16 results on '"Nicolas, Gourdin"'
Search Results
2. Blocking Antibodies Targeting the CD39/CD73 Immunosuppressive Pathway Unleash Immune Responses in Combination Cancer Therapies
- Author
-
Ivan Perrot, Henri-Alexandre Michaud, Marc Giraudon-Paoli, Séverine Augier, Aurélie Docquier, Laurent Gros, Rachel Courtois, Cécile Déjou, Diana Jecko, Ondine Becquart, Hélène Rispaud-Blanc, Laurent Gauthier, Benjamin Rossi, Stéphanie Chanteux, Nicolas Gourdin, Beatrice Amigues, Alain Roussel, Armand Bensussan, Jean-François Eliaou, Jérémy Bastid, François Romagné, Yannis Morel, Emilie Narni-Mancinelli, Eric Vivier, Carine Paturel, and Nathalie Bonnefoy
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: Immune checkpoint inhibitors have revolutionized cancer treatment. However, many cancers are resistant to ICIs, and the targeting of additional inhibitory signals is crucial for limiting tumor evasion. The production of adenosine via the sequential activity of CD39 and CD73 ectoenzymes participates to the generation of an immunosuppressive tumor microenvironment. In order to disrupt the adenosine pathway, we generated two antibodies, IPH5201 and IPH5301, targeting human membrane-associated and soluble forms of CD39 and CD73, respectively, and efficiently blocking the hydrolysis of immunogenic ATP into immunosuppressive adenosine. These antibodies promoted antitumor immunity by stimulating dendritic cells and macrophages and by restoring the activation of T cells isolated from cancer patients. In a human CD39 knockin mouse preclinical model, IPH5201 increased the anti-tumor activity of the ATP-inducing chemotherapeutic drug oxaliplatin. These results support the use of anti-CD39 and anti-CD73 monoclonal antibodies and their combination with immune checkpoint inhibitors and chemotherapies in cancer. : The production of adenosine via CD39 and CD73 ectoenzymes participates in an immunosuppressive tumor microenvironment. Perrot et al. generated two antibodies, IPH5201 and IPH5301, targeting human CD39 and CD73, respectively. In vitro and in vivo data support the use of anti-CD39 and anti-CD73 mAbs in combination cancer therapies. Keywords: CD39, CD73, cancer immunotherapies, therapeutic antibodies, adenosine pathway, tumor micro-environment, immunosuppression
- Published
- 2019
- Full Text
- View/download PDF
3. Figure S1 from Autocrine Adenosine Regulates Tumor Polyfunctional CD73+CD4+ Effector T Cells Devoid of Immune Checkpoints
- Author
-
Christine Ménétrier-Caux, Christophe Caux, Pedro Romero, Jérôme Guitton, Bertrand Dubois, Julien C. Marie, Olivier Tredan, Nicolas Chopin, Isabelle Durand, Julien Faget, David Bauché, Camilla Jandus, Christelle Machon, Selena Vigano, Céline Rodriguez, Marion Bossennec, and Nicolas Gourdin
- Abstract
Th1.17 characteristics of CD73+ CD4+ Teff based on the expression of CXCR3, CCR6, CRTH2, the migration in response to CXCL10 or CCL20 gradient and the MDR-1-specific Rhodamine 123 exclusion assay
- Published
- 2023
4. Data from Autocrine Adenosine Regulates Tumor Polyfunctional CD73+CD4+ Effector T Cells Devoid of Immune Checkpoints
- Author
-
Christine Ménétrier-Caux, Christophe Caux, Pedro Romero, Jérôme Guitton, Bertrand Dubois, Julien C. Marie, Olivier Tredan, Nicolas Chopin, Isabelle Durand, Julien Faget, David Bauché, Camilla Jandus, Christelle Machon, Selena Vigano, Céline Rodriguez, Marion Bossennec, and Nicolas Gourdin
- Abstract
The production of CD73-derived adenosine (Ado) by Tregs has been proposed as a resistance mechanism to anti-PD-1 therapy in murine tumor models. We reported that human Tregs express the ectonucleotidase CD39, which generates AMP from ATP, but do not express the AMPase CD73. In contrast, CD73 defined a subset of effector CD4+ T cells (Teffs) enriched in polyfunctional Th1.17 cells characterized by expression of CXCR3, CCR6, and MDR1, and production of IL17A/IFNγ/IL22/GM-CSF. CD39+ Tregs selectively targeted CD73+ Teffs through cooperative degradation of ATP into Ado inhibiting and restricting the ability of CD73+ Teffs to secrete IL17A. CD73+ Teffs infiltrating breast and ovarian tumors were functionally blunted by Tregs expressing upregulated levels of CD39 and ATPase activity. Moreover, tumor-infiltrating CD73+ Teffs failed to express inhibitory immune checkpoints, suggesting that CD73 might be selected under pressure from immune checkpoint blockade therapy and thus may represent a nonredundant target for restoring antitumor immunity.Significance: Polyfunctional CD73+ T-cell effectors lacking other immune checkpoints are selectively targeted by CD39 overexpressing Tregs that dominate the breast tumor environment. Cancer Res; 78(13); 3604–18. ©2018 AACR.
- Published
- 2023
5. Control of acute myeloid leukemia by a trifunctional NKp46-CD16a-NK cell engager targeting CD123
- Author
-
Laurent Gauthier, Angela Virone-Oddos, Jochen Beninga, Benjamin Rossi, Céline Nicolazzi, Céline Amara, Audrey Blanchard-Alvarez, Nicolas Gourdin, Jacqueline Courta, Alexandra Basset, Magali Agnel, Franceline Guillot, Gwendoline Grondin, Hélène Bonnevaux, Anne-Laure Bauchet, Ariane Morel, Yannis Morel, Marielle Chiron, and Eric Vivier
- Subjects
Biomedical Engineering ,Molecular Medicine ,Bioengineering ,Applied Microbiology and Biotechnology ,Biotechnology - Abstract
CD123, the alpha chain of the IL-3 receptor, is an attractive target for acute myeloid leukemia (AML) treatment. However, cytotoxic antibodies or T cell engagers targeting CD123 had insufficient efficacy or safety in clinical trials. We show that expression of CD64, the high-affinity receptor for human IgG, on AML blasts confers resistance to anti-CD123 antibody-dependent cell cytotoxicity (ADCC) in vitro. We engineer a trifunctional natural killer cell engager (NKCE) that targets CD123 on AML blasts and NKp46 and CD16a on NK cells (CD123-NKCE). CD123-NKCE has potent antitumor activity against primary AML blasts regardless of CD64 expression and induces NK cell activation and cytokine secretion only in the presence of AML cells. Its antitumor activity in a mouse CD123+ tumor model exceeds that of the benchmark ADCC-enhanced antibody. In nonhuman primates, it had prolonged pharmacodynamic effects, depleting CD123+ cells for more than 10 days with no signs of toxicity and very low inflammatory cytokine induction over a large dose range. These results support clinical development of CD123-NKCE.
- Published
- 2023
6. Abstract 2960: The novel trifunctional anti-BCMA NK cell engager SAR’514 has potent in-vitro, in-vivo and ex-vivo anti-myeloma effect through dual NK cell engagement
- Author
-
Alexandre Tang, Laurent Gauthier, Jochen Beninga, Benjamin Rossi, Nicolas Gourdin, Audrey Blanchard-Alvarez, Céline Amara, Jacqueline Courta, Alexandra Basset, Dorothée Bourges, Alexandre Desjonqueres, Emmanuelle Menoret, Catherine Pellat-Deceunynck, Philippe Moreau, Yannis Morel, Marielle Chiron, Eric Vivier, and Angela Virone-Oddos
- Subjects
Cancer Research ,Oncology - Abstract
Multiple Myeloma (MM) is the second most prevalent hematopoietic malignancy, representing 10% of total blood cancers. Despite the emergence of new therapies, it is still an incurable disease. Safe, potent and innovative approaches with long lasting beneficial effects are required. BCMA (B Cell Maturation Antigen) is a cell surface receptor selectively expressed on normal and malignant plasma cells and promote cell proliferation and survival upon binding of its ligands APRIL (A PRoliferation Inducing Ligand) and BAFF (B cell Activating Factor). BCMA expression is highly prevalent on myeloma tumor cells and is maintained after standard of care treatments such as anti-CD38 therapies, or even BCMA-targeting agents (Cohen AD et al. J Clin Invest 2019). The ability of NK cells to intrinsically kill tumor cells, leaving healthy cells unharmed, with minimal pro-inflammatory cytokine release induction as compared to T cell-based therapies makes NK cells ideal immune cells for a safe and efficacious therapeutic approach. We developed SAR’514, a trifunctional NK Cell Engager (NKCE) that activates NK cells through a dual engagement of NKp46 and CD16a, two major NK cell activating receptors highly expressed on NK cells in MM patients, and which redirects the activated NK cells to engage and kill BCMA+ tumor cells. We demonstrated that SAR’514 NK dual engagement was more potent than the single NK engagement with NKp46 or CD16a as well as the combination of NKp46 and CD16a engagement. SAR’514 leads to NK cell activation, degranulation and release of effector cytokines only in the presence of BCMA+ tumor cells. This antitumor activity is associated with very low IL-1β, IL-6, TNFα and IFNγ cytokine release as compared to a T cell engager targeting the same BCMA antigen, in PBMC and in whole blood settings in the presence of BCMA+ tumor target cells. In addition, the in vivo anti-tumor activity of an anti-murine NKp46 surrogate NKCE molecule was investigated in huFcgR transgenic mice engrafted with the EL4-huBCMA murine thymoma model. SAR’514 induced a significant mouse survival at 0.5 to 5 mg/kg with an overall survival of 90% as compared to the control group in which only 20% of mice survived. Importantly, SAR’514 exhibits ex vivo efficacy using bone marrow mononuclear cells (BMMC) from MM patients in an autologous setting showing an active and efficient primary MM cell killing against MM cells from patients that have failed diverse therapies, including standard of care treatments. In summary, these results demonstrate the efficacy of SAR’514 for controlling MM tumors in vivo and ex vivo, and provide consistent support for its clinical development. Citation Format: Alexandre Tang, Laurent Gauthier, Jochen Beninga, Benjamin Rossi, Nicolas Gourdin, Audrey Blanchard-Alvarez, Céline Amara, Jacqueline Courta, Alexandra Basset, Dorothée Bourges, Alexandre Desjonqueres, Emmanuelle Menoret, Catherine Pellat-Deceunynck, Philippe Moreau, Yannis Morel, Marielle Chiron, Eric Vivier, Angela Virone-Oddos. The novel trifunctional anti-BCMA NK cell engager SAR’514 has potent in-vitro, in-vivo and ex-vivo anti-myeloma effect through dual NK cell engagement [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 2960.
- Published
- 2023
7. The Novel Trifunctional Anti-BCMA NK Cell Engager SAR'514 Has Potent in-Vitro and in-Vivo Anti-Myeloma Effect through Dual NK Cell Engagement
- Author
-
Alexandre Tang, Laurent Gauthier, Jochen Beninga, Benjamin Rossi, Nicolas Gourdin, Audrey Blanchard-Alvarez, Céline Amara, Jacqueline Courta, Alexandra Basset, Dorothee Bourges, Alexandre Desjonqueres, Emmanuelle Menoret, Catherine Pellat-Deceunynck, Philippe Moreau, Yannis Morel, Marielle Chiron, Angela Virone-Oddos, and Eric Vivier
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
8. CD73-Mediated Immunosuppression Is Linked to a Specific Fibroblast Population That Paves the Way for New Therapy in Breast Cancer
- Author
-
Yann Kieffer, Monika Licaj, Rana Mhaidly, Nicolas Gourdin, Carine Paturel, Ariane Morel, Ilaria Magagna, Fatima Mechta-Grigoriou, Anne Vincent-Salomon, and Pascale Andre
- Subjects
Cancer Research ,medicine.medical_treatment ,Population ,Tregs ,Article ,NT5E ,Immune system ,Breast cancer ,breast cancer ,Stroma ,medicine ,education ,RC254-282 ,education.field_of_study ,immunosuppression ,biology ,anti-CD73 antibody ,business.industry ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Immunosuppression ,Immunotherapy ,medicine.disease ,CAF subsets ,Oncology ,Cancer research ,biology.protein ,Antibody ,business - Abstract
Simple Summary Recent findings have revealed the contribution of cancer-associated fibroblasts (CAF) in immune escape in breast cancer. Still, how to specifically target immunosuppressive CAF remains an unmet medical question. Here, we provide a promising therapeutic strategy by highlighting the role of CD73 in immunosuppressive CAF. By studying cohorts of breast cancer patients and performing functional assays, our study uncovers how CD73 contributes to immunosuppression by acting in a specific CAF subpopulation (referred to as CAF-S1) in breast cancer. In addition, we validate that using an anti-CD73 antibody significantly reduces CAF-S1-mediated immunosuppression, thereby highlighting a new interesting therapeutic strategy for breast cancer patients. Abstract Background: Cancer-associated fibroblasts (CAF) are heterogeneous with multiple functions in breast cancer. Recently, we identified a specific CAF subpopulation (referred to as CAF-S1), which promotes immunosuppression and immunotherapy resistance. Methods and Results: Here, by studying a large collection of human samples, we highlight the key function of CD73/NT5E in CAF-S1-mediated immunosuppression in breast cancer. We first reveal that CD73 protein level specifically accumulates in CAF-S1 in breast cancer patients. Interestingly, infiltration of regulatory T lymphocytes (Tregs) is significantly correlated with CD73 expression in stroma but not in epithelium, indicating that CD73 contributes to immunosuppression when expressed in CAF-S1 and not in tumor cells. By performing functional assays based on relevant systems using primary CAF-S1 isolated from patients, we demonstrate that CAF-S1 increase the content in both PD-1+ and CTLA-4+ Tregs. Importantly, the use of a blocking anti-CD73 antibody on CAF-S1 reduces CAF-S1-mediated immunosuppression by preventing expression of these immune checkpoints on Tregs. Conclusions: Our data support the potential clinical benefit of using both anti-CD73 and immune-checkpoint inhibitors in breast cancer patients for inhibiting CAF-S1-mediated immunosuppression and enhancing anti-tumor immune response.
- Published
- 2021
- Full Text
- View/download PDF
9. 852 Trifunctional NKp46/CD16a-NK cell engager targeting CD123 overcomes acute myeloid leukemia resistance to ADCC
- Author
-
Laurent Gauthier, Angela Virone-Oddos, Jochen Beninga, Benjamin Rossi, Céline Nicolazzi, Céline Amara, Audrey Blanchard-Alvarez, Nicolas Gourdin, Jacqueline Courta, Alexandra Basset, Franceline Guillot, Gwendoline Grondin, Hélène Bonnevaux, Anne-Laure Bauchet, Ariane Morel, Yannis Morel, Marielle Chiron, and Eric Vivier
- Subjects
Pharmacology ,Cancer Research ,Acute leukemia ,business.industry ,Immunology ,CD33 ,Myeloid leukemia ,Decitabine ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,NKG2D ,medicine.disease ,Natural killer cell ,Leukemia ,medicine.anatomical_structure ,Oncology ,Cancer research ,Molecular Medicine ,Immunology and Allergy ,Medicine ,Cytokine secretion ,business ,RC254-282 ,medicine.drug - Abstract
BackgroundThere is a clear need for targeted therapies to treat acute myeloid leukemia (AML), the most common acute leukemia in adults. CD123 (IL-3 receptor alpha chain) is an attractive target for AML treatment.1 However, cytotoxic antibody targeting CD123 proved insufficiently effective in a combination setting in phase II/III clinical trials.2 T-cell engagers targeting CD123 displayed some clinical efficacy but were often associated with cytokine release syndrome and neurotoxicity.3 Interest in the use of NK cells for therapeutic interventions has increased in recent years, as a potential safer alternative to T cells. Several NK-cell activating receptors, such as CD16a, NKG2D, and the natural cytotoxicity receptors NKp30 and NKp46, can be targeted to induce antitumor immunity. We previously reported the development of trifunctional NK-cell engagers (NKCEs) targeting a tumor antigen on cancer cells and co-engaging NKp46 and CD16a on NK cells.4MethodsWe report here the design, characterization and preclinical development of a novel trifunctional NK cell engager (NKCE) targeting CD123 on AML cells and engaging the activating receptors NKp46 and CD16a on NK cells. The CD123 NKCE therapeutic molecule was engineered with humanized antibodies targeting NKp464 and CD123.5 We compared CD123-NKCE and a cytotoxic ADCC-enhanced antibody (Ab) targeting CD123, in terms of antitumor activity in vitro, ex vivo and in vivo. Pharmacokinetic, pharmacodynamic and safety profile of CD123-NKCE were evaluated in non-human primate (NHP) studies.ResultsThe expression of the high affinity Fc gamma receptor CD64 on patient-derived AML cells inhibited the ADCC of the Ab targeting CD123 in vitro and ex vivo, but not the antitumor activity of CD123-NKCE. CD123-NKCE had potent antitumor activity against primary AML blasts and AML cell lines, promoted strong NK-cell activation and induced cytokine secretion only in the presence of AML target cells. Its antitumor activity in mouse model was greater than that of the comparator antibody. Moreover, CD123-NKCE had strong and prolonged pharmacodynamic effects in NHP when used at very low doses, was well-tolerated up to high 3 mg/kg dose and triggered only minor cytokine release.ConclusionsThe data for activity, safety, pharmacokinetics, and pharmacodynamics provided here demonstrate the superiority of CD123-NKCE over comparator cytotoxic antibody, in terms of antitumor activity in vitro, ex vivo, in vivo, and its favorable safety profile, as compared to T-cell therapies. These results constitute proof-of-principle for the efficacy of CD123-NKCE for controlling AML tumors in vivo, and provide consistent support for their clinical development.ReferencesEhninger A, Kramer M, Rollig C, et al. Distribution and levels of cell surface expression of CD33 and CD123 in acute myeloid leukemia. Blood Cancer J 2014;4:e218.Montesinos P, Gail J Roboz GJ, et al. Safety and efficacy of talacotuzumab plus decitabine or decitabine alone in patients with acute myeloid leukemia not eligible for chemotherapy: results from a multicenter, randomized, phase 2/3 study. Leukemia 2021;35(1):62–74.Uy GL, Aldoss I, Foster MC, et al. Flotetuzumab as salvage immunotherapy for refractory acute myeloid leukemia. Blood 2021;137(6):751–762.Gauthier L, Morel A, Anceriz N, et al. Multifunctional natural killer cell engagers targeting NKp46 trigger protective tumor immunity. Cell 2019;177(7):1701–13.Jin L, Lee EM, Ramshaw HS, et al. Monoclonal antibody-mediated targeting of CD123, IL-3 receptor alpha chain, eliminates human acute myeloid leukemic stem cells. Cell Stem Cell 2009;5:31–42.
- Published
- 2021
10. Antitumor immunity induced by antibody-based natural killer cell engager therapeutics armed with not-alpha IL-2 variant
- Author
-
Olivier Demaria, Laurent Gauthier, Marie Vetizou, Audrey Blanchard Alvarez, Constance Vagne, Guillaume Habif, Luciana Batista, William Baron, Nourhène Belaïd, Mathilde Girard-Madoux, Cedric Cesari, Melody Caratini, Frédéric Bosco, Olivier Benac, Julie Lopez, Aurore Fenis, Justine Galluso, Sylvia Trichard, Barbara Carrette, Florent Carrette, Aurélie Maguer, Solène Jaubert, Audrey Sansaloni, Robin Letay-Drouet, Camille Kosthowa, Naouel Lovera, Arnaud Dujardin, Fabien Chanuc, Mélanie Le Van, Sivan Bokobza, Nicolas Jarmuzynski, Camille Fos, Nicolas Gourdin, Romain Remark, Eric Lechevallier, Nicolas Fakhry, Sébastien Salas, Jean-Laurent Deville, Roger Le Grand, Cécile Bonnafous, Lukas Vollmy, Agnès Represa, Sabrina Carpentier, Benjamin Rossi, Ariane Morel, Stéphanie Cornen, Ivan Perrot, Yannis Morel, Eric Vivier, Innate Pharma, Recherche & Développement, Immunologie des tumeurs et immunothérapie (UMR 1015), Université Paris-Sud - Paris 11 (UP11)-Institut Gustave Roussy (IGR)-Institut National de la Santé et de la Recherche Médicale (INSERM), Institut Necker Enfants-Malades (INEM - UM 111 (UMR 8253 / U1151)), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université Paris Cité (UPCité), Centre d'Immunologie de Marseille - Luminy (CIML), Aix Marseille Université (AMU)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Aix-Marseille Université - École de médecine (AMU SMPM MED), Aix-Marseille Université - Faculté des sciences médicales et paramédicales (AMU SMPM), Aix Marseille Université (AMU)-Aix Marseille Université (AMU), Institut Cochin (UMR_S567 / UMR 8104), Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Méthodes computationnelles pour la prise en charge thérapeutique en oncologie : Optimisation des stratégies par modélisation mécaniste et statistique (COMPO), Inria Sophia Antipolis - Méditerranée (CRISAM), Institut National de Recherche en Informatique et en Automatique (Inria)-Institut National de Recherche en Informatique et en Automatique (Inria)-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)-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), and Assistance Publique - Hôpitaux de Marseille (APHM)
- Subjects
MESH: Killer Cells, Natural ,MESH: Cytokines ,Cancer immunotherapy ,MESH: Interleukin-2 ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Receptors, Interleukin-2 ,MESH: Chemokines ,General Biochemistry, Genetics and Molecular Biology ,MESH: Receptors, Interleukin-2 ,Killer Cells, Natural ,Natural Killer cells ,Neoplasms ,cytokine ,Animals ,Interleukin-2 ,Cytokines ,MESH: Neoplasms ,multispecific antibodies ,Chemokines - Abstract
International audience; Harnessing innate immunity is emerging as a promising therapeutic approach in cancer. We report here the design of tetraspecific molecules engaging natural killer (NK) cell-activating receptors NKp46 and CD16a, the β-chain of the interleukin-2 receptor (IL-2R), and a tumor-associated antigen (TAA). In vitro, these tetraspecific antibody-based natural killer cell engager therapeutics (ANKETs) induce a preferential activation and proliferation of NK cells, and the binding to the targeted TAA triggers NK cell cytotoxicity and cytokine and chemokine production. In vivo, tetraspecific ANKETs induce NK cell proliferation and their accumulation at the tumor bed, as well as the control of local and disseminated tumors. Treatment of non-human primates with CD20-directed tetraspecific ANKET leads to CD20+ circulating B cell depletion, with minimal systemic cytokine release and no sign of toxicity. Tetraspecific ANKETs, thus, constitute a technological platform for harnessing NK cells as next-generation cancer immunotherapies.
- Published
- 2021
11. Autocrine Adenosine Regulates Tumor Polyfunctional CD73+CD4+ Effector T Cells Devoid of Immune Checkpoints
- Author
-
Pedro Romero, Marion Bossennec, Isabelle Durand, Bertrand Dubois, Christelle Machon, David Bauché, Julien Faget, Nicolas Gourdin, Christophe Caux, Jérôme Guitton, Julien C. Marie, Camilla Jandus, Nicolas Chopin, Olivier Tredan, Christine Ménétrier-Caux, Céline Rodriguez, Selena Vigano, Centre de Recherche en Cancérologie de Lyon (UNICANCER/CRCL), Centre Léon Bérard [Lyon]-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Université de Lausanne (UNIL), Centre Hospitalier Lyon Sud [CHU - HCL] (CHLS), Hospices Civils de Lyon (HCL), Institut des Sciences Pharmaceutiques et Biologiques (ISPB), Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon, German Cancer Research Center - Deutsches Krebsforschungszentrum [Heidelberg] (DKFZ), Institut de Recherche en Cancérologie de Montpellier (IRCM - U1194 Inserm - UM), CRLCC Val d'Aurelle - Paul Lamarque-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Montpellier (UM), Department of Medical Oncology [Lyon], Centre Léon Bérard [Lyon], Centre de Recherche en Cancérologie de Lyon (CRCL), Université de Lyon-Université de Lyon-Centre Léon Bérard [Lyon]-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), CRLCC Val d'Aurelle - Paul Lamarque-Université de Montpellier (UM)-Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Lausanne = University of Lausanne (UNIL), and Herrada, Anthony
- Subjects
0301 basic medicine ,Cancer Research ,T-Lymphocytes ,Drug Resistance ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Antineoplastic Agents ,chemical and pharmacologic phenomena ,C-C chemokine receptor type 6 ,CXCR3 ,GPI-Linked Proteins/metabolism ,Helper-Inducer/immunology/metabolism ,5'-Nucleotidase/metabolism ,Adenosine/metabolism ,Antineoplastic Agents, Immunological/pharmacology ,Antineoplastic Agents, Immunological/therapeutic use ,Apyrase/metabolism ,Breast Neoplasms/drug therapy ,Breast Neoplasms/immunology ,Breast Neoplasms/pathology ,Costimulatory and Inhibitory T-Cell Receptors/antagonists & inhibitors ,Costimulatory and Inhibitory T-Cell Receptors/metabolism ,Drug Resistance, Neoplasm/immunology ,Female ,Humans ,Interleukin-17/metabolism ,Lymphocytes, Tumor-Infiltrating/immunology ,Lymphocytes, Tumor-Infiltrating/metabolism ,Ovarian Neoplasms/drug therapy ,Ovarian Neoplasms/immunology ,Ovarian Neoplasms/pathology ,T-Lymphocytes, Helper-Inducer/immunology ,T-Lymphocytes, Helper-Inducer/metabolism ,T-Lymphocytes, Regulatory/immunology ,T-Lymphocytes, Regulatory/metabolism ,Tumor Escape/immunology ,Regulatory/immunology/metabolism ,Interleukin 22 ,03 medical and health sciences ,Immune system ,[SDV.CAN] Life Sciences [q-bio]/Cancer ,Lymphocytes ,Autocrine signalling ,Costimulatory and Inhibitory T-Cell Receptors/antagonists & inhibitors/metabolism ,[SDV.MHEP] Life Sciences [q-bio]/Human health and pathology ,Breast Neoplasms/drug therapy/immunology/pathology ,Chemistry ,Effector ,Neoplasm/immunology ,Immunological/pharmacology/therapeutic use ,Immune checkpoint ,Tumor-Infiltrating/immunology/metabolism ,030104 developmental biology ,Oncology ,Tumor Escape ,Cancer research ,Ovarian Neoplasms/drug therapy/immunology/pathology ,[SDV.MHEP]Life Sciences [q-bio]/Human health and pathology - Abstract
The production of CD73-derived adenosine (Ado) by Tregs has been proposed as a resistance mechanism to anti-PD-1 therapy in murine tumor models. We reported that human Tregs express the ectonucleotidase CD39, which generates AMP from ATP, but do not express the AMPase CD73. In contrast, CD73 defined a subset of effector CD4 + T cells (Teffs) enriched in polyfunctional Th1.17 cells characterized by expression of CXCR3, CCR6, and MDR1, and production of IL17A/IFNγ/IL22/GM-CSF. CD39 + Tregs selectively targeted CD73 + Teffs through cooperative degradation of ATP into Ado inhibiting and restricting the ability of CD73 + Teffs to secrete IL17A. CD73 + Teffs infiltrating breast and ovarian tumors were functionally blunted by Tregs expressing upregulated levels of CD39 and ATPase activity. Moreover, tumor-infiltrating CD73 + Teffs failed to express inhibitory immune checkpoints, suggesting that CD73 might be selected under pressure from immune checkpoint blockade therapy and thus may represent a nonredundant target for restoring antitumor immunity.Significance: Polyfunctional CD73 + T-cell effectors lacking other immune checkpoints are selectively targeted by CD39 overexpressing Tregs that dominate the breast tumor environment. Cancer Res; 78(13); 3604-18. ©2018 AACR.
- Published
- 2018
12. Blocking Antibodies Targeting the CD39/CD73 Immunosuppressive Pathway Unleash Immune Responses in Combination Cancer Therapies
- Author
-
Hélène Rispaud-Blanc, Jean-François Eliaou, Cécile Dejou, Rachel Courtois, Béatrice Amigues, Emilie Narni-Mancinelli, Nathalie Bonnefoy, Jérémy Bastid, Armand Bensussan, Marc Giraudon-Paoli, Aurélie Docquier, François Romagné, O. Becquart, Carine Paturel, Ivan Perrot, Benjamin Rossi, Yannis Morel, Laurent Gauthier, Henri-Alexandre Michaud, Laurent Gros, Eric Vivier, Nicolas Gourdin, Alain Roussel, Diana Jecko, Stéphanie Chanteux, Severine Augier, Innate Pharma, Institut de Recherche en Cancérologie de Montpellier (IRCM - U1194 Inserm - UM), CRLCC Val d'Aurelle - Paul Lamarque-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Montpellier (UM), Centre Hospitalier Régional Universitaire [Montpellier] (CHRU Montpellier), Centre de Recherche en Cancérologie de Lyon (UNICANCER/CRCL), Centre Léon Bérard [Lyon]-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Architecture et fonction des macromolécules biologiques (AFMB), Centre National de la Recherche Scientifique (CNRS)-Aix Marseille Université (AMU)-Institut National de la Recherche Agronomique (INRA), Immunologie, dermatologie, oncologie, Oncodermatologie, immunologie et cellules souches cutanées (IDO (U976 / UMR_S 976)), Université Paris Diderot - Paris 7 (UPD7)-Institut National de la Santé et de la Recherche Médicale (INSERM), OREGA Biotech, Centre d'Immunologie de Marseille - Luminy (CIML), Aix Marseille Université (AMU)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris Diderot - Paris 7 (UPD7), CRLCC Val d'Aurelle - Paul Lamarque-Université de Montpellier (UM)-Institut National de la Santé et de la Recherche Médicale (INSERM), Biologie et physiopathologie cutanées : expression génique, signalisation et thérapie, Université Nice Sophia Antipolis (... - 2019) (UNS), Université Côte d'Azur (UCA)-Université Côte d'Azur (UCA)-IFR50-Institut National de la Santé et de la Recherche Médicale (INSERM), IFR50, Université Côte d'Azur (UCA)-Université Côte d'Azur (UCA)-Faculté de Médecine Nice, Dynamique Musculaire et Métabolisme (DMEM), Institut National de la Recherche Agronomique (INRA)-Université de Montpellier (UM), AB Science SA, Cellules Souches et Radiations (SCSR (U967 / UMR-E_008)), Institut National de la Santé et de la Recherche Médicale (INSERM)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Université Paris Diderot - Paris 7 (UPD7)-Université Paris-Sud - Paris 11 (UP11), Centre de Recherche en Cancérologie de Lyon (CRCL), Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Centre Léon Bérard [Lyon]-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Institut Curie, Institut National de la Recherche Agronomique (INRA)-Aix Marseille Université (AMU)-Centre National de la Recherche Scientifique (CNRS), Recherche & Développement, Université Paris-Sud - Paris 11 (UP11)-Université Paris Diderot - Paris 7 (UPD7)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Institut National de la Santé et de la Recherche Médicale (INSERM), and Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Aix Marseille Université (AMU)
- Subjects
0301 basic medicine ,Adenosine ,T-Lymphocytes ,medicine.medical_treatment ,Mice ,Adenosine Triphosphate ,0302 clinical medicine ,Tumor Microenvironment ,Medicine ,Gene Knock-In Techniques ,5'-Nucleotidase ,Melanoma ,lcsh:QH301-705.5 ,Mice, Knockout ,biology ,Apyrase ,Immunosuppression ,3. Good health ,Oxaliplatin ,Survival Rate ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,Antibody ,medicine.drug ,medicine.drug_class ,Antineoplastic Agents ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Monoclonal antibody ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Immune system ,Antigens, CD ,Cell Line, Tumor ,Blocking antibody ,Animals ,Humans ,Antibodies, Blocking ,business.industry ,Cancer ,[SDV.IMM.IMM]Life Sciences [q-bio]/Immunology/Immunotherapy ,medicine.disease ,Immune checkpoint ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,lcsh:Biology (General) ,Leukocytes, Mononuclear ,biology.protein ,Cancer research ,business ,030217 neurology & neurosurgery - Abstract
Summary: Immune checkpoint inhibitors have revolutionized cancer treatment. However, many cancers are resistant to ICIs, and the targeting of additional inhibitory signals is crucial for limiting tumor evasion. The production of adenosine via the sequential activity of CD39 and CD73 ectoenzymes participates to the generation of an immunosuppressive tumor microenvironment. In order to disrupt the adenosine pathway, we generated two antibodies, IPH5201 and IPH5301, targeting human membrane-associated and soluble forms of CD39 and CD73, respectively, and efficiently blocking the hydrolysis of immunogenic ATP into immunosuppressive adenosine. These antibodies promoted antitumor immunity by stimulating dendritic cells and macrophages and by restoring the activation of T cells isolated from cancer patients. In a human CD39 knockin mouse preclinical model, IPH5201 increased the anti-tumor activity of the ATP-inducing chemotherapeutic drug oxaliplatin. These results support the use of anti-CD39 and anti-CD73 monoclonal antibodies and their combination with immune checkpoint inhibitors and chemotherapies in cancer. : The production of adenosine via CD39 and CD73 ectoenzymes participates in an immunosuppressive tumor microenvironment. Perrot et al. generated two antibodies, IPH5201 and IPH5301, targeting human CD39 and CD73, respectively. In vitro and in vivo data support the use of anti-CD39 and anti-CD73 mAbs in combination cancer therapies. Keywords: CD39, CD73, cancer immunotherapies, therapeutic antibodies, adenosine pathway, tumor micro-environment, immunosuppression
- Published
- 2019
- Full Text
- View/download PDF
13. Autocrine Adenosine Regulates Tumor Polyfunctional CD73
- Author
-
Nicolas, Gourdin, Marion, Bossennec, Céline, Rodriguez, Selena, Vigano, Christelle, Machon, Camilla, Jandus, David, Bauché, Julien, Faget, Isabelle, Durand, Nicolas, Chopin, Olivier, Tredan, Julien C, Marie, Bertrand, Dubois, Jérôme, Guitton, Pedro, Romero, Christophe, Caux, and Christine, Ménétrier-Caux
- Subjects
Ovarian Neoplasms ,Adenosine ,Apyrase ,Interleukin-17 ,Breast Neoplasms ,T-Lymphocytes, Helper-Inducer ,GPI-Linked Proteins ,T-Lymphocytes, Regulatory ,Antineoplastic Agents, Immunological ,Lymphocytes, Tumor-Infiltrating ,Costimulatory and Inhibitory T-Cell Receptors ,Drug Resistance, Neoplasm ,Humans ,Female ,Tumor Escape ,5'-Nucleotidase - Abstract
The production of CD73-derived adenosine (Ado) by Tregs has been proposed as a resistance mechanism to anti-PD-1 therapy in murine tumor models. We reported that human Tregs express the ectonucleotidase CD39, which generates AMP from ATP, but do not express the AMPase CD73. In contrast, CD73 defined a subset of effector CD4
- Published
- 2017
14. Abstract 2718: Preclinical development of humanized CD39 and CD73 blocking antibodies targeting the ATP/adenosine immune checkpoint pathway for cancer immunotherapy
- Author
-
Paul Ricaut, Aurélie Docquier, Ariane Morel, Marc Giraudon Paoli, Diana Jecko, Frédéric Bosco, Caroline Denis, Romain Remark, Cécile Bonnafous, Laurent Gauthier, Carine Paturel, Jérémy Bastid, Benjamin Rossi, Nicolas Gourdin, Stéphanie Chanteux, Severine Augier, Nathalie Bonnefoy, Rachel Courtois, Marilyne Royannez Blemont, Cyril Perrier, Agnès Represa, Maryline Salin Agu, Yannis Morel, Ivan Perrot, Stephane Delahaye, and Marion Gaudin
- Subjects
0301 basic medicine ,Cancer Research ,Tumor microenvironment ,biology ,medicine.drug_class ,business.industry ,T cell ,medicine.medical_treatment ,Monoclonal antibody ,Immune checkpoint ,03 medical and health sciences ,030104 developmental biology ,Immune system ,medicine.anatomical_structure ,Oncology ,Cancer immunotherapy ,medicine ,Cancer research ,biology.protein ,Immunogenic cell death ,Antibody ,business - Abstract
Within the tumor microenvironment, adenosine causes immune suppression and dysregulation of immune cell infiltrates resulting in tumor spreading. Adenosine accumulation results from the hydrolysis of extracellular immunoactivating ATP and ADP into AMP by the CD39 (ENTPD1) ectonucleotidase. AMP can be further hydrolyzed into immunosuppressive Adenosine (Ado) by CD73 (NT5E), a cell membrane ectonucleotidase of the NTPDase family.The immunosuppressive role of CD39 expressed on both Tregs and tumor cells has been demonstrated in several reports. Conversely, CD73 expression in the tumor environment has been associated with poor disease outcome and/or with a pro-metastatic phenotype. Blockade of CD39 and CD73 may promote anti-tumor immunity directly by accumulating immunostimulating ATP for CD39 and indirectly by reducing adenosine accumulation for both targets.In cancer tissue, using IHC and flow cytometry analyses, we observed that while CD73 is often expressed by tumor cells, CD39 is more frequently up-regulated on tumor infiltrating cells compared to PBMC or adjacent non-tumor tissue. We next describe the discovery and preclinical development of a unique anti-huCD39 blocking antibody and of an anti-human CD73 antibody for cancer immunotherapy. These anti-CD39 and anti-CD73 antibodies specifically bind with high affinity to huCD39 and huCD73 proteins, respectively. They potently inhibit enzyme activity of their respective targets under their soluble and membrane-associated forms, without inducing down-modulation of these enzymes expressed at the cell surface. Innate's antibodies efficiently reverse Ado-mediated T cell suppression in vitro in presence of ATP and both CD39- and CD73-expressing immune cells and additionally exhibit unique features. The anti-CD39 Ab maintains high concentration of ATP in the extracellular compartment that enhances DC activation and subsequent T cell proliferation in vitro. The anti-CD73 blocking Ab exhibits a more potent ability to block soluble and membrane-associated CD73 enzyme activity than benchmark Abs currently in clinical development. Finally, in vivo blockade of ATP/Ado pathway in CD39ko mice resulted in improved anti-tumor efficacy of immunogenic cell death inducer chemotherapy and of immune checkpoint therapies, including PD1 and CTLA4.Taken together, these data support the clinical development of anti-CD39 and anti-CD73 neutralizing Abs for cancer immunotherapy, potentially in combination with chemotherapy or Immune Checkpoint therapy. The humanized anti-huCD39 and anti-huCD73 monoclonal antibodies are currently in preclinical development.The research leading to CD73 results were obtained within the TumAdoR collaborative consortium that received funding from the European Community's Seventh Framework Program (FP7/2007-2013) under grant agreement n°602200. Citation Format: Ivan Perrot, Marc Giraudon Paoli, Séverine Augier, Marilyne Royannez Blemont, Marion Gaudin, Frédéric Bosco, Rachel Courtois, Stephane Delahaye, Diana Jecko, Nicolas Gourdin, Maryline Salin Agu, Cyril Perrier, Paul Ricaut, Aurélie Docquier, Stéphanie Chanteux, Benjamin Rossi, Agnès Représa, Caroline Denis, Romain Remark, Cécile Bonnafous, Laurent Gauthier, Ariane Morel, Nathalie Bonnefoy, Jérémy Bastid, Yannis Morel, Carine Paturel. Preclinical development of humanized CD39 and CD73 blocking antibodies targeting the ATP/adenosine immune checkpoint pathway for cancer immunotherapy [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 2718.
- Published
- 2018
15. Abstract 2344: Discovery and characterization of new original blocking antibodies targeting the CD73 immune checkpoint for cancer immunotherapy
- Author
-
Céline Rodriguez, Marc Giraudon Paoli, Hélène Rispaud Blanc, Yannis Morel, Laurent Gauthier, Carine Paturel, Nicolas Gourdin, Christophe Caux, Ivan Perrot, Stéphanie Chanteux, Severine Augier, Christine Menetrier Caux, and Marilyne Royannez Blemont
- Subjects
0301 basic medicine ,Cancer Research ,Tumor microenvironment ,biology ,Chemistry ,T cell ,medicine.medical_treatment ,Virology ,Immune checkpoint ,03 medical and health sciences ,030104 developmental biology ,Immune system ,medicine.anatomical_structure ,Epitope mapping ,Oncology ,Cancer immunotherapy ,Blocking antibody ,biology.protein ,Cancer research ,medicine ,Antibody - Abstract
CD73 (NT5E) is a cell membrane ectoenzyme of the NTPDase family that plays a major role in the conversion of AMP into Adenosine (Ado). Within the tumor microenvironment, accumulation of Ado causes immune suppression and dysregulation of immune cell infiltrates resulting in tumor spreading. CD73 expression in the tumor environment has been associated with poor disease outcome and/or with a pro-metastatic phenotype. Thus, targeting CD73 may promote anti-tumor immunity by reducing Ado accumulation and may block tumor cell metastasis by inhibiting CD73 on tumor cells. Here, we describe the generation and characterization of novel anti-human CD73 antibodies, intended for the treatment of a wide range of cancers. The research leading to these results has received funding from the European Community's Seventh Framework Program (FP7/2007-2013) under grant agreement n°602200. Antibodies were discovered that inhibited CD73 function by different mechanisms, including the direct blocking of CD73 enzymatic activity or the down-modulation of membrane CD73 expression. Epitope mapping revealed that antibodies acting by these different modes of action bound to distinct sites on CD73. All selected antibodies cross-react with cynomolgus CD73 protein and have strong avidity and affinity for membrane or recombinant CD73, by flow cytometry and Surface Plasmon Resonance, respectively. Antibodies that inhibit CD73 enzymatic activity strongly reduce AMP catabolism by both recombinant and cellular CD73 with IC50 in the nanomolar range. They also efficiently reverse ATP- and AMP-mediated T cell suppression in in vitro assays in presence of both CD39+ and CD73+ cells. The antibodies that induce down-modulation of cellular CD73 expression do not block recombinant CD73 enzyme activity and partially inhibit cellular CD73 activity; they reverse ATP- but not AMP-dependent T cell suppression. The antibodies displaying the most interesting features were humanized. Evaluation of their activity in animal models is ongoing. Citation Format: Marc Giraudon Paoli, Severine Augier, Marilyne Royannez Blemont, Céline Rodriguez, Hélène Rispaud Blanc, Stéphanie Chanteux, Nicolas Gourdin, Laurent Gauthier, Christine Ménétrier Caux, Yannis Morel, Christophe Caux, Carine Paturel, Ivan Perrot. Discovery and characterization of new original blocking antibodies targeting the CD73 immune checkpoint for cancer immunotherapy. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 2344.
- Published
- 2016
16. Abstract 2338: CD39+ Treg cooperate with a CD73-expressing Th1/Th17 subset for Adenosine-mediated immunosuppression in human breast tumors
- Author
-
Nicolas Gourdin, Christine Ménétrier-Caux, Selena Vigano, Pedro Romero, Jean-Yves Blay, Camilla Jandus, Julien Faget, Isabelle Durand, Céline Rodriguez, and Christophe Caux
- Subjects
Cancer Research ,business.industry ,medicine.medical_treatment ,hemic and immune systems ,Immunosuppression ,C-C chemokine receptor type 6 ,CXCR3 ,Cytokine ,Oncology ,Immunology ,medicine ,Tumor necrosis factor alpha ,business ,Receptor ,CD8 ,CCL22 - Abstract
Our group and others have previously reported that Treg infiltrating (Ti-Treg) breast tumors have a negative impact on patients’ outcome. We further reported their selective recruitment in the tumor environment through CCL22 secretion and expansion upon ICOS engagement by plasmacytoïd dendritic cells. Here, we investigated the mechanism of Ti-Treg mediated suppression and observed that Ti-Treg express high CD39 levels. CD39 is an ectonucleotidase that cooperates with CD73 to degrade the danger signal ATP into immunosuppressive Adenosine (Ado). The potency of Ado mediated suppression is illustrated in Adenosine-Deaminase (ADA)-deficient patients unable to degrade Ado and developing severe immunodeficiency. We further show that, in contrast to murine Treg, human CD39+Treg do not express CD73 enabling them only to degrade ATP into AMP. Within T cells, CD73 expression was mainly associated with naïve CD8+ T cells and a subset of memory conventional CD4+ T cells (Tconv) that exhibit Th1/Th17 characteristics (CXCR3+CCR6+CCR4negIFNγhighIL17+). CD39+ Treg isolated from healthy donor blood, in presence of exogenous ATP, potently inhibit purified CD73+ but not CD73neg Tconv, proliferation and cytokine production (IFNγ, TNFα). By using enzymatic inhibitors, we demonstrated the involvement of CD39 and CD73 through Treg/Tconv cooperation for Ado mediated immunosuppression. Of importance, when integrated in [Treg-CD4+CD73+] high density coculture, CD4+CD73neg T cells expressing similar levels of Ado receptors (A2A, A2B) are also inhibited. In conclusion, our findings support the existence of an Ado mediated immunosuppression loop in the tumor through cooperation between CD39highTreg and CD73-expressing Th1/Th17 subsets in the breast tumor environment. The research leading to these results has received funding from the European Community's Seventh Framework Program (FP7/2007-2013) under grant agreement n°602200. Citation Format: Nicolas Gourdin, Céline Rodriguez, Selena Vigano, Isabelle Durand, Julien Faget, Camilla Jandus, Jean Yves Blay, Pedro Romero, Christine Menetrier-Caux, Christophe Caux. CD39+ Treg cooperate with a CD73-expressing Th1/Th17 subset for Adenosine-mediated immunosuppression in human breast tumors. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 2338.
- Published
- 2016
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.