Network


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

Hotspot


Dive into the research topics where Qiang Shen is active.

Publication


Featured researches published by Qiang Shen.


Cancers | 2014

Transcription Factor STAT3 as a Novel Molecular Target for Cancer Prevention

Ailian Xiong; Zhengduo Yang; Yicheng Shen; Jia Zhou; Qiang Shen

Signal Transducers and Activators of Transcription (STATs) are a family of transcription factors that regulate cell proliferation, differentiation, apoptosis, immune and inflammatory responses, and angiogenesis. Cumulative evidence has established that STAT3 has a critical role in the development of multiple cancer types. Because it is constitutively activated during disease progression and metastasis in a variety of cancers, STAT3 has promise as a drug target for cancer therapeutics. Recently, STAT3 was found to have an important role in maintaining cancer stem cells in vitro and in mouse tumor models, suggesting STAT3 is integrally involved in tumor initiation, progression and maintenance. STAT3 has been traditionally considered as nontargetable or undruggable, and the lag in developing effective STAT3 inhibitors contributes to the current lack of FDA-approved STAT3 inhibitors. Recent advances in cancer biology and drug discovery efforts have shed light on targeting STAT3 globally and/or specifically for cancer therapy. In this review, we summarize current literature and discuss the potential importance of STAT3 as a novel target for cancer prevention and of STAT3 inhibitors as effective chemopreventive agents.


Oncogene | 2008

The AP-1 transcription factor regulates breast cancer cell growth via cyclins and E2F factors

Qiang Shen; Ivan P. Uray; Yi Li; Tibor Krisko; Tracy Strecker; Heetae Kim; Powel H. Brown

The activating protein-1 (AP-1) transcription factor transduces growth signals through signal transduction pathways to the nucleus, leading to the expression of genes involved in growth and malignant transformation in many cell types. We have previously shown that overexpression of a dominant negative form of the cJun proto-oncogene, a cJun dominant negative mutant (Tam67), blocks AP-1 transcriptional activity, induces a G1 cell cycle block and inhibits breast cancer cell growth in vitro and in vivo. We found that AP-1 blockade by Tam67 in MCF-7 breast cancer cells downregulates cyclin D1 transcriptional activity by at least two mechanisms: by suppressing transcription at the known AP-1 binding site (−934/−928) and by suppressing growth factor-induced expression through suppressing E2F activation at the E2F-responsive site (−726/−719). AP-1 blockade also led to reduced expression of E2F1 and E2F2, but not E2F4, at the mRNA and protein levels. Chromatin immunoprecipitation and supershift assays demonstrated that AP-1 blockade caused decreased binding of E2F1 protein to the E2F site in the cyclin D1 promoter. We also found that Tam67 suppressed the expression of the E2F1 dimerizing partner, DP1 and E2F-upregulated cell cycle genes (cyclins E, A, B and D3) and enhanced the expression of E2F-downregulated cell cycle genes (cyclins G2 and I). Reduced expression of other E2F-regulated genes was also seen with AP-1 blockade and E2F suppression. Thus, the AP-1 factor regulates the expression of cyclin D and E2F (the latter in turn regulates E2F-downstream genes), leading to cell cycle progression and breast cancer cell proliferation.


Journal of Mammary Gland Biology and Neoplasia | 2003

Novel Agents for the Prevention of Breast Cancer: Targeting Transcription Factors and Signal Transduction Pathways

Qiang Shen; Powel H. Brown

Transformation of breast cells occurs through loss or mutation of tumor suppressor genes, or activation or amplification of oncogenes, leading to deregulation of signal transduction pathways, abnormal amplification of growth signals, and aberrant expression of genes that ultimately transform the cells into invasive cancer. The goal of cancer preventive therapy, or “chemoprevention,” is to eliminate premalignant cells or to block the progression of normal cells into cancer. Multiple alterations in signal pathways and transcription factors are observed in mammary gland tumorigenesis. In particular, estrogen receptor (ER) deregulation plays a critical role in breast cancer development and progress, and targeting ER with selective ER modulators (SERMs) has achieved significant reduction of breast cancer incidence in women at high risk for breast cancer. However, not all breast cancer is prevented by SERMs, because 30–40% of the tumors are ER-negative. Other receptors for retinoids, vitamin D analogs and peroxisome proliferator–activiator, along with transcription factors such as AP-1, NF-κB, and STATs (signal transducers and activators of transcription) affect breast tumorigenesis. This is also true for the signal transduction pathways, for example cyclooxygenase 2 (Cox-2), HER2/neu, mitogen-activated protein kinase (MAPK), and PI3K/Akt. Therefore, proteins in pathways that are altered during the process of mammary tumorigenesis may be promising targets of future chemopreventive drugs. Many newly-developed synthetic or natural compounds/agents are now under testing in preclinical studies and clinical trials. Receptor selective retinoids, receptor tyrosine kinase inhibitors (TKIs), SERMs, Cox-2 inhibitors, and others are some of the promising novel agents for the prevention of breast cancer. The chemopreventive activity of these agents and other novel signal transduction inhibitors are discussed in this chapter.


Oncogene | 2004

AP-1 blockade in breast cancer cells causes cell cycle arrest by suppressing G1 cyclin expression and reducing cyclin-dependent kinase activity

Yongmin Liu; Chunhua Lu; Qiang Shen; Debbie Munoz-Medellin; Heetae Kim; Powel H. Brown

The AP-1 transcription factor is a central component of signal transduction pathways in many cells, although the exact role of AP-1 in controlling cell growth and malignant transformation is unknown. We have previously shown that AP-1 complexes are activated by peptide and steroid growth factors in both normal and malignant breast cells, and that blocking AP-1 by overexpressing a dominant-negative form of cJun (cJun-DN, TAM67) inhibits breast cancer cell growth both in vivo and in vitro. We hypothesized that TAM67 inhibits cell growth by altering the expression of cell cycle regulatory proteins, thus causing a cell cycle block. In the present study, we used clones of MCF7 breast cancer cells that express TAM67 under the control of an inducible promoter. First, we determined the effect of AP-1 blockade on cell growth, then we performed 3H-thymidine incorporation and flow cytometry assays to investigate whether TAM67 inhibits the cell cycle. We observed that in the presence of serum TAM67 inhibited cell growth and caused a block in the G1 phase of the cell cycle. Next, we performed Western-blotting and CDK kinase assays to determine the effects of TAM67 on retinoblastoma (Rb) phosphorylation, the expression of cell cycle regulatory proteins, and CDK activity. We discovered that TAM67 inhibited Rb phosphorylation and reduced E2F activity. We also found that TAM67 decreased the expression of D and E cyclins, reduced CDK2 and CDK4 activity, and increased the CDK inhibitor p27. The studies of gene expression at the RNA level showed that TAM67 decreased cyclin Ds mRNA expression. Our study suggests that in the presence of serum, TAM67 inhibits breast cancer growth predominantly by inducing inhibitors of cyclin-dependent kinases (such as p27) and by reducing the expression of the cyclins involved in transitioning from G1 into S phase of the cell cycle. These studies lay the foundation for future attempt to develop new agents for the treatment and prevention of breast cancer.


Journal of Medicinal Chemistry | 2013

Novel Nitrogen-Enriched Oridonin Analogues with Thiazole-Fused A‑Ring: Protecting Group-Free Synthesis, Enhanced Anticancer Profile, and Improved Aqueous Solubility

Chunyong Ding; Yusong Zhang; Haijun Chen; Zhengduo Yang; Christopher Wild; Lili Chu; Huiling Liu; Qiang Shen; Jia Zhou

Oridonin (1), a complex ent-kaurane diterpenoid isolated from the traditional Chinese herb Isodon rubescens , has demonstrated great potential in the treatment of various human cancers due to its unique and safe anticancer pharmacological profile. Nevertheless, the clinical development of oridonin for cancer therapy has been hampered by its relatively moderate potency, limited aqueous solubility, and poor bioavailability. Herein, we report the concise synthesis of a series of novel nitrogen-enriched oridonin derivatives with thiazole-fused A-ring through an efficient protecting group-free synthetic strategy. Most of them, including compounds 7-11, 13, and 14, exhibited potent antiproliferative effects against breast, pancreatic, and prostate cancer cells with low micromolar to submicromolar IC50 values as well as markedly enhanced aqueous solubility. These new analogues obtained by rationally modifying the natural product have been demonstrated not only to significantly induce the apoptosis and suppress growth of triple-negative MDA-MB-231 breast cancer both in vitro and in vivo but also effective against drug-resistant ER-positive MCF-7 clones.


Oncogene | 2005

cFos is critical for MCF-7 breast cancer cell growth

Chunhua Lu; Qiang Shen; Elizabeth DuPré; Heetae Kim; Susan G. Hilsenbeck; Powel H. Brown

The activating protein-1 (AP-1) transcription factor is a converging point of multiple signal transduction pathways in many cells. We have previously demonstrated that overexpressing Tam67, a dominant-negative (DN) form of cJun, blocks AP-1 activity and inhibits breast cancer cell growth. We hypothesized that Tam67 forms dimers with other AP-1 proteins to suppress the growth of breast cancer cells. In the present study, we used immunoprecipitation-Western blotting to demonstrate that Tam67 binds all Jun and Fos proteins in breast cancer cells. In addition, we used two variants of the Tam67 mutant to investigate whether Jun or Fos protein was required for breast cancer cell growth. We created a Tam/Fos mutant in which the cJun dimerization domain was replaced by the cFos dimerization domain, and a Tam/Squelcher mutant in which the cJun dimerization domain was deleted. We then isolated MCF-7 cell lines that stably expressed these cJun-DN mutants under the control of an inducible promoter. Using AP-1-dependent reporter assays, we observed that Tam67 and Tam/Fos mutants inhibited AP-1 transcriptional activity, while the Tam/Squelcher mutant did not. We then determined whether Tam/Fos or Tam/Squelcher inhibited breast cell growth as well as Tam67. We found that while Tam67 repressed cell growth, neither Tam/Fos nor Tam/Squelcher mutant affected cell growth. These results indicate that Tam67 likely inactivates Fos family member proteins to suppress breast cancer cell growth. Finally, we performed antisense experiments to knock down the expression of individual family members (cJun or cFos). Our results demonstrated that antisense cFos inhibited breast cancer cell proliferation and colony formation, while antisense cJun did not. These results suggest that Tam67 suppresses breast cancer cell growth by interacting with Fos family members, specifically with cFos, to produce an inactive AP-1 complex.


Journal of the National Cancer Institute | 2009

Effect of Lapatinib on the Development of Estrogen Receptor–Negative Mammary Tumors in Mice

Tracy Strecker; Qiang Shen; Yun Zhang; Jamal Hill; Yuxin Li; Chunyu Wang; Hee Tae Kim; Tona M. Gilmer; Krystal Sexton; Susan G. Hilsenbeck; C. Kent Osborne; Powel H. Brown

Lapatinib, a selective orally available inhibitor of epidermal growth factor receptor (EGFR) and ErbB2 receptor tyrosine kinases, is a promising agent for the treatment of breast cancer. We examined the effect of lapatinib on the development of mammary tumors in MMTV-erbB2 transgenic mice, which express wild-type ErbB2 under the control of the mouse mammary tumor virus promoter and spontaneously develop estrogen receptor (ER)-negative and ErbB2-positive mammary tumors by 14 months of age. Mice were treated from age 3 months to age 15 months with vehicle (n = 17) or lapatinib (30 or 75 mg/kg body weight; n = 16 mice per group) by oral gavage twice daily (6 d/wk). All statistical tests were two-sided. By 328 days after the start of treatment, all 17 (100%) of the vehicle-treated mice vs five (31%) of the 16 mice treated with high-dose lapatinib developed mammary tumors (P < .001). Among MMTV-erbB2 mice treated for 5 months (n = 20 mice per group), those treated with lapatinib had fewer premalignant lesions and noninvasive cancers in their mammary glands than those treated with vehicle (P = .02). Lapatinib also effectively blocked epidermal growth factor-induced signaling through the EGFR and ErbB2 receptors, suppressed cyclin D1 and epiregulin mRNA expression, and stimulated p27 mRNA expression in human mammary epithelial cells and in mammary epithelial cells from mice treated for 5 months with high-dose lapatinib. Thus, cyclin D1, epiregulin, and p27 may represent useful biomarkers of lapatinib response in patients. These data suggest that lapatinib is a promising agent for the prevention of ER-negative breast cancer.


Clinical Cancer Research | 2007

The Rexinoid LG100268 prevents the development of preinvasive and invasive estrogen receptor negative tumors in MMTV-erbB2 mice.

Yuxin Li; Yun Zhang; Jamal Hill; Qiang Shen; Hee Tae Kim; Xiao Chun Xu; Susan G. Hilsenbeck; Reid P. Bissonnette; William W. Lamph; Powel H. Brown

Purpose: To test whether a novel rexinoid, LG100268, prevents the development of preinvasive and invasive estrogen receptor–negative mammary tumorigenesis in MMTV-erbB2 mice. Experimental Design: For invasive breast cancer prevention, MMTV-erbB2 mice were treated with daily gastric gavage of vehicle, LG100268 (10 mg/kg), or LG100268 (100 mg/kg) for long term starting at 3 months of age. For preinvasive lesion study, mice were treated with daily gastric gavage of vehicle or LG100268 (100 mg/kg) for 4 months. Results: Long-term treatment with LG100268 significantly prevented invasive mammary tumor development. Median time (age) to tumor development was delayed from 217 days in vehicle group to 357 days in low-dose group. In high-dose group, only 2 of 20 mice developed tumors after 430 days of treatment. Short-term treatment of LG100268 significantly prevented the development of preinvasive mammary lesions including hyperplasia and ductal carcinoma in situ. The cancer prevention effect was associated with reduced expression of Ki67 and cyclin D1 in mammary glands by >80%. Conclusion: Rexinoid LG100268 is an effective chemopreventive agent in preventing the development of both malignant and premalignant mammary lesions in MMTV-erbB2 mice.


European Journal of Medicinal Chemistry | 2013

Fragment-based drug design and identification of HJC0123, a novel orally bioavailable STAT3 inhibitor for cancer therapy

Haijun Chen; Zhengduo Yang; Chunyong Ding; Lili Chu; Yusong Zhang; Kristin Terry; Huiling Liu; Qiang Shen; Jia Zhou

Fragment-based drug design (FBDD) is a promising approach for the generation of lead molecules with enhanced activity and especially drug-like properties against therapeutic targets. Herein, we report the fragment-based drug design, systematic chemical synthesis and pharmacological evaluation of novel scaffolds as potent anticancer agents by utilizing six privileged fragments from known STAT3 inhibitors. Several new molecules such as compounds 5, 12, and 19 that may act as advanced chemical leads have been identified. The most potent compound 5 (HJC0123) has demonstrated to inhibit STAT3 promoter activity, downregulate phosphorylation of STAT3, increase the expression of cleaved caspase-3, inhibit cell cycle progression and promote apoptosis in breast and pancreatic cancer cells with low micromolar to nanomolar IC50 values. Furthermore, compound 5 significantly suppressed estrogen receptor (ER)-negative breast cancer MDA-MB-231 xenograft tumor growth in vivo (p.o.), indicating its great potential as an efficacious and orally bioavailable drug candidate for human cancer therapy.


British Journal of Cancer | 2008

The rexinoid, bexarotene, prevents the development of premalignant lesions in MMTV-erbB2 mice

Yi Li; Yun Zhang; Jamal Hill; Heetae Kim; Qiang Shen; Reid P. Bissonnette; William W. Lamph; Powel H Brown

Retinoids, vitamin A analogues that bind to retinoic acid receptor (RAR) or retinoid X receptor (RXR), play important roles in regulating cell proliferation, apoptosis, and differentiation. Recently, RXR-selective ligands, also referred to as rexinoids, have been investigated as potential chemopreventive agents for breast cancer. Our previous studies demonstrated that the rexinoid bexarotene significantly prevented ER-negative mammary tumourigenesis with less toxicity than naturally occurring retinoids in animal models. To determine whether bexarotene prevents cancer at the early stages during the multistage process of mammary carcinogenesis, we treated MMTV-erbB2 mice with bexarotene for 2 or 4 months. The development of preinvasive mammary lesions such as hyperplasias and carcinoma-in-situ was significantly inhibited. This inhibition was associated with reduced proliferation, but no induction of apoptosis. We also examined the regulation of a number of rexinoid-modulated genes including critical growth and cell cycle regulating genes using breast cell lines and mammary gland samples from mice treated with rexinoids. We showed that two of these genes (DHRS3 and DEC2) were modulated by bexarotene both in vitro and in vivo. Identification of these rexinoid-modulated genes will help us understand the mechanism by which rexinoid prevents cancer. Such rexinoid-regulated genes also represent potential biomarkers to assess the response of rexinoid treatment in clinical trials.

Collaboration


Dive into the Qiang Shen's collaboration.

Top Co-Authors

Avatar

Jia Zhou

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Na Ye

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Chunyong Ding

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Haijun Chen

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Zhengduo Yang

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Christopher Wild

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Powel H. Brown

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Ye Ding

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Haiying Chen

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Yusong Zhang

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge