447 research outputs found

    BRCA 1/2-Mutation Related and Sporadic Breast and Ovarian Cancers: More Alike than Different

    Get PDF
    No longer is histology solely predictive of cancer treatment and outcome. There is an increasing influence of tumor genomic characteristics on therapeutic options. Both breast and ovarian cancers are at higher risk of development in patients with BRCA 1/2-germline mutations. Recent data from the Cancer Genome Atlas (TCGA) and others have shown a number of genomic similarities between triple negative breast cancers and ovarian cancers. Recently, poly (ADP-ribose) polymerase (PARP) inhibitors have shown promising activity in hereditary BRCA 1/2-mutated and sporadic breast and ovarian cancers. In this review, we will summarize the current literature regarding the genomic and phenotypic similarities between BRCA 1/2-mutation related cancers, sporadic triple negative breast cancers, and sporadic ovarian cancers. We will also review phase I, II, and III data using PARP inhibitors for these malignancies and compare and contrast the results with respect to histology

    NCI-MATCH Arms N & P: Phase II study of PI3K beta inhibitor GSK2636771 in patients (pts) with cancers (ca) with PTEN mutation/deletion (mut/del) or PTEN protein loss

    Get PDF
    Background: The NCI-MATCH trial is the largest national study (1173 sites) for ptswith relapsed/ refractory solid tumors, lymphomas and myeloma, which assigns tar-geted therapies based on individual tumor molecular alterations detected using theadapted Oncomine AmpliSeq panel (143 genes) and immunohistochemistry (IHC).We hypothesized that patients with PTEN-deficient cancers enrolled to Arms N and Pmay benefit from treatment with the PI3K beta-selective inhibitor GSK2636771. Methods: Eligibility: relapsed/refractory ca, good end-organ function, and ECOG PS ≤ 1. Pts were screened for molecular alterations by centralized testing on fresh tumor biopsy and had deleterious PTEN mut/del without loss of expression (Arm N) or complete loss of cytoplasmic and nuclear PTEN staining on IHC (Arm P), and no other aberrations activating the PI3K/MTOR and MAPK pathways (mut in PIK3CA, PIK3R1, BRAF, KRAS, AKT1, TSC1/2, mTOR, RHEB, NF2, NRAS, HRAS). Pts received GSK2636771 400mg/day (28-days cycles). RECIST 1.1 overall response rate (ORR) was the primary endpoint. Results: Of 59 enrolled pts, 56 were eligible and received treatment. Of 22 pts with PTEN mut/del (Arm N: 6 uterine, 2 breast, 2 prostate, 2 head/neck ca, 10 other), all are off treatment as of analysis (14 disease progression, 4 for adverse events [AEs], 4 other). One pt (4.5%) with prostate ca (PTEN deletion, MPRSS2-ERG fusion) attained a partial response (-42%). Of 7 (32%) pts with stable disease (SD), 2 had SD \u3e 6 months (uterine leiomyosarcoma; endometrial carcinoma). Of 34 pts with loss of PTEN protein by IHC (Arm P: 7 prostate, 6 breast, 3 squamous anal ca, 2 cholangiocarcinoma, 16 other), all are off treatment as of analysis (26 disease progression, 4 for AE, 4 other). Of 9 (37.5%) pts with SD, 3 had SD \u3e 6 months (prostate cancer; squamous bladder cancer, squamous anal cancer). Median progression-free survival was 1.8 months for both arms. Gr ≥ 3 treatment-related (tr) reversible toxicities were experienced by 30% (7) and 20% (7) of pts in arms N and P, respectively. No tr Gr 5 toxicities were observed in either arm. Conclusions: Single agent GSK2636771 has very modest activity in ca with PTEN gene mutation/deletion and/or PTEN protein loss

    Veliparib with carboplatin and paclitaxel in BRCA-mutated advanced breast cancer (BROCADE3):a randomised, double-blind, placebo-controlled, phase 3 trial

    Get PDF
    BACKGROUND: BRCA1 or BRCA2-mutated breast cancers are sensitive to poly(ADP-ribose) polymerase (PARP) inhibitors and platinum agents owing to deficiency in homologous recombination repair of DNA damage. In this trial, we compared veliparib versus placebo in combination with carboplatin and paclitaxel, and continued as monotherapy if carboplatin and paclitaxel were discontinued before progression, in patients with HER2-negative advanced breast cancer and a germline BRCA1 or BRCA2 mutation. METHODS: BROCADE3 was a randomised, double-blind, placebo-controlled, phase 3 trial done at 147 hospitals in 36 countries. Eligible patients (aged ≥18 years) had deleterious germline BRCA1 or BRCA2 mutation-associated, histologically or cytologically confirmed advanced HER2-negative breast cancer, an Eastern Cooperative Oncology Group performance status of 0-2, and had received up to two previous lines of chemotherapy for metastatic disease. Patients were randomly assigned (2:1) by interactive response technology by means of permuted blocks within strata (block size of 3 or 6) to carboplatin (area under the concentration curve 6 mg/mL per min intravenously) on day 1 and paclitaxel (80 mg/m2 intravenously) on days 1, 8, and 15 of 21-day cycles combined with either veliparib (120 mg orally twice daily, on days -2 to 5) or matching placebo. If patients discontinued carboplatin and paclitaxel before progression, they could continue veliparib or placebo at an intensified dose (300 mg twice daily continuously, escalating to 400 mg twice daily if tolerated) until disease progression. Patients in the control group could receive open-label veliparib monotherapy after disease progression. Randomisation was stratified by previous platinum use, history of CNS metastases, and oestrogen and progesterone receptor status. The primary endpoint was investigator-assessed progression-free survival per Response Evaluation Criteria in Solid Tumors version 1.1. Efficacy analyses were done by intention to treat, which included all randomly assigned patients with a centrally confirmed BRCA mutation, and safety analyses included all patients who received at least one dose of velilparib or placebo. This study is ongoing and is registered with ClinicalTrials.gov, NCT02163694. FINDINGS: Between July 30, 2014, and Jan 17, 2018, 2202 patients were screened, of whom 513 eligible patients were enrolled and randomly assigned. In the intention-to-treat population (n=509), 337 patients were assigned to receive veliparib plus carboplatin-paclitaxel (veliparib group) and 172 were assigned to receive placebo plus carboplatin-paclitaxel (control group). Median follow-up at data cutoff (April 5, 2019) was 35·7 months (IQR 24·9-43·6) in the veliparib group and 35·5 months (23·1-45·9) in the control group. Median progression-free survival was 14·5 months (95% CI 12·5-17·7) in the veliparib group versus 12·6 months (10·6-14·4) in the control group (hazard ratio 0·71 [95% CI 0·57-0·88], p=0·0016). The most common grade 3 or worse adverse events were neutropenia (272 [81%] of 336 patients in the veliparib group vs 143 [84%] of 171 patients in the control group), anaemia (142 [42%] vs 68 [40%]), and thrombocytopenia (134 [40%] vs 48 [28%]). Serious adverse events occurred in 115 (34%) patients in the veliparib group versus 49 (29%) patients in the control group. There were no study drug-related deaths. INTERPRETATION: The addition of veliparib to a highly active platinum doublet, with continuation as monotherapy if the doublet were discontinued, resulted in significant and durable improvement in progression-free survival in patients with germline BRCA mutation-associated advanced breast cancer. These data indicate the utility of combining platinum and PARP inhibitors in this patient population. FUNDING: AbbVie

    Targeted Mutation Detection in Advanced Breast Cancer Using MammaSeq Identifies RET as a Potential Contributor to Breast Cancer Metastasis

    Get PDF
    The lack of any reported breast cancer specific diagnostic NGS tests inspired the development of MammaSeq, an amplicon based NGS panel built specifically for use in advanced breast cancer. In a pilot study to define the clinical utility of the panel, 46 solid tumor samples, plus an additional 14 samples of circulating-free DNA (cfDNA) from patients with advanced breast cancer were sequenced and analyzed using the OncoKB precision oncology database. We identified 26 clinically actionable variants (levels 1-3) annotated by the OncoKB precision oncology database, distributed across 20 out of 46 solid tumor cases (40%), and 4 clinically actionable mutations distributed across 4 samples in the 14 cfDNA sample cohort (29%). The mutation allele (MAF) frequencies of ESR1-D538G and FOXA1-Y175C mutations correlated with CA.27.29 levels in patient-matched blood, indicating that MAF may be a reliable marker for disease burden. Interestingly, 4 of the mutations found in metastatic samples occurred in the gene RET, an oncogenic receptor tyrosine kinase. In an orthogonal study, the lab has recently identified RET as one of the most recurrently upregulated genes in breast cancer brain metastases. Interestingly, the ligand for RET is the family of glial-cell derived neurotrophic factors (GDNF), a growth factor secreted by glial cells of the central nervous system. This lead to the hypothesis that RET overexpression facilitates breast cancer brain metastasis in response to the high levels of GDNF, while RET activating point mutations increase metastatic capacity without specific organ tropism. While the effect of GDNF treatment on proliferation in 2D was limited, in ultra-low attachment (ULA) plates we saw a significant increase in anchorage independent growth of MCF-7 cells. To determine if GDNF acts as a chemoattractant for RET positive BrCa cells, we utilized a transwell migration assay, with GDNF as the sole chemoattractant. When RET was overexpressed, there was a visual increase in cell migration. Together, these studies demonstrate the clinical feasibility of using MammaSeq to detect clinically actionable mutations in breast cancer patients, and provides provisional data supporting the investigation of RET signaling as a potentially targetable mediator of breast cancer brain metastasis

    The ER-alpha mutation Y537S confers Tamoxifen-resistance via enhanced mitochondrial metabolism, glycolysis and Rho-GDI/PTEN signaling : implicating TIGAR in somatic resistance to endocrine therapy

    Get PDF
    Naturally-occurring somatic mutations in the estrogen receptor gene (ESR1) have been previously implicated in the clinical development of resistance to hormonal therapies, such as Tamoxifen. For example, the somatic mutation Y537S has been specifically associated with acquired endocrine resistance. Briefly, we recombinantly-transduced MCF7 cells with a lentiviral vector encoding ESR1 (Y537S). As a first step, we confirmed that MCF7-Y537S cells are indeed functionally resistant to Tamoxifen, as compared with vector alone controls. Importantly, further phenotypic characterization of Y537S cells revealed that they show increased resistance to Tamoxifen-induced apoptosis, allowing them to form mammospheres with higher efficiency, in the presence of Tamoxifen. Similarly, Y537S cells had elevated basal levels of ALDH activity, a marker of "stemness", which was also Tamoxifen-resistant. Metabolic flux analysis of Y537S cells revealed a hyper-metabolic phenotype, with significantly increased mitochondrial respiration and high ATP production, as well as enhanced aerobic glycolysis. Finally, to understand which molecular signaling pathways that may be hyper-activated in Y537S cells, we performed unbiased label-free proteomics analysis. Our results indicate that TIGAR over-expression and the Rho-GDI/PTEN signaling pathway appear to be selectively activated by the Y537S mutation. Remarkably, this profile is nearly identical in MCF7-TAMR cells; these cells were independently-generated , suggesting a highly conserved mechanism underlying Tamoxifen-resistance. Importantly, we show that the Y537S mutation is specifically associated with the over-expression of a number of protein markers of poor clinical outcome (COL6A3, ERBB2, STAT3, AFP, TFF1, CDK4 and CD44). In summary, we have uncovered a novel metabolic mechanism leading to endocrine resistance, which may have important clinical implications for improving patient outcomes

    Data quality aware analysis of differential expression in RNA-seq with NOISeq R/Bioc package

    Get PDF
    This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by-nc/4.0/), which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original work is properly cited.[EN] As the use of RNA-seq has popularized, there is an increasing consciousness of the importance of experimental design, bias removal, accurate quantification and control of false positives for proper data analysis. We introduce the NOISeq R-package for quality control and analysis of count data. We show how the available diagnostic tools can be used to monitor quality issues, make pre-processing decisions and improve analysis. We demonstrate that the non-parametric NOISeqBIO efficiently controls false discoveries in experiments with biological replication and outperforms state-of-the-art methods. NOISeq is a comprehensive resource that meets current needs for robust data-aware analysis of RNA-seq differential expression.European Union Seventh Framework Programme [FP7/2007-2013, 306000]; Spanish Ministry of Science and Innovation [MICINN, BIO2008-04638-E], in the framework of ERA-Net Pathogenomics; MICINN [DPI2008-06880-C03-03/DPI]. Funding for open access charge: University of Florida, Publication Funds.Tarazona Campos, S.; Furió Tarí, P.; Turrà, D.; Di Pietro, A.; Nueda, MJ.; Ferrer, A.; Conesa, A. (2015). Data quality aware analysis of differential expression in RNA-seq with NOISeq R/Bioc package. Nucleic Acids Research. 43(21):1-13. https://doi.org/10.1093/nar/gkv711S1134321Malone, J. H., & Oliver, B. (2011). Microarrays, deep sequencing and the true measure of the transcriptome. BMC Biology, 9(1). doi:10.1186/1741-7007-9-34Robles, J. A., Qureshi, S. E., Stephen, S. J., Wilson, S. R., Burden, C. J., & Taylor, J. M. (2012). Efficient experimental design and analysis strategies for the detection of differential expression using RNA-Sequencing. BMC Genomics, 13(1), 484. doi:10.1186/1471-2164-13-484Marioni, J. C., Mason, C. E., Mane, S. M., Stephens, M., & Gilad, Y. (2008). RNA-seq: An assessment of technical reproducibility and comparison with gene expression arrays. Genome Research, 18(9), 1509-1517. doi:10.1101/gr.079558.108Mortazavi, A., Williams, B. A., McCue, K., Schaeffer, L., & Wold, B. (2008). Mapping and quantifying mammalian transcriptomes by RNA-Seq. Nature Methods, 5(7), 621-628. doi:10.1038/nmeth.1226Wang, Z., Gerstein, M., & Snyder, M. (2009). RNA-Seq: a revolutionary tool for transcriptomics. Nature Reviews Genetics, 10(1), 57-63. doi:10.1038/nrg2484Wagner, J. R., Ge, B., Pokholok, D., Gunderson, K. L., Pastinen, T., & Blanchette, M. (2010). Computational Analysis of Whole-Genome Differential Allelic Expression Data in Human. PLoS Computational Biology, 6(7), e1000849. doi:10.1371/journal.pcbi.1000849Bell, G. D. M., Kane, N. C., Rieseberg, L. H., & Adams, K. L. (2013). RNA-Seq Analysis of Allele-Specific Expression, Hybrid Effects, and Regulatory Divergence in Hybrids Compared with Their Parents from Natural Populations. Genome Biology and Evolution, 5(7), 1309-1323. doi:10.1093/gbe/evt072Łabaj, P. P., Leparc, G. G., Linggi, B. E., Markillie, L. M., Wiley, H. S., & Kreil, D. P. (2011). Characterization and improvement of RNA-Seq precision in quantitative transcript expression profiling. Bioinformatics, 27(13), i383-i391. doi:10.1093/bioinformatics/btr247Auer, P. L., & Doerge, R. W. (2010). Statistical Design and Analysis of RNA Sequencing Data. Genetics, 185(2), 405-416. doi:10.1534/genetics.110.114983Busby, M. A., Stewart, C., Miller, C. A., Grzeda, K. R., & Marth, G. T. (2013). Scotty: a web tool for designing RNA-Seq experiments to measure differential gene expression. Bioinformatics, 29(5), 656-657. doi:10.1093/bioinformatics/btt015Cai, G., Li, H., Lu, Y., Huang, X., Lee, J., Müller, P., … Liang, S. (2012). Accuracy of RNA-Seq and its dependence on sequencing depth. BMC Bioinformatics, 13(Suppl 13), S5. doi:10.1186/1471-2105-13-s13-s5Oshlack, A., & Wakefield, M. J. (2009). Transcript length bias in RNA-seq data confounds systems biology. Biology Direct, 4(1), 14. doi:10.1186/1745-6150-4-14Risso, D., Schwartz, K., Sherlock, G., & Dudoit, S. (2011). GC-Content Normalization for RNA-Seq Data. BMC Bioinformatics, 12(1), 480. doi:10.1186/1471-2105-12-480Robinson, M. D., & Oshlack, A. (2010). A scaling normalization method for differential expression analysis of RNA-seq data. Genome Biology, 11(3), R25. doi:10.1186/gb-2010-11-3-r25Bullard, J. H., Purdom, E., Hansen, K. D., & Dudoit, S. (2010). Evaluation of statistical methods for normalization and differential expression in mRNA-Seq experiments. BMC Bioinformatics, 11(1). doi:10.1186/1471-2105-11-94Dillies, M.-A., Rau, A., Aubert, J., Hennequet-Antier, C., Jeanmougin, M., … Servant, N. (2012). A comprehensive evaluation of normalization methods for Illumina high-throughput RNA sequencing data analysis. Briefings in Bioinformatics, 14(6), 671-683. doi:10.1093/bib/bbs046Zheng, W., Chung, L. M., & Zhao, H. (2011). Bias detection and correction in RNA-Sequencing data. BMC Bioinformatics, 12(1), 290. doi:10.1186/1471-2105-12-290Robinson, M. D., McCarthy, D. J., & Smyth, G. K. (2009). edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics, 26(1), 139-140. doi:10.1093/bioinformatics/btp616Love, M. I., Huber, W., & Anders, S. (2014). Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biology, 15(12). doi:10.1186/s13059-014-0550-8Soneson, C., & Delorenzi, M. (2013). A comparison of methods for differential expression analysis of RNA-seq data. BMC Bioinformatics, 14(1). doi:10.1186/1471-2105-14-91Young, M. D., Wakefield, M. J., Smyth, G. K., & Oshlack, A. (2010). Gene ontology analysis for RNA-seq: accounting for selection bias. Genome Biology, 11(2), R14. doi:10.1186/gb-2010-11-2-r14Bashir, A., Bansal, V., & Bafna, V. (2010). Designing deep sequencing experiments: detecting structural variation and estimating transcript abundance. BMC Genomics, 11(1), 385. doi:10.1186/1471-2164-11-385Russo, F., & Angelini, C. (2014). RNASeqGUI: a GUI for analysing RNA-Seq data. Bioinformatics, 30(17), 2514-2516. doi:10.1093/bioinformatics/btu308Gentleman, R. C., Carey, V. J., Bates, D. M., Bolstad, B., Dettling, M., Dudoit, S., … Zhang, J. (2004). Genome Biology, 5(10), R80. doi:10.1186/gb-2004-5-10-r80Tarazona, S., Garcia-Alcalde, F., Dopazo, J., Ferrer, A., & Conesa, A. (2011). Differential expression in RNA-seq: A matter of depth. Genome Research, 21(12), 2213-2223. doi:10.1101/gr.124321.111Ferreira, P. G., Patalano, S., Chauhan, R., Ffrench-Constant, R., Gabaldón, T., Guigó, R., & Sumner, S. (2013). Transcriptome analyses of primitively eusocial wasps reveal novel insights into the evolution of sociality and the origin of alternative phenotypes. Genome Biology, 14(2), R20. doi:10.1186/gb-2013-14-2-r20Nascimento, R., Queiroz, F., Rocha, A., Ren, T. I., Mello, V., & Peixoto, A. (2012). Computer-assisted coloring and illuminating based on a region-tree structure. SpringerPlus, 1(1). doi:10.1186/2193-1801-1-1Zhu, Q.-H., Stephen, S., Kazan, K., Jin, G., Fan, L., Taylor, J., … Wang, M.-B. (2013). Characterization of the defense transcriptome responsive to Fusarium oxysporum-infection in Arabidopsis using RNA-seq. Gene, 512(2), 259-266. doi:10.1016/j.gene.2012.10.036Shearman, J. R., Jantasuriyarat, C., Sangsrakru, D., Yoocha, T., Vannavichit, A., Tragoonrung, S., & Tangphatsornruang, S. (2013). Transcriptome analysis of normal and mantled developing oil palm flower and fruit. Genomics, 101(5), 306-312. doi:10.1016/j.ygeno.2013.02.012Chen, G., Chen, J., Shi, C., Shi, L., Tong, W., & Shi, T. (2013). Dissecting the Characteristics and Dynamics of Human Protein Complexes at Transcriptome Cascade Using RNA-Seq Data. PLoS ONE, 8(6), e66521. doi:10.1371/journal.pone.0066521Durban, J., Pérez, A., Sanz, L., Gómez, A., Bonilla, F., Rodríguez, S., … Calvete, J. J. (2013). Integrated «omics» profiling indicates that miRNAs are modulators of the ontogenetic venom composition shift in the Central American rattlesnake, Crotalus simus simus. BMC Genomics, 14(1), 234. doi:10.1186/1471-2164-14-234Liu, W.-Y., Chang, Y.-M., Chen, S. C.-C., Lu, C.-H., Wu, Y.-H., Lu, M.-Y. J., … Li, W.-H. (2013). Anatomical and transcriptional dynamics of maize embryonic leaves during seed germination. Proceedings of the National Academy of Sciences, 110(10), 3979-3984. doi:10.1073/pnas.1301009110Su, L., Zhou, L., Liu, J., Cen, Z., Wu, C., Wang, T., … Liu, C. (2014). Phenotypic, genomic, transcriptomic and proteomic changes in Bacillus cereus after a short-term space flight. Advances in Space Research, 53(1), 18-29. doi:10.1016/j.asr.2013.08.001Xia, J. H., Liu, P., Liu, F., Lin, G., Sun, F., Tu, R., & Yue, G. H. (2013). Analysis of Stress-Responsive Transcriptome in the Intestine of Asian Seabass (Lates calcarifer) using RNA-Seq. DNA Research, 20(5), 449-460. doi:10.1093/dnares/dst022Nookaew, I., Papini, M., Pornputtapong, N., Scalcinati, G., Fagerberg, L., Uhlén, M., & Nielsen, J. (2012). A comprehensive comparison of RNA-Seq-based transcriptome analysis from reads to differential gene expression and cross-comparison with microarrays: a case study in Saccharomyces cerevisiae. Nucleic Acids Research, 40(20), 10084-10097. doi:10.1093/nar/gks804Bi, Y., & Davuluri, R. V. (2013). NPEBseq: nonparametric empirical bayesian-based procedure for differential expression analysis of RNA-seq data. BMC Bioinformatics, 14(1). doi:10.1186/1471-2105-14-262Klambauer, G., Unterthiner, T., & Hochreiter, S. (2013). DEXUS: identifying differential expression in RNA-Seq studies with unknown conditions. Nucleic Acids Research, 41(21), e198-e198. doi:10.1093/nar/gkt834Li, J., & Tibshirani, R. (2011). Finding consistent patterns: A nonparametric approach for identifying differential expression in RNA-Seq data. Statistical Methods in Medical Research, 22(5), 519-536. doi:10.1177/0962280211428386Lin, B., Zhang, L.-F., & Chen, X. (2014). LFCseq: a nonparametric approach for differential expression analysis of RNA-seq data. BMC Genomics, 15(Suppl 10), S7. doi:10.1186/1471-2164-15-s10-s7Anders, S., McCarthy, D. J., Chen, Y., Okoniewski, M., Smyth, G. K., Huber, W., & Robinson, M. D. (2013). Count-based differential expression analysis of RNA sequencing data using R and Bioconductor. Nature Protocols, 8(9), 1765-1786. doi:10.1038/nprot.2013.099Nueda, M. j., Ferrer, A., & Conesa, A. (2011). ARSyN: a method for the identification and removal of systematic noise in multifactorial time course microarray experiments. Biostatistics, 13(3), 553-566. doi:10.1093/biostatistics/kxr042Efron, B., Tibshirani, R., Storey, J. D., & Tusher, V. (2001). Empirical Bayes Analysis of a Microarray Experiment. Journal of the American Statistical Association, 96(456), 1151-1160. doi:10.1198/016214501753382129(2012). An integrated encyclopedia of DNA elements in the human genome. Nature, 489(7414), 57-74. doi:10.1038/nature11247Harrow, J., Frankish, A., Gonzalez, J. M., Tapanari, E., Diekhans, M., Kokocinski, F., … Hubbard, T. J. (2012). GENCODE: The reference human genome annotation for The ENCODE Project. Genome Research, 22(9), 1760-1774. doi:10.1101/gr.135350.111Kim, D., Pertea, G., Trapnell, C., Pimentel, H., Kelley, R., & Salzberg, S. L. (2013). TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions. Genome Biology, 14(4), R36. doi:10.1186/gb-2013-14-4-r36Anders, S., Pyl, P. T., & Huber, W. (2014). HTSeq--a Python framework to work with high-throughput sequencing data. Bioinformatics, 31(2), 166-169. doi:10.1093/bioinformatics/btu638López-Berges, M. S., Capilla, J., Turrà, D., Schafferer, L., Matthijs, S., Jöchl, C., … Di Pietro, A. (2012). HapX-Mediated Iron Homeostasis Is Essential for Rhizosphere Competence and Virulence of the Soilborne Pathogen Fusarium oxysporum. The Plant Cell, 24(9), 3805-3822. doi:10.1105/tpc.112.098624Flicek, P., Amode, M. R., Barrell, D., Beal, K., Brent, S., Carvalho-Silva, D., … Fitzgerald, S. (2011). Ensembl 2012. Nucleic Acids Research, 40(D1), D84-D90. doi:10.1093/nar/gkr991Ren, S., Peng, Z., Mao, J.-H., Yu, Y., Yin, C., Gao, X., … Sun, Y. (2012). RNA-seq analysis of prostate cancer in the Chinese population identifies recurrent gene fusions, cancer-associated long noncoding RNAs and aberrant alternative splicings. Cell Research, 22(5), 806-821. doi:10.1038/cr.2012.30Trapnell, C., Pachter, L., & Salzberg, S. L. (2009). TopHat: discovering splice junctions with RNA-Seq. Bioinformatics, 25(9), 1105-1111. doi:10.1093/bioinformatics/btp120Li, S., Łabaj, P. P., Zumbo, P., Sykacek, P., Shi, W., Shi, L., … Mason, C. E. (2014). Detecting and correcting systematic variation in large-scale RNA sequencing data. Nature Biotechnology, 32(9), 888-895. doi:10.1038/nbt.3000Wang, L., Wang, S., & Li, W. (2012). RSeQC: quality control of RNA-seq experiments. Bioinformatics, 28(16), 2184-2185. doi:10.1093/bioinformatics/bts356DeLuca, D. S., Levin, J. Z., Sivachenko, A., Fennell, T., Nazaire, M.-D., Williams, C., … Getz, G. (2012). RNA-SeQC: RNA-seq metrics for quality control and process optimization. Bioinformatics, 28(11), 1530-1532. doi:10.1093/bioinformatics/bts196García-Alcalde, F., Okonechnikov, K., Carbonell, J., Cruz, L. M., Götz, S., Tarazona, S., … Conesa, A. (2012). Qualimap: evaluating next-generation sequencing alignment data. Bioinformatics, 28(20), 2678-2679. doi:10.1093/bioinformatics/bts503Liu, Y., Zhou, J., & White, K. P. (2013). RNA-seq differential expression studies: more sequence or more replication? Bioinformatics, 30(3), 301-304. doi:10.1093/bioinformatics/btt688Rapaport, F., Khanin, R., Liang, Y., Pirun, M., Krek, A., Zumbo, P., … Betel, D. (2013). Comprehensive evaluation of differential gene expression analysis methods for RNA-seq data. Genome Biology, 14(9), R95. doi:10.1186/gb-2013-14-9-r95(2014). A comprehensive assessment of RNA-seq accuracy, reproducibility and information content by the Sequencing Quality Control Consortium. Nature Biotechnology, 32(9), 903-914. doi:10.1038/nbt.295

    The molecular landscape of premenopausal breast cancer

    Get PDF
    Introduction: Breast cancer in premenopausal women (preM) is frequently associated with worse prognosis compared to that in postmenopausal women (postM), and there is evidence that preM estrogen receptor-positive (ER+) tumors may respond poorly to endocrine therapy. There is, however, a paucity of studies characterizing molecular alterations in premenopausal tumors, a potential avenue for personalizing therapy for this group of women. Methods: Using TCGA and METABRIC databases, we analyzed gene expression, copy number, methylation, somatic mutation, and reverse-phase protein array data in breast cancers from >2,500 preM and postM women. Results: PreM tumors showed unique gene expression compared to postM tumors, however, this difference was limited to ER+ tumors. ER+ preM tumors showed unique DNA methylation, copy number and somatic mutations. Integrative pathway analysis revealed that preM tumors had elevated integrin/laminin and EGFR signaling, with enrichment for upstream TGFβ-regulation. Finally, preM tumors showed three different gene expression clusters with significantly different outcomes. Conclusion: Together these data suggest that ER+ preM tumors have distinct molecular characteristics compared to ER+ postM tumors, particularly with respect to integrin/laminin and EGFR signaling, which may represent therapeutic targets in this subgroup of breast cancers

    Local origin of two vegetative compatibility groups of Fusarium oxysporum f. sp. vasinfectum in Australia

    Get PDF
    Pathogenicity and genetic diversity of Fusarium oxysporum from geographically widespread native Gossypium populations, including a cotton growing area believed to be the center of origin of VCG 01111 and VCG 01112 of F. oxysporum f. sp. vasinfectum (Fov) in Australia, was determined using glasshouse bioassays and AFLPs. Five lineages (A–E) were identified among 856 isolates. Of these, 12% were strongly pathogenic on cotton, 10% were weakly pathogenic and designated wild Fov, while 78% were nonpathogenic. In contrast to the occurrence of pathogenic isolates in all five lineages in soils associated with wild Gossypium, in cotton growing areas only three lineages (A, B, E) occurred and all pathogenic isolates belonged to two subgroups in lineage A. One of these contained VCG 01111 isolates while the other contained VCG 01112 isolates. Sequence analyses of translation elongation factor-1α, mitochondrial small subunit rDNA, nitrate reductase and phosphate permease confirmed that Australian Fov isolates were more closely related to lineage A isolates of native F. oxysporum than to Fov races 1–8 found overseas. These results strongly support a local evolutionary origin for Fov in Australian cotton growing regions

    TBCRC 002: A phase II, randomized, open-label trial of preoperative letrozole with or without bevacizumab in postmenopausal women with newly diagnosed stage 2/3 hormone receptor-positive and HER2-negative breast cancer

    Get PDF
    Background: In preclinical studies, the expression of vascular endothelial growth factor (VEGF) in hormone receptor-positive breast cancer is associated with estrogen-independent tumor growth and resistance to endocrine therapies. This study investigated whether the addition of bevacizumab, a monoclonal antibody against VEGF, to letrozole enhanced the antitumor activity of the letrozole in the preoperative setting. Methods: Postmenopausal women with newly diagnosed stage 2 or 3 estrogen and/or progesterone receptor-positive, HER2-negative breast cancer were randomly assigned (2:1) between letrozole 2.5 mg PO daily plus bevacizumab 15 mg/kg IV every 3 weeks (Let/Bev) and letrozole 2.5 mg PO daily (Let) for 24 weeks prior to definitive surgery. Primary objective was within-arm pathologic complete remission (pCR) rate. Secondary objectives were safety, objective response, and downstaging rate. Results: Seventy-five patients were randomized (Let/Bev n = 50, Let n = 25). Of the 45 patients evaluable for pathological response in the Let/Bev arm, 5 (11%; 95% CI, 3.7-24.1%) achieved pCR and 4 (9%; 95% CI, 2.5-21.2%) had microscopic residual disease; no pCRs or microscopic residual disease was seen in the Let arm (0%; 95% CI, 0-14.2%). The rates of downstaging were 44.4% (95% CI, 29.6-60.0%) and 37.5% (95% CI, 18.8-59.4%) in the Let/Bev and Let arms, respectively. Adverse events typically associated with letrozole (hot flashes, arthralgias, fatigue, myalgias) occurred in similar frequencies in the two arms. Hypertension, headache, and proteinuria were seen exclusively in the Let/Bev arm. The rates of grade 3 and 4 adverse events and discontinuation due to adverse events were 18% vs 8% and 16% vs none in the Let/Bev and Let arms, respectively. A small RNA-based classifier predictive of response to preoperative Let/Bev was developed and confirmed on an independent cohort. Conclusion: In the preoperative setting, the addition of bevacizumab to letrozole was associated with a pCR rate of 11%; no pCR was seen with letrozole alone. There was additive toxicity with the incorporation of bevacizumab. Responses to Let/Bev can be predicted from the levels of 5 small RNAs in a pretreatment biopsy. Trial registration: This trial is registered with ClinicalTrials.gov (Identifier: NCT00161291), first posted on September 12, 2005, and is completed
    corecore