Network


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

Hotspot


Dive into the research topics where David Fenstermacher is active.

Publication


Featured researches published by David Fenstermacher.


Nature Genetics | 2009

A genome-wide association study identifies a new ovarian cancer susceptibility locus on 9p22.2

Honglin Song; Susan J. Ramus; Jonathan Tyrer; Kelly L. Bolton; Aleksandra Gentry-Maharaj; Eva Wozniak; Hoda Anton-Culver; Jenny Chang-Claude; Daniel W. Cramer; Richard A. DiCioccio; Thilo Dörk; Ellen L. Goode; Marc T. Goodman; Joellen M. Schildkraut; Thomas A. Sellers; Laura Baglietto; Matthias W. Beckmann; Jonathan Beesley; Jan Blaakær; Michael E. Carney; Stephen J. Chanock; Zhihua Chen; Julie M. Cunningham; Ed Dicks; Jennifer A. Doherty; Matthias Dürst; Arif B. Ekici; David Fenstermacher; Brooke L. Fridley; Graham G. Giles

Epithelial ovarian cancer has a major heritable component, but the known susceptibility genes explain less than half the excess familial risk. We performed a genome-wide association study (GWAS) to identify common ovarian cancer susceptibility alleles. We evaluated 507,094 SNPs genotyped in 1,817 cases and 2,353 controls from the UK and ∼2 million imputed SNPs. We genotyped the 22,790 top ranked SNPs in 4,274 cases and 4,809 controls of European ancestry from Europe, USA and Australia. We identified 12 SNPs at 9p22 associated with disease risk (P < 10−8). The most significant SNP (rs3814113; P = 2.5 × 10−17) was genotyped in a further 2,670 ovarian cancer cases and 4,668 controls, confirming its association (combined data odds ratio (OR) = 0.82, 95% confidence interval (CI) 0.79–0.86, Ptrend = 5.1 × 10−19). The association differs by histological subtype, being strongest for serous ovarian cancers (OR 0.77, 95% CI 0.73–0.81, Ptrend = 4.1 × 10−21).


Scientific Reports | 2012

12-Chemokine Gene Signature Identifies Lymph Node-like Structures in Melanoma: Potential for Patient Selection for Immunotherapy?

Jane L. Messina; David Fenstermacher; Steven Eschrich; Xiaotao Qu; Anders Berglund; Mark C. Lloyd; Michael J. Schell; Vernon K. Sondak; Jeffrey S. Weber; James J. Mulé

We have interrogated a 12-chemokine gene expression signature (GES) on genomic arrays of 14,492 distinct solid tumors and show broad distribution across different histologies. We hypothesized that this 12-chemokine GES might accurately predict a unique intratumoral immune reaction in stage IV (non-locoregional) melanoma metastases. The 12-chemokine GES predicted the presence of unique, lymph node-like structures, containing CD20+ B cell follicles with prominent areas of CD3+ T cells (both CD4+ and CD8+ subsets). CD86+, but not FoxP3+, cells were present within these unique structures as well. The direct correlation between the 12-chemokine GES score and the presence of unique, lymph nodal structures was also associated with better overall survival of the subset of melanoma patients. The use of this novel 12-chemokine GES may reveal basic information on in situ mechanisms of the anti-tumor immune response, potentially leading to improvements in the identification and selection of melanoma patients most suitable for immunotherapy.


Molecular & Cellular Proteomics | 2008

Proteomic Contributions to Personalized Cancer Care

John M. Koomen; Eric B. Haura; Gerold Bepler; Rebecca Sutphen; Elizabeth Remily-Wood; Kaaron Benson; Mohamad A. Hussein; Lori A. Hazlehurst; Timothy J. Yeatman; Lynne T. Hildreth; Thomas A. Sellers; Paul B. Jacobsen; David Fenstermacher; William S. Dalton

Cancer impacts each patient and family differently. Our current understanding of the disease is primarily limited to clinical hallmarks of cancer, but many specific molecular mechanisms remain elusive. Genetic markers can be used to determine predisposition to tumor development, but molecularly targeted treatment strategies that improve patient prognosis are not widely available for most cancers. Individualized care plans, also described as personalized medicine, still must be developed by understanding and implementing basic science research into clinical treatment. Proteomics holds great promise in contributing to the prevention and cure of cancer because it provides unique tools for discovery of biomarkers and therapeutic targets. As such, proteomics can help translate basic science discoveries into the clinical practice of personalized medicine. Here we describe how biological mass spectrometry and proteome analysis interact with other major patient care and research initiatives and present vignettes illustrating efforts in discovery of diagnostic biomarkers for ovarian cancer, development of treatment strategies in lung cancer, and monitoring prognosis and relapse in multiple myeloma patients.


Cancer Research | 2011

LIN28B Polymorphisms Influence Susceptibility to Epithelial Ovarian Cancer

Jennifer Permuth-Wey; Donghwa Kim; Ya Yu Tsai; Hui-Yi Lin; Y. Ann Chen; Jill S. Barnholtz-Sloan; Michael J. Birrer; Gregory C. Bloom; Stephen J. Chanock; Zhihua Chen; Daniel W. Cramer; Julie M. Cunningham; Getachew A. Dagne; Judith Ebbert-Syfrett; David Fenstermacher; Brooke L. Fridley; Montserrat Garcia-Closas; Simon A. Gayther; William Ge; Aleksandra Gentry-Maharaj; Jesus Gonzalez-Bosquet; Ellen L. Goode; Edwin S. Iversen; Heather Jim; William Kong; John R. McLaughlin; Usha Menon; Alvaro N.A. Monteiro; Steven A. Narod; Paul Pharoah

Defective microRNA (miRNA) biogenesis contributes to the development and progression of epithelial ovarian cancer (EOC). In this study, we examined the hypothesis that single nucleotide polymorphisms (SNP) in miRNA biogenesis genes may influence EOC risk. In an initial investigation, 318 SNPs in 18 genes were evaluated among 1,815 EOC cases and 1,900 controls, followed up by a replicative joint meta-analysis of data from an additional 2,172 cases and 3,052 controls. Of 23 SNPs from 9 genes associated with risk (empirical P < 0.05) in the initial investigation, the meta-analysis replicated 6 SNPs from the DROSHA, FMR1, LIN28, and LIN28B genes, including rs12194974 (G>A), an SNP in a putative transcription factor binding site in the LIN28B promoter region (summary OR = 0.90, 95% CI: 0.82-0.98; P = 0.015) which has been recently implicated in age of menarche and other phenotypes. Consistent with reports that LIN28B overexpression in EOC contributes to tumorigenesis by repressing tumor suppressor let-7 expression, we provide data from luciferase reporter assays and quantitative RT-PCR to suggest that the inverse association among rs12194974 A allele carriers may be because of reduced LIN28B expression. Our findings suggest that variants in LIN28B and possibly other miRNA biogenesis genes may influence EOC susceptibility.


PLOS ONE | 2014

Clonal Architectures and Driver Mutations in Metastatic Melanomas

Li Ding; Minjung Kim; Krishna L. Kanchi; Nathan D. Dees; Charles Lu; Malachi Griffith; David Fenstermacher; Hyeran Sung; Christopher A. Miller; Brian D. Goetz; Michael C. Wendl; Obi L. Griffith; Lynn A. Cornelius; Gerald P. Linette; Joshua F. McMichael; Vernon K. Sondak; Ryan C. Fields; Timothy J. Ley; James J. Mulé; Richard Wilson; Jeffrey S. Weber

To reveal the clonal architecture of melanoma and associated driver mutations, whole genome sequencing (WGS) and targeted extension sequencing were used to characterize 124 melanoma cases. Significantly mutated gene analysis using 13 WGS cases and 15 additional paired extension cases identified known melanoma genes such as BRAF, NRAS, and CDKN2A, as well as a novel gene EPHA3, previously implicated in other cancer types. Extension studies using tumors from another 96 patients discovered a large number of truncation mutations in tumor suppressors (TP53 and RB1), protein phosphatases (e.g., PTEN, PTPRB, PTPRD, and PTPRT), as well as chromatin remodeling genes (e.g., ASXL3, MLL2, and ARID2). Deep sequencing of mutations revealed subclones in the majority of metastatic tumors from 13 WGS cases. Validated mutations from 12 out of 13 WGS patients exhibited a predominant UV signature characterized by a high frequency of C->T transitions occurring at the 3′ base of dipyrimidine sequences while one patient (MEL9) with a hypermutator phenotype lacked this signature. Strikingly, a subclonal mutation signature analysis revealed that the founding clone in MEL9 exhibited UV signature but the secondary clone did not, suggesting different mutational mechanisms for two clonal populations from the same tumor. Further analysis of four metastases from different geographic locations in 2 melanoma cases revealed phylogenetic relationships and highlighted the genetic alterations responsible for differential drug resistance among metastatic tumors. Our study suggests that clonal evaluation is crucial for understanding tumor etiology and drug resistance in melanoma.


BMC Bioinformatics | 2013

Iterative rank-order normalization of gene expression microarray data

Eric A. Welsh; Steven Eschrich; Anders Berglund; David Fenstermacher

BackgroundMany gene expression normalization algorithms exist for Affymetrix GeneChip microarrays. The most popular of these is RMA, primarily due to the precision and low noise produced during the process. A significant strength of this and similar approaches is the use of the entire set of arrays during both normalization and model-based estimation of signal. However, this leads to differing estimates of expression based on the starting set of arrays, and estimates can change when a single, additional chip is added to the set. Additionally, outlier chips can impact the signals of other arrays, and can themselves be skewed by the majority of the population.ResultsWe developed an approach, termed IRON, which uses the best-performing techniques from each of several popular processing methods while retaining the ability to incrementally renormalize data without altering previously normalized expression. This combination of approaches results in a method that performs comparably to existing approaches on artificial benchmark datasets (i.e. spike-in) and demonstrates promising improvements in segregating true signals within biologically complex experiments.ConclusionsBy combining approaches from existing normalization techniques, the IRON method offers several advantages. First, IRON normalization occurs pair-wise, thereby avoiding the need for all chips to be normalized together, which can be important for large data analyses. Secondly, the technique does not require similarity in signal distribution across chips for normalization, which can be important for maintaining biologically relevant differences in a heterogeneous background. Lastly, IRON introduces fewer post-processing artifacts, particularly in data whose behavior violates common assumptions. Thus, the IRON method provides a practical solution to common needs of expression analysis. A software implementation of IRON is available at [http://gene.moffitt.org/libaffy/].


Cancer Epidemiology, Biomarkers & Prevention | 2011

Inherited Variants in Mitochondrial Biogenesis Genes May Influence Epithelial Ovarian Cancer Risk

Jennifer Permuth-Wey; Y. Ann Chen; Ya Yu Tsai; Zhihua Chen; Xiaotao Qu; Johnathan M. Lancaster; Heather G. Stockwell; Getachew A. Dagne; Edwin S. Iversen; Harvey A. Risch; Jill S. Barnholtz-Sloan; Julie M. Cunningham; Robert A. Vierkant; Brooke L. Fridley; Rebecca Sutphen; John R. McLaughlin; Steven A. Narod; Ellen L. Goode; Joellen M. Schildkraut; David Fenstermacher; Catherine M. Phelan; Thomas A. Sellers

Background: Mitochondria contribute to oxidative stress, a phenomenon implicated in ovarian carcinogenesis. We hypothesized that inherited variants in mitochondrial-related genes influence epithelial ovarian cancer (EOC) susceptibility. Methods: Through a multicenter study of 1,815 Caucasian EOC cases and 1,900 controls, we investigated associations between EOC risk and 128 single nucleotide polymorphisms (SNPs) from 22 genes/regions within the mitochondrial genome (mtDNA) and 2,839 nuclear-encoded SNPs localized to 138 genes involved in mitochondrial biogenesis (BIO, n = 35), steroid hormone metabolism (HOR, n = 13), and oxidative phosphorylation (OXP, n = 90) pathways. Unconditional logistic regression was used to estimate OR and 95% CI between genotype and case status. Overall significance of each gene and pathway was evaluated by using Fishers method to combine SNP-level evidence. At the SNP level, we investigated whether lifetime ovulation, hormone replacement therapy (HRT), and cigarette smoking were confounders or modifiers of associations. Results: Interindividual variation involving BIO was most strongly associated with EOC risk (empirical P = 0.050), especially for NRF1, MTERF, PPARGC1A, ESRRA, and CAMK2D. Several SNP-level associations strengthened after adjustment for nongenetic factors, particularly for MTERF. Statistical interactions with cigarette smoking and HRT use were observed with MTERF and CAMK2D SNPs, respectively. Overall variation within mtDNA, HOR, and OXP was not statistically significant (empirical P > 0.10). Conclusion: We provide novel evidence to suggest that variants in mitochondrial biogenesis genes may influence EOC susceptibility. Impact: A deeper understanding of the complex mechanisms implicated in mitochondrial biogenesis and oxidative stress may aid in developing strategies to reduce morbidity and mortality from EOC. Cancer Epidemiol Biomarkers Prev; 20(6); 1131–45. ©2011 AACR.


Scientific Reports | 2013

JAK1 truncating mutations in gynecologic cancer define new role of cancer-associated protein tyrosine kinase aberrations

Yuan Ren; Yonghong Zhang; Richard Z. Liu; David Fenstermacher; Kenneth L. Wright; Jamie K. Teer; Jie Wu

Cancer-associated protein tyrosine kinase (PTK) mutations usually are gain-of-function (GOF) mutations that drive tumor growth and metastasis. We have found 50 JAK1 truncating mutations in 36 of 635 gynecologic tumors in the Total Cancer Care® (TCC®) tumor bank. Among cancer cell lines containing JAK1 truncating mutations in the Cancer Cell Line Encyclopedia databank, 68% are gynecologic cancer cells. Within JAK1 the K142, P430, and K860 frame-shift mutations were identified as hot spot mutation sites. Sanger sequencing of cancer cell lines, primary tumors, and matched normal tissues confirmed the JAK1 mutations and showed that these mutations are somatic. JAK1 mediates interferon (IFN)-γ-regulated tumor immune surveillance. Functional assays show that JAK1 deficient cancer cells are defective in IFN-γ-induced LMP2 and TAP1 expression, loss of which inhibits presentation of tumor antigens. These findings identify recurrent JAK1 truncating mutations that could contribute to tumor immune evasion in gynecologic cancers, especially in endometrial cancer.


Journal of Personalized Medicine | 2012

Developing a prototype system for integrating pharmacogenomics findings into clinical practice.

Casey Lynnette Overby; Peter Tarczy-Hornoch; Ira J. Kalet; Kenneth E. Thummel; Joe W. Smith; Guilherme Del Fiol; David Fenstermacher; Emily Beth Devine

Findings from pharmacogenomics (PGx) studies have the potential to be applied to individualize drug therapy to improve efficacy and reduce adverse drug events. Researchers have identified factors influencing uptake of genomics in medicine, but little is known about the specific technical barriers to incorporating PGx into existing clinical frameworks. We present the design and development of a prototype PGx clinical decision support (CDS) system that builds on existing clinical infrastructure and incorporates semi-active and active CDS. Informing this work, we updated previous evaluations of PGx knowledge characteristics, and of how the CDS capabilities of three local clinical systems align with data and functional requirements for PGx CDS. We summarize characteristics of PGx knowledge and technical needs for implementing PGx CDS within existing clinical frameworks. PGx decision support rules derived from FDA drug labels primarily involve drug metabolizing genes, vary in maturity, and the majority support the post-analytic phase of genetic testing. Computerized provider order entry capabilities are key functional requirements for PGx CDS and were best supported by one of the three systems we evaluated. We identified two technical needs when building on this system, the need for (1) new or existing standards for data exchange to connect clinical data to PGx knowledge, and (2) a method for implementing semi-active CDS. Our analyses enhance our understanding of principles for designing and implementing CDS for drug therapy individualization and our current understanding of PGx characteristics in a clinical context. Characteristics of PGx knowledge and capabilities of current clinical systems can help govern decisions about CDS implementation, and can help guide decisions made by groups that develop and maintain knowledge resources such that delivery of content for clinical care is supported.


Cancer Epidemiology, Biomarkers & Prevention | 2011

MicroRNA Processing and Binding Site Polymorphisms Are Not Replicated in the Ovarian Cancer Association Consortium

Jennifer Permuth-Wey; Zhihua Chen; Ya Yu Tsai; Hui-Yi Lin; Y. Ann Chen; Jill S. Barnholtz-Sloan; Michael J. Birrer; Stephen J. Chanock; Daniel W. Cramer; Julie M. Cunningham; David Fenstermacher; Brooke L. Fridley; Montserrat Garcia-Closas; Simon A. Gayther; Aleksandra Gentry-Maharaj; Jesus Gonzalez-Bosquet; Edwin S. Iversen; Heather Jim; John R. McLaughlin; Usha Menon; Steven A. Narod; Catherine M. Phelan; Susan J. Ramus; Harvey A. Risch; Honglin Song; Rebecca Sutphen; Kathryn L. Terry; Jonathan Tyrer; Robert A. Vierkant; Nicolas Wentzensen

Background: Single nucleotide polymorphisms (SNP) in microRNA-related genes have been associated with epithelial ovarian cancer (EOC) risk in two reports, yet associated alleles may be inconsistent across studies. Methods: We conducted a pooled analysis of previously identified SNPs by combining genotype data from 3,973 invasive EOC cases and 3,276 controls from the Ovarian Cancer Association Consortium. We also conducted imputation to obtain dense coverage of genes and comparable genotype data for all studies. In total, 226 SNPs within 15 kb of 4 miRNA biogenesis genes (DDX20, DROSHA, GEMIN4, and XPO5) and 23 SNPs located within putative miRNA binding sites of 6 genes (CAV1, COL18A1, E2F2, IL1R1, KRAS, and UGT2A3) were genotyped or imputed and analyzed in the entire dataset. Results: After adjustment for European ancestry, no overall association was observed between any of the analyzed SNPs and EOC risk. Conclusions: Common variants in these evaluated genes do not seem to be strongly associated with EOC risk. Impact: This analysis suggests earlier associations between EOC risk and SNPs in these genes may have been chance findings, possibly confounded by population admixture. To more adequately evaluate the relationship between genetic variants and cancer risk, large sample sizes are needed, adjustment for population stratification should be carried out, and use of imputed SNP data should be considered. Cancer Epidemiol Biomarkers Prev; 20(8); 1793–7. ©2011 AACR.

Collaboration


Dive into the David Fenstermacher's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Rebecca Sutphen

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Thomas A. Sellers

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Zhihua Chen

University of South Florida

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge