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)  (40)
  • 1
    In: Cancer Immunology Research, American Association for Cancer Research (AACR), Vol. 3, No. 3 ( 2015-03-01), p. 288-295
    Abstract: Activating NRAS mutations are found in 15% to 20% of melanomas. Immune therapies have become a mainstay in advanced melanoma treatment. We sought to evaluate whether tumor genotype (e.g., NRAS mutations) correlates with benefit from immune therapy in melanoma. We identified 229 patients with melanoma treated with immune therapies [IL2, ipilimumab, or anti-programmed cell death-1/ligand-1 (PD-1/PD-L1)] at three centers and compared clinical outcomes following immune therapy for patients with or without NRAS mutations. Of the 229 patients with melanoma, 60 had NRAS mutation, 53 had BRAF mutation, and 116 had NRAS/BRAF wild type. The NRAS-mutant cohort had superior or a trend to superior outcomes compared with the other cohorts in terms of response to first-line immune therapy (28% vs. 16%, P = 0.04), response to any line of immune therapy (32% vs. 20%, P = 0.07), clinical benefit (response + stable disease lasting ≥24 weeks; 50% vs. 31%, P & lt; 0.01), and progression-free survival (median, 4.1 vs. 2.9 months, P = 0.09). Benefit from anti–PD-1/PD-L1 was particularly marked in the NRAS cohort (clinical benefit rate 73% vs. 35%). In an independent group of patient samples, NRAS-mutant melanoma had higher PD-L1 expression (although not statistically significant) compared with other genotypes (8/12 vs. 9/20 samples with ≥1% expression; 6/12 vs. 6/20 samples with ≥5% expression), suggesting a potential mechanism for the clinical results. This retrospective study suggests that NRAS mutations in advanced melanoma correlate with increased benefit from immune-based therapies compared with other genetic subtypes. If confirmed by prospective studies, this may be explained in part by high rates of PD-L1 expression. Cancer Immunol Res; 3(3); 288–95. ©2015 AACR.
    Type of Medium: Online Resource
    ISSN: 2326-6066 , 2326-6074
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2015
    detail.hit.zdb_id: 2732517-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 74, No. 19_Supplement ( 2014-10-01), p. 2942-2942
    Abstract: Background: Immune-based therapies are playing an expanding role in the treatment of advanced melanoma. Reliable biomarkers to predict benefit from these agents have not yet been identified. Specifically, the relationship between melanoma genotype and immune-based therapy has not been well characterized. Approximately 15-20% of patients with melanoma harbor an NRAS mutation in their tumor tissue. We assessed whether NRAS mutations impact the response to immune-based therapy in patients with advanced melanoma. Methods: The current study involved 171 patients from three institutions with advanced melanoma and documented tumor genotype. All patients were treated with immune-based therapy (defined as IL-2, anti-CTLA-4, or anti-PD1/PD-L1 inhibitors). We compared the rates of clinical response between patients with NRAS mutant melanoma and those without NRAS or BRAF mutations (wild type, WT). The primary study end-point was complete or partial response (CR/PR); secondary end-points included clinical benefit (CB; defined as CR/PR or stable disease for & gt;24 weeks), progression-free survival (PFS), and overall survival (OS). Results: In our study cohort, 59 patients (35%) had NRAS-mutant metastatic melanoma and 112 patients (65%) had WT metastatic melanoma. We observed a significantly higher rate of CR/PR and CB in patients with NRAS-mutant melanoma treated with immune-based therapy compared to patients with WT melanoma (32% vs 18%, p=0.04, for CR/PR; 49% vs 29%, p=0.01, for CB). Within specific immune-therapy types, patients with NRAS-mutant melanoma experienced the greatest benefit in CR/PR compared to patients with WT melanoma when treated with anti-PD1/PD-L1 agents (70% vs 20%, p=0.01). No statistically significant differences in median PFS (4.1 months for NRAS-mutant vs 3.3 months for WT, p=0.22) or median OS (14.6 months for NRAS-mutant vs 15.4 months for WT, p=0.63) were observed. Conclusions: Patients with NRAS-mutant metastatic melanoma experience a higher rate of CR/PR and CB when treated with immune-based therapy compared to patients with WT metastatic melanoma. The underlying mechanism for this finding merits further exploration, and these data suggest that routine assessment for NRAS mutations in patients with melanoma is warranted. Citation Format: Wade T. Iams, Douglas B. Johnson, Marisa Flavin, Katherine Panageas, Gregory D. Ayers, Zhiguo Zhao, Anthony John Iafrate, Ryan J. Sullivan, Richard D. Carvajal, Jeffrey A. Sosman, Christine M. Lovly. NRAS mutation as a predictor of response to immune-based therapies in patients with metastatic melanoma. [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 2942. doi:10.1158/1538-7445.AM2014-2942
    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 ...
  • 3
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 22, No. 1_Supplement ( 2016-01-01), p. 14-14
    Abstract: Noninvasive detection of urine tumor DNA (utDNA) has the potential to augment the diagnosis and management of bladder cancer. We sought to identify whether patient specific molecular events identified initially in the primary tumor could be detected in their utDNA. We interrogated both genetic and epigenetic events by a number of different technologies. Single nucleotide variants (SNVs) were analyzed by Next Generation Sequencing (NGS) on both the Ion Torrent platform (using amplicon enrichment) and the Illumina platform (via hybridization capture). Thirteen differentially methylated regions (DMRs) in the genome were analyzed by methylation-specific quantitative polymerase chain reaction (MS-qPCR). The cohort included patients with newly diagnosed NMIBC and those undergoing surveillance cystoscopy following a prior diagnosis of NMIBC. DNA was isolated from fixed paraffin embedded (FPE) primary tumors, matched blood buffy coat (BC), urine sediment (US) and, in some cases, clarified urine (CU-a.k.a. urine supernatant). Tumor specific molecular events were identified by comparing primary tumor tissue to BC (for SNVs with NGS) or to pooled data from urine DNA from healthy controls (for DMRs with MS-qPCR). Initially, 8 patients were evaluated with all three technologies. A larger cohort of 66 patients undergoing surveillance for NMIBC was then analyzed using MS-qPCR and a subset was also analyzed using NGS on the Ion Torrent platform. Tumor specific DMRs and/or SNVs were detected in tumor tissue from 73 of 74 patients. In the 8 patients assessed using both NGS platforms, SNVs were found in the primary tumor for 8/8 (Ion Torrent) and 6/8 (Illumina) patients, and both NGS platforms identified utDNA in 4 of 5 biopsy confirmed NMIBC patients. In patients with sufficient DNA to be assessed with all three technologies, in both US and CU, there was 100% agreement for all positive/negative calls for utDNA. Targeted genomic regions methylated in NMIBC tissue were seen to be methylated in the corresponding utDNA at the time of diagnosis or recurrence. In the cohort of 66 patients undergoing surveillance for NMIBC using MS-qPCR, a negative predictive value for high risk recurrences of 95% (with a prevalence of 10%) was achievable. These results provide evidence that both genetic and epigenetic events are evident in utDNA and reflect the presence and genetics of the NMIBC. This proof of concept study demonstrates the potential for a personalized non-invasive means to detect and monitor bladder cancer, thus making a valuable contribution to the field of precision medicine. Citation Format: Matthew J. Resnick, Margarita Lopatin, Neal D. Shore, Po N. Lam, Brian Helfand, Richard D. Abramson, Michael Crager, Michael Bonham, Haluk Tezcan, Kim M. Clark-Langone, Chris Silk, Seth Michelson, Gregory Alexander, Phillip G. Febbo. Analysis of tumor DNA in urine as a highly sensitive liquid biopsy for patients with non-muscle invasive bladder cancer (NMIBC). [abstract]. In: Proceedings of the AACR Precision Medicine Series: Integrating Clinical Genomics and Cancer Therapy; Jun 13-16, 2015; Salt Lake City, UT. Philadelphia (PA): AACR; Clin Cancer Res 2016;22(1_Suppl):Abstract nr 14.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 9_Supplement ( 2015-05-01), p. P4-02-08-P4-02-08
    Abstract: Background: Analytically and clinically validated non-invasive blood tests that quantify breast cancer burden and clinical drug response/resistance are greatly needed. Many groups have successfully detected tumor markers in blood using a variety of technologies, including next generation sequencing (NGS). We performed a comprehensive NGS study on a small number of patients to evaluate the value of global versus individual markers for the quantitation of tumor-derived cell free DNA (cfDNA) in plasma. Methods: DNA isolated from formalin-fixed primary tumor, buffy coat cells, and plasma from 2 patients with metastatic breast cancer were characterized simultaneously for copy number aberrations (CNAs) and differentially methylated regions (DMRs) using whole genome bisulfite sequencing (WBGS), and targeted sequencing-based genotyping of 346 cancer-associated single nucleotide variations (SNVs). CNA and DMR regions were identified from log normalized, GC content corrected counts and DMR data using Poisson and binomial distribution theory and false discovery rate controlling methods. Percent tumor in cfDNA was estimated from the normalized ratio (plasma: primary tumor) of CNA or DMR compared to buffy coat, aggregating over genomic regions. Sample sets from 8 non-metastatic patients were also profiled using the targeted SNV panel in order to compare SNVs between samples and estimate percent tumor cfDNA. Results: WGBS detected tumor specific alterations in each primary tumor compared to buffy coat. By analyzing the genome using 100 Kb bins, we observed over 1000 bins with detectable CNA signal and, among 56 million CpG sites, over 30,000 DMRs. As expected, 5 or fewer informative somatic SNVs were detected in each patient. Analysis of these somatic changes in plasma revealed that the tumor fraction estimated from SNV detected in cfDNA varied widely between sites originally discovered in the patient’s primary tumor. In contrast, similar estimates of tumor fraction in cfDNA were obtained using CNA and DMR profiles within each patient; both methods yielded similar estimates of over 50% in one patient and less than 10% in the other. For the patient with high tumor fraction, both CNA and DMR profiles contained examples of individual large genomic regions that displayed additional clear aberrations in the plasma compared to the original tumor, such as a striking loss of a & gt;25 Mb region of chromosome 4. Conclusions: Although individual somatic SNV in cfDNA can be detected in metastatic disease, calculated allelic fraction based on individual SNVs varies greatly within the same patient. Measuring and integrating CNA or DMR across the genome provided more consistent and reliable estimates of tumor DNA fraction in plasma, and also revealed alterations in plasma from patients with metastatic disease that were not prominent in the primary tumor. Citation Format: Ellen M Beasley, Richard D Abramson, Gregory E Alexander, David Chan, Kristen Bradley, Francois Collin, Michael Crager, Andrew Dei Rossi, Joseph Dorado, Adam Friedman, William J Gibb, Jennie Jeong, Col Jones, C J Ku, Yan Ma, John Morlan, Kunbin Qu, Aibing Rao, Aaron Scott, Haluk Tezcan. Global quantitative measures using next-generation sequencing for breast cancer presence outperform individual tumor markers in plasma [abstract]. In: Proceedings of the Thirty-Seventh Annual CTRC-AACR San Antonio Breast Cancer Symposium: 2014 Dec 9-13; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2015;75(9 Suppl):Abstract nr P4-02-08.
    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 ...
  • 5
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 18, No. 12 ( 2019-12-01), p. 2207-2219
    Abstract: Although Aurora A, B, and C kinases share high sequence similarity, especially within the kinase domain, they function distinctly in cell-cycle progression. Aurora A depletion primarily leads to mitotic spindle formation defects and consequently prometaphase arrest, whereas Aurora B/C inactivation primarily induces polyploidy from cytokinesis failure. Aurora B/C inactivation phenotypes are also epistatic to those of Aurora A, such that the concomitant inactivation of Aurora A and B, or all Aurora isoforms by nonisoform–selective Aurora inhibitors, demonstrates the Aurora B/C-dominant cytokinesis failure and polyploidy phenotypes. Several Aurora inhibitors are in clinical trials for T/B-cell lymphoma, multiple myeloma, leukemia, lung, and breast cancers. Here, we describe an Aurora A–selective inhibitor, LY3295668, which potently inhibits Aurora autophosphorylation and its kinase activity in vitro and in vivo, persistently arrests cancer cells in mitosis, and induces more profound apoptosis than Aurora B or Aurora A/B dual inhibitors without Aurora B inhibition–associated cytokinesis failure and aneuploidy. LY3295668 inhibits the growth of a broad panel of cancer cell lines, including small-cell lung and breast cancer cells. It demonstrates significant efficacy in small-cell lung cancer xenograft and patient-derived tumor preclinical models as a single agent and in combination with standard-of-care agents. LY3295668, as a highly Aurora A–selective inhibitor, may represent a preferred approach to the current pan-Aurora inhibitors as a cancer therapeutic agent.
    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 ...
  • 6
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. 5789-5789
    Abstract: Background: The bromodomains and extra-terminal domain (BET) proteins are a family of 4 adapter proteins, BRD2, BRD3, BRD4, and BRDT, that bind to specific acetylated lysine residues on the histone tails of chromatin and recruit additional proteins to regulate gene transcription. The c-MYC oncogene, which is amplified and deregulated in 40% to 70% of all cancers, is directly regulated by BET proteins. Preclinical studies provide a strong rationale for pursuing transcriptional regulation via BET inhibition in cancer treatment (Lenhart, et al. Mol Cancer Ther. 2015;14:2167-2174; Filippakopoulos, et al. Nature. 2010;468:1067-1073). Here, we present results of crystal structure-guided structure-activity relationship (SAR) studies that resulted in the identification of BMS-986158, a highly potent BET inhibitor. Methods: Using fluorescence resonance energy transfer (FRET), we screened a library of compounds and identified a carbazole series of BET inhibitors. Alkylation of the carbazole nitrogen resulted in a 10-fold boost in potency against BET. We then created a differently oriented carbazole series and, subsequently, a carboline series of compounds to improve potency and pharmaceutical properties. A thermal shift assay was used to evaluate selectivity for binding to the BET family of bromodomains. Results: Crystal structure and subsequent SAR studies demonstrated that the isoxazole moiety formed critical interactions with the BET bromodomains. Lead compounds demonstrated potent binding to BRD4 and reduction in c-MYC expression and proliferation in cell lines such as KMS-11. Accessing a second lipophilic pocket in the BRD4 binding site increased potency significantly. Modification of the lead series from a carbazole carboxamide to a carboline resulted in significant improvement in pharmaceutical properties and led to the identification of BMS-986158, which demonstrated in vitro and in vivo potency against a variety of tumor types. In c-MYC-driven cancer cell lines, BMS-986158 caused dose-dependent downregulation of c-MYC expression and induced cancer cell death. BMS-986158 demonstrated & gt; 70% tumor growth inhibition at tolerated doses in patient-derived xenograft models (lung, colorectal, and triple-negative breast cancers). Antitumor activity in mice and pharmacokinetic properties in animal studies support oral dosing in humans. Conclusions: Structure-based drug design led to the discovery of BMS-986158, a highly potent BET inhibitor. With promising antitumor activity in preclinical studies, BMS-986158 is currently being evaluated in a phase 1/2a clinical trial in patients with advanced cancers. Citation Format: Ashvinikumar V. Gavai, Derek Norris, David Tortolani, Daniel O'Malley, Yufen Zhao, Claude Quesnelle, Patrice Gill, Wayne Vaccaro, Tram Huynh, Vijay Ahuja, Dharmpal Dodd, Christopher Mussari, Lalgudi Harikrishnan, Muthoni Kamau, John S. Tokarski, Steven Sheriff, Richard Rampulla, Dauh-Rurng Wu, Jianqing Li, Huiping Zhang, Peng Li, Dawn Sun, Henry Yip, Yingru Zhang, Arvind Mathur, Haiying Zhang, Christine Huang, Zheng Yang, Asoka Ranasinghe, Celia D'Arienzo, Ching Su, Gerry Everlof, Lisa Zhang, Nirmala Raghavan, John T. Hunt, Michael Poss, Gregory D. Vite, Richard A. Westhouse, Susan Wee. Discovery of clinical candidate BMS-986158, an oral BET inhibitor, for the treatment of cancer [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 5789.
    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 ...
  • 7
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 15, No. 22 ( 2009-11-15), p. 6891-6900
    Abstract: Purpose: Deregulation of the Janus kinase-signal transducers and activators of transcription (JAK-STAT) pathway is a hallmark for the Philadelphia chromosomenegative myeloproliferative diseases polycythemia vera, essential thrombocythemia, and primary myelofibrosis. We tested the efficacy of a selective JAK1/2 inhibitor in cellular and in vivo models of JAK2-driven malignancy. Experimental Design: A novel inhibitor of JAK1/2 was characterized using kinase assays. Cellular effects of this compound were measured in cell lines bearing the JAK2V617F or JAK1V658F mutation, and its antiproliferative activity against primary polycythemiavera patient cells was determined using clonogenic assays. Antineoplastic activity in vivo was determined using a JAK2V617F-driven xenograft model, and effects of the compound on survival, organomegaly, body weight, and disease-associated inflammatory markers were measured. Results: INCB16562 potently inhibited proliferation of cell lines and primary cells from PV patients carrying the JAK2V617F or JAK1V658F mutation by blocking JAK-STAT signaling and inducing apoptosis. In vivo, INCB16562 reduced malignant cell burden, reversed splenomegaly and normalized splenic architecture, improved body weight gains, and extended survival in a model of JAK2V617F-driven hematologic malignancy. Moreover, these mice suffered from markedly elevated levels of inflammatory cytokines, similar to advanced myeloproliferative disease patients, which was reversed upon treatment. Conclusions: These data showed that administration of the dual JAK1/2 inhibitor INCB16562 reduces malignant cell burden, normalizes spleen size and architecture, suppresses inflammatory cytokines, improves weight gain, and extends survival in a rodent model of JAK2V617F-driven hematologic malignancy. Thus, selective inhibitors of JAK1 and JAK2 represent a novel therapy for the patients with myeloproliferative diseases and other neoplasms associated with JAK dysregulation. (Clin Cancer Res 2009;15(22):6891900)
    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 ...
  • 8
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 18, No. 24 ( 2012-12-15), p. 6758-6770
    Abstract: Purpose: Adoptive cell therapy (ACT) using autologous tumor-infiltrating lymphocytes (TIL) is a promising treatment for metastatic melanoma unresponsive to conventional therapies. We report here on the results of an ongoing phase II clinical trial testing the efficacy of ACT using TIL in patients with metastatic melanoma and the association of specific patient clinical characteristics and the phenotypic attributes of the infused TIL with clinical response. Experimental Design: Altogether, 31 transiently lymphodepleted patients were treated with their expanded TIL, followed by two cycles of high-dose interleukin (IL)-2 therapy. The effects of patient clinical features and the phenotypes of the T cells infused on the clinical response were determined. Results: Overall, 15 of 31 (48.4%) patients had an objective clinical response using immune-related response criteria (irRC) with 2 patients (6.5%) having a complete response. Progression-free survival of more than 12 months was observed for 9 of 15 (60%) of the responding patients. Factors significantly associated with the objective tumor regression included a higher number of TIL infused, a higher proportion of CD8+ T cells in the infusion product, a more differentiated effector phenotype of the CD8+ population, and a higher frequency of CD8+ T cells coexpressing the negative costimulation molecule “B- and T-lymphocyte attenuator” (BTLA). No significant difference in the telomere lengths of TIL between responders and nonresponders was identified. Conclusion: These results indicate that the immunotherapy with expanded autologous TIL is capable of achieving durable clinical responses in patients with metastatic melanoma and that CD8+ T cells in the infused TIL, particularly differentiated effectors cells and cells expressing BTLA, are associated with tumor regression. Clin Cancer Res; 18(24); 6758–70. ©2012 AACR.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2012
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 73, No. 8_Supplement ( 2013-04-15), p. 3626-3626
    Abstract: Definitive radiotherapy combined with chemotherapy is associated with enhanced locoregional control and clinical outcomes in non-small cell lung cancer (NSCLC) patients who are not candidates for surgery. However, radiation-induced pneumonitis and esophagitis are common dose-limiting inflammatory conditions that occur in these patients. These events can compromise the effect of the overall treatment and reduce quality of life in these patients. It is not clear what factors contribute to differences in the development of toxicities among patients with same clinical or dosimetric parameters. In this study, we conducted a large scale pathway-based analysis to identify inflammation-related genetic variants associated with radiation-induced pneumonitis or esophagitis. In the initial discovery phase, 11,930 inflammation-related genetic variants were genotyped in 243 stage I-III NSCLC patients who were treated with definitive radiotherapy at MD Anderson Cancer Center. A fast-track validation was performed for the top candidate SNPs in an additional 212 NSCLC cases following the same criteria. A total of 14 SNPs and 12 SNPs were validated for pneumonitis and esophagitis, respectively. Meta-analysis was used to summarize the results from two phases. The most significant SNPs for esophagitis and pneumonitis mapped to chromosome 18p (OR=9.05, 95%CI=2.68-30.59, P-meta=3.90×10−4) and 14q (OR=4.75, 95%CI=2.17-10.41, P-meta =9.61×10−5), respectively. To explore the potential underlying mechanisms, we analyzed radiation sensitivity and eQTL relationships of the identified SNPs and their host genes in 277 ethnically defined human lymphoblastoid cell lines. Among the 24 host genes for the 26 identified SNPs, three genes (DDX58, GZMB, CDC2) showed significant association with radiation responses in these cell lines (P & lt;0.05). Moreover, a significant eQTL relationship was identified between CDC2 expression and the genotype of the identified CDC2 SNP (P=0.022). Interestingly, this CDC2 SNP is predicted to create a new miRNA binding site. Luciferase reporter assays to test the influence of this SNP on miRNA regulation is currently ongoing. Our results provide evidence that inflammation-related genetic variants contribute to the development of radiation-induced pneumonitis and esophagitis. The results also suggest that gene expression changes through altered miRNA binding may be an underlying mechanism for these events. Citation Format: Xia Pu, Liewei Wang, Michelle AT Hildebrandt, Yuanqing Ye, Joe Y. Chang, Charles Lu, Heath D. Skinner, Nifang Niu, Gregory D. Jenkins, Ritsuko Komaki, John D. Minna, Jack A. Roth, Richard M. Weinshilboum, Xifeng Wu. Inflammation-related genetic variants predict radiation-induced toxicity following definitive radiotherapy for non-small cell lung cancer. [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 3626. doi:10.1158/1538-7445.AM2013-3626
    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 ...
  • 10
    In: Cancer Discovery, American Association for Cancer Research (AACR), Vol. 3, No. 7 ( 2013-07-01), p. 812-825
    Abstract: The selective estrogen receptor modulators (SERM) tamoxifen and raloxifene can reduce the occurrence of breast cancer in high-risk women by 50%, but this U.S. Food and Drug Administration-approved prevention therapy is not often used. We attempted to identify genetic factors that contribute to variation in SERM breast cancer prevention, using DNA from the NSABP P-1 and P-2 breast cancer prevention trials. An initial discovery genome-wide association study identified common single-nucleotide polymorphisms (SNP) in or near the ZNF423 and CTSO genes that were associated with breast cancer risk during SERM therapy. We then showed that both ZNF423 and CTSO participated in the estrogen-dependent induction of BRCA1 expression, in both cases with SNP-dependent variation in induction. ZNF423 appeared to be an estrogen-inducible BRCA1 transcription factor. The OR for differences in breast cancer risk during SERM therapy for subjects homozygous for both protective or both risk alleles for ZNF423 and CTSO was 5.71. Significance: This study identified novel, functionally polymorphic genes involved in the estrogen-dependent regulation of BRCA1 expression, as well as a novel mechanism for genetic variation in SERM therapeutic effect. These observations, and definition of their underlying mechanisms, represent steps toward pharmacogenomically individualized SERM breast cancer prevention. Cancer Discov; 3(7); 812–25. ©2013 AACR. See related commentary by Machiela and Chanock, p. 728 This article is highlighted in the In This Issue feature, p. 705
    Type of Medium: Online Resource
    ISSN: 2159-8274 , 2159-8290
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2013
    detail.hit.zdb_id: 2607892-2
    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...