Network


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

Hotspot


Dive into the research topics where Nicolas Chomont is active.

Publication


Featured researches published by Nicolas Chomont.


Nature Medicine | 2006

Upregulation of PD-1 expression on HIV-specific CD8 + T cells leads to reversible immune dysfunction

Lydie Trautmann; Loury Janbazian; Nicolas Chomont; Elias A. Said; Sylvain Gimmig; Benoit Bessette; Mohamed Rachid Boulassel; Eric Delwart; Homero Sepulveda; Robert Balderas; Jean-Pierre Routy; Elias K. Haddad; Rafick Pierre Sekaly

The engagement of programmed death 1 (PD-1) to its ligands, PD-L1 and PD-L2, inhibits proliferation and cytokine production mediated by antibodies to CD3 (refs. 5,6,7). Blocking the PD-1–PD-L1 pathway in mice chronically infected with lymphocytic choriomeningitis virus restores the capacity of exhausted CD8+ T cells to undergo proliferation, cytokine production and cytotoxic activity and, consequently, results in reduced viral load. During chronic HIV infection, HIV-specific CD8+ T cells are functionally impaired, showing a reduced capacity to produce cytokines and effector molecules as well as an impaired capacity to proliferate. Here, we found that PD-1 was upregulated on HIV-specific CD8+ T cells; PD-1 expression levels were significantly correlated both with viral load and with the reduced capacity for cytokine production and proliferation of HIV-specific CD8+ T cells. Notably, cytomegalovirus (CMV)-specific CD8+ T cells from the same donors did not upregulate PD-1 and maintained the production of high levels of cytokines. Blocking PD-1 engagement to its ligand (PD-L1) enhanced the capacity of HIV-specific CD8+ T cells to survive and proliferate and led to an increased production of cytokines and cytotoxic molecules in response to cognate antigen. The accumulation of HIV-specific dysfunctional CD8+ T cells in the infected host could prevent the renewal of a functionally competent HIV-specific CD8+ repertoire.


Nature Medicine | 2009

HIV reservoir size and persistence are driven by T cell survival and homeostatic proliferation

Nicolas Chomont; Mohamed El-Far; Petronela Ancuta; Lydie Trautmann; Francesco A. Procopio; Bader Yassine-Diab; Geneviève Boucher; Mohamed Rachid Boulassel; Georges Ghattas; Jason M. Brenchley; Timothy W. Schacker; Brenna J. Hill; Jean-Pierre Routy; Elias K. Haddad; Rafick Pierre Sekaly

HIV persists in a reservoir of latently infected CD4+ T cells in individuals treated with highly active antiretroviral therapy (HAART). Here we identify central memory (TCM) and transitional memory (TTM) CD4+ T cells as the major cellular reservoirs for HIV and find that viral persistence is ensured by two different mechanisms. HIV primarily persists in TCM cells in subjects showing reconstitution of the CD4+ compartment upon HAART. This reservoir is maintained through T cell survival and low-level antigen-driven proliferation and is slowly depleted with time. In contrast, proviral DNA is preferentially detected in TTM cells from aviremic individuals with low CD4+ counts and higher amounts of interleukin-7–mediated homeostatic proliferation, a mechanism that ensures the persistence of these cells. Our results suggest that viral eradication might be achieved through the combined use of strategic interventions targeting viral replication and, as in cancer, drugs that interfere with the self renewal and persistence of proliferating memory T cells.


Nature Reviews Immunology | 2012

Towards an HIV cure: a global scientific strategy

Steven G. Deeks; Brigitte Autran; Ben Berkhout; Monsef Benkirane; Scott Cairns; Nicolas Chomont; Tae Wook Chun; Melissa Churchill; Michele Di Mascio; Christine Katlama; Alain Lafeuillade; Alan Landay; Michael M. Lederman; Sharon R. Lewin; Frank Maldarelli; David J. Margolis; Martin Markowitz; Javier Martinez-Picado; James I. Mullins; John W. Mellors; Santiago Moreno; Una O'Doherty; Sarah Palmer; Marie Capucine Penicaud; Matija Peterlin; Guido Poli; Jean-Pierre Routy; Christine Rouzioux; Guido Silvestri; Mario Stevenson

Given the limitations of antiretroviral therapy and recent advances in our understanding of HIV persistence during effective treatment, there is a growing recognition that a cure for HIV infection is both needed and feasible. The International AIDS Society convened a group of international experts to develop a scientific strategy for research towards an HIV cure. Several priorities for basic, translational and clinical research were identified. This Opinion article summarizes the groups recommended key goals for the international community.


Science | 2010

HIV Persistence and the Prospect of Long-Term Drug-Free Remissions for HIV-Infected Individuals

Didier Trono; Carine Van Lint; Christine Rouzioux; Eric Verdin; Françoise Barré-Sinoussi; Tae-Wook Chun; Nicolas Chomont

HIV infection can persist in spite of efficacious antiretroviral therapies. Although incomplete inhibition of viral replication may contribute to this phenomenon, this is largely due to the early establishment of a stable reservoir of latently infected cells. Thus, life-long antiviral therapy may be needed to control HIV. Such therapy is prone to drug resistance and cumulative side effects and is an unbearable financial burden for regions of the world hit hardest by the epidemic. This review discusses our current understanding of HIV persistence and the limitations of potential approaches to eradicate the virus and accordingly pleads for a joint multidisciplinary effort toward two highly related goals: the development of an HIV prophylactic vaccine and the achievement of long-term drug-free remissions in HIV-infected individuals.


PLOS Pathogens | 2014

Activation of HIV Transcription with Short-Course Vorinostat in HIV-Infected Patients on Suppressive Antiretroviral Therapy

Julian Elliott; Fiona Wightman; Ajantha Solomon; Khader Ghneim; Jeffrey D. Ahlers; Mark J. Cameron; Miranda Z. Smith; Tim Spelman; James H. McMahon; Pushparaj Velayudham; Gregor J. Brown; Janine Roney; Jo Watson; Miles Prince; Jennifer Hoy; Nicolas Chomont; Rémi Fromentin; Francesco A. Procopio; Joumana Zeidan; Sarah Palmer; Lina Odevall; Ricky W. Johnstone; Ben P. Martin; Elizabeth Sinclair; Steven G. Deeks; Daria J. Hazuda; Paul U. Cameron; Rafick-Pierre Sekaly; Sharon R. Lewin

Human immunodeficiency virus (HIV) persistence in latently infected resting memory CD4+ T-cells is the major barrier to HIV cure. Cellular histone deacetylases (HDACs) are important in maintaining HIV latency and histone deacetylase inhibitors (HDACi) may reverse latency by activating HIV transcription from latently infected CD4+ T-cells. We performed a single arm, open label, proof-of-concept study in which vorinostat, a pan-HDACi, was administered 400 mg orally once daily for 14 days to 20 HIV-infected individuals on suppressive antiretroviral therapy (ART). The primary endpoint was change in cell associated unspliced (CA-US) HIV RNA in total CD4+ T-cells from blood at day 14. The study is registered at ClinicalTrials.gov (NCT01365065). Vorinostat was safe and well tolerated and there were no dose modifications or study drug discontinuations. CA-US HIV RNA in blood increased significantly in 18/20 patients (90%) with a median fold change from baseline to peak value of 7.4 (IQR 3.4, 9.1). CA-US RNA was significantly elevated 8 hours post drug and remained elevated 70 days after last dose. Significant early changes in expression of genes associated with chromatin remodeling and activation of HIV transcription correlated with the magnitude of increased CA-US HIV RNA. There were no statistically significant changes in plasma HIV RNA, concentration of HIV DNA, integrated DNA, inducible virus in CD4+ T-cells or markers of T-cell activation. Vorinostat induced a significant and sustained increase in HIV transcription from latency in the majority of HIV-infected patients. However, additional interventions will be needed to efficiently induce virus production and ultimately eliminate latently infected cells. Trial Registration ClinicalTrials.gov NCT01365065


PLOS ONE | 2012

Impact of Multi-Targeted Antiretroviral Treatment on Gut T Cell Depletion and HIV Reservoir Seeding during Acute HIV Infection

Jintanat Ananworanich; Alexandra Schuetz; Claire Vandergeeten; Irini Sereti; Mark S. de Souza; Rungsun Rerknimitr; Robin L. Dewar; Mary Marovich; Frits van Griensven; Rafick Pierre Sekaly; Suteeraporn Pinyakorn; Nittaya Phanuphak; Rapee Trichavaroj; Wiriya Rutvisuttinunt; Nitiya Chomchey; Robert Paris; Sheila A. Peel; Victor Valcour; Frank Maldarelli; Nicolas Chomont; Nelson L. Michael; Praphan Phanuphak; Jerome H. Kim

Background Limited knowledge exists on early HIV events that may inform preventive and therapeutic strategies. This study aims to characterize the earliest immunologic and virologic HIV events following infection and investigates the usage of a novel therapeutic strategy. Methods and Findings We prospectively screened 24,430 subjects in Bangkok and identified 40 AHI individuals. Thirty Thais were enrolled (8 Fiebig I, 5 Fiebig II, 15 Fiebig III, 2 Fiebig IV) of whom 15 completed 24 weeks of megaHAART (tenofovir/emtricitabine/efavirenz/raltegravir/maraviroc). Sigmoid biopsies were completed in 24/30 at baseline and 13/15 at week 24. At baseline, the median age was 29 years and 83% were MSM. Most were symptomatic (87%), and were infected with R5-tropic (77%) CRF01_AE (70%). Median CD4 was 406 cells/mm3. HIV RNA was 5.5 log10 copies/ml. Median total blood HIV DNA was higher in Fiebig III (550 copy/106 PBMC) vs. Fiebig I (8 copy/106 PBMC) (p = 0.01) while the median %CD4+CCR5+ gut T cells was lower in Fiebig III (19%) vs. Fiebig I (59%) (p = 0.0008). After 24 weeks of megaHAART, HIV RNA levels of <50 copies were achieved in 14/15 in blood and 13/13 in gut. Total blood HIV DNA at week 0 predicted reservoir size at week 24 (p<0.001). Total HIV DNA declined significantly and was undetectable in 3 of 15 in blood and 3 of 7 in gut. Frequency of CD4+CCR5+ gut T cells increased from 41% at baseline to 64% at week 24 (p>0.050); subjects with less than 40% at baseline had a significant increase in CD4+CCR5+ T cells from baseline to week 24 (14% vs. 71%, p = 0.02). Conclusions Gut T cell depletion and HIV reservoir seeding increases with progression of AHI. MegaHAART was associated with immune restoration and reduced reservoir size. Our findings could inform research on strategies to achieve HIV drug-free remission.


Journal of Immunology | 2010

Peripheral Blood CCR4+CCR6+ and CXCR3+CCR6+ CD4+ T Cells Are Highly Permissive to HIV-1 Infection

Annie Gosselin; Patricia Monteiro; Nicolas Chomont; Felipe Diaz-Griffero; Elias A. Said; Simone Fonseca; Vanessa Sue Wacleche; Mohamed El-Far; Mohamed Rachid Boulassel; Jean-Pierre Routy; Rafick Pierre Sekaly; Petronela Ancuta

There is limited knowledge on the identity of primary CD4+ T cell subsets selectively targeted by HIV-1 in vivo. In this study, we established a link between HIV permissiveness, phenotype/homing potential, and lineage commitment in primary CD4+ T cells. CCR4+CCR6+, CCR4+CCR6−, CXCR3+CCR6+, and CXCR3+CCR6− T cells expressed cytokines and transcription factors specific for Th17, Th2, Th1Th17, and Th1 lineages, respectively. CCR4+CCR6+ and CXCR3+CCR6+ T cells expressed the HIV coreceptors CCR5 and CXCR4 and were permissive to R5 and X4 HIV replication. CCR4+CCR6− T cells expressed CXCR4 but not CCR5 and were permissive to X4 HIV only. CXCR3+CCR6− T cells expressed CCR5 and CXCR4 but were relatively resistant to R5 and X4 HIV in vitro. Total CCR6+ T cells compared with CCR6− T cells harbored higher levels of integrated HIV DNA in treatment-naive HIV-infected subjects. The frequency of total CCR6+ T cells and those of CCR4+CCR6+ and CXCR3+CCR6+ T cells were diminished in chronically infected HIV-positive subjects, despite viral-suppressive therapy. A high-throughput analysis of cytokine profiles identified CXCR3+CCR6+ T cells as a major source of TNF-α and CCL20 and demonstrated a decreased TNF-α/IL-10 ratio in CXCR3+CCR6− T cells. Finally, CCR4+CCR6+ and CXCR3+CCR6+ T cells exhibited gut- and lymph node-homing potential. Thus, we identified CCR4+CCR6+ and CXCR3+CCR6+ T cells as highly permissive to HIV replication, with potential to infiltrate and recruit more CCR6+ T cells into anatomic sites of viral replication. It is necessary that new therapeutic strategies against HIV interfere with viral replication/persistence in discrete CCR6+ T cell subsets.


PLOS Pathogens | 2015

The Depsipeptide Romidepsin Reverses HIV-1 Latency In Vivo.

Ole S. Søgaard; Mette E. Graversen; Steffen Leth; Christel R. Brinkmann; Sara K. Nissen; Anne Sofie Kjær; Mariane H. Schleimann; Paul W. Denton; William J. Hey-Cunningham; Kersten K. Koelsch; Giuseppe Pantaleo; Kim Krogsgaard; Maja A. Sommerfelt; Rémi Fromentin; Nicolas Chomont; Thomas A. Rasmussen; Lars Østergaard; Martin Tolstrup

Pharmacologically-induced activation of replication competent proviruses from latency in the presence of antiretroviral treatment (ART) has been proposed as a step towards curing HIV-1 infection. However, until now, approaches to reverse HIV-1 latency in humans have yielded mixed results. Here, we report a proof-of-concept phase Ib/IIa trial where 6 aviremic HIV-1 infected adults received intravenous 5 mg/m2 romidepsin (Celgene) once weekly for 3 weeks while maintaining ART. Lymphocyte histone H3 acetylation, a cellular measure of the pharmacodynamic response to romidepsin, increased rapidly (maximum fold range: 3.7–7.7 relative to baseline) within the first hours following each romidepsin administration. Concurrently, HIV-1 transcription quantified as copies of cell-associated un-spliced HIV-1 RNA increased significantly from baseline during treatment (range of fold-increase: 2.4–5.0; p = 0.03). Plasma HIV-1 RNA increased from <20 copies/mL at baseline to readily quantifiable levels at multiple post-infusion time-points in 5 of 6 patients (range 46–103 copies/mL following the second infusion, p = 0.04). Importantly, romidepsin did not decrease the number of HIV-specific T cells or inhibit T cell cytokine production. Adverse events (all grade 1–2) were consistent with the known side effects of romidepsin. In conclusion, romidepsin safely induced HIV-1 transcription resulting in plasma HIV-1 RNA that was readily detected with standard commercial assays demonstrating that significant reversal of HIV-1 latency in vivo is possible without blunting T cell-mediated immune responses. These finding have major implications for future trials aiming to eradicate the HIV-1 reservoir. Trial Registration clinicaltrials.gov NTC02092116


Science Translational Medicine | 2015

Virologic effects of broadly neutralizing antibody VRC01 administration during chronic HIV-1 infection

Rebecca M. Lynch; Eli Boritz; Emily E. Coates; Adam DeZure; Patrick Madden; Pamela Costner; Mary E. Enama; Sarah Plummer; LaSonji A. Holman; Cynthia S. Hendel; Ingelise J. Gordon; Joseph P. Casazza; Michelle Conan-Cibotti; Stephen A. Migueles; Randall Tressler; Robert T. Bailer; Adrian B. McDermott; Sandeep Narpala; Sijy O’Dell; Gideon Wolf; Jeffrey D. Lifson; Brandie A. Freemire; Robert J. Gorelick; Janardan P. Pandey; Sarumathi Mohan; Nicolas Chomont; Rémi Fromentin; Tae-Wook Chun; Anthony S. Fauci; Richard M. Schwartz

A single infusion with broadly neutralizing antibody VRC01 resulted in lowered plasma virus load in HIV-1–infected subjects. Passive aggression for HIV Antibodies that neutralize HIV could add to the therapeutic arsenal to prevent and treat disease. Lynch et al. have now tested one such antibody—VRC01—in HIV-infected individuals. Although little difference was observed in viral reservoir in individuals on antiretroviral therapy, plasma viremia was reduced in untreated subjects with a single infusion of VRC01, preferentially suppressing neutralization-sensitive strains. Passive immunization with neutralizing antibodies could therefore aid in viral suppression in HIV-infected individuals. Passive immunization with HIV-1–neutralizing monoclonal antibodies (mAbs) is being considered for prevention and treatment of HIV-1 infection. As therapeutic agents, mAbs could be used to suppress active virus replication, maintain suppression induced by antiretroviral therapy (ART), and/or decrease the size of the persistent virus reservoir. We assessed the impact of VRC01, a potent human mAb targeting the HIV-1 CD4 binding site, on ART-treated and untreated HIV-1–infected subjects. Among six ART-treated individuals with undetectable plasma viremia, two infusions of VRC01 did not reduce the peripheral blood cell–associated virus reservoir measured 4 weeks after the second infusion. In contrast, six of eight ART-untreated, viremic subjects infused with a single dose of VRC01 experienced a 1.1 to 1.8 log10 reduction in plasma viremia. The two subjects with minimal responses to VRC01 were found to have predominantly VRC01-resistant virus before treatment. Notably, two subjects with plasma virus load <1000 copies/ml demonstrated virus suppression to undetectable levels for over 20 days until VRC01 levels declined. Among the remaining four subjects with baseline virus loads between 3000 and 30,000 copies, viremia was only partially suppressed by mAb infusion, and we observed strong selection pressure for the outgrowth of less neutralization-sensitive viruses. In summary, a single infusion of mAb VRC01 significantly decreased plasma viremia and preferentially suppressed neutralization-sensitive virus strains. These data demonstrate the virological effect of this neutralizing antibody and highlight the need for combination strategies to maintain virus suppression.


Immunological Reviews | 2013

Immune activation and HIV persistence: implications for curative approaches to HIV infection

Nichole R. Klatt; Nicolas Chomont; Steven G. Deeks

Despite complete or near‐complete suppression of human immunodeficiency virus (HIV) replication with combination antiretroviral therapy, both HIV and chronic inflammation/immune dysfunction persist indefinitely. Untangling the association between the virus and the host immune environment during therapy might lead to novel interventions aimed at either curing the infection or preventing the development of inflammation‐associated end‐organ disease. Chronic inflammation and immune dysfunction might lead to HIV persistence by causing virus production, generating new target cells, enabling infecting of activated and resting target cells, altering the migration patterns of susceptible target cells, increasing the proliferation of infected cells, and preventing normal HIV‐specific clearance mechanisms from function. Chronic HIV production or replication might contribute to persistent inflammation and immune dysfunction. The rapidly evolving data on these issues strongly suggest that a vicious cycle might exist in which HIV persistence causes inflammation that in turn contributes to HIV persistence.

Collaboration


Dive into the Nicolas Chomont's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jean-Pierre Routy

McGill University Health Centre

View shared research outputs
Top Co-Authors

Avatar

Rafick Pierre Sekaly

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jintanat Ananworanich

Henry M. Jackson Foundation for the Advancement of Military Medicine

View shared research outputs
Top Co-Authors

Avatar

Nelson L. Michael

Walter Reed Army Institute of Research

View shared research outputs
Top Co-Authors

Avatar

Suteeraporn Pinyakorn

Walter Reed Army Institute of Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Rafick-Pierre Sekaly

Case Western Reserve University

View shared research outputs
Researchain Logo
Decentralizing Knowledge