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)  (3)
  • 1
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 73, No. 8_Supplement ( 2013-04-15), p. 354-354
    Abstract: It is hypothesized that many genetic changes are required for the transformation process of sporadic malignant peripheral nerve sheath tumor (MPNST). Currently, Schwann cells and/or their precursor cells are believed to be the primary pathogenic cell source in peripheral nerve sheath tumor (PNST) formation and malignant progression. Recently, it has been shown that phosphatase and tensin homolog (PTEN) and epidermal growth factor receptor (EGFR) both play important roles in the initiation of peripheral nerve sheath tumors (PNSTs). In human MPNSTs, PTEN expression is often reduced, while EGFR expression is often induced. We tested if these two genes cooperate in the evolution of PNSTs, because genetically modified mouse models for each gene alone resulted in the formation of low-grade PNSTs. Transgenic mice were generated carrying conditional floxed alleles of Pten, EGFR was expressed under the control of the 2′,3′-cyclic nucleotide 3’ phosphodiesterase (Cnp) promoter and a desert hedgehog (Dhh) regulatory element driving Cre recombinase transgenic mice (Dhh-Cre). Transgenic mice with EGFR overexpression and Pten inactivated have an early postnatal lethality (median survival age of 26-days) and displayed various peripheral nervous system phenotypes. These mice had multiple enlarged dorsal root ganglia, with high incidence of enlarged brachial plexus and trigeminal nerves at various stages of PNST tumorigenesis. In vitro experiments using immortalized human Schwann cells demonstrated that loss of PTEN and overexpression of EGFR cooperate to increase cellular proliferation and anchorage-independent colony formation. Taken together, our data suggests that reduced PTEN expression, together with EGFR overexpression, can drive malignant progression of low-grade to high-grade PNSTs. Importantly, our novel mouse model recapitulates sporadic human MPNST and will be useful for testing therapies to prevent or reverse tumor progression. Citation Format: Vincent W. Keng, Adrienne L. Watson, Eric P. Rahrmann, Hua Li, Barbara R. Tschida, Branden S. Moriarity, Kwangmin Choi, Tilat A. Rizvi, Margaret H. Collins, Margaret R. Wallace, Nancy Ratner, David A. Largaespada. Conditional inactivation of Pten and overexpression of EGFR in Schwann cells results in early high-grade peripheral nerve sheath tumor development. [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 354. doi:10.1158/1538-7445.AM2013-354
    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 ...
  • 2
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 21 ( 2017-11-01), p. 5900-5912
    Abstract: Traditional treatments for breast cancer fail to address therapy-resistant cancer stem–like cells that have been characterized by changes in epigenetic regulators such as the lysine demethylase KDM4. Here, we describe an orally available, selective and potent KDM4 inhibitor (QC6352) with unique preclinical characteristics. To assess the antitumor properties of QC6352, we established a method to isolate and propagate breast cancer stem–like cells (BCSC) from individual triple-negative tumors resected from patients after neoadjuvant chemotherapy. Limiting-dilution orthotopic xenografts of these BCSCs regenerated original patient tumor histology and gene expression. QC6352 blocked BCSC proliferation, sphere formation, and xenograft tumor formation. QC6352 also abrogated expression of EGFR, which drives the growth of therapy-resistant triple-negative breast cancer cells. Our findings validate a unique BCSC culture system for drug screening and offer preclinical proof of concept for KDM4 inhibition as a new strategy to treat triple-negative breast cancer. Cancer Res; 77(21); 5900–12. ©2017 AACR.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2017
    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 ...
  • 3
    In: Cancer Discovery, American Association for Cancer Research (AACR), Vol. 3, No. 6 ( 2013-06-01), p. 674-689
    Abstract: Genetic changes required for the formation and progression of human Schwann cell tumors remain elusive. Using a Sleeping Beauty forward genetic screen, we identified several genes involved in canonical Wnt signaling as potential drivers of benign neurofibromas and malignant peripheral nerve sheath tumors (MPNSTs). In human neurofibromas and MPNSTs, activation of Wnt signaling increased with tumor grade and was associated with downregulation of β-catenin destruction complex members or overexpression of a ligand that potentiates Wnt signaling, R-spondin 2 (RSPO2). Induction of Wnt signaling was sufficient to induce transformed properties in immortalized human Schwann cells, and downregulation of this pathway was sufficient to reduce the tumorigenic phenotype of human MPNST cell lines. Small-molecule inhibition of Wnt signaling effectively reduced the viability of MPNST cell lines and synergistically induced apoptosis when combined with an mTOR inhibitor, RAD-001, suggesting that Wnt inhibition represents a novel target for therapeutic intervention in Schwann cell tumors. Significance: We show canonical Wnt/β-catenin signaling is a novel genetic driver of Schwann cell tumor development and progression, due to downregulation of β-catenin destruction complex members and overexpression of RSPO2. Inhibitors of Wnt signaling alone, or in combination with RAD-001, may have therapeutic value for patients with MPNSTs or neurofibromas. Cancer Discov; 3(6); 674–89. ©2013 AACR. See related commentary by Reilly, p. 610 This article is highlighted in the In This Issue feature, p. 591
    Type of Medium: Online Resource
    ISSN: 2159-8274 , 2159-8290
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2013
    detail.hit.zdb_id: 2607892-2
    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...