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)  (11)
  • 1
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 7_Supplement ( 2023-04-04), p. 3886-3886
    Abstract: Background:The response of 1st-line EGFR-TKI treatment in advanced EGFR-mutated lung adenocarcinoma patients are highly effective, but its sustainability and clinical course are quite variable from patient to patient. In this study, we investigated the clinical impact of concomitant genetic mutations analyzed by targeted NGS on PFS and acquired resistance in advanced EGFR-mutated lung adenocarcinoma patients. Methods: Eighty-five advanced NSCLC patients harboring EGFR mutations were enrolled prospectively in multi-centers from 2019 to 2022 (NCT04122833). We performed the targeted next generation sequencing on 324 cancer-related genes by Foundation One CDx with pre-treated tumor samples. First- or second-generation EGFR-TKIs(gefitinib, afatinib, or erlotinib) were administered in 1st-line setting. After the progression, tissue re-biopsy or plasma liquid biopsy (FoundationOne Liquid CDx) for NGS if tissue biopsy is difficult or risk was performed. Results: Of the 85 patients (70.6% of female, 65.8% of nonsmoker, 56.4% of E19del and 38.8% of E21 L858R mutation), 50 patients experienced a disease progression in November, 2022. The median PFS was 20 months (95 %CI: 15.2-24.8). The most frequent co-mutations were TP53 (47.1%), CDKN2A/B loss (34.1%), MTAP loss (20%), NKX2-1amp(15.3%), MDM2amp (14.1%), RMB10, CCNE/CCND1 amp, NFKB1 amp (11.8%) and CDK4/6 amp (10.6%). Patients with TP53, CDK4/6 amp and MYC amp were independently associated with shorter PFS. In a multivariate analysis, tumors with copy number alterations such as CDK4/6 amp or Myc amp were also independently associated with shorter PFS. However, the CDKN2A/2B loss, MTAP loss, and MDM amp were not related with the PFS. In the number of co-mutations, patients harboring ≥5 co-mutations identified by NGS had shorter median PFS than patients with 0-1 or 2-4 co-mutations. (mPFS 0-1: 2-4: ≥5 co-mutations=35: 18: 9.3 months, p & lt;0.001). At progression, 22 patients harbored an acquired T790M mutation (25.8%). Before TKI treatment, patients with CDKN2A/B loss, MTP loss or CCND/CCNE1 amp in pretreatment tumor have more acquired T790M mutation after progression significantly (p & lt;0.05) Conclusion: We have demonstrated that concomitant mutations detected by targeted NGS analysis provide significant impact on the drug response and clinical course of advanced EGFR-mutated adenocarcinoma patients treated by 1st-line EGFR-TKIs. It is suggested that targeted NGS along with PCR-based detection will be necessary for precision medicine-based individualized practice of 1st-line EGFR-TKI-based combination treatment. Citation Format: In Ae Kim, Seung Joon Kim, Sung Yong Lee, Chang Min Choi, Jae Cheol Lee, Tae Won Jang, Seung Hun Jang, Chan Kwon Park, Wan Seop Kim, Jae Young Hur, Hee Joung Kim, Young Whan Kim, Key Young Lee. Analysis of concomitant genetic alterations in advanced EGFR-mutated lung adenocarcinoma by targeted NGS: A multicenter prospective and real world 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 3886.
    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 ...
  • 2
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 21, No. 11 ( 2015-06-01), p. 2613-2623
    Abstract: Purpose: To better understand the complete genomic architecture of lung adenocarcinoma. Experimental Design: We used array experiments to determine copy number variations and sequenced the complete exomes of the 247 lung adenocarcinoma tumor samples along with matched normal cells obtained from the same patients. Fully annotated clinical data were also available, providing an unprecedented opportunity to assess the impact of genomic alterations on clinical outcomes. Results: We discovered that genomic alternations in the RB pathway are associated with significantly shorter disease-free survival in early-stage lung adenocarcinoma patients. This association was also observed in our independent validation cohort. The current treatment guidelines for early-stage lung adenocarcinoma patients recommend follow-up without adjuvant therapy after complete resection, except for high-risk patients. However, our findings raise the interesting possibility that additional clinical interventions might provide medical benefits to early-stage lung adenocarcinoma patients with genomic alterations in the RB pathway. When examining the association between genomic mutation and histologic subtype, we uncovered the characteristic genomic signatures of various histologic subtypes. Notably, the solid and the micropapillary subtypes demonstrated great diversity in the mutated genes, while the mucinous subtype exhibited the most unique landscape. This suggests that a more tailored therapeutic approach should be used to treat patients with lung adenocarcinoma. Conclusions: Our analysis of the genomic and clinical data for 247 lung adenocarcinomas should help provide a more comprehensive genomic portrait of lung adenocarcinoma, define molecular signatures of lung adenocarcinoma subtypes, and lead to the discovery of useful prognostic markers that could be used in personalized treatments for early-stage lung adenocarcinoma patients. Clin Cancer Res; 21(11); 2613–23. ©2014 AACR. See related commentary by Collisson, p. 2418
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2015
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2019
    In:  Molecular Cancer Therapeutics Vol. 18, No. 12_Supplement ( 2019-12-01), p. C058-C058
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 18, No. 12_Supplement ( 2019-12-01), p. C058-C058
    Abstract: Vitamin C (L-Ascorbic acid, VC), an anti-oxidant has been studied as the possible potent chemotherapeutic drug in various cancers including colorectal cancer, breast cancer, ovarian cancer, prostate cancer and lung cancer. In particular, it is recently reported that VC kills efficiently colorectal cancer cells with KRAS or BRAF mutations. Give fact that pancreatic cancer has high frequency of KRAS mutation rate and is one of the most aggressive and lethal cancers with lack of effective therapy targeting the KRAS mutations, we would like to investigate whether VC effectively kills pancreatic cancer cells with various types of KRAS mutation. We first tested cytotoxicity and cell growth inhibition effect of VC on pancreatic cancer cell lines. In growth inhibition and colony formation assay, VC showed differential effect on cell growth and colony formation according to the KRAS genotypes. Treatment of the pancreatic cancer cell lines harboring KRAS G12D mutation with VC resulted in marked decrease of cell survival and colony formation, while same treatment of the cell lines with other KRAS mutations or wild type had no growth inhibitory effect. Interestingly, although there was no dramatic difference in well-known VC transporters, Glut1 expression according to the KRAS genotypes, sodium-dependent vitamin c transporter 2 (SVCT2) expression was relatively lower in KRAS G12D-mutant pancreatic cancer cells than other cells. To study possible mechanisms of VC-induced growth inhibition and cell death, we also tested whether glycolysis was inhibited or not though extracellular acidification rate (ECAR) measurement. After 1mM VC injection, ECAR decreased dramatically in KRAS G12D-mutant pancreatic cancer cells compared to the other genotypes. In cell cycle phospho-antibody array analysis, the results indicated that VC significantly increased the expression of DNA damage-associated proteins. Finally, we also found that cell death measured by apoptosis increased in KRAS G12D-mutant pancreatic cancer cells with VC through annexin V-PI staining FACS analysis. In conclusion, we showed VC selectively induces the apoptosis and cell growth inhibition in KRAS G12D-mutant pancreatic cancer cells than other KRAS mutations by inhibition of glycolysis and induction of DNA damage-associated proteins. Our findings suggest that VC is needed to further investigate as the potential novel agent with cytotoxic chemotherapies for the treatment of pancreatic cancer. Citation Format: Hye-Lim Jang, Jung Yong Hong, Seung Tae Kim, Jeeyun Lee, Se Hoon Park, Young Suk Park, Ho Yeong Lim, Won Ki Kang, Joon Oh Park. Vitamin C preferentially induces growth inhibition and cell death in KRAS G12D-mutant pancreatic cancer cells [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference on Molecular Targets and Cancer Therapeutics; 2019 Oct 26-30; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2019;18(12 Suppl):Abstract nr C058. doi:10.1158/1535-7163.TARG-19-C058
    Type of Medium: Online Resource
    ISSN: 1535-7163 , 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    detail.hit.zdb_id: 2062135-8
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 15, No. 18 ( 2009-09-15), p. 5861-5868
    Abstract: Purpose: This study aimed to investigate whether metabolic tumor volume (MTV) measured from [18F]-fluorodeoxyglucose positron emission tomography/computed tomography (FDG-PET/CT) predicts short-term outcome to radiotherapy with or without chemotherapy and disease-free survival (DFS) in patients with pharyngeal cancers. Experimental Design: The MTVs of primary sites with or without neck nodes were measured in 82 patients. Short-term outcome was assessed using the treatment response evaluation by the Response Evaluation Criteria in Solid Tumors and recurrence events during follow-up (complete response/no recurrence or residual disease/recurrence). Results: A total of 64 patients had complete response/no recurrence as of the last follow-up. A cutoff of 40 mL for the MTV was the best discriminative value for predicting treatment response. By univariate analyses, patients with MTV & gt;40 mL showed a significantly lower number of complete response/no recurrence than did patients with MTV ≤40 mL [68.2% versus 87.8%; hazard ratio (HR), 3.34; 95% confidence interval (95% CI), 1.09-10.08; P = 0.03], as is the same in tumor-node-metastasis stage (87.5% for I-II versus 90% for III versus 63.8% for IV; P = 0.02). However, MTV was only a significant predictor of short-term outcome by multivariate analyses (HR, 4.09; 95% CI, 1.02-16.43; P = 0.04). MTV & gt;40 mL indicated a significantly worse DFS than MTV ≤40 mL (HR, 3.42; 95% CI, 1.04-11.26;P = 0.04). The standardized uptake value for the primary tumor did not show any correlation with treatment outcome or DFS. Conclusion: MTV has a potential value in predicting short-term outcome and DFS in patients with pharyngeal cancers. (Clin Cancer Res 2009;15(18):5861–8)
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2009
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 70, No. 8 ( 2010-04-15), p. 3034-3041
    Abstract: Hepatocellular carcinoma (HCC) and cholangiocarcinoma (CC) are the major adult liver cancers. The existence of combined hepatocellular-cholangiocarcinoma (CHC), a histopathologic intermediate form between HCC and CC, suggests phenotypic overlap between these tumors. Here, we applied an integrative oncogenomic approach to address the clinical and functional implications of the overlapping phenotype between these tumors. By performing gene expression profiling of human HCC, CHC, and CC, we identified a novel HCC subtype, i.e., cholangiocarcinoma-like HCC (CLHCC), which expressed cholangiocarcinoma-like traits (CC signature). Similar to CC and CHC, CLHCC showed an aggressive phenotype with shorter recurrence-free and overall survival. In addition, we found that CLHCC coexpressed embryonic stem cell–like expression traits (ES signature) suggesting its derivation from bipotent hepatic progenitor cells. By comparing the expression of CC signature with previous ES-like, hepatoblast-like, or proliferation-related traits, we observed that the prognostic value of the CC signatures was independent of the expression of those signatures. In conclusion, we suggest that the acquisition of cholangiocarcinoma-like expression traits plays a critical role in the heterogeneous progression of HCC. Cancer Res; 70(8); 3034–41. ©2010 AACR.
    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 ...
  • 6
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 14, No. 10 ( 2015-10-01), p. 2238-2248
    Abstract: RET rearrangement is a newly identified oncogenic mutation in lung adenocarcinoma (LADC). Activity of dovitinib (TKI258), a potent inhibitor of FGFR, VEGFR, and PDGFR, in RET-rearranged LADC has not been reported. The aims of the study are to explore antitumor effects and mechanisms of acquired resistance of dovitinib in RET-rearranged LADC. Using structural modeling and in vitro analysis, we demonstrated that dovitinib induced cell-cycle arrest at G0–G1 phase and apoptosis by selective inhibition of RET kinase activity and ERK1/2 signaling in RET-rearranged LC-2/ad cells. Strong antitumor effect of dovitinib was observed in an LC-2/ad tumor xenograft model. To identify the acquired resistance mechanisms to dovitinib, LC-2/ad cells were exposed to increasing concentrations of dovitinib to generate LC-2/ad DR cells. Gene-set enrichment analysis of gene expression and phosphor-kinase revealed that Src, a central gene in focal adhesion, was activated in LC-2/ad DR cells. Saracatinib, an src kinase inhibitor, suppressed ERK1/2 phosphorylation and growth of LC-2/ad DR cells. Taken together, these findings suggest that dovitinib can be a potential therapeutic option for RET-rearranged LADC, in which acquired resistance to dovitinib can be overcome by targeting Src. Mol Cancer Ther; 14(10); 2238–48. ©2015 AACR.
    Type of Medium: Online Resource
    ISSN: 1535-7163 , 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2015
    detail.hit.zdb_id: 2062135-8
    SSG: 12
    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. 6597-6597
    Abstract: Pancreatic ductal adenocarcinoma (PDAC) is currently the 3rd leading cause of cancer-related deaths in the US. Although the all-stage 5-year survival rate is ~10%, early-stage 5-year survival is markedly superior and in excess of 40%. Hence, early detection of PDAC via blood-based liquid biopsies holds promise to reduce morbidity and mortality. PrognomiQ’s multi-omics platform performs deep and unbiased molecular profiling of blood samples to detect proteins, metabolites, lipids, mRNA, miRNA, cfDNA fragmentation and copy-number, and CpG methylation. Here we report results from training and validation of a classifier on a subset of that multi-omic data with the potential to enable the development of high sensitivity and specificity tests for early detection of PDAC.We conducted a case-control study comprising 146 subjects across 16 clinical sites, including 63 pathology-confirmed, untreated PDAC cases (12 stage I, 8 stage II, 4 stage III, 36 stage IV, and 3 stage unknown) and 83 age- and gender- matched controls without any known cancer. For each subject, venous blood samples including plasma were collected. Unbiased LCMS was used to detect and quantify proteins, and targeted, multiplexed MRM-LCMS assays were used for both metabolites and lipids. After data processing, we detected 54,114 proteomic features, 898 lipids, and 373 metabolites. 445 proteomic features, 170 lipids, and 37 metabolites were found to be significantly different as determined by Bonferroni-corrected Wilcoxon tests with FWER & lt; 0.05. For classification, the dataset was split into training (37 cases and 37 controls) and validation (26 cases and 46 controls) sets, with control for collection site and date, age, and gender. XGBoost models were constructed for each analyte class using ten repeats of 10-fold cross-validation. To improve specificity to PDAC, all proteomic features which mapped to GOBP terms associated with acute-phase response, inflammation, and immune response were excluded prior to training. The best-performing hyperparameters were used for a final model built on the full training set and then used for inference on the validation set. At 99% specificity, the proteomic classifier had sensitivities of 77%, 57%, and 88% for Stages 1-4, Stages 1-2, and Stages 3-4, respectively, estimated by bootstrap re-sampling of the validation results. Metabolomics had sensitivities of 81%, 71%, and 88%. Lipidomics had sensitivities of 65%, 71%, and 65%. A joint, multi-omic model was constructed by averaging the scaled probabilities of all models. This joint model improved performance at 99% specificity with sensitivities of 92%, 86%, and 94%, highlighting the synergy of multi-omics data, particularly phenotypically related omics such as those described here. Multi-omic classifiers such as these can serve as the foundation for blood-based liquid biopsies for the early detection of PDAC. Citation Format: John Blume, Ghristine Bundalian, Jessica Chan, Connie Chao-Shern, Jinlyung Choi, Rea Cuaresma, Kevin Dai, Sara N. Golmaei, Jun Heok Jang, Manoj Khadka, Ehdieh Khaledian, Thidar Khin, Yuya Kodama, Ajinkya Kokate, Joon-Yong Lee, Manway Liu, Hoda Malekpour, Megan Mora, Nithya Mudaliar, Preethi Prasad, Madhuvanthi Ramaiah, Saividya Ramaswamy, Peter Spiro, Kavya Swaminathan, Dijana Vitko, James Yee, Brian Young, Susan Zhang, Chinmay Belthangady, Bruce Wilcox, Brian Koh, Philip Ma. A multi-omics classifier achieves high sensitivity and specificity for pancreatic ductal adenocarcinoma in a case-control study of 146 subjects [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 6597.
    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
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 22_Supplement ( 2022-11-15), p. A038-A038
    Abstract: Pancreatic cancer is the third leading cause of cancer-related deaths in the United States. Disease biomarkers quantified from blood-based assays may help reduce mortality by enabling early detection, treatment selection, or response and resistance assessment. PrognomiQ has developed a multi-omics assay and analysis platform that comprehensively profiles blood samples to detect proteins, metabolites, lipids, mRNA, miRNA, cfDNA fragments, and methylation at CpG sites. This platform can provide deep insights into the biology of pancreatic cancer and could enable the development of high sensitivity and specificity tests for the early detection of pancreatic cancer. We conducted a case-control study comprising 196 subjects: 92 with untreated pancreatic cancer and 104 matched controls without pancreatic cancer. For each subject, blood was collected in assay-specific tubes and processed to provide 7 different `omics readouts. cfDNA and mRNA were isolated from samples and assayed following standard NGS protocols. cfDNA fragments were processed to estimate fragment-length disorder and copy-number variation along with CpG site methylation. In addition, targeted and untargeted LCMS were used to detect and quantify proteins, metabolites, and lipids. After normalization, non-parametric univariate analyses of cases versus controls were performed to identify differentially abundant features on all available samples for each assay. Unsupervised learning was used to investigate the separation of subjects into groups based on disease status for the subset of 157 subjects for which complete data on all 7 readouts were available. We detected 2,812 proteins, 811 lipids, 373 metabolites, and 110,864 mRNA transcripts in all samples where data for each assay was available. Of these, 275 proteins, 232 lipids, 49 metabolites, and 3385 mRNA transcripts were significantly different (FWER & lt; 0.05) in cases versus controls. From cfDNA data, we identified 35 non-contiguous genomic regions associated with fragment-length disorder, 557 with copy-number variation, and 5 with multiple, differentially methylated CpGs (FWER & lt; 0.05) that aggregately span 307 protein-coding genes; of these, the overlap with the differentially expressed proteins included E-cadherin (tumor suppressor) and N-cadherin (involved in epithelial-to-mesenchymal transition). Statistically significant genes and proteins were found to be associated with processes including Wnt signaling, regulation of focal adhesion assembly, and actin cytoskeleton organization. Multi-omics, unsupervised learning showed separation of early- and late-stage cases and controls. High-dimensional bioinformatics analyses systematically decomposed each `omics data type into joint and orthogonal components associated with pancreatic cancer. Ongoing multivariate analyses, including supervised machine learning, will further elucidate the biology of pancreatic cancer development, and serve as the foundation for high-sensitivity blood tests for the early detection and monitoring of pancreatic cancer. Citation Format: Ehdieh Khaledian, Preethi Prasad, John Blume, Ghristine Bundalian, Connie Chao-Shern, Jinlyung Choi, Rea Cuaresma, Jared Deyarmin, Jun Heok Jang, Manoj Khadka, Thidar Khin, Yuya Kodama, Ajinkya Kokate, Joon-Yong Lee, Manway Liu, Nithya Mudaliar, Madhuvanthi Ramaiah, Saividya Ramaswamy, Peter Spiro, Kavya Swaminathan, Preston Williams, Mi Yang, James Yee, Brian Young, Robert Zawada, Susan Zhang, Chinmay Belthangady, Bruce Wilcox, Philip Ma. High-dimensional, multi-omics analyses of proteins, metabolites, transcripts, and genes enable biomarker discovery in early- and late-stage pancreatic cancer [abstract]. In: Proceedings of the AACR Special Conference on Pancreatic Cancer; 2022 Sep 13-16; Boston, MA. Philadelphia (PA): AACR; Cancer Res 2022;82(22 Suppl):Abstract nr A038.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    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
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2015
    In:  Cancer Research Vol. 75, No. 15_Supplement ( 2015-08-01), p. 2810-2810
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 15_Supplement ( 2015-08-01), p. 2810-2810
    Abstract: The mammalian target of rapamycin (mTOR), a serine/threonine protein kinase, plays a critical role in the regulation of Akt signaling-mediated cell proliferation and transformation. Although several synthetic chemical compounds have developed to inhibit mTOR kinase activity, only few publications have been reported on the identification of natural compounds to target the mTOR kinase active pocket. In this study, we found that AME, a natural compound, inhibited cell proliferation by impairment of G1/S cell cycle transition when cells were co-treated with tumor promoters, such as epidermal growth factor. AME directly targeted the active pocket of mTOR kinase domain by competing with adenosine triphosphate (ATP), but not with PI3K and PDK1. We further confirmed that AME inhibited phosphorylation of Akt at Ser473, which is a target amino acid of mTOR complex 2 (mTORC2), and Akt-mediated GSK3β phosphorylation at Ser9, which resulted in activation of GSK3β. The activated GSK3β inhibited cell proliferation by c-Jun phosphorylation at Ser243, which facilitated destabilization and degradation of c-Jun through an ubiquitination-mediated proteasomal degradation pathway. Notably, decreased c-Jun stability by AME treatment suppressed EGF-induced neoplastic cell transformation in JB6 Cl41 mouse skin epidermal cells and HaCaT human skin keratinocytes in soft agar assay. Taken together, these results demonstrated that AME might be a natural chemopreventive agent targeting mTOR kinase active pocket. Key words: Natural compound, ATP-competitive mTOR kinase inhibitor, mTORC2/Akt/GSK3 signaling, chemoprevention Citation Format: Jeong-Hoon Jang, Cheol-Jung Lee, Mee-Hyun Lee, Young-Joon Surh, Yong-Yeon Cho. Identification of a natural compound as mammalian target of rapamycin kinase inhibitor. [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 2810. doi:10.1158/1538-7445.AM2015-2810
    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
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 14_Supplement ( 2016-07-15), p. 1176-1176
    Abstract: Aberrant Wnt/β-catenin signaling is implicated in the progression of several human cancers, including non-small-cell lung cancer (NSCLC). However, mutations in Wnt/β-catenin pathway com-ponents are uncommon in NSCLC, and epigenetic mechanisms controlling the Wnt/β-catenin path-way remain unclear. Here, we show that KIF3A, a member of the kinesin-2 motor family, plays a key role in suppressing Wnt/β-catenin signaling in NSCLC cells. Knockdown of KIF3A increases both β-catenin levels and transcriptional activity, with a concomitant promotion of malignant phenotypes, such as enhanced proliferation and migration, and upregulation of stemness markers. KIF3A binds to β-arrestin, and KIF3A depletion allows β-arrestin to form a complex with DVL2 and AXIN, result-ing in β-catenin stabilization. Although primary cilia, of which the biogenesis requires KIF3A, are thought to restrain the Wnt response, pharmacological inhibition of ciliogenesis does not enhance β-catenin activity in NSCLC cells. A correlation between KIF3A loss and worse NSCLC prognosis as well as upregulation of β-catenin and Cyclin D1 further suggests that KIF3A is a suppressor of Wnt/β-catenin signaling and tumorigenesis in NSCLC. Citation Format: Minsuh Kim, Yong-Ah Suh, Ju-hee Oh, Bo Ra Lee, Joon Kim, Se Jin Jang. The role of KIF3A in the suppression of canonical Wnt signaling through the KIF3A and β-arrestin complex, independent of the ciliary mechanism, in non-small cell lung cancer (NSCLC). [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 1176.
    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 ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...