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 for ImmunoTherapy of Cancer, BMJ, Vol. 3, No. S2 ( 2015-12)
    Type of Medium: Online Resource
    ISSN: 2051-1426
    Language: English
    Publisher: BMJ
    Publication Date: 2015
    detail.hit.zdb_id: 2719863-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2016
    In:  Cancer Immunology Research Vol. 4, No. 1_Supplement ( 2016-01-01), p. A058-A058
    In: Cancer Immunology Research, American Association for Cancer Research (AACR), Vol. 4, No. 1_Supplement ( 2016-01-01), p. A058-A058
    Abstract: GITRL, (Glucocorticoid-Induced Tumor Necrosis Factor Receptor Ligand, TNFSF18) is a member of the TNF family of ligands and naturally exists as a membrane-anchored type II protein that self assembles as a trimer. GITRL activates the co-stimulatory receptor GITR. A novel single-gene linkerless GITRL trimer was shown to be functional when fused to either the N- or C-terminus of an immunoglobulin Fc domain, offering a flexible strategy that may also be amenable to the production of bispecific agents. GITRL-Fc activated GITR signaling more effectively than prototype GITR agonist antibody DTA-1. GITRL-Fc promoted a robust anti-tumor immune response in several murine tumor graft models, including the apparent total regression of some treated tumors. GITRL-Fc potentiated tumor specific T-cell responses, particularly of the Th1 type, increased antigen-specific CD8 response, and promoted a reduction in Treg-mediated immune-suppressive activity. Citation Format: Fumiko Axelrod, Hyun-Bae Jie, Erin Mayes, Jorge Monteon, Minu Srivastava, Rui Yun, Inkyung Angie Park, Austin Gurney. GITRL-Fc, an immunotherapeutic agent that stimulates T-cell-mediated antitumor immune response. [abstract]. In: Proceedings of the CRI-CIMT-EATI-AACR Inaugural International Cancer Immunotherapy Conference: Translating Science into Survival; September 16-19, 2015; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2016;4(1 Suppl):Abstract nr A058.
    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 ...
  • 3
    Online Resource
    Online Resource
    Informa UK Limited ; 2023
    In:  Expert Opinion on Drug Discovery Vol. 18, No. 4 ( 2023-04-03), p. 417-428
    In: Expert Opinion on Drug Discovery, Informa UK Limited, Vol. 18, No. 4 ( 2023-04-03), p. 417-428
    Type of Medium: Online Resource
    ISSN: 1746-0441 , 1746-045X
    Language: English
    Publisher: Informa UK Limited
    Publication Date: 2023
    detail.hit.zdb_id: 2272966-5
    SSG: 15,3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    Online Resource
    Online Resource
    Elsevier BV ; 2004
    In:  Journal of Biological Chemistry Vol. 279, No. 29 ( 2004-07), p. 30540-30545
    In: Journal of Biological Chemistry, Elsevier BV, Vol. 279, No. 29 ( 2004-07), p. 30540-30545
    Type of Medium: Online Resource
    ISSN: 0021-9258
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2004
    detail.hit.zdb_id: 2141744-1
    detail.hit.zdb_id: 1474604-9
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: bchm, Walter de Gruyter GmbH, Vol. 379, No. 10 ( 1998), p. 1279-1286
    Type of Medium: Online Resource
    ISSN: 1437-4315 , 1431-6730
    Language: English
    Publisher: Walter de Gruyter GmbH
    Publication Date: 1998
    detail.hit.zdb_id: 1466062-3
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2004
    In:  Cancer Research Vol. 64, No. 6 ( 2004-03-15), p. 2070-2075
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 64, No. 6 ( 2004-03-15), p. 2070-2075
    Abstract: One of the fundamental principles of pharmacology is that most drugs have side effects. Although considerable attention is paid to detrimental side effects, drugs can also have beneficial side effects. Given the time and expense of drug development, it would be particularly exciting if a systematic method could be applied to reveal all of the activities, including the unappreciated actions, of a potential drug. The present study takes the first step along this path. An activity-based proteomics strategy was used to simultaneously identify targets and screen for their inhibitors in prostate cancer. Orlistat, a Food and Drug Administration-approved drug used for treating obesity, was included in this screen. Surprisingly, we find a new molecular target and a potential new application for Orlistat. Orlistat is a novel inhibitor of the thioesterase domain of fatty acid synthase, an enzyme strongly linked to tumor progression. By virtue of its ability to inhibit fatty acid synthase, Orlistat halts tumor cell proliferation, induces tumor cell apoptosis, and inhibits the growth of PC-3 tumors in nude mice.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2004
    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 Research, American Association for Cancer Research (AACR), Vol. 75, No. 15_Supplement ( 2015-08-01), p. 255-255
    Abstract: Blocking DLL4, a Notch ligand, effectively inhibits tumor growth by increasing non-functional angiogenesis and decreasing the cancer stem cells (CSC) population. We are currently testing an anti-DLL4 antibody, demcizumab, in Phase1B trials in NSCLC, pancreatic, and ovarian cancer. DLL4 is also known to modulate immune responses. In the current study we examine the impact of anti-DLL4 on anti-tumor immune responses as a single agent and in combination with the key immune checkpoint inhibitor Programmed Cell Death Protein 1 (PD1). While the recent clinical success of PD1 inhibitors represents a new and promising cancer immunotherapeutic approach, high initial response rates are often associated by a lack of long-term, durable effects in a significant number of patients. Therefore, we hypothesized that dual blockade of DLL4 and PD1 might further impact tumor growth by further enhancing anti-tumor immune immunity. Our data demonstrates that dual blockade of DLL4 and PD1 using antibodies not only reduces tumor growth, but also led to tumor rejection in ∼50% in CT26WT tumor-bearing mice, similar to those treated with anti-PD1 alone (no tumor rejection was observed with anti-DLL4 alone). Anti-PD1 increased specific CD8+ T cell-mediated IFN-γ production while decreasing IL6. Anti-DLL4 treatment reduced IL17 production. Interestingly, only the dual blockage led to increased production of IL2 by splenocytes. Since IL2 is required for secondary population expansion of CD8+ memory T cells, increased IL2 in the combination group suggests potential for increased T cell activation, maintenance and memory T cell function, as compared to single agent anti-DLL4 and anti-PD1. While anti-PD1 reduced inhibition of CD4+ T cell proliferation by Tregs, the dual blockade significantly reduced Treg-mediated CD8+ T cell suppression. Furthermore, both effector and memory CD8+ T cell frequencies were increased within the total CD8+ T cell population. Interestingly, anti-PD1 decreased granulocytic MDSCs, while anti-DLL4 reduced monocytic MDSCs. Mice cured with single-agent anti-PD1 and anti-DLL4/anti-PD1 combination treatments were protected from series of re-challenge with tumor cells, suggesting the existence of immunologic memory. Interestingly, more mice were protected from tumor re-challenge when both DLL4 and PD1 were blocked, as compared to PD1 alone. Surprisingly, mice previously treated with the anti-DLL4/anti-PD1 combination produced more IL2, clearly indicating the role of DLL4 blockade in enhancing anti-tumor immunity. Therefore, these results show that dual targeting of DLL4 and PD1 may be an effective and durable cancer therapy by increasing anti-tumor immune response and promoting long-term immunological memory. Citation Format: Minu Srivastava, Christopher L. Murriel, Julie Roda, Hyun-Bae Jie, Fumiko Axelrod, Ming-Hong Xie, Rui Yun, Erin Mayes, Trevor Bentley, Belinda Cancilla, Raymond Tam, Tracy Tang, Ann Kapoun, John Lewicki, Tim Hoey, Austin Gurney, Angie Inkyung Park. Dual targeting of Delta-like ligand 4 (DLL4) and programmed death 1(PD1) inhibits tumor growth and generates enhanced long-term immunological memory. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 255. doi:10.1158/1538-7445.AM2015-255
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2015
    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 ...
  • 8
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 74, No. 19_Supplement ( 2014-10-01), p. 1764-1764
    Abstract: Deregulation of the canonical Wnt/beta-catenin signaling pathway has long been associated with cancer. Intracellular components of this pathway, including Axin, APC, and beta-catenin are frequently mutated in a range of human tumors. The identity of specific extracellular ligands that contribute to human cancer development through this signaling axis has remained unclear. Molecular characterization of the secreted beta-catenin signaling activities produced by minimally passaged human tumor xenograft models identified RSPO family members produced by multiple tumor types including ovarian, pancreatic, colon, breast and non-small cell lung cancer. In human tumor xenograft models that had RSPO overexpression, in some instances due to genomic translocation, anti-RSPO treatment markedly inhibited tumor growth. In addition, striking combination activity with standard of care chemotherapy agents resulted in regression of established tumors. These results highlight the potential for therapeutic intervention with this newly appreciated signaling axis. Citation Format: Austin Gurney, Fumiko Axelrod, Chris Bond, Jennifer Cain, Cecile Chartier, Marcus Fischer, May Ji, Chris Murriel, Janak Raval, Jalpa Shah, Min Wang, Wan-Ching Yen, Ann Kapoun, John Lewicki, Timothy Hoey. Inhibition of R-spondin (RSPO) signaling reduces the growth of multiple human tumors. [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 1764. doi:10.1158/1538-7445.AM2014-1764
    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 ...
  • 9
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 14_Supplement ( 2016-07-15), p. 2214-2214
    Abstract: GITRL (Glucocorticoid-Induced Tumor Necrosis Factor Receptor Ligand, TNFSF18) is a member of the TNF superfamily and naturally exists as a membrane-anchored type II protein that self assembles as a trimer. GITRL activates the co-stimulatory receptor GITR. GITR is found primarily on activated T effector (Teff) cells and regulatory T (Treg) cells. Co-stimulation of GITR by agonist agents is hypothesized to promote anti-tumor immunity by enhancing Teff cell activity and inhibiting Treg suppression. We generated a novel single-gene GITRL trimer fused to an immunoglobulin Fc domain (GITRL-Fc). GITRL-Fc activated GITR signaling more effectively than prototype GITR agonist antibody DTA-1. GITRL-Fc promoted a robust anti-tumor immune response in multiple syngeneic mouse tumor models. GITRL-Fc enhanced tumor specific T-cell responses, particularly of the Th1 type, and also led to reduction in Treg-mediated immunesuppressive activity. GITRL-Fc displayed single agent activity in inhibiting tumor growth and promoting complete tumor rejection in the murine CT26 colon carcinoma model and combination activity with anti-PDL1 as compared to anti-PDL1 and control IgG2a alone. Mice “cured” with GITRL or GITRL/anti-PDL1 combination treatments were protected from re-challenge with tumor cells, suggesting the existence of immunologic memory. More mice were protected from tumor re-challenge with the combination of GITRL-Fc and anti-PDL1, as compared to GITRL-Fc alone. Our results demonstrate that agonist GITRL-Fc induces potent T cell responses, overcomes Treg inhibition, and promotes anti-tumor activity in preclinical models as a single agent or in combination with anti PDL1. The mechanism of tumor eradication and induction of long-term immune memory response by the combination is under investigation and will be discussed at the presentation. Citation Format: Minu K. Srivastava, Rui Yun, Erin Mayes, Hyun_Bae Jie, Fumiko Axelrod, Jorge Monteon, Ming-Hong Xie, John Lewicki, Tim Hoey, Austin Gurney, Angie Inkyung Park. GITR ligand fusion protein (GITRL-Fc) induces T cell mediated anti-tumor immune response and can combine with anti-PDL1 to enhance anti-tumor immunity and long-term immune memory. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 2214.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    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 ...
  • 10
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2018
    In:  Cancer Research Vol. 78, No. 13_Supplement ( 2018-07-01), p. 70-70
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. 70-70
    Abstract: Mouse tumor models have been successfully used to generate preclinical data for numerous clinical programs including immunotherapy. Preclinical in vivo studies are typically carried out using young mice (often less than 2 months old) to generate efficacy data, predictive biomarker and pharmacodynamic markers. In notable contrast, the majority of human cancer occur in adult and older patients. It has become increasingly clear that the immune system of young and old mice is quite different with regards to the relative abundance and functionality of different cell populations. Data generated using young mice could provide a distorted assessment of the potential activity of immuno-oncology drugs in the clinic. Therefore, we tested the activity of GITRL-Fc protein in both young and old mice ( & gt;9 months). Previously using extensive young mice experiments, we have shown that GITRL-Fc promoted a robust antitumor immune response and enhanced tumor-specific T-cell responses, particularly of the Th1 type, and also led to a reduction in Treg-mediated immunosuppressive activity. Compared to young mice, tumors grew faster in older mice, and peripheral blood of older tumor-bearing mice has fewer T cells and NK cells. The total MDSC population was increased in the blood and spleen of old tumor-bearing mice, with a significantly higher number of G-MDSCs in the blood. On the other hand, old mice had reduced “antigen-presenting cells” (macrophages/dendritic cells expressing MHCII) in the blood. Furthermore, splenocytes from old mice had impaired production of IL-2. GITRL-Fc significantly inhibited tumor growth in both older and younger mice. However, efficacy was more pronounced in young mice, which frequently exhibited complete tumor regression. There were fewer tumor-infiltrating immune cells with less CD8 T and NK cells in older mice compared to young mice, consistent with faster tumor growth. Interestingly, GITR expression in CD8 T cells in old mice was lower compared with young mice at the tumor site. In old mice, GITRL-Fc (mIgG2a) was still able to deplete Tregs in tumor and increase Tregs in the spleen as has been previously shown with GITRL-Fc in young mice. On the other hand, GITRL-Fc deficient in effector function (mIgG2a (N297A)) did not deplete Tregs in the tumor but did retain some antitumor growth activity, indicating a role for GITR signaling in the mechanism of efficacy by GITLR-Fc. In conclusion, the results demonstrate the potential for the aging of the immune system to impact the efficacy observed with immunotherapy agents and highlight the potential benefits of conducting efficacy studies with both young and older mice. Citation Format: Angie Inkyung Park, Minu K. Srivastava, Rui Yun, Jenny Pokorny, Janice Yu, Fumiko Axelrod, Austin Gurney. Effect of aging on the antitumor activity of GITRL-Fc [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 70.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    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 ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...