Network


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

Hotspot


Dive into the research topics where François Vergez is active.

Publication


Featured researches published by François Vergez.


Cancer Discovery | 2017

Chemotherapy-Resistant Human Acute Myeloid Leukemia Cells Are Not Enriched for Leukemic Stem Cells but Require Oxidative Metabolism

Thomas Farge; Estelle Saland; Fabienne De Toni; Nesrine Aroua; Moshen Hosseini; Robin Perry; Claudie Bosc; Mayumi Sugita; Lucille Stuani; Marine Fraisse; Sarah Scotland; Clément Larrue; Héléna Boutzen; Virginie Féliu; Marie-Laure Nicolau-Travers; Stephanie Cassant-Sourdy; Nicolas Broin; Marion David; Nizar Serhan; Audrey Sarry; Suzanne Tavitian; Tony Kaoma; Laurent Vallar; Jason Iacovoni; Laetitia Karine Linares; Camille Montersino; Rémy Castellano; Emmanuel Griessinger; Yves Collette; Olivier Duchamp

Chemotherapy-resistant human acute myeloid leukemia (AML) cells are thought to be enriched in quiescent immature leukemic stem cells (LSC). To validate this hypothesis in vivo, we developed a clinically relevant chemotherapeutic approach treating patient-derived xenografts (PDX) with cytarabine (AraC). AraC residual AML cells are enriched in neither immature, quiescent cells nor LSCs. Strikingly, AraC-resistant preexisting and persisting cells displayed high levels of reactive oxygen species, showed increased mitochondrial mass, and retained active polarized mitochondria, consistent with a high oxidative phosphorylation (OXPHOS) status. AraC residual cells exhibited increased fatty-acid oxidation, upregulated CD36 expression, and a high OXPHOS gene signature predictive for treatment response in PDX and patients with AML. High OXPHOS but not low OXPHOS human AML cell lines were chemoresistant in vivo. Targeting mitochondrial protein synthesis, electron transfer, or fatty-acid oxidation induced an energetic shift toward low OXPHOS and markedly enhanced antileukemic effects of AraC. Together, this study demonstrates that essential mitochondrial functions contribute to AraC resistance in AML and are a robust hallmark of AraC sensitivity and a promising therapeutic avenue to treat AML residual disease.Significance: AraC-resistant AML cells exhibit metabolic features and gene signatures consistent with a high OXPHOS status. In these cells, targeting mitochondrial metabolism through the CD36-FAO-OXPHOS axis induces an energetic shift toward low OXPHOS and strongly enhanced antileukemic effects of AraC, offering a promising avenue to design new therapeutic strategies and fight AraC resistance in AML. Cancer Discov; 7(7); 716-35. ©2017 AACR.See related commentary by Schimmer, p. 670This article is highlighted in the In This Issue feature, p. 653.


Cell Reports | 2014

The ROS/SUMO Axis Contributes to the Response of Acute Myeloid Leukemia Cells to Chemotherapeutic Drugs

Guillaume Bossis; Jean-Emmanuel Sarry; Chamseddine Kifagi; Marko Ristic; Estelle Saland; François Vergez; Tamara Salem; Héléna Boutzen; Hayeon Baik; Frédérique Brockly; Mireia Pelegrin; Tony Kaoma; Laurent Vallar; Christian Recher; Stéphane Manenti; Marc Piechaczyk

Chemotherapeutic drugs used in the treatment of acute myeloid leukemias (AMLs) are thought to induce cancer cell death through the generation of DNA double-strand breaks. Here, we report that one of their early effects is the loss of conjugation of the ubiquitin-like protein SUMO from its targets via reactive oxygen species (ROS)-dependent inhibition of the SUMO-conjugating enzymes. Desumoylation regulates the expression of specific genes, such as the proapoptotic gene DDIT3, and helps induce apoptosis in chemosensitive AMLs. In contrast, chemotherapeutics do not activate the ROS/SUMO axis in chemoresistant cells. However, pro-oxidants or inhibition of the SUMO pathway by anacardic acid restores DDIT3 expression and apoptosis in chemoresistant cell lines and patient samples, including leukemic stem cells. Finally, inhibition of the SUMO pathway decreases tumor growth in mice xenografted with AML cells. Thus, targeting the ROS/SUMO axis might constitute a therapeutic strategy for AML patients resistant to conventional chemotherapies.


Journal of Experimental Medicine | 2016

Isocitrate dehydrogenase 1 mutations prime the all-trans retinoic acid myeloid differentiation pathway in acute myeloid leukemia

Héléna Boutzen; Estelle Saland; Clément Larrue; Fabienne De Toni; Lara Gales; Florence Castelli; Mathilde Cathebas; Sonia Zaghdoudi; Lucille Stuani; Tony Kaoma; Romain Riscal; Guangli Yang; Pierre Hirsch; Marion David; Véronique De Mas-Mansat; Eric Delabesse; Laurent Vallar; François Delhommeau; Isabelle Jouanin; Ouathek Ouerfelli; Laurent Le Cam; Laetitia Karine Linares; Christophe Junot; Jean-Charles Portais; François Vergez; Christian Recher; Jean-Emmanuel Sarry

Boutzen et al. show that the IDH1 mutation and its oncometabolite, (R)-2-hydroxyglutarate, dysregulate downstream target pathways of myeloid-specific TFs, especially CEBPα, priming mutant IDH1-R132H AML blasts to the granulomonocytic lineage.


Blood Cancer Journal | 2015

A robust and rapid xenograft model to assess efficacy of chemotherapeutic agents for human acute myeloid leukemia

Estelle Saland; Helena Boutzen; Rémy Castellano; Laurent Pouyet; Emmanuel Griessinger; Clément Larrue; F. De Toni; S Scotland; Mathieu David; Gwenn-ael Danet-Desnoyers; François Vergez; Y Barreira; Yves Collette; Christian Recher; J-E Sarry

Relevant preclinical mouse models are crucial to screen new therapeutic agents for acute myeloid leukemia (AML). Current in vivo models based on the use of patient samples are not easy to establish and manipulate in the laboratory. Our objective was to develop robust xenograft models of human AML using well-characterized cell lines as a more accessible and faster alternative to those incorporating the use of patient-derived AML cells. Five widely used AML cell lines representing various AML subtypes were transplanted and expanded into highly immunodeficient non-obese diabetic/LtSz-severe combined immunodeficiency IL2Rγcnull mice (for example, cell line-derived xenografts). We show here that bone marrow sublethal conditioning with busulfan or irradiation has equal efficiency for the xenotransplantation of AML cell lines. Although higher number of injected AML cells did not change tumor engraftment in bone marrow and spleen, it significantly reduced the overall survival in mice for all tested AML cell lines. On the basis of AML cell characteristics, these models also exhibited a broad range of overall mouse survival, engraftment, tissue infiltration and aggressiveness. Thus, we have established a robust, rapid and straightforward in vivo model based on engraftment behavior of AML cell lines, all vital prerequisites for testing new therapeutic agents in preclinical studies.


Leukemia | 2013

The short form of RON is expressed in acute myeloid leukemia and sensitizes leukemic cells to cMET inhibitors.

Camille Fialin; Clément Larrue; François Vergez; Jean-Emmanuel Sarry; Sarah Bertoli; V Mansat-De Mas; Cécile Demur; Eric Delabesse; Bernard Payrastre; Stéphane Manenti; Serge Roche; Christian Recher

Several receptor tyrosine kinases (TKs) are involved in the pathogenesis of acute myeloid leukemia (AML). Here, we have assessed the expression of the Recepteur d’Origine Nantais (RON) in leukemic cell lines and samples from AML patients. In a series of 86 AML patients, we show that both the full length and/or the short form (sf) of RON are expressed in 51% and 43% of cases, respectively. Interestingly, sfRON is not expressed in normal CD34+ hematopoietic cells and induces part of its oncogenic signaling through interaction with the Src kinase Lyn. sfRON-mediated signaling in leukemic cells also involves mTORC1, the proapoptotic bcl2-family member, BAD, but not the phosphatidylinositol 3-kinase/Akt pathway. Furthermore, the expression of sfRON was specifically downregulated by 5-azacytidine (AZA). Conversely, AZA could induce the expression of sfRON in sfRON-negative leukemic cells suggesting that the activity of this drug in AML and myelodysplastic syndromes could involve modulation of TKs. cMET/RON inhibitors exhibited an antileukemic activity exclusively in AML samples and cell lines expressing sfRON. These results might support clinical trials evaluating cMET/RON inhibitors in AML patients expressing sfRON.


Molecular Cancer Therapeutics | 2015

Antileukemic Activity of 2-Deoxy-d-Glucose through Inhibition of N-Linked Glycosylation in Acute Myeloid Leukemia with FLT3-ITD or c-KIT Mutations

Clément Larrue; Estelle Saland; François Vergez; Nizar Serhan; Eric Delabesse; Véronique Mansat-De Mas; Jerome Tamburini; Stéphane Manenti; Jean Emmanuel Sarry; Christian Recher

We assessed the antileukemic activity of 2-deoxy-d-glucose (2-DG) through the modulation of expression of receptor tyrosine kinases (RTK) commonly mutated in acute myeloid leukemia (AML). We used human leukemic cell lines cells, both in vitro and in vivo, as well as leukemic samples from AML patients to demonstrate the role of 2-DG in tumor cell growth inhibition. 2-DG, through N-linked glycosylation inhibition, affected the cell-surface expression and cellular signaling of both FTL3-ITD and mutated c-KIT and induced apoptotic cell death. Leukemic cells harboring these mutated RTKs (MV4-11, MOLM-14, Kasumi-1, and TF-1 c-KIT D816V) were the most sensitive to 2-DG treatment in vitro as compared with nonmutated cells. 2-DG activity was also demonstrated in leukemic cells harboring FLT3-TKD mutations resistant to the tyrosine kinase inhibitor (TKI) quizartinib. Moreover, the antileukemic activity of 2-DG was particularly marked in c-KIT–mutated cell lines and cell samples from core binding factor–AML patients. In these cells, 2-DG inhibited the cell-surface expression of c-KIT, abrogated STAT3 and MAPK–ERK pathways, and strongly downregulated the expression of the receptor resulting in a strong in vivo effect in NOD/SCID mice xenografted with Kasumi-1 cells. Finally, we showed that 2-DG decreases Mcl-1 protein expression in AML cells and induces sensitization to both the BH3 mimetic inhibitor of Bcl-xL, Bcl-2 and Bcl-w, ABT-737, and cytarabine. In conclusion, 2-DG displays a significant antileukemic activity in AML with FLT3-ITD or KIT mutations, opening a new therapeutic window in a subset of AML with mutated RTKs. Mol Cancer Ther; 14(10); 2364–73. ©2015 AACR.


Leukemia Research | 2015

Hyperferritinemia at diagnosis predicts relapse and overall survival in younger AML patients with intermediate-risk cytogenetics.

Delphine Lebon; François Vergez; Sarah Bertoli; Veronique Harrivel; Stéphane de Botton; Jean-Baptiste Micol; Jean-Pierre Marolleau; Christian Recher

The prognostic value of ferritin level at diagnosis in AML patients is unknown. We studied 162 younger AML patients with intermediate-risk cytogenetics who received intensive chemotherapy. The median ferritin level at diagnosis was 633 μg/L and 128 (79%) patients had a ferritin level above the upper normal limit. Hyperferritinemia was significantly associated with a higher cumulative incidence of relapse as well as poorer disease-free and overall survival. In multivariate analysis, hyperferritinemia remained an independent poor prognosis factor. The level of ferritin at diagnosis has a major impact on relapse suggesting a link between inflammation, oxidative stress and chemoresistance in AML.


Leukemia | 2017

High mTORC1 activity drives glycolysis addiction and sensitivity to G6PD inhibition in acute myeloid leukemia cells

L Poulain; P Sujobert; F Zylbersztejn; S Barreau; Lucille Stuani; M Lambert; Tony Lionel Palama; V Chesnais; R Birsen; François Vergez; T Farge; C Chenevier-Gobeaux; M Fraisse; F Bouillaud; C Debeissat; O Herault; Christian Recher; C Lacombe; M Fontenay; P Mayeux; T T Maciel; J-C Portais; J-E Sarry; J Tamburini; D Bouscary; N Chapuis

Alterations in metabolic activities are cancer hallmarks that offer a wide range of new therapeutic opportunities. Here we decipher the interplay between mTORC1 activity and glucose metabolism in acute myeloid leukemia (AML). We show that mTORC1 signaling that is constantly overactivated in AML cells promotes glycolysis and leads to glucose addiction. The level of mTORC1 activity determines the sensitivity of AML cells to glycolysis inhibition as switch-off mTORC1 activity leads to glucose-independent cell survival that is sustained by an increase in mitochondrial oxidative phosphorylation. Metabolic analysis identified the pentose phosphate pathway (PPP) as an important pro-survival pathway for glucose metabolism in AML cells with high mTORC1 activity and provided a clear rational for targeting glucose-6-phosphate dehydrogenase (G6PD) in AML. Indeed, our analysis of the cancer genome atlas AML database pinpointed G6PD as a new biomarker in AML, as its overexpression correlated with an adverse prognosis in this cohort. Targeting the PPP using the G6PD inhibitor 6-aminonicotinamide induces in vitro and in vivo cytotoxicity against AML cells and synergistically sensitizes leukemic cells to chemotherapy. Our results demonstrate that high mTORC1 activity creates a specific vulnerability to G6PD inhibition that may work as a new AML therapy.


Haematologica | 2018

Dexamethasone in hyperleukocytic acute myeloid leukemia

Sarah Bertoli; Muriel Picard; Emilie Bérard; Emmanuel Griessinger; Clément Larrue; Pierre Luc Mouchel; François Vergez; Suzanne Tavitian; Edwige Yon; Jean Ruiz; Eric Delabesse; Isabelle Luquet; Laetitia Karine Linares; Estelle Saland; Martin Carroll; Gwenn Danet-Desnoyers; Audrey Sarry; Françoise Huguet; Jean Emmanuel Sarry; Christian Recher

Patients with acute myeloid leukemia and a high white blood cell count are at increased risk of early death and relapse. Because mediators of inflammation contribute to leukostasis and chemoresistance, dexamethasone added to chemotherapy could improve outcomes. This retrospective study evaluated the impact of adding or not adding dexamethasone to chemotherapy in a cohort of 160 patients with at least 50×109 white blood cells. In silico studies, primary samples, leukemic cell lines, and xenograft mouse models were used to explore the antileukemic activity of dexamethasone. There was no difference with respect to induction death rate, response, and infections between the 60 patients in the dexamethasone group and the 100 patients in the no dexamethasone group. Multivariate analysis showed that dexamethasone was significantly associated with improved relapse incidence (adjusted sub-HR: 0.30; 95% CI: 0.14–0.62; P=0.001), disease-free survival (adjusted HR: 0.50; 95% CI: 0.29–0.84; P=0.010), event-free survival (adjusted HR: 0.35; 95% CI: 0.21–0.58; P<0.001), and overall survival (adjusted HR: 0.41; 95% CI: 0.22–0.79; P=0.007). In a co-culture system, dexamethasone reduced the frequency of leukemic long-term culture initiating cells by 38% and enhanced the cytotoxicity of doxorubicin and cytarabine. In a patient-derived xenograft model treated with cytarabine, chemoresistant cells were enriched in genes of the inflammatory response modulated by dexamethasone. Dexamethasone also demonstrated antileukemic activity in NPM1-mutated samples. Dexamethasone may improve the outcome of acute myeloid leukemia patients receiving intensive chemotherapy. This effect could be due to the modulation of inflammatory chemoresistance pathways and to a specific activity in acute myeloid leukemia with NPM1 mutation.


Blood Cancer Journal | 2017

Signaling mechanisms that regulate ex vivo survival of human acute myeloid leukemia initiating cells

Dheeraj Bhavanasi; Kwun Wah Wen; Xiaolei Liu; François Vergez; Gwenn Danet-Desnoyers; Martin Carroll; Jian Huang; Peter S. Klein

Acute myeloid leukemia (AML) is an aggressive malignancy of the hematopoietic system that arises through clonal expansion of myeloid precursor cells that have arrested at an early stage of differentiation and ultimately causes death in over 50% of patients. Despite the aggressive growth characteristics of AML in vivo, AML blasts are difficult to culture once removed from the patient, suggesting that AML cells depend on signals from the microenvironment. AML initiating cells (LICs), defined functionally as cells capable of initiating AML in immunocompromised mice, are also challenging to maintain in culture, presenting a major obstacle to developing new therapies for AML that target the LIC. Co-culture with stromal cells can maintain and expand LICs; however these culture systems use multiple cytokines that may promote differentiation and loss of LICs ex vivo. Small molecules such as SR1 (an arylhydrocarbon receptor antagonist) and UM729 support AML viability without stromal co-culture, but also require multiple cytokines. In this work, we demonstrate an alternative approach that enhances the viability of primary AML cells and supports ex vivo maintenance of LICs in cytokine-free medium. We show that inhibition of GSK-3, which activates Wnt signaling, markedly improves the viability of primary human AML cells in cytokine-free medium and further that combined inhibition of GSK-3 and the nutrient sensor mTORC1 maintains LICs capable of engrafting immunocompromised mice, identifying a signaling network that regulates the viability of LICs. As GSK-3 inhibition expands hematopoietic stem and progenitor cells and Gsk3 knockout causes a myeloproliferative neoplasm suggestive of AML in mice, we tested whether direct inhibition of GSK-3 would enhance the viability of primary AML cells ex vivo. AML cells in culture do not proliferate and typically die over a few days. Cells from AML patients were cultured in defined medium without cytokines in the presence of the GSK-3 inhibitor CHIR99021 (Chiron) or vehicle. After 5 days, cells cultured with vehicle alone were small with fragmented nuclei whereas cells cultured with Chiron were uniformly larger with intact, round nuclei (Fig. 1a). The fraction of viable cells upon GSK-3 inhibition increased in a dose-dependent manner over 10 days (Fig. 1b, Supplementary Fig. 1a), with a maximal effect at 1–3 μM Chiron. The mechanistically distinct GSK-3 inhibitor, lithium chloride, also increased the fraction of viable cells compared to control at concentrations similar to the established IC50 for lithium inhibition of GSK-3 (Figs. 1c, Supplementary Fig. 1b). To address potential off-target effects with these inhibitors, we knocked down GSK3 with short interfering RNAs (siRNAs). Knockdown (30% of control for GSK3A and GSK3B, Supplementary Fig. 1c, d) of either GSK3A or GSK3B alone had no effect on viability, but combined knockdown significantly increased the viable fraction relative to control siRNA, similar to chemical GSK-3 inhibitors (Gsk3i) and consistent with redundant functions for GSK3A and GSK3B (Supplementary Fig. 1e). Thus GSK-3 inhibition improves the viability of AML cells ex vivo. To test the generality of the response to GSK-3 inhibition, primary AML blasts with distinct genetic and

Collaboration


Dive into the François Vergez's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Clément Larrue

French Institute of Health and Medical Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Françoise Huguet

Paris Descartes University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sarah Bertoli

French Institute of Health and Medical Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Laurent Vallar

University of Luxembourg

View shared research outputs
Researchain Logo
Decentralizing Knowledge