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










Base de données
Gamme d'année
1.
J Immunother Cancer ; 9(8)2021 08.
Article de Anglais | MEDLINE | ID: mdl-34389618

RÉSUMÉ

BACKGROUND: GWN323 is an IgG1 monoclonal antibody (mAb) against the glucocorticoid-induced tumor necrosis factor receptor-related protein. This first-in-human, open-label phase I/Ib study aimed to investigate the safety and tolerability and to identify the recommended doses of GWN323 with/without spartalizumab, an anti-programmed cell death receptor-1 agent, for future studies. Pharmacokinetics, preliminary efficacy and efficacy biomarkers were also assessed. METHODS: Patients (aged ≥18 years) with advanced/metastatic solid tumors with Eastern Cooperative Oncology Group performance status of ≤2 were included. GWN323 (10-1500 mg) or GWN323+spartalizumab (GWN323 10-750 mg+spartalizumab 100-300 mg) were administered intravenously at various dose levels and schedules during the dose-escalation phase. Dose-limiting toxicities (DLTs) were assessed during the first 21 days in a single-agent arm and 42 days in a combination arm. Adverse events (AEs) were graded per National Cancer Institute-Common Toxicity Criteria for Adverse Events V.4.03 and efficacy was assessed using Response Evaluation Criteria in Solid Tumors V.1.1. RESULTS: Overall, 92 patients (single-agent, n=39; combination, n=53) were included. The maximum administered doses (MADs) in the single-agent and combination arms were GWN323 1500 mg every 3 weeks (q3w) and GWN323 750 mg+spartalizumab 300 mg q3w, respectively. No DLTs were observed with single-agent treatment. Three DLTs (6%, all grade ≥3) were noted with combination treatment: blood creatine phosphokinase increase, respiratory failure and small intestinal obstruction. Serious AEs were reported in 30.8% and 34.0%, and drug-related AEs were reported in 82.1% and 77.4% of patients with single-agent and combination treatments, respectively. Disease was stable in 7 patients and progressed in 26 patients with single-agent treatment. In combination arm patients, 1 had complete response (endometrial cancer); 3, partial response (rectal cancer, adenocarcinoma of colon and melanoma); 14, stable disease; and 27, disease progression. GWN323 exhibited a pharmacokinetic profile typical of mAbs with a dose-dependent increase in the pharmacokinetic exposure. Inconsistent decreases in regulatory T cells and increases in CD8+ T cells were observed in the combination arm. Gene expression analyses showed no significant effect of GWN323 on interferon-γ or natural killer-cell signatures. CONCLUSIONS: GWN323, as a single agent and in combination, was well tolerated in patients with relapsed/refractory solid tumors. The MAD was 1500 mg q3w for single-agent and GWN323 750 mg+spartalizumab 300 mg q3w for combination treatments. Minimal single-agent activity and modest clinical benefit were observed with the spartalizumab combination. TRIAL REGISTRATION NUMBER: NCT02740270.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Lymphomes/traitement médicamenteux , Tumeurs/traitement médicamenteux , Adulte , Sujet âgé , Anticorps monoclonaux humanisés/administration et posologie , Antinéoplasiques immunologiques/administration et posologie , Antinéoplasiques immunologiques/immunologie , Relation dose-effet des médicaments , Femelle , Protéine associée au récepteur du TNF induit par les corticoïdes/antagonistes et inhibiteurs , Protéine associée au récepteur du TNF induit par les corticoïdes/immunologie , Humains , Inhibiteurs de points de contrôle immunitaires/administration et posologie , Mâle , Dose maximale tolérée , Adulte d'âge moyen
2.
Biochem Pharmacol ; 178: 114110, 2020 08.
Article de Anglais | MEDLINE | ID: mdl-32569630

RÉSUMÉ

The tumor necrosis factor (TNF) superfamily (TNFSF) includes about thirty structurally related receptors (TNFSFRs) and about twenty protein ligands that bind to one or more of these receptors. Receptors of the tumor necrosis factor (TNF) superfamily (TNFSFRs) are pharmacological targets for treatment of inflammatory and autoimmune diseases. Currently, drugs targeting TNFSFR signaling are biological drugs (monoclonal antibodies, decoy receptors) aimed at binding and sequestering TNFSFR ligands. The glucocorticoid-induced tumor necrosis factor receptor-related gene (GITR) signaling is involved in a series of inflammatory and autoimmune diseases, such as rheumatoid arthritis and Crohn's disease. Our study aimed at repurposing FDA approved small molecules as protein-protein disruptors at the GITR ligand (GITRL) trimer, in order to inhibit the binding of GITRL to its receptor (GITR). A structure based molecular modeling approach was carried out to identify, through high throughput virtual screening, GITRL monomer-monomer disruptors. We used a database of ~8,000 FDA approved drugs, and after virtual screening, we focused on two hit compounds, minocycline and oxytetracycline. These two compounds were tested for their capability to modulate IL-17, IL-21 and RORγT expression in T lymphocytes, isolated from wild-type and GITR knock-out (GITR-/-) mice. Minocycline showed immunomodulatory effects specific to GITR activation and could represent a novel pharmacological tool to treat inflammatory diseases.


Sujet(s)
Anti-inflammatoires/composition chimique , Protéine associée au récepteur du TNF induit par les corticoïdes/antagonistes et inhibiteurs , Minocycline/composition chimique , Oxytétracycline/composition chimique , Facteurs de nécrose tumorale/composition chimique , Animaux , Anti-inflammatoires/pharmacologie , Anticorps monoclonaux/pharmacologie , Sites de fixation , Antigènes CD3/antagonistes et inhibiteurs , Antigènes CD3/immunologie , Régulation de l'expression des gènes , Protéine associée au récepteur du TNF induit par les corticoïdes/composition chimique , Protéine associée au récepteur du TNF induit par les corticoïdes/déficit , Protéine associée au récepteur du TNF induit par les corticoïdes/immunologie , Tests de criblage à haut débit , Interleukine-17/génétique , Interleukine-17/immunologie , Interleukines/génétique , Interleukines/immunologie , Mâle , Souris , Souris knockout , Minocycline/pharmacologie , Membre-3 du groupe F de la sous-famille-1 de récepteurs nucléaires/génétique , Membre-3 du groupe F de la sous-famille-1 de récepteurs nucléaires/immunologie , Oxytétracycline/pharmacologie , Culture de cellules primaires , Liaison aux protéines , Structure en hélice alpha , Structure en brin bêta , Motifs et domaines d'intéraction protéique , Rate/cytologie , Rate/effets des médicaments et des substances chimiques , Rate/immunologie , Lymphocytes T/cytologie , Lymphocytes T/effets des médicaments et des substances chimiques , Lymphocytes T/immunologie , Facteurs de nécrose tumorale/immunologie
3.
Int Immunopharmacol ; 84: 106494, 2020 Jul.
Article de Anglais | MEDLINE | ID: mdl-32304993

RÉSUMÉ

Rheumatoid arthritis (RA) is an autoimmune disease classified by uncontrolled joint inflammation leading to the destruction of both cartilage and joints. Despite progress made in RA treatment in the past decade, new drugs with high efficacy and fewer long-term adverse effects are still needed; thus, safe anti-inflammatory therapies for RA are urgently needed. Previous results demonstrated that the CXCR3 antagonist is an extremely attractive therapeutic target for the treatment of several autoimmune diseases, suggesting that it might have an inhibitory effect on RA. In this study, we investigated the effect of AMG487, a selective CXCR3 antagonist, on collagen-induced arthritis (CIA) in mice and evaluated its potential therapeutic mechanism.Following induction of CIA, mice were treated with AMG487 (5 mg/kg, intraperitoneally), to investigate their protective effects against CIA. CD4, CD25, CCR6, IL-9, NF-κB, IL-6, IL-17A, IL-21, STAT6 and Foxp3 expressing GITR+ and CD45+ cells were measured in the spleen using flow cytometry to assess anti-inflammatory effects of AMG487. The mRNA and protein expression of GITR, CCR6, IL-9, and IL-21 were measured using quantitative real-time PCR and western blot analysis in knee tissue. AMG487 significantly alleviated joint inflammation by decreasing GITR+CD25+, GITR+CD45+, GITR+IL-9+, GITR+NF-κB+ CD45+CD4+, CD45+CCR6+, CD45+IL-6+ cells, CD45+IL-17A+, and CD45+IL-21+, and increasing GITR+Foxp3+ and GITR+STAT6+ cells. There was a significant decrease in mRNA and protein expression of GITR, CD4, CCR6, IL-6, IL-9, and IL-21 in knee tissue of CIA mice. This study demonstrates that AMG487 has a potential therapeutic effect on RA and could explore novel anti-inflammatory therapies for its treatment.


Sujet(s)
Acétamides/pharmacologie , Anti-inflammatoires/pharmacologie , Arthrite expérimentale/traitement médicamenteux , Protéine associée au récepteur du TNF induit par les corticoïdes/antagonistes et inhibiteurs , Sous-populations de lymphocytes/effets des médicaments et des substances chimiques , Pyrimidinones/pharmacologie , Récepteurs CXCR3/antagonistes et inhibiteurs , Acétamides/administration et posologie , Animaux , Anti-inflammatoires/administration et posologie , Arthrite expérimentale/induit chimiquement , Collagène/pharmacologie , Protéine associée au récepteur du TNF induit par les corticoïdes/métabolisme , Médiateurs de l'inflammation/antagonistes et inhibiteurs , Injections péritoneales , Sous-unité alpha du récepteur à l'interleukine-2/métabolisme , Interleukines/métabolisme , Antigènes CD45/métabolisme , Sous-populations de lymphocytes/métabolisme , Mâle , Souris , Souris de lignée DBA , Pyrimidinones/administration et posologie , Lymphocytes T régulateurs/métabolisme
4.
J Immunother Cancer ; 7(1): 311, 2019 11 20.
Article de Anglais | MEDLINE | ID: mdl-31747946

RÉSUMÉ

BACKGROUND: TNF receptor family agonists and checkpoint blockade combination therapies lead to minimal tumor clearance of poorly immunogenic tumors. Therefore, a need to enhance the efficacy of this combination therapy arises. Antigen-presenting cells (APCs) present antigen to T cells and steer the immune response through chemokine and cytokine secretion. DRibbles (DR) are tumor-derived autophagosomes containing tumor antigens and innate inflammatory adjuvants. METHODS: Using preclinical murine lung and pancreatic cancer models, we assessed the triple combination therapy of GITR agonist and PD-1 blocking antibodies with peritumoral injections of DRibbles-pulsed-bone marrow cells (BMCs), which consisted mainly of APCs, or CD103+ cross-presenting dendritic cells (DCs). Immune responses were assessed by flow cytometry. FTY720 was used to prevent T-cell egress from lymph nodes to assess lymph node involvement, and MHC-mismatched-BMCs were used to assess the necessity of antigen presentation by the peritumorally-injected DR-APCs. RESULTS: Tritherapy increased survival and cures in tumor-bearing mice compared to combined antibody therapy or peritumoral DR-BMCs alone. Peritumorally-injected BMCs remained within the tumor for at least 14 days and tritherapy efficacy was dependent on both CD4+ and CD8+ T cells. Although the overall percent of tumor-infiltrating T cells remained similar, tritherapy increased the ratio of effector CD4+ T cells-to-regulatory T cells, CD4+ T-cell cytokine production and proliferation, and CD8+ T-cell cytolytic activity in the tumor. Despite tritherapy-induced T-cell activation and cytolytic activity in lymph nodes, this T-cell activation was not required for tumor regression and enhanced survival. Replacement of DR-BMCs with DR-pulsed-DCs in the tritherapy led to similar antitumor effects, whereas replacement with DRibbles was less effective but delayed tumor growth. Interestingly, peritumoral administration of DR-pulsed MHC-mismatched-APCs in the tritherapy led to similar antitumor effects as MHC-matched-APCs, indicating that the observed enhanced antitumor effect was mediated independently of antigen presentation by the administered APCs. CONCLUSIONS: Overall, these results demonstrate that peritumoral DR-pulsed-BMC/DC administration synergizes with GITR agonist and PD-1 blockade to locally modulate and sustain tumor effector T-cell responses independently of T cell priming and perhaps through innate inflammatory modulations mediated by the DRibbles adjuvant. We offer a unique approach to modify the tumor microenvironment to benefit T-cell-targeted immunotherapies.


Sujet(s)
Anticorps/usage thérapeutique , Cellules présentatrices d'antigène/immunologie , Cellules de la moelle osseuse/immunologie , Protéine associée au récepteur du TNF induit par les corticoïdes/antagonistes et inhibiteurs , Tumeurs du poumon/thérapie , Tumeurs du pancréas/thérapie , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Animaux , Présentation d'antigène , Antigènes néoplasiques/immunologie , Lignée cellulaire tumorale , Cytokines/immunologie , Protéine associée au récepteur du TNF induit par les corticoïdes/agonistes , Tumeurs du poumon/immunologie , Lymphocytes TIL/immunologie , Souris de lignée BALB C , Souris de lignée C57BL , Tumeurs du pancréas/immunologie , Phagosomes/immunologie , Lymphocytes T/immunologie
5.
Clin Cancer Res ; 25(21): 6501-6510, 2019 11 01.
Article de Anglais | MEDLINE | ID: mdl-31358539

RÉSUMÉ

PURPOSE: Determine the differential effect of a FcγR-binding, mIgG2a anti-GITR antibody in mouse tumor models, and characterize the tumor microenvironment for the frequency of GITR expression in T-cell subsets from seven different human solid tumors.Experimental Design: For mouse experiments, wild-type C57BL/6 mice were subcutaneously injected with MC38 cells or B16 cells, and BALB/c mice were injected with CT26 cells. Mice were treated with the anti-mouse GITR agonist antibody 21B6, and tumor burden and survival were monitored. GITR expression was evaluated at the single-cell level using flow cytometry (FC). A total of 213 samples were evaluated for GITR expression by IHC, 63 by FC, and 170 by both in seven human solid tumors: advanced hepatocellular carcinoma, non-small cell lung cancer (NSCLC), renal cell carcinoma, pancreatic carcinoma, head and neck carcinoma, melanoma, and ovarian carcinoma. RESULTS: The therapeutic benefit of 21B6 was greatest in CT26 followed by MC38, and was least in the B16 tumor model. The frequency of CD8 T cells and effector CD4 T cells within the immune infiltrate correlated with response to treatment with GITR antibody. Analysis of clinical tumor samples showed that NSCLC, renal cell carcinoma, and melanoma had the highest proportions of GITR-expressing cells and highest per-cell density of GITR expression on CD4+ Foxp3+ T regulatory cells. IHC and FC data showed similar trends with a good correlation between both techniques. CONCLUSIONS: Human tumor data suggest that NSCLC, renal cell carcinoma, and melanoma should be the tumor subtypes prioritized for anti-GITR therapy development.


Sujet(s)
Anticorps anti-idiotypiques/pharmacologie , Protéine associée au récepteur du TNF induit par les corticoïdes/génétique , Mélanome expérimental/génétique , Sous-populations de lymphocytes T/immunologie , Animaux , Anticorps anti-idiotypiques/immunologie , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cytométrie en flux , Facteurs de transcription Forkhead/génétique , Protéine associée au récepteur du TNF induit par les corticoïdes/antagonistes et inhibiteurs , Humains , Mélanome expérimental/traitement médicamenteux , Mélanome expérimental/immunologie , Mélanome expérimental/anatomopathologie , Souris , Récepteurs du fragment Fc des IgG/immunologie , Sous-populations de lymphocytes T/anatomopathologie , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Microenvironnement tumoral/immunologie
6.
Nat Med ; 25(5): 759-766, 2019 05.
Article de Anglais | MEDLINE | ID: mdl-31036879

RÉSUMÉ

Modulating T cell homeostatic mechanisms with checkpoint blockade can efficiently promote endogenous anti-tumor T cell responses1-11. However, many patients still do not benefit from checkpoint blockade12, highlighting the need for targeting of alternative immune pathways13. Glucocorticoid-induced tumor necrosis factor receptor-related protein (GITR) is an attractive target for immunotherapy, owing to its capacity to promote effector T cell (Teff) functions14,15 and hamper regulatory T cell (Treg) suppression16-20. On the basis of the potent preclinical anti-tumor activity of agonist anti-GITR antibodies, reported by us and others16,21,22, we initiated the first in-human phase 1 trial of GITR agonism with the anti-GITR antibody TRX518 ( NCT01239134 ). Here, we report the safety profile and immune effects of TRX518 monotherapy in patients with advanced cancer and provide mechanistic preclinical evidence to rationally combine GITR agonism with checkpoint blockade in future clinical trials. We demonstrate that TRX518 reduces circulating and intratumoral Treg cells to similar extents, providing an easily assessable biomarker of anti-GITR activity. Despite Treg reductions and increased Teff:Treg ratios, substantial clinical responses were not seen. Similarly, in mice with advanced tumors, GITR agonism was not sufficient to activate cytolytic T cells due to persistent exhaustion. We demonstrate that T cell reinvigoration with PD-1 blockade can overcome resistance of advanced tumors to anti-GITR monotherapy. These findings led us to start investigating TRX518 with PD-1 pathway blockade in patients with advanced refractory tumors ( NCT02628574 ).


Sujet(s)
Anticorps monoclonaux humanisés/usage thérapeutique , Protéine associée au récepteur du TNF induit par les corticoïdes/agonistes , Immunothérapie/méthodes , Animaux , Marqueurs biologiques tumoraux/immunologie , Conception de médicament , Protéine associée au récepteur du TNF induit par les corticoïdes/antagonistes et inhibiteurs , Protéine associée au récepteur du TNF induit par les corticoïdes/immunologie , Humains , Mélanome expérimental/immunologie , Mélanome expérimental/thérapie , Souris , Souris de lignée C57BL , Tumeurs/immunologie , Tumeurs/thérapie , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Lymphocytes T régulateurs/immunologie
7.
Sci Rep ; 9(1): 5562, 2019 04 03.
Article de Anglais | MEDLINE | ID: mdl-30944344

RÉSUMÉ

An anti-glucocorticoid induced TNF receptor (GITR) agonistic antibody (Ab) induces an antitumor immunity with both stimulation of effector T cells and inhibition of regulatory T cell activity. To enhance GITR Ab-mediated tumor immunity, we focused on the intratumoral route, since a tumor-localized high concentration of Ab would confer activation of only tumor-infiltrating T cells. First, in a murine colon cancer model, we showed that the intratumoral delivery of Ab significantly increased the number of effector T cells infiltrated into tumors, and suppressed tumor growth more effectively than the intraperitoneal and intravenous injections did. Then, we found that the injection of Ab into the peritumoral area induced a systemic antitumor immunity at a similar level to the intratumoral injection. Therefore, we hypothesized that the transfer of locally administrated Ab into tumor-draining lymph nodes (TDLNs) plays an important role in inducing an effective immunity. In fact, intratumorally or peritumorally injected Ab was detected in TDLNs, and resection of Ab-injected TDLNs significantly reduced GITR Ab-mediated systemic tumor immunity. Intratumoral injection showed less number of auto-reactive T cells in the spleen than the intraperitoneal injection did. Intratumoral delivery of GITR Ab is a promising approach to induce an effective immunity compared to the systemic delivery.


Sujet(s)
Anticorps monoclonaux/administration et posologie , Antinéoplasiques immunologiques/administration et posologie , Tumeurs du côlon/traitement médicamenteux , Tumeurs du côlon/immunologie , Protéine associée au récepteur du TNF induit par les corticoïdes/antagonistes et inhibiteurs , Animaux , Anticorps monoclonaux/pharmacocinétique , Anticorps monoclonaux/pharmacologie , Antinéoplasiques immunologiques/pharmacocinétique , Antinéoplasiques immunologiques/pharmacologie , Tumeurs du côlon/anatomopathologie , Femelle , Protéine associée au récepteur du TNF induit par les corticoïdes/immunologie , Injections intralésionnelles , Injections péritoneales , Injections veineuses , Interféron gamma/métabolisme , Noeuds lymphatiques/effets des médicaments et des substances chimiques , Noeuds lymphatiques/immunologie , Noeuds lymphatiques/anatomopathologie , Souris de lignée BALB C , Lymphocytes T/effets des médicaments et des substances chimiques , Lymphocytes T/immunologie
8.
Biochem Biophys Res Commun ; 509(1): 96-101, 2019 01 29.
Article de Anglais | MEDLINE | ID: mdl-30579597

RÉSUMÉ

The lymphopenic condition following autologous hematopoietic stem cell transplantation (HSCT) enhances the proliferation of T cells by engaging tumor-associated antigens, leading to the alteration of the T-cell repertoire towards antitumor immunity. However, cure by autologous HSCT alone have rarely occurred in the clinical setting. Since tumor-reactive lymphocytes preferentially proliferate during reconstitution of the immune system, we examined whether the priming of donor lymphocytes can strengthen the antitumor effect by HSCT in a CT26 murine colon cancer model. The systemic administration of an anti-glucocorticoid-induced TNF receptor (GITR) agonistic antibody (Ab) significantly increased the number of CT26-responsive T cells but not that of auto-reactive lymphocytes in donor mice. The infusion of non-primed and GITR Ab-primed donor lymphocytes suppressed the CT26 tumor growth, and only the primed lymphocytes eliminated tumors in all the treated mice. The frequency of CT26-responsive T cells was elevated in recipient mice infused with both primed and non-primed lymphocytes until 4 weeks after transplantation, while the frequency in recipients with primed lymphocytes was markedly elevated compared with that in mice harboring non-primed lymphocytes at 2 weeks. The frequencies of regulatory T cells and myeloid-derived suppressor cells were elevated in recipient mice infused with primed and non-primed lymphocytes 2 weeks after transplantation, and returned to normal levels by week 4. The combination of autologous HSCT with pre-immunization of donor lymphocytes is a promising strategy to induce strong antitumor immunity.


Sujet(s)
Anticorps/usage thérapeutique , Tumeurs du côlon/thérapie , Protéine associée au récepteur du TNF induit par les corticoïdes/immunologie , Transplantation de cellules souches hématopoïétiques/méthodes , Lymphocytes/immunologie , Animaux , Anticorps/immunologie , Lignée cellulaire tumorale , Tumeurs du côlon/immunologie , Femelle , Protéine associée au récepteur du TNF induit par les corticoïdes/antagonistes et inhibiteurs , Immunité , Immunisation , Souris , Souris de lignée BALB C , Transplantation autologue/méthodes
9.
Front Immunol ; 9: 2170, 2018.
Article de Anglais | MEDLINE | ID: mdl-30294332

RÉSUMÉ

Despite the potential to cure metastatic disease, immunotherapy on its own often fails outright or early on due to tumor immune evasion. To address this obstacle, we investigated combinations of anti-GITR, anti-PD1 and radiation therapy (XRT) in our previously developed anti-PD1 resistant 344SQ non-small cell lung adenocarcinoma preclinical tumor model. We hypothesized that targeting multiple mechanisms of immune evasion with this triple therapy would lead to an enhanced tumor-specific immune response and improve survival more so than any mono- or dual therapy. In a two tumor 344SQR murine model, treatment with anti-GITR, anti-PD1, and XRT led to significantly improved survival and an abscopal response, with half of the mice becoming tumor free. These mice showed durable response and increased CD4+ and CD8+ effector memory on tumor rechallenge. Regulatory T cells (Tregs) expressed the highest level of GITR at the tumor site and anti-GITR therapy drastically diminished Tregs at the tumor site. Anti-tumor effects were largely dependent on CD4+ T cells and partially dependent on CD8+ T cells. Anti-GITR IgG2a demonstrated superior efficacy to anti-GITR IgG1 in driving antitumor effects. Collectively, these results suggest that combinatorial strategies targeting multiple points of tumor immune evasion may lead to a robust and lasting antitumor response.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Carcinome pulmonaire non à petites cellules/thérapie , Protéine associée au récepteur du TNF induit par les corticoïdes/antagonistes et inhibiteurs , Tumeurs du poumon/thérapie , Lymphocytes T régulateurs/immunologie , Échappement de la tumeur à la surveillance immunitaire/immunologie , Animaux , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Carcinome pulmonaire non à petites cellules/immunologie , Carcinome pulmonaire non à petites cellules/mortalité , Carcinome pulmonaire non à petites cellules/anatomopathologie , Lignée cellulaire tumorale/transplantation , Chimioradiothérapie/effets indésirables , Chimioradiothérapie/méthodes , Modèles animaux de maladie humaine , Tests de criblage d'agents antitumoraux , Protéine associée au récepteur du TNF induit par les corticoïdes/agonistes , Protéine associée au récepteur du TNF induit par les corticoïdes/immunologie , Humains , Tumeurs du poumon/immunologie , Tumeurs du poumon/mortalité , Tumeurs du poumon/anatomopathologie , Mâle , Souris , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/immunologie , Analyse de survie , Lymphocytes T régulateurs/effets des médicaments et des substances chimiques , Lymphocytes T régulateurs/effets des radiations , Résultat thérapeutique , Charge tumorale/effets des médicaments et des substances chimiques , Charge tumorale/effets des radiations , Échappement de la tumeur à la surveillance immunitaire/effets des médicaments et des substances chimiques , Échappement de la tumeur à la surveillance immunitaire/effets des radiations
10.
Blood Adv ; 2(17): 2230-2241, 2018 09 11.
Article de Anglais | MEDLINE | ID: mdl-30194137

RÉSUMÉ

Direct activation of tumor infiltrating antigen-presenting cells (APCs) by intratumoral injection of STING agonists (STINGa) leads to regression of the treated lymphoma tumor. Because STING activation induces apoptosis in lymphoma cells in vitro, we distinguished between the direct therapeutic vs the indirect immunotherapeutic properties of STINGa in vivo. Employing wild-type or STING knockout hosts bearing either wild-type or STING knockout tumor cells, we demonstrated that local tumor regression is totally dependent on STING expression by the host and is therefore immune mediated. However, distant untreated tumors are weakly affected after injection of STINGa to a single tumor site. Therefore, using the STINGa currently being tested in clinical trials, we screened for immunomodulatory agents that could synergize with the STING pathway to induce a systemic antitumor immune response and regression of distant tumors. We combined the STINGa with agents that improve APC or T-cell function. We found that modulation of both APCs and T cells can enhance control of distant lymphoma tumors by STINGa. In particular, adding an anti-GITR antibody induced lymphocyte expansion in the lymph node draining the treated site followed by increased T-cell infiltration in the distant tumor. Furthermore, more of these CD8 T cells at the distant site expressed PD-1. Therefore, blockade of PD-1 further enhanced tumor control at the distant site, leading to cure in 50% of the mice. These preclinical data provide the rationale for testing local injection of STINGa followed by agonistic anti-GITR and anti-PD-1 antibodies as immunotherapy for human lymphoma.


Sujet(s)
Anticorps monoclonaux/usage thérapeutique , Immunothérapie/méthodes , Lymphomes/traitement médicamenteux , Protéines membranaires/agonistes , Animaux , Lymphocytes T CD8+/composition chimique , Lignée cellulaire tumorale , Synergie des médicaments , Femelle , Protéine associée au récepteur du TNF induit par les corticoïdes/antagonistes et inhibiteurs , Protéine associée au récepteur du TNF induit par les corticoïdes/immunologie , Hétérogreffes , Humains , Protéines membranaires/génétique , Souris , Souris de lignée BALB C , Souris de lignée C57BL , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/immunologie
11.
Expert Opin Ther Targets ; 22(9): 783-797, 2018 09.
Article de Anglais | MEDLINE | ID: mdl-30107134

RÉSUMÉ

INTRODUCTION: Triggering of the glucocorticoid-induced TNFR-related gene (GITR) increases the activation of T lymphocytes and other immune system cells; furthermore, its ligand, GITRL, delivers signals in the cells where it is expressed. Areas covered: This review describes the effects of GITR/GITRL triggering/inhibition in conventional T cells, regulatory T cells (Tregs), monocytes/macrophages, endothelial cells and other cells of the immune system. GITR triggering appears to be an approach for promoting tumor rejection, treating infection and boosting vaccinations in several murine models. GITR inhibition may be useful for inhibiting inflammation and autoimmune disease development. Expert opinion: The exciting antitumor activity of anti-GITR mAbs depends on CD8+ effector T cell activation and inhibition/deletion of tumor-infiltrating Tregs. Whether one of these effects is more relevant is still under debate. Inhibition of GITR triggering plays an interesting anti-inflammatory role, but the potential effect of long-term treatment is to be investigated. The use of adjuvants able to trigger GITR is promising regarding new vaccines. Finally, caution is recommended when translating the findings of experimental murine models to human diseases; biologicals modulating human GITR/GITRL system can behave differently from those modulating the murine GITR/GITRL system.


Sujet(s)
Protéine associée au récepteur du TNF induit par les corticoïdes/antagonistes et inhibiteurs , Immunothérapie/méthodes , Thérapie moléculaire ciblée , Animaux , Maladies auto-immunes/immunologie , Maladies auto-immunes/thérapie , Modèles animaux de maladie humaine , Protéine associée au récepteur du TNF induit par les corticoïdes/génétique , Protéine associée au récepteur du TNF induit par les corticoïdes/immunologie , Humains , Inflammation/immunologie , Inflammation/thérapie , Souris , Tumeurs/immunologie , Tumeurs/thérapie , Spécificité d'espèce
12.
Ann N Y Acad Sci ; 1417(1): 104-115, 2018 04.
Article de Anglais | MEDLINE | ID: mdl-29566262

RÉSUMÉ

Cancer immunotherapy involving blockade of immune checkpoint molecules, such as CTLA-4 and PD-1, has shown remarkable clinical success across several types of malignancies. However, a fraction of patients experience disease progression after treatment; thus, exploring resistant mechanisms for immune checkpoint inhibitors and improving their treatment outcome with additional modalities are of great importance. CD4+ regulatory T (Treg ) cells characterized by expression of the master regulatory transcription factor FOXP3 are a highly immune-suppressive subset of CD4+ T cells that maintain immune homeostasis. Several preclinical and clinical studies suggest that Treg cells hamper immune surveillance against cancer in healthy individuals, prevent the development of effective antitumor immunity in tumor-bearing patients, and promote tumor progression. Therefore, targeting Treg cells should be crucial to improving the treatment outcomes of cancer immunotherapy. Several clinical studies directly or indirectly targeting Treg cells in combination with immune checkpoint inhibitors are ongoing or being planned. Understanding the characteristics and roles of Treg cells in cancer settings could make disease-specific Treg -targeted therapy more efficacious and reduce the incidence of immune-related adverse effects mediated by Treg cell inhibition.


Sujet(s)
Immunothérapie/méthodes , Tumeurs/immunologie , Tumeurs/thérapie , Lymphocytes T régulateurs/immunologie , Anticorps monoclonaux/usage thérapeutique , Antigène CTLA-4/antagonistes et inhibiteurs , Antigène CTLA-4/immunologie , Protéine associée au récepteur du TNF induit par les corticoïdes/antagonistes et inhibiteurs , Protéine associée au récepteur du TNF induit par les corticoïdes/immunologie , Protéines du choc thermique HSP70/antagonistes et inhibiteurs , Humains , Indoleamine-pyrrole 2,3,-dioxygenase/antagonistes et inhibiteurs , Sous-unité alpha du récepteur à l'interleukine-2/antagonistes et inhibiteurs , Sous-unité alpha du récepteur à l'interleukine-2/immunologie , Modèles immunologiques , Inhibiteurs des phosphoinositide-3 kinases , Récepteurs CCR4/antagonistes et inhibiteurs , Récepteurs CCR4/immunologie , Récepteur au OX40/antagonistes et inhibiteurs , Récepteur au OX40/immunologie , Lymphocytes T régulateurs/classification , Facteur de croissance transformant bêta/antagonistes et inhibiteurs , Facteur de croissance endothéliale vasculaire de type A/antagonistes et inhibiteurs , Facteur de croissance endothéliale vasculaire de type A/immunologie
13.
Oncotarget ; 8(24): 39117-39130, 2017 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-28388572

RÉSUMÉ

Tumor progression is facilitated immunologically by mechanisms that include low antigen expression, an absence of coimmunostimulatory signals, and the presence of regulatory T cells (Tregs), all of which act to suppress and restrict effector T cells in the tumor. It may be possible to overcome these conditions by a combination of modulatory immunotherapy agents and tumor-antigen targeting to activate and drive effective antitumor T cell responses. Here, we demonstrated that co-administration of aGITR and aPD-1 monoclonal antibodies (mAb) in combination with a peptide vaccine (Vax) in mice bearing established tumors significantly delayed tumor growth and induced complete regression in 50% of the mice. This response was associated with increased expansion and functionality of potent Ag-specific polyfunctional CD8+ T cells, reduced Tregs, and the generation of memory T cells. Tumor regression correlated with the expansion of tumor-infiltrating antigen-specific CD8+ effector memory T cells, as depletion of this cell population significantly reduced the effectiveness of the triple combination Vax/aGITR/aPD-1 therapy. These findings support the concept that dual aGITR/aPD-1 combination with cancer vaccines may be a novel strategy against poorly immunogenic tumors.


Sujet(s)
Anticorps monoclonaux/pharmacologie , Antigènes néoplasiques/immunologie , Vaccins anticancéreux/administration et posologie , Protéine associée au récepteur du TNF induit par les corticoïdes/antagonistes et inhibiteurs , Mélanome expérimental/immunologie , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Lymphocytes T régulateurs/immunologie , Animaux , Apoptose , Lymphocytes T CD8+/immunologie , Prolifération cellulaire , Femelle , Immunothérapie , Mélanome expérimental/métabolisme , Mélanome expérimental/thérapie , Souris , Souris de lignée C57BL , Cellules cancéreuses en culture , Vaccination
14.
PLoS One ; 11(3): e0152622, 2016.
Article de Anglais | MEDLINE | ID: mdl-27027302

RÉSUMÉ

Toxoplasma gondii is a widespread parasite responsible for causing clinical diseases especially in pregnant and immunosuppressed individuals. Glucocorticoid-induced TNF receptor (GITR), which is also known as TNFRS18 and belongs to the TNF receptor superfamily, is found to be expressed in various cell types of the immune system and provides an important costimulatory signal for T cells and myeloid cells. However, the precise role of this receptor in the context of T. gondii infection remains elusive. Therefore, the current study investigated the role of GITR activation in the immunoregulation mechanisms induced during the experimental infection of mice with T. gondii. Our data show that T. gondii infection slightly upregulates GITR expression in Treg cells and B cells, but the most robust increment in expression was observed in macrophages and dendritic cells. Interestingly, mice infected and treated with an agonistic antibody anti-GITR (DTA-1) presented a robust increase in pro-inflammatory cytokine production at preferential sites of parasite replication, which was associated with the decrease in latent brain parasitism of mice under treatment with DTA-1. Several in vivo and in vitro analysis were performed to identify the cellular mechanisms involved in GITR activation upon infection, however no clear alterations were detected in the phenotype/function of macrophages, Tregs and B cells under treatment with DTA-1. Therefore, GITR appears as a potential target for intervention during infection by the parasite Toxoplasma gondii, even though further studies are still necessary to better characterize the immune response triggered by GITR activation during T. gondii infection.


Sujet(s)
Lymphocytes B/immunologie , Protéine associée au récepteur du TNF induit par les corticoïdes/immunologie , Cellules myéloïdes/immunologie , Lymphocytes T régulateurs/immunologie , Toxoplasma/immunologie , Toxoplasmose/immunologie , Animaux , Anticorps neutralisants/immunologie , Anticorps neutralisants/pharmacologie , Lymphocytes B/parasitologie , Femelle , Protéine associée au récepteur du TNF induit par les corticoïdes/antagonistes et inhibiteurs , Mâle , Souris , Cellules myéloïdes/parasitologie , Grossesse , Lymphocytes T régulateurs/parasitologie , Toxoplasmose/traitement médicamenteux
15.
Immunology ; 147(2): 190-203, 2016 Feb.
Article de Anglais | MEDLINE | ID: mdl-26501838

RÉSUMÉ

Lymphatic filariasis leads to profound impairment of parasite-specific T helper type 1 (Th1) and Th2 immune responses and significantly increases the expression of regulatory networks and regulatory effectors like transforming growth factor-ß, CD25, cytotoxic T-lymphocyte antigen 4, glucocorticoid-induced tumour necrosis factor receptor (GITR) and regulatory T (Treg) cells, which together play an important role in immunosuppression. While Treg cells suppress the activity of effector cells, monocyte dysfunction, characterized by an alternatively activated immunoregulatory phenotype, is one hypothesis that explains the lack of an antigen-specific T-cell response in infected individuals. In the present study, we administered neutralizing antibodies against the Treg cell-associated markers CD25 and GITR and observed its effects on filaria-induced immunosuppression. Our results show that administration of anti-CD25 and anti-GITR in infected animals not only arrested the accumulation of Treg cells and reduced arginase activity, but also led to an increase in the percentages of Th17 cells in the secondary lymphoid organs of mice. Elevated levels of interferon-γ and decreased levels of interleukin-10 were also noted in the culture supernatants of mouse splenocytes that were treated with neutralizing antibodies. Furthermore, treatment with neutralizing antibodies enhanced the expression of inducible nitric oxide synthase on host macrophages and CD40 on host dendritic cells with concomitant decreased expression of alternative activation markers Arg1, Ym1 and Fizz1, which together lead to reduced parasite burden in treated animals. In summary, administration of neutralizing antibodies helps in breaking the regulatory network in mice and limits parasite-induced immunosuppression at the earliest host-parasite interface.


Sujet(s)
Anticorps neutralisants/pharmacologie , Filariose lymphatique/traitement médicamenteux , Protéine associée au récepteur du TNF induit par les corticoïdes/antagonistes et inhibiteurs , Médiateurs de l'inflammation/immunologie , Sous-unité alpha du récepteur à l'interleukine-2/antagonistes et inhibiteurs , Lymphocytes T régulateurs/effets des médicaments et des substances chimiques , Cellules Th17/effets des médicaments et des substances chimiques , Animaux , Protéines de la membrane externe bactérienne/immunologie , Antigènes CD40/immunologie , Antigènes CD40/métabolisme , Cellules cultivées , Cellules dendritiques/effets des médicaments et des substances chimiques , Cellules dendritiques/immunologie , Cellules dendritiques/parasitologie , Modèles animaux de maladie humaine , Filariose lymphatique/immunologie , Filariose lymphatique/métabolisme , Filariose lymphatique/parasitologie , Granulocytes éosinophiles/effets des médicaments et des substances chimiques , Granulocytes éosinophiles/immunologie , Granulocytes éosinophiles/parasitologie , Protéine associée au récepteur du TNF induit par les corticoïdes/immunologie , Protéine associée au récepteur du TNF induit par les corticoïdes/métabolisme , Interactions hôte-parasite , Immunisation , Médiateurs de l'inflammation/métabolisme , Interféron gamma/immunologie , Interféron gamma/métabolisme , Interleukine-10/immunologie , Interleukine-10/métabolisme , Sous-unité alpha du récepteur à l'interleukine-2/immunologie , Sous-unité alpha du récepteur à l'interleukine-2/métabolisme , Activation des macrophages/effets des médicaments et des substances chimiques , Souris de lignée BALB C , Lymphocytes T régulateurs/immunologie , Lymphocytes T régulateurs/métabolisme , Lymphocytes T régulateurs/parasitologie , Cellules Th17/immunologie , Cellules Th17/métabolisme , Cellules Th17/parasitologie , Facteurs temps
16.
Oncol Rep ; 34(4): 1995-2001, 2015 Oct.
Article de Anglais | MEDLINE | ID: mdl-26239052

RÉSUMÉ

We attempted to enhance the antitumor effects of tumor lysate-pulsed dendritic cells by eliminating regulatory T cells. The combinatorial effects of dendritic cells and agonist anti-glucocorticoid-induced tumor necrosis factor receptor (anti-GITR) antibodies were investigated with respect to enhancement of the systemic immune response, elimination of regulatory T cells, and inhibition of tumor growth. To determine whether the combination of dendritic cells and anti­GITR antibodies could enhance systemic immune responses and inhibit primary tumor growth in a murine osteosarcoma (LM8) model. We established the following 4 groups of C3H mice (20 mice in total): i), control IgG-treated mice; ii), tumor lysate-pulsed dendritic cell­treated mice; iii), agonist anti-GITR antibody-treated mice; and iv), agonist anti-GITR antibody- and tumor lysate-pulsed dendritic cell­treated mice.The mice that received the agonist anti-GITR antibodies and tumor lysate-pulsed dendritic cells displayed inhibited primary growth, prolonged life time, reduced numbers of regulatory T lymphocytes in the spleen, elevated serum interferon-γ levels, increased number of CD8+ T lymphocytes. The mice that received combined therapy had reduced level of immunosuppressive cytokines in tumor tissue and serum. Combining agonist anti-GITR antibodies with tumor lysate-pulsed dendritic cells enhanced the systemic immune response. These findings provide further support for the continued development of agonist anti-GITR antibodies as an immunotherapeutic strategy for osteosarcoma. We suggest that our proposed immunotherapy could be developed further to improve osteosarcoma treatment.


Sujet(s)
Anticorps anti-idiotypiques/administration et posologie , Cellules dendritiques/immunologie , Protéine associée au récepteur du TNF induit par les corticoïdes/immunologie , Immunité innée/immunologie , Ostéosarcome/immunologie , Animaux , Anticorps anti-idiotypiques/immunologie , Lymphocytes T CD8+/immunologie , Prolifération cellulaire/génétique , Thérapie cellulaire et tissulaire , Protéine associée au récepteur du TNF induit par les corticoïdes/antagonistes et inhibiteurs , Humains , Immunothérapie , Souris , Ostéosarcome/anatomopathologie , Ostéosarcome/thérapie , Lymphocytes T régulateurs/immunologie
17.
Cancer Immunol Res ; 2(5): 448-58, 2014 May.
Article de Anglais | MEDLINE | ID: mdl-24795357

RÉSUMÉ

Induction of potent immune responses to self-antigens remains a major challenge in tumor immunology. We have shown that a vaccine based on alphavirus replicon particles (VRP) activates strong cellular and humoral immunity to tyrosinase-related protein-2 (TRP2) melanoma antigen, providing prophylactic and therapeutic effects in stringent mouse models. Here, we report that the immunogenicity and efficacy of this vaccine is increased in combination with either antagonist anti-CTL antigen-4 (CTLA-4) or agonist anti-glucocorticoid-induced TNF family-related gene (GITR) immunomodulatory monoclonal antibodies (mAb). In the challenging therapeutic setting, VRP-TRP2 plus anti-GITR or anti-CTLA-4 mAb induced complete tumor regression in 90% and 50% of mice, respectively. These mAbs had similar adjuvant effects in priming an adaptive immune response against the vaccine-encoded antigen, augmenting, respectively, approximately 4- and 2-fold the TRP2-specific CD8(+) T-cell response and circulating Abs, compared with the vaccine alone. Furthermore, while both mAbs increased the frequency of tumor-infiltrating CD8(+) T cells, anti-CTLA-4 mAb also increased the quantity of intratumor CD4(+)Foxp3(-) T cells expressing the negative costimulatory molecule programmed death-1 (PD-1). Concurrent GITR expression on these cells suggests that they might be controlled by anti-GITR mAbs, thus potentially explaining their differential accumulation under the two treatment conditions. These findings indicate that combining immunomodulatory mAbs with alphavirus-based anticancer vaccines can provide therapeutic antitumor immune responses in a stringent mouse model, suggesting potential utility in clinical trials. They also indicate that tumor-infiltrating CD4(+)Foxp3(-)PD-1(+) T cells may affect the outcome of immunomodulatory treatments.


Sujet(s)
Alphavirus/génétique , Anticorps monoclonaux/pharmacologie , Facteurs immunologiques/pharmacologie , Mélanome expérimental/immunologie , Vaccins à pseudo-particules virales/immunologie , Animaux , Antigène CTLA-4/antagonistes et inhibiteurs , Modèles animaux de maladie humaine , Femelle , Protéine associée au récepteur du TNF induit par les corticoïdes/antagonistes et inhibiteurs , Immunoglobuline G/immunologie , Intramolecular oxidoreductases/génétique , Intramolecular oxidoreductases/immunologie , Lymphocytes TIL/immunologie , Lymphocytes TIL/métabolisme , Mélanome expérimental/thérapie , Souris , Phénotype , Réplicon , Sous-populations de lymphocytes T/immunologie , Sous-populations de lymphocytes T/métabolisme , Lymphocytes T cytotoxiques/effets des médicaments et des substances chimiques , Vaccins à pseudo-particules virales/administration et posologie
18.
J Exp Med ; 210(9): 1685-93, 2013 Aug 26.
Article de Anglais | MEDLINE | ID: mdl-23897982

RÉSUMÉ

Fc γ receptor (FcγR) coengagement can facilitate antibody-mediated receptor activation in target cells. In particular, agonistic antibodies that target tumor necrosis factor receptor (TNFR) family members have shown dependence on expression of the inhibitory FcγR, FcγRIIB. It remains unclear if engagement of FcγRIIB also extends to the activities of antibodies targeting immunoregulatory TNFRs expressed by T cells. We have explored the requirement for activating and inhibitory FcγRs for the antitumor effects of antibodies targeting the TNFR glucocorticoid-induced TNFR-related protein (GITR; TNFRSF18; CD357) expressed on activated and regulatory T cells (T reg cells). We found that although FcγRIIB was dispensable for the in vivo efficacy of anti-GITR antibodies, in contrast, activating FcγRs were essential. Surprisingly, the dependence on activating FcγRs extended to an antibody targeting the non-TNFR receptor CTLA-4 (CD152) that acts as a negative regulator of T cell immunity. We define a common mechanism that correlated with tumor efficacy, whereby antibodies that coengaged activating FcγRs expressed by tumor-associated leukocytes facilitated the selective elimination of intratumoral T cell populations, particularly T reg cells. These findings may have broad implications for antibody engineering efforts aimed at enhancing the therapeutic activity of immunomodulatory antibodies.


Sujet(s)
Anticorps antitumoraux/pharmacologie , Antinéoplasiques/pharmacologie , Protéine associée au récepteur du TNF induit par les corticoïdes/antagonistes et inhibiteurs , Facteurs immunologiques/pharmacologie , Tumeurs/immunologie , Récepteurs du fragment Fc des IgG/métabolisme , Animaux , Antigène CTLA-4/métabolisme , Femelle , Protéine associée au récepteur du TNF induit par les corticoïdes/métabolisme , Déplétion lymphocytaire , Souris , Souris de lignée BALB C , Tumeurs/anatomopathologie , Lymphocytes T régulateurs/effets des médicaments et des substances chimiques , Lymphocytes T régulateurs/métabolisme
19.
Br J Pharmacol ; 165(7): 2089-99, 2012 Apr.
Article de Anglais | MEDLINE | ID: mdl-22029729

RÉSUMÉ

Glucocorticoid-induced TNFR-related (gitr) is a gene coding for a member of the TNF receptor superfamily. GITR activation by its ligand (GITRL) influences the activity of effector and regulatory T cells, thus participating in the development of immune response against tumours and infectious agents, as well as in autoimmune and inflammatory diseases. Notably, treating animals with GITR-Fc fusion protein ameliorates autoimmune/inflammatory diseases while GITR triggering, by treatment with anti-GITR mAb, is effective in treating viral, bacterial and parasitic infections, as well in boosting immune response against tumours. GITR modulation has been indicated as one of the top 25 most promising research areas by the American National Cancer Institute, and a clinical trial testing the efficacy of an anti-GITR mAb in melanoma patients has been started. In this review, we summarize results regarding: (i) the mechanisms by which GITRL/GITR system modulates immune response; (ii) the structural and functional studies clearly demonstrating differences between GITRL/GITR systems of mice and humans; (iii) the molecules with pharmacological activities including anti-GITR mAbs, GITR-Fc and GITRL-Fc fusion proteins, GITRL in monomer or multimer conformation; and (iv) the possible risks deriving from GITRL/GITR system pharmacological modulation. In conclusion, GITR triggering and inhibition could be useful in treating tumours, infectious diseases, as well as autoimmune and inflammatory diseases. However, differences between mouse and human GITRL/GITR systems suggest that further preclinical studies are needed to better understand how safe therapeutic results can be obtained and to design appropriate clinical trials.


Sujet(s)
Protéine associée au récepteur du TNF induit par les corticoïdes/antagonistes et inhibiteurs , Inhibiteurs du facteur de nécrose tumorale , Animaux , Anticorps monoclonaux/pharmacologie , Protéine associée au récepteur du TNF induit par les corticoïdes/agonistes , Protéine associée au récepteur du TNF induit par les corticoïdes/composition chimique , Protéine associée au récepteur du TNF induit par les corticoïdes/immunologie , Humains , Ligands , Souris , Modèles immunologiques , Spécificité d'espèce , Lymphocytes T/immunologie , Facteurs de nécrose tumorale/agonistes , Facteurs de nécrose tumorale/composition chimique , Facteurs de nécrose tumorale/immunologie
20.
Immunol Rev ; 244(1): 197-217, 2011 Nov.
Article de Anglais | MEDLINE | ID: mdl-22017440

RÉSUMÉ

GITR [glucocorticoid inducible tumor necrosis factor receptor (TNFR)-related protein] and 4-1BB are costimulatory TNFR family members that are expressed on regulatory and effector T cells as well as on other cells of the immune system. Here we discuss the role of GITR and 4-1BB on T cells during viral infections and in cancer immunotherapy. Systemic treatment with agonistic anti-4-1BB antibody leads to a number of immune system abnormalities, and clinical trials of anti-4-1BB have been terminated. However, other modes of 4-1BB ligation may be less toxic. To date, similar toxicities have not been reported for anti-GITR treatment of mice, although anti-GITR antibodies can exacerbate mouse autoimmune models. Intrinsic effects of GITR and 4-1BB on effector T cells appear to predominate over their effects on other cell types in some models. Despite their similarities in enhancing T-cell survival, 4-1BB and GITR are clearly not redundant, and both pathways are required for maximal CD8(+) T-cell responses and mouse survival following severe respiratory influenza infection. GITR uses TNFR-associated factor (TRAF) 2 and TRAF5, whereas 4-1BB recruits TRAF1 and TRAF2 to mediate survival signaling in T cells. The differential use of signaling adapters combined with their differential expression may explain the non-redundant roles of GITR and 4-1BB in the immune system.


Sujet(s)
Anticorps/immunologie , Protéine associée au récepteur du TNF induit par les corticoïdes/immunologie , Immunité innée , Transduction du signal/effets des médicaments et des substances chimiques , Transduction du signal/immunologie , Lymphocytes T/immunologie , Antigènes CD137/immunologie , Protéines et peptides associés aux récepteurs des facteurs de nécrose tumorale/immunologie , Animaux , Anticorps/administration et posologie , Auto-immunité/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Expression des gènes/immunologie , Protéine associée au récepteur du TNF induit par les corticoïdes/antagonistes et inhibiteurs , Protéine associée au récepteur du TNF induit par les corticoïdes/génétique , Protéine associée au récepteur du TNF induit par les corticoïdes/métabolisme , Humains , Immunité innée/effets des médicaments et des substances chimiques , Immunothérapie/méthodes , Virus de la grippe A/immunologie , Souris , Souris knockout , Tumeurs/traitement médicamenteux , Tumeurs/immunologie , Tumeurs/métabolisme , Infections à Orthomyxoviridae/immunologie , Infections à Orthomyxoviridae/virologie , Lymphocytes T/effets des médicaments et des substances chimiques , Lymphocytes T/métabolisme , Antigènes CD137/génétique , Antigènes CD137/métabolisme , Protéines et peptides associés aux récepteurs des facteurs de nécrose tumorale/génétique , Protéines et peptides associés aux récepteurs des facteurs de nécrose tumorale/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...