Network


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

Hotspot


Dive into the research topics where Wan-Teck Lim is active.

Publication


Featured researches published by Wan-Teck Lim.


Nature Medicine | 2012

A common BIM deletion polymorphism mediates intrinsic resistance and inferior responses to tyrosine kinase inhibitors in cancer

King Pan Ng; Axel M. Hillmer; Charles Chuah; Wen Chun Juan; Tun Kiat Ko; Audrey S.M. Teo; Pramila Ariyaratne; Naoto Takahashi; Kenichi Sawada; Yao Fei; Sheila Soh; Wah Heng Lee; John Huang; John Carson Allen; Xing Yi Woo; Niranjan Nagarajan; Vikrant Kumar; Anbupalam Thalamuthu; Wan Ting Poh; Ai Leen Ang; Hae Tha Mya; Gee Fung How; Li Yi Yang; Liang Piu Koh; Balram Chowbay; Chia-Tien Chang; Veera S. Nadarajan; Wee Joo Chng; Hein Than; Lay Cheng Lim

Tyrosine kinase inhibitors (TKIs) elicit high response rates among individuals with kinase-driven malignancies, including chronic myeloid leukemia (CML) and epidermal growth factor receptor–mutated non–small-cell lung cancer (EGFR NSCLC). However, the extent and duration of these responses are heterogeneous, suggesting the existence of genetic modifiers affecting an individuals response to TKIs. Using paired-end DNA sequencing, we discovered a common intronic deletion polymorphism in the gene encoding BCL2-like 11 (BIM). BIM is a pro-apoptotic member of the B-cell CLL/lymphoma 2 (BCL2) family of proteins, and its upregulation is required for TKIs to induce apoptosis in kinase-driven cancers. The polymorphism switched BIM splicing from exon 4 to exon 3, which resulted in expression of BIM isoforms lacking the pro-apoptotic BCL2-homology domain 3 (BH3). The polymorphism was sufficient to confer intrinsic TKI resistance in CML and EGFR NSCLC cell lines, but this resistance could be overcome with BH3-mimetic drugs. Notably, individuals with CML and EGFR NSCLC harboring the polymorphism experienced significantly inferior responses to TKIs than did individuals without the polymorphism (P = 0.02 for CML and P = 0.027 for EGFR NSCLC). Our results offer an explanation for the heterogeneity of TKI responses across individuals and suggest the possibility of personalizing therapy with BH3 mimetics to overcome BIM-polymorphism–associated TKI resistance.


Journal of Clinical Oncology | 2006

Never-Smokers With Lung Cancer: Epidemiologic Evidence of a Distinct Disease Entity

Chee-Keong Toh; Fei Gao; Wan-Teck Lim; Swan-Swan Leong; Kam-Weng Fong; Swee-Peng Yap; Anne A.L. Hsu; Philip Eng; Heng-Nung Koong; Agasthian Thirugnanam; Eng-Huat Tan

PURPOSE Tobacco smoke is a definite causative agent for lung cancer. It is increasingly being recognized that never-smokers can be afflicted with non-small-cell lung cancer (NSCLC). We aim to assess survival differences between smokers and never-smokers with NSCLC. PATIENTS AND METHODS We analyzed 975 NSCLC patients who presented from January 1999 to December 2002. Clinical characteristics among current-, former- and never-smokers were tested using chi2 or Kruskal-Wallis test. The hazard ratio (HR) for death and its 95% CI were calculated by Cox regression. RESULTS Of 975 patients, 59 had no smoking history and 33 had no quit time recorded. Of 883 patients analyzed, 286 patients (32.4%) were never-smokers. One hundred ninety-six never-smokers (68.5%) were females compared with 12% among current- and 13% among former-smokers (P < .001). There was a significant difference in histologic subtype between never-smokers and smokers: 69.9% with adenocarcinoma versus 39.9% (current-smokers) versus 47.3% (former-smokers); 5.9% with squamous cell carcinoma versus 35.7% (current-smokers) versus 28% (former-smokers; P < .001). Smokers had significantly poorer performance status (P = .002) and higher median age at diagnosis (P < .001) while more never-smokers presented with advanced disease (P = .002). Eight hundred and five patients (82.6%) died by May 30, 2005. The HR for smokers was significantly higher on both univariate and multivariate analysis (HR, 1.297; 95% CI, 1.040 to 1.619). CONCLUSION Never-smokers comprised a high proportion of NSCLC patients in Singapore. Definite epidemiologic differences exist between never-smokers and smokers. Differences in survival outcome further suggest that the biology underlying the pathogenesis and behavior of the disease may be different for never-smokers.


Biosensors and Bioelectronics | 2010

Versatile label free biochip for the detection of circulating tumor cells from peripheral blood in cancer patients

Swee Jin Tan; Rumkumar Lalitha Lakshmi; Pengfei Chen; Wan-Teck Lim; Levent Yobas; Chwee Teck Lim

The isolation of circulating tumor cells (CTCs) using microfluidics is attractive as the flow conditions can be accurately manipulated to achieve an efficient separation. CTCs are rare events within the peripheral blood of metastatic cancer patients which makes them hard to detect. The presence of CTCs is likely to indicate the severity of the disease and increasing evidences show its use for prognostic and treatment monitoring purposes. We demonstrated an effective separation using a microfluidic device to utilize the unique differences in size and deformability of cancer cells to blood cells. Using physical structures placed in the path of blood specimens in a microchannel, CTCs which are generally larger and stiffer are retained while most blood constituents are removed. The placements of the structures are optimized by computational analysis to enhance the isolation efficiency. With blood specimens from metastatic lung cancer patients, we confirmed the successful detection of CTCs. The operations for processing blood are straightforward and permit multiplexing of the microdevices to concurrently work with different samples. The microfluidic device is optically transparent which makes it simple to be integrated to existing laboratory microscopes and immunofluorescence staining can be done in situ to distinguish cancer cells from hematopoietic cells. This also minimizes the use of expensive staining reagents, given the small size of the microdevice. Identification of CTCs will aid in the detection of malignancy and disease stage as well as understanding the phenotypic and genotypic expressions of cancer cells.


Annals of Oncology | 2010

Phase I pharmacokinetic study of a weekly liposomal paclitaxel formulation (Genexol®-PM) in patients with solid tumors

Wan-Teck Lim; Eng-Huat Tan; Chee-Keong Toh; Siew Wan Hee; Swan-Swan Leong; P. C. S. Ang; N. S. Wong; Balram Chowbay

BACKGROUND The aim of this study was to determine the maximum tolerated dose (MTD) and the pharmacokinetic profile of Genexol-PM in Asian cancer patients. MATERIALS AND METHODS Patients (N = 24) refractory to previous chemotherapy received Genexol-PM as an 1-h infusion on a weekly basis for 3 weeks followed by a resting week. The starting dose was 80 mg/m(2) and the maximum administered dose was 200 mg/m(2). RESULTS The majority of patients had lung, nasopharyngeal and breast cancers and in eleven patients (46%), taxane-based chemotherapy had previously failed. The MTD was defined at 180 mg/m(2). The most common grade 3 non-hematologic adverse events in cycle 1 were fatigue (4%) and neuropathy (4%) occurring mainly at 200 mg/m(2). Five (21%) patients had partial response, nine (38%) had stable disease and seven (29%) had disease progression. Five of 11 previously taxane-refractory patients showed clinical benefit to Genexol-PM. The pharmacokinetics of Genexol-PM displayed dose-proportionality, with both the maximum concentration (C(max)) and the area under the concentration-time curve from zero to infinity (AUC(0-infinity)) increasing by approximately four- and threefold, respectively, as the dose of Genexol-PM was escalated from 80 to 200 mg/m(2). The median total-body clearance of Genexol-PM for all patients was 43.9 l/h. CONCLUSION The weekly regimen of Genexol-PM was well tolerated and responses were observed in patients with refractory tumors, including patients who had failed taxane-based chemotherapy previously.


PLOS ONE | 2014

Clinical Validation of an Ultra High-Throughput Spiral Microfluidics for the Detection and Enrichment of Viable Circulating Tumor Cells

Bee Luan Khoo; Majid Ebrahimi Warkiani; Daniel Shao-Weng Tan; Ali Asgar S. Bhagat; Darryl Irwin; Dawn Pingxi Lau; Alvin S. T. Lim; Kiat Hon Lim; Sai Sakktee Krisna; Wan-Teck Lim; Yoon Sim Yap; Soo-Chin Lee; Ross A. Soo; Jongyoon Han; Chwee Teck Lim

Background Circulating tumor cells (CTCs) are cancer cells that can be isolated via liquid biopsy from blood and can be phenotypically and genetically characterized to provide critical information for guiding cancer treatment. Current analysis of CTCs is hindered by the throughput, selectivity and specificity of devices or assays used in CTC detection and isolation. Methodology/Principal Findings Here, we enriched and characterized putative CTCs from blood samples of patients with both advanced stage metastatic breast and lung cancers using a novel multiplexed spiral microfluidic chip. This system detected putative CTCs under high sensitivity (100%, n = 56) (Breast cancer samples: 12–1275 CTCs/ml; Lung cancer samples: 10–1535 CTCs/ml) rapidly from clinically relevant blood volumes (7.5 ml under 5 min). Blood samples were completely separated into plasma, CTCs and PBMCs components and each fraction were characterized with immunophenotyping (Pan-cytokeratin/CD45, CD44/CD24, EpCAM), fluorescence in-situ hybridization (FISH) (EML4-ALK) or targeted somatic mutation analysis. We used an ultra-sensitive mass spectrometry based system to highlight the presence of an EGFR-activating mutation in both isolated CTCs and plasma cell-free DNA (cf-DNA), and demonstrate concordance with the original tumor-biopsy samples. Conclusions/Significance We have clinically validated our multiplexed microfluidic chip for the ultra high-throughput, low-cost and label-free enrichment of CTCs. Retrieved cells were unlabeled and viable, enabling potential propagation and real-time downstream analysis using next generation sequencing (NGS) or proteomic analysis.


Journal of Clinical Oncology | 2017

Evaluation of BGJ398, a Fibroblast Growth Factor Receptor 1-3 Kinase Inhibitor, in Patients With Advanced Solid Tumors Harboring Genetic Alterations in Fibroblast Growth Factor Receptors: Results of a Global Phase I, Dose-Escalation and Dose-Expansion Study

Lucia Nogova; Lecia V. Sequist; Jose Manuel Perez Garcia; Fabrice Andre; Jean-Pierre Delord; Manuel Hidalgo; Jan H. M. Schellens; Philippe Cassier; D. Ross Camidge; Martin Schuler; Ulka N. Vaishampayan; Howard A. Burris; G. Gary Tian; Mario Campone; Zev A. Wainberg; Wan-Teck Lim; Patricia LoRusso; Geoffrey I. Shapiro; Katie Parker; Xueying Chen; Somesh Choudhury; Francois Ringeisen; Diana Graus-Porta; Dale Porter; Randi Isaacs; Reinhard Buettner; Jürgen Wolf

Purpose This two-part, first-in-human study was initiated in patients with advanced solid tumors harboring genetic alterations in fibroblast growth factor receptors (FGFRs) to determine the maximum tolerated dose (MTD), the recommended phase II dose (RP2D), and the schedule, safety, pharmacokinetics, pharmacodynamics, and antitumor activity of oral BGJ398, a selective FGFR1-3 tyrosine kinase inhibitor. Patients and Methods Adult patients were treated with escalating dosages of BGJ398 5 to 150 mg once daily or 50 mg twice daily continuously in 28-day cycles. During expansion at the MTD, patients with FGFR1-amplified squamous cell non-small-cell lung cancer (sqNSCLC; arm 1) or other solid tumors with FGFR genetic alterations (mutations/amplifications/fusions) received BGJ398 daily on a continuous schedule (arm 2), or on a 3-weeks-on/1-week-off schedule (arm 3). Results Data in 132 patients from the escalation and expansion arms are reported (May 15, 2015, cutoff). The MTD, 125 mg daily, was determined on the basis of dose-limiting toxicities in four patients (100 mg, grade 3 aminotransferase elevations [n = 1]; 125 mg, hyperphosphatemia [n = 1]; 150 mg, grade 1 corneal toxicity [n = 1] and grade 3 aminotransferase elevations [n = 1]). Common adverse events in patients treated at the MTD (n = 57) included hyperphosphatemia (82.5%), constipation (50.9%), decreased appetite (45.6%), and stomatitis (45.6%). A similar safety profile was observed using the 3-weeks-on/1-week-off schedule (RP2D). However, adverse event-related dose adjustments/interruptions were less frequent with the 3-weeks-on/1-week-off (50.0%) versus the continuous (73.7%) schedule. Antitumor activity (seven partial responses [six confirmed]) was demonstrated with BGJ398 doses ≥ 100 mg in patients with FGFR1-amplified sqNSCLC and FGFR3-mutant bladder/urothelial cancer. Conclusion BGJ398 at the MTD/RP2D had a tolerable and manageable safety profile and showed antitumor activity in several tumor types, including FGFR1-amplified sqNSCLC and FGFR3-mutant bladder/urothelial cancers.


Molecular Therapy | 2014

Adoptive T-cell Transfer and Chemotherapy in the First-line Treatment of Metastatic and/or Locally Recurrent Nasopharyngeal Carcinoma

Whay-Kuang Chia; Marissa Teo; Who-Whong Wang; Bernett Lee; Soo-Fan Ang; Wai‐Meng Tai; Chit-Lai Chee; Joanna Ng; Rebecca Kan; Wan-Teck Lim; Sze-Huey Tan; Whee-Sze Ong; Yin Bun Cheung; Eng-Huat Tan; John Connolly; Stephen Gottschalk; Han-Chong Toh

The outcomes for patients with metastatic or locally recurrent Epstein–Barr virus (EBV)-positive nasopharyngeal carcinoma (NPC) remain poor. Adoptive immunotherapy with EBV-specific cytotoxic T lymphocytes (EBV-CTLs) has proven clinical efficacy, but it has never been evaluated in the first-line treatment setting in combination with chemotherapy. To evaluate the safety and efficacy of a chemotherapy in combination with adoptive EBV-CTL transfer, we conducted a phase 2 clinical trial consisting of four cycles of gemcitabine and carboplatin (GC) followed by up to six doses of EBV-CTL. Thirty-eight patients were enrolled, and 35 received GC and EBV-CTL. GC-CTL therapy resulted in a response rate of 71.4% with 3 complete responses and 22 partial responses. With a median follow up of 29.9 months, the 2-year and 3-year overall survival (OS) rate was 62.9 and 37.1%, respectively. Five patients did not require further chemotherapy for more than 34 months since initiation of CTL. Infusion of CTL products containing T cells specific for LMP2 positively correlated with OS (hazard ratio: 0.35; 95% confidence interval: 0.14–0.84; P = 0.014). Our study achieved one of the best survival outcomes in patients with advanced NPC, setting the stage for a future randomized study of chemotherapy with and without EBV-CTL.


Journal of Clinical Pathology | 2006

Lung cancer in never-smokers

Chee-Keong Toh; Wan-Teck Lim

The mystery behind never-smokers being more prone to lung cancer is unlocked with regard to smoking status and sex Never-smokers with lung cancer constitute an understudied and under-represented subset of patients. Although there have been hints that never-smokers can be afflicted with lung cancer,1 especially among Asian Chinese women,2 it is only recently that attention has turned towards this much ignored group of patients. Sparking the attention was the finding that somatic mutations of the epidermal growth factor receptor (EGFR) were consistently more common among lung tumours of never-smokers,3 and that these mutations could possibly explain the higher response rates to single agent gefitinib.4,5 The attention was further intensified by media reports of a non-smoking wife of a celebrity who developed lung cancer. Most studies on never-smokers with lung cancer have emerged from Asia, as the smoking prevalence rates in Asia are lower compared with the West. In Singapore, where the population is predominantly Chinese, the smoking prevalence in the general population is 24.3% in men and 3.6% in women.6 About 10–15% of lung cancers occur in a lifetime among never-smokers in the West,7 whereas about 30–40% of patients with lung cancer are never-smokers among the Asian countries.8 Whether this represents a higher risk of lung cancer among never-smokers in Asia or is a mere reflection of the higher numbers of never-smokers at risk is unclear at present. A recent large prospective study by Thun et al 9 may help to put things in perspective. The study among African Americans and Whites provides estimates of mortality due to lung cancer among never-smokers, with rates of 17.1 and 14.7 per 100 000 person-years among men and women, respectively. These figures highlight that the burden of lung cancer among never-smokers is fairly significant among …


Annals of Oncology | 2012

A phase II study evaluating the safety and efficacy of an adenovirus-ΔLMP1-LMP2 transduced dendritic cell vaccine in patients with advanced metastatic nasopharyngeal carcinoma

W. K. Chia; W.-W. Wang; M. Teo; W. M. Tai; Wan-Teck Lim; Eng-Huat Tan; Swan-Swan Leong; Li Sun; J. J. Chen; Stephen Gottschalk; H. C. Toh

BACKGROUND Individuals with metastatic Epstein-Barr virus (EBV)-positive nasopharyngeal carcinoma (NPC) continue to have poor outcomes. To evaluate the ability of a dendritic cell (DC) vaccine to target subdominant EBV antigens LMP1 and LMP2 expressed by NPC cells, we vaccinated patients using autologous DCs transduced with an adenovirus encoding a truncated LMP1 (ΔLMP1) and full-length LMP2 (Ad-ΔLMP1-LMP2). MATERIALS AND METHODS Sixteen subjects with metastatic NPC received Ad-ΔLMP1-LMP2 DC vaccines i.d. biweekly for up to five doses. Toxicity, immune responses and clinical responses were determined. RESULTS Most patients had extensive disease, with a median of three visceral sites of involvement (range 1-7). No significant toxicity was observed. Ad-ΔLMP1-LMP2 DCs induced delayed type hypersensitivity responses in 9 out of 12 patients, but although these DCs activated LMP1/2-specific T cells in vitro, no such increase in the frequency of peripheral LMP1/2-specific T cells was detected. Three patients had clinical responses including one with partial response (for 7½ months) and two with stable disease (for 6½ and 7½ months). CONCLUSIONS Ad-ΔLMP1-LMP2 transduced DCs can be successfully generated and safely administered to patients with advanced NPC. Since efficacy was limited, future studies should focus on DC vaccines with greater potency administered to subjects with less tumor burden.


Scientific Reports | 2016

Microfluidic enrichment for the single cell analysis of circulating tumor cells

Trifanny Yeo; Swee Jin Tan; Chew Leng Lim; Dawn Pingxi Lau; Yong Wei Chua; Sai Sakktee Krisna; Gopal Iyer; Gek San Tan; Tony Kiat Hon Lim; Daniel S.W. Tan; Wan-Teck Lim; Chwee Teck Lim

Resistance to drug therapy is a major concern in cancer treatment. To probe clones resistant to chemotherapy, the current approach is to conduct pooled cell analysis. However, this can yield false negative outcomes, especially when we are analyzing a rare number of circulating tumor cells (CTCs) among an abundance of other cell types. Here, we develop a microfluidic device that is able to perform high throughput, selective picking and isolation of single CTC to 100% purity from a larger population of other cells. This microfluidic device can effectively separate the very rare CTCs from blood samples from as few as 1 in 20,000 white blood cells. We first demonstrate isolation of pure tumor cells from a mixed population and track variations of acquired T790M mutations before and after drug treatment using a model PC9 cell line. With clinical CTC samples, we then show that the isolated single CTCs are representative of dominant EGFR mutations such as T790M and L858R found in the primary tumor. With this single cell recovery device, we can potentially implement personalized treatment not only through detecting genetic aberrations at the single cell level, but also through tracking such changes during an anticancer therapy.

Collaboration


Dive into the Wan-Teck Lim's collaboration.

Top Co-Authors

Avatar

Chee-Keong Toh

National University of Singapore

View shared research outputs
Top Co-Authors

Avatar

Swan-Swan Leong

National University of Singapore

View shared research outputs
Top Co-Authors

Avatar

Angela Takano

Singapore General Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chwee Teck Lim

National University of Singapore

View shared research outputs
Top Co-Authors

Avatar

Balram Chowbay

Howard Hughes Medical Institute

View shared research outputs
Top Co-Authors

Avatar

Kiat Hon Lim

Singapore General Hospital

View shared research outputs
Top Co-Authors

Avatar

Alvin S. T. Lim

Singapore General Hospital

View shared research outputs
Top Co-Authors

Avatar

Balram Chowbay

Howard Hughes Medical Institute

View shared research outputs
Top Co-Authors

Avatar

Ravindran Kanesvaran

National University of Singapore

View shared research outputs
Researchain Logo
Decentralizing Knowledge