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)  (58)
  • 1
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2021
    In:  Cancer Research Vol. 81, No. 22_Supplement ( 2021-11-15), p. PO-065-PO-065
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 81, No. 22_Supplement ( 2021-11-15), p. PO-065-PO-065
    Abstract: SIWA318H is a novel monoclonal antibody that selectively targets a biomarker (an advanced glycation end product) found on the surface of cells exhibiting a combination of: (a) an abnormally high level of aerobic glycolysis, a process known to be associated with initiation and progression of cancer as well as metastasis; and (b) oxidative stress. Cells with this biomarker are often dysfunctional and associated with aging, cancer, and infectious diseases. In this study, we evaluated the antitumor activity of SIWA318H in a humanized xenograft model for pancreatic cancer. SIWA318H was found to react to pancreatic cancer cells (PSN1) and cancer associated fibroblasts (CAF08) using immunofluorescent staining. SIWA318H also showed strong reactivity with tumor xenografts derived from patients with pancreatic cancer as demonstrated by immunohistochemistry. Humanized CD34+ NSG mice were injected subcutaneously with PSN1 pancreatic cancer tumor cells. Ten days later, mice were randomized and enrolled in one of the following groups (n=16/group): 1) IgG1 isotype control (20mg/kg, BIWx1 followed by 10 mg/kg, BIWx2); 2) high dose SIWA318H (20mg/kg, BIWx1 followed by 10 mg/kg, BIWx2); and 3) low dose SIWA318H (10 mg/kg, BIWx1 followed by 5mg/kg, BIWx2). Tumors in mice treated with SIWA318H grew significantly slower than those in mice treated with isotype control antibody (p & lt;0.001). After 3 weeks of treatment with the high dose or the low dose of SIWA318H, the tumor growth was suppressed by 68.8% and 61.5%, respectively, when compared to the isotype antibody control (ANOVA p & lt;0.002). Moreover, a significant increase in complete remission (CR) rate was observed in mice receiving high (60%, p & lt;0.05) or low dose (77.8%, p & lt;0.02) of SIWA318H treatment compared with the control mice (6.7%). No significant antibody treatment related weight loss in mice was observed. These results provide compelling evidence that SIWA318H is a promising novel therapeutic for pancreatic cancer. Additional studies to better understand the mechanism of action of this novel immunotherapy agent are underway. Citation Format: Ashley Jensen, Gabriela R. Rossi, Ruben Muñoz, Kimberly Brothers, Lewis Gruber, Misty Gruber, Haiyong Han. SIWA318H, an advanced glycation end product (AGE) targeting antibody, is efficacious in a humanized mouse xenograft model for pancreatic cancer [abstract]. In: Proceedings of the AACR Virtual Special Conference on Pancreatic Cancer; 2021 Sep 29-30. Philadelphia (PA): AACR; Cancer Res 2021;81(22 Suppl):Abstract nr PO-065.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2016
    In:  Cancer Research Vol. 76, No. 4_Supplement ( 2016-02-15), p. PD7-01-PD7-01
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 4_Supplement ( 2016-02-15), p. PD7-01-PD7-01
    Abstract: Background: Emerging evidence demonstrates the effectiveness of targeted gene sequencing panels as a practical method for the diagnosis of inherited susceptibility to breast cancer. Sequencing of multiple high and moderate risk genes simultaneously accelerates the discovery of deleterious mutations (DM) or variants of unknown significance (VUS). However, a consequence of Multiplex Gene Panel (MGP) testing is the discovery of unexpected DMs in high or moderate risk genes other than BRCA1 or BRCA2 (BRCA1/2). The overall clinical utility and incremental gain of information conferred by MGP testing in hereditary cancer risk assessment is still unknown. Methods: We are conducting a multicenter prospective cohort study of patients undergoing cancer-risk assessment using a 25 gene sequencing panel, which includes APC, ATM, BARD1, BMPR1A, BRCA1, BRCA2, BRIP1, CDH1, CDK4, CDKN2A, CHEK2, EPCAM, MLH1, MSH2, MSH6, MUTYH, NBN, PALB2, PMS2, PTEN, RAD51C, RAD51D, SMAD4, STK11, and TP53. Patients were recruited from August 2014 to June 2015 at three medical centers. Patients are enrolled if they meet standard criteria for genetic testing or are predicted to have a ≥ 2.5% probability of inherited susceptibility to cancer calculated by validated risk prediction models. We present a planned interim analysis after enrolling 500 of 2000 total participants. Results: HCP testing was performed for 332 patients referred for clinical suspicion of hereditary breast and ovarian cancer (HBOC). In this cohort, 96.7% were female (n=321) and the mean age was 50 years (standard deviation, SD=12.2); race/ethnicity was 43.1% Hispanic (n=143), 37% Non-Hispanic White (n=123), 4.2% Black (n=14), 10.5% Asian (n=35), and 1.8% other (n=6). Among this cohort, 37 tested positive for one deleterious mutation (DM) (11.1%: 95% confidence interval (CI), 8.2% to 15%) and 118 patients carried at least one variant of uncertain significance (VUS) (35.5%: 95% CI, 30.6% to 69%). Excluding BRCA1 or BRCA2, 14 patients (4.3%: 95% CI, 2.6% to 7.2%) have a DM in ATM (n=3), CHEK2 (n=2), MSH6 (n=1), MUTYH (n=3), PALB2 (n=1), PMS2 (n=1), RAD51C (n=2), and TP53 (n=2). In a patient with an unexpected PMS2 mutation, enhanced cancer surveillance based on Lynch Syndrome guidelines was recommended. Among 160 patients with a history of invasive breast cancer or breast DCIS, 19 patients carried a DM (11.8 %: 95 CI, 7.7% to 17.8%). Conclusion: In this multicenter prospective cohort study among a diverse group of participants undergoing 25-gene MGP testing, 11.1% of participants tested positive for a DM. Among participants testing negative for BRCA1 and BRCA2, MGP testing identified DMs in 4.3% of participants prompting clinically appropriate risk reduction recommendations and enhanced cancer surveillance. Ongoing recruitment and long-term follow-up are in progress. Citation Format: Idos GE, Kurian AW, Mcdonnell KJ, Ricker CN, Sturgeon DY, Culver JO, Lowstuter K, Hartman A-R, Allen B, Teeter C-R, Kingham KE, Koff R, Lebensohn A, Chun NM, Mills MA, Petrovchich I, Hong C, Ladabaum U, Ford JM, Gruber SB. Interim analysis of multiplex gene panel testing for inherited susceptibility to breast cancer. [abstract]. In: Proceedings of the Thirty-Eighth Annual CTRC-AACR San Antonio Breast Cancer Symposium: 2015 Dec 8-12; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2016;76(4 Suppl):Abstract nr PD7-01.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Cancer Epidemiology, Biomarkers & Prevention, American Association for Cancer Research (AACR), Vol. 31, No. 5 ( 2022-05-04), p. 1077-1089
    Abstract: Currently known associations between common genetic variants and colorectal cancer explain less than half of its heritability of 25%. As alcohol consumption has a J-shape association with colorectal cancer risk, nondrinking and heavy drinking are both risk factors for colorectal cancer. Methods: Individual-level data was pooled from the Colon Cancer Family Registry, Colorectal Transdisciplinary Study, and Genetics and Epidemiology of Colorectal Cancer Consortium to compare nondrinkers (≤1 g/day) and heavy drinkers ( & gt;28 g/day) with light-to-moderate drinkers (1–28 g/day) in GxE analyses. To improve power, we implemented joint 2df and 3df tests and a novel two-step method that modifies the weighted hypothesis testing framework. We prioritized putative causal variants by predicting allelic effects using support vector machine models. Results: For nondrinking as compared with light-to-moderate drinking, the hybrid two-step approach identified 13 significant SNPs with pairwise r2 & gt; 0.9 in the 10q24.2/COX15 region. When stratified by alcohol intake, the A allele of lead SNP rs2300985 has a dose–response increase in risk of colorectal cancer as compared with the G allele in light-to-moderate drinkers [OR for GA genotype = 1.11; 95% confidence interval (CI), 1.06–1.17; OR for AA genotype = 1.22; 95% CI, 1.14–1.31], but not in nondrinkers or heavy drinkers. Among the correlated candidate SNPs in the 10q24.2/COX15 region, rs1318920 was predicted to disrupt an HNF4 transcription factor binding motif. Conclusions: Our study suggests that the association with colorectal cancer in 10q24.2/COX15 observed in genome-wide association study is strongest in nondrinkers. We also identified rs1318920 as the putative causal regulatory variant for the region. Impact: The study identifies multifaceted evidence of a possible functional effect for rs1318920.
    Type of Medium: Online Resource
    ISSN: 1055-9965 , 1538-7755
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    detail.hit.zdb_id: 2036781-8
    detail.hit.zdb_id: 1153420-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 15 ( 2023-08-01), p. 2572-2583
    Abstract: Colorectal cancer risk can be impacted by genetic, environmental, and lifestyle factors, including diet and obesity. Gene-environment interactions (G × E) can provide biological insights into the effects of obesity on colorectal cancer risk. Here, we assessed potential genome-wide G × E interactions between body mass index (BMI) and common SNPs for colorectal cancer risk using data from 36,415 colorectal cancer cases and 48,451 controls from three international colorectal cancer consortia (CCFR, CORECT, and GECCO). The G × E tests included the conventional logistic regression using multiplicative terms (one degree of freedom, 1DF test), the two-step EDGE method, and the joint 3DF test, each of which is powerful for detecting G × E interactions under specific conditions. BMI was associated with higher colorectal cancer risk. The two-step approach revealed a statistically significant G×BMI interaction located within the Formin 1/Gremlin 1 (FMN1/GREM1) gene region (rs58349661). This SNP was also identified by the 3DF test, with a suggestive statistical significance in the 1DF test. Among participants with the CC genotype of rs58349661, overweight and obesity categories were associated with higher colorectal cancer risk, whereas null associations were observed across BMI categories in those with the TT genotype. Using data from three large international consortia, this study discovered a locus in the FMN1/GREM1 gene region that interacts with BMI on the association with colorectal cancer risk. Further studies should examine the potential mechanisms through which this locus modifies the etiologic link between obesity and colorectal cancer. Significance: This gene-environment interaction analysis revealed a genetic locus in FMN1/GREM1 that interacts with body mass index in colorectal cancer risk, suggesting potential implications for precision prevention strategies.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: Cancer Epidemiology, Biomarkers & Prevention, American Association for Cancer Research (AACR), Vol. 32, No. 3 ( 2023-03-06), p. 315-328
    Abstract: Tobacco smoking is an established risk factor for colorectal cancer. However, genetically defined population subgroups may have increased susceptibility to smoking-related effects on colorectal cancer. Methods: A genome-wide interaction scan was performed including 33,756 colorectal cancer cases and 44,346 controls from three genetic consortia. Results: Evidence of an interaction was observed between smoking status (ever vs. never smokers) and a locus on 3p12.1 (rs9880919, P = 4.58 × 10−8), with higher associated risk in subjects carrying the GG genotype [OR, 1.25; 95% confidence interval (CI), 1.20–1.30] compared with the other genotypes (OR & lt;1.17 for GA and AA). Among ever smokers, we observed interactions between smoking intensity (increase in 10 cigarettes smoked per day) and two loci on 6p21.33 (rs4151657, P = 1.72 × 10−8) and 8q24.23 (rs7005722, P = 2.88 × 10−8). Subjects carrying the rs4151657 TT genotype showed higher risk (OR, 1.12; 95% CI, 1.09–1.16) compared with the other genotypes (OR & lt;1.06 for TC and CC). Similarly, higher risk was observed among subjects carrying the rs7005722 AA genotype (OR, 1.17; 95% CI, 1.07–1.28) compared with the other genotypes (OR & lt;1.13 for AC and CC). Functional annotation revealed that SNPs in 3p12.1 and 6p21.33 loci were located in regulatory regions, and were associated with expression levels of nearby genes. Genetic models predicting gene expression revealed that smoking parameters were associated with lower colorectal cancer risk with higher expression levels of CADM2 (3p12.1) and ATF6B (6p21.33). Conclusions: Our study identified novel genetic loci that may modulate the risk for colorectal cancer of smoking status and intensity, linked to tumor suppression and immune response. Impact: These findings can guide potential prevention treatments.
    Type of Medium: Online Resource
    ISSN: 1055-9965 , 1538-7755
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    detail.hit.zdb_id: 2036781-8
    detail.hit.zdb_id: 1153420-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 70, No. 24_Supplement ( 2010-12-15), p. P4-09-02-P4-09-02
    Abstract: Purpose: The aim of the present study was to assess the prognostic and predictive value of Ki67 labeling index in postmenopausal hormone receptor-positive early breast cancer patients who were treated with adjuvant tamoxifen or anastrozole after tamoxifen. Patients and Methods: We determined the expression of Ki67 by immunohistochemistry on whole tissue sections of breast carcinoma patients who had been enrolled in Austrian Breast and Colorectal Cancer Study Group (ABCSG) Trial 8 and received tamoxifen for 5 years or tamoxifen for 2 years followed by anastrozole for 3 years. Ki67 labeling index was evaluated as continuous variable or dichotomized at 10%. Distant recurrence and death were analyzed using Cox models adjusted for clinical and pathological factors. Results: High Ki67 labeling index was observed in 394 of 1587 (23%) tumors and was associated with poor outcome. Patients with high Ki67 labeling index had a significantly shorter distant recurrence-free survival (adjusted hazard ratio [HR] for distant recurrence 2.16, 95% confidence interval [CI] 1.43-3.25, P & lt; 0.001) and overall survival (adjusted HR for death 1.77, 95% CI 1.30-2.42, P & lt; 0.001) as compared to patients with low Ki67 labeling index. No interaction between Ki67 labeling index and treatment was observed (P = 0.84). Conclusion: High Ki67 labeling index is an independent poor prognostic factor for distant recurrence and death in postmenopausal women with early-stage, hormone receptor-positive breast cancer but is not predictive for outcome of adjuvant treatment with either tamoxifen or tamoxifen followed by anastrozole. Citation Information: Cancer Res 2010;70(24 Suppl):Abstract nr P4-09-02.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2010
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 14, No. 12_Supplement_2 ( 2015-12-01), p. C129-C129
    Abstract: While FGFR4 and its ligand FGF19 represent a promising target for cancers in which FGF19 is overexpressed or gene amplified, inhibition of this pathway carries a potential toxicity risk due to the role of FGFR4 in bile acid homeostasis. Treatment with an anti-FGF19 antibody caused significant toxicity in cynomolgus monkeys that was attributed to perturbations in the enterohepatic circulation of bile acids. Given that FGF19 signals through other FGFRs besides FGFR4, we hypothesized that the toxicity profile of an anti-FGFR4 antibody might be different than that of an anti-FGF19 antibody. To test this we set out to identify an anti-FGFR4 antibody that inhibits FGF19 binding and displays anti-tumor activity in models overexpressing FGF19, and then determine its toxicity profile. A phage screening effort yielded several anti-FGFR4 antibodies including H4, a high affinity fully human IgG1. H4 binds to the extracellular domain of FGFR4 (Kd = 12 pM) but does not cross react with FGFR1, FGFR2, or FGFR3. Furthermore, H4 efficiently blocks FGF19 binding to FGFR4 (IC50 = 589 pM) and inhibits FGF19-mediated downstream signaling in Hep3B2 cells. In human liver cancer cells that overexpress FGF19, H4 inhibits proliferation (IC50 250-1,100 nM). Anti-tumor activity was evaluated using cell line-derived xenograft tumors established from the FGF19 overexpressing and gene amplified cell lines HuH-7 and Hep3B2. H4 inhibited tumor growth in these models (T/C of 50% and 8%, respectively), and the antitumor effect was accompanied by alterations in several biomarkers associated with FGFR4 pathway inhibition including increased expression of CYP7A, the gene encoding the rate-limiting enzyme of bile acid synthesis. Finally, to assess the potential toxicity of H4 treatment, single dose intravenous toxicology studies were conducted in rats (20, 60, and 200 mg/kg) and cynomolgus monkeys (5, 20, and 100 mg/kg). Although H4 binds with similar affinity to monkey and rat FGFR4, the toxicology profiles were distinct. H4 was well tolerated in rats with no significant findings up to 200 mg/kg. Effects in monkeys included sporadic malformed feces, reduced food intake, elevated serum ALT (up to 44 fold over baseline) and AST activities, and elevated fecal bile acid concentrations at all doses. Hyperplasia of the gall bladder epithelium occurred at 100 mg/kg. There were no microscopic findings in the liver. Qualitatively this toxicity profile is similar to that of the anti-FGF19 antibody, although the maximum tolerated dose (MTD) of H4 in the monkey after a single dose was considered to be & gt;100 mg/kg. These data are relevant for drug development in Oncology in light of the efforts aimed at developing anti-FGFR4 antibodies and small molecule inhibitors specific for FGFR4. Citation Format: Timothy R. Mack, Colleen Burns Burns, Xuemei Guo, William John Feaver, Marie Prewett, Jennifer Gruber, Amelie Forest, Armando R. Irizarry, Ruslan Novosiadly, Nick Loizos, Andrew Dropsey. Antitumor efficacy and non-clinical safety of the high affinity anti-FGFR4 antibody H4: Implications for targeting the FGF19-FGFR4 axis in oncology. [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2015 Nov 5-9; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2015;14(12 Suppl 2):Abstract nr C129.
    Type of Medium: Online Resource
    ISSN: 1535-7163 , 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2015
    detail.hit.zdb_id: 2062135-8
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 17 ( 2016-09-01), p. 5103-5114
    Abstract: Identifying genetic variants with pleiotropic associations can uncover common pathways influencing multiple cancers. We took a two-stage approach to conduct genome-wide association studies for lung, ovary, breast, prostate, and colorectal cancer from the GAME-ON/GECCO Network (61,851 cases, 61,820 controls) to identify pleiotropic loci. Findings were replicated in independent association studies (55,789 cases, 330,490 controls). We identified a novel pleiotropic association at 1q22 involving breast and lung squamous cell carcinoma, with eQTL analysis showing an association with ADAM15/THBS3 gene expression in lung. We also identified a known breast cancer locus CASP8/ALS2CR12 associated with prostate cancer, a known cancer locus at CDKN2B-AS1 with different variants associated with lung adenocarcinoma and prostate cancer, and confirmed the associations of a breast BRCA2 locus with lung and serous ovarian cancer. This is the largest study to date examining pleiotropy across multiple cancer-associated loci, identifying common mechanisms of cancer development and progression. Cancer Res; 76(17); 5103–14. ©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: 2016
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 13_Supplement ( 2017-07-01), p. 1304-1304
    Abstract: Studies that estimate complex disease heritability based on genome-wide common SNP array data have shown that a large fraction of heritability is contributed from variants that do not reach genome-wide significance at current genome-wide association study (GWAS) sample sizes. The contribution of rare variants to heritability has yet to be explored for many complex diseases. Despite the decreasing cost of sequencing, it still remains prohibitively expensive to sequence sufficient samples for well-powered genetic association studies of rare variants. However, with increasingly large and denser imputation reference panels it has become feasible to accurately impute variants with minor allele frequencies (MAFs) as low as 0.1%, enabling study of a subset of rare risk variants. In previous work, we used restricted maximum likelihood (REML) to estimate the total additive heritability of colorectal cancer (CRC) based on common SNP array genotypes. Here, we expand this work using imputed genotype data and a larger sample size. We performed whole-genome sequencing of 1,961 CRC cases and 981 controls, and subsequently imputed these haplotypes into 11,895 unrelated CRC cases and 14,659 unrelated controls that are part of the Colorectal Cancer Family Registry (CCFR) and the Genetics and Epidemiology of Colorectal Cancer Consortium (GECCO). We estimated heritability from individual-level imputed genotype data using LD- and MAF-stratified GREML, as implemented in GCTA. In total, we analyzed 17,649,167 imputed genetic variants with minor allele count & gt;3. Published heritability estimates for CRC from family-based studies vary from 12% to 35%. Based on common genotyped SNPs, we previously estimated heritability to be 7.42% (95% CI: 4.71-10.12%) on the underlying liability scale, assuming a population prevalence of 0.004. For imputed genotypes, we estimate the total heritability to be 12.0% (95% CI: 9.65-14.35). Using a likelihood ratio test, we demonstrate a significant contribution of variants with MAF ≤1% to CRC genetic risk (P=0.003). Because of the imperfect imputation accuracy for very rare variants, their contribution is likely higher. These results suggest that with additional sequencing, improved imputation accuracy, and larger GWAS, we should expect to start discovering rare variant associations for CRC risk. Citation Format: Jeroen R. Huyghe, Sai Chen, Hyun M. Kang, Tabitha Harrison, Sonja I. Berndt, Stephane Bézieau, Hermann Brenner, Graham Casey, Andrew T. Chan, Jenny Chang-Claude, Steven J. Gallinger, Stephen B. Gruber, Andrea Gsur, Michael Hoffmeister, Thomas Hudson, Mark A. Jenkins, Loic Le Marchand, Polly A. Newcomb, John D. Potter, Conghui Qu, Martha L. Slattery, Joshua D. Smith, Emily White, Goncalo R. Abecasis, Li Hsu, Deborah A. Nickerson, Ulrike Peters, on behalf of CCFR and GECCO. The contribution of rare and low-frequency variants to colorectal cancer heritability [abstract]. In: Proceedings of the American Association for C ancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 1304. doi:10.1158/1538-7445.AM2017-1304
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    In: Cancer Epidemiology, Biomarkers & Prevention, American Association for Cancer Research (AACR), Vol. 26, No. 9 ( 2017-09-01), p. 1427-1435
    Abstract: Background: Many cancers share specific genetic risk factors, including both rare high-penetrance mutations and common SNPs identified through genome-wide association studies (GWAS). However, little is known about the overall shared heritability across cancers. Quantifying the extent to which two distinct cancers share genetic origin will give insights to shared biological mechanisms underlying cancer and inform design for future genetic association studies. Methods: In this study, we estimated the pair-wise genetic correlation between six cancer types (breast, colorectal, lung, ovarian, pancreatic, and prostate) using cancer-specific GWAS summary statistics data based on 66,958 case and 70,665 control subjects of European ancestry. We also estimated genetic correlations between cancers and 14 noncancer diseases and traits. Results: After adjusting for 15 pair-wise genetic correlation tests between cancers, we found significant (P & lt; 0.003) genetic correlations between pancreatic and colorectal cancer (rg = 0.55, P = 0.003), lung and colorectal cancer (rg = 0.31, P = 0.001). We also found suggestive genetic correlations between lung and breast cancer (rg = 0.27, P = 0.009), and colorectal and breast cancer (rg = 0.22, P = 0.01). In contrast, we found no evidence that prostate cancer shared an appreciable proportion of heritability with other cancers. After adjusting for 84 tests studying genetic correlations between cancer types and other traits (Bonferroni-corrected P value: 0.0006), only the genetic correlation between lung cancer and smoking remained significant (rg = 0.41, P = 1.03 × 10−6). We also observed nominally significant genetic correlations between body mass index and all cancers except ovarian cancer. Conclusions: Our results highlight novel genetic correlations and lend support to previous observational studies that have observed links between cancers and risk factors. Impact: This study demonstrates modest genetic correlations between cancers; in particular, breast, colorectal, and lung cancer share some degree of genetic basis. Cancer Epidemiol Biomarkers Prev; 26(9); 1427–35. ©2017 AACR.
    Type of Medium: Online Resource
    ISSN: 1055-9965 , 1538-7755
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2017
    detail.hit.zdb_id: 2036781-8
    detail.hit.zdb_id: 1153420-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages