Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 1966-1966
    Abstract: The importance of the tumor microenvironment in the development of B-cell chronic lymphocytic lekuemia (CLL) is now widely accepted. Previous studies within our and other groups revealed the establishment of an inflammatory milieu in CLL characterized by enhanced expression and secretion of several cytokines and their receptors. Using 250 CLL serum samples and 50 age-matched controls, we found significantly increased levels of the TNFα receptors TNFR1 and TNFR2 in the patients that positively correlated with an adverse clinical outcome. Based on these findings we aimed to investigate the functional role of TNFR signaling in CLL development. In contrast to healthy B cells that do not express TNFα receptors, we detected TNFR1 expression in CLL cells upon survival-inducing culture conditions and in lymph node sections of 80 CLL patients, where TNFR1 signals co-localized to the B cell marker CD20 and were mainly present within proliferation centers. These findings were confirmed in Eµ-TCL1 mice, a well-established mouse model for CLL. Here, CLL cells in the peripheral blood were negative for TNFR1. However, the cells upregulated the receptor upon entering the spleen where they get into contact with accessory cells, receive survival stimuli and undergo proliferation. In addition, increased levels of soluble TNFR1 in the serum were confirmed in the mice. After ligand binding, TNFR1 can activate two different signaling pathways: (1) the extrinsic apoptosis cascade through its death receptor domain, or (2) survival-inducing NFκB signaling. The latter pathway can be blocked by wogonin, a naturally occurring monoflavonoid. In a multitude of in vitro and in vivo studies, wogonin has been shown to exert anti-oxidant, anti-inflammatory and anti-tumor activities. By ex vivo treatment of CLL cells with TNFα we observed NFκB activation which was reversed by TNFα-blocking antibody, suggesting TNFR1-mediated survival signaling in CLL. Therefore, we aimed to test whether wogonin can prevent this signaling in CLL cells and might therefore represent a novel potential drug for CLL. In CLL cocultures, wogonin treatment resulted in a concentration-dependent apoptosis induction, which was significantly increased by the addition of TNFα. To test the effects of wogonin in vivo, we transplanted isogenic, immunocompetent wild-type mice with CLL cells from leukemic TCL1 animals and treated them daily with 40 mg/kg wogonin. When treatment was started 2 days after transplantation, wogonin significantly reduced spleen weights and lead to a reduced CLL content in the spleen, the bone marrow and the peritoneal cavity. If treatment was started in advanced disease stage, wogonin slightly lowered spleen weight and the CLL content in the spleen, whereas the percentage of CLL cells in the peripheral blood was increased. Interestingly, here wogonin treatment resulted in a loss of cell surface TNFR1 expression in splenic CLL cells and increased TNFR1 levels in the serum. These data suggest that wogonin induces a redistribution of CLL cells in vivo, preventing their homing to lymphoid organs and that loss of TNFR1 expression might be involved in this process. In summary, our results show that TNFR1 signaling is involved in CLL cell activation and survival. Targeting this pathway with wogonin reduces CLL cell viability in vitro and leukemia development in TCL1 mice. Disclosures No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2014
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    The American Association of Immunologists ; 2015
    In:  The Journal of Immunology Vol. 194, No. 1_Supplement ( 2015-05-01), p. 141.33-141.33
    In: The Journal of Immunology, The American Association of Immunologists, Vol. 194, No. 1_Supplement ( 2015-05-01), p. 141.33-141.33
    Abstract: Previous in vitro studies have shown that myeloid cells play a key role in apoptosis-resistance in chronic lymphocytic leukemia (CLL). However, their composition and function have not been thoroughly investigated in vivo. Using the Eµ-TCL1 mouse model of CLL, we observed severe skewing of myeloid cell populations along with CLL development. Monocytes and M2-like macrophages infiltrated the peritoneal cavity of leukemic mice. Monocytes expressing high levels of inflammatory factors like IL-10, TNFα and CXCL9 accumulated also in the spleen, the main site of disease, and were severely skewed towards Ly6Clow phenotype. Along with that, the percentage of MHC-IIhi dendritic cells (DCs) and macrophages significantly dropped in the spleen, suggesting monocyte differentiation arrest. Myeloid cell depletion using liposomal clodronate resulted in a significant control of CLL development, repair of innate immune cell phenotypes and partial resolution of systemic inflammation. Also, CLL-induced loss of naive T cells was resolved. CLL-associated monocytes and DCs aberrantly express high PD-L1 levels suggesting their contribution in T cell defects. Importantly, PD-L1 blockade very effectively prevented CLL development and was accompanied with a re-activation of immune effector cells. This included restoration of mature macrophages and MHC-IIhi DCs. Our preclinical data suggests that targeting myeloid cell survival and immunosuppression can serve as a novel strategy for CLL immunotherapy.
    Type of Medium: Online Resource
    ISSN: 0022-1767 , 1550-6606
    RVK:
    RVK:
    Language: English
    Publisher: The American Association of Immunologists
    Publication Date: 2015
    detail.hit.zdb_id: 1475085-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 126, No. 2 ( 2015-07-09), p. 203-211
    Abstract: In vivo PD-L1 blockade prevents CLL development in the Eµ-TCL1 adoptive transfer model. In vivo PD-L1 blockade normalizes T-cell and myeloid cell populations and immune effector functions.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2015
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 3298-3298
    Abstract: Chronic lymphocytic leukemia (CLL) is a malignancy of mature B cells that is characterized by apoptosis resistance and dysfunctional immune system. The chronic nature and slow development of the disease indicates a contribution of CLL-induced inflammation in the disease course. Previous reports suggested a potential role of myeloid cells in mediating these defects. However, the composition and function of CLL-associated myeloid cells have not been thoroughly investigated in an in vivo system. Here, we used the well-established CLL mouse model, Eµ-TCL1 mice (TCL1), to characterize changes within myeloid cell populations along with CLL development and the influence of their depletion on disease progression and immune dysfunction. We have recently shown that CLL development in TCL1 mice is associated with massive changes within myeloid cell populations. In the peritoneal cavity (PC) of leukemic mice we observed an infiltration of monocytes and an M2-like skewing of macrophages according to phenotypical and signaling signatures. Along this line, monocytes infiltrated the spleens of leukemic animals, both in primary CLL and adoptive transfer models, which is most likely due to high CCL2 serum levels. These monocytes lost the inflammatory Ly6Chi subset and were severely skewed towards Ly6Clow patrolling monocytes, accompanied by high expression of adhesion and angiogenic molecules like ICAM1, PECAM1 and MMP14. Gene expression profiling of splenic myeloid cells from TCL1 mice revealed an enrichment of various genes involved in dendritic cell (DC) maturation and MHC-II-mediated antigen presentation. However, the numbers of MHC-IIhi mature DCs and macrophages were significantly decreased, suggesting a monocyte differentiation arrest leading to impaired anti-tumor immune response. The observed transcriptional upregulation of multiple inflammatory cytokines like TNF-α, CXCL9, CXCL10 and CXCL16 in monocytes was confirmed by serum cytokine arrays, and is likely due to the overexpression of the pro-inflammatory regulator TREM-1. In addition, TCL1 monocytes upregulated the expression of several inhibitory molecules like PD-L1, IL-10, IL1ra and IL4i1 suggesting an impaired immune function. While CLL-induced immune dysfunction is a well-established phenomenon, the contribution of myeloid cells in this context was not clear. We therefore sought to determine the in vivo effects of myeloid cell depletion on CLL development and its associated immune defects. For that purpose we used liposomal clodronate to selectively ablate macrophages and monocytes from young wild-type mice adoptively transferred with murine CLL. Our data clearly show control of CLL development in clodronate-treated mice relative to control liposomes as demonstrated by decreased spleen weight (1.09 vs. 0.54 g, p 〈 0.0001) and a significant drop in tumor load, defined as CD5+CD19+ cells, in spleen (60.58% vs. 42.25%), peripheral blood (43% vs 11.8%), PC (66.2% vs 3.1%), lymph nodes (4.9% vs 1.2%) and bone marrow (1.9% vs 0.8%). In addition, we observed changes in immune effector cells in response to myeloid cell depletion suggesting better immune status in treated mice. Interestingly, the loss of macrophages/monocytes was compensated by increased splenic monocyte proliferation as shown by EdU incorporation in vivo. In contrast to control mice, the repopulating monocytes upon clodronate treatment were largely inflammatory Ly6Chi monocytes. In summary, our data show that skewing of myeloid cells actively contributes to CLL development via; 1) enhancing the survival of leukemic cells, and 2) suppressing anti-tumor immune functions. In the absence of monocytes and macrophages, disease development is delayed in mice adoptively transferred with murine CLL. Therefore, we suggest that targeting non-malignant myeloid cells in CLL might serve as a novel strategy for CLL immunotherapy. Disclosures No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2014
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 717-717
    Abstract: Background: Clinical studies have demonstrated that targeted immunotherapy using PD-1/PD-L1 antibodies induces tumor regression and prolongs disease stabilization in advanced solid cancers. Data on the clinical efficacy in hematological malignancies is largely missing, even though PD-L1/PD-1 interactions have been described as major mediators of immune dysfunction in several leukemias and lymphomas. They are therefore ideal to study if PD-L1/PD-1 blockade has the potential to control disease by restoring anti-tumor immune responses. Several groups showed that chronic lymphocytic leukemia (CLL) provokes immune evasion via PD-L1/PD-1 inhibitory signaling, and that this is very closely mirrored in the Eµ-TCL1 (TCL1) murine model for CLL. Our recent data suggest that in this model, aberrant PD-L1 expression in myeloid cells contributes to the immune defect in CLL. We further demonstrated that the T cell and myeloid cell immune defects in ageing leukemic mice can be induced in young wild-type (WT) mice by adoptive transfer (AT) of murine CLL. In the current study, we used the AT model to test if in vivoPD-L1 blockade corrects leukemia-induced cellular immune dysfunction in myeloid and T cells and enhances anti-tumor immunity. Methods: WT mice transplanted with 4x107 TCL1 splenocytes were randomized to treatment with 10 mg/kg α-murine-PD-L1 (n=15) or isotype antibody (n=10), which was administered i.p. every 3 days starting 1 day after AT, and sacrificed 31 days later. Matched non-transplanted WT mice (n=6) served as additional controls. Immune cell subsets, expression of immune checkpoint markers and T cell effector functions were analyzed by multicolor flow cytometry using cells isolated form spleen, peripheral blood (PB), bone marrow (BM) and peritoneal cavity (PC). Cell proliferation was measured by EdU incorporation in vivo. Immune synapse (IS) formation was assessed by confocal microscopy. Serum cytokines were quantified by multiplex bead arrays. Results: We first confirmed successful engraftment and presence of disease by immunohistochemistry. Compared to isotype controls, α-PD-L1 treated mice had significantly lower spleen weights (median 0.2 g vs 0.9 g, p 〈 0.0001) and a highly significant lower relative frequency of CD19+CD5+ CLL lymphocytes in spleen (1.55% vs 71.69%), PB (10.5% vs 63.53%) and BM (0.26% vs 2.74%) demonstrating very effective tumor control. Compared to non-transplanted animals, α-PD-L1 treated mice showed alterations in almost all phenotypical and functional immune cell parameters, especially in regards to immune cell activation, indicating encounter with and immunological challenge by CLL cells. Along with disease control, α-PD-L1 treated mice had improved immune status as multiple inflammatory cytokines in the serum, including IL-10, TNF-α, CCL2 and GM-CSF were decreased and splenic infiltration of monocytes was reduced. While CLL development skewed monocytes towards Ly6Clow patrolling monocytes, α-PD-L1 treatment restored the presence of Ly6Chi inflammatory monocytes and decreased the expression of adhesion molecules ICAM-1 and PECAM-1. These monocytes regained their differentiation capacity as shown by increased numbers of macrophages and mature MHC-IIhi dendritic cells in the spleens of treated mice. In the T cell compartment, in vivo PD-L1 blockade prevented the CLL-induced CD4/CD8 ratio inversion, the loss of naïve CD8 T cells and the shift towards antigen-experienced and terminally differentiated T cells in spleen, BM and PB. Aberrant expression of immune checkpoint markers PD-1, KLRG-1, LAG-3, and 2B4 was also significantly reduced. The CLL-associated loss of intracellular IL-2 and the increased secretion of IL-4 and IFN-γ in CD4 T cells were prevented in α-PD-L1 treated mice. Respective cytokine patterns were observed in the serum. Functionally, PD-L1 blockade restored CD8 degranulation and IS formation to the level of healthy T cells, and significantly improved both ex vivo and in vivoT cell proliferation. Conclusion: Our in vivodata demonstrate that early PD-L1 blockade very effectively controls CLL development and enables complex effector function of myeloid and T cells, thus restoring anti-tumor immune responses. Targeting PD-L1/PD-1 interactions should therefore be further explored in clinical studies, potentially in combination with novel substances. BH/FM and MS/JGG contributed equally to first and last authorship. Disclosures No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2014
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages