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: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 982-982
    Abstract: Current chemotherapy is effective in killing leukemic blasts in the periphery, but not leukemic stem cells (LSCs) in the bone marrow, which are thought to be responsible for the high relapse rate in leukemia. Thus, new therapies that effectively target LSCs are urgently needed to prevent cancer relapse. Accumulating evidence supports an essential role for adhesion pathways: particularly integrin beta 3 and its downstream focal adhesion kinase (FAK) in the maintenance of LSCs (Miller et al., Cancer Cell 2013; Despeaux et al., Stem Cells 2012). Previously, we have shown that FAK inhibitors preferentially target cancer stem cells in solid tumors (Shapiro et al., Sci Transl Med, 2014; Kolev et al., AACR 2013). We are extending our investigation of FAK inhibitors into hematological malignancies and report here that the FAK inhibitor VS-4718 displays anticancer activity in leukemia models both in vitro and in vivo. VS-4718 is a potent and orally bioavailable small molecule compound that targets cancer stem cells through inhibition of FAK kinase which is currently being tested in a Phase 1 clinical trial (NCT01849744). The anti-proliferative effect of VS-4718 was evaluated in a panel of 10 cell lines derived from patients with acute promyelocytic leukemia (APL), T-cell acute lymphocytic leukemia (T-ALL), B-cell acute lymphocytic leukemia (B-ALL), chronic myeloid leukemia (CML) and acute myeloid leukemia (AML) using a CellTiter-Glo cell viability assay on tissue culture or collagen coated plates. VS-4718 displayed anti-proliferative effects against most of these cell lines, with MV-4-11 AML being the most sensitive with an EC50 value of 200 nM. We further investigated the in vivo efficacy of VS-4718 against AML in both subcutaneous and disseminated models using the MV-4-11 cell line. Nude mice bearing MV-4-11 subcutaneous xenografts were treated orally twice daily with either vehicle control or VS-4718 for 14 days. 75 mg/kg VS-4718 caused 50% tumor growth delay and significantly extended median survival from 28 days to 48 days (p 〈 0.05). Moreover, tumor regression was observed in 4 out of 10 mice. We extended these observations to a disseminated MV-4-11 AML model to incorporate bone marrow stromal biology. When compared with vehicle control, VS-4718 dosed at 25 or 75 mg/kg, on a twice daily oral dosing schedule for 14 days, resulted in 40% and 76% increase in mouse life span, and significantly extended survival with p values 〈 0.05 and 〈 0.001 (log rank test), respectively. The effect of VS-4718 on leukemia stem cells and minimal residual disease (MRD) is currently under investigation. Taken together, results of our preclinical studies suggest that VS-4718 may have activity against leukemia that warrants further investigation. Disclosures Tam: Verastem: Employment. Ring:Verastem: Employment. Trombino:Verastem: Employment. Weaver:Verastem: Employment, Equity Ownership. Pachter:Verastem Inc.: Employment, Equity Ownership. Padval:Verastem: Employment. Xu:Verastem: Employment.
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 112, No. 11 ( 2008-11-16), p. 252-252
    Abstract: We have made the surprising discovery that agonism of adenosine A2A receptors has potent and synergistic antiproliferative effects in B-cell malignancies. Here we describe the preclinical evaluation of CRx-501, a potent selective adenosine A2A receptor agonist that synergizes with established anti-MM agents resulting in enhanced efficacy in pre-clinical models of MM. CRx-501 demonstrates potent single agent inhibition of proliferation in MM cell lines with EC50s ranging from 2–20 nM and maximum effects ranging from 20–75% inhibition of proliferation in the 10 MM cell lines surveyed. While CRx-501 demonstrates single agent effects, this molecule potently synergizes with glucocorticoids (dexamethasone and prednisolone), bortezomib, lenalidomide, melphalan and doxorubicin as well as emerging drug classes including HDAC inhibitors and HSP90 inhibitors. Substantial increases in overall effect levels and 2 to 100 fold potency shifts are observed with CRx-501 combinations in a broad panel of 10 MM cell lines including those both sensitive and resistant to current MM agents. In MM.1S cells, addition of 50 nM CRx-501 to 100 nM Dexamethasone results in 95% inhibition of proliferation as compared to approximately 60 and 75% inhibition with either CRx-501 or dexamethasone alone respectively. This combination results in a 10 fold shift in the dexamethasone IC50 and a combination index of 0.2 indicating high levels of synergy. Importantly, in cells resistant to dexamethasone up to the highest concentrations tested (2 microM; EJM, ANBL-6, MM.1R, KSM-12PE, MOLP-8), addition of CRx-501 can induce synergistic inhibition of proliferation converting resistance into sensitivity. Similarly, in combination with lenalidomide, CRx-501 results in a & gt;50 fold shift in IC50, an enhancement of efficacy from 50% to 90% inhibition of proliferation and activity against a lenolidamide resistant cell line (MOLP-8). Synergy is also observed with bortezomib resulting in a 2-fold shift in the IC50 of bortezomib in the presence of CRx-501. Importantly, CRx-501 is highly selective for B-cell malignancies and demonstrates no single agent activity or synergy in normal primary human cell types including peripheral blood mononuclear cells, umbilical vein endothelial cells, aortic smooth muscle cells, coronary artery endothelial cells and solid tumor cell lines at concentrations 2–3 orders of magnitude greater than the IC50 in MM cell lines. These antiproliferative effects occur through apoptosis. Treatment with CRx-501 in combination with dexamethasone causes a rapid and synergistic induction of Annexin V expression as compared to either single agent alone. Surprisingly, the potency and efficacy of CRx-501 is enhanced in the presence of 10 ng/mL interleukin-6 and HS-5 human bone marrow stromal cells. Concentrations of CRx-501 that are effective in vitro are achievable in mice with a 3 mg/kg s.c. dose resulting in a mean plasma AUC0–24 of 1228 ng*hr/mL. The synergistic antiproliferative effects of CRx-501 translate into xenograft models of MM with no significant body weight loss. Mice bearing subcutaneous H929 tumors show a 73% reduction in tumor volume after treatment with the combination of dexamethasone (1 mg/kg, s.c QD) and CRx-501 (3 mg/kg s.c. QD) for 34 days compared to a 36% or 37% decrease after treatment with either CRx-501or dexamethasone respectively. In summary, we report the preclinical evaluation of CRx-501, a potent and highly synergistic A2A agonist as a novel, selective, synergistic drug candidate for the treatment of MM. Our preclinical data provides compelling evidence in support of the further development of CRx-501 for use in multi-drug combination therapy of MM.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2008
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    American Society of Hematology ; 2006
    In:  Blood Vol. 108, No. 11 ( 2006-11-16), p. 1427-1427
    In: Blood, American Society of Hematology, Vol. 108, No. 11 ( 2006-11-16), p. 1427-1427
    Abstract: MOZ-TIF2 is expressed as a consequence of the chromosomal inversion inv(8)(p11q13) and is associated with AML FAB subtypes M4 and M5. Mice transplanted with MOZ-TIF2 succumb to monoclonal or oligoclonal leukemias with a long median disease latency, indicating that cooperating mutations are required for MOZ-TIF2 mediated leukemogenesis. The presence of a FLT3-ITD mutation in a patient with inv(8)(p11q13) has previously been reported, suggesting FLT3-ITD as a candidate cooperating mutation. Here we report that FLT3-ITD functionally cooperates with MOZ-TIF2 in co-transduction experiments in both a serial replating and murine bone marrow transplantation assay to induce AML that is transplantable to secondary recipients. At limit dilution, both MOZ-TIF2 and FLT3-ITD retroviruses are present, demonstrating cooperative effect. Moreover, tyrosine to phenylalanine mutations of FLT3-ITD residues 589 and 591, which we have previously reported to abrogate Stat5 signaling, also abolish the ability of FLT3-ITD to cooperate with MOZ-TIF2 both in vivo and in vitro. Furthermore, a constitutively active Stat5 mutant supports factor independent serial replating activity of MOZ-TIF2 in vitro. These data suggest a multi-step transformation model in which constitutive downstream Stat5 signaling by FLT3-ITD cooperates with MOZ-TIF2 in AML induction, and indicate that FLT3-ITD potentiates the properties of self-renewal in hematopoietic progenitors through activation of STAT5.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2006
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 114, No. 22 ( 2009-11-20), p. 3762-3762
    Abstract: Abstract 3762 Poster Board III-698 By using a high throughput combination screening strategy, we made the surprising discovery that adenosine A2A and beta-2 adrenergic receptor (b2AR) agonists have synergistic anti-proliferative activity in combination with dexamethasone, melphalan, lenalidomide, bortezomib and doxorubicin in preclinical multiple myeloma (MM) models. Both A2A and b2AR agonists are highly selective and demonstrate no single agent activity or synergy in normal cell types including human PBMCs, AoSMCs, HUVECs or HCAECs at concentrations 2-3 orders of magnitude greater than the IC50 in MM cell lines. To further examine the selectivity and breadth we have evaluated A2A and b2AR agonist combinations in a panel of 83 cell lines including solid tumor types and hematological malignancies. Single agents and combinations with dexamethasone and melphalan were systematically studied at multiple ratios of clinically relevant concentrations. Using a quantitative synergy score based on the Loewe model (Lehar et al. Nat Biotech 2009), we observe that combination activity for A2A or b2AR agonists is highly selective for hematologic malignancies with synergy observed most frequently in multiple myeloma and DLBCL cell lines. Synergy is also observed with the B-cell lines JM-1 (pre B-ALL) and GA-10 (Burkitt's lymphoma). Using a relative synergy cut-off (synergy score 〉 1), we find that 13 of the 18 MM cell lines tested demonstrate a synergistic interaction between the A2A agonist CGS-21680 and dexamethasone and 11 demonstrate a synergistic interaction between CGS-21680 and melphalan. Using this same measure, 9 of 18 MM cell lines demonstrate synergy with combinations of the b2AR agonist salmeterol with either dexamethasone or melphalan. Nine and ten of the cell lines in this MM panel are insensitive or respond weakly to dexamethasone and melphalan as single agents respectively. All cell lines were treated with the same concentrations of dexamethasone or melphalan, pointing to A2A agonists having a higher breadth of activity across the MM cell line panel. An interesting observation is the strong synergy observed for A2A or b2AR agonists with dexamethasone in the glucocorticoid-insensitive cell lines EJM and ANBL-6, which suggests that these agents may help restore steroid sensitivity in refractory patients. In general, drug resistance is a recurrent problem for cancer drugs and development of resistance after chronic exposure can reduce drug efficacy and promote refractory disease. We therefore examined the effects of chronic exposure to either A2A or b2AR agonists in the MM.1S cell line. Exposure of cells to CGS-21680 or salmeterol for one month reduced single agent sensitivity 〉 80%. Surprisingly, combinations of either agent with dexamethasone maintained similar amounts of synergy and cell killing as found with naïve untreated cells. As determined by Western blot analysis, the reduction in single agent activity after chronic exposure is not accompanied by a reduction in receptor levels. These data demonstrate that synergistic combinations of A2A and b2AR agonists are highly selective for B-cell malignancies and support the notion that synergistic drug combinations improve therapeutically relevant selectivity and circumvent drug resistance. This work further supports the rationalefor investigation of A2A and b2AR agonists in the treatment of B-cell malignancies and in particular, patients who have MM. Disclosures: Rickles: CombinatoRx, Inc.: Employment. Tam:CombinatoRx, Inc.: Employment. Necheva:CombinatoRx, Inc.: Employment. Giordano:CombinatoRx, Inc.: Employment. Borisy:CombinatoRx, Inc.: Employment. Lee:CombinatoRx, Inc.: Employment.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2009
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 124, No. 17 ( 2014-10-23), p. 2675-2686
    Abstract: Pyk2 plays a tumor-promoting role in MM progression via modulation of the Wnt/β-catenin signaling pathway. Pyk2 inhibitors represent a new therapeutic option against MM.
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    Elsevier BV ; 2001
    In:  Journal of Biological Chemistry Vol. 276, No. 11 ( 2001-03), p. 7701-7704
    In: Journal of Biological Chemistry, Elsevier BV, Vol. 276, No. 11 ( 2001-03), p. 7701-7704
    Type of Medium: Online Resource
    ISSN: 0021-9258
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2001
    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 ...
  • 7
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2013
    In:  Molecular Cancer Therapeutics Vol. 12, No. 11_Supplement ( 2013-11-01), p. C271-C271
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 12, No. 11_Supplement ( 2013-11-01), p. C271-C271
    Abstract: Focal adhesion kinase (FAK) is a non-receptor tyrosine kinase that orchestrates cell signaling through integrins and growth factor receptors and plays important roles in tumorigenesis. Amplification and overexpression of FAK have been observed in aggressive human cancers including ovarian and breast cancers. Defactinib is a potent, selective, and orally active FAK inhibitor with demonstrated tolerability and preliminary clinical activity as a single agent in a Phase 1 clinical trial. In an ongoing Phase 1/1b clinical trial (NCT01778803), we have demonstrated that defactinib can be combined with paclitaxel and have observed preliminary signs of clinical activity. We report here preclinical data supporting the clinical testing of defactinib in combination with paclitaxel in patients with ovarian cancer. In human ovarian cancer cell lines TOV-21G and OV-7, defactinib was found to enhance the efficacy of paclitaxel. Combination Index analyses demonstrated synergistic inhibition of tumor cell proliferation/survival with combination of defactinib and paclitaxel. We find that FAK inhibitors preferentially target breast and ovarian cancer stem cells (CSCs) relative to bulk tumor cells as evidenced by a diminished proportion of CSCs in multiple orthogonal CSC assays. In direct contrast, the standard-of-care cytotoxic agents, paclitaxel and carboplatin, increased the percentage of CSCs, indicating that these agents do not effectively target CSCs. Importantly, combination of FAK inhibitors with cytotoxic agents attenuated the enrichment of CSCs induced by the cytotoxic agents. We subsequently determined if defactinib reduces CSCs in tumor specimens from ovarian cancer patients who were previously treated with taxane and platinum. Ex vivo treatment of ovarian tumor tissue fragments with defactinib reduced the percentage of CSCs as assessed by CD24hi/CD117hi CSC markers and the Aldefluor assay. To assess drug effects on tumor-initiating capability, the ‘gold standard’ for CSC activity, TOV-21G cells were pretreated in vitro with the FAK inhibitor, paclitaxel or the combination of both agents and 1,000 of the resulting cells were injected into immunodeficient mice. The FAK inhibitor alone or in combination with paclitaxel prevented tumor initiation, while paclitaxel alone did not reduce tumor initiation, suggesting that treatment with a FAK inhibitor could effectively eliminate tumor initiating CSCs. In summary, our data indicate that defactinib preferentially targets cancer stem cells and enhances the activity of paclitaxel in preclinical models of ovarian cancer. These results provide additional support for the current clinical development of defactinib in combination with paclitaxel for the treatment of ovarian cancer. Citation Information: Mol Cancer Ther 2013;12(11 Suppl):C271. Citation Format: Christian M. Vidal, Vihren N. Kolev, Quentin G. Wright, Winnie F. Tam, David T. Weaver, Mahesh V. Padval, Qunli Xu, Jonathan A. Pachter. FAK inhibitor defactinib (VS-6063) enhances the efficacy of paclitaxel and preferentially targets ovarian cancer stem cells. [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 C271.
    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 ...
  • 8
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2019
    In:  Cancer Research Vol. 79, No. 13_Supplement ( 2019-07-01), p. 1963-1963
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 13_Supplement ( 2019-07-01), p. 1963-1963
    Abstract: One of the leading causes of non-responding tumors with existing immunotherapies is an immunosuppressive tumor micro-environment (TME). The suppressive nature of the TME is, in part, mediated by immunoregulatory cells (e.g. MDSC cells) and cytokines/chemokines. TGFB1 has recently been shown to be a key suppressive factor responsible for facilitating “cold” or immune-excluded tumors. In these tumors, T cell activity is restricted and by reducing TGFB1 levels we aim to support anti-tumor T cell activity. We evaluated if downregulation of TGFB1 with a self-delivering RNAi (sd-rxRNA) in MDSCs (in vitro) or TME (in vivo) promotes T cell activity and increase anti-tumor activity. sd-rxRNA is a chemically modified RNAi which incorporates drug like properties allowing for self-delivery of RNAi compounds to immune cells (including MDSC, DC, T and NK cells) in the absence of electroporation or formulation in a delivery vehicle. Tumor induced MDSCs were incubated with sd-rxRNA targeting TGFB1, its downstream target COX2 or in combination for 2 days. The cells were washed and added to a co-culture with OT-1 T cells and OVA expressing B16 melanoma. The anti-tumor activity of T-cells was monitored using a Real-Time Cell Activity assay (RTCA). TGFB1 and COX2 downregulated MDSCs had reduced immune-suppressive activity as shown by the enhanced B16-OVA tumor cell killing by CD8 OT-1 T cells in the RTCA assay while the combination of TGFB1 and COX2 MDSCs did not further improve the anti-tumor activity. To evaluate the effect of anti-TGFB1 sd-rxRNA in vivo, we first studied the tumor distribution and TGFB1 downregulation by intratumoral injection of sd-rxRNA to immunocompetent Balbc mice bearing 4T1-RL-GFP breast cancer in a fat pad model. DY547 labeled sd-rxRNA targeting PPIB or unlabeled sd-rxRNA targeting TGFB1 was injected into tumors. Two days later, tumors were resected, and fluorescence signal was analyzed by microscopy and TGFB1 downregulation was evaluated by RT-PCR. Efficient delivery of the DY547 labeled sd-rxRNA was observed in the vast majority of cells as shown by fluorescence microscopy. In addition, fluorescence signal could be detected in tumors up to 28 days after intratumoral injection, suggesting that sd-rxRNA was stable in tumors. Downregulation of TGFB1 in resected tumors, and an increased anti-tumor activity, was seen after three injections of sd-rxRNA. A follow up study was initiated investigating the efficacy of TGFB1 sd-rxRNA in combination with local tumor radiation on anti-tumor and metastatic activities in a 4T1 orthotopic breast cancer model. Preliminary data shows promising results. Our results suggest that sd-rxRNA compounds targeting TGFB1 can enhance the anti-tumor activity of T cells. Local injection of sd-rxRNA into tumors can effectively target TGFB1 in the TME resulting in less immunosuppression and increased anti-tumoral activity. Citation Format: Winnie F. Tam, Dingxue Yan, Melissa Maxwell, James Cardia, Gerrit Dispersyn. Feasibility and efficacy of using self-delivering RNAi against TGFB1 to reduce TME immunosuppression [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 1963.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    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: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 2101-2101
    Abstract: Background. Proline-rich tyrosine kinase (Pyk2) is a non-receptor tyrosine kinase which belongs to the focal adhesion kinase (FAK) family. It is known to facilitate the TNFα induced EMT process in solid tumors, but this has not been investigated in the field of hematologic malignancies. We therefore dissected the role of PyK2 in multiple myeloma (MM) by looking at its ability to modulate MM biology both in vitro and in vivo. Methods. Lentiviral packaged small hairpin RNA (shRNA), overexpression plasmid, related scrambled shRNA probe and empty vector were introduced into the MM1.S (GFP+/Luc+) cell line, to generate stable Pyk2 knock down (K.D.; #A2 and #A4), Pyk2 over-expression (Pyk2+), and control cells, respectively. The efficiency of K.D. or over-expression was validated by qPCR and immunoblotting. Cell viability and cell proliferation were detected by using CellTiter-Glo® luminescent assay and thymidine uptake, respectively. Gain- and loss-of function studies were also performed on MM cells in the presence of primary bone marrow stromal cells isolated from MM patients (MM-BMSCs). Adhesion of Pyk2 stable cells to fibronectin was measured by using an ECM cell adhesion assay kit. The synergistic effects of Pyk2 with Bortezomib were determined through calculating the DNA synthesis of Pyk2 K.D. cells treated with Bortezomib (2.5-5 µM), using Calcusyn software and Chou-Talalay method. Pyk2 K.D. stable cells were intravenously injected into SCID-Biege mice to generate a xenograft model. In vivo tumor growth was monitored by Bioluminescent Imaging. Pyk2-dependent-modulation of the Wnt/β-catenin pathway signaling was assessed using immunoblotting. The FAK/Pyk2 kinase inhibitor, VS-4718, was tested in vitro using both MM cell lines and primary bone marrow-derived MM cells; and in vivo using bioluminescence imaging. Results. Knockdown of Pyk2 in MM cells significantly repressed cell viability and proliferation, as well as their adhesive ability to BMSCs, compared to scrambled shRNA control cells. Moreover, Pyk2 knockdown induced de-adhesion of MM cells from BMSCs thus inducing chemosensitivity of tumor cells to Bortezomib. We next corroborated our findings by studying Pyk2+ MM cells, and showed that stably upregulated Pyk2 expression promoted MM cell growth as measured by either ATP quantitation or DNA synthesis. Upregulation of Pyk2 expression also stabilized the adhesion of MM cells to BMSCs, leading to drug resistance of MM cells to Bortezomib, compared with vector control cells. Pyk2-regulated tumor growth was further validated by establishing a xenograft mouse model. By using bioluminescence imaging, we found a significantly lower tumor burden in mice injected with Pyk2 K.D. cells, compared to control mice (injected with scrambled shRNA cells). We next dissected the effect of Pyk2 in modulation of cellular signaling in MM cells by using immunoblotting, and demonstrated that Pyk2 played an important role in regulating β-catenin signaling. Indeed, knockdown of Pyk2 induced GSK3β phosphorylation, leading to increased β-catenin phosphorylation, thus resulting in β-catenin degradation and inhibited translocation to the nucleus. Importantly, Pyk2 K.D. cells presented with reduced expression of c-myc and cyclin D1 at the protein level. Conversely, Pyk2 overexpression enhanced β-catenin expression together with c-myc and cyclin D1 up-regulation, thus confirming the role of Pyk2 in modulating Wnt/β-catenin signaling activity in MM. We finally determined that VS-4718, a potent FAK/Pyk2 kinase inhibitor currently in a phase I clinical study, induced apoptosis in MM cell lines and primary MM cells, and inhibited MM tumor growth in vivo. Conclusion. These findings indicate that Pyk2 exhibits pro-oncogenic properties in MM through modulation of Wnt/β-catenin signaling. The findings that the FAK/Pyk2 kinase inhibitor VS-4718 exerts anti-MM activity in cellular and in vivo models support MM as a potential clinical direction for this agent. Disclosures Ring: Verastem: Employment. Tam:Verastem: Employment. Xu:Verastem: Employment. Pachter:Verastem Inc.: Employment, Equity Ownership. Ghobrial:Onyx: Advisory board Other; BMS: Advisory board, Advisory board Other, Research Funding; Noxxon: Research Funding; Sanofi: Research Funding.
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    Online Resource
    Online Resource
    Informa UK Limited ; 2001
    In:  Molecular and Cellular Biology Vol. 21, No. 14 ( 2001-07-01), p. 4837-4846
    In: Molecular and Cellular Biology, Informa UK Limited, Vol. 21, No. 14 ( 2001-07-01), p. 4837-4846
    Type of Medium: Online Resource
    ISSN: 1098-5549
    Language: English
    Publisher: Informa UK Limited
    Publication Date: 2001
    detail.hit.zdb_id: 1474919-1
    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...