Network


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

Hotspot


Dive into the research topics where Vivian Wai Yan Lui is active.

Publication


Featured researches published by Vivian Wai Yan Lui.


Cancer Discovery | 2013

Frequent mutation of the PI3K pathway in head and neck cancer defines predictive biomarkers

Vivian Wai Yan Lui; Matthew L. Hedberg; Hua Li; Bhavana S. Vangara; Kelsey P. Pendleton; Yan Zeng; Yiling Lu; Qiuhong Zhang; Yu Du; Breean R. Gilbert; Maria L. Freilino; Sam Sauerwein; Noah D. Peyser; Dong Xiao; Brenda Diergaarde; Lin Wang; Simion I. Chiosea; Raja R. Seethala; Jonas T. Johnson; Seungwon Kim; Umamaheswar Duvvuri; Robert L. Ferris; Marjorie Romkes; Tomoko Nukui; Patrick Kwok Shing Ng; Levi A. Garraway; Peter S. Hammerman; Gordon B. Mills; Jennifer R. Grandis

Genomic findings underscore the heterogeneity of head and neck squamous cell carcinoma (HNSCC). Identification of mutations that predict therapeutic response would be a major advance. We determined the mutationally altered, targetable mitogenic pathways in a large HNSCC cohort. Analysis of whole-exome sequencing data from 151 tumors revealed the phosphoinositide 3-kinase (PI3K) pathway to be the most frequently mutated oncogenic pathway (30.5%). PI3K pathway-mutated HNSCC tumors harbored a significantly higher rate of mutations in known cancer genes. In a subset of human papillomavirus-positive tumors, PIK3CA or PIK3R1 was the only mutated cancer gene. Strikingly, all tumors with concurrent mutation of multiple PI3K pathway genes were advanced (stage IV), implicating concerted PI3K pathway aberrations in HNSCC progression. Patient-derived tumorgrafts with canonical and noncanonical PIK3CA mutations were sensitive to an mTOR/PI3K inhibitor (BEZ-235), in contrast to PIK3CA-wild-type tumorgrafts. These results suggest that PI3K pathway mutations may serve as predictive biomarkers for treatment selection.


Expert Opinion on Biological Therapy | 2006

STAT3 as a therapeutic target in head and neck cancer

Rebecca Leeman; Vivian Wai Yan Lui; Jennifer R. Grandis

The signal transducer and activator of transcription (STAT) proteins relay signals from cytokine receptors and receptor tyrosine kinases on the cell surface to the nucleus, where they affect the transcription of genes involved in normal cell functions, including growth, apoptosis and differentiation. STAT3 has been found to be constitutively active in head and neck squamous cell carcinoma (HNSCC) as well as in other epithelial malignancies. In HNSCC, STAT3 alters the cell cycle, prevents apoptosis, and mediates the proliferation and survival of tumour cells. Several therapeutic approaches are being developed to target STAT3, including molecules that block either dimerisation or DNA binding by STAT3, strategies to decrease STAT3 expression and drugs that inhibit STAT3 function. Strategies that block STAT3 may prove efficacious for cancer treatment.


Cancer Research | 2012

TMEM16A, induces MAPK and contributes directly to tumorigenesis and cancer progression

Umamaheswar Duvvuri; Daniel J. Shiwarski; Dong Xiao; Carol A. Bertrand; Xin Huang; Robert S. Edinger; Jason R. Rock; Brian D. Harfe; Brian J. Henson; Karl Kunzelmann; Rainer Schreiber; Raja S. Seethala; Ann Marie Egloff; Xing Chen; Vivian Wai Yan Lui; Jennifer R. Grandis; Susanne M. Gollin

Frequent gene amplification of the receptor-activated calcium-dependent chloride channel TMEM16A (TAOS2 or ANO1) has been reported in several malignancies. However, its involvement in human tumorigenesis has not been previously studied. Here, we show a functional role for TMEM16A in tumor growth. We found TMEM16A overexpression in 80% of head and neck squamous cell carcinoma (SCCHN), which correlated with decreased overall survival in patients with SCCHN. TMEM16A overexpression significantly promoted anchorage-independent growth in vitro, and loss of TMEM16A resulted in inhibition of tumor growth both in vitro and in vivo. Mechanistically, TMEM16A-induced cancer cell proliferation and tumor growth were accompanied by an increase in extracellular signal-regulated kinase (ERK)1/2 activation and cyclin D1 induction. Pharmacologic inhibition of MEK/ERK and genetic inactivation of ERK1/2 (using siRNA and dominant-negative constructs) abrogated the growth effect of TMEM16A, indicating a role for mitogen-activated protein kinase (MAPK) activation in TMEM16A-mediated proliferation. In addition, a developmental small-molecule inhibitor of TMEM16A, T16A-inh01 (A01), abrogated tumor cell proliferation in vitro. Together, our findings provide a mechanistic analysis of the tumorigenic properties of TMEM16A, which represents a potentially novel therapeutic target. The development of small-molecule inhibitors against TMEM16A may be clinically relevant for treatment of human cancers, including SCCHN.


Future Oncology | 2011

Targeting the PI3K/Akt/mTOR pathway in hepatocellular carcinoma

Qian Zhou; Vivian Wai Yan Lui; Winnie Yeo

Despite recent advances in the understanding of the biologic basis of hepatocellular carcinoma (HCC) development, the clinical management of the disease remains a major challenge. Deregulation of the PI3K/Akt/mTOR pathway, which is a prototypic survival pathway, is increasingly implicated in HCC carcinogenesis. In this article, we detailed the role of this pathway in the pathogenesis of HCC and provide an update on the preclinical and clinical development of various agents targeting this key survival/proliferation pathway, which include various PI3K inhibitors, Akt inhibitors and mTOR inhibitors for HCC. In addition, we highlighted the therapeutic potential of combination strategy for mTOR inhibitors with conventional chemotherapy, in particular, antimicrotubule agents, other molecular targeting agents, as well as radiotherapy.


Molecular Cancer Therapeutics | 2007

Antitumor mechanisms of combined gastrin-releasing peptide receptor and epidermal growth factor receptor targeting in head and neck cancer

Qing Zhang; Neil E. Bhola; Vivian Wai Yan Lui; Doris R. Siwak; Sufi M. Thomas; Christopher T. Gubish; Jill M. Siegfried; Gordon B. Mills; Dong M. Shin; Jennifer R. Grandis

Head and neck squamous cell carcinoma (HNSCC) is characterized by epidermal growth factor receptor (EGFR) overexpression, where EGFR levels correlate with survival. To date, EGFR targeting has shown limited antitumor effects in head and neck cancer when administrated as monotherapy. We previously identified a gastrin-releasing peptide/gastrin-releasing peptide receptor (GRP/GRPR) aurocrine regulatory pathway in HNSCC, where GRP stimulates Src-dependent cleavage of EGFR proligands with subsequent EGFR phosphorylation and mitogen-activated protein kinase (MAPK) activation. To determine whether GRPR targeting can enhance the antitumor efficacy of EGFR inhibition, we investigated the effects of a GRPR antagonist (PD176252) in conjunction with an EGFR tyrosine kinase inhibitor (erlotinib). Combined blockade of GRPR and EGFR pathways significantly inhibited HNSCC, but not immortalized mucosal epithelial cell, proliferation, invasion, and colony formation. In addition, the percentage of apoptotic cells increased upon combined inhibition. The enhanced antitumor efficacy was accompanied by increased expression of cleaved poly(ADP-ribose) polymerase (PARP) and decreased phospho-EGFR, phospho-MAPK, and proliferating cell nuclear antigen (PCNA). Using reverse-phase protein microarray (RPPA), we further detected decreased expression of phospho–c-Jun, phospho-p70S6K, and phospho-p38 with combined targeting. Cumulatively, these results suggest that GRPR targeting can enhance the antitumor effects of EGFR inhibitors in head and neck cancer. [Mol Cancer Ther 2007;6(4):1414–24]


Oncogene | 2003

Mitogenic effects of gastrin-releasing peptide in head and neck squamous cancer cells are mediated by activation of the epidermal growth factor receptor

Vivian Wai Yan Lui; Sufi M. Thomas; Qing Zhang; Abbey L. Wentzel; Jill M. Siegfried; Joyce Yan Li; Jennifer R. Grandis

Head and neck squamous cell carcinomas (HNSCC) are characterized by upregulation of the epidermal growth factor receptor (EGFR), where EGFR serves as a potential therapeutic target. We previously reported that a gastrin-releasing peptide/gastrin-releasing peptide receptor (GRP/GRPR) autocrine growth pathway is activated early in HNSCC carcinogenesis. In the present study, we examined the mechanism of EGFR activation by GRP/GRPR in HNSCC proliferation. In HNSCC cells that express elevated levels of both GRPR and EGFR, we found that GRP induced rapid phosphorylation of EGFR as well as p44/42-MAPK activation. Using several EGFR-specific tyrosine kinase inhibitors and cells derived from EGFR knockout mice, we demonstrated that GRP-induced p44/42-MAPK activation was dependent upon EGFR activation. Further investigation demonstrated that cleavage of transforming growth factor-alpha (TGF-α) by matrix metalloproteinases mediated GRP-induced MAPK activation. In addition, HNSCC proliferation stimulated by GRP was eliminated upon specific inhibition of EGFR or MEK, and GRP failed to stimulate proliferation in EGFR-deficient cells. These results imply that the mitogenic effects of GRP in HNSCC are mediated by extracellular release of TGF-α and require the activation of an EGFR-dependent MEK/MAPK-dependent pathway.


Proceedings of the National Academy of Sciences of the United States of America | 2012

Cyclin D1 overexpression supports stable EBV infection in nasopharyngeal epithelial cells

Chi Man Tsang; Yim Ling Yip; Kwok Wai Lo; Wen Deng; Ka Fai To; Pok Man Hau; Victoria Ming Yi Lau; Kenzo Takada; Vivian Wai Yan Lui; Maria Li Lung; Honglin Chen; Mu Sheng Zeng; Jaap M. Middeldorp; Annie Lai Man Cheung; Sai Wah Tsao

Undifferentiated nasopharyngeal carcinomas (NPCs) are commonly present with latent EBV infection. However, events regulating EBV infection at early stages of the disease and the role of EBV in disease pathogenesis are largely undefined. Genetic alterations leading to activation of cyclin D1 signaling in premalignant nasopharyngeal epithelial (NPE) cells have been postulated to predispose cells to EBV infection. We previously reported that loss of p16, a negative regulator of cyclin D1 signaling, is a frequent feature of NPC tumors. Here, we report that early premalignant lesions of nasopharyngeal epithelium overexpress cyclin D1. Furthermore, overexpression of cyclin D1 is closely associated with EBV infection. Therefore we investigated the potential role of cyclin D1 overexpression in dysplastic NPE cells in vitro. In human telomerase reverse transcriptase-immortalized NPE cells, overexpression of cyclin D1 or a p16-resistant form of CDK4 (CDK4R24C) suppressed differentiation. This suppression may have implications for the close association of EBV infection with undifferentiated NPC. In these in vitro models, we found that cellular growth arrest and senescence occurred in EBV-infected cell populations immediately after infection. Nevertheless, overexpression of cyclin D1 or a p16-resistant form of CDK4 or knockdown of p16 in the human telomerase reverse transcriptase-immortalized NPE cell lines could counteract the EBV-induced growth arrest and senescence. We conclude that dysregulated expression of cyclin D1 in NPE cells may contribute to NPC pathogenesis by enabling persistent infection of EBV.


Clinical Cancer Research | 2013

c-Src Activation Mediates Erlotinib Resistance in Head and Neck Cancer by Stimulating c-Met

Laura P. Stabile; Guoqing He; Vivian Wai Yan Lui; Cassandra Henry; Christopher T. Gubish; Sonali Joyce; Kelly Quesnelle; Jill M. Siegfried; Jennifer R. Grandis

Purpose: Strategies to inhibit the EGF receptor (EGFR) using the tyrosine kinase inhibitor erlotinib have been associated with limited clinical efficacy in head and neck squamous cell carcinoma (HNSCC). Co-activation of alternative kinases may contribute to erlotinib resistance. Experimental Design: We generated HNSCC cells expressing dominant-active c-Src (DA-Src) to determine the contribution of c-Src activation to erlotinib response. Results: Expression of DA-Src conferred resistance to erlotinib in vitro and in vivo compared with vector-transfected control cells. Phospho-Met was strongly upregulated by DA-Src, and DA-Src cells did not produce hepatocyte growth factor (HGF). Knockdown of c-Met enhanced sensitivity to erlotinib in DA-Src cells in vitro, as did combining a c-Met or c-Src inhibitor with erlotinib. Inhibiting EGFR resulted in minimal reduction of phospho-Met in DA-Src cells, whereas complete phospho-Met inhibition was achieved by inhibiting c-Src. A c-Met inhibitor significantly sensitized DA-Src tumors to erlotinib in vivo, resulting in reduced Ki67 labeling and increased apoptosis. In parental cells, knockdown of endogenous c-Src enhanced sensitivity to erlotinib, whereas treatment with HGF to directly induce phospho-Met resulted in erlotinib resistance. The level of endogenous phospho-c-Src in HNSCC cell lines was also significantly correlated with erlotinib resistance. Conclusions: Ligand-independent activation of c-Met contributes specifically to erlotinib resistance, not cetuximab resistance, in HNSCC with activated c-Src, where c-Met activation is more dependent on c-Src than on EGFR, providing an alternate survival pathway. Addition of a c-Met or c-Src inhibitor to erlotinib may increase efficacy of EGFR inhibition in patients with activated c-Src. Clin Cancer Res; 19(2); 380–92. ©2012 AACR.


Molecular Pharmacology | 2007

Antiproliferative Mechanisms of a Transcription Factor Decoy Targeting Signal Transducer and Activator of Transcription (STAT) 3: The Role of STAT1

Vivian Wai Yan Lui; Amanda L. Boehm; Priya Koppikar; Rebecca Leeman; Daniel E. Johnson; Michelene Ogagan; Erin E. Childs; Maria L. Freilino; Jennifer R. Grandis

We previously developed a transcription factor decoy targeting signal transducer and activator of transcription 3 (STAT3) and reported antitumor activity in both in vitro and in vivo models of squamous cell carcinoma of the head and neck (SCCHN). Based on the known existence of STAT1-STAT3 heterodimers, the high sequence homology between STAT1 and STAT3, as well as expression of both STAT1 and STAT3 in SCCHN, we examined whether the STAT3 decoy interferes with STAT1 signaling. SCCHN cell lines with different STAT1 expression levels (but similar STAT3 levels) were used. Both cell lines were sensitive to the growth-inhibitory effects of the STAT3 decoy compared with a mutant control decoy. Intact STAT1 signaling was demonstrated by interferon-γ (IFN-γ)-mediated induction of STAT1 phosphorylation (Tyr701) and interferon-regulatory factor-1 (IRF-1) expression. Treatment with the STAT3 decoy (but not a mutant control decoy) resulted in inhibition of IRF-1 protein expression in both cell lines, indicating specific inhibition of STAT1 signaling by the STAT3 decoy. Because STAT1 is a potential tumor suppressor, we also investigated whether STAT1 signaling mitigated the therapeutic efficacy of the STAT3 decoy. In both PCI-15B and UM-22B cells, STAT1 siRNA treatment resulted in decreased STAT1 expression, without altering the antitumor activity of the STAT3 decoy. Likewise, the antitumor effects of the STAT3 decoy were not altered by STAT1 activation upon IFN-γ treatment. These results suggest that the therapeutic mechanisms of STAT3 blockade using a transcription factor decoy are independent of STAT1 activation.


Journal of Clinical Oncology | 2009

Intratumoral Epidermal Growth Factor Receptor Antisense DNA Therapy in Head and Neck Cancer: First Human Application and Potential Antitumor Mechanisms

Stephen Y. Lai; Priya Koppikar; Sufi M. Thomas; Erin E. Childs; Ann Marie Egloff; Raja R. Seethala; Barton F. Branstetter; William E. Gooding; Ashok Muthukrishnan; James M. Mountz; Vivian Wai Yan Lui; Dong M. Shin; Sanjiv S. Agarwala; Rita Johnson; Larry A. Couture; Eugene N. Myers; Jonas T. Johnson; Gordon B. Mills; Athanassios Argiris; Jennifer R. Grandis

PURPOSE Squamous cell carcinoma of the head and neck (SCCHN) is characterized by upregulation of the epidermal growth factor receptor (EGFR). We developed a novel strategy to target EGFR by using a therapeutic gene that consisted of an EGFR antisense (AS) gene sequence under U6 promoter control. A phase I clinical trial was conducted to evaluate the safety and biologic effects of EGFR AS. PATIENTS AND METHODS Patients with advanced SCCHN who were refractory to standard therapies and who had at least one assessable and accessible lesion were enrolled. The EGFR AS dose was escalated in successive cohorts (six dose levels; 60 to 1,920 microg/injection). Patients received four weekly intratumoral EGFR AS injections. Tumor biopsies were performed before and after completion of therapy. Treatment response was assessed by tumor volume measurements (positron emission tomography/computed tomography), and levels of target proteins were assessed by immunohistochemistry. RESULTS Seventeen assessable patients were treated. No grades 3 to 4 or dose-limiting toxicities were noted, and a maximum-tolerated dose was not reached. Five patients (29%) achieved a clinical response, which included two complete responses (CRs) and three partial responses (PRs); two additional patients had stable disease (SD) as the best response. Patients with disease control (CR + PR + SD) had tumors with higher EGFR and lower STAT3 expression at baseline compared with patients who had progressive disease (P = .0312 and P = .095, respectively). CONCLUSION Intratumoral EGFR AS was safe and resulted in antitumor activity in patients with advanced SCCHN. Baseline levels of high EGFR and low STAT3 may be associated with antitumor effects.

Collaboration


Dive into the Vivian Wai Yan Lui's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anthony T.C. Chan

The Chinese University of Hong Kong

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Margaret H.L. Ng

The Chinese University of Hong Kong

View shared research outputs
Top Co-Authors

Avatar

Brigette Ma

The Chinese University of Hong Kong

View shared research outputs
Top Co-Authors

Avatar

Edwin P. Hui

The Chinese University of Hong Kong

View shared research outputs
Top Co-Authors

Avatar

Gordon B. Mills

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Hua Li

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar

Bo Hong

The Chinese University of Hong Kong

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge