Network


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

Hotspot


Dive into the research topics where Wee Joo Chng is active.

Publication


Featured researches published by Wee Joo Chng.


Lancet Oncology | 2016

International Myeloma Working Group consensus criteria for response and minimal residual disease assessment in multiple myeloma

Shaji Kumar; Bruno Paiva; Kenneth C. Anderson; Brian G. M. Durie; Ola Landgren; Philippe Moreau; Nikhil C. Munshi; Sagar Lonial; Joan Bladé; Maria-Victoria Mateos; Meletios A. Dimopoulos; Efstathios Kastritis; Mario Boccadoro; Robert Z. Orlowski; Hartmut Goldschmidt; Andrew Spencer; Jian Hou; Wee Joo Chng; Saad Z Usmani; Elena Zamagni; Kazuyuki Shimizu; Sundar Jagannath; Hans Erik Johnsen; Evangelos Terpos; Anthony Reiman; Robert A. Kyle; Pieter Sonneveld; Paul G. Richardson; Philip L. McCarthy; Heinz Ludwig

Treatment of multiple myeloma has substantially changed over the past decade with the introduction of several classes of new effective drugs that have greatly improved the rates and depth of response. Response criteria in multiple myeloma were developed to use serum and urine assessment of monoclonal proteins and bone marrow assessment (which is relatively insensitive). Given the high rates of complete response seen in patients with multiple myeloma with new treatment approaches, new response categories need to be defined that can identify responses that are deeper than those conventionally defined as complete response. Recent attempts have focused on the identification of residual tumour cells in the bone marrow using flow cytometry or gene sequencing. Furthermore, sensitive imaging techniques can be used to detect the presence of residual disease outside of the bone marrow. Combining these new methods, the International Myeloma Working Group has defined new response categories of minimal residual disease negativity, with or without imaging-based absence of extramedullary disease, to allow uniform reporting within and outside clinical trials. In this Review, we clarify several aspects of disease response assessment, along with endpoints for clinical trials, and highlight future directions for disease response assessments.


Cancer Research | 2014

T Lymphocytes Expressing a CD16 Signaling Receptor Exert Antibody-Dependent Cancer Cell Killing

Ko Kudo; Chihaya Imai; Paolo Lorenzini; Takahiro Kamiya; Koji Kono; Andrew M. Davidoff; Wee Joo Chng; Dario Campana

To expand applications for T-cell-based immunotherapy in cancer, we designed a receptor that binds the Fc portion of human immunoglobulins and delivers activation signals. The construct included the high-affinity CD16 (FCGR3A) V158 variant, CD8α hinge, and transmembrane domains, along with signaling domains from CD3ζ and 4-1BB (TNFRSF9), forming a chimeric receptor termed CD16V-BB-ζ. After retrovirus-mediated expression in human T cells, CD16V-BB-ζ bound humanized antibodies with higher affinity than a control receptor containing the more common F158 variant. Engagement of CD16V-BB-ζ provoked T-cell activation, exocytosis of lytic granules, and sustained proliferation, with a mean cell recovery after 4-week coculture with Daudi lymphoma cells and rituximab of nearly 70-fold relative to input cells. In contrast, unbound antibody alone produced no effect. CD16V-BB-ζ T cells specifically killed lymphoma cells and primary chronic lymphocytic leukemia cells in combination with rituximab at a low effector:target ratio, even when assayed on mesenchymal cells. Trastuzumab triggered CD16V-BB-ζ-mediated killing of HER2 (ERBB2)(+) breast and gastric cancer cells; similar results were obtained with an anti-GD2 antibody in neuroblastoma and osteosarcoma cells. Furthermore, coadministration of CD16V-BB-ζ T cells with immunotherapeutic antibodies exerted considerable antitumor activity in vivo. Signaling mediated by 4-1BB-CD3ζ induced higher T-cell activation, proliferation, and cytotoxicity than CD3ζ or FcεRIγ, and the receptor was expressed effectively after mRNA electroporation without viral vectors, facilitating clinical translation. Our results offer preclinical proof of concept for CD16V-BB-ζ as a universal, next-generation chimeric receptor with the potential to augment the efficacy of antibody therapies for cancer.


Lancet Oncology | 2017

Role of 18F-FDG PET/CT in the diagnosis and management of multiple myeloma and other plasma cell disorders: a consensus statement by the International Myeloma Working Group

Michele Cavo; Evangelos Terpos; Cristina Nanni; Philippe Moreau; Suzanne Lentzsch; Sonja Zweegman; Jens Hillengass; Monika Engelhardt; Saad Z Usmani; David H. Vesole; Jesús F. San-Miguel; Shaji Kumar; Paul G. Richardson; Joseph R. Mikhael; Fernando Leal da Costa; Meletios Athanassios Dimopoulos; Chiara Zingaretti; Niels Abildgaard; Hartmut Goldschmidt; Robert Z. Orlowski; Wee Joo Chng; Hermann Einsele; Sagar Lonial; Bart Barlogie; Kenneth C. Anderson; S. Vincent Rajkumar; Brian G. M. Durie; Elena Zamagni

The International Myeloma Working Group consensus aimed to provide recommendations for the optimal use of 18fluorodeoxyglucose (18F-FDG) PET/CT in patients with multiple myeloma and other plasma cell disorders, including smouldering multiple myeloma and solitary plasmacytoma. 18F-FDG PET/CT can be considered a valuable tool for the work-up of patients with both newly diagnosed and relapsed or refractory multiple myeloma because it assesses bone damage with relatively high sensitivity and specificity, and detects extramedullary sites of proliferating clonal plasma cells while providing important prognostic information. The use of 18F-FDG PET/CT is mandatory to confirm a suspected diagnosis of solitary plasmacytoma, provided that whole-body MRI is unable to be performed, and to distinguish between smouldering and active multiple myeloma, if whole-body X-ray (WBXR) is negative and whole-body MRI is unavailable. Based on the ability of 18F-FDG PET/CT to distinguish between metabolically active and inactive disease, this technique is now the preferred functional imaging modality to evaluate and to monitor the effect of therapy on myeloma-cell metabolism. Changes in FDG avidity can provide an earlier evaluation of response to therapy compared to MRI scans, and can predict outcomes, particularly for patients who are eligible to receive autologous stem-cell transplantation. 18F-FDG PET/CT can be coupled with sensitive bone marrow-based techniques to detect minimal residual disease (MRD) inside and outside the bone marrow, helping to identify those patients who are defined as having imaging MRD negativity.


Blood Cancer Journal | 2011

Pacritinib (SB1518), a JAK2/FLT3 inhibitor for the treatment of acute myeloid leukemia.

Stefan Hart; Kee Chuan Goh; Veronica Novotny-Diermayr; Yong Cheng Tan; Babita Madan; Chithra Amalini; Lai Chun Ong; B Kheng; A Cheong; Jianbiao Zhou; Wee Joo Chng; Jeanette Marjorie Wood

FMS-like tyrosine kinase 3 (FLT3) is the most commonly mutated gene found in acute myeloid leukemia (AML) patients and its activating mutations have been proven to be a negative prognostic marker for clinical outcome. Pacritinib (SB1518) is a tyrosine kinase inhibitor (TKI) with equipotent activity against FLT3 (IC50=22 n) and Janus kinase 2 (JAK2, IC50=23 n). Pacritinib inhibits FLT3 phosphorylation and downstream STAT, MAPK and PI3 K signaling in FLT3-internal-tandem duplication (ITD), FLT3-wt cells and primary AML blast cells. Oral administration of pacritinib in murine models of FLT3-ITD-driven AML led to significant inhibition of primary tumor growth and lung metastasis. Upregulation of JAK2 in FLT3-TKI-resistant AML cells was identified as a potential mechanism of resistance to selective FLT3 inhibition. This resistance could be overcome by the combined FLT3 and JAK2 activities of pacritinib in this cellular model. Our findings provide a rationale for the clinical evaluation of pacritinib in AML including patients resistant to FLT3-TKI therapy.


Molecular Cancer Therapeutics | 2013

VS-5584, a Novel and Highly Selective PI3K/mTOR Kinase Inhibitor for the Treatment of Cancer

Stefan Hart; Veronica Novotny-Diermayr; Kee Chuan Goh; Meredith Williams; Yong Cheng Tan; Lai Chun Ong; Albert Cheong; Bee Kheng Ng; Chithra Amalini; Babita Madan; Harish Nagaraj; Ramesh Jayaraman; Khalis M Pasha; Kantharaj Ethirajulu; Wee Joo Chng; Nurulhuda Mustafa; Boon Cher Goh; Cyril H. Benes; Ultan McDermott; Mathew J. Garnett; Brian W. Dymock; Jeanette Marjorie Wood

Dysregulation of the PI3K/mTOR pathway, either through amplifications, deletions, or as a direct result of mutations, has been closely linked to the development and progression of a wide range of cancers. Moreover, this pathway activation is a poor prognostic marker for many tumor types and confers resistance to various cancer therapies. Here, we describe VS-5584, a novel, low-molecular weight compound with equivalent potent activity against mTOR (IC50 = 37 nmol/L) and all class I phosphoinositide 3-kinase (PI3K) isoforms IC50: PI3Kα = 16 nmol/L; PI3Kβ = 68 nmol/L; PI3Kγ = 25 nmol/L; PI3Kδ = 42 nmol/L, without relevant activity on 400 lipid and protein kinases. VS-5584 shows robust modulation of cellular PI3K/mTOR pathways, inhibiting phosphorylation of substrates downstream of PI3K and mTORC1/2. A large human cancer cell line panel screen (436 lines) revealed broad antiproliferative sensitivity and that cells harboring mutations in PI3KCA are generally more sensitive toward VS-5584 treatment. VS-5584 exhibits favorable pharmacokinetic properties after oral dosing in mice and is well tolerated. VS-5584 induces long-lasting and dose-dependent inhibition of PI3K/mTOR signaling in tumor tissue, leading to tumor growth inhibition in various rapalog-sensitive and -resistant human xenograft models. Furthermore, VS-5584 is synergistic with an EGF receptor inhibitor in a gastric tumor model. The unique selectivity profile and favorable pharmacologic and pharmaceutical properties of VS-5584 and its efficacy in a wide range of human tumor models supports further investigations of VS-5584 in clinical trials. Mol Cancer Ther; 12(2); 151–61. ©2012 AACR.


Cancer Research | 2014

LEO1 Is Regulated by PRL-3 and Mediates Its Oncogenic Properties in Acute Myelogenous Leukemia

Phyllis Sy Chong; Jianbiao Zhou; Lip-Lee Cheong; Shaw-Cheng Liu; Jingru Qian; Tiannan Guo; Siu Kwan Sze; Qi Zeng; Wee Joo Chng

PRL-3, an oncogenic dual-specificity phosphatase, is overexpressed in 50% of acute myelogenous leukemia (AML) and associated with poor survival. We found that stable expression of PRL-3 confers cytokine independence and growth advantage of AML cells. However, how PRL-3 mediates these functions in AML is not known. To comprehensively screen for PRL3-regulated proteins in AML, we performed SILAC-based quantitative proteomics analysis and discovered 398 significantly perturbed proteins after PRL-3 overexpression. We show that Leo1, a component of RNA polymerase II-associated factor (PAF) complex, is a novel and important mediator of PRL-3 oncogenic activities in AML. We described a novel mechanism where elevated PRL-3 protein increases JMJD2C histone demethylase occupancy on Leo1 promoter, thereby reducing the H3K9me3 repressive signals and promoting Leo1 gene expression. Furthermore, PRL-3 and Leo1 levels were positively associated in AML patient samples (N=24; P<0.01). On the other hand, inhibition of Leo1 reverses PRL-3 oncogenic phenotypes in AML. Loss of Leo1 leads to destabilization of the PAF complex and downregulation of SOX2 and SOX4, potent oncogenes in myeloid transformation. In conclusion, we identify an important and novel mechanism by which PRL-3 mediates its oncogenic function in AML.


Bone Marrow Transplantation | 2016

Early relapse post autologous transplant is a stronger predictor of survival compared with pretreatment patient factors in the novel agent era: analysis of the Singapore Multiple Myeloma Working Group

Shin-Yeu Ong; S de Mel; Yunxin Chen; Melissa G. Ooi; Shilpa Surendran; Adeline Lin; Liang Piu Koh; Y C Linn; A Y L Ho; William Ying Khee Hwang; C Phipps; S M Y Loh; Y T Goh; Daryl Tan; Wee Joo Chng; Sathish Kumar Gopalakrishnan

The clinical outcome of multiple myeloma is heterogeneous. Both the depth of response to induction and transplant as well as early relapse within a year are correlated with survival, but it is unclear which factor is most relevant in Southeast Asian patients with multiple myeloma. We retrospectively analyzed outcomes of 215 patients who were treated with upfront autologous transplant in Singapore between 2000 and 2014. In patients who received novel agent (NA)-based induction, achieving only partial response (PR) post-induction was associated with poorer OS (HR 1.95, P=0.047) and PFS (HR 2.9, P<0.001), while achieving only PR post-transplant was strongly correlated with both OS (HR 3.3, P=0.001) and PFS (HR 7.6, P<0.001), compared with patients who achieved very good partial response (VGPR) or better. Early relapse was detected in 18% of all patients, although nearly half had initially achieved VGPR or better post-transplant. Early relapse after NA-based induction led to significantly shorter OS (median 22 months vs not reached, P<0.001), and was strongly associated with OS (HR 13.7, P<0.001). The impact of suboptimal post-transplant response and early relapse on survival may be more important than pretransplant factors, such as International Staging System or cytogenetics, and should be considered in risk stratification systems to rationalize therapy.


Oncology Letters | 2017

Single-cell genomic profiling of acute myeloid leukemia for clinical use: A pilot study.

Benedict Yan; Yongli Hu; Kenneth H. Ban; Zenia Tiang; Christopher Ng; Joanne Lee; Wilson Tan; Lily Chiu; Tin Wee Tan; Elaine Seah; Chin Hin Ng; Wee Joo Chng; Roger Foo

Although bulk high-throughput genomic profiling studies have led to a significant increase in the understanding of cancer biology, there is increasing awareness that bulk profiling approaches do not completely elucidate tumor heterogeneity. Single-cell genomic profiling enables the distinction of tumor heterogeneity, and may improve clinical diagnosis through the identification and characterization of putative subclonal populations. In the present study, the challenges associated with a single-cell genomics profiling workflow for clinical diagnostics were investigated. Single-cell RNA-sequencing (RNA-seq) was performed on 20 cells from an acute myeloid leukemia bone marrow sample. Putative blasts were identified based on their gene expression profiles and principal component analysis was performed to identify outlier cells. Variant calling was performed on the single-cell RNA-seq data. The present pilot study demonstrates a proof of concept for clinical single-cell genomic profiling. The recognized limitations include significant stochastic RNA loss and the relatively low throughput of the current proposed platform. Although the results of the present study are promising, further technological advances and protocol optimization are necessary for single-cell genomic profiling to be clinically viable.


Blood Cancer Journal | 2016

Thrombotic microangiopathy during carfilzomib use: case series in Singapore

Y Chen; M Ooi; S F Lim; A Lin; J Lee; C Nagarajan; Colin Phipps; Yuh Shan Lee; N F Grigoropoulos; Z Lao; S Surendran; E M Teh; Y T Goh; Wee Joo Chng; S K Gopalakrishnan

Carfilzomib is an irreversible proteasome inhibitor and is an effective treatment for multiple myeloma (MM).1, 2 It received US Food and Drug Administration approval based on a single-arm multicenter trial of carfilzomib monotherapy in 266 patients with relapsed MM following at least two prior lines of treatment including bortezomib and immunomodulators.1, 3 Safety analyses from four phase II carfilzomib trials (N=526) that included heavily pre-treated MM patients suggested a favorable risk-benefit profile with no specific signal of thrombotic microangiopathy (TMA) reported. However, serious adverse events of anemia, thrombocytopenia and increased serum creatinine comprised 1.3%, 1.1% and 1.3%, respectively.4 Over 350 subjects have been enrolled in randomized phase III trials in Asian countries. Herein, we report four cases of TMA related to carfilzomib use among 24 patients from 2 tertiary hospitals in Singapore (Table 1). All patients who started carfilzomib had a creatinine clearance >30 ml/min and platelet counts>50 × 109/l at the start of treatment.


bioRxiv | 2018

Recurrent PD-L1 Structural Rearrangements in Natural Killer/T Cell Lymphoma Patients with Complete Response to PD-1 Blockade Therapy

Jing Quan Lim; Tiffany Tang; Qingqing Cai; Daryl Tan; Maarja-Liisa Nairismagi; Yurike Laurensia; Burton K H Chia; Roujun Peng; Jabed Iqbal; Dachuan Huang; Tammy Song; Wan Lu Pang; Daryl Ming Zhe Cheah; Cedric Chuan Young Ng; Vikneswari Rajasegaran; Huangming Hong; Eric Tse; Benjamin Mow; Qi-Chun Cai; Limei Poon; Jing Tan; Nicholas Grigoropoulos; Yeow Tee Goh; Colin Phipps; Olaf Rotzschke; Chee Leong Cheng; Yuh Shan Lee; Yvonne Loh; Miriam Tao; Mohamad Farid

This study aims to identify recurrent genetic alterations in relapsed or refractory (RR) natural-killer/T-cell lymphoma (NKTL) patients who have achieved complete response (CR) with programmed cell death 1 (PD-1) blockade therapy. Seven of the eleven patients treated with pembrolizumab achieved CR while the remaining four had progressive disease (PD). Using whole genome sequencing (WGS), we found recurrent clonal structural rearrangements (SR) of the PD-L1 gene in four of the seven (57%) CR patients’ pretreated tumors. These PD-L1 SRs consist of inter-chromosomal translocations, tandem duplication and micro-inversion that disrupted the suppressive function of PD-L1 3’UTR. Interestingly, recurrent JAK3-activating (p.A573V) mutations were also validated in two CR patients’ tumors that did not harbor the PD-L1 SR. Importantly, these mutations were absent in the four PD cases. With immunohistochemistry (IHC), PD-L1 positivity could not discriminate patients who archived CR (range: 6%-100%) from patients who had PD (range: 35%-90%). PD-1 blockade with pembrolizumab is a potent strategy for RR NKTL patients and genomic screening could potentially accompany PD-L1 IHC positivity to better select patients for anti-PD-1 therapy.Abstract Natural killer/ T-cell lymphoma (NKTL) patients failing L-asparaginase regimens have extremely poor treatment outcomes. Previous case series showed promising activity when relapsed or refractory NKTL patients were treated with anti-programmed death 1 (PD1) inhibitors. Here, we continue to unravel the molecular profiles with whole-genome sequencing (WGS) on an extended cohort of 11 pembrolizumab-treated patients (median age at diagnosis, 42 years; range, 27-66 years) with a median follow-up of 11 months (range, 2 - 25 months) since starting anti-PD1 therapy. Seven patients achieved complete response (CR) and four patients had progressive disease (PD). Using WGS, we found structural rearrangements of the PD-L1 gene, JAK3-activating mutations and ARID1B homozygous insertion in four, two and one of the CR patients’ tumors, respectively. Interesting, these alterations, especially PD-L1 rearrangements, were absent in the four PD cases. Expression of PD1 ligand (PD-L1) was strong in nine patients (5 CR and 4 PD cases) and weak in two patients (both CR). PD1 blockade with pembrolizumab was a potent strategy for NKTL patients and genomic screening could potentially accompany PD-L1 immunohistochemical screening to better select patients for anti-PD1 therapy.

Collaboration


Dive into the Wee Joo Chng's collaboration.

Top Co-Authors

Avatar

Daryl Tan

Singapore General Hospital

View shared research outputs
Top Co-Authors

Avatar

Jianbiao Zhou

National University of Singapore

View shared research outputs
Top Co-Authors

Avatar

Siok Bian Ng

National University of Singapore

View shared research outputs
Top Co-Authors

Avatar

Yunxin Chen

Singapore General Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Brian G. M. Durie

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar

Babita Madan

National University of Singapore

View shared research outputs
Top Co-Authors

Avatar

Colin Phipps

Singapore General Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge