Network


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

Hotspot


Dive into the research topics where Chang Qi Zhu is active.

Publication


Featured researches published by Chang Qi Zhu.


Nature Medicine | 2008

Gene expression-based survival prediction in lung adenocarcinoma: A multi-site, blinded validation study

Kerby Shedden; Jeremy M. G. Taylor; Steven A. Enkemann; Ming-Sound Tsao; Timothy J. Yeatman; William L. Gerald; Steven Eschrich; Igor Jurisica; Thomas J. Giordano; David E. Misek; Andrew C. Chang; Chang Qi Zhu; Daniel Strumpf; Samir M. Hanash; Frances A. Shepherd; Keyue Ding; Lesley Seymour; Katsuhiko Naoki; Nathan A. Pennell; Barbara A. Weir; Roel G.W. Verhaak; Christine Ladd-Acosta; Todd R. Golub; Michael Gruidl; Anupama Sharma; Janos Szoke; Maureen F. Zakowski; Valerie W. Rusch; Mark G. Kris; Agnes Viale

Although prognostic gene expression signatures for survival in early-stage lung cancer have been proposed, for clinical application, it is critical to establish their performance across different subject populations and in different laboratories. Here we report a large, training–testing, multi-site, blinded validation study to characterize the performance of several prognostic models based on gene expression for 442 lung adenocarcinomas. The hypotheses proposed examined whether microarray measurements of gene expression either alone or combined with basic clinical covariates (stage, age, sex) could be used to predict overall survival in lung cancer subjects. Several models examined produced risk scores that substantially correlated with actual subject outcome. Most methods performed better with clinical data, supporting the combined use of clinical and molecular information when building prognostic models for early-stage lung cancer. This study also provides the largest available set of microarray data with extensive pathological and clinical annotation for lung adenocarcinomas.


Nature Genetics | 2009

SOX2 is an amplified lineage-survival oncogene in lung and esophageal squamous cell carcinomas

Adam J. Bass; Hideo Watanabe; Craig H. Mermel; Yu Ss; Sven Perner; Roeland Verhaak; So Young Kim; Leslie Wardwell; Pablo Tamayo; Irit Gat-Viks; Alex H. Ramos; Michele S. Woo; Barbara A. Weir; Gad Getz; Rameen Beroukhim; Michael O'Kelly; Amit Dutt; Orit Rozenblatt-Rosen; Piotr Dziunycz; Justin Komisarof; Lucian R. Chirieac; Christopher J. Lafargue; Veit Scheble; Theresia Wilbertz; Changqing Ma; Shilpa Rao; Hiroshi Nakagawa; Douglas B. Stairs; Lin Lin; Thomas J. Giordano

Lineage-survival oncogenes are activated by somatic DNA alterations in cancers arising from the cell lineages in which these genes play a role in normal development. Here we show that a peak of genomic amplification on chromosome 3q26.33 found in squamous cell carcinomas (SCCs) of the lung and esophagus contains the transcription factor gene SOX2, which is mutated in hereditary human esophageal malformations, is necessary for normal esophageal squamous development, promotes differentiation and proliferation of basal tracheal cells and cooperates in induction of pluripotent stem cells. SOX2 expression is required for proliferation and anchorage-independent growth of lung and esophageal cell lines, as shown by RNA interference experiments. Furthermore, ectopic expression of SOX2 here cooperated with FOXE1 or FGFR2 to transform immortalized tracheobronchial epithelial cells. SOX2-driven tumors show expression of markers of both squamous differentiation and pluripotency. These characteristics identify SOX2 as a lineage-survival oncogene in lung and esophageal SCC.


Journal of Clinical Oncology | 2010

Prognostic and Predictive Gene Signature for Adjuvant Chemotherapy in Resected Non–Small-Cell Lung Cancer

Chang Qi Zhu; Keyue Ding; Dan Strumpf; Barbara A. Weir; Matthew Meyerson; Nathan A. Pennell; Roman K. Thomas; Katsuhiko Naoki; Christine Ladd-Acosta; Ni Liu; Melania Pintilie; Sandy D. Der; Lesley Seymour; Igor Jurisica; Frances A. Shepherd; Ming Sound Tsao

PURPOSEnThe JBR.10 trial demonstrated benefit from adjuvant cisplatin/vinorelbine (ACT) in early-stage non-small-cell lung cancer (NSCLC). We hypothesized that expression profiling may identify stage-independent subgroups who might benefit from ACT.nnnPATIENTS AND METHODSnGene expression profiling was conducted on mRNA from 133 frozen JBR.10 tumor samples (62 observation [OBS], 71 ACT). The minimum gene set that was selected for the greatest separation of good and poor prognosis patient subgroups in OBS patients was identified. The prognostic value of this gene signature was tested in four independent published microarray data sets and by quantitative reverse-transcriptase polymerase chain reaction (RT-qPCR).nnnRESULTSnA 15-gene signature separated OBS patients into high-risk and low-risk subgroups with significantly different survival (hazard ratio [HR], 15.02; 95% CI, 5.12 to 44.04; P < .001; stage I HR, 13.31; P < .001; stage II HR, 13.47; P < .001). The prognostic effect was verified in the same 62 OBS patients where gene expression was assessed by qPCR. Furthermore, it was validated consistently in four separate microarray data sets (total 356 stage IB to II patients without adjuvant treatment) and additional JBR.10 OBS patients by qPCR (n = 19). The signature was also predictive of improved survival after ACT in JBR.10 high-risk patients (HR, 0.33; 95% CI, 0.17 to 0.63; P = .0005), but not in low-risk patients (HR, 3.67; 95% CI, 1.22 to 11.06; P = .0133; interaction P < .001). Significant interaction between risk groups and ACT was verified by qPCR.nnnCONCLUSIONnThis 15-gene expression signature is an independent prognostic marker in early-stage, completely resected NSCLC, and to our knowledge, is the first signature that has demonstrated the potential to select patients with stage IB to II NSCLC most likely to benefit from adjuvant chemotherapy with cisplatin/vinorelbine.


Journal of Clinical Oncology | 2007

Prognostic and Predictive Importance of p53 and RAS for Adjuvant Chemotherapy in Non–Small-Cell Lung Cancer

Ming-Sound Tsao; Sarit Aviel-Ronen; Keyue Ding; Davina Lau; Ni Liu; Akira Sakurada; Marlo Whitehead; Chang Qi Zhu; Robert B. Livingston; David Johnson; James R. Rigas; Lesley Seymour; Timothy Winton; Frances A. Shepherd

PURPOSEnp53 and RAS are multifunctional proteins that are critical to cell cycle regulation, apoptosis, cell survival, gene transcription, response to stress, and DNA repair. We have evaluated the prognostic and predictive value of p53 gene/protein aberrations using tumor samples from JBR.10, a North American phase III intergroup trial that randomly assigned 482 patients with completely resected stage IB and II non-small-cell lung cancer (NSCLC) to receive four cycles of adjuvant cisplatin plus vinorelbine or observation alone.nnnMETHODSnp53 protein expression was evaluated by immunohistochemistry. Mutations in exons 5 to 9 of the p53 gene were determined by denaturing high-performance liquid chromatography and confirmed by sequencing. RAS mutations were identified by allelic specific oligonucleotide hybridization.nnnRESULTSnOf 253 patients, 132 (52%) were positive for p53 protein overexpression. Untreated p53-positive patients had significantly shorter overall survival than did patients with p53-negative tumors (hazard ratio [HR] = 1.89; 95% CI, 1.07 to 3.34; P = .03). However, these p53-positive patients also had a significantly greater survival benefit from adjuvant chemotherapy (HR = 0.54; P = .02) compared with patients with p53-negative tumors (HR = 1.40; P = .26; interaction P = .02). Mutations of p53 and RAS genes were found in 124 (31%) of 397 and 117 (26%) of 450 patients, respectively. Mutations in these genes were neither prognostic for survival nor predictive of a differential benefit from adjuvant chemotherapy.nnnCONCLUSIONnp53 protein overexpression is a significant prognostic marker of shortened survival, and also a significant predictive marker for a differentially greater benefit from adjuvant chemotherapy in completely resected NSCLC patients.


Journal of Clinical Oncology | 2007

Three-gene prognostic classifier for early-stage non small-cell lung cancer.

Suzanne K. Lau; Paul C. Boutros; Melania Pintilie; Fiona Blackhall; Chang Qi Zhu; Dan Strumpf; Michael R. Johnston; Gail Darling; Shaf Keshavjee; Thomas K. Waddell; Ni Liu; Davina Lau; Linda Z. Penn; Frances A. Shepherd; Igor Jurisica; Sandy D. Der; Ming-Sound Tsao

PURPOSEnSeveral microarray studies have reported gene expression signatures that classify non-small-cell lung carcinoma (NSCLC) patients into different prognostic groups. However, the prognostic gene lists reported to date overlap poorly across studies, and few have been validated independently using more quantitative assay methods.nnnPATIENTS AND METHODSnThe expression of 158 putative prognostic genes identified in previous microarray studies was analyzed by reverse transcription quantitative polymerase chain reaction in the tumors of 147 NSCLC patients. Concordance indices and risk scores were used to identify a stage-independent set of genes that could classify patients with significantly different prognoses.nnnRESULTSnWe have identified a three-gene classifier (STX1A, HIF1A, and CCR7) for overall survival (hazard ratio = 3.8; 95% CI, 1.7 to 8.2; P < .001). The classifier was also able to stratify stage I and II patients and further improved the predictive ability of clinical factors such as histology and tumor stage. The predictive value of this three-gene classifier was validated in two large independent microarray data sets from Harvard and Duke Universities.nnnCONCLUSIONnWe have identified a new three-gene classifier that is independent of and improves on stage to stratify early-stage NSCLC patients with significantly different prognoses. This classifier may be tested further for its potential value to improve the selection of resected NSCLC patients in adjuvant therapy.


Nature Communications | 2014

Integrated Omic analysis of lung cancer reveals metabolism proteome signatures with prognostic impact

Lei Li; Yuhong Wei; Christine To; Chang Qi Zhu; Jiefei Tong; Nhu-An Pham; Paul Taylor; Ignatchenko; Alexandr Ignatchenko; Wei Zhang; Dennis Wang; Naoki Yanagawa; Ming Li; Melania Pintilie; Geoffrey Liu; Lakshmi Muthuswamy; Shepherd Fa; M. Tsao; Thomas Kislinger; Michael F. Moran

Cancer results from processes prone to selective pressure and dysregulation acting along the sequence-to-phenotype continuum DNA → RNA → protein → disease. However, the extent to which cancer is a manifestation of the proteome is unknown. Here we present an integrated omic map representing non-small cell lung carcinoma. Dysregulated proteins not previously implicated as cancer drivers are encoded throughout the genome including, but not limited to, regions of recurrent DNA amplification/deletion. Clustering reveals signatures composed of metabolism proteins particularly highly recapitulated between patient-matched primary and xenograft tumours. Interrogation of The Cancer Genome Atlas reveals cohorts of patients with lung and other cancers that have DNA alterations in genes encoding the signatures, and this was accompanied by differences in survival. The recognition of genome and proteome alterations as related products of selective pressure driving the disease phenotype may be a general approach to uncover and group together cryptic, polygenic disease drivers.


Cancer Research | 2011

Interaction between MYC and MCL1 in the Genesis and Outcome of Non–Small-Cell Lung Cancer

Thaddeus D. Allen; Chang Qi Zhu; Kirk D. Jones; Naoki Yanagawa; Ming-Sound Tsao; J. Michael Bishop

MYC exerts both positive and negative functions in cancer cells, such that its procancerous effects are unmasked only after its anticancer effects are blocked. Here we used multiple mouse models of lung adenocarcinoma to identify genetic events that can cooperate with MYC activation to promote the genesis of non-small-cell lung cancer (NSCLC), the most common form of lung cancer in humans. MYC overexpression targeted to pulmonary alveolar cells was sufficient to induce lung adenomas and carcinomas. Tumorigenesis was assisted by either spontaneous mutations in Kras or experimental introduction of activated RAS, but investigations revealed that additional events were required to circumvent apoptosis, one of the most significant negative functions exerted by MYC. We determined that overexpression of the antiapoptotic protein MCL1 was sufficient to circumvent apoptosis in this setting. Previous clinical studies have indicated that prognosis of human NSCLC is not associated with MCL1, despite its overexpression in many NSCLCs. In reexamining the prognostic value in this setting, we found that MCL1 overexpression does correlate with poor patient survival, but only when accompanied by MYC overexpression. Our findings therefore produce a convergence of mouse and human results that explain how MCL1 can block an important negative consequence of MYC overexpression in both experimental models and clinical cases of NSCLC.


International Journal of Cancer | 2017

Molecular heterogeneity of non-small cell lung carcinoma patient-derived xenografts closely reflect their primary tumors.

Dennis Wang; Nhu An Pham; Jiefei Tong; Shingo Sakashita; Ghassan Allo; Lucia Kim; Naoki Yanagawa; Vibha Raghavan; Yuhong Wei; Christine To; Quang M. Trinh; Maud H. W. Starmans; Michelle Chan-Seng-Yue; Dianne Chadwick; Lei Li; Chang Qi Zhu; Ni Liu; Ming Li; Sharon Lee; Dan Strumpf; Paul Taylor; Nadeem Moghal; Geoffrey Liu; Paul C. Boutros; Thomas Kislinger; Melania Pintilie; Igor Jurisica; Frances A. Shepherd; John D. McPherson; Lakshmi Muthuswamy

Availability of lung cancer models that closely mimic human tumors remains a significant gap in cancer research, as tumor cell lines and mouse models may not recapitulate the spectrum of lung cancer heterogeneity seen in patients. We aimed to establish a patient‐derived tumor xenograft (PDX) resource from surgically resected non‐small cell lung cancer (NSCLC). Fresh tumor tissue from surgical resection was implanted and grown in the subcutaneous pocket of non‐obese severe combined immune deficient (NOD SCID) gamma mice. Subsequent passages were in NOD SCID mice. A subset of matched patient and PDX tumors and non‐neoplastic lung tissues were profiled by whole exome sequencing, single nucleotide polymorphism (SNP) and methylation arrays, and phosphotyrosine (pY)‐proteome by mass spectrometry. The data were compared to published NSCLC datasets of NSCLC primary and cell lines. 127 stable PDXs were established from 441 lung carcinomas representing all major histological subtypes: 52 adenocarcinomas, 62 squamous cell carcinomas, one adeno‐squamous carcinoma, five sarcomatoid carcinomas, five large cell neuroendocrine carcinomas, and two small cell lung cancers. Somatic mutations, gene copy number and expression profiles, and pY‐proteome landscape of 36 PDXs showed greater similarity with patient tumors than with established cell lines. Novel somatic mutations on cancer associated genes were identified but only in PDXs, likely due to selective clonal growth in the PDXs that allows detection of these low allelic frequency mutations. The results provide the strongest evidence yet that PDXs established from lung cancers closely mimic the characteristics of patient primary tumors.


Journal of Clinical Oncology | 2007

Updated molecular analyses of exons 19 and 21 of the epidermal growth factor receptor (EGFR) gene and codons 12 and 13 of the KRAS gene in non-small cell lung cancer (NSCLC) patients treated with erlotinib in National Cancer Institute of Cancer

Frances A. Shepherd; Keyue Ding; Akira Sakurada; G. da Cunha Santos; Chang Qi Zhu; Lesley Seymour; Marlo Whitehead; Suzanne Kamel-Reid; Jeremy A. Squire; Ming Tsao


Lung Cancer | 2005

O-126 Expression profiling of matrix metalloproteinases (MMPs) and inhibitors reveals a novel prognostic role for MMP-19 in patients with non-small cell lung cancer

Fiona Blackhall; Melania Pintilie; Ni Liu; C. Pennington; Chang Qi Zhu; Frances A. Shepherd; D. Edwards; Ming-Sound Tsao

Collaboration


Dive into the Chang Qi Zhu's collaboration.

Top Co-Authors

Avatar

Frances A. Shepherd

Princess Margaret Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Ming-Sound Tsao

Princess Margaret Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Ni Liu

University Health Network

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Melania Pintilie

Princess Margaret Cancer Centre

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dan Strumpf

University Health Network

View shared research outputs
Top Co-Authors

Avatar

Geoffrey Liu

Princess Margaret Cancer Centre

View shared research outputs
Researchain Logo
Decentralizing Knowledge