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)  (21)
Material
Publisher
  • American Association for Cancer Research (AACR)  (21)
Language
Subjects(RVK)
  • 1
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 16_Supplement ( 2020-08-15), p. 3275-3275
    Abstract: Background: GLS-010 is a novel fully human anti-PD-1 mAb developed by the OMT transgenic rat platform. In Phase 1a study, GLS-010 exhibited good tolerance and 240mg (Q2W) was selected. Phase 1b study was conducted to evaluate the safety,anti-tumor activity and explore biomarkers of GLS-010 in pts with advanced solid tumors or lymphomas. Methods: All pts enrolled received GLS-010 240 mg every 2 weeks. Tumor response was assessed by RECIST 1.1 every 8 weeks. Adverse events (AEs) were graded by NCI CTCAE v4.03. Several biomarkers were explored, including PD-L1 by SP263 assay, tissue tumor mutation burden (tTMB) by whole exome sequencing (WES) from FFPE tissue, blood TMB (bTMB) by multi-gene panel based next-generation sequencing (NGS) from blood ctDNA. Results: As of 19 JUL 2019, 213 pts, median age of 55 (range: 21-75) years, were enrolled and 109 of 213 pts were still in treatment. The median dosing number was 7.5 (range: 1~41). Treatment-related AEs (TRAEs) occurred in 185 patients (70%), of which mostly were CTCAE grade 1-2. The most frequent TRAEs were related to hepatotoxicity, included “ALT increased” (32/213), “AST increased” (32/213), “blood bilirubin increased” (25/213). 53 of 163 pts, who received ≥1 response evaluation, achieved response (PR+CR, unconfirmed response included), including GC (4/21), EC (5/25), BTC (3/10), NSCLC (8/32), nasopharynx cancer (10/26),UC (2/8), HCC (0/12), cHL (18/21) and peripheral T/NK cell lymphoma (3/8). In all solid tumor types, pts with PD-L1-positive tumors experienced clinical benefit with significantly higher ORR (39.6% vs 14.5%, p=0.0025) and longer PFS (p=0.0241). ORR was higher for PD-L1 positive (TC≥25%) pts with lung cancer (66.7% vs 20.0%, p=0.051). For pan-caner analysis, ORR benefits of GLS-010 were also enhanced (51.9% versus 26.4%, p=0.0298) in pts with tTMB-high ( & gt;75th percentile, of each tumor type) versus tTMB-low. Pairing tTMB and survival data were analyzed in 129 pts, and tTMB-high pts benefit more with a significantly improved PFS (p=0.011). Also, improved PFS was observed in tTMB-high pts with EC (p=0.0059). For bTMB-high group, pts with EC achieved higher ORR (42.9% vs 6.3%, p=0.0672), which translated into PFS benefits (p=0.03). Conclusion: In conclusion, it is suggested that GLS-010 was well tolerated and had durable antitumor activity in Chinese tumor pts. PD-L1 has showed to be predictive of GLS-010 activity in solid tumors, especially in lung cancer. For pan-cancer analysis, it is preliminarily demonstrated that tTMB may be valuable biomarkers to predict the treatment response and benefit of GLS-010. For Chinese EC pts, tTMB and bTMB may be of value in predicting the response to PD-1 inhibitor. Citation Format: Lin Shen, Jifang Gong, Yuqin Song, Dingwei Ye, Zhihao Lu, Siyang Wang, Peijian Peng, Jianhua Chen, Ou Jiang, Guojun Zhang, Yuxian Bai, Jianji Pan, Chunguang Ma, Li Chen, Yi Ba, Qi Li, Ping Lu, Lingli Zhang, Xianli Yin, Shanzhi Gu, Huilai Zhang, Hang Su, Yongsheng Jiang, Bangwei Cao, Weiqing Han, Yan Sun, Feng Zhang, Weiwei Ouyang, Haiying Dong, Jianming Guo, Yabing Guo, Chongyuan Xu, Junyuan Qi, Li Wang, Jun Lv, Xiang Wang, Chris Chen, Jing Li, Yong Zheng, Ge Jin, Yining Yang, Guodong Zhao, Fan Yang, Kehui Xu, Xiangying Liang, Zhaoyang Pan, Haijin Meng. GLS-010, a novel fully human anti-PD-1 mAb in patients with advanced tumor: Preliminary results of a Phase Ib clinical trial [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 3275.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    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: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 25, No. 12 ( 2019-06-15), p. 3486-3494
    Abstract: No antiangiogenic treatment is yet approved for extrapancreatic neuroendocrine tumors (NET). Surufatinib (HMPL-012, previously named sulfatinib) is a small-molecule inhibitor targeting vascular endothelial growth factor receptors, fibroblast growth factor receptor 1 and colony-stimulating factor 1 receptor. We conducted a single-arm phase Ib/II study of surufatinib in advanced NETs. Patients and Methods: Patients with histologically well-differentiated, low or intermittent grade, inoperable or metastatic NETs were enrolled into a pancreatic or extrapancreatic NET cohort. Patients were treated with surufatinib 300 mg orally, once daily. The primary endpoints were safety and objective response rate (ORR) according to Response Evaluation Criteria in Solid Tumors (version 1.1). Results: Of the 81 patients enrolled, 42 had pancreatic NETs and 39 had extrapancreatic NETs. Most patients had radiologic progression within 1 year prior to enrollment (32 patients in each cohort). In the pancreatic and extrapancreatic NET cohorts, ORRs were 19% [95% confidence intervals (CI), 9–34] and 15% (95% CI, 6–31), disease control rates were 91% (95% CI, 77–97) and 92% (95% CI, 79–98), and median progression-free survival was 21.2 months (95% CI, 15.9–24.8) and 13.4 months (95% CI, 7.6–19.3), respectively. The most common grade ≥3 treatment-related adverse events were hypertension (33%), proteinuria (12%), hyperuricemia (10%), hypertriglyceridemia, and diarrhea (6% for each), and increased alanine aminotransferase (5%). Conclusions: Surufatinib showed encouraging antitumor activity and manageable toxicities in patients with advanced NETs. Two ongoing phase III studies, validating the efficacy of surufatinib in patients with NETs, will contribute to the clinical evidence.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 7_Supplement ( 2023-04-04), p. 5463-5463
    Abstract: Programmed death-ligand 1 (PD-L1) is a trans-membrane protein and co-inhibitory factor of the immune response. PD-L1 can combine with its receptor (PD-1) to reduce the proliferation of PD-1-positive cells and induce apoptosis. Therefore, PD-L1 was employed as a prognostic marker and a target for anti-cancer immunotherapy in blocking the PD-1 and PD-L1 checkpoints. PD-L1 is highly expressed in various malignancies, including head and neck squamous cell carcinoma (HNSCC). HNSCC is the sixth most common cancer worldwide, with high incidence and mortality rates in China. Although PD-L1 expression has been widely investigated, the PD-L1 expression status in Chinese HNSCC patients (pts) is largely unrevealed. The primary objective of this study was to determine the prevalence of PD-L1 expression with the Combined Positive Score (CPS) ≥20 in Chinese pts with recurrent or metastatic (R/M) HNSCC. The secondary objectives were to determine the prevalence of CPS≥1 in pts with R/M HNSCC and to study the difference in the demographic characteristics, clinicopathological parameters, treatment status, and other available biomarkers between the PD-L1 CPS≥20 group and the PD-L1 CPS & lt;20 group. This study was a multi-center retrospective analysis of data from six centers in China from August 9, 2021, to February 28, 2022. Pts with histologically/cytologically confirmed diagnoses of R/M HNSCC were included. PD-L1 expression was assessed by IHC using the 22C3 PharmDx assay (Agilent, Santa Clara, CA, USA) and was determined using a CPS. The Chi-square test, Fisher’s exact test, or Wilcoxon rank sum test would be used to compare the prevalence of these variables among pts between the PD-L1 CPS≥20 group and the PD-L1 CPS & lt;20 group. Out of 406 enrolled pts with R/M HNSCC, 402 testing pts were included in the final analysis. For all testing R/M HNSCC pts, 168 pts (41.8% [95% CI 36.92-46.78]) had PD-L1 expression with CPS ≥20. In addition, 337 pts (83.8% [95% CI 79.86-87.29] ) had PD-L1 expression (CPS≥1). For the testing pts, there were statistically significant differences in variables of gender (P & lt; 0.001), smoking habit (P = 0.0138 for non-smokers versus current smokers), and primary tumor site (P & lt; 0.001 for hypopharynx versus oral cavity and P = 0.0304 for larynx versus oral cavity) between the PD-L1 CPS≥20 group and the PD-L1 CPS & lt;20 group. In summary, in Chinese R/M HNSCC pts, most pts (83.8%) had PD-L1 expression, and two-fifths (41.8%) had PD-L1 expression with CPS ≥20. Prevalence of PD-L1 among Chinese pts with R/M HNSCC was consistent with that reported in the global KEYNOTE-048 study. PD-L1 expression was significantly associated with gender, smoking history and primary tumor site. Our findings of the variables related to the PD-L1 expression levels can supply adjuvant evidence for clinical practice and are a solid basis for future research on immunotherapy. Citation Format: Haizhen Lu, Dong Kuang, Lili Jiang, Jingping Yun, Qingxin Xia, Jian Wang, Pei Duan, Ping Zhou, Shengbing Zang, Yiping Jin, Xiangnan Jiang, Jielin Li, Wenmin Tang, Jiansong Zhou, Jihua Chen, Jianming Ying. The PD-L1 protein expression in Chinese patients with recurrent or metastatic head and neck squamous cell carcinoma: A multi-center retrospective study. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 5463.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    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: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 20, No. 1 ( 2021-01-01), p. 173-182
    Abstract: Although it is known that oncolytic viruses can inflame and recruit immune cells to otherwise immunosuppressed tumor microenvironments, the influence of the antiviral immune response on antitumor immunity is less clear across viral platforms and tumor types. CF33 is a recombinant orthopoxvirus backbone effective against colon cancer. We tested derivatives of CF33 with and without immune-checkpoint inhibition (anti–PD-L1) in mouse models of colon cancer. Results showed that the efficacy of CF33 backbone with J2R deletion (single-deleted) against colon cancer is not altered by additional deletion of F14.5L in vitro or in vivo. CF33 infection upregulated PD-L1 expression on tumor cells and led to an increased influx of lymphocytes and macrophages in tumors. Also, the levels of active CD8+ (IFNγ+) T cells in the virus-treated tumors were higher than those in control-treated tumors. Furthermore, a combination of CF33 derivatives with anti–PD-L1 resulted in durable tumor regression and long-term survival, resistant to tumor rechallenge. Analysis of immune cells from the treated mice showed that tumor-specific T cell activation occurred more robustly in tumors treated with the virus and that T cells were more strongly activated against the virus than against tumor, in an MHC-I–dependent manner. Our findings warrant further studies on the role of cross-priming of T cells against viral and tumor antigens, in the overall success of viroimmunotherapy.
    Type of Medium: Online Resource
    ISSN: 1535-7163 , 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2021
    detail.hit.zdb_id: 2062135-8
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2017
    In:  Cancer Research Vol. 77, No. 16 ( 2017-08-15), p. 4293-4304
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 16 ( 2017-08-15), p. 4293-4304
    Abstract: Tumor-initiating cells (TIC) represent cancer stem-like cell (CSC) subpopulations within tumors that are thought to give rise to recurrent cancer after therapy. Identifying key regulators of TIC/CSC maintenance is essential for the development of therapeutics designed to limit recurrence. The steroid receptor coactivator 3 (SRC-3) is overexpressed in a wide range of cancers, driving tumor initiation, cell proliferation, and metastasis. Here we report that SRC-3 supports the TIC/CSC state and induces an epithelial-to-mesenchymal transition (EMT) by driving expression of the master EMT regulators and stem cell markers. We also show that inhibition of SRC-3 and SRC-1 with SI-2, a second-generation SRC-3/SRC-1 small-molecule inhibitor, targets the CSC/TIC population both in vitro and in vivo. Collectively, these results identify SRC coactivators as regulators of stem-like capacity in cancer cells and that these coactivators can serve as potential therapeutic targets to prevent the recurrence of cancer. Cancer Res; 77(16); 4293–304. ©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 ...
  • 6
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2010
    In:  Cancer Research Vol. 70, No. 8_Supplement ( 2010-04-15), p. 2531-2531
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 70, No. 8_Supplement ( 2010-04-15), p. 2531-2531
    Abstract: Introduction: We previously reported the expression of CCR9 in pancreatic intraepithelial neoplasia (PanIN) and invasive pancreatic cancer tissues. Here, our objective was to investigate signaling downstream of CCR9 and to determine the anti-proliferative effects of the candidate therapeutic CCR9 antagonist CCX8037. Methods: PanIN and corresponding invasive pancreatic cancer (5143PDA) and liver metastasis (5143LM) cell lines were derived from Pdx-1-Cre;LSL-K-rasG12D and Pdx-1-Cre;LSL-K-rasG12D/p53R172H mice. In culture, murine and human pancreatic cancer cells (PANC-1) were exposed to CCL25, the specific CCR9 ligand. The phosphorylation of STAT3, an established regulator of pancreatic cancer proliferation, was measured by Western blot assay and proliferation assays were performed with the small molecule CCX8037 and the JAK/STAT inhibitor AG490. Results: First, we verified that exposure to CCL25 enhanced cell proliferation (20%-40%) in both murine and human pancreatic cancer cells. To investigate CCL25/CCR9 signaling, we exposed cells to CCL25 and observed increased phosphorylation of STAT3 in the cells. Blockade of STAT3 signaling with AG490 abrogated CCL25/CCR9-mediated proliferation in both murine and human pancreatic cancer cells. Pre-treatment of PanIN and pancreatic cancer cells with CCX8037 blocked CCL25-mediated phosphorylation of STAT3 and inhibited CCL25-mediated cell proliferation. Conclusion: Our study demonstrates for the first time that the CCL25/CCR9 axis activates downstream STAT3 signaling to mediate proliferation in murine and human PanIN and pancreatic cancer cells. Additionally, our data shows that the candidate therapeutic agent CCX8037 effectively blocks CCL25/CCR9-mediated proliferation in pancreatic cancer and warrants investigation in vivo. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 2531.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2010
    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. 83, No. 7_Supplement ( 2023-04-04), p. 3242-3242
    Abstract: Antibody-drug conjugates (ADCs) are effective anti-tumor therapies with high specificity, with little toxicity and side effects. Xenograft tumor models based on immunodeficient mice have been used for evaluating the human anti-tumor efficacy of ADCs. However, the antibody-mediated immune response of ADCs is not well evaluated in immunodeficient mice. To this end, we have developed BALB/c-hPD1 humanized mouse model to evaluate the efficacy of ADCs in combination with immunotherapy. Trastuzumab deruxtecan (T-DXd, DS-8201a) is a HER2-targeting ADC. In vitro efficacy studies showed that T-DXd effectively bound to the HER2-positive cell line SK-BR-3, resulting in strong endocytosis and induction of apoptosis. It has also been shown to kill HER2-negative MDA-MB-468 cells through a bystander effect both in vitro and in vivo. In vivo efficacy studies have demonstrated that ADCs can effectively inhibit human tumor cell growth in immunodeficient mice. In addition to human tumor cell lines, the efficacy of ADCs was also studied in BALB/c-hPD1mice engrafted with murine colon cancer cells model, and the results showed that T-DXd and Keytruda could significantly inhibit CT26-hHER2 tumor growth, with the tumor growth inhibition rates being 57% and 66%, respectively, Tumor growth inhibition rate of the combined treatment group was about 91%, which was superior compared to the efficacy of single drug treatment. The evaluation of Fc segment-mediated effect (such as ADCC, CDC and ADCP) of ADCs antibodies was carried out in humanized immune system mouse models, such as NCG-hIL15 and NCG-hSGM3. In summary, we have established a drug evaluation platform using various humanized mouse models to study the synergistic anti-tumor effects of ADCs combined with immune checkpoint blockade antibodies. Citation Format: Fang Zhu, Hongyan Sun, Qian Lu, Yuan Fang, Jianming Xu, Cunxiang Ju, Hongyu Wang, Santi Suryani Chen, Zhiying Li, Mark Wade Moore, Jing Zhao, Xiang Gao. Efficacy evaluation by novel humanized mouse models for preclinical study of ADCs combined with immunotherapy [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 3242.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    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
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2013
    In:  Cancer Research Vol. 73, No. 8_Supplement ( 2013-04-15), p. 4009-4009
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 73, No. 8_Supplement ( 2013-04-15), p. 4009-4009
    Abstract: Background: Expression of the chemokine receptor CCR9 is elevated in select solid organ cancers; and may contribute to drug resistance. However, there is no data for the role of CCR9 in GI cancers, aside from our prior work which demonstrated elevated expression in pancreatic cancer. Our current studies have uncovered activation of a novel CCR9-mediated molecular pathway and suggest that CCR9 may play a critical role in cancer progression through the aberrant activation of β-catenin. Methods: Immunohistochemistry and western blotting were used to assess CCR9 expression. We exposed human pancreatic cancer cell lines to CCL25 and evaluated levels of activated β-catenin and phosphorylated AKT via western blot. Proliferation and invasion were measured using an ATP-based assay and a Boyden chamber assay, respectively. IC50 of gemcitabine was determined using an acid phosphatase assay. Results: We have previously shown that CCR9 is expressed in human and murine PanIN lesions and in a human pancreatic cancer cell line. Here we expand upon these findings to show CCR9 expression in human pancreatic cancer tissue samples and in multiple cell lines. We determined that stimulation of CCR9 by its ligand, CCL25, signals through AKT and β-catenin to increase cell proliferation and invasion in pancreatic cancer cell lines. Following treatment with CCL25, cell proliferation was increased by an average of 20% across all cell lines (p & lt;0.01). Pre-treatment with the PI3K inhibitor, LY294002, inhibited the CCL25-mediated increase in proliferation. We further show a CCL25-mediated increase in invasion. Using a modified Boyden chamber matrigel invasion assay, we determined that CCL25 as a chemoattractant heightened the invasive abilities of our pancreatic cancer cell lines by 3-fold. Inhibiting PI3K activation blocked this increase. In addition, exposure to CCL25 led to reduced gemcitabine sensitivity. Pre-treatment with CCL25 increased the IC50 of gemcitabine by 7-fold in PANC-1 cells, from 0.1μM to 0.77μM (p=0.005) and 5.5-fold in AsPC-1 cells from 17.7μM to 96.7μM (p=0.008), indicating a drastic decrease in the effectiveness of gemcitabine when CCL25 is present. Conclusions: The current paucity of effective therapies for pancreatic cancer presents a significant clinical challenge. This highlights the need to identify and study mechanisms and pathways involved in pancreatic cancer progression. Determining the role of CC9R in pancreatic cancer and targeting the CCL25-CCR9-β-catenin pathway may therefore present an innovative, clinically significant therapeutic target for pancreatic cancer. Citation Format: Eileen L. Heinrich, Jianming Lu, Wendy Lee, Carrie Luu, Joseph Kim. CCL25-CCR9 axis mediates pancreatic cancer progression through beta-catenin activation. [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 4009. doi:10.1158/1538-7445.AM2013-4009
    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 ...
  • 9
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 15_Supplement ( 2015-08-01), p. 5382-5382
    Abstract: Lung cancer is the leading cause of cancer-related deaths worldwide and has a low 5-year survival rate. The current treatment for lung cancer patients is surgical resection followed by chemotherapy. However, the majority of patients will eventually experience disease progression and require further treatment. Increased evidence indicates that overexpression of survivin, the smallest member of IAP (inhibitor of apoptosis) family, results in poor prognosis, tumor recurrence, and drug resistance in various cancers, including non-small-cell lung cancer (NSCLC), which accounts for approximately 85% of all lung cancer cases. Although the precise mechanism leading to survivin overexpression in cancer cells is not fully understood, approach to effectively downregulate survivin is thought to be required to enhance chemotherapy, reduce relapse, and improve the survival of NSCLC patients. In the present study, we have explored the therapeutic potential of the class I HDAC inhibitor (HDACi) entinostat (or MS-275, SNDX-275, LC Laboratories, Woburn, MA) in combination with paclitaxel in the treatment of NSCLC. We show that entinostat significantly enhances paclitaxel-induced anti-proliferative/anti-survival effects and apoptosis in NSCLC cells. It appears that this HDACi selectively reduces the expression levels of survivin, but not the functionally-related molecules Mcl-1 and Bcl-xL. Further studies reveal that entinostat induces expression of two survivin-targeting miRNAs, miR-203 and miR-542-3p; and that both survivin-specific shRNAs and the mimics of miR-203 and/or miR-542-3p effectively downregulate survivin and dramatically promote NSCLC cells undergoing apoptosis upon paclitaxel treatment. Moreover, in a tumor xenografts model-established from NSCLC cells, entinostat maintains its capability to upregulate miR-203 and downregulate survivin in vivo. Importantly, the combinations of entinostat and paclitaxel exhibit a striking activity, as compared to either agent alone, to inhibit tumor growth. These data indicate that entinostat is efficacious to enhance paclitaxel-mediated anti-NSCLC activity. Our findings suggest that epigenetic targeting of survivin with entinostat or miRNA-replacement therapy may be a novel strategy to re-sensitize NSCLC to chemotherapy. Numerous studies demonstrate that entinostat exerts potent antitumor activity in various human cancers. Its clinic activity in NSCLC is currently being tested in combination with the DNA methyltransferase inhibitor Azacitidine or EGFR inhibitor erlotinib, but not conventional chemotherapy (http://www.clinicaltrial.gov/ct2/results?term = entinostat & Search = Search). Our studies provide a strong rationale to initiate clinical evaluation of the combinatorial effects of entinostat and paclitaxel on NSCLC patients. Citation Format: Shuiliang Wang, Ling Zhu, Zhiyong Zeng, Lianghu Huang, Fengjin Lin, Rong Lin, Jin Wang, Jun Lu, Qinghua Wang, Lingjing Lin, Huiyue Dong, Weizhen Wu, Kai Zheng, Jinquan Cai, Shunliang Yang, Yujie Ma, Shixin Ye, Wei Liu, Yinghao Yu, Bolin Liu, Jianming Tan. Epigenetic targeting of survivin enhances paclitaxel-mediated antitumor activity against non-small-cell lung cancer. [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 5382. doi:10.1158/1538-7445.AM2015-5382
    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 ...
  • 10
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2015
    In:  Cancer Research Vol. 75, No. 15_Supplement ( 2015-08-01), p. 780-780
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 15_Supplement ( 2015-08-01), p. 780-780
    Abstract: Introduction: MET overexpression has been shown to correlate with clinical outcomes in gastric cancer. The objective of this study was to evaluate MET expression and its downstream signaling mediators, STAT3, ERK, and AKT, as potential therapeutic targets in human gastric cancer. We evaluated cabozantinib, a multi-targeted receptor tyrosine kinase inhibitor with potent activity against MET and VEGFR2, in MET-expressing (MET(+)) gastric cancer cells. Methods: Surgical gastric cancer specimens were evaluated for expression of MET, phospho-ERK and phospho-STAT3 by immunohistochemistry (IHC). Cabozantinib was tested alone and in combination with a MEK inhibitor (U0126) and a JAK1/2 inhibitor (ruxolitinib). Half maximal inhibitory concentrations (IC50) for each drug were determined by MTS assay to determine appropriate treatment concentrations in two MET(+) human gastric cancer cell lines (Hs746T and SNU5). Protein phosphorylation levels of MET, STAT3, ERK, and AKT were determined by Western blot to assess downstream activation before and after drug treatment. Results: MET protein expression by IHC was detected in 18/47 (38.3%) gastric cancer specimens. One-half of MET(+) specimens stained with moderate or strong intensity, and 15/18 (83.3%) showed ≥ 25% membrane staining. In the MET(+) specimens, 9/18 (50.0%) samples expressed phospho-ERK and 3/18 (16.7%) expressed phospho-STAT3. Next, gastric cancer cell lines were examined by Western blot showing elevated levels of baseline phosphorylation of MET, STAT3 and ERK in Hs746T and SNU5 cells. Treatment of SNU5 (IC50 0.37 μM) and Hs746T (IC50 2.3 μM) with cabozantinib (0.1 μM) completely blocked downstream phosphorylation of MET, STAT3, AKT and ERK. Addition of MET's ligand hepatocyte growth factor (HGF, 25 ng/ml) overcame this observed inhibition at a lower cabozantinib concentration (0.01 μM), as evidenced by phosphorylation of STAT3 and ERK. Combining U0126 or ruxolitinib with cabozantinib (0.01 μM) effectively blocked ERK and STAT3 phosphorylation, respectively, even in the presence of HGF. This demonstrates that downstream signaling in MET(+) lines can be effectively and synergistically blocked by two targeted drug therapies. Conclusion: MET, ERK and STAT3 signaling are activated in gastric cancer cell lines, suggesting that ERK and STAT3 may serve as additional points of therapeutic intervention in MET(+) gastric cancer. Combining cabozantinib with a MEK or STAT3 inhibitor was necessary to inhibit phosphorylation of their respective targets in the presence of HGF, suggesting that drug synergy may be effective in blocking multiple activated downstream pathways. Citation Format: Audrey H. Choi, Jianming Lu, Sangjun Lee, Peiguo Chu, Vincent Chung, Ren-Jang Lin, Joseph Kim, Joseph Chao. Multi-targeted tyrosine kinase inhibitor Cabozantinib as a therapeutic agent in MET-overexpressing gastric cancer. [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 780. doi:10.1158/1538-7445.AM2015-780
    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 ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...