Network


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

Hotspot


Dive into the research topics where Cecilia S. Leung is active.

Publication


Featured researches published by Cecilia S. Leung.


Cancer Research | 2013

TGF-β Modulates Ovarian Cancer Invasion by Upregulating CAF-Derived Versican in the Tumor Microenvironment

Tsz Lun Yeung; Cecilia S. Leung; Kwong Kwok Wong; Goli Samimi; Melissa S. Thompson; Jinsong Liu; Tarrik M. Zaid; Sue Ghosh; Michael J. Birrer; Samuel C. Mok

TGF-β has limited effects on ovarian cancer cells, but its contributions to ovarian tumor growth might be mediated through elements of the tumor microenvironment. In the present study, we tested the hypothesis that TGF modulates ovarian cancer progression by modulating the contribution of cancer-associated fibroblasts (CAF) that are present in the microenvironment. Transcriptome profiling of microdissected stromal and epithelial components of high-grade serous ovarian tumors and TGF-β-treated normal ovarian fibroblasts identified versican (VCAN) as a key upregulated target gene in CAFs. Functional evaluations in coculture experiments showed that TGF-β enhanced the aggressiveness of ovarian cancer cells by upregulating VCAN in CAFs. VCAN expression was regulated in CAFs through TGF-β receptor type II and SMAD signaling. Upregulated VCAN promoted the motility and invasion of ovarian cancer cells by activating the NF-κB signaling pathway and by upregulating expression of CD44, matrix metalloproteinase-9, and the hyaluronan-mediated motility receptor. Our work identified a TGF-β-inducible gene signature specific to CAFs in advanced high-grade serous ovarian tumors, and showed how TGF-β stimulates ovarian cancer cell motility and invasion by upregulating the CAF-specific gene VCAN. These findings suggest insights to develop or refine strategies for TGF-β-targeted therapy of ovarian cancer.


Nature Communications | 2016

Exosomal transfer of stroma-derived miR21 confers paclitaxel resistance in ovarian cancer cells through targeting APAF1

Chi Lam Au Yeung; Ngai Na Co; Tetsushi Tsuruga; Tsz Lun Yeung; Suet Ying Kwan; Cecilia S. Leung; Yong Li; Edward S. Lu; Kenny Kwan; Kwong Kwok Wong; Rosemarie Schmandt; Karen H. Lu; Samuel C. Mok

Advanced ovarian cancer usually spreads to the visceral adipose tissue of the omentum. However, the omental stromal cell-derived molecular determinants that modulate ovarian cancer growth have not been characterized. Here, using next-generation sequencing technology, we identify significantly higher levels of microRNA-21 (miR21) isomiRNAs in exosomes and tissue lysates isolated from cancer-associated adipocytes (CAAs) and fibroblasts (CAFs) than in those from ovarian cancer cells. Functional studies reveal that miR21 is transferred from CAAs or CAFs to the cancer cells, where it suppresses ovarian cancer apoptosis and confers chemoresistance by binding to its direct novel target, APAF1. These data suggest that the malignant phenotype of metastatic ovarian cancer cells can be altered by miR21 delivered by exosomes derived from neighbouring stromal cells in the omental tumour microenvironment, and that inhibiting the transfer of stromal-derived miR21 is an alternative modality in the treatment of metastatic and recurrent ovarian cancer.


Clinical Cancer Research | 2013

Identification of FGFR4 as a Potential Therapeutic Target for Advanced-Stage, High-Grade Serous Ovarian Cancer

Tarrik M. Zaid; Tsz Lun Yeung; Melissa S. Thompson; Cecilia S. Leung; Tom Harding; Ngai Na Co; Rosie S. Schmandt; Suet Ying Kwan; Cristian Rodriguez-Aguay; Gabriel Lopez-Berestein; Anil K. Sood; Kwong Kwok Wong; Michael J. Birrer; Samuel C. Mok

Purpose: To evaluate the prognostic value of fibroblast growth factor receptor 4 (FGFR4) protein expression in patients with advanced-stage, high-grade serous ovarian cancer, delineate the functional role of FGFR4 in ovarian cancer progression, and evaluate the feasibility of targeting FGFR4 in serous ovarian cancer treatment. Experimental Design: Immunolocalization of FGFR4 was conducted on 183 ovarian tumor samples. The collected FGFR4 expression data were correlated with overall survival using Kaplan–Meier and Cox regression analyses. The effects of FGFR4 silencing on ovarian cancer cell growth, survival, invasiveness, apoptosis, and FGF1-mediated signaling pathway activation were evaluated by transfecting cells with FGFR4-specific siRNAs. An orthotopic mouse model was used to evaluate the effect of injection of FGFR4-specific siRNAs and FGFR4 trap protein encapsulated in nanoliposomes on ovarian tumor growth in vivo. Results: Overexpression of FGFR4 protein was significantly associated with decreased overall survival durations. FGFR4 silencing significantly decreased the proliferation, survival, and invasiveness and increased apoptosis of ovarian cancer cells. Also, downregulation of FGFR4 significantly abrogated the mitogen-activated protein kinase (MAPK), nuclear factor-κB (NF-κB), and WNT signaling pathways, which are activated by FGF1. Targeting FGFR4 with the FGFR4-specific siRNAs and FGFR4 trap protein significantly decreased ovarian tumor growth in vivo. Conclusions: FGFR4 is a prognostic marker for advanced-stage, high-grade serous ovarian carcinoma. Silencing FGFR4 and inhibiting ligand-receptor binding significantly decrease ovarian tumor growth both in vitro and in vivo, suggesting that targeting ovarian cancer cells with high levels of FGFR4 protein expression is a new therapeutic modality for this disease and will improve survival of it. Clin Cancer Res; 19(4); 809–20. ©2012 AACR.


Nature Communications | 2014

Calcium Dependent FAK/CREB/TNNC1 Signaling Mediates the Effect of Stromal MFAP5 on Ovarian Cancer Metastatic Potential

Cecilia S. Leung; Tsz Lun Yeung; Kay-Pong Yip; Sunila Pradeep; Lavanya Balasubramanian; Jinsong Liu; Kwong Kwok Wong; Lingegowda S. Mangala; Guillermo N. Armaiz-Pena; Gabriel Lopez-Berestein; Anil K. Sood; Michael J. Birrer; Samuel C. Mok

Ovarian cancer is the most lethal gynecologic malignancy in the United States, and advanced serous ovarian adenocarcinoma is responsible for most ovarian cancer deaths. However, the stroma-derived molecular determinants that modulate patient survival have yet to be characterized. Here we identify a stromal gene signature for advanced high-grade serous ovarian cancer using microdissected stromal ovarian tumor samples and find that stromal microfibrillar-associated protein 5 (MFAP5) is a prognostic marker for poor survival. Further functional studies reveal that FAK/CREB/TNNC1 signaling pathways mediate the effect of MFAP5 on ovarian cancer cell motility and invasion potential. Targeting stromal MFAP5 using MFAP5 specific siRNA encapsulated in chitosan nanoparticles significantly decreases ovarian tumor growth and metastasis in vivo, suggesting that it may be a new modality of ovarian cancer treatment.


Biomolecules | 2016

Targeting Stromal-Cancer Cell Crosstalk Networks in Ovarian Cancer Treatment

Tsz Lun Yeung; Cecilia S. Leung; Fuhai Li; Stephen S.T. Wong; Samuel C. Mok

Ovarian cancer is a histologically, clinically, and molecularly diverse disease with a five-year survival rate of less than 30%. It has been estimated that approximately 21,980 new cases of epithelial ovarian cancer will be diagnosed and 14,270 deaths will occur in the United States in 2015, making it the most lethal gynecologic malignancy. Ovarian tumor tissue is composed of cancer cells and a collection of different stromal cells. There is increasing evidence that demonstrates that stromal involvement is important in ovarian cancer pathogenesis. Therefore, stroma-specific signaling pathways, stroma-derived factors, and genetic changes in the tumor stroma present unique opportunities for improving the diagnosis and treatment of ovarian cancer. Cancer-associated fibroblasts (CAFs) are one of the major components of the tumor stroma that have demonstrated supportive roles in tumor progression. In this review, we highlight various types of signaling crosstalk between ovarian cancer cells and stromal cells, particularly with CAFs. In addition to evaluating the importance of signaling crosstalk in ovarian cancer progression, we discuss approaches that can be used to target tumor-promoting signaling crosstalk and how these approaches can be translated into potential ovarian cancer treatment.


Oncotarget | 2017

ELF3 is a negative regulator of epithelial-mesenchymal transition in ovarian cancer cells

Tsz Lun Yeung; Cecilia S. Leung; Kwong Kwok Wong; Arthur Gutierrez-Hartmann; Joseph Kwong; David M. Gershenson; Samuel C. Mok

Transcription factors are master switches for various biochemical pathways. However, transcription factors involved in the pathogenesis of ovarian cancer have yet to be explored thoroughly. Therefore, in the present study, we assessed the prognostic value of the transcription factor E74-like factor 3 (ELF3) identified via transcriptome profiling of the epithelial components of microdissected ovarian tumor samples isolated from long- and short-term survivors and determined its roles in ovarian cancer pathogenesis. Immunohistochemical analysis of ELF3 in tumor tissue sections suggested that ELF3 was exclusively expressed by epithelial ovarian cancer cells. Furthermore, using 112 high-grade ovarian cancer samples isolated from patients and The Cancer Genome Atlas (TCGA) data, we found that downregulation of ELF3 expression was markedly associated with reduced survival. Functional studies demonstrated that overexpression of ELF3 in ovarian cancer cells suppressed proliferation and anchorage-dependent growth of the cells and that ELF3 silencing increased cell proliferation. Furthermore, upregulation of ELF3 increased expression of epithelial markers, decreased expression of mesenchymal markers, and mediated translocation of epithelial-mesenchymal transition (EMT) signaling molecules in ovarian cancer cells. Finally, we validated the tumor-inhibitory roles of ELF3 using animal models. In conclusion, ELF3 is a favorable prognostic marker for ovarian cancer. As a negative regulator of EMT, ELF3-modulated reversal of EMT may be a new effective modality in the treatment of ovarian cancer.


Journal of Clinical Investigation | 2017

Cancer-associated fibroblasts regulate endothelial adhesion protein LPP to promote ovarian cancer chemoresistance

Cecilia S. Leung; Tsz Lun Yeung; Kay-Pong Yip; Kwong Kwok Wong; Samuel Y. Ho; Lingegowda S. Mangala; Anil K. Sood; Gabriel Lopez-Berestein; Jianting Sheng; Stephen T. C. Wong; Michael J. Birrer; Samuel C. Mok

The molecular mechanism by which cancer-associated fibroblasts (CAFs) confer chemoresistance in ovarian cancer is poorly understood. The purpose of the present study was to evaluate the roles of CAFs in modulating tumor vasculature, chemoresistance, and disease progression. Here, we found that CAFs upregulated the lipoma-preferred partner (LPP) gene in microvascular endothelial cells (MECs) and that LPP expression levels in intratumoral MECs correlated with survival and chemoresistance in patients with ovarian cancer. Mechanistically, LPP increased focal adhesion and stress fiber formation to promote endothelial cell motility and permeability. siRNA-mediated LPP silencing in ovarian tumor–bearing mice improved paclitaxel delivery to cancer cells by decreasing intratumoral microvessel leakiness. Further studies showed that CAFs regulate endothelial LPP via a calcium-dependent signaling pathway involving microfibrillar-associated protein 5 (MFAP5), focal adhesion kinase (FAK), ERK, and LPP. Thus, our findings suggest that targeting endothelial LPP enhances the efficacy of chemotherapy in ovarian cancer. Our data highlight the importance of CAF–endothelial cell crosstalk signaling in cancer chemoresistance and demonstrate the improved efficacy of using LPP-targeting siRNA in combination with cytotoxic drugs.


Journal of the National Cancer Institute | 2018

Systematic Identification of Druggable Epithelial-Stromal Crosstalk Signaling Networks in Ovarian Cancer

Tsz-Lun Yeung; Jianting Sheng; Cecilia S. Leung; Fuhai Li; Jaeyeon Kim; Samuel Y. Ho; Martin M. Matzuk; Karen H. Lu; Stephen T. C. Wong; Samuel C. Mok

Abstract Background Bulk tumor tissue samples are used for generating gene expression profiles in most research studies, making it difficult to decipher the stroma–cancer crosstalk networks. In the present study, we describe the use of microdissected transcriptome profiles for the identification of cancer–stroma crosstalk networks with prognostic value, which presents a unique opportunity for developing new treatment strategies for ovarian cancer. Methods Transcriptome profiles from microdissected ovarian cancer–associated fibroblasts (CAFs) and ovarian cancer cells from patients with high-grade serous ovarian cancer (n = 70) were used as input data for the computational systems biology program CCCExplorer to uncover crosstalk networks between various cell types within the tumor microenvironment. The crosstalk analysis results were subsequently used for discovery of new indications for old drugs in ovarian cancer by computational ranking of candidate agents. Survival analysis was performed on ovarian tumor–bearing Dicer/Pten double-knockout mice treated with calcitriol, a US Food and Drug Administration–approved agent that suppresses the Smad signaling cascade, or vehicle control (9–11 mice per group). All statistical tests were two-sided. Results Activation of TGF-β-dependent and TGF-β-independent Smad signaling was identified in a particular subtype of CAFs and was associated with poor patient survival (patients with higher levels of Smad-regulated gene expression by CAFs: median overall survival = 15 months, 95% confidence interval [CI] = 12.7 to 17.3 months; vs patients with lower levels of Smad-regulated gene expression: median overall survival = 26 months, 95% CI = 15.9 to 36.1 months, P = .02). In addition, the activated Smad signaling identified in CAFs was found to be targeted by repositioning calcitriol. Calcitriol suppressed Smad signaling in CAFs, inhibited tumor progression in mice, and prolonged the median survival duration of ovarian cancer–bearing mice from 36 to 48 weeks (P = .04). Conclusions Our findings suggest the feasibility of using novel multicellular systems biology modeling to identify and repurpose known drugs targeting cancer–stroma crosstalk networks, potentially leading to faster and more effective cures for cancers.


Cancer Research | 2014

Abstract 4799: Identification and characterization of stromal factors with clinical significance in the ovarian tumor microenvironment

Tsz-Lun Yeung; Cecilia S. Leung; Kwong Kwok Wong; Samuel C. Mok

Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CA Ovarian cancer is the most lethal gynecologic malignancy in the United States. While the tumor microenvironment has been shown to play important roles in cancer pathogenesis, large-scale transcriptome profiles of the stromal component of ovarian tumors and the identification of stromal prognostic markers and therapeutic targets are lacking. This study seeks to identify secreted stromal factors and evaluate their clinical significance. Transcriptome profiling and clustering analysis on microdissected ovarian cancer associated fibroblasts (CAFs) samples from 83 patients revealed a heterogeneous gene expression pattern. Since CAFs can be derived from mesenchymal stem cells (MSCs), we compared transcriptome profiles of MSC samples obtained from healthy individuals with those from CAFs. Results showed that CAF expression profiles could be classified into two major subtypes. The MSC subtype, which has gene expression pattern highly resembling to that of undifferentiated MSCs, is significantly associated with poorer patient survival when compared to the non-MSC subtype. Further analyses identified two expression clusters within the MSC subtype: 1) the CAF-C cluster which expressed high level of MSC chondrogenic differentiation associated genes including CSPG2, SFRP2 and COMP, and 2) the CAF-O cluster which expressed high level of osteogenic differentiation associated genes including RUNX2 and BGN. Survival analysis showed that patients with the CAF-O expression subtype had overall survival rates comparable to those with the non-MSC subtypes, while the CAF-C subtype is associated with worst clinical outcomes. Pathway analyses revealed strong interactions among these stromal genes. Among them, SFRP2, which has been shown to inhibit osteogenic differentiation of MSCs by inhibiting Wnt signaling and subsequently suppress RUNX2 expression, was selected for further validation studies. Stromal SFRP2 and RUNX2 protein expression levels were evaluated by immunohistochemistry on paraffin tissue sections from 94 patients. Results showed that high levels of stromal SFRP2 were significantly associated with poor patient survival (p < 0.001), while high levels of stromal RUNX2 were significantly associated with improved patient survival (p < 0.001). A significant inverse correlation between SFRP2 and RUNX2 expression levels was also observed (r = -0.279, p = 0.037), suggested that the interactions among these genes may generate different landscapes in the tumor microenvironment, which modulate MSC differentiation and subsequently cancer cell aggressiveness. This study presents a new paradigm of which osteogenic-like and chondrogenic-like CAF signatures are associated with better and poorer patient survival respectively. Understanding the relationship between the expression patterns of stromal factors and MSC differentiation would provide us with new insights into ovarian cancer pathogenesis. Citation Format: Tsz-Lun Yeung, Cecilia S. Leung, Kwong-Kwok Wong, Samuel C. Mok. Identification and characterization of stromal factors with clinical significance in the ovarian tumor microenvironment. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 4799. doi:10.1158/1538-7445.AM2014-4799


Cancer Research | 2017

Abstract LB-214: The immunosuppressive roles of ovarian stromal tumor microenvironment

Tsz-Lun Yeung; Cecilia S. Leung; Kwong Kwok Wong; Michael J. Birrer; Stephen T. C. Wong; Karen H. Lu; Samuel C. Mok

Ovarian cancer is the most lethal gynecologic malignancy in the US. Our group and others has shown that CD8+ lymphocyte infiltration in the ovarian tumor epithelium is associated with prolonged survival in patients with high-grade serous ovarian cancer. Despite the increasing evidence on stromal involvement in tumor progression, the interactions between stromal fibroblasts and cancer cells, as well as the underlying genetic composition of the stromal cells that could regulate the infiltration and activation of CD8+ cytotoxic T lymphocytes (CTLs) and affect patient survival is not fully understood. The present study seeks to evaluate the roles and to delineate the underlying mechanisms by which stromal cancer associated fibroblasts (CAFs) modulates immune response in ovarian cancer, particularly immune suppression by CAF-derived protein factors. By laser microdissection and transcriptome profiling of tumor tissue samples from ovarian cancer patients, we identified CAF-specific gene signatures for ovarian cancer that are associated with the survival duration of patients and with the differential tumor immune response. Among the differentially expressed genes identified, validation study by qPCR and immunohistochemistry indicated that stromal MFAP5 expression is significantly higher in patients with short survival duration and in patients with lower intratumoral CD8+ T cell density. Survival analysis result suggested that high stromal MFAP5 expression indicates poor disease prognosis (Hazard ratio=2.722, P Delineating the molecular mechanism by which MFAP5 modulates the progression and immune responses of ovarian cancer will shed light on the design of novel treatment modalities based on the blockade of tumor stroma-derived factors, which could promote activation and trafficking of cytotoxic CD8+ T cells and improve patient survival rates. Citation Format: Tsz-Lun Yeung, Cecilia S. Leung, Kwong-Kwok Wong, Michael J. Birrer, Stephen T. Wong, Karen H. Lu, Samuel C. Mok. The immunosuppressive roles of ovarian stromal tumor microenvironment [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr LB-214. doi:10.1158/1538-7445.AM2017-LB-214

Collaboration


Dive into the Cecilia S. Leung's collaboration.

Top Co-Authors

Avatar

Samuel C. Mok

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Kwong Kwok Wong

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Tsz-Lun Yeung

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Tsz Lun Yeung

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Chi Lam Au Yeung

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Karen H. Lu

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Kay-Pong Yip

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Ngai Na Co

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Anil K. Sood

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge