Network


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

Hotspot


Dive into the research topics where Thi Tran is active.

Publication


Featured researches published by Thi Tran.


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.


Nature Communications | 2017

Induction of resident memory T cells enhances the efficacy of cancer vaccine

Mevyn Nizard; Hélène Roussel; Mariana O. Diniz; Soumaya Karaki; Thi Tran; Thibault Voron; Estelle Dransart; Federico Sandoval; Marc Riquet; Bastien Rance; Elie Marcheteau; Elizabeth Fabre; Marion Mandavit; Magali Terme; Charlotte Blanc; Jean-Baptiste Escudié; Laure Gibault; Françoise Le Pimpec Barthes; Clémence Granier; Luís Carlos de Souza Ferreira; Cécile Badoual; Ludger Johannes; Eric Tartour

Tissue-resident memory T cells (Trm) represent a new subset of long-lived memory T cells that remain in tissue and do not recirculate. Although they are considered as early immune effectors in infectious diseases, their role in cancer immunosurveillance remains unknown. In a preclinical model of head and neck cancer, we show that intranasal vaccination with a mucosal vector, the B subunit of Shiga toxin, induces local Trm and inhibits tumour growth. As Trm do not recirculate, we demonstrate their crucial role in the efficacy of cancer vaccine with parabiosis experiments. Blockade of TFGβ decreases the induction of Trm after mucosal vaccine immunization, resulting in the lower efficacy of cancer vaccine. In order to extrapolate this role of Trm in humans, we show that the number of Trm correlates with a better overall survival in lung cancer in multivariate analysis. The induction of Trm may represent a new surrogate biomarker for the efficacy of cancer vaccine. This study also argues for the development of vaccine strategies designed to elicit them.


Journal of Leukocyte Biology | 2008

Stimulation of the primary anti-HIV antibody response by IFN-α in patients with acute HIV-1 infection

Laura Adalid-Peralta; Véronique Godot; Céline Colin; Roman Krzysiek; Thi Tran; Pascal Poignard; Alain Venet; Anne Hosmalin; Pierre Lebon; Christine Rouzioux; Geneviève Chêne; Dominique Emilie

Type I IFNs are needed for the production of antiviral antibodies in mice; whether they also stimulate primary antibody responses in vivo during human viral infections is unknown. This was assessed in patients acutely infected with HIV‐1 and treated with IFN‐α2b. Patients with acute HIV‐1 infection were randomized to receive antiretroviral therapy alone (Group A, n=60) or combined for 14 weeks with pegylated‐IFN‐α2b (Group B, n=30). Emergence of anti‐HIV antibodies was monitored during 32 weeks by Western blot (WB) analyses of serum samples. IFN‐α2b treatment stimulated the production of anti‐HIV antibodies. On Week 32, 19 weeks after the last IFN‐α2b administration, there were 8.5 (6.5–10.0) HIV WB bands (median, interquartile range) in Group B and 7.0 (5.0–10.0) bands in Group A (P=0.054), and band intensities were stronger in Group B (P<0.05 for p18, p24, p34, p40, and p55 HIV antigens). IFN‐α2b treatment also increased circulating concentrations of the B cell‐activating factor of the TNF family (P<0.001) and ex vivo production of IL‐12 (P<0.05), reflecting its effect on innate immune cells. Withdrawal of antiretroviral treatment on Week 36 resulted in a lower rebound of HIV replication in Group B than in Group A (P<0.05). Therefore, type I IFNs stimulate the emerging anti‐HIV immune response in patients with acute HIV‐1 infection, resulting in an improved control of HIV replication. Type I IFNs are thus critical in the development of efficient antiviral immune responses in humans, including the production of antiviral antibodies.


Vaccine | 2016

Is There Still Room for Cancer Vaccines at the Era of Checkpoint Inhibitors

Soumaya Karaki; Marie Anson; Thi Tran; Delphine Giusti; Charlotte Blanc; Stéphane Oudard; Eric Tartour

Checkpoint inhibitor (CPI) blockade is considered to be a revolution in cancer therapy, although most patients (70%–80%) remain resistant to this therapy. It has been hypothesized that only tumors with high mutation rates generate a natural antitumor T cell response, which could be revigorated by this therapy. In patients with no pre-existing antitumor T cells, a vaccine-induced T cell response is a rational option to counteract clinical resistance. This hypothesis has been validated in preclinical models using various cancer vaccines combined with inhibitory pathway blockade (PD-1-PDL1-2, CTLA-4-CD80-CD86). Enhanced T cell infiltration of various tumors has been demonstrated following this combination therapy. The timing of this combination appears to be critical to the success of this therapy and multiple combinations of immunomodulating antibodies (CPI antagonists or costimulatory pathway agonists) have reinforced the synergy with cancer vaccines. Only limited results are available in humans and this combined approach has yet to be validated. Comprehensive monitoring of the regulation of CPI and costimulatory molecules after administration of immunomodulatory antibodies (anti-PD1/PD-L1, anti-CTLA-4, anti-OX40, etc.) and cancer vaccines should help to guide the selection of the best combination and timing of this therapy.


Journal of Immunology | 2014

Glucocorticoid-Induced Leucine Zipper Enhanced Expression in Dendritic Cells Is Sufficient To Drive Regulatory T Cells Expansion In Vivo

Joseph Calmette; Mehdi Ellouze; Thi Tran; Soumaya Karaki; Emilie Ronin; Francis Capel; Marc Pallardy; Françoise Bachelerie; Roman Krzysiek; Dominique Emilie; Géraldine Schlecht-Louf; Véronique Godot

Tolerance induction by dendritic cells (DCs) is, in part, mediated by the activation of regulatory T cells (Tregs). We have previously shown in vitro that human DCs treated with glucocorticoids (GCs), IL-10, or TGF-β upregulate the GC-Induced Leucine Zipper protein (GILZ). GILZ overexpression promotes DC differentiation into regulatory cells that generate IL-10–producing Ag-specific Tregs. To investigate whether these observations extend in vivo, we have generated CD11c-GILZhi transgenic mice. DCs from these mice constitutively overexpress GILZ to levels observed in GC-treated wild-type DCs. In this article, we establish that GILZhi DCs display an accumulation of Foxp3+ Tregs in the spleens of young CD11c-GILZhi mice. In addition, we show that GILZhi DCs strongly increase the Treg pool in central and peripheral lymphoid organs of aged animals. Upon adoptive transfer to wild-type recipient mice, OVA-loaded GILZhi bone marrow–derived DCs induce a reduced activation and proliferation of OVA-specific T cells as compared with control bone marrow–derived DCs, associated with an expansion of thymus-derived CD25+Foxp3+ CD4 T cells. Transferred OVA-loaded GILZhi DCs produce significantly higher levels of IL-10 and express reduced levels of MHC class II molecules as compared with OVA-loaded control DCs, emphasizing the regulatory phenotype of GILZhi DCs in vivo. Thus, our work demonstrates in vivo that the GILZ overexpression alone is sufficient to promote a tolerogenic mode of function in DCs.


Allergy | 2014

Enhanced glucocorticoid-induced leucine zipper in dendritic cells induces allergen-specific regulatory CD4+T-cells in respiratory allergies

Soumaya Karaki; Gilles Garcia; Colas Tcherakian; Francis Capel; Thi Tran; Marc Pallardy; Marc Humbert; Dominique Emilie; V. Godot

Respiratory allergies rely on a defect of IL‐10‐secreting regulatory CD4+T‐cells (IL‐10‐Tregs) leading to excessive Th2‐biased immune responses to allergens. According to clinical data, the restoration of allergen‐specific IL‐10‐Tregs is required to control respiratory allergies and cure patients. The discovery of mechanisms involved in the generation of IL‐10‐Tregs will thus help to provide effective treatments. We previously demonstrated that dendritic cells (DCs) expressing high levels of the glucocorticoid‐induced leucine zipper protein (GILZ) generate antigen‐specific IL‐10‐Tregs.


Journal of Hepatology | 2016

Decreased expression of the glucocorticoid receptor-GILZ pathway in Kupffer cells promotes liver inflammation in obese mice.

Olivier Robert; Hédia Boujedidi; A. Bigorgne; Gladys Ferrere; Cosmin Sebastian Voican; Sabine Vettorazzi; Jan Tuckermann; Laurence Bouchet-Delbos; Thi Tran; Patrice Hemon; Virginie Puchois; Ibrahim Dagher; Richard Douard; Françoise Gaudin; Hélène Gary-Gouy; Francis Capel; Ingrid Durand-Gasselin; Sophie Prévot; Sophie Rousset; Sylvie Naveau; V. Godot; Dominique Emilie; Marc Lombès; Gabriel Perlemuter; Anne-Marie Cassard

BACKGROUND & AIMS Kupffer cells (KC) play a key role in the onset of inflammation in non-alcoholic steatohepatitis (NASH). The glucocorticoid receptor (GR) induces glucocorticoid-induced leucine zipper (GILZ) expression in monocytes/macrophages and is involved in several inflammatory processes. We hypothesized that the GR-GILZ axis in KC may contribute to the pathophysiology of obesity-induced liver inflammation. METHODS By using a combination of primary cell culture, pharmacological experiments, mice deficient for the Gr specifically in macrophages and transgenic mice overexpressing Gilz in macrophages, we explored the involvement of the Gr-Gilz axis in KC in the pathophysiology of obesity-induced liver inflammation. RESULTS Obesity was associated with a downregulation of the Gr and Gilz, and an impairment of Gilz induction by lipopolysaccharide (LPS) and dexamethasone (DEX) in KC. Inhibition of Gilz expression in isolated KC transfected with Gilz siRNA demonstrated that Gilz downregulation was sufficient to sensitize KC to LPS. Conversely, liver inflammation was decreased in obese transgenic mice specifically overexpressing Gilz in macrophages. Pharmacological inhibition of the Gr showed that impairment of Gilz induction in KC by LPS and DEX in obesity was driven by a downregulation of the Gr. In mice specifically deficient for Gr in macrophages, Gilz expression was low, leading to an exacerbation of obesity-induced liver inflammation. CONCLUSIONS Obesity is associated with a downregulation of the Gr-Gilz axis in KC, which promotes liver inflammation. The Gr-Gilz axis in KC is an important target for the regulation of liver inflammation in obesity.


Current Gene Therapy | 2014

A suicide gene therapy combining the improvement of cyclophosphamide tumor cytotoxicity and the development of an anti-tumor immune response.

Walid Touati; Thi Tran; Johanne Seguin; Monique Diry; Jean-Pierre Flinois; Claude Baillou; Géraldine Lescaille; François André; Eric Tartour; François M. Lemoine; Philippe Beaune; Isabelle de Waziers

Gene-directed enzyme prodrug therapy (GDEPT) consists in targeted delivery to tumor cells of a suicide gene responsible for in situ conversion of a prodrug into cytotoxic metabolites. One of the major limitations of this strategy in clinical application was the poor prodrug activation capacity of suicide gene. We built a highly efficient suicide gene capable of bioactivating the prodrug cyclophosphamide (CPA) by fusing a CYP2B6 triple mutant with NADPH cytochrome P450 reductase (CYP2B6TM-RED). Expression of this fusion gene via a recombinant lentivirus (LV) vector converted resistant human (A549) and murine (TC1) pulmonary cell lines into CPA-susceptible cell lines. We tested the efficiency of our GDEPT strategy in C57Bl/6 immunocompetent mice, using TC1 cells expressing the HPV-16 E6/E7 oncoproteins. In mice bearing tumors composed only of TC1-CYP2B6TM-RED cells, four CPA injections (140 mg/Kg once a week) completely eradicated the tumors for more than two months. Tumors having only 25% of TC1-CYP2B6TM-RED cells were also completely eradicated by five CPA injections, demonstrating a major in vivo bystander effect. Moreover, surviving mice were rechallenged with parental TC1 cells. The tumors regressed spontaneously 7 days after cell inoculation or grew more slowly than in control naive mice due to a strong immune response mediated by anti-E7CD8(+)T cells. These data suggest that combining the CYPB6TM-RED gene with CPA may hold promise as a highly effective treatment for solid tumors in humans.


Seminars in Immunopathology | 2018

Therapeutic cancer vaccine: building the future from lessons of the past

Thi Tran; C. Blanc; Clémence Granier; Antonin Saldmann; C. Tanchot; Eric Tartour

Anti-cancer vaccines have raised many hopes from the start of immunotherapy but have not yet been clinically successful. The few positive results of anti-cancer vaccines have been observed in clinical situations of low tumor burden or preneoplastic lesions. Several new concepts and new results reposition this therapeutic approach in the field of immunotherapy. Indeed, cancers that respond to anti-PD-1/PD-L1 (20–30%) are those that are infiltrated by anti-tumor T cells with an inflammatory infiltrate. However, 70% of cancers do not appear to have an anti-tumor immune reaction in the tumor microenvironment. To induce this anti-tumor immunity, therapeutic combinations between vaccines and anti-PD-1/PD-L1 are being evaluated. In addition, the identification of neoepitopes against which the immune system is less tolerated is giving rise to a new enthusiasm by the first clinical results of the vaccine including these neoepitopes in humans. The ability of anti-cancer vaccines to induce a population of anti-tumor T cells called memory resident T cells that play an important role in immunosurveillance is also a new criterion to consider in the design of therapeutic vaccines.


International Journal of Cancer | 2018

Rapalog combined with CCR4 antagonist improves anticancer vaccines efficacy: Cancer vaccine combined with rapalog

Laurent Beziaud; Laura Boullerot; Thi Tran; Laura Mansi; Elodie Lauret Marie-Joseph; Patrice Ravel; Ludger Johannes; Jagadeesh Bayry; Eric Tartour; Olivier Adotevi

mTOR pathway inhibitors such as rapalogs represent a promising tool to induce functional memory CD8 T cells. In our study, we investigated the combination of temsirolimus with anticancer vaccines. Using various designs of cancer vaccines (short and long peptides or the B subunit of Shiga toxin as an antigen delivery vector) and tumor models (melanoma, lung and colon cancer), we showed that the administration of temsirolimus efficiently decreased tumor growth and enhanced tumor‐specific CD8 T‐cell responses induced by vaccination. Furthermore, tumor‐specific CD8 T cells induced by the bi‐therapy (vaccine + temsirolimus) exhibit phenotypic characteristics of central memory (CD127+CD62L+) CD8 T cells compared to vaccination alone. We demonstrated that regulatory CD4 T cells (Tregs) expansion in vivo limits the efficacy of the bi‐therapy by altering the antitumor CD8 T‐cell responses. Finally, the use of a small molecule CCR4 antagonist to prevent Tregs induction considerably improved the efficacy of the bi‐therapy by enhancing CD8 T cells‐mediated antitumor immunity. Taken together, our study highlights the potential interest of combining cancer vaccines with drugs that promote memory CD8 T cells and inhibit Tregs.

Collaboration


Dive into the Thi Tran's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Cécile Badoual

Paris Descartes University

View shared research outputs
Top Co-Authors

Avatar

Charlotte Blanc

Paris Descartes University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Federico Sandoval

Paris Descartes University

View shared research outputs
Top Co-Authors

Avatar

François M. Lemoine

French Institute of Health and Medical Research

View shared research outputs
Top Co-Authors

Avatar

Marc Pallardy

Université Paris-Saclay

View shared research outputs
Top Co-Authors

Avatar

Mevyn Nizard

Paris Descartes University

View shared research outputs
Top Co-Authors

Avatar

Philippe Beaune

Paris Descartes University

View shared research outputs
Researchain Logo
Decentralizing Knowledge