Network


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

Hotspot


Dive into the research topics where Stanley W.K. Ng is active.

Publication


Featured researches published by Stanley W.K. Ng.


Nature | 2016

A 17-gene stemness score for rapid determination of risk in acute leukaemia.

Stanley W.K. Ng; Amanda Mitchell; James A. Kennedy; Weihsu C. Chen; Jessica McLeod; Narmin Ibrahimova; Andrea Arruda; Andreea C. Popescu; Vikas Gupta; Aaron D. Schimmer; Andre C. Schuh; Karen Yee; Lars Bullinger; Tobias Herold; Dennis Görlich; Thomas Büchner; Wolfgang Hiddemann; Wolfgang E. Berdel; Bernhard Wörmann; Meyling Cheok; Claude Preudhomme; Hervé Dombret; Klaus H. Metzeler; Christian Buske; Bob Löwenberg; Peter W. Zandstra; Mark D. Minden; John E. Dick; Jean C.Y. Wang

Refractoriness to induction chemotherapy and relapse after achievement of remission are the main obstacles to cure in acute myeloid leukaemia (AML). After standard induction chemotherapy, patients are assigned to different post-remission strategies on the basis of cytogenetic and molecular abnormalities that broadly define adverse, intermediate and favourable risk categories. However, some patients do not respond to induction therapy and another subset will eventually relapse despite the lack of adverse risk factors. There is an urgent need for better biomarkers to identify these high-risk patients before starting induction chemotherapy, to enable testing of alternative induction strategies in clinical trials. The high rate of relapse in AML has been attributed to the persistence of leukaemia stem cells (LSCs), which possess a number of stem cell properties, including quiescence, that are linked to therapy resistance. Here, to develop predictive and/or prognostic biomarkers related to stemness, we generated a list of genes that are differentially expressed between 138 LSC+ and 89 LSC− cell fractions from 78 AML patients validated by xenotransplantation. To extract the core transcriptional components of stemness relevant to clinical outcomes, we performed sparse regression analysis of LSC gene expression against survival in a large training cohort, generating a 17-gene LSC score (LSC17). The LSC17 score was highly prognostic in five independent cohorts comprising patients of diverse AML subtypes (n = 908) and contributed greatly to accurate prediction of initial therapy resistance. Patients with high LSC17 scores had poor outcomes with current treatments including allogeneic stem cell transplantation. The LSC17 score provides clinicians with a rapid and powerful tool to identify AML patients who do not benefit from standard therapy and who should be enrolled in trials evaluating novel upfront or post-remission strategies.


Cancer Cell | 2016

miR-126 Regulates Distinct Self-Renewal Outcomes in Normal and Malignant Hematopoietic Stem Cells

Eric R. Lechman; Bernhard Gentner; Stanley W.K. Ng; Erwin M. Schoof; James A. Kennedy; Silvia Nucera; Fabio Ciceri; Kerstin B Kaufmann; Naoya Takayama; Stephanie M. Dobson; Aaron Trotman-Grant; Gabriela Krivdova; Janneke Elzinga; Amanda Mitchell; Björn Nilsson; Karin G. Hermans; Kolja Eppert; Rene Marke; Ruth Isserlin; Veronique Voisin; Gary D. Bader; Peter W. Zandstra; Todd R. Golub; Benjamin L. Ebert; Jun Lu; Mark D. Minden; Jean C.Y. Wang; Luigi Naldini; John E. Dick

Summary To investigate miRNA function in human acute myeloid leukemia (AML) stem cells (LSC), we generated a prognostic LSC-associated miRNA signature derived from functionally validated subpopulations of AML samples. For one signature miRNA, miR-126, high bioactivity aggregated all in vivo patient sample LSC activity into a single sorted population, tightly coupling miR-126 expression to LSC function. Through functional studies, miR-126 was found to restrain cell cycle progression, prevent differentiation, and increase self-renewal of primary LSC in vivo. Compared with prior results showing miR-126 regulation of normal hematopoietic stem cell (HSC) cycling, these functional stem effects are opposite between LSC and HSC. Combined transcriptome and proteome analysis demonstrates that miR-126 targets the PI3K/AKT/MTOR signaling pathway, preserving LSC quiescence and promoting chemotherapy resistance.


Nature | 2017

Tracing the origins of relapse in acute myeloid leukaemia to stem cells

Liran I. Shlush; Amanda Mitchell; Lawrence E. Heisler; Sagi Abelson; Stanley W.K. Ng; Aaron Trotman-Grant; Jessie J. F. Medeiros; Abilasha Rao-Bhatia; Ivana Jaciw-Zurakowsky; Rene Marke; Jessica McLeod; Monica Doedens; Gary D. Bader; Veronique Voisin; ChangJiang Xu; John D. McPherson; Thomas J. Hudson; Jean C.Y. Wang; Mark D. Minden; John E. Dick

In acute myeloid leukaemia, long-term survival is poor as most patients relapse despite achieving remission. Historically, the failure of therapy has been thought to be due to mutations that produce drug resistance, possibly arising as a consequence of the mutagenic properties of chemotherapy drugs. However, other lines of evidence have pointed to the pre-existence of drug-resistant cells. For example, deep sequencing of paired diagnosis and relapse acute myeloid leukaemia samples has provided direct evidence that relapse in some cases is generated from minor genetic subclones present at diagnosis that survive chemotherapy, suggesting that resistant cells are generated by evolutionary processes before treatment and are selected by therapy. Nevertheless, the mechanisms of therapy failure and capacity for leukaemic regeneration remain obscure, as sequence analysis alone does not provide insight into the cell types that are fated to drive relapse. Although leukaemia stem cells have been linked to relapse owing to their dormancy and self-renewal properties, and leukaemia stem cell gene expression signatures are highly predictive of therapy failure, experimental studies have been primarily correlative and a role for leukaemia stem cells in acute myeloid leukaemia relapse has not been directly proved. Here, through combined genetic and functional analysis of purified subpopulations and xenografts from paired diagnosis/relapse samples, we identify therapy-resistant cells already present at diagnosis and two major patterns of relapse. In some cases, relapse originated from rare leukaemia stem cells with a haematopoietic stem/progenitor cell phenotype, while in other instances relapse developed from larger subclones of immunophenotypically committed leukaemia cells that retained strong stemness transcriptional signatures. The identification of distinct patterns of relapse should lead to improved methods for disease management and monitoring in acute myeloid leukaemia. Moreover, the shared functional and transcriptional stemness properties that underlie both cellular origins of relapse emphasize the importance of developing new therapeutic approaches that target stemness to prevent relapse.


Nature | 2018

A Myc enhancer cluster regulates normal and leukaemic haematopoietic stem cell hierarchies

Carsten Bahr; Lisa von Paleske; Veli Vural Uslu; Silvia Remeseiro; Naoya Takayama; Stanley W.K. Ng; Alex Murison; Katja Langenfeld; Massimo Petretich; Roberta Scognamiglio; Petra Zeisberger; Amelie S. Benk; Ido Amit; Peter W. Zandstra; Mathieu Lupien; John E. Dick; Andreas Trumpp; François Spitz

The transcription factor Myc is essential for the regulation of haematopoietic stem cells and progenitors and has a critical function in haematopoietic malignancies. Here we show that an evolutionarily conserved region located 1.7 megabases downstream of the Myc gene that has previously been labelled as a ‘super-enhancer’ is essential for the regulation of Myc expression levels in both normal haematopoietic and leukaemic stem cell hierarchies in mice and humans. Deletion of this region in mice leads to a complete loss of Myc expression in haematopoietic stem cells and progenitors. This caused an accumulation of differentiation-arrested multipotent progenitors and loss of myeloid and B cells, mimicking the phenotype caused by Mx1-Cre-mediated conditional deletion of the Myc gene in haematopoietic stem cells. This super-enhancer comprises multiple enhancer modules with selective activity that recruits a compendium of transcription factors, including GFI1b, RUNX1 and MYB. Analysis of mice carrying deletions of individual enhancer modules suggests that specific Myc expression levels throughout most of the haematopoietic hierarchy are controlled by the combinatorial and additive activity of individual enhancer modules, which collectively function as a ‘blood enhancer cluster’ (BENC). We show that BENC is also essential for the maintenance of MLL–AF9-driven leukaemia in mice. Furthermore, a BENC module, which controls Myc expression in mouse haematopoietic stem cells and progenitors, shows increased chromatin accessibility in human acute myeloid leukaemia stem cells compared to blasts. This difference correlates with MYC expression and patient outcome. We propose that clusters of enhancers, such as BENC, form highly combinatorial systems that allow precise control of gene expression across normal cellular hierarchies and which also can be hijacked in malignancies.


Nature | 2018

Author Correction: A Myc enhancer cluster regulates normal and leukaemic haematopoietic stem cell hierarchies

Carsten Bahr; Lisa von Paleske; Veli Vural Uslu; Silvia Remeseiro; Naoya Takayama; Stanley W.K. Ng; Alex Murison; Katja Langenfeld; Massimo Petretich; Roberta Scognamiglio; Petra Zeisberger; Amelie S. Benk; Ido Amit; Peter W. Zandstra; Mathieu Lupien; John E. Dick; Andreas Trumpp; François Spitz

In the originally published version of this Letter, ref. 43 was erroneously provided twice. In the ‘Estimation of relative cell-type-specific composition of AML samples’ section in the Methods, the citation to ref. 43 after the GEO dataset GSE24759 is correct. However, in the ‘Mice’ section of the Methods, the citation to ref. 43 after ‘TAMERE’ should have been associated with a new reference1. The original Letter has been corrected online (with the new reference included as ref. 49).


Leukemia | 2018

The stem cell-associated gene expression signature allows risk stratification in pediatric acute myeloid leukemia.

Nicolas Duployez; Alice Marceau-Renaut; Céline Villenet; Arnaud Petit; Alexandra Rousseau; Stanley W.K. Ng; Agnès Paquet; Fanny Gonzales; Adeline Barthelemy; Frédéric Leprêtre; Nicolas Pottier; Brigitte Nelken; Gérard Michel; André Baruchel; Yves Bertrand; Guy Leverger; Hélène Lapillonne; Martin Figeac; John E. Dick; Jean C.Y. Wang; Claude Preudhomme; Meyling Cheok

Despite constant progress in prognostic risk stratification, children with acute myeloid leukemia (AML) still relapse. Treatment failure and subsequent relapse have been attributed to acute myeloid leukemia-initiating cells (LSC), which harbor stem cell properties and are inherently chemoresistant. Although pediatric and adult AML represent two genetically very distinct diseases, we reasoned that common LSC gene expression programs are shared and consequently, the highly prognostic LSC17 signature score recently developed in adults may also be of clinical interest in childhood AML. Here, we demonstrated prognostic relevance of the LSC17 score in pediatric non-core-binding factor AML using Nanostring technology (ELAM02) and RNA-seq data from the NCI (TARGET-AML). AML were stratified by LSC17 quartile groups (lowest 25%, intermediate 50% and highest 25%) and children with low LSC17 score had significantly better event-free survival (EFS: HR = 3.35 (95%CI = 1.64–6.82), P < 0.001) and overall survival (OS: HR = 3.51 (95%CI = 1.38–8.92), P = 0.008) compared with patients with high LSC17 scores. More importantly, the high LSC17 score was an independent negative EFS and OS prognosticator determined by multivariate Cox model analysis (EFS: HR = 3.42 (95% CI = 1.63–7.16), P = 0.001; OS HR = 3.02 (95%CI = 1.16–7.85), P = 0.026). In conclusion, we have demonstrated the broad applicability of the LSC17 score in the clinical management of AML by extending its prognostic relevance to pediatric AML.


Blood Cancer Journal | 2018

Leukemic stem cell signatures identify novel therapeutics targeting acute myeloid leukemia

Isabelle Laverdière; Meaghan Boileau; Andrea Neumann; Héloïse Frison; Amanda Mitchell; Stanley W.K. Ng; Jean C.Y. Wang; Mark D. Minden; Kolja Eppert

Therapy for acute myeloid leukemia (AML) involves intense cytotoxic treatment and yet approximately 70% of AML are refractory to initial therapy or eventually relapse. This is at least partially driven by the chemo-resistant nature of the leukemic stem cells (LSCs) that sustain the disease, and therefore novel anti-LSC therapies could decrease relapses and improve survival. We performed in silico analysis of highly prognostic human AML LSC gene expression signatures using existing datasets of drug–gene interactions to identify compounds predicted to target LSC gene programs. Filtering against compounds that would inhibit a hematopoietic stem cell (HSC) gene signature resulted in a list of 151 anti-LSC candidates. Using a novel in vitro LSC assay, we screened 84 candidate compounds at multiple doses and confirmed 14 drugs that effectively eliminate human AML LSCs. Three drug families presenting with multiple hits, namely antihistamines (astemizole and terfenadine), cardiac glycosides (strophanthidin, digoxin and ouabain) and glucocorticoids (budesonide, halcinonide and mometasone), were validated for their activity against human primary AML samples. Our study demonstrates the efficacy of combining computational analysis of stem cell gene expression signatures with in vitro screening to identify novel compounds that target the therapy-resistant LSC at the root of relapse in AML.


Cancer Cell | 2016

Corrections: miR-126 Regulates Distinct Self-Renewal Outcomes in Normal and Malignant Hematopoietic Stem Cells

Eric R. Lechman; Bernhard Gentner; Stanley W.K. Ng; Erwin M. Schoof; James A. Kennedy; Silvia Nucera; Fabio Ciceri; Kerstin B. Kaufmann; Naoya Takayama; Stephanie M. Dobson; Aaron Trotman-Grant; Gabriela Krivdova; Janneke Elzinga; Amanda C. Mitchell; Björn Nilsson; Karin G. Hermans; Kolja Eppert; Rene Marke; Ruth Isserlin; Veronique Voisin; Gary D. Bader; Peter W. Zandstra; Todd R. Golub; Benjamin L. Ebert; Jun Lu; Mark D. Minden; Jean C.Y. Wang; Luigi Naldini; John E. Dick


Experimental Hematology | 2016

Identification of existing bioactive molecules that target acute myeloid leukemia stem cells

Meaghan Boileau; Isabelle Laverdière; Amanda Mitchell; Stanley W.K. Ng; Jean C.Y. Wang; Mark D. Minden; John E. Dick; Kolja Eppert


Blood | 2016

CD200 Is a Marker of LSC Activity in Acute Myeloid Leukemia

Jenny M. Ho; Stephanie M. Dobson; Jessica McLeod; Liqing Jin; Stanley W.K. Ng; Amanda Mitchell; Liran I. Shlush; Sasan Zandi; Kolja Eppert; Mark D. Minden; John E. Dick; Jean C.Y. Wang

Collaboration


Dive into the Stanley W.K. Ng's collaboration.

Top Co-Authors

Avatar

John E. Dick

Princess Margaret Cancer Centre

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mark D. Minden

Princess Margaret Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Amanda Mitchell

University Health Network

View shared research outputs
Top Co-Authors

Avatar

Kolja Eppert

McGill University Health Centre

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jessica McLeod

University Health Network

View shared research outputs
Top Co-Authors

Avatar

Naoya Takayama

Princess Margaret Cancer Centre

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge