Network


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

Hotspot


Dive into the research topics where Guoqing Qian is active.

Publication


Featured researches published by Guoqing Qian.


Molecular Cancer Therapeutics | 2014

Combination of Anti-HER3 Antibody MM-121/SAR256212 and Cetuximab Inhibits Tumor Growth in Preclinical Models of Head and Neck Squamous Cell Carcinoma

Ning Jiang; Dongsheng Wang; Zhongliang Hu; Hyung Ju C. Shin; Guoqing Qian; Mohammad Aminur Rahman; Hongzheng Zhang; A.R.M. Ruhul Amin; Sreenivas Nannapaneni; Xiaojing Wang; Zhengjia Chen; Gabriela Garcia; Gavin MacBeath; Dong M. Shin; Fadlo R. Khuri; Jun Ma; Zhuo Georgia Chen; Nabil F. Saba

The EGFR monoclonal antibody cetuximab is the only approved targeted agent for treating head and neck squamous cell carcinoma (HNSCC). Yet resistance to cetuximab has hindered its activity in this disease. Intrinsic or compensatory HER3 signaling may contribute to cetuximab resistance. To investigate the therapeutic benefit of combining MM-121/SAR256212, an anti-HER3 monoclonal antibody, with cetuximab in HNSCC, we initially screened 12 HNSCC cell lines for total and phosphorylated levels of the four HER receptors. We also investigated the combination of MM-121 with cetuximab in preclinical models of HNSCC. Our results revealed that HER3 is widely expressed and activated in HNSCC cell lines. MM-121 strongly inhibited phosphorylation of HER3 and AKT. When combined with cetuximab, MM-121 exerted a more potent antitumor activity through simultaneously inhibiting the activation of HER3 and EGFR and consequently the downstream PI3K/AKT and ERK pathways in vitro. Both high and low doses of MM-121 in combination with cetuximab significantly suppressed tumor growth in xenograft models and inhibited activations of HER3, EGFR, AKT, and ERK in vivo. Our work is the first report on this new combination in HNSCC and supports the concept that HER3 inhibition may play an important role in future therapy of HNSCC. Our results open the door for further mechanistic studies to better understand the role of HER3 in resistance to EGFR inhibitors in HNSCC. Mol Cancer Ther; 13(7); 1826–36. ©2014 AACR.


Cancer | 2015

Heregulin and HER3 are prognostic biomarkers in oropharyngeal squamous cell carcinoma

Guoqing Qian; Ning Jiang; Dongsheng Wang; Scott Newman; Sungjin Kim; Zhengjia Chen; Gabriela Garcia; Gavin MacBeath; Dong M. Shin; Fadlo R. Khuri; Zhuo Georgia Chen; Nabil F. Saba

Although heregulin and human epidermal growth factor receptor 3 (HER3) are frequently expressed at high levels in patients with head and neck cancer, their prognostic value remains unclear. The authors explored the prognostic significance of heregulin/HER3 expression in patients with oropharyngeal squamous cell carcinoma (OPSCC), taking into account other HER family members as well as p16 status.


Clinical Cancer Research | 2017

HER3 Targeting Sensitizes HNSCC to Cetuximab by Reducing HER3 Activity and HER2/HER3 Dimerization: Evidence from Cell Line and Patient-Derived Xenograft Models

Dongsheng Wang; Guoqing Qian; Hongzheng Zhang; Kelly R. Magliocca; Sreenivas Nannapaneni; A.R.M. Ruhul Amin; Michael R. Rossi; Mihir Patel; Mark W. El-Deiry; J. Trad Wadsworth; Zhengjia Chen; Fadlo R. Khuri; Dong M. Shin; Nabil F. Saba; Zhuo Georgia Chen

