Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 97
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Immunity ; 55(3): 527-541.e5, 2022 03 08.
Article in English | MEDLINE | ID: mdl-35231421

ABSTRACT

The presence of intratumoral tertiary lymphoid structures (TLS) is associated with positive clinical outcomes and responses to immunotherapy in cancer. Here, we used spatial transcriptomics to examine the nature of B cell responses within TLS in renal cell carcinoma (RCC). B cells were enriched in TLS, and therein, we could identify all B cell maturation stages toward plasma cell (PC) formation. B cell repertoire analysis revealed clonal diversification, selection, expansion in TLS, and the presence of fully mature clonotypes at distance. In TLS+ tumors, IgG- and IgA-producing PCs disseminated into the tumor beds along fibroblastic tracks. TLS+ tumors exhibited high frequencies of IgG-producing PCs and IgG-stained and apoptotic malignant cells, suggestive of anti-tumor effector activity. Therapeutic responses and progression-free survival correlated with IgG-stained tumor cells in RCC patients treated with immune checkpoint inhibitors. Thus, intratumoral TLS sustains B cell maturation and antibody production that is associated with response to immunotherapy, potentially via direct anti-tumor effects.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Tertiary Lymphoid Structures , Carcinoma, Renal Cell/therapy , Female , Humans , Immunoglobulin G , Kidney Neoplasms/therapy , Male , Plasma Cells , Tertiary Lymphoid Structures/pathology , Tumor Microenvironment
2.
Semin Immunol ; 48: 101406, 2020 04.
Article in English | MEDLINE | ID: mdl-33248905

ABSTRACT

Tumors progression is under the control of a heterogeneous microenvironment composed of immune cells, fibroblasts, blood and lymphatic vessels, in which T cells have been demonstrated to be major actors, through their cytotoxic and cytokine producing effector functions and their long term memory that protects against metastasis. In this scenario, lessons from mouse models taught that B cells exert a protumoral role, via macrophage-dependent activation of inflammation. However, it became progressively evident from studies in patients with human cancers that the anti-tumor responses can be generated and controlled in tertiary lymphoid structures (TLS) that concentrate most of the intratumoral B cells and where B cells can differentiate into plasma cells and memory cells. Furthermore, recent studies demonstrated that the presence in tumors of B cells and TLS are associated with favorable outcome in patients treated by immunotherapy, unraveling TLS as a new predictive marker of anti-tumor response human cancers. This review encompasses the characteristics and functions of TLS and of B cells in human tumors, their prognostic and theranostic impact and summarizes the mouse models used to induce TLS neogenesis in tumors.


Subject(s)
B-Lymphocytes/immunology , Biomarkers, Tumor/immunology , Immunotherapy/methods , Neoplasms/therapy , T-Lymphocytes/immunology , Tertiary Lymphoid Structures/immunology , Animals , Biomarkers, Pharmacological , Humans , Immunomodulation , Neoplasms/immunology , Tumor Microenvironment
3.
Lancet Oncol ; 23(5): 612-624, 2022 05.
Article in English | MEDLINE | ID: mdl-35390339

ABSTRACT

BACKGROUND: We previously reported a 35-gene expression classifier identifying four clear-cell renal cell carcinoma groups (ccrcc1 to ccrcc4) with different tumour microenvironments and sensitivities to sunitinib in metastatic clear-cell renal cell carcinoma. Efficacy profiles might differ with nivolumab and nivolumab-ipilimumab. We therefore aimed to evaluate treatment efficacy and tolerability of nivolumab, nivolumab-ipilimumab, and VEGFR-tyrosine kinase inhibitors (VEGFR-TKIs) in patients according to tumour molecular groups. METHODS: This biomarker-driven, open-label, non-comparative, randomised, phase 2 trial included patients from 15 university hospitals or expert cancer centres in France. Eligible patients were aged 18 years or older, had an Eastern Cooperative Oncology Group performance status of 0-2, and had previously untreated metastatic clear-cell renal cell carcinoma. Patients were randomly assigned (1:1) using permuted blocks of varying sizes to receive either nivolumab or nivolumab-ipilimumab (ccrcc1 and ccrcc4 groups), or either a VEGFR-TKI or nivolumab-ipilimumab (ccrcc2 and ccrcc3 groups). Patients assigned to nivolumab-ipilimumab received intravenous nivolumab 3 mg/kg plus ipilimumab 1 mg/kg every 3 weeks for four doses followed by intravenous nivolumab 240 mg every 2 weeks. Patients assigned to nivolumab received intravenous nivolumab 240 mg every 2 weeks. Patients assigned to VEGFR-TKIs received oral sunitinib (50 mg/day for 4 weeks every 6 weeks) or oral pazopanib (800 mg daily continuously). The primary endpoint was the objective response rate by investigator assessment per Response Evaluation Criteria in Solid Tumors version 1.1. The primary endpoint and safety were assessed in the population who received at least one dose of study drug. This trial is registered with ClinicalTrials.gov, NCT02960906, and with the EU Clinical Trials Register, EudraCT 2016-003099-28, and is closed to enrolment. FINDINGS: Between June 28, 2017, and July 18, 2019, 303 patients were screened for eligibility, 202 of whom were randomly assigned to treatment (61 to nivolumab, 101 to nivolumab-ipilimumab, 40 to a VEGFR-TKI). In the nivolumab group, two patients were excluded due to a serious adverse event before the first study dose and one patient was excluded from analyses due to incorrect diagnosis. Median follow-up was 18·0 months (IQR 17·6-18·4). In the ccrcc1 group, objective responses were seen in 12 (29%; 95% CI 16-45) of 42 patients with nivolumab and 16 (39%; 24-55) of 41 patients with nivolumab-ipilimumab (odds ratio [OR] 0·63 [95% CI 0·25-1·56]). In the ccrcc4 group, objective responses were seen in seven (44%; 95% CI 20-70) of 16 patients with nivolumab and nine (50% 26-74) of 18 patients with nivolumab-ipilimumab (OR 0·78 [95% CI 0·20-3·01]). In the ccrcc2 group, objective responses were seen in 18 (50%; 95% CI 33-67) of 36 patients with a VEGFR-TKI and 19 (51%; 34-68) of 37 patients with nivolumab-ipilimumab (OR 0·95 [95% CI 0·38-2·37]). In the ccrcc3 group, no objective responses were seen in the four patients who received a VEGFR-TKI, and in one (20%; 95% CI 1-72) of five patients who received nivolumab-ipilimumab. The most common treatment-related grade 3-4 adverse events were hepatic failure and lipase increase (two [3%] of 58 for both) with nivolumab, lipase increase and hepatobiliary disorders (six [6%] of 101 for both) with nivolumab-ipilimumab, and hypertension (six [15%] of 40) with a VEGFR-TKI. Serious treatment-related adverse events occurred in two (3%) patients in the nivolumab group, 38 (38%) in the nivolumab-ipilimumab group, and ten (25%) patients in the VEGFR-TKI group. Three deaths were treatment-related: one due to fulminant hepatitis with nivolumab-ipilimumab, one death from heart failure with sunitinib, and one due to thrombotic microangiopathy with sunitinib. INTERPRETATION: We demonstrate the feasibility and positive effect of a prospective patient selection based on tumour molecular phenotype to choose the most efficacious treatment between nivolumab with or without ipilimumab and a VEGFR-TKI in the first-line treatment of metastatic clear-cell renal cell carcinoma. FUNDING: Bristol Myers Squibb, ARTIC.


Subject(s)
Carcinoma, Renal Cell , Nivolumab , Angiogenesis Inhibitors/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Biomarkers , Carcinoma, Renal Cell/drug therapy , Female , Humans , Ipilimumab , Lipase , Male , Neoplasm Staging , Nivolumab/adverse effects , Prospective Studies , Protein Kinase Inhibitors/adverse effects , Sunitinib , Tumor Microenvironment
4.
Immunity ; 39(4): 782-95, 2013 Oct 17.
Article in English | MEDLINE | ID: mdl-24138885

ABSTRACT

The complex interactions between tumors and their microenvironment remain to be elucidated. Combining large-scale approaches, we examined the spatio-temporal dynamics of 28 different immune cell types (immunome) infiltrating tumors. We found that the immune infiltrate composition changed at each tumor stage and that particular cells had a major impact on survival. Densities of T follicular helper (Tfh) cells and innate cells increased, whereas most T cell densities decreased along with tumor progression. The number of B cells, which are key players in the core immune network and are associated with prolonged survival, increased at a late stage and showed a dual effect on recurrence and tumor progression. The immune control relevance was demonstrated in three endoscopic orthotopic colon-cancer mouse models. Genomic instability of the chemokine CXCL13 was a mechanism associated with Tfh and B cell infiltration. CXCL13 and IL21 were pivotal factors for the Tfh/B cell axis correlating with survival. This integrative study reveals the immune landscape in human colorectal cancer and the major hallmarks of the microenvironment associated with tumor progression and recurrence.


Subject(s)
B-Lymphocytes/immunology , Carcinoma/immunology , Chemokine CXCL13/immunology , Colorectal Neoplasms/immunology , Interleukins/immunology , Neoplasm Recurrence, Local/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , B-Lymphocytes/pathology , Carcinoma/genetics , Carcinoma/mortality , Carcinoma/pathology , Cell Movement , Chemokine CXCL13/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Gene Expression Profiling , Gene Expression Regulation , Humans , Immunity, Innate , Interleukins/genetics , Lymphocyte Count , Mice , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/mortality , Neoplasm Recurrence, Local/pathology , Neoplasm Staging , Protein Stability , Survival Analysis , T-Lymphocytes, Helper-Inducer/pathology , Tumor Microenvironment/immunology
5.
Immunol Rev ; 271(1): 260-75, 2016 May.
Article in English | MEDLINE | ID: mdl-27088920

ABSTRACT

The characterization of the microenvironment of human tumors led to the description of tertiary lymphoid structures (TLS) characterized by mature dendritic cells in a T-cell zone adjacent to B-cell follicle including a germinal center. TLS represent sites of lymphoid neogenesis that develop in most solid cancers. Analysis of the current literature shows that the TLS presence is associated with a favorable clinical outcome for cancer patients, regardless of the approach used to quantify TLS and the stage of the disease. Using several approaches that combine immunohistochemistry, gene expression assays, and flow cytometry on large series of lung tumors, our work demonstrated that TLS are important sites for the initiation and/or maintenance of the local and systemic T- and B-cell responses against tumors. Surrounded by high endothelial venules, they represent a privileged area for the recruitment of lymphocytes into tumors and generation of central-memory T and B cells that circulate and limit cancer progression. TLS can be considered as a novel biomarker to stratify the overall survival risk of untreated cancer patients and as a marker of efficient immunotherapies. The induction and manipulation of cancer-associated TLS using drug agonists and/or biotherapies should open new avenues to treat cancer patients.


Subject(s)
B-Lymphocytes/immunology , Dendritic Cells/immunology , Immunotherapy, Adoptive , Lung Neoplasms/immunology , Lymphoid Tissue/immunology , T-Lymphocytes/immunology , Antigens, Neoplasm/immunology , Cell Differentiation , Cell Movement , Humans , Immunologic Memory , Immunologic Surveillance , Lung Neoplasms/therapy , Lymphocyte Activation , Tumor Microenvironment
6.
Br J Cancer ; 120(1): 45-53, 2019 01.
Article in English | MEDLINE | ID: mdl-30413828

ABSTRACT

The highly complex and heterogenous ecosystem of a tumour not only contains malignant cells, but also interacting cells from the host such as endothelial cells, stromal fibroblasts, and a variety of immune cells that control tumour growth and invasion. It is well established that anti-tumour immunity is a critical hurdle that must be overcome for tumours to initiate, grow and spread and that anti-tumour immunity can be modulated using current immunotherapies to achieve meaningful anti-tumour clinical responses. Pioneering studies in melanoma, ovarian and colorectal cancer have demonstrated that certain features of the tumour immune microenvironment (TME)-in particular, the degree of tumour infiltration by cytotoxic T cells-can predict a patient's clinical outcome. More recently, studies in renal cell cancer have highlighted the importance of assessing the phenotype of the infiltrating T cells to predict early relapse. Furthermore, intricate interactions with non-immune cellular players such as endothelial cells and fibroblasts modulate the clinical impact of immune cells in the TME. Here, we review the critical components of the TME in solid tumours and how they shape the immune cell contexture, and we summarise numerous studies evaluating its clinical significance from a prognostic and theranostic perspective.


Subject(s)
Neoplasms/immunology , T-Lymphocytes, Cytotoxic/immunology , Tumor Microenvironment/immunology , Cancer-Associated Fibroblasts/immunology , Cancer-Associated Fibroblasts/pathology , Cell Proliferation/genetics , Endothelial Cells/immunology , Endothelial Cells/pathology , Humans , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasms/pathology , T-Lymphocytes, Cytotoxic/pathology
7.
Br J Cancer ; 120(8): 815-818, 2019 04.
Article in English | MEDLINE | ID: mdl-30862951

ABSTRACT

In colorectal cancer (CRC), T-cell checkpoint blockade is only effective in patients diagnosed with mismatch repair-deficient (MMR-d) cancers. However, defects in Human Leukocyte Antigen (HLA) class I expression were reported to occur in most MMR-d CRCs, which would preclude antigen presentation in these tumours, considered essential for the clinical activity of this immunotherapeutic modality. We revisited this paradox by characterising HLA class I expression in two independent cohorts of CRC. We determined that loss of HLA class I expression occurred in the majority (73-78%) of MMR-d cases. This phenotype was rare in CRC liver metastases, irrespective of MMR status, whereas weak, inducible expression of HLA class I molecules was frequent in liver lesions. We propose that HLA class I is an important determinant of metastatic homing in CRCs. This observation is paramount to understand CRC carcinogenesis and for the application of immunotherapies in the metastatic setting.


Subject(s)
Colorectal Neoplasms/therapy , Genes, MHC Class I/genetics , Immunotherapy , T-Lymphocytes/immunology , Antigen Presentation/immunology , Brain Neoplasms/genetics , Brain Neoplasms/immunology , Carcinogenesis/genetics , Carcinogenesis/immunology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Female , Gene Expression Regulation, Neoplastic/immunology , Genes, cdc/drug effects , Genes, cdc/immunology , Humans , Liver/metabolism , Liver/pathology , Male , Middle Aged , Neoplasm Metastasis , Neoplastic Syndromes, Hereditary/genetics , Neoplastic Syndromes, Hereditary/immunology
8.
J Hepatol ; 70(1): 58-65, 2019 01.
Article in English | MEDLINE | ID: mdl-30213589

ABSTRACT

BACKGROUND & AIMS: Tertiary lymphoid structures (TLSs) provide a local and critical microenvironment for generating anti-tumor cellular and humoral immune responses. TLSs are associated with improved clinical outcomes in most solid tumors investigated to date. However, their role in hepatocellular carcinoma (HCC) is debated, as they have recently been shown to promote the growth of malignant hepatocyte progenitors in the non-tumoral liver. METHODS: We aimed to determine, by pathological review, the prognostic significance of both intra-tumoral and non-tumoral TLSs in a series of 273 patients with HCC treated by surgical resection in Henri Mondor University Hospital. Findings were further validated by gene expression profiling using a public data set (LCI cohort). RESULTS: TLSs were identified in 47% of the tumors, by pathological review, with lymphoid aggregates, primary and secondary follicles in 26%, 16% and 5% of the cases, respectively. Univariate and multivariate analyses showed that intra-tumoral TLSs significantly correlated with a lower risk of early relapse (<2 years after surgery, hazard ratio 0.46, p = 0.005). Interestingly, the risk of recurrence was also related to the degree of TLS maturation (primary or secondary follicles vs. lymphoid aggregates, p = 0.01). A gene expression signature associated with the presence of intra-tumoral TLS was also independently associated with a lower risk of early relapse in the LCI cohort. No association between the density of TLSs located in the adjacent non-tumoral liver and early or late recurrence was observed. CONCLUSIONS: We have shown that intra-tumoral TLSs are associated with a lower risk of early relapse in 2 independent cohorts of patients with HCC treated by surgical resection. Thus, intra-tumoral TLSs may reflect the existence of ongoing, effective anti-tumor immunity. LAY SUMMARY: Tertiary lymphoid structures provide a critical microenvironment for generating anti-tumor immune responses, and are associated with improved clinical outcome in most cancers investigated. Their role in hepatocellular carcinoma is however debated. We show in the present study that intra-tumoral tertiary lymphoid structures are associated with a low risk of early relapse after surgical resection, suggesting that they reflect the existence of in situ, effective anti-tumor immunity.


Subject(s)
Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Neoplasm Recurrence, Local/pathology , Tertiary Lymphoid Structures/pathology , Tumor Microenvironment/immunology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Biopsy , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/metabolism , Follow-Up Studies , Gene Expression Profiling , Humans , Liver Neoplasms/immunology , Liver Neoplasms/metabolism , Male , Middle Aged , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/metabolism , Prognosis , Retrospective Studies , Tertiary Lymphoid Structures/immunology , Tertiary Lymphoid Structures/metabolism , Time Factors
9.
Trends Immunol ; 37(1): 1-2, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26706045

ABSTRACT

The role of tertiary lymphoid structures (TLS) in cancer has recently been illustrated and revisited using mouse models. Joshi et al. describe regulatory T (Treg) cell infiltration and functional heterogeneity of TLS in lung tumors. Finkin et al. report that inflammation-associated TLS serve as niche for tumor progenitor cells, which may lead to recurrence in hepatocellular-carcinoma.


Subject(s)
B-Lymphocytes/immunology , Carcinoma, Hepatocellular/immunology , Immunologic Surveillance , Lung Neoplasms/immunology , Neoplastic Stem Cells/physiology , T-Lymphocytes, Regulatory/immunology , Tertiary Lymphoid Structures/immunology , Animals , Disease Models, Animal , Humans , Mice , Stem Cell Niche
12.
Trends Immunol ; 35(11): 571-80, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25443495

ABSTRACT

Tertiary lymphoid structures (TLS) are ectopic lymphoid formations found in inflamed, infected, or tumoral tissues. They exhibit all the characteristics of structures in the lymph nodes (LN) associated with the generation of an adaptive immune response, including a T cell zone with mature dendritic cells (DC), a germinal center with follicular dendritic cells (FDC) and proliferating B cells, and high endothelial venules (HEV). In this review, we discuss evidence for the roles of TLS in chronic infection, autoimmunity, and cancer, and address the question of whether TLS present beneficial or deleterious effects in these contexts. We examine the relationship between TLS in tumors and patient prognosis, and discuss the potential role of TLS in building and/or maintaining local immune responses and how this understanding may guide therapeutic interventions.


Subject(s)
Neoplasms/immunology , Tumor Microenvironment/immunology , Animals , Biomarkers , Humans , Immunity, Cellular , Infections/immunology , Infections/pathology , Inflammation/immunology , Inflammation/pathology , Neoplasms/genetics , Neoplasms/pathology , Prognosis , Tumor Microenvironment/genetics
13.
Am J Respir Crit Care Med ; 191(4): 377-90, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25369536

ABSTRACT

Solid tumors, beyond mere accumulation of cancer cells, form a complex ecosystem consisting of normal epithelial cells, fibroblasts, blood and lymphatic vessels, structural components, and infiltrating hematopoietic cells including myeloid and lymphoid elements that impact tumor growth, tumor spreading, and clinical outcome. The composition of the immune microenvironment is diverse, including various populations of T cells, B cells, dendritic cells, natural killer cells, myeloid-derived suppressor cells, neutrophils, or macrophages. The immune contexture describes the density, location, and organization of these immune cells within solid tumors. In lung cancer, which is the deadliest type of cancer, and particularly in non-small cell lung cancer, its most prevalent form, reports have described some of the interactions between the tumor and the host. These data, in addition to articles on various types of tumors, provide a greater understanding of the tumor-host microenvironment interaction and stimulate the development of prognostic and predictive biomarkers, the identification of novel target antigens for therapeutic intervention, and the implementation of tools for long-term management of patients with cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung/immunology , Lung Neoplasms/immunology , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/diagnosis , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Humans , Immunologic Factors/therapeutic use , Lung Neoplasms/diagnosis , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Prognosis , Tumor Microenvironment/immunology
14.
Am J Respir Crit Care Med ; 189(7): 832-44, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24484236

ABSTRACT

RATIONALE: It is now well established that immune responses can take place outside of primary and secondary lymphoid organs. We previously described the presence of tertiary lymphoid structures (TLS) in patients with non-small cell lung cancer (NSCLC) characterized by clusters of mature dendritic cells (DCs) and T cells surrounded by B-cell follicles. We demonstrated that the density of these mature DCs was associated with favorable clinical outcome. OBJECTIVES: To study the role of follicular B cells in TLS and the potential link with a local humoral immune response in patients with NSCLC. METHODS: The cellular composition of TLS was investigated by immunohistochemistry. Characterization of B-cell subsets was performed by flow cytometry. A retrospective study was conducted in two independent cohorts of patients. Antibody specificity was analyzed by ELISA. MEASUREMENTS AND MAIN RESULTS: Consistent with TLS organization, all stages of B-cell differentiation were detectable in most tumors. Germinal center somatic hypermutation and class switch recombination machineries were activated, associated with the generation of plasma cells. Approximately half of the patients showed antibody reactivity against up to 7 out of the 33 tumor antigens tested. A high density of follicular B cells correlated with long-term survival, both in patients with early-stage NSCLC and with advanced-stage NSCLC treated with chemotherapy. The combination of follicular B cell and mature DC densities allowed the identification of patients with the best clinical outcome. CONCLUSIONS: B-cell density represents a new prognostic biomarker for NSCLC patient survival, and makes the link between TLS and a protective B cell-mediated immunity.


Subject(s)
B-Lymphocyte Subsets/metabolism , Carcinoma, Non-Small-Cell Lung/immunology , Immunity, Humoral , Lung Neoplasms/immunology , Biomarkers/metabolism , Carcinoma, Non-Small-Cell Lung/mortality , Cohort Studies , Dendritic Cells/immunology , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Humans , Immunohistochemistry , Lung Neoplasms/mortality , Male , Prognosis , Retrospective Studies
15.
Cancer Immunol Immunother ; 63(10): 991-7, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25112529

ABSTRACT

A high density of intratumoral effector memory CD8+/Th1 T cells is associated with favorable prognosis in most cancers and may be induced or increased by immunotherapy. Efficient adaptive immune reactions are shaped in tumor adjacent tertiary lymphoid structures, which exhibit all characteristics of immunity generating lymphoid formations in reactive lymph nodes. Malignant tumor cells impact favorably or deleteriously their immune microenvironment if they bear genetic mutations that result in neo-antigens or by producing chemokines and cytokines that recruit lymphocytes and myeloid cells or increase inflammation and neo-angiogenesis. This intricate network of interactions results in control or escape of tumors, and its understanding will help define goals to monitor efficiency of immunotherapies.


Subject(s)
Neoplasms/immunology , Tumor Microenvironment/immunology , Animals , Humans , Immunotherapy/methods , Neoplasms/therapy , Prognosis
16.
J Immunol ; 189(7): 3528-37, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22922817

ABSTRACT

Complement is a major innate immune surveillance system. One of its most important regulators is the plasma protein factor H (FH). FH inactivation by mutations or by autoantibodies is associated with a thrombotic microangiopathy disease, atypical hemolytic uremic syndrome. In this study, we report the characterization of blood samples from 19 anti-FH Ab-positive atypical hemolytic uremic syndrome patients collected at the acute phase of the disease. Analyses of the functional consequences and epitope mapping, using both fluid phase and solid phase approaches, were performed. The anti-FH Abs perturbed FH-mediated cell protection (100%), inhibited FH interaction with C3 (46%), and caused C3 consumption (47%). The Abs were directed against multiple FH epitopes located at the N and C termini. In all tested patients, high titers of FH-containing circulating immune complexes were detected. The circulating immune complex titers correlated with the disease stage better than did the Ab titers. Our results show that anti-FH autoantibodies induce neutralization of FH at acute phase of the disease, leading to an overall impairment of several functions of FH, extending the role of autoantibodies beyond the impairment of the direct cell surface protection.


Subject(s)
Autoantibodies/physiology , Autoimmune Diseases/immunology , Complement Factor H/antagonists & inhibitors , Complement Factor H/immunology , Hemolytic-Uremic Syndrome/immunology , Neutralization Tests , Acute Disease , Adult , Autoimmune Diseases/diagnosis , Autoimmune Diseases/pathology , Child , Complement C3/metabolism , Complement Factor H/metabolism , Hemolytic-Uremic Syndrome/diagnosis , Hemolytic-Uremic Syndrome/pathology , Humans , Neutralization Tests/methods , Protein Binding/immunology
17.
Front Immunol ; 15: 1343020, 2024.
Article in English | MEDLINE | ID: mdl-38318190

ABSTRACT

The intricate relationship between anti-tumor immunity and autoimmunity is a complex yet crucial aspect of cancer biology. Tumor microenvironment often exhibits autoimmune features, a phenomenon that involves natural autoimmunity and the induction of humoral responses against self-antigens during tumorigenesis. This induction is facilitated by the orchestration of anti-tumor immunity, particularly within organized structures like tertiary lymphoid structures (TLS). Paradoxically, a significant number of cancer patients do not manifest autoimmune features during the course of their illness, with rare instances of paraneoplastic syndromes. This discrepancy can be attributed to various immune-mediated locks, including regulatory or suppressive immune cells, anergic autoreactive lymphocytes, or induction of effector cells exhaustion due to chronic stimulation. Overcoming these locks holds the risk to induce autoimmune mechanisms during cancer progression, a phenomenon notably observed with anti-immune checkpoint therapies, in contrast to more conventional treatments like chemotherapy or radiotherapy. Therefore, the challenge arises in managing immune-related adverse events (irAEs) induced by immune checkpoint inhibitors treatment, as decoupling them from the anti-tumor activity poses a significant clinical dilemma. This review summarizes recent advances in understanding the link between B-cell driven anti-tumor responses and autoimmune reactions in cancer patients, and discusses the clinical implications of this relationship.


Subject(s)
Autoimmunity , Neoplasms , Humans , Autoantibodies , Neoplasms/drug therapy , Autoantigens , Antibodies, Monoclonal/therapeutic use , Tumor Microenvironment
18.
Blood ; 118(18): 4853-62, 2011 Nov 03.
Article in English | MEDLINE | ID: mdl-21908423

ABSTRACT

Regulatory T cells (Tregs) may impede cancer vaccine efficacy in hematologic malignancies and cancer. CCR4 antagonists, an emergent class of Treg inhibitor, have been shown to block recruitment of Tregs mediated by CCL22 and CCL17. Our aim was to demonstrate the ability of a CCR4 antagonist (a small chemical molecule identified in silico) when combined with vaccines to break peripheral tolerance controlled by Tregs, a prerequisite for the induction of CD8(+) T cells against self Ags. Immunization of transgenic or normal mice expressing tumor-associated self Ags (Her2/neu, OVA, gp100) with a CCR4 antagonist combined with various vaccines led to the induction of effector CD8(+) T cells and partial inhibition of tumor growth expressing self Ags in both prophylactic and therapeutic settings. The CCR4 antagonist was more efficient than cyclophosphamide to elicit anti-self CD8(+) T cells. We also showed that the population of Tregs expressing CCR4 corresponded to memory (CD44(high)) and activated (ICOS(+)) Tregs, an important population to be targeted to modulate Treg activity. CCR4 antagonist represents a competitive class of Treg inhibitor able to induce functional anti-self CD8(+) T cells and tumor growth inhibition when combined with vaccines. High expression of CCR4 on human Tregs also supports the clinical development of this strategy.


Subject(s)
Autoantigens/immunology , CD8-Positive T-Lymphocytes/drug effects , Cancer Vaccines/administration & dosage , Neoplasms/therapy , Receptors, CCR4/antagonists & inhibitors , Tumor Escape/drug effects , Animals , Antigens, Neoplasm/immunology , Antineoplastic Agents/administration & dosage , Autoantigens/drug effects , CD8-Positive T-Lymphocytes/immunology , Combined Modality Therapy , Disease Models, Animal , Female , Humans , Immunologic Factors/administration & dosage , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , T-Cell Antigen Receptor Specificity/drug effects , T-Cell Antigen Receptor Specificity/immunology , Tumor Escape/immunology
19.
Clin Cancer Res ; 29(7): 1279-1291, 2023 04 03.
Article in English | MEDLINE | ID: mdl-36374555

ABSTRACT

PURPOSE: Immune checkpoint inhibitors (ICI) have revolutionized the treatment of patients with clear-cell renal cell carcinomas (ccRCC). Although analyses of transcriptome, genetic alterations, and the tumor microenvironment (TME) have shed light into mechanisms of response and resistance to these agents, the role of epigenetic alterations in this process remains fully unknown. EXPERIMENTAL DESIGN: We investigated the methylome of six ccRCC cohorts as well as one cell line dataset. Of note, we took advantage of the BIONIKK trial aiming to tailor treatments according to Paris Descartes 4-gene expression subgroups, and performed Illumina EPIC profiling for 46 samples related to patients treated with ipilimumab plus nivolumab, and 17 samples related to patients treated with sunitinib. RESULTS: A group of tumors associated with enhancer demethylation was discovered, namely TED. TED was associated with tumors with sarcomatoid differentiation and poor clinical outcome. TED harbored TET1 promoter demethylation, activated the gene expression signature of epithelial-mesenchymal transition and IL6/JAK/STAT3 pathways, and displayed a TME characterized by both immune activation and suppressive populations, fibroblast infiltration, and endothelial depletion. In addition, TED was a predictive factor of resistance to the combination of first-line ipilimumab-nivolumab in the BIONIKK clinical trial. Finally, TED was associated with activation of specific regulons, which we also found to be predictive of resistance to immunotherapy in an independent cohort. CONCLUSIONS: We report on the discovery of a novel epigenetic phenotype associated with resistance to ICIs that may pave the way to better personalizing patients' treatments. See related commentary by Zhou and Kim, p. 1170.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Humans , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Nivolumab/administration & dosage , Kidney Neoplasms/drug therapy , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Ipilimumab/administration & dosage , DNA Methylation , Phenotype , Tumor Microenvironment/genetics , Mixed Function Oxygenases , Proto-Oncogene Proteins/genetics
20.
Blood ; 116(6): 926-34, 2010 Aug 12.
Article in English | MEDLINE | ID: mdl-20439625

ABSTRACT

The anti-CD20 monoclonal antibody (mAb) rituximab has been used successfully for lymphoma therapy for more than 10 years. Although several direct mechanisms by which anti-CD20 mAbs act have been characterized in vitro, their specific role in clinical efficacy is still debated. Little is known about the possible antitumor immune response that they may induce in patients, despite clinical data suggesting a "vaccinal" effect. We show here that an initial treatment with anti-CD20 induces protection against human CD20-expressing tumor cells and allows immunocompetent mice to survive tumor challenge. This long-lasting protection requires the presence of the Fc portion of the anti-CD20 mAb and is achieved through the induction of a cellular immune response. Only CD4(+) cells were needed at the beginning of the treatment, but both CD4(+) and CD8(+) cells were required after tumor challenge to achieve protection. Finally, we show that interleukin-2 treatment, given after tumor challenge, improves the overall survival rate, compared with that obtained by anti-CD20 treatment alone. These findings demonstrate that anti-CD20 mAbs exert therapeutic effects through the induction of an adaptive cellular immune response, aside from any direct mechanisms involving effectors from innate immunity.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Cancer Vaccines/pharmacology , Thymoma/drug therapy , Thymus Neoplasms/drug therapy , Adoptive Transfer , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Murine-Derived , Antigens, Heterophile/immunology , Antineoplastic Agents/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Cell Line, Tumor , Disease Models, Animal , Drug Therapy, Combination , Female , Flow Cytometry , Humans , Immunocompetence/immunology , Immunoglobulin Fc Fragments/immunology , Interleukin-2/immunology , Interleukin-2/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Transplantation , Rituximab , Survival Rate , Thymoma/immunology , Thymoma/prevention & control , Thymus Neoplasms/immunology , Thymus Neoplasms/prevention & control , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL