Network


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

Hotspot


Dive into the research topics where Tammy M. Havener is active.

Publication


Featured researches published by Tammy M. Havener.


Pharmacogenomics | 2011

Pharmacogenomic characterization of US FDA-approved cytotoxic drugs

Eric J Peters; Alison A. Motsinger-Reif; Tammy M. Havener; Lorraine Everitt; Nicholas E. Hardison; Venita Gresham Watson; Michael J. Wagner; Kristy L. Richards; M. A. Province; Howard L. McLeod

AIMS Individualization of cancer chemotherapy based on the patients genetic makeup holds promise for reducing side effects and improving efficacy. However, the relative contribution of genetics to drug response is unknown. MATERIALS & METHODS In this study, we investigated the cytotoxic effect of 29 commonly prescribed chemotherapeutic agents from diverse drug classes on 125 lymphoblastoid cell lines derived from 14 extended families. RESULTS The results of this systematic study highlight the variable role that genetics plays in response to cytotoxic drugs, ranging from a heritability of <0.15 for gemcitabine to >0.60 for epirubicin. CONCLUSION Putative quantitative trait loci for cytotoxic response were identified, as well as drug class-specific signatures, which could indicate possible shared genetic mechanisms. In addition to the identification of putative quantitative trait locis, the results of this study inform the prioritization of chemotherapeutic drugs with a sizable genetic response component for future investigation.


Pharmacogenetics and Genomics | 2012

A genome-wide association analysis of temozolomide response using lymphoblastoid cell lines shows a clinically relevant association with MGMT.

Chad Brown; Tammy M. Havener; Marisa W. Medina; J Todd Auman; Lara M. Mangravite; Ronald M. Krauss; Howard L. McLeod; Alison A. Motsinger-Reif

Objective Recently, lymphoblastoid cell lines (LCLs) have emerged as an innovative model system for mapping gene variants that predict the dose response to chemotherapy drugs. Methods In the current study, this strategy was expanded to the in-vitro genome-wide association approach, using 516 LCLs derived from a White cohort to assess the cytotoxic response to temozolomide. Results Genome-wide association analysis using ∼2.1 million quality-controlled single-nucleotide polymorphisms (SNPs) identified a statistically significant association (P<10−8) with SNPs in the O6-methylguanine-DNA methyltransferase (MGMT) gene. We also show that the primary SNP in this region is significantly associated with the differential gene expression of MGMT (P<10–26) in LCLs and differential methylation in glioblastoma samples from The Cancer Genome Atlas. Conclusion The previously documented clinical and functional relationships between MGMT and temozolomide response highlight the potential of well-powered genome-wide association studies of the LCL model system to identify meaningful genetic associations.


Pharmacogenomics | 2014

Genome-wide association and pharmacological profiling of 29 anticancer agents using lymphoblastoid cell lines

Chad Brown; Tammy M. Havener; Marisa W. Medina; John Jack; Ronald M. Krauss; Howard L. McLeod; Alison A. Motsinger-Reif

AIM Association mapping with lymphoblastoid cell lines (LCLs) is a promising approach in pharmacogenomics research, and in the current study we utilized LCLs to perform association mapping for 29 chemotherapy drugs. MATERIALS & METHODS Currently, we use LCLs to perform genome-wide association mapping of the cytotoxic response of 520 European-Americans to 29 different anticancer drugs; the largest LCL study to date. A novel association approach using a multivariate analysis of covariance design was employed with the software program MAGWAS, testing for differences in the dose-response profiles between genotypes without making assumptions about the response curve or the biologic mode of association. Additionally, by classifying 25 of the 29 drugs into eight families according to structural and mechanistic relationships, MAGWAS was used to test for associations that were shared across each drug family. Finally, a unique algorithm using multivariate responses and multiple linear regressions across pairs of response curves was used for unsupervised clustering of drugs. RESULTS Among the single-drug studies, suggestive associations were obtained for 18 loci, 12 within/near genes. Three of these, MED12L, CHN2 and MGMT, have been previously implicated in cancer pharmacogenomics. The drug family associations resulted in four additional suggestive loci (three contained within/near genes). One of these genes, HDAC4, associated with the DNA alkylating agents, shows possible clinical interactions with temozolomide. For the drug clustering analysis, 18 of 25 drugs clustered into the appropriate family. CONCLUSION This study demonstrates the utility of LCLs in identifying genes that have clinical importance in drug response and for assigning unclassified agents to specific drug families, and proposes new candidate genes for follow-up in a large number of chemotherapy drugs.


Biodata Mining | 2012

Multivariate methods and software for association mapping in dose-response genome-wide association studies.

Chad Brown; Tammy M. Havener; Marisa W. Medina; Ronald M. Krauss; Howard L. McLeod; Alison A. Motsinger-Reif

BackgroundThe large sample sizes, freedom of ethical restrictions and ease of repeated measurements make cytotoxicity assays of immortalized lymphoblastoid cell lines a powerful new in vitro method in pharmacogenomics research. However, previous studies may have over‐simplified the complex differences in dose‐response profiles between genotypes, resulting in a loss of power.MethodsThe current study investigates four previously studied methods, plus one new method based on a multivariate analysis of variance (MANOVA) design. A simulation study was performed using differences in cancer drug response between genotypes for biologically meaningful loci. These loci also showed significance in separate genome‐wide association studies. This manuscript builds upon a previous study, where differences in dose‐response curves between genotypes were constructed using the hill slope equation.ConclusionOverall, MANOVA was found to be the most powerful method for detecting real signals, and was also the most robust method for detection using alternatives generated with the previous simulation study. This method is also attractive because test statistics follow their expected distributions under the null hypothesis for both simulated and real data. The success of this method inspired the creation of the software program MAGWAS. MAGWAS is a computationally efficient, user‐friendly, open source software tool that works on most platforms and performs GWASs for individuals having multivariate responses using standard file formats.


Frontiers in Genetics | 2011

A Comparison of Association Methods for Cytotoxicity Mapping in Pharmacogenomics

Chad Brown; Tammy M. Havener; Lorraine Everitt; Howard L. McLeod; Alison A. Motsinger-Reif

Cytotoxicity assays of immortalized lymphoblastoid cell lines (LCLs) represent a promising new in vitro approach in pharmacogenomics research. However, previous studies employing LCLs in gene mapping have used simple association methods, which may not adequately capture the true differences in non-linear response profiles between genotypes. Two common approaches summarize each dose-response curve with either the IC50 or the slope parameter estimates from a hill slope fit and treat these estimates as the response in a linear model. The current study investigates these two methods, as well as four novel methods, and compares their power to detect differences between the response profiles of genotypes under a variety of different alternatives. The four novel methods include two methods that summarize each dose-response by its area under the curve, one method based off of an analysis of variance (ANOVA) design, and one method that compares hill slope fits for all individuals of each genotype. The power of each method was found to depend not only on the choice of alternative, but also on the choice for the set of dosages used in cytotoxicity measurements. The ANOVA-based method was found to be the most robust across alternatives and dosage sets for power in detecting differences between genotypes.


Clinical Pharmacology & Therapeutics | 2018

Genetic Variants in HSD17B3, SMAD3, and IPO11 Impact Circulating Lipids in Response to Fenofibrate in Individuals With Type 2 Diabetes

Daniel M. Rotroff; Sonja S. Pijut; Skylar W. Marvel; John Jack; Tammy M. Havener; Aurora Pujol; Agatha Schlüter; Gregory A. Graf; Henry N. Ginsberg; Hetal Shah; He Gao; Mario‐Luca Morieri; Alessandro Doria; Josyf C. Mychaleckyi; Howard L. McLeod; John B. Buse; Michael Wagner; Alison A. Motsinger-Reif

Individuals with type 2 diabetes (T2D) and dyslipidemia are at an increased risk of cardiovascular disease. Fibrates are a class of drugs prescribed to treat dyslipidemia, but variation in response has been observed. To evaluate common and rare genetic variants that impact lipid responses to fenofibrate in statin‐treated patients with T2D, we examined lipid changes in response to fenofibrate therapy using a genomewide association study (GWAS). Associations were followed‐up using gene expression studies in mice. Common variants in SMAD3 and IPO11 were marginally associated with lipid changes in black subjects (P < 5 × 10‐6). Rare variant and gene expression changes were assessed using a false discovery rate approach. AKR7A3 and HSD17B13 were associated with lipid changes in white subjects (q < 0.2). Mice fed fenofibrate displayed reductions in Hsd17b13 gene expression (q < 0.1). Associations of variants in SMAD3, IPO11, and HSD17B13, with gene expression changes in mice indicate that transforming growth factor‐beta (TGF‐β) and NRF2 signaling pathways may influence fenofibrate effects on dyslipidemia in patients with T2D.


BMC Research Notes | 2014

Application of next generation sequencing to CEPH cell lines to discover variants associated with FDA approved chemotherapeutics

Gunjan D. Hariani; Ernest J. Lam; Tammy M. Havener; Pui-Yan Kwok; Howard L. McLeod; Michael J. Wagner; Alison A. Motsinger-Reif

BackgroundThe goal of this study was to perform candidate gene association with cytotoxicity of chemotherapeutics in cell line models through resequencing and discovery of rare and low frequency variants along with common variations. Here, an association study of cytotoxicity response to 30 FDA approved drugs was conducted and we applied next generation targeted sequencing technology to discover variants from 103 candidate genes in 95 lymphoblastoid cell lines from 14 CEPH pedigrees. In this article, we called variants across 95 cell lines and performed association analysis for cytotoxic response using the Family Based Association Testing method and software.ResultsWe called 2281 variable SNP genotypes across the 103 genes for these cell lines and identified three genes of significant association within this marker set. Specifically, ATP-binding cassette, sub-family C, member 5 (ABCC5), metallothionein 1A (MT1A) and NAD(P)H dehydrogenase quinone1 (NQO1) were significantly associated with oxaliplatin drug response. The significant SNP on NQO1 (rs1800566) has been linked with poor survival rates in patients with non-small cell lung cancer treated with cisplatin (which belongs to the same class of drugs as oxaliplatin). A SNP (rs1846692) near the 5′ region of MT1A was associated with arsenic trioxide.ConclusionsThe results from this study are promising and this serves as a proof-of-principle demonstration of the use of sequencing data in the cytotoxicity models of human cell lines. With increased sample sizes, such studies will be a fast and powerful way to associate common and rare variants with drug response; while overcoming the cost and time limitations to recruit cohorts for association study.


PeerJ | 2017

Common and rare genetic markers of lipid variation in subjects with type 2 diabetes from the ACCORD clinical trial

Skylar W. Marvel; Daniel M. Rotroff; Michael J. Wagner; John B. Buse; Tammy M. Havener; Howard L. McLeod; Alison A. Motsinger-Reif

Background Individuals with type 2 diabetes are at an increased risk of cardiovascular disease. Alterations in circulating lipid levels, total cholesterol (TC), low-density lipoprotein (LDL), high-density lipoprotein (HDL), and triglycerides (TG) are heritable risk factors for cardiovascular disease. Here we conduct a genome-wide association study (GWAS) of common and rare variants to investigate associations with baseline lipid levels in 7,844 individuals with type 2 diabetes from the ACCORD clinical trial. Methods DNA extracted from stored blood samples from ACCORD participants were genotyped using the Affymetrix Axiom Biobank 1 Genotyping Array. After quality control and genotype imputation, association of common genetic variants (CV), defined as minor allele frequency (MAF) ≥ 3%, with baseline levels of TC, LDL, HDL, and TG was tested using a linear model. Rare variant (RV) associations (MAF < 3%) were conducted using a suite of methods that collapse multiple RV within individual genes. Results Many statistically significant CV (p < 1 × 10−8) replicate findings in large meta-analyses in non-diabetic subjects. RV analyses also confirmed findings in other studies, whereas significant RV associations with CNOT2, HPN-AS1, and SIRPD appear to be novel (q < 0.1). Discussion Here we present findings for the largest GWAS of lipid levels in people with type 2 diabetes to date. We identified 17 statistically significant (p < 1 × 10−8) associations of CV with lipid levels in 11 genes or chromosomal regions, all of which were previously identified in meta-analyses of mostly non-diabetic cohorts. We also identified 13 associations in 11 genes based on RV, several of which represent novel findings.


Diabetes | 2018

Genetic Variants in CPA6 and PRPF31 Are Associated With Variation in Response to Metformin in Individuals With Type 2 Diabetes

Daniel M. Rotroff; Sook Wah Yee; Kaixin Zhou; Skylar W. Marvel; Hetal Shah; John Jack; Tammy M. Havener; Monique M. Hedderson; Michiaki Kubo; Mark A. Herman; He Gao; Josyf C. Mychaleckyi; Howard L. McLeod; Alessandro Doria; Kathleen M. Giacomini; Ewan R. Pearson; Michael J. Wagner; John B. Buse; Alison A. Motsinger-Reif; MetGen Investigators; Accord; ACCORDion Investigators

Metformin is the first-line treatment for type 2 diabetes (T2D). Although widely prescribed, the glucose-lowering mechanism for metformin is incompletely understood. Here, we used a genome-wide association approach in a diverse group of individuals with T2D from the Action to Control Cardiovascular Risk in Diabetes (ACCORD) clinical trial to identify common and rare variants associated with HbA1c response to metformin treatment and followed up these findings in four replication cohorts. Common variants in PRPF31 and CPA6 were associated with worse and better metformin response, respectively (P < 5 × 10−6), and meta-analysis in independent cohorts displayed similar associations with metformin response (P = 1.2 × 10−8 and P = 0.005, respectively). Previous studies have shown that PRPF31(+/−) knockout mice have increased total body fat (P = 1.78 × 10−6) and increased fasted circulating glucose (P = 5.73 × 10−6). Furthermore, rare variants in STAT3 associated with worse metformin response (q <0.1). STAT3 is a ubiquitously expressed pleiotropic transcriptional activator that participates in the regulation of metabolism and feeding behavior. Here, we provide novel evidence for associations of common and rare variants in PRPF31, CPA6, and STAT3 with metformin response that may provide insight into mechanisms important for metformin efficacy in T2D.


Pharmacogenomics | 2010

Institutional Profile: UNC Institute for Pharmacogenomics and Individualized Therapy: interdisciplinary research for individual care

Tejinder Rakhra-Burris; J Todd Auman; Patricia A. Deverka; Lynn G. Dressler; James P. Evans; Richard M. Goldberg; Tammy M. Havener; Janelle M. Hoskins; Daniel E. Jonas; Kevin M. Long; Alison A. Motsinger-Reif; William J. Irvin; Kristy L. Richards; Mary W Roederer; John Valgus; Marcia van Riper; John A. Vernon; William C. Zamboni; Michael J. Wagner; Christine M. Walko; Karen E. Weck; Tim Wiltshire; Howard L. McLeod

The Institute for Pharmacogenomics and Individualized Therapy (IPIT) at the University of North Carolina at Chapel Hill (NC, USA) is a collaborative, multidisciplinary unit that brings together faculty from different disciplines and crosses the traditional departmental/school structure to perform pharmacogenomics research. IPIT investigators work together towards the goal of developing therapies to enable the delivery of individualized medical care. The NIH-supported Comprehensive Research on Expressed Alleles in Therapeutic Evaluation (CREATE) group leads the field in the evaluation of pathways regulating drug activity, and also provides a foundation for future IPIT research. IPIT members perform bench research, clinical cohort analysis and prospective clinical intervention studies, research on the integration of pharmacogenomic therapy into practice and research to foster global health pharmacogenomics application through the Pharmacogenetics for Every Nation Initiative. IPIT Investigators are actively incorporating a pharmacogenomics curriculum into existing teaching programs at all levels.

Collaboration


Dive into the Tammy M. Havener's collaboration.

Top Co-Authors

Avatar

Alison A. Motsinger-Reif

North Carolina State University

View shared research outputs
Top Co-Authors

Avatar

Howard L. McLeod

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

John Jack

North Carolina State University

View shared research outputs
Top Co-Authors

Avatar

Michael J. Wagner

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Chad Brown

North Carolina State University

View shared research outputs
Top Co-Authors

Avatar

Daniel M. Rotroff

North Carolina State University

View shared research outputs
Top Co-Authors

Avatar

John B. Buse

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Kristy L. Richards

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Skylar W. Marvel

North Carolina State University

View shared research outputs
Top Co-Authors

Avatar

Lorraine Everitt

University of North Carolina at Chapel Hill

View shared research outputs
Researchain Logo
Decentralizing Knowledge