Network


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

Hotspot


Dive into the research topics where Shuang Hou is active.

Publication


Featured researches published by Shuang Hou.


Advanced Materials | 2013

Capture and Stimulated Release of Circulating Tumor Cells on Polymer‐Grafted Silicon Nanostructures

Shuang Hou; Haichao Zhao; Libo Zhao; Qinglin Shen; Kevin S. Wei; Daniel Y. Suh; Aiko Nakao; Mitch A. Garcia; Min Song; Thomas H. Lee; Bin Xiong; Shyh-Chyang Luo; Hsian-Rong Tseng; Hsiao-hua Yu

A platform for capture and release of circulating tumor cells is demonstrated by utilizing polymer grafted silicon nanowires. In this platform, integration of ligand-receptor recognition, nanostructure amplification, and thermal responsive polymers enables a highly efficient and selective capture of cancer cells. Subsequently, these captured cells are released upon a physical stimulation with outstanding cell viability.


Advanced Materials | 2013

Specific Capture and Release of Circulating Tumor Cells Using Aptamer‐Modified Nanosubstrates

Qinglin Shen; Li Xu; Libo Zhao; Dongxia Wu; Yunshan Fan; Yiliang Zhou; Wei-Han OuYang; Xiaochun Xu; Zhen Zhang; Min Song; Thomas H. Lee; Mitch A. Garcia; Bin Xiong; Shuang Hou; Hsian-Rong Tseng; Xiaohong Fang

Circulating tumor cells (CTCs)[1] are cancer cells that have propagated from tumors, spreading into the bloodstream as the cellular origin of fatal metastasis. Besides conventional diagnostic approaches (e.g., tumor biopsy, anatomical/molecular imaging and serum marker detection), detecting CTCs in peripheral blood is of prognostic value in different types of solid tumors, especially for predicting patient survival. The fact is that CTC detection have been technically challenging because of the extremely low abundance (a few to hundreds per mL) of CTCs among a high number (109 cells mL-1) of hematologic cells.[2] Over the past decade, a diversity of diagnostic technologies has been demonstrated for CTC detection using different working mechanisms. The current FDA-cleared CellSearch™ Assay is based on immunomagnetic separation of CTCs. Due to its unsatisfactory efficiency and high cost, researchers have been exploiting new technologies,[3] e.g., flow cytometry, size-based filtration systems and microfluidic devices that may offer improved sensitivity and reduced cost for CTC detection. In addition to the prognostic utility of CTC-based diagnostics, it is conceivable that the molecular signatures and functional readouts derived from CTCs will shed much valuable insight into tumor biology during the critical window where therapeutic intervention could make a significant difference.


Angewandte Chemie | 2013

Polymer Nanofiber‐Embedded Microchips for Detection, Isolation, and Molecular Analysis of Single Circulating Melanoma Cells

Shuang Hou; Libo Zhao; Qinglin Shen; Juehua Yu; Charles Ng; Xiangju Kong; Dongxia Wu; Min Song; Xiaohong Shi; Xiaochun Xu; Wei-Han OuYang; Rongxian He; Xingzhong Zhao; Thomas H. Lee; F. Charles Brunicardi; Mitch A. Garcia; Antoni Ribas; Roger S. Lo; Hsian-Rong Tseng

Circulating tumor cells (CTCs)[1] are cancer cells shed from either the primary tumors or metastatic sites. The presence and number of CTCs in peripheral blood can provide clinically significant data on prognosis and therapeutic response patterns, respectively[2]. Thus, as with traditional invasive tumor biopsies that enable gold-standard pathological analysis, CTCs can be regarded as “liquid biopsies” of the tumor, which enable repeated and relatively non-invasive characterization of tumor evolution, especially important during therapeutic interventions. Currently, FDA-cleared CellSearch™ Assay is costly and inefficient in capturing CTCs, and the enriched CTCs are typically contaminated with a large number of white blood cells (WBCs). As a result, the diagnostic value of CTCs has been underutilized. Over the past decade, a diversity of CTC detection technologies[2d, 3] have been developed to overcome the challenges encountered by the immunomagnetic separation-based CellSearch™ Assay.


Accounts of Chemical Research | 2014

Nanostructure embedded microchips for detection, isolation, and characterization of circulating tumor cells.

Millicent Lin; Jie-Fu Chen; Yi-Tsung Lu; Yang Zhang; Jinzhao Song; Shuang Hou; Zunfu Ke; Hsian-Rong Tseng

Conspectus Circulating tumor cells (CTCs) are cancer cells that break away from either a primary tumor or a metastatic site and circulate in the peripheral blood as the cellular origin of metastasis. With their role as a “tumor liquid biopsy”, CTCs provide convenient access to all disease sites, including that of the primary tumor and the site of fatal metastases. It is conceivable that detecting and analyzing CTCs will provide insightful information in assessing the disease status without the flaws and limitations encountered in performing conventional tumor biopsies. However, identifying CTCs in patient blood samples is technically challenging due to the extremely low abundance of CTCs among a large number of hematologic cells. To address this unmet need, there have been significant research endeavors, especially in the fields of chemistry, materials science, and bioengineering, devoted to developing CTC detection, isolation, and characterization technologies. Inspired by the nanoscale interactions observed in the tissue microenvironment, our research team at UCLA pioneered a unique concept of “NanoVelcro” cell-affinity substrates, in which CTC capture agent-coated nanostructured substrates were utilized to immobilize CTCs with high efficiency. The working mechanism of NanoVelcro cell-affinity substrates mimics that of Velcro: when the two fabric strips of a Velcro fastener are pressed together, tangling between the hairy surfaces on two strips leads to strong binding. Through continuous evolution, three generations (gens) of NanoVelcro CTC chips have been established to achieve different clinical utilities. The first-gen NanoVelcro chip, composed of a silicon nanowire substrate (SiNS) and an overlaid microfluidic chaotic mixer, was created for CTC enumeration. Side-by-side analytical validation studies using clinical blood samples suggested that the sensitivity of first-gen NanoVelcro chip outperforms that of FDA-approved CellSearch. In conjunction with the use of the laser microdissection (LMD) technique, second-gen NanoVelcro chips (i.e., NanoVelcro-LMD), based on polymer nanosubstrates, were developed for single-CTC isolation. The individually isolated CTCs can be subjected to single-CTC genotyping (e.g., Sanger sequencing and next-generation sequencing, NGS) to verify the CTC’s role as tumor liquid biopsy. Created by grafting of thermoresponsive polymer brushes onto SiNS, third-gen NanoVelcro chips (i.e., Thermoresponsive NanoVelcro) have demonstrated the capture and release of CTCs at 37 and 4 °C, respectively. The temperature-dependent conformational changes of polymer brushes can effectively alter the accessibility of the capture agent on SiNS, allowing for rapid CTC purification with desired viability and molecular integrity. This Account summarizes the continuous evolution of NanoVelcro CTC assays from the emergence of the original idea all the way to their applications in cancer research. We envision that NanoVelcro CTC assays will lead the way for powerful and cost-efficient diagnostic platforms for researchers to better understand underlying disease mechanisms and for physicians to monitor real-time disease progression.


Advanced Materials | 2013

High‐Purity Prostate Circulating Tumor Cell Isolation by a Polymer Nanofiber‐Embedded Microchip for Whole Exome Sequencing

Libo Zhao; Yi-Tsung Lu; Fuqiang Li; Kui Wu; Shuang Hou; Juehua Yu; Qinglin Shen; Dongxia Wu; Min Song; Wei-Han OuYang; Zheng Luo; Thomas H. Lee; Xiaohong Fang; Chen Shao; Xun Xu; Mitch A. Garcia; Leland W.K. Chung; Matthew Rettig; Hsian-Rong Tseng; Edwin M. Posadas

Handpick single cancer cells: a modified NanoVelcro Chip is coupled with ArcturusXT laser capture microdissection (LCM) technology to enable the detection and isolation of single circulating tumor cells (CTCs) from patients with prostate cancer (PC). This new approach paves the way for conducting next-generation sequencing (NGS) on single CTCs.


Cancer Research | 2010

A Microfluidic Platform for Systems Pathology: Multiparameter Single-Cell Signaling Measurements of Clinical Brain Tumor Specimens

Jing Sun; Michael Masterman-Smith; Nicholas A. J. Graham; Jing Jiao; Jack Mottahedeh; Dan R. Laks; Minori Ohashi; Jason DeJesus; Ken-ichiro Kamei; Ki-Bum Lee; Hao Wang; Yi-Tsung Lu; Shuang Hou; Keyu Li; Max Liu; Nangang Zhang; Shutao Wang; Brigitte Angénieux; Eric R. Samuels; Jun Park; Dirk Williams; Vera Konkankit; David Nathanson; R. Michael van Dam; Michael E. Phelps; Hong Wu; Linda M. Liau; Paul S. Mischel; Jorge A. Lazareff; Harley I. Kornblum

The clinical practice of oncology is being transformed by molecular diagnostics that will enable predictive and personalized medicine. Current technologies for quantitation of the cancer proteome are either qualitative (e.g., immunohistochemistry) or require large sample sizes (e.g., flow cytometry). Here, we report a microfluidic platform-microfluidic image cytometry (MIC)-capable of quantitative, single-cell proteomic analysis of multiple signaling molecules using only 1,000 to 2,800 cells. Using cultured cell lines, we show simultaneous measurement of four critical signaling proteins (EGFR, PTEN, phospho-Akt, and phospho-S6) within the oncogenic phosphoinositide 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) signaling pathway. To show the clinical application of the MIC platform to solid tumors, we analyzed a panel of 19 human brain tumor biopsies, including glioblastomas. Our MIC measurements were validated by clinical immunohistochemistry and confirmed the striking intertumoral and intratumoral heterogeneity characteristic of glioblastoma. To interpret the multiparameter, single-cell MIC measurements, we adapted bioinformatic methods including self-organizing maps that stratify patients into clusters that predict tumor progression and patient survival. Together with bioinformatic analysis, the MIC platform represents a robust, enabling in vitro molecular diagnostic technology for systems pathology analysis and personalized medicine.


Methods | 2013

NanoVelcro Chip for CTC enumeration in prostate cancer patients

Yi-Tsung Lu; Libo Zhao; Qinglin Shen; Mitch A. Garcia; Dongxia Wu; Shuang Hou; Min Song; Xiaochun Xu; Wei-Han OuYang; William W.-L. OuYang; Jake Lichterman; Zheng Luo; Xuan Xuan; Jiaoti Huang; Leland W.K. Chung; Matthew Rettig; Hsian-Rong Tseng; Chen Shao; Edwin M. Posadas

Circulating tumor cells (CTCs) are one of the most crucial topics in rare cell biology and have become the focus of a significant and emerging area of cancer research. While CTC enumeration is a valid biomarker in prostate cancer, the current FDA-approved CTC technology is unable to detect CTCs in a large portion of late stage prostate cancer patients. Here we introduce the NanoVelcro CTC Chip, a device composed of a patterned silicon nanowire substrate (SiNW) and an overlaid polydimethylsiloxane (PDMS) chaotic mixer. Validated by two institutions participating in the study, the NanoVelcro Chip assay exhibits very consistent efficiency in CTC-capture from patient samples. The utilized protocol can be easily replicated at different facilities. We demonstrate the clinical utility of the NanoVelcro Chip by performing serial enumerations of CTCs in prostate cancer patients after undergoing systemic therapy. Changes in CTC numbers after 4-10 weeks of therapy were compared with their clinical responses. We observed a statistically significant reduction in CTCs counts in the clinical responders. We performed long-term follow up with serial CTC collection and enumeration in one patient observing variations in counts correlating with treatment response. This study demonstrates the consistency of the NanoVelcro Chip assay over time for CTC enumeration and also shows that continuous monitoring of CTC numbers can be employed to follow responses to different treatments and monitor disease progression.


ACS Nano | 2015

Programming thermoresponsiveness of NanoVelcro substrates enables effective purification of circulating tumor cells in lung cancer patients.

Zunfu Ke; Millicent Lin; Jie-Fu Chen; Jin-sil Choi; Yang Zhang; Anna Fong; An-Jou Liang; Shang-Fu Chen; Qingyu Li; Wenfeng Fang; Pingshan Zhang; Mitch A. Garcia; Thomas H. Lee; Min Song; Hsing-An Lin; Haichao Zhao; Shyh-Chyang Luo; Shuang Hou; Hsiao-hua Yu; Hsian-Rong Tseng

Unlike tumor biopsies that can be constrained by problems such as sampling bias, circulating tumor cells (CTCs) are regarded as the “liquid biopsy” of the tumor, providing convenient access to all disease sites, including primary tumor and fatal metastases. Although enumerating CTCs is of prognostic significance in solid tumors, it is conceivable that performing molecular and functional analyses on CTCs will reveal much significant insight into tumor biology to guide proper therapeutic intervention. We developed the Thermoresponsive NanoVelcro CTC purification system that can be digitally programmed to achieve an optimal performance for purifying CTCs from non-small cell lung cancer (NSCLC) patients. The performance of this unique CTC purification system was optimized by systematically modulating surface chemistry, flow rates, and heating/cooling cycles. By applying a physiologically endurable stimulation (i.e., temperature between 4 and 37 °C), the mild operational parameters allow minimum disruption to CTCs’ viability and molecular integrity. Subsequently, we were able to successfully demonstrate culture expansion and mutational analysis of the CTCs purified by this CTC purification system. Most excitingly, we adopted the combined use of the Thermoresponsive NanoVelcro system with downstream mutational analysis to monitor the disease evolution of an index NSCLC patient, highlighting its translational value in managing NSCLC.


Biomaterials | 2012

The Therapeutic Efficacy of Camptothecin-Encapsulated Supramolecular Nanoparticles

Kuan Ju Chen; Li Tang; Mitch A. Garcia; Hao Wang; Hua Lu; Wei-Yu Lin; Shuang Hou; Qian Yin; Clifton Kwang-Fu Shen; Jianjun Cheng; Hsian-Rong Tseng

Nanomaterials have been increasingly employed as drug(s)-incorporated vectors for drug delivery due to their potential of maximizing therapeutic efficacy while minimizing systemic side effects. However, there have been two main challenges for these vectors: (i) the existing synthetic approaches are cumbersome and incapable of achieving precise control of their structural properties, which will affect their biodistribution and therapeutic efficacies, and (ii) lack of an early checkpoint to quickly predict which drug(s)-incorporated vectors exhibit optimal therapeutic outcomes. In this work, we utilized a new rational developmental approach to rapidly screen nanoparticle (NP)-based cancer therapeutic agents containing a built-in companion diagnostic utility for optimal therapeutic efficacy. The approach leverages the advantages of a self-assembly synthetic method for preparation of two different sizes of drug-incorporated supramolecular nanoparticles (SNPs), and a positron emission tomography (PET) imaging-based biodistribution study to quickly evaluate the accumulation of SNPs at a tumor site in vivo and select the favorable SNPs for in vivo therapeutic study. Finally, the enhanced in vivo anti-tumor efficacy of the selected SNPs was validated by tumor reduction/inhibition studies. We foresee our rational developmental approach providing a general strategy in the search of optimal therapeutic agents among the diversity of NP-based therapeutic agents.


Oncotarget | 2015

A comparison of isolated circulating tumor cells and tissue biopsies using whole-genome sequencing in prostate cancer

Runze Jiang; Yi-Tsung Lu; Hao Ho; Bo Li; Jie-Fu Chen; Millicent Lin; Fuqiang Li; Kui Wu; Hanjie Wu; Jake Lichterman; Haolei Wan; Chia-Lun Lu; William W.-L. OuYang; Ming Ni; Linlin Wang; Guibo Li; Thomas H. Lee; Xiuqing Zhang; Jonathan Yang; Matthew Rettig; Leland W.K. Chung; Huanming Yang; Ker-Chau Li; Yong Hou; Hsian-Rong Tseng; Shuang Hou; Xun Xu; Jun Wang; Edwin M. Posadas

Previous studies have demonstrated focal but limited molecular similarities between circulating tumor cells (CTCs) and biopsies using isolated genetic assays. We hypothesized that molecular similarity between CTCs and tissue exists at the single cell level when characterized by whole genome sequencing (WGS). By combining the NanoVelcro CTC Chip with laser capture microdissection (LCM), we developed a platform for single-CTC WGS. We performed this procedure on CTCs and tissue samples from a patient with advanced prostate cancer who had serial biopsies over the course of his clinical history. We achieved 30X depth and ≥ 95% coverage. Twenty-nine percent of the somatic single nucleotide variations (SSNVs) identified were founder mutations that were also identified in CTCs. In addition, 86% of the clonal mutations identified in CTCs could be traced back to either the primary or metastatic tumors. In this patient, we identified structural variations (SVs) including an intrachromosomal rearrangement in chr3 and an interchromosomal rearrangement between chr13 and chr15. These rearrangements were shared between tumor tissues and CTCs. At the same time, highly heterogeneous short structural variants were discovered in PTEN, RB1, and BRCA2 in all tumor and CTC samples. Using high-quality WGS on single-CTCs, we identified the shared genomic alterations between CTCs and tumor tissues. This approach yielded insight into the heterogeneity of the mutational landscape of SSNVs and SVs. It may be possible to use this approach to study heterogeneity and characterize the biological evolution of a cancer during the course of its natural history.

Collaboration


Dive into the Shuang Hou's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Min Song

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jie-Fu Chen

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Edwin M. Posadas

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar

Libo Zhao

University of California

View shared research outputs
Top Co-Authors

Avatar

Yi-Tsung Lu

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Leland W.K. Chung

Cedars-Sinai Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge