Hung-Cheng Lai
National Defense Medical Center
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Hung-Cheng Lai.
Oncogene | 2013
W. K. Chau; Carman K.M. Ip; Abby Sin-Chi Mak; Hung-Cheng Lai; Alice S. T. Wong
Cisplatin and paclitaxel are standard chemotherapy for metastatic ovarian cancer, but with limited efficacy. Cancer stem/progenitor cells (or tumor-initiating cells, TICs) are hypothesized to be chemoresistant, and the existence of TICs in ovarian cancer has been previously demonstrated. However, the key signals and molecular events regulating the formation and expansion of ovarian tumor-initiating cells (OTICs) remain elusive. Here, we show that c-Kit is not just a marker of OTICs, but also a critical mediator of the phenotype that can be a viable target for the treatment of ovarian cancer. In contrast to non-OICs, c-Kit was overexpressed in OTICs. Moreover, the use of small interfering RNA to inhibit c-Kit expression markedly attenuated the number and size of OTIC subpopulations, inhibited the expression of stem cell markers and decreased the tumorigenic capabilities of OTICs. Imatinib (Gleevec), a clinical drug that blocks c-Kit kinase activity, also demonstrated its inhibition potency on OTICs. In addition, cisplatin/paclitaxel, which killed non-OTICs, with c-Kit knockdown or imatinib revealed that this was critically required for intervening ovarian cancer progression and recurrence in vitro and in xenograft tumors in vivo. Similar results were obtained with OTICs derived from ovarian carcinoma patients. Studies into the mechanisms suggest an important role for the activation of Wnt/β-catenin and ATP-binding cassette G2 downstream of c-Kit. The tumor-promoting microenvironment, such as hypoxia, could promote OTICs via upregulation of c-Kit expression. These results unravel an integral role for c-Kit in ovarian neoplastic processes and shed light on its mechanisms of action.
Molecular Cancer Therapeutics | 2012
Yi Te Yo; Ya W. Lin; Yu Chi Wang; Curt Balch; Rui Lan Huang; Michael W.Y. Chan; Huey-Kang Sytwu; Chi Kuan Chen; Cheng Chang Chang; Kenneth P. Nephew; Tim H M Huang; Mu Hsien Yu; Hung-Cheng Lai
A recent hypothesis for cancer chemoresistance posits that cytotoxic survival of a subpopulation of tumor progenitors drives the propagation of recurrent disease, underscoring the need for new therapeutics that target such primitive cells. To discover such novel compounds active against drug-resistant ovarian cancer, we identified a subset of chemoresistant ovarian tumor cells fulfilling current definitions of cancer-initiating cells from cell lines and patient tumors using multiple stemness phenotypes, including the expression of stem cell markers, membrane dye efflux, sphere formation, potent tumorigenicity, and serial tumor propagation. We then subjected such stem-like ovarian tumor-initiating cells (OTIC) to high-throughput drug screening using more than 1,200 clinically approved drugs. Of 61 potential compounds preliminarily identified, more stringent assessments showed that the antihelmintic niclosamide selectively targets OTICs in vitro and in vivo. Gene expression arrays following OTIC treatment revealed niclosamide to disrupt multiple metabolic pathways affecting biogenetics, biogenesis, and redox regulation. These studies support niclosamide as a promising therapy for ovarian cancer and warrant further preclinical and clinical evaluation of this safe, clinically proven drug for the management of this devastating gynecologic malignancy. Mol Cancer Ther; 11(8); 1703–12. ©2012 AACR.
International Journal of Cancer | 2014
Pei Ning Yu; Ming D. Yan; Hung-Cheng Lai; Rui Lan Huang; Yu Ching Chou; Wen Chi Lin; Li Tzu Yeh; Ya W. Lin
Drug resistance is an obstacle to the treatment of ovarian cancer. Using a unique cell model, we have proven previously that a subpopulation of ovarian cancer cells is more resistant to cisplatin than are the original cells. MicroRNAs (miRNAs), small noncoding RNAs, are involved in many biological events in cancer cells. In our study, we explored whether miRNAs are involved in cisplatin resistance of ovarian cancer cells. Cisplatin‐resistant cells expressed a lower level of miR‐29a/b/c. Manipulation of microRNA‐29 (miR‐29) expression modulated cisplatin sensitivity of CP70, HeyC2, SKOV3 and A2780 ovarian cancer cells. Knockdown of miR‐29a/b/c increased the ability of cells to escape cisplatin‐induced cell death partly through upregulation of collagen type I alpha 1 (COL1A1) and increased the activation of extracellular signal‐regulated kinase 1/2 and inactivation of glycogen synthase kinase 3 beta. When combined with cisplatin treatment, knockdown of miR‐29 decreased the amount of the active form of caspase‐9 and caspase‐3. Ectopic expression of miR‐29 alone or in combination with cisplatin treatment efficaciously reduced the tumorigenicity of CP70 cells in vivo. Our data show that downregulation of miR‐29 increases cisplatin resistance in ovarian cancer cells. Taken together, these data suggest that overexpression of miR‐29 is a potential sensitizer to cisplatin treatment that may have therapeutic implications.
Oncogene | 2013
Po-Hsuan Su; Yen-Shing Lin; Rui Lan Huang; Yu-Ping Liao; H. Y. Lee; Hui Chen Wang; Tai-Kuang Chao; C. K. Chen; Michael W.Y. Chan; Tang-Yuan Chu; Mu Hsien Yu; Hung-Cheng Lai
Epigenetic modifications are a driving force in carcinogenesis. However, their role in cancer metastasis remains poorly understood. The present study investigated the role of DNA methylation in the cervical cancer metastasis. Here, we report evidence of the overexpression of DNA methyltransferases 3B (DNMT3B) in invasive cervical cancer and of the inhibition of metastasis by DNMT3B interference. Using methyl-DNA immunoprecipitation coupled with microarray analysis, we found that the protein tyrosine phosphatase receptor type R (PTPRR) was silenced through DNMT3B-mediated methylation in the cervical cancer. PTPRR inhibited p44/42 MAPK signaling, the expression of the transcription factor AP1, human papillomavirus (HPV) oncogenes E6/E7 and DNMTs. The methylation status of PTPRR increased in cervical scrapings (n=358) in accordance with disease severity, especially in invasive cancer. Methylation of the PTPRR promoter has an important role in the metastasis and may be a biomarker of invasive cervical cancer.
Oncotarget | 2017
Ping-Ying Chang; Yu-Ping Liao; Hui-Chen Wang; Yu-Chih Chen; Rui Lan Huang; Yu-Chi Wang; Chiou-Chung Yuan; Hung-Cheng Lai
DNA methylation is a promising biomarker for cancer. The epigenetic effects of cell adhesion molecules may affect the therapeutic outcome and the present study examined their effects on survival in ovarian cancer. We integrated methylomics and genomics datasets in The Cancer Genome Atlas (n = 391) and identified 106 highly methylated adhesion-related genes in ovarian cancer tissues. Univariate analysis revealed the methylation status of eight genes related to progression-free survival. In multivariate Cox regression analysis, four highly methylated genes (CD97, CTNNA1, DLC1, HAPLN2) and three genes (LAMA4, LPP, MFAP4) with low methylation were significantly associated with poor progression-free survival. Low methylation of VTN was an independent poor prognostic factor for overall survival after adjustment for age and stage. Patients who carried any two of CTNNA1, DLC1 or MFAP4 were significantly associated with poor progression-free survival (hazard ratio: 1.59; 95% confidence interval: 1.23, 2.05). This prognostic methylation signature was validated in a methylomics dataset generated in our lab (n = 37, hazard ratio: 16.64; 95% confidence interval: 2.68, 103.14) and in another from the Australian Ovarian Cancer Study (n = 91, hazard ratio: 2.43; 95% confidence interval: 1.11, 5.36). Epigenetics of cell adhesion molecules is related to ovarian cancer prognosis. A more comprehensive methylomics of cell adhesion molecules is needed and may advance personalized treatment with adhesion molecule-related drugs.DNA methylation is a promising biomarker for cancer. The epigenetic effects of cell adhesion molecules may affect the therapeutic outcome and the present study examined their effects on survival in ovarian cancer. We integrated methylomics and genomics datasets in The Cancer Genome Atlas (n = 391) and identified 106 highly methylated adhesion-related genes in ovarian cancer tissues. Univariate analysis revealed the methylation status of eight genes related to progression-free survival. In multivariate Cox regression analysis, four highly methylated genes (CD97, CTNNA1, DLC1, HAPLN2) and three genes (LAMA4, LPP, MFAP4) with low methylation were significantly associated with poor progression-free survival. Low methylation of VTN was an independent poor prognostic factor for overall survival after adjustment for age and stage. Patients who carried any two of CTNNA1, DLC1 or MFAP4 were significantly associated with poor progression-free survival (hazard ratio: 1.59; 95% confidence interval: 1.23, 2.05). This prognostic methylation signature was validated in a methylomics dataset generated in our lab (n = 37, hazard ratio: 16.64; 95% confidence interval: 2.68, 103.14) and in another from the Australian Ovarian Cancer Study (n = 91, hazard ratio: 2.43; 95% confidence interval: 1.11, 5.36). Epigenetics of cell adhesion molecules is related to ovarian cancer prognosis. A more comprehensive methylomics of cell adhesion molecules is needed and may advance personalized treatment with adhesion molecule-related drugs.
International Journal of Cancer | 2018
Phui-Ly Liew; Rui Lan Huang; Yu-Chun Weng; Chia-Lang Fang; Tim H M Huang; Hung-Cheng Lai
Mucinous type of epithelial ovarian cancer (MuOC) is a unique subtype with a poor survival outcome in recurrent and advanced stages. The role of type‐specific epigenomics and its clinical significance remains uncertain. We analyzed the methylomic profiles of 6 benign mucinous adenomas, 24 MuOCs, 103 serous type of epithelial ovarian cancers (SeOCs) and 337 nonepithelial ovarian cancers. MuOC and SeOC exhibited distinct DNA methylation profiles comprising 101 genes, 81 of which exhibited low methylation in MuOC and were associated with the response to glucocorticoid, ATP hydrolysis‐coupled proton transport, proteolysis involved in the cellular protein catabolic process and ion transmembrane transport. Hierarchical clustering analysis showed that the profiles of MuOC were similar to colorectal adenocarcinoma and stomach adenocarcinoma. Genetic interaction network analysis of differentially methylated genes in MuOC showed a dominant network module is the proteasome subunit beta (PSMB) family. Combined functional module and methylation analysis identified PSMB8 as a candidate marker for MuOC. Immunohistochemical staining of PSMB8 used to validate in 94 samples of ovarian tumors (mucinous adenoma, MuOC or SeOC) and 62 samples of gastrointestinal cancer. PSMB8 was commonly expressed in MuOC and gastrointestinal cancer samples, predominantly as strong cytoplasmic and occasionally weak nuclei staining, but was not expressed in SeOC samples. Carfilzomib, a second‐generation proteasome inhibitor, suppressed MuOC cell growth in vitro. This study unveiled a mucinous‐type‐specific methylation profile and suggests the potential use of a proteasome inhibitor to treat MuOC.
Cancer Research | 2011
Lin Yu Chen; Rui Lan Huang; Han Lin Chou; Jian Liang Chou; Wei-Ting Deng; Hui Wen Yang; Ru Inn Lin; Chin Yang Li; Huey-Jen Lin; Tim Hui Ming Huang; Chien Chih Chiu; Hung-Cheng Lai; Michael W.Y. Chan
The TGF-β signaling pathway plays an important role in controlling cell growth and differentiation. In advanced ovarian cancer, frequent TGF-β-induced metastasis or epithelial-mesenchymal transition (EMT) can be observed. This phenomenon is often associated with epigenetic silencing of epithelial marker, E-cadherin which can also be observed in ovarian cancer cell lines that demethylation treatment restored E-cadherin expression. We recently hypothesized that long term activation of TGF-β signaling may induce EMT phenotype by epigenetic silencing of E-cadherin and that inhibition of the signaling may restore E-cadherin and reverse EMT in ovarian cancer (Chou et al., Expert Opin Ther Targets 2010). In this study, we cloned the cDNA of the inhibitory SMAD, SMAD7 from a human immortalized ovarian surface epithelial cell, IOSE into pcDNA3.1 mammalian expression vector. The inhibitory effect of this SMAD7 expression vector on TGF-β signaling has been confirmed by reporter assay. We then stably transfected the SMAD7 expression vector into a mesenchymal ovarian cancer cell, CP70 in which E-cadherin is silenced by complete promoter methylation. Cells over-expressing SMAD7 showed up-regulation of SMAD7 and a decrease in SMAD2 phosphorylation while the control cells maintained a hyperphosphorylation of SMAD2 thus suggesting that TGF-β signaling is disrupted in SMAD7-overexpressing cells. We further examined the expression of E-cadherin from passage 5 up to 30 of the stable transfectants. Surprisingly, stable restoration of E-cadherin can only be observed from passage 20 and thereafter, of the SMAD7-overexpressing cells, while E-cadherin remained silence in the control cells. To investigate if this restoration is due to promoter demethylation of E-cadherin, we performed bisulphite pyrosequencing on the E-cadherin promoter CpG island spanning -586 to -12 of the region. Compared with control cells, consistent demethylation of E-cadherin promoter can be observed at 2 CpG sites located at -214 and -235 of the promoter such that gradual demethylation occurred from passage 5 to 30 of the SMAD7-overexpressing cells (passage 20, methylation% control vs SMAD7: 91% vs 67% at -235; 80% vs 52% at -214); while the rest of the CpG sites remained heavily methylated. This demethylation may be due to down-regulation of transcriptional repressor, TWIST after SMAD7 transfection. Additionally, one of the SMAD7 stable expression clones with highest restoration of E-cadherin showed decreased migration and invasion ability as determined by wound healing and invasion assay. Taken together, disruption of TGF-β signaling can induce demethylation of E-cadherin promoter and reverse EMT phenotype in ovarian cancer. The therapeutic potential of targeting TGF-β signaling pathway in inhibiting metastasis of ovarian cancer deserves further investigation. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 3433. doi:10.1158/1538-7445.AM2011-3433
Nature Communications | 2018
Maggie K. S. Tang; Patrick Ying-Kit Yue; Philip P.C. Ip; Rui Lan Huang; Hung-Cheng Lai; Annie N.Y. Cheung; Ka Yu Tse; Hys Ngan; Alice S. T. Wong
The limitations of current anti-angiogenic therapies necessitate other targets with complimentary mechanisms. Here, we show for the first time that soluble E-cadherin (sE-cad) (an 80-kDa soluble form), which is highly expressed in the malignant ascites of ovarian cancer patients, is a potent inducer of angiogenesis. In addition to ectodomain shedding, we provide further evidence that sE-cad is abundantly released in the form of exosomes. Mechanistically, sE-cad-positive exosomes heterodimerize with VE-cadherin on endothelial cells and transduce a novel sequential activation of β-catenin and NFκB signaling. In vivo and clinical data prove the relevance of sE-cad-positive exosomes for malignant ascites formation and widespread peritoneal dissemination. These data advance our understanding of the molecular regulation of angiogenesis in ovarian cancer and support the therapeutic potential of targeting sE-cad. The exosomal release of sE-cad, which represents a common route for externalization in ovarian cancer, could potentially be biomarkers for diagnosis and prognosis.A soluble form E-cadherin is highly expressed in ovarian cancer. Here, the authors show that soluble E-cadherin is released by ovarian cancer cells packaged in exosomes and promotes tumor angiogenesis through β-catenin and NFkB signaling activation.
International Journal of Cancer | 2018
Rui Lan Huang; Hsiang-Ju Chen; Lin-Yu Chen; Tai-Kuang Chao; Wei-Yu Lin; Phui-Ly Liew; Po-Hsuan Su; Yu-Chun Weng; Yu-Chi Wang; Chi-Chun Liao; Yaw-Wen Hsu; Hui-Chen Wang; Hung-Cheng Lai
Precision medicine requires markers for therapeutic guidance. The purpose of this study was to determine whether epithelial ovarian cancer (EOC) epigenetics can lead to the identification of biomarkers for precision medicine. Through integrative methylomics, we discovered and validated the epigenetic signature of NEFH and HS3ST2 as an independent prognostic factor for type II EOC in our dataset (n = 84), and two independent methylomics datasets (total n = 467). Integrated transcriptomics dataset (n = 1147) and tissue microarrays (n = 54) of HS3ST2 also related to high‐methylation statuses and the EOC prognosis. Mechanistic explorations of HS3ST2 have assessed responses to oncogenic stimulations such as IL‐6, EGF, and FGF2 in cancer cells. The combination of HS3ST2 and various oncogenic ligands also confers the worse outcome. 3‐O‐sulfation of heparan sulfate by HS3ST2 makes ovarian cancer cells intrinsically sensitive to oncogenic signals, which sheds new light on the application of HS3ST2 as a companion diagnostic for targeted therapy using kinase inhibitors or therapeutic antibodies.
International Journal of Cancer | 2018
Hsiang-Ju Chen; Rui Lan Huang; Phui-Ly Liew; Po-Hsuan Su; Lin-Yu Chen; Yu-Chun Weng; Cheng-Chang Chang; Yu-Chi Wang; Michael Wing-Yan Chan; Hung-Cheng Lai
Ovarian high‐grade serous carcinoma (HGSC) is the most lethal gynecological malignancy. Prevailing evidences suggest that drug resistance and recurrence of ovarian HGSC are caused by the presence of cancer stem cells. Therefore, targeting cancer stems is appealing, however, all attempts to date, have failed. To circumvent this limit, we analyzed differential transcriptomes at early differentiation of ovarian HGSC stem cells and identified the developmental transcription factor GATA3 as highly expressed in stem, compared to progenitor cells. GATA3 expression associates with poor prognosis of ovarian HGSC patients, and was found to recruit the histone H3, lysine 27 (H3K27) demethylase, UTX, activate stemness markers, and promote stem‐like phenotypes in ovarian HGSC cell lines. Targeting UTX by its inhibitor, GSKJ4, impeded GATA3‐driven stemness phenotypes, and enhanced apoptosis of GATA3‐expressing cancer cells. Combinations of gemcitabine or paclitaxel with GSKJ4, resulted in a synergistic cytotoxic effect. Our findings provide evidence for a new role for GATA3 in ovarian HGSC stemness, and demonstrate that GATA3 may serve as a biomarker for precision epigenetic therapy in the future.