Network


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

Hotspot


Dive into the research topics where Nadine Benhamouda is active.

Publication


Featured researches published by Nadine Benhamouda.


Cancer Research | 2013

PD-1–Expressing Tumor-Infiltrating T Cells Are a Favorable Prognostic Biomarker in HPV-Associated Head and Neck Cancer

Cécile Badoual; Stéphane Hans; Nathalie Merillon; Cordélia Van Ryswick; Patrice Ravel; Nadine Benhamouda; Emeline Levionnois; Mevyn Nizard; Ali Si-Mohamed; Nicolas Besnier; Alain Gey; Rinat Rotem-Yehudar; Hélène Péré; Thi Tran; Coralie L. Guerin; Anne Chauvat; Estelle Dransart; Cécile Alanio; Sebastien Albert; Beatrix Barry; Federico Sandoval; Françoise Quintin-Colonna; Patrick Bruneval; Wolf H. Fridman; François M. Lemoine; S. Oudard; Ludger Johannes; Daniel Olive; Daniel Brasnu; Eric Tartour

Head and neck cancers positive for human papillomavirus (HPV) have a more favorable clinical outcome than HPV-negative cancers, but it is unknown why this is the case. We hypothesized that prognosis was affected by intrinsic features of HPV-infected tumor cells or differences in host immune response. In this study, we focused on a comparison of regulatory Foxp3(+) T cells and programmed death-1 (PD-1)(+) T cells in the microenvironment of tumors that were positive or negative for HPV, in two groups that were matched for various clinical and biologic parameters. HPV-positive head and neck cancers were more heavily infiltrated by regulatory T cells and PD-1(+) T cells and the levels of PD-1(+) cells were positively correlated with a favorable clinical outcome. In explaining this paradoxical result, we showed that these PD-1(+) T cells expressed activation markers and were functional after blockade of the PD-1-PD-L1 axis in vitro. Approximately 50% of PD-1(+) tumor-infiltrating T cells lacked Tim-3 expression and may indeed represent activated T cells. In mice, administration of a cancer vaccine increased PD-1 on T cells with concomitant tumor regression. In this setting, PD-1 blockade synergized with vaccine in eliciting antitumor efficacy. Our findings prompt a need to revisit the significance of PD-1-infiltrating T cells in cancer, where we suggest that PD-1 detection may reflect a previous immune response against tumors that might be reactivated by PD-1/PD-L1 blockade.


European Heart Journal | 2015

Human embryonic stem cell-derived cardiac progenitors for severe heart failure treatment: first clinical case report

Philippe Menasché; Valérie Vanneaux; Albert Hagège; Alain Bel; Bernard Cholley; Isabelle Cacciapuoti; Alexandre Parouchev; Nadine Benhamouda; Gérard Tachdjian; Lucie Tosca; Jean-Hugues Trouvin; Jean-Roch Fabreguettes; Valérie Bellamy; Romain Guillemain; Caroline Suberbielle Boissel; Eric Tartour; Michel Desnos; Jerome Larghero

AIMS Comparative studies suggest that stem cells committed to a cardiac lineage are more effective for improving heart function than those featuring an extra-cardiac phenotype. We have therefore developed a population of human embryonic stem cell (ESC)-derived cardiac progenitor cells. METHODS AND RESULTS Undifferentiated human ESCs (I6 line) were amplified and cardiac-committed by exposure to bone morphogenetic protein-2 and a fibroblast growth factor receptor inhibitor. Cells responding to these cardio-instructive cues express the cardiac transcription factor Isl-1 and the stage-specific embryonic antigen SSEA-1 which was then used to purify them by immunomagnetic sorting. The Isl-1(+) SSEA-1(+) cells were then embedded into a fibrin scaffold which was surgically delivered onto the infarct area in a 68-year-old patient suffering from severe heart failure [New York Heart Association [NYHA] functional Class III; left ventricular ejection fraction (LVEF): 26%]. A coronary artery bypass was performed concomitantly in a non-infarcted area. The implanted cells featured a high degree of purity (99% were SSEA-1(+)), had lost the expression of Sox-2 and Nanog, taken as markers for pluripotency, and strongly expressed Isl-1. The intraoperative delivery of the patch was expeditious. The post-operative course was uncomplicated either. After 3 months, the patient is symptomatically improved (NYHA functional Class I; LVEF: 36%) and a new-onset contractility is echocardiographically evident in the previously akinetic cell/patch-treated, non-revascularized area. There have been no complications such as arrhythmias, tumour formation, or immunosuppression-related adverse events. CONCLUSION This observation demonstrates the feasibility of generating a clinical-grade population of human ESC-derived cardiac progenitors and combining it within a tissue-engineered construct. While any conclusion pertaining to efficacy would be meaningless, the patients functional outcome yet provides an encouraging hint. Beyond this case, the platform that has been set could be useful for generating different ESC-derived lineage-specific progenies.


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.


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.


Clinical Cancer Research | 2012

Analysis of Spontaneous Tumor-Specific CD4 T-cell Immunity in Lung Cancer Using Promiscuous HLA-DR Telomerase-Derived Epitopes: Potential Synergistic Effect with Chemotherapy Response

Yann Godet; Elizabeth Fabre; Magalie Dosset; Michele Lamuraglia; Emeline Levionnois; Patrice Ravel; Nadine Benhamouda; Aurélie Cazes; Françoise Le Pimpec-Barthes; Béatrice Gaugler; Pierre Langlade-Demoyen; Xavier Pivot; Philippe Saas; Bernard Maillere; Eric Tartour; Christophe Borg; Olivier Adotevi

Purpose: To investigate the presence and impact of spontaneous telomerase-specific CD4 T-cell responses in cancer patients. Experimental Design: A multistep approach was used to design novel pan-HLA-DR–restricted peptides from telomerase. T-cell clones isolated from cancer patients were used to characterize the polarization of telomerase-specific CD4 response. The presence of spontaneous CD4 T-cell response against telomerase was monitored in 84 metastatic non–small cell lung cancer (NSCLC) patients before first-line chemotherapy (CT) using IFN-γ ELISPOT assay. Then we analyzed the impact of the pretherapeutic telomerase-specific CD4 T immunity on clinical outcome in patients according to their respective response to CT. Results: We described four novel telomerase-derived CD4 epitopes referred as universal cancer peptides (UCP) that effectively bind to most commonly found human MHC class II alleles. UCP-specific CD4 T-cell repertoire is present in human and UCP-specific CD4 T-cell clones generated from cancer patients exhibited high avidity and are Th1 polarized. Significant frequency (38%) of naturally occurring UCP-specific T-cell responses were detected before CT in advanced NSCLC but not in healthy volunteers. This response was shown to significantly increase overall survival (OS) of patients responding to CT (Median OS: 53 vs. 40 weeks, P = 0.034). Conclusions: These results show for the first time a potential synergistic effect of telomerase-specific CD4 T-cell response with CT response in NSCLC and underline the potential role of tumor-specific CD4 T-cell response on the efficiency of conventional anticancer therapy. Clin Cancer Res; 18(10); 2943–53. ©2012 AACR.


Cancer Research | 2017

Tim-3 Expression on Tumor-Infiltrating PD-1+CD8+ T Cells Correlates with Poor Clinical Outcome in Renal Cell Carcinoma

Clémence Granier; C. Dariane; Pierre Combe; Virginie Verkarre; Saïk Urien; Cécile Badoual; Hélène Roussel; Marion Mandavit; Patrice Ravel; Mathilde Sibony; Lucie Biard; Camelia Radulescu; Emeline Vinatier; Nadine Benhamouda; Michaël Peyromaure; S. Oudard; Arnaud Mejean; Marc Olivier Timsit; Alain Gey; Eric Tartour

Inhibitory receptors expressed by T cells mediate tolerance to tumor antigens, with coexpression of these receptors exacerbating this dysfunctional state. Using the VectraR automated multiparametric immunofluorescence technique, we quantified intratumoral CD8+ T cells coexpressing the inhibitory receptors PD-1 and Tim-3 from patients with renal cell carcinoma (RCC). A second validation cohort measured the same parameters by cytometry. The percentage of tumor-infiltrating CD8+ T cells coexpressing PD-1 and Tim-3 correlated with an aggressive phenotype and a larger tumor size at diagnosis. Coexpression of PD-1 and Tim-3 above the median conferred a higher risk of relapse and a poorer 36-month overall survival. Notably, other CD8+T-cell subsets did not exert a similar effect on overall survival. Moreover, only the PD-1+Tim-3+ subset of CD8+ T cells exhibited impaired function after stimulation. Our findings establish intratumoral Tim-3+PD1+CD8+ T cells as critical mediators of an aggressive phenotype in RCC. Use of the Vectra tool may be useful to identify similarly critical prognostic and predictive biomarkers in other tumor types and their response to immunotherapy. Cancer Res; 77(5); 1075-82. ©2016 AACR.


OncoImmunology | 2013

Immunotherapy of HPV-associated head and neck cancer: Critical parameters

Mevyn Nizard; Federico Sandoval; Cécile Badoual; Hélène Péré; Magali Terme; Stéphane Hans; Nadine Benhamouda; Clémence Granier; Daniel Brasnu; Eric Tartour

Various arguments support the development of a vaccine targeting human papillomavirus (HPV) for the treatment of HPV-associated head and neck cancer. However, the mucosal localization of this tumor, the HPV-driven downregulation of MHC Class I molecules and various other immunosuppressive mechanisms must be carefully considered to improve the clinical efficacy of such an immunotherapeutic strategy.


The Journal of Thoracic and Cardiovascular Surgery | 2012

Gene polymorphisms and cytokine plasma levels as predictive factors of complications after cardiopulmonary bypass

Jérôme Jouan; Lisa Golmard; Nadine Benhamouda; Nicolas Dürrleman; Jean-Louis Golmard; Raphaël Ceccaldi; Ludovic Trinquart; Jean-Noël Fabiani; Eric Tartour; Xavier Jeunemaitre; Philippe Menasché

OBJECTIVE Cardiopulmonary bypass remains associated with significant morbidity and mortality, in part caused by a systemic inflammatory response that is unpredictable and variable among patients. Several limited studies have suggested associations of cytokine plasma levels or gene polymorphisms with outcome after cardiopulmonary bypass. The present study was to determine the relationships between several circulating cytokines and their polymorphisms (single nucleotide polymorphisms), and the occurrence of postoperative clinical events in patients who underwent coronary artery bypass grafting under cardiopulmonary bypass. METHODS Patients were genotyped for single nucleotide polymorphisms of LTA (Cys13Arg, +252A>G), TNF (-308G>A), IL6 (-597G>A, -572G>C, -174G>C), IL10 (-592C>A, c.∗117C>T), and APOE (Cys112Arg, Arg158Cys). Serum samples were collected preoperatively, immediately after cardiopulmonary bypass, and at different postoperative time points to measure cytokine serum levels by enzyme-linked immunosorbent assay. The clinical end point was the composite of postoperative death, low cardiac output syndrome, myocardial infarction, sepsis, and acute renal insufficiency. RESULTS Single nucleotide polymorphisms IL6-572GC+CC/IL10-592CC were associated with the clinical end point (P=.032 and P=.009, respectively). In addition to preoperative clinical conditions, the other factor associated with the clinical end point was interleukin-10 plasma levels 24 hours after surgery (P=.017). On the basis of these results, a predictive model of postoperative complications after coronary artery bypass grafting was created. CONCLUSIONS Our data suggest that focused genetic testing of the IL6-572G>C and IL10-592C>A single nucleotide polymorphisms might be a tool for identifying patients at the highest risk of poor tolerance to the inflammatory response to cardiopulmonary bypass and for implementing strategies to mitigate it, provided the generalization of these tests makes them reasonably affordable and thus favorably shifts their cost-to-benefit ratio.


Cells | 2015

High Reproducibility of ELISPOT Counts from Nine Different Laboratories.

