Network


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

Hotspot


Dive into the research topics where Daniel J. Zabransky is active.

Publication


Featured researches published by Daniel J. Zabransky.


Clinical Cancer Research | 2014

Detection of Cancer DNA in Plasma of Patients with Early-Stage Breast Cancer

Julia A. Beaver; Danijela Jelovac; Sasidharan Balukrishna; Rory L. Cochran; Sarah Croessmann; Daniel J. Zabransky; Hong Yuen Wong; Patricia Valda Toro; Justin Cidado; Brian G. Blair; David Chu; Timothy F. Burns; Michaela J. Higgins; Vered Stearns; Lisa K. Jacobs; Mehran Habibi; Julie R. Lange; Paula J. Hurley; Josh Lauring; Dustin A. VanDenBerg; Jill Kessler; Stacie Jeter; Michael L. Samuels; Dianna Maar; Leslie Cope; Ashley Cimino-Mathews; Pedram Argani; Antonio C. Wolff; Ben Ho Park

Purpose: Detecting circulating plasma tumor DNA (ptDNA) in patients with early-stage cancer has the potential to change how oncologists recommend systemic therapies for solid tumors after surgery. Droplet digital polymerase chain reaction (ddPCR) is a novel sensitive and specific platform for mutation detection. Experimental Design: In this prospective study, primary breast tumors and matched pre- and postsurgery blood samples were collected from patients with early-stage breast cancer (n = 29). Tumors (n = 30) were analyzed by Sanger sequencing for common PIK3CA mutations, and DNA from these tumors and matched plasma were then analyzed for PIK3CA mutations using ddPCR. Results: Sequencing of tumors identified seven PIK3CA exon 20 mutations (H1047R) and three exon 9 mutations (E545K). Analysis of tumors by ddPCR confirmed these mutations and identified five additional mutations. Presurgery plasma samples (n = 29) were then analyzed for PIK3CA mutations using ddPCR. Of the 15 PIK3CA mutations detected in tumors by ddPCR, 14 of the corresponding mutations were detected in presurgical ptDNA, whereas no mutations were found in plasma from patients with PIK3CA wild-type tumors (sensitivity 93.3%, specificity 100%). Ten patients with mutation-positive ptDNA presurgery had ddPCR analysis of postsurgery plasma, with five patients having detectable ptDNA postsurgery. Conclusions: This prospective study demonstrates accurate mutation detection in tumor tissues using ddPCR, and that ptDNA can be detected in blood before and after surgery in patients with early-stage breast cancer. Future studies can now address whether ptDNA detected after surgery identifies patients at risk for recurrence, which could guide chemotherapy decisions for individual patients. Clin Cancer Res; 20(10); 2643–50. ©2014 AACR.


PLOS ONE | 2012

Phenotypic and Functional Properties of Helios+ Regulatory T Cells

Daniel J. Zabransky; Christopher Nirschl; Nicholas M. Durham; Ben V. Park; Christina M. Ceccato; Tullia C. Bruno; Ada J. Tam; Derese Getnet; Charles G. Drake

Helios, an Ikaros family transcription factor, is preferentially expressed at the mRNA and protein level in regulatory T cells. Helios expression previously appeared to be restricted to thymic-derived Treg. Consistent with recent data, we show here that Helios expression is inducible in vitro under certain conditions. To understand phenotypic and functional differences between Helios+ and Helios− Treg, we profiled cell-surface markers of FoxP3+ Treg using unmanipulated splenocytes. We found that CD103 and GITR are expressed at high levels on a subset of Helios+ Treg and that a Helios+ Treg population could be significantly enriched by FACS sorting using these two markers. Quantitative real-time PCR (qPCR) analysis revealed increased TGF-β message in Helios+ Treg, consistent with the possibility that this population possesses enhanced regulatory potential. In tumor-bearing mice, we found that Helios+ Treg were relatively over-represented in the tumor-mass, and BrdU studies showed that, in vivo, Helios+ Treg proliferated more than Helios− Treg. We hypothesized that Helios-enriched Treg might exert increased suppressive effects. Using in vitro suppression assays, we show that Treg function correlates with the absolute number of Helios+ cells in culture. Taken together, these data show that Helios+ Treg represent a functional subset with associated CD103 and GITR expression.


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.


Proceedings of the National Academy of Sciences of the United States of America | 2014

MACROD2 overexpression mediates estrogen independent growth and tamoxifen resistance in breast cancers

Morassa Mohseni; Justin Cidado; Sarah Croessmann; Karen Cravero; Ashley Cimino-Mathews; Hong Yuen Wong; Rob Scharpf; Daniel J. Zabransky; Abde M. Abukhdeir; Joseph P. Garay; Grace M. Wang; Julia A. Beaver; Rory L. Cochran; Brian G. Blair; D. Marc Rosen; Bracha Erlanger; Pedram Argani; Paula J. Hurley; Josh Lauring; Ben Ho Park

Significance Despite the widespread use and success of tamoxifen for treating ER-positive breast cancers, overcoming resistance to this drug remains an unmet need in clinical breast oncology. The results presented in this study demonstrate that overexpression of a novel gene, MACROD2, can mediate tamoxifen resistance and estrogen independent growth in human breast cancers, and that amplification of MACROD2 in primary breast tumors is associated with worse overall survival. Tamoxifen is effective for treating estrogen receptor-alpha (ER) positive breast cancers. However, few molecular mediators of tamoxifen resistance have been elucidated. Here we describe a previously unidentified gene, MACROD2 that confers tamoxifen resistance and estrogen independent growth. We found MACROD2 is amplified and overexpressed in metastatic tamoxifen-resistant tumors. Transgene overexpression of MACROD2 in breast cancer cell lines results in tamoxifen resistance, whereas RNAi-mediated gene knock down reverses this phenotype. MACROD2 overexpression also leads to estrogen independent growth in xenograft assays. Mechanistically, MACROD2 increases p300 binding to estrogen response elements in a subset of ER regulated genes. Primary breast cancers and matched metastases demonstrate MACROD2 expression can change with disease evolution, and increased expression and amplification of MACROD2 in primary tumors is associated with worse overall survival. These studies establish MACROD2 as a key mediator of estrogen independent growth and tamoxifen resistance, as well as a potential novel target for diagnostics and therapy.


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.


Oncotarget | 2016

Ki-67 is required for maintenance of cancer stem cells but not cell proliferation

Justin Cidado; Hong Yuen Wong; D. Marc Rosen; Ashley Cimino-Mathews; Joseph P. Garay; Abigail G. Fessler; Zeshaan Rasheed; Jessica Hicks; Rory L. Cochran; Sarah Croessmann; Daniel J. Zabransky; Morassa Mohseni; Julia A. Beaver; David Chu; Karen Cravero; Eric S. Christenson; Arielle Medford; Austin Mattox; Angelo M. De Marzo; Pedram Argani; Ajay Chawla; Paula J. Hurley; Josh Lauring; Ben Ho Park

Ki-67 expression is correlated with cell proliferation and is a prognostic marker for various cancers; however, its function is unknown. Here we demonstrate that genetic disruption of Ki-67 in human epithelial breast and colon cancer cells depletes the cancer stem cell niche. Ki-67 null cells had a proliferative disadvantage compared to wildtype controls in colony formation assays and displayed increased sensitivity to various chemotherapies. Ki-67 null cancer cells showed decreased and delayed tumor formation in xenograft assays, which was associated with a reduction in cancer stem cell markers. Immunohistochemical analyses of human breast cancers revealed that Ki-67 expression is maintained at equivalent or greater levels in metastatic sites of disease compared to matched primary tumors, suggesting that maintenance of Ki-67 expression is associated with metastatic/clonogenic potential. These results elucidate Ki-67s role in maintaining the cancer stem cell niche, which has potential diagnostic and therapeutic implications for human malignancies.


Clinical Cancer Research | 2017

HER2 Reactivation through Acquisition of the HER2 L755S Mutation as a Mechanism of Acquired Resistance to HER2-targeted Therapy in HER2+ Breast Cancer

Xiaowei Xu; Carmine De Angelis; Kathleen A. Burke; Agostina Nardone; Huizhong Hu; Lanfang Qin; Jamunarani Veeraraghavan; Vidyalakshmi Sethunath; Laura M. Heiser; Nicholas Wang; Charlotte K.Y. Ng; Edward S. Chen; Alexander Renwick; Tao Wang; Sarmistha Nanda; Martin Shea; Tamika Mitchell; Mahitha Rajendran; Ian Waters; Daniel J. Zabransky; Kenneth L. Scott; Carolina Gutierrez; Chandandeep Nagi; Felipe C. Geyer; Gary C. Chamness; Ben Ho Park; Chad A. Shaw; Susan G. Hilsenbeck; Mothaffar F. Rimawi; Joe W. Gray

Purpose: Resistance to anti-HER2 therapies in HER2+ breast cancer can occur through activation of alternative survival pathways or reactivation of the HER signaling network. Here we employed BT474 parental and treatment-resistant cell line models to investigate a mechanism by which HER2+ breast cancer can reactivate the HER network under potent HER2-targeted therapies. Experimental Design: Resistant derivatives to lapatinib (L), trastuzumab (T), or the combination (LR/TR/LTR) were developed independently from two independent estrogen receptor ER+/HER2+ BT474 cell lines (AZ/ATCC). Two derivatives resistant to the lapatinib-containing regimens (BT474/AZ-LR and BT474/ATCC-LTR lines) that showed HER2 reactivation at the time of resistance were subjected to massive parallel sequencing and compared with parental lines. Ectopic expression and mutant-specific siRNA interference were applied to analyze the mutation functionally. In vitro and in vivo experiments were performed to test alternative therapies for mutant HER2 inhibition. Results: Genomic analyses revealed that the HER2L755S mutation was the only common somatic mutation gained in the BT474/AZ-LR and BT474/ATCC-LTR lines. Ectopic expression of HER2L755S induced acquired lapatinib resistance in the BT474/AZ, SK-BR-3, and AU565 parental cell lines. HER2L755S-specific siRNA knockdown reversed the resistance in BT474/AZ-LR and BT474/ATCC-LTR lines. The HER1/2–irreversible inhibitors afatinib and neratinib substantially inhibited both resistant cell growth and the HER2 and downstream AKT/MAPK signaling driven by HER2L755S in vitro and in vivo. Conclusions: HER2 reactivation through acquisition of the HER2L755S mutation was identified as a mechanism of acquired resistance to lapatinib-containing HER2-targeted therapy in preclinical HER2-amplified breast cancer models, which can be overcome by irreversible HER1/2 inhibitors. Clin Cancer Res; 23(17); 5123–34. ©2017 AACR.


The Prostate | 2012

Lenalidomide Modulates IL-8 and Anti-Prostate Antibody Levels in Men with Biochemically Recurrent Prostate Cancer

Daniel J. Zabransky; Heath A. Smith; Christopher J. Thoburn; Marianna Zahurak; Daniel Keizman; Michael A. Carducci; Mario A. Eisenberger; Douglas G. McNeel; Charles G. Drake; Emmanuel S. Antonarakis

We retrospectively explored changes in immunological parameters in men with biochemically recurrent prostate cancer treated with either 5 or 25 mg of lenalidomide in a randomized phase 2 trial, and determined whether those changes correlated with disease progression.


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.

Collaboration


Dive into the Daniel J. Zabransky's collaboration.

Top Co-Authors

Avatar

Ben Ho Park

Johns Hopkins University

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

David Chu

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Karen Cravero

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
Top Co-Authors

Avatar

Hong Yuen Wong

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bracha Erlanger

Johns Hopkins University School of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge