Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Magali Terme is active.

Publication


Featured researches published by Magali Terme.


Journal of Experimental Medicine | 2005

CD4+CD25+ regulatory T cells inhibit natural killer cell functions in a transforming growth factor–β–dependent manner

François Ghiringhelli; Cédric Ménard; Magali Terme; Caroline Flament; Julien Taieb; Nathalie Chaput; Pierre Emmanuel Puig; Sophie Novault; Bernard Escudier; Eric Vivier; Axel Lecesne; Caroline Robert; Jean-Yves Blay; Jacky Bernard; Sophie Caillat-Zucman; Antônio C. Freitas; Thomas Tursz; Orianne Wagner-Ballon; Claude Capron; William Vainchencker; François Martin; Laurence Zitvogel

Tumor growth promotes the expansion of CD4+CD25+ regulatory T (T reg) cells that counteract T cell–mediated immune responses. An inverse correlation between natural killer (NK) cell activation and T reg cell expansion in tumor-bearing patients, shown here, prompted us to address the role of T reg cells in controlling innate antitumor immunity. Our experiments indicate that human T reg cells expressed membrane-bound transforming growth factor (TGF)–β, which directly inhibited NK cell effector functions and down-regulated NKG2D receptors on the NK cell surface. Adoptive transfer of wild-type T reg cells but not TGF-β −/− T reg cells into nude mice suppressed NK cell–mediated cytotoxicity, reduced NKG2D receptor expression, and accelerated the growth of tumors that are normally controlled by NK cells. Conversely, the depletion of mouse T reg cells exacerbated NK cell proliferation and cytotoxicity in vivo. Human NK cell–mediated tumor recognition could also be restored by depletion of T reg cells from tumor-infiltrating lymphocytes. These findings support a role for T reg cells in blunting the NK cell arm of the innate immune system.


Nature Medicine | 2006

A novel dendritic cell subset involved in tumor immunosurveillance

Julien Taieb; Nathalie Chaput; Cédric Ménard; Lionel Apetoh; Evelyn Ullrich; Mathieu Bonmort; Marie O. Pequignot; Noelia Casares; Magali Terme; Caroline Flament; Paule Opolon; Yann Lécluse; Didier Métivier; Elena Tomasello; Eric Vivier; François Ghiringhelli; François Martin; David Klatzmann; Thierry Poynard; Thomas Tursz; Graça Raposo; Hideo Yagita; Bernard Ryffel; Guido Kroemer; Laurence Zitvogel

The interferon (IFN)-γ–induced TRAIL effector mechanism is a vital component of cancer immunosurveillance by natural killer (NK) cells in mice. Here we show that the main source of IFN-γ is not the conventional NK cell but a subset of B220+Ly6C− dendritic cells, which are atypical insofar as they express NK cell-surface molecules. Upon contact with a variety of tumor cells that are poorly recognized by NK cells, B220+NK1.1+ dendritic cells secrete high levels of IFN-γ and mediate TRAIL-dependent lysis of tumor cells. Adoptive transfer of these IFN-producing killer dendritic cells (IKDCs) into tumor-bearing Rag2−/−Il2rg−/− mice prevented tumor outgrowth, whereas transfer of conventional NK cells did not. In conclusion, we identified IKDCs as pivotal sensors and effectors of the innate antitumor immune response.


Journal of Clinical Investigation | 2004

Novel mode of action of c-kit tyrosine kinase inhibitors leading to NK cell–dependent antitumor effects

Christophe Borg; Magali Terme; Julien Taieb; Cédric Ménard; Caroline Flament; Caroline Robert; Koji Maruyama; Hiro Wakasugi; Eric Angevin; Kris Thielemans; Axel Le Cesne; Véronique Chung-Scott; Vladimir Lazar; Isabelle Tchou; Florent Crépineau; François M. Lemoine; Jacky Bernard; Jonhantan A. Fletcher; Ali G. Turhan; Jean Yves Blay; Alain Spatz; Jean-François Emile; Michael C. Heinrich; Salah Mecheri; Thomas Tursz; Laurence Zitvogel

Mutant isoforms of the KIT or PDGF receptors expressed by gastrointestinal stromal tumors (GISTs) are considered the therapeutic targets for STI571 (imatinib mesylate; Gleevec), a specific inhibitor of these tyrosine kinase receptors. Case reports of clinical efficacy of Gleevec in GISTs lacking the typical receptor mutations prompted a search for an alternate mode of action. Here we show that Gleevec can act on host DCs to promote NK cell activation. DC-mediated NK cell activation was triggered in vitro and in vivo by treatment of DCs with Gleevec as well as by a loss-of-function mutation of KIT. Therefore, tumors that are refractory to the antiproliferative effects of Gleevec in vitro responded to Gleevec in vivo in an NK cell-dependent manner. Longitudinal studies of Gleevec-treated GIST patients revealed a therapy-induced increase in IFN-gamma production by NK cells, correlating with an enhanced antitumor response. These data point to a novel mode of antitumor action for Gleevec.


Nature Immunology | 2008

Natural killer cell-directed therapies: moving from unexpected results to successful strategies.

Magali Terme; Evelyn Ullrich; Nicolas F. Delahaye; Nathalie Chaput; Laurence Zitvogel

Natural killer (NK) cells influence innate and adaptive immune host defenses. Existing data indicate that manipulating the balance between inhibitory and activating NK receptor signals, the sensitivity of target cells to NK cell-mediated apoptosis, and NK cell cross-talk with dendritic cells might hold therapeutic promise. Efforts to modulate NK cell trafficking into inflamed tissues and/or lymph nodes, and to counteract NK cell suppressors, might also prove fruitful in the clinic. However, deeper investigation into the benefits of combination therapy, greater understanding of the functional distinctions between NK cell subsets, and design of new tools to monitor NK cell activity are needed to strengthen our ability to harness the power of NK cells for therapeutic aims.


Cancer and Metastasis Reviews | 2011

Angiogenesis and immunity: a bidirectional link potentially relevant for the monitoring of antiangiogenic therapy and the development of novel therapeutic combination with immunotherapy

Eric Tartour; Hélène Péré; Bernard Maillere; Magali Terme; Nathalie Merillon; Julien Taieb; Federico Sandoval; Françoise Quintin-Colonna; K. Lacerda; A. Karadimou; Cécile Badoual; A. Tedgui; Wolf-Hervé Fridman; S. Oudard

The immune system regulates angiogenesis in cancer with both pro- and antiangiogenic activities. The induction of angiogenesis is mediated by tumor-associated macrophages and myeloid-derived suppressor cells (MDSC) which produce proinflammatory cytokines, endothelial growth factors (VEGF, bFGF…), and protease (MMP9) implicated in neoangiogenesis. Some cytokines (IL-6, IL-17…) activated Stat3 which also led to the production of VEGF and bFGF. In contrast, other cytokines (IFN, IL-12, IL-21, and IL-27) display an antiangiogenic activity. Recently, it has been shown that some antiangiogenic molecules alleviates immunosuppression associated with cancer by decreasing immunosuppressive cells (MDSC, regulatory T cells), immunosuppressive cytokines (IL-10, TGFβ), and inhibitory molecules on T cells (PD-1). Some of these broad effects may result from the ability of some antiangiogenic molecules, especially cytokines to inhibit the Stat3 transcription factor. The association often observed between angiogenesis and immunosuppression may be related to hypoxia which induces both neoangiogenesis via activation of HIF-1 and VEGF and favors the intratumor recruitment and differentiation of regulatory T cells and MDSC. Preliminary studies suggest that modulation of immune markers (intratumoral MDSC and IL-8, peripheral regulatory T cells…) may predict clinical response to antiangiogenic therapy. In preclinical models, a synergy has been observed between antiangiogenic molecules and immunotherapy which may be explained by an improvement of immune status in tumor-bearing mice after antiangiogenic therapy. In preclinical models, antiangiogenic molecules promoted intratumor trafficking of effector cells, enhance endogenous anti-tumor response, and synergyzed with immunotherapy protocols to cure established murine tumors. All these results warrant the development of clinical trials combining antiangiogenic drugs and immunotherapy.


Journal of Immunology | 2006

Chemoimmunotherapy of tumors: cyclophosphamide synergizes with exosome based vaccines.

Julien Taieb; Nathalie Chaput; Nöel E. C. Schartz; Stephan Roux; Sophie Novault; Cédric Ménard; François Ghiringhelli; Magali Terme; Antoine F. Carpentier; Guillaume Darrasse-Jèse; François A. Lemonnier; Laurence Zitvogel

Dendritic cell-derived exosomes (DEX) are nanomeric vesicles harboring MHC/peptide complexes capable of promoting primary T cell responses and tumor rejection in the presence of adjuvants. In this study, we show that, in the absence of adjuvants, DEX mediate potent Ag-dependent antitumor effects against preestablished tumors in mice pretreated with immunopotentiating dosing of cyclophosphamide. Cyclophosphamide could 1) abolish the suppressive function of CD4+CD25+Foxp3+ regulatory T cells, 2) markedly enhance the magnitude of secondary but not primary CTL responses induced by DEX vaccines, 3) synergize with DEX in therapy but not prophylaxis tumor models. Therefore, therapeutic vaccines such as DEX aimed at boosting tumor-primed effector T cells could benefit procedures that minimize the effects of CD4+CD25+ regulatory T cells.


Cancer Research | 2013

VEGFA-VEGFR Pathway Blockade Inhibits Tumor-Induced Regulatory T-cell Proliferation in Colorectal Cancer

Magali Terme; Simon Pernot; Elie Marcheteau; Federico Sandoval; Nadine Benhamouda; Orianne Colussi; Olivier Dubreuil; Antoine F. Carpentier; Eric Tartour; Julien Taieb

Multitarget antiangiogenic tyrosine kinase inhibitors (TKI) have been shown to reduce regulatory T cells (Treg) in tumor-bearing animals and patients with metastatic renal carcinomas. However, a direct role of the VEGF-A/VEGFR pathway inhibition in this phenomenon is a matter of debate and molecular mechanisms leading to Treg modulation in this setting have not been explored to date. Treg proportion, number, and proliferation were analyzed by flow cytometry in peripheral blood of patients with metastatic colorectal cancer (mCRC) treated with bevacizumab, a monoclonal antibody targeting specifically VEGF-A, and in colon cancer-bearing mice (CT26) treated with drugs targeting the VEGF/VEGFR axis. The direct impact of VEGF-A on Treg induction was assessed together with specific blockade of different isoforms of VEGFRs that may be involved. In CT26-bearing mice, anti-VEGF antibody and sunitinib treatments reduced Treg but masitinib, a TKI not targeting VEGFR, did not. Targeting VEGF-A/VEGFR axis seems sufficient to affect Treg percentages, without any changes in their function. Similarly, bevacizumab inhibited Treg accumulation in peripheral blood of patients with mCRCs. In vitro, Treg expressing VEGFR from tumor-bearing mice directly proliferated in response to VEGF-A. Anti-VEGF-A treatment decreased Treg proliferation in mice as well as in patients with mCRCs. VEGFR-2- but not VEGFR-1-specific blockade led to the same results. We identified a novel mechanism of tumor escape by which VEGF-A directly triggers Treg proliferation. This proliferation is inhibited by VEGF-A/VEGFR-2 blockade. Anti-VEGF-A therapies also have immunologic effects that may be used with a therapeutic goal in the future.


PLOS ONE | 2009

Dendritic Cell-Derived Exosomes Promote Natural Killer Cell Activation and Proliferation: A Role for NKG2D Ligands and IL-15Rα

Sophie Viaud; Magali Terme; Caroline Flament; Julien Taieb; Fabrice Andre; Sophie Novault; Bernard Escudier; Caroline Robert; Sophie Caillat-Zucman; Thomas Tursz; Laurence Zitvogel; Nathalie Chaput

Dendritic cell (DC) derived-exosomes (Dex) are nanomeric vesicles harboring functional MHC/peptide complexes promoting T cell-dependent tumor rejection. In the first Phase I trial using peptide-pulsed Dex, the observation of clinical regressions in the absence of T cell responses prompted the search for alternate effector mechanisms. Mouse studies unraveled the bioactivity of Dex on NK cells. Indeed, Dex promoted an IL-15Rα- and NKG2D-dependent NK cell proliferation and activation respectively, resulting in anti-metastatic effects mediated by NK1.1+ cells. In humans, Dex express functional IL-15Rα which allow proliferation and IFNγ secretion by NK cells. In contrast to immature DC, human Dex harbor NKG2D ligands on their surface leading to a direct engagement of NKG2D and NK cell activation ex vivo. In our phase I clinical trial, we highlight the capacity of Dex based-vaccines to restore the number and NKG2D-dependent function of NK cells in 7/14 patients. Altogether, these data provide a mechanistic explanation on how Dex may stimulate non MHC restricted-anti-tumor effectors and induce tumor regression in vivo.


Journal of Experimental Medicine | 2015

VEGF-A modulates expression of inhibitory checkpoints on CD8+ T cells in tumors

Thibault Voron; Orianne Colussi; Elie Marcheteau; Simon Pernot; Mevyn Nizard; Anne-Laure Pointet; Sabrina Latreche; Sonia Bergaya; Nadine Benhamouda; Corinne Tanchot; Christian Stockmann; Pierre Combe; Anne Berger; Franck Zinzindohoue; Hideo Yagita; Eric Tartour; Julien Taieb; Magali Terme

VEGF-A production in the tumor microenvironment enhances expression of PD-1 and other inhibitory checkpoints involved with CD8+ T cell exhaustion, which can be reversed with anti-VEGF/VEGFR treatment.


Cancer Research | 2011

IL-18 Induces PD-1–Dependent Immunosuppression in Cancer

Magali Terme; Evelyn Ullrich; Laetitia Aymeric; Kathrin Meinhardt; Mélanie Desbois; Nicolas F. Delahaye; Sophie Viaud; Bernhard Ryffel; Hideo Yagita; G. Kaplanski; Armelle Prévost-Blondel; Masashi Kato; Joachim L. Schultze; Eric Tartour; Guido Kroemer; Nathalie Chaput; Laurence Zitvogel

Immunosuppressive cytokines subvert innate and adaptive immune responses during cancer progression. The inflammatory cytokine interleukin-18 (IL-18) is known to accumulate in cancer patients, but its pathophysiological role remains unclear. In this study, we show that low levels of circulating IL-18, either exogenous or tumor derived, act to suppress the NK cell arm of tumor immunosurveillance. IL-18 produced by tumor cells promotes the development of NK-controlled metastases in a PD-1-dependent manner. Accordingly, PD-1 is expressed by activated mature NK cells in lymphoid organs of tumor bearers and is upregulated by IL-18. RNAi-mediated knockdown of IL-18 in tumors, or its systemic depletion by IL-18-binding protein, are sufficient to stimulate NK cell-dependent immunosurveillance in various tumor models. Together, these results define IL-18 as an immunosuppressive cytokine in cancer. Our findings suggest novel clinical implementations of anti-PD-1 antibodies in human malignancies that produce IL-18.

Collaboration


Dive into the Magali Terme's collaboration.

Top Co-Authors

Avatar

Julien Taieb

Paris Descartes University

View shared research outputs
Top Co-Authors

Avatar

Eric Tartour

Paris Descartes University

View shared research outputs
Top Co-Authors

Avatar

Elie Marcheteau

Paris Descartes University

View shared research outputs
Top Co-Authors

Avatar

Simon Pernot

Paris Descartes University

View shared research outputs
Top Co-Authors

Avatar

Cécile Badoual

Paris Descartes University

View shared research outputs
Top Co-Authors

Avatar

Thibault Voron

French Institute of Health and Medical Research

View shared research outputs
Top Co-Authors

Avatar

Evelyn Ullrich

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar

Federico Sandoval

Paris Descartes University

View shared research outputs
Top Co-Authors

Avatar

Eric Angevin

Institut Gustave Roussy

View shared research outputs
Top Co-Authors

Avatar

Mevyn Nizard

Paris Descartes University

View shared research outputs
Researchain Logo
Decentralizing Knowledge