90 results on '"Adotevi, O."'
Search Results
2. LBA6 UCPVax therapeutic vaccination promotes cytotoxic and Th1 polarized antitumor CD4 T cells and epitope spreading in patients with advanced non-small cell lung cancer
- Author
-
Adotevi, O., Laheurte, C., Boullerot, L., Malfroy, M., Richard, T., Cuche, L., Seffar, E., Kaulek, V., Jacquin, M., Orillard, E., Eberst, G., Jandus, C., Westeel, V., Borg, C., and Godet, Y.
- Published
- 2022
- Full Text
- View/download PDF
3. 210P Anti-CTLA-4 overcomes inhibitory effect of PD1high regulatory T cell on circulating antitumor CD4+ Th1 response in patients treated by anti-PD(L)-1
- Author
-
Marguier, A., Perreira, V., Wespiser, M., Lecoester, B., Boullerot, L., Malfroy, M., Nardin, C., Laheurte, C., Orillard, E., and Adotevi, O.
- Published
- 2022
- Full Text
- View/download PDF
4. 203P Early remodeling of systemic antitumor T cell immunity in head and neck cancer patients treated by chemoradiation
- Author
-
Lecoester, B., Seffar, E., Marguier, A., Renaudin, A., Richard, T., Wespiser, M., Malfroy, M., Boullerot, L., Arnier, R., Martin, E., Sun, X., Laheurte, C., Boustani, J., and Adotevi, O.
- Published
- 2022
- Full Text
- View/download PDF
5. 180P Recalling pre-existing microbiota-specific T cells to target tumors
- Author
-
Carpier, J-M., Talpin, A., Kulakowski, G., Dos Santos Leite, A.M., Laviolette, K., Noguerol, J., Mersceman, T., Garnier, A., Gaal, C., Pereira Oliveira, C., Calderon, C., Boullerot, L., Malfroy, M., Strozzi, F., Gouttefangeas, C., Adotevi, O., Joffre, O., Gamelas Magalhaes, J., Chêne, L., and Bonny, C.
- Published
- 2022
- Full Text
- View/download PDF
6. Synthetic and natural non-live vectors: rationale for their clinical development in cancer vaccine protocols
- Author
-
Tartour, E, Benchetrit, F, Haicheur, N, Adotevi, O, and Fridman, W.H
- Published
- 2002
- Full Text
- View/download PDF
7. Use of cancer vaccine after immunotherapy failure: a promising strategy for advanced NSCLC patients with secondary resistance to checkpoint inhibitors
- Author
-
Adotévi, O.
- Published
- 2023
- Full Text
- View/download PDF
8. 1903P - Myeloid derived suppressor cells but not regulatory T cells are associated with adaptive immunity and clinical outcomes in anal squamous cell carcinoma
- Author
-
Borg, C., Spehner, L., Kroemer, M., Kim, S., Jary, M., Vienot, A., Klajer, E., Ramseyer, M., Adotevi, O., Godet, Y., Andre, T., Buecher, B., Francois, E., and Loyon, R.
- Published
- 2019
- Full Text
- View/download PDF
9. 1789P - Physicians’ satisfaction with health-related quality of life (HRQoL) assessment in daily clinical practice using electronic patient-reported outcome (ePRO) for cancer patients
- Author
-
Mouillet, G., Fritzsch, J., Thiery-Vuillemin, A., Meneveau, N., Almotlak, H., Mansi, L., Curtit, E., Kim, S., Jary, M., Maurina, T., Eberst, G., Barthelemy, P., Eymard, J.-C., Geoffrois, L., Djoumakh, O., Anota, A., Adotevi, O., Westeel, V., and Paget-Bailly, S.
- Published
- 2019
- Full Text
- View/download PDF
10. P1.14-19 Exome Analysis of Patients Treated with Afatinib Reveals Genetic Variations Discriminating Extreme Responders
- Author
-
Niogret, J., Kaderbhai, C., Chevrier, S., Chapusot, C., Truntzer, C., Mananet, H., Foucher, P., Coudert, B., Lagrange, A., Favier, L., Martin, L., Almotlak, H., Jacoulet, P., Lahourcade, J., Adotevi, O., Westeel, V., Arnould, L., and Boidot, R.
- Published
- 2019
- Full Text
- View/download PDF
11. 30TiP - Predictive markers of immune mediated adverse events and of treatment response in patients treated with durvalumab monotherapy or in combination with tremelimumab (IOPREDI study)
- Author
-
Marabelle, A., Ghiringhelli, F., Ayyoub, M., Tartour, E., Adotevi, O., Loriot, Y., Lambotte, O., Cupissol, D., Damotte, D., Adam, J., Bonvalet, M., Petre Lazar, B., Licour, M., and Chaput, N.
- Published
- 2018
- Full Text
- View/download PDF
12. 92P - Understanding the anti-tumor T cell responses modulation mediated by chemoradiation to improve immunotherapy efficacy
- Author
-
Lauret, E., Bonin, C., Rangan, L., Boullerot, L., Boustani, J., Fagnoni-Legat, C., Perrin, S., Mirjolet, C., Dosset, M., Boidot, R., Servagi, S., De Bari, B., and Adotevi, O.
- Published
- 2018
- Full Text
- View/download PDF
13. 40P - Interest and potential of genetically engineered cord blood derived T cells for cancer immunotherapy
- Author
-
Marton, C., Mercier-Letondal, P., Deschamps, M., Ferrand, C., Adotevi, O., Borg, C., Galaine, J., and Godet, Y.
- Published
- 2018
- Full Text
- View/download PDF
14. 1356P - The level of circulating NKp46+ CD56dim CD16+ natural killer cells predicts distinct survival in non-small cell lung cancer
- Author
-
Picard, E., Godet, Y., Laheurte, C., Boullerot, L., Lauret Marie Joseph, E., Jacquin, M., Kaulek, V., Eberst, G., Gaugler, B., Jacoulet, P., Gainet-Brun, M., Lahoucarde, J., Almotlak, H., Le Pimpec-Barthes, F., Fabre-Guillevin, E., Borg, C., Westeel, V., and Adotevi, O.
- Published
- 2018
- Full Text
- View/download PDF
15. 415PD - Clinical response and pharmacodynamic assessment of INVAC-1, a DNA plasmid encoding an inactive form of human telomerase reverse transcriptase (hTERT), on immune responses, immune tolerability, tumor burden and circulating tumor DNA (ctDNA) in patients with advanced solid tumors
- Author
-
Medioni, J., Brizard, M., Teixeira, L.O., Doucet, L., Ghrieb, Z., Angelergues, A., Oudard, S., Culine, S., Adotevi, O., Laheurte, C., Dragon-Durey, M.-A., Laurent-Puig, P., Kiladjian, J.-J., Doppler, V., Souttou, B., Wain-Hobson, S., Defrance, R., Huet, T., and Langlade-Demoyen, P.
- Published
- 2018
- Full Text
- View/download PDF
16. 1656P - Myeloid-derived suppressor cells are associated with a decrease of tumor antigen-specific Th1 immunity in non-small cell lung cancer
- Author
-
Adotevi, O., Lauret Marie Joseph, E., Laheurte, C., Dosset, M., Fabre-Guillevin, E., Jacoulet, P., Eberts, G., Helluin, G., Boullerot, L., Rangan, L., Godet, Y., and Westeel, V.
- Published
- 2017
- Full Text
- View/download PDF
17. B subunit of shiga toxin-based vaccines synergize with alpha-galactosylceramide to break tolerance against self antigen and elicit antiviral immunity
- Author
-
Adotevi, O., Vingert, B., Freyburger, L., Shrikant, P., Y.C., Lone, Quintin-Colonna, F., Haicheur, N., Amessou, M., Herbelin, A., Langlade-Demoyen, P., W.H., Fridman, Lemonnier, F., Cytokines, hématopoïèse et réponse immune (CHRI), Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Slama, Catherine, and Université Paris Descartes - Paris 5 (UPD5) - Institut National de la Santé et de la Recherche Médicale (INSERM) - Centre National de la Recherche Scientifique (CNRS)
- Subjects
ComputingMilieux_MISCELLANEOUS - Abstract
International audience
- Published
- 2007
18. 1081P - The efficacy of everolimus relies on a modulation of adaptative anti tumor T cell immunity
- Author
-
Adotévi, O., Mansi, L., Beziaud, L., Ravel, P., Marie-Joseph, E. Lauret, Laheurte, C., Rangan, L., Maurina, T., Mouillet, G., Hon, T. Nguyen Tan, Curtit, E., Pivot, X., Godet, Y., Borg, C., and Thiery-Vuillemin, A.
- Published
- 2016
- Full Text
- View/download PDF
19. 211P - Prognostic Value of Angiopoietin-2 for Overall Survival in Patients with Metastatic Colorectal Carcinoma Treated By Chemotherapy and Bevacizumab
- Author
-
Jary, M., Vernerey, D., Dobi, E., Monnien, F., Godet, Y., Kim, S., Bouché, O., Fratte, S., Ghiringhelli, F., Goncalvez, A., Lecomte, T., Leger, J., Queiroz, L., Adotevi, O., Bonnetain, F., and Borg, C.
- Published
- 2014
- Full Text
- View/download PDF
20. In situ delivery of allogeneic natural killer cell (NK) combined with Cetuximab in liver metastases of gastrointestinal carcinoma: A phase I clinical trial.
- Author
-
Adotevi, O., Godet, Y., Galaine, J., Lakkis, Z., Idirene, I., Certoux, J. M., Jary, M., Loyon, R., Laheurte, C., Kim, S., Dormoy, A., Pouthier, F., Barisien, C., Fein, F., Tiberghien, P., Pivot, X., Valmary-Degano, S., Ferrand, C., Morel, P., and Delabrousse, E.
- Subjects
- *
KILLER cells , *CETUXIMAB , *GASTROINTESTINAL cancer - Abstract
Despite successful introduction of NK-based cellular therapy in the treatment of myeloid leukemia, the potential use of NK alloreactivity in solid malignancies is still elusive. We performed a phase I clinical trial to assess the safety and efficacy of
in situ delivery of allogeneic NK cells combined with cetuximab in liver metastasis of gastrointestinal origin. The conditioning chemotherapy was administrated before the allogeneic NK cells injection via hepatic artery. Three escalating doses were tested (3.106, 8.106 and 12.106 NK cells/kg) following by a high-dose interleukin-2 (IL-2). Cetuximab was administered intravenously every week for 7 weeks. Nine patients with liver metastases of colorectal or pancreatic cancers were included, three per dose level. Hepatic artery injection was successfully performed in all patients with no report of dose-limiting toxicity. Two patients had febrile aplasia requiring a short-term antibiotherapy. Grade 3/4 anemia and thrombopenia were also observed related to the chemotherapy. Objective clinical responses were documented in 3 patients and among them 2 occurred in patients injected with cell products harboring two KIR ligand mismatches and one in a patient with one KIR ligand mismatch. Immune monitoring revealed that most patients presented an increase but transient of IL-15 and IL-7 cytokines levels one week after chemotherapy. Furthermore, a high expansion of FoxP3+regulatory T cells and PD-1+ T cells was observed in all patients, related to IL-2 administration. Our results demonstrated that combining allogeneic NK cells transfer via intra-hepatic artery, cetuximab and a high-dose IL-2 is feasible, well tolerated and may result in clinical responses. [ABSTRACT FROM AUTHOR]- Published
- 2018
- Full Text
- View/download PDF
21. PD-L1 expression is regulated by both DNA methylation and NF-kB during EMT signaling in non-small cell lung carcinoma.
- Author
-
Asgarova, A., Asgarov, K., Godet, Y., Peixoto, P., Nadaradjane, A., Boyer-Guittaut, M., Galaine, J., Guenat, D., Mougey, V., Perrard, J., Pallandre, J. R., Bouard, A., Balland, J., Tirole, C., Adotevi, O., Hendrick, E., Herfs, M., Cartron, P. F., Borg, C., and Hervouet, E.
- Subjects
DNA methylation ,NON-small-cell lung carcinoma - Abstract
Tumor cells, which undergo Epithelial-mesenchymal transition (EMT) acquire increased capacities of proliferation, invasion and have the ability to generate metastases by escaping the immune system during their systemic migration. To escape the immune system, cancer cells may induce tolerance or resist elimination by immune effectors
via multiple mechanisms and we hypothesized that EMT may control the expression of immune checkpoint inhibitors, then promoting immune evasion. PD-L1 (programmed cell death ligand 1) but not PD-L2 nor Galectin 9 or Death receptor (DR4, DR5 and Fas) and ligands (FasL and TRAIL) expression was up-regulated during cytokine-driven EMT in a reversible manner. Moreover PD-L1 is overexpressed in VIMENTIN positive NSCLC tissues. We also demonstrated that the expression of PD-L1 required both TNFα and TGFβ1. Indeed, TGFβ1 decreased DNMT1 content and that resulted inPD-L1 promoter demethylation whereas TNFα induced the NF-κB pathway that promoted expression of demethylatedPD-L1 promoter. [ABSTRACT FROM AUTHOR]- Published
- 2018
- Full Text
- View/download PDF
22. THBS1 + myeloid cells expand in SLD hepatocellular carcinoma and contribute to immunosuppression and unfavorable prognosis through TREM1.
- Author
-
Giraud J, Chalopin D, Ramel E, Boyer T, Zouine A, Derieppe MA, Larmonier N, Adotevi O, Le Bail B, Blanc JF, Laurent C, Chiche L, Derive M, Nikolski M, and Saleh M
- Subjects
- Humans, Triggering Receptor Expressed on Myeloid Cells-1, Immunosuppression Therapy, Myeloid Cells, Immunosuppressive Agents, Inflammation, Carcinoma, Hepatocellular, Liver Neoplasms
- Abstract
Hepatocellular carcinoma (HCC) is an inflammation-associated cancer arising from viral or non-viral etiologies including steatotic liver diseases (SLDs). Expansion of immunosuppressive myeloid cells is a hallmark of inflammation and cancer, but their heterogeneity in HCC is not fully resolved and might underlie immunotherapy resistance. Here, we present a high-resolution atlas of innate immune cells from patients with HCC that unravels an SLD-associated contexture characterized by influx of inflammatory and immunosuppressive myeloid cells, including a discrete population of THBS1
+ regulatory myeloid (Mreg ) cells expressing monocyte- and neutrophil-affiliated genes. THBS1+ Mreg cells expand in SLD-associated HCC, populate fibrotic lesions, and are associated with poor prognosis. THBS1+ Mreg cells are CD163+ but distinguished from macrophages by high expression of triggering receptor expressed on myeloid cells 1 (TREM1), which contributes to their immunosuppressive activity and promotes HCC tumor growth in vivo. Our data support myeloid subset-targeted immunotherapies to treat HCC., Competing Interests: Declaration of interests M.D. is co-founder, chief scientific officer, and employee of Inotrem SA, a French company that develops TREM1 inhibitors., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
23. First-line durvalumab and tremelimumab with chemotherapy in RAS-mutated metastatic colorectal cancer: a phase 1b/2 trial.
- Author
-
Thibaudin M, Fumet JD, Chibaudel B, Bennouna J, Borg C, Martin-Babau J, Cohen R, Fonck M, Taieb J, Limagne E, Blanc J, Ballot E, Hampe L, Bon M, Daumoine S, Peroz M, Mananet H, Derangère V, Boidot R, Michaud HA, Laheurte C, Adotevi O, Bertaut A, Truntzer C, and Ghiringhelli F
- Subjects
- Humans, Antibodies, Monoclonal therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Colorectal Neoplasms drug therapy, Colorectal Neoplasms genetics, Colorectal Neoplasms pathology
- Abstract
Although patients with microsatellite instable metastatic colorectal cancer (CRC) benefit from immune checkpoint blockade, chemotherapy with targeted therapies remains the only therapeutic option for microsatellite stable (MSS) tumors. The single-arm, phase 1b/2 MEDITREME trial evaluated the safety and efficacy of durvalumab plus tremelimumab combined with mFOLFOX6 chemotherapy in first line, in 57 patients with RAS-mutant unresectable metastatic CRC. Safety was the primary objective of phase Ib; no safety issue was observed. The phase 2 primary objective of efficacy in terms of 3-month progression-free survival (PFS) in patients with MSS tumors was met, with 3-month PFS of 90.7% (95% confidence interval (CI): 79.2-96%). For secondary objectives, response rate was 64.5%; median PFS was 8.2 months (95% CI: 5.9-8.6); and overall survival was not reached in patients with MSS tumors. We observed higher tumor mutational burden and lower genomic instability in responders. Integrated transcriptomic analysis underlined that high immune signature and low epithelial-mesenchymal transition were associated with better outcome. Immunomonitoring showed induction of neoantigen and NY-ESO1 and TERT blood tumor-specific T cell response associated with better PFS. The combination of durvalumab-tremelimumab with mFOLFOX6 was tolerable with promising clinical activity in MSS mCRC. Clinicaltrials.gov identifier: NCT03202758 ., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
24. Evaluation of the interest to combine a CD4 Th1-inducer cancer vaccine derived from telomerase and atezolizumab plus bevacizumab in unresectable hepatocellular carcinoma: a randomized non-comparative phase II study (TERTIO - PRODIGE 82).
- Author
-
Vienot A, Jacquin M, Rebucci-Peixoto M, Pureur D, Ghiringhelli F, Assenat E, Hammel P, Rosmorduc O, Stouvenot M, Allaire M, Bouattour M, Regnault H, Fratte S, Raymond E, Soularue E, Husson-Wetzel S, Di Martino V, Muller A, Clairet AL, Fagnoni-Legat C, Adotevi O, Meurisse A, Vernerey D, and Borg C
- Subjects
- Humans, Bevacizumab, Tumor Microenvironment, Cancer Vaccines adverse effects, Carcinoma, Hepatocellular drug therapy, Liver Neoplasms drug therapy, Lung Neoplasms, Telomerase
- Abstract
Background: Several cancer immunotherapies that target the PD-L1/PD-1 pathway show promising clinical activity in patients with hepatocellular carcinoma (HCC). However, the standard of care in first-line treatment with atezolizumab (anti-PD-L1 therapy) in combination with bevacizumab is associated with a limited objective response rate. Telomerase reverse transcriptase (TERT) activation meets the criteria of oncogenic addiction in HCC and could be actionable therapeutic target and a relevant tumor antigen. Therefore we hypothesized that combining anti-PD-1/PD-L1 therapy with an anti-telomerase vaccine might be an attractive therapy in HCC. UCPVax is a therapeutic cancer vaccine composed of two separate peptides derived from telomerase (human TERT). UCPVax has been evaluated in a multicenter phase I/II study in non-small cell lung cancers and has demonstrated to be safe and immunogenic, and is under evaluation in combination with atezolizumab in a phase II clinical trial in tumors where telomerase reactivation contributes to an oncogene addiction (HPV
+ cancers). The aim of the TERTIO study is to determine the clinical interest and immunological efficacy of a treatment combining the CD4 helper T-inducer cancer anti-telomerase vaccine (UCPVax) with atezolizumab and bevacizumab in unresectable HCC in a multicenter randomized phase II study., Methods: Patients with locally advanced, metastatic or unresectable HCC who have not previously received systemic anti-cancer treatment are eligible. The primary end point is the objective response rate at 6 months. Patients will be allocated to a treatment arm with a randomization 2:1. In both arms, patients will receive atezolizumab at fixed dose of 1200 mg IV infusion and bevacizumab at fixed dose of 15 mg/kg IV infusion, every 3 weeks, according to the standard of care. In the experimental arm, these treatments will be combined with the UCPVax vaccine at 0.5 mg subcutaneously., Discussion: Combining anti-PD-1/PD-L1 therapy with an anti-telomerase vaccine gains serious consideration in HCC, in order to extend the clinical efficacy of anti-PD-1/PD-L1. Indeed, anti-cancer vaccines can induce tumor-specific T cell expansion and activation and therefore restore the cancer-immunity cycle in patients lacking pre-existing anti-tumor responses. Thus, there is a strong rational to combine immune checkpoint blockade therapy and anticancer vaccine (UCPVax) in order to activate antitumor T cell immunity and bypass the immunosuppression in the tumor microenvironment in HCC. This pivotal proof of concept study will evaluate the efficacy and safety of the combination of a CD4 Th1-inducer cancer vaccine derived from telomerase (UCPVax) and atezolizumab plus bevacizumab in unresectable HCC, as well as confirming their synergic mechanism, and settling the basis for a new combination for future clinical trials., Trial Registration: NCT05528952., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
25. Durable response of lung carcinoma patients to EGFR tyrosine kinase inhibitors is determined by germline polymorphisms in some immune-related genes.
- Author
-
Dalens L, Niogret J, Richard C, Chevrier S, Foucher P, Coudert B, Lagrange A, Favier L, Westeel V, Kim S, Adotevi O, Chapusot C, Martin L, Arnould L, Kaderbhai CG, and Boidot R
- Subjects
- Humans, Tyrosine Kinase Inhibitors, Germ Cells, Lung, ErbB Receptors genetics, Lung Neoplasms drug therapy, Lung Neoplasms genetics, Carcinoma, Non-Small-Cell Lung drug therapy, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma
- Abstract
Background: Non-small cell lung cancer is a very poor prognosis disease. Molecular analyses have highlighted several genetic alterations which may be targeted by specific therapies. In clinical practice, progression-free survival on EGFR TKI treatment is between 12 and 14 months. However, some patients progress rapidly in less than 6 months, while others remain free of progression for 16 months or even longer during EGFR TKI treatment., Methods: We sequenced tumor exomes from 135 lung cancer patients (79 with EGFR-wildtype (WT), 56 with EGFR-mutant tumors) enrolled in the ALCAPONE trial (genomic analysis of lung cancers by next generation sequencing for personalized treatment)., Results: Some germline polymorphisms were enriched in the EGFR-mutant subset compared to EGFR-WT tumors or to a reference population. However, the most interesting observation was the negative impact of some germline SNPs in immunity-related genes on survival on EGFR TKI treatment. Indeed, the presence of one of three particular SNPs in the HLA-DRB5 gene was associated with a decreased PFS on EGFR TKI. Moreover, some SNPs in the KIR3DL1 and KIR3DL2 genes were linked to a decrease in both progression-free and overall survival of patients with EGFR-mutant tumors., Conclusion: Our data suggest that SNPs in genes expressed by immune cells may influence the response to targeted treatments, such as EGFR TKIs. This indicates that the impact of these cells may not be limited to modulating the response to immunotherapies. Further studies are needed to determine the exact mechanisms underlying this influence and to identify the associated predictive and prognostic markers that would allow to refine treatments and so improve lung cancer patient outcomes., Trial Registration: NCT02281214: NGS Genome Analysis in Personalization of Lung Cancer Treatment (ALCAPONE)., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
26. Production of functional plasmacytoid dendritic cells - targeted chimeric antigen receptor T cells from patients with immune-mediated inflammatory diseases.
- Author
-
Caël B, Bôle-Richard E, Galaine J, Adotevi O, Garnache-Ottou F, and Aubin F
- Subjects
- Humans, Dendritic Cells, Cells, Cultured, T-Lymphocytes, Receptors, Chimeric Antigen
- Abstract
Competing Interests: Conflicts of interest all authors declare no conflicts of interest.
- Published
- 2023
- Full Text
- View/download PDF
27. Development of a new heparan sulfate proteoglycan (HSPG) chromolith LC column to study the pH dependence binding of peptide vaccines to HSPG and role of human serum albumin on its binding.
- Author
-
André C, Lethier L, Adotevi O, and Guillaume YC
- Subjects
- Humans, Histidine, Cell Membrane, Peptides, Heparan Sulfate Proteoglycans, Serum Albumin, Human
- Abstract
Heparan sulfate proteoglycan (HSPG) expressed on immune cell surface participate in antitumor T-cell responses generated in the acidic lymph node (LN) microenvironment. In this work, HSPG was immobilized for the first time on a HPLC chromolith support for studying the effect of extra cellular acidosis in LNs on the binding to HSPG of two peptide vaccines (universal cancer peptide UCP2 and UCP4). This home-made HSPG column enabling to work at high flow-rates, was resistance to change in pH, had a long - life time, an excellent repeatability and negligible non-specific binding sites. The performance of this affinity HSPG column was confirmed by the evaluation of recognition assay for a series of known ligand of HSPG. It was shown that at 37 °C, the UCP2 binding to HSPG versus pH described a sigmoidal shape while UCP4 remained relatively constant in the pH range 5.0-7.5 and lower than the one of UCP2. By the use of an HSA HPLC column, it was shown at 37 °C and in acidic conditions a loss of affinity of UCP2 and UCP4 to HSA. It was demonstrated that upon UCP2/HSA binding, the protonation of the histidine residue in the cluster R(arg) Q(Gln) Hist (H) of the UCP2 peptide allowed to expose more favorably than UCP4 its polar and cationic groups to the negative net charge of HSPG on immune cells. Acidic pHs led to the protonation of the UCP2 residue histidine by flipping the His switch to the on position with a concomitant increase in affinity for the negative net charge of HSPG confirming that UCP2 was more immunogenic than UCP4. As well this HSPG chromolith LC column developed in this work could be used in the feature for other protein - HSPG binding studies or for a separative mode.
- Published
- 2023
- Full Text
- View/download PDF
28. Interplay between plasmacytoid dendritic cells and tumor-specific T cells in peripheral blood influences long-term survival in non-small cell lung carcinoma.
- Author
-
Laheurte C, Seffar E, Gravelin E, Lecuelle J, Renaudin A, Boullerot L, Malfroy M, Marguier A, Lecoester B, Gaugler B, Saas P, Truntzer C, Ghiringhelli F, and Adotevi O
- Subjects
- Humans, T-Lymphocytes, Immune Checkpoint Inhibitors, Dendritic Cells, Tumor Microenvironment, Carcinoma, Non-Small-Cell Lung, Lung Neoplasms
- Abstract
Plasmacytoid dendritic cells (pDCs) represent a subset of antigen-presenting cells that play an ambivalent role in cancer immunity. Here, we investigated the clinical significance of circulating pDCs and their interaction with tumor-specific T cell responses in patients with non-small cell lung cancer (NSCLC, n = 126) . The relation between intratumoral pDC signature and immune checkpoint inhibitors efficacy was also evaluated. Patients with NSCLC had low level but activated phenotype pDC compared to healthy donors. In overall population, patients with high level of pDC (pDC
high ) had improved overall survival (OS) compared to patients with pDClow , median OS 30.4 versus 20.7 months (P = 0.013). This clinical benefit was only observed in stage I to III patients, but not in metastatic disease. We showed that patients harboring pDChigh profile had high amount of Th1-diffentiation cytokine interleukin-12 (IL-12) in blood and had functional T cells directed against a broad range of tumor antigens. Furthermore, a high pDC signature in the tumor microenvironment was associated with improved clinical outcome in patients treated with anti-PD-(L)1 therapy. Overall, this study showed that circulating pDChigh is associated with long-term OS in NSCLC and highlighted the predictive value of intratumor pDC signature in the efficacy of immune checkpoint inhibitors., (© 2022. The Author(s), under exclusive licence to Springer-Verlag GmbH Germany, part of Springer Nature.)- Published
- 2023
- Full Text
- View/download PDF
29. Plasma CD27, a Surrogate of the Intratumoral CD27-CD70 Interaction, Correlates with Immunotherapy Resistance in Renal Cell Carcinoma.
- Author
-
Benhamouda N, Sam I, Epaillard N, Gey A, Phan L, Pham HP, Gruel N, Saldmann A, Pineau J, Hasan M, Quiniou V, Nevoret C, Verkarre V, Libri V, Mella S, Granier C, Broudin C, Ravel P, De Guillebon E, Mauge L, Helley D, Jabla B, Chaput N, Albiges L, Katsahian S, Adam J, Mejean A, Adotevi O, Vano YA, Oudard S, and Tartour E
- Subjects
- Humans, CD27 Ligand genetics, Tumor Necrosis Factor Receptor Superfamily, Member 7 genetics, Immunotherapy, Tumor Microenvironment, Carcinoma, Renal Cell drug therapy, Carcinoma, Renal Cell genetics, Kidney Neoplasms drug therapy, Kidney Neoplasms genetics
- 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., (©2022 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
30. TIE-2 Signaling Activation by Angiopoietin 2 On Myeloid-Derived Suppressor Cells Promotes Melanoma-Specific T-cell Inhibition.
- Author
-
Marguier A, Laheurte C, Lecoester B, Malfroy M, Boullerot L, Renaudin A, Seffar E, Kumar A, Nardin C, Aubin F, and Adotevi O
- Subjects
- Angiopoietin-2 metabolism, Humans, T-Lymphocytes, Melanoma, Myeloid-Derived Suppressor Cells
- Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous group of immune suppressive cells detected in several human cancers. In this study, we investigated the features and immune suppressive function of a novel subset of monocytic MDSC overexpressing TIE-2 (TIE-2
+ M-MDSC), the receptor for the pro-angiogenic factor angiopoietin 2 (ANGPT2). We showed that patients with melanoma exhibited a higher circulating rate of TIE-2+ M-MDSCs, especially in advanced stages, as compared to healthy donors. The distribution of the TIE-2+ M-MDSC rate toward the melanoma stage correlated with the serum level of ANGPT2. TIE-2+ M-MDSC from melanoma patients overexpressed immune suppressive molecules such as PD-L1, CD73, TGF-β, and IL-10, suggesting a highly immunosuppressive phenotype. The exposition of these cells to ANGPT2 increased the expression of most of these molecules, mainly Arginase 1. Hence, we observed a profound impairment of melanoma-specific T-cell responses in patients harboring high levels of TIE-2+ M-MDSC along with ANGPT2. This was confirmed by in vitro experiments indicating that the addition of ANGPT2 increased the ability of TIE-2+ M-MDSC to suppress antitumor T-cell function. Furthermore, by using TIE-2 kinase-specific inhibitors such as regorafenib or rebastinib, we demonstrated that an active TIE-2 signaling was required for optimal suppressive activity of these cells after ANGPT2 exposition. Collectively, these results support that TIE-2+ M-MDSC/ANGPT2 axis represents a potential immune escape mechanism in melanoma., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Marguier, Laheurte, Lecoester, Malfroy, Boullerot, Renaudin, Seffar, Kumar, Nardin, Aubin and Adotevi.)- Published
- 2022
- Full Text
- View/download PDF
31. A Phase II Study Evaluating the Interest to Combine UCPVax, a Telomerase CD4 T H 1-Inducer Cancer Vaccine, and Atezolizumab for the Treatment of HPV Positive Cancers: VolATIL Study.
- Author
-
Rebucci-Peixoto M, Vienot A, Adotevi O, Jacquin M, Ghiringhelli F, de la Fouchardière C, You B, Maurina T, Kalbacher E, Bazan F, Meynard G, Clairet AL, Fagnoni-Legat C, Spehner L, Bouard A, Vernerey D, Meurisse A, Kim S, Borg C, and Mansi L
- Abstract
Background: There is a strong rational of using anti-programmed cell death protein-1 and its ligand (anti-PD-1/L1) antibodies in human papillomavirus (HPV)-induced cancers. However, anti-PD-1/L1 as monotherapy induces a limited number of objective responses. The development of novel combinations in order to improve the clinical efficacy of an anti-PD-1/L1 is therefore of interest. Combining anti-PD-1/L1 therapy with an antitumor vaccine seems promising in HPV-positive (+) cancers. UCPVax is a therapeutic cancer vaccine composed of two separate peptides derived from telomerase (hTERT, human telomerase reverse transcriptase). UCPVax is being evaluated in a multicenter phase I/II study in NSCLC (non-small cell lung cancer) and has demonstrated to be safe and immunogenic. The aim of the VolATIL study is to evaluate the combination of atezolizumab (an anti-PD-L1) and UCPVax vaccine in a multicenter phase II study in patients with HPV
+ cancers., Methods: Patients with HPV+ cancer (anal canal, head and neck, and cervical or vulvar), at locally advanced or metastatic stage, and refractory to at least one line of systemic chemotherapy are eligible. The primary end point is the objective response rate (ORR) at 4 months. Patients will receive atezolizumab every 3 weeks at a fixed dose of 1,200 mg in combination with the UCPVax vaccine at 1 mg subcutaneously., Discussion: Anti-cancer vaccines can restore cancer-immunity via the expansion and activation of tumor-specific T cells in patients lacking pre-existing anti-tumor responses. Moreover, preclinical data showed that specific TH 1 CD4 T cells sustain the quality and homing of an antigen-specific CD8+ T-cell immunity. In previous clinical studies, the induction of anti-hTERT immunity was significantly correlated to survival in patients with advanced squamous anal cell carcinoma. Thus, there is a strong rational to combine an anti-cancer hTERT vaccine and an immune checkpoint inhibitor to activate and promote antitumor T-cell immunity. This pivotal proof of concept study will evaluate the efficacy and safety of the combination of a telomerase-based TH 1 inducing vaccine (UCPVax) and an anti-PD-L1 (atezolizumab) immunotherapy in HPV+ cancers, as well as confirming their synergic mechanism, and settling the basis for a new combination for future clinical trials., Clinical Trial Registration: https://www.clinicaltrials.gov/, identifier NCT03946358., Competing Interests: CB: Research grant from Bayer and Roche, advisory board for Bayer, Sanofi, MSD. FB: Novartis, Seagen, Daiichi Sankyo, Pierre Fabre, Astra-Zeneca, Clovis. SK: reports a consulting or advisory role for Ipsen, Incyte, Boehringer Ingelheim, Sanofi and BeiGene and has received research funding from Pfizer, Roche, Novartis, Bristol-Myers Squibb, Boehringer Ingelheim and Sanofi. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Rebucci-Peixoto, Vienot, Adotevi, Jacquin, Ghiringhelli, de la Fouchardière, You, Maurina, Kalbacher, Bazan, Meynard, Clairet, Fagnoni-Legat, Spehner, Bouard, Vernerey, Meurisse, Kim, Borg and Mansi.)- Published
- 2022
- Full Text
- View/download PDF
32. Umbilical Cord Blood as a Source of Less Differentiated T Cells to Produce CD123 CAR-T Cells.
- Author
-
Caël B, Galaine J, Bardey I, Marton C, Fredon M, Biichle S, Poussard M, Godet Y, Angelot-Delettre F, Barisien C, Bésiers C, Adotevi O, Pouthier F, Garnache-Ottou F, and Bôle-Richard E
- Abstract
Chimeric Antigen Receptor (CAR) therapy has led to great successes in patients with leukemia and lymphoma. Umbilical Cord Blood (UCB), stored in UCB banks, is an attractive source of T cells for CAR-T production. We used a third generation CD123 CAR-T (CD28/4-1BB), which was previously developed using an adult's Peripheral Blood (PB), to test the ability of obtaining CD123 CAR-T from fresh or cryopreserved UCB. We obtained a cell product with a high and stable transduction efficacy, and a poorly differentiated phenotype of CAR-T cells, while retaining high cytotoxic functions in vitro and in vivo. Moreover, CAR-T produced from cryopreserved UCB are as functional as CAR-T produced from fresh UCB. Overall, these data pave the way for the clinical development of UCB-derived CAR-T. UCB CAR-T could be transferred in an autologous manner (after an UCB transplant) to reduce post-transplant relapses, or in an allogeneic setting, thanks to fewer HLA restrictions which ease the requirements for a match between the donor and recipient.
- Published
- 2022
- Full Text
- View/download PDF
33. c-Maf enforces cytokine production and promotes memory-like responses in mouse and human type 2 innate lymphoid cells.
- Author
-
Trabanelli S, Ercolano G, Wyss T, Gomez-Cadena A, Falquet M, Cropp D, Imbratta C, Leblond MM, Salvestrini V, Curti A, Adotevi O, Jandus C, and Verdeil G
- Subjects
- Animals, Cytokines metabolism, Humans, Interleukin-33 genetics, Interleukin-33 metabolism, Lung metabolism, Mice, Papain metabolism, Proto-Oncogene Proteins c-maf metabolism, Immunity, Innate, Lymphocytes metabolism
- Abstract
Group-2 innate lymphoid cells (ILC2s), which are involved in type 2 inflammatory diseases such as allergy, can exhibit immunological memory, but the basis of this ILC2 "trained immunity" has remained unclear. Here, we found that stimulation with IL-33/IL-25 or exposure to the allergen papain induces the expression of the transcription factor c-Maf in mouse ILC2s. Chronic papain exposure results in high production of IL-5 and IL-13 cytokines and lung eosinophil recruitment, effects that are blocked by c-Maf deletion in ILCs. Transcriptomic analysis revealed that knockdown of c-Maf in ILC2s suppresses expression of type 2 cytokine genes, as well as of genes linked to a memory-like phenotype. Consistently, c-Maf was found highly expressed in human adult ILC2s but absent in cord blood and required for cytokine production in isolated human ILC2s. Furthermore, c-Maf-deficient mouse or human ILC2s failed to exhibit strengthened ("trained") responses upon repeated challenge. Thus, the expression of c-Maf is indispensable for optimal type 2 cytokine production and proper memory-like responses in group-2 innate lymphoid cells., (© 2022 The Authors. Published under the terms of the CC BY NC ND 4.0 license.)
- Published
- 2022
- Full Text
- View/download PDF
34. Prognostic value of CD4+ T lymphopenia in non-small cell lung Cancer.
- Author
-
Eberst G, Vernerey D, Laheurte C, Meurisse A, Kaulek V, Cuche L, Jacoulet P, Almotlak H, Lahourcade J, Gainet-Brun M, Fabre E, Le Pimpec-Barthes F, Adotevi O, and Westeel V
- Subjects
- CD4-Positive T-Lymphocytes pathology, Humans, Prognosis, Prospective Studies, Carcinoma, Non-Small-Cell Lung pathology, Lung Neoplasms, Lymphopenia
- Abstract
Background: There is a paucity of data regarding the prognostic influence of peripheral blood CD4+ T lymphopenia in non-small cell lung cancer (NSCLC). Therefore, we investigated the prognostic value of T lymphopenia in NSCLC., Materials: Treatment-naive patients with a pathological diagnosis of NSCLC, at clinical stage I to IV were included in the prospective TELOCAP1 study. Lymphocytes count was evaluated in peripheral blood by flow cytometry. CD4+ and CD8+ T lymphopenia were defined as an absolute count of < 500/μL and < 224/μL respectively. The prognostic value of T lymphopenia was analyzed in the whole population, in local/loco-regional (stage I-IIIB) and in advanced (stage IV) NSCLC disease, using the Kaplan-Meier method and Cox regression models for survival curves and multivariate analysis, respectively., Results: Between July 2010 and January 2014, 169 evaluable patients with clinical stage I to IV NSCLC were prospectively enrolled. The prevalence of CD4+ and CD8+ T lymphopenia was similar in the study population (around 29%). Patients with CD4+ T lymphopenia showed lower overall survival than those with CD4+ T lymphocytes count > 500/μL (median overall survival (OS) 16.1 versus 21.7 months, hazard ratio (HR): 1.616 [95% CI: 1.1-2.36], p = 0.012). This association with OS was especially marked in local/loco-regional NSCLC stages (median OS, 21.8 versus 72 months, respectively, HR: 1.88 [95% CI: 0.9-3.8], p = 0.035). Multivariate analysis confirmed the worse prognosis associated with CD4+ T lymphopenia in local/loco-regional NSCLC, but not in metastatic patients (HR 2.028 [95% CI = 1.065-3.817] p = 0.02). Restricted cubic spline analysis showed that patients with CD4+ T lymphocytes count ≤500/μL displayed a high risk of death regardless of NSCLC clinical stage. There was no obvious relationship between CD8+ T lymphopenia and clinical outcome., Conclusion: We identified CD4+ T lymphopenia as an independent prognostic factor in local/loco-regional stages of NSCLC and CD4+ T lymphopenia is also associated with a high risk of death, regardless of NSCLC clinical stage., Trial Registration: EUDRACT: 2009-A00642-55., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
35. Human Monocyte-Derived Suppressor Cell Supernatant Induces Immunoregulatory Effects and Mitigates xenoGvHD.
- Author
-
Gérard C, Thébault M, Lamarthée B, Genet C, Cattin F, Brazdova A, Janikashvili N, Cladière C, Ciudad M, Ouandji S, Ghesquière T, Greigert H, Tinel C, Adotevi O, Saas P, Samson M, Audia S, and Bonnotte B
- Subjects
- Animals, CD8-Positive T-Lymphocytes, Humans, Membrane Glycoproteins metabolism, Mice, Mice, Inbred NOD, Mice, SCID, Proteomics, Graft vs Host Disease metabolism, Graft vs Host Disease prevention & control, Monocytes metabolism
- Abstract
Recently developed cell-based therapies have shown potential for graft-versus-host disease (GvHD) mitigation. Our team previously developed a protocol to generate human monocyte-derived suppressor Cells (HuMoSC), a subpopulation of CD33+ suppressor cells of monocytic origin. CD33+HuMoSC successfully reduced xenoGvHD severity in NOD/SCID/IL-2Rγc
-/- (NSG) mice. While CD33+ HuMoSC culture supernatant inhibits T cell activation and proliferation, the recovery of CD33+ HuMoSC immunosuppressive cells and the subsequent production of their supernatant is limited. An attractive solution would be to use both the CD33+ and the large number of CD14+ cells derived from our protocol. Here, we assessed the immunoregulatory properties of the CD14+HuMoSC supernatant and demonstrated that it inhibited both CD4 and CD8 T cell proliferation and decreased CD8 cytotoxicity. In vivo , injection of CD14+HuMoSC supernatant reduced xenoGvHD in NSG mice. Furthermore, CD14+HuMoSC supernatant maintained its immunoregulatory properties in an inflammatory environment. Proteomic and multiplex analyses revealed the presence of immunosuppressive proteins such as GPNMB, galectin-3 and IL-1R(A) Finally, CD14+HuMoSC supernatant can be produced using good manufacturing practices and be used as complement to current immunosuppressive drugs. CD14+HuMoSC supernatant is thus a promising therapy for preventing GvHD. ., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Gérard, Thébault, Lamarthée, Genet, Cattin, Brazdova, Janikashvili, Cladière, Ciudad, Ouandji, Ghesquière, Greigert, Tinel, Adotevi, Saas, Samson, Audia and Bonnotte.)- Published
- 2022
- Full Text
- View/download PDF
36. Naturally Occurring Telomerase-Specific CD4 T-Cell Immunity in Melanoma.
- Author
-
Nardin C, Laheurte C, Puzenat E, Boullerot L, Ramseyer M, Marguier A, Jacquin M, Godet Y, Aubin F, and Adotevi O
- Subjects
- Adult, Aged, Drug Resistance, Neoplasm immunology, Female, Follow-Up Studies, Humans, Immune Checkpoint Inhibitors pharmacology, Male, Melanoma blood, Melanoma drug therapy, Melanoma mortality, Middle Aged, Neoplasm Staging, Progression-Free Survival, Prospective Studies, Skin Neoplasms blood, Skin Neoplasms drug therapy, Skin Neoplasms mortality, Immune Checkpoint Inhibitors therapeutic use, Melanoma immunology, Skin Neoplasms immunology, Telomerase immunology, Th1 Cells immunology
- Abstract
CD4 T cells play a key role in anticancer immunity. In this study, we investigate the clinical relevance of circulating CD4 T helper type 1 (Th1) response against telomerase (anti-TERT Th1 response) in patients with melanoma. The spontaneous anti-TERT Th1 response was detected in 54.5% (85/156) of patients with melanoma before treatment. The prevalence of this systemic response was inversely related to Breslow thickness >1 mm and American Joint Committee on Cancer stage ≥II (P = 0.001 and 0.032, respectively). In contrast to patients treated with targeted therapies, the anti-TERT Th1 immunity was associated with an objective response after immune checkpoint inhibitors treatment. Hence, 86% (18/21) of responder patients exhibited pre-existing anti-TERT Th1 versus 35% (6/19) in nonresponders (P = 0.001). This response was also associated with increased progression-free survival and overall survival in patients with melanoma treated with immune checkpoint inhibitors (P = 0.0008 and 0.012, respectively). Collectively, the presence of circulating anti-TERT Th1 response is inversely related to melanoma evolution and appears to be a predictive factor of response to immunotherapy. Our results highlight the interest in telomerase-specific CD4 Th1 response as a promising blood-based biomarker of immune checkpoint inhibitors therapy in melanoma., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
37. Plasmacytoid dendritic cells proliferation associated with acute myeloid leukemia: phenotype profile and mutation landscape
- Author
-
Zalmaï L, Viailly PJ, Biichle S, Cheok M, Soret L, Angelot-Delettre F, Petrella T, Collonge-Rame MA, Seilles E, Geffroy S, Deconinck E, Daguindau E, Bouyer S, Dindinaud E, Baunin V, Le Garff-Tavernier M, Roos-Weil D, Wagner-Ballon O, Salaun V, Feuillard J, Brun S, Drenou B, Mayeur-Rousse C, Okamba P, Dorvaux V, Tichionni M, Rose J, Rubio MT, Jacob MC, Raggueneau V, Preudhomme C, Saas P, Ferrand C, Adotevi O, Roumier C, Jardin F, Garnache-Ottou F, and Renosi F
- Subjects
- Cell Proliferation, Humans, Mutation, Phenotype, Dendritic Cells, Leukemia, Myeloid, Acute genetics
- Abstract
Neoplasms involving plasmacytoid Dendritic Cells (pDCs) include Blastic pDC Neoplasms (BPDCN) and other pDC proliferations, where pDCs are associated with myeloid malignancies: most frequently Chronic MyeloMonocytic Leukemia (CMML) but also Acute Myeloid Leukemia (AML), hereafter named pDC-AML. We aimed to determine the reactive or neoplastic origin of pDCs in pDC-AML, and their link with the CD34+ blasts, monocytes or conventional DCs (cDCs) associated in the same sample, by phenotypic and molecular analyses (targeted NGS, 70 genes). We compared 15 pDC-AML at diagnosis with 21 BPDCN and 11 normal pDCs from healthy donors. CD45low CD34+ blasts were found in all cases (10-80% of medullar cells), associated with pDCs (4-36%), monocytes in 14 cases (1-10%) and cDCs (2 cases, 4.8-19%). pDCs in pDC-AML harbor a clearly different phenotype from BPDCN: CD4+ CD56- in 100% of cases, most frequently CD303+, CD304+ and CD34+; lower expression of cTCL1 and CD123 with isolated lymphoid markers (CD22/CD7/CD5) in some cases, suggesting a pre-pDC stage. In all cases, pDCs, monocytes and cDC are neoplastic since they harbor the same mutations as CD34+ blasts. RUNX1 is the most commonly mutated gene: detected in all AML with minimal differentiation (M0-AML) but not in the other cases. Despite low number of cases, the systematic association between M0-AML, RUNX1 mutations and an excess of pDC is puzzling. Further evaluation in a larger cohort is required to confirm RUNX1 mutations in pDC-AML with minimal differentiation and to investigate whether it represents a proliferation of blasts with macrophage and DC progenitor potential.
- Published
- 2021
- Full Text
- View/download PDF
38. Feasibility of health-related quality of life (HRQoL) assessment for cancer patients using electronic patient-reported outcome (ePRO) in daily clinical practice.
- Author
-
Mouillet G, Falcoz A, Fritzsch J, Almotlak H, Jacoulet P, Pivot X, Villanueva C, Mansi L, Kim S, Curtit E, Meneveau N, Adotevi O, Jary M, Eberst G, Vienot A, Calcagno F, Pozet A, Djoumakh O, Borg C, Westeel V, Anota A, and Paget-Bailly S
- Subjects
- Electronics, Feasibility Studies, Female, Humans, Middle Aged, Patient Reported Outcome Measures, Prospective Studies, Surveys and Questionnaires, Breast Neoplasms, Quality of Life psychology
- Abstract
Introduction: Routine Electronic Monitoring of Health-Related Quality of Life (HRQoL) (REMOQOL) in clinical care with real-time feedback to physicians could help to enhance patient-centered care. We evaluated the feasibility of REMOQOL in the French context in the QOLIBRY study. The primary objective was to assess the patients' compliance with REMOQOL., Methods: The QOLIBRY study was a single-center, prospective study conducted in the University Hospital of Besançon (France). Eligible patients were those treated with systemic therapies for breast, lung or colorectal cancer at any stage. Patients were invited to complete the EORTC QLQ-C30 questionnaire and cancer-site-specific modules before each visit on tablets and/or computers in the hospital or at home. During the consultation, physicians had real-time access to visual summaries of HRQoL scores. Compliance was assessed as adequate if at least 66% of HRQoL assessments were completed during the 4 months of follow-up., Results: Between March 2016 and October 2018, 177 patients were included in the QOLIBRY study. Median age was 64 years (IQR 54-71). The proportion of patients with an adequate compliance rate was 95.5% (n = 63) in the breast cancer cohort, 98.2% (n = 55) in the colorectal cancer cohort, and 90.9% (n = 50) in the lung cancer cohort. The physicians checked the HRQoL results in 73.1% of visits and prescribed supportive care and adapted patient management in 8.3% and 5.2% of visits, respectively., Conclusion & Perspectives: The results of QOLIBRY study suggest that REMOQOL is feasible in the French context. However, information about HRQoL monitoring, training of the physicians in the use of the software, and recommendations for using HRQoL results to guide care are essential and must be improved., (© 2021. Springer Nature Switzerland AG.)
- Published
- 2021
- Full Text
- View/download PDF
39. Anti-PD-1/Anti-PD-L1 Drugs and Radiation Therapy: Combinations and Optimization Strategies.
- Author
-
Boustani J, Lecoester B, Baude J, Latour C, Adotevi O, Mirjolet C, and Truc G
- Abstract
Immune checkpoint inhibitors have been associated with long-term complete responses leading to improved overall survival in several cancer types. However, these novel immunotherapies are only effective in a small proportion of patients, and therapeutic resistance represents a major limitation in clinical practice. As with chemotherapy, there is substantial evidence that radiation therapy promotes anti-tumor immune responses that can enhance systemic responses to immune checkpoint inhibitors. In this review, we discuss the main preclinical and clinical evidence on strategies that can lead to an enhanced response to PD-1/PD-L1 blockade in combination with radiation therapy. We focused on central issues in optimizing radiation therapy, such as the optimal dose and fractionation for improving the therapeutic ratio, as well as the impact on immune and clinical responses of dose rate, target volume, lymph nodes irradiation, and type of radiation particle. We explored the addition of a third immunomodulatory agent to the combination such as other checkpoint inhibitors, chemotherapy, and treatment targeting the tumor microenvironment components. The strategies described in this review provide a lead for future clinical trials.
- Published
- 2021
- Full Text
- View/download PDF
40. Weak immunogenicity of SARS-CoV-2 vaccine in patients with hematologic malignancies.
- Author
-
Malard F, Gaugler B, Gozlan J, Bouquet L, Fofana D, Siblany L, Eshagh D, Adotevi O, Laheurte C, Ricard L, Dulery R, Stocker N, van de Wyngaert Z, Genthon A, Banet A, Memoli M, Ikhlef S, Sestilli S, Vekhof A, Brissot E, Marjanovic Z, Chantran Y, Cuervo N, Ballot E, Morand-Joubert L, and Mohty M
- Subjects
- Adaptive Immunity, Adult, Aged, Aged, 80 and over, Antibodies, Neutralizing immunology, Antibodies, Viral immunology, BNT162 Vaccine, COVID-19 Vaccines administration & dosage, Comorbidity, Female, Host-Pathogen Interactions immunology, Humans, Immunoglobulin G immunology, Male, Middle Aged, Risk Factors, T-Lymphocytes immunology, T-Lymphocytes metabolism, Young Adult, COVID-19 complications, COVID-19 immunology, COVID-19 Vaccines immunology, Hematologic Neoplasms complications, Hematologic Neoplasms immunology, Immunogenicity, Vaccine, SARS-CoV-2 immunology
- Abstract
This study evaluated the safety and immunogenicity of BNT162b2 vaccine in patients with hematological malignancies. Antibodies blocking spike binding to immobilized ACE-2 (NAb) correlated with anti-Spike (S) IgG d42 titers (Spearman r = 0.865, p < 0.0001), and an anti-S IgG d42 level ≥3100 UA/mL was predictive of NAb ≥ 30%, the positivity cutoff for NAb (p < 0.0001). Only 47% of the patients achieved an anti-S IgG d42 level ≥3100 UA/mL after the two BNT162b2 inocula, compared to 87% of healthy controls. In multivariable analysis, male patients, use of B-cell targeting treatment within the last 12 months prior to vaccination, and CD19
+ B-cell level <120/uL, were associated with a significantly decreased probability of achieving a protective anti-S IgG level after the second BNT162b2 inoculum. Finally, using the IFN-γ ELISPOT assay, we found a significant increase in T-cell response against the S protein, with 53% of patients having an anti-S IgG-positive ELISPOT after the second BNT162b2 inoculum. There was a correlation between the anti-S ELISPOT response and IgG d42 level (Spearman r = 0.3026, p = 0.012). These findings suggest that vaccination with two BNT162b2 inocula translates into a significant increase in humoral and cellular response in patients with hematological malignancies, but only around half of the patients can likely achieve effective immune protection against COVID-19., (© 2021. The Author(s).)- Published
- 2021
- Full Text
- View/download PDF
41. Inflammatory and immunological profile in COPD secondary to organic dust exposure.
- Author
-
Keddache S, Laheurte C, Boullerot L, Laurent L, Dalphin JC, Adotevi O, and Soumagne T
- Subjects
- Aged, Agriculture, B-Lymphocytes immunology, B-Lymphocytes pathology, Case-Control Studies, Cytokines biosynthesis, Female, Humans, Inflammation etiology, Inflammation immunology, Inflammation pathology, Killer Cells, Natural immunology, Killer Cells, Natural pathology, Leukocytes, Mononuclear immunology, Leukocytes, Mononuclear pathology, Male, Middle Aged, Occupational Exposure adverse effects, Pulmonary Disease, Chronic Obstructive etiology, Pulmonary Disease, Chronic Obstructive pathology, T-Lymphocytes immunology, T-Lymphocytes pathology, Tobacco Smoking adverse effects, Dust immunology, Organic Chemicals toxicity, Pulmonary Disease, Chronic Obstructive immunology
- Abstract
Inflammatory response in patients with COPD secondary to organic dust exposure (OD-COPD) is poorly understood. We therefore aimed to characterize inflammatory and immune profile from peripheral blood mononuclear cells (PBMC) in a group of patients with mild-to-moderate COPD secondary to organic dust exposure (OD-COPD), tobacco smoking (T-COPD), or both. We compared T, B and NK cells distribution and inflammatory (TNF-α, Il-1β, IL-6), type 1 (IFN-γ), type 2 (IL-4, IL-13) and type 3 (IL-17) immunity related cytokines at baseline, and after stimulation with LPS, flagellin and CD3/CD28 beads in all COPD groups. OD-COPD displayed significantly lower NK cells and CD8+ T cells compared with controls. After flagellin stimulation, T-COPD had significantly lower IL-13 levels than OD-COPD and controls (p < 0.05) whereas IFN-γ tended to be lower in OD-COPD. All COPD groups displayed higher IL-1β and IL-17 than controls after CD3/CD28 stimulation. Inflammatory responses in OD-COPD were different from T-COPD. OD-COPD displayed higher levels of type 2 immunity related cytokines., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
42. Chemoradiation triggers antitumor Th1 and tissue resident memory-polarized immune responses to improve immune checkpoint inhibitors therapy.
- Author
-
Lauret Marie Joseph E, Kirilovsky A, Lecoester B, El Sissy C, Boullerot L, Rangan L, Marguier A, Tochet F, Dosset M, Boustani J, Ravel P, Boidot R, Spehner L, Haicheur-Adjouri N, Marliot F, Pallandre JR, Bonnefoy F, Scripcariu V, Van den Eynde M, Cornillot E, Mirjolet C, Pages F, and Adotevi O
- Subjects
- Animals, Disease Models, Animal, Female, Humans, Immune Checkpoint Inhibitors pharmacology, Mice, Tumor Microenvironment, Chemoradiotherapy methods, Immune Checkpoint Inhibitors therapeutic use, Immunity immunology, Immunotherapy methods, Th1 Cells radiation effects
- Abstract
Background: Multiple synergistic combination approaches with cancer drugs are developed to overcome primary resistance to immunotherapy; however, the mechanistic rationale to combine chemoradiotherapy (CRT) with immune checkpoint inhibitors remains elusive., Methods: This study described the immunological landscape of tumor microenvironment (TME) exposed to CRT. Tumor samples from patients with rectal cancer (n=43) treated with neoadjuvant CRT or radiotherapy were analyzed by nanostring and immunohistochemistry. Studies in mice were performed using three syngeneic tumors (TC1, CT26 and MC38). Tumor-bearing mice were treated either with platinum-based CRT, radiotherapy or chemotherapy. Anti-CTLA-4 and/or anti-Programmed Cell Death Receptor-1 (PD-1) therapy was used in combination with CRT. The therapy-exposed TME was screened by RNA sequencing and flow cytometry and tumor-infiltrating T lymphocyte functionality was evaluated by interferon (IFN)-γ ELIspot and intracellular cytokine staining., Results: Front-to-front comparison analysis revealed the synergistic effect of CRT to establish a highly inflamed and Th1-polarized immune signature in the TME of patients and mice. In both settings, CRT-exposed TMEs were highly enriched in newly-infiltrated tumor-specific CD8
+ T cells as well as tissue resident memory CD103+ CD8+ T cells. In mice, CD8 T cells were involved in the antitumor response mediated by CRT and were primed by CRT-activated CD103+ dendritic cells. In the three tumor models, we showed that concurrent combination of CRT with a dual CTLA-4 and PD-1 blockade was required to achieve an optimal antitumor effect and to establish a broad and long-lasting protective antitumor T cell immunity., Conclusions: Our results highlight the ability of CRT to stimulate strong antitumor T-cell-mediated immunity and tissue resident memory T activation in TME, to foster immune checkpoint inhibitors action. These findings have implications in clinic for the design clinical trials combining chemoradiation with immunotherapy., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)- Published
- 2021
- Full Text
- View/download PDF
43. Polyploid giant cancer cells, stemness and epithelial-mesenchymal plasticity elicited by human cytomegalovirus.
- Author
-
Nehme Z, Pasquereau S, Haidar Ahmad S, Coaquette A, Molimard C, Monnien F, Algros MP, Adotevi O, Diab Assaf M, Feugeas JP, and Herbein G
- Subjects
- Humans, Female, Epithelial Cells virology, Epithelial Cells pathology, Epithelial Cells metabolism, Neoplastic Stem Cells pathology, Neoplastic Stem Cells virology, Neoplastic Stem Cells metabolism, Cytomegalovirus Infections virology, Cytomegalovirus Infections pathology, Cell Transformation, Neoplastic, Mammary Glands, Human virology, Mammary Glands, Human pathology, Cytomegalovirus physiology, Epithelial-Mesenchymal Transition, Polyploidy, Breast Neoplasms pathology, Breast Neoplasms virology, Giant Cells virology, Giant Cells pathology, Giant Cells metabolism
- Abstract
A growing body of evidence is recognizing human cytomegalovirus (HCMV) as a potential oncogenic virus. We hereby provide the first experimental in vitro evidence for HCMV as a reprogramming vector, through the induction of dedifferentiation of mature human mammary epithelial cells (HMECs), generation of a polyploid giant cancer cell (PGCC) phenotype characterized by sustained growth of blastomere-like cells, in concordance with the acquisition of embryonic stem cells characteristics and epithelial-mesenchymal plasticity. HCMV presence parallels the succession of the observed cellular and molecular events potentially ensuing the transformation process. Correlation between PGCCs detection and HCMV presence in breast cancer tissue further validates our hypothesis in vivo. Our study indicates that some clinical HCMV strains conserve the potential to transform HMECs and fit with a "blastomere-like" model of oncogenesis, which may be relevant in the pathophysiology of breast cancer and other adenocarcinoma, especially of poor prognosis.
- Published
- 2021
- Full Text
- View/download PDF
44. First immunotherapeutic CAR-T cells against the immune checkpoint protein HLA-G.
- Author
-
Anna F, Bole-Richard E, LeMaoult J, Escande M, Lecomte M, Certoux JM, Souque P, Garnache F, Adotevi O, Langlade-Demoyen P, Loustau M, and Caumartin J
- Subjects
- Animals, Antibodies, Monoclonal genetics, Antibodies, Monoclonal immunology, Antigens, CD metabolism, Cell Differentiation, Coculture Techniques, Cytotoxicity, Immunologic, HLA-G Antigens immunology, Humans, Immunologic Memory, K562 Cells, Leukemia, Erythroblastic, Acute genetics, Leukemia, Erythroblastic, Acute immunology, Leukemia, Erythroblastic, Acute metabolism, Leukocyte Immunoglobulin-like Receptor B1 metabolism, Memory T Cells immunology, Memory T Cells metabolism, Mice, Inbred NOD, Mice, SCID, Phenotype, Receptors, Chimeric Antigen metabolism, Time Factors, Tumor Microenvironment, Xenograft Model Antitumor Assays, Mice, Antibodies, Monoclonal metabolism, HLA-G Antigens metabolism, Immunotherapy, Adoptive, Leukemia, Erythroblastic, Acute therapy, Memory T Cells transplantation, Receptors, Chimeric Antigen genetics
- Abstract
Background: CAR-T cells immunotherapy is a breakthrough in the treatment of hematological malignancies such as acute lymphoblastic leukemia (ALL) and B-cell malignancies. However, CAR-T therapies face major hurdles such as the lack of tumor-specific antigen (TSA), and immunosuppressive tumor microenvironment sometimes caused by the tumorous expression of immune checkpoints (ICPs) such as HLA-G. Indeed, HLA-G is remarkable because it is both a potent ICP and a TSA. HLA-G tumor expression causes immune escape by impairing innate and adaptive immune responses and by inducing a suppressive microenvironment. Yet, to date, no immunotherapy targets it., Methods: We have developed two anti-HLA-G third-generation CARs based on new anti-HLA-G monoclonal antibodies., Results: Anti-HLA-G CAR-T cells were specific for immunosuppressive HLA-G isoforms. HLA-G-activated CAR-T cells polarized toward T helper 1, and became cytotoxic against HLA-G
+ tumor cells. In vivo, anti-HLA-G CAR-T cells were able to control and eliminate HLA-G+ tumor cells. The interaction of tumor-HLA-G with interleukin (IL)T2-expressing T cells is known to result in effector T cell functional inhibition, but anti-HLA-G CAR-T cells were insensitive to this inhibition and still exerted their function even when expressing ILT2. Lastly, we show that anti-HLA-G CAR-T cells differentiated into long-term memory effector cells, and seemed not to lose function even after repeated stimulation by HLA-G-expressing tumor cells., Conclusion: We report for the first time that HLA-G, which is both a TSA and an ICP, constitutes a valid target for CAR-T cell therapy to specifically target and eliminate both tumor cells and HLA-G+ suppressive cells., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)- Published
- 2021
- Full Text
- View/download PDF
45. PD-1 and TIGIT coexpression identifies a circulating CD8 T cell subset predictive of response to anti-PD-1 therapy.
- Author
-
Simon S, Voillet V, Vignard V, Wu Z, Dabrowski C, Jouand N, Beauvais T, Khammari A, Braudeau C, Josien R, Adotevi O, Laheurte C, Aubin F, Nardin C, Rulli S, Gottardo R, Ramchurren N, Cheever M, Fling SP, Church CD, Nghiem P, Dreno B, Riddell SR, and Labarriere N
- Subjects
- CD8-Positive T-Lymphocytes metabolism, Carcinoma, Merkel Cell blood, Carcinoma, Merkel Cell drug therapy, Humans, Melanoma blood, Melanoma drug therapy, Predictive Value of Tests, Programmed Cell Death 1 Receptor antagonists & inhibitors, Programmed Cell Death 1 Receptor blood, Programmed Cell Death 1 Receptor immunology, Receptors, CXCR5 immunology, Receptors, Immunologic blood, Receptors, Immunologic immunology, T-Lymphocyte Subsets immunology, CD8-Positive T-Lymphocytes immunology, Carcinoma, Merkel Cell immunology, Immune Checkpoint Inhibitors pharmacology, Melanoma immunology, Programmed Cell Death 1 Receptor biosynthesis, Receptors, Immunologic biosynthesis
- Abstract
Background: Clinical benefit from programmed cell death 1 receptor (PD-1) inhibitors relies on reinvigoration of endogenous antitumor immunity. Nonetheless, robust immunological markers, based on circulating immune cell subsets associated with therapeutic efficacy are yet to be validated., Methods: We isolated peripheral blood mononuclear cell from three independent cohorts of melanoma and Merkel cell carcinoma patients treated with PD-1 inhibitor, at baseline and longitudinally after therapy. Using multiparameter flow cytometry and cell sorting, we isolated four subsets of CD8
+ T cells, based on PD-1 and TIGIT expression profiles. We performed phenotypic characterization, T cell receptor sequencing, targeted transcriptomic analysis and antitumor reactivity assays to thoroughly characterize each of these subsets., Results: We documented that the frequency of circulating PD-1+ TIGIT+ (DPOS) CD8+ T-cells after 1 month of anti-PD-1 therapy was associated with clinical response and overall survival. This DPOS T-cell population was enriched in highly activated T-cells, tumor-specific and emerging T-cell clonotypes and T lymphocytes overexpressing CXCR5, a key marker of the CD8 cytotoxic follicular T cell population. Additionally, transcriptomic profiling defined a specific gene signature for this population as well as the overexpression of specific pathways associated with the therapeutic response., Conclusions: Our results provide a convincing rationale for monitoring this PD-1+ TIGIT+ circulating population as an early cellular-based marker of therapeutic response to anti-PD-1 therapy., Competing Interests: Competing interests: SRR has served as an advisor and has patents licensed to Juno Therapeutics, a Celgene/Bristol-Myers Squibb company; is a founder and employee of Lyell Immunopharma; and has served on advisory boards for Adaptive Biotechnologies and Nohla. PN serves as a paid consultant for EMD Serono. Bristol Myers Squibb has provided research support to PN’s institution. RG has received consulting income from Juno Therapeutics, Takeda, Infotech Soft, Celgene, has received research support from Janssen Pharmaceuticals and Juno Therapeutics, and declares ownership in Cellspace Biosciences. SR and ZW are employed by QIAGEN, however, the studies were conducted in the absence of any potential conflict of interest. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (© Author(s) (or their employer(s)) 2020. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)- Published
- 2020
- Full Text
- View/download PDF
46. Molecular description of ANGPT2 associated colorectal carcinoma.
- Author
-
Jary M, Hasanova R, Vienot A, Asgarov K, Loyon R, Tirole C, Bouard A, Orillard E, Klajer E, Kim S, Viot J, Colle E, Adotevi O, Bouché O, Lecomte T, Borg C, and Feugeas JP
- Subjects
- Angiopoietin-2 blood, Clinical Trials as Topic, Colorectal Neoplasms blood, Databases, Genetic, Drug Resistance, Neoplasm, Female, Gene Expression Profiling, Gene Expression Regulation, Neoplastic, Genetic Association Studies, Glycoproteins blood, Humans, Male, Neoplasm Staging, Prognosis, Prospective Studies, Survival Analysis, Syndecan-1 blood, Angiopoietin-2 genetics, Colorectal Neoplasms mortality, Colorectal Neoplasms pathology
- Abstract
Angiopoietin-2 (ANGPT2) is a prognostic factor in metastatic colorectal cancer (CRC). Nevertheless, it remains to be elucidated which molecular characteristics make up the ANGPT2-related poor-prognosis CRC subset. Public transcriptomic datasets were collected from Gene Expression Omnibus GEO and with the TCGAbiolinks R-package for the TCGA. After appropriate normalization, differential expression analysis was performed using Benjamini and Hochberg method for false discovery rate. Plasma from two prospective clinical trials were used to investigate the clinical impact of ANGPT2-related biomarkers. In the 935 samples included in four annotated platforms (GPL) and derived from localized CRC, ANGPT2
hi expression conferred a worst overall survival (HR = 1.20; p = 0.02). CRC stage, ANGPT2hi expression but not Consortium Molecular Subtype (CMS) predict overall survival in multivariate analysis. ANGPT2 expression was not correlated with a specific CMS nor to RAS, RAF, MSI, p53, CIN, CIMP genomic alterations. Gene expression analysis revealed that ANGPT2hi CRC subset is characterized by angiogenesis-related gene expression, presence of myeloid cells, stromal organization and resistance to chemotherapy. A prognostic model was proposed using seric levels of ANGPT2, STC1 and CD138 in 97 mCRC patients. Our results provide evidence that ANGPT2 is a prognostic factor in localized CRC and defined a specific CRC subset with potential clinical implementation., (© 2020 UICC.)- Published
- 2020
- Full Text
- View/download PDF
47. Anti-Telomerase CD4 + Th1 Immunity and Monocytic-Myeloid-Derived-Suppressor Cells Are Associated with Long-Term Efficacy Achieved by Docetaxel, Cisplatin, and 5-Fluorouracil (DCF) in Advanced Anal Squamous Cell Carcinoma: Translational Study of Epitopes-HPV01 and 02 Trials.
- Author
-
Spehner L, Kim S, Vienot A, François E, Buecher B, Adotevi O, Vernerey D, Abdeljaoued S, Meurisse A, and Borg C
- Subjects
- Adaptive Immunity, Adult, Aged, Aged, 80 and over, Anus Neoplasms pathology, Anus Neoplasms virology, CD4-Positive T-Lymphocytes, Carcinoma, Squamous Cell mortality, Carcinoma, Squamous Cell virology, Cisplatin therapeutic use, Docetaxel therapeutic use, Female, Fluorouracil therapeutic use, Humans, Male, Middle Aged, Papillomavirus Infections pathology, Prognosis, Prospective Studies, Th1 Cells immunology, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Anus Neoplasms drug therapy, Anus Neoplasms immunology, Carcinoma, Squamous Cell drug therapy, Carcinoma, Squamous Cell immunology, Papillomavirus Infections immunology, Telomerase immunology
- Abstract
Docetaxel, cisplatin and 5-fluorouracil (DCF) chemotherapy regimen is highly effective in advanced anal squamous cell carcinoma (SCCA), as demonstrated by the Epitopes-HPV02 study results. Here, we analyzed the impact of DCF regimen and the prognostic value of adaptive immune responses and immunosuppressive cells in SCCA patients included in two prospective studies (Epitopes-HPV01 and HPV02). The presence of T-cell responses against Human papillomavirus (HPV)16-E6/E7 and anti-telomerase (hTERT)-antigens was measured by IFNᵧ-ELISpot. Here, we showed that HPV-adaptive immune responses are increased in SCCA patients. SCCA patients also displayed enhanced circulating TH1 T-cells restricted by hTERT. Exposition to DCF increased hTERT immunity but not HPV or common viruses immune responses. Notably, the correlation of hTERT immune responses with SCCA patients' clinical outcomes highlights that hTERT is a relevant antigen in this HPV-related disease. The influence of peripheral immunosuppressive cells was investigated by flow cytometry. While both regulatory T-cells and monocytic-myeloid-derived suppressive cells (M-MDSC) accumulated in the peripheral blood of SCCA patients, only high levels of M-MDSC were negatively correlated with hTERT adaptive immune responses and predicted poor prognosis. Altogether, our results reveal that hTERT is a relevant antigen in HPV-driven SCCA disease and that M-MDSC levels influence TH1-adaptive immune responses and patients' survival.
- Published
- 2020
- Full Text
- View/download PDF
48. Radiotherapy Scheme Effect on PD-L1 Expression for Locally Advanced Rectal Cancer.
- Author
-
Boustani J, Derangère V, Bertaut A, Adotevi O, Morgand V, Charon-Barra C, Ghiringhelli F, and Mirjolet C
- Subjects
- Adult, Aged, Aged, 80 and over, Female, Humans, Male, Middle Aged, Neoadjuvant Therapy, Retrospective Studies, Adenocarcinoma metabolism, Adenocarcinoma radiotherapy, B7-H1 Antigen metabolism, Rectal Neoplasms metabolism, Rectal Neoplasms radiotherapy
- Abstract
In locally advanced rectal cancer, radiotherapy (RT) followed by surgery have improved locoregional control, but distant recurrences remain frequent. Although checkpoint inhibitors have demonstrated objective response in several cancers, the clinical benefit of PD-1/PD-L1 blockade remains uncertain in rectal cancer. We collected data from biopsies and surgical specimens in 74 patients. The main objective was to evaluate the impact of neoadjuvant RT and fractionation on PD-L1 expression. Secondary objectives were to study the relation between PD-L1 expression and tumor regression grade (TRG), progression-free survival (PFS), overall survival (OS), and CD8 TILs infiltration. Median rates of cells expressing PD-L1 pre- and post-RT were 0.15 (range, 0-17) and 0.5 (range, 0-27.5), respectively ( p = 0.0005). There was no effect of RT fractionation on PD-L1+ cell rates. We found no relation between CD8+ TILs infiltration and PD-L1 expression and no difference between high-PD-L1 or low-PD-L1 expression and TRG. High-to-high PD-L1 expression profile had none significant higher OS and PFS compared to all other groups ( p = 0.06). Median OS and PFS were higher in biopsies with >0.08 PD-L1+ cells. High-to-high PD-L1 profile and ypT0-2 were significantly associated with higher OS and PFS. This study did not show the differential induction of PD-L1 expression according to fractionation.
- Published
- 2020
- Full Text
- View/download PDF
49. Characterization of chronic obstructive pulmonary disease in dairy farmers.
- Author
-
Soumagne T, Degano B, Guillien A, Annesi-Maesano I, Andujar P, Hue S, Adotevi O, Jouneau S, Botebol M, Laplante JJ, Roche N, and Dalphin JC
- Subjects
- Dairying, Farmers, Humans, Prevalence, Quality of Life, Risk Factors, Spirometry, Occupational Exposure adverse effects, Pulmonary Disease, Chronic Obstructive epidemiology, Pulmonary Disease, Chronic Obstructive etiology
- Abstract
Background: Although farming is often considered a risk factor for COPD, data regarding the burden and characteristics of COPD in dairy farmers are sparse and conflicting., Objectives: To characterize COPD in dairy farmers., Methods: 4788 subjects entered two parallel COPD screening programs, one in agricultural workers and one in general practice from 2011 to 2015. Subjects with COPD were invited to participate in the characterization phase of the study. Those who accepted were included in two subgroups: dairy farmers with COPD (DF-COPD) (n = 101) and non-farmers with COPD (NF-COPD) (n = 85). Patients with COPD were frequency-matched with subjects with normal spirometry for age, sex and tobacco smoking (pack-years and status) (DF-controls n = 98, NF-controls n = 89). All subjects from these four groups underwent lung function and exercise testing, questionnaires and blood analysis., Results: The frequency of COPD in dairy farmers was 8.0% using the GOLD criterion and 6.2% using the lower limit of normal criterion and was similar in non-farming subjects (7.3% and 5.2%, respectively) although dairy farmers had lower tobacco consumption (screening phase). DF-COPD had better pulmonary function, exercise capacity and quality of life, fewer symptoms and comorbidities than NF-COPD, and higher levels of some Th2 biomarkers (MCP-2, periostin) (characterization phase). In farmers, COPD was not related to occupational exposure factors, supporting the role of host factors., Conclusion: COPD secondary to organic dust exposure (dairy farming) appears less severe and associated with fewer comorbidities than COPD secondary to tobacco smoking., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
50. A First-in-Human Phase I Study of INVAC-1, an Optimized Human Telomerase DNA Vaccine in Patients with Advanced Solid Tumors.
- Author
-
Teixeira L, Medioni J, Garibal J, Adotevi O, Doucet L, Durey MD, Ghrieb Z, Kiladjian JJ, Brizard M, Laheurte C, Wehbe M, Pliquet E, Escande M, Defrance R, Culine S, Oudard S, Wain-Hobson S, Doppler V, Huet T, and Langlade-Demoyen P
- Subjects
- DNA, Humans, Vaccination, Cancer Vaccines, Neoplasms, Telomerase, Vaccines, DNA
- 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 ., (©2019 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.