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)  (5)
Material
Publisher
  • American Association for Cancer Research (AACR)  (5)
Language
Years
Subjects(RVK)
  • 1
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 72, No. 5 ( 2012-03-01), p. 1092-1102
    Abstract: Chemokine CCL5/RANTES is highly expressed in cancer where it contributes to inflammation and malignant progression. In this study, we show that CCL5 plays a critical role in immune escape in colorectal cancer. We found that higher levels of CCL5 expression in human and murine colon tumor cells correlated with higher levels of apoptosis of CD8+ T cells and infiltration of T-regulatory cells (Treg). In mouse cells, RNA interference (RNAi)-mediated knockdown of CCL5 delayed tumor growth in immunocompetent syngeneic hosts but had no effect on tumor growth in immunodeficient hosts. Reduced tumor growth was correlated with a reduction in Treg infiltration and CD8+ T-cell apoptosis in tumors. Notably, we found that CCL5 enhanced the cytotoxicity of Treg against CD8+ T cells. We also found tumor growth to be diminished in mice lacking CCR5, a CCL5 receptor, where a similar decrease in both Treg cell infiltration and CD8+ T-cell apoptosis was noted. TGF-β signaling blockade diminished apoptosis of CD8+ T cells, implicating TGF-β as an effector of CCL5 action. In support of this concept, CCL5 failed to enhance the production of TGF-β by CCR5-deficient Treg or to enhance their cytotoxic effects against CD8+ T cells. CCR5 signaling blockade also diminished the in vivo suppressive capacity of Treg in inhibiting the antitumor responses of CD8+ T cells, in the same way as CCL5 signaling blockade. Together, our findings establish that CCL5/CCR5 signaling recruits Treg to tumors and enhances their ability to kill antitumor CD8+ T cells, thereby defining a novel mechanism of immune escape in colorectal cancer. Cancer Res; 72(5); 1092–102. ©2012 AACR.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2012
    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. 83, No. 7_Supplement ( 2023-04-04), p. 1225-1225
    Abstract: Epithelial-mesenchymal transition (EMT) has been considered the mechanism underlying distant metastasis in epithelial cancers. The hallmarks of the EMT phenomenon are mainly the loss of cell-cell adhesion, acquisition of a mesenchymal phenotype, gain of mobility and invasiveness, and attainment of enhanced tumorigenicity and anchorage-independent growth. After EMT, cancer cells can enter and survive in fluid-filled spaces and subsequently colonize distant sites. Despite a huge body of in vitro and in vivo evidence recognizing EMT as an important event in tumor progression, whether EMT exists in naturally-occurring experimental settings remains a contentious debate in the field of pathology. Lack of convincing histopathological evidence of EMT in tumor subtypes without spindle sarcomatous tumor components and lack of dynamic change demonstration of EMT are to blame. In this report, we devised a naturally-occurring experimental setting to prove the existence of EMT in tumors completely lacking histological evidence of EMT. Further, we demonstrated dynamic change of EMT in in vitro and in vivo settings. We performed primary culture 37H from a monophasic metaplastic squamous cell carcinoma tissue (MMSCC), which exhibited no histologic features reminiscent of EMT, to examine whether EMT exists in tumors without sarcomatous histology. Two dominant subtypes demonstrating different morphology were derived from parental 37H: polygonal epithelial-type (HE) and epithelioid mesenchymal-type (HM) cells. The HE cells were E-cadherin+vimentin- predominant; while the HM cells were E-cadherin-vimentin+ predominant, and functionally concomitant with increased migration and invasion and anchorage-independent tumorigenicity. Taking advantage of the inherited differences between the HE and HM cells, we performed head-to-head comparison by RNA-seq and subsequently identified key EMT transcription factors (EMT-TFs) ZEB1 and ZEB2 in this naturally-occurring experimental setting. In the primary MMSCC tissue where 37H was derived, the upregulation of ZEB1 and ZEB2 was also observed in vimentin+ carcinoma cells. Further, the dynamic change of EMT was demonstrated in the HE and HM cells in in vitro and in vivo settings. Spontaneous EMT was observed specifically in the ZEB1-or ZEB2-overexpressed polygonal epithelial-type cells; the knockdown of ZEB1 and ZEB2 reverted EMT in the HM cells; and reverse EMT was found a requirement for HM cells to grow in a xenograft mouse model. In conclusion, this report backed up the existence of EMT in naturally-occurring tumor without histologic evidence of EMT. The identification of underlying regulators contributing to spontaneous EMT, and the demonstration of EMT dynamic change were also included. Citation Format: Huang-Chun Lien, Hui-Chieh Yu, Tom Wei-Wu Chen, I-Chun Chen, Chiao Lo, Ling-Chun Yeh, Li-Ping Hsiao, Yen-Shen Lu. the existence, characteristics and underlying regulators of spontaneous epithelial-mesenchymal transition from a monophasic metaplastic squamous carcinoma patient-derived cancer cell model [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 1225.
    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 ...
  • 3
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2011
    In:  Cancer Research Vol. 71, No. 8_Supplement ( 2011-04-15), p. 2411-2411
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 71, No. 8_Supplement ( 2011-04-15), p. 2411-2411
    Abstract: Aberrant expression of hepatoma up-regulated protein, HURP, was found in hepatocellular carcinoma (HCC). Recent studies revealed that HURP is an essential factor during mitosis. However, the function of HURP in an in vivo context has not been well studied. In this study, we have concentrated our efforts on the pathological effects of Hurp deficiency during liver regeneration. We generated Hurp knockout (KO) mice to investigate the function of Hurp involved in hepatocyte cell cycle progression; we used the in vivo model of two-third partial-hepatectomy (PH) to induce synchronous cell cycle entry in liver. Our results revealed striking phenotypes of mitotic errors and chromosome instability in the Hurp KO livers. These phenotypes include impaired cell cycle progression as revealed by delayed entry of S-phase and M-phase of hepatocytes post-PH; mitotic infidelity as revealed by lagging chromosomes, multi-polar segregation and unequal segregation; aberrant nuclei of micro-nucleus, multi-nucleus and atypical nuclear morphology were frequently observed in the Hurp KO livers. Mechanistically, these abnormalities caused by Hurp deficiency did not turn on the spindle checkpoint machinery, such as BubR1 and Mad2, but up-regulated p21 expression to delay the cell cycle progression in liver. As an evidence of chronic liver damage, we observed continuously elevated serum ALT levels in Hurp KO mice post-PH. Together, these phenotypes in liver indicated that Hurp deficiency interferes spindle checkpoint (directly or indirectly), leading to mitotic infidelities, and possibly causes DNA breakage during chromatin segregation; this DNA breakage can further induce p21 to delay cell cycle during liver regeneration which is accompanying by chronic liver damages. All of these abnormalities may further lead to enhanced carcinogenesis in liver, which is currently under investigation. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 2411. doi:10.1158/1538-7445.AM2011-2411
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2011
    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
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2023
    In:  Cancer Research Vol. 83, No. 7_Supplement ( 2023-04-04), p. 2094-2094
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 7_Supplement ( 2023-04-04), p. 2094-2094
    Abstract: Realizing the full potential of precision oncology requires accurate prediction of treatment response. We previously developed a deep learning model, namely DeepDR, to predict drug sensitivity by integrating baseline profiles of mutations and gene expression of a cancer sample (Chiu et al.; DOI: 10.1186/s12920-018-0460-9). The model features a transfer learning design that captures two types of features: i) tumor relevant representations of mutation and expression data learned from tumor samples, and ii) predictive model for drug response (measured by IC50 values) learned from high-throughput drug screens of pan-cancer cell lines. DeepDR was applicable to both cell lines and tumors and achieved superior performance over conventional methods. To make the DeepDR model more accessible to biomedical researchers with limited programming skills, here we present a user-friendly platform implemented by using an R Shiny framework. The R Shiny app is a web interface that allows users to upload mutation and/or gene expression profiles of a cancer sample (cell or tumor model) and predict the sample’s response to 265 FDA-approved and investigational compounds covered by the screening library of the Genomics of Drug Sensitivity in Cancer (GDSC) project. The app provides an intuitive user interface to interactively visualize, search, and filter prediction results among 265 compounds. It also enables downstream analyses including statistical tests and provides links to external compound databases, such as PubChem. In the original publication of DeepDR, we have validated its performance across cell lines and tumors using well-known pharmacogenomic associations, such as estrogen receptor antagonist (tamoxifen) and EGFR-targeting drugs (afatinib and gefitinib), as well as a novel agent, CX-5461, in treating hematopoietic malignancies. Here we utilized the proposed Shiny app to further predict drug sensitivity of patients with hepatocellular carcinoma (HCC; n=356) of The Cancer Genome Atlas (TCGA). We investigated CTNNB1 as a demonstrating example for it is one of the most prevalent, yet undruggable, trunk mutations of HCC. A systematic search yielded nine compounds that were predicted to be significantly more effective in HCC tumors harboring CTNNB1 mutations (n=92) compared to others (n=264) (with one-tailed t-test P & lt; 0.0001). These top compounds target critical cancer pathways, such as cell apoptosis (p53 activator and Bcl-2 inhibitor), the PI3K/AKT and MAPK signaling pathways, and DNA-dependent protein kinases, suggesting candidates for further in vitro and in vivo investigations. In summary, the present R Shiny app is a user-friendly platform that enables in silico drug screening using deep learning with no requirement of programming experience. We expect the tool to foster accessibility of our deep learning prediction machine and facilitate the process of drug development in cancer. Citation Format: Li-Ju Wang, Michael Ning, Tapsya Nayak, Satdarshan P. Monga, Yufei Huang, Yidong Chen, Yu-Chiao Chiu. A user-friendly R Shiny app for predicting drug response of cancer using deep learning [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 2094.
    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 ...
  • 5
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 22, No. 15 ( 2016-08-01), p. 3950-3960
    Abstract: Purpose: FBW7 functions as a tumor suppressor by targeting oncoproteins for destruction. We previously reported that the oncogenic mutation of KRAS inhibits the tumor suppressor FBW7 via the Ras–Raf–MEK–ERK pathway, which facilitates the proliferation and survival of pancreatic cancer cells. However, the underlying mechanism by which FBW7 suppresses pancreatic cancer remains unexplored. Here, we sought to elucidate the function of FBW7 in pancreatic cancer glucose metabolism and malignancy. Experimental Design: Combining maximum standardized uptake value (SUVmax), which was obtained preoperatively via a PET/CT scan, with immunohistochemistry staining, we analyzed the correlation between SUVmax and FBW7 expression in pancreatic cancer tissues. The impact of FBW7 on glucose metabolism was further validated in vitro and in vivo. Finally, gene expression profiling was performed to identify core signaling pathways. Results: The expression level of FBW7 was negatively associated with SUVmax in pancreatic cancer patients. FBW7 significantly suppressed glucose metabolism in pancreatic cancer cells in vitro. Using a xenograft model, MicroPET/CT imaging results indicated that FBW7 substantially decreased 18F-fluorodeoxyglucose (18F-FDG) uptake in xenograft tumors. Gene expression profiling data revealed that TXNIP, a negative regulator of metabolic transformation, was a downstream target of FBW7. Mechanistically, we demonstrated that TXNIP was a c-Myc target gene and that FBW7 regulated TXNIP expression in a c-Myc–dependent manner. Conclusions: Our results thus reveal that FBW7 serves as a negative regulator of glucose metabolism through regulation of the c-Myc/TXNIP axis in pancreatic cancer. Clin Cancer Res; 22(15); 3950–60. ©2016 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...