53 results on '"Yannis Morel"'
Search Results
2. 852 Trifunctional NKp46/CD16a-NK cell engager targeting CD123 overcomes acute myeloid leukemia resistance to ADCC
- Author
-
Yannis Morel, Eric Vivier, Benjamin Rossi, Laurent Gauthier, Anne-Laure Bauchet, Ariane Morel, Marielle Chiron, Angela Virone-Oddos, Jochen Beninga, Céline Nicolazzi, Céline Amara, Audrey Blanchard-Alvarez, Nicolas Gourdin, Jacqueline Courta, Alexandra Basset, Franceline Guillot, Gwendoline Grondin, and Hélène Bonnevaux
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2021
- Full Text
- View/download PDF
3. Targeting MICA/B with cytotoxic therapeutic antibodies leads to tumor control [version 2; peer review: 2 approved]
- Author
-
Mathieu Bléry, Manel Mrabet-Kraiem, Ariane Morel, Florence Lhospice, Delphine Bregeon, Cécile Bonnafous, Laurent Gauthier, Benjamin Rossi, Romain Remark, Stéphanie Cornen, Nadia Anceriz, Nicolas Viaud, Sylvia Trichard, Sabrina Carpentier, Alix Joulin-Giet, Gwendoline Grondin, Veronika Liptakova, Younghoon Kim, Laurent Daniel, Aurélie Haffner, Nicolas Macagno, Laurent Pouyet, Ivan Perrot, Carine Paturel, Yannis Morel, Alexander Steinle, François Romagné, Emilie Narni-Mancinelli, and Eric Vivier
- Subjects
Science ,Social Sciences - Abstract
Background: MICA and MICB are tightly regulated stress-induced proteins that trigger the immune system by binding to the activating receptor NKG2D on cytotoxic lymphocytes. MICA and MICB are highly polymorphic molecules with prevalent expression on several types of solid tumors and limited expression in normal/healthy tissues, making them attractive targets for therapeutic intervention. Methods: We have generated a series of anti-MICA and MICB cross-reactive antibodies with the unique feature of binding to the most prevalent isoforms of both these molecules. Results: The anti-MICA and MICB antibody MICAB1, a human IgG1 Fc-engineered monoclonal antibody (mAb), displayed potent antibody-dependent cellular cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP) of MICA/B-expressing tumor cells in vitro. However, it showed insufficient efficiency against solid tumors in vivo, which prompted the development of antibody-drug conjugates (ADC). Indeed, optimal tumor control was achieved with MICAB1-ADC format in several solid tumor models, including patient-derived xenografts (PDX) and carcinogen-induced tumors in immunocompetent MICAgen transgenic mice. Conclusions: These data indicate that MICA and MICB are promising targets for cytotoxic immunotherapy.
- Published
- 2021
- Full Text
- View/download PDF
4. 483 Association of COVID-19 inflammation with activation of the C5a-C5aR1 axis
- Author
-
Mikael Ebbo, Nicolas Schleinitz, Yannis Morel, Luciana Batista, Sabrina Carpentier, Romain Remark, Eric Vivier, Frédéric Vély, Olivier Demaria, Julien Carvelli, Nassima Chouaki Benmansour, Joanna Fares, Marie-Laure Thibult, Ariane Morel, Agnes Represa, and Robert Zerbib
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2020
- Full Text
- View/download PDF
5. 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
6. Daratumumab-mediated lysis of primary multiple myeloma cells is enhanced in combination with the human anti-KIR antibody IPH2102 and lenalidomide
- Author
-
Inger S. Nijhof, Jeroen J. Lammerts van Bueren, Berris van Kessel, Pascale Andre, Yannis Morel, Henk M. Lokhorst, Niels W.C.J. van de Donk, Paul W.H.I. Parren, and Tuna Mutis
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Despite recent treatment improvements, multiple myeloma remains an incurable disease. Since antibody-dependent cell-mediated cytotoxicity is an important effector mechanism of daratumumab, we explored the possibility of improving daratumumab-mediated cell-mediated cytotoxicity by blocking natural killer cell inhibitory receptors with the human monoclonal anti-KIR antibody IPH2102, next to activation of natural killer cells with the immune modulatory drug lenalidomide. In 4-hour antibody-dependent cell-mediated cytotoxicity assays, IPH2102 did not induce lysis of multiple myeloma cell lines, but it did significantly augment daratumumab-induced myeloma cell lysis. Also in an ex vivo setting, IPH2102 synergistically improved daratumumab-dependent lysis of primary myeloma cells in bone marrow mononuclear cells (n=21), especially in patients carrying the FcγRIIIa-158F allele or the FcγRIIa-131R allele, who bind IgG1 with lower affinity than patients carrying the FcγRIIIa-158V allele or the FcγRIIa-131H allele. Finally, a further synergistically improved myeloma cell lysis with the daratumumab-IPH2102 combination was observed by adding lenalidomide, which suggests that more effective treatment strategies can be designed for multiple myeloma by combining daratumumab with agents that independently modulate natural killer cell function.
- Published
- 2015
- Full Text
- View/download PDF
7. The toll-like receptor agonist imiquimod is active against prions.
- Author
-
Nassima Oumata, Phu Hai Nguyen, Vincent Beringue, Flavie Soubigou, Yanhong Pang, Nathalie Desban, Catherine Massacrier, Yannis Morel, Carine Paturel, Marie-Astrid Contesse, Serge Bouaziz, Suparna Sanyal, Hervé Galons, Marc Blondel, and Cécile Voisset
- Subjects
Medicine ,Science - Abstract
Using a yeast-based assay, a previously unsuspected antiprion activity was found for imiquimod (IQ), a potent Toll-like receptor 7 (TLR7) agonist already used for clinical applications. The antiprion activity of IQ was first detected against yeast prions [PSI (+) ] and [URE3], and then against mammalian prion both ex vivo in a cell-based assay and in vivo in a transgenic mouse model for prion diseases. In order to facilitate structure-activity relationship studies, we conducted a new synthetic pathway which provides a more efficient means of producing new IQ chemical derivatives, the activity of which was tested against both yeast and mammalian prions. The comparable antiprion activity of IQ and its chemical derivatives in the above life forms further emphasizes the conservation of prion controlling mechanisms throughout evolution. Interestingly, this study also demonstrated that the antiprion activity of IQ and IQ-derived compounds is independent from their ability to stimulate TLRs. Furthermore, we found that IQ and its active chemical derivatives inhibit the protein folding activity of the ribosome (PFAR) in vitro.
- Published
- 2013
- Full Text
- View/download PDF
8. Data from Opposing Effects of Toll-like Receptor (TLR3) Signaling in Tumors Can Be Therapeutically Uncoupled to Optimize the Anticancer Efficacy of TLR3 Ligands
- Author
-
Laurence Zitvogel, Guido Kroemer, Fabrice André, Nathalie Chaput, Christophe Combadière, Robert Schreiber, Ravindra Uppaluri, Maria Ferrantini, Bernard Ryffel, Sophie Viaud, Magali Terme, Carine Paturel, Yannis Morel, Yuting Ma, and Rosa Conforti
- Abstract
Many cancer cells express Toll-like receptors (TLR) that offer possible therapeutic targets. Polyadenylic-polyuridylic acid [poly(A:U)] is an agonist of the Toll-like receptor TLR3 that displays anticancer properties. In this study, we illustrate how the immunostimulatory and immunosuppressive effects of this agent can be uncoupled to therapeutic advantage. We took advantage of two TLR3-expressing tumor models that produced large amounts of CCL5 (a CCR5 ligand) and CXCL10 (a CXCR3 ligand) in response to type I IFN and poly(A:U), both in vitro and in vivo. Conventional chemotherapy or in vivo injection of poly(A:U), alone or in combination, failed to reduce tumor growth unless an immunochemotherapeutic regimen of vaccination against tumor antigens was included. CCL5 blockade improved the efficacy of immunochemotherapy, whereas CXCR3 blockade abolished its beneficial effects. These findings show how poly(A:U) can elicit production of a range of chemokines by tumor cells that reinforce immunostimulatory or immunosuppressive effects. Optimizing the anticancer effects of TLR3 agonists may require manipulating these chemokines or their receptors. Cancer Res; 70(2); 490–500
- Published
- 2023
- Full Text
- View/download PDF
9. Supplementary Figure Legends 1-2, Table Legend and Methods from TLR3 as a Biomarker for the Therapeutic Efficacy of Double-stranded RNA in Breast Cancer
- Author
-
Fabrice Andre, Pedro Romero, Donata Rimoldi, Michel Pierres, Serge Lebecque, Aicha Goubar, Jean-Christophe Sabourin, Marie Pierre Chenard, Rosa Conforti, Catherine Massacrier, Clarisse Dubois, Karine Chemin, Yannis Morel, Carine Asselin-Paturel, Laurence Zitvogel, and Bruno Salaun
- Abstract
Supplementary Figure Legends 1-2, Table Legend and Methods from TLR3 as a Biomarker for the Therapeutic Efficacy of Double-stranded RNA in Breast Cancer
- Published
- 2023
- Full Text
- View/download PDF
10. Supplementary Figure Legends 1-6 from Opposing Effects of Toll-like Receptor (TLR3) Signaling in Tumors Can Be Therapeutically Uncoupled to Optimize the Anticancer Efficacy of TLR3 Ligands
- Author
-
Laurence Zitvogel, Guido Kroemer, Fabrice André, Nathalie Chaput, Christophe Combadière, Robert Schreiber, Ravindra Uppaluri, Maria Ferrantini, Bernard Ryffel, Sophie Viaud, Magali Terme, Carine Paturel, Yannis Morel, Yuting Ma, and Rosa Conforti
- Abstract
Supplementary Figure Legends 1-6 from Opposing Effects of Toll-like Receptor (TLR3) Signaling in Tumors Can Be Therapeutically Uncoupled to Optimize the Anticancer Efficacy of TLR3 Ligands
- Published
- 2023
- Full Text
- View/download PDF
11. Supplementary Figures 1-6 from Opposing Effects of Toll-like Receptor (TLR3) Signaling in Tumors Can Be Therapeutically Uncoupled to Optimize the Anticancer Efficacy of TLR3 Ligands
- Author
-
Laurence Zitvogel, Guido Kroemer, Fabrice André, Nathalie Chaput, Christophe Combadière, Robert Schreiber, Ravindra Uppaluri, Maria Ferrantini, Bernard Ryffel, Sophie Viaud, Magali Terme, Carine Paturel, Yannis Morel, Yuting Ma, and Rosa Conforti
- Abstract
Supplementary Figures 1-6 from Opposing Effects of Toll-like Receptor (TLR3) Signaling in Tumors Can Be Therapeutically Uncoupled to Optimize the Anticancer Efficacy of TLR3 Ligands
- Published
- 2023
- Full Text
- View/download PDF
12. Supplementary Table 1 from TLR3 as a Biomarker for the Therapeutic Efficacy of Double-stranded RNA in Breast Cancer
- Author
-
Fabrice Andre, Pedro Romero, Donata Rimoldi, Michel Pierres, Serge Lebecque, Aicha Goubar, Jean-Christophe Sabourin, Marie Pierre Chenard, Rosa Conforti, Catherine Massacrier, Clarisse Dubois, Karine Chemin, Yannis Morel, Carine Asselin-Paturel, Laurence Zitvogel, and Bruno Salaun
- Abstract
Supplementary Table 1 from TLR3 as a Biomarker for the Therapeutic Efficacy of Double-stranded RNA in Breast Cancer
- Published
- 2023
- Full Text
- View/download PDF
13. 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
- Full Text
- View/download PDF
14. Targeting MICA/B with cytotoxic therapeutic antibodies leads to tumor control [version 2; peer review: 2 approved]
- Author
-
Mathieu Bléry, Manel Mrabet-Kraiem, Ariane Morel, Florence Lhospice, Delphine Bregeon, Cécile Bonnafous, Laurent Gauthier, Benjamin Rossi, Romain Remark, Stéphanie Cornen, Nadia Anceriz, Nicolas Viaud, Sylvia Trichard, Sabrina Carpentier, Alix Joulin-Giet, Gwendoline Grondin, Veronika Liptakova, Younghoon Kim, Laurent Daniel, Aurélie Haffner, Nicolas Macagno, Laurent Pouyet, Ivan Perrot, Carine Paturel, Yannis Morel, Alexander Steinle, François Romagné, Emilie Narni-Mancinelli, and Eric Vivier
- Subjects
stomatognathic diseases ,Science ,Social Sciences - Abstract
Background: MICA and MICB are tightly regulated stress-induced proteins that trigger the immune system by binding to the activating receptor NKG2D on cytotoxic lymphocytes. MICA and MICB are highly polymorphic molecules with prevalent expression on several types of solid tumors and limited expression in normal/healthy tissues, making them attractive targets for therapeutic intervention. Methods: We have generated a series of anti-MICA and MICB cross-reactive antibodies with the unique feature of binding to the most prevalent isoforms of both these molecules. Results: The anti-MICA and MICB antibody MICAB1, a human IgG1 Fc-engineered monoclonal antibody (mAb), displayed potent antibody-dependent cellular cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP) of MICA/B-expressing tumor cells in vitro. However, it showed insufficient efficiency against solid tumors in vivo, which prompted the development of antibody-drug conjugates (ADC). Indeed, optimal tumor control was achieved with MICAB1-ADC format in several solid tumor models, including patient-derived xenografts (PDX) and carcinogen-induced tumors in immunocompetent MICAgen transgenic mice. Conclusions: These data indicate that MICA and MICB are promising targets for cytotoxic immunotherapy.
- Published
- 2022
15. 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
- Full Text
- View/download PDF
16. 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
17. 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
- Full Text
- View/download PDF
18. 483 Association of COVID-19 inflammation with activation of the C5a-C5aR1 axis
- Author
-
Olivier Demaria, Julien Carvelli, Nassima Chouaki Benmansour, Joanna Fares, Luciana Batista, Marie-Laure Thibult, Ariane Morel, Sabrina Carpentier, Romain Remark, Agnes Represa, Frederic Vely, Mikael Ebbo, Nicolas Schleinitz, Robert Zerbib, Yannis Morel, and Eric Vivier
- Subjects
ARDS ,Lung ,Myeloid ,medicine.drug_class ,business.industry ,Inflammation ,Lung injury ,Monoclonal antibody ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,lcsh:RC254-282 ,Pathophysiology ,medicine.anatomical_structure ,Immune system ,Immunology ,medicine ,medicine.symptom ,business - Abstract
Background Coronavirus disease 2019 (COVID-19) is a new pandemic disease caused by infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The C5a anaphylatoxin and its receptor C5aR1 (CD88) play a key role in the initiation and maintenance of several inflammatory responses, by recruiting and activating neutrophils and monocytes in the lungs. Methods We provide a longitudinal analysis of immune responses, including immune cell phenotyping and assessments of the soluble factors present in the blood and broncho-alveolar lavage fluid (BALF) of patients at various stages of COVID-19 severity: paucisymptomatic, pneumonia and acute respiratory distress syndrome (ARDS) Results We report an increase in soluble C5a levels proportional to COVID-19 severity and high levels of C5aR1 expression in blood and pulmonary myeloid cells, supporting a role for the C5a-C5aR1 axis in the pathophysiology of ARDS. Avdoralimab, an anti-C5aR1 therapeutic monoclonal antibodies (mAbs) prevented C5a-mediated human myeloid cell recruitment and activation, and inhibited acute lung injury (ALI) in human C5aR1 knockin mice. Conclusions These results support the evaluation of avdoralimab to block C5a-C5aR1 axis as a mean of limiting myeloid cell infiltration in damaged organs and preventing the excessive lung inflammation and endothelialitis associated with ARDS in COVID-19 patients Acknowledgements The Explore COVID-19 IPH group, the Explore COVID-19 Marseille Immunopole group. Ethics Approval Human study protocol was approved by the Committee for the Protection of Persons Ile-de-France III – France (#2020-A00757-32). Animal experiments were approved by the ministere de l’enseignement superieur, de la recherche et de l’innovation – France (APAFIS#25418-2020051512242806 v2).
- Published
- 2020
19. Identification of immune checkpoints in COVID-19
- Author
-
Sabrina Carpentier, Joanna Fares, Marie-Laure Thibult, Olivier Demaria, Marc Gainnier, Agnès Represa, Ariane Morel, Eric Vivier, Nassima Chouaki Benmansour, Frédéric Vély, Julien Carvelli, Mikael Ebbo, Nicolas Schleinitz, Pascale Andre, Erwan Le Dault, Pierre Simeone, Yannis Morel, Pierre Yves Cordier, Luciana Batista, Christelle Piperoglou, and Christophe Guervilly
- Subjects
Immune system ,Coronavirus disease 2019 (COVID-19) ,business.industry ,Medicine ,Identification (biology) ,Computational biology ,business - Abstract
Coronavirus disease 2019 (COVID-19) is a new pandemic acute respiratory disease caused by infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)11-3. We provide here a longitudinal analysis of immune responses, including immune cell phenotyping and assessments of the soluble factors present in the blood and broncho-alveolar lavage fluid (BALF) of patients at various stages of COVID-19 severity: paucisymptomatic, pneumonia and acute respiratory distress syndrome (ARDS). While we confirm a lymphopenia associated with COVID-19 severity4-7, we report an increase in expression of the NKG2A and PD-1 inhibitory receptors on T and natural killer (NK) cells, as well as an increase in CD39 expression on NK cells, suggesting that therapeutic blocking antibodies targeting these molecules already used for cancer immunotherapy8 could be repurposed as first line of defense to promote SARS-CoV-2 clearance. In addition, the C5a anaphylatoxin and its receptor C5aR1 (CD88) play a key role in the initiation and maintenance of inflammatory responses, by recruiting neutrophils and monocytes to the lungs9,10. We report an increase in soluble C5a levels proportional to COVID-19 severity and high levels of C5aR1 expression in blood and BALF myeloid cells, indicating a potential role of the C5a-C5aR1 axis in the pathophysiology of ARDS. Avdoralimab is a clinical-stage anti-C5aR1 therapeutic monoclonal antibody (mAb) that prevents C5a-mediated myeloid cell recruitment and activation. We propose the use of this antibody to limit myeloid cell infiltration in the lung and to prevent the excessive lung inflammation associated with ARDS in COVID-19 patients.
- Published
- 2020
- Full Text
- View/download PDF
20. [Natural killer cells: promising targets in cancer therapy]
- Author
-
Stéphanie, Cornen, Pascale, André, Laurent, Gauthier, Yannis, Morel, and Éric, Vivier
- Subjects
Cytotoxicity, Immunologic ,Killer Cells, Natural ,Neoplasms ,Animals ,Humans ,Molecular Targeted Therapy ,Medical Oncology ,Immunologic Surveillance - Abstract
Les cellules natural killer : des cibles prometteuses dans la thérapie contre le cancer.L’immuno-oncologie est une approche d’immunothérapie novatrice qui change le traitement des cancers en stimulant la capacité du système immunitaire à reconnaître et éliminer les cellules tumorales. Cette approche a pour but de mettre en place une immuno-surveillance anti-tumorale durable chez des patients pour lesquels les thérapies conventionnelles ont échoué.
- Published
- 2020
21. 851 Harnessing innate immunity in cancer therapies: the example of natural killer cell engagers
- Author
-
Sabrina Carpentier, Audrey Blanchard Alvarez, Guillaume Habif, Benjamin Rossi, Ariane Morel, Agnès Represa, Yannis Morel, Olivier Demaria, Marie Vetizou, Ivan Perrot, Luciana Batista, Cécile Bonnafous, Eric Vivier, Constance Vagne, Laurent Gauthier, Sivan Bokobza, and Stéphanie Cornen
- Subjects
Pharmacology ,Cancer Research ,Innate immune system ,biology ,Regulatory T cell ,medicine.medical_treatment ,Immunology ,Immunotherapy ,Tumor antigen ,Chimeric antigen receptor ,Natural killer cell ,medicine.anatomical_structure ,Immune system ,Oncology ,medicine ,biology.protein ,Cancer research ,Molecular Medicine ,Immunology and Allergy ,Antibody - Abstract
BackgroundMost immunomodulatory approaches have focused on enhancing T-cell responses, with immune checkpoint inhibitors, chimeric antigen receptor T cells or bispecific antibodies. Although these therapies have led to exceptional successes, only a minority of cancer patients benefit from these treatments, highlighting the need to identify new cells and molecules that could be exploited in the next generation of immunotherapy. Given the crucial role of innate immune responses in immunity, harnessing these responses opens up new possibilities for tumor control. Antibody engineering provides us with great opportunities to induce synthetic immunity and to optimize the biological functions of innate immune cells, in particular by boosting the capacity of Natural Killer (NK) cells to kill tumor cells directly and to stimulate T-cell responses indirectly.MethodsIn order to leverage the advantages of harnessing NK cell effector functions, we used our Antibody-based NK cell Engager Therapeutics (ANKET) molecular platform1 and designed a new generation of molecules that can engage activating receptors NKp46 and CD16, the IL-2Rβ chain and a tumor antigen in a single tetra-specific molecule (ANKET4). The variant of interleukin-2 (IL-2v) integrated in the ANKET4 molecule is unable to bind the α-subunit of its receptor to limit regulatory T cell activation and IL-2Rα-mediated toxicity.ResultsIn vitro, ANKET4 provides proliferation and activation signals targeted to NK cells and induces primary human NK cell cytolytic activity and the secretion of cytokines and chemokines only after binding to the tumor target. In mouse models of both invasive and solid tumors, ANKET4 induced NK cell proliferation and accumulation at the tumor bed, and had a higher anti-tumor efficacy than approved therapeutic antibodies targeting the same tumor antigen. Mechanistically, transcriptomic analysis and in-vivo studies revealed that the geometry of the ANKET4 molecule including NKp46, CD16 and IL-2 receptor binding moieties on the same molecule was essential for its strong activity which results from a synthetic cooperativity between immunoreceptor tyrosine-based activation motif (ITAM) and cytokine signaling pathways. In non-human primates, CD20-directed ANKET4 resulted in sustained CD20+ B-cell depletion with minimal systemic cytokine release and no clinical sign of toxicity.ConclusionsTetra-specific ANKET4 thus constitutes a technological platform combining the induction of NK cell proliferation and effector functions with a manageable safety profile, supporting its clinical development for next-generation cancer immunotherapies.ReferenceGauthier L, Morel A, Anceriz N, Rossi B, Blanchard-Alvarez A, Grondin G, et al. Multifunctional natural killer cell engagers targeting NKp46 trigger protective tumor immunity. Cell 2019;177(7):1701–13 e16.Ethics ApprovalPrimary immune cells were purified from buffy coats from healthy donors obtained from Etablissement Francais du Sang (EFS, Marseille) with written consent from each volunteer.All mouse experiments were performed in accordance with the rules of the Innate Pharma ethics committee and were approved by the Ministère de l’Enseignement Supérieur, de la Recherche et de l’Innovation – France (APAFIS# 19272 ).All non human-primate procedures were conducted according to European guidelines for animal care and use for scientific purposes (Directive 63-2010, ”Journal Officiel des Communautés Européennes”, L276, September 22, 2010) and according to CEA institutional guidelines. The study was approved by the local ethical committee under the number A18_080 and by the French Administration (APAFIS#20525-2019050616506478 v1)
- Published
- 2021
- Full Text
- View/download PDF
22. Targeting MICA/B with cytotoxic therapeutic antibodies leads to tumor control
- Author
-
Alix Joulin-Giet, Laurent Gauthier, Laurent Daniel, Veronika Liptakova, Younghoon Kim, Florence Lhospice, Eric Vivier, Nadia Anceriz, Nicolas Macagno, Mathieu Blery, Gwendoline Grondin, Delphine Bregeon, Laurent Pouyet, Ariane Morel, Sabrina Carpentier, Benjamin Rossi, Romain Remark, Yannis Morel, Sylvia Trichard, Manel Mrabet-Kraiem, Cécile Bonnafous, Aurélie Haffner, Ivan Perrot, François Romagné, Alexander Steinle, Carine Paturel, Emilie Narni-Mancinelli, Nicolas Viaud, and Stéphanie Cornen
- Subjects
medicine.drug_class ,medicine.medical_treatment ,Monoclonal antibody ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Cancer immunotherapy ,medicine ,Cytotoxic T cell ,030304 developmental biology ,Antibody-dependent cell-mediated cytotoxicity ,0303 health sciences ,cancer immunotherapy ,biology ,Chemistry ,Articles ,Immunotherapy ,NKG2D ,3. Good health ,stomatognathic diseases ,ADC ,MICA ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Antibody ,Research Article - Abstract
Background: MICA and MICB are tightly regulated stress-induced proteins that trigger the immune system by binding to the activating receptor NKG2D on cytotoxic lymphocytes. MICA and MICB are highly polymorphic molecules with prevalent expression on several types of solid tumors and limited expression in normal/healthy tissues, making them attractive targets for therapeutic intervention. Methods: We have generated a series of anti-MICA and MICB cross-reactive antibodies with the unique feature of binding to the most prevalent isoforms of both these molecules. Results: The anti-MICA and MICB antibody MICAB1, a human IgG1 Fc-engineered monoclonal antibody (mAb), displayed potent antibody-dependent cellular cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP) of MICA/B-expressing tumor cells in vitro. However, it showed insufficient efficiency against solid tumors in vivo, which prompted the development of antibody-drug conjugates (ADC). Indeed, optimal tumor control was achieved with MICAB1-ADC format in several solid tumor models, including patient-derived xenografts (PDX) and carcinogen-induced tumors in immunocompetent MICAgen transgenic mice. Conclusions: These data indicate that MICA and MICB are promising targets for cytotoxic immunotherapy.
- Published
- 2021
- Full Text
- View/download PDF
23. 1O Harnessing innate immunity in cancer therapies: The example of natural killer cell engagers
- Author
-
Yannis Morel, Agnès Represa, Cécile Bonnafous, Laurent Gauthier, M. Vetizou, Eric Vivier, Olivier Demaria, Ivan Perrot, Benjamin Rossi, A. Blanchard Alvarez, Guillaume Habif, Ariane Morel, and Stéphanie Cornen
- Subjects
Innate immune system ,medicine.anatomical_structure ,Oncology ,business.industry ,Immunology ,medicine ,Cancer ,Hematology ,medicine.disease ,business ,Natural killer cell - Published
- 2021
- Full Text
- View/download PDF
24. Harnessing innate immunity in cancer therapy
- Author
-
Stéphanie Cornen, Olivier Demaria, Ruslan Medzhitov, Yannis Morel, Marc Daëron, Eric Vivier, Innate Pharma, Centre d'Immunologie de Marseille - Luminy (CIML), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Aix Marseille Université (AMU), Yale University [New Haven], Aix Marseille Université (AMU)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), and ANR-17-RHUS-0007,PIONEER,Precision Immuno-Oncology for advanced Non small cell lung cancer patients with PD-1 ICI Resistance(2017)
- Subjects
0301 basic medicine ,Bispecific antibody ,medicine.medical_treatment ,T-Lymphocytes ,Cancer therapy ,03 medical and health sciences ,0302 clinical medicine ,Immunity ,Neoplasms ,medicine ,Animals ,Humans ,Multidisciplinary ,Innate immune system ,Antitumor immunity ,business.industry ,Cancer ,Immunotherapy ,medicine.disease ,Chimeric antigen receptor ,Immunity, Innate ,3. Good health ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer research ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,business - Abstract
New therapies that promote antitumour immunity have been recently developed. Most of these immunomodulatory approaches have focused on enhancing T-cell responses, either by targeting inhibitory pathways with immune checkpoint inhibitors, or by targeting activating pathways, as with chimeric antigen receptor T cells or bispecific antibodies. Although these therapies have led to unprecedented successes, only a minority of patients with cancer benefit from these treatments, highlighting the need to identify new cells and molecules that could be exploited in the next generation of immunotherapy. Given the crucial role of innate immune responses in immunity, harnessing these responses opens up new possibilities for long-lasting, multilayered tumour control.
- Published
- 2019
- Full Text
- View/download PDF
25. 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
26. Multifunctional Natural Killer Cell Engagers Targeting NKp46 Trigger Protective Tumor Immunity
- Author
-
Béatrice Amigues, Frédéric Bosco, Hélène Rispaud-Blanc, Gwendoline Grondin, Romain Remark, Guillaume Habif, Ariane Morel, Stéphanie Cornen, Flavien Caraguel, Nadia Anceriz, Laurent Gauthier, Emilie Narni-Mancinelli, Alain Roussel, Cédric Cesari, Franceline Guillot, Audrey Blanchard-Alvarez, Melody Sapet, Sylvia Trichard, Yannis Morel, Benjamin Rossi, Eric Vivier, François Romagné, Sandrine Arrufat, 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), Innate Pharma, Architecture et fonction des macromolécules biologiques (AFMB), Institut National de la Recherche Agronomique (INRA)-Aix Marseille Université (AMU)-Centre National de la Recherche Scientifique (CNRS), Institut Curie, Centre d'Immunologie de Marseille - Luminy (CIML), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Aix Marseille Université (AMU), Stabilité génétique, Cellules Souches et Radiations (SCSR (U_967)), Université Paris-Sud - Paris 11 (UP11)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris Cité (UPCité), Institut Curie [Paris], Aix Marseille Université (AMU)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), ANR-17-RHUS-0007,PIONEER,Precision Immuno-Oncology for advanced Non small cell lung cancer patients with PD-1 ICI Resistance(2017), 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), Université Paris-Sud - Paris 11 (UP11)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Paris (UP), and Centre National de la Recherche Scientifique (CNRS)-Aix Marseille Université (AMU)-Institut National de la Recherche Agronomique (INRA)
- Subjects
Cytotoxicity, Immunologic ,medicine.drug_class ,medicine.medical_treatment ,Biology ,Monoclonal antibody ,General Biochemistry, Genetics and Molecular Biology ,Natural killer cell ,Mice ,03 medical and health sciences ,Antineoplastic Agents, Immunological ,0302 clinical medicine ,Immune system ,Cancer immunotherapy ,Immunity ,Antibodies, Bispecific ,medicine ,Animals ,Antigens, Ly ,Humans ,030304 developmental biology ,0303 health sciences ,Natural Cytotoxicity Triggering Receptor 1 ,Neoplasms, Experimental ,Tumor antigen ,3. Good health ,Killer Cells, Natural ,medicine.anatomical_structure ,Immunoglobulin G ,Cancer cell ,Cancer research ,biology.protein ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,Antibody ,030217 neurology & neurosurgery - Abstract
Summary Over the last decade, various new therapies have been developed to promote anti-tumor immunity. Despite interesting clinical results in hematological malignancies, the development of bispecific killer-cell-engager antibody formats directed against tumor cells and stimulating anti-tumor T cell immunity has proved challenging, mostly due to toxicity problems. We report here the generation of trifunctional natural killer (NK) cell engagers (NKCEs), targeting two activating receptors, NKp46 and CD16, on NK cells and a tumor antigen on cancer cells. Trifunctional NKCEs were more potent in vitro than clinical therapeutic antibodies targeting the same tumor antigen. They had similar in vivo pharmacokinetics to full IgG antibodies and no off-target effects and efficiently controlled tumor growth in mouse models of solid and invasive tumors. Trifunctional NKCEs thus constitute a new generation of molecules for fighting cancer. Video Abstract
- Published
- 2019
- Full Text
- View/download PDF
27. 31st Annual Meeting and Associated Programs of the Society for Immunotherapy of Cancer (SITC 2016): part two
- Author
-
Casey Ager, Matthew Reilley, Courtney Nicholas, Todd Bartkowiak, Ashvin Jaiswal, Michael Curran, Tina C. Albershardt, Anshika Bajaj, Jacob F. Archer, Rebecca S. Reeves, Lisa Y. Ngo, Peter Berglund, Jan ter Meulen, Caroline Denis, Hormas Ghadially, Thomas Arnoux, Fabien Chanuc, Nicolas Fuseri, Robert W. Wilkinson, Nicolai Wagtmann, Yannis Morel, Pascale Andre, Michael B. Atkins, Matteo S. Carlino, Antoni Ribas, John A. Thompson, Toni K. Choueiri, F. Stephen Hodi, Wen-Jen Hwu, David F. McDermott, Victoria Atkinson, Jonathan S. Cebon, Bernie Fitzharris, Michael B. Jameson, Catriona McNeil, Andrew G. Hill, Eric Mangin, Malidi Ahamadi, Marianne van Vugt, Mariëlle van Zutphen, Nageatte Ibrahim, Georgina V. Long, Robyn Gartrell, Zoe Blake, Ines Simoes, Yichun Fu, Takuro Saito, Yingzhi Qian, Yan Lu, Yvonne M. Saenger, Sadna Budhu, Olivier De Henau, Roberta Zappasodi, Kyle Schlunegger, Bruce Freimark, Jeff Hutchins, Christopher A. Barker, Jedd D. Wolchok, Taha Merghoub, Elena Burova, Omaira Allbritton, Peter Hong, Jie Dai, Jerry Pei, Matt Liu, Joel Kantrowitz, Venus Lai, William Poueymirou, Douglas MacDonald, Ella Ioffe, Markus Mohrs, William Olson, Gavin Thurston, Cristian Capasso, Federica Frascaro, Sara Carpi, Siri Tähtinen, Sara Feola, Manlio Fusciello, Karita Peltonen, Beatriz Martins, Madeleine Sjöberg, Sari Pesonen, Tuuli Ranki, Lukasz Kyruk, Erkko Ylösmäki, Vincenzo Cerullo, Fabio Cerignoli, Biao Xi, Garret Guenther, Naichen Yu, Lincoln Muir, Leyna Zhao, Yama Abassi, Víctor Cervera-Carrascón, Mikko Siurala, João Santos, Riikka Havunen, Suvi Parviainen, Akseli Hemminki, Angus Dalgleish, Satvinder Mudan, Mark DeBenedette, Ana Plachco, Alicia Gamble, Elizabeth W. Grogan, John Krisko, Irina Tcherepanova, Charles Nicolette, Pooja Dhupkar, Ling Yu, Eugenie S. Kleinerman, Nancy Gordon, Italia Grenga, Lauren Lepone, Sofia Gameiro, Karin M. Knudson, Massimo Fantini, Kwong Tsang, James Hodge, Renee Donahue, Jeffrey Schlom, Elizabeth Evans, Holm Bussler, Crystal Mallow, Christine Reilly, Sebold Torno, Maria Scrivens, Cathie Foster, Alan Howell, Leslie Balch, Alyssa Knapp, John E. Leonard, Mark Paris, Terry Fisher, Siwen Hu-Lieskovan, Ernest Smith, Maurice Zauderer, William Fogler, Marilyn Franklin, Matt Thayer, Dan Saims, John L. Magnani, Jian Gong, Michael Gray, George Fromm, Suresh de Silva, Louise Giffin, Xin Xu, Jason Rose, Taylor H. Schreiber, Sofia R. Gameiro, Paul E. Clavijo, Clint T. Allen, James W. Hodge, Kwong Y. Tsang, Jane Grogan, Nicholas Manieri, Eugene Chiang, Patrick Caplazi, Mahesh Yadav, Patrick Hagner, Hsiling Chiu, Michelle Waldman, Anke Klippel, Anjan Thakurta, Michael Pourdehnad, Anita Gandhi, Ian Henrich, Laura Quick, Rob Young, Margaret Chou, Andrew Hotson, Stephen Willingham, Po Ho, Carmen Choy, Ginna Laport, Ian McCaffery, Richard Miller, Kimberly A. Tipton, Kenneth R. Wong, Victoria Singson, Chihunt Wong, Chanty Chan, Yuanhiu Huang, Shouchun Liu, Jennifer H. Richardson, W. Michael Kavanaugh, James West, Bryan A. Irving, Ritika Jaini, Matthew Loya, Charis Eng, Melissa L. Johnson, Alex A. Adjei, Mateusz Opyrchal, Suresh Ramalingam, Pasi A. Janne, George Dominguez, Dmitry Gabrilovich, Laura de Leon, Jeannette Hasapidis, Scott J. Diede, Peter Ordentlich, Scott Cruickshank, Michael L. Meyers, Matthew D. Hellmann, Pawel Kalinski, Amer Zureikat, Robert Edwards, Ravi Muthuswamy, Nataša Obermajer, Julie Urban, Lisa H. Butterfield, William Gooding, Herbert Zeh, David Bartlett, Olga Zubkova, Larissa Agapova, Marina Kapralova, Liudmila Krasovskaia, Armen Ovsepyan, Maxim Lykov, Artem Eremeev, Vladimir Bokovanov, Olga Grigoryeva, Andrey Karpov, Sergey Ruchko, Alexandr Shuster, Danny N. Khalil, Luis Felipe Campesato, Yanyun Li, Adam S. Lazorchak, Troy D. Patterson, Yueyun Ding, Pottayil Sasikumar, Naremaddepalli Sudarshan, Nagaraj Gowda, Raghuveer Ramachandra, Dodheri Samiulla, Sanjeev Giri, Rajesh Eswarappa, Murali Ramachandra, David Tuck, Timothy Wyant, Jasmin Leshem, Xiu-fen Liu, Tapan Bera, Masaki Terabe, Birgit Bossenmaier, Gerhard Niederfellner, Yoram Reiter, Ira Pastan, Leiming Xia, Yang Xia, Yangyang Hu, Yi Wang, Yangyi Bao, Fu Dai, Shiang Huang, Elaine Hurt, Robert E. Hollingsworth, Lawrence G. Lum, Alfred E. Chang, Max S. Wicha, Qiao Li, Thomas Mace, Neil Makhijani, Erin Talbert, Gregory Young, Denis Guttridge, Darwin Conwell, Gregory B. Lesinski, Rodney JM Macedo Gonzales, Austin P. Huffman, Ximi K. Wang, Ran Reshef, Andy MacKinnon, Jason Chen, Matt Gross, Gisele Marguier, Peter Shwonek, Natalija Sotirovska, Susanne Steggerda, Francesco Parlati, Amani Makkouk, Mark K. Bennett, Ethan Emberley, Tony Huang, Weiqun Li, Silinda Neou, Alison Pan, Jing Zhang, Winter Zhang, Netonia Marshall, Thomas U. Marron, Judith Agudo, Brian Brown, Joshua Brody, Christopher McQuinn, Matthew Farren, Hannah Komar, Reena Shakya, Thomas Ludwug, Y. Maurice Morillon, Scott A. Hammond, John W. Greiner, Pulak R. Nath, Anthony L. Schwartz, Dragan Maric, David D. Roberts, Aung Naing, Kyriakos P. Papadopoulos, Karen A. Autio, Deborah J. Wong, Manish Patel, Gerald Falchook, Shubham Pant, Patrick A. Ott, Melinda Whiteside, Amita Patnaik, John Mumm, Filip Janku, Ivan Chan, Todd Bauer, Rivka Colen, Peter VanVlasselaer, Gail L. Brown, Nizar M. Tannir, Martin Oft, Jeffrey Infante, Evan Lipson, Ajay Gopal, Sattva S. Neelapu, Philippe Armand, Stephen Spurgeon, John P. Leonard, Rachel E. Sanborn, Ignacio Melero, Thomas F. Gajewski, Matthew Maurer, Serena Perna, Andres A. Gutierrez, Raphael Clynes, Priyam Mitra, Satyendra Suryawanshi, Douglas Gladstone, Margaret K. Callahan, James Crooks, Sheila Brown, Audrey Gauthier, Marc Hillairet de Boisferon, Andrew MacDonald, Laura Rosa Brunet, William T. Rothwell, Peter Bell, James M. Wilson, Fumi Sato-Kaneko, Shiyin Yao, Shannon S. Zhang, Dennis A. Carson, Cristina Guiducci, Robert L. Coffman, Kazutaka Kitaura, Takaji Matsutani, Ryuji Suzuki, Tomoko Hayashi, Ezra E. W. Cohen, David Schaer, Yanxia Li, Julie Dobkin, Michael Amatulli, Gerald Hall, Thompson Doman, Jason Manro, Frank Charles Dorsey, Lillian Sams, Rikke Holmgaard, Krishnadatt Persaud, Dale Ludwig, David Surguladze, John S. Kauh, Ruslan Novosiadly, Michael Kalos, Kyla Driscoll, Hardev Pandha, Christy Ralph, Kevin Harrington, Brendan Curti, Wallace Akerley, Sumati Gupta, Alan Melcher, David Mansfield, David R. Kaufman, Emmett Schmidt, Mark Grose, Bronwyn Davies, Roberta Karpathy, Darren Shafren, Katerina Shamalov, Cyrille Cohen, Naveen Sharma, James Allison, Tala Shekarian, Sandrine Valsesia-Wittmann, Christophe Caux, Aurelien Marabelle, Brian M. Slomovitz, Kathleen M. Moore, Hagop Youssoufian, Marshall Posner, Poonam Tewary, Alan D. Brooks, Ya-Ming Xu, Kithsiri Wijeratne, Leslie A. A. Gunatilaka, Thomas J. Sayers, John P. Vasilakos, Tesha Alston, Simon Dovedi, James Elvecrog, Iwen Grigsby, Ronald Herbst, Karen Johnson, Craig Moeckly, Stefanie Mullins, Kristen Siebenaler, Julius SternJohn, Ashenafi Tilahun, Mark A. Tomai, Katharina Vogel, Eveline E. Vietsch, Anton Wellstein, Martin Wythes, Stefano Crosignani, Joseph Tumang, Shilpa Alekar, Patrick Bingham, Sandra Cauwenberghs, Jenny Chaplin, Deepak Dalvie, Sofie Denies, Coraline De Maeseneire, JunLi Feng, Kim Frederix, Samantha Greasley, Jie Guo, James Hardwick, Stephen Kaiser, Katti Jessen, Erick Kindt, Marie-Claire Letellier, Wenlin Li, Karen Maegley, Reece Marillier, Nichol Miller, Brion Murray, Romain Pirson, Julie Preillon, Virginie Rabolli, Chad Ray, Kevin Ryan, Stephanie Scales, Jay Srirangam, Jim Solowiej, Al Stewart, Nicole Streiner, Vince Torti, Konstantinos Tsaparikos, Xianxian Zheng, Gregory Driessens, Bruno Gomes, Manfred Kraus, Chunxiao Xu, Yanping Zhang, Giorgio Kradjian, Guozhong Qin, Jin Qi, Xiaomei Xu, Bo Marelli, Huakui Yu, Wilson Guzman, Rober Tighe, Rachel Salazar, Kin-Ming Lo, Jessie English, Laszlo Radvanyi, Yan Lan, Michael Postow, Yasin Senbabaoglu, Billel Gasmi, Hong Zhong, Cailian Liu, Daniel Hirschhorhn-Cymerman, Yuanyuan Zha, Gregory Malnassy, Noreen Fulton, Jae-Hyun Park, Wendy Stock, Yusuke Nakamura, Hongtao Liu, Xiaoming Ju, Rachelle Kosoff, Kimberly Ramos, Brandon Coder, Robert Petit, Michael Princiotta, Kyle Perry, Jun Zou, Ainhoa Arina, Christian Fernandez, Wenxin Zheng, Michael A. Beckett, Helena J. Mauceri, Yang-Xin Fu, Ralph R. Weichselbaum, Whitney Lewis, Yanyan Han, Yeting Wu, Chou Yang, Jing Huang, Dongyun Wu, Jin Li, Xiaoling Liang, Xiangjun Zhou, Jinlin Hou, Raffit Hassan, Thierry Jahan, Scott J. Antonia, Hedy L. Kindler, Evan W. Alley, Somayeh Honarmand, Weiqun Liu, Meredith L. Leong, Chan C. Whiting, Nitya Nair, Amanda Enstrom, Edward E. Lemmens, Takahiro Tsujikawa, Sushil Kumar, Lisa M. Coussens, Aimee L. Murphy, Dirk G. Brockstedt, Sven D. Koch, Martin Sebastian, Christian Weiss, Martin Früh, Miklos Pless, Richard Cathomas, Wolfgang Hilbe, Georg Pall, Thomas Wehler, Jürgen Alt, Helge Bischoff, Michael Geissler, Frank Griesinger, Jens Kollmeier, Alexandros Papachristofilou, Fatma Doener, Mariola Fotin-Mleczek, Madeleine Hipp, Henoch S. Hong, Karl-Josef Kallen, Ute Klinkhardt, Claudia Stosnach, Birgit Scheel, Andreas Schroeder, Tobias Seibel, Ulrike Gnad-Vogt, Alfred Zippelius, Ha-Ram Park, Yong-Oon Ahn, Tae Min Kim, Soyeon Kim, Seulki Kim, Yu Soo Lee, Bhumsuk Keam, Dong-Wan Kim, Dae Seog Heo, Shari Pilon-Thomas, Amy Weber, Jennifer Morse, Krithika Kodumudi, Hao Liu, John Mullinax, Amod A. Sarnaik, Luke Pike, Andrew Bang, Tracy Balboni, Allison Taylor, Alexander Spektor, Tyler Wilhite, Monica Krishnan, Daniel Cagney, Brian Alexander, Ayal Aizer, Elizabeth Buchbinder, Mark Awad, Leena Ghandi, Jonathan Schoenfeld, Elizabeth Lessey-Morillon, Lisa Ridnour, Neil H. Segal, Manish Sharma, Dung T. Le, Robert L. Ferris, Andrew D. Zelenetz, Ronald Levy, Izidore S. Lossos, Caron Jacobson, Radhakrishnan Ramchandren, John Godwin, A. Dimitrios Colevas, Roland Meier, Suba Krishnan, Xuemin Gu, Jaclyn Neely, John Timmerman, Claire I. Vanpouille-Box, Silvia C. Formenti, Sandra Demaria, Erik Wennerberg, Aranzazu Mediero, Bruce N. Cronstein, Michael P. Gustafson, AriCeli DiCostanzo, Courtney Wheatley, Chul-Ho Kim, Svetlana Bornschlegl, Dennis A. Gastineau, Bruce D. Johnson, Allan B. Dietz, Cameron MacDonald, Mark Bucsek, Guanxi Qiao, Bonnie Hylander, Elizabeth Repasky, William J. Turbitt, Yitong Xu, Andrea Mastro, Connie J. Rogers, Sita Withers, Ziming Wang, Lam T. Khuat, Cordelia Dunai, Bruce R. Blazar, Dan Longo, Robert Rebhun, Steven K. Grossenbacher, Arta Monjazeb, William J. Murphy, Scott Rowlinson, Giulia Agnello, Susan Alters, David Lowe, Nicole Scharping, Ashley V. Menk, Ryan Whetstone, Xue Zeng, Greg M. Delgoffe, Patricia M. Santos, Jian Shi, Greg Delgoffe, Misako Nagasaka, Ammar Sukari, Miranda Byrne-Steele, Wenjing Pan, Xiaohong Hou, Brittany Brown, Mary Eisenhower, Jian Han, Natalie Collins, Robert Manguso, Hans Pope, Yashaswi Shrestha, Jesse Boehm, W. Nicholas Haining, Kyle R. Cron, Ayelet Sivan, Keston Aquino-Michaels, Marco Orecchioni, Davide Bedognetti, Wouter Hendrickx, Claudia Fuoco, Filomena Spada, Francesco Sgarrella, Gianni Cesareni, Francesco Marincola, Kostas Kostarelos, Alberto Bianco, Lucia Delogu, Jessica Roelands, Sabri Boughorbel, Julie Decock, Scott Presnell, Ena Wang, Franco M. Marincola, Peter Kuppen, Michele Ceccarelli, Darawan Rinchai, Damien Chaussabel, Lance Miller, Andrew Nguyen, J. Zachary Sanborn, Charles Vaske, Shahrooz Rabizadeh, Kayvan Niazi, Steven Benz, Shashank Patel, Nicholas Restifo, James White, Sam Angiuoli, Mark Sausen, Sian Jones, Maria Sevdali, John Simmons, Victor Velculescu, Luis Diaz, Theresa Zhang, Jennifer S. Sims, Sunjay M. Barton, Angela Kadenhe-Chiweshe, Filemon Dela Cruz, Andrew T. Turk, Christopher F. Mazzeo, Andrew L. Kung, Jeffrey N. Bruce, Darrell J. Yamashiro, Eileen P. Connolly, Jason Baird, Marka Crittenden, David Friedman, Hong Xiao, Rom Leidner, Bryan Bell, Kristina Young, Michael Gough, Zhen Bian, Koby Kidder, Yuan Liu, Emily Curran, Xiufen Chen, Leticia P. Corrales, Justin Kline, Ethan G. Aguilar, Jennifer Guerriero, Alaba Sotayo, Holly Ponichtera, Alexandra Pourzia, Sara Schad, Ruben Carrasco, Suzan Lazo, Roderick Bronson, Anthony Letai, Richard S. Kornbluth, Sachin Gupta, James Termini, Elizabeth Guirado, Geoffrey W. Stone, Christina Meyer, Laura Helming, Nicholas Wilson, Robert Hofmeister, Natalie J. Neubert, Laure Tillé, David Barras, Charlotte Soneson, Petra Baumgaertner, Donata Rimoldi, David Gfeller, Mauro Delorenzi, Silvia A. Fuertes Marraco, Daniel E. Speiser, Tara S. Abraham, Bo Xiang, Michael S. Magee, Scott A. Waldman, Adam E. Snook, Wojciech Blogowski, Ewa Zuba-Surma, Marta Budkowska, Daria Salata, Barbara Dolegowska, Teresa Starzynska, Leo Chan, Srinivas Somanchi, Kelsey McCulley, Dean Lee, Nico Buettner, Feng Shi, Paisley T. Myers, Stuart Curbishley, Sarah A. Penny, Lora Steadman, David Millar, Ellen Speers, Nicola Ruth, Gabriel Wong, Robert Thimme, David Adams, Mark Cobbold, Remy Thomas, Mariam Al-Muftah, Michael KK Wong, Michael Morse, Joseph I. Clark, Howard L. Kaufman, Gregory A. Daniels, Hong Hua, Tharak Rao, Janice P. Dutcher, Kai Kang, Yogen Saunthararajah, Vamsidhar Velcheti, Vikas Kumar, Firoz Anwar, Amita Verma, Zinal Chheda, Gary Kohanbash, John Sidney, Kaori Okada, Shruti Shrivastav, Diego A. Carrera, Shuming Liu, Naznin Jahan, Sabine Mueller, Ian F. Pollack, Angel M. Carcaboso, Alessandro Sette, Yafei Hou, Hideho Okada, Jessica J. Field, Weiping Zeng, Vincent FS Shih, Che-Leung Law, Peter D. Senter, Shyra J. Gardai, Nicole M. Okeley, Jennifer G. Abelin, Abu Z. Saeed, Stacy A. Malaker, Jeffrey Shabanowitz, Stephen T. Ward, Donald F. Hunt, Pam Profusek, Laura Wood, Dale Shepard, Petros Grivas, Kerstin Kapp, Barbara Volz, Detlef Oswald, Burghardt Wittig, Manuel Schmidt, Julian P. Sefrin, Lars Hillringhaus, Valeria Lifke, Alexander Lifke, Anna Skaletskaya, Jose Ponte, Thomas Chittenden, Yulius Setiady, Eva Sivado, Vincent Thomas, Meddy El Alaoui, Sébastien Papot, Charles Dumontet, Mike Dyson, John McCafferty, Said El Alaoui, Praveen K. Bommareddy, Andrew Zloza, Frederick Kohlhapp, Ann W. Silk, Sachin Jhawar, Tomas Paneque, Jenna Newman, Pedro Beltran, Felicia Cao, Bang-Xing Hong, Tania Rodriguez-Cruz, Xiao-Tong Song, Stephen Gottschalk, Hugo Calderon, Sam Illingworth, Alice Brown, Kerry Fisher, Len Seymour, Brian Champion, Emma Eriksson, Jessica Wenthe, Ann-Charlotte Hellström, Gabriella Paul-Wetterberg, Angelica Loskog, Ioanna Milenova, Magnus Ståhle, Justyna Jarblad-Leja, Gustav Ullenhag, Anna Dimberg, Rafael Moreno, Ramon Alemany, Sharad Goyal, Ann Silk, Janice Mehnert, Nashat Gabrail, Jennifer Bryan, Daniel Medina, Leah Mitchell, Kader Yagiz, Fernando Lopez, Daniel Mendoza, Anthony Munday, Harry Gruber, Douglas Jolly, Steven Fuhrmann, Sasa Radoja, Wei Tan, Aldo Pourchet, Alan Frey, Ian Mohr, Matthew Mulvey, Robert H. I. Andtbacka, Merrick Ross, Sanjiv Agarwala, Kenneth Grossmann, Matthew Taylor, John Vetto, Rogerio Neves, Adil Daud, Hung Khong, Stephanie M. Meek, Richard Ungerleider, Scott Welden, Maki Tanaka, Matthew Williams, Sigrun Hallmeyer, Bernard Fox, Zipei Feng, Christopher Paustian, Carlo Bifulco, Sadia Zafar, Otto Hemminki, Simona Bramante, Lotta Vassilev, Hongjie Wang, Andre Lieber, Silvio Hemmi, Tanja de Gruijl, Anna Kanerva, Tameem Ansari, Srividya Sundararaman, Diana Roen, Paul Lehmann, Anja C. Bloom, Lewis H. Bender, Ian B. Walters, Jay A. Berzofsky, Fanny Chapelin, Eric T. Ahrens, Jeff DeFalco, Michael Harbell, Amy Manning-Bog, Alexander Scholz, Danhui Zhang, Gilson Baia, Yann Chong Tan, Jeremy Sokolove, Dongkyoon Kim, Kevin Williamson, Xiaomu Chen, Jillian Colrain, Gregg Espiritu Santo, Ngan Nguyen, Wayne Volkmuth, Norman Greenberg, William Robinson, Daniel Emerling, Charles G. Drake, Daniel P. Petrylak, Emmanuel S. Antonarakis, Adam S. Kibel, Nancy N. Chang, Tuyen Vu, Dwayne Campogan, Heather Haynes, James B. Trager, Nadeem A. Sheikh, David I. Quinn, Peter Kirk, Murali Addepalli, Thomas Chang, Ping Zhang, Marina Konakova, Katsunobu Hagihara, Steven Pai, Laurie VanderVeen, Palakshi Obalapur, Peiwen Kuo, Phi Quach, Lawrence Fong, Deborah H. Charych, Jonathan Zalevsky, John L. Langowski, Yolanda Kirksey, Ravi Nutakki, Shalini Kolarkar, Rhoneil Pena, Ute Hoch, Stephen K. Doberstein, John Cha, Zach Mallon, Myra Perez, Amanda McDaniel, Snjezana Anand, Darrin Uecker, Richard Nuccitelli, Eva Wieckowski, Ravikumar Muthuswamy, Roshni Ravindranathan, Ariana N. Renrick, Menaka Thounaojam, Portia Thomas, Samuel Pellom, Anil Shanker, Duafalia Dudimah, Alan Brooks, Yu-Lin Su, Tomasz Adamus, Qifang Zhang, Sergey Nechaev, Marcin Kortylewski, Spencer Wei, Clark Anderson, Chad Tang, Jonathan Schoenhals, Efrosini Tsouko, John Heymach, Patricia de Groot, Joe Chang, Kenneth R. Hess, Adi Diab, Padmanee Sharma, David Hong, James Welsh, Andrea J. Parsons, Jardin Leleux, Stephane Ascarateil, Marie Eve Koziol, Dina Bai, Peihong Dai, Weiyi Wang, Ning Yang, Stewart Shuman, Liang Deng, Patrick Dillon, Gina Petroni, David Brenin, Kim Bullock, Walter Olson, Mark E. Smolkin, Kelly Smith, Carmel Nail, Craig L. Slingluff, Meenu Sharma, Faisal Fa’ak, Louise Janssen, Hiep Khong, Zhilan Xiao, Yared Hailemichael, Manisha Singh, Christina Vianden, Willem W. Overwijk, Andrea Facciabene, Pierini Stefano, Fang Chongyung, Stavros Rafail, Michael Nielsen, Peter Vanderslice, Darren G. Woodside, Robert V. Market, Ronald J. Biediger, Upendra K. Marathi, Kevin Hollevoet, Nick Geukens, Paul Declerck, Nathalie Joly, Laura McIntosh, Eustache Paramithiotis, Magnus Rizell, Malin Sternby, Bengt Andersson, Alex Karlsson-Parra, Rui Kuai, Lukasz Ochyl, Anna Schwendeman, James Moon, Weiwen Deng, Thomas E. Hudson, Bill Hanson, Chris S. Rae, Joel Burrill, Justin Skoble, George Katibah, Michele deVries, Peter Lauer, Thomas W. Dubensky, Xin Chen, Li Zhou, Xiubao Ren, Charu Aggarwal, Drishty Mangrolia, Roger Cohen, Gregory Weinstein, Matthew Morrow, Joshua Bauml, Kim Kraynyak, Jean Boyer, Jian Yan, Jessica Lee, Laurent Humeau, Sandra Oyola, Susan Duff, David Weiner, Zane Yang, Mark Bagarazzi, Douglas G. McNeel, Jens Eickhoff, Robert Jeraj, Mary Jane Staab, Jane Straus, Brian Rekoske, Glenn Liu, Marit Melssen, William Grosh, Nikole Varhegyi, Nadejda Galeassi, Donna H. Deacon, Elizabeth Gaughan, Maurizio Ghisoli, Minal Barve, Robert Mennel, Gladice Wallraven, Luisa Manning, Neil Senzer, John Nemunaitis, Masahiro Ogasawara, Shuichi Ota, Kaitlin M. Peace, Diane F. Hale, Timothy J. Vreeland, Doreen O. Jackson, John S. Berry, Alfred F. Trappey, Garth S. Herbert, Guy T. Clifton, Mark O. Hardin, Anne Toms, Na Qiao, Jennifer Litton, George E. Peoples, Elizabeth A. Mittendorf, Lila Ghamsari, Emilio Flano, Judy Jacques, Biao Liu, Jonathan Havel, Vladimir Makarov, Timothy A. Chan, Jessica B. Flechtner, John Facciponte, Stefano Ugel, Francesco De Sanctis, George Coukos, Sébastien Paris, Agnes Pottier, Laurent Levy, Bo Lu, Federica Cappuccini, Emily Pollock, Richard Bryant, Freddie Hamdy, Adrian Hill, Irina Redchenko, Hussein Sultan, Takumi Kumai, Valentyna Fesenkova, Esteban Celis, Ingrid Fernando, Claudia Palena, Justin M. David, Elizabeth Gabitzsch, Frank Jones, James L. Gulley, Mireia Uribe Herranz, Hiroshi Wada, Atsushi Shimizu, Toshihiro Osada, Satoshi Fukaya, Eiji Sasaki, Milad Abolhalaj, David Askmyr, Kristina Lundberg, Ann-Sofie Albrekt, Lennart Greiff, Malin Lindstedt, Dallas B. Flies, Tomoe Higuchi, Wojciech Ornatowski, Jaryse Harris, Sarah F. Adams, Todd Aguilera, Marjan Rafat, Laura Castellini, Hussein Shehade, Mihalis Kariolis, Dadi Jang, Rie vonEbyen, Edward Graves, Lesley Ellies, Erinn Rankin, Albert Koong, Amato Giaccia, Reham Ajina, Shangzi Wang, Jill Smith, Mariaelena Pierobon, Sandra Jablonski, Emanuel Petricoin, Louis M. Weiner, Lorcan Sherry, John Waller, Mark Anderson, Alison Bigley, Chantale Bernatchez, Cara Haymaker, Harriet Kluger, Michael Tetzlaff, Natalie Jackson, Ivan Gergel, Mary Tagliaferri, Patrick Hwu, Mario Snzol, Michael Hurwitz, Theresa Barberi, Allison Martin, Rahul Suresh, David Barakat, Sarah Harris-Bookman, Charles Drake, Alan Friedman, Sara Berkey, Stephanie Downs-Canner, Robert P. Edwards, Tyler Curiel, Kunle Odunsi, Tullia C. Bruno, Brandon Moore, Olivia Squalls, Peggy Ebner, Katherine Waugh, John Mitchell, Wilbur Franklin, Daniel Merrick, Martin McCarter, Brent Palmer, Jeffrey Kern, Dario Vignali, Jill Slansky, Anissa S. H. Chan, Xiaohong Qiu, Kathryn Fraser, Adria Jonas, Nadine Ottoson, Keith Gordon, Takashi O. Kangas, Steven Leonardo, Kathleen Ertelt, Richard Walsh, Mark Uhlik, Jeremy Graff, Nandita Bose, Ravi Gupta, Nitin Mandloi, Kiran Paul, Ashwini Patil, Rekha Sathian, Aparna Mohan, Malini Manoharan, Amitabha Chaudhuri, Yu Chen, Jing Lin, Yun-bin Ye, Chun-wei Xu, Gang Chen, Zeng-qing Guo, Andrey Komarov, Alex Chenchik, Michael Makhanov, Costa Frangou, Yi Zheng, Carla Coltharp, Darryn Unfricht, Ryan Dilworth, Leticia Fridman, Linying Liu, Milind Rajopadhye, Peter Miller, Fernando Concha-Benavente, Julie Bauman, Sumita Trivedi, Raghvendra Srivastava, James Ohr, Dwight Heron, Uma Duvvuri, Seungwon Kim, Heather Torrey, Toshi Mera, Yoshiaki Okubo, Eva Vanamee, Rosemary Foster, Denise Faustman, Edward Stack, Daisuke Izaki, Kristen Beck, Dan Tong Jia, Paul Armenta, Ashley White-Stern, Douglas Marks, Bret Taback, Basil Horst, Laura Hix Glickman, David B. Kanne, Kelsey S. Gauthier, Anthony L. Desbien, Brian Francica, Justin L. Leong, Leonard Sung, Ken Metchette, Shailaja Kasibhatla, Anne Marie Pferdekamper, Lianxing Zheng, Charles Cho, Yan Feng, Jeffery M. McKenna, John Tallarico, Steven Bender, Chudi Ndubaku, Sarah M. McWhirter, Elena Gonzalez Gugel, Charles J. M. Bell, Adiel Munk, Luciana Muniz, Nina Bhardwaj, Fei Zhao, Kathy Evans, Christine Xiao, Alisha Holtzhausen, Brent A. Hanks, Nathalie Scholler, Catherine Yin, Pien Van der Meijs, Andrew M. Prantner, Cecile M. Krejsa, Leia Smith, Brian Johnson, Daniel Branstetter, Paul L. Stein, Juan C. Jaen, Joanne BL Tan, Ada Chen, Timothy Park, Jay P. Powers, Holly Sexton, Guifen Xu, Steve W. Young, Ulrike Schindler, Wentao Deng, David John Klinke, Hannah M. Komar, Gregory Serpa, Omar Elnaggar, Philip Hart, Carl Schmidt, Mary Dillhoff, Ming Jin, Michael C. Ostrowski, Madhuri Koti, Katrina Au, Nichole Peterson, Peter Truesdell, Gillian Reid-Schachter, Charles Graham, Andrew Craig, Julie-Ann Francis, Beatrix Kotlan, Timea Balatoni, Emil Farkas, Laszlo Toth, Mihaly Ujhelyi, Akos Savolt, Zoltan Doleschall, Szabolcs Horvath, Klara Eles, Judit Olasz, Orsolya Csuka, Miklos Kasler, Gabriella Liszkay, Eytan Barnea, Collin Blakely, Patrick Flynn, Reid Goodman, Raphael Bueno, David Sugarbaker, David Jablons, V. Courtney Broaddus, Brian West, Paul R. Kunk, Joseph M. Obeid, Kevin Winters, Patcharin Pramoonjago, Edward B. Stelow, Todd W. Bauer, Osama E. Rahma, Adam Lamble, Yoko Kosaka, Fei Huang, Kate A. Saser, Homer Adams, Christina E. Tognon, Ted Laderas, Shannon McWeeney, Marc Loriaux, Jeffery W. Tyner, Brian J. Druker, Evan F. Lind, Zhuqing Liu, Shanhong Lu, Lawrence P. Kane, Gulidanna Shayan, Julia Femel, Ryan Lane, Jamie Booth, Amanda W. Lund, Anthony Rodriguez, Victor H. Engelhard, Alessandra Metelli, Bill X. Wu, Caroline W. Fugle, Rachidi Saleh, Shaoli Sun, Jennifer Wu, Bei Liu, Zihai Li, Zachary S. Morris, Emily I. Guy, Clinton Heinze, Jasdeep Kler, Monica M. Gressett, Lauryn R. Werner, Stephen D. Gillies, Alan J. Korman, Hans Loibner, Jacquelyn A. Hank, Alexander L. Rakhmilevich, Paul M. Harari, Paul M. Sondel, Erica Huelsmann, Joseph Broucek, Dorothee Brech, Tobias Straub, Martin Irmler, Johannes Beckers, Florian Buettner, Elke Schaeffeler, Matthias Schwab, Elfriede Noessner, Alison Wolfreys, Andre Da Costa, John Silva, Andrea Crosby, Ludovicus Staelens, Graham Craggs, Annick Cauvin, Sean Mason, Alison M. Paterson, Andrew C. Lake, Caroline M. Armet, Rachel W. O’Connor, Jonathan A. Hill, Emmanuel Normant, Ammar Adam, Detlev M. Biniszkiewicz, Scott C. Chappel, Vito J. Palombella, Pamela M. Holland, Annette Becker, Manmohan R. Leleti, Eric Newcomb, Joanne B. L. Tan, Suthee Rapisuwon, Arash Radfar, Kellie Gardner, Geoffrey Gibney, Michael Atkins, Keith R. Rennier, Robert Crowder, Ping Wang, Russell K. Pachynski, Rosa M. Santana Carrero, Sarai Rivas, Figen Beceren-Braun, Scott Anthony, Kimberly S. Schluns, Deepali Sawant, Maria Chikina, Hiroshi Yano, Creg Workman, Elise Salerno, Ileana Mauldin, Donna Deacon, Sofia Shea, Joel Pinczewski, Thomas Gajewski, Stefani Spranger, Brendan Horton, Akiko Suzuki, Pamela Leland, Bharat H. Joshi, Raj K. Puri, Randy F. Sweis, Riyue Bao, Jason Luke, Marie-Nicole Theodoraki, Frances-Mary Mogundo, Haejung Won, Dayson Moreira, Chan Gao, Xingli Zhao, Priyanka Duttagupta, Jeremy Jones, Massimo D’Apuzzo, and Sumanta Pal
- Subjects
0301 basic medicine ,Pharmacology ,Cancer Research ,medicine.medical_specialty ,business.industry ,medicine.medical_treatment ,Immunology ,Cancer ,Immunotherapy ,medicine.disease ,3. Good health ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,030220 oncology & carcinogenesis ,Family medicine ,Molecular Medicine ,Immunology and Allergy ,Medicine ,business - Abstract
O1 IL-15 primes an mTOR-regulated gene-expression program to prolong anti-tumor capacity of human natural killer cells #### Andreas Lundqvist1, Vincent van Hoef1, Xiaonan Zhang1, Erik Wennerberg2, Julie Lorent1, Kristina Witt1, Laia Masvidal Sanz1, Shuo Liang1, Shannon Murray3, Ola Larsson1
- Published
- 2016
- Full Text
- View/download PDF
28. TLR3 as a Biomarker for the Therapeutic Efficacy of Double-stranded RNA in Breast Cancer
- Author
-
Michel Pierres, Aicha Goubar, Laurence Zitvogel, Catherine Massacrier, Jean-Christophe Sabourin, Serge Lebecque, Rosa Conforti, Yannis Morel, Marie Pierre Chenard, Pedro Romero, Fabrice Andre, Bruno Salaun, Karine Chemin, Carine Asselin-Paturel, Clarisse Dubois, Donata Rimoldi, Ludwig Institute for Cancer Research Ltd., 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), Centre d'Investigation Clinique en Biotherapie des cancers (CIC 1428 , CBT 507 ), Institut Gustave Roussy (IGR)-Institut National de la Santé et de la Recherche Médicale (INSERM), School of Medicine, Université Paris-Sud - Paris 11 (UP11), Schering-Plough Laboratories for Immunological Research, Schering Plough Laboratories, Innate Pharma, Différenciation des cellules B, hémopathies, lymphoïdes et déficit de l'immunité humorale, Université Paris Diderot - Paris 7 (UPD7), Département de Pathologie, CHU Strasbourg-Hôpital de Hautepierre [Strasbourg], CHU Rouen, Normandie Université (NU), Université Claude Bernard Lyon 1 (UCBL), Université de Lyon, 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), Département de médecine oncologique [Gustave Roussy], Institut Gustave Roussy (IGR), Institut Gustave Roussy (IGR)-Institut Curie [Paris]-Institut National de la Santé et de la Recherche Médicale (INSERM), Institut National de la Santé et de la Recherche Médicale (INSERM)-Institut Gustave Roussy (IGR)-Université Paris-Sud - Paris 11 (UP11), and Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Aix Marseille Université (AMU)
- Subjects
Cancer Research ,viruses ,medicine.medical_treatment ,MESH: Antibodies, Monoclonal ,Mice ,0302 clinical medicine ,Antibody Specificity ,Multicenter Studies as Topic ,MESH: Animals ,Randomized Controlled Trials as Topic ,Mice, Inbred BALB C ,Antibodies, Monoclonal ,virus diseases ,hemic and immune systems ,MESH: Toll-Like Receptor 3 ,Immunohistochemistry ,3. Good health ,RNA silencing ,Oncology ,030220 oncology & carcinogenesis ,Monoclonal ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,Biomarker (medicine) ,Female ,Breast disease ,Adjuvant ,MESH: Mice, Inbred BALB C ,Antineoplastic Agents ,Breast Neoplasms ,chemical and pharmacologic phenomena ,03 medical and health sciences ,Breast cancer ,MESH: RNA, Double-Stranded ,Biomarkers, Tumor ,medicine ,Animals ,Humans ,MESH: Antibody Specificity ,MESH: Mice ,RNA, Double-Stranded ,Retrospective Studies ,MESH: Humans ,business.industry ,Cancer ,MESH: Retrospective Studies ,MESH: Immunohistochemistry ,medicine.disease ,Toll-Like Receptor 3 ,MESH: Randomized Controlled Trials as Topic ,MESH: Tumor Markers, Biological ,TLR3 ,Immunology ,MESH: Multicenter Studies as Topic ,Cancer research ,MESH: Antineoplastic Agents ,business ,MESH: Female ,MESH: Breast Neoplasms ,030215 immunology - Abstract
The discovery of a targeted therapeutic compound along with its companion predictive biomarker is a major goal of clinical development for a personalized anticancer therapy to date. Here we present evidence of the predictive value of TLR3 expression by tumor cells for the efficacy of Poly (A:U) dsRNA in 194 breast cancer patients enrolled in a randomized clinical trial. Adjuvant treatment with double-stranded RNA (dsRNA) was associated with a significant decrease in the risk of metastatic relapse in TLR3 positive but not in TLR3-negative breast cancers. Moreover, we show the functional relevance of TLR3 expression by human tumor cells for the antitumor effects mediated by dsRNA in several preclinical mouse models carried out in immunocompromised animals. These 2 independent lines of evidence relied upon the generation of a novel tool, an anti-TLR3 antibody (40F9.6) validated for routine detection of TLR3 expression on paraffin-embedded tissues. Altogether, these data suggest that dsRNA mediates its therapeutic effect through TLR3 expressed on tumor cells, and could therefore represent an effective targeted treatment in patients with TLR3-positive cancers. Cancer Res; 71(5); 1607–14. ©2011 AACR.
- Published
- 2011
- Full Text
- View/download PDF
29. Desirable cell death during anticancer chemotherapy
- Author
-
Yuting Ma, François Ghiringhelli, Rosa Conforti, Yannis Morel, Takahiro Yamazaki, Sylvie Rusakiewicz, Clara Locher, Laurence Zitvogel, Antoine Tesniere, Lionel Apetoh, Guido Kroemer, Laetitia Aymeric, and Jean-Philippe Girard
- Subjects
Toll-like receptor ,Chemotherapy ,General Neuroscience ,medicine.medical_treatment ,T cell ,Immunogenicity ,Immunotherapy ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Immune system ,medicine.anatomical_structure ,History and Philosophy of Science ,Immunology ,medicine ,Cytotoxic T cell ,Immunogenic cell death - Abstract
The concept of immunogenic chemotherapy that has recently emerged relies upon the capacity of a cytotoxic compound to trigger a cell-death modality. This modality elicits cross-priming by dendritic cells of tumor antigen-specific T cells that will contribute to the tumoricidal activity of the compound and protect the host against relapse. In contrast, most anticancer drugs elicit nonimmunogenic apoptosis that is not accompanied with an immunizing property. This review will discuss some molecular and metabolic changes required at the level of the tumor that must engage key pathways at the level of the host for the induction of Tc1 polarized–protective T cell responses during chemotherapy. We will summarize the immune adjuvants that can boost the immunogenicity of cell death to augment the efficacy of chemotherapy.
- Published
- 2010
- Full Text
- View/download PDF
30. 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
- Full Text
- View/download PDF
31. Abstract 2713: Preclinical development of first-in-class antibodies targeting Siglec-9 immune checkpoint for cancer immunotherapy
- Author
-
Olivier Benac, Stéphanie Cornen, Carine Paturel, Mélody Ors, Aude Le Roy, Laurent Gauthier, Marion Gaudin, Ivan Perrot, Hélène Rispaud Blanc, Caroline Soulas, Benjamin Rossi, Yannis Morel, and Stéphanie Chanteux
- Subjects
0301 basic medicine ,Cancer Research ,biology ,MHC class I antigen ,medicine.medical_treatment ,Sialic acid binding ,Acquired immune system ,Immune checkpoint ,Epitope ,03 medical and health sciences ,030104 developmental biology ,Oncology ,Cancer immunotherapy ,MHC class I ,biology.protein ,Cancer research ,medicine ,CD8 - Abstract
Siglec-9 is a MHC class I-independent inhibitory receptor expressed on NK and myeloid cells (including dendritic cells, monocytes and neutrophils). Its ligands are sialic acid-containing carbohydrates which are over-expressed on various tumor types compared to normal tissues (1). Sialylation of tumor cells is involved in tumor cell malignancy and is reported for decades as a mechanism of escape from immune surveillance (2). The loss of beta-2-microglobulin, an essential component of MHC class I antigen presentation, was recently described as a common mechanism of resistance to checkpoint blockade in clinical trials and revealed the need for MHC class I-independent therapies (3-4). Thus, Siglec-9-sialic acid interaction disruption may promote anti-tumor immunity independently of MHC class I expression by tumors. Here, we describe the discovery and characterization of first-in class anti-human Siglec-9 antibodies as new checkpoint blockade therapy in a wide range of cancers. Antibodies were discovered that efficiently block the interaction between Siglec-9 and its ligands. Epitope mapping revealed that antibodies bind to distinct epitopes on Siglec-9 near the sialic acid binding site. In vitro assays showed that they potently reverse inhibitory functions of Siglec-9 on NK cells leading to subsequent sialic acid-expressing tumor cell killing. Interestingly, Siglec-9 is enhanced on both CD4+ and CD8+ T cells from RCC, melanoma and NSCLC PBMC patients suggesting a putative additional role on adaptive immunity. Siglec-9 was also co-expressed with other inhibitory receptors on NK cells and combination with other immune checkpoint blockers in in vitro assays is ongoing. The antibodies displaying the most interesting features were successfully humanized. (1) Pathol Oncol Res. 2016 Jul;22(3):443-7 (2) Nature. 1968 Jun 29;218(5148):1254-5 (3) Nat Commun. 2017 Oct 26;8(1):1136 (4) N Engl J Med. 2016 Sep 1;375(9):819-29 Citation Format: Olivier Bénac, Marion Gaudin, Mélody Ors, Aude Le Roy, Hélène Rispaud Blanc, Caroline Soulas, Stéphanie Chanteux, Benjamin Rossi, Laurent Gauthier, Carine Paturel, Yannis Morel, Ivan Perrot, Stéphanie Cornen. Preclinical development of first-in-class antibodies targeting Siglec-9 immune checkpoint 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 2713.
- Published
- 2018
- Full Text
- View/download PDF
32. Nucleic acid agonists for Toll-like receptor 7 are defined by the presence of uridine ribonucleotides
- Author
-
Caetano Reis e Sousa, Shizuo Akira, Catherine Massacrier, Carine Paturel, Sandra S. Diebold, and Yannis Morel
- Subjects
Poly U ,Amino Acid Motifs ,Deoxyribonucleotides ,Immunology ,RNA-dependent RNA polymerase ,Endosomes ,Biology ,Mice ,Sense (molecular biology) ,Animals ,Humans ,Immunology and Allergy ,Uridine ,Cells, Cultured ,Mice, Knockout ,Membrane Glycoproteins ,virus diseases ,RNA ,Non-coding RNA ,Mice, Inbred C57BL ,RNA silencing ,Toll-Like Receptor 7 ,Biochemistry ,RNA editing ,Nucleic acid ,Cytokines ,RNA, Viral ,Small nuclear RNA - Abstract
Toll-like receptor 7 (TLR7) mediates innate responses by responding to viral RNA in endocytic compartments. However, the molecular pattern recognised by TLR7 and whether it differs between RNA of viral and self origin remains unclear. Here, we identify nucleic acids that act as TLR7 agonists for mouse and human cells. We show that uridine and ribose, the two defining features of RNA, are both necessary and sufficient for TLR7 stimulation, and that short single-stranded RNA (ssRNA) act as TLR7 agonists in a sequence-independent manner as long as they contain several uridines in close proximity. Consistent with the notion that TLR7 lacks specificity for sequence motifs, we show that it is triggered equally efficiently by viral or self RNA delivered to endosomes. Our results support the notion that TLR7 recognises uracil repeats in RNA and that it discriminates between viral and self ligands on the basis of endosomal accessibility rather than sequence.
- Published
- 2006
- Full Text
- View/download PDF
33. Phosphostim-Activated γδ T Cells Kill Autologous Metastatic Renal Cell Carcinoma
- Author
-
Salem Chouaib, Emilie Viey, Bernard Escudier, Yannis Morel, Gaëlle Fromont, Sylvie Da Rocha, and Anne Caignard
- Subjects
T cell ,Immunology ,chemical and pharmacologic phenomena ,Biology ,NKG2D ,Natural killer T cell ,Natural killer cell ,Interleukin 21 ,medicine.anatomical_structure ,medicine ,Cancer research ,Immunology and Allergy ,Cytotoxic T cell ,IL-2 receptor ,Antigen-presenting cell - Abstract
Metastatic renal cell carcinoma, inherently resistant to conventional treatments, is considered immunogenic. Indeed, partial responses are obtained after treatment with cytokines such as IL-2 or IFN-alpha, suggesting that the immune system may control the tumor growth. In this study, we have investigated the ability of the main subset of peripheral gammadelta lymphocytes, the Vgamma9Vdelta2-TCR T lymphocytes, to induce an effective cytotoxic response against autologous primary renal cell carcinoma lines. These gammadelta T cells were expanded ex vivo using a Vgamma9Vdelta2 agonist, a synthetic phosphoantigen called Phosphostim. From 11 of 15 patients, the peripheral Vgamma9Vdelta2 T cells were amplified in vitro by stimulating PBMCs with IL-2 and Phosphostim molecule. These expanded Vgamma9Vdelta2 T cells express activation markers and exhibit an effector/memory phenotype. They display a selective lytic potential toward autologous primary renal tumor cells and not against renal NC. The lytic activity involves the perforin-granzyme pathway and is mainly TCR and NKG2D receptor dependent. Furthermore, an increased expression of MHC class I-related molecule A or B proteins, known ligands of NKG2D, are detected on primary renal tumor cells. Interestingly, from 2 of the 11 positive cultures in response to Phosphostim, expanded-Vgamma9Vdelta2 T cells present an expression of killer cell Ig-like receptors, suggesting their prior recruitment in vivo. Unexpectedly, on serial frozen sections from three tumors, we observe a gammadelta lymphocyte infiltrate that was mainly composed of Vgamma9Vdelta2 T cells. These results outline that Vgamma9Vdelta2-TCR effectors may represent a promising approach for the treatment of metastatic renal cell carcinoma.
- Published
- 2005
- Full Text
- View/download PDF
34. LIGHT, a new TNF superfamily member, is essential for memory T helper cell-mediated activation of dendritic cells
- Author
-
Daniel Olive, Yannis Morel, Alemseged Truneh, and Régis Costello
- Subjects
CD4-Positive T-Lymphocytes ,Receptors, CCR7 ,Tumor Necrosis Factor Ligand Superfamily Member 14 ,CD40 Ligand ,Immunology ,Priming (immunology) ,chemical and pharmacologic phenomena ,Biology ,Interleukin 21 ,immune system diseases ,medicine ,Humans ,Immunology and Allergy ,Cytotoxic T cell ,IL-2 receptor ,Antigen-presenting cell ,Tumor Necrosis Factor-alpha ,ZAP70 ,Membrane Proteins ,hemic and immune systems ,Dendritic Cells ,Dendritic cell ,T helper cell ,Interleukin-12 ,Cell biology ,medicine.anatomical_structure ,Receptors, Chemokine ,Immunologic Memory - Abstract
LIGHT is a recently identified member of the TNF superfamily that is up-regulated on activated T cells and can cooperate with CD40L to condition DC for the priming of CTL. In this report, we show that an intracellular pool of LIGHT is found selectively in some CD45RA-CD27+CD4+ "central" memory T cells and to a greater extent in some CD45RA-CD27-CD4+ effector memory T cells. LIGHT, like CD40L, is rapidly up-regulated on memory CD4+ T cells upon activation. Unlike CD40L, LIGHT up-regulation on naive T cells is delayed. Stimulation of DC with each subset of memory T cells in the presence of superantigen revealed that CD40L and LIGHT are required for optimal secretion of IL-12. Moreover, effector memory T cells, which can interact with DC in peripheral tissues, contribute to CCR7 induction on DC. LIGHT and CD40L can also regulate IL-12 production by CCR7+ DC capable of migration to lymph nodes. These data suggest that both LIGHT and CD40L may be involved in the maintenance or reactivation of secondary Th1 responses.
- Published
- 2003
- Full Text
- View/download PDF
35. Abstract 1491: IPH4301, an antibody targeting MICA and MICB exhibits potent cytotoxic activity and immunomodulatory properties for the treatment of cancer
- Author
-
Ariane Morel, Nicolas Viaud, Cécile Bonnafous, Sylvia Trichard, Alix Joulin-Giet, Samia Mizari, Gwendoline Grondin, Nadia Anceriz, J. Zhang, J. Jarzen, J. Wu, Laetitia Cohen-Tannoudji, Yannis Morel, Benjamin Rossi, Carine Paturel, Renaud Buffet, Laurent Gauthier, Nicolai Wagtmann, and Mathieu Blery
- Subjects
0301 basic medicine ,Antibody-dependent cell-mediated cytotoxicity ,Cancer Research ,biology ,Chemistry ,medicine.disease_cause ,NKG2D ,stomatognathic diseases ,03 medical and health sciences ,030104 developmental biology ,Oncology ,Cell culture ,Immunology ,medicine ,Myeloid-derived Suppressor Cell ,Cancer research ,biology.protein ,Cytotoxic T cell ,Antibody ,Carcinogenesis ,Cytotoxicity - Abstract
MICA and MICB, and ULPB1-6, are ligands for NKG2D, an activating receptor expressed on NK cells and subsets of T cells. Expression of MICA and MICB is induced by cellular stress in transformed tumor cells, upon infections or at sites of chronic inflammation. Their expression is tightly regulated by complex mechanisms both at the mRNA and protein levels. As markers of cellular stress and tumorigenesis, MICA and MICB proteins are attractive candidates for targeting by a cytotoxic antibody. Moreover, ionizing radiation and various chemotherapies that cause cellular stress have been shown to induce expression of NKG2D ligands, opening interesting options for combination therapies. MICA and MICB are also compelling targets for immunomodulation. MICA and -B cause internalization of NKG2D, leading to reduced cell surface NKG2D levels and desensitization of cytotoxic effector cells in cancer patients. It was recently reported that blockade of the interactions between NKG2D and its ligands could lead to significant anti-tumor responses in mouse models. Moreover, NKG2D ligand expression was induced on immunosuppressive macrophages in cancer patients and in mouse tumor models, raising the possibility that anti-MICA/B antibodies may be used to counter local immunosuppression by targeting myeloid derived suppressor cells. We have selected the IPH4301 antibody for its ability to bind to all allotypes of MICA and MICB, and for its dual action as an immunomodulatory agent, as well as direct cytotoxicity towards MICA/B-expressing tumor cells. First, IPH4301 induces killing of MICA/B expressing tumor cells through antibody-dependent cell cytotoxicity (ADCC) and antibody-dependent cell phagocytosis (ADCP) measured towards MICA expressing cells in vitro. In vivo ADCC/ADCP efficacy was demonstrated in several preventive and curative settings using MICA expressing cell lines or endogenous tumors. Second, IPH4301 blocks the binding of MICA/B to NKG2D. In a tumor context of chronic triggering, NKG2D downmodulation has been described in several studies of cancer patients. This modulation is mainly induced by expression of MICA/B, and less by the ULBPs. By blocking the MICA/NKG2D interaction, IPH4301 effectively restored NKG2D expression and function in vitro on primary NK and T cells. Third, we show that IPH4301 can override immunosuppression induced by suppressive myeloid cells. In vitro differentiated M2 macrophages, but not M1 macrophages, have the capacity to impair cytotoxic functions of autologous NK cells towards MICA expressing tumor cell lines. This suppression could be overcome by IPH4301, which triggered ADCC by these otherwise impaired NK cells. Altogether, IPH4301 is a novel, first-in-class anti-MICA/B mAb with both cytotoxic and immunomodulatory properties. Ongoing work aims to perform regulatory toxicology studies and manufacture a clinical grade product for testing in a clinical trial. Citation Format: Ariane Morel, Nicolas Viaud, Cécile Bonnafous, Sylvia Trichard, Alix Joulin-Giet, Samia Mizari, Gwendoline Grondin, Nadia Anceriz, J. Zhang, J. Jarzen, J. Wu, Gwendoline Grondin, Laetitia Cohen-Tannoudji, Yannis Morel, Benjamin Rossi, Carine Paturel, Renaud Buffet, Laurent Gauthier, Nicolai Wagtmann, Mathieu Blery. IPH4301, an antibody targeting MICA and MICB exhibits potent cytotoxic activity and immunomodulatory properties for the treatment of cancer. [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 1491.
- Published
- 2016
- Full Text
- View/download PDF
36. Abstract 3222: Preclinical development of a humanized blocking antibody targeting the CD39 immune checkpoint for cancer immunotherapy
- Author
-
Stephane Delahaye, Rachel Joly, Yannis Morel, Hélène Rispaud Blanc, Nathalie Bonnefoy, Ivan Perrot, Laurent Gauthier, Caroline Denis, Carine Paturel, Jérémy Bastid, Jean-François Eliaou, Cécile Dejou, Armand Bensussan, and Severine Augier
- Subjects
Cancer Research ,Tumor microenvironment ,medicine.drug_class ,medicine.medical_treatment ,Cell ,Cancer ,Biology ,Monoclonal antibody ,medicine.disease ,Molecular biology ,Immune checkpoint ,medicine.anatomical_structure ,Immune system ,Oncology ,Cancer immunotherapy ,Blocking antibody ,medicine ,Cancer research - Abstract
CD39 (ENTPD1) is a cell membrane ectonucleotidase that hydrolyzes extracellular immunoactivating ATP and ADP into AMP, which can be further hydrolyzed by ectonucleotidase CD73 into immunosuppressive adenosine. Within the tumor microenvironment, adenosine accumulation causes immune suppression and dysregulation of immune cell infiltrates resulting in tumor spreading. The role of CD39 expression on both Tregs and on tumor cells in promoting immunosuppression has been demonstrated in several reports. Blockade of CD39 may promote anti-tumor immunity by directly accumulating immunostimulating ATP and indirectly by reducing adenosine accumulation. Here, we describe the discovery and preclinical development of an anti-huCD39 blocking antibody for cancer immunotherapy. Parental anti-huCD39 mouse monoclonal antibody was humanized. The humanized mAb specifically binds huCD39 protein, but not CD39-like proteins. Nanomolar affinities for human CD39 were measured in SPR studies on recombinant CD39 protein and in flow cytometry titration studies on CD39 expressing transfectants and tumor cell lines. The humanized mAb blocked human CD39 ATPase activity in vitro in the nanomolar range, as demonstrated using transfected cells, CD39-expressing tumor cell lines, as well as human PBMC and ex-vivo isolated fresh tumor samples. The humanized mAb cross-reacted on cynomolgus CD39 and blocked ATPase activity on cynomolgus PBMC with similar efficacy as on human PBMC. Finally, treatment with blocking anti-CD39 mAb inhibited tumor growth in vivo in mouse tumor models. Taken together, these data support the clinical development of anti-CD39 neutralizing mAb for cancer immunotherapy. Citation Format: Severine Augier, Ivan Perrot, Cecile Dejou, Rachel Joly, Stephane Delahaye, Helene Rispaud Blanc, Caroline Denis, Laurent Gauthier, Armand Bensussan, Jean-francois Eliaou, Yannis Morel, Nathalie Bonnefoy, Jeremy Bastid, Carine Paturel. Preclinical development of a humanized blocking antibody targeting the CD39 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 3222.
- Published
- 2016
- Full Text
- View/download PDF
37. Abstract 2342: NKG2A immune checkpoint blockade enhances the anti-tumor efficacy of PD1/PD-L1 inhibitors in a preclinical model
- Author
-
Caroline Sola, Cécile Bonnafous, Nicolas Fuseri, Benjamin Rossi, Pascale Andre, Nicolai Wagtmann, Corinne Leget, Fabien Chanuc, Laurent Gauthier, Thomas Arnoux, and Yannis Morel
- Subjects
0301 basic medicine ,Cancer Research ,biology ,business.industry ,Pharmacology ,Immune checkpoint ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Immune system ,Oncology ,030220 oncology & carcinogenesis ,PD-L1 ,Cancer cell ,biology.protein ,Cancer research ,Monalizumab ,Medicine ,Cytotoxic T cell ,Antibody ,business ,CD8 - Abstract
Monalizumab (IPH2201) is a novel, first-in-class humanized IgG4 targeting the immune checkpoint receptor NKG2A (Natural Killer Group 2A). NKG2A is expressed as a heterodimer with CD94 on the surface of subsets of cytotoxic lymphocytes: NK (Natural Killer) cells, γδ T cells and tumor infiltrating CD8 T lymphocytes. CD94/NKG2A is an inhibitory receptor that binds to HLA-E (Human Leukocyte Antigen-E) in humans and orthologous Qa-1b in mice. Upon ligand binding, CD94/NKG2A triggers inhibitory signaling that reduces NK and CD8 T cell responses. HLA-E is frequently up-regulated on cancer cells of many solid tumors or hematological malignancies, protecting from killing by NKG2A+ immune cells. By blocking the binding of CD94/NKG2A to HLA-E, monalizumab leads to enhancement of NK and cytotoxic T cell responses. The immune checkpoint receptor programmed cell death 1 (PD-1) is another inhibitory receptor widely upregulated by tumor-infiltrating T lymphocytes (TILs). In many tumor types immune surveillance is hampered by the expression of PD-L1, one of the ligands of PD-1. Blocking the PD-1 pathway has proven efficient as anti-tumor therapy. Nevertheless many patients remain refractory to these therapeutics. Combination treatment with PD-1 blockers and mAb to a second checkpoint receptor, CTLA-4, have proven effective only for some patients, suggesting a need for combining with other checkpoint blockers. The A20 cell line is a mouse B cell lymphoma line that expresses PD-L1, but not Qa-1b. Upon subcutaneous injection into Balb/c mice, A20 formed solid tumors in which PD-L1 expression was retained and where Qa-1b expression was induced. A20 tumor growth was controlled by NK and CD8 T cells. In spite of high expression of PD-1 on many immune infiltrating cells, and high expression of PD-L1 on tumor cells, monotherapy with anti-PD-1 or -PD-L1 mAb resulted in only moderate reduction in tumor growth. Interestingly, more than 50% of A20 tumor infiltrating NK cells and about 10% of CD8 T cells expressed CD94/NKG2A. The NKG2A+ CD8 T cell population also co-expressed PD-1. Qa-1b expression was induced not only on the surface of tumor cells but also on infiltrating immune cells in vivo. Consistent with the hypothesis that Qa-1b protects tumor cells from killing by NKG2A+ effector NK and T cells, treatment with an antibody that blocks NKG2A significantly delayed A20 tumor growth. Mice were then treated with both anti-NKG2A and anti-PD-1 or -PD-L1 in combination. These combinations resulted in significantly higher anti-tumor responses compared to monotherapies, characterized by an increased frequency of complete tumor cell regression. Together, these data indicate that blocking NKG2A in conjunction with PD-1/PD-L1 checkpoint inhibitors could provide synergistic anti-tumor efficacy and support the rationale for investigating this combination in clinical trials. Citation Format: Caroline Sola, Thomas Arnoux, Fabien Chanuc, Nicolas Fuseri, Benjamin Rossi, Laurent Gauthier, Corinne Leget, Cécile Bonnafous, Nicolai Wagtmann, Yannis Morel, Pascale André. NKG2A immune checkpoint blockade enhances the anti-tumor efficacy of PD1/PD-L1 inhibitors in a preclinical model. [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 2342.
- Published
- 2016
- Full Text
- View/download PDF
38. 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
- Full Text
- View/download PDF
39. Toll-like receptor 3 in Epstein-Barr virus-associated nasopharyngeal carcinomas: consistent expression and cytotoxic effects of its synthetic ligand poly(A:U) combined to a Smac-mimetic
- Author
-
Mélanie Gressette, Yannis Morel, Anne-Sophie Jimenez-Pailhes, Carine Paturel, Michel Wassef, Philippe Herman, Kwok Wai Lo, Sai Wah Tsao, Benjamin Verillaud, and Philippe Busson
- Subjects
Cancer Research ,Epidemiology ,Nasopharyngeal cancer ,Poly(A:U) ,Inhibitor of apoptosis ,medicine.disease_cause ,Western blot ,Inhibitor of Apoptosis Protein ,medicine ,Cytotoxic T cell ,Epstein-Barr virus ,Clonogenic assay ,medicine.diagnostic_test ,business.industry ,Cell growth ,Epstein–Barr virus ,In vitro ,Toll-Like Receptor 3 ,Infectious Diseases ,Oncology ,Cell culture ,Immunology ,Cancer research ,business ,Smac-mimetic ,Research Article - Abstract
Background Nasopharyngeal carcinomas (NPC) are consistently associated with the Epstein-Barr virus (EBV). Though NPCs are more radiosensitive and chemosensitive than other tumors of the upper aero-digestive tract, many therapeutic challenges remain. In a previous report, we have presented data supporting a possible therapeutic strategy based on artificial TLR3 stimulation combined to the inhibition of the IAP protein family (Inhibitor of Apoptosis Proteins). The present study was designed to progress towards practical applications of this strategy pursuing 2 main objectives: 1) to formally demonstrate expression of the TLR3 protein by malignant NPC cells; 2) to investigate the effect of poly(A:U) as a novel TLR3-agonist more specific than poly(I:C) which was used in our previous study. Methods TLR3 expression was investigated in a series of NPC cell lines and clinical specimens by Western blot analysis and immunohistochemistry, respectively. The effects on NPC cells growth of the TLR3 ligand poly(A:U) used either alone or in combination with RMT5265, an IAP inhibitor based on Smac-mimicry, were assessed using MTT assays and clonogenic assays. Results TLR3 was detected at a high level in all NPC cell lines and clinical specimens. Low concentrations of poly(A:U) were applied to several types of NPC cells including cells from the C17 xenograft which for the first time have been adapted to permanent propagation in vitro. As a single agent, poly(A:U) had no significant effects on cell growth and cell survival. In contrast, dramatic effects were obtained when it was combined with the IAP inhibitor RMT5265. These effects were obtained using concentrations as low as 0.5 μg/ml (poly(A:U)) and 50 nM (RMT5265). Conclusion These data confirm that TLR3 expression is a factor of vulnerability for NPC cells. They suggest that in some specific pathological and pharmacological contexts, it might be worth to use Smac-mimetics at very low doses, allowing a better management of secondary effects. In light of our observations, combined use of both types of compounds should be considered for treatment of nasopharyngeal carcinomas.
- Published
- 2012
40. Desirable cell death during anticancer chemotherapy
- Author
-
Clara, Locher, Rosa, Conforti, Laetitia, Aymeric, Yuting, Ma, Takahiro, Yamazaki, Sylvie, Rusakiewicz, Antoine, Tesnière, François, Ghiringhelli, Lionel, Apetoh, Yannis, Morel, Jean-Philippe, Girard, Guido, Kroemer, and Laurence, Zitvogel
- Subjects
Mice ,Neoplasms ,Animals ,Humans ,Antineoplastic Agents ,Apoptosis ,Cancer Vaccines ,Combined Modality Therapy - Abstract
The concept of immunogenic chemotherapy that has recently emerged relies upon the capacity of a cytotoxic compound to trigger a cell-death modality. This modality elicits cross-priming by dendritic cells of tumor antigen-specific T cells that will contribute to the tumoricidal activity of the compound and protect the host against relapse. In contrast, most anticancer drugs elicit nonimmunogenic apoptosis that is not accompanied with an immunizing property. This review will discuss some molecular and metabolic changes required at the level of the tumor that must engage key pathways at the level of the host for the induction of Tc1 polarized-protective T cell responses during chemotherapy. We will summarize the immune adjuvants that can boost the immunogenicity of cell death to augment the efficacy of chemotherapy.
- Published
- 2010
41. TLR3 and Rig-like receptor on myeloid dendritic cells and Rig-like receptor on human NK cells are both mandatory for production of IFN-gamma in response to double-stranded RNA
- Author
-
Laurent Gauthier, Nathalie Bonnefoy-Berard, Mathieu Blery, Yannis Morel, Isabelle Durand, Ivan Perrot, Christophe Caux, Pierre Garrone, Lena Alexopoulou, Florence Deauvieau, Catherine Massacrier, Giorgio Trinchieri, Nicola Hughes, Nicolas Viaud, Jürg Tschopp, Carine Paturel, Olivier Demaria, Innate Pharma, Oncogénèse et progression tumorale, 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), Immunité infection vaccination (I2V), Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-IFR128-Institut National de la Santé et de la Recherche Médicale (INSERM), Centre d'Immunologie de Marseille - Luminy (CIML), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Aix Marseille Université (AMU), Institute of Biochemistry, Université de Lausanne (UNIL)-BIL Biomedical Research Center, Schering-Plough Laboratories for Immunological Research, Schering Plough Laboratories, Aix Marseille Université (AMU)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), and Université de Lausanne = University of Lausanne (UNIL)-BIL Biomedical Research Center
- Subjects
Interferon-Induced Helicase, IFIH1 ,viruses ,Lymphocyte Activation ,MESH: Mice, Knockout ,MESH: Dose-Response Relationship, Drug ,DEAD-box RNA Helicases ,Mice ,0302 clinical medicine ,Immunology and Allergy ,Interferon gamma ,Myeloid Cells ,MESH: Animals ,Receptors, Immunologic ,Receptor ,Cells, Cultured ,Mice, Knockout ,0303 health sciences ,MESH: Dendritic Cells ,virus diseases ,MDA5 ,hemic and immune systems ,Transfection ,MESH: Toll-Like Receptor 3 ,Killer Cells, Natural ,030220 oncology & carcinogenesis ,DEAD Box Protein 58 ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,Cell activation ,medicine.drug ,MESH: Cells, Cultured ,MESH: Killer Cells, Natural ,MESH: Interferon-gamma ,Immunology ,Adaptor Proteins, Signal Transducing/genetics ,Adaptor Proteins, Signal Transducing/metabolism ,Animals ,Cell Line ,DEAD-box RNA Helicases/genetics ,DEAD-box RNA Helicases/metabolism ,Dendritic Cells/cytology ,Dendritic Cells/drug effects ,Dose-Response Relationship, Drug ,Humans ,Interferon-gamma/metabolism ,Killer Cells, Natural/cytology ,Killer Cells, Natural/drug effects ,Lymphocyte Activation/drug effects ,Mice, Inbred C57BL ,Myeloid Cells/cytology ,Myeloid Cells/drug effects ,Poly A-U/pharmacology ,Poly I-C/pharmacology ,RNA, Double-Stranded/pharmacology ,Toll-Like Receptor 3/genetics ,Toll-Like Receptor 3/metabolism ,chemical and pharmacologic phenomena ,Biology ,MESH: Poly I-C ,Article ,03 medical and health sciences ,Interferon-gamma ,MESH: RNA, Double-Stranded ,MESH: Mice, Inbred C57BL ,medicine ,MESH: DEAD-box RNA Helicases ,MESH: Poly A-U ,MESH: Lymphocyte Activation ,MESH: Mice ,030304 developmental biology ,Adaptor Proteins, Signal Transducing ,RNA, Double-Stranded ,MESH: Adaptor Proteins, Signal Transducing ,MESH: Humans ,MESH: Transfection ,Dendritic Cells ,Molecular biology ,MESH: Myeloid Cells ,Toll-Like Receptor 3 ,MESH: Cell Line ,Cytolysis ,Poly I-C ,Cell culture ,TLR3 ,Poly A-U - Abstract
Cross-talk between NK cells and dendritic cells (DCs) is critical for the potent therapeutic response to dsRNA, but the receptors involved remained controversial. We show in this paper that two dsRNAs, polyadenylic-polyuridylic acid and polyinosinic-polycytidylic acid [poly(I:C)], similarly engaged human TLR3, whereas only poly(I:C) triggered human RIG-I and MDA5. Both dsRNA enhanced NK cell activation within PBMCs but only poly(I:C) induced IFN-γ. Although myeloid DCs (mDCs) were required for NK cell activation, induction of cytolytic potential and IFN-γ production did not require contact with mDCs but was dependent on type I IFN and IL-12, respectively. Poly(I:C) but not polyadenylic-polyuridylic acid synergized with mDC-derived IL-12 for IFN-γ production by acting directly on NK cells. Finally, the requirement of both TLR3 and Rig-like receptor (RLR) on mDCs and RLRs but not TLR3 on NK cells for IFN-γ production was demonstrated using TLR3- and Cardif-deficient mice and human RIG-I–specific activator. Thus, we report the requirement of cotriggering TLR3 and RLR on mDCs and RLRs on NK cells for a pathogen product to induce potent innate cell activation.
- Published
- 2010
- Full Text
- View/download PDF
42. Poly(I:C) induces intense expression of c-IAP2 and cooperates with an IAP inhibitor in induction of apoptosis in cancer cells
- Author
-
Carine Paturel, Claire Gourzones, Catherine Uzan, Stéphane Temam, Yannis Morel, Luc Friboulet, Sai Wah Tsao, and Philippe Busson
- Subjects
Adult ,Male ,Cell type ,Cancer Research ,Time Factors ,Ubiquitin-Protein Ligases ,Adaptor Proteins, Vesicular Transport - metabolism ,Apoptosis - drug effects ,Apoptosis ,Biology ,lcsh:RC254-282 ,Inhibitor of Apoptosis Proteins ,Neoplasms - genetics - metabolism - pathology ,Cell Line, Tumor ,Neoplasms ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Tumor Cells, Cultured ,Genetics ,Humans ,RNA, Messenger ,Clonogenic assay ,Receptor ,Cell Proliferation ,Dose-Response Relationship, Drug ,Cell growth ,Inhibitor of Apoptosis Proteins - antagonists and inhibitors - genetics - metabolism ,Antineoplastic Combined Chemotherapy Protocols - pharmacology ,Middle Aged ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Molecular biology ,Baculoviral IAP Repeat-Containing 3 Protein ,Toll-Like Receptor 3 ,Up-Regulation ,Gene Expression Regulation, Neoplastic ,Adaptor Proteins, Vesicular Transport ,Poly I-C ,Nasopharyngeal carcinoma ,Oncology ,Cancer cell ,Female ,RNA Interference ,Stem cell ,Research Article - Abstract
Background: There is increasing evidence that the toll-like receptor 3 (TLR3) is an interesting target for anti-cancer therapy. Unfortunately, most laboratory investigations about the impact of TLR3 stimulation on human malignant cells have been performed with very high concentrations - 5 to 100 μg/ml - of the prototype TLR3 ligand, poly(I:C). In a previous study focused on a specific type of human carcinoma - nasopharyngeal carcinoma - we have shown that concentrations of poly(I:C) as low as 100 ng/ml are sufficient to induce apoptosis of malignant cells when combined to a pharmacological antagonist of the IAP family based on Smac mimicry.Methods: This observation prompted us to investigate the contribution of the IAP family in cell response to poly(I:C) in a variety of human malignant cell types.Results: We report a rapid, intense and selective increase in c-IAP2 protein expression observed under stimulation by poly(I:C)(500 ng/ml) in all types of human malignant cells. In most cell types, this change in protein expression is underlain by an increase in c-IAP2 transcripts and dependent on the TLR3/TRIF pathway. When poly(I:C) is combined to the IAP inhibitor RMT 5265, a cooperative effect in apoptosis induction and/or inhibition of clonogenic growth is obtained in a large fraction of carcinoma and melanoma cell lines.Conclusions: Currently, IAP inhibitors like RMT 5265 and poly(I:C) are the subject of separate therapeutic trials. In light of our observations, combined use of both types of compounds should be considered for treatment of human malignancies including carcinomas and melanomas. © 2010 Friboulet et al; licensee BioMed Central Ltd., published_or_final_version
- Published
- 2010
43. Plasmacytoid dendritic cell–derived type I interferon is crucial for the adjuvant activity of Toll-like receptor 7 agonists
- Author
-
Satoshi Uematsu, Yannis Morel, Shizuo Akira, Sandra S. Diebold, Carine Paturel, and Deepa Rajagopal
- Subjects
medicine.medical_treatment ,Immunology ,Melanoma, Experimental ,Antineoplastic Agents ,Plasmacytoid dendritic cell ,Receptor, Interferon alpha-beta ,Biology ,Biochemistry ,Cancer Vaccines ,Mice ,Immune system ,Adjuvants, Immunologic ,medicine ,Tumor Cells, Cultured ,Animals ,Antigen-presenting cell ,Immunobiology ,Mice, Knockout ,Toll-like receptor ,Innate immune system ,Imiquimod ,Membrane Glycoproteins ,Dose-Response Relationship, Drug ,Imidazoles ,Cell Biology ,Hematology ,Immunotherapy ,Dendritic cell ,Dendritic Cells ,Mice, Inbred C57BL ,Toll-Like Receptor 7 ,Interferon Type I ,Aminoquinolines ,Quinolines ,RNA ,Interferon type I ,medicine.drug - Abstract
There is a high demand for the development of adjuvants that induce cytotoxic T lymphocytes, which are crucial for the elimination of intracellular pathogens and tumor cells. Toll-like receptor (TLR) agonists are prime candidates to fulfill this role because they induce innate immune activation and promote adaptive immune responses. The successful application of the TLR7 agonist R837 for treatment of basal cell carcinoma shows the potential for exploiting this pathway in tumor immunotherapy. Imidazoquinolines like R837 and stimulatory ssRNA oligonucleotides both trigger TLR7-mediated immune activation, but little is known about their comparative ability to promote immunity induction. We investigated differences in innate immune activation and adjuvant activity between the imidazoquinoline R848 and the ssRNA TLR7 agonist polyUs21. In contrast to R848, polyUs21 induced detectable levels of intracellular interferon-α (IFN-α) in plasmacytoid dendritic cells (PDCs). In immunization studies, only polyUs21 led to robust priming of type 1 T helper cells and cytotoxic T lymphocytes, and it was more efficient in inducing antitumor immunity than R848. Notably, exogenous IFN-α augmented the adjuvant activity of R848, whereas depletion of PDC abrogated the adjuvanticity of polyUs21. This study, therefore, identifies sufficient IFN-α production by PDC as an important determinant of vaccine efficacy.
- Published
- 2010
44. Identification, activation, and selective in vivo ablation of mouse NK cells via NKp46
- Author
-
Sébastien Jaeger, Thierry Walzer, Mathieu Blery, Michel Pierres, Laurent Daniel, Yannis Morel, François Romagné, Laurent Gauthier, Marc Dalod, Nicolas Fuseri, Julie Chaix, Scott H. Robbins, Lionel Chasson, Karine Chemin, Eric Vivier, Alessandro Moretta, Pascale Andre, Jean Imbert, Centre d'Immunologie de Marseille - Luminy (CIML), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Aix Marseille Université (AMU), and Aix Marseille Université (AMU)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
Cellular differentiation ,MESH: Membrane Glycoproteins ,MESH: Microscopy, Fluorescence ,MESH: Flow Cytometry ,CD49b ,Mice ,Interleukin 21 ,0302 clinical medicine ,Antigens, Ly ,Diphtheria Toxin ,MESH: Animals ,Receptors, Immunologic ,Promoter Regions, Genetic ,MESH: Haplorhini ,0303 health sciences ,Membrane Glycoproteins ,Multidisciplinary ,Janus kinase 3 ,Cell Differentiation ,Haplorhini ,Biological Sciences ,Flow Cytometry ,Natural killer T cell ,MESH: Diphtheria Toxin ,MESH: Gene Expression Regulation ,Cell biology ,Killer Cells, Natural ,MESH: Promoter Regions (Genetics) ,Interleukin 12 ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,MESH: Cell Differentiation ,MESH: Killer Cells, Natural ,MESH: Immunophenotyping ,MESH: Mice, Transgenic ,Green Fluorescent Proteins ,Mice, Transgenic ,Biology ,Immunophenotyping ,03 medical and health sciences ,MESH: Green Fluorescent Proteins ,Animals ,MESH: Mice ,MESH: Receptors, Immunologic ,030304 developmental biology ,Lymphokine-activated killer cell ,Natural Cytotoxicity Triggering Receptor 1 ,Molecular biology ,Gene Expression Regulation ,Microscopy, Fluorescence ,Myeloid-derived Suppressor Cell ,030215 immunology - Abstract
Natural killer (NK) cells contribute to a variety of innate immune responses to viruses, tumors and allogeneic cells. However, our understanding of NK cell biology is severely limited by the lack of consensus phenotypic definition of these cells across species, by the lack of specific marker to visualize them in situ , and by the lack of a genetic model where NK cells may be selectively ablated. NKp46/CD335 is an Ig-like superfamily cell surface receptor involved in human NK cell activation. In addition to human, we show here that NKp46 is expressed by NK cells in all mouse strains analyzed, as well as in three common monkey species, prompting a unifying phenotypic definition of NK cells across species based on NKp46 cell surface expression. Mouse NKp46 triggers NK cell effector function and allows the detection of NK cells in situ . NKp46 expression parallels cell engagement into NK differentiation programs because it is detected on all NK cells from the immature CD122 + NK1.1 + DX5 − stage and on a minute fraction of NK-like T cells, but not on CD1d-restricted NKT cells. Moreover, human NKp46 promoter drives NK cell selective expression both in vitro and in vivo . Using NKp46 promoter, we generated transgenic mice expressing EGFP and the diphtheria toxin (DT) receptor in NK cells. DT injection in these mice leads to a complete and selective NK cell ablation. This model paves a way for the in vivo characterization and preclinical assessment of NK cell biological function.
- Published
- 2007
- Full Text
- View/download PDF
45. Lirilumab Enhances Anti-Tumor Efficacy of Elotuzumab
- Author
-
Mathieu Blery, Robert F. Graziano, Elodie Bonnet, Nicolas Fuseri, Yannis Morel, Pascale Andre, Cécile Bonnafous, and Caroline Sola
- Subjects
Antibody-dependent cell-mediated cytotoxicity ,biology ,business.industry ,SLAMF7 ,Immunology ,Fc receptor ,Cell Biology ,Hematology ,Pharmacology ,Major histocompatibility complex ,Biochemistry ,biology.protein ,Medicine ,Tumor necrosis factor alpha ,Antibody ,Elotuzumab ,business ,Receptor ,medicine.drug - Abstract
Tumor cells that express reduced levels of Major Histocompatibility Complex (MHC) class I molecules may be recognized and killed by Natural Killer cells (NK cells), through a process known as “missing self” recognition. In humans, this is controlled by inhibitory receptors such as Killer Immunoglobulin-like Receptors (KIR) that recognize Human Leukocyte Antigen (HLA)-A, -B or –C. Engagement of KIR by HLA molecules results in inhibitory signaling that reduces NK cell-mediated natural killing and antibody-dependent cellular cytotoxicity (ADCC). Hence, antibodies that block interactions between inhibitory KIR and their HLA ligands are being evaluated as an anti-cancer therapeutic strategy. The anti-KIR2DL1/2/3-specific monoclonal antibody, lirilumab (BMS-986015 / IPH2102), is a fully human IgG4 that blocks binding of KIR to HLA-C, and is being developed for treating hematologic malignancies and solid tumors. Elotuzumab (BMS901608 / HuLuc63) is a humanized IgG1 anti-SLAMF7 (signaling lymphocyte activation molecule family member 7, CS-1) being developed for the treatment of Multiple Myeloma (MM). SLAMF7 is a cell surface glycoprotein highly expressed in myelomatous cells and only at low levels on normal cells. NK cell-mediated ADCC is one of the main mechanisms of action of elotuzumab, but ADCC is negatively regulated by KIR checkpoint receptors. Thus a combination of lirilumab and elotuzumab has strong scientific rationale. The aim of the present study was to assess whether lirilumab would enhance elotuzumab anti-MM activity in vitro with human peripheral blood NK cells and MM cell lines, and in vivo in a newly developed xenogenic mouse model. Two MM cell lines (OPM-2 and U266B1) were identified that express both HLA-C and SLAMF7. These MM cells were capable of activating peripheral blood NK cells from healthy donors in vitro, as assessed by three different endpoints (CD107 mobilization on NK cells surface and intracellular production of the cytokines IFN-g and TNF- a); each of these responses were significantly enhanced, in a dose-dependent manner, by both lirilumab and elotuzumab independently. Moreover, the elotuzumab-mediated functional activation of KIR2D+ NK cells could be further enhanced by the addition of increasing doses of lirilumab. The best combinatorial effect was observed in response to MM cells expressing low densities of SLAMF7. These data suggest that lirilumab treatment may increase the therapeutic efficacy of elotuzumab, particularly in MM patients with low SLAMF7 expression. In these experiments, it was not possible to clearly identify the impact of Fc receptor genotype or HLA-C genotype on the NK cell responses. To assess the therapeutic efficacy of lirilumab and elotuzumab in vivo, we generated a novel strain of double-transgenic mice expressing human KIR2DL3 as well as its ligand, HLA-cw3, on a Rag1-/- background (KIR-cw3-tgRAG mice), to allow engraftment of human MM tumor cells expressing SLAMF7. The OPM-2 MM cell line was subcutaneously engrafted in these mice and when high tumor volumes were reached, mice were treated with lirilumab, elotuzumab or a combination of both. As monotherapy, each of monoclonal antibody had some therapeutic effect while the combination of both resulted in a significantly stronger anti-tumor effect and increased survival of the mice. Median survival of mice treated with huIgG control was 38 days, 41 days with lirilumab, 42 days with elotuzumab and 51 days with both mAbs in combination (10 mice per group). In conclusion, we demonstrate that blockade of KIR checkpoint receptors with lirilumab was able to augment elotuzumab mediated ADCC in vitro and synergized with elotuzumab to mediate potent anti-MM activity in vivo. Taken together, these data provide a rationale for clinical trials to test combination treatment of lirilumab and elotuzumab in MM patients. Disclosures Sola: InnatePharma: Employment, Equity Ownership. Blery:Innate Pharma: Employment, Equity Ownership. Bonnafous:Innate Pharma: Employment, Equity Ownership. Bonnet:Innate Pharma: Employment, Equity Ownership. Fuseri:Innate Pharma: Employment, Equity Ownership. Graziano:Bristol-Myers Squibb: Employment; Bristol-Myers Squibb: Equity Ownership. Morel:Innate Pharma: Employment, Equity Ownership. André:Innate Pharma: Employment, Equity Ownership.
- Published
- 2014
- Full Text
- View/download PDF
46. Mechanisms regulating expression of the tumor necrosis factor-related light gene. Role of calcium-signaling pathway in the transcriptional control
- Author
-
Remy, Castellano, Carine, Van Lint, Valentine, Peri, Emmanuelle, Veithen, Yannis, Morel, Regis, Costello, Daniel, Olive, and Yves, Collette
- Subjects
Tumor Necrosis Factor Ligand Superfamily Member 14 ,Base Sequence ,NFATC Transcription Factors ,Transcription, Genetic ,Reverse Transcriptase Polymerase Chain Reaction ,Tumor Necrosis Factor-alpha ,T-Lymphocytes ,Molecular Sequence Data ,Membrane Proteins ,Nuclear Proteins ,Lymphocyte Activation ,Recombinant Proteins ,DNA-Binding Proteins ,Jurkat Cells ,Gene Expression Regulation ,Cyclosporine ,Mutagenesis, Site-Directed ,Humans ,Calcium Signaling ,Promoter Regions, Genetic ,Cells, Cultured ,Immunosuppressive Agents ,Signal Transduction ,Transcription Factors - Abstract
LIGHT (TNFSF14) is a newly identified tumor necrosis factor superfamily member involved in the regulation of immune responses by control of activation, maturation, and survival of immune effector cells. Despite the immunological relevance of the LIGHT protein, little knowledge is available as to how light gene expression is regulated. In T-lymphocytes, most LIGHT surface expression and transcript accumulation occurs after T cell activation. In this study, we have shown that these events are blocked at the transcriptional level by cyclosporin A, an immuno-suppressive drug. Besides, we identified a role for Ca2+ -signaling pathways and NFAT transcription factors in T cell activation-induced LIGHT expression. To further investigate this process, we have identified, cloned, and characterized a 2.1-kilobase 5'-flanking DNA genomic fragment from the human light gene. We have shown the transcriptional activity of the herein-identified minimal 5' regulatory region of human light gene parallels the endogenous expression of light in T cells. Moreover, we demonstrated that induced LIGHT promoter activity can be equally blocked by cyclosporin A treatment or dominant negative NFAT overexpression and further identified by site-directed mutagenesis and electrophoretic mobility supershift analysis of a NFAT transcription factor binding site within the human light minimal promoter. Finally, Sp1 and Ets1 binding sites were identified and shown to regulate light basal promoter activity. Thus, the present study establishes a molecular basis to further understand the mechanisms governing human light gene expression and, consequently, could potentially lead to novel therapeutic manipulations that control the signaling cascade, resulting in LIGHT production in conditions characterized by immunopathologic activation of T cells.
- Published
- 2002
47. Abstract 2514: Towards homogenous adcs: A new site-specific antibody conjugation using bacterial transglutaminase (btg-adc)
- Author
-
Agnès Represa, Patrick Dennler, Yannis Morel, François Romagné, Delphine Bregeon, Angelique Boedec, Florence Lhospice, Roger Schibli, and Christian Belmant
- Subjects
Cancer Research ,biology ,Chemistry ,Tissue transglutaminase ,Endogeny ,Molecular biology ,In vitro ,body regions ,Oncology ,Biochemistry ,Pharmacokinetics ,In vivo ,biology.protein ,Antibody ,Linker ,EC50 - Abstract
Purpose : Bacterial Transglutaminase (BTG) allows coupling on endogenous Q295 from an aglycosylated antibody. Here, the work relates to ADCs synthesis through BTG approach on mAbs with single point mutations and the study of stability, pharmacokinetics, as well as in vitro and in vivo efficacy of resulting BTG-ADCs. AdcetriS®, brentuximab vedotin, has been used as comparator, hence for consistency BTG-ADCs have been conjugated with the different linkage strategies (one step and two step) containing distally the same protease sensitive moiety compared to ADCETRIS®, i.e. valine-citrulline-PAB-MMAE. Methods : . For synthesis, a two-step process has been designed consisting of coupling first a linker and coming back with a toxin, and affording versatility and efficiency. The DAR stability was monitored over one week in human and cynomolgus plasma by affinity capture/LC/MS. The pharmacokinetic profile was studied in rat and monitored by affinity capture/LC/MS for conjugated antibody and ELISA for total antibody. In vitro efficacy of ADCs conjugated using BTG was compared side-by-side with ADCETRIS® on Karpas 299 cells. In vivo efficacy was studied in xenogenic nude mice engrafted with Karpas 299 in SC with BTG ADCs or ADCETRIS at doses of 0.6mg/kg. Results: BTG two-step process leads to ADCs with DAR of exactly 2.0 or 4.0 for antibodies with N297S or N297Q single point mutation respectively. The BTG ADCs were stable in human and cynomolgus plasma, with no degradation observed over 15 days in rats and furthermore with favorable total antibody clearance. In vitro efficacy for BTG ADCs DAR=4.0 was comparable to ADCETRIS (mean DAR=4) with respect to EC50 and plateau. In vivo efficacy for BTG ADC was demonstrated to be at least equivalent to ADCETRIS®. Conclusions: Pre-clinical proof of concept is demonstrated for BTG coupling. When considering versatility, speed and efficiency of the process, BTG-ADC coupling should provide a promising technology for the next generation of homogeneous ADCs. Citation Format: Florence Lhospice, Delphine Bregeon, Christian Belmant, Agnes Represa, Angelique Boedec, Yannis Morel, Patrick Dennler, Roger Schibli, François Romagne. Towards homogenous adcs: A new site-specific antibody conjugation using bacterial transglutaminase (btg-adc). [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 2514. doi:10.1158/1538-7445.AM2014-2514
- Published
- 2014
- Full Text
- View/download PDF
48. Abstract 5037: Targeting MICA with therapeutic antibodies for the treatment of cancer
- Author
-
Ariane Thielens, Stéphanie Cornen, Valentine Peri, Laurent Gauthier, Mathieu Blery, Ivan Perrot, Cécile Bonnafous, Sylvia Trichard, Yannis Morel, François Romagné, Benjamin Rossi, Carine Paturel, and Violette Breso
- Subjects
Antibody-dependent cell-mediated cytotoxicity ,Cancer Research ,biology ,business.industry ,medicine.drug_class ,Cancer ,NKG2D ,Monoclonal antibody ,medicine.disease ,medicine.disease_cause ,stomatognathic diseases ,Oncology ,Antigen ,Immunology ,Cancer research ,biology.protein ,Medicine ,Cytotoxic T cell ,Antibody ,business ,Carcinogenesis - Abstract
MICA and MICB, along with ULPBs, are ligands for the activating receptor NKG2D expressed on NK cells and subsets of T cells in Human. NKG2D ligands are induced by cellular stress and pathogen infections. Their expression is tightly regulated by complex mechanisms both at the mRNA and protein levels. In the case of MICA and MICB, more than 65 and 30 alleles respectively were described with different properties regarding to their cellular location adding to the complexity of this recognition system. Nevertheless, as markers of cellular stress, in particular in tumorigenesis, MICA and the closely related MICB proteins are candidates of choice to be targeted by a cytotoxic therapeutic antibody. We first evaluated MICA/B expression by immunohistochemistry on healthy tissues and tumors to validate these antigens as therapeutic targets. Then, using mouse immunization, we generated a panel of chimeric human IgG1 monoclonal antibodies targeting MICA and MICB. These mAbs have the ability to bind to several structurally different alleles and to cross-react on MIC proteins from cynomolgus macaques. Their capacity to block the MICA/NKG2D interaction was assessed by surface plasmon resonance as well as by using cell-based assays. In vitro efficacy was measured by the capacity to mediate complement-dependent cytotoxicity (CDC) and antibody-dependent cell cytotoxicity (ADCC) towards MICA expressing cells. In vivo efficacy of the anti-MICA mAbs was measured in both a preventive and a curative setting using MICA expressing cell lines. Altogether, we have generated a panel of anti-MICA mAbs with diverse functional properties. Ongoing work aims to choose the best candidate for humanization and further clinical development. Citation Format: Mathieu Blery, Cécile Bonnafous, Valentine Peri, Sylvia Trichard, Ivan Perrot, Stéphanie Cornen, Ariane Thielens, Violette Breso, Yannis Morel, François Romagne, Benjamin Rossi, Carine Paturel, Laurent Gauthier. Targeting MICA with therapeutic antibodies for the treatment of cancer. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 5037. doi:10.1158/1538-7445.AM2014-5037
- Published
- 2014
- Full Text
- View/download PDF
49. The TNF superfamily members LIGHT and CD154 (CD40 ligand) costimulate induction of dendritic cell maturation and elicit specific CTL activity
- Author
-
Yannis Morel, Alemseged Truneh, Régis T. Costello, Raymond W. Sweet, and Daniel Olive
- Subjects
Herpesvirus entry mediator ,Tumor Necrosis Factor Ligand Superfamily Member 14 ,T cell ,medicine.medical_treatment ,Immunology ,CD40 Ligand ,chemical and pharmacologic phenomena ,In Vitro Techniques ,Lymphocyte Activation ,Transfection ,Receptors, Tumor Necrosis Factor ,Cell Line ,Interferon-gamma ,Mice ,Neoplasms ,medicine ,Immunology and Allergy ,Animals ,Humans ,CD154 ,CD40 ,biology ,Tumor Necrosis Factor-alpha ,Membrane Proteins ,hemic and immune systems ,Cell Differentiation ,Dendritic cell ,Immunotherapy ,Dendritic Cells ,Cell biology ,CTL ,medicine.anatomical_structure ,biology.protein ,Cytokines ,Pinocytosis ,Receptors, Virus ,Cytokine secretion ,Receptors, Tumor Necrosis Factor, Member 14 ,Signal Transduction ,T-Lymphocytes, Cytotoxic - Abstract
LIGHT is a recently identified member of the TNF superfamily that is up-regulated upon activation of T cells. Herpesvirus entry mediator, one of its receptors, is constitutively expressed on immature dendritic cells (DCs). In this report, we demonstrate that LIGHT induces partial DC maturation as demonstrated by Ag presentation and up-regulation of adhesion and costimulatory molecules. LIGHT-stimulated DCs show reduced macropinocytosis and enhanced allogeneic stimulatory capacity but fail to produce significant amounts of IL-12, IL-6, IL-1β, or TNF-α compared with unstimulated DCs. However, LIGHT cooperates with CD154 (CD40 ligand) in DC maturation, with particular potentiation of allogeneic T cell proliferation and cytokine secretion of IL-12, IL-6, and TNF-α. Moreover, LIGHT costimulation allows DCs to prime in vitro-enhanced specific CTL responses. Our results suggest that LIGHT plays an important role in DC-mediated immune responses by regulating CD154 signals and represents a potential tool for DC-based cancer immunotherapy.
- Published
- 2001
50. Reciprocal expression of the TNF family receptor herpes virus entry mediator and its ligand LIGHT on activated T cells: LIGHT down-regulates its own receptor
- Author
-
Raymond W. Sweet, Sanjay S. Khandekar, Régis T. Costello, Jeremy A. Harrop, Jean-Marc Schiano de Colella, Jean-Albert Gastaut, Yannis Morel, Stephen D. Holmes, Daniel Olive, Alemseged Truneh, Trevor A. Wattam, and Keith Charles Deen
- Subjects
CD4-Positive T-Lymphocytes ,Herpesvirus entry mediator ,Tumor Necrosis Factor Ligand Superfamily Member 14 ,T cell ,Immunology ,Cell ,Down-Regulation ,Cell Separation ,Biology ,CD8-Positive T-Lymphocytes ,Ligands ,Lymphocyte Activation ,Receptors, Tumor Necrosis Factor ,Immune system ,T-Lymphocyte Subsets ,medicine ,Immunology and Allergy ,Humans ,Simplexvirus ,RNA, Messenger ,Cycloheximide ,Enzyme Inhibitors ,Receptor ,Cells, Cultured ,Protein Synthesis Inhibitors ,Microscopy, Confocal ,Tumor Necrosis Factor-alpha ,Membrane Proteins ,Metalloendopeptidases ,Fusion protein ,Cell biology ,Up-Regulation ,medicine.anatomical_structure ,Cytoplasm ,Receptors, Virus ,Receptors, Tumor Necrosis Factor, Member 14 ,CD8 - Abstract
The TNF receptor (TNFR) family plays a central role in the development of the immune response. Here we describe the reciprocal regulation of the recently identified TNFR superfamily member herpes virus entry mediator (HVEM) (TR2) and its ligand LIGHT (TL4) on T cells following activation and the mechanism of this process. T cell activation resulted in down-regulation of HVEM and up-regulation of LIGHT, which were both more pronounced in CD8+ than CD4+ T lymphocytes. The analysis of HVEM and LIGHT mRNA showed an increase in the steady state level of both mRNAs following stimulation. LIGHT, which was present in cytoplasm of resting T cells, was induced both in cytoplasm and at the cell surface. For HVEM, activation resulted in cellular redistribution, with its disappearance from cell surface. HVEM down-regulation did not rely on de novo protein synthesis, in contrast to the partial dependence of LIGHT induction. Matrix metalloproteinase inhibitors did not modify HVEM expression, but did enhance LIGHT accumulation at the cell surface. However, HVEM down-regulation was partially blocked by a neutralizing mAb to LIGHT or an HVEM-Fc fusion protein during activation. As a model, we propose that following stimulation, membrane or secreted LIGHT binds to HVEM and induces receptor down-regulation. Degradation or release of LIGHT by matrix metalloproteinases then contributes to the return to baseline levels for both LIGHT and HVEM. These results reveal a self-regulating ligand/receptor system that contributes to T cell activation through the interaction of T cells with each other and probably with other cells of the immune system.
- Published
- 2000
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.