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
  • 1
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. LB-165-LB-165
    Abstract: Breast cancer incidence rates, clinical features, and risk factors are known to vary significantly by race/ethnicity. Recently, tumor sequencing and profiling analyses have improved molecular classification and refined existing breast cancer subtypes. However, detailed genomic analyses are limited in Asian populations. The goal of this study was to profile fresh frozen breast tumors and paired normal tissue in breast cancer cases who had surgeries at two hospitals in Hong Kong. Only tumors containing 50% or more tumor cells were included for sequencing. RNA sequencing (RNASeq), whole exome sequencing, and SNP array were conducted in ~100 tumors. In 92 tumors with RNASeq data, the unsupervised clustering analysis based on 2000 most variable genes identified two subgroups in patients with luminal tumors (defined by PAM50). The most significantly differentially expressed genes between the two luminal subgroups were enriched in immune and inflammation pathways. We further conducted the clustering analysis within 72 luminal tumors (luminal A + luminal B) using 700 immune genes, which again classified the patients into two subgroups (high-immune luminal [HILum] and low-immune luminal [LILum] ). Three computational algorithms, CIBERSORT, MCP-counter, and ESTIMATE, were used to infer the fraction or abundance of immune cell populations in each tumor using RNASeq data. We found significant differences in the abundance of most immune cell populations between the two luminal subgroups. The HILum group, which consisted of 53% of all luminal tumors, had a higher overall immune score as well as a higher score for each immune cell subpopulation measured by MCP-counter, at levels similar to those in patients with HER2-positive or basal-like tumors. HILum tumors also expressed significantly higher levels of immune checkpoint genes such as PD1, PDL1, and CTLA4 compared with LILum tumors. Interestingly, HILum was also associated with lower ESR1 expression, earlier age onset, higher level of ploidy and loss-of-heterozygosity, and higher proportions of APOBEC signatures (mutation signatures 2 and 13 in COSMIC) and lower occurrence of the defective DNA mismatch repair signature (signature 6) compared with LILum. The HILum subtype is not specific to Hong Kong since similar results were seen in both TCGA white and Asian breast cancer patients. In summary, we identified a group of luminal patients who had higher levels of immune cell infiltration and immune checkpoint gene expression and are associated with distinct genomic features. These patients may potentially benefit from the immune checkpoint inhibitor therapies. Citation Format: Xiaohong (Rose) Yang, Bin Zhu, Difei Wang, Hela Koka, Tongwu Zhang, Feng Wang, Cherry Wu, Koon Ho Tsang, Wing-cheong Chan, Sze Hong Law, Priscilla Lee, Mengjie Li, Wentao Li, Suyang Wu, Zhiguang Liu, Mingyi Wang, Kristine Jones, Amy Hutchinson, Belynda Hicks, Jianxin Shi, Shelly Lap Ah Tse. Identification of a luminal subtype with high immune abundance among breast cancer patients in Hong Kong, China [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 LB-165.
    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 ...
  • 2
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 13_Supplement ( 2019-07-01), p. 1394-1394
    Abstract: Background Incidence rate of thyroid cancer is steadily increasing due to overdiagnosis and overtreatment. Thyroid ultrasound is commonly used to diagnose thyroid cancer. The aim of this study is to examine the accuracy of using deep convolutional neural network (DCNN) models to improve diagnosis of thyroid cancer by analyzing sonographic imaging data from clinical thyroid ultrasound. Methods A total of 131,731 sonographic images from 17,627 thyroid cancer patients and 180,668 sonographic images from 25,325 controls used as training set were obtained from Tianjin Cancer Hospital. Images from anatomical sites that did not have cancer according to location sign on the image were not included. All thyroid cancer patients and 13·2% of controls (51,255 images) were confirmed by pathological reports. DCNN is a specific type of neural network optimized for image recognition. We trained two DCNN models on the training set and subsequently evaluated the performance on one independent internal (Tianjin, 1,118 individuals) and two external (Jilin,154 individuals; Weihai, 1,420 individuals) validation sets. Individuals in the validation sets all have pathological examinations. We compared the specificity/sensitivity of DCNN models with the performance of six thyroid ultrasound radiologists on these three validation sets. Findings DCNN model achieved high performance in identifying thyroid cancer patients versus six experience radiologists: for Tianjin validation set, sensitivity was 92·2% versus 96·9% (95% CI 89·7% - 94·3% vs. 93·9% - 98·6%; p = 0·003), and specificity was 85·6% versus 59·4% (95% CI 82·4% - 88·4% vs. 53% - 65·6%; p & lt; 0·0001); for Jilin validation set, sensitivity was 84·3% versus 92·9% (95% CI 73·6% - 91·9% vs. 84·1% - 97·6%; p = 0·05), and specificity was 86·9% versus 57·1% (95% CI 77·8% - 93·3% vs. 45·9% - 67·9%; p & lt; 0·0001); for Weihai validation set, sensitivity was 84·5% versus 89% (95% CI 81·2% - 87·4% vs. 81·9% - 94%; p = 0·2), and specificity was 87·5% versus 68·6% (95% CI 85·1% - 89·6% vs. 60·7% - 75·8%; p & lt; 0·0001). Interpretation DCNN models exhibited high accuracy, sensitivity, and specificity in identifying thyroid cancer patients at levels comparable to or higher than six experienced radiologists. Conferred by the high specificity of DCNN models, the rate of overdiagnosis and overtreatment of patients with thyroid cancer is expected to decrease. This supports future application of the deep learning models to clinical practice for thyroid cancer diagnosis. However, further validation of these DCNN models in prospective clinical trials is warranted. Funding The Program for Changjiang Scholars and Innovative Research Team in University in China (IRT_14R40), National Natural Science Foundation of China (31801117). Citation Format: Xiangchun Li, Sheng Zhang, Qiang Zhang, Xi Wei, Yi Pan, Jing Zhao, Xiaojie Xin, Xiaoqing Wang, Fan Yang, Jianxin Li, Meng Yang, Qinghua Wang, Xiangqian Zheng, Yanhui Zhao, Lun Zhang, Xudong Wang, Zhimin Zheng, Christopher T. Whitlow, Metin N. Gurcan, Boris Pasche, Ming Gao, Wei Zhang, Kexin Chen. Diagnosis of thyroid cancer using deep convolutional neural network models applied to sonographic images from clinical ultrasound exams [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 1394.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. CT133-CT133
    Abstract: Background Although successful discovery and applications of genome editing system, CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats/associated nuclease 9), has been reported in basic science research, it has been unclear what happen to patients when moving CRISPR/Cas9 technology to bedside. We performed the first-in-man clinical trial (NCT02793856), phase I, to assess the safety of CRISPR/Cas9-mediated knockout of the programmed cell death 1 (PD-1) gene in autologous T lymphocytes in patients with metastatic non-small cell lung cancer (NSCLC). Methods Using a 3 + 3 + 3 dose-escalation study, we assigned patients with advanced NSCLC with PD-L1 expression positive on tumor cells that had progressed after the 3rd line standard therapeutic regimens. Pre-A cohort was used to enroll 2 patients who received PD-1 deficient gene-edited T cells (PD-1- T) with 2x107 cells/kg in one cycle via CRISPR/Cas9 for safety concern. Then 3 cohorts (A, B, C) enrolled 3 patients receiving total dose of PD-1- T cells with 1 ×107/kg, 2 x 107/kg, 4×107/kg each cycle every 4 weeks, respectively. The primary outcome was safety. Secondary end points were objective response rate, 8 weeks progression-free survival. In exploratory analyses, next-generation sequencing (NGS) was performed on PD-1 editing region of T cell and the CDR3 region of the T-cell receptor beta chain. Results 8 patients received totally 16 cycles of PD-1- T cell infusion in cohort Pre-A, A and B. 13 adverse events (AEs) related to the PD-1- T cell infusion occurred (84.6% of grade 1 and 15.4% of grade 2). Most common AEs were acute fever and hepatic dysfunction (both 15.4%). No DLT and 3-5 AEs were found. 92.3% (12/13) AEs presented within 1st cycle. 7 patients were response evaluable. 2/4 of patients receiving 2 x 107/kg PD-1- T cell experienced SD while other 3/3 patients with lower dose had disease progression. 8-week PFS were 28.6%. Of exploratory data, numerous novel T-cell clonotypes were detected in PD-1- T cell products but not in peripheral bloods before cell infusion. Intriguingly, the persisting novel T cell clones were found in patient peripheral bloods with the frequency of 0.005%-3.05% during the cell therapy, indicating the existence of potential responsive T cell clones. Conclusion The patients in this trial seemed safety when conducing the PD-1 deficient engineered T cells with CRISPR/Cas9 system. Further study should be performed to explore the effective dose and the related immune response. Citation Format: You Lu, Meijuan Huang, Tao Deng, Xiaojuan Zhou, Kun Yu, Maozhi Liang, Lei Deng, Jianxin Xue, Xin Yi, Zhenyu Ding, Youling Gong, Jiang Zhu, Yongsheng Wang, Yuqi Wang, Jin Song, Ruizhan Tong, Li Li, Jingwen Huang, Feifei Na, Min Zhao, Chong Chen, Yuquan Wei, Weimin Li. A phase I trial of PD-1 deficient engineered T cells with CRISPR/Cas9 in patients with advanced non-small cell lung cancer with PD-L1 expression [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AAC R; Cancer Res 2018;78(13 Suppl):Abstract nr CT133.
    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 ...
  • 4
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 72, No. 8_Supplement ( 2012-04-15), p. 2931-2931
    Abstract: While lung cancer is largely caused by tobacco smoking, inherited genetic factors play a role in its etiology. Genome-wide association studies (GWAS) in Europeans have robustly demonstrated only three polymorphic variations influencing lung cancer risk. Tumor heterogeneity may have hampered the detection of association signal when all lung cancer subtypes were analyzed together. In a GWAS of 5,355 European smoking lung cancer cases and 4,344 smoking controls, we conducted a pathway-based analysis in lung cancer histologic subtypes with 19,082 SNPs mapping to 917 genes in the HuGE-defined “inflammation” pathway. We identified a susceptibility locus for squamous cell lung carcinoma (SQ) at 12p13.33 (RAD52, rs6489769), and validated it in three independent samples totaling 3,359 SQ cases and 9,100 controls (odds ratio=1.20, Pcombined=2.3×10−8). The combination of pathway-based approaches and information on disease specific subtypes can improve the identification of cancer susceptibility loci in heterogeneous diseases. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 2931. doi:1538-7445.AM2012-2931
    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 ...
  • 5
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 14_Supplement ( 2016-07-15), p. 1369-1369
    Abstract: Aldo-keto reductase family 1 member C3 (AKR1C3) catalyzes the reduction of a diverse group of substrates, including prostaglandin (PG) D2 and PGH2. It has been reported that AKR1C3 is overexpressed in the majority of hepatocellular carcinomas (HCC) with 58% of HCC patient surgical tumor samples having strong expression of the enzyme1. Tumors overexpressing AKR1C3 can be resistant to radiation therapy2 and chemotherapies3. AKR1C3 is also expressed in normal tissues,but its expression is much lower than in HCC tissue1. HCC is the sixth most common cause of cancer. It has very poor prognosis, and is the second leading cause of cancer death in all cancers. Treatment options for late stage liver cancer are very limited, with Sorafenib, a multi-tyrosine kinase inhibitor, being the only approved drug with limited efficacy. More effective therapies are urgently needed. TH3424 is a prodrug which selectively releases a DNA alkylating agent upon exposure to activated AKR1C3. In vitro cell proliferation tests with TH3424 in several HCC cell lines showed that it is very potent (IC50 ≤ 10 nM with 2 hour exposure) in killing cancer cells with high levels of AKR1C3, but less active (IC50 ≥ 10 μM) in killing cells with low or no AKR1C3 reductase. The activity of TH3424 correlates with the expression level of AKR1C3 as the potency of TH3424 is inhibited when used with a specific AKR1C3 inhibitor:4 (IC50: 4 nM vs 6.3 μM with SN33638) in a non-small cell lung cancer cell line (H460). In vivo orthotopic and patient derived disease (PDX) liver cancer model studies have shown promising efficacy with TH3424 being administer weekly with doses as low as 1.5mg/kg. TH3424 showed better efficacy than Sorafenib in an orthotopic HepG2 mouse model. Three of 8 mice treated with TH3424 at 2.5 mg/kg, Q7Dx3, and 8 of 8 mice treated with TH3424 at 5 mg/kg, Q7Dx3 were tumor free at day 35. Reference:1: Guise C. P.; Abbattista M. R.; Singleton R. S.; Holford S. D.; Connolly J.; Dachs G. U.; Fox S. B.; Rollock R.; Harvey J.; Guiford P.; Daňate F.; Wilson W. R.; and Patterson A. V.; Cancer Res.70(4), 2010, 1573 2: Xiong W.; Zhao J.; Yu H.; Li X.; Sun S.; Li Y.; Xia Q.; Zhang C.; He Q.; Gao X.; Zhang L.; Zhou D.; Plos One, V. 9 (11), 2014, e111911 3: Liu C.; Lou W.; Zhu Y.; Yang J.; Nadiminty N.; Gaikwad N.; Evans C.; Gao A.; Cancer Res.; 2015, 75(7), 1413 4: Flanagan J. U.; Atwell G. J.; Heinrich D. M.; Brooke D. G.; Silva S.; Rigoreau L. J. M.; Trivier E.; Turnbull A. P.; Raynham T.; Jamieson S. M. F.; Denny W. A.; Bioorg. Med. Chem.; 22(2014); 967 Citation Format: Jianxin Duan, Zhong Wang, Qing Li, Yeyu Cao, Ping He, Fanying Meng, Changhua Zhou, Yanhong Wang, Gavin Qu, Henry Li, Jiang Li, Meng Yang, Hui Qi, Don Jung, Mei Song, Mark Matteucci. In vitro and in vivo antitumor activity of TH3424: Preclinical rationale for a highly selective AKR1C3 prodrug for treating hepatocellular carcinomas. [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 1369.
    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 ...
  • 6
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2011
    In:  Cancer Research Vol. 71, No. 24 ( 2011-12-15), p. 7547-7557
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 71, No. 24 ( 2011-12-15), p. 7547-7557
    Abstract: Of the three envelope glycoproteins encoded by hepatitis B virus (HBV) that are collectively referred to as HBV surface antigen (HBsAg), the large HBsAg (LHBs) glycoprotein is expressed preferentially in HBV-associated hepatocellular carcinoma. LHBs can act as an oncogene in transgenic mice, but how it contributes functionally to hepatocarcinogenesis remains unclear. In this study, we determined the molecular and functional roles of LHBs during HBV-associated hepatocarcinogenesis. LHBs increased tumor formation of hepatoma cells. Moreover, expression of LHBs but not other HBV envelope glycoproteins specifically promoted proliferation of hepatoma and hepatic cells in vitro. Mechanistic investigations revealed that these effects were caused by activation of the Src/PI3K/Akt pathway through proximal stimulation of PKCα/Raf1 signaling by LHBs. Proliferation induced by stable LHBs expression was associated with increased G1–S cell-cycle progression and apoptosis resistance mediated by Src kinase activation, as established in hepatocellular carcinoma clinical specimens. Importantly, LHBs-induced cellular proliferation and tumor formation were reversed by administration of the Src inhibitor saracatinib. Together, our findings suggest that LHBs promotes tumorigenesis of hepatoma cells by triggering a PKCα/Raf1 to Src/PI3K/Akt signaling pathway, revealing novel insights into the underlying mechanisms of HBV-associated hepatocarcinogenesis. Cancer Res; 71(24); 7547–57. ©2011 AACR.
    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 ...
  • 7
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 18 ( 2020-09-15), p. 4004-4013
    Abstract: Registry-based epidemiologic studies suggest associations between chronic inflammatory intestinal diseases and pancreatic ductal adenocarcinoma (PDAC). As genetic susceptibility contributes to a large proportion of chronic inflammatory intestinal diseases, we hypothesize that the genomic regions surrounding established genome-wide associated variants for these chronic inflammatory diseases are associated with PDAC. We examined the association between PDAC and genomic regions (±500 kb) surrounding established common susceptibility variants for ulcerative colitis, Crohn's disease, inflammatory bowel disease, celiac disease, chronic pancreatitis, and primary sclerosing cholangitis. We analyzed summary statistics from genome-wide association studies data for 8,384 cases and 11,955 controls of European descent from two large consortium studies using the summary data-based adaptive rank truncated product method to examine the overall association of combined genomic regions for each inflammatory disease group. Combined genomic susceptibility regions for ulcerative colitis, Crohn disease, inflammatory bowel disease, and chronic pancreatitis were associated with PDAC at P values & lt; 0.05 (0.0040, 0.0057, 0.011, and 3.4 × 10−6, respectively). After excluding the 20 PDAC susceptibility regions (±500 kb) previously identified by GWAS, the genomic regions for ulcerative colitis, Crohn disease, and inflammatory bowel disease remained associated with PDAC (P = 0.0029, 0.0057, and 0.0098, respectively). Genomic regions for celiac disease (P = 0.22) and primary sclerosing cholangitis (P = 0.078) were not associated with PDAC. Our results support the hypothesis that genomic regions surrounding variants associated with inflammatory intestinal diseases, particularly, ulcerative colitis, Crohn disease, inflammatory bowel disease, and chronic pancreatitis are associated with PDAC. Significance: The joint effects of common variants in genomic regions containing susceptibility loci for inflammatory bowel disease and chronic pancreatitis are associated with PDAC and may provide insights to understanding pancreatic cancer etiology.
    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 ...
  • 8
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 4 ( 2017-02-15), p. 926-936
    Abstract: Aberrant expression of thioredoxin 1 (Trx1) plays an important role in cancer initiation and progression and has gained attention as an anticancer drug target. Here we report that the recently discovered natural diterpenoid isoforretin A (IsoA) significantly inhibits Trx1 activity and mediates anticancer effects in multiple preclinical settings. The inhibitory effect of IsoA was antagonized by free radical scavengers polyethylene glycol-catalase, polyethylene glycol superoxide dismutase, thiol-based antioxidants N-acetylcysteine and glutathione. Mass spectrometry analysis revealed that the mechanism of action was based on direct conjugation of IsoA to the Cys32/Cys35 residues of Trx1. This conjugation event attenuated reversible thiol reduction of Trx1, leading to ROS accumulation and a broader degradation of thiol redox homeostasis in cancer cells. Extending these in vitro findings, we documented that IsoA administration inhibited the growth of HepG2 tumors in a murine xenograft model of hepatocellular carcinoma. Taken together, our findings highlight IsoA as a potent bioactive inhibitor of Trx1 and a candidate anticancer natural product. Cancer Res; 77(4); 926–36. ©2016 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 ...
  • 9
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 72, No. 8_Supplement ( 2012-04-15), p. 3184-3184
    Abstract: Prostate cancer is the most commonly diagnosed malignancy among men in the United States. Due to an aging population, prostate cancer incidence has been increasing, with an estimated 200,000 men being diagnosed in 2010 and more than 32,000 deaths resulting from this disease. Better predictors of patient prognosis and treatment outcome are required to individualize prostate cancer treatment. High-throughput genomic sequence-based approaches offer a unique opportunity to identify biomarkers of disease-progression, thereby enabling more individualized therapy. The Canadian Prostate Cancer Genome Network (CPC-GENE) is an outcomes-based initiative that will sequence 500 specimens from 350 prostate cancer patients over a 5-year time span. Previously, whole genome sequencing efforts from biopsy specimens have been hindered by insufficient quantities of extracted DNA required as input for sequencing library construction. As a proof of concept to demonstrate the ability to sequence low input amounts of DNA from prostate biopsies, whole genome sequencing has been initiated for 50 prostate tumor biopsy samples along with their matched blood-derived reference sample. An on-bead sample preparation protocol was optimized using decreasing quantities of input DNA and used to construct sequencing libraries from as low as 100ng of DNA derived from macrodissected fresh frozen prostate biopsies ( & gt;70% cellularity). Sequencing is performed on the Illumina HiSeq 2000 platform to generate coverage depths of 50x for tumor samples and 30x for reference samples. Following alignment using NovoAlign and variant-calling using GATK, we compared our results to genotyping-array results generated using the Affymetrix OncoScan platform. Single-nucleotide variants detected using arrays were validated & gt;99% of the time by sequence data, confirming that the use of a low-input library did not hinder mutation detection. Sequencing does not exhibit significant genome-wide coverage biases, and CNV calls were compared between the genotyping arrays and the next-generation sequencing data. Outcomes from the sequencing and analysis of the initial 50 sample sets will similarly be applied over a 5-year period to characterize an additional 450 prostate specimens. The ability to whole genome sequence specimens where minimal amounts of extracted DNA exist presents new opportunities to sequence many samples previously deemed unusable, while also providing encouraging prospects for whole genome sequencing applications for future studies using biopsy specimens. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 3184. doi:1538-7445.AM2012-3184
    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 ...
  • 10
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2008
    In:  Cancer Research Vol. 68, No. 10 ( 2008-05-15), p. 3854-3862
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 68, No. 10 ( 2008-05-15), p. 3854-3862
    Abstract: Modest clinical outcomes of dendritic cell (DC) vaccine trials call for novel strategies. In this study, we have created a chimeric CD40 molecule that incorporates a single chain Fv (scFv) molecule specific for human ErbB2 antigen and fusing to the membrane spanning and cytosolic domains of murine CD40. After adenoviral transfer to bone marrow–derived DC, this chimeric receptor (CR) induced nuclear factor-κB (NF-κB)–dependent DC activation and effector function when cultured with immobilized ErbB2 protein or ErbB2-positive tumor cells in vitro. In vivo migration assays showed that ∼40% injected CR-modified DC (scFv-CD40-DC) effectively migrated to ErbB2-positive tumors, where they were activated after ErbB2 antigen stimulation, and sequentially homed into the draining lymph nodes. In murine ErbB2-positive D2F2/E2 breast tumor (BALB/c) and EL4/E2 thymoma (C57BL/6) models, i.v. injection of 1 × 106 scFv-CD40-DC significantly inhibited tumor growth and cured established tumors. Importantly, the cured mice treated by injection of scFv-CD40-DC were effective in preventing both ErbB2-positive and parental ErbB2-negative tumor rechallenge. Analysis of the underlying mechanism revealed that i.v. infusion of scFv-CD40-DC elicited tumor-specific CTL responses, and the transfer of CTLs from scFv-CD40-DC–treated mice protected naive mice against a subsequent tumor challenge. These results support the concept that genetic modification of DC with tumor-associated antigen-specific CD40 chimeric receptor might be a useful strategy for treatment of human cancers. [Cancer Res 2008;68(10):3854–62]
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2008
    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...