Network


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

Hotspot


Dive into the research topics where Heather A. Parsons is active.

Publication


Featured researches published by Heather A. Parsons.


Journal of Biological Chemistry | 2004

DNA binding features of human POT1: a nonamer 5'-TAGGGTTAG-3' minimal binding site, sequence specificity, and internal binding to multimeric sites.

Diego Loayza; Heather A. Parsons; Jill R. Donigian; Kristina Hoke; Titia de Lange

The human telomeric protein POT1 is known to bind single-stranded telomeric DNA in vitro and to participate in the regulation of telomere maintenance by telomerase in vivo. We examined the in vitro DNA binding features of POT1. We report that deleting the oligosaccharide/oligonucleotide-binding fold of POT1 abrogates its DNA binding activity. The minimal binding site (MBS) for POT1 was found to be the telomeric nonamer 5′-TAGGGTTAG-3′, and the optimal substrate is [TTAGGG]n (n ≥ 2). POT1 displays exceptional sequence specificity when binding to MBS, tolerating changes only at position 7 (T7A). Whereas POT1 binding to MBS or [TTAGGG]2 was enhanced by the proximity of a 3′ end, POT1 was able to bind to a [TTAGGG]5 array when positioned internally. These data indicate that POT1 has a strong sequence preference for the human telomeric repeat tract and predict that POT1 can bind both the 3′ telomeric overhang and the displaced TTAGGG repeats at the base of the t-loop.


Clinical Cancer Research | 2016

ESR1 mutations in circulating plasma tumor DNA from metastatic breast cancer patients

David Chu; Costanza Paoletti; Christina Gersch; Dustin A. VanDenBerg; Daniel J. Zabransky; Rory L. Cochran; Hong Yuen Wong; Patricia Valda Toro; Justin Cidado; Sarah Croessmann; Bracha Erlanger; Karen Cravero; Kelly Kyker-Snowman; Berry Button; Heather A. Parsons; W. Brian Dalton; Riaz Gillani; Arielle Medford; Kimberly Aung; Nahomi Tokudome; Arul M. Chinnaiyan; Anne F. Schott; Dan R. Robinson; Karen S. Jacks; Josh Lauring; Paula J. Hurley; Daniel F. Hayes; James M. Rae; Ben Ho Park

Purpose: Mutations in the estrogen receptor (ER)α gene, ESR1, have been identified in breast cancer metastases after progression on endocrine therapies. Because of limitations of metastatic biopsies, the reported frequency of ESR1 mutations may be underestimated. Here, we show a high frequency of ESR1 mutations using circulating plasma tumor DNA (ptDNA) from patients with metastatic breast cancer. Experimental Design: We retrospectively obtained plasma samples from eight patients with known ESR1 mutations and three patients with wild-type ESR1 identified by next-generation sequencing (NGS) of biopsied metastatic tissues. Three common ESR1 mutations were queried for using droplet digital PCR (ddPCR). In a prospective cohort, metastatic tissue and plasma were collected contemporaneously from eight ER-positive and four ER-negative patients. Tissue biopsies were sequenced by NGS, and ptDNA ESR1 mutations were analyzed by ddPCR. Results: In the retrospective cohort, all corresponding mutations were detected in ptDNA, with two patients harboring additional ESR1 mutations not present in their metastatic tissues. In the prospective cohort, three ER-positive patients did not have adequate tissue for NGS, and no ESR1 mutations were identified in tissue biopsies from the other nine patients. In contrast, ddPCR detected seven ptDNA ESR1 mutations in 6 of 12 patients (50%). Conclusions: We show that ESR1 mutations can occur at a high frequency and suggest that blood can be used to identify additional mutations not found by sequencing of a single metastatic lesion. Clin Cancer Res; 22(4); 993–9. ©2015 AACR.


Clinical Biochemistry | 2015

Comparison of cell stabilizing blood collection tubes for circulating plasma tumor DNA

Patricia Valda Toro; Bracha Erlanger; Julia A. Beaver; Rory L. Cochran; Dustin A. VanDenBerg; Elizabeth Yakim; Karen Cravero; David Chu; Daniel J. Zabransky; Hong Yuen Wong; Sarah Croessmann; Heather A. Parsons; Paula J. Hurley; Josh Lauring; Ben Ho Park

OBJECTIVES Circulating plasma DNA is being increasingly used for biomedical and clinical research as a substrate for genetic testing. However, cell lysis can occur hours after venipuncture when using standard tubes for blood collection, leading to an increase in contaminating cellular DNA that may hinder analysis of circulating plasma DNA. Cell stabilization agents can prevent cellular lysis for several days, reducing the need for immediate plasma preparation after venipuncture, thereby facilitating the ease of blood collection and sample preparation for clinical research. However, the majority of cell stabilizing reagents have not been formally tested for their ability to preserve circulating plasma tumor DNA. DESIGN & METHODS In this study, we compared the properties of two cell stabilizing reagents, the cell-free DNA BCT tube and the PAXgene tube, by collecting blood samples from metastatic breast cancer patients and measuring genome equivalents of plasma DNA by droplet digital PCR. We compared wild type PIK3CA genome equivalents and also assayed for two PIK3CA hotspot mutations, E545K and H1047R. RESULTS Our results demonstrate that blood stored for 7 days in BCT tubes did not show evidence of cell lysis, whereas PAXgene tubes showed an order of magnitude increase in genome equivalents, indicative of considerable cellular lysis. CONCLUSIONS We conclude that BCT tubes can prevent lysis and cellular release of genomic DNA of blood samples from cancer patients when stored at room temperature, and could therefore be of benefit for blood specimen collections in clinical trials.


Nature Communications | 2017

Scalable whole-exome sequencing of cell-free DNA reveals high concordance with metastatic tumors

Viktor A. Adalsteinsson; Gavin Ha; Samuel S. Freeman; Atish D. Choudhury; Daniel G. Stover; Heather A. Parsons; Gregory Gydush; Sarah C. Reed; Denisse Rotem; Justin Rhoades; Denis Loginov; Dimitri Livitz; Daniel Rosebrock; Ignaty Leshchiner; Jaegil Kim; Chip Stewart; Mara Rosenberg; Joshua M. Francis; Cheng-Zhong Zhang; Ofir Cohen; Coyin Oh; Huiming Ding; Paz Polak; Max Lloyd; Sairah Mahmud; Karla Helvie; Margaret S. Merrill; Rebecca A. Santiago; Edward P. O’Connor; Seong Ho Jeong

Whole-exome sequencing of cell-free DNA (cfDNA) could enable comprehensive profiling of tumors from blood but the genome-wide concordance between cfDNA and tumor biopsies is uncertain. Here we report ichorCNA, software that quantifies tumor content in cfDNA from 0.1× coverage whole-genome sequencing data without prior knowledge of tumor mutations. We apply ichorCNA to 1439 blood samples from 520 patients with metastatic prostate or breast cancers. In the earliest tested sample for each patient, 34% of patients have ≥10% tumor-derived cfDNA, sufficient for standard coverage whole-exome sequencing. Using whole-exome sequencing, we validate the concordance of clonal somatic mutations (88%), copy number alterations (80%), mutational signatures, and neoantigens between cfDNA and matched tumor biopsies from 41 patients with ≥10% cfDNA tumor content. In summary, we provide methods to identify patients eligible for comprehensive cfDNA profiling, revealing its applicability to many patients, and demonstrate high concordance of cfDNA and metastatic tumor whole-exome sequencing.Identifying the mutational landscape of tumours from cell-free DNA in the blood could help diagnostics in cancer. Here, the authors present ichorCNA, software that quantifies tumour content in cell free DNA, and they demonstrate that cell-free DNA whole-exome sequencing is concordant with metastatic tumour whole-exome sequencing.


Proceedings of the National Academy of Sciences of the United States of America | 2015

HER2 missense mutations have distinct effects on oncogenic signaling and migration

Daniel J. Zabransky; Christopher L. Yankaskas; Rory L. Cochran; Hong Yuen Wong; Sarah Croessmann; David Chu; Shyam M. Kavuri; Monica Red Brewer; D. Marc Rosen; W. Brian Dalton; Ashley Cimino-Mathews; Karen Cravero; Berry Button; Kelly Kyker-Snowman; Justin Cidado; Bracha Erlanger; Heather A. Parsons; Kristen M. Manto; Ron Bose; Josh Lauring; Carlos L. Arteaga; Konstantinos Konstantopoulos; Ben Ho Park

Significance The discovery of human epidermal growth factor receptor 2 (HER2) missense mutations in breast and other cancers potentially make such tumors susceptible to current and future HER2-targeted therapies. However, the majority of HER2 mutations occur in HER2 nonamplified cancers, and whether these mutations will predict for sensitivity to HER2-directed therapies remains unknown. Using genome editing, the data presented here suggest that HER2 missense mutations are functionally distinct and require additional oncogenic input to impart cancerous phenotypes. These results suggest that HER2 missense mutations by themselves may not be reliable predictors of response to HER2-targeted therapies, a hypothesis currently being tested in genomically driven clinical trials. Recurrent human epidermal growth factor receptor 2 (HER2) missense mutations have been reported in human cancers. These mutations occur primarily in the absence of HER2 gene amplification such that most HER2-mutant tumors are classified as “negative” by FISH or immunohistochemistry assays. It remains unclear whether nonamplified HER2 missense mutations are oncogenic and whether they are targets for HER2-directed therapies that are currently approved for the treatment of HER2 gene-amplified breast cancers. Here we functionally characterize HER2 kinase and extracellular domain mutations through gene editing of the endogenous loci in HER2 nonamplified human breast epithelial cells. In in vitro and in vivo assays, the majority of HER2 missense mutations do not impart detectable oncogenic changes. However, the HER2 V777L mutation increased biochemical pathway activation and, in the context of a PIK3CA mutation, enhanced migratory features in vitro. However, the V777L mutation did not alter in vivo tumorigenicity or sensitivity to HER2-directed therapies in proliferation assays. Our results suggest the oncogenicity and potential targeting of HER2 missense mutations should be considered in the context of cooperating genetic alterations and provide previously unidentified insights into functional analysis of HER2 mutations and strategies to target them.


Clinical Cancer Research | 2017

Individualized Molecular Analyses Guide Efforts (IMAGE): A Prospective Study of Molecular Profiling of Tissue and Blood in Metastatic Triple-Negative Breast Cancer

Heather A. Parsons; Julia A. Beaver; Ashley Cimino-Mathews; Siraj M. Ali; Jennifer E. Axilbund; David Chu; Roisin M. Connolly; Rory L. Cochran; Sarah Croessmann; Travis A. Clark; Christopher D. Gocke; Stacie C. Jeter; Mark Kennedy; Josh Lauring; Justin Lee; Doron Lipson; Vincent A. Miller; Geoff Otto; Gary L. Rosner; Jeffrey S. Ross; Shannon Slater; Philip J. Stephens; Dustin A. VanDenBerg; Antonio C. Wolff; Lauren E. Young; Daniel J. Zabransky; Zhe Zhang; Jane Zorzi; Vered Stearns; Ben Ho Park

Purpose: The clinical utility of next-generation sequencing (NGS) in breast cancer has not been demonstrated. We hypothesized that we could perform NGS of a new biopsy from patients with metastatic triple-negative breast cancer (TNBC) in a clinically actionable timeframe. Experimental Design: We planned to enroll 40 patients onto a prospective study, Individualized Molecular Analyses Guide Efforts (IMAGE), to evaluate the feasibility of obtaining a new biopsy of a metastatic site, perform NGS (FoundationOne), and convene a molecular tumor board to formulate treatment recommendations within 28 days. We collected blood at baseline and at time of restaging to assess cell-free circulating plasma tumor DNA (ptDNA). Results: We enrolled 26 women with metastatic TNBC who had received ≥1 line of prior chemotherapy, and 20 (77%) underwent NGS of a metastatic site biopsy. Twelve (60%) evaluable patients received treatment recommendations within 28 days of consent. The study closed after 20 patients underwent NGS, based on protocol-specified interim futility analysis. Three patients went on to receive genomically directed therapies. Twenty-four of 26 patients had genetic alterations successfully detected in ptDNA. Among 5 patients, 4 mutations found in tumor tissues were not identified in blood, and 4 mutations found in blood were not found in corresponding tumors. In 9 patients, NGS of follow-up blood samples showed 100% concordance with baseline blood samples. Conclusions: This study demonstrates challenges of performing NGS on prospective tissue biopsies in patients with metastatic TNBC within 28 days, while also highlighting the potential use of blood as a more time-efficient and less invasive method of mutational assessment. Clin Cancer Res; 23(2); 379–86. ©2016 AACR.


Proceedings of the National Academy of Sciences of the United States of America | 2015

NDRG1 links p53 with proliferation-mediated centrosome homeostasis and genome stability

Sarah Croessmann; Hong Yuen Wong; Daniel J. Zabransky; David Chu; Janet Mendonca; Anup Sharma; Morassa Mohseni; D. Marc Rosen; Robert B. Scharpf; Justin Cidado; Rory L. Cochran; Heather A. Parsons; W. Brian Dalton; Bracha Erlanger; Berry Button; Karen Cravero; Kelly Kyker-Snowman; Julia A. Beaver; Sushant K. Kachhap; Paula J. Hurley; Josh Lauring; Ben Ho Park

Significance The mechanism of how loss of the tumor suppressor p53 can lead to genomic instability is not fully understood. This study demonstrates that under physiologic low levels of proliferation, homozygous loss of tumor protein 53 (TP53) via genome editing, but not common p53 missense mutations, results in an inability to increase expression of N-Myc down-regulated gene 1 (NDRG1). In turn, failure to upregulate NDRG1 protein under low proliferative states leads to supernumerary centrosome formation, a known mechanism of aneuploidy. These results provide a mechanistic link between loss of TP53, proliferation, NDRG1, and genomic instability and help explain how cells with a low proliferative index and p53 loss can acquire additional genetic alterations that lead to cancer. The tumor protein 53 (TP53) tumor suppressor gene is the most frequently somatically altered gene in human cancers. Here we show expression of N-Myc down-regulated gene 1 (NDRG1) is induced by p53 during physiologic low proliferative states, and mediates centrosome homeostasis, thus maintaining genome stability. When placed in physiologic low-proliferating conditions, human TP53 null cells fail to increase expression of NDRG1 compared with isogenic wild-type controls and TP53 R248W knockin cells. Overexpression and RNA interference studies demonstrate that NDRG1 regulates centrosome number and amplification. Mechanistically, NDRG1 physically associates with γ-tubulin, a key component of the centrosome, with reduced association in p53 null cells. Strikingly, TP53 homozygous loss was mutually exclusive of NDRG1 overexpression in over 96% of human cancers, supporting the broad applicability of these results. Our study elucidates a mechanism of how TP53 loss leads to abnormal centrosome numbers and genomic instability mediated by NDRG1.


Clinical Chemistry | 2017

Multiplexed Elimination of Wild-Type DNA and High-Resolution Melting Prior to Targeted Resequencing of Liquid Biopsies

Ioannis Ladas; Mariana Fitarelli-Kiehl; Chen Song; Viktor A. Adalsteinsson; Heather A. Parsons; Nan Lin; Nikhil Wagle; G. Mike Makrigiorgos

BACKGROUND The use of clinical samples and circulating cell-free DNA (cfDNA) collected from liquid biopsies for diagnostic and prognostic applications in cancer is burgeoning, and improved methods that reduce the influence of excess wild-type (WT) portion of the sample are desirable. Here we present enrichment of mutation-containing sequences using enzymatic degradation of WT DNA. Mutation enrichment is combined with high-resolution melting (HRM) performed in multiplexed closed-tube reactions as a rapid, cost-effective screening tool before targeted resequencing. METHODS We developed a homogeneous, closed-tube approach to use a double-stranded DNA-specific nuclease for degradation of WT DNA at multiple targets simultaneously. The No Denaturation Nuclease-assisted Minor Allele Enrichment with Probe Overlap (ND-NaME-PrO) uses WT oligonucleotides overlapping both strands on putative DNA targets. Under conditions of partial denaturation (DNA breathing), the oligonucleotide probes enhance double-stranded DNA-specific nuclease digestion at the selected targets, with high preference toward WT over mutant DNA. To validate ND-NaME-PrO, we used multiplexed HRM, digital PCR, and MiSeq targeted resequencing of mutated genomic DNA and cfDNA. RESULTS Serial dilution of KRAS mutation-containing DNA shows mutation enrichment by 10- to 120-fold and detection of allelic fractions down to 0.01%. Multiplexed ND-NaME-PrO combined with multiplexed PCR-HRM showed mutation scanning of 10-20 DNA amplicons simultaneously. ND-NaME-PrO applied on cfDNA from clinical samples enables mutation enrichment and HRM scanning over 10 DNA targets. cfDNA mutations were enriched up to approximately 100-fold (average approximately 25-fold) and identified via targeted resequencing. CONCLUSIONS Closed-tube homogeneous ND-NaME-PrO combined with multiplexed HRM is a convenient approach to efficiently enrich for mutations on multiple DNA targets and to enable prescreening before targeted resequencing.


Oncotarget | 2015

Functional isogenic modeling of BRCA1 alleles reveals distinct carrier phenotypes

Rory L. Cochran; Justin Cidado; Minsoo Kim; Daniel J. Zabransky; Sarah Croessmann; David Chu; Hong Yuen Wong; Julia A. Beaver; Karen Cravero; Bracha Erlanger; Heather A. Parsons; Christopher M. Heaphy; Alan K. Meeker; Josh Lauring; Ben Ho Park

Clinical genetic testing of BRCA1 and BRCA2 is commonly performed to identify specific individuals at risk for breast and ovarian cancers who may benefit from prophylactic therapeutic interventions. Unfortunately, it is evident that deleterious BRCA1 alleles demonstrate variable penetrance and that many BRCA1 variants of unknown significance (VUS) exist. In order to further refine hereditary risks that may be associated with specific BRCA1 alleles, we performed gene targeting to establish an isogenic panel of immortalized human breast epithelial cells harboring eight clinically relevant BRCA1 alleles. Interestingly, BRCA1 mutations and VUS had distinct, quantifiable phenotypes relative to isogenic parental BRCA1 wild type cells and controls. Heterozygous cells with known deleterious BRCA1 mutations (185delAG, C61G and R71G) demonstrated consistent phenotypes in radiation sensitivity and genomic instability assays, but showed variability in other assays. Heterozygous BRCA1 VUS cells also demonstrated assay variability, with some VUS demonstrating phenotypes more consistent with deleterious alleles. Taken together, our data suggest that BRCA1 deleterious mutations and VUS can differ in their range of tested phenotypes, suggesting they might impart varying degrees of risk. These results demonstrate that functional isogenic modeling of BRCA1 alleles could aid in classifying BRCA1 mutations and VUS, and determining BRCA allele cancer risk.


JCO Precision Oncology | 2017

Personalized Medicine in the Oncology Clinic: Implementation and Outcomes of the Johns Hopkins Molecular Tumor Board

W. Brian Dalton; Patrick M. Forde; Hyunseok Kang; Roisin M. Connolly; Vered Stearns; Christopher D. Gocke; James R. Eshleman; Jennifer E. Axilbund; Dana Petry; Cindy Geoghegan; Antonio C. Wolff; David M. Loeb; Christine A. Pratilas; Christian Meyer; Eric S. Christenson; Shannon Slater; Jennifer Ensminger; Heather A. Parsons; Ben Ho Park; Josh Lauring

Purpose Tumor genomic profiling for personalized oncology therapy is being widely applied in clinical practice even as it is being evaluated more formally in clinical trials. Given the complexities of genomic data and its application to clinical use, molecular tumor boards with diverse expertise can provide guidance to oncologists and patients seeking to implement personalized genetically targeted therapy in practice. Methods A multidisciplinary molecular tumor board reviewed tumor molecular profiling reports from consecutive referrals at the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins over a 3-year period. The tumor board weighed evidence for actionability of genomic alterations identified by molecular profiling and provided recommendations including US Food and Drug Administration-approved drug therapy, clinical trials of matched targeted therapy, off-label use of such therapy, and additional tumor or germline genetic testing. Results One hundred fifty-five patients were reviewed. Actionable genomic alterations were identified in 132 patients (85%). Off-label therapies were recommended in 37 patients (24%). Eleven patients were treated off-label, and 13 patients were enrolled onto clinical trials of matched targeted therapies. Median progression-free survival of patients treated with matched therapies was 5 months (95% CI, 2.9 months to not reached), and the progression-free survival probability at 6 months was 43%(95% CI, 26% to 71%). Lack of locally available clinical trials was the major limitation on clinical actionability of tumor profiling reports. Conclusion The molecular tumor board recommended off-label targeted therapies for a quarter of all patients reviewed. Outcomes were heterogeneous, although 43% of patients receiving genomically matched therapy derived clinical benefit lasting at least 6 months. Until more data become available from precision oncology trials, molecular tumor boards can help guide appropriate use of tumor molecular testing to direct therapy.

Collaboration


Dive into the Heather A. Parsons's collaboration.

Top Co-Authors

Avatar

Ben Ho Park

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Daniel J. Zabransky

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

David Chu

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Josh Lauring

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Rory L. Cochran

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Sarah Croessmann

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Bracha Erlanger

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge