Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 549
Filtrer
1.
J Neurosurg ; 140(6): 1549-1557, 2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38157532

RÉSUMÉ

OBJECTIVE: Malignancies of the CNS are difficult to treat because the blood-brain barrier (BBB) prevents most therapeutics from reaching the intracranial lesions at sufficiently high concentrations. This also applies to chimeric antigen receptor (CAR) T cells, for which systemic delivery is inferior to direct intratumoral or intraventricular injection of the cells. The authors previously reported on a novel approach to safely and reversibly open the BBB of mice by applying intra-arterial (IA) injections of NEO100, a pharmaceutical-grade version of the natural monoterpene perillyl alcohol. The authors hypothesized that this method would enable enhanced brain entry and therapeutic activity of intravenously delivered CAR T cells, which the authors tested in a mouse model of CNS lymphoma. METHODS: Human Raji lymphoma cells were implanted into the brains of immune-deficient mice. After tumor uptake was confirmed with bioluminescent imaging, 0.3% NEO100 was injected intra-arterially, which was followed by intravenous (IV) delivery of CD19-targeted CAR T cells. After this single intervention, tumor growth was monitored with imaging, long-term survival of mice was recorded, and select mice were euthanized to analyze the distribution of CAR T cells in brain tissue. RESULTS: Intravenously injected CAR T cells could be readily detected in brain tumor areas after IA injection of NEO100 but not after IA injection of the vehicle (without NEO100). Although all untreated control animals died within 3 weeks, all mice that received IA NEO100 followed by IV CAR T cells survived and thrived for 200 days, when the experiment was terminated. Of the mice that received IV CAR T cells without prior IA NEO100, 3 died within 3 weeks and 2 survived long-term. CONCLUSIONS: BBB opening by IA NEO100 facilitates brain entry of intravenously delivered CD19 CAR T cells. The long-term survival of all mice with CNS lymphoma, along with the disappearance of the tumor as determined with imaging, suggests that this one-time therapeutic intervention was curative. BBB opening by IA NEO100 may offer a novel option to increase brain access by CAR T cells.


Sujet(s)
Immunothérapie adoptive , Injections artérielles , Récepteurs chimériques pour l'antigène , Animaux , Souris , Immunothérapie adoptive/méthodes , Modèles animaux de maladie humaine , Barrière hémato-encéphalique , Humains , Tumeurs du cerveau/thérapie , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/immunologie , Lignée cellulaire tumorale/transplantation , Lymphomes/thérapie , Lymphomes/immunologie , Tumeurs du système nerveux central/thérapie , Tumeurs du système nerveux central/immunologie , Lymphocytes T/immunologie , Lymphocytes T/transplantation , Souris SCID
2.
Nat Commun ; 12(1): 6831, 2021 11 24.
Article de Anglais | MEDLINE | ID: mdl-34819502

RÉSUMÉ

Exhausted CD8+ T cells are key targets of immune checkpoint blockade therapy and their ineffective reinvigoration limits the durable benefit in some cancer patients. Here, we demonstrate that histone demethylase LSD1 acts to enforce an epigenetic program in progenitor exhausted CD8+ T cells to antagonize the TCF1-mediated progenitor maintenance and to promote terminal differentiation. Consequently, genetic perturbation or small molecules targeting LSD1 increases the persistence of the progenitor exhausted CD8+ T cells, which provide a sustained source for the proliferative conversion to numerically larger terminally exhausted T cells with tumor-killing cytotoxicity, thereby leading to effective and durable responses to anti-PD1 therapy. Collectively, our findings provide important insights into epigenetic mechanisms that regulate T cell exhaustion and have important implications for durable immunotherapy.


Sujet(s)
Lymphocytes T CD8+/immunologie , Régulation de l'expression des gènes tumoraux/immunologie , Histone Demethylases/métabolisme , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Tumeurs/immunologie , Animaux , Lymphocytes T CD8+/métabolisme , Lignée cellulaire tumorale/transplantation , Prolifération cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/génétique , Déméthylation de l'ADN/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Résistance aux médicaments antinéoplasiques/génétique , Épigenèse génétique/immunologie , Femelle , Cellules HEK293 , Facteur nucléaire hépatocytaire HNF-1 alpha/métabolisme , Histone Demethylases/antagonistes et inhibiteurs , Histone Demethylases/génétique , Humains , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Lymphocytes TIL , Mâle , Souris , Souris transgéniques , Tumeurs/traitement médicamenteux , Tumeurs/anatomopathologie , Culture de cellules primaires , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/métabolisme , Protéines recombinantes
3.
Int Immunopharmacol ; 101(Pt A): 108288, 2021 Dec.
Article de Anglais | MEDLINE | ID: mdl-34710844

RÉSUMÉ

PURPOSE: T-cell immunoglobulin and ITIM domain (TIGIT) is an immune checkpoint that is overexpressed on both immune cells and some cancer cells. TIGIT can alter the anti-tumor responses inside the tumor microenvironment. Hypoxia-inducible factor 1-alpha (HIF-1α) plays a significant role in the TME and involves suppressing the anti-tumor responses. Under hypoxic conditions, HIF-1α can enhance the expression of different immune checkpoints. Accordingly, hypoxic TME and TIGIT overexpression cause cancer development. Thus, we decided to inhibit tumor cell expansion by inhibiting TIGIT and HIF-1α molecules and discovering the relationship between TIGIT and HIF-1α. METHODS: In this research, we utilized superparamagnetic iron oxide-based NPs (SPIONs) combined with chitosan lactate (CL) and folic acid (FA) nanoparticles (NPs) loaded with TIGIT-siRNA and HIF-1α- siRNA for suppressing TIGIT and HIF-1α in tumor cells and evaluated the consequences of this treatment strategy on tumor growth, apoptosis, and metastasis. RESULTS: The results showed that cancer cells treated with TIGIT and HIF-1α siRNA-loaded SPIONs-CL-FA NPs, strongly suppressed the TIGIT and HIF-1α expression, colony formation ability, angiogenesis, and the growth rate of cancer cells. CONCLUSIONS: Present data suggest the combination treatment of TIGIT and HIF-1α as a novel treatment strategy against colorectal and breast cancer, but more researches are required to realize the complete role of TIGIT and HIF-1α inside the TME.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Sous-unité alpha du facteur-1 induit par l'hypoxie/antagonistes et inhibiteurs , Système d'administration de médicaments à base de nanoparticules/composition chimique , Tumeurs/traitement médicamenteux , Récepteurs immunologiques/antagonistes et inhibiteurs , Animaux , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Lignée cellulaire tumorale/transplantation , Prolifération cellulaire/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Synergie des médicaments , Femelle , Humains , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Nanoparticules magnétiques d'oxyde de fer/composition chimique , Souris , Invasion tumorale/prévention et contrôle , Tumeurs/immunologie , Tumeurs/anatomopathologie , Récepteurs immunologiques/métabolisme , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Microenvironnement tumoral/immunologie
4.
Nat Commun ; 12(1): 5733, 2021 09 30.
Article de Anglais | MEDLINE | ID: mdl-34593794

RÉSUMÉ

In addition to increasing the expression of programmed death-ligand 1 (PD-L1), tumor cells can also secrete exosomal PD-L1 to suppress T cell activity. Emerging evidence has revealed that exosomal PD-L1 resists immune checkpoint blockade, and may contribute to resistance to therapy. In this scenario, suppressing the secretion of tumor-derived exosomes may aid therapy. Here, we develop an assembly of exosome inhibitor (GW4869) and ferroptosis inducer (Fe3+) via amphiphilic hyaluronic acid. Cooperation between the two active components in the constructed nanounit induces an anti-tumor immunoresponse to B16F10 melanoma cells and stimulates cytotoxic T lymphocytes and immunological memory. The nanounit enhances the response to PD-L1 checkpoint blockade and may represent a therapeutic strategy for enhancing the response to this therapy.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Vecteurs de médicaments/composition chimique , Exosomes/effets des médicaments et des substances chimiques , Ferroptose/effets des médicaments et des substances chimiques , Mélanome expérimental/traitement médicamenteux , Tumeurs cutanées/traitement médicamenteux , Dérivés de l'aniline/pharmacologie , Dérivés de l'aniline/usage thérapeutique , Animaux , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Antigène CD274/antagonistes et inhibiteurs , Antigène CD274/métabolisme , Composés benzylidéniques/pharmacologie , Composés benzylidéniques/usage thérapeutique , Lignée cellulaire tumorale/transplantation , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/immunologie , Exosomes/immunologie , Exosomes/métabolisme , Femelle , Ferroptose/immunologie , Humains , Acide hyaluronique/composition chimique , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Mémoire immunologique , Activation des lymphocytes/effets des médicaments et des substances chimiques , Mélanome expérimental/immunologie , Mélanome expérimental/anatomopathologie , Souris , Tumeurs cutanées/immunologie , Tumeurs cutanées/anatomopathologie , Lymphocytes T cytotoxiques/effets des médicaments et des substances chimiques , Lymphocytes T cytotoxiques/immunologie , Échappement de la tumeur à la surveillance immunitaire/effets des médicaments et des substances chimiques , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Microenvironnement tumoral/immunologie
5.
Int Immunopharmacol ; 101(Pt A): 108173, 2021 Dec.
Article de Anglais | MEDLINE | ID: mdl-34607233

RÉSUMÉ

In previous studies, we have obtained a notable anti-tumor efficacy of the recombinant MUC1-MBP vaccine in the process of mouse B16-MUC1 melanoma treatment. However, the tumor cannot be eliminated completely. We found that the tumor inhibition rate decreased from 81.67% (five immunizations) to 43.67% (eight immunizations) after more than five immunizations, indicating persistent vaccine stimulation may activate immunosuppressive factors. In the present study, we revealed that programmed cell death 1 (PD1), an inhibitory molecule suppressing T cell function, expressed on splenic and tumor-infiltrating T cells were up-regulated by the vaccine. Therefore, to optimize the anti-tumor efficacy of the vaccine, we employed combination immunotherapy with MUC1-MBP vaccine and αPD1 (anti-PD1 antibody). Results showed that combination immunotherapy induced a more remarkable anti-tumor efficacy, the tumor clearance being increased to 80% from 20% which obtain by MUC1-MBP vaccine immunizations. To investigate the possible underlying mechanism, IFN-γ secretion and cytotoxic T lymphocyte (CTL) cytotoxicity were measured by enzyme-linked immunosorbent assay (ELISA) and xCELLigence real-time cell analyzer (RTCA) respectively. T cell subsets and immunosuppressive cells in the mouse spleen and tumor microenvironment were analyzed by FACS. Results showed that the proportion of splenic CD8+T cells and tumor infiltration was increased and the activity of CTL killing, T helper 1 (Th1), Type 1 CD8+T (Tc1) was enhanced, indicating that the anti-tumor efficacy enhanced by combination immunotherapy was mainly through boosting CD8+T cells mediated anti-tumor cellular immunity. Additionally, combination immunotherapy significantly decreased the splenic and tumor-infiltrating myeloid derived suppressor cells (MDSCs). These results demonstrated that combination immunotherapy with MUC1-MBP vaccine and αPD1 was capable to invoke a more potent anti-tumor immune response and provide a foundation for further research.


Sujet(s)
Vaccins anticancéreux/administration et posologie , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Mélanome expérimental/thérapie , Tumeurs cutanées/thérapie , Animaux , Lymphocytes T CD8+/effets des médicaments et des substances chimiques , Lymphocytes T CD8+/immunologie , Vaccins anticancéreux/génétique , Vaccins anticancéreux/immunologie , Lignée cellulaire tumorale/transplantation , Femelle , Humains , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Immunothérapie/méthodes , Mélanome expérimental/immunologie , Mélanome expérimental/anatomopathologie , Souris , Mucine-1/administration et posologie , Mucine-1/génétique , Mucine-1/immunologie , Protéine basique de la myéline/administration et posologie , Protéine basique de la myéline/génétique , Protéine basique de la myéline/immunologie , Cellules myéloïdes suppressives/effets des médicaments et des substances chimiques , Cellules myéloïdes suppressives/immunologie , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/métabolisme , Protéines de fusion recombinantes/administration et posologie , Protéines de fusion recombinantes/génétique , Protéines de fusion recombinantes/immunologie , Tumeurs cutanées/immunologie , Tumeurs cutanées/anatomopathologie , Lymphocytes auxiliaires Th1/effets des médicaments et des substances chimiques , Lymphocytes auxiliaires Th1/immunologie , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Microenvironnement tumoral/immunologie , Vaccins synthétiques/administration et posologie , Vaccins synthétiques/génétique , Vaccins synthétiques/immunologie
6.
Int Immunopharmacol ; 101(Pt A): 108265, 2021 Dec.
Article de Anglais | MEDLINE | ID: mdl-34715491

RÉSUMÉ

Dendritic cells (DCs) are recognized as the most potent antigen-presenting cells, capable of priming both naïve and memory T cells. Thus, tumor-resident DCs (tumor-associated DCs: TADCs) play a crucial role in the immune response against tumors. However, TADCs are also well known as a "double-edged sword" because an immunosuppressive environment, such as a tumor microenvironment, maintains the immature and tolerogenic properties of TADCs, resulting in the deterioration of the tumor. Therefore, it is essential to maintain and enhance the anti-tumoral activity of TADCs to aid tumor elimination. This study demonstrated the potential for tumor growth inhibition of Aureobasidium pullulan-derived ß-glucan (AP-BG). Administration of AP-BG dramatically limited the development of different types of tumor cell lines transplanted into mice. Examination of the tumor-infiltrating leukocytes revealed that AP-BG caused high expression of co-stimulatory molecules on TADCs and enhanced the production of cytolytic granules as well as pro-inflammatory cytokines by the tumor-resident T cells. Furthermore, the syngeneic mixed lymphoid reaction assay and popliteal lymph node assay showed the significant ability of AP-BG to improve DCs' antigen-specific priming of T cells in vitro and in vivo. Taken together, ß-glucan might be an immune-potentiating adjuvant for cancer treatment. This highly widely-used reagent will initiate a new way to activate DC-targeted cancer immune therapy.


Sujet(s)
Adjuvants immunologiques/pharmacologie , Aureobasidium (genre)/composition chimique , Cellules dendritiques/effets des médicaments et des substances chimiques , Tumeurs/traitement médicamenteux , bêta-Glucanes/pharmacologie , Adjuvants immunologiques/isolement et purification , Adjuvants immunologiques/usage thérapeutique , Animaux , Lignée cellulaire tumorale/transplantation , Cellules dendritiques/immunologie , Modèles animaux de maladie humaine , Tests de criblage d'agents antitumoraux , Humains , Activation des lymphocytes/effets des médicaments et des substances chimiques , Lymphocytes TIL/effets des médicaments et des substances chimiques , Lymphocytes TIL/immunologie , Souris , Souris transgéniques , Tumeurs/immunologie , Tumeurs/anatomopathologie , Lymphocytes T/effets des médicaments et des substances chimiques , Lymphocytes T/immunologie , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Microenvironnement tumoral/immunologie , bêta-Glucanes/isolement et purification , bêta-Glucanes/usage thérapeutique
7.
Sci Rep ; 11(1): 18923, 2021 09 23.
Article de Anglais | MEDLINE | ID: mdl-34556678

RÉSUMÉ

Advances in imaging and early cancer detection have increased interest in magnetic resonance (MR) guided focused ultrasound (MRgFUS) technologies for cancer treatment. MRgFUS ablation treatments could reduce surgical risks, preserve organ tissue and function, and improve patient quality of life. However, surgical resection and histological analysis remain the gold standard to assess cancer treatment response. For non-invasive ablation therapies such as MRgFUS, the treatment response must be determined through MR imaging biomarkers. However, current MR biomarkers are inconclusive and have not been rigorously evaluated against histology via accurate registration. Existing registration methods rely on anatomical features to directly register in vivo MR and histology. For MRgFUS applications in anatomies such as liver, kidney, or breast, anatomical features that are not caused by the treatment are often insufficient to drive direct registration. We present a novel MR to histology registration workflow that utilizes intermediate imaging and does not rely on anatomical MR features being visible in histology. The presented workflow yields an overall registration accuracy of 1.00 ± 0.13 mm. The developed registration pipeline is used to evaluate a common MRgFUS treatment assessment biomarker against histology. Evaluating MR biomarkers against histology using this registration pipeline will facilitate validating novel MRgFUS biomarkers to improve treatment assessment without surgical intervention. While the presented registration technique has been evaluated in a MRgFUS ablation treatment model, this technique could be potentially applied in any tissue to evaluate a variety of therapeutic options.


Sujet(s)
Ablation par ultrasons focalisés de haute intensité/méthodes , Traitement d'image par ordinateur , Imagerie interventionnelle par résonance magnétique , Tumeurs/thérapie , Animaux , Lignée cellulaire tumorale/transplantation , Modèles animaux de maladie humaine , Études de faisabilité , Humains , Nécrose/diagnostic , Nécrose/anatomopathologie , Tumeurs/imagerie diagnostique , Tumeurs/anatomopathologie , Lapins , Résultat thérapeutique
8.
Int J Cancer ; 149(12): 2116-2124, 2021 12 15.
Article de Anglais | MEDLINE | ID: mdl-34460096

RÉSUMÉ

Acidity in the tumor microenvironment has been reported to promote cancer growth and metastasis. In our study, we examined a potential relation between extracellular acidity and expression level of the immune checkpoint molecule programmed cell death protein 1 (PD-L1) in murine squamous cell carcinoma (SCC) and melanoma cell lines. PD-L1 expression in the tumor cells was upregulated by culturing in a low pH culture medium. Tumor-bearing mice were allowed to ingest sodium bicarbonate, resulting in neutralization of acidity in the tumor tissue, a decrease in PD-L1 expression in tumor cells and suppression of tumor growth in vivo. Proton-sensing G protein-coupled receptors, T-cell death-associated gene 8 (TDAG8) and ovarian cancer G-protein-coupled receptor 1 (OGR1), were upregulated by low pH, and essentially involved in the acidity-induced elevation of PD-L1 expression in the tumor cells. Human head and neck SCC RNAseq data from the Cancer Genome Atlas also suggested a statistically significant correlation between expression levels of the proton sensors and PD-L1 mRNA expression. These findings strongly suggest that neutralization of acidity in tumor tissue may result in reduction of PD-L1 expression, potentially leading to inhibition of an immune checkpoint and augmentation of antitumor immunity.


Sujet(s)
Antigène CD274/génétique , Tumeurs/immunologie , Récepteurs couplés aux protéines G/métabolisme , Animaux , Antigène CD274/métabolisme , Lignée cellulaire tumorale/transplantation , Jeux de données comme sujet , Modèles animaux de maladie humaine , Femelle , Régulation de l'expression des gènes tumoraux/immunologie , Humains , Concentration en ions d'hydrogène , Souris , Tumeurs/génétique , Tumeurs/anatomopathologie , Protons , RNA-Seq , Échappement de la tumeur à la surveillance immunitaire/génétique , Microenvironnement tumoral/génétique , Microenvironnement tumoral/immunologie , Régulation positive
9.
Front Immunol ; 12: 705381, 2021.
Article de Anglais | MEDLINE | ID: mdl-34349765

RÉSUMÉ

The role of B cells in the tumor microenvironment (TME) has largely been under investigated, and data regarding the antibody repertoire encoded by B cells in the TME and the adjacent lymphoid organs are scarce. Here, we utilized B cell receptor high-throughput sequencing (BCR-Seq) to profile the antibody repertoire signature of tumor-infiltrating lymphocyte B cells (TIL-Bs) in comparison to B cells from three anatomic compartments in a mouse model of triple-negative breast cancer. We found that TIL-Bs exhibit distinct antibody repertoire measures, including high clonal polarization and elevated somatic hypermutation rates, suggesting a local antigen-driven B-cell response. Importantly, TIL-Bs were highly mutated but non-class switched, suggesting that class-switch recombination may be inhibited in the TME. Tracing the distribution of TIL-B clones across various compartments indicated that they migrate to and from the TME. The data thus suggests that antibody repertoire signatures can serve as indicators for identifying tumor-reactive B cells.


Sujet(s)
Diversité des anticorps , Sous-populations de lymphocytes B/immunologie , Chaines lourdes des immunoglobulines/génétique , Lymphocytes TIL/immunologie , Tumeurs expérimentales de la mamelle/immunologie , Récepteurs pour l'antigène des lymphocytes B/immunologie , Tumeurs du sein triple-négatives/immunologie , Microenvironnement tumoral/immunologie , Animaux , Cellules sanguines/immunologie , Moelle osseuse/anatomopathologie , Lignée cellulaire tumorale/transplantation , Mouvement cellulaire , Femelle , Séquençage nucléotidique à haut débit , Humains , Immunoglobuline G/génétique , Immunoglobuline G/immunologie , Immunoglobuline M/génétique , Immunoglobuline M/immunologie , Région variable d'immunoglobuline/génétique , Noeuds lymphatiques/anatomopathologie , Tumeurs expérimentales de la mamelle/anatomopathologie , Souris , Souris de lignée BALB C , Spécificité d'organe , Récepteurs pour l'antigène des lymphocytes B/génétique , Hypermutation somatique des gènes des immunoglobulines , Tumeurs du sein triple-négatives/anatomopathologie
10.
Immunol Lett ; 239: 32-41, 2021 11.
Article de Anglais | MEDLINE | ID: mdl-34418488

RÉSUMÉ

Tumor-derived exosomes (TEXs) could be harnessed as an immunotherapeutic cancer vaccine. These nanovesicles are inherently possesses rich tumor antigen reservoirs. Due to their undesirable features such as poor or limited immunogenicity as well as facilitation of cancer development via mediating communication between tumor cells TEXs could be transformed into an effective immune adjuvant delivery system that initiates a strong humoral and cell-mediated tumor-specific immune response. Engineering TEXs to harbor immunostimulatory molecules still remains a challenge. Previously, we demonstrated that nucleic acid ligand encapsulated liposomes could trigger synergistic strong humoral, and cell mediated immune responses and provokes tumor regression to that of their standalone counterparts. In this study, we evaluated to immunogenicity of 4T1/Her2 cell-derived exosomes upon loading them with two potent immuno adjuvant, a TLR9 ligand, K-type CpG ODN and a TLR3 ligand, p(I:C). Engineered TEXs co-encapsulating both ligands displayed boosted immunostimulatory properties by activating antigen-specific primary and memory T cell responses. Furthermore, our exosome-based vaccine candidate elicited robust Th1-biased immunity as evidenced by elevated secretion of IgG2a and IFNγ. In a therapeutic cancer model, administration of4T1 tumor derived exosomes loaded with CpG ODN and p(I:C) to animals regress tumor growth in 4T1 tumor-bearing mice. Taken together this work implicated that an exosome-based therapeutic vaccine promoted strong cellular and humoral anti-tumor immunity that is sufficient to reverse established tumors. This approach offers a personalized tumor therapy strategy that could be implemented in the clinic.


Sujet(s)
Adjuvants immunologiques/administration et posologie , Antigènes néoplasiques/administration et posologie , Tumeurs du sein/thérapie , Vaccins anticancéreux/administration et posologie , Exosomes/immunologie , Animaux , Antigènes néoplasiques/immunologie , Tumeurs du sein/immunologie , Tumeurs du sein/anatomopathologie , Vaccins anticancéreux/immunologie , Lignée cellulaire tumorale/transplantation , Modèles animaux de maladie humaine , Femelle , Humains , Cellules T mémoire/immunologie , Souris , Oligodésoxyribonucléotides/administration et posologie , Oligodésoxyribonucléotides/immunologie , Poly I-C/administration et posologie , Poly I-C/immunologie , Lymphocytes auxiliaires Th1/immunologie , Récepteur de type Toll-3/métabolisme , Récepteur-9 de type Toll-like/métabolisme
11.
Hepatology ; 74(6): 3174-3193, 2021 12.
Article de Anglais | MEDLINE | ID: mdl-34288020

RÉSUMÉ

BACKGROUND AND AIMS: Because of a paucity of effective treatment options, metastasis is still a major cause for HCC-associated mortality. The molecular mechanism of inflammation-induced HCC metastasis is open for study. Here, we characterized the function of solute carrier family 7 member 11 (SLC7A11) in inflammation-related HCC metastasis and probed therapy strategies for this subpopulation of patients. APPROACH AND RESULTS: Elevated expression of SLC7A11 was positively correlated with poor tumor differentiation, and higher tumor-nodule-metastasis stage, and indicated poor prognosis in human HCC. SLC7A11 increased HIF1α expression through reducing α-ketoglutarate (αKG) level by exporting glutamate. SLC7A11 up-regulated programmed death ligand 1 (PD-L1) and colony-stimulating factor 1 (CSF1) expression through αKG-HIF1α cascade. SLC7A11 overexpression in HCC cells promoted intratumoral tumor-associated macrophage (TAM) and myeloid-derived suppressor cell (MDSC) infiltration through the CSF1/colony-stimulating factor 1 receptor (CSF1R) axis, whereas knockdown of CSF1 attenuated SLC7A11-mediated intratumoral TAM and MDSC infiltration and HCC metastasis. Depletion of either TAMs or MDSCs decreased SLC7A11-mediated HCC metastasis. Furthermore, the combination of CSF1R inhibitor BZL945 and anti-PD-L1 antibody blocked SLC7A11-induced HCC metastasis. In addition, IL-1ß up-regulated SLC7A11 expression through the interleukin-1 receptor type 1 (IL-1R1)/extracellular signal-regulated kinase/specificity protein 1 pathway. SLC7A11 knockdown impaired IL-1ß-promoted HCC metastasis. Anakinra, an IL-1R1 antagonist, reversed IL-1ß-promoted HCC metastasis. In human HCC tissues, SLC7A11 expression was positively associated with HIF1α, PD-L1, and CSF1 expression and intratumoral TAM and MDSC infiltration. CONCLUSIONS: IL-1ß-induced SLC7A11 overexpression up-regulated PD-L1 and CSF1 through the αKG/HIF1α axis, which promoted TAM and MDSC infiltration. Interruption of this oncogenic loop may provide a promising therapy strategy for the inhibition of SLC7A11-mediated HCC metastasis.


Sujet(s)
Système y+ de transport d'acides aminés/génétique , Carcinome hépatocellulaire/immunologie , Interleukine-1 bêta/métabolisme , Tumeurs du foie/immunologie , Système y+ de transport d'acides aminés/métabolisme , Animaux , Antigène CD274/antagonistes et inhibiteurs , Antigène CD274/métabolisme , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/mortalité , Carcinome hépatocellulaire/thérapie , Lignée cellulaire tumorale/transplantation , Modèles animaux de maladie humaine , Études de suivi , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux/immunologie , Techniques de knock-down de gènes , Hépatectomie , Humains , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Antagoniste du récepteur à l'interleukine-1/pharmacologie , Antagoniste du récepteur à l'interleukine-1/usage thérapeutique , Acides cétoglutariques/métabolisme , Foie/immunologie , Foie/anatomopathologie , Foie/chirurgie , Tumeurs du foie/génétique , Tumeurs du foie/mortalité , Tumeurs du foie/thérapie , Facteur de stimulation des colonies de macrophages/métabolisme , Cellules myéloïdes suppressives/immunologie , Pronostic , Récepteur de facteur de croissance granulocyte-macrophage/antagonistes et inhibiteurs , Récepteur de facteur de croissance granulocyte-macrophage/métabolisme , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Microenvironnement tumoral/génétique , Microenvironnement tumoral/immunologie , Macrophages associés aux tumeurs/immunologie , Régulation positive/effets des médicaments et des substances chimiques , Régulation positive/immunologie
12.
Nat Commun ; 12(1): 4405, 2021 07 20.
Article de Anglais | MEDLINE | ID: mdl-34285232

RÉSUMÉ

Stimulator of interferon genes (STING) promotes anti-tumour immunity by linking innate and adaptive immunity, but it remains unclear how intratumoural treatment with STING agonists yields anti-tumour effects. Here we demonstrate that intratumoural injection of the STING agonist cGAMP induces strong, rapid, and selective apoptosis of tumour endothelial cells (ECs) in implanted LLC tumour, melanoma and breast tumour, but not in spontaneous breast cancer and melanoma. In both implanted and spontaneous tumours, cGAMP greatly increases TNFα from tumour-associated myeloid cells. However, compared to spontaneous tumour ECs, implanted tumour ECs are more vulnerable to TNFα-TNFR1 signalling-mediated apoptosis, which promotes effective anti-tumour activity. The spontaneous tumour's refractoriness to cGAMP is abolished by co-treatment with AKT 1/2 inhibitor (AKTi). Combined treatment with cGAMP and AKTi induces extensive tumour EC apoptosis, leading to extensive tumour apoptosis and marked growth suppression of the spontaneous tumour. These findings propose an advanced avenue for treating primary tumours that are refractory to single STING agonist therapy.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Protéines membranaires/agonistes , Tumeurs/traitement médicamenteux , Nucléotides cycliques/pharmacologie , Protéines proto-oncogènes c-akt/antagonistes et inhibiteurs , Animaux , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Apoptose/effets des médicaments et des substances chimiques , Apoptose/immunologie , Lignée cellulaire tumorale/transplantation , Modèles animaux de maladie humaine , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Femelle , Techniques de knock-down de gènes , Cellules endothéliales de la veine ombilicale humaine , Humains , Immunité innée/effets des médicaments et des substances chimiques , Injections intralésionnelles , Mâle , Protéines membranaires/antagonistes et inhibiteurs , Protéines membranaires/génétique , Protéines membranaires/métabolisme , Souris , Souris transgéniques , Tumeurs/vascularisation , Tumeurs/immunologie , Tumeurs/anatomopathologie , Nucléotides cycliques/usage thérapeutique , Phosphorylation/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-akt/métabolisme , Récepteur au facteur de nécrose tumorale de type I/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Transduction du signal/immunologie , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Microenvironnement tumoral/immunologie , Facteur de nécrose tumorale alpha/métabolisme
13.
Int J Med Sci ; 18(13): 2981-2989, 2021.
Article de Anglais | MEDLINE | ID: mdl-34220326

RÉSUMÉ

Salmonella causes salmonellosis, is a facultative anaerobe and is one of the common Gram-negative bacteria. Salmonella has anti-tumor potential and tumor-targeting activity. The heparin sulfate on cell surfaces can be cleaved by heparanase that is an endo-ß-D-glucuronidase. Heparanase can destroy the extracellular matrix and is involved in tumor metastasis and angiogenic activity. Previously, Salmonella was demonstrated to inhibit tumor metastasis. It remains unclear whether Salmonella inhibits metastasis by regulating heparanase. The expression of heparanase in Salmonella-treated tumor cells was found to be decreased. Transwell and wound-healing assays demonstrated the inhibition of cell migration after Salmonella treatment. Salmonella was found to influence the levels of phosphate-protein kinase B (P-AKT) and phosphate-extracellular regulated protein kinases (P-ERK), which are involved in heparanase expression. Salmonella reduced the heparanase expression induced upregulating PERK and PAKT signaling pathways. The mice bearing an experimental metastasis tumor model was used to evaluate the anti-tumor metastatic effects of Salmonella. Compared with the control group, Salmonella significantly reduced the number of metastatic nodules and enhanced survival. The results of our study indicate that Salmonella plays a vital role in the inhibition of tumor metastasis through the downregulation of heparanase.


Sujet(s)
Régulation de l'expression des gènes tumoraux/immunologie , Glucuronidase/métabolisme , Tumeurs/thérapie , Vaccins antisalmonella/immunologie , Animaux , Lignée cellulaire tumorale/transplantation , Modèles animaux de maladie humaine , Régulation négative/immunologie , Humains , Souris , Tumeurs/immunologie , Tumeurs/anatomopathologie , Salmonella/immunologie , Vaccins antisalmonella/administration et posologie
14.
J Immunol Res ; 2021: 9965099, 2021.
Article de Anglais | MEDLINE | ID: mdl-34307695

RÉSUMÉ

BACKGROUND: Immune checkpoint inhibitor therapy targeting antiprogrammed cell death-1 (anti-PD-1) or its ligand (anti-PD-L1) is effective in the treatment of some hepatocellular carcinomas (HCC). Hence, further identification of biological targets related to PD-L1 regulation in HCC is beneficial to improve the clinical efficacy of immunotherapy. Some HCC cells express lysine-specific demethylase 1A (KDM1A), which is implicated in the reduced survival time of patients. Here, we studied whether the level of PD-L1 and the immunosuppression are regulated by KDM1A and its miRNA in HCC cells. METHODS: In the present study, we studied clinical data from The Cancer Genome Atlas (TCGA) database. We performed qPCR and western blotting assays to measure the expression level of genes of interest. PD-L1 expression was also analyzed by FACS. Clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 was used to generate gene knockout cells to investigate the relationships of genes of interest. We also developed a reporter gene assay (RGA) to explore the changes in T cell-induced antitumor immunity relative to PD-L1 expression in HCC cells. The binding between proteins and promoters or miRNAs and their target genes was explored by luciferase reporter assays. RESULTS: The results showed that PD-L1 and KDM1A were increased in HCC patients and cells, and KDM1A promoted the expression of PD-L1 in HCC cells. Our findings showed that the enhancement of PD-L1 expression was not attributed to mitochondrial dysfunction caused by increases in KDM1A in HCC cells. Furthermore, we observed a lower level of MEF2D methylation in HCC cells than in normal human liver cells. Demethylated MEF2D could bind to the promoter of PD-L1 and activate its expression, while KDM1A interacted with MEF2D and acted as a demethylase to reduce its methylation. Moreover, a new miRNA, miR-329-3p, targeting KDM1A was found to regulate the PD-L1 expression profile in HCC cells. In the xenograft model, the tumors treated with miR-329-3p showed growth inhibition. CONCLUSIONS: Mechanistically, miR-329-3p inhibits tumor cellular immunosuppression and reinforces the response of tumor cells to T cell-induced cytotoxic effect by targeting KDM1A mRNA and downregulating its expression, which contributed to MEF2D demethylation and activation of PD-L1 expression.


Sujet(s)
Antigène CD274/génétique , Carcinome hépatocellulaire/immunologie , Histone Demethylases/métabolisme , Tumeurs du foie/immunologie , Animaux , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/anatomopathologie , Lignée cellulaire tumorale/transplantation , Déméthylation de l'ADN , Modèles animaux de maladie humaine , Épigenèse génétique/immunologie , Régulation de l'expression des gènes tumoraux/immunologie , Techniques de knock-out de gènes , Histone Demethylases/génétique , Humains , Foie/anatomopathologie , Tumeurs du foie/génétique , Tumeurs du foie/anatomopathologie , Facteurs de transcription MEF2/génétique , Facteurs de transcription MEF2/métabolisme , Mâle , Souris , microARN/métabolisme , Régions promotrices (génétique) , Échappement de la tumeur à la surveillance immunitaire/génétique
15.
Mol Pharm ; 18(9): 3342-3351, 2021 09 06.
Article de Anglais | MEDLINE | ID: mdl-34324363

RÉSUMÉ

Poor distribution of nanocarriers at the tumor site and insufficient drug penetration into the tissue are major challenges in the development of effective and safe cancer therapy. Here, we aim to enhance the therapeutic effect of liposomes by accumulating doxorubicin-loaded liposomes at high concentrations in and around the tumor, followed by heat-triggered drug release to facilitate low-molecular-weight drug penetration throughout the tumor. A cyclic RGD peptide (cRGD) was incorporated into liposomes decorated with a thermosensitive polymer that allowed precise tuning of drug release temperature (i.e., Polymer-lip) to develop a targeted thermosensitive liposome (cRGD-Polymer-lip). Compared with conventional thermosensitive liposomes, cRGD-Polymer-lip enhanced the binding of liposomes to endothelial cells, leading to their accumulation at the tumor site upon intravenous administration in tumor-bearing mice. Drug release triggered by local heating strongly inhibited tumor growth. Notably, tumor remission was achieved via multiple administrations of cRGD-Polymer-lip and heat treatments. Thus, combining the advantages of tumor neovascular targeting and heat-triggered drug release, these liposomes offer high potential for minimally invasive and effective cancer chemotherapy.


Sujet(s)
Antibiotiques antinéoplasiques/administration et posologie , Système d'administration de médicaments à base de nanoparticules/composition chimique , Tumeurs/traitement médicamenteux , Néovascularisation pathologique/traitement médicamenteux , Animaux , Antibiotiques antinéoplasiques/pharmacocinétique , Lignée cellulaire tumorale/transplantation , Modèles animaux de maladie humaine , Doxorubicine/administration et posologie , Doxorubicine/analogues et dérivés , Doxorubicine/pharmacocinétique , Libération de médicament , Femelle , Température élevée , Humains , Liposomes , Souris , Tumeurs/vascularisation , Tumeurs/anatomopathologie , Néovascularisation pathologique/anatomopathologie , Peptides cycliques/composition chimique , Polyéthylène glycols/administration et posologie , Polyéthylène glycols/pharmacocinétique , Polymères/composition chimique
16.
Hepatology ; 74(5): 2652-2669, 2021 11.
Article de Anglais | MEDLINE | ID: mdl-34157147

RÉSUMÉ

BACKGROUND AND AIMS: Lenvatinib is an effective drug in advanced HCC. Its combination with the anti-PD1 (programmed cell death protein 1) immune checkpoint inhibitor, pembrolizumab, has generated encouraging results in phase Ib and is currently being tested in phase III trials. Here, we aimed to explore the molecular and immunomodulatory effects of lenvatinib alone or in combination with anti-PD1. APPROACH AND RESULTS: We generated three syngeneic models of HCC in C57BL/6J mice (subcutaneous and orthotopic) and randomized animals to receive placebo, lenvatinib, anti-PD1, or combination treatment. Flow cytometry, transcriptomic, and immunohistochemistry analyses were performed in tumor and blood samples. A gene signature, capturing molecular features associated with the combination therapy, was used to identify a subset of candidates in a cohort of 228 HCC patients who might respond beyond what is expected for monotherapies. In mice, the combination treatment resulted in tumor regression and shorter time to response compared to monotherapies (P < 0.001). Single-agent anti-PD1 induced dendritic and T-cell infiltrates, and lenvatinib reduced the regulatory T cell (Treg) proportion. However, only the combination treatment significantly inhibited immune suppressive signaling, which was associated with the TGFß pathway and induced an immune-active microenvironment (P < 0.05 vs. other therapies). Based on immune-related genomic profiles in human HCC, 22% of patients were identified as potential responders beyond single-agent therapies, with tumors characterized by Treg cell infiltrates, low inflammatory signaling, and VEGFR pathway activation. CONCLUSIONS: Lenvatinib plus anti-PD1 exerted unique immunomodulatory effects through activation of immune pathways, reduction of Treg cell infiltrate, and inhibition of TGFß signaling. A gene signature enabled the identification of ~20% of human HCCs that, although nonresponding to single agents, could benefit from the proposed combination.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Carcinome hépatocellulaire/traitement médicamenteux , Tumeurs du foie/traitement médicamenteux , Phénylurées/pharmacologie , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Quinoléines/pharmacologie , Animaux , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Carcinome hépatocellulaire/immunologie , Lignée cellulaire tumorale/transplantation , Modèles animaux de maladie humaine , Tests de criblage d'agents antitumoraux , Synergie des médicaments , Femelle , Humains , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Tumeurs du foie/immunologie , Tumeurs du foie/anatomopathologie , Mâle , Souris , Phénylurées/usage thérapeutique , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Quinoléines/usage thérapeutique , Échappement de la tumeur à la surveillance immunitaire/effets des médicaments et des substances chimiques , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Microenvironnement tumoral/immunologie
17.
Nat Commun ; 12(1): 3414, 2021 06 07.
Article de Anglais | MEDLINE | ID: mdl-34099731

RÉSUMÉ

Pancreatic ductal adenocarcinoma (PDAC) patients have a 5-year survival rate of only 8% largely due to late diagnosis and insufficient therapeutic options. Neutrophils are among the most abundant immune cell type within the PDAC tumor microenvironment (TME), and are associated with a poor clinical prognosis. However, despite recent advances in understanding neutrophil biology in cancer, therapies targeting tumor-associated neutrophils are lacking. Here, we demonstrate, using pre-clinical mouse models of PDAC, that lorlatinib attenuates PDAC progression by suppressing neutrophil development and mobilization, and by modulating tumor-promoting neutrophil functions within the TME. When combined, lorlatinib also improves the response to anti-PD-1 blockade resulting in more activated CD8 + T cells in PDAC tumors. In summary, this study identifies an effect of lorlatinib in modulating tumor-associated neutrophils, and demonstrates the potential of lorlatinib to treat PDAC.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Carcinome du canal pancréatique/traitement médicamenteux , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Lactames macrocycliques/pharmacologie , Granulocytes neutrophiles/effets des médicaments et des substances chimiques , Tumeurs du pancréas/traitement médicamenteux , Aminopyridines , Animaux , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Lymphocytes T CD8+/effets des médicaments et des substances chimiques , Lymphocytes T CD8+/immunologie , Carcinome du canal pancréatique/anatomopathologie , Lignée cellulaire tumorale/transplantation , Modèles animaux de maladie humaine , Synergie des médicaments , Femelle , Humains , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Lactames , Lactames macrocycliques/usage thérapeutique , Activation des lymphocytes/effets des médicaments et des substances chimiques , Lymphocytes TIL/effets des médicaments et des substances chimiques , Lymphocytes TIL/immunologie , Mâle , Souris , Souris transgéniques , Granulocytes neutrophiles/immunologie , Tumeurs du pancréas/génétique , Tumeurs du pancréas/immunologie , Tumeurs du pancréas/anatomopathologie , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/métabolisme , Pyrazoles , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Microenvironnement tumoral/immunologie
18.
Nat Commun ; 12(1): 3187, 2021 05 27.
Article de Anglais | MEDLINE | ID: mdl-34045459

RÉSUMÉ

Failure of conventional clinical therapies such as tumor resection and chemotherapy are mainly due to the ineffective control of tumor metastasis. Metastasis consists of three steps: (i) tumor cells extravasate from the primary sites into the circulation system via epithelial-mesenchymal transition (EMT), (ii) the circulating tumor cells (CTCs) form "micro-thrombi" with platelets to evade the immune surveillance in circulation, and (iii) the CTCs colonize in the pre-metastatic niche. Here, we design a systemic metastasis-targeted nanotherapeutic (H@CaPP) composed of an anti-inflammatory agent, piceatannol, and an anti-thrombotic agent, low molecular weight heparin, to hinder the multiple steps of tumor metastasis. H@CaPP is found efficiently impeded EMT, inhibited the formation of "micro-thrombi", and prevented the development of pre-metastatic niche. When combined with surgical resection or chemotherapy, H@CaPP efficiently inhibits tumor metastasis and prolonged overall survival of tumor-bearing mice. Collectively, we provide a simple and effective systemic metastasis-targeted nanotherapeutic for combating tumor metastasis.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/administration et posologie , Vecteurs de médicaments/composition chimique , Tumeurs expérimentales de la mamelle/thérapie , Métastase tumorale/thérapie , Nanomédecine théranostique/méthodes , Animaux , Anti-inflammatoires/administration et posologie , Anticoagulants/administration et posologie , Lignée cellulaire tumorale/transplantation , Traitement médicamenteux adjuvant/méthodes , Modèles animaux de maladie humaine , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Femelle , Héparine bas poids moléculaire/administration et posologie , Cellules endothéliales de la veine ombilicale humaine , Humains , Mâle , Glandes mammaires animales/anatomopathologie , Glandes mammaires animales/chirurgie , Tumeurs expérimentales de la mamelle/anatomopathologie , Souris , Nanoparticules/composition chimique , Cellules tumorales circulantes/effets des médicaments et des substances chimiques , Paclitaxel/administration et posologie , Étude de validation de principe , Rats , Stilbènes/administration et posologie
19.
Nat Commun ; 12(1): 3236, 2021 05 28.
Article de Anglais | MEDLINE | ID: mdl-34050151

RÉSUMÉ

Adenosine is an immunosuppressive factor that limits anti-tumor immunity through the suppression of multiple immune subsets including T cells via activation of the adenosine A2A receptor (A2AR). Using both murine and human chimeric antigen receptor (CAR) T cells, here we show that targeting A2AR with a clinically relevant CRISPR/Cas9 strategy significantly enhances their in vivo efficacy, leading to improved survival of mice. Effects evoked by CRISPR/Cas9 mediated gene deletion of A2AR are superior to shRNA mediated knockdown or pharmacological blockade of A2AR. Mechanistically, human A2AR-edited CAR T cells are significantly resistant to adenosine-mediated transcriptional changes, resulting in enhanced production of cytokines including IFNγ and TNF, and increased expression of JAK-STAT signaling pathway associated genes. A2AR deficient CAR T cells are well tolerated and do not induce overt pathologies in mice, supporting the use of CRISPR/Cas9 to target A2AR for the improvement of CAR T cell function in the clinic.


Sujet(s)
Immunothérapie adoptive/méthodes , Tumeurs/thérapie , Récepteur A2A à l'adénosine/génétique , Lymphocytes T/transplantation , Adénosine/métabolisme , Antagonistes des récepteurs A2 à l'adénosine/pharmacologie , Animaux , Systèmes CRISPR-Cas/génétique , Ingénierie cellulaire/méthodes , Lignée cellulaire tumorale/transplantation , Modèles animaux de maladie humaine , Femelle , Édition de gène , Régulation de l'expression des gènes tumoraux/immunologie , Techniques de knock-down de gènes , Techniques de knock-out de gènes , Humains , Lymphocytes TIL/immunologie , Souris , Souris transgéniques , Tumeurs/génétique , Tumeurs/immunologie , Petit ARN interférent/métabolisme , RNA-Seq , Récepteur A2A à l'adénosine/métabolisme , Récepteur ErbB-2/génétique , Récepteurs chimériques pour l'antigène/immunologie , Récepteurs chimériques pour l'antigène/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Transduction du signal/génétique , Transduction du signal/immunologie , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Échappement de la tumeur à la surveillance immunitaire/effets des médicaments et des substances chimiques , Échappement de la tumeur à la surveillance immunitaire/génétique
20.
Nat Commun ; 12(1): 2582, 2021 05 11.
Article de Anglais | MEDLINE | ID: mdl-33976133

RÉSUMÉ

Immune checkpoint blockers (ICBs) have failed in all phase III glioblastoma (GBM) trials. Here, we show that regulatory T (Treg) cells play a key role in GBM resistance to ICBs in experimental gliomas. Targeting glucocorticoid-induced TNFR-related receptor (GITR) in Treg cells using an agonistic antibody (αGITR) promotes CD4 Treg cell differentiation into CD4 effector T cells, alleviates Treg cell-mediated suppression of anti-tumor immune response, and induces potent anti-tumor effector cells in GBM. The reprogrammed GBM-infiltrating Treg cells express genes associated with a Th1 response signature, produce IFNγ, and acquire cytotoxic activity against GBM tumor cells while losing their suppressive function. αGITR and αPD1 antibodies increase survival benefit in three experimental GBM models, with a fraction of cohorts exhibiting complete tumor eradication and immune memory upon tumor re-challenge. Moreover, αGITR and αPD1 synergize with the standard of care treatment for newly-diagnosed GBM, enhancing the cure rates in these GBM models.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Glioblastome/traitement médicamenteux , Protéine associée au récepteur du TNF induit par les corticoïdes/agonistes , Lymphocytes T régulateurs/effets des médicaments et des substances chimiques , Animaux , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Lignée cellulaire tumorale/transplantation , Reprogrammation cellulaire/effets des médicaments et des substances chimiques , Reprogrammation cellulaire/immunologie , Modèles animaux de maladie humaine , Femelle , Glioblastome/immunologie , Humains , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Mémoire immunologique/effets des médicaments et des substances chimiques , Mâle , Souris , Souris knockout , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Lymphocytes T cytotoxiques/immunologie , Lymphocytes T régulateurs/immunologie , Lymphocytes T régulateurs/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...