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)  (186)
Material
Publisher
  • American Association for Cancer Research (AACR)  (186)
Language
Years
Subjects(RVK)
  • 11
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 74, No. 19_Supplement ( 2014-10-01), p. 1561-1561
    Abstract: Background Glioblastoma (GBM) is the most common malignant primary brain tumor in adults and is a leading cause of cancer-related death due to its invasive nature. Despite advances in radiation and chemotherapy following surgical resection of the tumor, the prognosis of GBM remains poor with an average survival time of less than one year. Accumulating evidence indicates the existence of tumor suppressor-like genes encoded on the 1p36 region. Recent investigations have reported Adherens Junctional Associated Protein-1 (AJAP1, also known as Shrew1) as another tumor suppressor-like gene on chromosome 1 in the 1p36 region. We and others also demonstrated that the AJAP1 promoter was highly methylated in a wide spectrum of glioma cell lines, and the loss of expression was associated with poorer survival in gliomas patients. However, the altered expression profiles of AJAP1 in gliomas as well as the underlying mechanisms of AJAP1 on glial cell migration and invasion are still poorly understood. Methods The gene profiles of AJAP1 in glioma patients were studied among four independent cohorts. Confocal imaging was used to analyze the AJAP1 localization. After AJAP1 over-expression in GBM cell lines, cellular polarity, cytoskeleton distribution, and anti-tumor effect were investigated in vitro and in vivo. Results AJAP1 expression was significantly decreased in gliomas compared with normal brain in REMBRANDT and CGCA cohorts. Additionally, low AJAP1 expression was associated with worse survival in GBMs in REMBRANDT and TCGA U133A cohorts and was significantly associated with classical and mesenchymal subtypes of GBMs among four cohorts. Confocal imaging indicated AJAP1 localized in cell membranes in low-grade gliomas and AJAP1 over-expressing GBM cells, but difficult to assess in high-grade gliomas due to its absence. AJAP1 over-expression altered the cytoskeleton and cellular morphology in vitro, and inhibited the tumor growth in vivo. Conclusion In summary, gene profiling of gliomas showed that dysregulated AJAP1 exists in the early stage of gliomagenesis. In particular, AJAP1 expression is associated with low and high tumor grades, as well as the clinical outcome of patients with GBMs. Its over-expression predicts poor clinical outcome and may serve as a promising biomarker for intensive therapy, especially in Classical and Mesenchymal GBM patients. Few studies have investigated the function of AJAP1 and cytoskeleton regulation in gliomas. Further studies are warranted to explore the biological functions of AJAP1 that may improve our understanding of the initiation of gliomas and development of new biomarker and therapeutic target for individualized therapy of GBM. Citation Format: Lei Han, Kai-Liang Zhang, Jun-Xia Zhang, Liang Zeng, Chun-Hui Di, Brian Fee, Miriam Rivas, Tao Jiang, Darrell Bigner, Chun-Sheng Kang, David Cory Adamson. AJAP1 is dysregulated at an early stage of gliomagenesis and suppresses invasion through cytoskeleton reorganization. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 1561. doi:10.1158/1538-7445.AM2014-1561
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2014
    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 ...
  • 12
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 13_Supplement ( 2017-07-01), p. 3269-3269
    Abstract: Esophageal cancer (EC) has been well documented with predominant occurrence in male and with major histopathological type of esophageal squamous cell carcinoma (ESCC) in China. However, the influence by gender on ESCC survival is largely unknown in China. The present study was thus undertaken to elucidate the impact by gender on survival of Chinese patients with ESCC. A total of 17,969 ESCC patients was enrolled in this study, including 11,127 males with a mean age of 60.2±8.6 and 6842 females with a mean age of 61.0±8.4.All the patients were from ESCC database in Henan Key Laboratory for Esophageal Cancer Research of the First Affiliated Hospital, Zhengzhou University (1975 to 2015). Only the patients with T2–4N0–1M0–1 were enrolled in this study. Based on the diagnosed time, the patients were classified into two groups, group I (1996–2005, 6885/38.3%) and group II (2006–2015, 11084/61.7%). The followed–up was undertaken until October, 2016. Kaplan–Meier models were for describing the survival curves and Cox proportional hazards models were undertaken to observe the risk of death of male and female. The results demonstrated that the male patients had a poor survival than female (X2=58.17, P & lt;0.001). Furthermore, similar results were observed for the two different periods of 1996–2005 (X2=21.08, P & lt;0.001) and 2006–2015 (X2=34.67, P & lt;0.001). The present results indicate that gender is an independent survival factor for ESCC and the female patients with ESCC have a dramatic prolonged survival than male patients in China. [Supported by Doctoral Team Foundation of the First Affiliated Hospital of Zhengzhou University (2016-BSTDJJ-03), Natural Science Foundation of Henan Province (20161110) and Correspondening author: Li Dong Wang, email:ldwang2007@126.com] Note: This abstract was not presented at the meeting. Citation Format: Jian Liang Lu, Jian Liang Lu, Rui Ping Xu, Bei Li, Li Xin Wan, Ming Yao Chen, Chun Jiang Liu, Zhan Qiang Han, Fang Fang Shen, Jian Bo Zhao, Hai Zhou Guo, Wei Cao, Bing Yu Fan, Li Dong Wang. Impact of gender on survival of the patients with esophageal squamous cell carcinoma at a single institute in China [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 3269. doi:10.1158/1538-7445.AM2017-3269
    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 ...
  • 13
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 7_Supplement ( 2023-04-04), p. 4444-4444
    Abstract: Hyperactivation of the PI3K pathway has been reported to correlate with resistance to immune checkpoint blockade therapy (ICB) in melanoma, highlighting the therapeutic potential of combining PI3K inhibition (PI3Ki) with ICB. To maximize the clinical benefit of PI3Ki-based immune oncology (IO) combination, we characterized the role of PI3K isoforms in tumor and T cells and determined the immunological impacts of PI3Ki alone or in combination with ICB. Inhibitions of PI3K were achieved by either genetic knockdown (KD) or the bioactive compound in PTEN-present (B16/MC38), PTEN-absent (BP/D4M) tumor cell lines, and CD8+ T cells (Pmel-1). Following PI3Ki, we determined the activation status of the PI3K pathway (p-AKT level), transcriptional profile, and cellular function of these cells. We found both in vitro KD and pharmacological inhibition of either PI3Kα or PI3Kβ displayed a dramatic reduction of the PI3K pathway in tumor cells but moderate or no reduction in T cells, whereas the PI3K pathway significantly decreased in T cells with PI3Kγ or PI3Kδ inhibition. KD of PI3Kα or β isoforms drastically sensitized both D4M and MC38 tumors to αPD1 in vivo. We also observed that only PI3Kγ or PI3Kδ inhibition profoundly suppressed cytokine production and cytotoxicity of CD8+ T cell, suggesting that PI3Kα or PI3Kβ isoform inhibition can achieve tumor specific PI3Ki with limited impacts on T cell function. Furthermore, we used multiple syngeneic melanoma models to determine whether PI3K isoform inhibition can synergize the antitumor activity of ICB in vivo. In PTEN-present tumors, BYL719 (BYL, a PI3Kα inhibitor) synergized with αPD1 to delay tumor growth and extend survival (median survival of MC38-bearing mice in control (Ctrl), BYL, αPD1, and combination (Comb) groups: 30, 36, 33, and & gt;45 respectively; p & lt;0.05: Ctrl/BYL/αPD1 vs Comb). However, a limited combinatorial effect between GSK2636771(a PI3Kβ inhibitor) and αPD1 was observed in PTEN-present tumor models. Moreover, the combination of BYL and αPD1 exhibits superior antitumor activity in a spontaneous Braf-mutant, PTEN-loss melanoma model when compared with either reagent. Mechanistically, the combination of BYL and αPD1 improved CD8+ T cells tumor infiltration (14 days treatment, mean CD8+ number/mg of the tumor, Ctrl:1392.9, BYL:2073.9, αPD1:1545.2, Comb:4691.8; p & lt;0.01: Ctrl/BYL/αPD1 vs Comb) and reduced MDSCs in MC38 tumors (p & lt;0.05: Ctrl vs Comb). Multi-omics profiling of tumor cells with in vitro and in vivo PI3K isoform inhibition is ongoing. Collectively, our results demonstrate that PI3Kα inhibitor can potentiate T cell-mediated antitumor immune responses regardless of PTEN status, providing a strong rationale for the clinical development of the BYL-based IO combination. In collaboration with Novartis, MD Anderson Cancer Center will launch a Phase I/II trial of the FDA-approved BYL in combination with αPD1 in advanced melanoma and breast cancer patients. Citation Format: Ritu Bohat, Xiaofang Liang, Chunyu Xu, Yitao Tang, Jiakai Hou, Nicholas A. Egan, Leilei Shi, Ashley Guerrero, Roshni Jaffery, Elizabeth M. Burton, Han Liang, Hussein Tawbi, Michael A. Davies, Weiyi Peng. Targeting PI3K isoforms to improve the effectiveness of T cell mediated 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 4444.
    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 ...
  • 14
    In: Cancer Immunology Research, American Association for Cancer Research (AACR), Vol. 8, No. 7 ( 2020-07-01), p. 952-965
    Abstract: Programmed cell death 1 ligand 1 (PD-L1) is a key driver of tumor-mediated immune suppression, and targeting it with antibodies can induce therapeutic responses. Given the costs and associated toxicity of PD-L1 blockade, alternative therapeutic strategies are needed. Using reverse-phase protein arrays to assess drugs in use or likely to enter trials, we performed a candidate drug screen for inhibitors of PD-L1 expression and identified verteporfin as a possible small-molecule inhibitor. Verteporfin suppressed basal and IFN-induced PD-L1 expression in vitro and in vivo through Golgi-related autophagy and disruption of the STAT1–IRF1–TRIM28 signaling cascade, but did not affect the proinflammatory CIITA-MHC II cascade. Within the tumor microenvironment, verteporfin inhibited PD-L1 expression, which associated with enhanced T-lymphocyte infiltration. Inhibition of chromatin-associated enzyme PARP1 induced PD-L1 expression in high endothelial venules (HEV) in tumors and, when combined with verteporfin, enhanced therapeutic efficacy. Thus, verteporfin effectively targets PD-L1 through transcriptional and posttranslational mechanisms, representing an alternative therapeutic strategy for targeting PD-L1.
    Type of Medium: Online Resource
    ISSN: 2326-6066 , 2326-6074
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    detail.hit.zdb_id: 2732517-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 15
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2013
    In:  Molecular Cancer Research Vol. 11, No. 3 ( 2013-03-01), p. 294-302
    In: Molecular Cancer Research, American Association for Cancer Research (AACR), Vol. 11, No. 3 ( 2013-03-01), p. 294-302
    Abstract: Pancreatic cancer significantly affects the quality of life due to the severe abdominal pain. However, the underlying mechanism is not clear. This study aimed to determine the relationship between Substance P (SP) and pancreatic cancer perineural invasion (PNI) as well as the mechanism of SP mediating pancreatic cancer PNI, which causes pain in patients with pancreatic cancer. Human pancreatic cancer cells and newborn dorsal root ganglions (DRG) were used to determine the expression of SP or NK-1R in pancreatic cancer cells and DRGs cells by QT-PCR and Western blotting. The effects of SP on pancreatic cancer cell proliferation and invasion were analyzed using MTT assay and Transwell Matrigel invasion assay, respectively. Alterations in the neurotropism of pancreatic cancer cells were assessed by coculture system, which mimics the interaction of tumor/neuron in vivo. SP is not only widely distributed in the neurite outgrowth from newborn DRGs but also expressed in MIA PaCa-2 and BxPC-3 cells. NK-1R is found to be overexpressed in the pancreatic cancer cell lines examined. SP induces cancer cell proliferation and invasion as well as the expression of matrix metalloproteinase (MMP)-2 in pancreatic cancer cells, and NK-1R antagonists inhibit these effects. Furthermore, SP promotes neurite outgrowth and the migration of pancreatic cancer cell cluster to the DRGs, which is blocked by NK-1R antagonists in the coculture model. Our results suggest that SP plays an important role in the development of pancreatic cancer metastasis and PNI, and blocking the SP/NK-1R signaling system is a novel strategy for the treatment of pancreatic cancer. Mol Cancer Res; 11(3); 294–302. ©2012 AACR.
    Type of Medium: Online Resource
    ISSN: 1541-7786 , 1557-3125
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2013
    detail.hit.zdb_id: 2097884-4
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 16
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 13_Supplement ( 2017-07-01), p. 3732-3732
    Abstract: Expanding the paradigm of solid tumor profiling from single-gene testing to comprehensive panels presents many challenges. One such challenges is the ability of these panels to detect genetic alterations from FFPE samples, where the DNA is of low abundance and often heavily compromised. Despite these challenges, next-generation sequencing (NGS) offers the ability to assess multiple variants simultaneously in an ever-expanding list of relevant tumor genes. To that end, Illumina developed a comprehensive, hybrid capture-based NGS assay targeting 170 key cancer genes that is FFPE optimized. The assay consists of a DNA workflow for the identification of single and multiple nucleotide variants (SNVs, MNVs), small insertions and deletions (indels), gene amplifications, as well as a RNA workflow for the identification of splice variants and gene fusions. Following sequencing on the NextSeq® or HiSeq® instruments, the analytical pipeline initiates variant calling. The DNA aligner and variant callers were first optimized against the simulated read data from & gt;40,000 COSMIC[1] mutations reported in the exons of the 170 genes. To reduce false positive variant calling due to systematic errors, each variant call was evaluated against its locus specific background error distribution. This distribution was compiled from a panel of FFPE normal samples and was also used to normalize against systematic bias in read coverage to increase the accuracy of amplification calling. Furthermore, gene amplification calling was improved by the addition of enhancer probes to the hybrid capture pool. The analytical sensitivity and specificity of TruSight® Tumor 170* was assessed on a large collection of FFPE samples and reference material. A panel of 72 cancer samples, including multiple tissue types, reference standards, and cell line and FFPE mixes were used to evaluate the limit of detection. The samples contained 533 SNVs, 80 indels including deletions up to 30 base pairs and insertions up to 31 base pairs, 4 MNVs, and 31 gene amplifications, characterized by orthogonal testing methods. Using 40 ng DNA input, detection sensitivity of the & gt;1000 variants (including replicates) tested at variant allele frequencies down to ~5% was at 99.6%, while detection sensitivity of gene amplifications as low as 1.45x to 2.2x was at 98%. For limit of blank samples, a panel of 24 normal samples was used. Again using 40 ng DNA input, we show & gt;99% specificity for small variant calling and & gt;95% specificity for gene amplification calling. These data demonstrates the TruSight® Tumor 170 is able to detect multiple variant types within a single sample at low nucleic acid input, while exhibiting high analytical sensitivity and specificity for low allele fraction detection. [1] Forbes, et al. (2015) *For Research Use Only. Not for use in diagnostic procedures. Citation Format: Danny Chou, Xiao Chen, Austin Purdy, Li Teng, Byron Luo, Chen Zhao, Laurel Ball, Allan Castaneda, Katie Clark, Brian Crain, Anthony Daulo, Manh Do, Tingting Du, Sarah Dumm, Yonmee Han, Michael Havern, Chia-Ling Hsieh, Tingting Jiang, Suzanne Johansen, Scott Lang, Rachel Liang, Jennifer S. LoCoco, Jaime McLean, Yousef Nassiri, Jason Rostron, Jennifer Silhavy, June Snedecor, Natasha Talago, Kevin Wu, Clare Zlatkov, Ali Kuraishy, Karen Gutekunst, Sohela De Rozieres, Matthew Friedenberg, Han-Yu Chuang, Anne C. Jager. Analytical performance of TruSight® Tumor 170 on small nucleotide variations and gene amplifications using DNA from formalin-fixed, paraffin-embedded (FFPE) solid tumor samples [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 3732. doi:10.1158/1538-7445.AM2017-3732
    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 ...
  • 17
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2018
    In:  Cancer Research Vol. 78, No. 13_Supplement ( 2018-07-01), p. 550-550
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. 550-550
    Abstract: Cancer cells employ multiple mechanisms to evade the recognition by the host immune system, including the expression of the negative T cell regulatory molecule PD-L1. PD-1/PD-L1-mediated immune escape plays key role during cancer development. It is crucial to unravel the regulatory mechanism of PD-1/PD-L1. In this study, we seek one enhancer (we named “enh9”) that regulates the transcription of PD-L1 by systematic exploration. First, we found that PD-L1 and enh9 were co-expressed in multiple cancer types, including both lung adenocarcinoma and lung squamous cell carcinoma, which was further validated in a cohort of 130 lung cancer cell lines from the Cancer Cell Line Encyclopedia database. Then the direct interaction between PD-L1 and enh9 was further confirmed by Hi-C dataset. To validate the transcriptional regulation of PD-L1 by enh9, we knocked out enh9 using CRISPRi (CRISPR/ clustered regularly interspaced short palindromic repeats interference) in lung adenocarcinoma cell line A549, and single cell clones with double-copy deletion of enh9 were screened out. The expression of PD-L1 was dramatically reduced at both mRNA and protein level in enh9 knockout cells. Furthermore, we found that the cell proliferation and metastasis of enh9 knockout cells were significantly decreased compared to the control cells. Based on the previous studies that PD-L1 intrinsically regulated cancer cells' proliferation in ovarian and melanoma cancer cells, ongoing work is to determine whether enh9 regulates the cancer cell hallmarks by direct regulation of the expression of PD-L1 in lung cancer cells. The regulatory mechanisms will be deeply studied. In conclusion, we found an enhancer regulated the expression of PD-L1, which was one famous immune checkpoint, and it may play key roles in the cancer cell hallmarks. Citation Format: Chunyan Li, Han Chen, Han Liang. One enhancer stimulates cancer cells proliferation and metastasis via the regulation of PDL1 expression [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 550.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    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 ...
  • 18
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2013
    In:  Cancer Research Vol. 73, No. 8_Supplement ( 2013-04-15), p. 677-677
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 73, No. 8_Supplement ( 2013-04-15), p. 677-677
    Abstract: Gene silencing mediated by aberrant promoter DNA hypermethylation is one of the key features of cancer. Although such modification is heritable, its dynamic nature and reversibility through pharmacological interventions make it an attractive therapeutic target. Over the past few decades, various DNA methyltransferase inhibitors have been developed with the goal of reactivating aberrantly silenced genes. Two FDA-approved demthylating agents, decitabine and azacitidine are efficacious for the treatment of myeloid malignancies (MDS). It has been shown that transient exposure of tumor cells to low dose decitabine and azacitidine results in reduced tumorigenecity. However, the mechanism underlying clinical efficacies of these DNA methyltransferase inhibitors remains unclear. Here we investigate the long-term effect (up to 10 weeks) on population doubling time and the returning of DNA methylation (rebound methylation) after transient exposure to decitabine in three cancer cell lines, HCT116, T24 and HL-60. We show that transient exposure results in inhibition of cell growth and reduction of their colony formation capacity (up to 42 days). Furthermore, we also show that the rebound methylation occurs at different rates depending on the region and the type of genes. The discrepancy in population doubling time between the vehicle and decitabine treated cells disappears when rebound methylation fully restores, suggesting the presence of a correlation between cell doubling and rebound methylation. The majority of probes, which exhibit fast rebound methylation, are located in gene bodies. We show a positive correlation between gene body methylation and expression. However, there is a small group of fast rebound probes located at the promoters. Many of them belong to cancer-testis antigens, which get reactivated and quickly re-silenced. On the other hand, the majority of probes, which exhibit slow rebound methylation are located at promoters and show an inverse correlation between methylation and expression. The presence of a nucleosome-depleted region and enrichments of active histone marks, H3K4me3 and H2A.Z are observed at the promoters of those genes up to 42 days after drug treatment. Interesting, this group is enriched for genes down-regulated in colon adenocarcinoma when compared to normal colon (p=3.3e−6). Taken together, the sustained reactivation of those genes may be the driving force in reducing tumorigenecity of cancer cells. Our study elucidates the mechanism of decitabine, thus advancing our understanding of epigenetic therapy. Citation Format: Gangning Liang, Han Han, Daniel D. De Carvalho, Xiaojing Yang, Peter A. Jones. Transient exposure to decitabine results in sustained cell growth inhibition and long term DNA demethylation at specific loci. [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 677. doi:10.1158/1538-7445.AM2013-677 Note: This abstract was not presented at the AACR Annual Meeting 2013 because the presenter was unable to attend.
    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 ...
  • 19
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2013
    In:  Cancer Research Vol. 73, No. 8_Supplement ( 2013-04-15), p. 5148-5148
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 73, No. 8_Supplement ( 2013-04-15), p. 5148-5148
    Abstract: Driver mutations are somatic mutations that provide growth advantage to tumor cells, while passenger mutations are those that are not functionally associated with oncogenesis. Distinguishing drivers from passengers is challenging because drivers occur much less frequently than passengers, they turn to have low prevalence, and their functions are multifactorial. Missense mutations are excellent candidates for driver identification, as they occur more frequently and are potentially easier to target than other types of mutations. Although several methods have been developed for predicting the functional impact of missense mutations, only a few are specifically designed for identifying driver mutations. As more mutations are being discovered, more accurate predictive models can be developed using machine learning approaches that systematically characterize the commonality and peculiarity of somatic mutations under specific cancer background. Here, we present such a Cancer Driver Annotation (CanDrA) tool that assesses the driver potential of missense somatic mutations based on a set of 95 structural and evolutionary features computed by over 10 functional prediction algorithms from SNVBOX, VEP, ANNOVAR and Mutation Assessor. Through feature optimization and supervised training, CanDrA outperforms existing tools in analysing the glioblastoma multiforme and the ovarian carcinoma data in The Cancer Genome Atlas (TCGA) and the Cancer Cell Line Encyclopedia project. Citation Format: Yong Mao, Han Chen, Han Liang, Funda Meric-Bernstam, Gordon Mills, Ken Chen. CanDrA: Cancer-specific driver missense mutation annotation with optimized features. [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 5148. doi:10.1158/1538-7445.AM2013-5148
    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 ...
  • 20
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 81, No. 13_Supplement ( 2021-07-01), p. CT158-CT158
    Abstract: Background: Savolitinib is a potent and selective MET tyrosine kinase inhibitor. It demonstrated clinical efficacy and a manageable safety profile in Chinese NSCLC pts with METex14 alterations in a phase 2 study (NCT028997479). Here, we report the post-hoc ctDNA analysis of METex14 at baseline and clearance upon treatment and the association of these findings with clinical outcome. In addition, concurrent gene alterations in ctDNA samples from the patients treated with savolitinib and impact on clinical efficacy is explored. Methods: Plasma samples were prospectively collected pre-dose and at tumor assessment visits, until disease progression or end of treatment. MET and other somatic gene alterations in the ctDNA samples were detected by next generation sequencing (425-gene panel, Geneseeq). Results: Sixty-six pts provided baseline plasma samples, of which METex14 ctDNA was detectable in 46 (70%) and undetectable in the remaining 20 pts (30%). Among the 46 ctDNA detectable pts, 19 were pulmonary sarcomatoid carcinoma (19/22, 86%) and 27 were other NSCLC (27/44, 61%), respectively. Of the 46 baseline-detectable pts, 24 were clearance evaluable and 22 had no qualified post baseline samples for clearance evaluation. Of the 24 clearance evaluable pts, 14 achieved ctDNA clearance (undetectable) with a median time to clearance of 1.4 months of treatment (Min 1.4 m, Max 4.2 m). The PFS and OS were compared for pts based on their METex14 ctDNA status at baseline and upon treatment. As shown in the table, METex14 baseline undetectable or clearance pts demonstrated significantly longer mPFS and mOS. Furthermore, in 21 pts with analyzable ctDNA samples at baseline and at disease progression, additional gene alterations were observed such as KRAS, NRAS, BRAF, PIK3CA as well as secondary MET mutations and FGF19 amplification in 12 pts (57%). These alterations might be associated with treatment resistance. Conclusions: The results suggest that ctDNA METex14 undetectable at baseline or clearance upon savolitinib treatment may define favorable treatment outcome. Confirmation of this finding and the predictive value of the ctDNA with larger sample size is desirable. METex14 ctDNAmPFS(mon, 95% CI)HR (95% CI)P valuemOS(mon, 95% CI)HR (95% CI)P valueBaseline undetectable (n=20)13.8(4.2, 22.1)--NC(10.9, NC)--Baseline detectable (n=46)5.6(4.1, 6.9)1.77(0.88, 3.57)0.10810.9(9.2, 14.0)3.26(1.35, 7.89)0.006- Clearance (n=14)30.3(6.8, NC)0.72(0.28, 1.87)0.50135.8(14, NC)0.89(0.25, 3.18)0.835- Non-clearance (n=10)5.5(0.66, 5.6)4.94(1.83, 13.36)0.0028.7(0.8, 10.6)7.06(2.39, 20.89) & lt;0.001- Non-evaluable (n=22)4.1(4.0, 6.9)3.45(1.48, 8.03)0.00610.6(4.8, 12.0)5.88(2.24, 15.47) & lt;0.001CI, confidence interval; HR, hazard ratio; NC, non-calculable Citation Format: Yongfeng Yu, Yongxin Ren, Jian Fang, Lejie Cao, Zongan Liang, Qisen Guo, Sen Han, Zimei Ji, Ye Wang, Yulan Sun, Yuan Chen, Xingya Li, Hua Xu, Jianying Zhou, Liyan Jiang, Ying Cheng, Zhigang Han, Jianhua Shi, Gongyan Chen, Rui Ma, Yun Fan, Sanyuan Sun, Longxian Jiao, Xiaoyun Jia, Linfang Wang, Puhan Lu, Jing Li, Qian Xu, Xian Luo, Weiguo Su, Shun Lu. ctDNA analysis in the savolitinib phase II study in Non-Small Cell Lung Cancer (NSCLC) patients (pts) harboring MET exon 14 skipping alterations (METex14) [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr CT158.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2021
    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...