Network


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

Hotspot


Dive into the research topics where Ching Lau is active.

Publication


Featured researches published by Ching Lau.


Nature Genetics | 2009

Genome-wide association study identifies five susceptibility loci for glioma.

Sanjay Shete; Fay J. Hosking; Lindsay B. Robertson; Sara E. Dobbins; Marc Sanson; Beatrice Malmer; Matthias Simon; Yannick Marie; Blandine Boisselier; Jean Yves Delattre; Khê Hoang-Xuan; Soufiane El Hallani; Ahmed Idbaih; Diana Zelenika; Ulrika Andersson; Roger Henriksson; A. Tommy Bergenheim; Maria Feychting; Stefan Lönn; Anders Ahlbom; Johannes Schramm; Michael Linnebank; Kari Hemminki; Rajiv Kumar; Sarah J. Hepworth; Amy Price; Georgina Armstrong; Yanhong Liu; Xiangjun Gu; Robert Yu

To identify risk variants for glioma, we conducted a meta-analysis of two genome-wide association studies by genotyping 550K tagging SNPs in a total of 1,878 cases and 3,670 controls, with validation in three additional independent series totaling 2,545 cases and 2,953 controls. We identified five risk loci for glioma at 5p15.33 (rs2736100, TERT; P = 1.50 × 10−17), 8q24.21 (rs4295627, CCDC26; P = 2.34 × 10−18), 9p21.3 (rs4977756, CDKN2A-CDKN2B; P = 7.24 × 10−15), 20q13.33 (rs6010620, RTEL1; P = 2.52 × 10−12) and 11q23.3 (rs498872, PHLDB1; P = 1.07 × 10−8). These data show that common low-penetrance susceptibility alleles contribute to the risk of developing glioma and provide insight into disease causation of this primary brain tumor.


Lancet Oncology | 2006

Risk-adapted craniospinal radiotherapy followed by high-dose chemotherapy and stem-cell rescue in children with newly diagnosed medulloblastoma (St Jude Medulloblastoma-96): long-term results from a prospective, multicentre trial

Amar Gajjar; Murali Chintagumpala; David M. Ashley; Stewart J. Kellie; Larry E. Kun; Thomas E. Merchant; Shaio Woo; Greg Wheeler; Valerie Ahern; Matthew J. Krasin; Maryam Fouladi; Alberto Broniscer; Robert A. Krance; Gregory A. Hale; Clinton F. Stewart; Robert C. Dauser; Robert A. Sanford; Christine E. Fuller; Ching Lau; James M. Boyett; Dana Wallace; Richard J. Gilbertson

BACKGROUND Current treatment for medulloblastoma, which includes postoperative radiotherapy and 1 year of chemotherapy, does not cure many children with high-risk disease. We aimed to investigate the effectiveness of risk-adapted radiotherapy followed by a shortened period of dose-intense chemotherapy in children with medulloblastoma. METHODS After resection, patients were classified as having average-risk medulloblastoma (< or = 1.5 cm2 residual tumour and no metastatic disease) or high-risk medulloblastoma (> 1.5 cm2 residual disease or metastatic disease localised to neuraxis) medulloblastoma. All patients received risk-adapted craniospinal radiotherapy (23.4 Gy for average-risk disease and 36.0-39.6 Gy for high-risk disease) followed by four cycles of cyclophosphamide-based, dose-intensive chemotherapy. Patients were assessed regularly for disease status and treatment side-effects. The primary endpoint was 5-year event-free survival; we also measured overall survival. This study is registered with ClinicalTrials.gov, number NCT00003211. FINDINGS Of 134 children with medulloblastoma who underwent treatment (86 average-risk, 48 high-risk), 119 (89%) completed the planned protocol. No treatment-related deaths occurred. 5-year overall survival was 85% (95% CI 75-94) in patients in the average-risk group and 70% (54-84) in those in the high-risk group (p=0.04); 5-year event-free survival was 83% (73-93) and 70% (55-85), respectively (p=0.046). For the 116 patients whose histology was reviewed centrally, histological subtype correlated with 5-year event-free survival (p=0.04): 84% (74-95) for classic histology, 77% (49-100) for desmoplastic tumours, and 57% (33-80) for large-cell anaplastic tumours. INTERPRETATION Risk-adapted radiotherapy followed by a shortened schedule of dose-intensive chemotherapy can be used to improve the outcome of patients with high-risk medulloblastoma.


Journal of Clinical Oncology | 2004

Clinical, Histopathologic, and Molecular Markers of Prognosis: Toward a New Disease Risk Stratification System for Medulloblastoma

Amar Gajjar; Roberto Hernan; Mehmet Kocak; Christine Fuller; Youngsoo Lee; Peter J. McKinnon; Dana Wallace; Ching Lau; Murali Chintagumpala; David M. Ashley; Stewart J. Kellie; L. E. Kun; Richard J. Gilbertson

PURPOSE To assess the feasibility of performing central molecular analyses of fresh medulloblastomas obtained from multiple institutions and using these data to identify prognostic markers for contemporaneously treated patients. MATERIALS AND METHODS Ninety-seven samples of medulloblastoma were collected. Tumor content in samples was judged by frozen section review. Tumor ERBB2 protein and MYCC, MYCN, and TRKC mRNA levels were measured blind to clinical details using Western blotting and real-time polymerase chain reaction, respectively. Histopathologic and clinical review of each case was also performed. All data were subjected to independent statistical analysis. RESULTS Sample acquisition and analysis times ranged from 3 to 6 days. Eighty-six samples contained sufficient tumor for analysis, including 38 classic, 30 nodular desmoplastic, and 18 large-cell anaplastic (LCA) medulloblastomas. Protein and mRNA were extracted from 81 and 49 tumors, respectively. ERBB2 was detected in 40% (n=32 of 81) of tumors, most frequently in LCA disease (P=.005), and was independently associated with a poor prognosis (P=.031). A combination of clinical characteristics and ERBB2 expression provided a highly accurate means of discriminating disease risk. One hundred percent (n=26) of children with clinical average-risk, ERBB2-negative disease were alive at 5 years, with a median follow-up of 5.6 years, compared with only 54% for children with average-risk, ERBB2-positive tumors (n=13; P=.0001). TRKC, MYCC, and MYCN expression and histopathologic subtype were not associated with prognosis in this study. CONCLUSION Central and rapid molecular analysis of frozen medulloblastomas collected from multiple institutions is feasible. ERBB2 expression and clinical risk factors together constitute a highly accurate disease risk stratification tool.


Annals of Oncology | 2015

EURAMOS-1, an international randomised study for osteosarcoma: results from pre-randomisation treatment.

Jeremy Whelan; S. Bielack; Neyssa Marina; Sigbjørn Smeland; Gordana Jovic; Jane Hook; Mark Krailo; Jakob K. Anninga; Trude Butterfass-Bahloul; Tom Böhling; Gabriele Calaminus; M. Capra; C. Deffenbaugh; Catharina Dhooge; Mikael Eriksson; Adrienne M. Flanagan; Hans Gelderblom; Allen M. Goorin; Richard Gorlick; G. Gosheger; Robert J. Grimer; Kirsten Sundby Hall; K. Helmke; P. C. W. Hogendoorn; Gernot Jundt; Leo Kager; T. Kuehne; Ching Lau; George Douglas Letson; James S. Meyer

This manuscript describes the experience from registration until randomisation for a cohort of 2260 patients with osteosarcoma who joined the EURAMOS-1 trial. This includes pre-operative chemotherapy and surgery. It sets out the practical issues in collaboration and in achieving randomisation.


Human Molecular Genetics | 2011

Chromosome 7p11.2 (EGFR) variation influences glioma risk

Marc Sanson; Fay J. Hosking; Sanjay Shete; Diana Zelenika; Sara E. Dobbins; Yussanne Ma; Victor Enciso-Mora; Ahmed Idbaih; Jean Yves Delattre; Khê Hoang-Xuan; Yannick Marie; Blandine Boisselier; Catherine Carpentier; Xiao Wei Wang; Anna Luisa Di Stefano; Marianne Labussière; Konstantinos Gousias; Johannes Schramm; Anne Boland; Doris Lechner; Ivo Gut; Georgina Armstrong; Yanhong Liu; Robert Yu; Ching Lau; Maria Chiara Di Bernardo; Lindsay B. Robertson; Kenneth Muir; Sarah J. Hepworth; Anthony J. Swerdlow

While gliomas are the most common primary brain tumors, their etiology is largely unknown. To identify novel risk loci for glioma, we conducted genome-wide association (GWA) analysis of two case-control series from France and Germany (2269 cases and 2500 controls). Pooling these data with previously reported UK and US GWA studies provided data on 4147 glioma cases and 7435 controls genotyped for 424 460 common tagging single-nucleotide polymorphisms. Using these data, we demonstrate two statistically independent associations between glioma and rs11979158 and rs2252586, at 7p11.2 which encompasses the EGFR gene (population-corrected statistics, P(c) = 7.72 × 10(-8) and 2.09 × 10(-8), respectively). Both associations were independent of tumor subtype, and were independent of EGFR amplification, p16INK4a deletion and IDH1 mutation status in tumors; compatible with driver effects of the variants on glioma development. These findings show that variation in 7p11.2 is a determinant of inherited glioma risk.


Cancer Epidemiology, Biomarkers & Prevention | 2007

GLIOGENE—an International Consortium to Understand Familial Glioma

Beatrice Malmer; Phyllis Adatto; Georgina Armstrong; Jill S. Barnholtz-Sloan; Jonine L. Bernstein; Elizabeth B. Claus; Faith G. Davis; Richard S. Houlston; Dora Il'yasova; Robert B. Jenkins; Christoffer Johansen; Rose Lai; Ching Lau; Bridget J. McCarthy; Hanne Nielsen; Sara H. Olson; Siegal Sadetzki; Sanjay Shete; Fredrik Wiklund; Margaret Wrensch; Ping Yang; Melissa L. Bondy

Evidence for familial aggregation of glioma has been documented in both case-control and cohort studies and occurs apart from the well-described rare inherited genetic syndromes involving glioma: neurofibromatosis type 1 and 2, tuberous sclerosis, Turcots syndrome, and Li-Fraumeni syndrome. Nonsyndromic glioma families have been studied but no genes have been identified in the two published linkage studies of familial glioma probably due to the small number of families. Because glioma is a rare but devastating cancer, and a family history of glioma has been observed in ∼5% of the cases, we initiated an international consortium to identify glioma families not affected by syndromes to better understand the inherited factors related to this disease. The international consortium GLIOGENE is an acronym for “glioma gene” and includes 15 research groups in North America, Europe, and Israel to study familial glioma. The overarching goal is to characterize genes in glioma families using a genome-wide single-nucleotide polymorphism approach and conducting linkage analysis to identify new genomic regions or loci that could harbor genes important for gliomagenesis. Here, we review the rationale for studying familial glioma and our proposed strategy for the GLIOGENE study. (Cancer Epidemiol Biomarkers Prev 2007;16(9):1730–4)


Neuro-oncology | 2009

A pilot study of risk-adapted radiotherapy and chemotherapy in patients with supratentorial PNET

Murali Chintagumpala; Tim Hassall; Shawna L. Palmer; David M. Ashley; Dana Wallace; Kimberly A. Kasow; Thomas E. Merchant; Matthew J. Krasin; Robert C. Dauser; Frederick A. Boop; Robert A. Krance; Shiao Y. Woo; Robyn Cheuk; Ching Lau; Richard J. Gilbertson; Amar Gajjar

We undertook this study to estimate the event-free survival (EFS) of patients with newly diagnosed supratentorial primitive neuroectodermal tumor (SPNET) treated with risk-adapted craniospinal irradiation (CSI) with additional radiation to the primary tumor site and subsequent high-dose chemotherapy supported by stem cell rescue. Between 1996 and 2003, 16 patients with SPNET were enrolled. High-risk (HR) disease was differentiated from average-risk (AR) disease by the presence of residual tumor (M(0) and tumor size > 1.5 cm(2)) or disseminated disease in the neuraxis (M(1)-M(3)). Patients received risk-adapted CSI: those with AR disease received 23.4 Gy; those with HR disease, 36-39.6 Gy. The tumor bed received a total of 55.8 Gy. Subsequently, all patients received four cycles of high-dose cyclophosphamide, cisplatin, and vincristine with stem cell support. The median age at diagnosis was 7.9 years; eight patients were female. Seven patients had pineal PNET. Twelve patients are alive at a median follow-up of 5.4 years. The 5-year EFS and overall survival (OS) estimates for all patients were 68% +/- 14% and 73% +/- 13%. The 5-year EFS and OS estimates were 75% +/- 17% and 88% +/- 13%, respectively, for the eight patients with AR disease and 60% +/- 19% and 58% +/- 19%, respectively, for the eight with HR disease. No deaths were due to toxicity. High-dose cyclophosphamide-based chemotherapy with stem cell support after risk-adapted CSI results in excellent EFS estimates for patients with newly diagnosed AR SPNET. Further, this chemotherapy allows for a reduction in the dose of CSI used to treat AR SPNET without compromising EFS.


Neuro-oncology | 2009

Glutathione S-transferase M1 and T1 polymorphisms may predict adverse effects after therapy in children with medulloblastoma.

Nadia Barahmani; Sarah Carpentieri; Xio Nan Li; Tao Wang; Yumei Cao; Laura Howe; Lindsay Kilburn; Murali Chintagumpala; Ching Lau; M. Fatih Okcu

Glutathione S-transferases (GSTs) are polymorphic enzymes that catalyze the glutathione conjugation of alkylating agents, platinum compounds, and free radicals formed by radiation used to treat medulloblastoma. We hypothesized that GST polymorphisms may be responsible, in part, for individual differences in toxicity and responses in pediatric medulloblastoma. We investigated the relationship between GSTM1 and GSTT1 polymorphisms and survival and toxicity in 42 children with medulloblastoma diagnosed and treated at the Texas Childrens Cancer Center. We conducted Kaplan-Meier analyses to determine if the GST polymorphisms were related to progression-free survival (PFS) and performed logistic regression to explore associations between GST polymorphisms and occurrence of grade 3 or greater (> or =Gr 3) myelosuppression, ototoxicity, nephrotoxicity, neurotoxicity, and intellectual impairment. Patients with at least one null genotype had a 4.3 (95% confidence interval, 1.1-16.8), 3.7 (1-13.6), and 6.4 (1.2-34) times increased risk for any > or =Gr 3 toxicity, any > or =Gr 3 toxicity excluding peripheral neuropathy, and any > or =Gr 3 toxicity requiring omission or cessation of chemotherapy, respectively. Compared with all others, patients with at least one null genotype had, on average, 27.2 (p x= 0.0002), 29 (p = 0.0004), and 21.7 (p = 0.002) lower full-scale, performance, and verbal intelligence quotient (IQ) scores, respectively. GSTM1 and GSTT1 polymorphisms may predict adverse events, including cognitive impairment after therapy, in patients with medulloblastoma. A larger study to validate these findings is under way.


Nature Genetics | 2017

Genome-wide association study of glioma subtypes identifies specific differences in genetic susceptibility to glioblastoma and non-glioblastoma tumors.

Beatrice Melin; Jill S. Barnholtz-Sloan; Margaret Wrensch; Christoffer Johansen; Dora Il'yasova; Ben Kinnersley; Quinn T. Ostrom; Karim Labreche; Yanwen Chen; Georgina Armstrong; Yanhong Liu; Jeanette E. Eckel-Passow; Paul A. Decker; Marianne Labussière; Ahmed Idbaih; Khê Hoang-Xuan; Anna-Luisa Di Stefano; Karima Mokhtari; Jean-Yves Delattre; Peter Broderick; Pilar Galan; Konstantinos Gousias; Johannes Schramm; Minouk J. Schoemaker; Sarah Fleming; Stefan Herms; Stefanie Heilmann; Markus M. Nöthen; Heinz-Erich Wichmann; Stefan Schreiber

Genome-wide association studies (GWAS) have transformed our understanding of glioma susceptibility, but individual studies have had limited power to identify risk loci. We performed a meta-analysis of existing GWAS and two new GWAS, which totaled 12,496 cases and 18,190 controls. We identified five new loci for glioblastoma (GBM) at 1p31.3 (rs12752552; P = 2.04 × 10−9, odds ratio (OR) = 1.22), 11q14.1 (rs11233250; P = 9.95 × 10−10, OR = 1.24), 16p13.3 (rs2562152; P = 1.93 × 10−8, OR = 1.21), 16q12.1 (rs10852606; P = 1.29 × 10−11, OR = 1.18) and 22q13.1 (rs2235573; P = 1.76 × 10−10, OR = 1.15), as well as eight loci for non-GBM tumors at 1q32.1 (rs4252707; P = 3.34 × 10−9, OR = 1.19), 1q44 (rs12076373; P = 2.63 × 10−10, OR = 1.23), 2q33.3 (rs7572263; P = 2.18 × 10−10, OR = 1.20), 3p14.1 (rs11706832; P = 7.66 × 10−9, OR = 1.15), 10q24.33 (rs11598018; P = 3.39 × 10−8, OR = 1.14), 11q21 (rs7107785; P = 3.87 × 10−10, OR = 1.16), 14q12 (rs10131032; P = 5.07 × 10−11, OR = 1.33) and 16p13.3 (rs3751667; P = 2.61 × 10−9, OR = 1.18). These data substantiate that genetic susceptibility to GBM and non-GBM tumors are highly distinct, which likely reflects different etiology.


British Journal of Cancer | 2013

Low penetrance susceptibility to glioma is caused by the TP53 variant rs78378222

Victor Enciso-Mora; Fay J. Hosking; A. L. Di Stefano; Diana Zelenika; Sanjay Shete; Peter Broderick; Ahmed Idbaih; Jean Yves Delattre; Khê Hoang-Xuan; Yannick Marie; Marianne Labussière; Agusti Alentorn; Pietro Ciccarino; Marta Rossetto; Georgina Armstrong; Yongmei Liu; Konstantinos Gousias; Johannes Schramm; Ching Lau; Sarah J. Hepworth; Minouk J. Schoemaker; Konstantin Strauch; Martina Müller-Nurasyid; Stefan Schreiber; Andre Franke; Susanne Moebus; Lewin Eisele; Anthony J. Swerdlow; Matthias Simon; Melissa L. Bondy

Background:Most of the heritable risk of glioma is presently unaccounted for by mutations in known genes. In addition to rare inactivating germline mutations in TP53 causing glioma in the context of the Li-Fraumeni syndrome, polymorphic variation in TP53 may also contribute to the risk of developing glioma.Methods:To comprehensively evaluate the impact of variation in TP53 on risk, we analysed 23 tagSNPs and imputed 2377 unobserved genotypes in four series totaling 4147 glioma cases and 7435 controls.Results:The strongest validated association signal was shown by the imputed single-nucleotide polymorphism (SNP) rs78378222 (P=6.86 × 10−24, minor allele frequency ∼0.013). Confirmatory genotyping confirmed the high quality of the imputation. The association between rs78378222 and risk was seen for both glioblastoma multiforme (GBM) and non-GBM tumours. We comprehensively examined the relationship between rs78378222 and overall survival in two of the case series totaling 1699 individuals. Despite employing statistical tests sensitive to the detection of differences in early survival, no association was shown.Conclusion:Our data provided strong validation of rs78378222 as a risk factor for glioma but do not support the tenet that the polymorphism being a clinically useful prognostic marker. Acquired TP53 inactivation is a common feature of glioma. As rs78378222 changes the polyadenylation signal of TP53 leading to impaired 3′-end processing of TP53 mRNA, the SNP has strong plausibility for being directly functional contributing to the aetiological basis of glioma.

Collaboration


Dive into the Ching Lau's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sanjay Shete

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Melissa L. Bondy

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Christoffer Johansen

Copenhagen University Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jill S. Barnholtz-Sloan

Case Western Reserve University

View shared research outputs
Researchain Logo
Decentralizing Knowledge