Network


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

Hotspot


Dive into the research topics where Kenneth W. Yip is active.

Publication


Featured researches published by Kenneth W. Yip.


Clinical Cancer Research | 2006

Prognostic significance of the epstein-barr virus, p53, Bcl-2, and survivin in nasopharyngeal cancer

Kenneth W. Yip; Wei Shi; Melania Pintilie; Joseph D. Martin; Joseph D. Mocanu; Derek Wong; Christine MacMillan; Pat Gullane; Brian O'Sullivan; Carlo Bastianutto; Fei-Fei Liu

Purpose: Nasopharyngeal cancer (NPC) is a malignant epithelial carcinoma which is intimately associated with EBV. The latent presence of EBV affects the function of p53, Bcl-2, and survivin. We thus investigated the relationship between EBV status, p53, Bcl-2, and survivin in biopsy specimens from patients with primary NPC. Experimental Design: Archival formalin-fixed, paraffin-embedded NPC biopsies were evaluated in 80 patients treated with curative radiation from a single institution. The presence of EBV was determined using EBER in situ hybridization, whereas p53, Bcl-2, and survivin were assessed using immunohistochemistry. Results: The majority of NPC specimens in this patient cohort were EBER-positive (64 of 78, or 82%), which in turn, was significantly associated with ethnicity (P = 0.0007), and WHO subtype 2A/2B (P = 0.04). EBER-positive tumors were also associated with p53 (P = 0.002), Bcl-2 (P = 0.04), and nuclear survivin (P = 0.03) expression. Patients with EBER-positive NPC fared better, with a 10-year overall survival of 68% versus 48% for EBER-negative patients (P = 0.03). For nuclear survivin, patients with either low or high nuclear survivin fared worse than patients with intermediate survivin expression (P = 0.05), suggesting that there is an optimal proportion of survivin-expressing cells for best function and clinical outcome. Conclusions: With an extended median follow-up time of 11.4 years, EBV status remains a strong predictor for overall survival in NPC. EBV-positive NPC has strong molecular associations with p53, Bcl-2, and survivin expression. Furthermore, we provide clinical data revealing the potentially dual nature of survivin in predicting clinical outcome.


Clinical Cancer Research | 2005

Combination Bcl-2 Antisense and Radiation Therapy for Nasopharyngeal Cancer

Kenneth W. Yip; Joseph D. Mocanu; P.Y. Billie Au; Gillian T. Sleep; Dolly P. Huang; Pierre Busson; Wen-Chen Yeh; Ralph W. Gilbert; Brian O'Sullivan; Patrick J. Gullane; Carlo Bastianutto; Fei-Fei Liu

Purpose: A wide variety of tumors depend on the dysregulation of Bcl-2 family proteins for survival. The resulting apoptotic block can often provide a mechanism for resistance to anticancer treatments, such as chemotherapy and radiation. This current study evaluates the efficacy of combining systemically delivered Bcl-2 phosphorothioate antisense (Bcl-2 ASO) and radiation for nasopharyngeal cancer therapy. Results: Antisense uptake was unaffected by 0, 3, or 6 Gy radiation. Radiation decreased the fraction of viable C666-1 cells to 60%, with a further decrease to 40% in combination with Bcl-2 ASO. Despite a modest in vitro effect, Bcl-2 ASO alone caused the regression of established xenograft tumors in mice, extending survival by 15 days in a C666-1 and by 6 days in a C15 model. The survival times for mice treated with both Bcl-2 ASO and radiation increased by 52 days in C666-1 and by 20 days in C15 tumors. This combination resulted in a more-than-additive effect in C666-1 tumors. Less impressive gains observed in C15 tumors might be attributable to higher expression of antiapoptotic Bcl-2 family proteins and limited drug distribution in the tumor. Retreatment of C666-1 tumors with the Bcl-2 ASO-radiation combination, however, was effective, resulting in mice surviving for >80 days relative to untreated controls. Conclusions: Our results show that the Bcl-2 ASO and radiation combination is a highly potent therapy for nasopharyngeal cancer. Further examination of combination therapy with radiation and other Bcl-2 family–targeted anticancer agents in both preclinical and clinical settings is definitely warranted.


Molecular Pharmacology | 2009

Potential Use of Cetrimonium Bromide as an Apoptosis-Promoting Anticancer Agent for Head and Neck Cancer

Emma Ito; Kenneth W. Yip; David Katz; Sonali B. Fonseca; David W. Hedley; Sue Chow; G. Wei Xu; Tabitha E. Wood; Carlo Bastianutto; Aaron D. Schimmer; Shana O. Kelley; Fei-Fei Liu

A potential therapeutic agent for human head and neck cancer (HNC), cetrimonium bromide (CTAB), was identified through a cell-based phenotype-driven high-throughput screen (HTS) of 2000 biologically active or clinically used compounds, followed by in vitro and in vivo characterization of its antitumor efficacy. The preliminary and secondary screens were performed on FaDu (hypopharyngeal squamous cancer) and GM05757 (primary normal fibroblasts), respectively. Potential hit compounds were further evaluated for their anticancer specificity and efficacy in combination with standard therapeutics on a panel of normal and cancer cell lines. Mechanism of action, in vivo antitumor efficacy, and potential lead compound optimizations were also investigated. In vitro, CTAB interacted additively with γ radiation and cisplatin, two standard HNC therapeutic agents. CTAB exhibited anticancer cytotoxicity against several HNC cell lines, with minimal effects on normal fibroblasts; a selectivity that exploits cancer-specific metabolic aberrations. The central mode of cytotoxicity was mitochondria-mediated apoptosis via inhibition of H+-ATP synthase activity and mitochondrial membrane potential depolarization, which in turn was associated with reduced intracellular ATP levels, caspase activation, elevated sub-G1 cell population, and chromatin condensation. In vivo, CTAB ablated tumor-forming capacity of FaDu cells and delayed growth of established tumors. Thus, using an HTS approach, CTAB was identified as a potential apoptogenic quaternary ammonium compound possessing in vitro and in vivo efficacy against HNC models.


Clinical Cancer Research | 2006

Benzethonium Chloride: A Novel Anticancer Agent Identified by Using a Cell-Based Small-Molecule Screen

Kenneth W. Yip; Xinliang Mao; P.Y. Billie Au; David W. Hedley; Sue Chow; Shadi Dalili; Joseph D. Mocanu; Carlo Bastianutto; Aaron D. Schimmer; Fei-Fei Liu

Purpose: This study aims to identify a novel therapeutic agent for head and neck cancer and to evaluate its antitumor efficacy. Experimental Design: A cell-based and phenotype-driven high-throughput screening of ∼2,400 biologically active or clinically used compounds was done using a tetrazolium-based assay on FaDu (hypopharyngeal squamous cancer) and NIH 3T3 (untransformed mouse embryonic fibroblast) cells, with secondary screening done on C666-1 (nasopharyngeal cancer) and GM05757 (primary normal human fibroblast) lines. The “hit” compound was assayed for efficacy in combination with standard therapeutics on a panel of human cancer cell lines. Furthermore, its mode of action (using transmission electron microscopy and flow cytometry) and its in vivo efficacy (using xenograft models) were evaluated. Results: Benzethonium chloride was identified as a novel cancer-specific compound. For benzethonium (48-hour incubation), the dose required to reduce cell viability by 50% was 3.8 μmol/L in FaDu, 42.2 μmol/L in NIH 3T3, 5.3 μmol/L in C666-1, and 17.0 μmol/L in GM05757. In vitro, this compound did not interfere with the effects of cisplatin, 5-fluorouracil, or γ-irradiation. Benzethonium chloride induced apoptosis and activated caspases after 12 hours. Loss of mitochondrial membrane potential (ΔΨM) preceded cytosolic Ca2+ increase and cell death. In vivo, benzethonium chloride ablated the tumor-forming ability of FaDu cells, delayed the growth of xenograft tumors, and combined additively with local tumor radiation therapy. Evaluation of benzethonium chloride on the National Cancer Institute/NIH Developmental Therapeutics Program 60 human cancer cell lines revealed broad-range antitumor activity. Conclusions: This high-throughput screening identified a novel antimicrobial compound with significant broad-spectrum anticancer activity.


Molecular Cancer Therapeutics | 2006

Potential use of alexidine dihydrochloride as an apoptosis-promoting anticancer agent

Kenneth W. Yip; Emma Ito; Xinliang Mao; P.Y. Billie Au; David W. Hedley; Joseph D. Mocanu; Carlo Bastianutto; Aaron D. Schimmer; Fei-Fei Liu

Despite advances in surgery, radiation, and chemotherapy, novel therapeutics are needed for head and neck cancer treatment. The objective of this current study was to evaluate alexidine dihydrochloride as a novel compound lead for head and neck cancers. Using a tetrazolium-based assay, the dose required to reduce cell viability by 50% (ED50) was found to be ∼1.8 μmol/L in FaDu (human hypopharyngeal squamous cancer) and ∼2.6 μmol/L in C666-1 (human undifferentiated nasopharyngeal cancer) cells. In contrast, the ED50 values were much higher in untransformed cells, specifically at ∼8.8 μmol/L in GM05757 (primary normal human fibroblast), ∼8.9 μmol/L in HNEpC (primary normal human nasal epithelial), and ∼19.6 μmol/L in NIH/3T3 (mouse embryonic fibroblast) cells. Alexidine dihydrochloride did not interfere with the activities of cisplatin, 5-fluorouracil, or radiation, and interacted in a less-than-additive manner. DNA content analyses and Hoechst 33342 staining revealed that this compound induced apoptosis. Alexidine dihydrochloride–induced mitochondrial damage was visualized using transmission electron microscopy. Mitochondrial membrane potential (ΔΨM) depolarization was detectable after only 3 hours of treatment, and was followed by cytosolic Ca2+ increase along with loss of membrane integrity/cell death. Caspase-2 and caspase-9 activities were detectable at 12 hours, caspase-8 at 24 hours, and caspase-3 at 48 hours. FaDu cell clonogenic survival was reduced to <5% with 1 μmol/L alexidine dihydrochloride, and, correspondingly, this compound decreased the in vivo tumor-forming potential of FaDu cells. Thus, we have identified alexidine dihydrochloride as the first bisbiguanide compound with anticancer specificity. [Mol Cancer Ther 2006;5(9):2234–40]


Journal of Clinical Oncology | 2015

Nasopharyngeal Cancer: Molecular Landscape

Jeff Bruce; Kenneth W. Yip; Scott V. Bratman; Emma Ito; Fei-Fei Liu

Nasopharyngeal carcinoma (NPC) is a unique epithelial malignancy arising from the superior aspect of the pharyngeal mucosal space, associated with latent Epstein-Barr virus infection in most cases. The capacity to characterize cancer genomes in unprecedented detail is now providing insights into the genesis and molecular underpinnings of this disease. Herein, we provide an overview of the molecular aberrations that likely drive nasopharyngeal tumor development and progression. The contributions of major Epstein-Barr virus-encoded factors, including proteins, small RNAs, and microRNAs, along with their interactions with pathways regulating cell proliferation and survival are highlighted. We review recent analyses that clearly define the role of genetic and epigenetic variations affecting the human genome in NPC. These findings point to the impact of DNA methylation and histone modifications on gene expression programs that promote this malignancy. The molecular interactions that allow NPC cells to evade immune recognition and elimination, which is crucial for the survival of cells expressing potentially immunogenic viral proteins, are also described. Finally, the potential utility of detecting host and viral factors for the diagnosis and prognosis of NPC is discussed. Altogether, the studies summarized herein have greatly expanded our knowledge of the molecular biology of NPC, yet much remains to be uncovered. Emerging techniques for using and analyzing well-annotated biospecimens from patients with NPC will ultimately lead to a greater level of understanding, and enable improvements in precision therapies and clinical outcomes.


Journal of Human Genetics | 2017

MicroRNAs in extracellular vesicles: potential cancer biomarkers

Takashi Kinoshita; Kenneth W. Yip; Tara Spence; Fei-Fei Liu

Extracellular vesicles (EV) are small membrane-bound structures that are secreted by various cell types, including tumor cells. Recent studies have shown that EVs are important for cell-to-cell communication, locally and distantly; horizontally transferring DNA, mRNA, microRNA (miRNA), proteins and lipids. In the context of cancer biology, tumor-derived EVs are capable of modifying the microenvironment, promoting tumor progression, immune evasion, angiogenesis and metastasis. miRNAs contained within EVs are functionally associated with cancer progression, metastasis and aggressive tumor phenotypes. These factors, along with their stability in bodily fluids, have led to extensive investigations on the potential role of circulating EV-derived miRNAs as tumor biomarkers. In this review, we summarize the current understanding of circulating EV miRNAs in human cancer, and discuss their clinical utility and challenges in functioning as biomarkers.


Oncotarget | 2016

MiR-449a promotes breast cancer progression by targeting CRIP2

Wei Shi; Jeff Bruce; Matthew Lee; Shijun Yue; Matthew Rowe; Melania Pintilie; Ryunosuke Kogo; Pierre Antoine Bissey; Anthony Fyles; Kenneth W. Yip; Fei-Fei Liu

The identification of prognostic biomarkers and their underlying mechanisms of action remain of great interest in breast cancer biology. Using global miRNA profiling of 71 lymph node-negative invasive ductal breast cancers and 5 normal mammary epithelial tissues, we identified miR-449a to be highly overexpressed in the malignant breast tissue. Its expression was significantly associated with increased incidence of patient relapse, decreased overall survival, and decreased disease-free survival. In vitro, miR-449a promoted breast cancer cell proliferation, clonogenic survival, migration, and invasion. By utilizing a tri-modal in silico approach for target identification, Cysteine-Rich Protein 2 (CRIP2; a transcription factor) was identified as a direct target of miR-449a, corroborated using qRT-PCR, Western blot, and luciferase reporter assays. MDA-MB-231 cells stably transfected with CRIP2 demonstrated a significant reduction in cell viability, migration, and invasion, as well as decreased tumor growth and angiogenesis in mouse xenograft models. Our data revealed that overexpression of miR-449a suppresses CRIP2, which then affects the tumor vasculature, likely via NF-κB/p65 complex-mediated transcription of VEGF. These finding define an oncogenic function of miR-449a in human breast cancer, and highlight the importance of this pathway in driving aggressive behaviour.


Molecular Therapy | 2007

Imaging the Modulation of Adenoviral Kinetics and Biodistribution for Cancer Gene Therapy

Joseph D. Mocanu; Kenneth W. Yip; Nehad M. Alajez; Wei Shi; Jian-Hua Li; Sarah Jane Lunt; Eduardo H. Moriyama; Brian C. Wilson; Michael Milosevic; Kwok Wai Lo; Nico van Rooijen; Pierre Busson; Carlo Bastianutto; Fei-Fei Liu

See page 841 To explore systemic utilization of Epstein-Barr virus (EBV)-specific transcriptionally targeted adenoviruses, three vectors were constructed to examine kinetics, specificity, and biodistribution: adv.oriP.luc, expressing luciferase under EBV-specific control; adv.SV40luc, expressing luciferase constitutively; and adv.oriP.E1A.oriP.luc, a conditionally replicating adenovirus, expressing both luciferase and E1A. Bioluminescence imaging (BLI) was conducted on tumor-bearing severe combined immunodeficient (SCID) mice (C666-1, EBV-positive human nasopharyngeal cancer) treated intravenously (i.v.) with 3 × 108 infectious units (ifu) of the adenoviral vectors. At 72 hours, adv.oriPluc demonstrated an 8.4-fold higher tumor signal than adv.SV40luc; adv.oriP.E1A.oriP.luc was 26.7-fold higher; however, a significant liver signal was also observed, necessitating further action to improve biodistribution. Several compounds were examined to this end, including norepinephrine, serotonin, clodronate liposomes, and STI571, to determine whether any of these measures could improve adenoviral biodistribution. Each of these interventions was assessed using BLI in mice i.v. injected with adv.oriP.luc. STI571 achieved the highest increase in tumor-to-liver ratio (TLR; 6.6-fold), which was associated with a 59% reduction in tumor interstitial fluid pressure (IFP) along with a decrease in platelet-derived growth factor-β receptor (PDGFβR) activation. This study reports the favorable modulation by STI571 of the biodistribution of adenoviral vectors, providing a potential approach to improving therapeutic outcome.


JAMA Oncology | 2016

Human Papillomavirus Genotype Association With Survival in Head and Neck Squamous Cell Carcinoma

Scott V. Bratman; Jeffrey P. Bruce; Brian O'Sullivan; Trevor J. Pugh; Wei Xu; Kenneth W. Yip; Fei-Fei Liu

Human Papillomavirus Genotype Association With Survival in Head and Neck Squamous Cell Carcinoma The presence of human papillomavirus (HPV) has recently been recognized as a strong positive prognostic factor for head and neck squamous cell carcinoma (HNSC) and is used to select patients for risk-adapted treatment regimens. A positive immunohistochemical (IHC) finding for p16 is used as a surrogate for HPV based on the high concordance between these 2 biomarkers1,2; however, p16 IHC analysis cannot distinguish between HPV-16 (the most common HPV genotype found in HNSC) and other HPV genotypes. We investigated the prognostic consequences of distinct HPV genotypes within HNSC. Methods | Patient data available through The Cancer Genome Atlas (TCGA)3 was collected after obtaining informed consent in accordance with institutional review board–approved studies at each participating TCGA partner institution. We downloaded RNA-seq bam files from the University of California, Santa Cruz, Cancer Genomics Hub. VirusFinder (version 2.0)4 was used to identify 179 distinct HPV genotypes. Viral gene expression was normalized to total HPV-mapped reads and gene size (reads per kilobase of transcript per million [RPKM]). Clinical data were downloaded from Broad Institute’s Firehose database (http://gdac.broadinstitute.org; std _data run 2015_06_01). The Kaplan-Meier method was used to estimate survival; survival between cohorts was compared using the log-rank test. Both χ2 and t tests were used to compare clinical and

Collaboration


Dive into the Kenneth W. Yip's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jeff Bruce

Princess Margaret Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Scott V. Bratman

Princess Margaret Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Wei Shi

University Health Network

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Brian O'Sullivan

Princess Margaret Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Emma Ito

University Health Network

View shared research outputs
Top Co-Authors

Avatar

John Waldron

Princess Margaret Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Aaron D. Schimmer

Princess Margaret Cancer Centre

View shared research outputs
Researchain Logo
Decentralizing Knowledge