Network


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

Hotspot


Dive into the research topics where Bee Luan Khoo is active.

Publication


Featured researches published by Bee Luan Khoo.


Scientific Reports | 2013

Isolation and retrieval of circulating tumor cells using centrifugal forces

Han Wei Hou; Majid Ebrahimi Warkiani; Bee Luan Khoo; Zi Rui Li; Ross A. Soo; Daniel Shao-Weng Tan; Wan Teck Lim; Jongyoon Han; Ali Asgar S. Bhagat; Chwee Teck Lim

Presence and frequency of rare circulating tumor cells (CTCs) in bloodstreams of cancer patients are pivotal to early cancer detection and treatment monitoring. Here, we use a spiral microchannel with inherent centrifugal forces for continuous, size-based separation of CTCs from blood (Dean Flow Fractionation (DFF)) which facilitates easy coupling with conventional downstream biological assays. Device performance was optimized using cancer cell lines (> 85% recovery), followed by clinical validation with positive CTCs enumeration in all samples from patients with metastatic lung cancer (n = 20; 5–88 CTCs per mL). The presence of CD133+ cells, a phenotypic marker characteristic of stem-like behavior in lung cancer cells was also identified in the isolated subpopulation of CTCs. The spiral biochip identifies and addresses key challenges of the next generation CTCs isolation assay including antibody independent isolation, high sensitivity and throughput (3 mL/hr); and single-step retrieval of viable CTCs.


Nature Protocols | 2016

Ultra-fast, label-free isolation of circulating tumor cells from blood using spiral microfluidics

Majid Ebrahimi Warkiani; Bee Luan Khoo; Lidan Wu; Andy Tay; Ali Asgar S. Bhagat; Jongyoon Han; Chwee Teck Lim

Circulating tumor cells (CTCs) are rare cancer cells that are shed from primary or metastatic tumors into the peripheral blood circulation. Phenotypic and genetic characterization of these rare cells can provide important information to guide cancer staging and treatment, and thus further research into their characteristics and properties is an area of considerable interest. In this protocol, we describe detailed procedures for the production and use of a label-free spiral microfluidic device to allow size-based isolation of viable CTCs using hydrodynamic forces that are present in curvilinear microchannels. This spiral system enables us to achieve ≥85% recovery of spiked cells across multiple cancer cell lines and 99.99% depletion of white blood cells in whole blood. The described spiral microfluidic devices can be produced at an extremely low cost using standard microfabrication and soft lithography techniques (2–3 d), and they can be operated using two syringe pumps for lysed blood samples (7.5 ml in 12.5 min for a three-layered multiplexed chip). The fast processing time and the ability to collect CTCs from a large patient blood volume allows this technique to be used experimentally in a broad range of potential genomic and transcriptomic applications.


ACS Nano | 2013

Isoporous Micro/Nanoengineered Membranes

Majid Ebrahimi Warkiani; Ali Asgar S. Bhagat; Bee Luan Khoo; Jongyoon Han; Chwee Teck Lim; Hai Qing Gong; Anthony G. Fane

Isoporous membranes are versatile structures with numerous potential and realized applications in various fields of science such as micro/nanofiltration, cell separation and harvesting, controlled drug delivery, optics, gas separation, and chromatography. Recent advances in micro/nanofabrication techniques and material synthesis provide novel methods toward controlling the detailed microstructure of membrane materials, allowing fabrication of membranes with well-defined pore size and shape. This review summarizes the current state-of-the-art for isoporous membrane fabrication using different techniques, including microfabrication, anodization, and advanced material synthesis. Various applications of isoporous membranes, such as protein filtration, pathogen isolation, cell harvesting, biosensing, and drug delivery, are also presented.


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.


Science Advances | 2016

Liquid biopsy and therapeutic response: Circulating tumor cell cultures for evaluation of anticancer treatment

Bee Luan Khoo; Gianluca Grenci; Tengyang Jing; Ying Bena Lim; Soo-Chin Lee; Jean Paul Thiery; Jongyoon Han; Chwee Teck Lim

A patient-derived CTC culture assay for anticancer drug evaluation to guide therapy for personalized treatment. The lack of a robust anticancer drug screening system to monitor patients during treatment delays realization of personalized treatment. We demonstrate an efficient approach to evaluate drug response using patient-derived circulating tumor cell (CTC) cultures obtained from liquid biopsy. Custom microfabricated tapered microwells were integrated with microfluidics to allow robust formation of CTC clusters without pre-enrichment and subsequent drug screening in situ. Rapid feedback after 2 weeks promotes immediate intervention upon detection of drug resistance or tolerance. The procedure was clinically validated with blood samples (n = 73) from 55 patients with early-stage, newly diagnosed, locally advanced, or refractory metastatic breast cancer. Twenty-four of these samples were used for drug evaluation. Cluster formation potential correlated inversely with increased drug concentration and therapeutic treatment. This new and robust liquid biopsy technique can potentially evaluate patient prognosis with CTC clusters during treatment and provide a noninvasive and inexpensive assessment that can guide drug discovery development or therapeutic choices for personalized treatment.


International Journal of Cancer | 2016

Single-cell profiling approaches to probing tumor heterogeneity.

Bee Luan Khoo; Parthiv Kant Chaudhuri; Naveen Ramalingam; Daniel Shao Weng Tan; Chwee Teck Lim; Majid Ebrahimi Warkiani

Tumor heterogeneity is a major hindrance in cancer classification, diagnosis and treatment. Recent technological advances have begun to reveal the true extent of its heterogeneity. Single‐cell analysis (SCA) is emerging as an important approach to detect variations in morphology, genetic or proteomic expression. In this review, we revisit the issue of inter‐ and intra‐tumor heterogeneity, and list various modes of SCA techniques (cell‐based, nucleic acid‐based, protein‐based, metabolite‐based and lipid‐based) presently used for cancer characterization. We further discuss the advantages of SCA over pooled cell analysis, as well as the limitations of conventional techniques. Emerging trends, such as high‐throughput sequencing, are also mentioned as improved means for cancer profiling. Collectively, these applications have the potential for breakthroughs in cancer treatment.


Biomicrofluidics | 2015

Hybrid capillary-inserted microfluidic device for sheathless particle focusing and separation in viscoelastic flow

Jeonghun Nam; Justin Kok Soon Tan; Bee Luan Khoo; Bumseok Namgung; Hwa Liang Leo; Chwee Teck Lim; Sangho Kim

A novel microfluidic device which consists of two stages for particle focusing and separation using a viscoelastic fluid has been developed. A circular capillary tube was used for three-dimensional particle pre-alignment before the separation process, which was inserted in a polydimethylsiloxane microchannel. Particles with diameters of 5 and 10 μm were focused at the centerline in the capillary tube, and the location of particles was initialized at the first bifurcation. Then, 5 and 10 μm particles were successfully separated in the expansion region based on size-dependent lateral migration, with ∼99% separation efficiency. The proposed device was further applied to separation of MCF-7 cells from leukocytes. Based on the cell size distribution, an approximate size cutoff for separation was determined to be 16 μm. At 200 μl/min, 94% of MCF-7 cells were separated with the purity of ∼97%. According to the trypan blue exclusion assay, high viability (∼90%) could be achieved for the separated MCF-7 cells. The use of a commercially available capillary tube enables the device to be highly versatile in dealing with particles in a wide size range by using capillary tubes with different inner diameters.


Nature Protocols | 2017

Expansion of patient-derived circulating tumor cells from liquid biopsies using a CTC microfluidic culture device

Bee Luan Khoo; Gianluca Grenci; Ying Bena Lim; Soo-Chin Lee; Jongyoon Han; Chwee Teck Lim

The development of personalized cancer therapy depends on a robust system to monitor the patients individual response to anticancer treatment. Anticancer drug efficacy has been tested on circulating tumor cells (CTCs) derived from patient blood samples after ex vivo expansion into CTC clusters. Current attempts to culture these primary cancer cells focus on long-term maintenance under growth factor supplements into cell lines, which usually takes >6 months and results in a CTC expansion efficiency of <20%. We recently developed a simple but unique microfluidics-based culture approach that requires minimal preprocessing (∼30 min) and does not require prior enrichment of CTCs or depend on the use of growth factor supplements. The approach capitalizes on co-culture of immune cells from the same patient blood sample within specially designed microwells that promote CTC cluster formation within 2 weeks, with an overall cluster formation success rate of ∼50%. Drug screening is facilitated by the incorporation of a gradient generator for parallel exposure to two or more drugs at various concentrations. Owing to the cost-effectiveness and less-invasive nature of this procedure, routine monitoring of disease progression can be achieved. The described microfluidics system can be operated with a single syringe pump to introduce drug compounds (which takes ∼6 min), followed by incubation of the CTC clusters for 48 h before analysis. In addition to its applications in biomedical research, the rapid readout of our platform will enable clinicians to assess or predict a patients response to various therapeutic strategies, so as to enable personalized or precision therapy.


Analytical Chemistry | 2018

Characterizing Deformability and Electrical Impedance of Cancer Cells in a Microfluidic Device

Ying Zhou; Dahou Yang; Yinning Zhou; Bee Luan Khoo; Jongyoon Han; Ye Ai

Mechanical properties of cells, reflective of various biochemical characteristics such as gene expression and cytoskeleton, are promising label-free biomarkers for studying and characterizing cells. Electrical properties of cells, dependent on the cellular structure and content, are also label-free indicators of cell states and phenotypes. In this work, we have developed a microfluidic device that is able to simultaneously characterize the mechanical and electrical properties of individual biological cells in a high-throughput manner (>1000 cells/min). The deformability of MCF-7 breast cancer cells was characterized based on the passage time required for an individual cell to pass through a constriction smaller than the cell size. The total passage time can be divided into two components: the entry time required for a cell to deform and enter a constriction, which is dominated by the deformability of cells, and the transit time required for the fully deformed cell to travel inside the constriction, which mainly relies on the surface friction between cells and the channel wall. The two time durations for individual cells to pass through the entry region and transit region have both been investigated. In addition, undeformed cells and fully deformed cells were simultaneously characterized via electrical impedance spectroscopy technique. The combination of mechanical and electrical properties serves as a unique set of intrinsic cellular biomarkers for single-cell analysis, providing better differentiation of cellular phenotypes, which are not easily discernible via single-marker analysis.


Archive | 2014

Ultra-High Throughput Enrichment of Viable Circulating Tumor Cells

Bee Luan Khoo; Majid Ebrahimi Warkiani; Guofeng Guan; Daniel Shao-Weng Tan; Alvin S. T. Lim; Wan-Teck Lim; Yoon Sim Yap; Soo-Chin Lee; Ross A. Soo; Jongyoon Han; Chwee Teck Lim

Detection, enumeration and characterization of rare circulating tumor cells (CTCs) from the peripheral blood of cancer patients potentially provide critical insights into tumor biology and is promising for cancer diagnosis and prognosis. Here, we present a novel multiplexed spiral microfluidic device for ultra-high throughput, label-free enrichment of CTCs from clinically relevant blood volumes. The fast processing time of the technique (7.5 mL blood in < 5 min) and high sensitivity of the device lends itself to a broad range of potential genomic and transcriptomic applications. The method can specifically separate and preserve all fractions of blood (i.e., plasma, CTCs and PBMC) for diverse downstream analysis. CTCs were detected from 100% (10/10) of blood samples collected from patients with advanced stage metastatic breast (12-56 CTC/ml) or lung cancer (30-153 CTC/ml). Cancer cells were characterized with immunostaining and fluorescence in situ hybridization (FISH) (HER2/neu). Retrieved cells were unlabelled and hence more viable for propagation and other informative analysis such as the next generation sequencing (NGS) to guide treatment and individualized patient care.

Collaboration


Dive into the Bee Luan Khoo's collaboration.

Top Co-Authors

Avatar

Chwee Teck Lim

National University of Singapore

View shared research outputs
Top Co-Authors

Avatar

Jongyoon Han

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Jean Paul Thiery

National University of Singapore

View shared research outputs
Top Co-Authors

Avatar

Ross A. Soo

National University of Singapore

View shared research outputs
Top Co-Authors

Avatar

Wan-Teck Lim

National University of Singapore

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andy Tay

University of California

View shared research outputs
Top Co-Authors

Avatar

Yoon Sim Yap

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Alvin S. T. Lim

Singapore General Hospital

View shared research outputs
Top Co-Authors

Avatar

Gianluca Grenci

National University of Singapore

View shared research outputs
Researchain Logo
Decentralizing Knowledge