Antagonistes des androgènes/pharmacologie , Antagonistes des androgènes/usage thérapeutique , Androgènes/usage thérapeutique , Animaux , Humains , Système immunitaire , Mâle , Souris , Tumeurs prostatiques résistantes à la castration/traitement médicamenteux
3.
Cancer Discov ; 12(4): 958-983, 2022 04 01.
Article de Anglais | MEDLINE | ID: mdl-35179201

RÉSUMÉ

Vaccination against coronavirus disease 2019 (COVID-19) relies on the in-depth understanding of protective immune responses to severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). We characterized the polarity and specificity of memory T cells directed against SARS-CoV-2 viral lysates and peptides to determine correlates with spontaneous, virus-elicited, or vaccine-induced protection against COVID-19 in disease-free and cancer-bearing individuals. A disbalance between type 1 and 2 cytokine release was associated with high susceptibility to COVID-19. Individuals susceptible to infection exhibited a specific deficit in the T helper 1/T cytotoxic 1 (Th1/Tc1) peptide repertoire affecting the receptor binding domain of the spike protein (S1-RBD), a hotspot of viral mutations. Current vaccines triggered Th1/Tc1 responses in only a fraction of all subject categories, more effectively against the original sequence of S1-RBD than that from viral variants. We speculate that the next generation of vaccines should elicit Th1/Tc1 T-cell responses against the S1-RBD domain of emerging viral variants. SIGNIFICANCE: This study prospectively analyzed virus-specific T-cell correlates of protection against COVID-19 in healthy and cancer-bearing individuals. A disbalance between Th1/Th2 recall responses conferred susceptibility to COVID-19 in both populations, coinciding with selective defects in Th1 recognition of the receptor binding domain of spike. See related commentary by McGary and Vardhana, p. 892. This article is highlighted in the In This Issue feature, p. 873.


Sujet(s)
Facteurs de restriction antiviraux , COVID-19 , Tumeurs , Lymphocytes T , Anticorps neutralisants , Facteurs de restriction antiviraux/immunologie , COVID-19/immunologie , Humains , Tumeurs/complications , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus/composition chimique , Glycoprotéine de spicule des coronavirus/génétique , Lymphocytes T/immunologie
4.
Cancer Discov ; 12(4): 1128-1151, 2022 04 01.
Article de Anglais | MEDLINE | ID: mdl-34930787

RÉSUMÉ

Gut dysbiosis has been associated with intestinal and extraintestinal malignancies, but whether and how carcinogenesis drives compositional shifts of the microbiome to its own benefit remains an open conundrum. Here, we show that malignant processes can cause ileal mucosa atrophy, with villous microvascular constriction associated with dominance of sympathetic over cholinergic signaling. The rapid onset of tumorigenesis induced a burst of REG3γ release by ileal cells, and transient epithelial barrier permeability that culminated in overt and long-lasting dysbiosis dominated by Gram-positive Clostridium species. Pharmacologic blockade of ß-adrenergic receptors or genetic deficiency in Adrb2 gene, vancomycin, or cohousing of tumor bearers with tumor-free littermates prevented cancer-induced ileopathy, eventually slowing tumor growth kinetics. Patients with cancer harbor distinct hallmarks of this stress ileopathy dominated by Clostridium species. Hence, stress ileopathy is a corollary disease of extraintestinal malignancies requiring specific therapies. SIGNIFICANCE: Whether gut dysbiosis promotes tumorigenesis and how it controls tumor progression remain open questions. We show that 50% of transplantable extraintestinal malignancies triggered a ß-adrenergic receptor-dependent ileal mucosa atrophy, associated with increased gut permeability, sustained Clostridium spp.-related dysbiosis, and cancer growth. Vancomycin or propranolol prevented cancer-associated stress ileopathy. This article is highlighted in the In This Issue feature, p. 873.


Sujet(s)
Dysbiose , Récepteurs bêta-adrénergiques , Carcinogenèse/anatomopathologie , Dysbiose/induit chimiquement , Dysbiose/complications , Dysbiose/anatomopathologie , Humains , Muqueuse intestinale/anatomopathologie , Transduction du signal
5.
Cell Death Differ ; 28(12): 3297-3315, 2021 12.
Article de Anglais | MEDLINE | ID: mdl-34230615

RÉSUMÉ

Patients with cancer are at higher risk of severe coronavirus infectious disease 2019 (COVID-19), but the mechanisms underlying virus-host interactions during cancer therapies remain elusive. When comparing nasopharyngeal swabs from cancer and noncancer patients for RT-qPCR cycle thresholds measuring acute respiratory syndrome coronavirus-2 (SARS-CoV-2) in 1063 patients (58% with cancer), we found that malignant disease favors the magnitude and duration of viral RNA shedding concomitant with prolonged serum elevations of type 1 IFN that anticorrelated with anti-RBD IgG antibodies. Cancer patients with a prolonged SARS-CoV-2 RNA detection exhibited the typical immunopathology of severe COVID-19 at the early phase of infection including circulation of immature neutrophils, depletion of nonconventional monocytes, and a general lymphopenia that, however, was accompanied by a rise in plasmablasts, activated follicular T-helper cells, and non-naive Granzyme B+FasL+, EomeshighTCF-1high, PD-1+CD8+ Tc1 cells. Virus-induced lymphopenia worsened cancer-associated lymphocyte loss, and low lymphocyte counts correlated with chronic SARS-CoV-2 RNA shedding, COVID-19 severity, and a higher risk of cancer-related death in the first and second surge of the pandemic. Lymphocyte loss correlated with significant changes in metabolites from the polyamine and biliary salt pathways as well as increased blood DNA from Enterobacteriaceae and Micrococcaceae gut family members in long-term viral carriers. We surmise that cancer therapies may exacerbate the paradoxical association between lymphopenia and COVID-19-related immunopathology, and that the prevention of COVID-19-induced lymphocyte loss may reduce cancer-associated death.


Sujet(s)
COVID-19/complications , COVID-19/virologie , Lymphopénie/complications , Tumeurs/complications , ARN viral/analyse , SARS-CoV-2/génétique , Excrétion virale , Adolescent , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Études de cohortes , ADN bactérien/sang , Enterobacteriaceae/génétique , Femelle , Humains , Interféron de type I/sang , Lymphopénie/virologie , Mâle , Micrococcaceae/génétique , Adulte d'âge moyen , Partie nasale du pharynx/virologie , Tumeurs/diagnostic , Tumeurs/mortalité , Pandémies , Pronostic , Facteurs temps , Jeune adulte
6.
Cell Death Differ ; 28(7): 2276-2295, 2021 07.
Article de Anglais | MEDLINE | ID: mdl-33976389

RÉSUMÉ

A deviated repertoire of the gut microbiome predicts resistance to cancer immunotherapy. Enterococcus hirae compensated cancer-associated dysbiosis in various tumor models. However, the mechanisms by which E. hirae restored the efficacy of cyclophosphamide administered with concomitant antibiotics remain ill defined. Here, we analyzed the multifaceted modes of action of this anticancer probiotic. Firstly, E. hirae elicited emigration of thymocytes and triggered systemic and intratumoral IFNγ-producing and CD137-expressing effector memory T cell responses. Secondly, E. hirae activated the autophagy machinery in enterocytes and mediated ATG4B-dependent anticancer effects, likely as a consequence of its ability to increase local delivery of polyamines. Thirdly, E. hirae shifted the host microbiome toward a Bifidobacteria-enriched ecosystem. In contrast to the live bacterium, its pasteurized cells or membrane vesicles were devoid of anticancer properties. These pleiotropic functions allow the design of optimal immunotherapies combining E. hirae with CD137 agonistic antibodies, spermidine, or Bifidobacterium animalis. We surmise that immunological, metabolic, epithelial, and microbial modes of action of the live E. hirae cooperate to circumvent primary resistance to therapy.


Sujet(s)
Antibactériens/pharmacologie , Enterococcus hirae/immunologie , Tumeurs/traitement médicamenteux , Probiotiques/pharmacologie , Animaux , Femelle , Microbiome gastro-intestinal/immunologie , Immunothérapie/méthodes , Cellules T mémoire/immunologie , Souris , Souris de lignée C57BL , Tumeurs/immunologie
7.
Cell Death Differ ; 28(5): 1532-1547, 2021 05.
Article de Anglais | MEDLINE | ID: mdl-33262469

RÉSUMÉ

Ileal epithelial cell apoptosis and the local microbiota modulate the effects of oxaliplatin against proximal colon cancer by modulating tumor immunosurveillance. Here, we identified an ileal immune profile associated with the prognosis of colon cancer and responses to chemotherapy. The whole immune ileal transcriptome was upregulated in poor-prognosis patients with proximal colon cancer, while the colonic immunity of healthy and neoplastic areas was downregulated (except for the Th17 fingerprint) in such patients. Similar observations were made across experimental models of implanted and spontaneous murine colon cancer, showing a relationship between carcinogenesis and ileal inflammation. Conversely, oxaliplatin-based chemotherapy could restore a favorable, attenuated ileal immune fingerprint in responders. These results suggest that chemotherapy inversely shapes the immune profile of the ileum-tumor axis, influencing clinical outcome.


Sujet(s)
Tumeurs du côlon/physiopathologie , Maladies de l'iléon/complications , Iléum/anatomopathologie , Animaux , Humains , Souris , Pronostic
8.
Oncoimmunology ; 9(1): 1794423, 2020 07 20.
Article de Anglais | MEDLINE | ID: mdl-32934888

RÉSUMÉ

Accumulating evidence from preclinical studies and human trials demonstrated the crucial role of the gut microbiota in determining the effectiveness of anticancer therapeutics such as immunogenic chemotherapy or immune checkpoint blockade. In summary, it appears that a diverse intestinal microbiota supports therapeutic anticancer responses, while a dysbiotic microbiota composition that lacks immunostimulatory bacteria or contains overabundant immunosuppressive species causes treatment failure. In this review, we explore preclinical and translational studies highlighting how eubiotic and dysbiotic microbiota composition can affect progression-free survival in cancer patients.


Sujet(s)
Microbiome gastro-intestinal , Tumeurs , Dysbiose , Humains , Inhibiteurs de points de contrôle immunitaires , Tumeurs/traitement médicamenteux , Symbiose
9.
Nat Med ; 26(6): 919-931, 2020 06.
Article de Anglais | MEDLINE | ID: mdl-32451498

RÉSUMÉ

The prognosis of colon cancer (CC) is dictated by tumor-infiltrating lymphocytes, including follicular helper T (TFH) cells and the efficacy of chemotherapy-induced immune responses. It remains unclear whether gut microbes contribute to the elicitation of TFH cell-driven responses. Here, we show that the ileal microbiota dictates tolerogenic versus immunogenic cell death of ileal intestinal epithelial cells (IECs) and the accumulation of TFH cells in patients with CC and mice. Suppression of IEC apoptosis led to compromised chemotherapy-induced immunosurveillance against CC in mice. Protective immune responses against CC were associated with residence of Bacteroides fragilis and Erysipelotrichaceae in the ileum. In the presence of these commensals, apoptotic ileal IECs elicited PD-1+ TFH cells in an interleukin-1R1- and interleukin-12-dependent manner. The ileal microbiome governed the efficacy of chemotherapy and PD-1 blockade in CC independently of microsatellite instability. These findings demonstrate that immunogenic ileal apoptosis contributes to the prognosis of chemotherapy-treated CC.


Sujet(s)
Adénocarcinome/traitement médicamenteux , Antinéoplasiques/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Tumeurs du côlon/traitement médicamenteux , Microbiome gastro-intestinal/immunologie , Iléum/effets des médicaments et des substances chimiques , Lymphocytes TIL/effets des médicaments et des substances chimiques , Oxaliplatine/pharmacologie , Adénocarcinome/immunologie , Adénocarcinome/microbiologie , Adénocarcinome/anatomopathologie , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Animaux , Antinéoplasiques/usage thérapeutique , Apoptose/immunologie , Bacteroides fragilis , Lignée cellulaire tumorale , Tumeurs du côlon/immunologie , Tumeurs du côlon/microbiologie , Tumeurs du côlon/anatomopathologie , Cellules épithéliales/effets des médicaments et des substances chimiques , Cellules épithéliales/immunologie , Cellules épithéliales/anatomopathologie , Femelle , Firmicutes , Microbiome gastro-intestinal/physiologie , Humains , Iléum/immunologie , Iléum/microbiologie , Iléum/anatomopathologie , Mort cellulaire immunogène/effets des médicaments et des substances chimiques , Mort cellulaire immunogène/immunologie , Surveillance immunologique/effets des médicaments et des substances chimiques , Surveillance immunologique/immunologie , Interleukine-12/immunologie , Muqueuse intestinale , Lymphocytes TIL/immunologie , Mâle , Souris , Adulte d'âge moyen , Oxaliplatine/usage thérapeutique , Pronostic , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur à l'interleukine-1 de type I/immunologie , Lymphocytes T auxiliaires/effets des médicaments et des substances chimiques , Lymphocytes T auxiliaires/immunologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...