Network


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

Hotspot


Dive into the research topics where Xiuqing Li is active.

Publication


Featured researches published by Xiuqing Li.


Clinical Cancer Research | 2013

Monoclonal Antibody against Cell Surface GRP78 as a Novel Agent in Suppressing PI3K/AKT Signaling, Tumor Growth, and Metastasis

Ren Liu; Xiuqing Li; Wenming Gao; Yue Zhou; Shiuan Wey; Satyajit K. Mitra; Valery Krasnoperov; Dezheng Dong; Shuanglong Liu; Dan Li; Genyuan Zhu; Stan G. Louie; Peter S. Conti; Zibo Li; Amy S. Lee; Parkash S. Gill

Purpose: The ER chaperone GRP78 translocates to the surface of tumor cells and promotes survival, metastasis, and resistance to therapy. An oncogenic function of cell surface GRP78 has been attributed to the activation of the phosphoinositide 3-kinase (PI3K) pathway. We intend to use a novel anti-GRP78 monoclonal antibody (MAb159) to attenuate PI3K signaling and inhibit tumor growth and metastasis. Experimental Design: MAb159 was characterized biochemically. Antitumor activity was tested in cancer cell culture, tumor xenograft models, tumor metastasis models, and spontaneous tumor models. Cancer cells and tumor tissues were analyzed for PI3K activity. MAb159 was humanized and validated for diagnostic and therapeutic application. Results: MAb159 specifically recognized surface GRP78, triggered GRP78 endocytosis, and localized to tumors but not to normal organs in vivo. MAb159 inhibited tumor cell proliferation and enhanced tumor cell death both in vitro and in vivo. In MAb159-treated tumors, PI3K signaling was inhibited without compensatory MAPK pathway activation. Furthermore, MAb159 halted or reversed tumor progression in the spontaneous PTEN–loss-driven prostate and leukemia tumor models, and inhibited tumor growth and metastasis in xenograft models. Humanized MAb159, which retains high affinity, tumor specific localization, and the antitumor activity, was nontoxic in mice, and had desirable pharmacokinetics. Conclusions: GRP78-specific antibody MAb159 modulates the PI3K pathway and inhibits tumor growth and metastasis. Humanized MAb159 will enter human trials shortly. Clin Cancer Res; 19(24); 6802–11. ©2013 AACR.


BMC Cancer | 2010

Combination of Dll4/Notch and Ephrin-B2/EphB4 targeted therapy is highly effective in disrupting tumor angiogenesis.

Dusan Djokovic; Alexandre Trindade; Joana Gigante; Marina Badenes; Lilliana Silva; Ren Liu; Xiuqing Li; Ming Gong; Valery Krasnoperov; Parkash S. Gill; Antonio Duarte

BackgroundDll4/Notch and Ephrin-B2/EphB4 pathways play critical roles in tumor vessel development and maturation. This study evaluates the efficacy of the inhibition of both signaling pathways, alone and in combination, in reducing the growth of an autochthonous mouse tumor and assesses potential adverse effects.MethodsWe used the transgenic RIP1-Tag2 tumor model to study the effects of 1) inhibition of Dll4/Notch by either Dll4 allelic deletion or use of a soluble extracellular Dll4 (sDll4), 2) inhibition of Ephrin-B2/EphB4 signaling by a soluble extracellular EphB4 fused to albumin (sEphB4-Alb), and 3) inhibition of both pathways by sEphB4-Alb combined with either Dll4 allelic deletion or sDll4. To investigate adverse effects, we used inducible endothelial-specific Dll4 knock-out mice, treated with sEphB4-Alb, and carried out histopathological analysis.ResultsDll4 allele deletion or soluble Dll4 treatment resulted in increased tumor vessel density, reduced mural cell recruitment and vessel perfusion which resulted in reduced tumor size. The soluble EphB4 instead reduced vessel density and vessel perfusion, leading to reduction of tumor size. Greater efficacy was observed when sEphB4-Alb was combined with either Dll4 allele deletion or sDll4 in regards to tumor size, vessel perfusion and mural cell recruitment. Induced endothelial specific Dll4 loss-of-function caused hepatic vascular alterations, which were prevented by concomitant sEphB4-Alb treatment.ConclusionCombination targeting of Dll4/Notch and Ephrin-B2/EphB4 has potential for clinical investigation, providing cumulative efficacy and increased safety over Dll4/Notch inhibition alone.


American Journal of Pathology | 2010

Novel EphB4 Monoclonal Antibodies Modulate Angiogenesis and Inhibit Tumor Growth

Valery Krasnoperov; S. Ram Kumar; Eric J. Ley; Xiuqing Li; Jeffrey S. Scehnet; Ren Liu; Sergey Zozulya; Parkash S. Gill

EphB4 receptor tyrosine kinase and its cognate ligand EphrinB2 regulate induction and maturation of newly forming vessels. Inhibition of their interaction arrests angiogenesis, vessel maturation, and pericyte recruitment. In addition, EphB4 is expressed in the vast majority of epithelial cancers and provides a survival advantage to most. Here, we describe two anti-EphB4 monoclonal antibodies that inhibit tumor angiogenesis and tumor growth by two distinct pathways. MAb131 binds to fibronectin-like domain 1 and induces degradation of human EphB4, but not murine EphB4. MAb131 inhibits human endothelial tube formation in vitro and growth of human tumors expressing EphB4 in vivo. In contrast, MAb47 targets fibronectin-like domain 2 of both human and murine EphB4 and does not alter EphB4 receptor levels, but inhibits angiogenesis and growth of both EphB4-positive and EphB4-negative tumors in a mouse s.c. xenograft model. Combination of MAb47 and bevacizumab enhances the antitumor activity and induces tumor regression. Indeed, humanized antibodies hAb47 and hAb131 showed similar affinity for EphB4 and retained efficacy in the inhibition of primary tumor development and experimental metastasis.


Blood | 2010

KSHV induced notch components render endothelial and mural cell characteristics and cell survival

Ren Liu; Xiuqing Li; Anil Tulpule; Yue Zhou; Jeffrey S. Scehnet; Shaobing Zhang; Jong-Soo Lee; Preet M. Chaudhary; Jae Jung; Parkash S. Gill

Kaposi sarcoma-associated herpesvirus (KSHV) infection is essential to the development of Kaposi sarcoma (KS). Notch signaling is also known to play a pivotal role in KS cell survival and lytic phase entrance of KSHV. In the current study, we sought to determine whether KSHV regulates Notch components. KSHV-infected lymphatic endothelial cells showed induction of receptors Notch3 and Notch4, Notch ligands Dll4 and Jagged1, and activated Notch receptors in contrast to uninfected lymphatic endothelial cells. In addition, KSHV induced the expression of endothelial precursor cell marker (CD133) and mural cell markers (calponin, desmin, and smooth muscle alpha actin), suggesting dedifferentiation and trans-differentiation. Overexpression of latency proteins (LANA, vFLIP) and lytic phase proteins (RTA, vGPCR, viral interleukin-6) further supported the direct regulatory capacity of KSHV viral proteins to induce Notch receptors (Notch2, Notch3), ligands (Dll1, Dll4, Jagged1), downstream targets (Hey, Hes), and endothelial precursor CD133. Targeting Notch pathway with gamma-secretase inhibitor and a decoy protein in the form of soluble Dll4 inhibited growth of KSHV-transformed endothelial cell line. Soluble Dll4 was also highly active in vivo against KS tumor xenograft. It inhibited tumor cell growth, induced tumor cell death, and reduced vessel perfusion. Soluble Dll4 is thus a candidate for clinical investigation.


Blood | 2010

Induction, regulation, and biologic function of Axl receptor tyrosine kinase in Kaposi sarcoma

Ren Liu; Ming Gong; Xiuqing Li; Yue Zhou; Wenming Gao; Anil Tulpule; Preet M. Chaudhary; Jae Jung; Parkash S. Gill

Axl is an oncogenic receptor tyrosine kinase that plays multiple roles in tumorigenesis and metastasis of many cancers. This study is the first to demonstrate that Axl is induced in Kaposi sarcoma and Kaposi sarcoma herpesvirus (KSHV) transformed endothelial cells. Conditionally, expression of one KSHV latency protein vFLIP induces Axl expression in endothelial cells. This induction can be blocked by nuclear factor-kappaB inhibitor, consistent with the known vFLIP mechanism of action. KS cell lines lacking KSHV also have elevated Axl expression, which probably resulted from hypomethylation of AXL promoter. Axl activation activates downstream phosphoinositol-3 kinase signaling, and Axl knockdown by siRNA impairs phosphoinositol-3 kinase signaling. Furthermore, Axl knockdown inhibits KS cell growth and invasion. To explore the potential for translation of these findings, we generated monoclonal antibodies to block the biologic functions of Axl. MAb173, which induces receptor degradation, showed activity in vitro to inhibit KS cell invasion. Moreover, in vivo xenograft studies with KS cells with or without KSHV infection showed that MAb173 reduced tumor growth, increased tumor cell apoptosis, and markedly decreased Axl protein level in tumors. Axl thus has a potential role in KS pathogenesis and is a candidate for prognostic and therapeutic investigations.


Cancer Research | 2013

Critical Role for the Receptor Tyrosine Kinase EPHB4 in Esophageal Cancers

Rifat Hasina; Nathan M. Mollberg; Ichiro Kawada; Karun Mutreja; Geetanjali Kanade; Soheil Yala; Mosmi Surati; Ren Liu; Xiuqing Li; Yue Zhou; Benjamin D. Ferguson; Vidya Nallasura; Kenneth Cohen; Elizabeth Hyjek; Jeffery Mueller; Rajani Kanteti; Essam El Hashani; Dorothy Kane; Yutaka Shimada; Mark W. Lingen; Aliya N. Husain; Mitchell C. Posner; Irving Waxman; Victoria M. Villaflor; Mark K. Ferguson; Lyuba Varticovski; Everett E. Vokes; Parkash S. Gill; Ravi Salgia

Esophageal cancer incidence is increasing and has few treatment options. In studying receptor tyrosine kinases associated with esophageal cancers, we have identified EPHB4 to be robustly overexpressed in cell lines and primary tumor tissues. In total, 94 squamous cell carcinoma, 82 adenocarcinoma, 25 dysplasia, 13 Barrett esophagus, and 25 adjacent or unrelated normal esophageal tissues were evaluated by immunohistochemistry. EPHB4 expression was significantly higher in all the different histologic categories than in adjacent normal tissues. In 13 esophageal cancer cell lines, 3 of the 9 SCC cell lines and 2 of the 4 adenocarcinomas expressed very high levels of EPHB4. An increased gene copy number ranging from 4 to 20 copies was identified in a subset of the overexpressing patient samples and cell lines. We have developed a novel 4-nitroquinoline 1-oxide (4-NQO)-induced mouse model of esophageal cancer that recapitulates the EPHB4 expression in humans. A specific small-molecule inhibitor of EPHB4 decreased cell viability in a time- and dose-dependent manner in 3 of the 4 cell lines tested. The small-molecule inhibitor and an EPHB4 siRNA also decreased cell migration (12%-40% closure in treated vs. 60%-80% in untreated), with decreased phosphorylation of various tyrosyl-containing proteins, EphB4, and its downstream target p125FAK. Finally, in a xenograft tumor model, an EPHB4 inhibitor abrogated tumor growth by approximately 60% compared with untreated control. EphB4 is robustly expressed and potentially serves as a novel biomarker for targeted therapy in esophageal cancers.


Molecular Pharmaceutics | 2014

Design, synthesis and validation of Axl-targeted monoclonal antibody probe for microPET imaging in human lung cancer xenograft

Shuanglong Liu; Dan Li; Jiacong Guo; Nicolette Canale; Xiuqing Li; Ren Liu; Valery Krasnoperov; Parkash S. Gill; Peter S. Conti; Hong Shan; Zibo Li

Accumulating experimental evidence indicates that overexpression of the oncogenic receptor tyrosine kinase, Axl, plays a key role in the tumorigenesis and metastasis of various types of cancer. The objective of this study is to design a novel imaging probe based on the monoclonal antibody, h173, for microPET imaging of Axl expression in human lung cancer. A bifunctional chelator, DOTA, was conjugated to h173, followed by radiolabeling with 64Cu. The binding of DOTA-h173 to the Axl receptor was first evaluated by a cell uptake assay and flow cytometry analysis using human lung cancer cell lines. The probe 64Cu-DOTA-h173 was further evaluated by microPET imaging, and ex vivo histology studies in the Axl-positive A549 tumors. In vitro cellular study showed that Axl probe, 64Cu-DOTA-h173, was highly immuno-reactive with A549 cells. Western blot analysis confirmed that Axl is highly expressed in the A549 cell line. For microPET imaging, the A549 xenografts demonstrated a significantly higher 64Cu-DOTA-h173 uptake compared to the NCI-H249 xenograft (a negative control model). Furthermore, 64Cu-DOTA-h173 uptake in A549 is significantly higher than that of 64Cu-DOTA-hIgG. Immuno-fluorescence staining was consistent with the in vivo micro-PET imaging results. In conclusion, 64Cu-DOTA-h173 could be potentially used as a probe for noninvasive imaging of Axl expression, which could collect important information regarding tumor response to Axl-targeted therapeutic interventions.


PLOS ONE | 2014

The differential expression of EphB2 and EphB4 receptor kinases in normal bladder and in transitional cell carcinoma of the bladder.

Xiuqing Li; Wesley W. Choi; Rui Yan; Haiyang Yu; Valery Krasnoperov; S. Ram Kumar; Anne Schuckman; David J. Klumpp; Chong Xian Pan; David I. Quinn; Inderbir S. Gill; Parkash S. Gill; Ren Liu

Effective treatment of transitional cell carcinoma (TCC) of the bladder requires early diagnosis. Identifying novel molecular markers in TCC would guide the development of diagnostic and therapeutic targets. Ephrins mediate signals via tyrosine kinase activity that modulates diverse physiologic and developmental processes, and ephrins are increasingly implicated in carcinogenesis. The aim of our study was to examine the differential regulation of EphB4 and EphB2 in normal bladder and in TCC of the bladder in 40 patients undergoing radical cystectomy for curative intent. Immunostaining and Western blotting revealed that normal urothelium expresses EphB2 (20 of 24 cases, 83% of the time) not EphB4 (0 of 24 cases, 0%). In sharp contrast, TCC specimens show loss of EphB2 expression (0 of 34 cases, 0%) and gain of EphB4 expression (32 of 34, 94%). Furthermore, EphB4 signal strength statistically correlated with higher tumor stage, and trended toward the presence of carcinoma in situ (CIS). These results are confirmed by analysis of normal urothelial and tumor cell lines. EphB2 is not a survival factor in normal urothelium, while EphB4 is a survival factor in TCC. Treatment of bladder tumor xenograft with an EphB4 inhibitor sEphB4-HSA leads to 62% tumor regression and complete remission when combined with Bevacizumab. Furthermore, tissue analysis revealed that sEphB4-HSA led to increased apoptosis, decreased proliferation, and reduced vessel density, implicating direct tumor cell targeting as well as anti-angiogenesis effect. In summary loss of EphB2 and gain of EphB4 expression represents an inflection point in the development, growth and possibly progression of TCC. Therapeutic compounds targeting EphB4 have potential for diagnosing and treating TCC.


PLOS ONE | 2015

Identification of Androgen Receptor Splice Variants in the Pten Deficient Murine Prostate Cancer Model

Mengmeng Liang; Helty Adisetiyo; Xiuqing Li; Ren Liu; Parkash S. Gill; Pradip Roy-Burman; Jeremy Jones; David J. Mulholland

Androgen receptor (AR) variants are associated with resistance to anti androgen therapy both in human prostate cancer cell lines and clinical samples. These observations support the hypothesis that AR isoform accumulation is a consequence of selective therapeutic pressure on the full length AR. The Pten deficient prostate cancer model proceeds with well-defined kinetics including progression to castration resistant prostate cancer (CRPC). While surgical castration and enzalutamide treatments yield an initial therapeutic response, Pten-/-epithelia continue to proliferate yielding locally invasive primary tumor pathology. That most epithelium remains AR positive, but ligand independent, suggests the presence of oncogenic AR variants. To address this hypothesis, we have used a panel of recently described Pten-/- tumor cell lines derived from both from hormone intact (E4, E8) and castrated Pten mutants (cE1, cE2) followed by RACE PCR to identify and characterize three novel truncated, amino terminus containing AR variants (mAR-Va, b, c). Variants appear not only conserved throughout progression but are correlated with nearly complete loss of full length AR (AR-FL) at castrate androgen levels. The overexpression of variants leads to enhanced transcriptional activity of AR while knock down studies show reduced transcriptional output. Collectively, the identification of truncated AR variants in the conditional PTEN deletion model supports a role for maintaining the CRPC phenotype and provides further therapeutic applications of this preclinical model.


Cancer Research | 2012

Abstract 1861: EPHB4 as a potential therapeutic target for esophageal cancer

Rifat Hasina; Karun Mutreja; Ichiro Kawada; Rajani Kanteti; Qudsia Arif; Ren Liu; Xiuqing Li; Yue Zhou; Elizabeth Hyjek; Mark W. Lingen; Jeffery Mueller; Irving Waxman; Victoria M. Villaflor; Mark K. Ferguson; Everett E. Vokes; Parkash S. Gill; Ravi Salgia

Proceedings: AACR 103rd Annual Meeting 2012‐‐ Mar 31‐Apr 4, 2012; Chicago, IL Despite substantial improvements in screening, diagnosis, and treatment of esophageal cancer, the prognosis of this disease remains bleak. Survival at 5 years for all esophageal cancer patients taken together, with amenable treatments and with or without surgery ranges from 5-20% (Cancer Facts and Figures. American Cancer Society 2011). Although a multidisciplinary approach to include surgery, radiation, and chemotherapy, alone or in combination, attempts to improve the survival of this aggressive disease, these statistics underscore the continued need for attention to this disease and for identification of new targets in its treatment. We have been studying the role of receptor tyrosine kinases (RTKs) in cancer, particularly the EphB4 receptor, which has become increasingly associated with the pathobiology of adult cancers over the past several years. It has been shown to be aberrantly expressed and/or to play an oncogenic role in cancers of the breast, bladder, ovary, uterus, colon, head and neck, and prostate through its effects on cellular motility, growth, and migration, as well as on tumors’ ability to induce neoangiogenesis. To date, its role and potential as a target for therapy in different cancers have not been thoroughly investigated and remain poorly understood. Targeted inactivation of EphB4 and its ligand Ephrin-B2 have demonstrated that both are essential for angiogenic remodeling and embryonic survival (Adams and Klein 2000; Kim, Hu et al. 2008). In this report, we determine that EphB4 is overexpressed, has increased gene copy number and is involved in enhanced motility and migration in esophageal cancer. Archival patient samples consisting of 93 squamous cell carcinoma, 100 adenocarcinoma and 25 adjacent normal control samples as well as four adenocarcinoma and nine squamous cell carcinoma cell lines were used for the studies. Extensive mouse modeling of esophageal cancer was also used. We have reported that there is consistently higher expression of EphB4 in both squamous and adenocarcinoma compared to adjacent normal tissue with a statistically significant correlation between EphB4 expression and higher grades of squamous cell carcinoma. In a chemically induced esophageal squamous cell carcinoma model in mice, EphB4 was found overexpressed compared to normal controls. This study identifies EphB4 to be an important pathway in esophageal neoplastic lesions. It would now be useful to bring this to clinical fruition. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 1861. doi:1538-7445.AM2012-1861

Collaboration


Dive into the Xiuqing Li's collaboration.

Top Co-Authors

Avatar

Parkash S. Gill

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Ren Liu

University of Southern California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yue Zhou

University of Toyama

View shared research outputs
Top Co-Authors

Avatar

Anil Tulpule

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Dan Li

University of Southern California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge