48 research outputs found

    Impact of p38 mitogen-activated protein kinase inhibition on immunostimulatory properties of human 6-sulfo LacNAc dendritic cells

    Get PDF
    p38 Mitogen-activated protein kinase (MAPK) plays a crucial role in the induction and regulation of innate and adaptive immunity. Furthermore, p38 MAPK can promote tumor invasion, metastasis, and angiogenesis. Based on these properties, p38 MAPK inhibitors emerged as interesting candidates for the treatment of immune-mediated disorders and cancer. However, the majority of p38 MAPK inhibitor-based clinical trials failed due to poor efficacy or toxicity. Further studies investigating the influence of p38 MAPK inhibitors on immunomodulatory capabilities of human immune cells may improve their therapeutic potential. Here, we explored the impact of the p38 MAPK inhibitor SB203580 on the pro-inflammatory properties of native human 6-sulfo LacNAc dendritic cells (slanDCs). SB203580 did not modulate maturation of slanDCs and their capacity to promote T-cell proliferation. However, SB203580 significantly reduced the production of pro-inflammatory cytokines by activated slanDCs. Moreover, inhibition of p38 MAPK impaired the ability of slanDCs to differentiate naïve CD4(+) T cells into T helper 1 cells and to stimulate interferon-γ secretion by natural killer cells. These results provide evidence that SB203580 significantly inhibits various important immunostimulatory properties of slanDCs. This may have implications for the design of p38 MAPK inhibitor-based treatment strategies for immune-mediated disorders and cancer

    A Novel Modular Antigen Delivery System for Immuno Targeting of Human 6-sulfo LacNAc-Positive Blood Dendritic Cells (SlanDCs)

    Get PDF
    Previously, we identified a major myeloid-derived proinflammatory subpopulation of human blood dendritic cells which we termed slanDCs (e.g. Schäkel et al. (2006) Immunity 24, 767-777). The slan epitope is an O-linked sugar modification (6-sulfo LacNAc, slan) of P-selectin glycoprotein ligand-1 (PSGL-1). As slanDCs can induce neoantigen-specific CD4+ T cells and tumor-reactive CD8+ cytotoxic T cells, they appear as promising targets for an in vivo delivery of antigens for vaccination. However, tools for delivery of antigens to slanDCs were not available until now. Moreover, it is unknown whether or not antigens delivered via the slan epitope can be taken up, properly processed and presented by slanDCs to T cells.Single chain fragment variables were prepared from presently available decavalent monoclonal anti-slan IgM antibodies but failed to bind to slanDCs. Therefore, a novel multivalent anti-slanDC scaffold was developed which consists of two components: (i) a single chain bispecific recombinant diabody (scBsDb) that is directed on the one hand to the slan epitope and on the other hand to a novel peptide epitope tag, and (ii) modular (antigen-containing) linker peptides that are flanked at both their termini with at least one peptide epitope tag. Delivery of a Tetanus Toxin-derived antigen to slanDCs via such a scBsDb/antigen scaffold allowed us to recall autologous Tetanus-specific memory T cells.In summary our data show that (i) the slan epitope can be used for delivery of antigens to this class of human-specific DCs, and (ii) antigens bound to the slan epitope can be taken up by slanDCs, processed and presented to T cells. Consequently, our novel modular scaffold system may be useful for the development of human vaccines

    Interaktionen von dendritischen Zellen und Effektorzellen der frühen antitumoralen Immunabwehr

    Get PDF
    In den letzten Jahren ergaben sich vermehrt Hinweise, dass dendritische Zellen (DCs) zu einer Stimulation von Natürlichen „Killer“ (NK)-Zellen in der Lage sind, die als zytotoxische Effektorzellen des angeborenen Immunsystems Tumorzellen eliminieren. Aus diesem Grund bestand ein wesentliches Ziel dieser Arbeit in der Analyse der Wechselwirkungen zwischen nativen DCs und NK-Zellen. Dazu wurden slanDCs verwendet, welche die größte DC-Subpopulation des Blutes repräsentieren. Zunächst wurde evaluiert, ob slanDCs eine effiziente Aktivierung von NK-Zellen bewirken. Als ein Ergebnis zeigte sich, dass Lipopolysaccharid (LPS)-stimulierte slanDCs sowohl zu einer verstärkten Expression des Aktivierungsmarkers CD69 auf der Oberfläche von NK-Zellen als auch zur Induktion der NK-Zell-Proliferation führen. Darüber hinaus wurde erstmals die slanDC-abhängige Erhöhung der Expression von aktivierenden Rezeptoren (NKp46, NKp44, NKp30) und Korezeptoren (2B4, DNAM-1) auf NK-Zellen demonstriert, welche essentiell für die NK-Zell-vermittelte Erkennung und Lyse von Tumorzellen sind. In weiteren Untersuchungen induzierten LPS-aktivierte slanDCs eine erhebliche Produktion von Interferon (IFN)-gamma in NK-Zellen, welches proliferationshemmend auf Tumorzellen und aktivierend auf T-Lymphozyten wirkt. Funktionelle Analysen ergaben, dass aktivierte slanDCs das zytotoxische Potential von NK-Zellen gegenüber der Tumorzelllinie K-562 deutlich verstärken. Untersuchungen der zugrunde liegenden Mechanismen zeigten die herausragende Bedeutung von IL-12, das sowohl die Steigerung der IFN-gamma-Sekretion als auch die Zunahme der zytolytischen Aktivität von NK-Zellen induzierte. Darüber hinaus konnte erstmals gezeigt werden, dass LPS-aktivierte slanDCs eine Zytotoxizität von NK-Zellen gegenüber frisch etablierten Blasten von Patienten mit akuter myeloischer Leukämie induzieren. In weiteren Untersuchungen wurde evaluiert, ob NK-Zellen ihrerseits die immunstimulatorischen Eigenschaften von slanDCs beeinflussen. Die Analysen zeigten erstmals, dass unstimulierte NK-Zellen die Expression von MHC-Klasse II-Molekülen, kostimulatorischen Molekülen und Adhäsionsmolekülen auf slanDCs deutlich erhöhen und somit ihre Fähigkeit zur Aktivierung von CD8+ T-Lymphozyten sowie CD4+ T-Helferzellen fördern. NK-Zellen führen ebenfalls zu einer deutlichen Verstärkung der Produktion von IL-12 durch LPS-stimulierte slanDCs. Darüber hinaus zeigte sich, dass NK-Zellen die Sekretion des immunsuppressiven Zytokins IL-10 durch LPS-stimulierte slanDCs reduzieren. In weiteren Analysen wurde demonstriert, dass die Interaktionen mit NK Zellen die Fähigkeit von LPS-aktivierten slanDCs zur Programmierung naiver CD4+ T-Lymphozyten in IFN-gamma-produzierende T-Helfer-1-Zellen deutlich verstärken. Diese Ergebnisse zeigten deutlich, dass stimulierte slanDCs und NK-Zellen in der Lage sind, sich wechselseitig zu aktivieren. NKT-Zellen repräsentieren eine weitere bedeutende Effektorzellpopulation der frühen antitumoralen Immunabwehr, die durch Sekretion von Zytokinen und ein ausgeprägtes zytolytisches Potential zur Elimination von Tumorzellen beiträgt. Deshalb wurden im Rahmen dieser Arbeit erstmals die Wechselwirkungen zwischen slanDCs und NKT-Zellen analysiert. Dabei verstärkten LPS-stimulierte slanDCs die Expression des Aktivierungsmarkers CD69 auf NKT-Zellen. Darüber hinaus induzierten LPS-aktivierte slanDCs eine deutliche IFN-gamma-Produktion in NKT-Zellen, wobei erneut die zentrale Rolle von IL-12 gezeigt wurde. Diese Ergebnisse demonstrierten, dass stimulierte slanDCs zu einer effektiven Aktivierung von NKT Zellen in der Lage sind. In abschließenden Untersuchungen wurde die Wirkung von NKT-Zellen auf slanDCs evaluiert. Dabei verstärkten NKT-Zellen die Maturierung von slanDCs erheblich und führten zu einer signifikanten Steigerung der IL-12-Produktion sowie zu einer Reduktion der IL-10-Freisetzung in Abhängigkeit von IFN-gamma. Die gewonnenen Daten demonstrierten, dass NKT-Zellen und slanDCs zu einer gegenseitigen Aktivierung befähigt sind. Die im Rahmen dieser Dissertation gewonnenen Erkenntnisse zu den Interaktionen von slanDCs und NK- bzw. NKT-Zellen können einen wesentlichen Beitrag zum Verständnis der Immunabwehr von Tumoren leisten und die Konzeption neuer antitumoraler Therapiestrategien unterstützen

    Prognostic impact of the post-treatment T cell composition and spatial organization in soft tissue sarcoma patients treated with neoadjuvant hyperthermic radio(chemo)therapy

    Get PDF
    Soft tissue sarcomas (STS) form a heterogeneous group of tumors sharing a mesenchymal origin. Despite good local control of the disease, the occurrence of distant metastases often limits survival of STS patients with localized, high-risk tumors of the extremities. Accumulating evidence suggests a central role for the tumor immune microenvironment in determining the clinical outcome and response to therapy. Thus, it has been reported that STS patients with a high immune signature and especially presence of B cells and tertiary lymphoid structures display improved overall survival and response to checkpoint inhibitor treatment. Here, we explored the effect of curative multimodal therapy on the T cell landscape of STS using multiplex immunohistochemistry. We analyzed the phenotype, frequency, and spatial distribution of STS-infiltrating CD8+ T cells by staining for CD8, 4-1BB, Granzyme B, Ki67, PD-1, and LAG-3 as well as CD3+ T helper cells using a panel consisting of CD3, T-bet, GATA3, RORγT, FoxP3, and Ki67. All patients received neoadjuvant radiotherapy plus locoregional hyperthermia with or without chemotherapy. While the treatment-naïve biopsy sample allows an analysis of baseline T cell infiltration levels, both intra- and peritumoral areas of the matched resected tissue were analyzed to assess composition and spatial distribution of the T cell compartment and its therapeutic modulation. Generally, post-treatment tissues displayed lower frequencies of CD3+ and CD8+ T cells. Association with clinical data revealed that higher post-treatment frequencies of peritumoral and intratumoral CD3+ T cells and intratumoral PD-1+ CD8+ T cells were significantly associated with improved disease-free survival (DFS), while these densities had no prognostic significance in the biopsy. Upon spatial analysis, a high ratio of intratumoral to peritumoral CD8+ T cells emerged as an independent prognostic marker for longer DFS. These results indicate that the STS T cell landscape is altered by multimodal therapy and may influence the clinical outcome of patients. An enhanced understanding of the STS immune architecture and its modulation by neoadjuvant therapy may pave the way towards novel treatment modalities and improve the long-term clinical outcome of STS patients

    Activation of Dendritic Cells by the Novel Toll-Like Receptor 3 Agonist RGC100

    Get PDF
    Toll-like receptor (TLR) 3 agonists emerged as attractive candidates for vaccination strategies against tumors and pathogens. An important mechanism of action of such agonists is based on the activation of TLR3-expressing dendritic cells (DCs), which display a unique capacity to induce and stimulate T-cell responses. In this context, it has been demonstrated that targeting of TLR3 by double-stranded RNA such as poly(I:C) results in potent activation of DCs. Major disadvantages of poly(I:C) comprise its undefined chemical structure and very poor homogeneity, with subsequent unpredictable pharmacokinetics and high toxicity. In the present study, we evaluated the physicochemical properties and biological activity of the novel TLR3 agonist RGC100. RGC100 has a defined chemical structure, with a defined length (100 bp) and molecular weight (64.9 KDa) and a good solubility. RGC100 is stable in serum and activates myeloid DCs through TLR3 targeting, as evidenced by gene silencing experiments. Activation of mouse and human myeloid CD1c+ DCs by RGC100 leads to secretion of several proinflammatory cytokines. In addition, RGC100 improves the ability of CD1c+ DCs to stimulate T-cell proliferation. Due to its physicochemical properties and its immunostimulatory properties, RGC100 may represent a promising adjuvant for prophylactic and therapeutic vaccination strategies

    Tumor-Associated Antigens for Specific Immunotherapy of Prostate Cancer

    Get PDF
    Prostate cancer (PCa) is the most common noncutaneous cancer diagnosis and the second leading cause of cancer-related deaths among men in the United States. Effective treatment modalities for advanced metastatic PCa are limited. Immunotherapeutic strategies based on T cells and antibodies represent interesting approaches to prevent progression from localized to advanced PCa and to improve survival outcomes for patients with advanced disease. CD8+ cytotoxic T lymphocytes (CTLs) efficiently recognize and destroy tumor cells. CD4+ T cells augment the antigen-presenting capacity of dendritic cells and promote the expansion of tumor-reactive CTLs. Antibodies mediate their antitumor effects via antibody-dependent cellular cytotoxicity, activation of the complement system, improving the uptake of coated tumor cells by phagocytes, and the functional interference of biological pathways essential for tumor growth. Consequently, several tumor-associated antigens (TAAs) have been identified that represent promising targets for T cell- or antibody-based immunotherapy. These TAAs comprise proteins preferentially expressed in normal and malignant prostate tissues and molecules which are not predominantly restricted to the prostate, but are overexpressed in various tumor entities including PCa. Clinical trials provide evidence that specific immunotherapeutic strategies using such TAAs represent safe and feasible concepts for the induction of immunological and clinical responses in PCa patients. However, further improvement of the current approaches is required which may be achieved by combining T cell- and/or antibody-based strategies with radio-, hormone-, chemo- or antiangiogenic therapy

    Immune Monitoring of Cancer Patients Prior to and During CTLA-4 or PD-1/PD-L1 Inhibitor Treatment

    No full text
    Targeting the immune checkpoint receptors cytotoxic T lymphocyte antigen 4 (CTLA-4), programmed cell death protein 1 (PD-1), or programmed cell death 1 ligand 1 (PD-L1) represents a very attractive treatment modality for tumor patients. The administration of antibodies against these receptors can promote efficient antitumor effects and can induce objective clinical responses in about 20–40% patients with various tumor types, accompanied by improved survival. Based on their therapeutic efficiency, several antibodies have been approved for the treatment of tumor patients. However, many patients do not respond to checkpoint inhibitor therapy. Therefore, the identification of biomarkers is required to guide patient selection for this treatment modality. Here, we summarize recent studies investigating the PD-L1 expression or mutational load of tumor tissues as well as the frequency and phenotype of immune cells in tumor patients prior to and during CTLA-4 or PD-1/PD-L1 inhibitor treatment

    Chimeric Antigen Receptor-Engineered T Cells for Immunotherapy of Cancer

    Get PDF
    CD4+ and CD8+ T lymphocytes are powerful components of adaptive immunity, which essentially contribute to the elimination of tumors. Due to their cytotoxic capacity, T cells emerged as attractive candidates for specific immunotherapy of cancer. A promising approach is the genetic modification of T cells with chimeric antigen receptors (CARs). First generation CARs consist of a binding moiety specifically recognizing a tumor cell surface antigen and a lymphocyte activating signaling chain. The CAR-mediated recognition induces cytokine production and tumor-directed cytotoxicity of T cells. Second and third generation CARs include signal sequences from various costimulatory molecules resulting in enhanced T-cell persistence and sustained antitumor reaction. Clinical trials revealed that the adoptive transfer of T cells engineered with first generation CARs represents a feasible concept for the induction of clinical responses in some tumor patients. However, further improvement is required, which may be achieved by second or third generation CAR-engrafted T cells
    corecore