Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Immunol Immunother ; 73(7): 133, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38753169

ABSTRACT

BACKGROUND: Glioblastoma (GBM) is a primary brain tumor with a dismal prognosis, often resistant to immunotherapy and associated with immune suppression. This study aimed to assess the impact of steroids and Stupp-regimen treatment on peripheral blood immune parameters in GBM patients and their association with outcomes. METHODS: Using cytometry panels and bioplex assays, we analyzed the immune phenotype and serum cytokines of 54 GBM patients and 21 healthy volunteers. RESULTS: GBM patients exhibited decreased lymphoid cell numbers (CD4, CD8 T cells, NKT cells) with heightened immune checkpoint expression and increased myeloid cell numbers (especially neutrophils), along with elevated pro-inflammatory cytokine levels. Steroid use decreased T and NK cell numbers, while radio-chemotherapy led to decreased lymphoid cell numbers, increased myeloid cell numbers, and heightened immune checkpoint expression. Certain immune cell subsets were identified as potential outcome predictors. CONCLUSION: Overall, these findings shed light on the peripheral immune landscape in GBM, emphasizing the immunosuppressive effects of treatment. Baseline immune parameters may serve as prognostic indicators for treatment response.


Subject(s)
Brain Neoplasms , Chemoradiotherapy , Glioblastoma , Humans , Glioblastoma/immunology , Glioblastoma/therapy , Glioblastoma/drug therapy , Male , Female , Middle Aged , Brain Neoplasms/immunology , Brain Neoplasms/therapy , Brain Neoplasms/drug therapy , Chemoradiotherapy/methods , Adult , Aged , Prognosis , Cytokines/metabolism , Cytokines/blood
2.
STAR Protoc ; 4(4): 102658, 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-37910510

ABSTRACT

Here, we present a protocol for the simultaneous analysis of peripheral and intratumoral lymphocyte function by flow cytometry. Using tumor and blood samples from patients with colorectal cancer, we describe steps for tumor digestion, pre-labeling of the tumor-infiltrating leukocytes (TILs), and activation and labeling of the total cells (TILs and whole blood cells). For complete details on the use and execution of this protocol, please refer to Thibaudin et al.1.


Subject(s)
Leukocytes , Neoplasms , Humans , Flow Cytometry , Lymphocytes
3.
Nat Med ; 29(8): 2087-2098, 2023 08.
Article in English | MEDLINE | ID: mdl-37563240

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 .


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Colorectal Neoplasms , Humans , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antibodies, Monoclonal/therapeutic use , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology
4.
Cancer Immunol Immunother ; 71(10): 2549-2563, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35292828

ABSTRACT

Microsatellite stable colorectal cancers (MSS-CRC) are resistant to anti-PD-1/PD-L1 therapy but the combination of immune checkpoints inhibitors (ICI) could be a clue to reverse resistance. Our aim was to evaluate ex vivo the capacity of the combination of atezolizumab (anti-PD-L1) and tiragolumab (anti-TIGIT) to reactivate the immune response of tumor infiltrating lymphocytes (TILs) in MSS-CRC. We analysed CRC tumor tissue and the associated blood sample in parallel. For each patient sample, extensive immunomonitoring and cytokine production were tested. We generated an ex vivo assay to study immune reactivity following immune stimulation with checkpoint inhibitors of tumor cell suspensions. Three microsatellite instable (MSI) and 13 MSS-CRC tumors were analysed. To generalize our observations, bioinformatics analyses were performed on public data of single cell RNA sequencing of CRC TILs and RNA sequencing data of TCGA. Atezolizumab alone could only reactivate T cells from MSI tumors. Atezolizumab and tiragolumab reactivated T cells in 46% of MSS-CRC samples. Reactivation by ICK was observed in patients with higher baseline frequency of Th1 and Tc1 cells, and was also associated with higher baseline T cell polyfunctionality and higher CD96 expression. We showed that a high frequency of CD96 expression on T cells could be a surrogate marker of atezolizumab and tiragolumab efficacy. Together these data suggest that the association of atezolizumab and tiragolumab could restore function of CD4 and CD8 TILs in MSS-CRC and could be tested in a clinical trial in colorectal cancer patients with MSS status.


Subject(s)
Colorectal Neoplasms , Microsatellite Instability , Antibodies, Monoclonal/genetics , Antigens, CD/metabolism , CD8-Positive T-Lymphocytes , Humans , Lymphocytes, Tumor-Infiltrating , Receptors, Immunologic/metabolism
5.
J Immunother Cancer ; 9(6)2021 06.
Article in English | MEDLINE | ID: mdl-34103351

ABSTRACT

BACKGROUND: T follicular helper cells (Tfh) are essential to shape B cell response during germinal center formation. Tfh accumulation has been reported in various human cancers, with positive or negative prognostic roles. However, the mechanisms explaining the accumulation of Tfh and their role in cancer remain obscure. METHODS: In vitro differentiated and mouse cell sorted Tfh phenotype was evaluated by flow cytometry and quantitative PCR (qPCR). Antitumor effect of Tfh was evaluated by adoptive transfer in different tumor-bearing mice models. The involvement of immune cells, cytokines and chemokines was evaluated, using depleting antibodies. Chemokines and cytokines expression and production were evaluated by qPCR and ELISA. In human, the impact of immune cells and chemokines on survival was evaluated by analyzing transcriptomic data from public databases and from our own patient cohorts. RESULTS: In this study, we show that Tfh exert an antitumor immune effect in a CD8+-dependent manner. Tfh produce interleukin-21, which sustains proliferation, viability, cytokine production and cytotoxic functions of exhausted T cells. The presence of Tfh is required for efficacy of antiprogrammed cell death ligand-1 therapy. Tfh accumulate in the tumor bed and draining lymph nodes in different mouse cancer models. This recruitment is due to the capacity of transforming growth factor ß to drive Chemokine (C-X-C motif) Ligand 13 expression, a chemoattractant of Tfh, by intratumor CD8+ T cells. Accumulation of Tfh and exhausted CD8+ T cells predicts cancer outcome in various cancer types. In patients treated with anti-programmed cell death-1 mAb, accumulation of Tfh and CD8+ at the tumor site is associated with outcome. CONCLUSION: This study provides evidence that CD8+/Tfh crosstalk is important in shaping antitumor immune response generated by immunotherapy.


Subject(s)
Brain Neoplasms/therapy , Breast Neoplasms/therapy , Glioblastoma/therapy , Immune Checkpoint Inhibitors/administration & dosage , T Follicular Helper Cells/transplantation , Adoptive Transfer , Animals , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Agents, Immunological/pharmacology , Brain Neoplasms/immunology , Breast Neoplasms/genetics , Breast Neoplasms/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Chemokine CXCL13/genetics , Chemokine CXCL13/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/genetics , Glioblastoma/immunology , Humans , Immune Checkpoint Inhibitors/pharmacology , Interleukins/genetics , Interleukins/metabolism , Mice , T Follicular Helper Cells/immunology , Treatment Outcome , Xenograft Model Antitumor Assays
6.
Eur J Cancer ; 139: 70-80, 2020 11.
Article in English | MEDLINE | ID: mdl-32977223

ABSTRACT

BACKGROUND: Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2, has caused a major pandemic. Patients with cancer are at higher risk of severe COVID-19. We aimed to describe and compare the immunological features of cancer patients hospitalised for COVID-19 or other concomitant, cancer-related illness. METHODS: In this prospective study, the clinical and immunological characteristics of 11 cancer patients with COVID-19 and 11 non-COVID-19 cancer patients hospitalised in the same unit at the same period for other medical issues were analysed. We also used 10 healthy volunteers as controls. Peripheral immune parameters were analysed using multiparametric flow cytometry. RESULTS: The median age of COVID-19-positive cancer patients was 71.1 years, and 66.4 years for controls. Compared with non-COVID-19 cancer patients, COVID-19-positive cancer patients had more extensive lymphopenia and hypoalbuminemia, with higher levels of C-reactive protein. In COVID-19 patients, elevated procalcitonin was associated with a higher risk of death. By phenotypic analysis, COVID-19-positive patients presented CD3 lymphopenia, with inversion of the CD4/CD8 ratio and modification of monocyte activation, with accumulation of mMDSC (monocytic Myeloid-Derived Suppressor Cells) -like cells and a decrease in activated monocytes. Analysis of the T-cell compartment revealed a T-dependent inflammatory response with accumulation of Th17 cells and cytotoxic CD8 T cells producing TNFα, a decrease in HLA-DR (Human Leukocyte Antigen - DR isotype)-positive CD8 T cells and Treg/CD8 ratio. CONCLUSION: SARS-CoV-2 infection in cancer patients is associated with CD4 T-cell lymphopenia with induction of an inflammatory T-cell response, accumulation of IFNγ+ TNFα+ CD8 T and Th17 cells, and a concomitant modification of monocyte activation status.


Subject(s)
Betacoronavirus/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Coronavirus Infections/immunology , Neoplasms/immunology , Pneumonia, Viral/immunology , T-Lymphocytes, Cytotoxic/immunology , Aged , Betacoronavirus/isolation & purification , COVID-19 , Coronavirus Infections/complications , Coronavirus Infections/transmission , Coronavirus Infections/virology , Female , France/epidemiology , Humans , Male , Neoplasms/epidemiology , Neoplasms/virology , Pandemics , Pneumonia, Viral/complications , Pneumonia, Viral/transmission , Pneumonia, Viral/virology , Prospective Studies , SARS-CoV-2 , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...