Srividya Sundararaman; Alexey Y. Karulin; Tameem Ansari; Nadine Benhamouda; Judith M. Gottwein; Sreenivas Laxmanan; Steven Levine; John Loffredo; Stephanie McArdle; Christine Neudoerfl; Diana Roen; Karina Silina; Mackenzie Welch; Paul V. Lehmann

The primary goal of immune monitoring with ELISPOT is to measure the number of T cells, specific for any antigen, accurately and reproducibly between different laboratories. In ELISPOT assays, antigen-specific T cells secrete cytokines, forming spots of different sizes on a membrane with variable background intensities. Due to the subjective nature of judging maximal and minimal spot sizes, different investigators come up with different numbers. This study aims to determine whether statistics-based, automated size-gating can harmonize the number of spot counts calculated between different laboratories. We plated PBMC at four different concentrations, 24 replicates each, in an IFN-γ ELISPOT assay with HCMV pp65 antigen. The ELISPOT plate, and an image file of the plate was counted in nine different laboratories using ImmunoSpot® Analyzers by (A) Basic Count™ relying on subjective counting parameters set by the respective investigators and (B) SmartCount™, an automated counting protocol by the ImmunoSpot® Software that uses statistics-based spot size auto-gating with spot intensity auto-thresholding. The average coefficient of variation (CV) for the mean values between independent laboratories was 26.7% when counting with Basic Count™, and 6.7% when counting with SmartCount™. Our data indicates that SmartCount™ allows harmonization of counting ELISPOT results between different laboratories and investigators.


Human Vaccines & Immunotherapeutics | 2014

Clinical validation of IFNγ/IL-10 and IFNγ/IL-2 FluoroSpot assays for the detection of Tr1 T cells and influenza vaccine monitoring in humans

Anne Chauvat; Nadine Benhamouda; Alain Gey; François M. Lemoine; Staffan Paulie; Fabrice Carrat; Marie-Lise Gougeon; Flore Rozenberg; Anne Krivine; Mustapha Cherai; Paul V. Lehmann; Françoise Quintin-Colonna; Odile Launay; Eric Tartour

The type of T cell polarization and simultaneous production of multiple cytokines have been correlated with vaccine efficacy. ELISpot is a T cell detection technique optimized for the measurement of a secreted cytokine at the single cell level. The FluoroSpot assay differs from ELISpot by the use of multiple fluorescent-labeled anticytokine detection antibodies, allowing optimal measurement of multiple cytokines. In the present study, we show that an IFNγ/IL-10 FluoroSpot assay is more sensitive than flow cytometry to detect Tr1 regulatory T cells, an immunosuppressive T cell population characterized by the production of IL-10 and IFNγ. As many tolerogenic vaccines are designed to induce these Tr1 cells, this FluoroSpot test could represent a standard method for the detection of these cells in the future. The use of an IFNγ/IL-2 FluoroSpot assay during influenza vaccine monitoring showed that the influenza-specific IL-2-producing T-cell response was the dominant response both before and after vaccine administration. This study therefore questions the rationale of using the single-color IFNγ ELISpot as the standard technique to monitor vaccine-specific T-cell response. Using this same test, a trend was also observed between baseline levels of IFNγ T cell response and T cell vaccine response. In addition, a lower IFNγ+IL-2+ T-cell response after vaccine was observed in the group of patients treated with TNFα inhibitors (P = 0.08). This study therefore supports the use of the FluoroSpot assay due to its robustness, versatility and the complementary information that it provides compared with ELISpot or flow cytometry to monitor vaccine-specific T-cell responses.

Collaboration


Dive into the Nadine Benhamouda's collaboration.

Top Co-Authors

Avatar

Eric Tartour

Institut Gustave Roussy

View shared research outputs
Top Co-Authors

Avatar

Cécile Badoual

Paris Descartes University

View shared research outputs
Top Co-Authors

Avatar

Federico Sandoval

Paris Descartes University

View shared research outputs
Top Co-Authors

Avatar

Mevyn Nizard

Paris Descartes University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Patrice Ravel

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar

Anne Chauvat

Paris Descartes University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Françoise Quintin-Colonna

École nationale vétérinaire d'Alfort

View shared research outputs
Top Co-Authors

Avatar

Hélène Péré

Paris Descartes University

View shared research outputs
Researchain Logo
Decentralizing Knowledge