Purpose: Our previous work suggested that HER3 inhibition sensitizes head and neck squamous cell carcinoma (HNSCC) to EGFR inhibition with cetuximab. This study aimed to define the role of HER3 in cetuximab resistance and the antitumor mechanisms of EGFR/HER3 dual targeting in HNSCC. Experimental Design: We treated cetuximab-resistant HNSCC UMSCC1-C and parental UMSCC1-P cell lines with anti-EGFR antibody cetuximab, anti-HER3 antibody MM-121, and their combination. We assessed activities of HER2, HER3, and downstream signaling pathways by Western blotting and cell growth by sulforhodamine B (SRB) and colony formation assays. HER3-specific shRNA was used to confirm the role of HER3 in cetuximab response. The combined efficacy and alterations in biomarkers were evaluated in UMSCC1-C xenograft and patient-derived xenograft (PDX) models. Results: Cetuximab treatment induced HER3 activation and HER2/HER3 dimerization in HNSCC cell lines. Combined treatment with cetuximab and MM-121 blocked EGFR and HER3 activities and inhibited the PI3K/AKT and ERK signaling pathways and HNSCC cell growth more effectively than each antibody alone. HER3 knockdown reduced HER2 activation and resensitized cells to cetuximab. Cetuximab-resistant xenografts and PDX models revealed greater efficacy of dual EGFR and HER3 inhibition compared with single antibodies. In PDX tissue samples, cetuximab induced HER3 expression and MM-121 reduced AKT activity. Conclusions: Clinically relevant PDX models demonstrate that dual targeting of EGFR and HER3 is superior to EGFR targeting alone in HNSCC. Our study illustrates the upregulation of HER3 by cetuximab as one mechanism underlying resistance to EGFR inhibition in HNSCC, supporting further clinical investigations using multiple targeting strategies in patients who have failed cetuximab-based therapy. Clin Cancer Res; 23(3); 677–86. ©2016 AACR.


Cancer | 2015

Human papillomavirus 16 oncoprotein regulates the translocation of β‐catenin via the activation of epidermal growth factor receptor

Zhongliang Hu; Susan Muller; Guoqing Qian; Jing Xu; Sungjin Kim; Zhengjia Chen; Ning Jiang; Dongsheng Wang; Hongzheng Zhang; Nabil F. Saba; Dong M. Shin; Zhuo Georgia Chen

To understand the mechanism of frequent and early lymph node metastasis in high‐risk human papillomavirus (HPV)–associated oropharyngeal squamous cell carcinoma (OPSCC), this study investigated whether β‐catenin is regulated by the HPV oncoprotein and contributes to OPSCC metastasis.Background To understand the mechanism of frequent and early lymph node metastasis in high risk human papillomavirus (HPV)-associated oropharyngeal squamous cell carcinoma (OPSCC), we investigated whether β-catenin is regulated by HPV oncoprotein and contributes to OPSCC metastasis.


Clinical Cancer Research | 2017

Overcoming acquired resistance to AZD9291, a third generation EGFR inhibitor, through modulation of MEK/ERK-dependent Bim and Mcl-1 degradation

Puyu Shi; You-Take Oh; Liang Deng; Guojing Zhang; Guoqing Qian; Shuo Zhang; Hui Ren; Grant Wu; Benjamin Legendre; Emily Anderson; Suresh S. Ramalingam; Taofeek K. Owonikoko; Mingwei Chen; Shi-Yong Sun

Purpose: The mechanisms accounting for anticancer activity of AZD9291 (osimertinib or TAGRISSO), an approved third-generation EGFR inhibitor, in EGFR-mutant non–small cell lung cancer (NSCLC) cells and particularly for the subsequent development of acquired resistance are unclear and thus are the focus of this study. Experimental Design: AZD9219-resistant cell lines were established by exposing sensitive cell lines to AZD9291. Protein alterations were detected with Western blotting. Apoptosis was measured with annexin V/flow cytometry. Growth-inhibitory effects of tested drugs were evaluated in vitro with cell number estimation and colony formation assay and in vivo with mouse xenograft models. Protein degradation was determined by comparing protein half-lives and inhibiting proteasome. Gene knockdown were achieved with siRNA or shRNA. Results: AZD9291 potently induced apoptosis in EGFR-mutant NSCLC cell lines, in which ERK phosphorylation was suppressed accompanied with Bim elevation and Mcl-1 reduction likely due to enhanced Mcl-1 degradation and increased Bim stability. Blocking Bim elevation by gene knockdown or enforcing Mcl-1 expression attenuated or abolished AZD9291-induced apoptosis. Moreover, AZD9291 lost its ability to modulate Bim and Mcl-1 levels in AZD9291-resistant cell lines. The combination of a MEK inhibitor with AZD9291 restores the sensitivity of AZD9291-resistant cells including those with C797S mutation to undergo apoptosis and growth regression in vitro and in vivo. Conclusions: Modulation of MEK/ERK-dependent Bim and Mcl-1 degradation critically mediates sensitivity and resistance of EGFR-mutant NSCLC cells to AZD9291 and hence is an effective strategy to overcome acquired resistance to AZD9291. Clin Cancer Res; 23(21); 6567–79. ©2017 AACR.


Oncotarget | 2016

Biomarker quantification by multiplexed quantum dot technology for predicting lymph node metastasis and prognosis in head and neck cancer

Zhongliang Hu; Guoqing Qian; Susan Muller; Jing Xu; Nabil F. Saba; Sungjin Kim; Zhengjia Chen; Ning Jiang; Dongsheng Wang; Hongzheng Zhang; Kristin Lane; Clifford C. Hoyt; Dong M. Shin; Zhuo Georgia Chen

Purpose To predict lymph node metastasis and prognosis in head and neck squamous cell carcinoma (HNSCC). Results The combination of membranous E-cadherin and membranous epidermal growth factor receptor (EGFR) quantified by QD technology with age, gender, and grade had greater predictive power than any of the single biomarkers or the two combined biomarkers quantified by conventional immunohistochemistry (IHC). The predictive power of this model was validated in another independent sample set; the predictive sensitivity of this model for LNM was 87.5%, with specificity up to 97.4%, and accuracy 92.9%. Furthermore, a higher membranous E-cadherin level was significantly correlated with better overall and disease-free survival (OS, DFS; P = 0.002, 0.033, respectively), while lower cytoplasmic vimentin and membranous EGFR levels were significantly correlated with better OS (P = 0.016 and 0.021, respectively). The combined biomarkers showed a stronger prognostic value for OS and DFS than any of the single biomarkers. Methods Multiplexed quantum dots (QDs) were used to simultaneously label E-cadherin, vimentin, and EGFR with β-actin as an internal control. Primary tissue samples from 97 HNSCC patients, 49 with and 48 without LNM were included in the training set. Levels of membranous E-cadherin, cytoplasmic vimentin, and membranous EGFR were quantified by InForm software and correlated with clinical characteristics. Conclusions Multiplexed subcellular QD quantification of EGFR and E-cadherin is a potential strategy for the prediction of LNM, DFS, and OS of HNSCC patients.


Oncotarget | 2017

Phenformin enhances the therapeutic effect of selumetinib in KRAS-mutant non-small cell lung cancer irrespective of LKB1 status

Jun Zhang; Sreenivas Nannapaneni; Dongsheng Wang; Fakeng Liu; Xu Wang; Rui Jin; Xiuju Liu; Mohammad Aminur Rahman; Xianghong Peng; Guoqing Qian; Zhuo Georgia Chen; Kwok-Kin Wong; Fadlo R. Khuri; Wei Zhou; Dong M. Shin

MEK inhibition is potentially valuable in targeting KRAS-mutant non-small cell lung cancer (NSCLC). Here, we analyzed whether concomitant LKB1 mutation alters sensitivity to the MEK inhibitor selumetinib, and whether the metabolism drug phenformin can enhance the therapeutic effect of selumetinib in isogenic cell lines with different LKB1 status. Isogenic pairs of KRAS-mutant NSCLC cell lines A549, H460 and H157, each with wild-type and null LKB1, as well as genetically engineered mouse-derived cell lines 634 (krasG12D/wt/p53-/-/lkb1wt/wt) and t2 (krasG12D/wt/p53-/-/lkb1-/-) were used in vitro to analyze the activities of selumetinib, phenformin and their combination. Synergy was measured and potential mechanisms investigated. The in vitro findings were then confirmed in vivo using xenograft models. The re-expression of wild type LKB1 increased phospho-ERK level, suggesting that restored dependency on MEK->ERK->MAPK signaling might have contributed to the enhanced sensitivity to selumetinib. In contrast, the loss of LKB1 sensitized cells to phenformin. At certain combination ratios, phenformin and selumetinib showed synergistic activity regardless of LKB1 status. Their combination reduced phospho-ERK and S6 levels and induced potent apoptosis, but was likely through different mechanisms in cells with different LKB1 status. Finally, in xenograft models bearing isogenic A549 cells, we confirmed that loss of LKB1 confers resistance to selumetinib, and phenformin significantly enhances the therapeutic effect of selumetinib. Irrespective of LKB1 status, phenformin may enhance the anti-tumor effect of selumetinib in KRAS-mutant NSCLC. The dual targeting of MEK and cancer metabolism may provide a useful strategy to treat this subset of lung cancer.


Journal of Oral Pathology & Medicine | 2016

A novel prediction model for human papillomavirus-associated oropharyngeal squamous cell carcinoma using p16 and subcellular β-catenin expression.

Guoqing Qian; Zhongliang Hu; Hong Xu; Susan Muller; Dongsheng Wang; Hongzheng Zhang; Sungjin Kim; Zhengjia Chen; Nabil F. Saba; Dong M. Shin; Andrew Y. Wang; Zhuo Georgia Chen

BACKGROUND p16 overexpression is a highly sensitive yet moderately specific biomarker for predicting human papillomavirus (HPV)-associated oropharyngeal squamous cell carcinoma (OPSCC). Nuclear β-catenin translocation has been linked to HPV-positive OPSCC. However, whether the strategy of combining β-catenin with p16 can better predict HPV-associated OPSCC remains unknown. METHODS We evaluated the expression of p16 and β-catenin (nuclear and membrane) by immunohistochemistry staining in 101 OPSCC tissues and HPV status by HPV DNA in situ hybridization. Logistic regression models were used to evaluate single or multiple biomarkers for HPV prediction. The prediction power, sensitivity, and specificity were determined by receiver operating characteristic (ROC) analyses. RESULTS Our data showed that upon univariate analysis, p16 and nuclear β-catenin were positively correlated with HPV status, while membrane β-catenin was inversely correlated with HPV status (P < 0.01). p16 showed the highest HPV predictive power, with area under the curve (AUC) of 0.9074 compared to 0.6762 for nuclear β-catenin and 0.7635 for membrane β-catenin, respectively, indicating differential accuracies for HPV prediction. Multivariable analysis showed that p16 was significantly correlated with HPV, while nuclear and membrane β-catenin showed marginal significance. The three-biomarker model was similarly sensitive (98.9% vs. 100%) but more specific (88.9% vs. 81%) than p16 alone, which also showed a good predictive value for overall (P = 0.0002) survival and disease-free (P = 0.0158) survival. CONCLUSION Our study suggests a novel model of combining p16 and subcellular β-catenin for prediction of HPV-associatred OPSCC, and this finding deserves further validation.


Oncogene | 2018

Monocyte chemotactic protein-induced protein-1 enhances DR5 degradation and negatively regulates DR5 activation-induced apoptosis through its deubiquitinase function

You-Take Oh; Guoqing Qian; Jiusheng Deng; Shi-Yong Sun

Monocyte chemotactic protein-induced protein-1 (MCPIP1; also called Regnase-1) encoded by the ZC3H12A gene critically regulates inflammatory responses and immune homeostasis primarily by RNase-dependent and -independent mechanisms. However, the relationship of MCPIP1 with apoptosis and cancer and the underlying mechanisms are largely unclear. The current study has demonstrated a previously uncovered connection between MCPIP1 and the negative regulation of death receptor 5 (DR5; also known as TRAIL-R2 or killer/DR5), a cell surface receptor for tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), which is produced endogenously by various immune cells such as T cells. Our findings have revealed that MCPIP1 decreases both total cellular and cell surface DR5, primarily through modulating DUB-mediated protein autophagic/lysosomal degradation. Suppression of MCPIP1 by gene knockdown induces the formation of death-induced signaling complex (DISC) and enhances TRAIL or DR5 activation-induced apoptosis in cancer cells. Moreover, we demonstrated an inverse correlation between MCPIP1 expression and DR5 expression/cell sensitivity to DR5 activation-induced apoptosis in cancer cells. Our findings warrant future investigation of the roles of negative regulation of DR5 by MCPIP1 in cancer and in T-cell immunity.


Cancer Research | 2015

Abstract 20: Honokiol radiosensitizes squamous cells carcinoma of head and neck by down-regulation of survivin

Xu Wang; Jonathan J. Beitler; Wen Huang; Guoqing Qian; Kelly R. Magliocca; Jun Zhang; Sreenivas Nannapaneni; Sungjin Kim; Zhengjia Chen; Saba F. Nabil; Zhuo Georgia Chen; Jack L. Arbiser; Dong M. Shin

Proceedings: AACR 106th Annual Meeting 2015; April 18-22, 2015; Philadelphia, PA Background: Overexpression of survivin is associated with resistance to radiotherapy and poor survival in many cancer types. However, in squamous cells carcinoma of head and neck (SCCHN), previous studies have revealed diverging results regarding survivins role in clinical outcome. In this study, we aim to 1) evaluate the clinical significance of survivin expression in SCCHN; 2) identify the function of survivin on DNA damage repair following ionizing radiation (IR) in SCCHN cells; 3) investigate the potential of using honokiol, a nature compound, to enhance radiation therapy through down regulation of survivin. Methods: Protein expression of survivin in SCCHN patient primary tissues (n = 99) was analyzed by immunohistochemistry and correlated with clinical parameters. IR-sensitive (JHU-022), IR -insensitive (PCI-15A, Tu212), and IR acquired resistance (PCI-15A-R) SCCHN cell lines were used to evaluate the role of survivin in DNA double strain break repairer following RT. The in vitro effects of and potential molecular targets of honokiol were also evaluated using those cell lines. Results: Overexpression of survivin is significantly associated with lymph node metastasis, worse overall survival and worse disease free survival in patients receiving radiation therapy (n = 65) and in all patients with SCCHN (n = 99). In SCCHN cells, depletion of survivin by siRNA inhibited, whereas overexpression of survivin increased, clonogenic survival following IR. IR induces nuclear accumulation of survivin and complexation with γ-H2X, and DNA-PKCs, suggesting survivin is involved in DNA damage response and repair induced by IR. Further, honokiol significantly reduces survivin expression and increases DNA damage induced by IR, as demonstrated by higher amount of DNA breaks and an increased amount of γ-H2X foci post IR. Conclusions: Survivin is a negative prognostic factor in SCCHN, and is involved in DNA damage response and repair induced by IR in SCCHN cells. Down regulation of survivin by honokiol enhances the efficacy of IR, and may provide a novel therapeutic approach to improve the efficacy of radiotherapy in SCCHN. (This research was supported by the National Cancer Institute award P50 CA128613, and GCC Distinguished Cancer Scholar to Dong M. Shin, Zhuo (Georgia) Chen, and Jonathan J Beitler) Citation Format: Xu Wang, Jonathan J. Beitler, Wen Huang, Guoqing Qian, Kelly Magliocca, Jun Zhang, Sreenivas Nannapaneni, Sungjin Kim, Zhengjia Chen, Saba F. Nabil, Zhuo G. Chen, Jack L. Arbiser, Dong M. Shin. Honokiol radiosensitizes squamous cells carcinoma of head and neck by down-regulation of survivin. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 20. doi:10.1158/1538-7445.AM2015-20

Collaboration


Dive into the Guoqing Qian's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sungjin Kim

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge