4,936 research outputs found

    Targeted delivery of TRAIL for immunotherapy of ovarian cancer

    Get PDF
    Het menselijk eiwit TRAIL helpt de vorming en verspreiding van kanker te onderdrukken. Het komt voor op de buitenkant van bepaalde immuuncellen, maar ook in vrije vorm in het lichaam. Deze vorm wordt sTRAIL genoemd. sTRAIL bindt zich wel aan gezonde cellen, maar richt er geen schade aan. Als het zich aan een kankercel bindt, kan het deze uitschakelen. Daardoor is het mogelijk geschikt om kanker te bestrijden. Recent onderzoek heeft echter laten zien dat sTRAIL veel minder effectief is dan werd gedacht. Dit komt onder andere doordat het snel door het lichaam wordt uitgescheiden. Promovendus Marco de Bruyn onderzocht de effectiviteit van een nieuwe aanpak, waarbij sTRAIL genetisch wordt gekoppeld aan een eiwit dat de kankercellen ‘herkent’ en zich eraan bindt. Hiermee kan sTRAIL selectief en met hoge concentraties worden afgeleverd aan het oppervlak van kankercellen. Deze methode blijkt zowel in een laboratoriumopstelling als in een diermodel zeer goed te werken, zo laat De Bruyn zien. Ook onderzocht De Bruyn of de antikankeractiviteit van immuuncellen versterkt wanneer sTRAIL gericht wordt afgeleverd aan het celoppervlak van immuuncellen. Dit bleek het geval te zijn: de immuuncellen bleken tot wel vijfhonderd maal krachtiger tegen kanker op te treden. In de toekomst kan deze methode wellicht worden ingezet in de strijd tegen kanker

    Tumor-infiltrating lymphocytes in the immunotherapy era

    Get PDF
    The clinical success of cancer immune checkpoint blockade (ICB) has refocused attention on tumor-infiltrating lymphocytes (TILs) across cancer types. The outcome of immune checkpoint inhibitor therapy in cancer patients has been linked to the quality and magnitude of T cell, NK cell, and more recently, B cell responses within the tumor microenvironment. State-of-the-art single-cell analysis of TIL gene expression profiles and clonality has revealed a remarkable degree of cellular heterogeneity and distinct patterns of immune activation and exhaustion. Many of these states are conserved across tumor types, in line with the broad responses observed clinically. Despite this homology, not all cancer types with similar TIL landscapes respond similarly to immunotherapy, highlighting the complexity of the underlying tumor-immune interactions. This observation is further confounded by the strong prognostic benefit of TILs observed for tumor types that have so far respond poorly to immunotherapy. Thus, while a holistic view of lymphocyte infiltration and dysfunction on a single-cell level is emerging, the search for response and prognostic biomarkers is just beginning. Within this review, we discuss recent advances in the understanding of TIL biology, their prognostic benefit, and their predictive value for therapy

    Automated causal inference in application to randomized controlled clinical trials

    Full text link
    Randomized controlled trials (RCTs) are considered the gold standard for testing causal hypotheses in the clinical domain; however, the investigation of prognostic variables of patient outcome in a hypothesized cause–effect route is not feasible using standard statistical methods. Here we propose a new automated causal inference method (AutoCI) built on the invariant causal prediction (ICP) framework for the causal reinterpretation of clinical trial data. Compared with existing methods, we show that the proposed AutoCI allows one to clearly determine the causal variables of two real-world RCTs of patients with endometrial cancer with mature outcome and extensive clinicopathological and molecular data. This is achieved via suppressing the causal probability of non-causal variables by a wide margin. In ablation studies, we further demonstrate that the assignment of causal probabilities by AutoCI remains consistent in the presence of confounders. In conclusion, these results confirm the robustness and feasibility of AutoCI for future applications in real-world clinical analysis

    The Novel Immune Checkpoint GPR56 Is Expressed on Tumor-Infiltrating Lymphocytes and Selectively Upregulated upon TCR Signaling

    Get PDF
    High levels of tumor-infiltrating lymphocytes (TILs) in the tumor microenvironment (TME) are associated with a survival benefit in various cancer types and the targeted (re)activation of TILs is an attractive therapeutic anti-cancer approach that yields curative responses. However, current T cell targeting strategies directed at known immune checkpoints have not increased objective response rates for all cancer types, including for epithelial ovarian cancer (EOC). For this reason, the identification of new immune checkpoints that regulate T cell immunity remains of great interest. One yet largely uninvestigated checkpoint of potential interest is the G protein-coupled receptor 56 (GPR56), which belongs to the adhesion GPCR family. GPR56 was originally reported to function in cerebral cortical development and in anti-depressant response, but also in cancer. Recently, GPR56 was identified as an inhibitory receptor expressed on human NK cells that by cis-interaction with the tetraspanin CD81 attenuated the cytotoxic activity of NK cells. This NK cell checkpoint could be blocked by an GPR56 antibody, leading to increased cytotoxicity. Interestingly, GPR56 expression has also been reported on cytokine producing memory CD8 T lymphocytes and may thus represent a T cell checkpoint as well. Here, GPR56 mRNA expression was characterized in the context of TILs, with GPR56 expression being detected predominantly in tumor infiltrating CD8 T cells with a cytotoxic and (pre-)exhausted phenotype. In accordance with this mRNA profile, TILs from ovarian cancer patients expressed GPR56 primarily within the effector memory and central memory T cell subsets. On T cells from healthy donors the expression was limited to effector memory and terminally differentiated T cells. Notably, GPR56 expression further increased on TILs upon T cell receptor (TCR)-mediated stimulation in co-cultures with cancer cells, whereas GPR56 expression on healthy primary human T cells did not. Further, the ectopic expression of GPR56 significantly reduced the migration of GPR56-positive T cells. Taken together, GPR56 is a potential immune-checkpoint in EOC found on (pre-)exhausted CD8 TILs that may regulate migratory behavior
    • 

    corecore