Network


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

Hotspot


Dive into the research topics where Laurent Poirot is active.

Publication


Featured researches published by Laurent Poirot.


Current Gene Therapy | 2011

Meganucleases and Other Tools for Targeted Genome Engineering: Perspectives and Challenges for Gene Therapy

George H. Silva; Laurent Poirot; Roman Galetto; Julianne Smith; Guillermo Montoya; Philippe Duchateau; Frédéric Paques

The importance of safer approaches for gene therapy has been underscored by a series of severe adverse events (SAEs) observed in patients involved in clinical trials for Severe Combined Immune Deficiency Disease (SCID) and Chromic Granulomatous Disease (CGD). While a new generation of viral vectors is in the process of replacing the classical gamma-retrovirus–based approach, a number of strategies have emerged based on non-viral vectorization and/or targeted insertion aimed at achieving safer gene transfer. Currently, these methods display lower efficacies than viral transduction although many of them can yield more than 1% engineered cells in vitro. Nuclease-based approaches, wherein an endonuclease is used to trigger site-specific genome editing, can significantly increase the percentage of targeted cells. These methods therefore provide a real alternative to classical gene transfer as well as gene editing. However, the first endonuclease to be in clinic today is not used for gene transfer, but to inactivate a gene (CCR5) required for HIV infection. Here, we review these alternative approaches, with a special emphasis on meganucleases, a family of naturally occurring rare-cutting endonucleases, and speculate on their current and future potential.


Nucleic Acids Research | 2012

Chromosomal context and epigenetic mechanisms control the efficacy of genome editing by rare-cutting designer endonucleases

Fayza Daboussi; Mikhail Zaslavskiy; Laurent Poirot; Mariana Loperfido; Agnès Gouble; Valérie Guyot; Sophie Leduc; Roman Galetto; Sylvestre Grizot; Danusia Oficjalska; Christophe Perez; Fabien Delacôte; Aurélie Dupuy; Isabelle Chion-Sotinel; Diane Le Clerre; Céline Lebuhotel; Olivier Danos; Frédéric Lemaire; Kahina Oussedik; Frédéric Cédrone; Jean-Charles Epinat; Julianne Smith; Rafael J. Yáñez-Muñoz; George Dickson; Linda Popplewell; Taeyoung Koo; Thierry Vandendriessche; Marinee K. Chuah; Aymeric Duclert; Philippe Duchateau

The ability to specifically engineer the genome of living cells at precise locations using rare-cutting designer endonucleases has broad implications for biotechnology and medicine, particularly for functional genomics, transgenics and gene therapy. However, the potential impact of chromosomal context and epigenetics on designer endonuclease-mediated genome editing is poorly understood. To address this question, we conducted a comprehensive analysis on the efficacy of 37 endonucleases derived from the quintessential I-CreI meganuclease that were specifically designed to cleave 39 different genomic targets. The analysis revealed that the efficiency of targeted mutagenesis at a given chromosomal locus is predictive of that of homologous gene targeting. Consequently, a strong genome-wide correlation was apparent between the efficiency of targeted mutagenesis (≤0.1% to ∼6%) with that of homologous gene targeting (≤0.1% to ∼15%). In contrast, the efficiency of targeted mutagenesis or homologous gene targeting at a given chromosomal locus does not correlate with the activity of individual endonucleases on transiently transfected substrates. Finally, we demonstrate that chromatin accessibility modulates the efficacy of rare-cutting endonucleases, accounting for strong position effects. Thus, chromosomal context and epigenetic mechanisms may play a major role in the efficiency rare-cutting endonuclease-induced genome engineering.


Molecular Therapy | 2015

A Multidrug-resistant Engineered CAR T Cell for Allogeneic Combination Immunotherapy

Julien Valton; Valérie Guyot; Alan Marechal; Jean-Marie Filhol; Alexandre Juillerat; Aymeric Duclert; Philippe Duchateau; Laurent Poirot

The adoptive transfer of chimeric antigen receptor (CAR) T cell represents a highly promising strategy to fight against multiple cancers. The clinical outcome of such therapies is intimately linked to the ability of effector cells to engraft, proliferate, and specifically kill tumor cells within patients. When allogeneic CAR T-cell infusion is considered, host versus graft and graft versus host reactions must be avoided to prevent rejection of adoptively transferred cells, host tissue damages and to elicit significant antitumoral outcome. This work proposes to address these three requirements through the development of multidrug-resistant T cell receptor αβ-deficient CAR T cells. We demonstrate that these engineered T cells displayed efficient antitumor activity and proliferated in the presence of purine and pyrimidine nucleoside analogues, currently used in clinic as preconditioning lymphodepleting regimens. The absence of TCRαβ at their cell surface along with their purine nucleotide analogues-resistance properties could prevent their alloreactivity and enable them to resist to lymphodepleting regimens that may be required to avoid their ablation via HvG reaction. By providing a basic framework to develop a universal T cell compatible with allogeneic adoptive transfer, this work is laying the foundation stone of the large-scale utilization of CAR T-cell immunotherapies.


Scientific Reports | 2016

Design of chimeric antigen receptors with integrated controllable transient functions

Alexandre Juillerat; Alan Marechal; Jean-Marie Filhol; Julien Valton; Aymeric Duclert; Laurent Poirot; Philippe Duchateau

The ability to control T cells engineered to permanently express chimeric antigen receptors (CARs) is a key feature to improve safety. Here, we describe the development of a new CAR architecture with an integrated switch-on system that permits to control the CAR T-cell function. This system offers the advantage of a transient CAR T-cell for safety while letting open the possibility of multiple cytotoxicity cycles using a small molecule drug.


Cancer Research | 2016

TALEN-Mediated Inactivation of PD-1 in Tumor-Reactive Lymphocytes Promotes Intratumoral T-cell Persistence and Rejection of Established Tumors

Laurie Menger; Anna Sledzinska; Katharina Bergerhoff; Frederick Arce Vargas; Julianne Smith; Laurent Poirot; Martin Pule; Javier Hererro; Karl S. Peggs; Sergio A. Quezada

Despite the promising efficacy of adoptive cell therapies (ACT) in melanoma, complete response rates remain relatively low and outcomes in other cancers are less impressive. The immunosuppressive nature of the tumor microenvironment and the expression of immune-inhibitory ligands, such as PD-L1/CD274 by the tumor and stroma are considered key factors limiting efficacy. The addition of checkpoint inhibitors (CPI) to ACT protocols bypasses some mechanisms of immunosuppression, but associated toxicities remain a significant concern. To overcome PD-L1-mediated immunosuppression and reduce CPI-associated toxicities, we used TALEN technology to render tumor-reactive T cells resistant to PD-1 signaling. Here, we demonstrate that inactivation of the PD-1 gene in melanoma-reactive CD8(+) T cells and in fibrosarcoma-reactive polyclonal T cells enhanced the persistence of PD-1 gene-modified T cells at the tumor site and increased tumor control. These results illustrate the feasibility and potency of approaches incorporating advanced gene-editing technologies into ACT protocols to silence immune checkpoints as a strategy to overcome locally active immune escape pathways. Cancer Res; 76(8); 2087-93. ©2016 AACR.


Scientific Reports | 2015

Optimized tuning of TALEN specificity using non-conventional RVDs

Alexandre Juillerat; Coline Pessereau; Gwendoline Dubois; Valérie Guyot; Alan Marechal; Julien Valton; Fayza Daboussi; Laurent Poirot; Aymeric Duclert; Philippe Duchateau

A key feature when designing DNA targeting tools and especially nucleases is specificity. The ability to control and tune this important parameter represents an invaluable advance to the development of such molecular scissors. Here, we identified and characterized new non-conventional RVDs (ncRVDs) that possess novel intrinsic targeting specificity features. We further report a strategy to control TALEN targeting based on the exclusion capacities of ncRVDs (discrimination between different nucleotides). By implementing such ncRVDs, we demonstrated in living cells the possibility to efficiently promote TALEN-mediated processing of a target in the HBB locus and alleviate undesired off-site cleavage. We anticipate that this method can greatly benefit to designer nucleases, especially for therapeutic applications and synthetic biology.


Scientific Reports | 2017

An oxygen sensitive self-decision making engineered CAR T-cell

Alexandre Juillerat; Alan Marechal; Jean Marie Filhol; Yannick Valogne; Julien Valton; Aymeric Duclert; Philippe Duchateau; Laurent Poirot

A key to the success of chimeric antigen receptor (CAR) T-cell based therapies greatly rely on the capacity to identify and target antigens with expression restrained to tumor cells. Here we present a strategy to generate CAR T-cells that are only effective locally (tumor tissue), potentially also increasing the choice of targetable antigens. By fusing an oxygen sensitive subdomain of HIF1α to a CAR scaffold, we generated CAR T-cells that are responsive to a hypoxic environment, a hallmark of certain tumors. Along with the development of oxygen-sensitive CAR T-cells, this work also provides a basic framework to use a multi-chain CAR as a platform to create the next generation of smarter self-decision making CAR T-cells.


Stem cell reports | 2015

Generation of a High Number of Healthy Erythroid Cells from Gene-Edited Pyruvate Kinase Deficiency Patient-Specific Induced Pluripotent Stem Cells

Zita Garate; Oscar Quintana-Bustamante; Ana M. Crane; Emmanuel Olivier; Laurent Poirot; Roman Galetto; Penelope Kosinski; Collin Hill; Charles Kung; Xabi Agirre; Israel Orman; Laura Cerrato; Omaira Alberquilla; Fatima Rodriguez-Fornes; Noemi Fusaki; Félix García-Sánchez; Tabita M. Maia; Ribeiro Ml; Julián Sevilla; Felipe Prosper; Shengfang Jin; Joanne C. Mountford; Guillermo Guenechea; Agnès Gouble; Juan A. Bueren; Brian R. Davis; José Segovia

Summary Pyruvate kinase deficiency (PKD) is a rare erythroid metabolic disease caused by mutations in the PKLR gene. Erythrocytes from PKD patients show an energetic imbalance causing chronic non-spherocytic hemolytic anemia, as pyruvate kinase defects impair ATP production in erythrocytes. We generated PKD induced pluripotent stem cells (PKDiPSCs) from peripheral blood mononuclear cells (PB-MNCs) of PKD patients by non-integrative Sendai viral vectors. PKDiPSCs were gene edited to integrate a partial codon-optimized R-type pyruvate kinase cDNA in the second intron of the PKLR gene by TALEN-mediated homologous recombination (HR). Notably, we found allele specificity of HR led by the presence of a single-nucleotide polymorphism. High numbers of erythroid cells derived from gene-edited PKDiPSCs showed correction of the energetic imbalance, providing an approach to correct metabolic erythroid diseases and demonstrating the practicality of this approach to generate the large cell numbers required for comprehensive biochemical and metabolic erythroid analyses.


Biotechnology and Bioengineering | 2013

Characterization of three loci for homologous gene targeting and transgene expression

Justin Eyquem; Laurent Poirot; Roman Galetto; Andrew M. Scharenberg; Julianne Smith

Integrative gene transfer is widely used for bioproduction, drug screening, and therapeutic applications but usual viral methods lead to random and multicopy insertions, contribute to unstable transgene expression and can disturb endogenous gene expression. Homologous targeting of an expression cassette using rare‐cutting endonucleases is a potential solution; however the number of studied loci remains limited. Furthermore, the behavior and performance of various types of gene cassettes following gene targeting is poorly defined. Here we have evaluated three loci for gene targeting, including one locus compatible with the proposed Safe Harbor criteria for human translational applications. Using optimized conditions for homologous gene targeting, reporter genes under the control of different promoters were efficiently inserted at each locus in both sense and antisense orientations. Sustainable expression was achieved at all three loci without detectable disturbance of flanking gene expression. However, the promoter, the integration locus and the cassette orientation have a strong impact on transgene expression. Finally, single targeted integrations exhibited greatly improved transgene expression stability versus multicopy or random integration. Taken together, our data suggest a potential set of loci for site‐specific transgene integration, suitable for a variety of biotechnological applications. Biotechnol. Bioeng. 2013; 110: 2225–2235.


Journal for ImmunoTherapy of Cancer | 2013

Treatment of B cells malignancies with anti-CD19 CAR+, TCR-, CD52- allogeneic T cells

Cécile Mannioui; Laetitia Lemaire; Laurent Poirot; Agnès Gouble; Sylvain Arnould; Roman Galetto; Julianne Smith; Andrew M. Scharenberg

Encouraging data have emerged from adoptive T-cell therapies in advanced forms of cancer. Anti-tumor immunity is found in tumor infiltrating lymphocytes as well as engineered T cells where exogenous expression of a chimeric antigen receptor (CAR) confers cancer recognition on the cells. Present adoptive immunotherapy methods are restricted to the use of autologous patient T-cells due to the limited persistence of allogeneic T cells and the potential for graft versus host disease (GvHD). The use of autologous patient T cells in cancer immunotherapy is however limited due to the fact that this approach is complex and time consuming. We propose a novel approach to treat B cell malignancies based on the use of genetically modified allogeneic T cells in conjunction with the conditioning regimen alemtuzumab. Allogeneic T cells were engineered to express an anti-CD19 CAR and to no longer express TCRalpha and CD52, responsible for GVHD and the sensitivity to alemtuzumab, respectively. The inactivation of the TCRalpha and CD52 genes in allogeneic T cells was realized by using TALEN TM ,an ovel class of sequence-specific nucleases created by the fusion of transcription activator-like effectors (TALEs) to the catalytic domain of an endonuclease. We have shown that anti-CD19 CAR+ TCR- CD52- allogeneic T cells did not respond to TCR stimulation, were resistant to alemtuzumab treatment and were able to kill target cells expressing CD19 in vitro and in vivo.

Collaboration


Dive into the Laurent Poirot's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Julien Valton

University of Copenhagen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrew M. Scharenberg

Seattle Children's Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alan Marechal

University of Copenhagen

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge