Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Sherri R. Davies is active.

Publication


Featured researches published by Sherri R. Davies.


Journal of the National Cancer Institute | 2009

Ki67 Index, HER2 Status, and Prognosis of Patients With Luminal B Breast Cancer

Maggie Cheang; Stephen Chia; David Voduc; Dongxia Gao; Samuel Leung; Jacqueline Snider; Mark A. Watson; Sherri R. Davies; Philip S. Bernard; Joel S. Parker; Charles M. Perou; Matthew J. Ellis; Torsten O. Nielsen

Background Gene expression profiling of breast cancer has identified two biologically distinct estrogen receptor (ER)-positive subtypes of breast cancer: luminal A and luminal B. Luminal B tumors have higher proliferation and poorer prognosis than luminal A tumors. In this study, we developed a clinically practical immunohistochemistry assay to distinguish luminal B from luminal A tumors and investigated its ability to separate tumors according to breast cancer recurrence-free and disease-specific survival. Methods Tumors from a cohort of 357 patients with invasive breast carcinomas were subtyped by gene expression profile. Hormone receptor status, HER2 status, and the Ki67 index (percentage of Ki67-positive cancer nuclei) were determined immunohistochemically. Receiver operating characteristic curves were used to determine the Ki67 cut point to distinguish luminal B from luminal A tumors. The prognostic value of the immunohistochemical assignment for breast cancer recurrence-free and disease-specific survival was investigated with an independent tissue microarray series of 4046 breast cancers by use of Kaplan–Meier curves and multivariable Cox regression. Results Gene expression profiling classified 101 (28%) of the 357 tumors as luminal A and 69 (19%) as luminal B. The best Ki67 index cut point to distinguish luminal B from luminal A tumors was 13.25%. In an independent cohort of 4046 patients with breast cancer, 2847 had hormone receptor–positive tumors. When HER2 immunohistochemistry and the Ki67 index were used to subtype these 2847 tumors, we classified 1530 (59%, 95% confidence interval [CI] = 57% to 61%) as luminal A, 846 (33%, 95% CI = 31% to 34%) as luminal B, and 222 (9%, 95% CI = 7% to 10%) as luminal–HER2 positive. Luminal B and luminal–HER2-positive breast cancers were statistically significantly associated with poor breast cancer recurrence-free and disease-specific survival in all adjuvant systemic treatment categories. Of particular relevance are women who received tamoxifen as their sole adjuvant systemic therapy, among whom the 10-year breast cancer–specific survival was 79% (95% CI = 76% to 83%) for luminal A, 64% (95% CI = 59% to 70%) for luminal B, and 57% (95% CI = 47% to 69%) for luminal–HER2 subtypes. Conclusion Expression of ER, progesterone receptor, and HER2 proteins and the Ki67 index appear to distinguish luminal A from luminal B breast cancer subtypes.


Nature | 2010

Genome remodelling in a basal-like breast cancer metastasis and xenograft.

Li Ding; Matthew J. Ellis; Shunqiang Li; David E. Larson; Ken Chen; John W. Wallis; Christopher C. Harris; Michael D. McLellan; Robert S. Fulton; Lucinda Fulton; Rachel Abbott; Jeremy Hoog; David J. Dooling; Daniel C. Koboldt; Heather K. Schmidt; Joelle Kalicki; Qunyuan Zhang; Lei Chen; Ling Lin; Michael C. Wendl; Joshua F. McMichael; Vincent Magrini; Lisa Cook; Sean McGrath; Tammi L. Vickery; Elizabeth L. Appelbaum; Katherine DeSchryver; Sherri R. Davies; Therese Guintoli; Li Lin

Massively parallel DNA sequencing technologies provide an unprecedented ability to screen entire genomes for genetic changes associated with tumour progression. Here we describe the genomic analyses of four DNA samples from an African-American patient with basal-like breast cancer: peripheral blood, the primary tumour, a brain metastasis and a xenograft derived from the primary tumour. The metastasis contained two de novo mutations and a large deletion not present in the primary tumour, and was significantly enriched for 20 shared mutations. The xenograft retained all primary tumour mutations and displayed a mutation enrichment pattern that resembled the metastasis. Two overlapping large deletions, encompassing CTNNA1, were present in all three tumour samples. The differential mutation frequencies and structural variation patterns in metastasis and xenograft compared with the primary tumour indicate that secondary tumours may arise from a minority of cells within the primary tumour.


Nature | 2014

Proteogenomic characterization of human colon and rectal cancer

Bing Zhang; Jing Wang; Xiaojing Wang; Jing Zhu; Qi Liu; Zhiao Shi; Matthew C. Chambers; Lisa J. Zimmerman; Kent Shaddox; Sangtae Kim; Sherri R. Davies; Sean Wang; Pei Wang; Christopher R. Kinsinger; Robert Rivers; Henry Rodriguez; R. Reid Townsend; Matthew J. Ellis; Steven A. Carr; David L. Tabb; Robert J. Coffey; Robbert J. C. Slebos; Daniel C. Liebler; Michael A. Gillette; Karl R. Klauser; Eric Kuhn; D. R. Mani; Philipp Mertins; Karen A. Ketchum; Amanda G. Paulovich

Extensive genomic characterization of human cancers presents the problem of inference from genomic abnormalities to cancer phenotypes. To address this problem, we analysed proteomes of colon and rectal tumours characterized previously by The Cancer Genome Atlas (TCGA) and perform integrated proteogenomic analyses. Somatic variants displayed reduced protein abundance compared to germline variants. Messenger RNA transcript abundance did not reliably predict protein abundance differences between tumours. Proteomics identified five proteomic subtypes in the TCGA cohort, two of which overlapped with the TCGA ‘microsatellite instability/CpG island methylation phenotype’ transcriptomic subtype, but had distinct mutation, methylation and protein expression patterns associated with different clinical outcomes. Although copy number alterations showed strong cis- and trans-effects on mRNA abundance, relatively few of these extend to the protein level. Thus, proteomics data enabled prioritization of candidate driver genes. The chromosome 20q amplicon was associated with the largest global changes at both mRNA and protein levels; proteomics data highlighted potential 20q candidates, including HNF4A (hepatocyte nuclear factor 4, alpha), TOMM34 (translocase of outer mitochondrial membrane 34) and SRC (SRC proto-oncogene, non-receptor tyrosine kinase). Integrated proteogenomic analysis provides functional context to interpret genomic abnormalities and affords a new paradigm for understanding cancer biology.


Clinical Cancer Research | 2010

A Comparison of PAM50 Intrinsic Subtyping with Immunohistochemistry and Clinical Prognostic Factors in Tamoxifen-Treated Estrogen Receptor-Positive Breast Cancer

Torsten O. Nielsen; Joel S. Parker; Samuel Leung; K. David Voduc; Mark T.W. Ebbert; Tammi L. Vickery; Sherri R. Davies; Jacqueline Snider; Inge J. Stijleman; Jerry S. Reed; Maggie Cheang; Elaine R. Mardis; Charles M. Perou; Philip S. Bernard; Matthew J. Ellis

Purpose: To compare clinical, immunohistochemical (IHC), and gene expression models of prognosis applicable to formalin-fixed, paraffin-embedded blocks in a large series of estrogen receptor (ER)–positive breast cancers from patients uniformly treated with adjuvant tamoxifen. Experimental Design: Quantitative real-time reverse transcription-PCR (qRT-PCR) assays for 50 genes identifying intrinsic breast cancer subtypes were completed on 786 specimens linked to clinical (median follow-up, 11.7 years) and IHC [ER, progesterone receptor (PR), HER2, and Ki67] data. Performance of predefined intrinsic subtype and risk-of-relapse scores was assessed using multivariable Cox models and Kaplan-Meier analysis. Harrells C-index was used to compare fixed models trained in independent data sets, including proliferation signatures. Results: Despite clinical ER positivity, 10% of cases were assigned to nonluminal subtypes. qRT-PCR signatures for proliferation genes gave more prognostic information than clinical assays for hormone receptors or Ki67. In Cox models incorporating standard prognostic variables, hazard ratios for breast cancer disease-specific survival over the first 5 years of follow-up, relative to the most common luminal A subtype, are 1.99 [95% confidence interval (CI), 1.09-3.64] for luminal B, 3.65 (95% CI, 1.64-8.16) for HER2-enriched subtype, and 17.71 (95% CI, 1.71-183.33) for the basal-like subtype. For node-negative disease, PAM50 qRT-PCR–based risk assignment weighted for tumor size and proliferation identifies a group with >95% 10-year survival without chemotherapy. In node-positive disease, PAM50-based prognostic models were also superior. Conclusion: The PAM50 gene expression test for intrinsic biological subtype can be applied to large series of formalin-fixed, paraffin-embedded breast cancers, and gives more prognostic information than clinical factors and IHC using standard cut points. Clin Cancer Res; 16(21); 5222–32. ©2010 AACR.


Cell Reports | 2013

Endocrine-Therapy-Resistant ESR1 Variants Revealed by Genomic Characterization of Breast-Cancer-Derived Xenografts

Shunqiang Li; Dong Shen; Jieya Shao; Robert Crowder; Wenbin Liu; Aleix Prat; Xiaping He; Shuying Liu; Jeremy Hoog; Charles Lu; Li Ding; Obi L. Griffith; Christopher A. Miller; Dave Larson; Robert S. Fulton; Michelle L. K. Harrison; Tom Mooney; Joshua F. McMichael; Jingqin Luo; Yu Tao; Rodrigo Franco Gonçalves; Christopher Schlosberg; Jeffrey F. Hiken; Laila Saied; César Sánchez; Therese Giuntoli; Caroline Bumb; Crystal Cooper; Robert T. Kitchens; Austin Lin

To characterize patient-derived xenografts (PDXs) for functional studies, we made whole-genome comparisons with originating breast cancers representative of the major intrinsic subtypes. Structural and copy number aberrations were found to be retained with high fidelity. However, at the single-nucleotide level, variable numbers of PDX-specific somatic events were documented, although they were only rarely functionally significant. Variant allele frequencies were often preserved in the PDXs, demonstrating that clonal representation can be transplantable. Estrogen-receptor-positive PDXs were associated with ESR1 ligand-binding-domain mutations, gene amplification, or an ESR1/YAP1 translocation. These events produced different endocrine-therapy-response phenotypes in human, cell line, and PDX endocrine-response studies. Hence, deeply sequenced PDX models are an important resource for the search for genome-forward treatment options and capture endocrine-drug-resistance etiologies that are not observed in standard cell lines. The originating tumor genome provides a benchmark for assessing genetic drift and clonal representation after transplantation.


Nature | 2016

Proteogenomics connects somatic mutations to signalling in breast cancer

Philipp Mertins; D. R. Mani; Kelly V. Ruggles; Michael A. Gillette; Karl R. Clauser; Pei Wang; Xianlong Wang; Jana W. Qiao; Song Cao; Francesca Petralia; Emily Kawaler; Filip Mundt; Karsten Krug; Zhidong Tu; Jonathan T. Lei; Michael L. Gatza; Matthew D. Wilkerson; Charles M. Perou; Venkata Yellapantula; Kuan Lin Huang; Chenwei Lin; Michael D. McLellan; Ping Yan; Sherri R. Davies; R. Reid Townsend; Steven J. Skates; Jing Wang; Bing Zhang; Christopher R. Kinsinger; Mehdi Mesri

Summary Somatic mutations have been extensively characterized in breast cancer, but the effects of these genetic alterations on the proteomic landscape remain poorly understood. We describe quantitative mass spectrometry-based proteomic and phosphoproteomic analyses of 105 genomically annotated breast cancers of which 77 provided high-quality data. Integrated analyses allowed insights into the somatic cancer genome including the consequences of chromosomal loss, such as the 5q deletion characteristic of basal-like breast cancer. The 5q trans effects were interrogated against the Library of Integrated Network-based Cellular Signatures, thereby connecting CETN3 and SKP1 loss to elevated expression of EGFR, and SKP1 loss also to increased SRC. Global proteomic data confirmed a stromal-enriched group in addition to basal and luminal clusters and pathway analysis of the phosphoproteome identified a G Protein-coupled receptor cluster that was not readily identified at the mRNA level. Besides ERBB2, other amplicon-associated, highly phosphorylated kinases were identified, including CDK12, PAK1, PTK2, RIPK2 and TLK2. We demonstrate that proteogenomic analysis of breast cancer elucidates functional consequences of somatic mutations, narrows candidate nominations for driver genes within large deletions and amplified regions, and identifies therapeutic targets.


Cancer Research | 2009

PIK3CA and PIK3CB Inhibition Produce Synthetic Lethality when Combined with Estrogen Deprivation in Estrogen Receptor-Positive Breast Cancer

Robert Crowder; Chanpheng Phommaly; Yu Tao; Jeremy Hoog; Jingqin Luo; Charles M. Perou; Joel S. Parker; Melinda A. Miller; David Huntsman; Li Lin; Jacqueline Snider; Sherri R. Davies; John A. Olson; Mark A. Watson; Anthony J. Saporita; Jason D. Weber; Matthew J. Ellis

Several phosphoinositide 3-kinase (PI3K) catalytic subunit inhibitors are currently in clinical trial. We therefore sought to examine relationships between pharmacologic inhibition and somatic mutations in PI3K catalytic subunits in estrogen receptor (ER)-positive breast cancer, in which these mutations are particularly common. RNA interference (RNAi) was used to determine the effect of selective inhibition of PI3K catalytic subunits, p110alpha and p110beta, in ER(+) breast cancer cells harboring either mutation (PIK3CA) or gene amplification (PIK3CB). p110alpha RNAi inhibited growth and promoted apoptosis in all tested ER(+) breast cancer cells under estrogen deprived-conditions, whereas p110beta RNAi only affected cells harboring PIK3CB amplification. Moreover, dual p110alpha/p110beta inhibition potentiated these effects. In addition, treatment with the clinical-grade PI3K catalytic subunit inhibitor BEZ235 also promoted apoptosis in ER(+) breast cancer cells. Importantly, estradiol suppressed apoptosis induced by both gene knockdowns and BEZ235 treatment. Our results suggest that PI3K inhibitors should target both p110alpha and p110beta catalytic subunits, whether wild-type or mutant, and be combined with endocrine therapy for maximal efficacy when treating ER(+) breast cancer.


Cell | 2016

Integrated proteogenomic characterization of human high-grade serous ovarian cancer

Hui Zhang; Tao Liu; Zhen Zhang; Samuel H. Payne; Bai Zhang; Jason E. McDermott; Jian-Ying Zhou; Vladislav A. Petyuk; Li Chen; Debjit Ray; Shisheng Sun; Feng Yang; Lijun Chen; Jing Wang; Punit Shah; Seong Won Cha; Paul Aiyetan; Sunghee Woo; Yuan Tian; Marina A. Gritsenko; Therese R. Clauss; Caitlin H. Choi; Matthew E. Monroe; Stefani N. Thomas; Song Nie; Chaochao Wu; Ronald J. Moore; Kun-Hsing Yu; David L. Tabb; David Fenyö

To provide a detailed analysis of the molecular components and underlying mechanisms associated with ovarian cancer, we performed a comprehensive mass-spectrometry-based proteomic characterization of 174 ovarian tumors previously analyzed by The Cancer Genome Atlas (TCGA), of which 169 were high-grade serous carcinomas (HGSCs). Integrating our proteomic measurements with the genomic data yielded a number of insights into disease, such as how different copy-number alternations influence the proteome, the proteins associated with chromosomal instability, the sets of signaling pathways that diverse genome rearrangements converge on, and the ones most associated with short overall survival. Specific protein acetylations associated with homologous recombination deficiency suggest a potential means for stratifying patients for therapy. In addition to providing a valuable resource, these findings provide a view of how the somatic genome drives the cancer proteome and associations between protein and post-translational modification levels and clinical outcomes in HGSC. VIDEO ABSTRACT.


Molecular & Cellular Proteomics | 2014

Ischemia in tumors induces early and sustained phosphorylation changes in stress kinase pathways but does not affect global protein levels

Philipp Mertins; Feng Yang; Tao Liu; D. R. Mani; Vladislav A. Petyuk; Michael A. Gillette; Karl R. Clauser; Jana W. Qiao; Marina A. Gritsenko; Ronald J. Moore; Douglas A. Levine; R. Reid Townsend; Petra Erdmann-Gilmore; Jacqueline Snider; Sherri R. Davies; Kelly V. Ruggles; David Fenyö; R. Thomas Kitchens; Shunqiang Li; Narcisco Olvera; Fanny Dao; Henry Rodriguez; Daniel W. Chan; Daniel C. Liebler; Forest M. White; Karin D. Rodland; Gordon B. Mills; Richard D. Smith; Amanda G. Paulovich; Matthew J. Ellis

Protein abundance and phosphorylation convey important information about pathway activity and molecular pathophysiology in diseases including cancer, providing biological insight, informing drug and diagnostic development, and guiding therapeutic intervention. Analyzed tissues are usually collected without tight regulation or documentation of ischemic time. To evaluate the impact of ischemia, we collected human ovarian tumor and breast cancer xenograft tissue without vascular interruption and performed quantitative proteomics and phosphoproteomics after defined ischemic intervals. Although the global expressed proteome and most of the >25,000 quantified phosphosites were unchanged after 60 min, rapid phosphorylation changes were observed in up to 24% of the phosphoproteome, representing activation of critical cancer pathways related to stress response, transcriptional regulation, and cell death. Both pan-tumor and tissue-specific changes were observed. The demonstrated impact of pre-analytical tissue ischemia on tumor biology mandates caution in interpreting stress-pathway activation in such samples and motivates reexamination of collection protocols for phosphoprotein analysis.


Osteoarthritis and Cartilage | 2008

Exuberant expression of chemokine genes by adult human articular chondrocytes in response to IL-1β

Linda J. Sandell; Xiaoyun Xing; Carl J. Franz; Sherri R. Davies; Li-Wei Chang; Debabrata Patra

OBJECTIVE To provide a more complete picture of the effect of interleukin-1 beta (IL-1beta) on adult human articular chondrocyte gene expression, in contrast to the candidate gene approach. DESIGN Chondrocytes from human knee cartilage were cultured in medium containing IL-1beta. Changes in gene expression were analyzed by microarray and reverse transcriptase-polymerase chain reaction analysis. The ability of transforming growth factor beta-1 (TGF-beta1), fibroblast growth factor (FGF)-18, and bone morphogenetic protein 2 (BMP-2) to alter the effects of IL-1beta was analyzed. Computational analysis of the promoter regions of differentially expressed genes for transcription factor binding motifs was performed. RESULTS IL-1beta-treated human chondrocytes showed significant increases in the expression of granulocyte colony stimulating factor-3, endothelial leukocyte adhesion molecule 1 and leukemia inhibitory factor as well as for a large group of chemokines that include CXCL1, CXCL2, CXCL3, CXCL5, CXCL6, CXCL8, CCL2, CCL3, CCL4, CCL5, CCL8, CCL20, CCL3L1, CX3CL1 and the cytokine IL-6. As expected, the mRNA for matrix metalloproteinase (MMP)-13 and BMP-2 also increased while mRNA for the matrix genes COL2A1 and aggrecan was down-regulated. A subset of chemokines increased rapidly at very low levels of IL-1beta. The phenotype induced by IL-1beta was partially reversed by TGF-beta1, but not by BMP-2. In the presence of IL-1beta, FGF-18 increased expression of ADAMTS-4, aggrecan, BMP-2, COL2A1, CCL3, CCL4, CCL20, CXCL1, CXCL3, CXCL6, IL-1beta, IL-6, and IL-8 and decreased ADAMTS-5, MMP-13, CCL2, and CCL8. Computational analysis revealed a high likelihood that the most up-regulated chemokines are regulated by the transcription factors myocyte enhancer binding factor-3 (MEF-3), CCAAT/enhancer binding protein (C/EBP) and nuclear factor-kappa B (NF-kappaB). CONCLUSION IL-1beta has a diverse effect on gene expression profile in human chondrocytes affecting matrix genes as well as chemokines and cytokines. TGF-beta1 has the ability to antagonize some of the phenotype induced by IL-1beta.

Collaboration


Dive into the Sherri R. Davies's collaboration.

Top Co-Authors

Avatar

Matthew J. Ellis

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Jacqueline Snider

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Charles M. Perou

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Jeremy Hoog

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Shunqiang Li

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

R. Reid Townsend

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Elaine R. Mardis

Nationwide Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Li Ding

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge