Network


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

Hotspot


Dive into the research topics where Minghui Lee is active.

Publication


Featured researches published by Minghui Lee.


Gut | 2012

A comprehensive survey of genomic alterations in gastric cancer reveals systematic patterns of molecular exclusivity and co-occurrence among distinct therapeutic targets

Niantao Deng; Liang Kee Goh; Hannah Wang; Kakoli Das; Jiong Tao; Iain Beehuat Tan; Shenli Zhang; Minghui Lee; Jeanie Wu; Kiat Hon Lim; Zhengdeng Lei; Glenn Goh; Qing-Yan Lim; Angie Lay-Keng Tan; Dianne Yu Sin Poh; Sudep Riahi; Sandra Bell; Michael M. Shi; Ronald Richard Linnartz; Feng-Cai Zhu; Khay Guan Yeoh; Han Chong Toh; Wei Peng Yong; Hyun Cheol Cheong; Sun Young Rha; Alex Boussioutas; Heike I. Grabsch; Steve Rozen; Patrick Tan

Objective Gastric cancer is a major gastrointestinal malignancy for which targeted therapies are emerging as treatment options. This study sought to identify the most prevalent molecular targets in gastric cancer and to elucidate systematic patterns of exclusivity and co-occurrence among these targets, through comprehensive genomic analysis of a large panel of gastric cancers. Design Using high-resolution single nucleotide polymorphism arrays, copy number alterations were profiled in a panel of 233 gastric cancers (193 primary tumours, 40 cell lines) and 98 primary matched gastric non-malignant samples. For selected alterations, their impact on gene expression and clinical outcome were evaluated. Results 22 recurrent focal alterations (13 amplifications and nine deletions) were identified. These included both known targets (FGFR2, ERBB2) and also novel genes in gastric cancer (KLF5, GATA6). Receptor tyrosine kinase (RTK)/RAS alterations were found to be frequent in gastric cancer. This study also demonstrates, for the first time, that these alterations occur in a mutually exclusive fashion, with KRAS gene amplifications highlighting a clinically relevant but previously underappreciated gastric cancer subgroup. FGFR2-amplified gastric cancers were also shown to be sensitive to dovitinib, an orally bioavailable FGFR/VEGFR targeting agent, potentially representing a subtype-specific therapy for FGFR2-amplified gastric cancers. Conclusion The study demonstrates the existence of five distinct gastric cancer patient subgroups, defined by the signature genomic alterations FGFR2 (9% of tumours), KRAS (9%), EGFR (8%), ERBB2 (7%) and MET (4%). Collectively, these subgroups suggest that at least 37% of gastric cancer patients may be potentially treatable by RTK/RAS directed therapies.


PLOS Genetics | 2009

Oncogenic pathway combinations predict clinical prognosis in gastric cancer.

Chia Huey Ooi; Tatiana Ivanova; Jeanie Wu; Minghui Lee; Iain Beehuat Tan; Jiong Tao; Lindsay C. Ward; Jun Hao Koo; Veena Gopalakrishnan; Yansong Zhu; Lai Ling Cheng; Julian Lee; Sun Young Rha; Hyun Cheol Chung; Kumaresan Ganesan; Jimmy So; Khee Chee Soo; Dennis T. H. Lim; Weng Hoong Chan; Wai Keong Wong; David Bowtell; Khay Guan Yeoh; Heike Grabsch; Alex Boussioutas; Patrick Tan

Many solid cancers are known to exhibit a high degree of heterogeneity in their deregulation of different oncogenic pathways. We sought to identify major oncogenic pathways in gastric cancer (GC) with significant relationships to patient survival. Using gene expression signatures, we devised an in silico strategy to map patterns of oncogenic pathway activation in 301 primary gastric cancers, the second highest cause of global cancer mortality. We identified three oncogenic pathways (proliferation/stem cell, NF-kappaB, and Wnt/beta-catenin) deregulated in the majority (>70%) of gastric cancers. We functionally validated these pathway predictions in a panel of gastric cancer cell lines. Patient stratification by oncogenic pathway combinations showed reproducible and significant survival differences in multiple cohorts, suggesting that pathway interactions may play an important role in influencing disease behavior. Individual GCs can be successfully taxonomized by oncogenic pathway activity into biologically and clinically relevant subgroups. Predicting pathway activity by expression signatures thus permits the study of multiple cancer-related pathways interacting simultaneously in primary cancers, at a scale not currently achievable by other platforms.


Science Translational Medicine | 2012

Methylation Subtypes and Large-Scale Epigenetic Alterations in Gastric Cancer

Hermioni Zouridis; Niantao Deng; Tatiana Ivanova; Yansong Zhu; Wong B; Dan Huang; Yingting Wu; Yonghui Wu; Iain Bee Huat Tan; Natalia Liem; Gopalakrishnan; Luo Q; Jeanie Wu; Minghui Lee; Wei Peng Yong; Liang Goh; Bin Tean Teh; Steve Rozen; Patrick Tan

A large-scale genomic survey of epigenetic alterations in gastric cancer identifies clinically relevant molecular subgroups. The Silent Treatment of Gastric Cancer A new study by Zouridis and colleagues refutes the old adage that “silence is golden”—at least in the realm of gene methylation and epigenetic silencing in cancer. To decipher the effects of “silence” on gastric cancer, the authors analyzed gene methylation patterns in 240 gastric tumors and compared them to those of 94 matched samples of adjacent normal tissue. Gastric cancer is one of the most common types of cancer worldwide—and one of the most deadly, with few effective treatment options available. As a possible source of therapeutic targets, scientists are searching for genetic and epigenetic alteration patterns characteristic of these tumors. Here, the authors extensively characterized methylation patterns in human gastric cancers, which revealed tumor-specific arrangements of hyper- and hypomethylation. Zouridis and colleagues also identified a subset of cancers that fell into the CpG island methylator phenotype (CIMP) subgroup, which is associated with more extensive methylation and lower chances of survival in younger patients. As a possible pharmaceutical intervention, the authors tested the effects of the demethylating drug 5-aza-2′-deoxycytidine in CIMP tumor cell lines and found that their proliferation was significantly decreased when compared with non-CIMP cell lines. A broader analysis of gene regions that undergo modifications in cancers likely will identify new therapeutic targets and corresponding treatments. But for patients with high-risk gastric cancers that fall into the CIMP subgroup, silenced DNA is golden because it serves as a target for currently available drugs. Epigenetic alterations are fundamental hallmarks of cancer genomes. We surveyed the landscape of DNA methylation alterations in gastric cancer by analyzing genome-wide CG dinucleotide (CpG) methylation profiles of 240 gastric cancers (203 tumors and 37 cell lines) and 94 matched normal gastric tissues. Cancer-specific epigenetic alterations were observed in 44% of CpGs, comprising both tumor hyper- and hypomethylation. Twenty-five percent of the methylation alterations were significantly associated with changes in tumor gene expression. Whereas most methylation-expression correlations were negative, several positively correlated methylation-expression interactions were also observed, associated with CpG sites exhibiting atypical transcription start site distances and gene body localization. Methylation clustering of the tumors revealed a CpG island methylator phenotype (CIMP) subgroup associated with widespread hypermethylation, young patient age, and adverse patient outcome in a disease stage–independent manner. CIMP cell lines displayed sensitivity to 5-aza-2′-deoxycytidine, a clinically approved demethylating drug. We also identified long-range regions of epigenetic silencing (LRESs) in CIMP tumors. Combined analysis of the methylation, gene expression, and drug treatment data suggests that certain LRESs may silence specific genes within the region, rather than all genes. Finally, we discovered regions of long-range tumor hypomethylation, associated with increased chromosomal instability. Our results provide insights into the epigenetic impact of environmental and biological agents on gastric epithelial cells, which may contribute to cancer.


Gut | 2013

Comprehensive genomic meta-analysis identifies intra-tumoural stroma as a predictor of survival in patients with gastric cancer

Yonghui Wu; Heike I. Grabsch; Tatiana Ivanova; Iain Beehuat Tan; Jacinta Murray; Chia Huey Ooi; Alexander Wright; Nicholas P. West; Gordon Hutchins; Jeanie Wu; Minghui Lee; Julian Lee; Jun Hao Koo; Khay Guan Yeoh; Nicole C.T. van Grieken; Bauke Ylstra; Sun Young Rha; Jaffer A. Ajani; Jae Ho Cheong; Sung Hoon Noh; Kiat Hon Lim; Alex Boussioutas; Ju Seog Lee; Patrick Tan

Objective Gastric adenocarcinoma (gastric cancer, GC) is a major cause of global cancer mortality. Identifying molecular programmes contributing to GC patient survival may improve our understanding of GC pathogenesis, highlight new prognostic factors and reveal novel therapeutic targets. The authors aimed to produce a comprehensive inventory of gene expression programmes expressed in primary GCs, and to identify those expression programmes significantly associated with patient survival. Design Using a network-modelling approach, the authors performed a large-scale meta-analysis of GC transcriptome data integrating 940 gastric transcriptomes from multiple independent patient cohorts. The authors analysed a training set of 428 GCs and 163 non-malignant gastric samples, and a validation set of 288 GCs and 61 non-malignant gastric samples. Results The authors identified 178 gene expression programmes (‘modules’) expressed in primary GCs, which were associated with distinct biological processes, chromosomal location patterns, cis-regulatory motifs and clinicopathological parameters. Expression of a transforming growth factor β (TGF-β) signalling associated ‘super-module’ of stroma-related genes consistently predicted patient survival in multiple GC validation cohorts. The proportion of intra-tumoural stroma, quantified by morphometry in tissue sections from gastrectomy specimens, was also significantly associated with stromal super-module expression and GC patient survival. Conclusion Stromal gene expression predicts GC patient survival in multiple independent cohorts, and may be closely related to the intra-tumoural stroma proportion, a specific morphological GC phenotype. These findings suggest that therapeutic approaches targeting the GC stroma may merit evaluation.


Gut | 2013

Integrated epigenomics identifies BMP4 as a modulator of cisplatin sensitivity in gastric cancer

Tatiana Ivanova; Hermioni Zouridis; Yonghui Wu; Lai Ling Cheng; Iain Beehuat Tan; Veena Gopalakrishnan; Chia Huey Ooi; Julian Lee; Luo Qin; Jeanie Wu; Minghui Lee; Sun Young Rha; Dan Huang; Natalia Liem; Khay Guan Yeoh; Wei Peng Yong; Bin Tean Teh; Patrick Tan

Objective Cisplatin is a widely used gastric cancer (GC) chemotherapy; however, genetic factors regulating GC responses to cisplatin remain obscure. Identifying genes regulating cisplatin resistance could aid clinicians in tailoring treatments, by distinguishing cisplatin sensitive patients from those who might benefit from alternative platinum therapies, and highlight novel targeted strategies for overcoming cisplatin resistance. Here integrated epigenomics is applied to identify genes associated with GC cisplatin resistance. Design 20 GC cell lines were subjected to gene expression profiling, DNA methylation profiling and drug response assays. The molecular data were integrated to identify genes highly expressed and unmethylated specifically in cisplatin-resistant lines. Candidate genes were functionally tested by several in vitro and in vivo assays. Clinical impact of candidate genes was also assessed in a cohort of 197 GC patients. Results Epigenomic analysis identified bone morphogenetic protein 4 (BMP4) as an epigenetically regulated gene highly expressed in cisplatin-resistant lines. Functional assays confirmed that BMP4 is necessary and sufficient for the expression of several prooncogenic traits, likely mediated through stimulation of the epithelial-mesenchymal transition. In primary tumours, BMP4 promoter methylation levels were inversely correlated with BMP4 expression, and patients with high BMP4-expressing tumours exhibited significantly worse prognosis. Therapeutically, targeted genetic inhibition of BMP4 caused significant sensitisation of GC cells to cisplatin. Notably, BMP4-expressing GCs also did not exhibit cross resistance to oxaliplatin. Conclusions BMP4 epigenetic and expression status may represent promising biomarkers for GC cisplatin resistance. Targeting BMP4 may sensitise GC cells to cisplatin. Oxaliplatin, a clinically acceptable cisplatin alternative, may represent a potential therapeutic option for BMP4-positive GCs.


Gut | 2015

Regulatory crosstalk between lineage-survival oncogenes KLF5, GATA4 and GATA6 cooperatively promotes gastric cancer development.

Na-Yu Chia; Niantao Deng; Kakoli Das; Dachuan Huang; Longyu Hu; Yansong Zhu; Kiat Hon Lim; Minghui Lee; Jeanie Wu; Xin Xiu Sam; Gek San Tan; Wei Keat Wan; Willie Yu; Anna Gan; Angie Lay Keng Tan; Tay St; Khee Chee Soo; Wai Keong Wong; Lourdes Trinidad M Dominguez; Huck-Hui Ng; Steve Rozen; Liang Kee Goh; Bin-Tean Teh; Patrick Tan

Objective Gastric cancer (GC) is a deadly malignancy for which new therapeutic strategies are needed. Three transcription factors, KLF5, GATA4 and GATA6, have been previously reported to exhibit genomic amplification in GC. We sought to validate these findings, investigate how these factors function to promote GC, and identify potential treatment strategies for GCs harbouring these amplifications. Design KLF5, GATA4 and GATA6 copy number and gene expression was examined in multiple GC cohorts. Chromatin immunoprecipitation with DNA sequencing was used to identify KLF5/GATA4/GATA6 genomic binding sites in GC cell lines, and integrated with transcriptomics to highlight direct target genes. Phenotypical assays were conducted to assess the function of these factors in GC cell lines and xenografts in nude mice. Results KLF5, GATA4 and GATA6 amplifications were confirmed in independent GC cohorts. Although factor amplifications occurred in distinct sets of GCs, they exhibited significant mRNA coexpression in primary GCs, consistent with KLF5/GATA4/GATA6 cross-regulation. Chromatin immunoprecipitation with DNA sequencing revealed a large number of genomic sites co-occupied by KLF5 and GATA4/GATA6, primarily located at gene promoters and exhibiting higher binding strengths. KLF5 physically interacted with GATA factors, supporting KLF5/GATA4/GATA6 cooperative regulation on co-occupied genes. Depletion and overexpression of these factors, singly or in combination, reduced and promoted cancer proliferation, respectively, in vitro and in vivo. Among the KLF5/GATA4/GATA6 direct target genes relevant for cancer development, one target gene, HNF4α, was also required for GC proliferation and could be targeted by the antidiabetic drug metformin, revealing a therapeutic opportunity for KLF5/GATA4/GATA6 amplified GCs. Conclusions KLF5/GATA4/GATA6 may promote GC development by engaging in mutual crosstalk, collaborating to maintain a pro-oncogenic transcriptional regulatory network in GC cells.


Science Translational Medicine | 2011

CD44-SLC1A2 Gene Fusions in Gastric Cancer

Jiong Tao; Niantao Deng; Kalpana Ramnarayanan; Baohua Huang; Hue Kian Oh; Siew Hong Leong; Seong Soo Lim; Iain Beehuat Tan; Chia Huey Ooi; Jeanie Wu; Minghui Lee; Shenli Zhang; Sun Young Rha; Hyun Cheol Chung; Duane T. Smoot; Hassan Ashktorab; Oi Lian Kon; Valere Cacheux; Celestial T. Yap; Nallasivam Palanisamy; Patrick Tan

One partner of a fusion gene found in gastric cancer, CD44-SLC1A2, may contribute to the tumor’s abnormal metabolism. Bad Drivers Steer Scientists Toward New Drug Targets It’s ironic, but cancer cells are notoriously bad at cell division, losing bits and rearranging chunks of the genome in the process. One result of this chaos is the birth of chimeric genes, wherein one gene segment gets erroneously fused to part of another, sometimes forming peculiar hybrid proteins that contribute to the cancer cell phenotype. For example, the fused aberrant BCR-ABL gene drives chronic myelogenous leukemia and has proven to be a vulnerable target for therapy. Gene fusions in solid cancers are not so easy to spot, but have been located in prostate and small cell lung cancers. Now, Tao and her co-workers have documented a fusion gene that forms in a small percentage of gastric tumor cells and may contribute to the development of cancer. The authors analyzed copy number variations of genes in more than 100 primary gastric tumors and 27 established gastric tumor cell lines and pinpointed a common breakpoint in three and one, respectively. The resulting chimeric gene fused most of the coding region of SLC1A2/EAAT2 (which encodes a glutamate transporter) to what is probably the strong transcriptional promoter of its neighboring gene, CD44, likely the result of a chromosome inversion. The fusion gene generated a truncated SLC1A2 protein in the original tumors and in a new group of gastric cancers created by the authors through overexpression of the fusion gene in normal gastric cells. But an abnormal protein that lives in tumor cells can be an innocent bystander. So, the authors asked whether the truncated SLC1A2 contributes to gastric cancer development, and their evidence suggested that the answer is yes. Cells in which shortened SLC1A2 expression was silenced with small interfering RNA were less proficient at dividing and invading soft substrates—hallmarks of cancer cells—and overexpression of the pruned protein enhanced these traits. Consistent with the function of SLC1A2 as a transporter of glutamate, the amino acid—which can act as a growth regulator—existed in higher concentrations in gastric cancer cells and cell lines than in normal cells. And in a final set of incriminating evidence, tumor cells that sported the CD44-SLC1A2 fusion gene had higher amounts of SLC1A2 than did wild-type cells, suggesting that this aberrant protein may trigger a pro-oncogenic phenotype. Most other genes that are fused in cancers encode kinase enzymes or transcriptional regulatory proteins. The implication of an overexpressed metabolism-related gene in some gastric tumors may define a new class of cancer-driving genes, although the protein could also augment other cancer-promoting genetic aberrations. The utility of this fusion gene as a drug target or prognostic tool will require more studies, but this particular mistake made by a dividing cancer cell may act as a GPS that directs researchers down a new therapeutic avenue for gastric cancer. Fusion genes are chimeric genes formed in cancers through genomic aberrations such as translocations, amplifications, and rearrangements. To identify fusion genes in gastric cancer, we analyzed regions of chromosomal imbalance in a cohort of 106 primary gastric cancers and 27 cell lines derived from gastric cancers. Multiple samples exhibited genomic breakpoints in the 5′ region of SLC1A2/EAAT2, a gene encoding a glutamate transporter. Analysis of a breakpoint-positive SNU16 cell line revealed expression of a CD44-SLC1A2 fusion transcript caused by a paracentric chromosomal inversion, which was predicted to produce a truncated but functional SLC1A2 protein. In primary tumors, CD44-SLC1A2 gene fusions were detected in 1 to 2% of gastric cancers, but not in adjacent matched normal gastric tissues. When we specifically silenced CD44-SLC1A2, cellular proliferation, invasion, and anchorage-independent growth were significantly reduced. Conversely, CD44-SLC1A2 overexpression in gastric cells stimulated these pro-oncogenic traits. CD44-SLC1A2 silencing caused significant reductions in intracellular glutamate concentrations and sensitized SNU16 cells to cisplatin, a commonly used chemotherapeutic agent in gastric cancer. We conclude that fusion of the SLC1A2 gene coding region to CD44 regulatory elements likely causes SLC1A2 transcriptional dysregulation, because tumors expressing high SLC1A2 levels also tended to be CD44-SLC1A2–positive. CD44-SLC1A2 may represent a class of gene fusions in cancers that establish a pro-oncogenic metabolic milieu favoring tumor growth and survival.


PLOS Genetics | 2011

A Densely Interconnected Genome-Wide Network of MicroRNAs and Oncogenic Pathways Revealed Using Gene Expression Signatures

Chia Huey Ooi; Hue Kian Oh; Hannah Zhu Ai Wang; Angie Lay Keng Tan; Jeanie Wu; Minghui Lee; Sun Young Rha; Hyun Cheol Chung; David M. Virshup; Patrick Tan

MicroRNAs (miRNAs) are important components of cellular signaling pathways, acting either as pathway regulators or pathway targets. Currently, only a limited number of miRNAs have been functionally linked to specific signaling pathways. Here, we explored if gene expression signatures could be used to represent miRNA activities and integrated with genomic signatures of oncogenic pathway activity to identify connections between miRNAs and oncogenic pathways on a high-throughput, genome-wide scale. Mapping >300 gene expression signatures to >700 primary tumor profiles, we constructed a genome-wide miRNA–pathway network predicting the associations of 276 human miRNAs to 26 oncogenic pathways. The miRNA–pathway network confirmed a host of previously reported miRNA/pathway associations and uncovered several novel associations that were subsequently experimentally validated. Globally, the miRNA–pathway network demonstrates a small-world, but not scale-free, organization characterized by multiple distinct, tightly knit modules each exhibiting a high density of connections. However, unlike genetic or metabolic networks typified by only a few highly connected nodes (“hubs”), most nodes in the miRNA–pathway network are highly connected. Sequence-based computational analysis confirmed that highly-interconnected miRNAs are likely to be regulated by common pathways to target similar sets of downstream genes, suggesting a pervasive and high level of functional redundancy among coexpressed miRNAs. We conclude that gene expression signatures can be used as surrogates of miRNA activity. Our strategy facilitates the task of discovering novel miRNA–pathway connections, since gene expression data for multiple normal and disease conditions are abundantly available.


Cancer Cell | 2018

Genomic and Epigenomic Profiling of High-Risk Intestinal Metaplasia Reveals Molecular Determinants of Progression to Gastric Cancer

Kie Kyon Huang; Kalpana Ramnarayanan; Feng Zhu; Supriya Srivastava; Chang Xu; Angie Lay Keng Tan; Minghui Lee; Tay St; Kakoli Das; Manjie Xing; Aliya Fatehullah; Syed Muhammad Fahmy Alkaff; Tony Kiat Hon Lim; Jonathan Wj Lee; Khek Yu Ho; Steven G. Rozen; Bin Tean Teh; Nick Barker; Chung King Chia; Christopher Jen Lock Khor; Choon Jin Ooi; Kwong Ming Fock; Jimmy So; Wee Chian Lim; Khoon Lin Ling; Tiing Leong Ang; Andrew Siang Yih Wong; Jaideepraj Rao; Andrea Rajnakova; Lee Guan Lim

Intestinal metaplasia (IM) is a pre-malignant condition of the gastric mucosa associated with increased gastric cancer (GC) risk. We performed (epi)genomic profiling of 138 IMs from 148 cancer-free patients, recruited through a 10-year prospective study. Compared with GCs, IMs exhibit low mutational burdens, recurrent mutations in certain tumor suppressors (FBXW7) but not others (TP53, ARID1A), chromosome 8q amplification, and shortened telomeres. Sequencing identified more IM patients with active Helicobacter pylori infection compared with histopathology (11%-27%). Several IMs exhibited hypermethylation at DNA methylation valleys; however, IMs generally lack intragenic hypomethylation signatures of advanced malignancy. IM patients with shortened telomeres and chromosomal alterations were associated with subsequent dysplasia or GC; conversely patients exhibiting normal-like epigenomic patterns were associated with regression.


Oncogene | 2014

RNH1 regulation of reactive oxygen species contributes to histone deacetylase inhibitor resistance in gastric cancer cells

Yansong Zhu; Kakoli Das; Jeanie Wu; Minghui Lee; Patrick Tan

Histone deacetylase inhibitors (HDACis) are a promising class of anticancer epigenetic drugs, however, molecular factors influencing the responses of individual tumors to HDACi therapies remain obscure. Here, we sought to identify genes associated with HDACi resistance in gastric cancer. Treating a panel of 17 gastric cancer cell lines with multiple HDACi compounds (trichostatin A, SAHA and MS275), we identified two distinct classes of lines exhibiting either HDACi sensitivity or resistance. Genomic comparisons between the sensitive and resistant classes using two independent microarray platforms identified RNH1, encoding a ribonuclease inhibitor, as a gene highly expressed in HDACi-resistant lines. Using genetic knockdown and overexpression assays, we show that RNH1 is both necessary and sufficient to induce HDACi resistance, and that RNH1 is likely to mediate this resistance through the dampening of HDACi-induced reactive oxygen species (ROS) in cancer cells. The discovery of RNH1 as a regulator of HDACi resistance in gastric cancer highlights a functional role for ROS induction in the cellular effects of this important drug class.

Collaboration


Dive into the Minghui Lee's collaboration.

Top Co-Authors

Avatar

Patrick Tan

National University of Singapore

View shared research outputs
Top Co-Authors

Avatar

Jeanie Wu

National University of Singapore

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chia Huey Ooi

National University of Singapore

View shared research outputs
Top Co-Authors

Avatar

Iain Beehuat Tan

National University of Singapore

View shared research outputs
Top Co-Authors

Avatar

Tatiana Ivanova

National University of Singapore

View shared research outputs
Top Co-Authors

Avatar

Khay Guan Yeoh

National University of Singapore

View shared research outputs
Top Co-Authors

Avatar

Shenli Zhang

National University of Singapore

View shared research outputs
Top Co-Authors

Avatar

Alex Boussioutas

Peter MacCallum Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Bin Tean Teh

National University of Singapore

View shared research outputs
Researchain Logo
Decentralizing Knowledge