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)  (7)
  • Gibbons, Don L.  (7)
  • Tong, Pan  (7)
Material
Publisher
  • American Association for Cancer Research (AACR)  (7)
Language
Years
Subjects(RVK)
  • 1
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 22, No. 7 ( 2016-04-01), p. 1674-1686
    Abstract: Purpose: To identify new therapeutic targets for non–small cell lung cancer (NSCLC), we systematically searched two cancer cell line databases for sensitivity data on a broad range of drugs. We identified polo-like kinase 1 (PLK1) as the most promising target for further investigation based on a subset of sensitive NSCLC cell lines and inhibitors that were in advanced clinical development. Experimental Design: To identify potential biomarkers of response of NSCLC to PLK1 inhibition and mechanisms of PLK1 inhibitor–induced apoptosis, integrated analysis of gene and protein expression, gene mutations, and drug sensitivity was performed using three PLK1 inhibitors (volasertib, BI2536, and GSK461364) with a large panel of NSCLC cell lines. Results: The NSCLC cell lines had different sensitivities to PLK1 inhibition, with a minority demonstrating sensitivity to all three inhibitors. PLK1 inhibition led to G2–M arrest, but only treatment-sensitive cell lines underwent substantial apoptosis following PLK1 inhibition. NSCLC lines with high epithelial–mesenchymal transition (EMT) gene signature scores (mesenchymal cell lines) were more sensitive to PLK1 inhibition than epithelial lines (P & lt; 0.02). Likewise, proteomic profiling demonstrated that E-cadherin expression was higher in the resistant cell lines than in the sensitive ones (P & lt; 0.01). Induction of an epithelial phenotype by expression of the miRNA miR-200 increased cellular resistance to PLK1 inhibition. Also, KRAS mutation and alterations in the tight-junction, ErbB, and Rho signaling pathways correlated with drug response of NSCLC. Conclusions: In this first reported large-scale integrated analysis of PLK1 inhibitor sensitivity, we demonstrated that EMT leads to PLK1 inhibition sensitivity of NSCLC cells. Our findings have important clinical implications for mesenchymal NSCLC, a significant subtype of the disease that is associated with resistance to currently approved targeted therapies. Clin Cancer Res; 22(7); 1674–86. ©2015 AACR.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    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 Research Vol. 76, No. 7_Supplement ( 2016-04-01), p. A27-A27
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 7_Supplement ( 2016-04-01), p. A27-A27
    Abstract: Introduction: Lung cancer is the leading cause of cancer-related death, primarily due to distant metastatic disease. Metastatic cancer cells undergo an epithelial-to-mesenchymal transition (EMT) regulated by a double-negative feedback loop between the microRNA-200 (miR-200) family and Zeb1, but the precise mechanisms of Zeb1-dependent EMT in promoting malignancy remain largely undefined. While the cell-intrinsic effects of EMT are important for tumor progression, the reciprocal dynamic crosstalk between mesenchymal cancer cells and the extracellular matrix (ECM) is equally critical in regulating invasion and metastasis. This study investigates the collaborative effect of EMT and ECM in the metastatic process. Methods: Bioinformatic analysis of TCGA dataset was done to correlate ECM-associated gene expression with EMT gene signature scores. Western blotting and qPCR analysis of epithelial and mesenchymal lung cancer cell lines were performed to determine expression levels of collagen, LOX, and LOXL2. Lung tumor tissues from non-metastatic KrasG12D and metastatic KrasG12D;p53R172H mutant mice were analyzed by immunohistochemistry, Masson's trichrome staining, and second harmonics generation for collagen, LOX, and LOXL2 expression as well as collagen fiber organization. Syngeneic primary tumors generated by subcutaneous injection of murine lung cancer cell lines were analyzed in a similar fashion. Promoter and 3′-UTR luciferase reporter assays were performed to determine direct regulation LOXL2 and LOX by Zeb1 and miR-200, respectively. Results: Our results reveal increased collagen deposition in metastatic tumor tissues as a direct consequence of amplified collagen gene expression in Zeb1-activated mesenchymal lung cancer cells. Additionally, collagen fibers in metastatic lung tumors exhibit greater linearity and organization as a result of collagen crosslinking by the lysyl oxidase (LOX) family of enzymes. Expression of the LOX and LOXL2 isoforms is directly regulated by miR-200 and Zeb1, respectively, and their upregulation in metastatic tumors and mesenchymal cell lines is coordinated to that of collagen. Functionally, LOXL2, as opposed to LOX, is the principle isoform driving lung cancer metastasis by crosslinking and stabilizing insoluble collagen deposition in primary tumor tissues. Conclusions: Our study demonstrates that mesenchymal lung cancer cells metastasize by modulating the compositional and structural properties of the ECM through LOXL-mediated collagen crosslinking and deposition. We are the first to validate direct regulation of LOX and LOXL2 by the miR-200/Zeb1 axis, delineate collagen as a prognostic marker for lung cancer, and identify LOXL2 as a potential therapeutic target against tumor progression. Citation Format: David H. Peng, Pan Tong, Lauren A. Byers, Jing Wang, Chad J. Creighton, Don L. Gibbons. Zeb1 induces LOXL2-mediated collagen stabilization and deposition in the extracellular matrix to drive lung cancer invasion and metastasis. [abstract]. In: Proceedings of the AACR Special Conference on Tumor Metastasis; 2015 Nov 30-Dec 3; Austin, TX. Philadelphia (PA): AACR; Cancer Res 2016;76(7 Suppl):Abstract nr A27.
    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 ...
  • 3
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2019
    In:  Cancer Research Vol. 79, No. 13_Supplement ( 2019-07-01), p. 276-276
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 13_Supplement ( 2019-07-01), p. 276-276
    Abstract: Therapeutic resistance limits effective treatment of non-small cell lung cancer (NSCLC) and a better understanding of mechanisms contributing to resistance and strategies to overcome these are urgently needed. AXL, a TAM family receptor tyrosine kinase, has emerged as a key determinant of intrinsic and acquired resistance to chemotherapy, radiation and targeted therapies in NSCLC and other cancers, through its roles in mediating epithelial-mesenchymal transition (EMT) and immune escape. We previously showed that AXL may also play a role in DNA damage repair and that AXL overexpression mediated primary as well as acquired resistance to inhibitors of WEE1, a replication stress response kinase, in small cell lung cancer. In the present study, we further investigated the role of AXL in replication stress response. We found that, in NSCLC cell lines, AXL inhibition with the selective small-molecule AXL inhibitor BGB324 caused replication stress checkpoint activation, as indicated by increased RPA32 hyper-phosphorylation and ATR-mediated CHK1 phosphorylation. We further screened ATR inhibitors, VX-970 and AZD6738, in a panel of 20 NSCLC cell lines and correlated drug sensitivity with baseline expression of over 200 phosphorylated and total proteins, measured by reverse phase protein array. Notably, AXL was one of the biomarkers of resistance to the ATR inhibitor VX-970 (rho=0.52, p & lt;0.05). Therefore, we hypothesized that AXL plays a unique role in regulating replication stress and targeting AXL will sensitize NSCLC cells to ATR inhibitors. Combination of BGB324 and ATR inhibitors (VX-970 and AZD6738) significantly decreased cell proliferation in a panel of human and GEMM-derived NSCLC cell lines as compared to single agents alone. In NSCLC cells with primary resistance to ATR inhibition, co-targeting AXL and ATR significantly increased RPA32 hyper-phosphorylation, concomitantly with increased DNA double strand breaks and inactivated G2/M checkpoint, resulting in mitotic catastrophe. AXL knockdown in a GEMM-derived Kras/Trp53 mutant NSCLC model also showed similar results. Notably, NSCLC cell lines with low levels of SLFN11 (a DNA/RNA helicase that induces replication arrest following DNA damage independently of ATR) were more sensitive to AXL/ATR co-targeting. In conclusion, these findings suggest that AXL may play a novel and unexpected role in regulating replication stress. Furthermore, our results show that targeting AXL sensitizes NSCLC cell lines with primary resistance to ATR inhibitors and that AXL/ATR inhibitor combinations could be useful in treating platinum- and PARP inhibitor-resistant SLFN11-low tumors. Citation Format: Kavya Ramkumar, Pan Tong, You-Hong Fan, David Peng, John V. Heymach, Don L. Gibbons, Jing Wang, Lauren A. Byers. Targeting AXL sensitizes non-small cell lung cancer to ATR inhibitors by enhancing replication stress [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 276.
    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 ...
  • 4
    In: Cancer Discovery, American Association for Cancer Research (AACR), Vol. 8, No. 9 ( 2018-09-01), p. 1156-1175
    Abstract: Although treatment with immune checkpoint inhibitors provides promising benefit for patients with cancer, optimal use is encumbered by high resistance rates and requires a thorough understanding of resistance mechanisms. We observed that tumors treated with PD-1/PD-L1 blocking antibodies develop resistance through the upregulation of CD38, which is induced by all-trans retinoic acid and IFNβ in the tumor microenvironment. In vitro and in vivo studies demonstrate that CD38 inhibits CD8+ T-cell function via adenosine receptor signaling and that CD38 or adenosine receptor blockade are effective strategies to overcome the resistance. Large data sets of human tumors reveal expression of CD38 in a subset of tumors with high levels of basal or treatment-induced T-cell infiltration, where immune checkpoint therapies are thought to be most effective. These findings provide a novel mechanism of acquired resistance to immune checkpoint therapy and an opportunity to expand their efficacy in cancer treatment. Significance: CD38 is a major mechanism of acquired resistance to PD-1/PD-L1 blockade, causing CD8+ T-cell suppression. Coinhibition of CD38 and PD-L1 improves antitumor immune response. Biomarker assessment in patient cohorts suggests that a combination strategy is applicable to a large percentage of patients in whom PD-1/PD-L1 blockade is currently indicated. Cancer Discov; 8(9); 1156–75. ©2018 AACR. See related commentary by Mittal et al., p. 1066. This article is highlighted in the In This Issue feature, p. 1047
    Type of Medium: Online Resource
    ISSN: 2159-8274 , 2159-8290
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    detail.hit.zdb_id: 2607892-2
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Molecular Cancer Research, American Association for Cancer Research (AACR), Vol. 14, No. 11_Supplement ( 2016-11-01), p. PR10-PR10
    Abstract: Background: Small cell lung cancer (SCLC), the most lethal form of lung cancer, is characterized by early metastasis and rapid development of drug resistance. SCLC is characterized by ubiquitous loss of TP53 and RB1 and mutually exclusive amplification of the MYC oncogene (CMYC, MYCN, MYCL). The loss of TP53 is thought to render SCLC cells deficient at the G1-S cell cycle checkpoint, thus making these cells completely dependent on the G2-M checkpoint for DNA repair, mainly governed by CHK1 and WEE1. CHK1 is a vital serine-threonine kinase regulating the G2-M checkpoint upon DNA damage. The WEE1 kinase is a regulator of CDK1/2 and inhibition of WEE1 leads to aberrant DNA replication and premature mitosis. In this study we investigated the effect of CHK1 (LY2606368) and WEE1 (AZD1775) inhibitors as therapeutic targets for SCLC. Methods: Protein and gene expression was determined by immunoblot and real-time-RT-PCR respectively. Effect of the drugs on cell viability was determined by CellTiter-Glo. Efficacy of LY2606368 in H69 (chemosensitive) and H69/CR (chemoresistant) models was further determined by in vivo flank tumor regression analysis. The effect of LY2606368 monotherapy was also tested in a spontaneous SCLC genetically engineered mouse model (GEMM) model. The mechanistic studies were conducted by immunoblot analysis, apoptosis assay and flow cytometry. Pre and post-treatment cell lysates were analyzed by reverse phase protein array (RPPA) for biomarker exploration. Result: Higher expression of CHK1 and WEE1 was observed in SCLC patient tumor samples compared to normal lung (p & lt;0.001) and in SCLC cells as compared to NSCLC cells (p & lt;0.05). LY2606368 showed efficacy in both primary and acquired cisplatin resistant in vitro and in vivo models. LY2606368 treatment showed striking single agent activity in a spontaneous GEMM model of SCLC. Proteomic analysis identified an association between LY2606368 sensitivity and high levels of the CMYC protein. The level of CMYC decreased upon LY2606368 treatment (p & lt;0.001) while pro-apoptotic pathways increased (p & lt;0.05). CHK1 was a top marker of cisplatin sensitivity and treatment of cells with LY2606368 reduced activation of the MEK/MAPK pathway which we show as markers of de novo cisplatin resistance. LY2606368 treatment caused accumulation of cells in the sub-G1 phase and increased cell death. WEE1 inhibitor AZD1775 treatment decreased cell viability (IC50 & lt;100nM in 70% SCLC cells) in majority of SCLC cells. We identified AXL (receptor tyrosine kinase) and phospho-S6K as markers of AZD1775 resistance and treatment with AXL inhibitor, TP0903, resensitized the cells to AZD1775. Cell cycle analysis revealed increased apoptosis upon treatment of AZD1775 in combination with TP0903. Western blot analysis of pre and post AZD1775 treated samples revealed sustained activation of mTOR pathway in AZD1775 primary resistant lines. Pre-treatment of the cells with the mTOR inhibitor everolimus sensitized SCLC cells to AZD1775 by causing downregulation of AKT/mTOR pathway. Significance: The study establishes CHK1 and WEE1 as therapeutic targets in SCLC, a disease for which the standard of care has remained unchanged for & gt;30 years. Together, these results strongly indicate the potency of G2-M cell cycle checkpoint inhibitors as effective targeted therapies in both chemosensitive and chemoresistant patient population. The CHK1 inhibitor, LY2606368, and the WEE1 inhibitor, AZD1775, are both in current Phase 1 clinical trials. The correlative biomarker data from this study will not only indicate the subset of the patient population who are most likely to respond to these treatments, but it will also guide the selection of most effective combination strategies with CHK1 and WEE1 inhibition. This abstract is also being presented as Poster A25. Citation Format: Triparna Sen, Pan Tong, Catherine Allison Stewart, Sandra Cristea, You Hong-Fan, Bonnie S. Glisson, Don L. Gibbons, Julien Sage, Jing Wang, Lauren A. Byers. Exploiting the G2-M cell cycle checkpoint dependency in small cell lung cancer (SCLC) using pharmacological inhibitors of CHK1 and WEE1. [abstract]. In: Proceedings of the AACR Precision Medicine Series: Cancer Cell Cycle - Tumor Progression and Therapeutic Response; Feb 28-Mar 2, 2016; Orlando, FL. Philadelphia (PA): AACR; Mol Cancer Res 2016;14(11_Suppl):Abstract nr PR10.
    Type of Medium: Online Resource
    ISSN: 1541-7786 , 1557-3125
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    detail.hit.zdb_id: 2097884-4
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 14 ( 2017-07-15), p. 3870-3884
    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 ...
  • 7
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 22, No. 3 ( 2016-02-01), p. 609-620
    Abstract: Purpose: We previously demonstrated the association between epithelial-to-mesenchymal transition (EMT) and drug response in lung cancer using an EMT signature derived in cancer cell lines. Given the contribution of tumor microenvironments to EMT, we extended our investigation of EMT to patient tumors from 11 cancer types to develop a pan-cancer EMT signature. Experimental Design: Using the pan-cancer EMT signature, we conducted an integrated, global analysis of genomic and proteomic profiles associated with EMT across 1,934 tumors including breast, lung, colon, ovarian, and bladder cancers. Differences in outcome and in vitro drug response corresponding to expression of the pan-cancer EMT signature were also investigated. Results: Compared with the lung cancer EMT signature, the patient-derived, pan-cancer EMT signature encompasses a set of core EMT genes that correlate even more strongly with known EMT markers across diverse tumor types and identifies differences in drug sensitivity and global molecular alterations at the DNA, RNA, and protein levels. Among those changes associated with EMT, pathway analysis revealed a strong correlation between EMT and immune activation. Further supervised analysis demonstrated high expression of immune checkpoints and other druggable immune targets, such as PD1, PD-L1, CTLA4, OX40L, and PD-L2, in tumors with the most mesenchymal EMT scores. Elevated PD-L1 protein expression in mesenchymal tumors was confirmed by IHC in an independent lung cancer cohort. Conclusions: This new signature provides a novel, patient-based, histology-independent tool for the investigation of EMT and offers insights into potential novel therapeutic targets for mesenchymal tumors, independent of cancer type, including immune checkpoints. Clin Cancer Res; 22(3); 609–20. ©2015 AACR.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    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...