GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
  • 1
    In: Journal of Leukocyte Biology, Oxford University Press (OUP), Vol. 102, No. 2 ( 2017-05-26), p. 475-486
    Abstract: Epigenetic changes in chromatin structure have been recently associated with the deregulated expression of critical genes in normal and malignant processes. HDAC11, the newest member of the HDAC family of enzymes, functions as a negative regulator of IL-10 expression in APCs, as previously described by our lab. However, at the present time, its role in other hematopoietic cells, specifically in neutrophils, has not been fully explored. In this report, for the first time, we present a novel physiologic role for HDAC11 as a multifaceted regulator of neutrophils. Thus far, we have been able to demonstrate a lineage-restricted overexpression of HDAC11 in neutrophils and committed neutrophil precursors (promyelocytes). Additionally, we show that HDAC11 appears to associate with the transcription machinery, possibly regulating the expression of inflammatory and migratory genes in neutrophils. Given the prevalence of neutrophils in the peripheral circulation and their central role in the first line of defense, our results highlight a unique and novel role for HDAC11. With the consideration of the emergence of new, selective HDAC11 inhibitors, we believe that our findings will have significant implications in a wide range of diseases spanning malignancies, autoimmunity, and inflammation.
    Type of Medium: Online Resource
    ISSN: 0741-5400 , 1938-3673
    RVK:
    Language: English
    Publisher: Oxford University Press (OUP)
    Publication Date: 2017
    detail.hit.zdb_id: 2026833-6
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Molecular Immunology, Elsevier BV, Vol. 63, No. 2 ( 2015-02), p. 579-585
    Type of Medium: Online Resource
    ISSN: 0161-5890
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2015
    detail.hit.zdb_id: 2013448-4
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2016
    In:  Cancer Immunology Research Vol. 4, No. 11_Supplement ( 2016-11-01), p. B109-B109
    In: Cancer Immunology Research, American Association for Cancer Research (AACR), Vol. 4, No. 11_Supplement ( 2016-11-01), p. B109-B109
    Abstract: Immunotherapy strategies for the treatment of melanoma have achieved impressive clinical outcomes over the past decade. Response rates to checkpoint blockade by PD-1 and CTLA-4 antibodies range from 15-40%, while in adoptive cell therapy using tumor infiltrating lymphocytes (TILs), anti-tumor response is observed in approximately 50%. However, the need to improve immunotherapies is evident as the majority of patients are unresponsive to treatment. Dysfunctional T-cells are thought to contribute to failed responses to checkpoint inhibition. As such we sought to investigate the ability of drugs targeting the epigenetic regulatory machinery as a means to alter T-cell function(s) and improve the anti-melanoma response. Here we demonstrate that the HDAC6 selective inhibitor ACY1215 disrupts mTORC signaling pathways in T-cells obtained from melanoma patients. Phosphorylation of mTOR, RAPTOR and the downstream molecules AKT, SGK1, PKCa and S6K were reduced on CD4 and CD8 T-cells after ACY1215 in vitro treatment (p & lt;0.05). The levels of the Th2 cytokines IL-4, IL-6, IL-10 generated by ACY1215-treated T-cells (p & lt;0.05) were also decreased. Similar results were achieved with an SGK1 inhibitor, in agreement with published data demonstrating SGK1 as a regulator of Th2 polarization. Since the mTOR/RAPTOR complex is known to be involved in determining T regulatory (Treg) function, the effects of ACY1215 on Tregs were evaluated. Treatment in vitro with ACY1215 decreased phosphorylated mTOR and RAPTOR in Tregs, and reduced the levels of FOXP3. In a functional suppression assay, ACY1215-treated Tregs displayed a reduced ability to impair proliferation of effector T-cells (Teff) compared to control (DMSO: 10% vs ACY1215: 25% Teff proliferation, p & lt;0.05). To explore whether HDAC inhibition during expansion of tumor infiltrating lymphocytes (TIL) for adoptive transfer would improve their quality and anti-tumor reactivity, TIL isolated from melanoma surgical biopsies were cultured in vitro with IL-2 and ACY1215. Treatment with ACY1215 led to an accumulation of central memory CD4 and CD8 TILs (p & lt;0.01 and p & lt;0.05, respectively), which was maintained even after rapid expansion with anti-CD3 and anti-CD28 stimulation in vitro. Similarly, ACY1215 treatment of T-cells derived from peripheral blood of melanoma patients and healthy donors also displayed an increased central memory phenotype, characterized by expression of CD45RO, CD62L and CCR7 (p & lt;0.05). Inhibition of AKT has been shown to increase T-cells with memory characteristics, and the use of an AKT inhibitor also resulted in accumulation of central memory T-cells. Confirming the observed phenotypic changes, microarray analysis of ACY1215-treated TILs revealed up-regulation of genes associated with a T-cell central memory and inflammatory response (e.g. SELL, LEF1, TNFRSF9) and downregulation of genes associated with Treg function (e.g. LGMN, CXCL8). Collectively these data suggest that reprogramming T-cells with epigenetic modulators may improve melanoma immunotherapy by reducing Treg suppression and production of immunosuppressive cytokines, while favoring generation of central memory T-cells. Citation Format: Andressa L. Sodre, David M. Woods, Amod Sarnaik, Brian C. Betts, Jeffrey S. Weber. Epigenetic reprogramming of T-cells from metastatic melanoma patients enhances central memory and decreases Th2/Treg phenotypes [abstract]. In: Proceedings of the Second CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; 2016 Sept 25-28; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2016;4(11 Suppl):Abstract nr B109.
    Type of Medium: Online Resource
    ISSN: 2326-6066 , 2326-6074
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    detail.hit.zdb_id: 2732517-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2015
    In:  Cancer Immunology Research Vol. 3, No. 12 ( 2015-12-01), p. 1375-1385
    In: Cancer Immunology Research, American Association for Cancer Research (AACR), Vol. 3, No. 12 ( 2015-12-01), p. 1375-1385
    Abstract: Expression of PD-1 ligands by tumors and interaction with PD-1–expressing T cells in the tumor microenvironment can result in tolerance. Therapies targeting this coinhibitory axis have proven clinically successful in the treatment of metastatic melanoma, non–small cell lung cancer, and other malignancies. Therapeutic agents targeting the epigenetic regulatory family of histone deacetylases (HDAC) have shown clinical success in the treatment of some hematologic malignancies. Beyond direct tumor cell cytotoxicity, HDAC inhibitors have also been shown to alter the immunogenicity and enhance antitumor immune responses. Here, we show that class I HDAC inhibitors upregulated the expression of PD-L1 and, to a lesser degree, PD-L2 in melanomas. Evaluation of human and murine cell lines and patient tumors treated with a variety of HDAC inhibitors in vitro displayed upregulation of these ligands. This upregulation was robust and durable, with enhanced expression lasting past 96 hours. These results were validated in vivo in a B16F10 syngeneic murine model. Mechanistically, HDAC inhibitor treatment resulted in rapid upregulation of histone acetylation of the PD-L1 gene leading to enhanced and durable gene expression. The efficacy of combining HDAC inhibition with PD-1 blockade for treatment of melanoma was also explored in a murine B16F10 model. Mice receiving combination therapy had a slower tumor progression and increased survival compared with control and single-agent treatments. These results highlight the ability of epigenetic modifiers to augment immunotherapies, providing a rationale for combining HDAC inhibitors with PD-1 blockade. Cancer Immunol Res; 3(12); 1375–85. ©2015 AACR.
    Type of Medium: Online Resource
    ISSN: 2326-6066 , 2326-6074
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2015
    detail.hit.zdb_id: 2732517-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2014
    In:  Cancer Research Vol. 74, No. 19_Supplement ( 2014-10-01), p. 643-643
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 74, No. 19_Supplement ( 2014-10-01), p. 643-643
    Abstract: It has become increasingly clear that histone deacetylases (HDACs), a family of epigenetic modifiers, play intimate roles in both tumor and immune biology. Indeed, two HDAC inhibitors (HDACi) are FDA approved for the treatment of cutaneous T-cell leukemia, and various others are currently undergoing clinical trials for the treatment of both solid and hematological malignancies. Beyond direct anti-tumor effects, HDACi have shown pronounced abilities to augment the immunogenicity of tumor cells leading to an improved ant-tumor immune response. Furthermore, in addition to anti-tumor effects, several studies have demonstrated a profound ability of HDACi to tip the scale between tolerance and inflammation in antigen presenting cells, leading to enhanced T-cell activation. However, less well studied is the ability of HDACi to directly influence the function and fate of T-cells. Here we report that the HDACi JNJ-264-81585 (Quisinostat) is able to augment the pro-inflammatory phenotype of T-cells in vitro and in vivo. Quisinostat is a hydroxamic acid with potent sub-nanomolar inhibition of several HDACs, and reported anti-tumor activity. Here we report that Quisinostat has a potent HDAC inhibition in T-cells, with sub-nanomolar doses increasing histone 3 acetylation levels in a dose dependent manner. Intriguingly, when treated with Quisinostat in vitro, activated murine CD4+ and CD8+ T-cells produced significantly higher levels of the pro-inflammatory cytokines IL-2, IFN-g, and TNF. Concomitantly, treated T-cells produced decreased levels of IL-6 and IL-17. To determine the efficacy of Quisinostat at augmenting T-cell function in vivo, a novel T-cell adoptive transfer model was utilized. In this model syngeneic T-cells from B6.SJL mice were stained with the proliferation tracking dye CellTrace™ Violet and activated in the presence of Quisinostat, then subsequently adoptively transferred into sub-lethally irradiated C57BL/6 recipient mice. Analysis of proliferation of transferred T-cells showed that CD8+ T-cells treated with Quisinostat had significantly higher rates of proliferation than those treated with DMSO control. As well, in a result similar to that seen in vitro, adoptively transferred T-cells had skewing towards a predominantly CD8+ composition. Finally, T-cells treated with Quisinostat displayed a reconstitution advantage over DMSO treated cells, with higher percentages of transferred T-cells relative to endogenous T-cells. Collectively, these results demonstrate a profound and important ability of HDAC inhibition to modulate the T-cell response, highlighting a role of HDACi, particularly Quisinostat, in augmenting the efficacy of adoptive cell therapy, giving rationale for clinical investigation. Citation Format: David M. Woods, Andressa L. Sodre, Jason B. Brayer, Eduardo M. Sotomayor. The histone deacetylase inhibitor Quisinostat augments the anti-tumor reponses of T-cells: Implications in adoptive cell therapy. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 643. doi:10.1158/1538-7445.AM2014-643
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2014
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 74, No. 19_Supplement ( 2014-10-01), p. 4090-4090
    Abstract: Histone deacetylase inhibitors (HDACi) have shown remarkable anti-tumor activity, leading to FDA approval of two HDACi for the treatment of CTCL and several others currently at various stages of clinical development for the treatment of both solid and hematological malignancies. Previous work from our lab has shown that treatment with HDACi results in increased expression of pro-inflammatory promoting surface markers on melanoma cells, promoting enhanced T-cell activation. Recent clinical trial data has shown that blockade of the PD1/PDL1 interaction is effective in the treatment of melanoma, renal cell and non-small cell lung cancer. Importantly, responses to PD1 blocking antibodies were preferentially seen in patients with tumors expressing PDL1. Here we report that HDACi targeting class I HDACs, but not class II, augments expression of PDL1 in melanoma cells. Two murine and five human melanoma cell lines were treated for up to 72 hours with DMSO, LBH589 (pan-HDACi), MS275 (class I inhibitor), MGCD0103 (class I inhibitor), an HDAC6 specific inhibitor, or a class IIa inhibitor. Using flow cytometry, dose dependent, increases in PDL1 expression were found in the LBH589, MS275 and MGCD0103 treated groups, but not in those receiving HDAC6i or class IIa inhibitor, relative to DMSO. Increased expression was noted as early as 24 hours after treatment and peaked at 72 to 96 hours post-treatment. As IFN-γ is known to upregulate the expression of PDL1 in both normal and transformed cells, we evaluated whether these results were associated with induction of IFN-γ expression by the melanoma cells. However, no detectable levels of IFN-γ were seen in either non-treated, class I HDACi, or class II HDACi-treated cells. Melanoma cells treated with HDACi in addition to IFN-γ have enhanced expression of PDL1 relative to either treatment alone. To further gain insight into the specific HDAC regulating the expression of PDL1, preliminary experiments utilizing knockdowns (KD) of individual class I HDACs were performed. In all KD melanoma cells no increase in PDL1 expression was seen, suggesting that the increased expression of PDL1 is dependent on inhibition of multiple class I HDACs. Supporting this conclusion, treatment of class I HDAC-KDs with HDACi recapitulates the increased PDL1 expression seen with WT melanoma. Finally, in preliminary in vivo experiments combining treatment of melanoma bearing mice with anti-PDL1 antibodies, mice receiving the combination treatment had a survival advantage over those receiving PDL1 blocking antibodies or HDACi alone. These results provide a strong rationale for the evaluation of combination therapies utilizing PDL1 or PD1 blocking antibodies in combination with HDACi. Furthermore, these results support the need for further development and investigation of iso-specific HDACi in order to obtain more directed therapeutic efficacy. Citation Format: David M. Woods, Andressa L. Sodre, Eva Sahakian, John Powers, Maritza Lienlaf-Moreno, Patricio Perez-Villarroel, Alejandro Villagra, Javier Pinilla-Ibarz, Eduardo Sotomayor. Inhibition of class I histone deacetylases promotes robust and durable enhancement of PDL1 expression in melanoma: Rationale for combination therapy. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 4090. doi:10.1158/1538-7445.AM2014-4090
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2014
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Cancer Immunology, Immunotherapy, Springer Science and Business Media LLC, Vol. 67, No. 3 ( 2018-3), p. 381-392
    Type of Medium: Online Resource
    ISSN: 0340-7004 , 1432-0851
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2018
    detail.hit.zdb_id: 1458489-X
    detail.hit.zdb_id: 195342-4
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    Online Resource
    Online Resource
    The American Association of Immunologists ; 2017
    In:  The Journal of Immunology Vol. 198, No. 1_Supplement ( 2017-05-01), p. 56.7-56.7
    In: The Journal of Immunology, The American Association of Immunologists, Vol. 198, No. 1_Supplement ( 2017-05-01), p. 56.7-56.7
    Abstract: PD-1 blockade has remarkable response rates in the treatment of melanoma; however, many patients fail to respond, and biomarkers and mechanisms of response remain unknown. We investigated the roles of Tregs, a population of immunosuppressive T-cells, in patient response to nivolumab. Tregs had a decrease in suppressive function in patients with positive outcomes (p=0.03), but not in patients with negative outcomes. Patients with no evidence of disease (NED) displayed increased percentages of Tregs post-nivolumab (p=0.04), but relapsing patients did not. RNA-seq analysis revealed genes significantly changed after treatment were distinct between Tcons and Tregs (~13% overlap) and between NED and relapsing patient Tregs (~2% overlap). Pathway analysis showed an increase in proliferation associated pathways in NED patient Tregs, but not relapsing patients. We found increased phosphoSTAT3 (pSTAT3) expression in Tregs from patients with positive outcomes (p=0.01), but not in patients with negative outcomes. Mechanistically, in vitro culturing of T-cells with αPD-1 resulted in increases in pSTAT3 expression (p=0.03) and increased percentages of Tregs (p=0.001). Culturing with αPD-1 also enhanced production of IL-10 (p=0.02), and the addition of a STAT3 inhibitor reduced the increases in IL-10 levels (p=0.01) and Treg percentages (p=0.01). The addition of an IL-10 neutralizing antibody also reduced the increased Tregs resulting from αPD-1 (p=0.01). These results support a model in which PD-1 blockade increases pSTAT3 expression leading to enhanced IL-10 production and Treg percentages, suggesting that pSTAT3 induction and reduced suppressive function are biomarkers of melanoma patient response to nivolumab.
    Type of Medium: Online Resource
    ISSN: 0022-1767 , 1550-6606
    RVK:
    RVK:
    Language: English
    Publisher: The American Association of Immunologists
    Publication Date: 2017
    detail.hit.zdb_id: 1475085-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Blood, American Society of Hematology, Vol. 130, No. 2 ( 2017-07-13), p. 146-155
    Abstract: T cells from HDAC11KO mice have increased effector functions and mediate more rapid and potent GVHD. HDAC11 associates with the Eomes and Tbet gene promoter regions in resting cells and disassociates upon activation.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2017
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2016
    In:  Cancer Immunology Research Vol. 4, No. 11_Supplement ( 2016-11-01), p. A067-A067
    In: Cancer Immunology Research, American Association for Cancer Research (AACR), Vol. 4, No. 11_Supplement ( 2016-11-01), p. A067-A067
    Abstract: Blockade of the co-inhibitory receptor PD-1 has had unprecedented success in the treatment of metastatic melanoma, with response rates of approximately 40% in previously untreated patients. However, since the majority of patients fail to respond, a better understanding of the mechanisms of action involved in response/resistance and novel approaches to overcome resistance are needed. We evaluated T-cells present in peripheral blood samples of resected stage III/IV melanoma patients treated with adjuvant nivolumab. Data from intra-patient, paired pre-treatment/post-treatment samples showed a significant (p & lt;0.05) increase in phosphorylated STAT3 (pSTAT3) in patients without relapse, but not in patients relapsing. Increases in regulatory T-cells (Tregs) (p & lt;0.05) in relapsers, but not in non-relapsers, and OX40 expression on Tregs (p & lt;0.05) in both relapsers and non-relapsers were also observed. Based on these data we evaluated the ability of PD-1 blocking antibodies to induce pSTAT3 signaling in vitro. Culturing with anti-PD-1 significantly increased levels of pSTAT3 in T-cells, including Tregs, both at the basal state (p & lt;0.05) and post-CD3/CD28 activation (p & lt;0.01). Additionally, culturing T-cells with PD-1 blocking antibodies increased the production of the STAT3 regulated cytokine IL-10 (p & lt;0.05), the number of Tregs (p & lt;0.01) and Treg expression of OX40 (p & lt;0.05). Increases in all these markers/populations were abrogated with the addition of a STAT3 inhibitor (p & lt;0.01). Similar increases in the number of Tregs and the expression of OX40 were also found when T-cells were treated in vitro with recombinant IL-10. Addition of IL-10 neutralizing antibodies ameliorated upregulation of Treg numbers and OX40 expression resulting from PD-1 blockade. Additionally, serum levels of IL-10 were found to be significantly elevated in non-relapsing patients relative to relapsers (p & lt;0.05). RNA-seq analysis comparing Tregs from relapsers to non-relapsers further supported the observed increase in Tregs resulting from nivolumab treatment, with pathway analyses demonstrating significantly increased cell cycle, DNA replication, mitosis and other proliferation related pathways in Tregs from non-relapsed patients. Next we evaluated changes in the function of patient Tregs in an allogenic mixed lymphocyte suppression assay. Tregs from non-relapsers had a significant reduction in suppressive capacity post-nivolumab treatment in paired analyses (p & lt;0.05), while Tregs from relapsers had no significant change. Post-treatment samples from relapses were also found to be significantly more suppressive (p & lt;0.01) than those of non-relapsers. Additionally, decreased percentages of CCR4+CD45RA+ Tregs, previously described as having enhanced suppressive capacity, were found in non-relapsers post-treatment, but not in relapsers. A similar decrease in CCR4+CD45RA+ Tregs was achieved in vitro using OX40 agonist antibodies. Therefore, we propose that PD-1 blockade triggers STAT3 signaling, a hereunto unknown effect. These data support a model in which STAT3 induction by PD-1 blockade results in IL-10 production, which induces proliferation of Tregs with reduced suppressive capacity and triggers OX40 expression. Based on these results and previous findings of others demonstrating that ligation of OX40 expressed on Tregs hinders suppressive capacity, combining OX40 agonist antibodies may enhance the effectiveness of PD-1 blockade by reducing Treg suppression. Citation Format: David M. Woods, Rupal Ramakrishnan, Andressa L. Sodré, Anders Berglund, Jeffrey Weber. PD-1 blockade enhances OX40 expression on regulatory T-cells and decreases suppressive function through induction of phospho-STAT3 signaling [abstract]. In: Proceedings of the Second CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; 2016 Sept 25-28; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2016;4(11 Suppl):Abstract nr A067.
    Type of Medium: Online Resource
    ISSN: 2326-6066 , 2326-6074
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    detail.hit.zdb_id: 2732517-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...