Network


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

Hotspot


Dive into the research topics where Tomoko Nukui is active.

Publication


Featured researches published by Tomoko Nukui.


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.


Pharmacogenetics | 2004

Maternal/newborn GSTT1 null genotype contributes to risk of preterm, low birthweight infants.

Tomoko Nukui; Richard D. Day; Cynthia S. Sims; Roberta B. Ness; Marjorie Romkes

OBJECTIVES Maternal cigarette smoke exposure during pregnancy has been identified as a risk factor for prematurity and low birthweight. However, little is known about genetic susceptibility and possible interactions with cigarette smoking which may increase risk of these events. METHODS Maternal peripheral and umbilical cord blood samples from 955 mother/newborn pairs were genotyped for a panel of phase I/II metabolic enzymes responsible for the metabolism of tobacco related mutagens and carcinogens in order to evaluate the association with premature birth. RESULTS As reported previously, maternal cigarette smoking during the last trimester was significantly associated with premature birth. In addition, maternal glutathione S-transferase T1 (GSTT1) null genotype also increased risk of premature birth. Risk was further elevated among subjects with the combination of maternal and newborn GSTT1 null genotype with or without maternal cigarette smoke. CONCLUSIONS These observations suggest that women and/or newborns with the GSTT1 null genotype who are exposed to cigarette smoke during pregnancy are at elevated risk for premature delivery. The ability to identify high-risk women by genotyping has potential for reducing the frequency of premature births, a major public health concern.


Journal of the National Cancer Institute | 2011

Genome-Wide Association Study of Survival in Non–Small Cell Lung Cancer Patients Receiving Platinum-Based Chemotherapy

Xifeng Wu; Yuanqing Ye; Rafael Rosell; Christopher I. Amos; David J. Stewart; Michelle A.T. Hildebrandt; Jack A. Roth; John D. Minna; Jian Gu; Jie Lin; Shama Buch; Tomoko Nukui; José Serrano; Miquel Taron; Adrian Cassidy; Charles Lu; Joe Y. Chang; Scott M. Lippman; Waun Ki Hong; Margaret R. Spitz; Marjorie Romkes; Ping Yang

BACKGROUND Interindividual variation in genetic background may influence the response to chemotherapy and overall survival for patients with advanced-stage non-small cell lung cancer (NSCLC). METHODS To identify genetic variants associated with poor overall survival in these patients, we conducted a genome-wide scan of 307,260 single-nucleotide polymorphisms (SNPs) in 327 advanced-stage NSCLC patients who received platinum-based chemotherapy with or without radiation at the University of Texas MD Anderson Cancer Center (the discovery population). A fast-track replication was performed for 315 patients from the Mayo Clinic followed by a second validation at the University of Pittsburgh in 420 patients enrolled in the Spanish Lung Cancer Group PLATAX clinical trial. A pooled analysis combining the Mayo Clinic and PLATAX populations or all three populations was also used to validate the results. We assessed the association of each SNP with overall survival by multivariable Cox proportional hazard regression analysis. All statistical tests were two-sided. RESULTS SNP rs1878022 in the chemokine-like receptor 1 (CMKLR1) was statistically significantly associated with poor overall survival in the MD Anderson discovery population (hazard ratio [HR] of death = 1.59, 95% confidence interval [CI] = 1.32 to 1.92, P = 1.42 × 10(-6)), in the PLATAX clinical trial (HR of death = 1.23, 95% CI = 1.00 to 1.51, P = .05), in the pooled Mayo Clinic and PLATAX validation (HR of death = 1.22, 95% CI = 1.06 to 1.40, P = .005), and in pooled analysis of all three populations (HR of death = 1.33, 95% CI = 1.19 to 1.48, P = 5.13 × 10(-7)). Carrying a variant genotype of rs10937823 was associated with decreased overall survival (HR of death = 1.82, 95% CI = 1.42 to 2.33, P = 1.73 × 10(-6)) in the pooled MD Anderson and Mayo Clinic populations but not in the PLATAX trial patient population (HR of death = 0.96, 95% CI = 0.69 to 1.35). CONCLUSION These results have the potential to contribute to the future development of personalized chemotherapy treatments for individual NSCLC patients.


Molecular Carcinogenesis | 2012

Genetic variability in DNA repair and cell cycle control pathway genes and risk of smoking-related lung cancer

Shama Buch; Brenda Diergaarde; Tomoko Nukui; Roger Day; Jill M. Siegfried; Marjorie Romkes; Joel L. Weissfeld

DNA repair and cell cycle control play an important role in the repair of DNA damage caused by cigarette smoking. Given this role, functionally relevant single nucleotide polymorphisms (SNPs) in genes in these pathways may well affect the risk of smoking‐related lung cancer. We examined the relationship between 240 SNPs in DNA repair and cell cycle control pathway genes and lung cancer risk in a case–control study of white current and ex‐cigarette smokers (722 cases and 929 controls). Additive, dominant, and recessive genetic models were evaluated for each SNP. A genetic risk summary score was also constructed. Odds ratios (OR) for lung cancer risk and 95% confidence intervals (95% CI) were estimated using logistic regression models. Thirty‐eight SNPs were associated with lung cancer risk in our study population at P < 0.05. The strongest associations were observed for rs2074508 in GTF2H4 (Padditive = 0.003), rs10500298 in LIG1 (Precessive = 2.7 × 10−4), rs747658 and rs3219073 in PARP1 (rs747658: Padditive = 5.8 × 10−5; rs3219073: Padditive = 4.6 × 10−5), and rs1799782 and rs3213255 in XRCC1 (rs1799782: Pdominant = 0.006; rs3213255: Precessive = 0.004). Compared to individuals with first quartile (lowest) risk summary scores, individuals with third and fourth quartile summary score results were at increased risk for lung cancer (OR: 2.21, 95% CI: 1.66–2.95 and OR: 3.44, 95% CI: 2.58–4.59, respectively; Ptrend < 0.0001). Our data suggests that variation in DNA repair and cell cycle control pathway genes is associated with smoking‐related lung cancer risk. Additionally, combining genotype information for SNPs in these pathways may assist in classifying current and ex‐cigarette smokers according to lung cancer risk.


Cancer | 2013

Pathologic and gene expression features of metastatic melanomas to the brain

Ronald L. Hamilton; Michal T. Krauze; Marjorie Romkes; Bernard Omolo; Panagiotis A. Konstantinopoulos; Todd A. Reinhart; Malgorzata Harasymczuk; Yangyang Wang; Yan Lin; Soldano Ferrone; Theresa L. Whiteside; Stephanie Bortoluzzi; Jonette Werley; Tomoko Nukui; Beth Fallert-Junecko; Douglas Kondziolka; Joseph G. Ibrahim; Dorothea Becker; John M. Kirkwood; Stergios J. Moschos

The prognosis of metastatic melanomas to the brain (MBM) is variable with prolonged survival in a subset. It is unclear whether MBM differ from extracranial metastases (EcM) and primary melanomas (PrM).


American Journal of Medical Genetics | 2009

The impact of genetic variation in DRD2 and SLC6A3 on smoking cessation in a cohort of participants 1 year after enrollment in a lung cancer screening study

Mindi A. Styn; Tomoko Nukui; Marjorie Romkes; Kenneth A. Perkins; Stephanie R. Land; Joel L. Weissfeld

Smoking cessation strategies continue to have disappointing results. By determining the interindividual genetic differences that influence smoking behaviors, we may be able to develop tailored strategies that increase the likelihood of successful cessation. This study attempts to determine genetic influences on the relationship between the dopamine pathway and smoking cessation by examining associations with a variable number tandem repeat variation in SLC6A3 and the DRD2 variants TaqIA (A2 vs. A1), TaqIB (B2 vs. B1), C957T (C vs. T), and −141C Ins/Del (C vs. Del). Baseline smokers in the Pittsburgh Lung Screening Study who provided information on smoking status 1 year later were evaluated. We frequency‐matched those who were not abstinent at 1 year to those who were abstinent at 1 year by gender, decade of age, and time of enrollment (3‐month intervals) in a 3:1 ratio (N = 881). Logistic regression was used to identify the effect of genotype on abstinence at 1 year. In a model containing the matching variables and other genotypes, DRD2 TaqIA was significantly associated with being abstinent at 1 year (P = 0.01). Compared to participants who were homozygous for the TaqIA major allele (A2A2), participants who carried at least one minor allele (A1) were less likely to quit (Odds Ratio: 0.47, 95% CI: 0.24–0.94). The other dopamine receptor genotypes and the SLC6A3 genotype were not associated with smoking status at 1 year. The association between DRD2 TaqIA and smoking cessation supports the hypothesis that genetic variation in the dopamine pathway influences smoking cessation.


Cancer Epidemiology, Biomarkers & Prevention | 2011

A sex-specific association between a 15q25 variant and upper aerodigestive tract cancers

Dan Chen; Thérèse Truong; Valerie Gaborieau; Graham Byrnes; Amelie Chabrier; Shu Chun Chuang; Andrew F. Olshan; Mark C. Weissler; Jingchun Luo; Marjorie Romkes; Shama Buch; Tomoko Nukui; Silvia Franceschi; Rolando Herrero; Renato Talamini; Karl T. Kelsey; Brock C. Christensen; Michael D. McClean; Martin Lacko; Johannes J. Manni; Wilbert H.M. Peters; Jan Lubinski; Joanna Trubicka; Marcin Lener; Joshua E. Muscat; Philip Lazarus; Qingyi Wei; Erich M. Sturgis; Zuo-Feng Zhang; Shen Chih Chang

Background: Sequence variants located at 15q25 have been associated with lung cancer and propensity to smoke. We recently reported an association between rs16969968 and risk of upper aerodigestive tract (UADT) cancers (oral cavity, oropharynx, hypopharynx, larynx, and esophagus) in women (OR = 1.24, P = 0.003) with little effect in men (OR = 1.04, P = 0.35). Methods: In a coordinated genotyping study within the International Head and Neck Cancer Epidemiology (INHANCE) consortium, we have sought to replicate these findings in an additional 4,604 cases and 6,239 controls from 10 independent UADT cancer case–control studies. Results: rs16969968 was again associated with UADT cancers in women (OR = 1.21, 95% CI = 1.08–1.36, P = 0.001) and a similar lack of observed effect in men [OR = 1.02, 95% CI = 0.95–1.09, P = 0.66; P-heterogeneity (Phet) = 0.01]. In a pooled analysis of the original and current studies, totaling 8,572 UADT cancer cases and 11,558 controls, the association was observed among females (OR = 1.22, 95% CI = 1.12–1.34, P = 7 × 10−6) but not males (OR = 1.02, 95% CI = 0.97–1.08, P = 0.35; Phet = 6 × 10−4). There was little evidence for a sex difference in the association between this variant and cigarettes smoked per day, with male and female rs16969968 variant carriers smoking approximately the same amount more in the 11,991 ever smokers in the pooled analysis of the 14 studies (Phet = 0.86). Conclusions: This study has confirmed a sex difference in the association between the 15q25 variant rs16969968 and UADT cancers. Impact: Further research is warranted to elucidate the mechanisms underlying these observations. Cancer Epidemiol Biomarkers Prev; 20(4); 658–64. ©2011 AACR.


American Journal of Respiratory and Critical Care Medicine | 2014

Matrix Metalloproteinase-19 Promotes Metastatic Behavior In Vitro and Is Associated with Increased Mortality in Non–Small Cell Lung Cancer

Guoying Yu; Jose D. Herazo-Maya; Tomoko Nukui; Marjorie Romkes; Anil V. Parwani; Brenda Juan-Guardela; Jennifer Robertson; Jack Gauldie; Jill M. Siegfried; Naftali Kaminski; Daniel J. Kass

RATIONALE Lung cancer is the leading cause of cancer death in both men and women in the United States and worldwide. Matrix metalloproteinases (MMPs) have been implicated in the development and progression of lung cancer, but their role in the molecular pathogenesis of lung cancer remains unclear. We have found that MMP19, a relatively novel member of the MMP family, is overexpressed in lung tumors when compared with control subjects. OBJECTIVES To test the hypothesis that MMP19 plays a significant role in the development and progression of non-small cell lung cancer (NSCLC). METHODS We have analyzed lung cancer gene expression data, immunostained lung tumors for MMP19, and performed in vitro assays to test the effects of MMP19 in NSCLC cells. MEASUREMENTS AND MAIN RESULTS We found that MMP19 gene and protein expression is increased in lung cancer tumors compared with adjacent and histologically normal lung tissues. In three independent datasets, increased MMP19 gene expression conferred a poorer prognosis in NSCLC. In vitro, we found that overexpression of MMP19 promotes epithelial-mesenchymal transition, migration, and invasiveness in multiple NSCLC cell lines. Overexpression of MMP19 with a mutation at the catalytic site did not impair epithelial-mesenchymal transition or expression of prometastasis genes. We also found that miR-30 isoforms, a microRNA family predicted to target MMP19, is markedly down-regulated in human lung cancer and regulates MMP19 expression. CONCLUSIONS Taken together, these findings suggest that MMP19 is associated with the development and progression of NSCLC and may be a potential biomarker of disease severity and outcome.


Journal of the National Cancer Institute | 2015

15q12 Variants, Sputum Gene Promoter Hypermethylation, and Lung Cancer Risk: A GWAS in Smokers

Shuguang Leng; Yushi Liu; Joel L. Weissfeld; Cynthia L. Thomas; Younghun Han; Maria A. Picchi; Christopher K. Edlund; Randall P. Willink; Autumn Gaither Davis; Kieu Do; Tomoko Nukui; Xiequn Zhang; Elizabeth A. Burki; David Van Den Berg; Marjorie Romkes; W. James Gauderman; Richard E. Crowell; Yohannes Tesfaigzi; Christine A. Stidley; Christopher I. Amos; Jill M. Siegfried; Frank D. Gilliland; Steven A. Belinsky

BACKGROUND Lung cancer is the leading cause of cancer-related mortality worldwide. Detection of promoter hypermethylation of tumor suppressor genes in exfoliated cells from the lung provides an assessment of field cancerization that in turn predicts lung cancer. The identification of genetic determinants for this validated cancer biomarker should provide novel insights into mechanisms underlying epigenetic reprogramming during lung carcinogenesis. METHODS A genome-wide association study using generalized estimating equations and logistic regression models was conducted in two geographically independent smoker cohorts to identify loci affecting the propensity for cancer-related gene methylation that was assessed by a 12-gene panel interrogated in sputum. All statistical tests were two-sided. RESULTS Two single nucleotide polymorphisms (SNPs) at 15q12 (rs73371737 and rs7179575) that drove gene methylation were discovered and replicated with rs73371737 reaching genome-wide significance (P = 3.3×10(-8)). A haplotype carrying risk alleles from the two 15q12 SNPs conferred 57% increased risk for gene methylation (P = 2.5×10(-9)). Rs73371737 reduced GABRB3 expression in lung cells and increased risk for smoking-induced chronic mucous hypersecretion. Furthermore, subjects with variant homozygote of rs73371737 had a two-fold increase in risk for lung cancer (P = .0043). Pathway analysis identified DNA double-strand break repair by homologous recombination (DSBR-HR) as a major pathway affecting susceptibility for gene methylation that was validated by measuring chromatid breaks in lymphocytes challenged by bleomycin. CONCLUSIONS A functional 15q12 variant was identified as a risk factor for gene methylation and lung cancer. The associations could be mediated by GABAergic signaling that drives the smoking-induced mucous cell metaplasia. Our findings also substantiate DSBR-HR as a critical pathway driving epigenetic gene silencing.


Journal of Thoracic Oncology | 2015

Lung Cancer Risk Prediction Using Common SNPs Located in GWAS-Identified Susceptibility Regions.

Joel L. Weissfeld; Yan Lin; Hui Min Lin; Brenda F. Kurland; David O. Wilson; Carl R. Fuhrman; Arjun Pennathur; Marjorie Romkes; Tomoko Nukui; Jian-Min Yuan; Jill M. Siegfried; Brenda Diergaarde

Introduction: Genome-wide association studies (GWAS) have consistently identified specific lung cancer susceptibility regions. We evaluated the lung cancer–predictive performance of single-nucleotide polymorphisms (SNPs) in these regions. Methods: Lung cancer cases (N = 778) and controls (N = 1166) were genotyped for 77 SNPs located in GWAS-identified lung cancer susceptibility regions. Variable selection and model development used stepwise logistic regression and decision-tree analyses. In a subset nested in the Pittsburgh Lung Screening Study, change in area under the receiver operator characteristic curve and net reclassification improvement were used to compare predictions made by risk factor models with and without genetic variables. Results: Variable selection and model development kept two SNPs in each of three GWAS regions, rs2736100 and rs7727912 in 5p15.33, rs805297 and rs1802127 in 6p21.33, and rs8034191 and rs12440014 in 15q25.1. The ratio of cases to controls was three times higher among subjects with a high-risk genotype in every one as opposed to none of the three GWAS regions (odds ratio, 3.14; 95% confidence interval, 2.02–4.88; adjusted for sex, age, and pack-years). Adding a three-level classified count of GWAS regions with high-risk genotypes to an age and smoking risk factor-only model improved lung cancer prediction by a small amount: area under the receiver operator characteristic curve, 0.725 versus 0.717 (p = 0.056); overall net reclassification improvement was 0.052 across low-, intermediate-, and high- 6-year lung cancer risk categories (<3.0%, 3.0%–4.9%, ≥5.0%). Conclusion: Specifying genotypes for SNPs in three GWAS-identified susceptibility regions improved lung cancer prediction, but probably by an extent too small to affect disease control practice.

Collaboration


Dive into the Tomoko Nukui's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Shama Buch

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sanja Dacic

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar

Petr Pancoska

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yan Lin

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge