7 research outputs found

    Mast cell-derived mediators promote murine neutrophil effector functions

    Get PDF
    Mast cells are able to trigger life-saving immune responses in murine models for acute inflammation. In such settings, several lines of evidence indicate that the rapid and protective recruitment of neutrophils initiated by the release of mast cell-derived pro-inflammatory mediators is a key element of innate immunity. Herein, we investigate the impact of mast cells on critical parameters of neutrophil effector function. In the presence of activated murine bone marrow-derived mast cells, neutrophils freshly isolated from bone marrow rapidly lose expression of CD62L and up-regulate CD11b, the latter being partly driven by mast cell-derived TNF and GM-CSF. Mast cells also strongly enhance neutrophil phagocytosis and generation of reactive oxygen species. All these phenomena partly depend on mast cell-derived TNF and to a greater extend on GM-CSF. Furthermore, spontaneous apoptosis of neutrophils is greatly diminished due to the ability of mast cells to deliver antiapoptotic GM-CSF. Finally, we show in a murine model for acute lung inflammation that neutrophil phagocytosis is impaired in mast cell-deficient Kit W-sh /Kit W-sh mice but can be restored upon mast cell engraftment. Thus, a previously underrated feature of mast cells is their ability to boost neutrophil effector functions in immune response

    Phase Ib evaluation of a self-adjuvanted protamine formulated mRNA-based active cancer immunotherapy, BI1361849 (CV9202), combined with local radiation treatment in patients with stage IV non-small cell lung cancer

    Get PDF
    Background: Preclinical studies demonstrate synergism between cancer immunotherapy and local radiation, enhancing anti-tumor effects and promoting immune responses. BI1361849 (CV9202) is an active cancer immunotherapeutic comprising protamine-formulated, sequence-optimized mRNA encoding six non-small cell lung cancer (NSCLC)-associated antigens (NY-ESO-1, MAGE-C1, MAGE-C2, survivin, 5T4, and MUC-1), intended to induce targeted immune responses. Methods: We describe a phase Ib clinical trial evaluating treatment with BI1361849 combined with local radiation in 26 stage IV NSCLC patients with partial response (PR)/stable disease (SD) after standard first-line therapy. Patients were stratified into three strata (1: non-squamous NSCLC, no epidermal growth factor receptor (EGFR) mutation, PR/SD after ≥4 cycles of platinum- and pemetrexed-based treatment [n = 16]; 2: squamous NSCLC, PR/SD after ≥4 cycles of platinum-based and non-platinum compound treatment [n = 8]; 3: non-squamous NSCLC, EGFR mutation, PR/SD after ≥3 and ≤ 6 months EGFR-tyrosine kinase inhibitor (TKI) treatment [n = 2]). Patients received intradermal BI1361849, local radiation (4 × 5 Gy), then BI1361849 until disease progression. Strata 1 and 3 also had maintenance pemetrexed or continued EGFR-TKI therapy, respectively. The primary endpoint was evaluation of safety; secondary objectives included assessment of clinical efficacy (every 6 weeks during treatment) and of immune response (on Days 1 [baseline], 19 and 61). Results: Study treatment was well tolerated; injection site reactions and flu-like symptoms were the most common BI1361849-related adverse events. Three patients had grade 3 BI1361849-related adverse events (fatigue, pyrexia); there was one grade 3 radiation-related event (dysphagia). In comparison to baseline, immunomonitoring revealed increased BI1361849 antigen-specific immune responses in the majority of patients (84%), whereby antigen-specific antibody levels were increased in 80% and functional T cells in 40% of patients, and involvement of multiple antigen specificities was evident in 52% of patients. One patient had a partial response in combination with pemetrexed maintenance, and 46.2% achieved stable disease as best overall response. Best overall response was SD in 57.7% for target lesions. Conclusion: The results support further investigation of mRNA-based immunotherapy in NSCLC including combinations with immune checkpoint inhibitors. Trial registration: ClinicalTrials.gov, Identifier: NCT01915524

    31st Annual Meeting and Associated Programs of the Society for Immunotherapy of Cancer (SITC 2016) : part two

    Get PDF
    Background The immunological escape of tumors represents one of the main ob- stacles to the treatment of malignancies. The blockade of PD-1 or CTLA-4 receptors represented a milestone in the history of immunotherapy. However, immune checkpoint inhibitors seem to be effective in specific cohorts of patients. It has been proposed that their efficacy relies on the presence of an immunological response. Thus, we hypothesized that disruption of the PD-L1/PD-1 axis would synergize with our oncolytic vaccine platform PeptiCRAd. Methods We used murine B16OVA in vivo tumor models and flow cytometry analysis to investigate the immunological background. Results First, we found that high-burden B16OVA tumors were refractory to combination immunotherapy. However, with a more aggressive schedule, tumors with a lower burden were more susceptible to the combination of PeptiCRAd and PD-L1 blockade. The therapy signifi- cantly increased the median survival of mice (Fig. 7). Interestingly, the reduced growth of contralaterally injected B16F10 cells sug- gested the presence of a long lasting immunological memory also against non-targeted antigens. Concerning the functional state of tumor infiltrating lymphocytes (TILs), we found that all the immune therapies would enhance the percentage of activated (PD-1pos TIM- 3neg) T lymphocytes and reduce the amount of exhausted (PD-1pos TIM-3pos) cells compared to placebo. As expected, we found that PeptiCRAd monotherapy could increase the number of antigen spe- cific CD8+ T cells compared to other treatments. However, only the combination with PD-L1 blockade could significantly increase the ra- tio between activated and exhausted pentamer positive cells (p= 0.0058), suggesting that by disrupting the PD-1/PD-L1 axis we could decrease the amount of dysfunctional antigen specific T cells. We ob- served that the anatomical location deeply influenced the state of CD4+ and CD8+ T lymphocytes. In fact, TIM-3 expression was in- creased by 2 fold on TILs compared to splenic and lymphoid T cells. In the CD8+ compartment, the expression of PD-1 on the surface seemed to be restricted to the tumor micro-environment, while CD4 + T cells had a high expression of PD-1 also in lymphoid organs. Interestingly, we found that the levels of PD-1 were significantly higher on CD8+ T cells than on CD4+ T cells into the tumor micro- environment (p < 0.0001). Conclusions In conclusion, we demonstrated that the efficacy of immune check- point inhibitors might be strongly enhanced by their combination with cancer vaccines. PeptiCRAd was able to increase the number of antigen-specific T cells and PD-L1 blockade prevented their exhaus- tion, resulting in long-lasting immunological memory and increased median survival

    Mastzellen als Verstärker wichtiger Effektorfunktionen neutrophiler Granulozyten

    No full text
    Mastzellen sind an der Entstehung und Regulation von Entzündungsreaktionen unterschiedlicher Genese beteiligt und leisten somit auch innerhalb der angeborenen Immunität einen wichtigen Beitrag zur Pathogenabwehr. Mastzellen sind jedoch auch in entzündliche Prozesse involviert, welche chronische Erkrankungen auslösen oder verstärken können. Zentral ist hierbei ihre Fähigkeit, durch Produktion inflammatorischer Mediatoren die lokalen Blutgefäße zu aktivieren und somit rasch neutrophile Granulozyten zum Entzündungsort zu rekrutieren. Über eine direkte Interaktion zwischen Mastzellen und Neutrophilen ist bisher jedoch wenig bekannt.rnIn der vorliegenden Arbeit konnte gezeigt werden, dass aktivierte Mastzellen essentielle Effektorfunktionen neutrophiler Granulozyten verstärken. So erhöhen Mastzellen den Aktivierungsstatus von Neutrophilen, was anhand der Modulation der Aktivierungsmarker CD11b und CD62L auf der Zelloberfläche von Neutrophilen demonstriert wurde. Die Phagozytoseaktivität von Neutrophilen wird in Anwesenheit aktivierter Mastzellen enorm gesteigert. Dies wird hauptsächlich durch Mastzell-produziertes TNF und GM-CSF vermittelt, wie in Experimenten mit Mastzellen aus TNF- und GM-CSF-defizienten Mäusen gezeigt wurde. Darüber hinaus wird die Produktion reaktiver Sauerstoffradikale von Neutrophilen in Gegenwart aktivierter Mastzellen erhöht. Dieser Effekt wird hauptsächlich durch GM-CSF der Mastzellen induziert, während TNF nur einen geringen Einfluss hat. Weiterhin zeigten in vitro Experimente, dass die Anwesenheit aktivierter Mastzellen die Lebensdauer von Neutrophilen erhöht. Die verminderte Apoptose von Neutrophilen wird einzig durch GM-CSF bewirkt. rnZur Validierung der in vitro Studien wurden in vivo Untersuchungen zur Mastzell-abhängigen Phagozytoseaktivität von Neutrophilen durchgeführt. Mit Hilfe des Modells einer LPS-induzierten akuten Atemwegsentzündung in Mastzell-defizienten und Wildtyp-Tieren des kongenen Stammes konnte bestätigt werden, dass Mastzellen auch in vivo die Phagozytosefunktion von Neutrophilen verstärken. Die Ergebnisse dieser Arbeit zeigen somit, dass Mastzellen in Entzündungsprozessen auch eine Rolle als lokaler Verstärker von Neutrophilenfunktionen einnehmen können. rnMast cells are able to induce and to regulate inflammatory responses in diverse settings, and thus play an essential role in the host defense against pathogens. However, mast cells are also involved in inflammatory processes, which can play an important role in the development or amplification of chronic diseases. In such settings, the key element of mast cells is the production of inflammatory mediators, leading to the activation of endothelial cells and the recruitment of neutrophils. Until now, it is not much known about a direct interaction between mast cells and neutrophils. rnThe results of this experimental work could demonstrate that activated mast cells are able to boost essential effector functions of neutrophils. Analyses revealed that mast cells promote the activation of neutrophils determined by modulation of the activation markers CD11b and CD62L on neutrophil surface. Furthermore, in the presence of activated mast cells neutrophil phagocytosis is strongly enhanced. These effects depend on mast cell-derived TNF and GM-CSF, which was demonstrated by using mast cells derived from TNF- and GM-CSF-deficient mice. Additionally, activated mast cells enhance the generation of reactive oxygen species by neutrophils. This is mainly due to mast cell-derived GM-CSF whereas TNF produced by mast cells showed little effect on neutrophil oxidative burst. Further in vitro experiments could show that activated mast cells can extend the lifespan of neutrophils. Mast cell-derived GM-CSF is solely responsible for the delayed apoptosis of neutrophils. To corroborate the in vitro findings, mast cells´ influence on neutrophil phagocytosis was also investigated in an in vivo mouse model. In a model for LPS-induced acute lung inflammation in mast cell-deficient and wildtype mice, it could be shown that mast cells enhance neutrophil phagocytosis in vivo.rnThese results demonstrate that mast cells have the capability to boost neutrophil effector functions in immune responses.r

    Mast cell-derived mediators promote murine neutrophil effector functions

    Full text link
    Mast cells are able to trigger life-saving immune responses in murine models for acute inflammation. In such settings, several lines of evidence indicate that the rapid and protective recruitment of neutrophils initiated by the release of mast cell-derived pro-inflammatory mediators is a key element of innate immunity. Herein, we investigate the impact of mast cells on critical parameters of neutrophil effector function. In the presence of activated murine bone marrow-derived mast cells, neutrophils freshly isolated from bone marrow rapidly lose expression of CD62L and up-regulate CD11b, the latter being partly driven by mast cell-derived TNF and GM-CSF. Mast cells also strongly enhance neutrophil phagocytosis and generation of reactive oxygen species. All these phenomena partly depend on mast cell-derived TNF and to a greater extend on GM-CSF. Furthermore, spontaneous apoptosis of neutrophils is greatly diminished due to the ability of mast cells to deliver antiapoptotic GM-CSF. Finally, we show in a murine model for acute lung inflammation that neutrophil phagocytosis is impaired in mast cell-deficient Kit W-sh /Kit W-sh mice but can be restored upon mast cell engraftment. Thus, a previously underrated feature of mast cells is their ability to boost neutrophil effector functions in immune response
    corecore