Network


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

Hotspot


Dive into the research topics where Ben Ho Park is active.

Publication


Featured researches published by Ben Ho Park.


Science | 2007

The genomic landscapes of human breast and colorectal cancers.

Laura D. Wood; D. Williams Parsons; Siân Jones; Jimmy Lin; Tobias Sjöblom; Rebecca J. Leary; Dong Shen; Simina M. Boca; Thomas D. Barber; Janine Ptak; Natalie Silliman; Steve Szabo; Zoltan Dezso; Vadim Ustyanksky; Tatiana Nikolskaya; Yuri Nikolsky; Rachel Karchin; Paul Wilson; Joshua S. Kaminker; Zemin Zhang; Randal Croshaw; Joseph Willis; Dawn Dawson; Michail Shipitsin; James K V Willson; Saraswati Sukumar; Kornelia Polyak; Ben Ho Park; Charit L. Pethiyagoda; P.V. Krishna Pant

Human cancer is caused by the accumulation of mutations in oncogenes and tumor suppressor genes. To catalog the genetic changes that occur during tumorigenesis, we isolated DNA from 11 breast and 11 colorectal tumors and determined the sequences of the genes in the Reference Sequence database in these samples. Based on analysis of exons representing 20,857 transcripts from 18,191 genes, we conclude that the genomic landscapes of breast and colorectal cancers are composed of a handful of commonly mutated gene “mountains” and a much larger number of gene “hills” that are mutated at low frequency. We describe statistical and bioinformatic tools that may help identify mutations with a role in tumorigenesis. These results have implications for understanding the nature and heterogeneity of human cancers and for using personal genomics for tumor diagnosis and therapy.


Nature | 2002

DNMT1 and DNMT3b cooperate to silence genes in human cancer cells

Ina Rhee; Kurtis E. Bachman; Ben Ho Park; Kam Wing Jair; Ray Whay Chiu Yen; Kornel E. Schuebel; Hengmi Cui; Andrew P. Feinberg; Christoph Lengauer; Kenneth W. Kinzler; Stephen B. Baylin; Bert Vogelstein

Inactivation of tumour suppressor genes is central to the development of all common forms of human cancer. This inactivation often results from epigenetic silencing associated with hypermethylation rather than intragenic mutations. In human cells, the mechanisms underlying locus-specific or global methylation patterns remain unclear. The prototypic DNA methyltransferase, Dnmt1, accounts for most methylation in mouse cells, but human cancer cells lacking DNMT1 retain significant genomic methylation and associated gene silencing. We disrupted the human DNMT3b gene in a colorectal cancer cell line. This deletion reduced global DNA methylation by less than 3%. Surprisingly, however, genetic disruption of both DNMT1 and DNMT3b nearly eliminated methyltransferase activity, and reduced genomic DNA methylation by greater than 95%. These marked changes resulted in demethylation of repeated sequences, loss of insulin-like growth factor II (IGF2) imprinting, abrogation of silencing of the tumour suppressor gene p16INK4a, and growth suppression. Here we demonstrate that two enzymes cooperatively maintain DNA methylation and gene silencing in human cancer cells, and provide compelling evidence that such methylation is essential for optimal neoplastic proliferation.


Cancer Biology & Therapy | 2004

The PIK3CA gene is mutated with high frequency in human breast cancers

Kurtis E. Bachman; Pedram Argani; Yardena Samuels; Natalie Silliman; Janine Ptak; Steve Szabo; Hiroyuki Konishi; Bedri Karakas; Brian G. Blair; Clarence Lin; Brock A. Peters; Victor E. Velculescu; Ben Ho Park

The phosphatidylinositol 3-kinases (PI3Ks) are known regulators of cellular growth and proliferation. It has recently been reported that somatic mutations within the PI3K subunit p110? (PIK3CA) are present in human colorectal and other cancers. Here we show that thirteen of fifty-three breast cancers (25%) contain somatic mutations in PIK3CA, with the majority of mutations located in the kinase domain. These results demonstrate that PIK3CA is the most mutated oncogene in breast cancer and support a role for PIK3CA in epithelial carcinogenesis.


Cancer Cell | 2003

Histone modifications and silencing prior to DNA methylation of a tumor suppressor gene.

Kurtis E. Bachman; Ben Ho Park; Ina Rhee; James G. Herman; Stephen B. Baylin; Kenneth W. Kinzler; Bert Vogelstein

We attempted to answer two central questions about epigenetic silencing of the tumor suppressor gene p16(INK4a) in this study: (1) whether the maintenance of associated histone modifications is dependent on DNA methylation and (2) whether such histone modifications can occur prior to DNA methylation. By coupling chromatin immunoprecipitation with gene targeting and the analysis of specific alleles, we found that elimination of DNA methylation from a p16(INK4a) allele resulted in profound changes in surrounding histones. After continued passage of such cells, methylation of histone H3 lysine-9 occurred in conjunction with re-silencing in the absence of DNA methylation. These results have important implications for understanding the biochemical events underlying the silencing of tumor suppressor genes and the resultant growth suppression.


British Journal of Cancer | 2006

Mutation of the PIK3CA oncogene in human cancers

Bedri Karakas; Kurtis E. Bachman; Ben Ho Park

It is now well established that cancer is a genetic disease and that somatic mutations of oncogenes and tumour suppressor genes are the initiators of the carcinogenic process. The phosphatidylinositol 3-kinase signalling pathway has previously been implicated in tumorigenesis, and evidence over the past year suggests a pivotal role for the phosphatidylinositol 3-kinase catalytic subunit, PIK3CA, in human cancers. In this review, we analyse recent reports describing PIK3CA mutations in a variety of human malignancies, and discuss their possible implications for diagnosis and therapy.


Proceedings of the National Academy of Sciences of the United States of America | 2001

Genetic disruption of PPARδ decreases the tumorigenicity of human colon cancer cells

Ben Ho Park; Bert Vogelstein; Kenneth W. Kinzler

Peroxisome proliferator-activated receptors (PPARs) are nuclear hormone receptors that have been implicated in a variety of biologic processes. The PPARδ isotype was recently proposed as a downstream target of the adenomatous polyposis coli (APC)/β-catenin pathway in colorectal carcinogenesis. To evaluate its role in tumorigenesis, a PPARδ null cell line was created by targeted homologous recombination. When inoculated as xenografts in nude mice, PPARδ −/− cells exhibited a decreased ability to form tumors compared with PPARδ +/− and wild-type controls. These data suggest that suppression of PPARδ expression contributes to the growth-inhibitory effects of the APC tumor suppressor.


Proceedings of the National Academy of Sciences of the United States of America | 2008

Integrated analysis of homozygous deletions, focal amplifications, and sequence alterations in breast and colorectal cancers

Rebecca J. Leary; Jimmy Lin; Jordan M. Cummins; Simina M. Boca; Laura D. Wood; D. Williams Parsons; Siân Jones; Tobias Sjöblom; Ben Ho Park; Ramon Parsons; Joseph Willis; Dawn Dawson; James K V Willson; Tatiana Nikolskaya; Yuri Nikolsky; Levy Kopelovich; Nick Papadopoulos; Len A. Pennacchio; Tian Li Wang; Sanford D. Markowitz; Giovanni Parmigiani; Kenneth W. Kinzler; Bert Vogelstein; Victor E. Velculescu

We have performed a genome-wide analysis of copy number changes in breast and colorectal tumors using approaches that can reliably detect homozygous deletions and amplifications. We found that the number of genes altered by major copy number changes, deletion of all copies or amplification to at least 12 copies per cell, averaged 17 per tumor. We have integrated these data with previous mutation analyses of the Reference Sequence genes in these same tumor types and have identified genes and cellular pathways affected by both copy number changes and point alterations. Pathways enriched for genetic alterations included those controlling cell adhesion, intracellular signaling, DNA topological change, and cell cycle control. These analyses provide an integrated view of copy number and sequencing alterations on a genome-wide scale and identify genes and pathways that could prove useful for cancer diagnosis and therapy.


Clinical Cancer Research | 2012

Detection of Tumor PIK3CA Status in Metastatic Breast Cancer Using Peripheral Blood

Michaela J. Higgins; Danijela Jelovac; Evan Barnathan; Brian G. Blair; Shannon Slater; Penny Powers; Jane Zorzi; Stacie Jeter; George Oliver; John H. Fetting; Leisha A. Emens; Carol D. Riley; Vered Stearns; Frank Diehl; Phillip Angenendt; Peng Huang; Leslie Cope; Pedram Argani; Kathleen M. Murphy; Kurtis E. Bachman; Joel Greshock; Antonio C. Wolff; Ben Ho Park

Purpose: We sought to evaluate the feasibility of detecting PIK3CA mutations in circulating tumor DNA (ctDNA) from plasma of patients with metastatic breast cancer using a novel technique called BEAMing. Experimental Design: In a retrospective analysis, 49 tumor and temporally matched plasma samples from patients with breast cancer were screened for PIK3CA mutations by BEAMing. We then prospectively screened the ctDNA of 60 patients with metastatic breast cancer for PIK3CA mutations by BEAMing and compared the findings with results obtained by screening corresponding archival tumor tissue DNA using both sequencing and BEAMing. Results: The overall frequency of PIK3CA mutations by BEAMing was similar in both patient cohorts (29% and 28.3%, respectively). In the retrospective cohort, the concordance of PIK3CA mutation status by BEAMing between formalin-fixed, paraffin-embedded (FFPE) samples and ctDNA from temporally matched plasma was 100% (34 of 34). In the prospective cohort, the concordance rate among 51 evaluable cases was 72.5% between BEAMing of ctDNA and sequencing of archival tumor tissue DNA. When the same archival tissue DNA was screened by both sequencing and BEAMing for PIK3CA mutations (n = 41 tissue samples), there was 100% concordance in the obtained results. Conclusions: Analysis of plasma-derived ctDNA for the detection of PIK3CA mutations in patients with metastatic breast cancer is feasible. Our results suggest that PIK3CA mutational status can change upon disease recurrence, emphasizing the importance of reassessing PIK3CA status on contemporary (not archival) biospecimens. These results have implications for the development of predictive biomarkers of response to targeted therapies. Clin Cancer Res; 18(12); 3462–9. ©2012 AACR.


Clinical Cancer Research | 2011

PTEN Protein Loss by Immunostaining: Analytic Validation and Prognostic Indicator for a High Risk Surgical Cohort of Prostate Cancer Patients

Tamara L. Lotan; Bora Gurel; Siobhan Sutcliffe; David Esopi; Wennuan Liu; Jianfeng Xu; Jessica Hicks; Ben Ho Park; Elizabeth B. Humphreys; Alan W. Partin; Misop Han; George J. Netto; William B. Isaacs; Angelo M. De Marzo

Purpose: Analytically validated assays to interrogate biomarker status in clinical samples are crucial for personalized medicine. PTEN is a tumor suppressor commonly inactivated in prostate cancer that has been mechanistically linked to disease aggressiveness. Though deletion of PTEN, as detected by cumbersome FISH spot counting assays, is associated with poor prognosis, few studies have validated immunohistochemistry (IHC) assays to determine whether loss of PTEN protein is associated with unfavorable disease. Experimental Design: PTEN IHC was validated by employing formalin fixed and paraffin-embedded isogenic human cell lines containing or lacking intact PTEN alleles. PTEN IHC was 100% sensitive and 97.8% specific for detecting genomic alterations in 58 additional cell lines. PTEN protein loss was then assessed on 376 prostate tumor samples, and PTEN FISH or high resolution single nucleotide polymorphism microarray analysis was done on a subset of these cases. Results: PTEN protein loss, as assessed as a dichotomous IHC variable, was highly reproducible, correlated strongly with adverse pathologic features (e.g., Gleason score and pathologic stage), detected between 75% and 86% of cases with PTEN genomic loss, and was found at times in the absence of apparent genomic loss. In a cohort of 217 high risk surgically treated patients, PTEN protein loss was associated with decreased time to metastasis. Conclusion: These studies validate a simple method to interrogate PTEN status in clinical specimens and support the utility of this test in future multicenter studies, clinical trials, and ultimately perhaps for routine clinical care. Clin Cancer Res; 17(20); 6563–73. ©2011 AACR.


Molecular Cell | 2011

NSD2 Links Dimethylation of Histone H3 at Lysine 36 to Oncogenic Programming

Alex J. Kuo; Peggie Cheung; Kaifu Chen; Barry M. Zee; Mitomu Kioi; Josh Lauring; Yuanxin Xi; Ben Ho Park; Xiaobing Shi; Benjamin A. Garcia; Wei Li; Or Gozani

The histone lysine methyltransferase NSD2 (MMSET/WHSC1) is implicated in diverse diseases and commonly overexpressed in multiple myeloma due to a recurrent t(4;14) chromosomal translocation. However, the precise catalytic activity of NSD2 is obscure, preventing progress in understanding how this enzyme influences chromatin biology and myeloma pathogenesis. Here, we show that dimethylation of histone H3 at lysine 36 (H3K36me2) is the principal chromatin-regulatory activity of NSD2. Catalysis of H3K36me2 by NSD2 is sufficient for gene activation. In t(4;14)-positive myeloma cells, the normal genome-wide and gene-specific distribution of H3K36me2 is obliterated, creating a chromatin landscape that selects for a transcription profile favorable for myelomagenesis. Catalytically active NSD2 confers xenograft tumor formation upon t(4;14)-negative cells and promotes oncogenic transformation of primary cells in an H3K36me2-dependent manner. Together, our findings establish H3K36me2 as the primary product generated by NSD2 and demonstrate that genomic disorganization of this canonical chromatin mark by NSD2 initiates oncogenic programming.

Collaboration


Dive into the Ben Ho Park's collaboration.

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

Pedram Argani

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar
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

Rory L. Cochran

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

Sarah Croessmann

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Abde M. Abukhdeir

Rush University Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge