Network


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

Hotspot


Dive into the research topics where Fong Chun Chan is active.

Publication


Featured researches published by Fong Chun Chan.


Blood | 2013

Mutational and structural analysis of diffuse large B-cell lymphoma using whole-genome sequencing

Ryan D. Morin; Karen Mungall; Erin Pleasance; Andrew J. Mungall; Rodrigo Goya; Ryan D. Huff; David W. Scott; Jiarui Ding; Andrew Roth; Readman Chiu; Richard Corbett; Fong Chun Chan; Maria Mendez-Lago; Diane L. Trinh; Madison Bolger-Munro; Greg Taylor; Alireza Hadj Khodabakhshi; Susana Ben-Neriah; Julia R. Pon; Barbara Meissner; Bruce Woolcock; Noushin Farnoud; Sanja Rogic; Emilia L. Lim; Nathalie A. Johnson; Sohrab P. Shah; Steven J.M. Jones; Christian Steidl; Robert A. Holt; Inanc Birol

Diffuse large B-cell lymphoma (DLBCL) is a genetically heterogeneous cancer composed of at least 2 molecular subtypes that differ in gene expression and distribution of mutations. Recently, application of genome/exome sequencing and RNA-seq to DLBCL has revealed numerous genes that are recurrent targets of somatic point mutation in this disease. Here we provide a whole-genome-sequencing-based perspective of DLBCL mutational complexity by characterizing 40 de novo DLBCL cases and 13 DLBCL cell lines and combining these data with DNA copy number analysis and RNA-seq from an extended cohort of 96 cases. Our analysis identified widespread genomic rearrangements including evidence for chromothripsis as well as the presence of known and novel fusion transcripts. We uncovered new gene targets of recurrent somatic point mutations and genes that are targeted by focal somatic deletions in this disease. We highlight the recurrence of germinal center B-cell-restricted mutations affecting genes that encode the S1P receptor and 2 small GTPases (GNA13 and GNAI2) that together converge on regulation of B-cell homing. We further analyzed our data to approximate the relative temporal order in which some recurrent mutations were acquired and demonstrate that ongoing acquisition of mutations and intratumoral clonal heterogeneity are common features of DLBCL. This study further improves our understanding of the processes and pathways involved in lymphomagenesis, and some of the pathways mutated here may indicate new avenues for therapeutic intervention.


Nature Medicine | 2014

Pharmacological and genomic profiling identifies NF-κB–targeted treatment strategies for mantle cell lymphoma

Rami Rahal; Mareike Frick; Rodrigo Romero; Joshua Korn; Robert Kridel; Fong Chun Chan; Barbara Meissner; Hyo-eun C. Bhang; Dave Ruddy; Audrey Kauffmann; Ali Farsidjani; Adnan Derti; Daniel Rakiec; Tara L. Naylor; Estelle Pfister; Steve Kovats; Sunkyu Kim; Kerstin Dietze; Bernd Dörken; Christian Steidl; Alexandar Tzankov; Michael Hummel; John E. Monahan; Michael Morrissey; Christine Fritsch; William R. Sellers; Vesselina G. Cooke; Randy D. Gascoyne; Georg Lenz; Frank Stegmeier

Mantle cell lymphoma (MCL) is an aggressive malignancy that is characterized by poor prognosis. Large-scale pharmacological profiling across more than 100 hematological cell line models identified a subset of MCL cell lines that are highly sensitive to the B cell receptor (BCR) signaling inhibitors ibrutinib and sotrastaurin. Sensitive MCL models exhibited chronic activation of the BCR-driven classical nuclear factor-κB (NF-κB) pathway, whereas insensitive cell lines displayed activation of the alternative NF-κB pathway. Transcriptome sequencing revealed genetic lesions in alternative NF-κB pathway signaling components in ibrutinib-insensitive cell lines, and sequencing of 165 samples from patients with MCL identified recurrent mutations in TRAF2 or BIRC3 in 15% of these individuals. Although they are associated with insensitivity to ibrutinib, lesions in the alternative NF-κB pathway conferred dependence on the protein kinase NIK (also called mitogen-activated protein 3 kinase 14 or MAP3K14) both in vitro and in vivo. Thus, NIK is a new therapeutic target for MCL treatment, particularly for lymphomas that are refractory to BCR pathway inhibitors. Our findings reveal a pattern of mutually exclusive activation of the BCR–NF-κB or NIK–NF-κB pathways in MCL and provide critical insights into patient stratification strategies for NF-κB pathway–targeted agents.


Blood | 2014

Genomic rearrangements involving programmed death ligands are recurrent in primary mediastinal large B-cell lymphoma.

David D.W. Twa; Fong Chun Chan; Susana Ben-Neriah; Bruce Woolcock; Anja Mottok; King Tan; Graham W. Slack; Jay Gunawardana; Raymond S. Lim; Andrew McPherson; Robert Kridel; Adele Telenius; David W. Scott; Kerry J. Savage; Sohrab P. Shah; Randy D. Gascoyne; Christian Steidl

The pathogenesis of primary mediastinal large B-cell lymphoma (PMBCL) is incompletely understood. Recently, specific genotypic and phenotypic features have been linked to tumor cell immune escape mechanisms in PMBCL. We studied 571 B-cell lymphomas with a focus on PMBCL. Using fluorescence in situ hybridization here, we report that the programmed death ligand (PDL) locus (9p24.1) is frequently and specifically rearranged in PMBCL (20%) as compared with diffuse large B-cell lymphoma, follicular lymphoma, and Hodgkin lymphoma. Rearrangement was significantly correlated with overexpression of PDL transcripts. Utilizing high-throughput sequencing techniques, we characterized novel translocations and chimeric fusion transcripts involving PDLs at base-pair resolution. Our data suggest that recurrent genomic rearrangement events underlie an immune privilege phenotype in a subset of B-cell lymphomas.


Journal of Clinical Oncology | 2013

Gene Expression–Based Model Using Formalin-Fixed Paraffin-Embedded Biopsies Predicts Overall Survival in Advanced-Stage Classical Hodgkin Lymphoma

David W. Scott; Fong Chun Chan; Fangxin Hong; Sanja Rogic; King Tan; Barbara Meissner; Susana Ben-Neriah; Merrill Boyle; Robert Kridel; Adele Telenius; Bruce Woolcock; Pedro Farinha; Richard I. Fisher; Lisa M. Rimsza; Nancy L. Bartlett; Bruce D. Cheson; Lois E. Shepherd; Ranjana H. Advani; Joseph M. Connors; Brad S. Kahl; Leo I. Gordon; Sandra J. Horning; Christian Steidl; Randy D. Gascoyne

PURPOSE Our aim was to reliably identify patients with advanced-stage classical Hodgkin lymphoma (cHL) at increased risk of death by developing a robust predictor of overall survival (OS) using gene expression measured in routinely available formalin-fixed paraffin-embedded tissue (FFPET). METHODS Expression levels of 259 genes, including those previously reported to be associated with outcome in cHL, were determined by digital expression profiling of pretreatment FFPET biopsies from 290 patients enrolled onto the E2496 Intergroup trial comparing doxorubicin, bleomycin, vinblastine, and dacarbazine (ABVD) and Stanford V regimens in locally extensive and advanced-stage cHL. A model for OS separating patients into low- and high-risk groups was produced using penalized Cox regression. The model was tested in an independent cohort of 78 patients enriched for treatment failure but otherwise similar to patients in a population-based registry of patients treated with ABVD. Weighted analysis methods generated unbiased estimates of predictor performance in the population-based registry. RESULTS A 23-gene outcome predictor was generated. The model identified a population at increased risk of death in the validation cohort. There was a 29% absolute difference in 5-year OS between the high- and low-risk groups (63% v 92%, respectively; log-rank P < .001; hazard ratio, 6.7; 95% CI, 2.6 to 17.4). The predictor was superior to the International Prognostic Score and CD68 immunohistochemistry in multivariate analyses. CONCLUSION A gene expression-based predictor, developed in and applicable to routinely available FFPET biopsies, identifies patients with advanced-stage cHL at increased risk of death when treated with standard-intensity up-front regimens.


Nature Genetics | 2014

Recurrent somatic mutations of PTPN1 in primary mediastinal B cell lymphoma and Hodgkin lymphoma

Jay Gunawardana; Fong Chun Chan; Adele Telenius; Bruce Woolcock; Robert Kridel; King Tan; Susana Ben-Neriah; Anja Mottok; Raymond S. Lim; Merrill Boyle; Sanja Rogic; Lisa M. Rimsza; Chrystelle Guiter; Karen Leroy; Philippe Gaulard; Corinne Haioun; Marco A. Marra; Kerry J. Savage; Joseph M. Connors; Sohrab P. Shah; Randy D. Gascoyne; Christian Steidl

Classical Hodgkin lymphoma and primary mediastinal B cell lymphoma (PMBCL) are related lymphomas sharing pathological, molecular and clinical characteristics. Here we discovered by whole-genome and whole-transcriptome sequencing recurrent somatic coding-sequence mutations in the PTPN1 gene. Mutations were found in 6 of 30 (20%) Hodgkin lymphoma cases, in 6 of 9 (67%) Hodgkin lymphoma–derived cell lines, in 17 of 77 (22%) PMBCL cases and in 1 of 3 (33%) PMBCL-derived cell lines, consisting of nonsense, missense and frameshift mutations. We demonstrate that PTPN1 mutations lead to reduced phosphatase activity and increased phosphorylation of JAK-STAT pathway members. Moreover, silencing of PTPN1 by RNA interference in Hodgkin lymphoma cell line KM-H2 resulted in hyperphosphorylation and overexpression of downstream oncogenic targets. Our data establish PTPN1 mutations as new drivers in lymphomagenesis.


Blood | 2012

Gene expression profiling of microdissected Hodgkin Reed-Sternberg cells correlates with treatment outcome in classical Hodgkin lymphoma.

Christian Steidl; Arjan Diepstra; Tang Lee; Fong Chun Chan; Pedro Farinha; King Tan; Adele Telenius; Lorena Barclay; Sohrab P. Shah; Joseph M. Connors; Anke van den Berg; Randy D. Gascoyne

In classical Hodgkin lymphoma (CHL), 20%-30% of patients experience relapse or progressive disease after initial treatment. The pathogenesis and biology of treatment failure are still poorly understood, in part because the molecular phenotype of the rare malignant Hodgkin Reed-Sternberg (HRS) cells is difficult to study. Here we examined microdissected HRS cells from 29 CHL patients and 5 CHL-derived cell lines by gene expression profiling. We found significant overlap of HL-specific gene expression in primary HRS cells and HL cell lines, but also differences, including surface receptor signaling pathways. Using integrative analysis tools, we identified target genes with expression levels that significantly correlated with genomic copy-number changes in primary HRS cells. Furthermore, we found a macrophage-like signature in HRS cells that significantly correlated with treatment failure. CSF1R is a representative of this signature, and its expression was significantly associated with progression-free and overall survival in an independent set of 132 patients assessed by mRNA in situ hybridization. A combined score of CSF1R in situ hybridization and CD68 immunohistochemistry was an independent predictor for progression-free survival in multivariate analysis. In summary, our data reveal novel insights into the pathobiology of treatment failure and suggest CSF1R as a drug target of at-risk CHL.


PLOS Medicine | 2016

Histological Transformation and Progression in Follicular Lymphoma: A Clonal Evolution Study

Robert Kridel; Fong Chun Chan; Anja Mottok; Merrill Boyle; Pedro Farinha; King Tan; Barbara Meissner; Ali Bashashati; Andrew McPherson; Andrew Roth; Karey Shumansky; Damian Yap; Susana Ben-Neriah; Jamie Rosner; Maia A. Smith; Cydney Nielsen; Eva Giné; Adele Telenius; Daisuke Ennishi; Andrew J. Mungall; Richard A. Moore; Ryan D. Morin; Nathalie A. Johnson; Laurie H. Sehn; Thomas Tousseyn; Ahmet Dogan; Joseph M. Connors; David W. Scott; Christian Steidl; Marco A. Marra

Background Follicular lymphoma (FL) is an indolent, yet incurable B cell malignancy. A subset of patients experience an increased mortality rate driven by two distinct clinical end points: histological transformation and early progression after immunochemotherapy. The nature of tumor clonal dynamics leading to these clinical end points is poorly understood, and previously determined genetic alterations do not explain the majority of transformed cases or accurately predict early progressive disease. We contend that detailed knowledge of the expansion patterns of specific cell populations plus their associated mutations would provide insight into therapeutic strategies and disease biology over the time course of FL clinical histories. Methods and Findings Using a combination of whole genome sequencing, targeted deep sequencing, and digital droplet PCR on matched diagnostic and relapse specimens, we deciphered the constituent clonal populations in 15 transformation cases and 6 progression cases, and measured the change in clonal population abundance over time. We observed widely divergent patterns of clonal dynamics in transformed cases relative to progressed cases. Transformation specimens were generally composed of clones that were rare or absent in diagnostic specimens, consistent with dramatic clonal expansions that came to dominate the transformation specimens. This pattern was independent of time to transformation and treatment modality. By contrast, early progression specimens were composed of clones that were already present in the diagnostic specimens and exhibited only moderate clonal dynamics, even in the presence of immunochemotherapy. Analysis of somatic mutations impacting 94 genes was undertaken in an extension cohort consisting of 395 samples from 277 patients in order to decipher disrupted biology in the two clinical end points. We found 12 genes that were more commonly mutated in transformed samples than in the preceding FL tumors, including TP53, B2M, CCND3, GNA13, S1PR2, and P2RY8. Moreover, ten genes were more commonly mutated in diagnostic specimens of patients with early progression, including TP53, BTG1, MKI67, and XBP1. Conclusions Our results illuminate contrasting modes of evolution shaping the clinical histories of transformation and progression. They have implications for interpretation of evolutionary dynamics in the context of treatment-induced selective pressures, and indicate that transformation and progression will require different clinical management strategies.


Blood | 2015

Cell-of-origin of transformed follicular lymphoma

Robert Kridel; Anja Mottok; Pedro Farinha; Susana Ben-Neriah; Daisuke Ennishi; Yvonne Zheng; Elizabeth Chavez; Hennady P. Shulha; King Tan; Fong Chun Chan; Merrill Boyle; Barbara Meissner; Adele Telenius; Laurie H. Sehn; Marco A. Marra; Sohrab P. Shah; Christian Steidl; Joseph M. Connors; David W. Scott; Randy D. Gascoyne

Follicular lymphoma (FL) is an indolent disease but transforms in 2% to 3% of patients per year into aggressive, large cell lymphoma, a critical event in the course of the disease associated with increased lymphoma-related mortality. Early transformation cannot be accurately predicted at the time of FL diagnosis and the biology of transformed FL (TFL) is poorly understood. Here, we assembled a cohort of 126 diagnostic FL specimens including 40 patients experiencing transformation (<5 years) and 86 patients not experiencing transformation for at least 5 years. In addition, we assembled an overlapping cohort of 155 TFL patients, including 114 cases for which paired samples were available, and assessed temporal changes of routinely available biomarkers, outcome after transformation, as well as molecular subtypes of TFL. We report that the expression of IRF4 is an independent predictor of early transformation (Hazard ratio, 13.3; P < .001). We also show that composite histology at the time of transformation predicts favorable prognosis. Moreover, applying the Lymph2Cx digital gene expression assay for diffuse large B-cell lymphoma (DLBCL) cell-of-origin determination to 110 patients with DLBCL-like TFL, we demonstrate that TFL is of the germinal-center B-cell-like subtype in the majority of cases (80%) but that a significant proportion of cases is of the activated B-cell-like (ABC) subtype (16%). These latter cases are commonly negative for BCL2 translocation and arise preferentially from BCL2 translocation-negative and/or IRF4-expressing FLs. Our study demonstrates the existence of molecular heterogeneity in TFL as well as its relationship to the antecedent FL.


Leukemia | 2014

The mutational landscape of Hodgkin lymphoma cell lines determined by whole-exome sequencing

Yuxuan Liu; F.R. Abdul Razak; M. Terpstra; Fong Chun Chan; Ali Saber; Marcel Nijland; van Gustaaf Imhoff; Lydia Visser; Randy D. Gascoyne; Christian Steidl; Joost Kluiver; Arjan Diepstra; Klaas Kok; van den Anke Berg

The mutational landscape of Hodgkin lymphoma cell lines determined by whole-exome sequencing


Blood | 2016

A Novel Prognostic Model Based on Tumor Microenvironment Biology in Relapse Biopsies Predicts Post-Autologous Stem Cell Transplantation Outcomes in Classical Hodgkin Lymphoma

Fong Chun Chan; Anja Mottok; Alina S. Gerrie; Maryse M. Power; Kerry J. Savage; Marcel Nijland; Arjan Diepstra; Anke van den Berg; Joseph M. Connors; Randy D. Gascoyne; Sohrab P. Shah; David W. Scott; Christian Steidl

Pediatric-type nodal follicular lymphoma (PTNFL) is a variant of follicular lymphoma (FL) characterized by limited-stage presentation and invariably benign behavior despite often high-grade histological appearance. It is important to distinguish PTNFL from typical FL in order to avoid unnecessary treatment; however, this distinction relies solely on clinical and pathological criteria, which may be variably applied. To define the genetic landscape of PTNFL, we performed copy number analysis and exome and/or targeted sequencing of 26 PTNFLs (16 pediatric and 10 adult). The most commonly mutated gene in PTNFL was MAP2K1, encoding MEK1, with a mutation frequency of 43%. All MAP2K1 mutations were activating missense mutations localized to exons 2 and 3, which encode negative regulatory and catalytic domains, respectively. Missense mutations in MAPK1 (2/22) and RRAS (1/22) were identified in cases that lacked MAP2K1 mutations. The second most commonly mutated gene in PTNFL was TNFRSF14, with a mutation frequency of 29%, similar to that seen in limited-stage typical FL (P = .35). PTNFL was otherwise genomically bland and specifically lacked recurrent mutations in epigenetic modifiers (eg, CREBBP, KMT2D). Copy number aberrations affected a mean of only 0.5% of PTNFL genomes, compared with 10% of limited-stage typical FL genomes (P < .02). Importantly, the mutational profiles of PTNFLs in children and adults were highly similar. Together, these findings define PTNFL as a biologically and clinically distinct indolent lymphoma of children and adults characterized by a high prevalence of MAPK pathway mutations and a near absence of mutations in epigenetic modifiers.

Collaboration


Dive into the Fong Chun Chan's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Joseph M. Connors

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sohrab P. Shah

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Robert Kridel

Princess Margaret Cancer Centre

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

David W. Scott

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Marco A. Marra

University of British Columbia

View shared research outputs
Researchain Logo
Decentralizing Knowledge