20 results on '"Sophie Agaugue"'
Search Results
2. 563 ICT01, an anti-BTN3A mAb, and NL-201, an alpha-independent IL-2/IL-15 agonist, combine to elicit a potent anti-tumor response by synergistically stimulating Vg9Vd2 T cell activation and proliferation
- Author
-
Ryan Swanson, Aude de Gassart, Patrick Brune, Sophie Agaugué, Paul Frohna, Loui Madakamutil, Maelle Mairesse, and Carl Walkey
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2021
- Full Text
- View/download PDF
3. 528 Correlation of baseline circulating Vg9Vd2 T cell counts and pharmacodynamic activity of ICT01 in cancer patients: preliminary results from EVICTION and a novel patient enrichment strategy
- Author
-
Aude de Gassart, Daniel Olive, Clement Ghigo, Patrick Brune, Emmanuel Valentin, Sophie Agaugué, and Paul Frohna
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2021
- Full Text
- View/download PDF
4. 687 Enhancement of anti-tumor immunity by ICT01: a novel g9d2 T cell-activating antibody targeting butyrophilin-3A (BTN3A)
- Author
-
Jennifer Sims, Patrick Brune, René Hoet, Emmanuel Valentin, LE Suong, Sophie Agaugué, Remy Castellano, and Alem Truneh
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2020
- Full Text
- View/download PDF
5. 442 ICT01, an anti-BTN3A mAb that activates Vg9Vd2 T cells, plus interleukin-2: a potent and promising combination for cancer immunotherapy
- Author
-
Jennifer Sims, Patrick Brune, René Hoet, Emmanuel Valentin, LE Suong, Sophie Agaugué, and Paul Frohna
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2020
- Full Text
- View/download PDF
6. Overcoming Target Driven Fratricide for T Cell Therapy
- Author
-
Eytan Breman, Benjamin Demoulin, Sophie Agaugué, Sebastien Mauën, Alexandre Michaux, Lorraine Springuel, Julien Houssa, Fanny Huberty, Céline Jacques-Hespel, Céline Marchand, Jérôme Marijsse, Thuy Nguyen, Nancy Ramelot, Benjamin Violle, Dorothée Daro, Peter De Waele, David E. Gilham, and Valérie Steenwinckel
- Subjects
chimeric antigen receptor ,T cells ,PI3K inhibitor ,NKG2D ,CD314 ,NKG2D blocking antibody ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Chimeric Antigen Receptor (CAR) T cells expressing the fusion of the NKG2D protein with CD3ζ (NKG2D-CAR T Cells) acquire a specificity for stress-induced ligands expressed on hematological and solid cancers. However, these stress ligands are also transiently expressed by activated T cells implying that NKG2D-based T cells may undergo self-killing (fratricide) during cell manufacturing or during the freeze thaw cycle prior to infusion in patients. To avoid target-driven fratricide and enable the production of NKG2D-CAR T cells for clinical application, two distinct approaches were investigated. The first focused upon the inclusion of a Phosphoinositol-3-Kinase inhibitor (LY294002) into the production process. A second strategy involved the inclusion of antibody blockade of NKG2D itself. Both processes impacted T cell fratricide, albeit at different levels with the antibody process being the most effective in terms of cell yield. While both approaches generated comparable NKG2D-CAR T cells, there were subtle differences, for example in differentiation status, that were fine-tuned through the phasing of the inhibitor and antibody during culture in order to generate a highly potent NKG2D-CAR T cell product. By means of targeted inhibition of NKG2D expression or generic inhibition of enzyme function, target-driven CAR T fratricide can be overcome. These strategies have been incorporated into on-going clinical trials to enable a highly efficient and reproducible manufacturing process for NKG2D-CAR T cells.
- Published
- 2018
- Full Text
- View/download PDF
7. NKG2D-based chimeric antigen receptor therapy induced remission in a relapsed/refractory acute myeloid leukemia patient
- Author
-
David A. Sallman, Jason Brayer, Elizabeth M. Sagatys, Caroline Lonez, Eytan Breman, Sophie Agaugué, Bikash Verma, David E. Gilham, Frédéric F. Lehmann, and Marco L Davila
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2018
- Full Text
- View/download PDF
8. Th1 disabled function in response to TLR4 stimulation of monocyte-derived DC from patients chronically-infected by hepatitis C virus.
- Author
-
Laure Perrin-Cocon, Sophie Agaugué, Olivier Diaz, Béatrice Vanbervliet, Sandra Dollet, Aurélie Guironnet-Paquet, Patrice André, and Vincent Lotteau
- Subjects
Medicine ,Science - Abstract
BACKGROUND: Lack of protective antibodies and inefficient cytotoxic responses are characteristics of chronic hepatitis C infection. A defect in dendritic cell (DC) function has thus been suspected, but this remains a controversial issue. METHODS AND FINDINGS: Here we show that monocyte-derived DC (MoDC) from chronically-infected patients can mature in response to TLR1/2, TLR2/6 or TLR3 ligands. In contrast, when stimulated with the TLR4 ligand LPS, MoDC from patients show a profound defect in inducing IFNgamma secretion by allogeneic T cells. This defect is not due to defective phenotypic maturation or to the presence of HCV-RNA in DC or monocytes but is correlated to reduced IL-12 secretion by DC. Restoration of DC ability to stimulate IFNgamma secretion can be obtained by blocking MEK activation in DC, indicating that MEK/ERK pathway is involved in the Th1 defect of MoDC. Monocytes from HCV patients present increased spontaneous secretion of cytokines and chemokines, especially MIP-1beta. Addition of MIP-1beta on healthy monocytes during differentiation results in DC that have Th1 defect characteristic of MoDC from HCV patients, suggesting that MIP-1beta secretion by HCV monocytes participates in the Th1 defect of DC. CONCLUSIONS: Our data indicate that monocytes from HCV patients are activated in vivo. This interferes with their differentiation into DC, leading to deficient TLR4 signaling in these cells that are enable to induce a Th1 response. This specific defect is linked to the activation of the MEK/ERK pathway.
- Published
- 2008
- Full Text
- View/download PDF
9. Hepatitis C lipo-Viro-particle from chronically infected patients interferes with TLR4 signaling in dendritic cell.
- Author
-
Sophie Agaugué, Laure Perrin-Cocon, Patrice André, and Vincent Lotteau
- Subjects
Medicine ,Science - Abstract
BACKGROUND: Hepatitis C virus (HCV) can be purified from serum of chronically-infected patients in the form of Lipo-Viro-Particles (LVP), which are triglycerid-rich lipoprotein-like particles containing viral RNA and proteins. Since LVP is a constant feature of chronically infected patients, we asked whether purified LVP could interfere with the immune response by acting directly on dendritic cell (DC) function. METHODS AND FINDINGS: We have analyzed the impact of LVP on the maturation monocyte-derived DC induced by TLR3 or TLR4 ligands. Following incubation with LVP, immature DC supported weak transient HCV-RNA replication and type I IFN synthesis. This, however, did not lead to viral particle production nor to maturation of DC. LVP-treatment prior to TLR3 stimulation by polyI:C only enhanced the secretion of IL-12, IL-6 and TNFalpha yielding typical mature DC. In contrast, LVP-treated DC activated by the TLR4 ligand LPS yielded phenotypically mature DC with reduced capacity to secrete both pro- and anti-inflammatory cytokines. Their ability to stimulate allogeneic T lymphocytes was strongly affected since activated T cells produced IL-5 and IL-13 instead of IFNgamma. Addition of IFNalpha prevented the effect of LVP on DC function. Restoration of IFNgamma secretion by T cells was obtained by blocking ERK activation in DC, while induction of IL-5 and IL-13 secretion was inhibited by blocking the p38-MAPK pathway in DC. CONCLUSIONS: LVP can interfere with TLR4-triggered maturation of DC, inducing a shift in DC function that stimulates Th2 cells instead of Th1, by a mechanism that is ERK- and p38-MAPK-dependent. The effect of LVP on DC polarization was reversed by IFNalpha, providing an additional rationale for the interferon therapy of chronically-infected patients. By acting on TLR4 pathway with LVP, HCV may thus exploit a natural protective mechanism of the liver and the intestine normally used to control inflammation and immunity to commensal microorganisms.
- Published
- 2007
- Full Text
- View/download PDF
10. Exploiting natural killer group 2D receptors for CAR T-cell therapy
- Author
-
Marie-Louise Sentman, Benjamin Demoulin, David E Gilham, Caroline Lonez, Sophie Agaugue, W. James Cook, Charles L Sentman, David J. Graber, and Joana M. Murad
- Subjects
Cytotoxicity, Immunologic ,0301 basic medicine ,Cancer Research ,T-Lymphocytes ,medicine.medical_treatment ,Drug Evaluation, Preclinical ,Receptors, Antigen, T-Cell ,Biology ,Immunotherapy, Adoptive ,Natural killer cell ,03 medical and health sciences ,0302 clinical medicine ,Antigens, Neoplasm ,Neoplasms ,Pancreatic cancer ,medicine ,Extracellular ,Animals ,Humans ,Receptor ,Clinical Trials as Topic ,General Medicine ,Immunotherapy ,medicine.disease ,NKG2D ,Chimeric antigen receptor ,Treatment Outcome ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,NK Cell Lectin-Like Receptor Subfamily K ,030220 oncology & carcinogenesis ,Immunology ,Cancer research ,Intracellular - Abstract
Chimeric antigen receptors (CARs) are genetically engineered proteins that combine an extracellular antigen-specific recognition domain with one or several intracellular T-cell signaling domains. When expressed in T cells, these CARs specifically trigger T-cell activation upon antigen recognition. While the clinical proof of principle of CAR T-cell therapy has been established in hematological cancers, CAR T cells are only at the early stages of being explored to tackle solid cancers. This special report discusses the concept of exploiting natural killer cell receptors as an approach that could broaden the specificity of CAR T cells and potentially enhance the efficacy of this therapy against solid tumors. New data demonstrating feasibility of this approach in humans and supporting the ongoing clinical trial are also presented.
- Published
- 2017
- Full Text
- View/download PDF
11. NKG2D-based chimeric antigen receptor therapy induced remission in a relapsed/refractory acute myeloid leukemia patient
- Author
-
Marco L. Davila, Bikash Verma, Sophie Agaugue, Caroline Lonez, Eytan Breman, David E. Gilham, Frederic Lehmann, David A. Sallman, Jason Brayer, and Elizabeth M. Sagatys
- Subjects
0301 basic medicine ,Male ,Myeloid ,medicine.medical_treatment ,Biopsy ,T-Lymphocytes ,Receptors, Antigen, T-Cell ,Immunotherapy, Adoptive ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,Bone Marrow ,Recurrence ,medicine ,Biomarkers, Tumor ,Neoplasm ,Humans ,Online Only Articles ,Receptors, Chimeric Antigen ,business.industry ,Remission Induction ,Myeloid leukemia ,Hematology ,Immunotherapy ,Middle Aged ,medicine.disease ,Immunohistochemistry ,Chimeric antigen receptor ,Leukemia ,Leukemia, Myeloid, Acute ,030104 developmental biology ,medicine.anatomical_structure ,Treatment Outcome ,Drug Resistance, Neoplasm ,NK Cell Lectin-Like Receptor Subfamily K ,030220 oncology & carcinogenesis ,Cancer research ,business - Published
- 2018
12. The high expression of NKG2D ligands on tumor and the lack of surface expression on healthy tissues provides a strong rationale to support NKG2D-based therapeutic approaches for cancer
- Author
-
A. Hargreaves, P. De Sousa, P. De Waele, David E. Gilham, and Sophie Agaugue
- Subjects
Oncology ,Expression (architecture) ,business.industry ,Cancer research ,medicine ,Nkg2d ligands ,Cancer ,Surface expression ,Hematology ,NKG2D ,medicine.disease ,business - Published
- 2018
- Full Text
- View/download PDF
13. Abstract 3583: NKG2D as a chimeric antigen receptor - DAP 10 provides optimal co stimulation for NKG2D based CARs
- Author
-
David E. Gilham, Thuy Nguyen, Julien Houssa, Céline Jacques-Hespel, Eytan Breman, Sophie Agaugue, Jennifer Bolsée, Fanny Huberty, Nancy Ramelot, Céline Marchand, Jérôme Marijsse, and Benjamin Violle
- Subjects
Cancer Research ,Chemistry ,medicine.medical_treatment ,CD28 ,NKG2D ,Chimeric antigen receptor ,Cell biology ,Cytokine ,Oncology ,Co-stimulation ,medicine ,Cytotoxic T cell ,CD8 ,K562 cells - Abstract
Chimeric antigen receptor (CAR) protein are artificial protein created by the fusion of an extracellular domain targeting one or several cell surface antigens, a transmembrane domain and an intracellular domain responsible for signal initiation and transmission. First-generation CARs consist of an intracellular signaling domain, often a cytoplasmic tail of the CD3ζ chain. The second and third generation include the addition of one or two co-stimulation domains (e.g. cytoplasmic domain of CD28 and/or 4-1BB). NKG2D is a receptor present on the cell surface of NK cells, NK T cells and subgroups of CD8+ T cells. The receptor exists as a dimer that interacts with a co-adaptor protein called DAP10. Upon interaction of NKG2D with one of its ligand (MICA, MICB, and ULBP1-ULBP6), the DAP10 cytoplasmic tail will induce downstream signaling. DAP10 has a similar signaling motif to CD28 and similarly gets phosphorylated and recruits kinases. NKG2D ligands are expressed on a vast majority of solid and liquid tumors and are thus a very attractive target for CAR T cells. In this study, multiple NKG2D constructs were created and compared to one another. The first CAR construct tested (chNKG2D.1) consists of a fusion of full-length wild-type NKG2D and the cytoplasmic domain of CD3ζ. This construct is currently tested in clinical settings. This CAR benefits from the co-stimulatory signal provided by DAP10 and can therefore be considered as a second generation CAR. Next to this, a new chNKG2D was constructed by combining DAP10 overexpression (chNKG2D.1.DAP10). Secondly classic second-generation CARs were created by adding CD28 (chNKG2D.2.1) or 4-1BB (chNKG2D.2.2) co-stimulatory domains to the chNKG2D. Co-expression of DAP10 increased chNKG2D expression at the cell surface of both CD4 and CD8 T cells (although the increase was more pronounced in CD4 T cells). Furthermore, co-cultures of T cells expressing this construct with K562 cells led to similar IFN-γ levels irrelevant of the CD4/CD8 ratio. Other CD4 specific cytokines were highly increased when the ratio was in favor of CD4 positive T cells. Interestingly, the addition of the co-stimulatory domains to the construct (chNKG2D.2.1 and .2.2) did not lead to higher levels of IFN-γ or any of the CD4 cytokines. Next to cytokine release, cytolytic activity was also assessed which interestingly showed no difference in any of the conditions even when the CD4/CD8 cell ratio was different. In conclusion NKG2D CAR T cells only showed increased cytokine release when DAP10 was overexpressed, all other conditions did not lead to any changes when compared to the ‘NKG2D CAR T cells currently tested in the clinic. This implies that NKG2D is optimally costimulated through the DAP 10 co-signaling, and that this co-signaling is a least as potent as traditional CD28 or 4-1BB based co stimulation. Citation Format: Jennifer BOLSEE, Eytan BREMAN, Fanny HUBERTY, Benjamin VIOLLE, Jerome MARIJSSE, Céline JACQUES-HESPEL, Celine MARCHAND, Nancy RAMELOT, Thuy NGUYEN, Julien HOUSSA, David E. GILHAM, Sophie AGAUGUE. NKG2D as a chimeric antigen receptor - DAP 10 provides optimal co stimulation for NKG2D based CARs [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 3583.
- Published
- 2018
- Full Text
- View/download PDF
14. Development of an allogeneic product by CAR-T cells TCR knock-down
- Author
-
Simon Bornschein, Thuy Nguyen, Céline Jacques-Hespel, Susanna Raitano, Fanny Huberty, Céline Marchand, Jennifer Bolsée, Jérôme Marijsse, Benjamin Violle, Nancy Ramelot, Julien Houssa, David E. Gilham, and Sophie Agaugue
- Subjects
Cancer Research ,Autologous cell ,business.industry ,media_common.quotation_subject ,T-cell receptor ,Cell therapy ,Oncology ,Cancer research ,Medicine ,Quality (business) ,Product (category theory) ,Car t cells ,business ,media_common - Abstract
e15040Background: The majority of CAR-T cell therapy studies use the patients’ own autologous cells which carry many potential restrictions related to manufacturing such as quantity and quality of ...
- Published
- 2018
- Full Text
- View/download PDF
15. TCR inhibitory molecule as a promising allogeneic NKG2D CAR-t cell approach
- Author
-
Céline Jacques-Hespel, Sophie Agaugue, Nancy Ramelot, Julien Houssa, David E. Gilham, Jennifer Bolsée, Eytan Breman, Sébastien Mauen, Benjamin Violle, Céline Marchand, Alexandre Michaux, Thuy Nguyen, Jérôme Marijsse, and Fanny Huberty
- Subjects
0301 basic medicine ,Cancer Research ,Autologous cell ,business.industry ,T-cell receptor ,NKG2D ,Inhibitory postsynaptic potential ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Medicine ,CAR T-cell therapy ,Healthy donor ,Car t cells ,business - Abstract
e15042Background: Most CAR T cell therapy relies on autologous cells with potential limitation in quality of the cells. Cells derived from an allogeneic healthy donor may be more consistent in qual...
- Published
- 2018
- Full Text
- View/download PDF
16. 425. Lentiviral Vector Platforrm To Develop Anti-Cancer Immunotherapies
- Author
-
Sophie Agaugue, Déborah Revaud, Cecile Bauche, Emeline Sarry, and Ana Bejanariu
- Subjects
Pharmacology ,medicine.drug_class ,business.industry ,medicine.medical_treatment ,T cell ,Immunotherapy ,Monoclonal antibody ,Epitope ,Viral vector ,Vaccination ,medicine.anatomical_structure ,Immune system ,Antigen ,Immunology ,Drug Discovery ,medicine ,Genetics ,Molecular Medicine ,business ,Molecular Biology - Abstract
In immunotherapy of cancer, different vaccination strategies have been developed and evaluated in clinical trials, e.g. monoclonal antibodies, injection of DNA, peptides, proteins, autologous cells (tumoral cells, dendritic cells (DC) or T cells) processed in vitro or even direct injection of modified viral vectors.THERAVECTYS is a Paris-based, privately-owned, fully integrated discovery & clinical development biotech company, developping lentiviral vectors for human vaccination and immunotherapy applications. THERAVECTYS’ vaccine candidates based on a proprietary lentiviral vector technology enable a direct vaccination mechanism in patients by inducing a broad, intense and long-lasting T cell mediated response. Theravectys’ technology presents several advantages compared to these various vaccination approaches: -Our lentivectors encompasse up to 15kb DNA, the resulting polypeptide hence targeting a combination of different antigens which mobilizes different actors of the anti-tumor immune system-No selection of patients is needed according to HLA haplotype since epitopes are physiologically processed in vivo.-Unlike all other vaccination approaches, this technology allows the transduction of non-dividing cells such as DC, the most potent antigen-presenting cells (APC) of the immune system.-Break of immunotolerance: lentivectors not only provide sufficient amounts of antigens to DCs but also ensure an efficient processing and prolonged presentation of the antigens, an efficient maturation of DCs that possess all the stimulating abilities to induce an efficient activation of T cells-Non replicative and non persistent lentivectors-antigen expressing cells and transduced cells are eliminated by the immune response elicited after injection.-Direct injections in patients, without ex vivo process of autologous cells.-Different pseudotypes of lentivectors are developed to avoid specific immune reaction against the vector during boost injection(s).-Vaccination with lentiviral vectors are more potent and efficient than DNA or peptide or protein vaccination.The phase I/II clinical trial with our first vaccine candidate has been completed in HIV-infected patients. The vaccine is safe, as demonstrated by the clinical trial ongoing in 38 patients (no Serious Adverse Events have been reported during the trial). The interim analysis of the immunological data from the first two cohorts of patients performed by THERAVECTYS demonstrates the ability of the vaccine candidate to elicit multi-specific and poly-functional CD4+ and CD8+ cellular immune responses in vaccinated patients, even at the lowest dose.The interim analysis of this trial supports the potential of the lentiviral vector platform developed by THERAVECTYS for the future development of therapeutic vaccines and immunotherapies in oncology and infectious diseases. As a second indication, THERAVECTYS is developing an anti-HTLV-1 vaccine candidate to treat patients who have developed a virally-induced adult T cell leukemia. This vaccine candidate should enter into the clinics in mid-2015. Other indications in the pipeline are urogenital cancers, triple-negative breast cancer, EBV-induced nasopharyngeal cancers, HPV-induced cancers (cervix, oropharyngeal and anal cancers) and multiple myeloma.
- Published
- 2015
- Full Text
- View/download PDF
17. 224. Development of Safer & Optimized CAR-T Cells Using Lentiviral Vectors
- Author
-
Abdel Zemmar, Sophie Agaugue, Cecile Bauche, Ana Bejanariu, Emeline Sarry, and Klervi Even-Desrumeaux
- Subjects
Pharmacology ,Streptavidin ,biology ,Chemistry ,Binding protein ,Endoplasmic reticulum ,CD28 ,Virology ,CD19 ,Chimeric antigen receptor ,Viral vector ,Cell biology ,chemistry.chemical_compound ,Drug Discovery ,Genetics ,biology.protein ,Molecular Medicine ,Antibody ,human activities ,Molecular Biology - Abstract
Chimeric antigen receptors (CAR) modified T cells have shown very promising clinical results in hematological malignancies. CAR are usually composed of a scFv antigen binding domain linked to a spacer and intracellular signaling domains. CAR introduction into T cells leads to cells with optimal effector functions and with a new antigen binding ability. This strategy is well suited to skirt immune escape mechanisms.Despite their efficacy, CAR-T cells still show toxicity due to a cytokine storm, even if it is manageable. Improving the safety while maintaining or even improving the anti-tumor efficacy of CAR-T cells is therefore a crucial therapeutic challenge.Based on our proprietary lentiviral vector technology, we are developing a CAR-T cell platform aiming at producing innovative CAR-T cells. The development conducted by Theravectys are: -Development of proprietary lentiviral vectors coding for CAR of the second (containing the CD3ζ and the 4-1BB cosignaling domains) and third generations (containing the CD3ζ, the CD28 and the 4-1BB domains) directed against CD19 (for CD19+ leukemias and lymphomas), LMP-1 and -2 (for EBV-induced leukemias). Our lentiviral vectors will allow optimal expression of CAR therefore leading to a better efficacy of CAR-T cells. Hematological malignancies will be used as a benchmark.-Development of an inducible RUSH (Retention Using Selective Hook) system which is based on the streptavidin anchored to the membrane of the endoplasmic reticulum (ER) through a hook, and on the SBP (streptavidin binding protein) introduced into the CAR structure. The interaction between the streptavidin-hook and the SBP-CAR allows the CAR retention inside the ER. The addition of biotin displaces the equilibrium of binding of streptavidin towards biotin instead of SBP, thus leading to the release of the CAR from the ER and its expression to the cytoplasmic membrane. View Large Image | Download PowerPoint SlideThe release of the CAR will stop with drug exhaustion (or antagonist) and remaining cells can be easily reactivated through reintroduction of biotin. This system will increase safety of CAR-T cells. A CD19-CAR RUSH will be used as a proof-of-concept of the RUSH system.Most of the scFv used to date are of murine origin. Neutralizing antibodies against these murine scFv can limit the efficacy of CAR. As an alternative, we are screening and engineering specific humanized camelid nanobodies to be used as binding domains since they are highly homologous to the human VH domain of antibodies and they display high antigen binding capacities. The proof-of-concept will be made with a second generation CAR containing a nanobody directed against Her2 as a binding domain.Theravectys’ technological platforms allows flexibility and reactivity in the CAR design, production and evaluation, thus leading to the fast generation of safe and optimized CAR-T cells.
- Published
- 2015
- Full Text
- View/download PDF
18. Acute myeloid leukemia transcription profile based on natural killer cell status at diagnosis correlates with clinical outcome (TUM10P.1047)
- Author
-
Antoine Toubert, Zena Khaznadar, Nicolas Boissel, Sophie Agaugue, Guylaine Henry, Meyling Cheok, Marguerite Vignon, Daniela Geromin, Jean Michel Cayuela, Joel Lachuer, Claude Preudhomme, Herve Dombret, and Nicolas Dulphy
- Subjects
Immunology ,Immunology and Allergy - Abstract
Acute myeloid leukemia (AML) is a heterogeneous group of malignancies sensitive to natural killer (NK) cell anti-tumor responses. However, NK cells are frequently defective in AML. Here, we found in an exploratory cohort (n = 46) that a different expression of activating receptors (NCR) on NK-cells identified a subgroup of patients with reduced expression of DNAM-1, NKG2D, NCRs and perforin especially in CD56bright NK-cells. Patients with a deficient NK-cell profile had a significantly higher risk of relapse (P = 0.03) independently of cytogenetic classification in multivariate analysis. We hypothesized that the transcription patterns in AML cells could give a clue on the NK/AML crosstalk. Analysis of the gene expression in AML blasts following classification according to their NK-cell status showed extensive differences in immune-related and oncogenic pathways. A favorable NK status was associated with pro-inflammatory AML transcription patterns whereas patients with defective NK cells showed a profound gene expression decrease in AML blasts for cytokine and chemokine signaling, antigen processing and adhesion molecule pathways. A set of 388 leukemic classifier genes defined in the exploratory cohort was independently validated in a multicentric cohort of 194 AML patients (ALFA-0701). In total, these data evidenced the prognostic value of NK-cell responses at diagnosis of AML and the relationships between NK-cells and AML independently of known cytogenetic status.
- Published
- 2015
- Full Text
- View/download PDF
19. Multi-step defect in NK cells and acute myeloid leukemia interaction
- Author
-
Zena, Khaznadar, primary, Guylaine, Henry, primary, Niclas, Setterblad, primary, Sophie, Agaugue, primary, Emmanuel, Raffoux, primary, Nicolas, Boissel, primary, Herve, Dombret, primary, Antoine, Toubert, primary, and Nicolas, Dulphy, primary
- Published
- 2013
- Full Text
- View/download PDF
20. Major pathways involved in macrophage polarization in cancer
- Author
-
Kerneur, Clément, Cano, Carla, Olive, Daniel, Centre de Recherche en Cancérologie de Marseille (CRCM), Aix Marseille Université (AMU)-Institut Paoli-Calmettes, Fédération nationale des Centres de lutte contre le Cancer (FNCLCC)-Fédération nationale des Centres de lutte contre le Cancer (FNCLCC)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), ImCheck Therapeutics, and This work was sponsored by ImCheck Therapeutics and CRCM. We thank our colleagues from ImCheck Therapeuticsfor providing insight and knowledge that substantially helpedfor writing the review. We gratefully acknowledge, EtienneFoucher, Sophie Agaugue and Loui Madakamutil forsuggestions that significantly improved the text as well asAude De Gassart for assistance withfigure conception.
- Subjects
[SDV]Life Sciences [q-bio] ,Macrophages ,Neoplasms ,Immunology ,Tumor Microenvironment ,Immunology and Allergy ,Humans ,Macrophage Activation ,Signal Transduction - Abstract
International audience; Macrophages play an important role in tissue homeostasis, tissue remodeling, immune response, and progression of cancer. Consequently, macrophages exhibit significant plasticity and change their transcriptional profile and function in response to environmental, tissue, and inflammatory stimuli resulting in pro- and anti-tumor effects. Furthermore, the categorization of tissue macrophages in inflammatory situations remains difficult; however, there is an agreement that macrophages are predominantly polarized into two different subtypes with pro- and anti-inflammatory properties, the so-called M1-like and M2-like macrophages, respectively. These two macrophage classes can be considered as the extreme borders of a continuum of many intermediate subsets. On one end, M1 are pro-inflammatory macrophages that initiate an immunological response, damage tissue integrity, and dampen tumor progression by fostering robust T and natural killer (NK) cell anti-tumoral responses. On the other end, M2 are anti-inflammatory macrophages involved in tissue remodeling and tumor growth, that promote cancer cell proliferation, invasion, tumor metastasis, angiogenesis and that participate to immune suppression. These decisive roles in tumor progression occur through the secretion of cytokines, chemokines, growth factors, and matrix metalloproteases, as well as by the expression of immune checkpoint receptors in the case of M2 macrophages. Moreover, macrophage plasticity is supported by stimuli from the Tumor Microenvironment (TME) that are relayed to the nucleus through membrane receptors and signaling pathways that result in gene expression reprogramming in macrophages, thus giving rise to different macrophage polarization outcomes. In this review, we will focus on the main signaling pathways involved in macrophage polarization that are activated upon ligand-receptor recognition and in the presence of other immunomodulatory molecules in cancer.
- Published
- 2022
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.