Network


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

Hotspot


Dive into the research topics where Julie W. Reeser is active.

Publication


Featured researches published by Julie W. Reeser.


Human Mutation | 2015

Evaluation of Hybridization Capture Versus Amplicon-Based Methods for Whole-Exome Sequencing

Eric Samorodnitsky; Benjamin M. Jewell; Raffi Hagopian; Jharna Miya; Michele R. Wing; Ezra Lyon; Senthilkumar Damodaran; Darshna Bhatt; Julie W. Reeser; Jharna Datta; Sameek Roychowdhury

Next‐generation sequencing has aided characterization of genomic variation. While whole‐genome sequencing may capture all possible mutations, whole‐exome sequencing remains cost‐effective and captures most phenotype‐altering mutations. Initial strategies for exome enrichment utilized a hybridization‐based capture approach. Recently, amplicon‐based methods were designed to simplify preparation and utilize smaller DNA inputs. We evaluated two hybridization capture‐based and two amplicon‐based whole‐exome sequencing approaches, utilizing both Illumina and Ion Torrent sequencers, comparing on‐target alignment, uniformity, and variant calling. While the amplicon methods had higher on‐target rates, the hybridization capture‐based approaches demonstrated better uniformity. All methods identified many of the same single‐nucleotide variants, but each amplicon‐based method missed variants detected by the other three methods and reported additional variants discordant with all three other technologies. Many of these potential false positives or negatives appear to result from limited coverage, low variant frequency, vicinity to read starts/ends, or the need for platform‐specific variant calling algorithms. All methods demonstrated effective copy‐number variant calling when evaluated against a single‐nucleotide polymorphism array. This study illustrates some differences between whole‐exome sequencing approaches, highlights the need for selecting appropriate variant calling based on capture method, and will aid laboratories in selecting their preferred approach.


The Journal of Molecular Diagnostics | 2015

Comparison of custom capture for targeted next-generation DNA sequencing.

Eric Samorodnitsky; Jharna Datta; Benjamin M. Jewell; Raffi Hagopian; Jharna Miya; Michele R. Wing; Senthilkumar Damodaran; Juliana M. Lippus; Julie W. Reeser; Darshna Bhatt; Cynthia Timmers; Sameek Roychowdhury

Targeted, capture-based DNA sequencing is a cost-effective method to focus sequencing on a coding region or other customized region of the genome. There are multiple targeted sequencing methods available, but none has been systematically investigated and compared. We evaluated four commercially available custom-targeted DNA technologies for next-generation sequencing with respect to on-target sequencing, uniformity, and ability to detect single-nucleotide variations (SNVs) and copy number variations. The technologies that used sonication for DNA fragmentation displayed impressive uniformity of capture, whereas the others had shorter preparation times, but sacrificed uniformity. One of those technologies, which uses transposase for DNA fragmentation, has a drawback requiring sample pooling, and the last one, which uses restriction enzymes, has a limitation depending on restriction enzyme digest sites. Although all technologies displayed some level of concordance for calling SNVs, the technologies that require restriction enzymes or transposase missed several SNVs largely because of the lack of coverage. All technologies performed well for copy number variation calling when compared to single-nucleotide polymorphism arrays. These results enable laboratories to compare these methods to make informed decisions for their intended applications.


Molecular Cancer Therapeutics | 2017

Akt activation mediates acquired resistance to fibroblast growth factor receptor inhibitor BGJ398

Jharna Datta; Senthilkumar Damodaran; Hannah Parks; Cristina Ocrainiciuc; Jharna Miya; Lianbo Yu; Elijah P. Gardner; Eric Samorodnitsky; Michele R. Wing; Darshna Bhatt; John L. Hays; Julie W. Reeser; Sameek Roychowdhury

Activation of FGFR signaling through mutations, amplifications, or fusions involving FGFR1, 2, 3, or 4 is seen in multiple tumors, including lung, bladder, and cholangiocarcinoma. Currently, several clinical trials are evaluating the role of novel FGFR inhibitors in solid tumors. As we move forward with FGFR inhibitors clinically, we anticipate the emergence of resistance with treatment. Consequently, we sought to study the mechanism(s) of acquired resistance to FGFR inhibitors using annotated cancer cell lines. We identified cancer cell lines that have activating mutations in FGFR1, 2, or 3 and treated them chronically with the selective FGFR inhibitor, BGJ398. We observed resistance to chronic BGJ398 exposure in DMS114 (small-cell lung cancer, FGFR1 amplification) and RT112 (urothelial carcinoma, FGFR3 fusion/amplification) cell lines based on viability assays. Reverse-phase protein array (RPPA) analysis showed increased phosphorylation of Akt (T308 and S473) and its downstream target GSK3 (S9 and S21) in both the resistant cell lines when compared with matching controls. Results of RPPA were confirmed using immunoblots. Consequently, the addition of an Akt inhibitor (GSK2141795) or siRNA was able to restore sensitivity to BGJ398 in resistant cell lines. These data suggest a role for Akt pathway in mediating acquired resistance to FGFR inhibition. Mol Cancer Ther; 16(4); 614–24. ©2017 AACR.


Oncotarget | 2017

Performance evaluation for rapid detection of pan-cancer microsatellite instability with MANTIS

Esko A. Kautto; Russell Bonneville; Jharna Miya; Lianbo Yu; Melanie A. Krook; Julie W. Reeser; Sameek Roychowdhury

In current clinical practice, microsatellite instability (MSI) and mismatch repair deficiency detection is performed with MSI-PCR and immunohistochemistry. Recent research has produced several computational tools for MSI detection with next-generation sequencing (NGS) data; however a comprehensive analysis of computational methods has not yet been performed. In this study, we introduce a new MSI detection tool, MANTIS, and demonstrate its favorable performance compared to the previously published tools mSINGS and MSISensor. We evaluated 458 normal-tumor sample pairs across six cancer subtypes, testing classification performance on variable numbers of target loci ranging from 10 to 2539. All three computational methods were found to be accurate, with MANTIS exhibiting the highest accuracy with 98.91% of samples from all six diseases classified correctly. MANTIS displayed superior performance among the three tools, having the highest overall sensitivity (MANTIS 97.18%, MSISensor 96.48%, mSINGS 76.06%) and specificity (MANTIS 99.68%, mSINGS 99.68%, MSISensor 98.73%) across six cancer types, even with loci panels of varying size. Additionally, MANTIS also had the lowest resource consumption (<1% of the space and <7% of the memory required by mSINGS) and fastest running times (49.6% and 8.7% of the running times of MSISensor and mSINGS, respectively). This study highlights the potential utility of MANTIS in classifying samples by MSI-status, allowing its incorporation into existing NGS pipelines.


JCO Precision Oncology | 2017

Landscape of Microsatellite Instability Across 39 Cancer Types

Russell Bonneville; Melanie A. Krook; Esko A. Kautto; Jharna Miya; Michele R. Wing; Hui-Zi Chen; Julie W. Reeser; Lianbo Yu; Sameek Roychowdhury

Purpose Microsatellite instability (MSI) is a pattern of hypermutation that occurs at genomic microsatellites and is caused by defects in the mismatch repair system. Mismatch repair deficiency that leads to MSI has been well described in several types of human cancer, most frequently in colorectal, endometrial, and gastric adenocarcinomas. MSI is known to be both predictive and prognostic, especially in colorectal cancer; however, current clinical guidelines only recommend MSI testing for colorectal and endometrial cancers. Therefore, less is known about the prevalence and extent of MSI among other types of cancer. Methods Using our recently published MSI-calling software, MANTIS, we analyzed whole-exome data from 11,139 tumor-normal pairs from The Cancer Genome Atlas and Therapeutically Applicable Research to Generate Effective Treatments projects and external data sources across 39 cancer types. Within a subset of these cancer types, we assessed mutation burden, mutational signatures, and somatic variants associated with MSI. Results We identified MSI in 3.8% of all cancers assessed-present in 27 of tumor types-most notably adrenocortical carcinoma (ACC), cervical cancer (CESC), and mesothelioma, in which MSI has not yet been well described. In addition, MSI-high ACC and CESC tumors were observed to have a higher average mutational burden than microsatellite-stable ACC and CESC tumors. Conclusion We provide evidence of as-yet-unappreciated MSI in several types of cancer. These findings support an expanded role for clinical MSI testing across multiple cancer types as patients with MSI-positive tumors are predicted to benefit from novel immunotherapies in clinical trials.


The Journal of Molecular Diagnostics | 2017

Validation of a Targeted RNA Sequencing Assay for Kinase Fusion Detection in Solid Tumors

Julie W. Reeser; Dorrelyn Martin; Jharna Miya; Esko A. Kautto; Ezra Lyon; Eliot Zhu; Michele R. Wing; Amy Smith; Matthew Reeder; Eric Samorodnitsky; Hannah Parks; Karan R. Naik; Joseph M. Gozgit; Nicholas Nowacki; Kurtis D. Davies; Marileila Varella-Garcia; Lianbo Yu; Aharon G. Freud; Joshua Coleman; Dara L. Aisner; Sameek Roychowdhury

Kinase gene fusions are important drivers of oncogenic transformation and can be inhibited with targeted therapies. Clinical grade diagnostics using RNA sequencing to detect gene rearrangements in solid tumors are limited, and the few that are available require prior knowledge of fusion break points. To address this, we have analytically validated a targeted RNA sequencing assay (OSU-SpARKFuse) for fusion detection that interrogates complete transcripts from 93 kinase and transcription factor genes. From a total of 74 positive and 36 negative control samples, OSU-SpARKFuse had 93.3% sensitivity and 100% specificity for fusion detection. Assessment of repeatability and reproducibility revealed 96.3% and 94.4% concordance between intrarun and interrun technical replicates, respectively. Application of this assay on prospective patient samples uncovered OLFM4 as a novel RET fusion partner in a small-bowel cancer and led to the discovery of a KLK2-FGFR2 fusion in a patient with prostate cancer who subsequently underwent treatment with a pan-fibroblast growth factor receptor inhibitor. Beyond fusion detection, OSU-SpARKFuse has built-in capabilities for discovery research, including gene expression analysis, detection of single-nucleotide variants, and identification of alternative splicing events.


Leukemia & Lymphoma | 2018

Trametinib for the treatment of IGHV4-34, MAP2K1-mutant variant hairy cell leukemia

Leslie A. Andritsos; Nicole R. Grieselhuber; Mirela Anghelina; Kerry A. Rogers; Sameek Roychowdhury; Julie W. Reeser; Cynthia Timmers; Aharon G. Freud; James S. Blachly; David M. Lucas; Gerard Lozanski; Jeffrey A. Jones; Katie Williams; Christopher C. Oakes; Daniel Jones; Michael R. Grever

Leslie A. Andritsos , Nicole R. Grieselhuber , Mirela Anghelina, Kerry A. Rogers, Sameek Roychowdhury, Julie W. Reeser, Cynthia D. Timmers, Aharon G. Freud, James S. Blachly, David M. Lucas, Gerard Lozanski, Jeffrey A. Jones, Katie Williams, Christopher Oakes, Dan Jones and Michael R. Grever Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA; Division of Medical Oncology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA; Department of Pathology, The Ohio State University, Columbus, OH, USA


Journal of The National Comprehensive Cancer Network | 2018

Targeting BRAF Mutations in High-Grade Neuroendocrine Carcinoma of the Colon

Jarred Burkart; Dwight Owen; Manisha H. Shah; Sherif Abdel-Misih; Sameek Roychowdhury; Robert Wesolowski; Sigurdis Haraldsdottir; Julie W. Reeser; Eric Samorodnitsky; Amy Smith; Bhavana Konda

Mutations in the RAS/RAF/MEK/ERK pathway leading to constitutive activation and uncontrolled cellular growth have been identified in various human malignancies, making this pathway a target for potential therapeutics. The activating BRAFV600E mutation is one well-characterized oncogenic mutation that has been described and targeted with clinical success in various malignancies, including melanoma and hairy cell leukemia. Although BRAF-directed treatments have yielded clinical benefit in a subset of tumor types, such as melanoma, thyroid cancer, and lung cancer, BRAF inhibition fails to confer a clinical benefit in colon cancer. Identification of patients for whom BRAF inhibition may produce clinically meaningful outcomes is imperative. The incidence of BRAF mutations in neuroendocrine carcinoma (NEC) is estimated to be 5% to 10%. A recent case series demonstrated benefit in targeting the BRAFV600E mutation in metastatic high-grade rectal NECs. Combination BRAF and MEK inhibition is known to yield improved outcomes compared with BRAF inhibition alone in melanoma. This report presents 2 patients with high-grade colorectal NECs who had different responses to treatment with combined BRAF/MEK inhibition after experiencing disease progression through first-line platinum-based chemotherapy. One patient experienced an excellent initial response to therapy before ultimately experiencing progression, and in the other patient initially had stable disease before eventually experiencing progression. These cases highlight the complicated role BRAF mutations play in gastrointestinal NECs, and the need for further research to identify not only patients who may benefit from BRAF-directed therapies but also strategies to avoid development of resistance.


Cold Spring Harb Mol Case Stud | 2018

Metaplastic breast cancer in a patient with neurofibromatosis type 1 and somatic loss of heterozygosity

Lorena P. Suarez-Kelly; Keiko Akagi; Julie W. Reeser; Eric Samorodnitsky; Matthew Reeder; Amy Smith; Sameek Roychowdhury; David E. Symer; William E. Carson

Metaplastic breast carcinoma (MBC) is rare and has a poor prognosis. Here we describe genetic analysis of a 41-yr-old female patient with MBC and neurofibromatosis type I (NF1). She initially presented with pT3N1a, grade 3 MBC, but lung metastases were discovered subsequently. To identify the molecular cause of her NF1, we screened for germline mutations disrupting NF1 or SPRED1, revealing a heterozygous germline single-nucleotide variant (SNV) in exon 21 of NF1 at c.2709G>A, Chr 17: 29556342. By report, this variant disrupts pre-mRNA splicing of NF1 transcripts. No pathogenic mutations were identified in SPRED1. A potential association between MBC and NF1 was reported in eight previous cases, but none underwent detailed genomics analysis. To identify additional candidate germline variants potentially predisposing to MBC, we conducted targeted exome sequencing of 279 established cancer-causing genes in a control blood sample, disclosing four rare SNVs. Analysis of her breast tumor showed markedly altered variant allelic fractions (VAFs) for two (50%) of them, revealing somatic loss of heterozygosity (LOH) at germline SNVs. Of these, only the VAF of the pathogenic SNV in NF1 was increased in the tumor. Tumor sequencing demonstrated five somatic mutations altering TP53, BRCA1, and other genes potentially contributing to cancer formation. Because somatic LOH at certain germline SNVs can enhance their impacts, we conclude that increased allelic imbalance of the pathogenic SNV in NF1 likely contributed to tumorigenesis. Our results highlight a need to assess predisposing genetic factors and LOH that can cause rare, aggressive diseases such as MBC in NF1.


Oncotarget | 2017

Analytic validation and real-time clinical application of an amplicon-based targeted gene panel for advanced cancer

Michele R. Wing; Julie W. Reeser; Amy Smith; Matthew Reeder; Dorrelyn Martin; Benjamin M. Jewell; Jharna Datta; Jharna Miya; J. Paul Monk; Amir Mortazavi; Gregory A. Otterson; Richard M. Goldberg; Jeffrey B. VanDeusen; Sharon Cole; Kristin Dittmar; Sunny Jaiswal; Matthew Kinzie; Suraj Waikhom; Aharon G. Freud; Xiao-Ping Zhou; Wei Chen; Darshna Bhatt; Sameek Roychowdhury

Multiplex somatic testing has emerged as a strategy to test patients with advanced cancer. We demonstrate our analytic validation approach for a gene hotspot panel and real-time prospective clinical application for any cancer type. The TruSight Tumor 26 assay amplifies 85 somatic hotspot regions across 26 genes. Using cell line and tumor mixes, we observed that 100% of the 14,715 targeted bases had at least 1000x raw coverage. We determined the sensitivity (100%, 95% CI: 96-100%), positive predictive value (100%, 95% CI: 96-100%), reproducibility (100% concordance), and limit of detection (3% variant allele frequency at 1000x read depth) of this assay to detect single nucleotide variants and small insertions and deletions. Next, we applied the assay prospectively in a clinical tumor sequencing study to evaluate 174 patients with metastatic or advanced cancer, including frozen tumors, formalin-fixed tumors, and enriched peripheral blood mononuclear cells in hematologic cancers. We reported one or more somatic mutations in 89 (53%) of the sequenced tumors (167 passing quality filters). Forty-three of these patients (26%) had mutations that would enable eligibility for targeted therapies. This study demonstrates the validity and feasibility of applying TruSight Tumor 26 for pan-cancer testing using multiple specimen types.

Collaboration


Dive into the Julie W. Reeser's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Amy Smith

Ohio State University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge