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
Filter
  • American Association for Cancer Research (AACR)  (2)
Material
Publisher
  • American Association for Cancer Research (AACR)  (2)
Language
Years
Subjects(RVK)
  • 1
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 12, No. 11_Supplement ( 2013-11-01), p. C1-C1
    Abstract: Background: The solid tumor microvasculature is characterized by structural immaturity and functional abnormality, and mediates several deleterious aspects of tumor behavior. The vascular endothelial protein tyrosine phosphatase (VE-PTP) attenuates the activity of the endothelial cell (EC) Tie-2 receptor tyrosine kinase, a key mediator of vessel maturation. Here we determine the role of VE-PTP in the vasculature of primary and metastatic mammary carcinomas. Methods: AKB-9778 is a first-in-class pharmacologic VE-PTP inhibitor. We systematically examined its effects in vitro and in vivo. First we studied the effects of AKB-9778 on Tie-2 signaling in two endothelial cell lines in vitro, and also measured its impact on Tie-2 activation in normal and tumor ECs in mice. We next used embryonic zebrafish assays to determine the effects of AKB-9778 on embryonic angiogenesis. We also studied the impact of AKB-9778 therapy on the tumor vasculature, tumor growth and metastatic progression using orthotopic models of murine mammary carcinoma as well as spontaneous and experimental metastasis models (4T1, E0771, P0008, MMTV-PyMT). Finally, we used endothelial nitric oxide synthase (eNOS) deficient mice to establish the role of eNOS in mediating the effects of VE-PTP inhibition. Results: AKB-9778 induced ligand-independent Tie-2 activation in ECs in vitro and in vivo, and impaired embryonic zebrafish angiogenesis. In mouse models of breast cancer, AKB-9778 (i) delayed the early phase of mammary tumor growth by maintaining vascular maturity; (ii) slowed the growth of spontaneous micrometastases by preventing extravasation of tumor cells into distant organ parenchyma (prolonging survival above adjuvant chemotherapy alone); and (iii) matured established primary tumor blood vessels (increased pericyte coverage, reduced permeability) in turn enhancing tumor perfusion, reducing hypoxia, and improving radiation response. Experiments using eNOS knockout mice showed that the effects of AKB-9778 on tumor vessels were mediated in part by eNOS activation. Conclusions: Phosphatase inhibition is a relatively unexplored field in cancer, and this is to our knowledge the first demonstration of the effects of VE-PTP inhibition in any disease. Our results demonstrate that pharmacological VE-PTP inhibition can normalize the structure and function of tumor vessels through Tie-2 activation, which delays tumor growth, slows metastatic progression, and enhances response to concomitant cytotoxic treatments. Furthermore we provide genetic evidence for the causal role of eNOS in mediating the beneficial effects of VEPTP inhibition in established tumors. By using models of adjuvant therapy (combined with chemotherapy) and radiation treatment, our results indicate possible avenues for further studying the clinical translatability of VE-PTP inhibition in cancer. Citation Information: Mol Cancer Ther 2013;12(11 Suppl):C1. Citation Format: Shom Goel, Nisha Gupta, Brian P. Walcott, Matija Snuderl, Cristina T. Kesler, Benjamin J. Vakoc, Randall T. Peterson, Timothy P. Padera, Dan G. Duda, Dai Fukumura, Rakesh K. Jain. Functional normalization of the breast cancer vasculature through activation of Tie2 using a vascular-endothelial protein tyrosine phosphatase inhibitor. [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2013 Oct 19-23; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2013;12(11 Suppl):Abstract nr C1.
    Type of Medium: Online Resource
    ISSN: 1535-7163 , 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2013
    detail.hit.zdb_id: 2062135-8
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 73, No. 8_Supplement ( 2013-04-15), p. LB-338-LB-338
    Abstract: CD36 is an endothelial cell (EC) receptor that mediates angiostatic activity and oxidative stress, which are important in ischemic disease and tumor progression. CD36 interacts with TSR domain proteins including thrombospondin-1. This results in a signal that is likely to initiate an antiangiogenic “switch," subsequently converting a growth factor-mediated proangiogenic response to an antiangiogenic response. However, in pathologic settings including malignant tumors, robust angiogenesis occurs despite the abundance of TSR-containing proteins in the microenvironment. This suggests that there is a mechanism by which TSR-mediated antiangiogenesis could be blunted via the localized downregulation of EC CD36 transcription. We previously reported that lysophosphatidic acid (LPA), a bioactive signaling phospholipid mediator, down-regulates microvascular EC (MVEC) CD36 transcription by protein kinase PKD-1 signaling via G protein coupled receptor LPA1,3. However, further elucidating the CD36 transcriptional mechanisms is critical to understanding the antiangiogenic switch. To this end, we recently discovered that obesity-derived LPA inhibited CD36 expression in tumor-associated ECs (TAECs). Similarly, inducing the constitutively active PKD-1 or PKD-CA expression also resulted in CD36 downregulation. Mechanistically, LPA treatment led to nuclear accumulation of PKD-1, HDAC7 and FoxO1. Furthermore, LPA-mediated PKD-1 signaling enhanced FoxO1-HDAC7 interaction in the nucleus. This interaction was attenuated with PKD-1 silencing. Intriguingly, doxycycline induced PKD-CA expression increased ephrin B2 expression and ERK activation, which are two critical “molecular signatures” involved in arteriogenesis. Functionally, FGF-2 induced a potent proangiogenic response even in the presence of TSP-1 in ECs pre-treated with LPA. Doxycycline induced PKD-CA expression stimulated robust arterial EC branching in a 3D spheroid assay. This angiogenic response was inhibited by a novel PKD-1 specific inhibitor. Moreover, in a mouse Lewis lung carcinoma model, we detected a minimal level of endothelial CD36 expression with robust tumor angiogenesis. The role of PKD-1 signaling in CD36 transcriptional repression and tumor arteriogenesis is a critical yet underexplored and underappreciated field. We propose that the LPA-PKD-FoxO1 signaling axis is essential for CD36 transcriptional repression and arteriogenic signaling and responses. The PKD-1 stimulated nuclear signaling network is critical to epigenetic suppression of CD36 transcription and silencing of CD36 antiangiogenic switch, subsequently turning on the angiogenic switch and stimulating the arteriogenic responses. Targeting the LPA-PKD1-FoxO1 signaling axis could have therapeutic potential in the ischemic disease and malignant tumors. Citation Format: Jacob D. Kohlenberg, Yiliang Chen, Brad Best, Peter Storz, Randall T. Peterson, Roy Silverstein, Bin Ren. A novel LPA-PKD1-FoxO1 pathway in endothelial cells provides an angiogenic switch via down-regulation of CD36 transcription and induction of arteriogenic responses . [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr LB-338. doi:10.1158/1538-7445.AM2013-LB-338
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2013
    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...