Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Genet Med ; 26(1): 101007, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37860968

ABSTRACT

PURPOSE: BCL11B-related disorder (BCL11B-RD) arises from rare genetic variants within the BCL11B gene, resulting in a distinctive clinical spectrum encompassing syndromic neurodevelopmental disorder, with or without intellectual disability, associated with facial features and impaired immune function. This study presents an in-depth clinico-biological analysis of 20 newly reported individuals with BCL11B-RD, coupled with a characterization of genome-wide DNA methylation patterns of this genetic condition. METHODS: Through an international collaboration, clinical and molecular data from 20 individuals were systematically gathered, and a comparative analysis was conducted between this series and existing literature. We further scrutinized peripheral blood DNA methylation profile of individuals with BCL11B-RD, contrasting them with healthy controls and other neurodevelopmental disorders marked by established episignature. RESULTS: Our findings unveil rarely documented clinical manifestations, notably including Rubinstein-Taybi-like facial features, craniosynostosis, and autoimmune disorders, all manifesting within the realm of BCL11B-RD. We refine the intricacies of T cell compartment alterations of BCL11B-RD, revealing decreased levels naive CD4+ T cells and recent thymic emigrants while concurrently observing an elevated proportion of effector-memory expressing CD45RA CD8+ T cells (TEMRA). Finally, a distinct DNA methylation episignature exclusive to BCL11B-RD is unveiled. CONCLUSION: This study serves to enrich our comprehension of the clinico-biological landscape of BCL11B-RD, potentially furnishing a more precise framework for diagnosis and follow-up of individuals carrying pathogenic BCL11B variant. Moreover, the identification of a unique DNA methylation episignature offers a valuable diagnosis tool for BCL11B-RD, thereby facilitating routine clinical practice by empowering physicians to reevaluate variants of uncertain significance within the BCL11B gene.


Subject(s)
Intellectual Disability , Neurodevelopmental Disorders , Humans , CD8-Positive T-Lymphocytes/metabolism , Transcription Factors/genetics , Neurodevelopmental Disorders/genetics , Intellectual Disability/genetics , DNA Methylation/genetics , Tumor Suppressor Proteins/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism
2.
Nat Commun ; 8: 15221, 2017 05 24.
Article in English | MEDLINE | ID: mdl-28537262

ABSTRACT

Tissue-resident memory T cells (Trm) represent a new subset of long-lived memory T cells that remain in tissue and do not recirculate. Although they are considered as early immune effectors in infectious diseases, their role in cancer immunosurveillance remains unknown. In a preclinical model of head and neck cancer, we show that intranasal vaccination with a mucosal vector, the B subunit of Shiga toxin, induces local Trm and inhibits tumour growth. As Trm do not recirculate, we demonstrate their crucial role in the efficacy of cancer vaccine with parabiosis experiments. Blockade of TFGß decreases the induction of Trm after mucosal vaccine immunization, resulting in the lower efficacy of cancer vaccine. In order to extrapolate this role of Trm in humans, we show that the number of Trm correlates with a better overall survival in lung cancer in multivariate analysis. The induction of Trm may represent a new surrogate biomarker for the efficacy of cancer vaccine. This study also argues for the development of vaccine strategies designed to elicit them.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Carcinoma, Non-Small-Cell Lung/therapy , Immunologic Memory , Lung Neoplasms/therapy , Administration, Inhalation , Animals , Biomarkers, Tumor/metabolism , Cancer Vaccines/administration & dosage , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/metabolism , Female , Gene Expression Profiling , Genetic Vectors , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/therapy , Humans , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Mice, Inbred C57BL , Mucous Membrane/immunology , Prognosis , Retrospective Studies , Treatment Outcome
3.
Clin Cancer Res ; 22(3): 530-2, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26542056

ABSTRACT

Cancer vaccine boost via the cervicovaginal rather than the intramuscular route of immunization appears to be crucial to induce genital CD8(+) T cells and tumor regression. This clinical activity is correlated with the ability of the mucosal boost to elicit resident memory T cells in the genital tract.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines , Biomarkers , Humans , Immunization , Immunologic Memory , Neoplasms
4.
Mol Cancer Ther ; 14(6): 1336-45, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25833837

ABSTRACT

There is growing interest in the association of radiotherapy and immunotherapy for the treatment of solid tumors. Here, we report an extremely effective combination of local irradiation (IR) and Shiga Toxin B (STxB)-based human papillomavirus (HPV) vaccination for the treatment of HPV-associated head and neck squamous cell carcinoma (HNSCC). The efficacy of the irradiation and vaccine association was tested using a model of HNSCC obtained by grafting TC-1/luciferase cells at a submucosal site of the inner lip of immunocompetent mice. Irradiation and the STxB-E7 vaccine acted synergistically with both single and fractionated irradiation schemes, resulting in complete tumor clearance in the majority of the treated mice. A dose threshold of 7.5 Gy was required to elicit the dramatic antitumor response. The combined treatment induced high levels of tumor-infiltrating, antigen-specific CD8(+) T cells, which were required to trigger the antitumor activity. Treatment with STxB-E7 and irradiation induced CD8(+) T-cell memory, which was sufficient to exert complete antitumor responses in both local recurrences and distant metastases. We also report for the first time that a combination therapy based on local irradiation and vaccination induces an increased pericyte coverage (as shown by αSMA and NG2 staining) and ICAM-1 expression on vessels. This was associated with enhanced intratumor vascular permeability that correlated with the antitumor response, suggesting that the combination therapy could also act through an increased accessibility for immune cells. The combination strategy proposed here offers a promising approach that could potentially be transferred into early-phase clinical trials.


Subject(s)
Cancer Vaccines/administration & dosage , Head and Neck Neoplasms/therapy , Papillomavirus Infections/therapy , Radiotherapy/methods , Vaccination/methods , Actins/metabolism , Animals , Antigens/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/radiation effects , Cancer Vaccines/immunology , Cell Line , Combined Modality Therapy , Female , Flow Cytometry , Fluorescent Antibody Technique , Head and Neck Neoplasms/complications , Head and Neck Neoplasms/immunology , Intercellular Adhesion Molecule-1/metabolism , Mice, Inbred C57BL , Muscle, Smooth/chemistry , Papillomavirus Infections/complications , Papillomavirus Infections/immunology , Papillomavirus Vaccines/administration & dosage , Papillomavirus Vaccines/immunology , Pericytes/drug effects , Pericytes/metabolism , Pericytes/radiation effects , Proteoglycans/metabolism , Shiga Toxins/immunology
5.
J Exp Med ; 212(2): 139-48, 2015 Feb 09.
Article in English | MEDLINE | ID: mdl-25601652

ABSTRACT

Immune escape is a prerequisite for tumor development. To avoid the immune system, tumors develop different mechanisms, including T cell exhaustion, which is characterized by expression of immune inhibitory receptors, such as PD-1, CTLA-4, Tim-3, and a progressive loss of function. The recent development of therapies targeting PD-1 and CTLA-4 have raised great interest since they induced long-lasting objective responses in patients suffering from advanced metastatic tumors. However, the regulation of PD-1 expression, and thereby of exhaustion, is unclear. VEGF-A, a proangiogenic molecule produced by the tumors, plays a key role in the development of an immunosuppressive microenvironment. We report in the present work that VEGF-A produced in the tumor microenvironment enhances expression of PD-1 and other inhibitory checkpoints involved in CD8(+) T cell exhaustion, which could be reverted by anti-angiogenic agents targeting VEGF-A-VEGFR. In view of these results, association of anti-angiogenic molecules with immunomodulators of inhibitory checkpoints may be of particular interest in VEGF-A-producing tumors.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Neoplasms/immunology , Neoplasms/metabolism , Receptors, Vascular Endothelial Growth Factor/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , CD8-Positive T-Lymphocytes/drug effects , CTLA-4 Antigen/genetics , CTLA-4 Antigen/metabolism , Disease Models, Animal , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Mice , Neoplasms/genetics , Neoplasms/pathology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , Protein Binding , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/pharmacology
6.
Hum Vaccin Immunother ; 10(8): 2175-87, 2014.
Article in English | MEDLINE | ID: mdl-25424921

ABSTRACT

The mucosal immune system displays several adaptations reflecting the exposure to the external environment. The efficient induction of mucosal immune responses also requires specific approaches, such as the use of appropriate administration routes and specific adjuvants and/or delivery systems. In contrast to vaccines delivered via parenteral routes, experimental, and clinical evidences demonstrated that mucosal vaccines can efficiently induce local immune responses to pathogens or tumors located at mucosal sites as well as systemic response. At least in part, such features can be explained by the compartmentalization of mucosal B and T cell populations that play important roles in the modulation of local immune responses. In the present review, we discuss molecular and cellular features of the mucosal immune system as well as novel immunization approaches that may lead to the development of innovative and efficient vaccines targeting pathogens and tumors at different mucosal sites.


Subject(s)
Immunity, Mucosal , Vaccines/administration & dosage , Vaccines/immunology , Administration, Mucosal , Animals , Humans
7.
Oncoimmunology ; 2(6): e24534, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23894716

ABSTRACT

Various arguments support the development of a vaccine targeting human papillomavirus (HPV) for the treatment of HPV-associated head and neck cancer. However, the mucosal localization of this tumor, the HPV-driven downregulation of MHC Class I molecules and various other immunosuppressive mechanisms must be carefully considered to improve the clinical efficacy of such an immunotherapeutic strategy.

8.
Sci Transl Med ; 5(172): 172ra20, 2013 Feb 13.
Article in English | MEDLINE | ID: mdl-23408053

ABSTRACT

Although many human cancers are located in mucosal sites, most cancer vaccines are tested against subcutaneous tumors in preclinical models. We therefore wondered whether mucosa-specific homing instructions to the immune system might influence mucosal tumor outgrowth. We showed that the growth of orthotopic head and neck or lung cancers was inhibited when a cancer vaccine was delivered by the intranasal mucosal route but not the intramuscular route. This antitumor effect was dependent on CD8⁺ T cells. Indeed, only intranasal vaccination elicited mucosal-specific CD8⁺ T cells expressing the mucosal integrin CD49a. Blockade of CD49a decreased intratumoral CD8⁺ T cell infiltration and the efficacy of cancer vaccine on mucosal tumor. We then showed that after intranasal vaccination, dendritic cells from lung parenchyma, but not those from spleen, induced the expression of CD49a on cocultured specific CD8⁺ T cells. Tumor-infiltrating lymphocytes from human mucosal lung cancer also expressed CD49a, which supports the relevance and possible extrapolation of these results in humans. We thus identified a link between the route of vaccination and the induction of a mucosal homing program on induced CD8⁺ T cells that controlled their trafficking. Immunization route directly affected the efficacy of the cancer vaccine to control mucosal tumors.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/administration & dosage , Chemotaxis, Leukocyte , Head and Neck Neoplasms/therapy , Immunity, Mucosal , Lung Neoplasms/therapy , Nasal Mucosa/immunology , Papillomavirus Vaccines/administration & dosage , Adjuvants, Immunologic/administration & dosage , Administration, Intranasal , Animals , Antigens, CD/metabolism , Cancer Vaccines/immunology , Cell Proliferation , Cells, Cultured , Dendritic Cells/immunology , Female , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/pathology , Humans , Injections, Intramuscular , Integrin alpha Chains/metabolism , Integrin alpha1/metabolism , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lymph Nodes/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Mice, Inbred C57BL , Papillomavirus Vaccines/immunology , Shiga Toxins/administration & dosage , Spleen/immunology , Tumor Burden
9.
Cancer Res ; 73(1): 128-38, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23135914

ABSTRACT

Head and neck cancers positive for human papillomavirus (HPV) have a more favorable clinical outcome than HPV-negative cancers, but it is unknown why this is the case. We hypothesized that prognosis was affected by intrinsic features of HPV-infected tumor cells or differences in host immune response. In this study, we focused on a comparison of regulatory Foxp3(+) T cells and programmed death-1 (PD-1)(+) T cells in the microenvironment of tumors that were positive or negative for HPV, in two groups that were matched for various clinical and biologic parameters. HPV-positive head and neck cancers were more heavily infiltrated by regulatory T cells and PD-1(+) T cells and the levels of PD-1(+) cells were positively correlated with a favorable clinical outcome. In explaining this paradoxical result, we showed that these PD-1(+) T cells expressed activation markers and were functional after blockade of the PD-1-PD-L1 axis in vitro. Approximately 50% of PD-1(+) tumor-infiltrating T cells lacked Tim-3 expression and may indeed represent activated T cells. In mice, administration of a cancer vaccine increased PD-1 on T cells with concomitant tumor regression. In this setting, PD-1 blockade synergized with vaccine in eliciting antitumor efficacy. Our findings prompt a need to revisit the significance of PD-1-infiltrating T cells in cancer, where we suggest that PD-1 detection may reflect a previous immune response against tumors that might be reactivated by PD-1/PD-L1 blockade.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/immunology , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/virology , Lymphocytes, Tumor-Infiltrating/immunology , Programmed Cell Death 1 Receptor/biosynthesis , T-Lymphocytes/immunology , Animals , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/virology , Female , Flow Cytometry , Fluorescent Antibody Technique , Head and Neck Neoplasms/metabolism , Humans , Lymphocyte Activation/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Mice , Mice, Inbred C57BL , Papillomavirus Infections/complications , Papillomavirus Infections/immunology , Papillomavirus Infections/metabolism , Prognosis , Programmed Cell Death 1 Receptor/immunology , T-Lymphocyte Subsets/immunology , Tumor Microenvironment/immunology
10.
Oncoimmunology ; 1(3): 326-333, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22737608

ABSTRACT

CD4(+)CD25(+)Foxp3(+) regulatory T cells (Treg) have emerged as a dominant T cell population inhibiting anti-tumor effector T cells. Initial strategies used for Treg-depletion (cyclophosphamide, anti-CD25 mAb…) also targeted activated T cells, as they share many phenotypic markers. Current, ameliorated approaches to inhibit Treg aim to either block their function or their migration to lymph nodes and the tumor microenvironment. Various drugs originally developed for other therapeutic indications (anti-angiogenic molecules, tyrosine kinase inhibitors,etc) have recently been discovered to inhibit Treg. These approaches are expected to be rapidly translated to clinical applications for therapeutic use in combination with immunomodulators.

SELECTION OF CITATIONS
SEARCH DETAIL