30 results on '"Olivier Adotevi"'
Search Results
2. Data from Plasma CD27, a Surrogate of the Intratumoral CD27–CD70 Interaction, Correlates with Immunotherapy Resistance in Renal Cell Carcinoma
- Author
-
Eric Tartour, Stéphane Oudard, Yann A. Vano, Olivier Adotevi, Arnaud Mejean, Julien Adam, Sandrine Katsahian, Laurence Albiges, Nathalie Chaput, Bernd Jabla, Dominique Helley, Laetitia Mauge, Eléonore De Guillebon, Patrice Ravel, Chloe Broudin, Clémence Granier, Sebastien Mella, Valentina Libri, Virginie Verkarre, Camille Nevoret, Valentin Quiniou, Milena Hasan, Joséphine Pineau, Antonin Saldmann, Nadège Gruel, Hang Phuong Pham, Letuan Phan, Alain Gey, Nicolas Epaillard, Ikuan Sam, and Nadine Benhamouda
- Abstract
Purpose:CD70 is a costimulatory molecule known to activate CD27-expressing T cells. CD27–CD70 interaction leads to the release of soluble CD27 (sCD27). Clear-cell renal cell carcinoma (ccRCC) expresses the highest levels of CD70 among all solid tumors; however, the clinical consequences of CD70 expression remain unclear.Experimental Design:Tumor tissue from 25 patients with ccRCC was assessed for the expression of CD27 and CD70 in situ using multiplex immunofluorescence. CD27+ T-cell phenotypes in tumors were analyzed by flow cytometry and their gene expression profile were analyzed by single-cell RNA sequencing then confirmed with public data. Baseline sCD27 was measured in 81 patients with renal cell carcinoma (RCC) treated with immunotherapy (35 for training cohort and 46 for validation cohort).Results:In the tumor microenvironment, CD27+ T cells interacted with CD70-expressing tumor cells. Compared with CD27− T cells, CD27+ T cells exhibited an apoptotic and dysfunctional signature. In patients with RCC, the intratumoral CD27–CD70 interaction was significantly correlated with the plasma sCD27 concentration. High sCD27 levels predicted poor overall survival in patients with RCC treated with anti–programmed cell death protein 1 in both the training and validation cohorts but not in patients treated with antiangiogenic therapy.Conclusions:In conclusion, we demonstrated that sCD27, a surrogate marker of T-cell dysfunction, is a predictive biomarker of resistance to immunotherapy in RCC. Given the frequent expression of CD70 and CD27 in solid tumors, our findings may be extended to other tumors.
- Published
- 2023
3. Supplementary Table from Plasma CD27, a Surrogate of the Intratumoral CD27–CD70 Interaction, Correlates with Immunotherapy Resistance in Renal Cell Carcinoma
- Author
-
Eric Tartour, Stéphane Oudard, Yann A. Vano, Olivier Adotevi, Arnaud Mejean, Julien Adam, Sandrine Katsahian, Laurence Albiges, Nathalie Chaput, Bernd Jabla, Dominique Helley, Laetitia Mauge, Eléonore De Guillebon, Patrice Ravel, Chloe Broudin, Clémence Granier, Sebastien Mella, Valentina Libri, Virginie Verkarre, Camille Nevoret, Valentin Quiniou, Milena Hasan, Joséphine Pineau, Antonin Saldmann, Nadège Gruel, Hang Phuong Pham, Letuan Phan, Alain Gey, Nicolas Epaillard, Ikuan Sam, and Nadine Benhamouda
- Abstract
Supplementary Table from Plasma CD27, a Surrogate of the Intratumoral CD27–CD70 Interaction, Correlates with Immunotherapy Resistance in Renal Cell Carcinoma
- Published
- 2023
4. Supplementary Data from Plasma CD27, a Surrogate of the Intratumoral CD27–CD70 Interaction, Correlates with Immunotherapy Resistance in Renal Cell Carcinoma
- Author
-
Eric Tartour, Stéphane Oudard, Yann A. Vano, Olivier Adotevi, Arnaud Mejean, Julien Adam, Sandrine Katsahian, Laurence Albiges, Nathalie Chaput, Bernd Jabla, Dominique Helley, Laetitia Mauge, Eléonore De Guillebon, Patrice Ravel, Chloe Broudin, Clémence Granier, Sebastien Mella, Valentina Libri, Virginie Verkarre, Camille Nevoret, Valentin Quiniou, Milena Hasan, Joséphine Pineau, Antonin Saldmann, Nadège Gruel, Hang Phuong Pham, Letuan Phan, Alain Gey, Nicolas Epaillard, Ikuan Sam, and Nadine Benhamouda
- Abstract
Supplementary Data from Plasma CD27, a Surrogate of the Intratumoral CD27–CD70 Interaction, Correlates with Immunotherapy Resistance in Renal Cell Carcinoma
- Published
- 2023
5. Data from A First-in-Human Phase I Study of INVAC-1, an Optimized Human Telomerase DNA Vaccine in Patients with Advanced Solid Tumors
- Author
-
Pierre Langlade-Demoyen, Thierry Huet, Valérie Doppler, Simon Wain-Hobson, Stephane Oudard, Stephane Culine, Rémy Defrance, Marie Escande, Elodie Pliquet, Maria Wehbe, Caroline Laheurte, Mara Brizard, Jean-Jacques Kiladjian, Zineb Ghrieb, Marie-Agnès Dragon Durey, Ludovic Doucet, Olivier Adotevi, Julie Garibal, Jacques Medioni, and Luis Teixeira
- Abstract
Purpose:Human telomerase reverse transcriptase (hTERT) is highly expressed in >85% of human tumors and is thus considered as a good tumor-associated antigen candidate for vaccine development. We conducted a phase I study to investigate the safety, tolerability, clinical response, and immunogenicity of INVAC-1, a DNA plasmid encoding a modified hTERT protein in patients with relapsed or refractory solid tumors.Patients and Methods:INVAC-1 was either administered by intradermal route followed by electroporation or by Tropis, a needle-free injection system. Safety and tolerability were monitored by clinical and laboratory assessments. Progression-free survival and overall survival were reported using Kaplan–Meier survival analysis. Immunogenicity was studied by ELISpot, Luminex, and Flow Cytometry.Results:Twenty-six patients were treated with INVAC-1 administered at three dose levels (100, 400, and 800 μg). Vaccination was well tolerated and no dose-limiting toxicity was reported. One treatment-related grade 3 SAE was reported. Fifty-eight percent of patients experienced disease stabilization. PFS was 2.7 months, median OS was 15 months, and 1-year survival was reached for 65% of patients. INVAC-1 vaccination stimulated specific anti-hTERT CD4 T-cell response as well as cytotoxic CD8 T-cell response. No evidence of peripheral vaccine-induced immunosuppression was observed.Conclusions:INVAC-1 vaccination was safe, well tolerated, and immunogenic when administered intradermally at the three tested doses in patients with relapsed or refractory cancers. Disease stabilization was observed for the majority of patients (58%) during the treatment period and beyond.See related commentary by Slingluff Jr, p. 529
- Published
- 2023
6. Supplementary Data from CML Hematopoietic Stem Cells Expressing IL1RAP Can Be Targeted by Chimeric Antigen Receptor–Engineered T Cells
- Author
-
Christophe Ferrand, Marina Deschamps, Olivier Adotevi, Sebastien Tabruyn, Francine Garnache-Ottou, Etienne Daguindau, Sabeha Biichle, Severine Valmary-Degano, Marius Moldovan, Denis Caillot, Cyril Faure, Ziad Fajloun, Eric Deconinck, Rim Trad, Mathieu Neto Da Rocha, Fabrice Larosa, and Walid Warda
- Abstract
Additonnal data (Material & Method)
- Published
- 2023
7. Supplementary Table 2 from A First-in-Human Phase I Study of INVAC-1, an Optimized Human Telomerase DNA Vaccine in Patients with Advanced Solid Tumors
- Author
-
Pierre Langlade-Demoyen, Thierry Huet, Valérie Doppler, Simon Wain-Hobson, Stephane Oudard, Stephane Culine, Rémy Defrance, Marie Escande, Elodie Pliquet, Maria Wehbe, Caroline Laheurte, Mara Brizard, Jean-Jacques Kiladjian, Zineb Ghrieb, Marie-Agnès Dragon Durey, Ludovic Doucet, Olivier Adotevi, Julie Garibal, Jacques Medioni, and Luis Teixeira
- Abstract
Histological diagnosis, age, number of cycles received and Overall Survival
- Published
- 2023
8. Supplementary Figure 3 from A First-in-Human Phase I Study of INVAC-1, an Optimized Human Telomerase DNA Vaccine in Patients with Advanced Solid Tumors
- Author
-
Pierre Langlade-Demoyen, Thierry Huet, Valérie Doppler, Simon Wain-Hobson, Stephane Oudard, Stephane Culine, Rémy Defrance, Marie Escande, Elodie Pliquet, Maria Wehbe, Caroline Laheurte, Mara Brizard, Jean-Jacques Kiladjian, Zineb Ghrieb, Marie-Agnès Dragon Durey, Ludovic Doucet, Olivier Adotevi, Julie Garibal, Jacques Medioni, and Luis Teixeira
- Abstract
Immunophenotyping of circulating CD4 and CD8 T cells
- Published
- 2023
9. Supplementary Table 3 from A First-in-Human Phase I Study of INVAC-1, an Optimized Human Telomerase DNA Vaccine in Patients with Advanced Solid Tumors
- Author
-
Pierre Langlade-Demoyen, Thierry Huet, Valérie Doppler, Simon Wain-Hobson, Stephane Oudard, Stephane Culine, Rémy Defrance, Marie Escande, Elodie Pliquet, Maria Wehbe, Caroline Laheurte, Mara Brizard, Jean-Jacques Kiladjian, Zineb Ghrieb, Marie-Agnès Dragon Durey, Ludovic Doucet, Olivier Adotevi, Julie Garibal, Jacques Medioni, and Luis Teixeira
- Abstract
Number and percentage of patients reporting studytreatment-related AEs per cycle
- Published
- 2023
10. Data from Prognostic Value of Angiopoietin-2 for Death Risk Stratification in Patients with Metastatic Colorectal Carcinoma
- Author
-
Christophe Borg, Franck Bonnetain, Olivier Adotevi, Lise Queiroz, Julie Leger, Anthony Gonçalves, Serge Fratte, Olivier Bouché, Stefano Kim, Yann Godet, Franck Monnien, François Ghiringhelli, Erion Dobi, Thierry Lecomte, Dewi Vernerey, and Marine Jary
- Abstract
Background: Baseline prognostic biomarkers stratifying treatment strategies in first-line metastatic colorectal cancer (mCRC) are lacking. Angiopoietin-2 (Ang-2) is proposed as a potential biomarker in several cancers. We therefore decided to establish the additional prognostic value of Ang-2 for overall survival (OS) in patients with first-line mCRC.Methods: We enrolled 177 patients treated with a bevacizumab containing chemotherapy in two prospective phase II clinical trials. Patient plasma samples were collected at baseline. ELISAs were used to measure Ang-2.Results: The multivariable Cox model identified increased lactate dehydrogenase [HR, 1.60; 95% confidence interval (CI), 1.04–2.45; P = 0.03] and Ang-2 log-transformation level (HR, 1.59; 95% CI, 1.14–2.21; P = 0.0065) as two significant independent OS prognostic factors. It exhibited good calibration (P = 0.8) and discrimination (C-index: 0.64; 95% CI, 0.58–0.68). Ang-2 parameter inclusion in the GERCOR reference model significantly and strongly improved its discriminative ability because the C-statistic increased significantly from 0.61 to 0.63 (bootstrap mean difference = 0.07; 95% CI, 0.069–0.077). Interestingly, the addition of Ang-2 binary information with a 5 ng/mL cutoff value to the GERCOR model allowed the reclassification of intermediate-risk profile patients (41%) into two subsets of low and high risks.Conclusions: Our study provides robust evidence in favor of baseline Ang-2 prognostic value for OS adding to the conventional factors. Its assessment appears to be useful for the improvement in risk stratification for patients with intermediate-risk profile.Impact: Ang-2 ability to predict OS at diagnosis could be of interest in the selection of patients eligible for intermittent or sequential therapeutic strategies dedicated to the optimization of patients' quality of life and chemotherapy cost-effectiveness. Cancer Epidemiol Biomarkers Prev; 24(3); 603–12. ©2015 AACR.
- Published
- 2023
11. Supplementary Data. Figure Legends S1-S12 & Tables S1-S4 from CML Hematopoietic Stem Cells Expressing IL1RAP Can Be Targeted by Chimeric Antigen Receptor–Engineered T Cells
- Author
-
Christophe Ferrand, Marina Deschamps, Olivier Adotevi, Sebastien Tabruyn, Francine Garnache-Ottou, Etienne Daguindau, Sabeha Biichle, Severine Valmary-Degano, Marius Moldovan, Denis Caillot, Cyril Faure, Ziad Fajloun, Eric Deconinck, Rim Trad, Mathieu Neto Da Rocha, Fabrice Larosa, and Walid Warda
- Abstract
Figure S1. In vitro detection (using #A3C3) of recombinant IL-1RAP protein by ELISA Figure S2. Procedure for generating IL-1RAP CART cells and CD4/CD8 ratio at the end of the process Figure S3. Staining of IL-1RAP CAR at the cell surface of T cell line or primary T -lymphocytes Figure S4. Impact of the IL-1RAP soluble form on IL-1RAP CART-cells toxicity Figure S5. Tissue macroarray (TMA) using #A3C3 monoclonal antibody Figure S6. Experimental immunosafety human CD34+ engrafted NOG murine model Figure S7. Colony Forming Unit (CFU-GM) experiment after coculture of autologous CART-cells with CD34+ HSC. Figure S8. In-vitro and in-vivo iCASP/AP1903 suicide gene safety switch Figure S9. Production and characterization of IL-1RAP (soluble and membrane) transfected cell line Figure S10. Cytokine secretion profiling of IL-1RAP CART-cells after co-culturing with targets Figure S11. Tumoral xenograft murine model Figure S12. Autologous cytotoxicity of IL-1RAP CART-cells directed against initial CML disease Table S1: Antibodies and kits Table S2: IL-1RAP (mAb#A3C3) immunostaining of normal tissues Table S3: Percentage of alive cells in different subpopulations according co-culture with different ratio of E (MockT-cells or IL-1RAP CART-cells) :T. Table S4: CML patient's characteristics with available PBSC cryopreserved autografts
- Published
- 2023
12. Supplementary Table 5 from A First-in-Human Phase I Study of INVAC-1, an Optimized Human Telomerase DNA Vaccine in Patients with Advanced Solid Tumors
- Author
-
Pierre Langlade-Demoyen, Thierry Huet, Valérie Doppler, Simon Wain-Hobson, Stephane Oudard, Stephane Culine, Rémy Defrance, Marie Escande, Elodie Pliquet, Maria Wehbe, Caroline Laheurte, Mara Brizard, Jean-Jacques Kiladjian, Zineb Ghrieb, Marie-Agnès Dragon Durey, Ludovic Doucet, Olivier Adotevi, Julie Garibal, Jacques Medioni, and Luis Teixeira
- Abstract
Clinical features of patients with OS 1 year
- Published
- 2023
13. Supplementary data (Figures S1 to S12) from CML Hematopoietic Stem Cells Expressing IL1RAP Can Be Targeted by Chimeric Antigen Receptor–Engineered T Cells
- Author
-
Christophe Ferrand, Marina Deschamps, Olivier Adotevi, Sebastien Tabruyn, Francine Garnache-Ottou, Etienne Daguindau, Sabeha Biichle, Severine Valmary-Degano, Marius Moldovan, Denis Caillot, Cyril Faure, Ziad Fajloun, Eric Deconinck, Rim Trad, Mathieu Neto Da Rocha, Fabrice Larosa, and Walid Warda
- Abstract
Figure S1. In vitro detection (using #A3C3) of recombinant IL-1RAP protein by ELISA Figure S2. Procedure for generating IL-1RAP CART cells and CD4/CD8 ratio at the end of the process Figure S3. Staining of IL-1RAP CAR at the cell surface of T cell line or primary T -lymphocytes Figure S4. Impact of the IL-1RAP soluble form on IL-1RAP CART-cells toxicity Figure S5. Tissue macroarray (TMA) using #A3C3 monoclonal antibody Figure S6. Experimental immunosafety human CD34+ engrafted NOG murine model Figure S7. Colony Forming Unit (CFU-GM) experiment after coculture of autologous CART-cells with CD34+ HSC. Figure S8. In-vitro and in-vivo iCASP/AP1903 suicide gene safety switch Figure S9. Production and characterization of IL-1RAP (soluble and membrane) transfected cell line Figure S10. Cytokine secretion profiling of IL-1RAP CART-cells after co-culturing with targets Figure S11. Tumoral xenograft murine model Figure S12. Autologous cytotoxicity of IL-1RAP CART-cells directed against initial CML disease
- Published
- 2023
14. Supplementary Figure 1 from A First-in-Human Phase I Study of INVAC-1, an Optimized Human Telomerase DNA Vaccine in Patients with Advanced Solid Tumors
- Author
-
Pierre Langlade-Demoyen, Thierry Huet, Valérie Doppler, Simon Wain-Hobson, Stephane Oudard, Stephane Culine, Rémy Defrance, Marie Escande, Elodie Pliquet, Maria Wehbe, Caroline Laheurte, Mara Brizard, Jean-Jacques Kiladjian, Zineb Ghrieb, Marie-Agnès Dragon Durey, Ludovic Doucet, Olivier Adotevi, Julie Garibal, Jacques Medioni, and Luis Teixeira
- Abstract
Flow Chart
- Published
- 2023
15. Data from CML Hematopoietic Stem Cells Expressing IL1RAP Can Be Targeted by Chimeric Antigen Receptor–Engineered T Cells
- Author
-
Christophe Ferrand, Marina Deschamps, Olivier Adotevi, Sebastien Tabruyn, Francine Garnache-Ottou, Etienne Daguindau, Sabeha Biichle, Severine Valmary-Degano, Marius Moldovan, Denis Caillot, Cyril Faure, Ziad Fajloun, Eric Deconinck, Rim Trad, Mathieu Neto Da Rocha, Fabrice Larosa, and Walid Warda
- Abstract
Chronic myeloid leukemia (CML) is a chronic disease resulting in myeloid cell expansion through expression of the BCR-ABL1 fusion transcript. Tyrosine kinase inhibitors (TKI) have significantly increased survival of patients with CML, and deep responders may consider stopping the treatment. However, more than 50% of patients relapse and restart TKI, subsequently suffering unknown toxicity. Because CML is a model immune system–sensitive disease, we hypothesize that chimeric antigen receptor (CAR) T cells targeting IL1 receptor-associated protein (IL1RAP) in quiescent CML stem cells may offer an opportunity for a permanent cure. In this study, we produced and molecularly characterized a specific monoclonal anti-IL1RAP antibody from which fragment antigen-binding nucleotide coding sequences were cloned as a single chain into a lentiviral backbone and secured with the suicide gene iCASP9/rimiducid system. Our CAR T-cell therapy exhibited cytotoxicity against both leukemic stem cells and, to a lesser extent, monocytes expressing IL1RAP, with no apparent effect on the hematopoietic system, including CD34+ stem cells. This suggests IL1RAP as a tumor-associated antigen for immunotherapy cell targeting. IL1RAP CAR T cells were activated in the presence of IL1RAP+ cell lines or primary CML cells, resulting in secretion of proinflammatory cytokines and specifically killing in vitro and in a xenograft murine model. Overall, we demonstrate the proof of concept of a CAR T-cell immunotherapy approach in the context of CML that is applicable for young patients and primary TKI-resistant, intolerant, or allograft candidate patients.Significance:These findings present the first characterization and proof of concept of a chimeric antigen receptor directed against IL1RAP expressed by leukemic stem cells in the context of CML.
- Published
- 2023
16. Supplementary Figure 2 from A First-in-Human Phase I Study of INVAC-1, an Optimized Human Telomerase DNA Vaccine in Patients with Advanced Solid Tumors
- Author
-
Pierre Langlade-Demoyen, Thierry Huet, Valérie Doppler, Simon Wain-Hobson, Stephane Oudard, Stephane Culine, Rémy Defrance, Marie Escande, Elodie Pliquet, Maria Wehbe, Caroline Laheurte, Mara Brizard, Jean-Jacques Kiladjian, Zineb Ghrieb, Marie-Agnès Dragon Durey, Ludovic Doucet, Olivier Adotevi, Julie Garibal, Jacques Medioni, and Luis Teixeira
- Abstract
Kaplan-Meier representation of Overall Survival and Progression Free Survival
- Published
- 2023
17. Data not shown from A First-in-Human Phase I Study of INVAC-1, an Optimized Human Telomerase DNA Vaccine in Patients with Advanced Solid Tumors
- Author
-
Pierre Langlade-Demoyen, Thierry Huet, Valérie Doppler, Simon Wain-Hobson, Stephane Oudard, Stephane Culine, Rémy Defrance, Marie Escande, Elodie Pliquet, Maria Wehbe, Caroline Laheurte, Mara Brizard, Jean-Jacques Kiladjian, Zineb Ghrieb, Marie-Agnès Dragon Durey, Ludovic Doucet, Olivier Adotevi, Julie Garibal, Jacques Medioni, and Luis Teixeira
- Abstract
This file compiles all the results identified as "data not shown" in the manuscript
- Published
- 2023
18. Supplementary Table 1 from A First-in-Human Phase I Study of INVAC-1, an Optimized Human Telomerase DNA Vaccine in Patients with Advanced Solid Tumors
- Author
-
Pierre Langlade-Demoyen, Thierry Huet, Valérie Doppler, Simon Wain-Hobson, Stephane Oudard, Stephane Culine, Rémy Defrance, Marie Escande, Elodie Pliquet, Maria Wehbe, Caroline Laheurte, Mara Brizard, Jean-Jacques Kiladjian, Zineb Ghrieb, Marie-Agnès Dragon Durey, Ludovic Doucet, Olivier Adotevi, Julie Garibal, Jacques Medioni, and Luis Teixeira
- Abstract
Flow Cytometry antibody references
- Published
- 2023
19. Supplementary Table 4 from A First-in-Human Phase I Study of INVAC-1, an Optimized Human Telomerase DNA Vaccine in Patients with Advanced Solid Tumors
- Author
-
Pierre Langlade-Demoyen, Thierry Huet, Valérie Doppler, Simon Wain-Hobson, Stephane Oudard, Stephane Culine, Rémy Defrance, Marie Escande, Elodie Pliquet, Maria Wehbe, Caroline Laheurte, Mara Brizard, Jean-Jacques Kiladjian, Zineb Ghrieb, Marie-Agnès Dragon Durey, Ludovic Doucet, Olivier Adotevi, Julie Garibal, Jacques Medioni, and Luis Teixeira
- Abstract
Best Overall Response
- Published
- 2023
20. Supplementary Methods, Table 1 from Prognostic Value of Angiopoietin-2 for Death Risk Stratification in Patients with Metastatic Colorectal Carcinoma
- Author
-
Christophe Borg, Franck Bonnetain, Olivier Adotevi, Lise Queiroz, Julie Leger, Anthony Gonçalves, Serge Fratte, Olivier Bouché, Stefano Kim, Yann Godet, Franck Monnien, François Ghiringhelli, Erion Dobi, Thierry Lecomte, Dewi Vernerey, and Marine Jary
- Abstract
Supplementary Methods, Table 1. Supplementary methods: - Ang-2 plasma sample measurement - Statistical analysis interpretation Supplementary table 1: - Cox Univariate analyses for OS prediction with and without the stratified approach (sensitivity analysis) Supplementary references
- Published
- 2023
21. Antitumor CAR T-cell Screening Platform: Many Are Called, but Few Are Chosen
- Author
-
Jeanne Galaine and Olivier Adotevi
- Subjects
Cancer Research ,Oncology - Abstract
Treatment with T cells expressing chimeric antigen receptors (CAR) is a promising anticancer therapy. However, this approach has several limitations and has not yet been effectively applied to treat solid tumors. The study by Panowski and colleagues represents the first comparative analysis of multiple single chain fragment variable (scFv)-based anti-CD70 CAR T-cell clones for the development of a clinical product to treat renal cell carcinoma (RCC). Despite the risk of T-cell fratricide due to CD70 expression on T cells, CD70 CAR T cells were produced successfully thanks to the protective CD70 masking phenomenon. Two distinct classes of CAR T cells were identified with different memory phenotypes, activation statuses, and cytotoxic activity. CD70 CAR T cells presented high cytotoxic activity against RCC both in vitro in RCC cell lines and in vivo in patient-derived xenograft mouse models. The off-target effects expected on the lymphoid compartment were confirmed by tissue cross-reactivity staining and in a cynomolgus monkey preclinical model with CD3-CD70 bispecific antibody treatment. The efficacy and the toxicity profile of the lead CD70 CAR T-cell candidate instigated the researchers to proceed with upscaled clinical production. This article emphasizes the influence of the scFv of the CARs on their efficacy:toxicity balance. Ultimately, they successfully managed to develop a highly effective CAR T-cell candidate to treat a solid tumor by an allogeneic approach, thereby overcoming two major hurdles to broaden application of CAR T-cell therapy. See related article by Panowski et al., p. 2610
- Published
- 2022
22. A First-in-Human Phase I Study of INVAC-1, an Optimized Human Telomerase DNA Vaccine in Patients with Advanced Solid Tumors
- Author
-
Zineb Ghrieb, Ludovic Doucet, Stéphane Culine, Mara Brizard, Marie-Agnès Dragon Durey, Jacques Medioni, Thierry Huet, Rémy DeFrance, Maria Wehbe, Luis Augusto Teixeira, Julie Garibal, Pierre Langlade-Demoyen, Olivier Adotevi, Jean-Jacques Kiladjian, Elodie Pliquet, Marie Escande, Valérie Doppler, Caroline Laheurte, Stéphane Oudard, and Simon Wain-Hobson
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,ELISPOT ,medicine.medical_treatment ,Immunogenicity ,Immunosuppression ,DNA vaccination ,Vaccination ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Tolerability ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,Telomerase reverse transcriptase ,business ,Survival analysis - Abstract
Purpose: Human telomerase reverse transcriptase (hTERT) is highly expressed in >85% of human tumors and is thus considered as a good tumor-associated antigen candidate for vaccine development. We conducted a phase I study to investigate the safety, tolerability, clinical response, and immunogenicity of INVAC-1, a DNA plasmid encoding a modified hTERT protein in patients with relapsed or refractory solid tumors. Patients and Methods: INVAC-1 was either administered by intradermal route followed by electroporation or by Tropis, a needle-free injection system. Safety and tolerability were monitored by clinical and laboratory assessments. Progression-free survival and overall survival were reported using Kaplan–Meier survival analysis. Immunogenicity was studied by ELISpot, Luminex, and Flow Cytometry. Results: Twenty-six patients were treated with INVAC-1 administered at three dose levels (100, 400, and 800 μg). Vaccination was well tolerated and no dose-limiting toxicity was reported. One treatment-related grade 3 SAE was reported. Fifty-eight percent of patients experienced disease stabilization. PFS was 2.7 months, median OS was 15 months, and 1-year survival was reached for 65% of patients. INVAC-1 vaccination stimulated specific anti-hTERT CD4 T-cell response as well as cytotoxic CD8 T-cell response. No evidence of peripheral vaccine-induced immunosuppression was observed. Conclusions: INVAC-1 vaccination was safe, well tolerated, and immunogenic when administered intradermally at the three tested doses in patients with relapsed or refractory cancers. Disease stabilization was observed for the majority of patients (58%) during the treatment period and beyond. See related commentary by Slingluff Jr, p. 529
- Published
- 2020
23. Immunoregulation and Clinical Implications of ANGPT2/TIE2+ M-MDSC Signature in Non–Small Cell Lung Cancer
- Author
-
Caroline Laheurte, Laura Boullerot, Olivier Adotevi, L. Rangan, Magalie Dosset, Elodie Lauret Marie Joseph, Marine Jary, Adeline Bouard, Eléonore Gravelin, Christophe Borg, and Kamal Asgarov
- Subjects
0301 basic medicine ,Cancer Research ,business.industry ,medicine.medical_treatment ,Immunology ,Immunosuppression ,Immunotherapy ,medicine.disease ,Immune tolerance ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Immune system ,Antigen ,Cancer immunotherapy ,030220 oncology & carcinogenesis ,cardiovascular system ,medicine ,Cancer research ,Carcinoma ,Lung cancer ,business - Abstract
Myeloid-derived suppressor cells (MDSC) promote immunosuppression and are a target in the field of immuno-oncology. Accumulation of MDSCs is associated with poor prognosis and resistance to immunotherapy for several cancers. Here, we describe an accumulation of a subset of circulating monocytic MDSCs (M-MDSC) overexpressing TIE2, the receptor for angiopoietin-2 (ANGPT2), in patients with non–small cell lung cancer (NSCLC). Greater numbers of circulating TIE2+ M-MDSCs were detected in patients with NSCLC compared with healthy subjects, and this accumulation correlated with ANGPT2 concentration in blood. The presence of an ANGPT2-rich environment was associated with impairment of preexisting T-cell responses against tumor-associated antigens (TAA) in patients with NSCLC. We demonstrated that ANGPT2 sensitizes TIE2+ M-MDSCs such that these cells suppress TAA-specific T cells. In patients with NSCLC, upregulation of the ANGPT2/TIE2+ M-MDSC signature in blood was associated with a poor prognosis. Our results identify the ANGPT2/TIE2+ M-MDSC axis as a participant in tumor immune evasion that should be taken into account in future cancer immunotherapy.
- Published
- 2020
24. CML Hematopoietic Stem Cells Expressing IL1RAP Can Be Targeted by Chimeric Antigen Receptor–Engineered T Cells
- Author
-
Christophe Ferrand, Etienne Daguindau, Francine Garnache-Ottou, Sabeha Biichle, Cyril Faure, Mathieu Neto Da Rocha, Denis Caillot, Eric Deconinck, Fabrice Larosa, Séverine Valmary-Degano, Olivier Adotevi, Marina Deschamps, Rim Trad, Sébastien Tabruyn, Ziad Fajloun, Marius Moldovan, and Walid Warda
- Subjects
Cytotoxicity, Immunologic ,0301 basic medicine ,Cancer Research ,Myeloid ,T-Lymphocytes ,medicine.medical_treatment ,CD34 ,Biology ,Immunotherapy, Adoptive ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,hemic and lymphatic diseases ,medicine ,Animals ,Humans ,Molecular Targeted Therapy ,Cell Engineering ,Mice, Inbred BALB C ,Receptors, Chimeric Antigen ,Antibodies, Monoclonal ,Immunotherapy ,Suicide gene ,Hematopoietic Stem Cells ,Xenograft Model Antitumor Assays ,Chimeric antigen receptor ,Haematopoiesis ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Stem cell ,Interleukin-1 Receptor Accessory Protein - Abstract
Chronic myeloid leukemia (CML) is a chronic disease resulting in myeloid cell expansion through expression of the BCR-ABL1 fusion transcript. Tyrosine kinase inhibitors (TKI) have significantly increased survival of patients with CML, and deep responders may consider stopping the treatment. However, more than 50% of patients relapse and restart TKI, subsequently suffering unknown toxicity. Because CML is a model immune system–sensitive disease, we hypothesize that chimeric antigen receptor (CAR) T cells targeting IL1 receptor-associated protein (IL1RAP) in quiescent CML stem cells may offer an opportunity for a permanent cure. In this study, we produced and molecularly characterized a specific monoclonal anti-IL1RAP antibody from which fragment antigen-binding nucleotide coding sequences were cloned as a single chain into a lentiviral backbone and secured with the suicide gene iCASP9/rimiducid system. Our CAR T-cell therapy exhibited cytotoxicity against both leukemic stem cells and, to a lesser extent, monocytes expressing IL1RAP, with no apparent effect on the hematopoietic system, including CD34+ stem cells. This suggests IL1RAP as a tumor-associated antigen for immunotherapy cell targeting. IL1RAP CAR T cells were activated in the presence of IL1RAP+ cell lines or primary CML cells, resulting in secretion of proinflammatory cytokines and specifically killing in vitro and in a xenograft murine model. Overall, we demonstrate the proof of concept of a CAR T-cell immunotherapy approach in the context of CML that is applicable for young patients and primary TKI-resistant, intolerant, or allograft candidate patients. Significance: These findings present the first characterization and proof of concept of a chimeric antigen receptor directed against IL1RAP expressed by leukemic stem cells in the context of CML.
- Published
- 2019
25. Abstract CT021: INVAC-1, an optimized telomerase DNA vaccine in patients with advanced solid tumors: Final results of first-in-human phase I study
- Author
-
Valérie Doppler, Olivier Adotevi, Thierry Huet, Jacques Medioni, Claire Germain, Luis Augusto Teixeira, Caroline Laheurte, Marie Escande, Julie Garibal, Pierre Langlade Demoyen, Marie-Agnès Dragon-Durey, and Maria Wehbe
- Subjects
Cancer Research ,Telomerase ,business.industry ,medicine.medical_treatment ,Cancer ,Immunosuppression ,medicine.disease ,Tumor antigen ,DNA vaccination ,Immune system ,Oncology ,medicine ,Cancer research ,Telomerase reverse transcriptase ,business ,Survival rate - Abstract
Background INVAC-1 is an optimized DNA plasmid encoding an inactive form of human Telomerase Reverse Transcriptase (hTERT), a universal tumor antigen expressed in most of human tumors with little or no expression in normal somatic cells. Primary pharmacodynamics, safety and toxicology studies showed that INVAC-1 was enzymatically inactive, immunogenically safe and well tolerated. In murine models, we demonstrated that INVAC-1 was able to induce hTERT specific cellular immune responses with CD4+ Th1 effector and memory CD8+ T-cells as well as slow tumor growth and increase survival rate by 50% in tumor-bearing mice. Methods We conducted a First-In-Human (FIH) study, 2-centre, Phase I, open label, 3+3 escalation design and multiple dose study examining the safety and tolerability of INVAC-1 administered at three dose levels (100, 400 and 800 µg) in 26 patients with relapsed or solid refractory tumors. INVAC-1 was administered either by intradermal (ID) injection followed by electroporation (EP) (n=20) or by Tropis® Needle Free Injection System (n=6). Results INVAC-1 vaccination was safe and well tolerated when administered ID (either with EP or by Tropis®) at the three tested doses. Only one treatment-related grade 3 SAE was reported. 58% of patients experienced disease stabilization up to 9.9 months. One-year survival was reached for 65% of patients. INVAC-1 elicited both hTERT specific Th1-dominant CD4 and cytotoxic CD8 T cell responses with no vaccine-induced peripheral immunosuppression. Anti-hTERT immune responses were enhanced by adding anti-PD-1 immune checkpoint inhibitor ex vivo. In addition, INVAC-1 vaccination was able to promote epitope spreading. Finally, correlation analysis between clinical and immunological data showed that patients with OS >1 year presented a significantly higher hTERT immune response after INVAC-1 vaccination compared to patients with OS Citation Format: Julie Garibal, Jacques Medioni, Luis Teixeira, Olivier Adotevi, Marie-Agnès Dragon-Durey, Caroline Laheurte, Claire Germain, Marie Escande, Maria Wehbe, Valérie Doppler, Thierry Huet, Pierre Langlade Demoyen. INVAC-1, an optimized telomerase DNA vaccine in patients with advanced solid tumors: Final results of first-in-human phase I study [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr CT021.
- Published
- 2021
26. Abstract 4476: PD-1 and TIGIT co-expression identifies a circulating CD8 T cell population predictive of response to anti-PD-1 therapy in melanoma and Merkel-cell carcinoma patients
- Author
-
Stanley R. Riddell, Régis Josien, Camille Dabrowski, Olivier Adotevi, Virginie Vignard, Nathalie Labarrière, Raphael Gottardo, Amir Khammari, Tiffany Beauvais, Valentin Voillet, Cécile Braudeau, Steven P. Fling, Nirasha Ramchurren, Charles Nardin, Cheever Martin, Samuel Rulli, Brigitte Dréno, Paul Nghiem, Zhong Wu, Caroline Laheurte, Francçois Aubin, Nicolas Jouand, Sylvain Simon, and Candice D. Church
- Subjects
Cancer Research ,education.field_of_study ,Tumor microenvironment ,business.industry ,T cell ,Melanoma ,Population ,Cancer ,medicine.disease ,medicine.anatomical_structure ,Oncology ,TIGIT ,Cancer research ,Medicine ,Cytotoxic T cell ,business ,education ,CD8 - Abstract
Immunotherapies targeting the PD-1 pathway have profoundly transformed the clinical care of cancer patients for a growing variety of cancer types. However, most patients do not experience durable clinical benefit. The definition of robust and convenient biomarkers of PD-1 therapy efficacy to stratify patients beforehand or early after initiation of the therapy that could guide therapeutic management is still lacking while being a very active research field. Biomarkers described to date include tumor burden, neoantigen load, presence and number of PD-1+ CD8+ at the tumor margin, T-cell inflamed tumor microenvironment and PD-L1 expression by the tumor cells or other immune cells and composition of the gut microbiota. Most of these parameters are closely related/influenced by the presence, activation status and functional capacities of CD8+ T cells infiltrating the tumor site demonstrating their pivotal role for anti-PD-1 mediated anti-tumor efficacy. A population of PD-1high CD8 TILs was consequently described as predictive of PD-1 blockade in NSCLC. The exact contribution for clinical efficacy of TILs versus distinct CD8+ T cells from peripheral origins recirculating to the tumor site remains to be elucidated. Notably, immunological responses to PD-1 blockade at the periphery were described within the very first days following the first therapy dose. Therefore, describing circulating cellular population predictive of PD-1 inhibitor efficacy could represent a convenient, non-invasive and rapid method to assess anti-tumor benefits. Original findings reported in this study identified a circulating CD8 T cell population delineated by the co-expression of TIGIT and PD-1 inhibitory receptors as an early immune marker of anti-PD-1 efficacy in three independent cohorts of cancer patients (two melanoma patient's cohorts and one Merkel-cell carcinoma patient's cohort). The frequency of this double positive (DPOS) population even appeared predictive of PD-1 inhibitor therapy efficacy at baseline in the MCC cohort. Furthermore, to understand the mechanistical relevance of this subset for PD-1 blockade efficacy, we thoroughly described this DPOS T cell subset by flow cytometry, gene expression analysis, anti-tumor reactivity assay and TCR repertoire analysis, and compared it to its double negative (DNEG), PD-1 and TIGIT single positive counterparts. This DPOS subset was enriched in activated and proliferative T cells, retained expression of co-stimulatory molecules and was enriched for common features with Tfc. Moreover, this subpopulation exhibited a specific gene signature, strongly predictive of long-term survival in melanoma patients (TCGA analyses). We demonstrated that this subpopulation was enriched in tumor-specific T-cells (ELISPOT analysis against 11 antigen-derived peptides). Finally, clustering of TCR clonotypes revealed that the DPOS T cell population was significantly enriched in emerging clonotypes in responding patients, after 1 month of anti-PD-1 therapy echoing recent work from others. Our findings provide a compelling rationale to measure PD-1+TIGIT+ CD8 T-cell subset in the blood of cancer patients to monitor early anti-PD1 mediated clinical efficacy, and to use DPOS T cells as a window to study the dynamic changes that underly successful antitumor immunity. Citation Format: Sylvain Simon, Valentin Voillet, Virginie Vignard, Zhong Wu, Camille Dabrowski, Nicolas Jouand, Tiffany Beauvais, Amir Khammari, Cecile Braudeau, Regis Josien, Olivier Adotevi, Caroline Laheurte, Francçois Aubin, Charles Nardin, Samuel Rulli, Raphaël Gottardo, Nirasha Ramchurren, Cheever Martin, Steven P. Fling, Candice D. Church, Paul Nghiem, Brigitte Dreno, Stanley R. Riddell, Nathalie Labarriere. PD-1 and TIGIT co-expression identifies a circulating CD8 T cell population predictive of response to anti-PD-1 therapy in melanoma and Merkel-cell carcinoma patients [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 4476.
- Published
- 2020
27. Abstract 575: Adaptive CD4 Th1 response against telomerase in blood counteracts T-cell exhaustion in non-small cell lung cancer
- Author
-
Caroline Laheurte, Elodie Lauret Marie Joseph, Magalie Dosset, Olivier Adotevi, Laurie Cuche, Dewi Vernerey, Marion Jacquin, Laura Boullerot, Virginie Westeel, P. Jacoulet, Vincent Kaulec, and Guillaume Eberst
- Subjects
Cancer Research ,Telomerase ,business.industry ,ELISPOT ,T cell ,Cancer ,Human leukocyte antigen ,medicine.disease ,Immune system ,medicine.anatomical_structure ,Oncology ,Cancer research ,medicine ,Cytotoxic T cell ,business ,Lung cancer - Abstract
Despite the critical roles played by IFN-γ+ CD4 Th1 response in tumor immunity, the translation of their potential in clinic remains challenging. Here, we evaluate the clinical significance of circulating anti-tumor CD4 Th1 response in non-small cell lung cancer (NSCLC) patients. 170 naïve-treatment patients were enrolled in this immune monitoring study. The antitumor adaptive Th1 response was assessed by IFN-γ ELISPOT assay in blood lymphocytes using a mixture of eight highly promiscuous and Th1-polarized HLA class II-restricted epitopes from telomerase (TERT). The presence of anti-TERT Th1 response was detected in 59/170 patients (35%). We found an opposite link between anti-TERT Th1 and hyper exhausted T cells co-expressing PD-1+ and TIM-3+ in NSCLC patients. In contrast to hyper exhausted CD8 T cells, the presence of anti-TERT Th1 response was associated with a low rate of hyper exhausted CD4 T cells. We showed that NSCLC stage dissemination is associated with a decrease of anti-TERT Th1 response but an increase of circulating hyper exhausted PD-1+/TIM-3+ CD4 T cells. Notably, anti-telomerase Th1 response and hyper exhausted CD4 T-cells displayed opposite prognosis value in NSCLC. While high level of anti-TERT Th1 cells play a protective role, hyper-exhausted PD-1+TIM-3+ CD4 T cells negatively affect patients’ survival. By using these two circulating immune factors, we stratified patients in distinct prognostic group. Patients with anti-TERT Th1high/CD4 PD-1 TIM-3low immune profile had better overall survival than anti-TERT Th1low/CD4 PD-1 TIM-3high group (median OS : not reached versus 4 months respectively p Citation Format: Caroline Laheurte, Magalie Dosset, Dewi Vernerey, Elodie Lauret Marie Joseph, Laura Boullerot, Vincent Kaulec, Marion Jacquin, Laurie Cuche, Guillaume Eberst, Pascale Jacoulet, Virginie Westeel, Olivier Adotevi. Adaptive CD4 Th1 response against telomerase in blood counteracts T-cell exhaustion in non-small cell lung cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 575.
- Published
- 2019
28. Immunogenic HLA-B*0702-Restricted Epitopes Derived from Human Telomerase Reverse Transcriptase That Elicit Antitumor Cytotoxic T-Cell Responses
- Author
-
Emmanuelle Boulanger, François Lemonnier, Karine Mollier, Pierre Langlade-Demoyen, Pierre Charneau, Olivier Adotevi, Sylvain Cardinaud, Wolf-Hervé Fridman, Blandine Mignen, Maurizio Zanetti, Christine Neuveut, and Eric Tartour
- Subjects
Cancer Research ,Telomerase ,medicine.medical_treatment ,Epitopes, T-Lymphocyte ,Mice, Transgenic ,Cancer Vaccines ,Epitope ,Cell Line ,HLA-B7 Antigen ,Mice ,Cancer immunotherapy ,Neoplasms ,MHC class I ,medicine ,Animals ,Humans ,Cytotoxic T cell ,Telomerase reverse transcriptase ,Vaccines, Synthetic ,biology ,Immunogenicity ,Vaccination ,DNA ,Virology ,Peptide Fragments ,DNA-Binding Proteins ,CTL ,Oncology ,HLA-B Antigens ,Cancer research ,biology.protein ,Immunotherapy ,Plasmids - Abstract
Purpose: The human telomerase reverse transcriptase (hTERT) is considered as a potential target for cancer immunotherapy because it is preferentially expressed in tumor cells. To increase the applicability of hTERT-based immunotherapy, we set out to identify CTL epitopes in hTERT restricted by HLA-B*0702 molecule, a common MHC class I allele.Experimental Design: HLA-B*0702-restricted peptides from hTERT were selected by using a method of epitope prediction and tested for their immunogenicity in human (in vitro) and HLA-B*0702 transgenic mice (in vivo).Results: All the six hTERT peptides that were predicted to bind to HLA-B*0702 molecule were found to induce primary human CTL responses in vitro. The peptide-specific CD8+ CTL lines were tested against various hTERT+ tumor cells. Although differences were observed according to the tumor origin, only three CTL lines specific for p277, p342, and p351 peptides exhibited cytotoxicity against tumor cells in a HLA-B*0702-restricted manner. In addition, this cytotoxicity was inhibited by the addition of peptide-loaded cold target cells and indicated that these epitopes are naturally processed and presented on the tumor cells. Further, in vivo studies using humanized HLA-B*0702 transgenic mice showed that all the candidate peptides were able to induce CTL responses after peptide immunization. Furthermore, vaccination with a plasmid DNA encoding full-length hTERT elicited peptide-specific CTL responses, indicating that these epitopes are efficiently processed in vivo.Conclusions: Together with previously reported hTERT epitopes, the identification of new CTL epitopes presented by HLA-B*0702 increases the applicability of hTERT-based immunotherapy to treating cancer.
- Published
- 2006
29. Abstract 2588: Antiangiogenic and immunomodulatory effects of metronomic cyclophosphamide (CPM) treatment in prostate cancer patients with PSA failure
- Author
-
Yann Godet, Caroline Laheurte, Olivier Adotevi, Christophe Borg, Fabien Calcagno, Tristan Maurina, Stefano Kim, and Antoine Thiery-Vuillemin
- Subjects
Oncology ,Gynecology ,Cancer Research ,medicine.medical_specialty ,Performance status ,Cyclophosphamide ,business.industry ,Prostatectomy ,medicine.medical_treatment ,Cancer ,Phases of clinical research ,medicine.disease ,Metronomic Chemotherapy ,Prostate cancer ,PSA Failure ,Internal medicine ,Medicine ,business ,medicine.drug - Abstract
Background: After curative local therapy, patients (pts) will experience rising PSA as an early indicator of recurrent prostate cancer and no standard of care exists. Previous studies demonstrated the clinical interest of cyclophosphamide metronomic chemotherapy (CMC) in several cancers. Multifactorial modes of action including immunological and anti-angiogenic effects have been well described by Kerbel et al and Ghiringhelli et al (Cancer Research 2006 and European Journal of immunology 2004, respectively). In the current study, we investigated the safety and immuno-modulatory effects of CMC in prostate cancer patients with PSA failure (biochemical relapse). Methods: We conducted a prospective phase II study to characterize the clinical and immunological interest of CMC in histologically proven prostate cancer patients previously treated by prostatectomy and with biochemical relapse (defined as PSA > 1 and < 20ng/mL and progressive PSA level on 3 different measures). CMC was administered per os, at daily dose of 50 mg during 6 months. PSA level, serum VEGF and immune parameters were monitored every month in blood samples. Results: Thirty-four consecutive pts were enrolled in this study and all received CMC. The median age was 68,3 (range, 56,9-82,1) and all pts had good performance status (ECOG-PS = 0). Gleason Score was 7 in 58%, ≤6 in 13%, and ≥ 8 in 13%. No serious adverse events (grade 3-5) were observed. The most common drug-related adverse event was grade 1 lymphopenia. Baseline PSA was 2,92 ng/mL (range, 1,1-9,35). Twenty-two pts presented stable PSA (64.7%), one patient had partial response defined by >50% decline in the serum PSA (3%), and 11 pts (32%) had PSA progression before 6 months. The immuno-monitoring performed in all pts who completed 6 months CMC. A decrease of immunosuppressive regulatory T cells (Treg) was observed in 56% (10/18). In addition, high rate of activated HLA-DR+ cytotoxic CD8 T cells was also detected in these pts. Furthermore, serum VEGF level decreased at month 1 but it returned to baseline level at the end of treatment in 9/17 pts evaluated (52%). In some patients the correlation was observed between the control of PSA level, the decrease of Treg and VEGF parameters. Conclusions: This study demonstrated the clinical benefit and the ability of CMC to promote the blockage of angiogenesis and immuno-modulatory effect in prostate cancer patients treated for a biochemical relapse. It will be of interest to combine CMC to vaccination in these patients. Citation Format: Olivier Adotevi, Fabien Calcagno, Tristan Maurina, Stefano Kim, Antoine Thiery-Vuillemin, Christophe Borg, Yann Godet, Caroline Laheurte. Antiangiogenic and immunomodulatory effects of metronomic cyclophosphamide (CPM) treatment in prostate cancer patients with PSA failure. [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 2588. doi:10.1158/1538-7445.AM2014-2588
- Published
- 2014
30. Abstract 4780: CD4 T-lymphocyte responses to the human telomerase reverse transcriptase (hTERT) in patients (pts) with advanced non small cell lung cancer (NSCLC)
- Author
-
Olivier Adotevi, Patrice Ravel, Marc Riquet, Magali Dosset, Guy Meyer, Jacques Medioni, Aurélie Cazes, Emeline Leviennois, Eric Tartour, and Elizabeth Fabre
- Subjects
Oncology ,Cancer Research ,Chemotherapy ,medicine.medical_specialty ,business.industry ,medicine.medical_treatment ,Cancer ,non-small cell lung cancer (NSCLC) ,Immunotherapy ,medicine.disease ,Internal medicine ,Immunology ,medicine ,Adenocarcinoma ,Telomerase reverse transcriptase ,Stage (cooking) ,business ,Progressive disease - Abstract
Background: hTERT is a potential target for cancer immunotherapy because it is highly expressed in tumor cells. To assess the applicability of hTERT-based immunotherapy in NSCLC, we aimed to analyse the natural anti-hTERT CD4 T-lymphocyte responses in advanced NSCLC pts. Methods: We included in a prospective, monocentric study chemonaive NSCLC pts. Pts were required to present stage III or IV tumor, without any immune deficiency or immunosuppressive drug. Before start of chemotherapy, the anti-hTERT T-lymphocyte responses were assessed in whole blood by ELISPOT test. We evaluated the presence or absence of hTERT-specific CD4 T lymphocytes. Thereafter, we analyzed its link with age (< 70 vs. ≥ 70 yrs), ECOG performance status PS (0-1 vs. 2-3), tumoral stage (IIIA vs. IIIb + IV), histological subtype (adenocarcinoma vs. other) and smoking status (never smoker vs. smoker). Statistical analysis was performed using chi-Square or Fischer test. Results: Between February 2008 and October 2009, fifty-one NSCLC pts were included. They presented a stage IIIa (n = 5), IIIb (n = 5) or IV (n = 41) disease. Median age was 66 yrs [39-89]. Nineteen pts presented anti-telomerase CD4 T lymphocytes (37 %); among them we found 14 stage IV (73 %), 13 pts < 70 yrs (68 %), 12 pts with PS 0/1 (63 %), 18 adenocarcinoma/undifferentiated carcinoma (94 %), 13 pts with stable disease/ partial response (68%). Furthermore, all non smokers have anti-telomerase immune response. Conclusions: These preliminary results demonstrate that natural anti-hTERT immune response exists in NSCLC pts, even in case of advanced disease. However the presence of immune response is more frequently found in pts less than 70 yrs with good PS, in non squamous cell carcinoma and in pts with non progressive disease. The study is ongoing to evaluate if baseline anti-hTERT immune response could be used as selection criteria for telomerase vaccination in NSCLC. Note: This abstract was not presented at the AACR 101st Annual Meeting 2010 because the presenter was unable to attend. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 4780.
- Published
- 2010
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.