Network


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

Hotspot


Dive into the research topics where Zhiyong Liao is active.

Publication


Featured researches published by Zhiyong Liao.


Cancer Research | 2008

Transcription Factor Stat5 Synergizes with Androgen Receptor in Prostate Cancer Cells

Shyh-Han Tan; Ayush Dagvadorj; Feng Shen; Lei Gu; Zhiyong Liao; Junaid Abdulghani; Ying Zhang; Edward P. Gelmann; Tobias Zellweger; Zoran Culig; Tapio Visakorpi; Lukas Bubendorf; Robert A. Kirken; James G. Karras; Marja T. Nevalainen

The molecular mechanisms underlying progression of prostate cancer to the hormone-independent state are poorly understood. Signal transducer and activator of transcription 5a and 5b (Stat5a/b) is critical for the viability of human prostate cancer cells. We have previously shown that Stat5a/b is constitutively active in high-grade human prostate cancer, but not in normal prostate epithelium. Furthermore, activation of Stat5a/b in primary human prostate cancer predicted early disease recurrence. We show here that transcription factor Stat5a/b is active in 95% of clinical hormone-refractory human prostate cancers. We show for the first time that Stat5a/b synergizes with androgen receptor (AR) in prostate cancer cells. Specifically, active Stat5a/b increases transcriptional activity of AR, and AR, in turn, increases transcriptional activity of Stat5a/b. Liganded AR and active Stat5a/b physically interact in prostate cancer cells and, importantly, enhance nuclear localization of each other. The work presented here provides the first evidence of synergy between AR and the prolactin signaling protein Stat5a/b in human prostate cancer cells.


Clinical Cancer Research | 2008

Androgen-regulated and highly tumorigenic human prostate cancer cell line established from a transplantable primary CWR22 tumor

Ayush Dagvadorj; Shyh-Han Tan; Zhiyong Liao; Luciane R. Cavalli; Bassem R. Haddad; Marja T. Nevalainen

Purpose: One of the major obstacles in understanding the molecular mechanisms underlying the transition of prostate cancer growth from androgen dependency to a hormone-refractory state is the lack of androgen-regulated and tumorigenic human prostate cancer cell lines. Experimental Design: We have established and characterized a new human prostate cancer cell line, CWR22Pc, derived from the primary CWR22 human prostate xenograft tumors. Results: The growth of CWR22Pc cells is induced markedly by dihydrotestosterone, and CWR22Pc cells express high levels of androgen receptor (AR) and prostate-specific antigen (PSA). Importantly, PSA expression in CWR22Pc cells is regulated by androgens. Stat5a/b, Stat3, Akt, and mitogen-activated protein kinase were constitutively active or cytokine inducible in CWR22Pc cells. The AR in CWR22Pc cells contains the H874Y mutation, but not the exon 3 duplication or other mutations. When inoculated subcutaneously into dihydrotestosterone-supplemented castrated nude mice, large tumors formed rapidly in 20 of 20 mice, whereas no tumors developed in mice without circulating dihydrotestosterone. Moreover, the serum PSA levels correlated with the tumor volumes. When androgens were withdrawn from the CWR22Pc tumors grown in nude mice, the tumors initially shrank but regrew back as androgen-independent tumors. Conclusions: This androgen-regulated and tumorigenic human prostate cancer cell line provides a valuable tool for studies on androgen regulation of prostate cancer cells and on the molecular mechanisms taking place in growth promotion of prostate cancer when androgens are withdrawn from the growth environment. CWR22Pc cells also provide a model system for studies on the regulation of transcriptional activity of mutated H874YAR in a prostate cancer cell context.


Clinical Cancer Research | 2013

Pharmacologic Inhibition of Jak2–Stat5 Signaling By Jak2 Inhibitor AZD1480 Potently Suppresses Growth of Both Primary and Castrate-Resistant Prostate Cancer

Lei Gu; Zhiyong Liao; David T. Hoang; Ayush Dagvadorj; Shilpa Gupta; Shauna Blackmon; Elyse Ellsworth; Pooja Talati; Benjamin E. Leiby; Michael Zinda; Edouard J. Trabulsi; Peter McCue; Leonard G. Gomella; Dennis Huszar; Marja T. Nevalainen

Purpose: Progression of prostate cancer to the lethal castrate-resistant stage coincides with loss of responsiveness to androgen deprivation and requires development of novel therapies. We previously provided proof-of-concept that Stat5a/b is a therapeutic target protein for prostate cancer. Here, we show that pharmacologic targeting of Jak2-dependent Stat5a/b signaling by the Jak2 inhibitor AZD1480 blocks castrate-resistant growth of prostate cancer. Experimental Design: Efficacy of AZD1480 in disrupting Jak2–Stat5a/b signaling and decreasing prostate cancer cell viability was evaluated in prostate cancer cells. A unique prostate cancer xenograft mouse model (CWR22Pc), which mimics prostate cancer clinical progression in patients, was used to assess in vivo responsiveness of primary and castrate-resistant prostate cancer (CRPC) to AZD1480. Patient-derived clinical prostate cancers, grown ex vivo in organ explant cultures, were tested for responsiveness to AZD1480. Results: AZD1480 robustly inhibited Stat5a/b phosphorylation, dimerization, nuclear translocation, DNA binding, and transcriptional activity in prostate cancer cells. AZD1480 reduced prostate cancer cell viability sustained by Jak2–Stat5a/b signaling through induction of apoptosis, which was rescued by constitutively active Stat5a/b. In mice, pharmacologic targeting of Stat5a/b by AZD1480 potently blocked growth of primary androgen-dependent as well as recurrent castrate-resistant CWR22Pc xenograft tumors, and prolonged survival of tumor-bearing mice versus vehicle or docetaxel-treated mice. Finally, nine of 12 clinical prostate cancers responded to AZD1480 by extensive apoptotic epithelial cell loss, concurrent with reduced levels of nuclear Stat5a/b. Conclusions: We report the first evidence for efficacy of pharmacologic targeting of Stat5a/b as a strategy to inhibit castrate-resistant growth of prostate cancer, supporting further clinical development of Stat5a/b inhibitors as therapy for advanced prostate cancer. Clin Cancer Res; 19(20); 5658–74. ©2013 AACR.


Molecular Cancer Therapeutics | 2014

Pharmacologic Suppression of JAK1/2 by JAK1/2 Inhibitor AZD1480 Potently Inhibits IL-6–Induced Experimental Prostate Cancer Metastases Formation

Lei Gu; Pooja Talati; Paraskevi Vogiatzi; Ana L. Romero-Weaver; Junaid Abdulghani; Zhiyong Liao; Benjamin E. Leiby; David T. Hoang; Tuomas Mirtti; Kalle Alanen; Michael Zinda; Dennis Huszar; Marja T. Nevalainen

Metastatic prostate cancer is lethal and lacks effective strategies for prevention or treatment, requiring novel therapeutic approaches. Interleukin-6 (IL-6) is a cytokine that has been linked with prostate cancer pathogenesis by multiple studies. However, the direct functional roles of IL-6 in prostate cancer growth and progression have been unclear. In the present study, we show that IL-6 is produced in distant metastases of clinical prostate cancers. IL-6–activated signaling pathways in prostate cancer cells induced a robust 7-fold increase in metastases formation in nude mice. We further show that IL-6 promoted migratory prostate cancer cell phenotype, including increased prostate cancer cell migration, microtubule reorganization, and heterotypic adhesion of prostate cancer cells to endothelial cells. IL-6–driven metastasis was predominantly mediated by Stat3 and to lesser extent by ERK1/2. Most importantly, pharmacologic inhibition of Jak1/2 by AZD1480 suppressed IL-6–induced signaling, migratory prostate cancer cell phenotypes, and metastatic dissemination of prostate cancer in vivo in nude mice. In conclusion, we demonstrate that the cytokine IL-6 directly promotes prostate cancer metastasis in vitro and in vivo via Jak–Stat3 signaling pathway, and that IL-6–driven metastasis can be effectively suppressed by pharmacologic targeting of Jak1/2 using Jak1/2 inhibitor AZD1480. Our results therefore provide a strong rationale for further development of Jak1/2 inhibitors as therapy for metastatic prostate cancer. Mol Cancer Ther; 13(5); 1246–58. ©2014 AACR.


The International Journal of Biochemistry & Cell Biology | 2010

Transcription factor Stat5a/b as a therapeutic target protein for prostate cancer

Zhiyong Liao; Jacqueline Lutz; Marja T. Nevalainen

Prostate cancer is the most common non-cutaneous cancer in Western males. The majority of prostate cancer fatalities are caused by development of castration-resistant growth and metastatic spread of the primary tumor. The average duration of the response of primary prostate cancer to hormonal ablation is less than 3 years, and 75% of prostate cancers in the United States progress to castration-resistant disease. The existing pharmacological therapies for metastatic and/or castration-resistant prostate cancer do not provide significant survival benefit. This review summarizes the importance of transcription factor Stat5 signaling in the pathogenesis of prostate cancer and discusses the molecular basis of Stat5a/b inhibition as a therapeutic strategy for prostate cancer.


Molecular Cancer Therapeutics | 2015

Structure-Based Screen Identifies a Potent Small Molecule Inhibitor of Stat5a/b with Therapeutic Potential for Prostate Cancer and Chronic Myeloid Leukemia

Zhiyong Liao; Lei Gu; Jenny Vergalli; Samanta A. Mariani; Marco De Dominici; Ravi K. Lokareddy; Ayush Dagvadorj; Puranik Purushottamachar; Peter McCue; Edouard J. Trabulsi; Shilpa Gupta; Elyse Ellsworth; Shauna Blackmon; Adam Ertel; Paolo Fortina; Benjamin E. Leiby; Guanjun Xia; Hallgeir Rui; David T. Hoang; Leonard G. Gomella; Gino Cingolani; Vincent C. O. Njar; Nagarajan Pattabiraman; Bruno Calabretta; Marja T. Nevalainen

Bypassing tyrosine kinases responsible for Stat5a/b phosphorylation would be advantageous for therapy development for Stat5a/b-regulated cancers. Here, we sought to identify small molecule inhibitors of Stat5a/b for lead optimization and therapy development for prostate cancer and Bcr-Abl–driven leukemias. In silico screening of chemical structure databases combined with medicinal chemistry was used for identification of a panel of small molecule inhibitors to block SH2 domain–mediated docking of Stat5a/b to the receptor-kinase complex and subsequent phosphorylation and dimerization. We tested the efficacy of the lead compound IST5-002 in experimental models and patient samples of two known Stat5a/b-driven cancers, prostate cancer and chronic myeloid leukemia (CML). The lead compound inhibitor of Stat5-002 (IST5-002) prevented both Jak2 and Bcr-Abl–mediated phosphorylation and dimerization of Stat5a/b, and selectively inhibited transcriptional activity of Stat5a (IC50 = 1.5μmol/L) and Stat5b (IC50 = 3.5 μmol/L). IST5-002 suppressed nuclear translocation of Stat5a/b, binding to DNA and Stat5a/b target gene expression. IST5-002 induced extensive apoptosis of prostate cancer cells, impaired growth of prostate cancer xenograft tumors, and induced cell death in patient-derived prostate cancers when tested ex vivo in explant organ cultures. Importantly, IST5-002 induced robust apoptotic death not only of imatinib-sensitive but also of imatinib-resistant CML cell lines and primary CML cells from patients. IST5-002 provides a lead structure for further chemical modifications for clinical development for Stat5a/b-driven solid tumors and hematologic malignancies. Mol Cancer Ther; 14(8); 1777–93. ©2015 AACR.


Molecular Cancer Therapeutics | 2015

Inhibition of Stat5a/b Enhances Proteasomal Degradation of Androgen Receptor Liganded by Antiandrogens in Prostate Cancer

David T. Hoang; Lei Gu; Zhiyong Liao; Feng Shen; Pooja Talati; Mateusz Koptyra; Shyh-Han Tan; Elyse Ellsworth; Shilpa Gupta; Heather L. Montie; Ayush Dagvadorj; Saija Savolainen; Benjamin E. Leiby; Tuomas Mirtti; Diane E. Merry; Marja T. Nevalainen

Although poorly understood, androgen receptor (AR) signaling is sustained despite treatment of prostate cancer with antiandrogens and potentially underlies development of incurable castrate-resistant prostate cancer. However, therapies targeting the AR signaling axis eventually fail when prostate cancer progresses to the castrate-resistant stage. Stat5a/b, a candidate therapeutic target protein in prostate cancer, synergizes with AR to reciprocally enhance the signaling of both proteins. In this work, we demonstrate that Stat5a/b sequesters antiandrogen-liganded (MDV3100, bicalutamide, flutamide) AR in prostate cancer cells and protects it against proteasomal degradation in prostate cancer. Active Stat5a/b increased nuclear levels of both unliganded and antiandrogen-liganded AR, as demonstrated in prostate cancer cell lines, xenograft tumors, and clinical patient-derived prostate cancer samples. Physical interaction between Stat5a/b and AR in prostate cancer cells was mediated by the DNA-binding domain of Stat5a/b and the N-terminal domain of AR. Moreover, active Stat5a/b increased AR occupancy of the prostate-specific antigen promoter and AR-regulated gene expression in prostate cancer cells. Mechanistically, both Stat5a/b genetic knockdown and antiandrogen treatment induced proteasomal degradation of AR in prostate cancer cells, with combined inhibition of Stat5a/b and AR leading to maximal loss of AR protein and prostate cancer cell viability. Our results indicate that therapeutic targeting of AR in prostate cancer using antiandrogens may be substantially improved by targeting of Stat5a/b. Mol Cancer Ther; 14(3); 713–26. ©2014 AACR.


Clinical Cancer Research | 2010

Abstract B20: Ex vivo prostate cancer organ culture: Development and validation of an experimental model system to test novel targeted therapies in prostate cancer and offer a patient-tailored approach

Shilpa Gupta; Lei Gu; Zhiyong Liao; Peter Mc Cue; Edourd J. Trabulsi; Marja T. Nevalainen

Introduction: There is no cure for metastatic prostate cancer and novel targeted therapies are urgently required to improve survival and offer a patient-tailored approach. The purpose of this study is to validate the ex vivo organ culture model system of clinical human prostate cancers from patients for analysis of the effects of novel therapeutic agents in prostate cancer. In the studies presented here, we exploited ex vivo organ cultures to target the Jak2-Stat5a/b signaling pathway by a pharmacologic inhibitor of Stat5a/b IST5-002 in clinical prostate cancers. Development and validation of this ex vivo organ culture system may provide a more physiologic preclinical model than cell cultures or xenograft tumors to establish the efficacy of various novel targeted therapies and provide information on the probability of a drug9s overall success in advancing through the drug development stages. In addition, ex vivo organ cultures may provide a tool to identify patients specifically sensitive to a given targeted therapy. Experimental Procedures: Organ cultures of prostate cancers from 10 patients undergoing radical prostatectomy were started within 2 hours of surgery. Tissue explants were cultured in a medium containing the pharmacologic inhibitor of Stat5a/b IST5-002 or the control at various concentrations starting at 10 µM for 7 days and analyzed for viability by hematoxylin-eosin staining and “in situ DNA end-labeling” assay. Stat5a/b activation was analyzed by immunohistochemistry for phosphorylated and nuclear Stat5a/b. In addition, qPCR was performed for the downstream target genes of Stat5a/b. Results: The overall morphology of malignant human prostate was well maintained in all the ten ex vivo organ cultures. All the prostate cancers analyzed by organ cultures in this study showed an effective dose-dependant drug response to the pharmacologic inhibitor of Stat5a/b by decreased Stat5 phosphorylation and downregulation of Stat5-target genes in cultured prostate cancers demonstrating the inhibitory effect of IST5-002 on the Jak-Stat5a/b signaling pathway. The minimum dose of the IST5-002 found to be effective was 25µM Conclusions: The pharmacologic inhibitor of Stat5a/b, IST5- 002 shows promising preclinical activity in clinical prostate cancers demonstrated by 3D-ex vivo organ cultures of clinical human prostate cancers as the experimental model system and would be a promising drug for use in a phase I clinical trial. Ex vivo organ culture system may serve as a tool to analyze the effects of various novel targeted therapies in human prostate cancer to supplement the data obtained from prostate cancer cell lines, and xenograft prostate tumors. Finally, the use of ex vivo prostate organ cultures from individual patients may help to select those patients who would respond to a given targeted therapy providing a patient-tailored approach. Citation Information: Clin Cancer Res 2010;16(14 Suppl):B20.


American Journal of Translational Research | 2011

Targeting transcription factor Stat5a/b as a therapeutic strategy for prostate cancer

Zhiyong Liao; Marja T. Nevalainen


Journal of Clinical Oncology | 2011

Identification of a small-molecule inhibitor of Stat5a/b through structure-based screen for therapy development for prostate cancer.

Zhiyong Liao; Lei Gu; F. Shen; A. Dagvadorj; Shilpa Gupta; Nagarajan Pattabiraman; Marja T. Nevalainen; Edouard J. Trabulsi; Leonard G. Gomella; Peter McCue

Collaboration


Dive into the Zhiyong Liao's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lei Gu

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Shilpa Gupta

University of Minnesota

View shared research outputs
Top Co-Authors

Avatar

Ayush Dagvadorj

Thomas Jefferson University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Peter McCue

Thomas Jefferson University

View shared research outputs
Top Co-Authors

Avatar

Leonard G. Gomella

Thomas Jefferson University

View shared research outputs
Top Co-Authors

Avatar

Benjamin E. Leiby

Thomas Jefferson University

View shared research outputs
Top Co-Authors

Avatar

David T. Hoang

Thomas Jefferson University

View shared research outputs
Top Co-Authors

Avatar

Shyh-Han Tan

Uniformed Services University of the Health Sciences

View shared research outputs
Researchain Logo
Decentralizing Knowledge