Network


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

Hotspot


Dive into the research topics where Virginia Chen is active.

Publication


Featured researches published by Virginia Chen.


Transplantation | 2010

Whole blood biomarkers of acute cardiac allograft rejection: double-crossing the biopsy.

Zsuzsanna Hollander; David Lin; Virginia Chen; Raymond T. Ng; J. Wilson-McManus; Andrew Ignaszewski; Gabriela V. Cohen Freue; Rob Balshaw; Alice Mui; R. McMaster; Paul Keown; Bruce M. McManus

Background. Acute rejection is still a significant barrier to long-term survival of the allograft. Current acute rejection diagnostic methods are not specific enough or are invasive. There have been a number of studies that have explored the blood or the biopsy to discover genomic biomarkers of acute rejection; however, none of the studies to date have used both. Methods. We analyzed endomyocardial biopsy tissue and whole blood-derived messenger RNA from 11 acute rejection and 20 nonrejection patients using Affymetrix Human Genome U133 Plus 2.0 chips. We used a novel approach and gained insight into the biology of rejection based on gene expression in the biopsy, and applied this knowledge to the blood analysis to identify novel blood biomarkers. Results. We identified probesets that are differentially expressed between acute rejection and nonrejection patients in the biopsy and blood, and developed three biomarker panels: (1) based on biopsy-only (area under the curve=0.85), (2) based on biopsy-targeted whole blood (area under the curve=0.83), and (3) based on whole blood-only (area under the curve=0.60) analyses. Conclusions. Most of the probesets replicated between biopsy and blood are regulated in opposite direction between the two sources of information. We also observed that the biopsy-targeted blood biomarker discovery approach can improve performance of the biomarker panel. The biomarker panel developed using this targeted approach is able to diagnose acute cardiac allograft rejection almost as well as the biopsy-only based biomarker panel.


Journal of Heart and Lung Transplantation | 2013

Predicting acute cardiac rejection from donor heart and pre-transplant recipient blood gene expression

Zsuzsanna Hollander; Virginia Chen; Keerat Sidhu; David Lin; Raymond T. Ng; Robert Balshaw; Gabriela Cohen-Freue; Andrew Ignaszewski; C. Imai; A. Kaan; Scott J. Tebbutt; J. Wilson-McManus; R. McMaster; Paul Keown; Bruce M. McManus

BACKGROUND Acute rejection in cardiac transplant patients remains a contributory factor to limited survival of implanted hearts. Currently, there are no biomarkers in clinical use that can predict, at the time of transplantation, the likelihood of post-transplant acute cellular rejection. Such a development would be of great value in personalizing immunosuppressive treatment. METHODS Recipient age, donor age, cold ischemic time, warm ischemic time, panel-reactive antibody, gender mismatch, blood type mismatch and human leukocyte antigens (HLA-A, -B and -DR) mismatch between recipients and donors were tested in 53 heart transplant patients for their power to predict post-transplant acute cellular rejection. Donor transplant biopsy and recipient pre-transplant blood were also examined for the presence of genomic biomarkers in 7 rejection and 11 non-rejection patients, using non-targeted data mining techniques. RESULTS The biomarker based on the 8 clinical variables had an area under the receiver operating characteristic curve (AUC) of 0.53. The pre-transplant recipient blood gene-based panel did not yield better performance, but the donor heart tissue gene-based panel had an AUC = 0.78. A combination of 25 probe sets from the transplant donor biopsy and 18 probe sets from the pre-transplant recipient whole blood had an AUC = 0.90. Biologic pathways implicated include VEGF- and EGFR-signaling, and MAPK. CONCLUSIONS Based on this study, the best predictive biomarker panel contains genes from recipient whole blood and donor myocardial tissue. This panel provides clinically relevant prediction power and, if validated, may personalize immunosuppressive treatment and rejection monitoring.


BMC Bioinformatics | 2012

A computational pipeline for the development of multi-marker bio-signature panels and ensemble classifiers

Oliver P. Günther; Virginia Chen; Gabriela V. Cohen Freue; Robert Balshaw; Scott J. Tebbutt; Zsuzsanna Hollander; Mandeep Takhar; W. Robert McMaster; Bruce M. McManus; Paul Keown; Raymond T. Ng

BackgroundBiomarker panels derived separately from genomic and proteomic data and with a variety of computational methods have demonstrated promising classification performance in various diseases. An open question is how to create effective proteo-genomic panels. The framework of ensemble classifiers has been applied successfully in various analytical domains to combine classifiers so that the performance of the ensemble exceeds the performance of individual classifiers. Using blood-based diagnosis of acute renal allograft rejection as a case study, we address the following question in this paper: Can acute rejection classification performance be improved by combining individual genomic and proteomic classifiers in an ensemble?ResultsThe first part of the paper presents a computational biomarker development pipeline for genomic and proteomic data. The pipeline begins with data acquisition (e.g., from bio-samples to microarray data), quality control, statistical analysis and mining of the data, and finally various forms of validation. The pipeline ensures that the various classifiers to be combined later in an ensemble are diverse and adequate for clinical use. Five mRNA genomic and five proteomic classifiers were developed independently using single time-point blood samples from 11 acute-rejection and 22 non-rejection renal transplant patients. The second part of the paper examines five ensembles ranging in size from two to 10 individual classifiers. Performance of ensembles is characterized by area under the curve (AUC), sensitivity, and specificity, as derived from the probability of acute rejection for individual classifiers in the ensemble in combination with one of two aggregation methods: (1) Average Probability or (2) Vote Threshold. One ensemble demonstrated superior performance and was able to improve sensitivity and AUC beyond the best values observed for any of the individual classifiers in the ensemble, while staying within the range of observed specificity. The Vote Threshold aggregation method achieved improved sensitivity for all 5 ensembles, but typically at the cost of decreased specificity.ConclusionProteo-genomic biomarker ensemble classifiers show promise in the diagnosis of acute renal allograft rejection and can improve classification performance beyond that of individual genomic or proteomic classifiers alone. Validation of our results in an international multicenter study is currently underway.


European Journal of Heart Failure | 2014

Proteomic biomarkers of recovered heart function

Zsuzsanna Hollander; Marie Lazárová; Karen K.Y. Lam; Andrew Ignaszewski; Gavin Y. Oudit; Jason R. B. Dyck; George E. Schreiner; Julie Pauwels; Virginia Chen; Gabriela V. Cohen Freue; Raymond T. Ng; J. Wilson-McManus; Robert Balshaw; Scott J. Tebbutt; R. McMaster; Paul Keown; Bruce M. McManus

Chronic heart failure is a costly epidemic that affects up to 2% of people in developed countries. The purpose of this study was to discover novel blood proteomic biomarker signatures of recovered heart function that could lead to more effective heart failure patient management by both primary care and specialty physicians.


PLOS ONE | 2017

Clinical utility of C-reactive protein to predict treatment response during cystic fibrosis pulmonary exacerbations

Ashutosh Sharma; Gordon Kirkpatrick; Virginia Chen; Kate Skolnik; Zsuzsanna Hollander; Pearce G. Wilcox; Bradley S. Quon

Rationale C-reactive protein (CRP) is a systemic marker of inflammation that correlates with disease status in cystic fibrosis (CF). The clinical utility of CRP measurement to guide pulmonary exacerbation (PEx) treatment decisions remains uncertain. Objectives To determine whether monitoring CRP during PEx treatment can be used to predict treatment response. We hypothesized that early changes in CRP can be used to predict treatment response. Methods We reviewed all PEx events requiring hospitalization for intravenous (IV) antibiotics over 2 years at our institution. 83 PEx events met our eligibility criteria. CRP levels from admission to day 5 were evaluated to predict treatment non-response, using a modified version of a prior published composite definition. CRP was also evaluated to predict time until next exacerbation (TUNE). Measurements and main results 53% of 83 PEx events were classified as treatment non-response. Paradoxically, 24% of PEx events were characterized by a ≥ 50% increase in CRP levels within the first five days of treatment. Absolute change in CRP from admission to day 5 was not associated with treatment non-response (p = 0.58). Adjusted for FEV1% predicted, admission log10 CRP was associated with treatment non-response (OR: 2.39; 95% CI: 1.14 to 5.91; p = 0.03) and shorter TUNE (HR: 1.60; 95% CI: 1.13 to 2.27; p = 0.008). The area under the receiver operating characteristics (ROC) curve of admission CRP to predict treatment non-response was 0.72 (95% CI 0.61–0.83; p<0.001). 23% of PEx events were characterized by an admission CRP of > 75 mg/L with a specificity of 90% for treatment non-response. Conclusions Admission CRP predicts treatment non-response and time until next exacerbation. A very elevated admission CRP (>75mg/L) is highly specific for treatment non-response and might be used to target high-risk patients for future interventional studies aimed at improving exacerbation outcomes.


PLOS ONE | 2017

C-reactive protein and N-terminal prohormone brain natriuretic peptide as biomarkers in acute exacerbations of COPD leading to hospitalizations

Yu‐Wei Roy Chen; Virginia Chen; Zsuzsanna Hollander; Jonathon Leipsic; Cameron J. Hague; Mari L. DeMarco; J. Mark FitzGerald; Bruce McManus; Raymond T. Ng; Don D. Sin

There are currently no accepted and validated blood tests available for diagnosing acute exacerbations of chronic obstructive pulmonary disease (AECOPD). In this study, we sought to determine the discriminatory power of blood C-reactive protein (CRP) and N-terminal prohormone brain natriuretic peptide (NT-proBNP) in the diagnosis of AECOPD requiring hospitalizations. The study cohort consisted of 468 patients recruited in the COPD Rapid Transition Program who were hospitalized with a primary diagnosis of AECOPD, and 110 stable COPD patients who served as controls. Logistic regression was used to build a classification model to separate AECOPD from convalescent or stable COPD patients. Performance was assessed using an independent validation set of patients who were not included in the discovery set. Serum CRP and whole blood NT-proBNP concentrations were highest at the time of hospitalization and progressively decreased over time. Of the 3 classification models, the one with both CRP and NT-proBNP had the highest AUC in discriminating AECOPD (cross-validated AUC of 0.80). These data were replicated in a validation cohort with an AUC of 0.88. A combination of CRP and NT-proBNP can reasonably discriminate AECOPD requiring hospitalization versus clinical stability and can be used to rapidly diagnose patients requiring hospitalization for AECOPD.


BMC Genomics | 2017

Enumerateblood – an R package to estimate the cellular composition of whole blood from Affymetrix Gene ST gene expression profiles

Casey P. Shannon; Robert Balshaw; Virginia Chen; Zsuzsanna Hollander; Mustafa Toma; Bruce McManus; J. Mark FitzGerald; Don D. Sin; Raymond T. Ng; Scott J. Tebbutt

BackgroundMeasuring genome-wide changes in transcript abundance in circulating peripheral whole blood is a useful way to study disease pathobiology and may help elucidate the molecular mechanisms of disease, or discovery of useful disease biomarkers. The sensitivity and interpretability of analyses carried out in this complex tissue, however, are significantly affected by its dynamic cellular heterogeneity. It is therefore desirable to quantify this heterogeneity, either to account for it or to better model interactions that may be present between the abundance of certain transcripts, specific cell types and the indication under study. Accurate enumeration of the many component cell types that make up peripheral whole blood can further complicate the sample collection process, however, and result in additional costs. Many approaches have been developed to infer the composition of a sample from high-dimensional transcriptomic and, more recently, epigenetic data. These approaches rely on the availability of isolated expression profiles for the cell types to be enumerated. These profiles are platform-specific, suitable datasets are rare, and generating them is expensive. No such dataset exists on the Affymetrix Gene ST platform.ResultsWe present ‘Enumerateblood’, a freely-available and open source R package that exposes a multi-response Gaussian model capable of accurately predicting the composition of peripheral whole blood samples from Affymetrix Gene ST expression profiles, outperforming other current methods when applied to Gene ST data.Conclusions‘Enumerateblood’ significantly improves our ability to study disease pathobiology from whole blood gene expression assayed on the popular Affymetrix Gene ST platform by allowing a more complete study of the various components of this complex tissue without the need for additional data collection. Future use of the model may allow for novel insights to be generated from the ~400 Affymetrix Gene ST blood gene expression datasets currently available on the Gene Expression Omnibus (GEO) website.


PLOS ONE | 2016

COPD Exacerbation Biomarkers Validated Using Multiple Reaction Monitoring Mass Spectrometry.

Janice M. Leung; Virginia Chen; Zsuzsanna Hollander; Darlene Dai; Scott J. Tebbutt; Shawn D. Aaron; Kathy L. Vandemheen; Stephen I. Rennard; J. Mark FitzGerald; Prescott G. Woodruff; Stephen C. Lazarus; John E. Connett; Harvey O. Coxson; Christoph H. Borchers; Bruce McManus; Raymond T. Ng; Don D. Sin

Background Acute exacerbations of chronic obstructive pulmonary disease (AECOPD) result in considerable morbidity and mortality. However, there are no objective biomarkers to diagnose AECOPD. Methods We used multiple reaction monitoring mass spectrometry to quantify 129 distinct proteins in plasma samples from patients with COPD. This analytical approach was first performed in a biomarker cohort of patients hospitalized with AECOPD (Cohort A, n = 72). Proteins differentially expressed between AECOPD and convalescent states were chosen using a false discovery rate <0.01 and fold change >1.2. Protein selection and classifier building were performed using an elastic net logistic regression model. The performance of the biomarker panel was then tested in two independent AECOPD cohorts (Cohort B, n = 37, and Cohort C, n = 109) using leave-pair-out cross-validation methods. Results Five proteins were identified distinguishing AECOPD and convalescent states in Cohort A. Biomarker scores derived from this model were significantly higher during AECOPD than in the convalescent state in the discovery cohort (p<0.001). The receiver operating characteristic cross-validation area under the curve (CV-AUC) statistic was 0.73 in Cohort A, while in the replication cohorts the CV-AUC was 0.77 for Cohort B and 0.79 for Cohort C. Conclusions A panel of five biomarkers shows promise in distinguishing AECOPD from convalescence and may provide the basis for a clinical blood test to diagnose AECOPD. Further validation in larger cohorts is necessary for future clinical translation.


Esc Heart Failure | 2016

Circulating biomarker responses to medical management vs. mechanical circulatory support in severe inotrope‐dependent acute heart failure

Anna Meredith; Darlene L.Y. Dai; Virginia Chen; Zsuzsanna Hollander; Raymond T. Ng; A. Kaan; Scott J. Tebbutt; Krishnan Ramanathan; Anson Cheung; Bruce M. McManus

Severe inotrope‐dependent acute heart failure (AHF) is associated with poor clinical outcomes. There are currently no well‐defined blood biomarkers of response to treatment that can guide management or identify recovery in this patient population. In the present study, we characterized the levels of novel and emerging circulating biomarkers of heart failure in patients with AHF over the first 30 days of medical management or mechanical circulatory support (MCS). We hypothesized a shared a plasma proteomic treatment response would be identifiable in both patient groups, representing reversal of the AHF phenotype.


PLOS ONE | 2015

The Effect of Statins on Blood Gene Expression in COPD

Ma’en Obeidat; Nick Fishbane; Yunlong Nie; Virginia Chen; Zsuzsanna Hollander; Scott J. Tebbutt; Yohan Bossé; Raymond T. Ng; Bruce M. McManus; Stephen I. Rennard; Peter D. Paré; Don D. Sin

Background COPD is currently the fourth leading cause of death worldwide. Statins are lipid lowering agents with documented cardiovascular benefits. Observational studies have shown that statins may have a beneficial role in COPD. The impact of statins on blood gene expression from COPD patients is largely unknown. Objective Identify blood gene signature associated with statin use in COPD patients, and the pathways underpinning this signature that could explain any potential benefits in COPD. Methods Whole blood gene expression was measured on 168 statin users and 451 non-users from the ECLIPSE study using the Affymetrix Human Gene 1.1 ST microarray chips. Factor Analysis for Robust Microarray Summarization (FARMS) was used to process the expression data. Differential gene expression analysis was undertaken using the Linear Models for Microarray data (Limma) package adjusting for propensity score and surrogate variables. Similarity of the expression signal with published gene expression profiles was performed in ProfileChaser. Results 25 genes were differentially expressed between statin users and non-users at an FDR of 10%, including LDLR, CXCR2, SC4MOL, FAM108A1, IFI35, FRYL, ABCG1, MYLIP, and DHCR24. The 25 genes were significantly enriched in cholesterol homeostasis and metabolism pathways. The resulting gene signature showed correlation with Huntington’s disease, Parkinson’s disease and acute myeloid leukemia gene signatures. Conclusion The blood gene signature of statins’ use in COPD patients was enriched in cholesterol homeostasis pathways. Further studies are needed to delineate the role of these pathways in lung biology.

Collaboration


Dive into the Virginia Chen's collaboration.

Top Co-Authors

Avatar

Raymond T. Ng

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Zsuzsanna Hollander

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Bruce M. McManus

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Scott J. Tebbutt

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Robert Balshaw

BC Centre for Disease Control

View shared research outputs
Top Co-Authors

Avatar

Paul Keown

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Andrew Ignaszewski

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

R. McMaster

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

J. Wilson-McManus

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Casey P. Shannon

University of British Columbia

View shared research outputs
Researchain Logo
Decentralizing Knowledge