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.
J Immunother Cancer ; 11(10)2023 10.
Article de Anglais | MEDLINE | ID: mdl-37802604

RÉSUMÉ

BACKGROUND: Adoptive cell therapy (ACT) with tumor-infiltrating lymphocytes (TILs) is a promising immunotherapeutic approach for patients with advanced solid tumors. While numerous advances have been made, the contribution of neoantigen-specific CD4+T cells within TIL infusion products remains underexplored and therefore offers a significant opportunity for progress. METHODS: We analyzed infused TIL products from metastatic melanoma patients previously treated with ACT for the presence of neoantigen-specific T cells. TILs were enriched on reactivity to neoantigen peptides derived and prioritized from patient sample-directed mutanome analysis. Enriched TILs were further investigated to establish the clonal neoantigen response with respect to function, transcriptomics, and persistence following ACT. RESULTS: We discovered that neoantigen-specific TIL clones were predominantly CD4+ T cells and were present in both therapeutic responders and non-responders. CD4+ TIL demonstrated an effector T cell response with cytotoxicity toward autologous tumor in a major histocompatibility complex class II-dependent manner. These results were validated by paired TCR and single cell RNA sequencing, which elucidated transcriptomic profiles distinct to neoantigen-specific CD4+ TIL. CONCLUSIONS: Despite methods which often focus on CD8+T cells, our study supports the importance of prospective identification of neoantigen-specific CD4+ T cells within TIL products as they are a potent source of tumor-specific effectors. We further advocate for the inclusion of neoantigen-specific CD4+ TIL in future ACT protocols as a strategy to improve antitumor immunity.


Sujet(s)
Lymphocytes TIL , Mélanome , Humains , Immunothérapie adoptive/méthodes , Études prospectives , Lymphocytes T CD4+
2.
Front Immunol ; 12: 702726, 2021.
Article de Anglais | MEDLINE | ID: mdl-34177968

RÉSUMÉ

Regulatory T cells (Tregs) are key immunosuppressive cells that promote tumor growth by hindering the effector immune response. Tregs utilize multiple suppressive mechanisms to inhibit pro-inflammatory responses within the tumor microenvironment (TME) by inhibition of effector function and immune cell migration, secretion of inhibitory cytokines, metabolic disruption and promotion of metastasis. In turn, Tregs are being targeted in the clinic either alone or in combination with other immunotherapies, in efforts to overcome the immunosuppressive TME and increase anti-tumor effects. However, it is now appreciated that Tregs not only suppress cells intratumorally via direct engagement, but also serve as key interactors in the peritumor, stroma, vasculature and lymphatics to limit anti-tumor immune responses prior to tumor infiltration. We will review the suppressive mechanisms that Tregs utilize to alter immune and non-immune cells outside and within the TME and discuss how these mechanisms collectively allow Tregs to create and promote a physical and biological barrier, resulting in an immune-excluded or limited tumor microenvironment.


Sujet(s)
Lymphocytes T régulateurs/immunologie , Échappement de la tumeur à la surveillance immunitaire/immunologie , Microenvironnement tumoral/immunologie , Animaux , Humains
3.
Nat Immunol ; 21(9): 1010-1021, 2020 09.
Article de Anglais | MEDLINE | ID: mdl-32661362

RÉSUMÉ

Robust CD8+ T cell memory is essential for long-term protective immunity but is often compromised in cancer, where T cell exhaustion leads to loss of memory precursors. Immunotherapy via checkpoint blockade may not effectively reverse this defect, potentially underlying disease relapse. Here we report that mice with a CD8+ T cell-restricted neuropilin-1 (NRP1) deletion exhibited substantially enhanced protection from tumor rechallenge and sensitivity to anti-PD1 immunotherapy, despite unchanged primary tumor growth. Mechanistically, NRP1 cell-intrinsically limited the self-renewal of the CD44+PD1+TCF1+TIM3- progenitor exhausted T cells, which was associated with their reduced ability to induce c-Jun/AP-1 expression on T cell receptor restimulation, a mechanism that may contribute to terminal T cell exhaustion at the cost of memory differentiation in wild-type tumor-bearing hosts. These data indicate that blockade of NRP1, a unique 'immune memory checkpoint', may promote the development of long-lived tumor-specific Tmem that are essential for durable antitumor immunity.


Sujet(s)
Lymphocytes T CD8+/immunologie , Protéines de points de contrôle immunitaires/métabolisme , Mélanome expérimental/immunologie , Neuropiline 1/métabolisme , Précurseurs lymphoïdes T/immunologie , Animaux , Lignée cellulaire tumorale , Humains , Protéines de points de contrôle immunitaires/génétique , Tolérance immunitaire , Immunité , Mémoire immunologique , Souris , Souris knockout , Neuropiline 1/génétique , Récepteur-1 de mort cellulaire programmée/métabolisme , Transduction du signal
4.
Cancer Immunol Immunother ; 69(12): 2465-2476, 2020 Dec.
Article de Anglais | MEDLINE | ID: mdl-32556443

RÉSUMÉ

Emm55 is a bacterial gene derived from Streptococcus pyogenes (S. pyogenes) that was cloned into a plasmid DNA vaccine (pAc/emm55). In this study, we investigated the anti-tumor efficacy of pAc/emm55 in a B16 murine melanoma model. Intralesional (IL) injections of pAc/emm55 significantly delayed tumor growth compared to the pAc/Empty group. There was a significant increase in the CD8+ T cells infiltrating into the tumors after pAc/emm55 treatment compared to the control group. In addition, we observed that IL injection of pAc/emm55 increased antigen-specific T cell infiltration into tumors. Depletion of CD4+ or CD8+ T cells abrogated the anti-tumor effect of pAc/emm55. Combination treatment of IL injection of pAc/emm55 with anti-PD-1 antibody significantly delayed tumor growth compared to either monotherapy. pAc/emm55 treatment combined with PD-1 blockade enhanced anti-tumor immune response and improved systemic anti-tumor immunity. Together, these strategies may lead to improvements in the treatment of patients with melanoma.


Sujet(s)
Antigènes bactériens/immunologie , Antinéoplasiques immunologiques/administration et posologie , Protéines de la membrane externe bactérienne/immunologie , Immunothérapie/méthodes , Mélanome expérimental/thérapie , Animaux , Antigènes bactériens/génétique , Protéines de la membrane externe bactérienne/génétique , Lymphocytes T CD8+/immunologie , Lignée cellulaire tumorale/transplantation , Association thérapeutique/méthodes , Femelle , Humains , Injections intralésionnelles , Lymphocytes TIL/immunologie , Mélanome expérimental/immunologie , Souris , Plasmides/administration et posologie , Plasmides/génétique , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/immunologie
5.
PLoS One ; 13(4): e0196033, 2018.
Article de Anglais | MEDLINE | ID: mdl-29694419

RÉSUMÉ

Intralesional (IL) injection of Rose Bengal (PV-10) induces regression of injected and uninjected lesions in several murine tumor models. In this study, we investigated the anti-tumor response of combining IL PV-10 with blockade of the PD-1 / PD-L1 pathway and the role of immune cell populations in eliciting this response. To investigate the role of T cell subsets in mediating an immune response, B16 or M05 melanoma-bearing mice received combination therapy as well as CD8+, CD4+, or CD25+ depleting antibodies. Tumor growth was measured. T cells were collected from spleens or tumors, and phenotype, activation markers, and reactivity were measured. Splenocytes from mice treated with combination therapy had increased OVA antigen-specific CD8+ T cells in M05-tumor-bearing mice. Depletion of CD4+ T cells or regulatory T cells (Tregs) in combination with IL PV-10 and anti-PD-1 antibody treatment resulted in an enhanced anti-tumor effect. Treatment with CD8+ depleting antibody abrogated anti-tumor immunity. These results support a clinical study for the safety and anti-tumor immune responses with combination therapy of IL PV-10 and PD-1/PD-L1 blockade.


Sujet(s)
Anticorps monoclonaux/administration et posologie , Protocoles de polychimiothérapie antinéoplasique/administration et posologie , Mélanome expérimental/traitement médicamenteux , Rose de Bengale/administration et posologie , Tumeurs cutanées/traitement médicamenteux , Lymphocytes T/immunologie , Animaux , Anticorps monoclonaux/pharmacologie , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Antigène CD274/antagonistes et inhibiteurs , Antigènes CD4/métabolisme , Antigènes CD8/métabolisme , Lignée cellulaire tumorale , Cytotoxicité immunologique , Humains , Injections intralésionnelles , Injections péritoneales , Sous-unité alpha du récepteur à l'interleukine-2/métabolisme , Mélanome expérimental/immunologie , Souris , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Rose de Bengale/pharmacologie , Tumeurs cutanées/immunologie , Lymphocytes T/effets des médicaments et des substances chimiques , Résultat thérapeutique
6.
PLoS One ; 11(4): e0153053, 2016.
Article de Anglais | MEDLINE | ID: mdl-27050669

RÉSUMÉ

Tumor-infiltrating lymphocytes (TIL) has been associated with improved survival in cancer patients. Within the tumor microenvironment, regulatory cells and expression of co-inhibitory immune checkpoint molecules can lead to the inactivation of TIL. Hence, there is a need to develop strategies that disrupt these negative regulators to achieve robust anti-tumor immune responses. We evaluated the blockade of immune checkpoints and their effect on T cell infiltration and function. We examined the ability of TIL to induce tumor-specific immune responses in vitro and in vivo. TIL isolated from tumor bearing mice were tumor-specific and expressed co-inhibitory immune checkpoint molecules. Administration of monoclonal antibodies against immune checkpoints led to a significant delay in tumor growth. However, anti-PD-L1 antibody treated mice had a significant increase in T cell infiltration and IFN-γ production compared to other groups. Adoptive transfer of in vitro expanded TIL from tumors of anti-PD-L1 antibody treated mice led to a significant delay in tumor growth. Blockade of co-inhibitory immune checkpoints could be an effective strategy to improve TIL infiltration and function.


Sujet(s)
Transfert adoptif , Lymphocytes TIL/immunologie , Tumeurs expérimentales/thérapie , Animaux , Antigène CD274/antagonistes et inhibiteurs , Antigène CD274/immunologie , Femelle , Souris , Souris de lignée C57BL , Tumeurs expérimentales/immunologie
7.
Healthc Financ Manage ; 65(9): 80-3, 2011 Sep.
Article de Anglais | MEDLINE | ID: mdl-21923045

RÉSUMÉ

The current statutory and regulatory guidance for recovery audit contractor (RAC) extrapolation leaves providers with minimal protection against the process and a limited ability to challenge overpayment demands. Providers not only should understand the statutory and regulatory basis for extrapolation forecast, but also should be able to assess their extrapolation risk and their recourse through regulatory safeguards against contractor error. Providers also should aggressively appeal all incorrect RAC denials to minimize the potential impact of extrapolation.


Sujet(s)
, Examen des demandes de remboursement d'assurance/organisation et administration , Audit médical/économie , Services externalisés , États-Unis
8.
Healthc Financ Manage ; 65(3): 62-6, 68, 2011 Mar.
Article de Anglais | MEDLINE | ID: mdl-21449307

RÉSUMÉ

Review by an administrative law judge (ALJ) constitutes the third level of appeal for healthcare providers seeking to overturn reverse recovery audit contractor (RAC) findings of overpayment of Medicare claims. An analysis of the results of RAC appeals submitted by 30 New York hospitals during the demonstration project has disclosed two deficiencies in the ALJ review process: inconsistent ALJ decision making and a lack of an appropriate feedback mechanism to correct erroneous overpayment determinations. The Centers for Medicare & Medicaid Services should take advantage of feedback from such studies as an impetus to reevaluate and streamline the RAC appeals process.


Sujet(s)
Efficacité fonctionnement , Examen des demandes de remboursement d'assurance/organisation et administration , Medicare (USA)/économie , Services externalisés , Humains , Medicare (USA)/organisation et administration , État de New York , États-Unis
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE