Network


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

Hotspot


Dive into the research topics where Jie-Fu Chen is active.

Publication


Featured researches published by Jie-Fu Chen.


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.


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.


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.


Cancer | 2015

Subclassification of prostate cancer circulating tumor cells by nuclear size reveals very small nuclear circulating tumor cells in patients with visceral metastases.

Jie-Fu Chen; Hao Ho; Jake Lichterman; Yi-Tsung Lu; Yang Zhang; Mitch A. Garcia; Shang-Fu Chen; An-Jou Liang; Elisabeth Hodara; Haiyen E. Zhau; Shuang Hou; Rafi S. Ahmed; Daniel Luthringer; Jiaoti Huang; Ker‐Chau Li; Leland W.K. Chung; Zunfu Ke; Hsian-Rong Tseng; Edwin M. Posadas

Although enumeration of circulating tumor cells (CTCs) has shown some clinical value, the pool of CTCs contains a mixture of cells that contains additional information that can be extracted. The authors subclassified CTCs by shape features focusing on nuclear size and related this with clinical information.


Theranostics | 2016

Clinical Applications of NanoVelcro Rare-Cell Assays for Detection and Characterization of Circulating Tumor Cells

Jie-Fu Chen; Yazhen Zhu; Yi-Tsung Lu; Elisabeth Hodara; Shuang Hou; Vatche G. Agopian; James S. Tomlinson; Edwin M. Posadas; Hsian-Rong Tseng

Liquid biopsy of tumor through isolation of circulating tumor cells (CTCs) allows non-invasive, repetitive, and systemic sampling of disease. Although detecting and enumerating CTCs is of prognostic significance in metastatic cancer, it is conceivable that performing molecular and functional characterization on CTCs will reveal unprecedented insight into the pathogenic mechanisms driving lethal disease. Nanomaterial-embedded cancer diagnostic platforms, i.e., NanoVelcro CTC Assays represent a unique rare-cell sorting method that enables detection isolation, and characterization of CTCs in peripheral blood, providing an opportunity to noninvasively monitor disease progression in individual cancer patients. Over the past decade, a series of NanoVelcro CTC Assays has been demonstrated for exploring the full potential of CTCs as a clinical biomarker, including CTC enumeration, phenotyping, genotyping and expression profiling. In this review article, the authors will briefly introduce the development of three generations of NanoVelcro CTC Assays, and highlight the clinical applications of each generation for various types of solid cancers, including prostate cancer, pancreatic cancer, lung cancer, and melanoma.


Oncotarget | 2016

Detecting ALK-rearrangement of CTC enriched by nanovelcro chip in advanced NSCLC patients

Weiling He; Di Xu; Zhuo Wang; Xianhong Xiang; Bing Tang; Shuhua Li; Minzhi Hou; Yang Zhang; Jie-Fu Chen; Millicent Lin; Liantang Wang; Shuang Hou; Hsian-Rong Tseng; Ming Kuang; Zunfu Ke

Circulating tumor cells (CTC) shows great prospect to realize precision medicine in cancer patients. We developed the NanoVelcro Chip integrating three functional mechanisms. NanoVelcro CTC capture efficiency was tested in advanced NSCLC. Further, ALK-rearrangement status was examined through fluorescent in situ hybridization in CTCs enriched by NanoVelcro. NanoVelcro system showed higher CTC-capture efficiency than CellSearch in advanced NSCLC. CTC counts obtained by both methods were positively correlated (r=0.45, p<0.05). Further, CTC counts enriched by NanoVelcro correlated to the pTNM stage but CellSearch. All ALK-positive patients had 3 or more ALK-rearranged CTC per mL of blood. Less than 3 ALK-rearranged CTC was detected in ALK-negative patients. NanoVelcro can detect the ALK-rearranged status with consistent sensitivity and specificity compared to biopsy test. Furthermore, the ALK-rearranged CTC ratio correlated to the pTNM stage in ALK-positive patients. Following up with CTC enumeration by NanoVelcro and CellSearch observed their variations in reflecting clinical response of crizotinib treatment. NanoVelcro provides a sensitive method for CTC counts and characterization in advanced NSCLC. ALK-rearrangement can be detected in CTCs collected from advanced NSCLC patients by NanoVelcro, facilitating diagnostic test and prognosis analysis, most importantly offering one noninvasive method for real-time monitoring of treatment reaction.


ACS Nano | 2017

Imprinted NanoVelcro Microchips for Isolation and Characterization of Circulating Fetal Trophoblasts: Toward Noninvasive Prenatal Diagnostics

Shuang Hou; Jie-Fu Chen; Min Song; Yazhen Zhu; Yu Jen Jan; Szu Hao Chen; Tzu-Hua Weng; Dean-An Ling; Shang-Fu Chen; Tracy Ro; An-Jou Liang; Thomas H. Lee; Helen Jin; Man Li; L. Liu; Yu-Sheng Hsiao; Peilin Chen; Hsiao-hua Yu; Ming-Song Tsai; Margareta D. Pisarska; Angela Chen; Li-Ching Chen; Hsian-Rong Tseng

Circulating fetal nucleated cells (CFNCs) in maternal blood offer an ideal source of fetal genomic DNA for noninvasive prenatal diagnostics (NIPD). We developed a class of nanoVelcro microchips to effectively enrich a subcategory of CFNCs, i.e., circulating trophoblasts (cTBs) from maternal blood, which can then be isolated with single-cell resolution by a laser capture microdissection (LCM) technique for downstream genetic testing. We first established a nanoimprinting fabrication process to prepare the LCM-compatible nanoVelcro substrates. Using an optimized cTB-capture condition and an immunocytochemistry protocol, we were able to identify and isolate single cTBs (Hoechst+/CK7+/HLA-G+/CD45-, 20 μm > sizes > 12 μm) on the imprinted nanoVelcro microchips. Three cTBs were polled to ensure reproducible whole genome amplification on the cTB-derived DNA, paving the way for cTB-based array comparative genomic hybridization (aCGH) and short tandem repeats analysis. Using maternal blood samples collected from expectant mothers carrying a single fetus, the cTB-derived aCGH data were able to detect fetal genders and chromosomal aberrations, which had been confirmed by standard clinical practice. Our results support the use of nanoVelcro microchips for cTB-based noninvasive prenatal genetic testing, which holds potential for further development toward future NIPD solution.


Advanced Drug Delivery Reviews | 2018

NanoVelcro rare-cell assays for detection and characterization of circulating tumor cells.

Yu Jen Jan; Jie-Fu Chen; Yazhen Zhu; Yi-Tsung Lu; Szu Hao Chen; Howard Chung; Matthew Smalley; Yen-Wen Huang; Jiantong Dong; Li-Ching Chen; Hsiao-hua Yu; James S. Tomlinson; Shuang Hou; Vatche G. Agopian; Edwin M. Posadas; Hsian-Rong Tseng

Abstract Circulating tumor cells (CTCs) are cancer cells shredded from either a primary tumor or a metastatic site and circulate in the blood as the potential cellular origin of metastasis. By detecting and analyzing CTCs, we will be able to noninvasively monitor disease progression in individual cancer patients and obtain insightful information for assessing disease status, thus realizing the concept of “tumor liquid biopsy”. However, it is technically challenging to identify CTCs in patient blood samples because of the extremely low abundance of CTCs among a large number of hematologic cells. In order to address this challenge, 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 remarkable efficiency. Four generations of NanoVelcro CTC assays have been developed over the past decade for a variety of clinical utilities. The 1st‐gen NanoVelcro Chips, composed of a silicon nanowire substrate (SiNS) and an overlaid microfluidic chaotic mixer, were created for CTC enumeration. The 2nd‐gen NanoVelcro Chips (i.e., NanoVelcro‐LMD), based on polymer nanosubstrates, were developed for single‐CTC isolation in conjunction with the use of the laser microdissection (LMD) technique. By grafting thermoresponsive polymer brushes onto SiNS, the 3rd‐gen Thermoresponsive NanoVelcro Chips have demonstrated the capture and release of CTCs at 37 and 4 °C respectively, thereby allowing for rapid CTC purification while maintaining cell viability and molecular integrity. Fabricated with boronic acid‐grafted conducting polymer‐based nanomaterial on chip surface, the 4th‐gen NanoVelcro Chips (Sweet chip) were able to purify CTCs with well‐preserved RNA transcripts, which could be used for downstream analysis of several cancer specific RNA biomarkers. In this review article, we will summarize the development of the four generations of NanoVelcro CTC assays, and the clinical applications of each generation of devices. Graphical abstract Figure. No Caption available.


Small | 2015

Supramolecular Nanosubstrate-Mediated Delivery for Reprogramming and Transdifferentiation of Mammalian Cells

Shuang Hou; Jin-sil Choi; Kuan-Ju Chen; Yang Zhang; Jinliang Peng; Mitch A. Garcia; Juehua Yu; Kaushali Thakore-Shah; Tracy Ro; Jie-Fu Chen; Parham Peyda; Guoping Fan; April D. Pyle; Hao Wang; Hsian-Rong Tseng

Supramolecular nanosubstrate-mediated delivery (SNSMD) leverages the power of molecular self-assembly and a nanostructured substrate platform for the low toxicity, highly efficient co-delivery of biological factors encapsulated in a nanovector. Human fibroblasts are successfully reprogrammed into induced pluripotent stems and transdifferentiated into induced neuronal-like cells.


Journal of Medicinal Chemistry | 2018

Reduction of Circulating Cancer Cells and Metastases in Breast-Cancer Models by a Potent EphA2-Agonistic Peptide–Drug Conjugate

Ahmed Salem; Si Wang; Sandrine Billet; Jie-Fu Chen; Parima Udompholkul; Luca Gambini; Carlo Baggio; Hsian-Rong Tseng; Edwin M. Posadas; Neil A. Bhowmick; Maurizio Pellecchia

EphA2 overexpression has been associated with metastasis in multiple cancer types, including melanomas and ovarian, prostate, lung, and breast cancers. We have recently proposed the development of peptide–drug conjugates (PDCs) using agonistic EphA2-targeting agents, such as the YSA peptide or its optimized version, 123B9. Although our studies indicated that YSA– and 123B9–drug conjugates can selectively deliver cytotoxic drugs to cancer cells in vivo, the relatively low cellular agonistic activities (i.e., the high micromolar concentrations required) of the agents toward the EphA2 receptor remained a limiting factor to the further development of these PDCs in the clinic. Here, we report that a dimeric version of 123B9 can induce receptor activation at nanomolar concentrations. Furthermore, we demonstrated that the conjugation of dimeric 123B9 with paclitaxel is very effective at targeting circulating tumor cells and inhibiting lung metastasis in breast-cancer models. These studies represent an important step toward the development of effective EphA2-targeting PDCs.

Collaboration


Dive into the Jie-Fu Chen's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Edwin M. Posadas

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar

Shuang Hou

University of California

View shared research outputs
Top Co-Authors

Avatar

Leland W.K. Chung

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar

Yi-Tsung Lu

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar

Yang Zhang

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Shirley Cheng

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Zunfu Ke

Sun Yat-sen University

View shared research outputs
Researchain Logo
Decentralizing Knowledge