Network


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

Hotspot


Dive into the research topics where Eric Tartour is active.

Publication


Featured researches published by Eric Tartour.


Oncogene | 2010

Immune infiltration in human tumors: a prognostic factor that should not be ignored

Franck Pagès; Jérôme Galon; Marie-Caroline Dieu-Nosjean; Eric Tartour; Wolf-Herman Fridman

The natural history of a tumor includes phases of ‘in situ’ growth, invasion, extravasation and metastasis. During these phases, tumor cells interact with their microenvironment and are influenced by signals coming from stromal, endothelial, inflammatory and immune cells. Indeed, tumors are often infiltrated by various numbers of lymphocytes, macrophages or mast cells. It is generally believed that the latter produce factors that maintain chronic inflammation and promote tumor growth, whereas lymphocytes may control cancer outcome, as evidenced in mouse models. In this study, we analyze data from large cohorts of human tumors, clearly establishing that infiltration of the primary tumor by memory T cells, particularly of the Th1 and cytotoxic types, is the strongest prognostic factor in terms of freedom from disease and overall survival at all stages of clinical disease. We review data suggesting that tertiary lymphoid structures adjacent to tumors and composed of mature dendritic cells (T and B cells organized as germinal centers) may be the site of an antitumor reaction. We propose an immune scoring based on the type, density and location of lymphocyte infiltrates as a novel prognostic factor for use in addition to tumor node metastasis staging to predict disease-free survival and to aid in decisions regarding adjuvant therapies in early stage human cancers.


OncoImmunology | 2012

Trial watch: Dendritic cell-based interventions for cancer therapy.

Erika Vacchelli; Ilio Vitale; Alexander Eggermont; Wolf Hervé Fridman; Jitka Fucikova; Isabelle Cremer; Jérôme Galon; Eric Tartour; Laurence Zitvogel; Guido Kroemer; Lorenzo Galluzzi

Dendritic cells (DCs) occupy a privileged position at the interface between innate and adaptive immunity, orchestrating a large panel of responses to both physiological and pathological cues. In particular, whereas the presentation of antigens by immature DCs generally results in the development of immunological tolerance, mature DCs are capable of priming robust, and hence therapeutically relevant, adaptive immune responses. In line with this notion, functional defects in the DC compartment have been shown to etiologically contribute to pathological conditions including (but perhaps not limited to) infectious diseases, allergic and autoimmune disorders, graft rejection and cancer. Thus, the possibility of harnessing the elevated immunological potential of DCs for anticancer therapy has attracted considerable interest from both researchers and clinicians over the last decade. Alongside, several methods have been developed not only to isolate DCs from cancer patients, expand them, load them with tumor-associated antigens and hence generate highly immunogenic clinical grade infusion products, but also to directly target DCs in vivo. This intense experimental effort has culminated in 2010 with the approval by the US FDA of a DC-based preparation (sipuleucel-T, Provenge®) for the treatment of asymptomatic or minimally symptomatic metastatic castration-refractory prostate cancer. As an update to the latest Trial Watch dealing with this exciting field of research (October 2012), here we summarize recent advances in DC-based anticancer regimens, covering both high-impact studies that have been published during the last 13 mo and clinical trials that have been launched in the same period to assess the antineoplastic potential of this variant of cellular immunotherapy.


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 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.


OncoImmunology | 2012

Trial watch: Monoclonal antibodies in cancer therapy

Lorenzo Galluzzi; Erika Vacchelli; Wolf Hervé Fridman; Jérôme Galon; Eric Tartour; Jessica Zucman-Rossi; Laurence Zitvogel; Guido Kroemer

Since the advent of hybridoma technology, dating back to 1975, monoclonal antibodies have become an irreplaceable diagnostic and therapeutic tool for a wide array of human diseases. During the last 15 years, several monoclonal antibodies (mAbs) have been approved by FDA for cancer therapy. These mAbs are designed to (1) activate the immune system against tumor cells, (2) inhibit cancer cell-intrinsic signaling pathways, (3) bring toxins in the close proximity of cancer cells, or (4) interfere with the tumor-stroma interaction. More recently, major efforts have been made for the development of immunostimulatory mAbs that either enhance cancer-directed immune responses or limit tumor- (or therapy-) driven immunosuppression. Some of these antibodies, which are thought to facilitate tumor eradication by initiating or sustaining a tumor-specific immune response, have already entered clinical trials. In this Trial Watch, we will review and discuss the clinical progress of the most important mAbs that are have entered clinical trials after January 2008.


Blood | 2011

A CCR4 antagonist combined with vaccines induces antigen-specific CD8+ T cells and tumor immunity against self antigens.

Hélène Péré; Yves Montier; Jagadeesh Bayry; Françoise Quintin-Colonna; Nathalie Merillon; Estelle Dransart; Cécile Badoual; Alain Gey; Patrice Ravel; Elie Marcheteau; Frédéric Batteux; Federico Sandoval; Olivier Adotevi; Christopher Chiu; Sylvie Garcia; Corinne Tanchot; Yu-Chun Lone; Luís Carlos de Souza Ferreira; Brad H. Nelson; Douglas Hanahan; Wolf H. Fridman; Ludger Johannes; Eric Tartour

Regulatory T cells (Tregs) may impede cancer vaccine efficacy in hematologic malignancies and cancer. CCR4 antagonists, an emergent class of Treg inhibitor, have been shown to block recruitment of Tregs mediated by CCL22 and CCL17. Our aim was to demonstrate the ability of a CCR4 antagonist (a small chemical molecule identified in silico) when combined with vaccines to break peripheral tolerance controlled by Tregs, a prerequisite for the induction of CD8(+) T cells against self Ags. Immunization of transgenic or normal mice expressing tumor-associated self Ags (Her2/neu, OVA, gp100) with a CCR4 antagonist combined with various vaccines led to the induction of effector CD8(+) T cells and partial inhibition of tumor growth expressing self Ags in both prophylactic and therapeutic settings. The CCR4 antagonist was more efficient than cyclophosphamide to elicit anti-self CD8(+) T cells. We also showed that the population of Tregs expressing CCR4 corresponded to memory (CD44(high)) and activated (ICOS(+)) Tregs, an important population to be targeted to modulate Treg activity. CCR4 antagonist represents a competitive class of Treg inhibitor able to induce functional anti-self CD8(+) T cells and tumor growth inhibition when combined with vaccines. High expression of CCR4 on human Tregs also supports the clinical development of this strategy.


British Journal of Cancer | 1994

Serum interleukin 6 and C-reactive protein levels correlate with resistance to IL-2 therapy and poor survival in melanoma patients.

Eric Tartour; T. Dorval; Véronique Mosseri; L. Deneux; C. Mathiot; H. Brailly; F. Montero; I. Joyeux; P Pouillart; Wolf-Herman Fridman

Interleukin 6 and C-reactive protein (CRP) were determined prior to IL-2 therapy in sera from metastatic melanoma patients. Patients with elevated serum IL-6 (> 20 pg ml-1) and/or CRP (> 10 mg l-1) levels were associated with resistance to IL-2 therapy. A correlation between high serum IL-6 levels and a shorter median survival was also observed.


Journal of Clinical Oncology | 1996

Predictors of clinical response to interleukin-2--based immunotherapy in melanoma patients: a French multiinstitutional study.

Eric Tartour; J Y Blay; T. Dorval; B Escudier; Véronique Mosseri; J Y Douillard; L Deneux; I Gorin; Sylvie Négrier; C Mathiot; P. Pouillart; Wolf-Herman Fridman

PURPOSE Various parameters have been reported to be correlated with response to interleukin-2 (IL-2) therapy. A multiinstitutional study was performed to assess by multivariate analysis the predictive value of known clinical and biologic melanoma prognostic markers recorded before the onset of IL-2 therapy on the likelihood of objective clinical response. PATIENTS AND METHODS Serum C-reactive protein (CRP), IL-6, and lactate dehydrogenase (LDH) levels were measured in 81 metastatic melanoma patients included in different IL-2-based regimens before the starting of IL-2-therapy. Clinically defined prognostic groups, i.e., patients with superficial or visceral metastases, were also analyzed for response correlates. Patients were evaluated for response to treatment 4 to 6 weeks after completion of one course of therapy. RESULTS On univariate analysis, the pretreatment values of CRP (P = .001), IL-6 (P = .007), and LDH (P = .02) and site of metastases (P = .0004) were correlated with clinical response. However, only CRP (P < .007) and clinically defined group (P < .004) were independent predictors on multifactorial analysis. Indeed, when adjusted to CRP, IL-6 tended to improve patient selection, but did not reach statistical significance (P = .07). Furthermore, using multivariate survival analysis based on the Cox proportional hazards model, only CRP was found to be an independent prognostic factor for survival (P < .0001). CONCLUSION In this study, patients with high serum levels of CRP and/or visceral organ involvement before therapy were unlikely to respond to IL-2 therapy. Therefore, clinical classification based on the site of metastases and serum CRP determination before the start of IL-2 therapy may help to improve selection of melanoma patients who may benefit from IL-2 and could prevent unnecessary morbidity.


Frontiers in Oncology | 2014

Control of the Immune Response by Pro-Angiogenic Factors

Thibault Voron; Elie Marcheteau; Simon Pernot; Orianne Colussi; Eric Tartour; Julien Taieb; Magali Terme

The progressive conversion of normal cells into cancer cells is characterized by the acquisition of eight hallmarks. Among these criteria, the capability of the cancer cell to avoid the immune destruction has been noted. Thus, tumors develop mechanisms to become invisible to the immune system, such as the induction of immunosuppressive cells, which are able to inhibit the development of an efficient immune response. Molecules produced in the tumor microenvironment are involved in the occurrence of an immunosuppressive microenvironment. Recently, it has been shown that vascular endothelial growth factor A (VEGF-A) exhibits immunosuppressive properties in addition to its pro-angiogenic activities. VEGF-A can induce the accumulation of immature dendritic cells, myeloid-derived suppressor cells, regulatory T cells, and inhibit the migration of T lymphocytes to the tumor. Other pro-angiogenic factors such as placental growth factor (PlGF) could also participate in tumor-induced immunosuppression, but only few works have been performed on this point. Here, we review the impact of pro-angiogenic factors (especially VEGF-A) on immune cells. Anti-angiogenic molecules, which target VEGF-A/VEGFR axis, have been developed in the last decades and are commonly used to treat cancer patients. These drugs have anti-angiogenic properties but can also counteract the tumor-induced immunosuppression. Based on these immunomodulatory properties, anti-angiogenic molecules could be efficiently associated with immunotherapeutic strategies in preclinical models. These combinations are currently under investigation in cancer patients.


European Journal of Immunology | 1998

Major histocompatibility complex class I presentation of exogenous soluble tumor antigen fused to the B-fragment of Shiga toxin

Ren-Shiang Lee; Eric Tartour; Pierre van der Bruggen; Valérie Vantomme; Isabelle Joyeux; Bruno Goud; Wolf H. Fridman; Ludger Johannes

Targeting exogenous antigen into the MHC class I‐restricted presentation pathway is a prerequisite for the induction of cytotoxic T lymphocytes (CTL) which have been shown to represent an important component of the protective and therapeutic immune response to viral infections and tumors. In this study, we produced recombinant proteins composed of the receptor‐binding non‐toxic B‐fragment of bacterial Shiga toxin derived from Shigella dysenteriae associated with an epitope from a model tumor antigen, Mage 1. We show that Shiga B‐Mage 1 fusion proteins carrying an active or inactive endoplasmic reticulum retrieval signal (the C‐terminal peptides KDEL or KDELGL, respectively) could be presented by peripheral blood mononuclear cells in an MHC class I‐restricted manner to Mage 1‐specific CTL. After pulsing B lymphoblastoid cells or dendritic cells with Shiga B‐Mage 1 fusion protein, activation of the MHC class I‐restricted Mage 1‐specific CTL was also demonstrated. In further analysis, we showed that treatment with brefeldin A or paraformaldehyde fixation of Epstein‐Barr virus‐transformed B cells prevented the presentation of the Mage 1 T cell epitope, which excluded extracellular processing of the antigen. Immunofluorescence analysis also revealed that the Shiga B‐Mage 1 fusion protein was largely excluded from Lamp‐2‐positive lysosomal structures. Therefore, the ability of Shiga toxin B‐fragment to target dendritic cells and B cells and to direct antigen into the exogenous class I‐restricted pathway makes it an attractive non‐living and non‐toxic vaccine vector.

Collaboration


Dive into the Eric Tartour's collaboration.

Top Co-Authors

Avatar

Cécile Badoual

Paris Descartes University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Wolf H. Fridman

Paris Descartes University

View shared research outputs
Top Co-Authors

Avatar

Magali Terme

Paris Descartes University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Clémence Granier

French Institute of Health and Medical Research

View shared research outputs
Top Co-Authors

Avatar

Hélène Péré

Paris Descartes University

View shared research outputs
Top Co-Authors

Avatar

Hélène Roussel

Paris Descartes University

View shared research outputs
Researchain Logo
Decentralizing Knowledge